EP1259236A1 - Tyrosine kinase inhibitors - Google Patents

Tyrosine kinase inhibitors

Info

Publication number
EP1259236A1
EP1259236A1 EP01914424A EP01914424A EP1259236A1 EP 1259236 A1 EP1259236 A1 EP 1259236A1 EP 01914424 A EP01914424 A EP 01914424A EP 01914424 A EP01914424 A EP 01914424A EP 1259236 A1 EP1259236 A1 EP 1259236A1
Authority
EP
European Patent Office
Prior art keywords
optionally substituted
substituents selected
alkyl
compound
inhibitor
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.)
Withdrawn
Application number
EP01914424A
Other languages
German (de)
French (fr)
Other versions
EP1259236A4 (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 EP1259236A1 publication Critical patent/EP1259236A1/en
Publication of EP1259236A4 publication Critical patent/EP1259236A4/en
Withdrawn legal-status Critical Current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

Definitions

  • 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 like in mammals.
  • tyrosine kinase-dependent diseases and conditions such as angiogenesis, cancer, tumor growth, atherosclerosis, age related macular degeneration, diabetic retinopathy, inflammatory diseases, and the 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 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 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, HER3, and HER4.
  • Ligands of this subfamily of receptors include epithileal growth factor, TGF- , amphiregulin, HB-EGF, betacellulin and heregulin.
  • TGF- epithileal growth factor
  • amphiregulin amphiregulin
  • HB-EGF betacellulin
  • heregulin Another subfamily of these receptor-type tyrosine kinases is the insulin subfamily, which includes INS-R, IGF-IR, and IR-R.
  • the PDGF subfamily includes the PDGF- ⁇ and ⁇ receptors, CSFIR, c-kit and FLK-II.
  • 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 fms-like tyrosine kinase-1 (flt-1).
  • KDR kinase insert domain receptor
  • FLK-1 fetal liver kinase- 1
  • FLK-4 fetal liver kinase-4
  • flt-1 fms-like tyrosine kinase-1
  • the non-receptor type of tyrosine kinases is also comprised of numerous subfamilies, including Src, Frk, Btk, Csk, Abl, Zap70, Fes/Fps, Fak, Jak, Ack, and LIMK. Each of these subfamilies is further sub-divided into varying receptors.
  • 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.
  • 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.
  • receptor-type tyrosine kinases and the growth factors that 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.
  • 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 it binds VEGF with high affinity.
  • VEGF and KDR are a ligand-receptor pair that play an important role in the proliferation of vascular endo- thelial cells, and the formation and sprouting of blood vessels, termed vasculogenesis and angiogenesis, respectively.
  • VEGF vascular endothelial growth factor
  • KDR 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.
  • 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). 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 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 prolif erative diseases dependent on these enzymes.
  • the angiogenic activity of VEGF is not limited to tumors.
  • VEGF 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 culminating in blindness. Ocular VEGF mRNA and protein are elevated by conditions such as retinal vein occlusion in primates and decreased ⁇ 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 retinopathy, inhibition of ocular VEGF is useful in treating the disease.
  • VEGF vascular endothelial growth factor
  • oncogenes ras, raf, src and mutant p53 all of which are relevant to targeting cancer.
  • Monoclonal anti- VEGF antibodies inhibit the growth 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 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.
  • VEGF-binding construct of Flk-1, Flt-1, the mouse KDR receptor homologue truncated to eliminate the cytoplasmic tyrosine 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, these data indicate the role of VEGF in the growth of solid tumors.
  • KDR or Flt-1 are implicated in pathological angiogenesis, and these receptors are useful in the treatment of diseases in which angiogenesis is part of the overall pathology, e.g., inflammation, diabetic retinal vascularization, as well as various forms of cancer since tumor growth is known to be dependent on angiogenesis.
  • the present invention relates to compounds that are capable of 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:
  • the compounds of this invention are useful in the inhibition of kinases and are illustrated by a compound of Formula I :
  • Rl, Rla R4 and R5 are independently selected from:
  • R2 and R3 are independently selected from the group consisting of:
  • aryl said alkyl, aryl, alkenyl and alkynyl is optionally substituted with one to three substituents selected from R6;
  • R4a is selected from the group consisting of:
  • alkyl, aryl, alkenyl and alkynyl is optionally substituted with one to three substituents selected from R6;
  • R6 is:
  • R ⁇ a is:
  • R8 are independently selected from:
  • R7 and R8 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, O and S, said heterocycle optionally substituted with one to three substituents selected from R° ⁇
  • Rl, R la , R4 and R5 are independently selected from: 1) H,
  • R4a i s (C O)O a C ⁇ -C6 alkyl or C1-C6 alkyl;
  • R6 is:
  • R ⁇ a is: 1) H,
  • R8 are independently selected from:
  • R7 and R 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, O and S, said heterocycle optionally substituted with one to three substituents selected from R6a.
  • Rl and R are independently selected from:
  • R2 and R are independently selected from H and methyl
  • R4a is methyl
  • R5 and Rla are H
  • R6 is: 1) H
  • R6a is:
  • R7 and R are independently selected from:
  • Ci-Cio alkyl optionally substituted with one to three substituents selected from R6a, 5) aryl, optionally substituted with one to three substituents selected from
  • R7 and R8 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 R° ⁇
  • Yet another embodiment of the present invention is a compound which is 3-(4-methyl-5-oxo-4,5-dihydro-lH-pyrrolo[3,2-b]pyridin-2-yl)-lH-quinolin-2-one or a pharmaceutically acceptable salt or stereoisomer thereof.
  • a 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 selected from cancers of the brain, genitourinary tract, lymphatic system, stomach, larynx and lung.
  • Another set of preferred forms of cancer are histiocytic lymphoma, lung adenocarcinoma, small cell lung cancers, pancreatic cancer, gioblastomas and breast carcinoma.
  • 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 Formula I.
  • 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 mammal in need of such treatment a therapeutically effective amount of a compound of Formula I.
  • 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 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 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 dermatitis and delayed hypersensitivity reactions, as well as treatment or prevention of bone associated pathologies selected from osteosarcoma, osteoarthritis, and rickets.
  • the invention also contemplates the use of the instantly claimed compounds in combination with a second compound selected from:
  • Preferred angiogenesis inhibitors are selected from the group 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- ⁇ , interleukin-12, pentosan polysulfate, a cyclooxygenase inhibitor, carboxyamidotriazole, combretastatin A-4, squalamine, 6-O-chloroacetyl-carbonyl)- fumagillol, thalidomide, angiostatin, troponin-1, and an antibody to VEGF.
  • Preferred estrogen receptor modulators are tamoxifen and raloxifene.
  • 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 compound selected from:
  • 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 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.
  • 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 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- 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, Stereochemistry 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.
  • the compounds disclosed herein may exist as tautomers and both tautomeric forms are intended to be encompassed by the 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.
  • any variable e.g. aryl, heterocycle, Rl, R2 etc.
  • its definition on each occurrence is independent at every other occurrence.
  • combinations of substituents and variables are permissible only if such combinations result in stable compounds.
  • Lines drawn into the ring systems from substituents indicate that the 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.
  • X and Z are C
  • R5 is CH3
  • R3 is H
  • 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 by techniques known in the art, as well as those methods set forth below, from readily available starting materials.
  • alkyl is intended to include both branched, straight- chain, and cyclic saturated aliphatic hydrocarbon groups having the specified number of carbon atoms.
  • Ci-Cio as in “Ci-Cio 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.
  • Ci-Cio 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- naphthalene, methylenecylohexyl, and so on.
  • Alkoxy represents an alkyl group of indicated number of carbon atoms attached through an oxygen bridge.
  • 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 carbon to carbon double bond is present, and up to 4 non-aromatic carbon-carbon double bonds may be present.
  • 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 if a substituted alkenyl group is indicated.
  • 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.
  • C2-C6 alkynyl means an alkynyl radical having from 2 to 6 carbon atoms.
  • Alkynyl groups include ethynyl, propynyl and butynyl. As described above with respect to alkyl, the straight, branched or cyclic portion of the alkynyl group may contain triple bonds and may be substituted if a substituted alkynyl group is indicated.
  • 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 aromatic.
  • aryl elements include phenyl, naphthyl, tetrahydro- naphthyl, indanyl, biphenyl, phenanthryl, anthryl or acenaphthyl.
  • the aryl substituent is bicyclic and one ring is non-aromatic, it is understood that attachment is via the aromatic ring.
  • heteroaryl represents a stable monocyclic or 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 O, 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, isoxazolyl, indolyl, pyrazinyl, pyridazinyl, pyridinyl, pyrimidinyl, pyrrolyl, tetrahydroquinoline.
  • 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.
  • halo or halogen as used herein is intended to include chloro, fluoro, bromo and iodo.
  • 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 O, N and S, and includes bicyclic groups. "Heterocyclyl” therefore includes the above mentioned heteroaryls, as well as dihydro and tetrathydro analogs thereof.
  • heterocyclyl include, but are not limited to the following: benzoimidazolyl, benzofuranyl, benzofurazanyl, benzopyrazolyl, benzotriazolyl, benzothiophenyl, benzoxazolyl, carbazolyl, carbolinyl, cinnolinyl, furanyl, imidazolyl, indolinyl, indolyl, indolazinyl, indazolyl, isobenzofuranyl, isoindolyl, isoquinolyl, isothiazolyl, isoxazolyl, naphthpyridinyl, oxadiazolyl, oxazolyl, oxazoline, isoxazoline, oxetanyl, pyranyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridopyridinyl, pyridazinyl, pyridazinyl
  • 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.
  • such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the 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.
  • R7 and R are defined such that they 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, O and S, said heterocycle optionally substituted with one to three substituents selected from R6a
  • 5-7 membered ring systems that can thus be formed include, but are not limited to the following:
  • Rl is H, Ci-C ⁇ alkyl, or aryl. Most preferably Rl is H or C1-C6 alkyl.
  • Rla The preferred definition of Rla is H.
  • R4 is OH, OC1-C6 alkyl, C1-C6 alkyl.
  • R4a i s C1-C6 alkyl.
  • R5 is H or C1-C6 alkyl. Most preferably R5 is H.
  • the pharmaceutically acceptable salts of the compounds of this invention can be synthesized from the compounds of this invention which contain a basic or acidic moiety by conventional chemical methods.
  • 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- orming inorganic or organic acid in a suitable solvent or various combinations of solvents.
  • 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 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.
  • 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- 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.
  • the ⁇ -alkyloxy pyridino-pyroles 3-1 can be converted to the corresponding pyrimidinone analogs 3-2 by heating with aqueous HBr.
  • the pyrimidinone analogs can be synthesized via the N-oxide 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.
  • step 1 t-BuLi
  • the instant compounds are useful as pharmaceutical agents for mammals, especially for humans, in the treatment of tyrosine kinase dependent diseases.
  • 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).
  • 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 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.
  • bone-related pathologies such as osteosarcoma, osteoarthritis, and rickets
  • oncogenic osteomalacia also known as oncogenic osteomalacia.
  • VEGF directly promotes osteoclastic bone resorption through KDR/Flk-1 expressed in mature osteoclasts (FEBS Let.
  • the instant compounds are also useful to treat and prevent conditions related to bone resorption, such as osteoporosis and Paget's disease.
  • 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); /. Clin. Invest. 104:1613-1620 (1999)).
  • the compounds of this invention may be administered to mammals, 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 administration.
  • the selected compound may be administered, for example, in the form of tablets or capsules, or as an aqueous solution or suspension.
  • carriers which are commonly used include lactose and corn starch, and lubricating agents, such as magnesium stearate, are commonly added.
  • useful diluents include lactose and dried corn starch.
  • 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.
  • sterile solutions of the active ingredient are usually prepared, and the pH of the solutions should be suitably adjusted and buffered.
  • 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 usefulness against the condition that is being treated.
  • combinations that would be useful include those with antiresorptive bisphosphonates, such as alendronate and risedronate; integrin blockers (defined further below), such as oc v ⁇ 3 antagonists; conjugated estrogens used in hormone replacement therapy, such as PREMPRO®, PREMARTN® and ENDOMETRION®; selective estrogen receptor modulators (SERMs), such as raloxifene, droloxifene, CP-336,156 (Pfizer) and lasofoxifene; cathespin K inhibitors; and ATP proton pump inhibitors.
  • SERMs selective estrogen receptor modulators
  • the instant compounds are also useful in combination with known anti-cancer agents.
  • known anti-cancer agents include the following: estrogen receptor modulators, androgen receptor modulators, retinoid receptor modulators, cytotoxic agents, antiproliferative agents, prenyl-protein transferase inhibitors, HMG-
  • CoA reductase inhibitors HJV protease inhibitors, reverse transcriptase inhibitors, and other angiogenesis inhibitors.
  • the instant compounds are particularly useful 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 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-l- oxopropoxy-4-methyl-2-[4-[2-(l-piperidinyl)ethoxy]phenyl]-2H-l-benzopyran-3-yl]- phenyl-2,2-dimethylpropanoate, 4,4'-dihydroxybenzophenone-2,4-dinitrophenyl- 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 5 ⁇ -reductase inhibitors, nilutamide, flutamide, bicalutamide, liarozole, and abiraterone acetate.
  • Retinoid receptor modulators refers to compounds which interfere or inhibit the binding of retinoids to the receptor, regardless of mechanism.
  • retinoid receptor modulators examples include bexarotene, tretinoin, 13-cis-retinoic acid, 9-cis-retinoic acid, ⁇ -difluoromethylornithine, ILX23-7553, trans-N-(4'- hydroxyphenyl) retinamide, N-4-carboxyphenyl retinamide, "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.
  • cytotoxic agents include, but are not limited to, tirapazimine, sertenef, cachectin, ifosfamide, tasonermin, lonidamine, carboplatin, altretamine, prednimustine, dibromodulcitol, ranimustine, fotemustine, nedaplatin, oxaliplatin, temozolomide, heptaplatin, estramustine, improsulfan tosilate, 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-
  • 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) benzene sulfonamide, anhydrovinblastine, N,N-dimethyl-L-valyl-L-valyl-N-methyl- L-valyl-L-prolyl-L-proline-t-butylamide, TDX258, and BMS188797.
  • 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- (6H)propanamine, l-amino-9-ethyl-5-fluoro-2,3-dihydro-9-hydroxy-4-methyl- lH,12H-benzo[de]pyrano[3',4':b,7]indolizino[l,2b]quinoline-10,13(9H,15H) dione, lurtotecan, 7-[2-(N-isopropylamino)ethyl]-(20S)camptothecin, BNP1350, BNPIllOO, BN80915, BN80942, etoposide phosphat
  • Antiproliferative agents includes antisense RNA and DNA oligonucleotides such as G3139, ODN698, RVASKRAS, GEM231 , and INX3001 , and antimetabolites such as enocitabine, carmofur, tegafur, pentostatin, doxifluridine, trimetrexate, fmdarabine, capecitabine, galocitabine, cytarabine ocfosfate, fosteabine sodium hydrate, raltitrexed, paltitrexid, emitefur, tiazofurin, decitabine, nolatrexed, pemetrexed, nelzarabine, 2'-deoxy-2'-methy ⁇ idenecytidine, 2'-fluoromethylene-2'- deoxycytidine, N-[5-(2,3-dihydro-benzofuryl)sulfonyl]-N'-(3,4-
  • Antiproliferative agents also includes monoclonal antibodies to growth factors, other than those listed under “angiogenesis inhibitors”, such as trastuzumab, and tumor suppressor genes, such as p53, which can be delivered via recombinant virus-mediated gene transfer (see U.S. Patent No. 6,069,134, for example).
  • 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. 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.
  • HMG-CoA reductase inhibitors examples include but are not limited to lovastatin (MEVACOR®; see US Patent No. 4,231,938;
  • HMG- CoA reductase inhibitor as used herein includes all pharmaceutically acceptable 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 ⁇ .
  • HMG-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 are included within the meaning of the term "HMG-CoA reductase inhibitor" as used herein.
  • the HMG-CoA reductase inhibitor is selected from lovastatin and simvastatin, and most preferably simvastatin.
  • 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 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, procaine, N-benzylphenethylamine, l-p-chlorobenzyl-2-pyrrolidine- -yl-methylbenz- imidazole, diethylamine, piperazine, and tris(hydroxymethyl) aminomethane.
  • a suitable organic or inorganic base particularly those formed from cations
  • 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, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynapthoate, iodide, isothionate, lactate, lactobionate, laurate, malate, maleate, mandelate, mesylate, methylsulfate, mucate, napsylate, nitrate, oleate, oxalate, pamao
  • 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.
  • 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-IJ (GGPTase-II, also called Rab GGPTase).
  • FPTase farnesyl-protein transferase
  • GGPTase-I geranylgeranyl-protein transferase type I
  • GGPTase-II geranylgeranyl-protein transferase type-IJ
  • prenyl-protein transferase inhibiting compounds examples include ( ⁇ )-6-[amino(4-chlorophenyl)(l-methyl-lH-imidazol-5- yl)methyl]-4-(3-chlorophenyl)-l-methyl-2(lH)-quinolinone, (-)-6-[amino(4- chlorophenyl) ( 1 -methyl- 1 ⁇ -imidazol-5-yl)methyl] -4-(3 -chlorophenyl)- 1 -methyl- 2(lH)-quinolinone, (+)-6-[amino(4-chlorophenyl)(l-methyl-l ⁇ -imidazol-5- yl)methyl]-4-(3-chlorophenyl)-l-methyl-2(lH)-quinolinone, 5(S)-n-butyl-l-(2,3- dimethylphenyl)-4-[l-(4-cyanobenzyl)-5
  • prenyl-protein transferase inhibitors can be 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, European Patent Publ.
  • HTV protease inhibitors examples include amprenavir, abacavir, CGP-73547, CGP-61755, DMP-450, indinavir, nelfinavir, tipranavir, ritonavir, saquinavir, ABT-378, AG 1776, and BMS-232,632.
  • reverse transcriptase inhibitors include delaviridine, efavirenz, GS-840, HB Y097, lamivudine, nevirapine, AZT, 3TC, ddC, and ddl.
  • Angiogenesis inhibitors refers to compounds that inhibit the formation of new blood vessels, regardless of mechanism.
  • angiogenesis inhibitors include, but are not limited to, tyrosine kinase inhibitors, such as inhibitors of the tyrosine kinase receptors Flt-1 (VEGFR1) and Flk-1/KDR (VEGFR20), inhibitors of epidermal-derived, fibroblast-derived, or platelet derived growth factors, MMP (matrix metalloprotease) inhibitors, integrin blockers, interferon- ⁇ , interleukin- 12, pentosan polysulfate, cyclooxygenase inhibitors, including nonsteroidal anti- inflammatories (NS AIDs) like aspirin and ibuprofen as well as selective cyclooxygenase-2 inhibitors like celecoxib and rofecoxib (PNAS, Vol.
  • tyrosine kinase inhibitors such as inhibitors of the tyrosine
  • NSAID's which are potent COX-2 inhibiting agents.
  • an NSAID is potent if it possess an IC50 for the inhibition of COX-2 of l ⁇ M or less as measured by the cell or microsomal assay disclosed herein.
  • NSAID's which are selective COX-2 inhibitors are defined as those which possess a specificity for 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 rnicromsal 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, U.S.
  • inhibitors of COX-2 include the following: 3-(3-fluorophenyl)-4-(4-(methylsulfonyl)phenyl)-2-(5H)-furanone;
  • Inhibitors of COX-2 that are particularly useful in the instant method of treatment are:
  • angiogenesis inhibitors include, but are not limited to, endostation, ukrain, ranpirnase, EVI862, 5-methoxy-4-[2-methyl-3-(3-methyl-2- butenyl)oxiranyl]-l-oxaspiro[2,5]oct-6-yl(chloroacetyl)carbamate, acetyldinanaline, 5-amino-l-[[3,5-dichloro-4-(4-chlorobenzoyl)phenyl]methyl]-lH-l,2,3-triazole-4- carboxamide,CM101, squalamine, combretastatin, RPI4610, NX31838, sulfated mannopentaose phosphate, 7 ,7-(carbonyl-bis [imino-N-methyl-4,2-pyrrolocarbonyl- imino[N-methyl-4,2-pyrrole]-carbonylimino]-bis-(
  • integralin Mockers refers to compounds which selectively antagonize, inhibit or counteract binding of a physiological ligand to the oc v ⁇ 3 integrin, to compounds which selectively antagonize, inhibit or counteract binding of a physiological ligand to the ⁇ v ⁇ 5 integrin, to compounds which antagonize, inhibit or counteract binding of a physiological ligand to both the ⁇ v ⁇ 3 integrin and the v ⁇ 5 integrin, and to compounds which antagonize, inhibit or counteract the activity of the particular integrin(s) expressed on capillary endothelial cells.
  • the term also refers to antagonists of the 0Cv ⁇ 6 > ⁇ v 8 > ⁇ i ⁇ i, 0C2 ⁇ l> 0C5 ⁇ l > g ⁇ i and oc ⁇ -i integrins.
  • the term also refers to antagonists of any combination of
  • tyrosine kinase inhibitors include N- (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, 2,3,9,10,1 l,12-hexahydro-10-(hydroxymethyl)-10-hydroxy-9-methyl-9,12-epoxy- lH-diindolo[l,2,3-fg:3',2',l'-kl]pyrrolo[3,
  • the instant compounds are also useful, alone or in combination with platelet fibrinogen receptor (GP Ilb/IIIa) 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 VEGF enhances metastasis by increasing extravasation at points of adhesion to vascular endothelium (Amirkhosravi, Platelets 10, 285-292, 1999). Therefore, the present compounds can serve to inhibit metastasis, alone or in combination with GP ⁇ b/HIa) antagonists.
  • fibrinogen receptor antagonists examples include abciximab, eptifibatide, sibrafiban, lamifiban, lotrafiban, cromofiban, and CT50352. If formulated as a fixed dose, such combination products employ the 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.
  • administration means introducing the compound or a prodrug of the compound into the system of the animal in need of treatment.
  • 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 sequential introduction of the compound or prodrug thereof and other agents.
  • 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.
  • therapeutically effective amount 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.
  • treating cancer refers to 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.
  • 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 pharmaceutically acceptable carriers or diluents.
  • Suitable compositions of this invention include 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.
  • the daily dosage will normally be determined by the prescribing 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.
  • 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 weight per day, preferably of between 0.5 mg/kg of body weight to about 40 mg/kg of body weight per day.
  • VEGF receptor kinase activity is measured by incorporation of radio-labeled phosphate into polyglutamic acid, tyrosine, 4:1 (pEY) substrate.
  • the phosphorylated pEY product is trapped onto a filter membrane and the incorporation of radio-labeled phosphate quantified by scintillation counting.
  • 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 domain of the KDR kinase as an in frame fusion at the carboxy terminus of the GST gene.
  • GST glutathione S-transferase
  • Soluble recombinant GST-kinase domain fusion proteins were expressed in Spodoptera frugiperda (Sf21) insect cells (Invitrogen) using a baculovirus expression vector (pAcG2T, Pharmingen).
  • wash buffer 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 lmM phenylmethylsulfonyl fluoride.
  • Enzyme dilution buffer 50 mM Tris, pH 7.4, 0.1 M NaCl, 1 mM DTT, 10 % glycerol, 100 mg/mL BSA.
  • 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.
  • Human Umbilical Vein Endothelial Cell Mitogenesis Assay 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.
  • quiescent HUVEC monolayers are treated 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 [3H]thymidine into cellular DNA.
  • 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.
  • EGM Endothelial Growth Medium
  • NUNCLON 96-well polystyrene tissue culture plates (NUNC #167008). Assay Medium
  • HUVEC monolayers maintained in EGM are harvested by trypsinization and plated at a density of 4000 cells per 100 ⁇ L Assay Medium per well in 96-well plates. Cells are growth-arrested for 24 hours at 37°C in a humidified atmosphere containing 5% CO2-
  • Growth-arrest medium is replaced by 100 ⁇ L 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% CO2 for 2 hours to allow test compounds to enter cells. 3. After the 2-hour pretreatment period, cells are stimulated by addition of 10 ⁇ L/well of either Assay Medium, 10X VEGF solution or 10X bFGF solution. Cells are then incubated at 37°C/5% CO2.
  • 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 of cancers, e.g., solid tumors.
  • the instant compounds inhibit VEGF-stimulated mitogenesis of human vascular endothelial cells in culture with IC50 values between 0.01 - 5.0 ⁇ M.
  • These compounds also show selectivity over related tyrosine kinases (e.g., FGFRl 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).
  • Step 1 Synthesis of 2-chloro-3-iodo-quinoline (Intermediate A) 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, 1589-1594) 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 hours.
  • 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, 1589-1594
  • N-iodosuccinimide 5.48 g, 24.4 mmol
  • Step 1 A solution of tert-butyllithium in pentane (1.7 M, 3.95 mL, 6.72 mmol, 1.20 equiv) was added to a solution of intermediate B (1.39 g, 5.60 mmol, 1 equiv) in THE (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 (100 mL).
  • 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, then partitioned between brine (150 mL) and ethyl acetate (150 mL). The organic layer was dried over sodium sulfate, then concentrated.
  • Step 4 Synthesis of 3-(5-methoxy-lH-pyrrolo[3.2-b1pyridin-2-yl)-lH-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 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.
  • Examples 2-4 were synthesized in analogous fashion to Example 1 starting from the corresponding azaindoles prepared by the method of Hands, D.; Bishop, B.; Cameron, M.; Edwards, J. S.; Cottrell, I. F.; Wright, S. H. B Synthesis 1996, 887-882.
  • 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 a colorless oil which solidified upon standing (180 mg, 56%).
  • Step 2 Synthesis of 3-(5-methoxy-lH-pyrrolo[2.3-c]pyridin-2-yl)-lH-quinolin-2-one
  • Step 1 A solution of tert-butyllithium in pentane (1.7 M, 0.45 mL,
  • Step 2 A deoxygenated solution of this solid, intermediate A (150 mg, 0.51 mmol, 1.0 equiv), te ⁇ akis(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 2P hours. The reaction mixture was cooled,
  • IP 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 (4P% EtOAc in hexanes initially, grading tolPP% EtOAc). The fractions containing primarily the desired coupled product were concentrated.
  • Step 3 A solution of this residue in a 1 : 1 mixture of acetic acid and water was heated at reflux for 2P hours. The reaction mixture was concentrated, and the residue was purified by reverse-phase column chromatography (5% acetonitrile in water initially, grading to 1PP% acetonitrile). The desired fractions were concentrated, and the residue was partitioned between saturated aqueous sodium bicarbonate P solution and ethyl acetate. The organic layer was dried over sodium sulfate and concentrated to afford the titled compound as a yellow solid.
  • the titled compound can be made by the reaction of the corresponding methyl ether with HBr according to the procedure in Example 3 above.
  • the titled compound can be made via oxidation of the product from Example 3 followed by rearrangement (see Scheme 4).
  • Examples 7-9 were synthesized in analogous fashion to Example 1 starting from the corresponding azaindoles prepared by the method of Hands, D.; 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.

Abstract

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 like in mammals.

Description

TITLE OF THE INVENTION TYROSΓNE KΓNASE INHIBITORS
BACKGROUND OF THE INVENTION 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 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 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 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, HER3, and HER4. Ligands of this subfamily of receptors include epithileal growth factor, TGF- , amphiregulin, HB-EGF, betacellulin and heregulin. Another subfamily of these receptor-type tyrosine kinases is the insulin subfamily, which includes INS-R, IGF-IR, and IR-R. The PDGF subfamily includes the PDGF-α and β receptors, CSFIR, c-kit and FLK-II. 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 fms-like tyrosine kinase-1 (flt-1). The PDGF and FLK 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, 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 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 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 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- 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 (Klagsburn and D'Amore, Cytokine άGrσwih 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 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). 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 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 prolif erative diseases dependent on these enzymes. The angiogenic activity of VEGF is not limited to tumors. VEGF 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 culminating in blindness. Ocular VEGF mRNA and protein are elevated by conditions such as retinal vein occlusion in primates and decreased θ2 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 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 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 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 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, 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 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 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 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:
DETAILED DESCRIPTION OF THE INVENTION
The compounds of this invention are useful in the inhibition of kinases and are illustrated by a compound of Formula I :
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
Qis S,O, or-E=D- selected from the following:
a is Oorl; bis 0 or 1 ; s is 1 or 2; m is 0, 1, or 2;
E=D is C=N, N=C, or C=C;
Rl, Rla R4 and R5 are independently selected from:
1) H,
2) (C=O)aObCι-Cio alkyl, optionally substituted with one to three substituents selected from R6, 3) (C=O)aObaryl, optionally substituted with one to three substituents selected from R6,
4) (C=O)aO C2-Cιo alkenyl, optionally substituted with one to three substituents selected from R6,
5) (C=O)aObC2-Cio alkynyl, optionally substituted with one to three substituents selected from R6,
6) SOmCι-Cιo alkyl, optionally substituted with one to three substituents selected from R6,
7) SOmaryl, optionally substituted with one to three substituents selected from R6, 8) CO2H,
9) halo,
10) CN,
11) OH,
12) ObCi-C6 perfluoroalkyl, and 13) (C=O)aNR7R8; R2 and R3 are independently selected from the group consisting of:
1) H,
2) (C=O)OaCι-Cιo alkyl, 3) (C=O)Oaaryl,
4) Ci-Cio alkyl,
5) SOmCi-Cio alkyl,
6) SOmaryl,
7) (C=O)aObC2-Cio alkenyl, 8) (C=O)aObC2-Cio alkynyl, and
9) aryl, said alkyl, aryl, alkenyl and alkynyl is optionally substituted with one to three substituents selected from R6;
R4a is selected from the group consisting of:
1) (C=O)OaCι-Cio alkyl,
2) (C=O)Oaaι l,
3) Ci-Cio alkyl,
4) SOmCι-Cιo alkyl, 5) SOmaryl,
6) (C=O)aObC2-Cιo alkenyl,
7) (C=O)aObC2-Cιo alkynyl, and
8) aryl, said alkyl, aryl, alkenyl and alkynyl is optionally substituted with one to three substituents selected from R6;
R6 is:
1) H,
2) (C=O)aObCi-C6 alkyl,
3) (C=O)aObaryl, optionally substituted with one to three substituents selected from R6a
4) C2-C 10 alkenyl,
5) C2-C10 alkynyl,
6) heterocyclyl, optionally substituted with one to three substituents selected from R6a,
7) CO2H,
8) halo,
9) CN,
10) OH, ll) oxo,
12) ObCi-C6 perfluoroalkyl, or
13) NR7R8;
Rόa is:
1) H,
2) SOmaryl,
3) SOmCl-C6 alkyl,
4) (C=O)aObCι-C6 alkyl,
5) (C=O)aObaryl, 6) C2-C10 alkenyl,
7) C2-C10 alkynyl,
8) heterocyclyl,
9) CO2H,
10) halo,
11) CN,
12) OH,
13) oxo,
14) ObCι-C6 perfluoroalkyl, or
15) N(Cι-C6 alkyl)2;
R8 are independently selected from:
1) H,
2) alkyl, optionally substituted with one to three substituents selected from R°
3) (C=O)Obaryl, optionally substituted with one to three substituents selected from R6a
4) C1-C10 alkyl, optionally substituted with one to three substituents selected from R6a; 5) aryl, optionally substituted with one to three substituents selected from
R6a
6) C2-C10 alkenyl, optionally substituted with one to three substituents selected from R6a;
7) C2-C10 alkynyl, optionally substituted with one to three substituents selected from R6a? and 8) heterocyclyl, or
R7 and R8 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, O and S, said heterocycle optionally substituted with one to three substituents selected from R°\
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 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;
Rl, Rla, R4 and R5 are independently selected from: 1) H,
2) (C=O)aObCι-C6 alkyl, optionally substituted with one to three substituents selected from R6,
3) (C=O)aObaryl, optionally substituted with one to three substituents selected from R6, 4) (C=O)aObC2-C6 alkenyl, optionally substituted with one to three substituents selected from R6,
5) CO2H,
6) halo,
7) CN, 8) OH,
9) ObCi-C3 perfluoroalkyl, and 10) (C=O)aNR7R8;
R2 and R3 are independently selected from the group consisting of: 1) H, 2) (C=O)OaCι-C6 alkyl, and
3) C1-C6 alkyl;
R4a is (C=O)OaCι-C6 alkyl or C1-C6 alkyl;
R6 is:
1) H,
2) (C=O)aObCi-C6 alkyl,
3) (C=O) Obaryl, optionally substituted with one to three substituents selected from R6a, 4) C2-C6 alkenyl,
5) heterocyclyl, optionally substituted with one to three substituents selected from R6a
6) CO2H,
7) halo, 8) CN,
9) OH, 10) oxo,
11) ObCi-C3 perfluoroalkyl, or
12) NR7R8;
Rόa is: 1) H,
2) SOmaryl,
3) SOmCι-C6 alkyl,
4) (C=O)aObCι-C6 alkyl,
5) (C=O)aObaryl,
6) C2-C6 alkenyl,
7) heterocyclyl,
8) CO2H,
9) halo,
10) CN,
11) OH,
12) oxo,
13) ObCι-C3 perfluoroalkyl, or
14) N(Cι-C6 alkyl)2;
R8 are independently selected from:
1) H,
2) (C=O)ObCi-C6 alkyl, optionally substituted with one to three substituents selected from R° 3) (C=O)Obaryl, optionally substituted with one to three substituents selected from R°
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 6) C2-C6 alkenyl, optionally substituted with one to three substituents selected from R°Λ and
7) heterocyclyl, or
R7 and R 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, O and S, said heterocycle optionally substituted with one to three substituents selected from R6a.
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 O or l; b is 0 or 1 ; s is 1;
Rl and R are independently selected from:
1) H,
2) (C=O)aObCi-C6 alkyl, optionally substituted with one to three substituents selected from R6, 3) (C=O)aObaryl, optionally substituted with one to three substituents selected from R6,
4) (C=O)aObC2~C6 alkenyl, optionally substituted with one to three substituents selected from R6,
5) (C=O)aObC2-C6 alkynyl, optionally substituted with one to three substituents selected from R6, 6) CO2H,
7) halo,
8) CN,
9) OH, 10) ObCι-C3 perfluoroalkyl, and
11) (C=O)aNR7R8;
R2 and R are independently selected from H and methyl;
R4a is methyl;
R5 and Rla are H;
R6 is: 1) H,
2) (C=O)aObCi-C6 alkyl,
3) (C=O)aObaryl, optionally substituted with one to three substituents selected from R°
4) C2-C10 alkenyl, 5) C2-C 10 alkynyl,
6) heterocyclyl, optionally substituted with one to three substituents selected from R6a,
7) CO2H,
8) halo, 9) CN,
10) OH, ll) oxo,
12) ObCι-C3 perfluoroalkyl, or
13) NR7R8;
R6a is:
1) H,
2) SOmaryl,
3) SOmCl-C6 alkyl,
4) (C=O)aObCι-C6 alkyl,
5) (C=O)aObaryl,
6) C2-C10 alkenyl,
7) C2-C10 alkynyl,
8) heterocyclyl,
9) CO2H,
10) halo,
11) CN,
12) OH,
13) oxo,
14) ObCι-C3 perfluoroalkyl, or
15) N(Cι-C6 alkyl)2;
R7 and R are independently selected from:
1) H,
2) (C=O)ObCι-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-Cio alkyl, optionally substituted with one to three substituents 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,
7) C2-C6 alkynyl, optionally substituted with one to three substituents selected from R6a, and
8) heterocyclyl, or
R7 and R8 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 R°\
Yet another embodiment of the present invention is a compound which is 3-(4-methyl-5-oxo-4,5-dihydro-lH-pyrrolo[3,2-b]pyridin-2-yl)-lH-quinolin-2-one or a pharmaceutically acceptable salt or stereoisomer thereof.
Also included within the scope of the present invention is a 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 selected from cancers of the brain, genitourinary tract, lymphatic system, stomach, larynx and lung. Another set of preferred forms of cancer are histiocytic lymphoma, 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 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 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 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 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 dermatitis and delayed hypersensitivity reactions, as well as treatment or prevention of bone associated pathologies selected from osteosarcoma, osteoarthritis, and rickets. 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, 3) retinoid receptor modulator,
4) a cytotoxic agent,
5) an antiproliferative agent,
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.
Preferred angiogenesis inhibitors are selected from the group 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-α, interleukin-12, pentosan polysulfate, a cyclooxygenase inhibitor, carboxyamidotriazole, combretastatin A-4, squalamine, 6-O-chloroacetyl-carbonyl)- 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 compound selected from:
1) an estrogen receptor modulator,
2) an androgen receptor modulator, 3) retinoid receptor modulator,
4) a cytotoxic agent,
5) an antiproliferative agent,
6) a prenyl-protein transferase inhibitor, 7) an HMG-CoA reductase inhibitor,
8) an HTV protease inhibitor,
9) a reverse transcriptase inhibitor, and
10) another angiogenesis inhibitor.
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 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 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 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- 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, Stereochemistry 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 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.
When any variable (e.g. aryl, heterocycle, Rl, R2 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 Rl, R2, R , R4 etc_) indicate that the 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,
can be, inter alia, any of the following when
X and Z are C,
Y is N,
R5 is CH3,
R3 is H, and
(R4)t is as definec in the claims:
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 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 of carbon atoms. For example, Ci-Cio, as in "Ci-Cio 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, "Ci-Cio 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- naphthalene, methylenecylohexyl, and so on. "Alkoxy" represents an alkyl group of indicated number of carbon atoms attached through an oxygen bridge. li 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 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 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 include ethynyl, propynyl and butynyl. As described above with respect to alkyl, the 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 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 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 O, 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, 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 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 O, N and S, and includes bicyclic groups. "Heterocyclyl" therefore includes the above mentioned heteroaryls, as well as dihydro and tetrathydro analogs thereof. Further examples of "heterocyclyl" include, but are not limited to the following: benzoimidazolyl, benzofuranyl, benzofurazanyl, benzopyrazolyl, benzotriazolyl, benzothiophenyl, benzoxazolyl, carbazolyl, carbolinyl, cinnolinyl, 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, 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, dihydrooxadiazolyl, dihydrooxazolyl, dihydropyrazinyl, dihydropyrazolyl, dihydropyridinyl, dihydropyrimidinyl, dihydropyrrolyl, dihydroquinolinyl, dihydrotetrazolyl, dihydrothiadiazolyl, dihydrothiazolyl, dihydrothienyl, dihydrotriazolyl, dihydroazetidinyl, methylenedioxybenzoyl, tetrahydrofuranyl, and tetrahydrothienyl, and N-oxides thereof. 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 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, R7 and R are defined such that they 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, O and S, said heterocycle optionally substituted with 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:
Preferably Q is E=D and E=D is C=C.
Preferably Rl is H, Ci-Cβ alkyl, or aryl. Most preferably Rl is H or C1-C6 alkyl.
The preferred definition of Rla is H.
Preferably R2 and R3 are independently H, C1-C6 alkyl, or (C=O)Ci-C6 alkyl. Most preferably R and R3 are independently H or C1-C6 alkyl.
Preferably R4 is OH, OC1-C6 alkyl, C1-C6 alkyl. Preferably R4a is C1-C6 alkyl.
Preferably R5 is H or C1-C6 alkyl. Most preferably R5 is H.
The pharmaceutically acceptable salts of the compounds of this invention can be synthesized from the compounds of this invention which contain a 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- orming inorganic or organic acid in a suitable solvent or various combinations of 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 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 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- 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. As shown in Scheme 3, the α-alkyloxy pyridino-pyroles 3-1 can be converted to the corresponding pyrimidinone analogs 3-2 by heating with aqueous HBr. Alternatively, the pyrimidinone analogs can be synthesized via the N-oxide 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.
SCHEME 1
1-1 1-2
step 1 : t-BuLi;
Me3SnCI step 2: Intermediate 1-2,
Pd(PPh3)4, Cul dioxane, reflux
SCHEME 2
SCHEME 3
SCHEME 4
SCHEME 5
UTILITY
The instant compounds are useful as pharmaceutical agents for mammals, especially for humans, in the treatment of tyrosine kinase dependent 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). 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 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, 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. 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); /. Clin. Invest. 104:1613-1620 (1999)).
The compounds of this invention may be administered to mammals, 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 administration.
For oral use of a chemotherapeutic compound according to this invention, the selected compound may be administered, for example, in the form 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. 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 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 usefulness against the condition that is being treated. For example, in the case of bone- related disorders, combinations that would be useful include those with antiresorptive bisphosphonates, such as alendronate and risedronate; integrin blockers (defined further below), such as ocvβ3 antagonists; conjugated estrogens used in hormone replacement therapy, such as PREMPRO®, PREMARTN® and ENDOMETRION®; selective estrogen receptor modulators (SERMs), such as raloxifene, droloxifene, 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, cytotoxic agents, antiproliferative agents, prenyl-protein transferase inhibitors, HMG-
CoA reductase inhibitors, HJV protease inhibitors, reverse transcriptase inhibitors, and other angiogenesis inhibitors. The instant compounds are particularly useful 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 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-l- oxopropoxy-4-methyl-2-[4-[2-(l-piperidinyl)ethoxy]phenyl]-2H-l-benzopyran-3-yl]- phenyl-2,2-dimethylpropanoate, 4,4'-dihydroxybenzophenone-2,4-dinitrophenyl- 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 5α-reductase inhibitors, nilutamide, flutamide, bicalutamide, liarozole, and abiraterone acetate. "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, α-difluoromethylornithine, ILX23-7553, trans-N-(4'- hydroxyphenyl) retinamide, N-4-carboxyphenyl retinamide, "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, tirapazimine, sertenef, cachectin, ifosfamide, tasonermin, lonidamine, carboplatin, altretamine, prednimustine, dibromodulcitol, ranimustine, fotemustine, nedaplatin, oxaliplatin, temozolomide, heptaplatin, estramustine, improsulfan tosilate, 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(II)]bis [diamine(chloro)platinum (II)]tetrachloride, diarizidinylspermine, arsenic trioxide, l-(ll-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- 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) benzene sulfonamide, anhydrovinblastine, N,N-dimethyl-L-valyl-L-valyl-N-methyl- L-valyl-L-prolyl-L-proline-t-butylamide, TDX258, and BMS188797.
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- (6H)propanamine, l-amino-9-ethyl-5-fluoro-2,3-dihydro-9-hydroxy-4-methyl- lH,12H-benzo[de]pyrano[3',4':b,7]indolizino[l,2b]quinoline-10,13(9H,15H) dione, lurtotecan, 7-[2-(N-isopropylamino)ethyl]-(20S)camptothecin, BNP1350, BNPIllOO, BN80915, BN80942, etoposide phosphate, teniposide, sobuzoxane, 2'-dimethylamino-2'-deoxy-etoposide, GL331, N-[2-(dimethylamino)ethyl]-9- hydroxy-5,6-dimethyl-6H-pyrido[4,3-b]carbazole-l-carboxamide, asulacrine, (5a,
5aB, 8aa,9b)-9-[2-[N-[2-(dimethylamino)ethyl]-N-methylamino]ethyl]-5-[4-hydroxy- 3,5-dimethoxyphenyl]-5,5a,6,8,8a,9-hexohydrofuro(3',4':6,7)naphtho(2,3-d)-l,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- 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,l- c]quinolin-7-one, and dimesna.
"Antiproliferative agents" includes antisense RNA and DNA oligonucleotides such as G3139, ODN698, RVASKRAS, GEM231 , and INX3001 , and antimetabolites such as enocitabine, carmofur, tegafur, pentostatin, doxifluridine, trimetrexate, fmdarabine, capecitabine, galocitabine, cytarabine ocfosfate, fosteabine sodium hydrate, raltitrexed, paltitrexid, emitefur, tiazofurin, decitabine, nolatrexed, pemetrexed, nelzarabine, 2'-deoxy-2'-methyιidenecytidine, 2'-fluoromethylene-2'- 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-pyrirnidino[5,4-b][l,4]thiazin-6-yl-(S)-ethyl]- 2,5-thienoyl-L-glutamic acid, aminopterin, 5-flurouracil, alanosine, ll-acetyl-8- (carbamoyloxymethyl)-4-formyl-6-methoxy-14-oxa-l,ll-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-l-B-D-arabino furanosyl cytosine, and 3-aminopyridine-2-carboxaldehyde thiosemicarbazone. "Antiproliferative agents" also includes monoclonal antibodies to growth factors, other than those listed under "angiogenesis inhibitors", such as trastuzumab, and tumor suppressor genes, such as p53, which can be delivered via recombinant virus-mediated gene transfer (see U.S. Patent No. 6,069,134, for example). "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. 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; 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 (LrPiTOR®; see US Patent Nos. 5,273,995; 4,681,893;
5,489,691; 5,342,952) and cerivastatin (also known as rivastatin and BAYCHOL®; 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 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 π.
Lactone Open-Acid I II
In HMG-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 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 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, procaine, N-benzylphenethylamine, l-p-chlorobenzyl-2-pyrrolidine- -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, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrabamine, hydrobromide, 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, 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. "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-IJ (GGPTase-II, also called Rab GGPTase). Examples of prenyl-protein transferase inhibiting compounds include (±)-6-[amino(4-chlorophenyl)(l-methyl-lH-imidazol-5- yl)methyl]-4-(3-chlorophenyl)-l-methyl-2(lH)-quinolinone, (-)-6-[amino(4- chlorophenyl) ( 1 -methyl- 1 Η-imidazol-5-yl)methyl] -4-(3 -chlorophenyl)- 1 -methyl- 2(lH)-quinolinone, (+)-6-[amino(4-chlorophenyl)(l-methyl-lΗ-imidazol-5- yl)methyl]-4-(3-chlorophenyl)-l-methyl-2(lH)-quinolinone, 5(S)-n-butyl-l-(2,3- dimethylphenyl)-4-[l-(4-cyanobenzyl)-5-imidazolylmethyl]-2-piperazinone, (S)-l- (3-chlorophenyl) -4- [ 1 -(4-cyanobenzyl)-5-imidazolylmethyl] -5-[2-(ethanesulf onyl) methyl)-2-piperazinone, 5(S)-n~Butyl-l-(2-methylphenyl)-4-[l-(4-cyanobenzyl)-5- imidazolylmethyl]-2-piperazinone, 1 -(3-chlorophenyl) -4- [ 1 -(4-cyanobenzyl)-2- methyl-5-imidazolylmethyl]-2-piperazinone, l-(2,2-diphenylethyl)-3-[N-(l-(4- cyanobenzyl)- lΗ-imidazol-5-ylethyl)carbamoyl]piperidine, 4- { 5- [4-Hydroxymethyl- 4-(4-chloropyridin-2-ylmethyl)-piperidine- 1 -ylmethyl] -2-methylimidazol- 1 -ylmethyl } benzonitrile, 4-{5-[4-hydroxymethyl-4-(3-chlorobenzyl)-piperidine-l-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-[l,2']bipyridin-5'- ylmethyl]-3H-imidazol-4-ylmethyl jbenzonitrile, 4-{ 3-[4-(2-Oxo-2H-[l ,2' ]bipyridin- 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-lH-imidazo[4,3-c][l,ll,4]dioxaazacyclo - nonadecine-9-carbonitrile, (±)- 19,20-Dihydro- 19-oxo-5H- 18,21 -ethano- 12, 14-etheno- 6,10-metheno-22H-benzo[. ]imidazo[4,3-A:][l,6,9,12]oxatriaza-cyclooctadecine-9- carbonitrile, 19,20-dihydro-19-oxo-5H,17H-18,21-ethano-6,10:12,16-dimetheno- 22H-imidazo[3,4-/ϊ][l,8,ll,14]oxatriazacycloeicosine-9-carbonitrile, and (±)-19,20- Dihydro-3-methyl- 19-oxo-5H- 18 ,21 -ethano- 12, 14-etheno-6, 10-metheno-22H- benzo[i]imidazo[4,3-^][l,6,9,12]oxa-triazacyclooctadecine-9-carbonitrile.
Other examples of prenyl-protein transferase inhibitors can be 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, 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, 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, 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 see European J. of Cancer, Vol. 35, No. 9, pp.1394-1401 (1999).
Examples of HTV 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, 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 (VEGFR1) and Flk-1/KDR (VEGFR20), inhibitors of epidermal-derived, fibroblast-derived, or platelet derived growth factors, MMP (matrix metalloprotease) inhibitors, integrin blockers, interferon-α, interleukin- 12, pentosan polysulfate, cyclooxygenase inhibitors, including nonsteroidal anti- inflammatories (NS AIDs) like aspirin and ibuprofen as well as selective cyclooxygenase-2 inhibitors like celecoxib and rofecoxib (PNAS, Vol. 89, p. 7384 (1992); JNCI, Vol. 69, p. 475 (1982); Arch. Opthalmol., Vol. 108, p.573 (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.107 (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. 274, p. 9116 (1999)), carboxyamidotriazole, combretastatin A-4, squalamine, 6-O- chloroacetyl-carbonyl)-fumagillol, thalidomide, angiostatin, troponin-1, angiotensin U antagonists (see Fernandez et al, J. Lab. Clin. Med. 105:141-145 (1985)), and antibodies to VEGF. (see, Nature Biotechnology, Vol. 17, pp.963-968 (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 specification an NSAID is potent if it possess an IC50 for the inhibition of COX-2 of lμM 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 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 rnicromsal 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, 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, 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: 3-(3-fluorophenyl)-4-(4-(methylsulfonyl)phenyl)-2-(5H)-furanone;
3-(3,4-difluorophenyl)-4-(4-(methylsulfonyl)phenyl)-2-(5H)-furanone; 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; 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; 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; 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; 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; 5-chloro-3-(4-methylsulfonyl)phenyl-2-(2-ethyl-5-pyridinyl)pyridine hydromethanesulf onate ;
3-(3,4-difluorophenoxy)-5,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5Η-furan-2-one; 3-(3-fluorophenoxy)-5,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-one;
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; 3-(4-chlorophenoxy)-5,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-one;
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-fluorophenylthio)-5,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-one;
3-(3,5-difluorophenylthio)-5,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2- one;
3-phenylthio-5,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-one;
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; 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-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-one;
3-(3,4-difluorophenylhydroxymethyl)-5,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H- furan-2-one; 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-(methylsulf onyl)phenyl)-3 -phenoxy- 1 -oxaspiro [4.4] non-3 -en- 2-one ;
4-(4-(methylsulfonyl)phenyl)-3-phenylthio-l-oxaspiro[4.4]non-3-en-2-one;
4-(2-oxo-3-phenylthio-l-oxa-spiro[4,4]non-3-en-4-yl) benzenesulfonamide; 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; 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-(methylsulfonyl) phenyl)-5H-furan-2- one;
3-(3-isoquinolinoxy)-5,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-one;
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-bromoρyridin-2-yloxy)-5H-furan-2- one;
5,5-dimethyl-4-(4-methylsulfonylphenyl)-3-(2-propoxy)-5H-furan-2-one;
2-(3,4-difluorophenoxy)-3-(4-methylsulfonylphenyl)-cyclopent-2-enone;
3-(5-benzothiophenyloxy)-5,5-dimethyl-4-(4-(methylsulfonyl) phenyl)-5H-furan-2- one; 5,5-dimethyl-4-(4-methylsulfonyl-phenyl)-3-(pyridyl-4-oxy)-5H-furan-2-one;
5,5-dimethyl-4-(4-methylsulfonyl-phenyl)-3-(pyridyl-3-oxy)-5H-furan-2-one;
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- 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; 3-(3-chloro-5-pyridyloxy)-5,5-dimethyl-4-(4-(methylsulfonyl) phenyl)-5H-furan-2- one; 3-(3-(l,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- 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; 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- 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; 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)phenyl-5H-furan-2- one;
3-(2-methyl-3-pyridyloxy)-5,5-dimethyl-4-(4-methylsulfonyl)phenyl-5H-furan-2-one; 3-(4-methyl-5-nitro-2-pyridyloxy)-5,5-dimethyl-4-(4-methylsulfonyl)phenyl-5H- furan-2-one;
3-(5-chloro-4-methyl-2-pyridyloxy)-5,5-dimethyl-4-(4-methylsulfonyl)phenyl-5H- furan-2-one; 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-furan-2-one;
3-(4-fluorophenoxy)-5-methyl-4-(4-methylsulfonyl)phenyl-5-propyl-5H-furan-2-one;
3-(N,N-diethylamino)-5,5-dimethyl-4-(4-(methylsulfonyl) phenyl)-5H-furan-2-one; 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)phenyl-5H-furan-2- one;
(5R)-3-(4-methoxyphenoxy)-5-ethyl-5-methyl-4-(4-methylsulfonyl)phenyl-5H-furan- 2-one;
(5R)-3-(5-chloro-2-pyridyloxy)-5-methyl-4-(4-methylsulfonyl)phenyl-5-(2,2,2- trifluoroethyl)-5H-furan-2-one;
3-(5-chloro-2-pyridyloxy)-5-methyl-4-(4-methylsulfonyl)phenyl-5-propyl-5H-furan-2- one; 3-(l-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- one;
5(R)-3-(l-cyclopropyl-ethoxy)-5-ethyl-5-methyl-4-(4-(methyl sulfonyl)phenyl-5H- furan-2-one; 5(S)-5-ethyl-5-methyl-4-(4-(methylsulfonyl)phenyl-3-(2-propoxy)-5H-furan-2-one;
3-(l-cyclopropylethoxy)-5,5-dimethyl-4-(4-(methylsulfonyl)phenyl)-5H-furan-2-one;
3-(l-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; 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; 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; (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-(l-isoquinolinyloxy)-5,5-dimethyl-4-(methylsulfonyl)phenyl-5H-furan-2-one; (5R)-3-(4-fluorophenoxy)-5-methyl-4-(4-methylsulfonyl)phenyl-5-(2,2,2- trifluoroethyl)-5H-furan-2-one;
3-(3-fluoro-2-pyridyloxy)-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 ; (5R)-3-(5-chloro-2-pyridyloxy)-5-ethyl-5-methyl-4-(4-methylsulfonyl)phenyl-5H- furan-2-one; 3-(3,4-difluorophenoxy)-5-methyl-5-trifluoromethyl-4-(4-methylsulfonyl)phenyl-5H- furan-2-one;
3-(3,4-difluorophenoxy)-5-methyl-4-(4-(methylsulfonyl)phenyl)-5-propyl-5H-furan-2- one; 3-cyclobutyloxy-5,5-dimethyl-4-(4-methylsulfonylphenyl-5H-furan-2-one;
3-(l-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- 2-one;
3-isopropoxy-5-methyl-4-(4-methylsulfonylphenyl)-5-propyl-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; (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- 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; 5-cyclopropylmethyl-3-(3,4-difluorophenoxy)-5-methyl-(4-methylsulfonyl)phenyl-
5H-furan-2-one; (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; (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; (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- furan-2-one;
3-(5-cyclopropyl-2-pyridyloxy)-5,5-dimethyl-4-(4-methylsulfonyl)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; 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
5-chloro-3-(4-methylsulfonyl)phenyl-2-(2-methyl-5-pyridinyl)pyridine;
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, 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 following:
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 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, 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 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. 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, EVI862, 5-methoxy-4-[2-methyl-3-(3-methyl-2- butenyl)oxiranyl]-l-oxaspiro[2,5]oct-6-yl(chloroacetyl)carbamate, acetyldinanaline, 5-amino-l-[[3,5-dichloro-4-(4-chlorobenzoyl)phenyl]methyl]-lH-l,2,3-triazole-4- carboxamide,CM101, squalamine, combretastatin, RPI4610, NX31838, sulfated mannopentaose phosphate, 7 ,7-(carbonyl-bis [imino-N-methyl-4,2-pyrrolocarbonyl- imino[N-methyl-4,2-pyrrole]-carbonylimino]-bis-(l,3-naphthalene disulfonate), and 3-[(2,4-dimethylpyrrol-5-yl)methylene]-2-indolinone (SU5416).
As used above, "integrin Mockers" refers to compounds which selectively antagonize, inhibit or counteract binding of a physiological ligand to the ocvβ3 integrin, to compounds which selectively antagonize, inhibit or counteract binding of a physiological ligand to the αvβ5 integrin, to compounds which antagonize, inhibit or counteract binding of a physiological ligand to both the αvβ3 integrin and the vβ5 integrin, and to compounds which antagonize, inhibit or counteract the activity of the particular integrin(s) expressed on capillary endothelial cells. The term also refers to antagonists of the 0Cvβ6> αv 8> ^iβi, 0C2βl> 0C5βl> gβi and ocββ-i integrins. The term also refers to antagonists of any combination of
αγβ3, αvβ5, ocvβ6, γβ8' al 2 5^1 ; ^l and a6^4 ∞teg™8-
Some specific examples of tyrosine kinase inhibitors include N- (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, 2,3,9,10,1 l,12-hexahydro-10-(hydroxymethyl)-10-hydroxy-9-methyl-9,12-epoxy- lH-diindolo[l,2,3-fg:3',2',l'-kl]pyrrolo[3,4-i][l,6]benzodiazocin-l-one, SH268, genistein, STI571, 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, STI571A, N-4-chlorophenyl-4-(4-pyridylmethyl)-l-phthalazinamine, and EMD121974.
The instant compounds are also useful, alone or in combination with platelet fibrinogen receptor (GP Ilb/IIIa) 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 VEGF enhances metastasis by increasing extravasation at points of adhesion to vascular endothelium (Amirkhosravi, Platelets 10, 285-292, 1999). Therefore, the present compounds can serve to inhibit metastasis, alone or in combination with GP πb/HIa) 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 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" 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 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. 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 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. 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 pharmaceutically acceptable carriers or diluents. Suitable compositions of this invention include 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 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 weight per day, preferably of between 0.5 mg/kg of body weight to about 40 mg/kg of body weight per day.
ASSAYS
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. Chem. 274:9116-9121; Sheu et si., AntkancerRes. 18:4435-4441; Ausprunk et al., Dev. Biol. 38:237-248; Gimbrone et al., J. Natl. Cancer Inst. 52:413- 427; Nicosia et al., In Vitro 18:538-549). 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 phosphorylated pEY product is trapped onto a filter membrane and the incorporation of radio-labeled phosphate quantified by scintillation counting.
MATERIALS
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 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).
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 lmM phenylmethylsulfonyl fluoride (all Sigma).
Wash buffer 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 lmM phenylmethylsulfonyl fluoride.
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 lmM phenylmethylsuflonyl fluoride.
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 50 mM Tris, pH 7.4, 0.1 M NaCl, 1 mM DTT, 10 % glycerol, 100 mg/mL BSA.
10 X Substrate
750 μg/mLpoly (glutamic acid, tyrosine; 4:1) (Sigma).
Stop solution
30% trichloroacetic acid, 0.2 M sodium pyrophosphate (both Fisher).
Wash solution 15% trichloroacetic acid, 0.2 M sodium pyrophosphate.
Filter plates Millipore #MAFC NOB, GF/C glass fiber 96 well plate.
METHOD
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 buffer followed by centrifugation at 100,000Xg 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.
B. VEGF receptor kinase assay
1. Add 5 μ\ of inhibitor or control to the assay in 50% DMSO.
2. Add 35 μl of reaction mix containing 5 μl of 10 X reaction buffer, 5 μl 25 mM ATP/10 μCi [33p]ATP (Amersham), and 5 μl 10 X substrate. 3. Start the reaction by the addition of 10 μl of KDR (25 nM) in enzyme dilution buffer.
4. Mix and incubate at room temperature for 15 minutes.
5. Stop by the addition of 50 μl stop solution.
6. Incubate for 15 minutes at 4°C. 7. Transfer a 90 μl aliquot to filter plate.
8. Aspirate and wash 3 times with wash solution. 9. Add 30 μl of scintillation cocktail, seal plate and count in a Wallac Microbeta scintillation counter.
Human Umbilical Vein Endothelial Cell Mitogenesis Assay 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 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 [3H]thymidine into cellular DNA.
MATERIALS
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.
Culture Plates
NUNCLON 96-well polystyrene tissue culture plates (NUNC #167008). 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).
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 IX concentration are made directly into Assay Medium immediately prior to addition to cells.
IPX 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.
IPX r3HlThvmidine
[Methyl-3H]Thymidine (20 Ci/mmol; Dupont-NEN) is diluted to 80 uCi/mL in low-glucose DMEM.
Cell Wash Medium
Hank's balanced salt solution (Mediatech) containing 1 mg/mL bovine serum albumin (Boehringer-Mannheim).
Cell Lysis Solution 1 N NaOH, 2% (w/v) Na2CO3. METHOD
1. HUVEC monolayers maintained in EGM are harvested by trypsinization and plated at a density of 4000 cells per 100 μL Assay Medium per well in 96-well plates. Cells are growth-arrested for 24 hours at 37°C in a humidified atmosphere containing 5% CO2-
2. Growth-arrest medium is replaced by 100 μL 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% CO2 for 2 hours to allow test compounds to enter cells. 3. After the 2-hour pretreatment period, cells are stimulated by addition of 10 μL/well of either Assay Medium, 10X VEGF solution or 10X bFGF solution. Cells are then incubated at 37°C/5% CO2.
4. After 24 hours in the presence of growth factors, 10X [3H] Thymidine (10 μL/well) is added. 5. Three days after addition of [3H]thymidine, medium is removed by aspiration, and cells are washed twice with Cell Wash Medium (400 μL/well followed by 200 μL/well). The washed, adherent cells are then solubilized by addition of Cell Lysis Solution (100 μL/well) and warming to 37°C for 30 minutes. Cell lysates are transferred to 7-mL glass scintillation vials containing 150 μL of water. Scintillation 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 of cancers, e.g., solid tumors. The instant compounds inhibit VEGF-stimulated mitogenesis of human vascular endothelial cells in culture with IC50 values between 0.01 - 5.0 μM. These compounds also show selectivity over related tyrosine kinases (e.g., FGFRl 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).
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 thereof.
EXAMPLE 1
3-(5-methoxy-lH-pyrrolor3.2-b]pyridin-2-yl)-lH-quinolin-2-one
Step 1: Synthesis of 2-chloro-3-iodo-quinoline (Intermediate A) 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, 1589-1594) 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 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. 1H NMR (400 MHz, CDC13) 5 8.67 (s, 1H), 7.99 (br d, 1H, J = 8.4 Hz), 7.75 (br t, 1H, / = 7.7 Hz), 7.72 (br d, 1H, J = 7.8 Hz), 7.57 (br t, 1H, / = 7.6 Hz).
Step 2: Synthesis of Intermediate B
Intermediate B
A solution of 5-methoxy-lH-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 Heterocycles 1999, 50, 1065-1080), di-tert-butyl dicarbonate (1.64 g, 4.05 mmol, 1.20 equiv), and 4-dimethylaminopyridine (IP 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 a colorless oil. 1H NMR (300 MHz, CDC13) δ 8.24 (br d, 1H, / = 9.0 Hz), 7.72 (br d, 1H, / = 3.4 Hz), 6.69 (d, 1H, / = 9.0 Hz), 6.63 (d, 1H, /= 3.9 Hz), 3.99 (s, 3H), 1.67 (s, 9H).
Step 3: Synthesis of Intermediate C
Intermediate C
Step 1: A solution of tert-butyllithium in pentane (1.7 M, 3.95 mL, 6.72 mmol, 1.20 equiv) was added to a solution of intermediate B (1.39 g, 5.60 mmol, 1 equiv) in THE (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 (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, 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. 1H NMR (300 MHz, CDC13) δ 8.44 (d, IH, J = 9.2 Hz), 8.18 (s, IH), 8.08 (d, IH, J = 8.5 Hz), 7.88 (d, IH, J= 8.2 Hz), 7.79 (ddd, IH, /= 8.5, 7.0, 1.5 Hz), 7.63 (ddd, IH, J= 8.5, 7.0, 1.5 Hz), 6.78 (d, IH, /= 8.8 Hz), 6.72 (s, IH), 4.02 (s, 3H), 1.27 (s, 9H).
Step 4: Synthesis of 3-(5-methoxy-lH-pyrrolo[3.2-b1pyridin-2-yl)-lH-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 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.
1H NMR (300 MHz, (CD3)2SO) δ 12.23 (s, IH), 11.75 (s, IH), 8.58 (s, IH), 7.86 (br d, IH, J = 9.2 Hz), 7.75 (br d, IH, /= 7.6, Hz), 7.54 (br t, IH, J = 7.8 Hz), 7.39 (d, IH, /= 8.2 Hz), 7.26 (br t, IH, J= 7.6 Hz), 7.18 (br s, IH), 6.57 (d, IH, J= 8.5 Hz), 3.88 (s, 3H). HRMS (electrospray FT/ICR) calcd for C17H14N3O2 [M+H]+ 292.1081, found 292.1059.
Examples 2-4 were synthesized in analogous fashion to Example 1 starting from the corresponding azaindoles prepared by the method of Hands, D.; Bishop, B.; Cameron, M.; Edwards, J. S.; Cottrell, I. F.; Wright, S. H. B Synthesis 1996, 887-882.
EXAMPLE 2
3-(5-methoxy-lH-pyrrolo[2.3-clpyridin-2-yl)-lH-quinolin-2-one
Step 1: Synthesis of Intermediate D
Intermediate D
A solution of 5-methoxy-lH-pyrrolo[2,3-c]pyridine (190 mg, 1.28 mmol, 1 equiv, prepared by the method of Mazeas, D.; Guillaumet, G.; Viaud, M-C 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 a colorless oil which solidified upon standing (180 mg, 56%).
1H NMR (300 MHz, CDC13) δ 8.89 (br s, IH), 7.70 (br d, IH, /= 4.0 Hz), 6.86 (s, IH), 6.48 (d, IH, J = 3.9 Hz), 3.98 (s, 3H), 1.68 (s, 9H).
Step 2: Synthesis of 3-(5-methoxy-lH-pyrrolo[2.3-c]pyridin-2-yl)-lH-quinolin-2-one 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 mmol, 1 equiv) in TH (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 O°C, 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), teμakis(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 2P hours. The reaction mixture was cooled,
IP 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 (4P% EtOAc in hexanes initially, grading tolPP% 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 2P hours. The reaction mixture was concentrated, and the residue was purified by reverse-phase column chromatography (5% acetonitrile in water initially, grading to 1PP% acetonitrile). The desired fractions were concentrated, and the residue was partitioned between saturated aqueous sodium bicarbonate P solution and ethyl acetate. The organic layer was dried over sodium sulfate and concentrated to afford the titled compound as a yellow solid. 1H NMR (4PP MHz, (CD3)2SO) δ 12.22 (s, IH), 12.PP (s, IH), 8.62 (s, IH), 8.49 (s, IH), 7.74 (br d, IH, / = 7.7 Hz), 7.53 (br t, IH, J = 7.7 Hz), 7.37 (br d, IH, / = 8.2 Hz), 7.23 (br t, IH, J= 7.5 Hz), 7.12 (s, IH), 6.84 (s, IH), 3.84 (s, 3H). HRMS 5 (electrospray FT/ICR) calcd for C17H14N3O2 [M+H]+ 292.1P81, found 292.1P68. EXAMPLE 3
3-(5-oxo-4.5-dihvdro-lH-pyrrolor3.2-b]pyridin-2-yl)-lH-quinolin-2-one
A solution of the product from Example 1 (100 mg, 0.343 mmol) in aqueous 48% ΗBr 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 aqueous 1 N hydrochloric acid solution. The filtered solid was then dried under vacuum to afford the titled product as a yellow solid.
1H NMR (300 MHz, (CD3)2SO) δ 14.20 (br s, IH), 12.51 (s, IH), 12.40 (s, IH), 8.81 (s, IH), 8.29 (br d, IH, J= 9.2 Hz), 7.81 (br d, IH, /= 7.9 Hz), 7.60 (br t, IH, J= 7.0 Hz), 7.41 (d, IH, J= 8.2 Hz), 7.30 (br t, IH, J = 1.6 Hz), 7.14 (br s, IH), 6.70 (d, IH, 7= 8.8 Hz).
EXAMPLE 4
3-(5-oxo-5.6-dihvdro-lH-pyrrolo[2.3-c1pyridin-2-yl)-lH-quinolin-2-one
The titled compound can be made by the reaction of the corresponding methyl ether with HBr according to the procedure in Example 3 above.
EXAMPLE 5
3-(4-oxo-4.5-dihydro-lH-pyrrolo[3.2-c1pyridin-2-yl)-lH-quinolin-2-one
The titled compound can be made via oxidation of the product from Example 3 followed by rearrangement (see Scheme 4).
EXAMPLE 6
3-(4-methyl-5-oxo-4,5-dihydro-lH-pyrrolo[3,2-b1pyridin-2-yl)-lH-quinolin-2-one
Sodium tert-butoxide (10 mg, 0.11 mmol, 1.0 equiv) and iodomethane (13 μL, 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 THE (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 μL, 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 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.
1H NMR (300 MHz, (CD3)2SO) δ 12.28 (s, IH), 11.94 (s, IH), 8.56 (s, IH), 7.78 (d, IH, J = 9.5 Hz), 7.73 (br d, IH, J = 7.9 Hz), 7.53 (br t, IH, J = 7.0 Hz), 7.38 (br d, IH, J = 8.2 Hz), 7.26 (br t, IH, / = 7.3 Hz), 7.03 ( br d, /= 1.5 Hz), 6.18 (d, IH, 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.; 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.
EXAMPLE 7 3-(lH-pyrrolo[2,3-c1pyridin-2-yl)-lH-quinolin-2-one
1H NMR (400 MHz, (CD3)2SO) δ 12.26 (s, IH), 12.03 (s, IH), 8.89 (br s, IH), 8.70 (br s, IH), 8.09 (br d, IH, / = 5.0 Hz), 7.78 (br d, IH, J = 7.7 Hz), 7.57 (br t, IH, J = 8.0 Hz), 7.53 (br d, IH, J= 5.3 Hz), 7.40 (br d, IH, / = 8.3 Hz), 7.31 (br s, IH), 7.28 (br t, IH, 7 = 7.6 Hz).
EXAMPLE 8
3-(lH-pyrrolor3.2-c1pyridin-2-yl)-lH-quinolin-2-one
1H NMR (400 MHz, (CD3)2SO) δ 12.23 (s, IH), 11.97 (s, IH), 8.87 (br s, IH), 8.61 (s, IH), 8.18 (br s, IH), 7.75 (br d, IH, J= 7.7 Hz), 7.55 (br t, IH, J= 8.0 Hz), 7.50 (br s, IH), 7.45 (br s, IH), 7.39 (br d, IH, J = 8.2 Hz), 7.27 (br t, IH, J = 7.6 Hz). EXAMPLE 9
3-(lH-pyrrolo[3,2-b1pyridin-2-yl)-lH-quinolin-2-one
1H NMR (400 MHz, (CD3OD) δ 8.62 (s, IH), 8.31 (dd, IH, J- 4.7, 1.3 Hz), 7.92 (br d, IH, J= 8.2 Hz), 7.81 (br d, IH, J= 7.8 Hz), 7.58 (br t, IH, J= 7.6 Hz), 7.40 (br d, IH, J = 8.0 Hz), 7.34 (br s, IH), 7.31 (br t, IH, J = 8.0 Hz), 7.18 (dd, IH, J = 8.2, 4.7 Hz). HRMS (electrospray FT/ICR) calcd for C16H11N3O [M+H]+ 262.0975, found 262.0975.

Claims

WHAT IS CLAIMED IS:
1. A compound of Formula I
or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
Q is S, O, or -E=D- is selected from the following:
a is O or l; b is 0 or 1; s is 1 or 2; m is 0, 1, or 2;
E=D is C=N, N=C, or C=C;
Rl, Rla, R4 and R5 are independently selected from:
1) H,
2) (C=O)aObCi-Cio alkyl, optionally substituted with one to three substituents selected from R6,
3) (C=O)aObaryl, optionally substituted with one to three substituents selected from R6 ,
4) (C=O)aObC2-Cio alkenyl, optionally substituted with one to three substituents selected from R6,
5) (C=O)aObC2-Ciθ alkynyl, optionally substituted with one to three substituents selected from R6, 6) SOmCl-Cio alkyl, optionally substituted with one to three substituents selected from R6,
7) SOmaryl, optionally substituted with one to three substituents selected from R6, 8) CO2H,
9) halo,
10) CN,
11) OH,
12) ObCχ-C6 perfluoroalkyl, and 13) (C=O)aNR7R8;
R2 and R3 are independently selected from the group consisting of:
1) H,
2) (C=O)OaCι-Cio alkyl,
3) (C=O)Oaaryl,
4) C1-C10 alkyl,
5) SOmCι-Cio alkyl,
6) SOmaryl,
7) (C=O)aObC2-Cιo alkenyl,
8) (C=O)aObC2-Cio alkynyl, and
9) aryl,
said alkyl, aryl, alkenyl and alkynyl is optionally substituted with one to three substituents selected from R6; R4a is selected from the group consisting of:
1) (C=O)OaCι-Cιo alkyl,
2) (C=O)Oaaryl,
3) Ci-Cio alkyl,
4) SOmCi-Cio alkyl,
5) SOmaryl,
6) (C=O)aObC2-Cιo alkenyl,
7) (C=O)aObC2-Cιo alkynyl, and
8) aryl,
said alkyl, aryl, alkenyl and alkynyl is optionally substituted with one to three substituents selected from R6;
R6 IS:
1) H,
2) (C=O)aObCι-C6 alkyl,
3) (C=O) Obaryl, optionally substituted with one to three substituents selected from R6
4) C2-C10 alkenyl,
5) C2-C10 alkynyl,
6) heterocyclyl, optionally substituted with one to three substituents selected from R6a,
7) CO H,
8) halo,
9) CN, 10) OH,
11) oxo,
12) ObCχ-C6 perfluoroalkyl, or
13) NR7R8;
R6a is:
1) H,
2) SOmaryl,
3) SOmCl-C6 alkyl, 4) (C=O)aObCi-C6 alkyl,
5) (C=O)aObaryl,
6) C2-C10 alkenyl,
7) C2-C10 alkynyl,
8) heterocyclyl, 9) CO2H,
10) halo,
11) CN,
12) OH,
13) oxo, 14) ObCχ-C6 perfluoroalkyl, or
15) N(Ci-C6 alkyl)2;
R7 and R are independently selected from:
1) H, 2) (C=O)ObCι-Cιo alkyl, optionally substituted with one to three substituents selected from R°
3) (C=O)Obaryl, optionally substituted with one to three substituents selected from R° ; 4) Ci-Cio alkyl, optionally substituted with one to three substituents selected from R6a,
5) aryl, optionally substituted with one to three substituents selected from R6a
6) C2-C10 alkenyl, optionally substituted with one to three substituents selected from R6a,
7) C2-C10 alkynyl, optionally substituted with one to three substituents selected from R6 ; and
8) heterocyclyl, or
R7 and R8 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, O and S, said heterocycle optionally substituted with one to three substituents selected from R \
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 Rl, Rla, R4 and R5 are independently selected from:
1) H,
2) (C=O)aObCχ-C6 alkyl, optionally substituted with one to three substituents selected from R6, 3) (C=O)aObaryl, optionally substituted with one to three substituents selected from R6,
4) (C=O)aObC2-C6 alkenyl, optionally substituted with one to three substituents selected from R6,
5) CO2H, 6) halo,
7) CN,
8) OH,
9) ObCl-C3 perfluoroalkyl, and
10) (C=O)aNR7R8;
R2 and R3 are independently selected from the group consisting of:
1) H,
2) (C=O)OaCι-C6 alkyl, and
3) C1-C6 alkyl;
R4a is (C=O)OaCi-C6 alkyl or C1.-C6 alkyl;
R6 is:
1) H, 2) (C=O)aObCι-C6 alkyl, 3) (C=O) Obaryl, optionally substituted with one to three substituents selected from R6a
4) C2-C6 alkenyl,
5) heterocyclyl, optionally substituted with one to three substituents selected from R6a
6) CO2H,
7) halo,
8) CN,
9) OH, 10) oxo, ll) O Cι-C3 perfluoroalkyl, or
12) NR7R8;
R6a is:
1) H,
2) SOmaryl,
3) SOmCι-C6 alkyl,
4) (C=O)aObCi-C6 alkyl,
5) (C=O)aObaryl,
6) C2-C6 alkenyl,
7) heterocyclyl,
8) CO2H,
9) halo,
10) CN,
11) OH, 12) oxo,
13) ObCi-C3 perfluoroalkyl, or
14)N(Ci-C6 alkyl)2;
5 R7 and R8 are independently selected from:
1) H,
2) (C=O)ObCχ-C6 alkyl, optionally substituted with one to three substituents selected from R6a;
3) (C=O)Obaryl, optionally substituted with one to three substituents
IP selected from R° ;
4) Ci-Cβ 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 R6 , and 7) heterocyclyl, or
R7 and R can be taken together with the nitrogen to which they are attached to form 2P a 5-7 membered heterocycle containing, in addition to the nitrogen, one or two additional heteroatoms selected from N, O and S, said heterocycle optionally substituted with one to three substituents selected from R6a.
5. The compound of Claim 3, wherein
25 a is O or l; b is 0 or 1 ; s is 1;
Rl and R4 ^e independently selected from:
1) H,
2) (C=O)aObCχ-C6 alkyl, optionally substituted with one to three substituents selected from R6,
3) (C=O)aObaryl, optionally substituted with one to three substituents selected from R6,
4) (C=O)aObC2-C6 alkenyl, optionally substituted with one to three substituents selected from R6,
5) (C=O)aObC2-C6 alkynyl, optionally substituted with one to three substituents selected from R6, 6) CO2H,
7) halo,
8) CN,
9) OH,
10) ObCχ-C3 perfluoroalkyl, and 11) (C=O)aNR7R8;
R2 and R3 ^e independently selected from H and methyl;
R4a is methyl;
R5 and Rla are H; R6 is:
1) H,
2) (C=O)aObCi-C6 alkyl,=O)aObaryl, optionally substituted with one to three substituents selected from R6 ,C2-Cl0 alkenyl, 3) C2-C ιo alkynyl,
4) heterocyclyl, optionally substituted with one to three substituents selected from R6a
5) CO2H,
6) halo, 7) CN,
8) OH,
9) oxo,
10) ObCχ-C3 perfluoroalkyl, or
11) NR7R8;
'a is:
1) H,
2) SOmaryl,
3) SOmCl-C6 alkyl,
4) (C=O)aO Cι-C6 alkyl,
5) (C=O)aO aryl,
6) C2-C10 alkenyl,
7) C2-C10 alkynyl,
8) heterocyclyl,
9) CO2H, 10) halo,
11) CN,
12) OH,
13) oxo, 14) ObCχ-C3 perfluoroalkyl, or
15)N(Cι-C6 alkyl)2;
R are independently selected from: 1) H, 2) (C=O)ObCι-C6 alkyl, optionally substituted with one to three substituents selected from R°
3) (C=O)Obaryl, optionally substituted with one to three substituents selected from R6a,
4) Ci-Cio alkyl, optionally substituted with one to three substituents 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 R \ 7) C2-C6 alkynyl, optionally substituted with one to three substituents selected from R6a; and 8) heterocyclyl, or R7 and R8 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.
6. A compound which is 3-(4-methyl-5-oxo-4,5-dihydro-lH- pyrrolo[3,2-b]pyridin-2-yl)-lH-quinolin-2-one or a pharmaceutically acceptable salt or stereoisomer thereof.
7. A pharmaceutical composition which is comprised of a 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.
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.
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.
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.
12. A method in accordance with Claim 11 wherein the disease is an ocular disease.
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 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 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.
17. A method according to Claim 16 wherein the inflammatory disease is selected from rheumatoid arthritis, psoriasis, contact dermatitis and delayed hypersensitivity reactions.
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.
19. A pharmaceutical composition made by combining the compound of Claim 1 and a pharmaceutically acceptable carrier.
20. A process for making a pharmaceutical composition which comprises combining a compound of Claim 1 with a pharmaceutically acceptable carrier.
21. A method of treating or preventing bone associated pathologies 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: 1) an estrogen receptor modulator,
2) an androgen receptor modulator,
3) retinoid receptor modulator,
4) a cytotoxic agent,
5) an antiproliferative agent, 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.
23. The composition of Claim 22, wherein the second compound is another angiogenesis inhibitor selected from the group 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 inhibitor, an integrin blocker, interferon-α, interleukin-12, pentosan polysulfate, a cyclooxygenase inhibitor, carboxyamidotriazole, combretastatin A-4, squalamine, 6-O-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.
25. A method of treating cancer which comprises administering a therapeutically effective amount of a compound of Claim 1 in combination with radiation therapy.
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: an estrogen receptor modulator, an androgen receptor modulator, retinoid receptor modulator, a cytotoxic agent, an antiproliferative agent, a prenyl-protein transferase inhibitor, an HMG-CoA reductase inhibitor, an HTV protease inhibitor, a reverse transcriptase inhibitor, and
10) another angiogenesis inhibitor.
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: 1) an estrogen receptor modulator,
2) an androgen receptor modulator,
3) retinoid receptor modulator,
4) a cytotoxic agent,
5) an antiproliferative agent, 6) a prenyl-protein transferase inhibitor,
7) an HMG-CoA reductase inhibitor,
8) an HTV protease inhibitor,
9) a reverse transcriptase inhibitor, and
10) another angiogenesis inhibitor.
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.
29. A method of treating or preventing cancer which comprises administering a therapeutically effective amount of a compound of Claim 1 and a GPHb/UJa antagonist.
30. The method of Claim 29 wherein the GPlTb/IIIa antagonist is tirofiban.
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.
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.
EP01914424A 2000-02-25 2001-02-21 Tyrosine kinase inhibitors Withdrawn EP1259236A4 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US18502400P 2000-02-25 2000-02-25
US185024P 2000-02-25
PCT/US2001/005567 WO2001062252A1 (en) 2000-02-25 2001-02-21 Tyrosine kinase inhibitors

Publications (2)

Publication Number Publication Date
EP1259236A1 true EP1259236A1 (en) 2002-11-27
EP1259236A4 EP1259236A4 (en) 2004-11-03

Family

ID=22679242

Family Applications (1)

Application Number Title Priority Date Filing Date
EP01914424A Withdrawn EP1259236A4 (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
BR0017038A (en) 1999-12-24 2003-01-07 Aventis Pharma Ltd Azzaindols
AU2001293233A1 (en) 2000-09-01 2002-03-13 Chiron Corporation Aza heterocyclic derivatives and their therapeutic use
BRPI0113757B8 (en) 2000-09-11 2017-11-07 Chiron Corp quinolinone derivatives as tyrosine kinase inhibitors
GB0115109D0 (en) 2001-06-21 2001-08-15 Aventis Pharma Ltd Chemical compounds
RU2333754C2 (en) * 2001-10-25 2008-09-20 Новартис Аг Compositions containing selective inhibitor of cyclooxygenase-2
US6797825B2 (en) 2001-12-13 2004-09-28 Abbott Laboratories Protein kinase inhibitors
US20030187026A1 (en) 2001-12-13 2003-10-02 Qun Li Kinase inhibitors
US6822097B1 (en) 2002-02-07 2004-11-23 Amgen, Inc. Compounds and methods of uses
EP2573079A3 (en) 2002-08-23 2015-03-11 Novartis AG 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.
WO2005046590A2 (en) 2003-11-07 2005-05-26 Chiron Corporation Methods for synthesizing quinolinone compounds
WO2006071035A1 (en) * 2004-12-31 2006-07-06 Lg Life Sciences, Ltd. Novel ([1,3]thiazolo[5,4-b]pyridin-2-yl)-2-carboxamide derivatives
KR101387985B1 (en) 2005-01-27 2014-04-25 노바티스 백신즈 앤드 다이아그노스틱스 인코포레이티드 Treatment of metastasized tumors
MX2007014381A (en) 2005-05-17 2008-02-06 Novartis Ag Methods for synthesizing heterocyclic compounds.
GB0614471D0 (en) 2006-07-20 2006-08-30 Syngenta Ltd Herbicidal Compounds
NZ587906A (en) * 2008-03-10 2011-12-22 Sanofi Aventis Treatment for ocular-related disorders
CA2897651C (en) 2013-01-10 2021-09-21 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
CA3041679A1 (en) 2016-10-27 2018-05-03 Lawrence S. ZISMAN Combination therapy for treating pulmonary hypertension

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001062251A1 (en) * 2000-02-25 2001-08-30 Merck & Co., Inc. Tyrosine kinase inhibitors

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE69222637T2 (en) * 1991-05-10 1998-02-26 Rhone Poulenc Rorer Int UP TO MONO AND BICYCLIC ARYL AND HETEROARYL DERIVATIVES WITH INHIBITING EFFECT ON EGF AND / OR PDGF RECEPTOR TYROSINKINASE
US6162804A (en) * 1997-09-26 2000-12-19 Merck & Co., Inc. Tyrosine kinase inhibitors

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001062251A1 (en) * 2000-02-25 2001-08-30 Merck & Co., Inc. Tyrosine kinase inhibitors

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
No further relevant documents disclosed *
See also references of WO0162252A1 *

Also Published As

Publication number Publication date
EP1259236A4 (en) 2004-11-03
CA2400923A1 (en) 2001-08-30
JP2003523390A (en) 2003-08-05
AU778417B2 (en) 2004-12-02
AU3981401A (en) 2001-09-03
WO2001062252A1 (en) 2001-08-30

Similar Documents

Publication Publication Date Title
EP1259235B1 (en) Tyrosine kinase inhibitors
US6313138B1 (en) Tyrosine kinase inhibitors
US6958340B2 (en) Tyrosine kinase inhibitors
US6927293B2 (en) Tyrosine kinase inhibitors
US6544988B1 (en) Tyrosine kinase inhibitors
EP1226119B1 (en) Tyrosine kinase inhibitors
US7109204B2 (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
WO2003024969A1 (en) Tyrosine kinase inhibitors
EP1226136A2 (en) Tyrosine kinase inhibitors
AU2002346053A1 (en) Tyrosine kinase inhibitors
WO2003024931A1 (en) Tyrosine kinase inhibitors
AU778417B2 (en) Tyrosine kinase inhibitors
US20040181066A1 (en) Tyrosine kinase inhibitors
WO2003015717A2 (en) Tyrosine kinase inhibitors

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20020925

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE TR

AX Request for extension of the european patent

Free format text: AL PAYMENT 20020925;LT PAYMENT 20020925;LV PAYMENT 20020925;MK PAYMENT 20020925;RO PAYMENT 20020925;SI PAYMENT 20020925

A4 Supplementary search report drawn up and despatched

Effective date: 20040921

RIC1 Information provided on ipc code assigned before grant

Ipc: 7C 07D 519/00 B

Ipc: 7A 61K 31/4355 A

Ipc: 7C 07D 209:00 J

Ipc: 7A 61K 31/437 B

Ipc: 7A 61P 35/00 B

Ipc: 7A 61K 31/4375 B

Ipc: 7C 07D 471/04 B

Ipc: 7A 61K 31/4365 B

Ipc: 7C 07D 471/04 J

Ipc: 7C 07D 221:00 J

RIN1 Information on inventor provided before grant (corrected)

Inventor name: HUNGATE, RANDALL, W.

Inventor name: HARTMAN, GEORGE, D.

Inventor name: FRALEY, MARK, E.

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

RIC1 Information provided on ipc code assigned before grant

Ipc: A61K 31/4355 20060101AFI20070227BHEP

Ipc: C07D 221/00 20060101ALN20070227BHEP

Ipc: A61P 35/00 20060101ALI20070227BHEP

Ipc: A61K 31/4365 20060101ALI20070227BHEP

Ipc: A61K 31/437 20060101ALI20070227BHEP

Ipc: C07D 471/04 20060101ALI20070227BHEP

Ipc: C07D 519/00 20060101ALI20070227BHEP

Ipc: A61K 31/4375 20060101ALI20070227BHEP

Ipc: C07D 209/00 20060101ALN20070227BHEP

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20070724