AU3981401A - Tyrosine kinase inhibitors - Google Patents

Tyrosine kinase inhibitors Download PDF

Info

Publication number
AU3981401A
AU3981401A AU39814/01A AU3981401A AU3981401A AU 3981401 A AU3981401 A AU 3981401A AU 39814/01 A AU39814/01 A AU 39814/01A AU 3981401 A AU3981401 A AU 3981401A AU 3981401 A AU3981401 A AU 3981401A
Authority
AU
Australia
Prior art keywords
optionally substituted
alkyl
substituents selected
compound
uso1
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
AU39814/01A
Other versions
AU778417B2 (en
Inventor
Mark E. Fraley
George D. Hartman
Randall W. Hungate
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Merck and Co Inc
Original Assignee
Merck and Co 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 Merck and Co Inc filed Critical Merck and Co Inc
Publication of AU3981401A publication Critical patent/AU3981401A/en
Application granted granted Critical
Publication of AU778417B2 publication Critical patent/AU778417B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Diabetes (AREA)
  • Rheumatology (AREA)
  • Dermatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Emergency Medicine (AREA)
  • Ophthalmology & Optometry (AREA)
  • Pain & Pain Management (AREA)
  • Obesity (AREA)
  • Immunology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Hematology (AREA)
  • Endocrinology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)

Description

WO 01/62252 PCT/USO1/05567 TITLE OF THE INVENTION TYROSINE KINASE INHIBITORS BACKGROUND OF THE INVENTION 5 The present invention relates to compounds which inhibit, regulate and/or modulate tyrosine kinase signal transduction, compositions which contain these compounds, and methods of using them to treat tyrosine kinase-dependent diseases and conditions, such as angiogenesis, cancer, tumor growth, atherosclerosis, age related macular degeneration, diabetic retinopathy, inflammatory diseases, and the 10 like in mammals. Tyrosine kinases are a class of enzymes that catalyze the transfer of the terminal phosphate of adenosine triphosphate to tyrosine residues in protein substrates. Tyrosine kinases are believed, by way of substrate phosphorylation, to play critical roles in signal transduction for a number of cell functions. Though the 15 exact mechanisms of signal transduction is still unclear, tyrosine kinases have been shown to be important contributing factors in cell proliferation, carcinogenesis and cell differentiation. Tyrosine kinases can be categorized as receptor type or non-receptor type. Receptor type tyrosine kinases have an extracellular, a transmembrane, and an 20 intracellular portion, while non-receptor type tyrosine kinases are wholly intracellular. The receptor-type tyrosine kinases are comprised of a large number of transmembrane receptors with diverse biological activity. In fact, about 20 different subfamilies of receptor-type tyrosine kinases have been identified. One tyrosine kinase subfamily, designated the HER subfamily, is comprised of EGFR, HER2, 25 HER3, and BER4. Ligands of this subfamily of receptors include epithileal growth factor, TGF-a, amphiregulin, BB-EGF, betacellulin and heregulin. Another subfamily of these receptor-type tyrosine kinases is the insulin subfamily, which - 1 - WO 01/62252 PCT/USO1/05567 includes INS-R, IGF-IR, and IR-R. The PDGF subfamily includes the PDGF-a and $ receptors, CSFIR, c-kit and FLK-Il. Then there is the FLK family which is comprised of the kinase insert domain receptor (KDR), fetal liver kinase-1 (FLK-1), fetal liver kinase-4 (FLK-4) and the fins-like tyrosine kinase-1 (fit-1). The PDGF and FLK 5 families are usually considered together due to the similarities of the two groups. For a detailed discussion of the receptor-type tyrosine kinases, see Plowman et al., DN&P 7(6):334-339, 1994, which is hereby incorporated by reference. The non-receptor type of tyrosine kinases is also comprised of numerous subfamilies, including Src, Frk, Btk, Csk, Abl, Zap70, Fes/Fps, Fak, 10 Jak, Ack, and LIMK. Each of these subfamilies is further sub-divided into varying receptors. For example, the Src subfamily is one of the largest and includes Src, Yes, Fyn, Lyn, Lck, Blk, Hck, Fgr, and Yrk. The Src subfamily of enzymes has been linked to oncogenesis. For a more detailed discussion of the non-receptor type of tyrosine kinases, see Bolen Oncogene, 8:2025-2031 (1993), which is hereby 15 incorporated by reference. Both receptor-type and non-receptor type tyrosine kinases are implicated in cellular signaling pathways leading to numerous pathogenic conditions, including cancer, psoriasis and hyperimmune responses. Several receptor-type tyrosine kinases, and the growth factors that 20 bind thereto, have been suggested to play a role in angiogenesis, although some may promote angiogenesis indirectly (Mustonen and Alitalo, J. Cell Biol. 129:895-898, 1995). One such receptor-type tyrsoine kinase is fetal liver kinase 1 or FLK-1. The human analog of FLK-1 is the kinase insert domain-containing receptor KDR, which is also known as vascular endothelial cell growth factor receptor 2 or VEGFR-2, since 25 it binds VEGF with high affinity. Finally, the murine version of this receptor has also been called NYK (Oelrichs et al., Oncogene 8(1):11-15, 1993). VEGF and KDR are a ligand-receptor pair that play an important role in the proliferation of vascular endo -2- WO 01/62252 PCT/USO1/05567 thelial cells, and the formation and sprouting of blood vessels, termed vasculogenesis and angiogenesis, respectively. Angiogenesis is characterized by excessive activity of vascular endothelial growth factor (VEGF). VEGF is actually comprised of a family of ligands 5 (Klagsburn and D'Amore, Cytokine &Growth Factor Reviews 7:259-270, 1996). VEGF binds the high affinity membrane-spanning tyrosine kinase receptor KDR and the related fms-like tyrosine kinase-1, also known as Flt-1 or vascular endothelial cell growth factor receptor 1 (VEGFR-1). Cell culture and gene knockout experiments indicate that each receptor contributes to different aspects of angiogenesis. KDR 10 mediates the mitogenic function of VEGF whereas Flt-1 appears to modulate non mitogenic functions such as those associated with cellular adhesion. Inhibiting KDR thus modulates the level of mitogenic VEGF activity. In fact, tumor growth has been shown to be susceptible to the antiangiogenic effects of VEGF receptor antagonists. (Kim et al., Nature 362, pp. 841-844, 1993). 15 Solid tumors can therefore be treated by tyrosine kinase inhibitors since these tumors depend on angiogenesis for the formation of the blood vessels necessary to support their growth. These solid tumors include histiocytic lymphoma, cancers of the brain, genitourinary tract, lymphatic system, stomach, larynx and lung, including lung adenocarcinoma and small cell lung cancer. Additional examples 20 include cancers in which overexpression or activation of Raf-activating oncogenes (e.g., K-ras, erb-B) is observed. Such cancers include pancreatic and breast carcinoma. Accordingly, inhibitors of these tyrosine kinases are useful for the prevention and treatment of proliferative diseases dependent on these enzymes. The angiogenic activity of VEGF is not limited to tumors. VEGF 25 accounts for most of the angiogenic activity produced in or near the retina in diabetic retinopathy. This vascular growth in the retina leads to visual degeneration culminat ing in blindness. Ocular VEGF mRNA and protein are elevated by conditions such -3- WO 01/62252 PCT/USO1/05567 as retinal vein occlusion in primates and decreased P 0 2 levels in mice that lead to neovascularization. Intraocular injections of anti-VEGF monoclonal antibodies or VEGF receptor immunofusions inhibit ocular neovascularization in both primate and rodent models. Regardless of the cause of induction of VEGF in human diabetic 5 retinopathy, inhibition of ocular VEGF is useful in treating the disease. Expression of VEGF is also significantly increased in hypoxic regions of animal and human tumors adjacent to areas of necrosis. VEGF is also upregulated by the expression of the oncogenes ras, raf, src and mutant p53 (all of which are relevant to targeting cancer). Monoclonal anti-VEGF antibodies inhibit the growth 10 of human tumors in nude mice. Although these same tumor cells continue to express VEGF in culture, the antibodies do not diminish their mitotic rate. Thus tumor derived VEGF does not function as an autocrine mitogenic factor. Therefore, VEGF contributes to tumor growth in vivo by promoting angiogenesis through its paracrine vascular endothelial cell chemotactic and mitogenic activities. These monoclonal 15 antibodies also inhibit the growth of typically less well vascularized human colon cancers in athymic mice and decrease the number of tumors arising from inoculated cells. Viral expression of a VEGF-binding construct of Flk-1, Flt-1, the mouse KDR receptor homologue, truncated to eliminate the cytoplasmic tyrosine 20 kinase domains but retaining a membrane anchor, virtually abolishes the growth of a transplantable glioblastoma in mice presumably by the dominant negative mechanism of heterodimer formation with membrane spanning endothelial cell VEGF receptors. Embryonic stem cells, which normally grow as solid tumors in nude mice, do not produce detectable tumors if both VEGF alleles are knocked out. Taken together, 25 these data indicate the role of VEGF in the growth of solid tumors. Inhibition of KDR or Flt-1 is implicated in pathological angiogenesis, and these receptors are useful in the treatment of diseases in which angiogenesis is part of the overall -4- WO 01/62252 PCT/USO1/05567 pathology, e.g., inflammation, diabetic retinal vascularization, as well as various forms of cancer since tumor growth is known to be dependent on angiogenesis. (Weidner et al., N. Engl. J. Med., 324, pp. 1-8, 1991). Accordingly, the identification of small compounds which specifically 5 inhibit, regulate and/or modulate the signal transduction of tyrosine kinases is desirable and is an object of this invention. SUMMARY OF THE INVENTION The present invention relates to compounds that are capable of 10 inhibiting, modulating and/or regulating signal transduction of both receptor-type and non-receptor type tyrosine kinases. One embodiment of the present invention is illustrated by a compound of Formula I, and the pharmaceutically acceptable salts and stereoisomers thereof: (R R R N O N O 15 DETAILED DESCRIPTION OF TEE INVENTION The compounds of this invention are useful in the inhibition of kinases and are illustrated by a compound of Formula I: 20 - 5- WO 01/62252 PCT/USO1/05567 R 5 )!R la|A (RR Q N O or a pharmaceutically acceptable salt or stereoisomer thereof, wherein 5 Q is S, 0, or -E=D- ; A is selected from the following:
R
4 a 0
OR
4 a RO4a "5R 4"R 4 R40 R 4 R4 N 4 a N' R 4 a and 0 R 10 ais Gorl; bis Oorl; -6- WO 01/62252 PCT/USO1/05567 s is 1 or 2; m is 0, 1, or 2; E=D is C=N, N=C, or C=C; 5 R1, Ria, R 4 and R 5 are independently selected from: 1) H, 2) (C=O)aObC1-C1O alkyl, optionally substituted with one to three substituents selected from R 6 , 10 3) (C=O)aObaryl, optionally substituted with one to three substituents selected from R 6 , 4) (C=O)aObC2-C10 alkenyl, optionally substituted with one to three substituents selected from R 6 , 5) (C=O)aObC2-C1O alkynyl, optionally substituted with one to three 15 substituents selected from R 6 , 6) SOmC1-C1O alkyl, optionally substituted with one to three substituents selected from R 6 , 7) SOmaryl, optionally substituted with one to three substituents selected from R 6 , 20 8) CO2H, 9) halo, 10) CN, 11) OH, 12) ObC1-C6 perfluoroalkyl, and 25 13) (C=0)aNR 7
R
8 ; -7- WO 01/62252 PCT/USO1/05567
R
2 and R 3 are independently selected from the group consisting of: 1) H, 2) (C=O)OaC1-C1O alkyl, 5 3) (C=O)Oaaryl, 4) C1-C10 alkyl, 5) SOmC1-C1O alkyl, 6) SOmaryl, 7) (C=O)aObC2-C1O alkenyl, 10 8) (C=O)aObC2-C1O alkynyl, and 9) aryl, said alkyl, aryl, alkenyl and alkynyl is optionally substituted with one to three substituents selected from R 6 ; 15 R4a is selected from the group consisting of: 1) (C=O)OaC1-CIO alkyl, 2) (C=O)Oaaryl, 3) C1-C10 alkyl, 4) SOmC1-C1O alkyl, 20 5) SOmaryl, 6) (C=O)aObC2-C1O alkenyl, 7) (C=O)aObC2-C1O alkynyl, and 8) aryl, -8- WO 01/62252 PCT/USO1/05567 said alkyl, aryl, alkenyl and alkynyl is optionally substituted with one to three substituents selected from R 6 ;
R
6 is: 5 1) H, 2) (C=O)aObCl-C6 alkyl, 3) (C=O)aObaryl, optionally substituted with one to three substituents selected from R6a, 4) C2-C1O alkenyl, 10 5) C2-C10 alkynyl, 6) heterocyclyl, optionally substituted with one to three substituents selected from R6a, 7) CO2H, 8) halo, 15 9) CN, 10) OH, 11) oxo, 12) ObC1-C6 perfluoroalkyl, or 13) NR 7
R
8 ; 20
R
6 a is: 1) H, 2) SOmaryl, 3) SOmC1-C6 alkyl, 25 4) (C=O)aObCl-C6 alkyl, 5) (C=O)aObaryl, -9- WO 01/62252 PCT/USO1/05567 6) C2-C10 alkenyl, 7) C2-C10 alkynyl, 8) heterocyclyl, 9) CO2H, 5 10) halo, 11) CN, 12) OH, 13) oxo, 14) ObC1-C6 perfluoroalkyl, or 10 15) N(C1-C6 alkyl)2;
R
7 and R 8 are independently selected from: 1) H, 2) (C=O)ObC1-C10 alkyl, optionally substituted with one to three 15 substituents selected from R6a, 3) (C=O)Obaryl, optionally substituted with one to three substituents selected from R6a, 4) C1-CI alkyl, optionally substituted with one to three substituents selected from R6a, 20 5) aryl, optionally substituted with one to three substituents selected from R6a, 6) C 2 -C1O alkenyl, optionally substituted with one to three substituents selected from R6a, 7) C2-C10 alkynyl, optionally substituted with one to three substituents 25 selected from R6a, and - 10 - WO 01/62252 PCT/USO1/05567 8) heterocyclyl, or
R
7 and R 8 can be taken together with the nitrogen to which they are attached to form a 5-7 membered heterocycle containing, in addition to the nitrogen, one or two 5 additional heteroatoms selected from N, 0 and S, said heterocycle optionally substituted with one to three substituents selected from R 6 a. A second embodiment is illustrated by the compound of Formula I, as described above, wherein Q is E=D. In a third embodiment, E=D is further defined as 10 C=C. A fourth embodiment of the invention is the compound of Formula I, as described above, wherein Q is E=D; E=D is C=C; RI, R1a, R 4 and R 5 are independently selected from: 15 1) H, 2) (C=O)aObCl-C6 alkyl, optionally substituted with one to three substituents selected from R 6 , 3) (C=O)aObaryl, optionally substituted with one to three substituents selected from R 6 , 20 4) (C=0)aObC2-C6 alkenyl, optionally substituted with one to three substituents selected from R 6 , 5) CO2H, 6) halo, 7) CN, 25 8) OH, 9) ObC1-C3 perfluoroalkyl, and - 11 - WO 01/62252 PCT/USO1/05567 10) (C=0)aNR 7
R
8 ;
R
2 and R 3 are independently selected from the group consisting of: 1) H, 5 2) (C=O)OaCi-C6 alkyl, and 3) C1-C6 alkyl; R4a is (C=O)OaC1-C6 alkyl or C1-C6 alkyl; 10 R 6 is: 1) H, 2) (C=O)aObCl-C6 alkyl, 3) (C=O)aObaryl, optionally substituted with one to three substituents selected from R6a, 15 4) C2-C6 alkenyl, 5) heterocyclyl, optionally substituted with one to three substituents selected from R6a, 6) CO2H, 7) halo, 20 8) CN, 9) OH, 10) oxo, 11) ObC1-C3 perfluoroalkyl, or 12) NR 7
R
8 ; 25
R
6 a is: - 12 - WO 01/62252 PCT/USO1/05567 1) H, 2) SOmaryl, 3) SOmC1-C6 alkyl, 4) (C=O)aObCl-C6 alkyl, 5 5) (C=O)aObaryl, 6) C 2 -C6 alkenyl, 7) heterocyclyl, 8) CO2H, 9) halo, 10 10) CN, 11) OH, 12) oxo, 13) ObC1-C3 perfluoroalkyl, or 14) N(C1-C6 alkyl)2; 15
R
7 and R 8 are independently selected from: 1) H, 2) (C=O)ObC1-C6 alkyl, optionally substituted with one to three substituents selected from R6a, 20 3) (C=O)Obaryl, optionally substituted with one to three substituents selected from R 6 a, 4) C1-C6 alkyl, optionally substituted with one to three substituents selected from R6a, 5) aryl, optionally substituted with one to three substituents selected from 25 R6a, - 13 - WO 01/62252 PCT/USO1/05567 6) C2-C6 alkenyl, optionally substituted with one to three substituents selected from R6a, and 7) heterocyclyl, or 5 R 7 and R 8 can be taken together with the nitrogen to which they are attached to form a 5-7 membered heterocycle containing, in addition to the nitrogen, one or two additional heteroatoms selected from N, 0 and S, said heterocycle optionally substituted with one to three substituents selected from R6a. 10 In a fifth embodiment of the invention, the compound of Formula I is defined such that Q is E=D; E=D is C=C; a is 0 or 1; b is 0 or 1; s is 1; 15 R1 and R 4 are independently selected from: 1) H, 2) (C=O)aObCl-C6 alkyl, optionally substituted with one to three substituents selected from R 6 , 20 3) (C=O)aObaryl, optionally substituted with one to three substituents selected from R 6 , 4) (C=O)aObC2-C6 alkenyl, optionally substituted with one to three substituents selected from R 6 , 5) (C=O)aObC2-C6 alkynyl, optionally substituted with one to three 25 substituents selected from R 6 , - 14 - WO 01/62252 PCT/USO1/05567 6) CO2H, 7) halo, 8) CN, 9) OH, 5 10) ObC1-C3 perfluoroalkyl, and 11) (C=0)aNR 7
R
8 ;
R
2 and R 3 are independently selected from H and methyl; 10 R 4 a is methyl;
R
5 and Ria are H;
R
6 is: 15 1) H, 2) (C=O)aObCl-C6 alkyl, 3) (C=O)aObaryl, optionally substituted with one to three substituents selected from R6a, 4) C2-C10 alkenyl, 20 5) C2-C10 alkynyl, 6) heterocyclyl, optionally substituted with one to three substituents selected from R6a, 7) CO2H, 8) halo, 25 9) CN, 10) OH, - 15 - WO 01/62252 PCT/USO1/05567 11) oxo, 12) ObC1-C3 perfluoroalkyl, or 13) NR 7
R
8 ; 5 R 6 a is: 1) H, 2) SOmaryl, 3) SOmC1-C6 alkyl, 4) (C=O)aObC1-C6 alkyl, 10 5) (C=O)aObaryl, 6) C2-C10 alkenyl, 7) C2-C10 alkynyl, 8) heterocyclyl, 9) CO2H, 15 10) halo, 11) CN, 12) OH, 13) oxo, 14) ObC1-C3 perfluoroalkyl, or 20 15) N(C1-C6 alkyl)2;
R
7 and R 8 are independently selected from: 1) H, 2) (C=O)ObC1-C6 alkyl, optionally substituted with one to three 25 substituents selected from R6a, - 16 - WO 01/62252 PCT/USO1/05567 3) (C=O)Obaryl, optionally substituted with one to three substituents selected from R6a, 4) C1-CI alkyl, optionally substituted with one to three substituents selected from R6a, 5 5) aryl, optionally substituted with one to three substituents selected from R6a, 6) C2-C6 alkenyl, optionally substituted with one to three substituents selected from R6a, 7) C2-C6 alkynyl, optionally substituted with one to three substituents 10 selected from R6a, and 8) heterocyclyl, or
R
7 and R 8 can be taken together with the nitrogen to which they are attached to form a piperidinyl, piperazinyl, morpholinyl or pyrrolidinyl group, optionally substituted 15 with one or two substituents selected from R6a. Yet another embodiment of the present invention is a compound which is 3-(4-methyl-5-oxo-4,5-dihydro-1H-pyrrolo[3,2-b]pyridin-2-yl)-1H-quinolin-2-one or a pharmaceutically acceptable salt or stereoisomer thereof. Also included within the scope of the present invention is a 20 pharmaceutical composition which is comprised of a compound of Formula I as described above and a pharmaceutically acceptable carrier. The present invention also encompasses a method of treating or preventing cancer in a mammal in need of such treatment which is comprised of administering to said mammal a therapeutically effective amount of a compound of Formula I. Preferred cancers for treatment are 25 selected from cancers of the brain, genitourinary tract, lymphatic system, stomach, larynx and lung. Another set of preferred forms of cancer are histiocytic lymphoma, - 17 - WO 01/62252 PCT/USO1/05567 lung adenocarcinoma, small cell lung cancers, pancreatic cancer, gioblastomas and breast carcinoma. Also included is a method of treating or preventing a disease in which angiogenesis is implicated, which is comprised of administering to a mammal in need 5 of such treatment a therapeutically effective amount of a compound of Formula I. Such a disease in which angiogenesis is implicated is ocular diseases such as retinal vascularization, diabetic retinopathy, age-related macular degeneration, and the like. Also included within the scope of the present invention is a method of treating or preventing inflammatory diseases which comprises administering to a 10 mammal in need of such treatment a therapeutically effective amount of a compound of Formula I. Examples of such inflammatory diseases are rheumatoid arthritis, psoriasis, contact dermatitis, delayed hypersensitivity reactions, and the like. Also included is a method of treating or preventing a tyrosine kinase dependent disease or condition in a mammal which comprises administering to a 15 mammalian patient in need of such treatment a therapeutically effective amount of a compound of Formula I. The therapeutic amount varies according to the specific disease and is discernable to the skilled artisan without undue experimentation. A method of treating or preventing retinal vascularization which is comprised of administering to a mammal in need of such treatment a therapeutically 20 effective amount of compound of Formula I is also encompassed by the present invention. Methods of treating or preventing ocular diseases, such as diabetic retinopathy and age-related macular degeneration, are also part of the invention. Also included within the scope of the present invention is a method of treating or preventing inflammatory diseases, such as rheumatoid arthritis, psoriasis, contact 25 dermatitis and delayed hypersensitivity reactions, as well as treatment or prevention of bone associated pathologies selected from osteosarcoma, osteoarthritis, and rickets. - 18 - WO 01/62252 PCT/USO1/05567 The invention also contemplates the use of the instantly claimed compounds in combination with a second compound selected from: 1) an estrogen receptor modulator, 2) an androgen receptor modulator, 5 3) retinoid receptor modulator, 4) a cytotoxic agent, 5) an antiproliferative agent, 6) a prenyl-protein transferase inhibitor, 7) an HMG-CoA reductase inhibitor, 10 8) an HIV protease inhibitor, 9) a reverse transcriptase inhibitor, and 10) another angiogenesis inhibitor. Preferred angiogenesis inhibitors are selected from the group 15 consisting of a tyrosine kinase inhibitor, an inhibitor of epidermal-derived growth factor, an inhibitor of fibroblast-derived growth factor, an inhibitor of platelet derived growth factor, an MMP (matrix metalloprotease) inhibitor, an integrin blocker, interferon-a, interleukin-12, pentosan polysulfate, a cyclooxygenase inhibitor, carboxyamidotriazole, combretastatin A-4, squalamine, 6-0-chloroacetyl-carbonyl) 20 fumagillol, thalidomide, angiostatin, troponin-1, and an antibody to VEGF. Preferred estrogen receptor modulators are tamoxifen and raloxifene. Also included in the scope of the claims is a method of treating cancer which comprises administering a therapeutically effective amount of a compound of Formula I in combination with radiation therapy and/or in combination with a 25 compound selected from: 1) an estrogen receptor modulator, 2) an androgen receptor modulator, - 19 - WO 01/62252 PCT/USO1/05567 3) retinoid receptor modulator, 4) a cytotoxic agent, 5) an antiproliferative agent, 6) a prenyl-protein transferase inhibitor, 5 7) an HMG-CoA reductase inhibitor, 8) an HIV protease inhibitor, 9) a reverse transcriptase inhibitor, and 10) another angiogenesis inhibitor. 10 And yet another embodiment of the invention is a method of treating cancer which comprises administering a therapeutically effective amount of a compound of Formula I in combination with paclitaxel or trastuzumab. Also within the scope of the invention is a method of reducing or preventing tissue damage following a cerebral ischemic event which comprises 15 administering a therapeutically effective amount of a compound of Formula I. Another embodiment of the invention is a method of treating or preventing cancer which comprises administering a therapeutically effective amount of a compound of Formula I in combination with a COX-2 inhibitor. These and other aspects of the invention will be apparent from the 20 teachings contained herein. "Tyrosine kinase-dependent diseases or conditions" refers to pathologic conditions that depend on the activity of one or more tyrosine kinases. Tyrosine kinases either directly or indirectly participate in the signal transduction pathways of a variety of cellular activities including proliferation, adhesion and 25 migration, and differentiation. Diseases associated with tyrosine kinase activities include the proliferation of tumor cells, the pathologic neovascularization that supports solid tumor growth, ocular neovascularization (diabetic retinopathy, age - 20 - WO 01/62252 PCT/USO1/05567 related macular degeneration, and the like) and inflammation (psoriasis,,rheumatoid arthritis, and the like). The compounds of the present invention may have asymmetric centers, chiral axes, and chiral planes (as described in: E.L. Eliel and S.H. Wilen, Stereo 5 chemistry of Carbon Compounds, John Wiley & Sons, New York, 1994, pages 1119 1190), and occur as racemates, racemic mixtures, and as individual diastereomers, with all possible isomers and mixtures thereof, including optical isomers, being included in the present invention. In addition, the compounds disclosed herein may exist as tautomers and both tautomeric forms are intended to be encompassed by the 10 scope of the invention, even though only one tautomeric structure is depicted. For example, any claim to compound A below is understood to include tautomeric structure B, and vice versa, as well as mixtures thereof. 1i Rla 1 Rla R R N O N OH H A B 15 When any variable (e.g. aryl, heterocycle, R 1 , R 2 etc.) occurs more than one time in any constituent, its definition on each occurrence is independent at every other occurrence. Also, combinations of substituents and variables are permissible only if such combinations result in stable compounds. Lines drawn into the ring systems from substituents (such as from R 1 , R 2 , R 3 , R 4 etc.) indicate that the 20 indicated bond may be attached to any of the substitutable ring carbon atoms. If the ring system is polycyclic, it is intended that the bond be attached to any of the suitable carbon atoms on the proximal ring only. For example, - 21 - WO 01/62252 PCT/USO1/05567 R5 (R 4) R N , R3 can be, inter alia, any of the following when: X and Z are C, 5 Y is N,
R
5 is CH3,
R
3 is H, and
(R
4 )t is as defined in the claims:
H
3 C OCH 3
H
3 C N N N N OH H
CH
3 H OH
H
3 O
CH
3
H
3 C H-C N N H3N CO2Et N OH 3 I H Br H 10 It is understood that substituents and substitution patterns on the compounds of the instant invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized - 22 - WO 01/62252 PCT/USO1/05567 by techniques known in the art, as well as those methods set forth below, from readily available starting materials. As used herein, "alkyl" is intended to include both branched, straight chain, and cyclic saturated aliphatic hydrocarbon groups having the specified number 5 of carbon atoms. For example, CI-Clo, as in "CI-Clo alkyl" is defined to include groups having 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 carbons in a linear, branched, or cyclic arrangement. For example, "C1-C1O alkyl" specifically includes methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, and so on, as well as cycloalkyls such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, tetrahydro 10 naphthalene, methylenecylohexyl, and so on. "Alkoxy" represents an alkyl group of indicated number of carbon atoms attached through an oxygen bridge. If no number of carbon atoms is specified, the term "alkenyl" refers to a non-aromatic hydrocarbon radical, straight, branched or cyclic, containing from 2 to 10 carbon atoms and at least one carbon to carbon double bond. Preferably one 15 carbon to carbon double bond is present, and up to 4 non-aromatic carbon-carbon double bonds may be present. Thus, "C2-C6 alkenyl" means an alkenyl radical having from 2 to 6 carbon atoms. Alkenyl groups include ethenyl, propenyl, butenyl and cyclohexenyl. As described above with respect to alkyl, the straight, branched or cyclic portion of the alkenyl group may contain double bonds and may be substituted 20 if a substituted alkenyl group is indicated. The term "alkynyl" refers to a hydrocarbon radical straight, branched or cyclic, containing from 2 to 10 carbon atoms and at least one carbon to carbon triple bond. Up to 3 carbon-carbon triple bonds may be present. Thus, "C2-C6 alkynyl" means an alkynyl radical having from 2 to 6 carbon atoms. Alkynyl groups 25 include ethynyl, propynyl and butynyl. As described above with respect to alkyl, the - 23 - WO 01/62252 PCT/USO1/05567 straight, branched or cyclic portion of the alkynyl group may contain triple bonds and may be substituted if a substituted alkynyl group is indicated. As used herein, "aryl" is intended to mean any stable monocyclic or bicyclic carbon ring of up to 7 atoms in each ring, wherein at least one ring is 5 aromatic. Examples of such aryl elements include phenyl, naphthyl, tetrahydro naphthyl, indanyl, biphenyl, phenanthryl, anthryl or acenaphthyl. In cases where the aryl substituent is bicyclic and one ring is non-aromatic, it is understood that attachment is via the aromatic ring. The term heteroaryl, as used herein, represents a stable monocyclic or 10 bicyclic ring of up to 7 atoms in each ring, wherein at least one ring is aromatic and contains from 1 to 4 heteroatoms selected from the group consisting of 0, N and S. Heteroaryl groups within the scope of this definition include but are not limited to: acridinyl, carbazolyl, cinnolinyl, quinoxalinyl, pyrrazolyl, indolyl, benzotriazolyl, furanyl, thienyl, benzothienyl, benzofuranyl, quinolinyl, isoquinolinyl, oxazolyl, 15 isoxazolyl, indolyl, pyrazinyl, pyridazinyl, pyridinyl, pyrimidinyl, pyrrolyl, tetrahydroquinoline. In cases where the heteroaryl substituent is bicyclic and one ring is non-aromatic or contains no heteroatoms, it is understood that attachment is via the aromatic ring or via the heteroatom containing ring, respectively. As appreciated by those of skill in the art, "halo" or "halogen" as used 20 herein is intended to include chloro, fluoro, bromo and iodo. The term "heterocycle" or "heterocyclyl" as used herein is intended to mean a 5- to 10-membered aromatic or nonaromatic heterocycle containing from 1 to 4 heteroatoms selected from the group consisting of 0, N and S, and includes bicyclic groups. "Heterocyclyl" therefore includes the above mentioned heteroaryls, as well as dihydro and tetrathydro analogs 25 thereof. Further examples of "heterocyclyl" include, but are not limited to the following: benzoimidazolyl, benzofuranyl, benzofurazanyl, benzopyrazolyl, benzotriazolyl, benzothiophenyl, benzoxazolyl, carbazolyl, carbolinyl, cinnolinyl, - 24- WO 01/62252 PCT/USO1/05567 furanyl, imidazolyl, indolinyl, indolyl, indolazinyl, indazolyl, isobenzofuranyl, isoindolyl, isoquinolyl, isothiazolyl, isoxazolyl, naphthpyridinyl, oxadiazolyl, oxazolyl, oxazoline, isoxazoline, oxetanyl, pyranyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridopyridinyl, pyridazinyl, pyridyl, pyrimidyl, pyrrolyl, quinazolinyl, quinolyl, 5 quinoxalinyl, tetrahydropyranyl, tetrazolyl, tetrazolopyridyl, thiadiazolyl, thiazolyl, thienyl, triazolyl, azetidinyl, 1,4-dioxanyl, hexahydroazepinyl, piperazinyl, piperidinyl, pyrrolidinyl, morpholinyl, thiomorpholinyl, dihydrobenzoimidazolyl, dihydrobenzofuranyl, dihydrobenzothiophenyl, dihydrobenzoxazolyl, dihydrofuranyl, dihydroimidazolyl, dihydroindolyl, dihydroisooxazolyl, dihydroisothiazolyl, 10 dihydrooxadiazolyl, dihydrooxazolyl, dihydropyrazinyl, dihydropyrazolyl, dihydropyridinyl, dihydropyrimidinyl, dihydropyrrolyl, dihydroquinolinyl, dihydrotetrazolyl, dihydrothiadiazolyl, dihydrothiazolyl, dihydrothienyl, dihydrotriazolyl, dihydroazetidinyl, methylenedioxybenzoyl, tetrahydrofuranyl, and tetrahydrothienyl, and N-oxides thereof. 15 The pharmaceutically acceptable salts of the compounds of this invention include the conventional non-toxic salts of the compounds of this invention as formed, e.g., from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the 20 salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxy-benzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, trifluoroacetic and the like. In certain instances, R 7 and R 8 are defined such that they can be 25 taken together with the nitrogen to which they are attached to form a 5-7 membered heterocycle containing, in addition to the nitrogen, one or two additional heteroatoms selected from N, 0 and S, said heterocycle optionally substituted with - 25 - WO 01/62252 PCT/USO1/05567 one to three substituents selected from R6a. Examples of the 5-7 membered ring systems that can thus be formed include, but are not limited to the following:
R
6 a
R
6 a Ra R6a N-N 6-N O N - Rea -N N N N OI 5 Ra R a
R
6 a
R
6 a I ' N 1 /-O -N -N ) -NN
R
6 a R 6 a R 6 a Preferably Q is E=D and E=D is C=C. Preferably RI is H, C1-C6 alkyl, or aryl. Most preferably RI is H or 10 C1-C6 alkyl. The preferred definition of Ria is H. Preferably R 2 and R 3 are independently H, C1-C6 alkyl, or (C=O)C1-C6 alkyl. Most preferably R 2 and R 3 are independently H or C1-C6 alkyl. Preferably R 4 is OH, OC1-C6 alkyl, C1-C6 alkyl. 15 Preferably R4a is C1-C6 alkyl. Preferably R 5 is H or C1-C6 alkyl. Most preferably R 5 is H. The pharmaceutically acceptable salts of the compounds of this invention can be synthesized from the compounds of this invention which contain a - 26 - WO 01/62252 PCT/USO1/05567 basic or acidic moiety by conventional chemical methods. Generally, the salts of the basic compounds are prepared either by ion exchange chromatography or by reacting the free base with stoichiometric amounts or with an excess of the desired salt forming inorganic or organic acid in a suitable solvent or various combinations of 5 solvents. Similarly, the salts of the acidic compounds are formed by reactions with the appropriate inorganic or organic base. The compounds of this invention may be prepared by employing reactions as shown in the following schemes, in addition to other standard manipulations that are known in the literature or exemplified in the experimental 10 procedures. These schemes, therefore, are not limited by the compounds listed nor by any particular substituents employed for illustrative purposes. Substituent numbering as shown in the schemes does not necessarily correlate to that used in the claims. Synopsis of Schemes 15 As shown in Scheme 1, the quinoline reagent 1-2 can be synthesized by the general procedures taught in Marsais, F; Godard, A.; Queguiner, G. J. Hetero cyclic Chem. 1989, 26, 1589-1594). Derivatives with varying substitution can be made by modifying this procedure and use of standard synthetic protocols known in the art. Intermediate 1-2 is then coupled with the appropriate N-protected pyrollo 20 compound, structure 1-4, to produce a chlorinated intermediate of structure 1-5. At least one of the R4 substituents would be OH or OR on the carbon adjacent to the ring nitrogen. Heating of 1-5 in aqueous acetic acid produces the desired de-chlorinated product, 1-6. Scheme 2 shows an example using this route to arrive at a [3,2] pyridno-pyrole, 2-3. 25 As shown in Scheme 3, the a-alkyloxy pyridino-pyroles 3-1 can be converted to the corresponding pyrimidinone analogs 3-2 by heating with aqueous HMr. Alternatively, the pyrimidinone analogs can be synthesized via the N-oxide - 27 - WO 01/62252 PCT/US01/05567 intermediates 4-2 as shown in Scheme 4. Scheme 5 shows the N-alkylation of the pyrimidinone-pyrole 3-2 to arrive at the compounds of Formula I. Other electrophilic reagents can be employed to alkylate or acylate the nitrogen, as will be apparent to the skilled artisan. 5 SCHEME 1 1 s la ) la
B(OH)
2 NIS I
X
N CI CH 3 CN N CI 1-1 1-2
RR
5 SBoc 2 0, Yx DMAP Z N CH 2
CI
2 N H / Boc (R 4)t 1-3H (R 4)t 1-4 o step 1: t-BuLi; Me 3 SnCI step 2: Intermediate 1-2, Pd(PPh 3
)
4 , Cul dioxane, reflux
R
5 X= (R 4 )t
R
5 X=Y (R4t (R )s Rla Z (RI)s Rla \ N AcOH Bo N O H 2 0, reflux N CI H 1-6 1-5 - 28 - WO 01/62252 PCT/USO1/05567 SCHEME 2 I Boc2O, N 2Nc N CH 2
CI
2 Bo (R 4)t H (R4)t 2-1 2-2 (R4)t -l=\ R) RlaN step 1: t-BuLi; (R), R N (MeO) 3 B IN I H step 2: Intermediate 1-2, N O Pd(PPh 3
)
4 , K 3
PO
4 , H dioxane, reflux 2-3 step 3: AcOH (aq), reflux 5 SCHEME 3 0 0 R N l HN '. N ~-. N H 48% HBr (aq) ( H N 0 reflIux N 0 3-1 H H 3-2 -29- WO 01/62252 PCT/USO1/05567 SCHEME 4 (N CN 0 NH 1 -la RR NmPBA N 1N TFAA X N HH IH N ~ N 2. NaHC 3 N H H 4-1 4-2 H 5 5 SCHEME 5 0 R 0 HN N 'N N IN (R) 1 NaO-t-Bu - N N H H N 0RI ~ N 0 H 3-2 H 5-1 10 UTILITY The instant compounds are useful as pharmaceutical agents for mammals, especially for humans, in the treatment of tyrosine kinase dependent 15 diseases. Such diseases include the proliferation of tumor cells, the pathologic neovascularization (or angiogenesis) that supports solid tumor growth, ocular neovascularization (diabetic retinopathy, age-related macular degeneration, and the like) and inflammation (psoriasis, rheumatoid arthritis, and the like). - 30 - WO 01/62252 PCT/USO1/05567 The compounds of the instant invention may be administered to patients for use in the treatment of cancer. The instant compounds inhibit tumor angiogenesis, thereby affecting the growth of tumors (J. Rak et al. Cancer Research, 55:4575-4580, 1995). The anti-angiogenesis properties of the instant compounds 5 are also useful in the treatment of certain forms of blindness related to retinal vascularization. The disclosed compounds are also useful in the treatment of certain bone-related pathologies, such as osteosarcoma, osteoarthritis, and rickets, also known as oncogenic osteomalacia. (Hasegawa et al., Skeletal Radiol., 28, pp.41-45, 10 1999; Gerber et al., Nature Medicine, Vol. 5, No. 6, pp.623-628, June 1999). And since VEGF directly promotes osteoclastic bone resorption through KDR/Flk-1 expressed in mature osteoclasts (FEBS Let. 473:161-164 (2000); Endocrinology, 141:1667 (2000)), the instant compounds are also useful to treat and prevent conditions related to bone resorption, such as osteoporosis and Paget's disease. 15 The claimed compounds can also be used to reduce or prevent tissue damage which occurs after cerebral ischemic events, such as stroke, by reducing cerebral edema, tissue damage, and reperfusion injury following ischemia. (Drug News Perspect 11:265-270 (1998); J. Clin. Invest. 104:1613-1620 (1999)). The compounds of this invention may be administered to mammals, 20 preferably humans, either alone or, preferably, in combination with pharmaceutically acceptable carriers or diluents, optionally with known adjuvants, such as alum, in a pharmaceutical composition, according to standard pharmaceutical practice. The compounds can be administered orally or parenterally, including the intravenous, intramuscular, intraperitoneal, subcutaneous, rectal and topical routes of 25 administration. For oral use of a chemotherapeutic compound according to this invention, the selected compound may be administered, for example, in the form - 31 - WO 01/62252 PCT/USO1/05567 of tablets or capsules, or as an aqueous solution or suspension. In the case of tablets for oral use, carriers which are commonly used include lactose and corn starch, and lubricating agents, such as magnesium stearate, are commonly added. For oral administration in capsule form, useful diluents include lactose and dried corn starch. 5 When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring agents may be added. For intramuscular, intraperitoneal, subcutaneous and intravenous use, sterile solutions of the active ingredient are usually prepared, and the pH of the solutions should be suitably adjusted and buffered. For 10 intravenous use, the total concentration of solutes should be controlled in order to render the preparation isotonic. The compounds of the instant invention may also be co-administered with other well known therapeutic agents that are. selected for their particular useful ness against the condition that is being treated. For example, in the case of bone 15 related disorders, combinations that would be useful include those with antiresorptive bisphosphonates, such as alendronate and risedronate; integrin blockers (defined further below), such as cv33 antagonists; conjugated estrogens used in hormone replacement therapy, such as PREMPRO@, PREMARIN@ and ENDOMETRION@; selective estrogen receptor modulators (SERMs), such as raloxifene, droloxifene, 20 CP-336,156 (Pfizer) and lasofoxifene; cathespin K inhibitors; and ATP proton pump inhibitors. The instant compounds are also useful in combination with known anti-cancer agents. Such known anti-cancer agents include the following: estrogen receptor modulators, androgen receptor modulators, retinoid receptor modulators, 25 cytotoxic agents, antiproliferative agents, prenyl-protein transferase inhibitors, HMG CoA reductase inhibitors, HIV protease inhibitors, reverse transcriptase inhibitors, and other angiogenesis inhibitors. The instant compounds are particularly useful - 32 - WO 01/62252 PCT/USO1/05567 when coadminsitered with radiation therapy. The synergistic effects of inhibiting VEGF in combination with radiation therapy have been described in the art. (see WO 00/61186). "Estrogen receptor modulators" refers to compounds which interfere 5 or inhibit the binding of estrogen to the receptor, regardless of mechanism. Examples of estrogen receptor modulators include, but are not limited to, tamoxifen, raloxifene, idoxifene, LY353381, LY117081 , toremifene, fulvestrant, 4-[7-(2,2-dimethyl-1 oxopropoxy-4-methyl-2-[4-[2-(1-piperidinyl)ethoxy]phenyl]-2H-1-benzopyran-3-yl] phenyl-2,2-dimethylpropanoate, 4,4'-dihydroxybenzophenone-2,4-dinitrophenyl 10 hydrazone, and SH646. "Androgen receptor modulators" refers to compounds which interfere or inhibit the binding of androgens to the receptor, regardless of mechanism. Examples of androgen receptor modulators include finasteride and other 5a-reductase inhibitors, nilutamide, flutamide, bicalutamide, liarozole, and abiraterone acetate. 15 "Retinoid receptor modulators" refers to compounds which interfere or inhibit the binding of retinoids to the receptor, regardless of mechanism. Examples of such retinoid receptor modulators include bexarotene, tretinoin, 13-cis-retinoic acid, 9-cis-retinoic acid, a-difluoromethylornithine, ]LX23-7553, trans-N-(4' hydroxyphenyl) retinamide, N-4-carboxyphenyl retinamide, 20 "Cytotoxic agents" refer to compounds which cause cell death primarily by interfering directly with the cell's functioning or inhibit or interfere with cell myosis, including alkylating agents, tumor necrosis factors, intercalators, microtubulin inhibitors, and topoisomerase inhibitors. Examples of cytotoxic agents include, but are not limited to, 25 tirapazimine, sertenef, cachectin, ifosfamide, tasonermin, lonidamine, carboplatin, altretamine, prednimustine, dibromodulcitol, ranimustine, fotemustine, nedaplatin, oxaliplatin, temozolomide, heptaplatin, estramustine, improsulfan tosilate, - 33 - WO 01/62252 PCT/USO1/05567 trofosfamide, nimustine, dibrospidium chloride, pumitepa, lobaplatin, satraplatin, profiromycin, cisplatin, irofulven, dexifosfamide, cis-aminedichloro(2-methyl pyridine) platinum, benzylguanine, glufosfamide, GPX100, (trans, trans, trans) bis-mu-(hexane-1,6-diamine)-mu-[diamine-platinum(Il)lbis[diamine(chloro)platinum 5 (II)]tetrachloride, diarizidinylspermine, arsenic trioxide, 1-(1 1-dodecylamino-10 hydroxyundecyl)-3,7-dimethylxanthine, zorubicin, idarubicin, daunorubicin, bisantrene, mitoxantrone, pirarubicin, pinafide, valrubicin, amrubicin, antineoplaston, 3'-deamino-3'-morpholino-13-deoxo-10-hydroxycarminomycin, annamycin, galarubicin, elinafide, MEN10755, and 4-demethoxy-3-deamino-3-aziridinyl-4 10 methylsulphonyl-daunorubicin (see WO 00/50032). Examples of microtubulin inhibitors include paclitaxel, vindesine sulfate, 3',4'-didehydro-4'-deoxy-8'-norvincaleukoblastine, docetaxol, rhizoxin, dolastatin, mivobulin isethionate, auristatin, cemadotin, RPR109881, BMS184476, vinflunine, cryptophycin, 2,3,4,5,6-pentafluoro-N-(3-fluoro-4-methoxyphenyl) 15 benzene sulfonamide, anhydrovinblastine, N,N-dimethyl-L-valyl-L-valyl-N-methyl L-valyl-L-prolyl-L-proline-t-butylamide, TDX25 8, and BMS 188797. Some examples of topoisomerase inhibitors are topotecan, hycaptamine, irinotecan, rubitecan, 6-ethoxypropionyl-3',4'-O-exo-benzylidene chartreusin, 9-methoxy-N,N-dimethyl-5-nitropyrazolo[3,4,5-kl]acridine-2 20 (6H)propanamine, 1-anino-9-ethyl-5-fluoro-2,3-dihydro-9-hydroxy-4-methyl 1H,12H-benzo[de]pyrano[3',4':b,7]indolizino[1,2b]quinoline-10,13(9H,15H) dione, lurtotecan, 7-[2-(N-isopropylamino)ethyl]-(20S)camptothecin, BNP1350, BNPI1 100, BN80915, BN80942, etoposide phosphate, teniposide, sobuzoxane, 2'-dimethylamino-2'-deoxy-etoposide, GL331, N-[2-(dimethylamino)ethyl]-9 25 hydroxy-5,6-dimethyl-6H-pyrido[4,3-b]carbazole-1-carboxamide, asulacrine, (5a, 5aB, 8aa,9b)-9- [2-[N-[2-(dimethylamino)ethyl]-N-methylamino]ethyl] -5- [4-hydroxy - 34 - WO 01/62252 PCT/USO1/05567 3,5-dimethoxyphenyl]-5,5a,6,8,8a,9-hexohydrofuro(3',4':6,7)naphtho(2,3-d)-1,3 dioxol-6-one, 2,3-(methylenedioxy)-5-methyl-7-hydroxy-8-methoxybenzo[c] phenanthridinium, 6,9-bis[(2-aminoethyl)amino]benzo[g]isoguinoline-5,10-dione, 5-(3-aminopropylamino)-7,10-dihydroxy-2-(2-hydroxyethylaminomethyl)-6H 5 pyrazolo[4,5,1-de]acridin-6-one, N-[1-[2(diethylamino)ethylamino]-7-methoxy-9 oxo-9H-thioxanthen-4-ylmethyl]formamide, N-(2-(dimethylamino)ethyl)acridine 4-carboxamide, 6-[[2-(dimethylamino)ethyl] amino]-3-hydroxy-7H-indeno [2,1 c]quinolin-7-one, and dimesna. "Antiproliferative agents" includes antisense RNA and DNA 10 oligonucleotides such as G3139, ODN698, RVASKRAS, GEM231, and INX3001, and antimetabolites such as enocitabine, carmofur, tegafur, pentostatin, doxifluridine, trimetrexate, fludarabine, capecitabine, galocitabine, cytarabine ocfosfate, fosteabine sodium hydrate, raltitrexed, paltitrexid, emitefur, tiazofurin, decitabine, nolatrexed, pemetrexed, nelzarabine, 2'-deoxy-2'-methylidenecytidine, 2'-fluoromethylene-2' 15 deoxycytidine, N-[5-(2,3-dihydro-benzofuryl)sulfonyl]-N'-(3,4-dichlorophenyl) urea, N6-[4-deoxy-4-[N2-[2(E),4(E)-tetradecadienoyl]glycylamino]-L-glycero B-L-manno-heptopyranosyl] adenine, aplidine, ecteinascidin, troxacitabine, 4-[2 amino-4-oxo-4,6,7,8-tetrahydro-3H-pyrimidino[5,4-b][1,4]thiazin-6-yl-(S)-ethyl] 2,5-thienoyl-L-glutamic acid, aminopterin, 5-flurouracil, alanosine, 11-acetyl-8 20 (carbamoyloxymethyl)-4-formyl-6-methoxy-14-oxa-1,11-diazatetracyclo(7.4.1.0.0) tetradeca-2,4,6-trien-9-yl acetic acid ester, swainsonine, lometrexol, dexrazoxane, methioninase, 2'-cyano-2'-deoxy-N4-palmitoyl-1-B-D-arabino furanosyl cytosine, and 3-aminopyridine-2-carboxaldehyde thiosemicarbazone. "Antiproliferative agents" also includes monoclonal antibodies to growth factors, other than those listed 25 under "angiogenesis inhibitors", such as trastuzumab, and tumor suppressor genes, such as p5 3 , which can be delivered via recombinant virus-mediated gene transfer (see U.S. Patent No. 6,069,134, for example). - 35 - WO 01/62252 PCT/USO1/05567 "HMG-CoA reductase inhibitors" refers to inhibitors of 3-hydroxy 3-methylglutaryl-CoA reductase. Compounds which have inhibitory activity for HMG-CoA reductase can be readily identified by using assays well-known in the art. For example, see the assays described or cited in U.S. Patent 4,231,938 at col. 5 6, and WO 84/02131 at pp. 30-33. The terms "HMG-CoA reductase inhibitor" and "inhibitor of HMG-CoA reductase" have the same meaning when used herein. Examples of HMG-CoA reductase inhibitors that may be used include but are not limited to lovastatin (MEVACOR®; see US Patent No. 4,231,938; 4,294,926; 4,319,039), simvastatin (ZOCOR@; see US Patent No. 4,444,784; 10 4,820,850; 4,916,239), pravastatin (PRAVACHOL®; see US Patent Nos. 4,346,227; 4,537,859; 4,410,629; 5,030,447 and 5,180,589), fluvastatin (LESCOL@; see US Patent Nos. 5,354,772; 4,911,165; 4,929,437; 5,189,164; 5,118,853; 5,290,946; 5,356,896), atorvastatin (LIPITOR@; see US Patent Nos. 5,273,995; 4,681,893; 5,489,691; 5,342,952) and cerivastatin (also known as rivastatin and BAYCHOL@; 15 see US Patent No. 5,177,080). The structural formulas of these and additional HMG CoA reductase inhibitors that may be used in the instant methods are described at page 87 of M. Yalpani, "Cholesterol Lowering Drugs", Chemistry & Industry, pp. 85 89 (5 February 1996) and US Patent Nos. 4,782,084 and 4,885,314. The term HMG CoA reductase inhibitor as used herein includes all pharmaceutically acceptable 20 lactone and open-acid forms (i.e., where the lactone ring is opened to form the free acid) as well as salt and ester forms of compounds which have HMG-CoA reductase inhibitory activity, and therefor the use of such salts, esters, open-acid and lactone forms is included within the scope of this invention. An illustration of the lactone portion and its corresponding open-acid form is shown below as structures I and II. 25 - 36 - WO 01/62252 PCT/USO1/05567 HO 0 HO COH Lactone Open-Acid I II In HiMG-CoA reductase inhibitors where an open-acid form can exist, salt and ester forms may preferably be formed from the open-acid, and all such forms 5 are included within the meaning of the term "HMG-CoA reductase inhibitor" as used herein. Preferably, the HMG-CoA reductase inhibitor is selected from lovastatin and simvastatin, and most preferably simvastatin. Herein, the term "pharmaceutically acceptable salts" with respect to the HMG-CoA reductase inhibitor shall mean non toxic salts of the compounds employed in this invention which are generally prepared 10 by reacting the free acid with a suitable organic or inorganic base, particularly those formed from cations such as sodium, potassium, aluminum, calcium, lithium, magnesium, zinc and tetramethylammonium, as well as those salts formed from amines such as ammonia, ethylenediamine, N-methylglucamine, lysine, arginine, ornithine, choline, N,N'-dibenzylethylenediamine, chloroprocaine, diethanolamine, 15 procaine, N-benzylphenethylamine, 1-p-chlorobenzyl-2-pyrrolidine-1'-yl-methylbenz imidazole, diethylamine, piperazine, and tris(hydroxymethyl) aminomethane. Further examples of salt forms of HMG-CoA reductase inhibitors may include, but are not limited to, acetate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate, bromide, calcium edetate, camsylate, carbonate, chloride, clavulanate, citrate, 20 dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrabamine, hydrobromide, - 37 - WO 01/62252 PCT/USO1/05567 hydrochloride, hydroxynapthoate, iodide, isothionate, lactate, lactobionate, laurate, malate, maleate, mandelate, mesylate, methylsulfate, mucate, napsylate, nitrate, oleate, oxalate, pamaote, palmitate, panthothenate, phosphate/diphosphate, polygalacturonate, salicylate, stearate, subacetate, succinate, tannate, tartrate, teoclate, 5 tosylate, triethiodide, and valerate. Ester derivatives of the described HMG-CoA reductase inhibitor compounds may act as prodrugs which, when absorbed into the bloodstream of a warm-blooded animal, may cleave in such a manner as to release the drug form and permit the drug to afford improved therapeutic efficacy. 10 "Prenyl-protein transferase inhibitor" refers to a compound which inhibits any one or any combination of the prenyl-protein transferase enzymes, including farnesyl-protein transferase (FPTase), geranylgeranyl-protein transferase type I (GGPTase-I), and geranylgeranyl-protein transferase type-Il (GGPTase-II, also called Rab GGPTase). Examples of prenyl-protein transferase inhibiting 15 compounds include ( )-6-[amino(4-chlorophenyl)(1-methyl-1IH-imidazol-5 yl)methyl]-4-(3-chlorophenyl)-1-methyl-2(1H)-quinolinone, (-)-6-[amino(4 chlorophenyl)(1-methyl-1H-imidazol-5-yl)methyl]-4-(3-chlorophenyl)-1-methyl 2(1H)-quinolinone, (+)-6-[amino(4-chlorophenyl)(1-methyl-1H-imidazol-5 yl)methyl]-4-(3-chlorophenyl)-1-methyl-2(1H)-quinolinone, 5(S)-n-butyl-1-(2,3 20 dimethylphenyl)-4-[1-(4-cyanobenzyl)-5-imidazolylmethyl]-2-piperazinone, (S)-1 (3-chlorophenyl) -4-[1-(4-cyanobenzyl)-5-imidazolylmethyl]-5-[2-(ethanesulfonyl) methyl)-2-piperazinone, 5(S)-n-Butyl-1-(2-methylphenyl)-4-[1-(4-cyanobenzyl)-5 imidazolylmethyl]-2-piperazinone, 1-(3-chlorophenyl) -4-[i-(4-cyanobenzyl)-2 methyl-5-imidazolylmethyl]-2-piperazinone, 1-(2,2-diphenylethyl)-3-[N-(1-(4 25 cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl]piperidine, 4-{5-[4-Hydroxymethyl 4-(4-chloropyridin-2-ylmethyl)-piperidine-1-ylmethyl]-2-methylimidazol-1-ylmethyl} - 38 - WO 01/62252 PCT/USO1/05567 benzonitrile, 4-{5-[4-hydroxymethyl-4-(3-chlorobenzyl)-piperidine-1-ylmethyl]-2 methylimidazol-1-ylmethyl}benzonitrile, 4-{3-[4-(2-oxo-2H-pyridin-1-yl)benzyl] 3H-imidazol-4-ylmethyl}benzonitrile, 4-{3-[4-(5-chloro-2-oxo-2H-[1,2']bipyridin-5' ylmethyl]-3H-imidazol-4-ylmethyl}benzonitrile, 4-{3-[4-(2-Oxo-2H-[1,2']bipyridin 5 5'-ylmethyl]-3H-imidazol-4-ylmethyl}benzonitrile, 4-[3-(2-Oxo-1-phenyl-1,2 dihydropyridin-4-ylmethyl)-3H-imidazol-4-ylmethyl}benzonitrile, 18,19-dihydro 19-oxo-5H,17H-6,10:12,16-dimetheno-1H-imidazo[4,3-c][1,11,4]dioxaazacyclo nonadecine-9-carbonitrile, (±)-19,20-Dihydro-19-oxo-5H-18,21-ethano-12,14-etheno 6,10-metheno-22H-benzo[d]imidazo[4,3-k][1,6,9,12]oxatriaza-cyclooctadecine-9 10 carbonitrile, 19,20-dihydro-19-oxo-5H,17H-18,21-ethano-6,10:12,16-dimetheno 22H-imidazo[3,4-h][1,8,11,14]oxatriazacycloeicosine-9-carbonitrile, and (±)-19,20 Dihydro-3-methyl-19-oxo-5H-18,21-ethano-12,14-etheno-6,10-metheno-22H benzo[d]imidazo[4,3-k][1,6,9,12]oxa-triazacyclooctadecine-9-carbonitrile. Other examples of prenyl-protein transferase inhibitors can be 15 found in the following publications and patents: WO 96/30343, WO 97/18813, WO 97/21701, WO 97/23478, WO 97/38665, WO 98/28980, WO 98/29119, WO 95/32987, U. S. Patent No. 5,420,245, U. S. Patent No. 5,523,430, U. S. Patent No. 5,532,359, U. S. Patent No. 5,510,510, U. S. Patent No. 5,589,485, U. S. Patent No. 5,602,098, European Patent Publ. 0 618 221, European Patent Publ. 0 675 112, 20 European Patent Publ. 0 604 181, European Patent Publ. 0 696 593, WO 94/19357, WO 95/08542, WO 95/11917, WO 95/12612, WO 95/12572, WO 95/10514, U.S. Patent No. 5,661,152, WO 95/10515, WO 95/10516, WO 95/24612, WO 95/34535, WO 95/25086, WO 96/05529, WO 96/06138, WO 96/06193, WO 96/16443, WO 96/21701, WO 96/21456, WO 96/22278, WO 96/24611, WO 96/24612, 25 WO 96/05168, WO 96/05169, WO 96/00736, U.S. Pat. No. 5,571,792, WO 96/17861, WO 96/33159, WO 96/34850, WO 96/34851, WO 96/30017, WO 96/30018, WO 96/30362, WO 96/30363, WO 96/31111, WO 96/31477, - 39 - WO 01/62252 PCT/USO1/05567 WO 96/31478, WO 96/31501, WO 97/00252, WO 97/03047, WO 97/03050, WO 97/04785, WO 97/02920, WO 97/17070, WO 97/23478, WO 97/26246, WO 97/30053, WO 97/44350, WO 98/02436, and U. S. Pat. No. 5,532,359. For an example of the role of a prenyl-protein transferase inhibitor on angiogenesis 5 see European J. of Cancer, Vol. 35, No. 9, pp.
13 9 4
-
14 0 1 (1999). Examples of HIV protease inhibitors include amprenavir, abacavir, CGP-73547, CGP-61755, DMP-450, indinavir, nelfinavir, tipranavir, ritonavir, saquinavir, ABT-378, AG 1776, and BMS-232,632. Examples of reverse transcriptase inhibitors include delaviridine, efavirenz, GS-840, HB Y097, 10 lamivudine, nevirapine, AZT, 3TC, ddC, and ddl. "Angiogenesis inhibitors" refers to compounds that inhibit the formation of new blood vessels, regardless of mechanism. Examples of angiogenesis inhibitors include, but are not limited to, tyrosine kinase inhibitors, such as inhibitors of the tyrosine kinase receptors Flt-1 (VEGFRI) and Flk-1/KDR (VEGFR20), 15 inhibitors of epidermal-derived, fibroblast-derived, or platelet derived growth factors, MMP (matrix metalloprotease) inhibitors, integrin blockers, interferon-a, interleukin 12, pentosan polysulfate, cyclooxygenase inhibitors, including nonsteroidal anti inflammatories (NSAIDs) like aspirin and ibuprofen as well as selective cyclooxygenase-2 inhibitors like celecoxib and rofecoxib (PNAS, Vol. 89, p. 7384 20 (1992); JNCI, Vol. 69, p. 475 (1982); Arch. Opthalmol., Vol. 108, p.
57 3 (1990); Anat. Rec., Vol. 238, p. 68 (1994); FEBS Letters, Vol. 372, p. 83 (1995); Clin, Orthop. Vol. 313, p. 76 (1995); J. Mol. Endocrinol., Vol. 16, p.
10 7 (1996); Jpn. J. Pharmacol., Vol. 75, p. 105 (1997); Cancer Res., Vol. 57, p. 1625 (1997); Cell, Vol. 93, p. 705 (1998); Intl. J. Mol. Med., Vol. 2, p. 715 (1998); J. Biol. Chem., Vol. 25 274, p. 9116 (1999)), carboxyamidotriazole, combretastatin A-4, squalamine, 6-0 chloroacetyl-carbonyl)-fumagillol, thalidomide, angiostatin, troponin-1, angiotensin II antagonists (see Fernandez et al., J. Lab. Clin. Med. 105:141-145 (1985)), and - 40 - WO 01/62252 PCT/USO1/05567 antibodies to VEGF. (see, Nature Biotechnology, Vol. 17, pp.
9 6 3
-
9 6 8 (October 1999); Kim et al., Nature, 362, 841-844 (1993); WO 00/44777; and WO 00/61186). As described above, the combinations with NSAID's are directed to the use of NSAID's which are potent COX-2 inhibiting agents. For purposes of this 5 specification an NSAID is potent if it possess an IC50 for the inhibition of COX-2 of 1[tM or less as measured by the cell or microsomal assay disclosed herein. The invention also encompasses combinations with NSAID's which are selective COX-2 inhibitors. For purposes of this specification NSAID's which are selective inhibitors of COX-2 are defined as those which possess a specificity for 10 inhibiting COX-2 over COX-1 of at least 100 fold as measured by the ratio of IC50 for COX-2 over IC50 for COX-1 evaluated by the cell or micromsal assay disclosed hereinunder. Such compounds include, but are not limited to those disclosed in U.S. 5,474,995, issued December 12, 1995, U.S. 5,861,419, issued January 19, 1999, U.S. 6,001,843, issued December 14, 1999, U.S. 6,020,343, issued February 1, 2000, 15 U.S. 5,409,944, issued April 25, 1995, U.S. 5,436,265, issued July 25, 1995, U.S. 5,536,752, issued July 16, 1996 , U.S. 5,550,142, issued August 27, 1996, U.S. 5,604,260, issued February 18, 1997, U.S. 5,698,584, issued December 16, 1997, U.S. 5,710,140, issued January 20,1998, WO 94/15932, published July 21, 1994, U.S. 5,344,991, issued June 6, 1994, U.S. 5,134,142, issued July 28, 1992, 20 U.S. 5,380,738, issued January 10, 1995, U.S. 5,393,790, issued February 20, 1995, U.S. 5,466,823, issued November 14, 1995, U.S. 5,633,272, issued May 27, 1997, and U.S. 5,932,598, issued August 3, 1999, all of which are hereby incorporated by reference. Other examples of specific inhibitors of COX-2 include the following: 25 3-(3-fluorophenyl)-4-(4-(methylsulfonyl)phenyl)-2-(5H)-furanone; 3-(3,4-difluorophenyl)-4-(4-(methylsulfonyl)phenyl)-2-(5H)-furanone; - 41 - WO 01/62252 PCT/USO1/05567 3-(3,4-dichlorophenyl)-4-(4-(methylsulfonyl)phenyl)-2-(5H)-furanone; 3-phenyl-4-(4-(methylsulfonyl)phenyl)-2-(5H)-furanone; 5,5-dimethyl-3-(3-fluorophenyl)-4-(methylsulfonyl)phenyl)-2-(5H)-furanone; 3-(4-methylsulfonyl)phenyl-2-phenyl-5-trifluoromethylpyridine; 5 2-(3-chlorophenyl)-3-(4-methylsulfonyl)phenyl-5-trifluoromethyl-pyridine; 2-(4-chlorophenyl)-3-(4-methylsulfonyl)phenyl-5-trifluoromethyl-pyridine; 2-(4-fluorophenyl)-3-(4-methylsulfonyl)phenyl-5-trifluoromethyl-pyridine; 3-(4-methylsulfonyl)phenyl-2-(3-pyridinyl)-5-trifluoromethylpyridine; 5-methyl-3-(4-methylsulfonyl)phenyl-2-phenylpyridine; 10 2-(4-chlorophenyl)-5-methyl-3-(4-methylsulfonyl) phenylpyridine; 5-methyl-3-(4-methylsulfonyl)phenyl-2-(3-pyridinyl) pyridine; 5-chloro-2-(4-chlorophenyl)-3-(4-methylsulfonyl) phenylpyridine; 5-chloro-3-(4-methylsulfonyl)phenyl-2-(2-pyridinyl) pyridine; 5-chloro-3-(4-methylsulfonyl)phenyl-2-(3-pyridinyl) pyridine; 15 5-chloro-3-(4-methylsulfonyl)phenyl-2-(4-pyridinyl) pyridine; 5-chloro-3-(4-methylsulfonyl)phenyl-2-(2-methyl-5-pyridinyl)pyridine; 2-(4-chlorophenyl)-3-(4-methylsulfonyl)phenylpyridinyl-5-carboxylic acid methyl ester; 2-(4-chlorophenyl)-3-(4-methylsulfonyl)phenylpyridinyl-5-carboxylic acid; 20 5-cyano-2-(4-chlorophenyl)-3-(4-methylsulfonyl) phenylpyridine; 5-chloro-3-(4-methylsulfonyl)phenyl-2-(3-pyridyl)pyridine hydromethanesulfonate; 5-chloro-3-(4-methylsulfonyl)phenyl-2-(3-pyridyl)pyridine hydrochloride; 5-chloro-3-(4-methylsulfonyl)phenyl-2-(2-methyl-5-pyridinyl)pyridine hydrochloride; 5-chloro-3-(4-methylsulfonyl)phenyl-2-(2-ethyl-5-pyridinyl)pyridine; 25 5-chloro-3-(4-methylsulfonyl)phenyl-2-(2-ethyl-5-pyridinyl)pyridine hydromethanesulfonate; 3-(3,4-difluorophenoxy)-5,5-dimethyl-4-(4-(methylsulfony)phenyl)-5H-furan-2-one; -42 - WO 01/62252 PCT/USO1/05567 3-(3-fluorophenoxy)-5,5-dimethy-4-(4-(methysufonly)phel-5I-fural-2-ole; 3-(3 ,5-difluorophenoxy)-5 ,5-dimethyl-4-(methylsulfonyl) phenyl)-5H-furan-2-one; 3-phenoxy-5 ,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-one; 3-(2,4-difluorophenoxy)-5 ,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-one; 5 3-(4-chlorophenoxy)-5 ,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-ofle; 3-(3 ,4-dichlorophenoxy)-5 ,5-dimethyl-4-(methylsulfonyl) phenyl)-5H-furan-2-one; 3-(4-fluorophenoxy)-5 ,5-dimethyl-4-(4-(methylsulfonyl) phenyl)-5H-furan-2-one; 3-(4-fluorophenythio)-5,5-dimethy-4-(4-(methysufofly)pheyl)-5H-fural-2-ole; 3-(3 ,5-difluorophenylthio)-5 ,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2 10 one; 3-phenylthio-5 ,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-ole; 3-(N-phenylamino)-5 ,5-dimethyl-4-(4-(methylsulfonyl) phenyl)-5H-furan-2-one; 3-(N-methyl-N-phenylamino)-5 ,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2 one; 15 3-cyclohexyloxy-5 ,5-.dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-one; 3-phenylthio-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-one; 3-benzyl-5 ,5-dimethy1-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-one; 3.-(3 ,4-difluorophenylhydroxymethyl)-5 ,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H furan-2-one; 20 3-(3 ,4-difluorobenzoyl)-5 ,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-one; 3-benzoyl-5 ,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-one; 4-(4-(methylsulfonyl)phenyl)-3-phenoxy-l1-oxaspiro[4.4]non-3-en- 2-one; 4-(4-(methylsulfonyl)phenyl)-3-phenylthio-l1-oxaspiroll4.4]non-3-en-2-one; 4-(2-oxo-3 -phenylthio-l1-oxa-spiro[4,4]non-3-en-4-yl) benzenesulfonamide; 25 3-(4-fluorobenzyl)-5 ,5-dimethyl-4-(4-(methylsulfonyl) phenyl)-5H-furan-2-one; 3-(3 ,4-difluorophenoxy)-5-methoxy-5-methyl-4-(4- (methylsulfonyl)phenyl)-5H furan-2-one; - 43 - WO 01/62252 PCT/USO1/05567 3-(5-chloro-2-pyridyloxy)-5 ,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2 one; 3-(2-pyridyloxy)-5 ,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-one; 3-(6-methyl-2-pyridyloxy)-5,5-dimethyl-4-(4-(methylsulfoflyl) phenyl)-5H-furan-2 5 one; 3-(3-isoquinolinoxy)-5 ,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-fural-2-ole; 3-(4-(methylsulfonyl)phenyl)-2-phenoxycyclopent-2-enone; 3-(4-(methylsulfonyl)phenyl)-2-(3 ,4-difluorophenoxy)cyclopent-2-enone; 5 ,5-dimethyl-4-(4-methylsulfonylphenyl)-3-(5-bromopyridin-2-yloxy)-5H-fural-2 10 one; 5 ,5-dimethyl-4-(4-methylsulfonylphenyl)-3-(2-propoxy)-5H-furan-2-one; 2-(3 ,4-difluorophenoxy)-3-(4-methylsulfonylphenyl)-cyclopent-2-enole; 3-(5-benzothiophenyloxy)-5 ,5-dimethyl-4-(4-(methylsulfonyl) phenyl)-5H-furan-2 one; 15 5 ,5-dimethyl-4-(4-methylsulfonyl-phenyl)-3-(pyridyl-4-oxy)-5H-furan-2-ofle; 5 ,5-dimethyl-4-(4-methylsulfonyl-phenyl)-3-(pyridyl-3-oxy)-5H-furan- 2 -ofle; 3-(2-methyl-5-pyridyloxy)-5 ,5-dimethyl-4-(4-(methylsulfonyl) phenyl)-5H-furan-2 one; 3-(2-fluoro-4-trifluoromethyl)phenoxy-4-(4-methylsulfonyl)phenyl)-5 ,5-dimethyl-5H 20 furan-2-one; 3-.(5-chloro-2-pyridylthio)-5 ,5-dimethyl-4-(4-methylsulfonyl)phenyl-5H-furan-2-one; 2-(3 ,5-difluorophenoxy)-3-(4-methylsulfonylphenyl)-cyclopent-2-enone; 3-(2-pyrimidinoxy)-5 ,5-dimethyl-4-(4-methylsulfonyl)phenyl-5H-furan-2-one; 3-(3-methyl-2-pyridyloxy)-5 ,5-dimethyl-4-(4-methylsulfonyl)phenyl-5H-furan-2-one; 25 3-(3-chloro-5-pyridyloxy)-5 ,5-dimethyl.-4-.(4-(methylsulfonyl) phenyl)-5H-furan-2 one; - 44- WO 01/62252 PCT/USO1/05567 3-(3-(1,2,5-thiadiazolyl)oxy)-4-(4-(methylsulfonyl)phenyl)-5,5-dimethyl-5H-furan-2 one; 3-(5-isoquinolinoxy)-5,5-dimethyl-4-(4-methylsulfonyl)phenyl-5H-furan-2-one; 3-(6-amino-2-pyridyloxy)-5,5-dimethyl-4-(4-(methylsulfonyl) phenyl)-5H-furan-2 5 one; 3-(3-chloro-4-fluoro)phenoxy-4-(methylsulfonyl)phenyl)-5,5-dimethyl-5H-furan-2 one; 3-(6-quinolinoxy)-5,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-one; 3-(5-nitro-2-pyridyloxy)-5,5-dimethyl-4-(4-methylsulfonyl)phenyl-5H-furan-2-one; 10 3-(2-thiazolylthio)-5,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-one; 3-(3-chloro-5-pyridyloxy)-5,5-dimethyl-4-(4-(methylsulfonyl) phenyl)-5H-furan-2 one; 5,5-dimethyl-4-(4-methylsulfonylphenyl)-3-(2-propoxy)-5H-furan-2-one; 3-(3-trifluoromethyl)phenoxy-4-(4-methylsulfonyl)phenyl)-5,5-dimethyl-5H-furan-2 15 one; 5,5-dimethyl-(4-(4-methylsulfonyl)phenyl)-3-(piperidine-1-carbonyl)-5-H-furan-2 one; 5,5-dimethyl-3-(2-Butoxy)-4-(4-methylsulfonylphenyl)-5H-furan-2-one; 5,5-dimethyl-4-(4-methylsulfonylphenyl)-3-(3-pentoxy)-5H-furan-2-one; 20 2-(5-chloro-2-pyridyloxy)-3-(4-methylsulfonyl)phenylcyclopent-2-enone; 3-(4-methyl-2-pyridyloxy)-5,5-dimethyl-4-(4-methylsulfonyl)phenyl-5H-furan-2-one; (5R)-3-(3,4-difluorophenoxy)-5-ethyl-5-methyl-4-(4-methylsulfonyl)phenyl-5H-furan 2-one; (5R)-3-(4-chlorophenoxy)-5-ethyl-5-methyl-4-(4-methylsulfonyl)pheny-5H-furan-2 25 one; 3-(2-methyl-3-pyridyloxy)-5,5-dimethyl-4-(4-methylsulfonyl)phenyl-5H-furan-2-one; -45- WO 01/62252 PCT/USO1/05567 3-(4-methyl-5-nitro-2-pyridyloxy)-5 ,5-dimethyl-4-(4-methylsulfonyl)phenyl-5H furan-2-one; 3-(5-chloro-4-methyl-2-pyrtidyloxy)-5 ,5-dimethyl-4-(4-methylsulfonyl)phenyl-5H furan-2-one; 5 3-(5-fluoro-4-methyl-2-pyridyloxy)-5 ,5-dimethyl-4-(4-methylsulfonyl)phenyl-5H furan-2-one; 3-(3-chloro-2-pyridyloxy)-5,5-dimethyl-4-(4-methylsulfonyl)phenyl-5H-fural-2-ole; 3-(4-fluorophenoxy)-5-methyl-4-(4-methylsulfonyl)phenyl-5-propyl-51-furan-2-one; 3-(N,N-diethylamino)-5 ,5-dimethyl-4-(4-(methylsulfonyl) phenyl)-5H-furan-2-one; 10 5 ,5-dimethyl-4-(4-methylsulfonyl-phenyl)-3-(3 ,5-dichloro-2-pyridyloxy)--5H-furan-2 one; (5R)-3-(4-bromophenoxy)-5-ethyl-5-methyl-4-(4-methylsulfonyl)phelyl-5H-fural-2 one; (5R)-3-(4-methoxyphenoxy)-5-ethyl-5-methyl-4-(4-methylsulfonyl)phelyl-5H-fural 15 2-one; (5R)-3-(5-chloro-2-pyridyloxy)-5-methyl-4-(4-methylsulfonyl)phenyl-5-(2,2,2 trifluoroethyl)-511-furan-2-one; 3-(5-chloro-2-pyridyloxy)--5-methyl-4-(4-methylsulfonyl)phenyl-5-propyl-5H-furan-2 one; 20 3-( 1-cyclopropyl-ethoxy)-5 ,5-dimethyl-4-(4-methyl sulfonyl)phenyl)-5H-furan-2-one; 5-methyl-4-(4-(methylsulfonyl)phenyl)-3-(2-(propoxy)-5-(2-trifluoroethyl)-5H-furan 2-one; 5(R)-5-ethyl-5-methyl-4-(4-(methylsulfonyl)phenyl)-3-(2-propoxy)-5H-furan-2-one; 5,5-dimethyl-3-(2,2-dimethylpropyloxy)-4-(4-(methylsulfonyl)phenyl)-5H-furan-2 25 one; 5(R)-3-( 1-cyclopropyl-ethoxy)-5-ethyl-5-methyl-4-(4-(methyl sulfonyl)phenyl-5H furan-2-one; -46 - WO 01/62252 PCT/USO1/05567 5(S)-5-ethyl-5-methyl-4-(4-(methylsulfonyl)phenyl-3-(2-propoxy)-51-furan-2-ofle; 3-(1 -cyclopropylethoxy)-5 ,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-one; 3-( 1-cyclopropylethoxy)-5 ,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-one; 5,5-dimethyl-3-(isobutoxy)-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-one; 5 3-(4-bromophenoxy)-5 ,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-one; 3-(2-quinolinoxy)-5 ,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-one; 3-(2-chloro-5-pyridyloxy)-5 ,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2 one; 3-(6-benzothiazolyloxy)-5,5-dimethyl-4-(4-(methylsulfonyl) phenyl)-5H-furan-2-one; 10 3-(6-chloro--2-pyridyloxy)-5 ,5-dimethyl-4-(4-(methylsulfonyl) phenyl)-5H-furan-2 one; 3-(4-quinazolyloxy)-5 ,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-one; (5R)-3-(5-fluoro-2-pyridyloxy)-5-ethyl-5-methyl-4-(4-methylsulfonyl)phenyl-5H furan-2-one; 15 (5R)-3-(4-fluorophenoxy)-5-ethyl-5-methyl-4-(4-methylsulfonyl)phenyl-5H-furan-2 one; (5R)-3-(5-fluoro-2-pyridyloxy)-5-methyl-4-(4-methylsulfonyl)phenyl-5-(2,2,2 trifluoroethyl)-5H-furan-2-one; 3-(1 -isoquinolinyloxy)-5 ,5-dimethyl-4-(methylsulfonyl)phenyl-5H-furan-2-one; 20 (5R)-3-(4-fluorophenoxy)-5-methyl-4-(4-methylsulfonyl)phenyl-5-(2,2,2 trifluoroethyl)-5H-furan-2-one; 3-(3-fluoro-2-pyrtidyloxy)-5 ,5-dimethyl-4-(4-methylsulfonyl) phenyl-5H-furan-2-one; (5R)-3-(3 ,4-difluorophenoxy)-5-methyl-4-(4-methylsulfonyl) phenyl-5-(2,2,2 trifluoroethyl)-5H-furan-2-one; 25 (5R)-3-(5-chloro-2-pyridyloxy)-5-ethyl-5-methyl-4-(4-methylsulfonyl)phenyl-5H furan-2-one; - 47 - WO 01/62252 PCT/USO1/05567 3-(3 ,4-difluorophenoxy)--5-methyl-5-trifluoromethyl-4-(4-methylsulfolyl)phelyl-5H furan-2-one; 3-.(3 ,4-difluorophenoxy)-5-methy-4-(4-(methysufonyl)phenyl)-5-propyl-5H-furafl-2 one; 5 3-cyclobutyloxy-5 ,5-dimethyl-4-(4-methylsulfonylphenyl-5H-furan-2-one; 3-(1-indanyloxy)-5 ,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-one; 3-(2-indanyloxy)-5 ,5-dimethyl-4-(4-methylsulfonyl)phenyl)-5H-furan-2-one; 3-cyclopentyloxy-5 ,5-dimethyl-4-(4-methylsulfonylphenyl)5H-furan-2-one; 3-(3 ,3-dimethylcyclopentyloxy)-5 ,5-dimethyl-4-(4-methylsulfonyl-phenyl)-5H-furan 10 2-one; 3-isopropoxy-5-methy-4-(4-methysufonylpheny)-5-propy-5H-furan-2-one; 3-(2-methoxy-5-pyridyloxy)-5 ,5-dimethyl-4-(4-methylsulfonyl)phenyl-5H-furan-2 one; 3-(5-methyl-2-pyridyloxy)-5 ,5-dimethyl.-4-(4-methylsulfonyl)phenyl-5H-furan-2-one; 15 (5RS)-3-(3 ,4-difluorophenoxy)-5-methyl-4-(4-methylsulfonyl)phenyl-5-( 2
,
2
,
2 trifluoroethyl)-5H-furan-2-one; 3-(3-chloro-4-methoxyphenoxy)-5 ,5-dimethyl-4-(4-methylsulfonyl)phenyl-5H-furan 2-one; (5R)-3-(3-chloro-4-methoxyphenoxy)-5-ethyl-5-methyl-4-(4-methylsulfonyl)phenyl 20 5H-furan-2-one; (5R)-3-(4-chlorophenoxy)-5-trifluoroethyl-5-methyl-4-(4-methylsulfonyl)phenyl-5H furan-2-one; (5R)-3-(4-bromophenoxy)-5-trifluoroethyl-5-methyl-4-(4-methylsulfonyl)phenyl-5H furan-2-one; 25 5-cyclopropylmethyl-3-(3 ,4-difluorophenoxy)-5-methyl-(4-methylsulfonyl)phenyl 5H-furan-2-one; - 48 - WO 01/62252 PCT/USO1/05567 (5R)-3-(3-fluorophenoxy)-5-ethyl-5-methyl-4-(4-methylsulfonyl)phenyl-5H-furan-2 one; (5R)-3-(4-chloro-3-fluorophenoxy)-5-ethyl-5-methyl-4-(4-methylsulfonyl)phenyl-5H furan-2-one; 5 (5R)-3-phenoxy-5-ethyl-5-methyl-4-(4-methylsulfonyl)phenyl-5H-furan-2-one; (5R)-3-(4-chloro-3-methylphenoxy)-5-ethyl-5-methyl-4-(4-methylsulfonyl)phenyl 5H-furan-2-one; 3-(4-chloro-3-methylphenoxy)-5-5-dimethyl-4-(4-methylsulfonyl)phenyl-5H-furan-2 one; 10 (5R)-3-(5-bromo-2-pyridyloxy)-4-(4-methylsulfonylphenyl)-5-methyl-5-(2,2,2 trifluoroethyl)-5H-furan-2-one; (5R)-3-(5-bromo-2-pyridyloxy)-4-(4-methylsulfonylphenyl)-5-ethyl-5-methyl-5H furan-2-one; 3-(5-chloro-6-methyl-2-pyridyloxy)-5,5-dimethyl-4-(4-methylsulfonyl)phenyl-5H 15 furan-2-one; 3-(5-cyclopropyl-2-pyridyloxy)-5,5-dimethyl-4-(4-methylsulfony)phenyl-5H-furan-2 one; 3-(1-cyclopropylethoxy)-4-(4-methylsulfonyl)phenyl-5H-furan-2-one; and 3-(cyclopropylmethoxy)-4-(4-methylsulfonyl)phenyl-5H-furan-2-one; 20 or a pharmaceutically acceptable salt or stereoisomer thereof. Inhibitors of COX-2 that are particularly useful in the instant method of treatment are: 3-phenyl-4-(4-(methylsulfonyl)phenyl)-2-(5H)-furanone; and 25 -49 - WO 01/62252 PCT/USO1/05567 S0 2 CH 3 0I o/ 5-chloro-3-(4-methylsulfonyl)phenyl-2-(2-methyl-5-pyridinyl)pyridine; S0 2 CH 3 CI N 5 N CH 3 or a pharmaceutically acceptable salt thereof. General and specific synthetic procedures for the preparation of the COX-2 inhibitor compounds described above are found in U.S. Patent No. 5,474,995, 10 issued December 12, 1995, U.S. 5,861,419, issued January 19, 1999, and U.S. Patent No. 6,001,843, issued December 14, 1999, all of which are herein incorporated by reference. Compounds that have been described as specific inhibitors of COX-2 and are therefore useful in the present invention include, but are not limited to, the 15 following: -50- WO 01/62252 PCT/USO1/05567
H
2 N N
CF
3
H
3 C
H
3 C O'N
H
2 N-S IO 0
H
3 C O N
H
Et N S 5 O O or a pharmaceutically acceptable salt thereof. Compounds which are described as specific inhibitors of COX-2 and are therefore useful in the present invention, and methods of synthesis thereof, can - 51 - WO 01/62252 PCT/USO1/05567 be found in the following patents, pending applications and publications, which are herein incorporated by reference: WO 94/15932, published July 21, 1994, U.S. Patent No. 5,344,991, issued June 6, 1994, U.S. Patent No. 5,134,142, issued July 28, 1992, U.S. Patent No. 5,380,738, issued January 10, 1995, U.S. Patent No. 5,393,790, 5 issued February 20, 1995, U.S. Patent No. 5,466,823, issued November 14, 1995, U.S. Patent No. 5,633,272, issued May 27, 1997, and U.S. Patent No. 5,932,598, issued August 3, 1999. Compounds which are specific inhibitors of COX-2 and are therefore useful in the present invention, and methods of synthesis thereof, can be found 10 in the following patents, pending applications and publications, which are herein incorporated by reference: U.S. Patent 5,474,995, issued December 12, 1995 , U.S. Patent 5,861,419, issued January 19, 1999, U.S. Patent 6,001,843, issued December 14, 1999, U.S. Patent 6,020,343, issued February 1, 2000, U.S. Patent 5,409,944, issued April 25, 1995 , U.S. Patent 5,436,265, issued July 25, 1995 , U.S. 15 Patent 5,536,752, issued July 16, 1996 , U.S. Patent 5,550,142, issued August 27, 1996, U.S. Patent 5,604,260, issued February 18, 1997, U.S. Patent 5,698,584, issued December 16, 1997, and U.S. Patent 5,710,140, issued January 20,1998. Other examples of angiogenesis inhibitors include, but are not limited to, endostation, ukrain, ranpirnase, IM862, 5-methoxy-4-[2-methyl-3-(3-methyl-2 20 butenyl)oxiranyl]-1-oxaspiro[2,5]oct-6-yl(chloroacetyl)carbamate, acetyldinanaline, 5-amino-1-[[3,5-dichloro-4-(4-chlorobenzoyl)phenyl]methyl]-lH-1,2,3-triazole-4 carboxamide,CM101, squalamine, combretastatin, RP14610, NX31838, sulfated mannopentaose phosphate, 7,7-(carbonyl-bis[imino-N-methyl-4,2-pyrrolocarbonyl imino[N-methyl-4,2-pyrrole]-carbonylimino]-bis-(1,3-naphthalene disulfonate), and 25 3-[(2,4-dimethylpyrrol-5-yl)methylene]-2-indolinone (SU5416). As used above, "integrin blockers" refers to compounds which selectively antagonize, inhibit or counteract binding of a physiological ligand to - 52 - WO 01/62252 PCT/USO1/05567 the avI3 integrin, to compounds which selectively antagonize, inhibit or counter act binding of a physiological ligand to the av@5 integrin, to compounds which antagonize, inhibit or counteract binding of a physiological ligand to both the cavP3 integrin and the av5 integrin, and to compounds which antagonize, inhibit or 5 counteract the activity of the particular integrin(s) expressed on capillary endothelial cells. The term also refers to antagonists of the av6, Uv08, aI01, a201, U501, a61 and a64 integrins. The term also refers to antagonists of any combination of av3, 6v05, v06, av@8, U1, a201, a511, a6pl and a64 integrins. Some specific examples of tyrosine kinase inhibitors include N 10 (trifluoromethylphenyl)-5-methylisoxazol-4-carboxamide, 3-[(2,4-dimethylpyrrol-5 yl)methylidenyl)indolin-2-one, 17-(allylamino)-17-demethoxygeldanamycin, 4-(3 chloro-4-fluorophenylamino)-7-methoxy-6-[3-(4-morpholinyl)propoxyl]quinazoline, N-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)-4-quinazolinamine, BIBX1382, 2,3,9,10,11,12-hexahydro-10-(hydroxymethyl)-10-hydroxy-9-methyl-9,12-epoxy 15 1H-diindolol[1,2,3-fg:3',2', 1'-kl]pyrrolo [3,4-i] [1,6]benzodiazocin-1-one, SH268, genistein, ST157 1, CEP2563, 4-(3-chlorophenylamino)-5,6-dimethyl-7H-pyrrolo [2,3-d]pyrimidinemethane sulfonate, 4-(3-bromo-4-hydroxyphenyl)amino-6,7 dimethoxyquinazoline, 4-(4'-hydroxyphenyl)amino-6,7-dimethoxyquinazoline, SU6668, ST1571A, N-4-chlorophenyl-4-(4-pyridylmethyl)-1-phthalazinamine, and 20 EMD121974. The instant compounds are also useful, alone or in combination with platelet fibrinogen receptor (GP fIb/IfIa) antagonists, such as tirofiban, to inhibit metastasis of cancerous cells. Tumor cells can activate platelets largely via thrombin generation. This activation is associated with the release of VEGF. The release of 25 VEGF enhances metastasis by increasing extravasation at points of adhesion to vascular endothelium (Amirkhosravi, Platelets 10, 285-292, 1999). Therefore, - 53 - WO 01/62252 PCT/USO1/05567 the present compounds can serve to inhibit metastasis, alone or in combination with GP Ib/Ia) antagonists. Examples of other fibrinogen receptor antagonists include abciximab, eptifibatide, sibrafiban, lamifiban, lotrafiban, cromofiban, and CT50352. If formulated as a fixed dose, such combination products employ the 5 compounds of this invention within the dosage range described below and the other pharmaceutically active agent(s) within its approved dosage range. Compounds of the instant invention may alternatively be used sequentially with known pharmaceutically acceptable agent(s) when a combination formulation is inappropriate. The term "administration" and variants thereof (e.g., "administering" 10 a compound) in reference to a compound of the invention means introducing the compound or a prodrug of the compound into the system of the animal in need of treatment. When a compound of the invention or prodrug thereof is provided in combination with one or more other active agents (e.g., a cytotoxic agent, etc.), "administration" and its variants are each understood to include concurrent and 15 sequential introduction of the compound or prodrug thereof and other agents. As used herein, the term "composition" is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts. 20 The term "therapeutically effective amount" as used herein means that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue, system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician. The term "treating cancer" or "treatment of cancer" refers to 25 administration to a mammal afflicted with a cancerous condition and refers to an effect that alleviates the cancerous condition by killing the cancerous cells, but also to an effect that results in the inhibition of growth and/or metastasis of the cancer. - 54- WO 01/62252 PCT/USO1/05567 The present invention also encompasses a pharmaceutical composition useful in the treatment of cancer, comprising the administration of a therapeutically effective amount of the compounds of this invention, with or without pharmaceutic ally acceptable carriers or diluents. Suitable compositions of this invention include 5 aqueous solutions comprising compounds of this invention and pharmacologically acceptable carriers, e.g., saline, at a pH level, e.g., 7.4. The solutions may be introduced into a patient's bloodstream by local bolus injection. When a compound according to this invention is administered into a human subject, the daily dosage will normally be determined by the prescribing 10 physician with the dosage generally varying according to the age, weight, and response of the individual patient, as well as the severity of the patient's symptoms. In one exemplary application, a suitable amount of compound is administered to a mammal undergoing treatment for cancer. Administration occurs in an amount between about 0.1 mg/kg of body weight to about 60 mg/kg of body 15 weight per day, preferably of between 0.5 mg/kg of body weight to about 40 mg/kg of body weight per day. ASSAYS 20 The compounds of the instant invention described in the Examples were tested by the assays described below and were found to have kinase inhibitory activity. Other assays are known in the literature and could be readily performed by those of skill in the art. (see, for example, Dhanabal et al., Cancer Res. 59:189-197; Xin et al., J. Biol. Chein. 274:9116-9121; Sheu et al., Anticancer Res. 18:4435-4441; 25 Ausprunk et al., Dev. Biol. 38:237-248; Gimbrone et al., J. Nati. Cancer Inst. 52:413 427; Nicosia et al., In Vitro 18:538-549). - 55 - WO 01/62252 PCT/USO1/05567 VEGF RECEPTOR KINASE ASSAY VEGF receptor kinase activity is measured by incorporation of radio-labeled phosphate into polyglutamic acid, tyrosine, 4:1 (pEY) substrate. The 5 phosphorylated pEY product is trapped onto a filter membrane and the incorporation of radio-labeled phosphate quantified by scintillation counting. MATERIALS 10 VEGF receptor kinase The intracellular tyrosine kinase domains of human KDR (Terman, B.I. et al. Oncogene (1991) vol. 6, pp. 1677-1683.) and Flt-1 (Shibuya, M. et al. Oncogene (1990) vol. 5, pp. 519-524) were cloned as glutathione S-transferase (GST) gene fusion proteins. This was accomplished by cloning the cytoplasmic 15 domain of the KDR kinase as an in frame fusion at the carboxy terminus of the GST gene. Soluble recombinant GST-kinase domain fusion proteins were expressed in Spodoptera frugiperda (Sf21) insect cells (Invitrogen) using a baculovirus expression vector (pAcG2T, Pharmingen). 20 Lysis buffer 50 mM Tris pH 7.4, 0.5 M NaCl, 5 mM DTT, 1 mM EDTA, 0.5% triton X-100, 10% glycerol, 10 mg/mL of each leupeptin, pepstatin and aprotinin and 1mM phenylmethylsulfonyl fluoride (all Sigma). 25 Wash buffer - 56 - WO 01/62252 PCT/USO1/05567 50 mM Tris pH 7.4, 0.5 M NaCl, 5 mM DTT, 1 mM EDTA, 0.05% triton X-100, 10 % glycerol, 10 mg/mL of each leupeptin, pepstatin and aprotinin and 1mM phenylmethylsulfonyl fluoride. 5 Dialysis buffer 50 mM Tris pH 7.4, 0.5 M NaCl, 5 mM DTT, 1 mM EDTA, 0.05% triton X-100, 50% glycerol, 10 mg/mL of each leupeptin, pepstatin and aprotinin and 1mM phenylmethylsuflonyl fluoride. 10 10 X reaction buffer 200 mM Tris, pH 7.4, 1.0 M NaCl, 50 mM MnCl2, 10 mM DTT and 5 mg/mL bovine serum albumin (Sigma). Enzyme dilution buffer 15 50 mM Tris, pH 7.4, 0.1 M NaCl, 1 mM DTT, 10 % glycerol, 100 mg/mL BSA. 10 X Substrate 750 sg/mL poly (glutamic acid, tyrosine; 4:1) (Sigma). 20 Stop solution 30% trichloroacetic acid, 0.2 M sodium pyrophosphate (both Fisher). Wash solution 25 15% trichloroacetic acid, 0.2 M sodium pyrophosphate. Filter plates - 57 - WO 01/62252 PCT/USO1/05567 Millipore #MAFC NOB, GF/C glass fiber 96 well plate. METHOD 5 A. Protein purification 1. Sf21 cells were infected with recombinant virus at a multiplicity of infection of 5 virus particles/ cell and grown at 27'C for 48 hours. 2. All steps were performed at 4"C. Infected cells were harvested by centrifugation at 1000 X g and lysed at 4'C for 30 minutes with 1/10 volume of lysis 10 buffer followed by centrifugation at 100,00OXg for 1 hour. The supernatant was then passed over a glutathione Sepharose column (Pharmacia) equilibrated in lysis buffer and washed with 5 volumes of the same buffer followed by 5 volumes of wash buffer. Recombinant GST-KDR protein was eluted with wash buffer/10 mM reduced glutathione (Sigma) and dialyzed against dialysis buffer. 15 B. VEGF receptor kinase assay 1. Add 5 pl of inhibitor or control to the assay in 50% DMSO. 2. Add 35 sl of reaction mix containing 5 gl of 10 X reaction buffer, 5 pl 25 mM ATP/10 Ci [3 3 P]ATP (Amersham), and 5 pl 10 X substrate. 20 3. Start the reaction by the addition of 10 pl of KDR (25 nM) in enzyme dilution buffer. 4. Vix and incubate at room temperature for 15 minutes. 5. Stop by the addition of 50 pl stop solution. 6. Incubate for 15 minutes at 4*C. 25 7. Transfer a 90 pl aliquot to filter plate. 8. Aspirate and wash 3 times with wash solution. - 58 - WO 01/62252 PCT/USO1/05567 9. Add 30 d of scintillation cocktail, seal plate and count in a Wallac Microbeta scintillation counter. Human Umbilical Vein Endothelial Cell Mitogenesis Assay 5 Expression of VEGF receptors that mediate mitogenic responses to the growth factor is largely restricted to vascular endothelial cells. Human umbilical vein endothelial cells (HUVECs) in culture proliferate in response to VEGF treatment and can be used as an assay system to quantify the effects of KDR kinase inhibitors on VEGF stimulation. In the assay described, quiescent HUVEC monolayers are treated 10 with vehicle or test compound 2 hours prior to addition of VEGF or basic fibroblast growth factor (bFGF). The mitogenic response to VEGF or bFGF is determined by measuring the incorporation of [ 3 H]thymidine into cellular DNA. MATERIALS 15 HUVECs HUVECs frozen as primary culture isolates are obtained from Clonetics Corp. Cells are maintained in Endothelial Growth Medium (EGM; Clonetics) and are used for mitogenic assays at passages 3-7. 20 Culture Plates NUNCLON 96-well polystyrene tissue culture plates (NUNC #167008). - 59 - WO 01/62252 PCT/USO1/05567 Assay Medium Dulbecco's modification of Eagle's medium containing 1 g/mL glucose (low-glucose DMEM; Mediatech) plus 10% (v/v) fetal bovine serum (Clonetics). 5 Test Compounds Working stocks of test compounds are diluted serially in 100% dimethylsulfoxide (DMSO) to 400-fold greater than their desired final concentrations. Final dilutions to 1X concentration are made directly into Assay Medium immediately 10 prior to addition to cells. 1oX Growth factors Solutions of human VEGF165 (500 ng/mL; R&D Systems) and bFGF (10 ng/mL; R&D Systems) are prepared in Assay Medium. 15 lox r 3 H1Thymidine [Methyl- 3 H]Thymidine (20 Ci/mmol; Dupont-NEN) is diluted to 80 uCi/mL in low-glucose DMEM. 20 Cell Wash Medium Hank's balanced salt solution (Mediatech) containing 1 mg/mL bovine serum albumin (Boehringer-Mannheim). Cell Lysis Solution 25 1 N NaOH, 2% (w/v) Na2CO3. - 60 - WO 01/62252 PCT/USO1/05567 METHOD 1. HUVEC monolayers maintained in EGM are harvested by trypsinization and plated at a density of 4000 cells per 100 ptL Assay Medium per well in 96-well plates. Cells are growth-arrested for 24 hours at 37"C in a humidified 5 atmosphere containing 5% C02. 2. Growth-arrest medium is replaced by 100 pL Assay Medium containing either vehicle (0.25% [v/v] DMSO) or the desired final concentration of test compound. All determinations are performed in triplicate. Cells are then incubated at 37"C/5% C02 for 2 hours to allow test compounds to enter cells. 10 3. After the 2-hour pretreatment period, cells are stimulated by addition of 10 piL/well of either Assay Medium, lOX VEGF solution or 1OX bFGF solution. Cells are then incubated at 37"C/5% C02. 4. After 24 hours in the presence of growth factors, 1OX [ 3 H] Thymidine (10 pL/well) is added. 15 5. Three days after addition of [ 3 H]thymidine, medium is removed by aspiration, and cells are washed twice with Cell Wash Medium (400 pL/well followed by 200 pL/well). The washed, adherent cells are then solubilized by addition of Cell Lysis Solution (100 pL/well) and warming to 37"C for 30 minutes. Cell lysates are transferred to 7-mL glass scintillation vials containing 150 piL of water. Scintillation 20 cocktail (5 mL/vial) is added, and cell-associated radioactivity is determined by liquid scintillation spectroscopy. Based upon the foregoing assays the compounds of formula I are inhibitors of VEGF and thus are useful for the inhibition of angiogenesis, such as in the treatment of ocular disease, e.g., diabetic retinopathy and in the treatment 25 of cancers, e.g., solid tumors. The instant compounds inhibit VEGF-stimulated mitogenesis of human vascular endothelial cells in culture with IC50 values between - 61 - WO 01/62252 PCT/USO1/05567 0.01 - 5.0 pM. These compounds also show selectivity over related tyrosine kinases (e.g., FGFR1 and the Src family; for relationship between Src kinases and VEGFR kinases, see Eliceiri et al., Molecular Cell, Vol. 4, pp.915-924, December 1999). 5 EXAMPLES Examples provided are intended to assist in a further understanding of the invention. Particular materials employed, species and conditions are intended to be further illustrative of the invention and not limiting of the reasonable scope 10 thereof. EXAMPLE 1 3-(5-methoxy-1H-pyrrolo[3,2-blpyridin-2-yl)-1H-quinolin-2-one 15 0
N
\ / N H N O H Step 1: Synthesis of 2-chloro-3-iodo-quinoline (Intermediate A) 20 N CI - 62 - WO 01/62252 PCT/USO1/05567 A suspension of 3-(2-chloro)-quinolineboronic acid (5.05 g, 24.3 mmol, 1 equiv, prepared by the method of Marsais, F; Godard, A.; Queguiner, G. J. Heterocyclic Chem. 1989, 26, 15 89-1594) and N-iodosuccinimide (5.48 g, 24.4 mmol, 1.00 equiv) in acetonitrile (300 mL) was stirred at 23"C in the dark for 20 5 hours. The reaction mixture was concentrated to dryness, and the resulting yellow solid was partitioned between saturated aqueous sodium bicarbonate solution and dichloromethane. The organic layer was washed with water, then dried over magnesium sulfate and concentrated to give 2-chloro-3-iodo-quinoline (intermediate A) as a pale yellow solid. 10 'H NMR (400 MHz, CDCl 3 ) 8 8.67 (s, 1H), 7.99 (br d, IH, J = 8.4 Hz), 7.75 (br t, 1H, J = 7.7 Hz), 7.72 (br d, iH, J = 7.8 Hz), 7.57 (br t, IH, J = 7.6 Hz). Step 2: Synthesis of Intermediate B 15 Boc Intermediate B A solution of 5-methoxy-1H-pyrrolo[3,2-b]pyridine (0.930 g, 6.28 mmol, 1 equiv, prepared by the method of Mazeas, D.; Guillaumet, G.; Viaud, M-C 20 Heterocycles 1999, 50, 1065-1080), di-tert-butyl dicarbonate (1.64 g, 4.05 mmol, 1.20 equiv), and 4-dimethylaminopyridine (10 mg, 0.082 mmol, 0.013 equiv) in dichloromethane (30 mL) was stirred at 23"C for 1 hour. The reaction mixture was concentrated, and the residue was purified by flash column chromatography (100% hexanes initially, grading to 30% ethyl acetate in hexanes) to afford intermediate B as 25 a colorless oil. - 63 - WO 01/62252 PCT/USO1/05567 'H NMR (300 MHz, CDCl 3 ) 8 8.24 (br d, 1H, J= 9.0 Hz), 7.72 (br d, 111, J= 3.4 Hz), 6.69 (d, 1H, J= 9.0 Hz), 6.63 (d, 1H, J= 3.9 Hz), 3.99 (s, 311), 1.67 (s, 9H). Step 3: Synthesis of Intermediate C 5 0
N
Boo N CI Intermediate C Step 1: A solution of tert-butyllithium in pentane (1.7 M, 3.95 mL, 10 6.72 mmol, 1.20 equiv) was added to a solution of intermediate B (1.39 g, 5.60 mmol, 1 equiv) in THFE (70 mL) at -78'C. The orange solution was stirred for 15 min, then a solution of trimethyltin chloride (2.23 g, 11.2 mmol, 2.00 equiv) in THF (4.0 mL) was added. The reaction mixture was warmed to 23*C, then partitioned between aqueous pH 7 phosphate buffer and a 1:1 mixture of ethyl acetate and hexane 15 (100 mL). The organic layer was dried over sodium sulfate and concentrated. Step 2: A deoxygenated solution of this residue, intermediate A (0.800 g, 2.76 mmol, 0.500 equiv), tetrakis(triphenylphosphine)palladium (0.160 g, 0.140 mmol, 0.025 equiv), and cuprous iodide (0.053 g, 0.28 mmol, 0.05 equiv) in dioxane (40 mL) was heated at 90"C for 20 hours. The reaction mixture was cooled, 20 then partitioned between brine (150 mL) and ethyl acetate (150 mL). The organic layer was dried over sodium sulfate, then concentrated. The residue was purified by flash column chromatography (100% hexanes initially, grading to 30% ethyl acetate in hexanes) to afford intermediate C as a light yellow foam. - 64 - WO 01/62252 PCT/USO1/05567 'H NMR (300 MEIz, CDCl 3 ) 8 8.44 (d, 1H, J = 9.2 Hz), 8.18 (s, 1H), 8.08 (d, 1H, J= 8.5 Hz), 7.88 (d, 1H, J= 8.2 Hz), 7.79 (ddd, 1H, J= 8.5, 7.0, 1.5 Hz), 7.63 (ddd, 1H, J= 8.5, 7.0, 1.5 Hz), 6.78 (d, 1H, J= 8.8 Hz), 6.72 (s, 1H), 4.02 (s, 3H), 1.27 (s, 9H). 5 Step 4: Synthesis of 3-(5-methoxy-1H-pyrrolor3,2-blpyridin-2-yl)-1H-quinolin-2-one A solution of intermediate C (900 mg, 2.20 mmol) was heated in a 1:1 mixture of acetic acid and water (50 mL) at reflux for 16 hours. The reaction mixture was concentrated, and the residue was partitioned between aqueous saturated sodium bicarbonate solution (150 mL) and hot ethyl acetate (3 x 200 mL). The combined 10 organic layers were dried over sodium sulfate and concentrated. The residue was suspended in ethyl ether (200 mL), filtered, then air-dried to give the titled compound as a yellow solid. 'H NMR (300 MiHz, (CD 3
)
2 SO) 8 12.23 (s, 1H), 11.75 (s, 1H), 8.58 (s, 1H), 7.86 (br d, 1H, J = 9.2 Hz), 7.75 (br d, 1H, J = 7.6, Hz), 7.54 (br t, 1H, J = 7.8 Hz), 7.39 15 (d, 1H, J= 8.2 Hz), 7.26 (br t, 1H, J= 7.6 Hz), 7.18 (br s, 1H), 6.57 (d, 1H, J= 8.5 Hz), 3.88 (s, 3H). HRMS (electrospray FT/ICR) calcd for C17H14N302 [M+H]* 292.1081, found 292.1059. Examples 2-4 were synthesized in analogous fashion to Example 1 20 starting from the corresponding azaindoles prepared by the method of Hands, D.; Bishop, B.; Cameron, M.; Edwards, I. S.; Cottrell, I. F.; Wright, S. H. B Synthesis 1996, 887-882. EXAMPLE 2 25 3-(5-methoxy-1H-pyrrolo[2,3-clpyridin-2-yl)-1H-quinolin-2-one - 65 - WO 01/62252 PCT/USO1/05567 0 /N N H N O H Step 1: Synthesis of Intermediate D N N 5 Boc Intermediate D A solution of 5-methoxy-1H-pyrrolo[2,3-c]pyridine (190 mg, 1.28 mmol, 1 equiv, prepared by the method of Mazeas, D.; Guillaumet, G.; Viaud, M-C 10 Heterocycles 1999, 50, 1065-1080), di-tert-butyl dicarbonate (336 mg, 1.54 mmol, 1.20 equiv), and 4-dimethylaminopyridine (10 mg, 0.082 mmol, 0.064 equiv) in dichloromethane (20 mL) was stirred at 23"C for 2 hours. The reaction mixture was concentrated, and the residue was purified by flash column chromatography (100% hexanes initially, grading to 20% ethyl acetate in hexanes) to afford intermediate E as 15 a colorless oil which solidified upon standing (180 mg, 56%). 'H NMR (300 MlHz, CDCl 3 ) 5 8.89 (br s, 1H), 7.70 (br d, 1H, J = 4.0 Hz), 6.86 (s, 1H), 6.48 (d, 1H, J= 3.9 H~z), 3.98 (s, 3H), 1.68 (s, 9H). Step 2: Synthesis of 3-(5-methoxy-1H-pyrrolo[2,3-clpyridin-2-yl-1H-quinolin-2-one 20 Step 1: A solution of tert-butyllithium in pentane (1.7 M, 0.45 mL, 0.77 mmol, 1.20 equiv) was added to a solution of intermediate D (160 mg, 0.644 - 66 - WO 01/62252 PCT/USO1/05567 mmol, 1 equiv) in THF (15 mL) at -78'C. The bright-yellow solution was stirred for 10 min, then trimethylborate (0.144 mL, 1.29 mmol, 2.00 equiv) was added. The reaction mixture was warmed to OC, then partitioned between aqueous half saturated ammonium chloride solution and ethyl acetate (2 x 75 mL). The organic 5 layer was dried over sodium sulfate and concentrated to give a white solid (160 mg). Step 2: A deoxygenated solution of this solid, intermediate A (150 mg, 0.51 mmol, 1.0 equiv), tetrakis(triphenylphosphine)palladium (30 mg, 0.026 mmol, 0.05 equiv), and potassium phosphate (327 mg, 1.54 mmol, 3.00 equiv) in dioxane (15 mL) was heated at reflux for 20 hours. The reaction mixture was cooled, 10 then partitioned between water (75 mL) and ethyl acetate (2 x 75 mL). The organic layer was dried over sodium sulfate, then concentrated. The residue was passed through a column of flash-grade silica gel (40% EtOAc in hexanes initially, grading tolOO% EtOAc). The fractions containing primarily the desired coupled product were concentrated. 15 Step 3: A solution of this residue in a 1:1 mixture of acetic acid and water was heated at reflux for 20 hours. The reaction mixture was concentrated, and the residue was purified by reverse-phase column chromatography (5% acetonitrile in water initially, grading to 100% acetonitrile). The desired fractions were concen trated, and the residue was partitioned between saturated aqueous sodium bicarbonate 20 solution and ethyl acetate. The organic layer was dried over sodium sulfate and concentrated to afford the titled compound as a yellow solid. 'H NMR (400 MHz, (CD 3
)
2 SO) 8 12.22 (s, 1H), 12.00 (s, 1H), 8.62 (s, 1H), 8.49 (s, 1H), 7.74 (br d, 1H, J = 7.7 Hz), 7.53 (br t, 1H, J = 7.7 Hz), 7.37 (br d, 1H, J = 8.2 Hz), 7.23 (br t, 1H, J= 7.5 Hz), 7.12 (s, 1H), 6.84 (s, 1H), 3.84 (s, 3H). HRMS 25 (electrospray FT/ICR) calcd for C17H14N302 [M+H]* 292.1081, found 292.1068. - 67 - WO 01/62252 PCT/USO1/05567 EXAMPLE 3 3-(5-oxo-4,5-dihydro-1H-pyrrolo[3,2-blpyridin-2-yl)-1H-quinolin-2-one 0 HN \ / N H 5 H A solution of the product from Example 1 (100 mg, 0.343 mmol) in aqueous 48% Hir solution was heated at reflux for 20 hours. The reaction mixture was cooled and the yellow solid which had precipitated was filtered and washed with 10 aqueous 1 N hydrochloric acid solution. The filtered solid was then dried under vacuum to afford the titled product as a yellow solid. 1 H NMR (300 MHz, (CD 3
)
2 SO) 8 14.20 (br s, 1H), 12.51 (s, 1H), 12.40 (s, 1), 8.81 (s, 1H), 8.29 (br d, 1H, J= 9.2 Hz), 7.81 (br d, 1H, J= 7.9 Hz), 7.60 (br t, 1H, J= 7.0 Hz), 7.41 (d, 1H, J = 8.2 Hz), 7.30 (br t, 1H, J = 7.6 Hz), 7.14 (br s, 1H), 6.70 (d, 1H, 15 J= 8.8 Hz). EXAMPLE 4 3-(5-oxo-5,6-dihydro-1H-pyrrolo[2,3-clpyridin-2-yl)-1H-quinolin-2-one 20 - 68 - WO 01/62252 PCT/USO1/05567 0 NH N H N O H The titled compound can be made by the reaction of the corresponding methyl ether with HBr according to the procedure in Example 3 above. 5 EXAMPLE 5 3-(4-oxo-4,5-dihydro-1H-pyrrolo[3,2-clpyridin-2-yl)-1H-quinolin-2-one 0 NH \ / N H N O 10 H The titled compound can be made via oxidation of the product from Example 3 followed by rearrangement (see Scheme 4). 15 EXAMPLE 6 3-(4-methyl-5-oxo-4,5-dihydro-1H-pyrrolo[3,2-blpyridin-2-yl)-1H-quinolin-2-one - 69 - WO 01/62252 PCT/USO1/05567 0 N \ / H NO H Sodium tert-butoxide (10 mg, 0.11 mmol, 1.0 equiv) and iodomethane 5 (13 pL, 0.22 mmol, 2.0 equiv) were added sequentially to a suspension of the product from Example 3 (30 mg, 0.11 mmol, 1 equiv) in THF (10 mL) at 23"C. The reaction mixture was stirred for 20 hours. Additional sodium tert-butoxide (20 mg, 0.22mmol, 2.0 equiv) and iodomethane (26 gL, 0.44 mmol, 4.0 equiv) were added in 2 equal portions over 1 hour, and then the reaction mixture was allowed to stir an additional 10 1 hour. Water (5 mL) was added, and the resulting mixture was concentrated to a volume of 2 mL. The mixture was purified by reverse phase HPLC to afford the titled compound as a yellow solid. 1 H NMR (300 MHz, (CD 3
)
2 SO) 8 12.28 (s, 1H), 11.94 (s, 1H), 8.56 (s, 1H), 7.78 (d, 1H, J = 9.5 Hz), 7.73 (br d, 1H, J = 7.9 Hz), 7.53 (br t, 1H, J = 7.0 Hz), 7.38 (br d, 15 1H, J = 8.2 Hz), 7.26 (br t, 1H, J = 7.3 Hz), 7.03 ( br d, J= 1.5 Hz), 6.18 (d, 1H, J= 9.2 Hz). Examples 7-9 were synthesized in analogous fashion to Example 1 starting from the corresponding azaindoles prepared by the method of Hands, D.; 20 Bishop, B.; Cameron, M.; Edwards, J. S.; Cottrell, I. F.; Wright, S. H. B Synthesis 1996, 887-882. The products can be converted to the N-alkylated compounds of Formula I via the procedure in Scheme 4 followed by alkylation as in Scheme 5, or by other procedures readily available in the chemical literature. 25 EXAMPLE 7 - 70 - WO 01/62252 PCT/USO1/05567 3-(1H-pyrrolo[2,3-clpyridin-2-yl)-1H-quinolin-2-one /N N H N O H 5 H NMR (400 MHz, (CD 3
)
2 SO) 8 12.26 (s, 1H), 12.03 (s, 1H), 8.89 (br s, 1H), 8.70 (br s, 1H), 8.09 (br d, 1H, J = 5.0 Hz), 7.78 (br d, 1H, J = 7.7 Hz), 7.57 (br t, 1H, J = 8.0 Hz), 7.53 (br d, 1H, J = 5.3 Hz), 7.40 (br d, 1H, J = 8.3 Hz), 7.31 (br s, 1H), 7.28 (br t, 1H, J= 7.6 Hz). 10 EXAMPLE 8 3-(1H-pyrrolo[3,2-clpyridin-2-yl)-1H-quinolin-2-one -N N H N 0 15 H lH NMR (400 MHz, (CD 3
)
2 SO) 8 12.23 (s, 1H), 11.97 (s, 1H), 8.87 (br s, 1H), 8.61 (s, 1H), 8.18 (br s, 1H), 7.75 (br d, 1H, J= 7.7 Hz), 7.55 (br t, 1H, J= 8.0 Hz), 7.50 (br s, 1H), 7.45 (br s, 1H), 7.39 (br d, 1H, J = 8.2 Hz), 7.27 (br t, 1H, J = 7.6 Hz). 20 - 71 - WO 01/62252 PCT/USO1/05567 EXAMPLE 9 3-(1H-pyrrolo[3,2-blpyridin-2-yl)-1H-quinolin-2-one
N
N H N1 0 5 H H NMR (400 MHz, (CD30D) 8 8.62 (s, 1H), 8.31 (dd, 1H, J= 4.7, 1.3 Hz), 7.92 (br d, 1H, J = 8.2 Hz), 7.81 (br d, 1H, J = 7.8 Hz), 7.58 (br t, 1H, J = 7.6 Hz), 7.40 (br d, 1H, J = 8.0 Hz), 7.34 (br s, 1H), 7.31 (br t, 1H, J = 8.0 Hz), 7.18 (dd, 1H, J = 8.2, 4.7 10 Hz). HRMS (electrospray FT/ICR) calcd for C16H1 1N30 [M+H]* 262.0975, found 262.0975. - 72 -

Claims (32)

1. A compound of Formula I (R RR N Q N O 5 or a pharmaceutically acceptable salt or stereoisomer thereof, wherein Q is S, O, or -E=D-; 10 A is selected from the following: - 73 - WO 01/62252 PCT/USO1/05567 R 4 a4a R4a N 0 NRW R4 R 4 R 4 0R 4a R 4 a andN O R4a ais Oor1; bis 0orl; 5 sis lor2; mis 0, 1, or
2; E=D is C=N, N=C, or C=C; 10 R1, Ria, R 4 and R 5 are independently selected from: 1) H, 2) (C=0)aObCl-C1O alkyl, optionally substituted with one to three substituents selected from R 6 , 3) (C=O)aObaryl, optionally substituted with one to three substituents 15 selected from R 6 , 4) (C=O)aObC2-C10 alkenyl, optionally substituted with one to three substituents selected from R 6 , 5) (C=O)aObC2-C1O alkynyl, optionally substituted with one to three substituents selected from R 6 , - 74- WO 01/62252 PCT/USO1/05567 6) SOmC1-C1O alkyl, optionally substituted with one to three substituents selected from R 6 , 7) SOmaryl, optionally substituted with one to three substituents selected from R 6 , 5 8) CO2H, 9) halo, 10) CN, 11) OH, 12) ObC1-C6 perfluoroalkyl, and 10 13) (C=O)aNR 7 R 8 ; R 2 and R 3 are independently selected from the group consisting of: 1) H, 2) (C=O)OaCi-C1O alkyl, 15 3) (C=O)Oaaryl, 4) C1-C10 alkyl, 5) SOmCi-C1O alkyl, 6) SOmaryl, 7) (C=O)aObC2-C1O alkenyl, 20 8) (C=O)aObC2-C1O alkynyl, and 9) aryl, said alkyl, aryl, alkenyl and alkynyl is optionally substituted with one to three substituents selected from R 6 ; 25 - 75 - WO 01/62252 PCT/USO1/05567 R4a is selected from the group consisting of: 1) (C=O)OaC1-C1O alkyl, 2) (C=O)Oaaryl, 3) C1-Cl alkyl, 5 4) SOmC1-C1O alkyl, 5) SOmaryl, 6) (C=O)aObC2-C1O alkenyl, 7) (C=O)aObC2-C1O alkynyl, and 8) aryl, 10 said alkyl, aryl, alkenyl and alkynyl is optionally substituted with one to three substituents selected from R 6 ; R 6 is: 15 1) H, 2) (C=O)aObCl-C6 alkyl, 3) (C=O)aObalyl, optionally substituted with one to three substituents selected from R6a, 4) C2-C1O alkenyl, 20 5) C2-C10 alkynyl, 6) heterocyclyl, optionally substituted with one to three substituents selected from R6a, 7) CO2H, 8) halo, 25 9) CN, -76- WO 01/62252 PCT/USO1/05567 10) OH, 11) oxo, 12) ObC1-C6 perfluoroalkyl, or 13) NR 7 R 8 ; 5 R 6 a is: 1) H, 2) SOmaryl, 3) SOmC1-C6 alkyl, 10 4) (C=O)aObC1-C6 alkyl, 5) (C=O)aObaryl, 6) C2-C10 alkenyl, 7) C2-C10 alkynyl, 8) heterocyclyl, 15 9) CO2H, 10) halo, 11) CN, 12) OH, 13) oxo, 20 14) ObCl-C6 perfluoroalkyl, or 15) N(C1-C6 alkyl)2; R 7 and R 8 are independently selected from: 1) H, - 77 - WO 01/62252 PCT/USO1/05567 2) (C=O)ObCi-C1O alkyl, optionally substituted with one to three substituents selected from R6a, 3) (C=O)Obaryl, optionally substituted with one to three substituents selected from R6a, 5 4) C1-C 10 alkyl, optionally substituted with one to three substituents selected from R 6 a, 5) aryl, optionally substituted with one to three substituents selected from R 6 a, 6) C2-Clo alkenyl, optionally substituted with one to three substituents 10 selected from R 6 a, 7) C2-C1O alkynyl, optionally substituted with one to three substituents selected from R 6 a, and 8) heterocyclyl, or 15 R 7 and R 8 can be taken together with the nitrogen to which they are attached to form a 5-7 membered heterocycle containing, in addition to the nitrogen, one or two additional heteroatoms selected from N, 0 and S, said heterocycle optionally substituted with one to three substituents selected from R6a. 20 2. The compound of Claim 1, wherein Q is E=D.
3. The compound of Claim 2, wherein E=D is C=C.
4. The compound of Claim 3, wherein 25 - 78 - WO 01/62252 PCT/USO1/05567 RI, Ria, R 4 and R 5 are independently selected from: 1) H, 2) (C=O)aObCl-C6 alkyl, optionally substituted with one to three substituents selected from R 6 , 5 3) (C=0)aObaryl, optionally substituted with one to three substituents selected from R 6 , 4) (C=O)aObC2-C6 alkenyl, optionally substituted with one to three substituents selected from R 6 , 5) CO2H, 10 6) halo, 7) CN, 8) OH, 9) ObC1-C3 perfluoroalkyl, and 10) (C=0)aNR 7 R 8 ; 15 R 2 and R 3 are independently selected from the group consisting of: 1) H, 2) (C=O)OaC1-C6 alkyl, and 3) C1-C6 alkyl; 20 R4a is (C=O)OaC1-C6 alkyl or C1-C6 alkyl; R 6 is: 1) H, 25 2) (C=O)aObCl-C6 alkyl, - 79 - WO 01/62252 PCT/USO1/05567 3) (C=O)aObaryl, optionally substituted with one to three substituents selected from R6a, 4) C2-C6 alkenyl, 5) heterocyclyl, optionally substituted with one to three substituents selected 5 from R6a, 6) CO2H, 7) halo, 8) CN, 9) OH, 10 10) oxo, 11) ObC1-C3 perfluoroalkyl, or 12) NR 7 R8; R 6 a is: 15 1) H, 2) SOmaryl, 3) SOmC1-C6 alkyl, 4) (C=O)aObCl-C6 alkyl, 5) (C=O)aObaryl, 20 6) C2-C6 alkenyl, 7) heterocyclyl, 8) CO2H, 9) halo, 10) CN, 25 11) OH, - 80 - WO 01/62252 PCT/USO1/05567 12) oxo, 13) ObC1-C3 perfluoroalkyl, or 14) N(C1-C6 alkyl)2; 5 R 7 and R 8 are independently selected from: 1) H, 2) (C=O)ObC1-C6 alkyl, optionally substituted with one to three substituents selected from R6a, 3) (C=O)Obaryl, optionally substituted with one to three substituents 10 selected from R 6 a, 4) C1-C6 alkyl, optionally substituted with one to three substituents selected from R6a, 5) aryl, optionally substituted with one to three substituents selected from R6a, 15 6) C2-C6 alkenyl, optionally substituted with one to three substituents selected from R6a, and 7) heterocyclyl, or R 7 and R 8 can be taken together with the nitrogen to which they are attached to form 20 a 5-7 membered heterocycle containing, in addition to the nitrogen, one or two additional heteroatoms selected from N, 0 and S, said heterocycle optionally substituted with one to three substituents selected from R6a.
5. The compound of Claim 3, wherein 25 - 81 - WO 01/62252 PCT/USO1/05567 a is 0 or 1; b is 0 or 1; s is 1; 5 R1 and R 4 are independently selected from: 1) H, 2) (C=O)aObCl-C6 alkyl, optionally substituted with one to three substituents selected from R 6 , 3) (C=O)aObaryl, optionally substituted with one to three substituents 10 selected from R 6 , 4) (C=O)aObC2-C6 alkenyl, optionally substituted with one to three substituents selected from R 6 , 5) (C=O)aObC2-C6 alkynyl, optionally substituted with one to three substituents selected from R 6 , 15
6) CO2H, 7) halo, 8) CN, 9) OH, 10) ObC1-C3 perfluoroalkyl, and 20 11) (C=0)aNR 7 R 8 ; R 2 and R 3 are independently selected from H and methyl; R4a is methyl; R 5 and Ria are H; 25 - 82 - WO 01/62252 PCT/USO1/05567 R 6 is: 1) H, 2) (C=O)aObCl-C6 alkyl,=O)aObaryl, optionally substituted with one to three substituents selected from R6a,C 2 -C10 alkenyl, 5 3) C2-C10 alkynyl, 4) heterocyclyl, optionally substituted with one to three substituents selected from R6a, 5) C02H, 6) halo, 10 7) CN, 8) OH, 9) oxo, 10) ObC1-C3 perfluoroalkyl, or 11) NR 7 R 8 ; 15 R 6 a is: 1) H, 2) SOmaryl, 3) SOmC1-C6 alkyl, 20 4) (C=O)aObC1-C6 alkyl, 5) (C=O)aObaryl, 6) C2-C1O alkenyl, 7) C2-C10 alkynyl, 8) heterocyclyl, 25 9) C02H, - 83 - WO 01/62252 PCT/USO1/05567 10) halo, 11) CN, 12) OH, 13) oxo, 5 14) ObC1-C3 perfluoroalkyl, or 15) N(C1-C6 alkyl)2; R 7 and R 8 are independently selected from: 1) H, 10 2) (C=O)ObC1-C6 alkyl, optionally substituted with one to three substituents selected from R6a, 3) (C=O)Obaryl, optionally substituted with one to three substituents selected from R6a, 4) CI-C10 alkyl, optionally substituted with one to three substituents 15 selected from R6a, 5) aryl, optionally substituted with one to three substituents selected from R6a, 6) C2-C6 alkenyl, optionally substituted with one to three substituents selected from R6a, 20 7) C2-C6 alkynyl, optionally substituted with one to three substituents selected from R6a, and 8) heterocyclyl, or - 84- WO 01/62252 PCT/USO1/05567 R 7 and R 8 can be taken together with the nitrogen to which they are attached to form a piperidinyl, piperazinyl, morpholinyl or pyrrolidinyl group, optionally substituted with one or two substituents selected from R6a. 5 6. A compound which is 3-(4-methyl-5-oxo-4,5-dihydro-1H pyrrolo[3,2-b]pyridin-2-yl)-1H-quinolin-2-one or a pharmaceutically acceptable salt or stereoisomer thereof.
7. A pharmaceutical composition which is comprised of a 10 compound in accordance with Claim 1 and a pharmaceutically acceptable carrier.
8. A method of treating or preventing cancer in a mammal in need of such treatment which is comprised of administering to said mammal a therapeutically effective amount of a compound of Claim 1. 15
9. A method of treating cancer or preventing cancer in accordance with Claim 8 wherein the cancer is selected from cancers of the brain, genitourinary tract, lymphatic system, stomach, larynx and lung. 20
10. A method of treating or preventing cancer in accordance with Claim 8 wherein the cancer is selected from histiocytic lymphoma, lung adenocarcinoma, small cell lung cancers, pancreatic cancer, gioblastomas and breast carcinoma. 25
11. A method of treating or preventing a disease in which angiogenesis is implicated, which is comprised of administering to a mammal in need of such treatment a therapeutically effective amount of a compound of Claim 1. - 85 - WO 01/62252 PCT/USO1/05567
12. A method in accordance with Claim 11 wherein the disease is an ocular disease. 5
13. A method of treating or preventing retinal vascularization which is comprised of administering to a mammal in need of such treatment a therapeutically effective amount of compound of Claim 1.
14. A method of treating or preventing diabetic retinopathy which 10 is comprised of administering to a mammal in need of such treatment a therapeutically effective amount of compound of Claim 1.
15. A method of treating or preventing age-related macular degeneration which is comprised of administering to a mammal in need of such 15 treatment a therapeutically effective amount of a compound of Claim 1.
16. A method of treating or preventing inflammatory diseases which comprises administering to a mammal in need of such treatment a therapeutically effective amount of a compound of Claim 1. 20
17. A method according to Claim 16 wherein the inflammatory disease is selected from rheumatoid arthritis, psoriasis, contact dermatitis and delayed hypersensitivity reactions. 25
18. A method of treating or preventing a tyrosine kinase-dependent disease or condition which comprises administering a therapeutically effective amount of a compound of Claim 1. - 86 - WO 01/62252 PCT/USO1/05567
19. A pharmaceutical composition made by combining the compound of Claim 1 and a pharmaceutically acceptable carrier. 5
20. A process for making a pharmaceutical composition which comprises combining a compound of Claim 1 with a pharmaceutically acceptable carter.
21. A method of treating or preventing bone associated pathologies 10 selected from osteosarcoma, osteoarthritis, and rickets which comprises administering a therapeutically effective amount of a compound of Claim 1.
22. The composition of Claim 7 further comprising a second compound selected from: 15 1) an estrogen receptor modulator, 2) an androgen receptor modulator, 3) retinoid receptor modulator, 4) a cytotoxic agent, 5) an antiproliferative agent, 20 6) a prenyl-protein transferase inhibitor, 7) an HMG-CoA reductase inhibitor, 8) an IRV protease inhibitor, 9) a reverse transcriptase inhibitor, and 10) another angiogenesis inhibitor. 25
23. The composition of Claim 22, wherein the second compound is another angiogenesis inhibitor selected from the group consisting of a tyrosine kinase - 87 - WO 01/62252 PCT/USO1/05567 inhibitor, an inhibitor of epidermal-derived growth factor, an inhibitor of fibroblast derived growth factor, an inhibitor of platelet derived growth factor, an MMP inhibitor, an integrin blocker, interferon-a, interleukin-12, pentosan polysulfate, a cyclooxygenase inhibitor, carboxyamidotriazole, combretastatin A-4, squalamine, 5 6-0-chloroacetyl-carbonyl)-fumagillol, thalidomide, angiostatin, troponin-1, and an antibody to VEGF.
24. The composition of Claim 22, wherein the second compound is an estrogen receptor modulator selected from tamoxifen and raloxifene. 10
25. A method of treating cancer which comprises administering a therapeutically effective amount of a compound of Claim 1 in combination with radiation therapy. 15
26. A method of treating or preventing cancer which comprises administering a therapeutically effective amount of a compound of Claim 1 in combination with a compound selected from: 1) an estrogen receptor modulator, 2) an androgen receptor modulator, 20 3) retinoid receptor modulator, 4) a cytotoxic agent, 5) an antiproliferative agent, 6) a prenyl-protein transferase inhibitor, 7) an HMG-CoA reductase inhibitor, 25 8) an IIIV protease inhibitor, 9) a reverse transcriptase inhibitor, and 10) another angiogenesis inhibitor. - 88 - WO 01/62252 PCT/USO1/05567
27. A method of treating cancer which comprises administering a therapeutically effective amount of a compound of Claim 1 in combination with radiation therapy and a compound selected from: 5 1) an estrogen receptor modulator, 2) an androgen receptor modulator, 3) retinoid receptor modulator, 4) a cytotoxic agent, 5) an antiproliferative agent, 10 6) a prenyl-protein transferase inhibitor, 7) an HMG-CoA reductase inhibitor, 8) an HIV protease inhibitor, 9) a reverse transcriptase inhibitor, and 10) another angiogenesis inhibitor. 15
28. A method of treating or preventing cancer which comprises administering a therapeutically effective amount of a compound of Claim 1 and paclitaxel or trastuzumab. 20
29. A method of treating or preventing cancer which comprises administering a therapeutically effective amount of a compound of Claim 1 and a GPIIbIIa antagonist.
30. The method of Claim 29 wherein the GPIIb/IIa antagonist is 25 tirofiban. -89- WO 01/62252 PCT/USO1/05567
31. A method of reducing or preventing tissue damage following a cerebral ischemic event which comprises administering a therapeutically effective amount of a compound of Claim 1. 5
32. A method of treating or preventing cancer which comprises administering a therapeutically effective amount of a compound of Claim 1 in combination with a COX-2 inhibitor. - 90 -
AU39814/01A 2000-02-25 2001-02-21 Tyrosine kinase inhibitors Ceased AU778417B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US18502400P 2000-02-25 2000-02-25
US60/185024 2000-02-25
PCT/US2001/005567 WO2001062252A1 (en) 2000-02-25 2001-02-21 Tyrosine kinase inhibitors

Publications (2)

Publication Number Publication Date
AU3981401A true AU3981401A (en) 2001-09-03
AU778417B2 AU778417B2 (en) 2004-12-02

Family

ID=22679242

Family Applications (1)

Application Number Title Priority Date Filing Date
AU39814/01A Ceased AU778417B2 (en) 2000-02-25 2001-02-21 Tyrosine kinase inhibitors

Country Status (5)

Country Link
EP (1) EP1259236A4 (en)
JP (1) JP2003523390A (en)
AU (1) AU778417B2 (en)
CA (1) CA2400923A1 (en)
WO (1) WO2001062252A1 (en)

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
OA12514A (en) 1999-12-24 2006-05-29 Aventis Pharma Ltd Azaindoles.
DE60140456D1 (en) 2000-09-01 2009-12-24 Novartis Vaccines & Diagnostic AZA HETEROCYCLIC DERIVATIVES AND ITS THERAPEUTIC USE
KR100732206B1 (en) 2000-09-11 2007-06-27 노바티스 백신즈 앤드 다이아그노스틱스 인코포레이티드 Quinolinone derivatives as tyrosine kinase inhibitors
GB0115109D0 (en) 2001-06-21 2001-08-15 Aventis Pharma Ltd Chemical compounds
MXPA04003878A (en) * 2001-10-25 2004-07-08 Novartis Ag Combinations comprising a selective cyclooxygenase-2 inhibitor.
US20030187026A1 (en) 2001-12-13 2003-10-02 Qun Li Kinase inhibitors
US6797825B2 (en) 2001-12-13 2004-09-28 Abbott Laboratories Protein kinase inhibitors
US6822097B1 (en) 2002-02-07 2004-11-23 Amgen, Inc. Compounds and methods of uses
EP1539754A4 (en) 2002-08-23 2009-02-25 Novartis Vaccines & Diagnostic Benzimidazole quinolinones and uses thereof
US20060228331A1 (en) 2003-10-10 2006-10-12 Novo Nordisk A/S IL-21 Derivatives and variants
MXPA06004199A (en) * 2003-10-17 2006-06-28 Novo Nordisk As Combination therapy.
BRPI0416143A (en) 2003-11-07 2007-01-02 Chiron Corp pharmaceutically acceptable salts of quinolinone compounds having improved pharmaceutical properties
KR20060079098A (en) * 2004-12-31 2006-07-05 주식회사 엘지생명과학 Novel ([1,3]thiazolo[5,4-b]pyridin-2-yl)-2-carboxamide derivatives
PL2301546T3 (en) 2005-01-27 2015-03-31 Novartis Ag Treatment of metastasized tumors
RU2437878C2 (en) 2005-05-17 2011-12-27 Новартис Аг Methods of producing heterocyclic compounds
GB0614471D0 (en) 2006-07-20 2006-08-30 Syngenta Ltd Herbicidal Compounds
BRPI0909659A2 (en) * 2008-03-10 2015-09-22 Sanofi Aventis treatment for eye related disorders
EP3007689B1 (en) 2013-01-10 2018-03-07 Pulmokine, Inc. Non-selective kinase inhibitors
CN106132403B (en) 2013-10-11 2020-04-28 普尔莫凯恩股份有限公司 Spray-dried formulations
MX2019004855A (en) 2016-10-27 2019-09-19 Pulmokine Inc Combination therapy for treating pulmonary hypertension.

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2102780C (en) * 1991-05-10 2007-01-09 Alfred P. Spada Bis mono-and bicyclic aryl and heteroaryl compounds which inhibit egf and/or pdgf receptor tyrosine kinase
US6162804A (en) * 1997-09-26 2000-12-19 Merck & Co., Inc. Tyrosine kinase inhibitors
US6420382B2 (en) * 2000-02-25 2002-07-16 Merck & Co., Inc. Tyrosine kinase inhibitors

Also Published As

Publication number Publication date
EP1259236A4 (en) 2004-11-03
EP1259236A1 (en) 2002-11-27
JP2003523390A (en) 2003-08-05
WO2001062252A1 (en) 2001-08-30
CA2400923A1 (en) 2001-08-30
AU778417B2 (en) 2004-12-02

Similar Documents

Publication Publication Date Title
EP1259235B1 (en) Tyrosine kinase inhibitors
US6313138B1 (en) Tyrosine kinase inhibitors
US6958340B2 (en) Tyrosine kinase inhibitors
EP1226119B1 (en) Tyrosine kinase inhibitors
US6245759B1 (en) Tyrosine kinase inhibitors
US7109204B2 (en) Tyrosine kinase inhibitors
US6927293B2 (en) Tyrosine kinase inhibitors
US20040171630A1 (en) Tyrosine kinase inhibitors
US20040235867A1 (en) Tyrosine kinase inhibitors
US20030100567A1 (en) Tyrosine kinase inhibitors
AU2001238575A1 (en) Tyrosine kinase inhibitors
US20040220216A1 (en) Tyrosine kinase inhibitors
US20040192926A1 (en) Tyrosine kinase inhibitors
AU778417B2 (en) Tyrosine kinase inhibitors
WO2003024931A1 (en) Tyrosine kinase inhibitors
US20040181066A1 (en) Tyrosine kinase inhibitors
US20040242637A1 (en) Tyrosine kinase inhibitors