WO2000074634A2 - Methods and compositions for modulating cell proliferation and cell death - Google Patents

Methods and compositions for modulating cell proliferation and cell death Download PDF

Info

Publication number
WO2000074634A2
WO2000074634A2 PCT/US2000/040103 US0040103W WO0074634A2 WO 2000074634 A2 WO2000074634 A2 WO 2000074634A2 US 0040103 W US0040103 W US 0040103W WO 0074634 A2 WO0074634 A2 WO 0074634A2
Authority
WO
WIPO (PCT)
Prior art keywords
fgf
cell
cytotoxic agent
tumor
subject
Prior art date
Application number
PCT/US2000/040103
Other languages
French (fr)
Other versions
WO2000074634A9 (en
WO2000074634A3 (en
Inventor
Jessie L. S. Au
M. Guillaume Wientjes
Original Assignee
Au Jessie L S
Wientjes M Guillaume
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 Au Jessie L S, Wientjes M Guillaume filed Critical Au Jessie L S
Priority to AU57903/00A priority Critical patent/AU780454B2/en
Priority to EP00943429A priority patent/EP1206234A4/en
Priority to CA002377385A priority patent/CA2377385A1/en
Priority to IL14687200A priority patent/IL146872A0/en
Priority to JP2001501171A priority patent/JP2003503313A/en
Publication of WO2000074634A2 publication Critical patent/WO2000074634A2/en
Publication of WO2000074634A3 publication Critical patent/WO2000074634A3/en
Priority to IL146872A priority patent/IL146872A/en
Publication of WO2000074634A9 publication Critical patent/WO2000074634A9/en

Links

Classifications

    • 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
    • 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
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1825Fibroblast growth factor [FGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/00113Growth factors
    • A61K39/001132Fibroblast growth factors [FGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies

Definitions

  • Cancer therapy such as chemotherapy and radiation, targets proliferating cells and thereby causes undesired toxicity to normal host tissues that undergo continuous renewal, including the hematopoietic cells, cells in the lining of the gastrointestinal tract, and hair follicles.
  • Bone marrow suppression induced by cancer therapy is, at least in part, overcome by the use of hematopoietic growth factors, including erythropoietin, granulocytes colony-stimulating factor, and granulocyte-macrophage colony- stimulating factor (Gabrilove, J.L. and Goldie, D.W. (1993) In: Cancer, Principles & Practice of Oncology (eds. DeVita, V.T.
  • the invention is based, at least in part, on the elucidation of the role played by basic Fibroblast Growth Factor (bFGF) in the induction of broad spectrum resistance to anticancer agents in a number of tumors and metastatic lesions, and the role played by acidic FGF (aFGF) in amplifying the bFGF-induced resistance.
  • bFGF basic Fibroblast Growth Factor
  • aFGF acidic FGF
  • Inhibitors of aFGF/bFGF enhance the in vitro and in vivo activity of anticancer agents, and result in shrinkage and eradication of human xenograft tumors including lung metastasis and subcutaneous tumors in mice.
  • Methods of the invention use FGF antagonists to potentiate the antitumor effect of anticancer agents.
  • FGF agonists e.g., aFGF, e.g., bFGF
  • Methods of the invention use FGF agonists to protect normal cells from the cytotoxic effects of anticancer agents.
  • the invention features, a method of inhibiting unwanted cell growth or division, e.g., reducing or inhibiting the proliferation of, or enhancing the killing of, a cell, e.g., a hyperproliferative cell, e.g., a malignant cell or a benign hyperproliferative cell.
  • the method includes: contacting the cell with at least one cytotoxic agent, (e.g., a cytostatic agent, e.g., an agent that causes cell death) and at least one FGF antagonist, in an amount, which together, is effective to reduce or inhibit the proliferation of the cell, or induce cell killing.
  • the unwanted cell is the cell of an established tumor.
  • the invention features a method of improving the efficacy of an agent, e.g., a cytotoxic agent, in a subject.
  • the method includes: administering to the subject at least one agent, e.g., a cytotoxic agent; administering to the subject at least one FGF antagonist.
  • the FGF antagonist enhances the efficacy of the agent, e.g., a cytotoxic agent, relative to the effect of the cytotoxic agent in the absence of the FGF antagonist.
  • the FGF antagonist improves the efficacy of the cytotoxic agent against a cancer, e.g., an established tumor.
  • the invention features, a method of inhibiting unwanted cell growth or division, or inducing the killing of an unwanted cell (e.g., a hyperproliferative cell), e.g., a cell of an established tumor or a benign hyperproliferative cell, in a subject.
  • the method can be used to treat or prevent, in a subject, a disorder characterized by unwanted cell growth or division.
  • the method includes: administering to the subject at least one cytotoxic agent, (e.g., a cytostatic agent, an agent that causes cell death), and at least one FGF antagonist, in an amount, which together, is effective (e.g., therapeutically or prophylactically) to reduce or inhibit the growth or division of, or induce the killing of, the unwanted cell.
  • the unwanted cell is the cell of an established tumor.
  • the FGF antagonist inhibits or reduces the FGF- induced resistance to a broad spectrum of cytotoxic agents, i.e., agents with diverse structures and mechanisms of action, including but not limited to, antimicrotubule agents, topoisomerase I inhibitors, topoisomerase II inhibitors, antimetabohtes, mitotic inhibitors, alkylating agents, intercalating agents, agents capable of interfering with a signal transduction pathway (e.g., protein kinase C inhibitors, e.g., anti-hormones, e.g., antibodies against growth factor receptors), agents that promote apoptosis and/or necrosis, interferons, interleukins, tumor necrosis factors, and radiation.
  • cytotoxic agents i.e., agents with diverse structures and mechanisms of action, including but not limited to, antimicrotubule agents, topoisomerase I inhibitors, topoisomerase II inhibitors, antimetabohtes, mitotic inhibitors, alkylating agents, inter
  • the FGF antagonist comprises an inhibitor of bFGF. In a preferred embodiment, the FGF antagonist comprises an inhibitor of aFGF.
  • the FGF antagonist includes at least one bFGF inhibitor and at least one aFGF inhibitor.
  • the aFGF inhibitor potentiates the action of the bFGF inhibitor.
  • the FGF antagonist acts extracellularly, e.g., inhibits the binding of an FGF molecule to its receptor.
  • the FGF antagonist acts intracellularly, e.g., interacts with the intracellular domain of the FGF receptor, inhibits the intracellular effects of FGF.
  • the FGF antagonist is capable of binding to an FGF molecule or an FGF receptor; blocks the binding of FGF to a receptor; blocks the interaction of FGF with molecules that facilitate the binding of FGF to a receptor; and/or down regulates FGF receptor action.
  • the FGF molecule is bFGF and/or aFGF.
  • the FGF antagonist is other than suramin. In a preferred embodiment, the FGF antagonist is other than an antibody, e.g., an antibody against FGF or an FGF receptor.
  • the FGF antagonist inhibits or reverses the resistance to anticancer drugs induced by FGF (e.g. aFGF and/or bFGF) in cultured tumor cells under in vitro conditions.
  • FGF e.g. aFGF and/or bFGF
  • the determination of effect on cultured cells can be determined using the system described in Example XV.
  • the FGF antagonist improves the efficacy of an agent, e.g., a cytotoxic agent, in the subject, relative to the effect of the cytotoxic agent in the absence of the FGF antagonist.
  • the FGF antagonist improves the efficacy of the cytotoxic agent against an established tumor.
  • the FGF antagonist is present in an amount sufficient to block FGF (e.g., bFGF and/or aFGF) action, but is not sufficient to cause one or more of: (i) significant inhibition of cell proliferation; (ii) significant cell death in human and/or animal tumor cells, (iii) a measurable antitumor effect in a subject, e.g., a human subject; and/or (iv) significant cell cycle arrest.
  • FGF e.g., bFGF and/or aFGF
  • the determination of effect on cultured cells can be determined with the system described in Example XV.
  • the FGF antagonist is administered at levels such that the plasma concentration of the FGF antagonist that is present when the cytotoxic agent is present in plasma at pharmacologically active concentration does not result in one or more of: (i) significant cell cycle arrest, (ii) significant cell death, or (iii) significant inhibition of cell growth, e.g., the concentration in plasma is of a level that if the same concentration of FGF antagonist is provided in cultured cells at least 10, more preferably at least 25, more preferably at least 50, more preferably at least 70, more preferably at least 80, more preferably at least 90, and most preferably at least 99% of the treated cultured cells continue to be involved in one or more of: (i) cycling cells continue to progress through the cell cycle, (ii) cells remain viable, or (iii) cells remain capable of proliferating, following treatment with the FGF antagonist.
  • the determination of effect on cultured cells should be determined with the system described in Example XV.
  • the FGF antagonist is administered at levels that, although not resulting in significant cell cycle arrest, significant cell death, or significant inhibition of cell growth as described above, sensitizes the tumor cells to treatment with cytotoxic agents.
  • the levels of FGF antagonist result in a significant inhibition of FGF-mediated chemoresistance, e.g., the resistance of tumor cells to cytotoxic agents that is mediated by FGF.
  • the FGF antagonist inhibits at least 10, more preferably, at least 25, more preferably at least, 50, more preferably at least 70, more preferably at least 80, more preferably at least 90, and most preferably at least 99% of the FGF-mediated chemoresistance.
  • the determination of effect on cultured cells can be determined with the system described in Example XV.
  • the time period over which the FGF antagonist is administered or over which the FGF antagonist is maintained at a therapeutic level, e.g., at a plasma concentration that is sufficient to enhance the cytotoxic effect of the cytotoxic agent is less than 180, more preferably less than 90, more preferably less than 60, and most preferably less than 30 days.
  • the time period over which the FGF antagonist is administered or over which the FGF antagonist is maintained at a therapeutic level e.g., at a plasma concentration that is sufficient to enhance the cytotoxic effect of the cytotoxic agent does not begin substantially earlier or end substantial later than the period over which the cytotoxic agent is administered or over which the cytotoxic agent is maintained at a therapeutic level.
  • the time period over which the FGF antagonist is administered or over which the FGF antagonist is maintained at a therapeutic level e.g., at a plasma concentration that is sufficient to enhance the cytotoxic effect of the cytotoxic agent ends less than 180, more preferably less than 90, more preferably less than 60, and most preferably less than 30 days after the last day on which the cytotoxic agent is administered or the last day on which the cytotoxic agent is present at therapeutic levels.
  • the time period over which the FGF antagonist is administered or over which the FGF antagonist is maintained at a therapeutic level, e.g., at a plasma concentration that is sufficient to enhance the cytotoxic effect of the cytotoxic agent begins less than 180, more preferably less than 90, more preferably less than 60, and most preferably less than 30 days before the first day on which the cytotoxic agent is administered or the first day on which the cytotoxic agent is present at therapeutic levels.
  • the FGF antagonist is a protein or a peptide; is an antibody, e.g., a monoclonal, a murine antibody or a human antibody, or an antigen- binding fragment thereof.
  • the monoclonal antibody is a human antibody.
  • the antibodies can be of the various isotypes, including: IgG (e.g., IgGl, IgG2, IgG3, IgG4), IgM, IgAl, IgA2, IgD, of IgE.
  • the antibody is an IgG isotype.
  • the antibodies can be full-length (e.g., an IgGl or IgG4 antibody) or can include only an antigen-binding fragment (e.g., a Fab, F(ab')2, Fv or a single chain Fv fragment).
  • the FGF antagonist is a recombinant antibody, e.g., a chimeric or a humanized antibody, or an antigen binding fragment thereof, e.g., has a variable region, or at least a complementarity determining region (CDR), derived from a non-human antibody (e.g., murine), while the remaining portion(s) are human in origin.
  • a recombinant antibody e.g., a chimeric or a humanized antibody, or an antigen binding fragment thereof, e.g., has a variable region, or at least a complementarity determining region (CDR), derived from a non-human antibody (e.g., murine), while the remaining portion(s) are human in origin.
  • CDR complementarity determining region
  • the FGF antagonist is a fragment of the FGF molecule.
  • the FGF fragment competes with an FGF molecule for binding to the receptor.
  • the FGF antagonist is a small molecule, (e.g., is selected from a combinatorial library).
  • the FGF antagonist is chosen from those disclosed herein, e.g., suramin, structural analogs of suramin, anti-FGF antibodies, anti-FGF receptor antibodies, pentosan polysulfate, scopolamine, angiostatin, sprouty, estradiol, carboxymethylbenzylamme dextran (CMDB7), suradista, insulin-like growth factor binding protein-3, ethanol, heparin (e.g., 6-O-desulfated heparin), low molecular weight heparin, heparan sulfate, protamine sulfate, transforming growth factor beta, cyclosporin A, or RNA ligands for bFGF.
  • the FGF antagonist is heparin.
  • the FGF antagonist is low molecular weight heparin.
  • the FGF antagonist is heparan sulfate. In a preferred embodiment, the FGF antagonist is an anti-FGF antibody.
  • the FGF antagonist is suramin.
  • the FGF antagonist is suramin and the suramin is present in a concentration that is sufficient to block the resistance to anticancer agents induced by FGF (e.g., bFGF and/or aFGF), but is not sufficient to produce one or more of: (i) significant inhibition of cell proliferation; (ii) significant cell death in human and/or animal tumor cells, (iii) a measurable antitumor effect in a subject, e.g., a human subject, and/or (iv) cell cycle arrest.
  • the determination of effect on cultured cells can be determined with the system described in Example XV.
  • the FGF antagonist is suramin and it is administered at levels such that the plasma concentration of suramin that is present when the cytotoxic agent is present in plasma at pharmacologically active concentration does not result in one or more of: (i) significant cell cycle arrest, (ii) significant cell death, or (iii) significant inhibition of cell growth, e.g., the concentration in plasma is of a level that, if the same concentration of suramin is provided in cultured cells, at least 10, more preferably at least 25, more preferably at least 50, more preferably at least 70, more preferably at least 80, more preferably at least 90, more preferably at least 99, and most preferably at least 99% of the treated cultured cells continue to be involved in one or more of: (i) cycling cells continue to progress through the cell cycle, (ii) cells remain viable, or (iii) cells remain capable of proliferating, following treatment with suramin.
  • the determination of effect on cultured cells can be determined with the system described in Example XV.
  • suramin is administered in an amount that results in a concentration ranging from about 0.1 to 100 ⁇ g/ml, preferably about 1 to 85 ⁇ g/ml, more preferably, about 5 to 60 ⁇ g/ml, even more preferably, about 10 to 50 ⁇ g/ml, and most preferably, 15 to 45 ⁇ g/ml.
  • the pharmacokinetics of suramin is characterized by a triphasic concentration decline, with half-lives of 5.5 hours, 4.1 days and 78 days. The total body clearance is 0.0095 liter/hour/m 2 (Jodrell et al (1994) J Clin Oncol 12:166-175).
  • an initial dose of approximately 240 mg/m 2 should be administered to the average patient to achieve plasma concentrations declining from about 90 ⁇ g/ml (63 ⁇ M) to about 18 ⁇ g/ml (13 ⁇ M) over 96 hours.
  • the 96 hour duration is chosen as an example, as the plasma concentrations of many commonly used cytotoxic agents to be administered with suramin will have declined below their therapeutic levels at 96 hours.
  • Similar calculations can be performed to identify the initial suramin dose to deliver the preferred suramin plasma concentrations over other desired treatment durations. Maintenance doses to adjust the plasma concentrations for later treatment cycles can be similarly calculated.
  • the total suramin exposure is preferably less than
  • 800 ⁇ M-day over 96 hours preferably less than 600 ⁇ M-day over 96 hours, preferably less than 500 ⁇ M-day over 96 hours, preferably less than 400 ⁇ M-day over 96 hours, preferably less than 300 ⁇ M-day over 96 hours, preferably less than 252 ⁇ M-day over 96 hours, preferably less than 200 ⁇ M-day over 96 hours, preferably less than 150 ⁇ M- day over 96 hours, preferably less than 100 ⁇ M-day over 96 hours, and most preferably less than 52 ⁇ M-day over 96 hours.
  • the total suramin exposure is a product of the drug plasma concentration in ⁇ M-day (e.g., the average micromolarity over 24 hours) and the treatment duration in days. For example, treatment of a subject with 13 ⁇ M of suramin for four days would result in a total drug exposure of 52 ⁇ M-day over 96 hours.
  • suramin is administered in an amount that results in a plasma concentration of less than 100, preferably less than 80, preferably less than 50, preferably less than 25, more preferably less than 15, and most preferably less than 10 ⁇ M.
  • this plasma concentration is maintained for less than 20 days, preferably less than 15 days, preferably less than 12 days, preferably less than 10 days, more preferably less than 8 days, and most preferably less than 5 days beyond the time duration where therapeutic concentrations of a cytotoxic agent are maintained.
  • the FGF antagonist is suramin and the time period over which the suramin is administered or over which the suramin is maintained at the plasma concentration sufficient to inhibit or reverse the FGF-mediated resistance or to enhance the efficacy of the cytotoxic agent is less than 180, more preferably less than 90, more preferably less than 60, and most preferably less than 30 days.
  • suramin is given immediately or within 3, 2, or 1 day before the administration of the cytotoxic agent, ending immediately after plasma concentrations of the cytotoxic agent are below the therapeutic level.
  • the FGF antagonist is suramin and the time period over which the suramin is administered or over which the suramin is maintained at the plasma concentration sufficient to inhibit or reverse the FGF-mediated resistance or to enhance the efficacy of the cytotoxic agent does not begin substantially earlier or end substantial later than the period over which the cytotoxic agent is administered or over which the cytotoxic agent is maintained at a therapeutic level.
  • the FGF antagonist is suramin and the time period over which the suramin is administered or over which the suramin is maintained at the plasma concentration sufficient to inhibit or reverse the FGF-mediated resistance or to enhance the efficacy of the cytotoxic agent ends less than 180, more preferably less than 90, more preferably less than 60, and most preferably less than 30 days after the last day on which the cytotoxic agent is administered or the last day on which the cytotoxic agent is present at therapeutic levels.
  • the FGF antagonist is suramin and the time period over which the suramin is administered or over which the suramin is maintained at the plasma concentration sufficient to inhibit or reverse the FGF-mediated resistance or to enhance the efficacy of the cytotoxic agent begins less than 180, more preferably less than 90, more preferably less than 60, and most preferably less than 30 days before the first day on which the cytotoxic agent is administered or the first day on which the cytotoxic agent is present at therapeutic levels.
  • Methods described herein use suramin to enhance the antitumor effect of a cytotoxic agent (e.g., agents described in Table 2), where the suramin dose is selected to deliver a plasma concentration of below 100 ⁇ g/ml, preferably below 75 ⁇ g/ml, most preferably below 50 ⁇ g/ml, in a mammal while the cytotoxic agent is present in plasma at a pharmacologically active concentration.
  • the suramin dose is administered before, simultaneously with, or after the administration of the at least one anticancer agent.
  • mice with two weekly intravenous bolus suramin doses of 10 mg/kg for 3 weeks enhances the antitumor effect of anticancer drugs (e.g., paclitaxel, doxorubicin) but does not result in additional body weight loss.
  • This dose is calculated to result in a plasma suramin concentration of about 10 ⁇ M (-14 ⁇ g/ml) at 72 hours after dose administration (see Example IX, Tables 4 and 5, Figure 7).
  • the methods of the art use high dose suramin, either alone or in combination with a cytotoxic agent, where for a human subject, maintenance of plasma suramin concentrations of between 150 to 300 ⁇ g/ml is needed to produce a measurable antitumor effect (Eisenberger et al (1995) J Clin Oncol 13:2174-2186).
  • a typical suramin dosing schedule aimed at maintaining suramin plasma concentrations between 150 and 300 ⁇ g/ml consists of an initial administration of 2100 mg/m 2 over the first week with the subsequent doses repeated every 28 days for 6 months or longer; the subsequent doses are adjusted using the Bayesian pharmacokinetic method (Dawson et al (1998) Clin Cancer Res 4:37-44, Falcone et al (1999) Cancer 86:470- 476).
  • the methods of the art for using suramin in combination with other cytotoxic agents often administer suramin at a more frequent schedule or a longer duration than the frequency and the treatment duration for the other cytotoxic agents.
  • the duration of doxorubicin treatment was up to 20 weeks whereas the duration of the suramin treatment was up to 45 weeks (Tu et al (1998) Clin Cancer Res 4:1193-1201).
  • suramin was given weekly whereas mitomycin C was given only every 5 weeks (Rapoport et al (1993) Ann Oncol 4:567-573).
  • suramin causes the following toxicity in a human patient: adrenal insufficiency, coagulopathy, peripheral neuropathy, and proximal muscle weakness (Dorr and Von Hoff, Cancer Chemotherapy Handbook, 1994, pp 859-866).
  • the incidence and severity of these toxicities are positively related to cumulated dose and are minimized in the methods described herein.
  • the FGF antagonist is an anti-FGF antibody.
  • the anti-FGF antibody is present in a concentration that is sufficient to block the resistance to anticancer agents induced by FGF (e.g., bFGF and/or aFGF), but is not sufficient to produce one or more of: (i) significant inhibition of cell proliferation; (ii) significant cell death in human and/or animal tumor cells, (iii) a measurable antitumor effect in a subject, e.g., a human subject, and/or (iv) cell cycle arrest.
  • the FGF antagonist is an anti-FGF antibody and it is administered at levels such that the plasma concentration of the anti-FGF antibody that is present when the cytotoxic agent is present in plasma at pharmacologically active concentration does not result in one or more of: (i) significant cell cycle arrest, (ii) significant cell death, or (iii) significant inhibition of cell growth, e.g., the concentration in plasma is of a level that if the same concentration of the anti-FGF antibody is provided in cultured cells at least 10, more preferably at least 25, more preferably at least 50, more preferably at least 70, more preferably at least 80, more preferably at least 90, more preferably at least 99, and most preferably at least 99% of the treated cultured cells, continue to be involved in one or more of: (i) cycling cells continue to progress through the cell cycle, (ii) cells remain viable, or (iii) cells remain capable of proliferating, following treatment with the FGF antibody.
  • the determination of effect on cultured cells can be determined with the system described
  • the FGF antagonist is an anti-FGF antibody and the time period over which the anti-FGF antibody is administered or over which the anti- FGF antibody is maintained at the plasma concentration sufficient to inhibit or reverse the FGF-mediated resistance or to enhance the efficacy of the cytotoxic agent is less than 180, more preferably less than 90, more preferably less than 60, and most preferably less than 30 days.
  • the FGF antagonist is an anti-FGF antibody and the time period over which the anti-FGF antibody is administered or over which the anti- FGF antibody is maintained at the plasma concentration sufficient to inhibit or reverse the FGF-mediated resistance or to enhance the efficacy of the cytotoxic agent does not begin substantially earlier or end substantial later than the period over which the cytotoxic agent is administered or over which the cytotoxic agent is maintained at a therapeutic level.
  • the FGF antagonist is an anti-FGF antibody and the time period over which the anti-FGF antibody is administered or over which the anti- FGF antibody is maintained at the plasma concentration sufficient to inhibit or reverse the FGF-mediated resistance or to enhance the efficacy of the cytotoxic agent ends less than 180, more preferably less than 90, more preferably less than 60, and most preferably less than 30 days after the last day on which the cytotoxic agent is administered or the last day on which the cytotoxic agent is present at therapeutic levels.
  • the FGF antagonist is an anti-FGF antibody and the time period over which the anti-FGF antibody is administered or over which the anti- FGF antibody is maintained at the plasma concentration sufficient to inhibit or reverse the FGF-mediated resistance or to enhance the efficacy of the cytotoxic agent begins less than 180, more preferably less than 90, more preferably less than 60, and most preferably less than 30 days before the first day on which the cytotoxic agent is administered or the first day on which the cytotoxic agent is present at therapeutic levels.
  • the method inhibits the proliferation of, or enhances the killing of, a hyperproliferative cell selected from the group consisting of a solid tumor cell, a soft-tissue tumor cell, a metastatic tumor cell, a leukemic tumor cell, and a lymphoid tumor cell.
  • a hyperproliferative cell selected from the group consisting of a solid tumor cell, a soft-tissue tumor cell, a metastatic tumor cell, a leukemic tumor cell, and a lymphoid tumor cell.
  • the method inhibits the proliferation of, or enhances the killing of, a hyperproliferative cell in a fibrotic tumor.
  • the disorder is a cancer, e.g., a sarcoma, a carcinoma, an adenocarcinoma, a lymphoma, or a leukemia.
  • the disorder is a cancer which includes an established tumor.
  • the disorder is a cancer which includes a solid tumor.
  • the disorder is a cancer which includes a metastatic lesion.
  • the disorder is a cancer which includes a leukemia.
  • the disorder is a cancer which includes a lymphoma.
  • the disorder is a cancer, e.g., a fibrosarcoma, myosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, gastric cancer, esophageal cancer, colon carcinoma, rectal cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, uterine cancer, cancer of the head and neck, skin cancer, brain cancer, squamous cell carcinoma, sebaceous gland carcinoma, papillary carcinoma
  • the disorder includes a cancer which includes cells, e.g., tumor or metastatic cells, which form from a tissue where an FGF molecule is expressed, preferably at high levels, or cells that are in contact or exposed to aFGF, bFGF and/or FGF-producing cells or tissues.
  • a cancer which includes cells, e.g., tumor or metastatic cells, which form from a tissue where an FGF molecule is expressed, preferably at high levels, or cells that are in contact or exposed to aFGF, bFGF and/or FGF-producing cells or tissues.
  • the disorder includes a cancer which includes cells, e.g., metastatic cells, which form from a tissue of the breast, prostate, kidney, bladder, liver, lungs, lymph nodes, colon, rectum, skin, brain, pancreas, cervix, ovary, larynx, pharynx, oral mucosa, cancers of the head and neck, cancers of hematopoietic origin, or cancers of the lymphoid system.
  • cells e.g., metastatic cells, which form from a tissue of the breast, prostate, kidney, bladder, liver, lungs, lymph nodes, colon, rectum, skin, brain, pancreas, cervix, ovary, larynx, pharynx, oral mucosa, cancers of the head and neck, cancers of hematopoietic origin, or cancers of the lymphoid system.
  • the disorder is a cancer which includes an established tumor, e.g., a tumor that has been growing in a subject for at least one week, preferably two weeks, preferably one month, preferably two months, more preferably three months, more preferably six months, and most preferably longer than six months.
  • an established tumor e.g., a tumor that has been growing in a subject for at least one week, preferably two weeks, preferably one month, preferably two months, more preferably three months, more preferably six months, and most preferably longer than six months.
  • an established tumor can be diagnosed clinically.
  • an established tumor can be visualized on diagnostic media, e.g., an X-ray, a CAT scan, or MRI.
  • diagnostic media e.g., an X-ray, a CAT scan, or MRI.
  • an established tumor can be diagnosed by the detection of tumor markers, e.g., prostate specific antigen for prostate cancer, Her2/neu for certain breast cancers, CA125 for ovarian cancer, or genetic alterations.
  • an established tumor can be diagnosed by the detection of pathological changes, e.g., blood in sputum for lung cancer, blood in feces in colon cancer, blood in urine for bladder cancer, a lump in breasts for breast cancer, pain, or headache.
  • pathological changes e.g., blood in sputum for lung cancer, blood in feces in colon cancer, blood in urine for bladder cancer, a lump in breasts for breast cancer, pain, or headache.
  • an established tumor can be diagnosed by biochemical analysis of a patient sample, e.g., a patient's sputum, feces, urine, etc.
  • an established tumor can be diagnosed by morphological analysis of cells derived from a subject, e.g., morphological abnormalities, abnormal nucleus-to- cytoplasmic ratios, abnormal tissue architecture, abnormal tissue organization, or abnormal tissue composition.
  • an established tumor is one that is visible, palpable, or found during necropsy or autopsy.
  • the disorder includes a cancer, e.g., lung cancer, renal cancer, glioma, melanoma, or chemotherapy-refractory cancer, a metastatic cancer, which is chemoresistant, e.g., shows little or no significant response to chemotherapy.
  • a cancer e.g., lung cancer, renal cancer, glioma, melanoma, or chemotherapy-refractory cancer, a metastatic cancer, which is chemoresistant, e.g., shows little or no significant response to chemotherapy.
  • the hyperproliferative cell is found in a benign lesion.
  • the disorder is selected from the group consisting of psoriasis, cysts, benign prostatic hyperplasia, and endometriosis.
  • the disorder is selected from the group consisting of benign hyperplastic diseases, e.g., oral papillomas, central giant cell granulomas of the mouth or pharynx, benign cementoblastomas of the oral cavity, oral plakia, gastric polyps, gastric adenomas, small intestinal adenomas, small intestinal granulomas, small intestinal papillomas, small intestinal oncocytomas, small intestinal Schwannomas, colonic polyps, colonic adenomas, Crohn's disease, hepatic adenoma, hepatic cirrhosis, biliary papillomatosis, pancreatic adenomas, pancreatic ductal hyperplasia, renal oncocytomas, renal papillomas, adenomas of the group consisting of benign hyperplastic diseases,
  • the subject is a mammal, e.g., a human.
  • the subject is a patient, e.g., a cancer patient.
  • the subject can be a patient with non-small cell lung cancer, is treated with a combination of paclitaxel, carboplatin and an FGF antagonist, e.g., suramin, or with a combination of gemcitabine, cisplatin, and an FGF antagonist, e.g., suramin.
  • the subject can be a patient with hormone refractory prostate cancer, who is treated with a combination of estramustine phosphate, taxotere and an FGF antagonist, e.g., suramin, or with a combination of doxorubicin, ketoconazole and an FGF antagonist, e.g., suramin.
  • the subject can be a patient with metastatic breast cancer, who is treated with a combination of cyclophosphamide, doxorubicin, 5- fluorouracil and an FGF antagonist, e.g., suramin, or a combination of doxorubicin, taxotere and an FGF antagonist, e.g., suramin.
  • the subject can be a patient with advanced breast cancer that overexpresses the HER2/neu oncogene, who is treated with a HER2/neu inhibitor (e.g., a HER2/neu antibody) and an FGF antagonist, e.g., suramin, with or without paclitaxel or cisplatin.
  • a HER2/neu inhibitor e.g., a HER2/neu antibody
  • an FGF antagonist e.g., suramin
  • the subject can be a patient with advanced or metastatic colorectal cancer, who is treated with a combination of irinotecan and an FGF antagonist, e.g., suramin.
  • the subject can be a patient with advanced colon cancer, who is treated with a combination of 5-fluorouracil, leucovorin and an FGF antagonist, e.g., suramin.
  • the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an agent that promotes apoptosis and/or necrosis, an interferon, an interleukin, a tumor necrosis factor, and radiation.
  • a signal transduction pathway e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors
  • an agent that promotes apoptosis and/or necrosis e.g., an interferon, an interleukin, a tumor necros
  • the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interferon, an interleukin, a tumor necrosis factor, and radiation.
  • a signal transduction pathway e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors
  • an interferon e.g., an interleukin, a tumor necrosis factor, and radiation.
  • the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interferon, an interleukin, a tumor necrosis factor, and radiation.
  • a signal transduction pathway e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors
  • an interferon e.g., an interleukin, a tumor necrosis factor, and radiation.
  • the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interferon, an interleukin, a tumor necrosis factor, and radiation.
  • a signal transduction pathway e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors
  • an interferon e.g., an interleukin, a tumor necrosis factor, and radiation.
  • the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interferon, an interleukin, a tumor necrosis factor, and radiation.
  • a signal transduction pathway e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors
  • an interferon e.g., an interleukin, a tumor necrosis factor, and radiation.
  • the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interferon, an interleukin, and radiation.
  • a signal transduction pathway e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors
  • an interferon e.g., an interleukin, and radiation.
  • the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interleukin, a tumor necrosis factor, and radiation.
  • a signal transduction pathway e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors
  • an interleukin e.g., a tumor necrosis factor, and radiation.
  • the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a mitotic inhibitor, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interleukin, and radiation.
  • a signal transduction pathway e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors
  • an interleukin e.g., interleukin, and radiation.
  • the cytotoxic agent is selected from those disclosed below.
  • the cytotoxic agent is selected from those disclosed below.
  • the cytotoxic agent is selected from those disclosed below.
  • the cytotoxic agent is selected from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine
  • Ara-C trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N-Phosphoracetyl- L-Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza-2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5- fluoro-2'-deoxyuridine, 5-fluorouridine, 5'-deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphat
  • the cytotoxic agent is selected from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-Asparate (PALA)
  • the cytotoxic agent is selected from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-Asparate (PALA)
  • the cytotoxic agent is selected from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-Asparate (PALA)
  • the cytotoxic agent is selected from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), topotecan, irinotecan hydrochloride (e.g., Camptosar), etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N-Phosphoracetyl-L- Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza-2'-deoxy
  • the cytotoxic agent is selected from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere(e.g., Docetaxel), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N-Phosphoracetyl-L-Asparate (PALA), pentostatin,
  • paclitaxel paclitaxel, vincristine, vinblastine, vindesine, vinore
  • the cytotoxic agent is selected from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N-Phosphoracetyl-L-Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza-2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil
  • the cytotoxic agent is selected from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard
  • the cytotoxic agent is selected from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-Asparate (PALA)
  • the cytotoxic agent is selected from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-Asparate (PALA)
  • the cytotoxic agent is selected from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano-3-(4-fluorophenylsulphonyl)-2-hydroxy- 2-methyl-3 '-(trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interleukin 2, interleukin 4, interleukin 12, and radiation.
  • anti-Her2/neu antibody e.g., Herceptin
  • anti-CD20 e.g
  • the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, 5-fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
  • the cytotoxic agent is: paclitaxel, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, 5- fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
  • paclitaxel gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, 5- fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
  • Her2/neu antibody e.g
  • the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, taxotere, doxorubicin, epirubicin, 5- fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
  • the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, epirubicin, 5- fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
  • the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
  • the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, 5-fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
  • anti-Her2/neu antibody e.g., Herceptin
  • anti-CD20 anti-CD20
  • leuprolide e.g., Lupron
  • flutamide flutamide
  • the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, taxotere, doxorubicin, epirubicin, 5-fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
  • anti-Her2/neu antibody e.g., Herceptin
  • anti-CD20 anti-CD20
  • leuprolide e.g., Lupron
  • flutamide flutamide
  • the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
  • the cytotoxic agent is: paclitaxel, gemcitabine, fludarabine, irinotecan, taxotere, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
  • the cytotoxic agent is present in an amount equal to or lower than the one used in conventional chemotherapy.
  • the dose of paclitaxel is equal to or below 225 mg/m 2
  • the dose of carboplatin is chosen to achieve a total concentration-time product of equal to or below 6-7 mg/ml.min in previously untreated patients, or equal to or below 4-5 mg/ml.min in patients that have received chemotherapy previously; the treatment is repeated every 3 weeks.
  • estramustine phosphate and taxotere with an FGF antagonist
  • the daily oral dose of estramustine is equal to or below 1400 mg
  • the dose of taxotere is equal to or below 70 mg/m over 1 hour; the treatment is repeated every 3 weeks.
  • the weekly dose of doxorubicin is equal to or below 20 mg/m 2 by 24 hr infusion
  • the total daily oral dose of ketoconazole is equal to or below 1200 mg.
  • the dose of intravenous cyclophosphamide is equal to or below 500 mg/m 2
  • the dose of doxorubicin is equal to or below 50 mg/m 2
  • the dose of 5-fluorouracil is equal to or below 500 mg/m 2
  • the treatment is repeated every 4 weeks (the 5-fluorouracil dose is given once per week for two weeks whereas the doses of doxorubicin and cyclophosphamide are given once every 4 weeks).
  • the Herceptin dose is equal to or below 250 mg on day 0, followed by 9 weekly doses of equal to or below 100 mg, and the cisplatin dose is equal to or below 75 mg/m 2 on days 1, 29, and 57.
  • the four weekly doses of irinotecan are equal to or below 125 mg/m 2 ; the treatment cycle is 4 weeks on and 2 weeks off.
  • the dose of irinotecan is 350 mg/m 2 every 3 weeks.
  • the five daily intravenous bolus doses of 5-fluorouracil are equal to or below 425 mg/m 2
  • the five daily intravenous bolus doses of leucovorin are equal to or below 20 mg/m 2 ;
  • the treatment cycle is 1 week on and 4 weeks off.
  • the three weekly doses of gemcitabine are equal to or below 1000 mg/m 2
  • the single cisplatin dose given on day 2 is equal to or below 100 mg/m ; the treatment is repeated every 4 weeks.
  • the method further includes repeated dosages of the same, or a different cytotoxic agent.
  • the method further includes repeated dosages of the same, or a different FGF antagonist.
  • the cytotoxic agent and the FGF antagonist are administered at the same time or in overlapping time periods; the agent and the FGF antagonist are administered at different times; the agent is administered first and the FGF antagonist is administered subsequently; the FGF antagonist is administered first and the agent is administered subsequently.
  • the cytotoxic agent is administered systemically or locally.
  • the agent can be administered parenterally (e.g., subcutaneously, intravenously, intramuscularly, intraperitoneally, intradermally, intrathecally, etc.), intravesically (i.e., urinary bladder), intraprostatically, orally, nasally, rectally, topically, and/or transdermally.
  • the FGF antagonist is administered systemically or locally.
  • the FGF antagonist can be administered parenterally (e.g., subcutaneously, intravenously, intramuscularly, intraperitoneally, intradermally, intrathecally, etc.), intravesically (i.e., urinary bladder), intraprostatically, orally, nasally, rectally, topically, and/or transdermally.
  • the methods described herein further comprise monitoring the FGF levels (e.g., a-FGF and/or b-FGF) of a subject prior to or during treatment.
  • the amount of an FGF antagonist administered to the subject is determined based upon the levels of FGF present in the subject, e.g., low concentrations of FGF in the subject require decreased doses of the FGF antagonist to overcome the chemoresistance mediated by FGF.
  • the invention features, a method of treating a cell, e.g., inhibiting cell killing, or protecting the ability of a cell (e.g., a dividing cells, preferably, a rapidly dividing cell) to proliferate, e.g., a method of protecting a rapidly dividing cell, in a subject, from one or more of killing, inhibition of growth or division or other damage caused, e.g., by a cytotoxic agent (e.g., a cytostatic agent, e.g., an agent that causes cell death).
  • the method includes: administering, to the subject, an effective amount of at least one FGF agonist, thereby treating the cell, e.g., protecting or reducing the damage to the dividing cell from said cytotoxic agent.
  • the cell is: a cell from a body surface or cavity, e.g., a cell of the gastrointestinal or esophageal tract; a hair follicle cell; a hematopoietic cell, e.g., a hematopoietic stem cell.
  • the cell is part of the lining of the gastrointestinal tract or the esophageal tract.
  • the method inhibits hair loss; inhibits weight loss; inhibits the loss of gastrointestinal function; inhibits the loss of hematopoiesis.
  • the method further includes administering at least one cytotoxic agent (e.g., a cytostatic agent, e.g., an agent that causes cell death), e.g., an antiproliferative agent, e.g., an anticancer drug, e.g. radiation, e.g., an interferon, e.g., an interleukin, e.g., a tumor necrosis factor, to said subject.
  • cytotoxic agent e.g., a cytostatic agent, e.g., an agent that causes cell death
  • an antiproliferative agent e.g., an anticancer drug
  • radiation e.g., an interferon, e.g., an interleukin, e.g., a tumor necrosis factor
  • the cytotoxic agent is other than radiation, e.g., is an administered compound.
  • the cytotoxic agent is a compound other than an antimetabolite.
  • the FGF agonist is administered orally, locally, e.g., topically, or ex vivo.
  • the FGF agonist can be applied topically to treat hair follicles.
  • the cell e.g., a bone marrow cell, can be treated ex vivo.
  • the FGF agonist is administered locally to a site, e.g., gastrointestinal tract, bone marrow, or the skin.
  • a site e.g., gastrointestinal tract, bone marrow, or the skin.
  • the FGF agonist is directly injected into the bone marrow.
  • the FGF agonist is applied topically, e.g. externally to the scalp.
  • the application of the FGF agonist does not significantly result in absorption into systemic circulation, and/or does not significantly result in a blood or plasma concentration that is sufficient to increase the proliferation of a tumor cell in the subject.
  • the FGF agonist is administered to a subject by a method that does not result in significant systemic administration or significant systemic levels of the FGF agonist, e.g. does not result in an increase in FGF agonist blood levels that promotes the chemoresistance of the tumor.
  • Systemic administration or levels of the FGF agonist can be evaluated by determining the level of the FGF agonist in the blood before and after its administration to a subject. These two levels can then be tested in vitro and compared for their effect on the antitumor effect mediated by a cytotoxic agent against tumor cells.
  • the FGF agonist blood level after administration should increase the IC o of the cytotoxic agent by less than 30, preferably less than 20, preferably less than 10, preferably less than 5, and most preferably less than 1%, as compared to the IC 5 o of the cytotoxic agent in the presence of the blood levels of the FGF agonist prior to administration of the FGF agonist to the subject.
  • the determination of effect on cultured cells can be determined with the system described in Example XV.
  • the subject is a mammal, e.g., a human.
  • the subject is a patient, e.g., a cancer patient.
  • the subject is a patient with non-small cell lung cancer, who is treated with a combination of two or more of: paclitaxel, carboplatin, or an FGF antagonist, e.g., suramin, or with a combination of two or more of: gemcitabine, cisplatin, or an FGF antagonist, e.g., suramin.
  • the patient is a patient with hormone refractory prostate cancer, who is treated with a combination of two or more of: estramustine phosphate, taxotere, or an FGF antagonist, e.g., suramin, or with a combination of two or more of: doxorubicin, ketoconazole, an FGF antagonist, e.g., suramin.
  • the patient is a patient with metastatic breast cancer, who is treated with a combination of two or more of: cyclophosphamide, doxorubicin, 5-fluorouracil, or an FGF antagonist, e.g., suramin.
  • the patient is a patient with advanced breast cancer that overexpresses the HER2/neu oncogene, who is treated with Herceptin and suramin, with or without paclitaxel or cisplatin.
  • the patient is a patient with advanced or metastatic colorectal cancer, who is treated with one or more of: irinotecan or an FGF antagonist, e.g., suramin.
  • the patient is a patient with advanced colon cancer, who is treated with a combination of two or more of: 5-fluorouracil, leucovorin, or an FGF antagonist, e.g., suramin.
  • the treatment protects said cell from an effect, e.g., a cytotoxic effect (e.g., cytostasis, e.g., cell kill, e.g., hair loss), of said cytotoxic treatment.
  • a cytotoxic effect e.g., cytostasis, e.g., cell kill, e.g., hair loss
  • the FGF agonist comprises bFGF, aFGF, or bFGF and aFGF, or a fragment or an analog thereof.
  • bFGF and aFGF may have an additive effect, preferably, a synergistic effect.
  • the FGF agonist is administered in an amount that results in concentrations sufficient to protect said cell from an effect, e.g., a cytotoxic effect (e.g., cytostasis, e.g., cell kill, e.g., hair loss), of said cytotoxic treatment.
  • a cytotoxic effect e.g., cytostasis, e.g., cell kill, e.g., hair loss
  • Administration of the FGF agonist may be repeated to provide protection throughout the duration of said cytotoxic treatment and/or throughout the duration when the plasma concentrations of the cytotoxic agent remain sufficient to produce a cytotoxic effect.
  • the time period over which the FGF agonist is administered or over which the FGF agonist is maintained at a therapeutic level is less than 180, more preferably less than 90, more preferably less than 60, and most preferably less than 30 days.
  • the time period over which the FGF agonist is administered or over which the FGF agonist is maintained at a therapeutic level does not begin substantially earlier or end substantial later than the period over which the cytotoxic agent is administered or over which the cytotoxic agent is maintained at a therapeutic level.
  • the time period over which the FGF agonist is administered or over which the FGF agonist is maintained at a therapeutic level ends less than 180, more preferably less than 90, more preferably less than 60, and most preferably less than 30 days after the last day on which the cytotoxic agent is administered or the last day on which the cytotoxic agent is present at therapeutic levels.
  • the time period over which the FGF agonist is administered or over which the FGF agonist is maintained at a therapeutic level begins less than 180, more preferably less than 90, more preferably less than 60, and most preferably less than 30 days before the first day on which the cytotoxic agent is administered or the first day on which the cytotoxic agent is present at therapeutic levels.
  • the FGF agonist is a peptide, or a small molecule.
  • the FGF agonist is nonproteinaceous.
  • the cytotoxic agent used in the methods described herein is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti- hormone, e.g., an antibody against growth factor receptors), an agent that promotes apoptosis and/or necrosis, an interferon, an interleukin, a tumor necrosis factor, and radiation.
  • a signal transduction pathway e.g., a protein kinase C inhibitor, e.g., an anti- hormone, e.g., an antibody against growth factor receptors
  • an agent that promotes apoptosis and/or necrosis e.g., an anti- hormone, e.g.,
  • the cytotoxic agent used in the methods described herein is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interferon, an interleukin, a tumor necrosis factor, and radiation.
  • a signal transduction pathway e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors
  • an interferon e.g., an interleukin, a tumor necrosis factor, and radiation.
  • the cytotoxic agent used in the methods described herein is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti- hormone, e.g., an antibody against growth factor receptors, an interferon, an interleukin, and a tumor necrosis factor.
  • a signal transduction pathway e.g., a protein kinase C inhibitor, e.g., an anti- hormone, e.g., an antibody against growth factor receptors, an interferon, an interleukin, and a tumor necrosis factor.
  • the cytotoxic agent used in the methods described herein is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interferon, an interleukin, a tumor necrosis factor.
  • the cytotoxic agent used in the methods described herein is chosen from those disclosed below.
  • cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), topotecan, camptothecin, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N-Phosphoracetyl-L-Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza-2'-deoxycytidine, adenosine,
  • cytotoxic agent used in the methods described herein, is chosen from those disclosed below.
  • the cytotoxic agent used in the methods described herein is chosen from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), topotecan, camptothecin, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N-Phosphoracetyl-L- Asparate (PALA), 5-fluorouracil, met
  • the cytotoxic agent used in the methods described herein is chosen from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), topotecan, camptothecin, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphat
  • taxotere e.
  • the cytotoxic agent used in the methods described herein is chosen from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), topotecan, camptothecin, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphat
  • taxotere e.
  • the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, 5-fluororuacil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (Lupron) and flutamide.
  • the invention features, a pharmaceutical composition which includes at least one FGF antagonist, at least one cytotoxic agent (e.g., a cytostatic agent, e.g., an agent that causes cell death), and a pharmaceutically acceptable carrier, wherein said FGF antagonist is present in an amount effective to enhance the efficacy of the cytotoxic agent, in reducing or inhibiting the proliferation, or in enhancing the killing, of a hyperproliferative cell.
  • cytotoxic agent e.g., a cytostatic agent, e.g., an agent that causes cell death
  • a pharmaceutically acceptable carrier wherein said FGF antagonist is present in an amount effective to enhance the efficacy of the cytotoxic agent, in reducing or inhibiting the proliferation, or in enhancing the killing, of a hyperproliferative cell.
  • the pharmaceutical composition is packaged with directions to practice the methods described herein.
  • the pharmaceutical composition includes: an inhibitor of bFGF; an inhibitor of aFGF; or a bFGF inhibitor and an aFGF inhibitor.
  • the FGF antagonist inhibits or reverses the resistance to anticancer drugs induced by FGF (e.g. aFGF and/or bFGF) in cultured tumor cells under in vitro conditions.
  • FGF e.g. aFGF and/or bFGF
  • the determination of effect on cultured cells can be determined with the system described in Example XV.
  • the FGF antagonist inhibits or reduces the resistance of tumor cells to a broad spectrum of cytotoxic agents.
  • the FGF antagonist is capable of binding to an FGF molecule or an FGF receptor; blocks the binding of FGF to a receptor; blocks the interaction of FGF to molecules that facilitate the binding of FGF to a receptor; down regulates FGF receptor action; is a protein or a peptide; is an antibody, e.g., a monoclonal, diabody, a murine antibody, a human antibody, a humanized or a chimeric antibody, or an antigen-binding fragment thereof, e.g., an Fab, F(ab')2, Fv or a single chain Fv fragment; is a truncated FGF molecule, or a fragment thereof.
  • the FGF antagonist acts extracellularly, e.g., inhibits the binding of an FGF molecule to the extracellular domain of the FGF receptors.
  • the FGF antagonist acts intracellularly, e.g., inhibits the binding of an FGF molecule to the intracellular domain of the FGF receptors.
  • the FGF antagonist acts intracellularly, e.g., inhibits the intracellular effects of FGF.
  • the FGF antagonist acts extracellularly, e.g., inhibits the binding of an FGF molecule to its receptor.
  • the FGF antagonist is chosen from those disclosed herein, e.g., suramin, structural analogs of suramin, anti-FGF antibodies, anti-FGF receptor antibodies, pentosan polysulfate, scopolamine, angiostatin, sprouty, estradiol, carboxymethylbenzylamine dextran (CMDB7), suradista, insulin-like growth factor binding protein-3, ethanol, heparin (e.g., 6-O-desulfated heparin), low molecular weight heparin, heparan sulfate, protamine sulfate, transforming growth factor beta, cyclosporin A, or RNA ligands for bFGF.
  • CMDB7 carboxymethylbenzylamine dextran
  • the FGF antagonist is suramin.
  • the FGF antagonist is a fragment of the FGF molecule which competes with an FGF molecule for binding to the receptor; the FGF antagonist is a small molecule.
  • the FGF antagonist is a small molecule, is chosen from a combinatorial library.
  • the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an agent that promotes apoptosis and/or necrosis, an interferon, an interleukin, a tumor necrosis factor, and radiation.
  • a signal transduction pathway e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors
  • the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interferon, an interleukin, a tumor necrosis factor, and radiation.
  • a signal transduction pathway e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors
  • an interferon e.g., an interleukin, a tumor necrosis factor, and radiation.
  • the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interferon, an interleukin, a tumor necrosis factor, and radiation.
  • a signal transduction pathway e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors
  • an interferon e.g., an interleukin, a tumor necrosis factor, and radiation.
  • the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interferon, an interleukin, a tumor necrosis factor, and radiation.
  • a signal transduction pathway e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors
  • an interferon e.g., an interleukin, a tumor necrosis factor, and radiation.
  • the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interferon, an interleukin, a tumor necrosis factor, and radiation.
  • a signal transduction pathway e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors
  • an interferon e.g., an interleukin, a tumor necrosis factor, and radiation.
  • the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interferon, an interleukin, and radiation.
  • a signal transduction pathway e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors
  • an interferon e.g., an interleukin, and radiation.
  • the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interleukin, a tumor necrosis factor, and radiation.
  • a signal transduction pathway e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors
  • an interleukin e.g., a tumor necrosis factor, and radiation.
  • the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a mitotic inhibitor, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interleukin, and radiation.
  • a signal transduction pathway e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors
  • an interleukin e.g., interleukin, and radiation.
  • the cytotoxic agent is selected from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L- As
  • the cytotoxic agent is selected from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-Asparate (PALA),
  • paclitaxel
  • the cytotoxic agent is selected from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-Asparate (PALA), pen
  • the cytotoxic agent is selected from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N-Phosphoracetyl- L- Asparate (PALA), pen
  • the cytotoxic agent is selected from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-A
  • the cytotoxic agent is selected from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L- As
  • the cytotoxic agent is selected from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-A
  • the cytotoxic agent is selected from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), topotecan, irinotecan hydrochloride (e.g., Camptosar), etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N-Phosphoracetyl-L- Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza
  • the cytotoxic agent is selected from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N-Phosphoracetyl-L- Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza-2'-deoxycytidine, a
  • the cytotoxic agent is selected from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N-Phosphoracetyl-L- Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza-2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination
  • the cytotoxic agent is selected from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-A
  • the cytotoxic agent is selected from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-A
  • the cytotoxic agent is selected from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-A
  • the cytotoxic agent is selected from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano-3-(4-fluorophenylsulphonyl)-2-hydroxy- 2-methyl-3'-(trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti- CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, and radiation.
  • cytotoxic agents include:
  • the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, 5-fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
  • the cytotoxic agent is: paclitaxel, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, 5- fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
  • paclitaxel gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, 5- fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
  • Her2/neu antibody e.g
  • the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, taxotere, doxorubicin, epirubicin, 5- fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
  • the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, epirubicin, 5- fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
  • anti-Her2/neu antibody e.g., Herceptin
  • anti-CD20 anti-CD20
  • leuprolide e.g., Lupron
  • flutamide flutamide
  • the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
  • the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, 5-fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
  • anti-Her2/neu antibody e.g., Herceptin
  • anti-CD20 anti-CD20
  • leuprolide e.g., Lupron
  • flutamide flutamide
  • the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, taxotere, doxorubicin, epirubicin, 5-fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
  • anti-Her2/neu antibody e.g., Herceptin
  • anti-CD20 anti-CD20
  • leuprolide e.g., Lupron
  • flutamide flutamide
  • the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
  • anti-Her2/neu antibody e.g., Herceptin
  • anti-CD20 anti-CD20
  • leuprolide e.g., Lupron
  • flutamide flutamide
  • the cytotoxic agent is: paclitaxel, gemcitabine, fludarabine, irinotecan, taxotere, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
  • the cytotoxic agent is present at an amount equal to or lower than the one used in conventional chemotherapy.
  • the hyperproliferative cell is a cancer cell.
  • the hyperproliferative cell is found in a benign lesion.
  • the disorder is selected from the group consisting of psoriasis, cysts, benign prostatic hyperplasia, and endometriosis.
  • the disorder is selected from the group consisting of benign hyperplastic diseases, e.g., oral papillomas, central giant cell granulomas of the mouth or pharynx, benign cementoblastomas of the oral cavity, oral plakia, gastric polyps, gastric adenomas, small intestinal adenomas, small intestinal granulomas, small intestinal papillomas, small intestinal oncocytomas, small intestinal Schwannomas, colonic polyps, colonic adenomas, Crohn's disease, hepatic adenoma, hepatic ci ⁇ hosis, biliary papillomatosis, pancreatic adenomas, pancreatic ductal hyperplasia, renal oncocytomas, renal papillomas, adenomas of the bladder, malakoplakia of the bladder, pseudosarcomas of the bladder, endometriosis, benign prostatic hyperplasia,
  • benign hyperplastic diseases e.g
  • the invention features, a pharmaceutical composition which includes at least one FGF agonist, and a pharmaceutically acceptable carrier, wherein said FGF agonist is present in an amount effective to protect, or reduce the damage, a cell, e.g., a rapidly dividing cell, from said cytotoxic agent.
  • the pharmaceutical composition is packaged with directions to practice the methods described herein.
  • the FGF agonist comprises bFGF; aFGF; or bFGF and aFGF, or a fragment or an analog thereof.
  • the FGF agonist comprises bFGF, or a fragment or an analog thereof.
  • the FGF agonist is a peptide, or a small molecule.
  • the FGF agonist is nonproteinaceous.
  • the cell is: a cell from a body surface or cavity, e.g., a cell of the gastrointestinal or esophageal tract; a hair follicle cell; a hematopoietic cell, e.g., a hematopoietic stem cell.
  • the cell is a part of the lining of the gastrointestinal tract or the esophageal tract.
  • kits for carrying out the combined administration of the FGF antagonist/agonist with other cytotoxic agents comprises an FGF antagonist/agonist formulated in a pharmaceutical carrier, and at least one cytotoxic agent, formulated, as appropriate, in one or more separate pharmaceutical preparations.
  • the kit includes directions to practice the methods described herein.
  • the invention features a vaccine comprising an FGF polypeptide, or a FGF receptor-derived polypeptide, or a fragment of a proteoglycan that facilitates the binding of FGF to its receptors, and a pharmaceutically acceptable carrier, in an amount effective to immunize a subject against a neoplastic disease.
  • the immunization against the neoplastic disease can be partial or complete.
  • Said vaccine can be used to treat a cancer patient, or to prevent one or more of: the occu ⁇ ence of, recidivism of, and/or metastasis of, the neoplastic disease. For example, it could be administered to a patient who has had a tumor surgically removed, to prevent recurrence of the tumor.
  • the subject is a mammal, e.g., a human.
  • the subject is not a cancer patient.
  • the subject is a pateint that is afflicted with an ailment caused by a benign hyperplasia of a normal tissue.
  • the subject is a patient, e.g., a cancer patient.
  • the subject can be a patient in remission, or a cancer patient undergoing treatment (e.g., conventional chemotherapy, alone or in combination, with the methods described herein).
  • the subject can be a patient with non-small cell lung cancer, treated with a combination of two or more of: paclitaxel, carboplatin or an FGF antagonist, e.g., suramin, or with a combination of two or more of: gemcitabine, cisplatin, or an FGF antagonist, e.g., suramin.
  • the subject can be a patient with hormone refractory prostate cancer, who is treated with a combination of two or more of: estramustine phosphate, taxotere, or an FGF antagonist, e.g., suramin, or with a combination of two or more of: doxorubicin, ketoconazole, or an FGF antagonist, e.g., suramin.
  • the subject can be a patient with metastatic breast cancer, who is treated with a combination of two or more of: cyclophosphamide, doxorubicin, 5-fluorouracil, or an FGF antagonist, e.g., suramin, or a combination of two or more of: doxorubicin, taxotere, or an FGF antagonist, e.g., suramin.
  • the subject can be a patient with advanced breast cancer that overexpresses the HER2/neu oncogene, who is treated with a HER2/neu inhibitor (e.g., a HER2/neu antibody) and/or an FGF antagonist, e.g., suramin, with or without paclitaxel or cisplatin.
  • the subject can be a patient with advanced or metastatic colorectal cancer, who is treated with one or more of: irinotecan or an FGF antagonist, e.g., suramin.
  • the subject can be a patient with advanced colon cancer, who is treated with a combination of two or more of: 5- fluorouracil, leucovorin, or an FGF antagonist, e.g., suramin.
  • the subject can be a patient that is in remission but is with a cancer that has a good likelihood to recur, e.g., adult leukemia, e.g., early disseminated prostate cancer.
  • the invention features, a method for evaluating the effectiveness of a compound, e.g., for treating a disorder, e.g., a proliferative disorder, e.g., a malignant disorder, or for protecting cells, e.g., from a cytotoxic agent.
  • the method includes: contacting the compound with an FGF, e.g., aFGF or bFGF; and evaluating the ability of the compound to inhibit or promote FGF activity, inhibition being co ⁇ elated with effectiveness of treating a disorder, and promotion being co ⁇ elated with cell protection.
  • an FGF e.g., aFGF or bFGF
  • the method further comprises testing the compound to determine if it can modulate, e.g., increase, the efficacy of an agent which kills cells or inhibits cell growth, e.g., an anticancer agent. This can be done by administering the compound and the agent, together or separately, to a test cell or organism.
  • the method further comprises testing the compound to determine if it can modulate, e.g., increase, the efficacy of an agent which kills cells or inhibits cell growth, e.g., an anticancer agent, in the presence of FGF (e.g., bFGF, and or aFGF).
  • FGF e.g., bFGF, and or aFGF
  • the method further comprises testing the compound to determine if it can protect cells from an agent that kills cells or inhibits cell growth, e.g., a cytotoxic or an anticancer agent. This can be done by administering both the compound and the agent, together or separately, to a test cell or organism.
  • the method includes: providing a cell, e.g., a cultured cell, a transformed cell, a cell from a cancer, or a test organism; administering said compound to said cell (or test organism) and evaluating FGF activity, cell proliferation, cell death, or tumor growth, e.g., metastatic tumor growth.
  • the disorder is a cancer which includes a sarcoma, a carcinoma, an adenocarcinoma, a lymphoma, or a leukemia.
  • the disorder is a cancer which includes a solid tumor.
  • the disorder is a cancer which includes a leukemia. In a prefe ⁇ ed embodiment, the disorder is a cancer which includes a lymphoma. In a prefe ⁇ ed embodiment, the disorder is a cancer which includes a metastatic lesion.
  • the disorder includes a cancer which includes cells, e.g., metastatic cells, which form from a tissue where an FGF molecule is expressed, or cells that are in contact or exposed to aFGF, bFGF and/or FGF-producing cells or tissues.
  • cells e.g., metastatic cells, which form from a tissue where an FGF molecule is expressed, or cells that are in contact or exposed to aFGF, bFGF and/or FGF-producing cells or tissues.
  • the disorder includes a cancer which includes cells, e.g., metastatic cells, which form from a tissue of the breast, prostate, kidney, bladder, liver, lungs, lymph nodes, colon, rectum, skin, brain, pancreas, cervix, ovary, larynx, pharynx, oral mucosa, cancers of the head and neck, cancers of hematopoietic origin, or cancers of the lymphoid system.
  • cells e.g., metastatic cells, which form from a tissue of the breast, prostate, kidney, bladder, liver, lungs, lymph nodes, colon, rectum, skin, brain, pancreas, cervix, ovary, larynx, pharynx, oral mucosa, cancers of the head and neck, cancers of hematopoietic origin, or cancers of the lymphoid system.
  • the disorder is a cancer, e.g., a fibrosarcoma, myosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, gastric cancer, esophageal cancer, colon carcinoma, rectal cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, uterine cancer, cancer of the head and neck, skin cancer, brain cancer, squamous cell carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinoma, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma
  • a cancer e.g.
  • the disorder is selected from the group consisting of benign hyperplastic diseases, e.g., oral papillomas, central giant cell granulomas of the mouth or pharynx, benign cementoblastomas of the oral cavity, oral plakia, gastric polyps, gastric adenomas, small intestinal adenomas, small intestinal granulomas, small intestinal papillomas, small intestinal oncocytomas, small intestinal Schwannomas, colonic polyps, colonic adenomas, Crohn's disease, hepatic adenoma, hepatic ci ⁇ hosis, biliary papillomatosis, pancreatic adenomas, pancreatic ductal hyperplasia, renal oncocytomas, renal papillomas, adenomas of the bladder, malakoplakia of the bladder, pseudosarcomas of the bladder, endometriosis, benign prostatic hyperplasia,
  • benign hyperplastic diseases e.g
  • the compound is a chemical, e.g., a small molecule (e.g., a member of a combinatorial library).
  • the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an agent that promotes apoptosis and/or necrosis, an interferon, an interleukin, a tumor necrosis factor, or radiation.
  • a signal transduction pathway e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors
  • an agent that promotes apoptosis and/or necrosis e.g., an interferon, an interleukin, a tumor necros
  • the cytotoxic agent is chosen from those disclosed below.
  • Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), topotecan, camptothecin, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-A
  • the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, 5-fluorouracil, UFT, tamoxifen, goserelin, a HER2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (Lupron) and flutamide.
  • a HER2/neu antibody e.g., Herceptin
  • anti-CD20 e.g., leuprolide (Lupron) and flutamide.
  • the cell is a rapidly dividing cell, a gastrointestinal cell, or a hair follicle cell, or a hematopoietic cell, e.g., a hematopoietic stem cell.
  • the invention features, a method of analyzing a sample, e.g., for the level of tumor resistance to anticancer agents.
  • the method includes: evaluating the level of one or more FGF genes or gene products, wherein an increase or decrease in the level of one or more FGF genes or gene products, relative to a control, indicates the presence of tumor resistance to anticancer agents.
  • the sample e.g. a sample form a subject, is a sample of tissue having unwanted proliferation, e.g., a sample of a benign hype ⁇ lasic tissue, a sample from a primary tumor, a metastatic tumor, or a leukemia.
  • the expression of FGF genes or gene products is selected from the group consisting of bFGF, aFGF, TSC22, VEGF, GAFA1, GAFA2, GAFA3 (originally designated as FSC1, FSC2, FSC3 in the U.S. Provisional
  • the method is performed: on a sample from a mammal, a sample from a human subject; and a sample from a cancer patient; as part of therapeutic decision making; to determine if the individual from which the target FGF is taken should receive a drug or other treatment; to diagnose an individual for a disorder or for predisposition to resistance to treatment, to stage a disease or disorder.
  • the method further includes choosing a therapeutic modality, e.g., a particular anticancer treatment, e.g. a particular FGF antagonist, or a dosage thereof, based on the level of FGF expression.
  • nucleic acid (or protein) from the cell or sample is analyzed on positional a ⁇ ays, e.g., DNA-chip a ⁇ ays.
  • the method further includes: analyzing the sample by providing an a ⁇ ay of a plurality of capture probes, wherein each of the capture probes is positionally distinguishable from other capture probes of the plurality on the a ⁇ ay, and wherein each positional distinguishable capture probe includes a unique reagent e.g., an antibody or a nucleic acid probe which can identify an FGF gene or gene product; hybridizing the sample with the a ⁇ ay of capture probes, thereby analyzing the sample sequence.
  • a unique reagent e.g., an antibody or a nucleic acid probe which can identify an FGF gene or gene product
  • the method includes a method of staging a disorder, e.g., a proliferative disorder, e.g., a benign hype ⁇ lastic disorder, e.g., a malignant disorder, in a subject.
  • the method includes: providing a sample, e.g., cancerous sample, e.g.
  • a tissue e.g., urine, blood, or CSF, a biopsy, from said subject; evaluating the expression of one or more FGF genes, e.g., by contacting said cancerous sample with, a nucleic acid probe that selectively hybridizes to one or more FGF gene products; wherein an increase in the level of said one or more FGF genes or gene products, relative to a control, indicates a stage in the disorder, e.g., the malignant disorder.
  • a tissue e.g., a bodily fluid, e.g., urine, blood, or CSF, a biopsy
  • evaluating the expression of one or more FGF genes e.g., by contacting said cancerous sample with, a nucleic acid probe that selectively hybridizes to one or more FGF gene products; wherein an increase in the level of said one or more FGF genes or gene products, relative to a control, indicates a stage in the disorder, e.g., the malignant disorder.
  • the FGF genes or gene products are selected from the group consisting of bFGF, aFGF, TSC22, VEGF, GAFA1, GAFA2, GAFA3 (originally designated as FSC1, FSC2, FSC3 in the U.S. Provisional Application No. 60/137,345), and TFII-I.
  • the method is performed: on a sample from a mammal, a sample from a human subject; e.g., a sample of a patient suffering from a benign hype ⁇ lastic disorder, e.g., a sample from a cancer patient; to determine if the individual from which the target nucleic acid or protein is taken should receive a drug or other treatment; to diagnose an individual for a disorder or for predisposition to resistance to treatment, to stage a disease or disorder.
  • the method further includes choosing a therapeutic modality, e.g., a particular anticancer treatment, or a dosage thereof, based on the level of FGF expression.
  • the expression of an FGF gene is evaluated by evaluating the expression of a signal entity, e.g., a green fluorescent protein or other marker protein, which is under the control or an FGF gene control element e.g., promoter.
  • a signal entity e.g., a green fluorescent protein or other marker protein
  • nucleic acid (or protein) from the cell or sample is analyzed on positional a ⁇ ays, e.g., DNA-chip arrays.
  • the method further includes: analyzing the sample by providing an a ⁇ ay of a plurality of capture probes, wherein each of the capture probes is positionally distinguishable from other capture probes of the plurality on the a ⁇ ay, and wherein each positional distinguishable capture probe includes a unique reagent, e.g., an antibody or a nucleic acid probe which can identify an FGF gene or gene product; hybridizing the sample with the a ⁇ ay of capture probes, thereby analyzing the sample sequence.
  • a unique reagent e.g., an antibody or a nucleic acid probe which can identify an FGF gene or gene product
  • the invention features, a method of diagnosing a disorder, e.g., a proliferative disorder, e.g., a malignant disorder, e.g., a benign hype ⁇ roliferative disorder, in a subject.
  • a proliferative disorder e.g., a malignant disorder, e.g., a benign hype ⁇ roliferative disorder
  • the method includes: providing a sample e.g., a cancerous sample, e.g., a tissue, a bodily fluid (e.g., blood, urine, sputum, CSF), a biopsy, from said patient; evaluating the expression of one or more FGF genes, e.g., by contacting said sample with, a nucleic acid probe that selectively hybridizes to one or more FGF genes, or an antibody that specifically binds to one or more FGF gene products; wherein an increase or decrease in the level of said one or more FGF genes or gene products, relative to a control, indicates the presence or absence of the disorder, e.g., the malignant disorder.
  • a sample e.g., a cancerous sample, e.g., a tissue, a bodily fluid (e.g., blood, urine, sputum, CSF), a biopsy
  • evaluating the expression of one or more FGF genes e.g., by contacting said sample with, a nucle
  • the FGF related genes or gene product is selected from the group consisting of bFGF, aFGF, TSC22, VEGF, GAFA1, GAFA2, GAFA3 (originally designated as FSC1, FSC2, FSC3 in the U.S. Provisional Application No. 60/137,345), and TFII-I.
  • the expression of an FGF gene is evaluated by evaluating the expression of a signal entity, e.g., a green fluorescent protein or other marker protein, which is under the control of or an FGF gene control element, e.g., promoter.
  • a signal entity e.g., a green fluorescent protein or other marker protein
  • an FGF gene control element e.g., promoter.
  • nucleic acid (or protein) from the cell or sample is analyzed on positional a ⁇ ays, e.g., DNA-chip a ⁇ ays.
  • the method further includes: analyzing the sample by providing an a ⁇ ay of a plurality of capture probes, wherein each of the capture probes is positionally distinguishable from other capture probes of the plurality on the a ⁇ ay, and wherein each positional distinguishable capture probe includes a unique reagent, e.g., an antibody or a nucleic acid probe which can identify an FGF gene or gene product; hybridizing the sample with the a ⁇ ay of capture probes, thereby analyzing the sample sequence.
  • a unique reagent e.g., an antibody or a nucleic acid probe which can identify an FGF gene or gene product
  • the invention features, a method for evaluating the efficacy of a treatment of a disorder, e.g., a proliferative disorder, e.g., a malignant disorder, in a patient, comprising: providing a sample, e.g., a cancerous sample, e.g., a tissue, a bodily fluid (e.g., blood, urine, sputum, CSF), a biopsy, from said patient; evaluating the expression of one or more FGF genes, e.g., by contacting said cancerous sample with, a nucleic acid probe that selectively hybridizes to one or more FGF genes, or an antibody that specifically binds to one or more FGF gene products; wherein a change, e.g., decrease, e.g., increase, in the level of said one or more FGF genes or gene products in a sample obtained after treatment, relative to the level of expression before treatment, is indicative of the efficacy of the treatment of said disorder.
  • a change e
  • the method further includes choosing a therapeutic modality, e.g., a particular anticancer treatment, or a dosage thereof, based on the level of FGF expression.
  • a therapeutic modality e.g., a particular anticancer treatment, or a dosage thereof
  • the FGF genes or gene product is selected from the group consisting of bFGF, aFGF, TSC22, VEGF, GAFA1, GAFA2, GAFA3 (originally designated as FSC1, FSC2, FSC3 in the U.S. Provisional Application No. 60/137,345), and TFII-I.
  • the expression of an FGF gene is evaluated by evaluating the expression of a signal entity, e.g., a green fluorescent protein or other marker protein, which is under the control or an FGF gene control element, e.g., promoter.
  • a signal entity e.g., a green fluorescent protein or other marker protein
  • an FGF gene control element e.g., promoter.
  • the nucleic acid (or protein) from the cell or sample is analyzed on positional a ⁇ ays, e.g., DNA-chip a ⁇ ays.
  • the method further includes: analyzing the sample by providing an a ⁇ ay of a plurality of capture probes, wherein each of the capture probes is positionally distinguishable from other capture probes of the plurality on the a ⁇ ay, and wherein each positional distinguishable capture probe includes a unique reagent, e.g., an antibody or a nucleic acid probe which can identify an FGF gene or gene product; hybridizing the sample with the a ⁇ ay of capture probes, thereby analyzing the sample sequence.
  • a unique reagent e.g., an antibody or a nucleic acid probe which can identify an FGF gene or gene product
  • the invention features, a method for evaluating the effectiveness of a treatment, e.g., the administration of a compound, for treating a disorder, e.g., a proliferative disorder, e.g., a malignant disorder.
  • a treatment e.g., the administration of a compound
  • a disorder e.g., a proliferative disorder, e.g., a malignant disorder.
  • the method includes: providing a cell, e.g., a cultured cell, a transformed cell, a cell from a cancer, or a test organism; administering said treatment to said cell (or test organism) and evaluating the expression of one or more FGF genes, e.g., by contacting a sample from said cell (or test organism) with, a nucleic acid probe that selectively hybridizes to one or more FGF genes, or an antibody that specifically binds to one or more FGF gene products; wherein a change, e.g., decrease, in the level of said one or more FGF genes or gene products in a sample given said treatment, e.g., relative to the level of expression without the treatment, indicative of the effectiveness of the compound for treating said disorder.
  • a change e.g., decrease
  • the FGF genes or gene product is selected from the group consisting of bFGF, aFGF, TSC22, VEGF, GAFA1, GAFA2, GAFA3 (originally designated as FSC1, FSC2, FSC3 in the U.S. Provisional Application No. 60/137,345), and TFII-I.
  • an FGF gene is evaluated by evaluating the expression of a signal entity, e.g., a green fluorescent protein or other marker protein, which is under the control or an FGF gene control element, e.g., promoter.
  • a signal entity e.g., a green fluorescent protein or other marker protein
  • an FGF gene control element e.g., promoter.
  • nucleic acid (or protein) from the cell or sample is analyzed on positional a ⁇ ays, e.g., DNA-chip a ⁇ ays.
  • the method further includes: analyzing the sample by providing an a ⁇ ay of a plurality of capture probes, wherein each of the capture probes is positionally distinguishable from other capture probes of the plurality on the array, and wherein each positional distinguishable capture probe includes a unique reagent, e.g., an antibody or a nucleic acid probe which can identify an FGF gene or gene product; hybridizing the sample with the a ⁇ ay of capture probes, thereby analyzing the sample sequence.
  • a unique reagent e.g., an antibody or a nucleic acid probe which can identify an FGF gene or gene product
  • Figure 1 depicts a panel of graphs showing the induction of resistance to paclitaxel, doxorubicin and 5-fluorouracil in monolayer cultures of rat MAT-LyLu tumor cells using conditioned medium (CM) collected from tumor cultures.
  • CM conditioned medium
  • the induction of drug resistance was detected as changes in proliferative activity by measuring the bromodeoxyuridine (BrdU) inco ⁇ oration in treated samples relative to controls.
  • Top panels CM of primary tumors.
  • Middle panels CM of lymph node metastases.
  • Bottom panels CM of lung metastases.
  • Figures 2A-2B depict a panel of graphs showing the reversal of CM-induced resistance by inhibiting or removing aFGF/bFGF.
  • Figure 2 A shows the reversal of CM-induced resistance (detected as changes in proliferative activity of rat MAT-LyLu cells by measuring the changes in BrdU inco ⁇ oration in treated samples relative to control samples) using a monoclonal antibody against aFGF or bFGF as indicated.
  • the source of CM was lung histocultures for the aFGF experiment, and early monolayer lung cultures for the bFGF experiment.
  • Figure 2B shows (a) the reversal of the CM-induced resistance to doxorubicin by removing aFGF and/or bFGF via immunoprecipitation, and (b) the restoration of the resistance by reconstituting the FGF-immunoprecipitated CM using human recombinant proteins (0.16 ng/ml r-aFGF and/or 0.9 ng/ml r-bFGF).
  • the study was performed using rat MAT-LyLu cells.
  • the reversal in CM-induced resistance was detected by measuring the changes in BrdU inco ⁇ oration in treated samples relative to control samples.
  • the source of CM was lung histocultures for both aFGF and bFGF experiments.
  • CM minus aFGF and bFGF filled square, overlaps with left control curve
  • CM minus aFGF and bFGF plus r-bFGF filled circle
  • Lower right CM minus aFGF and bFGF (filled square, overlaps with left control curve); CM minus aFGF and bFGF plus r-aFGF and r-bFGF (filled circle, overlaps with the right CM-control curve).
  • Figure 3 is a panel of graphs showing induction of resistance to paclitaxel, doxorubicin and 5-fluorouracil in human prostate PC3 tumor cells using r-aFGF alone, r-bFGF alone or a combination of r-aFGF and r-bFGF, in the absence of CM.
  • the study was performed using human prostate PC3 tumor cells (shown here) and rat MAT- LyLu tumor cells.
  • the induction of resistance by FGF was detected by measuring the changes in BrdU inco ⁇ oration in treated samples relative to control samples.
  • the study used two controls: no CM (left solid curves, empty circles); with CM of lung histocultures (right solid curves, open squares).
  • Top panels addition of r-aFGF: 1 (filled circle), 10 (filled square) and 50 (filled inverted triangle) ng/ml. All three dotted curves overlap with the left control curve.
  • Middle panels addition of r-bFGF. Dotted curves from left to right: 1 (filled circle, overlaps with the left control curve), 10 (filled square) and 50 (filled inverted triangle, overlaps with the right CM-control curve) ng/ml.
  • Bottom panels addition of r-aFGF/r-bFGF combinations.
  • Dotted curves from left to right 0.04 and 1 ng/ml (filled square, overlaps with the left control curve); 0.08 and 1 ng/ml (open diamond); 0.16 and 1 ng/ml (filled inverted triangle); 0.32 and 1 ng/ml (filled diamond); 0.64 and 1 ng/ml (filled circle); and 1 ng/ml each (open inverted triangle). Similar results were obtained for rat MAT-LyLu tumor cells.
  • Figure is a graph depicting the FGF-induced resistance to cell kill caused by doxorubicin treatment.
  • Drug-induced cell death in PC3 cells was monitored by the release of lactate dehydrogenase (LDH) to the culture medium. Results are expressed as a ratio of treated samples to untreated controls. Values for control samples varied by less than 10%.
  • LDH lactate dehydrogenase
  • Curves from top to bottom Without CM or FGF (open circle); addition of 0.9 ng/ml aFGF (filled inverted triangle, overlaps with top control curve); addition of 0.9 ng/ml bFGF (filled circle); addition of 0.16 ng/ml aFGF plus 0.9 ng/ml bFGF (filled diamond); CM of lung tumor histocultures (open square); 0.9 ng/ml aFGF plus 0.9 ng/ml bFGF (filled square). Similar results were found for paclitaxel, and 5- fluorouracil.
  • Figure 5 shows three graphs depicting the cytotoxicity of increasing concentrations of suramin in rat MAT-LyLu tumor cells, human prostate PC3 and PC3- LN tumor cells. These results show that suramin did not produce measurable cytotoxicity (i.e., ⁇ 10% reduction in BrdU inco ⁇ oration) at the 1-15 micromolar concentration.
  • the co ⁇ esponding IC 5 o values i.e., suramin concentration required to result in 50% reduction in BrdU inco ⁇ oration
  • Figure 6 shows three graphs depicting the enhancement of the cytotoxicity of doxorubicin, paclitaxel and 5-fluorouracil using suramin, under in vitro conditions.
  • the study was performed using rat MAT-LyLu tumor cells (shown here) and human prostate PC3 and PC3-LN tumor cells.
  • the cytotoxic effect was detected by measuring the changes in BrdU inco ⁇ oration in treated samples relative to control samples.
  • the study used two controls: no CM and no suramin (left solid curves, open circle); with CM of lung histocultures but no suramin (right solid curves, open square). Dotted curves from right to left, addition of suramin 5 ⁇ M (filled circle); 10 ⁇ M (filled square); and 15 ⁇ M (filled inverted triangle). Similar results were obtained for PC3 and PC3-LN cells.
  • Figures 7A-7B are two graphs depicting the suramin-mediated enhancement of doxorubicin activity in immunodeficient mice bearing subcutaneous bulky human prostate PC3 xenograft tumors.
  • Figure 7 A shows the changes in tumor weight (depicted as experimental weight relative to initial weight) with respect to duration of treatment.
  • Figure 7B shows the changes in the animal body weight with respect to duration of treatment.
  • the present invention is based, at least in part, on the discovery that bFGF induces broad spectrum resistance to anticancer agents, in a number of solid and soft- tissue tumors, metastatic lesions, as well as normal noncancerous intestinal epithelium.
  • the bFGF-induced resistance was amplified by aFGF.
  • the invention shows that inhibitors of aFGF/bFGF enhanced the in vitro and in vivo activity of chemotherapy, resulting in shrinkage and eradication of well-established human lung metastasis and human subcutaneous xenograft tumors in mice. Accordingly, the present invention provides, in part, methods and compositions for inhibiting FGF- induced resistance to anticancer agents.
  • the FGF inhibitor is administered with other cytotoxic agents, e.g., an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti- hormone, e.g., an antibody against growth factor receptors), an agent that promotes apoptosis and/or necrosis, an interferon, an interleukin, a tumor necrosis factor, and radiation.
  • cytotoxic agents e.g., an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g.,
  • the enhanced, and sometimes synergistic, effect of the FGF inhibitors) e.g., suramin, e.g., FGF antibody
  • anticancer agents e.g., paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, 5-fluorouracil, UFT, tamoxifen, goserelin, Herceptin, anti- CD20, leuprolide (Lupron) and flutamide
  • FGF inhibitors e.g., suramin, e.g., FGF antibody
  • anticancer agents e.g., paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, 5-fluorouracil, UFT,
  • the subject is a patient with non-small cell lung cancer, who is treated with a combination of paclitaxel, carboplatin and suramin, or with a combination of gemcitabine, cisplatin, and suramin.
  • the patient is a patient with hormone refractory prostate cancer, who is treated with a combination of estramustine phosphate, taxotere and suramin, or with a combination of doxorubicin, ketoconazole and suramin.
  • the patient is a patient with metastatic breast cancer, who is treated with a combination of cyclophosphamide, doxorubicin, 5- fluorouracil and suramin.
  • the patient is a patient with advanced breast cancer that overexpresses the HER2/neu oncogene, who is treated with a Her2/neu antibody (e.g., Herceptin) and suramin, with or without paclitaxel or cisplatin.
  • a Her2/neu antibody e.g., Herceptin
  • suramin e.g., paclitaxel or cisplatin.
  • the patient is a patient with advanced or metastatic colorectal cancer, who is treated with a combination of irinotecan and suramin.
  • the patient is a patient with advanced colon cancer, who is treated with a combination of 5- fluorouracil, leucovorin and suramin.
  • CM conditioned medium
  • solid, soft-tissue tumors, and metastatic tumors induced resistance to multiple anticancer agents, in human and rodent tumor cells.
  • CM contained elevated levels of aFGF and bFGF (see Examples II- III).
  • Inhibition of bFGF using a bFGF-specific monoclonal antibody and removal of bFGF by immunoprecipitation resulted in complete reversal of the CM-induced resistance to anticancer agents, whereas inhibition/removal of aFGF resulted in partial reversal (see Example IV).
  • CM that had been depleted of aFGF and/or bFGF and subsequently reconstituted with respective human recombinant proteins
  • bFGF but not aFGF was found to induce resistance.
  • aFGF was found to amplify the bFGF effect.
  • aFGF and bFGF fully accounted for the CM effect, indicating these proteins are involved in the induction and maintenance of resistance to anticancer agents (see Example IV).
  • the FGF-induced resistance was not due to reduced intracellular drug accumulation, nor altered cell proliferation (see Example XII), thus representing a novel epigenetic resistance mechanism.
  • aFGF and bFGF as found in the active CM, induced broad spectrum resistance to at least 58 anticancer agents having different mechanisms of actions, including antitubulin/antimicrotubule agents, topoisomerase I inhibitors, topoisomerase II inhibitors, antimetabohtes, alkylating agents, intercalating agents, agents that interfere with signal transduction pathways (e.g., protein kinase C inhibitors), and agents that promote apoptosis and/or necrosis (see Examples II and VII).
  • anticancer agents having different mechanisms of actions, including antitubulin/antimicrotubule agents, topoisomerase I inhibitors, topoisomerase II inhibitors, antimetabohtes, alkylating agents, intercalating agents, agents that interfere with signal transduction pathways (e.g., protein kinase C inhibitors), and agents that promote apoptosis and/or necrosis (see Examples II and VII).
  • aFGF/bFGF induction of resistance to paclitaxel an antimicrotubule agent
  • doxorubicin a topoisomerase I inhibitor
  • 5-fluorouracil an antimetabolite
  • inhibitors of FGF including FGF antibodies and suramin, an inhibitor of multiple growth factors, including aFGF/bFGF, reverse the FGF-induced resistance and enhance the in vitro activity of anticancer agents, including at least 58 anticancer agents having different mechanisms of actions, including antitubulin antimicrotubule agents, topoisomerase I inhibitors, topoisomerase II inhibitors, antimetabohtes, alkylating agents, intercalating agents, agents that interfere with signal transduction pathways (e.g., protein kinase C inhibitors), and agents that promote apoptosis and/or necrosis (see Examples V and VII).
  • anticancer agents including antitubulin antimicrotubule agents, topoisomerase I inhibitors, topoisomerase II inhibitors, antimetabohtes, alkylating agents, intercalating agents, agents that interfere with signal transduction pathways (e.g., protein kinase C inhibitors), and agents that promote apoptosis and/or necrosis (
  • the appended examples further demonstrate that (a) presence of tumor cells enhanced the level of extracellular bFGF in host tissues, (b) bFGF level in tumors is determined by the location and the size of the tumor, and (c) bFGF level determines the sensitivity of human patient tumors to anticancer agents (see Example X).
  • the appended examples show that treatment of human prostate cells with several anticancer agents with diverse structures and action mechanisms (e.g., antitubulin/antimicrotubule agents, topoisomerase I inhibitors, topoisomerase II inhibitors, alkylating agents, and antimetabohtes) significantly enhanced the level of extracellular bFGF (see Example XI).
  • these examples indicate (a) the use of FGF gene expression and level to detect the presence of tumor cells, (b) the use of FGF expression and level in tumors to determine the extent of resistance, (c) the use of FGF gene expression and level in tumors to determine the dosage of the FGF inhibitors, and (d) that patients that are previously treated with chemotherapy are likely to exhibit chemoresistance. The latter indicates the need of using FGF inhibitors to treat patients who have failed cancer therapy and have recurrent tumors.
  • FGF antagonists e.g., suramin, e.g., heparin, e.g., low molecular weight heparin, e.g., heparan sulfate
  • FGF antagonists e.g., suramin, e.g., heparin, e.g., low molecular weight heparin, e.g., heparan sulfate
  • FGF agonists e.g., aFGF, bFGF
  • anticancer agents e.g., paclitaxel and doxorubicin
  • the invention also provides methods and compositions for treating or preventing the undesired host toxicity of anticancer agents using aFGF and bFGF, alone or in combination.
  • the findings described herein demonstrate, inter alia, (a) a novel epigenetic mechanism by which cancer cells utilize the unique microenvironment of solid tumors and metastases to elude cytotoxic insult, (b) an important role of extracellular growth factors in tumor sensitivity to anticancer agents, (c) a new cancer treatment paradigm using combinations of anticancer agents with aFGF/bFGF inhibitors, (d) a novel mechanism and methods by which normal noncancerous host cells can be protected from the undesired toxicity of anticancer agents, and (e) methods to use the bFGF level for making therapeutic decision for individual patients.
  • fibroblast growth factor refers to a member of a family of polypeptides that are potent regulators of a variety of cellular processes including proliferation, differentiation, migration, mo ⁇ hogenesis, tissue maintenance and in wound healing and repair (Clarke et al. (1993) J. Cell Sci.106: 121-133; Cuevas et al. (1988) Biochem. Biophys. Res. Commun. 156: 611-618; Burgess, W.H. and Maciag, T (1989) Ann. Rev. Biochem. 58: 575-606; Rifkin, D.B.
  • FGF fibroblast growth factor
  • the FGF family cu ⁇ ently includes at least 19 structurally and functionally related proteins, including acidic and basic FGF, FGF-1 and FGF-2 respectively; int2 (FGF-3); hst (FGF-4); FGF-5; hst2 (FGF-6); keratinocyte growth factor (FGF-7); androgen-induced growth factor (FGF-8); glia-activating factor (FGF- 9); FGF-10-19 (Galzie, Z. et al. (1997) Biochem. Cell. Biol., 75:669-685; Yamasaki, M. et al. (1996) J. Biol.
  • FGF refers to acidic and basic FGF, FGF-1 and FGF-2, respectively (reviewed in (Galzie, Z. et al. (1997) Biochem. Cell. Biol., 75:669-685 and Burgess, W.H. and Maciag, T (1989) Ann. Rev. Biochem. 58: 575-606).
  • FGF receptor refers to FGF -binding polypeptides that possess intrinsic tyrosine kinase activity or heparan sulfate proteoglycans in the extracellular matrix.
  • tyrosine kinase receptors there are cu ⁇ ently four known genes encoding FGF receptors (FGFR-1, FGFR-2, FGFR-3, and FGFR-4), which can give rise to a variety of protein isoforms via alternative RNA splicing (Galzie, Z. et al. (1997) Biochem. Cell. Biol., 75:669-685).
  • the structure of the FGFR consists of an extracellular region with three immunoglobulin-like domains, a transmembrane region, and a cytosolic tyrosine kinase domain that is activated upon ligand binding. FGF binding causes dimerization of the receptors, resulting in receptor autophosphorylation on tyrosine residues and the activation of intracellular signal transduction cascades.
  • the term "FGFR” also includes heparan sulfate proteoglycans in the extracellular matrix which mediate the actions of FGF, e.g., protection from proteolysis, localization, storage, and internalization of growth factors (Faham, S. et al. (1998) Curr. Opin. Struct.
  • Heparan sulfate proteoglycans may serve as low affinity FGF receptors that act to present FGF to its cognate FGFR, and/or to facilitate receptor oligomerization (Galzie, Z. et al. (1997) Biochem. Cell. Biol, 75:669-685).
  • an "FGF antagonist" refers to an agent that inhibits (completely or partially) the activity, production, stability, of an FGF molecule.
  • the FGF antagonist acts extracellularly or intracellularly, e.g., inhibits the binding of an FGF molecule to an extracellular or intracellular domain of an FGF receptor, e.g., blocks the intracellular signalling initiated by activation of the FGF receptor.
  • the inhibitor binds to the intracellular domain of the FGF receptor, or blocks a downstream action that is consequence of activation of the FGF receptor.
  • an "FGF agonist” refers to an agent that potentiates the activity, production, stability, of an FGF molecule, or activates FGF receptors.
  • telomere telomere telomere telomere telomere telomere telomere telomere telomere telomere telomere telomere telomere telomere telomere telomere telomere telomere telomere telomere telomere telomere telomere telomere telomere telomere telomere telomere telomeres As used herein, the term “specific inhibitor of FGF” refers to an FGF antagonist other than suramin.
  • cytotoxic agent As used herein, the terms “cytotoxic agent”, “anticancer agent” and “antitumor agent” are used interchangeably herein and refer to agents that have the property of inhibiting the growth or proliferation (e.g., a cytostatic agent), or inducing the killing, of hype ⁇ roliferative cells.
  • the cytotoxic agent inhibits or reverses the development or progression of a neoplasm, such as a solid tumor, a soft tissue tumor, a metastatic lesion, a lymphoma, or a leukemia, or the cytotoxic agent inhibits or reverses the development or progression of a benign hype ⁇ lastic growth.
  • a "therapeutically effective amount" of an FGF antagonist and a cytotoxic agent refers to an amount of such agents which in combination is effective, upon single- or multiple-dose administration to the subject, e.g., a patient, at inhibiting the growth or proliferation, or inducing the killing, of hype ⁇ roliferative cells.
  • Such growth inhibition or killing can be reflected as a prolongation of the survival of the subject, e.g., a patient beyond that expected in the absence of such treatment, or any improvement in the prognosis of the subject relative to the absence of such treatment.
  • the phrase refers to an amount of such agent which is effective, upon single- or multiple-dose administration to protect a cell in a subject, e.g., a patient, from damage by an agent, e.g., a cytotoxic agent.
  • a prophylactically effective amount of an FGF antagonist and a cytotoxic agent refers to an amount of such agents which in combination is effective, upon single or multiple dose administration to the subject, e.g., a patient, in preventing or delaying the occu ⁇ ence of the onset or recu ⁇ ence of a neoplastic disease state.
  • the phrase refers to an amount of such agent which is effective, upon single- or multiple-dose administration, in preventing or delaying damage to a cell in a subject, e.g., a patient, by an agent, e.g., a cytotoxic agent.
  • the language "subject” is intended to include human and non- human animals.
  • Prefe ⁇ ed human animals include a human patient having a disorder characterized by the abe ⁇ ant activity of a hype ⁇ roliferative cell.
  • the term "non- human animals” of the invention includes all vertebrates, e.g., mammals and non- mammals, such as non-human primates, sheep, dog, cat, horse, cow, chickens, amphibians, reptiles, etc.
  • the subject is a human patient, e.g., a cancer patient, e.g., a patient with a benign hype ⁇ lastic disorder.
  • neoplastic cell e.g., benign hype ⁇ lastic cell
  • inhibiting the growth or proliferation of the hype ⁇ roliferative cell refers to slowing, interrupting, a ⁇ esting or stopping its growth and metastasis, and does not necessarily indicate a total elimination of the neoplastic growth.
  • inducing the killing of the hype ⁇ roliferative cell refers to the partial or complete elimination of such cells, and does not necessarily indicate a total elimination of the neoplastic growth.
  • hypo ⁇ roliferative As used herein, the terms “hype ⁇ roliferative”, “hype ⁇ lastic”, “malignant”, and “neoplastic” are used interchangeably, and refer to those cells in an abnormal state or condition characterized by rapid proliferation or neoplasm. The terms are meant to include all types of hype ⁇ roliferative growth, hype ⁇ lastic growth, cancerous growths or oncogenic processes, metastatic tissues or malignantly transformed cells, tissues, or organs, i ⁇ espective of histopathologic type or stage of invasiveness. "Pathologic hype ⁇ roliferative" cells occur in disease states characterized by malignant tumor growth or in disease states characterized by benign hype ⁇ roliferative and hype ⁇ lastic cells.
  • Neoplasia refers to "new cell growth” that results as a loss of responsiveness to normal growth controls, e.g., neoplastic cell growth.
  • a “hype ⁇ lasia” refers to cells undergoing an abnormally high rate of growth.
  • neoplasia and hype ⁇ lasia can be used interchangeably, as their context will reveal, referring generally to cells experiencing abnormal cell growth rates.
  • Neoplasias and hype ⁇ lasias include “tumors,” which may be either benign, premalignant or malignant.
  • carcinoma is recognized by those skilled in the art and refers to malignancies of epithelial or endocrine tissues including respiratory system carcinomas, gastrointestinal system carcinomas, genitourinary system carcinomas, testicular carcinomas, breast carcinomas, prostatic carcinomas, endocrine system carcinomas, and melanomas.
  • Exemplary carcinomas include those forming from tissue of the cervix, lung, prostate, breast, head and neck, colon and ovary.
  • carcinosarcomas e.g., which include malignant tumors composed of carcinomatous and sarcomatous tissues.
  • An "adenocarcinoma” refers to a carcinoma derived from glandular tissue or in which the tumor cells form recognizable glandular structures.
  • leukemia or leukemic cancer refers to all cancers or neoplasias of the hemopoietic and immune systems (blood and lymphatic system). These terms refer to a progressive, malignant disease of the blood-forming organs, marked by distorted proliferation and development of leukocytes and their precursors in the blood and bone marrow.
  • Myelolomas refer to other types of tumors of the blood, bone ma ⁇ ow cells.
  • Lymphomas refer to tumors of the lymph tissue.
  • immunoglobuhn and “antibody” refer to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds.
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as HCVR or VH) and a heavy chain constant region.
  • the heavy chain constant region is comprised of three domains, CHI, CH2 and CH3.
  • Each light chain is comprised of a light chain variable region (abbreviated herein as LCVR or VL) and a light chain constant region.
  • the light chain constant region is comprised of one domain, CL.
  • VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDR complementarity determining regions
  • FR framework regions
  • Each VH and VL is composed of three CDRs and four FRs, a ⁇ anged from amino-terminus to carboxy-terminus in the following order: FRl, CDRl, FR2, CDR2, FR3, CDR3, FR4.
  • the variable regions of the heavy and light chains contain a binding domain that interacts with an antigen.
  • the constant regions of the antibodies may mediate the binding of the immunoglobuhn to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clq) of the classical complement system.
  • an antibody refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., a target antigen).
  • binding fragments encompassed within the term “antigen-binding portion” of an antibody include (a) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHI domains; (b) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (c) a Fd fragment consisting of the VH and CHI domains; (d) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (e) a dAb fragment (Ward et al, (1989) Nature 341:544-546), which consists of a VH domain; and (f) an isolated complementarity determining region (
  • the two domains of the Fv fragment, VL and VH are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl Acad. Sci. USA 85:5879-5883).
  • single chain Fv single chain Fv
  • Such single chain antibodies are encompassed within the term "antigen-binding portion" of an antibody.
  • the term "monoclonal antibody” as used herein refers to an antibody molecule of single molecular composition.
  • a monoclonal antibody composition displays a single binding specificity and affinity for a particular epitope.
  • the term “human monoclonal antibody” refers to antibodies displaying a single binding specificity which have variable and constant regions derived from human germline immunoglobuhn sequences.
  • the human monoclonal antibodies are produced by a hybridoma which includes a B cell obtained from a transgenic non-human animal, e.g., a transgenic mouse, having a genome comprising a human heavy chain transgene and a light chain transgene fused to an immortalized cell.
  • recombinant human antibody is intended to include all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies isolated from an animal (e.g., a mouse) that is transgenic for human immunoglobuhn genes; antibodies expressed using a recombinant expression vector transfected into a host cell, antibodies isolated from a recombinant, combinatorial human antibody library, or antibodies prepared, expressed, created or isolated by any other means that involves splicing of human immunoglobuhn gene sequences to other DNA sequences.
  • Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobuhn sequences.
  • such recombinant human antibodies are subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo.
  • this invention features methods for inhibiting the proliferation, and/or enhancing the killing of, hype ⁇ roliferative cells, by contacting the cells with at least one cytotoxic agent and at least one FGF antagonist.
  • the method includes a step of contacting pathological hype ⁇ roliferative cells with an amount of at least one cytotoxic agent and at least one FGF antagonist, which, in combination, is effective to reduce or inhibit the proliferation of the cell, or induce cell killing.
  • the present method can be performed on cells in culture, e.g., in vitro or ex vivo, or can be performed on cells present in a subject, e.g., as part of an in vivo therapeutic protocol.
  • the therapeutic regimen can be carried out on a human or on other animal subjects.
  • the enhanced therapeutic effectiveness of the combination therapy of the present invention represents a promising alternative to conventional highly toxic regimens of anticancer agents.
  • the subject methods are preferably combined with other anticancer agents, e.g., antimicrotubule agents, topoisomerase I inhibitors, topoisomerase II inhibitors, antimetabohtes, mitotic inhibitors, alkylating agents, intercalating agents, agents capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), agents that promotes apoptosis and/or necrosis, an interferon, an interleukin, a tumor necrosis factor, and radiation.
  • a signal transduction pathway e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors
  • agents that promotes apoptosis and/or necrosis e.g., an interferon, an interleukin, a tumor necrosis factor
  • the enhanced, and sometimes synergistic, effect of the FGF inhibitor(s) e.g., suramin, e.g., FGF antibody
  • anticancer agents e.g., paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, 5-fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, Herceptin, anti- CD20, leuprolide (Lupron) and flutamide), in addition to improving the efficacy of these anticancer agents, may allow for the administration of lower doses of these anticancer agents, thus reducing the induction of side effects in a subject.
  • FGF inhibitor(s) e.g., suramin, e.g., FGF antibody
  • anticancer agents e.g., paclitaxel, interferon alpha,
  • the subject method can be useful in treating malignancies of the various organ systems, such as those affecting lung, breast, lymphoid, gastrointestinal (e.g., colon), and genitourinary tract (e.g., prostate), pharynx, as well as adenocarcinomas which include malignancies such as most colon cancer, rectal cancer, renal cell carcinoma, prostate cancer and/or testicular tumors, non-small cell carcinoma of the lung, cancer of the small intestine and cancer of the esophagus.
  • malignancies of the various organ systems such as those affecting lung, breast, lymphoid, gastrointestinal (e.g., colon), and genitourinary tract (e.g., prostate), pharynx, as well as adenocarcinomas which include malignancies such as most colon cancer, rectal cancer, renal cell carcinoma, prostate cancer and/or testicular tumors, non-small cell carcinoma of the lung, cancer of the small intestine and cancer of the esophagus.
  • Exemplary solid tumors that can be treated include: fibrosarcoma, myosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, gastric cancer, esophageal cancer, colon carcinoma, rectal cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, uterine cancer, cancer of the head and neck, skin cancer, brain cancer, squamous cell carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinoma, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma,
  • the subject method may also be used to inhibit the proliferation or induce the killing of hype ⁇ lastic/neoplastic cells of hematopoietic origin, e.g., arising from myeloid, lymphoid or erythroid lineages, or precursor cells thereof.
  • the present invention contemplates the treatment of various myeloid disorders including, but not limited to, acute promyeloid leukemia (APML), acute myelogenous leukemia (AML) and chronic myelogenous leukemia (CML) (reviewed in Vaickus, L. (1991) Crit. Rev. Oncol/Hemotol. 11:267-97).
  • APML acute promyeloid leukemia
  • AML acute myelogenous leukemia
  • CML chronic myelogenous leukemia
  • Lymphoid malignancies which may be treated by the subject method include, but are not limited to acute lymphoblastic leukemia (ALL), which includes B-lineage ALL and T-lineage ALL, chronic lymphocytic leukemia (CLL), prolymphocytic leukemia (PLL), hairy cell leukemia (HLL) and
  • ALL acute lymphoblastic leukemia
  • CLL chronic lymphocytic leukemia
  • PLL prolymphocytic leukemia
  • HLL hairy cell leukemia
  • WM Waldenstrom's macroglobulinemia
  • Additional forms of malignant lymphomas contemplated by the treatment method of the present invention include, but are not limited to, non-Hodgkin's lymphoma and variants thereof, peripheral T-cell lymphomas, adult T-cell leukemia/lymphoma (ATL), cutaneous T-cell lymphoma (CTCL), large granular lymphocytic leukemia (LGF) and Hodgkin's disease.
  • the subject method may also be used to inhibit the proliferation or induce the killing of benign hype ⁇ lastic growth.
  • FGF antagonists are also be used to inhibit the proliferation or induce the killing of benign hype ⁇ lastic growth.
  • the FGF antagonist used in the methods and compositions of the present invention can be a chemical, e.g., a protein or a peptide, or a small molecule.
  • the FGF antagonist is an inhibitor of bFGF, aFGF, or an inhibitor of both.
  • the FGF antagonist can be capable of binding to an FGF molecule, a molecule that facilitates the binding of FGF to a receptor, or an FGF receptor and, thereby, blocking the binding of an FGF molecule to a receptor (e.g., a tyrosine kinase receptor and/or a heparan sulfate proteoglycan).
  • the FGF antagonist may also down regulate FGF receptor action, e.g., blocks the intracellular signalling initiated by activation of the FGF receptor.
  • the inhibitor binds to the intracellular domain of the FGF receptor, or blocks a downstream action that is consequence of activation of the FGF receptor.
  • Examples of chemicals that may antagonize FGF action include suramin, structural analogs of suramin, pentosan polysulfate, scopolamine, angiostatin, sprouty, estradiol, carboxymethylbenzylamme dextran (CMDB7), suradista, insulin-like growth factor binding protein-3, ethanol, heparin (e.g., 6-O-desulfated heparin), low molecular weight heparin, protamine sulfate, cyclosporin A, or RNA ligands for bFGF.
  • the FGF antagonist may also be a small molecule, e.g., a member of a combinatorial library.
  • a prefe ⁇ ed FGF antagonist is suramin.
  • suramin is present in a concentration sufficient to block the resistance to anticancer agents induced by FGF (e.g., bFGF and/or aFGF), but is not sufficient to produce significant cytotoxicity (e.g., significant inhibition of cell proliferation, e.g., significant cell death) in human and/or animal tumor cells, not sufficient to cause significant cell cycle a ⁇ est, and/or not sufficient to produce measurable antitumor effect in a subject, e.g., a human subject.
  • FGF e.g., bFGF and/or aFGF
  • suramin is administered in an amount that results in a plasma concentration ranging from about 0.1 to 100 microgram/ml, preferably about 1 to 85 microgram/ml, more preferably, about 5 to 60 microgram/ml, even more preferably, about 10 to 50 microgram/ml, and most preferably, 15 to 45 microgram/ml, of the plasma concentration.
  • proteins or peptide that may antagonize FGF activity include antibodies, e.g., a monoclonal antibody or an antigen fragment thereof, as described below.
  • the FGF antagonist is a fragment of the FGF molecule.
  • the FGF fragment competes with an FGF molecule for binding to the receptor.
  • the FGF antagonist may also be transforming growth factor beta or a fragment thereof.
  • proteins or peptide that may antagonize FGF activity include antibodies, e.g., a monoclonal antibody, a murine antibody or a human antibody, or an antigen-binding fragment thereof.
  • the monoclonal antibody is a human antibody.
  • the human antibody can be produced by a hybridoma which includes a B cell obtained from a transgenic non-human animal, e.g., a transgenic mouse, having a genome comprising a human heavy chain transgene and a light chain transgene fused to an immortalized cell.
  • the antibodies can be of the various isotypes, including: IgG (e.g., IgGl, IgG2, IgG3, IgG4), IgM, IgNl, IgA2, IgD, of IgE.
  • the antibody is an IgG isotype.
  • the antibodies can be full-length (e.g., an IgGl or IgG4 antibody) or can include only an antigen-binding fragment (e.g., a Fab, F(ab')2, Fv or a single chain Fv fragment).
  • antibodies against bFGF and FGF are commercially available, and can be readily used in the methods and compositions of the invention.
  • Examples of antibodies to bFGF that are commercially available include: (a) Sigma F 9666, anti- human, monoclonal, clone no. FA-88, host: mouse, type:IgG2a; (b) Sigma F 5521, polyclonal, host: rabbit; and (c) Oncogene PC316L, anti-bovine+anti-human, polyclonal, host: rabbit, type IgG.
  • antibodies to aFGF examples include: (a) Sigma F 6162, anti-human, monoclonal, clone no. FB-8, host: mouse, type: IgGl; (b) Sigma F 5537, anti-human, polyclonal, host: rabbit, type: IgG; (c) Sigma F 3393, anti- bovine, polyclonal, host: rabbit; (d) Oncogene PC15, anti-human, polyclonal, AB-1, host: rabbit, type: IgG; (e) Oncogene PC 16, anti-human, polyclonal, AB-2, host: rabbit, type: IgG; (f) Oncogene GF22, anti-human, monoclonal, clone no.
  • 3H3, AB-3 host: mouse, type: IgGl;
  • Oncogene GF23L anti-human, monoclonal, clone no. 98, AB-4, host: mouse, type: IgGl;
  • Oncogene GF24 anti-human, monoclonal, clone no. 52, AB-5, host: mouse, type: IgG2b; and
  • the anti-FGF antibody can be made by methods known in the art.
  • an anti-FGF antibody can be produced by a variety of techniques, including conventional monoclonal antibody methodology e.g., the standard somatic cell hybridization technique of Kohler and Milstein (1975) Nature 256: 495.
  • somatic cell hybridization procedures are prefe ⁇ ed, in principle, other techniques for producing monoclonal antibody can be employed, e.g., viral or oncogenic transformation of B lymphocytes.
  • the prefe ⁇ ed animal system for preparing hybridomas is the murine system. Hybridoma production in the mouse is a very well- established procedure. Immunization protocols and techniques for isolation of immunized splenocytes for fusion are known in the art.
  • Fusion partners e.g., murine myeloma cells
  • Human monoclonal antibodies (mAbs) directed against human proteins can be generated using transgenic mice carrying the complete human immune system rather than the mouse system. Splenocytes from these transgenic mice immunized with the antigen of interest are used to produce hybridomas that secrete human mAbs with specific affinities for epitopes from a human protein (see, e.g., Wood et al. International Application WO 91/00906, Kucherlapati et al. PCT publication WO 91/10741; Lonberg et al. International Application WO 92/03918; Kay et al.
  • Monoclonal antibodies can also be generated by other methods known to those skilled in the art of recombinant DNA technology.
  • An alternative method, refe ⁇ ed to as the "combinatorial antibody display” method, has been developed to identify and isolate antibody fragments having a particular antigen specificity, and can be utilized to produce monoclonal antibodies (for descriptions of combinatorial antibody display see e.g., Sastry et al. (1989) Proc. Natl. Acad. Sci. USA 86:5728; Huse et al. (1989) Science 246:1275; and Orlandi et al. (1989) Proc. Natl Acad. Sci. USA 86:3833; Larrick et al. (1991), Biotechniques 11 :152-156; Larrick et al (1991), Methods: Companion to Methods in Enzymology 2:106-110).
  • RNA is isolated from B lymphocytes, for example, peripheral blood cells, bone ma ⁇ ow, or spleen preparations, using standard protocols (e.g., U.S. Patent No. 4,683,202; Orlandi, et ⁇ /.(1989) Proc. Natl. Acad. Sci. USA 86:3833-3837; Sastry et al. (1989) Proc. Natl Acad. Sci. USA 86:5728-5732; and Huse et al.
  • standard protocols e.g., U.S. Patent No. 4,683,202; Orlandi, et ⁇ /.(1989) Proc. Natl. Acad. Sci. USA 86:3833-3837; Sastry et al. (1989) Proc. Natl Acad. Sci. USA 86:5728-5732; and Huse et al.
  • First-strand cDNA is synthesized using primers specific for the constant region of the heavy chain(s) and each of the K and ⁇ light chains, as well as primers for the signal sequence.
  • variable region PCR primers the variable regions of both heavy and light chains are amplified, each alone or in combination, and ligated into appropriate vectors for further manipulation in generating the display packages.
  • Ohgonucleotide primers useful in amplification protocols may be unique or degenerate or inco ⁇ orate inosine at degenerate positions. Restriction endonuclease recognition sequences may also be inco ⁇ orated into the primers to allow for the cloning of the amplified fragment into a vector in a predetermined reading frame for expression.
  • the V-gene library cloned from the immunization-derived antibody repertoire can be expressed by a population of display packages, preferably derived from filamentous phage, to form an antibody display library.
  • the display package comprises a system that allows the sampling of very large variegated antibody display libraries, rapid sorting after each affinity separation round, and easy isolation of the antibody gene from purified display packages.
  • kits for generating phage display libraries e.g., the Pharmacia Recombinant Phage
  • the V region domains of heavy and light chains can be expressed on the same polypeptide, joined by a flexible linker to form a single-chain Fv fragment, and the scFV gene subsequently cloned into the desired expression vector or phage genome.
  • a flexible linker As generally described in McCafferty et al. (1990) Nature 348:552- 554, complete VH and VL domains of an antibody, joined by a flexible (Gly_ ⁇ -Ser)3 linker can be used to produce a single chain antibody which can render the display package separable based on antigen affinity. Isolated scFV antibodies immunoreactive with the antigen can subsequently be formulated into a pharmaceutical preparation for use in the subject method.
  • the antibody library is screened with the target antigen, or peptide fragment thereof, to identify and isolate packages that express an antibody having specificity for the target antigen.
  • Nucleic acid encoding the selected antibody can be recovered from the display package (e.g., from the phage genome) and subcloned into other expression vectors by standard recombinant DNA techniques.
  • Specific antibody molecules with high affinities for a surface protein can be made according to methods known to those in the art, e.g., methods involving screening of libraries (Ladner, R.C., et al, U.S. Patent 5,233,409; Ladner, R.C., et al, U.S. Patent 5,403,484). Further, the methods of these libraries can be used in screens to obtain binding determinants that are mimetics of the structural determinants of antibodies.
  • the Fv binding surface of a particular antibody molecule interacts with its target ligand according to principles of protein-protein interactions, hence sequence data for VH and VL (the latter of which may be of the K or ⁇ chain type) is the basis for protein engineering techniques known to those with skill in the art.
  • sequence data for VH and VL is the basis for protein engineering techniques known to those with skill in the art.
  • Details of the protein surface that comprises the binding determinants can be obtained from antibody sequence information, by a modeling procedure using previously determined three-dimensional structures from other antibodies obtained from NMR studies or crytallographic data. See for example Bajorath, J. and S. Sheriff, (1996) Proteins: Struct., Fund., and Genet. 24, 152-157; Webster, D.M. and A. R. Rees (1995) "Molecular modeling of antibody-combining sites" in S.
  • a variegated peptide library is expressed by a population of display packages to form a peptide display library.
  • the display package comprises a system that allows the sampling of very large variegated peptide display libraries, rapid sorting after each affinity separation round, and easy isolation of the peptide-encoding gene from purified display packages.
  • Peptide display libraries can be in, e.g., prokaryotic organisms and viruses, which can be amplified quickly, are relatively easy to manipulate, and which allows the creation of large number of clones.
  • Prefe ⁇ ed display packages include, for example, vegetative bacterial cells, bacterial spores, and most preferably, bacterial viruses (especially DNA viruses).
  • modified or “recombinant” antibody is also intended to include antibodies, such as monoclonal antibodies, chimeric antibodies, and humanized antibodies which have been modified by, e.g., deleting, adding, or substituting portions of the antibody.
  • an antibody can be modified by deleting the hinge region, thus generating a monovalent antibody. Any modification is within the scope of the invention so long as the antibody has at least one antigen binding region specific.
  • Chimeric mouse-human monoclonal antibodies can be produced by recombinant DNA techniques known in the art (see Robinson et al, International Patent Publication PCT/US86/02269; Akira, et al, European Patent Application 184,187; Taniguchi, M., European Patent Application 171,496; Morrison et al, European Patent Application 173,494; Neuberger et al, International Application WO 86/01533; Cabilly et al. U.S. Patent No. 4,816,567; Cabilly et al, European Patent Application 125,023; Better et al. (1988) Science 240:1041-1043); Liu et al.
  • the chimeric antibody can be further humanized by replacing sequences of the Fv variable region which are not directly involved in antigen binding with equivalent sequences from human Fv variable regions.
  • General reviews of humanized chimeric antibodies are provided by Morrison, S. L. (1985) Science 229:1202-1207 and by Oi et al. (1986) BioTechniques 4:214. Those methods include isolating, manipulating, and expressing the nucleic acid sequences that encode all or part of immunoglobuhn Fv variable regions from at least one of a heavy or light chain. Sources of such nucleic acid are well known to those skilled in the art and, for example, may be obtained from 7E3, an anti-GPIIi,III a antibody producing hybridoma.
  • Suitable humanized antibodies can alternatively be produced by CDR substitution U.S. Patent 5,225,539; Jones et al. (1986) Nature 321 :552-525; Verhoeyan et al. (1988) Science 239:1534; and Beidler et al. (1988) J. Immunol. 141 :4053-4060.
  • All of the CDRs of a particular human antibody may be replaced with at least a portion of a non-human CDR or only some of the CDRs may be replaced with non- human CDRs. It is only necessary to replace the number of CDRs required for binding of the humanized antibody to the Fc receptor.
  • An antibody can be humanized by any method, which is capable of replacing at least a portion of a CDR of a human antibody with a CDR derived from a non-human antibody.
  • Winter describes a method which may be used to prepare the humanized antibodies of the present invention (UK Patent Application GB 2188638A).
  • the human CDRs may be replaced with non-human CDRs using ohgonucleotide site- directed mutagenesis.
  • chimeric and humanized antibodies in which specific amino acids have been substituted, deleted or added.
  • prefe ⁇ ed humanized antibodies have amino acid substitutions in the framework region, such as to improve binding to the antigen.
  • amino acids located in the human framework region can be replaced with the amino acids located at the co ⁇ esponding positions in the mouse antibody. Such substitutions are known to improve binding of humanized antibodies to the antigen in some instances.
  • Antibodies in which amino acids have been added, deleted, or substituted are refe ⁇ ed to herein as modified antibodies or altered antibodies. Cytotoxic Agents
  • the FGF antagonist is preferably administered in combination with at least one cytotoxic agent.
  • the term "in combination" in this context means that the agents are given substantially contemporaneously, either simultaneously or sequentially. If given sequentially, at the onset of administration of the second compound, the first of the two compounds is preferably still detectable at effective concentrations at the site where treatment effect is desired.
  • the FGF antagonists can be used in combination therapy with conventional cancer chemotherapeutics.
  • Conventional treatment regimens for leukemia and for other tumors include radiation, antitumor agents, interferons, interleukins, tumor necrosis factors, or a combination of two or more of these agents.
  • the subject method may involve, in addition to the use of at least one FGF inhibitor, one or more other antitumor agents.
  • exemplary combination therapies include the use of such agents as, e.g., the ones described in Table 2 below, including but not limited to: antitubulin/antimicrotubule agents, e.g., paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel); topoisomerase I inhibitors, e.g., topotecan, camptothecin, irinotecan hydrochloride (e.g., Camptosar); topoisomerase II inhibitors, e.g., doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride; antimetabohtes
  • Examples of additional agents that can be used in combination with the FGF inhibitors include hydroxyurea, azathioprine, aminopterin, trimethoprin, pyrimethamine, pyritrexim, DDMP (2,4 diamino- 5(3',4' dichlorophenyl)6 methylpyrimidine), 5,10- dideazatetrahydrofolate,10-propargyl-5,8 dideazafolate (CB3717), 10-ethyl-lO-deaza- aminopterin, deoxycytidine, 5-aza-cytosine arabinoside, N-4-palmitoyl-ara C, 2'-azido-2'- deoxy-ara C, N4-behenoyl-ara C, CCNU (lomustine), estramustine, MeCCNU, triethylene melamine, trenimon, dimethyl busulfan, streptozotocin, chlorozotocin, procarbazine, hexamethylmelamine (
  • cytotoxic agents can be used depending on the type of hype ⁇ roliferative disorder to be treated.
  • the following drugs usually in combinations with each other, are often used to treat acute leukemias: vincristine, prednisone, methotrexate, mercaptopurine, cyclophosphamide, and cytarabine.
  • chronic leukemia for example, fludarabine, busulfan, melphalan, and chlorambucil can be used in combination.
  • paclitaxel and carboplatin or a combination of gemcitabine and cisplatin is used.
  • a combination of estramustine phosphate and taxotere or a combination of doxorubicin and ketoconazole is used.
  • a combination of cyclophosphamide, doxorubicin and 5-fluorouracil is used.
  • an anti-Her2/neu antibody e.g., Herceptin
  • cisplatin is used.
  • 5-fluorouracil and leucovorin is used.
  • Cytotoxic agents are also used to treat benign hype ⁇ lasia disorders. For example, psoriasis is treated with 5-fluorouracil. Treatment can be initiated with smaller dosages that are less than the optimum dose of the agent. Thereafter, the dosage should be increased by small increments until the optimum effect under the circumstances is reached. For convenience, the total daily dosage may be divided and administered in portions during the day if desired.
  • a therapeutically effective antitumor amount and a prophylactically effective antitumor amount of an FGF inhibitor or a cytotoxic agent is expected to vary from about 0.1 milligram per kilogram of body weight per day (mg/kg/day) to about 100 mg/kg/day.
  • Determination of a therapeutically effective antitumor amount and a prophylactically effective antitumor amount of a FGF inhibitor and cytotoxic agent can be readily made by the physician or veterinarian (the "attending clinician").
  • the dosages may be varied depending upon the requirements of the patient, the severity of the condition being treated and the particular agent being employed.
  • a number of factors are considered by the attending clinician, including, but not limited to: the specific hype ⁇ lastic/neoplastic cell involved; pharmacodynamic characteristics of the particular agent and its mode and route of administration; the desired time course of treatment; the species of mammal; its size, age, and general health; the specific disease involved; the degree of or involvement or the severity of the disease; the response of the individual patient; the particular compound administered; the mode of administration; the bioavailability characteristics of the preparation administered; the dose regimen selected; the kind of concu ⁇ ent treatment; and other relevant circumstances.
  • the present invention shows that the bFGF level determines the tumor sensitivity to anticancer agents, thus indicating the use of bFGF level in tumors for therapeutic decision making, including the selection of a therapeutic modality, e.g., a particular anticancer treatment , or a dosage thereof, based on the level of FGF expression.
  • a therapeutic modality e.g., a particular anticancer treatment
  • a dosage thereof based on the level of FGF expression.
  • kits for carrying out the combined administration of the FGF antagonist/agonist with other therapeutic compounds comprises an FGF antagonist/agonist formulated in a pharmaceutical carrier, and at least one cytotoxic agent, formulated as appropriate, in one or more separate pharmaceutical preparations.
  • compositions which comprise a therapeutically-effective amount of at least one FGF antagonist/agonist, formulated together with or without at least one cytotoxic agent, in a pharmaceutically acceptable carrier(s).
  • the pharmaceutical compositions of the invention may be specially formulated for administration in solid or liquid form, including those adapted for the following: (a) oral administration, for example, drenches (aqueous or non-aqueous solutions or suspensions), tablets, boluses, powders, granules, pastes, mouthwash, hydrogels; (b) parenteral administration, for example, by subcutaneous, intramuscular or intravenous injection as, for example, a sterile solution or suspension; (c) intracavity administration, e.g., intraperitoneal instillation, intravesical (i.e., urinary bladder) instillation, intrathecal administration, (d) intraorgan administration, e.g., intraprostatical administration, (e) topical application, for example, as a cream, ointment or spray applied to the skin; (f) intravaginal or intrarectal administration, for example, as a pessary, cream, foam
  • FGF antagonist/agonist can be administered to the subject prior to, simultaneously with, or after the administration of the other cytotoxic agent(s). Further, several divided dosages, as well as staggered dosages, can be administered daily or sequentially, or the dose can be proportionally increased or decreased as indicated by the exigencies of the therapeutic situation.
  • phrases "pharmaceutically acceptable” is employed herein to refer to those FGF antagonist/agonist and/or cytotoxic agent, compositions containing such compounds, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically-acceptable carrier means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject chemical from one organ, or portion of the body, to another organ, or portion of the body.
  • a pharmaceutically-acceptable material such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject chemical from one organ, or portion of the body, to another organ, or portion of the body.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
  • materials which can serve as pharmaceutically-acceptable carriers include: (a) sugars, such as lactose, glucose and sucrose; (b) starches, such as com starch and potato starch; (c) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (d) powdered tragacanth; (e) malt; (f) gelatin; (g) talc; (h) excipients, such as cocoa butter and suppository waxes; (i) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (j) glycols, such as propylene glycol; (k) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (1) esters, such as ethyl oleate and ethyl laurate; (m) agar; (
  • wetting agents such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.
  • antioxidants examples include: (a) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (b) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (c) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
  • water soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like
  • oil-soluble antioxidants such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluen
  • compositions containing the FGF antagonist/agonist and/or cytotoxic agent of the present invention may conveniently be presented in unit dosage form.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration. Generally, out of one hundred per cent, this dosage will range from about 1 per cent to about ninety-nine percent of active ingredient, preferably from about 5 per cent to about 70 per cent, most preferably from about 10 per cent to about 30 per cent.
  • compositions of the invention suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, hydrogels, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia), and/or as mouthwashes and the like, each containing a predetermined amount of a FGF antagonist/agonist and/or cytotoxic agent as an active ingredient.
  • a compound may also be administered as a bolus, electuary or paste.
  • solid dosage forms of the invention for oral administration capsules, tablets, pills, dragees, powders, granules, hydrogels and the like
  • the active ingredient is mixed with one or more pharmaceutically-acceptable carriers, such as sodium citrate or dicalcium phosphate.
  • the pharmaceutical compositions may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.
  • the tablets, and other solid dosage forms of the pharmaceutical compositions of the present invention may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes, nanoparticles, hydrogels, and/or microspheres.
  • compositions may be sterilized by, for example, filtration through a bacteria-retaining filter, or by inco ⁇ orating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile injectable medium immediately before use.
  • These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner.
  • embedding compositions which can be used include polymeric substances and waxes.
  • the active ingredient can also be in micro- encapsulated form, if appropriate, with one or more of the above-described excipients.
  • Liquid dosage forms for oral administration of the FGF antagonist/agonist and/or cytotoxic agent of the invention include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents commonly used in the art, such as, for example, water
  • Suspensions in addition to the active FGF antagonist/agonist and/or cytotoxic agent may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • compositions of the invention for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more FGF antagonist/agonist and/or cytotoxic agent with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active agent.
  • suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active agent.
  • compositions of the present invention which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.
  • Dosage forms for the topical or transdermal administration of a FGF antagonist/agonist and/or cytotoxic agent include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants.
  • the active FGF antagonist/agonist and/or cytotoxic agent may be mixed under sterile conditions with a pharmaceutically-acceptable carrier, and with any preservatives, buffers, or propellants which may be required.
  • the ointments, pastes, creams and gels may contain, in addition to FGF antagonist/agonist and/or cytotoxic agent, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • the FGF antagonist/agonist and/or cytotoxic agent can be alternatively administered by aerosol. This is accomplished by preparing an aqueous aerosol, liposomal preparation or solid particles containing the compound. A nonaqueous (e.g., fluorocarbon propellant) suspension could be used. Sonic nebulizers are prefe ⁇ ed because they minimize exposing the agent to shear, which can result in degradation of the compound. Transdermal patches have the added advantage of providing controlled delivery of a FGF antagonist/agonist and/or cytotoxic agent to the body. Such dosage forms can be made by dissolving or dispersing the agent in the proper medium. Abso ⁇ tion enhancers can also be used to increase the flux of the peptidomimetic across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the peptidomimetic in a polymer matrix or gel.
  • compositions of this invention suitable for parenteral and/or intracavity administration comprise one or more FGF antagonist/agonist and/or cytotoxic agent in combination with one or more pharmaceutically-acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
  • the abso ⁇ tion of the drug in order to prolong the effect of a drug, it is desirable to slow the abso ⁇ tion of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amo ⁇ hous material having poor water solubility. The rate of abso ⁇ tion of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed abso ⁇ tion of a parenterally-administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.
  • Injectable or implantable depot forms are made by forming matrices (e.g., microcapsules, e.g., cylinders, e.g., discs) of FGF antagonist/agonist and/or cytotoxic agent in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue.
  • matrices e.g., microcapsules, e.g., cylinders, e.g., discs
  • biodegradable polymers such as polylactide-polyglycolide.
  • examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot
  • Depot formulations can also be administered directly to tumor-bearing organs and/or cavity, e.g., urinary bladder, e.g., intraperitoneal cavity, e.g., intrathecally to brain tissue, to achieve localized delivery of FGF antagonist/agonist and/or cytotoxic agent.
  • tumor-bearing organs and/or cavity e.g., urinary bladder, e.g., intraperitoneal cavity, e.g., intrathecally to brain tissue, to achieve localized delivery of FGF antagonist/agonist and/or cytotoxic agent.
  • administration is intended to include routes of introducing to a subject of the FGF antagonist/agonist and/or cytotoxic agent to perform their intended function.
  • routes of administration which can be used include injection (subcutaneous, intravenous, parenterally, intraperitoneally, intrathecally, etc.), intravesically (i.e., urinary bladder), intraprostatically, oral, inhalation, rectal and transdermal.
  • the pharmaceutical preparations are of course given by forms suitable for each administration route. For example, these preparations are administered in tablets or capsule form, by injection, inhalation, eye lotion, ointment, suppository, etc.
  • the injection can be bolus or can be bycontinuous infusion.
  • the FGF antagonist/agonist and/or cytotoxic agent can be administered alone, or in conjunction with either another agent as described above or with a pharmaceutically acceptable carrier, or both.
  • the FGF antagonist/agonist and/or cytotoxic agent can be administered prior to the administration of the other agent, simultaneously with the agent, or after the administration of the agent.
  • the FGF antagonist/agonist and/or cytotoxic agent can also be administered in a proform which is converted into its active metabolite, or more active metabolite in vivo.
  • parenteral administration and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticulare, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion.
  • systemic administration means the administration of a FGF antagonist/agonist and/or cytotoxic agent, such that it enters the patient's system and, thus, is subject to metabolism and other like processes.
  • the FGF antagonist/agonist and/or cytotoxic agent which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically-acceptable dosage forms by conventional methods known to those of skill in the art.
  • the present invention also concerns vaccines comprising an immunogenic amount of an FGF polypeptide, or a fragment thereof; an FGF receptor-derived polypeptide (FGFR polypeptide), or a fragment thereof, or a fragment of a proteoglycan (PG) that facilitates the binding of FGF to its receptors, dispersed in a physiologically acceptable, nontoxic vehicle, which amount is effective to immunize a subject, preferably a mammal, more preferably a human, against a neoplastic disease.
  • FGFR polypeptide FGF receptor-derived polypeptide
  • PG proteoglycan
  • the FGF or FGFR polypeptides, or the PG may be synthesized or prepared recombinantly or otherwise biologically, to comprise one or more amino acid sequences co ⁇ esponding to one or more epitopes of the FGF or FGFR polypeptides, or the PG either in monomeric or multimeric form. Those polypeptides may then be inco ⁇ orated into vaccines capable of inducing protective immunity. Techniques for enhancing the antigenicity of such polypeptides include inco ⁇ oration into a multimeric structure, binding to a highly immunogenic protein carrier, for example, keyhole limpet hemocyanin (KLH), or diphtheria toxoid, and administration in combination with adjuvants or any other enhancers of immune response.
  • KLH keyhole limpet hemocyanin
  • an amino acid sequence co ⁇ esponding to an epitope of an FGF polypeptide either in monomeric or multimeric form may also be obtained by chemical synthetic means or by purification from biological sources including genetically modified microorganisms or their culture media. See Lerner, "Synthetic Vaccines", Sci. Am. 248(2): 66-74 (1983).
  • the polypeptide may be combined in an amino acid sequence with other proteins/polypeptides including fragments of other proteins, as for example, when synthesized as a fusion protein, or linked to other antigenic or non-antigenic polypeptides of synthetic or biological origin.
  • an epitope of an FGF or FGFR polypeptides, or the PG will be understood to include the practical possibility that, in some instances, amino acid sequence variations of a naturally occurring protein or polypeptide may be antigenic and confer protective immunity against neoplastic disease and/or anti-tumorigenic effects.
  • Possible sequence variations include, without limitation, amino acid substitutions, extensions, deletions, truncations, inte ⁇ olations and combinations thereof.
  • Such variations fall within the contemplated scope of the invention provided the polypeptide containing these variations is immunogenic and antibodies elicited by such a polypeptide cross-react with naturally occurring FGF polypeptides to a sufficient extent to provide protective immunity and/or anti-tumorigenic activity when administered as a vaccine.
  • Such vaccine compositions can be combined with a physiologically acceptable medium, including immunologically acceptable diluents and carriers as well as commonly employed adjuvants such as Freund's Complete Adjuvant, saponin, alum, and the like.
  • Administration would be in immunologically effective amounts of the FGF or FGFR polypeptides, or the PG, preferably in quantities providing unit doses of from 0.01 to 10.0 micrograms of immunologically active FGF or FGFR polypeptide, or the PG per kilogram of the recipient's body weight.
  • Total protective doses may range from 0.1 to about 100 micrograms of antigen.
  • anti-idiotype antibodies to FGF or FGFR polypeptide, or the PG are also useful as vaccines and can be similarly formulated.
  • Mouse anti-human aFGF and bFGF monoclonal antibodies were obtained from Sigma; lactate dehydrogenase (LDH) assay, bromodeoxyuridine (BrdU) ELISA and human recombinant r-aFGF and r-bFGF from Boehringer Mannheim.
  • LDH lactate dehydrogenase
  • BrdU bromodeoxyuridine
  • the anti-aFGF antibody reacts with naturally occurring aFGF and human recombinant aFGF and does not cross-react with bFGF (Ichimori, Y. et al (1991) Biochem. Biophys. Res. Commun. 17 '5:291-297).
  • the anti-bFGF antibody was generated using the 18 kDa human r- bFGF, is specific for bFGF and does not cross-react with aFGF (Watanabe, H. et al. (1991) Biochem. Biophys. Res. Commun. 175:229-235).
  • r-aFGF and r-bFGF are monomeric peptides and are identical to the 14 kDa human aFGF or the 18 kDa human bFGF, except for the extra methionine at the amino terminus (Jaye, M. et al. (1986) Science, 233:541-545; Bohlen, P. et al. (1985) EERS Letters 18:177-181).
  • Human prostate PC3 tumor cells and rat normal intestinal epithelial I ⁇ C6 cells were obtained from the American Type Culture Collection.
  • the rat MAT-LyLu tumor cells and PC3-LN cells were obtained from Dr. John Isaacs and Dr. Joy Ware, respectively.
  • Paired primary and metastatic tumors were surgically removed from the same host. Fragments of the non-necrotic portions were cultured as histocultures. Single cell suspensions (>95% viability) were obtained by incubating tumor fragments ( ⁇ 1 g) with collagenase, EDTA, and trypsin. Histocultures (tumor fragments of ⁇ 1 mm 3 ) and monolayers were cultured in medium supplemented with 9% heat-inactivated fetal bovine serum (FBS) (Yen, W.C. et al. (1996; Pharm. Res. 13:1305-1312).
  • FBS heat-inactivated fetal bovine serum
  • CM Conditioned medium
  • proteins were transfe ⁇ ed from polyacrylamide gel to a nitrocellulose filter by electrophoresis, followed by sequential incubation with 5% nonfat dry milk in 100 mM Tris/150 mM NaCl/0.1% Tween 20 (pH 7.6), and 5 microgram/ml aFGF or bFGF monoclonal antibody.
  • the antibody-immunoreactive band was visualized using chemiluminescence blotting.
  • the levels of aFGF and bFGF in the concentrated CM were quantified by comparing the intensity of their bands on Western blots to the intensity of the bands derived from standard curve samples of r-aFGF and r-bFGF.
  • the standard curves were linear between 3 and 100 ng r-aFGF and between 1 and 160 ng r-bFGF.
  • CM Pretreatment with CM and recombinant r-bFGF/r-aFGF
  • CM Pretreatment with CM and recombinant r-bFGF/r-aFGF
  • r-aFGF ad/or r-bFGF-containing medium, supplemented with 1% FBS (final protein concentration adjusted to 2 mg/ml).
  • FBS final protein concentration adjusted to 2 mg/ml
  • aFGF or bFGF was immunoprecipitated with their respective monoclonal antibody (1 microgram/ml), in the presence of Protein G PLUS/protein A agarose (Oncogene). This procedure reduced the bFGF level in the original CM to below the detection limit of 5 pg/ml by the ELISA (Oncogene), and the aFGF level in the concentrated CM to below the detection limit by Western blotting.
  • This example describes three studies showing (a) a novel epigenetic mechanism of broad spectrum chemoresistance that is mediated by extracellular factors present in solid and metastatic tumors, and (b) loss of these factors upon removing tumors from metastatic sites and/or disrupting the tumor microenvironment.
  • the results supporting these conclusions are provided in Table 1 below.
  • the studies were performed using anticancer agents that have diverse structures and action mechanism, including paclitaxel (an antimicrotubule agent), doxorubicin (a topoisomerase II inhibitor) and 5- fluorouracil (an antimetabolite). Similar results were found for all three drugs.
  • the first study compared the chemosensitivity in histocultures of rat primary and metastatic tumors, where the heterogeneous cell types and the 3-dimensional structure of solid tumors are maintained, to the chemosensitivity in the co ⁇ esponding monolayer cultures of cells obtained by trypsin-disaggregation of the same tumors.
  • the 2- to 40-fold lower chemosensitivity in histocultures indicates that the unique environment in solid tumors played a role in chemoresistance.
  • the second study shows that histocultures and early monolayer cultures of lung metastases were more resistant than lymph node metastases which in turn were more resistant than subcutaneous primary tumors, indicating that tumor location determined the extent of chemoresistance.
  • Table 1 Loss of drug resistance, aFGF and bFGF upon disaggregation of metastatic tumors and upon passaging in monolayer cultures.
  • Paired rat primary and metastatic tumors were cultured as histocultures or monolayers.
  • tumors were treated with paclitaxel for 24 hours (12 pairs of primary and lymph node tumors and 2 pairs of primary and lung tumors), and with doxorubicin (3 pairs of primary and lymph node tumors) and 5-fluorouracil (2 pairs of primary and lung tumors) for 96 hours.
  • monolayers cells were treated with drugs for 96 hours, and drug effect was measured as inhibition of bromodeoxyuridine inco ⁇ oration. NM, not measured.
  • EXAMPLE II Induction of resistance to anticancer agents due to extracellular factors in solid and metastatic tumors
  • This example describes the experiments confirming the induction of chemoresistance by extracellular factors in metastatic tumors, and indicating a progressive loss of these factors upon passaging metastatic tumor cells in monolayers.
  • CM from rat tumor cultures was collected and evaluated for its effect on chemosensitivity. This and subsequent experiments were performed using rat primary tumor cells and human prostate tumor cells. Qualitatively similar results were obtained for both cell types, indicating the general nature of the observations ( Figure 1). CM derived from histocultures and early monolayer cultures of metastases (refe ⁇ ed to as active metastatic tumor CM) induced a 3- to 10-fold resistance to drugs (p ⁇ 0.05), whereas CM from late monolayer cultures of metastatic tumors and CM from primary tumors (refe ⁇ ed to as inactive CM) had no effect.
  • EXAMPLE III Identification of bFGF and aFGF as some of the extracellular factors that induce resistance to anticancer agents
  • CM conditioned media
  • the active CM showed 2- to 7-fold higher concentrations of two proteins, which were identified by immunoblotting as aFGF (approximately 14 kDa) and bFGF (approximately 18 kDa).
  • aFGF approximately 14 kDa
  • bFGF approximately 18 kDa
  • This example describes six studies which collectively show that (a) extracellular aFGF and bFGF are involved in the induction and maintenance of broad spectrum resistance to anticancer agents; and (b) bFGF but not aFGF was required to induce resistance whereas aFGF amplified the bFGF effect.
  • the first study used specific inhibitors of extracellular aFGF and bFGF, i.e., monoclonal antibodies (Figure 2A).
  • aFGF or bFGF did not alter drug activity, indicating no effect of the antibodies on the baseline chemosensitivity.
  • the bFGF antibody produced a concentration-dependent reversal of the CM-induced chemoresistance, with complete reversal by 5 microgram/ml antibody, whereas a nonspecific antibody (i.e., non-immune rabbit IgG) had no effect.
  • aFGF For aFGF, the antibody treatment partially reversed the CM-induced resistance; the maximum reversal was about 60% that was obtained with 1 microgram/ml antibody with no significant additional reversal at a 5 -fold higher concentration.
  • the third study determined whether aFGF and/or bFGF are required for the resistance.
  • Endogenous aFGF and/or bFGF were removed from the active CM by immunoprecipitation, and then reconstituted the CM using recombinant proteins.
  • Combinations of r-aFGF/r-bFGF at concentrations as the endogenous proteins in the CM i.e., between 0.16 and 0.32 ng/ml r-aFGF plus 1 ng/ml r-bFGF
  • CM i.e., between 0.16 and 0.32 ng/ml r-aFGF plus 1 ng/ml r-bFGF
  • the chemoresistance observed above was measured primarily as antiproliferative drug effect.
  • the sixth study measured the cell kill effect of drugs. Similar findings were observed, i.e., the active metastatic tumor CM and r-bFGF induced resistance to cell kill by drugs, whereas r-aFGF did not induce resistance but enhanced the effect of r-bFGF ( Figure 4). Therefore, the FGF-induced resistance applies to both antiproliferative and cell kill effects.
  • EXAMPLE V In vitro data on the enhancement of sensitivity of tumor cells to anticancer agents using inhibitors of aFGF and bFGF
  • This example shows that inhibitors of aFGF and/or bFGF enhanced the tumor sensitivity to anticancer agents.
  • Figure 2A shows that inhibition of aFGF and/or bFGF using their respective monoclonal antibodies enhanced the cytotoxicity of paclitaxel, doxorubicin and 5- fluorouracil against human and rodent tumor cells.
  • Figure 2B shows the same findings when anti-aFGF monoclonal antibody and/or anti-bFGF monoclonal antibody were used to remove aFGF and/or bFGF from the cell culture medium.
  • Another inhibitor of aFGF and bFGF i.e., suramin, also reverses the FGF-induced resistance and enhances the activity of at least 58 anticancer agents under in vitro and/or in vivo conditions (see also Example VIII).
  • Suramin is a negatively charged molecule that inhibits the action of multiple growth factors including aFGF and bFGF (Middaugh, C.R. et al. (1992) Biochem. 31 :9016-9024).
  • Figure 5 shows the results of in vitro experiments demonstrating that addition of suramin produced a concentration-dependent reversal of the CM-induced resistance to paclitaxel, doxorubicin and 5-fluorouracil in rat MAT-LyLu tumor cells, human prostate PC3 tumor cells, and a metastatic subline of PC3, i.e., PC3-LN cells. Complete reversal was attained at between 10 to 15 ⁇ M suramin, which, as shown in Example VI, had no antitumor effect when used alone.
  • FIG. 6 shows the concentration-response curves of suramin in the tumor cell lines used in the studies described in Examples V and VII.
  • the concentration of suramin, when used alone, required to result in 50% reduction of bromodeoxyuridine inco ⁇ oration (i.e., IC50) were 235 micromolar in rat MAT-LyLu tumor cells, 93 micromolar in human prostate PC3 tumor cells, and 98 micromolar in human prostate PC3-LN tumor cells.
  • IC 50 values are between 6- to 200-fold higher than its concentration needed to fully reverse the FGF-induced resistance (between 1 to 15 micromolar, see Examples V and VII and Table 2).
  • suramin did not produce measurable cytotoxicity (i.e., ⁇ 10% reduction in cell number and/or proliferation index).
  • the enhancement of the antitumor effect of anticancer drugs by suramin occurs at nontoxic concentrations of suramin.
  • EXAMPLE VII Broad spectrum of anticancer agents exhibit aFGF/bFGF- induced resistance that is reversed by suramin
  • EXAMPLE VIII aFGF bFGF-induced resistance to anticancer agents is detected in multiple cancer cell lines
  • the drug effect was determined by the inhibition of BrdU incorporation.
  • the drug effect was measured by the MTT assay, which measures the number of metabolically active cells (Alley, M.C. et al (1988) Cancer Res 48:589-601).
  • r-aFGF amplified the r-bFGF effect by more than 50-fold (e.g. 1 ng/ml r-aFGF plus 1 ng/ml r-bFGF induced greater resistance than 50 ng/ml r-bFGF alone).
  • CCRF-CEM human leukemic cell line
  • r-aFGF 1 ng/ml
  • r-bFGF 3 ng/ml
  • FGF-induced resistance occurs in multiple human and rodent tumor cell lines.
  • the BC 19 cell line is a subline of MCF7 transfected with m rl. * Indicates p ⁇ 0.05 compared to control.
  • This example describes four studies that provide additional data on the enhancement of the antitumor effect of paclitaxel and doxorubicin by suramin in immunodeficient mice bearing several human xenograft tumors.
  • mice Male Balb/c nu/nu mice, 6-8 weeks old. Animal care was in accordance with the guidelines at the Ohio State University.
  • human tumor models were used, including (a) lung metastases derived from intravenous injection of human PC3-LN tumor cells, (b) lung tumors derived from direct injection of human PC3 cells in lungs, and (c) subcutaneously implanted PC3 tumors. Four studies were performed. In all studies, drug treatment was administered intravenously via a tail vein, after the tumor was established. The first and second studies used the intravenous PC3-LN lung metastasis model.
  • human PC3-LN cells (10 6 cells in 0.1 ml physiologic saline) were injected intravenously, via a tail vein, into immunodeficient mice. This procedure results in metastasis to lungs in 100% of the animals (Ware, J.L. et al. (1985) Exp. Cell Biol. 53:163-169). After 4 weeks, tumor establishment was determined by visual examination of the lungs of two randomly selected animals, and drug treatment in the remaining animals was initiated when these two animals showed at least 5 tumor nodules of ⁇ 1 mm diameter.
  • mice received intravenous injection (over 1 min) of 200 ⁇ l of either physiologic saline or a saline solution delivering a chemotherapeutic, 10 mg/kg suramin, or a combination of chemotherapeutic and suramin.
  • the chemotherapeutic was doxorubicin (5 mg/kg) for the first study and paclitaxel (Taxol, 15 mg/kg) for the second study. Treatment was given twice weekly for three weeks. Preliminary pharmacokinetic data in rodents indicate that these doses would result in plasma concentrations of approximately 10 nM for doxorubicin, 5-7 nM for paclitaxel and 10 micromolar for suramin at 72 hours.
  • the suramin concentration was sufficient to reverse the FGF- induced chemoresistance.
  • the doxorubicin and paclitaxel concentrations are near or above their IC 5 oin the monolayer cultures of PC3-LN cells (i.e., 17 nM for doxorubicin and 8 nM for paclitaxel).
  • the selected suramin dose has no in vivo antitumor activity against other mouse tumors (Chahinian, A.P. et al. (1998) J. Surg. Oncol. 67 : 104- 111 ; Shin, R., et al. (1997) Scand. J. Gastroenterol 32:824-828).
  • mice were euthanized and their lungs removed, fixed in Bouin's solution to visualize tumor nodules, and then processed for histologic evaluation. Histologic sections (5 micron thickness) at a depth of between 200-300 ⁇ m from the ventral surface and containing all 5 lobes of the lungs were obtained. The lung surface area (counted as number of pixel) occupied by the tumor was calculated as a fraction of the total lung area, using Adobe PhotoShop. The number of tumor cells in residual tumors and the fraction of apoptotic cells in each tumor were also determined microscopically.
  • apoptotic cells disappear over time, a second measure of the extent of apoptosis was the density of non-apoptotic cells in the residual tumors. This was determined by counting the number of non-apoptotic tumor cells in randomly selected microscopic fields at 400 x magnification. On average, ten fields per animal, or >1,500 cells in the control and suramin groups and >600 cells in the doxorubicin group were counted. In the case of combination therapy where fewer than 5 tumor nodules remained per animal, all residual cells (between 20 to 600 cells per animal) were counted.
  • the average pretreatment weights for the four groups ranged from 21 g to 22 g. Animals that did not show visible tumors on the lung surface nor microscopic lesions in 5 random histologic sections are considered tumor-free. Mean ⁇ SD. *p ⁇ 0.05 compared to the control and suramin groups. t p ⁇ 0.05 compared to all other groups.
  • I l l Table 5 Suramin enhances the in vivo antitumor effect of paclitaxel in mice bearing human lung metastases established by intravenous injection of tumor cells.
  • the average pretreatment weights for the four groups ranged from 21 g to 22 g. Mean ⁇ SD. *p ⁇ 0.05 compared to the control and suramin groups. t p ⁇ 0.05 compared to all other groups.
  • the third study shows enhancement of doxorubicin activity in lung tumors established by intra-organ tumor cell injection.
  • PC3 cells were directly injected into the lungs of immunodeficient mice (2.5xl0 6 cells per lung). Tumor establishment (on day 5) was verified by visual examination of a randomly selected animal. The remaining animals were given intravenous bolus injection of doxorubicin (three doses of 10 mg/kg each given on day 5, 8 and 11) alone, suramin alone (three doses of 20 mg/kg), or a combination of both drugs. Animals were euthanized on day 12. Lungs were removed and examined visually for residual tumors. Histologic sections were obtained and examined microscopically, as described for the first two studies.
  • the fourth study shows that suramin enhanced the antitumor effect of doxorubicin in subcutaneous bulky tumors.
  • PC3 cells (5xl0 6 cells) were injected subcutaneously.
  • Drug treatments suramin 10 mg/kg, doxorubicin 5 mg/kg, or a combination of both drugs, given twice weekly for 3 weeks.
  • the average tumor size was 150 mg at the time of first treatment.
  • Figure 7 A shows (a) a 3- to 4-fold increase of tumor size in the untreated control group and suramin group, indicating that suramin alone had no antitumor activity, (b) doxorubicin alone reduced the tumor growth but did not cause tumor regression, (c) doxorubicin + suramin combination reduced the tumor growth during the first week, followed by tumor regression until the end of the 3-week treatment.
  • Results on the animal body weight ( Figure 7B) indicate (a) suramin alone did not cause body weight loss, (b) doxorubicin produced a 17% body weight loss, and (c) addition of suramin to doxorubicin did not enhance body weight loss.
  • EXAMPLE X Relationship between aFGF and bFGF levels in tumor, tumor size, and tumor sensitivity to anticancer agents
  • This example demonstrates a correlation between aFGF and bFGF level in tumor and tumor size, and a correlation between bFGF level in tumor and tumor sensitivity to anticancer agents.
  • the first study examined the effect of the presence of tumor cells on the extracellular bFGF levels in tissues. Briefly, human prostate PC3 tumor cells were implanted intradermally or directly into the lungs of immunodeficient mice. Tumors were removed from animals and -10 mg were cultured as histocultures. The culture medium was collected at 24 hours and analyzed for bFGF.
  • the bFGF level in PC3 cells was 2 pg/10 6 cells (approximately 2 pg/mg), 0.13 pg/mg in normal lung tissue, 0.62 pg/mg in normal skin tissue, 3.2 pg/mg in PC3-containing lung tumor, and 11.3 pg/mg in PC3-containing skin tumor.
  • the bFGF levels in tumor-bearing tissues exceeded the sum of bFGF levels in tumor cells and normal tissues, indicating that the presence of tumor cells enhanced bFGF level.
  • the enhancement was 25-fold in lungs and 18-fold in skin.
  • the mouse brain contained 0.027 pg bFGF per mg tissue and the liver contained 0.636 pg bFGF per mg tissue. The results indicate (a) different bFGF levels in different tissues, and (b) presence of tumor cells significantly enhanced the bFGF levels.
  • the second study examined the relationship between the extracellular bFGF level and tumor size.
  • Human prostate PC3 tumor cells were implanted subcutaneously in immunodeficient mice. Subcutaneous tumor were removed from host animals after 1 to 7 weeks, and -200 mg were cultured as histocultures. The culture medium was collected at 24 hours and analyzed for bFGF concentration by ELISA.
  • the bFGF level increased with time, from 27 pg/ml in week 1 tumor, to 56 pg/ml in week 2 tumor, to 77 pg/ml in week 3 tumor, and reached a plateau level of between 80 to 87 pg/ml between week 4 and week 7 tumors.
  • the differences between the bFGF levels in weeks 1 , 2 and 3 are statistically significant. This indicates a positive correlation between bFGF level and tumor size.
  • the third study examined the relationship between the levels of aFGF and bFGF in human tumors and tumor sensitivity to anticancer agents.
  • This study was performed using 96 tumors obtained from human patients, including include bladder, breast, head and neck, ovarian and prostate tumors. Paclitaxel was used as the model drug. Briefly, histocultures of bladder tumors were treated with paclitaxel for 2 hours and the remaining tumors for 96 hours. The drug-induced antiproliferation was measured by inhibition of DNA precursor inco ⁇ oration, and apoptosis was measured by morphological changes and by labeling of nicked DNA.
  • aFGF, bFGF and other proteins known to contribute to drug resistance i.e. the mdrl p-glycoprotein, p53 and bcl-2) were detected by immunohistochemical staining.
  • bFGF expression was the best indicator of resistance to the antiproliferative effect and the overall cytotoxicity (i.e., antiproliferation plus apoptosis) of paclitaxel in tumors obtained from human patients (see Table 6 set forth below). Addition of aFGF to bFGF improves the correlation (Table 6).
  • aFGF and bFGF in tumors are determined by the location of the tumor and the size of the tumor, and that the aFGF/bFGF level, in turn, determines the tumor sensitivity to anticancer agents.
  • the varying FGF level in tumors indicates the use of FGF level for making therapeutic decision for individual patients.
  • the best prognostic indicators are those that give the highest r 2 values as well as the lowest AIC values.
  • Statistical analysis was performed using linear regression analysis using REG software. A p value of less than 0.05 is statistically significant.
  • FGF-induced resistance to anticancer agents is a novel mechanism that is different from the previously reported mechanisms of chemoresistance that involve an enhancement of drug efflux and altered cell proliferation.
  • Human prostate PC3 tumor cells and rat MAT-LyLu tumor cells were treated with 3 H-paclitaxel (1 nM for human PC3 cells and 10 nM for rat tumor cells), 14 C-doxorubicin (50 and 100 nM, respectively), and 3 H-5-fluorouracil (500 nM for both cells) (Au, J.L.-S. et al. (1998) supra), with or without with the active CM collected from histocultures of lung metastases or with r-bFGF.
  • the extracellular drug concentrations were selected based on their IC 50 (50% inhibitory concentration) in human and rat cells.
  • the plateau intracellular drug concentrations which were attained at 4 hours for all three drugs in PC3 cells, and 1 (doxorubicin and 5-fluorouracil) and 4 hours (paclitaxel) in rat cells, were measured.
  • CM or r-bFGF Treatment with the CM or r-bFGF also did not alter the doubling time of exponentially growing cells, which remained unchanged at 17 and 24 hours for rat and PC3 tumor cells, respectively. Hence, the FGF-induced chemoresistance is not due to altered cell proliferation.
  • the findings described herein demonstrate (a) a novel epigenetic mechanism by which cancer cells utilize the unique microenvironment of solid tumors and metastases to elude cytotoxic insult, (b) establish an important role of extracellular growth factors in tumor sensitivity to chemotherapy, and (c) indicate a new treatment paradigm using combinations of chemotherapy with aFGF/bFGF inhibitors.
  • This example shows that treatment of non-cancerous intestinal epithelium with aFGF and bFGF protects these cells from the toxicity of anticancer agents, such as paclitaxel and doxorubicin.
  • IEC6 normal rat intestinal epithelial cells
  • IEC6 normal rat intestinal epithelial cells
  • Cell culture assay experiments can be performed in the rat prostate MAT-LyLu tumor cells, and the human prostate PC3 tumor cells or its metastatic subline PC3-LN cells. If the requirements of the invention are met in any of the three cell cells, the choice and the dosage of the FGF antagonist is suitable for use in the invention. Preferably the PC3 cells are used.
  • Human prostate PC3 tumor cells can be obtained from the American Type Culture Collection.
  • the rat MAT-LyLu tumor cells and PC3-LN cells were obtained from Dr. John Isaacs (Johns Hopkins University) and Dr. Joy Ware (University of Virginia), respectively.
  • the doubling time of PC3 and PC3-LN cells is approximately 24-hour, the doubling time ofMAT- MAT-LyLu cells is approximately 15-17 hr. All three cell lines should be cultured as monolayers in a humidified environment containing 5% CO2 and 95% air, at 37°C.
  • PC 3 cells and PC3-LN cells should be maintained in RPMI 1640 medium supplemented with 9% heat-inactivated fetal bovine serum, 2 mM 1- glutamine, 100 ⁇ g/ml gentamicin, and 95 ⁇ g/ml cefotaxime.
  • MAT-LyLu cells should be maintained in RPMI 1640 medium containing 100 ⁇ g/ml gentamicin, 95 ⁇ g/ml cefotaxime and 9% heat-inactivated fetal bovine serum.
  • Cells are harvested from subconfluent cultures using trypsin and resuspended in fresh medium before plating.
  • Cells with > 90% viability, as determined by trypan blue exclusion, are used to evaluate the cytotoxicity of an FGF antagonist, e.g., suramin.
  • FGF antagonist e.g., suramin.
  • Cells are plated in 96 well microtiter plates at a density such that confluence would not be achieved at the end of the drug treatment period. Cells are allowed to attach to the plate surface by growing in drug-free medium for 20 to 24 hr. Afterward, cells are incubated with the FGF antagonist (one example used 0.2 ml of suramin)-containing culture medium, at concentrations spanning at least 4 log scales.
  • the drug effect should be measured as inhibition of BrdU inco ⁇ oration, e.g., according to the Cell Proliferation ELISA BrdU (Boehringer Mannheim).
  • EXAMPLE XVI Inhibition of FGF binding to FGF receptors This example describes the inhibition of the binding of FGF to FGF receptors by FGF antagonists.
  • FGF antagonists i.e., suramin, heparin, low molecular weight heparin, heparan sulfate
  • FGF receptors FGF receptors in human prostate cancer PC3 cells and human breast cancer MCF7 cells
  • FGF antagonists i.e., suramin, heparin, low molecular weight heparin, heparan sulfate
  • FGF receptors in human prostate cancer PC3 cells and human breast cancer MCF7 cells were studied. Briefly, cells were plated at 1 X 10 5 cells per 16 mm well on 24-well plates and were allowed to settle for 48 hours before binding experiment. On the day of the experiment, cells were incubated for 2 hours at 37°C in serum-free DMEM supplemented with 0.15%) gelatin. Cells were then washed twice with ice-cold phosphate buffered saline.
  • Differential display was used to identify the down-stream genes that are involved in the bFGF-induced multidrug resistance. These genes were identified by comparing the effect of bFGF on the expression of these genes in a cell line that demonstrates bFGF-induced chemoresistance (i.e. human prostate cancer PC3 cells) with that in a cell line which does not show bFGF-induced chemoresistance (i.e. human head and neck cancer FaDu cells). By this method, the genes that were altered by bFGF treatment in PC3 cells but not in FaDu cells were identified. The identified genes are likely to be associated with the bFGF-induced multidrug resistance.
  • bFGF-induced chemoresistance i.e. human prostate cancer PC3 cells
  • FaDu cells i.e. human head and neck cancer FaDu cells

Abstract

Methods and compositions for modulating the FGF effect on the sensitivity of malignant and normal cells to anticancer agents are provided. In particular, methods and compostions for inhibiting FGF-induced resistance to a broad spectrum of anticancer agents in solid and soft-tissue tumors, metastatic lesions, leukemia and lymphoma are provided. Preferably, the compositions include at least one FGF inhibitor in combination with a cytotoxic agents, e.g., antimicrotubule agents, topoisomerase I inhibitors, topoisomerase II inhibitors, antimetabolites, mitotic inhibitors, alkylating agents, intercalating agents, agents capable of interfering with a signal transduction pathway (e.g., g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an agent that promote apoptosis and/or necrosis, an interferon, an interleukin, a tumor necrosis factor, and radiation. In other embodiments, methods and composition for protecting a cell in a subject, from one or more of killing, inhibition of growth or division or other damage caused, e.g., by a cytotoxic agent, are provided. Preferably, the method includes: administering, to the subject, an effective amount of at least one FGF agonist, thereby treating the cell, e.g., protecting or reducing the damage to the dividing cell from said cytotoxic agent.

Description

METHODS AND COMPOSITIONS FOR MODULATING CELL PROLIFERATION AND CELL DEATH
Related Applications This application claims priority to U.S. Provisional Application No. 60/172,031 , entitled "Modulation of Cell Proliferation" filed on December 23, 1999; U.S. Provisional Application No. 60/137,345, entitled "Methods to Diagnose, Prevent, and Overcome FGF-Dependent or FGF-Induced Chemoresistance in Metastatic Sites" filed on June 3, 1999; U.S. Provisional Application No. 60/165,983, entitled "Genes and Gene Products Conferring Reversible Broad Spectrum Anticancer Drug Resistance and Methods of Inhibiting the Same" filed on November 17, 1999; and U.S. Provisional Application No. 60/187,445, entitled "Methods and Compositions for Modulating Cell Proliferation and Cell Death" filed on March 7, 2000, the contents of all of which are hereby incorporated by reference.
Background of the Invention
Resistance of tumor cells to cancer therapy, limited efficacy of cancer therapy in metastatic disease, and undesired host toxicity of cancer therapy are three significant challenges in patient management. A common resistance mechanism to chemotherapy observed in preclinical studies is the overexpression of drug efflux proteins (Lum, B.L. et al. (1993) Cancer 72, 3502-3514; Barrand, M.A. et al. (1997) Gen. Pharmacol. 28, 639-645; Fidler, IJ. (1999) Cancer Chemother. Pharmacol. 43:S3-S10.). However, at least some clinical studies show that inhibition of the drug efflux proteins does not significantly improve the effectiveness of chemotherapy in patients (Ferry, D.R., et al. (1996) Ewr. J. Cancer 32:1070-1081; Broxterman, H.J., et al. (1996) Eur. J. Cancer. 32:1024-1033), suggesting the existence of other resistance mechanisms.
Cancer therapy, such as chemotherapy and radiation, targets proliferating cells and thereby causes undesired toxicity to normal host tissues that undergo continuous renewal, including the hematopoietic cells, cells in the lining of the gastrointestinal tract, and hair follicles. Bone marrow suppression induced by cancer therapy is, at least in part, overcome by the use of hematopoietic growth factors, including erythropoietin, granulocytes colony-stimulating factor, and granulocyte-macrophage colony- stimulating factor (Gabrilove, J.L. and Goldie, D.W. (1993) In: Cancer, Principles & Practice of Oncology (eds. DeVita, V.T. et al, J.B. Lippincott Co., Philadelphia). On the other hand, no treatment is available to overcome the gastrointestinal toxicity and alopecia induced by anticancer agents. Therefore, there exists a need to identify new mechanisms by which tumor and normal cells elude cytotoxicity of anticancer agents, to identify methods and agents to overcome such resistance in tumor cells, and to utilize these resistance mechanisms to protect normal host tissues from the undesired toxicity of cancer therapy.
Summary of the Invention
The invention is based, at least in part, on the elucidation of the role played by basic Fibroblast Growth Factor (bFGF) in the induction of broad spectrum resistance to anticancer agents in a number of tumors and metastatic lesions, and the role played by acidic FGF (aFGF) in amplifying the bFGF-induced resistance. Inhibitors of aFGF/bFGF enhance the in vitro and in vivo activity of anticancer agents, and result in shrinkage and eradication of human xenograft tumors including lung metastasis and subcutaneous tumors in mice. Methods of the invention use FGF antagonists to potentiate the antitumor effect of anticancer agents. FGF agonists (e.g., aFGF, e.g., bFGF) reduce the cytotoxicity of anticancer agents to normal noncancerous intestinal epithelial cells. Methods of the invention use FGF agonists to protect normal cells from the cytotoxic effects of anticancer agents.
Accordingly, in general, the invention features, a method of inhibiting unwanted cell growth or division, e.g., reducing or inhibiting the proliferation of, or enhancing the killing of, a cell, e.g., a hyperproliferative cell, e.g., a malignant cell or a benign hyperproliferative cell. The method includes: contacting the cell with at least one cytotoxic agent, (e.g., a cytostatic agent, e.g., an agent that causes cell death) and at least one FGF antagonist, in an amount, which together, is effective to reduce or inhibit the proliferation of the cell, or induce cell killing. Preferably, the unwanted cell is the cell of an established tumor.
In another aspect, the invention features a method of improving the efficacy of an agent, e.g., a cytotoxic agent, in a subject. The method includes: administering to the subject at least one agent, e.g., a cytotoxic agent; administering to the subject at least one FGF antagonist.
The FGF antagonist enhances the efficacy of the agent, e.g., a cytotoxic agent, relative to the effect of the cytotoxic agent in the absence of the FGF antagonist. In a preferred embodiment, the FGF antagonist improves the efficacy of the cytotoxic agent against a cancer, e.g., an established tumor.
In another aspect, the invention features, a method of inhibiting unwanted cell growth or division, or inducing the killing of an unwanted cell (e.g., a hyperproliferative cell), e.g., a cell of an established tumor or a benign hyperproliferative cell, in a subject. The method can be used to treat or prevent, in a subject, a disorder characterized by unwanted cell growth or division. The method includes: administering to the subject at least one cytotoxic agent, (e.g., a cytostatic agent, an agent that causes cell death), and at least one FGF antagonist, in an amount, which together, is effective (e.g., therapeutically or prophylactically) to reduce or inhibit the growth or division of, or induce the killing of, the unwanted cell. Preferably, the unwanted cell is the cell of an established tumor.
In a preferred embodiment, the FGF antagonist inhibits or reduces the FGF- induced resistance to a broad spectrum of cytotoxic agents, i.e., agents with diverse structures and mechanisms of action, including but not limited to, antimicrotubule agents, topoisomerase I inhibitors, topoisomerase II inhibitors, antimetabohtes, mitotic inhibitors, alkylating agents, intercalating agents, agents capable of interfering with a signal transduction pathway (e.g., protein kinase C inhibitors, e.g., anti-hormones, e.g., antibodies against growth factor receptors), agents that promote apoptosis and/or necrosis, interferons, interleukins, tumor necrosis factors, and radiation.
In a preferred embodiment, the FGF antagonist comprises an inhibitor of bFGF. In a preferred embodiment, the FGF antagonist comprises an inhibitor of aFGF.
In a preferred embodiment, the FGF antagonist includes at least one bFGF inhibitor and at least one aFGF inhibitor.
In a preferred embodiment, the aFGF inhibitor potentiates the action of the bFGF inhibitor. In a preferred embodiment, the FGF antagonist acts extracellularly, e.g., inhibits the binding of an FGF molecule to its receptor.
In a preferred embodiment, the FGF antagonist acts intracellularly, e.g., interacts with the intracellular domain of the FGF receptor, inhibits the intracellular effects of FGF.
In a preferred embodiment, the FGF antagonist: is capable of binding to an FGF molecule or an FGF receptor; blocks the binding of FGF to a receptor; blocks the interaction of FGF with molecules that facilitate the binding of FGF to a receptor; and/or down regulates FGF receptor action. Preferably, the FGF molecule is bFGF and/or aFGF.
In a preferred embodiment, the FGF antagonist is other than suramin. In a preferred embodiment, the FGF antagonist is other than an antibody, e.g., an antibody against FGF or an FGF receptor.
In a preferred embodiment, the FGF antagonist inhibits or reverses the resistance to anticancer drugs induced by FGF (e.g. aFGF and/or bFGF) in cultured tumor cells under in vitro conditions. The determination of effect on cultured cells can be determined using the system described in Example XV.
In a preferred embodiment, the FGF antagonist improves the efficacy of an agent, e.g., a cytotoxic agent, in the subject, relative to the effect of the cytotoxic agent in the absence of the FGF antagonist. Preferably, the FGF antagonist improves the efficacy of the cytotoxic agent against an established tumor.
In a preferred embodiment, the FGF antagonist is present in an amount sufficient to block FGF (e.g., bFGF and/or aFGF) action, but is not sufficient to cause one or more of: (i) significant inhibition of cell proliferation; (ii) significant cell death in human and/or animal tumor cells, (iii) a measurable antitumor effect in a subject, e.g., a human subject; and/or (iv) significant cell cycle arrest. The determination of effect on cultured cells can be determined with the system described in Example XV. In a preferred embodiment, the FGF antagonist is administered at levels such that the plasma concentration of the FGF antagonist that is present when the cytotoxic agent is present in plasma at pharmacologically active concentration does not result in one or more of: (i) significant cell cycle arrest, (ii) significant cell death, or (iii) significant inhibition of cell growth, e.g., the concentration in plasma is of a level that if the same concentration of FGF antagonist is provided in cultured cells at least 10, more preferably at least 25, more preferably at least 50, more preferably at least 70, more preferably at least 80, more preferably at least 90, and most preferably at least 99% of the treated cultured cells continue to be involved in one or more of: (i) cycling cells continue to progress through the cell cycle, (ii) cells remain viable, or (iii) cells remain capable of proliferating, following treatment with the FGF antagonist. The determination of effect on cultured cells should be determined with the system described in Example XV.
In a preferred embodiment, the FGF antagonist is administered at levels that, although not resulting in significant cell cycle arrest, significant cell death, or significant inhibition of cell growth as described above, sensitizes the tumor cells to treatment with cytotoxic agents. Preferably the levels of FGF antagonist result in a significant inhibition of FGF-mediated chemoresistance, e.g., the resistance of tumor cells to cytotoxic agents that is mediated by FGF. Preferably the FGF antagonist inhibits at least 10, more preferably, at least 25, more preferably at least, 50, more preferably at least 70, more preferably at least 80, more preferably at least 90, and most preferably at least 99% of the FGF-mediated chemoresistance. The determination of effect on cultured cells can be determined with the system described in Example XV. In a preferred embodiment, the time period over which the FGF antagonist is administered or over which the FGF antagonist is maintained at a therapeutic level, e.g., at a plasma concentration that is sufficient to enhance the cytotoxic effect of the cytotoxic agent, is less than 180, more preferably less than 90, more preferably less than 60, and most preferably less than 30 days.
In a preferred embodiment, the time period over which the FGF antagonist is administered or over which the FGF antagonist is maintained at a therapeutic level, e.g., at a plasma concentration that is sufficient to enhance the cytotoxic effect of the cytotoxic agent does not begin substantially earlier or end substantial later than the period over which the cytotoxic agent is administered or over which the cytotoxic agent is maintained at a therapeutic level.
In a preferred embodiment, the time period over which the FGF antagonist is administered or over which the FGF antagonist is maintained at a therapeutic level, e.g., at a plasma concentration that is sufficient to enhance the cytotoxic effect of the cytotoxic agent ends less than 180, more preferably less than 90, more preferably less than 60, and most preferably less than 30 days after the last day on which the cytotoxic agent is administered or the last day on which the cytotoxic agent is present at therapeutic levels.
In a preferred embodiment, the time period over which the FGF antagonist is administered or over which the FGF antagonist is maintained at a therapeutic level, e.g., at a plasma concentration that is sufficient to enhance the cytotoxic effect of the cytotoxic agent begins less than 180, more preferably less than 90, more preferably less than 60, and most preferably less than 30 days before the first day on which the cytotoxic agent is administered or the first day on which the cytotoxic agent is present at therapeutic levels. In a preferred embodiment, the FGF antagonist: is a protein or a peptide; is an antibody, e.g., a monoclonal, a murine antibody or a human antibody, or an antigen- binding fragment thereof. Preferably, the monoclonal antibody is a human antibody. The antibodies can be of the various isotypes, including: IgG (e.g., IgGl, IgG2, IgG3, IgG4), IgM, IgAl, IgA2, IgD, of IgE. Preferably, the antibody is an IgG isotype. The antibodies can be full-length (e.g., an IgGl or IgG4 antibody) or can include only an antigen-binding fragment (e.g., a Fab, F(ab')2, Fv or a single chain Fv fragment).
In a preferred embodiment, the FGF antagonist: is a recombinant antibody, e.g., a chimeric or a humanized antibody, or an antigen binding fragment thereof, e.g., has a variable region, or at least a complementarity determining region (CDR), derived from a non-human antibody (e.g., murine), while the remaining portion(s) are human in origin.
In a preferred embodiment, the FGF antagonist is a fragment of the FGF molecule. Preferably, the FGF fragment competes with an FGF molecule for binding to the receptor. In a preferred embodiment, the FGF antagonist is a small molecule, (e.g., is selected from a combinatorial library).
In a preferred embodiment, the FGF antagonist is chosen from those disclosed herein, e.g., suramin, structural analogs of suramin, anti-FGF antibodies, anti-FGF receptor antibodies, pentosan polysulfate, scopolamine, angiostatin, sprouty, estradiol, carboxymethylbenzylamme dextran (CMDB7), suradista, insulin-like growth factor binding protein-3, ethanol, heparin (e.g., 6-O-desulfated heparin), low molecular weight heparin, heparan sulfate, protamine sulfate, transforming growth factor beta, cyclosporin A, or RNA ligands for bFGF. In a preferred embodiment, the FGF antagonist is heparin. In a preferred embodiment, the FGF antagonist is low molecular weight heparin.
In a preferred embodiment, the FGF antagonist is heparan sulfate. In a preferred embodiment, the FGF antagonist is an anti-FGF antibody.
In a preferred embodiment, the FGF antagonist is suramin. In a preferred embodiment, the FGF antagonist is suramin and the suramin is present in a concentration that is sufficient to block the resistance to anticancer agents induced by FGF (e.g., bFGF and/or aFGF), but is not sufficient to produce one or more of: (i) significant inhibition of cell proliferation; (ii) significant cell death in human and/or animal tumor cells, (iii) a measurable antitumor effect in a subject, e.g., a human subject, and/or (iv) cell cycle arrest. The determination of effect on cultured cells can be determined with the system described in Example XV.
In a preferred embodiment, the FGF antagonist is suramin and it is administered at levels such that the plasma concentration of suramin that is present when the cytotoxic agent is present in plasma at pharmacologically active concentration does not result in one or more of: (i) significant cell cycle arrest, (ii) significant cell death, or (iii) significant inhibition of cell growth, e.g., the concentration in plasma is of a level that, if the same concentration of suramin is provided in cultured cells, at least 10, more preferably at least 25, more preferably at least 50, more preferably at least 70, more preferably at least 80, more preferably at least 90, more preferably at least 99, and most preferably at least 99% of the treated cultured cells continue to be involved in one or more of: (i) cycling cells continue to progress through the cell cycle, (ii) cells remain viable, or (iii) cells remain capable of proliferating, following treatment with suramin. The determination of effect on cultured cells can be determined with the system described in Example XV.
Preferably, suramin is administered in an amount that results in a concentration ranging from about 0.1 to 100 μg/ml, preferably about 1 to 85 μg/ml, more preferably, about 5 to 60 μg/ml, even more preferably, about 10 to 50 μg/ml, and most preferably, 15 to 45 μg/ml. The pharmacokinetics of suramin is characterized by a triphasic concentration decline, with half-lives of 5.5 hours, 4.1 days and 78 days. The total body clearance is 0.0095 liter/hour/m2 (Jodrell et al (1994) J Clin Oncol 12:166-175). Based on pharmacokinetic principles, a person skilled in the art can calculate that an initial dose of approximately 240 mg/m2 should be administered to the average patient to achieve plasma concentrations declining from about 90 μg/ml (63 μM) to about 18 μg/ml (13 μM) over 96 hours. The 96 hour duration is chosen as an example, as the plasma concentrations of many commonly used cytotoxic agents to be administered with suramin will have declined below their therapeutic levels at 96 hours. Similar calculations can be performed to identify the initial suramin dose to deliver the preferred suramin plasma concentrations over other desired treatment durations. Maintenance doses to adjust the plasma concentrations for later treatment cycles can be similarly calculated. In a preferred embodiment, the total suramin exposure is preferably less than
800 μM-day over 96 hours, preferably less than 600 μM-day over 96 hours, preferably less than 500 μM-day over 96 hours, preferably less than 400 μM-day over 96 hours, preferably less than 300 μM-day over 96 hours, preferably less than 252 μM-day over 96 hours, preferably less than 200 μM-day over 96 hours, preferably less than 150 μM- day over 96 hours, preferably less than 100 μM-day over 96 hours, and most preferably less than 52 μM-day over 96 hours. The total suramin exposure, as expressed in μM- day, is a product of the drug plasma concentration in μM-day (e.g., the average micromolarity over 24 hours) and the treatment duration in days. For example, treatment of a subject with 13 μM of suramin for four days would result in a total drug exposure of 52 μM-day over 96 hours.
Preferably, suramin is administered in an amount that results in a plasma concentration of less than 100, preferably less than 80, preferably less than 50, preferably less than 25, more preferably less than 15, and most preferably less than 10 μM. Preferably, this plasma concentration is maintained for less than 20 days, preferably less than 15 days, preferably less than 12 days, preferably less than 10 days, more preferably less than 8 days, and most preferably less than 5 days beyond the time duration where therapeutic concentrations of a cytotoxic agent are maintained.
In a preferred embodiment the FGF antagonist is suramin and the time period over which the suramin is administered or over which the suramin is maintained at the plasma concentration sufficient to inhibit or reverse the FGF-mediated resistance or to enhance the efficacy of the cytotoxic agent is less than 180, more preferably less than 90, more preferably less than 60, and most preferably less than 30 days. In a preferred embodiment, suramin is given immediately or within 3, 2, or 1 day before the administration of the cytotoxic agent, ending immediately after plasma concentrations of the cytotoxic agent are below the therapeutic level.
In a preferred embodiment, the FGF antagonist is suramin and the time period over which the suramin is administered or over which the suramin is maintained at the plasma concentration sufficient to inhibit or reverse the FGF-mediated resistance or to enhance the efficacy of the cytotoxic agent does not begin substantially earlier or end substantial later than the period over which the cytotoxic agent is administered or over which the cytotoxic agent is maintained at a therapeutic level. In a preferred embodiment, the FGF antagonist is suramin and the time period over which the suramin is administered or over which the suramin is maintained at the plasma concentration sufficient to inhibit or reverse the FGF-mediated resistance or to enhance the efficacy of the cytotoxic agent ends less than 180, more preferably less than 90, more preferably less than 60, and most preferably less than 30 days after the last day on which the cytotoxic agent is administered or the last day on which the cytotoxic agent is present at therapeutic levels.
In a preferred embodiment, the FGF antagonist is suramin and the time period over which the suramin is administered or over which the suramin is maintained at the plasma concentration sufficient to inhibit or reverse the FGF-mediated resistance or to enhance the efficacy of the cytotoxic agent begins less than 180, more preferably less than 90, more preferably less than 60, and most preferably less than 30 days before the first day on which the cytotoxic agent is administered or the first day on which the cytotoxic agent is present at therapeutic levels.
Methods described herein use suramin to enhance the antitumor effect of a cytotoxic agent (e.g., agents described in Table 2), where the suramin dose is selected to deliver a plasma concentration of below 100 μg/ml, preferably below 75 μg/ml, most preferably below 50 μg/ml, in a mammal while the cytotoxic agent is present in plasma at a pharmacologically active concentration. The suramin dose is administered before, simultaneously with, or after the administration of the at least one anticancer agent. Animal trials presented herein show that treatment of mice with two weekly intravenous bolus suramin doses of 10 mg/kg for 3 weeks enhances the antitumor effect of anticancer drugs (e.g., paclitaxel, doxorubicin) but does not result in additional body weight loss. This dose is calculated to result in a plasma suramin concentration of about 10 μM (-14 μg/ml) at 72 hours after dose administration (see Example IX, Tables 4 and 5, Figure 7). The methods of the art use high dose suramin, either alone or in combination with a cytotoxic agent, where for a human subject, maintenance of plasma suramin concentrations of between 150 to 300 μg/ml is needed to produce a measurable antitumor effect (Eisenberger et al (1995) J Clin Oncol 13:2174-2186). A typical suramin dosing schedule aimed at maintaining suramin plasma concentrations between 150 and 300 μg/ml consists of an initial administration of 2100 mg/m2 over the first week with the subsequent doses repeated every 28 days for 6 months or longer; the subsequent doses are adjusted using the Bayesian pharmacokinetic method (Dawson et al (1998) Clin Cancer Res 4:37-44, Falcone et al (1999) Cancer 86:470- 476). Moreover, the methods of the art for using suramin in combination with other cytotoxic agents often administer suramin at a more frequent schedule or a longer duration than the frequency and the treatment duration for the other cytotoxic agents. For example, in the combination of suramin and doxorubicin for the treatment of androgen-independent prostate cancer, the duration of doxorubicin treatment was up to 20 weeks whereas the duration of the suramin treatment was up to 45 weeks (Tu et al (1998) Clin Cancer Res 4:1193-1201). For example, in the combination of suramin and mitomycin C for the treatment of hormone-resistant prostate cancer, suramin was given weekly whereas mitomycin C was given only every 5 weeks (Rapoport et al (1993) Ann Oncol 4:567-573). At these doses and chronic treatments, suramin causes the following toxicity in a human patient: adrenal insufficiency, coagulopathy, peripheral neuropathy, and proximal muscle weakness (Dorr and Von Hoff, Cancer Chemotherapy Handbook, 1994, pp 859-866). The incidence and severity of these toxicities are positively related to cumulated dose and are minimized in the methods described herein.
In a preferred embodiment, the FGF antagonist is an anti-FGF antibody. Preferably, the anti-FGF antibody is present in a concentration that is sufficient to block the resistance to anticancer agents induced by FGF (e.g., bFGF and/or aFGF), but is not sufficient to produce one or more of: (i) significant inhibition of cell proliferation; (ii) significant cell death in human and/or animal tumor cells, (iii) a measurable antitumor effect in a subject, e.g., a human subject, and/or (iv) cell cycle arrest.
In a preferred embodiment, the FGF antagonist is an anti-FGF antibody and it is administered at levels such that the plasma concentration of the anti-FGF antibody that is present when the cytotoxic agent is present in plasma at pharmacologically active concentration does not result in one or more of: (i) significant cell cycle arrest, (ii) significant cell death, or (iii) significant inhibition of cell growth, e.g., the concentration in plasma is of a level that if the same concentration of the anti-FGF antibody is provided in cultured cells at least 10, more preferably at least 25, more preferably at least 50, more preferably at least 70, more preferably at least 80, more preferably at least 90, more preferably at least 99, and most preferably at least 99% of the treated cultured cells, continue to be involved in one or more of: (i) cycling cells continue to progress through the cell cycle, (ii) cells remain viable, or (iii) cells remain capable of proliferating, following treatment with the FGF antibody. The determination of effect on cultured cells can be determined with the system described in Example XV.
In a preferred embodiment, the FGF antagonist is an anti-FGF antibody and the time period over which the anti-FGF antibody is administered or over which the anti- FGF antibody is maintained at the plasma concentration sufficient to inhibit or reverse the FGF-mediated resistance or to enhance the efficacy of the cytotoxic agent is less than 180, more preferably less than 90, more preferably less than 60, and most preferably less than 30 days.
In a preferred embodiment, the FGF antagonist is an anti-FGF antibody and the time period over which the anti-FGF antibody is administered or over which the anti- FGF antibody is maintained at the plasma concentration sufficient to inhibit or reverse the FGF-mediated resistance or to enhance the efficacy of the cytotoxic agent does not begin substantially earlier or end substantial later than the period over which the cytotoxic agent is administered or over which the cytotoxic agent is maintained at a therapeutic level.
In a preferred embodiment, the FGF antagonist is an anti-FGF antibody and the time period over which the anti-FGF antibody is administered or over which the anti- FGF antibody is maintained at the plasma concentration sufficient to inhibit or reverse the FGF-mediated resistance or to enhance the efficacy of the cytotoxic agent ends less than 180, more preferably less than 90, more preferably less than 60, and most preferably less than 30 days after the last day on which the cytotoxic agent is administered or the last day on which the cytotoxic agent is present at therapeutic levels. In a preferred embodiment, the FGF antagonist is an anti-FGF antibody and the time period over which the anti-FGF antibody is administered or over which the anti- FGF antibody is maintained at the plasma concentration sufficient to inhibit or reverse the FGF-mediated resistance or to enhance the efficacy of the cytotoxic agent begins less than 180, more preferably less than 90, more preferably less than 60, and most preferably less than 30 days before the first day on which the cytotoxic agent is administered or the first day on which the cytotoxic agent is present at therapeutic levels.
In a preferred embodiment, the method inhibits the proliferation of, or enhances the killing of, a hyperproliferative cell selected from the group consisting of a solid tumor cell, a soft-tissue tumor cell, a metastatic tumor cell, a leukemic tumor cell, and a lymphoid tumor cell.
In a preferred embodiment, the method inhibits the proliferation of, or enhances the killing of, a hyperproliferative cell in a fibrotic tumor. In a preferred embodiment, the disorder is a cancer, e.g., a sarcoma, a carcinoma, an adenocarcinoma, a lymphoma, or a leukemia.
In a preferred embodiment, the disorder is a cancer which includes an established tumor.
In a preferred embodiment, the disorder is a cancer which includes a solid tumor.
In a preferred embodiment, the disorder is a cancer which includes a metastatic lesion.
In a preferred embodiment, the disorder is a cancer which includes a leukemia. In a preferred embodiment, the disorder is a cancer which includes a lymphoma. In a preferred embodiment, the disorder is a cancer, e.g., a fibrosarcoma, myosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, gastric cancer, esophageal cancer, colon carcinoma, rectal cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, uterine cancer, cancer of the head and neck, skin cancer, brain cancer, squamous cell carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinoma, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, testicular cancer, lung carcinoma, small cell lung carcinoma, non-small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, meningioma, melanoma, neuroblastoma, retinoblastoma, leukemia, lymphoma, or Kaposi sarcoma.
In a preferred embodiment, the disorder includes a cancer which includes cells, e.g., tumor or metastatic cells, which form from a tissue where an FGF molecule is expressed, preferably at high levels, or cells that are in contact or exposed to aFGF, bFGF and/or FGF-producing cells or tissues.
In a preferred embodiment, the disorder includes a cancer which includes cells, e.g., metastatic cells, which form from a tissue of the breast, prostate, kidney, bladder, liver, lungs, lymph nodes, colon, rectum, skin, brain, pancreas, cervix, ovary, larynx, pharynx, oral mucosa, cancers of the head and neck, cancers of hematopoietic origin, or cancers of the lymphoid system.
In a preferred embodiment, the disorder is a cancer which includes an established tumor, e.g., a tumor that has been growing in a subject for at least one week, preferably two weeks, preferably one month, preferably two months, more preferably three months, more preferably six months, and most preferably longer than six months.
Preferably, an established tumor can be diagnosed clinically. Preferably, an established tumor can be visualized on diagnostic media, e.g., an X-ray, a CAT scan, or MRI. Preferably, an established tumor can be diagnosed by the detection of tumor markers, e.g., prostate specific antigen for prostate cancer, Her2/neu for certain breast cancers, CA125 for ovarian cancer, or genetic alterations.
Preferably, an established tumor can be diagnosed by the detection of pathological changes, e.g., blood in sputum for lung cancer, blood in feces in colon cancer, blood in urine for bladder cancer, a lump in breasts for breast cancer, pain, or headache.
Preferably, an established tumor can be diagnosed by biochemical analysis of a patient sample, e.g., a patient's sputum, feces, urine, etc. Preferably, an established tumor can be diagnosed by morphological analysis of cells derived from a subject, e.g., morphological abnormalities, abnormal nucleus-to- cytoplasmic ratios, abnormal tissue architecture, abnormal tissue organization, or abnormal tissue composition. Preferably, an established tumor is one that is visible, palpable, or found during necropsy or autopsy.
In a preferred embodiment, the disorder includes a cancer, e.g., lung cancer, renal cancer, glioma, melanoma, or chemotherapy-refractory cancer, a metastatic cancer, which is chemoresistant, e.g., shows little or no significant response to chemotherapy.
In a preferred embodiment, the hyperproliferative cell is found in a benign lesion.
In a preferred embodiment, the disorder is selected from the group consisting of psoriasis, cysts, benign prostatic hyperplasia, and endometriosis. In a preferred embodiment, the disorder is selected from the group consisting of benign hyperplastic diseases, e.g., oral papillomas, central giant cell granulomas of the mouth or pharynx, benign cementoblastomas of the oral cavity, oral plakia, gastric polyps, gastric adenomas, small intestinal adenomas, small intestinal granulomas, small intestinal papillomas, small intestinal oncocytomas, small intestinal Schwannomas, colonic polyps, colonic adenomas, Crohn's disease, hepatic adenoma, hepatic cirrhosis, biliary papillomatosis, pancreatic adenomas, pancreatic ductal hyperplasia, renal oncocytomas, renal papillomas, adenomas of the bladder, malakoplakia of the bladder, pseudosarcomas of the bladder, endometriosis, benign prostatic hyperplasia, erythroplasia of the penis, polyps and papillomas of the vulva, vagina, or cervix, endometrial polyps, adenomas, papillomas, or leimyomas, ovarian cysts, fibrocystic disease of the breast, lipoma of the breast, sclerosing adenosis, hemangioma, ductal hyperplasia of the breast, fibroadenomas, adenomyoepitheliomas, hamartoma, nevus of the skin, genodermatoses, fibrosis of the bone, fibrous dysplasia, chondrodysplasisa, sclerosing bone dysplasia, axial osteomalacia, fibrogenesis imperfecta, osteomas, osteoid osteomas, osteoblastomas, osteochondomas, enchondromas, chondromyxoid fibromas, chondroblastomas, synovial lipomas, adenomas of endocrine organs, goiter, Graves' disease, adrenal hyperplasia, adrenal adenomas, adrenal MEN I syndrome, adrenal myelolipomas. In a preferred embodiment, the subject is a mammal, e.g., a human. E.g., the subject is a patient, e.g., a cancer patient. The subject can be a patient with non-small cell lung cancer, is treated with a combination of paclitaxel, carboplatin and an FGF antagonist, e.g., suramin, or with a combination of gemcitabine, cisplatin, and an FGF antagonist, e.g., suramin. The subject can be a patient with hormone refractory prostate cancer, who is treated with a combination of estramustine phosphate, taxotere and an FGF antagonist, e.g., suramin, or with a combination of doxorubicin, ketoconazole and an FGF antagonist, e.g., suramin. The subject can be a patient with metastatic breast cancer, who is treated with a combination of cyclophosphamide, doxorubicin, 5- fluorouracil and an FGF antagonist, e.g., suramin, or a combination of doxorubicin, taxotere and an FGF antagonist, e.g., suramin. The subject can be a patient with advanced breast cancer that overexpresses the HER2/neu oncogene, who is treated with a HER2/neu inhibitor (e.g., a HER2/neu antibody) and an FGF antagonist, e.g., suramin, with or without paclitaxel or cisplatin. The subject can be a patient with advanced or metastatic colorectal cancer, who is treated with a combination of irinotecan and an FGF antagonist, e.g., suramin. The subject can be a patient with advanced colon cancer, who is treated with a combination of 5-fluorouracil, leucovorin and an FGF antagonist, e.g., suramin.
In a preferred embodiment, the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an agent that promotes apoptosis and/or necrosis, an interferon, an interleukin, a tumor necrosis factor, and radiation.
In a preferred embodiment, the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interferon, an interleukin, a tumor necrosis factor, and radiation.
In a preferred embodiment, the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interferon, an interleukin, a tumor necrosis factor, and radiation. In a preferred embodiment, the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interferon, an interleukin, a tumor necrosis factor, and radiation.
In a preferred embodiment, the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interferon, an interleukin, a tumor necrosis factor, and radiation.
In a preferred embodiment, the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interferon, an interleukin, and radiation.
In a preferred embodiment, the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interleukin, a tumor necrosis factor, and radiation.
In a preferred embodiment, the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a mitotic inhibitor, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interleukin, and radiation. In a preferred embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-Asparate=PALA,N-Phosphoracetyl-L-Asparate (PALA), pentostatin, N-Phosphoracetyl-L-Asparate=PALA, pentostatin,5-azacitidine, 5-Aza-C, BCNU=Carmustine,5-azacitidine,5-Aza- 5-Aza-2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5- fluoro-2'-deoxyuridine, 5-fluorouridine, 5'-deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU=Carmustine,BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, Lupron,leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (Zoledax), flutamide, 4'-cyano-3-(4-(e.g., Zoladex), flutamide, 4'-cyano-3-(4-fluorophenylsulphonyl)-2-hydroxy-2-methyl-3'- (trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, tumor necrosis factors, and radiation.
In a preferred embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (Docetaxel), topotecan, irinotecan hydrochloride=Camptosar, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, (e.g., Docetaxel), topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, epirubicin, merbarone, piroxantrone hydrochloride, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N-Phosphoracetyl-L-Asparate=PALA, pentostatin, 5-azacitidine, 5-Aza-2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'-deoxyuridine, 5- fluorouridine, 5'-deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, carboplatin, oxaliplatin, mitomycin C, BCNU=Carmustine, melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, Lupron, ketoconazole, tamoxifen, goserelin (Zoledax), flutamide, 4'-cyano-3-(4- fluorophenylsulphonyl)-2-hydroxy-2-methyl-3'-(trifluoromethyl) propionanilide, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza- 2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'-deoxyuridine, 5-fluorouridine, 5'- deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano- 3-(4-fluorophenylsulphonyl)-2-hydroxy-2-methyl-3'-(trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, tumor necrosis factors, and radiation.
In a preferred embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere amsacrine, (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), etoposide, mitoxantrone, daunorubicin, epirubicin, merbarone, piroxantrone hydrochloride, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N-Phosphoracetyl-L-Asparate=PALA, pentostatin, 5-azacitidine, 5-Aza-2'-deoxycytidine, adenosine arabinoside (Ara-A), idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza- 2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'-deoxyuridine, 5-fluorouridine, 5'- deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano- 3-(4-fluorophenylsulphonyl)-2-hydroxy-2-methyl-3'-(trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, tumor necrosis factors, and radiation. In a preferred embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine
(Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N-Phosphoracetyl- L-Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza-2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5- fluoro-2'-deoxyuridine, 5-fluorouridine, 5'-deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano-3-(4-fluorophenylsulphonyl)-2-hydroxy- 2-methyl-3 '-(trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, tumor necrosis factors, and radiation. In a preferred embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza- 2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'-deoxyuridine, 5-fluorouridine, 5'- deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano-3-(4- fluorophenylsulphonyl)-2-hydroxy-2-methyl-3'-(trifluoromethyl) propionanilide, anti- Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, tumor necrosis factors, and radiation.
In a preferred embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza- 2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'-deoxyuridine, 5-fluorouridine, 5'- deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano- 3 -(4-fluorophenylsulphonyl)-2-hydroxy-2-methyl-3 '-(trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, and radiation.
In a preferred embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza- 2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'-deoxyuridine, 5-fluorouridine, 5'- deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano- 3-(4-fluorophenylsulphonyl)-2-hydroxy-2-methyl-3'-(trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, tumor necrosis factors, and radiation.
In a preferred embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), topotecan, irinotecan hydrochloride (e.g., Camptosar), etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N-Phosphoracetyl-L- Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza-2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5- fluoro-2'-deoxyuridine, 5-fluorouridine, 5'-deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano-3-(4-fluorophenylsulphonyl)-2-hydroxy- 2-methyl-3'-(trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, and radiation.
In a preferred embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere(e.g., Docetaxel), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N-Phosphoracetyl-L-Asparate (PALA), pentostatin,
5-azacitidine, 5-Aza-2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'-deoxyuridine, 5- fluorouridine, 5'-deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano-3-(4-fluorophenylsulphonyl)-2-hydroxy-2-methyl-3'- (trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, tumor necrosis factors, and radiation. In a preferred embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N-Phosphoracetyl-L-Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza-2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'-deoxyuridine, 5- fluorouridine, 5'-deoxy-5- fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano-3-(4-fluorophenylsulphonyl)-2-hydroxy-2-methyl-3'- (trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, tumor necrosis factors, and radiation.
In a preferred embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano- 3-(4-fluorophenylsulphonyl)-2-hydroxy-2-methyl-3'-(trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin- 12, tumor necrosis factors, and radiation. In a preferred embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza- 2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'-deoxyuridine, 5-fluorouridine, 5'- deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano-3-(4-fluorophenylsulphonyl)- 2-hydroxy-2-methyl-3'-(trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin- 12, tumor necrosis factors, and radiation.
In a preferred embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza- 2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'-deoxyuridine, 5-fluorouridine, 5'- deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano- 3-(4-fiuorophenylsulphonyl)-2-hydroxy-2-methyl-3'-(trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interleukin 2, interleukin 4, interleukin 12, tumor necrosis factors, and radiation.
In a preferred embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano-3-(4-fluorophenylsulphonyl)-2-hydroxy- 2-methyl-3 '-(trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interleukin 2, interleukin 4, interleukin 12, and radiation. In a preferred embodiment, the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, 5-fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide. In a preferred embodiment, the cytotoxic agent is: paclitaxel, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, 5- fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
In a preferred embodiment, the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, taxotere, doxorubicin, epirubicin, 5- fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
In a preferred embodiment, the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, epirubicin, 5- fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
In a preferred embodiment, the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
In a preferred embodiment, the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, 5-fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
In a preferred embodiment, the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, taxotere, doxorubicin, epirubicin, 5-fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
In a preferred embodiment, the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
In a preferred embodiment, the cytotoxic agent is: paclitaxel, gemcitabine, fludarabine, irinotecan, taxotere, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
In a preferred embodiment, the cytotoxic agent is present in an amount equal to or lower than the one used in conventional chemotherapy. For example, for the combination of paclitaxel and carboplatin with an FGF antagonist, the dose of paclitaxel is equal to or below 225 mg/m2, and the dose of carboplatin is chosen to achieve a total concentration-time product of equal to or below 6-7 mg/ml.min in previously untreated patients, or equal to or below 4-5 mg/ml.min in patients that have received chemotherapy previously; the treatment is repeated every 3 weeks. For example, for the combination of estramustine phosphate and taxotere with an FGF antagonist, the daily oral dose of estramustine is equal to or below 1400 mg, and the dose of taxotere is equal to or below 70 mg/m over 1 hour; the treatment is repeated every 3 weeks. For example, for the combination of doxorubicin and ketoconazole with an FGF antagonist, the weekly dose of doxorubicin is equal to or below 20 mg/m2 by 24 hr infusion, and the total daily oral dose of ketoconazole is equal to or below 1200 mg. For example, for the combination of cyclophosphamide, doxorubicin and 5- fluorouracil with an FGF antagonist, the dose of intravenous cyclophosphamide is equal to or below 500 mg/m2, the dose of doxorubicin is equal to or below 50 mg/m2, and the dose of 5-fluorouracil is equal to or below 500 mg/m2; the treatment is repeated every 4 weeks (the 5-fluorouracil dose is given once per week for two weeks whereas the doses of doxorubicin and cyclophosphamide are given once every 4 weeks). For example, for the combination of Herceptin and cisplatin with an FGF antagonist, the Herceptin dose is equal to or below 250 mg on day 0, followed by 9 weekly doses of equal to or below 100 mg, and the cisplatin dose is equal to or below 75 mg/m2 on days 1, 29, and 57. For example, for the combination of irinotecan with an FGF antagonist, the four weekly doses of irinotecan are equal to or below 125 mg/m2; the treatment cycle is 4 weeks on and 2 weeks off. For example, for the combination of irinotecan with an FGF antagonist, the dose of irinotecan is 350 mg/m2 every 3 weeks. For example, for the combination of 5-fluorouracil and leucovorin with an FGF antagonist, the five daily intravenous bolus doses of 5-fluorouracil are equal to or below 425 mg/m2, and the five daily intravenous bolus doses of leucovorin are equal to or below 20 mg/m2; the treatment cycle is 1 week on and 4 weeks off. For example, for the combination of gemcitabine and cisplatin with an FGF antagonist, the three weekly doses of gemcitabine are equal to or below 1000 mg/m2, and the single cisplatin dose given on day 2 is equal to or below 100 mg/m ; the treatment is repeated every 4 weeks. In a preferred embodiment, the method further includes repeated dosages of the same, or a different cytotoxic agent.
In a preferred embodiment, the method further includes repeated dosages of the same, or a different FGF antagonist.
In a preferred embodiment, the cytotoxic agent and the FGF antagonist are administered at the same time or in overlapping time periods; the agent and the FGF antagonist are administered at different times; the agent is administered first and the FGF antagonist is administered subsequently; the FGF antagonist is administered first and the agent is administered subsequently.
In a preferred embodiment, the cytotoxic agent is administered systemically or locally. For example, the agent can be administered parenterally (e.g., subcutaneously, intravenously, intramuscularly, intraperitoneally, intradermally, intrathecally, etc.), intravesically (i.e., urinary bladder), intraprostatically, orally, nasally, rectally, topically, and/or transdermally.
In a preferred embodiment, the FGF antagonist is administered systemically or locally. For example, the FGF antagonist can be administered parenterally (e.g., subcutaneously, intravenously, intramuscularly, intraperitoneally, intradermally, intrathecally, etc.), intravesically (i.e., urinary bladder), intraprostatically, orally, nasally, rectally, topically, and/or transdermally. In a prefeπed embodiment, the methods described herein further comprise monitoring the FGF levels (e.g., a-FGF and/or b-FGF) of a subject prior to or during treatment. Preferably, the amount of an FGF antagonist administered to the subject is determined based upon the levels of FGF present in the subject, e.g., low concentrations of FGF in the subject require decreased doses of the FGF antagonist to overcome the chemoresistance mediated by FGF.
In another aspect, the invention features, a method of treating a cell, e.g., inhibiting cell killing, or protecting the ability of a cell (e.g., a dividing cells, preferably, a rapidly dividing cell) to proliferate, e.g., a method of protecting a rapidly dividing cell, in a subject, from one or more of killing, inhibition of growth or division or other damage caused, e.g., by a cytotoxic agent (e.g., a cytostatic agent, e.g., an agent that causes cell death). The method includes: administering, to the subject, an effective amount of at least one FGF agonist, thereby treating the cell, e.g., protecting or reducing the damage to the dividing cell from said cytotoxic agent.
In a preferred embodiment, the cell is: a cell from a body surface or cavity, e.g., a cell of the gastrointestinal or esophageal tract; a hair follicle cell; a hematopoietic cell, e.g., a hematopoietic stem cell.
In a preferred embodiment, the cell is part of the lining of the gastrointestinal tract or the esophageal tract.
In a preferred embodiment, the method inhibits hair loss; inhibits weight loss; inhibits the loss of gastrointestinal function; inhibits the loss of hematopoiesis.
In a preferred embodiment, the method further includes administering at least one cytotoxic agent (e.g., a cytostatic agent, e.g., an agent that causes cell death), e.g., an antiproliferative agent, e.g., an anticancer drug, e.g. radiation, e.g., an interferon, e.g., an interleukin, e.g., a tumor necrosis factor, to said subject.
In a preferred embodiment the cytotoxic agent is other than radiation, e.g., is an administered compound.
In a preferred embodiment, the cytotoxic agent is a compound other than an antimetabolite.
In a preferred embodiment, the FGF agonist is administered orally, locally, e.g., topically, or ex vivo. For example, the FGF agonist can be applied topically to treat hair follicles. In other embodiments, the cell, e.g., a bone marrow cell, can be treated ex vivo.
In a preferred embodiment, the FGF agonist is administered locally to a site, e.g., gastrointestinal tract, bone marrow, or the skin. In a prefeπed embodiment, the FGF agonist is directly injected into the bone marrow.
In a prefeπed embodiment, the FGF agonist is applied topically, e.g. externally to the scalp.
In a prefeπed embodiment, the application of the FGF agonist does not significantly result in absorption into systemic circulation, and/or does not significantly result in a blood or plasma concentration that is sufficient to increase the proliferation of a tumor cell in the subject.
In a prefeπed embodiment, the FGF agonist is administered to a subject by a method that does not result in significant systemic administration or significant systemic levels of the FGF agonist, e.g. does not result in an increase in FGF agonist blood levels that promotes the chemoresistance of the tumor. Systemic administration or levels of the FGF agonist can be evaluated by determining the level of the FGF agonist in the blood before and after its administration to a subject. These two levels can then be tested in vitro and compared for their effect on the antitumor effect mediated by a cytotoxic agent against tumor cells. Preferably the FGF agonist blood level after administration should increase the IC o of the cytotoxic agent by less than 30, preferably less than 20, preferably less than 10, preferably less than 5, and most preferably less than 1%, as compared to the IC5o of the cytotoxic agent in the presence of the blood levels of the FGF agonist prior to administration of the FGF agonist to the subject. The determination of effect on cultured cells can be determined with the system described in Example XV.
In a prefeπed embodiment, the subject is a mammal, e.g., a human. E.g., the subject is a patient, e.g., a cancer patient. For example, the subject is a patient with non-small cell lung cancer, who is treated with a combination of two or more of: paclitaxel, carboplatin, or an FGF antagonist, e.g., suramin, or with a combination of two or more of: gemcitabine, cisplatin, or an FGF antagonist, e.g., suramin. For example, the patient is a patient with hormone refractory prostate cancer, who is treated with a combination of two or more of: estramustine phosphate, taxotere, or an FGF antagonist, e.g., suramin, or with a combination of two or more of: doxorubicin, ketoconazole, an FGF antagonist, e.g., suramin. For example, the patient is a patient with metastatic breast cancer, who is treated with a combination of two or more of: cyclophosphamide, doxorubicin, 5-fluorouracil, or an FGF antagonist, e.g., suramin. For example, the patient is a patient with advanced breast cancer that overexpresses the HER2/neu oncogene, who is treated with Herceptin and suramin, with or without paclitaxel or cisplatin. For example, the patient is a patient with advanced or metastatic colorectal cancer, who is treated with one or more of: irinotecan or an FGF antagonist, e.g., suramin. For example, the patient is a patient with advanced colon cancer, who is treated with a combination of two or more of: 5-fluorouracil, leucovorin, or an FGF antagonist, e.g., suramin.
In a prefeπed embodiment, the treatment protects said cell from an effect, e.g., a cytotoxic effect (e.g., cytostasis, e.g., cell kill, e.g., hair loss), of said cytotoxic treatment. In a prefeπed embodiment, the FGF agonist comprises bFGF, aFGF, or bFGF and aFGF, or a fragment or an analog thereof. When bFGF and aFGF are given in combination, they may have an additive effect, preferably, a synergistic effect. The FGF agonist is administered in an amount that results in concentrations sufficient to protect said cell from an effect, e.g., a cytotoxic effect (e.g., cytostasis, e.g., cell kill, e.g., hair loss), of said cytotoxic treatment. Administration of the FGF agonist may be repeated to provide protection throughout the duration of said cytotoxic treatment and/or throughout the duration when the plasma concentrations of the cytotoxic agent remain sufficient to produce a cytotoxic effect.
In a prefeπed embodiment, the time period over which the FGF agonist is administered or over which the FGF agonist is maintained at a therapeutic level, e.g., a plasma concentration that is sufficient to protect cells from the cytotoxic effects of the cytotoxic agent, is less than 180, more preferably less than 90, more preferably less than 60, and most preferably less than 30 days.
In a prefeπed embodiment, the time period over which the FGF agonist is administered or over which the FGF agonist is maintained at a therapeutic level, e.g., a plasma concentration that is sufficient to protect cells from the cytotoxic effects of the cytotoxic agent, does not begin substantially earlier or end substantial later than the period over which the cytotoxic agent is administered or over which the cytotoxic agent is maintained at a therapeutic level.
In a prefeπed embodiment, the time period over which the FGF agonist is administered or over which the FGF agonist is maintained at a therapeutic level, e.g., plasma concentration that is sufficient to protect cells from the cytotoxic effects of the cytotoxic agent, ends less than 180, more preferably less than 90, more preferably less than 60, and most preferably less than 30 days after the last day on which the cytotoxic agent is administered or the last day on which the cytotoxic agent is present at therapeutic levels. In a prefeπed embodiment, the time period over which the FGF agonist is administered or over which the FGF agonist is maintained at a therapeutic level, e.g., a plasma concentration that is sufficient to protect cells from the cytotoxic effects of the cytotoxic agent, begins less than 180, more preferably less than 90, more preferably less than 60, and most preferably less than 30 days before the first day on which the cytotoxic agent is administered or the first day on which the cytotoxic agent is present at therapeutic levels.
In a prefeπed embodiment, the FGF agonist: is a peptide, or a small molecule.
In a prefeπed embodiment, the FGF agonist is nonproteinaceous.
In a prefeπed embodiment, the cytotoxic agent used in the methods described herein is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti- hormone, e.g., an antibody against growth factor receptors), an agent that promotes apoptosis and/or necrosis, an interferon, an interleukin, a tumor necrosis factor, and radiation.
In a prefeπed embodiment, the cytotoxic agent used in the methods described herein is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interferon, an interleukin, a tumor necrosis factor, and radiation. In a prefeπed embodiment, the cytotoxic agent used in the methods described herein is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti- hormone, e.g., an antibody against growth factor receptors, an interferon, an interleukin, and a tumor necrosis factor.
In a prefeπed embodiment, the cytotoxic agent used in the methods described herein is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interferon, an interleukin, a tumor necrosis factor. In a prefeπed embodiment, the cytotoxic agent used in the methods described herein, is chosen from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), topotecan, camptothecin, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N-Phosphoracetyl-L-Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza-2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'-deoxyuridine, 5- fluorouridine, 5'-deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (Zoladex), flutamide, 4'-cyano-3-(4-fluorophenylsulphonyl)-2-hydroxy-2-methyl-3'-
(trifluoromethyl) propionanilide, anti-Her 2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, tumor necrosis factors, and radiation. In a prefeπed embodiment, the cytotoxic agent used in the methods described herein, is chosen from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), topotecan, camptothecin, irinotecan hydrochloride (e.g., Camptosar), pyrazofurin, doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N-Phosphoracetyl-L- Asparate=PALA, Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza-2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'-deoxyuridine, 5-fluorouridine, 5'-deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (Zoladex), flutamide, 4'-cyano-3-(4-fluorophenylsulphonyl)-2- hydroxy-2-methyl-3 '-(trifluoromethyl) propionanilide, anti -Her 2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, and tumor necrosis factors.
In a prefeπed embodiment, the cytotoxic agent used in the methods described herein, is chosen from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), topotecan, camptothecin, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N-Phosphoracetyl-L- Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza-2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'-deoxyuridine, 5- fluorouridine, 5'-deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (Zoladex), flutamide, 4'-cyano-3-(4-fluorophenylsulphonyl)-2-hydroxy-2-methyl-3'- (trifluoromethyl) propionanilide, anti-Her 2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, and tumor necrosis factors, and radiation.
In a prefeπed embodiment, the cytotoxic agent used in the methods described herein, is chosen from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), topotecan, camptothecin, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano-3-(4-fluorophenylsulphonyl)-2-hydroxy- 2-methyl-3'-(trifluoromethyl) propionanilide, anti-Her 2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, tumor necrosis factors, and radiation.
In a prefeπed embodiment, the cytotoxic agent used in the methods described herein, is chosen from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), topotecan, camptothecin, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (Zoladex), flutamide, 4'-cyano-3-(4-fluorophenylsulphonyl)-2-hydroxy-2- methyl-3 '-(trifluoromethyl) propionanilide, anti-Her 2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, and tumor necrosis factors.
In a prefeπed embodiment, the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, 5-fluororuacil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (Lupron) and flutamide.
In another aspect, the invention features, a pharmaceutical composition which includes at least one FGF antagonist, at least one cytotoxic agent (e.g., a cytostatic agent, e.g., an agent that causes cell death), and a pharmaceutically acceptable carrier, wherein said FGF antagonist is present in an amount effective to enhance the efficacy of the cytotoxic agent, in reducing or inhibiting the proliferation, or in enhancing the killing, of a hyperproliferative cell.
In a prefeπed embodiment, the pharmaceutical composition is packaged with directions to practice the methods described herein.
In a prefeπed embodiment, the pharmaceutical composition includes: an inhibitor of bFGF; an inhibitor of aFGF; or a bFGF inhibitor and an aFGF inhibitor.
In a prefeπed embodiment, the FGF antagonist inhibits or reverses the resistance to anticancer drugs induced by FGF (e.g. aFGF and/or bFGF) in cultured tumor cells under in vitro conditions. The determination of effect on cultured cells can be determined with the system described in Example XV.
In a prefeπed embodiment, the FGF antagonist inhibits or reduces the resistance of tumor cells to a broad spectrum of cytotoxic agents.
In a prefeπed embodiment, the FGF antagonist: is capable of binding to an FGF molecule or an FGF receptor; blocks the binding of FGF to a receptor; blocks the interaction of FGF to molecules that facilitate the binding of FGF to a receptor; down regulates FGF receptor action; is a protein or a peptide; is an antibody, e.g., a monoclonal, diabody, a murine antibody, a human antibody, a humanized or a chimeric antibody, or an antigen-binding fragment thereof, e.g., an Fab, F(ab')2, Fv or a single chain Fv fragment; is a truncated FGF molecule, or a fragment thereof.
In a prefeπed embodiment, the FGF antagonist acts extracellularly, e.g., inhibits the binding of an FGF molecule to the extracellular domain of the FGF receptors. In a prefeπed embodiment, the FGF antagonist acts intracellularly, e.g., inhibits the binding of an FGF molecule to the intracellular domain of the FGF receptors.
In a prefeπed embodiment, the FGF antagonist acts intracellularly, e.g., inhibits the intracellular effects of FGF. In a prefeπed embodiment, the FGF antagonist acts extracellularly, e.g., inhibits the binding of an FGF molecule to its receptor.
In a prefeπed embodiment, the FGF antagonist is chosen from those disclosed herein, e.g., suramin, structural analogs of suramin, anti-FGF antibodies, anti-FGF receptor antibodies, pentosan polysulfate, scopolamine, angiostatin, sprouty, estradiol, carboxymethylbenzylamine dextran (CMDB7), suradista, insulin-like growth factor binding protein-3, ethanol, heparin (e.g., 6-O-desulfated heparin), low molecular weight heparin, heparan sulfate, protamine sulfate, transforming growth factor beta, cyclosporin A, or RNA ligands for bFGF.
In a prefeπed embodiment, the FGF antagonist is suramin. In a prefeπed embodiment, the FGF antagonist is a fragment of the FGF molecule which competes with an FGF molecule for binding to the receptor; the FGF antagonist is a small molecule.
In a prefeπed embodiment, the FGF antagonist is a small molecule, is chosen from a combinatorial library. In a prefeπed embodiment, the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an agent that promotes apoptosis and/or necrosis, an interferon, an interleukin, a tumor necrosis factor, and radiation.
In a prefeπed embodiment, the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interferon, an interleukin, a tumor necrosis factor, and radiation. In a prefeπed embodiment, the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interferon, an interleukin, a tumor necrosis factor, and radiation.
In a prefeπed embodiment, the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interferon, an interleukin, a tumor necrosis factor, and radiation.
In a prefeπed embodiment, the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interferon, an interleukin, a tumor necrosis factor, and radiation.
In a prefeπed embodiment, the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interferon, an interleukin, and radiation. In a prefeπed embodiment, the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interleukin, a tumor necrosis factor, and radiation.
In a prefeπed embodiment, the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a mitotic inhibitor, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an interleukin, and radiation.
In a prefeπed embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L- Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza- 2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'-deoxyuridine, 5-fluorouridine, 5'- deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano- 3-(4-fluorophenylsulphonyl)-2-hydroxy-2-methyl-3'-(trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, tumor necrosis factors, and radiation.
In a prefeπed embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza- 2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'-deoxyuridine, 5-fluorouridine, 5'- deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano- 3-(4-fluorophenylsulphonyl)-2-hydroxy-2-methyl-3'-(trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, tumor necrosis factors, and radiation. In a prefeπed embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza- 2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'-deoxyuridine, 5-fluorouridine, 5'- deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano- 3-(4-fluorophenylsulphonyl)-2-hydroxy-2-methyl-3'-(trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, tumor necrosis factors, and radiation. In a prefeπed embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N-Phosphoracetyl- L- Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza-2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5- fluoro-2'-deoxyuridine, 5-fluorouridine, 5'-deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano-3-(4-fluorophenylsulphonyl)-2-hydroxy- 2-methyl-3'-(trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, tumor necrosis factors, and radiation.
In a prefeπed embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza- 2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'-deoxyuridine, 5-fluorouridine, 5'- deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano-3-(4- fluorophenylsulphonyl)-2-hydroxy-2-methyl-3'-(trifluoromethyl) propionanilide, anti- Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, tumor necrosis factors, and radiation.
In a prefeπed embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L- Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza- 2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'- deoxyuridine, 5-fluorouridine, 5'- deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano- 3-(4-fluorophenylsulphonyl)-2-hydroxy-2-methyl-3'-(trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, and radiation.
In a prefeπed embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza-
2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'-deoxyuridine, 5-fluorouridine, 5'- deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano- 3-(4-fluorophenylsulphonyl)-2-hydroxy-2-methyl-3'-(trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, tumor necrosis factors, and radiation. In a prefeπed embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), topotecan, irinotecan hydrochloride (e.g., Camptosar), etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N-Phosphoracetyl-L- Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza-2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5- fluoro-2'-deoxyuridine, 5-fluorouridine, 5'-deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano-3-(4-fluorophenylsulphonyl)-2-hydroxy- 2-methy 1-3 '-(trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, and radiation.
In a prefeπed embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N-Phosphoracetyl-L- Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza-2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'-deoxyuridine, 5- fluorouridine, 5'-deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano-3-(4-fluorophenylsulphonyl)-2-hydroxy-2-methyl-3'- (trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, tumor necrosis factors, and radiation. In a prefeπed embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N-Phosphoracetyl-L- Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza-2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'-deoxyuridine, 5- fluorouridine, 5 '-deoxy-5 -fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano-3-(4-fluorophenylsulphonyl)-2-hydroxy-2-methyl-3'- (trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, tumor necrosis factors, and radiation. In a prefeπed embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza- 2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'-deoxyuridine, 5-fluorouridine, 5'- deoxy-5 -fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano-3-(4-fluorophenylsulphonyl)- 2-hydroxy-2-methyl-3'-(trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, tumor necrosis factors, and radiation.
In a prefeπed embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza- 2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'-deoxyuridine, 5-fluorouridine, 5'- deoxy-5-fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano- 3-(4-fluorophenylsulphonyl)-2-hydroxy-2-methyl-3'-(trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, and radiation. In a prefeπed embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), camptothecin, topotecan, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza- 2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'- deoxyuridine, 5-fluorouridine, 5'- deoxy-5 -fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano- 3-(4-fluorophenylsulphonyl)-2-hydroxy-2-methyl-3'-(trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20 (e.g., Rituxan), interleukin 2, interleukin 4, interleukin 12, tumor necrosis factors, and radiation.
In a prefeπed embodiment, the cytotoxic agent is selected from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano-3-(4-fluorophenylsulphonyl)-2-hydroxy- 2-methyl-3'-(trifluoromethyl) propionanilide, anti-Her2/neu antibody (e.g., Herceptin), anti- CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, and radiation.
In a prefeπed embodiment, the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, 5-fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
In a prefeπed embodiment, the cytotoxic agent is: paclitaxel, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, 5- fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
In a prefeπed embodiment, the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, taxotere, doxorubicin, epirubicin, 5- fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
In a prefeπed embodiment, the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, epirubicin, 5- fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide. In a prefeπed embodiment, the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
In a prefeπed embodiment, the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, 5-fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
In a prefeπed embodiment, the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, taxotere, doxorubicin, epirubicin, 5-fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
In a prefeπed embodiment, the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide.
In a prefeπed embodiment, the cytotoxic agent is: paclitaxel, gemcitabine, fludarabine, irinotecan, taxotere, tamoxifen, goserelin, ketoconazole, anti-Her2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (e.g., Lupron) and flutamide. In a prefeπed embodiment, the cytotoxic agent is present at an amount equal to or lower than the one used in conventional chemotherapy.
In a prefeπed embodiment, the hyperproliferative cell is a cancer cell.
In a prefeπed embodiment, the hyperproliferative cell is found in a benign lesion.
In a prefeπed embodiment, the disorder is selected from the group consisting of psoriasis, cysts, benign prostatic hyperplasia, and endometriosis.
In a prefeπed embodiment, the disorder is selected from the group consisting of benign hyperplastic diseases, e.g., oral papillomas, central giant cell granulomas of the mouth or pharynx, benign cementoblastomas of the oral cavity, oral plakia, gastric polyps, gastric adenomas, small intestinal adenomas, small intestinal granulomas, small intestinal papillomas, small intestinal oncocytomas, small intestinal Schwannomas, colonic polyps, colonic adenomas, Crohn's disease, hepatic adenoma, hepatic ciπhosis, biliary papillomatosis, pancreatic adenomas, pancreatic ductal hyperplasia, renal oncocytomas, renal papillomas, adenomas of the bladder, malakoplakia of the bladder, pseudosarcomas of the bladder, endometriosis, benign prostatic hyperplasia, erythroplasia of the penis, polyps and papillomas of the vulva, vagina, or cervix, endometrial polyps, adenomas, papillomas, or leimyomas, ovarian cysts, fibrocystic disease of the breast, lipoma of the breast, sclerosing adenosis, hemangioma, ductal hyperplasia of the breast, fibroadenomas, adenomyoepitheliomas, hamartoma, nevus of the skin, genodermatoses, fibrosis of the bone, fibrous dysplasia, chondrodysplasisa, sclerosing bone dysplasia, axial osteomalacia, fibrogenesis imperfecta, osteomas, osteoid osteomas, osteoblastomas, osteochondomas, enchondromas, chondromyxoid fibromas, chondroblastomas, synovial lipomas, adenomas of endocrine organs, goiter, Graves' disease, adrenal hyperplasia, adrenal adenomas, adrenal MEN I syndrome, adrenal myelolipomas.
In another aspect, the invention features, a pharmaceutical composition which includes at least one FGF agonist, and a pharmaceutically acceptable carrier, wherein said FGF agonist is present in an amount effective to protect, or reduce the damage, a cell, e.g., a rapidly dividing cell, from said cytotoxic agent.
In a prefeπed embodiment, the pharmaceutical composition is packaged with directions to practice the methods described herein. In a prefeπed embodiment, the FGF agonist comprises bFGF; aFGF; or bFGF and aFGF, or a fragment or an analog thereof.
In a prefeπed embodiment, the FGF agonist comprises bFGF, or a fragment or an analog thereof. In a prefeπed embodiment, the FGF agonist: is a peptide, or a small molecule.
In a prefeπed embodiment the FGF agonist is nonproteinaceous.
In a prefeπed embodiment, the cell is: a cell from a body surface or cavity, e.g., a cell of the gastrointestinal or esophageal tract; a hair follicle cell; a hematopoietic cell, e.g., a hematopoietic stem cell. In a prefeπed embodiment, the cell is a part of the lining of the gastrointestinal tract or the esophageal tract.
In another aspect, the invention relates to kits for carrying out the combined administration of the FGF antagonist/agonist with other cytotoxic agents. In one embodiment, the kit comprises an FGF antagonist/agonist formulated in a pharmaceutical carrier, and at least one cytotoxic agent, formulated, as appropriate, in one or more separate pharmaceutical preparations.
In a prefeπed embodiment, the kit includes directions to practice the methods described herein.
In another aspect, the invention features a vaccine comprising an FGF polypeptide, or a FGF receptor-derived polypeptide, or a fragment of a proteoglycan that facilitates the binding of FGF to its receptors, and a pharmaceutically acceptable carrier, in an amount effective to immunize a subject against a neoplastic disease. The immunization against the neoplastic disease can be partial or complete. Said vaccine can be used to treat a cancer patient, or to prevent one or more of: the occuπence of, recidivism of, and/or metastasis of, the neoplastic disease. For example, it could be administered to a patient who has had a tumor surgically removed, to prevent recurrence of the tumor. In a prefeπed embodiment, the subject is a mammal, e.g., a human.
In a prefeπed embodiment, the subject is not a cancer patient. In a prefeπed embodiment, the subject is a pateint that is afflicted with an ailment caused by a benign hyperplasia of a normal tissue. In a prefeπed embodiment, the subject is a patient, e.g., a cancer patient. For example, the subject can be a patient in remission, or a cancer patient undergoing treatment (e.g., conventional chemotherapy, alone or in combination, with the methods described herein). For example, the subject can be a patient with non-small cell lung cancer, treated with a combination of two or more of: paclitaxel, carboplatin or an FGF antagonist, e.g., suramin, or with a combination of two or more of: gemcitabine, cisplatin, or an FGF antagonist, e.g., suramin. The subject can be a patient with hormone refractory prostate cancer, who is treated with a combination of two or more of: estramustine phosphate, taxotere, or an FGF antagonist, e.g., suramin, or with a combination of two or more of: doxorubicin, ketoconazole, or an FGF antagonist, e.g., suramin. The subject can be a patient with metastatic breast cancer, who is treated with a combination of two or more of: cyclophosphamide, doxorubicin, 5-fluorouracil, or an FGF antagonist, e.g., suramin, or a combination of two or more of: doxorubicin, taxotere, or an FGF antagonist, e.g., suramin. The subject can be a patient with advanced breast cancer that overexpresses the HER2/neu oncogene, who is treated with a HER2/neu inhibitor (e.g., a HER2/neu antibody) and/or an FGF antagonist, e.g., suramin, with or without paclitaxel or cisplatin. The subject can be a patient with advanced or metastatic colorectal cancer, who is treated with one or more of: irinotecan or an FGF antagonist, e.g., suramin. The subject can be a patient with advanced colon cancer, who is treated with a combination of two or more of: 5- fluorouracil, leucovorin, or an FGF antagonist, e.g., suramin. The subject can be a patient that is in remission but is with a cancer that has a good likelihood to recur, e.g., adult leukemia, e.g., early disseminated prostate cancer.
In another aspect, the invention features, a method for evaluating the effectiveness of a compound, e.g., for treating a disorder, e.g., a proliferative disorder, e.g., a malignant disorder, or for protecting cells, e.g., from a cytotoxic agent. The method includes: contacting the compound with an FGF, e.g., aFGF or bFGF; and evaluating the ability of the compound to inhibit or promote FGF activity, inhibition being coπelated with effectiveness of treating a disorder, and promotion being coπelated with cell protection. In a prefeπed embodiment, the method further comprises testing the compound to determine if it can modulate, e.g., increase, the efficacy of an agent which kills cells or inhibits cell growth, e.g., an anticancer agent. This can be done by administering the compound and the agent, together or separately, to a test cell or organism. In a prefeπed embodiment, the method further comprises testing the compound to determine if it can modulate, e.g., increase, the efficacy of an agent which kills cells or inhibits cell growth, e.g., an anticancer agent, in the presence of FGF (e.g., bFGF, and or aFGF). This can be done by administering the compound, the agent and the FGF, together or separately, to a test cell or organism. In a prefeπed embodiment, the method further comprises testing the compound to determine if it can protect cells from an agent that kills cells or inhibits cell growth, e.g., a cytotoxic or an anticancer agent. This can be done by administering both the compound and the agent, together or separately, to a test cell or organism. In a prefeπed embodiment the method includes: providing a cell, e.g., a cultured cell, a transformed cell, a cell from a cancer, or a test organism; administering said compound to said cell (or test organism) and evaluating FGF activity, cell proliferation, cell death, or tumor growth, e.g., metastatic tumor growth. In a prefeπed embodiment, the disorder is a cancer which includes a sarcoma, a carcinoma, an adenocarcinoma, a lymphoma, or a leukemia.
In a prefeπed embodiment, the disorder is a cancer which includes a solid tumor.
In a prefeπed embodiment, the disorder is a cancer which includes a leukemia. In a prefeπed embodiment, the disorder is a cancer which includes a lymphoma. In a prefeπed embodiment, the disorder is a cancer which includes a metastatic lesion.
In a prefeπed embodiment, the disorder includes a cancer which includes cells, e.g., metastatic cells, which form from a tissue where an FGF molecule is expressed, or cells that are in contact or exposed to aFGF, bFGF and/or FGF-producing cells or tissues.
In a prefeπed embodiment, the disorder includes a cancer which includes cells, e.g., metastatic cells, which form from a tissue of the breast, prostate, kidney, bladder, liver, lungs, lymph nodes, colon, rectum, skin, brain, pancreas, cervix, ovary, larynx, pharynx, oral mucosa, cancers of the head and neck, cancers of hematopoietic origin, or cancers of the lymphoid system.
In a prefeπed embodiment, the disorder is a cancer, e.g., a fibrosarcoma, myosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, gastric cancer, esophageal cancer, colon carcinoma, rectal cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, uterine cancer, cancer of the head and neck, skin cancer, brain cancer, squamous cell carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinoma, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, testicular cancer, lung carcinoma, small cell lung carcinoma, non-small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, meningioma, melanoma, neuroblastoma, retinoblastoma, leukemia, lymphoma, or Kaposi sarcoma.
In a prefeπed embodiment, the disorder is selected from the group consisting of benign hyperplastic diseases, e.g., oral papillomas, central giant cell granulomas of the mouth or pharynx, benign cementoblastomas of the oral cavity, oral plakia, gastric polyps, gastric adenomas, small intestinal adenomas, small intestinal granulomas, small intestinal papillomas, small intestinal oncocytomas, small intestinal Schwannomas, colonic polyps, colonic adenomas, Crohn's disease, hepatic adenoma, hepatic ciπhosis, biliary papillomatosis, pancreatic adenomas, pancreatic ductal hyperplasia, renal oncocytomas, renal papillomas, adenomas of the bladder, malakoplakia of the bladder, pseudosarcomas of the bladder, endometriosis, benign prostatic hyperplasia, erythroplasia of the penis, polyps and papillomas of the vulva, vagina, or cervix, endometrial polyps, adenomas, papillomas, or leimyomas, ovarian cysts, fibrocystic disease of the breast, lipoma of the breast, sclerosing adenosis, hemangioma, ductal hyperplasia of the breast, fibroadenomas, adenomyoepitheliomas, hamartoma, nevus of the skin, genodermatoses, fibrosis of the bone, fibrous dysplasia, chondrodysplasisa, sclerosing bone dysplasia, axial osteomalacia, fibrogenesis imperfecta, osteomas, osteoid osteomas, osteoblastomas, osteochondomas, enchondromas, chondromyxoid fibromas, chondroblastomas, synovial lipomas, adenomas of endocrine organs, goiter, Graves= disease, adrenal hyperplasia, adrenal adenomas, adrenal MEN I syndrome, adrenal myelolipomas. In a prefeπed embodiment, the compound is a protein or a peptide.
In a prefeπed embodiment, the compound is a chemical, e.g., a small molecule (e.g., a member of a combinatorial library).
In a prefeπed embodiment, the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), an agent that promotes apoptosis and/or necrosis, an interferon, an interleukin, a tumor necrosis factor, or radiation. In a prefeπed embodiment, the cytotoxic agent is chosen from those disclosed below. Exemplary cytotoxic agents include: paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel), topotecan, camptothecin, irinotecan hydrochloride (e.g., Camptosar), doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride, 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N- Phosphoracetyl-L-Asparate=PALA, N-Phosphoracetyl-L- Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza- 5-Aza-2'-deoxycytidine, adenosine arabinoside (Ara- A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'- deoxyuridine, 5-fluorouridine, 5 '-deoxy-5 -fluorouridine, hydroxyurea, dihydrolenchlorambucil, tiazofurin, cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol, dihydrolenperone, spiromustine, geldenamycin, cytochalasins, depsipeptide, leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (Zoladex), flutamide, 4'-cyano-3-(4-fluorophenylsulphonyl)-2-hydroxy-2- methyl-3'-(trifluoromethyl) propionanilide, Herceptin, anti-CD20 (e.g., Rituxan), interferon alpha, interferon beta, interferon gamma, interleukin 2, interleukin 4, interleukin 12, tumor necrosis factors, and radiation.
In a prefeπed embodiment, the cytotoxic agent is: paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, 5-fluorouracil, UFT, tamoxifen, goserelin, a HER2/neu antibody (e.g., Herceptin), anti-CD20, leuprolide (Lupron) and flutamide.
In a prefeπed embodiment the cell is a rapidly dividing cell, a gastrointestinal cell, or a hair follicle cell, or a hematopoietic cell, e.g., a hematopoietic stem cell.
In another aspect, the invention features, a method of analyzing a sample, e.g., for the level of tumor resistance to anticancer agents. The method includes: evaluating the level of one or more FGF genes or gene products, wherein an increase or decrease in the level of one or more FGF genes or gene products, relative to a control, indicates the presence of tumor resistance to anticancer agents. In a prefeπed embodiment, the sample, e.g. a sample form a subject, is a sample of tissue having unwanted proliferation, e.g., a sample of a benign hypeφlasic tissue, a sample from a primary tumor, a metastatic tumor, or a leukemia.
In a prefeπed embodiment, the expression of FGF genes or gene products is selected from the group consisting of bFGF, aFGF, TSC22, VEGF, GAFA1, GAFA2, GAFA3 (originally designated as FSC1, FSC2, FSC3 in the U.S. Provisional
Application No. 60/137,345), and TFII-I.
In a prefeπed embodiment, the method is performed: on a sample from a mammal, a sample from a human subject; and a sample from a cancer patient; as part of therapeutic decision making; to determine if the individual from which the target FGF is taken should receive a drug or other treatment; to diagnose an individual for a disorder or for predisposition to resistance to treatment, to stage a disease or disorder. In a prefeπed embodiment, the method further includes choosing a therapeutic modality, e.g., a particular anticancer treatment, e.g. a particular FGF antagonist, or a dosage thereof, based on the level of FGF expression. In some embodiments, nucleic acid (or protein) from the cell or sample is analyzed on positional aπays, e.g., DNA-chip aπays. Accordingly, in prefeπed embodiments, the method further includes: analyzing the sample by providing an aπay of a plurality of capture probes, wherein each of the capture probes is positionally distinguishable from other capture probes of the plurality on the aπay, and wherein each positional distinguishable capture probe includes a unique reagent e.g., an antibody or a nucleic acid probe which can identify an FGF gene or gene product; hybridizing the sample with the aπay of capture probes, thereby analyzing the sample sequence.
In another aspect, the method includes a method of staging a disorder, e.g., a proliferative disorder, e.g., a benign hypeφlastic disorder, e.g., a malignant disorder, in a subject. The method includes: providing a sample, e.g., cancerous sample, e.g. a tissue, a bodily fluid, e.g., urine, blood, or CSF, a biopsy, from said subject; evaluating the expression of one or more FGF genes, e.g., by contacting said cancerous sample with, a nucleic acid probe that selectively hybridizes to one or more FGF gene products; wherein an increase in the level of said one or more FGF genes or gene products, relative to a control, indicates a stage in the disorder, e.g., the malignant disorder. In a prefeπed embodiment the FGF genes or gene products are selected from the group consisting of bFGF, aFGF, TSC22, VEGF, GAFA1, GAFA2, GAFA3 (originally designated as FSC1, FSC2, FSC3 in the U.S. Provisional Application No. 60/137,345), and TFII-I.
In prefeπed embodiments the method is performed: on a sample from a mammal, a sample from a human subject; e.g., a sample of a patient suffering from a benign hypeφlastic disorder, e.g., a sample from a cancer patient; to determine if the individual from which the target nucleic acid or protein is taken should receive a drug or other treatment; to diagnose an individual for a disorder or for predisposition to resistance to treatment, to stage a disease or disorder. In a prefeπed embodiment, the method further includes choosing a therapeutic modality, e.g., a particular anticancer treatment, or a dosage thereof, based on the level of FGF expression.
In a prefeπed embodiment, the expression of an FGF gene is evaluated by evaluating the expression of a signal entity, e.g., a green fluorescent protein or other marker protein, which is under the control or an FGF gene control element e.g., promoter.
In some embodiments, nucleic acid (or protein) from the cell or sample is analyzed on positional aπays, e.g., DNA-chip arrays. Accordingly, in prefeπed embodiments the method further includes: analyzing the sample by providing an aπay of a plurality of capture probes, wherein each of the capture probes is positionally distinguishable from other capture probes of the plurality on the aπay, and wherein each positional distinguishable capture probe includes a unique reagent, e.g., an antibody or a nucleic acid probe which can identify an FGF gene or gene product; hybridizing the sample with the aπay of capture probes, thereby analyzing the sample sequence.
In another aspect, the invention features, a method of diagnosing a disorder, e.g., a proliferative disorder, e.g., a malignant disorder, e.g., a benign hypeφroliferative disorder, in a subject. The method includes: providing a sample e.g., a cancerous sample, e.g., a tissue, a bodily fluid (e.g., blood, urine, sputum, CSF), a biopsy, from said patient; evaluating the expression of one or more FGF genes, e.g., by contacting said sample with, a nucleic acid probe that selectively hybridizes to one or more FGF genes, or an antibody that specifically binds to one or more FGF gene products; wherein an increase or decrease in the level of said one or more FGF genes or gene products, relative to a control, indicates the presence or absence of the disorder, e.g., the malignant disorder. In a prefeπed embodiment, the FGF related genes or gene product is selected from the group consisting of bFGF, aFGF, TSC22, VEGF, GAFA1, GAFA2, GAFA3 (originally designated as FSC1, FSC2, FSC3 in the U.S. Provisional Application No. 60/137,345), and TFII-I.
In a prefeπed embodiment, the expression of an FGF gene is evaluated by evaluating the expression of a signal entity, e.g., a green fluorescent protein or other marker protein, which is under the control of or an FGF gene control element, e.g., promoter. In some embodiments, nucleic acid (or protein) from the cell or sample is analyzed on positional aπays, e.g., DNA-chip aπays. Accordingly, in prefeπed embodiments the method further includes: analyzing the sample by providing an aπay of a plurality of capture probes, wherein each of the capture probes is positionally distinguishable from other capture probes of the plurality on the aπay, and wherein each positional distinguishable capture probe includes a unique reagent, e.g., an antibody or a nucleic acid probe which can identify an FGF gene or gene product; hybridizing the sample with the aπay of capture probes, thereby analyzing the sample sequence.
In another aspect, the invention features, a method for evaluating the efficacy of a treatment of a disorder, e.g., a proliferative disorder, e.g., a malignant disorder, in a patient, comprising: providing a sample, e.g., a cancerous sample, e.g., a tissue, a bodily fluid (e.g., blood, urine, sputum, CSF), a biopsy, from said patient; evaluating the expression of one or more FGF genes, e.g., by contacting said cancerous sample with, a nucleic acid probe that selectively hybridizes to one or more FGF genes, or an antibody that specifically binds to one or more FGF gene products; wherein a change, e.g., decrease, e.g., increase, in the level of said one or more FGF genes or gene products in a sample obtained after treatment, relative to the level of expression before treatment, is indicative of the efficacy of the treatment of said disorder.
In a prefeπed embodiment, the method further includes choosing a therapeutic modality, e.g., a particular anticancer treatment, or a dosage thereof, based on the level of FGF expression.
In a prefeπed embodiment the FGF genes or gene product is selected from the group consisting of bFGF, aFGF, TSC22, VEGF, GAFA1, GAFA2, GAFA3 (originally designated as FSC1, FSC2, FSC3 in the U.S. Provisional Application No. 60/137,345), and TFII-I.
In a prefeπed embodiment, the expression of an FGF gene is evaluated by evaluating the expression of a signal entity, e.g., a green fluorescent protein or other marker protein, which is under the control or an FGF gene control element, e.g., promoter. In some embodiments, the nucleic acid (or protein) from the cell or sample is analyzed on positional aπays, e.g., DNA-chip aπays. Accordingly, in prefeπed embodiments the method further includes: analyzing the sample by providing an aπay of a plurality of capture probes, wherein each of the capture probes is positionally distinguishable from other capture probes of the plurality on the aπay, and wherein each positional distinguishable capture probe includes a unique reagent, e.g., an antibody or a nucleic acid probe which can identify an FGF gene or gene product; hybridizing the sample with the aπay of capture probes, thereby analyzing the sample sequence.
In another aspect, the invention features, a method for evaluating the effectiveness of a treatment, e.g., the administration of a compound, for treating a disorder, e.g., a proliferative disorder, e.g., a malignant disorder. The method includes: providing a cell, e.g., a cultured cell, a transformed cell, a cell from a cancer, or a test organism; administering said treatment to said cell (or test organism) and evaluating the expression of one or more FGF genes, e.g., by contacting a sample from said cell (or test organism) with, a nucleic acid probe that selectively hybridizes to one or more FGF genes, or an antibody that specifically binds to one or more FGF gene products; wherein a change, e.g., decrease, in the level of said one or more FGF genes or gene products in a sample given said treatment, e.g., relative to the level of expression without the treatment, indicative of the effectiveness of the compound for treating said disorder. In a prefeπed embodiment the FGF genes or gene product is selected from the group consisting of bFGF, aFGF, TSC22, VEGF, GAFA1, GAFA2, GAFA3 (originally designated as FSC1, FSC2, FSC3 in the U.S. Provisional Application No. 60/137,345), and TFII-I.
In a prefeπed embodiment the expression of an FGF gene is evaluated by evaluating the expression of a signal entity, e.g., a green fluorescent protein or other marker protein, which is under the control or an FGF gene control element, e.g., promoter. In some embodiments, nucleic acid (or protein) from the cell or sample is analyzed on positional aπays, e.g., DNA-chip aπays. Accordingly, in prefeπed embodiments the method further includes: analyzing the sample by providing an aπay of a plurality of capture probes, wherein each of the capture probes is positionally distinguishable from other capture probes of the plurality on the array, and wherein each positional distinguishable capture probe includes a unique reagent, e.g., an antibody or a nucleic acid probe which can identify an FGF gene or gene product; hybridizing the sample with the aπay of capture probes, thereby analyzing the sample sequence.
Other features and advantages of the invention will be apparent from the following detailed description, and from the claims.
Brief Description of the Drawings
Figure 1 depicts a panel of graphs showing the induction of resistance to paclitaxel, doxorubicin and 5-fluorouracil in monolayer cultures of rat MAT-LyLu tumor cells using conditioned medium (CM) collected from tumor cultures. The induction of drug resistance was detected as changes in proliferative activity by measuring the bromodeoxyuridine (BrdU) incoφoration in treated samples relative to controls. Top panels: CM of primary tumors. Middle panels: CM of lymph node metastases. Bottom panels: CM of lung metastases. For each panel: control with no CM, (open circles); addition of histoculture CM (filled circle); addition of CM of early monolayer culture (passage 0, filled square); addition of CM of late monolayer culture (passage 3, filled inverted triangle). The curves for CM of primary tumor cultures and late monolayer metastatic tumor cultures overlap with the control curves. Similar results were obtained for human PC3 tumor cells.
Figures 2A-2B depict a panel of graphs showing the reversal of CM-induced resistance by inhibiting or removing aFGF/bFGF.
Figure 2 A shows the reversal of CM-induced resistance (detected as changes in proliferative activity of rat MAT-LyLu cells by measuring the changes in BrdU incoφoration in treated samples relative to control samples) using a monoclonal antibody against aFGF or bFGF as indicated. The source of CM was lung histocultures for the aFGF experiment, and early monolayer lung cultures for the bFGF experiment. The study used four controls: no CM (left solid curves, open circles); addition of 5 μg/ml aFGF or bFGF antibody but no CM (inverted filled triangle, overlaps with the left control curve); addition of CM but no antibody (right solid curves, open square); addition of CM plus a nonspecific antibody, IgG (5 μg/ml, filled diamond, overlaps with the right CM-control curve). Dotted curves, from right to left, addition of aFGF or bFGF antibody concentrations of 0.05 μg/ml (filled circle); 0.1 μg/ml (filled square); 1 or 0.5 μg/ml (filled triangle); and 5 μg/ml (open inverted triangle). Similar results were found for doxorubicin (shown here), paclitaxel and 5-fluorouracil. Similar results were also found for human PC3 prostate tumor cells.
Figure 2B shows (a) the reversal of the CM-induced resistance to doxorubicin by removing aFGF and/or bFGF via immunoprecipitation, and (b) the restoration of the resistance by reconstituting the FGF-immunoprecipitated CM using human recombinant proteins (0.16 ng/ml r-aFGF and/or 0.9 ng/ml r-bFGF). The study was performed using rat MAT-LyLu cells. The reversal in CM-induced resistance was detected by measuring the changes in BrdU incoφoration in treated samples relative to control samples. The source of CM was lung histocultures for both aFGF and bFGF experiments. The study used two controls: no CM (left solid curves, empty circles); with CM (right solid curves, empty squares). Upper left: CM minus bFGF (filled square); CM minus aFGF plus r-aFGF (filled circles, overlaps with the right CM- control curve). Upper right: CM minus bFGF (filled square, overlaps with the left control curve); CM minus bFGF plus r-bFGF (filled circle, overlaps with the right CM control curve). Lower left: Both the CM minus aFGF and bFGF (filled square) and the CM minus aFGF and bFGF plus aFGF (filled circle) curves overlap with the left control curve. Lower center: CM minus aFGF and bFGF (filled square, overlaps with left control curve); CM minus aFGF and bFGF plus r-bFGF (filled circle). Lower right: CM minus aFGF and bFGF (filled square, overlaps with left control curve); CM minus aFGF and bFGF plus r-aFGF and r-bFGF (filled circle, overlaps with the right CM-control curve).
Figure 3 is a panel of graphs showing induction of resistance to paclitaxel, doxorubicin and 5-fluorouracil in human prostate PC3 tumor cells using r-aFGF alone, r-bFGF alone or a combination of r-aFGF and r-bFGF, in the absence of CM. The study was performed using human prostate PC3 tumor cells (shown here) and rat MAT- LyLu tumor cells. The induction of resistance by FGF was detected by measuring the changes in BrdU incoφoration in treated samples relative to control samples. The study used two controls: no CM (left solid curves, empty circles); with CM of lung histocultures (right solid curves, open squares). Top panels: addition of r-aFGF: 1 (filled circle), 10 (filled square) and 50 (filled inverted triangle) ng/ml. All three dotted curves overlap with the left control curve. Middle panels: addition of r-bFGF. Dotted curves from left to right: 1 (filled circle, overlaps with the left control curve), 10 (filled square) and 50 (filled inverted triangle, overlaps with the right CM-control curve) ng/ml. Bottom panels: addition of r-aFGF/r-bFGF combinations. Dotted curves from left to right: 0.04 and 1 ng/ml (filled square, overlaps with the left control curve); 0.08 and 1 ng/ml (open diamond); 0.16 and 1 ng/ml (filled inverted triangle); 0.32 and 1 ng/ml (filled diamond); 0.64 and 1 ng/ml (filled circle); and 1 ng/ml each (open inverted triangle). Similar results were obtained for rat MAT-LyLu tumor cells.
Figure is a graph depicting the FGF-induced resistance to cell kill caused by doxorubicin treatment. Drug-induced cell death in PC3 cells was monitored by the release of lactate dehydrogenase (LDH) to the culture medium. Results are expressed as a ratio of treated samples to untreated controls. Values for control samples varied by less than 10%. Curves from top to bottom: Without CM or FGF (open circle); addition of 0.9 ng/ml aFGF (filled inverted triangle, overlaps with top control curve); addition of 0.9 ng/ml bFGF (filled circle); addition of 0.16 ng/ml aFGF plus 0.9 ng/ml bFGF (filled diamond); CM of lung tumor histocultures (open square); 0.9 ng/ml aFGF plus 0.9 ng/ml bFGF (filled square). Similar results were found for paclitaxel, and 5- fluorouracil.
Figure 5 shows three graphs depicting the cytotoxicity of increasing concentrations of suramin in rat MAT-LyLu tumor cells, human prostate PC3 and PC3- LN tumor cells. These results show that suramin did not produce measurable cytotoxicity (i.e., <10% reduction in BrdU incoφoration) at the 1-15 micromolar concentration. The coπesponding IC5o values (i.e., suramin concentration required to result in 50% reduction in BrdU incoφoration) are 235 micromolar in rat tumor cells, 93 micromolar in PC3 cells and 98 micromolar in PC3-LN cells.
Figure 6 shows three graphs depicting the enhancement of the cytotoxicity of doxorubicin, paclitaxel and 5-fluorouracil using suramin, under in vitro conditions. The study was performed using rat MAT-LyLu tumor cells (shown here) and human prostate PC3 and PC3-LN tumor cells. The cytotoxic effect was detected by measuring the changes in BrdU incoφoration in treated samples relative to control samples. The study used two controls: no CM and no suramin (left solid curves, open circle); with CM of lung histocultures but no suramin (right solid curves, open square). Dotted curves from right to left, addition of suramin 5 μM (filled circle); 10 μM (filled square); and 15 μM (filled inverted triangle). Similar results were obtained for PC3 and PC3-LN cells.
Figures 7A-7B are two graphs depicting the suramin-mediated enhancement of doxorubicin activity in immunodeficient mice bearing subcutaneous bulky human prostate PC3 xenograft tumors.
Figure 7 A shows the changes in tumor weight (depicted as experimental weight relative to initial weight) with respect to duration of treatment. Figure 7B shows the changes in the animal body weight with respect to duration of treatment.
Detailed Description of the Invention The present invention is based, at least in part, on the discovery that bFGF induces broad spectrum resistance to anticancer agents, in a number of solid and soft- tissue tumors, metastatic lesions, as well as normal noncancerous intestinal epithelium. The bFGF-induced resistance was amplified by aFGF. In one embodiment, the invention shows that inhibitors of aFGF/bFGF enhanced the in vitro and in vivo activity of chemotherapy, resulting in shrinkage and eradication of well-established human lung metastasis and human subcutaneous xenograft tumors in mice. Accordingly, the present invention provides, in part, methods and compositions for inhibiting FGF- induced resistance to anticancer agents. In one embodiment, the FGF inhibitor is administered with other cytotoxic agents, e.g., an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti- hormone, e.g., an antibody against growth factor receptors), an agent that promotes apoptosis and/or necrosis, an interferon, an interleukin, a tumor necrosis factor, and radiation. The enhanced, and sometimes synergistic, effect of the FGF inhibitors) (e.g., suramin, e.g., FGF antibody) with anticancer agents (e.g., paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, 5-fluorouracil, UFT, tamoxifen, goserelin, Herceptin, anti- CD20, leuprolide (Lupron) and flutamide), in addition to improving the efficacy of these anticancer agents, may allow for the administration of lower doses of these anticancer agents, thus reducing the induction of side effects in a subject, (e.g., a patient). For example, the subject is a patient with non-small cell lung cancer, who is treated with a combination of paclitaxel, carboplatin and suramin, or with a combination of gemcitabine, cisplatin, and suramin. For example, the patient is a patient with hormone refractory prostate cancer, who is treated with a combination of estramustine phosphate, taxotere and suramin, or with a combination of doxorubicin, ketoconazole and suramin. For example, the patient is a patient with metastatic breast cancer, who is treated with a combination of cyclophosphamide, doxorubicin, 5- fluorouracil and suramin. For example, the patient is a patient with advanced breast cancer that overexpresses the HER2/neu oncogene, who is treated with a Her2/neu antibody (e.g., Herceptin) and suramin, with or without paclitaxel or cisplatin. For example, the patient is a patient with advanced or metastatic colorectal cancer, who is treated with a combination of irinotecan and suramin. For example, the patient is a patient with advanced colon cancer, who is treated with a combination of 5- fluorouracil, leucovorin and suramin.
It is shown that the conditioned medium (CM) of solid, soft-tissue tumors, and metastatic tumors induced resistance to multiple anticancer agents, in human and rodent tumor cells. These CM contained elevated levels of aFGF and bFGF (see Examples II- III). Inhibition of bFGF using a bFGF-specific monoclonal antibody and removal of bFGF by immunoprecipitation resulted in complete reversal of the CM-induced resistance to anticancer agents, whereas inhibition/removal of aFGF resulted in partial reversal (see Example IV). Using CM that had been depleted of aFGF and/or bFGF and subsequently reconstituted with respective human recombinant proteins, bFGF but not aFGF was found to induce resistance. aFGF was found to amplify the bFGF effect. aFGF and bFGF fully accounted for the CM effect, indicating these proteins are involved in the induction and maintenance of resistance to anticancer agents (see Example IV). The FGF-induced resistance was not due to reduced intracellular drug accumulation, nor altered cell proliferation (see Example XII), thus representing a novel epigenetic resistance mechanism.
It is shown that the elevated levels of aFGF and bFGF, as found in the active CM, induced broad spectrum resistance to at least 58 anticancer agents having different mechanisms of actions, including antitubulin/antimicrotubule agents, topoisomerase I inhibitors, topoisomerase II inhibitors, antimetabohtes, alkylating agents, intercalating agents, agents that interfere with signal transduction pathways (e.g., protein kinase C inhibitors), and agents that promote apoptosis and/or necrosis (see Examples II and VII). aFGF/bFGF induction of resistance to paclitaxel (an antimicrotubule agent), doxorubicin (a topoisomerase I inhibitor) and 5-fluorouracil (an antimetabolite) was detected in multiple cancer cell lines derived from different human solid tumors, including prostate, lung, pharynx, colon and breast, as well as a human breast cancer cell line derived from stable transfection of the mdrl gene encoding for the drug efflux protein, i.e., p-glycoprotein (see Example VIII). aFGF/bFGF also induced resistance of human leukemic cells to fludarabine (an antimetabolite) (see Example VIII).
It is further shown that inhibitors of FGF, including FGF antibodies and suramin, an inhibitor of multiple growth factors, including aFGF/bFGF, reverse the FGF-induced resistance and enhance the in vitro activity of anticancer agents, including at least 58 anticancer agents having different mechanisms of actions, including antitubulin antimicrotubule agents, topoisomerase I inhibitors, topoisomerase II inhibitors, antimetabohtes, alkylating agents, intercalating agents, agents that interfere with signal transduction pathways (e.g., protein kinase C inhibitors), and agents that promote apoptosis and/or necrosis (see Examples V and VII). The reversal of FGF- induced resistance was attained at suramin concentrations that do not produce measurable cytotoxicity under in vitro conditions (see Example VI). Under in vivo conditions, addition of suramin, at doses that do not produce measurable antitumor activity nor host toxicity, enhances the treatment efficacy of chemotherapy, resulting in shrinkage and eradication of well-established human lung metastases in mice and shrinkage of subcutaneous bulky human prostate xenograft in mice, without enhancing the host toxicity of chemotherapy (see Examples IX).
The appended examples further demonstrate that (a) presence of tumor cells enhanced the level of extracellular bFGF in host tissues, (b) bFGF level in tumors is determined by the location and the size of the tumor, and (c) bFGF level determines the sensitivity of human patient tumors to anticancer agents (see Example X). In addition, the appended examples show that treatment of human prostate cells with several anticancer agents with diverse structures and action mechanisms (e.g., antitubulin/antimicrotubule agents, topoisomerase I inhibitors, topoisomerase II inhibitors, alkylating agents, and antimetabohtes) significantly enhanced the level of extracellular bFGF (see Example XI). Collectively, these examples indicate (a) the use of FGF gene expression and level to detect the presence of tumor cells, (b) the use of FGF expression and level in tumors to determine the extent of resistance, (c) the use of FGF gene expression and level in tumors to determine the dosage of the FGF inhibitors, and (d) that patients that are previously treated with chemotherapy are likely to exhibit chemoresistance. The latter indicates the need of using FGF inhibitors to treat patients who have failed cancer therapy and have recurrent tumors.
An appended example demonstrates that FGF antagonists, e.g., suramin, e.g., heparin, e.g., low molecular weight heparin, e.g., heparan sulfate, inhibit the binding of bFGF to its binding sites (Example XVI).
An additional appended example demonstrates that FGF agonists (e.g., aFGF, bFGF) reduced the toxicity of anticancer agents, e.g., paclitaxel and doxorubicin, to non-cancerous intestinal epithelium (Example XIV). Hence, the invention also provides methods and compositions for treating or preventing the undesired host toxicity of anticancer agents using aFGF and bFGF, alone or in combination.
In sum, the findings described herein demonstrate, inter alia, (a) a novel epigenetic mechanism by which cancer cells utilize the unique microenvironment of solid tumors and metastases to elude cytotoxic insult, (b) an important role of extracellular growth factors in tumor sensitivity to anticancer agents, (c) a new cancer treatment paradigm using combinations of anticancer agents with aFGF/bFGF inhibitors, (d) a novel mechanism and methods by which normal noncancerous host cells can be protected from the undesired toxicity of anticancer agents, and (e) methods to use the bFGF level for making therapeutic decision for individual patients.
Before further description of the invention, certain terms employed in the specification, examples and appended claims are, for convenience, collected here.
The terms peptides, proteins, and polypeptides are used interchangeably herein. As used herein, the term "fibroblast growth factor" or "FGF" refers to a member of a family of polypeptides that are potent regulators of a variety of cellular processes including proliferation, differentiation, migration, moφhogenesis, tissue maintenance and in wound healing and repair (Clarke et al. (1993) J. Cell Sci.106: 121-133; Cuevas et al. (1988) Biochem. Biophys. Res. Commun. 156: 611-618; Burgess, W.H. and Maciag, T (1989) Ann. Rev. Biochem. 58: 575-606; Rifkin, D.B. and Moscatelli, D. (1989) J. Cell Biol. 109:1-6). The FGF family cuπently includes at least 19 structurally and functionally related proteins, including acidic and basic FGF, FGF-1 and FGF-2 respectively; int2 (FGF-3); hst (FGF-4); FGF-5; hst2 (FGF-6); keratinocyte growth factor (FGF-7); androgen-induced growth factor (FGF-8); glia-activating factor (FGF- 9); FGF-10-19 (Galzie, Z. et al. (1997) Biochem. Cell. Biol., 75:669-685; Yamasaki, M. et al. (1996) J. Biol. Chem, 271:15918-15921; Smallwood, P.M. et al. (1996) Proc. Natl. Acad. Sci USA, 93:9850-9857; McWhirter, J.R. et al. (1997) Development, 124:3221-3232; Hoshikawa, M. et al. (1998) Biochem. Biophys. Res. Comm., 244:187- 191; Hu, M.C.T. et al. (1998) Mol. Cell. Biol., 18:6063-6074; and Nishimura, T. et al. (1999) Biochim. Biophy. Ada, 1444:148-151). Preferably, the term FGF refers to acidic and basic FGF, FGF-1 and FGF-2, respectively (reviewed in (Galzie, Z. et al. (1997) Biochem. Cell. Biol., 75:669-685 and Burgess, W.H. and Maciag, T (1989) Ann. Rev. Biochem. 58: 575-606).
As used herein, the term "FGF receptor" or "FGFR" refers to FGF -binding polypeptides that possess intrinsic tyrosine kinase activity or heparan sulfate proteoglycans in the extracellular matrix. With respect to the tyrosine kinase receptors, there are cuπently four known genes encoding FGF receptors (FGFR-1, FGFR-2, FGFR-3, and FGFR-4), which can give rise to a variety of protein isoforms via alternative RNA splicing (Galzie, Z. et al. (1997) Biochem. Cell. Biol., 75:669-685). The structure of the FGFR consists of an extracellular region with three immunoglobulin-like domains, a transmembrane region, and a cytosolic tyrosine kinase domain that is activated upon ligand binding. FGF binding causes dimerization of the receptors, resulting in receptor autophosphorylation on tyrosine residues and the activation of intracellular signal transduction cascades. The term "FGFR" also includes heparan sulfate proteoglycans in the extracellular matrix which mediate the actions of FGF, e.g., protection from proteolysis, localization, storage, and internalization of growth factors (Faham, S. et al. (1998) Curr. Opin. Struct. Biol, 8:578-586). Heparan sulfate proteoglycans may serve as low affinity FGF receptors that act to present FGF to its cognate FGFR, and/or to facilitate receptor oligomerization (Galzie, Z. et al. (1997) Biochem. Cell. Biol, 75:669-685). As used herein, an "FGF antagonist" refers to an agent that inhibits (completely or partially) the activity, production, stability, of an FGF molecule. Preferably, the FGF antagonist acts extracellularly or intracellularly, e.g., inhibits the binding of an FGF molecule to an extracellular or intracellular domain of an FGF receptor, e.g., blocks the intracellular signalling initiated by activation of the FGF receptor. E.g., the inhibitor binds to the intracellular domain of the FGF receptor, or blocks a downstream action that is consequence of activation of the FGF receptor. In contrast, an "FGF agonist" refers to an agent that potentiates the activity, production, stability, of an FGF molecule, or activates FGF receptors.
As used herein, the term "specific inhibitor of FGF" refers to an FGF antagonist other than suramin.
As used herein, the terms "cytotoxic agent", "anticancer agent" and "antitumor agent" are used interchangeably herein and refer to agents that have the property of inhibiting the growth or proliferation (e.g., a cytostatic agent), or inducing the killing, of hypeφroliferative cells. Preferably, the cytotoxic agent inhibits or reverses the development or progression of a neoplasm, such as a solid tumor, a soft tissue tumor, a metastatic lesion, a lymphoma, or a leukemia, or the cytotoxic agent inhibits or reverses the development or progression of a benign hypeφlastic growth.
As used herein, a "therapeutically effective amount" of an FGF antagonist and a cytotoxic agent refers to an amount of such agents which in combination is effective, upon single- or multiple-dose administration to the subject, e.g., a patient, at inhibiting the growth or proliferation, or inducing the killing, of hypeφroliferative cells. Such growth inhibition or killing can be reflected as a prolongation of the survival of the subject, e.g., a patient beyond that expected in the absence of such treatment, or any improvement in the prognosis of the subject relative to the absence of such treatment. As applied to FGF agonists, the phrase refers to an amount of such agent which is effective, upon single- or multiple-dose administration to protect a cell in a subject, e.g., a patient, from damage by an agent, e.g., a cytotoxic agent.
As used herein, "a prophylactically effective amount" of an FGF antagonist and a cytotoxic agent refers to an amount of such agents which in combination is effective, upon single or multiple dose administration to the subject, e.g., a patient, in preventing or delaying the occuπence of the onset or recuπence of a neoplastic disease state. As applied to FGF agonists, the phrase refers to an amount of such agent which is effective, upon single- or multiple-dose administration, in preventing or delaying damage to a cell in a subject, e.g., a patient, by an agent, e.g., a cytotoxic agent.
As used herein, the language "subject" is intended to include human and non- human animals. Prefeπed human animals include a human patient having a disorder characterized by the abeπant activity of a hypeφroliferative cell. The term "non- human animals" of the invention includes all vertebrates, e.g., mammals and non- mammals, such as non-human primates, sheep, dog, cat, horse, cow, chickens, amphibians, reptiles, etc. Preferably, the subject is a human patient, e.g., a cancer patient, e.g., a patient with a benign hypeφlastic disorder. As used herein, "inhibiting the growth or proliferation" of the hypeφroliferative cell, e.g., neoplastic cell, e.g., benign hypeφlastic cell, refers to slowing, interrupting, aπesting or stopping its growth and metastasis, and does not necessarily indicate a total elimination of the neoplastic growth.
As used herein, "inducing the killing" of the hypeφroliferative cell, e.g., neoplastic cell, e.g., benign hypeφlastic cell, refers to the partial or complete elimination of such cells, and does not necessarily indicate a total elimination of the neoplastic growth.
The terms "induce", "inhibit", "potentiate", "elevate", "increase", "decrease" or the like, denote quantitative differences between two states, refer to at least statistically significant differences between the two states. For example, "an amount effective to inhibit growth of hypeφroliferative cells" means that the rate of growth of the cells will at least statistically significantly different from the untreated cells. Such terms are applied herein to, for example, rates of cell proliferation.
As used herein, the terms "hypeφroliferative", "hypeφlastic", "malignant", and "neoplastic" are used interchangeably, and refer to those cells in an abnormal state or condition characterized by rapid proliferation or neoplasm. The terms are meant to include all types of hypeφroliferative growth, hypeφlastic growth, cancerous growths or oncogenic processes, metastatic tissues or malignantly transformed cells, tissues, or organs, iπespective of histopathologic type or stage of invasiveness. "Pathologic hypeφroliferative" cells occur in disease states characterized by malignant tumor growth or in disease states characterized by benign hypeφroliferative and hypeφlastic cells.
The common medical meaning of the term "neoplasia" refers to "new cell growth" that results as a loss of responsiveness to normal growth controls, e.g., neoplastic cell growth. A "hypeφlasia" refers to cells undergoing an abnormally high rate of growth. However, as used herein, the terms neoplasia and hypeφlasia can be used interchangeably, as their context will reveal, referring generally to cells experiencing abnormal cell growth rates. Neoplasias and hypeφlasias include "tumors," which may be either benign, premalignant or malignant.
The term "carcinoma" is recognized by those skilled in the art and refers to malignancies of epithelial or endocrine tissues including respiratory system carcinomas, gastrointestinal system carcinomas, genitourinary system carcinomas, testicular carcinomas, breast carcinomas, prostatic carcinomas, endocrine system carcinomas, and melanomas. Exemplary carcinomas include those forming from tissue of the cervix, lung, prostate, breast, head and neck, colon and ovary. The term also includes carcinosarcomas, e.g., which include malignant tumors composed of carcinomatous and sarcomatous tissues. An "adenocarcinoma" refers to a carcinoma derived from glandular tissue or in which the tumor cells form recognizable glandular structures.
The term "sarcoma" is recognized by those skilled in the art and refers to malignant tumors of mesenchymal derivation. As used herein, the terms "leukemia" or "leukemic cancer" refers to all cancers or neoplasias of the hemopoietic and immune systems (blood and lymphatic system). These terms refer to a progressive, malignant disease of the blood-forming organs, marked by distorted proliferation and development of leukocytes and their precursors in the blood and bone marrow. Myelolomas refer to other types of tumors of the blood, bone maπow cells. Lymphomas refer to tumors of the lymph tissue. As used herein, the terms "immunoglobuhn" and "antibody" refer to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds. Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as HCVR or VH) and a heavy chain constant region. The heavy chain constant region is comprised of three domains, CHI, CH2 and CH3. Each light chain is comprised of a light chain variable region (abbreviated herein as LCVR or VL) and a light chain constant region. The light chain constant region is comprised of one domain, CL. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL is composed of three CDRs and four FRs, aπanged from amino-terminus to carboxy-terminus in the following order: FRl, CDRl, FR2, CDR2, FR3, CDR3, FR4. The variable regions of the heavy and light chains contain a binding domain that interacts with an antigen. The constant regions of the antibodies may mediate the binding of the immunoglobuhn to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clq) of the classical complement system.
The term "antigen-binding portion" of an antibody (or simply "antibody portion"), as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., a target antigen). Examples of binding fragments encompassed within the term "antigen-binding portion" of an antibody include (a) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHI domains; (b) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (c) a Fd fragment consisting of the VH and CHI domains; (d) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (e) a dAb fragment (Ward et al, (1989) Nature 341:544-546), which consists of a VH domain; and (f) an isolated complementarity determining region (CDR). Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl Acad. Sci. USA 85:5879-5883). Such single chain antibodies are encompassed within the term "antigen-binding portion" of an antibody. These antibody fragments are obtained using conventional techniques known to those with skill in the art, and the fragments are screened for utility in the same manner as are intact antibodies.
The term "monoclonal antibody" as used herein refers to an antibody molecule of single molecular composition. A monoclonal antibody composition displays a single binding specificity and affinity for a particular epitope. Accordingly, the term "human monoclonal antibody" refers to antibodies displaying a single binding specificity which have variable and constant regions derived from human germline immunoglobuhn sequences. In one embodiment, the human monoclonal antibodies are produced by a hybridoma which includes a B cell obtained from a transgenic non-human animal, e.g., a transgenic mouse, having a genome comprising a human heavy chain transgene and a light chain transgene fused to an immortalized cell.
The term "recombinant human antibody", as used herein, is intended to include all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies isolated from an animal (e.g., a mouse) that is transgenic for human immunoglobuhn genes; antibodies expressed using a recombinant expression vector transfected into a host cell, antibodies isolated from a recombinant, combinatorial human antibody library, or antibodies prepared, expressed, created or isolated by any other means that involves splicing of human immunoglobuhn gene sequences to other DNA sequences. Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobuhn sequences. In certain embodiments, however, such recombinant human antibodies are subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo. Methods for inhibiting the activity of hyperproliferative cells
In one aspect, this invention features methods for inhibiting the proliferation, and/or enhancing the killing of, hypeφroliferative cells, by contacting the cells with at least one cytotoxic agent and at least one FGF antagonist. In general, the method includes a step of contacting pathological hypeφroliferative cells with an amount of at least one cytotoxic agent and at least one FGF antagonist, which, in combination, is effective to reduce or inhibit the proliferation of the cell, or induce cell killing. The present method can be performed on cells in culture, e.g., in vitro or ex vivo, or can be performed on cells present in a subject, e.g., as part of an in vivo therapeutic protocol. The therapeutic regimen can be carried out on a human or on other animal subjects. The enhanced therapeutic effectiveness of the combination therapy of the present invention represents a promising alternative to conventional highly toxic regimens of anticancer agents.
While the FGF antagonists can be utilized alone, the subject methods are preferably combined with other anticancer agents, e.g., antimicrotubule agents, topoisomerase I inhibitors, topoisomerase II inhibitors, antimetabohtes, mitotic inhibitors, alkylating agents, intercalating agents, agents capable of interfering with a signal transduction pathway (e.g., a protein kinase C inhibitor, e.g., an anti-hormone, e.g., an antibody against growth factor receptors), agents that promotes apoptosis and/or necrosis, an interferon, an interleukin, a tumor necrosis factor, and radiation. The enhanced, and sometimes synergistic, effect of the FGF inhibitor(s) (e.g., suramin, e.g., FGF antibody) with anticancer agents (e.g., paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, 5-fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, Herceptin, anti- CD20, leuprolide (Lupron) and flutamide), in addition to improving the efficacy of these anticancer agents, may allow for the administration of lower doses of these anticancer agents, thus reducing the induction of side effects in a subject.
The subject method can be useful in treating malignancies of the various organ systems, such as those affecting lung, breast, lymphoid, gastrointestinal (e.g., colon), and genitourinary tract (e.g., prostate), pharynx, as well as adenocarcinomas which include malignancies such as most colon cancer, rectal cancer, renal cell carcinoma, prostate cancer and/or testicular tumors, non-small cell carcinoma of the lung, cancer of the small intestine and cancer of the esophagus. Exemplary solid tumors that can be treated include: fibrosarcoma, myosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, gastric cancer, esophageal cancer, colon carcinoma, rectal cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, uterine cancer, cancer of the head and neck, skin cancer, brain cancer, squamous cell carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinoma, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, testicular cancer, lung carcinoma, small cell lung carcinoma, non-small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, meningioma, melanoma, neuroblastoma, retinoblastoma, leukemia, lymphoma, or Kaposi sarcoma.
The subject method may also be used to inhibit the proliferation or induce the killing of hypeφlastic/neoplastic cells of hematopoietic origin, e.g., arising from myeloid, lymphoid or erythroid lineages, or precursor cells thereof. For instance, the present invention contemplates the treatment of various myeloid disorders including, but not limited to, acute promyeloid leukemia (APML), acute myelogenous leukemia (AML) and chronic myelogenous leukemia (CML) (reviewed in Vaickus, L. (1991) Crit. Rev. Oncol/Hemotol. 11:267-97). Lymphoid malignancies which may be treated by the subject method include, but are not limited to acute lymphoblastic leukemia (ALL), which includes B-lineage ALL and T-lineage ALL, chronic lymphocytic leukemia (CLL), prolymphocytic leukemia (PLL), hairy cell leukemia (HLL) and
Waldenstrom's macroglobulinemia (WM). Additional forms of malignant lymphomas contemplated by the treatment method of the present invention include, but are not limited to, non-Hodgkin's lymphoma and variants thereof, peripheral T-cell lymphomas, adult T-cell leukemia/lymphoma (ATL), cutaneous T-cell lymphoma (CTCL), large granular lymphocytic leukemia (LGF) and Hodgkin's disease.
The subject method may also be used to inhibit the proliferation or induce the killing of benign hypeφlastic growth. FGF antagonists
The FGF antagonist used in the methods and compositions of the present invention can be a chemical, e.g., a protein or a peptide, or a small molecule. Preferably, the FGF antagonist is an inhibitor of bFGF, aFGF, or an inhibitor of both. For example, the FGF antagonist can be capable of binding to an FGF molecule, a molecule that facilitates the binding of FGF to a receptor, or an FGF receptor and, thereby, blocking the binding of an FGF molecule to a receptor (e.g., a tyrosine kinase receptor and/or a heparan sulfate proteoglycan). The FGF antagonist may also down regulate FGF receptor action, e.g., blocks the intracellular signalling initiated by activation of the FGF receptor. E.g., the inhibitor binds to the intracellular domain of the FGF receptor, or blocks a downstream action that is consequence of activation of the FGF receptor.
Examples of chemicals that may antagonize FGF action include suramin, structural analogs of suramin, pentosan polysulfate, scopolamine, angiostatin, sprouty, estradiol, carboxymethylbenzylamme dextran (CMDB7), suradista, insulin-like growth factor binding protein-3, ethanol, heparin (e.g., 6-O-desulfated heparin), low molecular weight heparin, protamine sulfate, cyclosporin A, or RNA ligands for bFGF. The FGF antagonist may also be a small molecule, e.g., a member of a combinatorial library. A prefeπed FGF antagonist is suramin. Preferably, suramin is present in a concentration sufficient to block the resistance to anticancer agents induced by FGF (e.g., bFGF and/or aFGF), but is not sufficient to produce significant cytotoxicity (e.g., significant inhibition of cell proliferation, e.g., significant cell death) in human and/or animal tumor cells, not sufficient to cause significant cell cycle aπest, and/or not sufficient to produce measurable antitumor effect in a subject, e.g., a human subject. Preferably, suramin is administered in an amount that results in a plasma concentration ranging from about 0.1 to 100 microgram/ml, preferably about 1 to 85 microgram/ml, more preferably, about 5 to 60 microgram/ml, even more preferably, about 10 to 50 microgram/ml, and most preferably, 15 to 45 microgram/ml, of the plasma concentration. Examples of proteins or peptide that may antagonize FGF activity include antibodies, e.g., a monoclonal antibody or an antigen fragment thereof, as described below. Alternatively, the FGF antagonist is a fragment of the FGF molecule. Preferably, the FGF fragment competes with an FGF molecule for binding to the receptor. The FGF antagonist may also be transforming growth factor beta or a fragment thereof.
Immunoglobulins Examples of proteins or peptide that may antagonize FGF activity include antibodies, e.g., a monoclonal antibody, a murine antibody or a human antibody, or an antigen-binding fragment thereof. Preferably, the monoclonal antibody is a human antibody. For example, the human antibody can be produced by a hybridoma which includes a B cell obtained from a transgenic non-human animal, e.g., a transgenic mouse, having a genome comprising a human heavy chain transgene and a light chain transgene fused to an immortalized cell. The antibodies can be of the various isotypes, including: IgG (e.g., IgGl, IgG2, IgG3, IgG4), IgM, IgNl, IgA2, IgD, of IgE. Preferably, the antibody is an IgG isotype. The antibodies can be full-length (e.g., an IgGl or IgG4 antibody) or can include only an antigen-binding fragment (e.g., a Fab, F(ab')2, Fv or a single chain Fv fragment).
Several antibodies against bFGF and FGF are commercially available, and can be readily used in the methods and compositions of the invention. Examples of antibodies to bFGF that are commercially available include: (a) Sigma F 9666, anti- human, monoclonal, clone no. FA-88, host: mouse, type:IgG2a; (b) Sigma F 5521, polyclonal, host: rabbit; and (c) Oncogene PC316L, anti-bovine+anti-human, polyclonal, host: rabbit, type IgG.
Examples of antibodies to aFGF that are commercially available include: (a) Sigma F 6162, anti-human, monoclonal, clone no. FB-8, host: mouse, type: IgGl; (b) Sigma F 5537, anti-human, polyclonal, host: rabbit, type: IgG; (c) Sigma F 3393, anti- bovine, polyclonal, host: rabbit; (d) Oncogene PC15, anti-human, polyclonal, AB-1, host: rabbit, type: IgG; (e) Oncogene PC 16, anti-human, polyclonal, AB-2, host: rabbit, type: IgG; (f) Oncogene GF22, anti-human, monoclonal, clone no. 3H3, AB-3, host: mouse, type: IgGl; (g) Oncogene GF23L, anti-human, monoclonal, clone no. 98, AB-4, host: mouse, type: IgGl; (h) Oncogene GF24, anti-human, monoclonal, clone no. 52, AB-5, host: mouse, type: IgG2b; and (i) Oncogene pC194L, anti-human, polyclonal, AB-6, host: goat, type: IgG.
Alternatively, the anti-FGF antibody can be made by methods known in the art. For example, an anti-FGF antibody can be produced by a variety of techniques, including conventional monoclonal antibody methodology e.g., the standard somatic cell hybridization technique of Kohler and Milstein (1975) Nature 256: 495. Although somatic cell hybridization procedures are prefeπed, in principle, other techniques for producing monoclonal antibody can be employed, e.g., viral or oncogenic transformation of B lymphocytes. The prefeπed animal system for preparing hybridomas is the murine system. Hybridoma production in the mouse is a very well- established procedure. Immunization protocols and techniques for isolation of immunized splenocytes for fusion are known in the art. Fusion partners (e.g., murine myeloma cells) and fusion procedures are also known. Human monoclonal antibodies (mAbs) directed against human proteins can be generated using transgenic mice carrying the complete human immune system rather than the mouse system. Splenocytes from these transgenic mice immunized with the antigen of interest are used to produce hybridomas that secrete human mAbs with specific affinities for epitopes from a human protein (see, e.g., Wood et al. International Application WO 91/00906, Kucherlapati et al. PCT publication WO 91/10741; Lonberg et al. International Application WO 92/03918; Kay et al. International Application 92/03917; Lonberg, N. et al (1994) Nature 368:856-859; Green, L.L. et al. (1994) Nature Genet. 7:13-21; Morrison, S.L. et al. (1994) Proc. Natl. Acad. Sci. USA 91 :6851-6855; Bruggeman et al. (1993) Year Immunol 7:33-40; Tuaillon et al. (1993) Proc. Natl Acad. Sci. USA 90:3720-3724; Bruggeman et al. (1991) Eur J Immunol 21 :1323-1326).
Monoclonal antibodies can also be generated by other methods known to those skilled in the art of recombinant DNA technology. An alternative method, refeπed to as the "combinatorial antibody display" method, has been developed to identify and isolate antibody fragments having a particular antigen specificity, and can be utilized to produce monoclonal antibodies (for descriptions of combinatorial antibody display see e.g., Sastry et al. (1989) Proc. Natl. Acad. Sci. USA 86:5728; Huse et al. (1989) Science 246:1275; and Orlandi et al. (1989) Proc. Natl Acad. Sci. USA 86:3833; Larrick et al. (1991), Biotechniques 11 :152-156; Larrick et al (1991), Methods: Companion to Methods in Enzymology 2:106-110).
In an illustrative embodiment, RNA is isolated from B lymphocytes, for example, peripheral blood cells, bone maπow, or spleen preparations, using standard protocols (e.g., U.S. Patent No. 4,683,202; Orlandi, et α/.(1989) Proc. Natl. Acad. Sci. USA 86:3833-3837; Sastry et al. (1989) Proc. Natl Acad. Sci. USA 86:5728-5732; and Huse et al. (1989) Science 246:1275-1281.) First-strand cDNA is synthesized using primers specific for the constant region of the heavy chain(s) and each of the K and λ light chains, as well as primers for the signal sequence. Using variable region PCR primers, the variable regions of both heavy and light chains are amplified, each alone or in combination, and ligated into appropriate vectors for further manipulation in generating the display packages. Ohgonucleotide primers useful in amplification protocols may be unique or degenerate or incoφorate inosine at degenerate positions. Restriction endonuclease recognition sequences may also be incoφorated into the primers to allow for the cloning of the amplified fragment into a vector in a predetermined reading frame for expression.
The V-gene library cloned from the immunization-derived antibody repertoire can be expressed by a population of display packages, preferably derived from filamentous phage, to form an antibody display library. Ideally, the display package comprises a system that allows the sampling of very large variegated antibody display libraries, rapid sorting after each affinity separation round, and easy isolation of the antibody gene from purified display packages. In addition to commercially available kits for generating phage display libraries (e.g., the Pharmacia Recombinant Phage
Antibody System, catalog no. 27-9400-01; and the Stratagene SurfZAPτM phage display kit, catalog no. 240612), examples of methods and reagents particularly amenable for use in generating a variegated antibody display library can be found in, for example, Ladner et al U.S. Patent No. 5,223,409; Kang et al. International Publication No. WO 92/18619; Dower et al. International Publication No. WO 91/17271 ; Winter et al International Publication WO 92/20791; Markland et al. International Publication No. WO 92/15679; Breitling et al. International Publication WO 93/01288; McCafferty et al. International Publication No. WO 92/01047; Gaπard et al. International Publication No. WO 92/09690; Ladner et al. International Publication No. WO 90/02809; Fuchs et al. (1991) Bio/Technology 9:1370-1372; Hay et al. (1992) Hum Antibody Hybridomas 3:81-85; Huse et al. (1989) Science 246:1275- 1281; Griffiths et al. (1993) EMBO J 12:725-734; Hawkins et al. (1992) J Mol Biol 226:889-896; Clackson et al. (1991) Nature 352:624-628; Gram et al. (1992) Proc. Natl. Acad. Sci. USA 89:3576-3580; Gaπad et al. (1991) Bio/Technology 9:1373-1377; Hoogenboom et al. (1991) Nuc Acid Res 19:4133-4137; and Barbas et al. (1991) Proc. Natl Acad. Sci. USA 88:7978-7982.
In certain embodiments, the V region domains of heavy and light chains can be expressed on the same polypeptide, joined by a flexible linker to form a single-chain Fv fragment, and the scFV gene subsequently cloned into the desired expression vector or phage genome. As generally described in McCafferty et al. (1990) Nature 348:552- 554, complete VH and VL domains of an antibody, joined by a flexible (Gly_ι-Ser)3 linker can be used to produce a single chain antibody which can render the display package separable based on antigen affinity. Isolated scFV antibodies immunoreactive with the antigen can subsequently be formulated into a pharmaceutical preparation for use in the subject method.
Once displayed on the surface of a display package (e.g., filamentous phage), the antibody library is screened with the target antigen, or peptide fragment thereof, to identify and isolate packages that express an antibody having specificity for the target antigen. Nucleic acid encoding the selected antibody can be recovered from the display package (e.g., from the phage genome) and subcloned into other expression vectors by standard recombinant DNA techniques.
Specific antibody molecules with high affinities for a surface protein can be made according to methods known to those in the art, e.g., methods involving screening of libraries (Ladner, R.C., et al, U.S. Patent 5,233,409; Ladner, R.C., et al, U.S. Patent 5,403,484). Further, the methods of these libraries can be used in screens to obtain binding determinants that are mimetics of the structural determinants of antibodies.
In particular, the Fv binding surface of a particular antibody molecule interacts with its target ligand according to principles of protein-protein interactions, hence sequence data for VH and VL (the latter of which may be of the K or λ chain type) is the basis for protein engineering techniques known to those with skill in the art. Details of the protein surface that comprises the binding determinants can be obtained from antibody sequence information, by a modeling procedure using previously determined three-dimensional structures from other antibodies obtained from NMR studies or crytallographic data. See for example Bajorath, J. and S. Sheriff, (1996) Proteins: Struct., Fund., and Genet. 24, 152-157; Webster, D.M. and A. R. Rees (1995) "Molecular modeling of antibody-combining sites" in S. Paul, Ed., Methods in Molecular Biol. 51, Antibody Engineering Protocols, Humana Press, Totowa, NJ, pp 17-49; and Johnson, G. et al. (1995) "Seqhunt: A program to screen aligned nucleotide and amino acid sequences," in Methods in Molecular Biol.51 , op. cit., pp 1-15.
In one embodiment, a variegated peptide library is expressed by a population of display packages to form a peptide display library. Ideally, the display package comprises a system that allows the sampling of very large variegated peptide display libraries, rapid sorting after each affinity separation round, and easy isolation of the peptide-encoding gene from purified display packages. Peptide display libraries can be in, e.g., prokaryotic organisms and viruses, which can be amplified quickly, are relatively easy to manipulate, and which allows the creation of large number of clones. Prefeπed display packages include, for example, vegetative bacterial cells, bacterial spores, and most preferably, bacterial viruses (especially DNA viruses). However, the present invention also contemplates the use of eukaryotic cells, including yeast and their spores, as potential display packages. Phage display libraries are described above. The term "modified" or "recombinant" antibody is also intended to include antibodies, such as monoclonal antibodies, chimeric antibodies, and humanized antibodies which have been modified by, e.g., deleting, adding, or substituting portions of the antibody. For example, an antibody can be modified by deleting the hinge region, thus generating a monovalent antibody. Any modification is within the scope of the invention so long as the antibody has at least one antigen binding region specific. Chimeric mouse-human monoclonal antibodies (i.e., chimeric antibodies) can be produced by recombinant DNA techniques known in the art (see Robinson et al, International Patent Publication PCT/US86/02269; Akira, et al, European Patent Application 184,187; Taniguchi, M., European Patent Application 171,496; Morrison et al, European Patent Application 173,494; Neuberger et al, International Application WO 86/01533; Cabilly et al. U.S. Patent No. 4,816,567; Cabilly et al, European Patent Application 125,023; Better et al. (1988) Science 240:1041-1043); Liu et al. (1987) Proc. Natl. Acad. Sci. USA 84:3439-3443; Liu et al. (1987) J. Immunol. 139:3521- 3526; Sun et al. (1987) Proc. Natl. Acad. Sci. USA 84:214-218; Nishimura et al. (1987) Cane. Res. 47:999-1005; Wood et al. (1985) Nature 314:446-449; and Shaw et al. (1988) J. Natl Cancer Inst. 80:1553-1559).
The chimeric antibody can be further humanized by replacing sequences of the Fv variable region which are not directly involved in antigen binding with equivalent sequences from human Fv variable regions. General reviews of humanized chimeric antibodies are provided by Morrison, S. L. (1985) Science 229:1202-1207 and by Oi et al. (1986) BioTechniques 4:214. Those methods include isolating, manipulating, and expressing the nucleic acid sequences that encode all or part of immunoglobuhn Fv variable regions from at least one of a heavy or light chain. Sources of such nucleic acid are well known to those skilled in the art and, for example, may be obtained from 7E3, an anti-GPIIi,IIIa antibody producing hybridoma. The recombinant DNA encoding the chimeric antibody, or fragment thereof, can then be cloned into an appropriate expression vector. Suitable humanized antibodies can alternatively be produced by CDR substitution U.S. Patent 5,225,539; Jones et al. (1986) Nature 321 :552-525; Verhoeyan et al. (1988) Science 239:1534; and Beidler et al. (1988) J. Immunol. 141 :4053-4060.
All of the CDRs of a particular human antibody may be replaced with at least a portion of a non-human CDR or only some of the CDRs may be replaced with non- human CDRs. It is only necessary to replace the number of CDRs required for binding of the humanized antibody to the Fc receptor.
An antibody can be humanized by any method, which is capable of replacing at least a portion of a CDR of a human antibody with a CDR derived from a non-human antibody. Winter describes a method which may be used to prepare the humanized antibodies of the present invention (UK Patent Application GB 2188638A). The human CDRs may be replaced with non-human CDRs using ohgonucleotide site- directed mutagenesis.
Also within the scope of the invention are chimeric and humanized antibodies in which specific amino acids have been substituted, deleted or added. In particular, prefeπed humanized antibodies have amino acid substitutions in the framework region, such as to improve binding to the antigen. For example, in a humanized antibody having mouse CDRs, amino acids located in the human framework region can be replaced with the amino acids located at the coπesponding positions in the mouse antibody. Such substitutions are known to improve binding of humanized antibodies to the antigen in some instances. Antibodies in which amino acids have been added, deleted, or substituted are refeπed to herein as modified antibodies or altered antibodies. Cytotoxic Agents
The FGF antagonist is preferably administered in combination with at least one cytotoxic agent. The term "in combination" in this context means that the agents are given substantially contemporaneously, either simultaneously or sequentially. If given sequentially, at the onset of administration of the second compound, the first of the two compounds is preferably still detectable at effective concentrations at the site where treatment effect is desired.
For example, the FGF antagonists can be used in combination therapy with conventional cancer chemotherapeutics. Conventional treatment regimens for leukemia and for other tumors include radiation, antitumor agents, interferons, interleukins, tumor necrosis factors, or a combination of two or more of these agents.
For example, the subject method may involve, in addition to the use of at least one FGF inhibitor, one or more other antitumor agents. Exemplary combination therapies include the use of such agents as, e.g., the ones described in Table 2 below, including but not limited to: antitubulin/antimicrotubule agents, e.g., paclitaxel, vincristine, vinblastine, vindesine, vinorelbin, taxotere (e.g., Docetaxel); topoisomerase I inhibitors, e.g., topotecan, camptothecin, irinotecan hydrochloride (e.g., Camptosar); topoisomerase II inhibitors, e.g., doxorubicin, etoposide, mitoxantrone, daunorubicin, idarubicin, teniposide, amsacrine, epirubicin, merbarone, piroxantrone hydrochloride; antimetabohtes, e.g., 5-fluorouracil, methotrexate, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, cytarabine (Ara-C), trimetrexate, gemcitabine, acivicin, alanosine, pyrazofurin, N-Phosphoracetyl-L- Asparate (PALA), pentostatin, 5-azacitidine, 5-Aza-2'-deoxycytidine, adenosine arabinoside (Ara-A), cladribine, ftorafur, UFT (combination of uracil and ftorafur), 5-fluoro-2'-deoxyuridine, 5'-deoxy- 5-fluorouridine, tiazofurin; alkylating agents, e.g., cisplatin, carboplatin, oxaliplatin, mitomycin C, BCNU (e.g., Carmustine), melphalan, thiotepa, busulfan, chlorambucil, plicamycin, dacarbazine, ifosfamide phosphate, cyclophosphamide, nitrogen mustard, uracil mustard, pipobroman, 4-ipomeanol; agents acting via interfering with signal transduction pathway an or yet-unknown action mechanisms, e.g., dihydrolenperone, spiromustine, geldanamycins, cytochalasins, depsipeptide; anti-hormones, e.g., leuprolide (e.g., Lupron), ketoconazole, tamoxifen, goserelin (e.g., Zoladex), flutamide, 4'-cyano-3-(4-fluorophenylsulphonyl)-2-hydroxy-2-methyl-3'-(trifluoromethyl) propionanilide; agents that inhibit the binding of a growth factor to a growth factor receptor, e.g., Herceptin, anti-CD20 (Rituxan); interferons, e.g., interferon alpha, interferon beta, interferon gamma; interleukins, e.g., interleukin 2, interleukin 4, interleukin 12; tumor necrosis factors; and radiation.
Examples of additional agents that can be used in combination with the FGF inhibitors include hydroxyurea, azathioprine, aminopterin, trimethoprin, pyrimethamine, pyritrexim, DDMP (2,4 diamino- 5(3',4' dichlorophenyl)6 methylpyrimidine), 5,10- dideazatetrahydrofolate,10-propargyl-5,8 dideazafolate (CB3717), 10-ethyl-lO-deaza- aminopterin, deoxycytidine, 5-aza-cytosine arabinoside, N-4-palmitoyl-ara C, 2'-azido-2'- deoxy-ara C, N4-behenoyl-ara C, CCNU (lomustine), estramustine, MeCCNU, triethylene melamine, trenimon, dimethyl busulfan, streptozotocin, chlorozotocin, procarbazine, hexamethylmelamine (Altretamine), pentamethylmelamine (PMM), tetraplatin, oxaliplatin, platinum-DACH, aziridinylbenzoqumone (AZQ), bleomycin, tallysomycin Sιob, liblomycin, pepleomycin, asparaginase (Elspar), pegaspargase (Oncaspar), Cladrbine (leustatin), porfimer sodium (Photofrin), amonofide, deoxyspergualin, dihydrolenperone, flavone acetic acid, gallium nitrate, and hexamethylene bisacetamine (HMB A).
Particular combinations of cytotoxic agents can be used depending on the type of hypeφroliferative disorder to be treated. For example, in addition to radiation, the following drugs, usually in combinations with each other, are often used to treat acute leukemias: vincristine, prednisone, methotrexate, mercaptopurine, cyclophosphamide, and cytarabine. In chronic leukemia, for example, fludarabine, busulfan, melphalan, and chlorambucil can be used in combination. In lung cancer, a combination of paclitaxel and carboplatin, or a combination of gemcitabine and cisplatin is used. In hormone refractory prostate cancer, a combination of estramustine phosphate and taxotere or a combination of doxorubicin and ketoconazole is used. For metastatic breast cancer, a combination of cyclophosphamide, doxorubicin and 5-fluorouracil is used. For advanced breast cancer that overexpresses the HER2/neu oncogene, a combination of an anti-Her2/neu antibody (e.g., Herceptin) and cisplatin is used. For advanced or metastatic colorectal cancer, a combination of 5-fluorouracil and leucovorin is used. All of the conventional anticancer drugs are highly toxic and tend to make patients quite ill while undergoing treatment; vigorous therapy is based on the premise that unless every cancer cell is destroyed, the residual cells will multiply and cause a relapse. Cytotoxic agents are also used to treat benign hypeφlasia disorders. For example, psoriasis is treated with 5-fluorouracil. Treatment can be initiated with smaller dosages that are less than the optimum dose of the agent. Thereafter, the dosage should be increased by small increments until the optimum effect under the circumstances is reached. For convenience, the total daily dosage may be divided and administered in portions during the day if desired. A therapeutically effective antitumor amount and a prophylactically effective antitumor amount of an FGF inhibitor or a cytotoxic agent is expected to vary from about 0.1 milligram per kilogram of body weight per day (mg/kg/day) to about 100 mg/kg/day.
Compounds which are determined to be effective for the prevention or treatment of tumors or for the prevention or treatment of benign hypeφroliferative disorders in animals, e.g., dogs, rodents (e.g., see the Examples below), may also be useful in treatment of tumors in humans. Those skilled in the art of treating tumor in humans will know, based upon the data obtained in animal studies, the dosage and route of administration of the compound to humans. In general, the dosage and route of administration in humans is expected to be similar to that in animals, when adjusted for body surface area.
Determination of a therapeutically effective antitumor amount and a prophylactically effective antitumor amount of a FGF inhibitor and cytotoxic agent can be readily made by the physician or veterinarian (the "attending clinician"). The dosages may be varied depending upon the requirements of the patient, the severity of the condition being treated and the particular agent being employed. In determining the dose, a number of factors are considered by the attending clinician, including, but not limited to: the specific hypeφlastic/neoplastic cell involved; pharmacodynamic characteristics of the particular agent and its mode and route of administration; the desired time course of treatment; the species of mammal; its size, age, and general health; the specific disease involved; the degree of or involvement or the severity of the disease; the response of the individual patient; the particular compound administered; the mode of administration; the bioavailability characteristics of the preparation administered; the dose regimen selected; the kind of concuπent treatment; and other relevant circumstances. U.S. Patent 5,427,916, for example, describes method for predicting the effectiveness of antineoplastic therapy in individual patients. In one embodiment, the present invention shows that the bFGF level determines the tumor sensitivity to anticancer agents, thus indicating the use of bFGF level in tumors for therapeutic decision making, including the selection of a therapeutic modality, e.g., a particular anticancer treatment , or a dosage thereof, based on the level of FGF expression.
Another aspect of the present invention accordingly relates to kits for carrying out the combined administration of the FGF antagonist/agonist with other therapeutic compounds. In one embodiment, the kit comprises an FGF antagonist/agonist formulated in a pharmaceutical carrier, and at least one cytotoxic agent, formulated as appropriate, in one or more separate pharmaceutical preparations.
Pharmaceutical Compositions In another aspect, the present invention provides pharmaceutically acceptable compositions which comprise a therapeutically-effective amount of at least one FGF antagonist/agonist, formulated together with or without at least one cytotoxic agent, in a pharmaceutically acceptable carrier(s).
In a prefeπed embodiment, as described in detail below, the pharmaceutical compositions of the invention may be specially formulated for administration in solid or liquid form, including those adapted for the following: (a) oral administration, for example, drenches (aqueous or non-aqueous solutions or suspensions), tablets, boluses, powders, granules, pastes, mouthwash, hydrogels; (b) parenteral administration, for example, by subcutaneous, intramuscular or intravenous injection as, for example, a sterile solution or suspension; (c) intracavity administration, e.g., intraperitoneal instillation, intravesical (i.e., urinary bladder) instillation, intrathecal administration, (d) intraorgan administration, e.g., intraprostatical administration, (e) topical application, for example, as a cream, ointment or spray applied to the skin; (f) intravaginal or intrarectal administration, for example, as a pessary, cream, foam, enema, suppository; or (g) aerosol, for example, as an aqueous aerosol, liposomal preparation or solid particles containing the agent(s).
In vitro assays and animal studies as described in the Examples below, or an assay similar thereto (e.g., differing in choice of cells or animal used) can be used to determine an "effective amount" of the FGF antagonist/agonist and/or cytotoxic agent, or combinations thereof. The ordinarily skilled artisan would select an appropriate amount of each individual compound in the combination for use in the aforementioned assays or similar assays. Changes in cell activity or cell proliferation can be used to determine whether the selected amounts are "effective amount" for the particular combination of compounds. The regimen of administration also can affect what constitutes an effective amount. As described, FGF antagonist/agonist can be administered to the subject prior to, simultaneously with, or after the administration of the other cytotoxic agent(s). Further, several divided dosages, as well as staggered dosages, can be administered daily or sequentially, or the dose can be proportionally increased or decreased as indicated by the exigencies of the therapeutic situation.
The phrase "pharmaceutically acceptable" is employed herein to refer to those FGF antagonist/agonist and/or cytotoxic agent, compositions containing such compounds, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
The phrase "pharmaceutically-acceptable carrier" as used herein means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject chemical from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient. Some examples of materials which can serve as pharmaceutically-acceptable carriers include: (a) sugars, such as lactose, glucose and sucrose; (b) starches, such as com starch and potato starch; (c) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (d) powdered tragacanth; (e) malt; (f) gelatin; (g) talc; (h) excipients, such as cocoa butter and suppository waxes; (i) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (j) glycols, such as propylene glycol; (k) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (1) esters, such as ethyl oleate and ethyl laurate; (m) agar; (n) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (o) alginic acid; (p) pyrogen-free water; (q) isotonic saline; (r) Ringer's solution; (s) ethyl alcohol; (t) phosphate buffer solutions; and (u) other non- toxic compatible substances employed in pharmaceutical formulations. Wetting agents, emulsifiers and lubricants, such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.
Examples of pharmaceutically-acceptable antioxidants include: (a) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (b) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (c) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like. Compositions containing the FGF antagonist/agonist and/or cytotoxic agent of the present invention may conveniently be presented in unit dosage form. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration. Generally, out of one hundred per cent, this dosage will range from about 1 per cent to about ninety-nine percent of active ingredient, preferably from about 5 per cent to about 70 per cent, most preferably from about 10 per cent to about 30 per cent.
Compositions of the invention suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, hydrogels, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia), and/or as mouthwashes and the like, each containing a predetermined amount of a FGF antagonist/agonist and/or cytotoxic agent as an active ingredient. A compound may also be administered as a bolus, electuary or paste.
In solid dosage forms of the invention for oral administration (capsules, tablets, pills, dragees, powders, granules, hydrogels and the like), the active ingredient is mixed with one or more pharmaceutically-acceptable carriers, such as sodium citrate or dicalcium phosphate. In the case of capsules, tablets and pills, the pharmaceutical compositions may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.
The tablets, and other solid dosage forms of the pharmaceutical compositions of the present invention, such as dragees, capsules, pills, granules and hydrogels, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes, nanoparticles, hydrogels, and/or microspheres. They may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incoφorating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile injectable medium immediately before use. These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. The active ingredient can also be in micro- encapsulated form, if appropriate, with one or more of the above-described excipients. Liquid dosage forms for oral administration of the FGF antagonist/agonist and/or cytotoxic agent of the invention include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
Suspensions, in addition to the active FGF antagonist/agonist and/or cytotoxic agent may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof. Pharmaceutical compositions of the invention for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more FGF antagonist/agonist and/or cytotoxic agent with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active agent.
Compositions of the present invention which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate. Dosage forms for the topical or transdermal administration of a FGF antagonist/agonist and/or cytotoxic agent include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. The active FGF antagonist/agonist and/or cytotoxic agent may be mixed under sterile conditions with a pharmaceutically-acceptable carrier, and with any preservatives, buffers, or propellants which may be required. The ointments, pastes, creams and gels may contain, in addition to FGF antagonist/agonist and/or cytotoxic agent, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
The FGF antagonist/agonist and/or cytotoxic agent can be alternatively administered by aerosol. This is accomplished by preparing an aqueous aerosol, liposomal preparation or solid particles containing the compound. A nonaqueous (e.g., fluorocarbon propellant) suspension could be used. Sonic nebulizers are prefeπed because they minimize exposing the agent to shear, which can result in degradation of the compound. Transdermal patches have the added advantage of providing controlled delivery of a FGF antagonist/agonist and/or cytotoxic agent to the body. Such dosage forms can be made by dissolving or dispersing the agent in the proper medium. Absoφtion enhancers can also be used to increase the flux of the peptidomimetic across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the peptidomimetic in a polymer matrix or gel.
Ophthalmic formulations, eye ointments, powders, solutions and the like, are also contemplated as being within the scope of this invention. Pharmaceutical compositions of this invention suitable for parenteral and/or intracavity administration comprise one or more FGF antagonist/agonist and/or cytotoxic agent in combination with one or more pharmaceutically-acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
In some cases, in order to prolong the effect of a drug, it is desirable to slow the absoφtion of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amoφhous material having poor water solubility. The rate of absoφtion of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absoφtion of a parenterally-administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.
Injectable or implantable depot forms are made by forming matrices (e.g., microcapsules, e.g., cylinders, e.g., discs) of FGF antagonist/agonist and/or cytotoxic agent in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue. Depot formulations can also be administered directly to tumor-bearing organs and/or cavity, e.g., urinary bladder, e.g., intraperitoneal cavity, e.g., intrathecally to brain tissue, to achieve localized delivery of FGF antagonist/agonist and/or cytotoxic agent.
The term "administration," is intended to include routes of introducing to a subject of the FGF antagonist/agonist and/or cytotoxic agent to perform their intended function. Examples of routes of administration which can be used include injection (subcutaneous, intravenous, parenterally, intraperitoneally, intrathecally, etc.), intravesically (i.e., urinary bladder), intraprostatically, oral, inhalation, rectal and transdermal. The pharmaceutical preparations are of course given by forms suitable for each administration route. For example, these preparations are administered in tablets or capsule form, by injection, inhalation, eye lotion, ointment, suppository, etc. The injection can be bolus or can be bycontinuous infusion. The FGF antagonist/agonist and/or cytotoxic agent can be administered alone, or in conjunction with either another agent as described above or with a pharmaceutically acceptable carrier, or both. The FGF antagonist/agonist and/or cytotoxic agent can be administered prior to the administration of the other agent, simultaneously with the agent, or after the administration of the agent. Furthermore, the FGF antagonist/agonist and/or cytotoxic agent can also be administered in a proform which is converted into its active metabolite, or more active metabolite in vivo. The phrases "parenteral administration" and "administered parenterally" as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticulare, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion.
The phrases "systemic administration," "administered systemically", "peripheral administration" and "administered peripherally" as used herein mean the administration of a FGF antagonist/agonist and/or cytotoxic agent, such that it enters the patient's system and, thus, is subject to metabolism and other like processes. Regardless of the route of administration selected, the FGF antagonist/agonist and/or cytotoxic agent, which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically-acceptable dosage forms by conventional methods known to those of skill in the art.
Vaccines
The present invention also concerns vaccines comprising an immunogenic amount of an FGF polypeptide, or a fragment thereof; an FGF receptor-derived polypeptide (FGFR polypeptide), or a fragment thereof, or a fragment of a proteoglycan (PG) that facilitates the binding of FGF to its receptors, dispersed in a physiologically acceptable, nontoxic vehicle, which amount is effective to immunize a subject, preferably a mammal, more preferably a human, against a neoplastic disease. The FGF or FGFR polypeptides, or the PG may be synthesized or prepared recombinantly or otherwise biologically, to comprise one or more amino acid sequences coπesponding to one or more epitopes of the FGF or FGFR polypeptides, or the PG either in monomeric or multimeric form. Those polypeptides may then be incoφorated into vaccines capable of inducing protective immunity. Techniques for enhancing the antigenicity of such polypeptides include incoφoration into a multimeric structure, binding to a highly immunogenic protein carrier, for example, keyhole limpet hemocyanin (KLH), or diphtheria toxoid, and administration in combination with adjuvants or any other enhancers of immune response.
An amino acid sequence coπesponding to an epitope of an FGF polypeptide either in monomeric or multimeric form may also be obtained by chemical synthetic means or by purification from biological sources including genetically modified microorganisms or their culture media. See Lerner, "Synthetic Vaccines", Sci. Am. 248(2): 66-74 (1983). The polypeptide may be combined in an amino acid sequence with other proteins/polypeptides including fragments of other proteins, as for example, when synthesized as a fusion protein, or linked to other antigenic or non-antigenic polypeptides of synthetic or biological origin. In some instances, it may be desirable to fuse an FGF or FGFR polypeptides, or the PG to an immunogenic and/or antigenic protein or polypeptide, for example, to stimulate efficacy of the vaccine.
The term "an epitope of an FGF or FGFR polypeptides, or the PG " will be understood to include the practical possibility that, in some instances, amino acid sequence variations of a naturally occurring protein or polypeptide may be antigenic and confer protective immunity against neoplastic disease and/or anti-tumorigenic effects. Possible sequence variations include, without limitation, amino acid substitutions, extensions, deletions, truncations, inteφolations and combinations thereof. Such variations fall within the contemplated scope of the invention provided the polypeptide containing these variations is immunogenic and antibodies elicited by such a polypeptide cross-react with naturally occurring FGF polypeptides to a sufficient extent to provide protective immunity and/or anti-tumorigenic activity when administered as a vaccine. Such vaccine compositions can be combined with a physiologically acceptable medium, including immunologically acceptable diluents and carriers as well as commonly employed adjuvants such as Freund's Complete Adjuvant, saponin, alum, and the like. Administration would be in immunologically effective amounts of the FGF or FGFR polypeptides, or the PG, preferably in quantities providing unit doses of from 0.01 to 10.0 micrograms of immunologically active FGF or FGFR polypeptide, or the PG per kilogram of the recipient's body weight. Total protective doses may range from 0.1 to about 100 micrograms of antigen.
Routes of administration, antigen dose, number and frequency of injections are all matters of optimization within the scope of the ordinary skill in the art.
It will further be appreciated that anti-idiotype antibodies to FGF or FGFR polypeptide, or the PG are also useful as vaccines and can be similarly formulated.
EXAMPLES
The invention now being generally described, it will be more readily understood by reference to the following examples which are included merely for puφoses of illustration of certain aspects and embodiments of the present invention, and are not intended to limit the invention. The reagents and experimental protocols used in the appended examples are briefly described below.
Chemicals and reagents
Mouse anti-human aFGF and bFGF monoclonal antibodies were obtained from Sigma; lactate dehydrogenase (LDH) assay, bromodeoxyuridine (BrdU) ELISA and human recombinant r-aFGF and r-bFGF from Boehringer Mannheim. The anti-aFGF antibody reacts with naturally occurring aFGF and human recombinant aFGF and does not cross-react with bFGF (Ichimori, Y. et al (1991) Biochem. Biophys. Res. Commun. 17 '5:291-297). The anti-bFGF antibody was generated using the 18 kDa human r- bFGF, is specific for bFGF and does not cross-react with aFGF (Watanabe, H. et al. (1991) Biochem. Biophys. Res. Commun. 175:229-235). r-aFGF and r-bFGF are monomeric peptides and are identical to the 14 kDa human aFGF or the 18 kDa human bFGF, except for the extra methionine at the amino terminus (Jaye, M. et al. (1986) Science, 233:541-545; Bohlen, P. et al. (1985) EERS Letters 18:177-181).
Tumors and cultures
Human prostate PC3 tumor cells and rat normal intestinal epithelial IΕC6 cells were obtained from the American Type Culture Collection. The rat MAT-LyLu tumor cells and PC3-LN cells were obtained from Dr. John Isaacs and Dr. Joy Ware, respectively.
The original clone of the rat MAT-LyLu tumor cells, upon implantation in the hind limbs of male Copenhagen rats, yielded primary tumors at the implantation site and, as the primary tumor reaches a size equal to, or greater than, 0.5 g, metastasized initially in inguinal lymph nodes and subsequently in the lungs in 50% of animals (Isaacs, J.T. et al. (1981) Invest. Urol 19:20-23). Using serial reimplantation of the lymph node and lung metastases, we obtained subclones that yielded more rapidly growing metastases in 100% of animals were obtained (n=24 for lymph node metastases and n=12 for lung metastases).
Paired primary and metastatic tumors were surgically removed from the same host. Fragments of the non-necrotic portions were cultured as histocultures. Single cell suspensions (>95% viability) were obtained by incubating tumor fragments (~1 g) with collagenase, EDTA, and trypsin. Histocultures (tumor fragments of ~1 mm3) and monolayers were cultured in medium supplemented with 9% heat-inactivated fetal bovine serum (FBS) (Yen, W.C. et al. (1996; Pharm. Res. 13:1305-1312).
Collection and analysis of proteins in conditioned medium
Conditioned medium (CM) was collected from tumor histocultures (50 ml per 50 to 100 mg tumor fragments) and monolayer cultures (40 ml per 8x107 cells), after incubation in serum-free medium for 24 hours (Cavanaugh, P.G. & Nicolson, G.L. (1989) Cancer Res. 49:3928-3933). An aliquot of CM was concentrated 10,000-fold using lyophilization followed by reconstitution in 0.1 mM phenylmethylsulfonyl fluoride in water, and then analyzed on 15% SDS-PAGE (85 V for 2.5 hr). Protein bands were visualized with silver stain reagents. For Western blotting, proteins were transfeπed from polyacrylamide gel to a nitrocellulose filter by electrophoresis, followed by sequential incubation with 5% nonfat dry milk in 100 mM Tris/150 mM NaCl/0.1% Tween 20 (pH 7.6), and 5 microgram/ml aFGF or bFGF monoclonal antibody. The antibody-immunoreactive band was visualized using chemiluminescence blotting.
The levels of aFGF and bFGF in the concentrated CM were quantified by comparing the intensity of their bands on Western blots to the intensity of the bands derived from standard curve samples of r-aFGF and r-bFGF. The standard curves were linear between 3 and 100 ng r-aFGF and between 1 and 160 ng r-bFGF.
Pretreatment with CM and recombinant r-bFGF/r-aFGF Prior to drug treatment, cells were incubated for 4 days with tumor CM or with r-aFGF ad/or r-bFGF-containing medium, supplemented with 1% FBS (final protein concentration adjusted to 2 mg/ml). The medium was renewed every other day.
In vitro antitumor activity evaluation The antiproliferative drug effect in histocultures was measured as inhibition of
3H-thymidine incoφoration quantified by autoradiography (Yen, W.C. et al . (1996; Pharm. Res. 13:1305-1312) and, for monolayers, as inhibition of BrdU incoφoration or as reduction of total proteins by the sulforhodamine B assay (Au, J.L.-S. et al. (1998) Cancer Res. 58:2141-2148). For the latter, the two assays yielded qualitatively similar results. Cell kill induced by 96-hour drug treatment was monitored by the release of LDH into the culture medium. LDH activity was monitored by the conversion of tetrazolium to formazan (detected at 490 nm).
Removal of aFGF and bFGF from CM by immunoprecipitation aFGF or bFGF was immunoprecipitated with their respective monoclonal antibody (1 microgram/ml), in the presence of Protein G PLUS/protein A agarose (Oncogene). This procedure reduced the bFGF level in the original CM to below the detection limit of 5 pg/ml by the ELISA (Oncogene), and the aFGF level in the concentrated CM to below the detection limit by Western blotting.
EXAMPLE I: Contribution of tumor environment and tumor location to resistance to anticancer agents
This example describes three studies showing (a) a novel epigenetic mechanism of broad spectrum chemoresistance that is mediated by extracellular factors present in solid and metastatic tumors, and (b) loss of these factors upon removing tumors from metastatic sites and/or disrupting the tumor microenvironment. The results supporting these conclusions are provided in Table 1 below. The studies were performed using anticancer agents that have diverse structures and action mechanism, including paclitaxel (an antimicrotubule agent), doxorubicin (a topoisomerase II inhibitor) and 5- fluorouracil (an antimetabolite). Similar results were found for all three drugs. The first study compared the chemosensitivity in histocultures of rat primary and metastatic tumors, where the heterogeneous cell types and the 3-dimensional structure of solid tumors are maintained, to the chemosensitivity in the coπesponding monolayer cultures of cells obtained by trypsin-disaggregation of the same tumors. The 2- to 40-fold lower chemosensitivity in histocultures indicates that the unique environment in solid tumors played a role in chemoresistance. The second study shows that histocultures and early monolayer cultures of lung metastases were more resistant than lymph node metastases which in turn were more resistant than subcutaneous primary tumors, indicating that tumor location determined the extent of chemoresistance. The third study shows that monolayers derived from lung and lymph node metastases lost their chemoresistance after three passages and became equally sensitive to drugs as monolayers of primary tumor cells where chemosensitivity remained constant for all passages. This indicates a reversal of chemoresistance in metastatic tumors when cells were removed from the metastatic milieu.
Table 1. Loss of drug resistance, aFGF and bFGF upon disaggregation of metastatic tumors and upon passaging in monolayer cultures. Paired rat primary and metastatic tumors were cultured as histocultures or monolayers. For histocultures, tumors were treated with paclitaxel for 24 hours (12 pairs of primary and lymph node tumors and 2 pairs of primary and lung tumors), and with doxorubicin (3 pairs of primary and lymph node tumors) and 5-fluorouracil (2 pairs of primary and lung tumors) for 96 hours. For monolayers, cells were treated with drugs for 96 hours, and drug effect was measured as inhibition of bromodeoxyuridine incoφoration. NM, not measured. *In histocultures, paclitaxel at 10,000 nM produced <50% inhibition, data shown are the concentrations that produced 30% inhibition. +p< 0.05 for differences between primary and metastatic tumors, two-tailed Student's t-test.
Figure imgf000096_0001
EXAMPLE II: Induction of resistance to anticancer agents due to extracellular factors in solid and metastatic tumors
This example describes the experiments confirming the induction of chemoresistance by extracellular factors in metastatic tumors, and indicating a progressive loss of these factors upon passaging metastatic tumor cells in monolayers.
CM from rat tumor cultures was collected and evaluated for its effect on chemosensitivity. This and subsequent experiments were performed using rat primary tumor cells and human prostate tumor cells. Qualitatively similar results were obtained for both cell types, indicating the general nature of the observations (Figure 1). CM derived from histocultures and early monolayer cultures of metastases (refeπed to as active metastatic tumor CM) induced a 3- to 10-fold resistance to drugs (p<0.05), whereas CM from late monolayer cultures of metastatic tumors and CM from primary tumors (refeπed to as inactive CM) had no effect.
EXAMPLE III: Identification of bFGF and aFGF as some of the extracellular factors that induce resistance to anticancer agents
This example describes the experiments that show the presence of elevated concentrations of aFGF and bFGF in conditioned media (CM) from tumor cells. The identity of the extracellular factors that cause tumor cells to become resistant to anticancer agents was established by comparing the proteins in the active and inactive CM (Table 1). Briefly, the active CM collected from histocultures of primary, lymph node and lung tumors and from early monolayer cultures (passage 0) of same tumors, were compared to the inactive CM collected from late monolayer cultures (passage 3) of same tumors. Immunoblotting was performed with mouse anti-human aFGF and bFGF antibodies. Control lanes with r-aFGF (20 ng) or r-bFGF (5 ng) were processed as a comparison.
The active CM showed 2- to 7-fold higher concentrations of two proteins, which were identified by immunoblotting as aFGF (approximately 14 kDa) and bFGF (approximately 18 kDa). Several observations suggest a cause-and-effect relationship between these proteins and resistance. First, the rank order of aFGF/bFGF concentrations in CM of different tumor culture systems was identical to the rank order of the chemoresistance in these cultures. Second, the progressive loss of these proteins upon passaging the metastatic tumors in monolayers coincided with the diminishing ability of these monolayer CM to induce chemoresistance. Third, as the protein levels in monolayers of metastatic tumors (passage 3) were reduced to the same levels as in monolayers of primary tumors, equal drug sensitivity was attained in both cultures. Fourth, in monolayer cultures, the aFGF/bFGF concentrations in CM significantly coπelated with the relative chemoresistance (pO.OOOl , Pearson test).
EXAMPLE IV: Role of extracellular aFGF and bFGF in resistance to anticancer agents
This example describes six studies which collectively show that (a) extracellular aFGF and bFGF are involved in the induction and maintenance of broad spectrum resistance to anticancer agents; and (b) bFGF but not aFGF was required to induce resistance whereas aFGF amplified the bFGF effect.
The first study used specific inhibitors of extracellular aFGF and bFGF, i.e., monoclonal antibodies (Figure 2A). In the absence of the active CM, treatment with antibodies to aFGF or bFGF did not alter drug activity, indicating no effect of the antibodies on the baseline chemosensitivity. In the presence of the active CM, the bFGF antibody produced a concentration-dependent reversal of the CM-induced chemoresistance, with complete reversal by 5 microgram/ml antibody, whereas a nonspecific antibody (i.e., non-immune rabbit IgG) had no effect. For aFGF, the antibody treatment partially reversed the CM-induced resistance; the maximum reversal was about 60% that was obtained with 1 microgram/ml antibody with no significant additional reversal at a 5 -fold higher concentration. The second study evaluated the effect of removing aFGF and/or bFGF from the active CM by immunoprecipitation. Removal of bFGF abolished the resistance, whereas removal of aFGF only partially reversed the CM effect. These two studies indicate that both aFGF and bFGF were involved in the resistance, but had different roles.
The third study determined whether aFGF and/or bFGF are required for the resistance. Endogenous aFGF and/or bFGF were removed from the active CM by immunoprecipitation, and then reconstituted the CM using recombinant proteins. When either the endogenous aFGF or bFGF were removed from the CM, addition of their respective recombinant counteφarts, at concentrations comparable to the endogenous levels, fully restored the resistance (Figure 2B). When both proteins were removed from the CM, (a) addition of r-aFGF did not induce resistance, whereas addition of r-bFGF produced a concentration-dependent resistance, indicating that bFGF but not aFGF was required for the resistance, and (b) the concentration of exogenous r-bFGF needed to restore the resistance was 55-fold higher than the endogenous bFGF concentration in the native CM or when endogenous aFGF was present (i.e., 50 ng/ml versus about 0.9 ng/ml). The latter finding suggests an amplification of bFGF effect by aFGF. This was confirmed in the fourth study, which shows a complete restoration of chemoresistance when the aFGF/bFGF-depleted CM was reconstituted with both r-aFGF and r-bFGF at physiological concentrations of the endogenous proteins. The fifth study confirmed that aFGF and bFGF were the main cause of chemoresistance, by using only r-aFGF and r-bFGF (i.e., without the immunoprecipitated CM) to induce resistance (Figure 3). When given alone, r-aFGF had no effect whereas r-bFGF induced a concentration-dependent resistance. Combinations of r-aFGF/r-bFGF at concentrations as the endogenous proteins in the CM (i.e., between 0.16 and 0.32 ng/ml r-aFGF plus 1 ng/ml r-bFGF) produced the same extent of resistance as the CM, with an even greater resistance at higher protein concentrations.
The chemoresistance observed above was measured primarily as antiproliferative drug effect. The sixth study measured the cell kill effect of drugs. Similar findings were observed, i.e., the active metastatic tumor CM and r-bFGF induced resistance to cell kill by drugs, whereas r-aFGF did not induce resistance but enhanced the effect of r-bFGF (Figure 4). Therefore, the FGF-induced resistance applies to both antiproliferative and cell kill effects.
EXAMPLE V: In vitro data on the enhancement of sensitivity of tumor cells to anticancer agents using inhibitors of aFGF and bFGF
This example shows that inhibitors of aFGF and/or bFGF enhanced the tumor sensitivity to anticancer agents.
Figure 2A shows that inhibition of aFGF and/or bFGF using their respective monoclonal antibodies enhanced the cytotoxicity of paclitaxel, doxorubicin and 5- fluorouracil against human and rodent tumor cells. Likewise, Figure 2B shows the same findings when anti-aFGF monoclonal antibody and/or anti-bFGF monoclonal antibody were used to remove aFGF and/or bFGF from the cell culture medium. The applicants further show that another inhibitor of aFGF and bFGF, i.e., suramin, also reverses the FGF-induced resistance and enhances the activity of at least 58 anticancer agents under in vitro and/or in vivo conditions (see also Example VIII).
Suramin is a negatively charged molecule that inhibits the action of multiple growth factors including aFGF and bFGF (Middaugh, C.R. et al. (1992) Biochem. 31 :9016-9024). Figure 5 shows the results of in vitro experiments demonstrating that addition of suramin produced a concentration-dependent reversal of the CM-induced resistance to paclitaxel, doxorubicin and 5-fluorouracil in rat MAT-LyLu tumor cells, human prostate PC3 tumor cells, and a metastatic subline of PC3, i.e., PC3-LN cells. Complete reversal was attained at between 10 to 15 μM suramin, which, as shown in Example VI, had no antitumor effect when used alone.
EXAMPLE VI: In vitro data on the enhancement of the activity of anticancer agents using low concentrations of suramin that do not produce significant cytotoxicity
This example shows that the enhancement of the activity of anticancer agents by suramin occurs at low suramin concentrations that were sufficient to fully inhibit the FGF-induced chemoresistance but were not sufficient to produce significant cytotoxicity. Figure 6 shows the concentration-response curves of suramin in the tumor cell lines used in the studies described in Examples V and VII. The concentration of suramin, when used alone, required to result in 50% reduction of bromodeoxyuridine incoφoration (i.e., IC50) were 235 micromolar in rat MAT-LyLu tumor cells, 93 micromolar in human prostate PC3 tumor cells, and 98 micromolar in human prostate PC3-LN tumor cells. These IC50 values are between 6- to 200-fold higher than its concentration needed to fully reverse the FGF-induced resistance (between 1 to 15 micromolar, see Examples V and VII and Table 2). At 1-15 micromolar concentration, suramin did not produce measurable cytotoxicity (i.e., <10% reduction in cell number and/or proliferation index). Hence, the enhancement of the antitumor effect of anticancer drugs by suramin occurs at nontoxic concentrations of suramin.
A second study was performed to determine whether suramin, at concentrations sufficient to inhibit the FGF-induced resistance to anticancer agents, fully inhibited the bFGF binding to its receptor. This study was performed using the rat MAT-LyLu tumor cells as the model tumor cells and using doxorubicin and paclitaxel as the model anticancer agents. The CM used to induce resistance was collected from histocultures of lung metastases (see Table 1). Five microgram/ml anti-bFGF antibody was added.
The results show that the IC5o of doxorubicin and paclitaxel, in the presence of 1.6 micromolar suramin, remained unchanged at 13 and 14 nM, respectively, in the absence or presence of the antibody. This indicates that suramin and the bFGF antibody shared the same action mechanism and that suramin alone at 1-2 micromolar concentrations completely inhibited the bFGF-induced resistance.
EXAMPLE VII: Broad spectrum of anticancer agents exhibit aFGF/bFGF- induced resistance that is reversed by suramin
This example summarizes in Table 2 (set forth below) the data showing that aFGF and bFGF induce resistance to at least 58 anticancer agents with a broad spectrum of mechanisms of actions, including antitubulin/antimicrotubule, topoisomerase I inhibitors, topoisomerase II inhibitors, antimetabohtes, alkylating agents, and agents that interfere with signal transduction pathways.
The experimental conditions are described briefly below. Human prostate PC3 tumor cells were pretreated with aFGF plus bFGF for 96 hours, followed by drug treatment for 96 hours, with or without 15 micromolar suramin. Cell number was measured using the sulforhodamine B assay which measures the total protein concentration. All drugs listed in Table 2 showed (a) resistance induced by aFGF plus bFGF (i.e. higher IC5o), and (b) reversal of the resistance by suramin treatment (i.e. suramin treatment reduced the IC50 to the original values as without FGF treatment). The FGF-induced resistance was dependent on the FGF concentrations, with a higher resistance at higher FGF concentrations.
Table 2. List of anticancer agents which show resistance induced by aFGF/bFGF and reversal of FGF-induced resistance by suramin
Figure imgf000102_0001
Table 2 (continued)
Figure imgf000103_0001
Table 2 (continued)
Figure imgf000104_0001
Table 2 (continued)
Figure imgf000105_0001
Table 2 (continued)
Figure imgf000106_0001
EXAMPLE VIII: aFGF bFGF-induced resistance to anticancer agents is detected in multiple cancer cell lines
This example summarizes in Table 3 (set forth below) the data showing that aFGF and bFGF induce resistance to paclitaxel, doxorubicin, 5-fluorouracil, and/or fludarabine in multiple cancer cell lines derived from different human and animal organs, including prostate, lung, pharynx, colon, breast and blood, and a human breast cancer cell line that is stably transfected with the mdrl gene encoding for p- glycoprotein that is responsible for drug efflux. The experimental conditions are described briefly below. Cells were pretreated with human recombinant r-aFGF and/or r-bFGF for 96 hours and then with drugs for 96 hours. For the studies on cells derived from the human prostate, lung, pharynx, colon and breast cancer cells, the drug effect was determined by the inhibition of BrdU incorporation. For the study on the human leukemic cells, the drug effect was measured by the MTT assay, which measures the number of metabolically active cells (Alley, M.C. et al (1988) Cancer Res 48:589-601).
For the cell lines derived from solid tumors, the results indicate that aFGF alone did not induce resistance, whereas bFGF alone induced resistance in five cell lines A549,(prostate PC3, breast MCF7, mdrl -transfected breast BC19, lung A549, prostate MAT-LyLu) but not in the remaining two cell lines (pharynx FaDu, colon HT29).
However, combination of r-aFGF and r-bFGF induced resistance in all seven cell lines, indicating different cells have different requirements of aFGF and bFGF. The results further show that r-aFGF amplified the r-bFGF effect by more than 50-fold (e.g. 1 ng/ml r-aFGF plus 1 ng/ml r-bFGF induced greater resistance than 50 ng/ml r-bFGF alone).
For the human leukemic cell line, i.e., CCRF-CEM, combination of r-aFGF (1 ng/ml) and r-bFGF (3 ng/ml) induced resistance to fludarabine; for a 96 hour treatment, the fludarabine concentration required to produce 50% was increased from 4 micromolar in the absence of FGF to 6 micromolar in the presence of FGF. Table 3. FGF-induced resistance occurs in multiple human and rodent tumor cell lines. Pac - paclitaxel; Dox - doxorubicin; 5- VI) -- 5-iluorouracil. The BC 19 cell line is a subline of MCF7 transfected with m rl. * Indicates p< 0.05 compared to control.
o
—1
Figure imgf000108_0001
Figure imgf000109_0001
EXAMPLE IX: In vivo data on the enhancement of the activity of anticancer drugs, against well-established tumors, using nontoxic dose of suramin
This example describes four studies that provide additional data on the enhancement of the antitumor effect of paclitaxel and doxorubicin by suramin in immunodeficient mice bearing several human xenograft tumors.
The activity of paclitaxel and doxorubicin, with or without suramin, was evaluated in immunodeficient mice (male Balb/c nu/nu mice, 6-8 weeks old). Animal care was in accordance with the guidelines at the Ohio State University. Several human tumor models were used, including (a) lung metastases derived from intravenous injection of human PC3-LN tumor cells, (b) lung tumors derived from direct injection of human PC3 cells in lungs, and (c) subcutaneously implanted PC3 tumors. Four studies were performed. In all studies, drug treatment was administered intravenously via a tail vein, after the tumor was established. The first and second studies used the intravenous PC3-LN lung metastasis model. Briefly, human PC3-LN cells (106 cells in 0.1 ml physiologic saline) were injected intravenously, via a tail vein, into immunodeficient mice. This procedure results in metastasis to lungs in 100% of the animals (Ware, J.L. et al. (1985) Exp. Cell Biol. 53:163-169). After 4 weeks, tumor establishment was determined by visual examination of the lungs of two randomly selected animals, and drug treatment in the remaining animals was initiated when these two animals showed at least 5 tumor nodules of ~1 mm diameter. Mice received intravenous injection (over 1 min) of 200 μl of either physiologic saline or a saline solution delivering a chemotherapeutic, 10 mg/kg suramin, or a combination of chemotherapeutic and suramin. The chemotherapeutic was doxorubicin (5 mg/kg) for the first study and paclitaxel (Taxol, 15 mg/kg) for the second study. Treatment was given twice weekly for three weeks. Preliminary pharmacokinetic data in rodents indicate that these doses would result in plasma concentrations of approximately 10 nM for doxorubicin, 5-7 nM for paclitaxel and 10 micromolar for suramin at 72 hours. As shown in Example VI, the suramin concentration was sufficient to reverse the FGF- induced chemoresistance. The doxorubicin and paclitaxel concentrations are near or above their IC5oin the monolayer cultures of PC3-LN cells (i.e., 17 nM for doxorubicin and 8 nM for paclitaxel). The selected suramin dose has no in vivo antitumor activity against other mouse tumors (Chahinian, A.P. et al. (1998) J. Surg. Oncol. 67 : 104- 111 ; Shin, R., et al. (1997) Scand. J. Gastroenterol 32:824-828). Three days after completion of drug treatments, animals were euthanized and their lungs removed, fixed in Bouin's solution to visualize tumor nodules, and then processed for histologic evaluation. Histologic sections (5 micron thickness) at a depth of between 200-300 μm from the ventral surface and containing all 5 lobes of the lungs were obtained. The lung surface area (counted as number of pixel) occupied by the tumor was calculated as a fraction of the total lung area, using Adobe PhotoShop. The number of tumor cells in residual tumors and the fraction of apoptotic cells in each tumor were also determined microscopically. Because apoptotic cells disappear over time, a second measure of the extent of apoptosis was the density of non-apoptotic cells in the residual tumors. This was determined by counting the number of non-apoptotic tumor cells in randomly selected microscopic fields at 400 x magnification. On average, ten fields per animal, or >1,500 cells in the control and suramin groups and >600 cells in the doxorubicin group were counted. In the case of combination therapy where fewer than 5 tumor nodules remained per animal, all residual cells (between 20 to 600 cells per animal) were counted.
Both studies show that suramin enhanced the in vivo antitumor effect of drugs in mice bearing well-established human lung metastases. Table 4 shows the results for doxorubicin. Suramin alone had no significant antitumor effect, nor toxicity. Doxorubicin alone did not eradicate tumors, but reduced the tumor size by -80%, tripled the fraction of apoptotic cells and halved the density of nonapoptotic cells in residual tumors, and caused a -20% loss in body weight. All animals in the control and single agent groups had visible tumors on the ventral and dorsal surfaces of the lungs, whereas only 40% of animals in the doxorubicin/suramin combination group showed visible tumors on the lung surface. Histologic section of lung tissues in all animals in the control and single agent groups showed microscopic tumor lesions, whereas only 58% of animals in the doxorubicin/suramin group showed microscopic tumor lesions. Hence, the addition of suramin to doxorubicin therapy significantly enhanced the antitumor effect, resulting in (a) tumor eradication in 42% of animals, and (b) in the remaining 58% of animals which showed residual tumors, further reduction of the tumor size (additional 10-fold), reduction of density of non- apoptotic tumor cells (additional 4-fold), and enhancement of the apoptotic cell fraction (additional 3-fold). Addition of suramin to doxorubicin did not enhance the body weight loss, indicating that suramin did not enhance the host toxicity of doxorubicin.
Table 4. Suramin enhances the in vivo antitumor effect of doxorubicin in mice bearing human lung metastases established by intravenous injection of tumor cells.
The average pretreatment weights for the four groups ranged from 21 g to 22 g. Animals that did not show visible tumors on the lung surface nor microscopic lesions in 5 random histologic sections are considered tumor-free. Mean±SD. *p<0.05 compared to the control and suramin groups. tp<0.05 compared to all other groups.
Figure imgf000112_0001
Similar findings were observed for the effect of suramin on paclitaxel. Table 5 shows that suramin alone had no significant antitumor effect, nor toxicity. Paclitaxel alone reduced the density of nonapoptotic cells by -70%, increased the fraction of apoptotic cells by 13-fold, and caused a -7% loss in body weight. The addition of suramin to paclitaxel therapy significantly enhanced the antitumor effect, resulting in further reduction of the density of non-apoptotic tumor cells (additional 30-fold), and nearly doubled the apoptotic cell fraction. Addition of suramin to paclitaxel therapy did not enhance the body weight loss, indicating that suramin did not enhance the host toxicity of paclitaxel.
I l l Table 5. Suramin enhances the in vivo antitumor effect of paclitaxel in mice bearing human lung metastases established by intravenous injection of tumor cells. The average pretreatment weights for the four groups ranged from 21 g to 22 g. Mean±SD. *p<0.05 compared to the control and suramin groups. tp<0.05 compared to all other groups.
Figure imgf000113_0001
The third study shows enhancement of doxorubicin activity in lung tumors established by intra-organ tumor cell injection. Briefly, PC3 cells were directly injected into the lungs of immunodeficient mice (2.5xl06 cells per lung). Tumor establishment (on day 5) was verified by visual examination of a randomly selected animal. The remaining animals were given intravenous bolus injection of doxorubicin (three doses of 10 mg/kg each given on day 5, 8 and 11) alone, suramin alone (three doses of 20 mg/kg), or a combination of both drugs. Animals were euthanized on day 12. Lungs were removed and examined visually for residual tumors. Histologic sections were obtained and examined microscopically, as described for the first two studies. No animals in the untreated control group (n=4) and the suramin group (n=3) were free of tumors, whereas the doxorubicin group (n=4) showed one tumor- free animal (25%), and all animals in the doxorubicin/suramin combination group (n=5) was tumor- free (100%). The differences between the combination group and the other three groups were significant (p<0.04). These data show that suramin enhanced the antitumor activity of doxorubicin.
The fourth study shows that suramin enhanced the antitumor effect of doxorubicin in subcutaneous bulky tumors. Briefly, PC3 cells (5xl06 cells) were injected subcutaneously. Drug treatments (suramin 10 mg/kg, doxorubicin 5 mg/kg, or a combination of both drugs, given twice weekly for 3 weeks) were initiated after tumor establishment. The average tumor size was 150 mg at the time of first treatment.
Figure 7 A shows (a) a 3- to 4-fold increase of tumor size in the untreated control group and suramin group, indicating that suramin alone had no antitumor activity, (b) doxorubicin alone reduced the tumor growth but did not cause tumor regression, (c) doxorubicin + suramin combination reduced the tumor growth during the first week, followed by tumor regression until the end of the 3-week treatment. Results on the animal body weight (Figure 7B) indicate (a) suramin alone did not cause body weight loss, (b) doxorubicin produced a 17% body weight loss, and (c) addition of suramin to doxorubicin did not enhance body weight loss.
EXAMPLE X: Relationship between aFGF and bFGF levels in tumor, tumor size, and tumor sensitivity to anticancer agents
This example demonstrates a correlation between aFGF and bFGF level in tumor and tumor size, and a correlation between bFGF level in tumor and tumor sensitivity to anticancer agents.
The first study examined the effect of the presence of tumor cells on the extracellular bFGF levels in tissues. Briefly, human prostate PC3 tumor cells were implanted intradermally or directly into the lungs of immunodeficient mice. Tumors were removed from animals and -10 mg were cultured as histocultures. The culture medium was collected at 24 hours and analyzed for bFGF. The bFGF level in PC3 cells was 2 pg/106 cells (approximately 2 pg/mg), 0.13 pg/mg in normal lung tissue, 0.62 pg/mg in normal skin tissue, 3.2 pg/mg in PC3-containing lung tumor, and 11.3 pg/mg in PC3-containing skin tumor. Hence, the bFGF levels in tumor-bearing tissues exceeded the sum of bFGF levels in tumor cells and normal tissues, indicating that the presence of tumor cells enhanced bFGF level. The enhancement was 25-fold in lungs and 18-fold in skin. For comparison, the mouse brain contained 0.027 pg bFGF per mg tissue and the liver contained 0.636 pg bFGF per mg tissue. The results indicate (a) different bFGF levels in different tissues, and (b) presence of tumor cells significantly enhanced the bFGF levels.
The second study examined the relationship between the extracellular bFGF level and tumor size. Human prostate PC3 tumor cells were implanted subcutaneously in immunodeficient mice. Subcutaneous tumor were removed from host animals after 1 to 7 weeks, and -200 mg were cultured as histocultures. The culture medium was collected at 24 hours and analyzed for bFGF concentration by ELISA. The bFGF level increased with time, from 27 pg/ml in week 1 tumor, to 56 pg/ml in week 2 tumor, to 77 pg/ml in week 3 tumor, and reached a plateau level of between 80 to 87 pg/ml between week 4 and week 7 tumors. The differences between the bFGF levels in weeks 1 , 2 and 3 are statistically significant. This indicates a positive correlation between bFGF level and tumor size.
The third study examined the relationship between the levels of aFGF and bFGF in human tumors and tumor sensitivity to anticancer agents. This study was performed using 96 tumors obtained from human patients, including include bladder, breast, head and neck, ovarian and prostate tumors. Paclitaxel was used as the model drug. Briefly, histocultures of bladder tumors were treated with paclitaxel for 2 hours and the remaining tumors for 96 hours. The drug-induced antiproliferation was measured by inhibition of DNA precursor incoφoration, and apoptosis was measured by morphological changes and by labeling of nicked DNA. aFGF, bFGF and other proteins known to contribute to drug resistance (i.e. the mdrl p-glycoprotein, p53 and bcl-2) were detected by immunohistochemical staining.
Sixty- four % (61/96) of the tumors showed positive bFGF staining. aFGF is highly expressed in bladder tumors. Ovarian and prostate tumors have intermediate aFGF levels with low positive rates in head and neck and breast tumors. aFGF was only found in the cytoplasm. Paclitaxel produced antiproliferation in 84% of the tumors and induced apoptosis in 96% of the tumors. bFGF expression correlated with resistance to paclitaxel-induced antiproliferation (pO.OOl) and paclitaxel-induced apoptosis (p=0.13). Statistical analyses using multiple regression and the Akaike Information
Criterion show that among the pathobiological parameters including tumor stage, grade, proliferation index, and expression of bFGF, p-glycoprotein, p53 and bcl-2 proteins, bFGF expression was the best indicator of resistance to the antiproliferative effect and the overall cytotoxicity (i.e., antiproliferation plus apoptosis) of paclitaxel in tumors obtained from human patients (see Table 6 set forth below). Addition of aFGF to bFGF improves the correlation (Table 6).
Collectively, these results indicate the levels of aFGF and bFGF in tumors is determined by the location of the tumor and the size of the tumor, and that the aFGF/bFGF level, in turn, determines the tumor sensitivity to anticancer agents. The varying FGF level in tumors indicates the use of FGF level for making therapeutic decision for individual patients.
Table 6. Correlation between pathobiological parameters and chemosensitivity.
A higher r2 value (from multiple regression analysis) and a lower AIC value (from Akaike Information Criterion analysis) indicate a better predictive value. For multiple- parameter combinations, the best prognostic indicators are those that give the highest r2 values as well as the lowest AIC values. Statistical analysis was performed using linear regression analysis using REG software. A p value of less than 0.05 is statistically significant.
Figure imgf000117_0001
EXAMPLE XI: Treatment of cancer cells with anticancer agents enhanced extracellular bFGF levels
This example summarizes in Table 7 (set forth below) the data showing that treatment of human prostate PC3 tumor cells with at least 7 anticancer agents with diverse action mechanisms and chemical structures (e.g., antimicrotubules, topoisomerase I inhibitor, topoisomerase II inhibitor, DNA alkylating agents) significantly enhanced the levels of extracellular bFGF.
The experimental conditions are described briefly below. Human prostate PC3 tumor cells were treated with drugs for 72 hours. The remaining cell number at the end of treatment was counted using a Coulter counter. The extracellular bFGF concentration was measured using ELISA. The release of lactate dehydrogenase (LDH) into the extracellular culture medium is indicative of cell death and gives a measure of the release of cellular content into the culture medium. If the increase in extracellular bFGF level is due to the release of bFGF upon cell death and lysis, the increase in bFGF will be accompanied by an increase in LDH in the same extent. This would result in an higher bFGF level and a constant bFGF-to-LDH ratio. On the other hand, an increase in extracellular bFGF due to an increase in bFGF production and release of bFGF from cells by processes other than cell lysis will result in an higher bFGF level and an higher bFGF-to-LDH ratio.
The results, listed in Table 7, show that treatments with all of the tested drugs resulted in 3- to 30-fold higher extracellular bFGF levels (p<0.05). These higher bFGF levels were attained in spite of the up to 80% reduction in cell number, indicating an even greater bFGF production in the drug-treated cells on a per cell basis, relative to untreated controls. The increases in bFGF level were accompanied by higher bFGF-to- LDH ratios, indicating that the increase in extracellular bFGF level was at least in part due to increased production and release of bFGF from live cells.
The enhancement of extracellular bFGF level in tumor cells due to drug treatment indicates that patients that have been treated with chemotherapy are likely to exhibit chemoresistance. This in turn indicates the need of using inhibitors of FGF to treat patients who have failed cancer therapy and have recurrent tumors. Table 7. Drug treatment enhances bFGF production and release into extracellular space
Figure imgf000119_0001
EXAMPLE XII: FGF-induced resistance to anticancer agents is not due to altered drug accumulation nor altered cell proliferation
This example demonstrates that FGF-induced resistance to anticancer agents is a novel mechanism that is different from the previously reported mechanisms of chemoresistance that involve an enhancement of drug efflux and altered cell proliferation.
The experimental conditions are briefly described below. Human prostate PC3 tumor cells and rat MAT-LyLu tumor cells were treated with 3H-paclitaxel (1 nM for human PC3 cells and 10 nM for rat tumor cells), 14C-doxorubicin (50 and 100 nM, respectively), and 3H-5-fluorouracil (500 nM for both cells) (Au, J.L.-S. et al. (1998) supra), with or without with the active CM collected from histocultures of lung metastases or with r-bFGF. The extracellular drug concentrations were selected based on their IC50 (50% inhibitory concentration) in human and rat cells. The plateau intracellular drug concentrations which were attained at 4 hours for all three drugs in PC3 cells, and 1 (doxorubicin and 5-fluorouracil) and 4 hours (paclitaxel) in rat cells, were measured.
The results show that these treatments did not alter the drug accumulation in tumor cells; the respective intracellular concentrations of paclitaxel, doxorubicin and 5- fluorouracil remained unchanged at -0.5, 40, and -0.1 pmol/106 cells in human PC3 tumor cells; and -1, 80, and 24 pmol/106 cells in rat tumor cells (n=6 each). Therefore, the FGF-induced chemoresistance is not due to reduced drug accumulation, as previously reported for overexpression of drug efflux proteins (Lum, B.L. et al. (1993) Cancer 72, 3502-3514; Barrand, M.A. et al. (1997) Gen. Pharmacol. 28, 639-645; Fidler, IJ. (1999) Cancer Chemother. Pharmacol. 43:S3-S10).
Treatment with the CM or r-bFGF also did not alter the doubling time of exponentially growing cells, which remained unchanged at 17 and 24 hours for rat and PC3 tumor cells, respectively. Hence, the FGF-induced chemoresistance is not due to altered cell proliferation.
EXAMPLE XIII: FGFs induce a novel epigenetic mechanism of broad spectrum resistance to anticancer drugs
The findings described herein demonstrate (a) a novel epigenetic mechanism by which cancer cells utilize the unique microenvironment of solid tumors and metastases to elude cytotoxic insult, (b) establish an important role of extracellular growth factors in tumor sensitivity to chemotherapy, and (c) indicate a new treatment paradigm using combinations of chemotherapy with aFGF/bFGF inhibitors.
These results establish a new mechanism of broad spectrum anticancer drug resistance that is mediated primarily by extracellular aFGF/bFGF and does not involve significant changes in intracellular drug accumulation, nor cell proliferation. The reversal of the resistance by inhibition of extracellular aFGF/bFGF suggests a mechanism that involves binding of these proteins to their receptors.
The correlation between the concentrations of extracellular aFGF/bFGF and tumor location and the progressive loss of these proteins upon disaggregation of metastatic tumors and passaging the disaggregated cells in monolayers indicate a relationship between the aFGF/bFGF concentrations and the tumor growth environment. The findings disclosed herein of higher bFGF concentrations in metastases, compared to primary tumors, is consistent with the higher urinary bFGF concentration observed in patients with metastatic cancer compared to localized cancer (Huang, A. et al. (1994) Exp. Cell Res. 213, 335-339). The significant resistance induced by aFGF/bFGF at concentrations found in the active metastatic tumor CM and in patients (Huang, A. et al. (1994, supra) indicates the importance and clinical relevance of this resistance mechanism, and suggests elevated aFGF/bFGF concentrations in metastases as an important mechanism that limits the efficacy of chemotherapy in metastatic disease. The extent of in vivo chemoresistance may be higher than the 3- to 10-fold resistance induced by the CM, because the aFGF/bFGF concentrations in the local environment within solid tumors are likely to exceed their concentrations in the CM which was diluted 500- to 1 , 000-fold during collection.
EXAMPLE XIV: Protection of Gastrointestinal Cells from Cytotoxicity of
Anticancer Agents by FGF
This example shows that treatment of non-cancerous intestinal epithelium with aFGF and bFGF protects these cells from the toxicity of anticancer agents, such as paclitaxel and doxorubicin.
Briefly, normal rat intestinal epithelial cells (IEC6) were used. These cells were derived from normal, noncancerous intestinal epithelium. Cells were pretreated with 1 ng/ml aFGF and 3 ng/ml bFGF for 96 hours and then with doxorubicin or paclitaxel for 24 hours. Drug effect was measured as the reduction of bromodeoxyuridine incorporation in treated cells relative to untreated controls. The results show that the IC5o for doxorubicin was increased from 292 nM in the absence of aFGF and bFGF to 544 nM in the presence of aFGF and bFGF, and the IC50 for paclitaxel was increased from 491 nM to 1446 nM. This indicates that aFGF/bFGF treatment protects normal intestinal cells from the toxicity of anticancer agents.
EXAMPLE XV: Cell Culture Systems
Cell culture assay experiments can be performed in the rat prostate MAT-LyLu tumor cells, and the human prostate PC3 tumor cells or its metastatic subline PC3-LN cells. If the requirements of the invention are met in any of the three cell cells, the choice and the dosage of the FGF antagonist is suitable for use in the invention. Preferably the PC3 cells are used.
Human prostate PC3 tumor cells can be obtained from the American Type Culture Collection. The rat MAT-LyLu tumor cells and PC3-LN cells were obtained from Dr. John Isaacs (Johns Hopkins University) and Dr. Joy Ware (University of Virginia), respectively. The doubling time of PC3 and PC3-LN cells is approximately 24-hour, the doubling time ofMAT- MAT-LyLu cells is approximately 15-17 hr. All three cell lines should be cultured as monolayers in a humidified environment containing 5% CO2 and 95% air, at 37°C. PC 3 cells and PC3-LN cells should be maintained in RPMI 1640 medium supplemented with 9% heat-inactivated fetal bovine serum, 2 mM 1- glutamine, 100 μg/ml gentamicin, and 95 μg/ml cefotaxime. MAT-LyLu cells should be maintained in RPMI 1640 medium containing 100 μg/ml gentamicin, 95 μg/ml cefotaxime and 9% heat-inactivated fetal bovine serum. Cells are harvested from subconfluent cultures using trypsin and resuspended in fresh medium before plating. Cells with > 90% viability, as determined by trypan blue exclusion, are used to evaluate the cytotoxicity of an FGF antagonist, e.g., suramin. Cells are plated in 96 well microtiter plates at a density such that confluence would not be achieved at the end of the drug treatment period. Cells are allowed to attach to the plate surface by growing in drug-free medium for 20 to 24 hr. Afterward, cells are incubated with the FGF antagonist (one example used 0.2 ml of suramin)-containing culture medium, at concentrations spanning at least 4 log scales. The drug effect should be measured as inhibition of BrdU incoφoration, e.g., according to the Cell Proliferation ELISA BrdU (Boehringer Mannheim).
EXAMPLE XVI: Inhibition of FGF binding to FGF receptors This example describes the inhibition of the binding of FGF to FGF receptors by FGF antagonists.
The effects of several FGF antagonists (i.e., suramin, heparin, low molecular weight heparin, heparan sulfate) on the binding of 125I-bFGF on FGF receptors in human prostate cancer PC3 cells and human breast cancer MCF7 cells were studied. Briefly, cells were plated at 1 X 105 cells per 16 mm well on 24-well plates and were allowed to settle for 48 hours before binding experiment. On the day of the experiment, cells were incubated for 2 hours at 37°C in serum-free DMEM supplemented with 0.15%) gelatin. Cells were then washed twice with ice-cold phosphate buffered saline. Aliquots of bFGF antagonist in cold DMEM binding buffer (100 μl, containing 25 mM HEPES, 0.15% gelatin, pH 7.5) at various concentration and 125I-bFGF (100 μl) at various concentrations in the same buffer were added in each well. At the end of the incubation, the cells were either washed three times with 1 ml ice-cold phosphate-buffered saline or washed twice with ice-cold phosphate-buffered saline followed by one wash of 1 ml 2 M NaCl in 25 mM HEPES, pH 7.5. The cells were then extracted with 0.25 ml 0.5% Triton X-100 and the radioactivity in the extract was determined in a gamma scintillation counter.
The results, summarized in Table 8, show that the FGF antagonists inhibited the high affinity and total binding of 125I-bFGF in PC3 and MCF7 cells.
Table 8. Inhibition of binding of 125I-bFGF to FGF receptors by FGF antagonists.
FGF antagonist IC50 for high affinity binding IC50 for total binding PC3 MCF7 PC3 MCF7
Suramin 87 μg/ml 21 μg/ml 87 μg/ml 17 μg/ml
Heparin O.001 μg/ml — 0.001 μg/ml —
Low mol wt heparin — 10 μg/ml — 0.5 μg/ml
Heparan sulfate ~ 1 μg/ml — 5 μg/ml EXAMPLE XVII: Genetic changes induced by extracellular bFGF
Differential display was used to identify the down-stream genes that are involved in the bFGF-induced multidrug resistance. These genes were identified by comparing the effect of bFGF on the expression of these genes in a cell line that demonstrates bFGF-induced chemoresistance (i.e. human prostate cancer PC3 cells) with that in a cell line which does not show bFGF-induced chemoresistance (i.e. human head and neck cancer FaDu cells). By this method, the genes that were altered by bFGF treatment in PC3 cells but not in FaDu cells were identified. The identified genes are likely to be associated with the bFGF-induced multidrug resistance.
For differential display, total RNA was isolated from the cells and reverse- transcripted in single strand cDNA. PCR was performed using a pair of T and P primers. One pair of primers displayed about 50 - 100 bands. 90 T and P primer combinations which displayed about 6000 cDNA bands were used. Fourteen bands showed either increase or decrease in density in PC3 cells after bFGF treatment. Five of these bands were reproducibly altered in PC3 cells, but not in FaDu cells. These bands were cut from the gels, reamplified and cloned into pGEM-T vector. Nine different clones were identified and sequenced. The gene sequence was compared to the data in the NCBI GenBank. The comparison showed that 5 clones corresponded to previously reported genes, and 4 clones were not-yet-reported and therefore potentially new genes. Primers were synthesized for drug resistant cancer cells. Among the nine clones, five were altered reproducibly by bFGF treatment while bFGF had no effect on the remaining four clones. The expression of two of the known genes, TSC22 and VEGF, were increased by 20 to 40%, whereas the expression of the three new genes (temporarily named FSC-1 , FSC-2, and FSC-3) were induced by 65%, 300%, and 40%. TSC22 and VEGF are not known to be regulated by bFGF.
Expression of the transcription factor TFII-I was also identified to be upregulated by extracellular bFGF. Using differential display, cloning and sequencing techniques, another cDNA (256 bp) was identified which showed only partial (i.e. - 60%) matches with several segments in the human PAC and BAC clones, indicating this as a not-yet reported gene. Equivalents
Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents of the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

Claims

We claim:
1. A method of improving the efficacy of a cytotoxic agent, in a subj ect, comprising administering to the subject at least one cytotoxic agent; administering to the subject at least one FGF antagonist, wherein the FGF antagonist enhances the efficacy of the cytotoxic agent, relative to the effect of the cytotoxic agent in the absence of the FGF antagonist.
2. A method of inhibiting growth or proliferation of hypeφroliferative cells in a subject, comprising administering to the subject at least one cytotoxic agent and at least one FGF antagonist, in an amount which, together, is effective to inhibit growth or proliferation of the hypeφroliferative cells, or is effective to induce death of the hypeφroliferative cells.
3. A method of inhibiting growth or proliferation of, or inducing killing of tumor cells in a subject, comprising administering to the subject at least one cytotoxic agent and at least one FGF antagonist, in an amount which, together, is effective to inhibit growth or proliferation of the tumor cells.
4. A method of inhibiting unwanted growth or proliferation of, or inducing killing of tumor cells in a human subject, comprising administering to the human subject at least one cytotoxic agent and at least one FGF antagonist, in an amount, which together, is effective to reduce or inhibit the growth or proliferation of the established tumor, induce cell death of the established tumors, or to reduce the size of the established tumors.
5. A method of inhibiting growth or proliferation of tumor cells or inducing killing of tumor cells in a subject, comprising administering to the subject at least one cytotoxic agent and at least one FGF antagonist, in an amount which, together, is effective to inhibit growth or proliferation of the tumor cells, or induce cell death of the tumors, wherein said FGF antagonist is other than suramin.
6. A method of inhibiting unwanted proliferation of, or inducing killing of, an established tumor in a subject, comprising administering to the subject at least one cytotoxic agent and at least one FGF antagonist, in an amount, which together, is effective to reduce or inhibit the growth or proliferation of the established tumor, induce cell death of the established tumors, or to reduce the size of the established tumors, wherein the established tumor is selected from the group consisting of lung carcinoma, renal cell carcinoma, glioma, melanoma, chemotherapy resistant tumors and metastatic tumors.
7. A method of inhibiting unwanted proliferation of, or inducing killing of, an established tumor in a subject, comprising administering to the subject at least one cytotoxic agent and at least one FGF antagonist, in an amount, which together, is effective to reduce or inhibit the growth or proliferation of the established lung tumor, induce cell death of the lung established tumors, or to reduce the size of the established lung tumors,
8. The method of any of claims 1, 2, 3, 4, or 5, wherein the FGF antagonist comprises a bFGF inhibitor or an aFGF inhibitor.
9. The method of any of claims 1, 2, 3, 4, or 5, wherein the FGF antagonist comprises a bFGF inhibitor and an aFGF inhibitor.
10. The method of any of claims 1, 2, 3, 4, or 5, wherein the FGF antagonist is capable of binding to an FGF molecule or an FGF receptor.
11. The method of any of claims 1, 2, 3, 4, or 5, wherein the FGF antagonist is a protein or a peptide.
12. The method of any of claims 1, 2, 3, 4, or 5 wherein the FGF antagonist is a monoclonal antibody or an antigen-binding fragment thereof.
13. The method of claim 12, wherein the monoclonal antibody is an anti-FGF antibody.
14. The method of claim 10, wherein the antagonist is a truncated FGF molecule, or a fragment thereof.
15. The method of either of claims 4 or 6, wherein the FGF antagonist is suramin.
16. The method of claim 15, wherein the total suramin exposure is less than 52 micromolar-day.
17. The method of claim 15, wherein the total suramin exposure is less than 252 micromolar-day.
18. The method of claim 15, wherein suramin is administered in an amount that results in a concentration ranging from about 0.1 to 100 μg/ml.
19. The method of claim 15, wherein suramin is administered in an amount that results in a concentration ranging from about 15 to 45 μg/ml.
20. The method of claim 15, wherein suramin is administered over a time period which ends less than 180 days after the last day on which the cytotoxic agent is administered.
21. The method of claim 15, wherein suramin is administered over a time period which ends less than 30 days after the last day on which the cytotoxic agent is administered.
22. The method of claim 15, wherein suramin is administered over a time period which begins less than 180 days before the first day on which the cytotoxic agent is administered.
23. The method of claim 15, wherein suramin is administered over a time period which begins less than 30 days before the first day on which the cytotoxic agent is administered.
24. The method of either of claims 4 or 6, wherein the FGF antagonist is administered at levels such that the plasma concentration of the FGF antagonist does not result in significant cell cycle arrest, significant cell death, or significant inhibition of cell growth.
25. The method of either of claims 4 or 6, wherein the FGF antagonist is administered at levels such that the plasma concentration of the FGF antagonist sensitizes the established tumor to treatment with cytotoxic agents.
26. The method of any of claims 1 , 2, 3, 4, or 5, wherein the FGF antagonist is heparin.
27. The method of any of claims 1, 2, 3, 4, or 5, wherein the FGF antagonist is low molecular weight heparin.
28. The method of any of claims 1 , 2, 3, 4, or 5, wherein the FGF antagonist is heparan sulfate.
29. The method of any of claims 3, 4, or 5, wherein the tumor is selected from the group consisting of a fibrosarcoma, myosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyo sarcoma, gastric cancer, esophageal cancer, colon carcinoma, rectal cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, uterine cancer, cancer of the head and neck, skin cancer, brain cancer, squamous cell carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinoma, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, testicular cancer, lung carcinoma, small cell lung carcinoma, non-small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, meningioma, melanoma, neuroblastoma, retinoblastoma, leukemia, lymphoma, or Kaposi sarcoma.
30. The method of claim 4, wherein the established tumor comprises a cancer forming from a tissue or cell selected from the group consisting of a tissue or cell where an FGF molecule is expressed, a tissue or cell that is in contact or exposed to aFGF, bFGF or both, and an FGF-producing tissue or cells.
31. The method of claim 2, wherein the hypeφroliferive disorder comprises unwanted growth forming from a tissue or cell selected from the group consisting of a tissue or cell where an FGF molecule is expressed, a tissue or cell that is in contact or exposed to aFGF, bFGF or both, and an FGF-producing tissue or cells.
32. The method of any of claims 1, 2, 3, 4, or 5, wherein the subject is a mammal.
33. The method of claim 32, wherein the subject is a human.
34. The method of any of claims 1, 2, 3, 4, or 5, wherein the cytotoxic agent is selected from the group consisting of an antimicrotubule agent, a topoisomerase I inhibitor, a topoisomerase II inhibitor, an antimetabolite, a mitotic inhibitor, an alkylating agent, an intercalating agent, an agent capable of interfering with a signal transduction pathway, an agent that promotes apoptosis and/or necrosis, an interferon, an interleukin, a tumor necrosis factor, and radiation.
35. The method of claim 34, wherein the cytotoxic agent is paclitaxel, interferon alpha, gemcitabine, fludarabine, irinotecan, carboplatin, cisplatin, taxotere, doxorubicin, epirubicin, 5-fluorouracil, UFT, tamoxifen, goserelin, ketoconazole, an anti-Her2/neu receptor, anti-CD20, leuprolide (Lupron) and flutamide.
36. The method of any of claims 1, 2, 3, 4, or 5, wherein the cytotoxic agent and the FGF antagonist are administered simultaneously.
37. The method of any of claims 1, 2, 3, 4, or 5, wherein repeated dosages of the same, or a different cytotoxic agent are administered.
38. The method of any of claims 1, 2, 3, 4, or 5, wherein repeated dosages of the same, or a different FGF antagonist are administered.
39. The method of any of claims 1, 2, 3, 4, or 5, wherein the cytotoxic agent is administered parenterally, orally, locoregionally, nasally, rectally, topically, or transdermally.
40. The method of any of claims 1, 2, 3, 4, or 5, wherein the FGF antagonist is administered parenterally, orally, locoregionally, nasally, rectally, topically, or transdermally.
41. The method of any of claims 1 , 2, 3, 4, or 5, further comprising monitoring the FGF levels of the subject prior to administering the FGF antagonist to the subject.
42. The method of claim 41 , wherein the amount of the FGF antagonist administered to the subject is determined, at least in part, based upon the levels of FGF present in the subject as determined by said monitoring step prior to administering the FGF antagonist.
43. The method of claim 1, wherein, the FGF antagonist improves the efficacy of the cytotoxic agent against an established tumor.
44. A method for inhibiting chemoresistance of a tumor cell to a cytotoxic agent, comprising administering to the tumor cell an FGF antagonist such that chemoresistance of the tumor cell to the cytotoxic agent is inhibited.
45. The method of claim 44, wherein the tumor cell is in a subject.
46. The method of claim 45, wherein the tumor cell is in an established tumor in a subject.
47. The method of claim 44, wherein the FGF antagonist is a specific FGF inhibitor.
48. A method for treating or preventing a disorder involving aberrant growth or proliferation of benign proliferative tissue, comprising contacting the tissue with at least one cytotoxic agent and at least one specific inhibitor of FGF, in an amount which, together, is effective to inhibit growth or proliferation of the tissue or to induce death of hypeφroliferative cells.
49. The method of claim 48, wherein the disorder is selected from the group consisting of psoriasis, cysts, benign prostatic hypeφlasia, and endometriosis.
50. The method of claim 48, wherein the disorder is selected from the group consisting of oral papillomas, central giant cell granulomas of the mouth or pharynx, benign cementoblastomas of the oral cavity, oral plakia, gastric polyps, gastric adenomas, small intestinal adenomas, small intestinal granulomas, small intestinal papillomas, small intestinal oncocytomas, small intestinal Schwannomas, colonic polyps, colonic adenomas, Crohn's disease, hepatic adenoma, hepatic cirrhosis, biliary papillomatosis, pancreatic adenomas, pancreatic ductal hypeφlasia, renal oncocytomas, renal papillomas, adenomas of the bladder, malakoplakia of the bladder, pseudosarcomas of the bladder, endometriosis, benign prostatic hypeφlasia, erythroplasia of the penis, polyps and papillomas of the vulva, vagina, or cervix, endometrial polyps, adenomas, papillomas, or leimyomas, ovarian cysts, fibrocystic disease of the breast, lipoma of the breast, sclerosing adenosis, hemangioma, ductal hypeφlasia of the breast, fibroadenomas, adenomyoepitheliomas, hamartoma, nevus of the skin, genodermatoses, fibrosis of the bone, fibrous dysplasia, chondrodysplasisa, sclerosing bone dysplasia, axial osteomalacia, fibrogenesis imperfecta, osteomas, osteoid osteomas, osteoblastomas, osteochondomas, enchondromas, chondromyxoid fibromas, chondroblastomas, synovial lipomas, adenomas of endocrine organs, goiter, Graves' disease, adrenal hypeφlasia, adrenal adenomas, adrenal MEN I syndrome, and adrenal myelolipomas.
51. A method of inhibiting cell killing, or protecting the ability of a cell to proliferate, from damage caused by a cytotoxic agent, comprising administering to the subject an effective amount of at least one FGF agonist, thereby protecting or reducing the damage to the cell from said cytotoxic agent, wherein the cell is selected from the group consisting of a cell from a body surface or cavity and a hematopoietic cell, and wherein the cytotoxic agent is an agent other than radiation.
52. The method of claim 51 , wherein the body surface is the lining of the gastrointestinal tract.
53. The method of claim 51, wherein bFGF, aFGF, or a combination of both, are administered.
54. The method of claim 51 , wherein the at least one FGF agonist is administered orally or ex vivo.
55. A method of inhibiting cell killing, or protecting the ability of a hair follicle cell to proliferate, from damage caused by a cytotoxic agent, comprising administering to the subject an effective amount of at least one FGF agonist, thereby protecting or reducing the damage to the hair follicle cell from said cytotoxic agent, wherein administration of the FGF agonist does not result in significant systemic administration of the FGF agonist.
56. The method of claim 55, wherein the at least one FGF agonist is administered topically.
57. A pharmaceutical composition comprising at least one FGF antagonist, at least one cytotoxic agent, and a pharmaceutically acceptable carrier, wherein said FGF antagonist is present in an amount effective to enhance the efficacy of the cytotoxic agent, in reducing or inhibiting the proliferation, or in enhancing the killing, of a hypeφroliferative cell.
58. The method of claim 57, wherein the pharmaceutical composition includes an inhibitor of bFGF; an inhibitor of aFGF; or a bFGF inhibitor and an aFGF inhibitor.
59. A pharmaceutical composition comprising at least one FGF agonist, and a pharmaceutically acceptable carrier, wherein said FGF agonist is present in an amount effective to protect, or reduce the damage, of a rapidly dividing cell, from said cytotoxic agent.
60. The pharmaceutical composition of claim 59, wherein the FGF agonist comprises bFGF; aFGF; or bFGF and aFGF, or a fragment or an analog thereof.
61. A vaccine comprising an FGF polypeptide and a pharmaceutically acceptable carrier, in an amount effective to immunize a subject against a neoplastic disease.
62. A vaccine comprising a FGF receptor-derived polypeptide and a pharmaceutically acceptable carrier, in an amount effective to immunize a subject against a neoplastic disease.
63. A vaccine comprising a fragment of a proteoglycan that facilitates the binding of FGF to its receptors and a pharmaceutically acceptable carrier, in an amount effective to immunize a subject against a neoplastic disease.
64. A kit for carrying out the combined administration of the FGF antagonist with other cytotoxic agents, comprising an FGF antagonist formulated in a pharmaceutical carrier, and at least one cytotoxic agent, formulated, as appropriate, in one or more separate pharmaceutical preparations.
65. A kit for carrying out the combined administration of the FGF agonist with other cytotoxic agents, comprising an FGF agonist formulated in a pharmaceutical carrier, and at least one cytotoxic agent, formulated, as appropriate, in one or more separate pharmaceutical preparations.
66. A method of diagnosing a proliferative disorder in a subject, the method comprising providing a sample from the subject, and evaluating the expression of one or more FGF genes or gene products in the sample; wherein an increase or a decrease in the level of said one or more FGF genes or gene products indicates the presence or absence of the disorder.
67. The method of claim 66, wherein said one or more of the FGF genes or gene products is selected from the group consisting of bFGF, aFGF, TSC22, VEGF, GAFA1, GAFA2, GAFA3, and TFII-I.
68. The method of claim 66, wherein the disorder is a malignant disorder.
69. A method for evaluating the effectiveness of a treatment for treating a proliferative disorder, comprising providing a test organism, administering said treatment to said test organism and evaluating the expression of one or more FGF genes; wherein a change in the level of said one or more FGF genes or gene products in a sample given said treatment, relative to the level of expression without the treatment, indicative of the effectiveness of the compound for treating said disorder.
70. The method of claim 69, wherein said one or more of the FGF genes or gene product is selected from the group consisting of bFGF, aFGF, TSC22, VEGF, GAFA1, GAFA2, GAFA3, and TFII-I.
71. A method for evaluating the efficacy of a treatment of a proliferative disorder, in a patient, comprising providing a sample from said patient, and evaluating the expression of one or more FGF genes; wherein a decrease or increase in the level of said one or more FGF genes or gene products in a sample obtained after treatment, relative to the level of expression before treatment, is indicative of the efficacy of the treatment of said disorder.
72. The method of claim 71, wherein said one or more of the FGF genes is selected from the group consisting of bFGF, aFGF, TSC22, VEGF, GAFA1, GAFA2, GAFA3, and TFII-I.
73. A method of analyzing a sample for the level of resistance to cytotoxic agents in a tumor, comprising evaluating the level of one or more FGF genes or gene products, wherein an increase or a decrease in the level of one or more FGF genes or gene products, relative to a control, indicates the presence of resistance to cytotoxic agents.
74. The method of claim 73, wherein the sample is a sample of tissue having unwanted proliferation.
75. The method of claim 73, wherein the FGF genes or gene products is selected from the group consisting of bFGF, aFGF, TSC22, VEGF, GAFAl, GAFA2, GAFA3, and TFII-I.
76. A method of staging a proliferative disorder, in a subject, comprising providing a sample from said subject and evaluating the expression of one or more FGF genes; wherein an increase or a decrease in the level of said one or more FGF genes or gene products, relative to a control, indicates a stage in the disorder.
77. The method of claim 73, wherein the one or more FGF genes is selected from the group consisting of bFGF, aFGF, TSC22, VEGF, GAFAl , GAFA2, GAFA3, and
TFII-I.
PCT/US2000/040103 1999-06-03 2000-06-05 Methods and compositions for modulating cell proliferation and cell death WO2000074634A2 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
AU57903/00A AU780454B2 (en) 1999-06-03 2000-06-05 Methods and compositions for modulating cell proliferation and cell death
EP00943429A EP1206234A4 (en) 1999-06-03 2000-06-05 Methods and compositions for modulating cell proliferation and cell death
CA002377385A CA2377385A1 (en) 1999-06-03 2000-06-05 Methods and compositions for modulating cell proliferation and cell death
IL14687200A IL146872A0 (en) 1999-06-03 2000-06-05 Methods and compositions for modulating cell proliferation and cell death
JP2001501171A JP2003503313A (en) 1999-06-03 2000-06-05 Methods and compositions for modulating cell proliferation and cell death
IL146872A IL146872A (en) 1999-06-03 2001-12-02 Use of cytotoxic agent and suramin for the manufacture of pharmaceutical compositions

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US13734599P 1999-06-03 1999-06-03
US60/137,345 1999-06-03
US16598399P 1999-11-17 1999-11-17
US60/165,983 1999-11-17
US17203199P 1999-12-23 1999-12-23
US60/172,031 1999-12-23
US18744500P 2000-03-07 2000-03-07
US60/187,445 2000-03-07

Publications (3)

Publication Number Publication Date
WO2000074634A2 true WO2000074634A2 (en) 2000-12-14
WO2000074634A3 WO2000074634A3 (en) 2001-08-23
WO2000074634A9 WO2000074634A9 (en) 2002-09-26

Family

ID=27495263

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2000/040103 WO2000074634A2 (en) 1999-06-03 2000-06-05 Methods and compositions for modulating cell proliferation and cell death

Country Status (8)

Country Link
US (2) US6599912B1 (en)
EP (1) EP1206234A4 (en)
JP (1) JP2003503313A (en)
KR (1) KR100903243B1 (en)
AU (1) AU780454B2 (en)
CA (1) CA2377385A1 (en)
IL (2) IL146872A0 (en)
WO (1) WO2000074634A2 (en)

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1328264A1 (en) * 2000-07-25 2003-07-23 PHARMACIA &amp; UPJOHN COMPANY Use of estramustine phosphate in the treatment of bone metastasis
JP2005508920A (en) * 2001-10-03 2005-04-07 セレーター テクノロジーズ インコーポレイテッド Composition for delivering a concomitant drug
US7071164B2 (en) 2001-08-16 2006-07-04 Kimberly-Clark Worldwide, Inc. Anti-cancer and wound healing compounds
US7094754B2 (en) 2001-08-16 2006-08-22 Kimberly-Clark Worldwide, Inc. Anti-aging and wound healing compounds
US7148194B2 (en) 2002-12-30 2006-12-12 Kimberly-Clark Worldwide, Inc. Method to increase fibronectin
WO2006009805A3 (en) * 2004-06-18 2007-01-04 Genentech Inc Combination of a chemotherapeutic agent and an antagonist of a gene product for treating tumors
EP1742961A2 (en) * 2004-02-13 2007-01-17 Boston Biomedical Research Institute Inhibition of fgf signaling
JP2007504194A (en) * 2003-09-05 2007-03-01 ゲンチウム エスピーエー Anti-tumor preparation containing defibrotide alone or in combination with other anti-tumor agents
US7186693B2 (en) 2001-08-16 2007-03-06 Kimberly - Clark Worldwide, Inc. Metalloproteinase inhibitors for wound healing
US7189700B2 (en) 2003-06-20 2007-03-13 Kimberly-Clark Worldwide, Inc. Anti-chrondrosarcoma compounds
US7196162B2 (en) 2001-08-16 2007-03-27 Kimberly-Clark Worldwide, Inc. Anti-aging and wound healing compounds
US7250416B2 (en) 2005-03-11 2007-07-31 Supergen, Inc. Azacytosine analogs and derivatives
US7276228B2 (en) 2001-04-24 2007-10-02 Supergen, Inc. Methods for treating hematological disorders through inhibition of DNA methylation and histone deacetylase
US7700567B2 (en) 2005-09-29 2010-04-20 Supergen, Inc. Oligonucleotide analogues incorporating 5-aza-cytosine therein
WO2010140795A2 (en) * 2009-06-01 2010-12-09 (주)차바이오앤디오스텍 Human-placenta-like composition containing a human-placenta-derived growth factor and cytokines, and cosmetic use thereof
US7850990B2 (en) 2001-10-03 2010-12-14 Celator Pharmaceuticals, Inc. Compositions for delivery of drug combinations
US8980862B2 (en) 2010-11-12 2015-03-17 Gentium S.P.A. Defibrotide for use in prophylaxis and/or treatment of Graft versus Host Disease (GVHD)
US9381207B2 (en) 2011-08-30 2016-07-05 Astex Pharmaceuticals, Inc. Drug formulations
US9902952B2 (en) 2012-06-22 2018-02-27 Gentrum S.R.L. Euglobulin-based method for determining the biological activity of defibrotide
US10058507B2 (en) 2001-10-03 2018-08-28 Celator Pharmaceuticals, Inc. Compositions for delivery of drug combinations
US10393731B2 (en) 2014-11-27 2019-08-27 Gentium S.R.L. Cellular-based method for determining the biological activity of defibrotide
US10485764B2 (en) 2015-07-02 2019-11-26 Otsuka Pharmaceutical Co., Ltd. Lyophilized pharmaceutical compositions
US10519190B2 (en) 2017-08-03 2019-12-31 Otsuka Pharmaceutical Co., Ltd. Drug compound and purification methods thereof
RU2723091C2 (en) * 2015-03-23 2020-06-08 Дойчес Кребсфоршунгсцентрум Штифтунг Дес Эффентлихен Рехтс Oligonucleotide sequences aimed at the transcription factor tsc22d4, for treating insulin resistance

Families Citing this family (75)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030180290A1 (en) * 1995-06-07 2003-09-25 Idec Pharmaceuticals Corporation Anti-CD80 antibody having ADCC activity for ADCC mediated killing of B cell lymphoma cells alone or in combination with other therapies
JP2003531149A (en) * 2000-04-13 2003-10-21 ザ・ロツクフエラー・ユニバーシテイ Enhancement of antibody-derived immune response
CA2436180C (en) * 2001-01-31 2011-11-08 Idec Pharmaceutical Corporation Immunoregulatory antibodies and uses thereof
US20030103971A1 (en) * 2001-11-09 2003-06-05 Kandasamy Hariharan Immunoregulatory antibodies and uses thereof
US20030059375A1 (en) * 2001-08-20 2003-03-27 Transave, Inc. Method for treating lung cancers
CA2457148A1 (en) * 2001-08-20 2003-02-27 Transave, Inc. Treatment of cancers by inhalation of stable platinum-containing formulations
WO2003026574A2 (en) * 2001-09-24 2003-04-03 Au Jessie L-S Methods and compositions to determine the chemosensitizing dose of suramin used in combination therapy
JP4694128B2 (en) * 2002-01-30 2011-06-08 ザ ブライハム アンド ウイメンズ ホスピタル, インコーポレイテッド Compositions and methods related to TIM-3, Th1-specific cell surface molecules
EP1490027A4 (en) * 2002-03-05 2010-11-10 Transave Inc Methods for entrapment of bioactive agent in a liposome or lipid complex
US9186322B2 (en) * 2002-08-02 2015-11-17 Insmed Incorporated Platinum aggregates and process for producing the same
US20040101553A1 (en) * 2002-08-02 2004-05-27 Transave, Inc. Platinum aggregates and process for producing the same
US7718189B2 (en) * 2002-10-29 2010-05-18 Transave, Inc. Sustained release of antiinfectives
US7879351B2 (en) * 2002-10-29 2011-02-01 Transave, Inc. High delivery rates for lipid based drug formulations, and methods of treatment thereof
CN1747738B (en) * 2002-10-29 2010-11-24 川塞夫有限公司 Sustained release of antifectives
KR20050114241A (en) * 2003-03-19 2005-12-05 가부시키가이샤 디나벡크 겐큐쇼 Method of treating inflammtory disease associated with bone destruction
KR20050116166A (en) * 2003-04-28 2005-12-09 파마시아 앤드 업존 캄파니 엘엘씨 Use of irinotecan for treatment of resistant breast cancer
US20050282893A1 (en) * 2004-01-30 2005-12-22 Au Jessie L Methods and compositions for using suramin, pentosan, polysulfate, telomerase antisense and telomerase inhibitors
US20050249822A1 (en) * 2004-03-18 2005-11-10 Transave, Inc. Administration of cisplatin by inhalation
WO2005112957A1 (en) * 2004-05-21 2005-12-01 Transave, Inc. Treatment of lung diseases and pre-lung disease conditions
EP2290071B1 (en) * 2004-05-28 2014-12-31 Asuragen, Inc. Methods and compositions involving microRNA
RO121676B1 (en) * 2004-08-18 2008-02-28 Institutul Oncologic "Prof.Dr. Al. Trestioreanu" Method for improving cytostatic tolerance of the body
US20060246124A1 (en) * 2004-11-08 2006-11-02 Pilkiewicz Frank G Methods of treating cancer with lipid-based platinum compound formulations administered intraperitoneally
EP2281887A1 (en) 2004-11-12 2011-02-09 Asuragen, Inc. Methods and compositions involving miRNA and miRNA inhibitor molecules
US20060183712A1 (en) * 2005-02-17 2006-08-17 The Texas A&M University System Affinity purified heparin/heparan sulfate for controlling the biological activity of the FGF receptor
US8999706B2 (en) * 2005-04-12 2015-04-07 The Trustees Of The University Of Pennsylvania Methods for preparation of human hair-follicle derived multipotent adult stem cells
US20060283742A1 (en) * 2005-06-16 2006-12-21 Canel Lightning Co. Ltd. Multi-lamp display packaging for lamps with collapsible lampshades
CA2619568A1 (en) * 2005-08-19 2007-03-01 Government Of The United States Of America, Represented By The Secretary , Department Of Health And Human Services Topical formulations of histone deacetylase inhibitors and methods of using the same
US9107824B2 (en) 2005-11-08 2015-08-18 Insmed Incorporated Methods of treating cancer with high potency lipid-based platinum compound formulations administered intraperitoneally
US20070190181A1 (en) * 2005-11-08 2007-08-16 Pilkiewicz Frank G Methods of treating cancer with lipid-based platinum compound forumulations administered intravenously
US20070190180A1 (en) * 2005-11-08 2007-08-16 Pilkiewicz Frank G Methods of treating cancer with high potency lipid-based platinum compound formulations administered intravenously
US20070190182A1 (en) * 2005-11-08 2007-08-16 Pilkiewicz Frank G Methods of treating cancer with high potency lipid-based platinum compound formulations administered intraperitoneally
WO2007064658A2 (en) * 2005-11-30 2007-06-07 Transave, Inc. Safe and effective methods of administering therapeutic agents
WO2007065016A2 (en) * 2005-12-02 2007-06-07 Au Jessie L S Methods and compositions to improve activity and reduce toxicity of stents
DK1962805T3 (en) 2005-12-08 2016-09-26 Insmed Inc Lipid-based compositions of the anti-infective agents for the treatment of lung infections
US20070197599A1 (en) * 2006-02-02 2007-08-23 Matier William L Hydroxylamines and derivatives as anti-angiogenic agents
JP2010510964A (en) * 2006-09-19 2010-04-08 アシュラジェン インコーポレイテッド MiR-15, miR-26, miR-31, miR-145, miR-147, miR-188, miR-215, miR-216, miR-331, mmu-miR-292 as targets for therapeutic intervention Genes and pathways regulated by 3p
AU2007299828C1 (en) * 2006-09-19 2014-07-17 Interpace Diagnostics, Llc MicroRNAs differentially expressed in pancreatic diseases and uses thereof
US20080085881A1 (en) * 2006-10-06 2008-04-10 Aimery De Gramont Stop-and-go oxaliplatin treatment regimens
EP1918376A1 (en) * 2006-11-03 2008-05-07 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. FGFR4 promotes cancer cell resistance in response to chemotherapeutic drugs
AU2007333109A1 (en) * 2006-12-08 2008-06-19 Asuragen, Inc. Functions and targets of let-7 micro RNAs
CA2671294A1 (en) * 2006-12-08 2008-06-19 Asuragen, Inc. Mir-21 regulated genes and pathways as targets for therapeutic intervention
CN101622349A (en) * 2006-12-08 2010-01-06 奥斯瑞根公司 miR-20 regulated genes and pathways as targets for therapeutic intervention
US20090175827A1 (en) * 2006-12-29 2009-07-09 Byrom Mike W miR-16 REGULATED GENES AND PATHWAYS AS TARGETS FOR THERAPEUTIC INTERVENTION
WO2008137717A1 (en) * 2007-05-04 2008-11-13 Transave, Inc. Compositions of multicationic drugs for reducing interactions with polyanionic biomolecules and methods and uses thereof
US9333214B2 (en) 2007-05-07 2016-05-10 Insmed Incorporated Method for treating pulmonary disorders with liposomal amikacin formulations
US9119783B2 (en) 2007-05-07 2015-09-01 Insmed Incorporated Method of treating pulmonary disorders with liposomal amikacin formulations
US9114081B2 (en) 2007-05-07 2015-08-25 Insmed Incorporated Methods of treating pulmonary disorders with liposomal amikacin formulations
US20090131354A1 (en) * 2007-05-22 2009-05-21 Bader Andreas G miR-126 REGULATED GENES AND PATHWAYS AS TARGETS FOR THERAPEUTIC INTERVENTION
US20090232893A1 (en) * 2007-05-22 2009-09-17 Bader Andreas G miR-143 REGULATED GENES AND PATHWAYS AS TARGETS FOR THERAPEUTIC INTERVENTION
WO2008154333A2 (en) * 2007-06-08 2008-12-18 Asuragen, Inc. Mir-34 regulated genes and pathways as targets for therapeutic intervention
WO2008156644A2 (en) * 2007-06-14 2008-12-24 Frank David A Stat modulators
US8361714B2 (en) 2007-09-14 2013-01-29 Asuragen, Inc. Micrornas differentially expressed in cervical cancer and uses thereof
WO2009052386A1 (en) * 2007-10-18 2009-04-23 Asuragen, Inc. Micrornas differentially expressed in lung diseases and uses thereof
WO2009070805A2 (en) * 2007-12-01 2009-06-04 Asuragen, Inc. Mir-124 regulated genes and pathways as targets for therapeutic intervention
WO2009086156A2 (en) * 2007-12-21 2009-07-09 Asuragen, Inc. Mir-10 regulated genes and pathways as targets for therapeutic intervention
EP2260110B1 (en) * 2008-02-08 2014-11-12 Asuragen, INC. miRNAs DIFFERENTIALLY EXPRESSED IN LYMPH NODES FROM CANCER PATIENTS
WO2009111643A2 (en) * 2008-03-06 2009-09-11 Asuragen, Inc. Microrna markers for recurrence of colorectal cancer
WO2009154835A2 (en) * 2008-03-26 2009-12-23 Asuragen, Inc. Compositions and methods related to mir-16 and therapy of prostate cancer
WO2009126172A1 (en) * 2008-04-11 2009-10-15 The Trustees Of Columbia University In The City Of New York Resistance to polyphenon e due to increased bcl-2 expression
US8258111B2 (en) 2008-05-08 2012-09-04 The Johns Hopkins University Compositions and methods related to miRNA modulation of neovascularization or angiogenesis
WO2009155504A2 (en) * 2008-06-20 2009-12-23 The Children's Medical Center Corporation Methods for the modulation of angiogenesis
WO2010056737A2 (en) * 2008-11-11 2010-05-20 Mirna Therapeutics, Inc. Methods and compositions involving mirnas in cancer stem cells
US8491927B2 (en) 2009-12-02 2013-07-23 Nimble Epitech, Llc Pharmaceutical composition containing a hypomethylating agent and a histone deacetylase inhibitor
US8445517B2 (en) 2009-12-11 2013-05-21 Dana-Farber Cancer Institute Stat modulators
WO2012116328A2 (en) 2011-02-24 2012-08-30 H. Lee Moffitt Cancer Center And Research Institute, Inc. Bad phosphorylation determines ovarian cancer chemo-sensitivity and patient survival
US9644241B2 (en) 2011-09-13 2017-05-09 Interpace Diagnostics, Llc Methods and compositions involving miR-135B for distinguishing pancreatic cancer from benign pancreatic disease
EP2776043B1 (en) * 2011-11-11 2018-02-21 Millennium Pharmaceuticals, Inc. Biomarkers of response to proteasome inhibitors
LT2852391T (en) 2012-05-21 2022-03-10 Insmed Incorporated Systems for treating pulmonary infections
MX356830B (en) 2012-07-13 2018-06-15 Shin Nippon Biomedical Laboratories Ltd Chiral nucleic acid adjuvant.
US11291644B2 (en) 2012-09-04 2022-04-05 Eleison Pharmaceuticals, Llc Preventing pulmonary recurrence of cancer with lipid-complexed cisplatin
AU2013352259B2 (en) 2012-11-29 2018-06-14 Insmed Incorporated Stabilized vancomycin formulations
WO2014124412A2 (en) * 2013-02-11 2014-08-14 President And Fellows Of Harvard College Methods and compounds for the inhibition of cellular proliferation
ES2926985T3 (en) 2014-05-15 2022-10-31 Insmed Inc Methods for treating nontuberculous mycobacterial lung infections
US11129906B1 (en) 2016-12-07 2021-09-28 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria
EP3773505A4 (en) 2018-03-30 2021-12-22 Insmed Incorporated Methods for continuous manufacture of liposomal drug products

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5574026A (en) * 1994-12-13 1996-11-12 American Cyanamid Company Methods for inhibiting angiogenesis proliferation of endothelial or tumor cells and tumor growth
US5716981A (en) * 1993-07-19 1998-02-10 Angiogenesis Technologies, Inc. Anti-angiogenic compositions and methods of use

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5252718A (en) * 1986-04-22 1993-10-12 The Salk Institute For Biological Studies Fibroblast growth factor antagonists
US5102870A (en) 1989-04-14 1992-04-07 Schering Ag Treatment and prevention of oral mucositis with growth factors
JPH04224522A (en) 1990-04-27 1992-08-13 Merck & Co Inc Therapeutic or prophylactic method for alopecia using composition containing fibroblast growth factor
US5486509A (en) 1991-06-28 1996-01-23 University Of Miami Method of preventing and treating chemotherapy-induced alopecia
US5587368A (en) * 1993-11-30 1996-12-24 New York University Administration of a 27-hydroxycholesterol or related compound or sterol-27-hydroxylase stimulant to prevent restenosis following vascular endothelial injury
US5783568A (en) 1994-06-10 1998-07-21 Sugen, Inc. Methods for treating cancer and other cell proliferative diseases
US5597830A (en) 1994-12-20 1997-01-28 Warner-Lambert Company Combination chemotherapy
US5773252A (en) 1995-06-05 1998-06-30 Human Genome Sciences, Inc. Fibroblast growth factor 15
JPH1149701A (en) * 1997-08-04 1999-02-23 Toagosei Co Ltd Anticancer agent
GB9727524D0 (en) 1997-12-31 1998-02-25 Pharmacia & Upjohn Spa Synergistic antitumor composition containing a biologically active ureido compound
PL348634A1 (en) 1998-05-15 2002-06-03 Imclone Systems Inc Treatment of human tumors with radiation and inhibitors of growth factor receptor tyrosine kinases
US6420335B1 (en) 1998-06-15 2002-07-16 Dana Farber Cancer Institute, Inc. Combination of radiotherapy and anti-angiogenic factors

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5716981A (en) * 1993-07-19 1998-02-10 Angiogenesis Technologies, Inc. Anti-angiogenic compositions and methods of use
US5574026A (en) * 1994-12-13 1996-11-12 American Cyanamid Company Methods for inhibiting angiogenesis proliferation of endothelial or tumor cells and tumor growth

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
See also references of EP1206234A2 *
Stratagene catalog, "Gene Characterization Kits", 1988, page 39, XP002938362 *

Cited By (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1328264A4 (en) * 2000-07-25 2004-01-28 Upjohn Co Use of estramustine phosphate in the treatment of bone metastasis
EP1328264A1 (en) * 2000-07-25 2003-07-23 PHARMACIA &amp; UPJOHN COMPANY Use of estramustine phosphate in the treatment of bone metastasis
US7276228B2 (en) 2001-04-24 2007-10-02 Supergen, Inc. Methods for treating hematological disorders through inhibition of DNA methylation and histone deacetylase
US7186693B2 (en) 2001-08-16 2007-03-06 Kimberly - Clark Worldwide, Inc. Metalloproteinase inhibitors for wound healing
US7071164B2 (en) 2001-08-16 2006-07-04 Kimberly-Clark Worldwide, Inc. Anti-cancer and wound healing compounds
US7094754B2 (en) 2001-08-16 2006-08-22 Kimberly-Clark Worldwide, Inc. Anti-aging and wound healing compounds
US7196162B2 (en) 2001-08-16 2007-03-27 Kimberly-Clark Worldwide, Inc. Anti-aging and wound healing compounds
US9271931B2 (en) 2001-10-03 2016-03-01 Celator Pharmaceuticals, Inc. Compositions for delivery of drug combinations
US10058507B2 (en) 2001-10-03 2018-08-28 Celator Pharmaceuticals, Inc. Compositions for delivery of drug combinations
US10722464B2 (en) 2001-10-03 2020-07-28 Celator Pharmaceuticals, Inc. Compositions for delivery of drug combinations
US7850990B2 (en) 2001-10-03 2010-12-14 Celator Pharmaceuticals, Inc. Compositions for delivery of drug combinations
JP2005508920A (en) * 2001-10-03 2005-04-07 セレーター テクノロジーズ インコーポレイテッド Composition for delivering a concomitant drug
JP2010180210A (en) * 2001-10-03 2010-08-19 Celator Pharmaceuticals Inc Composition for delivery of drug combination
US7148194B2 (en) 2002-12-30 2006-12-12 Kimberly-Clark Worldwide, Inc. Method to increase fibronectin
US7189700B2 (en) 2003-06-20 2007-03-13 Kimberly-Clark Worldwide, Inc. Anti-chrondrosarcoma compounds
US7795225B2 (en) 2003-06-20 2010-09-14 Kimberly-Clark Worldwide, Inc. Anti-chrondrosarcoma compounds
JP2007504194A (en) * 2003-09-05 2007-03-01 ゲンチウム エスピーエー Anti-tumor preparation containing defibrotide alone or in combination with other anti-tumor agents
US8551967B2 (en) 2003-09-05 2013-10-08 Gentium Spa Formulations with anti-tumour action
US7968527B2 (en) 2004-02-13 2011-06-28 Boston Biomedical Research Institute Inhibition of FGF signaling
EP1742961A4 (en) * 2004-02-13 2009-07-15 Boston Biomedical Res Inst Inhibition of fgf signaling
EP1742961A2 (en) * 2004-02-13 2007-01-17 Boston Biomedical Research Institute Inhibition of fgf signaling
AU2005265067B2 (en) * 2004-06-18 2010-12-16 Genentech, Inc. Combination of a chemotherapeutic agent and an antagonist of a gene product for treating tumors
WO2006009805A3 (en) * 2004-06-18 2007-01-04 Genentech Inc Combination of a chemotherapeutic agent and an antagonist of a gene product for treating tumors
US8147827B2 (en) 2004-06-18 2012-04-03 Genentech, Inc. Tumor treatment
US7250416B2 (en) 2005-03-11 2007-07-31 Supergen, Inc. Azacytosine analogs and derivatives
US8461123B2 (en) 2005-09-29 2013-06-11 Astex Pharmaceuticals, Inc. Oligonucleotide analogues incorporating 5-aza-cytosine therein
US10456415B2 (en) 2005-09-29 2019-10-29 Astex Pharmaceuticals, Inc. Oligonucleotide analogues incorporating 5-aza-cytosine therein
US9358248B2 (en) 2005-09-29 2016-06-07 Astex Pharmaceuticals, Inc. Oligonucleotide analogues incorporating 5-aza-cytosine therein
US10933079B2 (en) 2005-09-29 2021-03-02 Astex Pharmaceuticals, Inc. Oligonucleotide analogues incorporating 5-aza-cytosine therein
US9480698B2 (en) 2005-09-29 2016-11-01 Astex Pharmaceuticals, Inc. Oligonucleotide analogues incorporating 5-aza-cytosine therein
US7700567B2 (en) 2005-09-29 2010-04-20 Supergen, Inc. Oligonucleotide analogues incorporating 5-aza-cytosine therein
WO2010140795A2 (en) * 2009-06-01 2010-12-09 (주)차바이오앤디오스텍 Human-placenta-like composition containing a human-placenta-derived growth factor and cytokines, and cosmetic use thereof
WO2010140795A3 (en) * 2009-06-01 2011-04-21 (주)차바이오앤디오스텍 Human-placenta-like composition containing a human-placenta-derived growth factor and cytokines, and cosmetic use thereof
US8980862B2 (en) 2010-11-12 2015-03-17 Gentium S.P.A. Defibrotide for use in prophylaxis and/or treatment of Graft versus Host Disease (GVHD)
US9539277B2 (en) 2010-11-12 2017-01-10 Gentium S.R.L. Defibrotide for use in prophylaxis and/or treatment of graft versus host disease (GVHD)
US9867843B2 (en) 2010-11-12 2018-01-16 Gentium S.R.L. Defibrotide for use in prophylaxis and/or treatment of graft versus host disease (GVHD)
US10517886B2 (en) 2011-08-30 2019-12-31 Astex Pharmaceuticals, Inc. Drug formulations
US9913856B2 (en) 2011-08-30 2018-03-13 Astex Pharmaceuticals, Inc. Drug formulations
US9381207B2 (en) 2011-08-30 2016-07-05 Astex Pharmaceuticals, Inc. Drug formulations
US9902952B2 (en) 2012-06-22 2018-02-27 Gentrum S.R.L. Euglobulin-based method for determining the biological activity of defibrotide
US11085043B2 (en) 2012-06-22 2021-08-10 Gentium S.R.L. Euglobulin-based method for determining the biological activity of defibrotide
US11236328B2 (en) 2012-06-22 2022-02-01 Gentium S.R.L. Euglobulin-based method for determining the biological activity of defibrotide
US11746348B2 (en) 2012-06-22 2023-09-05 Gentium S.R.L. Euglobulin-based method for determining the biological activity of defibrotide
US10393731B2 (en) 2014-11-27 2019-08-27 Gentium S.R.L. Cellular-based method for determining the biological activity of defibrotide
RU2723091C2 (en) * 2015-03-23 2020-06-08 Дойчес Кребсфоршунгсцентрум Штифтунг Дес Эффентлихен Рехтс Oligonucleotide sequences aimed at the transcription factor tsc22d4, for treating insulin resistance
US10485764B2 (en) 2015-07-02 2019-11-26 Otsuka Pharmaceutical Co., Ltd. Lyophilized pharmaceutical compositions
US10519190B2 (en) 2017-08-03 2019-12-31 Otsuka Pharmaceutical Co., Ltd. Drug compound and purification methods thereof

Also Published As

Publication number Publication date
US20040010001A1 (en) 2004-01-15
JP2003503313A (en) 2003-01-28
IL146872A (en) 2006-10-31
AU780454B2 (en) 2005-03-24
US6599912B1 (en) 2003-07-29
US7625860B2 (en) 2009-12-01
EP1206234A4 (en) 2005-06-01
WO2000074634A9 (en) 2002-09-26
CA2377385A1 (en) 2000-12-14
KR100903243B1 (en) 2009-06-17
IL146872A0 (en) 2002-08-14
WO2000074634A3 (en) 2001-08-23
EP1206234A2 (en) 2002-05-22
KR20020019924A (en) 2002-03-13
AU5790300A (en) 2000-12-28

Similar Documents

Publication Publication Date Title
US7625860B2 (en) Methods and compositions for modulating cell proliferation and cell death
ES2929614T3 (en) Anti-MUC16 antibodies and uses thereof
JP7001731B2 (en) Use of bacteria, bacterial products, and other immunomodulatory entities in combination with anti-CTLA-4 and / or anti-PD-1 antibodies to treat solid tumor malignancies
JP6385277B2 (en) Anti-CEACAM1 recombinant antibody for cancer treatment
DE60317677T2 (en) OX40 (= CD134) RECEPTOR AGONISTS AND THERAPEUTIC USES
JP4857259B2 (en) Use of anti-α5β1 antibodies to inhibit cancer cell growth
CN107135654A (en) Giant cell drinks the anti-CD46 antibody of the mankind and target on cancer therapy
US20030082188A1 (en) Treatment of prostate cancer by inhibitors of NCAM2
JP2002539076A (en) Methods and compositions for inhibiting angiogenesis
CN104768579A (en) Methods of treating neuroendocrine tumors using Wnt pathway-binding agents
US20220289831A1 (en) Use of beta-catenin as a biomarker for treating cancers using anti-dkk-1 antibody
WO2005019270A2 (en) Endotheliase-2 ligands
JP2003527441A (en) Drugs that block cell cycle and drugs that contain antibodies
JP6941565B2 (en) Compounds and compositions useful for treating or preventing cancer metastasis, and their usage
EP2984108B1 (en) Anti-s100a7 antibodies for the treatment and diagnosis of cancer
CN112739716A (en) Methods of treating cancer using anti-PD-1 antibodies in combination with anti-tissue factor antibody-drug conjugates
KR20220131223A (en) How to use a DKK-1 inhibitor to treat cancer
US20050281821A1 (en) Method and composition for angiogenesis inhibition
KR20120123619A (en) Polypeptide compounds for inhibiting angiogenesis and tumor growth
RU2361614C2 (en) APPLICATION OF ANTIBODIES AGAINST α5β1 FOR INHIBITION OF CANCER CELLS PROLIFERATION

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

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

AL Designated countries for regional patents

Kind code of ref document: A2

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

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

Ref document number: 2377385

Country of ref document: CA

ENP Entry into the national phase

Ref country code: JP

Ref document number: 2001 501171

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1020017015591

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 57903/00

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2000943429

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1020017015591

Country of ref document: KR

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 2000943429

Country of ref document: EP

AK Designated states

Kind code of ref document: C2

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

AL Designated countries for regional patents

Kind code of ref document: C2

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

COP Corrected version of pamphlet

Free format text: PAGES 1-124, DESCRIPTION, REPLACED BY NEW PAGES 1-124; PAGES 125-135, CLAIMS, REPLACED BY NEW PAGES125-135; PAGES 1/7-7/7, DRAWINGS, ADDED

WWG Wipo information: grant in national office

Ref document number: 57903/00

Country of ref document: AU