US20040248781A1 - Therapeutic method for reducing angiogenesis - Google Patents

Therapeutic method for reducing angiogenesis Download PDF

Info

Publication number
US20040248781A1
US20040248781A1 US10/203,399 US20339903A US2004248781A1 US 20040248781 A1 US20040248781 A1 US 20040248781A1 US 20339903 A US20339903 A US 20339903A US 2004248781 A1 US2004248781 A1 US 2004248781A1
Authority
US
United States
Prior art keywords
receptor
kit
chemotherapeutic agent
vegf
dose
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
US10/203,399
Inventor
Robert Kerbel
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.)
Sunnybrook Health Sciences Centre
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
Application filed by Individual filed Critical Individual
Priority to US10/203,399 priority Critical patent/US20040248781A1/en
Assigned to SUNNYBROOK HEALTH SCIENCE CENTER reassignment SUNNYBROOK HEALTH SCIENCE CENTER ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KERBEL, ROBERT
Publication of US20040248781A1 publication Critical patent/US20040248781A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies

Definitions

  • the present invention relates to the inhibition or prevention of angiogenesis as a means to control or treat an angiogenic dependent condition, a condition characterized by, or dependent upon, blood vessel proliferation.
  • the invention further relates to the use of an anti-angiogenic molecule in combination with a chemotherapeutic agent.
  • Angiogenesis is a highly complex process of developing new blood vessels that involves the proliferation and migration of, and tissue infiltration by capillary endothelial cells from pre-existing blood vessels, cell assembly into tubular structures, joining of newly forming tubular assemblies to closed-circuit vascular systems, and maturation of newly formed capillary vessels.
  • the molecular bases of many of these aspects are still not understood.
  • Angiogenesis is important in normal physiological processes including embryonic development, follicular growth, and wound healing, as well as in pathological conditions such as tumor growth and in non-neoplastic diseases involving abnormal neovascularization, including neovascular glaucoma (Folkman, J. and Klagsbrun, M. Science 235:442-447 (1987).
  • neoplastic diseases including but not limited to solid tumors, autoimmune diseases and collagen vascular diseases such as, for example, rheumatoid arthritis, and ophthalmalogical conditions such as diabetic retinopathy, retrolental fibroplasia and neovascular glaucoma
  • ophthalmalogical conditions such as diabetic retinopathy, retrolental fibroplasia and neovascular glaucoma
  • Conditions or diseases to which persistent or uncontrolled angiogenesis contribute have been termed angiogenic dependent or angiogenic associated diseases.
  • One means of controlling such diseases and pathological conditions comprises restricting the blood supply to those cells involved in mediating or causing the disease or condition.
  • solid tumors develop to a size of about a few millimeters, and further growth is not possible, absent angiogenesis within the tumor.
  • strategies to limit the blood supply to tumors have included occluding blood vessels supplying portions of organs in which tumors are present. Such approaches require the site of the tumor to be identified and are generally limited to treatment to a single site, or small number of sites.
  • An additional disadvantage of direct mechanical restriction of a blood supply is that collateral blood vessels develop, often quite rapidly, restoring the blood supply to the tumor.
  • TGF ⁇ transforming growth factor beta
  • aFGF and bFGF acidic and basic fibroblast growth factor
  • PDGF platelet derived growth factor
  • VEGF vascular endothelial growth factor
  • VEGF growth factor of particular interest
  • An endothelial-cell specific mitogen, VEGF acts as an angiogenesis inducer by specifically promoting the proliferation of endothelial cells. It is a homodimeric glycoprotein consisting of two 23 kD subunits with structural similarity to PDGF.
  • Four different monomeric isoforms of VEGF resulting from alternative splicing of mRNA have been identified. These include two membrane bound forms (VEGF 206 and VEGF 189 ) and two soluble forms (VEGF 165 and VEGF121).
  • VEGF 165 is the most abundant isoform in all human tissues except placenta.
  • VEGF is expressed in embryonic tissues (Breier et al., Development (Camb.) 114:521 (1992)), macrophages, and proliferating epidermal keratinocytes during wound healing (Brown et al., J. Exp. Med., 176:1375 (1992)), and maybe responsible for tissue edema associated with inflammation (Fenrara et al., Endocr. Rev. 13:18 (1992)).
  • VEGF vascular endothelial growth factor
  • human tumor lines including glioblastoma multiforme, hemangioblastoma, other central nervous system neoplasms and AIDS-associated Kaposi's sarcoma (Plate, K. et al. (1992) Nature 359: 845-848; Plate, K. et al. (1993) Cancer Res. 53: 5822-5827; Berkman, R. et al. (1993) J. Clin. Invest. 91: 153-159; Nakamura, S. et al. (1992) AIDS Weekly, 13 (1)).
  • High levels of VEGF also have been reported in hypoxia induced angiogenesis (Shweiki, D. et al. (1992) Nature 359: 843-845).
  • VEGF mediates its biological effect through high affinity VEGF receptors which are selectively expressed on endothelial cells during, for example, embryogenesis (Millauer, B., et al. (1993) Cell 72: 835-846) and tumor formation.
  • VEGF receptors typically are class III receptor-type tyrosine kinases characterized by having several, typically 5 or 7, immunoglobulin-like loops in their amino-terminal extracellular receptor ligand-binding domains (Kaipainen et al., J. Exp. Med. 178:2077-2088 (1993)).
  • VEGF receptors include flt-1, sequenced by Shibuya M. et al., Oncogene 5, 519-524 (1990); flk-1, sequenced by Matthews W. et al. Proc. Natl. Acad. Sci. USA, 88:9026-9030 (1991) and KDR, the human homologue of flk-1, described in PCT/US92/01300, filed Feb. 20, 1992, and in Terman et al., Oncogene 6:1677-1683 (1991).
  • High levels of flk-1 are expressed by endothelial cells that infiltrate gliomas (Plate, K. et al., (1992) Nature 359: 845-848), and are specifically upregulated by VEGF produced by human glioblastomas (Plate, K et al. (1993) Cancer Res. 53: 5822-5827).
  • the finding of high levels of flk-1 expression in glioblastoma associated endothelial cells (GAEC) suggests that receptor activity is induced during tumor formation, since flk-1 transcripts are barely detectable in normal brain endothelial cells. This upregulation is confined to the vascular endothelial cells in close proximity to the tumor.
  • VEGF activity results in inhibition of the growth of human tumor xenografts in nude mice (Kim, K. et al. (1993) Nature 362: 841-844), suggesting a direct role for VEGF in tumor-related angiogenesis.
  • chemotherapeutic drugs also have been shown to block functions of activated, dividing endothelial cells critical to angiogenesis, or to kill such cells.
  • Such collateral damaging effects on a genetically stable normal host cell in addition to the chemotherapeutic agent's effect upon the tumor cells, contribute significantly to the in vivo anti-tumor efficacy of chemotherapy.
  • the standard use of chemotherapeutic agents has obvious undesirable side-effects upon the normal cells of patients, limiting its use.
  • Administration of chemotherapeutic agents in their usual doses and at usual dosage frequencies are commonly associated with side-effects, including, but not limited to, myelosuppression, neurotoxicity, cardiotoxicity, alopecia, nausea and vomiting, nephrotoxicity, and gastrointestinal toxicity.
  • patients' tumors often also develop resistance to the chemotherapeutic agents after initial exposure to the drugs.
  • a desirable method and composition for controlling angiogenesis should be well tolerated, have few or no side-effects, and prevent new vessel formation at sites of disease without interfering with required physiologic angiogenesis in normal sites. It should be effective and, in the case of neoplastic disease, overcome the problem of the development of drug resistance by tumor cells. In so doing, it should permit targeted therapy without the accurate identification of all disease sites.
  • the present invention addresses many of the problems with existing materials and methods.
  • the present invention provides a method of treating an angiogenic dependent condition in a mammal comprising administering an anti-angiogenic molecule and a chemotherapeutic agent to the mammal, in an amount and frequency effective, in combination, to produce a regression or arrest of the condition without significant toxicity from the chemotherapeutic agent.
  • the angiogenic dependent condition may be selected from the group consisting of neoplasm, collagen-vascular disease or auto-immune disease, including a solid tumor neoplasm, including breast carcinoma, lung carcinoma, prostate carcinoma, colon carcinoma, prostate carcinoma, ovarian carcinoma, neuroblastoma, central nervous system tumor, neuroblastoma, glioblastoma multiforme or melanoma.
  • the mammal receiving the treatment is preferably a human.
  • the anti-angiogenic molecules inhibit the action of a vascular endothelium survival factor, which include receptors and their ligands.
  • Vascular endothelium survival factors include receptors, including angiogenic growth factors such as VEGF receptor, including flk-1/KDR receptor, or flt-4 receptor and VEGF. Examples of other vascular endothelial survival factors are integrin ⁇ v ⁇ 3 .
  • ⁇ v ⁇ 3 ligand Tie2/tek ligand, Tie2/tek, endoglin ligand, endoglin, neuropilin ligand, neuropilin, thrombospondin ligand, thrombospondin, PDGF ⁇ , PDGF ⁇ receptor, PDGF ⁇ , PDGF ⁇ receptor, aFGF, aFGF receptor, bFGF, bFGF receptor, TGF ⁇ , TGF ⁇ receptor, EGF, EGF receptor, angiostatin, angiostatin receptor, angiopoetin, angiopoeitin receptor, PLGF, PLGF receptor, VPF, or VPF receptor.
  • the ligand is selected from the group consisting of VEGF (VEGF-A), VEGF-B, VEGF-C, or VEGF-D.
  • VEGF-A VEGF
  • VEGF-B VEGF-B
  • VEGF-C VEGF-C
  • VEGF-D VEGF-D
  • the anti-angiogenic molecule may be selected from the group consisting of antibody, antibody fragment, small molecule or peptide.
  • Preferred embodiments of the present invention include antibodies selected from the group consisting of mouse antibody, rat antibody, chimeric antibody, humanized antibody or human antibody.
  • a preferred antibody is IMC-1C11.
  • IMC-1C11 is administered in a dose of from about 5 mg/m 2 to about 700 mg/m 2 about daily to about every 7 days, more preferably a dose of from about 7.5 mg/m 2 to about 225 mg/m 2 , about twice per week.
  • the IMC-1C11 is administered at a dose and frequency sufficient to substantially saturate the VEGF receptor.
  • the anti-angiogenic molecule is administered in a dose and frequency sufficient to substantially saturate the target of the anti-angiogenic molecule.
  • the anti-angiogenic molecule is administered in a dose equivalent to that of IMC-1C11, administered in a dose of from about 5 mg/m 2 to about 700 mg/m 2 about daily to about every 7 days, more preferably a dose of from about 7.5 mg/m 2 to about 225 mg/m 2 , about twice per week.
  • the chemotherapeutic agent may be selected from the group consisting of vinca alkaloid, camptothecan, taxane, or platinum analogue, including vincristine, vinblastine, vinorelbine, vindesine, paclitaxel, docetaxel, 5 FU, cisplatin, carboplatin, iranotecan, topotecan or cyclophosphamide.
  • the chemotherapeutic agent is administered in a low-dose regimen.
  • the chemotherapeutic agent is administered at less than about 50% of the maximum tolerated dose, more preferably at less than about 20% of the maximum tolerated dose, most preferably at less than about 10% of the maximum tolerated dose.
  • the vinblastine is administered in a dose from about 0.5 mg/m 2 to about 3 mg/m 2 from about once every 3 days to about once every 7 days.
  • the chemotherapeutic agent is administered in a dosage and frequency that is of substantially equivalent efficacy to vinblastine in a dose from about 0.5 mg/m 2 to about 3 mg/m 2 from about once every 3 days to about once every 7 days.
  • the chemotherapeutic agent is administered more frequently than about every three weeks, or more frequently than about every seven days.
  • the present invention also includes a kit for treating an angiogenic dependent condition in a mammal comprising the anti-angiogenic molecule and the chemotherapeutic agent, which are provided to be administered in an amount and frequency effective, in combination, to produce a regression or arrest of the condition while minimizing or preventing significant toxicity of the chemotherapeutic agent.
  • FIG. 1 is the encoding nucleotide sequence and deduced amino acid sequence of V H and V L domains of IMC-1C11 (c-p1C11).
  • the present invention comprises a method of treating or controlling an angiogenic dependent condition in a mammal, comprising administering an anti-angiogenic molecule and a chemotherapeutic agent in combined amounts effective to produce a regression or arrest to the angiogenic dependent condition, while minimizing or preventing significant toxicity.
  • the benefits of the combination of an anti-angiogenic molecule and a chemotherapeutic agent of the present invention include an improvement in the treatment and control of an angiogenic dependant condition with reduced doses of a chemotherapeutic agent administered at increased frequency, without significant toxicity, together with a anti-angiogenic molecule.
  • the combination can be administered for a prolonged period of time, or optionally a shorter duration of treatment may be administered due to the increased effectiveness of the combination. Toxicity is reduced or eliminated without a loss of effectiveness.
  • the administration of the treatment of the invention can overcome the problems of drug resistance that develops with standard chemotherapeutic regimens.
  • the anti-angiogenic molecule functions to inhibit or prevent angiogenesis and thereby treating the angiogenic dependent condition by inhibiting, blocking, or antagonizing the effect of vascular endothelial survival factors.
  • survival factors are receptors and their ligands, upon which vascular endothelium depends, either directly or indirectly, for growth and/or survival. They play a role in allowing vascular endothelial cells to recovery from injury or insult, which, absent the effect of the survival factor would result in cell death or apoptosis.
  • Survival factors include vascular endothelial cell growth factors or mitogens, as well as those factors which do not appear to have a direct growth-stimulatory effect but allow the cells to recover from injury.
  • survival factors include VEGF receptors, including but not limited to flt-1 (VEGFR1), flk-1/KDR (VEGFR2), flt-4 (VEGFR3), their ligands VEGF, VEGF-B, VEGF-C, and VEGF-D, integrin ⁇ V ⁇ 3, Tie2/tek, endoglin (CD105), neuropilin, thrombospondin and their ligands, and PDGF ⁇ , PDGF ⁇ , aFGF, bFGF, and TGF ⁇ , as well as EGF, angiostatin, and angiopoeitin, vascular permiability factor (VPF), and placenta-like growth factor (PLGF) and their receptors.
  • VEGF receptors including but not limited to flt-1 (VEGFR1), flk-1/KDR (VEGFR2), flt-4 (VEGFR3), their ligands VEGF, VEGF-B
  • the survival factors that are receptors are located on vascular endothelial cells or may be located on other cell types including, but not limited to, tumor cells.
  • the anti-angiogenic molecule inhibit binding to ,and/or activation of, receptors, inhibit their expression, or inhibit the binding of or expression of ligands.
  • Suitable anti-angiogenic molecules include, but are not limited to antibody, antibody fragment, small molecule or peptide.
  • An antibody can be derived from any mammalian species.
  • the antibody is of mouse, rat, rabbit, or human origin.
  • the antibody is chimeric, more preferably the antibody is humanized, and even more preferably the antibody is human.
  • Suitable antibody fragments include, for example, Fab fragment, Fab′ fragment, F(ab′) 2 fragment, monovalent single chain antibody (scFv), and diabodies (DAB).
  • VEGF receptor antagonist or VEGF antagonist
  • aFGF receptor antagonist as disclosed in U.S. Pat. Nos. 5,840,301, 5,861,499, 5,874,542, 5,955,311, and 5,730,977, which are incorporated in their entirety by reference, aFGF receptor antagonist, aFGF antagonist, bFGF receptor antagonist, bFGF antagonist, PDGF receptor antagonist, PDGF antagonist, TGF ⁇ antagonist, Tie2/tek antagonist (P. Lin et al., Inhibition of Tumor Angiogenesis Using a Soluble Receptor Establishes a Role for Tie2 in Pathologic Vascular Growth.
  • endoglin (CD105) antagonist as disclosed in U.S. Pat. Nos. 5,855,866, and 5,660,827, neuropilin antagonist, thrombospondin antagonist, and antagonists to the receptors for PDGF ⁇ , PDGF ⁇ , aFGF, bFGF, or TGF ⁇ , as well as antagonists to the receptors for EGF, angiostatin, angiopoeitin, or VPF (Vascular Permeability Factor) as disclosed in U.S. Pat. Nos. 5,036,003 and 5,659,013. Also encompassed within the scope of the present invention are integrin receptor antagonists as disclosed in U.S. Pat. Nos.
  • integrin ⁇ V ⁇ 3 antagonists as disclosed in U.S. Pat. Nos. 5,780,426, 5,773,412, 5,767,071, 5,759,996, 5,753,230, 5,652,110, and 5,652,109, antagonists to placenta-like growth factor (PLGF) as disclosed in European Patent Application EP506477A1, thrombospondin antagonists as disclosed in U.S. Pat. Nos.
  • PLGF placenta-like growth factor
  • the expression of a receptor and/or ligand is upregulated in an region of angiogenesis.
  • the cells of the vascular endothelium are largely normal and responsive to normal regulatory mechanisms.
  • An advantage to blocking a receptor, rather than the ligand, is that are that fewer anti-angiogenic molecules may be needed to achieve such inhibition, as levels of receptor expression may be more constant than those of the environmentally induced ligand. Because the receptors exist on essentially normal endothelial cells, their behavior is less likely to escape normal regulatory control.
  • antagonism of the receptor is combined with antagonism of the ligand in order to achieve even more efficient inhibition of angiogenesis.
  • the anti-angiogenic molecule is an antagonist to VEGF or the VEGF receptor.
  • the expression of the VEGF receptor and ligand is low in normal endothelial cells that are not in or near a region of angiogenesis.
  • VEGF receptors present on tumor infiltrating vascular endothelial cells are upregulated, as is the expression of the VEGF ligand by tumor cells.
  • VEGF (or VEGF-A) is the ligand for VEGFR1 and VEGFR2
  • VEGF-B is the ligand for VEGFR2
  • VEGF-C is the ligand for VEGFR3, VEGFR4, and possibly VEGFR2
  • VEGF-D is the ligand for VEGFR2 and VEGFR3.
  • Blocking the interaction between VEGF and its receptors can inhibit angiogenesis, and thereby tumor growth, while not significantly effecting normal endothelial cells at other sites, where vascular endothelial cell receptors have not been upregulated.
  • antagonism of the VEGF receptor is combined with antagonism of the VEGF ligand in order to achieve even more efficient inhibition of angiogenesis.
  • VEGF VEGF-A
  • VEGF-B VEGF-B
  • VEGF-C VEGF-D
  • VEGF-D is the ligand which is inhibited by the anti-angiogenesis molecule.
  • the effect of more than one form of VEGF is inhibited.
  • Examples of an antagonist to a receptor are the antibodies DC101, described in the Examples, an antagonist of flk-1, and A.4.6.1 and its chimeric and humanized form as disclosed in L. G. Presta, Humanization of an Anti-vascular Endothelial Growth Factor Monoclonal Antibody for the Therapy of Solid Tumors and Other Disorders. Cancer Research, 57, 4593-4599 (1997), which is hereby incorporated by reference.
  • a preferred antibody is the mouse-human chimeric antibody IMC-1C11 which is a KDR antagonist, and is disclosed in U.S. application Ser. No. 09/240,736, which is hereby incorporated by reference.
  • the encoding nucleotide sequences and deduced amino acid sequences of the V H and V L domains are shown in FIG. 1.
  • the present invention provides a low dose application of a chemotherapeutic agent that provides effective therapy without significant side-effects.
  • An effective amount of a chemotherapeutic agent is an amount sufficient to inhibit or contribute to the inhibition of angiogenesis, when administered with an anti-angiogenic molecule, without resulting in significant toxicity.
  • the meaning of significant toxicity is well known to one of ordinary skill in the art, and includes toxicities that cumulatively or acutely effect a patient's quality of life and/or limit the amount of chemotherapeutic agent than can be administered.
  • Examples of chemotherapy induced toxicity that can be minimized or prevented by the present invention include, but are not limited to, myelosuppression, neurotoxicity, cardiotoxicity, alopecia, nausea and vomiting, nephrotoxicity, and gastrointestinal toxicity.
  • the low dose administration of a chemotherapeutic agent without significant toxicity permits prolonged treatment if desired.
  • the low dose manner of chemotherapy administration in the present invention can overcome the problem of the development of chemotherapeutic drug resistance by the patient's tumor cells that occurs with current chemotherapeutic regimens.
  • the present invention delays, reduces, or even circumvents the problem of acquired drug resistance by targeting the genetically stable endothelial cells of newly formed tumor blood vessels, rather than genetically unstable tumor cells which are prone to mutate and develop resistance.
  • Encompassed within the scope of the present invention is the administration of amounts of chemotherapy that are insufficient to have a cytotoxic effect on tumor cells yet have anti-angiogenic properties as a result of the drug's effect on vascular endothelial cells.
  • the chemotherapeutic agent of the present invention functions, in combination with the anti-angiogenic molecule, to cause a cytotoxic effect on the vascular endothelial cells involved in angiogenesis.
  • a number of chemotherapeutic agents have been identified as having anti-angiogenic activity and are suitable for use in the practice of the present invention.
  • Examples include, but are not limited to, taxanes, including but not limited to paclitaxel and docetaxel, camptothecin analogues, including but not limited to iranotecan and topotecan, platinum analogues including but not limited to cisplatin and carboplatin, 5FU, and vinca alkaloid, including but not limited to vinblastine, vincristine vindesine and vinorelbine.
  • taxanes including but not limited to paclitaxel and docetaxel
  • camptothecin analogues including but not limited to iranotecan and topotecan
  • platinum analogues including but not limited to cisplatin and carboplatin
  • 5FU and vinca alkaloid, including but not limited to vinblastine, vincristine vindesine and vinorelbine.
  • chemotherapeutic agents to achieve therapeutic effects without significant toxicity (side effects) is routinely possible by the practice of the present invention.
  • a low-dose regimen will administer the chemotherapy at frequent intervals or continually, at less than about 50% [Should this be lower?] of the maximum tolerated dose (MTD), more preferably less than about 20% of the MTD, and most preferably, less than about 10% of the MTD, although the preferred dose depends on the particular chemotherapy.
  • MTD maximum tolerated dose
  • the preferred dose will be a dose effective to inhibit or prevent progression of the angiogenic dependent condition, when administered in combination with the anti-angiogenic molecule of the present invention, while minimizing or preventing the development of significant chemotherapy related toxicity.
  • the dose of chemotherapy will be effective to inhibit or prevent progression of the angiogenic dependent condition even when administered alone.
  • the dose of chemotherapy will be one which does not exert a direct cytotoxic effect on tumor cells, yet has an antitumor effect mediated by its anti-angiogenic properties.
  • the low-dose regimen of the present invention will administer chemotherapy at a dose intensity of less than 50% of that when administered alone to treat a particular neoplasm. More preferably the low-dose regimen of the present invention will administer chemotherapy at a dose intensity of less than 20% of that when administered alone to treat a particular neoplasm.
  • chemotherapy is usually given intermittently, commonly in the form of a bolus infusion or an infusion lasting from about 20 minutes to about three hours, at about the maximum tolerated dose (MTD) with long rest periods (e.g., 3 weeks) between successive drug exposures. It has been suggested that these rest periods provide the endothelial cell compartment of a tumor an opportunity to repair some of the damage inflicted by the chemotherapy (T. Browder, et al., Antiangiogenic Scheduling of Chemotherapy Improves Efficacy against Experimental Drug-Resistant Cancer. Cancer Res . (In press) 1999).
  • MTD maximum tolerated dose
  • a chemotherapeutic drug such as cyclophosphamide
  • Administering lower doses of a chemotherapeutic drug, such as cyclophosphamide, more frequently such as weekly, enables circumvention of many problems associated with standard chemotherapeutic doses.
  • This anti-angiogenic scheduling of chemotherapy optimizes antitumor/anti-vascular effects.
  • a sub-line of the Lewis Lung Carcinoma previously selected in vivo for acquired resistance to the MTD of cyclophosphamide, is rendered sensitive again to the drug in vivo by employing continuous low dose therapy of the same drug.
  • the invention provides low-dose administration of chemotherapy administered at short intervals, for example, from about every 4 to about every 6 hours, to about daily to weekly, or optionally administered continuously.
  • the preferred time interval between administration of successive doses of chemotherapeutic agent is that amount of time that is of sufficiently short duration that the blood levels of the chemotherapeutic agent (or its active metabolite) will remain at about a concentration sufficient to exert an anti-angiogenic effect for substantially the duration of treatment.
  • a blood level will be maintained for at least about 30% [Should this be lower] of the time between doses, more preferably for at least 50% of the time between doses, most preferably for at least about 70% of the time between doses.
  • Treatment is continued for a period of time from about 10 days to about 6 months, or as determined by one of skill in the art.
  • treatment will continue chronically for a period longer than six months for as long as is needed.
  • the present invention reduces host toxicity, allows for longer term administration of the chemotherapeutic agent in diseases or pathological conditions requiring it, and does not sacrifice, and perhaps even improves, antitumor efficacy.
  • increased efficacy will permit the use of shorter durations of therapy for selected angiogenic dependent conditions.
  • the anti-angiogenic molecule is administered in dosages and dose frequencies sufficient to substantially saturate the selected target receptor or ligand.
  • Substantial saturation is saturation of at least about 50% of targeted receptors.
  • a more preferred level of saturation is at least about 80%, and a most preferred level of saturation is about 100%.
  • the anti-angiogenic molecule is administered at a dose and frequency sufficiently short to maintain a blood level sufficient to saturate the targeted survival factor for at least about 50% of the time between doses, more preferably at least about 70% of the time and most preferably at least about 90% of the time interval between doses.
  • concentrations required to achieve receptor saturation or ligand neutralization in vitro, and by analysis of serum concentrations of anti-angiogenic molecule in vivo both the appropriate dose and schedule can be determined readily by one of skill in the art.
  • a preferred embodiment of the invention is the combination of a chemotherapeutic agent and a VEGF receptor antagonist. It has been shown that a major function of VEGF is to promote the survival of endothelial cells comprising newly formed vessels (L. E. Benjamin, et Al., Selective Ablation of Immature Blood Vessels in Established Human Tumors followss Vascular Endothelial Growth Factor Withdrawal. J.Clin.Invest . 103:159-165 (1999), T. Alon, et al., Vascular Endothelial Growth Factor Acts as a Survival Factor for Newly Formed Retinal Vessels and Has Implications for Retinopathy of Prematurity. Nature Med .
  • a preferred embodiment of the present invention comprises the administration of the antibody IMC-1C11, a KDR receptor antagonist with a chemotherapeutic agent.
  • a preferred dose of IMC-1C11 is an amount that is sufficient to adequately saturate the targeted receptors or ligand.
  • 50% saturation of VEGF receptors was obtained as an IMC-1C11 concentration of 0.2 ⁇ g/ml, and 100% at a concentration of 3 ⁇ g/ml.
  • a preferred level of saturation is about at least 50%, a more preferred level is about at least 80%, and a most preferred level is about 100% saturation.
  • a preferred dose regimen of IMC-1C11 is from about 5 mg/m 2 to about 700 mg/M 2 , more preferably from about 7.5 mg/M 2 to about 225 mg/m 2 , administered about twice per week.
  • Another preferred embodiment of the invention combines IMC-1C11 in the doses described above with, vinblastine, administered in a low dose regimen, at a dose from about 0.5 mg /m 2 to about 3 mg/m 2 from about every 3 days to about every 7 days.
  • a suitable chemotherapeutic agent other than vinblastine is administered in a dosage and frequency that is of substantially equivalent efficacy to vinblastine (in the combination) at a dose from about 0.5 mg /m 2 to about 3 mg/m 2 from about every 3 days to about every 7 days.
  • doses of an anti-angiogenic molecule in amounts and dosing frequencies sufficient to provide levels of receptor or ligand saturation equivalent to that of IMC-1C11 in the doses about are combined with a chemotherapeutic agent in a dose and frequency equivalent to that of vinblastine above, and therapy is carried out for as long as is needed.
  • An equivalent dose is one that, in the combination, is substantially as effective in arresting or inhibiting the angiogenic dependent condition, while being substantially as effective in minimizing or preventing significant chemotherapy induced toxicity.
  • an equivalent dose of another chemotherapeutic agent is determined based on data derived from an animal model, an example of which is included herein, utilizing a chemotherapy-resistant cell line so that any observed antitumor effect is due to an effect on the vascular endothelium.
  • a preferred dose of vinblastine in a mouse is from about 1 mg/m 2 to about 2 mg/m 2 more preferably about 1.5 mg/m 2 administered every three days.
  • the MTD of this drug in mice is approximately 4-5 times that of a human, and a preferred dose is ⁇ fraction (1/16) ⁇ - ⁇ fraction (1/20) ⁇ of the MTD in mice.
  • a preferred dose of DC011 in a mouse is about 800 ⁇ g administered intraperitoneally every three days.
  • DC101 and vinblastine showed a therapeutic effect upon neuroblastoma cell lines grown as xenografts in SCID mice.
  • L. Witte, L, et al. Monoclonal antibodies targeting the VEGF receptor-2 (flk1/KDR) as an anti-angiogenic therapeutic strategy. Cancer Metastasis Rev . 17:155-161. (1998); Prewitt, 1999).
  • low-dose vinblastine is administered every 3 days in combination with IMC-1C11 (p1C11).
  • kits comprising an anti-angiogenic molecule and a chemotherapeutic agent to be administered to a mammal in an amount effective to produce a regression or arrest of an angiogenic dependent condition while minimizing or preventing significant toxicity of the chemotherapeutic agent.
  • a kit optionally comprises an anti-angiogenic molecule and a chemotherapeutic agent in one or more than one containers for administration at about the same time points or at different times.
  • the anti-angiogenic molecule is administered intermittently and the chemotherapeutic agent is administered continuously or in a manner that permits the maintenance of a suitable blood concentration.
  • Such treatment optionally is administered for a prolonged period or chronically, without substantial chemotherapy induced toxicity.
  • Routes of administration include but are not limited to oral and parenteral, including but not limited to intravenous, subcutaneous, percutaneous, intrathecal and intraperitoneal.
  • Patients that may be treated with the methods and compositions of the present invention include any patients with an angiogenic dependent disease.
  • the angiogenic dependent diseases encompassed by the scope of the present invention include, but are not limited to neoplasms, collegen vascular diseases or autoimmune diseases. All neoplasms are suitable for treatment with the present invention, however preferred neoplasms are solid tumors. More preferred are breast carcinoma, lung carcinoma, prostate carcinoma, colon carcinoma, prostate carcinoma, ovarian carcinoma, neuroblastoma, central nervous system tumor, neuroblastoma, glioblastoma multiforme or melanoma, and a preferred mammal to receive treatment is a human.
  • Antibodies used in this invention may be produced in a eukaryotic cell. Techniques for the creation of and production of such antibodies, or portions thereof are well know in the field and are within the knowledge of one of ordinary skill in the art. Techniques used for preparation of monoclonal antibodies, include but are not limited to, the hybridoma technique (Kohler & Milstein, Nature, 256:495-497 (1975)), the trioma technique, the human B-cell hybridoma technique (Kozbor et al., Immunology Today 4:72, (1983)), and the EBV-hybridoma technique to produce human monoclonal antibodies (Cole, et al., 1985, In Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96).
  • DNA encoding chimerized antibodies may be prepared by recombining DNA substantially or exclusively encoding human constant regions and DNA encoding variable regions derived substantially or exclusively from the sequence of the variable region of a mammal other than a human.
  • DNA encoding humanized antibodies may be prepared by recombining DNA encoding constant regions and variable regions, other than the CDRs, derived substantially or exclusively from the corresponding human antibody regions and DNA encoding CDRs derived substantially or exclusively from a mammal other than a human.
  • Each domain of the antibodies of this invention may be a complete immunoglobulin heavy or light chain variable domain, or it may be a functional equivalent or a mutant or derivative of a naturally occurring domain, or a synthetic domain constructed, for example, in vitro using a technique such as one described in WO 93/11236 (Medical Research Council et al./Griffiths et al.). For instance, it is possible to join together domains corresponding to antibody variable domains which are missing at least one amino acid.
  • the important characterizing feature is the ability of each domain to associate with a complementary domain to form an antigen binding site. Accordingly, the terms “variable heavy/light chain fragment” should not be construed to exclude variants which do not have a material effect on how the invention works.
  • DNA deletions and recombinations of the present invention may be carried out by known methods, such as those described in PCT applications WO 93/21319, WO 89/09622, European Patent applications 239,400, 338,745 and 332,424 and/or other standard recombinant DNA techniques.
  • Conventional methods such as those employed in the construction of vectors and plasmids, the insertion of genes encoding polypeptides into such vectors and plasmids, or the introduction of plasmids into host cells, are well known to those of ordinary skill in the art and are described in numerous publications including Sambrook, J., Fritsch, E. F. and Maniatis, T. (1989) Molecular Cloning: A Laboratory Manual, 2nd edition, Cold Spring Harbor Laboratory Press, and in Ausubel et al. (Eds) Current Protocols in Molecular Biology, Green Publishing Associates/ Wiley-Interscience, New York (1990).
  • the invention also includes functional equivalents of the antibodies described in this specification.
  • Functional equivalents have binding characteristics comparable to those of the antibodies, and include, for example, chimerized, humanized and single chain antibodies as well as fragments thereof. Methods of producing such functional equivalents are disclosed in PCT Application No. WO 93/21319, European Patent Application No. EPO 239,400; PCT Application WO 89/09622; European Patent Application No. EP338,745; and European Patent Application EPO 332,424.
  • Functional equivalents include polypeptides with amino acid sequences substantially the same as the amino acid sequence of the variable or hypervariable regions of the antibodies of the invention. “Substantially the same” amino acid sequence is defined herein as a sequence with at least 70% percent homology to an amino acid sequence of an antibody of the invention, as determined by the FASTA search method in accordance with Pearson and Lipman, Proc. Natl. Acad. Sci. USA 85, 2444-2448 (1988).
  • Neuroblastoma cell lines SK-N-MC, SK-N-AS were obtained from American Type Culture Collection (ATCC) and expanded as a monolayer culture by serial passage on tissue culture plates (Nunc, Denmark) in DMEM, 5% fetal bovine serum (Gibco, Grand Island, N.Y., USA).
  • Human umbilical vein endothelial cells (HUVEC) (Clonetics, San Diego, Calif.) were expanded on 1% gelatin-coated tissue culture plates in MCDB131 culture medium (JRH Biosciences, Lenexa, Kans., USA) supplemented with 5 ng/ml bFGF (R&D, Minneapolis, Minn.), 10 units/ml heparin (Wyeth-Ayerst Canada), 10 ng/ml EGF (UBI, Lake Placid, N.Y.) and 10% fetal bovine serum.
  • MCDB131 culture medium JRH Biosciences, Lenexa, Kans., USA
  • the plates were frozen, thawed and the DNA harvested onto a filtermat using a Titertek Cell Harvester. Incorporated radioactivity was measured on Wallac 1205 BetaPlate Scintillation Counter (Wallac Oy, Finland) and proliferation was expressed as a percentage of 3 H-thymidine in treated cells vs. that in controls.
  • SK-N-MC cells were harvested using 1% Trypsin EDTA (GibcoBRL, Gaithesburg, Md.), and single cell suspension of 2 ⁇ 106 cells in 0.2 ml of growth media was injected subcutaneously into the flanks of 4-6 week old CB-17 SCID mice (Charles River, St-Constant, Quebec). Approximately 3 weeks later, most tumors had grown to ⁇ 0.75 cm 3 , and mice were randomized into groups of 5 animals. Two independent experiments were performed, each totaling 20 animals in 4 groups. The treatment was as follows:
  • Group I (Control)—0.4 ml of PBS (DC101 vehicle) i.p. every three days and 0.15 ml injectable saline (vinblastine vehicle) i.p. every three days.
  • Group II 0.4 ml of 2 mg/ml DC101 antibody (800 ⁇ g/mouse) (24) every three days and 0.15 ml of injectable saline i.p. every three days
  • Group III vinblastine sulfate 0.75 mg/m 2 i.p. bolus at the start of therapy, followed by 1 mg/m 2 /day via subcutaneous Alzet osmotic pumps (Alza Corp,Palo Alto, Calif.) for 3 weeks, followed by maintenance therapy with 0.15 ml of 0.067 mg/ml vinblastine sulfate (1.5 mg/m 2 ) i.p. every three days, and 0.4 ml of PBS i.p. every three days
  • Group IV combination of DC101 and vinblastine at doses identical to the single agent groups.
  • Relative tumor vascularity assessed by an FITC-Dextran perfusion assay The method was designed to assess the relative functionality of the tumor vasculature. 2 ⁇ 10 6 SK-N-AS neuroblastoma cells were injected into the flanks of CB-17 SCID mice. Tumors were allowed to grow to approximately 0.75 cm 3 at which point tumor bearing mice were then treated with 1 mg/m 2 vinblastine i.p. every three days, 800 ⁇ g DC101 i.p. every three days, combination of the two agents or saline as a control. At 14 days, when divergence in tumor growth between the treatment groups was clearly evident, 0.2 ml of 25 mg/ml FITC-Dextran in PBS (Sigma, St.
  • the tissue was homogenized, centrifuged at 5000 rpm for 10 minutes, and the supernatant was collected and stored in the dark until further analysis. Blood samples were centrifuged immediately following collection, plasma separated and protected from light at 4° C. until analysis. Fluorescence readings were obtained on a FL600 Fluorescence Plate Reader (Bio-tek Instruments Inc., Winooski, Vt., USA), from a standard curve created by serial dilution of the FITC-dextran used for injection. The ratio of tumor fluorescence: plasma fluorescence was assumed to be reflective of the degree of tumor perfusion.
  • Group I saline i.p.
  • Group II 800 ⁇ g DC101 i.p.
  • Group III 1 mg/m 2 vinblastine i.p.
  • Group IV combination therapy.
  • mice All 25 mice were treated on day 4 and 7 and sacrificed on day 10. Blood samples were collected into heparinized tubes by cardiac puncture, centrifuged immediately following their collection, plasma separated and protected from light at 4° C.
  • the Matrigel plugs were resected from surrounding connective tissues, placed into tubes containing 1 ml of 1:10 dispase and incubated in the dark in a 37° C. shaker overnight. The following day, the plugs were homogenized, centrifuged at 5000 rpm for 10 minutes and supematant saved in the dark for analysis of fluorescence. Fluorescence readings were obtained on FL600 Fluorescence Plate Reader using a standard curve created by serial dilution of FITC-dextran used for injection. Angiogenic response was expressed as a ratio of Matrigel plug fluorescence: plasma fluorescence.
  • the first treatment group treated with DC101, an anti-Flk1 receptor antibody shown previously to inhibit growth of different kinds of human xenografts in mice and in mouse tumor models (5), showed an anticipated effectiveness in inhibiting tumor growth, but, its effect was not sustained.
  • the findings in the second treatment group (vinblastine alone), were even more surprising.
  • This agent traditionally thought to act by inhibiting tumor cell proliferation through inhibition of tubulin assembly, produced significant, albeit not sustained, regression of tumor growth even though used at subclinical low-dose,.
  • This growth delay in the vinblastine group was further potentiated with the simultaneous treatment with the anti-flk-1 antibody, DC101.
  • the combination treatment induced an initial response comparable to the other treatment groups but then caused further, long term, tumor regression.
  • mice in combination therapy group have not manifested any resistance to the treatment or recurrence of disease, despite almost seven months of continuous treatment.
  • the mice remain healthy, with almost no evidence of tumor, except for a small, barely palpable remnant in one of the mice. [What were the doses used?]
  • Toxicity evaluation Anti-vascular therapy would be expected to show minimal toxicity in the post-natal stage of development.
  • DC101/vinblastine combination therapy the health status of the mice was monitored. Weight was plotted at regular intervals and considered a surrogate for evaluation of systemic well being, anorexia, or failure to thrive. No significant differences in weights were seen between the four groups. The weight curve of the DC101 group parallels very closely that of the control group. The vinblastine group showed some weight gain retardation, but the differences never became significantly different from controls. Similarly, the toxicity profile in the combination treatment group was very similar to those in the single agent groups, with the exception of a transient episode of weight loss associated with diarrhea.
  • mice recovered without interruption of treatment were not seen at the doses used in our experiments.
  • Diarrhea a common sign of vinblastine toxicity when doses of 10 mg/m 2 are used, was generally not observed, except for the above mentioned episode.
  • Vinblastine alone or DC101 treatment alone both show an increase in the width of the apoptotic rims, suggesting the cells most distal to the tumor vasculature are primarily affected, but a large percentage of viable tumor cells still survive in the center of the cuff.
  • histology of the combined therapy group shows overwhelming loss of both cell viability and pre-existing tumor architecture.
  • H/E and TUNEL stain There is a close similarity of the appearance of H/E and TUNEL stain.
  • Tumor perfusion by assessment of intravascular fluorescence To further explore the possibility that tumor regression induced with treatment using DC101 and vinblastine was indeed due to the vascular injury, rather than a direct anti-tumor cell effect, we assessed tumor perfusion directly by using a FITC-Dextran fluorescence method. Mice carrying established subcutaneous SK- N-AS human neuroblastoma xenografts ( ⁇ 0.75 cm 3 ) were randomized into four groups and treated systemically with either saline control, DC101, vinblastine or combination therapy for 10 days. FITC-Dextran was injected into the lateral tail vein and equilibrated throughout the vascular compartment.
  • the dose of vinblastine used in our experiments was in the range of 1.5 mg/m 2 , every 3 days, which is approximately 3 times the MTD of this drug in humans, and ⁇ fraction (1/16) ⁇ - ⁇ fraction (1/20) ⁇ of the MTD in mice, given the fact that the MTD of vinblastine in mice is 4-5 times higher than in humans (33,34).
  • Using the Matrigel plug assay we demonstrated that continuous low dose vinblastine administration can cause a direct anti-angiogenic effect in vivo. The combined effect with DC101 was significant.

Abstract

A method of controlling or treating an angiogenic dependent condition in a mammal, preferably in a human by administering an anti-angiogenic molecule such as an angiogenesis growth factor antagonist, and a chemotherapeutic agent in amounts and frequencies effective, in combination, to produce a regression or arrest of said condition while minimizing or preventing significant toxicity of the chemotherapeutic agent. Also a kit for controlling or treating an angiogenic dependent condition in a mammal, preferably in a human, comprising an anti-angiogenic molecule, such as an angiogenesis growth factor antagonist, and a chemotherapeutic agent in amounts effective, in combination, to produce a regression or arrest of said condition while minimizing or preventing significant toxicity of the chemotherapeutic agent.

Description

  • The present application claims the benefit of priority from U.S. Provisional Application No. 60/178791, filed on Jan. 28, 2000, which is hereby incorporated in its entirety by reference.[0001]
  • FIELD OF THE INVENTION
  • The present invention relates to the inhibition or prevention of angiogenesis as a means to control or treat an angiogenic dependent condition, a condition characterized by, or dependent upon, blood vessel proliferation. The invention further relates to the use of an anti-angiogenic molecule in combination with a chemotherapeutic agent. [0002]
  • BACKGROUND OF THE INVENTION
  • Angiogenesis is a highly complex process of developing new blood vessels that involves the proliferation and migration of, and tissue infiltration by capillary endothelial cells from pre-existing blood vessels, cell assembly into tubular structures, joining of newly forming tubular assemblies to closed-circuit vascular systems, and maturation of newly formed capillary vessels. The molecular bases of many of these aspects are still not understood. [0003]
  • Angiogenesis is important in normal physiological processes including embryonic development, follicular growth, and wound healing, as well as in pathological conditions such as tumor growth and in non-neoplastic diseases involving abnormal neovascularization, including neovascular glaucoma (Folkman, J. and Klagsbrun, M. Science 235:442-447 (1987). Other disease states include but are not limited to, neoplastic diseases, including but not limited to solid tumors, autoimmune diseases and collagen vascular diseases such as, for example, rheumatoid arthritis, and ophthalmalogical conditions such as diabetic retinopathy, retrolental fibroplasia and neovascular glaucoma Conditions or diseases to which persistent or uncontrolled angiogenesis contribute have been termed angiogenic dependent or angiogenic associated diseases. [0004]
  • One means of controlling such diseases and pathological conditions comprises restricting the blood supply to those cells involved in mediating or causing the disease or condition. For example, in the case of neoplastic disease, solid tumors develop to a size of about a few millimeters, and further growth is not possible, absent angiogenesis within the tumor. In the past, strategies to limit the blood supply to tumors have included occluding blood vessels supplying portions of organs in which tumors are present. Such approaches require the site of the tumor to be identified and are generally limited to treatment to a single site, or small number of sites. An additional disadvantage of direct mechanical restriction of a blood supply is that collateral blood vessels develop, often quite rapidly, restoring the blood supply to the tumor. [0005]
  • Other approaches have focused on the modulation of factors that are involved in the regulation of angiogenesis. While usually quiescent, vascular endothelial proliferation is highly regulated, even during angiogenesis. Examples of factors that have been implicated as possible regulators of angiogenesis in vivo include, but are not limited to, transforming growth factor beta (TGFβ), acidic and basic fibroblast growth factor (aFGF and bFGF), platelet derived growth factor (PDGF), and vascular endothelial growth factor (VEGF) (Klagsbrun, M. and D'Amore, P. (1 991) Annual Rev. Physiol. 53: 217-239). [0006]
  • One growth factor of particular interest is VEGF. An endothelial-cell specific mitogen, VEGF acts as an angiogenesis inducer by specifically promoting the proliferation of endothelial cells. It is a homodimeric glycoprotein consisting of two 23 kD subunits with structural similarity to PDGF. Four different monomeric isoforms of VEGF resulting from alternative splicing of mRNA have been identified. These include two membrane bound forms (VEGF[0007] 206 and VEGF189) and two soluble forms (VEGF165 and VEGF121). VEGF165 is the most abundant isoform in all human tissues except placenta.
  • VEGF is expressed in embryonic tissues (Breier et al., Development (Camb.) 114:521 (1992)), macrophages, and proliferating epidermal keratinocytes during wound healing (Brown et al., J. Exp. Med., 176:1375 (1992)), and maybe responsible for tissue edema associated with inflammation (Fenrara et al., Endocr. Rev. 13:18 (1992)). In situ hybridization studies have demonstrated high levels of VEGF expression in a number of human tumor lines including glioblastoma multiforme, hemangioblastoma, other central nervous system neoplasms and AIDS-associated Kaposi's sarcoma (Plate, K. et al. (1992) Nature 359: 845-848; Plate, K. et al. (1993) Cancer Res. 53: 5822-5827; Berkman, R. et al. (1993) J. Clin. Invest. 91: 153-159; Nakamura, S. et al. (1992) AIDS Weekly, 13 (1)). High levels of VEGF also have been reported in hypoxia induced angiogenesis (Shweiki, D. et al. (1992) Nature 359: 843-845). [0008]
  • VEGF mediates its biological effect through high affinity VEGF receptors which are selectively expressed on endothelial cells during, for example, embryogenesis (Millauer, B., et al. (1993) Cell 72: 835-846) and tumor formation. VEGF receptors typically are class III receptor-type tyrosine kinases characterized by having several, typically 5 or 7, immunoglobulin-like loops in their amino-terminal extracellular receptor ligand-binding domains (Kaipainen et al., J. Exp. Med. 178:2077-2088 (1993)). The other two regions include a transmembrane region and a carboxy-terminal intracellular catalytic domain interrupted by an insertion of hydrophilic interkinase sequences of variable lengths, called the kinase insert domain (Terman et al., Oncogene 6:1677-1683 (1991)). VEGF receptors include flt-1, sequenced by Shibuya M. et al., Oncogene 5, 519-524 (1990); flk-1, sequenced by Matthews W. et al. Proc. Natl. Acad. Sci. USA, 88:9026-9030 (1991) and KDR, the human homologue of flk-1, described in PCT/US92/01300, filed Feb. 20, 1992, and in Terman et al., Oncogene 6:1677-1683 (1991). [0009]
  • High levels of flk-1 are expressed by endothelial cells that infiltrate gliomas (Plate, K. et al., (1992) Nature 359: 845-848), and are specifically upregulated by VEGF produced by human glioblastomas (Plate, K et al. (1993) Cancer Res. 53: 5822-5827). The finding of high levels of flk-1 expression in glioblastoma associated endothelial cells (GAEC) suggests that receptor activity is induced during tumor formation, since flk-1 transcripts are barely detectable in normal brain endothelial cells. This upregulation is confined to the vascular endothelial cells in close proximity to the tumor. Blocking VEGF activity with neutralizing anti-VEGF monoclonal antibodies (mAbs) results in inhibition of the growth of human tumor xenografts in nude mice (Kim, K. et al. (1993) Nature 362: 841-844), suggesting a direct role for VEGF in tumor-related angiogenesis. [0010]
  • Various chemotherapeutic drugs also have been shown to block functions of activated, dividing endothelial cells critical to angiogenesis, or to kill such cells. Such collateral damaging effects on a genetically stable normal host cell, in addition to the chemotherapeutic agent's effect upon the tumor cells, contribute significantly to the in vivo anti-tumor efficacy of chemotherapy. However, the standard use of chemotherapeutic agents has obvious undesirable side-effects upon the normal cells of patients, limiting its use. Administration of chemotherapeutic agents in their usual doses and at usual dosage frequencies are commonly associated with side-effects, including, but not limited to, myelosuppression, neurotoxicity, cardiotoxicity, alopecia, nausea and vomiting, nephrotoxicity, and gastrointestinal toxicity. Further, patients' tumors often also develop resistance to the chemotherapeutic agents after initial exposure to the drugs. [0011]
  • A desirable method and composition for controlling angiogenesis should be well tolerated, have few or no side-effects, and prevent new vessel formation at sites of disease without interfering with required physiologic angiogenesis in normal sites. It should be effective and, in the case of neoplastic disease, overcome the problem of the development of drug resistance by tumor cells. In so doing, it should permit targeted therapy without the accurate identification of all disease sites. The present invention addresses many of the problems with existing materials and methods. [0012]
  • SUMMARY OF THE INVENTION
  • The present invention provides a method of treating an angiogenic dependent condition in a mammal comprising administering an anti-angiogenic molecule and a chemotherapeutic agent to the mammal, in an amount and frequency effective, in combination, to produce a regression or arrest of the condition without significant toxicity from the chemotherapeutic agent. The angiogenic dependent condition may be selected from the group consisting of neoplasm, collagen-vascular disease or auto-immune disease, including a solid tumor neoplasm, including breast carcinoma, lung carcinoma, prostate carcinoma, colon carcinoma, prostate carcinoma, ovarian carcinoma, neuroblastoma, central nervous system tumor, neuroblastoma, glioblastoma multiforme or melanoma. The mammal receiving the treatment is preferably a human. [0013]
  • The anti-angiogenic molecules inhibit the action of a vascular endothelium survival factor, which include receptors and their ligands. Vascular endothelium survival factors include receptors, including angiogenic growth factors such as VEGF receptor, including flk-1/KDR receptor, or flt-4 receptor and VEGF. Examples of other vascular endothelial survival factors are integrin α[0014] vβ3. α 3 ligand, Tie2/tek ligand, Tie2/tek, endoglin ligand, endoglin, neuropilin ligand, neuropilin, thrombospondin ligand, thrombospondin, PDGFα, PDGFα receptor, PDGFβ, PDGFβ receptor, aFGF, aFGF receptor, bFGF, bFGF receptor, TGFβ, TGFβ receptor, EGF, EGF receptor, angiostatin, angiostatin receptor, angiopoetin, angiopoeitin receptor, PLGF, PLGF receptor, VPF, or VPF receptor. Optionally, the ligand is selected from the group consisting of VEGF (VEGF-A), VEGF-B, VEGF-C, or VEGF-D. The anti-angiogenic molecule may be selected from the group consisting of antibody, antibody fragment, small molecule or peptide.
  • Preferred embodiments of the present invention include antibodies selected from the group consisting of mouse antibody, rat antibody, chimeric antibody, humanized antibody or human antibody. A preferred antibody is IMC-1C11. [0015]
  • Preferably, IMC-1C11 is administered in a dose of from about 5 mg/m[0016] 2 to about 700 mg/m2 about daily to about every 7 days, more preferably a dose of from about 7.5 mg/m2 to about 225 mg/m2, about twice per week. Optionally, the IMC-1C11 is administered at a dose and frequency sufficient to substantially saturate the VEGF receptor. Optionally, the anti-angiogenic molecule is administered in a dose and frequency sufficient to substantially saturate the target of the anti-angiogenic molecule. In another embodiment, the anti-angiogenic molecule is administered in a dose equivalent to that of IMC-1C11, administered in a dose of from about 5 mg/m2 to about 700 mg/m2 about daily to about every 7 days, more preferably a dose of from about 7.5 mg/m2 to about 225 mg/m2, about twice per week.
  • The chemotherapeutic agent may be selected from the group consisting of vinca alkaloid, camptothecan, taxane, or platinum analogue, including vincristine, vinblastine, vinorelbine, vindesine, paclitaxel, docetaxel, 5 FU, cisplatin, carboplatin, iranotecan, topotecan or cyclophosphamide. The chemotherapeutic agent is administered in a low-dose regimen. Preferably the chemotherapeutic agent is administered at less than about 50% of the maximum tolerated dose, more preferably at less than about 20% of the maximum tolerated dose, most preferably at less than about 10% of the maximum tolerated dose. In one preferred embodiment the vinblastine is administered in a dose from about 0.5 mg/m[0017] 2 to about 3 mg/m2 from about once every 3 days to about once every 7 days. In another embodiment, the chemotherapeutic agent is administered in a dosage and frequency that is of substantially equivalent efficacy to vinblastine in a dose from about 0.5 mg/m2 to about 3 mg/m2 from about once every 3 days to about once every 7 days.
  • Optionally the chemotherapeutic agent is administered more frequently than about every three weeks, or more frequently than about every seven days. [0018]
  • The present invention also includes a kit for treating an angiogenic dependent condition in a mammal comprising the anti-angiogenic molecule and the chemotherapeutic agent, which are provided to be administered in an amount and frequency effective, in combination, to produce a regression or arrest of the condition while minimizing or preventing significant toxicity of the chemotherapeutic agent.[0019]
  • BRIEF DESCRIPTION OF FIGURES
  • FIG. 1 is the encoding nucleotide sequence and deduced amino acid sequence of V[0020] H and VL domains of IMC-1C11 (c-p1C11).
  • DETAILED DESCRIPTION OF THE INVENTION
  • Throughout this application, various articles and patents are referenced. Disclosures of these publications in their entireties are hereby incorporated by reference into this application. [0021]
  • The present invention comprises a method of treating or controlling an angiogenic dependent condition in a mammal, comprising administering an anti-angiogenic molecule and a chemotherapeutic agent in combined amounts effective to produce a regression or arrest to the angiogenic dependent condition, while minimizing or preventing significant toxicity. [0022]
  • The benefits of the combination of an anti-angiogenic molecule and a chemotherapeutic agent of the present invention include an improvement in the treatment and control of an angiogenic dependant condition with reduced doses of a chemotherapeutic agent administered at increased frequency, without significant toxicity, together with a anti-angiogenic molecule. The combination can be administered for a prolonged period of time, or optionally a shorter duration of treatment may be administered due to the increased effectiveness of the combination. Toxicity is reduced or eliminated without a loss of effectiveness. The administration of the treatment of the invention can overcome the problems of drug resistance that develops with standard chemotherapeutic regimens. [0023]
  • The anti-angiogenic molecule functions to inhibit or prevent angiogenesis and thereby treating the angiogenic dependent condition by inhibiting, blocking, or antagonizing the effect of vascular endothelial survival factors. These survival factors are receptors and their ligands, upon which vascular endothelium depends, either directly or indirectly, for growth and/or survival. They play a role in allowing vascular endothelial cells to recovery from injury or insult, which, absent the effect of the survival factor would result in cell death or apoptosis. Survival factors include vascular endothelial cell growth factors or mitogens, as well as those factors which do not appear to have a direct growth-stimulatory effect but allow the cells to recover from injury. [0024]
  • Examples of survival factors include VEGF receptors, including but not limited to flt-1 (VEGFR1), flk-1/KDR (VEGFR2), flt-4 (VEGFR3), their ligands VEGF, VEGF-B, VEGF-C, and VEGF-D, integrin αVβ3, Tie2/tek, endoglin (CD105), neuropilin, thrombospondin and their ligands, and PDGFα, PDGFβ, aFGF, bFGF, and TGFβ, as well as EGF, angiostatin, and angiopoeitin, vascular permiability factor (VPF), and placenta-like growth factor (PLGF) and their receptors. [0025]
  • The survival factors that are receptors are located on vascular endothelial cells or may be located on other cell types including, but not limited to, tumor cells. The anti-angiogenic molecule inhibit binding to ,and/or activation of, receptors, inhibit their expression, or inhibit the binding of or expression of ligands. Suitable anti-angiogenic molecules include, but are not limited to antibody, antibody fragment, small molecule or peptide. An antibody can be derived from any mammalian species. Optionally, the antibody is of mouse, rat, rabbit, or human origin. Preferably the antibody is chimeric, more preferably the antibody is humanized, and even more preferably the antibody is human. Suitable antibody fragments include, for example, Fab fragment, Fab′ fragment, F(ab′)[0026] 2 fragment, monovalent single chain antibody (scFv), and diabodies (DAB).
  • Examples of suitable anti-angiogenic molecules that are antagonists to vascular endothelium survival factors include, but are not limited to, VEGF receptor antagonist or VEGF antagonist, as disclosed in U.S. Pat. Nos. 5,840,301, 5,861,499, 5,874,542, 5,955,311, and 5,730,977, which are incorporated in their entirety by reference, aFGF receptor antagonist, aFGF antagonist, bFGF receptor antagonist, bFGF antagonist, PDGF receptor antagonist, PDGF antagonist, TGFβ antagonist, Tie2/tek antagonist (P. Lin et al., Inhibition of Tumor Angiogenesis Using a Soluble Receptor Establishes a Role for Tie2 in Pathologic Vascular Growth. J. Clin. Invest. 100(8) 2072 (1997)), endoglin (CD105) antagonist, as disclosed in U.S. Pat. Nos. 5,855,866, and 5,660,827, neuropilin antagonist, thrombospondin antagonist, and antagonists to the receptors for PDGFα, PDGFβ, aFGF, bFGF, or TGFβ, as well as antagonists to the receptors for EGF, angiostatin, angiopoeitin, or VPF (Vascular Permeability Factor) as disclosed in U.S. Pat. Nos. 5,036,003 and 5,659,013. Also encompassed within the scope of the present invention are integrin receptor antagonists as disclosed in U.S. Pat. Nos. 6,017,926, 6,017,925, 5,981,546, 5,952,341, and 5,919,792, integrin αVβ[0027] 3 antagonists, as disclosed in U.S. Pat. Nos. 5,780,426, 5,773,412, 5,767,071, 5,759,996, 5,753,230, 5,652,110, and 5,652,109, antagonists to placenta-like growth factor (PLGF) as disclosed in European Patent Application EP506477A1, thrombospondin antagonists as disclosed in U.S. Pat. Nos. 5,840,692, 5,770,563, 5,654,277, 5,648,461, 5,506,208, 5,399,667, 5,200,397, 5,192,744, and 5,190,918, as well as those disclosed in U.S. Pat. Nos. 5,965,132, 6,004,555 and 5,877,289, and PCT Applications Nos. WO 99/16465, WO 97/05250, WO 98/33917. Also included, are molecules such as thalidomide, TNP-470, interferon-a (INF-α), and interleukin-12 (IL-12).
  • In many cases, the expression of a receptor and/or ligand is upregulated in an region of angiogenesis. However, although located in an area of abnormal cells responsible for the specific disease, exposed to high levels of ligand, and having upregulated receptors, the cells of the vascular endothelium are largely normal and responsive to normal regulatory mechanisms. An advantage to blocking a receptor, rather than the ligand, is that are that fewer anti-angiogenic molecules may be needed to achieve such inhibition, as levels of receptor expression may be more constant than those of the environmentally induced ligand. Because the receptors exist on essentially normal endothelial cells, their behavior is less likely to escape normal regulatory control. Although there are advantages to targeting receptors, it is also possible, and within the scope of the present invention, to inhibit angiogenesis by targeting the ligand for the receptor, either alone or in combination with blockade of the receptor. Optionally, antagonism of the receptor is combined with antagonism of the ligand in order to achieve even more efficient inhibition of angiogenesis. [0028]
  • In a preferred embodiment of the invention, the anti-angiogenic molecule is an antagonist to VEGF or the VEGF receptor. The expression of the VEGF receptor and ligand is low in normal endothelial cells that are not in or near a region of angiogenesis. In contrast, VEGF receptors present on tumor infiltrating vascular endothelial cells are upregulated, as is the expression of the VEGF ligand by tumor cells. VEGF (or VEGF-A) is the ligand for VEGFR1 and VEGFR2, VEGF-B is the ligand for VEGFR2, VEGF-C is the ligand for VEGFR3, VEGFR4, and possibly VEGFR2, and VEGF-D is the ligand for VEGFR2 and VEGFR3. Blocking the interaction between VEGF and its receptors can inhibit angiogenesis, and thereby tumor growth, while not significantly effecting normal endothelial cells at other sites, where vascular endothelial cell receptors have not been upregulated. In one embodiment of the present invention, antagonism of the VEGF receptor is combined with antagonism of the VEGF ligand in order to achieve even more efficient inhibition of angiogenesis. In other embodiments of the invention, VEGF (VEGF-A), VEGF-B, VEGF-C or VEGF-D is the ligand which is inhibited by the anti-angiogenesis molecule. Optionally, the effect of more than one form of VEGF is inhibited. [0029]
  • Examples of an antagonist to a receptor are the antibodies DC101, described in the Examples, an antagonist of flk-1, and A.4.6.1 and its chimeric and humanized form as disclosed in L. G. Presta, Humanization of an Anti-vascular Endothelial Growth Factor Monoclonal Antibody for the Therapy of Solid Tumors and Other Disorders. Cancer Research, 57, 4593-4599 (1997), which is hereby incorporated by reference. A preferred antibody is the mouse-human chimeric antibody IMC-1C11 which is a KDR antagonist, and is disclosed in U.S. application Ser. No. 09/240,736, which is hereby incorporated by reference. The encoding nucleotide sequences and deduced amino acid sequences of the V[0030] H and VL domains are shown in FIG. 1.
  • The present invention provides a low dose application of a chemotherapeutic agent that provides effective therapy without significant side-effects. An effective amount of a chemotherapeutic agent is an amount sufficient to inhibit or contribute to the inhibition of angiogenesis, when administered with an anti-angiogenic molecule, without resulting in significant toxicity. The meaning of significant toxicity is well known to one of ordinary skill in the art, and includes toxicities that cumulatively or acutely effect a patient's quality of life and/or limit the amount of chemotherapeutic agent than can be administered. Examples of chemotherapy induced toxicity that can be minimized or prevented by the present invention include, but are not limited to, myelosuppression, neurotoxicity, cardiotoxicity, alopecia, nausea and vomiting, nephrotoxicity, and gastrointestinal toxicity. The low dose administration of a chemotherapeutic agent without significant toxicity permits prolonged treatment if desired. Additionally, the low dose manner of chemotherapy administration in the present invention can overcome the problem of the development of chemotherapeutic drug resistance by the patient's tumor cells that occurs with current chemotherapeutic regimens. The present invention delays, reduces, or even circumvents the problem of acquired drug resistance by targeting the genetically stable endothelial cells of newly formed tumor blood vessels, rather than genetically unstable tumor cells which are prone to mutate and develop resistance. Encompassed within the scope of the present invention is the administration of amounts of chemotherapy that are insufficient to have a cytotoxic effect on tumor cells yet have anti-angiogenic properties as a result of the drug's effect on vascular endothelial cells. [0031]
  • The chemotherapeutic agent of the present invention functions, in combination with the anti-angiogenic molecule, to cause a cytotoxic effect on the vascular endothelial cells involved in angiogenesis. A number of chemotherapeutic agents have been identified as having anti-angiogenic activity and are suitable for use in the practice of the present invention. Examples include, but are not limited to, taxanes, including but not limited to paclitaxel and docetaxel, camptothecin analogues, including but not limited to iranotecan and topotecan, platinum analogues including but not limited to cisplatin and carboplatin, 5FU, and vinca alkaloid, including but not limited to vinblastine, vincristine vindesine and vinorelbine. [0032]
  • The low-dose administration of chemotherapeutic agents, to achieve therapeutic effects without significant toxicity (side effects) is routinely possible by the practice of the present invention. Applying standard methods of defining optimal dosage levels and schedules to the teachings of the present invention, one of ordinary skill in the art readily can determine a more or most desirable low-dose regimen for a selected chemotherapeutic agent when used in combination with an anti-angiogenic molecule, as detailed in the present application. A low-dose regimen will administer the chemotherapy at frequent intervals or continually, at less than about 50% [Should this be lower?] of the maximum tolerated dose (MTD), more preferably less than about 20% of the MTD, and most preferably, less than about 10% of the MTD, although the preferred dose depends on the particular chemotherapy. The preferred dose will be a dose effective to inhibit or prevent progression of the angiogenic dependent condition, when administered in combination with the anti-angiogenic molecule of the present invention, while minimizing or preventing the development of significant chemotherapy related toxicity. Optionally the dose of chemotherapy will be effective to inhibit or prevent progression of the angiogenic dependent condition even when administered alone. Optionally the dose of chemotherapy will be one which does not exert a direct cytotoxic effect on tumor cells, yet has an antitumor effect mediated by its anti-angiogenic properties. Optionally, the low-dose regimen of the present invention will administer chemotherapy at a dose intensity of less than 50% of that when administered alone to treat a particular neoplasm. More preferably the low-dose regimen of the present invention will administer chemotherapy at a dose intensity of less than 20% of that when administered alone to treat a particular neoplasm. [0033]
  • In the prior art, chemotherapy is usually given intermittently, commonly in the form of a bolus infusion or an infusion lasting from about 20 minutes to about three hours, at about the maximum tolerated dose (MTD) with long rest periods (e.g., 3 weeks) between successive drug exposures. It has been suggested that these rest periods provide the endothelial cell compartment of a tumor an opportunity to repair some of the damage inflicted by the chemotherapy (T. Browder, et al., Antiangiogenic Scheduling of Chemotherapy Improves Efficacy Against Experimental Drug-Resistant Cancer. [0034] Cancer Res. (In press) 1999). Administering lower doses of a chemotherapeutic drug, such as cyclophosphamide, more frequently such as weekly, enables circumvention of many problems associated with standard chemotherapeutic doses. This anti-angiogenic scheduling of chemotherapy optimizes antitumor/anti-vascular effects. For example, a sub-line of the Lewis Lung Carcinoma, previously selected in vivo for acquired resistance to the MTD of cyclophosphamide, is rendered sensitive again to the drug in vivo by employing continuous low dose therapy of the same drug.
  • The invention provides low-dose administration of chemotherapy administered at short intervals, for example, from about every 4 to about every 6 hours, to about daily to weekly, or optionally administered continuously. The preferred time interval between administration of successive doses of chemotherapeutic agent is that amount of time that is of sufficiently short duration that the blood levels of the chemotherapeutic agent (or its active metabolite) will remain at about a concentration sufficient to exert an anti-angiogenic effect for substantially the duration of treatment. Preferably, such a blood level will be maintained for at least about 30% [Should this be lower] of the time between doses, more preferably for at least 50% of the time between doses, most preferably for at least about 70% of the time between doses. Therapy is continued for a period of time from about 10 days to about 6 months, or as determined by one of skill in the art. Optionally, treatment will continue chronically for a period longer than six months for as long as is needed. The present invention reduces host toxicity, allows for longer term administration of the chemotherapeutic agent in diseases or pathological conditions requiring it, and does not sacrifice, and perhaps even improves, antitumor efficacy. Optionally, increased efficacy will permit the use of shorter durations of therapy for selected angiogenic dependent conditions. [0035]
  • The anti-angiogenic molecule is administered in dosages and dose frequencies sufficient to substantially saturate the selected target receptor or ligand. Substantial saturation is saturation of at least about 50% of targeted receptors. A more preferred level of saturation is at least about 80%, and a most preferred level of saturation is about 100%. Optionally, the anti-angiogenic molecule is administered at a dose and frequency sufficiently short to maintain a blood level sufficient to saturate the targeted survival factor for at least about 50% of the time between doses, more preferably at least about 70% of the time and most preferably at least about 90% of the time interval between doses. Using the concentrations required to achieve receptor saturation or ligand neutralization in vitro, and by analysis of serum concentrations of anti-angiogenic molecule in vivo, both the appropriate dose and schedule can be determined readily by one of skill in the art. [0036]
  • A preferred embodiment of the invention is the combination of a chemotherapeutic agent and a VEGF receptor antagonist. It has been shown that a major function of VEGF is to promote the survival of endothelial cells comprising newly formed vessels (L. E. Benjamin, et Al., Selective Ablation of Immature Blood Vessels in Established Human Tumors Follows Vascular Endothelial Growth Factor Withdrawal. [0037] J.Clin.Invest. 103:159-165 (1999), T. Alon, et al., Vascular Endothelial Growth Factor Acts as a Survival Factor for Newly Formed Retinal Vessels and Has Implications for Retinopathy of Prematurity. Nature Med. 1:1024-1028 (1995), R. K. Jain, et al., Endothelial Cell Death, Angiogenesis, and Microvascular Function after Castration in an Androgen-Dependent Tumor: Role of Vascular Endothelial Growth Factor. Proc.Natl.Acad.Sci. U.S.A. 95:10820-10825 (1998)) Hence, the ability of such cells to cope with the damage inflicted by continuous or frequent exposure to a chemotherapeutic drug is selectively and significantly impaired, (M. J. Prewett, et al., Antivascular Endothelial Growth Factor Receptor (Fetal Liver Kinase 1) Monoclonal Antibody Inhibits Tumor Angiogenesis and Growth of Several Mouse and Human Tumors. Cancer Res 59:5209-5218. (1999); T. A. Fong, et al., SU5416 Is a Potent and Selective Inhibitor of the Vascular Endothelial Growth Factor Receptor (Flk-1/kdr) That Inhibits Tyrosine Kinase Catalysis, Tumor Vascularization, and Growth of Multiple Tumor Types. Cancer Res 59:99-106 (1999); N. Ferrara, et al., Clinical Applications of Angiogenic Growth Factors and Their Inhibitors. Nat.Med. 5:1359-1364. (1999)). It is believed that the combination of continuous chemotherapy with, for example, interruption of the cell rescue mechanisms provided by activation of the VEGF receptor plays a role in inducing vascular endothelial cell apoptosis.
  • A preferred embodiment of the present invention comprises the administration of the antibody IMC-1C11, a KDR receptor antagonist with a chemotherapeutic agent. A preferred dose of IMC-1C11, is an amount that is sufficient to adequately saturate the targeted receptors or ligand. In in vitro experiments, 50% saturation of VEGF receptors was obtained as an IMC-1C11 concentration of 0.2 μg/ml, and 100% at a concentration of 3 μg/ml. A preferred level of saturation is about at least 50%, a more preferred level is about at least 80%, and a most preferred level is about 100% saturation. For therapy, a preferred dose regimen of IMC-1C11 is from about 5 mg/m[0038] 2 to about 700 mg/M2, more preferably from about 7.5 mg/M2 to about 225 mg/m2, administered about twice per week.
  • Another preferred embodiment of the invention combines IMC-1C11 in the doses described above with, vinblastine, administered in a low dose regimen, at a dose from about 0.5 mg /m[0039] 2 to about 3 mg/m2 from about every 3 days to about every 7 days. Optionally, a suitable chemotherapeutic agent other than vinblastine is administered in a dosage and frequency that is of substantially equivalent efficacy to vinblastine (in the combination) at a dose from about 0.5 mg /m2 to about 3 mg/m2 from about every 3 days to about every 7 days.
  • In other embodiments of the present invention, doses of an anti-angiogenic molecule in amounts and dosing frequencies sufficient to provide levels of receptor or ligand saturation equivalent to that of IMC-1C11 in the doses about are combined with a chemotherapeutic agent in a dose and frequency equivalent to that of vinblastine above, and therapy is carried out for as long as is needed. An equivalent dose is one that, in the combination, is substantially as effective in arresting or inhibiting the angiogenic dependent condition, while being substantially as effective in minimizing or preventing significant chemotherapy induced toxicity. In one preferred embodiment of the present invention, an equivalent dose of another chemotherapeutic agent is determined based on data derived from an animal model, an example of which is included herein, utilizing a chemotherapy-resistant cell line so that any observed antitumor effect is due to an effect on the vascular endothelium. A preferred dose of vinblastine in a mouse is from about 1 mg/m[0040] 2 to about 2 mg/m2 more preferably about 1.5 mg/m2 administered every three days. The MTD of this drug in mice is approximately 4-5 times that of a human, and a preferred dose is {fraction (1/16)}-{fraction (1/20)} of the MTD in mice. A preferred dose of DC011 in a mouse is about 800 μg administered intraperitoneally every three days. The use of DC101 and vinblastine showed a therapeutic effect upon neuroblastoma cell lines grown as xenografts in SCID mice. (L. Witte, L, et al., Monoclonal antibodies targeting the VEGF receptor-2 (flk1/KDR) as an anti-angiogenic therapeutic strategy. Cancer Metastasis Rev. 17:155-161. (1998); Prewitt, 1999). In yet another preferred embodiment of the present invention, low-dose vinblastine is administered every 3 days in combination with IMC-1C11 (p1C11).
  • In one aspect of the present invention, there is provided a kit comprising an anti-angiogenic molecule and a chemotherapeutic agent to be administered to a mammal in an amount effective to produce a regression or arrest of an angiogenic dependent condition while minimizing or preventing significant toxicity of the chemotherapeutic agent. Such a kit optionally comprises an anti-angiogenic molecule and a chemotherapeutic agent in one or more than one containers for administration at about the same time points or at different times. Optionally, the anti-angiogenic molecule is administered intermittently and the chemotherapeutic agent is administered continuously or in a manner that permits the maintenance of a suitable blood concentration. It is an aspect of the present invention that such treatment optionally is administered for a prolonged period or chronically, without substantial chemotherapy induced toxicity. Routes of administration include but are not limited to oral and parenteral, including but not limited to intravenous, subcutaneous, percutaneous, intrathecal and intraperitoneal. Patients that may be treated with the methods and compositions of the present invention include any patients with an angiogenic dependent disease. [0041]
  • The angiogenic dependent diseases encompassed by the scope of the present invention include, but are not limited to neoplasms, collegen vascular diseases or autoimmune diseases. All neoplasms are suitable for treatment with the present invention, however preferred neoplasms are solid tumors. More preferred are breast carcinoma, lung carcinoma, prostate carcinoma, colon carcinoma, prostate carcinoma, ovarian carcinoma, neuroblastoma, central nervous system tumor, neuroblastoma, glioblastoma multiforme or melanoma, and a preferred mammal to receive treatment is a human. [0042]
  • Antibodies used in this invention may be produced in a eukaryotic cell. Techniques for the creation of and production of such antibodies, or portions thereof are well know in the field and are within the knowledge of one of ordinary skill in the art. Techniques used for preparation of monoclonal antibodies, include but are not limited to, the hybridoma technique (Kohler & Milstein, Nature, 256:495-497 (1975)), the trioma technique, the human B-cell hybridoma technique (Kozbor et al., Immunology Today 4:72, (1983)), and the EBV-hybridoma technique to produce human monoclonal antibodies (Cole, et al., 1985, In Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96). [0043]
  • DNA encoding chimerized antibodies may be prepared by recombining DNA substantially or exclusively encoding human constant regions and DNA encoding variable regions derived substantially or exclusively from the sequence of the variable region of a mammal other than a human. DNA encoding humanized antibodies may be prepared by recombining DNA encoding constant regions and variable regions, other than the CDRs, derived substantially or exclusively from the corresponding human antibody regions and DNA encoding CDRs derived substantially or exclusively from a mammal other than a human. [0044]
  • Each domain of the antibodies of this invention may be a complete immunoglobulin heavy or light chain variable domain, or it may be a functional equivalent or a mutant or derivative of a naturally occurring domain, or a synthetic domain constructed, for example, in vitro using a technique such as one described in WO 93/11236 (Medical Research Council et al./Griffiths et al.). For instance, it is possible to join together domains corresponding to antibody variable domains which are missing at least one amino acid. The important characterizing feature is the ability of each domain to associate with a complementary domain to form an antigen binding site. Accordingly, the terms “variable heavy/light chain fragment” should not be construed to exclude variants which do not have a material effect on how the invention works. The DNA deletions and recombinations of the present invention may be carried out by known methods, such as those described in PCT applications WO 93/21319, WO 89/09622, European Patent applications 239,400, 338,745 and 332,424 and/or other standard recombinant DNA techniques. Conventional methods, such as those employed in the construction of vectors and plasmids, the insertion of genes encoding polypeptides into such vectors and plasmids, or the introduction of plasmids into host cells, are well known to those of ordinary skill in the art and are described in numerous publications including Sambrook, J., Fritsch, E. F. and Maniatis, T. (1989) Molecular Cloning: A Laboratory Manual, 2nd edition, Cold Spring Harbor Laboratory Press, and in Ausubel et al. (Eds) Current Protocols in Molecular Biology, Green Publishing Associates/ Wiley-Interscience, New York (1990). [0045]
  • The invention also includes functional equivalents of the antibodies described in this specification. Functional equivalents have binding characteristics comparable to those of the antibodies, and include, for example, chimerized, humanized and single chain antibodies as well as fragments thereof. Methods of producing such functional equivalents are disclosed in PCT Application No. WO 93/21319, European Patent Application No. EPO 239,400; PCT Application WO 89/09622; European Patent Application No. EP338,745; and European Patent Application EPO 332,424. [0046]
  • Functional equivalents include polypeptides with amino acid sequences substantially the same as the amino acid sequence of the variable or hypervariable regions of the antibodies of the invention. “Substantially the same” amino acid sequence is defined herein as a sequence with at least 70% percent homology to an amino acid sequence of an antibody of the invention, as determined by the FASTA search method in accordance with Pearson and Lipman, [0047] Proc. Natl. Acad. Sci. USA 85, 2444-2448 (1988).
  • The examples which follow are set forth to aid in understanding the invention, but are not intended to, and should not be construed as, limiting the scope of the invention in any manner. [0048]
  • EXAMPLES
  • Cells and culture conditions: Neuroblastoma cell lines SK-N-MC, SK-N-AS were obtained from American Type Culture Collection (ATCC) and expanded as a monolayer culture by serial passage on tissue culture plates (Nunc, Denmark) in DMEM, 5% fetal bovine serum (Gibco, Grand Island, N.Y., USA). Human umbilical vein endothelial cells (HUVEC) (Clonetics, San Diego, Calif.) were expanded on 1% gelatin-coated tissue culture plates in MCDB131 culture medium (JRH Biosciences, Lenexa, Kans., USA) supplemented with 5 ng/ml bFGF (R&D, Minneapolis, Minn.), 10 units/ml heparin (Wyeth-Ayerst Canada), 10 ng/ml EGF (UBI, Lake Placid, N.Y.) and 10% fetal bovine serum. [0049]
  • In vitro determination of drug sensitivity: Three thousand cells in 200 μl growth media per well were plated in 96-well flat bottom tissue culture plates (Nunc, Denmark) and incubated at 37° C., 5% CO[0050] 2 for 24 hours prior to initiation of treatment. The cells were then washed with PBS and treated with 1-500 ng/ml vinblastine sulphate (Calbiochem, La Jolla, Calif.) for 24 hours, in groups of eight wells per dose. The cells were then pulsed for 6 hrs with 2 μCi/well of methyl-3H-thymidine (Amersham Life Science, Buckinghamshire, England). The plates were frozen, thawed and the DNA harvested onto a filtermat using a Titertek Cell Harvester. Incorporated radioactivity was measured on Wallac 1205 BetaPlate Scintillation Counter (Wallac Oy, Finland) and proliferation was expressed as a percentage of 3H-thymidine in treated cells vs. that in controls.
  • In vivo tumor growth assessment: SK-N-MC, cells were harvested using 1% Trypsin EDTA (GibcoBRL, Gaithesburg, Md.), and single cell suspension of 2×106 cells in 0.2 ml of growth media was injected subcutaneously into the flanks of 4-6 week old CB-17 SCID mice (Charles River, St-Constant, Quebec). Approximately 3 weeks later, most tumors had grown to ˜0.75 cm[0051] 3, and mice were randomized into groups of 5 animals. Two independent experiments were performed, each totaling 20 animals in 4 groups. The treatment was as follows:
  • Group I (Control)—0.4 ml of PBS (DC101 vehicle) i.p. every three days and 0.15 ml injectable saline (vinblastine vehicle) i.p. every three days. [0052]
  • Group II—0.4 ml of 2 mg/ml DC101 antibody (800 μg/mouse) (24) every three days and 0.15 ml of injectable saline i.p. every three days [0053]
  • Group III—vinblastine sulfate 0.75 mg/m[0054] 2 i.p. bolus at the start of therapy, followed by 1 mg/m2/day via subcutaneous Alzet osmotic pumps (Alza Corp,Palo Alto, Calif.) for 3 weeks, followed by maintenance therapy with 0.15 ml of 0.067 mg/ml vinblastine sulfate (1.5 mg/m2) i.p. every three days, and 0.4 ml of PBS i.p. every three days
  • Group IV—combination of DC101 and vinblastine at doses identical to the single agent groups. [0055]
  • The body weight, tumor size and general clinical status of the animals were recorded every 2-3 days. Perpendicular tumor diameters were measured using a vernier scale caliper and tumor volume was estimated using the formula for ellipsoid: (width[0056] 2×length)/2. Growth curves were statistically analyzed using repeated measures ANOVA. All animal care was in accordance with institutional guidelines. As required by institutional guidelines, the mice were sacrificed when tumor size reached 1.5 cm3 or 7.5-10% of their body weight.
  • Histology: All tumors were excised, fixed in 10% (v/v) formalin and processed for immunohistochemical analysis. To obtain adequate tissue for the combination treatment group, two mice were sacrificed at 7.5 weeks of treatment. Paraffin blocks were cut to 5 μm sections and stained with haematoxylin/eosin for morphology evaluation and with Apoptosis Detection System (Promega, Madison, Wis.) for assessment of programmed cell death. [0057]
  • Relative tumor vascularity assessed by an FITC-Dextran perfusion assay: The method was designed to assess the relative functionality of the tumor vasculature. 2×10[0058] 6 SK-N-AS neuroblastoma cells were injected into the flanks of CB-17 SCID mice. Tumors were allowed to grow to approximately 0.75 cm3 at which point tumor bearing mice were then treated with 1 mg/m2 vinblastine i.p. every three days, 800 μg DC101 i.p. every three days, combination of the two agents or saline as a control. At 14 days, when divergence in tumor growth between the treatment groups was clearly evident, 0.2 ml of 25 mg/ml FITC-Dextran in PBS (Sigma, St. Louis, Mo.) was injected systemically into the lateral tail vein of each mouse and allowed to circulate for 20-30 minutes. Mice were then sacrificed by cervical dislocation and blood samples were collected into heparinized tubes by cardiac puncture for assessment of systemic fluorescein levels. Tumors were resected from the surrounding connective tissue being careful to avoid spillage of intra-vascular contents, weighed and placed into tubes containing 1:10 dispase (Collaborative Research, Two Oaks, Bedford, Mass.). To normalize for dilution caused by the difference in tumor sizes, 1 ml of 1:10 dispase was added per 0.5 g of tissue. Tumors were incubated in a dark 37° C. shaker overnight. The tissue was homogenized, centrifuged at 5000 rpm for 10 minutes, and the supernatant was collected and stored in the dark until further analysis. Blood samples were centrifuged immediately following collection, plasma separated and protected from light at 4° C. until analysis. Fluorescence readings were obtained on a FL600 Fluorescence Plate Reader (Bio-tek Instruments Inc., Winooski, Vt., USA), from a standard curve created by serial dilution of the FITC-dextran used for injection. The ratio of tumor fluorescence: plasma fluorescence was assumed to be reflective of the degree of tumor perfusion.
  • In vivo angiogenesis assessment by the Matrigel plug assay (5,25): Matrigel (Collaborative Biomedical Products, Bedford, Mass.) stored at -20° C., was thawed at 4° C. overnight and mixed with 500 ng/ml bFGF. 0.5 ml of this mixture was then injected subcutaneously into the shaved flanks of twenty 6-8 week old female Balb/cJ mice (Jackson Labs, Bar Harbor, Me.). Five mice, used as negative controls, were injected with Matrigel alone. Three days later, treatment mice were randomized into four groups as follows: [0059]
  • Group I—saline i.p., [0060]
  • Group II—800 μg DC101 i.p. [0061]
  • Group III—1 mg/m[0062] 2 vinblastine i.p.
  • Group IV—combination therapy. [0063]
  • All 25 mice were treated on day 4 and 7 and sacrificed on day 10. Blood samples were collected into heparinized tubes by cardiac puncture, centrifuged immediately following their collection, plasma separated and protected from light at 4° C. The Matrigel plugs were resected from surrounding connective tissues, placed into tubes containing 1 ml of 1:10 dispase and incubated in the dark in a 37° C. shaker overnight. The following day, the plugs were homogenized, centrifuged at 5000 rpm for 10 minutes and supematant saved in the dark for analysis of fluorescence. Fluorescence readings were obtained on FL600 Fluorescence Plate Reader using a standard curve created by serial dilution of FITC-dextran used for injection. Angiogenic response was expressed as a ratio of Matrigel plug fluorescence: plasma fluorescence. [0064]
  • In vitro determination of differential drug sensitivity: Prior to undertaking our in vivo experiments we established a dose of vinblastine, at which significant toxicity of endothelial, but not tumor, cells was observed. To do so, we optimized growth conditions to achieve comparable levels of mitotic activity in two human neuroblastoma cell lines (SK-NM-C and SK-N-AS) and HUVEC. All three cell lines were grown in DMEM with 10% bovine serum, but the HUVEC were grown on gelatinized plates and in the presence of additional growth factors (bFGF and EGF). The untreated controls show similar levels of [0065] 3H-Thymidine incorporation for all three cell lines thus eliminating the concern that the differences in proliferation may be inherent. At the higher concentrations of vinblastine used (e.g.100-400 ng/ml) all three cell populations were strongly inhibited, especially HUVEC. In striking contrast, at the lowest concentrations (e.g. 0.78 ng/ml) vinblastine retained almost the same degree of inhibitory activity against HUVEC, whereas anti-proliferative activity against two tumor cell lines was not. The source of this differential sensitivity is not clear, but it should be noted that at least one of the tumor cell lines, SK-N-MC, is positive for multidrug resistance-associated protein (MRP) (26). These in vitro findings suggest that the lowering of the usual maximum tolerated dose (MTD) used in the clinic may allow retention of good vinblastine activity against dividing endothelial cells present in tumors.
  • In vivo tumor growth assessment: Building on this in vitro difference in sensitivity to vinblastine, we went on to evaluate lower doses of vinblastine in an in vivo model, using an increased dose frequency to maximize the endothelial injury. Xenografts of either SK-N-MC neuroepithelioma or SK-N-AS neuroblastoma cell lines were implanted subcutaneously in the flanks of 4-6 week old. CB-17 SCID mice and grown to ˜0.75 cm[0066] 3 before initiation of treatment. The first treatment group, treated with DC101, an anti-Flk1 receptor antibody shown previously to inhibit growth of different kinds of human xenografts in mice and in mouse tumor models (5), showed an anticipated effectiveness in inhibiting tumor growth, but, its effect was not sustained. The findings in the second treatment group (vinblastine alone), were even more surprising. This agent, traditionally thought to act by inhibiting tumor cell proliferation through inhibition of tubulin assembly, produced significant, albeit not sustained, regression of tumor growth even though used at subclinical low-dose,. This growth delay in the vinblastine group was further potentiated with the simultaneous treatment with the anti-flk-1 antibody, DC101. The combination treatment induced an initial response comparable to the other treatment groups but then caused further, long term, tumor regression. To date, the mice in combination therapy group have not manifested any resistance to the treatment or recurrence of disease, despite almost seven months of continuous treatment. The mice remain healthy, with almost no evidence of tumor, except for a small, barely palpable remnant in one of the mice. [What were the doses used?]
  • Toxicity evaluation: Anti-vascular therapy would be expected to show minimal toxicity in the post-natal stage of development. To evaluate this aspect of DC101/vinblastine combination therapy, the health status of the mice was monitored. Weight was plotted at regular intervals and considered a surrogate for evaluation of systemic well being, anorexia, or failure to thrive. No significant differences in weights were seen between the four groups. The weight curve of the DC101 group parallels very closely that of the control group. The vinblastine group showed some weight gain retardation, but the differences never became significantly different from controls. Similarly, the toxicity profile in the combination treatment group was very similar to those in the single agent groups, with the exception of a transient episode of weight loss associated with diarrhea. The episode lasted approximately 2-3 weeks and was unlikely to be due to the therapy as the mice recovered without interruption of treatment. Other usual signs of drug toxicity in mice such as ruffled fur, anorexia, cachexia, skin tenting (due to dehydration), skin ulcerations or toxic deaths, were not seen at the doses used in our experiments. Diarrhea, a common sign of vinblastine toxicity when doses of 10 mg/m[0067] 2 are used, was generally not observed, except for the above mentioned episode.
  • Histopathologic analysis: To further elucidate the mechanisms involved in the tumor regression following treatment with vinblastine, DC101, or the combined therapy, tissue histopathology assessment was undertaken. Cancer cells with high nuclear to cytoplasmic ratio form cuffs around central vessels, and apoptotic cells characterized by pyknotic nuclei and cytoplasmic blebbing, are only evident as a thin rim at the periphery of the cuffs. The nuclei of these cells stain strongly for terminal deoxynucleotidyl transferase (TUNEL) reactivity, as expected for cells undergoing apoptosis. Vinblastine alone or DC101 treatment alone both show an increase in the width of the apoptotic rims, suggesting the cells most distal to the tumor vasculature are primarily affected, but a large percentage of viable tumor cells still survive in the center of the cuff. In contrast, histology of the combined therapy group, as would be predicted by the regression in tumor size in this treatment group at the time of analysis, shows overwhelming loss of both cell viability and pre-existing tumor architecture. There is a close similarity of the appearance of H/E and TUNEL stain. Interestingly, we observed signs of endothelial cell toxicity in all of the treatment groups. Rather than a typical single layer of flattened endothelial cells surrounding the vascular lumen in untreated group, we observed edema, and detachment from surrounding basement membrane and leading to complete vascular wall disintegration and tumor cell death. [0068]
  • Tumor perfusion by assessment of intravascular fluorescence: To further explore the possibility that tumor regression induced with treatment using DC101 and vinblastine was indeed due to the vascular injury, rather than a direct anti-tumor cell effect, we assessed tumor perfusion directly by using a FITC-Dextran fluorescence method. Mice carrying established subcutaneous SK- N-AS human neuroblastoma xenografts (˜0.75 cm[0069] 3) were randomized into four groups and treated systemically with either saline control, DC101, vinblastine or combination therapy for 10 days. FITC-Dextran was injected into the lateral tail vein and equilibrated throughout the vascular compartment. The majority of the blood-borne dextran, because of its 150 kDa size, remains intravascular, and despite some perivascular losses due to changes in vascular permeability and the possibility of interstitial hemorrhages, the fluorescence is reflective of the overall volume of blood passing through the tumor vasculature. Since our therapy is chronic in its nature, changes in intra-tumoral vascular/blood volume are likely to represent structural changes rather than transient fluxes in vascular permeability. By these criteria DC101 alone caused a 47 % decrease in tumor perfusion, whereas vinblastine alone resulted in a 41% decrease, and the combination of the two drugs resulted in 65% perfusion inhibition. Of interest is the appreciable difference in gross vascularity in the corresponding tumor specimens.
  • Effects of chemotherapy treatments on in vivo angiogenesis: The direct assessment of tumor vasculature does not provide any clues as to whether the apparent vascular inhibition within the tumor is a primary cause or a secondary consequence of the tumor regression. Evidence for the former would provide support for the hypothesis that low-dose vinblastine treatment alone is potentially anti-angiogenic, and the extent of this anti-angiogenic effect may be further enhanced by concurrent treatment with DC101. Again, the ratio between intra- and extra-vascular volume within the tumor could be also somewhat affected by transient changes in vascular permeability (27). To address these questions, we repeated the same fluorescence measurement using an in vivo Matrigel plug angiogenesis assay. Four treatment groups were treated with an identical therapeutic regimen as in the tumor perfusion experiment of example 11. The regression of vascularity in subcutaneously implanted Matrigel pellets was quantitatively assessed by measuring the fluorescence of circulating FITC-labeled dextran. DC101 administration inhibited bFGF induced vascularization to 50% of the positive control group, and vinblastine administration inhibited vascularization to 62.5% of the positive control group. There was again an enhanced effect with combination therapy, which reduced the Matrigel pellet fluorescence, and by inference vascularization to 29.2% of control, a level only marginally different to the negative control (Matrigel not supplemented with growth factors). [0070]
  • Thus, large (0.75 cm[0071] 3) established human neuroblastoma xenografts could be induced to completely regress with this combination strategy, whereas either agent alone caused only partial and temporary regressions with relapses observed in all animals treated at between 30-50 days after initiation of the individual therapy treatments. In striking contrast, a fully regressed state could be induced and maintained for as long as the combination therapy was maintained, which in our case was 200 days, in the absence of any significant toxicity, as assessed by lack of weight loss. No myelosuppression has been observed.
  • The dose of vinblastine used in our experiments was in the range of 1.5 mg/m[0072] 2, every 3 days, which is approximately 3 times the MTD of this drug in humans, and {fraction (1/16)}- {fraction (1/20)} of the MTD in mice, given the fact that the MTD of vinblastine in mice is 4-5 times higher than in humans (33,34). Using the Matrigel plug assay, we demonstrated that continuous low dose vinblastine administration can cause a direct anti-angiogenic effect in vivo. The combined effect with DC101 was significant.
  • 1 16 1 10 PRT Mouse 1 Gly Phe Asn Ile Lys Asp Phe Tyr Met His 1 5 10 2 17 PRT Mouse 2 Trp Ile Asp Pro Glu Asn Gly Asp Ser Asp Tyr Ala Pro Lys Phe Gln Gly 1 5 10 15 3 8 PRT Mouse 3 Tyr Tyr Gly Asp Tyr Glu Gly Tyr 1 5 4 10 PRT Mouse 4 Ser Ala Ser Ser Ser Val Ser Tyr Met His 1 5 10 5 7 PRT Mouse 5 Ser Thr Ser Asn Leu Ala Ser 1 5 6 9 PRT Mouse 6 Gln Gln Arg Ser Ser Tyr Pro Phe Thr 1 5 7 117 PRT Mouse 7 Gln Val Lys Leu Gln Gln Ser Gly Ala Glu Leu Val Gly Ser Gly Ala 1 5 10 15 Ser Val Lys Leu Ser Cys Thr Thr Ser Gly Phe Asn Ile Lys Asp Phe 20 25 30 Tyr Met His Trp Val Lys Gln Arg Pro Glu Gln Gly Leu Glu Trp Ile 35 40 45 Gly Trp Ile Asp Pro Glu Asn Gly Asp Ser Asp Tyr Ala Pro Lys Phe 50 55 60 Gln Gly Lys Ala Thr Met Thr Ala Asp Ser Ser Ser Asn Thr Ala Tyr 65 70 75 80 Leu Gln Leu Ser Ser Leu Thr Ser Glu Asp Thr Ala Val Tyr Tyr Cys 85 90 95 Asn Ala Tyr Tyr Gly Asp Tyr Glu Gly Tyr Trp Gly Gln Gly Thr Thr 100 105 110 Val Thr Val Ser Ser 115 8 108 PRT Mouse 8 Asp Ile Glu Leu Thr Gln Ser Pro Ala Ile Met Ser Ala Ser Pro Gly 1 5 10 15 Glu Lys Val Thr Ile Thr Cys Ser Ala Ser Ser Ser Val Ser Tyr Met 20 25 30 His Trp Phe Gln Gln Lys Pro Gly Thr Ser Pro Lys Leu Trp Ile Tyr 35 40 45 Ser Thr Ser Asn Leu Ala Ser Gly Val Pro Ala Arg Phe Ser Gly Ser 50 55 60 Gly Ser Gly Thr Ser Tyr Ser Leu Thr Ile Ser Arg Met Glu Ala Glu 65 70 75 80 Asp Ala Ala Thr Tyr Tyr Cys Gln Gln Arg Ser Ser Tyr Pro Phe Thr 85 90 95 Phe Gly Ser Gly Thr Lys Leu Glu Ile Lys Arg Ala 100 105 9 30 DNA Mouse 9 ggcttcaaca ttaaagactt ctatatgcac 30 10 51 DNA Mouse 10 tggattgatc ctgagaatgg tgattctgat tatgccccga agttccaggg c 51 11 24 DNA Mouse 11 tactatggtg actacgaagg ctac 24 12 30 DNA Mouse 12 agtgccagct caagtgtaag ttacatgcac 30 13 21 DNA Mouse 13 agcacatcca acctggcttc t 21 14 27 DNA Mouse 14 cagcaaagga gtagttaccc attcacg 27 15 351 DNA Mouse 15 caggtcaagc tgcagcagtc tggggcagag cttgtggggt caggggcctc agtcaaattg 60 tcctgcacaa cttctggctt caacattaaa gacttctata tgcactgggt gaagcagagg 120 cctgaacagg gcctggagtg gattggatgg attgatcctg agaatggtga ttctgattat 180 gccccgaagt tccagggcaa ggccaccatg actgcagact catcctccaa cacagcctac 240 ctgcagctca gcagcctgac atctgaggac actgccgtct attactgtaa tgcatactat 300 ggtgactacg aaggctactg gggccaaggg accacggtca ccgtctcctc a 351 16 324 DNA Mouse 16 gacatcgagc tcactcagtc tccagcaatc atgtctgcat ctccagggga gaaggtcacc 60 ataacctgca gtgccagctc aagtgtaagt tacatgcact ggttccagca gaagccaggc 120 acttctccca aactctggat ttatagcaca tccaacctgg cttctggagt ccctgctcgc 180 ttcagtggca gtggatctgg gacctcttac tctctcacaa tcagccgaat ggaggctgaa 240 gatgctgcca cttattactg ccagcaaagg agtagttacc cattcacgtt cggctcgggg 300 accaagctgg aaataaaacg ggcg 324

Claims (58)

What is claimed is:
1. A method of treating or controlling an angiogenic dependent condition in a mammal comprising:
administering an anti-angiogenic molecule and
a chemotherapeutic agent to the mammal, in amounts and frequencies effective, in combination, to produce a regression or arrest of said condition while minimizing or preventing significant toxicity of the chemotherapeutic agent.
2. The method of claim 1, wherein the anti-angiogenic condition is selected from the group consisting of a neoplasm, a collagen-vascular disease or an auto-immune disease.
3. The method of claim 2, wherein the neoplasm is a solid tumor.
4. The method of claim 3, wherein the solid tumor is selected from the group consisting of breast carcinoma, lung carcinoma, prostate carcinoma, colon carcinoma, prostate carcinoma, ovarian carcinoma, neuroblastoma, central nervous system tumor, neuroblastoma, glioblastoma multiforme or melanoma.
5. The method of claim 1, wherein the mammal is a human.
6. The method of claim 1, wherein the anti-angiogenic molecule inhibits or blocks the action of an vascular endothelium survival factor.
7. The method of claim 6, wherein the vascular endothelial survival factor is selected from the group consisting of VEGF, VEGF receptor,αvβ3. αvβ3 receptor, Tie2/tek ligand, Tie2/tek, endoglin ligand, endoglin, neuropilin ligand, neuropilin, thrombospondin ligand, thrombospondin, PDGFα, PDGFα receptor, PDGFβ, PDGFβ receptor, aFGF, aFGF receptor, bFGF, bFGF receptor, TGFβ, TGFβ receptor, EGF, EGF receptor, angiostatin, angiostatin receptor, angiopoetin, angiopoeitin receptor, PLGF, PLGF receptor, VPF, or VPF receptor.
8. The method of claim 6, wherein the vascular endothelial survival factor is a receptor.
9. The method of claim 8, wherein the vascular endothelial survival factor is an angiogenesis growth factor receptor.
10. The method of claim 9, wherein the angiogenesis growth factor receptor is a VEGF receptor.
11. The method of claim 10, wherein the VEGF receptor is selected from the group consisting of flk-1/KDR receptor, or flt-4 receptor.
12. The method of claim 6, wherein the vascular endothelial survival factor is a ligand to a receptor.
13. The method of claim 10, wherein the ligand is selected from the group consisting of VEGF, VEGF-B, VEGF-C, or VEGF-D.
14. The method of claim 6, wherein the anti-angiogenic molecule is selected from the group consisting of an antibody, antibody fragment, small molecule or peptide.
15. The method of claim 14, wherein the molecule is an antibody or fragment selected from the group consisting of mouse, rat, rabbit, chimeric, humanized or human antibody or fragment.
16. The method of claim 6, wherein the anti-angiogenic molecule is IMC-1C11.
17. The method of claim 16, wherein the IMC-1C11 is administered in a dose of from about 5 mg/m2 to about 700 mg/m2 from about daily to about every 7 days.
18. The method of claim 17, wherein the IMC-1C11 is administered in a dose of from about 7.5 mg/m2 to about 225 mg/m2, about twice per week.
19. The method of claim 16, wherein the IMC-1C11 is administered at a dose and frequency sufficient to substantially saturate the VEGF receptor.
20. The method of claim 6, wherein the anti-angiogenic molecule is administered in a dose and frequency sufficient to substantially saturate the target of the anti-angiogenic molecule.
21. The method of claim 1, wherein the chemotherapeutic agent is selected from the group consisting of vinca alkaloid, camptothecan, taxane, or platinum analogue.
22. The method of claim 21, wherein the chemotherapeutic agent is selected from the group consisting of vincristine, vinblastine, vinorelbine, vindesine, paclitaxel, docetaxel, 5 FU, cisplatin, carboplatin, iranotecan, topotecan or cyclophosphamide.
23. The method of claim 22, wherein the chemotherapeutic agent is administered at less than about 50% of the maximum tolerated dose.
24. The method of claim 23, wherein the chemotherapeutic agent is administered at less than about 20% of the maximum tolerated dose.
25. The method of claim 24, wherein the chemotherapeutic agent is administered at less than about 10% of the maximum tolerated dose.
26. The method of claim 22, wherein the chemotherapeutic agent is vinblastine administered in a dose from about 0.5 mg/m2 to about 3 mg/m2 from about once every 3 days to about once every 7 days.
27. The method of claim 1, wherein the chemotherapeutic agent is administered in a dosage and frequency that is of substantially equivalent efficacy to vinblastine in a dose from about 0.5 mg/M2 to about 3 mg/m2 from about once every 3 days to about once every 7 days.
28. The method of claim 1, wherein the chemotherapeutic agent is administered more frequently than about once every three weeks.
29. The method of claim 28, wherein the chemotherapeutic agent is administered more frequently than about every seven days.
30. A kit for treating an angiogenic dependent condition in a mammal comprising:
an anti-angiogenic molecule; and,
a chemotherapeutic agent, to be administered in amounts and frequencies effective, in combination, to produce a regression or arrest of the condition while minimizing or preventing significant toxicity of the chemotherapeutic agent, when administered in combination.
31. The kit of claim 30, wherein the angiogenic dependent condition is selected from the group consisting of neoplasm, collagen-vascular disease or autoimmune disease.
32. The kit of claim 31, wherein the neoplasm is a solid tumor.
33. The kit claim 32, wherein the solid tumor is selected from the group consisting of breast carcinoma, lung carcinoma, prostate carcinoma, colon carcinoma, prostate carcinoma, ovarian carcinoma, neuroblastoma, central nervous system tumor, neuroblastoma, glioblastoma multiforme or melanoma.
34. The kit of claim 30, wherein the mammal is a human.
35. The kit of claim 30, wherein the anti-angiogenic molecule inhibits or blocks the action of a vascular endothelium survival factor.
36. The kit of claim 35, wherein the vascular endothelial survival factor is selected from the group consisting of VEGF, VEGF receptor,αvβ3 . αvβ3 receptor, Tie2/tek ligand, Tie2/tek, endoglin ligand, endoglin, neuropilin ligand, neuropilin, thrombospondin ligand, thrombospondin, PDGFα, PDGFα receptor, PDGFβ, PDGFβ receptor, aFGF, aFGF receptor, bFGF, bFGF receptor, TGFβ, TGFβ receptor, EGF, EGF receptor, angiostatin, angiostatin receptor, angiopoetin, angiopoeitin receptor, PLGF, PLGF receptor, VPF, or VPF receptor.
37. The kit of claim 30, wherein the vascular endothelial survival factor is a receptor.
38. The kit of claim 37, wherein the vascular endothelial survival factor is a an angiogenesis growth factor receptor.
39. The kit of claim 37, wherein the angiogenesis growth factor receptor is a VEGF receptor.
40. The kit of claim 39, wherein the VEGF receptor is selected from the group consisting of flk-1/KDR receptor, or flt-4 receptor.
41. The kit of claim 35, wherein the vascular endothelial survival factor is a ligand for a receptor.
42. The kit of claim 39, wherein the ligand is selected from the group consisting of VEGF, VEGF-B, VEGF-C, or VEGF-D.
43. The kit of claim 35, wherein the anti-angiogenic molecule is selected from the group consisting of an antibody, antibody fragment, small molecule or peptide.
44. The kit of claim 43, wherein the molecule is an antibody or fragment selected from the group consisting of mouse, rat, rabbit, chimeric, humanized or human antibody or fragment.
45. The kit of claim 35, wherein the antibody is IMC-1C11.
46. The kit of claim 45, wherein the IMC-1C11 is provided for administration in a dose of from about 5 mg/m2 to about 700 mg/m2 about every 1 day to about every 7 days.
47. The kit of claim 46, wherein the IMC-1C11 is provided for administration in a dose of from about 7.5 mg/m2 to about 225 mg/m2, about twice per week.
48. The kit of claim 46, wherein the IMC-1C11 is provided for administration at a dose and frequency sufficient to substantially saturate the VEGF receptor.
49. The kit of claim 35, wherein the anti-angiogenic molecule is provided for administration in a dose and frequency sufficient to substantially saturate the target of the anti-angiogenic molecule.
50. The kit of claim 30, wherein the chemotherapeutic agent is selected from the group consisting of a vinca alkaloid, a camptothecan, a taxane, or a platinum analogue.
51. The kit of claim 50, wherein the chemotherapeutic agent is selected from the group consisting of vincristine, vinblastine, vinorelbine, vindesine, paclitaxel, docetaxel, 5 FU, cisplatin, carboplatin, iranotecan, topotecan or cyclophosphamide.
52. The kit of claim 51, wherein the chemotherapeutic agent is provided for administration at less than about 50% of the maximum tolerated dose.
53. The kit of claim 49, wherein the chemotherapeutic agent is provided for administration at less than about 20% of the maximum tolerated dose.
54. The kit of claim 52, wherein the chemotherapeutic agent is provided for administration at less than about 10% of the maximum tolerated dose.
55. The kit of claim 51, wherein the chemotherapeutic agent is vinblastine, provided for administration in a dose from about 0.5 mg/m2 to about 3 mg/m2 from about once every 3 days to about once every 7 days.
56. The kit of claim 30, wherein the chemotherapeutic agent is provided for administration at a dosage and frequency that is of substantially equivalent efficacy to vinblastine is a dose from about 0.5 mg/m2 to about 3 mg/m2 from about once every 3 days to about once every 7 days.
57. The kit of claim 30, wherein the chemotherapeutic agent is provided for administration more frequently than about every three weeks.
58. The kit of claim 57, wherein the chemotherapeutic agent is provided for administration more frequently than about every seven days.
US10/203,399 2000-01-28 2001-01-29 Therapeutic method for reducing angiogenesis Abandoned US20040248781A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/203,399 US20040248781A1 (en) 2000-01-28 2001-01-29 Therapeutic method for reducing angiogenesis

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US17879100P 2000-01-28 2000-01-28
US09/539,692 US7740841B1 (en) 2000-01-28 2000-03-31 Therapeutic method for reducing angiogenesis
US10/203,399 US20040248781A1 (en) 2000-01-28 2001-01-29 Therapeutic method for reducing angiogenesis
PCT/US2001/002839 WO2001054723A1 (en) 2000-01-28 2001-01-29 Therapeutic method for reducing angiogenesis

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/539,692 Continuation-In-Part US7740841B1 (en) 2000-01-28 2000-03-31 Therapeutic method for reducing angiogenesis

Publications (1)

Publication Number Publication Date
US20040248781A1 true US20040248781A1 (en) 2004-12-09

Family

ID=26874652

Family Applications (3)

Application Number Title Priority Date Filing Date
US09/539,692 Expired - Fee Related US7740841B1 (en) 2000-01-28 2000-03-31 Therapeutic method for reducing angiogenesis
US10/203,399 Abandoned US20040248781A1 (en) 2000-01-28 2001-01-29 Therapeutic method for reducing angiogenesis
US12/817,676 Abandoned US20100260776A1 (en) 2000-01-28 2010-06-17 Therapeutic method for reducing angiogenesis

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/539,692 Expired - Fee Related US7740841B1 (en) 2000-01-28 2000-03-31 Therapeutic method for reducing angiogenesis

Family Applications After (1)

Application Number Title Priority Date Filing Date
US12/817,676 Abandoned US20100260776A1 (en) 2000-01-28 2010-06-17 Therapeutic method for reducing angiogenesis

Country Status (6)

Country Link
US (3) US7740841B1 (en)
EP (2) EP2301579A1 (en)
JP (1) JP2003520826A (en)
AU (1) AU2001231223A1 (en)
CA (1) CA2398503A1 (en)
WO (1) WO2001054723A1 (en)

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030175274A1 (en) * 2001-04-13 2003-09-18 Rosen Craig A. Vascular endothelial growth factor 2
US20030215921A1 (en) * 2000-08-04 2003-11-20 Timothy Coleman Vascular endothelial growth factor-2
US20040018974A1 (en) * 2002-03-01 2004-01-29 Christophe Arbogast Multivalent constructs for therapeutic and diagnostic applications
US20040210041A1 (en) * 2002-03-01 2004-10-21 Christophe Arbogast Multivalent constructs for therapeutic and diagnostic applications
US20050059117A1 (en) * 2001-04-13 2005-03-17 Human Genome Sciences, Inc. Vascular endothelial growth factor 2
US20050100963A1 (en) * 2002-03-01 2005-05-12 Dyax Corporation KDR and VEGF/KDR binding peptides and their use in diagnosis and therapy
US20050147555A1 (en) * 2002-03-01 2005-07-07 Hong Fan Methods for preparing multivalent constructs for therapeutic and diagnostic applications and methods of preparing the same
US20050181979A1 (en) * 1999-02-08 2005-08-18 Human Genome Sciences, Inc. Vascular endothelial growth factor-2
US20050232921A1 (en) * 2001-04-13 2005-10-20 Rosen Craig A Vascular endothelial growth factor 2
US20050250700A1 (en) * 2002-03-01 2005-11-10 Sato Aaron K KDR and VEGF/KDR binding peptides
US20060024297A1 (en) * 2003-08-12 2006-02-02 Dyax Corp. Tie complex binding proteins
US20060057138A1 (en) * 2003-08-12 2006-03-16 Dyax Corporation Tie complex binding proteins
US20060263434A1 (en) * 2005-02-18 2006-11-23 Desai Neil P Combinations and modes of administration of therapeutic agents and combination therapy
US20080107607A1 (en) * 2002-03-01 2008-05-08 Bracco International B.V. Targeting vector-phospholipid conjugates
US20080152594A1 (en) * 2002-03-01 2008-06-26 Philippe Bussat Targeting vector-phospholipid conjugates
US20080213253A1 (en) * 2007-01-12 2008-09-04 Dyax Corp. Combination therapy for the treatment of cancer
US20080254021A1 (en) * 2003-08-12 2008-10-16 Dyax Corp. Tie1-binding ligands
US20080287384A1 (en) * 2005-09-19 2008-11-20 Marsha Rosner Methods of Identifying Agents Having Antiangiogenic Activity
US8623822B2 (en) 2002-03-01 2014-01-07 Bracco Suisse Sa KDR and VEGF/KDR binding peptides and their use in diagnosis and therapy
US8735394B2 (en) 2005-02-18 2014-05-27 Abraxis Bioscience, Llc Combinations and modes of administration of therapeutic agents and combination therapy
US9393318B2 (en) 2010-03-29 2016-07-19 Abraxis Bioscience, Llc Methods of treating cancer
US9399071B2 (en) 2010-06-04 2016-07-26 Abraxis Bioscience, Llc Methods of treatment of pancreatic cancer
US9540439B2 (en) 2012-10-08 2017-01-10 St. Jude Children's Research Hospital Therapies based on control of regulatory T cell stability and function via a neuropilin-1:semaphorin axis
US10660965B2 (en) 2010-03-29 2020-05-26 Abraxis Bioscience, Llc Methods of enhancing drug delivery and effectiveness of therapeutic agents
US20200330566A1 (en) * 2006-09-07 2020-10-22 Stemline Therapeutics, Inc. Cancer Stem Cell-Targeted Cancer Therapy

Families Citing this family (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6749853B1 (en) 1992-03-05 2004-06-15 Board Of Regents, The University Of Texas System Combined methods and compositions for coagulation and tumor treatment
US5965132A (en) 1992-03-05 1999-10-12 Board Of Regents, The University Of Texas System Methods and compositions for targeting the vasculature of solid tumors
WO1998031394A2 (en) 1997-01-22 1998-07-23 Board Of Regents, The University Of Texas System Tissue factor methods and compositions for coagulation and tumor treatment
EP1098665B9 (en) 1998-07-13 2003-08-13 The Board Of Regents, The University Of Texas System Cancer treatment methods using therapeutic conjugates that bind to aminophospholipids
DK1520588T3 (en) 1998-07-13 2015-03-23 Univ Texas Use of antibodies to aminophospholipids for cancer treatment
US6406693B1 (en) 1998-07-13 2002-06-18 Board Of Regents, The University Of Texas System Cancer treatment methods using antibodies to aminophospholipids
US6818213B1 (en) 1998-07-13 2004-11-16 Board Of Regents, The University Of Texas System Cancer treatment compositions comprising therapeutic conjugates that bind to aminophospholipids
AU2725000A (en) 1999-01-13 2000-08-01 Bayer Corporation Omega-carboxy aryl substituted diphenyl ureas as p38 kinase inhibitors
US8124630B2 (en) 1999-01-13 2012-02-28 Bayer Healthcare Llc ω-carboxyaryl substituted diphenyl ureas as raf kinase inhibitors
ES2223705T3 (en) 1999-04-28 2005-03-01 Board Of Regents, The University Of Texas System COMPOSITIONS AND METHODS FOR THE TREATMENT OF CANCER THROUGH INHIBITION SELECTIVE OF VEGF.
JP2004511430A (en) * 2000-05-24 2004-04-15 イムクローン システムズ インコーポレイティド Bispecific immunoglobulin-like antigen binding protein and production method
EP2286843A3 (en) 2000-06-02 2011-08-03 Bracco Suisse SA Compounds for targeting endothelial cells
US8263739B2 (en) 2000-06-02 2012-09-11 Bracco Suisse Sa Compounds for targeting endothelial cells, compositions containing the same and methods for their use
US7071164B2 (en) 2001-08-16 2006-07-04 Kimberly-Clark Worldwide, Inc. Anti-cancer and wound healing compounds
US6906036B2 (en) * 2001-08-16 2005-06-14 Kimberly-Clark Worldwide, Inc. Anti-aging and wound healing compounds
US7186693B2 (en) 2001-08-16 2007-03-06 Kimberly - Clark Worldwide, Inc. Metalloproteinase inhibitors for wound healing
SI1478358T1 (en) 2002-02-11 2013-09-30 Bayer Healthcare Llc Sorafenib tosylate for the treatment of diseases characterized by abnormal angiogenesis
ES2362931T3 (en) 2002-03-04 2011-07-15 Imclone Llc SPECIFIC HUMAN ANTIBODIES AGAINST KDR AND USES OF THE SAME.
GB0210783D0 (en) * 2002-05-10 2002-06-19 Polonelli Luciano Anti-microbial polypeptides
US7148194B2 (en) 2002-12-30 2006-12-12 Kimberly-Clark Worldwide, Inc. Method to increase fibronectin
SI2949658T1 (en) 2003-03-03 2018-10-30 Dyax Corp. Peptides that specifically bind HGF receptor (cMet) and uses thereof
US8796250B2 (en) 2003-05-20 2014-08-05 Bayer Healthcare Llc Diaryl ureas for diseases mediated by PDGFR
US7189700B2 (en) 2003-06-20 2007-03-13 Kimberly-Clark Worldwide, Inc. Anti-chrondrosarcoma compounds
KR101139557B1 (en) 2003-07-23 2012-04-30 바이엘 파마슈티칼스 코포레이션 Fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention of diseases and conditions
GB0406446D0 (en) * 2004-03-23 2004-04-28 Astrazeneca Ab Combination therapy
ATE531388T1 (en) * 2004-03-24 2011-11-15 Abbott Biotherapeutics Corp USE OF ANTI-ALPHA5BETA1 ANTIBODIES TO INHIBIT CANCER CELL PROLIFERATION
EP2329838A3 (en) 2004-11-22 2013-02-13 Centre National de la Recherche Scientifique Mutated netrin 4, fragments thereof and uses thereof as drugs
FR2878164B1 (en) * 2004-11-22 2007-03-23 Centre Nat Rech Scient Cnrse NEW USES OF NEOGENINE LIGANDS
BRPI0717964A2 (en) 2006-11-02 2013-12-17 Acceleron Pharma Inc ALK1 BINDER RECEIVER AND ANTAGONISTS AND USES OF THE SAME.
US10059756B2 (en) 2006-11-02 2018-08-28 Acceleron Pharma Inc. Compositions comprising ALK1-ECD protein
US8642031B2 (en) 2006-11-02 2014-02-04 Acceleron Pharma, Inc. Antagonists of BMP9, BMP10, ALK1 and other ALK1 ligands, and uses thereof
EA019595B1 (en) 2007-04-17 2014-04-30 Имклоун Элэлси PDGFRβ-SPECIFIC INHIBITORS
BRPI0911853A8 (en) 2008-05-02 2018-03-06 Acceleron Pharma Inc compositions and methods for angiogenesis modulation and pericyte composition
WO2012118969A2 (en) * 2011-03-01 2012-09-07 The U.S.A., As Represented By The Secretary, Department Of Health And Human Services Method of selecting cancer patients for anti-angiogenesis therapy in combination with chemotherapy

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4853221A (en) * 1980-11-13 1989-08-01 Warner-Lambert Company Method for treating non-small cell lung cancer, head and neck cancers and breast cancer
US5696153A (en) * 1994-05-16 1997-12-09 Napro Biotherapeutics, Inc. Therapeutic regimen for treating patients
US5811447A (en) * 1993-01-28 1998-09-22 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
US5840301A (en) * 1994-02-10 1998-11-24 Imclone Systems Incorporated Methods of use of chimerized, humanized, and single chain antibodies specific to VEGF receptors
US6302838B1 (en) * 1998-02-25 2001-10-16 Novartis Ag Cancer treatment with epothilones
US6342219B1 (en) * 1999-04-28 2002-01-29 Board Of Regents, The University Of Texas System Antibody compositions for selectively inhibiting VEGF
US6610688B2 (en) * 1999-12-21 2003-08-26 Sugen, Inc. 4-substituted 7-aza-indolin-2-ones and their use as protein kinase inhibitors

Family Cites Families (60)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4946718A (en) * 1985-12-31 1990-08-07 Protectaire Systems Co. Air curtain housing for conveyor mechanism
GB8607679D0 (en) 1986-03-27 1986-04-30 Winter G P Recombinant dna product
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5036003A (en) 1987-08-21 1991-07-30 Monsanto Company Antibodies to VPF
WO1989006692A1 (en) * 1988-01-12 1989-07-27 Genentech, Inc. Method of treating tumor cells by inhibiting growth factor receptor function
IL89489A0 (en) 1988-03-09 1989-09-10 Hybritech Inc Chimeric antibodies directed against human carcinoembryonic antigen
JPH02503867A (en) 1988-04-15 1990-11-15 プロテイン デザイン ラブズ インコーポレーテッド IL-2 receptor-specific chimeric antibody
ATE119198T1 (en) 1988-04-16 1995-03-15 Celltech Ltd METHOD FOR PRODUCING PROTEINS USING RECOMBINANT DNA.
US5036033A (en) * 1989-05-26 1991-07-30 Union Oil Company Of California Alkylation catalyst and processes for preparing
US5192744A (en) 1990-01-12 1993-03-09 Northwestern University Method of inhibiting angiogenesis of tumors
US5840692A (en) 1990-02-22 1998-11-24 Allegheny University Of The Health Sciences Peptide fragments and analogs of thrombospondin
US5200397A (en) 1990-02-22 1993-04-06 W. R. Grace & Co.-Conn. Use of peptide analogs of thrombospondin for the inhibition of angiogenic activity
US5648461A (en) 1990-02-22 1997-07-15 W.R. Grace & Co.-Conn Synthetic analogs of thrombospondin and therapeutic use thereof
US5190920A (en) 1990-09-24 1993-03-02 W. R. Grace & Co.-Conn. Method for using synthetic analogs of thrombospondin for inhibiting metastasis activity
US5190918A (en) 1990-02-22 1993-03-02 W. R. Grace & Co.-Conn. Peptide fragments and analogs of thrombospondin and methods of use
US5165922A (en) * 1990-05-22 1992-11-24 Bristol-Myers Squibb Company Synergistic tumor therapy with combinations of biologically active anti-tumor antibodies and chemotherapy
DE69232718T2 (en) 1991-02-22 2003-04-03 American Cyanamid Co Identification of a human receptor tyrosine kinase gene
US5190198A (en) 1991-03-27 1993-03-02 John A. Bott Vehicle article carrier
CA2064331C (en) 1991-03-28 2003-02-18 Marvin L. Bayne Vascular endothelial cell growth factor c subunit
CA2063055A1 (en) 1991-05-22 1992-11-23 Jacob Eyal Peptides having thrombospondin-like activity and their therapeutic use
AU2861692A (en) 1991-10-18 1993-05-21 Beth Israel Hospital Association, The Vascular permeability factor targeted compounds
DK1024191T3 (en) 1991-12-02 2008-12-08 Medical Res Council Preparation of autoantibodies displayed on phage surfaces from antibody segment libraries
US5770563A (en) 1991-12-06 1998-06-23 The United States Of America As Represented By The Department Of Health And Human Services Heparin- and sulfatide binding peptides from the type I repeats of human thrombospondin and conjugates thereof
US5877289A (en) 1992-03-05 1999-03-02 The Scripps Research Institute Tissue factor compositions and ligands for the specific coagulation of vasculature
US5660827A (en) 1992-03-05 1997-08-26 Board Of Regents, The University Of Texas System Antibodies that bind to endoglin
US6004555A (en) 1992-03-05 1999-12-21 Board Of Regents, The University Of Texas System Methods for the specific coagulation of vasculature
US5965132A (en) 1992-03-05 1999-10-12 Board Of Regents, The University Of Texas System Methods and compositions for targeting the vasculature of solid tumors
ATE239506T1 (en) 1992-03-05 2003-05-15 Univ Texas USE OF IMMUNOCONJUGATES FOR THE DIAGNOSIS AND/OR THERAPY OF VASCULARIZED TUMORS
WO1993020229A1 (en) 1992-04-03 1993-10-14 Genentech, Inc. ANTIBODIES TO ALPHAvBETA3 INTEGRIN
AU4025193A (en) 1992-04-08 1993-11-18 Cetus Oncology Corporation Humanized C-erbB-2 specific antibodies
US5696131A (en) * 1993-01-22 1997-12-09 Xenova Limited Treatment of cancers
US6824777B1 (en) * 1992-10-09 2004-11-30 Licentia Ltd. Flt4 (VEGFR-3) as a target for tumor imaging and anti-tumor therapy
US5399667A (en) 1993-03-05 1995-03-21 Washington University Thrombospondin receptor binding peptides
WO1994024163A2 (en) * 1993-04-09 1994-10-27 Chiron Corporation Bispecific antigen-binding molecules
US5399677A (en) 1993-12-07 1995-03-21 Genetics Institute, Inc. Mutants of bone morphogenetic proteins
US5861499A (en) 1994-02-10 1999-01-19 Imclone Systems Incorporated Nucleic acid molecules encoding the variable or hypervariable region of a monoclonal antibody that binds to an extracellular domain
US6448077B1 (en) 1994-02-10 2002-09-10 Imclone Systems, Inc. Chimeric and humanized monoclonal antibodies specific to VEGF receptors
US5753230A (en) 1994-03-18 1998-05-19 The Scripps Research Institute Methods and compositions useful for inhibition of angiogenesis
US5770565A (en) 1994-04-13 1998-06-23 La Jolla Cancer Research Center Peptides for reducing or inhibiting bone resorption
US6645933B1 (en) 1995-08-01 2003-11-11 Helsinki University Licensing Ltd. Oy Receptor ligand VEGF-C
WO1998033917A1 (en) 1994-11-14 1998-08-06 The Ludwig Institute For Cancer Research Vascular endothelial growth factor c (vegf-c) protein and gene, mutants thereof, and uses thereof
US5780426A (en) 1995-06-07 1998-07-14 Ixsys, Incorporated Fivemer cyclic peptide inhibitors of diseases involving αv β3
US5767071A (en) 1995-06-07 1998-06-16 Ixsys Incorporated Sevenmer cyclic peptide inhibitors of diseases involving αv β3
US5730977A (en) 1995-08-21 1998-03-24 Mitsui Toatsu Chemicals, Inc. Anti-VEGF human monoclonal antibody
US5981546A (en) 1996-08-29 1999-11-09 Merck & Co., Inc. Integrin antagonists
JPH10114680A (en) * 1996-10-08 1998-05-06 Toagosei Co Ltd Carcinostatic agent
US5684461A (en) 1996-10-18 1997-11-04 Jones; Richard Sidney Sleep prevention device for a driver of a vehicle
US5952341A (en) 1996-10-30 1999-09-14 Merck & Co., Inc. Integrin antagonists
US5919792A (en) 1996-10-30 1999-07-06 Merck & Co., Inc. Integrin antagonists
US6017925A (en) 1997-01-17 2000-01-25 Merck & Co., Inc. Integrin antagonists
US6884879B1 (en) * 1997-04-07 2005-04-26 Genentech, Inc. Anti-VEGF antibodies
WO1999016465A1 (en) 1997-09-30 1999-04-08 Beth Israel Deaconess Medical Center Method for inhibiting tumor angiogenesis in a living subject
US6100254A (en) * 1997-10-10 2000-08-08 Board Of Regents, The University Of Texas System Inhibitors of protein tyrosine kinases
EP1027437B8 (en) * 1997-10-29 2008-10-29 Genentech, Inc. Uses of the wnt-1 induced secreted polypeptide wisp-1
US6017926A (en) 1997-12-17 2000-01-25 Merck & Co., Inc. Integrin receptor antagonists
US6372719B1 (en) 1998-03-04 2002-04-16 Jay Cunningham ανβ3 integrin antagonists in combination with chemotherapeutic agents
DE19842415A1 (en) * 1998-09-16 2000-03-23 Merck Patent Gmbh Pharmaceutical preparation for treating e.g. tumors, thrombosis or inflammation, contains cyclic pentapeptide integrin inhibitor and chemotherapeutic agent and/or angiogenesis inhibitor
WO2000034337A1 (en) 1998-12-10 2000-06-15 Tsukuba Research Laboratory, Toagosei Co., Ltd. Humanized monoclonal antibodies against vascular endothelial cell growth factor
IL146480A0 (en) 1999-05-14 2002-07-25 Imclone Systems Inc Treatment of refractory human tumors with epidermal growth factor receptor antagonists
JP5835204B2 (en) 2012-12-20 2015-12-24 信越化学工業株式会社 Resist material and pattern forming method using the same

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4853221A (en) * 1980-11-13 1989-08-01 Warner-Lambert Company Method for treating non-small cell lung cancer, head and neck cancers and breast cancer
US5811447A (en) * 1993-01-28 1998-09-22 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
US5840301A (en) * 1994-02-10 1998-11-24 Imclone Systems Incorporated Methods of use of chimerized, humanized, and single chain antibodies specific to VEGF receptors
US5874542A (en) * 1994-02-10 1999-02-23 Imclone Systems Incorporated Single chain antibodies specific to VEGF receptors
US5696153A (en) * 1994-05-16 1997-12-09 Napro Biotherapeutics, Inc. Therapeutic regimen for treating patients
US6302838B1 (en) * 1998-02-25 2001-10-16 Novartis Ag Cancer treatment with epothilones
US6342219B1 (en) * 1999-04-28 2002-01-29 Board Of Regents, The University Of Texas System Antibody compositions for selectively inhibiting VEGF
US6610688B2 (en) * 1999-12-21 2003-08-26 Sugen, Inc. 4-substituted 7-aza-indolin-2-ones and their use as protein kinase inhibitors

Cited By (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7524501B2 (en) 1999-02-08 2009-04-28 Human Genome Sciences, Inc. Methods of treating an injury to, or disorder of, the eye involving photoreceptor proliferation by administering VEGF2 antibodies
US7223724B1 (en) 1999-02-08 2007-05-29 Human Genome Sciences, Inc. Use of vascular endothelial growth factor to treat photoreceptor cells
US20050181979A1 (en) * 1999-02-08 2005-08-18 Human Genome Sciences, Inc. Vascular endothelial growth factor-2
US20030215921A1 (en) * 2000-08-04 2003-11-20 Timothy Coleman Vascular endothelial growth factor-2
US7273751B2 (en) 2000-08-04 2007-09-25 Human Genome Science, Inc. Vascular endothelial growth factor-2
US20050232921A1 (en) * 2001-04-13 2005-10-20 Rosen Craig A Vascular endothelial growth factor 2
US20030175274A1 (en) * 2001-04-13 2003-09-18 Rosen Craig A. Vascular endothelial growth factor 2
US20050059117A1 (en) * 2001-04-13 2005-03-17 Human Genome Sciences, Inc. Vascular endothelial growth factor 2
US7208582B2 (en) 2001-04-13 2007-04-24 Human Genome Sciences, Inc. Vascular endothelial growth factor 2
US7794693B2 (en) 2002-03-01 2010-09-14 Bracco International B.V. Targeting vector-phospholipid conjugates
US8623822B2 (en) 2002-03-01 2014-01-07 Bracco Suisse Sa KDR and VEGF/KDR binding peptides and their use in diagnosis and therapy
US8642010B2 (en) 2002-03-01 2014-02-04 Dyax Corp. KDR and VEGF/KDR binding peptides and their use in diagnosis and therapy
US8632753B2 (en) 2002-03-01 2014-01-21 Bracco Suisse Sa Multivalent constructs for therapeutic and diagnostic applications
US9629934B2 (en) 2002-03-01 2017-04-25 Dyax Corp. KDR and VEGF/KDR binding peptides and their use in diagnosis and therapy
US20050147555A1 (en) * 2002-03-01 2005-07-07 Hong Fan Methods for preparing multivalent constructs for therapeutic and diagnostic applications and methods of preparing the same
US7211240B2 (en) 2002-03-01 2007-05-01 Bracco International B.V. Multivalent constructs for therapeutic and diagnostic applications
US20050100963A1 (en) * 2002-03-01 2005-05-12 Dyax Corporation KDR and VEGF/KDR binding peptides and their use in diagnosis and therapy
US7261876B2 (en) 2002-03-01 2007-08-28 Bracco International Bv Multivalent constructs for therapeutic and diagnostic applications
US20050027105A9 (en) * 2002-03-01 2005-02-03 Christophe Arbogast Multivalent constructs for therapeutic and diagnostic applications
US20080107607A1 (en) * 2002-03-01 2008-05-08 Bracco International B.V. Targeting vector-phospholipid conjugates
US20080152594A1 (en) * 2002-03-01 2008-06-26 Philippe Bussat Targeting vector-phospholipid conjugates
US9446155B2 (en) 2002-03-01 2016-09-20 Bracco Suisse Sa KDR and VEGF/KDR binding peptides and their use in diagnosis and therapy
US9056138B2 (en) 2002-03-01 2015-06-16 Bracco Suisse Sa Multivalent constructs for therapeutic and diagnostic applications
US9408926B2 (en) 2002-03-01 2016-08-09 Bracco Suisse S.A. KDR and VEGF/KDR binding peptides and their use in diagnosis and therapy
US8663603B2 (en) 2002-03-01 2014-03-04 Bracco Suisse Sa Multivalent constructs for therapeutic and diagnostic applications
US9381258B2 (en) 2002-03-01 2016-07-05 Bracco Suisse S.A. Targeting vector-phospholipid conjugates
US20040210041A1 (en) * 2002-03-01 2004-10-21 Christophe Arbogast Multivalent constructs for therapeutic and diagnostic applications
US7666979B2 (en) 2002-03-01 2010-02-23 Bracco International B.V. Methods for preparing multivalent constructs for therapeutic and diagnostic applications and methods of preparing the same
US9295737B2 (en) 2002-03-01 2016-03-29 Bracco Suisse Sa Targeting vector-phospholipid conjugates
US20040018974A1 (en) * 2002-03-01 2004-01-29 Christophe Arbogast Multivalent constructs for therapeutic and diagnostic applications
US7854919B2 (en) 2002-03-01 2010-12-21 Bracco, Suisse SA Multivalent constructs for therapeutic and diagnostic applications
US8551450B2 (en) 2002-03-01 2013-10-08 Philippe Bussat Targeting vector-phospholipid conjugates
US7910088B2 (en) 2002-03-01 2011-03-22 Bracco Suisse Sa Multivalent constructs for therapeutic and diagnostic applications
US7985402B2 (en) 2002-03-01 2011-07-26 Bracco Suisse Sa Targeting vector-phospholipid conjugates
US20050250700A1 (en) * 2002-03-01 2005-11-10 Sato Aaron K KDR and VEGF/KDR binding peptides
US7485297B2 (en) 2003-08-12 2009-02-03 Dyax Corp. Method of inhibition of vascular development using an antibody
US7871610B2 (en) 2003-08-12 2011-01-18 Dyax Corp. Antibodies to Tie1 ectodomain
US20080254021A1 (en) * 2003-08-12 2008-10-16 Dyax Corp. Tie1-binding ligands
US20060057138A1 (en) * 2003-08-12 2006-03-16 Dyax Corporation Tie complex binding proteins
US20060024297A1 (en) * 2003-08-12 2006-02-02 Dyax Corp. Tie complex binding proteins
US8034375B2 (en) 2005-02-18 2011-10-11 Abraxis Bioscience, Llc Combinations and modes of administration of therapeutic agents and combination therapy
US8735394B2 (en) 2005-02-18 2014-05-27 Abraxis Bioscience, Llc Combinations and modes of administration of therapeutic agents and combination therapy
US8268348B2 (en) 2005-02-18 2012-09-18 Abraxis Bioscience, Llc Combinations and modes of administration of therapeutic agents and combination therapy
US9101543B2 (en) 2005-02-18 2015-08-11 Abraxis Bioscience, Llc Combinations and modes of administration of therapeutic agents and combination therapy
US7758891B2 (en) 2005-02-18 2010-07-20 Abraxis Bioscience, Llc Combinations and modes of administration of therapeutic agents and combination therapy
US20090098210A1 (en) * 2005-02-18 2009-04-16 Desai Neil P Combinations and modes of administration of therapeutic agents and combination therapy
US20060263434A1 (en) * 2005-02-18 2006-11-23 Desai Neil P Combinations and modes of administration of therapeutic agents and combination therapy
US9561288B2 (en) 2005-02-18 2017-02-07 Abraxis Bioscience, Llc Combinations and modes of administration of therapeutic agents and combination therapy
US20080287384A1 (en) * 2005-09-19 2008-11-20 Marsha Rosner Methods of Identifying Agents Having Antiangiogenic Activity
US20200330566A1 (en) * 2006-09-07 2020-10-22 Stemline Therapeutics, Inc. Cancer Stem Cell-Targeted Cancer Therapy
US20080213253A1 (en) * 2007-01-12 2008-09-04 Dyax Corp. Combination therapy for the treatment of cancer
US9597409B2 (en) 2010-03-29 2017-03-21 Abraxis Bioscience, Llc Methods of treating cancer
US9393318B2 (en) 2010-03-29 2016-07-19 Abraxis Bioscience, Llc Methods of treating cancer
US10660965B2 (en) 2010-03-29 2020-05-26 Abraxis Bioscience, Llc Methods of enhancing drug delivery and effectiveness of therapeutic agents
US9399072B2 (en) 2010-06-04 2016-07-26 Abraxis Bioscience, Llc Methods of treatment of pancreatic cancer
US9399071B2 (en) 2010-06-04 2016-07-26 Abraxis Bioscience, Llc Methods of treatment of pancreatic cancer
US9820949B2 (en) 2010-06-04 2017-11-21 Abraxis Bioscience, Llc Methods of treatment of pancreatic cancer
US9540439B2 (en) 2012-10-08 2017-01-10 St. Jude Children's Research Hospital Therapies based on control of regulatory T cell stability and function via a neuropilin-1:semaphorin axis

Also Published As

Publication number Publication date
EP1261370A4 (en) 2004-08-25
EP1261370A1 (en) 2002-12-04
EP2301579A1 (en) 2011-03-30
JP2003520826A (en) 2003-07-08
AU2001231223A1 (en) 2001-08-07
WO2001054723A1 (en) 2001-08-02
US20100260776A1 (en) 2010-10-14
WO2001054723A9 (en) 2002-10-17
US7740841B1 (en) 2010-06-22
CA2398503A1 (en) 2001-08-02

Similar Documents

Publication Publication Date Title
US20040248781A1 (en) Therapeutic method for reducing angiogenesis
Davis et al. A phase 1b dose escalation study of Wnt pathway inhibitor vantictumab in combination with nab-paclitaxel and gemcitabine in patients with previously untreated metastatic pancreatic cancer
Veronese et al. Monoclonal antibodies in the treatment of colorectal cancer
JP4364510B2 (en) Combination therapy with receptor tyrosine kinase inhibitors and angiogenesis inhibitors
Lockhart et al. The epidermal growth factor receptor as a target for colorectal cancer therapy
von Baumgarten et al. Bevacizumab has differential and dose-dependent effects on glioma blood vessels and tumor cells
Manley et al. Therapies directed at vascular endothelial growth factor
US20020076408A1 (en) Combination radiation therapy and chemotherapy in conjunction with administration of growth factor receptor antibody
Galer et al. Dual inhibition of epidermal growth factor receptor and insulin‐like growth factor receptor I: Reduction of angiogenesis and tumor growth in cutaneous squamous cell carcinoma
AU2018253461A1 (en) Combination therapy for the treatment of glioblastoma
JP2009523813A (en) Specific therapies using integrin ligands to treat cancer
TW200838875A (en) Combination therapy with angiogenesis inhibitors
Costa et al. Developmental therapeutics for patients with breast cancer and central nervous system metastasis: current landscape and future perspectives
JP6563197B2 (en) Combination therapy for the treatment of ovarian cancer
Nielsen et al. Antiangiogenic therapy for breast cancer
Idbaih et al. Therapeutic application of noncytotoxic molecular targeted therapy in gliomas: growth factor receptors and angiogenesis inhibitors
CN102448497A (en) Continuous administration of integrin ligands for treating cancer
Kawaguchi et al. MEK inhibitor trametinib in combination with gemcitabine regresses a patient-derived orthotopic xenograft (PDOX) pancreatic cancer nude mouse model
MX2013000521A (en) Peptide for use in the treatment of breast cancer and/or bone metastases.
A Bareschino et al. The role of antiangiogenetic agents in the treatment of breast cancer
Beaudry et al. Potent antitumor effects of ZD6474 on neuroblastoma via dual targeting of tumor cells and tumor endothelium
Sánchez-Muñoz et al. Targeted therapy of metastatic breast cancer
Kochar et al. Contemporary updates on clinical trials of antiangiogenic agents in the treatment of glioblastoma multiforme
CN114340679A (en) Methods and medicaments for treating cancers that are non-responsive to inhibitors of PD-1/PD-L1 signaling
Modi et al. An update on epidermal growth factor receptor inhibitors

Legal Events

Date Code Title Description
AS Assignment

Owner name: SUNNYBROOK HEALTH SCIENCE CENTER, CANADA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:KERBEL, ROBERT;REEL/FRAME:014593/0979

Effective date: 20030429

STCB Information on status: application discontinuation

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