US20120029475A1 - Drug Combinations Useful for Prevention of Restenosis - Google Patents

Drug Combinations Useful for Prevention of Restenosis Download PDF

Info

Publication number
US20120029475A1
US20120029475A1 US13/227,002 US201113227002A US2012029475A1 US 20120029475 A1 US20120029475 A1 US 20120029475A1 US 201113227002 A US201113227002 A US 201113227002A US 2012029475 A1 US2012029475 A1 US 2012029475A1
Authority
US
United States
Prior art keywords
stent
combination
restenosis
rapamycin
dexamethasone
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/227,002
Inventor
Gregory A. Kopia
Gerald H. Llanos
Robert Falotico
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=26899464&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20120029475(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Individual filed Critical Individual
Priority to US13/227,002 priority Critical patent/US20120029475A1/en
Publication of US20120029475A1 publication Critical patent/US20120029475A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2/00Filters implantable into blood vessels; Prostheses, i.e. artificial substitutes or replacements for parts of the body; Appliances for connecting them with the body; Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/82Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/86Stents in a form characterised by the wire-like elements; Stents in the form characterised by a net-like or mesh-like structure
    • A61F2/90Stents in a form characterised by the wire-like elements; Stents in the form characterised by a net-like or mesh-like structure characterised by a net-like or mesh-like structure
    • A61F2/91Stents in a form characterised by the wire-like elements; Stents in the form characterised by a net-like or mesh-like structure characterised by a net-like or mesh-like structure made from perforated sheet material or tubes, e.g. perforated by laser cuts or etched holes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2/00Filters implantable into blood vessels; Prostheses, i.e. artificial substitutes or replacements for parts of the body; Appliances for connecting them with the body; Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/82Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/86Stents in a form characterised by the wire-like elements; Stents in the form characterised by a net-like or mesh-like structure
    • A61F2/90Stents in a form characterised by the wire-like elements; Stents in the form characterised by a net-like or mesh-like structure characterised by a net-like or mesh-like structure
    • A61F2/91Stents in a form characterised by the wire-like elements; Stents in the form characterised by a net-like or mesh-like structure characterised by a net-like or mesh-like structure made from perforated sheet material or tubes, e.g. perforated by laser cuts or etched holes
    • A61F2/915Stents in a form characterised by the wire-like elements; Stents in the form characterised by a net-like or mesh-like structure characterised by a net-like or mesh-like structure made from perforated sheet material or tubes, e.g. perforated by laser cuts or etched holes with bands having a meander structure, adjacent bands being connected to each other
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/436Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having oxygen as a ring hetero atom, e.g. rapamycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • A61K31/726Glycosaminoglycans, i.e. mucopolysaccharides
    • A61K31/727Heparin; Heparan
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/14Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L31/16Biologically active materials, e.g. therapeutic substances
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2/00Filters implantable into blood vessels; Prostheses, i.e. artificial substitutes or replacements for parts of the body; Appliances for connecting them with the body; Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/82Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/86Stents in a form characterised by the wire-like elements; Stents in the form characterised by a net-like or mesh-like structure
    • A61F2/90Stents in a form characterised by the wire-like elements; Stents in the form characterised by a net-like or mesh-like structure characterised by a net-like or mesh-like structure
    • A61F2/91Stents in a form characterised by the wire-like elements; Stents in the form characterised by a net-like or mesh-like structure characterised by a net-like or mesh-like structure made from perforated sheet material or tubes, e.g. perforated by laser cuts or etched holes
    • A61F2/915Stents in a form characterised by the wire-like elements; Stents in the form characterised by a net-like or mesh-like structure characterised by a net-like or mesh-like structure made from perforated sheet material or tubes, e.g. perforated by laser cuts or etched holes with bands having a meander structure, adjacent bands being connected to each other
    • A61F2002/91533Stents in a form characterised by the wire-like elements; Stents in the form characterised by a net-like or mesh-like structure characterised by a net-like or mesh-like structure made from perforated sheet material or tubes, e.g. perforated by laser cuts or etched holes with bands having a meander structure, adjacent bands being connected to each other characterised by the phase between adjacent bands
    • A61F2002/91541Adjacent bands are arranged out of phase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2250/00Special features of prostheses classified in groups A61F2/00 - A61F2/26 or A61F2/82 or A61F9/00 or A61F11/00 or subgroups thereof
    • A61F2250/0058Additional features; Implant or prostheses properties not otherwise provided for
    • A61F2250/0067Means for introducing or releasing pharmaceutical products into the body
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2250/00Special features of prostheses classified in groups A61F2/00 - A61F2/26 or A61F2/82 or A61F9/00 or A61F11/00 or subgroups thereof
    • A61F2250/0058Additional features; Implant or prostheses properties not otherwise provided for
    • A61F2250/0067Means for introducing or releasing pharmaceutical products into the body
    • A61F2250/0068Means for introducing or releasing pharmaceutical products into the body the pharmaceutical product being in a reservoir
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2310/00Prostheses classified in A61F2/28 or A61F2/30 - A61F2/44 being constructed from or coated with a particular material
    • A61F2310/00389The prosthesis being coated or covered with a particular material
    • A61F2310/0097Coating or prosthesis-covering structure made of pharmaceutical products, e.g. antibiotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/40Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a specific therapeutic activity or mode of action
    • A61L2300/41Anti-inflammatory agents, e.g. NSAIDs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/40Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a specific therapeutic activity or mode of action
    • A61L2300/416Anti-neoplastic or anti-proliferative or anti-restenosis or anti-angiogenic agents, e.g. paclitaxel, sirolimus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/40Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a specific therapeutic activity or mode of action
    • A61L2300/43Hormones, e.g. dexamethasone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/40Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a specific therapeutic activity or mode of action
    • A61L2300/45Mixtures of two or more drugs, e.g. synergistic mixtures
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/60Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a special physical form
    • A61L2300/606Coatings

Definitions

  • This invention describes the delivery of different drug combinations, either systemically or locally, particularly from an intravascular stent, directly from micropores in the stent body or mixed or bound to a polymer coating applied on stent, to inhibit neointimal tissue proliferation and thereby prevent restenosis.
  • This invention given either systemically or locally also facilitates the performance of the stent in inhibiting restenosis.
  • Atherosclerotic lesions which limit or obstruct coronary blood flow, are the major cause of ischemic heart disease related mortality, resulting in 500,000-600,000 deaths annually.
  • Percutaneous transluminal coronary angioplasty (PTCA) to open the obstructed artery was performed in over 550,000 patients in the U.S. and 945,000+ patients worldwide in 1996 (Lemaitre et al., 1996).
  • PTCA Percutaneous transluminal coronary angioplasty
  • a major limitation of this technique is the problem of post-PTCA closure of the vessel, both immediately after PTCA (acute occlusion) and in the long term (restenosis): 30% of patients with subtotal lesions and 50% of patients with chronic total lesions will go on to restenosis after angioplasty.
  • restenosis is a significant problem in patients undergoing saphenous vein bypass graft.
  • the mechanism of acute occlusion appears to involve several factors and may result from vascular recoil with resultant closure of the artery and/or deposition of blood platelets along the damaged length of the newly opened blood vessel followed by formation of a fibrin/red blood cell thrombus.
  • Restenosis after angioplasty is a more gradual process and involves initial formation of a subcritical thrombosis with release from adherent platelets of cell derived growth factors with subsequent proliferation of intimal smooth muscle cells and local infiltration of inflammatory cells contributing to vascular hyperplasia. It is important to note that multiple processes, among those including thrombosis, cell proliferation, cell migration and inflammation each seem to contribute to the restenotic process.
  • a 30-50% restenosis rate translates to 120,000-200,000 U.S. patients at risk from restenosis. If only 80% of such patients elect repeat angioplasty (with the remaining 20% electing coronary artery bypass graft) is added to the cost of coronary artery bypass graft for the remaining 20%, the total cost for restenosis easily reaches into billions of dollars. Thus, successful prevention of restenosis could result not only in significant therapeutic benefit but also in significant health care savings.
  • heparin and heparin fragments include: heparin and heparin fragments (Clowes, A. W. and Karnovsky M., Nature, 265: 25-26, 1977; Guyton, J. R. et al., Circ. Res., 46: 625-634, 1980; Clowes, A. W. and Clowes, M. M., Lab. Invest. 52: 611-616, 1985; Clowes, A. W. and Clowes, M. M., Circ. Res.
  • angiopeptin (Lundergan, C. F. et al., Am. J. Cardiol. 17 (Suppl. B); 132B-136B, 1991), cyclosporin A (Jonasson, L. et al., Proc. Natl., Acad. Sci., 85: 2303, 1988), goat-anti-rabbit PDGF antibody (Ferns, G. A. A. et al., Science 253: 1129-1132, 1991), terbinafine (Nemecek, G. M. et al., J. Pharmacol. Exp. Thera.
  • agents with diverse mechanisms of SMC inhibition may have therapeutic utility in reducing intimal hyperplasia.
  • the 7E3 humanized monoclonal antibody fragment to the platelet GP IIb/IIIa receptor is still under study but has not shown promising results for the reduction in restenosis following angioplasty and stenting.
  • Other agents which have also been unsuccessful in the prevention of restenosis, include the calcium channel antagonists, prostacyclin mimetics, angiotensin converting enzyme inhibitors, serotonin receptor antagonists, and antiproliferative agents.
  • antiproliferative (or anti-restenosis) concentrations may exceed the known toxic concentrations of these agents so that levels sufficient to produce smooth muscle inhibition may not be reached (Mak and Topol, 1997; Lang et al., 1991; Popma et al., 1991).
  • stents Unlike systemic pharmacologic therapy, stents have proven useful in partially preventing restenosis. Stents, are balloon-expandable slotted metal tubes (usually, but not limited to, stainless steel), which, when expanded within the lumen of an angioplastied coronary artery, provide structural support to the arterial wall. This support is helpful in maintaining vessel lumen patency. In two randomized clinical trials, stents increased angiographic success after PTCA, by increasing minimal lumen diameter and reducing, (but not eliminating), the incidence of restenosis at 6 months (Serruys et al., 1994; Fischman et al., 1994).
  • heparin coated stents appear to possess the same benefit of reduction in stenosis diameter at follow-up as was observed with non-heparin coated stents. Heparin coating also appears to have the added benefit of producing a reduction in sub-acute thrombosis after stent implantation (Serruys et al., 1996).
  • sustained mechanical expansion of a stenosed coronary artery with a stent has been shown to provide some measure of restenosis prevention
  • coating of stents with heparin has demonstrated both the feasibility and the clinical usefulness of delivering drugs locally, at the site of injured tissue.
  • Post-angioplasty restenosis is a multifactorial process that involves numerous interactive mechanisms. This means that effective prevention of restenosis may not be feasible with agents possessing a single mechanism of action; positive therapeutic results may be best achieved through application of several agents with differing therapeutic targets. Thus, potential therapeutic benefit could be found with the co-delivery of agents with different mechanisms of action targeting different components of the restenosis process.
  • the current invention comprises an approach to solving the clinical problem of restenosis, which involves the administration of drug combinations, either locally or systemically.
  • a combination would be the addition of the antiinflammatory corticosteroid, dexamethasone, with an antiproliferative agent such as cladribine, rapamycin, vincristine, taxol, or a nitric oxide donor.
  • an antiproliferative agent such as cladribine, rapamycin, vincristine, taxol, or a nitric oxide donor.
  • Such combination therapies might result in a better therapeutic effect (less proliferation as well as less inflammation, a stimulus for proliferation) than would occur with either agent alone.
  • Such agents could be administered systemically in their respective therapeutic doses, or, alternatively, could be bound to the surface of a stent by means of incorporation within either a biodegradable or biostable polymeric coating.
  • these agents could be incorporated into a stent constructed with a grooved reservoir.
  • delivery of a stent containing both an antiproliferative agent and an anti-inflammatory agent to a coronary artery injured during the process of angioplasty would provide the added therapeutic benefit of 1) limiting the degree of local smooth muscle cell proliferation; 2) reducing a stimulus for proliferation, i.e., inflammation, and thus enhance the restenosis-limiting action of the stent.
  • growth factor or cytokine signal transduction inhibitor such as the ras inhibitor, R115777, or a tyrosine kinase inhibitor, such as tyrphostin
  • an antiproliferative agent such as taxol, vincristine or rapamycin so that proliferation of SMC could be inhibited by different mechanisms.
  • an antiproliferative agent such as taxol, vincristine or rapamycin could be combined with an inhibitor or extracellular matrix synthesis such as halofuginone.
  • agents acting by different mechanisms could act synergistically to reduce SMC proliferation and vascular hyperplasia.
  • This invention is also intended to cover other combinations of two or more such drug agents.
  • such agents could be administered systemically, delivered locally via drug delivery catheter, or formulated for delivery from the surface of a stent, or given as a combination or systemic and local therapy.
  • FIGS. 1 and 1 a are top views and section views of a stent containing reservoirs as described in the present invention
  • FIGS. 2 a and 2 b are similar views of an alternate embodiment of the stent with open ends
  • FIGS. 3 a and 3 b are further alternate figures of a device containing a grooved reservoir
  • FIG. 4 is a layout view of a device containing a reservoir as in FIG. 3 ;
  • FIGS. 5 and 6 are a graph of the performance characteristics of stents coated according to this invention.
  • FIGS. 7 , 8 and 9 are additional release diagrams displaying results of various tests performed on stents made in accordance with the disclosure.
  • implantation of a coronary stent in conjunction with balloon angioplasty is highly effective in treating acute vessel closure and may reduce the risk of restensosis.
  • Intravascular ultrasound studies suggest that coronary stenting effectively prevents vessel constriction and that most of the late luminal loss after stent implantation is due to plaque growth, probably related to neointimal hyperplasia.
  • the late luminal loss after coronary stenting is almost two times higher than that observed after conventional balloon angioplasty.
  • a combination of agents, which prevent inflammation and proliferation, or prevents proliferation by multiple mechanisms, combined with a stent may provide the most efficacious by multiple mechanisms, combined with a stent may provide the most efficacious treatment for post-angioplasty restenosis.
  • a stent in conjunction with systemic treatment with the drug combinations suggested above or local delivery of such drug combinations is an attractive treatment. Either systemic or local delivery of multiple drugs from a stent has the following advantages:
  • Combination therapy is therefore a means of improving the therapeutic ratio (efficacy/toxicity) of an antirestenosis agent.
  • any stent strut 10 , 20 , 30 can be modified to have a certain reservoir 11 , 21 , 31 .
  • Each of these reservoirs can be open or closed as desired. These reservoirs can hold the drug to be delivered.
  • FIG. 4 shows a stent 40 with a reservoir 45 created at the apex of a flexible connector.
  • this reservoir 45 is intended to be useful to delivery any drug at a specific point of flexibility of the stent. Accordingly, this concept can be useful for “second generation” type stents.
  • the reservoir size in the stent struts must be kept at a size of about 0.1 mm to about 1 mm depth, and 7 mm to 15 mm length or enough to hold at least a therapeutic amount of the drug. Then, it should be possible to adequately apply the drug dosage at the desired location and in the desired amount.
  • human smooth muscle cells (Clonetics, Walkersville, Md.) were seeded at a density of 10,000 cells/well) into each well of 24-well plates and cultured in growth medium containing herapin, EGF (epidermal growth factor), FGF (fibroblast growth factor) and serum. After 24 hours, the growth medium was changed and fresh medium containing various concentrations of test agents (0.01-10 mcg/mL) were added to triplicate wells. Medium was replaced with fresh medium (plus test agents) after 3 days. On day five, cells were detached by trypsin/EDTA and counted using a hemacytometer. Cell viability was assessed by trypan blue exclusion.
  • Table 1 provides the percent of control growth of the various tested concentrations of the anti-inflammatory agent, dexamethasone, on human smooth muscle cells, either in the absence or presence of 2 concentrations of the antiproliferative/antiimmune agent, rapamycin.
  • Dexamethasone produced a concentration-related decrease in the proliferation of smooth muscle cells in this model system.
  • the IC 50 value concentration required to produce a reduction in proliferation by 50% of the control cell count
  • the IC 50 value concentration required to produce a reduction in proliferation by 50% of the control cell count
  • rapamycin concentration was increased in the incubation media, less dexamethasone was required to produce a 50% inhibition of cell growth. Since the amounts of rapamycin employed did not achieve a 50% inhibition of cell growth, Table 1 demonstrates that concentrations of both rapamycin or dexamethasone below their respective IC 50 amounts may combine to produce an effect on cell growth greater than either agent individually. Such a drug combination may be therapeutically useful for inhibition of the intimal smooth muscle cell proliferation that accompanies stent implantation. While efficacy could be maintained at these lower doses, toxicities associated with each of these agents might be ameliorated.
  • Base coat Rapamycin mixed with polymer.
  • Overcoat Dexamethasone mixed with polymer.
  • Base coat Dexamethasone mixed with polymer.
  • This example describes the preparation of a base coating that contains rapamycin.
  • Stents were coated with Parylene CTM using a vapor deposition method provided by the manufacturer of the parylene-coating instrument (SCS Madison, Wis.). The stent is weighed and then mounted for coating. While the stent is rotating a solution of 1.75 mg/ml Poly (ethylene-covinyl acetate) (PEVA), 1.75 mg/ml polybutyl methacrylate, and 1.5 mg/ml rapamycin dissolved in tetrahydrofuran is sprayed onto it. The coated stent is removed from the spray and allowed to air-dry. After a final weighing the amount of coating on the stent is determined.
  • PEVA poly (ethylene-covinyl acetate)
  • rapamycin dissolved in tetrahydrofuran is sprayed onto it.
  • the coated stent is removed from the spray and allowed to air-dry. After a final weighing the amount of coating on the stent is determined.
  • This example describes the preparation of a base coating that contains dexamethasone.
  • Stents were coated with Parylene CTM using a vapor deposition method provided by the manufacturer of the parylene-coating instrument (SCS Madison, Wis.). The stent is weighed and then mounted for coating. While the stent is rotating a solution of 1.75 mg/ml Poly (ethylene-covinyl acetate) (PEVA), 1.75 mg/ml polybutyl methacrylate, and 1.5 mg/ml dexamethasone dissolved in tetrahydrofuran is sprayed onto it. The coated stent is removed from the spray and allowed to air-dry. After a final weighing the amount of coating on the stent is determined.
  • PEVA poly (ethylene-covinyl acetate)
  • dexamethasone dissolved in tetrahydrofuran is sprayed onto it.
  • the coated stent is removed from the spray and allowed to air-dry. After a final weighing the amount of coating on the stent is determined.
  • This example describes the preparation of a base coating that contains rapamycin and dexamethasone.
  • Stents were coated with Parylene CTM using a vapor deposition method provided by the manufacturer of the parylene-coating instrument (SCS Madison, Wis.). The stent is weighed and then mounted for coating. While the stent is rotating a solution of 1.75 mg/ml Poly (ethylene-covinyl-acetate) (PEVA), 1.75 mg/ml polybutyl methacrylate, 0.75 mg/ml rapamycin and 0.75 mg/ml dexamethasone dissolved in tetrahydrofuran is sprayed onto it. The coated stent is removed from the spray and allowed to air-dry. After a final weighing the amount of coating on the stent is determined.
  • PEVA poly (ethylene-covinyl-acetate)
  • This example describes a stent coating that consists of a base coat containing rapamycin and dexamethasone and a drug-free barrier overcoat.
  • a stent is coated as in Example 4. After the coating is thoroughly dried a solution of 2.5 mg/ml polybutyl methacrylate dissolved in tetrahydrofuran is sprayed onto it. It is then air-dried for a final overcoat weight of 150 ⁇ g.
  • This example describes a stent coating, which consists of a base containing rapamycin and an overlayer with dexamethasone.
  • a stent is coated as in Example 2.
  • a solution of 1.75 mg/ml Poly (ethylene-covinyl-acetate) (PEVA), 1.75 mg/ml polybutyl methacrylate, and 1.5 mg/ml dexamethasone dissolved in tetrahyrdofuran is sprayed onto it.
  • the coated stent is removed from the spray and allowed to air-dry.
  • the final weight of each layer is typically 250 ⁇ g for a total coating weight of 500 ⁇ g.
  • This example describes a stent coating, which consists of a base containing dexamethasone and an overlayer with rapamycin.
  • a stent is coated as in Example 3.
  • a solution of 1.75 mg/ml Poly (ethylene-covinyl-acetate) (PEVA), 1.75 mg/ml polybutyl methacrylate, and 1.5 mg/ml rapamycin dissolved in tetrahydrofuran is sprayed onto it.
  • the coated stent is removed from the spray and allowed to air-dry.
  • the final weight of each layer is typically 250 ⁇ g for a total coating weight of 500 ⁇ g.
  • This example describes the method for performing the in vitro release of rapamycin and dexamethasone from coated stent.
  • Each stent was placed in a 2.5 ml of release medium (aqueous ethanol, 25 percent by volume at room temperature) contained in a 13 ⁇ 100 mm culture tube with a screw cap. The tube was shaken in a water bath (INNOVATM 3100, New Brunswick Scientific) at 200 rpm while maintaining ambient conditions. After a given time interval (ranging from 15 minutes to one day) the tubes were removed from the shaker and the respective stents carefully transferred to a fresh 2.5 ml Aliquot of release medium. The new tube was placed on the shaker and agitation resumed. A sample was removed from the aliquot, which had previously contained the stent and placed in a HPLC vial for determination of the rapamycin content and dexamethasone, by HPLC.
  • release medium aqueous ethanol, 25 percent by volume at room temperature
  • This example describes the method for analyzing the release medium for rapamycin.
  • the HPLC system used to analyze the samples was a Waters Alliance with a PDA 996. This system is equipped with a photodiode array detector. 204 of each sample was withdrawn and analyzed on a C 18 -reverse phase column (Waters SymmetryTM Column: 4.6 mm ⁇ 100 mm RP 18 3.5 ⁇ m with a matching guard column) using a mobile phase consisting of acetonitrile/methanol/water (38:34:28 v/v) delivered at a flow rate of 1.2 mL/min. The column was maintained at 60° C. through the analysis. Under these analytical conditions rapamycin had a retention time of 4.75 ⁇ 0.1 minutes. The concentration was determined from a standard curve of concentration versus response (area-under the curve) generated from rapamycin standards in the range of from 50 mg/mL to 50 ⁇ g/mL.
  • This example describes the method for analyzing the release medium for dexamethasone.
  • the HPLC system used to analyze the samples was a Shimadzu Class-VP Chromatography Laboratory System. This system is equipped with a photodiode array detector. 204 of each sample was withdrawn and analyzed on a C 18 -reverse phase column (Waters SymmetryTM Column: 4.6 mm ⁇ 100 mm RP 18 3.5 ⁇ ). An isocratic mobile phase consisting of methanol/water (55:45 v/v) delivered at a flow rate of 0.8 mL/min. was used for the first 6.5 mins of analysis followed by 100% methanol for 2 minutes; the latter was to ensure removal of rapamycin which is retained on the column. The column was maintained at 25° C. throughout the analysis.
  • dexamethasone had a retention time of 5.9 ⁇ 0.1 minutes.
  • the concentration was determined from a standard curve of concentration versus response (area-under the curve) generated from dexamethasone standards in the range of from 40 mg/mL to 4.0 ⁇ g/mL.

Abstract

The current invention comprises an approach to solving the clinical problem of restenosis, which involves the administration of combinations of drugs to patients undergoing PTCA or stent implantation. In one embodiment of the invention, an antiproliferative agent such as rapamycin, vincristine or taxol is administered in combination with the anti-inflammatory agent, dexamethasone, to patients systemically, either subcutaneously or intravenously. In another embodiment of the invention, the antiproliferative and anti-inflammatory agents are bound in a single formulation to the surface of a stent by means of incorporation within either a biodegradeable or biostable polymeric coating. Alternatively, such drug combinations could be incorporated into a stent constructed with a grooved reservoir.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application claims priority from application Ser. No. 09/575,480 filed May 19, 2000, now U.S. Pat. No. 8,029,561; which claims priority from a Provisional Application of the same title, Application No. 60/204,417, filed May 12, 2000.
  • FIELD OF THE INVENTION
  • This invention describes the delivery of different drug combinations, either systemically or locally, particularly from an intravascular stent, directly from micropores in the stent body or mixed or bound to a polymer coating applied on stent, to inhibit neointimal tissue proliferation and thereby prevent restenosis. This invention given either systemically or locally also facilitates the performance of the stent in inhibiting restenosis.
  • BACKGROUND OF THE INVENTION
  • Atherosclerotic lesions, which limit or obstruct coronary blood flow, are the major cause of ischemic heart disease related mortality, resulting in 500,000-600,000 deaths annually. Percutaneous transluminal coronary angioplasty (PTCA) to open the obstructed artery was performed in over 550,000 patients in the U.S. and 945,000+ patients worldwide in 1996 (Lemaitre et al., 1996). A major limitation of this technique is the problem of post-PTCA closure of the vessel, both immediately after PTCA (acute occlusion) and in the long term (restenosis): 30% of patients with subtotal lesions and 50% of patients with chronic total lesions will go on to restenosis after angioplasty. Additionally, restenosis is a significant problem in patients undergoing saphenous vein bypass graft. The mechanism of acute occlusion appears to involve several factors and may result from vascular recoil with resultant closure of the artery and/or deposition of blood platelets along the damaged length of the newly opened blood vessel followed by formation of a fibrin/red blood cell thrombus.
  • Restenosis after angioplasty is a more gradual process and involves initial formation of a subcritical thrombosis with release from adherent platelets of cell derived growth factors with subsequent proliferation of intimal smooth muscle cells and local infiltration of inflammatory cells contributing to vascular hyperplasia. It is important to note that multiple processes, among those including thrombosis, cell proliferation, cell migration and inflammation each seem to contribute to the restenotic process.
  • In the U.S., a 30-50% restenosis rate translates to 120,000-200,000 U.S. patients at risk from restenosis. If only 80% of such patients elect repeat angioplasty (with the remaining 20% electing coronary artery bypass graft) is added to the cost of coronary artery bypass graft for the remaining 20%, the total cost for restenosis easily reaches into billions of dollars. Thus, successful prevention of restenosis could result not only in significant therapeutic benefit but also in significant health care savings.
  • While the exact mechanism for restenosis is still uncertain, the general aspects of the restenosis process have been identified:
      • 1) In the normal arterial wall, smooth muscle cells (SMC) proliferate at a low rate (<0.1%/day). SMC in vessel wall exists in a ‘contractile’ phenotype characterized by 80-90% of the cell cytoplasmic volume occupied with the contractile apparatus. Endoplasmic reticulum, Golgi, and free ribosomes are few and located in the perinuclear region. Extracellular matrix surrounds SMC and is rich in heparin-like glycosylaminoglycans which are believed to be responsible for maintaining SMC in the contractile phenotype state (Campbell and Campbell, 1985).
      • 2) Upon pressure expansion of an intracoronary balloon catheter during angioplasty, smooth muscle cells within the arterial wall become injured, initiating a thrombotic and inflammatory response. Cell derived growth factors such as platelet derived growth factor (PDGF), basic fibroblast growth factor (bfGF), epidermal growth factor (EGF), thrombin, etc., released from platelets (i.e., PDGF) adhering to the damaged arterial luminal surface, invading macrophages and/or leukocytes, or directly from SMC (i.e., bFGF) provoke a proliferation and migratory response in medial SMC. These cells undergo a phenotypic change from the contractile phenotype to a ‘synthetic’ phenotype characterized by only few contractile filament bundles but extensive rough endoplasmic reticulum, Golgi and free ribosomes. Proliferation/migration usually begins with 1-2 days post-injury and peaks at 2 days in the media, declining thereafter (Campbell and Campbell, 1987; Clowes and Schwartz, 1985).
      • 3) Daughter synthetic cells migrate to the intimal layer of arterial smooth muscle and continue to proliferate and begin to secrete significant amounts of extracellular matrix proteins. Proliferation, migration and inflammation continue until the damaged luminal endothelial layer regenerates at which time proliferation slows within the intima, usually within 7-14 days postinjury. The further increase in intimal thickening that occurs over the next 3-6 months is due primarily to an increase in extracellular matrix rather than cell number. Thus, SMC migration and proliferation is an acute response to vessel injury while intimal hyperplasia is a more chronic response. (Liu et al., 1989).
      • 4) Simultaneous with local proliferation and migration, inflammatory cells adhere to the site of vascular injury. Within 3-7 days post injury, luminal adherent cells decline due to migration of inflammatory to the deeper layers of the vessel wall. In animal models employing either balloon injury or stent implantation, inflammatory cells may persist at the site of vascular injury for at least 30 days (Tanaka et al., 1993; Edelman et al., 1998). Inflammatory cells therefore are present and may contribute to both the acute and chronic phases of restenosis.
  • Numerous agents have been examined for presumed antiproliferative actions in restenosis and have shown activity in experimental animal models. Some of the agents which have been shown to successfully reduce the extent of intimal hyperplasia in animal models include: heparin and heparin fragments (Clowes, A. W. and Karnovsky M., Nature, 265: 25-26, 1977; Guyton, J. R. et al., Circ. Res., 46: 625-634, 1980; Clowes, A. W. and Clowes, M. M., Lab. Invest. 52: 611-616, 1985; Clowes, A. W. and Clowes, M. M., Circ. Res. 58: 839-845, 1986; Majesky et al., Circ. Res. 61: 296-300, 1985; Snow et al., Am. J. Pathol. 137: 313-330, 1990; Okada, T. et al., Neurosurgery 25: 92-98, 1989), colchicine (Currier, J. W. et al., Circulation 80: 11-66, 1989, taxol (Sollott, S. J. et al., J. Clin. Invest. 95: 1869-1876, 1995), angiotensin converting enzyme (ACE) inhibitors (Powell, J. S. et al., Science, 245: 186-188, 1989), angiopeptin (Lundergan, C. F. et al., Am. J. Cardiol. 17 (Suppl. B); 132B-136B, 1991), cyclosporin A (Jonasson, L. et al., Proc. Natl., Acad. Sci., 85: 2303, 1988), goat-anti-rabbit PDGF antibody (Ferns, G. A. A. et al., Science 253: 1129-1132, 1991), terbinafine (Nemecek, G. M. et al., J. Pharmacol. Exp. Thera. 248: 1167-1174, 1989), trapidil (Liu, M. W. et al., Circulation 81: 1089-1093, 1990), tranilast (Fukuyama, J. et al., Eur. J. Pharmacol, 318: 327-332, 1996), interferon-gamma (Hansson, G. K. and Holm, J., Circulation 84: 1266-1272, 1991), rapamycin (Marx, S. O. et al., Circ. Res. 76: 412-417, 1995), steroids (Colburn, M. D. et al., J. Vasc. Surg. 15: 510-518, 1992), see also Berk, B. C. et al., J. Am. Coll. Cardiol. 17: 111B-117B, 1991), ionizing radiation (Weinberger, J. et al., Int. J. Rad. Onc. Biol. Phys. 36: 767-775, 1996), fusion toxins (Farb, A. et al., Circ. Res. 80: 542-550, 1997) antisense oligonucleotides (Simons, M. et al., Nature 359: 67-70, 1992) and gene vectors (Chang, M. W. et al., J. Clin. Invest. 96: 2260-2268, 1995). Antiproliferative action on SMC in vitro has been demonstrated for many of these agents, including heparin and heparin conjugates, taxol, tranilast, colchicine, ACE inhibitors, fusion toxins, antisense oligonucleotides, rapamycin and ionizing radiation. Thus, agents with diverse mechanisms of SMC inhibition may have therapeutic utility in reducing intimal hyperplasia.
  • However, unlike animal models, attempts in human angioplasty patients to prevent restenosis by systemic pharmacologic means have thus far been unsuccessful. Neither aspirin-dipyrdamole, ticlopidine, anticoagulant therapy (acute heparin, chronic warfarin, hirudin or hirulog), thromboxane receptor antagonism nor steroids have been effective in preventing restenosis, although platelet inhibitors have been effective in preventing acute reocclusion after angioplasty (Mak and Topol, 1997; Lang et al., 1991; Popma et al., 1991). Additionally, the 7E3 humanized monoclonal antibody fragment to the platelet GP IIb/IIIa receptor is still under study but has not shown promising results for the reduction in restenosis following angioplasty and stenting. Other agents, which have also been unsuccessful in the prevention of restenosis, include the calcium channel antagonists, prostacyclin mimetics, angiotensin converting enzyme inhibitors, serotonin receptor antagonists, and antiproliferative agents. These agents must be given systemically, however, and attainments of a therapeutically effective dose may not be possible; antiproliferative (or anti-restenosis) concentrations may exceed the known toxic concentrations of these agents so that levels sufficient to produce smooth muscle inhibition may not be reached (Mak and Topol, 1997; Lang et al., 1991; Popma et al., 1991).
  • Additional clinical trials in which the effectiveness for preventing restenosis of dietary fish oil supplements or cholesterol lowering agents has been examined have shown either conflicting or negative results so that no pharmacological agents are as yet clinically available to prevent post-angioplasty restenosis (Mak and Topol, 1997; Franklin and Faxon, 1993; Serruys, P. W. et al., 1993). Recent observations suggest that the antilipid/antioxidant agent, probucol may be useful in preventing restenosis but this work requires confirmation (Tardif et al., 1997; Yokoi et al., 1997). Probucol is presently not approved for use in the United States and a 30-day pretreatment period would preclude its use in emergency angioplasty. Additionally, application of ionizing radiation has shown significant promise in reducing or preventing restenosis after angioplasty in patients with stents (Teirstein et al., 1997). Currently, however, the most effective treatments for restenosis are repeat angioplasty, atherectomy or coronary artery bypass grafting, because no therapeutic agents currently have US Federal Regulatory Agency (FDA) approval for use for the preventing of post-angioplasty restenosis.
  • Unlike systemic pharmacologic therapy, stents have proven useful in partially preventing restenosis. Stents, are balloon-expandable slotted metal tubes (usually, but not limited to, stainless steel), which, when expanded within the lumen of an angioplastied coronary artery, provide structural support to the arterial wall. This support is helpful in maintaining vessel lumen patency. In two randomized clinical trials, stents increased angiographic success after PTCA, by increasing minimal lumen diameter and reducing, (but not eliminating), the incidence of restenosis at 6 months (Serruys et al., 1994; Fischman et al., 1994).
  • Additionally, in a preliminary trial, heparin coated stents appear to possess the same benefit of reduction in stenosis diameter at follow-up as was observed with non-heparin coated stents. Heparin coating also appears to have the added benefit of producing a reduction in sub-acute thrombosis after stent implantation (Serruys et al., 1996). Thus, 1) sustained mechanical expansion of a stenosed coronary artery with a stent has been shown to provide some measure of restenosis prevention, and 2) coating of stents with heparin has demonstrated both the feasibility and the clinical usefulness of delivering drugs locally, at the site of injured tissue.
  • Post-angioplasty restenosis is a multifactorial process that involves numerous interactive mechanisms. This means that effective prevention of restenosis may not be feasible with agents possessing a single mechanism of action; positive therapeutic results may be best achieved through application of several agents with differing therapeutic targets. Thus, potential therapeutic benefit could be found with the co-delivery of agents with different mechanisms of action targeting different components of the restenosis process.
  • SUMMARY OF THE INVENTION
  • The current invention comprises an approach to solving the clinical problem of restenosis, which involves the administration of drug combinations, either locally or systemically. One example of such a combination would be the addition of the antiinflammatory corticosteroid, dexamethasone, with an antiproliferative agent such as cladribine, rapamycin, vincristine, taxol, or a nitric oxide donor. Such combination therapies might result in a better therapeutic effect (less proliferation as well as less inflammation, a stimulus for proliferation) than would occur with either agent alone. Such agents could be administered systemically in their respective therapeutic doses, or, alternatively, could be bound to the surface of a stent by means of incorporation within either a biodegradable or biostable polymeric coating. Alternatively, these agents could be incorporated into a stent constructed with a grooved reservoir. Thus, delivery of a stent containing both an antiproliferative agent and an anti-inflammatory agent to a coronary artery injured during the process of angioplasty would provide the added therapeutic benefit of 1) limiting the degree of local smooth muscle cell proliferation; 2) reducing a stimulus for proliferation, i.e., inflammation, and thus enhance the restenosis-limiting action of the stent.
  • In other embodiments of the inventions, growth factor or cytokine signal transduction inhibitor, such as the ras inhibitor, R115777, or a tyrosine kinase inhibitor, such as tyrphostin, might be combined with an antiproliferative agent such as taxol, vincristine or rapamycin so that proliferation of SMC could be inhibited by different mechanisms. Alternatively, an antiproliferative agent such as taxol, vincristine or rapamycin could be combined with an inhibitor or extracellular matrix synthesis such as halofuginone. In the above cases, agents acting by different mechanisms could act synergistically to reduce SMC proliferation and vascular hyperplasia. This invention is also intended to cover other combinations of two or more such drug agents. As mentioned above, such agents could be administered systemically, delivered locally via drug delivery catheter, or formulated for delivery from the surface of a stent, or given as a combination or systemic and local therapy.
  • DETAILED DESCRIPTION OF THE DRAWINGS
  • The invention will be better understood in connection with the following figures in which:
  • FIGS. 1 and 1 a are top views and section views of a stent containing reservoirs as described in the present invention;
  • FIGS. 2 a and 2 b are similar views of an alternate embodiment of the stent with open ends;
  • FIGS. 3 a and 3 b are further alternate figures of a device containing a grooved reservoir;
  • FIG. 4 is a layout view of a device containing a reservoir as in FIG. 3;
  • FIGS. 5 and 6 are a graph of the performance characteristics of stents coated according to this invention; and
  • FIGS. 7, 8 and 9 are additional release diagrams displaying results of various tests performed on stents made in accordance with the disclosure.
  • DETAILED DESCRIPTION OF THE INVENTION Multiple Drug Therapy Combined with a Stent
  • As stated previously, implantation of a coronary stent in conjunction with balloon angioplasty is highly effective in treating acute vessel closure and may reduce the risk of restensosis. Intravascular ultrasound studies (Mintz et al., 1996) suggest that coronary stenting effectively prevents vessel constriction and that most of the late luminal loss after stent implantation is due to plaque growth, probably related to neointimal hyperplasia. The late luminal loss after coronary stenting is almost two times higher than that observed after conventional balloon angioplasty. Thus, inasmuch as stents prevent at least a portion of the restenosis process, a combination of agents, which prevent inflammation and proliferation, or prevents proliferation by multiple mechanisms, combined with a stent may provide the most efficacious by multiple mechanisms, combined with a stent may provide the most efficacious treatment for post-angioplasty restenosis. In this regard, a stent in conjunction with systemic treatment with the drug combinations suggested above or local delivery of such drug combinations is an attractive treatment. Either systemic or local delivery of multiple drugs from a stent has the following advantages:
  • 1. Prevention of vessel recoil remodeling through the scaffolding action of the stent;
    2. Prevention of multiple components of neointimal hyperplasia, the vascular response to injury
  • Local administration of drug combinations to stented coronary arteries might have additional therapeutic benefit:
      • 1) higher tissue concentrations would be achievable than would occur with systemic administration;
      • 2) reduced systemic toxicity; and
      • 3) single treatment/ease of administration
  • An additional benefit of combination drug therapy may be to reduce the dose of each of the therapeutic components and thus limiting their toxicity, while still achieving a reduction in restenosis. Combination therapy is therefore a means of improving the therapeutic ratio (efficacy/toxicity) of an antirestenosis agent.
  • As seen in the accompanying FIGS. 1-4, it is possible to modify currently manufactured stents in order to provide adequate drug delivery. As seen in FIGS. 1 a, 2 a and 3 a, any stent strut 10, 20, 30 can be modified to have a certain reservoir 11, 21, 31. Each of these reservoirs can be open or closed as desired. These reservoirs can hold the drug to be delivered. FIG. 4 shows a stent 40 with a reservoir 45 created at the apex of a flexible connector. Of course, this reservoir 45 is intended to be useful to delivery any drug at a specific point of flexibility of the stent. Accordingly, this concept can be useful for “second generation” type stents. Processes for coating such stents are described, for instance, in Ser. No. 09/061,568, filed 16 Apr. 1998, and 09/512,432 filed 25 Feb. 2000, both of which are assigned to a common assignee and are incorporated herein by reference.
  • In any of the foregoing devices, however, it is useful to have the drug dosage applied with enough specificity and a sufficient concentration to provide an effective dosage in the lesion area. In this regard, the reservoir size in the stent struts must be kept at a size of about 0.1 mm to about 1 mm depth, and 7 mm to 15 mm length or enough to hold at least a therapeutic amount of the drug. Then, it should be possible to adequately apply the drug dosage at the desired location and in the desired amount.
  • Example 1
  • To assess the ability of a drug combination to prevent cell proliferation, human smooth muscle cells (Clonetics, Walkersville, Md.) were seeded at a density of 10,000 cells/well) into each well of 24-well plates and cultured in growth medium containing herapin, EGF (epidermal growth factor), FGF (fibroblast growth factor) and serum. After 24 hours, the growth medium was changed and fresh medium containing various concentrations of test agents (0.01-10 mcg/mL) were added to triplicate wells. Medium was replaced with fresh medium (plus test agents) after 3 days. On day five, cells were detached by trypsin/EDTA and counted using a hemacytometer. Cell viability was assessed by trypan blue exclusion.
  • Table 1 provides the percent of control growth of the various tested concentrations of the anti-inflammatory agent, dexamethasone, on human smooth muscle cells, either in the absence or presence of 2 concentrations of the antiproliferative/antiimmune agent, rapamycin. Dexamethasone produced a concentration-related decrease in the proliferation of smooth muscle cells in this model system. The IC50 value (concentration required to produce a reduction in proliferation by 50% of the control cell count) for the inhibition of smooth muscle cells with dexamethasone alone estimated from Table 1 is 5 μg/mL. Addition of 0.2 μg/mL of rapamycin to the incubation media was found to reduce the IC50 estimate of dexamethasone to 0.05 μgmL. A greater added concentration of rapamycin (2 μg/mL) further reduced the IC50 estimate for dexamethasone to less than 0.01 μg/mL.
  • Thus, as the rapamycin concentration was increased in the incubation media, less dexamethasone was required to produce a 50% inhibition of cell growth. Since the amounts of rapamycin employed did not achieve a 50% inhibition of cell growth, Table 1 demonstrates that concentrations of both rapamycin or dexamethasone below their respective IC50 amounts may combine to produce an effect on cell growth greater than either agent individually. Such a drug combination may be therapeutically useful for inhibition of the intimal smooth muscle cell proliferation that accompanies stent implantation. While efficacy could be maintained at these lower doses, toxicities associated with each of these agents might be ameliorated.
  • TABLE 1
    Inhibition of human vascular smooth muscle cell proliferation with
    dexamethasone or dexamethasone + rapamycin.
    Concentration of Dexamethasone (μg/ml)
    % of Control Growth 0 0.01 0.05 0.1 0.5 1 5 10 50 100
    Rapamycin 0 μg/ml 100.0 75.2 76.5 72.2 50.0 36.1 18.3 11.7
    Standard Deviation 4.2 0.8 16.3 9.3 7.6 5.9 6.0 1.3
    Rapamycin 0.2 μg/ml 85.7 63.4 57.6 49.7 48.9 48.2 41.2 31.1 31.2 29.0
    Standard Deviation 6.6 3.2 2.1 4.6 2.2 1.7 3.0 2.7 1.0 1.8
    Rapamycin 1 μg/ml 67.4 48.3 45.1 38.1 39.2 37.8 33.9 25.8 20.7 18.5
    Standard Deviation 2.6 3.3 13.3 9.5 4.4 4.5 3.1 8.1 6.4 3.7
  • The following examples are used to demonstrate the various configurations of medicated stent coatings containing one or more drugs. These are summarized in Table 2.
  • TABLE 2
    Coating configurations used to demonstrate controlled
    release of rapamycin and dexamethasone from a stent.
    Drug Content
    Sample I.D. Rapa Dexb Coating Configuration
    50/50  82 μg  82 μg Drugs are co-mixed with polymer.
    Total coating wt: 548 μg
     0/100  0 μg 100 μg Drugs are co-mixed with polymer.
    Total coating wt.: 641 μg
    100/0  150 μg  0 μg Drugs are co-mixed with polymer.
    Total coating wt.: 500 μg
    67/33 103  51 Drugs are co-mixed with polymer.
    Ttoal coating wt.: 513 μg
    33/67  53 107 Drugs are co-mixed with polymer.
    Total coating wt.: 534 μg
    33/67-3Xc 182 μg 363 μg Drugs are mixed with polymer.
    Total coating wt.: 1817 μg
    50/50-OLDd  77 μg  80 μg Base coat: Rapamycin mixed with
    polymer. Overcoat: Dexamethasone
    mixed with polymer. Total coating
    wt.: 520 μg
    50/50-OLRe  79 μg  81 μg Base coat: Dexamethasone mixed
    with polymer.
    Overcoat: Rapamycin mixed with
    polymer.
    Total coating wt.: 536 μg
    50/50-TCf 100 μg 100 μg Base coat: Drugs are mixed with
    polymer blend
    Barrier coat: 158 μg polybutyl
    methacrylate.
    Total coating wt.: 811 μg
    0/100-TC f  0 μg 196 μg Base coat: Drugs are mixed with
    polymer blend
    Barrier coat: 168 μg polybutyl
    methacrylate
    Total coating wt.: 839 μg
    aRapamycin;
    bDexamethasone;
    c3 time coating thickness;
    dDexamethasone overlayer;
    eRapamycin overlayer;
    fTop coated
  • Example 2
  • This example describes the preparation of a base coating that contains rapamycin.
  • Stents were coated with Parylene C™ using a vapor deposition method provided by the manufacturer of the parylene-coating instrument (SCS Madison, Wis.). The stent is weighed and then mounted for coating. While the stent is rotating a solution of 1.75 mg/ml Poly (ethylene-covinyl acetate) (PEVA), 1.75 mg/ml polybutyl methacrylate, and 1.5 mg/ml rapamycin dissolved in tetrahydrofuran is sprayed onto it. The coated stent is removed from the spray and allowed to air-dry. After a final weighing the amount of coating on the stent is determined.
  • Example 3
  • This example describes the preparation of a base coating that contains dexamethasone.
  • Stents were coated with Parylene C™ using a vapor deposition method provided by the manufacturer of the parylene-coating instrument (SCS Madison, Wis.). The stent is weighed and then mounted for coating. While the stent is rotating a solution of 1.75 mg/ml Poly (ethylene-covinyl acetate) (PEVA), 1.75 mg/ml polybutyl methacrylate, and 1.5 mg/ml dexamethasone dissolved in tetrahydrofuran is sprayed onto it. The coated stent is removed from the spray and allowed to air-dry. After a final weighing the amount of coating on the stent is determined.
  • Example 4
  • This example describes the preparation of a base coating that contains rapamycin and dexamethasone.
  • Stents were coated with Parylene C™ using a vapor deposition method provided by the manufacturer of the parylene-coating instrument (SCS Madison, Wis.). The stent is weighed and then mounted for coating. While the stent is rotating a solution of 1.75 mg/ml Poly (ethylene-covinyl-acetate) (PEVA), 1.75 mg/ml polybutyl methacrylate, 0.75 mg/ml rapamycin and 0.75 mg/ml dexamethasone dissolved in tetrahydrofuran is sprayed onto it. The coated stent is removed from the spray and allowed to air-dry. After a final weighing the amount of coating on the stent is determined.
  • Example 5
  • This example describes a stent coating that consists of a base coat containing rapamycin and dexamethasone and a drug-free barrier overcoat.
  • A stent is coated as in Example 4. After the coating is thoroughly dried a solution of 2.5 mg/ml polybutyl methacrylate dissolved in tetrahydrofuran is sprayed onto it. It is then air-dried for a final overcoat weight of 150 μg.
  • Example 6
  • This example describes a stent coating, which consists of a base containing rapamycin and an overlayer with dexamethasone.
  • A stent is coated as in Example 2. A solution of 1.75 mg/ml Poly (ethylene-covinyl-acetate) (PEVA), 1.75 mg/ml polybutyl methacrylate, and 1.5 mg/ml dexamethasone dissolved in tetrahyrdofuran is sprayed onto it. The coated stent is removed from the spray and allowed to air-dry. The final weight of each layer is typically 250 μg for a total coating weight of 500 μg.
  • Example 7
  • This example describes a stent coating, which consists of a base containing dexamethasone and an overlayer with rapamycin.
  • A stent is coated as in Example 3. A solution of 1.75 mg/ml Poly (ethylene-covinyl-acetate) (PEVA), 1.75 mg/ml polybutyl methacrylate, and 1.5 mg/ml rapamycin dissolved in tetrahydrofuran is sprayed onto it. The coated stent is removed from the spray and allowed to air-dry. The final weight of each layer is typically 250 μg for a total coating weight of 500 μg.
  • The following examples describe the method and results for testing the in vitro release of rapamycin and dexamethasone from coated stent.
  • Example 8
  • This example describes the method for performing the in vitro release of rapamycin and dexamethasone from coated stent.
  • Each stent was placed in a 2.5 ml of release medium (aqueous ethanol, 25 percent by volume at room temperature) contained in a 13×100 mm culture tube with a screw cap. The tube was shaken in a water bath (INNOVA™ 3100, New Brunswick Scientific) at 200 rpm while maintaining ambient conditions. After a given time interval (ranging from 15 minutes to one day) the tubes were removed from the shaker and the respective stents carefully transferred to a fresh 2.5 ml Aliquot of release medium. The new tube was placed on the shaker and agitation resumed. A sample was removed from the aliquot, which had previously contained the stent and placed in a HPLC vial for determination of the rapamycin content and dexamethasone, by HPLC.
  • Example 9
  • This example describes the method for analyzing the release medium for rapamycin.
  • The HPLC system used to analyze the samples was a Waters Alliance with a PDA 996. This system is equipped with a photodiode array detector. 204 of each sample was withdrawn and analyzed on a C18-reverse phase column (Waters Symmetry™ Column: 4.6 mm×100 mm RP18 3.5 μm with a matching guard column) using a mobile phase consisting of acetonitrile/methanol/water (38:34:28 v/v) delivered at a flow rate of 1.2 mL/min. The column was maintained at 60° C. through the analysis. Under these analytical conditions rapamycin had a retention time of 4.75±0.1 minutes. The concentration was determined from a standard curve of concentration versus response (area-under the curve) generated from rapamycin standards in the range of from 50 mg/mL to 50 μg/mL.
  • The results from testing the coated stents described above are shown in FIG. 5.
  • Example 10
  • This example describes the method for analyzing the release medium for dexamethasone.
  • The HPLC system used to analyze the samples was a Shimadzu Class-VP Chromatography Laboratory System. This system is equipped with a photodiode array detector. 204 of each sample was withdrawn and analyzed on a C18-reverse phase column (Waters Symmetry™ Column: 4.6 mm×100 mm RP18 3.5 μ). An isocratic mobile phase consisting of methanol/water (55:45 v/v) delivered at a flow rate of 0.8 mL/min. was used for the first 6.5 mins of analysis followed by 100% methanol for 2 minutes; the latter was to ensure removal of rapamycin which is retained on the column. The column was maintained at 25° C. throughout the analysis. Under these analytical conditions dexamethasone had a retention time of 5.9±0.1 minutes. The concentration was determined from a standard curve of concentration versus response (area-under the curve) generated from dexamethasone standards in the range of from 40 mg/mL to 4.0 μg/mL.
  • The results from testing the coated stents described above are shown in FIG. 6.
  • These and other concepts are disclosed herein. It would be apparent to the reader that modifications are possible to the stent or the drug dosage applied. In any event, however, any obvious modifications should be perceived to fall within the scope of the invention, which is to be realized from the attached claims and their equivalents.

Claims (7)

1-15. (canceled)
16. In combination:
a catheter for the delivery of drugs to a blood vessel lumen of a patient; and
a therapeutic dosage amount of the combination of an anti-proliferative agent for inhibiting smooth muscle cell growth comprising rapamycin or an analogue thereof and an anti-inflammatory agent for inhibiting smooth muscle growth, both said agents contained in therapeutic dosage amounts.
17. The combination of claim 16 wherein the anti-inflammatory agent comprises dexamethasone.
18. The combination of claim 16 wherein the combination of at least two agents further includes a growth factor or cytokine signal transduction inhibitor.
19. The combination of claim 16 wherein the combination of at least two agents further includes a tyrosine kinase inhibitor.
20. The combination of claim 16 wherein the combination of at least two agents further includes an inhibitor of extracellular matrix synthesis.
21. The combination of claim 20 wherein the inhibitor of extracellular matrix synthesis comprises halofuginone and the anti-proliferative agent is taken from a group consisting of rapamycin, taxol, or vincristine.
US13/227,002 2000-05-12 2011-09-07 Drug Combinations Useful for Prevention of Restenosis Abandoned US20120029475A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/227,002 US20120029475A1 (en) 2000-05-12 2011-09-07 Drug Combinations Useful for Prevention of Restenosis

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US20441700P 2000-05-12 2000-05-12
US09/575,480 US8029561B1 (en) 2000-05-12 2000-05-19 Drug combination useful for prevention of restenosis
US13/227,002 US20120029475A1 (en) 2000-05-12 2011-09-07 Drug Combinations Useful for Prevention of Restenosis

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/575,480 Division US8029561B1 (en) 1998-04-16 2000-05-19 Drug combination useful for prevention of restenosis

Publications (1)

Publication Number Publication Date
US20120029475A1 true US20120029475A1 (en) 2012-02-02

Family

ID=26899464

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/575,480 Expired - Fee Related US8029561B1 (en) 1998-04-16 2000-05-19 Drug combination useful for prevention of restenosis
US13/227,002 Abandoned US20120029475A1 (en) 2000-05-12 2011-09-07 Drug Combinations Useful for Prevention of Restenosis

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/575,480 Expired - Fee Related US8029561B1 (en) 1998-04-16 2000-05-19 Drug combination useful for prevention of restenosis

Country Status (12)

Country Link
US (2) US8029561B1 (en)
EP (1) EP1289576B1 (en)
JP (1) JP2003533493A (en)
AT (1) ATE298592T1 (en)
AU (3) AU6295701A (en)
BR (1) BR0110778A (en)
CA (1) CA2408606A1 (en)
DE (1) DE60111743T2 (en)
ES (1) ES2244622T3 (en)
MX (2) MXPA02011186A (en)
PT (1) PT1289576E (en)
WO (1) WO2001087372A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050249775A1 (en) * 2003-12-19 2005-11-10 Robert Falotico Intraluminal medical devices in combination with therapeutic agents
US20050270864A1 (en) * 2004-05-18 2005-12-08 Peter Poechmueller Memory cell arrangement having dual memory cells
US10182928B2 (en) 2013-04-16 2019-01-22 Kaneka Corporation Medical tubular body

Families Citing this family (117)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7445792B2 (en) 2003-03-10 2008-11-04 Abbott Laboratories Medical device having a hydration inhibitor
US6890546B2 (en) * 1998-09-24 2005-05-10 Abbott Laboratories Medical devices containing rapamycin analogs
US7399480B2 (en) 1997-09-26 2008-07-15 Abbott Laboratories Methods of administering tetrazole-containing rapamycin analogs with other therapeutic substances using medical devices
US20030129215A1 (en) * 1998-09-24 2003-07-10 T-Ram, Inc. Medical devices containing rapamycin analogs
US7378105B2 (en) 1997-09-26 2008-05-27 Abbott Laboratories Drug delivery systems, kits, and methods for administering zotarolimus and paclitaxel to blood vessel lumens
US8394398B2 (en) 1997-09-26 2013-03-12 Abbott Laboratories Methods of administering rapamycin analogs with anti-inflammatories using medical devices
US8257726B2 (en) 1997-09-26 2012-09-04 Abbott Laboratories Compositions, systems, kits, and methods of administering rapamycin analogs with paclitaxel using medical devices
US7357942B2 (en) 1997-09-26 2008-04-15 Abbott Laboratories Compositions, systems, and kits for administering zotarolimus and paclitaxel to blood vessel lumens
US8057816B2 (en) 1997-09-26 2011-11-15 Abbott Laboratories Compositions and methods of administering paclitaxel with other drugs using medical devices
US8257725B2 (en) 1997-09-26 2012-09-04 Abbott Laboratories Delivery of highly lipophilic agents via medical devices
US7208010B2 (en) * 2000-10-16 2007-04-24 Conor Medsystems, Inc. Expandable medical device for delivery of beneficial agent
US20060240070A1 (en) * 1998-09-24 2006-10-26 Cromack Keith R Delivery of highly lipophilic agents via medical devices
US7455853B2 (en) 1998-09-24 2008-11-25 Abbott Cardiovascular Systems Inc. Medical devices containing rapamycin analogs
US7960405B2 (en) 1998-09-24 2011-06-14 Abbott Laboratories Compounds and methods for treatment and prevention of diseases
US8257724B2 (en) 1998-09-24 2012-09-04 Abbott Laboratories Delivery of highly lipophilic agents via medical devices
US6790228B2 (en) 1999-12-23 2004-09-14 Advanced Cardiovascular Systems, Inc. Coating for implantable devices and a method of forming the same
CA2408997C (en) 2000-04-11 2008-08-05 Universitat Heidelberg Poly-tri-fluoro-ethoxypolyphosphazene coverings and films for medical devices
US7419678B2 (en) * 2000-05-12 2008-09-02 Cordis Corporation Coated medical devices for the prevention and treatment of vascular disease
US6534693B2 (en) 2000-11-06 2003-03-18 Afmedica, Inc. Surgically implanted devices having reduced scar tissue formation
US9080146B2 (en) 2001-01-11 2015-07-14 Celonova Biosciences, Inc. Substrates containing polyphosphazene as matrices and substrates containing polyphosphazene with a micro-structured surface
US6752829B2 (en) 2001-01-30 2004-06-22 Scimed Life Systems, Inc. Stent with channel(s) for containing and delivering a biologically active material and method for manufacturing the same
DK2269603T3 (en) * 2001-02-19 2015-08-24 Novartis Ag TREATMENT OF BREAST TUMORS WITH A RAPAMYCIN DERIVATIVE IN COMBINATION WITH EXEMESTAN
US7247313B2 (en) * 2001-06-27 2007-07-24 Advanced Cardiovascular Systems, Inc. Polyacrylates coatings for implantable medical devices
CA2457018C (en) 2001-08-17 2010-12-14 Polyzenix Gmbh Device based on nitinol, a process for its production and its use
IL147416A (en) * 2001-12-31 2008-11-26 Israel State Combined modalities for improved cancer treatment
KR20040076278A (en) * 2002-01-10 2004-08-31 노파르티스 아게 Drug delivery systems for the prevention and treatment of vascular diseases comprising rapamycin and derivatives thereof
CN101717410B (en) 2002-02-01 2015-04-29 阿里亚德医药股份有限公司 Phosphorus-containing compounds & uses thereof
AU2003220390A1 (en) * 2002-03-18 2003-10-08 Medtronic Ave Inc. Medical devices for delivering anti-proliferative compositions to anatomical sites at risk for restenosis
EP1523345A1 (en) * 2002-07-18 2005-04-20 Medtronic AVE Inc. Medical devices comprising a protein-tyrosine kinase inhibitor to inhibit restonosis
US7491233B1 (en) * 2002-07-19 2009-02-17 Advanced Cardiovascular Systems Inc. Purified polymers for coatings of implantable medical devices
DE10237571A1 (en) * 2002-08-13 2004-02-26 Biotronik Meß- und Therapiegeräte GmbH & Co. Ingenieurbüro Berlin Endovascular implant with active coating
WO2004017939A1 (en) * 2002-08-20 2004-03-04 Terumo Kabushiki Kaisha Medical instrument to be implanted in the body
JP4588986B2 (en) * 2002-08-20 2010-12-01 テルモ株式会社 Implantable medical device
JP2005537854A (en) 2002-09-06 2005-12-15 アボット・ラボラトリーズ Medical device comprising a hydration inhibitor
DE10244847A1 (en) * 2002-09-20 2004-04-01 Ulrich Prof. Dr. Speck Medical device for drug delivery
WO2004032947A1 (en) * 2002-10-09 2004-04-22 Unibioscreen S.A. Extract with anti-tumor and anti-poisonous activity
ATE374652T1 (en) 2002-10-22 2007-10-15 Medtronic Vascular Inc STENT WITH INTERMITTENT COATING
WO2004060283A2 (en) 2002-12-16 2004-07-22 Nitromed, Inc. Nitrosated and nitrosylated rapamycin compounds, compositions and methods of use
JP2004222953A (en) * 2003-01-22 2004-08-12 Kanegafuchi Chem Ind Co Ltd Indwelling stent
WO2004087045A2 (en) * 2003-03-28 2004-10-14 Kosan Biosciences, Inc. Devices, methods, and compositions to prevent restenosis
US7279002B2 (en) * 2003-04-25 2007-10-09 Boston Scientific Scimed, Inc. Cutting stent and balloon
US20050118344A1 (en) 2003-12-01 2005-06-02 Pacetti Stephen D. Temperature controlled crimping
US20050033417A1 (en) * 2003-07-31 2005-02-10 John Borges Coating for controlled release of a therapeutic agent
US20060286139A1 (en) * 2003-08-19 2006-12-21 Kadem Ai-Lamee Polymeric drug release system for medical devices
US8652502B2 (en) * 2003-12-19 2014-02-18 Cordis Corporation Local vascular delivery of trichostatin A alone or in combination with sirolimus to prevent restenosis following vascular injury
US7303758B2 (en) * 2004-01-20 2007-12-04 Cordis Corporation Local vascular delivery of mycophenolic acid in combination with rapamycin to prevent restenosis following vascular injury
US7806924B2 (en) 2004-02-18 2010-10-05 Cordis Corporation Implantable structures for local vascular delivery of cladribine in combination with rapamycin for restenosis
US8431145B2 (en) 2004-03-19 2013-04-30 Abbott Laboratories Multiple drug delivery from a balloon and a prosthesis
US7695731B2 (en) * 2004-03-22 2010-04-13 Cordis Corporation Local vascular delivery of etoposide in combination with rapamycin to prevent restenosis following vascular injury
US7875282B2 (en) * 2004-03-22 2011-01-25 Cordis Corporation Coated medical device for local vascular delivery of Panzem® in combination with rapamycin to prevent restenosis following vascular injury
US20050220836A1 (en) * 2004-03-31 2005-10-06 Robert Falotico Drug delivery device
US20050244459A1 (en) * 2004-04-06 2005-11-03 Dewitt David M Coating compositions for bioactive agents
US7976557B2 (en) 2004-06-23 2011-07-12 Boston Scientific Scimed, Inc. Cutting balloon and process
US8709469B2 (en) 2004-06-30 2014-04-29 Abbott Cardiovascular Systems Inc. Anti-proliferative and anti-inflammatory agent combination for treatment of vascular disorders with an implantable medical device
EP1773336A1 (en) * 2004-07-16 2007-04-18 Novartis AG Use of a steroid for enhancement of skin permeability
CN101052362A (en) * 2004-09-08 2007-10-10 株式会社钟化 Organism indwelling support
US9107850B2 (en) 2004-10-25 2015-08-18 Celonova Biosciences, Inc. Color-coded and sized loadable polymeric particles for therapeutic and/or diagnostic applications and methods of preparing and using the same
US9114162B2 (en) 2004-10-25 2015-08-25 Celonova Biosciences, Inc. Loadable polymeric particles for enhanced imaging in clinical applications and methods of preparing and using the same
US20210299056A9 (en) 2004-10-25 2021-09-30 Varian Medical Systems, Inc. Color-Coded Polymeric Particles of Predetermined Size for Therapeutic and/or Diagnostic Applications and Related Methods
US8066726B2 (en) 2004-11-23 2011-11-29 Boston Scientific Scimed, Inc. Serpentine cutting blade for cutting balloon
EP1877004A4 (en) * 2005-03-23 2012-01-25 Abbott Lab Compositions and methods of administering rapamycin analogs using medical devices for long-term efficacy
WO2006102359A2 (en) * 2005-03-23 2006-09-28 Abbott Laboratories Delivery of highly lipophilic agents via medical devices
AU2006270221B2 (en) 2005-07-15 2012-01-19 Micell Technologies, Inc. Polymer coatings containing drug powder of controlled morphology
US20090062909A1 (en) 2005-07-15 2009-03-05 Micell Technologies, Inc. Stent with polymer coating containing amorphous rapamycin
KR101492545B1 (en) * 2005-07-15 2015-02-12 미셀 테크놀로지즈, 인코포레이티드 Polymer coatings containing drug powder of controlled morphology
TWI469771B (en) * 2005-10-14 2015-01-21 Abbott Lab System for providing controlled release delivery of drugs for treatment of neointimal hyperplasia and pharmaceutical composition for reducing neointimal hyperplasia
WO2007046935A2 (en) * 2005-10-14 2007-04-26 Abbott Laboratories Compositions, systems, kits, and methods of administering rapamycin analogs with paclitaxel using medical devices
KR100778020B1 (en) 2005-10-24 2007-11-28 사회복지법인 삼성생명공익재단 Vascular stent which is specially designed for the multiple drug loading and better drug elution
US20070134163A1 (en) 2005-12-13 2007-06-14 Zhao Jonathon Z Radiographic contrasting agents and radio-opaque polymeric materials for medical devices
US10029034B2 (en) * 2005-12-15 2018-07-24 CARDINAL HEALTH SWITZERLAND 515 GmbH Drug-eluting articles with improved drug release profiles
BRPI0600275A (en) * 2006-01-03 2007-10-02 Brz Biotecnologia Ltda Coronary prosthesis releasing drug composition for prevention and treatment of restenosis and manufacturing process
US8043358B2 (en) * 2006-03-29 2011-10-25 Boston Scientific Scimed, Inc. Stent with overlap and high extension
ES2540059T3 (en) 2006-04-26 2015-07-08 Micell Technologies, Inc. Coatings containing multiple drugs
US7985441B1 (en) 2006-05-04 2011-07-26 Yiwen Tang Purification of polymers for coating applications
US9028859B2 (en) 2006-07-07 2015-05-12 Advanced Cardiovascular Systems, Inc. Phase-separated block copolymer coatings for implantable medical devices
US8088789B2 (en) 2006-09-13 2012-01-03 Elixir Medical Corporation Macrocyclic lactone compounds and methods for their use
US10695327B2 (en) 2006-09-13 2020-06-30 Elixir Medical Corporation Macrocyclic lactone compounds and methods for their use
EP2083834B1 (en) * 2006-09-13 2017-06-21 Elixir Medical Corporation Macrocyclic lactone compounds and methods for their use
CA2690539C (en) 2006-10-10 2014-10-07 Celonova Biosciences, Inc. Bioprosthetic heart valve with polyphosphazene
US9539593B2 (en) 2006-10-23 2017-01-10 Micell Technologies, Inc. Holder for electrically charging a substrate during coating
US9737642B2 (en) 2007-01-08 2017-08-22 Micell Technologies, Inc. Stents having biodegradable layers
US11426494B2 (en) 2007-01-08 2022-08-30 MT Acquisition Holdings LLC Stents having biodegradable layers
AU2008256684B2 (en) 2007-05-25 2012-06-14 Micell Technologies, Inc. Polymer films for medical device coating
US20090104240A1 (en) * 2007-10-19 2009-04-23 Abbott Cardiovascular Systems Inc. Dual Drug Formulations For Implantable Medical Devices For Treatment of Vascular Diseases
WO2009061787A1 (en) * 2007-11-05 2009-05-14 Nanocopoeia, Inc. Coated devices and method of making coated devices that reduce smooth muscle cell proliferation and platelet activity
US8216600B2 (en) 2007-11-14 2012-07-10 Cordis Corporation Polymeric materials for medical devices
FR2927815B1 (en) * 2008-02-21 2011-01-14 Hexacath MEDICAL DEVICE COMPRISING A NO PRECURSOR AGENT, SUCH AS L-ARGININE OR L-LYSINE, AND A PROTECTIVE AND / OR RETENTIVE LAYER THEREOF
FR2927814B1 (en) * 2008-02-21 2011-01-14 Hexacath IMPLANTABLE MEDICAL DEVICE COMPRISING THE OCTREOTIDE AND A PROTECTIVE AND / OR RETENTIVE LAYER THEREOF
FR2927813B1 (en) * 2008-02-21 2017-07-21 Hexacath MEDICAL DEVICE IMPLANTABLE ON A PROTECTIVE LAYER / RETENTION OF AN ACTIVE AGENT OR MEDICAMENT, ESPECIALLY WATER SOLUBLE
FR2927812B1 (en) * 2008-02-21 2011-09-23 Hexacath IMPLANTABLE MEDICAL DEVICE WITH DRUG AND PROTECTIVE OR RETENTIVE LAYER THEREOF
EP2249892B1 (en) 2008-02-21 2017-11-15 Hexacath Implantable medical device including a protection/retaining layer for an active ingredient or drug, in particular a water-soluble one
CA2721832C (en) 2008-04-17 2018-08-07 Micell Technologies, Inc. Stents having bioabsorbable layers
KR101104901B1 (en) 2008-05-23 2012-01-12 김영곤 manufacturing method of thermo-rod for active drug release the thermo-rod
JP2011528275A (en) 2008-07-17 2011-11-17 ミセル テクノロジーズ,インク. Drug delivery medical device
US9510856B2 (en) 2008-07-17 2016-12-06 Micell Technologies, Inc. Drug delivery medical device
US20100092534A1 (en) * 2008-10-10 2010-04-15 Medtronic Vascular, Inc. Combination Local Delivery Using a Stent
US20100161039A1 (en) * 2008-12-23 2010-06-24 Vipul Dave Adhesion promoting temporary mask for coated surfaces
US8834913B2 (en) 2008-12-26 2014-09-16 Battelle Memorial Institute Medical implants and methods of making medical implants
CN102481195B (en) 2009-04-01 2015-03-25 米歇尔技术公司 Drug delivery medical device
EP3366326A1 (en) 2009-04-17 2018-08-29 Micell Technologies, Inc. Stents having controlled elution
US8951595B2 (en) 2009-12-11 2015-02-10 Abbott Cardiovascular Systems Inc. Coatings with tunable molecular architecture for drug-coated balloon
WO2011097103A1 (en) 2010-02-02 2011-08-11 Micell Technologies, Inc. Stent and stent delivery system with improved deliverability
US8795762B2 (en) 2010-03-26 2014-08-05 Battelle Memorial Institute System and method for enhanced electrostatic deposition and surface coatings
WO2011133655A1 (en) 2010-04-22 2011-10-27 Micell Technologies, Inc. Stents and other devices having extracellular matrix coating
CA2805631C (en) 2010-07-16 2018-07-31 Micell Technologies, Inc. Drug delivery medical device
JP5985474B2 (en) * 2010-08-04 2016-09-06 メリル ライフ サイエンシズ ピーブィティ.エルティディ Process for the preparation of novel 42-O- (heteroalkoxyalkyl) rapamycin compounds with antiproliferative properties
WO2012166819A1 (en) 2011-05-31 2012-12-06 Micell Technologies, Inc. System and process for formation of a time-released, drug-eluting transferable coating
US10117972B2 (en) 2011-07-15 2018-11-06 Micell Technologies, Inc. Drug delivery medical device
US10188772B2 (en) 2011-10-18 2019-01-29 Micell Technologies, Inc. Drug delivery medical device
DE102012001188A1 (en) * 2012-01-24 2013-07-25 Qualimed Innovative Medizinprodukte Gmbh balloon catheter
US9220584B2 (en) 2012-03-30 2015-12-29 Abbott Cardiovascular Systems Inc. Treatment of diabetic patients with a stent and locally administered adjunctive therapy
US20130259921A1 (en) * 2012-03-30 2013-10-03 Abbott Cardiovascular Systems Inc. Treatment Of Diabetic Patients With A Stent And An Adjunctive Drug Formulation
US20130303496A1 (en) * 2012-05-08 2013-11-14 Abbott Cardiovascular Systems Inc. Method Of Treating Vascular Lesions
AU2014248508B2 (en) 2013-03-12 2018-11-08 Micell Technologies, Inc. Bioabsorbable biomedical implants
JP2016519965A (en) 2013-05-15 2016-07-11 マイセル・テクノロジーズ,インコーポレイテッド Bioabsorbable biomedical implant
EP2997977A1 (en) * 2014-09-19 2016-03-23 Fundación de la Comunidad Valenciana Centro de Investigación Principe Felipe Specific mtor inhibitors in the treatment of x-linked adrenoleukodystrophy
EP3629774A4 (en) * 2017-05-26 2021-03-03 Mercator Medsystems, Inc. Combination therapy for treatment of restenosis

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5516781A (en) * 1992-01-09 1996-05-14 American Home Products Corporation Method of treating restenosis with rapamycin
US6159488A (en) * 1997-08-14 2000-12-12 Agricultural Research Org. Ministry Of Agriculture (Gov.) Intracoronary stents containing quinazolinone derivatives
US6335029B1 (en) * 1998-08-28 2002-01-01 Scimed Life Systems, Inc. Polymeric coatings for controlled delivery of active agents
US6369039B1 (en) * 1998-06-30 2002-04-09 Scimed Life Sytems, Inc. High efficiency local drug delivery
US6368658B1 (en) * 1999-04-19 2002-04-09 Scimed Life Systems, Inc. Coating medical devices using air suspension
US6562829B1 (en) * 1997-05-23 2003-05-13 Hadasit Medical Research Services & Development Co., Ltd. Treatment of hepatic cirrhosis
US8592036B2 (en) * 2006-06-23 2013-11-26 Abbott Cardiovascular Systems Inc. Nanoshells on polymers

Family Cites Families (158)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1205743A (en) 1966-07-15 1970-09-16 Nat Res Dev Surgical dilator
US3657744A (en) 1970-05-08 1972-04-25 Univ Minnesota Method for fixing prosthetic implants in a living body
US4388735A (en) 1980-11-03 1983-06-21 Shiley Inc. Low profile prosthetic xenograft heart valve
AT392733B (en) 1981-09-16 1991-05-27 Medinvent Sa DEVICE FOR TREATING BLOOD VESSELS OR THE LIKE.
DE3205942A1 (en) 1982-02-19 1983-09-08 Ljubomir Dr. Skopje Vasilev Balloon-tipped catheter with a mobile tip which permits complete voiding of the bladder
US4503569A (en) 1983-03-03 1985-03-12 Dotter Charles T Transluminally placed expandable graft prosthesis
US5275622A (en) 1983-12-09 1994-01-04 Harrison Medical Technologies, Inc. Endovascular grafting apparatus, system and method and devices for use therewith
US4580568A (en) 1984-10-01 1986-04-08 Cook, Incorporated Percutaneous endovascular stent and method for insertion thereof
US4733665C2 (en) 1985-11-07 2002-01-29 Expandable Grafts Partnership Expandable intraluminal graft and method and apparatus for implanting an expandable intraluminal graft
US4907336A (en) 1987-03-13 1990-03-13 Cook Incorporated Method of making an endovascular stent and delivery system
US5041126A (en) 1987-03-13 1991-08-20 Cook Incorporated Endovascular stent and delivery system
US4800882A (en) 1987-03-13 1989-01-31 Cook Incorporated Endovascular stent and delivery system
US4969458A (en) 1987-07-06 1990-11-13 Medtronic, Inc. Intracoronary stent and method of simultaneous angioplasty and stent implant
US4990131A (en) 1987-09-01 1991-02-05 Herbert Dardik Tubular prostheses for vascular reconstructive surgery and process for preparing same
US5131908A (en) 1987-09-01 1992-07-21 Herbert Dardik Tubular prosthesis for vascular reconstructive surgery and process for preparing same
US5133732A (en) 1987-10-19 1992-07-28 Medtronic, Inc. Intravascular stent
US4886062A (en) 1987-10-19 1989-12-12 Medtronic, Inc. Intravascular radially expandable stent and method of implant
US5266073A (en) 1987-12-08 1993-11-30 Wall W Henry Angioplasty stent
US5192307A (en) 1987-12-08 1993-03-09 Wall W Henry Angioplasty stent
US4856516A (en) 1989-01-09 1989-08-15 Cordis Corporation Endovascular stent apparatus and method
CH678393A5 (en) 1989-01-26 1991-09-13 Ulrich Prof Dr Med Sigwart
US5163958A (en) 1989-02-02 1992-11-17 Cordis Corporation Carbon coated tubular endoprosthesis
US4990155A (en) 1989-05-19 1991-02-05 Wilkoff Howard M Surgical stent method and apparatus
US4994071A (en) 1989-05-22 1991-02-19 Cordis Corporation Bifurcating stent apparatus and method
US5171262A (en) 1989-06-15 1992-12-15 Cordis Corporation Non-woven endoprosthesis
US5015253A (en) 1989-06-15 1991-05-14 Cordis Corporation Non-woven endoprosthesis
US5292331A (en) 1989-08-24 1994-03-08 Applied Vascular Engineering, Inc. Endovascular support device
IE73670B1 (en) 1989-10-02 1997-07-02 Medtronic Inc Articulated stent
US5035706A (en) 1989-10-17 1991-07-30 Cook Incorporated Percutaneous stent and method for retrieval thereof
US5176660A (en) 1989-10-23 1993-01-05 Cordis Corporation Catheter having reinforcing strands
DK0441516T3 (en) 1990-02-08 1995-06-12 Howmedica Inflatable catheter
US5545208A (en) 1990-02-28 1996-08-13 Medtronic, Inc. Intralumenal drug eluting prosthesis
US6004346A (en) 1990-02-28 1999-12-21 Medtronic, Inc. Intralumenal drug eluting prosthesis
IL94138A (en) 1990-04-19 1997-03-18 Instent Inc Device for the treatment of constricted fluid conducting ducts
US5064435A (en) 1990-06-28 1991-11-12 Schneider (Usa) Inc. Self-expanding prosthesis having stable axial length
US5122154A (en) 1990-08-15 1992-06-16 Rhodes Valentine J Endovascular bypass graft
US5449372A (en) 1990-10-09 1995-09-12 Scimed Lifesystems, Inc. Temporary stent and methods for use and manufacture
DE9116881U1 (en) 1990-10-09 1994-07-07 Cook Inc Percutaneous stent
US5222971A (en) 1990-10-09 1993-06-29 Scimed Life Systems, Inc. Temporary stent and methods for use and manufacture
US5217483A (en) 1990-11-28 1993-06-08 Numed, Inc. Intravascular radially expandable stent
US5178618A (en) 1991-01-16 1993-01-12 Brigham And Womens Hospital Method and device for recanalization of a body passageway
US5354257A (en) 1991-01-29 1994-10-11 Med Institute, Inc. Minimally invasive medical device for providing a radiation treatment
US5135536A (en) 1991-02-05 1992-08-04 Cordis Corporation Endovascular stent and method
US5116365A (en) 1991-02-22 1992-05-26 Cordis Corporation Stent apparatus and method for making
US5304200A (en) 1991-05-29 1994-04-19 Cordis Corporation Welded radially expandable endoprosthesis and the like
USD359802S (en) 1991-06-28 1995-06-27 Cook Incorporated Vascular stent
US5314472A (en) 1991-10-01 1994-05-24 Cook Incorporated Vascular stent
US5443498A (en) 1991-10-01 1995-08-22 Cook Incorporated Vascular stent and method of making and implanting a vacsular stent
US5500013A (en) 1991-10-04 1996-03-19 Scimed Life Systems, Inc. Biodegradable drug delivery vascular stent
US5464450A (en) 1991-10-04 1995-11-07 Scimed Lifesystems Inc. Biodegradable drug delivery vascular stent
WO1993006792A1 (en) 1991-10-04 1993-04-15 Scimed Life Systems, Inc. Biodegradable drug delivery vascular stent
US5366504A (en) 1992-05-20 1994-11-22 Boston Scientific Corporation Tubular medical prosthesis
US5387235A (en) 1991-10-25 1995-02-07 Cook Incorporated Expandable transluminal graft prosthesis for repair of aneurysm
CA2380683C (en) 1991-10-28 2006-08-08 Advanced Cardiovascular Systems, Inc. Expandable stents and method for making same
US5258021A (en) 1992-01-27 1993-11-02 Duran Carlos G Sigmoid valve annuloplasty ring
CA2087132A1 (en) 1992-01-31 1993-08-01 Michael S. Williams Stent capable of attachment within a body lumen
US5405377A (en) 1992-02-21 1995-04-11 Endotech Ltd. Intraluminal stent
DE4206843C2 (en) 1992-03-04 1994-03-24 Heraeus Elektrochemie Electrochemical cells for performing electrochemical processes
EP0566245B1 (en) 1992-03-19 1999-10-06 Medtronic, Inc. Intraluminal stent
US5591224A (en) 1992-03-19 1997-01-07 Medtronic, Inc. Bioelastomeric stent
US5571166A (en) 1992-03-19 1996-11-05 Medtronic, Inc. Method of making an intraluminal stent
US5599352A (en) 1992-03-19 1997-02-04 Medtronic, Inc. Method of making a drug eluting stent
US5282823A (en) 1992-03-19 1994-02-01 Medtronic, Inc. Intravascular radially expandable stent
US5510077A (en) 1992-03-19 1996-04-23 Dinh; Thomas Q. Method of making an intraluminal stent
US5370683A (en) 1992-03-25 1994-12-06 Cook Incorporated Vascular stent
FR2689388B1 (en) 1992-04-07 1999-07-16 Celsa Lg PERFECTIONALLY RESORBABLE BLOOD FILTER.
CA2094858C (en) 1992-04-28 2004-06-15 Robert D. Mitchell Method of treating hyperproliferative vascular disease
US5354308A (en) 1992-05-01 1994-10-11 Beth Israel Hospital Association Metal wire stent
WO1995014500A1 (en) 1992-05-01 1995-06-01 Beth Israel Hospital A stent
US5383928A (en) 1992-06-10 1995-01-24 Emory University Stent sheath for local drug delivery
US5342387A (en) 1992-06-18 1994-08-30 American Biomed, Inc. Artificial support for a blood vessel
DE4222380A1 (en) * 1992-07-08 1994-01-13 Ernst Peter Prof Dr M Strecker Endoprosthesis implantable percutaneously in a patient's body
US5382261A (en) 1992-09-01 1995-01-17 Expandable Grafts Partnership Method and apparatus for occluding vessels
US5578075B1 (en) 1992-11-04 2000-02-08 Daynke Res Inc Minimally invasive bioactivated endoprosthesis for vessel repair
US5449382A (en) 1992-11-04 1995-09-12 Dayton; Michael P. Minimally invasive bioactivated endoprosthesis for vessel repair
US5342348A (en) 1992-12-04 1994-08-30 Kaplan Aaron V Method and device for treating and enlarging body lumens
BE1006440A3 (en) 1992-12-21 1994-08-30 Dereume Jean Pierre Georges Em Luminal endoprosthesis AND METHOD OF PREPARATION.
US5370691A (en) 1993-01-26 1994-12-06 Target Therapeutics, Inc. Intravascular inflatable stent
US5981568A (en) 1993-01-28 1999-11-09 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
JPH08507715A (en) 1993-03-18 1996-08-20 シーダーズ サイナイ メディカル センター Drug-inducing and releasable polymeric coatings for bioartificial components
US5607463A (en) 1993-03-30 1997-03-04 Medtronic, Inc. Intravascular medical device
US5441515A (en) 1993-04-23 1995-08-15 Advanced Cardiovascular Systems, Inc. Ratcheting stent
US5824048A (en) 1993-04-26 1998-10-20 Medtronic, Inc. Method for delivering a therapeutic substance to a body lumen
US5464650A (en) 1993-04-26 1995-11-07 Medtronic, Inc. Intravascular stent and method
US5411549A (en) 1993-07-13 1995-05-02 Scimed Life Systems, Inc. Selectively expandable, retractable and removable stent
PT711158E (en) * 1993-07-29 2004-04-30 Us Gov Health & Human Serv METHOD FOR TREATING ATHEROSCLEROSIS OR RESTENING USING A MICROTUBLE STABILIZING AGENT
WO1995008355A1 (en) * 1993-09-24 1995-03-30 Baxter International Inc. Methods for enhancing vascularization of implant devices
WO1995010989A1 (en) 1993-10-19 1995-04-27 Scimed Life Systems, Inc. Intravascular stent pump
US5389106A (en) 1993-10-29 1995-02-14 Numed, Inc. Impermeable expandable intravascular stent
JP2703510B2 (en) 1993-12-28 1998-01-26 アドヴァンスド カーディオヴァスキュラー システムズ インコーポレーテッド Expandable stent and method of manufacturing the same
US5519042A (en) * 1994-01-13 1996-05-21 Hoechst Aktiengesellschaft Method of treating hyperproliferative vascular disease
US5403341A (en) 1994-01-24 1995-04-04 Solar; Ronald J. Parallel flow endovascular stent and deployment apparatus therefore
US5609627A (en) 1994-02-09 1997-03-11 Boston Scientific Technology, Inc. Method for delivering a bifurcated endoluminal prosthesis
US5443477A (en) 1994-02-10 1995-08-22 Stentco, Inc. Apparatus and method for deployment of radially expandable stents by a mechanical linkage
US5643312A (en) 1994-02-25 1997-07-01 Fischell Robert Stent having a multiplicity of closed circular structures
US5441516A (en) 1994-03-03 1995-08-15 Scimed Lifesystems Inc. Temporary stent
US5449373A (en) 1994-03-17 1995-09-12 Medinol Ltd. Articulated stent
CA2188563C (en) 1994-04-29 2005-08-02 Andrew W. Buirge Stent with collagen
US5629077A (en) 1994-06-27 1997-05-13 Advanced Cardiovascular Systems, Inc. Biodegradable mesh and film stent
US5397355A (en) 1994-07-19 1995-03-14 Stentco, Inc. Intraluminal stent
US5788979A (en) 1994-07-22 1998-08-04 Inflow Dynamics Inc. Biodegradable coating with inhibitory properties for application to biocompatible materials
US5891108A (en) 1994-09-12 1999-04-06 Cordis Corporation Drug delivery stent
US5649977A (en) 1994-09-22 1997-07-22 Advanced Cardiovascular Systems, Inc. Metal reinforced polymer stent
EP1181904B1 (en) 1994-10-17 2009-06-24 Kabushikikaisha Igaki Iryo Sekkei Stent for liberating drug
US5707385A (en) 1994-11-16 1998-01-13 Advanced Cardiovascular Systems, Inc. Drug loaded elastic membrane and method for delivery
US5637113A (en) 1994-12-13 1997-06-10 Advanced Cardiovascular Systems, Inc. Polymer film for wrapping a stent structure
NL9500094A (en) 1995-01-19 1996-09-02 Industrial Res Bv Y-shaped stent and method of deployment.
US5605696A (en) 1995-03-30 1997-02-25 Advanced Cardiovascular Systems, Inc. Drug loaded polymeric material and method of manufacture
US5709713A (en) 1995-03-31 1998-01-20 Cardiovascular Concepts, Inc. Radially expansible vascular prosthesis having reversible and other locking structures
EP0734698B9 (en) 1995-04-01 2006-07-05 Variomed AG Stent for transluminal implantation into hollow organs
US5837313A (en) 1995-04-19 1998-11-17 Schneider (Usa) Inc Drug release stent coating process
US6099562A (en) * 1996-06-13 2000-08-08 Schneider (Usa) Inc. Drug coating with topcoat
US5674242A (en) 1995-06-06 1997-10-07 Quanam Medical Corporation Endoprosthetic device with therapeutic compound
US5603722A (en) 1995-06-06 1997-02-18 Quanam Medical Corporation Intravascular stent
US5609629A (en) 1995-06-07 1997-03-11 Med Institute, Inc. Coated implantable medical device
AU716005B2 (en) 1995-06-07 2000-02-17 Cook Medical Technologies Llc Implantable medical device
US5820917A (en) 1995-06-07 1998-10-13 Medtronic, Inc. Blood-contacting medical device and method
US5679659A (en) 1995-08-22 1997-10-21 Medtronic, Inc. Method for making heparinized biomaterials
US5672638A (en) 1995-08-22 1997-09-30 Medtronic, Inc. Biocompatability for solid surfaces
US5607475A (en) 1995-08-22 1997-03-04 Medtronic, Inc. Biocompatible medical article and method
US5669924A (en) 1995-10-26 1997-09-23 Shaknovich; Alexander Y-shuttle stent assembly for bifurcating vessels and method of using the same
DE19614160A1 (en) 1996-04-10 1997-10-16 Variomed Ag Stent for transluminal implantation in hollow organs
NZ331269A (en) 1996-04-10 2000-01-28 Advanced Cardiovascular System Expandable stent, its structural strength varying along its length
US5728420A (en) 1996-08-09 1998-03-17 Medtronic, Inc. Oxidative method for attachment of glycoproteins to surfaces of medical devices
UA58485C2 (en) 1996-05-03 2003-08-15 Медінол Лтд. Method for manufacture of bifurcated stent (variants) and bifurcated stent (variants)
US5697971A (en) 1996-06-11 1997-12-16 Fischell; Robert E. Multi-cell stent with cells having differing characteristics
US5820918A (en) 1996-07-11 1998-10-13 Hercules Incorporated Medical devices containing in-situ generated medical compounds
DE69714281T2 (en) 1996-08-30 2003-02-27 Agricultural Res Org INTRACORONARY STENT CONTAINS THE QUINAZOLINONE DERIVATIVES
US5807404A (en) 1996-09-19 1998-09-15 Medinol Ltd. Stent with variable features to optimize support and method of making such stent
US6387121B1 (en) * 1996-10-21 2002-05-14 Inflow Dynamics Inc. Vascular and endoluminal stents with improved coatings
US5833651A (en) 1996-11-08 1998-11-10 Medtronic, Inc. Therapeutic intraluminal stents
KR100526913B1 (en) 1997-02-20 2005-11-09 쿡 인코포레이티드 Coated implantable medical device
US20020133222A1 (en) * 1997-03-05 2002-09-19 Das Gladwin S. Expandable stent having a plurality of interconnected expansion modules
US5843172A (en) 1997-04-15 1998-12-01 Advanced Cardiovascular Systems, Inc. Porous medicated stent
US6273913B1 (en) 1997-04-18 2001-08-14 Cordis Corporation Modified stent useful for delivery of drugs along stent strut
US5879697A (en) 1997-04-30 1999-03-09 Schneider Usa Inc Drug-releasing coatings for medical devices
US5916910A (en) * 1997-06-04 1999-06-29 Medinox, Inc. Conjugates of dithiocarbamates with pharmacologically active agents and uses therefore
US6306166B1 (en) 1997-08-13 2001-10-23 Scimed Life Systems, Inc. Loading and release of water-insoluble drugs
US6890546B2 (en) * 1998-09-24 2005-05-10 Abbott Laboratories Medical devices containing rapamycin analogs
US6225346B1 (en) * 1997-10-24 2001-05-01 Sugen, Inc. Tyrphostin like compounds
US5932580A (en) * 1997-12-01 1999-08-03 Yissum Research And Development Company Of The Hebrew University Of Jerusalem PDGF receptor kinase inhibitory compounds their preparation and compositions
US7208010B2 (en) * 2000-10-16 2007-04-24 Conor Medsystems, Inc. Expandable medical device for delivery of beneficial agent
US20020099438A1 (en) * 1998-04-15 2002-07-25 Furst Joseph G. Irradiated stent coating
CA2340652C (en) * 1998-08-20 2013-09-24 Cook Incorporated Coated implantable medical device comprising paclitaxel
US6187024B1 (en) 1998-11-10 2001-02-13 Target Therapeutics, Inc. Bioactive coating for vaso-occlusive devices
US6730349B2 (en) * 1999-04-19 2004-05-04 Scimed Life Systems, Inc. Mechanical and acoustical suspension coating of medical implants
US6287628B1 (en) * 1999-09-03 2001-09-11 Advanced Cardiovascular Systems, Inc. Porous prosthesis and a method of depositing substances into the pores
US6899731B2 (en) * 1999-12-30 2005-05-31 Boston Scientific Scimed, Inc. Controlled delivery of therapeutic agents by insertable medical devices
US6379382B1 (en) * 2000-03-13 2002-04-30 Jun Yang Stent having cover with drug delivery capability
US7419678B2 (en) 2000-05-12 2008-09-02 Cordis Corporation Coated medical devices for the prevention and treatment of vascular disease
US6776796B2 (en) 2000-05-12 2004-08-17 Cordis Corportation Antiinflammatory drug and delivery device
US7077836B2 (en) * 2000-07-21 2006-07-18 Vein Rx, Inc. Methods and apparatus for sclerosing the wall of a varicose vein
US6545097B2 (en) * 2000-12-12 2003-04-08 Scimed Life Systems, Inc. Drug delivery compositions and medical devices containing block copolymer
US20020103526A1 (en) * 2000-12-15 2002-08-01 Tom Steinke Protective coating for stent
US6939375B2 (en) * 2000-12-22 2005-09-06 Avantac Vascular Corporation Apparatus and methods for controlled substance delivery from implanted prostheses
US7179251B2 (en) * 2001-01-17 2007-02-20 Boston Scientific Scimed, Inc. Therapeutic delivery balloon
US20020119178A1 (en) * 2001-02-23 2002-08-29 Luc Levesque Drug eluting device for treating vascular diseases
US20020133224A1 (en) * 2001-03-13 2002-09-19 Clara Bajgar Drug eluting encapsulated stent

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5516781A (en) * 1992-01-09 1996-05-14 American Home Products Corporation Method of treating restenosis with rapamycin
US6562829B1 (en) * 1997-05-23 2003-05-13 Hadasit Medical Research Services & Development Co., Ltd. Treatment of hepatic cirrhosis
US6159488A (en) * 1997-08-14 2000-12-12 Agricultural Research Org. Ministry Of Agriculture (Gov.) Intracoronary stents containing quinazolinone derivatives
US6369039B1 (en) * 1998-06-30 2002-04-09 Scimed Life Sytems, Inc. High efficiency local drug delivery
US6335029B1 (en) * 1998-08-28 2002-01-01 Scimed Life Systems, Inc. Polymeric coatings for controlled delivery of active agents
US6368658B1 (en) * 1999-04-19 2002-04-09 Scimed Life Systems, Inc. Coating medical devices using air suspension
US8592036B2 (en) * 2006-06-23 2013-11-26 Abbott Cardiovascular Systems Inc. Nanoshells on polymers

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050249775A1 (en) * 2003-12-19 2005-11-10 Robert Falotico Intraluminal medical devices in combination with therapeutic agents
US8747881B2 (en) 2003-12-19 2014-06-10 Cordis Corporation Intraluminal medical devices in combination with therapeutic agents
US9265598B2 (en) 2003-12-19 2016-02-23 Cordis Corporation Local vascular delivery of sirolimus to prevent restenosis following vascular injury
US9265597B2 (en) 2003-12-19 2016-02-23 Cordis Corporation; Wyeth LLC Local vascular delivery of probucol in combination with sirolimus
US20050270864A1 (en) * 2004-05-18 2005-12-08 Peter Poechmueller Memory cell arrangement having dual memory cells
US10182928B2 (en) 2013-04-16 2019-01-22 Kaneka Corporation Medical tubular body

Also Published As

Publication number Publication date
AU2001262957B2 (en) 2004-12-02
EP1289576A1 (en) 2003-03-12
DE60111743T2 (en) 2005-12-15
ATE298592T1 (en) 2005-07-15
EP1289576B1 (en) 2005-06-29
AU6158101A (en) 2001-11-26
JP2003533493A (en) 2003-11-11
DE60111743D1 (en) 2005-08-04
ES2244622T3 (en) 2005-12-16
BR0110778A (en) 2007-05-08
AU6295701A (en) 2001-11-26
US8029561B1 (en) 2011-10-04
WO2001087372A1 (en) 2001-11-22
PT1289576E (en) 2005-10-31
MXPA02011186A (en) 2004-09-09
MXPA02011099A (en) 2004-08-19
CA2408606A1 (en) 2001-11-22

Similar Documents

Publication Publication Date Title
US8029561B1 (en) Drug combination useful for prevention of restenosis
AU2001262957A1 (en) Drug combinations useful for prevention of restenosis
AU2001263113B2 (en) Drug/drug delivery systems for the prevention and treatment of vascular disease
US7300662B2 (en) Drug/drug delivery systems for the prevention and treatment of vascular disease
US20020007213A1 (en) Drug/drug delivery systems for the prevention and treatment of vascular disease
US20020007215A1 (en) Drug/drug delivery systems for the prevention and treatment of vascular disease
US20020005206A1 (en) Antiproliferative drug and delivery device
US20020007214A1 (en) Drug/drug delivery systems for the prevention and treatment of vascular disease
US20090204204A1 (en) Drug/Drug Deliver Systems For The Prevention And Treatment Of Vascular Disease
AU2001261580A1 (en) Delivery devices for treatment of vascular disease
JP2005334646A (en) Antiproliferative drug and delivery device
EP1591135A1 (en) Drug/drug delivery systems for the prevention and treatment of vascular disease

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION