WO2008124393A1 - Benzothiazole derivatives and their use as protein kinase inhibitors - Google Patents

Benzothiazole derivatives and their use as protein kinase inhibitors Download PDF

Info

Publication number
WO2008124393A1
WO2008124393A1 PCT/US2008/059024 US2008059024W WO2008124393A1 WO 2008124393 A1 WO2008124393 A1 WO 2008124393A1 US 2008059024 W US2008059024 W US 2008059024W WO 2008124393 A1 WO2008124393 A1 WO 2008124393A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
independently
alkyl
kinase
compounds
Prior art date
Application number
PCT/US2008/059024
Other languages
French (fr)
Inventor
Yun He
Zuosheng Liu
Shenlin Huang
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 WO2008124393A1 publication Critical patent/WO2008124393A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/60Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings condensed with carbocyclic rings or ring systems
    • C07D277/62Benzothiazoles
    • C07D277/68Benzothiazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2
    • C07D277/82Nitrogen atoms

Definitions

  • the invention relates to protein kinase inhibitors, and methods of using such compounds.
  • 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 (PDGFR), the nerve growth factor receptor, TrkB, Met, and the fibroblast growth factor receptor, FGFR-3 ; non-receptor tyrosine kinases such AbI and the fusion kinase Bcr-Abl, Lck, Csk, Fes, Bmx and Src; and serine/threonine kinases such as B-Raf, C-Raf, Sgk, MAP kinases (e.g., MKK4, MKK6, etc.) and SAPK2 ⁇ , SAPK2 ⁇ and SAPK3.
  • Aberrant kinase activity has been observed in many disease states including benign and malignant prolife
  • the invention provides compounds and pharmaceutical compositions thereof, which may be useful as protein kinase inhibitors.
  • the invention provides compounds having Formula (1):
  • X is a 5-7 membered aryl or heteroaryl containing N, O or S;
  • L is NHCO or CONH;
  • R 1 is H, halo, NR 4 CONR 5 R 6 , NR 4 CO 2 R 5 , NR 4 (CR 2 ) P NR 5 R 6 or NR 4 (CR 2 ) P R 5 ;
  • R 2 is halo, or an optionally halogenated Ci_ 6 alkyl;
  • R 3 is halo, an optionally halogenated Ci_ 6 alkyl, (CR 2 ) q R 7 or O(CR 2 ) q R 8 ;
  • R and R 4 are independently H or C 1-6 alkyl;
  • R and R are independently H, an optionally halogenated Ci_ 6 alkyl, C 2 _ 6 alkenyl or C 2 _ 6 alkynyl; Ci_ 6 alkanol, (CR 2 ) q -R 7 or O(CR 2 ) q R 8 ; or R 5 and R 6 together with N in NR 5 R 6 may form an optionally substituted ring;
  • R 7 and R 8 are independently an optionally substituted C 3 _ 7 cycloalkyl, 5-7 membered aryl, heterocyclic or heteroaryl; or R 8 is H; m is 1-4; and n, p and q are independently 0-4.
  • X may be phenyl, thienyl or pyridyl.
  • R is Ci- 6 alkyl.
  • R 3 is halo, an optionally halogenated C 1-6 alkyl, (CR 2 ) q R 7 , O(CR 2 ) q R 8 ; and R 7 and R 8 are independently an optionally substituted piperidinyl, piperazinyl or imidazolyl.
  • the invention provides compounds of Formula (1), wherein: R 1 is halo, NR 4 CONR 5 R 6 , NR 4 CO 2 R 5 , NR 4 (CR 2 ) P NR 5 R 6 or NR 4 (CR 2 ) P R 5 ; R 5 and R 6 are independently H, Ci_ 6 alkyl, Ci_ 6 alkanol, or (CR 2 ) q R 7 ; R 7 is an optionally substituted C 5 - 7 cycloalkyl pyridyl, phenyl, naphthalenyl, morpholinyl, piperazinyl, pyrimidinyl, imidazolyl, triazolyl, isothiazolyl, isoxazolyl, pyrazolyl or pyrazinyl; and q is 0-1.
  • R 1 is halo, NR 4 CONR 5 R 6 , NR 4 CO 2 R 5 , NR 4 (CR 2 ) P NR 5 R 6 or NR 4 (CR 2
  • R 1 is halo, NR 4 CONR 5 R 6 , NR 4 CO 2 R 5 , NR 4 (CR 2 ) P NR 5 R 6 or NR 4 (CR 2 ) P R 5 ;
  • R 9 is halo or CF 3 ;
  • R 10 is halo, an optionally halogenated C 1-6 alkyl, or OCi_6 alkyl;
  • R 11 is halo, (CR 2 ) q R 7 or O(CR 2 ) q R 8 ;
  • R and R 4 are independently H or Ci_ 6 alkyl;
  • R 5 and R 6 are independently H, an optionally halogenated C 1-6 alkyl, C 2 _6 alkenyl or C 2 _6 alkynyl; Ci_ 6 alkanol, (CR 2 ) q -R 7 or O(CR 2 ) q R 8 ; or R 5 and R 6 together with N in NR 5 R 6 may form an optionally substituted ring;
  • R 7 and R 8 are independently an optionally substituted C 3 _ 7 cycloalkyl, 5-7 membered aryl, heterocyclic or heteroaryl; or R is H; m and n are independently 0-1; and p and q are independently 0-4.
  • R 1 in Formula (1) and (2) is NR 4 CONR 5 R 6 , NR 4 CO 2 R 5 or NR 4 (CR 2 ) P NR 5 R 6 .
  • R 5 and R 6 are independently H, Ci_ 6 alkyl, or (CR 2 ) q R 7 ; and R 7 is an optionally substituted Cs_ 7 cycloalkyl, pyridyl, phenyl, naphthalenyl, morpholinyl, piperazinyl, pyrimidinyl, imidazolyl, triazolyl, isothiazolyl, isoxazolyl, pyrazolyl or pyrazinyl.
  • R 10 is CF 3 .
  • R 11 is halo
  • (CR 2 ) q R 7 , O(CR 2 ) q R 8 and R 7 and R 8 are independently an optionally substituted piperidinyl, piperazinyl or imidazolyl.
  • m and n in compounds of Formula (2) are each 0.
  • suitable substituents will be known to those of ordinary skill in the art, including but not limited to halo, optionally halogenated C 1-6 alkyl, C 2 _ 6 alkenyl, C 2-6 alkynyl, cyano, nitro or (CR 2 ) P R 12 ; wherein R 12 is O(CR 2 ) q R 13 , S(CR 2 ) q R 13 , (CR 2 )XO ⁇ 2 R 13 , CONR 13 (CR 2 ) p R 13 , SO 2 NR 13 (CR 2 ) P R 13 or NR 13 (CR 2 ) q R 13 or R 13 ; R 13 is H, optionally halogenated C 1-6 alkyl, or an optionally substituted C 3 - 7 cycloalkyl, 5-7 membered aryl, heterocyclic or heteroaryl.
  • the present invention provides pharmaceutical compositions comprising a compound having Formula (1) or (2), and a pharmaceutically acceptable excipient.
  • the invention also provides methods for modulating a protein kinase, comprising administering to a system or a subject in need thereof, a therapeutically effective amount of a compound having Formula (1) or (2), or pharmaceutically acceptable salts or pharmaceutical compositions thereof, thereby modulating said protein kinase.
  • protein kinases which may be modulated using the compounds of the invention include but are not limited to AIk, AbI, Aurora-A, B-Raf, Bcr-Abl, BRK, BIk, Bmx, c-Kit, c-Raf, , c-Src, CSK, EphB, FLTl, Fms, Fyn, JAK2, KDR, Lck, Lyn, PDGFR ⁇ , PDGFR ⁇ , PKC ⁇ , p38 (p38 MAP kinase, SAPK2 ⁇ ), SIK, Src, Syk, Tie2 and TrkB. More particularly, the compounds of Formula (1) or (2) may be used for inhibiting a protein kinase, such as B-Raf, Lck or a combination thereof.
  • a protein kinase such as B-Raf, Lck or a combination thereof.
  • the invention provides methods for ameliorating a condition mediated by a protein kinase, such as a B-Raf or Lck-mediated condition, comprising administering to a system or subject in need of such treatment an effective amount of a compound having Formula (1) or (2) or pharmaceutically acceptable salts or pharmaceutical compositions thereof, and optionally in combination with a second therapeutic agent, thereby treating said condition.
  • a protein kinase such as a B-Raf or Lck-mediated condition
  • the compounds of the invention may be used in combination with a chemotherapeutic agent to treat a cell proliferative disorder, including but not limited to, lymphoma, osteosarcoma, melanoma, or a tumor of breast, renal, prostate, colorectal, thyroid, ovarian, pancreatic, neuronal, lung, uterine or gastrointestinal tumor.
  • a chemotherapeutic agent to treat a cell proliferative disorder, including but not limited to, lymphoma, osteosarcoma, melanoma, or a tumor of breast, renal, prostate, colorectal, thyroid, ovarian, pancreatic, neuronal, lung, uterine or gastrointestinal tumor.
  • a compound having Formula (1) or (2) may be administered to a system comprising cells or tissues. In other embodiments, a compound having Formula (1) or (2) may be administered to a human or animal subject.
  • the invention also provides for the use of a compound of Formula (1) or (2), or pharmaceutically acceptable salts or pharmaceutical compositions thereof, for treating a protein kinase-mediated condition.
  • the invention also provides for the use of a compound of Formula (1) or (2) in the manufacture of a medicament for treating a condition mediated by a protein kinase.
  • the protein kinases include but are not limited to AIk, AbI, Aurora- A, B-Raf, Bcr-Abl, BRK, BIk, Bmx, c-Kit, c-Raf, , c-Src, CSK, EphB, FLTl, Fms, Fyn, JAK2, KDR, Lck, Lyn, PDGFR ⁇ , PDGFR ⁇ , PKC ⁇ , p38 (p38 MAP kinase, SAPK2 ⁇ ), SIK, Src, Syk, Tie2 and TrkB. More particularly, the protein kinase is B-Raf, Lck or a combination thereof.
  • the condition includes, but is not limited to, lymphoma, osteosarcoma, melanoma, or a tumor of breast, renal, prostate, colorectal, thyroid, ovarian, pancreatic, neuronal, lung, uterine or gastrointestinal tumor.
  • the invention also provides for the use of a compound of Formula (1) or (2) in the manufacture of a medicament for treating a cell-proliferative condition, wherein the cell- proliferative condition is lymphoma, osteosarcoma, melanoma, or a tumor of breast, renal, prostate, colorectal, thyroid, ovarian, pancreatic, neuronal, lung, uterine or gastrointestinal tumor.
  • a cell-proliferative condition is lymphoma, osteosarcoma, melanoma, or a tumor of breast, renal, prostate, colorectal, thyroid, ovarian, pancreatic, neuronal, lung, uterine or gastrointestinal tumor.
  • Alkyl refers to a moiety and as a structural element of other groups, for example halo-substituted-alkyl and alkoxy, and may be straight-chained or branched.
  • An optionally substituted alkyl, alkenyl or alkynyl as used herein may be optionally halogenated (e.g., CF 3 ), or may have one or more carbons that is substituted or replaced with a heteroatom, such as NR, O or S (e.g., -OCH 2 CH 2 O-, alkylthiol, thioalkoxy, alkylamine, etc).
  • Aryl refers to a monocyclic or fused bicyclic aromatic ring containing carbon atoms.
  • aryl may be phenyl or naphthyl.
  • Arylene means a divalent radical derived from an aryl group.
  • Heteroaryl as used herein is as defined for aryl above, where one or more of the ring members are a heteroatom.
  • heteroaryls include but are not limited to pyridyl, indolyl, indazolyl, quinoxalinyl, quinolinyl, benzofuranyl, benzopyranyl, benzothiopyranyl, benzo[l,3]dioxole, imidazolyl, benzoimidazolyl, pyrimidinyl, furanyl, oxazolyl, isoxazolyl, triazolyl, tetrazolyl, pyrazolyl, thienyl, etc.
  • Examples of carbocyclic rings include but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclopropylene, cyclohexanone, etc.
  • a "heterocyclic ring” as used herein is as defined for a carbocyclic ring above, wherein one or more ring carbons is a heteroatom.
  • heterocyclic rings include but are not limited to morpholino, pyrrolidinyl, pyrrolidin-2-one, piperazinyl, piperidinyl, piperidinone, l,4-dioxa-8-aza-spiro[4.5]dec-8-yl, etc.
  • co- administration or “combined administration” or the like as used 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 refers to a product obtained from mixing or combining active ingredients, 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 (1) 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 (1) 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 active ingredients in the body of the patient.
  • cocktail therapy e.g. the administration of three or more active ingredients.
  • terapéuticaally effective amount means the amount of the subject compound that will elicit a biological or medical response in a cell, tissue, organ, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician.
  • administration means providing a compound of the invention and prodrugs thereof to a subject in need of treatment.
  • Kinase Panel is a list of kinases including but not limited to Abl(human), Abl(T315I), JAK2, JAK3, ALK, JNKl ⁇ l, ALK4, KDR, Aurora- A, Lck, BIk, MAPKl, Bmx, MAPKAP- K2, BRK, MEKl, CaMKII(rat), Met, CDKl/cyclinB, p70S6K, CHK2, PAK2, CKl, PDGFR ⁇ , CK2, PDKl, c-Kit, Pim-2, C-Raf, PKA(h), CSK, PKB ⁇ , Src, PKC ⁇ , DYRK2, Plk3, EGFR, ROCK-I, Fes, Ron, FGFR-3, Ros, Flt3, SAPK2 ⁇ , Fms, SGK, Fyn, SIK, GSK3 ⁇ , Syk, IGFR, Tie-2, IKKB, TrkB, IR
  • the present invention provides compounds and pharmaceutical compositions thereof, which may be useful as protein kinase inhibitors.
  • the invention provides compounds having Formula (1):
  • X is a 5-7 membered aryl or heteroaryl containing N, O or S;
  • L is NHCO or CONH
  • R 1 is H, halo, NR 4 CONR 5 R 6 , NR 4 CO 2 R 5 , NR 4 (CR 2 ) P NR 5 R 6 or NR 4 (CR 2 ) P R 5 ;
  • R 2 is halo, or an optionally halogenated Ci_ 6 alkyl
  • R 3 is halo, an optionally halogenated Ci_ 6 alkyl, (CR 2 ) q R 7 or O(CR 2 ) q R 8 ;
  • R and R 4 are independently H or Ci_ 6 alkyl
  • R 5 and R 6 are independently H, an optionally halogenated C]_ 6 alkyl, C 2 - 6 alkenyl or C 2 _ 6 alkynyl; Ci_ 6 alkanol, (CR 2 ) q -R 7 or O(CR 2 ) q R 8 ; or R 5 and R 6 together with N in NR 5 R 6 may form an optionally substituted ring;
  • R 7 and R 8 are independently an optionally substituted C 3 _ 7 cycloalkyl, 5-7 membered aryl, heterocyclic or heteroaryl; or R 8 is H; m is 1-4; and n, p and q are independently 0-4. [0032] In one embodiment, the invention provides compounds of Formula (2):
  • R 1 is halo, NR 4 CONR 5 R 6 , NR 4 CO 2 R 5 , NR 4 (CR 2 ) P NR 5 R 6 or NR 4 (CR 2 ) P R 5 ;
  • R 9 is halo or CF 3 ;
  • R 10 is halo, an optionally halogenated C 1-6 alkyl, or OCi_6 alkyl;
  • R 11 is halo, (CR 2 ) q R 7 or O(CR 2 ) q R 8 ;
  • R and R 4 are independently H or C 1-6 alkyl
  • R and R are independently H, an optionally halogenated Ci_ 6 alkyl, C 2 _ 6 alkenyl or C 2 _ 6 alkynyl; C 1-6 alkanol, (CR 2 ) q -R 7 or O(CR 2 ) q R 8 ; or R 5 and R 6 together with N in NR 5 R 6 may form an optionally substituted ring;
  • R and R are independently an optionally substituted C 3 _ 7 cycloalkyl, 5-7 membered aryl, heterocyclic or heteroaryl; or R 8 is H; m and n are independently 0- 1 ; and p and q are independently 0-4.
  • the present invention also includes all suitable isotopic variations of the compounds of the invention, or pharmaceutically acceptable salts thereof.
  • An isotopic variation of a compound of the invention or a pharmaceutically acceptable salt thereof is defined as one in which at least one atom is replaced by an atom having the same atomic number but an atomic mass different from the atomic mass usually found in nature.
  • isotopes that may be incorporated into the compounds of the invention and pharmaceutically acceptable salts thereof include but are not limited to isotopes of hydrogen, carbon, nitrogen and oxygen such as as 2 H, 3 H, 11 C, 13 C, 14 C, 15 N, 17 O, 18 0, 35 S, 18 F, 36 Cl and 123 I.
  • isotopic variations of the compounds of the invention and pharmaceutically acceptable salts thereof are useful in drug and/or substrate tissue distribution studies.
  • 3 H and 14 C isotopes may be used for their ease of preparation and detectability.
  • substitution with isotopes such as H may afford certain therapeutic advantages resulting from greater metabolic stability, such as increased in vivo half-life or reduced dosage requirements.
  • Isotopic variations of the compounds of the invention or pharmaceutically acceptable salts thereof can generally be prepared by conventional procedures using appropriate isotopic variations of suitable reagents.
  • Compounds having Formula (1) or (2) may be useful as protein kinase inhibitors.
  • compounds having Formula (1) or (2), and pharmaceutically acceptable salts, solvates, N-oxides, prodrugs and isomers thereof may be used for the treatment of a kinase-mediated condition or disease, such as diseases mediated by AIk, AbI, Aurora-A, B-Raf, Bcr-Abl, BRK, BIk, Bmx, c-Kit, c-Raf, c-Src, CSK, EphB, FLTl, Fms, Fyn, JAK2, KDR, Lck, Lyn, PDGFR ⁇ , PDGFR ⁇ , PKC ⁇ , p38 (p38 MAP kinase, SAPK2 ⁇ ), SIK, Src, Syk, Tie2 and TrkB kinases, or a combination thereof.
  • a kinase-mediated condition or disease such as diseases mediated by AIk, AbI,
  • the compounds of the invention may also be used in combination with a second therapeutic agent, for ameliorating a condition mediated by a protein kinase, such as a B-Raf or Lck-mediated condition.
  • a second therapeutic agent for ameliorating a condition mediated by a protein kinase, such as a B-Raf or Lck-mediated condition.
  • the compounds of the invention may be used in combination with a chemotherapeutic agent to treat a cell proliferative disorder, including but not limited to, lymphoma, osteosarcoma, melanoma, or a tumor of breast, renal, prostate, colorectal, thyroid, ovarian, pancreatic, neuronal, lung, uterine or gastrointestinal tumor.
  • chemotherapeutic agents which may be used in the compositions and methods of the invention include but are not limited to anthracyclines, alkylating agents (e.g., mitomycin C), alkyl sulfonates, aziridines, ethylenimines, methylmelamines, nitrogen mustards, nitrosoureas, antibiotics, antimetabolites, folic acid analogs (e.g., dihydrofolate reductase inhibitors such as methotrexate), purine analogs, pyrimidine analogs, enzymes, podophyllotoxins, platinum-containing agents, interferons, and interleukins.
  • alkylating agents e.g., mitomycin C
  • alkyl sulfonates e.g., aziridines, ethylenimines, methylmelamines, nitrogen mustards, nitrosoureas, antibiotics, antimetabolites, folic acid analogs (e.g., dihydrofolate reduc
  • chemotherapeutic agents which may be used in the compositions and methods of the invention include, but are not limited to, busulfan, improsulfan, piposulfan, benzodepa, carboquone, meturedepa, uredepa, altretamine, triethylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide, trimethylolomelamine, chlorambucil, chlornaphazine, cyclophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard, carmustine, chlorozotocin, fotemustine, lomustine, nimustine, ranimustine, dacarbazine, mannomustine, mitobronitol, mitolactol, pipobroman, aclacinomycin
  • Compounds of the invention are screened against the kinase panel (wild type and/or mutation thereof) and may modulate the activity of at least one panel kinase panel member. As such, compounds of the invention may be useful for treating diseases or disorders in which kinases contribute to the pathology and/or symptomology of the disease.
  • kinases that may be inhibited by the compounds and compositions described herein and against which the methods described herein may be useful include, but are not limited to AIk, AbI, Aurora-A, B- Raf, Bcr-Abl, BRK, BIk, Bmx, c-Kit, c-Raf, c-Src, CSK, EphB, FLTl, Fms, Fyn, JAK2, KDR, Lck, Lyn, PDGFR ⁇ , PDGFR ⁇ , PKC ⁇ , p38 (p38 MAP kinase, SAPK2 ⁇ ), SIK, Src, Syk, Tie2 and TrkB kinases, and mutant forms thereof.
  • the Ras-Raf- MEK-ERK signaling pathway mediates cellular response to growth signals. Ras is mutated to an oncogenic form in approximately 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 (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, such as melanoma.
  • 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 compounds of the present invention may 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 Src family of kinases is implicated in cancer, immune system dysfunction and bone remodeling diseases.
  • Members of the Src family include the following eight kinases in mammals: Src, Fyn, Yes, Fgr, Lyn, Hck, Lck, and BIk.
  • Src, Fyn, Yes, Fgr, Lyn, Hck, Lck, and BIk For general reviews, see Thomas and Brugge, Annu. Rev. Cell Dev. Biol. (1997) 13, 513; Lawrence and Niu, Pharmacol. Ther. (1998) 77, 81; Tatosyan and Mizenina, Biochemistry (Moscow) (2000) 65, 49; Boschelli et al., Drugs of the Future 2000, 25(7), 717.
  • 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. Molina et al., Nature, 357, 161 (1992). Hck, Fgr and Lyn have been identified as important mediators of integrin signaling in myeloid leukocytes. Lowell et al., J. Leukoc. Diol., 65, 313 (1999). Inhibition of these kinase mediators may therefore be useful for treating inflammation. Boschelli et al., Drugs of the Future 2000, 25(7), 717.
  • Lyn a member of the Src family, plays a role in the regulation of B -cell immune responses. Lyn-deficient mice display disrupted B-cell function, leading to autoimmunity and defective mast cell degranulation. Studies have also suggested that Lyn is a negative regulator of apoptosis in various cell systems. In leukemic cells, Lyn is constitutively activated, and the inhibition of Lyn expression reversed proliferation. In addition, Lyn has been shown to be expressed in colon and PC cells, and that overexpression of a dominant active Lyn in colon cancer cell lines induced chemoresistance. (Goldenberg-Furmanov et al., Cancer Res. 64:1058-1066 (2004)).
  • c-Src transmits oncogenic signals of many receptors.
  • 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.
  • c-Src tyrosine kinase (CSK) influences the metastatic potential of cancer cells, particularly colon cancer.
  • c-Kit has a substantial homology to the PDGF receptor and to the CSF- 1 receptor (c- Fms). Investigations on various erythroid and myeloid cell lines indicate an expression of the c- Kit gene in early stages of differentiation (Andre et al., Oncogene 4 (1989), 1047-1049). Certain tumors such as glioblastoma cells likewise exhibit a pronounced expression of the c-Kit gene.
  • Eph receptors which include EphA and EphB subfamily, consist of the largest group of receptor tyrosine kinases. EphB was found to be overexpressed in several tumors including ovarian tumors, liver tumors, kidney tumors as well as melanomas. Downregulation of EphB signaling has shown to inhibit tumor growth and metastasis. Therefore, EphB may be an important target for anti-tumorigenic therapies. (Clevers et al., Cancer Res. 66:2-5 (2006); Heroult et al., Experimental Cell Res. 312: 642-650 (2006); and Batlle et al., Nature 435:1126- 1130 (2005)).
  • KDR Kinase insert domain-containing receptor
  • Fms-like tyrosine kinase referred to as "Fltl” hereinafter
  • Oncogene, 5: 519 (1990); Science, 255: 989 (1992)] belong to the receptor type tyrosine kinase family.
  • VEGF specifically binds to Fit- 1 and KDR at Kd values of 20 pM and 75 pM and that Fltl and KDR are expressed in vascular endothelial cells in a specific manner [Proc. Natl. Acad. Sci. USA, 90: 7533 (1993); Proc. Natl. Acad. Sci. USA, 90: 8915 (1993)].
  • FIt-I in various diseases, it has been reported that, in comparison with vascular endothelial cells in normal tissues, expression of FIt-I mRNA increases in tumor vascular endothelial cells of human glioblastoma tissues [Nature, 359: 845 (1992)] and tumor vascular endothelial cells of human digestive organ cancer tissues [Cancer Research, 53: 4727 (1993)]. Additionally, it has been reported that expression of FIt-I mRNA is observed by in situ hybridization in vascular endothelial cells of joints of patients with rheumatoid arthritis [J. Experimental Medicine, 180: 341 (1994)]. Studies also suggest that FIt-I plays an important role in tumor angiogenesis.
  • Flt3 is a member of the type III receptor tyrosine kinase (RTK) family.
  • Flt3 Flt3 (Fms-like tyrosine kinase) is also known as FLk-2 (fetal liver kinase T).
  • FLk-2 fetal liver kinase T.
  • Aberrant expression of the Flt3 gene has been documented in both adult and childhood leukemias including acute myeloid leukemia (AML), AML with trilineage myelodysplasia (AML/TMDS), acute lymphoblastic leukemia (ALL), and myelodysplastic syndrome (MDS).
  • AML acute myeloid leukemia
  • AML/TMDS AML with trilineage myelodysplasia
  • ALL acute lymphoblastic leukemia
  • MDS myelodysplastic syndrome
  • 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. Inhibition of p-FLT3 kinase activity induces apoptosis (programmed cell death) of the leukemic cells.
  • Abelson tyrosine kinase i.e. AbI, c-Abl
  • AbI AbI
  • c-Abl Abelson tyrosine kinase
  • the AbI protein appears to serve 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 important in the pathogenesis of 95% of chronic myelogenous leukemia (CML) and 10% of acute lymphocytic leukemia.
  • Compounds of the present invention may inhibit AbI kinase, for example, v-Abl kinase.
  • the compounds of the present invention may also inhibit wild-type Bcr-Abl kinase and mutations of Bcr-Abl kinase, and thus may be suitable for the treatment of Bcr-Abl-positive cancer and tumor diseases, such as leukemias (e.g., chronic myeloid leukemia and acute lymphoblastic leukemia, where especially apoptotic mechanisms of action are found) and other proliferation disorders related to Bcr-Abl.
  • leukemias e.g., chronic myeloid leukemia and acute lymphoblastic leukemia, where especially apoptotic mechanisms of action are found
  • other proliferation disorders related to Bcr-Abl such as leukemias (e.g., chronic myeloid leukemia and acute lymphoblastic leukemia, where especially apoptotic mechanisms of action are found) and other proliferation disorders related to Bcr
  • Compounds of the present invention may also be effective against leukemic stem cells, and may be potentially useful 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).
  • Anaplastic lymphoma kinase (ALK), a member of the insulin receptor superfamily of receptor tyrosine kinases, has been implicated in oncogenesis in hematopoietic and non- hematopoietic tumors.
  • ALK insulin receptor superfamily of receptor tyrosine kinases
  • the aberrant expression of full-length ALK receptor proteins has been reported in neuroblastomas and glioblastomas; and ALK fusion proteins have occurred in anaplastic large cell lymphoma.
  • the study of ALK fusion proteins has also raised the possibility of new therapeutic treatments for patients with ALK-positive malignancies. (Pulford et al., Cell. MoI. Life Sci. 61:2939-2953 (2004)).
  • Aurora-A a serine/threonine mitotic kinase
  • Aurora-A has been reported to be overexpressed in various human cancers, and its overexpression induces aneuploidy, centrosome amplification and tumorigenic transformation in cultured human and rodent cells.
  • Bmx/Etk non-receptor tyrosine protein kinase has been implicated in endothelial cell migration and tube formation in vitro.
  • Bmx in endothelium and bone marrow has also been reported to play an important role in arteriogenesis and angiogenesis in vivo, suggesting that Bmx may be a novel target for the treatment of vascular diseases such as coronary artery disease and peripheral arterial disease. (He et al., J. Clin. Invest. 116:2344-2355 (2006)).
  • Breast tumor kinase (Brk) is a soluble protein-tyrosine kinase overexpressed in the majority of breast cancers and also in normal skin and gut epithelium, but not in normal breast epithelial cells. (Zhang et al., J Biol. Chem. 280:1982-1991 (2005)).
  • the Janus kinases are a family of tyrosine kinases consisting of JAKl, JAK2, JAK3 and TYK2.
  • the JAKs play an important role in cytokine signaling.
  • the down-stream substrates of the JAK family of kinases include the signal transducer and activator of transcription (STAT) proteins.
  • STAT signal transducer and activator of transcription
  • JAK/STAT signaling has been implicated in the mediation of many abnormal immune responses such as allergies, asthma, autoimmune diseases such as transplant rejection, rheumatoid arthritis, amyotrophic lateral sclerosis and multiple sclerosis as well as in solid and hematologic malignancies such as leukemias and lymphomas.
  • VEGF vascular endothelium growth factor
  • VEC vascular endothelial cell
  • TC tumor cell
  • KDR KDR is the main receptor which gives play to VEGF functions, while other receptors play little role in cell growth. KDR is highly expressed on the TC and tumor VEC while lowly expressed on the normal tissues.
  • MAPKs Mitogen- activated protein kinases
  • MKKs mitogen- activated protein kinase kinases
  • cytokines including but not limited to TNF ⁇ , IL-6, IL-8 and IL-l ⁇ .
  • Peripheral blood monocytes PBMCs
  • LPS lipopoly saccharide
  • 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.
  • PDGF Platinum-derived Growth Factor
  • PDGFR PDGF receptor
  • Compounds of the present invention may 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.
  • Compounds of the present invention may also be useful 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 may especially be used for the treatment of diseases, which respond to an inhibition of the PDGF receptor kinase.
  • Compounds of the present invention may exhibit useful effects in the treatment of disorders arising as a result of transplantation, for example, allogenic transplantation, especially tissue rejection, such as obliterative bronchiolitis (OB), i.e. a chronic rejection of allogenic lung transplants.
  • OB obliterative bronchiolitis
  • OB obliterative bronchiolitis
  • PDGF PDGF
  • Compounds of the present invention may also be effective against diseases associated with vascular smooth-muscle cell migration and proliferation (where PDGF and PDGFR often also play a role), such as restenosis and atherosclerosis.
  • diseases associated with vascular smooth-muscle cell migration and proliferation where PDGF and PDGFR often also play a role
  • diseases associated with vascular smooth-muscle cell migration and proliferation such as restenosis and atherosclerosis.
  • PKC Protein kinase C
  • 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.
  • SNFlLK locus also known as SIK
  • Snf Ilk is also expressed in skeletal muscle progenitor cells of the somite beginning at 9.5 dpc, suggesting a more general role for snf Ilk in the earliest stages of muscle growth and/or differentiation.
  • Syk is a tyrosine kinase that plays an important role in mast cell degranulation and eosinophil activation. Accordingly, Syk kinase is implicated in various allergic disorders, in particular asthma. It has been shown that Syk binds to the phosphorylated gamma chain of the Fc ⁇ Rl receptor via N-terminal SH 2 domains and is important for downstream signaling.
  • Tie2 inhibitors can be used in situations where neovascularization takes place inappropriately (i.e.
  • TrkA, TrkB, TrkC 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. Moreover, TrkB is 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-gamma transduction pathway (Sugimoto et al., Jpn J. Cancer Res. 2001 Feb; 92(2): 152-60).
  • RTKs The class III receptor tyrosine kinases (RTKs), which include c-FMS, c-KIT, FLT3, platelet-derived growth factor receptor ⁇ (PDGFR ⁇ ) and / ⁇ PDGFR/)), have been reported to be associated with the pathogenesis of an increasing number of malignancies. (Blume-Jensen et al., Nature 411:355-565 (2001); Scheijin et al., Oncogene 21:3314-3333 (2002)).
  • 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 (1) 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 may 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.
  • 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 may 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, together with c) binders, e.g., magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose and or polyvinylpyrrolidone; and if desired, d) disintegrants, e.g., starches,
  • 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.
  • adjuvants such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure and/or buffers.
  • 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, may be 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 may 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.
  • a compound of the invention may 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 may 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 may be prepared using salts of the starting materials or intermediates.
  • the free acid or free base forms of the compounds of the invention may 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 may 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 may 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 may 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 may 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 may 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 may 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 may be conveniently prepared or formed during the process of the invention, as solvates (e.g., hydrates). Hydrates of compounds of the present invention may be conveniently prepared by recrystallization from an aqueous/organic solvent mixture, using organic solvents such as dioxin, tetrahydrofuran or methanol.
  • Compounds of the invention may 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.
  • Resolution of enantiomers may be carried out using covalent diastereomeric derivatives of the compounds of the invention, or by using dissociable complexes (e.g., crystalline diastereomeric salts).
  • Diastereomers have distinct physical properties (e.g., melting points, boiling points, solubility, reactivity, etc.) and may be readily separated by taking advantage of these dissimilarities.
  • the diastereomers may be separated by chromatography, or 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 solution is added via cannula to a solution of N-(3-amino-4- methyl-phenyl)-3-trifluoromethyl-benzamide (1.12 g, 3.81 mmol) and diisopropanylethylamine (1.65 mL, 9.52 mmol) in 40 mL CH 2 Cl 2 at 0 0 C over 30 min.
  • the mixture is stirred at rt for 1 h.
  • the mixture is extracted with ethyl acetate.
  • the organic phase is washed with saturated ⁇ aHCU 3 solution and brine.
  • Compounds of the present invention may be assayed to measure their capacity to inhibit a kinase panel, including but not limited to AIk, AbI, Aurora-A, B-Raf, Bcr-Abl, BRK, BIk, Bmx, c-Kit, c-Raf, cSRC, CSK, EphB, FLTl, Fms, Fyn, JAK2, KDR, Lck, Lyn, PDGFR ⁇ , PDGFR ⁇ , PKC ⁇ , p38 (p38 MAP kinase, SAPK2 ⁇ ), SIK, Src, Syk, Tie2 and TrkB kinases.
  • Compounds of the invention may be 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, I ⁇ B ⁇ is diluted in DPBS (1:750) and 15 ⁇ l is added to each well.
  • the plates are incubated at 4 0 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 0 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 fluorescent Attophos AP substrate (Promega) 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 Program (Excitation 455 nm, Emission 580 nm).
  • 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.
  • the murine cell line 32D hemopoietic progenitor cell line may be 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.
  • test compounds of the present invention may demonstrate an apoptotic effect on the 32D-p210 cells but not induce apoptosis in the 32D parental cells.
  • 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 2x10 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 may inhibit the proliferation of the Bcr-Abl expressing cells, inhibiting the cellular Bcr-Abl autophosphorylation in a dose-dependent manner.
  • Compounds of the invention may be 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 may be tested at 10, 3.3, 1.1 and 0.37 ⁇ M as described above (in media lacking IL3).
  • the IC 50 values of the compounds lacking toxicity on the untransformed cells are determined from the dose response curves obtained as described above.
  • Kinase activity assay with purified FGFR-3 (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 pH7.5, 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 FGFR-3 enzyme in kinase buffer was first dispensed into 384-well 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 pH 7.5, 0.5 M KF, 50 mM
  • Compounds of the invention are tested for their ability to inhibit transformed Ba/F3- TEL-FGFR3 cells proliferation, which is depended on FGFR-3 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 dimethylsulf oxide (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 dimethylsulf oxide
  • 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 0 C cell culture incubator, fluorescence signals from reduced AlamarBlue® (Excitation at 530 nm, Emission at 580 nm) are quantified 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.
  • Compounds of the invention may be tested for their ability to inhibit transformed Ba/F3-FLT3-ITD or Ba/F3-Tel-PDGFR ⁇ cells proliferation, which is dependent on FLT3 or PDGFR ⁇ cellular kinase activity.
  • Ba/F3-FLT3-ITD or Ba/F3-Tel-PDGFR ⁇ 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 dimethylsulfoxide (DMSO).
  • DMSO dimethylsulfoxide
  • Compounds of the invention may be assessed for their ability to inhibit individual members of a panel of kinases (a partial, non-limiting list of kinases includes: AIk, AbI, Aurora- A, B-Raf, Bcr-Abl, BRK, BIk, Bmx, c-Kit, c-Raf, cSRC, CSK, EphB, FLTl, Fms, Fyn, JAK2, KDR, Lck, Lyn, PDGFR ⁇ , PDGFR ⁇ , PKC ⁇ , p38 (p38 MAP kinase, SAPK2 ⁇ ), SIK, Src, Syk, Tie2 and TrkB kinases).
  • a partial, non-limiting list of kinases includes: AIk, AbI, Aurora- A, B-Raf, Bcr-Abl, BRK, BIk, Bmx, c-Kit, c-Raf, cSRC, CSK, Eph
  • kinase buffer composition and the substrates vary for the different kinases included in the "Upstate KinaseProfilerTM" panel.
  • Kinase buffer (2.5 ⁇ L, 10x - 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 .01 mg/ml) in kinase buffer and kinase buffer (50 ⁇ M; 5 ⁇ L) are mixed in an eppendorf on ice.
  • 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 (1) or (2) in free form or in pharmaceutically acceptable salt form may exhibit valuable pharmacological properties, for example, as indicated by the in vitro tests described in this application.
  • the IC 50 value in those experiments is given as that concentration of the test compound in question that results in a cell count that is 50 % lower than that obtained using the control without inhibitor.
  • compounds of the invention have IC 50 values from 1 nM to 10 ⁇ M.
  • compounds of the invention have IC 50 values from 0.01 ⁇ M to 5 ⁇ M.
  • compounds of the invention have IC 50 values from 0.01 ⁇ M to 1 ⁇ M, or more particularly from 1 nM to 1 ⁇ M.
  • compounds of the invention have IC 50 values of less than 1 nM or more than 10 ⁇ M.
  • Compounds of Formula (1) or (2) may exhibit a percentage inhibition of greater than 50%, or in other embodiments, may exhibit a percentage inhibition greater than about 70%, against one or more of the following kinases at 10 ⁇ M: AIk, AbI, Aurora-A, B-Raf, Bcr-Abl, BRK, BIk, Bmx, c-Kit, c-Raf, cSRC, CSK, EphB, FLTl, Fms, Fyn, JAK2, KDR, Lck, Lyn, PDGFR ⁇ , PDGFR ⁇ , PKC ⁇ , p38 (p38 MAP kinase, SAPK2 ⁇ ), SIK, Src, Syk, Tie2 and TrkB kinases.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention provides compounds of formula (1 ) and pharmaceutical compositions thereof, which are useful as protein kinase inhibitors, and methods for using such compounds to treat, ameliorate or prevent a condition associated with abnormal or deregulated kinase activity. In some embodiments, the invention provides methods for using such compounds to treat, ameliorate or prevent diseases or disorders that involve abnormal activation of AIk, AbI, Aurora-A, B-Raf, Bcr-Abl, BRK, BIk, Bmx, c-Kit, c-RAF, cSRC. CSK, FLTl, Fms, Fyn, JAK2, KDR, Lck, Lyn, PDGFRα, PDGFRβ, PKCα. p38 (p38 MAP kinase, SAPK2α), Src, SIK, Syk, Tie2 and TrkB kinases. FORMULE (I)

Description

PROTEIN KINASE INHIBITORS AND METHODS FOR USING THEREOF
Cross-Reference to Related Applications
[0001] This application claims the benefit of U.S. provisional application serial number 60/910,185, filed April 4, 2007, which is incorporated herein by reference in its entirety.
Technical Field
[0002] The invention relates to protein kinase inhibitors, and methods of using such compounds.
Background Art
[0003] 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 (PDGFR), the nerve growth factor receptor, TrkB, Met, and the fibroblast growth factor receptor, FGFR-3 ; non-receptor tyrosine kinases such AbI and the fusion kinase Bcr-Abl, Lck, Csk, Fes, Bmx and Src; and serine/threonine kinases such as B-Raf, C-Raf, Sgk, MAP kinases (e.g., MKK4, MKK6, etc.) and SAPK2α, SAPK2β and SAPK3. 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.
Disclosure of the Invention
[0004] The invention provides compounds and pharmaceutical compositions thereof, which may be useful as protein kinase inhibitors.
[0005] In one aspect, the invention provides compounds having Formula (1):
Figure imgf000003_0001
or pharmaceutically acceptable salts thereof, wherein: X is a 5-7 membered aryl or heteroaryl containing N, O or S; L is NHCO or CONH;
R1 is H, halo, NR4CONR5R6, NR4CO2R5, NR4(CR2)PNR5R6 or NR4(CR2)PR5; R2 is halo, or an optionally halogenated Ci_6 alkyl; R3 is halo, an optionally halogenated Ci_6 alkyl, (CR2)qR7 or O(CR2)qR8; R and R4 are independently H or C1-6 alkyl;
R and R are independently H, an optionally halogenated Ci_6 alkyl, C2_6 alkenyl or C2_6 alkynyl; Ci_6 alkanol, (CR2)q-R7 or O(CR2)qR8; or R5 and R6 together with N in NR5R6 may form an optionally substituted ring;
R7 and R8 are independently an optionally substituted C3_7 cycloalkyl, 5-7 membered aryl, heterocyclic or heteroaryl; or R8 is H; m is 1-4; and n, p and q are independently 0-4.
[0006] In the above Formula (1), X may be phenyl, thienyl or pyridyl. In some examples, R is Ci-6 alkyl. In other examples, R3 is halo, an optionally halogenated C1-6 alkyl, (CR2)qR7, O(CR2)qR8; and R7 and R8 are independently an optionally substituted piperidinyl, piperazinyl or imidazolyl.
[0007] In one embodiment, the invention provides compounds of Formula (1), wherein: R1 is halo, NR4CONR5R6, NR4CO2R5, NR4(CR2)PNR5R6 or NR4(CR2)PR5; R5 and R6 are independently H, Ci_6 alkyl, Ci_6 alkanol, or (CR2)qR7; R7 is an optionally substituted C5-7 cycloalkyl pyridyl, phenyl, naphthalenyl, morpholinyl, piperazinyl, pyrimidinyl, imidazolyl, triazolyl, isothiazolyl, isoxazolyl, pyrazolyl or pyrazinyl; and q is 0-1. [0008] In another embodiment, the invention provides compounds of Formula (2):
Figure imgf000004_0001
wherein R1 is halo, NR4CONR5R6, NR4CO2R5, NR4(CR2)PNR5R6 or NR4(CR2)PR5; R9 is halo or CF3;
R10 is halo, an optionally halogenated C1-6 alkyl, or OCi_6 alkyl; R11 is halo, (CR2)qR7 or O(CR2)qR8; R and R4 are independently H or Ci_6 alkyl;
R5 and R6 are independently H, an optionally halogenated C1-6 alkyl, C2_6 alkenyl or C2_6 alkynyl; Ci_6 alkanol, (CR2)q-R7 or O(CR2)qR8; or R5 and R6 together with N in NR5R6 may form an optionally substituted ring;
R7 and R8 are independently an optionally substituted C3_7 cycloalkyl, 5-7 membered aryl, heterocyclic or heteroaryl; or R is H; m and n are independently 0-1; and p and q are independently 0-4.
[0009] In some examples, R1 in Formula (1) and (2) is NR4CONR5R6, NR4CO2R5 or NR4(CR2)PNR5R6. For example, R5 and R6 are independently H, Ci_6 alkyl, or (CR2)qR7; and R7 is an optionally substituted Cs_7 cycloalkyl, pyridyl, phenyl, naphthalenyl, morpholinyl, piperazinyl, pyrimidinyl, imidazolyl, triazolyl, isothiazolyl, isoxazolyl, pyrazolyl or pyrazinyl. In some examples, R10 is CF3. In other examples, R11 is halo, (CR2)qR7, O(CR2)qR8 and R7 and R8 are independently an optionally substituted piperidinyl, piperazinyl or imidazolyl. In yet other examples, m and n in compounds of Formula (2) are each 0.
[0010] In the compounds of the invention, suitable substituents will be known to those of ordinary skill in the art, including but not limited to halo, optionally halogenated C1-6 alkyl, C2_6 alkenyl, C2-6 alkynyl, cyano, nitro or (CR2)PR12; wherein R12 is O(CR2)qR13, S(CR2)qR13, (CR2)XO^2R13, CONR13(CR2)pR13, SO2NR13(CR2)PR13 or NR13(CR2)qR13 or R13; R13 is H, optionally halogenated C1-6 alkyl, or an optionally substituted C3-7 cycloalkyl, 5-7 membered aryl, heterocyclic or heteroaryl.
[0011] In another aspect, the present invention provides pharmaceutical compositions comprising a compound having Formula (1) or (2), and a pharmaceutically acceptable excipient.
[0012] The invention also provides methods for modulating a protein kinase, comprising administering to a system or a subject in need thereof, a therapeutically effective amount of a compound having Formula (1) or (2), or pharmaceutically acceptable salts or pharmaceutical compositions thereof, thereby modulating said protein kinase. Examples of protein kinases which may be modulated using the compounds of the invention include but are not limited to AIk, AbI, Aurora-A, B-Raf, Bcr-Abl, BRK, BIk, Bmx, c-Kit, c-Raf, , c-Src, CSK, EphB, FLTl, Fms, Fyn, JAK2, KDR, Lck, Lyn, PDGFRα, PDGFRβ, PKCα, p38 (p38 MAP kinase, SAPK2α), SIK, Src, Syk, Tie2 and TrkB. More particularly, the compounds of Formula (1) or (2) may be used for inhibiting a protein kinase, such as B-Raf, Lck or a combination thereof.
[0013] In yet another aspect, the invention provides methods for ameliorating a condition mediated by a protein kinase, such as a B-Raf or Lck-mediated condition, comprising administering to a system or subject in need of such treatment an effective amount of a compound having Formula (1) or (2) or pharmaceutically acceptable salts or pharmaceutical compositions thereof, and optionally in combination with a second therapeutic agent, thereby treating said condition. For example, the compounds of the invention may be used in combination with a chemotherapeutic agent to treat a cell proliferative disorder, including but not limited to, lymphoma, osteosarcoma, melanoma, or a tumor of breast, renal, prostate, colorectal, thyroid, ovarian, pancreatic, neuronal, lung, uterine or gastrointestinal tumor.
[0014] In the above methods for using the compounds of the invention, a compound having Formula (1) or (2) may be administered to a system comprising cells or tissues. In other embodiments, a compound having Formula (1) or (2) may be administered to a human or animal subject.
[0015] The invention also provides for the use of a compound of Formula (1) or (2), or pharmaceutically acceptable salts or pharmaceutical compositions thereof, for treating a protein kinase-mediated condition.
[0016] The invention also provides for the use of a compound of Formula (1) or (2) in the manufacture of a medicament for treating a condition mediated by a protein kinase. [0017] In the above used the protein kinases include but are not limited to AIk, AbI, Aurora- A, B-Raf, Bcr-Abl, BRK, BIk, Bmx, c-Kit, c-Raf, , c-Src, CSK, EphB, FLTl, Fms, Fyn, JAK2, KDR, Lck, Lyn, PDGFRα, PDGFRβ, PKCα, p38 (p38 MAP kinase, SAPK2α), SIK, Src, Syk, Tie2 and TrkB. More particularly, the protein kinase is B-Raf, Lck or a combination thereof.
[0018] In the above used the condition includes, but is not limited to, lymphoma, osteosarcoma, melanoma, or a tumor of breast, renal, prostate, colorectal, thyroid, ovarian, pancreatic, neuronal, lung, uterine or gastrointestinal tumor.
[0019] The invention also provides for the use of a compound of Formula (1) or (2) in the manufacture of a medicament for treating a cell-proliferative condition, wherein the cell- proliferative condition is lymphoma, osteosarcoma, melanoma, or a tumor of breast, renal, prostate, colorectal, thyroid, ovarian, pancreatic, neuronal, lung, uterine or gastrointestinal tumor.
Definitions
[0020] "Alkyl" refers to a moiety and as a structural element of other groups, for example halo-substituted-alkyl and alkoxy, and may be straight-chained or branched. An optionally substituted alkyl, alkenyl or alkynyl as used herein may be optionally halogenated (e.g., CF3), or may have one or more carbons that is substituted or replaced with a heteroatom, such as NR, O or S (e.g., -OCH2CH2O-, alkylthiol, thioalkoxy, alkylamine, etc).
[0021] "Aryl" refers to a monocyclic or fused bicyclic aromatic ring containing carbon atoms. For example, aryl may be phenyl or naphthyl. "Arylene" means a divalent radical derived from an aryl group.
[0022] "Heteroaryl" as used herein is as defined for aryl above, where one or more of the ring members are a heteroatom. Examples of heteroaryls include but are not limited to pyridyl, indolyl, indazolyl, quinoxalinyl, quinolinyl, benzofuranyl, benzopyranyl, benzothiopyranyl, benzo[l,3]dioxole, imidazolyl, benzoimidazolyl, pyrimidinyl, furanyl, oxazolyl, isoxazolyl, triazolyl, tetrazolyl, pyrazolyl, thienyl, etc.
[0023] A "carbocyclic ring" as used herein refers to a saturated or partially unsaturated, monocyclic, fused bicyclic or bridged polycyclic ring containing carbon atoms, which may optionally be substituted, for example, with =0. Examples of carbocyclic rings include but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclopropylene, cyclohexanone, etc. [0024] A "heterocyclic ring" as used herein is as defined for a carbocyclic ring above, wherein one or more ring carbons is a heteroatom. For example, a heterocyclic ring may contain N, O, S, -N=, -S-, -S(O), -S(O)2-, or -NR- wherein R may be hydrogen, Ci^alkyl or a protecting group. Examples of heterocyclic rings include but are not limited to morpholino, pyrrolidinyl, pyrrolidin-2-one, piperazinyl, piperidinyl, piperidinone, l,4-dioxa-8-aza-spiro[4.5]dec-8-yl, etc.
[0025] The terms "co- administration" or "combined administration" or the like as used 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.
[0026] The term "pharmaceutical combination" as used herein refers to a product obtained from mixing or combining active ingredients, 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 (1) 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 (1) 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 active ingredients in the body of the patient. The latter also applies to cocktail therapy, e.g. the administration of three or more active ingredients.
[0027] The term "therapeutically effective amount" means the amount of the subject compound that will elicit a biological or medical response in a cell, tissue, organ, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician.
[0028] The term "administration" or "administering" of the subject compound means providing a compound of the invention and prodrugs thereof to a subject in need of treatment.
[0029] "Kinase Panel" is a list of kinases including but not limited to Abl(human), Abl(T315I), JAK2, JAK3, ALK, JNKl αl, ALK4, KDR, Aurora- A, Lck, BIk, MAPKl, Bmx, MAPKAP- K2, BRK, MEKl, CaMKII(rat), Met, CDKl/cyclinB, p70S6K, CHK2, PAK2, CKl, PDGFRα, CK2, PDKl, c-Kit, Pim-2, C-Raf, PKA(h), CSK, PKBα, Src, PKCα, DYRK2, Plk3, EGFR, ROCK-I, Fes, Ron, FGFR-3, Ros, Flt3, SAPK2α, Fms, SGK, Fyn, SIK, GSK3β, Syk, IGFR, Tie-2, IKKB, TrkB, IR, WNK3, IRAK4, ZAP-70, ITK, AMPK(rat), LIMKl, Rsk2, AxI, LKBl, SAPK2β, BrSK2, Lyn (h), SAPK3, BTK, MAPKAP-K3, SAPK4, CaMKIV, MARKl, Snk, CDK2/cyclinA, MINK, SRPKl, CDK3/cyclinE, MKK4(m), TAKl, CDK5/p25, MKK6(h), TBKl, CDKό/cyclinDS, MLCK, TrkA, CD^/cyclinH/MATl, MRCKβ, TSSKl, CHKl, MSKl, Yes, CKId, MST2, ZIPK, c-Kit (D816V), MuSK, DAPK2, NEK2, DDR2, NEK6, DMPK, PAK4, DRAKl, PAR-lBα, EphAl, PDGFRβ, EphA2, Pim-1, EphA5, PKBβ, EphB2, PKCβl, EphB4, PKCδ, FGFRl, PKCη, FGFR2, PKCΘ, FGFR4, PKD2, Fgr, PKGlβ, Fltl, PRK2, Hck, PYK2, HIPK2, Ret, IKKα, RIPK2, IRR, ROCK-II(human), JNK2α2, Rse, JNK3, Rskl(h), PI3 Kγ, PD Kδ and PI3-Kβ.
Modes of Carrying Out the Invention
[0030] The present invention provides compounds and pharmaceutical compositions thereof, which may be useful as protein kinase inhibitors.
[0031] In one aspect, the invention provides compounds having Formula (1):
Figure imgf000008_0001
or pharmaceutically acceptable salts thereof, wherein:
X is a 5-7 membered aryl or heteroaryl containing N, O or S;
L is NHCO or CONH;
R1 is H, halo, NR4CONR5R6, NR4CO2R5, NR4(CR2)PNR5R6 or NR4(CR2)PR5;
R2 is halo, or an optionally halogenated Ci_6 alkyl;
R3 is halo, an optionally halogenated Ci_6 alkyl, (CR2)qR7 or O(CR2)qR8;
R and R4 are independently H or Ci_6 alkyl;
R5 and R6 are independently H, an optionally halogenated C]_6 alkyl, C2-6 alkenyl or C2_6 alkynyl; Ci_6 alkanol, (CR2)q-R7 or O(CR2)qR8; or R5 and R6 together with N in NR5R6 may form an optionally substituted ring;
R7 and R8 are independently an optionally substituted C3_7 cycloalkyl, 5-7 membered aryl, heterocyclic or heteroaryl; or R8 is H; m is 1-4; and n, p and q are independently 0-4. [0032] In one embodiment, the invention provides compounds of Formula (2):
Figure imgf000009_0001
wherein R1 is halo, NR4CONR5R6, NR4CO2R5, NR4(CR2)PNR5R6 or NR4(CR2)PR5;
R9 is halo or CF3;
R10 is halo, an optionally halogenated C1-6 alkyl, or OCi_6 alkyl;
R11 is halo, (CR2)qR7 or O(CR2)qR8;
R and R4 are independently H or C1-6 alkyl;
R and R are independently H, an optionally halogenated Ci_6 alkyl, C2_6 alkenyl or C2_6 alkynyl; C1-6 alkanol, (CR2)q-R7 or O(CR2)qR8; or R5 and R6 together with N in NR5R6 may form an optionally substituted ring;
R and R are independently an optionally substituted C3_7 cycloalkyl, 5-7 membered aryl, heterocyclic or heteroaryl; or R8 is H; m and n are independently 0- 1 ; and p and q are independently 0-4.
[0033] The present invention also includes all suitable isotopic variations of the compounds of the invention, or pharmaceutically acceptable salts thereof. An isotopic variation of a compound of the invention or a pharmaceutically acceptable salt thereof is defined as one in which at least one atom is replaced by an atom having the same atomic number but an atomic mass different from the atomic mass usually found in nature. Examples of isotopes that may be incorporated into the compounds of the invention and pharmaceutically acceptable salts thereof include but are not limited to isotopes of hydrogen, carbon, nitrogen and oxygen such as as 2H, 3H, 11C, 13C, 14C, 15N, 17O, 180, 35S, 18F, 36Cl and 123I. Certain isotopic variations of the compounds of the invention and pharmaceutically acceptable salts thereof, for example, those in which a radioactive isotope such as 3H or 14C is incorporated, are useful in drug and/or substrate tissue distribution studies. In particular examples, 3H and 14C isotopes may be used for their ease of preparation and detectability. In other examples, substitution with isotopes such as H may afford certain therapeutic advantages resulting from greater metabolic stability, such as increased in vivo half-life or reduced dosage requirements. Isotopic variations of the compounds of the invention or pharmaceutically acceptable salts thereof can generally be prepared by conventional procedures using appropriate isotopic variations of suitable reagents.
[0034] Compounds having Formula (1) or (2) may be useful as protein kinase inhibitors. For example, compounds having Formula (1) or (2), and pharmaceutically acceptable salts, solvates, N-oxides, prodrugs and isomers thereof, may be used for the treatment of a kinase-mediated condition or disease, such as diseases mediated by AIk, AbI, Aurora-A, B-Raf, Bcr-Abl, BRK, BIk, Bmx, c-Kit, c-Raf, c-Src, CSK, EphB, FLTl, Fms, Fyn, JAK2, KDR, Lck, Lyn, PDGFRα, PDGFRβ, PKCα, p38 (p38 MAP kinase, SAPK2α), SIK, Src, Syk, Tie2 and TrkB kinases, or a combination thereof.
[0035] The compounds of the invention may also be used in combination with a second therapeutic agent, for ameliorating a condition mediated by a protein kinase, such as a B-Raf or Lck-mediated condition. For example, the compounds of the invention may be used in combination with a chemotherapeutic agent to treat a cell proliferative disorder, including but not limited to, lymphoma, osteosarcoma, melanoma, or a tumor of breast, renal, prostate, colorectal, thyroid, ovarian, pancreatic, neuronal, lung, uterine or gastrointestinal tumor.
[0036] Examples of chemotherapeutic agents which may be used in the compositions and methods of the invention include but are not limited to anthracyclines, alkylating agents (e.g., mitomycin C), alkyl sulfonates, aziridines, ethylenimines, methylmelamines, nitrogen mustards, nitrosoureas, antibiotics, antimetabolites, folic acid analogs (e.g., dihydrofolate reductase inhibitors such as methotrexate), purine analogs, pyrimidine analogs, enzymes, podophyllotoxins, platinum-containing agents, interferons, and interleukins. Particular examples of known chemotherapeutic agents which may be used in the compositions and methods of the invention include, but are not limited to, busulfan, improsulfan, piposulfan, benzodepa, carboquone, meturedepa, uredepa, altretamine, triethylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide, trimethylolomelamine, chlorambucil, chlornaphazine, cyclophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard, carmustine, chlorozotocin, fotemustine, lomustine, nimustine, ranimustine, dacarbazine, mannomustine, mitobronitol, mitolactol, pipobroman, aclacinomycins, actinomycin F(I), anthramycin, azaserine, bleomycin, cactinomycin, carubicin, carzinophilin, chromomycin, dactinomycin, daunorubicin, daunomycin, 6-diazo-5-oxo-l-norleucine, doxorubicin, epirubicin, mitomycin C, mycophenolic acid, nogalamycin, olivomycin, peplomycin, plicamycin, porfiromycin, puromycin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin, denopterin, methotrexate, pteropterin, trimetrexate, fludarabine, 6-mercaptopurine, thiamiprine, thioguanine, ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine, fluorouracil, tegafur, L-asparaginase, pulmozyme, aceglatone, aldophosphamide glycoside, aminolevulinic acid, amsacrine, bestrabucil, bisantrene, carboplatin, cisplatin, defofamide, demecolcine, diaziquone, elfornithine, elliptinium acetate, etoglucid, etoposide, flutamide, gallium nitrate, hydroxyurea, interferon-alpha, interferon-beta, interferon-gamma, interleukin-2, lentinan, lonidamine, mitoguazone, mitoxantrone, mopidamol, nitracrine, pentostatin, phenamet, pirarubicin, podophyllinic acid, 2-ethylhydrazide, procarbazine, razoxane, sizofiran, spirogermanium, paclitaxel, tamoxifen, teniposide, tenuazonic acid, triaziquone, 2,2',2"-trichlorotriethylamine, urethane, vinblastine, vincristine, and vindesine.
Pharmacology and Utility
[0037] Compounds of the invention are screened against the kinase panel (wild type and/or mutation thereof) and may modulate the activity of at least one panel kinase panel member. As such, compounds of the invention may be useful for treating diseases or disorders in which kinases contribute to the pathology and/or symptomology of the disease. Examples of kinases that may be inhibited by the compounds and compositions described herein and against which the methods described herein may be useful include, but are not limited to AIk, AbI, Aurora-A, B- Raf, Bcr-Abl, BRK, BIk, Bmx, c-Kit, c-Raf, c-Src, CSK, EphB, FLTl, Fms, Fyn, JAK2, KDR, Lck, Lyn, PDGFRα, PDGFRβ, PKCα, p38 (p38 MAP kinase, SAPK2α), SIK, Src, Syk, Tie2 and TrkB kinases, and mutant forms thereof.
[0038] The Ras-Raf- MEK-ERK signaling pathway mediates cellular response to growth signals. Ras is mutated to an oncogenic form in approximately 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 (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, such as melanoma.
[0039] 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. Further examples of 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.
[0040] The compounds of the present invention may 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)].
[0041] The Src family of kinases is implicated in cancer, immune system dysfunction and bone remodeling diseases. Members of the Src family include the following eight kinases in mammals: Src, Fyn, Yes, Fgr, Lyn, Hck, Lck, and BIk. For general reviews, see Thomas and Brugge, Annu. Rev. Cell Dev. Biol. (1997) 13, 513; Lawrence and Niu, Pharmacol. Ther. (1998) 77, 81; Tatosyan and Mizenina, Biochemistry (Moscow) (2000) 65, 49; Boschelli et al., Drugs of the Future 2000, 25(7), 717.
[0042] 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. Molina et al., Nature, 357, 161 (1992). Hck, Fgr and Lyn have been identified as important mediators of integrin signaling in myeloid leukocytes. Lowell et al., J. Leukoc. Diol., 65, 313 (1999). Inhibition of these kinase mediators may therefore be useful for treating inflammation. Boschelli et al., Drugs of the Future 2000, 25(7), 717.
[0043] Lyn, a member of the Src family, plays a role in the regulation of B -cell immune responses. Lyn-deficient mice display disrupted B-cell function, leading to autoimmunity and defective mast cell degranulation. Studies have also suggested that Lyn is a negative regulator of apoptosis in various cell systems. In leukemic cells, Lyn is constitutively activated, and the inhibition of Lyn expression reversed proliferation. In addition, Lyn has been shown to be expressed in colon and PC cells, and that overexpression of a dominant active Lyn in colon cancer cell lines induced chemoresistance. (Goldenberg-Furmanov et al., Cancer Res. 64:1058-1066 (2004)).
[0044] 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. On the other hand, 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. c-Src tyrosine kinase (CSK) influences the metastatic potential of cancer cells, particularly colon cancer.
[0045] c-Kit has a substantial homology to the PDGF receptor and to the CSF- 1 receptor (c- Fms). Investigations on various erythroid and myeloid cell lines indicate an expression of the c- Kit gene in early stages of differentiation (Andre et al., Oncogene 4 (1989), 1047-1049). Certain tumors such as glioblastoma cells likewise exhibit a pronounced expression of the c-Kit gene.
[0046] Eph receptors, which include EphA and EphB subfamily, consist of the largest group of receptor tyrosine kinases. EphB was found to be overexpressed in several tumors including ovarian tumors, liver tumors, kidney tumors as well as melanomas. Downregulation of EphB signaling has shown to inhibit tumor growth and metastasis. Therefore, EphB may be an important target for anti-tumorigenic therapies. (Clevers et al., Cancer Res. 66:2-5 (2006); Heroult et al., Experimental Cell Res. 312: 642-650 (2006); and Batlle et al., Nature 435:1126- 1130 (2005)).
[0047] Kinase insert domain-containing receptor (referred to as "KDR" hereinafter) [WO 92/14748; Proc. Natl. Acad. Sci. USA, 88: 9026 (1991)]; Biochem. Biophys. Res. Comm., 187: 1579 (1992); WO 94/11499) and Fms-like tyrosine kinase (referred to as "Fltl" hereinafter) [Oncogene, 5: 519 (1990); Science, 255: 989 (1992)] belong to the receptor type tyrosine kinase family. It has been reported that VEGF specifically binds to Fit- 1 and KDR at Kd values of 20 pM and 75 pM and that Fltl and KDR are expressed in vascular endothelial cells in a specific manner [Proc. Natl. Acad. Sci. USA, 90: 7533 (1993); Proc. Natl. Acad. Sci. USA, 90: 8915 (1993)]. With regard to FIt-I in various diseases, it has been reported that, in comparison with vascular endothelial cells in normal tissues, expression of FIt-I mRNA increases in tumor vascular endothelial cells of human glioblastoma tissues [Nature, 359: 845 (1992)] and tumor vascular endothelial cells of human digestive organ cancer tissues [Cancer Research, 53: 4727 (1993)]. Additionally, it has been reported that expression of FIt-I mRNA is observed by in situ hybridization in vascular endothelial cells of joints of patients with rheumatoid arthritis [J. Experimental Medicine, 180: 341 (1994)]. Studies also suggest that FIt-I plays an important role in tumor angiogenesis.
[0048] Flt3 is a member of the type III receptor tyrosine kinase (RTK) family. Flt3 (Fms-like tyrosine kinase) is also known as FLk-2 (fetal liver kinase T). Aberrant expression of the Flt3 gene has been documented in both adult and childhood leukemias including acute myeloid leukemia (AML), AML with trilineage myelodysplasia (AML/TMDS), acute lymphoblastic leukemia (ALL), and myelodysplastic syndrome (MDS). 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. Inhibition of p-FLT3 kinase activity induces apoptosis (programmed cell death) of the leukemic cells.
[0049] Abelson tyrosine kinase (i.e. AbI, c-Abl) is involved in the regulation of the cell cycle, in the cellular response to geno toxic stress, and in the transmission of information about the cellular environment through integrin signaling. The AbI protein appears to serve 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 important in the pathogenesis of 95% of chronic myelogenous leukemia (CML) and 10% of acute lymphocytic leukemia.
[0050] Compounds of the present invention may inhibit AbI kinase, for example, v-Abl kinase. The compounds of the present invention may also inhibit wild-type Bcr-Abl kinase and mutations of Bcr-Abl kinase, and thus may be suitable for the treatment of Bcr-Abl-positive cancer and tumor diseases, such as leukemias (e.g., chronic myeloid leukemia and acute lymphoblastic leukemia, where especially apoptotic mechanisms of action are found) and other proliferation disorders related to Bcr-Abl. Compounds of the present invention may also be effective against leukemic stem cells, and may be potentially useful 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).
[0051] Anaplastic lymphoma kinase (ALK), a member of the insulin receptor superfamily of receptor tyrosine kinases, has been implicated in oncogenesis in hematopoietic and non- hematopoietic tumors. The aberrant expression of full-length ALK receptor proteins has been reported in neuroblastomas and glioblastomas; and ALK fusion proteins have occurred in anaplastic large cell lymphoma. The study of ALK fusion proteins has also raised the possibility of new therapeutic treatments for patients with ALK-positive malignancies. (Pulford et al., Cell. MoI. Life Sci. 61:2939-2953 (2004)).
[0052] Aurora-A, a serine/threonine mitotic kinase, has been reported to be overexpressed in various human cancers, and its overexpression induces aneuploidy, centrosome amplification and tumorigenic transformation in cultured human and rodent cells. (Zhang et al., Oncogene 23:8720-30 (2004)).
[0053] Bmx/Etk non-receptor tyrosine protein kinase has been implicated in endothelial cell migration and tube formation in vitro. Bmx in endothelium and bone marrow has also been reported to play an important role in arteriogenesis and angiogenesis in vivo, suggesting that Bmx may be a novel target for the treatment of vascular diseases such as coronary artery disease and peripheral arterial disease. (He et al., J. Clin. Invest. 116:2344-2355 (2006)).
[0054] Breast tumor kinase (Brk) is a soluble protein-tyrosine kinase overexpressed in the majority of breast cancers and also in normal skin and gut epithelium, but not in normal breast epithelial cells. (Zhang et al., J Biol. Chem. 280:1982-1991 (2005)).
[0055] The Janus kinases (JAK) are a family of tyrosine kinases consisting of JAKl, JAK2, JAK3 and TYK2. The JAKs play an important role in cytokine signaling. The down-stream substrates of the JAK family of kinases include the signal transducer and activator of transcription (STAT) proteins. JAK/STAT signaling has been implicated in the mediation of many abnormal immune responses such as allergies, asthma, autoimmune diseases such as transplant rejection, rheumatoid arthritis, amyotrophic lateral sclerosis and multiple sclerosis as well as in solid and hematologic malignancies such as leukemias and lymphomas.
[0056] One of the most important factors in the tumor angiogenesis is vascular endothelium growth factor(VEGF). VEGF can promote and maintain the establishment of tumor vascular system, and can also promote the tumor growth directly. VEGF can induce the mitogenesis and chemotaxis of vascular endothelial cell (VEC) and tumor cell (TC). Almost all types of TC and tumor VEC can secret VEGF, but the expression of VEGF in the normal tissue is very low. In the four VEGF receptors, KDR is the main receptor which gives play to VEGF functions, while other receptors play little role in cell growth. KDR is highly expressed on the TC and tumor VEC while lowly expressed on the normal tissues. (Ren et al., World J. Gastroentrol. 8:596-601 (2002)).
[0057] Mitogen- activated protein kinases (MAPKs) are members of conserved signal transduction pathways that activate transcription factors, translation factors and other target molecules in response to a variety of extracellular signals. MAPKs are activated by phosphorylation at a dual phosphorylation motif having the sequence Thr-X-Tyr by mitogen- activated protein kinase kinases (MKKs). In higher eukaryotes, the physiological role of MAPK signaling has been correlated with cellular events such as proliferation, oncogenesis, development and differentiation. Accordingly, the ability to regulate signal transduction via these pathways (particularly via MKK4 and MKK6) could lead to the development of treatments and preventive therapies for human diseases associated with MAPK signaling, such as inflammatory diseases, autoimmune diseases and cancer.
[0058] 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-lβ. Peripheral blood monocytes (PBMCs) have been shown to express and secrete pro-inflammatory cytokines when stimulated with lipopoly saccharide (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.
[0059] 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.
[0060] 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 may inhibit PDGF receptor (PDGFR) activity, and may therefore be suitable for the treatment of tumor diseases, such as gliomas, sarcomas, prostate tumors, and tumors of the colon, breast, and ovary.
[0061] Compounds of the present invention, may 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. Compounds of the present invention may also be useful 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 may especially be used for the treatment of diseases, which respond to an inhibition of the PDGF receptor kinase.
[0062] Compounds of the present invention may exhibit useful effects in the treatment of disorders arising as a result of transplantation, for example, allogenic transplantation, especially tissue rejection, such as 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.
[0063] Compounds of the present invention may also be effective against diseases associated with vascular smooth-muscle cell migration and proliferation (where PDGF and PDGFR 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 may 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.
[0064] Protein kinase C (PKC) functions in processes relevant to carcinogenesis, tumor cell metastasis, and apoptosis. PKCα is associated with a diverse range of cancers and is previously shown to be overexpressed in three out of four antiestrogen resistant breast cancer cell lines. (Frankel et al., Breast Cancer Res Treat. 2006 Oct. 24 (ePub)).
[0065] The stress activated protein kinases (SAPKs) 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. In particular, 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.
[0066] The region encompassing the SNFlLK locus (also known as SIK) has been implicated in congenital heart defects often observed in patients with Down syndrome. Snf Ilk is also expressed in skeletal muscle progenitor cells of the somite beginning at 9.5 dpc, suggesting a more general role for snf Ilk in the earliest stages of muscle growth and/or differentiation. (Genomics 83:1105-15 (2004)).
[0067] Syk is a tyrosine kinase that plays an important role in mast cell degranulation and eosinophil activation. Accordingly, Syk kinase is implicated in various allergic disorders, in particular asthma. It has been shown that Syk binds to the phosphorylated gamma chain of the FcεRl receptor via N-terminal SH2 domains and is important for downstream signaling.
[0068] Lin et al., J. Clin. Invest. 100, 8: 2072-2078 (1997) and P. Lin, PNAS 95, 8829-8834, (1998) have shown an inhibition of tumor growth and vascularization and also a decrease in lung metastases during adenoviral infections or during injections of the extracellular domain of Tie-2 (Tek) in breast tumor and melanoma xenograft models. 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).
[0069] The Trk family of neurotrophin receptors (TrkA, TrkB, TrkC) 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. Moreover, TrkB is 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-gamma transduction pathway (Sugimoto et al., Jpn J. Cancer Res. 2001 Feb; 92(2): 152-60).
[0070] The class III receptor tyrosine kinases (RTKs), which include c-FMS, c-KIT, FLT3, platelet-derived growth factor receptor α(PDGFRα) and /^PDGFR/)), have been reported to be associated with the pathogenesis of an increasing number of malignancies. (Blume-Jensen et al., Nature 411:355-565 (2001); Scheijin et al., Oncogene 21:3314-3333 (2002)).
[0071] 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 (1) 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
[0072] 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.
[0073] Compounds of the invention may 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.
[0074] 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 may 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, together with c) binders, e.g., magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose and or polyvinylpyrrolidone; and 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.
[0075] 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, may be aqueous solutions, ointments, creams or gels well-known in the art. Such may contain solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives.
[0076] Compounds of the invention may 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.
[0077] 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.
Processes for Making Compounds of the Invention
[0078] General procedures for preparing compounds of the invention are described in the Examples, infra. In the reactions described, reactive functional groups, for example hydroxy, amino, imino, thio or carboxy groups, where these are desired in the final product, may be protected to avoid their unwanted participation in the reactions. Conventional protecting groups may be used in accordance with standard practice (see e.g., T. W. Greene and P. G. M. Wuts in "Protective Groups in Organic Chemistry", John Wiley and Sons, 1991).
[0079] A compound of the invention may 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 may 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 may be prepared using salts of the starting materials or intermediates.
[0080] The free acid or free base forms of the compounds of the invention may 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 may 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 may be converted to the corresponding free acid by treating with a suitable acid (e.g., hydrochloric acid, etc.). [0081] Compounds of the invention in unoxidized form may 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 8O0C.
[0082] Prodrug derivatives of the compounds of the invention may 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 may 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).
[0083] Protected derivatives of the compounds of the invention may 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.
[0084] Compounds of the present invention may be conveniently prepared or formed during the process of the invention, as solvates (e.g., hydrates). Hydrates of compounds of the present invention may be conveniently prepared by recrystallization from an aqueous/organic solvent mixture, using organic solvents such as dioxin, tetrahydrofuran or methanol.
[0085] Compounds of the invention may 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. Resolution of enantiomers may be carried out using covalent diastereomeric derivatives of the compounds of the invention, or by using dissociable complexes (e.g., crystalline diastereomeric salts). Diastereomers have distinct physical properties (e.g., melting points, boiling points, solubility, reactivity, etc.) and may be readily separated by taking advantage of these dissimilarities. The diastereomers may be separated by chromatography, or 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.
[0086] In summary, compounds having Formula (1) or (2) may be made by a process as described in the Examples; and
(a) optionally converting a compound of the invention into a pharmaceutically acceptable salt;
(b) optionally converting a salt form of a compound of the invention to a non-salt form;
(c) optionally converting an unoxidized form of a compound of the invention into a pharmaceutically acceptable N-oxide;
(d) optionally converting an N-oxide form of a compound of the invention to its unoxidized form;
(e) optionally resolving an individual isomer of a compound of the invention from a mixture of isomers;
(f) optionally converting a non-derivatized compound of the invention into a pharmaceutically acceptable prodrug derivative; and
(g) optionally converting a prodrug derivative of a compound of the invention to its non- derivatized form.
[0087] Insofar as the production of the starting materials is not particularly described, the compounds are known or may 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.
[0088] The following examples are offered to illustrate but not to limit the invention.
Example 1
2-(3-tert-Butyl-ureido)-benzothiazole-6-carboxylic acid r2-methyl-5-(3-trifluoromethyl- benzoylamino)-phenyll-amide
Figure imgf000024_0001
Figure imgf000024_0002
2-(3-fe?t-Butylureido)benzothiazole-6-carboxylic acid r2-methyl-5-(3- trifluoromethylbenzoylamino)phenyllamide
Figure imgf000024_0003
[0089] To a flask is added 2-te?t-butoxycarbonylaminobenzothiazole-6-carboxylic acid (3.0 g), N-(3-amino-4-methylphenyl)-3-trifluoromethylbenzamide (3.6 g), HATU (4.7 g), NN- diisopropylethylamine (2.1 mL) and DMF (30 mL). The mixture is stirred at rt overnight, then poured into water. The yellow precipitate is collected by filtration, air dried, then washed with EtOAc to afford the desired product. 1H ΝMR (400 MHz, DMSO) δ 12.05 (s, IH), 10.50 (s, IH), 10.00 (s, IH), 8.65 (s, IH), 8.40 (s, IH), 8.30 (d, J = 7.0 Hz, IH), 8.05 (d, J = 7.2 Hz, IH), 8.00 (d, J = 7.0 Hz, IH), 7.90 (s, IH), 7.80 (t, J = 7.0 Hz, 2H), 7.70 (d, J = 7.0 Hz, IH), 7.25 (d, J = 7.2 Hz, IH), 2.30 (s, 3H), 1.55 (s, 9H). MS (ESI) m/z: 571 (M+H)+. 2-Amino-benzothiazole-6-carboxylic acid r2-methyl-5-(3-trifluoromethyl-benzoylamino)- phenyll -amide
Figure imgf000025_0001
[0090] {6-[2-Methyl-5-(3-trifluoromethyl-benzoylamino)-phenylcarbamoyl]-benzothiazol-2- yl}-carbamic acid tert-butyl ester (100 mg) is dissolved in TFA (5 mL) and the mixture is stirred at rt for 30 min, then concentrated in vacuo. The residue is dissolved in EtOAc, then washed with aqueous NaHCU3. The organic phase is separated, dried with MgSO4, then concentrated and the residue is washed with DCM to afford 2-amino-benzothiazole-6-carboxylic acid [2-methyl-5-(3- trifluoromethyl-benzoylamino)-phenyl]-amide. 1H NMR (400 MHz, DMSO) δ 10.60 (s, IH), 9.90 (s, IH), 8.40 (s, IH), 8.35 (d, J = 7.0 Hz, IH), 8.05 (d, J = 7.0 Hz, IH), 8.00 (d, J = 7.0 Hz, IH), 7.90 (m, 2H), 7.70 (d, IH), 7.50 (d, J = 7.2 Hz, IH), 7.35 (d, J = 7.0 Hz, IH), 2.30 (s, 3H). MS (ESI) m/z: 471 (M+H)+.
2-(3-tert-Butyl-ureido)-benzothiazole-6-carboxylic acid r2-methyl-5-(3-trifluoromethyl- benzoylamino)-phenyll -amide
Figure imgf000025_0002
[0091] To a sealed vial is added 2-amino-benzothiazole-6-carboxylic acid [2-methyl-5-(3- trifluoromethyl-benzoylamino)-phenyl]-amide (50 mg), te?t-butylisocyanate (42 mg) and THF (2 mL). The mixture is heated to 70 0C overnight, then concentrated in vacuo and the residue is washed with DCM to afford 2-(3-tert-butyl-ureido)-benzothiazole-6-carboxylic acid [2-methyl-5- (3-trifluoromethyl-benzoylamino)-phenyl]-amide. 1H NMR (400 MHz, DMSO) δ 9.98 (s, IH), 8.55 (s, IH), 8.35 (s, IH), 8.30 (d, J = 7.0 Hz, IH), 8.00 (m, 2H), 7.85 (s, IH), 7.80 (t, J = 7.0 Hz, IH), 7.70 (d, J = 7.1 Hz, IH), 7.61 (d, J = 7.0 Hz, IH), 7.25 (d, J = 7.0 Hz, IH), 6.70 (s, IH), 2.20 (s, 3H), 0.35 (s, 9H). MS (ESI) m/z: 570 (M+H)+. Example 2
{6-r2-Methyl-5-(3-trifluoromethylbenzoylamino)phenylcarbamoyllbenzothiazol-2-yllcarbamic acid methyl ester
Figure imgf000026_0001
[0092] To a vial is added 2-amino-benzothiazole-6-carboxylic acid [2-methyl-5-(3- trifluoromethyl-benzoylamino)-phenyl]-amide (30 mg), methyl chloroformate (7.2 mg) and THF (2 mL). The mixture is stirred at rt overnight, then concentrated in vacuo and the residue is purified by reverse phase HPLC to afford {6-[2-methyl-5-(3-trifluoromethyl-benzoylamino)- phenylcarbamoyl]-benzothiazol-2-yl}-carbamic acid methyl ester. 1H NMR (400 MHz, DMSO) δ 10.50 (s, IH), 10.00 (s, IH), 8.60 (s, IH), 8.30 (m, 2H), 8.05 (d, J = 7.0 Hz, IH), 7.95 (d, J = 7.0 Hz, IH), 7.85 (s, IH), 7.80 (m, IH), 7.60 (d, J = 7.0 Hz, IH), 7.45 (d, J = 7.0 Hz, IH), 7.25 (m, IH), 3.85 (s, 3H), 2.22 (s, 3H). MS (ESI) m/z: 529 (M+H)+.
Example 3
2-(3-Ethyl-ureido)-benzothiazole-6-carboxylic acid r2-methyl-5-(3-methyl-benzoylamino)- phenyll- amide
Figure imgf000026_0002
Figure imgf000026_0003
2-(3-Ethylureido)benzothiazole-6-carboxylic acid r2-methyl-5-(3-methylbenzoylamino) phenyll amide
Figure imgf000027_0001
[0093] To a screw cap tube is added 2-aminobenzothiazole-6-carboxylic acid ethyl ester (2.0 g), ethyl isocyanate (3.2 g) and THF (30 niL). The mixture is heated to 70 0C for 3 hrs, then concentrated in vacuo and the residue is washed with EtOAc to afford 2-(3-ethyl-ureido)- benzothiazole-6-carboxylic acid ethyl ester. 1H NMR (400 MHz, DMSO) δ 10.98 (s, IH), 8.50 (s, IH), 7.95 (d, J = 6.8 Hz, IH), 7.70 (d, J = 7.0 Hz, IH), 6.80 (s, IH), 4.30 (q, J = 6.8 Hz, 2H), 3.18 (t, J = 6.8 Hz, 2H), 1.32 (t, J = 6.8 Hz, 3H), 1.10 (t, J = 6.8 Hz, 3H). MS (ESI) m/z: 294 (M+H)+.
2-(3-Ethyl-ureido)-benzothiazole-6-carboxylic acid:
Figure imgf000027_0002
[0094] 2-(3-Ethyl-ureido)-benzothiazole-6-carboxylic acid ethyl ester (2.0 g) is dissolved in 1,4-dioxane (50 niL) at 60 0C, then aqueous LiOH (0.82 g in 30 mL H2O) is added. The mixture is kept at 60 0C for 6 hrs, and then acetic acid is added slowly until the solution became slightly acidic. After being concentrated in vacuo to remove the organic phase, a white solid is precipitated out, which is collected by filtration and air dried to afford 2-(3-ethyl-ureido)- benzothiazole-6-carboxylic acid. 1H NMR (400 MHz, DMSO) δ 8.45 (s, IH), 7.95 (d, J = 7.0 Hz, IH), 7.60 (d, 7 = 7.0 Hz, IH), 3.15 (p, J = 6.7 Hz, 2H), 1.10 (t, 7 = 6.7 Hz, 3H). MS (ESI) m/z: 266 (M+H)+.
(3-{ r2-(3-Ethyl-ureido)-benzothiazole-6-carbonyll-aminol-4-methyl-phenyl)-carbamic acid tert- butyl ester
Figure imgf000028_0001
[0095] To a flask is added 2-(3-ethyl-ureido)-benzothiazole-6-carboxylic acid (0.10 g), 3- amino-4-methylphenyl carbamic acid tert-butyl ester (0.083 g), HATU (0.14 g), Et3N (65 μL) and DMF (3 mL). After being stirred at rt overnight, the mixture is poured into water and extracted with EtOAc. The organic layer is separated, then concentrated and the residue is washed with DCM to afford (3-{ [2-(3-ethyl-ureido)-benzothiazole-6-carbonyl]-amino}-4-methyl-phenyl)- carbamic acid tert-butyl ester. 1H NMR (400 MHz, DMSO) δ 10.90 (s, IH), 9.85 (s, IH), 9.30 (s, IH), 8.50 (s, IH), 8.00 (d, J = 6.8 Hz, IH), 7.70 (d, J = 6.8 Hz, IH), 7.50 (s, IH), 7.20 (d, J = 6.8 Hz, IH), 7.10 (d, J = 7.0 Hz, IH), 6.80 (s, IH), 3.20 (p, J = 6.7 Hz, 2H), 2.15 (s, 3H), 1.45 (s, 9H), 1.10 (t, J = 6.7 Hz, 2H). MS (ESI) m/z: 470 (M+H)+.
2-(3-Ethyl-ureido)-benzothiazole-6-carboxylic acid (5-amino-2-methyl-phenyl)-amide
Figure imgf000028_0002
[0096] (3-{ [2-(3-Ethyl-ureido)-benzothiazole-6-carbonyl]-amino}-4-methyl-phenyl)- carbamic acid tert-butyl ester (0.20 g) is dissolved in TFA (10 mL). After being stirred for 1 hr, the mixture is concentrated in vacuo. The residue is dissolved in EtOAc and washed with aqueous NaHCO3. The organic phase is separated then concentrated, and the residue is further washed with EtOAc to afford 2-(3-ethyl-ureido)-benzothiazole-6-carboxylic acid (5-amino-2-methyl- phenyl)-amide. 1H NMR (400 MHz, DMSO) δ 9.45 (s, IH), 8.30 (s, IH), 7.85 (s, IH), 7.60 (s, IH), 7.40 (s, IH), 6.85 (d, J = 7.0 Hz, IH), 6.67 (s, IH), 6.35 (d, J = 7.0 Hz, IH), 4.90 (s, 2H), 3.15 (p, J = 6.7 Hz, 2H), 2.05 (s, 3H), 1.15 (t, J = 6.7 Hz, 3H). MS (ESI) m/z: 370 (M+H)+. 2-(3-Ethyl-ureido)-benzothiazole-6-carboxylic acid r2-methyl-5-(3-methyl-benzoylamino)- phenyll -amide
Figure imgf000029_0001
[0097] To a vial is added 2-(3-ethyl-ureido)-benzothiazole-6-carboxylic acid (5-amino-2- methyl-phenyl)-amide (20 mg), HATU (21 mg), Hϋnig base (10 μL), 3-methylbenzoic acid (7.4 mg) and DMF (0.4 mL). The mixture is stirred at rt overnight, then purified by HPLC to afford 2- (3-ethyl-ureido)-benzothiazole-6-carboxylic acid [2-methyl-5-(3-methyl-benzoylamino)-phenyl]- amide. 1H NMR (400 MHz, DMSO) δ 10.95 (s, IH), 10.25 (s, IH), 9.95 (s, IH), 8.55 (s, IH), 8.00 (d, J = 7.0 Hz, IH), 7.85 (s, IH), 7.75 (m, 2H), 7.60 (d, J = 7.0 Hz, IH), 7.40 (s, IH), 7.25 (d, J = 7.0 Hz, IH), 6.80 (m, IH), 3.20 (p, J = 6.7 Hz, 2H), 2.42 (s, 3H), 2.25 (s, 3H), 1.10 (t, J = 6.7 Hz, 3H). MS (ESI) m/z: 488 (M+H)+.
Example 4
2-(3-Ethyl-l-methyl-ureido)-benzothiazole-6-carboxylic acid r2-methyl-5-(3-trifluoromethyl- benzoylamino)-phenyll-amide
B
Figure imgf000029_0002
Methyl-{6-r2-methyl-5-(3-trifluoromethyl-benzoylamino)-phenylcarbamoyll-benzothiazol-2- yll-carbamic acid tert-butyl ester
Figure imgf000030_0001
[0098] {6-[2-Methyl-5-(3-trifluoromethyl-benzoylamino)-phenylcarbamoyl]-benzothiazol-2- yl}-carbamic acid tert-butyl ester (57 mg, 0.1 mmol) is dissolved in 2 mL of anhydrous THF, triphenyl phosphine (39 mg, 0.15 mmol), methanol (4 μL, 0.1 mmol) and DIAD (28 μL, 0.15 mmol) is added. The reaction is stirred at rt overnight. The solvent is removed in vacuo and the crude product is purified using flash chromatography (hexane: ethyl acetate/4: 1) to give methyl- {6-[2-methyl-5-(3-trifluoromethyl-benzoylamino)-phenylcarbamoyl]-benzothiazol-2-yl}- carbamic acid tert-butyl ester. 1H NMR (400 MHz, DMSO) δ 10.50 (s, IH), 10.06 (s, IH), 8.59 (d, IH), 8.31 (s, IH), 8.27 (d, IH), 8.05 (dd, IH), 7.97 (d, IH), 7.87 (d, IH), 7.86 (s, IH), 7.79 (t, IH), 7.61 (dd, IH), 7.28 (d, IH), 3.61 (s, 3H), 2.25 (s, 3H), 1.58 (s, 9H). MS(ESI) m/z: 585.2 (M+ 1) +.
2-Methylamino-benzothiazole-6-carboxylic acid r2-methyl-5-(3-trifluoromethyl-benzoylamino)- phenyll -amide
Figure imgf000030_0002
[0099] Methyl-{6-[2-methyl-5-(3-trifluoromethyl-benzoylamino)-phenylcarbamoyl]- benzothiazol-2-yl}-carbamic acid tert-butyl ester (28 mg, 0.0479 mmol) is dissolved in trifluoroacetic acid (3 mL). The mixture is stirred at rt for 2 hrs. The solvent is removed in vacuo to give 2-methylamino-benzothiazole-6-carboxylic acid [2-methyl-5-(3-trifluoromethyl- benzoylamino)-phenyl]-amide. 1H NMR (400 MHz, DMSO) δ 10.48 (s, IH), 9.81 (s, IH), 8.36- 8.35 (br s, IH), 8.34 (d, IH), 8.31 (s, IH), 8.27 (d, IH), 7.97 (d, IH), 7.90 (dd, IH), 7.85 (d, IH), 7.79 (t, IH), 7.61 (dd, IH), 7.48 (d, IH), 7.26 (d, IH), 2.99 (d, 3H), 2.23 (s, 3H). MS(ESI) m/z: 485.1 (M+l) +.
2-(3-Ethyl-l-methyl-ureido)-benzothiazole-6-carboxyric acid r2-methyl-5-(3-trifluoromethyl- benzoylaminoVphenyll -amide
Figure imgf000031_0001
[0010O] To a solution of 2-methylamino-benzothiazole-6-carboxylic acid [2-methyl-5-(3- trifluoromethyl-benzoylamino)-phenyl]-amide (10.4 mg, 0.0214 mmol) in THF (1 mL) is added triethylamine (9 μL, 0.064 mmol) and ethyl isocyanate (3.4 μL, 0.043 mmol). The mixture is stirred at 65 0C overnight. The solvent is removed in vacuo and the residue is purified by prep HPLC to give 2-(3-ethyl-l-methyl-ureido)-benzothiazole-6-carboxylic acid [2-methyl-5-(3- trifluoromethyl-benzoylamino)-phenyl]-amide. MS(ESI)m/z: 556.2 (M+l) +.
Example 5
2-(2-Morpholin-4-yl-ethylamino)-benzothiazole-6-carboxylic acid r4-bromo-2-methyl-5-(3- trifluoromethyl-benzoylamino)-phenyll-amide
Figure imgf000032_0001
Figure imgf000032_0002
Figure imgf000032_0003
Benzothiazole-6-carboxylic acid r2-methyl-5-(3-trifluoromethyl-benzoylamino)-phenyl1-amide
Figure imgf000032_0004
[0010I] To a solution of 2-amino-benzothiazole-6-carboxylic acid [2-methyl-5-(3- trifluoromethyl-benzoylamino)-phenyl]-amide (100 mg, 0.212 mmol) in dioxane (3 mL) is added tert-butyl nitrite (62 μL, 90%, 0.468 mmol). The mixture is stirred at 85 0C for 1 h. The solvent is removed in vacuo and the residue is purified using flash chromatography (hexane:ethyl acetate/2: 1) to give benzothiazole-6-carboxylic acid [2-methyl-5-(3-trifluoromethyl- benzoylamino)-phenyl]-amide. 1H NMR (400 MHz, DMSO) δ 10.50 (s, IH), 10.10 (s, IH), 9.57 (s, IH), 8.31 (s, IH), 8.27 (d, IH), 8.22 (d, IH), 7.97 (d, IH), 7.88 (d, IH), 7.78 (t, IH), 7.78 (t, IH), 7.62 (dd, IH), 7.28 (d, IH), 2.25 (s, 3H). MS(ESI) m/z: 456.1 (M+l) +. 2-Bromo-benzothiazole-6-carboxylic acid r4-bromo-2-methyl-5-(3-trifluoromethyl- benzoylamino)-phenyll -amide
Figure imgf000033_0001
[00102] To a solution of 2-amino-benzothiazole-6-carboxylic acid [2-methyl-5-(3- trifluoromethyl-benzoylamino)-phenyl]-amide (117 mg, 0.25 mmol) and CuBr2 (112 mg, 0.5 mmol) in dioxane (10 mL) is added tert-butyl nitrite (66 μL, 90%, 0.5 mmol) at 0 0C. The mixture is stirred at rt overnight. The mixture is extracted with ethyl acetate. The organic phase is washed with saturated NaHCθ3 solution and brine. The solvent is removed in vacuo and the crude product is purified using flash chromatography (hexane: ethyl acetate/2: 1) to give 2-bromo- benzothiazole-6-carboxylic acid [4-bromo-2-methyl-5-(3-trifluoromethyl-benzoylamino)- phenyl]-amide. 1H NMR (400 MHz, CD3OD) δ 8.61 (s, IH), 8.29 (s, IH), 8.24 (d, IH), 8.09 (s, IH), 8.08 (d, IH), 7.92 (dd, IH), 7.77 (s, IH), 7.78 (t, IH), 7.67 (s, IH), 2.25 (s, 3H). MS(ESI) m/z: 611.9 (M+ 1) +.
2-(2-Morpholin-4-yl-ethylamino)-benzothiazole-6-carboxylic acid r4-bromo-2-methyl-5-(3- trifluoromethyl-benzoylamino)-phenvH-amide
Figure imgf000033_0002
[00103] A mixture of 2-bromo-benzothiazole-6-carboxylic acid [4-bromo-2-methyl-5-(3- trifluoromethyl-benzoylamino)-phenyl]-amide (20 mg, 0.0326mmol), 2-morpholinoethylamine (44 μL, 0.326 mmol), and DMF (0.8 mL) is heated to 100 0C for 2 hrs. The mixture is cooled down to room temperature and the crude mixture is separated with prep HPLC to yield 2-(2- Morpholin-4-yl-ethylamino)-benzothiazole-6-carboxylic acid [4-bromo-2-methyl-5-(3- trifluoromethyl-benzoylamino)-phenyl]-amide. MS(ESI) m/z: 662.1 (M+l) +.
Example 6 2-(5-Fluoro-pyridin-2-ylamino)-benzothiazole-6-carboxylic acid r2-methyl-5-(3-trifluoromethyl- benzoylamino)-phenyll-amide
Figure imgf000034_0001
2-Bromo-benzothiazole-6-carboxylic acid methyl ester
Figure imgf000034_0002
[00104] To a solution of 2-amino-benzothiazole-6-carboxylic acid methyl ester (1.11 g, 5 mmol) and CuBr2 (2.23 g, 10 mmol) in CH3CN (50 mL) is added tert-butyl nitrite (1.32 mL, 90%, 10 mmol) at 0 0C. The mixture is stirred at rt overnight. The mixture is extracted with ethyl acetate. The organic phase is washed with saturated NaHCO3 solution and brine. The solvent is removed in vacuo and the crude product is purified using flash chromatography (hexane: ethyl acetate/10: 1) to give 2-bromo-benzothiazole-6-carboxylic acid methyl ester. 1H NMR (400 MHz, CDCl3) δ 8.53 (d, IH), 8.15 (dd, IH), 8.01 (d, IH), 4.42 (q, 2H), 1.42 (t, 3H). MS(ESI) m/z: 271.9 (M+l) +.
2-Bromo-benzothiazole-6-carboxylic acid
Figure imgf000035_0001
[00105] To a solution of 2-bromo-benzothiazole-6-carboxylic acid methyl ester (1.256 g, 4.39 mmol) in THF (60 mL) and H2O (20 mL) is added lithium hydroxide monohydrate (920 mg, 21.9 mmol) at 0 0C. The mixture is stirred at rt overnight. The mixture is treated with IN HCl solution to adjust pH to 2 and extracted with ethyl acetate. The organic phase is washed with brine. The solvent is removed in vacuo and the crude product is used for next step without further purification. MS(ESI) m/z: 257.9 (M+l) +.
2-Chloro-benzothiazole-6-carboxylic acid r2-methyl-5-(3-trifluoromethyl-benzoylamino)- phenyll -amide
Figure imgf000035_0002
[0100] To a solution of 2-bromo-benzothiazole-6-carboxylic acid (984 mg, 3.81 mmol) in CH2Cl2 (40 mL) is added DMF (70 μL) and oxalyl chloride (6.6 mL, 76 mmol). After the addition is complete, the reaction is stirred at rt for an additional hour. After solvent removal in vacuo, the crude product is used without further purification. The corresponding acetyl chloride is dissolved in 20 mL CH2Cl2. The solution is added via cannula to a solution of N-(3-amino-4- methyl-phenyl)-3-trifluoromethyl-benzamide (1.12 g, 3.81 mmol) and diisopropanylethylamine (1.65 mL, 9.52 mmol) in 40 mL CH2Cl2 at 0 0C over 30 min. The mixture is stirred at rt for 1 h. The mixture is extracted with ethyl acetate. The organic phase is washed with saturated ΝaHCU3 solution and brine. The solvent is removed in vacuo and the crude product is purified by recrystallization (hexane:CH2Cl2/10:l) to give 2-chloro-benzothiazole-6-carboxylic acid [2- methyl-5-(3-trifluoromethyl-benzoylamino)-phenyl]-amide. 1H NMR (400 MHz, DMSO) δ 10.50 (s, IH), 10.12 (s, IH), 8.75 (s, IH), 8.30 (s, IH), 8.26 (d, IH), 8.13 (s, 2H), 7.97 (d, IH), 7.87 (d, IH), 7.78 (d, IH), 7.61 (dd, IH), 7.29 (d, IH), 2.24 (s, 3H). MS(ESI) m/z: 490.1 (M+ 1)
2-(2-Morpholin-4-yl-ethylamino)-benzothiazole-6-carboxylic acid r2-methyl-5-(3- trifluoromethyl-benzoylamino)-phenvH-amide
Figure imgf000036_0001
[0101] A mixture of 2-chloro-benzothiazole-6-carboxylic acid [2-methyl-5-(3- trifluoromethyl-benzoylamino)-phenyl]-amide (16 mg, 0.03 mmol), 2-morpholinoethylamine (39 μL, 0.3 mmol), and DMF (0.8 mL) is heated to 100 0C for 2 hrs. The mixture is cooled down to room temperature and the crude mixture is separated with prep HPLC to yield 2-(2- morpholin-4-yl-ethylamino)-benzothiazole-6-carboxylic acid [2-methyl-5-(3-trifluoromethyl- benzoylamino)-phenyl] -amide. 1H NMR (400 MHz, DMSO) δ 10.48 (s, IH), 9.84 (s, IH), 8.54 (t, IH), 8.37 (d, IH), 8.30 (s, IH), 8.26 (d, IH), 7.97 (d, IH), 7.93 (dd, IH), 7.87 (d, IH), 7.78 (t, IH), 7.57 (dd, IH), 7.52 (d, IH), 7.26 (d, IH), 3.90-3.80 (m, 2H), 3.82 (m, 4H), 3.60-3.43 (m, 6H), 2.20 (s, 3H). MS(ESI) m/z: 584.2 (M+ 1) +. 2-(5-Fluoro-pyridin-2-ylamino)-benzothiazole-6-carboxyric acid r2-methyl-5-(3-trifluoromethyl- benzoylamino)-phenyll -amide
Figure imgf000037_0001
[0102] A mixture of 2-chloro-benzothiazole-6-carboxylic acid [2-methyl-5-(3- trifluoromethyl-benzoylamino)-phenyl]-amide (30 mg, 0.061 mmol), 2-amino-5-fluoropyridine (27 mg, 0.245 mmol) in DMF (0.8 niL) is treated with sodium hydride (60%, 15 mg, 0.366 mmol) and heated to 80 0C for 2 hrs. The mixture is cooled down to room temperature and treated with acetic acid (60 μL). The solvent is removed in vacuo and the crude mixture is separated with prep HPLC to yield 2-(5-fluoro-pyridin-2-ylamino)-benzothiazole-6-carboxylic acid [2-methyl-5-(3-trifluoromethyl-benzoylamino)-phenyl]-amide. 1H NMR (400 MHz, DMSO) δ 11.88 (s, IH), 10.49 (s, IH), 9.92 (s, IH), 8.54 (d, 1H),8.41 (d, IH), 8.31 (s, IH), 8.27 (d, IH), 8.02 (dd, IH), 7.97 (d, IH), 7.87 (d, IH), 7.82 (dd, IH), 7.79 (t, IH), 7.71 (d, IH), 7.61 (d, IH), 7.29 (d, IH), 7.26 (d, IH), 2.25 (s, 3H). MS(ESI) m/z: 566.1 (M+l) +.
Example 7
2-(3-Ethyl-ureido)-benzothiazole-6-carboxylic acid r2-methyl-5-(3-trifluoromethyl- phenylcarbamovD-phenyll-amide
Figure imgf000038_0001
{6-r2-Methyl-5-(3-trifluoromethyl-phenylcarbamoyl)-phenylcarbamoyll-benzothiazol-2-yl|- carbamic acid tert-butyl ester
BocH
Figure imgf000038_0002
[0103] To a solution of 2-fe?t-butoxycarbonylamino-benzothiazole-6-carboxylic acid (294 mg, 1 mmol) and N-(3-amino-4-methyl-phenyl)-3-trifluoromethyl-benzamide (324 mg, 1.1 mmol) in CH2Cl2 (6 mL) and DMF (3 mL) is added iPr2ΝEt (0.52 mL, 3 mmol) and HATU (570 mg, 1.5 mmol). The mixture is stirred at rt overnight. Solvent is removed in vacuo and the crude product is purified using flash chromatography (hexane: ethyl acetate/2: 1) to give {6-[2-Methyl- 5-(3-trifluoromethyl-phenylcarbamoyl)-phenylcarbamoyl]-benzothiazol-2-yl}-carbamic acid te?t-butyl ester. 1H NMR (400 MHz, DMSO) δ 12.01 (s, IH), 10.53 (s, IH), 10.13 (s, IH), 8.60 (s, IH), 8.25 (s, IH), 8.08 (d, IH), 8.04 (dd, IH), 8.01 (d, IH), 7.84 (dd, IH), 7.79 (d, IH), 7.60 (t, IH), 7.46 (t, 2H), 2.34 (s, 3H), 1.58 (s, 9H). MS(ESI) m/z: 571.1 (M+l) +. 2-Amino-benzothiazole-6-carboxylic acid r2-methyl-5-(3-trifluoromethyl-phenylcarbamoyl)- phenyll -amide
Figure imgf000039_0001
[0104] {6-[2-Methyl-5-(3-trifluoromethyl-phenylcarbamoyl)-phenylcarbamoyl]- benzothiazol-2-yl}-carbamic acid tert-buty\ ester (285 mg, 0.5 mmol) is dissolved in trifluoroacetic acid (10 mL). The mixture is stirred at rt for 2 hrs. The solvent is removed in vacuo to give 2-amino-benzothiazole-6-carboxylic acid [2-methyl-5-(3-trifluoromethyl- phenylcarbamoyl)-phenyl]-amide as a TFA salt. 1H NMR (400 MHz, DMSO) δ 10.52 (s, IH), 9.94 (s, IH), 8.32 (d, IH), 8.25 (s, IH), 8.08 (d, IH), 8.00 (d, IH), 7.90 (dd, IH), 7.82-7.80 (m, 3H), 7.60 (t, IH), 7.45 (d, 2H), 7.42 (d, IH), 2.32 (s, 3H). MS(ESI) m/z: 471.1 (M+ 1) +.
2-(3-Ethyl-ureido)-benzothiazole-6-carboxylic acid r2-methyl-5-(3-trifluoromethyl- phenylcarbamovD-phenyll-amide
Figure imgf000039_0002
[0105] To a solution of 2-amino-benzothiazole-6-carboxylic acid [2-methyl-5-(3- trifluoromethyl-phenylcarbamoyl)-phenyl]-amide (20 mg, 0.0425 mmol) in THF (2 mL) is added ethyl isocyanate (4 μL, 0.051 mmol). The mixture is stirred at 60 0C overnight. The solvent is removed in vacuo and the residue is purified by prep HPLC to give 2-(3-ethyl-ureido)- benzothiazole-6-carboxylic acid [2-methyl-5-(3-trifluoromethyl-phenylcarbamoyl)-phenyl]- amide. 1H NMR (400 MHz, DMSO) δ 10.94 (s, IH), 10.53 (s, IH), 10.09 (s, IH), 8.65 (s, IH), 8.25 (s, IH), 8.08 (d, IH), 8.03-8.00 (m, 2H), 7.83 (dd, IH), 7.73 (d, IH), 7.60 (t, IH), 7.46 (t, 2H), 6.77 (brs, IH), 3.20 (q, 2H), 2.34 (s, 3H), 1.10 (t, 3H). MS(ESI) m/z: 542.1 (M+l) +. [0106] Representative compounds of the invention, prepared following the procedures described above, are set forth in Tables 1-7.
Table 1 (urea tail)
J =
J = J
J = J
(d, J
Figure imgf000040_0001
7.1
(s, =
(s, =
(m,
Figure imgf000041_0001
(s,
J = J
m/z:
(s, 7.0
(s,
(s, IH),
Figure imgf000042_0001
=
(s,
(m,
(m, (m,
Figure imgf000043_0001
(s, 8.00
2.20
(s,
(t, J
(s,
(s, (t, 654
Figure imgf000044_0001
(s,
(m, =
(s,
(m,
(s,
(m,
(s,
J =
MS
Figure imgf000045_0001
(s,
2.25 MS
(s, 6.8
(t, J
(s,
J =
Figure imgf000046_0001
(s, =
(s,
(s,
J (d, Hz, = 6.7
(s,
J =
Figure imgf000047_0001
(d,
(d, 3H),
Figure imgf000048_0001
Table 2 (urethane tail)
J
(s, 7.0 (d, Hz,
J Hz,
J (s,
Figure imgf000049_0001
(s, 7.0 (d, Hz, (s,
J 7.0
=
Figure imgf000050_0001
(s, 6.8
7
7 7.0
Figure imgf000051_0001
J Hz,
10.50 (dd,
(s,
Figure imgf000052_0001
Table 3 (substituted amino and amido tail)
(s, 7.0
(s,
(d,
(s,
(d,
8.42 (s, (s, 0.5 +
Figure imgf000053_0001
10.48
(t, (q,
(t,
s, (m,
(t,
Figure imgf000054_0001
(t,
7.90 (d, 2.22 +
(t,
(s,
IH), (dd,
Figure imgf000055_0001
(t,
(t,
(t,
(t, (q,
(t,
(t, (q,
Figure imgf000056_0001
(q,
(t,
(s, +
(t,
3H), (s, +
IH), (dd,
(s,
Figure imgf000057_0001
(s,
+ (s,
Figure imgf000058_0001
(s,
(t,
(s,
(s, 8.31-
(d,
Figure imgf000059_0001
(s,
(t, (d,
IH), (dd,
m/z:
(s,
IH), (d,
(s, (dd,
Figure imgf000060_0001
(s, IH), (d,
(s, +
(s,
(d,
7.26
Figure imgf000061_0001
(s, (t, 3H).
(s,
(d,
(s, (d,
Figure imgf000062_0001
(s,
(t,
(s,
(d,
(s, (dd,
(s,
Figure imgf000063_0001
(s,
(t, (s,
(d, (d,
(s, +
(s, (d,
(s,
(t,
Figure imgf000064_0001
(s,
(s, +
(d,
+
9.95 IH), (d, s, 3H),
(d,
(t,
Figure imgf000065_0001
(s,
(dd,
(s,
(d, IH),
Figure imgf000066_0001
(d,
(s, (s,
Figure imgf000067_0001
Table 4
(s,
8.61
(s, (d,
(s, (d,
Figure imgf000068_0001
(d, m/z:
(d,
Figure imgf000069_0001
Table 5
(d,
(d, (m,
(t, (t, +
(s,
Figure imgf000070_0001
(t, IH),
(t, (m,
(t, (d, +
Figure imgf000071_0001
(t, (m, 3H).
(d,
(t,
Figure imgf000072_0001
Physical Data
Structure 1H NMR 400 MHz (DMSO) and/or
MS(ESI) m/z
119 MS(ESI) m/z: 582.2 (M+l) +
120 MS(ESI) m/z: 596.2 (M+1) H
121 MS(ESI) m/z: 622.1 (M+1) H
Figure imgf000073_0001
Physical Data
Structure 1H NMR 400 MHz (DMSO) and/or
MS(ESI) m/z
122 MS(ESI) m/z: 614.2 (M+l) "
123 MS(ESI) m/z: 618.2 (M+l) +
Figure imgf000074_0001
Assays
[0107] Compounds of the present invention may be assayed to measure their capacity to inhibit a kinase panel, including but not limited to AIk, AbI, Aurora-A, B-Raf, Bcr-Abl, BRK, BIk, Bmx, c-Kit, c-Raf, cSRC, CSK, EphB, FLTl, Fms, Fyn, JAK2, KDR, Lck, Lyn, PDGFRα, PDGFRβ, PKCα, p38 (p38 MAP kinase, SAPK2α), SIK, Src, Syk, Tie2 and TrkB kinases.
B-Raf (Enzymatic assay)
[0108] Compounds of the invention may be 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 15μl is added to each well. The plates are incubated at 4 0C 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 (15μl/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 0C 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 fluorescent Attophos AP substrate (Promega) 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 Program (Excitation 455 nm, Emission 580 nm).
b-Raf (Cellular Assay)
[0109] Compounds of the invention are tested in A375 cells for their ability to inhibit phosphorylation of MEK. 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 370C 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.
Inhibition of cellular Bcr-Abl dependent proliferation (High Throughput method) [0110] The murine cell line 32D hemopoietic progenitor cell line may be 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. [0111] 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
[0112] 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
[0113] 32D and 32D-p210 cells are plated into 6 well TC plates at 2.5xlO6 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). In some embodiments, test compounds of the present invention may demonstrate an apoptotic effect on the 32D-p210 cells but not induce apoptosis in the 32D parental cells.
Effect on Cellular Bcr-Abl Autophosphorylation
[0114] 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 2x10 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). In some embodiments, test compounds of the invention may inhibit the proliferation of the Bcr-Abl expressing cells, inhibiting the cellular Bcr-Abl autophosphorylation in a dose-dependent manner.
Effect on proliferation of cells expressing mutant forms of Bcr-Abl
[0115] Compounds of the invention may be 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 may be 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 are determined from the dose response curves obtained as described above.
FGFR-3 (Enzymatic Assay)
[0116] Kinase activity assay with purified FGFR-3 (Upstate) is carried out in a final volume of 10 μL containing 0.25 μg/mL of enzyme in kinase buffer (30 mM Tris-HCl pH7.5, 15 mM MgCl2, 4.5 mM MnCl2, 15 μM Na3VO4 and 50 μg/mL BSA), and substrates (5 μg/mL biotin- poly-EY(Glu, Tyr) (CIS-US, Inc.) and 3μM ATP). Two solutions are made: the first solution of 5 μl contains the FGFR-3 enzyme in kinase buffer was first dispensed into 384-well format ProxiPlate® (Perkin-Elmer) followed by adding 50 nL of compounds dissolved in DMSO, then 5 μl 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 μL of HTRF detection mixture, which contains 30 mM Tris-HCl pH 7.5, 0.5 M KF, 50 mM
ETDA, 0.2 mg/mL BSA, 15 μg/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.). IC50 values are calculated by linear regression analysis of the percentage inhibition of each compound at 12 concentrations (1:3 dilution from 50 μM to 0.28 nM). In this assay, compounds of the invention have an IC50 in the range of 10 nM to 2 μM.
FGFR-3 (Cellular Assay)
[0117] Compounds of the invention are tested for their ability to inhibit transformed Ba/F3- TEL-FGFR3 cells proliferation, which is depended on FGFR-3 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 μL culture medium. Compounds of the invention are dissolved and diluted in dimethylsulf oxide (DMSO). Twelve points 1:3 serial dilutions are made into DMSO to create concentrations gradient ranging typically from 10 mM to 0.05 μM. Cells are added with 50 nL of diluted compounds and incubated for 48 hours in cell culture incubator. 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 0C cell culture incubator, fluorescence signals from reduced AlamarBlue® (Excitation at 530 nm, Emission at 580 nm) are quantified on Analyst GT (Molecular Devices Corp.). IC50 values are calculated by linear regression analysis of the percentage inhibition of each compound at 12 concentrations.
FLT3 and PDGFRβ
[0118] The effects of compounds of the invention on the cellular activity of FLT3 and PDGFRβ may be conducted following identical methods as described above for FGFR3 cellular activity, using Ba/F3-FLT3-ITD and Ba/F3-Tel-PDGFRβ.
[0119] Compounds of the invention may be tested for their ability to inhibit transformed Ba/F3-FLT3-ITD or Ba/F3-Tel-PDGFRβ cells proliferation, which is dependent on FLT3 or PDGFRβ cellular kinase activity. Ba/F3-FLT3-ITD or Ba/F3-Tel-PDGFRβ 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 μL culture medium. Compounds of the invention are dissolved and diluted in dimethylsulfoxide (DMSO). Twelve points 1:3 serial dilutions are made into DMSO to create concentrations gradient ranging typically from 10 mM to 0.05 μM. Cells are added with 50 nL of diluted compounds and incubated for 48 hours in cell culture incubator. 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 0C cell culture incubator, fluorescence signals from reduced AlamarBlue® (Excitation at 530 nm, Emission at 580 nm) are quantified on Analyst GT (Molecular Devices Corp.). IC50 values are calculated by linear regression analysis of the percentage inhibition of each compound at 12 concentrations.
c-Kit
[0120] Compounds of the invention may be tested for inhibition of SCF dependent proliferation using Mo7e cells which endogenously express c-Kit in a 96-well format. Briefly, two-fold serially diluted test compounds (Cmax = 10 μM) are evaluated for their antiproliferative activity on Mo7e cells stimulated with human recombinant SCF. After 48 hour incubation at 37 0C, cell viability is measured by using a MTT colorimetric assay from Promega.
Upstate KinaseProfiler™ - Radio-enzymatic filter binding assay
[0121] Compounds of the invention may be assessed for their ability to inhibit individual members of a panel of kinases (a partial, non-limiting list of kinases includes: AIk, AbI, Aurora- A, B-Raf, Bcr-Abl, BRK, BIk, Bmx, c-Kit, c-Raf, cSRC, CSK, EphB, FLTl, Fms, Fyn, JAK2, KDR, Lck, Lyn, PDGFRα, PDGFRβ, PKCα, p38 (p38 MAP kinase, SAPK2α), SIK, Src, Syk, Tie2 and TrkB kinases). 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, 10x - containing MnCl2 when required), active kinase (0.001-0.01 Units; 2.5 μL), specific or Poly(Glu4-Tyr) peptide (5-500 μM or .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 (3000Ci/mmol)) is added and the reaction is incubated at about 30 0C for about 10 minutes. The reaction mixture is spotted (20 μL) 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 32P incorporation (cpm) to the peptide substrate is quantified with a Beckman scintillation counter. Percentage inhibition is calculated for each reaction.
[0122] Compounds of Formula (1) or (2) in free form or in pharmaceutically acceptable salt form, may exhibit valuable pharmacological properties, for example, as indicated by the in vitro tests described in this application. The IC50 value in those experiments is given as that concentration of the test compound in question that results in a cell count that is 50 % lower than that obtained using the control without inhibitor. In general, compounds of the invention have IC50 values from 1 nM to 10 μM. In some examples, compounds of the invention have IC50 values from 0.01 μM to 5 μM. In other examples, compounds of the invention have IC50 values from 0.01 μM to 1 μM, or more particularly from 1 nM to 1 μM. In yet other examples, compounds of the invention have IC50 values of less than 1 nM or more than 10 μM. Compounds of Formula (1) or (2) may exhibit a percentage inhibition of greater than 50%, or in other embodiments, may exhibit a percentage inhibition greater than about 70%, against one or more of the following kinases at 10 μM: AIk, AbI, Aurora-A, B-Raf, Bcr-Abl, BRK, BIk, Bmx, c-Kit, c-Raf, cSRC, CSK, EphB, FLTl, Fms, Fyn, JAK2, KDR, Lck, Lyn, PDGFRα, PDGFRβ, PKCα, p38 (p38 MAP kinase, SAPK2α), SIK, Src, Syk, Tie2 and TrkB kinases.
[0123] 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

Claims
1. A compound of Formula ( 1) :
Figure imgf000081_0001
or pharmaceutically acceptable salts thereof, wherein:
X is a 5-7 membered aryl or heteroaryl containing N, O or S;
L is NHCO or CONH;
R1 is halo, NR4CONR5R6, NR4CO2R5, NR4(CR2)PNR5R6 or NR4(CR2)PR5;
R2 is halo, or an optionally halogenated C1-6 alkyl;
R3 is halo, an optionally halogenated Ci_6 alkyl, (CR2)qR7 or O(CR2)qR8;
R and R4 are independently H or Ci_6 alkyl;
R5 and R6 are independently H, an optionally halogenated C1-6 alkyl, C2_6 alkenyl or C2_6 alkynyl; Ci_6 alkanol, (CR2)q-R7 or O(CR2)qR8; or R5 and R6 together with N in NR5R6 may form an optionally substituted ring;
R7 and R8 are independently an optionally substituted Q^.η cycloalkyl, 5-7 membered aryl, heterocyclic or heteroaryl; or R8 is H; m is 1-4; and n, p and q are independently 0-4.
2. The compound of claim 1, wherein X is phenyl, thienyl or pyridyl.
3. The compound of claim 1 , wherein R1 is halo, NR4CONR5R6, NR4CO2R5, NR4(CR2)PNR5R6 or NR4(CR2)PR5;
R5 and R6 are independently H, Ci_6 alkyl, Ci_6 alkanol, or (CR2)qR7; R7 is an optionally substituted Cs_7 cycloalkyl pyridyl, phenyl, naphthalenyl, morpholinyl, piperazinyl, pyrimidinyl, imidazolyl, triazolyl, isothiazolyl, isoxazolyl, pyrazolyl or pyrazinyl; and q is 0-1.
4. The compound of claim 1, wherein R2 is Ci_6 alkyl.
5. The compound of claim 1, wherein R3 is halo, an optionally halogenated Ci_6 alkyl, (CR2)qR7, O(CR2)qR8; and
R and R are independently an optionally substituted piperidinyl, piperazinyl or imidazolyl.
6. The compound of claim 1, wherein said compound is of Formula (2):
Figure imgf000082_0001
wherein R1 is halo, NR4CONR5R6, NR4CO2R5, NR4(CR2)PNR5R6 or NR4(CR2)PR5;
R9 is halo or CF3;
R10 is halo, an optionally halogenated Ci_6 alkyl, or OCi_6 alkyl;
R11 is halo, (CR2)qR7 or O(CR2)qR8;
R and R4 are independently H or Ci_6 alkyl;
R and R are independently H, an optionally halogenated Ci_6 alkyl, C2_6 alkenyl or C2_6 alkynyl; Ci_6 alkanol, (CR2)q-R7 or O(CR2)qR8; or R5 and R6 together with N in NR5R6 may form an optionally substituted ring;
R7 and R8 are independently an optionally substituted C3_7 cycloalkyl, 5-7 membered aryl, heterocyclic or heteroaryl; or R8 is H; m and n are independently 0- 1 ; and p and q are independently 0-4.
7. The compound of claim 6, wherein R1 is NR4CONR5R6, NR4CO2R5 or
NR4(CR2)pNR5R6
8. The compound of claim 6, wherein R5 and R6 are independently H, C1-6 alkyl, or (CR2)qR7; and
R7 is an optionally substituted Cs_7 cycloalkyl, pyridyl, phenyl, naphthalenyl, morpholinyl, piperazinyl, pyrimidinyl, imidazolyl, triazolyl, isothiazolyl, isoxazolyl, pyrazolyl or pyrazinyl.
9. The compound of claim 6, wherein R10 is CF3.
10. The compound of claim 6, wherein R11 is halo, (CR2)qR7, O(CR2)qR8 and R7 and R8 are independently an optionally substituted piperidinyl, piperazinyl or imidazolyl.
11. The compound of claim 6, wherein m and n are 0.
12. A pharmaceutical composition comprising a therapeutically effective amount of a compound of claim 1 and a pharmaceutically acceptable carrier.
13. A method for inhibiting a kinase, comprising administering to a system or a subject in need thereof, a therapeutically effective amount of a compound of claim 1, or pharmaceutically acceptable salts or pharmaceutical compositions thereof, thereby inhibiting said kinase.
14. The method of claim 13, wherein said kinase is AIk, AbI, Aurora-A, B-Raf, Bcr- AbI, BRK, BIk, Bmx, c-Kit, c-Raf, cSRC, CSK, EphB, FLTl, Fms, Fyn, JAK2, KDR, Lck, Lyn, PDGFRα, PDGFRβ, PKCα, p38 (p38 MAP kinase, SAPK2α), SIK, Src, Syk, Tie2 and TrkB kinases, or a combination thereof.
15. The method of claim 13, wherein said kinase is B-Raf, Lck or a combination thereof.
16. The method of claim 13, comprising administering said compound to a cell or tissue system; or to a human or animal subject.
17. A method for treating a B-Raf or Lck-mediated condition, comprising administering to a system or subject in need of such treatment an effective amount of a compound of claim 1 , or pharmaceutically acceptable salts or pharmaceutical compositions thereof, thereby treating said condition.
18. The method of claim 17, comprising administering said compound to a cell or tissue system; or to a human or animal subject.
19. The method of claim 17, wherein said condition is lymphoma, osteosarcoma, melanoma, or a tumor of breast, renal, prostate, colorectal, thyroid, ovarian, pancreatic, neuronal, lung, uterine or gastrointestinal tumor.
20. A method for treating a cell-proliferative condition, comprising administering to a system or subject in need of such treatment an effective amount of a compound of claim 1, or pharmaceutically acceptable salts or pharmaceutical compositions thereof; wherein said cell- proliferative condition is lymphoma, osteosarcoma, melanoma, or a tumor of breast, renal, prostate, colorectal, thyroid, ovarian, pancreatic, neuronal, lung, uterine or gastrointestinal tumor.
21. The use of a compound of any one of claims 1-11, or pharmaceutically acceptable salts or pharmaceutical compositions thereof, for treating a protein kinase-mediated condition.
22. The use of a compound of any one of claims 1- 11 , or pharmaceutically acceptable salts or pharmaceutical compositions thereof, for the manufacture of a medicament for treating a condition mediated by a protein kinase.
23. The use of claims 21 or 22, wherein the protein kinase is B-Raf, Lck or a combination thereof.
24. The use of claim 21 or 22, wherein said condition is lymphoma, osteosarcoma, melanoma, or a tumor of breast, renal, prostate, colorectal, thyroid, ovarian, pancreatic, neuronal, lung, uterine or gastrointestinal tumor.
25. The use of a compound of any one of claims 1- 11 , or pharmaceutically acceptable salts or pharmaceutical compositions thereof, for the manufacture of a medicament for treating a cell-proliferative condition, wherein said cell-proliferative condition is lymphoma, osteosarcoma, melanoma, or a tumor of breast, renal, prostate, colorectal, thyroid, ovarian, pancreatic, neuronal, lung, uterine or gastrointestinal tumor.
PCT/US2008/059024 2007-04-04 2008-04-01 Benzothiazole derivatives and their use as protein kinase inhibitors WO2008124393A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US91018507P 2007-04-04 2007-04-04
US60/910,185 2007-04-04

Publications (1)

Publication Number Publication Date
WO2008124393A1 true WO2008124393A1 (en) 2008-10-16

Family

ID=39494987

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/059024 WO2008124393A1 (en) 2007-04-04 2008-04-01 Benzothiazole derivatives and their use as protein kinase inhibitors

Country Status (1)

Country Link
WO (1) WO2008124393A1 (en)

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101701018B (en) * 2009-11-05 2012-06-13 东南大学 2-(4-aminoquinazoline) benzo [d] thiazole derivative and application thereof
JP2012520845A (en) * 2009-03-17 2012-09-10 グラクソ グループ リミテッド Pyrimidine derivatives used as Itk inhibitors
JP2012522734A (en) * 2009-04-02 2012-09-27 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツング Autotaxin inhibitor
US8338452B2 (en) 2008-02-29 2012-12-25 Array Biopharma Inc. Raf inhibitor compounds and methods of use thereof
US20130040949A1 (en) * 2009-12-29 2013-02-14 Dana-Farber Cancer Institute, Inc. Type ii raf kinase inhibitors
US8394795B2 (en) 2008-02-29 2013-03-12 Array Biopharma Inc. Pyrazole [3, 4-B] pyridine Raf inhibitors
CN103058954A (en) * 2013-01-07 2013-04-24 盛世泰科生物医药技术(苏州)有限公司 Preparation method of [6-(5-amino-2-methyl-phenyl carbamoyloxy-benzothiazol]-tert-butyl carbamate
US8829195B2 (en) 2012-05-15 2014-09-09 Novartis Ag Compounds and compositions for inhibiting the activity of ABL1, ABL2 and BCR-ABL1
US9278981B2 (en) 2012-05-15 2016-03-08 Novartis Ag Compounds and compositions for inhibiting the activity of ABL1, ABL2 and BCR-ABL1
US9315489B2 (en) 2012-05-15 2016-04-19 Novartis Ag Compounds and compositions for inhibiting the activity of ABL1, ABL2 and BCR-ABL1
US9340537B2 (en) 2012-05-15 2016-05-17 Novatis Ag Benzamide derivatives for inhibiting the activity of ABL1, ABL2 and BCR-ABL1
US9382239B2 (en) 2011-11-17 2016-07-05 Dana-Farber Cancer Institute, Inc. Inhibitors of c-Jun-N-terminal kinase (JNK)
US9505784B2 (en) 2009-06-12 2016-11-29 Dana-Farber Cancer Institute, Inc. Fused 2-aminothiazole compounds
US9758522B2 (en) 2012-10-19 2017-09-12 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged small molecules as inducers of protein degradation
WO2017167182A1 (en) * 2016-04-01 2017-10-05 合肥中科普瑞昇生物医药科技有限公司 Selective c-kit kinase inhibitor
US9862688B2 (en) 2014-04-23 2018-01-09 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged janus kinase inhibitors and uses thereof
US10000483B2 (en) 2012-10-19 2018-06-19 Dana-Farber Cancer Institute, Inc. Bone marrow on X chromosome kinase (BMX) inhibitors and uses thereof
US10017477B2 (en) 2014-04-23 2018-07-10 Dana-Farber Cancer Institute, Inc. Janus kinase inhibitors and uses thereof
US10112927B2 (en) 2012-10-18 2018-10-30 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US10550121B2 (en) 2015-03-27 2020-02-04 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
US10702527B2 (en) 2015-06-12 2020-07-07 Dana-Farber Cancer Institute, Inc. Combination therapy of transcription inhibitors and kinase 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)
US11040957B2 (en) 2013-10-18 2021-06-22 Dana-Farber Cancer Institute, Inc. Heteroaromatic compounds useful for the treatment of proliferative diseases
US11142507B2 (en) 2015-09-09 2021-10-12 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
US11447480B2 (en) 2018-05-09 2022-09-20 Lg Chem, Ltd. Compound exhibiting enteropeptidase inhibitory activity

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999024035A1 (en) * 1997-11-10 1999-05-20 Bristol-Myers Squibb Company Benzothiazole protein tyrosine kinase inhibitors

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999024035A1 (en) * 1997-11-10 1999-05-20 Bristol-Myers Squibb Company Benzothiazole protein tyrosine kinase inhibitors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
HUANG ET AL: "Discovery of 2-amino-6-carboxamidobenzothiazoles as potent Lck inhibitors", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, OXFORD, GB, vol. 18, no. 7, 6 March 2008 (2008-03-06), pages 2324 - 2328, XP022574955, ISSN: 0960-894X *

Cited By (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8394795B2 (en) 2008-02-29 2013-03-12 Array Biopharma Inc. Pyrazole [3, 4-B] pyridine Raf inhibitors
US8338452B2 (en) 2008-02-29 2012-12-25 Array Biopharma Inc. Raf inhibitor compounds and methods of use thereof
JP2012520845A (en) * 2009-03-17 2012-09-10 グラクソ グループ リミテッド Pyrimidine derivatives used as Itk inhibitors
JP2012522734A (en) * 2009-04-02 2012-09-27 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツング Autotaxin inhibitor
US9505784B2 (en) 2009-06-12 2016-11-29 Dana-Farber Cancer Institute, Inc. Fused 2-aminothiazole compounds
CN101701018B (en) * 2009-11-05 2012-06-13 东南大学 2-(4-aminoquinazoline) benzo [d] thiazole derivative and application thereof
US11826365B2 (en) 2009-12-29 2023-11-28 Dana-Farber Cancer Institute, Inc. Type II raf kinase inhibitors
US9180127B2 (en) * 2009-12-29 2015-11-10 Dana-Farber Cancer Institute, Inc. Type II Raf kinase inhibitors
US20130040949A1 (en) * 2009-12-29 2013-02-14 Dana-Farber Cancer Institute, Inc. Type ii raf kinase inhibitors
US9358231B2 (en) 2009-12-29 2016-06-07 Dana-Farber Cancer Institute, Inc. Type II RAF kinase inhibitors
US9382239B2 (en) 2011-11-17 2016-07-05 Dana-Farber Cancer Institute, Inc. Inhibitors of c-Jun-N-terminal kinase (JNK)
US10144730B2 (en) 2011-11-17 2018-12-04 Dana-Farber Cancer Institute, Inc. Inhibitors of c-Jun-N-terminal kinase (JNK)
US10981903B2 (en) 2011-11-17 2021-04-20 Dana-Farber Cancer Institute, Inc. Inhibitors of c-Jun-N-terminal kinase (JNK)
US9278981B2 (en) 2012-05-15 2016-03-08 Novartis Ag Compounds and compositions for inhibiting the activity of ABL1, ABL2 and BCR-ABL1
US9458112B2 (en) 2012-05-15 2016-10-04 Novartis Ag Compounds and compositions for inhibiting the activity of ABL1, ABL2 and BCR-ABL1
US9340537B2 (en) 2012-05-15 2016-05-17 Novatis Ag Benzamide derivatives for inhibiting the activity of ABL1, ABL2 and BCR-ABL1
US9315489B2 (en) 2012-05-15 2016-04-19 Novartis Ag Compounds and compositions for inhibiting the activity of ABL1, ABL2 and BCR-ABL1
US8829195B2 (en) 2012-05-15 2014-09-09 Novartis Ag Compounds and compositions for inhibiting the activity of ABL1, ABL2 and BCR-ABL1
US9896444B2 (en) 2012-05-15 2018-02-20 Novartis Ag Benzamide derivatives for inhibiting the activity of ABL1, ABL2 and BCR-ABL1
US10112927B2 (en) 2012-10-18 2018-10-30 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US10787436B2 (en) 2012-10-18 2020-09-29 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
USRE48175E1 (en) 2012-10-19 2020-08-25 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged small molecules as inducers of protein degradation
US10000483B2 (en) 2012-10-19 2018-06-19 Dana-Farber Cancer Institute, Inc. Bone marrow on X chromosome kinase (BMX) inhibitors and uses thereof
US9758522B2 (en) 2012-10-19 2017-09-12 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged small molecules as inducers of protein degradation
CN103058954A (en) * 2013-01-07 2013-04-24 盛世泰科生物医药技术(苏州)有限公司 Preparation method of [6-(5-amino-2-methyl-phenyl carbamoyloxy-benzothiazol]-tert-butyl carbamate
US11040957B2 (en) 2013-10-18 2021-06-22 Dana-Farber Cancer Institute, Inc. Heteroaromatic compounds useful for the treatment of proliferative diseases
US10906889B2 (en) 2013-10-18 2021-02-02 Dana-Farber Cancer Institute, Inc. Polycyclic inhibitors of cyclin-dependent kinase 7 (CDK7)
US10017477B2 (en) 2014-04-23 2018-07-10 Dana-Farber Cancer Institute, Inc. Janus kinase inhibitors and uses thereof
US9862688B2 (en) 2014-04-23 2018-01-09 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged janus kinase inhibitors and uses thereof
US10870651B2 (en) 2014-12-23 2020-12-22 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US11325910B2 (en) 2015-03-27 2022-05-10 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
US10550121B2 (en) 2015-03-27 2020-02-04 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
US10702527B2 (en) 2015-06-12 2020-07-07 Dana-Farber Cancer Institute, Inc. Combination therapy of transcription inhibitors and kinase inhibitors
US11142507B2 (en) 2015-09-09 2021-10-12 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
US10793543B2 (en) 2016-04-01 2020-10-06 Precedo Pharmaceuticals Co., Ltd Selective C-KIT kinase inhibitor
WO2017167182A1 (en) * 2016-04-01 2017-10-05 合肥中科普瑞昇生物医药科技有限公司 Selective c-kit kinase inhibitor
CN107286077B (en) * 2016-04-01 2021-04-02 合肥中科普瑞昇生物医药科技有限公司 Selective C-KIT kinase inhibitor
CN107286077A (en) * 2016-04-01 2017-10-24 合肥中科普瑞昇生物医药科技有限公司 A kind of selective C-KIT kinase inhibitors
US11447480B2 (en) 2018-05-09 2022-09-20 Lg Chem, Ltd. Compound exhibiting enteropeptidase inhibitory activity

Similar Documents

Publication Publication Date Title
WO2008124393A1 (en) Benzothiazole derivatives and their use as protein kinase inhibitors
AU2008265843B2 (en) Protein kinase inhibitors and methods for using thereof
WO2008112695A2 (en) Pyrazolo [3,4-d] pyrimidines and 1, 2, 5, 6-tetraaza- as- indacenes as protein kinase inhibitors for cancer treatment
US20100184765A1 (en) Protein Kinase Inhibitors and Methods for Using Thereof
WO2008144253A1 (en) Protein kinase inhibitors and methods for using thereof
EP2044036B1 (en) [4,5']bipyrimidinyl-6,4'-diamine derivatives as protein kinase inhbitors
RU2368602C2 (en) Compounds and compositions as protein kinase inhibitors
CA2583737C (en) Compounds and compositions as protein kinase inhibitors
AU2008289135B2 (en) 2-heteroarylamino-pyrimidine derivatives as kinase inhibitors
US7678792B2 (en) Compositions and methods for modulating c-kit and PDGFR receptors
AU2008289037B2 (en) 5- (4- (haloalkoxy) phenyl) pyrimidine-2-amine compounds and compositions as kinase inhibitors
US20100087464A1 (en) Protein kinase inhibitors and methods for using thereof
WO2006124731A2 (en) Compounds and compositions as protein kinase inhibitors
WO2007056151A2 (en) Protein kinase inhbitors
KR20080033526A (en) Compounds and compositions as protein kinase inhibitors
MXPA06015147A (en) Compounds and compositions as protein kinase inhibitors.
KR20070030848A (en) Compounds and compositions as protein kinase inhibitors

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 08744862

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08744862

Country of ref document: EP

Kind code of ref document: A1