WO2006089150A2 - Antiangiogenic agents with aldesleukin - Google Patents

Antiangiogenic agents with aldesleukin Download PDF

Info

Publication number
WO2006089150A2
WO2006089150A2 PCT/US2006/005720 US2006005720W WO2006089150A2 WO 2006089150 A2 WO2006089150 A2 WO 2006089150A2 US 2006005720 W US2006005720 W US 2006005720W WO 2006089150 A2 WO2006089150 A2 WO 2006089150A2
Authority
WO
WIPO (PCT)
Prior art keywords
aldesleukin
ril
amine
cells
des
Prior art date
Application number
PCT/US2006/005720
Other languages
French (fr)
Other versions
WO2006089150A3 (en
Inventor
Sharon Lea Aukerman
Kimberly Denis-Mize
Laurence Elias
Bahija Jallal
Daniel Menezes
Gary W. Witherell
Original Assignee
Novartis Vaccines And Diagnostics Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Novartis Vaccines And Diagnostics Inc. filed Critical Novartis Vaccines And Diagnostics Inc.
Priority to EP06735400A priority Critical patent/EP1853302A2/en
Priority to BRPI0608880-5A priority patent/BRPI0608880A2/en
Priority to AU2006214138A priority patent/AU2006214138A1/en
Priority to JP2007556337A priority patent/JP2008530239A/en
Priority to MX2007010037A priority patent/MX2007010037A/en
Priority to CA002598448A priority patent/CA2598448A1/en
Publication of WO2006089150A2 publication Critical patent/WO2006089150A2/en
Publication of WO2006089150A3 publication Critical patent/WO2006089150A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/502Pyridazines; Hydrogenated pyridazines ortho- or peri-condensed with carbocyclic ring systems, e.g. cinnoline, phthalazine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4412Non condensed pyridines; Hydrogenated derivatives thereof having oxo groups directly attached to the heterocyclic ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2013IL-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/02Non-specific cardiovascular stimulants, e.g. drugs for syncope, antihypotensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives

Definitions

  • the present invention relates to methods of therapy for diseases associated with abnormal cellular proliferation.
  • recombinant IL-2 is combined with antangiogenic agents for use in the treatment of cancer.
  • Capillaries reach into almost all tissues of the human body and supply tissues with oxygen and nutrients as well as removing waste products. Under typical conditions, the endothelial cells lining the capillaries do not divide, and capillaries, therefore, do not normally increase in number or size in a human adult. Under certain conditions, however, such as when a tissue is damaged, or during certain parts of the menstrual cycle, the capillaries begin to proliferate rapidly. This process of forming new capillaries from pre-existing blood vessels is known as angiogenesis or neovascularization. See Folkman, J. Scientific American 275, 150-154 (1996). Angiogenesis during wound healing is an example of pathophysiological neovascularization during adult life.
  • the additional capillaries provide a supply of oxygen and nutrients, promote granulation tissue, and aid in waste removal. After termination of the healing process, the capillaries normally regress. Lymboussaki, A. "Vascular Endothelial Growth Factors and their Receptors in Embryos, Adults, and in Tumors" Academic Dissertation, University of Helsinki, Molecular/Cancer Biology Laboratory and Department of Pathology, Haartman Institute, (1999).
  • Angiogenesis also plays an important role in the growth of cancer cells. It is known that once a nest of cancer cells reaches a certain size, roughly 1 to 2 mm in diameter, the cancer cells must develop a blood supply in order for the tumor to grow larger as ⁇ iltusion will not be sufficient to supply the cancer cells with enough oxygen and nutrients. Thus, inhibition of angiogenesis is expected to halt the growth of cancer cells.
  • VEGF vascular endothelial growth factor
  • EGF epidermal growth factor
  • FGF fibroblast growth factor
  • PDGF platelet derived growth factor
  • anti-tumor immunity involves activation of multiple cell types in the immune system, the most efficient being cytolytic T lymphocytes.
  • cytolytic T lymphocytes To induce specific, anti-tumor T lymphocyte mediated immune response, recognition of not only the tumor antigen of interest, but also costimulatory interactions between specific ligands present on either the tumor cell or the antigen presenting cell, and the target T lymphocytes are required.
  • This second, co-stimulatory signal may also be provided by soluble factors, such as cytokines or other peptide molecules which bind to specific, cell surface receptors and initiate various signal transduction pathways, resulting in augmentation of effector function.
  • Interleukin-2 is a T lymphocyte-derived cytokine which binds to specific receptors present on T cells and natural killer (NK) cells, and will activate them for tumor cytolysis, cytokine secretion, and other effector functions.
  • Both CD4 and CD8 T lymphocytes express the receptor for IL-2, and develop increased cytolytic effector and cytokine synthetic function after exposure to the biologic.
  • the functional effects of IL-2 are mediated via engagement with the IL-2 receptor (IL- 2R), which is structurally composed of three subunits: ⁇ , ⁇ , and a common ⁇ chain that is shared with other cytokine receptors (Caligiuri et al., J. Exp. Med.
  • T cells express the high affinity IL-2R ⁇ , whereas NK cells, monocytes, and macrophages express predominantly the intermediate-affinity form, IL-2R ⁇ (Caligiuri et al., J. Exp. Med. 1990 171(5): 1509-1526; Voss et al., J. Exp. Med. 1992 176(2):531-541; Theze et al., Immunol. Today 1996 17(10):481-486; Nagler et al, J. Exp. Med. 1990 171(5): 1527-1533).
  • IL-2 activates the JAKs (JAKl, JAK3), which phosphorylate key tyrosines on IL-2R ⁇ , which serve as docking sites for downstream signaling molecules, including She and Stat5, which then catalyze the activation of two distinct pathways: Shc/Ras/Raf/MAPK, and the JAK/STAT5, which translocates to the nucleus and directly regulates a family of gene-regulating transcription factors- STATs (O'Shea, J.J., Immunity 1997 7(1):1-11).
  • Grb-2/Sos complex and activates the Ras/Raf/MAPK pathway, and Grb-2/Gab-2, which activates the phosphatidylinositol 3-kinase (PI3K) pathway. Together these signaling proteins regulate gene transcription factors, ultimately controlling cell growth, division, differentiation and immune activity.
  • PI3K phosphatidylinositol 3-kinase
  • IL- 2 also induces anti-apoptotic effects by regulating pro-mitogenic genes leading to increased bcl-2, bcl-xl, c-myb, which affects cell cycle control through activation of cdk 2,4,6 and inhibition of p27kipl or may be negatively regulated by SOCS (suppressors of cytokine signaling) (Smith, K.A., Science 1988 240(4856): 1169-1176; Theze et al., Immunol. Today 1996 17(10):481-486).
  • SOCS suppressors of cytokine signaling
  • recombinant human IL-2 when administered to tumor-bearing animals for periods of 10 to 14 days can result in regression of tumor burdens, long-term survival, and increased resistance to tumor regrowth.
  • Analysis of splenic lymphocytes obtained from these animals has shown that the anti-tumor effects are due at least in part to augmentation of cytolytic T cell function. This effect includes activation of both direct cytolysis of tumor cells by the CTL, as well an increased synthesis of other, T lymphocyte derived cytokines, which may have further direct or indirect anti-tumor effects as well.
  • RRCC renal cell carcinoma
  • RTK small molecule receptor tyrosine kinase
  • BAY 43-9006 is a potent inhibitor of Raf- 1 , a member of the Raf/MEK/ERK signaling pathway (Richly et al., Int. J. Clin. Pharmacol. Ther.
  • SUl 1248 is a highly potent, selective RTK inhibitor of VEGFR-I- 3, PDGFR ⁇ , cKIT, FLT3 and PDGFR ⁇ (Abrams et al., MoI Cancer Ther. 2003 2(5):471-478; Mendel et al., Clin. Cancer Res. 2003 9(l):327-337; O'Farrell et al., Clin. Cancer Res. 2003 9(15):5465-5476).
  • SU11248 has shown antitumor activity in a number of advanced solid tumors (RCC, neuroendocrine, stromal and adenocarninomas) (Faivre et al., J. Clin. Oncol.
  • Clinical data of SUl 1248 also indicate that the drug is generally tolerated with manageable toxicities, which include fatigue, lymphopenia, neutropenia, hyperlipasemia (Faivre et al., J. Clin. Oncol. 2006 24(l):23-35; Motzer et al., J. Clin. Oncol. 2006 24(1): 16-24).
  • VHL von Hippel-Lindau tumor suppressor gene
  • Vascular endothelial growth factor represents a good target for treatment of clear-cell kidney cancer because mutations in the von Hippel-Lindau tumor- suppressor gene, result in overproduction of this growth factor by the tumors.
  • a randomized, double-blind, placebo-controlled study of the humanized monoclonal antibody (bevacizumab) in patients with metastatic clear-cell renal cancer was conducted by Yang et al.
  • Recombinant human interleukin-2 (aldesleukin) was approved by the FDA for the treatment of metastatic renal cancer based on the results of several multicenter trials in 255 patients who received an intermittent high dose bolus regimen. In these trials, objective responses were seen in 15% of patients, and median survival was 16.3 months (Fisher and Rosenberg 2000 Cancer J Sci Am 6Sl :S55-57). Owing to the significant toxicity associated with this regimen, a series of phase I and phase II trials employing rIL-2 at different doses and using different routes of administration were undertaken. A recent review of the efficacy of rIL-2 as a single agent indicated an overall response of 15% in more than 1700 patients with metastatic RCC who were treated in this series of studies.
  • rIL-2 may also prevent tumor proliferation by causing secondary cytokine release (IFN ⁇ ) from activated NK cells.
  • IFN ⁇ secondary cytokine release
  • Saraya KLA Balkwill FR. Temporal sequence and cellular origin of interleukin-2 stimulated cytokine gene expression. British Journal of Cancer. 1993;67(3):514-21. The response rates and outcomes of clear cell renal cancer patients can be improved. Clinical trials employing novel drug combinations are needed.
  • the combination of rIL2 and antiangiogenic agents, such as bevacizumab may have additive effects that could translate to added clinical benefit for patients with metastatic renal cell carcinoma, as well as other cancers. Another advantage to the complimentary approach to cancer regression is that it provides a platform less easily bypassed by resistance mutations.
  • a single point mutation in the disease state may render it unaffected by a drug resulting in even harsher strains of the disease in future generations.
  • Novel methods and mechanisms for treating patients having disorders associated with abnormal proliferation that are resistant to, or inadequately treated by conventional approaches, utilizing agents targeting immune response mechanisms in the body and disease-state substrates, are needed.
  • the present invention provides such therapeutic agents, and further provides other related advantages.
  • the present invention is based in part on unique combination therapies using small molecule receptor tyrosine kinase inhibitors and rIL-2 with non-overlapping toxicities.
  • melanomas and RCC are generally responsive to immunotherapy such as rIL-2
  • rational combinations of rIL-2 with potential receptor tyrosine kinase inhibitors, based on pharmacology of the individual agents, have been evaluated herein.
  • the invention provides clinically applicable drug schedules to circumvent potential drug interactions based on the toxicological profile of these agents.
  • the potential of adverse pharmacokintic/pharmacodynamic interations and adverse pharmacological interactions are also elucidated.
  • the effects of rIL-2 and small molecule receptor tyrosine kinase inhibitors, such as BAY 43-9006, on T cells and the impact on IL-2 -mediated signaling pathways (MAPK and JAK/STAT5) is described.
  • the present invention expands the indication of the anti-tumor efficacy of IL-2 compounds, such as recombinant IL-2 (rIL-2, also known as aldesleukin) against cancer cell lines that respond poorly to conventional therapies, resulting in increased long-term survival and immunity to tumor rechallenge.
  • IL-2 compounds such as recombinant IL-2 (rIL-2, also known as aldesleukin) against cancer cell lines that respond poorly to conventional therapies, resulting in increased long-term survival and immunity to tumor rechallenge.
  • rIL-2 also known as aldesleukin
  • the immunostimulotory effects of rIL-2 aim to alleviate existing side effects caused by administration of antiangiogenic compositions for co-administration therewith.
  • Methods of treating a subject suffering from abnormal cellular proliferation, particularly renal cell carcinoma using a combination of an IL-2 compound, such as rIL-2 and at least one antiangiogenic agent is provided.
  • IL-2 compound such as rIL-2
  • small molecules of the present invention are listed in Tables 1-5. Particularly preferred molecules are N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-a small molecule.
  • Aldesleukin and antiangiogenic agents may be administered together or separately as individual pharmaceutical compositions. If administered separately, Aldesleukin can be administered prior to, concurrent with, or subsequent to the antiangiogenic agent.
  • aldesleukin with antiangiogenic agent such as 6,7-bis(2-methoxyethoxy)-N-(3- ethynylphenyl)quinazolm-4-amine, 6-(3-mo ⁇ holinopropoxy)-N-(3-chloro-4- fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2-(dimethylamino)ethyl)-5-((5- fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl-lH-pyrrole-3-carboxamide, N-(4- chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin-l -amine, and l-(4-(2- (methylcarbamoyl) ⁇ yridin-4-yloxy
  • the antiangiogenic agents preferably one of 6,7-bis(2- methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6-(3-morpholinopropoxy)- N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2-
  • antiangiogenic proteins such as monoclonal antibodies capable of inhibiting VEGF.
  • antiangiogenic proteins of the present invention are bevacizumab or VEGF-Trap.
  • the protein is an EGF inhibitor, such as cetuximab.
  • administering potentiates the effectiveness of the antiangiogenic agent, resulting in a positive/synergistic therapeutic response that is improved with respect to that observed with the inhibitor alone.
  • a therapeutic package suitable for commercial sale for treating a patient suffering from cancer comprising a container, a therapeutically effective amount of aldesleukin, and a therapeutically effective amount of an antiangiogenic agent and/or small molecule, preferably listed in Tables 1-5, such as 6,7-bis(2-methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6-(3- morpholinopropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N- (2-fdimethvlamino ') ethvlV5-ff5-fli ] nro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl- lH-pyrrole-3-carboxamide, N-(4-chlorophenyl)-4-((pyridin-4-yl)methyl)phthal
  • kits comprises a combination of medicaments for the treatment of a patient suffering from cancer, comprising: (a) aldesleukin, and (b) an antiangiogenic agent selected from 6,7-bis(2- methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6-(3-morpholinopropoxy)- N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2- (dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl-lH- pyrrole-3-carboxamide, N-(4-chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin-l- amine, or l-(4-(2-(methylcarbamoyl)pyr
  • Figure 1 shows single agent activity of rIL-2, BAY 43-9006 or SUl 1248 in IL-2-responsive, T-cell competent murine tumor models.
  • B16-F10 melanoma (2 x 10 6 ), CT26 colon (2 x 10 6 ) or RENCA renal carcinoma (Ix 10 6 ) cells were implanted s.c. into the right flank of female C57BL6 or BALB/c mice. Treatments were initiated when tumors were established to a mean size of 50 - 225 mm 3 , as outlined in methods. Mice were randomized into treatment cohorts (10 mice/group). rIL-2 was administered daily subcutaneously (0.2-3 mg/kg/d).
  • FIG. 1 illustrates the mean tumor growth inhibition (calculated as [l-(mean tumor volume of treated group/mean tumor volume of vehicle group) x 100]) of single agents in the B 16-F 10, CT26 and RENCA tumor model between days 10-14. The data is compiled from multiple independent studies with each study of 10 mice/group. * denotes statistical significance vs. Vehicle treatment (p ⁇ 0.05, ANOVA).
  • Figure 2 shows efficacy of concomitant rIL-2 and BAY 43-9006 therapy in the B16-F10 murine melanoma model.
  • B16-F10 cells (2 x 10 6 cells) were implanted subcutaneously in the right flank of female C57BL6 mice. Mice were randomized into groups when tumors were ⁇ 50 mm 3 , and treatments initiated on day 0 with either vehicle, days 0-6 ( ⁇ ), rIL-2 (3.3 mg/kg, s.c, days 0-6) ( ⁇ ) or BAY 43-9006 (30 mg/kg, p.o. days 0-6) (x) or combined rIL-2 (3.3 mg/kg, s.c, days 0-6) + BAY 43- 9006 (30 mg/kg, p.o. days 0-6) ( ⁇ ).
  • Figures 3 A and 3B show efficacy of sequential rIL-2 and BAY 43-9006 therapy in the B 16-F 10 murine melanoma model.
  • B 16-F 10 cells (2 x 10 6 cells) were implanted subcutaneously in the right flank of female C57BL6 mice. Mice were randomized into groups when tumors were -50 mm3, and treatments initiated on day 0.
  • Figure 3A shows results from sequential regimen with rIL-2 administered first, then BAY 43-9006. Panel illustrates: vehicle ( ⁇ ) days 0-6, 7-13; rIL-2 (3.3 mg/kg, s.c, days 0-6) ( ⁇ ) or BAY 43-9006 (30 mg/kg, p.o.
  • FIG. 3B shows results from sequential regimen with BAY 43-9006 administered first, followed by rIL-2.
  • Panel illustrates: vehicle ( ⁇ ) days 0-6, 7-13; BAY 43-9006 (30 mg/kg, p.o. days 0-6) (x); rIL-2 (3.3 mg/kg, s.c, days 7-13) (D) or combined BAY 43-9006 (30 mg/kg, p.o. days 0-6) + rIL-2 (3.3 mg/kg, s.c, days 7-13) (A).
  • FIG 4 shows efficacy of concomitant rIL-2 and SUl 1248 therapy in the B16-F10 murine melanoma model.
  • B16-F10 cells (2 x 10 6 cells) were implanted subcutaneously in the right flank of female C57BL6 mice. Mice were randomized into groups when tumors were ⁇ 50 mm 3 , and treatments initiated on day 0 with either vehicle, days 0-6 (0), rIL-2 (3.3 mg/kg, s.c, days 0-6) (D) or SUl 1248 (40 mg/kg, p.o. days 0-6) (O) or combined rIL-2 (3.3 mg/kg, s.c, days 0-6) + SUl 1248 (40 mg/kg, p.o. days 0-6) (A).
  • Figures 5 A and 5B show efficacy of sequential rIL-2 and SUl 1248 therapy in the Bl 6-F 10 murine melanoma model.
  • Bl 6-F 10 cells (2 x 10 6 cells) were implanted subcutaneously in the right flank of female C57BL6 mice. Mice were randomized into groups when tumors were ⁇ 50 mm J , and treatments initiated on day 0.
  • Figure 5A shows results from sequential regimen with rIL-2 administered first, then SUl 1248. Panel illustrates: vehicle (0) days 0-6, 7-13; rIL-2 (3.3 mg/kg, s.c, days 0-6) ( ⁇ ) or SUl 1248 (40 mg/kg, p.o.
  • FIG. 5B shows results from sequential regimen with SUl 1248 administered first, followed by rIL-2.
  • Panel illustrates: vehicle (0) days 0-6, 7-13; SUl 1248 (40 mg/kg, p.o. days 0-6) (O); rIL-2 (3.3 mg/kg, s.c, days 7-13) ( ⁇ ) or combined SUl 1248 (40 mg/kg, p.o. days 0-6) + rIL-2 (3.3 mg/kg, s.c, days 7-13) ( ⁇ ).
  • Figure 6 shows efficacy of sequential rIL-2 and BAY 43-9006 therapy in the CT26 murine colon adenocarcinoma model.
  • CT26 cells (2 x 10 6 cells) were implanted subcutaneously in the right flank of female BALB/c mice. Mice were randomized into groups when tumors were -225 mm 3 , and treatments initiated on day 0.
  • Panel illustrates the sequential regimen with rIL-2 administered first, then BAY 43- 9006: vehicle ( ⁇ ) days 0-6, 7-13; rIL-2 (1 mg/kg, s.c, days 0-6) (D) or BAY 43- 9006 (40 mg/kg, p.o. days 7-13) ( ⁇ ) or combined rIL-2 (1 mg/kg, s.c, days 0-6) + BAY 43-9006 (40 mg/kg, p.o. days 7-13) (•).
  • Figure 7 shows efficacy of concomitant treatment of rIL-2 and SUl 1248 in the CT26 murine colon adenocarcinoma model.
  • CT26 cells (2 x 10 6 cells) were implanted subcutaneously in the right flank of female BALB/c mice. Mice were randomized into groups when tumors were -225 mm 3 , and treatments initiated on day 0.
  • Panel illustrates: vehicle ( ⁇ ) days 0-6; rIL-2 (1 mg/kg, s.c, days 0-6) (D) or SUl 1248 (40 mg/kg, p.o. days 0-6) (O) or combined rIL-2 (1 mg/kg, s.c, days 0-6) + SUl 1248 (40 mg/kg, p.o. days 0-6) (•).
  • Figures 8 A and 8B shows efficacy of sequential rIL-2 and SUl 1248 therapy in the CT26 murine colon adenocarcinoma model.
  • CT26 cells (2 x 10 6 cells) were implanted subcutaneously in the right flank of female BALB/c mice. Mice were randomized into groups when tumors were -225 mm 3 , and treatments initiated on day 0.
  • Figure 8A shows results from the sequential regimen with rIL-2 administered first, then SUl 1248: vehicle ( ⁇ ) days 0-6, 7-13; rIL-2 (1 mg/kg, s.c, days 0-6) (D) or SUl 1248 (40 mg/kg, p.o.
  • FIG. 8B shows results irom sequential regimen with SUl 1248 administered first, followed by rIL-2.
  • Panel illustrates: vehicle ( ⁇ ) days 0-6, 7-13; SUl 1248 (40 mg/kg, p.o. days 0-6) (O); rIL-2 (3.3 mg/kg, s.c, days 7-13) (D) or combined SUl 1248(40 mg/kg, p.o. days 0-6) + rIL-2 (3.3 mg/kg, s.c, days 7-13) (A).
  • Figure 9 shows efficacy of concomitant rIL-2 and BAY 43-9006 therapy in the murine RCC RENCA tumor model.
  • RENCA cells (1 x 10 6 cells) were implanted subcutaneously in the right flank of female BALB/c mice. Mice were randomized into groups when tumors were ⁇ 50 mm 3 , and treatments initiated on day 0 with either vehicle, days 0-8 ( ⁇ ), rIL-2 (1 mg/kg, s.c, days 0-4, 7-11) ( ⁇ ) or BAY 43-9006 (30 mg/kg, p.o. days 0-8) (x) or combined rIL-2 (1 mg/kg, s.c, days 0-4, 7-11) + BAY 43-9006 (30 mg/kg, p.o. days 0-8 ( ⁇ ).
  • Figure 10 shows efficacy of concomitant treatment of rIL-2 and SUl 1248 in the therapy in the murine RCC RENCA tumor model.
  • RENCA cells (1 x 10 6 cells) were implanted subcutaneously in the right flank of female BALB/c mice. Mice were randomized into groups when tumors were ⁇ 50 mm 3 , and treatments initiated on day 0.
  • Panel illustrates: vehicle ( ⁇ ) days 0-6; rIL-2 (1 mg/kg, s.c, days 0-6) ( ⁇ ) or SUl 1248 (40 mg/kg, p.o. days 0-6) (x) or combined rIL-2 (1 mg/kg, s.c, days 0-6) + SUl 1248 (40 mg/kg, p.o. days 0-6) (A).
  • cancers include, for example, CML, AML, breast, gastric, endometrial, salivary gland, adrenal, non-small cell lung, pancreatic, renal, rectal, skin, melanoma, multiple myeloma, brain/CNS, cervix, nasopharynx, malignant mesothelioma, hypopharynx, gastroinstestinal carcinoid, peritoneum, omentum, mesentery, gallbladder, testis, esophageal, lung, thyroid, ovarian, peritoneal, prostate, head and neck, bladder, colon, colorectal, lymphomas, and glioblastomas.
  • the methods described herein are useful in the treatment of any such cancer.
  • Therapy with a combination of aldesleukin and at least one antiangiogenic agent in the manner set forth herein causes a physiological response that is beneficial with respect to treatment of cancers whose unabated proliferating cells are highly dependent on vascularization, such as VEGF.
  • One embodiment of the invention provides a method of treating a cancer patient suffering from hypotension from the administration of aldesleukin, comprising: co-administering to the patient a therapeutically effective amount of an antiangiogenic agent to attenuate hypotension. Another more particular embodiment thereof comprises amelioration of cancer in the patient.
  • Another embodiment of the invention provides a method of treating a cancer patient suffering from hypertension from the administration of an antiangiogenic agent, comprising: co-administering to the patient a therapeutically effective amount of aldesleukin to attenuate hypertension.
  • Another more particular embodiment thereof comprises amelioration of cancer in the patient.
  • the cancer is susceptible to inhibition of angiogenesis and/or immune stimulation.
  • said antiangiogenic agent is selected from 6,7-bis(2-methoxyethoxy)-N-(3- ethynylphenyl)quinazolin-4-amine, 6-(3 -morpholinopropoxy)-N-(3 -chloro-4- fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2-(dimethylamino)ethyl)-5-((5- fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl-lH-pyrrole-3-carboxamide, N-(4- chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin- 1 -amine, or 1 -(4-(2- (methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluoromethyl)phenyl
  • Another embodiment of the invention provides a method of treating a patient suffering from cancer comprising administering to said patient aldesleukin and a compound selected from 6,7-bis(2-methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4- amine, 6-(3-morpholinopropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin- 4-amine, N-(2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)- 2,4-diethyl- 1 H-pyrrole-3 -carboxamide, N-(4-chlorophenyl)-4-((pyridin-4- yl)methyl)phthalazin- 1 -amine, or 1 -(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)- 3-(4-ch
  • Another embodiment of the invention provides a method of increasing efficacy of aldesleukin in a cancer patient comprising first administering an antiangiogenic agent in a dose capable of inducing hypoxia in the patient then administering the aldesleukin.
  • Another embodiment provides a method of increasing efficacy of an antiangiogenic agent in a cancer patient comprising reducing nitric oxide synthase by administration of aldesleukin to the patient.
  • Another embodiment provides a method of increasing efficacy of an antiangiogenic agent in a cancer patient comprising administration aldesleukin to the patient wherein, nitric oxide synthase is thereby reduced by administration of aldesleukin to the patient.
  • said antangiogenic agent is selected from 6,7-bis(2-methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6-(3- morpholinopropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N- (2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl- 1 H-pyrrole-3 -carboxamide, N-(4-chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin- 1 - amine, or 1 -(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3 -(4-chloro-3 - (trifluoromethyl)phenyl)urea
  • Another embodiment of the invention provides a method for treating a patient suffering from cancer, comprising the steps of first administering to said patient a therapeutically effective amount of aldesleukin followed by administration of an antiangiogenic agent (such as) selected from 6,7-bis(2-methoxyethoxy)-N-(3- ethynylphenyl)quinazolin-4-amine, 6-(3-morpholinopropoxy)-N-(3-chloro-4- fluorophenyl)-7-methoxyquinazolm-4-amine, N-(2-(dimethylamino)ethyl)-5-((5- fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl-lH-pyrrole-3-carboxamide, N-(4- chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin-l -amine, or l-(4-(2- (methylcarbamoyl)
  • Another embodiment of the invention provides a method for treating a patient suffering from cancer, comprising the steps of first administering to said patient a therapeutically effective amount of an antiangiogenic agent, such as, 6,7-bis(2- methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6-(3-morpholinopropoxy)- N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2- (dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl-lH- pyrrole-3 -carboxamide, N-(4-chloro ⁇ henyl)-4-((pyridin-4-yl)methyl)phthalazin- 1 - amine, or l-(4-(2-(methylcarbamoyl)pyridin-4-yl
  • Another embodiment of the invention provides a method for treating a patient suffering from cancer, comprising separately administering to said patient a therapeutically effective amount of an antiangiogenic agent and aldesleukin according to a dosing schedule, wherein the aldesleukin is administered from 1 to 3 times daily in a dose between about 9 and about 130 MIU/day for a period of at least 3 consecutive days, optionally followed by a rest period of at least 3 consecutive days.
  • said antiangiogenic agent is administered from 1 to 6 times every 2-3 weeks.
  • said antiangiogenic agent is a VEGF inhibitor.
  • the antiangiogenic agent is selected from 6,7-bis(2-methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6-(3- morpholinopropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N- (2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl- 1 H-pyrrole-3 -carboxamide, N-(4-chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin- 1 - amine, or 1 -(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluoromethyl)phenyl)urea
  • the aldesleukin is administered subcutaneously and said rest period is absent.
  • the aldesleukin is administered 3 times daily in a dose of about 30-100 MIU/day.
  • the aldesleukin is administered for a period of 5 consecutive days followed by a 9-day rest period.
  • said dosing schedule is repeated for at least two courses.
  • each course consists of 2-5 day treatments followed by a rest period of 9-15 days.
  • the aldesleukin is administered 3 times for the first day and once daily each proceeding day.
  • said dosing schedule is repeated for at least two courses, or 3 courses, or 4 courses, or 5 courses, or 6 courses, or 7 courses, or 8 courses, or 9 courses, or 10 courses.
  • the cancer is colon cancer, renal cell carcinoma or malignant melanoma.
  • At least one compound selected from acetaminophen, meperidine, indomethacin, ranitidine, nizatidine, diastop, loperamide, diphenhydramine, or furosemide is also administered to said patient.
  • said antangiogenic agent is 6,7-bis(2-methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4- amine, 6-(3 -morpholinopropoxy)-N-(3 -chloro-4-fluorophenyl)-7-methoxyquinazolin- 4-amine, N-(2-(dimethylammo)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)- 2,4-diethyl- 1 H-pyrrole-3 -carboxamide, N-(4-chlorophenyl)-4-((pyridin-4- yl)methyl)phthalazin-l -amine, or l-(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)- 3 -(4-chloro-3 -(trifluoromethyl
  • onfffirin p from cancer comprising administering aldesleukin and: a tautomer thereof, a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable salt of the tautomer.
  • Ri is alkyl, -aryl(Rj) p , or heterocyclyl;
  • R 2 is H or alkyl; or,
  • Ri and R 2 are bound together to form R] -2 ;
  • R 3 is H, -CN, -OH, halogen, alkyl, aryl, alkoxy, -NR a R b , -C(O)R c , -S(O) n Ra, or heterocyclyl;
  • R 4 Is H, -CN, -OH, halogen, alkyl, aryl, -0-(CH 2 ) q -R g , -O-(CH 2 ) q -O-Re, -NR 3 R b , -S(O) n R d , or -heterocyclyl-R f ;
  • R 5 is H, -CN, -OH, halogen, alkyl, aryl, -0-(CH 2 ) q -R g , -O-(CH 2 ) q -O-Re, -NR 3 R b ,
  • R 6 is H, -CN, -OH, halogen, alkyl, aryl, alkoxy, -NR a R b , -C(O)R c , -S(O) n R d , or heterocyclyl
  • R 7 is H, -OH, halogen, alkyl, aryl, alkoxy, -NR 3 R b , -S(O) n R d , or heterocyclyl
  • each R a and R b is independently H, alkyl, -C(O)R 0 , aryl, heterocyclyl, or alkoxy; or,
  • R 3 and R b are bound together to form Ri -2 ; each R 0 is independently H, alkyl, alkoxy, -C(O)alkyl, -C(O)aryl, -CHO, aryl, or heterocyclyl; each Rj is independently H, alkyl, alkenyl, aryl, or -NR 3 R b ; each R 3 is independently H or alkyl;
  • Rf is H, halogen, -OH, -CN, -(CH 2 ) q NR a R h , alkoxy, -C(O)R 0 , -(CH 2 ) q CH 3 .
  • each Rg is independently H, halogen, -C(O)R 0 , aryl, heterocyclyl, or -NR a R b ;
  • R h is H, or -(CH 2 ) q S(O) n Rd; each Rj is independently H, halo, alkyl, alkenyl, alkynyl, or -0(CH 2 ) q -R g ; each n is independently 0, 1, or 2; each p is independently 0, 1, 2, or 3; each q is independently 0, 1, or 2; Ri -2 has the general structure as shown:
  • said compound is:
  • said compound is:
  • R- 2U R 22 , R 23 , and R 24 are each independently H, -CN, -OH, halogen, alkyl, aryl, alkoxy, -NR a R b , -C(O)R 2 , -S(O) n R d , or heterocyclyl;
  • R 25 is H, alkyl, or -C(O)R 2 ;
  • R 2 6 and R 27 are each independently H, -OH, halogen, alkyl, -NR a R b , -C(O)R 2 , or
  • R 28 is H, -CH 3 , or halogen; each R a and R b is independently H, alkyl, -C(O)R 2 , aryl, heterocyclyl, or alkoxy; each R 2 is independently H, alkyl, alkoxy, -NH 2 , -NH(alkyl), -N(alkyl) 2 , aryl, or heterocyclyl; each Ra is independently H, alkyl, alkenyl, aryl, or -NR a Rt,; each n is independently O, 1 , or 2; and each q is independently O, 1 , or 2; or a tautomer thereof, a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable salt of the tautomer.
  • said compound is:
  • said compound is:
  • t au t omer thereof a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable salt of the tautomer.
  • Another embodiment of the invention provides a method of decreasing the toxicity associated with the administration of aldesleukin to a cancer patient comprising administering a quinolinone, preferably an antiangiogenic agent, preferably selected from 6,7-bis(2-methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4- amine, 6-(3-morpholinopropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin- 4-amine, N-(2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)- 2,4-diethyl- 1 H-pyrrole-3 -carboxamide, N-(4-chlorophenyl)-4-((pyridin-4- yl)methyl)phthalazin- 1 -amine, or 1 -(4-(2-(methylcarbamoyl)
  • Another embodiment of the invention provides a method of decreasing the toxicity associated with the administration of an antiangiogenic agent, preferably selected from 6,7-bis(2-methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6- (3-mo ⁇ holinopropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl- lH-pyrrole-3-carboxamide, N-(4-chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin-l- amine, or 1 -(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (
  • Another embodiment of the invention provides a method of decreasing IL-2 resistance in a patient suffering from cancer comprising administering an antiangiogenic agent, preferably selected from 6,7-bis(2-methoxyethoxy)-N-(3- ethynylphenyl)quinazolin-4-amine, 6-(3 -morpholinopropoxy)-N-(3 -chloro-4- fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2-(dimethylamino)ethyl)-5-((5- fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl-lH-pyrrole-3-carboxamide, N-(4- chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin-l -amine, or l-(4-(2- (methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-
  • Another embodiment of the invention provides a method for treating a patient suffering from cancer, comprising the steps of first administering to said patient a therapeutically effective amount of aldesleukin followed by administration of an antiangiogenic agent, preferably selected from 6,7-bis(2-methoxyethoxy)-N-(3- ethynylphenyl)quinazolin-4-amine, 6-(3 -morpholinopropoxy) -N-(3 -chloro-4- fluoiOphenyl)-7-methoxyquinazolin-4-amine, N-(2-(dimethylamino)ethyl)-5-((5- fiuoro-2-oxoindolin-3 -ylidene)methyl)-2,4-diethyl- 1 H-pyrrole-3 -carboxamide, N-(4-
  • Another embodiment of the invention provides a method for treating a patient suffering from cancer, comprising separately administering to said patient a therapeutically effective amount of an antiangiogenic agent, preferably selected from 6,7-bis(2-methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6-(3- mo ⁇ holino ⁇ ropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N- (2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl- lH- ⁇ yrrole-3-carboxamide, N-(4-chlorophenyl)-4-((pyridm-4-yl)methyl)phthalazin-l- amine, or 1 -(4-(2-(methylcarbamoyl)pyridin-4-y
  • the dose is between about 30 and about 100 MIU/day. More specifically the dose is between about 30 and about 60 MIU/day. More specifically the dose is between about 30 and about 40 MIU/day. More specifically the dose is between about 17 and about 30 MIU/day. More specifically the dose is between about 9 and about 30 MIU/day.
  • said antiangiogenic agent preferably selected from 6,7-bis(2-methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6- (3-morpholinopropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl- lH- ⁇ yrrole-3-carboxamide, N-(4-chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin-l- amine, or l-(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3-
  • said antiangiogenic agent preferably selected from 6,7- bis(2-methoxyethoxy)-N-(3 -ethynylphenyl)quinazolin-4-amine, 6-(3 - morpholinopropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N- (2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl- lH-pyrrole-3-carboxamide, N-(4-chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin-l- amine, or 1 -(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)
  • the antiangiogenic agent preferably selected from 6,7- bis(2-methoxyethoxy)-N-(3-ethynyl ⁇ henyl)quinazolin-4-amine, 6-(3- morpholinopropoxy)-N-(3-chloro-4-fluoro ⁇ henyl)-7-methoxyquinazolin-4-amine, N- (2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl- 1 H-pyrrole-3 -carboxamide, N-(4-chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin- 1 - amine, or l-(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluoromethyl)phenyl)ure
  • said bevacizumab is administered in a dose between 1 and 12 mg/kg once every 12-16 days.
  • the antiangiogenic agent is administered within 72 hours of the administration of rIL-2; wherein "within” is meant to indicate before or after, as in: the antiangiogenic agent is administered 72 hours before or 72 hours after the administration of rIL-2.
  • the antiangiogenic agent is administered within 14 days of the administration of rIL-2.
  • the antiangiogenic agent is administered within 7 days of the administration of rIL-2.
  • the antiangiogenic agent is administered within 6 days of the administration of rIL-2.
  • the antiangiogenic agent is administered within 5 days of the administration of rIL-2.
  • the antiangiogenic agent is administered within 4 days of the administration of rIL-2. In another embodiment, the antiangiogenic agent is administered within 48 hours of the administration of rIL-2. In another embodiment, the antiangiogenic agent is administered within 36 hours of the administration of rIL-2. In another embodiment, the antiangiogenic agent is administered within 24 hours of the administration of rlL- 2. In another embodiment, the antiangiogenic agent is administered within 12 hours of the administration of rIL-2. In another embodiment, the antiangiogenic agent is administered within 6 hours of the administration of rIL-2. In another embodiment, the antiangiogenic agent is administered within 1 hour of the administration of rIL-2.
  • the antiangiogenic agent is administered at the same time as the administration of rIL-2. In another embodiment, the antiangiogenic agent is administered in combination with rIL-2. In a more particular embodiment thereof the antiangiogenic agent is selected from 6,7-bis(2-methoxyethoxy)-N-(3 ⁇ ethynylphenyl)quinazolin-4-amine, 6-(3-morpholinopropoxy)-N-(3-chloro-4- fiuorophenyl)-7-methoxyquinazolin-4-amine, N-(2-(dimethylamino)ethyl)-5-((5- fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl-lH-pyrrole-3-carboxamide, N-(4-
  • Another embodiment of the invention provides a method of treating a patient suffering from cancer comprising co-administering aldesleukin and an antiangiogenic agent, preferably selected from 6,7-bis(2-methoxyethoxy)-N-(3- ethynylphenyl)quinazolin-4-amine, 6-(3-morpholinopropoxy)-N-(3-chloro-4- fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2-(dimethylamino)ethyl)-5 ⁇ ((5- fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl-lH-pyrrole-3-carboxamide, N-(4- chloro ⁇ henyl) ⁇ 4-((pvridin-4-yl)methyl)phthalazin-l -amine, or l-(4-(2- (methylcarbainoyl)pyridin-4-yloxy)phenyl
  • Another embodiment of the invention provides a therapeutic package suitable for commercial sale for treating a patient suffering from cancer, comprising a container, a therapeutically effective amount of aldesleukin, and a therapeutically effective amount of an antiangiogenic agent, preferably selected from 6,7-bis(2- methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6-(3-mo ⁇ holinopropoxy)- N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2- (dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl-lH- pyrrole-3 -carboxamide, N-(4-chlorophenyl)-4-((pyridin-4-yl)methyl) ⁇ hthalazin- 1 - amine, or 1 -(4
  • a therapeutic package thereof wherein said patient is suffering from renal cell carcinoma.
  • a therapeutic package thereof further comprising written matter instructing that the patient receive treatment with aldesleukin prior to treatment with the antiangiogenic agent, preferably selected from 6,7-bis(2-methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6-(3- morpholinopropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N- (2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl- lH-pyrrole-3-carboxamide, N-(4-chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin-l- amine, or l-(4-(2-(methylcarbamoyl)pyr
  • Another embodiment provides a pharmaceutical composition
  • a pharmaceutical composition comprising a an antiangiogenic agent, preferably selected from 6,7-bis(2-methoxyethoxy)-N-(3- ethynylphenyl)quinazolin-4-amine, 6-(3-morpholinopropoxy)-N-(3-chloro-4-
  • Clear cell renal cell carcinoma is associated with increased vascularization, particularly VEGF expression.
  • VEGF is a protein that plays a crucial role in tumor angiogenesis (the formation of new blood vessels to the tumor) and maintaining established tumor blood vessels. It binds to specific receptors on blood vessels to stimulate extensions to existing blood vessels. Some but not all cases of RCCa have increased serum VEGF levels.
  • VEGF increases vascular permeability and treatment with anti- VEGF has been noted to lead to increased blood pressure of some patients. It is therefore likely that some of the major toxicities of anti-VEGF (e.g. hypertension) and rIL-2 (e.g. hypotension) would be mutually counteracted and lead to an overall improved therapeutic index if the combinations of the present invention were administered to patients suffering from cancer, such as RCCa.
  • anti-VEGF e.g. hypertension
  • rIL-2 e.g. hypotension
  • aldesleukin with bevacizumab or cetuximab yield a higher proportion and/or better durability of responses compared to the agents administered alone.
  • the particular aldesleukin dosing regimens disclosed herein provide for intermittent stimulation of natural killer (NK) cell activity and decreased risk of rIL-2- related side effects that can be associated with long term exposure to rIL-2 dosing.
  • NK natural killer
  • hypotension typically associated with rIL-2 dosing is offset by the administration of the antiangiogenic compositions of the present invention, since they are generally associated with hypertension.
  • hypoxia typically associated with VEGF inhibitors will increase sensitivity to rIL-2 treatment efficacy, while simultaneously providing palliative benefits as well.
  • Another embodiment of the invention provides the use of aldesleukin in the manufacture of a medicament for treating cancer, wherein said medicament is for
  • an antiangiogenic agent preferably selected from 6,7-bis(2-methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6- (3-morpholinopropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2-(dime1hylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl- 1 H-pyrrole-3 -carboxamide, N-(4-chloropb.enyl)-4-((pyridin-4-yl)methyl)phtb.alazin- 1 - amine, or l-(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (
  • said antiangiogenic agent preferably selected from 6,7-bis(2-methoxyethoxy)-N-(3- ethynylphenyl)quinazolin-4-amine, 6-(3-morpholinopropoxy)-N-(3-chloro-4- fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2-(dimethylamino)ethyl)-5-((5- fluoro-2-oxomdolin-3-ylidene)methyl)-2,4-diethyl-lH-pyrrole-3-carboxamide, N-(4- chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin- 1 -amine, or 1 ⁇ (4-(2- (methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro ⁇ 3- (trifluoromethyl)phenyl)urea is a
  • an antiangiogenic agent preferably selected from 6,7-bis(2-methoxyethoxy)-N-(3- ethynylphenyl)quinazolin-4-amine, 6-(3-morpholino ⁇ ropoxy)-N-(3-chloro-4- fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2-(dimethylamino)ethyl)-5-((5- fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl-lH-pyrrole-3-carboxamide, N-(4- chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin- 1 -amine, or 1 ⁇ (4-(2- (methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3-
  • Preferred molecules associated with angiogenesis, modulated (such as inhibition) by the compositions of the present invention include: Vascular Endothelial Growth Factor (VEGF), Fibroblast Growth Factor (FGF), Interleukin-8 (IL-8), Angiogenin, Angiotropin, Epidermal Growth Factor (EGF), Platelet Derived Endothelial Cell Growth Factor, Transforming Growth Factor a (TGF-a),
  • VEGF Vascular Endothelial Growth Factor
  • FGF Fibroblast Growth Factor
  • IL-8 Interleukin-8
  • Angiogenin Angiotropin
  • EGF Epidermal Growth Factor
  • TGF-a Transforming Growth Factor a
  • TGF-b Transforming Growth Factor b
  • Nitric Oxide a compound that modulates TGF-b
  • modulation such as potentiation
  • Thrombospondin, Angiostatin, and Endostatin is contemplated within the present invention.
  • Ri is alkyl, -aryl(RO P , or heterocyclyl
  • R 2 is H or alkyl
  • Ri and R 2 are bound together to form Ri -2 ;
  • R 3 is H, -CN, -OH, halogen, alkyl, aryl, alkoxy, -NR a R b , -C(O)Rc, -S(O) n Rd, or heterocyclyl;
  • R 4 Is H, -CN, -OH, halogen, alkyl, aryl, -0-(CH 2 ) q -R g , -0-(CH 2 ) C1 -O-R 6 , -NR a R b ,
  • R 5 is H, -CN, -OH, halogen, alkyl, aryl, -0-(CH 2 ) q -R g> -0-(CH 2 ) q -0-Re, -NR a R b , -S(O) n Rd, or heterocyclyl;
  • R 6 is H, -CN, -OH, halogen, alkyl, aryl, alkoxy, -NR 3 Rb, -C(O)R 0 , -S(O) n Rd, or heterocyclyl;
  • R 7 is H, -OH, halogen, alkyl, aryl, alkoxy, -NR 3 R b , -S(O) n R d , or heterocyclyl; each R a and R b is independently H, alkyl, -C(O)R 0 , aryl, heterocyclyl, or alkoxy; or, R a and R b are bound together to form Ri -2 ; each R 0 is independently H, alkyl, alkoxy, -C(O)alkyl, -C(O)aryl, -CHO, aryl, or heterocyclyl; each Rd is independently H, alkyl, alkenyl, aryl, or -NR a R b ; each R e is independently H or alkyl; each R f is H, halogen, -OH, -CN, -(CH 2 ) q NR a R h , alkoxy, -C(O)Rc
  • Rh is H, or -(CH 2 ) q S(O) n Rd; each Ri is independently H, halo, alkyl, alkenyl, alkynyl, or -0(CH 2 ) q -R g ; each n is independently O, 1, or 2; each p is independently O, 1 , 2, or 3;
  • Ri -2 has the general structure as shown:
  • R 2 is H.
  • Ri is -aryl(Ri) p .
  • Ri is -aryl(Rj) P; aryl within Rj is phenyl, p within Ri is 2 and both Ri groups within Ri are halo.
  • Ri is -aryl(Ri) p
  • aryl within Ri is phenyl
  • p within Ri is 1 and R
  • groups within Ri is alkynyl, preferably ethynyl.
  • Ri is -aryl(Rj) P) aryl within Ri is phenyl, p within Ri is 2, one R; group within Ri is halo and the other R; group within Ri is - O(CH 2 ) q -R g .
  • q within Ri is 1 and R g within R] is halophenyl.
  • Ri is -aryl(Ri) p
  • p within Ri is 1 and K ⁇ within Ri is alkynyl.
  • Ri and R 2 are bound together to form Ri -2 :
  • R 8 is H
  • X is N
  • R 9 is -C(O)NHR b
  • R b within R 9 is -phenyl-O-CH 2 (CH 3 ) 2
  • R 3 and R 6 are H.
  • R 4 is -O-(CH 2 ) q -R g .
  • q within R 4 is 1 and R g is H.
  • R 4 is -O-(CH 2 ) q -R g
  • R g within R 4 is heterocyclyl.
  • R 4 and R 5 are each -O- (CH 2 ) q -O-R e .
  • q within both R 4 and R 5 is 2 and R e within both R 4 and R 5 is methyl.
  • R 4 is -heterocyclyl-R f and R 5 is H.
  • said heterocyclyl within R 4 is furanyl.
  • R f within R 4 is -(CH 2 ) q NHR h .
  • R h is -(CH 2 ) q NHR h .
  • R 5 is -O-(CH 2 ) q -R g .
  • R g within R 5 is heterocyclyl.
  • R 7 is H.
  • R 3 , R 6 , and R 7 are all H. INDOLINONE
  • Ri i is alkyl, aryl or heterocyclyl
  • Ri 2 , Ri 3 Ri 4 , and Ri 5 are each independently H, -CN, -OH, halogen, alkyl, aryl, alkoxy, -NR a R b , -C(O)R 2 , -S(O) n Rd, or heterocyclyl; each R 3 and R b is independently H, alkyl, -C(O)R 2 , aryl, heterocyclyl, or alkoxy; each R z is independently H, alkyl, alkoxy, -NH 2 , -NH(alkyl), -N(alkyl) 2 , aryl, or heterocyclyl; each Rd is independently H, alkyl, alkenyl, aryl, or -NR 3 R b ", each n is independently 0, 1, or 2; and each q is independently 0, 1, or 2.
  • Rn is heterocyclyl.
  • Rn is Ri i a :
  • Ri 6 , Ri 7 and Rj 8 are each independently H, -CN, -OH, halogen, alkyl, aryl, alkoxy, - NR 3 Rb, -C(O)R 2 , -S(O) n R d , or heterocyclyl.
  • R 7 is -C(O)-(CH 2 ) p -N(H)( 2-r) (alkyl) r , wherein p is O, 1, 2, 3, 4, or 5 and r is 0, 1 , or 2.
  • Ri i is Ri ia
  • RB is F.
  • Ri i is R l la
  • Ri 7 is -(CH 2 ) t COOH, wherein t is 1, 2, 3, or 4.
  • Rn is R l la
  • R 6 and R 8 are both methyl.
  • Rn is Rn 3
  • Ri 7 is H.
  • Ri i is aryl. More particular still, Ri i is substituted or unsubstituted phenyl. In another embodiment R is iodide.
  • dotted line represents an optional placement of an additional bond
  • R 2 I, R 22 , R 23 , and R 24 are each independently H, -CN, -OH, halogen, alkyl, aryl, alkoxy, -NR 3 Rb, -C(O)R 2 , -S(O) n Rd, or heterocyclyl;
  • R 25 is H, alkyl, or -C(O)R 2 ;
  • R 2 6 and R 27 are each independently H, -OH, halogen, alkyl, -NR 3 R b , -C(O)R 2 , or
  • R 28 is H, -CH 3 , or halogen; each R 3 and Rb is independently H, alkyl, -C(O)R 2 , aryl, heterocyclyl, or alkoxy;
  • references to a certain element such as hydrogen or H is meant to include all isotopes of that element.
  • an R group is defined to include hydrogen or H, it also includes deuterium and tritium.
  • antiangiogenic agent any small molecule, more specifically, any compound having a molecular weight less than 1,100 g/mol that has been shown or will be shown to suppress angiogensis in a system.
  • VEGF Vascular Endothelial Growth Factor
  • FGF Fibroblast Growth Factor
  • IL-8 Interleukin-8
  • Angiogenin Angiotropin
  • EGF Epidermal Growth Factor
  • EGF Platelet Derived Endothelial Cell Growth Factor
  • TGF-a Transforming Growth Factor b
  • Nitric Oxide Nitric Oxide
  • compositions of the present invention include: 6,7-bis(2-methoxyethoxy)-N-(3 -ethynylphenyl)quinazolin-4-amine, 6-(3 - mo ⁇ holinopropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N- (2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl- 1 H- ⁇ yrrole-3-carboxamide, N-(4-chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin- 1 - amine, and l-(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluoromethyl)phenyl)urea.
  • an effective amount of a compound to treat renal cell carcinoma may be an amount necessary to cause regression of tumor growth in renal cells.
  • the effective amount may vary, depending, for example, upon the condition treated, weight of the subject and severity of the disease. One of skill in the art can readily determine the effective amount empirically without undue experimentation.
  • an effective amount for treatment refers to an amount sufficient to palliate, ameliorate, stabilize, reverse, slow or delay progression of a condition such as a disease state.
  • a “subject” or “patient” is meant to describe a human or vertebrate animal including a dog, cat, pocket pet, marmoset, horse, cow, pig, sheep, goat, elephant, giraffe, chicken, lion, monkey, owl, rat, squirrel, slender loris, and mouse.
  • a “pocket pet” refers to a group of vertebrate animals capable of fitting into a commodious coat pocket such as, for example, hamsters, chinchillas, ferrets, rats, guinea pigs, gerbils, rabbits and sugar gliders.
  • pharmaceutically acceptable ester refers to esters, which hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof. Suitable ester groups include, for example, those derived from pharmaceutically acceptable aliphatic carboxylic acids, particularly alkanoic, alkenoic, cycloalkanoic and alkanedioic acids, in which each
  • esters include, but are not limited to, formates, acetates, propionates, butyrates, acrylates and ethylsuccinates.
  • the compounds of the present invention can be used in the form of salts as in "pharmaceutically acceptable salts" derived from inorganic or organic acids.
  • These salts include but are not limited to the following: acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphorsulfonate, digluconate, cyclopentanepropionate, dodecylsulfate, ethanesulfonate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, fumarate, hydrochloride, hydrobromide, hydroiodide,
  • 2-hydroxyethanesulfonate lactate, maleate, methanesulfonate, nicotinate, 2-napth- alenesulfonate, oxalate, pamoate, pectinate, sulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, p-toluenesulfonate and undecanoate.
  • the basic nitrogen-containing groups can be quaternized with such agents as loweralkyl halides, such as methyl, ethyl, propyl, and butyl chloride, bromides, and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides, and others. " Water or oil-soluble or dispersible products are thereby obtained.
  • loweralkyl halides such as methyl, ethyl, propyl, and butyl chloride, bromides, and iodides
  • dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates
  • long chain halides
  • prodrugs refers to those prodrugs of the compounds of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals with undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the invention.
  • prodrug refers to compounds that are rapidly transformed in vivo to yield the parent compound of the above formula, for example by hydrolysis in blood. A thorough discussion is provided in T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems, Vol. 14 of the A.C.S. Symposium Series, and in Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical
  • halo refers to F, Cl, Br, or I atoms.
  • alkyl refers to substituted and unsubstituted alky groups such as methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, undecyl, dodecyl and the like.
  • the phrase also includes branched chain isomers of straight chain alkyl groups, including but not limited to, the following which are provided by way of example: -CH(CH 3 ) 2 , -CH(CH 3 )(CH 2 CH 3 ), -CH(CH 2 CH 3 ) 2 , -C(CH 3 ) 3 , -C(CH 2 CH 3 ) 3) -CH 2 CH(CH 3 ) 2 , -CH 2 CH(CH 3 )(CH 2 CH 3 ), -CH 2 CH(CH 2 CH 3 ) 2 , -CH 2 C(CH 3 ) 3 , -CH 2 C(CH 2 CH 3 ) 3 , -CH(CH 3 )CH(CH 3 )(CH 2 CH 3 ), -CH 2 CH 2 CH(CH 3 ) 2 , -CH 2 CH 2 CH(CH 3 )(CH 2 CH 3 ), -CH 2 CH 2 CH(CH 3 )S, -CH 2 CH 2 C(CH 3 ) 3 ,
  • the phrase also includes cyclic alkyl groups such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl and such rings substituted with straight and branched chain alkyl groups as defined above.
  • cyclic alkyl groups such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl and such rings substituted with straight and branched chain alkyl groups as defined above.
  • polycyclic alkyl groups such as, but not limited to, adamantyl norbornyl, and bicyclo[2.2.2]octyl and such rings substituted with straight and branched chain alkyl groups as defined above.
  • alkyl also includes groups in which one or more bonds to a carbon(s) or hydrogen(s) are replaced by a bond to non-hydrogen and non-carbon atoms such as, but not limited to, a halogen atom in halides such as F, Cl, Br, and I; and oxygen atom in groups such as hydroxyl groups, alkoxy groups, aryloxy groups, and ester groups; a sulfur atom in groups such as thiol groups, alkyl and aryl sulfide groups, sulfone groups, sulfonyl groups, and sulfoxide groups; a nitrogen atom in groups such as amines, amides, alkylamines, dialkylamines, arylamines, alkylarylamines, diarylamines, N-oxides, imides, and enamines; a silicon atom in groups such as in trialkylsilyl groups, dialkylarylsilyl groups, alkyldiarylsilyl groups
  • aryl refers to substituted and unsubstituted aryl groups that do not contain heteroatoms.
  • the phrase includes, but is not limited to, groups such as phenyl, biphenyl, anthracenyl, naphthenyl by way of example.
  • Aryl groups also include those in which one of the aromatic carbons is bonded to a non-carbon or non- hydrogen atoms described above (in the alkyl definition) and also includes aryl groups in which one or more aromatic carbons of the aryl group is bonded to a substituted
  • aryl includes, but is not limited to tolyl, and hydroxyphenyl among others.
  • Alkenyl groups are those limited to having 2 to 15 carbon atoms and as many as 4 additional heteroatoms as described above. More preferred alkenyl groups have from 2 to 5 carbon atoms and as many as 2 heteroatoms.
  • alkoxy refers to substituted or unsubstituted alkoxy groups of the formula -O-alkyl, wherein the point of attachment is the oxy group and the alkyl group is as defined above.
  • Alkoxy groups are those limited to having 1 to 20 carbon atoms and as many as 5 additional heteroatoms, including the oxygen atom. More preferred alkoxy groups have from 1 to 5 carbon atoms and as many as 2 heteroatoms, including the oxygen atom.
  • alkynyl groups in which a non-carbon or non-hydrogen atom is bonded to a carbon triple bonded to another carbon and those in which a non-carbon or non-hydrogen atom is bonded to a carbon not involved in a triple bond to another carbon.
  • Alkynyl groups are those limited to having 2 to 15 carbon atoms and as many as 4 additional heteroatoms as described
  • More preferred alkynyl groups have from 2 to 5 carbon atoms and as many as 2 heteroatoms.
  • heterocyclyl refers to both aromatic and nonaromatic ring compounds including monocyclic, bicyclic, and polycyclic ring compounds such as, but not limited to, quinuclidyl, containing 3 or more ring members of which one or more is a heteroatom such as, but not limited to, N, O, and S.
  • heterocyclyl groups include, but are not limited to: unsaturated 3 to 8 membered rings containing 1 to 4 nitrogen atoms such as, but not limited to pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, pyridyl, dihydropyridyl, pyrimidyl, pyrazinyl, pyridazinyl, triazolyl (e.g. 4H-l,2,4-triazolyl, lH-l,2,3-triazolyl, 2H-l,2,3-triazolyl etc.), tetrazolyl, (e.g.
  • saturated 3 to 8 membered rings containing 1 to 4 nitrogen atoms such as, but not limited to, pyrrolidinyl, imidazolidinyl, piperidinyl, piperazinyl; condensed unsaturated heterocyclic groups containing 1 to 4 nitrogen atoms such as, but not limited to, indolyl, isoindolyl, indolinyl, indolizinyl, benzimidazolyl, quinolyl, isoquinolyl, indazolyl, benzotriazolyl; unsaturated 3 to 8 membered rings containing 1 to 2 oxygen atoms such as, but not limited to furanyl; unsaturated 3 to 8 membered rings containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms such as, but not limited to, oxazolyl, isoxazolyl, oxadiazolyl (e.g.
  • unsaturated 3 to 8 membered rings containing 1 to 3 sulfur atoms and 1 to 3 nitrogen atoms such as, but not limited to, thiazolyl, isothiazolyl, thiadiazolyl (e.g.
  • Ri i is alkyl, aryl or heterocyclyl
  • Ri 2 , Ri 3 Ri 4 , and R 15 are each independently H, -CN, -OH, halogen, alkyl, aryl, alkoxy, -NR a R b , -C(O)R 2 , -S(O) n Rd, or heterocyclyl; each R a and Rb is independently H, alkyl, -C(O)R 2 , aryl, heterocyclyl, or alkoxy; each R z is independently H, alkyl, alkoxy, -NH 2 , -NH(alkyl), -N(alkyl) 2 , aryl, or heterocyclyl; each Rd is independently H, alkyl, alkenyl, aryl, or -NR a R b ; each n is independently 0, 1, or 2; and each q is independently 0, 1, or 2; or a tautomer thereof, a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable salt of the tautomer.
  • Ri 1 is Rj i a :
  • Ri 6, Ri 7 and Ri 8 are each independently H, -CN, -OH, halogen, alkyl, aryl, alkoxy, - NR 3 Rb, -C(O)R 2 , -S(O) n Rd, or heterocyclyl.
  • said compound is:
  • oxygen atoms such as benzodioxolyl (e.g. 1,3-benzodioxoyl, etc.); unsaturated 3 to 8 membered rings containing an oxygen atom and 1 to 2 sulfur atoms such as, but not limited to, dihydrooxathiinyl; saturated 3 to 8 membered rings containing 1 to 2 oxygen atoms and 1 to 2 sulfur atoms such as 1,4-oxathiane; unsaturated condensed rings containing 1 to 2 sulfur atoms such as benzothienyl, benzodithiinyl; and unsaturated condensed heterocyclic rings containing an oxygen atom and 1 to 2 oxygen atoms such as benzoxathiinyl.
  • benzodioxolyl e.g. 1,3-benzodioxoyl, etc.
  • unsaturated 3 to 8 membered rings containing an oxygen atom and 1 to 2 sulfur atoms such as, but not limited to, dihydroo
  • Heterocyclyl group also include those described above in which one or more S atoms in the ring is double-bonded to one or two oxygen atoms (sulfoxides and sulfones).
  • heterocyclyl groups include tetrahydrothiophene, tetrahydrothiophene oxide, and tetrahydrothiophene 1,1- dioxide.
  • Preferred heterocyclyl groups contain 5 or 6 ring members.
  • More preferred heterocyclyl groups include morpholine, piperazine, piperidine, pyrrolidine, imidazole, pyrazole, 1,2,3-triazole, 1,2,4-triazole, tetrazole, thiomorpholine, thiomorpholine in which the S atom of the thiomorpholine is bonded to one or more O atoms, pyrrole, homopiperazine, oxazolidin-2-one, pyrrolidin-2-one, oxazole, quinuclidine, thiazole, isoxazole, furan, and tetrahydrofuran.
  • Heterocyclyl also refers to those groups as defined above in which one of the ring members is bonded to a non-hydrogen atom such as described above with respect to substituted alkyl groups and substituted aryl groups. Examples, include, but are not limited to, 2- methylbenzimidazolyl, 5-methylbenzimidazolyl, 5-chlorobenzthiazolyl, 1 -methyl piperazinyl, and 2-chloropyridyl among others. Heterocyclyl groups are those limited to having 2 to 15 carbon atoms and as many as 6 additional heteroatoms as described above. More preferred heterocyclyl groups have from 3 to 5 carbon atoms and as many as 2 heteroatoms.
  • substituted as applied to an undefined, yet well known in the art group, such as phenyl, will have the same meaning with respect to the optional appendages as described in the definition of alkyl.
  • the invention also includes isotopically-labeled compounds, that are structurally identical to those disclosed above, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulfur, fluorine and chlorine, such as 2 H, 3 H, 13 C, 14 C, 15 N, 18 0, 17 0, 31 P, 32 P, 35 S, 18 F and 36 Cl, respectively.
  • Compounds of the present invention, prodrugs thereof, and pharmaceutically acceptable salts of said compounds and of said prodrugs that contain the aforementioned isotopes and/or other isotopes of other atoms are within the scope of this invention.
  • Certain isotopically labeled compounds of the present invention, for example those into which radioactive isotopes such as H and C are incorporated, are useful in drug and/or substrate tissue distribution assays. Tritiated, i.e., 3 H, and carbon-14, i.e., 14 C, isotopes are particularly preferred for their ease of preparation and detectability.
  • isotopically labeled compounds of this invention and prodrugs thereof can generally be prepared by carrying out known or referenced procedures and by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent.
  • IL-2 or "interleukin-2” indicates a lymphocyte that is produced by normal peripheral blood lymphocytes and is present in the body at low concentrations.
  • IL-2 was first described by Morgan et al. (1976) Science 193:1007- 1008 and originally called T-cell growth factor because of it's ability to induce proliferation of stimulated T lymphocytes. It is a protein with a reported molecular weight in the range of 13,000 to 17,000 (Gillis and Watson (1980) J. Exp. Med. 159: 1709) and has an isoelectric point in the range of 6-8.5.
  • IL-2 is intended to encompass any source of IL-2, including mammalian sources such as, e.g., mouse, rat, rabbit, primate, pig, pocket pet, and human, and may be native or obtained by recombinant techniques, such as recombinant IL-2 polypeptides produced by microbial hosts.
  • the IL-2 may be the native polypeptide sequence, or can be a variant of the native IL-2 polypeptide as described herein below, so long as the variant IL-2 polypeptide retains the IL-2 biological activity of interest as defined herein.
  • the IL-2 polypeptide or variant thereof is derived from a human source, and includes human IL-2 that is , recombinantly produced, such as recombinant human IL-2 polypeptides produced by microbial hosts, and variants thereof that retain the IL-2 biological activity of interest.
  • Any pharmaceutical composition comprising IL-2 as a therapeutically active component can be used to practice the present invention.
  • compositions of the present invention may be administered in conjunction with other anticancer agents.
  • compositions will either be formulated together as a combination therapeutic or administered separately.
  • Anticancer agents for use with the invention include, but are not limited to, one or more of the following set forth below: A. Kinase Inhibitors
  • EGFR Epidermal Growth Factor Receptor
  • small molecule quinazolines for example gefitinib (US 5457105, US 5616582, and US 5770599), ZD-6474 (WO 01/32651), erlotinib (Tarceva®, US 5,747,498 and WO 96/30347), and lapatinib (US 6,727,256 and WO 02/02552); Vascular Endothelial Growth Factor Receptor (VEGFR) kinase inhibitors, including SU-11248 (WO 01/60814), SU 5416 (US 5,883,113 and WO 99/61422), SU 6668 (US 5,883,113 and WO 99/61422), CHIR-258 (US 6,605,617 and US 6,774,237), vatalanib or PTK-787 (US 6,258,812), VEGF-787 (US 6,258,812), VEGF-787 (US 6,258,812), VEGF-787 (US 6,
  • Estrogen-targeting agents for use in anticancer therapy in conjunction with the compositions of the present invention include Selective Estrogen Receptor Modulators (SERMs) including tamoxifen, toremifene, raloxifene; aromatase inhibitors including Arimidex® or anastrozole; Estrogen Receptor Downregulators (ERDs) including Faslodex® or fulvestrant.
  • SERMs Selective Estrogen Receptor Modulators
  • ESDs Estrogen Receptor Downregulators
  • Androgen-targeting agents for use in anticancer therapy in conjunction with the compositions of the present invention include flutamide, bicalutamide, finasteride, aminoglutethamide, ketoconazole, and corticosteroids.
  • inhibitors for use as anticancer agents in conjunction with the compositions of the present invention include protein farnesyl transferase inhibitors " including tipifarnib or R-115777 (US 2003134846 and WO 97/21701), BMS-214662, AZD-3409, and FTI-277; topoisomerase inhibitors including merbarone and diflomotecan (BN-80915); mitotic kinesin spindle protein (KSP) inhibitors including SB-743921 and MKI-833; protease modulators such as bortezomib or Velcade® (US 5,780,454), XL-784; and cyclooxygenase 2 (COX-2) inhibitors including nonsteroidal antiinflammatory drugs I (NSAIDs).
  • protein farnesyl transferase inhibitors including tipifarnib or R-115777 (US 2003134846 and WO 97/21701), BMS-214662, AZD-3409, and FTI-277
  • Alkylating agents for use in conjunction with the compositions of the present invention for anticancer therapeutics include VNP-40101M or cloretizine, oxaliplatin (US 4,169,846, WO 03/24978 and WO 03/04505), glufosfamide, mafosfamide, etopophos (US 5,041,424), prednimustine; treosulfan; busulfan; irofluven (acylfulvene); penclomedine; pyrazoloacridine (PD-115934); O6-benzylguanine; decitabine (5-aza-2-deoxycytidine); brostallicin; mitomycin C (MitoExtra); TLK-286 (Telcyta®); temozolomide; trabectedin (US 5,478,932); AP-5280 (Platinate formulation of Cisplatin); porfiromycin; and clearazide (meclorethamine).
  • Chelating agents for use in conjunction with the compositions of the present invention for anticancer therapeutics include tetrathiomolybdate (WO 01/60814); RP- 697; Chimeric T84.66 (cT84.66); gadofosveset (Vasovist®); deferoxamine; and bleomycin optionally in combination with electorporation (EPT).
  • tetrathiomolybdate WO 01/60814
  • RP- 697 Chimeric T84.66 (cT84.66); gadofosveset (Vasovist®); deferoxamine; and bleomycin optionally in combination with electorporation (EPT).
  • EPT electorporation
  • Biological response modifiers for use in conjunction with the compositions of the present invention for anticancer therapeutics include staurosprine and macrocyclic analogs thereof, including UCN-01, CEP-701 and midostaurin (see WO 02/30941, WO 97/07081, WO 89/07105, US 5,621,100, WO 93/07153, WO 01/04125, WO 02/30941, WO 93/08809, WO 94/06799, WO 00/27422, WO 96/13506 and WO 88/07045); squalamine (WO 01/79255); DA-9601 (WO 98/04541 and US 6,025,387); alemtuzumab; interferons (e.g.
  • interleukins specifically IL-2 or aldesleukin as well as IL-I, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL- 10, IL- 11 , IL- 12, and active biological variants thereof having amino acid sequences greater than 70% of the native human sequence; altretamine ( ⁇ exalen®); SU 101 or leflunomide (WO 04/06834 and US 6,331,555); imidazoquinolines such as resiquimod and imiquimod (US 4,689,338, 5,389,640, 5,268,376, 4,929,624, 5,266,575, 5,352,784, 5,494,916, 5,482,936, 5,346,905, 5,395,937, 5,238,944, and 5,525,612); and SMIPs, including benzazoles, anthraquinones, thiosemicarbazones, and tryptanthrins (WO 04
  • Anticancer vaccines for use in conjunction with the compositions of the present invention include Avicine® (Tetrahedron Letters 26, 1974 2269-70); oregovomab (OvaRex®); Theratope® (STn-KL ⁇ ); Melanoma Vaccines; GI-4000 series (GI-4014, GI-4015, and GI-4016), which are directed to five mutations in the Ras protein; GlioVax-1; MelaVax; Advexin® or INGN-201 (WO 95/12660); Sig/E7/LAMP-1, encoding ⁇ PV-16 E7; MAGE-3 Vaccine or M3TK (WO 94/05304); HER-2VAX; ACTIVE, which stimulates T-cells specific for tumors; GM-CSF cancer vaccine; and Listeria monocytogenes-based vaccines.
  • J. Antisense Therapy :
  • Anticancer agents for use in conjunction with the compositions of the present invention also include antisense compositions, such as AEG-35156 (GEM-640); AP- 12009 and AP-11014 (TGF-beta2-specific antisense oligonucleotides); AVI-4126; AVI-4557; AVI-4472; oblimersen (Genasense®); JFS2; aprinocarsen (WO 97/29780); GTI-2040 (R2 ribonucleotide reductase mRNA antisense oligo) (WO 98/05769); GTI-2501 (WO 98/05769); liposome-encapsulated c-Raf antisense oligodeoxynucleotides (LErafAON) (WO 98/43095); and Sirna-027 (RNAi-based therapeutic targeting VEGFR-I mRNA).
  • AEG-35156 GEM-640
  • IL-2 compound is "aldesleukin” or "Proleukin®", manufactured by Chiron Corporation of Emeryville, California.
  • the IL-2 in this formulation is a recombinantly produced, unglycosylated human IL-2 mutein which differs from the native human IL-2 amino acid sequence in having the initial alanine residue eliminated and the cysteine residue at position 125 replaced by a serine residue (referred to as des-alanyl-1, serine-125 human interleukin-2).
  • This IL-2 mutein can be expressed in E. coli, and subsequently purified by diafiltration and cation exchange chromatography as described in U.S. Patent No. 4,931,543.
  • Aldesleukin has been compared to native (Jurkat) IL-2 in vitro. No significant differences have been seen and in vivo induction of cytolytic cells in mice and serum half-life following IV administration is equivalent for aldesleukin and native (Jurkat) IL-2; although there are beneficial attributes of the form of IL-2 encompassed by aldesleukin and therefore reference to "aldesleukin" encompasses only that composition and not all possible forms of the IL-2 protein.
  • the pharmaceutical compositions useful in the methods of the invention may comprise biologically active variants of IL-2.
  • Such variants should retain the desired biological activity of the native polypeptide such that the pharmaceutical composition comprising the variant polypeptide has the same therapeutic effect as the pharmaceutical composition comprising the native polypeptide when administered to a subject. That is, the variant polypeptide will serve as a therapeutically active component in the pharmaceutical composition in a manner similar to that observed for the native polypeptide.
  • Methods are available in the art for determining whether a variant polypeptide retains the desired biological activity, and hence serves as a therapeutically active component in the pharmaceutical composition.
  • Biological activity can be measured using assays specifically designed for measuring activity of the native polypeptide or protein, including assays described in the present inventions.
  • antibodies raised against a biologically active native polypeptide can be tested for their ability to bind to the variant polypeptide, where effective binding is indicative of a polypeptide having a conformation similar to that of the native polypeptide.
  • the IL-2 biological activity of interest is the ability of IL-2 to activate and/or expand natural killer (NK) cells to mediate lymphokine activated killer (LAK) activity and antibody-dependent cellular cytotoxicity (ADCC).
  • NK natural killer
  • LAK lymphokine activated killer
  • ADCC antibody-dependent cellular cytotoxicity
  • an IL-2 variant for example, a mutein of human IL-2
  • Assays to determine IL-2 activation or expansion of NK cells and mediation of LAC or ADCC activity are well known in the art.
  • Suitable biologically active variants of native or naturally occurring IL-2 can be fragments, analogues, and derivatives of that polypeptide.
  • fragment is intended a polypeptide consisting of only a part of the intact polypeptide sequence and structure, and can be a C-terminal deletion or N-terminal deletion of the native polypeptide.
  • analogue is intended an analogue of either the native polypeptide or of a fragment of the native polypeptide, where the analogue comprises a native polypeptide sequence and structure having one or more amino acid substitutions, insertions, or deletions.
  • “Muteins”, such as those described herein, and peptides having one or more peptoids (peptide mimics) are also encompassed by the term analogue (see International Publication No. WO 91/04282).
  • “derivative” is intended any suitable modification of the native polypeptide of interest, of a fragment of the native polypeptide, or of their respective analogues, such as glycosylation, phosphorylation, polymer conjugation (such as with polyethylene glycol), or other addition of foreign moieties, so long as the desired biological activity of the native polypeptide is retained.
  • Methods for making polypeptide fragments, analogues, and derivatives are generally available in the art.
  • amino acid sequence variants of the polypeptide can be prepared by mutations in the cloned DNA sequence encoding the native polypeptide of interest.
  • Methods for mutagenesis and nucleotide sequence alterations are well known in the art. See, for example, Walker and Gaastra, eds. (1983) Techniques in Molecular Biology (MacMillan Publishing Company, New York); Kunkel (1985) Proc. Natl. Acad. Sci. USA 82:488-492; Kunkel et al. (1987) Methods Enzymol. 154:367-382; Sambrook et al. ( 1989) Molecular Cloning: A Laboratory Manual (Cold Spring
  • variants of the IL-2 polypeptide of interest modifications are made such that variants continue to possess the desired activity. Any mutations made in the DNA encoding the variant polypeptide must not place the sequence out of reading frame and preferably will not create complementary regions that could produce a secondary mRNA structure.
  • Biologically active variants of IL-2 will generally have at least 70%, preferably at least 80%, more preferably about 90% to 95% or more, and most preferably about 98% or more amino acid sequence identity to the amino acid sequence of the reference polypeptide molecule, which serves as the basis for comparison.
  • a biologically active variant thereof will have at least 70%, preferably at least 80%, more preferably about 90% to 95% or more, and most preferably about 98% or more sequence identity to the amino acid sequence for human IL-2.
  • a biologically active variant of a native polypeptide of interest may differ from the native polypeptide by as few as 1-15 amino acids, as few as 1-10, such as 6-10, as few as 5, as few as 4, 3, 2, or even 1 amino acid residue.
  • sequence identity is intended the same amino acid residues are found within the variant polypeptide and the polypeptide molecule that serves as a reference when a specified, contiguous segment of the amino acid sequence of the variants is aligned and compared to the amino acid sequence of the reference molecule.
  • the percentage sequence identity between two amino acid sequences is calculated by determining the number of positions at which the identical amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the segment undergoing comparison to the reference molecule, and multiplying the result by 100 to yield the percentage of sequence identity.
  • the determination of percent identity between any two sequences can be accomplished using a mathematical algorithm.
  • naturally or non-naturally occurring variants of IL-2 have amino acid sequences that are at least 70%, preferably 80%, more preferably, 85%, 90%, 91%, 92 %, 93%, 94% or 95% identical to the amino acid sequence to the reference molecule, for example, the native human IL-2, or to a shorter portion of the reference IL-2 molecule. More preferably, the molecules are 96%, 97%, 98% or 99% identical.
  • Percent sequence identity is determined using the Smith- Waterman homology search algorithm using an affined gap search with a gap open penalty of 12 and a gap extension penalty of 2, BLOSUM matrix of 62.
  • a variant may, for example, differ by as few as 1 to 10 amino acid residues, such as 6-10, as few as 5, as few as 4, 3, 2, or even 1 amino aid residue.
  • the contiguous segment of the variant amino acid sequence may have additional amino acid residues or deleted amino acid residues with respect to the reference amino acid sequence.
  • the contiguous segment used for comparison to the reference amino acid sequence will include at least twenty (20) contiguous amino acid residues, and may be 30, 40, 50, or more amino acid residues. Corrections for sequence identity associated with conservative residue substitutions or gaps can be made (see Smith-Waterman homology search algorithm).
  • polypeptide having IL-2 activity depends on a number of factors. As ionizable amino and carboxyl groups are present in the molecule, a particular polypeptide may be obtained as an acidic or basic salt, or in neutral form. All such preparations that retain their biological activity when placed in suitable environmental conditions are included in the definition of polypeptides having IL-2 activity as used herein.
  • the primary amino acid sequence of the polypeptide may be augmented by derivatization using sugar moieties (glycosylation) or by other supplementary molecules such as lipids, phosphate, acetyl groups and the like. It may also be augmented by conjugation with saccharides. Certain aspects of such augmentation are accomplished through post-translational processing systems of the producing host; other such modifications may be introduced in vitro. In any event, such modifications are included in the definition of an IL-2 polypeptide used herein so long as the IL-2 activity of the polypeptide is not destroyed. It is expected that such modifications may quantitatively or qualitatively affect the activity, either by enhancing or diminishing the activity of the polypeptide, in the various assays. Further, individual amino acid residues in the chain may be modified by oxidation, reduction, or other derivatization, and the polypeptide may be cleaved to obtain fragments that retain activity.
  • IL-2 variants are suitable for use as a therapeutically active component of a pharmaceutical composition used in the methods of the present invention.
  • the IL-2 or variants thereof for use in the methods of the present invention may be from any source, but preferably is recombinant IL-2.
  • recombinant IL-2 is intended interleukin-2 that has comparable biological activity to native-sequence IL-2 and that has been prepared by recombinant DNA techniques as described, for example, by Taniguchi et al.
  • IL-2 nucleic Acids Research 11 :4307-4323 or mutationally altered IL-2 as described by Wang et al. (1984) Science 224:1431-1433.
  • the gene coding for IL-2 is cloned and then expressed in transformed organisms, preferably a microorganism, and most preferably E. coli, as described herein.
  • the host organism expresses the foreign gene to produce IL-2 under expression conditions.
  • Synthetic recombinant IL-2 can also be made in eukaryotes, such as yeast or human cells. Processes for growing, harvesting, disrupting, or extracting the IL-2 from cells are substantially described in, for example, U.S. Patent Nos.
  • EP 136,489 (which discloses one or more of the following alterations in the amino acid sequence of naturally occurring IL-2: Asn26 to Gln26; Trpl21 to Phel21; Cys58 to Ser58 or Ala58, CyslO5 to SerlO5 or AlalO5; Cysl25 to Serl25 or Alal25; deletion of all residues following Arg 120; and the Met-1 forms thereof); and the recombinant IL-2 muteins described in European Patent Application No. 83306221.9, filed October 13, 1983 (published May 30, 1984 under Publication No. EP 109,748), which is the equivalent to Belgian Patent No. 893,016, and commonly owned U.S. Patent No.
  • 4,752,585 (which discloses the following variant IL-2 proteins: alalO4 serl25 IL- 2, alalO4 IL-2, alalO4 alal25 IL-2, vallO4 ser!25 IL-2, vallO4 IL-2, vallO4 alal25 IL-2, des-alal alalO4 serl25 IL-2, des-alal alalO4 IL-2, 10 des-alal alalO4 alal25 IL-2, des-alal vallO4 serl25 IL-2, des-alal vallO4 IL-2, des-alal vallO4 alal25 IL-2, des-alal des-pro2 alalO4 serl25 IL-2, des-alal des-pro2 alalO4 serl25 IL-2, des-alal des-pro2 alalO4 serl25 IL-2, des-alal des-pro2 alalO4 IL- 2, des-alal des-pro2 alalO4 aial25 IL
  • Patent No. 4,931 ,543 (which discloses the IL-2 mutein des-alanyl- 1 , serine 30 125 human IL-2 used in the examples herein, as well as the other IL-2 muteins). Also see European Patent Publication No. EP 200,280 (published December 10, 1986), which discloses recombinant IL-2 muteins wherein the methionine at position 104 has been replaced by a conservative amino acid.
  • Examples include the following muteins: ser4 des-ser5 alalO4 IL-2; des- alal des-pro2 des-thr3 des-ser4 des-ser5 alalO4 alal25 IL-2; des-alal des-pro2 des-thr3 des-ser4 des-ser5 glulO4 serl25 IL-2; des-alal des-pro2 des-thr3 des ser4 des-ser5 glulO4 IL-2; des-alal des-pro2 des-thr3 des-ser4 des-ser5 glul04 alal25 IL-2; des-alal des-pro2 des-thr3 des-ser4 des-ser5 des-ser6 alalO4 alal25 IL-2; des-alal des-pro2 des-thr3 des-ser4 des-ser5 des-ser6 alalO4 alal25 IL-2; des-alal des-pro2 des-thr3 des-ser4 des
  • IL-2 muteins include the those disclosed in WO 99/60128 (substitutions of the aspartate at position 20 with histidine or isoleucine, the asparagine at position 88 with arginine, glycine, or isoleucine, or the glutamine at positionl26 with leucine or gulatamic acid), which reportedly have selective activity for high affinity IL-2 receptors expressed by cells expressing T cell receptors in preference to NK cells and reduced IL-2 toxicity; the muteins disclosed in U.S Patent No. 5,229, 109
  • WO 00/04048 (corresponding to the first 30 amino acids of IL-2, which contains the entire a-helix A of IL-2 and interacts with the b chain of the IL-2 receptor), which reportedly stimulates NK cells and induction of LAK cells; and a mutant form of the IL-2 pl-30 peptide also disclosed in WO 00/04048 (substitution of aspartic acid at position 20 with lysine), which reportedly is unable to induce vascular bleeds but remains capable of generating LAK cells.
  • IL-2 can be modified with polyethylene glycol to provide enhanced solubility and an altered pharmokinetic profile (see U.S. Patent No. 4,766,106).
  • IL-2 as used herein is also intended to include IL-2 fusions or conjugates comprising IL-2 fused to a second protein or covalently conjugated to polyproline or a water soluble polymer to reduce dosing frequencies or to improve IL- 2 tolerability.
  • the IL-2 (or a variant thereof as defined herein) can be fused to human albumin or an albumin fragment using methods known in the art (see WO 01/79258).
  • the IL-2 can be covalently conjugated to polyproline or polyethylene glycol homopolymers and polyoxyethylated polyols, wherein the homopolymer is unsubstituted or substituted at one end with an alkyl group and the poplyol is unsubstituted, using methods known in the art (see, for example, U.S.
  • compositions comprising IL-2 as the therapeutically active component can be used in the methods of the invention.
  • Such pharmaceutical compositions are known in the art and include, but are not limited to, those disclosed in U.S. Patent Nos. 4,745,180; 4,766,106; 4,816,440; 4,894,226; 4,931 ,544; and 5,078,997.
  • liquid, lyophilized, or spray- dried compositions comprising IL-2 or variants thereof that are known in the art may be prepared as an aqueous or nonaqueous solution or suspension for subsequent administration to a subject in accordance with the methods of the invention.
  • compositions will comprise IL-2 or variants thereof as a therapeutically or prophylactically active component.
  • therapeutically or prophylactically active component is intended the IL-2 or variants thereof is specifically incorporated into the composition to bring about a desired therapeutic or prophylactic response with regard to treatment, prevention, or diagnosis of a disease or condition within a subject when the pharmaceutical composition is administered to that subject.
  • the pharmaceutical compositions comprise appropriate stabilizing agents, bulking agents, or both to minimize problems associated with loss of protein stability and biological activity during preparation and storage.
  • the IL-2 containing pharmaceutical compositions useful in the methods of the invention are compositions comprising stabilized monomelic IL-2 or variants thereof, compositions comprising multimeric IL-2 or variants thereof, and compositions comprising stabilized lyophilized or spray-dried IL-2 or variants thereof.
  • compositions comprising stabilized monomeric IL-2 or variants thereof are disclosed in International Publication No. WO 01/24814, entitled “Stabilized Liquid Polypeptide-Containing Pharmaceutical Compositions.”
  • IL-2 is intended the protein molecules are present substantially in their monomer form, not in an aggregated form, in the pharmaceutical compositions described herein. Hence covalent or hydrophobic oligomers or aggregates of IL-2 are not present.
  • the IL-2 in these liquid compositions is formulated with an amount of an amino acid base sufficient to decrease aggregate formation of IL-2 during storage.
  • the amino acid base is an amino acid or a combination of amino acids, where any given amino acid is present either in its free base form or in its salt form.
  • Preferred amino acids are selected from the group consisting of arginine, lysine, aspartic acid, and glutamic acid.
  • compositions further comprise a buffering agent to maintain phi of the liquid compositions within an acceptable range for stability of IL-2, where the buffering agent is an acid substantially free of its salt form, an acid in its salt form, or a mixture of an acid and its salt fond.
  • the acid is selected from the group consisting of succinic acid, citric acid, phosphoric acid, and glutamic acid.
  • Such compositions are referred to herein as stabilized monomeric IL-2 pharmaceutical compositions.
  • the amino acid base in these compositions serves to stabilize the IL-2 against aggregate formation during storage of the liquid pharmaceutical composition, while use of an acid substantially free of its salt form, an acid in its salt form, or a mixture of an acid and its salt form as the buffering agent results in a liquid composition having an osmolality that is nearly isotonic.
  • the liquid pharmaceutical composition may additionally incorporate other stabilizing agents, more particularly methionine, a nonionic surfactant such as polysorbate 80, and EDTA, to further increase stability of the polypeptide.
  • Such liquid pharmaceutical compositions are said to be stabilized, as addition of amino acid base in combination with an acid substantially free of its salt fond, an acid in its salt fonn, or a mixture of an acid and its salt form, results in the compositions having increased storage stability relative to liquid pharmaceutical compositions formulated in the absence of the combination of these two components.
  • These liquid pharmaceutical compositions comprising stabilized monomelic IL-2 may either be used in an aqueous liquid form, or stored for later use in a frozen state, or in a dried form for later reconstitution into a liquid fond or other form suitable for administration to a subject in accordance with the methods of present invention.
  • liquid pharmaceutical composition or fonnulation is dried either by freeze drying (i.e., lyophilization; see, for example, Williams and Polli (1984) J. Parenteral Sci. Technol. 38:48 59), spray drying (see Masters (1991) in Spray- Drying Handbook (Sth ed; Longman Scientific and Technical, Essez, U.K.), pp. 491-676; Broadhead et al. (1992) Drug Devel. Ind. Pharm. 18:1169-1206; and Mumenthaler et al. (1994) Pharm. Res. 11 :12- 20, or air drying (Carpenter and Crowe (1988) Cryobiology 25:459-470; and Roser (1991) Biopharm. 4:47-53).
  • freeze drying i.e., lyophilization
  • spray drying see Masters (1991) in Spray- Drying Handbook (Sth ed; Longman Scientific and Technical, Essez, U.K.), pp. 491-676; Broadhead et al. (19
  • IL-2 formulations that comprise IL-2 in its nonaggregated monomeric state include those described in Whittington and Faulds (1993) Drugs 46(3):446- 514.
  • These formulations include the recombinant IL-2 product in which the recombinant IL- 2 mutein Teceleukin (unglycosylated human IL-2 with a methionine residue added at the amino-terminal) is formulated with 0.25% human serum albumin in a lyophilized powder that is reconstituted in isotonic saline, and the recombinant IL-2 mutein Bioleukin (human IL-2 with a methionine residue added at the amino-terminal, and a substitution of the cysteine residue at position 125 of the human IL-2 sequence with alanine) fonnulated such that 0.1 to 1.0 mg/ml IL-2 mutein is combined with acid, wherein the fonnulation has a pH of 3.0 to 4.0, 10
  • multimeric IL-2 examples of pharmaceutical compositions comprising multimeric IL-2 are disclosed in commonly owned U.S. Patent No.4,604,377.
  • multimeric is intended the protein molecules are present in the pharmaceutical composition in a microaggregated form having an average molecular association of 10-50 molecules. These multimers are present as loosely bound, physically associated IL-2 molecules. A lyophilized form of these compositions is available commercially under the tradename Proleukin® IL-2 (Chiron Corporation, Emeryville, California).
  • the lyophilized formulations disclosed in this reference comprise selectively oxidized, microbially produced recombinant IL-2 in which the recombinant IL-2 is admixed with a water soluble carrier such as mannitol that provides bulk, and a sufficient amount of sodium dodecyl sulfate to ensure the solubility of the recombinant IL-2 in water.
  • a water soluble carrier such as mannitol that provides bulk
  • compositions are suitable for reconstitution in aqueous injections for parenteral administration and are stable and well tolerated in human patients.
  • the IL-2 When reconstituted, the IL-2 retains its multimeric state.
  • Such lyophilized or liquid compositions comprising multimeric IL-2 are encompassed by the methods of the present invention.
  • Such compositions are referred to herein as multimeric IL-2 pharmaceutical compositions.
  • the methods of the present invention may also use stabilized lyophilized or spray-dried pharmaceutical compositions comprising IL-2, which may be reconstituted into a liquid or other suitable form for administration in accordance with methods of the invention.
  • Such pharmaceutical compositions are disclosed in International Publication No. W O 01/49274 entitled "Methods for Pulmonary Del vefy of InterleuklTz-2.”
  • These compositions may further comprise at least one bulking agent, at least one agent in an amount sufficient to stabilize the protein during the drying process, or both.
  • IL-2 protein or variants thereof retains its monomelic or multimeric form as well as its other key properties of quality, purity, and potency following lyophilization or spray-drying to obtain the solid or dry powder form of the composition.
  • preferred carrier materials for use as a bulking agent include glycine, mannitol, alanine, valine, or any combination thereof, most preferably glycine.
  • the bulking agent is present in the formulation in the range of 0% to about 10% (w/v), depending upon the agent used.
  • Preferred carrier materials for use as a stabilizing agent include any sugar or sugar alcohol or any amino acid.
  • Preferred sugars include sucrose, trehalose, raffinose, stachyose, sorbitol, glucose, lactose, dextrose or any combination thereof, preferably sucrose.
  • the stabilizing agent is a sugar, it is present in the range of about 0% to about 9.0% (w/v), preferably about 0.5% to about 5.0%, more preferably about 1.0% to about 3.0%, most preferably about 1.0%.
  • the stabilizing agent is an amino acid, it is present in the range of about 0% to about 1.0% (w/v), preferably about 0.3% to about 0.7%, most preferably about 0.5%.
  • These stabilized lyophilized or spray- dried compositions may optionally comprise methionine, ethylenediaminetetracetic acid (EDTA) or one of its salts such as disodium EDTA or other chelating agent, which protect the IL-2 or variants thereof against methionine oxidation. Use of these agents in this manner is described in U.S. Application Serial No. 09/677,643, herein incorporated by reference.
  • the stabilized lyophilized or spray- dried compositions may be formulated using a buffering agent, which maintains the pH of the pharmaceutical composition within an acceptable range, preferably between about pH 4.0 to about pH 8.5, when in a liquid phase, such as during the formulation process or following reconstitution of the dried form of the composition. Buffers are chosen such that they are compatible with the drying process and do not affect the quality, purity, potency, and stability of the protein during processing and upon storage.
  • IL-2 pharmaceutical compositions represent suitable compositions for use in the methods of the invention.
  • any pharmaceutical composition comprising an IL-2 compound as a therapeutically active component is encompassed by the methods of the invention.
  • kits or package containing at least one combination composition of the invention, accompanied by instructions for use.
  • the kit comprises each of the drugs, along with instructions for use.
  • the drug components may be packaged in any manner suitable for administration, so long as the packaging, when considered along with the instructions for administration, clearly indicates the manner in which each of the drug components is to be administered.
  • each of the drug components of the combination may be combined into a single administrable dosage form such as a single composition.
  • kits comprising rIL-2 and an antiangiogenic agent
  • the kit may be organized by any appropriate time period, such as by day.
  • a representative kit may comprise unit dosages of each of rIL-2 and the antiangiogenic agent. If each of the drugs is to be administered twice daily, then the kit may contain, corresponding to Day 1, two rows of unit dosage forms of each of rIL-2 and the antiangiogenic agent, with instructions for the timing of administration.
  • the kit may contain, corresponding to Day 1, two rows of unit dosage forms of each of rIL-2 and the antiangiogenic agent, with instructions for the timing of administration.
  • one or more of the drugs differs in the timing or quantity of drug to be administered in comparison to the other drug members of the combination, then such would be reflected in the packaging and instructions.
  • exemplary Day 1 packaging might correspond to unit dosage forms of rIL-2 as "Day 1, Dose 1", along with dosage forms for the antiangiogenic agent corresponding to "Day 1, Dose 2".
  • the packaging may be in any form commonly employed for the packaging of pharmaceuticals, and may utilize any of a number of features such as different colors, wrapping, tamper-resistant packaging, blister paks, dessicants, and the like.
  • compositions for Co-administration with rIL-2 A Compositions for Co-administration with rIL-2 A. COMPOUNDS
  • Scheme 1 describes a modular method of synthesizing a myriad of substituted quinazoline compounds.
  • Reference to AG indicates an activating group such as, for example a halide, triflate, or ketone, wherein as many as four AG groups may be present. Since the 4-chloro position, shown in the third step, is more active than positions 5-8 on the benzyl ring, displacement with NHRR' may proceed without much concomitant displacement of AG in any of positions 5-8. Subsequent displacement of the AG group for R" in the final step may proceed in the presence of a weak to moderate base.
  • the AG group(s) may be modified to yield the desired product, for example a NO 2 group may be reduced with Fe and AcOH in EtOH and H 2 O to yield an amino substituent, which may be further substituted, for example by reductive amination with paraformaldehyde.
  • the AG group(s) in the starting material may be a substituent desired in the final product (as in R"), such as, for example an alkoxy group(s) or a substituted alkyl group(s).
  • a number functionalized 2-nitrobenzamide starting materials are available, which are easily converted to the 2-aminobenzamide starting material in the presence of a reducing agent, such as H 2 /Pd/C in EtOH.
  • Scheme 2a is performed as a one-pot procedure, with reagents in step a being NH 4 OH, CuCl in H 2 O.
  • reagents in step a being NH 4 OH, CuCl in H 2 O.
  • R 2 -R 5 are as defined herein.
  • reaction may be heated to enhance the yield, depending on reactivity of the particular starting materials.
  • R 9 as shown in scheme 2, is H, -OH, -CN, alkyl, aryl, heterocyclyl, alkoxy, or -NR 3 R b as defined herein. It is contemplated that the above structure may replace Formula II to allow substitution at R ⁇ >, whereby all other substituents are as defined herein.
  • the filter residue is dissolved in about 200 ml hot methanol, the solution is treated with 0.75 g activated carbon and filtered Via a Hyfio Super CeI, and the pH of the filtrate is adjusted to about 2.5 with 7 ml 3N methanolic HCl.
  • the filtrate is evaporated to about half the original volume and ether added until slight turbidity occurs; cooling then leads to the precipitation of crystals.
  • the crystallizate is filtered off, washed with a mixture of methanol/ether (1:2) as well as ether, dried for 8 h at 110° C. under HV, and equilibrated for 72 h at 20° C. and in room atmosphere.
  • Messenger RNA from human Jurkat cell line is used to create double stranded cDNA, which is hybridized into pBR322 plasmids.
  • a clone containing the IL-2 gene is identified using a 32 P-labeled oligonucleotide probe corresponding to a short length IL-2 base sequence.
  • the gene is inserted into a region of the pBR322 plasmid that has a convenient restriction site.
  • the appropriate promoter and ribosome-binding site is inserted in front of the IL-2 gene, and the resulting expression clone encodes a modified recombinant rIL-2.
  • Proleukin ® is as a lyophilized cake in 5 cc vials containing 1.3 mg of protein. Vials of Proleukin ® for injection are reconstituted with 1.2 niL of Sterile Water for Injection, USP. The diluent is directed against the side of the vial to avoid excess foaming, swirling contents gently until completely dissolved, while avoiding shaking. When reconstituted each mL contains 1.1 mg (18 million IU) of Proleukin ® . Reconstituted Proleukin® is suitable for intravenous injection directly or may be diluted as necessary in volumes of 50 mL to 500 mL with 5% Dextrose Injection, USP with 0.1% Albumin Human, USP. When diluting, the Albumin Human, USP is added to the 5% Dextrose Injection, USP prior to the addition of the reconstituted Proleukin ® .
  • rIL-2 recombinant human interIeukin-2; MIU: million International Units; CR: complete response; PR: partial response; NS: not stated; i.v.: intravenous.
  • Antiangiogenic agent is given again on day 1 and then every 2 weeks continuously in an 8-weeks treatment cycle.
  • Treatment with rIL-2 is continued for 6 consecutive weeks (days 1 - 42, Monday- Friday of each week) followed by a 2-week rest period resulting in an 8- week treatment cycle.
  • Example 4 Combination Therapy with rIL-2 and Small Molecule Receptor Tyrosine Kinase
  • Inhibitors BAY 43-9006 and SU11248 A Material and Methods Drugs
  • Recombinant human Interleukin-2 (Proleukin ® , Aldesleukin/rIL-2); 18 MIU/ ml, Chiron Corporation, Emeryville, CA) was reconstituted with sterile water for injection and formulated in 5% dextrose prior to administration.
  • Vincristine (vincristine sulfate) was from Mayne Pharma Ltd (Mulgrave, Australia).
  • CHIR-258 is 4-amino-5-fluoro-3-[5-(4-methylpiperazin- 1 -yl)- 1 H-benzimidazol-2-yl]quinolin- 2(lH)-one (Chiron).
  • BAY 43-9006 (Sorafanib/Nexavar®) (Riedl et al., Proc. Am. Assoc. Cancer Res. 2001 42(Abs 4956); Lowinger et al., Curr. Pharm. Des. 2002 8(25):2269-2278; WO 9932455) and SUl 1248 (Sunitinib/Sutent®) (Sun et al., J. Med. Chem. 2003 46(7):1116-1119; WO 0160814) were synthesized and purified in- house according to published procedures and patents.
  • BAY 43- 9006 or SUl 1248 (20 mM) were prepared in DMSO, and aliquots will be stored at - 20 0 C prior to use.
  • all drugs were diluted in optimal culture medium.
  • BAY 43-9006 was formulated in 100% PEG 400 vehicle, whereas, SUl 1248 dosing solutions were prepared in 5 mM citrate buffer. All other chemicals used were of research grade.
  • CTLL-2 IL-2 dependent T cell line
  • B16-F10 melanoma CT26 colon
  • renal carcinoma RENCA renal carcinoma RENCA
  • CTLL-2 were grown in RPMIl 640 supplemented with 10% FBS (fetal bovine serum, Gibco Life Technologies, Gaithersburg, MD), 2 mM L-glutamine, 1 mM sodium pyruvate, 25 mM HEPES, 0.5 nM rIL-2, 2 mM /3-mercaptoethanol.
  • RENCA cells were cultured in a media containing EMEM with 10% FBS, 2% IOOX vitamin, 1% 200 mM glutmine,l% 100 mM NaPy, 1% nonessential amino acids.
  • media contained EMEM, 10% FBS, 2% vitamins, 1% 200 mM glutamine, 1% 100 mM sodium pyruvate, 1% nonessential amino acids.
  • B16-F10 cells were grown in RPMI 1640 with 10% FBS, 1% nonessential amino acids, 1% 10OmM sodium pyruvate 2% vitamins; 2 mM l-glutamine; 2% sodium bicarbonate.
  • Yac-1 cells were cultured in RPMI + 10% FBS and subcultured 1-2 days prior to assay to ensure log- phase growth. Cells were maintained as suspension or adherent cultures in a humidified atmosphere at 37 0 C and 5% CO 2 . Cells were used in exponential growth phase (not exceeding 6-8 passages) with viability >98% (assessed using trypan blue staining) and determined free of mycoplasma.
  • mice Female BALB/c or C57BL6 mice (4-6 week-old, 18-22 g) were obtained from Charles River (Wilmington, MA) and acclimated for 1 week in pathogen-free enclosures prior to the start of the study. Animals received sterile rodent chow and water ad libitum and were housed in sterile filter-top cages with 12 hour light/dark cycles. All experiments were under the guidelines of the Association for Assessment and Accreditation of Laboratory Animal Care International.
  • B16-F10 (2 x 10 6 ), CT26 (2 x 10 6 ) or RENCA (1 x 10 6 ) cells were harvested, washed three times, and resuspended in PBS. Mice were shaved on the flank, and implanted (0.2 ml) subcutaneously (s.c.) into the right flank of mice.
  • B16-F10 tumor model C57BL6 mice were used, whereas CT26 and RENCA tumors were implanted in BALB/c mice. Treatments were initiated when tumors were established to a mean size of 50-250 mm 3 (day 0) as outlined in specific study designs. Mice were randomized into treatment cohorts (typically 10 mice/group).
  • rIL-2 was administered daily s.c. (0.2-3 mg/kg/day) on days 0-6 or 7-13 or days 0-4, 7-11.
  • BAY 43-9006 or SUl 1248 (1-100 mg/kg) were administered daily (for 5-12 days) as a solution via oral gavage, starting day 0 or day 7. All monotherapy and drug combinations at selected doses, outlined in individual studies were well tolerated.
  • Tumor volumes and body weights were assessed 2-3 times weekly. Caliper measurements of tumors were converted into mean tumor volume (mm 3 ) using the formula: 1 A (length (mm) x [width (mm)] 2 ). Tumor growth inhibition (TGI) was calculated as [l-(mean tumor volume of treated group/mean tumor volume of control group) x 100]. Responses were defined as either a complete response (CR, no measurable tumor), or partial response (PR 5 50-99% tumor volume reduction) compared to tumor volume for each animal at treatment initiation. Tumor growth delay analysis was calculated as: [(number of days for a treated group to reach a mean tumor volume of 1000 mm 3 ) - (number of days for the control group to reach a mean tumor volume of 1000 mm 3 )].
  • mice were treated with a single s.c. dose of rIL-2 (6 mg/kg, 0.2 ml) or BAY 43-9006 (20 mg/kg, p.o., 0.2 ml) and blood was collected at various times after drug administration. Plasma levels of BAY 43- 9006 and rIL-2 were determined using HPLC or an ELISA bioassay.
  • pERK detection was done with a mouse antibody to pERK (1 : 1000, Cell Signaling, Beverly, MA) and incubated at 4 0 C overnight.
  • Detection of pSTAT5 antibody (1 : 1000 Upstate) and pAKT (1 : 1000 Cell Signaling) was performed with the same amount of protein by probing with appropriate anti-phosphotyrosine antibodies for 2 hours at room temperature.
  • the membranes were then incubated for 1 hour at room temperature with 1 : 5000 horseradish peroxidase-conjugated anti-rabbit IgG (Jackson Immunoresearch, West Grove, PA).
  • blots were stripped and re-probed with anti- ERK (Cell Signaling), anti-STAT5 (BD Biosciences), and anti-AKT (Cell Signaling) antibodies to measure total ERK, STAT5 and AKT protein, respectively. Proteins were detected using enhanced chemiluminescence (ECL; Amersham Biosciences, Buckinghamshire, England) and visualized after exposure to Kodak film. Scanning densitometry was performed to quantify band intensities. The amount pERK, pSTAT5 or pAKT was normalized to total ERK, STAT5 or AKT protein levels, and compared with vehicle or untreated controls.
  • ECL enhanced chemiluminescence
  • Murine T cells were detected with a rabbit anti-CD3 antibody (1 :60 dilution, Dako Norden A/S, Glostrup Denmark), and monocytes/macrophages were detected using F4/80 (Serotec).
  • mouse tumors were stained for Ki-67 using a monoclonal rat anti-mouse antibody (1: 15 dilution, DAKO). Heat-induced epitope retrieval was performed using CC 1 (Ventana Medical Systems). Samples were then incubated with the appropriate secondary antibodies (goat anti-rabbit IgG biotinylated antibody, 1 : 100 dilution, Jackson ImmunoReseach Laboratories).
  • a horseradish peroxidase-labelled streptavidin biotin system with 3-3'-diaminobenzidine chromogen (Ventana Medical Systems) was used for localizing the antibodies. Sections were counterstained with nuclear fast red to enhance visualization of tissue morphology.
  • the Ventana Bluemap kit - an alternative NBT/BCIP staining method was used due to observed melanin deposits in H&E tissues, which aided in visualization of T cells in tumors.
  • CTLL-2 T cells were preincubated with or without BAY 43-9006 (3 ⁇ M) at 37 0 C for 2 hours. The cells were then washed and plated in 96-well microtiter plates, 5,000 cells/well in culture media with serial dilutions of rhIL-2 (1 pM to 100 nM). At the end of the incubation period (72 hours at 37 0 C), cell viability was determined by a tetrazolium dye assay using cell proliferation reagent WST-I (Roche Applied Science, Indianapolis, IN).
  • CTLL-2 5000 cells/well; RENCA: 1500/well; B16-F10: 1000/well; MV4;11 : 5000/well
  • CTLL-2 is an IL-2 dependent cell line, and hence for these cells, the cytoxicity assay was conducted in media containing 5 nM rIL-2.
  • % specific lysis 100 x ((mean experimental - mean spontaneous release)/(mean maximal release - mean spontaneous release)). The spontaneous release was determined from wells containing labeled target cells and no effector cells, and maximal release was determined from wells containing labeled target cells in 1% Triton X-100.
  • CTLL-2 Activates JAK/STAT and MAPK Signaling in CTLL-2 Cells in vitro
  • CTLL-2 cells IxIO 7 cells
  • rIL-2 Activates JAK/STAT and MAPK Signaling in CTLL-2 Cells in vitro
  • CTLL-2 cells IxIO 7 cells
  • key signaling pathways MAPK, STAT5, AKT were evaluated using western blot analyses.
  • IL-2R ⁇ KD 10 "11 M
  • IL-2 ⁇ KD 10 "9 M
  • cells were exposed to free anti-IL-2 ⁇ antibody (>1000-fold excess; 10 nM) incubated for 1 hour prior to addition of rIL-2.
  • serum starved CTLL-2 cells were activated with 10 nM rIL-2, and IL-2 signaling was examined at various times from 10 minutes up to 48 hours.
  • IL-2R downstream phosphorylation of ERK1/2, STAT5 and AKT was evaluated in rIL-2-treated cells using Western blot analysis.
  • the relative levels of pERK, pSTAT5 or pAKT were compared to the total protein levels for ERK, STAT5 or AKT, respectively.
  • pERKl/2 Activation of pERKl/2 was observed following exposure of CTLL-2 cells with rIL-2.
  • Phospho ERK was slightly activated at 1 pM at 2 hours however, maximal activation was seen at > 100 pM concentrations.
  • the JAK/STAT5 pathway was maximally activated at 1 pM concentration; and increasing concentrations of rlL- 2 did not change levels of pSTAT5 (up to 100 nM).
  • the basal pAKT pathway in CTLL-2 appeared to be activated in T cells under serum starved conditions. Furthermore pAKT levels remained largely unchanged at the rIL-2 concentrations tested (1 pM to 100 nM; using a pAKT antibody to phosphorylation site 483).
  • a blocking antibody to IL-2R ⁇ was used to confirm that the IL-2 signal pathways are mediated specifically by the binding to the IL-2R.
  • the pSTAT5 levels in CTLL-2 cells were analyzed by Western blot. CTLL cells were serum starved and treated with excess free anti-IL-2R ⁇ antibody (10 nM, > 1000-fold) for 1 hour prior to treatment with rIL-2 (0.1 pM to 10 nM). In the absence of the blocking IL-2R ⁇ antibody, pSTAT5 was activated at 1 pM following rIL-2 treatment, however, in the presence of IL-2R ⁇ inhibition, pSTAT5 signaling was abrogated (> 95% inhibition), confirming the requirement of the IL-2R ⁇ in STAT5 signaling.
  • rIL-2 Activation of pSTAT5 is Rapid and Sustained in CTLL-2 Cells To evaluate the time of onset and duration of IL-2R signaling responses in
  • CTLL-2 cells serum starved cells were treated in vitro with rIL-2 (10 nM) and effects on phosphorylation of ERKl /2, STAT5 and AKT was evaluated using Western blot analyses. Phosphorylation of STAT5 was activated in minutes ( ⁇ 10 minutes) after addition of rIL-2 to CTLL-2 cells and the duration of pSTAT5 response was maintained up to 48 hours. Activation of the MAPK pathway (pERK) pathway by rIL-2 appeared to be slightly delayed and was activated by 1 hour. The intensity of pERK was lower than the maximal response obtained by stimulation of serum starved CTLL-2 cells with PMA (50 ng/ml) + ionomycin (0.4 ⁇ g/ml) for 15 minutes.
  • PMA 50 ng/ml
  • ionomycin 0.4 ⁇ g/ml
  • pERK levels were sustained up to 24 hours and returned to background levels by 48 hours. No discernable effects were observed on the pAKT, confirming that signaling via PI-3K/AKT pathway was continually active in CTLL-2 cells.
  • BAY-43-9006 is a potent inhibitor of Raf-1, which also inhibits both wild-type and mutant BRAF.
  • BAY-43-9006 inhibits multiple kinases particularly VEGF2, 3; PDGFR ⁇ ; FLT3 and cKIT (Wilhelm, SM et al. Cancer Res 2004; and Chiron's kinase profiling data), and inhibits to some extent Lck and Fyn, two kinases that are involved in T cell functional responses.
  • Lck and Fyn two kinases that are involved in T cell functional responses.
  • BAY 43-9006 Since the concentration of BAY 43-9006 required to inhibit the MAPK pathway in different cell types is quite variable, the effects of various concentrations of BAY 43-9006 (ranging from 0 to 20 ⁇ M) were evaluated on serum starved CTLL- 2 cells for 2 hours with or without PMA + Ionomycin stimulation. At the end of the incubation period, treated CTLL-2 cells were then lysed and the protein lysates were subjected to Western blot analyses for determination of levels of pERK. BAY 43- 9006 substantially inhibited pERK at concentrations > 1 ⁇ M in murine CTLL-2 cells and the human Jurkat T cell line. The effects of BAY 43-9006 treatment on CTLL-2 cells did not alter pSTAT5 and pAKT levels in cells indicating that the JAK/STAT5 pathway and PI-3K/AKT in T cells were largely unaffected.
  • rIL-2 and BAY 43-9006 were studied.
  • the IL-2 mediated T cell signaling effects with concomitant or sequential regimens of the two drugs were investigated in CTLL-2 cells.
  • Serum starved CTLL-2 cells were treated with BAY 43-9006 (3 ⁇ M) for 2 hours and then treated with either vehicle, rIL-2 (10 nM) or PMA+Ionomycin.
  • rIL-2 (10 nM, 2 hours) treatment followed by BAY 43-9006 (3 ⁇ M, 2 hours) was also investigated.
  • the effects of BAY 43-9006 treatment on murine tumor cell lines- B16-F10 melanoma, CT26 colon and the RCC RENCA model was determined.
  • the murine cell lines were selected based on their responsiveness to in vivo rIL-2 therapy in immunocompetent models (T-/NK-/monocyte-/macrophage-competent mice), where the effects of combined rIL-2 and BAY 43-9006 therapy could be investigated.
  • Serum starved B16-F10 melanoma and RENCA cells were exposed to a range of concentrations of BAY 43-9006 from 0 to 20 ⁇ M.
  • Phospho-ERK levels in cell lysates following drug exposure were determined by Western blot analyses.
  • BAY 43-9006 inhibited pERK levels in both cell lines tested (B16-F10 and RENCA) at very high concentrations of > 5 ⁇ M, with almost complete abolition of pERK seen at 20 ⁇ M.
  • proliferation assays were conducted by pre- incubating CTLL-2 cells (5000 cells/well) at concentrations of BAY 43-9006 (3 ⁇ M, 2 hours) that inhibit pERK. After a 2 hour incubation of cells with BAY 43-9006 (3 ⁇ M), cells were plated in 96-well plates and exposed to various concentrations of rlL- 2 (0 - 100 nM) for 72 hours. Untreated cells were given sham treatments prior to incubation with rIL-2 (at the same concentrations).
  • MV4;11 human FLT3 ITD AML cell line
  • BAY 43-9006 (0 to 50 ⁇ M)
  • B16-F10 cells were treated with vincristine (0 to 1 ⁇ M) to confirm cytotoxicity of agents and validity of assays.
  • the inhibitory concentrations of the drugs were expressed as an EC 5O value, which was defined as the concentration needed for a 50% reduction in proliferative response measured as absorbance of drug- treated cells vs. untreated/vehicle controls.
  • CTLL-2 5000 cells/well; RENCA: 1500/well; B16-F10: 1000/well; CT26: 1000 cells/well; MV4;11 : 5000/well
  • WST-I tetrazolium dye
  • a EC50 concentration needed for a 50% reduction in proliferative response measured as absorbance of drug-treated cells vs. untreated/vehicle controls.
  • b MV4;11 is a human acute myelogenous leukemia (AML) cell line that expresses a FLT3 Internal Tandem Duplication (ITD).
  • d Cell viability was assessed using the Promega Cell-Titer GloTM assay that measured ATP content of cells
  • BAY 43-9006 or SUl 1248 were needed to inhibit proliferation of CTLL-2 cells as well as various cells lines (B 16- FlO, RENCA, CT26), compared to the antimitotic agent vincristine (defined by the relative EC50's; see Table 9).
  • the cytotoxicity of BAY 43-9006 effects on RENCA cells was also examined using the BrdU assay to evaluate the effect on DNA synthesis (vs. antiproliferative activity using the mitochondrial tetrazolium dye assay).
  • the EC 5 O for BAY 43-9006 ( ⁇ 5 ⁇ M) on RENCA cells obtained using the BrdU method was similar to that observed with the WST-I assay.
  • a single oral dose of 30 mg/kg BAY 43-9006 in mice achieved a C max of about 5500 to 8000 ng/ml (-10 ⁇ M) at a t max of 2 hours.
  • the plasma elimination rate of BAY 43-9006 was fairly slow and the consequent half-life was about 4 hours.
  • the PK profile of rIL-2 following subcutaneous administration of 6 mg/kg demonstrated a C max of about 550 - 850 ng/ml at a t max of about 30 minutes.
  • the rlL- 2 ti /2 in mice was approximately 1 hour, with exposures of >50 ng/ml achieved for 4 hours.
  • rIL-2 and BAY 43-9006 treatment decreases immune effector function ex vivo Since inhibition of one or multiple T lymphocytic kinases (Lck, Fyn, Syk, Btk, Src, Tck2, MAPK, JAKs) may abrogate T cell expansion and immune effector function, the effects of rIL-2 and BAY 43-9006 on T cell proliferative and functional responses in vivo was investigated.
  • T lymphocytic kinases Lck, Fyn, Syk, Btk, Src, Tck2, MAPK, JAKs
  • BALB/c mice bearing RENCA tumors were treated with rIL-2 (lmg/kg/day, s.c. days 6-10), BAY 43-9006 (30 mg/kg/day, p.o., days 6-10) or combinations of rIL-2 and BAY 43-9006 administered either concomitantly or sequentially (rIL-2, lmg/kg/day, s.c, days 1-5 + BAY 43-9006, 30 mg/kg/day, p.o., days 6-10; or BAY 43-9006, 30 mg/kg/day, p.o., days 1-5 + rIL-2, lmg/kg/day, s.c, days 6-10).
  • Isolated splenocytes from treated mice were then subjected to ex vivo killing assays against Yac-1 target cells at various effector: target (E:T) ratios, and percent specific lysis of 51 Cr-labeled Yac-1 cells were determined.
  • Mice treated with rIL-2 (1 mg/kg) significantly increased splenocyte-mediated killing of Yac-1 targets compared to vehicle treatment (23% with rIL-2 vs. 0% with vehicle treatment).
  • T cells 1575 cells/ ⁇ l with vehicle
  • T cells 1550 cells/ ⁇ l vs. 664 cells/ ⁇ l with vehicle
  • a significantly increased ratio of CD4:CD8 cells was observed with rIL-2 therapy compared to vehicle treatment (5:3; rIL-2: Vehicle, i.e., 1.7-fold), as indicative of rIL-2 mechanism of action in expanding T cell numbers.
  • the relative numbers of non-T and monocytic cells (CD45+CD3-) following rIL-2 treatment were similar to vehicle treatment (837 cells/ ⁇ l vs. 911 cells/ ⁇ l with vehicle).
  • single agent BAY 43-9006 or BAY 43-9006 combined with rlL-
  • lymphocytes and monocytes had little impact or increased the absolute numbers of lymphocytes and monocytes (range of 2417-3577 cells/ ⁇ l vs. 2387 cells/ ⁇ l with vehicle), and also increased non- T cells and monocyte populations (1241-1563 cells/ ⁇ l vs. 837 cells/ ⁇ l with vehicle).
  • the effect of BAY 43-9006 therapy on total numbers of T cells was similar to vehicle treatment (1827 cells/ ⁇ l vs. 1550 cells/ ⁇ l with vehicle). Slightly increased total T cell numbers (including both CD4+ and CD8+ populations) observed with the sequential regimen of rIL-2 and BAY 43-9006 when commenced with rIL-2 (2081 T cells/ ⁇ l vs. 1550 cells/ ⁇ l with vehicle).
  • BAY 43-9006 or SUl 1248 therapy was determined.
  • tumor infiltrating T cells were detected with a mouse anti-CD3 antibody.
  • Ki67 staining With rIL-2 treatment, increased numbers of T cells were seen infiltrating both RENCA tumors (and B16-F10 tumors) compared to vehicle treatment. Generally, fewer T cells were detected in the Bayer 43-9006-treated group in the RENCA model.
  • n 10 BALB/c mice/group in each study.
  • CT26 tumor-bearing mice were treated when tumors were established to a mean size of -225 mm 3 .
  • b T/Cobs erv e d (O) % T/C
  • Vehicle 712 1.00 N/A N/A N/A
  • n 10 BALB/c mice/group in each study.
  • RENCA model tumored mice were treated tumors were established to a mean size of -50-70 mm 3 .
  • Concomitant regimens are presented as RENCA tumor model exhibits severe cachexia (animal wasting) making sequential administration of agents not possible, and not evaluable.
  • b T/Cobserved (O) % T/C

Abstract

The present invention relates to combination therapies with IL-2 compositions and antiangiogenic agents for the treatment of cancer. Further provided are methods of alleviating toxicities and increasing the efficacy associated with the administration of IL-2 compositions or antiangiogenic compositions.

Description

ANTIANGIOGENIC AGENTS WITHALDESLEUKIN
CROSS-REFERENCE TO RELATED APPLICATION This application claims benefit under 35 U.S.C. § 119(e) of provisional application 60/654,341, filed February 18, 2005, which application is hereby incorporated by reference in its entirety.
FIELD OF THE INVENTION The present invention relates to methods of therapy for diseases associated with abnormal cellular proliferation. In some embodiments, recombinant IL-2 is combined with antangiogenic agents for use in the treatment of cancer.
BACKGROUND OF THE INVENTION Capillaries reach into almost all tissues of the human body and supply tissues with oxygen and nutrients as well as removing waste products. Under typical conditions, the endothelial cells lining the capillaries do not divide, and capillaries, therefore, do not normally increase in number or size in a human adult. Under certain conditions, however, such as when a tissue is damaged, or during certain parts of the menstrual cycle, the capillaries begin to proliferate rapidly. This process of forming new capillaries from pre-existing blood vessels is known as angiogenesis or neovascularization. See Folkman, J. Scientific American 275, 150-154 (1996). Angiogenesis during wound healing is an example of pathophysiological neovascularization during adult life. During wound healing, the additional capillaries provide a supply of oxygen and nutrients, promote granulation tissue, and aid in waste removal. After termination of the healing process, the capillaries normally regress. Lymboussaki, A. "Vascular Endothelial Growth Factors and their Receptors in Embryos, Adults, and in Tumors" Academic Dissertation, University of Helsinki, Molecular/Cancer Biology Laboratory and Department of Pathology, Haartman Institute, (1999).
Angiogenesis also plays an important role in the growth of cancer cells. It is known that once a nest of cancer cells reaches a certain size, roughly 1 to 2 mm in diameter, the cancer cells must develop a blood supply in order for the tumor to grow larger as αiltusion will not be sufficient to supply the cancer cells with enough oxygen and nutrients. Thus, inhibition of angiogenesis is expected to halt the growth of cancer cells. Of the identified angiogenic factors, vascular endothelial growth factor (VEGF) is the most potent and specific and has been identified as a crucial regulator of both normal and pathologic angiogenesis; although other factors such as epidermal growth factor (EGF), fibroblast growth factor (FGF), and platelet derived growth factor (PDGF) have been implicated in angiogenesis and cancer cell survival. Curr Opin Investig Drugs. 2001 Feb;2(2):280-6.
Specific, anti-tumor immunity involves activation of multiple cell types in the immune system, the most efficient being cytolytic T lymphocytes. To induce specific, anti-tumor T lymphocyte mediated immune response, recognition of not only the tumor antigen of interest, but also costimulatory interactions between specific ligands present on either the tumor cell or the antigen presenting cell, and the target T lymphocytes are required. This second, co-stimulatory signal may also be provided by soluble factors, such as cytokines or other peptide molecules which bind to specific, cell surface receptors and initiate various signal transduction pathways, resulting in augmentation of effector function.
Interleukin-2 (IL-2) is a T lymphocyte-derived cytokine which binds to specific receptors present on T cells and natural killer (NK) cells, and will activate them for tumor cytolysis, cytokine secretion, and other effector functions. Both CD4 and CD8 T lymphocytes express the receptor for IL-2, and develop increased cytolytic effector and cytokine synthetic function after exposure to the biologic. The functional effects of IL-2 are mediated via engagement with the IL-2 receptor (IL- 2R), which is structurally composed of three subunits: α, β, and a common γ chain that is shared with other cytokine receptors (Caligiuri et al., J. Exp. Med. 1990 171(5):1509-1526; Bazan, J.F., Science 1992 257(5068):410-413; Theze et al., Immunol. Today 1996 17(10):481-486). Activation of IL-2 receptors and consequent signaling is dependent upon IL-2 receptor types on cells (Caligiuri et al., J. Exp. Med. 1990 171(5):1509-1526; Voss et al., J. Exp. Med. 1992 176(2): 531-541; Expinoza et al., J. Leukoc. Biol. 1995 57(l):13-29; Theze et al., Immunol. Today 1996 17(10):481- 486). T cells express the high affinity IL-2Rαβγ, whereas NK cells, monocytes, and macrophages express predominantly the intermediate-affinity form, IL-2Rβγ (Caligiuri et al., J. Exp. Med. 1990 171(5): 1509-1526; Voss et al., J. Exp. Med. 1992 176(2):531-541; Theze et al., Immunol. Today 1996 17(10):481-486; Nagler et al, J. Exp. Med. 1990 171(5): 1527-1533). Central to its mechanism, IL-2 activates the JAKs (JAKl, JAK3), which phosphorylate key tyrosines on IL-2Rβ, which serve as docking sites for downstream signaling molecules, including She and Stat5, which then catalyze the activation of two distinct pathways: Shc/Ras/Raf/MAPK, and the JAK/STAT5, which translocates to the nucleus and directly regulates a family of gene-regulating transcription factors- STATs (O'Shea, J.J., Immunity 1997 7(1):1-11). She recruits the Grb-2/Sos complex and activates the Ras/Raf/MAPK pathway, and Grb-2/Gab-2, which activates the phosphatidylinositol 3-kinase (PI3K) pathway. Together these signaling proteins regulate gene transcription factors, ultimately controlling cell growth, division, differentiation and immune activity. In addition, IL- 2 also induces anti-apoptotic effects by regulating pro-mitogenic genes leading to increased bcl-2, bcl-xl, c-myb, which affects cell cycle control through activation of cdk 2,4,6 and inhibition of p27kipl or may be negatively regulated by SOCS (suppressors of cytokine signaling) (Smith, K.A., Science 1988 240(4856): 1169-1176; Theze et al., Immunol. Today 1996 17(10):481-486). It is reported that interaction between IL-2 and IL-2R triggers downstream activation of the MAPK, PI-3K, and JAK/STAT signaling pathways, leading to cell survival and proliferation (Miyazaki et al., Science 1994 266(5187):1045-1047; Beadling et al., Embo J. 1996 15(8):1902- 1913; Nakajima et al., Immunity 1997 7(5):691-701; Moriggl et al., Immunity 1999 l l(2):225-230).
Preclinical studies in various murine tumor models have demonstrated that recombinant human IL-2, when administered to tumor-bearing animals for periods of 10 to 14 days can result in regression of tumor burdens, long-term survival, and increased resistance to tumor regrowth. Analysis of splenic lymphocytes obtained from these animals has shown that the anti-tumor effects are due at least in part to augmentation of cytolytic T cell function. This effect includes activation of both direct cytolysis of tumor cells by the CTL, as well an increased synthesis of other, T lymphocyte derived cytokines, which may have further direct or indirect anti-tumor effects as well.
Metastatic melanoma and renal cell carcinoma (RCC) are largely incurable diseases and are resistant to most types of systemic chemotherapies, therefore there remains a considerable need for developing more effective therapies. Recent clinical data have supported the development of small molecule receptor tyrosine kinase (RTK) inhibitors particularly BAY 43-9006 or SUl 1248 in melanoma and renal cell carcinoma. Both BAY 43-9006 and SUl 1248 inhibit multiple kinases that are involved in tumor growth, proliferation and antiangiogenesis. BAY 43-9006 is a potent inhibitor of Raf- 1 , a member of the Raf/MEK/ERK signaling pathway (Richly et al., Int. J. Clin. Pharmacol. Ther. 2003 41(12):620-621; Wilhelm et al., Cancer Res. 2004 64(19):7099-7109; Awada et al., Br. J. Cancer 2005 92(10):1855-1861), and inhibits both WT B-Raf and mutant V599E B-Raf. In addition to the effects of BAY 43-9006 on Raf, it is believed that a majority of its activity may be mediated via inhibition of other RTKs, particularly VEGFRs (VEGFR-2 and VEGFR-3), PDGFRβ, and other key kinases FLT-3, and cKIT (Wilhelm et al., Cancer Res. 2004 64(19):7099-7109). Responses with BAY 43-9006 in early solid tumor trials have established that daily or twice daily administrations may result in disease stabilizations including some partial responses. To date, Phase I studies have indicated that BAY 43-9006 is generally well tolerated in patients. The most common toxicities with Bay 43-9006 involved gastrointestinal tract effects (diarrhea, nausea, abdominal cramping) or derniatologic reactions including skin pruritus, or rash (Richly et al., Int. J. CHn. Pharmacol. Ther. 2003 41(12):620-621; Ahmad and Eisen, Clin. Cancer Res. 2004 10(18 Pt 2):6388S-92S; Awada et al., Br. J. Cancer 2005 92(10):1855-1861; Strumberg et al., J. Clin. Oncol. 2005 23(5):965-972).
In contrast, SUl 1248 is a highly potent, selective RTK inhibitor of VEGFR-I- 3, PDGFRα, cKIT, FLT3 and PDGFRβ (Abrams et al., MoI Cancer Ther. 2003 2(5):471-478; Mendel et al., Clin. Cancer Res. 2003 9(l):327-337; O'Farrell et al., Clin. Cancer Res. 2003 9(15):5465-5476). SU11248 has shown antitumor activity in a number of advanced solid tumors (RCC, neuroendocrine, stromal and adenocarninomas) (Faivre et al., J. Clin. Oncol. 2006 24(l):23-35; Motzer et al., J. Clin. Oncol. 2006 24(1): 16-24). Clinical data of SUl 1248 also indicate that the drug is generally tolerated with manageable toxicities, which include fatigue, lymphopenia, neutropenia, hyperlipasemia (Faivre et al., J. Clin. Oncol. 2006 24(l):23-35; Motzer et al., J. Clin. Oncol. 2006 24(1): 16-24).
Studies of the hereditary form of clear-cell renal carcinoma, which occurs in the von Hippel-Lindau syndrome, led to the identification of the von Hippel-Lindau tumor suppressor gene (VHL). The gene is mutated both in hereditary renal-cell carcinoma (where one mutation is a germ-line mutation) and in most cases of sporadic clear-cell renal carcinoma (where both alleles have acquired mutations or deletions). Gnarra JR, Duan DR, Weng Y, et al. Molecular cloning of the von Hippel-Lindau tumor suppressor gene and its role in renal carcinoma. Biochimica et Biophysica Acta. 1996;1242(3):201-10. One consequence of these mutations is the overproduction of vascular endothelial growth factor through mechanism involving hypoxia-inducible factor. O Iliopoulous AL, C Jiang, et al. Negative Regulation of Hypoxia-inducable genes by the Von-Hippel Lindu protein. Proc Natl Acad Sci USA. 1996;93: 10595- 10599. Thus, by its regulation of vascular endothelial growth factor, the von Hippel- Lindau protein is tightly linked to angiogenesis. Vascular endothelial growth factor stimulates the growth of endothelial cells and, as mentioned, appears to be a central factor in angiogenesis, particularly during embryogenesis, ovulation, wound healing, and tumor growth. Ferrara N. Molecular and biological properties of vascular endothelial growth factor. J. MoI. Med. 1999;77:527-543. Vascular endothelial growth factor represents a good target for treatment of clear-cell kidney cancer because mutations in the von Hippel-Lindau tumor- suppressor gene, result in overproduction of this growth factor by the tumors. A randomized, double-blind, placebo-controlled study of the humanized monoclonal antibody (bevacizumab) in patients with metastatic clear-cell renal cancer, was conducted by Yang et al. A randomized trial of bevacizumab, an anti-vascular endothelial growth factor antibody, for metastatic renal cancer. New England Journal of Medicine. 2003;349(5):427-34. The time to tumor progression was prolonged by a factor of 2.55 in patients given bevacizumab per kilogram (10 mg/kg every two weeks), as compared with patients in the placebo group. Survival was not a primary end point in this trial, which allowed patients to cross over from placebo to bevacizumab therapy at the time of disease progression. Id. Treatments for renal cancer that target angiogenic mechanisms may also be effective through pathways other than that mediated by vascular endothelial growth factor. There are other proteins in the local microenvironment of some tumors that can promote angiogenesis. As such, a need exists for antiangiogenic therapy utilizing a rational combination of inhibitors, directed by the understanding of the biology of renal cell carcinoma.
Recombinant human interleukin-2 (aldesleukin) was approved by the FDA for the treatment of metastatic renal cancer based on the results of several multicenter trials in 255 patients who received an intermittent high dose bolus regimen. In these trials, objective responses were seen in 15% of patients, and median survival was 16.3 months (Fisher and Rosenberg 2000 Cancer J Sci Am 6Sl :S55-57). Owing to the significant toxicity associated with this regimen, a series of phase I and phase II trials employing rIL-2 at different doses and using different routes of administration were undertaken. A recent review of the efficacy of rIL-2 as a single agent indicated an overall response of 15% in more than 1700 patients with metastatic RCC who were treated in this series of studies. Complete responses were noted in 3 to 5% of patients. Bukowski RM. Natural history and therapy of metastatic renal cell carcinoma: the role of interleukin-2. Cancer. 1997;80(7):l 198-220. The rates do not appear to differ between these routes. A randomized comparison between high dose bolus intravenous regimen and low dose bolus IV regimen and subcutaneous outpatient regimen showed no difference in median survival between the groups, but low dose and outpatient regimens were significantly less toxic. Yang JC, Sherry RM, Steinberg SM, et al. Randomized study of high-dose and low-dose interleukin-2 in patients with metastatic renal cancer. Journal of Clinical Oncology. 2003;21(16):3127-32.
Besides having direct immunomodulating effects, rIL-2 may also prevent tumor proliferation by causing secondary cytokine release (IFNγ) from activated NK cells. Saraya KLA, Balkwill FR. Temporal sequence and cellular origin of interleukin-2 stimulated cytokine gene expression. British Journal of Cancer. 1993;67(3):514-21. The response rates and outcomes of clear cell renal cancer patients can be improved. Clinical trials employing novel drug combinations are needed. The combination of rIL2 and antiangiogenic agents, such as bevacizumab may have additive effects that could translate to added clinical benefit for patients with metastatic renal cell carcinoma, as well as other cancers. Another advantage to the complimentary approach to cancer regression is that it provides a platform less easily bypassed by resistance mutations. Where therapeutic targets are so polarized and specific (which may be necessary in order to avoid targeting host cells), such as a particular substrate in a viral replicon or a kinase in a cancer cell line, a single point mutation in the disease state may render it unaffected by a drug resulting in even harsher strains of the disease in future generations.
Novel methods and mechanisms for treating patients having disorders associated with abnormal proliferation that are resistant to, or inadequately treated by conventional approaches, utilizing agents targeting immune response mechanisms in the body and disease-state substrates, are needed. The present invention provides such therapeutic agents, and further provides other related advantages.
BRIEF SUMMARY OF THE INVENTION
The present invention is based in part on unique combination therapies using small molecule receptor tyrosine kinase inhibitors and rIL-2 with non-overlapping toxicities. Given that melanomas and RCC are generally responsive to immunotherapy such as rIL-2, rational combinations of rIL-2 with potential receptor tyrosine kinase inhibitors, based on pharmacology of the individual agents, have been evaluated herein. The invention provides clinically applicable drug schedules to circumvent potential drug interactions based on the toxicological profile of these agents. The potential of adverse pharmacokintic/pharmacodynamic interations and adverse pharmacological interactions are also elucidated. The effects of rIL-2 and small molecule receptor tyrosine kinase inhibitors, such as BAY 43-9006, on T cells and the impact on IL-2 -mediated signaling pathways (MAPK and JAK/STAT5) is described.
Thus, the present invention expands the indication of the anti-tumor efficacy of IL-2 compounds, such as recombinant IL-2 (rIL-2, also known as aldesleukin) against cancer cell lines that respond poorly to conventional therapies, resulting in increased long-term survival and immunity to tumor rechallenge. Furthermore, the immunostimulotory effects of rIL-2 aim to alleviate existing side effects caused by administration of antiangiogenic compositions for co-administration therewith. Methods of treating a subject suffering from abnormal cellular proliferation, particularly renal cell carcinoma using a combination of an IL-2 compound, such as rIL-2 and at least one antiangiogenic agent is provided. Further, methods of treating a subject suffering from abnormal cellular proliferation, particularly renal cell carcinoma using a combination of an IL-2 compound, such as rIL-2 and a small molecule are provided as well. Preferred small molecules of the present invention are listed in Tables 1-5. Particularly preferred molecules are N-(2-
(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl-lH- pyrrole-3-carboxamide (also known as SUl 1248) and l-(4-(2- (methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluoromethyl)phenyl)urea (also known as Bay 43-9006). Aldesleukin and antiangiogenic agents may be administered together or separately as individual pharmaceutical compositions. If administered separately, Aldesleukin can be administered prior to, concurrent with, or subsequent to the antiangiogenic agent. Provided are particular regimens comprising daily, weekly, and monthly dosing schedules (or iterations thereof) for the co-administration of aldesleukin with antiangiogenic agent, such as 6,7-bis(2-methoxyethoxy)-N-(3- ethynylphenyl)quinazolm-4-amine, 6-(3-moφholinopropoxy)-N-(3-chloro-4- fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2-(dimethylamino)ethyl)-5-((5- fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl-lH-pyrrole-3-carboxamide, N-(4- chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin-l -amine, and l-(4-(2- (methylcarbamoyl)ρyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluoromethyl)phenyl)urea.
In one embodiment the antiangiogenic agents, preferably one of 6,7-bis(2- methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6-(3-morpholinopropoxy)- N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2-
(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl-lH- pyrrole-3-carboxamide, N-(4-chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin- 1 - amine, and l-(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluoromethyl)phenyl)urea are co-administered with antiangiogenic proteins, such as monoclonal antibodies capable of inhibiting VEGF. In a more particular embodiment the antiangiogenic proteins of the present invention are bevacizumab or VEGF-Trap. In another more particular embodiment the protein is an EGF inhibitor, such as cetuximab.
In another embodiment, administration of aldesleukin and antiangiogenic agent(s) together in the manner set forth herein potentiates the effectiveness of the antiangiogenic agent, resulting in a positive/synergistic therapeutic response that is improved with respect to that observed with the inhibitor alone.
Other embodiments provide a therapeutic package suitable for commercial sale for treating a patient suffering from cancer, comprising a container, a therapeutically effective amount of aldesleukin, and a therapeutically effective amount of an antiangiogenic agent and/or small molecule, preferably listed in Tables 1-5, such as 6,7-bis(2-methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6-(3- morpholinopropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N- (2-fdimethvlamino')ethvlV5-ff5-fli]nro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl- lH-pyrrole-3-carboxamide, N-(4-chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin-l- amine, or 1 -(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluoromethyl)ρhenyl)urea.
In yet further embodiments, a kit is provided. The kit comprises a combination of medicaments for the treatment of a patient suffering from cancer, comprising: (a) aldesleukin, and (b) an antiangiogenic agent selected from 6,7-bis(2- methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6-(3-morpholinopropoxy)- N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2- (dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl-lH- pyrrole-3-carboxamide, N-(4-chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin-l- amine, or l-(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluorornethyl)phenyl)urea, for simultaneous, sequential or separate use.
Methods of manufacturing the combinations described herein are provided and contemplated to fall within the scope of the invention as is the use of the combinations in methods for manufacturing medicaments for use in the methods of the invention.
Further embodiments of the invention include those described in the detailed description.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1 shows single agent activity of rIL-2, BAY 43-9006 or SUl 1248 in IL-2-responsive, T-cell competent murine tumor models. B16-F10 melanoma (2 x 106), CT26 colon (2 x 106) or RENCA renal carcinoma (Ix 106) cells were implanted s.c. into the right flank of female C57BL6 or BALB/c mice. Treatments were initiated when tumors were established to a mean size of 50 - 225 mm3, as outlined in methods. Mice were randomized into treatment cohorts (10 mice/group). rIL-2 was administered daily subcutaneously (0.2-3 mg/kg/d). BAY 43-9006 or SUl 1248 (1 - 100 mg/kg) were administered daily as a solution via oral gavage. Figure 1 illustrates the mean tumor growth inhibition (calculated as [l-(mean tumor volume of treated group/mean tumor volume of vehicle group) x 100]) of single agents in the B 16-F 10, CT26 and RENCA tumor model between days 10-14. The data is compiled from multiple independent studies with each study of 10 mice/group. * denotes statistical significance vs. Vehicle treatment (p< 0.05, ANOVA). Figure 2 shows efficacy of concomitant rIL-2 and BAY 43-9006 therapy in the B16-F10 murine melanoma model. B16-F10 cells (2 x 106 cells) were implanted subcutaneously in the right flank of female C57BL6 mice. Mice were randomized into groups when tumors were ~50 mm3, and treatments initiated on day 0 with either vehicle, days 0-6 (♦), rIL-2 (3.3 mg/kg, s.c, days 0-6) (Δ) or BAY 43-9006 (30 mg/kg, p.o. days 0-6) (x) or combined rIL-2 (3.3 mg/kg, s.c, days 0-6) + BAY 43- 9006 (30 mg/kg, p.o. days 0-6) (■). Figure 2 illustrates the mean tumor volume (mm3 + SE) vs. days post randomization (n= 10 mice/group).
Figures 3 A and 3B show efficacy of sequential rIL-2 and BAY 43-9006 therapy in the B 16-F 10 murine melanoma model. B 16-F 10 cells (2 x 106 cells) were implanted subcutaneously in the right flank of female C57BL6 mice. Mice were randomized into groups when tumors were -50 mm3, and treatments initiated on day 0. Figure 3A shows results from sequential regimen with rIL-2 administered first, then BAY 43-9006. Panel illustrates: vehicle (♦) days 0-6, 7-13; rIL-2 (3.3 mg/kg, s.c, days 0-6) (Δ) or BAY 43-9006 (30 mg/kg, p.o. days 7-13) (x) or combined rIL-2 (3.3 mg/kg, s.c, days 0-6) + BAY 43-9006 (30 mg/kg, p.o. days 7-13) (•). Figure 3B shows results from sequential regimen with BAY 43-9006 administered first, followed by rIL-2. Panel illustrates: vehicle (♦) days 0-6, 7-13; BAY 43-9006 (30 mg/kg, p.o. days 0-6) (x); rIL-2 (3.3 mg/kg, s.c, days 7-13) (D) or combined BAY 43-9006 (30 mg/kg, p.o. days 0-6) + rIL-2 (3.3 mg/kg, s.c, days 7-13) (A). Graphs show the mean tumor volume (mm3 + SE) vs. days post randomization (n= 10 mice/group).
Figure 4 shows efficacy of concomitant rIL-2 and SUl 1248 therapy in the B16-F10 murine melanoma model. B16-F10 cells (2 x 106 cells) were implanted subcutaneously in the right flank of female C57BL6 mice. Mice were randomized into groups when tumors were ~50 mm3, and treatments initiated on day 0 with either vehicle, days 0-6 (0), rIL-2 (3.3 mg/kg, s.c, days 0-6) (D) or SUl 1248 (40 mg/kg, p.o. days 0-6) (O) or combined rIL-2 (3.3 mg/kg, s.c, days 0-6) + SUl 1248 (40 mg/kg, p.o. days 0-6) (A). Figure 4 illustrates the mean tumor volume (mm3 ± SE) vs. days post randomization (n= 10 mice/group).
Figures 5 A and 5B show efficacy of sequential rIL-2 and SUl 1248 therapy in the Bl 6-F 10 murine melanoma model. Bl 6-F 10 cells (2 x 106 cells) were implanted subcutaneously in the right flank of female C57BL6 mice. Mice were randomized into groups when tumors were ~50 mmJ, and treatments initiated on day 0. Figure 5A shows results from sequential regimen with rIL-2 administered first, then SUl 1248. Panel illustrates: vehicle (0) days 0-6, 7-13; rIL-2 (3.3 mg/kg, s.c, days 0-6) (□) or SUl 1248 (40 mg/kg, p.o. days 7-13) (x) or combined rIL-2 (3.3 mg/kg, s.c, days 0-6) + SUl 1248 (40 mg/kg, p.o. days 7-13) (•). Figure 5B shows results from sequential regimen with SUl 1248 administered first, followed by rIL-2. Panel illustrates: vehicle (0) days 0-6, 7-13; SUl 1248 (40 mg/kg, p.o. days 0-6) (O); rIL-2 (3.3 mg/kg, s.c, days 7-13) (Δ) or combined SUl 1248 (40 mg/kg, p.o. days 0-6) + rIL-2 (3.3 mg/kg, s.c, days 7-13) (■). Graphs show mean tumor volume (mm3 + SE) vs. days post randomization (n= 10 mice/group).
Figure 6 shows efficacy of sequential rIL-2 and BAY 43-9006 therapy in the CT26 murine colon adenocarcinoma model. CT26 cells (2 x 106 cells) were implanted subcutaneously in the right flank of female BALB/c mice. Mice were randomized into groups when tumors were -225 mm3, and treatments initiated on day 0. Panel illustrates the sequential regimen with rIL-2 administered first, then BAY 43- 9006: vehicle (♦) days 0-6, 7-13; rIL-2 (1 mg/kg, s.c, days 0-6) (D) or BAY 43- 9006 (40 mg/kg, p.o. days 7-13) (Δ) or combined rIL-2 (1 mg/kg, s.c, days 0-6) + BAY 43-9006 (40 mg/kg, p.o. days 7-13) (•).
Figure 7 shows efficacy of concomitant treatment of rIL-2 and SUl 1248 in the CT26 murine colon adenocarcinoma model. CT26 cells (2 x 106 cells) were implanted subcutaneously in the right flank of female BALB/c mice. Mice were randomized into groups when tumors were -225 mm3, and treatments initiated on day 0. Panel illustrates: vehicle (♦) days 0-6; rIL-2 (1 mg/kg, s.c, days 0-6) (D) or SUl 1248 (40 mg/kg, p.o. days 0-6) (O) or combined rIL-2 (1 mg/kg, s.c, days 0-6) + SUl 1248 (40 mg/kg, p.o. days 0-6) (•).
Figures 8 A and 8B shows efficacy of sequential rIL-2 and SUl 1248 therapy in the CT26 murine colon adenocarcinoma model. CT26 cells (2 x 106 cells) were implanted subcutaneously in the right flank of female BALB/c mice. Mice were randomized into groups when tumors were -225 mm3, and treatments initiated on day 0. Panel illustrates: Figure 8A shows results from the sequential regimen with rIL-2 administered first, then SUl 1248: vehicle (♦) days 0-6, 7-13; rIL-2 (1 mg/kg, s.c, days 0-6) (D) or SUl 1248 (40 mg/kg, p.o. days 7-13) (O) or combined rIL-2 (1 mg/kg, s.c, days 0-6) + SUl 1248 (40 mg/kg, p.o. days 7-13) (■ ). Figure 8B shows results irom sequential regimen with SUl 1248 administered first, followed by rIL-2. Panel illustrates: vehicle (♦) days 0-6, 7-13; SUl 1248 (40 mg/kg, p.o. days 0-6) (O); rIL-2 (3.3 mg/kg, s.c, days 7-13) (D) or combined SUl 1248(40 mg/kg, p.o. days 0-6) + rIL-2 (3.3 mg/kg, s.c, days 7-13) (A). Graphs show the mean tumor volume (mm3 + SE) vs. days post randomization (n= 10 mice/group).
Figure 9 shows efficacy of concomitant rIL-2 and BAY 43-9006 therapy in the murine RCC RENCA tumor model. RENCA cells (1 x 106 cells) were implanted subcutaneously in the right flank of female BALB/c mice. Mice were randomized into groups when tumors were ~50 mm3, and treatments initiated on day 0 with either vehicle, days 0-8 (♦), rIL-2 (1 mg/kg, s.c, days 0-4, 7-11) (Δ) or BAY 43-9006 (30 mg/kg, p.o. days 0-8) (x) or combined rIL-2 (1 mg/kg, s.c, days 0-4, 7-11) + BAY 43-9006 (30 mg/kg, p.o. days 0-8 (■). Figure 9 illustrates the mean tumor volume (mm3 + SE) vs. days post randomization (n= 10 mice/group).
Figure 10 shows efficacy of concomitant treatment of rIL-2 and SUl 1248 in the therapy in the murine RCC RENCA tumor model. RENCA cells (1 x 106 cells) were implanted subcutaneously in the right flank of female BALB/c mice. Mice were randomized into groups when tumors were ~50 mm3, and treatments initiated on day 0. Panel illustrates: vehicle (♦) days 0-6; rIL-2 (1 mg/kg, s.c, days 0-6) (■) or SUl 1248 (40 mg/kg, p.o. days 0-6) (x) or combined rIL-2 (1 mg/kg, s.c, days 0-6) + SUl 1248 (40 mg/kg, p.o. days 0-6) (A).
DETAILED DESCRIPTION OF THE INVENTION
Utilizing a detection assay, several types of cancers have been characterized as susceptible to treatment with antiangiogenisis/VEGF inhibitors and cytokine administration or modulation with rIL-2. Such cancers include, for example, CML, AML, breast, gastric, endometrial, salivary gland, adrenal, non-small cell lung, pancreatic, renal, rectal, skin, melanoma, multiple myeloma, brain/CNS, cervix, nasopharynx, malignant mesothelioma, hypopharynx, gastroinstestinal carcinoid, peritoneum, omentum, mesentery, gallbladder, testis, esophageal, lung, thyroid, ovarian, peritoneal, prostate, head and neck, bladder, colon, colorectal, lymphomas, and glioblastomas. The methods described herein are useful in the treatment of any such cancer. Therapy with a combination of aldesleukin and at least one antiangiogenic agent in the manner set forth herein causes a physiological response that is beneficial with respect to treatment of cancers whose unabated proliferating cells are highly dependent on vascularization, such as VEGF. One embodiment of the invention provides a method of treating a cancer patient suffering from hypotension from the administration of aldesleukin, comprising: co-administering to the patient a therapeutically effective amount of an antiangiogenic agent to attenuate hypotension. Another more particular embodiment thereof comprises amelioration of cancer in the patient.
Another embodiment of the invention provides a method of treating a cancer patient suffering from hypertension from the administration of an antiangiogenic agent, comprising: co-administering to the patient a therapeutically effective amount of aldesleukin to attenuate hypertension.
Another more particular embodiment thereof comprises amelioration of cancer in the patient. In certain embodiments, the cancer is susceptible to inhibition of angiogenesis and/or immune stimulation.
In a more particular embodiment of any of the previous embodiments, said antiangiogenic agent is selected from 6,7-bis(2-methoxyethoxy)-N-(3- ethynylphenyl)quinazolin-4-amine, 6-(3 -morpholinopropoxy)-N-(3 -chloro-4- fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2-(dimethylamino)ethyl)-5-((5- fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl-lH-pyrrole-3-carboxamide, N-(4- chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin- 1 -amine, or 1 -(4-(2- (methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluoromethyl)phenyl)urea.
Another embodiment of the invention provides a method of treating a patient suffering from cancer comprising administering to said patient aldesleukin and a compound selected from 6,7-bis(2-methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4- amine, 6-(3-morpholinopropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin- 4-amine, N-(2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)- 2,4-diethyl- 1 H-pyrrole-3 -carboxamide, N-(4-chlorophenyl)-4-((pyridin-4- yl)methyl)phthalazin- 1 -amine, or 1 -(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)- 3-(4-chloro-3-(trifluoromethyl)phenyl)urea. Another embodiment of the invention provides a method of increasing efficacy of aldesleukin in a cancer patient comprising first administering an antiangiogenic agent in a dose capable of inducing hypoxia in the patient then administering the aldesleukin. Another embodiment provides a method of increasing efficacy of an antiangiogenic agent in a cancer patient comprising reducing nitric oxide synthase by administration of aldesleukin to the patient. Another embodiment provides a method of increasing efficacy of an antiangiogenic agent in a cancer patient comprising administration aldesleukin to the patient wherein, nitric oxide synthase is thereby reduced by administration of aldesleukin to the patient.
In a more particular embodiment thereof, said antangiogenic agent is selected from 6,7-bis(2-methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6-(3- morpholinopropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N- (2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl- 1 H-pyrrole-3 -carboxamide, N-(4-chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin- 1 - amine, or 1 -(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3 -(4-chloro-3 - (trifluoromethyl)phenyl)urea.
Another embodiment of the invention provides a method for treating a patient suffering from cancer, comprising the steps of first administering to said patient a therapeutically effective amount of aldesleukin followed by administration of an antiangiogenic agent (such as) selected from 6,7-bis(2-methoxyethoxy)-N-(3- ethynylphenyl)quinazolin-4-amine, 6-(3-morpholinopropoxy)-N-(3-chloro-4- fluorophenyl)-7-methoxyquinazolm-4-amine, N-(2-(dimethylamino)ethyl)-5-((5- fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl-lH-pyrrole-3-carboxamide, N-(4- chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin-l -amine, or l-(4-(2- (methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluoromethyl)phenyl)urea.
Another embodiment of the invention provides a method for treating a patient suffering from cancer, comprising the steps of first administering to said patient a therapeutically effective amount of an antiangiogenic agent, such as, 6,7-bis(2- methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6-(3-morpholinopropoxy)- N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2- (dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl-lH- pyrrole-3 -carboxamide, N-(4-chloroρhenyl)-4-((pyridin-4-yl)methyl)phthalazin- 1 - amine, or l-(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluoromethyl)phenyl)urea followed by administration of aldesleukin.
Another embodiment of the invention provides a method for treating a patient suffering from cancer, comprising separately administering to said patient a therapeutically effective amount of an antiangiogenic agent and aldesleukin according to a dosing schedule, wherein the aldesleukin is administered from 1 to 3 times daily in a dose between about 9 and about 130 MIU/day for a period of at least 3 consecutive days, optionally followed by a rest period of at least 3 consecutive days.
In a more particular embodiment thereof, said antiangiogenic agent is administered from 1 to 6 times every 2-3 weeks.
In a more particular embodiment thereof, said antiangiogenic agent is a VEGF inhibitor.
In a more particular embodiment thereof, the antiangiogenic agent is selected from 6,7-bis(2-methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6-(3- morpholinopropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N- (2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl- 1 H-pyrrole-3 -carboxamide, N-(4-chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin- 1 - amine, or 1 -(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluoromethyl)phenyl)urea In a more particular embodiment thereof, the aldesleukin is administered intravenously and said rest period is present.
In a more particular embodiment thereof, the aldesleukin is administered subcutaneously and said rest period is absent.
In a more particular embodiment thereof, the aldesleukin is administered 3 times daily in a dose of about 30-100 MIU/day.
In a more particular embodiment thereof, the aldesleukin is administered for a period of 5 consecutive days followed by a 9-day rest period.
In a more particular embodiment said dosing schedule is repeated for at least two courses. In a more particular embodiment still, each course consists of 2-5 day treatments followed by a rest period of 9-15 days.
In another more particular embodiment thereof, the aldesleukin is administered 3 times for the first day and once daily each proceeding day. In a more particular embodiment thereof, said dosing schedule is repeated for at least two courses, or 3 courses, or 4 courses, or 5 courses, or 6 courses, or 7 courses, or 8 courses, or 9 courses, or 10 courses.
In a more particular embodiment of any of the previous embodiments, the cancer is colon cancer, renal cell carcinoma or malignant melanoma.
In a more particular embodiment of any of the previous embodiments, upon administration of aldesleukin at least one compound selected from acetaminophen, meperidine, indomethacin, ranitidine, nizatidine, diastop, loperamide, diphenhydramine, or furosemide is also administered to said patient. In a preferred embodiment of any of the preceding embodiments, said antangiogenic agent is 6,7-bis(2-methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4- amine, 6-(3 -morpholinopropoxy)-N-(3 -chloro-4-fluorophenyl)-7-methoxyquinazolin- 4-amine, N-(2-(dimethylammo)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)- 2,4-diethyl- 1 H-pyrrole-3 -carboxamide, N-(4-chlorophenyl)-4-((pyridin-4- yl)methyl)phthalazin-l -amine, or l-(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)- 3 -(4-chloro-3 -(trifluoromethyl)phenyl)urea.
Another embodiment of the invention provides a method of treating a patient suffering from cancer comprising administering aldesleukin and:
Figure imgf000018_0001
, a tautomer thereof, a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable salt of the tautomer.
Another embodiment of the invention provides a method of treating a patient suffering from cancer comprising administering aldesleukin and:
6 005720
Figure imgf000019_0001
, a tautomer thereof, a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable salt of the tautomer.
Another embodiment of the invention provides a method of treating a patient suffering from cancer comprising administering aldesleukin and:
Figure imgf000019_0002
, a tautomer thereof, a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable salt of the tautomer.
Another embodiment of the invention provides a method of treating a patient suffering from cancer comprising administering aldesleukin and:
Figure imgf000019_0003
, a tautomer thereof, a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable salt of the tautomer.
Another embodiment of the invention provides a method of treating a patient
onfffirinp from cancer comprising administering aldesleukin and:
Figure imgf000019_0004
a tautomer thereof, a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable salt of the tautomer.
Another embodiment of the invention provides a method of treating a patient suffering from cancer comprising administering aldesleukin and a compound of formula I:
Figure imgf000020_0001
I wherein,
Ri is alkyl, -aryl(Rj)p, or heterocyclyl; R2 is H or alkyl; or,
Ri and R2 are bound together to form R]-2;
R3 is H, -CN, -OH, halogen, alkyl, aryl, alkoxy, -NRaRb, -C(O)Rc, -S(O)nRa, or heterocyclyl;
R4 Is H, -CN, -OH, halogen, alkyl, aryl, -0-(CH2)q-Rg, -O-(CH2)q-O-Re, -NR3Rb, -S(O)nRd, or -heterocyclyl-Rf;
R5 is H, -CN, -OH, halogen, alkyl, aryl, -0-(CH2)q-Rg, -O-(CH2)q-O-Re, -NR3Rb,
-S(O)nRd, or heterocyclyl;
R6 is H, -CN, -OH, halogen, alkyl, aryl, alkoxy, -NRaRb, -C(O)Rc, -S(O)nRd, or heterocyclyl; R7 is H, -OH, halogen, alkyl, aryl, alkoxy, -NR3Rb, -S(O)nRd, or heterocyclyl; each Ra and Rb is independently H, alkyl, -C(O)R0, aryl, heterocyclyl, or alkoxy; or,
R3 and Rb are bound together to form Ri-2; each R0 is independently H, alkyl, alkoxy, -C(O)alkyl, -C(O)aryl, -CHO, aryl, or heterocyclyl; each Rj is independently H, alkyl, alkenyl, aryl, or -NR3Rb; each R3 is independently H or alkyl;
Rf is H, halogen, -OH, -CN, -(CH2)qNRaRh, alkoxy, -C(O)R0, -(CH2)qCH3. each Rg is independently H, halogen, -C(O)R0, aryl, heterocyclyl, or -NRaRb;
Rh is H, or -(CH2)qS(O)nRd; each Rj is independently H, halo, alkyl, alkenyl, alkynyl, or -0(CH2)q-Rg; each n is independently 0, 1, or 2; each p is independently 0, 1, 2, or 3; each q is independently 0, 1, or 2; Ri-2 has the general structure as shown:
Figure imgf000021_0001
wherein, each R8 is independently H, -OH, halogen, alkyl, alkoxy, -NRaRb, or -S(O)nRa; each R9 is independently H, alkyl, -C(O)Rc, or absent if X is O, S, or absent; each X is independently O, S, N, CH, or absent, thereby forming a covalent bond; and each m is independently 0, 1, or 2; or a tautomer thereof, a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable salt of the tautomer.
In a more particular embodiment thereof, said compound is:
Figure imgf000021_0002
, a tautomer thereof, a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable salt of the tautomer. In a more particular embodiment thereof, said compound is:
Figure imgf000021_0003
, a tautomer thereof, a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable salt of the tautomer.
Another embodiment of the invention provides a method of treating a patient suffering from cancer comprising administering aldesleukin and a compound of formula II:
Figure imgf000022_0001
III wherein, the dotted line represents an optional placement of an additional bond;
R20 is H or =0;
R-2U R22, R23, and R24 are each independently H, -CN, -OH, halogen, alkyl, aryl, alkoxy, -NRaRb, -C(O)R2, -S(O)nRd, or heterocyclyl;
R25 is H, alkyl, or -C(O)R2; R26 and R27 are each independently H, -OH, halogen, alkyl, -NRaRb, -C(O)R2, or
-S(O)nRd;
R28 is H, -CH3, or halogen; each Ra and Rb is independently H, alkyl, -C(O)R2, aryl, heterocyclyl, or alkoxy; each R2 is independently H, alkyl, alkoxy, -NH2, -NH(alkyl), -N(alkyl)2, aryl, or heterocyclyl; each Ra is independently H, alkyl, alkenyl, aryl, or -NRaRt,; each n is independently O, 1 , or 2; and each q is independently O, 1 , or 2; or a tautomer thereof, a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable salt of the tautomer.
In a more particular embodiment thereof, said compound is:
Figure imgf000022_0002
, a tautomer thereof, a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable salt of the tautomer.
In a more particular embodiment thereof, said compound is:
Figure imgf000022_0003
a tautomer thereof, a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable salt of the tautomer.
-21- Another embodiment of the invention provides a method of decreasing the toxicity associated with the administration of aldesleukin to a cancer patient comprising administering a quinolinone, preferably an antiangiogenic agent, preferably selected from 6,7-bis(2-methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4- amine, 6-(3-morpholinopropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin- 4-amine, N-(2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)- 2,4-diethyl- 1 H-pyrrole-3 -carboxamide, N-(4-chlorophenyl)-4-((pyridin-4- yl)methyl)phthalazin- 1 -amine, or 1 -(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)- 3-(4-chloro-3-(trifluoromethyl)phenyl)urea, to the patient. Another embodiment of the invention provides a method of decreasing the toxicity associated with the administration of an antiangiogenic agent, preferably selected from 6,7-bis(2-methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6- (3-moφholinopropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl- lH-pyrrole-3-carboxamide, N-(4-chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin-l- amine, or 1 -(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluoromethyl)phenyl)urea, to a cancer patient comprising administering aldesleukin to the patient.
Another embodiment of the invention provides a method of decreasing IL-2 resistance in a patient suffering from cancer comprising administering an antiangiogenic agent, preferably selected from 6,7-bis(2-methoxyethoxy)-N-(3- ethynylphenyl)quinazolin-4-amine, 6-(3 -morpholinopropoxy)-N-(3 -chloro-4- fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2-(dimethylamino)ethyl)-5-((5- fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl-lH-pyrrole-3-carboxamide, N-(4- chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin-l -amine, or l-(4-(2- (methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluoromethyl)phenyl)urea to said patient.
Another embodiment of the invention provides a method for treating a patient suffering from cancer, comprising the steps of first administering to said patient a therapeutically effective amount of aldesleukin followed by administration of an antiangiogenic agent, preferably selected from 6,7-bis(2-methoxyethoxy)-N-(3- ethynylphenyl)quinazolin-4-amine, 6-(3 -morpholinopropoxy) -N-(3 -chloro-4- fluoiOphenyl)-7-methoxyquinazolin-4-amine, N-(2-(dimethylamino)ethyl)-5-((5- fiuoro-2-oxoindolin-3 -ylidene)methyl)-2,4-diethyl- 1 H-pyrrole-3 -carboxamide, N-(4-
22- chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin- 1 -amine, or 1 -(4-(2-
(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3-
(trifluoromethyl)phenyl)urea.
Another embodiment of the invention provides a method for treating a patient suffering from cancer, comprising separately administering to said patient a therapeutically effective amount of an antiangiogenic agent, preferably selected from 6,7-bis(2-methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6-(3- moφholinoρropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N- (2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl- lH-ρyrrole-3-carboxamide, N-(4-chlorophenyl)-4-((pyridm-4-yl)methyl)phthalazin-l- amine, or 1 -(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluoromethyl)phenyl)urea and aldesleukin according to a dosing schedule, wherein the aldesleukin is administered from 1 to 3 times daily in a dose between about 9 and about 130 MIU/day for a period of at least 3 consecutive days, optionally followed by a rest period of at least 3 consecutive days. More specifically the dose is between about 30 and about 100 MIU/day. More specifically the dose is between about 30 and about 60 MIU/day. More specifically the dose is between about 30 and about 40 MIU/day. More specifically the dose is between about 17 and about 30 MIU/day. More specifically the dose is between about 9 and about 30 MIU/day. In a more particular embodiment said antiangiogenic agent, preferably selected from 6,7-bis(2-methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6- (3-morpholinopropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl- lH-ρyrrole-3-carboxamide, N-(4-chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin-l- amine, or l-(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3-
(trifluoromethyl)phenyl)urea is administered from 1 to 9 times every 2-3 weeks. In a more particular embodiment said antiangiogenic agent, preferably selected from 6,7- bis(2-methoxyethoxy)-N-(3 -ethynylphenyl)quinazolin-4-amine, 6-(3 - morpholinopropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N- (2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl- lH-pyrrole-3-carboxamide, N-(4-chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin-l- amine, or 1 -(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluoromethyl)phenyl)urea is a VEGF inhibitor.
-23- In another embodiment the antiangiogenic agent, preferably selected from 6,7- bis(2-methoxyethoxy)-N-(3-ethynylρhenyl)quinazolin-4-amine, 6-(3- morpholinopropoxy)-N-(3-chloro-4-fluoroρhenyl)-7-methoxyquinazolin-4-amine, N- (2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl- 1 H-pyrrole-3 -carboxamide, N-(4-chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin- 1 - amine, or l-(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluoromethyl)phenyl)urea is also administered with bevacizumab, or cetuximab, or VEGF-Trap. More specifically, said bevacizumab is administered in a dose between 1 and 12 mg/kg once every 12-16 days. In another embodiment, the antiangiogenic agent is administered within 72 hours of the administration of rIL-2; wherein "within" is meant to indicate before or after, as in: the antiangiogenic agent is administered 72 hours before or 72 hours after the administration of rIL-2. In another embodiment, the antiangiogenic agent is administered within 14 days of the administration of rIL-2. In another embodiment, the antiangiogenic agent is administered within 7 days of the administration of rIL-2. In another embodiment, the antiangiogenic agent is administered within 6 days of the administration of rIL-2. In another embodiment, the antiangiogenic agent is administered within 5 days of the administration of rIL-2. In another embodiment, the antiangiogenic agent is administered within 4 days of the administration of rIL-2. In another embodiment, the antiangiogenic agent is administered within 48 hours of the administration of rIL-2. In another embodiment, the antiangiogenic agent is administered within 36 hours of the administration of rIL-2. In another embodiment, the antiangiogenic agent is administered within 24 hours of the administration of rlL- 2. In another embodiment, the antiangiogenic agent is administered within 12 hours of the administration of rIL-2. In another embodiment, the antiangiogenic agent is administered within 6 hours of the administration of rIL-2. In another embodiment, the antiangiogenic agent is administered within 1 hour of the administration of rIL-2. In another embodiment, the antiangiogenic agent is administered at the same time as the administration of rIL-2. In another embodiment, the antiangiogenic agent is administered in combination with rIL-2. In a more particular embodiment thereof the antiangiogenic agent is selected from 6,7-bis(2-methoxyethoxy)-N-(3~ ethynylphenyl)quinazolin-4-amine, 6-(3-morpholinopropoxy)-N-(3-chloro-4- fiuorophenyl)-7-methoxyquinazolin-4-amine, N-(2-(dimethylamino)ethyl)-5-((5- fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl-lH-pyrrole-3-carboxamide, N-(4-
-24- chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin- 1 -amine, or 1 -(4-(2-
(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3-
(trifluoromethyl)phenyl)urea.
Another embodiment of the invention provides a method of treating a patient suffering from cancer comprising co-administering aldesleukin and an antiangiogenic agent, preferably selected from 6,7-bis(2-methoxyethoxy)-N-(3- ethynylphenyl)quinazolin-4-amine, 6-(3-morpholinopropoxy)-N-(3-chloro-4- fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2-(dimethylamino)ethyl)-5~((5- fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl-lH-pyrrole-3-carboxamide, N-(4- chloroρhenyl)~4-((pvridin-4-yl)methyl)phthalazin-l -amine, or l-(4-(2- (methylcarbainoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluoromethyl)phenyl)urea.
Another embodiment of the invention provides a therapeutic package suitable for commercial sale for treating a patient suffering from cancer, comprising a container, a therapeutically effective amount of aldesleukin, and a therapeutically effective amount of an antiangiogenic agent, preferably selected from 6,7-bis(2- methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6-(3-moφholinopropoxy)- N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2- (dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl-lH- pyrrole-3 -carboxamide, N-(4-chlorophenyl)-4-((pyridin-4-yl)methyl)ρhthalazin- 1 - amine, or 1 -(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifiuoromethyl)phenyl)urea. A therapeutic package thereof, wherein said patient is suffering from renal cell carcinoma. A therapeutic package thereof, further comprising written matter instructing that the patient receive treatment with aldesleukin prior to treatment with the antiangiogenic agent, preferably selected from 6,7-bis(2-methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6-(3- morpholinopropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N- (2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl- lH-pyrrole-3-carboxamide, N-(4-chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin-l- amine, or l-(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluoromethyl)phenyl)urea.
Another embodiment provides a pharmaceutical composition comprising a an antiangiogenic agent, preferably selected from 6,7-bis(2-methoxyethoxy)-N-(3- ethynylphenyl)quinazolin-4-amine, 6-(3-morpholinopropoxy)-N-(3-chloro-4-
-25- fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2-(dimethylamino)ethyl)-5-((5- fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl-lH-pyrrole-3-carboxamide, N-(4- chlorophenyl)-4-((pyridin-4-yl)methyl)phtlialazin-l -amine, or 1 -(4-(2- (memylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (1xifluoromethyl)phenyl)urea and aldesleukin.
Clear cell renal cell carcinoma (RCC) is associated with increased vascularization, particularly VEGF expression. VEGF is a protein that plays a crucial role in tumor angiogenesis (the formation of new blood vessels to the tumor) and maintaining established tumor blood vessels. It binds to specific receptors on blood vessels to stimulate extensions to existing blood vessels. Some but not all cases of RCCa have increased serum VEGF levels.
There is evidence that increased VEGF correlates with resistance to IL-2 (Lissoni et al, Anticancer Res. 2001; 21: 777-9). The immunosuppressive effects of VEGF on T-cells and dendritic cells (Gabrilovich et al, J Leukoc Biol. 2002; 72: 285- 96) could represent a mechanism for VEGF induced resistance to IL-2 treatment.
In addition, VEGF increases vascular permeability and treatment with anti- VEGF has been noted to lead to increased blood pressure of some patients. It is therefore likely that some of the major toxicities of anti-VEGF (e.g. hypertension) and rIL-2 (e.g. hypotension) would be mutually counteracted and lead to an overall improved therapeutic index if the combinations of the present invention were administered to patients suffering from cancer, such as RCCa.
Further combinations of the present invention, such as aldesleukin with bevacizumab or cetuximab yield a higher proportion and/or better durability of responses compared to the agents administered alone. The particular aldesleukin dosing regimens disclosed herein provide for intermittent stimulation of natural killer (NK) cell activity and decreased risk of rIL-2- related side effects that can be associated with long term exposure to rIL-2 dosing. Of note, hypotension typically associated with rIL-2 dosing is offset by the administration of the antiangiogenic compositions of the present invention, since they are generally associated with hypertension. Further, hypoxia typically associated with VEGF inhibitors will increase sensitivity to rIL-2 treatment efficacy, while simultaneously providing palliative benefits as well.
Another embodiment of the invention provides the use of aldesleukin in the manufacture of a medicament for treating cancer, wherein said medicament is for
-26- separate, simultaneous, or sequential use with an antiangiogenic agent, preferably selected from 6,7-bis(2-methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6- (3-morpholinopropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2-(dime1hylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl- 1 H-pyrrole-3 -carboxamide, N-(4-chloropb.enyl)-4-((pyridin-4-yl)methyl)phtb.alazin- 1 - amine, or l-(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluoromethyl)phenyl)urea. In a more particular embodiment thereof said antiangiogenic agent, preferably selected from 6,7-bis(2-methoxyethoxy)-N-(3- ethynylphenyl)quinazolin-4-amine, 6-(3-morpholinopropoxy)-N-(3-chloro-4- fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2-(dimethylamino)ethyl)-5-((5- fluoro-2-oxomdolin-3-ylidene)methyl)-2,4-diethyl-lH-pyrrole-3-carboxamide, N-(4- chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin- 1 -amine, or 1 ~(4-(2- (methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro~3- (trifluoromethyl)phenyl)urea is a lactate salt. In another embodiment said cancer is renal cell carcinoma.
Another embodiment of the invention provides the use of an antiangiogenic agent, preferably selected from 6,7-bis(2-methoxyethoxy)-N-(3- ethynylphenyl)quinazolin-4-amine, 6-(3-morpholinoρropoxy)-N-(3-chloro-4- fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2-(dimethylamino)ethyl)-5-((5- fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl-lH-pyrrole-3-carboxamide, N-(4- chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin- 1 -amine, or 1 ~(4-(2- (methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3-
(trifluoromethyl)phenyi)urea in the manufacture of a medicament for treating cancer, wherein said medicament is for separate, simultaneous, or sequential use with aldesleukin.
Preferred molecules associated with angiogenesis, modulated (such as inhibition) by the compositions of the present invention include: Vascular Endothelial Growth Factor (VEGF), Fibroblast Growth Factor (FGF), Interleukin-8 (IL-8), Angiogenin, Angiotropin, Epidermal Growth Factor (EGF), Platelet Derived Endothelial Cell Growth Factor, Transforming Growth Factor a (TGF-a),
Transforming Growth Factor b (TGF-b), or Nitric Oxide. Also modulation (such as potentiation) by the compositions of the present invention of Thrombospondin, Angiostatin, and Endostatin is contemplated within the present invention.
-27- T/US2006/005720
General Compositions for (Jo-administration with rIL-2: QUINAZOLINE
Compounds of Formula I have the following structure:
Figure imgf000029_0001
wherein,
Ri is alkyl, -aryl(ROP, or heterocyclyl;
R2 is H or alkyl; or,
Ri and R2 are bound together to form Ri-2; R3 is H, -CN, -OH, halogen, alkyl, aryl, alkoxy, -NRaRb, -C(O)Rc, -S(O)nRd, or heterocyclyl;
R4Is H, -CN, -OH, halogen, alkyl, aryl, -0-(CH2)q-Rg, -0-(CH2)C1-O-R6, -NRaRb,
-S(O)nRd, or -heterocyclyl-Rf;
R5 is H, -CN, -OH, halogen, alkyl, aryl, -0-(CH2)q-Rg> -0-(CH2)q-0-Re, -NRaRb, -S(O)nRd, or heterocyclyl;
R6 is H, -CN, -OH, halogen, alkyl, aryl, alkoxy, -NR3Rb, -C(O)R0, -S(O)nRd, or heterocyclyl;
R7 is H, -OH, halogen, alkyl, aryl, alkoxy, -NR3Rb, -S(O)nRd, or heterocyclyl; each Ra and Rb is independently H, alkyl, -C(O)R0, aryl, heterocyclyl, or alkoxy; or, Ra and Rb are bound together to form Ri-2; each R0 is independently H, alkyl, alkoxy, -C(O)alkyl, -C(O)aryl, -CHO, aryl, or heterocyclyl; each Rd is independently H, alkyl, alkenyl, aryl, or -NRaRb; each Re is independently H or alkyl; Rf is H, halogen, -OH, -CN, -(CH2)qNRaRh, alkoxy, -C(O)Rc, -(CH2)qCH3. each Rg is independently H, halogen, -C(O)R0, aryl, heterocyclyl, or -NR3Rb;
Rh is H, or -(CH2)qS(O)nRd; each Ri is independently H, halo, alkyl, alkenyl, alkynyl, or -0(CH2)q-Rg; each n is independently O, 1, or 2; each p is independently O, 1 , 2, or 3;
-28- each q is independently 0, 1, or 2; Ri-2 has the general structure as shown:
Figure imgf000030_0001
wherein, each Rg is independently H, -OH, halogen, alkyl, alkoxy, -NR8Rb, or -S(O)nRa; each R9 is independently H, alkyl, -C(O)R0, or absent if X is O, S, or absent; each X is independently O, S, N, CH, or absent, thereby forming a covalent bond; and each m is independently O, 1, or 2.
In a more particular embodiment of the invention R2 is H. In a more particular embodiment still, Ri is -aryl(Ri)p. In a further embodiment wherein Ri is -aryl(Rj)P; aryl within Rj is phenyl, p within Ri is 2 and both Ri groups within Ri are halo.
In a further embodiment wherein Ri is -aryl(Ri)p, aryl within Ri is phenyl, p within Ri is 1 and R; groups within Ri is alkynyl, preferably ethynyl.
In a further embodiment wherein Ri is -aryl(Rj)P) aryl within Ri is phenyl, p within Ri is 2, one R; group within Ri is halo and the other R; group within Ri is - O(CH2)q-Rg. In a more particular embodiment thereof, q within Ri is 1 and Rg within R] is halophenyl.
In a further embodiment wherein Ri is -aryl(Ri)p, p within Ri is 1 and K\ within Ri is alkynyl. Another more particular embodiment of the invention is provided, wherein Ri and R2 are bound together to form Ri-2:
Figure imgf000030_0002
wherein, R8 is H, X is N, and R9 is -C(O)NHRb. In a more particular embodiment thereof, Rb within R9 is -phenyl-O-CH2(CH3)2. In a more particular embodiment of the invention, R3 and R6 are H.
In a more particular embodiment of the invention R4 is -O-(CH2)q-Rg. In a more particular embodiment thereof, q within R4 is 1 and Rg is H.
In another embodiment wherein R4 is -O-(CH2)q-Rg, Rg within R4 is heterocyclyl.
-29- In a more particular embodiment of the invention R4 and R5 are each -O- (CH2)q-O-Re. In a more particular embodiment thereof, q within both R4 and R5 is 2 and Re within both R4 and R5 is methyl.
In a more particular embodiment of the invention R4 is -heterocyclyl-Rf and R5 is H. In a more particular embodiment thereof, said heterocyclyl within R4 is furanyl. In a more particular embodiment still, Rf within R4 is -(CH2)qNHRh. Further still, Rh
Figure imgf000031_0001
In a more particular embodiment of the invention R5 is -O-(CH2)q-Rg. In another embodiment thereof, Rg within R5 is heterocyclyl. Another embodiment is provided wherein R7 is H.
Another embodiment is provided wherein R3, R6, and R7 are all H. INDOLINONE
Compounds of Formula II have the following structure:
Figure imgf000031_0002
II wherein,
Ri i is alkyl, aryl or heterocyclyl;
Ri2, Ri 3 Ri4, and Ri5 are each independently H, -CN, -OH, halogen, alkyl, aryl, alkoxy, -NRaRb, -C(O)R2, -S(O)nRd, or heterocyclyl; each R3 and Rb is independently H, alkyl, -C(O)R2, aryl, heterocyclyl, or alkoxy; each Rz is independently H, alkyl, alkoxy, -NH2, -NH(alkyl), -N(alkyl)2, aryl, or heterocyclyl; each Rd is independently H, alkyl, alkenyl, aryl, or -NR3Rb", each n is independently 0, 1, or 2; and each q is independently 0, 1, or 2.
-30- in a more particular embodiment, Rn is heterocyclyl. In a more particular embodiment still, Rn is Ri ia:
Figure imgf000032_0001
wherein,
Ri6, Ri7 and Rj8 are each independently H, -CN, -OH, halogen, alkyl, aryl, alkoxy, - NR3Rb, -C(O)R2, -S(O)nRd, or heterocyclyl. In yet another more particular embodiment, wherein Ri is Ri a, R7 is -C(O)-(CH2)p-N(H)(2-r)(alkyl)r, wherein p is O, 1, 2, 3, 4, or 5 and r is 0, 1 , or 2. In yet another more particular embodiment, wherein Ri i is Ri ia, RB is F. In yet another more particular embodiment, wherein Ri i is Rl la, Ri7 is -(CH2)tCOOH, wherein t is 1, 2, 3, or 4. In yet another more particular embodiment, wherein Rn is Rl la, R6 and R8 are both methyl. In yet another more particular embodiment, wherein Rn is Rn3, Ri7 is H.
In another more particular embodiment of the invention, Ri i is aryl. More particular still, Ri i is substituted or unsubstituted phenyl. In another embodiment R is iodide. ISOINDOLINONES
Compounds of Formula III have the following structure:
Figure imgf000032_0002
wherein, the dotted line represents an optional placement of an additional bond;
R20 is H or =0;
R2I, R22, R23, and R24 are each independently H, -CN, -OH, halogen, alkyl, aryl, alkoxy, -NR3Rb, -C(O)R2, -S(O)nRd, or heterocyclyl; R25 is H, alkyl, or -C(O)R2;
R26 and R27 are each independently H, -OH, halogen, alkyl, -NR3Rb, -C(O)R2, or
-S(O)nRd;
R28 is H, -CH3, or halogen; each R3 and Rb is independently H, alkyl, -C(O)R2, aryl, heterocyclyl, or alkoxy;
P/ """ ™ -31-
Ai each Rz is independently H, aiicyl, alkoxy, -NH2, -NH(alkyl), -N(alkyl)2, aryl, or heterocyclyl; each Ra is independently H, alkyl, alkenyl, aryl, or -NRaRb; each n is independently 0, 1, or 2; and each q is independently 0, 1, or 2.
DEFINITIONS:
AUC Area under the curve
CR Complete response DLT Dose limiting toxicity
IL Interleukin
IL-2 Interleukin-2
IU International units
IV Intravenous (as a mode of administration) LAK Lymphokine activated killer
MTD Maximum tolerated dosage
MIU Million international units
NK Natural killer
PK Pharmacokinetic PR Partial response
RCC Renal cell carcinoma
RCCa Clear cell renal cell carcinoma rIL-2 Recombinant interleukin-2
RTK Receptor tyrosine kinase TNF Tumor necrosis factor
VEGF Vascular endothelial growth factor
Generally, reference to a certain element such as hydrogen or H is meant to include all isotopes of that element. For example, if an R group is defined to include hydrogen or H, it also includes deuterium and tritium.
By "antiangiogenic agent" is meant any small molecule, more specifically, any compound having a molecular weight less than 1,100 g/mol that has been shown or will be shown to suppress angiogensis in a system. Preferred molecules associated with angiogenesis, modulated (such as inhibition) by the compositions of the present
-32- invention include: Vascular Endothelial Growth Factor (VEGF), Fibroblast Growth Factor (FGF), Interleukin-8 (IL-8), Angiogenin, Angiotropin, Epidermal Growth Factor (EGF), Platelet Derived Endothelial Cell Growth Factor, Transforming Growth Factor a (TGF-a), Transforming Growth Factor b (TGF -b), or Nitric Oxide. Also modulation (such as potentiation) by the compositions of the present invention of Thrombospondin, Angiostatin, and Endostatin is contemplated within the present invention. Preferred antiangiogenic compositions of the present invention include: 6,7-bis(2-methoxyethoxy)-N-(3 -ethynylphenyl)quinazolin-4-amine, 6-(3 - moφholinopropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N- (2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl- 1 H-ρyrrole-3-carboxamide, N-(4-chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin- 1 - amine, and l-(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluoromethyl)phenyl)urea.
The term "effective amount" is an amount necessary or sufficient to realize a desired biological effect. For example, an effective amount of a compound to treat renal cell carcinoma may be an amount necessary to cause regression of tumor growth in renal cells. The effective amount may vary, depending, for example, upon the condition treated, weight of the subject and severity of the disease. One of skill in the art can readily determine the effective amount empirically without undue experimentation.
As used herein "an effective amount for treatment" refers to an amount sufficient to palliate, ameliorate, stabilize, reverse, slow or delay progression of a condition such as a disease state.
A "subject" or "patient" is meant to describe a human or vertebrate animal including a dog, cat, pocket pet, marmoset, horse, cow, pig, sheep, goat, elephant, giraffe, chicken, lion, monkey, owl, rat, squirrel, slender loris, and mouse.
A "pocket pet" refers to a group of vertebrate animals capable of fitting into a commodious coat pocket such as, for example, hamsters, chinchillas, ferrets, rats, guinea pigs, gerbils, rabbits and sugar gliders. As used herein, the term "pharmaceutically acceptable ester" refers to esters, which hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof. Suitable ester groups include, for example, those derived from pharmaceutically acceptable aliphatic carboxylic acids, particularly alkanoic, alkenoic, cycloalkanoic and alkanedioic acids, in which each
-33- /vπy jJOCKet JNo. FFUU23636.0004 alkyl or alkenyl moiety advantageously has not more than 6 carbon atoms. Representative examples of particular esters include, but are not limited to, formates, acetates, propionates, butyrates, acrylates and ethylsuccinates.
The compounds of the present invention can be used in the form of salts as in "pharmaceutically acceptable salts" derived from inorganic or organic acids. These salts include but are not limited to the following: acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphorsulfonate, digluconate, cyclopentanepropionate, dodecylsulfate, ethanesulfonate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, fumarate, hydrochloride, hydrobromide, hydroiodide,
2-hydroxyethanesulfonate, lactate, maleate, methanesulfonate, nicotinate, 2-napth- alenesulfonate, oxalate, pamoate, pectinate, sulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, p-toluenesulfonate and undecanoate. Also, the basic nitrogen-containing groups can be quaternized with such agents as loweralkyl halides, such as methyl, ethyl, propyl, and butyl chloride, bromides, and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides, and others. "Water or oil-soluble or dispersible products are thereby obtained. The term "pharmaceutically acceptable prodrugs" as used herein refers to those prodrugs of the compounds of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals with undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the invention. The term "prodrug" refers to compounds that are rapidly transformed in vivo to yield the parent compound of the above formula, for example by hydrolysis in blood. A thorough discussion is provided in T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems, Vol. 14 of the A.C.S. Symposium Series, and in Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical
Association and Pergamon Press, 1987. Prodrugs as described in U.S. Patent No. 6,284,772 for example may be used.
Reference to "halo," "halide," or "halogen" refers to F, Cl, Br, or I atoms.
-34- The phrase "alkyl" refers to substituted and unsubstituted alky groups such as methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, undecyl, dodecyl and the like. The phrase also includes branched chain isomers of straight chain alkyl groups, including but not limited to, the following which are provided by way of example: -CH(CH3)2, -CH(CH3)(CH2CH3), -CH(CH2CH3)2, -C(CH3)3, -C(CH2CH3)3) -CH2CH(CH3)2, -CH2CH(CH3)(CH2CH3), -CH2CH(CH2CH3)2, -CH2C(CH3)3, -CH2C(CH2CH3)3, -CH(CH3)CH(CH3)(CH2CH3), -CH2CH2CH(CH3)2, -CH2CH2CH(CH3)(CH2CH3), -CH2CH2CH(CH2CH3)S, -CH2CH2C(CH3)3, - CH2CH2C(CH2CH3)3, -CH(CH3)CH2CH(CH3)2, -CH(CH3)CH(CH3)CH(CH3)2, - CH(CH2CH3)CH(CH3)CH(CH3)(CH2CH3), and others. The phrase also includes cyclic alkyl groups such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl and such rings substituted with straight and branched chain alkyl groups as defined above. The phrase also includes polycyclic alkyl groups such as, but not limited to, adamantyl norbornyl, and bicyclo[2.2.2]octyl and such rings substituted with straight and branched chain alkyl groups as defined above. The phrase alkyl also includes groups in which one or more bonds to a carbon(s) or hydrogen(s) are replaced by a bond to non-hydrogen and non-carbon atoms such as, but not limited to, a halogen atom in halides such as F, Cl, Br, and I; and oxygen atom in groups such as hydroxyl groups, alkoxy groups, aryloxy groups, and ester groups; a sulfur atom in groups such as thiol groups, alkyl and aryl sulfide groups, sulfone groups, sulfonyl groups, and sulfoxide groups; a nitrogen atom in groups such as amines, amides, alkylamines, dialkylamines, arylamines, alkylarylamines, diarylamines, N-oxides, imides, and enamines; a silicon atom in groups such as in trialkylsilyl groups, dialkylarylsilyl groups, alkyldiarylsilyl groups, and triarylsilyl groups; and other heteroatoms in various other groups. Alkyl groups are those limited to having 1 to 20 carbon atoms and as many as 5 additional heteroatoms as described above. More preferred alkyl groups have from 1 to 5 carbon atoms and as many as 2 heteroatoms.
The phrase "aryl" refers to substituted and unsubstituted aryl groups that do not contain heteroatoms. Thus the phrase includes, but is not limited to, groups such as phenyl, biphenyl, anthracenyl, naphthenyl by way of example. Aryl groups also include those in which one of the aromatic carbons is bonded to a non-carbon or non- hydrogen atoms described above (in the alkyl definition) and also includes aryl groups in which one or more aromatic carbons of the aryl group is bonded to a substituted
-35- and/or unsubstituted alkyl, alkenyl, or alkynyl group as defined herein. This includes bonding arrangements in which two carbon atoms of an aryl group are bonded to two atoms of an alkyl, alkenyl, or alkynyl group to define a fused ring system (e.g. dihydronaphthyl or tetrahydronaphthyl). Thus, the phrase "aryl" includes, but is not limited to tolyl, and hydroxyphenyl among others.
The phrase "alkenyl" refers to straight and branched chain and cyclic groups such as those described with respect to alkyl groups as defined above, except that at least one double bond exists between two carbon atoms. Examples include, but are not limited to vinyl, -CH=C(H)(CH3), -CH=C(CH3)2, -C(CH3)=C(H)2, - C(CHs)=C(H)(CH3), -C(CH2CHs)=CH2, cyclohexenyl, cyclopentenyl, cyclohexadienyl, butadienyl, pentadienyl, and hexadienyl among others. Included as well are groups in which a non-carbon or non-hydrogen atom is bonded to a carbon double bonded to another carbon and those in which one of the non-carbon or non- hydrogen atoms are bonded to a carbon not involved in a double bond to another carbon. Alkenyl groups are those limited to having 2 to 15 carbon atoms and as many as 4 additional heteroatoms as described above. More preferred alkenyl groups have from 2 to 5 carbon atoms and as many as 2 heteroatoms.
The phrase "alkoxy" refers to substituted or unsubstituted alkoxy groups of the formula -O-alkyl, wherein the point of attachment is the oxy group and the alkyl group is as defined above. Alkoxy groups are those limited to having 1 to 20 carbon atoms and as many as 5 additional heteroatoms, including the oxygen atom. More preferred alkoxy groups have from 1 to 5 carbon atoms and as many as 2 heteroatoms, including the oxygen atom.
The phrase "alkynyl" refers to straight and branched chain groups such as those described with respect to alkyl groups as defined above, except that at least one triple bond exists between two carbon atoms. Examples include, but are not limited to -C≡C(H), -CsC(CH3), -CsC(CH2CH3), -C(H2)C≡C(H), -C(H)2C=C(CH3), and - C(H)2C≡C(CH2CH3) among others. Included as well are alkynyl groups in which a non-carbon or non-hydrogen atom is bonded to a carbon triple bonded to another carbon and those in which a non-carbon or non-hydrogen atom is bonded to a carbon not involved in a triple bond to another carbon. Alkynyl groups are those limited to having 2 to 15 carbon atoms and as many as 4 additional heteroatoms as described
-36- above. More preferred alkynyl groups have from 2 to 5 carbon atoms and as many as 2 heteroatoms.
The phrase "heterocyclyl" refers to both aromatic and nonaromatic ring compounds including monocyclic, bicyclic, and polycyclic ring compounds such as, but not limited to, quinuclidyl, containing 3 or more ring members of which one or more is a heteroatom such as, but not limited to, N, O, and S. Examples of heterocyclyl groups include, but are not limited to: unsaturated 3 to 8 membered rings containing 1 to 4 nitrogen atoms such as, but not limited to pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, pyridyl, dihydropyridyl, pyrimidyl, pyrazinyl, pyridazinyl, triazolyl (e.g. 4H-l,2,4-triazolyl, lH-l,2,3-triazolyl, 2H-l,2,3-triazolyl etc.), tetrazolyl, (e.g. lH-tetrazolyl, 2H tetrazolyl, etc.); saturated 3 to 8 membered rings containing 1 to 4 nitrogen atoms such as, but not limited to, pyrrolidinyl, imidazolidinyl, piperidinyl, piperazinyl; condensed unsaturated heterocyclic groups containing 1 to 4 nitrogen atoms such as, but not limited to, indolyl, isoindolyl, indolinyl, indolizinyl, benzimidazolyl, quinolyl, isoquinolyl, indazolyl, benzotriazolyl; unsaturated 3 to 8 membered rings containing 1 to 2 oxygen atoms such as, but not limited to furanyl; unsaturated 3 to 8 membered rings containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms such as, but not limited to, oxazolyl, isoxazolyl, oxadiazolyl (e.g. 1,2,4-oxadiazolyl, 1,3,4-oxadiazolyl, 1,2,5-oxadiazolyl, etc.); saturated 3 to 8 membered rings containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms such as, but not limited to, morpholinyl; unsaturated condensed heterocyclic groups containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms, for example, benzoxazolyl, benzoxadiazolyl, benzoxazinyl (e.g. 2H-l,4-benzoxazinyl etc.); unsaturated 3 to 8 membered rings containing 1 to 3 sulfur atoms and 1 to 3 nitrogen atoms such as, but not limited to, thiazolyl, isothiazolyl, thiadiazolyl (e.g. 1,2,3-thiadiazolyl, 1,2,4-thiadiazolyl, 1,3,4-thiadiazolyl, 1,2,5-thiadiazolyl, etc.); saturated 3 to 8 membered rings containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms such as, but not limited to, thiazolodinyl; saturated and unsaturated 3 to 8 membered rings containing 1 to 2 sulfur atoms such as, but not limited to, thienyl, ) dihydrodithiinyl, dihydrodithionyl, tetrahydrothiophene, tetrahydrothiopyran; unsaturated condensed heterocyclic rings containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms such as, but not limited to, benzothiazolyl, benzothiadiazolyl, benzothiazinyl (e.g. 2H-l,4-benzothiazinyl, etc.), dihydrobenzothiazinyl (e.g. 2H-3,4- dihydrobenzothiazinyl, etc.), unsaturated condensed heterocyclic rings containing 1 to
-37-
Figure imgf000039_0001
wherein,
Ri i is alkyl, aryl or heterocyclyl;
Ri2, Ri3 Ri4, and R15 are each independently H, -CN, -OH, halogen, alkyl, aryl, alkoxy, -NRaRb, -C(O)R2, -S(O)nRd, or heterocyclyl; each Ra and Rb is independently H, alkyl, -C(O)R2, aryl, heterocyclyl, or alkoxy; each Rz is independently H, alkyl, alkoxy, -NH2, -NH(alkyl), -N(alkyl)2, aryl, or heterocyclyl; each Rd is independently H, alkyl, alkenyl, aryl, or -NRaRb; each n is independently 0, 1, or 2; and each q is independently 0, 1, or 2; or a tautomer thereof, a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable salt of the tautomer.
In a more particular embodiment thereof, Ri 1 is Rj ia:
Rla wherein,
Ri 6, Ri 7 and Ri8 are each independently H, -CN, -OH, halogen, alkyl, aryl, alkoxy, - NR3Rb, -C(O)R2, -S(O)nRd, or heterocyclyl.
In a more particular embodiment thereof, said compound is:
Figure imgf000039_0003
, a tautomer thereof, a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable salt of the tautomer.
Another embodiment of the invention provides a method of treating a patient suffering from cancer comprising administering aldesleukin and a compound of formula III:
-20- 2 oxygen atoms such as benzodioxolyl (e.g. 1,3-benzodioxoyl, etc.); unsaturated 3 to 8 membered rings containing an oxygen atom and 1 to 2 sulfur atoms such as, but not limited to, dihydrooxathiinyl; saturated 3 to 8 membered rings containing 1 to 2 oxygen atoms and 1 to 2 sulfur atoms such as 1,4-oxathiane; unsaturated condensed rings containing 1 to 2 sulfur atoms such as benzothienyl, benzodithiinyl; and unsaturated condensed heterocyclic rings containing an oxygen atom and 1 to 2 oxygen atoms such as benzoxathiinyl. Heterocyclyl group also include those described above in which one or more S atoms in the ring is double-bonded to one or two oxygen atoms (sulfoxides and sulfones). For example, heterocyclyl groups include tetrahydrothiophene, tetrahydrothiophene oxide, and tetrahydrothiophene 1,1- dioxide. Preferred heterocyclyl groups contain 5 or 6 ring members. More preferred heterocyclyl groups include morpholine, piperazine, piperidine, pyrrolidine, imidazole, pyrazole, 1,2,3-triazole, 1,2,4-triazole, tetrazole, thiomorpholine, thiomorpholine in which the S atom of the thiomorpholine is bonded to one or more O atoms, pyrrole, homopiperazine, oxazolidin-2-one, pyrrolidin-2-one, oxazole, quinuclidine, thiazole, isoxazole, furan, and tetrahydrofuran. "Heterocyclyl" also refers to those groups as defined above in which one of the ring members is bonded to a non-hydrogen atom such as described above with respect to substituted alkyl groups and substituted aryl groups. Examples, include, but are not limited to, 2- methylbenzimidazolyl, 5-methylbenzimidazolyl, 5-chlorobenzthiazolyl, 1 -methyl piperazinyl, and 2-chloropyridyl among others. Heterocyclyl groups are those limited to having 2 to 15 carbon atoms and as many as 6 additional heteroatoms as described above. More preferred heterocyclyl groups have from 3 to 5 carbon atoms and as many as 2 heteroatoms. The term "substituted" as applied to an undefined, yet well known in the art group, such as phenyl, will have the same meaning with respect to the optional appendages as described in the definition of alkyl.
Within the present invention it is to be understood that compounds described herein may exhibit the phenomenon of tautomerism and that the formulae drawings within this specification can represent only one of the possible tautomeric forms. It is to be understood that the invention encompasses any tautomeric form which possesses antiangiogenic activity and is not to be limited merely to any one tautomeric form utilised within the formulae drawings. It is also to be understood that certain compounds and embodiments of the invention can exist in solvated as well as unsolvated forms such as, for example, hydrated forms. It is to be understood that the invention encompasses all such solvated forms which possess antiangiogenic activity. The invention also includes isotopically-labeled compounds, that are structurally identical to those disclosed above, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulfur, fluorine and chlorine, such as 2H, 3H, 13C, 14C, 15N, 180, 170, 31P, 32P, 35S, 18F and 36Cl, respectively. Compounds of the present invention, prodrugs thereof, and pharmaceutically acceptable salts of said compounds and of said prodrugs that contain the aforementioned isotopes and/or other isotopes of other atoms are within the scope of this invention. Certain isotopically labeled compounds of the present invention, for example those into which radioactive isotopes such as H and C are incorporated, are useful in drug and/or substrate tissue distribution assays. Tritiated, i.e., 3H, and carbon-14, i.e., 14C, isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium, i.e., 2H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements and, hence, may be preferred in some circumstances. Isotopically labeled compounds of this invention and prodrugs thereof can generally be prepared by carrying out known or referenced procedures and by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent.
Reference to "IL-2" or "interleukin-2" indicates a lymphocyte that is produced by normal peripheral blood lymphocytes and is present in the body at low concentrations. IL-2 was first described by Morgan et al. (1976) Science 193:1007- 1008 and originally called T-cell growth factor because of it's ability to induce proliferation of stimulated T lymphocytes. It is a protein with a reported molecular weight in the range of 13,000 to 17,000 (Gillis and Watson (1980) J. Exp. Med. 159: 1709) and has an isoelectric point in the range of 6-8.5. For purposes of the present invention, the term "IL-2" is intended to encompass any source of IL-2, including mammalian sources such as, e.g., mouse, rat, rabbit, primate, pig, pocket pet, and human, and may be native or obtained by recombinant techniques, such as recombinant IL-2 polypeptides produced by microbial hosts. The IL-2 may be the native polypeptide sequence, or can be a variant of the native IL-2 polypeptide as described herein below, so long as the variant IL-2 polypeptide retains the IL-2 biological activity of interest as defined herein. Preferably the IL-2 polypeptide or variant thereof is derived from a human source, and includes human IL-2 that is , recombinantly produced, such as recombinant human IL-2 polypeptides produced by microbial hosts, and variants thereof that retain the IL-2 biological activity of interest. Any pharmaceutical composition comprising IL-2 as a therapeutically active component can be used to practice the present invention.
Anticancer Agents:
Compositions of the present invention may be administered in conjunction with other anticancer agents. In particular, compositions will either be formulated together as a combination therapeutic or administered separately. Anticancer agents for use with the invention include, but are not limited to, one or more of the following set forth below: A. Kinase Inhibitors
Kinase inhibitors for use as anticancer agents in conjunction with the compositions of the present invention include inhibitors of Epidermal Growth Factor Receptor (EGFR) kinases such as small molecule quinazolines, for example gefitinib (US 5457105, US 5616582, and US 5770599), ZD-6474 (WO 01/32651), erlotinib (Tarceva®, US 5,747,498 and WO 96/30347), and lapatinib (US 6,727,256 and WO 02/02552); Vascular Endothelial Growth Factor Receptor (VEGFR) kinase inhibitors, including SU-11248 (WO 01/60814), SU 5416 (US 5,883,113 and WO 99/61422), SU 6668 (US 5,883,113 and WO 99/61422), CHIR-258 (US 6,605,617 and US 6,774,237), vatalanib or PTK-787 (US 6,258,812), VEGF-Trap (WO 02/57423), B43- Genistein (WO-09606116), fenretinide (retinoic acid p-hydroxyphenylamine) (US 4,323,581), IM-862 (WO 02/62826), bevacizumab or Avastin® (WO 94/10202), KRN-951, 3-[5-(methylsulfonylpiperadine methyl)-indolyl]-quinolone, AG-13736 and AG-13925, pyrrolo[2,l-fj[l,2,4]triazines, ZK-304709, Veglin®, VMDA-3601, EG-004, CEP-701 (US 5,621,100), Cand5 (WO 04/09769); Erb2 tyrosine kinase inhibitors such as pertuzumab (WO 01/00245), trastuzumab, and rituximab; AKT protein kinase inhibitors, such as RX-0201; Protein Kinase C (PKC) inhibitors, such as LY-317615 (WO 95/17182), and perifosine (US 2003171303); Phosphoinositide 3- Kinase (PI3K) Inhibitors including SF-1126 and PI- 103, PI-509, PI-516 and PI-540 (produced by PIramed); Raf/Map/MEK/Ras kinase inhibitors including sorafenib (BAY 43-9006), ARQ-350RP, LErafAON, BMS-354825 AMG-548, and others disclosed in WO 03/82272; Fibroblast Growth Factor Receptor (FGFR) kinase inhibitors; Cell Dependent Kinase (CDK) inhibitors, including CYC-202 or roscovitine (WO 97/20842 and WO 99/02162); Platelet-Derived Growth Factor Receptor (PGFR) kinase inhibitors such as CHIR-258, 3G3 mAb, AG-13736, SU- 11248 and SU6668; and Bcr-Abl kinase inhibitors and fusion proteins such as STI- 571 or Gleevec®.
B. Anti-Estrogens
Estrogen-targeting agents for use in anticancer therapy in conjunction with the compositions of the present invention include Selective Estrogen Receptor Modulators (SERMs) including tamoxifen, toremifene, raloxifene; aromatase inhibitors including Arimidex® or anastrozole; Estrogen Receptor Downregulators (ERDs) including Faslodex® or fulvestrant.
C. Anti-Androgens
Androgen-targeting agents for use in anticancer therapy in conjunction with the compositions of the present invention include flutamide, bicalutamide, finasteride, aminoglutethamide, ketoconazole, and corticosteroids.
D. Other Inhibitors
Other inhibitors for use as anticancer agents in conjunction with the compositions of the present invention include protein farnesyl transferase inhibitors" including tipifarnib or R-115777 (US 2003134846 and WO 97/21701), BMS-214662, AZD-3409, and FTI-277; topoisomerase inhibitors including merbarone and diflomotecan (BN-80915); mitotic kinesin spindle protein (KSP) inhibitors including SB-743921 and MKI-833; protease modulators such as bortezomib or Velcade® (US 5,780,454), XL-784; and cyclooxygenase 2 (COX-2) inhibitors including nonsteroidal antiinflammatory drugs I (NSAIDs). E. Cancer Chemotherapeutic Drugs
Particular cancer chemotherapeutic agents for use as anticancer agents in conjunction with the compositions of the present invention include anastrozole (Arimidex®), bicalutamide (Casodex®), bleomycin sulfate (Blenoxane®), busulfan (Myleran®), busulfan injection (Busulfex®), capecitabine (Xeloda®), N4- pentoxycarbonyl-S-deoxy-S-fluorocytidine, carboplatin (Paraplatin®), carmustine (BiCNU®), chlorambucil (Leukeran®), cisplatin (Platinol®), cladribine (Leustatin®), cyclophosphamide (Cytoxan® or Neosar®), cytarabine, cytosine arabinoside (Cytosar-U®), cytarabine liposome injection (DepoCyt®), dacarbazine (DTIC- Dome®), dactinomycin (Actinomycin D, Cosmegan), daunorubicin hydrochloride (Cerubidine®), daunorubicin citrate liposome injection (DaunoXome®), dexamethasone, docetaxel (Taxotere®, US 2004073044), doxorubicin hydrochloride (Adriamycin®, Rubex®), etoposide (Vepesid®), fludarabine phosphate (Fludara®), 5-fluorouracil (Adrucil®, Efudex®), flutamide (Eulexin®), tezacitibine, Gemcitabine (difluorodeoxycitidine), hydroxyurea (Hydrea®), Idarubicin (Idamycin®), ifosfamide (IFEX®), irinotecan (Camptosar®), L-asparaginase (ELSPAR®), leucovorin calcium, melphalan (Alkeran®), 6-mercaptopurine (Purinethol®), methotrexate (Folex®), mitoxantrone (Novantrone®), mylotarg, paclitaxel (Taxol®), phoenix (Yttrium90/MX-DTPA), pentostatin, polifeprosan 20 with carmustine implant (Gliadel®), tamoxifen citrate (Nolvadex®), teniposide (Vumon®), 6-thioguanine, thiotepa, tirapazamine (Tirazone®), topotecan hydrochloride for injection (Hycamptin®), vinblastine (Velban®), vincristine (Oncovin®), and vinorelbine (Navelbine®).
F. Alkylating Agents Alkylating agents for use in conjunction with the compositions of the present invention for anticancer therapeutics include VNP-40101M or cloretizine, oxaliplatin (US 4,169,846, WO 03/24978 and WO 03/04505), glufosfamide, mafosfamide, etopophos (US 5,041,424), prednimustine; treosulfan; busulfan; irofluven (acylfulvene); penclomedine; pyrazoloacridine (PD-115934); O6-benzylguanine; decitabine (5-aza-2-deoxycytidine); brostallicin; mitomycin C (MitoExtra); TLK-286 (Telcyta®); temozolomide; trabectedin (US 5,478,932); AP-5280 (Platinate formulation of Cisplatin); porfiromycin; and clearazide (meclorethamine).
G, Chelating Agents
Chelating agents for use in conjunction with the compositions of the present invention for anticancer therapeutics include tetrathiomolybdate (WO 01/60814); RP- 697; Chimeric T84.66 (cT84.66); gadofosveset (Vasovist®); deferoxamine; and bleomycin optionally in combination with electorporation (EPT). H. Biological Response Modifiers
Biological response modifiers, such as immune modulators, for use in conjunction with the compositions of the present invention for anticancer therapeutics include staurosprine and macrocyclic analogs thereof, including UCN-01, CEP-701 and midostaurin (see WO 02/30941, WO 97/07081, WO 89/07105, US 5,621,100, WO 93/07153, WO 01/04125, WO 02/30941, WO 93/08809, WO 94/06799, WO 00/27422, WO 96/13506 and WO 88/07045); squalamine (WO 01/79255); DA-9601 (WO 98/04541 and US 6,025,387); alemtuzumab; interferons (e.g. IFN-a, IFN-b etc.); interleukins, specifically IL-2 or aldesleukin as well as IL-I, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL- 10, IL- 11 , IL- 12, and active biological variants thereof having amino acid sequences greater than 70% of the native human sequence; altretamine (Ηexalen®); SU 101 or leflunomide (WO 04/06834 and US 6,331,555); imidazoquinolines such as resiquimod and imiquimod (US 4,689,338, 5,389,640, 5,268,376, 4,929,624, 5,266,575, 5,352,784, 5,494,916, 5,482,936, 5,346,905, 5,395,937, 5,238,944, and 5,525,612); and SMIPs, including benzazoles, anthraquinones, thiosemicarbazones, and tryptanthrins (WO 04/87153, WO 04/64759, and WO 04/60308). /. Cancer Vaccines:
Anticancer vaccines for use in conjunction with the compositions of the present invention include Avicine® (Tetrahedron Letters 26, 1974 2269-70); oregovomab (OvaRex®); Theratope® (STn-KLΗ); Melanoma Vaccines; GI-4000 series (GI-4014, GI-4015, and GI-4016), which are directed to five mutations in the Ras protein; GlioVax-1; MelaVax; Advexin® or INGN-201 (WO 95/12660); Sig/E7/LAMP-1, encoding ΗPV-16 E7; MAGE-3 Vaccine or M3TK (WO 94/05304); HER-2VAX; ACTIVE, which stimulates T-cells specific for tumors; GM-CSF cancer vaccine; and Listeria monocytogenes-based vaccines. J. Antisense Therapy:
Anticancer agents for use in conjunction with the compositions of the present invention also include antisense compositions, such as AEG-35156 (GEM-640); AP- 12009 and AP-11014 (TGF-beta2-specific antisense oligonucleotides); AVI-4126; AVI-4557; AVI-4472; oblimersen (Genasense®); JFS2; aprinocarsen (WO 97/29780); GTI-2040 (R2 ribonucleotide reductase mRNA antisense oligo) (WO 98/05769); GTI-2501 (WO 98/05769); liposome-encapsulated c-Raf antisense oligodeoxynucleotides (LErafAON) (WO 98/43095); and Sirna-027 (RNAi-based therapeutic targeting VEGFR-I mRNA). K. IL-2:
One preferred IL-2 compound is "aldesleukin" or "Proleukin®", manufactured by Chiron Corporation of Emeryville, California. The IL-2 in this formulation is a recombinantly produced, unglycosylated human IL-2 mutein which differs from the native human IL-2 amino acid sequence in having the initial alanine residue eliminated and the cysteine residue at position 125 replaced by a serine residue (referred to as des-alanyl-1, serine-125 human interleukin-2). This IL-2 mutein can be expressed in E. coli, and subsequently purified by diafiltration and cation exchange chromatography as described in U.S. Patent No. 4,931,543.
Aldesleukin has been compared to native (Jurkat) IL-2 in vitro. No significant differences have been seen and in vivo induction of cytolytic cells in mice and serum half-life following IV administration is equivalent for aldesleukin and native (Jurkat) IL-2; although there are beneficial attributes of the form of IL-2 encompassed by aldesleukin and therefore reference to "aldesleukin" encompasses only that composition and not all possible forms of the IL-2 protein.
In 1983 when Cetus' lymphocyte proliferation bioassay was developed, there was no official IL-2 reference preparation available. One Cetus unit was defined for this preparation as the amount of IL-2 in 1 mL that induced the IL-2 dependent murine T-cells to incorporate tritiated-thymidine at 50% of their maximum level after 24 hours of incubation. The IL-2 containing product was assigned a specific activity of 3 x 106 Cetus units per mg. In 1988, the National Institute of Biological Standards and Controls (NIBSC), which is a WHO laboratory for Biological Standards in England, established an IL-2 International Standard. Also in 1988, the assay procedure for Cetus' lymphocyte proliferation bioassay was changed. The Cetus' in- house standard was calibrated against the IL-2 International Standard in the new bioassay procedure. The following correction factor was established: International Units = Cetus Units x 6 Quantities of aldesleukin will be designated with mass units based on 1 mg of aldesleukin having a nominal specific activity of 18 x 106 International Units (18 MIU). The protocol incorporates International Units.
The pharmaceutical compositions useful in the methods of the invention may comprise biologically active variants of IL-2. Such variants should retain the desired biological activity of the native polypeptide such that the pharmaceutical composition comprising the variant polypeptide has the same therapeutic effect as the pharmaceutical composition comprising the native polypeptide when administered to a subject. That is, the variant polypeptide will serve as a therapeutically active component in the pharmaceutical composition in a manner similar to that observed for the native polypeptide. Methods are available in the art for determining whether a variant polypeptide retains the desired biological activity, and hence serves as a therapeutically active component in the pharmaceutical composition. Biological activity can be measured using assays specifically designed for measuring activity of the native polypeptide or protein, including assays described in the present inventions. Additionally, antibodies raised against a biologically active native polypeptide can be tested for their ability to bind to the variant polypeptide, where effective binding is indicative of a polypeptide having a conformation similar to that of the native polypeptide. For purposes of the present invention, the IL-2 biological activity of interest is the ability of IL-2 to activate and/or expand natural killer (NK) cells to mediate lymphokine activated killer (LAK) activity and antibody-dependent cellular cytotoxicity (ADCC). Thus, an IL-2 variant (for example, a mutein of human IL-2) for use in the methods of the present invention will activate and/or expand NK cells to mediate LAK activity and ADCC. Assays to determine IL-2 activation or expansion of NK cells and mediation of LAC or ADCC activity are well known in the art.
Suitable biologically active variants of native or naturally occurring IL-2 can be fragments, analogues, and derivatives of that polypeptide. By "fragment" is intended a polypeptide consisting of only a part of the intact polypeptide sequence and structure, and can be a C-terminal deletion or N-terminal deletion of the native polypeptide. By "analogue" is intended an analogue of either the native polypeptide or of a fragment of the native polypeptide, where the analogue comprises a native polypeptide sequence and structure having one or more amino acid substitutions, insertions, or deletions. "Muteins", such as those described herein, and peptides having one or more peptoids (peptide mimics) are also encompassed by the term analogue (see International Publication No. WO 91/04282). By "derivative" is intended any suitable modification of the native polypeptide of interest, of a fragment of the native polypeptide, or of their respective analogues, such as glycosylation, phosphorylation, polymer conjugation (such as with polyethylene glycol), or other addition of foreign moieties, so long as the desired biological activity of the native polypeptide is retained. Methods for making polypeptide fragments, analogues, and derivatives are generally available in the art.
For example, amino acid sequence variants of the polypeptide can be prepared by mutations in the cloned DNA sequence encoding the native polypeptide of interest. Methods for mutagenesis and nucleotide sequence alterations are well known in the art. See, for example, Walker and Gaastra, eds. (1983) Techniques in Molecular Biology (MacMillan Publishing Company, New York); Kunkel (1985) Proc. Natl. Acad. Sci. USA 82:488-492; Kunkel et al. (1987) Methods Enzymol. 154:367-382; Sambrook et al. ( 1989) Molecular Cloning: A Laboratory Manual (Cold Spring
Harbor Laboratory Press, Plainview, New York); U.S. Patent No. 4,873,192; and the references cited therein; herein incorporated by reference. Guidance as to appropriate amino acid substitutions that do not affect biological activity of the polypeptide of interest may be found in the model of Dayhoff et al. (1978) in Atlas of Protein Sequence and Structure (Natl. Biomed. Res. Found., Washington, D. C), herein incorporated by reference. Conservative substitutions, such as exchanging one amino acid with another having similar properties, may be preferred. Examples of conservative substitutions include, but are not limited to, GIy : Ala, VaI : He : Leu, Asp : GIu, Lys : Arg, Asn : GIn, and Phe : Trp : Tyr. In constructing variants of the IL-2 polypeptide of interest, modifications are made such that variants continue to possess the desired activity. Any mutations made in the DNA encoding the variant polypeptide must not place the sequence out of reading frame and preferably will not create complementary regions that could produce a secondary mRNA structure. Biologically active variants of IL-2 will generally have at least 70%, preferably at least 80%, more preferably about 90% to 95% or more, and most preferably about 98% or more amino acid sequence identity to the amino acid sequence of the reference polypeptide molecule, which serves as the basis for comparison. Thus, where the IL-2 reference molecule is human IL-2, a biologically active variant thereof will have at least 70%, preferably at least 80%, more preferably about 90% to 95% or more, and most preferably about 98% or more sequence identity to the amino acid sequence for human IL-2. A biologically active variant of a native polypeptide of interest may differ from the native polypeptide by as few as 1-15 amino acids, as few as 1-10, such as 6-10, as few as 5, as few as 4, 3, 2, or even 1 amino acid residue. By "sequence identity" is intended the same amino acid residues are found within the variant polypeptide and the polypeptide molecule that serves as a reference when a specified, contiguous segment of the amino acid sequence of the variants is aligned and compared to the amino acid sequence of the reference molecule. The percentage sequence identity between two amino acid sequences is calculated by determining the number of positions at which the identical amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the segment undergoing comparison to the reference molecule, and multiplying the result by 100 to yield the percentage of sequence identity.
Thus, the determination of percent identity between any two sequences can be accomplished using a mathematical algorithm. Preferably, naturally or non-naturally occurring variants of IL-2 have amino acid sequences that are at least 70%, preferably 80%, more preferably, 85%, 90%, 91%, 92 %, 93%, 94% or 95% identical to the amino acid sequence to the reference molecule, for example, the native human IL-2, or to a shorter portion of the reference IL-2 molecule. More preferably, the molecules are 96%, 97%, 98% or 99% identical. Percent sequence identity is determined using the Smith- Waterman homology search algorithm using an affined gap search with a gap open penalty of 12 and a gap extension penalty of 2, BLOSUM matrix of 62. The Smith-Waterman homology search algorithm is taught in Smith and Waterman, Adv. Appl. Math. (1981) 2:482-489. A variant may, for example, differ by as few as 1 to 10 amino acid residues, such as 6-10, as few as 5, as few as 4, 3, 2, or even 1 amino aid residue.
With respect to optimal alignment of two amino acid sequences, the contiguous segment of the variant amino acid sequence may have additional amino acid residues or deleted amino acid residues with respect to the reference amino acid sequence. The contiguous segment used for comparison to the reference amino acid sequence will include at least twenty (20) contiguous amino acid residues, and may be 30, 40, 50, or more amino acid residues. Corrections for sequence identity associated with conservative residue substitutions or gaps can be made (see Smith-Waterman homology search algorithm).
The precise chemical structure of a polypeptide having IL-2 activity depends on a number of factors. As ionizable amino and carboxyl groups are present in the molecule, a particular polypeptide may be obtained as an acidic or basic salt, or in neutral form. All such preparations that retain their biological activity when placed in suitable environmental conditions are included in the definition of polypeptides having IL-2 activity as used herein.
Further, the primary amino acid sequence of the polypeptide may be augmented by derivatization using sugar moieties (glycosylation) or by other supplementary molecules such as lipids, phosphate, acetyl groups and the like. It may also be augmented by conjugation with saccharides. Certain aspects of such augmentation are accomplished through post-translational processing systems of the producing host; other such modifications may be introduced in vitro. In any event, such modifications are included in the definition of an IL-2 polypeptide used herein so long as the IL-2 activity of the polypeptide is not destroyed. It is expected that such modifications may quantitatively or qualitatively affect the activity, either by enhancing or diminishing the activity of the polypeptide, in the various assays. Further, individual amino acid residues in the chain may be modified by oxidation, reduction, or other derivatization, and the polypeptide may be cleaved to obtain fragments that retain activity.
Such alterations that do not destroy activity do not remove the polypeptide sequence from the definition of IL-2 polypeptides of interest as used herein.
The art provides substantial guidance regarding the preparation and use of polypeptide variants. In preparing the IL-2 variants, one of skill in the art can readily determine which modifications to the native protein nucleotide or amino acid sequence will result in a variant that is suitable for use as a therapeutically active component of a pharmaceutical composition used in the methods of the present invention. The IL-2 or variants thereof for use in the methods of the present invention may be from any source, but preferably is recombinant IL-2. By "recombinant IL-2" is intended interleukin-2 that has comparable biological activity to native-sequence IL-2 and that has been prepared by recombinant DNA techniques as described, for example, by Taniguchi et al. (1983) Nature 302:305-310 and Devos (1983) Nucleic Acids Research 11 :4307-4323 or mutationally altered IL-2 as described by Wang et al. (1984) Science 224:1431-1433. In general, the gene coding for IL-2 is cloned and then expressed in transformed organisms, preferably a microorganism, and most preferably E. coli, as described herein. The host organism expresses the foreign gene to produce IL-2 under expression conditions. Synthetic recombinant IL-2 can also be made in eukaryotes, such as yeast or human cells. Processes for growing, harvesting, disrupting, or extracting the IL-2 from cells are substantially described in, for example, U.S. Patent Nos. 4,604,377; 4,738,927; 4,656,132; 4,569, 790; 4,748,234; 4,530,787; 4,572,798; 4,748,234; and 4,931,543. For examples of variant IL-2 proteins, see European Patent (EP) Publication
No. EP 136,489 (which discloses one or more of the following alterations in the amino acid sequence of naturally occurring IL-2: Asn26 to Gln26; Trpl21 to Phel21; Cys58 to Ser58 or Ala58, CyslO5 to SerlO5 or AlalO5; Cysl25 to Serl25 or Alal25; deletion of all residues following Arg 120; and the Met-1 forms thereof); and the recombinant IL-2 muteins described in European Patent Application No. 83306221.9, filed October 13, 1983 (published May 30, 1984 under Publication No. EP 109,748), which is the equivalent to Belgian Patent No. 893,016, and commonly owned U.S. Patent No. 4,518,584 (which disclose recombinant human IL-2 mutein wherein the cysteine at position 125, numbered in accordance with native human IL-2, is deleted or replaced by a neutral amino acid; alanyl-serl25-IL-2; and des alanayl-serl25-IL-2). See also U.S. Patent No. 4,752,585 (which discloses the following variant IL-2 proteins: alalO4 serl25 IL- 2, alalO4 IL-2, alalO4 alal25 IL-2, vallO4 ser!25 IL-2, vallO4 IL-2, vallO4 alal25 IL-2, des-alal alalO4 serl25 IL-2, des-alal alalO4 IL-2, 10 des-alal alalO4 alal25 IL-2, des-alal vallO4 serl25 IL-2, des-alal vallO4 IL-2, des-alal vallO4 alal25 IL-2, des-alal des-pro2 alalO4 serl25 IL-2, des-alal des-pro2 alalO4 IL- 2, des-alal des-pro2 alalO4 aial25 IL-2, des- alal des-pro2 vallO4 serl25 IL-2, des-alal des pro2 vallO4 IL-2, des-alal des-pro2 vallO4 alal25 IL-2, des-alal des-pro2 des-thr3 alalO4 serl25 IL- 2, des-alal des-pro2 des-thr3 alalO4 IL-2, des-alal des-pro2 des-thr3 alalO4 15 alal25 IL-2, des-alal des-pro2 des-thr3 vallO4 serl25 IL-2, des-alal des-pro2 des-thr3 vallO4 IL-2, des-alal des-pro2 des-thr3 vallO4 alal25 IL-2, des-alal des-pro2 des-thr3 des ser4 alalO4 serl25 IL-2, des- alal des-pro2 des-thr3 des-ser4 alalO4 IL-2, des-alal des-pro2 des-thr3 des-ser4 alalO4 alal25 IL-2, des-alal des-pro2 des-thr3 des- ser4 vallO4 serl25 IL-2, des-alal des-pro2 des-thr3 des-ser4 vallO4 IL-2, des-alal des- pro2 des-thr3 des-ser4 vallO420 alal25 IL-2, des-alal des-pro2 des- thr3 des-ser4 des- ser5 alalO4 serl25 IL-2, des-alal des pro2 des-thr3 des-ser4 des-ser5 alalO4 IL-2, des- alal des-pro2 des-thr3 des-ser4 des-ser5 alalO4 alal25 IL-2, des-alal des-pro2 des-thr3 des-ser4 des-ser5 vallO4 serl25 IL-2, des alal des-pro2 des-thr3 des-ser4 des-ser5 val 104 IL-2, des-alal des-pro2 des-thr3 des-ser4 des-ser5 vallO4 ala!25 IL-2, des-alal des-pro2 des-thr3 des-ser4 des-ser5 des-ser6 alalO4 25 alal25 IL-2, des- alal des-pro2 des-thr3 des-ser4 des-ser5 des-ser6 alalO4 IL-2, des-alal des pro2 des-thr3 des-ser4 des-ser5 des-ser6 alalO4 serl25 IL-2, des-alal des-pro2 des-thr3 des ser4 des-ser5 des- ser6 vallO4 serl25 IL-2, des-alal des-pro2 des-thr3 des-ser4 des-ser5 des serό vallO4 IL-2, and des-alal des-pro2 des-thr3 des-ser4 des-ser5 des-ser6 vallO4 alal25 IL-2) and U.S. Patent No. 4,931 ,543 (which discloses the IL-2 mutein des-alanyl- 1 , serine 30 125 human IL-2 used in the examples herein, as well as the other IL-2 muteins). Also see European Patent Publication No. EP 200,280 (published December 10, 1986), which discloses recombinant IL-2 muteins wherein the methionine at position 104 has been replaced by a conservative amino acid. Examples include the following muteins: ser4 des-ser5 alalO4 IL-2; des- alal des-pro2 des-thr3 des-ser4 des-ser5 alalO4 alal25 IL-2; des-alal des-pro2 des-thr3 des-ser4 des-ser5 glulO4 serl25 IL-2; des-alal des-pro2 des-thr3 des ser4 des-ser5 glulO4 IL-2; des-alal des-pro2 des-thr3 des-ser4 des-ser5 glul04 alal25 IL-2; des-alal des-pro2 des-thr3 des-ser4 des-ser5 des-ser6 alalO4 alal25 IL-2; des-alal des-pro2 des-thr3 des-ser4 des-ser5 des-ser6 alalO4 IL-2; des-alal des-pro2 des-thr3 des-ser4 des- ser5 5 des-ser6 alalO4 serl25 IL-2; des-alal des-pro2 des-thr3 des-ser4 des-ser5 des-ser6 glulO4 serl25 IL-2; des-alal des-pro2 des-thr3 des-ser4 des-ser5 des-ser6 glulO4 IL-2; and des-alal des-pro2 des-thr3 des-ser4 des-ser5 des-ser6 glulO4 alal25 IL-2. See also European Patent Publication No. EP 118,617 and U.S. Patent No. 5,700,913, which disclose unglycosylated human IL-2 variants bearing alanine instead of native IL- 2's methionine as the N-terminal amino acid; an unglycosylated human IL-2 with the initial methionine deleted such that proline is the N- terminal amino acid; and an unglycosylated human IL-2 with an alanine inserted between the N- terminal methionine and proline amino acids. Other IL-2 muteins include the those disclosed in WO 99/60128 (substitutions of the aspartate at position 20 with histidine or isoleucine, the asparagine at position 88 with arginine, glycine, or isoleucine, or the glutamine at positionl26 with leucine or gulatamic acid), which reportedly have selective activity for high affinity IL-2 receptors expressed by cells expressing T cell receptors in preference to NK cells and reduced IL-2 toxicity; the muteins disclosed in U.S Patent No. 5,229, 109
(substitutions of arginine at position 38 with alanine, or substitutions of phenylalanine at position 42 with lysine), which exhibit reduced binding to the high affinity IL-2 receptor when compared to native IL-2 while maintaining the ability to stimulate LAK cells; the muteins disclosed in International Publication No. WO 00/58456 (altering or deleting a naturally occurring (x)D(y) sequence in native IL-2 where D is aspartic acid, (x) is leucine, isoleucine, glycine, or valine, and (y) is valine, leucine or serine), which are claimed to reduce vascular leak syndrome; the IL-2 pl-30 peptide disclosed in International Publication No. WO 00/04048 (corresponding to the first 30 amino acids of IL-2, which contains the entire a-helix A of IL-2 and interacts with the b chain of the IL-2 receptor), which reportedly stimulates NK cells and induction of LAK cells; and a mutant form of the IL-2 pl-30 peptide also disclosed in WO 00/04048 (substitution of aspartic acid at position 20 with lysine), which reportedly is unable to induce vascular bleeds but remains capable of generating LAK cells. Additionally, IL-2 can be modified with polyethylene glycol to provide enhanced solubility and an altered pharmokinetic profile (see U.S. Patent No. 4,766,106). The term IL-2 as used herein is also intended to include IL-2 fusions or conjugates comprising IL-2 fused to a second protein or covalently conjugated to polyproline or a water soluble polymer to reduce dosing frequencies or to improve IL- 2 tolerability. For example, the IL-2 (or a variant thereof as defined herein) can be fused to human albumin or an albumin fragment using methods known in the art (see WO 01/79258). Alternatively, the IL-2 can be covalently conjugated to polyproline or polyethylene glycol homopolymers and polyoxyethylated polyols, wherein the homopolymer is unsubstituted or substituted at one end with an alkyl group and the poplyol is unsubstituted, using methods known in the art (see, for example, U.S.
Patent Nos.4,766, 106, 5,206,344, and 4,894,226). Any pharmaceutical composition comprising IL-2 as the therapeutically active component can be used in the methods of the invention. Such pharmaceutical compositions are known in the art and include, but are not limited to, those disclosed in U.S. Patent Nos. 4,745,180; 4,766,106; 4,816,440; 4,894,226; 4,931 ,544; and 5,078,997. Thus liquid, lyophilized, or spray- dried compositions comprising IL-2 or variants thereof that are known in the art may be prepared as an aqueous or nonaqueous solution or suspension for subsequent administration to a subject in accordance with the methods of the invention. Each of these compositions will comprise IL-2 or variants thereof as a therapeutically or prophylactically active component. By "therapeutically or prophylactically active component" is intended the IL-2 or variants thereof is specifically incorporated into the composition to bring about a desired therapeutic or prophylactic response with regard to treatment, prevention, or diagnosis of a disease or condition within a subject when the pharmaceutical composition is administered to that subject. Preferably the pharmaceutical compositions comprise appropriate stabilizing agents, bulking agents, or both to minimize problems associated with loss of protein stability and biological activity during preparation and storage. In preferred embodiments of the invention, the IL-2 containing pharmaceutical compositions useful in the methods of the invention are compositions comprising stabilized monomelic IL-2 or variants thereof, compositions comprising multimeric IL-2 or variants thereof, and compositions comprising stabilized lyophilized or spray-dried IL-2 or variants thereof.
Pharmaceutical compositions comprising stabilized monomeric IL-2 or variants thereof are disclosed in International Publication No. WO 01/24814, entitled "Stabilized Liquid Polypeptide-Containing Pharmaceutical Compositions." By
"monomeric" IL-2 is intended the protein molecules are present substantially in their monomer form, not in an aggregated form, in the pharmaceutical compositions described herein. Hence covalent or hydrophobic oligomers or aggregates of IL-2 are not present. Briefly, the IL-2 in these liquid compositions is formulated with an amount of an amino acid base sufficient to decrease aggregate formation of IL-2 during storage. The amino acid base is an amino acid or a combination of amino acids, where any given amino acid is present either in its free base form or in its salt form. Preferred amino acids are selected from the group consisting of arginine, lysine, aspartic acid, and glutamic acid. These compositions further comprise a buffering agent to maintain phi of the liquid compositions within an acceptable range for stability of IL-2, where the buffering agent is an acid substantially free of its salt form, an acid in its salt form, or a mixture of an acid and its salt fond. Preferably the acid is selected from the group consisting of succinic acid, citric acid, phosphoric acid, and glutamic acid. Such compositions are referred to herein as stabilized monomeric IL-2 pharmaceutical compositions. The amino acid base in these compositions serves to stabilize the IL-2 against aggregate formation during storage of the liquid pharmaceutical composition, while use of an acid substantially free of its salt form, an acid in its salt form, or a mixture of an acid and its salt form as the buffering agent results in a liquid composition having an osmolality that is nearly isotonic. The liquid pharmaceutical composition may additionally incorporate other stabilizing agents, more particularly methionine, a nonionic surfactant such as polysorbate 80, and EDTA, to further increase stability of the polypeptide. Such liquid pharmaceutical compositions are said to be stabilized, as addition of amino acid base in combination with an acid substantially free of its salt fond, an acid in its salt fonn, or a mixture of an acid and its salt form, results in the compositions having increased storage stability relative to liquid pharmaceutical compositions formulated in the absence of the combination of these two components. These liquid pharmaceutical compositions comprising stabilized monomelic IL-2 may either be used in an aqueous liquid form, or stored for later use in a frozen state, or in a dried form for later reconstitution into a liquid fond or other form suitable for administration to a subject in accordance with the methods of present invention. By "dried form" is intended the liquid pharmaceutical composition or fonnulation is dried either by freeze drying (i.e., lyophilization; see, for example, Williams and Polli (1984) J. Parenteral Sci. Technol. 38:48 59), spray drying (see Masters (1991) in Spray- Drying Handbook (Sth ed; Longman Scientific and Technical, Essez, U.K.), pp. 491-676; Broadhead et al. (1992) Drug Devel. Ind. Pharm. 18:1169-1206; and Mumenthaler et al. (1994) Pharm. Res. 11 :12- 20, or air drying (Carpenter and Crowe (1988) Cryobiology 25:459-470; and Roser (1991) Biopharm. 4:47-53).
Other examples of IL-2 formulations that comprise IL-2 in its nonaggregated monomeric state include those described in Whittington and Faulds (1993) Drugs 46(3):446- 514. These formulations include the recombinant IL-2 product in which the recombinant IL- 2 mutein Teceleukin (unglycosylated human IL-2 with a methionine residue added at the amino-terminal) is formulated with 0.25% human serum albumin in a lyophilized powder that is reconstituted in isotonic saline, and the recombinant IL-2 mutein Bioleukin (human IL-2 with a methionine residue added at the amino-terminal, and a substitution of the cysteine residue at position 125 of the human IL-2 sequence with alanine) fonnulated such that 0.1 to 1.0 mg/ml IL-2 mutein is combined with acid, wherein the fonnulation has a pH of 3.0 to 4.0, 10 advantageously no buffer, and a conductivity of less than 1000 mmhos/cm (advantageously less than 500 mmhos/cm). See EP 373,679; Xhang et al. (1996) Pharmaceut. Res. 13(4):643- 644, and Prestrelski et al. (1995) Pharmaceut. Res. 12(9): 1250-1258. Examples of pharmaceutical compositions comprising multimeric IL-2 are disclosed in commonly owned U.S. Patent No.4,604,377. By "multimeric" is intended the protein molecules are present in the pharmaceutical composition in a microaggregated form having an average molecular association of 10-50 molecules. These multimers are present as loosely bound, physically associated IL-2 molecules. A lyophilized form of these compositions is available commercially under the tradename Proleukin® IL-2 (Chiron Corporation, Emeryville, California). The lyophilized formulations disclosed in this reference comprise selectively oxidized, microbially produced recombinant IL-2 in which the recombinant IL-2 is admixed with a water soluble carrier such as mannitol that provides bulk, and a sufficient amount of sodium dodecyl sulfate to ensure the solubility of the recombinant IL-2 in water.
These compositions are suitable for reconstitution in aqueous injections for parenteral administration and are stable and well tolerated in human patients. When reconstituted, the IL-2 retains its multimeric state. Such lyophilized or liquid compositions comprising multimeric IL-2 are encompassed by the methods of the present invention. Such compositions are referred to herein as multimeric IL-2 pharmaceutical compositions.
The methods of the present invention may also use stabilized lyophilized or spray-dried pharmaceutical compositions comprising IL-2, which may be reconstituted into a liquid or other suitable form for administration in accordance with methods of the invention. Such pharmaceutical compositions are disclosed in International Publication No. W O 01/49274 entitled "Methods for Pulmonary Del vefy of InterleuklTz-2." These compositions may further comprise at least one bulking agent, at least one agent in an amount sufficient to stabilize the protein during the drying process, or both. By "stabilized" is intended the IL-2 protein or variants thereof retains its monomelic or multimeric form as well as its other key properties of quality, purity, and potency following lyophilization or spray-drying to obtain the solid or dry powder form of the composition. In these compositions, preferred carrier materials for use as a bulking agent include glycine, mannitol, alanine, valine, or any combination thereof, most preferably glycine. The bulking agent is present in the formulation in the range of 0% to about 10% (w/v), depending upon the agent used. Preferred carrier materials for use as a stabilizing agent include any sugar or sugar alcohol or any amino acid. Preferred sugars include sucrose, trehalose, raffinose, stachyose, sorbitol, glucose, lactose, dextrose or any combination thereof, preferably sucrose. When the stabilizing agent is a sugar, it is present in the range of about 0% to about 9.0% (w/v), preferably about 0.5% to about 5.0%, more preferably about 1.0% to about 3.0%, most preferably about 1.0%. When the stabilizing agent is an amino acid, it is present in the range of about 0% to about 1.0% (w/v), preferably about 0.3% to about 0.7%, most preferably about 0.5%. These stabilized lyophilized or spray- dried compositions may optionally comprise methionine, ethylenediaminetetracetic acid (EDTA) or one of its salts such as disodium EDTA or other chelating agent, which protect the IL-2 or variants thereof against methionine oxidation. Use of these agents in this manner is described in U.S. Application Serial No. 09/677,643, herein incorporated by reference. The stabilized lyophilized or spray- dried compositions may be formulated using a buffering agent, which maintains the pH of the pharmaceutical composition within an acceptable range, preferably between about pH 4.0 to about pH 8.5, when in a liquid phase, such as during the formulation process or following reconstitution of the dried form of the composition. Buffers are chosen such that they are compatible with the drying process and do not affect the quality, purity, potency, and stability of the protein during processing and upon storage.
The previously described stabilized monomelic, multimeric, and stabilized lyophilized or spray-dried IL-2 pharmaceutical compositions represent suitable compositions for use in the methods of the invention. However, any pharmaceutical composition comprising an IL-2 compound as a therapeutically active component is encompassed by the methods of the invention.
Also provided herein is a kit or package containing at least one combination composition of the invention, accompanied by instructions for use. For example, in instances in which each of the drugs themselves are administered as individual or separate dosage forms, the kit comprises each of the drugs, along with instructions for use. The drug components may be packaged in any manner suitable for administration, so long as the packaging, when considered along with the instructions for administration, clearly indicates the manner in which each of the drug components is to be administered. Alternatively, each of the drug components of the combination may be combined into a single administrable dosage form such as a single composition.
For example, for an illustrative kit comprising rIL-2 and an antiangiogenic agent, the kit may be organized by any appropriate time period, such as by day. As an example, for Day 1, a representative kit may comprise unit dosages of each of rIL-2 and the antiangiogenic agent. If each of the drugs is to be administered twice daily, then the kit may contain, corresponding to Day 1, two rows of unit dosage forms of each of rIL-2 and the antiangiogenic agent, with instructions for the timing of administration. Alternatively, if one or more of the drugs differs in the timing or quantity of drug to be administered in comparison to the other drug members of the combination, then such would be reflected in the packaging and instructions. For example, if the rIL-2 is to be administered twice daily, and the antiangiogenic agent is to be taken only once daily, exemplary Day 1 packaging might correspond to unit dosage forms of rIL-2 as "Day 1, Dose 1", along with dosage forms for the antiangiogenic agent corresponding to "Day 1, Dose 2".
Various embodiments according to the above may be readily envisioned, and would of course depend upon the particular combination of drugs employed for treatment, their corresponding dosage forms, recommended dosages, intended patient population, and the like. The packaging may be in any form commonly employed for the packaging of pharmaceuticals, and may utilize any of a number of features such as different colors, wrapping, tamper-resistant packaging, blister paks, dessicants, and the like.
EXAMPLES
Below are examples of specific embodiments for carrying out the present invention. The examples are offered for illustrative purposes only, and are not intended to limit the scope of the present invention in any way.
Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperatures, etc.), but some experimental error and deviation should, of course, be allowed for.
Example 1
Compositions for Co-administration with rIL-2 A. COMPOUNDS
Table 1: Quinazoline Examples
Figure imgf000059_0001
6,7-bis(2-methoxyethoxy)-N-(3-
Figure imgf000060_0001
ethynylphenyl)quinazoIin-4-amine, US 5,747,498 erlotinib WO 96/30347
Figure imgf000060_0002
5 Table 3: Additional Compounds
Compound Structure Name Patent
Figure imgf000060_0003
N-(4-chlorophenyl)-4-((pyridin-4-
10 yl)methyl)phthalazin-1-amine US 6,258,812
Figure imgf000061_0001
Figure imgf000062_0001
Table 4: Macrocyclic Compound
Figure imgf000062_0002
Table 5: Isoindolinone Compounds
Compound Structure Name Patent y. CH3
Figure imgf000062_0003
[3-(1 ,3-dioxo-1 ,3-dihydro-2H-isoindol-2-yl)- 2,6-dioxopiperidin-1-yl]methyl 2-amino-3-
19 methylbutanoate hydrochloride WO 97/37988
Figure imgf000062_0004
[3-(1 ,3-dioxo-1 ,3-dihydro-2H-isoindol-2-yl)- 2,6-dioxopiperidin-1-yl]methyl ({[(1 ,1-
20 dimethylethyl)oxy]carbonyl}amino)acetate WO 97/37988
Figure imgf000062_0005
[3-(1 ,3-dioxo-1 ,3-dihydro-2H-isoindol-2-yl)- 2,6-dioxopiperidin-1 -yl]methyl
21 aminoacetate hydrochloride WO 97/37988
Figure imgf000062_0006
[3-(1 ,3-dioxo-1 ,3-dihydro-2H-isoindol-2-yl)- 2,6-dioxopiperidin-1-yl]methyl
22 methylamino)acetate hydrochloride WO 97/37988 4-({[3-(1 ,3-dioxo-1 ,3-dihydro-2H-isoindoI-2- yl)-2,6-dioxopiperidin-1-yl]methyl}oxy)-4-
23 oxobutanoic acid WO 97/37988
3-methyl-3-(4,5,6,7-tetrafluoro-1 -oxo-1 ,3- dihydro-2H-isoindol-2-yl)piperidine-2,6-
24 dione WO 98/54170
2-(3-fluoro-2,6-dioxopiperidin-3-yl)-1 H-
25 isoindole-1 ,3(2H)-dione US 5,874,448
Figure imgf000063_0001
3-fluoro-3-(1-oxo-1 ,3-dihydro-2H-isoindol-
26 2-yl)piperidine-2,6-dione
B. Syntheses
QUINAZOLINES Scheme 1:
Figure imgf000063_0002
Figure imgf000063_0003
Scheme 1 describes a modular method of synthesizing a myriad of substituted quinazoline compounds. Reference to AG indicates an activating group such as, for example a halide, triflate, or ketone, wherein as many as four AG groups may be present. Since the 4-chloro position, shown in the third step, is more active than positions 5-8 on the benzyl ring, displacement with NHRR' may proceed without much concomitant displacement of AG in any of positions 5-8. Subsequent displacement of the AG group for R" in the final step may proceed in the presence of a weak to moderate base. Alternatively, the AG group(s) may be modified to yield the desired product, for example a NO2 group may be reduced with Fe and AcOH in EtOH and H2O to yield an amino substituent, which may be further substituted, for example by reductive amination with paraformaldehyde.
As will be apparent by one skilled in the art, a plethora of functionalized 2- aminobenzamide starting materials are either commercially available, or easily synthesized by known procedures. Subsequently, the AG group(s) in the starting material may be a substituent desired in the final product (as in R"), such as, for example an alkoxy group(s) or a substituted alkyl group(s). Further, a number functionalized 2-nitrobenzamide starting materials are available, which are easily converted to the 2-aminobenzamide starting material in the presence of a reducing agent, such as H2/Pd/C in EtOH.
Alternative methods of making the quinazolines of the invention are described in WO 04/24703, WO 01/32651, US 5,457,105, US 5,616,582, US 5,770,599, WO 02/16351, US 6,727,256, WO 02/02552, US 5,747,498, and WO 96/30347. INDOLINONES Scheme 2a:
Figure imgf000064_0001
Scheme 2a is performed as a one-pot procedure, with reagents in step a being NH4OH, CuCl in H2O. Followed by addition of aq. HCl in step b. R2-R5 are as defined herein. Scheme 2b:
Figure imgf000065_0001
In Scheme 2b, the reagents are stirred in EtOH in the presence of piperidine
(a) to afford the final product. As will be apparent to a skilled artisan, the reaction may be heated to enhance the yield, depending on reactivity of the particular starting materials.
Scheme 2c:
Figure imgf000065_0002
In Scheme 2c, the reagents are refluxed in EtOH in the presence of NaOBu-t (a) to afford the final product. R9 as shown in scheme 2, is H, -OH, -CN, alkyl, aryl, heterocyclyl, alkoxy, or -NR3Rb as defined herein. It is contemplated that the above structure may replace Formula II to allow substitution at Rς>, whereby all other substituents are as defined herein.
Scheme 2d (Synthesis of Compound 6):
Figure imgf000065_0003
74% PHTHALAZINES
Preparation of substituted phthalazines as in Compound 10 are described as follows, as drawn from in US 6,258,812, which also includes other reaction schemes that may be helpful in synthesizing the compounds of the present invention. Scheme 3a: l-(4-Chloroanilino)-4-(4-pyridylmethyl)phthalazine dihydrochloride
A mixture of 15.22 g (59.52 mmol) l-chloro-4-(4- pyridylmethyl)phthalazine (for preparation see German Auslegeschriftno. 1 061 788 published JuI. 23, 1959]), 7.73 g (60.59 mmol) 4-chloroaniline and 200 ml 1-butanol is heated for 2 h under reflux. The crystallizate which is obtained when the mixture slowly cools to 5° C. is then filtered off and washed with 1-butanol and ether. The filter residue is dissolved in about 200 ml hot methanol, the solution is treated with 0.75 g activated carbon and filtered Via a Hyfio Super CeI, and the pH of the filtrate is adjusted to about 2.5 with 7 ml 3N methanolic HCl. The filtrate is evaporated to about half the original volume and ether added until slight turbidity occurs; cooling then leads to the precipitation of crystals. The crystallizate is filtered off, washed with a mixture of methanol/ether (1:2) as well as ether, dried for 8 h at 110° C. under HV, and equilibrated for 72 h at 20° C. and in room atmosphere. In this way, the title compound is obtained with a water content of 8.6%; m.p. >270° C. ; ' H NMR (DMSO-d 6 ) 11.05-12.20 (br), 9.18-9.23 (m, IH), 8. 88 (d, 2H), 8.35-8.40 (m, IH), 8.18-8.29 (m, 2H), 8.02 (d, 2H), 7.73 (d, 2H), 7.61 (d, 2H), 5.02 (s, 2H); ESI-MS: (M+H) + =347. Scheme 3b: l-(4-Chloroanilino)-4-(4-pyridylmethyl)phthalazine hydrochloride
A mixture of 0.972 g (3.8 mmol) l-chloro-4-(4- pyridylmethyl)phthalazine, 0.656 g (4 mmol) 4-chloroaniline hydrochloride (Research Organics, Inc., Cleveland, Ohio, USA) and 20 ml ethanol is heated for 2 h under reflux. The reaction mixture is cooled in an ice bath, filtered, and the crystallizate washed with a little ethanol and ether. After drying under HV for 8 h at 110° C. and for 10 h at 150° C, the title compound is obtained as a result of thermal removal of HCl; m.p. >270° C; ' H NMR (DMSO-d 6 ) 9.80-11.40 (br), 8.89-8.94 (m, IH), 8.67 (d, 2H), 8.25-8.30 (m, IH), 8.06-8.17 (m, 2H), 7.87 (d, 2H), 7. 69 (d, 2H), 7.49 (d, 2H), 4.81 (s, 2H); ESI-MS: (M+H) +=347. Scheme 3c:
1 -(4-Chloroanilino)-4-(4-pyridylmethyl)phthalazine hydrochloride A mixture of 1.28 g (5 mmol) l-chloro-4-(4-pyridylmethyl)phthalazine, 0.67 g (5.25 mmol) 4-chloroaniline and 15 ml 1-butanol is heated for 0.5 h at 100 C while stirring in a nitrogen atmosphere. The mixture is then cooled to RT, filtered, and the filtrate washed with 1-butanol and ether. For purification, the crystallizate is dissolved in 40 ml of hot methanol, the solution treated with activated carbon, filtered via Hyflo
Super CeI, and the filtrate evaporated to about half its original volume, resulting in the formation of a crystalline precipitate. Aftercooling to 0° C, filtration, washing of the filter residue with ether, and drying under HV for 8 h at 130° C, the title compound is obtained; m.p. >270° C; ' H NMR (DMSO-d 6 ) 9.80-11.40 (br), 8.89-8.94 (m, IH), 8.67 (d, 2H), 8.25-8.30 (m, IH), 8.06-8.17 (m, 2H), 7.87 (d, 2H), 7.69 (d, 2H), 7. 49 (d, 2H), 4.81 (s, 2H); ESI-MS: (M+H) + =347.
Example 2 Preparation of rIL-2
Preparation of Aldesleukin, rIL-2 (NSC3773364) (Proleukin®, Chiron) :
Messenger RNA from human Jurkat cell line is used to create double stranded cDNA, which is hybridized into pBR322 plasmids. A clone containing the IL-2 gene is identified using a 32P-labeled oligonucleotide probe corresponding to a short length IL-2 base sequence. The gene is inserted into a region of the pBR322 plasmid that has a convenient restriction site. The appropriate promoter and ribosome-binding site is inserted in front of the IL-2 gene, and the resulting expression clone encodes a modified recombinant rIL-2. In vitro mutagenesis of the cloned IL-2 is used to make a conservative substitution of serine for cysteine at position 125. The resultant molecule is indistinguishable from native IL-2 in its in vitro biological activity. A production strain of E. coli carrying the aldesleukin gene is grown in fermenters. The culture is harvested and the aldesleukin is extracted. A series of chromatographic steps are performed to purify the aldesleukin. The formulated product is adjusted to pH 7.2 - 7.8. The molecular weight of Proleukin® is approximately 15,600 daltons. Analysis by amino acid composition and N-terminal sequencing has confirmed that aldesleukin has the predicted protein sequence. Reconstitution and Dilution Procedure:
Proleukin® is as a lyophilized cake in 5 cc vials containing 1.3 mg of protein. Vials of Proleukin® for injection are reconstituted with 1.2 niL of Sterile Water for Injection, USP. The diluent is directed against the side of the vial to avoid excess foaming, swirling contents gently until completely dissolved, while avoiding shaking. When reconstituted each mL contains 1.1 mg (18 million IU) of Proleukin®. Reconstituted Proleukin® is suitable for intravenous injection directly or may be diluted as necessary in volumes of 50 mL to 500 mL with 5% Dextrose Injection, USP with 0.1% Albumin Human, USP. When diluting, the Albumin Human, USP is added to the 5% Dextrose Injection, USP prior to the addition of the reconstituted Proleukin®.
Example 3
Therapy with Single Agent rIL-2 Composition Versus Combination Therapy with rIL-2 Composition and Antiangiogenic Agents
Results of high dose rIL-2 have been summarized in Table 6.
Table 6: Trials of Single-Agent rIL-2 Bolus Infusion Schedules1
Dose range Number Median response duration
Authors Schedule rIL-2 (MIU) of CR PR /median survival (mos) patients
Atkins et al (7) Q 8 hrs Days 1-5 24/m2 71 4 8 16+/15 5
Fyfe et al (8) Q 8 hrs Days 1-5 0.6-0 72/kg 255 12 24 20 3/16 3
Yang et al (9) Q 8 hrs Days 1-5 0 72/kg 65 2 11 NS
Q 8 hrs Days 1-5 0 072/kg 60 4 5 NS
Rosenberg Q 8 hrs Days 1-5 0 72/kg 149 10 20 15 / 20 etal (lθ)
Rosenberg Q 8 hrs Days 1-5 0 72/kg 48 4 6 NS etal (l l)
Taneja et al (12) Q 8 hrs Days 1-5 0 6-0 72/kg 28 1 4 NS
Bukowski et 3 x per week 60/m2 41 1 5 5 / 10 8 al (13)
Abrams et Q 8 hrs Days 1-5 0 06/kg 16 0 0 NS i [Abrams, 1990 #30]
Total i 733 38 83 (5 2) (11 3) rIL-2 recombinant mterleukin-2, MIU million International Units, CR. complete response, PR partial response, q every, NS not stated
Complete response + Partial response = 16 5% (95% confidence interval, 13 8%-19 2%)
'Adapted in part from Bukowski (39)
A number of factors have given πse to the impetus for combination therapies with antiangiogenic compositions. The significant morbidity associated with high dose rIL-2 required careful patient selection, dramatically decreasing the number of potential patients who might benefit from therapy. Unfortunately, concomitant illness is most frequently found in those who have the highest incidence of RCC. The requirement for careful patient monitoring and occasional medical intensive care have made single agent high dose rIL-2 administration costly and restricted its use to large medical centers. The practical effect of which is to restrict availability to only a minority of patients.
Lower doses of rIL-2 have been critically evaluated, as seen in Table 7. Table 7: Phase II Trials of Single-Agent rIL-2: Subcutaneous Schedules1
Duration/median Median
Dose range" No of response/
Authors Schedule rIL-2 flVIIU) patients CR PR
Survival
Schomburg et al. 5 days/wk x 8 wks 4.8-14.4/m2/day 15 0 0 NS
Lissoni et al. Days 1 and 2 18.0/m2/day 14 0 4
9+/NS
5 days/wk x 6 wks 3.6/m2/day
Lissoni et al. Days 1 and 2 18.0/m2/day 50 1 13
13+/ 14+
5 days/wk x 6 wks 6.0/m2/day
Lopez Hanninen et al. 5 days/wk, wks 1-6 18.0/m2/day 16 0 1 NS
Buter et al. Days 1-5 18.0/m2/day 47 2 7
11.0/12.0
Days 8-12, etc. 9.0/m2/day
Casamassima et al. Days 1 and 2 18.0/m2/day 11 0 2 NS
Days 8-12, etc. 3.6/m2/day deLena et al. Days 1 and 2 18.0/m2/day 10 0 1 NS
Days 8-12, etc. 3.6/m2/day
Whitehead et al. Dose escalation every 3.0-30.0/m2/day 15 0 0 NS
2 wks
Marumo et al. i.v. (2 hr, Days 1 -28) 1.0/m2/day 12 3 0
31.0/NS then every day 1.0/m2/day
Total (%) 190 6(3.2) 28(14 t.7)
rIL-2: recombinant human interIeukin-2; MIU: million International Units; CR: complete response; PR: partial response; NS: not stated; i.v.: intravenous.
Complete response + partial response rate = 18.6% (95% confidence interval, 15.8-21.5%). "Total dose. bUnit type not specified.
'Adapted in part from Bukowski
The most commonly utilized subcutaneous regimen has been published by Buter et al. In Buter, rIL-2 was given once a day, 5 days per week for 6 weeks. During the first 5 day cycle, 18 x 106 IU (MIU) was given once daily; in the following cycles, the doses after the first 2 days were reduced to 9 MIU. Response rates and survival data may be similar to those published for high dose IV bolus administration of rIL-2. The Buter / Sleijfer regimen has recently been compared to high dose rIL-2 in a prospective randomized trial. Ninety six patients were randomized to high dose IV rIL-2 and 92 patients to SQ rIL-2( Buter / Sleijfer dose schedule). Previously, the response to high dose rIL-2 was 20% and to SQ rIL-2 was 10%. However overall survival was not different (p=0.34) from the high dose. As such, a catalyst for combination with an antiangiogenic compostions exists to increase efficacy and overall patient responsiveness to the lower dose regimen.
Regimens for combinations of aldesleukin with antiangiogenic agents are critically evaluated, as seen in Table 8.
Table 8: Treatment, Dose and Duration
Figure imgf000071_0001
One dose of antiangiogenic agent (equal to planed dose according to dose level) is given on Day -7.
Antiangiogenic agent is given again on day 1 and then every 2 weeks continuously in an 8-weeks treatment cycle.
Treatment with rIL-2 is continued for 6 consecutive weeks (days 1 - 42, Monday- Friday of each week) followed by a 2-week rest period resulting in an 8- week treatment cycle. Example 4 Combination Therapy with rIL-2 and Small Molecule Receptor Tyrosine Kinase
Inhibitors BAY 43-9006 and SU11248 A. Material and Methods Drugs
Recombinant human Interleukin-2 (Proleukin®, Aldesleukin/rIL-2); 18 MIU/ ml, Chiron Corporation, Emeryville, CA) was reconstituted with sterile water for injection and formulated in 5% dextrose prior to administration. Vincristine (vincristine sulfate) was from Mayne Pharma Ltd (Mulgrave, Australia). CHIR-258 is 4-amino-5-fluoro-3-[5-(4-methylpiperazin- 1 -yl)- 1 H-benzimidazol-2-yl]quinolin- 2(lH)-one (Chiron). BAY 43-9006 (Sorafanib/Nexavar®) (Riedl et al., Proc. Am. Assoc. Cancer Res. 2001 42(Abs 4956); Lowinger et al., Curr. Pharm. Des. 2002 8(25):2269-2278; WO 9932455) and SUl 1248 (Sunitinib/Sutent®) (Sun et al., J. Med. Chem. 2003 46(7):1116-1119; WO 0160814) were synthesized and purified in- house according to published procedures and patents. Stock solutions of BAY 43- 9006 or SUl 1248 (20 mM) were prepared in DMSO, and aliquots will be stored at - 20 0C prior to use. For in vitro assays, all drugs were diluted in optimal culture medium. For in vivo administration, BAY 43-9006 was formulated in 100% PEG 400 vehicle, whereas, SUl 1248 dosing solutions were prepared in 5 mM citrate buffer. All other chemicals used were of research grade.
Cell Lines
AU murine cell lines, CTLL-2 (IL-2 dependent T cell line), B16-F10 melanoma, CT26 colon, and renal carcinoma RENCA were obtained from American Tissue Culture Collection (Rockville, MD). CTLL-2 were grown in RPMIl 640 supplemented with 10% FBS (fetal bovine serum, Gibco Life Technologies, Gaithersburg, MD), 2 mM L-glutamine, 1 mM sodium pyruvate, 25 mM HEPES, 0.5 nM rIL-2, 2 mM /3-mercaptoethanol. RENCA cells were cultured in a media containing EMEM with 10% FBS, 2% IOOX vitamin, 1% 200 mM glutmine,l% 100 mM NaPy, 1% nonessential amino acids. For growing CT26 cells, media contained EMEM, 10% FBS, 2% vitamins, 1% 200 mM glutamine, 1% 100 mM sodium pyruvate, 1% nonessential amino acids. B16-F10 cells were grown in RPMI 1640 with 10% FBS, 1% nonessential amino acids, 1% 10OmM sodium pyruvate 2% vitamins; 2 mM l-glutamine; 2% sodium bicarbonate. Yac-1 cells (ATCC) were cultured in RPMI + 10% FBS and subcultured 1-2 days prior to assay to ensure log- phase growth. Cells were maintained as suspension or adherent cultures in a humidified atmosphere at 37 0C and 5% CO2. Cells were used in exponential growth phase (not exceeding 6-8 passages) with viability >98% (assessed using trypan blue staining) and determined free of mycoplasma.
In vivo Efficacy Studies
Female BALB/c or C57BL6 mice (4-6 week-old, 18-22 g) were obtained from Charles River (Wilmington, MA) and acclimated for 1 week in pathogen-free enclosures prior to the start of the study. Animals received sterile rodent chow and water ad libitum and were housed in sterile filter-top cages with 12 hour light/dark cycles. All experiments were under the guidelines of the Association for Assessment and Accreditation of Laboratory Animal Care International.
For tumor implantation, B16-F10 (2 x 106), CT26 (2 x 106) or RENCA (1 x 106) cells were harvested, washed three times, and resuspended in PBS. Mice were shaved on the flank, and implanted (0.2 ml) subcutaneously (s.c.) into the right flank of mice. For the B16-F10 tumor model, C57BL6 mice were used, whereas CT26 and RENCA tumors were implanted in BALB/c mice. Treatments were initiated when tumors were established to a mean size of 50-250 mm3 (day 0) as outlined in specific study designs. Mice were randomized into treatment cohorts (typically 10 mice/group). rIL-2 was administered daily s.c. (0.2-3 mg/kg/day) on days 0-6 or 7-13 or days 0-4, 7-11. BAY 43-9006 or SUl 1248 (1-100 mg/kg) were administered daily (for 5-12 days) as a solution via oral gavage, starting day 0 or day 7. All monotherapy and drug combinations at selected doses, outlined in individual studies were well tolerated.
Assessment of Tumor Inhibition and Responses
Tumor volumes and body weights were assessed 2-3 times weekly. Caliper measurements of tumors were converted into mean tumor volume (mm3) using the formula: 1A (length (mm) x [width (mm)]2). Tumor growth inhibition (TGI) was calculated as [l-(mean tumor volume of treated group/mean tumor volume of control group) x 100]. Responses were defined as either a complete response (CR, no measurable tumor), or partial response (PR5 50-99% tumor volume reduction) compared to tumor volume for each animal at treatment initiation. Tumor growth delay analysis was calculated as: [(number of days for a treated group to reach a mean tumor volume of 1000 mm3) - (number of days for the control group to reach a mean tumor volume of 1000 mm3)].
Synergistic effects were defined when the ratio of expected % tumor growth inhibition of combination therapy (%T/Cexp = %T/C treatment 1 x %T/C treatment 2) divided by observed % T/C (%T/Cobs) of the combination treatment was >1. Additive effects were defined when %T/Cexp/%T/Cobs =1, and antagonism when %T/Cexp/%T/Cobs <1 (Yokoyama et al., Cancer Res. 2000 60(80):2190-2196).
Pharmacokinetics
For evaluation of drug pharmacokinetics, mice were treated with a single s.c. dose of rIL-2 (6 mg/kg, 0.2 ml) or BAY 43-9006 (20 mg/kg, p.o., 0.2 ml) and blood was collected at various times after drug administration. Plasma levels of BAY 43- 9006 and rIL-2 were determined using HPLC or an ELISA bioassay.
Western blot analysis
After drug incubations under indicated conditions, cells were harvested, washed with ice-cold PBS and lysed with RIPA buffer (1% Nonidet P-40, 0.5% sodium deoxycholate, 0.1% sodium dodecylsulphate in IX phosphate buffered saline, pH 7.2) containing protease inhibitors (Roche Molecular Biochemicals, Indianapolis, IN) and phosphatase inhibitors (Sigma, St. Louis, MO). Protein content lysates were determined using the BCA assay (Bio-Rad, Hercules, CA). For western blot analysis, 60 μg of protein was electrophoresed and pERK detection was done with a mouse antibody to pERK (1 : 1000, Cell Signaling, Beverly, MA) and incubated at 4 0C overnight. Detection of pSTAT5 antibody (1 : 1000 Upstate) and pAKT (1 : 1000 Cell Signaling) was performed with the same amount of protein by probing with appropriate anti-phosphotyrosine antibodies for 2 hours at room temperature. The membranes were then incubated for 1 hour at room temperature with 1 : 5000 horseradish peroxidase-conjugated anti-rabbit IgG (Jackson Immunoresearch, West Grove, PA). To verify equal loading, blots were stripped and re-probed with anti- ERK (Cell Signaling), anti-STAT5 (BD Biosciences), and anti-AKT (Cell Signaling) antibodies to measure total ERK, STAT5 and AKT protein, respectively. Proteins were detected using enhanced chemiluminescence (ECL; Amersham Biosciences, Buckinghamshire, England) and visualized after exposure to Kodak film. Scanning densitometry was performed to quantify band intensities. The amount pERK, pSTAT5 or pAKT was normalized to total ERK, STAT5 or AKT protein levels, and compared with vehicle or untreated controls.
Cellular Immunophenotvping in BALB/c Nude Mice After Drug Treatment Blood samples were collected at various times after indicated treatments. Whole blood (100 μ\) was transferred to FACS/TruCount tubes (BD BioSciences) and kept on ice. Samples were treated with 0.5 μg mouse Fc block (anti-mouse CD16/CD32; BD BioSciences), and incubated on ice 20 minutes. Flourochrome- conjugated antibodies, as indicated below, were added to samples and incubated on ice for 20 minutes protected from light. Blood samples were vortexed while adding 2 ml of Ix FACS lysing solution (BD BioSciences), followed by incubation at room temperature for 10 minutes, then centrifuged at 1250 rpm. All samples were washed twice, suspended in PBS and 2% FBS, and stored at 4 0C prior to sample acquisition on BD FACScalibur, and subsequent analysis by CellQuest Pro software. Absolute numbers of cells were determined relative to TruCount bead reference. Populations of cells were identified and gated based on FSC and SSC characteristics, as well as markers for total lymphocytes (CD45, BD BioSciences); T cell lymphocyte populations were identified based on CD3 staining, and subpopulations identified by CD4 or CD8 staining (BD Biosciences). Individual cell populations were identified by appropriately gated events.
Histopathology and Immunohistochemistry Mouse tumors were fixed in 10% neutral buffered formalin and then transferred to 70% ethanol and subsequently processed for paraffin embedding using an Excelsior tissue processor (Thermo Electron Corporation, Pittsburgh, PA). Tissue sections (4 μm) were cut on a rotary microtome (RM2125, Leica Microsystems, Nussloch, Germany). Hemotoxylin and eosin (H&E) stained sections were prepared. Immunostains were then performed using a Discovery XT automated slide staining system (Ventana Medical Systems, Tucson, AZ). Murine T cells were detected with a rabbit anti-CD3 antibody (1 :60 dilution, Dako Norden A/S, Glostrup Denmark), and monocytes/macrophages were detected using F4/80 (Serotec). For cell proliferation, mouse tumors were stained for Ki-67 using a monoclonal rat anti-mouse antibody (1: 15 dilution, DAKO). Heat-induced epitope retrieval was performed using CC 1 (Ventana Medical Systems). Samples were then incubated with the appropriate secondary antibodies (goat anti-rabbit IgG biotinylated antibody, 1 : 100 dilution, Jackson ImmunoReseach Laboratories). A horseradish peroxidase-labelled streptavidin biotin system with 3-3'-diaminobenzidine chromogen (Ventana Medical Systems) was used for localizing the antibodies. Sections were counterstained with nuclear fast red to enhance visualization of tissue morphology. For B16-F10 tumors, the Ventana Bluemap kit - an alternative NBT/BCIP staining method was used due to observed melanin deposits in H&E tissues, which aided in visualization of T cells in tumors.
CTLL-2 T Cell Proliferaton Assay
CTLL-2 T cells were preincubated with or without BAY 43-9006 (3 μM) at 37 0C for 2 hours. The cells were then washed and plated in 96-well microtiter plates, 5,000 cells/well in culture media with serial dilutions of rhIL-2 (1 pM to 100 nM). At the end of the incubation period (72 hours at 37 0C), cell viability was determined by a tetrazolium dye assay using cell proliferation reagent WST-I (Roche Applied Science, Indianapolis, IN).
In vitro Cytotoxicity Assays
Cells were plated in 96-well microtiter plates (CTLL-2: 5000 cells/well; RENCA: 1500/well; B16-F10: 1000/well; MV4;11 : 5000/well) and treated with serial dilutions of BAY 43-9006/SU11248 or vincristine. CTLL-2 is an IL-2 dependent cell line, and hence for these cells, the cytoxicity assay was conducted in media containing 5 nM rIL-2. At the end of the incubation period (72 hours at 370C), cell viability was determined by either a tetrazolium dye (WST-I) assay or a chemiluminescent (BrdU) assay (Roche Applied Science, Indianapolis, IN). EC50 values were defined as the concentration needed for a 50% reduction in absorbance of treated vs. untreated control cells. Ex vivo Splenocvte Cytotoxicity Assays
Splenocytes were obtained from drug-treated BALB/c mice (n= 3-5/group) under aseptic conditions and homogenized in cold PBS. Following passage through a 70 μm nylon cell strainer, cells were briefly centrifuged at 4 0C. Red blood cells were lysed using RBC lysis buffer (Sigma, St. Louis, MO). Cells were washed and plated with 51Cr-labeled Yac-1 target cells at various E:T ratios (100:1, 50:1, 25:1, 12/5:1, 6.25:1, 3:1) in a 4 hour cytotoxicity assay. Data were obtained using a Wallac plate reader and expressed in counts per minute (cpm). Quantification is expressed as percent specific lysis, and was calculated as: % specific lysis = 100 x ((mean experimental - mean spontaneous release)/(mean maximal release - mean spontaneous release)). The spontaneous release was determined from wells containing labeled target cells and no effector cells, and maximal release was determined from wells containing labeled target cells in 1% Triton X-100.
Statistical analysis
Multiple comparisons were performed using one-way analysis of variance (ANOVA), and post-test to compare different treatment means was done using Student-Newman Keuls test (SigmaStat). Differences were considered statistically significant at p < 0.05.
B. In vitro Studies with rIL-2 and BAY 43-9006 on T cell and Tumor cell Signaling Pathways.
rIL-2 Activates JAK/STAT and MAPK Signaling in CTLL-2 Cells in vitro To elucidate the onset, time course and duration of IL-2/IL-2R signal transduction pathways in T cells, CTLL-2 cells (IxIO7 cells) were serum starved for 24 hours prior to treatment with various concentrations of rIL-2 (1 pM to 100 nM) for 2 hours and key signaling pathways: MAPK, STAT5, AKT were evaluated using western blot analyses. The serum-starved CTLL-2 cells were treated in vitro with a wide range of concentrations of rIL-2 (from 1 pM to 100 nM) in order to delineate the interactions of the two predominant IL-2R complexes: the high affinity (IL-2Rαβγ KD = 10"11M) and the intermediate affinity (IL-2βγ KD = 10"9M) receptor. In some experiments, cells were exposed to free anti-IL-2α antibody (>1000-fold excess; 10 nM) incubated for 1 hour prior to addition of rIL-2. To evaluate the time course of IL-2/IL-2R activation, serum starved CTLL-2 cells were activated with 10 nM rIL-2, and IL-2 signaling was examined at various times from 10 minutes up to 48 hours. IL-2R downstream phosphorylation of ERK1/2, STAT5 and AKT was evaluated in rIL-2-treated cells using Western blot analysis. The relative levels of pERK, pSTAT5 or pAKT were compared to the total protein levels for ERK, STAT5 or AKT, respectively.
Activation of pERKl/2 was observed following exposure of CTLL-2 cells with rIL-2. Phospho ERK was slightly activated at 1 pM at 2 hours however, maximal activation was seen at > 100 pM concentrations. The JAK/STAT5 pathway was maximally activated at 1 pM concentration; and increasing concentrations of rlL- 2 did not change levels of pSTAT5 (up to 100 nM). The basal pAKT pathway in CTLL-2 appeared to be activated in T cells under serum starved conditions. Furthermore pAKT levels remained largely unchanged at the rIL-2 concentrations tested (1 pM to 100 nM; using a pAKT antibody to phosphorylation site 483).
A blocking antibody to IL-2Rα was used to confirm that the IL-2 signal pathways are mediated specifically by the binding to the IL-2R. The pSTAT5 levels in CTLL-2 cells were analyzed by Western blot. CTLL cells were serum starved and treated with excess free anti-IL-2Rα antibody (10 nM, > 1000-fold) for 1 hour prior to treatment with rIL-2 (0.1 pM to 10 nM). In the absence of the blocking IL-2Rα antibody, pSTAT5 was activated at 1 pM following rIL-2 treatment, however, in the presence of IL-2Rα inhibition, pSTAT5 signaling was abrogated (> 95% inhibition), confirming the requirement of the IL-2Rα in STAT5 signaling. Interestingly, pSTAT5 levels were restored in CTLL-2 cells with increasing concentrations of rIL-2 (>10 pM), suggesting rIL-2 either competitively displaces the anti-IL-2Rα antibody and/or activates pSTAT5 signaling by binding to the low affinity IL-2Rβγ chain (KQ = 10-9 M).
rIL-2 Activation of pSTAT5 is Rapid and Sustained in CTLL-2 Cells To evaluate the time of onset and duration of IL-2R signaling responses in
CTLL-2 cells, serum starved cells were treated in vitro with rIL-2 (10 nM) and effects on phosphorylation of ERKl /2, STAT5 and AKT was evaluated using Western blot analyses. Phosphorylation of STAT5 was activated in minutes (< 10 minutes) after addition of rIL-2 to CTLL-2 cells and the duration of pSTAT5 response was maintained up to 48 hours. Activation of the MAPK pathway (pERK) pathway by rIL-2 appeared to be slightly delayed and was activated by 1 hour. The intensity of pERK was lower than the maximal response obtained by stimulation of serum starved CTLL-2 cells with PMA (50 ng/ml) + ionomycin (0.4 μg/ml) for 15 minutes.
Additionally, pERK levels were sustained up to 24 hours and returned to background levels by 48 hours. No discernable effects were observed on the pAKT, confirming that signaling via PI-3K/AKT pathway was continually active in CTLL-2 cells.
BAY 43-9006 Treatment Inhibits pERK and not pSTAT5 in CTLL-2 Cells
BAY-43-9006 is a potent inhibitor of Raf-1, which also inhibits both wild-type and mutant BRAF. In addition, BAY-43-9006 inhibits multiple kinases particularly VEGF2, 3; PDGFRβ; FLT3 and cKIT (Wilhelm, SM et al. Cancer Res 2004; and Chiron's kinase profiling data), and inhibits to some extent Lck and Fyn, two kinases that are involved in T cell functional responses. Given the inhibitory kinase profile of BAY 43-9006, and the potential impact on MAPK signaling/T cell signaling, it was thought that BAY 43-9006 could potentially interfere with IL-2/IL-2R signaling in T cells. Therefore, the potential interactions of BAY 43-9006 on IL-2 signaling were evaluated in CTLL-2, B16-F10, or RENCA cells in vitro. To examine the effects of BAY 43-9006 on CTLL-2, B16-F10 or RENCA model cells, serum starved cells were treated with various concentrations of BAY 43- 9006 (0.01- 20 μM). As an appropriate control, cells were stimulated with PMA (50 ng/ml) and ionomycin (0.4 μg/ml) for 15 minutes. To examine the effects of concomitant and sequential treatments of rIL-2 and BAY 43-9006 in CTLL-2 cells in vitro, serum starved CTLL-2 cells were treated with rIL-2 (10 nM) in the presence or absence of BAY 43-9006 (3 μM) for 2 hours. For sequential treatments, BAY 43- 9006 was treated for 2 hours. The cells were then washed and then treated with rIL-2, and vice versa. The inhibitory effects of BAY 43-9006 (3 μM) were also examined with or without stimulation with PMA (50 ng/ml) and ionomycin (0.4 μg/ml) for 15 minutes.
Since the concentration of BAY 43-9006 required to inhibit the MAPK pathway in different cell types is quite variable, the effects of various concentrations of BAY 43-9006 (ranging from 0 to 20 μM) were evaluated on serum starved CTLL- 2 cells for 2 hours with or without PMA + Ionomycin stimulation. At the end of the incubation period, treated CTLL-2 cells were then lysed and the protein lysates were subjected to Western blot analyses for determination of levels of pERK. BAY 43- 9006 substantially inhibited pERK at concentrations > 1 μM in murine CTLL-2 cells and the human Jurkat T cell line. The effects of BAY 43-9006 treatment on CTLL-2 cells did not alter pSTAT5 and pAKT levels in cells indicating that the JAK/STAT5 pathway and PI-3K/AKT in T cells were largely unaffected.
To investigate the therapeutic basis of combining rIL-2 and BAY 43-9006 in vitro, the effects of rIL-2 and BAY 43-9006 on T cells and the impact on the two IL- 2-mediated signaling pathways (MAPK and JAK/STAT5) in CTLL-2 cells were studied. The IL-2 mediated T cell signaling effects with concomitant or sequential regimens of the two drugs were investigated in CTLL-2 cells. Serum starved CTLL-2 cells were treated with BAY 43-9006 (3 μM) for 2 hours and then treated with either vehicle, rIL-2 (10 nM) or PMA+Ionomycin. Alternatively, rIL-2 (10 nM, 2 hours) treatment followed by BAY 43-9006 (3 μM, 2 hours) was also investigated. After drug exposure and/or PMA+ Ionomycin stimulation, Western blot analyses of pERK and pSTAT5 in cell lysates were performed (as described earlier). BAY 43-9006 (3 μM) treatment inhibited pERK levels, whereas, rIL-2 activated pERK levels in serum starved CTLL-2 cells (vs. baseline pERK levels), confirming the opposing effects of the two drugs on the MAPK pathway. AU treatments with BAY 43-9006 combinations (concomitant and sequential drug exposures) substantially inhibited pERK in CTLL-2 cells. No effects were observed on the STAT5 pathway or AKT pathway, with all combination treatments tested (as outlined in methods).
BAY 43-9006 at High Concentrations Inhibits pERK Levels in Tumor Cell
Lines
The effects of BAY 43-9006 treatment on murine tumor cell lines- B16-F10 melanoma, CT26 colon and the RCC RENCA model was determined. The murine cell lines were selected based on their responsiveness to in vivo rIL-2 therapy in immunocompetent models (T-/NK-/monocyte-/macrophage-competent mice), where the effects of combined rIL-2 and BAY 43-9006 therapy could be investigated. Serum starved B16-F10 melanoma and RENCA cells were exposed to a range of concentrations of BAY 43-9006 from 0 to 20 μM. Phospho-ERK levels in cell lysates following drug exposure were determined by Western blot analyses. BAY 43-9006 inhibited pERK levels in both cell lines tested (B16-F10 and RENCA) at very high concentrations of > 5μM, with almost complete abolition of pERK seen at 20 μM.
C. In vitro Effects of BAY 43-9006 on IL-2-Mediated Cell Proliferation
To examine if pERK inhibition by BAY 43-9006 affected IL-2-mediated proliferative responses in CTLL-2 cells, proliferation assays were conducted by pre- incubating CTLL-2 cells (5000 cells/well) at concentrations of BAY 43-9006 (3 μM, 2 hours) that inhibit pERK. After a 2 hour incubation of cells with BAY 43-9006 (3 μM), cells were plated in 96-well plates and exposed to various concentrations of rlL- 2 (0 - 100 nM) for 72 hours. Untreated cells were given sham treatments prior to incubation with rIL-2 (at the same concentrations). Proliferative responses of BAY 43-9006-treated and untreated CTLL-2 cells were assessed using the WST-I assay. Pre-incubation of cells with BAY 43-9006 (3 μM) inhibited pERK signaling but did not affect IL-2 induced proliferative responses in the CTLL-2 cell line.
D. Antiproliferative Activity of BAY 43-9006 or SUl 1248 on CTLL-2 and Tumor cells in Vitro
To assess the in vitro cytotoxic activity of BAY 43-9006 or SUl 1248 in each of these cell types (CTLL-2, B16-F10, RENCA, CT26), cells were plated in 96-well microtiter plates and treated with serial dilutions of BAY 43-9006 (from 0 to 50 μM) for 72 hours at 37 0C. Since the CTLL-2 cell line are dependent on IL-2 for proliferation, cytotoxicity against these cells were conducted in media containing 5 nM rIL-2. At the end of the 72 hours incubation period, cell viability was determined by either a tetrazolium dye (WST-I) assay or a chemiluminescent (BrdU) assay. As controls, MV4;11 (human FLT3 ITD AML cell line) were treated with BAY 43-9006 (0 to 50 μM) or B16-F10 cells were treated with vincristine (0 to 1 μM) to confirm cytotoxicity of agents and validity of assays. The inhibitory concentrations of the drugs were expressed as an EC5O value, which was defined as the concentration needed for a 50% reduction in proliferative response measured as absorbance of drug- treated cells vs. untreated/vehicle controls.
The relative EC50 of BAY 43-9006 or SUl 1248 are presented in Table 9. Table 9. Antiproliferative activity of BAY 43-9006 or SU11248 on various cell lines.
Figure imgf000082_0001
Cells were plated in 96-well microtiter plates (CTLL-2: 5000 cells/well; RENCA: 1500/well; B16-F10: 1000/well; CT26: 1000 cells/well; MV4;11 : 5000/well) and treated with serial dilutions of BAY 43-9006, SUl 1248 or Vincristine. CTLL-2 is an IL-2 dependent cell line and hence the cytoxicity assay was conducted in media containing 5 nM rIL-2. At the end of the incubation period (72 hours at 37 0C), cell viability was determined by either a tetrazolium dye (WST-I) assay. aEC50 = concentration needed for a 50% reduction in proliferative response measured as absorbance of drug-treated cells vs. untreated/vehicle controls. b MV4;11 is a human acute myelogenous leukemia (AML) cell line that expresses a FLT3 Internal Tandem Duplication (ITD). BAY 43-9006 (potent FLT3 kinase inhibitor; IC50 = 58 nM) demonstrates potent in vitro antiproliferative activity against MV4; 11 cells. d Cell viability was assessed using the Promega Cell-Titer Glo™ assay that measured ATP content of cells
In general, relatively high concentrations of BAY 43-9006 (or SUl 1248) were needed to inhibit proliferation of CTLL-2 cells as well as various cells lines (B 16- FlO, RENCA, CT26), compared to the antimitotic agent vincristine (defined by the relative EC50's; see Table 9). The cytotoxicity of BAY 43-9006 effects on RENCA cells was also examined using the BrdU assay to evaluate the effect on DNA synthesis (vs. antiproliferative activity using the mitochondrial tetrazolium dye assay). The EC5O for BAY 43-9006 (~5 μM) on RENCA cells obtained using the BrdU method was similar to that observed with the WST-I assay. No direct proliferative responses (or cytotoxicity) was observed when RENCA tumor cells were exposed to range of rIL-2 at concentrations (from 0 to 1 μM), confirming the mechanism of rIL-2 relies on activation of the immune effector cell component. E. In vivo Efficacy Studies rIL-2 and BAY 43-9006 Pharmacokinetics in Mice
In published phase I reports of BAY 43-9006 (at a dose of 400 mg), high Cmax of 10-20 μM and long drug half lives of about 24 hours were achieved in patients (Strumberg, et al., J. Clin. Oncol. 2005 23(5):965-972). To determine if plasma drug exposure may impact T cell viability and proliferative responses in vivo, the single dose pharmacokinetics of rIL-2 and BAY 43-9006 in mice were measured.
A single oral dose of 30 mg/kg BAY 43-9006 in mice achieved a Cmax of about 5500 to 8000 ng/ml (-10 μM) at a tmax of 2 hours. The plasma elimination rate of BAY 43-9006 was fairly slow and the consequent half-life was about 4 hours. In contrast, the PK profile of rIL-2 following subcutaneous administration of 6 mg/kg demonstrated a Cmax of about 550 - 850 ng/ml at a tmax of about 30 minutes. The rlL- 2 ti/2 in mice was approximately 1 hour, with exposures of >50 ng/ml achieved for 4 hours. Interestingly, both treatments after a single dose and at respective routes of administration demonstrated non-overlapping Cmax (at tmax), eluding that both concomitant and sequential treatments may be feasible as supported by the PK findings. Given that BAY 43-9006 may be largely protein bound in blood, the impact of these drug exposures on T cell viability in vivo remains to be addressed.
rIL-2 and BAY 43-9006 treatment decreases immune effector function ex vivo Since inhibition of one or multiple T lymphocytic kinases (Lck, Fyn, Syk, Btk, Src, Tck2, MAPK, JAKs) may abrogate T cell expansion and immune effector function, the effects of rIL-2 and BAY 43-9006 on T cell proliferative and functional responses in vivo was investigated.
In these experiments, BALB/c mice bearing RENCA tumors were treated with rIL-2 (lmg/kg/day, s.c. days 6-10), BAY 43-9006 (30 mg/kg/day, p.o., days 6-10) or combinations of rIL-2 and BAY 43-9006 administered either concomitantly or sequentially (rIL-2, lmg/kg/day, s.c, days 1-5 + BAY 43-9006, 30 mg/kg/day, p.o., days 6-10; or BAY 43-9006, 30 mg/kg/day, p.o., days 1-5 + rIL-2, lmg/kg/day, s.c, days 6-10). Isolated splenocytes from treated mice were then subjected to ex vivo killing assays against Yac-1 target cells at various effector: target (E:T) ratios, and percent specific lysis of 51Cr-labeled Yac-1 cells were determined. Mice treated with rIL-2 (1 mg/kg) significantly increased splenocyte-mediated killing of Yac-1 targets compared to vehicle treatment (23% with rIL-2 vs. 0% with vehicle treatment). The specific killing effects observed with BAY 43-9006 treatment (1%) was negligible and not different from vehicle treatment. All treatments with rf and BAY 43-9006 yielded reduced splenocyte-mediated lytic activity (concomitant treatment = 16%; sequential treatments < 9% vs. rIL-2 monotherapy = 23%).
Effect of rIL-2 and BAY 43-9006 Therapy on Circulating Immune Effector Cell Populations The pharmacodynamic effects of rIL-2 and/or BAY-43-9006 therapy on circulating lymphocyte and monocyte populations tumor-bearing BALB/c mice was examined. In these studies, blood was collected after single agent or combination of rIL-2 and BAY 43-9006 therapy (concomitant or sequential treatments as described earlier). Absolute counts of lymphocytes and T cell sub-populations (CD4+ or CD8+) were quantified in whole blood using TruCount™ tubes and appropriate immunostaining. rIL-2 treatment decreased the absolute numbers of circulating lymphocytes and monocytes (CD45+ cells: 2387 cells/μl vs. 1575 cells/μl with vehicle), and T cells (1550 cells/μl vs. 664 cells/μl with vehicle) in rumored mice. A significantly increased ratio of CD4:CD8 cells was observed with rIL-2 therapy compared to vehicle treatment (5:3; rIL-2: Vehicle, i.e., 1.7-fold), as indicative of rIL-2 mechanism of action in expanding T cell numbers. The relative numbers of non-T and monocytic cells (CD45+CD3-) following rIL-2 treatment were similar to vehicle treatment (837 cells/μl vs. 911 cells/μl with vehicle). In contrast, single agent BAY 43-9006 or BAY 43-9006 combined with rlL-
2 had little impact or increased the absolute numbers of lymphocytes and monocytes (range of 2417-3577 cells/μl vs. 2387 cells/μl with vehicle), and also increased non- T cells and monocyte populations (1241-1563 cells/μl vs. 837 cells/μl with vehicle). The effect of BAY 43-9006 therapy on total numbers of T cells was similar to vehicle treatment (1827 cells/μl vs. 1550 cells/μl with vehicle). Slightly increased total T cell numbers (including both CD4+ and CD8+ populations) observed with the sequential regimen of rIL-2 and BAY 43-9006 when commenced with rIL-2 (2081 T cells/μl vs. 1550 cells/μl with vehicle). When rIL-2 and BAY 43-9006 were given concomitantly or sequenced as BAY 43-9006 then rIL-2, the numbers of total T cells slightly decreased compared to vehicle treatment (-1170-1244 T cells/μl vs. 1550 cells/μl with vehicle). Similar trends were observed in individual CD4+ and CD8+ T cell populations. Additionally, the histology of tumors following treatment with rIL-2 and
BAY 43-9006 or SUl 1248 therapy (as described previously) was determined. To examine the pharmacodynamics of T cells in vivo, tumor infiltrating T cells were detected with a mouse anti-CD3 antibody. The antiproliferative effects in tumors following drug treatments were evaluated using Ki67 staining. With rIL-2 treatment, increased numbers of T cells were seen infiltrating both RENCA tumors (and B16-F10 tumors) compared to vehicle treatment. Generally, fewer T cells were detected in the Bayer 43-9006-treated group in the RENCA model. The effects of rIL-2 and BAY 43-9006 or SUl 1248 therapy on infiltrating T cells in B16-F10 tumors were equivocal, as some T cells were detected in all treated groups. Tumors that demonstrated increased necrosis (tumor inhibition), generally showed higher numbers of T cells interdispersed between tumor cells. Collectively, the data suggests that rIL-2 activates T cells in circulation and traffics cells to the extravascular sites including tumors, and that BAY43-9006 or SUl 1248 treatment may partially abrogate T cell proliferative responses and trafficking.
rIL-2 and BAY 43-9006 Therapy Augments Efficacy of IL-2-Responsive Murine Tumor Models
Drug interactions of rIL-2 with either BAY 43-9006 or SUl 1248 were examined (see Figures 1-10 and Table 10-12). The combination treatments were evaluated in three experimental T-cell competent, murine IL-2 -responsive tumor models (B16-F10 melanoma, CT26 colon, RCC RENCA model) (Figures 1-10). In these studies, mice were randomized when tumors were established to a size of 50- 225 mm3, and the growth of tumors were monitored by calliper measurements following daily oral dosing of BAY 43-9006 or SUl 1248 or subcutaneous daily administration of rIL-2 (as indicated in methods). Single agent efficacy and tolerability of rIL-2, BAY 43-9006 and SUl 1248 were investigated at a range of doses and treatment schedules (Figure 1). In all models, rIL-2 demonstrated potent efficacy and tumor inhibitions at effective doses were generally in the range of 40- 60% (vs. vehicle treatment) (Figure 1). The minimum effective dose for BAY 43- 9006 was >30 mg/kg/day (vs. vehicle treatment). SUl 1248 was generally effective at doses > 40 mg/kg/day, with the exception of the Bl 6-F 10 tumor model (Figure
I)-
Based on the single agent efficacy and tolerability, rIL-2 was then combined with BAY 43-9006 or SUl 1248 at doses that were tolerated and exhibited no deleterious effects on body weight or any adverse clinical symptoms. Both sequential and concomitant treatment schedules were examined in the Bl 6-F 10 and CT26 tumor models (Figures 2-8, Tables 10 and 11). In the B16-F10 and CT26 tumor models, almost all combination therapies (concomitant or sequential regimens) investigated with rIL-2 and BAY 43-9006 or rIL-2 and SU12248 augmented antitumor activity compared to monotherapy or vehicle treatments (Figures 2-5). A summary of the combination drug interactions of the various combination therapies is summarized in Tables 10 and 11.
Table 10. Efficacy of single agent rIL-2, BAY 43-9006, SU11248 and combinations of rIL-2 and BAY 43-9006/SU11248 in the murine melanoma B16- FlO tumor model in C57BL6 mice.
Figure imgf000086_0001
8. rIL-2 (3.3 mg/kg, s.c, days 848 0.40 0.31 1.02 (day Additive
0-6) + BAY 43-9006 (30 8) mg/kg, p. o. days 0-6)
B. rIL-2 + SU11248 efficacy:
7. Vehicle 2415 1 -00 N/A N/A N/A
2. rIL-2 3.3mg/kg/d, s.c, 934 0.39 N/A N/A N/A
3. rIL-2 3.3mg/kg/d, s.c, 1042 0.43 N/A N/A N/A days 7-13
4. SU11248 40mg/kg/d, p.o. 1756 0.72 N/A N/A N/A
5. SU11248 40mg/kg/d, p.o., 1701 0.70 N/A N/A N/A days 7-75
6. rIL-2 3.3mg/kg/d, s.c. days 583 0.24 0.28 1.20 Additive
0-5 + SUl 1248 40mg/kg/d, s.c, days 7-13
7. SU1124840mg/kg/d, p.o. 583 0.24 0.31 1.30 Additive days 0-6 + rIL-2 3.3mg/kg/d, s.c. days 7-13
8. rIL-2 3.3mg/kg/d, s.c. days 743 0.31 0.28 0.90 Additive 0-6 + SUIl 248 40mg/kg/d, p.o., days 0-6 an= 10 C57BL6 mice/group in each study. B16-F10 tumored mice were treated tumors were established to a mean size of ~50 mm3. b T/Cobserved (O) = % T/C
C T/CeXpected (E) = %T/C treatment 1 x %T/C treatment 2 d Synergistic effects were defined when the ratio of expected % tumor growth inhibition of combination therapy (%T/Cexp = %T/C treatment 1 x %T/C treatment 2) divided by observed % T/C (%T/Cobs) of the combination treatment was >1. e Drug interactions were defined as additive when %T/Cexp/%T/Cobs =1, and antagonism when %T/Cexp/%T/Cobs <1. N/A, not applicable
Table 11. Efficacy of rIL-2, BAY 43-9006, SU11248 and combinations of rIL-2 and BAY 43-9006/SU11248 in the murine CT26 colon tumor model in BALB/c mice.
Figure imgf000087_0001
2462 0.95 N/A N/A N/A
1773 0.68 N/A N/A N/A
2572 0.99 N/A N/A N/A
2127 0.83 N/A N/A N/A
1581 0.61 N/A N/A N/A
1114 0.43 0.76 1.77 Additive/
Synergistic
1250 0.49 0.47 0.97 Additive
2336 0.91 0.66 0.72 Sub- Additive
1006 0.39 0.79 2.02 Additive/
Synergistic
1502 0.58 0.53 0.90 Additive
1137 0.44 0.63 1.43 Additive
Figure imgf000088_0001
an= 10 BALB/c mice/group in each study. CT26 tumor-bearing mice were treated when tumors were established to a mean size of -225 mm3. bT/Cobserved (O)= % T/C
CT/Cexpected (E) = %T/C treatment 1 x %T/C treatment 2 d Synergistic effects were defined when the ratio of expected % tumor growth inhibition of combination therapy (%T/Cexp = %T/C treatment 1 x %T/C treatment 2) divided by observed % T/C (%T/Cobs) of the combination treatment was >1. e Drug interactions were defined as additive when %T/Cexp/%T/Cobs =1, and antagonism when %T/Cexp/%T/Cobs <1.
N/A, not applicable
In only one case in the CT26 model, when BAY 43-9006 (40 mg/kg/day, p.o. days 1-7) was administered prior to rIL-2 (1 mg/kg/day, s.c. days 8-13), the effects of the combined treatment was sub-optimal.
In the RENCA model, only concomitant schedules were evaluated (Figures 9- 10; Table 12), and combinations of rIL-2 with either BAY 43-9006 or SUl 1248 demonstrated greater tumor inhibition compared to single agent therapy. Table 12. Efficacy of single agent rIL-2, BAY 43-9006, SU11248 and combinations of rIL-2 and BAY 43-9006/SU11248 in the murine RCC RENCA tumor model in BALB/c mice.
Treatmenf Mean O" Ec E/θ" Drug tumor (T/Cobs) (TZCex,,) (0ZoTZCex/ Interaction1* volume %TZCobJ
(mm3) day 10 or
17
A. rIL-2 + BAY 43-9006 efficacy:
1. Vehicle, days 0-8 579 1.00 N/A N/A N/A
2. rIL-2 lmg/kg/d, s.c, days 288 0.50 N/A N/A N/A 0-4, 7-11
3. BAY 43-90063 Omg/kg/d, 273 0.47 N/A N/A N/A p.o., days 0-8
4. rIL-2 lmg/kg/d, s.c. days 154 0.27 0.23 0.88 Additive 0-4, 7-11 + BAY 43-9006
30mg/kg/d, p.o., days 0-8 (concomitant)
B. rIL-2 + SU11248 efficacy:
1. Vehicle 712 1.00 N/A N/A N/A
2. rIL-2 1 mg/kg/d, days 0-6 525 0.74 N/A N/A N/A
3. 169549 40 mg/kg/d, days 527 0.74 N/A N/A N/A 0-6
4. 169549 40 mg/kg/d, days 414 0.58 0.55 0.94 Additive 0-6ML-2 1.0 mg/kg/d, days 0-6 (concomitant) an= 10 BALB/c mice/group in each study. RENCA model tumored mice were treated tumors were established to a mean size of -50-70 mm3. Concomitant regimens are presented as RENCA tumor model exhibits severe cachexia (animal wasting) making sequential administration of agents not possible, and not evaluable. b T/Cobserved (O) = % T/C
C T/Ceχpected (E) = %T/C treatment 1 x %T/C treatment 2 d Synergistic effects were defined when the ratio of expected % tumor growth inhibition of combination therapy (%T/Cexp = %T/C treatment 1 x %T/C treatment 2) divided by observed % T/C (%T/Cobs) of the combination treatment was >1. eDrug interactions were defined as additive when %T/Cexp/%T/Cobs =1, and antagonism when %T/Cexp/%T/Cobs <1. N/A, not applicable
Sequential treatments in the RENCA model were not feasible due to the short model duration and mouse cachexia (animal wasting/body weight loss). Collectively, the data demonstrate enhanced activity when rIL-2 was combined with targeted small molecule therapies like BAY 43-9006 and SUl 1248 in preclinical models, indicating that such therapeutic strategies could be translated to the clinic. While the invention has been described with respect to specific examples including presently preferred modes of carrying out the invention, those skilled in the art will appreciate that there are numerous variations and permutations of the above described systems and techniques that fall within the spirit and scope of the invention.
INCORPORATION BY REFERENCE
The contents of all of the above cited patents, patent applications and journal articles are incorporated by reference as if set forth fully herein.

Claims

1. A method of treating a patient suffering from cancer comprising administering to said patient a therapeutically effective amount of aldesleukin and an antiangiogenic agent selected from 6,7-bis(2-methoxyethoxy)-N-(3- ethynylphenyl)quinazolin-4-amine, 6-(3-morpholinopropoxy)-N-(3-chloro-4- fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2-(dimethylamino)ethyl)-5-((5- fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl- 1 H-pyrrole-3-carboxamide, N-(4- chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin- 1 -amine, or 1 -(4-(2- (methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluoromethyl)phenyl)urea.
2. The method of claim 1, wherein the aldesleukin is administered prior to the antiangiogenic agent.
3. The method of claim 1, wherein the aldesleukin is administered subsequent to the antiangiogenic agent.
4. The method of claim 1, wherein the aldesleukin is administered concurrent with the antiangiogenic agent.
5. The method of claim 4, wherein the aldesleukin is administered in a separate composition from the antiangiogenic agent.
6. The method of any one of claims 1-5, comprising separately administering to said patient a therapeutically effective amount of aldesleukin and an antiangiogenic agent according to a dosing schedule, wherein the aldesleukin is administered from 1 to 3 times daily in a dose between about 9 and about 130 MIU/day for a period of at least 3 consecutive days, optionally followed by a rest period of at least 3 consecutive days.
7. The method of claim 6, wherein said antiangiogenic agent is administered from 1 to 6 times every 2-3 weeks.
8. The method of claim 6, wherein the aldesleukin is administered intravenously and said rest period is present.
9. The method of claim 6, wherein the aldesleukin is administered subcutaneously and said rest period is absent.
10. The method of claim 9, wherein the aldesleukin is administered 1-3 times daily in a dose of about 9-30 MIU/day.
11. The method of claim 6, wherein the aldesleukin is administered 3 times daily in a dose of about 30-130 MILJ/day.
12. The method of claim 6, wherein the aldesleukin is administered for a period of 5 consecutive days followed by a 9-day rest period.
13. The method of claim 6, wherein said dosing schedule is repeated for at least two courses.
14. The method of claim 6, wherein said dosing schedule is repeated for 3 courses.
15. The method of claim 6, wherein said dosing schedule is repeated for 4 courses.
16. The method of claim 6, wherein the aldesleukin is administered 3 times for the first day and once daily each proceeding day.
17. The method of any one of claims 1-16, wherein the cancer is renal cell carcinoma, melanoma or colon cancer.
18. The method of claim 17, further comprising administering to the patient at least one compound selected from acetaminophen, meperidine, indomethacin, ranitidine, nizatidine, diastop, loperamide, diphenhydramine, or furosemide subsequent to or concurrently with administration of aldesleukin.
19. The method of any one of claims 1-18, wherein said method results in the amelioration of cancer in the patient.
20. The method of any one of claims 1-18, wherein said method results in the attenuation of hypotension in the patient.
21. The method of any one of claims 1-18, wherein said method results in the attenuation of hypertension in the patient.
22. The method of any one of claims 1-18, wherein said method results in the reduction of nitric oxide synthase in the patient.
23. The method of any one of claims 1-18, wherein the cancer is susceptible to inhibition of angiogenesis and/or immune stimulation.
24. The method of any one of claims 1-23, wherein said antiangiogenic agent is N-(2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4- diethyl-lH-pyrrole-3-carboxamide.
25. The method of any one of claims 1-23, wherein said antiangiogenic agent is l-(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluoromethyl)phenyl)urea .
26. A composition comprising: (a) a therapeutically effective amount of aldesleukin; (b) a therapeutically effective amount of an antiangiogenic agent selected from 6,7-bis(2-methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6-(3- morpholinopropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N- (2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl- lH-pyrrole-3-carboxarnide, N-(4-chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin-l- amine, or l-(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluoromethyl)phenyl)urea; and (c) a pharmaceutically acceptable excipient.
27. A kit comprising a combination of medicaments for the treatment of a patient suffering from cancer, comprising: (a) aldesleukin, and (b) an antiangiogenic agent selected from 6,7-bis(2-methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4- amine, 6-(3-morpholinopropoxy)-N-(3-chloro-4-fluorophenyl)-7-methoxyqumazolin- 4-amine, N-(2-(dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)- 2,4-diethyl-lH-pyrrole-3-carboxamide, N-(4-chlorophenyl)-4-((pyridin-4- yl)methyl)phthalazin- 1 -amine, or 1 -(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)- 3-(4-chloro-3-(trifluoromethyl)phenyl)urea, for simultaneous, sequential or separate use.
28. The kit of claim 26, wherein each of said medicaments is separately packaged.
29. Use of aldesleukin and an antiangiogenic agent selected from 6,7-bis(2- methoxyethoxy)-N-(3-ethynylphenyl)quinazolin-4-amine, 6-(3-morpholinopropoxy)- N-(3-chloro-4-fluorophenyl)-7-methoxyquinazolin-4-amine, N-(2- (dimethylamino)ethyl)-5-((5-fluoro-2-oxoindolin-3-ylidene)methyl)-2,4-diethyl-lH- pyrrole-3-carboxamide, N-(4-chlorophenyl)-4-((pyridin-4-yl)methyl)phthalazin- 1 - amine, or 1 -(4-(2-(methylcarbamoyl)pyridin-4-yloxy)phenyl)-3-(4-chloro-3- (trifluoromethyl)phenyl)urea, in the manufacture of one or more medicaments for treating a patient suffering from cancer.
30. The use of claim 29, wherein the aldesleukin is formulated for administration prior to the antiangiogenic agent.
31. The use of claim 29, wherein the aldesleukin is formulated for administration subsequent to the antiangiogenic agent.
32. The use of claim 29, wherein the aldesleukin is formulated for administration concurrent with the antiangiogenic agent.
PCT/US2006/005720 2005-02-18 2006-02-17 Antiangiogenic agents with aldesleukin WO2006089150A2 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
EP06735400A EP1853302A2 (en) 2005-02-18 2006-02-17 Antiangiogenic agents with aldesleukin
BRPI0608880-5A BRPI0608880A2 (en) 2005-02-18 2006-02-17 anti-angiogenic agents with aldesleukin
AU2006214138A AU2006214138A1 (en) 2005-02-18 2006-02-17 Antiangiogenic agents with aldesleukin
JP2007556337A JP2008530239A (en) 2005-02-18 2006-02-17 Anti-angiogenic agents containing aldesleukin
MX2007010037A MX2007010037A (en) 2005-02-18 2006-02-17 Antiangiogenic agents with aldesleukin.
CA002598448A CA2598448A1 (en) 2005-02-18 2006-02-17 Antiangiogenic agents with aldesleukin

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US65434105P 2005-02-18 2005-02-18
US60/654,341 2005-02-18

Publications (2)

Publication Number Publication Date
WO2006089150A2 true WO2006089150A2 (en) 2006-08-24
WO2006089150A3 WO2006089150A3 (en) 2006-11-02

Family

ID=36569884

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/005720 WO2006089150A2 (en) 2005-02-18 2006-02-17 Antiangiogenic agents with aldesleukin

Country Status (10)

Country Link
EP (1) EP1853302A2 (en)
JP (1) JP2008530239A (en)
KR (1) KR20070108909A (en)
CN (1) CN101146549A (en)
AU (1) AU2006214138A1 (en)
BR (1) BRPI0608880A2 (en)
CA (1) CA2598448A1 (en)
MX (1) MX2007010037A (en)
RU (1) RU2007134570A (en)
WO (1) WO2006089150A2 (en)

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2011528676A (en) * 2008-07-24 2011-11-24 バイエル ファーマ アクチエンゲゼルシャフト Sulfone-substituted quinazoline derivatives as immunomodulators, their preparation and use as pharmaceuticals
WO2012079075A1 (en) 2010-12-10 2012-06-14 Concert Pharmaceuticals, Inc. Deuterated phthalimide derivatives
CN101497607B (en) * 2008-01-29 2012-11-28 上海百灵医药科技有限公司 Process for synthesizing sunitinib
WO2013130849A1 (en) 2012-02-29 2013-09-06 Concert Pharmaceuticals, Inc. Substituted dioxopiperidinyl phthalimide derivatives
WO2013159026A1 (en) 2012-04-20 2013-10-24 Concert Pharmaceuticals, Inc. Deuterated rigosertib
US8575184B2 (en) 2009-09-03 2013-11-05 Bristol-Myers Squibb Company Quinazolines as potassium ion channel inhibitors
WO2014066243A1 (en) 2012-10-22 2014-05-01 Concert Pharmaceuticals, Inc. Solid forms of {s-3-(4-amino-1-oxo-isoindolin-2yl)(piperidine-3,4,4,5,5-d5)-2,6-dione}
WO2014110322A2 (en) 2013-01-11 2014-07-17 Concert Pharmaceuticals, Inc. Substituted dioxopiperidinyl phthalimide derivatives
WO2014170910A1 (en) 2013-04-04 2014-10-23 Natco Pharma Limited Process for the preparation of lapatinib
US9045453B2 (en) 2008-11-14 2015-06-02 Concert Pharmaceuticals, Inc. Substituted dioxopiperidinyl phthalimide derivatives
US9145411B2 (en) 2012-08-02 2015-09-29 Asana Biosciences, Llc Substituted amino-pyrimidine derivatives
US9353122B2 (en) 2013-02-15 2016-05-31 Kala Pharmaceuticals, Inc. Therapeutic compounds and uses thereof
US9353123B2 (en) 2013-02-20 2016-05-31 Kala Pharmaceuticals, Inc. Therapeutic compounds and uses thereof
US9688688B2 (en) 2013-02-20 2017-06-27 Kala Pharmaceuticals, Inc. Crystalline forms of 4-((4-((4-fluoro-2-methyl-1H-indol-5-yl)oxy)-6-methoxyquinazolin-7-yl)oxy)-1-(2-oxa-7-azaspiro[3.5]nonan-7-yl)butan-1-one and uses thereof
US9790232B2 (en) 2013-11-01 2017-10-17 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
US9890173B2 (en) 2013-11-01 2018-02-13 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
US10253036B2 (en) 2016-09-08 2019-04-09 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
US10336767B2 (en) 2016-09-08 2019-07-02 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
US10766907B2 (en) 2016-09-08 2020-09-08 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
US10844039B2 (en) 2018-11-13 2020-11-24 Biotheryx, Inc. Substituted isoindolinones
CN114920704A (en) * 2019-07-26 2022-08-19 暨南大学 Phenyl piperazine quinazoline compound or pharmaceutically acceptable salt thereof, preparation method and application

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5647615B2 (en) * 2008-11-14 2015-01-07 コンサート ファーマシューティカルズ インコーポレイテッド Substituted dioxopiperidinylphthalimide derivatives
KR101478925B1 (en) * 2013-12-31 2015-01-08 아주대학교산학협력단 Pharmaceutical composition for treating or preventing cancer disease comprising proteasome inhibitor and loperamide
CN108069913B (en) * 2016-11-18 2022-03-01 陕西师范大学 Bis (morpholinylalkoxy) quinazoline derivative and application thereof in anti-tumor aspect
CN111499583B (en) * 2020-05-22 2022-02-15 沈阳工业大学 Quinazoline derivative and application thereof as antitumor drug
CN112538071A (en) * 2020-11-30 2021-03-23 新乡医学院第一附属医院 Compounds useful as RAF-1 inhibitors, methods of preparation and uses

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
SONI SANDEEP ET AL: "Treatment of pediatric ocular melanoma with high-dose interleukin-2 and thalidomide." JOURNAL OF PEDIATRIC HEMATOLOGY/ONCOLOGY : OFFICIAL JOURNAL OF THE AMERICAN SOCIETY OF PEDIATRIC HEMATOLOGY/ONCOLOGY. 2002 AUG-SEP, vol. 24, no. 6, August 2002 (2002-08), pages 488-491, XP009067555 ISSN: 1077-4114 *
STERNBERG C N: "Metastatic renal cell cancer treatments." DRUGS OF TODAY, vol. 39, no. Suppl. C, November 2003 (2003-11), pages 39-59, XP009067549 ISSN: 0025-7656 *

Cited By (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101497607B (en) * 2008-01-29 2012-11-28 上海百灵医药科技有限公司 Process for synthesizing sunitinib
JP2011528676A (en) * 2008-07-24 2011-11-24 バイエル ファーマ アクチエンゲゼルシャフト Sulfone-substituted quinazoline derivatives as immunomodulators, their preparation and use as pharmaceuticals
US9045453B2 (en) 2008-11-14 2015-06-02 Concert Pharmaceuticals, Inc. Substituted dioxopiperidinyl phthalimide derivatives
US11008306B2 (en) 2009-09-03 2021-05-18 Bristol-Myers Squibb Company Quinazolines as potassium ion channel inhibitors
US9822096B2 (en) 2009-09-03 2017-11-21 Bristol-Myers Squibb Company Quinazolines as potassium ion channel inhibitors
US9458114B2 (en) 2009-09-03 2016-10-04 Bristol-Myers Squibb Company Quinazolines as potassium ion channel inhibitors
US8575184B2 (en) 2009-09-03 2013-11-05 Bristol-Myers Squibb Company Quinazolines as potassium ion channel inhibitors
US10676460B2 (en) 2009-09-03 2020-06-09 Bristol-Myers Squibb Company Quinazolines as potassium ion channel inhibitors
US10214511B2 (en) 2009-09-03 2019-02-26 Bristol-Myers Squibb Company Quinazolines as potassium ion channel inhibitors
WO2012079075A1 (en) 2010-12-10 2012-06-14 Concert Pharmaceuticals, Inc. Deuterated phthalimide derivatives
WO2013130849A1 (en) 2012-02-29 2013-09-06 Concert Pharmaceuticals, Inc. Substituted dioxopiperidinyl phthalimide derivatives
WO2013159026A1 (en) 2012-04-20 2013-10-24 Concert Pharmaceuticals, Inc. Deuterated rigosertib
US9145411B2 (en) 2012-08-02 2015-09-29 Asana Biosciences, Llc Substituted amino-pyrimidine derivatives
WO2014066243A1 (en) 2012-10-22 2014-05-01 Concert Pharmaceuticals, Inc. Solid forms of {s-3-(4-amino-1-oxo-isoindolin-2yl)(piperidine-3,4,4,5,5-d5)-2,6-dione}
US9643950B2 (en) 2012-10-22 2017-05-09 Concert Pharmaceuticals, Inc. Solid forms of {s-3-(4-amino-1-oxo-isoindolin-2-yl)(piperidine-3,4,4,5,5-d5)-2,6-dione}
WO2014110322A2 (en) 2013-01-11 2014-07-17 Concert Pharmaceuticals, Inc. Substituted dioxopiperidinyl phthalimide derivatives
US9877970B2 (en) 2013-02-15 2018-01-30 Kala Pharmaceuticals, Inc. Therapeutic compounds and uses thereof
US10398703B2 (en) 2013-02-15 2019-09-03 Kala Pharmaceuticals, Inc. Therapeutic compounds and uses thereof
US9827248B2 (en) 2013-02-15 2017-11-28 Kala Pharmaceuticals, Inc. Therapeutic compounds and uses thereof
US10966987B2 (en) 2013-02-15 2021-04-06 Kala Pharmaceuticals, Inc. Therapeutic compounds and uses thereof
US9353122B2 (en) 2013-02-15 2016-05-31 Kala Pharmaceuticals, Inc. Therapeutic compounds and uses thereof
US9833453B2 (en) 2013-02-20 2017-12-05 Kala Pharmaceuticals, Inc. Therapeutic compounds and uses thereof
US9353123B2 (en) 2013-02-20 2016-05-31 Kala Pharmaceuticals, Inc. Therapeutic compounds and uses thereof
US9861634B2 (en) 2013-02-20 2018-01-09 Kala Pharmaceuticals, Inc. Therapeutic compounds and uses thereof
US11369611B2 (en) 2013-02-20 2022-06-28 Kala Pharmaceuticals, Inc. Therapeutic compounds and uses thereof
US10285991B2 (en) 2013-02-20 2019-05-14 Kala Pharmaceuticals, Inc. Therapeutic compounds and uses thereof
US10758539B2 (en) 2013-02-20 2020-09-01 Kala Pharmaceuticals, Inc. Therapeutic compounds and uses thereof
US9688688B2 (en) 2013-02-20 2017-06-27 Kala Pharmaceuticals, Inc. Crystalline forms of 4-((4-((4-fluoro-2-methyl-1H-indol-5-yl)oxy)-6-methoxyquinazolin-7-yl)oxy)-1-(2-oxa-7-azaspiro[3.5]nonan-7-yl)butan-1-one and uses thereof
WO2014170910A1 (en) 2013-04-04 2014-10-23 Natco Pharma Limited Process for the preparation of lapatinib
US9890173B2 (en) 2013-11-01 2018-02-13 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
US10975090B2 (en) 2013-11-01 2021-04-13 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
US10618906B2 (en) 2013-11-01 2020-04-14 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
US11713323B2 (en) 2013-11-01 2023-08-01 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
US9790232B2 (en) 2013-11-01 2017-10-17 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
US10160765B2 (en) 2013-11-01 2018-12-25 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
US10766907B2 (en) 2016-09-08 2020-09-08 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
US10253036B2 (en) 2016-09-08 2019-04-09 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
US11021487B2 (en) 2016-09-08 2021-06-01 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
US11104685B2 (en) 2016-09-08 2021-08-31 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
US10626121B2 (en) 2016-09-08 2020-04-21 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
US10336767B2 (en) 2016-09-08 2019-07-02 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
US10844039B2 (en) 2018-11-13 2020-11-24 Biotheryx, Inc. Substituted isoindolinones
US11352338B2 (en) 2018-11-13 2022-06-07 Biotheryx, Inc. Substituted isoindolinones
CN114920704A (en) * 2019-07-26 2022-08-19 暨南大学 Phenyl piperazine quinazoline compound or pharmaceutically acceptable salt thereof, preparation method and application
CN114920704B (en) * 2019-07-26 2023-11-03 暨南大学 Phenyl piperazine quinazoline compound or pharmaceutically acceptable salt thereof, preparation method and application

Also Published As

Publication number Publication date
AU2006214138A1 (en) 2006-08-24
WO2006089150A3 (en) 2006-11-02
MX2007010037A (en) 2008-02-15
RU2007134570A (en) 2009-03-27
EP1853302A2 (en) 2007-11-14
JP2008530239A (en) 2008-08-07
KR20070108909A (en) 2007-11-13
CA2598448A1 (en) 2006-08-24
BRPI0608880A2 (en) 2010-02-02
CN101146549A (en) 2008-03-19

Similar Documents

Publication Publication Date Title
EP1853302A2 (en) Antiangiogenic agents with aldesleukin
CA2707554C (en) Use of 3-(indolyl)-or 3-(azaindolyl)-4-arylmaleimide derivatives in leukemia management
JPWO2008111441A1 (en) Pharmaceutical composition
EA027654B1 (en) Combination of panobinostat and ruxolitinib in the treatment of cancer such as a myeloproliferative neoplasm
US20100041668A1 (en) Compositions and methods for treating thrombocytopenia
EP2885003B1 (en) Combination of pi3k inhibitor and c-met inhibitor
JP2019038821A (en) PIM kinase inhibitor combinations
TW201806600A (en) Combination of a BCL-2 inhibitor and a MCL1 inhibitor, uses and pharmaceutical compositions thereof
Zauli et al. State of the art of the therapeutic perspective of sorafenib against hematological malignancies
JP2019528250A (en) Combination of BCL-2 inhibitor and MCL-1 inhibitor, use thereof and pharmaceutical composition
TW202342047A (en) Combination therapy using substituted pyrimidin-4(3h)-ones and sotorasib
KR20220011562A (en) Methods and compositions for reducing the risk of recurrence and prolonging survival in acute myeloid leukemia
WO2022063134A1 (en) Csf1r kinase inhibitor and use thereof
KR20070083705A (en) Use of 4-(4-methylpiperazin-1-ylmethyl)-n-[4-methyl-3-(4-(pyridin-3-yl)pyrimidin-2-ylamino)phenyl]-benzamide to inhibit the tyrosine kinase receptor c-fms
TW202404599A (en) Combination therapy using substituted pyrimidin-4(3h)-ones and nivolumab
KR20230110751A (en) CXCR1/CXCR2 inhibitors for use in the treatment of myelofibrosis
Kulasekararaj et al. The Non-transplant Treatment of Myelodysplastic Syndromes—What's on the Horizon?
TW202342043A (en) Combination therapy using a ptpn11 inhibitor and an egfr inhibitor
WO2023059785A9 (en) Substituted pyrimidin-4(3h)-ones for use in treating cancer
Testa Membrane tyrosine kinase receptors kit and FLT3 are an important targets for the therapy of acute myeloid leukemia
CN114450008A (en) Use of MDM2 inhibitors for the treatment of myelofibrosis
Mesa et al. Conventional and experimental drug therapy in myelofibrosis with myeloid metaplasia
EA045240B1 (en) CANCER TREATMENT METHODS
EA039621B1 (en) Combination of a bcl-2 inhibitor and a mcl1 inhibitor, uses and pharmaceutical compositions thereof
JP2005508846A (en) Use of N-phenyl-2-pyrimidinamine derivatives for mast cell diseases such as allergic diseases

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200680009316.0

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref document number: 2598448

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: MX/a/2007/010037

Country of ref document: MX

Ref document number: 2007556337

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2006214138

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 3324/KOLNP/2007

Country of ref document: IN

ENP Entry into the national phase

Ref document number: 2006214138

Country of ref document: AU

Date of ref document: 20060217

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1020077021118

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 2006735400

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2007134570

Country of ref document: RU

ENP Entry into the national phase

Ref document number: PI0608880

Country of ref document: BR

Kind code of ref document: A2