WO2006124731A2 - Composes et compositions utilise(e)s en tant qu'inhibiteurs de proteines-kinases - Google Patents

Composes et compositions utilise(e)s en tant qu'inhibiteurs de proteines-kinases Download PDF

Info

Publication number
WO2006124731A2
WO2006124731A2 PCT/US2006/018644 US2006018644W WO2006124731A2 WO 2006124731 A2 WO2006124731 A2 WO 2006124731A2 US 2006018644 W US2006018644 W US 2006018644W WO 2006124731 A2 WO2006124731 A2 WO 2006124731A2
Authority
WO
WIPO (PCT)
Prior art keywords
ethyl
tetraaza
tetrahydro
oxo
cyclopenta
Prior art date
Application number
PCT/US2006/018644
Other languages
English (en)
Other versions
WO2006124731A3 (fr
Inventor
Pingda Ren
Nathanael S. Gray
Xia Wang
Guobao Zhang
Taebo Sim
Songchun Jiang
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
Publication of WO2006124731A2 publication Critical patent/WO2006124731A2/fr
Publication of WO2006124731A3 publication Critical patent/WO2006124731A3/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains three hetero rings
    • C07D471/14Ortho-condensed systems
    • 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

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 AbI, Bcr-Abl, BMX, BTK, CHK2, b-RAF, c-RAF, CSK, c-SRC, Fes, FGFR3, Flt3, IKK ⁇ , IKK ⁇ , JNK2 ⁇ 2, Lck, Met, MKK4, MKK6, MST2, NEK2, p70S6K, PDGFR ⁇ , PKA, PKB ⁇ , PKD2, Rskl, SAPK2 ⁇ , SAPK2 ⁇ , SAPK3, SGK, Tie2 and TrkB 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), the nerve growth factor receptor, trkB, Met, and the fibroblast growth factor receptor, FGFR3; non-receptor tyrosine kinases such AbI and the fusion kinase BCR-AbI, Lck, Csk, Fes, Bmx and c-src; and serine/threonine kinases such as b-RAF, c-RAF, sgk, MAP kinases (e.g., MKK4, MKK6, etc.) and SAPK2 ⁇ , SAPK2 ⁇ and SAPK3.
  • PDGF-R platelet-derived growth factor receptor kinase
  • 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:
  • n is selected from 0, 1, 2, 3 and 4;
  • Ri is selected from hydrogen and Ci. 6 alkyl
  • R 2 is selected from hydrogen and Ci -6 alkyl
  • R 3 is selected from halo, C h alky! and Ci -6 alkoxy;
  • R 4 is selected from NR 5 C(O)NR 5 R 6 , NR 5 C(O)R 6 , C(O)NR 5 R 6 ,
  • any alkyl or alkylene of R 6 is optionally substituted with -XOR 5 ; wherein any aryl, heteroaryl, cycloalkyl and heterocycloalkyl of R 6 is optionally substituted by 1 to 3 radicals independently selected from halo, cyano, nitro, Ci -6 alkyl, C 1-6 alkoxy, halo- substituted-Ci -6 alkyl, halo-substituted-Ci -6 alkoxy, C 5- i 2 heteroaryl-Co- 6 alkyl and C 3- i 2 heterocycloalkyl-Co- 6 alkyl; wherein any heteroaryl or heterocycloalkyl substituents of R 6 can optionally be substituted by a radical independently selected from Ci -6 alkyl and C 3- i 2 heterocycloalkyl; and the N-oxide derivatives, prodrug derivatives, protected derivatives, individual isomers and mixture of isomers thereof; and the pharmaceutically acceptable salts and
  • 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 AbI, Bcr-Abl, BMX, BTK, CHK2, b-RAF, c-RAF, CSK, c-SRC, Fes, FGFR3, Flt3, IKK ⁇ , IKK ⁇ , JNK2 ⁇ 2, Lck, Met, MKK4, MKK6, MST2, NEK2, p70S6K, PDGFR ⁇ , PKA, PKB ⁇ , PKD2, Rskl, SAPK2 ⁇ , SAPK2 ⁇ , SAPK3, SGK, Tie2 and/or TrkB 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.
  • kinase activity particularly AbI, Bcr-Abl, BM
  • the present invention provides the use of a compound of
  • kinase activity particularly AbI, Bcr-Abl, BMX, BTK, CHK2, b-RAF, c-RAF, CSK, c-SRC, Fes, FGFR3, Flt3, IKK ⁇ , IKK ⁇ , JNK2 ⁇ 2, Lck, Met, M
  • 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. Ci- 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 above where one or more of the carbon ring members indicated can be replaced by a heteroatom.
  • C 5-8 heteroaryl includes pyridyl, indolyl, indazolyl, quinoxalinyl, quinolinyl, benzofuranyl, benzopyranyl, benzothiopyranyl, benzo[l,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- iocycloalkyl includes cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, etc.
  • C 3- sheterocycloalkyl as used in this application to describe compounds of the invention includes morpholino, pyrrolidinyl, pyrrolidinyl-2-one, piperazinyl, piperidinyl, piperidinylone, l,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.
  • Kease Panel is a list of kinases comprising Abl(human), Abl(T3151),
  • mutant forms of BCR-AbI means single or multiple amino acid changes from the wild-type sequence. Mutations in BCR-ABL act by disrupting critical contact points between protein and inhibitor (for example, Gleevec, and the like), more often, by inducing a transition from the inactive to the active state, i.e. to a conformation to which BCR-ABL and Gleevec is unable to bind. From analyses of clinical samples, the repertoire of mutations found in association with the resistant phenotype has been increasing slowly but inexorably over time.
  • One group of mutations (G250E, Q252R, Y253F/H, E255K/V) includes amino acids that form the phosphate-binding loop for ATP (also known as the P-loop).
  • a second group (V289A, F31 IL, T315I, F317L) can be found in the Gleevec binding site and interacts directly with the inhibitor via hydrogen bonds or Van der Waals' interactions.
  • the third group of mutations (M351T, E355G) clusters in close proximity to the catalytic domain.
  • the fourth group of mutations (H396R/P) is located in the activation loop, whose conformation is the molecular switch controlling kinase activation/inactivation.
  • BCR-ABL point mutations associated with Gleevec resistance detected in CML and ALL patients include: M224V, L248V, G250E, G250R, Q252R, Q252H, Y253H, Y253F, E255K, E255V, D276G, T277A, V289A, F311L, T315I, T315N, F317L, M343T, M315T, E355G, F359V, F359A, V379I, F382L, L387M, L387F, H396P, H396R, A397P, S417Y, E459K, and F486S (Amino acid positions, indicated by the single letter code, are those for the GenBank sequence, accession number
  • Treatment refers to a method of alleviating or abating a disease and/or its attendant symptoms. uesc ⁇ ptton ot the Preferred Embodiments
  • the fusion protein BCR-AbI is a result of a reciprocal translocation that fuses the AbI proto-oncogene with the Bcr gene. BCR-AbI is then capable of transforming B-cells through the increase of mitogenic activity. This increase results in a reduction of sensitivity to apoptosis, as well as altering the adhesion and homing of CML progenitor cells.
  • the present invention provides compounds, compositions and methods for the treatment of kinase related disease, particularly AbI, Bcr-Abl, BMX, BTK, CHK2, b-RAF, c-RAF, CSK, c-SRC, Fes, FGFR3, Flt3, IKK ⁇ , IKK ⁇ , JNK2 ⁇ 2, Lck, Met, MKK4, MKK6, MST2, NEK2, p70S6K, PDGFR ⁇ , PKA, PKB ⁇ , PKD2, RsId, SAPK2 ⁇ , SAPK2 ⁇ , SAPK3, SGK, Tie2 and TrkB kinase related diseases.
  • leukemia and other proliferation disorders related to BCR-AbI can be treated through the inhibition of wild type and mutant forms of Bcr-Abl.
  • Ri is hydrogen
  • R 2 is selected from methyl and ethyl
  • R 3 is selected from methyl and methoxy.
  • R 4 is selected from C(O)NR 5 R 6 , C(O)OR 6 ,
  • R 5 is independently selected from hydrogen and Ci -6 alkyl; and R 6 is selected from hydrogen, Ci -6 alkyl, -XOR 5 , -XNR 5 R 5 , C 6 -i 2 aryl-Co- 4 alkyl, Cs-sheteroaryl-Co ⁇ alkyl, Cs- ⁇ cycloalkyl-Co ⁇ alkyl and C 3- 8 heterocycloalkyl-Co -4 alkyl; wherein X is selected from a bond and Ci 4 alkylene; wherein any alkyl or alkylene OfR 6 is optionally substituted with -XOR 5 ; wherein any aryl, heteroaryl, cycloalkyl and heterocycloalkyl of R 6 is optionally substituted by 1 to 3 radicals independently selected from halo, cyano, nitro, Ci-ealkyl,
  • R 4 is selected from C(O)NHR 6 , NHC(O)R 6 , OR 6 and NHR 6 ; wherein R 6 is selected from methyl, ethyl, cyclopropyl, phenyl, pyrrolidinyl- ethyl and morpholino-ethyl; wherein said phenyl is optionally substituted with 1 to 3 radicals independently selected from halo, trifluoromethyl, imidazolyl, morpholino, piperazinyl, piperidinyl and piperazinyl-ethyl; wherein said imidazolyl, morpholino, piperazinyl, piperidinyl and piperazinyl-ethyl can be optionally substituted with 1 to 3 radicals independently selected from methyl, ethyl and pyrrolidinyl.
  • Compounds of the invention modulate the activity of kinases and, as such, are useful for treating diseases or disorders in which kinases, contribute to the pathology and/or symptomology of the disease.
  • kinases that are inhibited by the compounds and compositions described herein and against which the methods described herein are useful include, but are not limited to, AbI, Bcr-Abl (wild type and mutant forms), BMX, BTK, CHK2, b-RAF, c-RAF, CSK, c-SRC, Fes, FGFR3, Flt3, IKK ⁇ , IKK ⁇ , JNK2 ⁇ 2, Lck, Met, MKK4, MKK6, MST2, NEK2, p70S6K, PDGFR ⁇ , PKA, PKB ⁇ , PKD2, Rskl, SAPK2 ⁇ , SAPK2 ⁇ , SAPK3, SGK, Tie2 and TrIcB.
  • Abelson tyrosine kinase (i.e. AbI, 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 AbI 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-AbI with deregulated tyrosine kinase activity or the v- AbI.
  • BCR-AbI 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-AbI tyrosine kinase and is used for the treatment of chronic myeloid leukemia (CML).
  • CML chronic myeloid leukemia
  • STI-571 is an inhibitor of the oncogenic BCR-AbI 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 mutations in the BCR-AbI kinase. Over 22 mutations have been reported to date with the most common being G250E, E255V, T315L 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-AbI kinase and mutations of BCR-AbI 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)
  • 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 (01 JuI 2002).
  • 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.
  • PDGF Platinum-derived Growth Factor
  • PDGF 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 rumor diseases, such as gliomas, sarcomas, prostate tumors, and tumors of the colon, breast, and ovary.
  • rumor 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 rumor- 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 bronchoalveolar lavage fluids.
  • 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.
  • the trk family of neurotrophin receptors promotes the survival, growth and differentiation of the neuronal and non-neuronal tissues.
  • the TrkB protein is expressed in neuroendocrine-type cells in the small intestine and colon, in the alpha cells of the pancreas, in the monocytes and macrophages of the lymph nodes and of the spleen, and in the granular layers of the epidermis (Shibayama and Koizumi, 1996). Expression of the TrkB protein has been associated with an unfavorable progression of Wilms tumors and of neuroblastomas.
  • TkrB is, moreover, expressed in cancerous prostate cells but not in normal cells.
  • the signaling pathway downstream of the trk receptors involves the cascade of MAPK activation through the She, activated Ras, ERK-I and ERK-2 genes, and the PLC-gammal transduction pathway (Sugimoto et al., 2001).
  • the kinase, c-Src transmits oncogenic signals of many receptors. For example, over-expression of EGFR or HER2/neu in tumors leads to the constitutive activation of c-src, which is characteristic for the malignant cell but absent from the normal cell.
  • mice deficient in the expression of c-src exhibit an osteopetrotic phenotype, indicating a key participation of c-src in osteoclast function and a possible involvement in related disorders.
  • Fibroblast growth factor receptor 3 was shown to exert a negative regulatory effect on bone growth and an inhibition of chondrocyte proliferation. Thanatophoric dysplasia is caused by different mutations in fibroblast growth factor receptor 3, and one mutation, TDII FGFR3, has a constitutive tyrosine kinase activity which activates the transcription factor Statl, leading to expression of a cell-cycle inhibitor, growth arrest and abnormal bone development (Su et al., Nature, 1997, 386, 288-292).
  • FGFR3 is also often expressed in multiple myeloma-type cancers.
  • Inhibitors of FGFR3 activity are useful in the treatment of T-cell mediated inflammatory or autoimmune diseases including but not limited to rheumatoid arthritis (RA), collagen II arthritis, multiple sclerosis (MS), systemic lupus erythematosus (SLE), psoriasis, juvenile onset diabetes, Sjogren's disease, thyroid disease, sarcoidosis, autoimmune uveitis, inflammatory bowel disease (Crohn's and ulcerative colitis), celiac disease and myasthenia gravis.
  • RA rheumatoid arthritis
  • MS multiple sclerosis
  • SLE systemic lupus erythematosus
  • psoriasis juvenile onset diabetes
  • Sjogren's disease thyroid disease
  • sarcoidosis autoimmune uveitis
  • inflammatory bowel disease Crohn's and ulcerative colitis
  • Tie2 inhibitors can be used in situations where neovascularization takes place inappropriately (i.e. in diabetic retinopathy, chronic inflammation, psoriasis, Kaposi's sarcoma, chronic neovascularization due to macular degeneration, rheumatoid arthritis, infantile haemangioma and cancers).
  • Lck plays a role in T-cell signaling. Mice that lack the Lck gene have a poor ability to develop thymocytes. The function of Lck as a positive activator of T-cell signaling suggests that Lck inhibitors may be useful for treating autoimmune disease such as rheumatoid arthritis.
  • JNKs have been implicated in having a role in mediating cellular response to cancer, thrombin-induced platelet aggregation, immunodeficiency disorders, autoimmune diseases, cell death, allergies, osteoporosis and heart disease.
  • the therapeutic targets related to activation of the JNK pathway include chronic myelogenous leukemia (CML), rheumatoid arthritis, asthma, osteoarthritis, ischemia, cancer and neurodegenerative diseases.
  • CML chronic myelogenous leukemia
  • rheumatoid arthritis rheumatoid arthritis
  • asthma rheumatoid arthritis
  • ischemia ischemia
  • compounds of the invention may also be useful to treat various hepatic disorders.
  • JNK Kaposi's sarcoma
  • VEGF vascular endothelial growth factor
  • IL-6 IL-6
  • TNF ⁇ vascular endothelial growth factor
  • Certain abnormal proliferative conditions are believed to be associated with raf expression and are, therefore, believed to be responsive to inhibition of raf expression. Abnormally high levels of expression of the raf protein are also implicated in transformation and abnormal cell proliferation. These abnormal proliferative conditions are also believed to be responsive to inhibition of raf expression. For example, expression of the c-raf protein is believed to play a role in abnormal cell proliferation since it has been reported that 60% of all lung carcinoma cell lines express unusually high levels of c-raf mRNA and protein.
  • abnormal proliferative conditions are hyper- proliferative disorders such as cancers, tumors, hyperplasia, pulmonary fibrosis, angiogenesis, psoriasis, atherosclerosis and smooth muscle cell proliferation in the blood vessels, such as stenosis or restenosis following angioplasty.
  • the cellular signaling pathway of which raf is a part has also been implicated in inflammatory disorders characterized by T- cell proliferation (T-cell activation and growth), such as tissue graft rejection, endotoxin shock, and glomerular nephritis, for example.
  • the stress activated protein kinases are a family of protein kinases that represent the penultimate step in signal transduction pathways that result in activation of the c-jun transcription factor and expression of genes regulated by c-jun.
  • c-jun is involved in the transcription of genes that encode proteins involved in the repair of DNA that is damaged due to genotoxic insults. Therefore, agents that inhibit SAPK activity in a cell prevent DNA repair and sensitize the cell to agents that induce DNA damage or inhibit DNA synthesis and induce apoptosis of a cell or that inhibit cell proliferation.
  • MAPKs Mitogen-activated protein kinases
  • MKKs mitogen- activated protein kinase kinases
  • Ribosomal protein S6 protein kinases consist of at least 8 members (RSKl, RSK2, RSK3, RSK4, MSKl, MSK2, p70S6K and p70S6 Kb). Ribosomal protein S6 protein kinases play important pleotropic functions, among them is a key role in the regulation of mRNA translation during protein biosynthesis (Eur. J. Biochem 2000 November; 267(21): 6321-30, Exp Cell Res. Nov. 25, 1999; 253 (1): 100-9, MoI Cell Endocrinol. May 25, 1999;151(l-2):65-77).
  • the phosphorylation of the S6 ribosomal protein by p70S6 has also been implicated in the regulation of cell motility (Immunol. Cell Biol. 2000 August;78(4):447-51) and cell growth (Prog. Nucleic Acid Res. MoI. Biol., 2000;65: 101-27), and hence, may be important in tumor metastasis, the immune response and tissue repair as well as other disease conditions.
  • SAPK's also called "jun N-terminal kinases” or “JNK's”
  • JNK's are a family of protein kinases that represent the penultimate step in signal transduction pathways that result in activation of the c-jun transcription factor and expression of genes regulated by c- jun.
  • c-jun is involved in the transcription of genes that encode proteins involved in the repair of DNA that is damaged due to genotoxic insults.
  • Agents that inhibit SAPK activity in a cell prevent DNA repair and sensitize the cell to those cancer therapeutic modalities that act by inducing DNA damage.
  • BTK plays a role in autoimmune and/or inflammatory disease such as systemic lupus erythematosus (SLE), rheumatoid arthritis, multiple vasculitides, idiopathic thrombocytopenic purpura (ITP), myasthenia gravis, and asthma.
  • SLE systemic lupus erythematosus
  • ITP idiopathic thrombocytopenic purpura
  • myasthenia gravis myasthenia gravis
  • asthma chronic lung disease
  • inhibitors of BTK are useful as inhibitors of B-cell mediated pathogenic activity, such as autoantibody production, and are useful for the treatment of B-cell lymphoma and leukemia.
  • CHK2 is a member of the checkpoint kinase family of serine/threonine protein kinases and is involved in a mechanism used for surveillance of DNA damage, such as damage caused by environmental mutagens and endogenous reactive oxygen species. As a result, it is implicated as a tumor suppressor and target for cancer therapy.
  • CSK influences the metastatic potential of cancer cells, particularly colon cancer.
  • Fes is a non-receptor protein tyrosine kinase that has been implicated in a variety of cytokine signal transduction pathways, as well as differentiation of myeloid cells. Fes is also a key component of the granulocyte differentiation machinery.
  • Flt3 receptor tyrosine kinase activity is implicated in leukemias and myelodysplastic syndrome. In approximately 25% of AML the leukemia cells express a constitutively active form of auto-phosphorylated (p) FLT3 tyrosine kinase on the cell surface. The activity of p-FLT3 confers growth and survival advantage on the leukemic cells.
  • Inhibitors of IKK ⁇ and IKK ⁇ (1 & 2) are therapeutics for diseases which include rheumatoid arthritis, transplant rejection, inflammatory bowel disease, osteoarthritis, asthma, chronic obstructive pulmonary disease, atherosclerosis, psoriasis, multiple sclerosis, stroke, systemic lupus erythematosus, Alzheimer's disease, brain ischemia, traumatic brain injury, Parkinson's disease, amyotrophic lateral sclerosis, subarachnoid hemorrhage or other diseases or disorders associated with excessive production of inflammatory mediators in the brain and central nervous system.)
  • Met is associated with most types of the major human cancers and expression is often correlated with poor prognosis and metastasis.
  • Inhibitors of Met are therapeutics for diseases which include cancers such as lung cancer, NSCLC (non small cell lung cancer), bone cancer, pancreatic cancer, skin cancer, cancer of the head and neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, colon cancer, breast cancer, gynecologic tumors (e.
  • uterine sarcomas carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina or carcinoma of the vulva
  • Hodgkin's Disease cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system (e. g., cancer of the thyroid, parathyroid or adrenal glands), sarcomas of soft tissues, cancer of the urethra, cancer of the penis, prostate cancer, chronic or acute leukemia, solid tumors of childhood, lymphocytic lymphomas, cancer of the bladder, cancer of the kidney or ureter (e.
  • neoplasms of the central nervous system e. g., primary CNS lymphoma, spinal axis tumors, brain stem glioma or pituitary adenomas
  • cancers of the blood such as acute myeloid leukemia, chronic myeloid leukemia, etc, Barrett's esophagus (pre-malignant syndrome) neoplastic cutaneous disease, psoriasis, mycoses fungoides and benign prostatic hypertrophy
  • diabetes related diseases such as diabetic retinopathy, retinal ischemia and retinal neovascularization, hepatic cirrhosis
  • cardiovascular disease such as atherosclerosis
  • immunological disease such as autoimmune disease and renal disease.
  • the disease is cancer such as acute myeloid leukemia and colorectal cancer.
  • the Nima-related kinase 2 (Nek2) is a cell cycle-regulated protein kinase with maximal activity at the onset of mitosis that localizes to the centrosome. Functional studies have implicated Nelc2 in regulation of centrosome separation and spindle formation. Nek2 protein is elevated 2- to 5-fold in cell lines derived from a range of human tumors including those of cervical, ovarian, prostate, and particularly breast.
  • p70S6K-mediated diseases or conditions include, but are not limited to, proliferative disorders, such as cancer and tuberous sclerosis.
  • 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) of a compound of Formula I or a pharmaceutically acceptable salt thereof.
  • 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 2.5mg/kg per body weight.
  • An indicated daily dosage in the larger mammal, e.g. humans, is in the range from about 0.5mg to about lOOmg, 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 50mg 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 combination 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 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.
  • 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
  • n, Ri, R 2j R 3 and R 4 are as defined in the Summary of the
  • a compound of Formula I can be synthesized by reacting a compound of formula 2 in the presence of a suitable solvent (for example, THF, and the like), a suitable reactive chemical intermediate (for example, phenyl chloroformate, and the like) and a suitable base (for example, triethylamine, and the like).
  • a suitable solvent for example, THF, and the like
  • a suitable reactive chemical intermediate for example, phenyl chloroformate, and the like
  • a suitable base for example, triethylamine, and the like.
  • the reaction proceeds in a temperature range of about O 0 C to about 4O 0 C and can take up to about 10 hours to complete.
  • the reaction mixture is further reacted in the presence of a suitable strong base (for example, sodium hydroxide, and the like), proceeding at a temperature range of about 6O 0 C to about 8O 0 C and can take up to about 24 hours to complete.
  • a suitable strong base for example, sodium hydroxide, and 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
  • 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 8O 0 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 carbamylating 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: (a) that of reaction scheme I;
  • the reaction mixture is cooled to room temperature and condensed to remove most of the T ⁇ F solvent.
  • the precipitate is collected by filtration and washed with water.
  • the resultant solid is then suspended in 3 ml water and acidified to a p ⁇ between 4 and 5 using 2N HCl aqueous solution.
  • the mixture is kept stirring for 30 minutes, the undissolved solid was collected via filtration and washed with water and ethyl ether, and dried to afford the title compound as pale yellow powder.
  • reaction mixture After stirring for 1.5 hours at 5O 0 C, the reaction mixture is neutralized with saturated sodium bicarbonate solution to a p ⁇ of 8. The solid is collected by filtration and washed with water, then hexanes, and dried to afford the title compound as a light yellow solid.
  • Compounds of the present invention are assayed to measure their capacity to selectively inhibit cell proliferation of 32D cells expressing BCR-AbI (32D-p210) compared with parental 32D cells. Compounds selectively inhibiting the proliferation of these BCR-AbI transformed cells are tested for antiproliferative activity on Ba/F3 cells expressing either wild type or the mutant forms of Bcr-abl.
  • compounds are assayed to measure their capacity to inhibit FGFR3, b-RAF, AbI, BMX, BTK, CHK2, c- RAF, CSK, c-SRC, Fes, FItS, IKK ⁇ , IKK ⁇ , JNK2 ⁇ 2, Lck, Met, MKK4, MKK6, MST2, NEK2, p70S6K, PDGFR ⁇ , PKA, PKB ⁇ , PKD2, Rskl, SAPK2 ⁇ , SAPK2 ⁇ , SAPK3, SGK, Tie2 and TrkB kinases.
  • the murine cell line used is the 32D hemopoietic progenitor cell line transformed with BCR-AbI 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.
  • 50nl 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 nni) 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 570nm is quantified spectrophotometrically and IC 50 values, the concentration of compound required for 50% inhibition, determined from a dose response curve.
  • BCR-AbI 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 2x10 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-AbI expressing cells, inhibit the cellular BCR-AbI autophosphorylation in a dose-dependent manner.
  • Ba/F3 cells expressing either wild type or the mutant forms of BCR-AbI (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 ⁇ M as described above (in media lacking IL3).
  • the IC5 0 values of the compounds lacking toxicity on the untransformed cells were determined from the dose response curves obtained as describe above.
  • Kinase activity assay with purified FGFR3 (Upstate) is carried out in a final volume of 10 ⁇ L containing 0.25 ⁇ g/mL of enzyme in kinase buffer (30 mM Tris-HCl ⁇ H7.5, 15 mM MgCl 2 , 4.5 mM MnCl 2 , 15 ⁇ M Na 3 VO 4 and 50 ⁇ g/mL BSA), and substrates (5 ⁇ g/mL biotin-poly-EY(Glu, Tyr) (CIS-US, Inc.) and 3 ⁇ M ATP).
  • the first solution of 5 ⁇ l contains the FGFR3 enzyme in kinase buffer was first dispensed into 384- format ProxiPlate® (Perkin-Elmer) followed by adding 50 nL of compounds dissolved in DMSO, then 5 ⁇ l of second solution contains the substrate (poly- EY) and ATP in kinase buffer was added to each wells.
  • the reactions are incubated at room temperature for one hour, stopped by adding 10 ⁇ L of HTRF detection mixture, which contains 30 mM Tris-HCl pH7.5, 0.5 M KF, 50 mM ETDA, 0.2 mg/mL BSA, 15 ⁇ g/mL streptavidin-XL665 (CIS-US, Inc.) and 150 ng/mL cryptate conjugated anti-phosphotyrosine antibody (CIS-US, Inc.). After one hour of room temperature incubation to allow for streptavidin-biotin interaction, time resolved florescent signals are read on Analyst GT (Molecular Devices Corp.).
  • IC 50 values are calculated by linear regression analysis of the percentage inhibition of each compound at 12 concentrations (1:3 dilution from 50 ⁇ M to 0.28 nM). In this assay, compounds of the invention have an IC 50 in the range of 10 nM to 2 ⁇ M.
  • Compounds of the invention are tested for their ability to inhibit transformed Ba/F3-TEL-FGFR3 cells proliferation, which is depended on FGFR3 cellular kinase activity.
  • Ba/F3-TEL-FGFR3 are cultured up to 800,000 cells/mL in suspension, with RPMI 1640 supplemented with 10% fetal bovine serum as the culture medium. Cells are dispensed into 384- well format plate at 5000 cell/well in 50 ⁇ L culture medium.
  • Compounds of the invention are dissolved and diluted in dimethylsufoxide (DMSO). Twelve points 1:3 serial dilutions are made into DMSO to create concentrations gradient ranging typically from 10 mM to 0.05 ⁇ M.
  • DMSO dimethylsufoxide
  • AlamarBlue® (TREK Diagnostic Systems), which can be used to monitor the reducing environment created by proliferating cells, are added to cells at final concentration of 10%. After additional four hours of incubation in a 37 °C cell culture incubator, fluorescence signals from reduced AlamarBlue® (Excitation at 530 nm, Emission at 580 ran) are quantified on Analyst GT (Molecular Devices Corp.). IC 5 O values are calculated by linear regression analysis of the percentage inhibition of each compound at 12 concentrations.
  • Assay buffer containing 20 ⁇ M ATP (lO ⁇ l) is added to each well followed by lOOnl or 500nl of compound.
  • B-Raf is diluted in the assay buffer (l ⁇ l into 25 ⁇ l) and lO ⁇ 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 0 C overnight and washed 6 times with TBST.
  • A375 cell line (ATCC) is derived from a human melanoma patient and it has a V599E mutation on the B-Raf gene. The levels of phosphorylated MEK are elevated due to the mutation of B-Raf.
  • Sub-confluent to confluent A375 cells are incubated with compounds for 2 hours at 37 0 C in serum free medium. Cells are then washed once with cold PBS and lysed with the lysis buffer containing 1% Triton XlOO. After centrifugation, the supernatants are subjected to SDS-PAGE, and then transferred to nitrocellulose membranes.
  • the membranes are then subjected to western blotting with anti-phospho-MEK antibody (ser217/221) (Cell Signaling).
  • the amount of phosphorylated MEK is monitored by the density of phospho-MEK bands on the nitrocellulose membranes.
  • Upstate KinaseProfilerTM Radio-enzymatic filter binding assay
  • Kinase buffer (2.5 ⁇ L, 1Ox - 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 .01mg/ml) in kinase buffer and kinase buffer (50 ⁇ M; 5 ⁇ L) are mixed in an eppendorf on ice.
  • a Mg/ATP mix (lO ⁇ L; 67.5 (or 33.75) mM MgCl 2 , 450 (or 225) ⁇ M ATP and 1 ⁇ Ci/ ⁇ l [ ⁇ - 32 P]-ATP (3000Ci/mmol)) is added and the reaction is incubated at about 3O 0 C for about 10 minutes.
  • the reaction mixture is spotted (20 ⁇ L) onto a 2cm x 2cm 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 x 10 "10 to 1 x 10 "5 M, preferably less than 50OnM, 25OnM, 10OnM and 5OnM for wild type BCR-AbI and G250E, E255V, T315I, F317L and M351T BCR-AbI mutants.
  • Compounds of Formula I preferably, at a concentration of lO ⁇ M, preferably show a percentage inhibition of greater than 50%, preferably greater than about 70%, against AbI, BMX, BTK, CHK2, b-RAF, c-RAF, CSK, c-SRC, Fes, FGFR3, Flt3, IKK ⁇ , IKK ⁇ , JNK2 ⁇ 2, LcIc, Met, MKK4, MKK6, MST2, NEK2, p70S6K, PDGFR ⁇ , PKA, PKB ⁇ , PKD2, Rskl, SAPK2 ⁇ , SAPK2 ⁇ , SAPK3, SGK, Tie2 and/or TrkB kinases.
  • N-ethyl-3-r9-ethyl-8-oxo-3.6.8.9-tetrahvdro-3.4,7.9-tetraaza- cyclopenta[ " a1naphthalen-7-vD-5-methoxy-benzamide (Example 1) shows an IC50 of 1OnM, 1OnM, 6nm and 1 InM in the FGFR3 en2ymatic assay, FGFR3 cellular assay, FGFRl cellular assay and FGFR4 cellular assay, respectively.
  • a) has an IC 50 of ⁇ 0.5 nM, 59 nM, 44 nM, 38 nM ⁇ 0.5 nM and ⁇ 0.5 nM for wild type, G250E, E255V, T315I, F317L and M351T Bcr-abl, respectively;
  • b has an IC 50 of 15 nM, ⁇ 260 nM for b-RAF en ⁇ ymatic and cellular assays, respectively;

Abstract

L'invention concerne une nouvelle classe de composés, des compositions pharmaceutiques comprenant ces composés, et des procédés d'utilisation desdits composés pour traiter ou prévenir des maladies ou des troubles qui sont associé(e)s à une activité anormale ou déréglée de kinases, en particulier des maladies ou des troubles impliquant une activation anormale des kinases Abl, Bcr-Abl, BMX, BTK, CHK2, b-RAF, c-RAF, CSK, c-SRC, Fes, FGFR3, Flt3, IKKα, IKKβ, JNK2α2, Lck, Met, MKK4, MKK6, MST2, NEK2, p70S6K, PDGFRβ, PKA, PKBα, PKD2, Rsk1, SAPK2α, SAPK2β, SAPK3, SGK, Tie2, et TrkB.
PCT/US2006/018644 2005-05-12 2006-05-12 Composes et compositions utilise(e)s en tant qu'inhibiteurs de proteines-kinases WO2006124731A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US68089205P 2005-05-12 2005-05-12
US60/680,892 2005-05-12

Publications (2)

Publication Number Publication Date
WO2006124731A2 true WO2006124731A2 (fr) 2006-11-23
WO2006124731A3 WO2006124731A3 (fr) 2007-03-29

Family

ID=37074595

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/018644 WO2006124731A2 (fr) 2005-05-12 2006-05-12 Composes et compositions utilise(e)s en tant qu'inhibiteurs de proteines-kinases

Country Status (1)

Country Link
WO (1) WO2006124731A2 (fr)

Cited By (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009098144A1 (fr) * 2008-02-05 2009-08-13 F. Hoffmann-La Roche Ag Nouvelles pyridinones et pyridazinones
US7683064B2 (en) 2008-02-05 2010-03-23 Roche Palo Alto Llc Inhibitors of Bruton's tyrosine kinase
JP2010514690A (ja) * 2006-12-22 2010-05-06 インサイト・コーポレイション Janusキナーゼ阻害剤としての置換複素環
US7902194B2 (en) 2008-06-24 2011-03-08 Roche Palo Alto Inhibitors of bruton's tyrosine kinase
CN102399233A (zh) * 2010-09-15 2012-04-04 山东轩竹医药科技有限公司 PI3K和mTOR双重抑制剂类化合物
US8338452B2 (en) 2008-02-29 2012-12-25 Array Biopharma Inc. Raf inhibitor compounds and methods of use thereof
US8394795B2 (en) 2008-02-29 2013-03-12 Array Biopharma Inc. Pyrazole [3, 4-B] pyridine Raf inhibitors
US8481540B2 (en) 2010-08-12 2013-07-09 Hoffmann-La Roche Inc. Inhibitors of bruton's tyrosine kinase
US8536166B2 (en) 2008-07-02 2013-09-17 Roche Palo Alto Llc Inhibitors of Burton's tyrosine kinase
EP2671891A2 (fr) 2008-06-27 2013-12-11 Amgen Inc. Inhibition d'ang-2 pour traiter la sclérose en plaques
US20130338134A1 (en) * 2012-06-13 2013-12-19 Incyte Corporation Substituted tricyclic compounds as fgfr inhibitors
US9708318B2 (en) 2015-02-20 2017-07-18 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9745311B2 (en) 2012-08-10 2017-08-29 Incyte Corporation Substituted pyrrolo[2,3-b]pyrazines as FGFR inhibitors
US9801889B2 (en) 2015-02-20 2017-10-31 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9890156B2 (en) 2015-02-20 2018-02-13 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10040790B2 (en) 2013-04-19 2018-08-07 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US10213427B2 (en) 2010-12-22 2019-02-26 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US10611762B2 (en) 2017-05-26 2020-04-07 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
US10851105B2 (en) 2014-10-22 2020-12-01 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10870651B2 (en) 2014-12-23 2020-12-22 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US10906889B2 (en) 2013-10-18 2021-02-02 Dana-Farber Cancer Institute, Inc. Polycyclic inhibitors of cyclin-dependent kinase 7 (CDK7)
US10981903B2 (en) 2011-11-17 2021-04-20 Dana-Farber Cancer Institute, Inc. Inhibitors of c-Jun-N-terminal kinase (JNK)
US11091447B2 (en) 2020-01-03 2021-08-17 Berg Llc UBE2K modulators and methods for their use
US11142507B2 (en) 2015-09-09 2021-10-12 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
US11174250B2 (en) 2016-03-01 2021-11-16 Propellon Therapeutics Inc. Substituted carboxamides as inhibitors of WDR5 protein-protein binding
US11174257B2 (en) 2018-05-04 2021-11-16 Incyte Corporation Salts of an FGFR inhibitor
US11319299B2 (en) 2016-03-01 2022-05-03 Propellon Therapeutics Inc. Substituted carboxamides as inhibitors of WDR5 protein-protein binding
US11325910B2 (en) 2015-03-27 2022-05-10 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
US11407750B2 (en) 2019-12-04 2022-08-09 Incyte Corporation Derivatives of an FGFR inhibitor
WO2022193227A1 (fr) * 2021-03-18 2022-09-22 Nutshell Biotech (Shanghai) Co., Ltd. Composés cycliques fusionnés utiles en tant qu'inhibiteurs de tyrosine kinases fgfr
US11466004B2 (en) 2018-05-04 2022-10-11 Incyte Corporation Solid forms of an FGFR inhibitor and processes for preparing the same
US11607416B2 (en) 2019-10-14 2023-03-21 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11628162B2 (en) 2019-03-08 2023-04-18 Incyte Corporation Methods of treating cancer with an FGFR inhibitor
US11826365B2 (en) 2009-12-29 2023-11-28 Dana-Farber Cancer Institute, Inc. Type II raf kinase inhibitors
US11897891B2 (en) 2019-12-04 2024-02-13 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors
US11939331B2 (en) 2021-06-09 2024-03-26 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9266892B2 (en) 2012-12-19 2016-02-23 Incyte Holdings Corporation Fused pyrazoles as FGFR inhibitors
WO2021007269A1 (fr) 2019-07-09 2021-01-14 Incyte Corporation Hétérocycles bicycliques en tant qu'inhibiteurs de fgfr
WO2021076728A1 (fr) 2019-10-16 2021-04-22 Incyte Corporation Hétérocycles bicycliques en tant qu'inhibiteurs de fgfr

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005034869A2 (fr) * 2003-10-08 2005-04-21 Irm Llc Composes et compositions convenant comme inhibiteurs de proteine-kinases
WO2005105097A2 (fr) * 2004-04-28 2005-11-10 Gpc Biotech Ag Derives de pyridopyrimidine

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005034869A2 (fr) * 2003-10-08 2005-04-21 Irm Llc Composes et compositions convenant comme inhibiteurs de proteine-kinases
WO2005105097A2 (fr) * 2004-04-28 2005-11-10 Gpc Biotech Ag Derives de pyridopyrimidine

Cited By (92)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2010514690A (ja) * 2006-12-22 2010-05-06 インサイト・コーポレイション Janusキナーゼ阻害剤としての置換複素環
US8513270B2 (en) 2006-12-22 2013-08-20 Incyte Corporation Substituted heterocycles as Janus kinase inhibitors
JP2011511027A (ja) * 2008-02-05 2011-04-07 エフ.ホフマン−ラ ロシュ アーゲー 新規ピリジノン及びピリダジノン
RU2505538C2 (ru) * 2008-02-05 2014-01-27 Ф.Хоффманн-Ля Рош Аг Новые пиридиноны и пиридазиноны
US7683064B2 (en) 2008-02-05 2010-03-23 Roche Palo Alto Llc Inhibitors of Bruton's tyrosine kinase
US7906509B2 (en) 2008-02-05 2011-03-15 Roche Palo Alto Llc Inhibitors of bruton's tyrosine kinase
AU2009211514B2 (en) * 2008-02-05 2014-02-20 F. Hoffmann-La Roche Ag Novel pyridinones and pyridazinones
KR101308803B1 (ko) 2008-02-05 2013-09-13 에프. 호프만-라 로슈 아게 신규한 피리디논 및 피리다지논
CN101932573A (zh) * 2008-02-05 2010-12-29 霍夫曼-拉罗奇有限公司 新型吡啶酮类和哒嗪酮类
WO2009098144A1 (fr) * 2008-02-05 2009-08-13 F. Hoffmann-La Roche Ag Nouvelles pyridinones et pyridazinones
US8338452B2 (en) 2008-02-29 2012-12-25 Array Biopharma Inc. Raf inhibitor compounds and methods of use thereof
US8394795B2 (en) 2008-02-29 2013-03-12 Array Biopharma Inc. Pyrazole [3, 4-B] pyridine Raf inhibitors
US7902194B2 (en) 2008-06-24 2011-03-08 Roche Palo Alto Inhibitors of bruton's tyrosine kinase
US8124604B2 (en) 2008-06-24 2012-02-28 Roche Palo Alto Llc Inhibitors of bruton's tyrosine kinase
US8618098B2 (en) 2008-06-24 2013-12-31 Roche Palo Alto Llc Inhibitors of bruton's tyrosine kinase
US8822457B2 (en) 2008-06-24 2014-09-02 Roche Palo Alto Llc Inhibitors of bruton's tyrosine kinase
EP2671891A2 (fr) 2008-06-27 2013-12-11 Amgen Inc. Inhibition d'ang-2 pour traiter la sclérose en plaques
US8536166B2 (en) 2008-07-02 2013-09-17 Roche Palo Alto Llc Inhibitors of Burton's tyrosine kinase
US11826365B2 (en) 2009-12-29 2023-11-28 Dana-Farber Cancer Institute, Inc. Type II raf kinase inhibitors
US8481540B2 (en) 2010-08-12 2013-07-09 Hoffmann-La Roche Inc. Inhibitors of bruton's tyrosine kinase
US8940741B2 (en) 2010-08-12 2015-01-27 Hoffmann-La Roche Inc. Inhibitors of bruton's tyrosine kinase
CN102399233A (zh) * 2010-09-15 2012-04-04 山东轩竹医药科技有限公司 PI3K和mTOR双重抑制剂类化合物
US10813930B2 (en) 2010-12-22 2020-10-27 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US10213427B2 (en) 2010-12-22 2019-02-26 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US10981903B2 (en) 2011-11-17 2021-04-20 Dana-Farber Cancer Institute, Inc. Inhibitors of c-Jun-N-terminal kinase (JNK)
CN104507943B (zh) * 2012-06-13 2017-11-21 因塞特控股公司 作为fgfr抑制剂的取代的三环化合物
AU2013287176C1 (en) * 2012-06-13 2023-01-19 Incyte Holdings Corporation Substituted tricyclic compounds as FGFR inhibitors
US20170137424A1 (en) * 2012-06-13 2017-05-18 Incyte Corporation Substituted tricyclic compounds as fgfr inhibitors
EP3176170A1 (fr) * 2012-06-13 2017-06-07 Incyte Holdings Corporation Composés tricycliques substitués servant d'inhibiteurs des fgfr
IL289834B1 (en) * 2012-06-13 2024-03-01 Incyte Holdings Corp Conversion of tricyclic compounds as FGFR inhibitors
KR20220080213A (ko) * 2012-06-13 2022-06-14 인사이트 홀딩스 코포레이션 Fgfr 억제제로서 치환된 트리사이클릭 화합물
US11840534B2 (en) 2012-06-13 2023-12-12 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
KR102406771B1 (ko) * 2012-06-13 2022-06-13 인사이트 홀딩스 코포레이션 Fgfr 억제제로서 치환된 트리사이클릭 화합물
CN107383009A (zh) * 2012-06-13 2017-11-24 因塞特控股公司 作为fgfr抑制剂的取代的三环化合物
JP2017222709A (ja) * 2012-06-13 2017-12-21 インサイト・ホールディングス・コーポレイションIncyte Holdings Corporation Fgfr阻害剤としての置換三環式化合物
CN107652289A (zh) * 2012-06-13 2018-02-02 因塞特控股公司 作为fgfr抑制剂的取代的三环化合物
JP7392096B2 (ja) 2012-06-13 2023-12-05 インサイト・ホールディングス・コーポレイション Fgfr阻害剤としての置換三環式化合物
JP2015521600A (ja) * 2012-06-13 2015-07-30 インサイト・コーポレイションIncyte Corporation Fgfr阻害剤としての置換三環式化合物
TWI769529B (zh) * 2012-06-13 2022-07-01 美商英塞特控股公司 作為纖維母細胞生長因子受體抑制劑之經取代三環化合物
AU2013287176B2 (en) * 2012-06-13 2018-10-04 Incyte Holdings Corporation Substituted tricyclic compounds as FGFR inhibitors
US10131667B2 (en) * 2012-06-13 2018-11-20 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
KR102556118B1 (ko) * 2012-06-13 2023-07-19 인사이트 홀딩스 코포레이션 Fgfr 억제제로서 치환된 트리사이클릭 화합물
CN104507943A (zh) * 2012-06-13 2015-04-08 因塞特公司 作为fgfr抑制剂的取代的三环化合物
TWI801156B (zh) * 2012-06-13 2023-05-01 美商英塞特控股公司 作為纖維母細胞生長因子受體抑制劑之經取代三環化合物
EP3495367A1 (fr) * 2012-06-13 2019-06-12 Incyte Holdings Corporation Composés tricycliques substitués servant d'inhibiteurs des fgfr
US11053246B2 (en) * 2012-06-13 2021-07-06 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
US9611267B2 (en) * 2012-06-13 2017-04-04 Incyte Holdings Corporation Substituted tricyclic compounds as FGFR inhibitors
KR20150036044A (ko) * 2012-06-13 2015-04-07 인사이트 코포레이션 Fgfr 억제제로서 치환된 트리사이클릭 화합물
CN107383009B (zh) * 2012-06-13 2020-06-09 因塞特控股公司 作为fgfr抑制剂的取代的三环化合物
CN107652289B (zh) * 2012-06-13 2020-07-21 因塞特控股公司 作为fgfr抑制剂的取代的三环化合物
KR102140426B1 (ko) * 2012-06-13 2020-08-04 인사이트 홀딩스 코포레이션 Fgfr 억제제로서 치환된 트리사이클릭 화합물
KR20200093696A (ko) * 2012-06-13 2020-08-05 인사이트 홀딩스 코포레이션 Fgfr 억제제로서 치환된 트리사이클릭 화합물
AU2020270520B2 (en) * 2012-06-13 2023-01-05 Incyte Holdings Corporation Substituted tricyclic compounds as fgfr inhibitors
AU2019200066B2 (en) * 2012-06-13 2020-08-27 Incyte Holdings Corporation Substituted tricyclic compounds as fgfr inhibitors
WO2014007951A3 (fr) * 2012-06-13 2014-03-06 Incyte Corporation Composés tricycliques substitués utilisés comme inhibiteurs de fgfr
EA036592B1 (ru) * 2012-06-13 2020-11-26 Инсайт Холдингс Корпорейшн Замещенные трициклические соединения как ингибиторы fgfr
EP3822273A1 (fr) * 2012-06-13 2021-05-19 Incyte Holdings Corporation Composés tricycliques substitués servant d'inhibiteurs des fgfr
US20130338134A1 (en) * 2012-06-13 2013-12-19 Incyte Corporation Substituted tricyclic compounds as fgfr inhibitors
US9745311B2 (en) 2012-08-10 2017-08-29 Incyte Corporation Substituted pyrrolo[2,3-b]pyrazines as FGFR inhibitors
US10947230B2 (en) 2013-04-19 2021-03-16 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11530214B2 (en) 2013-04-19 2022-12-20 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US10450313B2 (en) 2013-04-19 2019-10-22 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US10040790B2 (en) 2013-04-19 2018-08-07 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US10906889B2 (en) 2013-10-18 2021-02-02 Dana-Farber Cancer Institute, Inc. Polycyclic inhibitors of cyclin-dependent kinase 7 (CDK7)
US10851105B2 (en) 2014-10-22 2020-12-01 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10870651B2 (en) 2014-12-23 2020-12-22 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US10632126B2 (en) 2015-02-20 2020-04-28 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10214528B2 (en) 2015-02-20 2019-02-26 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9708318B2 (en) 2015-02-20 2017-07-18 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9801889B2 (en) 2015-02-20 2017-10-31 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9890156B2 (en) 2015-02-20 2018-02-13 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11173162B2 (en) 2015-02-20 2021-11-16 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10016438B2 (en) 2015-02-20 2018-07-10 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11667635B2 (en) 2015-02-20 2023-06-06 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10251892B2 (en) 2015-02-20 2019-04-09 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10738048B2 (en) 2015-02-20 2020-08-11 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11014923B2 (en) 2015-02-20 2021-05-25 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11325910B2 (en) 2015-03-27 2022-05-10 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
US11142507B2 (en) 2015-09-09 2021-10-12 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
US11174250B2 (en) 2016-03-01 2021-11-16 Propellon Therapeutics Inc. Substituted carboxamides as inhibitors of WDR5 protein-protein binding
US11319299B2 (en) 2016-03-01 2022-05-03 Propellon Therapeutics Inc. Substituted carboxamides as inhibitors of WDR5 protein-protein binding
US10611762B2 (en) 2017-05-26 2020-04-07 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
US11472801B2 (en) 2017-05-26 2022-10-18 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
US11466004B2 (en) 2018-05-04 2022-10-11 Incyte Corporation Solid forms of an FGFR inhibitor and processes for preparing the same
US11174257B2 (en) 2018-05-04 2021-11-16 Incyte Corporation Salts of an FGFR inhibitor
US11628162B2 (en) 2019-03-08 2023-04-18 Incyte Corporation Methods of treating cancer with an FGFR inhibitor
US11607416B2 (en) 2019-10-14 2023-03-21 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11407750B2 (en) 2019-12-04 2022-08-09 Incyte Corporation Derivatives of an FGFR inhibitor
US11897891B2 (en) 2019-12-04 2024-02-13 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors
US11091447B2 (en) 2020-01-03 2021-08-17 Berg Llc UBE2K modulators and methods for their use
WO2022193227A1 (fr) * 2021-03-18 2022-09-22 Nutshell Biotech (Shanghai) Co., Ltd. Composés cycliques fusionnés utiles en tant qu'inhibiteurs de tyrosine kinases fgfr
US11939331B2 (en) 2021-06-09 2024-03-26 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors

Also Published As

Publication number Publication date
WO2006124731A3 (fr) 2007-03-29

Similar Documents

Publication Publication Date Title
US7868018B2 (en) Compounds and compositions as protein kinase inhibitors
US8183248B2 (en) Substituted pyrrolo[2,3-d]pyrimidines and compositions as protein kinase inhibitors
US7589101B2 (en) Compounds and compositions as protein kinase inhibitors
EP1940844B1 (fr) Composes et compositions utilises comme inhibiteurs des kinases des proteines
EP2027123B1 (fr) Composes inhibiteurs de proteines kinases et compositions comprenant de tels composes
WO2006124731A2 (fr) Composes et compositions utilise(e)s en tant qu'inhibiteurs de proteines-kinases
EP1896470A2 (fr) Composes et compositions en tant qu'inhibiteurs de proteine kinase
WO2006002367A1 (fr) Composes et compositions utilises en tant qu'inhibiteurs des proteines kinases
KR20070030848A (ko) 단백질 키나제 억제제 화합물 및 조성물

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase in:

Ref country code: DE

NENP Non-entry into the national phase in:

Ref country code: RU

122 Ep: pct application non-entry in european phase

Ref document number: 06759800

Country of ref document: EP

Kind code of ref document: A2