WO2009152901A1 - Methods for treatment of malignancies - Google Patents

Methods for treatment of malignancies Download PDF

Info

Publication number
WO2009152901A1
WO2009152901A1 PCT/EP2009/003372 EP2009003372W WO2009152901A1 WO 2009152901 A1 WO2009152901 A1 WO 2009152901A1 EP 2009003372 W EP2009003372 W EP 2009003372W WO 2009152901 A1 WO2009152901 A1 WO 2009152901A1
Authority
WO
WIPO (PCT)
Prior art keywords
fusion protein
administered
rgd peptide
angiogenic
patient
Prior art date
Application number
PCT/EP2009/003372
Other languages
French (fr)
Inventor
Stefan Krueger
Matthias Dotzauer
Karl Josef Kallen
Ralph A. Reisfeld
Original Assignee
Merck Patent Gmbh,
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 Merck Patent Gmbh, filed Critical Merck Patent Gmbh,
Publication of WO2009152901A1 publication Critical patent/WO2009152901A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3076Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells against structure-related tumour-associated moieties
    • C07K16/3084Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells against structure-related tumour-associated moieties against tumour-associated gangliosides
    • 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
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/55IL-2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the invention relates to methods for inhibition of malignancies, more preferably of primary malignancies and especially of primary disseminated malignancies, such as melanomas and neuroblastomas, and metastases thereof, using a therapy based on the combined administration of an anti- angiogenic agent and a targeted anti-tumor agent.
  • angiogenesis plays a key role in the growth of malignant disease and has generated much interest in developing agents that inhibit angiogenesis (see for example Holmgren, L., O'Reilly, M.S. & Folkman, J. (1995) "Dormancy of micrometastases: balanced proliferation and apoptosis in the presence of angiogenesis suppression", Nature Medicine 1 , 149-153; Folkman, J.
  • Angiogenesis is characterized by invasion, migration and proliferation of endothelial cells, processes that depend on cell interactions with extracellular matrix components.
  • the endothelial adhesion receptor of integrin ⁇ v ⁇ 3 was shown to be a key player by providing a vasculature- specific target for anti-angiogenic treatment strategies.
  • Murine hybridoma LM609 has been deposited with the American Type Culture Collection (ATCC, Manassas, VA, USA) as the International Depository Authority under the Budapest Treaty, and assigned the ATCC Designation HB 9537, on September 15, 1987.
  • ATCC American Type Culture Collection
  • Such antagonists block the ligation of integrin ⁇ v ⁇ 3 which promotes apoptosis of the proliferative angiogenic vascular cells and thereby disrupt the maturation of newly forming blood vessels, an event essential for the proliferation of tumors.
  • a major obstacle for effective treatment of malignancies and/or disseminated malignancies includes minimal residual disease characterized by micrometastases that lack a well-established vascular supply for delivery of therapeutics.
  • a novel immunotherapeutic strategy proved efficient in using tumor compartment-specific monoclonal antibodies to direct cytokines to the tumor microenvironment.
  • Combination therapies using antiangiogenic agents with other chemotherapeutic agents has been used to enhance treatment of various types of cancer, although the ultimate effectiveness of such combinations can be difficult to predict.
  • ch14.18-IL2 The chimeric anti-GD2-IL-2 fusion protein known as ch14.18-IL2 has been investigated as a treatment for melanoma, alone, and in combination with cilengitide, with some limited degree of success, however, ch14.18-IL2 is known to elicit undesirably high levels of human anti-chimeric antibody (HACA) responses and allergic reactions in humans.
  • HACA human anti-chimeric antibody
  • the present invention is directed to a method for treating a malignant neoplasm, more preferably a disseminated malignant neoplasm such as melanoma or neuroblastoma in a patient.
  • the method comprises administering to the patient in need of such treatment a malignant cell proliferation inhibiting amount of an anti-angiogenic RGD peptide and a humanized anti-GD2 antibody-interleukin-2 (IL-2) fusion protein, preferably a fusion protein of IL-2 with the humanized anti-GD2 antibody designated as hu14.18.
  • IL-2 humanized anti-GD2 antibody-interleukin-2
  • Inhibition of malignant cell proliferation can encompass inhibition of the growth of malignant cells such as neuroblastoma cells, melanoma cells, and the like in an existing tumor or in tumor metastases, inhibition of the formation of additional tumor metastases, and even tumor cell death.
  • the anti-angiogenic RGD peptide and the humanized anti-GD2-IL-2 fusion protein can be administered substantially concurrently, as well as sequentially.
  • compositions i.e., the anti-angiogenic RGD peptide and the humanized anti-GD2-IL-2 fusion protein
  • administration can be accomplished via direct immersion; systemic or localized intravenous (i.v.), intraperitoneal (i p ), subcutaneous (s.c), intramuscular (i.m.), or direct injection into a tumor mass; and/or by oral administration of the appropriate formulations.
  • Anti-angiogenic RGD peptides suitable for use in the methods of the present invention include linear and cyclic polypeptides containing the Arg-Gly-Asp (RGD) integrin binding motif, which peptides bind to the ⁇ v ⁇ 3 and/or ⁇ v ⁇ s receptor and inhibit angiogenesis.
  • Anti-angiogenic RGD peptides are described in U.S. Patent No. 5,262,520 (Plow et al.) and WO 97/45137.
  • a particularly preferred RGD peptide is cilengitide, i.e., cyclo-(Arg-Gly-Asp-D-Phe-N Me-VaI), SEQ ID NO: 1.
  • a preferred humanized anti-GD2-IL-2 fusion protein comprises an immunoglobulin (Ig) polypeptide that specifically binds to the GD2 antigen, bound to IL-2, either directly or through a linker peptide.
  • the antibody fusion protein comprises two Ig polypeptide chains with a IL-2 polypeptide fused to each chain.
  • the humanized anti-GD2-IL-2 fusion protein comprises a GD2-binding antibody variable domain (e.g., at least one light chain variable region peptide and at least one heavy-chain variable region peptide that includes complementarity determining regions (CDRs) that specifically bind to GD2).
  • CDRs complementarity determining regions
  • the GD2-targeting Ig portion of the fusion protein can comprise an entire immunoglobulin chain amino acid sequence, or at least the fragment thereof that comprises the GD2 antigen binding specificity portion of the protein.
  • the GD2-binding portion of the fusion protein is humanized to reduce the potential for adverse immunological effects when administering the fusion protein to a human subject.
  • FIG. 1 shows the amino acid sequence of the light chain (SEQ ID NO: 2) of hu14.18-IL2.
  • FIG. 1 shows the amino acid sequence of the heavy chain (SEQ ID NO: 3) of hu14.18-IL2, the C-terminal region of which includes the IL-2 amino acid sequence.
  • the suppression and eradication of primary tumors and distant metastases is a major goal of alternative treatment strategies for cancer, such as inhibition of angiogenesis and targeted immunotherapy.
  • the methods of the invention utilize the combined administration of at least one angiogenesis inhibiting RGD peptide with at least one, humanized anti-GD2-IL-2 fusion protein in a therapeutic amount sufficient to inhibit proliferation of melanoma cells, neuroblastoma cells, and the like.
  • a preferred RGD peptide for use in the present invention is the cyclic pentapeptide known as cilengitide, i.e., cyclo-(Arg-Gly-Asp-D-Phe-NMe-Val), SEQ ID NO: 1 , which can be administered as a pharmaceutically acceptable salt thereof, e.g., a hydrochloride salt, or as the inner salt, which is cyclo- (Arg-Gly-Asp-D-Phe-NMe-Val), SEQ ID NO: 1 , perse.
  • cilengitide i.e., cyclo-(Arg-Gly-Asp-D-Phe-NMe-Val)
  • SEQ ID NO: 1 which can be administered as a pharmaceutically acceptable salt thereof, e.g., a hydrochloride salt, or as the inner salt, which is cyclo- (Arg-Gly-Asp-D-Phe-NMe-Val), SEQ ID NO: 1 ,
  • hybridomas typically are mouse-derived, or are derived from other rodents.
  • HAMA human anti-mouse antibodies
  • Methods to overcome this barrier include humanization of murine antibody proteins by substituting antigenic amino acids of the mouse protein with human protein sequences that are assumed to be less antigenic.
  • Other methods involve the grafting of the binding specificity determining amino acid residues or regions into human protein frameworks.
  • the ability to express an antibody protein in phage display systems allows for selection of antibodies that have been subjected to mutagenesis to either improve binding or to reduce immunogenicity (See for example Antibody Engineering (McCafferty, J. et al. editors, Oxford University Press, 1996). Recently, the substitution of human immunoglobulin genes into transgenic mice has allowed the use of murine hosts to generate antibodies, and thus monoclonal antibodies, that are of human nucleic acid sequence origin.
  • Antibodies can also be reduced in size by fragmentation to allow for reduced antigenicity, or to create smaller therapeutic molecules.
  • a whole antibody protein can be reduced, either by digestion with the appropriate enzymes or by producing the smaller protein by recombinant DNA methods.
  • Suitable fragments include at least an antigen binding portion of the whole molecule, and can comprise Fab, F(ab)2, F(ab)3, Fv or single chain Fv (single chain antibody; SCA) constructs.
  • Al! monoclonal antibody-based components of the therapeutic agents for use in the methods of the invention are humanized to reduce potential allergic reactions and HACA responses when administered to humans.
  • anti-GD2-IL2 fusion protein encompasses an anti- GD2 Ig polypeptide or the antigen-binding portion thereof, bound to IL-2.
  • the IL-2 can be directly bound to the Ig polypeptide or can be bound thereto via a linker peptide.
  • Hu14.18-IL2 comprises the immunoglobulin light chain sequence of SEQ ID NO: 2 and the immunoglobulin heavy chain sequence of SEQ ID NO: 3 (see FIG. 1 and
  • the Ig polypeptide chain is an Ig heavy chain polypeptide, a light chain polypeptide, or both, which comprise an N-terminal variable region specific for the GD2 antigen.
  • the fusion protein typically has the Ig polypeptide joined at its carboxy-terminus by a peptide bond to the amino terminus of IL-2.
  • the Ig heavy chain further typically comprises CH1 and CH2 domains, and may optionally further contain a CH3 domain.
  • such constant region domains can be eliminated to reduce immunogenicity, size, or non-specific binding of the resultant fusion protein construct.
  • the humanized anti- GD2-IL-2 fusion protein comprises a complete antibody with one IL-2 peptide attached to the carboxy-terminus of each Ig polypeptide chain of the antibody.
  • Therapeutic methods of the present invention for treating malignant tumor cells in tumors and tumor metastases are based on the combined use of an anti-angiogenic RGD peptide and a humanized anti-GD2-IL-2 fusion protein.
  • the combined use can be performed simultaneously, sequentially, or with the intervention of a period of time between the treatments. Any of the specific therapeutics may be administered more than once during a course of treatment.
  • the method of the invention provides a synergistic potentiation of the tumor cell proliferation inhibition effect of each individual therapeutic, yielding more effective treatment than found by administering an individual component alone.
  • the method of the invention comprises a variety of modalities for practicing the invention in terms of the steps.
  • the RGD peptide and the humanized anti-GD2-IL-2 fusion protein can be administered following admixture, i.e., simultaneously, or can be administered sequentially, i.e., separately.
  • the RGD peptide and the fusion protein can be separately administered within a time interval of about 3 weeks between administrations, i.e., from substantially immediately after the first active agent is administered to up to about 3 weeks after the first agent is administered.
  • the order of administration can be varied, i.e., that the RGD peptide can be administered prior to administration of the fusion protein, or that administration can be conducted in the reverse order.
  • terapéuticaally effective refers to an amount of a drug effective to treat a disease or disorder in a mammal.
  • the therapeutically effective amount of a therapeutic agent may reduce the number of malignant cells; reduce the tumor size; inhibit (i.e., slow to some extent and preferably stop) malignant cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve to some extent one or more of the symptoms associated with the cancer.
  • the drug may prevent growth and/or kill existing cancer cells, it may be cytostatic and/or cytotoxic.
  • efficacy can, for example, be measured by assessing the time to disease progression (TTP) and/or determining the response rate (RR).
  • the method of the invention also encompasses administering to a patient suffering from a melanoma, a neuroblastoma, or the metastases thereof, an angiogenesis inhibiting amount of the anti- angiogenic RGD peptide, and an amount of humanized anti-GD2-IL-2 fusion protein sufficient to elicit a biological response.
  • a therapeutically effective amount of a humanized anti-GD2-IL-2 fusion protein comprising a complete antibody fused to IL-2 is an amount such that when administered in physiologically tolerable composition is sufficient to achieve a plasma concentration of about 0.01 microgram ( ⁇ g) per milliliter (ml) to about 100 ⁇ g/ml, preferably about 1 ⁇ g/ml to about 5 ⁇ g/ml and usually about 5 ⁇ g/ml based on the antibody portion of the fusion protein.
  • the dosage can vary from about 0.1 mg/kg to about 300 mg/kg, preferably about 0.2 mg/kg to about 200 mg/kg, most preferably about 0.5 mg/kg to about 20 mg/kg, in one or more dose administrations daily, for one or several days, again based on the antibody portion of the fusion protein.
  • the Ig polypeptide portion of the fusion protein is in the form of a fragment of a monoclonal antibody or a conjugate
  • the amount can readily be adjusted based on the mass of the fragment / conjugate relative to the mass of the complete antibody.
  • a preferred plasma concentration, expressed as molarity is about 2 micromolar ( ⁇ M) to about 5 millimolar (mM), and preferably, about 100 ⁇ M to about 1 mM of the fusion protein.
  • a therapeutically effective amount of an RGD peptide according of this invention typically is an amount of RGD peptide that, when administered in a physiologically tolerable composition, is sufficient to achieve a plasma concentration of from about 0.1 microgram ( ⁇ g) per milliliter (ml) to about 200 ⁇ g/ml, preferably from about 1 ⁇ g/ml to about 150 ⁇ g/ml.
  • the preferred plasma concentration expressed as molarity, is about 2 micromolar ( ⁇ M) to about 5 millimolar (mM), and preferably about 100 ⁇ M to 1 mM of the RGD peptide.
  • Cilengitide has a molecular mass of about 589 g/mol.
  • the preferred dosage of an active agent, e.g., the RGD peptide or anti-GD2- IL-2 fusion protein, for administration to a patient is about 0.5 mg to about
  • the preferred active agent dosage level preferably is about 0.1 to about 100 mg/kg, and more preferably about 1 mg to about 50 mg/kg, preferably per dosage unit and more preferably per day.
  • the preferred dosage level of an active agent is about 0.5 mg to about 2000 mg/m 2 , more preferably about 5 to about 1500 mg/m 2 ' and especially about 50 to about 1000 mg/m 2 , preferably per dosage unit and more preferably per day.
  • the preferred dosage of the RGD peptide for administration to a patient is about 500 mg, about 1000 mg or about 2000 mg per patient and day, preferably if given "flat", e.g. as a flat dosing, which preferably means that the amount of RGD peptide given is not adjusted to the body weight or surface area of the respective patient.
  • the afore described dosage of about 500 mg, about 1000 mg or about 2000 mg per patient and day (flat) is typically administered at intervals ranging from once a month to once a day, more preferably ranging from once a week to five times a week and especially about once a week, about twice a week or about five times a week.
  • this dosing is applied to a patient for two or more consecutive weeks, more preferably four or more consecutive weeks.
  • this dosing can be applied to the patient until healing, stable disease or progression of the patient occurs.
  • this dosing can be applied to a patient for up to 52 weeks, up to 104 weeks, up to 156 weeks or even longer.
  • the treatment of malignancies, at least the treatment of malignancies with cancer, at least the treatment of cancer with therapeutic agents in the broadest sense, is a protracted issue.
  • the treatment of cancer with therapeutic agents generally includes a prolonged exposure to one or more respective therapeutic agents.
  • a treatment regimen comprises the administration time period of the respective therapeutic agent and the recovery time period after the administration of the respective therapeutic agent is repeated one or more times, preferably several times. Each repeat of the course of treatment is usually referred to by the skilled artisan as a "cycle" (or "treatment course").
  • Each cycle comprises the administration time period of the respective therapeutic agent and the recovery time period after the administration of the respective therapeutic agent.
  • the duration of the administration time period and/or the recovery time period after the administration of the therapeutic usually depend on the properties of the respective therapeutic agent. Accordingly, different therapeutic agents can have different durations of the administration time period and/or the recovery time period after the therapeutic agent is administered. Thus, the length or duration of a cycle can be different for different therapeutic agents.
  • the length of a cycle is in the range of about one week to about 12 weeks, more preferably one week to six weeks, and especially 2 to 4 weeks.
  • the dosing of the respective therapeutic agent is given in an amount per cycle, allowing a medical practitioner to adapt the actual administration regimen to the medical status of the patient, i.e., the amount of a particular agent administered per cycle can be a single administration or can be divided into two or more portions administered at different times within the cycle, if desired.
  • a combination treatment comprising two or more therapeutic agents, as in the present invention, generally two or more cycles, one for each agent, having the same or a different length, run in parallel.
  • each portion is preferably given on a different day within said cycle.
  • each of the therapeutic agents administered generally more than one cycle, preferably two or more cycles, even more preferably three or more cycles are applied to the patient, preferably substantially with out a pause. Generally, not more than 24 cycles are applied to the patient substantially without a pause.
  • the administration of about six cycles substantially without a pause to the patient for each of the therapeutic agents administered is generally a standard.
  • the RGD peptide is administered to a patient in an amount of about 250 mg to about 12500 mg per week, preferably for one or more weeks of a cycle/treatment course.
  • One cycle/treatment course according to this invention preferably consists of 21 to 35 sequential days, and more preferably of about 28 days or about 4 weeks.
  • the RGD peptide preferably cilengitide
  • the RGD peptide can be administered to the patient by any method or mode or administration known in the art.
  • the RGD peptide can be administered to the patient in an amount of about 250 mg to about 12500 mg, more preferably about 450 to about 10500 mg, within a time period of about one week. This is also referred to as the weekly administration with respect to the RGD peptide.
  • a weekly administration of the given amounts of RGD peptide takes place once or twice or more times, preferably once, twice, or three times, within a time period of about three weeks.
  • a weekly administration of the given amounts of RGD peptide takes place once or twice, preferably once, within a time period of about four weeks.
  • the weekly administration of the RGD peptide takes place during two or more weeks within the cycle or the cycles of administration of the anti-GD2-IL-2 fusion protein. In another preferred embodiment, the weekly administration of the RGD peptide takes place during every week within the cycle or the cycles of administration of the anti- GD2-IL-2 fusion protein. In another more preferred embodiment, the weekly administration of the RGD peptide takes place during one week within the cycle or cycles of administration of the anti-GD2-IL-2 fusion protein.
  • the weekly administration of the RGD peptide takes place once in the cycle or cycles of administration of the anti-GD2-IL-2 fusion protein, and more preferably within one week in which the anti-GD2-IL-2 fusion protein is also administered.
  • the amount of RGD peptide to be administered in the weekly administration can be the same or different in each week.
  • the following dosages or regimens are preferred in this respect: (a) the RGD peptide, preferably cilengitide, is administered to the patient in an amount of about 500 mg to about 2000 mg in a single dose each week during the cycle of administration of the anti-GD2-IL-2 fusion protein; (b) the RGD peptide, preferably cilengitide, is administered to the patient in an amount of about 500 mg to about 2000 mg twice a week each week during the cycle of administration of the anti-GD2-IL-2 fusion protein; (c) the RGD peptide, preferably cilengitide, is administered to the patient in an amount of about 500 mg each day on five consecutive days within one first week and in an amount of about 500 mg on one day within each additional week during the cycle of administration of the anti-GD2-IL-2 fusion protein; (d) the RGD peptide, preferably cilengitide, is administered to the patient
  • more than one cycle of the RGD peptide is administered to the patient. More preferably 2 to 12 cycles, and especially about 6 cycles are administered to the patient, preferably comprising one or more of regimens (a) to (f) discussed above.
  • the more than one cycle of RGD peptide administration comprises only one of the regimens selected from (a) to (f), i.e., the same regimen selected from (a) to (f) is applied to the patient in each cycle of administration. More preferably, the same regimen selected from (a) to (f) is administered to the patient in each of about 6 cycles.
  • the more than one cycles of RGD peptide administration comprises two or more of the regimens selected from (a) to (f) above, i.e., in different cycles, a different regimen is selected from (a) to (f) for administration to the patient.
  • regimens (a) to (f) are also administered to the patient.
  • regimen (c) can be applied to the patient for the first cycle, followed by regimen (a) for 1 to 11 cycles, and especially about 5 cycles.
  • the weekly administration of the RGD peptide consists of about 500 mg.
  • regimen (d) can be applied to the patient for the first cycle, followed by regimen (a) for 1 to 11 cycles, and especially about 5 cycles.
  • the weekly administration of the RGD peptide consists of about 2000 mg.
  • the amount of RGD peptide to be administered in the weekly administration can be the same or different in each week.
  • the dosages of the RGD peptide, preferably cilengitide are selected from the dosages or regimen (a) to (f) as described before and mixtures thereof and more preferably are selected from the dosages or regimen (a), (b), (d), (e) and (T); especially preferred in this respect is regimen or dosing (f).
  • more than one such cycle e.g., 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 of such cycles with respect to the RGD peptide are performed substantially without a pause.
  • the duration of one RGD peptide administration cycle is about three weeks (about 21 days) or about four weeks (about 28 days), more preferably about four weeks (about 28 days).
  • this RGD peptide also can be administered to the patient outside the administration cycles of the anti-GD2-IL-2 fusion protein. This is especially advantageous as a maintenance therapy consisting of or comprising, preferably consisting of, the administration of cilengitide, the pharmaceutically acceptable derivatives, solvates and/or salts thereof, for one or several months, for example, for up to about 24 months, even substantially without pause.
  • a dosage of both the humanized anti-GD2-IL-2 fusion protein and the RGD peptide preferably is based on an amount per square meter of body surface area.
  • the anti-GD2-IL-2 fusion protein e.g., hu14.18-IL2
  • the anti-GD2-IL-2 fusion protein (e.g., hu14.18-IL2) is administered in an amount of about 1 - 100 mg/m 2 , more preferably about 2 - 80 mg/m 2 , and even more preferably about 3 - 60 mg/m 2 , e.g. in an amount of about 12 mg/m 2 , of about 24 mg/m 2 , or about 36 mg/m 2 , per week solely in the first week during one cycle.
  • the amounts of anti-GD2-IL-2 fusion protein (e.g., hu14.18-IL2) given above are administered in 1 to 5, more preferably about 3 approximately equal portions, each portion being given on a separate day, preferably each portion being given on a separate, but consecutive day.
  • the anti-GD2-IL-2 fusion protein (e.g., hu14.18-IL2) is administered in an amount of about 1 - 20 mg/m 2 per day, more preferably about 2 - 15 mg/m 2 per day, and even more preferably about 4 - 12 mg/m 2 per day, e.g.
  • the anti-GD2-IL-2 fusion protein (e.g., hu14.18-IL2) is administered in an amount of about 4 mg/m 2 per day, of about 8 mg/m 2 per day, or about 12 mg/m 2 per day on 3 consecutive days within one week during solely one week of one cycle, preferably solely during the first week of the cycle.
  • more than one such cycle e.g., 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 such cycles are performed substantially without a pause.
  • the RGD peptide e.g., cilengitide
  • the RGD peptide is administered in an amount of about 0.5 - 2.5 g/m 2 per day, more preferably about 1 - 2 g/m 2 per day, and even more preferably about 1.1 - 1.9 g/m 2 per day, e.g. in an amount of about 1.2 g/m 2 per day, of about 1.4 g/m 2 per day, or about 1.8 g/m 2 per day, on 1 to 3 days and preferably about 2 days within one week during solely one week of one cycle, preferably solely during the first week of the cycle.
  • the RGD peptide e.g., cilengitide
  • the RGD peptide (e.g., cilengitide) is administered in an amount of about 0.5 - 2.5 g/m 2 per day, more preferably about 1 - 2 g/m 2 per day, and even more preferably about 1.1 - 1.9 g/m 2 per day, e.g. in an amount of about 1.2 g/m 2 per day, of about 1.4 g/m 2 per day, or about 1.8 g/m 2 per day, on Day 1 and Day 3 within one week, during solely one week of one cycle, preferably on Day 1 and Day 3 solely during the first week of the cycle.
  • the RGD peptide e.g., cilengitide
  • more than one such cycle e.g., 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 such cycles are performed substantially without a pause.
  • more than one such cycle e.g. 2 to 12, more preferably 2 to 8 and especially about 3 or about 6 of such cycles with respect to both the anti-GD2-IL-2 fusion protein and the RGD peptide are performed substantially without a pause, preferably substantially in parallel.
  • the amounts of the RGD peptide (e.g., cilengitide) given above are administered in 1 to 5, more preferably into about 2 or about 3, preferably approximately equal portions, each portion given on a separate day, preferably each given on a separate day within one week.
  • the invention can be practiced in conjunction with surgical procedures where portions or all of a tumor mass has been removed.
  • the method can be practiced following a surgical procedure.
  • the surgical procedure can be practiced during the interval between administration of the first active agent and the second active agent.
  • Exemplary of this method is the combination of the present method with surgical tumor removal described below.
  • Administration can be accomplished by periodic unit dosages, by continuous infusion, peristaltic delivery, by bolus injection, and the like. Routes can include intravenous, subcutaneous, intramuscular, orthotopic injection, orthotopic infusion, oral application, and the like.
  • Another aspect of treating melanoma is the use of an anti-angiogenic RGD peptide for the manufacture of a medicament for the treatment of melanomas, wherein the medicament is to be applied in combination with a humanized anti-GD2-IL-2 fusion protein.
  • the humanized anti-GD2-IL-2 fusion protein preferably comprises, or more preferably is, hu14.18-IL2; and/or the RGD peptide comprises, or more preferably is, cilengitide or a pharmaceutically acceptable salt thereof.
  • both the humanized anti-GD2-IL-2 fusion protein and the anti-angiogenic RGD peptide are preferably administered to the patient as separate pharmaceutical compositions.
  • the humanized anti-GD2-IL-2 fusion protein and the anti-angiogenic RGD peptide, or the pharmaceutical compositions containing them, can be administered to the patient on separate days and/or both on the same day, whereby the administration on separate days and the administration of both on the same day can alternate.
  • the anti-angiogenic RGD peptide or the pharmaceutical composition containing it is preferably administered to the patient prior to the administration of the humanized anti-GD2-IL-2 fusion protein or the pharmaceutical composition containing it, preferably 30 minutes to 6 hours and more preferably 1 hour to 3 hours, e.g., about one hour, about two hours or about three hours, prior to the administration of the humanized anti-GD2-IL-2 fusion protein or the pharmaceutical composition containing it.
  • the anti-angiogenic RGD peptide or the pharmaceutical composition containing it is then administered to the patient over a time period of 15 minutes to 2 hours, more preferably 30 minutes to 90 minutes, e.g., over a time period of about 1 hour, for example by i.v. infusion, and then, about directly or alternatively with a pause, preferably a pause of 10 minutes to 90 minutes, more preferably 20 to 70 minutes, e.g., a pause of about one hour, the humanized anti-GD2-IL-2 fusion protein or the pharmaceutical composition containing it is administered.
  • the dosage regimens to be administered to the patient and the treatment regimens related thereto discussed herein preferably also apply here.
  • the patient can be an adult or a child.
  • the dosages to be administered to the patient and the treatment regimens related thereto in the treatment of melanoma in children preferably comprise the following:
  • the RGD peptide (e.g., cilengitide) is administered in an amount of about 0.5 - 2.5 g/m 2 per day, more preferably about 1 - 2 g/m 2 per day, and even more preferably about 1.1 - 1.9 g/m 2 per day, e.g., in an amount of about 1.2 g/m 2 per day, of about 1.4 g/m 2 per day, or about 1.8 g/m 2 per day, on 1 to 3 days and preferably about 2 days, during solely one week of one cycle, preferably solely during the first week of the cycle.
  • the RGD peptide e.g., cilengitide
  • the RGD peptide (e.g., cilengitide) is administered in an amount of about 0.5 - 2.5 g/m 2 per day, more preferably about 1 - 2 g/m 2 per day, and even more preferably about 1.1 - 1.9 g/m 2 per day, e.g., in an amount of about 1.2 g/m 2 per day, of about 1.4 g/m 2 per day, or about 1.8 g/m 2 per day, on Day 1 and Day 3 during solely one week of one cycle, preferably on Day 1 and Day 3 solely during the first week of the cycle. Also especially preferably, more than one such cycle, e.g.
  • the anti-GD2-IL-2 fusion protein (e.g., hu14.18-IL2) is administered in an amount of about 1 - 20 mg/m 2 per day, more preferably about 2 - 15 mg/m 2 per day, and even more preferably about 4 - 12 mg/m 2 per day, e.g., in an amount of about 4 mg/m 2 per day, of about 8 mg/m 2 per day, or about 12 mg/m 2 per day, on 2-5 consecutive days and preferably about 3 consecutive days within solely one week of one cycle, preferably solely during the first week of the cycle.
  • the anti-GD2-IL-2 fusion protein e.g., hu14.18-IL2
  • the anti-GD2-IL-2 fusion protein is administered in an amount of about 1 - 20 mg/m 2 per day, more preferably about 2 - 15 mg/m 2 per day, and even more preferably about 4 - 12 mg/m 2 per day, e.g., in an amount of about 4 mg/m 2 per day
  • the anti-GD2- IL-2 fusion protein (e.g., hu14.18-IL2) is administered in an amount of about 4 mg/m 2 per day, of about 8 mg/m 2 per day, or about 12 mg/m 2 per day on 3 consecutive days during solely one week of one cycle, preferably solely during the first week of the cycle. Also especially preferably, more than one such cycle, e.g., 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 of such cycles with respect to the anti-GD2-IL-2 fusion protein are performed substantially without a pause.
  • more than one such cycle e.g., 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 of such cycles with respect to the anti-GD2-IL-2 fusion protein are performed substantially without a pause.
  • more than one such cycle e.g., 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 of such cycles with respect to both the anti- GD2-IL-2 fusion protein and the RGD peptide are performed substantially without a pause, preferably substantially in parallel.
  • the dosages to be administered to the patient and the treatment regimens related thereto in the treatment of melanoma in adults preferably comprise the following:
  • the amount of RGD peptide to be administered in the weekly administration can be the same or different in each week.
  • the dosages of the RGD peptide, preferably cilengitide are selected from the dosages or regimens (a) to (f) as described before and mixtures thereof and more preferably are selected from the dosages or regimens (a), (b), (d), (e) and (f); especially preferred in this respect is regimen or dosage (f).
  • more than one such cycle e.g. 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 of such cycles with respect to the RGD peptide are performed substantially without a pause.
  • the anti-GD2-IL-2 fusion protein (e.g., hu14.18-IL2) is administered in an amount of about 1 - 20 mg/m 2 per day, more preferably about 2 - 15 mg/m 2 per day, and even more preferably about 4 - 12 mg/m 2 per day, e.g. in an amount of about 4 mg/m 2 per day, of about 8 mg/m 2 per day, or about 12 mg/m 2 per day, on 2-5 consecutive days, and preferably about 3 consecutive days, during solely one week of one cycle, preferably solely during the first week of the cycle.
  • the anti-GD2-IL-2 fusion protein e.g., hu14.18-IL2
  • the anti-GD2-IL-2 fusion protein is administered in an amount of about 1 - 20 mg/m 2 per day, more preferably about 2 - 15 mg/m 2 per day, and even more preferably about 4 - 12 mg/m 2 per day, e.g. in an amount of about 4 mg/m 2 per day
  • the anti-GD2- IL-2 fusion protein (e.g., hu14.18-IL2) is administered in an amount of about 4 mg/m 2 per day, of about 8 mg/m 2 per day, or about 12 mg/m 2 per day on 3 consecutive days during solely one week of one cycle, preferably solely during the first week of the cycle. Also especially preferably, more than one such cycle, e.g., 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 of such cycles with respect to the anti-GD2-IL-2 fusion protein are performed substantially without a pause.
  • more than one such cycle e.g., 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 of such cycles with respect to the anti-GD2-IL-2 fusion protein are performed substantially without a pause.
  • more than one such cycle e.g., 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 of such cycles with respect to both the anti- GD2-IL-2 fusion protein and the RGD peptide are performed substantially without a pause, preferably substantially in parallel.
  • One preferred aspect of the instant invention is a method of the treatment of melanoma wherein the above described use, medicaments and/or pharmaceutical compositions are applied to the patient as described with respect to said use, medicaments and/or pharmaceutical compositions, as described above.
  • Human experience with hu14.18-IL2 Human experience with hu14.18-IL2
  • hu14.18-IL2 An initial Phase I protocol for hu14.18-IL2 was performed. A total of 33 subjects with melanoma were enrolled to establish the maximum tolerated dose (MTD). Subjects were enrolled in cohorts of 3 or 6 subjects and administered hu14.18-IL2 at one of the following dose levels: 0.8, 1.6, 3.2, 4.8, 6.0 or 7.5 mg/m 2 /day. Hu14.18-IL2 was administered as a 4-hour i.v. infusion over 3 consecutive days during the first week of each course. Subjects with tumor regression or stable disease were given a second course of treatment with hu14.18-IL2 four weeks after Course 1.
  • MTD maximum tolerated dose
  • hu14.18-IL2 was well tolerated, with most subjects experiencing mild to moderate adverse events (AEs) 1 mainly known IL-2 side effects.
  • Laboratory analyses showed lymphocytosis, expansion of CD16+ and CD56+ NK cells, marked peripheral lymphocyte activation (evidenced by a dose-dependent elevation of circulating slL2R alpha chain), and enhanced ability of peripheral blood mononuclear cells (PBMC) to mediate GD2+ tumor cell killing through ADCC.
  • PBMC peripheral blood mononuclear cells
  • PK results indicate the serum t1/2 of hu14.18-IL2 to be 3.7 hours.
  • All subjects preferably are scheduled to receive at least 3 courses of treatment and more preferably a total of 3 courses of treatment. Treatment can be stopped if it can not be tolerated or if there is recurrent disease.
  • Hu14.18-IL2 is administered as a 4 hour i.v. infusion on Days 1 , 2, and 3 in the first week of each treatment cycle. Each treatment cycle is 4 weeks (+/- 7 days). Subjects in this study will be administered 6 mg/m 2 /d of hu14.18-IL2, a dose corresponding to one dose below the maximal tolerated dose (MTD).
  • MTD maximal tolerated dose
  • Cilengitide therapy is administered on Days 1 and 3 of the 28-day treatment course as a 1-hour i.v. infusion at a fixed dose of 2000 mg/dose. Each treatment cycle is 4 weeks (+/- 7 days).
  • compositions used in a method of this invention comprise the active agent (i.e., the anti-angiogenic RGD peptide or the anti-GD2-IL-2 fusion protein) in a pharmaceutically acceptable carrier, as is well known, and therefore the invention is not to be construed as limited with regards to the composition so long as the concentration of the active agent(s) in the composition is sufficient for delivery (administration) of the recited active agent in the amounts described herein.
  • the active agent i.e., the anti-angiogenic RGD peptide or the anti-GD2-IL-2 fusion protein
  • the RGD peptide e.g., cilengitide
  • the RGD peptide is administered in an amount of about 1 g - 8 g/m 2 , more preferably about 1.5 g - 6 g/m 2 , and even more preferably about 2 g - 4 g/m 2 , e.g. in an amount of about 2.2 g/m 2 , 2.4 g/m 2 , of about 2.8 g/m 2 , or about 3.6 g/m 2 , per week for one or more weeks during one cycle, preferably one, two, three or four weeks during one cycle, more preferably one or two weeks during one cycle and even more preferably for one week during one cycle.
  • the RGD peptide e.g., cilengitide
  • the RGD peptide (e.g., cilengitide) is administered in an amount of about 1 g - 8 g/m 2 , more preferably about 1.5 g - 6 g/m 2 , and even more preferably about 2 g - 4 g/m 2 , e.g. in an amount of about 2.2 g/m 2 , 2.4 g/m 2 , about 2.8 g/m 2 , or about 3.6 g/m 2 , per week solely in the first week during one cycle.
  • more than one such cycle e.g., 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 such cycles are performed substantially without a pause.
  • the amounts of the RGD peptide (e.g., cilengitide) given above are administered in 1 to 5, more preferably into about 2 or about 3, preferably approximately equal portions, each portion given on a separate day, preferably each given on a separate day within one week.
  • the RGD peptide (e.g., cilengitide) is administered in an amount of about 0.5 - 2.5 g/m 2 per day, more preferably about 1 - 2 g/m 2 per day, and even more preferably about 1.1 - 1.9 g/m 2 per day, e.g. in an amount of about 1.2 g/m 2 per day, of about 1.4 g/m 2 per day, or about 1.8 g/m 2 per day, on 1 to 3 days and preferably about 2 days within one week during solely one week of one cycle, preferably solely during the first week of the cycle.
  • the RGD peptide e.g., cilengitide
  • the RGD peptide (e.g., cilengitide) is administered in an amount of about 0.5 - 2.5 g/m 2 per day, more preferably about 1 - 2 g/m 2 per day, and even more preferably about 1.1 - 1.9 g/m 2 per day, e.g. in an amount of about 1.2 g/m 2 per day, of about 1.4 g/m 2 per day, or about 1.8 g/m 2 per day, on Day 1 and Day 3 within one week, during solely one week of one cycle, preferably on Day 1 and Day 3 solely during the first week of the cycle.
  • more than one such cycle e.g., 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 such cycles are performed substantially without a pause.
  • the invention can be practiced in conjunction with surgical procedures where portions or all of a tumor mass has been removed.
  • the method can be practiced following a surgical procedure.
  • the surgical procedure can be practiced during the interval between administration of the first active agent and the second active agent. Exemplary of this method is the combination of the present method with surgical tumor removal described below.
  • Administration can be accomplished by periodic unit dosages, by continuous infusion, peristaltic delivery, by bolus injection, and the like. Routes can include intravenous, subcutaneous, intramuscular, orthotopic injection, orthotopic infusion, oral application, and the like.
  • an anti-angiogenic RGD peptide for the manufacture of a medicament for the treatment of neuroblastomas, wherein the medicament is to be applied in combination with a humanized anti-GD2-IL-2 fusion protein.
  • the humanized anti-GD2-IL-2 fusion protein preferably comprises, or more preferably is, hu14.18-IL2; and/or the RGD peptide comprises, or more preferably is, cilengitide or a pharmaceutically acceptable salt thereof.
  • both the humanized anti-GD2-IL-2 fusion protein and the anti-angiogenic RGD peptide are preferably administered to the patient as separate pharmaceutical compositions.
  • the humanized anti-GD2-IL-2 fusion protein and the anti-angiogenic RGD peptide, or the pharmaceutical compositions containing them, can be administered to the patient on separate days and/or both on the same day, whereby the administration on separate days and the administration of both on the same day can alternate.
  • the anti-angiogenic RGD peptide or the pharmaceutical composition containing it is preferably administered to the patient prior to the administration of the humanized anti-GD2-IL-2 fusion protein or the pharmaceutical composition containing it, preferably 30 minutes to 6 hours and more preferably 1 hour to 3 hours, e.g., about one hour, about two hours or about three hours, prior to the administration of the humanized anti-GD2-IL-2 fusion protein or the pharmaceutical composition containing it.
  • the anti-angiogenic RGD peptide or the pharmaceutical composition containing it is then administered to the patient over a time period of 15 minutes to 2 hours, more preferably 30 minutes to 90 minutes, e.g. over a time period of about 1 hour, for example by i.v. infusion, and then, about directly or alternatively with a pause, preferably a pause of 10 minutes to 90 minutes, more preferably 20 to 70 minutes, e.g., a pause of about one hour, the humanized anti-GD2-IL-2 fusion protein or the pharmaceutical composition containing it is administered.
  • the dosage regimens to be administered to the patient and the treatment regimens related thereto discussed herein preferably also apply here.
  • the patient can be an adult or a child.
  • the dosages to be administered to the patient and the treatment regimens related thereto in the treatment of neuroblastoma in children preferably comprise the following:
  • the RGD peptide (e.g., cilengitide) is administered in an amount of about 0.5 - 2.5 g/m 2 per day, more preferably about 1 - 2 g/m 2 per day, and even more preferably about 1.1 - 1.9 g/m 2 per day, e.g. in an amount of about 1.2 g/m 2 per day, of about 1.4 g/m 2 per day, or about 1.8 g/m 2 per day, on 1 to 3 days and preferably about 2 days, during solely one week of one cycle, preferably solely during the first week of the cycle.
  • the RGD peptide e.g., cilengitide
  • the RGD peptide (e.g., cilengitide) is administered in an amount of about 0.5 - 2.5 g/m 2 per day, more preferably about 1 - 2 g/m 2 per day, and even more preferably about 1.1 - 1.9 g/m 2 per day, e.g., in an amount of about 1.2 g/m 2 per day, of about 1.4 g/m 2 per day, or about 1.8 g/m 2 per day, on Day 1 and Day 3 during solely one week of one cycle, preferably on Day 1 and Day 3 solely during the first week of the cycle.
  • more than one such cycle e.g., 2 to 12, more preferably 2 to 8, and especially about 3 or about 6 of such cycles with respect to the RGD peptide are performed substantially without a pause.
  • the anti-GD2-IL-2 fusion protein (e.g., hu14.18-IL2) is administered in an amount of about 1 - 20 mg/m 2 per day, more preferably about 2 - 15 mg/m 2 per day, and even more preferably about 4 - 12 mg/m 2 per day, e.g., in an amount of about 4 mg/m 2 per day, of about 8 mg/m 2 per day, or about 12 mg/m 2 per day, on 2-5 consecutive days and preferably about 3 consecutive days within solely one week of one cycle, preferably solely during the first week of the cycle.
  • the anti-GD2- IL-2 fusion protein (e.g., hu14.18-IL2) is administered in an amount of about 4 mg/m 2 per day, of about 8 mg/m 2 per day, or about 12 mg/m 2 per day on 3 consecutive days during solely one week of one cycle, preferably solely during the first week of the cycle.
  • more than one such cycle e.g. 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 of such cycles with respect to the anti-GD2-IL-2 fusion protein are performed substantially without a pause. Even more preferably, more than one such cycle, e.g.
  • the dosages to be administered to the patient and the treatment regimens related thereto in the treatment of neuroblastoma in adults preferably comprise the following:
  • the amount of RGD peptide to be administered in the weekly administration can be the same or different in each week.
  • the dosages of the RGD peptide, preferably cilengitide are selected from the dosages or regimens (a) to (f) as described before and mixtures thereof and more preferably are selected from the dosages or regimens (a), (b), (d), (e) and (f); especially preferred in this respect is regimen or dosage (f).
  • more than one such cycle e.g., 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 of such cycles with respect to the RGD peptide are performed substantially without a pause.
  • the anti-GD2-IL-2 fusion protein (e.g., hu14.18-IL2) is administered in an amount of about 1 - 20 mg/m 2 per day, more preferably about 2 - 15 mg/m 2 per day, and even more preferably about 4 - 12 mg/m 2 per day, e.g., in an amount of about 4 mg/m 2 per day, of about 8 mg/m 2 per day, or about 12 mg/m 2 per day, on 2-5 consecutive days, and preferably about 3 consecutive days, during solely one week of one cycle, preferably solely during the first week of the cycle.
  • the anti-GD2-IL-2 fusion protein e.g., hu14.18-IL2
  • the anti-GD2-IL-2 fusion protein is administered in an amount of about 1 - 20 mg/m 2 per day, more preferably about 2 - 15 mg/m 2 per day, and even more preferably about 4 - 12 mg/m 2 per day, e.g., in an amount of about 4 mg/m 2
  • the anti-GD2- IL-2 fusion protein (e.g., hu14.18-IL2) is administered in an amount of about 4 mg/m 2 per day, of about 8 mg/m 2 per day, or about 12 mg/m 2 per day on 3 consecutive days during solely one week of one cycle, preferably solely during the first week of the cycle.
  • more than one such cycle e.g., 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 of such cycles with respect to the anti-GD2-IL-2 fusion protein are performed substantially without a pause.
  • more than one such cycle e.g. 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 of such cycles with respect to both the anti- GD2-IL-2 fusion protein and the RGD peptide are performed substantially without a pause, preferably substantially in parallel.
  • a preferred aspect of the instant invention is a method of treatment, wherein the above described use, medicaments and/or pharmaceutical compositions are applied to the patient as described with respect to said use, medicaments and/or pharmaceutical compositions, as described above.
  • Hu14.18-IL2 has preclinical activity in mouse models of neuroblastoma (NB), particularly when given to mice with a smaller tumor burden.
  • the anti-tumor activity of hu14.18-IL2 was also determined in two strata of human patients with recurrent or refractory NB.
  • hu14.18-IL2 was given intravenously at a dose of 12 mg/m 2 /day over 4 hours for three consecutive days every 28 days.
  • Responses were evaluated every 2 cycles for patients with disease measurable by standard radiographical criteria (stratum 1) and for patients with disease evaluable only by meta-iodobenzylguanidine (MIBG) imaging and/or bone marrow histology (stratum 2).
  • MIBG meta-iodobenzylguanidine
  • stratum 2 bone marrow histology
  • compositions used in a method of this invention comprise the active agent (i.e., the anti-angiogenic RGD peptide or the anti-GD2-IL-2 fusion protein) in a pharmaceutically acceptable carrier, as is well known, and therefore the invention is not to be construed as limited with regards to the composition so long as the concentration of the active agent(s) in the respective compositions is sufficient for delivery (administration) of the recited active agent in the amounts described herein. All aspects of the instant invention can be combined with other (further) treatment regimens known in the art of treating cancerous diseases in the broadest sense.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Mycology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The invention provides methods for treating malignant tumors and tumor metastases in a mammal comprising administering, to a mammal in need of treatment, a therapeutic amount of an anti-angiogenic RGD peptide (e.g., cilengitide) sufficient to inhibit angiogenesis in combination with a therapeutic amount of a humanized anti-GD2-IL-2 fusion protein (e.g., hu14.18-IL2). The present methods are particularly well suited for the treatment of melanoma and neuroblastoma.

Description

METHODS FOR TREATMENT OF MALIGNANCIES
Technical Field
The invention relates to methods for inhibition of malignancies, more preferably of primary malignancies and especially of primary disseminated malignancies, such as melanomas and neuroblastomas, and metastases thereof, using a therapy based on the combined administration of an anti- angiogenic agent and a targeted anti-tumor agent.
Background
The generation of new blood vessels, or angiogenesis, plays a key role in the growth of malignant disease and has generated much interest in developing agents that inhibit angiogenesis (see for example Holmgren, L., O'Reilly, M.S. & Folkman, J. (1995) "Dormancy of micrometastases: balanced proliferation and apoptosis in the presence of angiogenesis suppression", Nature Medicine 1 , 149-153; Folkman, J. (1995) "Angiogenesis in cancer, vascular, rheumatoid and other disease", Nature Medicine 1 , 27-31 ; O'Reilly, M.S., et a/., (1994) "Angiostatin: a novel angiogenesis inhibitor that mediates the suppression of metastases by a Lewis lung carcinoma", Cell 79, 315-328; Kerbel, R. S. (1997) "A cancer therapy resistant to resistance", Nature 390,335-336; Boehm, T., et al., (1997) "Antiangiogenic therapy of experimental cancer does not induce acquired drug resistance", Nature 390,404-7; and Volpert, O.V., et al., (1998) "A human fibrosarcoma inhibits systemic angiogenesis and the growth of experimental metastases via thrombospondin-1", Proc. Natl. Acad. Sci. (U.S.A.) 95, 6343-6348).
Angiogenesis is characterized by invasion, migration and proliferation of endothelial cells, processes that depend on cell interactions with extracellular matrix components. In this context, the endothelial adhesion receptor of integrin αvβ3 was shown to be a key player by providing a vasculature- specific target for anti-angiogenic treatment strategies. (Brooks, P. C, Clark, R.A. & Cheresh, D.A. (1994) "Requirement of vascular integrin alpha v beta 3 for angiogenesis", Science 264, 569-571 ; Friedlander, M., et al., (1995) "Definition of two angiogenic pathways by distinct alpha v integrins", Science 270, 1500-1502). The requirement for vascular integrin αvβ3 in angiogenesis was demonstrated by several in vivo models where the generation of new blood vessels by transplanted human tumors was entirely inhibited either by systemic administration of peptide antagonists of integrin αvβ3 or anti- αvβ3 antibody LM609. (Brooks, P. C1 et al., (1994) Science supra; Brooks, P. C, et al., (1994) "Integrin alpha v beta 3 antagonists promote tumor regression by inducing apoptosis of angiogenic blood vessels", Cell 79, 1157-1164). Murine hybridoma LM609 has been deposited with the American Type Culture Collection (ATCC, Manassas, VA, USA) as the International Depository Authority under the Budapest Treaty, and assigned the ATCC Designation HB 9537, on September 15, 1987. Such antagonists block the ligation of integrin αvβ3 which promotes apoptosis of the proliferative angiogenic vascular cells and thereby disrupt the maturation of newly forming blood vessels, an event essential for the proliferation of tumors.
A major obstacle for effective treatment of malignancies and/or disseminated malignancies includes minimal residual disease characterized by micrometastases that lack a well-established vascular supply for delivery of therapeutics. In this regard, a novel immunotherapeutic strategy proved efficient in using tumor compartment-specific monoclonal antibodies to direct cytokines to the tumor microenvironment. Combination therapies using antiangiogenic agents with other chemotherapeutic agents has been used to enhance treatment of various types of cancer, although the ultimate effectiveness of such combinations can be difficult to predict.
The chimeric anti-GD2-IL-2 fusion protein known as ch14.18-IL2 has been investigated as a treatment for melanoma, alone, and in combination with cilengitide, with some limited degree of success, however, ch14.18-IL2 is known to elicit undesirably high levels of human anti-chimeric antibody (HACA) responses and allergic reactions in humans. Summary of the Invention
The present invention is directed to a method for treating a malignant neoplasm, more preferably a disseminated malignant neoplasm such as melanoma or neuroblastoma in a patient. The method comprises administering to the patient in need of such treatment a malignant cell proliferation inhibiting amount of an anti-angiogenic RGD peptide and a humanized anti-GD2 antibody-interleukin-2 (IL-2) fusion protein, preferably a fusion protein of IL-2 with the humanized anti-GD2 antibody designated as hu14.18. The hu14.18 antibody and the IL-2 fusion protein thereof (hu14.18- IL2) are described in detail in co-owned U.S. Patent No. 7,169,904 (Gillies et a/.), which is incorporated herein by reference in its entirety. Inhibition of malignant cell proliferation can encompass inhibition of the growth of malignant cells such as neuroblastoma cells, melanoma cells, and the like in an existing tumor or in tumor metastases, inhibition of the formation of additional tumor metastases, and even tumor cell death. The anti-angiogenic RGD peptide and the humanized anti-GD2-IL-2 fusion protein can be administered substantially concurrently, as well as sequentially.
The aforementioned therapeutic compositions (i.e., the anti-angiogenic RGD peptide and the humanized anti-GD2-IL-2 fusion protein) can be administered to the patient prior to, during or after surgical intervention to remove all or part of a tumor, if desired. Administration can be accomplished via direct immersion; systemic or localized intravenous (i.v.), intraperitoneal (i p ), subcutaneous (s.c), intramuscular (i.m.), or direct injection into a tumor mass; and/or by oral administration of the appropriate formulations.
Anti-angiogenic RGD peptides suitable for use in the methods of the present invention include linear and cyclic polypeptides containing the Arg-Gly-Asp (RGD) integrin binding motif, which peptides bind to the αvβ3 and/or αvβs receptor and inhibit angiogenesis. Anti-angiogenic RGD peptides are described in U.S. Patent No. 5,262,520 (Plow et al.) and WO 97/45137. A particularly preferred RGD peptide is cilengitide, i.e., cyclo-(Arg-Gly-Asp-D-Phe-N Me-VaI), SEQ ID NO: 1.
A preferred humanized anti-GD2-IL-2 fusion protein comprises an immunoglobulin (Ig) polypeptide that specifically binds to the GD2 antigen, bound to IL-2, either directly or through a linker peptide. Preferably, the antibody fusion protein comprises two Ig polypeptide chains with a IL-2 polypeptide fused to each chain. Minimally, the humanized anti-GD2-IL-2 fusion protein comprises a GD2-binding antibody variable domain (e.g., at least one light chain variable region peptide and at least one heavy-chain variable region peptide that includes complementarity determining regions (CDRs) that specifically bind to GD2).
The GD2-targeting Ig portion of the fusion protein can comprise an entire immunoglobulin chain amino acid sequence, or at least the fragment thereof that comprises the GD2 antigen binding specificity portion of the protein. In addition, the GD2-binding portion of the fusion protein is humanized to reduce the potential for adverse immunological effects when administering the fusion protein to a human subject.
Brief Description of the Drawings
FIG. 1 shows the amino acid sequence of the light chain (SEQ ID NO: 2) of hu14.18-IL2. FIG. 1 shows the amino acid sequence of the heavy chain (SEQ ID NO: 3) of hu14.18-IL2, the C-terminal region of which includes the IL-2 amino acid sequence.
Detailed Description of the Invention The suppression and eradication of primary tumors and distant metastases is a major goal of alternative treatment strategies for cancer, such as inhibition of angiogenesis and targeted immunotherapy. The methods of the invention utilize the combined administration of at least one angiogenesis inhibiting RGD peptide with at least one, humanized anti-GD2-IL-2 fusion protein in a therapeutic amount sufficient to inhibit proliferation of melanoma cells, neuroblastoma cells, and the like.
A variety of therapeutic compositions are described which are useful in practicing the methods of the invention.
A preferred RGD peptide for use in the present invention is the cyclic pentapeptide known as cilengitide, i.e., cyclo-(Arg-Gly-Asp-D-Phe-NMe-Val), SEQ ID NO: 1 , which can be administered as a pharmaceutically acceptable salt thereof, e.g., a hydrochloride salt, or as the inner salt, which is cyclo- (Arg-Gly-Asp-D-Phe-NMe-Val), SEQ ID NO: 1 , perse.
Since the advent of methods for generating monoclonal antibodies first published by Kohler and Milstein, improved such methods have become well known in the art. (See for example, Dean, CJ. in Methods in Molecular Biology Vol. ^.Immunochemical Protocols, 2nd ed. (Pound, J. D. editor, Humana Press, Inc., Totowa NJ, 1998) chapter 4; and Ausbel, F. M. et al., Short Protocols in Molecular Biology, 2nd ed. (Current Protocols, John Wiley & Sons, NY NY, 1992) chapter 11). It is now routine practice to generate a monoclonal antibody (an Ig) which binds to a particular antigen. Screening protocols have also improved for the selection of high-affinity binding antibodies, if desired.
Typically the host cells for production of hybridomas are mouse-derived, or are derived from other rodents.
One barrier to repeated treatment with murine monoclonals to human patients is the HAMA (human anti-mouse antibodies) response generated by the patient in response to the treatment. Methods to overcome this barrier include humanization of murine antibody proteins by substituting antigenic amino acids of the mouse protein with human protein sequences that are assumed to be less antigenic. Other methods involve the grafting of the binding specificity determining amino acid residues or regions into human protein frameworks.
The ability to express an antibody protein in phage display systems allows for selection of antibodies that have been subjected to mutagenesis to either improve binding or to reduce immunogenicity (See for example Antibody Engineering (McCafferty, J. et al. editors, Oxford University Press, 1996). Recently, the substitution of human immunoglobulin genes into transgenic mice has allowed the use of murine hosts to generate antibodies, and thus monoclonal antibodies, that are of human nucleic acid sequence origin.
Antibodies can also be reduced in size by fragmentation to allow for reduced antigenicity, or to create smaller therapeutic molecules. For example, a whole antibody protein can be reduced, either by digestion with the appropriate enzymes or by producing the smaller protein by recombinant DNA methods. Suitable fragments include at least an antigen binding portion of the whole molecule, and can comprise Fab, F(ab)2, F(ab)3, Fv or single chain Fv (single chain antibody; SCA) constructs.
Al! monoclonal antibody-based components of the therapeutic agents for use in the methods of the invention are humanized to reduce potential allergic reactions and HACA responses when administered to humans.
As used herein, the term "anti-GD2-IL2 fusion protein" encompasses an anti- GD2 Ig polypeptide or the antigen-binding portion thereof, bound to IL-2. The IL-2 can be directly bound to the Ig polypeptide or can be bound thereto via a linker peptide.
Anti-tumor antigen/cytokine fusion proteins are known in the art, and in particular are described, for example, in U.S. Patent No. 5,650,150 (Gillies), and particularly in U.S. Patent No. 7,169,904 (Gillies et a/.), which describes hu14.18-IL2, the disclosures of which relating to the preparation and use of fusion proteins are expressly incorporated herein by reference. Hu14.18-IL2 comprises the immunoglobulin light chain sequence of SEQ ID NO: 2 and the immunoglobulin heavy chain sequence of SEQ ID NO: 3 (see FIG. 1 and
FIG. 2, respectively).
Typically, the Ig polypeptide chain is an Ig heavy chain polypeptide, a light chain polypeptide, or both, which comprise an N-terminal variable region specific for the GD2 antigen. The fusion protein typically has the Ig polypeptide joined at its carboxy-terminus by a peptide bond to the amino terminus of IL-2. Where the Ig polypeptide is a heavy chain, the Ig heavy chain further typically comprises CH1 and CH2 domains, and may optionally further contain a CH3 domain. However, if desired, such constant region domains can be eliminated to reduce immunogenicity, size, or non-specific binding of the resultant fusion protein construct. In order to facilitate heavy and light chain domain association, it may be desired to construct a linked Fv molecule, wherein a heavy chain Fv is tethered to a light chain Fv. The tether is between the carboxy end of one chain to the amino terminal of the other, and is long enough so as not to drastically sterically alter the antigen binding pocket after refolding/domain association. Preferably, the humanized anti- GD2-IL-2 fusion protein comprises a complete antibody with one IL-2 peptide attached to the carboxy-terminus of each Ig polypeptide chain of the antibody.
Therapeutic methods of the present invention for treating malignant tumor cells in tumors and tumor metastases are based on the combined use of an anti-angiogenic RGD peptide and a humanized anti-GD2-IL-2 fusion protein. The combined use can be performed simultaneously, sequentially, or with the intervention of a period of time between the treatments. Any of the specific therapeutics may be administered more than once during a course of treatment. The method of the invention provides a synergistic potentiation of the tumor cell proliferation inhibition effect of each individual therapeutic, yielding more effective treatment than found by administering an individual component alone.
The method of the invention comprises a variety of modalities for practicing the invention in terms of the steps. For example, the RGD peptide and the humanized anti-GD2-IL-2 fusion protein can be administered following admixture, i.e., simultaneously, or can be administered sequentially, i.e., separately. Furthermore, the RGD peptide and the fusion protein can be separately administered within a time interval of about 3 weeks between administrations, i.e., from substantially immediately after the first active agent is administered to up to about 3 weeks after the first agent is administered. Additionally, the order of administration can be varied, i.e., that the RGD peptide can be administered prior to administration of the fusion protein, or that administration can be conducted in the reverse order.
The term "therapeutically effective" or "therapeutically effective amount" refers to an amount of a drug effective to treat a disease or disorder in a mammal. In the case of a malignancy, e.g., cancer, the therapeutically effective amount of a therapeutic agent may reduce the number of malignant cells; reduce the tumor size; inhibit (i.e., slow to some extent and preferably stop) malignant cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve to some extent one or more of the symptoms associated with the cancer. To the extent the drug may prevent growth and/or kill existing cancer cells, it may be cytostatic and/or cytotoxic. For cancer therapy, efficacy can, for example, be measured by assessing the time to disease progression (TTP) and/or determining the response rate (RR).
In one embodiment, the method of the invention also encompasses administering to a patient suffering from a melanoma, a neuroblastoma, or the metastases thereof, an angiogenesis inhibiting amount of the anti- angiogenic RGD peptide, and an amount of humanized anti-GD2-IL-2 fusion protein sufficient to elicit a biological response.
Typically, a therapeutically effective amount of a humanized anti-GD2-IL-2 fusion protein comprising a complete antibody fused to IL-2, is an amount such that when administered in physiologically tolerable composition is sufficient to achieve a plasma concentration of about 0.01 microgram (μg) per milliliter (ml) to about 100 μg/ml, preferably about 1 μg/ml to about 5 μg/ml and usually about 5 μg/ml based on the antibody portion of the fusion protein. Stated differently, the dosage can vary from about 0.1 mg/kg to about 300 mg/kg, preferably about 0.2 mg/kg to about 200 mg/kg, most preferably about 0.5 mg/kg to about 20 mg/kg, in one or more dose administrations daily, for one or several days, again based on the antibody portion of the fusion protein. Where the Ig polypeptide portion of the fusion protein is in the form of a fragment of a monoclonal antibody or a conjugate, the amount can readily be adjusted based on the mass of the fragment / conjugate relative to the mass of the complete antibody. A preferred plasma concentration, expressed as molarity, is about 2 micromolar (μM) to about 5 millimolar (mM), and preferably, about 100 μM to about 1 mM of the fusion protein.
A therapeutically effective amount of an RGD peptide according of this invention typically is an amount of RGD peptide that, when administered in a physiologically tolerable composition, is sufficient to achieve a plasma concentration of from about 0.1 microgram (μg) per milliliter (ml) to about 200 μg/ml, preferably from about 1 μg/ml to about 150 μg/ml. Based on a polypeptide having a molecular mass of about 500 grams per mole, the preferred plasma concentration, expressed as molarity, is about 2 micromolar (μM) to about 5 millimolar (mM), and preferably about 100 μM to 1 mM of the RGD peptide. One of ordinary skill in the art can readily adjust the amount administered based on the actual molecular mass of the particular RGD peptide being used. Cilengitide has a molecular mass of about 589 g/mol.
The preferred dosage of an active agent, e.g., the RGD peptide or anti-GD2- IL-2 fusion protein, for administration to a patient, is about 0.5 mg to about
3000 mg per patient and day, more preferably about 10 to about 2500 mg per patient and per day, and especially about 50 to about 1000 mg per patient and per day. On a per kilogram body weight basis, the preferred active agent dosage level, preferably is about 0.1 to about 100 mg/kg, and more preferably about 1 mg to about 50 mg/kg, preferably per dosage unit and more preferably per day. On a per square meter of the body surface area basis, the preferred dosage level of an active agent is about 0.5 mg to about 2000 mg/m2, more preferably about 5 to about 1500 mg/m2' and especially about 50 to about 1000 mg/m2, preferably per dosage unit and more preferably per day.
Alternatively, the preferred dosage of the RGD peptide for administration to a patient is about 500 mg, about 1000 mg or about 2000 mg per patient and day, preferably if given "flat", e.g. as a flat dosing, which preferably means that the amount of RGD peptide given is not adjusted to the body weight or surface area of the respective patient. The afore described dosage of about 500 mg, about 1000 mg or about 2000 mg per patient and day (flat) is typically administered at intervals ranging from once a month to once a day, more preferably ranging from once a week to five times a week and especially about once a week, about twice a week or about five times a week. Preferably, this dosing is applied to a patient for two or more consecutive weeks, more preferably four or more consecutive weeks. However, this dosing can be applied to the patient until healing, stable disease or progression of the patient occurs. For example, this dosing can be applied to a patient for up to 52 weeks, up to 104 weeks, up to 156 weeks or even longer. The treatment of malignancies, at least the treatment of malignancies with cancer, at least the treatment of cancer with therapeutic agents in the broadest sense, is a protracted issue. Thus, the treatment of cancer with therapeutic agents generally includes a prolonged exposure to one or more respective therapeutic agents. Taking into account that most of the therapeutic agents, when administered in an efficient dose, are toxic for the body of the patient, the therapeutic agents (unless they show any or hardly any acute toxicity) are generally administered over a certain, limited time, followed by a time period without the administration of the respective therapeutic agent, during which time the patient's body is allowed to recover from the toxicity of said therapeutic agent. Generally, a treatment regimen comprises the administration time period of the respective therapeutic agent and the recovery time period after the administration of the respective therapeutic agent is repeated one or more times, preferably several times. Each repeat of the course of treatment is usually referred to by the skilled artisan as a "cycle" (or "treatment course").
Each cycle comprises the administration time period of the respective therapeutic agent and the recovery time period after the administration of the respective therapeutic agent. The duration of the administration time period and/or the recovery time period after the administration of the therapeutic usually depend on the properties of the respective therapeutic agent. Accordingly, different therapeutic agents can have different durations of the administration time period and/or the recovery time period after the therapeutic agent is administered. Thus, the length or duration of a cycle can be different for different therapeutic agents.
Generally, the length of a cycle is in the range of about one week to about 12 weeks, more preferably one week to six weeks, and especially 2 to 4 weeks. Preferably, the dosing of the respective therapeutic agent is given in an amount per cycle, allowing a medical practitioner to adapt the actual administration regimen to the medical status of the patient, i.e., the amount of a particular agent administered per cycle can be a single administration or can be divided into two or more portions administered at different times within the cycle, if desired. In the setting of a combination treatment comprising two or more therapeutic agents, as in the present invention, generally two or more cycles, one for each agent, having the same or a different length, run in parallel. If a therapeutic agent is administered to the patient in two or more portions within one cycle, each portion is preferably given on a different day within said cycle. With respect to each of the therapeutic agents administered, generally more than one cycle, preferably two or more cycles, even more preferably three or more cycles are applied to the patient, preferably substantially with out a pause. Generally, not more than 24 cycles are applied to the patient substantially without a pause. The administration of about six cycles substantially without a pause to the patient for each of the therapeutic agents administered is generally a standard.
RGD peptides that are useful in the compositions and methods of the present invention include anti-angiogenic cyclic RGD peptides, such as cilengitide, anti-angiogenic linear RGD peptides, as well as pharmaceutically acceptable derivatives, solvates, and/or salts thereof of such peptides. In some preferred embodiments, the RGD peptide is administered to a patient in an amount of about 250 mg to about 12500 mg per week, preferably for one or more weeks of a cycle/treatment course.
One cycle/treatment course according to this invention preferably consists of 21 to 35 sequential days, and more preferably of about 28 days or about 4 weeks.
Generally, the RGD peptide, preferably cilengitide, can be administered to the patient by any method or mode or administration known in the art. For example, the RGD peptide can be administered to the patient in an amount of about 250 mg to about 12500 mg, more preferably about 450 to about 10500 mg, within a time period of about one week. This is also referred to as the weekly administration with respect to the RGD peptide. Preferably, a weekly administration of the given amounts of RGD peptide takes place once or twice or more times, preferably once, twice, or three times, within a time period of about three weeks. Preferably, a weekly administration of the given amounts of RGD peptide takes place once or twice, preferably once, within a time period of about four weeks.
In some preferred embodiments, the weekly administration of the RGD peptide takes place during two or more weeks within the cycle or the cycles of administration of the anti-GD2-IL-2 fusion protein. In another preferred embodiment, the weekly administration of the RGD peptide takes place during every week within the cycle or the cycles of administration of the anti- GD2-IL-2 fusion protein. In another more preferred embodiment, the weekly administration of the RGD peptide takes place during one week within the cycle or cycles of administration of the anti-GD2-IL-2 fusion protein.
Preferably, the weekly administration of the RGD peptide takes place once in the cycle or cycles of administration of the anti-GD2-IL-2 fusion protein, and more preferably within one week in which the anti-GD2-IL-2 fusion protein is also administered.
The amount of RGD peptide to be administered in the weekly administration can be the same or different in each week. The following dosages or regimens are preferred in this respect: (a) the RGD peptide, preferably cilengitide, is administered to the patient in an amount of about 500 mg to about 2000 mg in a single dose each week during the cycle of administration of the anti-GD2-IL-2 fusion protein; (b) the RGD peptide, preferably cilengitide, is administered to the patient in an amount of about 500 mg to about 2000 mg twice a week each week during the cycle of administration of the anti-GD2-IL-2 fusion protein; (c) the RGD peptide, preferably cilengitide, is administered to the patient in an amount of about 500 mg each day on five consecutive days within one first week and in an amount of about 500 mg on one day within each additional week during the cycle of administration of the anti-GD2-IL-2 fusion protein; (d) the RGD peptide, preferably cilengitide, is administered to the patient in an amount of about 2000 mg each day on three consecutive days within one first week and in an amount of about 2000 mg on one day within each further week during the cycle of administration of the anti-GD2-IL-2 fusion protein;(e) the RGD peptide, preferably cilengitide, is administered to the patient in an amount of about 2000 mg once a week each week during the cycle of administration of the anti-GD2-IL-2 fusion protein; or (f) the RGD peptide, preferably cilengitide, is administered to the patient on two different days, in an amount of about 2000 mg on each of the two days, within the first week during the cycle of administration of the anti-GD2-IL-2 fusion protein.
Preferably, more than one cycle of the RGD peptide is administered to the patient. More preferably 2 to 12 cycles, and especially about 6 cycles are administered to the patient, preferably comprising one or more of regimens (a) to (f) discussed above. In one preferred embodiment, the more than one cycle of RGD peptide administration comprises only one of the regimens selected from (a) to (f), i.e., the same regimen selected from (a) to (f) is applied to the patient in each cycle of administration. More preferably, the same regimen selected from (a) to (f) is administered to the patient in each of about 6 cycles.
Alternatively, the more than one cycles of RGD peptide administration comprises two or more of the regimens selected from (a) to (f) above, i.e., in different cycles, a different regimen is selected from (a) to (f) for administration to the patient.
In some preferred embodiments, in cases wherein more than one cycle of the anti-GD2-IL-2 fusion protein is applied to the patient, combinations of one or more of regimens (a) to (f) are also administered to the patient. For example, regimen (c) can be applied to the patient for the first cycle, followed by regimen (a) for 1 to 11 cycles, and especially about 5 cycles. Preferably, during the regimen (a), the weekly administration of the RGD peptide consists of about 500 mg. Alternatively, regimen (d) can be applied to the patient for the first cycle, followed by regimen (a) for 1 to 11 cycles, and especially about 5 cycles.
Preferably, during the regimen (a), the weekly administration of the RGD peptide consists of about 2000 mg. Preferably, the amount of RGD peptide to be administered in the weekly administration can be the same or different in each week. More preferably, the dosages of the RGD peptide, preferably cilengitide, are selected from the dosages or regimen (a) to (f) as described before and mixtures thereof and more preferably are selected from the dosages or regimen (a), (b), (d), (e) and (T); especially preferred in this respect is regimen or dosing (f). Also especially preferably, more than one such cycle, e.g., 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 of such cycles with respect to the RGD peptide are performed substantially without a pause.
Preferably, the duration of one RGD peptide administration cycle, preferably each cycle, is about three weeks (about 21 days) or about four weeks (about 28 days), more preferably about four weeks (about 28 days). However, due to the extremely low toxicity of the cilengitide, the pharmaceutically acceptable derivatives, solvates, and/or salts thereof, this RGD peptide also can be administered to the patient outside the administration cycles of the anti-GD2-IL-2 fusion protein. This is especially advantageous as a maintenance therapy consisting of or comprising, preferably consisting of, the administration of cilengitide, the pharmaceutically acceptable derivatives, solvates and/or salts thereof, for one or several months, for example, for up to about 24 months, even substantially without pause.
Due to different pharmacokinetics in children, a dosage of both the humanized anti-GD2-IL-2 fusion protein and the RGD peptide preferably is based on an amount per square meter of body surface area. Preferably, the anti-GD2-IL-2 fusion protein (e.g., hu14.18-IL2) is administered in an amount of about 1 - 100 mg/m2, more preferably about 2 - 80 mg/m2, and even more preferably about 3 - 60 mg/m2, e.g. in an amount of about 12 mg/m2, of about 24 mg/m2, or about 36 mg/m2, per week, for one or more weeks during one cycle, preferably one or two weeks during one cycle and more preferably for one week during one cycle. More preferably, the anti-GD2-IL-2 fusion protein (e.g., hu14.18-IL2) is administered in an amount of about 1 - 100 mg/m2, more preferably about 2 - 80 mg/m2, and even more preferably about 3 - 60 mg/m2, e.g. in an amount of about 12 mg/m2, of about 24 mg/m2, or about 36 mg/m2, per week solely in the first week during one cycle.
Preferably, the amounts of anti-GD2-IL-2 fusion protein (e.g., hu14.18-IL2) given above are administered in 1 to 5, more preferably about 3 approximately equal portions, each portion being given on a separate day, preferably each portion being given on a separate, but consecutive day. Even more preferably, the anti-GD2-IL-2 fusion protein (e.g., hu14.18-IL2) is administered in an amount of about 1 - 20 mg/m2 per day, more preferably about 2 - 15 mg/m2 per day, and even more preferably about 4 - 12 mg/m2 per day, e.g. in an amount of about 4 mg/m2 per day, of about 8 mg/m2 per day, or about 12 mg/m2 per day, on 2-5 consecutive days and preferably about 3 consecutive days within one week during solely one week of one cycle, preferably solely during the first week of the cycle. Especially preferably, the anti-GD2-IL-2 fusion protein (e.g., hu14.18-IL2) is administered in an amount of about 4 mg/m2 per day, of about 8 mg/m2 per day, or about 12 mg/m2 per day on 3 consecutive days within one week during solely one week of one cycle, preferably solely during the first week of the cycle. Preferably, more than one such cycle, e.g., 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 such cycles are performed substantially without a pause.
For the treatment of melanoma, preferably the RGD peptide (e.g., cilengitide) is administered in an amount of about 0.5 - 2.5 g/m2 per day, more preferably about 1 - 2 g/m2 per day, and even more preferably about 1.1 - 1.9 g/m2 per day, e.g. in an amount of about 1.2 g/m2 per day, of about 1.4 g/m2 per day, or about 1.8 g/m2 per day, on 1 to 3 days and preferably about 2 days within one week during solely one week of one cycle, preferably solely during the first week of the cycle. Especially preferably, the RGD peptide (e.g., cilengitide) is administered in an amount of about 0.5 - 2.5 g/m2 per day, more preferably about 1 - 2 g/m2 per day, and even more preferably about 1.1 - 1.9 g/m2 per day, e.g. in an amount of about 1.2 g/m2 per day, of about 1.4 g/m2 per day, or about 1.8 g/m2 per day, on Day 1 and Day 3 within one week, during solely one week of one cycle, preferably on Day 1 and Day 3 solely during the first week of the cycle. Preferably, more than one such cycle, e.g., 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 such cycles are performed substantially without a pause. Even more preferably, more than one such cycle, e.g. 2 to 12, more preferably 2 to 8 and especially about 3 or about 6 of such cycles with respect to both the anti-GD2-IL-2 fusion protein and the RGD peptide are performed substantially without a pause, preferably substantially in parallel.
Preferably, the amounts of the RGD peptide (e.g., cilengitide) given above are administered in 1 to 5, more preferably into about 2 or about 3, preferably approximately equal portions, each portion given on a separate day, preferably each given on a separate day within one week.
In another embodiment, the invention can be practiced in conjunction with surgical procedures where portions or all of a tumor mass has been removed. In this regard, the method can be practiced following a surgical procedure. Alternatively, the surgical procedure can be practiced during the interval between administration of the first active agent and the second active agent. Exemplary of this method is the combination of the present method with surgical tumor removal described below. Administration can be accomplished by periodic unit dosages, by continuous infusion, peristaltic delivery, by bolus injection, and the like. Routes can include intravenous, subcutaneous, intramuscular, orthotopic injection, orthotopic infusion, oral application, and the like.
Another aspect of treating melanoma is the use of an anti-angiogenic RGD peptide for the manufacture of a medicament for the treatment of melanomas, wherein the medicament is to be applied in combination with a humanized anti-GD2-IL-2 fusion protein. According to this aspect of the invention, the humanized anti-GD2-IL-2 fusion protein preferably comprises, or more preferably is, hu14.18-IL2; and/or the RGD peptide comprises, or more preferably is, cilengitide or a pharmaceutically acceptable salt thereof.
According to this aspect of the invention, both the humanized anti-GD2-IL-2 fusion protein and the anti-angiogenic RGD peptide are preferably administered to the patient as separate pharmaceutical compositions. The humanized anti-GD2-IL-2 fusion protein and the anti-angiogenic RGD peptide, or the pharmaceutical compositions containing them, can be administered to the patient on separate days and/or both on the same day, whereby the administration on separate days and the administration of both on the same day can alternate. On the days where both the humanized anti- GD2-IL-2 fusion protein and the anti-angiogenic RGD peptide, or the pharmaceutical compositions containing them, are administered to the patient on the same day, the anti-angiogenic RGD peptide or the pharmaceutical composition containing it is preferably administered to the patient prior to the administration of the humanized anti-GD2-IL-2 fusion protein or the pharmaceutical composition containing it, preferably 30 minutes to 6 hours and more preferably 1 hour to 3 hours, e.g., about one hour, about two hours or about three hours, prior to the administration of the humanized anti-GD2-IL-2 fusion protein or the pharmaceutical composition containing it. Typically, the anti-angiogenic RGD peptide or the pharmaceutical composition containing it is then administered to the patient over a time period of 15 minutes to 2 hours, more preferably 30 minutes to 90 minutes, e.g., over a time period of about 1 hour, for example by i.v. infusion, and then, about directly or alternatively with a pause, preferably a pause of 10 minutes to 90 minutes, more preferably 20 to 70 minutes, e.g., a pause of about one hour, the humanized anti-GD2-IL-2 fusion protein or the pharmaceutical composition containing it is administered.
According to this aspect of the invention, the dosage regimens to be administered to the patient and the treatment regimens related thereto discussed herein preferably also apply here. The patient can be an adult or a child.
According to this aspect of the invention, the dosages to be administered to the patient and the treatment regimens related thereto in the treatment of melanoma in children preferably comprise the following:
Preferably, the RGD peptide (e.g., cilengitide) is administered in an amount of about 0.5 - 2.5 g/m2 per day, more preferably about 1 - 2 g/m2 per day, and even more preferably about 1.1 - 1.9 g/m2 per day, e.g., in an amount of about 1.2 g/m2 per day, of about 1.4 g/m2 per day, or about 1.8 g/m2 per day, on 1 to 3 days and preferably about 2 days, during solely one week of one cycle, preferably solely during the first week of the cycle. Especially preferably, the RGD peptide (e.g., cilengitide) is administered in an amount of about 0.5 - 2.5 g/m2 per day, more preferably about 1 - 2 g/m2 per day, and even more preferably about 1.1 - 1.9 g/m2 per day, e.g., in an amount of about 1.2 g/m2 per day, of about 1.4 g/m2 per day, or about 1.8 g/m2 per day, on Day 1 and Day 3 during solely one week of one cycle, preferably on Day 1 and Day 3 solely during the first week of the cycle. Also especially preferably, more than one such cycle, e.g. 2 to 12, more preferably 2 to 8, and especially about 3 or about 6 of such cycles with respect to the RGD peptide are performed substantially without a pause. Preferably, the anti-GD2-IL-2 fusion protein (e.g., hu14.18-IL2) is administered in an amount of about 1 - 20 mg/m2 per day, more preferably about 2 - 15 mg/m2 per day, and even more preferably about 4 - 12 mg/m2 per day, e.g., in an amount of about 4 mg/m2 per day, of about 8 mg/m2 per day, or about 12 mg/m2 per day, on 2-5 consecutive days and preferably about 3 consecutive days within solely one week of one cycle, preferably solely during the first week of the cycle. Especially preferably, the anti-GD2- IL-2 fusion protein (e.g., hu14.18-IL2) is administered in an amount of about 4 mg/m2 per day, of about 8 mg/m2 per day, or about 12 mg/m2 per day on 3 consecutive days during solely one week of one cycle, preferably solely during the first week of the cycle. Also especially preferably, more than one such cycle, e.g., 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 of such cycles with respect to the anti-GD2-IL-2 fusion protein are performed substantially without a pause. Even more preferably, more than one such cycle, e.g., 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 of such cycles with respect to both the anti- GD2-IL-2 fusion protein and the RGD peptide are performed substantially without a pause, preferably substantially in parallel.
According to this aspect of the invention, the dosages to be administered to the patient and the treatment regimens related thereto in the treatment of melanoma in adults preferably comprise the following:
Preferably, the amount of RGD peptide to be administered in the weekly administration can be the same or different in each week. More preferably, the dosages of the RGD peptide, preferably cilengitide, are selected from the dosages or regimens (a) to (f) as described before and mixtures thereof and more preferably are selected from the dosages or regimens (a), (b), (d), (e) and (f); especially preferred in this respect is regimen or dosage (f).
Also especially preferably, more than one such cycle, e.g. 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 of such cycles with respect to the RGD peptide are performed substantially without a pause.
Preferably, the anti-GD2-IL-2 fusion protein (e.g., hu14.18-IL2) is administered in an amount of about 1 - 20 mg/m2 per day, more preferably about 2 - 15 mg/m2 per day, and even more preferably about 4 - 12 mg/m2 per day, e.g. in an amount of about 4 mg/m2 per day, of about 8 mg/m2 per day, or about 12 mg/m2 per day, on 2-5 consecutive days, and preferably about 3 consecutive days, during solely one week of one cycle, preferably solely during the first week of the cycle. Especially preferably, the anti-GD2- IL-2 fusion protein (e.g., hu14.18-IL2) is administered in an amount of about 4 mg/m2 per day, of about 8 mg/m2 per day, or about 12 mg/m2 per day on 3 consecutive days during solely one week of one cycle, preferably solely during the first week of the cycle. Also especially preferably, more than one such cycle, e.g., 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 of such cycles with respect to the anti-GD2-IL-2 fusion protein are performed substantially without a pause. Even more preferably, more than one such cycle, e.g., 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 of such cycles with respect to both the anti- GD2-IL-2 fusion protein and the RGD peptide are performed substantially without a pause, preferably substantially in parallel.
One preferred aspect of the instant invention is a method of the treatment of melanoma wherein the above described use, medicaments and/or pharmaceutical compositions are applied to the patient as described with respect to said use, medicaments and/or pharmaceutical compositions, as described above. Human experience with hu14.18-IL2
An initial Phase I protocol for hu14.18-IL2 was performed. A total of 33 subjects with melanoma were enrolled to establish the maximum tolerated dose (MTD). Subjects were enrolled in cohorts of 3 or 6 subjects and administered hu14.18-IL2 at one of the following dose levels: 0.8, 1.6, 3.2, 4.8, 6.0 or 7.5 mg/m2/day. Hu14.18-IL2 was administered as a 4-hour i.v. infusion over 3 consecutive days during the first week of each course. Subjects with tumor regression or stable disease were given a second course of treatment with hu14.18-IL2 four weeks after Course 1. Thirty-one subjects completed one course of treatment with hu14.18-IL2, and 19 subjects had stable disease following Course 1 and proceeded to receive a second course of treatment. The dose of 7.5 mg/m2/day was found to be the MTD, as 2 of 6 subjects showed reversible dose-limiting toxicity at this dose level.
Overall, hu14.18-IL2 was well tolerated, with most subjects experiencing mild to moderate adverse events (AEs)1 mainly known IL-2 side effects. Laboratory analyses showed lymphocytosis, expansion of CD16+ and CD56+ NK cells, marked peripheral lymphocyte activation (evidenced by a dose-dependent elevation of circulating slL2R alpha chain), and enhanced ability of peripheral blood mononuclear cells (PBMC) to mediate GD2+ tumor cell killing through ADCC. Thus hu14.18-IL2 has demonstrated biological activities. PK results indicate the serum t1/2 of hu14.18-IL2 to be 3.7 hours.
In a clinical treatment setting, the following exemplary protocol set forth in Table 1 can be followed.
Table 1.
Figure imgf000024_0001
Notes for Table 1 :
1. All subjects preferably are scheduled to receive at least 3 courses of treatment and more preferably a total of 3 courses of treatment. Treatment can be stopped if it can not be tolerated or if there is recurrent disease.
2. Hu14.18-IL2 is administered as a 4 hour i.v. infusion on Days 1 , 2, and 3 in the first week of each treatment cycle. Each treatment cycle is 4 weeks (+/- 7 days). Subjects in this study will be administered 6 mg/m2/d of hu14.18-IL2, a dose corresponding to one dose below the maximal tolerated dose (MTD).
3. Cilengitide therapy is administered on Days 1 and 3 of the 28-day treatment course as a 1-hour i.v. infusion at a fixed dose of 2000 mg/dose. Each treatment cycle is 4 weeks (+/- 7 days).
The therapeutic compositions used in a method of this invention comprise the active agent (i.e., the anti-angiogenic RGD peptide or the anti-GD2-IL-2 fusion protein) in a pharmaceutically acceptable carrier, as is well known, and therefore the invention is not to be construed as limited with regards to the composition so long as the concentration of the active agent(s) in the composition is sufficient for delivery (administration) of the recited active agent in the amounts described herein. For the treatment of neuroblastoma, preferably the RGD peptide (e.g., cilengitide) is administered in an amount of about 1 g - 8 g/m2, more preferably about 1.5 g - 6 g/m2, and even more preferably about 2 g - 4 g/m2, e.g. in an amount of about 2.2 g/m2, 2.4 g/m2, of about 2.8 g/m2, or about 3.6 g/m2, per week for one or more weeks during one cycle, preferably one, two, three or four weeks during one cycle, more preferably one or two weeks during one cycle and even more preferably for one week during one cycle. More preferably, the RGD peptide (e.g., cilengitide) is administered in an amount of about 1 g - 8 g/m2, more preferably about 1.5 g - 6 g/m2, and even more preferably about 2 g - 4 g/m2, e.g. in an amount of about 2.2 g/m2, 2.4 g/m2, about 2.8 g/m2, or about 3.6 g/m2, per week solely in the first week during one cycle. Preferably, more than one such cycle, e.g., 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 such cycles are performed substantially without a pause.
Preferably, the amounts of the RGD peptide (e.g., cilengitide) given above are administered in 1 to 5, more preferably into about 2 or about 3, preferably approximately equal portions, each portion given on a separate day, preferably each given on a separate day within one week.
Even more preferably, the RGD peptide (e.g., cilengitide) is administered in an amount of about 0.5 - 2.5 g/m2 per day, more preferably about 1 - 2 g/m2 per day, and even more preferably about 1.1 - 1.9 g/m2 per day, e.g. in an amount of about 1.2 g/m2 per day, of about 1.4 g/m2 per day, or about 1.8 g/m2 per day, on 1 to 3 days and preferably about 2 days within one week during solely one week of one cycle, preferably solely during the first week of the cycle. Especially preferably, the RGD peptide (e.g., cilengitide) is administered in an amount of about 0.5 - 2.5 g/m2 per day, more preferably about 1 - 2 g/m2 per day, and even more preferably about 1.1 - 1.9 g/m2 per day, e.g. in an amount of about 1.2 g/m2 per day, of about 1.4 g/m2 per day, or about 1.8 g/m2 per day, on Day 1 and Day 3 within one week, during solely one week of one cycle, preferably on Day 1 and Day 3 solely during the first week of the cycle. Preferably, more than one such cycle, e.g., 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 such cycles are performed substantially without a pause.
In another embodiment, the invention can be practiced in conjunction with surgical procedures where portions or all of a tumor mass has been removed. In this regard, the method can be practiced following a surgical procedure. Alternatively, the surgical procedure can be practiced during the interval between administration of the first active agent and the second active agent. Exemplary of this method is the combination of the present method with surgical tumor removal described below.
Administration can be accomplished by periodic unit dosages, by continuous infusion, peristaltic delivery, by bolus injection, and the like. Routes can include intravenous, subcutaneous, intramuscular, orthotopic injection, orthotopic infusion, oral application, and the like.
Another aspect of the instant invention is the use of an anti-angiogenic RGD peptide for the manufacture of a medicament for the treatment of neuroblastomas, wherein the medicament is to be applied in combination with a humanized anti-GD2-IL-2 fusion protein. According to this aspect of the invention, the humanized anti-GD2-IL-2 fusion protein preferably comprises, or more preferably is, hu14.18-IL2; and/or the RGD peptide comprises, or more preferably is, cilengitide or a pharmaceutically acceptable salt thereof.
According to this aspect of the invention, both the humanized anti-GD2-IL-2 fusion protein and the anti-angiogenic RGD peptide are preferably administered to the patient as separate pharmaceutical compositions. The humanized anti-GD2-IL-2 fusion protein and the anti-angiogenic RGD peptide, or the pharmaceutical compositions containing them, can be administered to the patient on separate days and/or both on the same day, whereby the administration on separate days and the administration of both on the same day can alternate. On the days where both the humanized anti- GD2-IL-2 fusion protein and the anti-angiogenic RGD peptide, or the pharmaceutical compositions containing them, are administered to the patient on the same day, the anti-angiogenic RGD peptide or the pharmaceutical composition containing it is preferably administered to the patient prior to the administration of the humanized anti-GD2-IL-2 fusion protein or the pharmaceutical composition containing it, preferably 30 minutes to 6 hours and more preferably 1 hour to 3 hours, e.g., about one hour, about two hours or about three hours, prior to the administration of the humanized anti-GD2-IL-2 fusion protein or the pharmaceutical composition containing it. Typically, the anti-angiogenic RGD peptide or the pharmaceutical composition containing it is then administered to the patient over a time period of 15 minutes to 2 hours, more preferably 30 minutes to 90 minutes, e.g. over a time period of about 1 hour, for example by i.v. infusion, and then, about directly or alternatively with a pause, preferably a pause of 10 minutes to 90 minutes, more preferably 20 to 70 minutes, e.g., a pause of about one hour, the humanized anti-GD2-IL-2 fusion protein or the pharmaceutical composition containing it is administered.
According to this aspect of the invention, the dosage regimens to be administered to the patient and the treatment regimens related thereto discussed herein preferably also apply here. The patient can be an adult or a child.
According to this aspect of the invention, the dosages to be administered to the patient and the treatment regimens related thereto in the treatment of neuroblastoma in children preferably comprise the following:
Preferably, the RGD peptide (e.g., cilengitide) is administered in an amount of about 0.5 - 2.5 g/m2 per day, more preferably about 1 - 2 g/m2 per day, and even more preferably about 1.1 - 1.9 g/m2 per day, e.g. in an amount of about 1.2 g/m2 per day, of about 1.4 g/m2 per day, or about 1.8 g/m2 per day, on 1 to 3 days and preferably about 2 days, during solely one week of one cycle, preferably solely during the first week of the cycle. Especially preferably, the RGD peptide (e.g., cilengitide) is administered in an amount of about 0.5 - 2.5 g/m2 per day, more preferably about 1 - 2 g/m2 per day, and even more preferably about 1.1 - 1.9 g/m2 per day, e.g., in an amount of about 1.2 g/m2 per day, of about 1.4 g/m2 per day, or about 1.8 g/m2 per day, on Day 1 and Day 3 during solely one week of one cycle, preferably on Day 1 and Day 3 solely during the first week of the cycle. Also especially preferably, more than one such cycle, e.g., 2 to 12, more preferably 2 to 8, and especially about 3 or about 6 of such cycles with respect to the RGD peptide are performed substantially without a pause.
Preferably, the anti-GD2-IL-2 fusion protein (e.g., hu14.18-IL2) is administered in an amount of about 1 - 20 mg/m2 per day, more preferably about 2 - 15 mg/m2 per day, and even more preferably about 4 - 12 mg/m2 per day, e.g., in an amount of about 4 mg/m2 per day, of about 8 mg/m2 per day, or about 12 mg/m2 per day, on 2-5 consecutive days and preferably about 3 consecutive days within solely one week of one cycle, preferably solely during the first week of the cycle. Especially preferably, the anti-GD2- IL-2 fusion protein (e.g., hu14.18-IL2) is administered in an amount of about 4 mg/m2 per day, of about 8 mg/m2 per day, or about 12 mg/m2 per day on 3 consecutive days during solely one week of one cycle, preferably solely during the first week of the cycle. Also especially preferably, more than one such cycle, e.g. 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 of such cycles with respect to the anti-GD2-IL-2 fusion protein are performed substantially without a pause. Even more preferably, more than one such cycle, e.g. 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 of such cycles with respect to both the anti- GD2-IL-2 fusion protein and the RGD peptide are performed substantially without a pause, preferably substantially in parallel. According to this aspect of the invention, the dosages to be administered to the patient and the treatment regimens related thereto in the treatment of neuroblastoma in adults preferably comprise the following:
Preferably, the amount of RGD peptide to be administered in the weekly administration can be the same or different in each week. More preferably, the dosages of the RGD peptide, preferably cilengitide, are selected from the dosages or regimens (a) to (f) as described before and mixtures thereof and more preferably are selected from the dosages or regimens (a), (b), (d), (e) and (f); especially preferred in this respect is regimen or dosage (f).
Also especially preferably, more than one such cycle, e.g., 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 of such cycles with respect to the RGD peptide are performed substantially without a pause.
Preferably, the anti-GD2-IL-2 fusion protein (e.g., hu14.18-IL2) is administered in an amount of about 1 - 20 mg/m2 per day, more preferably about 2 - 15 mg/m2 per day, and even more preferably about 4 - 12 mg/m2 per day, e.g., in an amount of about 4 mg/m2 per day, of about 8 mg/m2 per day, or about 12 mg/m2 per day, on 2-5 consecutive days, and preferably about 3 consecutive days, during solely one week of one cycle, preferably solely during the first week of the cycle. Especially preferably, the anti-GD2- IL-2 fusion protein (e.g., hu14.18-IL2) is administered in an amount of about 4 mg/m2 per day, of about 8 mg/m2 per day, or about 12 mg/m2 per day on 3 consecutive days during solely one week of one cycle, preferably solely during the first week of the cycle. Also especially preferably, more than one such cycle, e.g., 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 of such cycles with respect to the anti-GD2-IL-2 fusion protein are performed substantially without a pause. Even more preferably, more than one such cycle, e.g. 2 to 12 cycles, more preferably 2 to 8 cycles, and especially about 3 or about 6 of such cycles with respect to both the anti- GD2-IL-2 fusion protein and the RGD peptide are performed substantially without a pause, preferably substantially in parallel.
A preferred aspect of the instant invention is a method of treatment, wherein the above described use, medicaments and/or pharmaceutical compositions are applied to the patient as described with respect to said use, medicaments and/or pharmaceutical compositions, as described above.
Hu14.18-IL2 has preclinical activity in mouse models of neuroblastoma (NB), particularly when given to mice with a smaller tumor burden. The anti-tumor activity of hu14.18-IL2 was also determined in two strata of human patients with recurrent or refractory NB.
In the human trial, hu14.18-IL2 was given intravenously at a dose of 12 mg/m2/day over 4 hours for three consecutive days every 28 days. Responses were evaluated every 2 cycles for patients with disease measurable by standard radiographical criteria (stratum 1) and for patients with disease evaluable only by meta-iodobenzylguanidine (MIBG) imaging and/or bone marrow histology (stratum 2). In each stratum, a one-stage design was powered to conclude efficacy with 4 or more responders
(complete remission, CR, or partial remission, PR) out of 20. Response was established by independent radiology review as well as bone marrow histology and immuno-cytochemistry, and durability assessed by repeat evaluation after greater than 3 weeks.
Of the 39 patients that were enrolled, 37 were evaluable. No responses were seen in stratum 1 (n=13). In stratum 2, response rate was 21% (5/24 overall; 95% confidence interval (Cl: 5%, 37%), all 5 in first 20. All 5 responders had CR (3/5 bone marrow disease only at study entry, 1/5 MIBG-avid disease only and 1/5 marrow and MIBG-avid disease) of 12, 8, 8+, 7+ and 3+ month duration. Two additional stratum 2 patients had partial improvement, but did not meet criteria for a PR. Two patients required dopamine for hypotension; 1 patient required ventilatory support for capillary leak and hypoxia. Other grade 3 and 4 non-hematologic toxicities included pain, rash, allergic reaction, fever and transaminitis. Most toxicities were reversible within 1-2 days of completing a treatment course and were expected based on phase 1 results. Patients with MIBG and/or bone marrow only disease had a 21% CR rate to hu14.8-IL2, while patients with bulky disease did not respond.
In a clinical treatment setting, the following exemplary protocol set forth in Table 2 can be followed.
Table 2.
Figure imgf000031_0001
Notes for Table 2:
1. Day 29 of each course = Day 1 of subsequent course.
2. The next course of treatment is initiated if there is no progression of disease by tumor measurement and no adverse events requiring removal from study has occurred.
The therapeutic compositions used in a method of this invention comprise the active agent (i.e., the anti-angiogenic RGD peptide or the anti-GD2-IL-2 fusion protein) in a pharmaceutically acceptable carrier, as is well known, and therefore the invention is not to be construed as limited with regards to the composition so long as the concentration of the active agent(s) in the respective compositions is sufficient for delivery (administration) of the recited active agent in the amounts described herein. All aspects of the instant invention can be combined with other (further) treatment regimens known in the art of treating cancerous diseases in the broadest sense.
The foregoing examples describing certain embodiments of the invention are illustrative and should not, of course, be construed as specifically limiting the invention. The aspects, features, properties and advantages exemplified for the compounds, pharmaceutical compositions, methods of treatments and uses defined in the examples, tables and/or figures may be assigned to other compounds, pharmaceutical compositions, methods of treatments and uses not specifically described and/or defined in the examples, tables and/or figures, but falling under the scope of what is defined in the claims. Moreover, such variations of the invention, now known or later developed, which would be within the purview of one skilled in the art are to be considered to fall within the scope of the present invention hereinafter claimed.

Claims

What is claimed is:
1. A method for treating a malignant neoplasm, preferably a disseminated malignant neoplasm, which comprises administering to a patent in need of such treatment a malignant cell proliferation inhibiting amount of: a) an anti-angiogenic RGD peptide; b) a humanized anti-GD2-IL-2 fusion protein.
2. The method of claim 1 wherein the humanized anti-GD2-IL-2 fusion protein comprises hu14.18-IL2.
3. The method of claim 1 or claim 2 wherein the RGD peptide comprises cilengitide or a pharmaceutically acceptable salt thereof.
4. The method for treating a melanoma tumor cell in a patient comprising administering to the patient a tumor cell proliferation inhibiting amount of: a) an anti-angiogenic RGD peptide; and b) a humanized anti-GD2-IL-2 fusion protein.
5. The method of claim 4 wherein the humanized anti-GD2-IL-2 fusion protein comprises hul14.18-IL2.
6. The method of claim 4 or claim 5 wherein the RGD peptide comprises cilengitide or a pharmaceutically acceptable salt thereof.
7. The method of any one of claims 4-6 wherein the anti-angiogenic RGD peptide and the humanized anti-GD2-IL-2 fusion protein are administered substantially concurrently.
8. The method of any one of claims 4-6 wherein the anti-angiogenic RGD peptide and the humanized anti-GD2-IL-2 fusion protein are administered sequentially.
9. The method of claim 8 wherein the anti-angiogenic RGD peptide is administered before the humanized anti-GD2-IL-2 fusion protein.
10. The method of claim 8 wherein the humanized anti-GD2-IL-2 fusion protein is administered before the anti-angiogenic RGD peptide.
11. The method of any one of claims 4-10 wherein the administration is effected during a time period when a tumor or tumor metastases is surgically removed from the patient.
12. The method of any on of claims 4-10 wherein the administration is effected after a tumor or tumor metastases has been surgically removed from the patient.
13. The method of any one of claims 4-12 wherein the anti-angiogenic RGD peptide is administered to a child at a dosage of about 1 to about 8 grams per square meter of body surface area per week, for one or more weeks during at least one treatment cycle.
14. The method of any one of claims 1-13 wherein the humanized anti-
GD2-IL-2 fusion protein is administered to a child at a dosage of about 1 to about 100 mg per square meter of body surface area per week, for one or more weeks during at least one treatment cycle.
15. The method of any one of claims 4-12 wherein the anti-angiogenic RGD peptide is administered to an adult at a dosage sufficient to provide a plasma concentration of about 2 micromolar to about 5 millimolar.
16. The method of any one of claims 4-12 and 15 wherein the humanized anti-GD2-IL-2 fusion protein is administered to an adult at a dosage sufficient to provide a plasma concentration of about 2 micromolar to about 5 millimolar.
17. A method for treating a neuroblastoma tumor cell in a patient comprising administering to the patient a tumor cell proliferation inhibiting amount of: a) an anti-angiogenic RGD peptide; and b) a humanized anti-GD2-IL-2 fusion protein.
18. The method of claim 17 wherein the humanized anti-GD2-IL-2 fusion protein comprises hu14.18-IL2.
19. The method of claim 17 or claim 18 wherein the RGD peptide comprises cilengitide or a pharmaceutically acceptable salt thereof.
20. The method of any one of claims 17-19 wherein the anti-agiogenic RGD peptide and the humanized anti-GD2-IL-2 fusion protein are administered substantially concurrently.
21. The method of any one of claims 17-20 wherein the anti-angiogenic RGD peptide and the humanized anti-GD2-IL-2 fusion protein are administered sequentially.
22. The method of claim 21 wherein the anti-angiogenic RGD peptide is administered before the humanized anti-GD2-IL-2 fusion protein.
23. The method of claim 21 wherein the humanized anti-GD2-IL-2 fusion protein is administered before the anti-angiogenic RGD peptide.
24. The method of any one of claims 17-23 wherein the administration is effected during a time period when a tumor or tumor metastases is surgically removed from the patient.
25. The method of any one of claims 17-23 wherein the administration is effected after a tumor or tumor metastases is surgically removed from the patient.
26. The method of any one of claims 17-25 wherein the anti-angiogenic
RGD peptide is administered to a child at a dosage of about 1 to about 8 grams per square meter of body surface area per week, for one or more weeks during at least one treatment cycle.
27. The method of any one of claims 17-26 wherein the humanized anti- GD2-IL-2 fusion protein is administered to a child at a dosage of about 1 to about 100 mg per square meter of body surface area per week, for one or more weeks during at least one treatment cycle.
28. The method of any one of claims 17-25 wherein the anti-angiogenic RGD peptide is administered to an adult at a dosage sufficient to provide a plasma concentration of about 2 micromolar to about 5 millimolar.
29. The method of any one of claims 17-25 and 28 wherein the humanized anti-GD2-IL-2 fusion protein is administered to an adult at a dosage sufficient to provide a plasma concentration of about 2 micromolar to about 5 millimolar.
PCT/EP2009/003372 2008-05-30 2009-05-12 Methods for treatment of malignancies WO2009152901A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US13037708P 2008-05-30 2008-05-30
US61/130,377 2008-05-30
US61/130,378 2008-05-30
US13037808P 2008-12-19 2008-12-19

Publications (1)

Publication Number Publication Date
WO2009152901A1 true WO2009152901A1 (en) 2009-12-23

Family

ID=41100588

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2009/003372 WO2009152901A1 (en) 2008-05-30 2009-05-12 Methods for treatment of malignancies

Country Status (1)

Country Link
WO (1) WO2009152901A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018093797A1 (en) * 2016-11-15 2018-05-24 The Schepens Eye Research Institute, Inc. Compositions and methods for the treatment of aberrant angiogenesis
US11739326B2 (en) 2017-11-14 2023-08-29 Massachusetts Eye And Ear Infirmary RUNX1 inhibition for treatment of proliferative vitreoretinopathy and conditions associated with epithelial to mesenchymal transition

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5650150A (en) * 1990-11-09 1997-07-22 Gillies; Stephen D. Recombinant antibody cytokine fusion proteins
US20040203100A1 (en) * 2002-12-17 2004-10-14 Emd Lexigen Research Center Corp. Immunocytokine sequences and uses thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5650150A (en) * 1990-11-09 1997-07-22 Gillies; Stephen D. Recombinant antibody cytokine fusion proteins
US20040203100A1 (en) * 2002-12-17 2004-10-14 Emd Lexigen Research Center Corp. Immunocytokine sequences and uses thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
BURKE P A ET AL: "Cilengitide Targeting of alphavbeta3 Integrin Receptor Synergizes with Radioimmunotherapy to Increase Efficacy and Apoptosis in Brest Cancer Xenografts1", CANCER RESEARCH, AMERICAN ASSOCIATION FOR CANCER RESEARCH, BALTIMORE, MD., US, vol. 62, no. 15, 1 August 2002 (2002-08-01), pages 4263 - 4272, XP002903574, ISSN: 0008-5472 *
LODE H N ET AL: "Synergy between an antiangiogenic integrin alphav antagonist and an antibody-cytokine fusion protein eradicates spontaneous tumor metastases", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA, NATIONAL ACADEMY OF SCIENCE, WASHINGTON, DC, US, vol. 96, no. 4, 16 February 1999 (1999-02-16), pages 1591 - 1596, XP002134006, ISSN: 0027-8424 *
MITJANS FRANCESC ET AL: "In vivo therapy of malignant melanoma by means of antagonists of alphav integrins", INTERNATIONAL JOURNAL OF CANCER, vol. 87, no. 5, 1 September 2000 (2000-09-01), pages 716 - 723, XP002547772, ISSN: 0020-7136 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018093797A1 (en) * 2016-11-15 2018-05-24 The Schepens Eye Research Institute, Inc. Compositions and methods for the treatment of aberrant angiogenesis
US11229662B2 (en) 2016-11-15 2022-01-25 The Schepens Eye Research Institute, Inc. Compositions and methods for the treatment of aberrant angiogenesis
US11739326B2 (en) 2017-11-14 2023-08-29 Massachusetts Eye And Ear Infirmary RUNX1 inhibition for treatment of proliferative vitreoretinopathy and conditions associated with epithelial to mesenchymal transition

Similar Documents

Publication Publication Date Title
US20210155700A1 (en) ANTI-DEspR INHIBITORS AS THERAPEUTICS FOR INHIBITION OF PATHOLOGICAL ANGIOGENESIS AND TUMOR CELL INVASIVENESS AND FOR MOLECULAR IMAGING AND TARGETED DELIVERY
CA2436326C (en) Combination therapy using receptor tyrosine kinase inhibitors and angiogenesis inhibitors
US8034904B2 (en) Anti-IGF-I receptor antibody
JP7102670B2 (en) Fusion of anti-PD-L1 antibody and IL-7
EP2759297B1 (en) Combination therapy using p53 activator and c-Met inhibitor
US9931400B2 (en) Method of combination therapy for prevention or treatment of c-Met or angiogenesis factor induced diseases
JP2021130668A (en) Treatment for multiple myeloma (mm)
JP2012518664A (en) Anti-ICAM-1 antibodies, uses and methods
JP2018516950A (en) Intensive interferon immunotherapy for cancer treatment
KR20220024035A (en) Combination of 4-1BB co-stimulation with dual specific antigen binding molecules that bind MUC16 and CD3
WO2009152901A1 (en) Methods for treatment of malignancies
BR112020026159A2 (en) DOSAGE SCHEMES FOR INHIBITION OF TARGET TGF-BETA FOR USE IN THE TREATMENT OF BILIARY TRACT CANCER
TW202003577A (en) Dosing regimens for targeted tgf-β inhibition for use in treating cancer in treatment naïve subjects
US20230034186A1 (en) Methods of treating cancer using multi-specific binding proteins that bind nkg2d, cd16 and a tumor-associated antigen
US20240010729A1 (en) Combination therapy of a pd-1 antagonist and lag3 antagonist and lenvatinib or a pharmaceutically acceptable salt thereof for treating patients with cancer
US20230141413A1 (en) Immunotherapy with combination therapy comprising an immunotoxin
JP5514388B2 (en) Preparation method of medicine, preparation method of cancer therapeutic agent, pharmaceutical composition for cancer treatment, preparation method of pharmaceutical composition for cancer treatment, neutralization method of complement activity inhibitory action of CD55 and complement deposition How to make
KR20230129467A (en) Combination therapy using anti-fucosyl-GM1 antibodies
JP2023504630A (en) Bispecific fusion proteins for tumor therapy
KR20200072507A (en) Combination products for cancer treatment
WO2023232140A1 (en) Cancer treatment with a pd-1 or pd-l1 inhibitor and an antibody-drug conjugates targeting claudin 18.2
TW202306588A (en) Use of antibody-drug conjugate in combination with immune checkpoint inhibitor in treatment of urothelial cancer
JP3845735B2 (en) A treatment for pemphigus comprising a CD40L antagonist as an active ingredient
TW201729827A (en) Peptides and peptidomimetics in combination with T cell activating and/or checkpoint inhibiting agents for cancer treatment

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09765481

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09765481

Country of ref document: EP

Kind code of ref document: A1