US20090258910A1 - Compounds and compositions as protein kinase inhibitors - Google Patents

Compounds and compositions as protein kinase inhibitors Download PDF

Info

Publication number
US20090258910A1
US20090258910A1 US12/492,081 US49208109A US2009258910A1 US 20090258910 A1 US20090258910 A1 US 20090258910A1 US 49208109 A US49208109 A US 49208109A US 2009258910 A1 US2009258910 A1 US 2009258910A1
Authority
US
United States
Prior art keywords
compounds
compound
abl
phenyl
raf
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.)
Abandoned
Application number
US12/492,081
Inventor
Nathanael S. Gray
Jiqing Jiang
Yi Liu
Ruo Steensma
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.)
IRM LLC
Original Assignee
IRM LLC
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 IRM LLC filed Critical IRM LLC
Priority to US12/492,081 priority Critical patent/US20090258910A1/en
Publication of US20090258910A1 publication Critical patent/US20090258910A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings

Definitions

  • the invention provides a novel class of compounds, pharmaceutical compositions comprising such compounds and methods of using such compounds to treat or prevent diseases or disorders associated with abnormal or deregulated kinase activity, particularly diseases or disorders that involve abnormal activation of the Abl, BCR-Abl, lck, SAPK2 ⁇ , PDGF-R, p38, TGF ⁇ , KDR, c-Kit, b-RAF, c-RAF, FLT1 and FLT4 kinases.
  • the protein kinases represent a large family of proteins, which play a central role in the regulation of a wide variety of cellular processes and maintaining control over cellular function.
  • a partial, non-limiting, list of these kinases include: receptor tyrosine kinases such as platelet-derived growth factor receptor kinase (PDGF-R), TGF ⁇ , VEGF-receptor kinase (e.g.
  • KDR, Flt-1 and Flt-4 the receptor kinase for stem cell factor, c-kit
  • non-receptor tyrosine kinases such Abl and the fusion kinase BCR-Abl
  • serine/threonine kinases such as p38, b-RAF and c-RAF.
  • Aberrant kinase activity has been observed in many disease states including benign and malignant proliferative disorders as well as diseases resulting from inappropriate activation of the immune and nervous systems.
  • novel compounds of this invention inhibit the activity of one or more protein kinases and are, therefore, expected to be useful in the treatment of kinase-associated diseases.
  • the present invention provides compounds of Formula I:
  • R 1 and R 2 are independently selected from hydrogen, —XR 3 , —XC(O)R 3 , —XSR 3 , —XS(O)R 3 , —XS(O) 2 R 3 , —XOR 4 , —XNC(O)NHR 3 R 4 and —XOC(O)NR 3 R 4 ; wherein X is a bond or C 1-4 alkylene; R 4 is selected from hydrogen and C 1-6 alkyl; R 3 is selected from C 6-10 aryl, C 5-10 heteroaryl, C 3-12 cycloalkyl and C 3-8 heterocycloalkyl; wherein any aryl, heteroaryl, cycloalkyl or heterocycloalkyl of R 3 is optionally substituted by 1 to 3 radicals selected from hydroxy, halo, nitro, C 1-6 alkyl, C 1-6 alkoxy, halo-substituted-C 1-6 alkyl, halo-substituted-C
  • the present invention provides a pharmaceutical composition which contains a compound of Formula I or a N-oxide derivative, individual isomers and mixture of isomers thereof; or a pharmaceutically acceptable salt thereof, in admixture with one or more suitable excipients.
  • the present invention provides a method of treating a disease in an animal in which inhibition of kinase activity, particularly Abl, BCR-Abl, PDGF-R, lck, SAPK2 ⁇ , p38, TGF ⁇ , KDR, c-Kit, b-RAF, c-RAF, FLT1 and FLT4 activity, can prevent, inhibit or ameliorate the pathology and/or symptomology of the diseases, which method comprises administering to the animal a therapeutically effective amount of a compound of Formula I or a N-oxide derivative, individual isomers and mixture of isomers thereof, or a pharmaceutically acceptable salt thereof.
  • the present invention provides the use of a compound of Formula I in the manufacture of a medicament for treating a disease in an animal in which kinase activity, particularly Abl, BCR-Abl, PDGF-R, lck, SAPK2 ⁇ , p38, TGF ⁇ , KDR, c-Kit, b-RAF, c-RAF, FLT1 and FLT4 activity, contributes to the pathology and/or symptomology of the disease.
  • kinase activity particularly Abl, BCR-Abl, PDGF-R, lck, SAPK2 ⁇ , p38, TGF ⁇ , KDR, c-Kit, b-RAF, c-RAF, FLT1 and FLT4 activity
  • the present invention provides a process for preparing compounds of Formula I and the N-oxide derivatives, prodrug derivatives, protected derivatives, individual isomers and mixture of isomers thereof, and the pharmaceutically acceptable salts thereof.
  • Alkyl as a group and as a structural element of other groups, for example halo-substituted-alkyl and alkoxy, can be either straight-chained or branched.
  • C 1-4 -alkoxy includes, methoxy, ethoxy, and the like.
  • Halo-substituted alkyl includes trifluoromethyl, pentafluoroethyl, and the like.
  • Aryl means a monocyclic or fused bicyclic aromatic ring assembly containing six to ten ring carbon atoms.
  • aryl may be phenyl or naphthyl, preferably phenyl.
  • Arylene means a divalent radical derived from an aryl group.
  • Heteroaryl is as defined for aryl where one or more of the ring members are a heteroatom.
  • heteroaryl includes pyridyl, indolyl, indazolyl, quinoxalinyl, quinolinyl, benzofuranyl, benzopyranyl, benzothiopyranyl, benzo[1,3]dioxole, imidazolyl, benzo-imidazolyl, pyrimidinyl, furanyl, oxazolyl, isoxazolyl, triazolyl, tetrazolyl, pyrazolyl, thienyl, etc.
  • Cycloalkyl means a saturated or partially unsaturated, monocyclic, fused bicyclic or bridged polycyclic ring assembly containing the number of ring atoms indicated.
  • C 3-10 cycloalkyl includes cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, etc.
  • Heterocycloalkyl means cycloalkyl, as defined in this application, provided that one or more of the ring carbons indicated, are replaced by a moiety selected from —O—, —N ⁇ , —NR—, —C(O)—, —S—, —S(O)— or —S(O) 2 —, wherein R is hydrogen, C 1-4 alkyl or a nitrogen protecting group.
  • C 3-8 heterocycloalkyl as used in this application to describe compounds of the invention includes morpholino, pyrrolidinyl, piperazinyl, piperidinyl, piperidinylone, 1,4-dioxa-8-aza-spiro[4.5]dec-8-yl, etc.
  • Halogen (or halo) preferably represents chloro or fluoro, but may also be bromo or iodo.
  • Treating refers to a method of alleviating or abating a disease and/or its attendant symptoms.
  • the present invention provides compounds, compositions and methods for the treatment of kinase related disease, particularly Abl, BCR-Abl, PDGF-R, lck, SAPK2 ⁇ , p38, TGF ⁇ , KDR, c-Kit, b-RAF, c-RAF, FLT1 and FLT4 kinase related diseases.
  • kinase related disease particularly Abl, BCR-Abl, PDGF-R, lck, SAPK2 ⁇ , p38, TGF ⁇ , KDR, c-Kit, b-RAF, c-RAF, FLT1 and FLT4 kinase related diseases.
  • leukemia and other proliferation disorders related to BCR-Abl can be treated through the inhibition of wild type and mutant forms of Bcr-Abl.
  • R 1 and R 2 are independently selected from hydrogen, —XR 3 and —XOC(O)NR 3 R 4 ; wherein X is a bond or C 1-4 alkylene; R 4 is selected from hydrogen and C 1-6 alkyl; R 3 is selected from C 6-10 aryl, C 5-10 heteroaryl and C 3-8 heterocycloalkyl; wherein any aryl, heteroaryl or heterocycloalkyl of R 3 is optionally substituted by 1 to 3 radicals selected from halo, nitro, C 1-6 alkyl, C 1-6 alkoxy, halo-substituted-C 1-6 alkyl, —XC(O)OR 4 and —NR 4 R 5 ; wherein X is a bond or C 1-4 alkylene and R 4 and R 5 are independently selected from hydrogen and C 1-6 alkyl.
  • R 1 is selected from hydrogen and phenyl; wherein said phenyl is optionally substituted by 1 to 3 radicals selected from trifluoromethyl, dimethylamino, methoxy, halo, ethoxy and nitro.
  • R 2 is selected from hydrogen, —OH and —OC(O)NHR 4 ; wherein R 4 is selected from phenyl and benzo[1,3]dioxol-5-yl; wherein said phenyl of R 4 is optionally substituted by 1 to 3 radicals selected from —C(O)OCH 3 and dimethylamino.
  • Preferred compounds of Formula I are selected from: Phenyl-carbamic acid 8-(2-phenyl-5-pyridin-4-yl-3H-imidazol-4-yl)-naphthalen-2-yl ester; 4-(5-Naphthalen-1-yl-2-phenyl-3H-imidazol-4-yl)-pyridine; 4-[2-(3,5-Bis-trifluoromethyl-phenyl)-5-naphthalen-1-yl-3H-imidazol-4-yl]-pyridine; Dimethyl-[4-(4-naphthalen-1-yl-5-pyridin-4-yl-1H-imidazol-2-yl)-phenyl]-amine; 4-[5-Naphthalen-1-yl-2-(2,4,6-trimethoxy-phenyl)-3H-imidazol-4-yl]-pyridine; 4-[2-(2-Fluoro-phenyl)-5-naphthalen-1-yl-3
  • Protein tyrosine kinases modulate the activity of protein tyrosine kinases and, as such, are useful for treating diseases or disorders in which protein tyrosine kinases, particularly Abl, BCR-Abl, PDGF-R, lck, SAPK2 ⁇ , p38, TGF ⁇ , KDR, c-Kit, b-RAF, c-RAF, FLT1 and FLT4 kinases, contribute to the pathology and/or symptomology of the disease.
  • protein tyrosine kinases particularly Abl, BCR-Abl, PDGF-R, lck, SAPK2 ⁇ , p38, TGF ⁇ , KDR, c-Kit, b-RAF, c-RAF, FLT1 and FLT4 kinases, contribute to the pathology and/or symptomology of the disease.
  • Abelson tyrosine kinase (i.e. Abl, c-Abl) is involved in the regulation of the cell cycle, in the cellular response to genotoxic stress, and in the transmission of information about the cellular environment through integrin signaling. Overall, it appears that the Abl protein serves a complex role as a cellular module that integrates signals from various extracellular and intracellular sources and that influences decisions in regard to cell cycle and apoptosis.
  • Abelson tyrosine kinase includes sub-types derivatives such as the chimeric fusion (oncoprotein) BCR-Abl with deregulated tyrosine kinase activity or the v-Abl.
  • BCR-Abl is critical in the pathogenesis of 95% of chronic myelogenous leukemia (CML) and 10% of acute lymphocytic leukemia.
  • STI-571 (Gleevec) is an inhibitor of the oncogenic BCR-Abl tyrosine kinase and is used for the treatment of chronic myeloid leukemia (CML).
  • CML chronic myeloid leukemia
  • some patients in the blast crisis stage of CML are resistant to STI-571 due to mutations in the BCR-Abl kinase. Over 22 mutations have been reported to date with the most common being G250E, E255V, T3151, F317L and M351T.
  • Compounds of the present invention inhibit abl kinase, especially v-abl kinase.
  • the compounds of the present invention also inhibit wild-type BCR-Abl kinase and mutations of BCR-Abl kinase and are thus suitable for the treatment of Bcr-abl-positive cancer and tumor diseases, such as leukemias (especially chronic myeloid leukemia and acute lymphoblastic leukemia, where especially apoptotic mechanisms of action are found), and also shows effects on the subgroup of leukemic stem cells as well as potential for the purification of these cells in vitro after removal of said cells (for example, bone marrow removal) and reimplantation of the cells once they have been cleared of cancer cells (for example, reimplantation of purified bone marrow cells).
  • Bcr-abl-positive cancer and tumor diseases such as leukemias (especially chronic myeloid leukemia and acute lymphoblastic leukemia, where especially apoptotic mechanisms of action are found)
  • PDGF Platinum-derived Growth Factor
  • PDGFR PDGF receptor
  • Compounds of the present invention can be used not only as a tumor-inhibiting substance, for example in small cell lung cancer, but also as an agent to treat non-malignant proliferative disorders, such as atherosclerosis, thrombosis, psoriasis, scleroderma and fibrosis, as well as for the protection of stem cells, for example to combat the hemotoxic effect of chemotherapeutic agents, such as 5-fluoruracil, and in asthma.
  • Compounds of the invention can especially be used for the treatment of diseases, which respond to an inhibition of the PDGF receptor kinase.
  • Compounds of the present invention show useful effects in the treatment of disorders arising as a result of transplantation, for example, allogenic transplantation, especially tissue rejection, such as especially obliterative bronchiolitis (OB), i.e. a chronic rejection of allogenic lung transplants.
  • allogenic transplantation especially tissue rejection, such as especially obliterative bronchiolitis (OB), i.e. a chronic rejection of allogenic lung transplants.
  • OB obliterative bronchiolitis
  • OB obliterative bronchiolitis
  • Compounds of the present invention are also effective in diseases associated with vascular smooth-muscle cell migration and proliferation (where PDGF and PDGF-R often also play a role), such as restenosis and atherosclerosis.
  • diseases associated with vascular smooth-muscle cell migration and proliferation where PDGF and PDGF-R often also play a role
  • PDGF and PDGF-R often also play a role
  • These effects and the consequences thereof for the proliferation or migration of vascular smooth-muscle cells in vitro and in vivo can be demonstrated by administration of the compounds of the present invention, and also by investigating its effect on the thickening of the vascular intima following mechanical injury in vivo.
  • the compounds of the present invention also inhibit cellular processes involving stem-cell factor (SCF, also known as the c-kit ligand or steel factor), such as inhibiting SCF receptor (kit) autophosphorylation and SCF-stimulated activation of MAPK kinase (mitogen-activated protein kinase).
  • SCF stem-cell factor
  • kit SCF receptor
  • MAPK kinase mitogen-activated protein kinase
  • MO7e cells are a human promegakaryocytic leukemia cell line, which depends on SCF for proliferation.
  • Compounds of the invention can inhibit the autophosphorylation of SCF receptors.
  • the Ras-Raf-MEK-ERK signaling pathway mediates cellular response to growth signals. Ras is mutated to an oncogenic form in ⁇ 15% of human cancer.
  • the Raf family belongs to the serine/threonine protein kinase and it includes three members, A-Raf, B-Raf and c-Raf (or Raf-1).
  • the focus on Raf being a drug target has centered on the relationship of Raf as a downstream effector of Ras.
  • B-Raf may have a prominent role in the formation of certain tumors with no requirement for an activated Ras allele (Nature 417, 949-954 (1 Jul. 2002).
  • B-Raf mutations have been detected in a large percentage of malignant melanomas.
  • the compounds of the present invention also inhibit cellular processes involving c-Raf kinase.
  • c-Raf is activated by the ras oncogene, which is mutated in a wide number of human cancers. Therefore inhibition of the kinase activity of c-Raf may provide a way to prevent ras mediated tumor growth [Campbell, S. L., Oncogene, 17, 1395 (1998)].
  • the compounds of the present invention also inhibit cellular processes involving KDR, Flt-1 and Flt-4.
  • Flt-4 is expressed in developing lymphatic vessels. Only the lymphatic endothelia and some high endothelial venules express Flt4 mRNA in adult human tissues and increased expression occurs in lymphatic sinuses in metastatic lymph nodes and in lymphangioma. Inhibition of KDR-mediated functional effects by inhibiting KDR's catalytic activity is considered to be an important therapeutic strategy in the treatment of angiogenized disease states including cancer.
  • p38 MAPK ⁇ , ⁇ , ⁇ , ⁇
  • kinase cascade involved in the response of cells to a variety of stimuli, including osmotic stress, UV light and cytokine mediated events.
  • osmotic stress e.g., UV light
  • cytokine mediated events e.g., UV light
  • p38 kinase cascade involved in the response of cells to a variety of stimuli, including osmotic stress, UV light and cytokine mediated events.
  • p38 is thought to regulate different aspects of intracellular signaling. Its activation is part of a cascade of signaling events that lead to the synthesis and production of pro-inflammatory cytokines like TNF ⁇ .
  • P38 functions by phosphorylating downstream substrates that include other kinases and transcription factors.
  • cytokines including but not limited to TNF ⁇ , IL-6, IL-8 and IL-1 ⁇ .
  • Peripheral blood monocytes PBMCs
  • LPS lipopolysaccharide
  • P38 inhibitors efficiently block this effect when PBMCs are pretreated with such compounds prior to stimulation with LPS.
  • P38 inhibitors are efficacious in animal models of inflammatory disease. The destructive effects of many disease states are caused by the over production of pro-inflammatory cytokines. The ability of p38 inhibitors to regulate this overproduction makes them useful as disease modifying agents.
  • Molecules that block p38's function have been shown to be effective in inhibiting bone resorption, inflammation, and other immune and inflammation-based pathologies.
  • a safe and effective p38 inhibitor would provide a means to treat debilitating diseases that can be regulated by modulation of p38 signaling like, for example, RA. Therefore, compounds of the invention that inhibit p38 activity are useful for the treatment of inflammation, osteoarthritis, rheumatoid arthritis, cancer, autoimmune diseases, and for the treatment of other cytokine mediated diseases.
  • TGF ⁇ Transforming growth factor-beta
  • TGF ⁇ 1, TGF ⁇ 2, and TGF ⁇ 3 which are pleotropic modulators of cell growth and differentiation, embryonic and bone development, extracellular matrix formation, hematopoiesis, immune and inflammatory responses.
  • the members of the TGF ⁇ family initiate intracellular signaling pathways leading ultimately to the expression of genes that regulate the cell cycle, control proliferative responses, or relate to extracellular matrix proteins that mediate outside-in cell signaling, cell adhesion, migration and intercellular communication.
  • compounds of the invention that are inhibitors of the TGF ⁇ intracellular signaling pathway are useful treatments for fibroproliferative diseases, including kidney disorders associated with unregulated TGF ⁇ activity and excessive fibrosis including glomerulonephritis (GN), such as mesangial proliferative GN, immune GN, and crescentic GN.
  • GN glomerulonephritis
  • Other renal conditions include diabetic nephropathy, renal interstitial fibrosis, renal fibrosis in transplant patients receiving cyclosporin, and HIV-associated nephropathy.
  • Collagen vascular disorders include progressive systemic sclerosis, polymyositis, scleroderma, dermatomyositis, eosinophilic fascitis, morphea, or those associated with the occurrence of Raynaud's syndrome.
  • Lung fibroses resulting from excessive TGF ⁇ activity include adult respiratory distress syndrome, COPD, idiopathic pulmonary fibrosis, and interstitial pulmonary fibrosis often associated with autoimmune disorders, such as systemic lupus erythematosus and scleroderma, chemical contact, or allergies.
  • Another autoimmune disorder associated with fibroproliferative characteristics is rheumatoid arthritis. Fibroproliferative conditions can be associated with surgical eye procedures. Such procedures include retinal reattachment surgery accompanying proliferative vitreoretinopathy, cataract extraction with intraocular lens implantation, and post glaucoma drainage surgery.
  • the present invention further provides a method for preventing or treating any of the diseases or disorders described above in a subject in need of such treatment, which method comprises administering to said subject a therapeutically effective amount (See, “Administration and Pharmaceutical Compositions”, infra) of a compound of Formula I or a pharmaceutically acceptable salt thereof.
  • a therapeutically effective amount See, “Administration and Pharmaceutical Compositions”, infra
  • the required dosage will vary depending on the mode of administration, the particular condition to be treated and the effect desired.
  • compounds of the invention will be administered in therapeutically effective amounts via any of the usual and acceptable modes known in the art, either singly or in combination with one or more therapeutic agents.
  • a therapeutically effective amount may vary widely depending on the severity of the disease, the age and relative health of the subject, the potency of the compound used and other factors. In general, satisfactory results are indicated to be obtained systemically at daily dosages of from about 0.03 to 2.5 mg/kg per body weight.
  • An indicated daily dosage in the larger mammal, e.g. humans is in the range from about 0.5 mg to about 100 mg, conveniently administered, e.g. in divided doses up to four times a day or in retard form.
  • Suitable unit dosage forms for oral administration comprise from ca. 1 to 50 mg active ingredient.
  • Compounds of the invention can be administered as pharmaceutical compositions by any conventional route, in particular enterally, e.g., orally, e.g., in the form of tablets or capsules, or parenterally, e.g., in the form of injectable solutions or suspensions, topically, e.g., in the form of lotions, gels, ointments or creams, or in a nasal or suppository form.
  • Pharmaceutical compositions comprising a compound of the present invention in free form or in a pharmaceutically acceptable salt form in association with at least one pharmaceutically acceptable carrier or diluent can be manufactured in a conventional manner by mixing, granulating or coating methods.
  • oral compositions can be tablets or gelatin capsules comprising the active ingredient together with a) diluents, e.g., lactose, dextrose, sucrose, mannitol, sorbitol, cellulose and/or glycine; b) lubricants, e.g., silica, talcum, stearic acid, its magnesium or calcium salt and/or polyethyleneglycol; for tablets also c) binders, e.g., magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose and or polyvinylpyrrolidone; if desired d) disintegrants, e.g., starches, agar, alginic acid or its sodium salt, or effervescent mixtures; and/or e) absorbents, colorants, flavors and sweeteners.
  • diluents e.g., lactose, dextrose, sucrose,
  • compositions can be aqueous isotonic solutions or suspensions, and suppositories can be prepared from fatty emulsions or suspensions.
  • the compositions may be sterilized and/or contain adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure and/or buffers. In addition, they may also contain other therapeutically valuable substances.
  • Suitable formulations for transdermal applications include an effective amount of a compound of the present invention with a carrier.
  • a carrier can include absorbable pharmacologically acceptable solvents to assist passage through the skin of the host.
  • transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound to the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin.
  • Matrix transdermal formulations may also be used.
  • Suitable formulations for topical application, e.g., to the skin and eyes, are preferably aqueous solutions, ointments, creams or gels well-known in the art. Such may contain solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives.
  • Compounds of the invention can be administered in therapeutically effective amounts in combination with one or more therapeutic agents (pharmaceutical combinations).
  • therapeutic agents for example, synergistic effects can occur with other immunomodulatory or anti-inflammatory substances, for example when used in combination with cyclosporin, rapamycin, or ascomycin, or immunosuppressant analogues thereof, for example cyclosporin A (CsA), cyclosporin G, FK-506, rapamycin, or comparable compounds, corticosteroids, cyclophosphamide, azathioprine, methotrexate, brequinar, leflunomide, mizoribine, mycophenolic acid, mycophenolate mofetil, 15-deoxyspergualin, immunosuppressant antibodies, especially monoclonal antibodies for leukocyte receptors, for example MHC, CD2, CD3, CD4, CD7, CD25, CD28, B7, CD45, CD58 or their ligands, or other immunomodulatory compounds, such as CT
  • the invention also provides for a pharmaceutical combinations, e.g. a kit, comprising a) a first agent which is a compound of the invention as disclosed herein, in free form or in pharmaceutically acceptable salt form, and b) at least one co-agent.
  • a pharmaceutical combinations e.g. a kit, comprising a) a first agent which is a compound of the invention as disclosed herein, in free form or in pharmaceutically acceptable salt form, and b) at least one co-agent.
  • the kit can comprise instructions for its administration.
  • co-administration or “combined administration” or the like as utilized herein are meant to encompass administration of the selected therapeutic agents to a single patient, and are intended to include treatment regimens in which the agents are not necessarily administered by the same route of administration or at the same time.
  • pharmaceutical combination means a product that results from the mixing or combining of more than one active ingredient and includes both fixed and non-fixed combinations of the active ingredients.
  • fixed combination means that the active ingredients, e.g. a compound of Formula I and a co-agent, are both administered to a patient simultaneously in the form of a single entity or dosage.
  • non-fixed combination means that the active ingredients, e.g. a compound of Formula I and a co-agent, are both administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific time limits, wherein such administration provides therapeutically effective levels of the 2 compounds in the body of the patient.
  • cocktail therapy e.g. the administration of 3 or more active ingredients.
  • the present invention also includes processes for the preparation of compounds of the invention.
  • reactive functional groups for example hydroxy, amino, imino, thio or carboxy groups, where these are desired in the final product, to avoid their unwanted participation in the reactions.
  • Conventional protecting groups can be used in accordance with standard practice, for example, see T. W. Greene and P. G. M. Wuts in “Protective Groups in Organic Chemistry”, John Wiley and Sons, 1991.
  • a compound of the invention (Formula I) can be prepared by reacting a compound of formula 2 with a compound of formula 3 in the presence of a suitable reactant (e.g., ammonium acetate, and the like) and a suitable solvent (e.g., acetic acid, or the like). The reaction is carried out in the temperature range of 100 to 140° C. and can take up to 24 hours to complete.
  • a suitable reactant e.g., ammonium acetate, and the like
  • a suitable solvent e.g., acetic acid, or the like.
  • a compound of the invention can be prepared as a pharmaceutically acceptable acid addition salt by reacting the free base form of the compound with a pharmaceutically acceptable inorganic or organic acid.
  • a pharmaceutically acceptable base addition salt of a compound of the invention can be prepared by reacting the free acid form of the compound with a pharmaceutically acceptable inorganic or organic base.
  • the salt forms of the compounds of the invention can be prepared using salts of the starting materials or intermediates.
  • the free acid or free base forms of the compounds of the invention can be prepared from the corresponding base addition salt or acid addition salt from, respectively.
  • a compound of the invention in an acid addition salt form can be converted to the corresponding free base by treating with a suitable base (e.g., ammonium hydroxide solution, sodium hydroxide, and the like).
  • a suitable base e.g., ammonium hydroxide solution, sodium hydroxide, and the like.
  • a compound of the invention in a base addition salt form can be converted to the corresponding free acid by treating with a suitable acid (e.g., hydrochloric acid, etc.).
  • Compounds of the invention in unoxidized form can be prepared from N-oxides of compounds of the invention by treating with a reducing agent (e.g., sulfur, sulfur dioxide, triphenyl phosphine, lithium borohydride, sodium borohydride, phosphorus trichloride, tribromide, or the like) in a suitable inert organic solvent (e.g. acetonitrile, ethanol, aqueous dioxane, or the like) at 0 to 80° C.
  • a reducing agent e.g., sulfur, sulfur dioxide, triphenyl phosphine, lithium borohydride, sodium borohydride, phosphorus trichloride, tribromide, or the like
  • a suitable inert organic solvent e.g. acetonitrile, ethanol, aqueous dioxane, or the like
  • Prodrug derivatives of the compounds of the invention can be prepared by methods known to those of ordinary skill in the art (e.g., for further details see Saulnier et al., (1994), Bioorganic and Medicinal Chemistry Letters, Vol. 4, p. 1985).
  • appropriate prodrugs can be prepared by reacting a non-derivatized compound of the invention with a suitable carbamoylating agent (e.g., 1,1-acyloxyalkylcarbanochloridate, para-nitrophenyl carbonate, or the like).
  • Protected derivatives of the compounds of the invention can be made by means known to those of ordinary skill in the art. A detailed description of techniques applicable to the creation of protecting groups and their removal can be found in T. W. Greene, “Protecting Groups in Organic Chemistry”, 3 rd edition, John Wiley and Sons, Inc., 1999.
  • Hydrates of compounds of the present invention can be conveniently prepared, or formed during the process of the invention, as solvates (e.g., hydrates). Hydrates of compounds of the present invention can be conveniently prepared by recrystallization from an aqueous/organic solvent mixture, using organic solvents such as dioxin, tetrahydrofuran or methanol.
  • Compounds of the invention can be prepared as their individual stereoisomers by reacting a racemic mixture of the compound with an optically active resolving agent to form a pair of diastereoisomeric compounds, separating the diastereomers and recovering the optically pure enantiomers. While resolution of enantiomers can be carried out using covalent diastereomeric derivatives of the compounds of the invention, dissociable complexes are preferred (e.g., crystalline diastereomeric salts). Diastereomers have distinct physical properties (e.g., melting points, boiling points, solubilities, reactivity, etc.) and can be readily separated by taking advantage of these dissimilarities.
  • the diastereomers can be separated by chromatography, or preferably, by separation/resolution techniques based upon differences in solubility.
  • the optically pure enantiomer is then recovered, along with the resolving agent, by any practical means that would not result in racemization.
  • a more detailed description of the techniques applicable to the resolution of stereoisomers of compounds from their racemic mixture can be found in Jean Jacques, Andre Collet, Samuel H. Wilen, “Enantiomers, Racemates and Resolutions”, John Wiley And Sons, Inc., 1981.
  • the compounds of Formula I can be made by a process, which involves:
  • the present invention is further exemplified, but not limited, by the following examples that illustrate the preparation of compounds of Formula I according to the invention.
  • a mixture of dimethylsulfoxide (1.64 mL, 1.81 g, 23.1 mmol) and anhydrous methylene chloride (100 mL) is cooled to ⁇ 70° C. and oxalyl chloride (2.0 mL, 2.94 g, 23.1 mmol) is added dropwise.
  • the mixture is stirred for 15 minutes at this temperature and a solution of the above diol in methylene chloride (40 mL) and dimethylsulfoxide (10 mL) is added dropwise.
  • the mixture is stirred at ⁇ 60° C. for 2 hours before triethylamine (6.45 mL, 4.68 g, 46.3 mmol) is added.
  • Compounds of the present invention are assayed to measure their capacity to selectively inhibit cell proliferation of 32D cells expressing BCR-Abl (32D-p210) compared with parental 32D cells. Compounds selectively inhibiting the proliferation of these BCR-Abl transformed cells are tested for anti-proliferative activity on Ba/F3 cells expressing either wild type or the mutant forms of Bcr-abl. In addition, compounds are assayed to measure their capacity to inhibit b-Raf.
  • the murine cell line used is the 32D hemopoietic progenitor cell line transformed with BCR-Abl cDNA (32D-p210). These cells are maintained in RPMI/10% fetal calf serum (RPMI/FCS) supplemented with penicillin 50 ⁇ g/mL, streptomycin 50 ⁇ g/mL and L-glutamine 200 mM. Untransformed 32D cells are similarly maintained with the addition of 15% of WEHI conditioned medium as a source of IL3.
  • RPMI/10% fetal calf serum RPMI/FCS
  • Untransformed 32D cells are similarly maintained with the addition of 15% of WEHI conditioned medium as a source of IL3.
  • 50 ⁇ l of a 32D or 32D-p210 cells suspension are plated in Greiner 384 well microplates (black) at a density of 5000 cells per well.
  • 50 nl of test compound (1 mM in DMSO stock solution) is added to each well (STI571 is included as a positive control).
  • the cells are incubated for 72 hours at 37° C., 5% CO 2 .
  • 10 ⁇ l of a 60% Alamar Blue solution (Tek diagnostics) is added to each well and the cells are incubated for an additional 24 hours.
  • the fluorescence intensity (Excitation at 530 nm, Emission at 580 nm) is quantified using the AcquestTM system (Molecular Devices).
  • 32D-p210 cells are plated into 96 well TC plates at a density of 15,000 cells per well. 50 ⁇ L of two fold serial dilutions of the test compound (C max is 40 ⁇ M) are added to each well (STI571 is included as a positive control). After incubating the cells for 48 hours at 37° C., 5% CO 2 , 15 ⁇ L of MTT (Promega) is added to each well and the cells are incubated for an additional 5 hours. The optical density at 570 nm is quantified spectrophotometrically and IC 50 values, the concentration of compound required for 50% inhibition, determined from a dose response curve.
  • BCR-Abl autophosphorylation is quantified with capture Elisa using a c-abl specific capture antibody and an antiphosphotyrosine antibody.
  • 32D-p210 cells are plated in 96 well TC plates at 2 ⁇ 10 5 cells per well in 50 ⁇ L of medium. 50 ⁇ L of two fold serial dilutions of test compounds (C max is 10 ⁇ M) are added to each well (STI571 is included as a positive control). The cells are incubated for 90 minutes at 37° C., 5% CO 2 .
  • the cells are then treated for 1 hour on ice with 150 ⁇ L of lysis buffer (50 mM Tris-HCl, pH 7.4, 150 mM NaCl, 5 mM EDTA, 1 mM EGTA and 1% NP-40) containing protease and phosphatase inhibitors.
  • 50 ⁇ L of cell lysate is added to 96 well optiplates previously coated with anti-abl specific antibody and blocked. The plates are incubated for 4 hours at 4° C. After washing with TBS-Tween 20 buffer, 50 ⁇ L of alkaline-phosphatase conjugated anti-phosphotyrosine antibody is added and the plate is further incubated overnight at 4° C.
  • Test compounds of the invention that inhibit the proliferation of the BCR-Abl expressing cells, inhibit the cellular BCR-Abl autophosphorylation in a dose-dependent manner.
  • the assay is carried out in 384-well MaxiSorp plates (NUNC) with black walls and clear bottom.
  • the substrate, I ⁇ B ⁇ is diluted in DPBS (1:750) and 15 ⁇ l is added to each well.
  • the plates are incubated at 4° C. overnight and washed 3 times with TBST (25 mM Tris, pH 8.0, 150 mM NaCl and 0.05% Tween-20) using the EMBLA plate washer. Plates are blocked by Superblock (15 ⁇ l/well) for 3 hours at room temperature, washed 3 times with TBST and pat-dried.
  • Assay buffer containing 20 ⁇ M ATP (10 ⁇ l) is added to each well followed by 100 nl or 500 nl of compound.
  • B-Raf is diluted in the assay buffer (1 ⁇ l into 25 ⁇ l) and 10 ⁇ l of diluted b-Raf is added to each well (0.4 ⁇ g/well).
  • the plates are incubated at room temperature for 2.5 hours.
  • the kinase reaction is stopped by washing the plates 6 times with TBST.
  • Phosph-I ⁇ B ⁇ (Ser32/36) antibody is diluted in Superblock (1:10,000) and 15 ⁇ l is added to each well. The plates are incubated at 4° C. overnight and washed 6 times with TBST.
  • AP-conjugated goat-anti-mouse IgG is diluted in Superblock (1:1,500) and 15 ⁇ l is added to each well. Plates are incubated at room temperature for 1 hour and washed 6 times with TBST. 15 ⁇ l of Attophos AP substrate is added to each well and plates are incubated at room temperature for 15 minutes. Plates are read on Acquest or Analyst GT using a Fluorescence Intensity Nanxin BBT anion (505 dichroic mirror).
  • kinases are assessed for their ability to inhibit individual members of a panel of kinases (a partial, non-limiting list of kinases includes: Abl, BCR-Abl, EGF-R, c-erbB2 kinase (HER-2), PDGF-R, lck, SAPK2 ⁇ , p38, TGF ⁇ , KDR, c-Kit, b-RAF, c-RAF, FLT1 and FLT4).
  • the compounds are tested in duplicates at a final concentration of 10 ⁇ M following this generic protocol. Note that the kinase buffer composition and the substrates vary for the different kinases included in the “Upstate KinaseProfilerTM” panel.
  • kinase buffer composition and the substrates vary for the different kinases included in the “Upstate KinaseProfilerTM” panel.
  • Kinase buffer (2.5 ⁇ L, 10 ⁇ -containing MnCl 2 when required), active kinase (0.001-0.01 Units; 2.5 ⁇ L), specific or Poly (Glu4-Tyr) peptide (5-500 ⁇ M or 0.01 mg/ml) in kinase buffer and kinase buffer (50 ⁇ M; 5 ⁇ L) are mixed in an eppendorf on ice.
  • a Mg/ATP mix (10 ⁇ L; 67.5 (or 33.75) mM MgCl 2 , 450 (or 225) ⁇ M ATP and 1 ⁇ Ci/ ⁇ l [ ⁇ - 32 P]-ATP (3000 Ci/mmol)) is added and the reaction is incubated at about 30° C. for about 10 minutes.
  • the reaction mixture is spotted (20 ⁇ L) onto a 2 cm ⁇ 2 cm P81 (phosphocellulose, for positively charged peptide substrates) or Whatman No. 1 (for Poly (Glu4-Tyr) peptide substrate) paper square.
  • the assay squares are washed 4 times, for 5 minutes each, with 0.75% phosphoric acid and washed once with acetone for 5 minutes.
  • the assay squares are transferred to a scintillation vial, 5 ml scintillation cocktail are added and 32 P incorporation (cpm) to the peptide substrate is quantified with a Beckman scintillation counter. Percentage inhibition is calculated for each reaction.
  • compounds of Formula I in free form or in pharmaceutically acceptable salt form, exhibit valuable pharmacological properties, for example, as indicated by the in vitro tests described in this application.
  • compounds of Formula I preferably show an IC 50 in the range of 1 ⁇ 10 ⁇ 10 to 1 ⁇ 10 ⁇ 5 M, preferably less than 500 nM for wild type BCR-Abl and b-Raf.
  • 6-(2-phenyl-5-pyridin-4-yl-1H-imidazol-4-yl)-naphthalen-2-ol compound 11
  • IC 50 300 nM and 879 nM against b-Raf, respectively.
  • Compounds of Formula I at a concentration of 10 ⁇ M, preferably show a percentage inhibition of greater than 50%, preferably greater than about 70%, against Abl, BCR-Abl, PDGF-R, lck, SAPK20 ⁇ , p38, TGF ⁇ , KDR, c-Kit, b-RAF, c-RAF, FLT1 and/or FLT4 kinases.
  • 6-(2-Phenyl-5-pyridin-4-yl-1H-imidazol-4-yl)-naphthalen-2-ol (compound 11) at a concentration of 10 ⁇ M, inhibits the following kinases by the percentage shown in brackets (for example, 100% means complete inhibition, 0% means no inhibition): Abl (98%); c-RAF (98%), Lck (61%); and SAPK2 ⁇ (74%).

Abstract

The invention provides a novel class of compounds, pharmaceutical compositions comprising such compounds and methods of using such compounds to treat or prevent diseases or disorders associated with abnormal or deregulated kinase activity, particularly diseases or disorders that involve abnormal activation of the Abl, BCR-Abl, PDGF-R, lck, SAPK2α, p38, TGFβ, KDR, c-Kit, b-RAF, c-RAF, FLT1 and FLT4 kinases.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is U.S. Divisional Patent Application of U.S. patent application Ser. No. 10/956,880, filed Oct. 1, 2004 which claims the benefit of priority under 35 U.S.C. § 119(e) to U.S. Provisional Patent Application No. 60/508,450, filed Oct. 2, 2003. The disclosures of which are incorporated herein by reference in their entirety and for all purposes.
  • BACKGROUND OF THE INVENTION
  • 1. Field of the Invention
  • The invention provides a novel class of compounds, pharmaceutical compositions comprising such compounds and methods of using such compounds to treat or prevent diseases or disorders associated with abnormal or deregulated kinase activity, particularly diseases or disorders that involve abnormal activation of the Abl, BCR-Abl, lck, SAPK2α, PDGF-R, p38, TGFβ, KDR, c-Kit, b-RAF, c-RAF, FLT1 and FLT4 kinases.
  • 2. Background
  • The protein kinases represent a large family of proteins, which play a central role in the regulation of a wide variety of cellular processes and maintaining control over cellular function. A partial, non-limiting, list of these kinases include: receptor tyrosine kinases such as platelet-derived growth factor receptor kinase (PDGF-R), TGFβ, VEGF-receptor kinase (e.g. KDR, Flt-1 and Flt-4), the receptor kinase for stem cell factor, c-kit; non-receptor tyrosine kinases such Abl and the fusion kinase BCR-Abl; and serine/threonine kinases such as p38, b-RAF and c-RAF. Aberrant kinase activity has been observed in many disease states including benign and malignant proliferative disorders as well as diseases resulting from inappropriate activation of the immune and nervous systems.
  • The novel compounds of this invention inhibit the activity of one or more protein kinases and are, therefore, expected to be useful in the treatment of kinase-associated diseases.
  • SUMMARY OF THE INVENTION
  • In one aspect, the present invention provides compounds of Formula I:
  • Figure US20090258910A1-20091015-C00001
  • in which:
  • R1 and R2 are independently selected from hydrogen, —XR3, —XC(O)R3, —XSR3, —XS(O)R3, —XS(O)2R3, —XOR4, —XNC(O)NHR3R4 and —XOC(O)NR3R4; wherein X is a bond or C1-4alkylene; R4 is selected from hydrogen and C1-6alkyl; R3 is selected from C6-10aryl, C5-10heteroaryl, C3-12cycloalkyl and C3-8heterocycloalkyl; wherein any aryl, heteroaryl, cycloalkyl or heterocycloalkyl of R3 is optionally substituted by 1 to 3 radicals selected from hydroxy, halo, nitro, C1-6alkyl, C1-6alkoxy, halo-substituted-C1-6alkyl, halo-substituted-C1-6alkoxy, —XC(O)OR4 and —NR4R5; wherein X is a bond or C1-4alkylene and R4 and R5 are independently selected from hydrogen and C1-6alkyl; wherein the pyridinyl ring A can have up to three —C═ groups replaced with —N═ groups; wherein the naphthyl ring B can have up to four —C═ groups replaced with —N═ groups; and the N-oxide derivatives, prodrug derivatives, protected derivatives, individual isomers and mixture of isomers thereof; and the pharmaceutically acceptable salts and solvates (e.g. hydrates) of such compounds.
  • In a second aspect, the present invention provides a pharmaceutical composition which contains a compound of Formula I or a N-oxide derivative, individual isomers and mixture of isomers thereof; or a pharmaceutically acceptable salt thereof, in admixture with one or more suitable excipients.
  • In a third aspect, the present invention provides a method of treating a disease in an animal in which inhibition of kinase activity, particularly Abl, BCR-Abl, PDGF-R, lck, SAPK2α, p38, TGFβ, KDR, c-Kit, b-RAF, c-RAF, FLT1 and FLT4 activity, can prevent, inhibit or ameliorate the pathology and/or symptomology of the diseases, which method comprises administering to the animal a therapeutically effective amount of a compound of Formula I or a N-oxide derivative, individual isomers and mixture of isomers thereof, or a pharmaceutically acceptable salt thereof.
  • In a fourth aspect, the present invention provides the use of a compound of Formula I in the manufacture of a medicament for treating a disease in an animal in which kinase activity, particularly Abl, BCR-Abl, PDGF-R, lck, SAPK2α, p38, TGFβ, KDR, c-Kit, b-RAF, c-RAF, FLT1 and FLT4 activity, contributes to the pathology and/or symptomology of the disease.
  • In a fifth aspect, the present invention provides a process for preparing compounds of Formula I and the N-oxide derivatives, prodrug derivatives, protected derivatives, individual isomers and mixture of isomers thereof, and the pharmaceutically acceptable salts thereof.
  • DETAILED DESCRIPTION OF THE INVENTION Definitions
  • “Alkyl” as a group and as a structural element of other groups, for example halo-substituted-alkyl and alkoxy, can be either straight-chained or branched. C1-4-alkoxy includes, methoxy, ethoxy, and the like. Halo-substituted alkyl includes trifluoromethyl, pentafluoroethyl, and the like.
  • “Aryl” means a monocyclic or fused bicyclic aromatic ring assembly containing six to ten ring carbon atoms. For example, aryl may be phenyl or naphthyl, preferably phenyl. “Arylene” means a divalent radical derived from an aryl group. “Heteroaryl” is as defined for aryl where one or more of the ring members are a heteroatom. For example heteroaryl includes pyridyl, indolyl, indazolyl, quinoxalinyl, quinolinyl, benzofuranyl, benzopyranyl, benzothiopyranyl, benzo[1,3]dioxole, imidazolyl, benzo-imidazolyl, pyrimidinyl, furanyl, oxazolyl, isoxazolyl, triazolyl, tetrazolyl, pyrazolyl, thienyl, etc.
  • “Cycloalkyl” means a saturated or partially unsaturated, monocyclic, fused bicyclic or bridged polycyclic ring assembly containing the number of ring atoms indicated. For example, C3-10cycloalkyl includes cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, etc. “Heterocycloalkyl” means cycloalkyl, as defined in this application, provided that one or more of the ring carbons indicated, are replaced by a moiety selected from —O—, —N═, —NR—, —C(O)—, —S—, —S(O)— or —S(O)2—, wherein R is hydrogen, C1-4alkyl or a nitrogen protecting group. For example, C3-8heterocycloalkyl as used in this application to describe compounds of the invention includes morpholino, pyrrolidinyl, piperazinyl, piperidinyl, piperidinylone, 1,4-dioxa-8-aza-spiro[4.5]dec-8-yl, etc.
  • “Halogen” (or halo) preferably represents chloro or fluoro, but may also be bromo or iodo.
  • “Treat”, “treating” and “treatment” refer to a method of alleviating or abating a disease and/or its attendant symptoms.
  • DESCRIPTION OF THE PREFERRED EMBODIMENTS
  • The present invention provides compounds, compositions and methods for the treatment of kinase related disease, particularly Abl, BCR-Abl, PDGF-R, lck, SAPK2α, p38, TGFβ, KDR, c-Kit, b-RAF, c-RAF, FLT1 and FLT4 kinase related diseases. For example, leukemia and other proliferation disorders related to BCR-Abl can be treated through the inhibition of wild type and mutant forms of Bcr-Abl.
  • In one embodiment, with reference to compounds of Formula I, R1 and R2 are independently selected from hydrogen, —XR3 and —XOC(O)NR3R4; wherein X is a bond or C1-4alkylene; R4 is selected from hydrogen and C1-6alkyl; R3 is selected from C6-10aryl, C5-10heteroaryl and C3-8heterocycloalkyl; wherein any aryl, heteroaryl or heterocycloalkyl of R3 is optionally substituted by 1 to 3 radicals selected from halo, nitro, C1-6alkyl, C1-6alkoxy, halo-substituted-C1-6alkyl, —XC(O)OR4 and —NR4R5; wherein X is a bond or C1-4alkylene and R4 and R5 are independently selected from hydrogen and C1-6alkyl.
  • In another embodiment, R1 is selected from hydrogen and phenyl; wherein said phenyl is optionally substituted by 1 to 3 radicals selected from trifluoromethyl, dimethylamino, methoxy, halo, ethoxy and nitro.
  • In a further embodiment, R2 is selected from hydrogen, —OH and —OC(O)NHR4; wherein R4 is selected from phenyl and benzo[1,3]dioxol-5-yl; wherein said phenyl of R4 is optionally substituted by 1 to 3 radicals selected from —C(O)OCH3 and dimethylamino.
  • Preferred compounds of Formula I are selected from: Phenyl-carbamic acid 8-(2-phenyl-5-pyridin-4-yl-3H-imidazol-4-yl)-naphthalen-2-yl ester; 4-(5-Naphthalen-1-yl-2-phenyl-3H-imidazol-4-yl)-pyridine; 4-[2-(3,5-Bis-trifluoromethyl-phenyl)-5-naphthalen-1-yl-3H-imidazol-4-yl]-pyridine; Dimethyl-[4-(4-naphthalen-1-yl-5-pyridin-4-yl-1H-imidazol-2-yl)-phenyl]-amine; 4-[5-Naphthalen-1-yl-2-(2,4,6-trimethoxy-phenyl)-3H-imidazol-4-yl]-pyridine; 4-[2-(2-Fluoro-phenyl)-5-naphthalen-1-yl-3H-imidazol-4-yl]-pyridine; 4-[2-(2-Ethoxy-phenyl)-5-naphthalen-1-yl-3H-imidazol-4-yl]-pyridine; 4-[5-Naphthalen-1-yl-2-(3-nitro-phenyl)-3H-imidazol-4-yl]-pyridine; 4-(5-Naphthalen-1-yl-3H-imidazol-4-yl)-pyridine; Phenyl-carbamic acid 6-(2-phenyl-5-pyridin-4-yl-1H-imidazol-4-yl)-naphthalen-2-yl ester; 6-(2-Phenyl-5-pyridin-4-yl-1H-imidazol-4-yl)-naphthalen-2-ol; 4-[6-(2-Phenyl-5-pyridin-4-yl-1H-imidazol-4-yl)-naphthalen-2-yloxycarbonylamino]-benzoic acid methyl ester; Benzo[1,3]dioxol-5-yl-carbamic acid 6-(2-phenyl-5-pyridin-4-yl-1H-imidazol-4-yl)-naphthalen-2-yl ester; 5-[6-(2-Phenyl-5-pyridin-4-yl-1H-imidazol-4-yl)-naphthalen-2-yloxycarbonylamino]-isophthalic acid dimethyl ester; (4-Dimethylamino-phenyl)-carbamic acid 6-(2-phenyl-5-pyridin-4-yl-1H-imidazol-4-yl)-naphthalen-2-yl ester; Benzo[1,3]dioxol-5-yl-carbamic acid 8-(2-phenyl-5-pyridin-4-yl-1H-imidazol-4-yl)-naphthalen-2-yl ester; 5-[8-(2-Phenyl-5-pyridin-4-yl-1H-imidazol-4-yl)-naphthalen-2-yloxycarbonylamino]-isophthalic acid dimethyl ester; and (4-Dimethylamino-phenyl)-carbamic acid 8-(2-phenyl-5-pyridin-4-yl-1H-imidazol-4-yl)-naphthalen-2-yl ester.
  • Further preferred compounds of Formula I are detailed in the Examples and Table I, infra.
  • Pharmacology and Utility
  • Compounds of the invention modulate the activity of protein tyrosine kinases and, as such, are useful for treating diseases or disorders in which protein tyrosine kinases, particularly Abl, BCR-Abl, PDGF-R, lck, SAPK2α, p38, TGFβ, KDR, c-Kit, b-RAF, c-RAF, FLT1 and FLT4 kinases, contribute to the pathology and/or symptomology of the disease.
  • Abelson tyrosine kinase (i.e. Abl, c-Abl) is involved in the regulation of the cell cycle, in the cellular response to genotoxic stress, and in the transmission of information about the cellular environment through integrin signaling. Overall, it appears that the Abl protein serves a complex role as a cellular module that integrates signals from various extracellular and intracellular sources and that influences decisions in regard to cell cycle and apoptosis. Abelson tyrosine kinase includes sub-types derivatives such as the chimeric fusion (oncoprotein) BCR-Abl with deregulated tyrosine kinase activity or the v-Abl. BCR-Abl is critical in the pathogenesis of 95% of chronic myelogenous leukemia (CML) and 10% of acute lymphocytic leukemia. STI-571 (Gleevec) is an inhibitor of the oncogenic BCR-Abl tyrosine kinase and is used for the treatment of chronic myeloid leukemia (CML). However, some patients in the blast crisis stage of CML are resistant to STI-571 due to mutations in the BCR-Abl kinase. Over 22 mutations have been reported to date with the most common being G250E, E255V, T3151, F317L and M351T.
  • Compounds of the present invention inhibit abl kinase, especially v-abl kinase. The compounds of the present invention also inhibit wild-type BCR-Abl kinase and mutations of BCR-Abl kinase and are thus suitable for the treatment of Bcr-abl-positive cancer and tumor diseases, such as leukemias (especially chronic myeloid leukemia and acute lymphoblastic leukemia, where especially apoptotic mechanisms of action are found), and also shows effects on the subgroup of leukemic stem cells as well as potential for the purification of these cells in vitro after removal of said cells (for example, bone marrow removal) and reimplantation of the cells once they have been cleared of cancer cells (for example, reimplantation of purified bone marrow cells).
  • PDGF (Platelet-derived Growth Factor) is a very commonly occurring growth factor, which plays an important role both in normal growth and also in pathological cell proliferation, such as is seen in carcinogenesis and in diseases of the smooth-muscle cells of blood vessels, for example in atherosclerosis and thrombosis. Compounds of the invention can inhibit PDGF receptor (PDGFR) activity and are, therefore, suitable for the treatment of tumor diseases, such as gliomas, sarcomas, prostate tumors, and tumors of the colon, breast, and ovary.
  • Compounds of the present invention, can be used not only as a tumor-inhibiting substance, for example in small cell lung cancer, but also as an agent to treat non-malignant proliferative disorders, such as atherosclerosis, thrombosis, psoriasis, scleroderma and fibrosis, as well as for the protection of stem cells, for example to combat the hemotoxic effect of chemotherapeutic agents, such as 5-fluoruracil, and in asthma. Compounds of the invention can especially be used for the treatment of diseases, which respond to an inhibition of the PDGF receptor kinase.
  • Compounds of the present invention show useful effects in the treatment of disorders arising as a result of transplantation, for example, allogenic transplantation, especially tissue rejection, such as especially obliterative bronchiolitis (OB), i.e. a chronic rejection of allogenic lung transplants. In contrast to patients without OB, those with OB often show an elevated PDGF concentration in bronchioalveolar lavage fluids.
  • Compounds of the present invention are also effective in diseases associated with vascular smooth-muscle cell migration and proliferation (where PDGF and PDGF-R often also play a role), such as restenosis and atherosclerosis. These effects and the consequences thereof for the proliferation or migration of vascular smooth-muscle cells in vitro and in vivo can be demonstrated by administration of the compounds of the present invention, and also by investigating its effect on the thickening of the vascular intima following mechanical injury in vivo.
  • The compounds of the present invention also inhibit cellular processes involving stem-cell factor (SCF, also known as the c-kit ligand or steel factor), such as inhibiting SCF receptor (kit) autophosphorylation and SCF-stimulated activation of MAPK kinase (mitogen-activated protein kinase). MO7e cells are a human promegakaryocytic leukemia cell line, which depends on SCF for proliferation. Compounds of the invention can inhibit the autophosphorylation of SCF receptors.
  • The Ras-Raf-MEK-ERK signaling pathway mediates cellular response to growth signals. Ras is mutated to an oncogenic form in ˜15% of human cancer. The Raf family belongs to the serine/threonine protein kinase and it includes three members, A-Raf, B-Raf and c-Raf (or Raf-1). The focus on Raf being a drug target has centered on the relationship of Raf as a downstream effector of Ras. However, recent data suggests that B-Raf may have a prominent role in the formation of certain tumors with no requirement for an activated Ras allele (Nature 417, 949-954 (1 Jul. 2002). In particular, B-Raf mutations have been detected in a large percentage of malignant melanomas.
  • Existing medical treatments for melanoma are limited in their effectiveness, especially for late stage melanomas. The compounds of the present invention also inhibit cellular processes involving b-Raf kinase, providing a new therapeutic opportunity for treatment of human cancers, especially for melanoma.
  • The compounds of the present invention also inhibit cellular processes involving c-Raf kinase. c-Raf is activated by the ras oncogene, which is mutated in a wide number of human cancers. Therefore inhibition of the kinase activity of c-Raf may provide a way to prevent ras mediated tumor growth [Campbell, S. L., Oncogene, 17, 1395 (1998)].
  • The compounds of the present invention also inhibit cellular processes involving KDR, Flt-1 and Flt-4. A number of diseases are known which are associated with deregulated angiogenesis, for example diseases caused by ocular neovascularisation, especially retinopathies (diabetic retinopathy, age-related macular degeneration); psoriasis; haemangioblastomas, such as “strawberry-marks” (=haemangioma); various inflammatory diseases, such as arthritis, especially rheumatoid arthritis, arterial atherosclerosis and atherosclerosis occurring after transplants, endometriosis or chronic asthma; and, especially, tumor diseases (solid tumors, but also leukemias and other liquid tumors, since many primitive blood cells and leukemia cells express c-kit, KDR, Flt-1 and Flt-4). Flt-4 is expressed in developing lymphatic vessels. Only the lymphatic endothelia and some high endothelial venules express Flt4 mRNA in adult human tissues and increased expression occurs in lymphatic sinuses in metastatic lymph nodes and in lymphangioma. Inhibition of KDR-mediated functional effects by inhibiting KDR's catalytic activity is considered to be an important therapeutic strategy in the treatment of angiogenized disease states including cancer.
  • Multiple forms of p38 MAPK (α, β, γ, δ), each encoded by a separate gene, form part of a kinase cascade involved in the response of cells to a variety of stimuli, including osmotic stress, UV light and cytokine mediated events. These four isoforms of p38 are thought to regulate different aspects of intracellular signaling. Its activation is part of a cascade of signaling events that lead to the synthesis and production of pro-inflammatory cytokines like TNFα. P38 functions by phosphorylating downstream substrates that include other kinases and transcription factors. Agents that inhibit p38 kinase have been shown to block the production of cytokines including but not limited to TNFα, IL-6, IL-8 and IL-1β. Peripheral blood monocytes (PBMCs) have been shown to express and secrete pro-inflammatory cytokines when stimulated with lipopolysaccharide (LPS) in vitro. P38 inhibitors efficiently block this effect when PBMCs are pretreated with such compounds prior to stimulation with LPS. P38 inhibitors are efficacious in animal models of inflammatory disease. The destructive effects of many disease states are caused by the over production of pro-inflammatory cytokines. The ability of p38 inhibitors to regulate this overproduction makes them useful as disease modifying agents.
  • Molecules that block p38's function have been shown to be effective in inhibiting bone resorption, inflammation, and other immune and inflammation-based pathologies. Thus, a safe and effective p38 inhibitor would provide a means to treat debilitating diseases that can be regulated by modulation of p38 signaling like, for example, RA. Therefore, compounds of the invention that inhibit p38 activity are useful for the treatment of inflammation, osteoarthritis, rheumatoid arthritis, cancer, autoimmune diseases, and for the treatment of other cytokine mediated diseases.
  • Transforming growth factor-beta (TGFβ) denotes a superfamily of proteins that includes, for example, TGFβ1, TGFβ2, and TGFβ3, which are pleotropic modulators of cell growth and differentiation, embryonic and bone development, extracellular matrix formation, hematopoiesis, immune and inflammatory responses. The members of the TGFβ family initiate intracellular signaling pathways leading ultimately to the expression of genes that regulate the cell cycle, control proliferative responses, or relate to extracellular matrix proteins that mediate outside-in cell signaling, cell adhesion, migration and intercellular communication. Consequently, compounds of the invention that are inhibitors of the TGFβ intracellular signaling pathway are useful treatments for fibroproliferative diseases, including kidney disorders associated with unregulated TGFβ activity and excessive fibrosis including glomerulonephritis (GN), such as mesangial proliferative GN, immune GN, and crescentic GN. Other renal conditions include diabetic nephropathy, renal interstitial fibrosis, renal fibrosis in transplant patients receiving cyclosporin, and HIV-associated nephropathy. Collagen vascular disorders include progressive systemic sclerosis, polymyositis, scleroderma, dermatomyositis, eosinophilic fascitis, morphea, or those associated with the occurrence of Raynaud's syndrome. Lung fibroses resulting from excessive TGFβ activity include adult respiratory distress syndrome, COPD, idiopathic pulmonary fibrosis, and interstitial pulmonary fibrosis often associated with autoimmune disorders, such as systemic lupus erythematosus and scleroderma, chemical contact, or allergies. Another autoimmune disorder associated with fibroproliferative characteristics is rheumatoid arthritis. Fibroproliferative conditions can be associated with surgical eye procedures. Such procedures include retinal reattachment surgery accompanying proliferative vitreoretinopathy, cataract extraction with intraocular lens implantation, and post glaucoma drainage surgery.
  • In accordance with the foregoing, the present invention further provides a method for preventing or treating any of the diseases or disorders described above in a subject in need of such treatment, which method comprises administering to said subject a therapeutically effective amount (See, “Administration and Pharmaceutical Compositions”, infra) of a compound of Formula I or a pharmaceutically acceptable salt thereof. For any of the above uses, the required dosage will vary depending on the mode of administration, the particular condition to be treated and the effect desired.
  • Administration and Pharmaceutical Compositions
  • In general, compounds of the invention will be administered in therapeutically effective amounts via any of the usual and acceptable modes known in the art, either singly or in combination with one or more therapeutic agents. A therapeutically effective amount may vary widely depending on the severity of the disease, the age and relative health of the subject, the potency of the compound used and other factors. In general, satisfactory results are indicated to be obtained systemically at daily dosages of from about 0.03 to 2.5 mg/kg per body weight. An indicated daily dosage in the larger mammal, e.g. humans, is in the range from about 0.5 mg to about 100 mg, conveniently administered, e.g. in divided doses up to four times a day or in retard form. Suitable unit dosage forms for oral administration comprise from ca. 1 to 50 mg active ingredient.
  • Compounds of the invention can be administered as pharmaceutical compositions by any conventional route, in particular enterally, e.g., orally, e.g., in the form of tablets or capsules, or parenterally, e.g., in the form of injectable solutions or suspensions, topically, e.g., in the form of lotions, gels, ointments or creams, or in a nasal or suppository form. Pharmaceutical compositions comprising a compound of the present invention in free form or in a pharmaceutically acceptable salt form in association with at least one pharmaceutically acceptable carrier or diluent can be manufactured in a conventional manner by mixing, granulating or coating methods. For example, oral compositions can be tablets or gelatin capsules comprising the active ingredient together with a) diluents, e.g., lactose, dextrose, sucrose, mannitol, sorbitol, cellulose and/or glycine; b) lubricants, e.g., silica, talcum, stearic acid, its magnesium or calcium salt and/or polyethyleneglycol; for tablets also c) binders, e.g., magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose and or polyvinylpyrrolidone; if desired d) disintegrants, e.g., starches, agar, alginic acid or its sodium salt, or effervescent mixtures; and/or e) absorbents, colorants, flavors and sweeteners. Injectable compositions can be aqueous isotonic solutions or suspensions, and suppositories can be prepared from fatty emulsions or suspensions. The compositions may be sterilized and/or contain adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure and/or buffers. In addition, they may also contain other therapeutically valuable substances. Suitable formulations for transdermal applications include an effective amount of a compound of the present invention with a carrier. A carrier can include absorbable pharmacologically acceptable solvents to assist passage through the skin of the host. For example, transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound to the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin. Matrix transdermal formulations may also be used. Suitable formulations for topical application, e.g., to the skin and eyes, are preferably aqueous solutions, ointments, creams or gels well-known in the art. Such may contain solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives.
  • Compounds of the invention can be administered in therapeutically effective amounts in combination with one or more therapeutic agents (pharmaceutical combinations). For example, synergistic effects can occur with other immunomodulatory or anti-inflammatory substances, for example when used in combination with cyclosporin, rapamycin, or ascomycin, or immunosuppressant analogues thereof, for example cyclosporin A (CsA), cyclosporin G, FK-506, rapamycin, or comparable compounds, corticosteroids, cyclophosphamide, azathioprine, methotrexate, brequinar, leflunomide, mizoribine, mycophenolic acid, mycophenolate mofetil, 15-deoxyspergualin, immunosuppressant antibodies, especially monoclonal antibodies for leukocyte receptors, for example MHC, CD2, CD3, CD4, CD7, CD25, CD28, B7, CD45, CD58 or their ligands, or other immunomodulatory compounds, such as CTLA41g. Where the compounds of the invention are administered in conjunction with other therapies, dosages of the co-administered compounds will of course vary depending on the type of co-drug employed, on the specific drug employed, on the condition being treated and so forth.
  • The invention also provides for a pharmaceutical combinations, e.g. a kit, comprising a) a first agent which is a compound of the invention as disclosed herein, in free form or in pharmaceutically acceptable salt form, and b) at least one co-agent. The kit can comprise instructions for its administration.
  • The terms “co-administration” or “combined administration” or the like as utilized herein are meant to encompass administration of the selected therapeutic agents to a single patient, and are intended to include treatment regimens in which the agents are not necessarily administered by the same route of administration or at the same time.
  • The term “pharmaceutical combination” as used herein means a product that results from the mixing or combining of more than one active ingredient and includes both fixed and non-fixed combinations of the active ingredients. The term “fixed combination” means that the active ingredients, e.g. a compound of Formula I and a co-agent, are both administered to a patient simultaneously in the form of a single entity or dosage. The term “non-fixed combination” means that the active ingredients, e.g. a compound of Formula I and a co-agent, are both administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific time limits, wherein such administration provides therapeutically effective levels of the 2 compounds in the body of the patient. The latter also applies to cocktail therapy, e.g. the administration of 3 or more active ingredients.
  • Processes for Making Compounds of the Invention
  • The present invention also includes processes for the preparation of compounds of the invention. In the reactions described, it can be necessary to protect reactive functional groups, for example hydroxy, amino, imino, thio or carboxy groups, where these are desired in the final product, to avoid their unwanted participation in the reactions. Conventional protecting groups can be used in accordance with standard practice, for example, see T. W. Greene and P. G. M. Wuts in “Protective Groups in Organic Chemistry”, John Wiley and Sons, 1991.
  • Compounds of Formula I can be prepared by proceeding as in the following Reaction Scheme I:
  • Figure US20090258910A1-20091015-C00002
  • in which A, B, R1 and R2 are as defined for Formula I in the Summary of the Invention.
  • A compound of the invention (Formula I) can be prepared by reacting a compound of formula 2 with a compound of formula 3 in the presence of a suitable reactant (e.g., ammonium acetate, and the like) and a suitable solvent (e.g., acetic acid, or the like). The reaction is carried out in the temperature range of 100 to 140° C. and can take up to 24 hours to complete. A detailed description of the synthesis of a compound of Formula I is set forth in the examples, infra.
  • Additional Processes for Making Compounds of the Invention
  • A compound of the invention can be prepared as a pharmaceutically acceptable acid addition salt by reacting the free base form of the compound with a pharmaceutically acceptable inorganic or organic acid. Alternatively, a pharmaceutically acceptable base addition salt of a compound of the invention can be prepared by reacting the free acid form of the compound with a pharmaceutically acceptable inorganic or organic base. Alternatively, the salt forms of the compounds of the invention can be prepared using salts of the starting materials or intermediates.
  • The free acid or free base forms of the compounds of the invention can be prepared from the corresponding base addition salt or acid addition salt from, respectively. For example a compound of the invention in an acid addition salt form can be converted to the corresponding free base by treating with a suitable base (e.g., ammonium hydroxide solution, sodium hydroxide, and the like). A compound of the invention in a base addition salt form can be converted to the corresponding free acid by treating with a suitable acid (e.g., hydrochloric acid, etc.).
  • Compounds of the invention in unoxidized form can be prepared from N-oxides of compounds of the invention by treating with a reducing agent (e.g., sulfur, sulfur dioxide, triphenyl phosphine, lithium borohydride, sodium borohydride, phosphorus trichloride, tribromide, or the like) in a suitable inert organic solvent (e.g. acetonitrile, ethanol, aqueous dioxane, or the like) at 0 to 80° C.
  • Prodrug derivatives of the compounds of the invention can be prepared by methods known to those of ordinary skill in the art (e.g., for further details see Saulnier et al., (1994), Bioorganic and Medicinal Chemistry Letters, Vol. 4, p. 1985). For example, appropriate prodrugs can be prepared by reacting a non-derivatized compound of the invention with a suitable carbamoylating agent (e.g., 1,1-acyloxyalkylcarbanochloridate, para-nitrophenyl carbonate, or the like).
  • Protected derivatives of the compounds of the invention can be made by means known to those of ordinary skill in the art. A detailed description of techniques applicable to the creation of protecting groups and their removal can be found in T. W. Greene, “Protecting Groups in Organic Chemistry”, 3rd edition, John Wiley and Sons, Inc., 1999.
  • Compounds of the present invention can be conveniently prepared, or formed during the process of the invention, as solvates (e.g., hydrates). Hydrates of compounds of the present invention can be conveniently prepared by recrystallization from an aqueous/organic solvent mixture, using organic solvents such as dioxin, tetrahydrofuran or methanol.
  • Compounds of the invention can be prepared as their individual stereoisomers by reacting a racemic mixture of the compound with an optically active resolving agent to form a pair of diastereoisomeric compounds, separating the diastereomers and recovering the optically pure enantiomers. While resolution of enantiomers can be carried out using covalent diastereomeric derivatives of the compounds of the invention, dissociable complexes are preferred (e.g., crystalline diastereomeric salts). Diastereomers have distinct physical properties (e.g., melting points, boiling points, solubilities, reactivity, etc.) and can be readily separated by taking advantage of these dissimilarities. The diastereomers can be separated by chromatography, or preferably, by separation/resolution techniques based upon differences in solubility. The optically pure enantiomer is then recovered, along with the resolving agent, by any practical means that would not result in racemization. A more detailed description of the techniques applicable to the resolution of stereoisomers of compounds from their racemic mixture can be found in Jean Jacques, Andre Collet, Samuel H. Wilen, “Enantiomers, Racemates and Resolutions”, John Wiley And Sons, Inc., 1981.
  • In summary, the compounds of Formula I can be made by a process, which involves:
  • (a) that of reaction schemes I; and
  • (b) optionally converting a compound of the invention into a pharmaceutically acceptable salt;
  • (c) optionally converting a salt form of a compound of the invention to a non-salt form;
  • (d) optionally converting an unoxidized form of a compound of the invention into a pharmaceutically acceptable N-oxide;
  • (e) optionally converting an N-oxide form of a compound of the invention to its unoxidized form;
  • (f) optionally resolving an individual isomer of a compound of the invention from a mixture of isomers;
  • (g) optionally converting a non-derivatized compound of the invention into a pharmaceutically acceptable prodrug derivative; and
  • (h) optionally converting a prodrug derivative of a compound of the invention to its non-derivatized form.
  • Insofar as the production of the starting materials is not particularly described, the compounds are known or can be prepared analogously to methods known in the art or as disclosed in the Examples hereinafter.
  • One of skill in the art will appreciate that the above transformations are only representative of methods for preparation of the compounds of the present invention, and that other well known methods can similarly be used.
  • EXAMPLES
  • The present invention is further exemplified, but not limited, by the following examples that illustrate the preparation of compounds of Formula I according to the invention.
  • Example 1 Phenyl-carbamic acid 8-(2-phenyl-5-pyridin-4-yl-3H-imidazol-4-yl)-naphthalen-2-yl ester
  • Figure US20090258910A1-20091015-C00003
  • A mixture of 8-amino-2-naphthol (14.63 g, 91.9 mmol) and di-tert-butyl dicarbonate (21.0 g, 96.2 mmol) in methylene chloride (200 mL) and tetrahydrofuran (200 mL) is stirred under reflux condition for 24 hours. The mixture is then cooled to ambient temperature, filtered and the filtrate is concentrated to dryness. Silica gel chromatography (CH2Cl2/EtOEt=5/95 to 10/90) afforded (7-hydroxy-naphthalen-1-yl)-carbamic acid tert-butyl ester (21.50 g, 90% yield); MS m/z (M+Na+) 282.05.
  • To a mixture of (7-hydroxy-naphthalen-1-yl)-carbamic acid tert-butyl ester (5.11 g, 19.7 mmol) and potassium carbonate (3.26 g, 23.6 mmol) in N,N-dimethylformamide is added benzyl bromide (3 mL, 4.31 g, 25.2 mmol) slowly. The mixture is then stirred at room temperature over night and poured into ice-water (150 mL). The resulting mixture is extracted with ethyl acetate (4×100 mL) and the organic portions are combined, washed with water and brine, dried over sodium sulfate, concentrated and purified by silica gel chromatography (Hexanes/EtOAc=15/1) to give (7-benzyloxy-naphthalen-1-yl)-carbamic acid tert-butyl ester (5.93 g, 86.3%); 1H NMR 400 Hz (CDCl3) δ 1.60 (s, 9H), 5.22 (s, 2H), 6.63 (br, 1H), 7.27 (m, 2H), 7.38 (m, 2H), 7.45 (m, 2H), 7.44 (d, J=7.44 Hz, 2H), 7.61 (d, J=8.11 Hz, 1H), 7.80 (m, 2H); MS m/z (M+Na+) 372.10.
  • To a solution of (7-benzyloxy-naphthalen-1-yl)-carbamic acid tert-butyl ester (9.89 g, 28.3 mmol) in methylene chloride (200 mL) is added trifluoroacetic acid (50 mL, 73 g, 649 mmol) and the mixture is stirred at room temperature for 30 minutes. The solvent and excessive trifluoroacetic acid is evaporated under reduced pressure and the crude product is dried on high vacuum over night and the resulting 7-benzyloxy-naphthalen-1-ylamine (6.98 g, 99%) was used for next step without further purification.
  • A solution of 7-benzyloxy-naphthalen-1-ylamine (6.98 g, 28.02 mmol) in concentrated hydrogen chloride (30 mL), water (30 mL) and tetrahydrofuran (30 mL) is cooled with an ice-salt (sodium chloride) bath. A solution of sodium nitrite (2.32 g, 33.6 mmol) in water (20 mL) is then added in a dropwise manner over a 15-minute period of time and the reaction temperature is kept under 5° C. The mixture is stirred for another 30 minutes before potassium iodide (9.3 g, 56.04 mmol) in water (30 mL) is added. The resulting mixture is stirred for three hours at room temperature before being extracted with ethyl acetate, dried over sodium sulfate, filtered, concentrated and purified by silica gel chromatography (Hexanes/CH2Cl2=25/1-20/1) to give 7-benzyloxy-1-iodo-naphthalene (6.26 g, 61%); MS m/z (M+Na+) 361.00.
  • To a solution of 7-benzyloxy-1-iodo-naphthalene (4.60 g, 12.77 mmol) in ether (120 mL) is added 1.7 M tert-butyl-lithium in pentane (8.26 mL, 14.0 mmol) at −78° C. under nitrogen atmosphere and the resulting mixture is stirred for one hour while being warmed up to 5° C. The mixture is cooled to −78° C. again before tetramethylethylene-diamine (1.93 mL, 1.49 g, 12.79 mmol) is added. The mixture is stirred for 10 minutes before N,N-dimethylformamide (0.99 mL, 0.93 g, 12.78 mmol) is added. The mixture is gradually warmed up to room temperature and stirred over night. Ethyl acetate is added to reaction mixture, washed with saturated ammonium chloride, dried over sodium sulfate, filtered and concentrated. The crude product is purified by silica gel chromatography (Hexanes/EtOAc=20/1) to give 7-benzyloxy-naphthalene-1-carbaldehyde (1.95 g, 58.3% yield); 1H NMR 400 Hz (CDCl3) δ 5.27 (s, 2H), 7.33 (m, 2H), 7.42 (m, 2H), 7.52 (m, 3H), 7.83 (d, J=8.98 Hz, 1H), 7.95 (d, J=7.11 Hz, 1H), 8.03 (d, J=8.10 Hz, 1H), 10.32 (s, 1H); MS m/z (M+H+) 263.05.
  • To a solution of 4-(tert-butyl-dimethyl-silanyloxymethyl)-pyridine (1.59 g, 7.09 mmol) in tetrahydrofuran (40 mL) is added 2M lithium diisopropylamide in hexanes (3.55 mL, 7.11 mmol) over 5 minutes at −50° C. under a nitrogen atmosphere. The mixture is stirred at −40° C. for one hour and 7-benzyloxy-naphthalene-1-carbaldehyde (1.69 g, 6.45 mmol) in tetrahydrofuran (20 mL) is added over 10 minutes. The reaction is gradually warmed up to room temperature and stirred at that temperature over night before being quenched by saturated sodium bicarbonate and extracted with ethyl acetate. The organic layers are combined, washed with brine, dried over sodium sulfate, filtered and concentrated in a vacuum. Silica gel chromatography (Hexanes/EtOAc=2/1-1/1) afforded 1-(7-benzyloxy-naphthalen-1-yl)-2-(tert-butyl-dimethyl-silanyloxy)-2-pyridin-4-yl-ethanol (3.85 g, 89% yield); MS m/z (M+H+) 486.20.
  • To a solution of 1-(7-benzyloxy-naphthalen-1-yl)-2-(tert-butyl-dimethyl-silanyloxy)-2-pyridin-4-yl-ethanol (3.75 g, 7.72 mmol) in tetrahydrofuran (60 mL) is added 1.0 M tetrabutylammonium fluoride in tetrahydrofuran (10 mL, 10 mmol) and the mixture is stirred at room temperature for 1 hour. The mixture is concentrated and ethyl acetate is added to the residue. The solution is washed with water, dried over sodium sulfate, filtered, concentrated and dried on a high vacuum. The resulting diol is used for the following oxidation without further purification.
  • A mixture of dimethylsulfoxide (1.64 mL, 1.81 g, 23.1 mmol) and anhydrous methylene chloride (100 mL) is cooled to −70° C. and oxalyl chloride (2.0 mL, 2.94 g, 23.1 mmol) is added dropwise. The mixture is stirred for 15 minutes at this temperature and a solution of the above diol in methylene chloride (40 mL) and dimethylsulfoxide (10 mL) is added dropwise. The mixture is stirred at −60° C. for 2 hours before triethylamine (6.45 mL, 4.68 g, 46.3 mmol) is added. The reaction mixture is gradually warmed up to room temperature and stirred over night. Water is then added to the mixture and organic phase is separated, washed with water, dried over sodium sulfate, filtered and concentrated. The crude product is purified by silica gel chromatography (Hexanes/EtOAC=3/1-2/1) to give 1-(7-benzyloxy-naphthalen-1-yl)-2-pyridin-4-yl-ethane-1,2-dione (2.26 g, 79.7% yield); 1H NMR 400 Hz (CDCl3) δ 5.31 (s, 2H), 7.40 (m, 5H), 7.55 (d, J=7.30 Hz, 2H), 7.81 (dd, J=1.66, 4.42 Hz, 2H), 7.87 (m, 2H), 8.10 (d, J=8.08 Hz, 1H), 8.89 (dd, J=1.55, 4.48 Hz, 2H), 8.93 (d, J=2.48 Hz, 1H); MS m/z (M+H+) 368.20.
  • A mixture of 1-(7-benzyloxy-naphthalen-1-yl)-2-pyridin-4-yl-ethane-1,2-dione (576 mg, 1.57 mmol), benzaldehyde (0.2 mL, 0.21 g, 1.98 mmol) and ammonium acetate (1.21 g, 15.7 mmol) in acetic acid (15 mL) is stirred at 120° C. over night. The reaction mixture is then cooled to room temperature, poured into ice cold ammonium hydroxide solution and extracted with ethyl acetate (3×100 mL). The organic extracts are combined, dried over sodium sulfate, filtered and concentrated in vacuum. Silica gel chromatography (CH2Cl2/EtOAc=2/1) of the crude product afforded 4-[5-(7-benzyloxy-naphthalen-1-yl)-2-phenyl-1H-imidazol-4-yl]-pyridine (700 mg, 98% yield); 1H NMR 400 Hz (DMSO) δ 3.30 (s, 2H), 6.99 (s, 1H), 7.21 (m, 5H), 7.24 (m, 3H), 7.43 (t, J=7.31 Hz, 1H), 7.52 (t, J=7.37 Hz, 3H), 7.66 (m, 1H), 8.00 (m, 2H), 8.13 (d, J=7.30 Hz, 2H), 8.32 (d, J=4.43 Hz, 2H); MS m/z (M+H+) 454.20.
  • To a solution of 4-[5-(7-benzyloxy-naphthalen-1-yl)-2-phenyl-1H-imidazol-4-yl]-pyridine (810 mg, 1.79 mmol) in methanol (50 mL) and tetrahydrofuran (10 mL) is added 10% Pd/C (100 mg) and the mixture is stirred for 3 hours under hydrogen atmosphere (1 atm). The catalyst is filtered and washed with ethyl acetate. The filtrate is concentrated and silica gel chromatography (CH2Cl2/EtOAc=1/1) afforded 8-(2-phenyl-5-pyridin-4-yl-3H-imidazol-4-yl)-naphthalen-2-ol (520 mg, 80% yield) as a syrup; 1H NMR 400 Hz (CD3OD) δ 6.89 (d, J=2.32 Hz, 1H), 7.17 (dd, J=2.38, 8.89 Hz, 1H), 7.48 (dd, J=7.15, 8.21 Hz, 1H), 7.55 (m, 3H), 7.66 (dd, J=1.09, 7.04 Hz, 1H), 7.91 (d, J=8.93 Hz, 1H), 7.95 (d, J=7.03 Hz, 3H), 8.04 (d, J=8.24 Hz, 1H), 8.10 (m, 2H), 8.47 (d, J=7.02 Hz, 2H); MS m/z (M+H+) 364.20.
  • A mixture of 8-(2-phenyl-5-pyridin-4-yl-3H-imidazol-4-yl)-naphthalen-2-ol (31 mg, 0.085 mmol), phenyl isocyanate (11 mg, 0.094 mmol) and triethylamine (17.8 μL, 12.9 mg, 0.128 mmol) in N,N-dimethylformamide (1.5 mL) is stirred for 2 hours at room temperature. Preparative LCMS afforded phenyl-carbamic acid 8-(2-phenyl-5-pyridin-4-yl-3H-imidazol-4-yl)-naphthalen-2-yl ester (17 mg, 41% yield); 1H NMR 400 Hz (CD3OD) δ 7.01 (m, 2H), 7.25 (m, 4H), 7.39 (m, 5H), 7.45 (dd, J=2.28, 8.86 Hz, 1H), 7.53 (m, 3H), 7.72 (dd, J=7.14, 8.23 Hz, 1H), 7.83 (d, J=6.02 Hz, 1H), 7.90 (d, J=6.99 Hz, 2H), 8.10 (m, 3H), 8.19 (d, J=8.29 Hz, 1H), 8.43 (d, J=7.00 Hz, 2H); MS m/z (M+H+) 483.20.
  • By repeating the procedures described in the above example, using appropriate starting materials, the following compounds of Formula I, as identified in Table 1, are obtained.
  • TABLE 1
    Physical Data
    1H NMR 400 MHz
    Compound (DMSO-d6)
    Number Structure and/or MS (m/z)
    2
    Figure US20090258910A1-20091015-C00004
    1H NMR 400 Hz (DMSO) δ 7.31 (d, J = 4.40 Hz, 2 H), 7.44 (dd, J = 7.27, 7.31 Hz, 1 H), 7.53 (m, 3 H), 7.61 (m, 2 H), 7.72 (m, 2 H), 8.14 (m, 4 H), 8.32 (br, 2 H). MS m/z (M + H+) 348.10
    3
    Figure US20090258910A1-20091015-C00005
    1H NMR 400 Hz (CD3OD) δ 7.50 (m, 1 H), 7.60 (dd, J = 7.17, 7.83 Hz, 1 H), 7.73 (m, 3 H), 7.94 (d, J = 6.83 Hz, 2 H), 8.06 (d, J = 7.91 Hz, 2 H), 8.18 (d, J = 8.21 Hz, 1 H), 8.45 (d, J = 6.79 Hz, 2 H), 8.72 (s, 2 H). MS m/z (M + H+) 484.10
    4
    Figure US20090258910A1-20091015-C00006
    1H NMR 400 Hz (CD3OD) δ 3.10 (s, 6 H), 6.95 (m, 2 H), 7.52 (m, 1 H), 7.60 (m, 1 H), 7.72 (m, 5 H), 7.97 (d, J = 8.94 Hz, 2 H), 8.06 (d, J = 8.14 Hz, 1 H), 8.18 (d, J = 8.20 Hz, 1 H), 8.49 (d, J = 6.28 Hz, 2 H). MS m/z (M + H+) 391.20
    5
    Figure US20090258910A1-20091015-C00007
    1H NMR 400 Hz (CD3OD) δ 3.94 (s, 3 H), 3.97 (s, 6 H), 6.46 (s, 2 H), 7.63 (m, 7 H), 8.07 (d, J = 7.99 Hz, 1 H), 8.18 (d, J = 8.19 Hz, 1 H), 8.52 (m, 2 H). MS m/z (M + H+) 438.20
    6
    Figure US20090258910A1-20091015-C00008
    1H NMR 400 Hz (CD3OD) δ 7.28 (m, 1 H), 7.38 (m, 1 H), 7.57 (m, 4 H), 7.72 (m, 2 H), 7.90 (d, J = 6.99 Hz, 2 H), 8.05 (d, J = 8.13 Hz, 1 H), 8.17 (m, 2 H), 8.43 (d, J = 7.02 Hz, 2 H). MS m/z (M + H+) 366.10
    7
    Figure US20090258910A1-20091015-C00009
    1H NMR 400 Hz (CDCl3) δ 1.43 (t, J = 6.96 Hz, 3 H), 4.27 (q, J = 6.96 Hz, 2 H), 7.07 (d, J = 8.12 Hz, 1 H), 7.22 (m, 1 H), 7.44 (m, 2 H), 7.67 (m, 4 H), 7.90 (d, J = 6.77 Hz, 2 H), 8.05 (d, J = 8.20 Hz, 1 H), 8.13 (m, 1 H), 8.43 (d, J = 6.79 Hz, 2 H), 8.53 (dd, J = 1.74, 7.82 Hz, 1 H). MS m/z (M + H+) 392.20
    8
    Figure US20090258910A1-20091015-C00010
    1H NMR 400 Hz (DMSO) δ 7.49 (m, 1 H), 7.67 (m, 6 H), 7.82 (t, J = 8.05 Hz, 1 H), 8.09 (d, J = 7.96 Hz, 1 H), 8.18 (d, J = 7.98 Hz, 1 H), 8.27 (m, 1 H), 8.52 (m, 3 H), 8.96 (t, J = 1.87 Hz, 1 H). MS m/z (M + H+) 393.10
    9
    Figure US20090258910A1-20091015-C00011
    MS m/z (M + H+) 272.10
    10
    Figure US20090258910A1-20091015-C00012
    1H NMR 400 Hz (CD3OD) δ 7.15 (m, 2 H), 7.49 (m, 6 H), 7.68 (m, 3 H), 7.86 (d, J = 5.73 Hz, 2 H), 7.91 (s, 1 H), 8.08 (d, J = 6.75 Hz, 2 H), 8.22 (d, J = 5.28 Hz, 2 H). MS m/z (M + H+) 483.20.
    11
    Figure US20090258910A1-20091015-C00013
    1H NMR 400 Hz (DMSO) δ 7.15 (m, 2 H), 7.45 (m, 1 H), 7.52 (m, 5 H), 7.81 (m, 2 H), 8.01 (s, 1 H), 8.11 (d, J = 7.32 Hz, 2 H), 8.45 (br, 2 H). MS m/z (M + H+) 364.20
    12
    Figure US20090258910A1-20091015-C00014
    1H NMR 400 Hz (CD3OD) δ 3.89 (s, 3 H), 7.51 (m, 4 H), 7.68 (m, 3 H), 7.84 (d, J = 2.0 Hz, 1 H), 8.01 (m, 3 H), 8.11 (m, 5 H), 8.23 (s, 1 H), 8.56 (d, J = 6.89 Hz, 2 H). MS m/z (M + H+) 541.20
    13
    Figure US20090258910A1-20091015-C00015
    1H NMR 400 Hz (CD3OD) δ 5.92 (s, 2 H), 6.77 (d, J = 8.36 Hz, 1 H), 6.90 (dd, J = 2.09, 8.36 Hz, 1 H), 7.17 (s, 1 H), 7.46 (dd, J = 2.29, 8.86 Hz, 1 H), 7.53 (m, 3 H), 7.66 (dd, J = 1.68, 8.47 Hz, 1 H), 7.78 (d, J = 2.11 Hz, 1 H), 8.09 (m, 6 H), 8.19 (s, 1 H), 8.54 (d, J = 6.97 Hz, 2 H). MS m/z (M + H+) 527.20
    14
    Figure US20090258910A1-20091015-C00016
    1H NMR 400 Hz (CD3OD) δ 3.92 (s, 6 H), 7.52 (m, 4 H), 7.70 (dd, J = 1.64, 8.32 Hz, 1 H), 7.85 (d, J = 2.03 Hz, 1 H), 8.04 (d, J = 9.06 Hz, 1 H), 8.10 (m, 5 H), 8.23 (br, 1 H), 8.34 (m, 1 H), 8.45 (d, J = 1.43 Hz, 2 H), 8.55 (d, J = 7.85 Hz, 2 H). MS m/z (M + H+) 599.20.
    15
    Figure US20090258910A1-20091015-C00017
    1H NMR 400 Hz (CD3OD) δ 3.31 (s, 6 H), 7.53 (m, 5 H), 7.64 (d, J = 9.06 Hz, 2 H), 7.70 (dd, J = 1.65, 8.47 Hz, 1 H), 7.79 (d, J = 9.09 Hz, 2 H), 7.84 (d, J = 1.97 Hz, 1 H), 8.11 (m, 7 H), 8.24 (s, 1 H), 8.59 (s, br, 2 H). MS m/z (M + H+) 526.30
    16
    Figure US20090258910A1-20091015-C00018
    1H NMR 400 Hz (CD3OD) δ 5.89 (s, 2 H), 6.72 (m, 2 H), 6.70 (s, 1 H), 7.43 (m, 2 H), 7.55 (m, 3 H), 7.71 (t, J = 8.19 Hz, 1 H), 7.82 (d, J = 6.17 Hz, 1 H), 7.89 (d, J = 6.97 Hz, 2 H), 8.09 (m, 3 H), 8.18 (d, J = 8.23 Hz, 1 H), 8.42 (d, J = 6.96 Hz, 2 H). MS m/z (M + H+) 527.20
    17
    Figure US20090258910A1-20091015-C00019
    1H NMR 400 Hz (CD3OD) δ 3.92 (s, 6 H), 7.43 (dd, J = 2.23, 8.84 Hz, 1 H), 7.50 (m, 4 H), 7.74 (dd, J = 7.20, 8.20 Hz, 1 H), 7.90 (m, 3 H), 8.10 (m, 3 H), 8.19 (d, J = 8.29 Hz, 1 H), 8.25(m, 3 H), 8.49 (d, J = 6.94 Hz, 2 H). MS m/z (M + H+) 599.10
    18
    Figure US20090258910A1-20091015-C00020
    1H NMR 400 Hz (CD3OD) δ 3.24 (s, 6 H), 7.47 (m, 2 H), 7.54 (m, 5 H), 7.61 (m, 2 H), 7.73 (dd, J = 7.38, 8.20 Hz, 1 H), 7.83 (m, 1 H), 7.91 (d, J = 6.98 Hz, 2 H), 8.11 (m, 3 H), 8.20 (d, J = 8.28 Hz, 1 H), 8.46 (d, J = 6.96 Hz, 2 H). MS m/z (M + H+) 526.30
  • Assays
  • Compounds of the present invention are assayed to measure their capacity to selectively inhibit cell proliferation of 32D cells expressing BCR-Abl (32D-p210) compared with parental 32D cells. Compounds selectively inhibiting the proliferation of these BCR-Abl transformed cells are tested for anti-proliferative activity on Ba/F3 cells expressing either wild type or the mutant forms of Bcr-abl. In addition, compounds are assayed to measure their capacity to inhibit b-Raf.
  • Inhibition of Cellular BCR-Abl Dependent Proliferation (High Throughput Method)
  • The murine cell line used is the 32D hemopoietic progenitor cell line transformed with BCR-Abl cDNA (32D-p210). These cells are maintained in RPMI/10% fetal calf serum (RPMI/FCS) supplemented with penicillin 50 μg/mL, streptomycin 50 μg/mL and L-glutamine 200 mM. Untransformed 32D cells are similarly maintained with the addition of 15% of WEHI conditioned medium as a source of IL3.
  • 50 μl of a 32D or 32D-p210 cells suspension are plated in Greiner 384 well microplates (black) at a density of 5000 cells per well. 50 nl of test compound (1 mM in DMSO stock solution) is added to each well (STI571 is included as a positive control). The cells are incubated for 72 hours at 37° C., 5% CO2. 10 μl of a 60% Alamar Blue solution (Tek diagnostics) is added to each well and the cells are incubated for an additional 24 hours. The fluorescence intensity (Excitation at 530 nm, Emission at 580 nm) is quantified using the Acquest™ system (Molecular Devices).
  • Inhibition of Cellular BCR-Abl Dependent Proliferation
  • 32D-p210 cells are plated into 96 well TC plates at a density of 15,000 cells per well. 50 μL of two fold serial dilutions of the test compound (Cmax is 40 μM) are added to each well (STI571 is included as a positive control). After incubating the cells for 48 hours at 37° C., 5% CO2, 15 μL of MTT (Promega) is added to each well and the cells are incubated for an additional 5 hours. The optical density at 570 nm is quantified spectrophotometrically and IC50 values, the concentration of compound required for 50% inhibition, determined from a dose response curve.
  • Effect on Cell Cycle Distribution
  • 32D and 32D-p210 cells are plated into 6 well TC plates at 2.5×106 cells per well in 5 ml of medium and test compound at 1 or 10 μM is added (STI571 is included as a control). The cells are then incubated for 24 or 48 hours at 37° C., 5% CO2. 2 ml of cell suspension is washed with PBS, fixed in 70% EtOH for 1 hour and treated with PBS/EDTA/RNase A for 30 minutes. Propidium iodide (Cf=10 μg/ml) is added and the fluorescence intensity is quantified by flow cytometry on the FACScalibur™ system (BD Biosciences). Test compounds of the present invention demonstrate an apoptotic effect on the 32D-p210 cells but do not induce apoptosis in the 32D parental cells.
  • Effect on Cellular BCR-Abl Autophosphorylation
  • BCR-Abl autophosphorylation is quantified with capture Elisa using a c-abl specific capture antibody and an antiphosphotyrosine antibody. 32D-p210 cells are plated in 96 well TC plates at 2×105 cells per well in 50 μL of medium. 50 μL of two fold serial dilutions of test compounds (Cmax is 10 μM) are added to each well (STI571 is included as a positive control). The cells are incubated for 90 minutes at 37° C., 5% CO2. The cells are then treated for 1 hour on ice with 150 μL of lysis buffer (50 mM Tris-HCl, pH 7.4, 150 mM NaCl, 5 mM EDTA, 1 mM EGTA and 1% NP-40) containing protease and phosphatase inhibitors. 50 μL of cell lysate is added to 96 well optiplates previously coated with anti-abl specific antibody and blocked. The plates are incubated for 4 hours at 4° C. After washing with TBS-Tween 20 buffer, 50 μL of alkaline-phosphatase conjugated anti-phosphotyrosine antibody is added and the plate is further incubated overnight at 4° C. After washing with TBS-Tween 20 buffer, 90 μL of a luminescent substrate are added and the luminescence is quantified using the Acquest™ system (Molecular Devices). Test compounds of the invention that inhibit the proliferation of the BCR-Abl expressing cells, inhibit the cellular BCR-Abl autophosphorylation in a dose-dependent manner.
  • Effect on Proliferation of Cells Expressing Mutant Forms of Bcr-Abl
  • Compounds of the invention are tested for their antiproliferative effect on Ba/F3 cells expressing either wild type or the mutant forms of BCR-Abl (G250E, E255V, T315I, F317L, M351T) that confers resistance or diminished sensitivity to STI571. The antiproliferative effect of these compounds on the mutant-BCR-Abl expressing cells and on the non transformed cells were tested at 10, 3.3, 1.1 and 0.37 μM as described above (in media lacking IL3). The IC50 values of the compounds lacking toxicity on the untransformed cells were determined from the dose response curves obtained as describe above.
  • b-Raf
  • Compounds of the invention are tested for their ability to inhibit the activity of b-Raf. The assay is carried out in 384-well MaxiSorp plates (NUNC) with black walls and clear bottom. The substrate, IκBα is diluted in DPBS (1:750) and 15 μl is added to each well. The plates are incubated at 4° C. overnight and washed 3 times with TBST (25 mM Tris, pH 8.0, 150 mM NaCl and 0.05% Tween-20) using the EMBLA plate washer. Plates are blocked by Superblock (15 μl/well) for 3 hours at room temperature, washed 3 times with TBST and pat-dried. Assay buffer containing 20 μM ATP (10 μl) is added to each well followed by 100 nl or 500 nl of compound. B-Raf is diluted in the assay buffer (1 μl into 25 μl) and 10 μl of diluted b-Raf is added to each well (0.4 μg/well). The plates are incubated at room temperature for 2.5 hours. The kinase reaction is stopped by washing the plates 6 times with TBST. Phosph-IκBα (Ser32/36) antibody is diluted in Superblock (1:10,000) and 15 μl is added to each well. The plates are incubated at 4° C. overnight and washed 6 times with TBST. AP-conjugated goat-anti-mouse IgG is diluted in Superblock (1:1,500) and 15 μl is added to each well. Plates are incubated at room temperature for 1 hour and washed 6 times with TBST. 15 μl of Attophos AP substrate is added to each well and plates are incubated at room temperature for 15 minutes. Plates are read on Acquest or Analyst GT using a Fluorescence Intensity Nanxin BBT anion (505 dichroic mirror).
  • Upstate KinaseProfiler™-Radio-Enzymatic Filter Binding Assay
  • Compounds of the invention are assessed for their ability to inhibit individual members of a panel of kinases (a partial, non-limiting list of kinases includes: Abl, BCR-Abl, EGF-R, c-erbB2 kinase (HER-2), PDGF-R, lck, SAPK2α, p38, TGFβ, KDR, c-Kit, b-RAF, c-RAF, FLT1 and FLT4). The compounds are tested in duplicates at a final concentration of 10 μM following this generic protocol. Note that the kinase buffer composition and the substrates vary for the different kinases included in the “Upstate KinaseProfiler™” panel. The compounds are tested in duplicates at a final concentration of 10 μM following this generic protocol. Note that the kinase buffer composition and the substrates vary for the different kinases included in the “Upstate KinaseProfiler™” panel. Kinase buffer (2.5 μL, 10×-containing MnCl2 when required), active kinase (0.001-0.01 Units; 2.5 μL), specific or Poly (Glu4-Tyr) peptide (5-500 μM or 0.01 mg/ml) in kinase buffer and kinase buffer (50 μM; 5 μL) are mixed in an eppendorf on ice. A Mg/ATP mix (10 μL; 67.5 (or 33.75) mM MgCl2, 450 (or 225) μM ATP and 1 μCi/μl [γ-32P]-ATP (3000 Ci/mmol)) is added and the reaction is incubated at about 30° C. for about 10 minutes. The reaction mixture is spotted (20 μL) onto a 2 cm×2 cm P81 (phosphocellulose, for positively charged peptide substrates) or Whatman No. 1 (for Poly (Glu4-Tyr) peptide substrate) paper square. The assay squares are washed 4 times, for 5 minutes each, with 0.75% phosphoric acid and washed once with acetone for 5 minutes. The assay squares are transferred to a scintillation vial, 5 ml scintillation cocktail are added and 32P incorporation (cpm) to the peptide substrate is quantified with a Beckman scintillation counter. Percentage inhibition is calculated for each reaction.
  • Compounds of Formula I, in free form or in pharmaceutically acceptable salt form, exhibit valuable pharmacological properties, for example, as indicated by the in vitro tests described in this application. For example, compounds of Formula I preferably show an IC50 in the range of 1×10−10 to 1×10−5 M, preferably less than 500 nM for wild type BCR-Abl and b-Raf. For example, 6-(2-phenyl-5-pyridin-4-yl-1H-imidazol-4-yl)-naphthalen-2-ol (compound 11) and (4-dimethylamino-phenyl)-carbamic acid 6-(2-phenyl-5-pyridin-4-yl-1H-imidazol-4-yl)-naphthalen-2-yl ester (compound 15) have an IC50 of 300 nM and 879 nM against b-Raf, respectively.
  • Compounds of Formula I, at a concentration of 10 μM, preferably show a percentage inhibition of greater than 50%, preferably greater than about 70%, against Abl, BCR-Abl, PDGF-R, lck, SAPK20α, p38, TGFβ, KDR, c-Kit, b-RAF, c-RAF, FLT1 and/or FLT4 kinases. For example, 6-(2-Phenyl-5-pyridin-4-yl-1H-imidazol-4-yl)-naphthalen-2-ol (compound 11) at a concentration of 10 μM, inhibits the following kinases by the percentage shown in brackets (for example, 100% means complete inhibition, 0% means no inhibition): Abl (98%); c-RAF (98%), Lck (61%); and SAPK2α (74%).
  • It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims. All publications, patents, and patent applications cited herein are hereby incorporated by reference for all purposes.

Claims (6)

1. A compound of Formula I:
Figure US20090258910A1-20091015-C00021
in which:
R1 is selected from hydrogen, —XR3, —XC(O)R3, —XSR3, —XS(O)R3, —XS(O)2R3, —XOR4, and —XOC(O)NR3R4; wherein X is a bond or C1-4alkylene;
R2 is selected from —XR3, —XC(O)R3, —XSR3, and —XOC(O)NR3R4; wherein X is a bond or C1-4alkylene;
R4 is selected from hydrogen and C1-6alkyl;
R3 is selected from C5-10heteroaryl, and C3-8heterocycloalkyl; wherein any heteroaryl, or heterocycloalkyl of R3 is optionally substituted by 1 to 3 radicals selected from hydroxy, halo, nitro, C1-6alkyl, C1-6alkoxy, halo-substituted-C1-6alkyl, halo-substituted-C1-6alkoxy, —XC(O)OR4 and —NR4R5; wherein X is a bond or C1-4alkylene and R4 and R5 are independently selected from hydrogen and C1-6alkyl;
and pharmaceutically acceptable salts thereof.
2. The compound of claim 1, wherein:
R1 is selected from hydrogen, —XR3, and —XOC(O)NR3R4; wherein X is a bond or C1-4alkylene;
R2 is selected from —XR3 and —XOC(O)NR3R4; wherein X is a bond or C1-4alkylene;
R4 is selected from hydrogen and C1-6alkyl;
R3 is C5-10heteroaryl or C3-8heterocycloalkyl optionally substituted by 1 to 3 radicals selected from halo, nitro, C1-6alkyl, C1-6alkoxy, halo-substituted-C1-6alkyl, —XC(O)OR4 and —NR4R5; wherein X is a bond or C1-4alkylene and R4 and R5 are independently selected from hydrogen and C1-6alkyl.
3. The compound of claim 2, wherein R1 is selected from hydrogen and phenyl; wherein said phenyl is optionally substituted by 1 to 3 radicals selected from trifluoromethyl, dimethylamino, methoxy, halo, ethoxy and nitro.
4. The compound of claim 2, wherein in which R2 is —OC(O)NHR3 and R3 is benzo[1,3]dioxol-5-yl optionally substituted by 1 to 3 radicals selected from —C(O)OCH3 and dimethylamino.
5. The compound of claim 1 selected from benzo[1,3]dioxol-5-yl-carbamic acid 6-(2-phenyl-5-pyridin-4-yl-1H-imidazol-4-yl)-naphthalen-2-yl ester and benzo[1,3]dioxol-5-yl-carbamic acid 8-(2-phenyl-5-pyridin-4-yl-1H-imidazol-4-yl)-naphthalen-2-yl ester.
6. A pharmaceutical composition comprising a therapeutically effective amount of a compound of claim 1 in combination with a pharmaceutically acceptable excipient.
US12/492,081 2003-10-02 2009-06-25 Compounds and compositions as protein kinase inhibitors Abandoned US20090258910A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/492,081 US20090258910A1 (en) 2003-10-02 2009-06-25 Compounds and compositions as protein kinase inhibitors

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US50845003P 2003-10-02 2003-10-02
US10/956,880 US7569593B2 (en) 2003-10-02 2004-10-01 Compounds and compositions as protein kinase inhibitors
US12/492,081 US20090258910A1 (en) 2003-10-02 2009-06-25 Compounds and compositions as protein kinase inhibitors

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/956,880 Division US7569593B2 (en) 2003-10-02 2004-10-01 Compounds and compositions as protein kinase inhibitors

Publications (1)

Publication Number Publication Date
US20090258910A1 true US20090258910A1 (en) 2009-10-15

Family

ID=34393249

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/956,880 Expired - Fee Related US7569593B2 (en) 2003-10-02 2004-10-01 Compounds and compositions as protein kinase inhibitors
US12/492,081 Abandoned US20090258910A1 (en) 2003-10-02 2009-06-25 Compounds and compositions as protein kinase inhibitors

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/956,880 Expired - Fee Related US7569593B2 (en) 2003-10-02 2004-10-01 Compounds and compositions as protein kinase inhibitors

Country Status (9)

Country Link
US (2) US7569593B2 (en)
EP (1) EP1670780A4 (en)
JP (1) JP2007507540A (en)
CN (1) CN100455579C (en)
AU (1) AU2004275888B2 (en)
BR (1) BRPI0414869A (en)
CA (1) CA2540518A1 (en)
MX (1) MXPA06003644A (en)
WO (1) WO2005030151A2 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9012462B2 (en) 2008-05-21 2015-04-21 Ariad Pharmaceuticals, Inc. Phosphorous derivatives as kinase inhibitors
US9273077B2 (en) 2008-05-21 2016-03-01 Ariad Pharmaceuticals, Inc. Phosphorus derivatives as kinase inhibitors
US9611283B1 (en) 2013-04-10 2017-04-04 Ariad Pharmaceuticals, Inc. Methods for inhibiting cell proliferation in ALK-driven cancers
US9834571B2 (en) 2012-05-05 2017-12-05 Ariad Pharmaceuticals, Inc. Compounds for inhibiting cell proliferation in EGFR-driven cancers
US9834518B2 (en) 2011-05-04 2017-12-05 Ariad Pharmaceuticals, Inc. Compounds for inhibiting cell proliferation in EGFR-driven cancers

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005102325A1 (en) * 2004-04-20 2005-11-03 Ab Science Use of c-kit inhibitors for treating inflammatory muscle disorders including myositis and muscular dystrophy
EP1992344A1 (en) 2007-05-18 2008-11-19 Institut Curie P38 alpha as a therapeutic target in pathologies linked to FGFR3 mutation
US8178526B2 (en) * 2007-06-15 2012-05-15 Irm Llc Compounds and compositions as ITPKb inhibitors
JP5370472B2 (en) * 2009-02-19 2013-12-18 富士通株式会社 Novel compounds and binding inhibitors, antiallergic agents, antiasthmatic agents, and antiinflammatory agents
WO2010095227A1 (en) * 2009-02-19 2010-08-26 富士通株式会社 Novel compound, and binding inhibitor, anti-allergic agent, anti-asthma agent and anti-inflammatory agent
CN103159735B (en) * 2011-12-10 2015-12-09 通化济达医药有限公司 The imidazoles kinase inhibitor replaced
BR112019000868A2 (en) 2016-07-22 2019-05-07 Syngenta Participations Ag urea and thiourea substituted bicyclic derivatives as pesticides
CN108103186B (en) * 2018-02-28 2021-07-02 固安博健生物技术有限公司 Molecular markers for diagnosis of rheumatoid arthritis and osteoarthritis

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6207687B1 (en) * 1998-07-31 2001-03-27 Merck & Co., Inc. Substituted imidazoles having cytokine inhibitory activity
US6235760B1 (en) * 1996-03-25 2001-05-22 Smithkline Beecham Corporation Treatment for CNS injuries
US20030165873A1 (en) * 2001-03-02 2003-09-04 Come Jon H. Three hybrid assay system
US7199137B2 (en) * 2000-09-21 2007-04-03 Smithkline Beecham Plc Imidazole derivatives as Raf kinase inhibitors

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5916891A (en) * 1992-01-13 1999-06-29 Smithkline Beecham Corporation Pyrimidinyl imidazoles
WO1995003297A1 (en) 1993-07-21 1995-02-02 Smithkline Beecham Corporation Imidazoles for treating cytokine mediated disease
CZ292896B6 (en) * 1994-02-04 2003-12-17 Smithkline Beecham P. L. C. Process for enantioselectively epoxidising and compounds suitable therefor as catalysts
GB2306108A (en) * 1995-10-13 1997-04-30 Merck & Co Inc Treatment of Raf-mediated cancers with imidazole derivatives
WO1997036587A1 (en) 1996-04-03 1997-10-09 Merck & Co., Inc. A method of treating cancer
GB9713726D0 (en) * 1997-06-30 1997-09-03 Ciba Geigy Ag Organic compounds
EP1233769B1 (en) * 1999-11-22 2007-12-26 SmithKline Beecham plc Imidazole derivatives
US20040053943A1 (en) * 2000-11-20 2004-03-18 Adams Jerry L. Novel compounds

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6235760B1 (en) * 1996-03-25 2001-05-22 Smithkline Beecham Corporation Treatment for CNS injuries
US6207687B1 (en) * 1998-07-31 2001-03-27 Merck & Co., Inc. Substituted imidazoles having cytokine inhibitory activity
US7199137B2 (en) * 2000-09-21 2007-04-03 Smithkline Beecham Plc Imidazole derivatives as Raf kinase inhibitors
US20030165873A1 (en) * 2001-03-02 2003-09-04 Come Jon H. Three hybrid assay system

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9012462B2 (en) 2008-05-21 2015-04-21 Ariad Pharmaceuticals, Inc. Phosphorous derivatives as kinase inhibitors
US9273077B2 (en) 2008-05-21 2016-03-01 Ariad Pharmaceuticals, Inc. Phosphorus derivatives as kinase inhibitors
US9834518B2 (en) 2011-05-04 2017-12-05 Ariad Pharmaceuticals, Inc. Compounds for inhibiting cell proliferation in EGFR-driven cancers
US9834571B2 (en) 2012-05-05 2017-12-05 Ariad Pharmaceuticals, Inc. Compounds for inhibiting cell proliferation in EGFR-driven cancers
US9611283B1 (en) 2013-04-10 2017-04-04 Ariad Pharmaceuticals, Inc. Methods for inhibiting cell proliferation in ALK-driven cancers

Also Published As

Publication number Publication date
MXPA06003644A (en) 2006-06-20
WO2005030151A2 (en) 2005-04-07
EP1670780A2 (en) 2006-06-21
AU2004275888A1 (en) 2005-04-07
CN1860111A (en) 2006-11-08
WO2005030151A3 (en) 2005-09-09
JP2007507540A (en) 2007-03-29
US7569593B2 (en) 2009-08-04
US20050209285A1 (en) 2005-09-22
CA2540518A1 (en) 2005-04-07
CN100455579C (en) 2009-01-28
EP1670780A4 (en) 2008-12-17
AU2004275888B2 (en) 2008-09-18
BRPI0414869A (en) 2006-11-28

Similar Documents

Publication Publication Date Title
US20090258910A1 (en) Compounds and compositions as protein kinase inhibitors
US7189729B2 (en) Methods and compositions as protein kinase inhibitors
US7449582B2 (en) Compounds and compositions as protein kinase inhibitors
US8106068B2 (en) Compositions and methods for modulating c-kit and PDGFR receptors
US7968557B2 (en) Substituted pyrrolo[2,3-2]pyrimidines as protein kinase inhibitors
AU2004227943B2 (en) Novel compounds and compositions as protein kinase inhibitors
ES2477878T3 (en) Compounds and compositions of 5- (4- (aloalkoxy) phenyl) pyrimidin-2-amine as kinase inhibitors
KR101280809B1 (en) Anthranilamide pyridinureas as vegf receptor kinase inhibitors
JP2010512405A (en) Compounds and compositions as kinase inhibitors
MXPA06009158A (en) Compounds and compositions as protein kinase inhibitors

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION