WO2002056692A1 - Methods for modulating tumor growth and metastasis - Google Patents

Methods for modulating tumor growth and metastasis Download PDF

Info

Publication number
WO2002056692A1
WO2002056692A1 PCT/US2001/050261 US0150261W WO02056692A1 WO 2002056692 A1 WO2002056692 A1 WO 2002056692A1 US 0150261 W US0150261 W US 0150261W WO 02056692 A1 WO02056692 A1 WO 02056692A1
Authority
WO
WIPO (PCT)
Prior art keywords
combretastatin
agent
pharmaceutical composition
compound
cancer agent
Prior art date
Application number
PCT/US2001/050261
Other languages
English (en)
French (fr)
Inventor
Francis Y. Lee
Ronald Peck
David Chaplin
Ronald Pero
Klaus Edvardsen
Original Assignee
Bristol-Myers Squibb Company
Oxigene, Inc.
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
Application filed by Bristol-Myers Squibb Company, Oxigene, Inc. filed Critical Bristol-Myers Squibb Company
Priority to CA2432792A priority Critical patent/CA2432792C/en
Priority to EP01994435A priority patent/EP1351573A4/en
Priority to JP2002557211A priority patent/JP2004523517A/ja
Priority to AU2002246827A priority patent/AU2002246827B2/en
Publication of WO2002056692A1 publication Critical patent/WO2002056692A1/en

Links

Classifications

    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/045Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
    • A61K31/05Phenols
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Definitions

  • This invention relates to the fields of oncology and improved chemotherapy regimens .
  • oncogenes function through the normal cellular signaling pathways required for organismal growth and cellular function (reviewed in McCormick, Nature 363:15-16, (1993))
  • additional alterations in the oncogenic signaling pathways may also contribute to tumor malignancy (Gilks et al . , Mol. Cell Biol. 13_: 1759-1768, (1993)), even though mutations in the signaling pathways alone may not cause cancer.
  • chemotherapeutic forms predominantly inter-strand DNA cross links in the nuclei of cells, thereby preventing cellular replication. As a result, tumor growth is initially repressed, and then reversed.
  • Another type of chemotherapeutic is referred to as an alkylating agent. These compounds function by inserting foreign compositions or molecules into the DNA of dividing cancer cells. As a result of these foreign moieties, the normal functions of cancer cells are disrupted and proliferation is prevented.
  • Another type of chemotherapeutic is an antineoplastic agent. This type of agent prevents, kills, or blocks the growth and spread of cancer cells. Still other types of anticancer agents include nonsteroidal aromastase inhibitors, bifunctional alkylating agents, etc.
  • fluorouracil a commonly used antineoplastic agent causes swelling or redness of normal skin, black or tarry stools, blood in the urine, chest pain, confusion, diarrhea, shortness of breath, and drowsiness.
  • Administration of fluorouracil has also been associated with fever, chills, cough, sore throat, lower back pain, mouth sores, nausea, vomiting, pain and/or difficulty passing urine.
  • Taxane administration has been associated with cardiovascular events such as syncope, rhythm abnormalities, hypertension and venous thrombosis; bone marrow suppression; neutropenia; anemia; peripheral neuropathy arthralgia/myalgia; nausea/vomiting and alopecia, to name only a few.
  • Combretastatins are another class of anticancer agents.
  • Combretastatins have been isolated from stem wood of the African tree combretum caffrum (Combretaceae) , and are potent inhibitors of microtubulin assembly.
  • Combretastatin A-4 (CA-4) is significantly active against the US National Cancer Institute's (NCI) murine L1210 and P338 lymphocytic leukemia cell lines.
  • NCI National Cancer Institute's
  • CA-4 was found to compete with combretastatin A-1 (CA-1) , another compound isolated from Combretum caffrum, as a potent inhibitor of colchicine binding to tubulin.
  • CA-4 also strongly retards the growth of certain cell lines (ED50 ⁇ 0.01 (g/ml), and is a powerful anti-mitotic agent. See US Patent 4,996,237. Furthermore, an "anti-vascular" mechanism of action for both CA-4 and CA-1 has recently been discovered. Since the solubility of the combretastatins is very limited, prodrugs have been developed, such as combretastatin A-4 phosphate disodium salt and combretastatin A-1 phosphate disodium salt (hereinafter "CA4P” and “CAlP” respectively), to increase the solubility, and thus the efficacy of CA-4 and CA-1.
  • CA4P combretastatin A-4 phosphate disodium salt
  • CAlP combretastatin A-1 phosphate disodium salt
  • the present invention provides effective therapeutic methods for modulating tumor growth or metastasis wherein a combination of agents is employed.
  • the methods of the present invention provide advantages such as greater overall efficacy, for example, in achieving synergy or avoiding antagonism, and allow, where desired, a reduction in the amount of one or more of the individual agents employed with a concomitant reduction in side effects. Further, where the tumor to be treated is not optimally responsive to a given anticancer agent, use of the present combination therapy methods can nonetheless provide effective treatment.
  • the present invention provides a method for modulating tumor growth or metastasis in an animal, especially a human, in need thereof, comprising sequential or simultaneous administration of a combretastatin A-4 compound or combretastatin A-1 compound and at least one other anticancer agent, in amounts effective therefor. Preferred such agents are described further below.
  • the method of the present invention can provide the aforementioned advantages . Further, the present inventors have found that certain sequences of administering the combretastatin A- 4 compound or combretastatin A-1 compound and the other anticancer agent can, in vivo, potentiate the overall efficacy of the combination.
  • Combretastatin A-4 compounds or combretastatin A-1 compounds modulate blood flow to tumor tissue.
  • the overall efficacy of the combination is potentiated.
  • certain anticancer agents are most efficacious at relatively high tumor concentrations, but are rapidly cleared from tumor tissue.
  • the present inventors have found that simultaneous administration of the combretastatin A-4 compound or combretastatin A-1 compound and the other anticancer agent potentiates the effect of the combination.
  • Peak Tumor Concentration Agents are thus preferably administered simultaneously with, or within close temporal proximity to, the combretastatin A-4 compound or combretastatin A-1 compound.
  • agents for example, need not be present at high concentrations, but are effective during a relatively short period of the overall cell cycle. As such agents can become protein-bound and inactive over time when remaining in contact with tumor tissue, they are therefore most efficacious under conditions where a continuing supply of the agent reaches the tumor.
  • Potentiation of the efficacy of combination therapy in these cases can be obtained by administering the anticancer agent and combretastatin A-4 compound or combretastatin A-1 compound sequentially, with sufficient delay between administrations to allow the action of one of the agents before the other.
  • the anticancer agent when such anticancer agent is administered first, followed by a delay before administering the combretastatin A-4 compound or combretastatin A-1 compound, the anticancer agent reaches the tumor tissue over a sufficient duration to allow action of the compound, with subsequent administration of the combretatatin A-4 compound or combretastatin A-1 compound further damaging tumor tissue.
  • the tumor is initially weakened by the combretastatin A-4 compound or combretastatin A-1 compound, followed by further damage to the tumor by the anticancer agent .
  • duration of anticancer agent tumor concentration is more significant than peak concentration.
  • the damage to tumor vasculature by the initial administration of the combretastin A-4 compound or combretastatin A-1 compound does not prevent the relatively low concentration of anticancer agent needed from reaching the tumor tissue once blood flow resumes.
  • Such agents are termed herein "Duration Exposure Agents”.
  • Duration Exposure Agents and the combretastatin A-4 compound or combretastatin A-1 compound are thus preferably administered sequentially, with either administration of the combretastatin A-4 compound or combretastatin A-1 compound first, followed by the anticancer agent, or vice versa, provided that a sufficient delay is allowed between administrations to potentiate the combination.
  • Administration of the anticancer agent after the administration of combretastatin A-4 compound or combretastatin A-1 compound is most preferred for Duration Exposure Agents.
  • certain agents are most efficacious when present at relatively high concentrations in tumor tissue over a longer duration (i.e., maximizing the "area under the curve" (AUC) of a plot of concentration over time) .
  • Administering such agents first, followed by a delay before administering the combretastatin A-4 compound or combretastatin A-1 compound allows action of the anticancer agent, with subsequent administration of the combretastatin A-4 compound or combretastatin A-1 compound further weakening the tumor tissue.
  • administration of the anticancer agent first avoids premature damage to tumor vasculature and allows sufficient concentrations of anticancer agent to reach the tumor.
  • High AUC Agents Such agents are termed herein "High AUC Agents".
  • High AUC Agents and the combretastatin A-4 compound or combretastatin A-1 compound are thus preferably administered sequentially, with administration of the High AUC Agent preceding administration of the combretastatin A-4 compound or combretastatin A-1 compound, provided that a sufficent delay is allowed between administrations to potentatiate the combination.
  • the present invention therefore provides as a further embodiment, a method for modulating tumor growth or metastasis in an animal in need thereof, especially a human, comprising administration of a combretastatin A-4 compound or combretastatin A-1 compound and at least one anticancer agent, in amounts effective therefor, wherein said combretastatin A-4 compound or combretastatin A-1 compound is administered at a time relative to administration of said anticancer agent sufficient to modulate blood flow to said tumor to provide a time-dependent effective tumor concentration of said anticancer agent .
  • the method of the present invention allows potentiation of the overall efficacy of the combination employed.
  • time-dependent effective tumor concentration denotes a concentration of the other anticancer agent in the tumor tissue over time (i.e, from administration until the agent is cleared from the body) which potentiates the action of the combination of the combretastatin A-4 compound or combretastatin A-1 compound and other anticancer agent.
  • potentiation can denote use of a combination without antagonistic results.
  • Patentiation can also denote achieving an unexpected improvement in the overall efficacy of the combination, such as synergy.
  • the present invention also provides pharmaceutical compositions comprising at least one anticancer agent and a combretastatin A-4 compound or combretastatin A-1 compound.
  • the anticancer agent and/or combretastatin A-4 compound or combretastatin A-1 compound can be present in a subtherapeutic dose for the individual agent, the agents being effective in combination, providing reduced side effects while maintaining efficacy.
  • each agent can be provided at higher doses for the individual agent, such as those found in the Physician's Desk Reference .
  • kits of the invention comprise a first pharmaceutical composition comprising at least one anticancer agent and a second pharmaceutical composition comprising a combretastatin A-4 compound or combretastatin A-1 compound together in a package.
  • the anticancer agent and/or combretastatin A4 compound or combretastatin A-1 compound can be present, for example, in a subtherapeutic dose for the individual agent, the agents being effective in combination and providing reduced side effects while maintaining efficacy.
  • each agent can be provided at a higher dose, such as those found for the agent in the Physician's Desk Reference.
  • combretastatin A-4 compound denotes at least one of combretastatin A-4, prodrugs (preferably phosphate prodrugs) and derivatives thereof, and salts of these compounds.
  • prodrugs preferably phosphate prodrugs
  • salts of these compounds include without limitation, combretastatin A-4, and various prodrugs of combretastatin A-4 exemplified by combretastatin A-4 phosphate and salts thereof, especially combretastatin A-4 phosphate disodium salt.
  • OR 1 and OR 2 are -0 " QH + , and the other is hydroxyl or -0 " QH +
  • Q is an amino acid containing at least two nitrogen atoms where one of the nitrogen atoms, together with a proton, forms a quaternary ammonium cation QH+, preferably, where one of OR 1 and OR 2 is hydroxyl, and the other is -0 ⁇ QH + where Q is L- histidine;
  • combretastatin A-1 compound denotes as least one of combretastatin A-1, prodrugs (preferably phosphate prodrugs) and derivatives thereof, and salts of these compounds.
  • Combretastatin A-1 is described in US Patent 5,409,953 to Pettit et al . and has the following general structure:
  • the terms “modulate”, “modulating” or “modulation” refer to changing the rate at which a particular process occurs, inhibiting a particular process, reversing a particular process, and/or preventing the initiation of a particular process. Accordingly, if the particular process is tumor growth or metastasis, the term “modulation” includes, without limitation, decreasing the rate at which tumor growth and/or metastasis occurs; inhibiting tumor growth and/or metastasis; reversing tumor growth and/or metastasis (including tumor shrinkage and/or eradication) and/or preventing tumor growth and/or metastasis.
  • anticancer agent denotes a chemical compound or electromagnetic radiation (especially, X-rays) which is capable of modulating tumor growth or metastasis.
  • X-rays especially, X-rays
  • the term refers to an agent other than a combretastatin A-4 compound or combretastatin A-1 compound. Unless otherwise indicated, this term can include one, or more than one, such agents.
  • the term “anticancer agent” encompasses the use of one or more chemical compounds and/or electomagnetic radiation in the present methods and compositions.
  • the relative time for administration of the combretastatin A-4 compound or combretastatin A-1 compound can, as desired, be selected to provide a time-dependent effective tumor concentration of one, or more than one, of the anticancer agents.
  • anticancer agents are exemplary of those having applications in a composition or method of the present invention.
  • Such classes of anticancer agents are described below:
  • Alkylating agent a compound that donates an alkyl group to nucleotides. Alkylated DNA is unable to replicate itself and cell proliferation is stopped. Examples of such compounds include, but are not limited to, busulfan, coordination metal complexes (such as carboplatin, oxaliplatin, and cisplatin) , cyclophosphamide (cytoxan) , dacarbazine, ifosfamide, mechlorethamine (mustargen) , and melphalan;
  • Bifunctional alkylating agent a compound having two labile methanesulfonate groups that are attached to opposite ends of a four carbon alkyl chain.
  • the methanesulfonate groups interact with, and cause damage to DNA in cancer cells, preventing their replication.
  • Examples of such compounds include, without limitation, chlorambucil and melphalan;
  • Non-steroidal aromatase inhibitor a compound that inhibits the enzyme aromatase, which is involved in estrogen production. Thus, blockage of aromatase results in the prevention of the production of estrogen. Examples of such compounds include anastrozole and exemstane; 4.
  • Immunotherapeutic agent an antibody or antibody fragment which targets cancer cells that produce proteins associated with malignancy. Exemplary immunotherapeutic agents include Herceptin which targets HER2 or HER2/neu, which occurs in high numbers in about 25 percent to 30 percent of breast cancers; and anti- CD20 which triggers apoptosis in B cell lymphomas .
  • Additional immunotherapeutic agents include immunotoxins , wherein toxin molecules such as ricin, diphtheria toxin and pseudomonas toxins are conjugated to antibodies which recognize tumor specific antigens. Conjugation can be achieved biochemically or via recombinant DNA methods .
  • Nitrosurea compound inhibits enzymes that are needed for DNA repair. These agents are able to travel to the brain so they are used to treat brain tumors, as well as non-Hodgkin' s lymphomas, multiple myeloma, and malignant melanoma. Examples of nitrosureas include carmustine and lomustine;
  • Antimetabolite a class of drugs that interfere with DNA and ribonucleic acid (RNA) elongation. These agents are phase specific (S phase) and are used to treat chronic leukemias as well as tumors of breast, ovary and the gastrointestinal tract. Examples of antimetabolites include 5-fluorouracil, methotrexate, ge citabine (GEMZAR) , cytarabine (Ara-C) , and fludarabine.
  • S phase phase specific
  • antimetabolites include 5-fluorouracil, methotrexate, ge citabine (GEMZAR) , cytarabine (Ara-C) , and fludarabine.
  • Antitumor antibiotic a compound having antimicrobial and cytotoxic activity. Such compounds also may interfere with DNA by chemically inhibiting enzymes and mitosis or altering cellular membranes. Examples include, but certainly are not limited to bleomycin, dactinomycin, daunorubicin, doxorubicin (Adriamycin) , and idarubicin; 8.
  • Mitotic inhibitor a compound that can inhibit mitosis (e.g., tubulin binding compounds) or inhibit enzymes that prevent protein synthesis needed for reproduction of the cell. Examples of mitotic inhibitors include taxanes such as paclitaxel and docetaxel, epothilones, etoposide, vinblastine, vincristine, and vinorelbine;
  • Radiation therapy includes but is not limited to X-rays or gamma rays which are delivered from either an externally supplied source such as a beam or by implantation of small radioactive sources.
  • Topoisomerase I inhibitors agents which interfere with topoisomerase activity thereby inhibiting DNA replication. Such agents include, without limitation, CPT-11 and topotecan.
  • Hormonal therapy includes, but is not limited to anti-estrogens, such as Tamoxifen, GnRH agonists, such as Lupron, and Progestin agents, such as Megace.
  • anti-estrogens such as Tamoxifen
  • GnRH agonists such as Lupron
  • Progestin agents such as Megace.
  • anticancer agents that function via a large variety of mechanisms have application in the pharmaceutical compositions and methods of the present invention.
  • Additional such agents include for example, leuocovorin, kinase inhibitors, such as Iressa and Flavopiridol, analogues of conventional chemotherapeutic agents such as taxane analogs and epothilone analogues, antiangiogenics such as matrix metalloproteinase inhibitors, and other VEGF inhibitors, such as ZD6474 and SU6668.
  • Retinoids such as Targretin can also be employed in the pharmaceutical compositions and methods of the invention.
  • Signal transduction inhibitors which interfere with farnesyl transferase activity and chemotherapy resistance modulators, e.g., Valspodar can also be employed.
  • Monoclonal antibodies such as C225 and anti-VEGFr antibodies can also be employed.
  • the phrase "effective amount" of a compound or pharmaceutical composition refers to an amount sufficient to modulate tumor growth or metastasis in an animal, especially a human, including without limitation decreasing tumor growth or size or preventing formation of tumor growth in an animal lacking any tumor formation prior to administration, i.e., prophylactic administration .
  • prodrug refers to a precursor form of the drug which is metabolically converted in vivo to produce the active drug.
  • combretastatin A-4 phosphate prodrug salts or combretastatin A-1 phosphate prodrug salts administered to an animal in accordance with the present invention undergo metabolic activation and regenerate combretastatin A-4 or combretastatin A-1 in vivo, e.g., following dissociation and exposure to endogenous non- specific phosphatases in the body.
  • the present invention is directed towards a pharmaceutical composition that modulates growth or metastasis of tumors, particularly solid tumors, using a pharmaceutical composition of the present invention, along with methods of modulating tumor growth or metastasis, for example, with a pharmaceutical composition of the present invention.
  • tumor As used herein, the terms “tumor”, “tumor growth” or “tumor tissue” can be used interchangeably, and refer to an abnormal growth of tissue resulting from uncontrolled progressive multiplication of cells and serving no physiological function.
  • a solid tumor can be malignant, e.g. tending to metastasize and being life threatening, or benign.
  • tumors comprising dysproliferative changes can be treated or prevented with a pharmaceutical composition or method of the present invention in epithelial tissues such as those in the cervix, esophagus, and lung.
  • the present invention provides for treatment of conditions known or suspected of preceding progression to neoplasia or cancer, in particular, where non- neoplastic cell growth consisting of hyperplasia, metaplasia, or most particularly, dysplasia has occurred (for review of such abnormal growth conditions, see Robbins and Angel1, 1976, Basic Pathology, 2d Ed., W.B. Saunders Co., Philadelphia, pp. 68 to 79).
  • Hyperplasia is a form of controlled cell proliferation involving an increase in cell number in a tissue or organ, without significant alteration in structure or function. For example, endometrial hyperplasia often precedes endometrial cancer. Metaplasia is a form of controlled cell growth in which one type of adult or fully differentiated cell substitutes for another type of adult cell. Metaplasia can occur in epithelial or connective tissue cells. Atypical metaplasia involves a somewhat disorderly metaplastic epithelium. Dysplasia is frequently a forerunner of cancer, and is found mainly in the epithelia; it is the most disorderly form of non-neoplastic cell growth, involving a loss in individual cell uniformity and in the architectural orientation of cells.
  • Dysplastic cells often have abnormally large, deeply stained nuclei, and exhibit pleomorphism. Dysplasia characteristically occurs where there exists chronic irritation or inflammation, and is often found in the cervix, respiratory passages, oral cavity, and gall bladder. For a review of such disorders, see Fishman et al . , 1985, Medicine, 2d Ed., J. B. Lippincott Co., Philadelphia.
  • AV arteriovenous
  • Peak Tumor Concentration Agents refers to anticancer agents which are most efficacious at high tumor concentrations yet are rapidly cleared from the tumor tissue. Such agents are preferably administered simultaneously with or in close temporal proximity to (e.g., as is clinically feasible, especially within one hour of) the administration of the combretastatin A-4 compound or combretastatin A-1 compound in accordance with the invention.
  • Exemplary Peak tumor Concentration Agents include, without limitation, alklating agents such as cytoxan and mitomycin C and metal coordination complexes such as cisplatin, oxaliplatin and carboplatin.
  • Duration Exposure Agents refers to agents which can be effective at relatively low tumor concentrations yet which require certain tumor tissue exposure times to be most effective. Such agents are preferably administered sequentially in any order with a combretastatin A-4 compound or combretastatin A-1 compound in accordance with the invention, provided that a sufficient delay is allowed between administrations to potentiate the combination.
  • the Duration Exposure Agent is administered after the administration of the combretastatin A-4 compound or combretastatin A-1 compound.
  • Exemplary Duration Exposure Agents include, without limitation, taxanes such as paclitaxel and docetaxel, etoposide, etoposide phosphate, immunotoxins, and epothilones.
  • High AUC Agents refers to those agents which show greatest efficacy when present at high concentrations in tumor tissue for extended time periods. Such agents are preferably administered sequentially with a combretastatin A-4 compound or combretastatin A-1 compound in accordance with the invention, wherein the High AUC Agent is administered first, followed by the combretastatin A-4 compound or combretastatin A-1 compound, provided that a sufficient delay is allowed between administrations to potentiate the combination.
  • Exemplary High AUC Agents include, without limitation, adriamycin, CPT-11 (irinotecan) , and topotecan.
  • Peak Tumor Concentration Agents such as platinum based anticancer agents, including cisplatin or carboplatin are administered essentially simultaneously with a combretastatin A-4 compound or combretastatin A-1 compound, such as combretastatin A-4 phosphate disodium salt or combretastatin A-1 phosphate disodium salt.
  • Duration Exposure Agents including immunotoxins, and taxanes, such as paclitaxel and docetaxel are administered after the combretastatin A-4 compound or combretastatin A-1 compound.
  • Administration of a combretastatin A-4 compound or combretastatin A-1 compound prior to the Duration Exposure Agent extends the exposure time of the tumor tissue to the Duration Exposure Agent.
  • High AUC Agents such as CPT-11 are administered prior to the administration of the combretastatin A-4 compound or combretastatin A-1 compound (e.g., combretastatin A-4 phosphate disodium salt or combretastatin A-1 phosphate disodium salt) .
  • Such agents can preferably be administered, for example, within 24 hours of the administration of the combretastatin A-4 compound or combretastatin A-1 compound, such as within 2-24 hours prior, 3-24 hours prior, 6-24 hours prior, 8-24 hours prior, or 12 to 24 hours prior to administration.
  • combinations such as those described above potentiate the efficacy of the combination and can provide the advantages described above.
  • the present methods permit the clinician to administer a combretastatin A-4 compound or combretastatin A-1 compound, such as the phosphate disodium salts of these compounds, and/or anticancer agent, at dosages which are significantly lower than those employed for the single agent.
  • Preferred dosages suitable for administration of the anticancer and combretastatin A-4 compounds or combretastatin A-1 compounds in accordance with the invention are set forth hereinbelow.
  • the combretastatin A-4 compound or combretastatin A-1 compound and the at least one anticancer agent can be administered in any amount or by any route of administration effective for the modulation of tumor growth or metastasis, especially treatment of cancer as described herein.
  • chemotherapeutically effective amount refers to a sufficient amount of the compounds of the invention to provide the desired anticancer effect. The exact amount required will vary from subject to subject, the mode of administration of the chemotherapeutic compounds and the like.
  • the present invention further provides chemotherapeutic pharmaceutical compositions comprising both a combretastatin A-4 compound or combretastatin A-1 compound, and at least one selected anticancer agent and the use thereof in the present methods.
  • the method of the present invention can be carried out using chemotherapeutic pharmaceutical compositions which comprise one of the above-described compounds as the active ingredient, in combination with a pharmaceutically acceptable carrier medium or an auxiliary agent.
  • the combretastatin A-4 compound or combretastatin A-1 compound such as combretastatin A-4 phosphate disodium salt or combretastatin A-1 phosphate disodium salt, and the anticancer agent, such as cisplatin are formulated and administered separately.
  • FIG. 1 Graph of the antitumor activity of cisplatin and combretastain A-4 phosphate disodium salt (CA4P) administered singly in the moderately platinum-resistant M5076DDP murine fibrosarcoma. Tumor was staged to 300 mg at treatment initiation. Cisplatin was administered intravenously (iv) , every 4 days for 3 doses (Q4D x 3). CA4P was given iv, every day for 10 days (Monday through Friday) .
  • FIG. 2 (A) Graph of therapeutic synergy observed with the combination of CA4P and the Peak Tumor Concentration Agent, cisplatin in the M5076DDP tumor model. Drug treatment was iv, Q4D x3. Drug combinations were administered simultaneously. (B) Graph showing CA4P significantly enhanced the antitumor activity of an otherwise inactive dose of cisplatin (3 mg/kg/inj ) .
  • FIG. 3 (A) Graph of therapeutic synergy observed with the combination of CA4P and the peak tumor concentration agent, carboplatin in the M5076 murine fibrosarcoma model. Drug treatment was intraperitoneal (ip) , Q4D x3. Drug combinations were administered simultaneously ip (admixed) . (B) Graph showing that CA4P, at three different dose levels (90-250 mg/kg/inj), significantly improved the antitumor activity of carboplatin.
  • FIG. 4 A graph showing antitumor activity in log cell kill indicating that the CA4P and carboplatin should essentially be administered simultaneously.
  • FIG. 5 Graph of inhibition of tumor blood flow by CA4P in the sc A2780 human ovarian carcinoma grown in nude mice (A) or nude rats (B) .
  • Fig. 6 Graph showing the antitumor effects of combined High AUC Agent, CPT-11, and CA4P chemotherapy in human ovarian carcinoma cells (A2780) .
  • CPT-11 is administered 3-24 hours prior to the administration of the combretastatin compound.
  • Fig. 7 Enhancement of the antitumor efficacy of carboplatin by low dose CA4P in the M5076/DDP tumors.
  • Panels A-C depict results for the combination of various doses of CA4P with 90, 60 and 40 mg/m 2 of carboplatin, respectively.
  • Fig. 8 Combination chemotherapy with CA4P and paclitaxel versus the 16/c murine mammary carcinoma.
  • Fig. 9 Combination chemotherapy with CA4P and paclitaxel versus A2780 human ovarian carcinoma. Simultaneous administration of the agents is antagonistic in this model.
  • Fig. 10 Combination chemotherapy with CA4P and paclitaxel versus A2780 human ovarian carcinoma. An interval of 3 hours between treatments abrogates negative interaction.
  • Fig. 11 A bar graph showing that combined administration of an immunotoxin BR96-sFv-PE40 with combretastatin A4P acts synergistically to reduce tumor size in a colon cancer xenograft model in an allogeneic Brown-Norway rat host.
  • Figs. 12A and 12B A pair of graphs showing that combretastatin A-lP inhibits blood flow in human tumor xenografts in nude mice in a manner comparable to that observed for combretastatin A-4P.
  • Fig. 12A N87 gastric cancer xenograft model
  • Fig. 12B A2780 ovarian cancer xenograft model .
  • Figs. 13A-13D are a series of graphs showing dose response curves of tumor size reduction in response to administration of combretastatin A-lP and carboplatin alone and in combination against an M5076 fibrosarcoma xenograft model. Combined administration of combretastatin A-lP and carboplatin acted synergistically to reduce tumor size.
  • Fig. 14 Graph showing that combined administration of combretastatin A-lP and carboplatin produces a synergistic antitumor effect, producing a complete response (disappearance of tumors) not observed in single agent therapy.
  • Fig. 15 A graph showing that combined administration of cisplatin and combretastatin A1P act synergistically to reduce tumor size in a CaNT breast tumor model in CBA mice.
  • improved chemotherapeutic regimens are provided for the treatment of cancer.
  • the improved chemotherapeutic regimens can lower side effects and enhance efficacy for the treatment of neoplastic disease.
  • CA-4 combretastatin A-4
  • CA4P there is considerable interest in exploiting the novel anti-vascular action of CA4P for cancer treatment .
  • Combretastatin A-1 and prodrugs thereof are also potent inhibitors of tubulin polymerization. CAlP has also been shown to cause shut down of blood flow to the tumor vasculature.
  • the present methods can, for example, be carried out using a single pharmaceutical composition comprising both combretastatin A-4 compound or combretastatin A-1 compound and anticancer agent (s) (when administration is to be simultaneous) or using two or more pharmaceutical compositions separately comprising combretastatin A-4 compound or combretastatin A-1 compound and anticancer agent (s) (when administration is to be simultaneous or sequential) .
  • Such pharmaceutical compositions can comprise, inter alia, at least one anticancer agent and/or a combretastatin A-4 compound or combretastatin A-1 compound, such as combretastatin A-4 phosphate disodium salt or combretastatin A-1 phosphate disodium salt and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable refers to molecular entities and compositions that are physiologically tolerable and preferably do not produce an allergic or similar untoward reaction, such as gastric upset, dizziness and the like, when administered to a human.
  • the term “pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S.
  • carrier refers, for example to a diluent, adjuvant, excipient, auxilliary agent or vehicle with which an active agent of the present invention is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oil. including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water or aqueou saline solutions and aqueous dextrose and glycerol solutions are preferably employed as carriers, particularly for injectable solutions. Suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E.W. Martin.
  • compositions contemplated to be withi the scope of the invention comprise, inter alia, pharmaceutically acceptable diluents, preservatives, solubilizers, emulsifiers, adjuvants and/or carrier;
  • Such compositions can include diluents of various - content (e.g., Tris-HCl, acetate, phosphate), pH and ionic strength; additives such as detergents and solubilizing agents (e.g., Tween 80, Polysorbate 80), anti-oxidants (e.g., ascorbic acid, sodium metabisulfite) , preservatives (e.g., Thimersol, benzyl alcohol) and bulking substances (e.g., lactose, mannitol) ; incorporation of the material into particulate preparations of polymeric compounds such as polylactic acid, polylactic acid
  • compositions may influence the physical state, stability, rate of in vivo release, and rate of in vivo clearance of components of a pharmaceutical composition of the present invention. See, e.g., Remington's Pharmaceutical Sciences, 18th Ed. (1990, Mack Publishing Co., Easton, PA 18042) pages 1435-1712 which are herein incorporated by reference.
  • a pharmaceutical composition of the present invention can be prepared, for example, in liquid form, or can be in dried powder, such as lyophilized form. Particular methods of administering such compositions are described infra.
  • the present invention is directed towards methods for modulating tumor growth and metastasis comprising, inter alia, the administration of a combretastatin A-4 compound or combretastatin A-1 compound, such as combretastatin A-4 phosphate disodium salt or combretastatin A-1 phosphate disodium salt, and at least one anticancer agent.
  • the agents of the invention can be administered separately (e.g, formulated and administered separately) , or in combination as a pharmaceutical composition of the present invention. Administration can be achieved by any suitable route, such as parenterally, transmucosally, e.g., orally, nasally, or rectally, or transdermally.
  • administration is parenteral, e.g., via intravenous injection.
  • Alternative means of administration also include, but are not limited to, intra-arteriole, intramuscular, intradermal, subcutaneous, intraperitoneal, intraventricular, and intracranial administration, or by injection into the tumor (s) being treated or into tissues surrounding the tumor (s) .
  • the combretastatin A-4 compound or combretastatin A-1 compound such as combretastatin A-4 phosphate disodium salt or combretastatin A-1 phosphate disodium salt and anticancer agent may be employed in any suitable pharmaceutical formulation, as described above, including in a vesicle, such as a liposome [see Langer, Science 249:1527-1533 (1990); Treat et al . , in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez- Berestein and Fidler (eds.), Liss: New York, pp.
  • administration of liposomes containing the agents of the invention is parenteral, e.g., via intravenous injection, but also may include, without limitation, intra-arteriole, intramuscular, intradermal, subcutaneous, intraperitoneal, intraventricular, and intracranial administration, or by injection into the tumor (s) being treated or into tissues surrounding the tumor (s).
  • a pharmaceutical composition of the present invention can be delivered in a controlled release system, such as using an intravenous infusion, an implantable osmotic pump, a transdermal patch, liposomes, or other modes of administration.
  • a pump may be used [see Langer, supra; Se ton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et al . , Surgery 88:507 (1980); Saudek et al . , N. Engl. J. Med. 321:574 (1989) ] .
  • polymeric materials can be used [see Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Press: Boca Raton, Florida (1974) ; Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley: New York (1984); Ranger and Peppas, J. Macromol . Sci. Rev.
  • a controlled release system can be placed in proximity of the target tissues of the animal, thus requiring only a fraction of the systemic dose [see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)].
  • a controlled release device can be introduced into an animal in proximity of the site of inappropriate immune activation or a tumor.
  • Combretastatin A-4 1-100 mg/m2 +5FU and optionally 5-5000 mg/m2 + Leucovorin 5-1000 mg/m2
  • Combretastatin A-4 1-100 mg/m2 + UFT and optionally 50-800 mg/m2 + leucovorin 5-1000 mg/m2
  • Combretastatin A-4 1-100 mg/m2 + Gemcitabine 100-3000 mg/m2 + Cisplatin 5-150 mg/m2
  • Combretastatin A-4 1-100 mg/m2 + Cisplatin 5-150 mg/m2 + paclitaxel 40-250 mg/m2
  • Combretastatin A-4 1-100 mg/m2 + radiation 200-8000 cGy + 5FU 5-5000 mg/m2 + Cisplatin 5-150 mg/m2
  • Combretastatin A-4 1-100 mg/m2 + paclitaxel 40-250 mg/m2 + 5-FU 5-5000 mg/m2
  • 5FU denotes 5-fluorouracil
  • Leucovorin can be employed as leucovorin calcium
  • UFT is a 1:4 molar ratio of tegafur :uracil
  • Ephilone is preferably a compound described in WO 99/02514 or WO 00/50423, both incorporated by reference herein in their entirety.
  • Table I provides exemplary dosage ranges of the combretastatin A-4 compounds or combretastatin ' A-1 compounds and certain anticancer agents of the invention
  • the clinician may utilize preferred dosages as warranted by the condition of the patient being treated.
  • combretastatin A-4 compounds or combretastatin A-1 compounds may preferably be administered at a dosage ranging from 30-70 mg/m2 every three weeks for as long as treatment is required.
  • Preferred dosages for cisplatin are 75-120 mg/m2 administered every three weeks.
  • Preferred dosages for carboplatin are within the range of 200-600 mg/m2 or an AUC of 0.5-8 mg/ml x min; most preferred is an AUC of 4- 6 mg/ml x min.
  • preferred dosages are within the range of 200-6000 cGY.
  • Preferred dosages for CPT-11 are within 100-125 mg/m2 , once a week.
  • Preferred dosages for paclitaxel are 130-225 mg/m2 every 21 days.
  • Preferred dosages for gemcitabine are within the range of 80-1500 mg/m2 administered weekly.
  • Preferably UFT is used within a range of 300-400 mg/m2 per day when combined with leucovorin administration.
  • Preferred dosages for leucovorin are 10-600 mg/m2 administered weekly.
  • a preferred dose of the Br96-sFv-PE40 immunotoxin is 420 mg/m2.
  • the use of the BR96-sFv-PE40 immunotoxin in combination with combretastatin A4 and its prodrugs in immune enhancing therapy is described in US Provisional Application 60/258,283, filed December 26, 2000, the entire disclosure of which is incorporated by reference herein.
  • Certain cancers can be treated effectively with combretastatin A-4 or combretastatin A-1 and a plurality of anticancer agents .
  • Such triple and quadruple combinations can provide greater efficacy.
  • the dosages set forth above can be utilized.
  • Other such combinations in the above Table I can therefore include "combretastatin A-4 or combretastatin A-1" in combination with (1) mitoxantrone + prednisone; (2) doxorubicin + taxane; or (3) herceptin + taxane.
  • 5-FU can be replaced by UFT in any of the above combinations.
  • agents used in the modulation of tumor growth or metastasis in a clinical setting such as antiemetics, can also be administered as desired.
  • Drug administration For administration to rodents, CA4P was dissolved in normal saline ( 0 . 9% NaCl) . Paclitaxel was dissolved in a 50/50 mixture of ethanol and Cremophor® and stored at 4°C; final dilution of paclitaxel was obtained immediately before drug administration with NaCl 0.9%. Fresh preparation of paclitaxel was employed to avoid precipitation. CPT-11 was dissolved in normal saline. The volume of all compounds injected was 0.01 ml/g of mice, and 0.005 ml/g of rats.
  • the human tumors were maintained in Balb/c nu/nu nude mice.
  • M5076 and M5076ddp was maintained in C57BL/6 mice .
  • Tumors were propagated as subcutaneous transplants in the appropriate mouse strain using tumor fragments obtained from donor mice.
  • M5076 fibrosarcoma M5076 fibrosarcoma
  • human A2780 ovarian carcinomas in nude mice. Tumor passage occurred biweekly for murine tumors and approximately every two to three weeks for the human tumor line.
  • M5076 and M5076ddp tumors were implanted in (C57B1/6 x DBA/2 )F1 hybrid mice, and human tumors were implanted in nude mice. All tumor implants for efficacy testing were subcutaneous (sc) .
  • the required number of animals needed to detect a meaningful response were pooled at the start of the experiment and each was given a subcutaneous implant of a tumor fragment (* 50 mg) with a 13-gauge trocar.
  • the animals were again pooled before distribution to the various treatment and control groups.
  • tumors were allowed to grow to the predetermined size window (tumors outside the range were excluded) and animals were evenly distributed to various treatment and control groups.
  • Treatment of each animal was based on individual body weight. Treated animals were checked daily for treatment related toxicity/mortality. Each group of animals was weighed before the initiation of treatment (Wtl) and then again following the last treatment dose (Wt2) . The difference in body weight (Wt2-Wtl) provides a measure of treatment-related toxicity.
  • Tumor response was determined by measurement of tumors with a caliper twice a week, until the tumors reach a predetermined "target" size of 1 gm.
  • Tumor weights (mg) were estimated from the formula:
  • Tumor weight (length x width2) ⁇ 2
  • Antitumor activity was evaluated at the maximum tolerated dose (MTD) which is defined as the dose level immediately below which excessive toxicity (i.e. more than one death) occurred.
  • MTD maximum tolerated dose
  • OD OD
  • Treated mice dying prior to having their tumors reach target size were considered to have died from drug toxicity.
  • Treatment groups with more than one death caused by drug toxicity were considered to have had excessively toxic treatments and their data were not included in the evaluation of a compound's antitumor efficacy.
  • Tumor response end-point was expressed in terms of tumor growth delay (T-C value) , defined as the difference in time (days) required for the treated tumors (T) to reach a predetermined target size compared to those of the control group (C) .
  • T-C value tumor growth delay
  • the tumor volume doubling time was first calculated with the formula:
  • Combretastatin A-4 phosphate disodium salt an agent with a dual mechanism of action, was evaluated for in vivo antitumor activity with the Peak Concentration Agents, cisplatin and carboplatin.
  • combretastatin A-4 phosphate disodium salt demonstrated significant antitumor activity against the cisplatin-resistant M5076DDP murine fibrosarcoma, producing 1.1 log cell kill. See Figure 1.
  • M5076DDP tumor model M5076DDP is a murine fibrosarcoma that has developed resistance to cisplatin and cross-resistance to carboplatin.
  • Combretastatin A-4 phosphate disodium salt treatment of mice bearing staged M5076DDP tumors using an everyday x 10 (Monday thru Friday) schedule produced moderate but significant antitumor effects.
  • combretastatin A-4 phosphate disodium salt yielded 1.1 log cell kill (LCK) .
  • Therapeutic synergy was achieved when combretastatin A-4 phosphate disodium salt was combined with cisplatin (administered simultaneously) in the treatment of advanced staged (300 mg) sc M5076DDP tumors.
  • Single agent cisplatin produced 0.8 LCK at its maximum tolerated dose (MTD) of 7.5 mg/kg/inj, q4dx3.
  • MMD maximum tolerated dose
  • the maximally tolerated combination of combretastatin A-4 phosphate disodium salt (250 mg/kg/inj) + Cisplatin (5 mg/kg/inj) yielded 2.0 LCK (Fig. 2A) .
  • Combretastatin A-4 phosphate disodium salt also produced synergistic antitumor activity when used in combination with carboplatin against large sc M5076 tumors (H300 mg) .
  • carboplatin produced 1.4 LCK, but with no tumor regression, at its MTD of 90 mg/kg/inj, iv, q4dx3.
  • the best combination yielded 2.0 LCK which was accompanied by significant tumor shrinkage (Fig 3A) .
  • Carboplatin (“CB-pt”) and CA4P are preferably administered more or less simultaneously. Most preferably carboplatin is administered immediately before CA4P.
  • the tumor model shown in this graph is M5076ddp (a platinum resistant variant of M5076 murine fibrosarcoma) .
  • Combretastatin A-4 has demonstrated robust therapeutic synergism with cisplatin and carboplatin as shown herein.
  • the doses of combretastatin A-4 phosphate disodium salt (CA4P) used in these previous combination studies has in general been between 100-250 mg/kg (200- 750 mg/m 2 ) .
  • Current human pharmacokinetics data indicate that preferred CA4P dosing is considerably lower (-50-60 mg/m 2 ) .
  • a study was therefore conducted to determine the minimum CA4P dose needed for combination therapy with carboplatin in the modestly carboplatin resistant murine fibrosarcoma M5076/DDP.
  • CA4P at doses as low as 12.5-25 mg/m 2 were sufficient to enhance the antitumor activity of carboplatin administered at a range of dose levels. See Figures 7A, 7B and 7C.
  • the present invention contemplates, for example, the administration of a combretastatin A-4 compound, such as CA4P with paclitaxel or with paclitaxel and carboplatin.
  • a combretastatin A-4 compound such as CA4P with paclitaxel or with paclitaxel and carboplatin.
  • a number of studies were conducted to determine an optimal treatment schedule, i.e, the sequence or the order, in which the two agents, CA4P and paclitaxel are administered.
  • CA4P is a tubulin depolymerizer while paclitaxel is a tubulin polymerizer, thus there may be potential for interaction at the tubulin level; and 2) CA4P inhibits tumor blood flow which may affect the regional micro-pharmacokinetics of paclitaxel in the tumors as well as the tumoral proliferative state.
  • Fig. 8 the 16/c mammary carcinoma model
  • BR96-sFv-PE40 Combination chemotherapy with immunotoxin Studies assessing the efficacy of combined administration of CA4P with the immunotoxin BR96-sFv- PE40 were also conducted. The construction of the immunotoxin is described in Siegall et al . (1994) J. of Immunology 152:2377-2384. Five groups of 5 rats each were inoculated intrahepatically with 1.5 x 10 5 wild type colon cancer cells (BN7005-H1D2) on day 0. BR96-sFv-PE40 is an immunoconjugate of BR96 monoclonal antibody and Pseudomonas toxin PE40. BR96 recognizes Lewis y antigen on the colon carcinoma BN7005 rat tumor.
  • the immunotoxin was inoculated at 2 different dose levels as indicated (125 ⁇ g/kg or 150 ⁇ g/kg respectively) , on days 9, 12, 14, 16, 19, 21 and 23.
  • Combretastatin A4- phosphate prodrug was administered ip 4-6 hours prior to the administration of the immunotoxin when administered on the same day as the immunotoxin on days 7, 8, 9, 12, 13, 14, 14, 16, 19 and 20. All rats were lapartomized on day 28 and liver tumor sizes measured by a caliper and tumor volume calculated.
  • Combretastatin A-1 phosphate disodium salt an agent with a dual mechanism of action, was evaluated for in vivo antitumor activity with the Peak Concentration Agents, carboplatin, and cisplatin.
  • combretastatin A-1 phosphate disodium salt demonstrated modest antitumor activity against the cisplatin-resistant M5076DDP murine fibrosarcoma.
  • therapeutic synergy was observed with both cisplatin and carboplatin.
  • combretastatin A-1 phosphate disodium salt significantly inhibited tumor blood flow in both A2780 human ovarian tumor xenografts in mice and N87 gastric cancer tumor xenografts .
  • CAlP demonstrated equivalent blood flow inhibition to that observed with CA4P in human tumor xenografts in nude mice but was 5-10 times more potent. Additionally, CAlP has demonstrated improved single agent activity in human tumor xenograft models, including N87 human gastric carcinoma, and the A2780 ovarian carcinoma. In A2780, CAlP achieved 2.1 LCK at its MTD of 9 mg/kg, ip, qldx ⁇ , compared to 1.1 LCK for CA4P at 150 mg/kg, ip. See Figure 12. Combination chemotherapy with carboplatin and cisplatin As shown in Figure 13, combination chemotherapy demonstrated that CAlP enhanced the antitumor activity of carboplatin in a manner similar to what had been observed for CA4P.
  • Synergistic antitumor activity was also demonstrated.
  • the minimum effective dose required for synergistic enhancement was considerably lower for CAlP (4-8 mg/kg) as compared to CA4P (25-50 mg/kg) .
  • CAlP is administered in combination with carboplatin
  • synergistic antitumor activity producing complete response was observed. When either agent was administered alone, this response was not observed. See Figure 14.
  • CAlP was administered in combination with cisplatin in a CaNT breasts tumor model I .
  • combined administration of cisplatin and CAlP acted synergistically to reduce tumor size.
PCT/US2001/050261 2000-12-22 2001-12-20 Methods for modulating tumor growth and metastasis WO2002056692A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
CA2432792A CA2432792C (en) 2000-12-22 2001-12-20 Methods for modulating tumor growth and metastasis
EP01994435A EP1351573A4 (en) 2000-12-22 2001-12-20 METHOD FOR MODULATING TUMOR WATCHTOWER AND METASTASIS
JP2002557211A JP2004523517A (ja) 2000-12-22 2001-12-20 腫瘍増殖および転移を調節するための方法
AU2002246827A AU2002246827B2 (en) 2000-12-22 2001-12-20 Methods for modulating tumor growth and metastasis

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US25819500P 2000-12-22 2000-12-22
US60/258,195 2000-12-22

Publications (1)

Publication Number Publication Date
WO2002056692A1 true WO2002056692A1 (en) 2002-07-25

Family

ID=22979500

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2001/050261 WO2002056692A1 (en) 2000-12-22 2001-12-20 Methods for modulating tumor growth and metastasis

Country Status (5)

Country Link
EP (1) EP1351573A4 (it)
JP (2) JP2004523517A (it)
AU (1) AU2002246827B2 (it)
CA (1) CA2432792C (it)
WO (1) WO2002056692A1 (it)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1068870A1 (en) * 1998-04-03 2001-01-17 Ajinomoto Co., Inc. Antitumor agents
US6562834B2 (en) 2000-10-27 2003-05-13 Aventis Pharma S. A. Combination comprising camptothecin and a stilbene derivative for the treatment of cancer
US6933320B2 (en) * 2001-03-15 2005-08-23 Aventis Pharma S. A. Combination comprising combretastatin and anticancer agents
JP2006503872A (ja) * 2002-10-09 2006-02-02 ユニバイオスクリーン エス.アー. 抗腫瘍活性および抗毒活性のある抽出物
JP2006503852A (ja) * 2002-09-25 2006-02-02 ユニバーシティー オブ ロチェスター 抗癌薬としてのカスパーゼインヒビター
JP2006508980A (ja) * 2002-11-15 2006-03-16 テリック,インコーポレイテッド Gst活性化された抗癌化合物と他の抗癌治療との併用癌治療
WO2006078422A2 (en) * 2004-12-22 2006-07-27 Oxigene, Inc. Methods for modulating tumor growth and metastasis
WO2009103076A1 (en) * 2008-02-15 2009-08-20 Oxigene, Inc. Methods and compositions for enhancing the efficacy of rtk inhibitors
WO2010067027A1 (fr) 2008-12-12 2010-06-17 Sanofi-Aventis Combinaison antitumorale associant l'ave8062 et le docetaxel
EP2219451A1 (en) * 2007-11-21 2010-08-25 Oxigene, Inc. Method for treating hematopoietic neoplasms
US9050378B2 (en) 2003-12-10 2015-06-09 Board Of Regents, The University Of Texas System N2S2 chelate-targeting ligand conjugates
EP3378473A4 (en) * 2016-08-26 2019-07-31 Dongguan Anhao Pharmaceutical Co., Ltd. MEDICINE COMBINATION WITH ANTITUMOR EFFECT
US10814013B2 (en) 2006-10-05 2020-10-27 The Board Of Regents Of The University Of Texas System Efficient synthesis of chelators for nuclear imaging and radiotherapy: compositions and applications
US11419934B2 (en) 2015-08-18 2022-08-23 Oncotelic Therapeutics, Inc. Use of VDAS to enhance immunomodulating therapies against tumors

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1351573A4 (en) * 2000-12-22 2007-03-28 Bristol Myers Squibb Co METHOD FOR MODULATING TUMOR WATCHTOWER AND METASTASIS
EP2047858A1 (en) * 2007-10-10 2009-04-15 Institut National De La Sante Et De La Recherche Medicale (Inserm) Combination products for treating cancer

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4996237A (en) 1987-01-06 1991-02-26 Arizona Board Of Regents Combretastatin A-4
US5409953A (en) 1987-01-06 1995-04-25 Arizona Board Of Regents Acting On Behalf Of Arizona State University Isolation, structural elucidation and synthesis of novel antineoplastic substances denominated "combretastatins"
US5561122A (en) 1994-12-22 1996-10-01 Arizona Board Of Regents Acting On Behalf Of Arizona State University Combretastatin A-4 prodrug
WO1999035150A1 (en) 1998-01-09 1999-07-15 Arizona Board Of Regents, A Body Corporate, Acting On Behalf Of Arizona State University Synthesis of combretastatin a-4 prodrugs and trans-isomers thereo f
WO2000048606A1 (en) 1999-02-18 2000-08-24 Oxigene, Inc. Compositions and methods for use in targeting vascular destruction

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9903404D0 (en) * 1999-02-16 1999-04-07 Angiogene Pharm Ltd Methods of treatment and compositions useful for the treatment of diseases involving angiogenesis
EP1351573A4 (en) * 2000-12-22 2007-03-28 Bristol Myers Squibb Co METHOD FOR MODULATING TUMOR WATCHTOWER AND METASTASIS

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4996237A (en) 1987-01-06 1991-02-26 Arizona Board Of Regents Combretastatin A-4
US5409953A (en) 1987-01-06 1995-04-25 Arizona Board Of Regents Acting On Behalf Of Arizona State University Isolation, structural elucidation and synthesis of novel antineoplastic substances denominated "combretastatins"
US5561122A (en) 1994-12-22 1996-10-01 Arizona Board Of Regents Acting On Behalf Of Arizona State University Combretastatin A-4 prodrug
WO1999035150A1 (en) 1998-01-09 1999-07-15 Arizona Board Of Regents, A Body Corporate, Acting On Behalf Of Arizona State University Synthesis of combretastatin a-4 prodrugs and trans-isomers thereo f
WO2000048606A1 (en) 1999-02-18 2000-08-24 Oxigene, Inc. Compositions and methods for use in targeting vascular destruction

Non-Patent Citations (19)

* Cited by examiner, † Cited by third party
Title
"Controlled Drug Bioavailability", 1984, WILEY, article "Drug Product Design and Performance"
"Medical Applications of Controlled Release", 1974, CRC PRESS
"Remington's Pharmaceutical Sciences", 1990, MACK PUBLISHING CO., pages: 1435 - 1712
BEAR ET AL., PROC. NATL. ACAD. SCI. USA, vol. 86, 1989, pages 7495 - 7499
BUCHWALD ET AL., SURGERY, vol. 88, 1980, pages 507
DURING ET AL., ANN. NEUROL., vol. 25, 1989, pages 351
FISHMAN ET AL.: "Medicine", 1985, J. B. LIPPINCOTT CO.
GILKS ET AL., MOL. CELL BIOL., vol. 13, 1993, pages 1759 - 1768
GOODSON, MEDICAL APPLICATIONS OF CONTROLLED RELEASE, vol. 2, 1984, pages 115 - 138
HOWARD ET AL., J. NEUROSURG., vol. 71, 1989, pages 105
LANGER, SCIENCE, vol. 249, 1990, pages 1527 - 1533
LEVY ET AL., SCIENCE, vol. 228, 1985, pages 190
MCCORMICK, NATURE, vol. 363, 1993, pages 15 - 16
RANGER; PEPPAS, J. MACROMOL. SD. REV. MACROMOL. CHEM., vol. 23, 1983, pages 61
ROBBINS; ANGELL: "Basic Pathology", 1976, W.B. SAUNDERS CO., pages: 68 - 79
SAUDEK ET AL., N. ENGL. J. MED., vol. 321, 1989, pages 574
See also references of EP1351573A4
SEFTON, CRC CRIT. REF. BIOMED. ENG., vol. 14, 1987, pages 201
TREAT ET AL.: "Liposomes in the Therapy of Infectious Disease and Cancer", LISS, pages: 317 - 327

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7973076B2 (en) 1998-04-03 2011-07-05 Ajinomoto Co., Inc. Anti-tumor composition
EP1068870A4 (en) * 1998-04-03 2003-02-12 Ajinomoto Kk ANTITUMOR AGENTS
US7655696B2 (en) 1998-04-03 2010-02-02 Ajinomoto Co., Inc. Anti-tumor composition
US6992106B2 (en) 1998-04-03 2006-01-31 Ajinomoto Co., Inc. Anti-tumor composition
EP1068870A1 (en) * 1998-04-03 2001-01-17 Ajinomoto Co., Inc. Antitumor agents
US6562834B2 (en) 2000-10-27 2003-05-13 Aventis Pharma S. A. Combination comprising camptothecin and a stilbene derivative for the treatment of cancer
US6933320B2 (en) * 2001-03-15 2005-08-23 Aventis Pharma S. A. Combination comprising combretastatin and anticancer agents
JP2006503852A (ja) * 2002-09-25 2006-02-02 ユニバーシティー オブ ロチェスター 抗癌薬としてのカスパーゼインヒビター
JP4944380B2 (ja) * 2002-10-09 2012-05-30 ユニバイオスクリーン エス.アー. 抗腫瘍活性および抗毒活性のある抽出物
JP2006503872A (ja) * 2002-10-09 2006-02-02 ユニバイオスクリーン エス.アー. 抗腫瘍活性および抗毒活性のある抽出物
JP2006508980A (ja) * 2002-11-15 2006-03-16 テリック,インコーポレイテッド Gst活性化された抗癌化合物と他の抗癌治療との併用癌治療
JP2010265305A (ja) * 2002-11-15 2010-11-25 Telik Inc Gst活性化された抗癌化合物と他の抗癌治療との併用癌治療
US9050378B2 (en) 2003-12-10 2015-06-09 Board Of Regents, The University Of Texas System N2S2 chelate-targeting ligand conjugates
WO2006078422A2 (en) * 2004-12-22 2006-07-27 Oxigene, Inc. Methods for modulating tumor growth and metastasis
WO2006078422A3 (en) * 2004-12-22 2007-02-08 Oxigene Inc Methods for modulating tumor growth and metastasis
US10814013B2 (en) 2006-10-05 2020-10-27 The Board Of Regents Of The University Of Texas System Efficient synthesis of chelators for nuclear imaging and radiotherapy: compositions and applications
US10925977B2 (en) 2006-10-05 2021-02-23 Ceil>Point, LLC Efficient synthesis of chelators for nuclear imaging and radiotherapy: compositions and applications
US9040500B2 (en) 2007-11-21 2015-05-26 Oxigene, Inc. Method for treating hematopoietic neoplasms
EP2219451A4 (en) * 2007-11-21 2011-01-19 Oxigene Inc METHOD FOR THE TREATMENT OF HEMOPOETIC NEOPLASMS
EP2219451A1 (en) * 2007-11-21 2010-08-25 Oxigene, Inc. Method for treating hematopoietic neoplasms
WO2009103076A1 (en) * 2008-02-15 2009-08-20 Oxigene, Inc. Methods and compositions for enhancing the efficacy of rtk inhibitors
WO2010067027A1 (fr) 2008-12-12 2010-06-17 Sanofi-Aventis Combinaison antitumorale associant l'ave8062 et le docetaxel
US11419934B2 (en) 2015-08-18 2022-08-23 Oncotelic Therapeutics, Inc. Use of VDAS to enhance immunomodulating therapies against tumors
EP3378473A4 (en) * 2016-08-26 2019-07-31 Dongguan Anhao Pharmaceutical Co., Ltd. MEDICINE COMBINATION WITH ANTITUMOR EFFECT

Also Published As

Publication number Publication date
JP2004523517A (ja) 2004-08-05
EP1351573A4 (en) 2007-03-28
CA2432792C (en) 2012-04-03
JP2009102350A (ja) 2009-05-14
EP1351573A1 (en) 2003-10-15
CA2432792A1 (en) 2002-07-25
AU2002246827B2 (en) 2008-02-21

Similar Documents

Publication Publication Date Title
JP2009102350A (ja) 腫瘍増殖および転移を調節するための方法
US20090258937A1 (en) Methods for Modulating Tumor Growth and Metastasis
AU2002246827A1 (en) Methods for modulating tumor growth and metastasis
US20090209496A1 (en) Methods and compositions for enhancing the efficacy of rtk inhibitors
WO2006078422A2 (en) Methods for modulating tumor growth and metastasis
US20070265213A1 (en) Use of Docetaxel/Doxorubicin/Cyclophosphamide in Adjuvant Therapy
US20030087839A1 (en) Combined preparations comprising morpholine anthracyclines and anticancer agent
PL205728B1 (pl) Kombinacja zawierająca kombretastatynę i czynniki przeciwnowotworowe
AU779922B2 (en) Formulations for parenteral use of estramustine phosphate and amino acids
JP2706371B2 (ja) 癌治療用補助剤及びそれを用いたキット
KR20090130098A (ko) 이소포스포르아미드 머스타드의 염 및 이의 동족체
EP1229916A2 (en) Combination chemotherapy
JP2006528696A (ja) 抗癌剤の抗腫瘍活性を増強する方法
AU2002216029B2 (en) A combination comprising camptothecin and a stilbene derivative for the treatment of cancer
AU2002216029A1 (en) A combination comprising camptothecin and a stilbene derivative for the treatment of cancer
AU5063302A (en) Combination chemotherapy
US20110110940A1 (en) Methods for Enhancing the Efficacy of Vascular Disrupting Agents
US20040014729A1 (en) Use of estramustine phosphate in the treatment of bone metastasis
US20030134893A1 (en) Combination chemotherapy

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US UZ VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2002557211

Country of ref document: JP

Ref document number: 2432792

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2001994435

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2002246827

Country of ref document: AU

WWP Wipo information: published in national office

Ref document number: 2001994435

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642