US20110092538A1 - Compounds and methods for kinase modulation, and indications therefor - Google Patents

Compounds and methods for kinase modulation, and indications therefor Download PDF

Info

Publication number
US20110092538A1
US20110092538A1 US12/981,427 US98142710A US2011092538A1 US 20110092538 A1 US20110092538 A1 US 20110092538A1 US 98142710 A US98142710 A US 98142710A US 2011092538 A1 US2011092538 A1 US 2011092538A1
Authority
US
United States
Prior art keywords
lower alkyl
optionally substituted
pyrrolo
pyridine
fluoro
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/981,427
Inventor
Wayne Spevak
Hanna Cho
Prabha N. Ibrahim
Shenghua Shi
Shumeye Mamo
Samuel J. Gillette
Hongyao Zhu
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Plexxikon Inc
Original Assignee
Plexxikon Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=39281754&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20110092538(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Plexxikon Inc filed Critical Plexxikon Inc
Priority to US12/981,427 priority Critical patent/US20110092538A1/en
Publication of US20110092538A1 publication Critical patent/US20110092538A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/10Drugs for genital or sexual disorders; Contraceptives for impotence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/12Drugs for disorders of the metabolism for electrolyte homeostasis
    • A61P3/14Drugs for disorders of the metabolism for electrolyte homeostasis for calcium homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • the present invention relates to kinases and compounds which modulate kinases, and uses therefor.
  • Particular embodiments contemplate disease indications which are amenable to treatment by modulation of kinase activity by the compounds of the present invention.
  • Receptor protein kinases regulate key signal transduction cascades that control or are involved in the control of a plethora of physiological functions including cellular growth and proliferation, cell differentiation, cellular development, cell division, cell adhesion, stress response, short-range contact-mediated axonal guidance, transcription regulation, aberrant mitogenesis, angiogenesis, abnormal endothelial cell-cell or cell-matrix interactions during vascular development, inflammation, lymphohematopoietic stem cell activity, protective immunity against specific bacteria, allergic asthma, aberrant tissue-specific responses to the activation of the JNK signal transduction pathway, cell transformation, memory, apoptosis, competitive activity-dependent synapse modification at the neuromuscular synapse, immunological mediation of disease, and calcium regulation.
  • Specific disease states associated with aberrant regulation of protein kinases include, for example without limitation, acrocephalo-syndactyl type I, acute myeloid leukemia, AIDS-induced non-Hodgkin's lymphoma, Alzheimer's disease, amyotrophic lateral sclerosis, arthritis, asthma, atherosclerosis, atopic dermatitis, autoimmune diseases, bacterial infection, bladder cancer, cancer of the breast, cancer of the central nervous system, cancer of the colon, cancer of the endometrium, cancer of the fallopian tube, cancer of the gastrointestinal tract, cancer of the ovary, heart failure, chronic myeloid leukemia, colon carcinoma, colorectal cancer, chronic obstructive pulmonary disease (COPD), Crouzon Syndrome, diabetes, diabetic nephropathy, emphysema, endometriosis, epidermoid cancer, fibrotic disorders, gastrointestinal stromal tumor (GIST), glomerulonephritis, Graves' disease, head
  • Compounds are contemplated that are active on protein kinases in general, including, but not limited to, Abl, Akt1, Akt2, Akt3, ALK, Alk5, A-Raf, B-Raf, Brk, Btk, Cdk2, CDK4, CDK5, CDK6, CHK1, c-Raf-1, Csk, EGFR, EphA1, EphA2, EphB2, EphB4, Erk2, Fak, FGFR1, FGFR2, FGFR3, FGFR4, Flt1, Flt3, Flt4, Fms, Frk, Fyn, Gsk3a, Gsk3 ⁇ , HCK, Her2/Erbb2, Her4/Erbb4, IGF1R, IKK beta, Irak4, Itk, Jak1, Jak2, Jak3, Jnk1, Jnk2, Jnk3, Kdr, Kit, Lck, Lyn, MAP2K1, MAP2K2, MAP4K4, MAPKAPK2, Met, Mnk
  • the compounds are active on protein kinases including A-Raf, B-Raf and/or c-Raf-1, including any mutations thereof.
  • compounds are of Formula I, Formula II, or Formula III, as described below.
  • Also contemplated in accordance with the present invention are methods for the use of the above-described compounds in treating diseases and conditions associated with regulation of the activity of the above-described kinases.
  • the use of compounds for therapeutic methods involving modulation of protein kinases are provided, as well as compounds that can be used for therapeutic methods involving modulation of protein kinases.
  • compounds have the structure according to the following Formula I:
  • R 1 is selected from the group consisting of hydrogen, halogen, optionally substituted lower alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, —CN, —OR 4 , —SR 4 , —NR 5 R 4 , —C(O)R 4 , —C(O)NR 5 R 4 , —S(O) 2 NR 5 R 4 , —NR 5 C(O)R 4 , —NR 5 S(O) 2 R 4 , —S(O)R 4 , and —S(O) 2 R 4 , and R 2 is selected from the group consisting of halogen, optionally substituted lower alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, —CN, —OR 4 , —SR 4 , —NR 5 R 4
  • R 1 is selected from the group consisting of hydrogen, halogen, lower alkyl, lower alkenyl, lower alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, —OH, —NH 2 , —CN, —NO 2 , —C(O)OH, —S(O) 2 NH 2 , —C(O)NH 2 , —C(S)NH 2 , —NHC(O)NH 2 , —NHC(S)NH 2 , —NHS(O) 2 NH 2 , —OR 7 , —SR 7 , —NR 8 R 7 , —C(O)R 7 , —C(S)R 7 , —C(O)OR 7 , —C(O)NR 8 R 7 , —C(S)NR 8 R 7 , —S(O) 2 NR 8 R 7 , —NR 8 C(
  • R 1 is selected from the group consisting of hydrogen, —CN, —OR 7 , —SR 7 , —NR 8 R 7 , —NR 8 C(O)R 7 , —NR 8 S(O) 2 R 7 , —C(O)NR 8 R 7 , —C(O)R 7 , —S(O) 2 NR 8 R 7 , —S(O)R 7 , —S(O) 2 R 7 , halogen, lower alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, aryl
  • R 1 is hydrogen, —CN, —NR 8 R 7 , —OR 7 , —S(O) 2 R 7 , fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, cycloalkyl, heterocycloalkyl, aryl or heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl or heteroaryl are optionally substituted with one or more substituents selected from the group consisting of halogen, lower alkyl, fluoro substituted lower alkyl, —NR 8 R 7 , —OR 7 and —S(O) 2 R 7 , and R 2 is —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, —NR 8 R 7 , —OR 7 or —S(O) 2 R 7 ; wherein R 7 and R 8 are as defined in paragraph [0011].
  • R 1 is hydrogen, —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower alkoxy substituted C 2-6 alkoxy, aryl or heteroaryl, wherein aryl or heteroaryl are optionally substituted with one or more substituents selected from the group consisting of fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, mono-alkylamino, di-alkylamino, and cycloalkylamino
  • R 2 is —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, mono-alkylamino, di-alkylamino, or cycloalkylamino.
  • the compound is selected from the group consisting of:
  • compounds have the structure according to the following Formula II:
  • R 1 is selected from the group consisting of hydrogen, halogen, optionally substituted lower alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, —CN, —OR 4 , —SR 4 , —NR 5 R 4 , —C(O)R 4 , —C(O)NR 5 R 4 , —S(O) 2 NR 5 R 4 , —NR 5 C(O)R 4 , —NR 5 S(O) 2 R 4 , —S(O)R 4 , and —S(O) 2 R 4 , and R 2a is selected from the group consisting of halogen, optionally substituted lower alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, —CN, —OR 4 , —SR 4 , —NR 5 R
  • R 1 is selected from the group consisting of hydrogen, halogen, lower alkyl, lower alkenyl, lower alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, —OH, —NH 2 , —CN, —NO 2 , —C(O)OH, —S(O) 2 NH 2 , —C(O)NH 2 , —C(S)NH 2 , —NHC(O)NH 2 , —NHC(S)NH 2 , —NHS(O) 2 NH 2 , —OR 7 , —SR 7 , —NR 8 R 7 , —C(O)R 7 , —C(S)R 7 , —C(O)OR 7 , —C(O)NR 8 R 7 , —C(S)NR 8 R 7 , —S(O) 2 NR 8 R 7 , —NR 8 C(
  • R 1 is selected from the group consisting of hydrogen, —CN, —OR 7 , —SR 7 , —NR 8 R 7 , —NR 8 C(O)R 7 , —NR 8 S(O) 2 R 7 , —C(O)NR 8 R 7 , —C(O)R 7 , —S(O) 2 NR 8 R 7 , —S(O)R 7 , —S(O) 2 R 7 , halogen, lower alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, aryl
  • R 1 is hydrogen, —CN, —NR 8 R 7 , —OR 7 , —S(O) 2 R 7 , fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, cycloalkyl, heterocycloalkyl, aryl or heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl or heteroaryl are optionally substituted with one or more substituents selected from the group consisting of halogen, lower alkyl, fluoro substituted lower alkyl, —NR 8 R 7 , —OR 7 and —S(O) 2 R 7 , and R 2a is —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, —NR 8 R 7 , —OR 7 or —S(O) 2 R 7 ; wherein R 7 and R 8 are as defined in paragraph [0011].
  • R 1 is hydrogen, —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower alkoxy substituted C 2-6 alkoxy, aryl or heteroaryl, wherein aryl or heteroaryl are optionally substituted with one or more substituents selected from the group consisting of fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, mono-alkylamino, di-alkylamino, and cycloalkylamino, and R 2a is —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, mono-alkylamino, di-alkylamino, or cycloalkylamino.
  • the compound is selected from the group consisting of:
  • compounds have the structure according to the following Formula III:
  • R 1 is selected from the group consisting of hydrogen, halogen, optionally substituted lower alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, —CN, —OR 4 , —SR 4 , —NR 5 R 4 , —C(O)R 4 , —C(O)NR 5 R 4 , —S(O) 2 NR 5 R 4 , —NR 5 C(O)R 4 , —NR 5 S(O) 2 R 4 , —S(O)R 4 , and —S(O) 2 R 4 , and R 6 is heteroaryl optionally substituted with one or more substituents selected from the group consisting of halogen, optionally substituted lower alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl
  • R 1 is selected from the group consisting of hydrogen, halogen, lower alkyl, lower alkenyl, lower alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, —OH, —NH 2 , —CN, —NO 2 , —C(O)OH, —S(O) 2 NH 2 , —C(O)NH 2 , —C(S)NH 2 , —NHC(O)NH 2 , —NHC(S)NH 2 , —NHS(O) 2 NH 2 , —OR 7 , —SR 7 , —NR 8 R 7 , —C(O)R 7 , —C(S)R 7 , —C(O)OR 7 , —C(O)NR 8 R 7 , —C(S)NR 8 R 7 , —S(O) 2 NR 8 R 7 , —NR 8 C(
  • R 1 is selected from the group consisting of hydrogen, —CN, —OR 7 , —SR 7 , —NR 8 R 7 , —NR 8 C(O)R 7 , —NR 8 S(O) 2 R 7 , —C(O)NR 8 R 7 , —C(O)R 7 , —S(O) 2 NR 8 R 7 , —S(O)R 7 , —S(O) 2 R 7 , halogen, lower alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, aryl
  • R 1 is hydrogen, —CN, —NR 8 R 7 , —OR 7 , —S(O) 2 R 7 , fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, cycloalkyl, heterocycloalkyl, aryl or heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl or heteroaryl are optionally substituted with one or more substituents selected from the group consisting of halogen, lower alkyl, fluoro substituted lower alkyl, —NR 8 R 7 , —OR 7 and —S(O) 2 R 7 , and R 6 is heteroaryl optionally substituted with one or more of —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, —NR 8 R 7 , —OR 7 or —S(O) 2 R 7 ; wherein R 7 and R 8 are as defined in paragraph [0011].
  • R 1 is hydrogen, —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower alkoxy substituted C 2-6 alkoxy, aryl or heteroaryl, wherein aryl or heteroaryl are optionally substituted with one or more substituents selected from the group consisting of fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, mono-alkylamino, di-alkylamino, and cycloalkylamino, and R 6 is heteroaryl optionally substituted with one or more of —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, mono-alkylamino, di-alkylamino, or cycloalkylamino.
  • the compound is selected from the group consisting of:
  • compounds are excluded where N (except where N is a heteroaryl ring atom), O, or S is bound to a carbon that is also bound to N (except where N is a heteroaryl ring atom), O, or S; or where N (except where N is a heteroaryl ring atom), O, C(S), C(O), or S(O) n (n is 0-2) is bound to an alkene carbon of an alkenyl group or bound to an alkyne carbon of an alkynyl group; accordingly, in some embodiments compounds which include linkages such as the following are excluded from the compounds provided: —NR—CH 2 —NR—, —O—CH 2 —NR—, —S—CH 2 —NR—, —NR—CH 2 —O—, —O—CH 2 —O—, —S—CH 2 —O—, —NR—CH 2 —S—, —O—CH 2 —S—, —S—CH 2 —, —S—CH
  • methods for treating a protein kinase mediated disease or condition in an animal subject, wherein the method involves administering to the subject an effective amount of one or more compounds of Formula I, Formula II, or Formula III.
  • treat refers to the administration of material, e.g., one or more compound of Formula I, Formula II, or Formula III, in an amount effective to prevent, alleviate, or ameliorate one or more symptoms of a disease or condition, i.e., indication, and/or to prolong the survival of the subject being treated.
  • protein kinase mediated disease or condition refers to a disease or condition in which the biological function of a protein kinase affects the development, course, and/or symptoms of the disease or condition, and/or in which modulation of the protein kinase alters the development, course, and/or symptoms of the disease or condition.
  • a protein kinase mediated disease or condition includes a disease or condition for which modulation provides a therapeutic benefit, e.g. wherein treatment with protein kinase inhibitors, including compounds described herein, provides a therapeutic benefit to the subject suffering from or at risk of the disease or condition.
  • the method involves administering to the subject an effective amount of a compound of Formula I, Formula II, or Formula III in combination with one or more other therapies for the disease or condition.
  • methods for treating a protein kinase mediated disease or condition in an animal subject, wherein the method involves administering to the subject an effective amount of any one or more compound of Formula I, Formula II, or Formula III.
  • the invention provides methods for treating a Raf protein kinase mediated disease or condition in an animal subject, wherein the method involves administering to the subject an effective amount of one or more compound of Formula I, Formula II, or Formula III.
  • the terms “Raf protein kinase mediated disease or condition,” “Raf mediated disease or condition,” and the like refer to a disease or condition in which the biological function of a Raf kinase, including any mutations thereof, affects the development, course, and/or symptoms of the disease or condition, and/or in which modulation of the Raf protein kinase alters the development, course, and/or symptoms of the disease or condition.
  • the Raf protein kinase includes, but is not limited to, A-Raf, A-Raf mutations, B-Raf, mutations of B-Raf, c-Raf-1 and mutations of c-Raf-1.
  • the Raf protein kinase is B-Raf mutation V600E.
  • the Raf protein kinase is B-Raf mutation V600E/T5291.
  • the disease or condition is a cancer that is amenable to treatment by an inhibitor of the V600E mutant B-Raf.
  • the disease or condition is a cancer that is amenable to treatment by an inhibitor of the V600E/T5291 mutant B-Raf.
  • the Raf protein kinase mediated disease or condition includes a disease or condition for which Raf inhibition provides a therapeutic benefit, e.g. wherein treatment with Raf inhibitors, including compounds described herein, provides a therapeutic benefit to the subject suffering from or at risk of the disease or condition.
  • the method involves administering to the subject an effective amount of a compound of Formula I, Formula II, or Formula III in combination with one or more other therapies for the disease or condition.
  • A-Raf, B-Raf or c-Raf-1 protein kinase mediated disease or condition refers to a disease or condition in which the biological function of an A-Raf, B-Raf or c-Raf-1 kinase, respectively, including any mutations thereof, affects the development, course and/or symptoms of the disease or condition, and/or in which modulation of the A-Raf, B-Raf or c-Raf-1 protein kinase, respectively, alters the development, course, and/or symptoms of the disease or condition.
  • a compound of Formula I, Formula II, or Formula III will have an IC 50 of less than 500 nm, less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 1 nM as determined in a generally accepted kinase activity assay.
  • a compound of any of Formula I, Formula II, or Formula III will have an IC 50 of less than 500 nm, less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 1 nM with respect to at least one kinase selected from the group consisting of Abl, Akt1, Akt2, Akt3, ALK, Alk5, A-Raf, B-Raf, Brk, Btk, Cdk2, CDK4, CDK5, CDK6, CHK1, c-Raf-1, Csk, EGFR, EphA1, EphA2, EphB2, EphB4, Erk2, Fak, FGFR1, FGFR2, FGFR3, FGFR4, Flt1, Flt3, Flt4, Fms, Frk, Fyn, Gsk3 ⁇ , Gsk3 ⁇ , HCK, Her2/Erbb2, Her4/Erbb4, IGF1R, IGF1R,
  • a compound of Formula I, Formula II, or Formula III will have an IC 50 of less than 500 nm, less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 1 nM with respect to at least one kinase selected from the group consisting of Abl, Akt 1, Akt2, Akt3, ALK, Alk5, A-Raf, B-Raf, Btk, Cdk2, CDK4, CDK5, CDK6, CHK1, c-Raf-1, Csk, EGFR, EphA1, EphA2, EphB2, EphB4, Erk2, Fak, Fms, Fyn, Gsk3 ⁇ , Gsk3 ⁇ , HCK, Her2/Erbb2, Her4/Erbb4, IGF1R, IKK beta, Irak4, Itk, Jak1, Jak2, Jak3, Jnk1, Jnk2, Jnk3, Kit, L
  • a compound of Formula I, Formula II, or Formula III will have an IC 50 of less than 500 nm, less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 1 nM with respect to at least one kinase selected from the group consisting of Abl, A-Raf, B-Raf, Btk, c-Raf-1, EGFR, EphB2, Erk2, Fak, FGFR1, Flt1, Flt3, Flt4, Fms, Irak4, Jnk1, Jnk2, Jnk3, Kdr, Kit, Lck, Lyn, MAP2K1, MAPKAP kinase 2, Met, p38, PDGFRB, Pim1, PKC theta, Pyk2, Ret, Src, Stk6, Yes, and Zap70, including any mutations thereof.
  • kinase selected from the group consisting of Abl, A-Raf
  • a compound of Formula I, Formula II, or Formula III will have an IC 50 of less than 500 nm, less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 1 nM with respect to at least one kinase selected from the group consisting of Abl, A-Raf, B-Raf, Btk, c-Raf-1, EGFR, EphB2, Erk2, Fak, Fms, Irak4, Jnk1, Jnk2, Jnk3, Kit, Lck, Lyn, MAP2K1, MAPKAP kinase 2, Met, p38, Pim1, PKC theta, Pyk2, Src, Stk6, Yes, and Zap70, including any mutations thereof.
  • kinase selected from the group consisting of Abl, A-Raf, B-Raf, Btk, c-Raf-1, EGFR, Eph
  • a compound of Formula I, Formula II, or Formula III will have an IC 50 of less than 500 nm, less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 1 nM with respect to at least one kinase selected from the group consisting of A-Raf, B-Raf, B-Raf V600E mutant, B-Raf V600E/T529I mutant, c-Raf-1, Fak, FGFR1, FGFR2, FGFR3, FGFR4, Jnk1, Jnk2, Jnk3, Lck, Lyn, Met, Pim1, Pim2, Pim3, Pyk2, Kdr, Src and Ret, including any mutations thereof.
  • a compound of Formula I, Formula II, or Formula III is an inhibitor of a Raf kinase and has an IC 50 of less than 500 nm, less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 1 nM as determined in a generally accepted Raf kinase activity assay.
  • a compound of Formula I, Formula II, or Formula III will have an IC 50 of less than 500 nm, less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 1 nM with respect to B-Raf, c-Raf-1, B-Raf V600E mutant, or B-Raf V600E/T529I mutant.
  • a compound of Formula I, Formula II, or Formula III will selectively inhibit one Raf kinase relative to one or more other Raf kinases.
  • the compound of Formula I, Formula II, or Formula III will selectively inhibit a mutation of the Raf kinase relative to the wild type kinase, for example B-Raf V600E mutant relative to wild type B-Raf.
  • a compound will also inhibit the effects of a mutation of the kinase, including, but not limited to, a mutation that is related to a disease state, such as a cancer.
  • a mutation that is related to a disease state such as a cancer.
  • B-Raf V600E mutant is present in a high percentage of some cancers, such as melanoma, and compounds will inhibit the kinase activity of this mutant.
  • a compound may selectively inhibit one kinase relative to one or more other kinases, where preferably inhibition is selective with respect to any of the other kinases, whether a kinase discussed herein, or other kinases.
  • the compound may selectively inhibit the effects of a mutation of the kinase relative to the wild type kinase, for example B-Raf V600E mutant relative to wild type B-Raf.
  • the compound may selectively inhibit Fms relative to Kit.
  • IC 50 for the one kinase may be at least about 2-fold, also 5-fold, also 10-fold, also 20-fold, also 50-fold, or at least about 100-fold less than the IC 50 for any of the other kinases as determined in a generally accepted kinase activity assay.
  • compositions that include a therapeutically effective amount of one or more compound of Formula I, Formula II, or Formula III and at least one pharmaceutically acceptable carrier, excipient, and/or diluent, including combinations of any two or more compounds of Formula I, Formula II or Formula III.
  • the composition can further include a plurality of different pharmacologically active compounds, which can include a plurality of compounds of Formula I, Formula II, or Formula III.
  • the composition can include one or more compounds of Formula I, Formula II, or Formula III along with one or more compounds that are therapeutically effective for the same disease indication.
  • the composition includes one or more compounds of Formula I, Formula II, or Formula III along with one or more compounds that are therapeutically effective for the same disease indication, wherein the compounds have a synergistic effect on the disease indication.
  • the composition includes one or more compounds of Formula I, Formula II, or Formula III effective in treating a cancer and one or more other compounds that are effective in treating the cancer, further wherein the compounds are synergistically effective in treating the cancer.
  • methods for modulating the activity of a protein kinase selected from the group consisting of Abl, Akt1, Akt2, Akt3, ALK, Alk5, A-Raf, B-Raf, Brk, Btk, Cdk2, CDK4, CDK5, CDK6, CHK1, c-Raf-1, Csk, EGFR, EphA1, EphA2, EphB2, EphB4, Erk2, Fak, FGFR1, FGFR2, FGFR3, FGFR4, Flt1, Flt3, Flt4, Fms, Frk, Fyn, Gsk3 ⁇ , Gsk3 ⁇ , HCK, Her2/Erbb2, Her4/Erbb4, IGF1R, IKK beta, Irak4, Itk, Jak1, Jak2, Jak3, Jnk1, Jnk2, Jnk3, Kdr, Kit, Lck, Lyn, MAP2K1, MAP2K2, MAP4K4, MAPKAPK2, Met, Mn
  • methods for treating a protein kinase mediated disease or condition in an animal subject, wherein the method involves administering to the subject an effective amount of a composition including one or more compound of Formula I, Formula II, or Formula III.
  • methods for treating a disease or condition mediated by a protein kinase selected from the group consisting of Abl, Akt 1, Akt2, Akt3, ALK, Alk5, A-Raf, B-Raf, Btk, Cdk2, CDK4, CDK5, CDK6, CHK1, c-Raf-1, Csk, EGFR, EphA1, EphA2, EphB2, EphB4, Erk2, Fak, FGFR1, FGFR2, FGFR3, FGFR4, Flt1, Flt3, Flt4, Fms, Fyn, Gsk3 ⁇ , Gsk3 ⁇ , HCK, Her2/Erbb2, Her4/Erbb4, IGF1R, IKK beta, Irak4, Itk, Jak1, Jak2, Jak3, Jnk1, Jnk2, Jnk3, Kdr, Kit, Lck, Lyn, MAP2K1, MAP2K2, MAP4K4, MAPKAPK2, Met, Mnk1,
  • the invention provides methods for treating a disease or condition mediated by a protein kinase selected from the group consisting of Abl, Akt 1, Akt2, Akt3, ALK, Alk5, A-Raf, B-Raf, Btk, Cdk2, CDK4, CDK5, CDK6, CHK1, c-Raf-1, Csk, EGFR, EphA1, EphA2, EphB2, EphB4, Erk2, Fak, Fms, Fyn, Gsk3 ⁇ , Gsk3 ⁇ , HCK, Her2/Erbb2, Her4/Erbb4, IGF1R, IKK beta, Irak4, Itk, Jak1, Jak2, Jak3, Jnk1, Jnk2, Jnk3, Kit, Lck, Lyn, MAP2K1, MAP2K2, MAP4K4, MAPKAPK2, Met, Mnk1, MLK1, p38, PDPK1, Pim1, Pim2, Pim3, PKC alpha, PKC beta
  • the invention provides methods for treating a disease or condition mediated by a protein kinase selected from the group consisting of Abl, A-Raf, B-Raf, Btk, c-Raf-1, EGFR, EphB2, Erk2, Fak, FGFR1, Flt1, Flt3, Flt4, Fms, Irak4, Jnk1, Jnk2, Jnk3, Kdr, Kit, Lck, Lyn, MAP2K1, MAPKAPK2, Met, p38, PDGFRB, Pim1, PKC theta, Pyk2, Ret, Src, Stk6, Yes, and Zap70 by administering to the subject an effective amount of a composition including one or more compound of Formula I, Formula II, or Formula III.
  • a protein kinase selected from the group consisting of Abl, A-Raf, B-Raf, Btk, c-Raf-1, EGFR, EphB2, Erk2, Fak, FGFR1,
  • the invention provides methods for treating a disease or condition mediated by a protein kinase selected from the group consisting of Abl, A-Raf, B-Raf, Btk, c-Raf-1, EGFR, EphB2, Erk2, Fak, Fms, Irak4, Jnk1, Jnk2, Jnk3, Kit, Lck, Lyn, MAP2K1, MAPKAPK2, Met, p38, Pim1, PKC theta, Pyk2, Src, Stk6, Yes, and Zap70 by administering to the subject an effective amount of a composition including one or more compound of Formula I, Formula II, or Formula III.
  • a protein kinase selected from the group consisting of Abl, A-Raf, B-Raf, Btk, c-Raf-1, EGFR, EphB2, Erk2, Fak, Fms, Irak4, Jnk1, Jnk2, Jnk3, Kit, Lck, Lyn, MAP
  • the invention provides methods for treating a disease or condition mediated by a protein kinase selected from the group consisting of A-Raf, B-Raf, B-Raf V600E mutant, B-Raf V600E/T529I mutant, c-Raf-1, Fak, FGFR1, FGFR2, FGFR3, FGFR4, Jnk1, Jnk2, Jnk3, Lck, Lyn, Met, Pim1, Pim2, Pim3, Pyk2, Kdr, Src and Ret by administering to the subject an effective amount of a composition including one or more compound of Formula I, Formula II, or Formula III.
  • a protein kinase selected from the group consisting of A-Raf, B-Raf, B-Raf V600E mutant, B-Raf V600E/T529I mutant, c-Raf-1, Fak, FGFR1, FGFR2, FGFR3, FGFR4, Jnk1, Jnk2, J
  • the invention provides methods for treating a disease or condition mediated by A-Raf, B-Raf, c-Raf-1, B-Raf V600E mutant, or B-Raf V600E/T529I mutant by administering to the subject an effective amount of a composition including one or more compound of Formula I, Formula II or Formula III.
  • the invention provides methods for treating a disease or condition mediated by A-Raf, B-Raf, c-Raf-1, B-Raf V600E mutant, or B-Raf V600E/T5291 mutant by administering to the subject an effective amount of a composition including one or more compound of Formula I, Formula II, or Formula III in combination with one or more other suitable therapies for treating the disease.
  • the invention provides methods for treating a cancer mediated by B-Raf V600E mutant or B-Raf V600E/T5291 mutant by administering to the subject an effective amount of a composition including one or more compound of Formula I, Formula II, or Formula III in combination with one or more suitable anticancer therapies, such as one or more chemotherapeutic drugs.
  • the invention provides a method of treating a cancer by administering to the subject an effective amount of a composition including one or more compound of Formula I, Formula II, or Formula III, in combination with one or more other therapies or medical procedures effective in treating the cancer.
  • Other therapies or medical procedures include suitable anticancer therapy (e.g. drug therapy, vaccine therapy, gene therapy, photodynamic therapy) or medical procedure (e.g. surgery, radiation treatment, hyperthermia heating, bone marrow or stem cell transplant).
  • the one or more suitable anticancer therapies or medical procedures is selected from treatment with a chemotherapeutic agent (e.g. chemotherapeutic drug), radiation treatment (e.g.
  • x-ray, y-ray, or electron, proton, neutron, or a particle beam hyperthermia heating (e.g. microwave, ultrasound, radiofrequency ablation),
  • Vaccine therapy e.g. AFP gene hepatocellular carcinoma vaccine, AFP adenoviral vector vaccine, AG-858, allogeneic GM-CSF-secretion breast cancer vaccine, dendritic cell peptide vaccines
  • gene therapy e.g. Ad5CMV-p53 vector, adenovector encoding MDA7, adenovirus 5-tumor necrosis factor alpha
  • photodynamic therapy e.g. aminolevulinic acid, motexafin lutetium
  • the invention provides a method of treating a cancer by administering to the subject an effective amount of a composition including one or more compound of Formula I, Formula II, or Formula III, in combination with one or more suitable chemotherapeutic agents.
  • the one or more suitable chemotherapeutic agents is selected from an alkylating agent, including, but not limited to, adozelesin, altretamine, bizelesin, busulfan, carboplatin, carboquone, carmustine, chlorambucil, cisplatin, cyclophosphamide, dacarbazine, estramustine, fotemustine, hepsulfam, ifosfamide, improsulfan, irofulven, lomustine, mechlorethamine, melphalan, oxaliplatin, piposulfan, semustine, streptozocin, temozolomide, thiotepa, and treosulfan;
  • the method of treating a cancer involves administering to the subject an effective amount of a composition including one or more compound of Formula I, Formula II, or Formula III in combination with a chemotherapeutic agent selected from 5-fluorouracil, carboplatin, dacarbazine, gefitinib, oxaliplatin, paclitaxel, SN-38, temozolomide, vinblastine, bevacizumab, cetuximab, or erlotinib.
  • a chemotherapeutic agent selected from 5-fluorouracil, carboplatin, dacarbazine, gefitinib, oxaliplatin, paclitaxel, SN-38, temozolomide, vinblastine, bevacizumab, cetuximab, or erlotinib.
  • the invention provides a method of treating or prophylaxis of a disease or condition in a mammal, by administering to the mammal a therapeutically effective amount of one or more compound of Formula I, Formula II, or Formula III, a prodrug of such compound, or a pharmaceutically acceptable salt of such compound or prodrug.
  • the compound can be alone or can be part of a composition.
  • the invention provides a method of treating or prophylaxis of a disease or condition in a mammal, by administering to the mammal a therapeutically effective amount of one or more compound of Formula I, Formula II, or Formula III, a prodrug of such compound, or a pharmaceutically acceptable salt of such compound or prodrug in combination with one or more other suitable therapies for the disease or condition.
  • kits that include a composition as described herein.
  • the composition is packaged, e.g., in a vial, bottle, flask, which may be further packaged, e.g., within a box, envelope, or bag; the composition is approved by the U.S.
  • the composition is approved for administration to a mammal, e.g., a human, for a protein kinase mediated disease or condition;
  • the invention kit includes written instructions for use and/or other indication that the composition is suitable or approved for administration to a mammal, e.g., a human, for a protein kinase-mediated disease or condition; and the composition is packaged in unit dose or single dose form, e.g., single dose pills, capsules, or the like.
  • the disease or condition is, for example without limitation, neurologic diseases, including, but not limited to, cerebrovascular ischemia, multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease (AD), Parkinson's disease, amyotrophic lateral sclerosis, dementia, senile chorea, and Huntington's disease; neoplastic diseases and associated complications, including, but not limited to, chemotherapy-induced hypoxia, gastrointestinal stromal tumors (GISTs), prostate tumors, mast cell tumors (including canine mast cell tumors), acute myeloid myelofibrosis, leukemia, acute lymphocytic leukemia, chronic myeloid leukemia, chronic lymphocytic leukemia, multiple myeloma, melanoma, mastocytosis, gliomas, glioblastoma, astrocytoma, neuroblastoma
  • neurologic diseases including, but not limited to, cerebrovascular ischemia, multi-infarct dementia, head injury,
  • sarcomas of neuroectodermal origin leiomyosarcoma
  • carcinomas e.g. lung, breast, pancreatic, colon, hepatocellular, renal, female genital tract, squamous cell, carcinoma in situ
  • lymphoma e.g. histiocytic lymphoma, non-Hodgkin's lymphoma
  • MEN2 syndromes neurofibromatosis (including Schwann cell neoplasia), myelodysplastic syndrome, leukemia, tumor angiogenesis, cancers of the thyroid, liver, bone, skin, brain, central nervous system, pancreas, lung (e.g.
  • small cell lung cancer non small cell lung cancer
  • pain of neuropathic or inflammatory origin including, but not limited to, acute pain, chronic pain, bone pain, cancer-related pain and migraine
  • cardiovascular diseases including, but not limited to, heart failure, ischemic stroke, cardiac hypertrophy, thrombosis (e.g. thrombotic microangiopathy syndromes), atherosclerosis, reperfusion injury and ischemia (e.g.
  • inflammation including, but not limited to, age-related macular degeneration, rheumatoid arthritis, allergic rhinitis, inflammatory bowel disease (IBD), ulcerative colitis, Crohn's disease, systemic lupus erythematosis, Sjogren's Syndrome, Wegener's granulomatosis, Kawasaki's Disease, hemophagocytic syndrome (macrophage activation syndrome), multicentric reticulohistiocytosis, psoriasis, scleroderma, chronic thyroiditis, Grave's disease, myasthenia gravis, multiple sclerosis, osteoarthritis, endometriosis, scarring (e.g.
  • immunodeficiency diseases including, but not limited to, severe combined immunodeficiency (SCID), organ transplant rejection, and graft versus host disease
  • SCID severe combined immunodeficiency
  • renal or prostatic diseases including, but not limited to, diabetic nephropathy, polycystic kidney disease, nephrosclerosis, glomerulonephritis, interstitial nephritis, Lupus nephritis, prostate hyperplasia, chronic renal failure, tubular necrosis, diabetes-associated renal complications, and renal hypertrophy
  • metabolic diseases including, but not limited to, type 1 diabetes, type 2 diabetes, metabolic syndrome, obesity, hepatic steatosis, insulin resistance, hyperglycemia, lipolysis and obesity
  • infection including, but not limited to, Helicobacter pylor
  • the invention provides methods for treating an A-Raf-mediated, B-Raf-mediated and/or c-Raf-1-mediated disease or condition in an animal subject (e.g. a mammal such as a human, other primates, sports animals, animals of commercial interest such as cattle, farm animals such as horses, or pets such as dogs and cats), e.g., a disease or condition characterized by abnormal A-Raf, B-Raf, and/or c-Raf-1 activity (e.g. kinase activity).
  • an animal subject e.g. a mammal such as a human, other primates, sports animals, animals of commercial interest such as cattle, farm animals such as horses, or pets such as dogs and cats
  • a disease or condition characterized by abnormal A-Raf, B-Raf, and/or c-Raf-1 activity e.g. kinase activity.
  • invention methods involve administering to the subject suffering from or at risk of an A-Raf-mediated, B-Raf-mediated and/or c-Raf-1-mediated disease or condition an effective amount of compound of Formula I, Formula II or Formula III.
  • the A-Raf-mediated, B-Raf-mediated, and/or c-Raf-1-mediated disease is selected from the group consisting of neurologic diseases, including, but not limited to, multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease (AD), Parkinson's disease; neoplastic diseases including, but not limited to, melanoma, glioma, sarcoma, carcinoma (e.g.
  • lymphoma e.g. histiocytic lymphoma
  • neurofibromatosis acute myeloid leukemia, myelodysplastic syndrome, leukemia, tumor angiogenesis, neuroendocrine tumors such as medullary thyroid cancer, carcinoid, small cell lung cancer and pheochromocytoma
  • pain of neuropathic or inflammatory origin including, but not limited to, acute pain, chronic pain, cancer-related pain, and migraine
  • cardiovascular diseases including, but not limited to, heart failure, ischemic stroke, cardiac hypertrophy, thrombosis (e.g.
  • inflammation including, but not limited to, psoriasis, arthritis and autoimmune diseases and conditions, osteoarthritis, endometriosis, scarring, vascular restenosis, fibrotic disorders, rheumatoid arthritis, inflammatory bowel disease (IBD); immunodeficiency diseases, including, but not limited to, organ transplant rejection, graft versus host disease; renal or prostatic diseases, including, but not limited to, diabetic nephropathy, polycystic kidney disease, nephrosclerosis, glomerulonephritis, prostate hyperplasia; metabolic disorders, including, but not limited to, obesity; infection, including, but not limited to Helicobacter pylori , Hepatitis and Influenza viruses, fever, and sepsis; pulmonary diseases including, but not limited to, chronic obstructive pulmonary disease (COPD) and acute respiratory distress syndrome (ARDS); genetic developmental diseases, including, but not limited to, COPD) and chronic respiratory distress syndrome (ARDS); genetic developmental diseases, including,
  • compounds of Formula I, Formula II, or Formula III can be used in the preparation of a medicament for the treatment of an A-Raf-mediated, B-Raf-mediated or c-Raf-1-mediated disease or condition selected from the group consisting of neurologic diseases, including, but not limited to, multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease (AD), Parkinson's disease; neoplastic diseases including, but not limited to, melanoma, glioma, sarcoma, carcinoma (e.g. colorectal, lung, breast, pancreatic, thyroid, renal, ovarian), lymphoma (e.g.
  • A-Raf-mediated, B-Raf-mediated or c-Raf-1-mediated disease or condition selected from the group consisting of neurologic diseases, including, but not limited to, multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease (AD), Parkinson's disease; neoplastic diseases including, but not limited to,
  • histiocytic lymphoma neurofibromatosis, acute myeloid leukemia, myelodysplastic syndrome, leukemia, tumor angiogenesis, neuroendocrine tumors such as medullary thyroid cancer, carcinoid, small cell lung cancer and pheochromocytoma
  • pain of neuropathic or inflammatory origin including, but not limited to, acute pain, chronic pain, cancer-related pain, and migraine
  • cardiovascular diseases including, but not limited to, heart failure, ischemic stroke, cardiac hypertrophy, thrombosis (e.g.
  • inflammation including, but not limited to, psoriasis, arthritis and autoimmune diseases and conditions, osteoarthritis, endometriosis, scarring, vascular restenosis, fibrotic disorders, rheumatoid arthritis, inflammatory bowel disease (IBD); immunodeficiency diseases, including, but not limited to, organ transplant rejection, graft versus host disease; renal or prostatic diseases, including, but not limited to, diabetic nephropathy, polycystic kidney disease, nephrosclerosis, glomerulonephritis, prostate hyperplasia; metabolic disorders, including, but not limited to, obesity; infection, including, but not limited to, Helicobacter pylori , Hepatitis and Influenza viruses, fever, and sepsis; pulmonary diseases, including, but not limited to, chronic obstructive pulmonary disease (COPD) and acute respiratory distress syndrome (ARDS); genetic developmental diseases, including,
  • COPD chronic obstructive pulmonary disease
  • ARDS acute respiratory distress
  • the compounds of Formula I, Formula II, or Formula III with kinase activity IC 50 less than 10 ⁇ M as determined in a standard assay described herein can be used to treat protein kinase mediated diseases and conditions related to the following protein kinases, including any mutations thereof, for example without limitation:
  • Halogen refer to all halogens, that is, chloro (Cl), fluoro (F), bromo (Br), or iodo (I).
  • Haldroxyl or “hydroxy” refer to the group —OH.
  • Thiol refers to the group —SH.
  • “Lower alkyl” alone or in combination means an alkane-derived radical containing from 1 to 6 carbon atoms (unless specifically defined) that includes a straight chain alkyl or branched alkyl.
  • the straight chain or branched alkyl group is attached at any available point to produce a stable compound.
  • a lower alkyl is a straight or branched alkyl group containing from 1-6, 1-4, or 1-2, carbon atoms, such as methyl, ethyl, propyl, isopropyl, butyl, t-butyl, and the like.
  • a “substituted lower alkyl” denotes lower alkyl that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, attached at any available atom to produce a stable compound, wherein the substituents are selected from the group consisting of —F, —OH, —NH 2 , —NO 2 , —CN, —C(O)OH, —C(S)OH, —C(O)NH 2 , —C(S)NH 2 , —S(O) 2 NH 2 , —NHC(O)NH 2 , —NHC(S)NH 2 , —NHS(O) 2 NH 2 , —C(NH)NH 2 , —OR o , —SR o , —OC(O)R o , —OC(S)R o , —C(O)R o , —C(S)R o ,
  • substitutions include subsets of these substitutions, such as are indicated herein, for example, in the description of compounds of Formula I, Formula II, or Formula III, attached at any available atom to produce a stable compound.
  • fluoro substituted lower alkyl denotes a lower alkyl group substituted with one or more fluoro atoms, such as perfluoroalkyl, where preferably the lower alkyl is substituted with 1, 2, 3, 4 or 5 fluoro atoms, also 1, 2, or 3 fluoro atoms.
  • substitutions are attached at any available atom to produce a stable compound, when optionally substituted alkyl is an R group of a moiety such as —OR (e.g.
  • substitution of the alkyl R group is such that substitution of the alkyl carbon bound to any O, S, or N of the moiety (except where N is a heteroaryl ring atom) excludes substituents that would result in any O, S, or N of the substituent (except where N is a heteroaryl ring atom) being bound to the alkyl carbon bound to any O, S, or N of the moiety.
  • C 2-6 alkyl denotes lower alkyl containing 2-6 carbon atoms.
  • a “substituted C 2-6 alkyl” denotes optionally substituted lower alkyl containing 2-6 carbon atoms.
  • a “substituted methyl” denotes methyl that is independently substituted, unless indicated otherwise, with 1, 2, or 3 substituents, wherein the substituents are selected as per optionally substituted lower alkyl.
  • C 1-3 alkylene refers to a divalent alkane-derived radical containing 1-3 carbon atoms, straight chain or branched, from which two hydrogen atoms are taken from the same carbon atom or from different carbon atoms.
  • C 1-3 alkylene includes methylene —CH 2 —, ethylene —CH 2 CH 2 —, propylene —CH 2 CH 2 CH 2 —, and isopropylene —CH(CH 3 )CH 2 — or —CH 2 CH(CH 3 )—.
  • C 1-3 alkylene substituted with one or more substituents indicates C 1-3 alkylene that is independently substituted, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents as indicated, attached at any available atom to produce a stable compound.
  • “Lower alkenyl” alone or in combination means a straight or branched hydrocarbon containing 2-6 carbon atoms (unless specifically defined) and at least one, preferably 1-3, more preferably 1-2, most preferably one, carbon to carbon double bond. Carbon to carbon double bonds may be either contained within a straight chain or branched portion. Examples of lower alkenyl groups include ethenyl, propenyl, isopropenyl, butenyl, and the like.
  • a “substituted lower alkenyl” denotes lower alkenyl that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, attached at any available atom to produce a stable compound, wherein the substituents are selected from the group consisting of —F, —OH, —NH 2 , —NO 2 , —CN, —C(O)OH, —C(S)OH, —C(O)NH 2 , —C(S)NH 2 , —S(O) 2 NH 2 , —NHC(O)NH 2 , —NHC(S)NH 2 , —NHS(O) 2 NH 2 , —C(NH)NH 2 , —OR o , —SR o , —OC(O)R o , —OC(S)R o , —C(O)R o , —C(S)R o
  • substitutions include subsets of these substitutions, such as are indicated herein, for example, in the description of compounds of Formula I, Formula II, or Formula III, attached at any available atom to produce a stable compound.
  • fluoro substituted lower alkenyl denotes a lower alkenyl group substituted with one or more fluoro atoms, where preferably the lower alkenyl is substituted with 1, 2, 3, 4 or 5 fluoro atoms, also 1, 2, or 3 fluoro atoms.
  • substitutions are attached at any available atom to produce a stable compound
  • substitution of alkenyl groups are such that —F, —C(O)—, —C(S)—, —C(NH)—, —S(O)—, —S(O) 2 —, —O—, —S—, or N (except where N is a heteroaryl ring atom), are not bound to an alkene carbon thereof.
  • alkenyl is a substituent of another moiety or an R group of a moiety such as —OR, —NHR, —C(O)R, and the like
  • substitution of the moiety is such that any —C(O)—, —C(S)—, —S(O)—, —S(O) 2 —, —O—, —S—, or N thereof (except where N is a heteroaryl ring atom) are not bound to an alkene carbon of the alkenyl substituent or R group.
  • alkenyl is a substituent of another moiety or an R group of a moiety such as —OR, —NHR, —C(O)NHR, and the like
  • substitution of the alkenyl R group is such that substitution of the alkenyl carbon bound to any O, S, or N of the moiety (except where N is a heteroaryl ring atom) excludes substituents that would result in any O, S, or N of the substituent (except where N is a heteroaryl ring atom) being bound to the alkenyl carbon bound to any O, S, or N of the moiety.
  • An “alkenyl carbon” refers to any carbon within an alkenyl group, whether saturated or part of the carbon to carbon double bond.
  • An “alkene carbon” refers to a carbon within an alkenyl group that is part of a carbon to carbon double bond.
  • “Lower alkynyl” alone or in combination means a straight or branched hydrocarbon containing 2-6 carbon atoms (unless specifically defined) containing at least one, preferably one, carbon to carbon triple bond.
  • alkynyl groups include ethynyl, propynyl, butynyl, and the like.
  • a “substituted lower alkynyl” denotes lower alkynyl that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, attached at any available atom to produce a stable compound, wherein the substituents are selected from the group consisting of —F, —OH, —NH 2 , —NO 2 , —CN, —C(O)OH, —C(S)OH, —C(O)NH 2 , —C(S)NH 2 , —S(O) 2 NH 2 , —NHC(O)NH 2 , —NHC(S)NH 2 , —NHS(O) 2 NH 2 , —C(NH)NH 2 , —OR o , —SR o , —OC(O)R o , —OC(S)R o , —C(O)R o , —C(S)R
  • substitutions include subsets of these substitutions, such as are indicated herein, for example, in the description of compounds of Formula I, Formula II, or Formula III, attached at any available atom to produce a stable compound.
  • fluoro substituted lower alkynyl denotes a lower alkynyl group substituted with one or more fluoro atoms, where preferably the lower alkynyl is substituted with 1, 2, 3, 4 or 5 fluoro atoms, also 1, 2, or 3 fluoro atoms.
  • substitutions are attached at any available atom to produce a stable compound
  • substitution of alkynyl groups are such that —F, —C(O)—, —C(S)—, —C(NH)—, —S(O)—, —S(O) 2 —, —O—, —S—, or N (except where N is a heteroaryl ring atom) are not bound to an alkyne carbon thereof.
  • alkynyl is a substituent of another moiety or an R group of a moiety such as —OR—, —NHR—, —C(O)R, and the like
  • substitution of the moiety is such that any —C(O)—, —C(S)—, —S(O)—, —S(O) 2 —, —O—, —S—, or N thereof (except where N is a heteroaryl ring atom) are not bound to an alkyne carbon of the alkynyl substituent or R group.
  • alkynyl is a substituent of another moiety or an R group of a moiety such as —OR, —NHR, —C(O)NHR, and the like
  • substitution of the alkynyl R group is such that substitution of the alkynyl carbon bound to any O, S, or N of the moiety (except where N is a heteroaryl ring atom) excludes substituents that would result in any O, S, or N of the substituent (except where N is a heteroaryl ring atom) being bound to the alkynyl carbon bound to any O, S, or N of the moiety.
  • alkynyl carbon refers to any carbon within an alkynyl group, whether saturated or part of the carbon to carbon triple bond.
  • alkyne carbon refers to a carbon within an alkynyl group that is part of a carbon to carbon triple bond.
  • Cycloalkyl refers to saturated or unsaturated, non-aromatic monocyclic, bicyclic or tricyclic carbon ring systems of 3-10, also 3-8, more preferably 3-6, ring members per ring, such as cyclopropyl, cyclopentyl, cyclohexyl, adamantyl, and the like.
  • a “substituted cycloalkyl” is a cycloalkyl that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, attached at any available atom to produce a stable compound, wherein the substituents are selected from the group consisting of halogen, —OH, —NH 2 , —NO 2 , —CN, —C(O)OH, —C(S)OH, —C(O)NH 2 , —C(S)NH 2 , —S(O) 2 NH 2 , —NHC(O)NH 2 , —NHC(S)NH 2 , —NHS(O) 2 NH 2 , —C(NH)NH 2 , —OR o , —SR o , —OC(O)R o , —OC(S)R o , —C(O)R o , —C(S)R
  • Heterocycloalkyl refers to a saturated or unsaturated non-aromatic cycloalkyl group having from 5 to 10 atoms in which from 1 to 3 carbon atoms in the ring are replaced by heteroatoms of O, S or N, and are optionally fused with benzo or heteroaryl of 5-6 ring members. Heterocycloalkyl is also intended to include oxidized S or N, such as sulfinyl, sulfonyl and N-oxide of a tertiary ring nitrogen. Heterocycloalkyl is also intended to include compounds in which a ring carbon may be oxo substituted, i.e.
  • the ring carbon is a carbonyl group, such as lactones and lactams.
  • the point of attachment of the heterocycloalkyl ring is at a carbon or nitrogen atom such that a stable ring is retained.
  • heterocycloalkyl groups include, but are not limited to, morpholino, tetrahydrofuranyl, dihydropyridinyl, piperidinyl, pyrrolidinyl, pyrrolidonyl, piperazinyl, dihydrobenzofuryl, and dihydroindolyl.
  • a “substituted heterocycloalkyl” is a heterocycloalkyl that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, attached at any available atom to produce a stable compound, wherein the substituents are selected from the group consisting of halogen, —OH, —NH 2 , —NO 2 , —CN, —C(O)OH, —C(S)OH, —C(O)NH 2 , —C(S)NH 2 , —S(O) 2 NH 2 , —NHC(O)NH 2 , —NHC(S)NH 2 , —NHS(O) 2 NH 2 , —C(NH)NH 2 , —OR o , —SR o , —OC(O)R o , —OC(S)R o , —C(O)R o , —C(S)R
  • Aryl alone or in combination refers to a monocyclic or bicyclic ring system containing aromatic hydrocarbons such as phenyl or naphthyl, which may be optionally fused with a cycloalkyl of preferably 5-7, more preferably 5-6, ring members.
  • aromatic hydrocarbons such as phenyl or naphthyl
  • Arylene is a divalent aryl.
  • a “substituted aryl” is an aryl that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, attached at any available atom to produce a stable compound, wherein the substituents are selected from the group consisting of halogen, —OH, —NH 2 , —NO 2 , —CN, —C(O)OH, —C(S)OH, —C(O)NH 2 , —C(S)NH 2 , —S(O) 2 NH 2 , —NHC(O)NH 2 , —NHC(S)NH 2 , —NHS(O) 2 NH 2 , —C(NH)NH 2 , —OR o , —SR o , —OC(O)R o , —OC(S)R o , —C(O)R o , —C(S)R o , —C
  • Heteroaryl alone or in combination refers to a monocyclic aromatic ring structure containing 5 or 6 ring atoms, or a bicyclic aromatic group having 8 to 10 atoms, containing one or more, preferably 1-4, more preferably 1-3, even more preferably 1-2, heteroatoms independently selected from the group consisting of O, S, and N. Heteroaryl is also intended to include oxidized S or N, such as sulfinyl, sulfonyl and N-oxide of a tertiary ring nitrogen. A carbon or nitrogen atom is the point of attachment of the heteroaryl ring structure such that a stable compound is produced.
  • heteroaryl groups include, but are not limited to, pyridinyl, pyridazinyl, pyrazinyl, quinaoxalyl, indolizinyl, benzo[b]thienyl, quinazolinyl, purinyl, indolyl, quinolinyl, pyrimidinyl, pyrrolyl, pyrazolyl, oxazolyl, thiazolyl, thienyl, isoxazolyl, oxathiadiazolyl, isothiazolyl, tetrazolyl, imidazolyl, triazolyl, furanyl, benzofuryl, and indolyl.
  • “Nitrogen containing heteroaryl” refers to heteroaryl wherein any heteroatoms are N.
  • Heteroarylene is a divalent heteroaryl.
  • a “substituted heteroaryl” is a heteroaryl that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, attached at any available atom to produce a stable compound, wherein the substituents are selected from the group consisting of halogen, —OH, —NH 2 , —NO 2 , —CN, —C(O)OH, —C(S)OH, —C(O)NH 2 , —C(S)NH 2 , —S(O) 2 NH 2 , —NHC(O)NH 2 , —NHC(S)NH 2 , —NHS(O) 2 NH 2 , —C(NH)NH 2 , —OR o , —SR o , —
  • R o , R p , R e , R d , R e , R f and R g as used in the description of optional substituents for alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl are defined as follows:
  • each R o , R p , and R c are independently selected from the group consisting of R d , R e , R f , and R g , or R p and R c combine with the nitrogen to which they are attached to form a 5-7 membered heterocycloalkyl or a 5 or 7 membered nitrogen containing heteroaryl, wherein the 5-7 membered heterocycloalkyl or 5 or 7 membered nitrogen containing heteroaryl are optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents selected from the group consisting of halogen, —NO 2 , —CN, —OH, —NH 2 , —OR u , —SR u , —NHR u , —NR u R u , —R x , and —R y ; each R d is independently lower alkyl, wherein lower alkyl is optionally substituted with one or more, preferably 1,
  • all occurrences of optionally substituted lower alkyl, optionally substituted C 2-6 alkyl, optionally substituted lower alkenyl, or optionally substituted lower alkynyl are optionally substituted with one or more, also 1, 2 or 3 groups or substituents selected from the group consisting of fluoro, —NO 2 , —CN, —OR 1a , —SR 1a , —NR 1a R 1a , —OC(O)R 1a , —OC(S)R 1a , —C(O)R 1a , —C(S)R 1a , —C(O)OR 1a , —C(S)OR 1a , —C(S)OR 1a , —C(O)NR 1a R 1a , —C(S)NR 1a R 1a , —S(O) 2 NR 1a R 1a , —C(NH)NR 1a R 1a , —NR 1a C(O
  • all occurrences of optionally substituted lower alkyl, optionally substituted C 2-6 alkyl, optionally substituted lower alkenyl, or optionally substituted lower alkynyl are optionally substituted with one or more, also 1, 2 or 3 groups or substituents selected from the group consisting of fluoro, —CN, —OR 1a , —SR 1a , —NR 1a R 1a , —C(O)R 1a , —C(S)R 1a , —C(O)OR 1a , —C(O)NR 1a R 1a , —C(S)NR 1a R 1a , —S(O) 2 NR 1a R 1a , —NR 1a C(O)R 1a , —NR 1a C(S)R 1a , —NR 1a S(O) 2 R 1a , —S(O)R 1a , —S(O) 2 R 1a , cycl
  • “Lower alkoxy” denotes the group —OR z , where R z is lower alkyl. “Substituted lower alkoxy” denotes lower alkoxy in which R z is lower alkyl substituted with one or more substituents as indicated herein, for example, in the description of compounds of Formula I, Formula II, or Formula III, including descriptions of substituted cycloalkyl, cycloheteroalkyl, aryl and heteroaryl, attached at any available atom to produce a stable compound. Preferably, substitution of lower alkoxy is with 1, 2, 3, 4, or 5 substituents, also 1, 2, or 3 substituents.
  • fluoro substituted lower alkoxy denotes lower alkoxy in which the lower alkyl is substituted with one or more fluoro atoms, where preferably the lower alkoxy is substituted with 1, 2, 3, 4 or 5 fluoro atoms, also 1, 2, or 3 fluoro atoms. While it is understood that substitutions on alkoxy are attached at any available atom to produce a stable compound, substitution of alkoxy is such that O, S, or N (except where N is a heteroaryl ring atom), are not bound to the alkyl carbon bound to the alkoxy O.
  • alkoxy is described as a substituent of another moiety
  • the alkoxy oxygen is not bound to a carbon atom that is bound to an O, S, or N of the other moiety (except where N is a heteroaryl ring atom), or to an alkene or alkyne carbon of the other moiety.
  • “Lower alkylthio” denotes the group —SR aa , where R aa is lower alkyl. “Substituted lower alkylthio” denotes lower alkylthio in which R aa is lower alkyl substituted with one or more substituents as indicated herein, for example, in the description of compounds of Formula I, Formula II, or Formula III, including descriptions of substituted cycloalkyl, cycloheteroalkyl, aryl and heteroaryl, attached at any available atom to produce a stable compound. Preferably, substitution of lower alkylthio is with 1, 2, 3, 4, or 5 substituents, also 1, 2, or 3 substituents.
  • fluoro substituted lower alkylthio denotes lower alkylthio in which the lower alkyl is substituted with one or more fluoro atoms, where preferably the lower alkylthio is substituted with 1, 2, 3, 4 or 5 fluoro atoms, also 1, 2, or 3 fluoro atoms. While it is understood that substitutions on alkylthio are attached at any available atom to produce a stable compound, substitution of alkylthio is such that O, S, or N (except where N is a heteroaryl ring atom), are not bound to the alkyl carbon bound to the alkylthio S.
  • alkylthio is described as a substituent of another moiety
  • the alkylthio sulfur is not bound to a carbon atom that is bound to an O, S, or N of the other moiety (except where N is a heteroaryl ring atom), or to an alkene or alkyne carbon of the other moiety.
  • Amino or “amine” denotes the group —NH 2 .
  • Mono-alkylamino denotes the group —NHR bb where R bb is lower alkyl.
  • Di-alkylamino denotes the group —NR bb R ee , where R dd and R cc are independently lower alkyl.
  • “Cycloalkylamino” denotes the group —NR dd R ee , where R dd and R ee combine with the nitrogen to form a 5-7 membered heterocycloalkyl, where the heterocycloalkyl may contain an additional heteroatom within the ring, such as O, N, or S, and may also be further substituted with lower alkyl.
  • Examples of 5-7 membered heterocycloalkyl include, but are not limited to, piperidine, piperazine, 4-methylpiperazine, morpholine, and thiomorpholine.
  • composition refers to a formulation suitable for administration to an intended animal subject for therapeutic purposes that contains at least one pharmaceutically active compound and at least one pharmaceutically acceptable carrier or excipient.
  • pharmaceutically acceptable indicates that the indicated material does not have properties that would cause a reasonably prudent medical practitioner to avoid administration of the material to a patient, taking into consideration the disease or conditions to be treated and the respective route of administration. For example, it is commonly required that such a material be essentially sterile, e.g., for injectibles.
  • the term “therapeutically effective” or “effective amount” indicates that the materials or amount of material is effective to prevent, alleviate, or ameliorate one or more symptoms of a disease or medical condition, and/or to prolong the survival of the subject being treated.
  • the terms “synergistically effective” or “synergistic effect” indicate that two or more compounds that are therapeutically effective, when used in combination, provide improved therapeutic effects greater than the additive effect that would be expected based on the effect of each compound used by itself.
  • ligand and “modulator” are used equivalently to refer to a compound that changes (i.e., increases or decreases) the activity of a target biomolecule, e.g., an enzyme such as a kinase.
  • a ligand or modulator will be a small molecule, where “small molecule refers to a compound with a molecular weight of 1500 daltons or less, or preferably 1000 daltons or less, 800 daltons or less, or 600 daltons or less.
  • an “improved ligand” is one that possesses better pharmacological and/or pharmacokinetic properties than a reference compound, where “better” can be defined by one skilled in the relevant art for a particular biological system or therapeutic use.
  • the terms “greater affinity” and “selective” indicates that the compound binds more tightly than a reference compound, or than the same compound in a reference condition, i.e., with a lower dissociation constant.
  • the greater affinity is at least 2, 3, 4, 5, 8, 10, 50, 100, 200, 400, 500, 1000, or 10,000-fold greater affinity.
  • the term “synthesizing” and like terms means chemical synthesis from one or more precursor materials.
  • enzymes can be assayed based on their ability to act upon a detectable substrate.
  • a compound or ligand can be assayed based on its ability to bind to a particular target molecule or molecules.
  • the term “modulating” or “modulate” refers to an effect of altering a biological activity, especially a biological activity associated with a particular biomolecule such as a protein kinase.
  • a biological activity associated with a particular biomolecule such as a protein kinase.
  • an agonist or antagonist of a particular biomolecule modulates the activity of that biomolecule, e.g., an enzyme, by either increasing (e.g. agonist, activator), or decreasing (e.g. antagonist, inhibitor) the activity of the biomolecule, such as an enzyme.
  • Such activity is typically indicated in terms of an inhibitory concentration (IC 50 ) or excitation concentration (EC 50 ) of the compound for an inhibitor or activator, respectively, with respect to, for example, an enzyme.
  • IC 50 inhibitory concentration
  • EC 50 excitation concentration
  • the term “contacting” means that the compound(s) are caused to be in sufficient proximity to a particular molecule, complex, cell, tissue, organism, or other specified material that potential binding interactions and/or chemical reaction between the compound and other specified material can occur.
  • isolated indicates that the sequence is separated from at least a portion of the amino acid and/or nucleic acid sequences with which it would normally be associated.
  • the term “purified” indicates that the subject molecule constitutes a significantly greater proportion of the biomolecules in a composition than the proportion observed in a prior composition, e.g., in a cell culture.
  • the greater proportion can be 2-fold, 5-fold, 10-fold, or more than 10-fold, with respect to the proportion found in the prior composition.
  • the present invention concerns compounds of Formula I, and all sub-generic formulae, compounds of Formula II and all sub-generic formulae, and compounds of Formula III and all sub-generic formulae that are modulators of protein kinases, for example without limitation, the compounds are modulators of at least one of the kinases selected from the group consisting of Abl, Akt1, Akt2, Akt3, ALK, Alk5, B-Raf, Brk, Btk, Cdk2, CDK4, CDK5, CDK6, CHK1, c-Raf-1, Csk, EGFR, EphA1, EphA2, EphB2, EphB4, Erk2, Fak, FGFR1, FGFR2, FGFR3, FGFR4, Flt1, Flt3, Flt4, Fms, Frk, Fyn, Gsk3 ⁇ , Gsk3 ⁇ , HCK, Her2/Erbb2, Her4/Erbb4, IGF1R, IKK beta, Irak4, Itk, Jak1, Jak
  • Protein kinases play key roles in propagating biochemical signals in diverse biological pathways. More than 500 kinases have been described, and specific kinases have been implicated in a wide range of diseases or conditions (i.e., indications), including for example without limitation, cancer, cardiovascular disease, inflammatory disease, neurological disease, and other diseases. As such, kinases represent important control points for small molecule therapeutic intervention. Description of specific target protein kinases contemplated by the present invention may be found, for example, in U.S. patent application Ser. No. 11/473,347 (PCT publication WO2007002433), the disclosure of which is hereby incorporated by reference in its entirety, in addition to the following:
  • A-Raf Target kinase
  • A-Raf i.e., v-raf murine sarcoma 3611 viral oncogene homolog 1
  • ARAF ARAF
  • the mature protein comprises RBD (i.e., Ras binding domain) and phorbol-ester/DAG-type zinc finger domain and is involved in the transduction of mitogenic signals from the cell membrane to the nucleus.
  • A-Raf inhibitors may be useful in treating neurologic diseases such as multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease (AD), Parkinson's disease; neoplastic diseases including, but not limited to, melanoma, glioma, sarcoma, carcinoma (e.g. colorectal, lung, breast, pancreatic, thyroid, renal, ovarian), lymphoma (e.g.
  • neurologic diseases such as multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease (AD), Parkinson's disease
  • neoplastic diseases including, but not limited to, melanoma, glioma, sarcoma, carcinoma (e.g. colorectal, lung, breast, pancreatic, thyroid, renal, ovarian), lymphoma (e.g.
  • muscle regeneration or degeneration including, but not limited to, vascular restenosis, sarcopenia, muscular dystrophies (including, but not limited to, Duchenne, Becker, Emery-Dreifuss, Limb-Girdle, Facioscapulohumeral, Myotonic, Oculopharyngeal, Distal and Congenital Muscular Dystrophies), motor neuron diseases (including, but not limited to, amyotrophic lateral sclerosis, infantile progressive spinal muscular atrophy, intermediate spinal muscular atrophy, juvenile spinal muscular atrophy, spinal bulbar muscular atrophy, and adult spinal muscular atrophy), inflammatory myopathies (including, but not limited to, dermatomyositis, polymyositis, and inclusion body myos
  • B-Raf Target kinase B-Raf (i.e., v-raf murine sarcoma viral oncogene homolog B1) is a 84.4 kDa serine/threonine kinase encoded by chromosome 7q34 (symbol: BRAF).
  • the mature protein comprises RBD (i.e., Ras binding domain), C1 (i.e., protein kinase C conserved region 1) and STK (i.e., serine/threonine kinase) domains.
  • Target kinase B-Raf is involved in the transduction of mitogenic signals from the cell membrane to the nucleus and may play a role in the postsynaptic responses of hippocampal neurons.
  • genes of the RAF family encode kinases that are regulated by Ras and mediate cellular responses to growth signals.
  • B-Raf kinase is a key component of the RAS->Raf->MEK->ERK/MAP kinase signaling pathway, which plays a fundamental role in the regulation of cell growth, division and proliferation, and, when constitutively activated, causes tumorigenesis.
  • the B-type, or B-Raf is the strongest activator of the downstream MAP kinase signaling.
  • the BRAF gene is frequently mutated in a variety of human tumors, especially in malignant melanoma and colon carcinoma.
  • the most common reported mutation was a missense thymine (T) to adenine (A) transversion at nucleotide 1796 (T1796A; amino acid change in the B-Raf protein is Val ⁇ 600> to Glu ⁇ 600>) observed in 80% of malignant melanoma tumors.
  • T1796A missense thymine
  • A adenine transversion at nucleotide 1796
  • Functional analysis reveals that this transversion is the only detected mutation that causes constitutive activation of B-Raf kinase activity, independent of RAS activation, by converting B-Raf into a dominant transforming protein.
  • Niihori et al. report that in 43 individuals with cardio-facio-cutaneous (CFC) syndrome, they identified two heterozygous KRAS mutations in three individuals and eight BRAF mutations in 16 individuals, suggesting that dysregulation of the RAS-RAF-ERK pathway is a common molecular basis for the three related disorders (Niihori et al., Nat. Genet. 2006, 38(3):294-6).
  • c-Raf-1 Target kinase c-Raf-1 (i.e., v-raf murine sarcoma viral oncogene homolog 1) is a 73.0 kDa STK encoded by chromosome 3p25 (symbol: RAF1).
  • c-Raf-1 can be targeted to to the mitochondria by BCL2 (i.e., oncogene B-cell leukemia 2) which is a regulator of apoptotic cell death. Active c-Raf-1 improves BCL2-mediated resistance to apoptosis, and c-Raf-1 phosphorylates BAD (i.e., BCL2-binding protein).
  • BAD i.e., BCL2-binding protein
  • c-Raf-1 is implicated in carcinomas, including colorectal, ovarian, lung and renal cell carcinoma. C-Raf-1 is also implicated as an important mediator of tumor angiogenesis (Hood, J. D. et al., 2002, Science 296, 2404). C-Raf-1 inhibitors may also be useful for the treatment of acute myeloid leukemia and myelodysplastic syndromes (Crump, Curr Pharm Des 2002, 8(25):2243-8).
  • Raf-1 activators may be useful as treatment for neuroendocrine tumors, such as medullary thyroid cancer, carcinoid, small cell lung cancer and pheochromocytoma (Kunnimalaiyaan et al., Anticancer Drugs 2006, 17(2):139-42).
  • B-Raf and/or C-Raf inhibitors may be useful in treating A-Raf-mediated, B-Raf-mediated or c-Raf-1-mediated disease or condition selected from the group consisting of neurologic diseases, including, but not limited to, multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease (AD), Parkinson's disease; neoplastic diseases including, but not limited to, melanoma, glioma, sarcoma, carcinoma (e.g. colorectal, lung, breast, pancreatic, thyroid, renal, ovarian), lymphoma (e.g.
  • histiocytic lymphoma neurofibromatosis, acute myeloid leukemia, myelodysplastic syndrome, leukemia, tumor angiogenesis, neuroendocrine tumors such as medullary thyroid cancer, carcinoid, small cell lung cancer and pheochromocytoma
  • pain of neuropathic or inflammatory origin including, but not limited to, acute pain, chronic pain, cancer-related pain, and migraine
  • cardiovascular diseases including, but not limited to, heart failure, ischemic stroke, cardiac hypertrophy, thrombosis (e.g.
  • inflammation including, but not limited to, psoriasis, arthritis and autoimmune diseases and conditions, osteoarthritis, endometriosis, scarring, vascular restenosis, fibrotic disorders, rheumatoid arthritis, inflammatory bowel disease (IBD); immunodeficiency diseases, including, but not limited to, organ transplant rejection, graft versus host disease; renal or prostatic diseases, including, but not limited to, diabetic nephropathy, polycystic kidney disease, nephrosclerosis, glomerulonephritis, prostate hyperplasia; metabolic disorders, including, but not limited to, obesity; infection, including, but not limited to, Helicobacter pylori , Hepatitis and Influenza viruses, fever, and sepsis; pulmonary diseases, including, but not limited to, chronic obstructive pulmonary disease (COPD) and acute respiratory distress syndrome (ARDS); genetic developmental diseases, including,
  • COPD chronic obstructive pulmonary disease
  • ARDS acute respiratory distress
  • the compounds described herein are useful for treating disorders related to c-kit e.g., diseases related to unregulated kinase signal transduction, including cell proliferative disorders, fibrotic disorders and metabolic disorders, among others.
  • disorders related to c-kit e.g., diseases related to unregulated kinase signal transduction, including cell proliferative disorders, fibrotic disorders and metabolic disorders, among others.
  • cell proliferative disorders which can be treated by the present invention include cancers, and mast cell proliferative disorders.
  • c-kit has also been associated with a number of different types of cancers.
  • association between abnormalities in c-kit and disease are not restricted to cancer.
  • c-kit has been associated with malignancies, including, but not limited to, mast cell tumors, small cell lung cancer, testicular cancer, gastrointestinal stromal tumors (GISTs), glioblastoma, astrocytoma, neuroblastoma, carcinomas of the female genital tract, sarcomas of neuroectodermal origin, colorectal carcinoma, carcinoma in situ, Schwann cell neoplasia associated with neurofibromatosis, acute myelocytic leukemia, acute lymphocytic leukemia, chronic myelogenous leukemia, mastocytosis, melanoma, and canine mast cell tumors, and inflammatory diseases, including, but not limited to, asthma, rheumatoid arthritis, allergic rhinitis, multiple sclerosis,
  • c-fins has been associated with a number of different types of diseases. As such, c-fins has been associated with immune disorders, including rheumatoid arthritis, systemic lupus erythematosis (SLE), and transplant rejection; inflammatory diseases including, but not limited to, osteoarthritis, inflammatory bowel syndrome, ulcerative colitis, Crohn's disease, chronic obstructive pulmonary disease (COPD), emphysema, Kawasaki's Disease, hemophagocytic syndrome (macrophage activation syndrome), multicentric reticulohistiocytosis, and atherosclerosis; metabolic disorders, including, but not limited to, Type I diabetes, Type II diabetes, insulin resistance, hyperglycemia, obesity, and lipolysis; disorders of bone structure, mineralization and bone formation and resorption, including, but not limited to, osteoporosis, increased risk of fracture, Paget's disease, hypercalcemia, infection-mediated osteolysis (e.g.
  • kidney and genitourinary diseases including, but not limited to, endometriosis, nephritis (e.g. glomerulonephritis, interstitial nephritis, Lupus nephritis), tubular necrosis, diabetes-associated renal complications (e.g.
  • diabetic nephropathy and renal hypertrophy
  • disorders of the central nervous system including, but not limited to, multiple sclerosis, stroke, Alzheimer's disease and Parkinson's disease
  • inflammatory and chronic pain including, but not limited to, bone pain
  • cancers including, but not limited to, multiple myeloma, acute myeloid leukemia (AML), chronic myeloid leukemia (CML), prostate cancer, breast cancer, ovarian cancer, melanoma, glioblastoma multiforme, metastasis of tumors to other tissues, and other chronic myeloproliferative diseases such as myelofibrosis.
  • TrkA and TrkB Exemplary Diseases Associated with TrkA and TrkB
  • TrkA Target kinase TrkA (i.e., neurotrophic tyrosine kinase, receptor, type 1) is a 140 kDa tyrosine kinase encoded by chromosome 1q21-q22 (symbol: NTRK1). TrkA inhibitors may be useful in treating pain (e.g. chronic pain, neuropathic pain), cancer (e.g. prostate cancer, lung cancer, myeloid leukemia, pancreatic cancer), allergic disorders (e.g. asthma), arthritis, diabetic retinopathy, macular degeneration and psoriasis.
  • pain e.g. chronic pain, neuropathic pain
  • cancer e.g. prostate cancer, lung cancer, myeloid leukemia, pancreatic cancer
  • allergic disorders e.g. asthma
  • arthritis diabetic retinopathy
  • macular degeneration psoriasis.
  • TrkA is a plasma member receptor composed of an extracellular domain (responsible for high affinity binding to nerve growth factor, NGF), a transmembrane segment and an intracellular protein tyrosine kinase domain (responsible to transmit the NGF signal to initiate and coordinate neuronal responses).
  • NGF binding induces TrkA clustering on the membrane and activates the kinase.
  • the kinase initiates a cascade of protein phosphorylation events through multiple pathways including SHC/Ras/MAPK, PI3K and PLCg1.
  • a TrkA kinase inhibitor would not prevent NGF/TrkA binding, but could prevent down-stream signal transduction.
  • Nerve Growth Factor is produced by a number of tissues and inflammatory cells during tissue injury and host immune response. It initiates and maintains hypersensitivity to incoming stimulus (hyperalgesia) and the perception of non-noxious stimuli (allodynia). Through its high-affinity receptor TrkA, NGF increases the excitation state of sensory neurons leading to the central nervous system (peripheral sensitization), and increases transmitter release from the dorsal spinal cord (central sensitization). In clinical trials, a single NGF subcutaneous injection generated local hyperalgesia persisting up to 7 weeks. At doses above 0.1 microgram/kg, NGF caused muscle pain that varied from mild to moderate, primarily in the bulbar and truncal musculature. Intravenous NGF produced earlier and more pronounced systemic effects (Petty et al, 1994, Ann Neurol. 36: 244-6). Conversely, TrkA kinase inhibitors could be used to treat diseases of enhanced states of nociception.
  • TrkA mRNA and protein levels in bronchial biopsies were increased significantly after allergen exposure in infiltrating mast cells before the onset of symptoms (Kassel et al, 2001, Clin Exp Allergy 31:1432-40).
  • TrkB Target kinase TrkB (i.e., neurotrophic tyrosine kinase, receptor, type 2) is a 145 kDa tyrosine kinase encoded by chromosome 9q22.1 (symbol: NTRK2). TrkB inhibitors may be useful in treating various cancers and their metastases (e.g. prostate cancer, lung cancer, Wilms tumors, neuroblastoma, and pancreatic cancer), and various neuropathies (e.g. stroke, multiple sclerosis, transverse myelitis, and encephalitis).
  • various cancers and their metastases e.g. prostate cancer, lung cancer, Wilms tumors, neuroblastoma, and pancreatic cancer
  • various neuropathies e.g. stroke, multiple sclerosis, transverse myelitis, and encephalitis.
  • paresthesia was developed at the site of subcutaneous injection (Coulie et al, 2000, Gastroenterology 119:41-50). Intrathecal infusion of BDNF in humans also induced paresthesia and warmth as side effects (Ochs et al, 2000, Amyotroph Lateral Scler Other Motor Neuron Disord. 1:201-6). Chronic paresthesia is often a symptom of an underlying neurological disease or traumatic nerve damage. Paresthesia can be caused by disorders affecting the central nervous system, such as stroke and transient ischemic attacks (mini-strokes), multiple sclerosis, transverse myelitis, and encephalitis. Since BDNF binds to TrkB specifically with high affinity these neuropath effects are mediated through TrkB signaling. Thus Trkb kinase inhibitors could be used to treat certain patients with neuropathy.
  • BDNF is known to act at the synapses between primary sensory and spinal dorsal horn neurons to affect pain transmission during inflammation.
  • the primary afferent is the only source of BDNF in the spinal cord, and it is up-regulated in the dorsal root ganglion (DRG) by peripheral NGF a few days after inflammation, and is transported and released into the superficial dorsal horn in an activity-dependent manner. TrkB expression in the dorsal horn also increases for a few days after inflammation.
  • TrkB transient receptor potential canonicals
  • TRPC3 transient receptor potential canonicals
  • Na + channel which mediates a rapid depolarization in the hippocampus.
  • TPC3 transient receptor potential canonicals
  • Anti-BDNF significantly increased the withdrawal threshold in CFA-treated rats, a model of inflammatory pain. Since the swelling at the site of CFA injection was not affected by antiserum, the residual component might be due to peripheral sensitization (Matayoshi et al, 2005, J. Physiol. 569:685-95).
  • TrkB and BDNF co-expression of TrkB and BDNF
  • TrkB with N-Myc amplification co-expression of TrkB with N-Myc amplification
  • TrkB truncations found in these tumors generate activated forms of the intracellular protein tyrosine kinase.
  • the constitutively active TrkB signals through multiple pathways to promote cancer initiation, progression and metastasis.
  • TrkB inhibitors could be used to treat a sub-population of cancer patients with an activated TrkB pathway.
  • TrkB expression is correlated with perineural invasion, positive retroperitoneal margin, and shorter latency to development of liver metastasis (Sclabas et al, 2005, Clin. Cancer. Res V11:440-449).
  • TrkB activates the PI3K pathway to suppress anoikis (apoptosis resulting from loss of cell-matrix interactions) which is one of the physiological barriers to metastasis.
  • TrkB kinase inhibition could break down resistance to anoikis of metastasizing tumors (Douma et al, 2004, Nature 430:1034-9). Therefore, TrkB inhibitors could have utility in a broad range of tumor types.
  • MAP4K4 Target kinase MAP4K4 (i.e., Mitogen-activated protein kinase kinase 4, aka Hematopoietic progenitor kinase/Germinal center kinase-like Kinase) is a 130 kDa serine/threonine kinase encoded by chromosome 2q11.2-q12 (symbol: MAP4K4) and is also known as HGK.
  • Mitogen-activated protein kinase kinase 4 aka Hematopoietic progenitor kinase/Germinal center kinase-like Kinase
  • MAP4K4 is a 130 kDa serine/threonine kinase encoded by chromosome 2q11.2-q12 (symbol: MAP4K4) and is also known as HGK.
  • HGK human STE20/mitogen-activated protein kinase kinase kinase kinase kinase kinase (MAP4K) family of serine/threonine kinases and is the human ortholog of mouse NIK (Nck-interacting kinase).
  • the N-terminus of the mature HGK protein has a catalytic kinase domain that shares 47% and 48% amino acid sequence identity to the catalytic domain of Hematopoietic progenitor kinase 1 (HPK1) and Germinal center kinase (GCK), respectively.
  • HPK1 Hematopoietic progenitor kinase 1
  • GCK Germinal center kinase
  • HGK-induced JNK activation was inhibited by dominant-negative MAP2K4, MAP2K7, and TAK1 mutants.
  • TNF-alpha also stimulated HGK kinase activity.
  • HGK was identified as a putative effect of Rap2 to activate JNK (Machida et al. J. Biol. Chem. 279: 15711-15714, 2004). This link establishes HGK as a potential target for a range of metabolic indications, since the JNK pathway clearly antagonizes insulin signaling.
  • An HGK inhibitor could re-sensitize fat and muscle cells to insulin.
  • HGK is found to be broadly expressed in human tumor cells and can modulate cellular transformation, invasion, and adhesion (Wright et al. Mol. Cell. Biol. 23: 2068-2082, 2003). Wright et al showed HGK to be highly expressed in most tumor cell lines relative to normal tissue. An active role for this kinase in transformation was suggested by an inhibition of H-Ras(V12)-induced focus formation by expression of inactive, dominant-negative mutants of HGK in both fibroblast and epithelial cell lines. Expression of an inactive mutant of HGK also inhibited the anchorage-independent growth of cells yet had no effect on proliferation in monolayer culture.
  • HGK mutants modulated integrin receptor expression and had a striking effect on hepatocyte growth factor-stimulated epithelial cell invasion. Together, these results suggest an important role for HGK in cell transformation and invasiveness. More recently, a small interfering RNA screen for modulators of tumor cell motility identifies MAP4K4 as a promigratory kinase (Collins et al. Proc. Natl. Acad. Sci. USA, 103: 3775-3780, 2006). Collins et al. showed that the knockdown of the HGK transcript inhibited the migration of multiple carcinoma cell lines, indicating a broad role in cell motility, and potently suppressed the invasion of SKOV-3 cells in vitro.
  • Insulin-regulated glucose transporter GLUT4 is a key modulator of whole body glucose homeostasis, and its selective loss in adipose tissue or skeletal muscle causes insulin resistance and diabetes.
  • RNA interference-based screen Tang et al. (Proc Natl Acad Sci USA. 103:2087-2092, 2006) found 4 negative regulators of insulin-responsive glucose transport in mouse adipocytes: Pctk1, Pftk1, Ikbka (CHUK), and HGK.
  • HGK suppressed expression of adipogenic transcription factors, C/EBPA, C/EBPB, and PPARG, and it suppressed surface expression of GLUT4 (SLC2A4), resulting in attenuated membrane hexose transport activity.
  • RNA interference-mediated depletion of HGK early in differentiation enhanced adipogenesis and triglyceride deposition; in fully differentiated adipocytes, loss of HGK upregulated GLUT4 expression.
  • conditions that inhibited adipogenesis such as TNF-alpha treatment or PPARG depletion, markedly upregulated HGK. Tang et al. (Proc Natl Acad Sci USA.
  • MAP4K4-dependent signaling inhibited PPARG-responsive gene expression, adipogenesis, and insulin-stimulated glucose transport. Furthermore, TNF-alpha signaling to down-regulate GLUT4 is impaired in the absence of HGK, indicating that HGK expression is required for optimal TNF-alpha action. This study further supports HGK as a target in metabolic disease, and suggests a role for HGK inhibition in ameliorating the pathology in adipocytes.
  • HGK silencing inhibited TNF-alpha-induced negative signaling inputs by preventing excessive JNK and ERK-1/2 phosphorylation, as well as IRS-1 serine phosphorylation.
  • HGK TNF-alpha-induced insulin resistance on AKT and the AKT substrate 160 (AS160), providing evidence that appropriate insulin signaling inputs for glucose metabolism were rescued.
  • siRNA against HGK restored insulin action on glucose uptake to levels observed in healthy subjects. This study further supports HGK as a target in metabolic diseases such as type II diabetes, and suggests a role for HGK inhibition in ameliorating the pathology in muscle cells.
  • HGK inhibitors may be useful in treating metabolic indications, including re-sensitizing fat and muscle cells to insulin, ameliorating the pathology in adipocytes, ameliorating the pathology in muscle cells, metabolic syndrome and type II diabetes; a broad range of oncology indications, including blocking the migration, invasion and metastasis in many different tumor types; and T-cell mediated autoimmune diseases.
  • a number of different assays for kinase activity can be utilized for assaying for active modulators and/or determining specificity of a modulator for a particular kinase or group or kinases.
  • assays for kinase activity can be utilized for assaying for active modulators and/or determining specificity of a modulator for a particular kinase or group or kinases.
  • one of ordinary skill in the art will know of other assays that can be utilized and can modify an assay for a particular application. For example, numerous papers concerning kinases describe assays that can be used.
  • Additional alternative assays can employ binding determinations.
  • this sort of assay can be formatted either in a fluorescence resonance energy transfer (FRET) format, or using an AlphaScreen (amplified luminescent proximity homogeneous assay) format by varying the donor and acceptor reagents that are attached to streptavidin or the phosphor-specific antibody.
  • FRET fluorescence resonance energy transfer
  • AlphaScreen amplified luminescent proximity homogeneous assay
  • solvents include polar and non-polar solvents known to those of skill in the art, including polar aprotic and polar protic solvents.
  • Polar solvents include, without limitation, protic solvents such as methanol, ethanol, isopropyl alcohol, t-butanol, n-butanol, acetic acid, formic acid or water, or aprotic solvents such as tetrahydrofuran (THF), acetonitrile, dioxane, methylene chloride, dimethylsulfoxide (DMSO), acetone, N,N-dimethylformamide (DMF), N,N-dimethylacetamide (DMA), ethyl acetate, 1,2-dimethoxyethane, 1,2-dichloroethane, chloroform, 1,2-dichloroethane, or pyridine.
  • protic solvents such as methanol, ethanol, isopropyl alcohol, t-butanol, n-
  • Polar solvents include a mixture of water with any of the above, or a mixture of any two or more of the above.
  • Apolar solvents include, without limitation, toluene, benzene, chlorobenzene, xylenes and hexanes.
  • reducing agent includes, without limitation, a reducing agent such as catalytic reducing agents using hydrogen and transition metal catalysts such as palladium, platinum, rhodium, etc. (e.g. Pt/acetic acid/H 2 ); a mixture of trifluoroacetic acid and triethylsilane, borane tetrahydrofuran complex, diborane, borane dimethylsulfide complex, and a combination of sodium borohydride and boron trifluoride; metals such as reduced iron, zinc powder, magnesium etc.; metal hydrogen complex compounds such as alkali metal borohydrides (for example, potassium borohydride, sodium borohydride, lithium borohydride, zinc borohydride, sodium triacetoxyborohydride, etc.), aluminum lithium hydride, etc.; metal hydrides such as sodium hydride, etc.; organic tin compounds (triphenyltin hydride, etc.); and metal salts such as nickel
  • oxidizing agent includes, without limitation, an oxidizing agent such as Dess-Martin reagent, TEMPO (2,2,6,6-tetramethylpiperidine-N-oxide), DDQ (2,3-Dichloro-5,6-dicyano-1,4-benzoquinone), PDC (pyridinium dichromate), PCC (pyridinium chlorochromate), Pyridine.SO3, Chromium trioxide, p-nitroperbenzoic acid, magnesium monoperoxyphthalate, sodium periodate, potassium periodate, hydrogen peroxide, urea peroxide, alkali metal bromates, cumene hydroperoxide, tert-butyl peroxide, peracids such as performic acid, peracetic acid, pertrifluoroacetic acid, perbenzoic acid, m-chloroperbenzoic acid, o-carboxyperbenzoic acid and the like; sodium metaperiodate, bichromic acid; bichromates
  • an oxidizing agent such
  • a nitrogen protecting group is a chemical group covalently bound to a nitrogen atom of a compound that is used to protect the nitrogen from reaction during a synthetic step.
  • the nitrogen protecting group may be added to a compound and removed in a subsequent step by methods known to those of skill in the art.
  • Nitrogen protecting groups include, without limitation, carbamates, amides, N-sulfonyl derivatives, groups of formula —C(O)OR, wherein R is, for example, methyl, ethyl, t-butyl, benzyl, phenylethyl, CH 2 ⁇ CHCH 2 —, and the like, groups of the formula —C(O)R′, wherein R′ is, for example, methyl, phenyl, trifluoromethyl, and the like, groups of the formula —SO 2 R′′, wherein R′′ is, for example, tolyl, phenyl, trifluoromethyl, 2,2,5,7,8-pentamethylchroman-6-yl, 2,3,6-trimethyl-4-methoxyphenyl, and the like, and silanyl containing groups, such as 2-trimethylsilylethoxymethyl, t-butyldimethylsilyl, triisopropylsilyl, and the like.
  • R
  • the methods and compounds will typically be used in therapy for human subjects. However, they may also be used to treat similar or identical indications in other animal subjects.
  • the terms “subject,” “animal subject,” and the like refer to human and non-human vertebrates, e.g. mammals, such as non-human primates, sports and commercial animals, e.g., equines, bovines, porcines, ovines, rodents, and pets, e.g., canines and felines.
  • a description of possible methods and routes of administration may be found, for example, in U.S. patent application Ser. No. 11/473,347 (see also, PCT publication WO2007002433), the disclosure of which is hereby incorporated by reference in its entirety.
  • the mass spectrometry result indicated for a compound may have more than one value due to the isotope distribution of an atom in the molecule, such as a compound having a bromo or chloro substituent.
  • R 2 and R 3 are as defined in paragraph [0009], and R 6 is as defined in paragraph [0023], where indicates the attachment point to the carbonyl carbon
  • an appropriate solvent e.g. methanol
  • an appropriate base e.g. potassium hydroxide, sodium methoxide
  • the reaction is typically allowed to stir at room temperature overnight. Isolation by conventional means (e.g. extraction, washing and filtering) affords a mixture containing compound of Formula Xc, which may be isolated by silica gel chromatography if desired.
  • an oxidizing agent e.g. Dess-Martin periodane, TEMPO, DDQ.
  • TEMPO tetrahydrofuran
  • DDQ oxidizing agent
  • the reaction is allowed to stir at room temperature for 20 minutes. Isolation by conventional means (e.g. extraction and silica gel column chromatography) affords compounds of Formula X.
  • Compound of Formula X is synthesized by reacting a compound of Formula Xa (see Example 1) with a compound of Formula Xd (Y is as defined in Example 1), e.g. benzoyl chloride, in the presence of a Lewis acid (e.g. aluminum trichloride) in an inert solvent (e.g. dichloromethane) under an inert atmosphere (e.g. argon) at room temperature or with heating up to reflux for 1-18 hours.
  • a Lewis acid e.g. aluminum trichloride
  • an inert solvent e.g. dichloromethane
  • an inert atmosphere e.g. argon
  • Compound of Formula Xe can be synthesized by reacting a compound of Formula Xa (see Example 1) with hexamethyltetramine and acetic acid in water with heating to reflux for two hours. After cooling, the desired compound precipitates and may be collected by filtration.
  • Compound of Formula Xf, where P is a nitrogen protecting group is synthesized by reacting a compound Xe with an appropriate reagent to introduce a protecting group (e.g. triisopropylsilylchloride) and a base (e.g. sodium hydride) in a solvent (e.g. tetrahydrofuran) typically at room temperature for 8-12 hours.
  • a protecting group e.g. triisopropylsilylchloride
  • a base e.g. sodium hydride
  • a solvent e.g. tetrahydrofuran
  • Compound of Formula Xh is synthesized by reacting a compound of Formula Xf in a solvent (e.g. tetrahydrofuran) with an organolithium reagent (e.g. phenyl lithium) in a solvent (e.g. tetrahydrofuran) under an inert atmosphere, cooled to ⁇ 78° C.
  • a solvent e.g. tetrahydrofuran
  • an organolithium reagent e.g. phenyl lithium
  • a solvent e.g. tetrahydrofuran
  • An appropriate organolithium reagent can also be prepared by reacting compounds of Formula Xg, where Y 1 is
  • R 6 indicates attachment point to the sulfone
  • R 2 , R 3 and R 6 are as defined in Example 1, with an organolithium reagent (e.g. butyllithium) in a solvent (e.g. tetrahydrofuran) under an inert atmosphere, cooled to ⁇ 78° C.
  • organolithium reagent e.g. butyllithium
  • solvent e.g. tetrahydrofuran
  • the reaction is typically allowed to warm to room temperature and stirred for 30 minutes.
  • the desired compound is isolated by conventional means (e.g. extraction).
  • An intermediate of compound of Formula X is synthesized by reacting a compound of Formula Xh with an appropriate reagent to remove the protecting group, P, (e.g. tetra-n-butyl ammonium fluoride) in an appropriate solvent (e.g. tetrahydrofuran).
  • P protecting group
  • solvent e.g. tetrahydrofuran
  • Compound of Formula X is synthesized by reacting the intermediate from Step 4 with an oxidizing agent (e.g. Dess-Martin periodane, TEMPO) in an aprotic solvent (e.g. tetrahydrofuran) typically at room temperature for 20 minutes.
  • an oxidizing agent e.g. Dess-Martin periodane, TEMPO
  • an aprotic solvent e.g. tetrahydrofuran
  • the desired compound is isolated by conventional means (e.g. extraction and silica gel chromatography).
  • Phenyl-(-1H-pyrrolo[2,3-b]pyridine-5-yl)-amine 1 was synthesized in three steps from 5-bromo-7-azaindole 1 as shown in Scheme 1.
  • 5-Chloro-1H-pyrrolo[2,3-b]pyridine 6 was synthesized in two steps from 5-bromo-1-triisopropylsilyl-7-azaindole 2 as shown in Scheme 2.
  • 5-Isopropyl-1H-pyrrolo[2,3-b]pyridine 13 was synthesized in three steps from 5-bromo-1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridine 2 as described in Scheme 4.
  • 5-Methyl-1H-pyrrolo[2,3-b]pyridine 15 was synthesized in two steps from 5-bromo-1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridine 2 as described in Scheme 5.
  • 5-Methoxy-1H-pyrrolo[2,3-b]pyridine 17 was synthesized in one step from 5-bromo-1H-pyrrolo[2,3-b]pyridine 1 as described in Scheme 6.
  • 5-Pyridin-3-yl-1H-pyrrolo[2,3-b]pyridine 22 was synthesized in one step from 5-bromo-1H-pyrrolo[2,3-b]pyridine 1 as described in Scheme 7.
  • 3-Iodo-1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridine 37 was synthesized in one step from 3-Iodo-1H-pyrrolo[2,3-b]pyridine 36 as shown in Scheme 8.
  • 5-Chloro-3-iodo-1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridine 40 was synthesized in one step from 5-Chloro-3-iodo-1H-pyrrolo[2,3-b]pyridine 39 as shown in Scheme 10.
  • 5-(1-Methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine 46 was synthesized in one step from 5-bromo-1H-pyrrolo[2,3-b]pyridine 1 as shown in Scheme 12.
  • Step 3 Preparation of propane-2-sulfonic acid ⁇ 2,4-difluoro-3-[hydroxy-(5-pyridin-3-yl-1H-pyrrolo[2,3-b]pyridin-3-yl)-methyl]-phenyl ⁇ -amide (50)
  • Step 5 Preparation of (3-Amino-2,6-difluoro-phenyl)-(5-chloro-1H-pyrrolo[2,3-b]pyridin-3-yl)-methanone (56)
  • Step 1 Preparation of 3- ⁇ 3-[2,6-difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl ⁇ -propionic acid methyl ester (P-0033)
  • Step 2 Preparation of 3- ⁇ 3-[2,6-difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl ⁇ -propionic acid (P-0034)
  • Step 3 Preparation of 3- ⁇ 3-[2,6-difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl ⁇ -N-ethyl-propionamide (P-0035)
  • B-Raf P-0001, P-0002, P-0003, P-0005, P-0006, P-0007, P-0009, P-0011, P-0013, P-0014, P-0015, P-0016, P-0017, P-0018, P-0019, P-0020, P-0021, P-0022, P-0023, P-0024, P-0025, P-0026, P-0027, P-0028, P-0030, P-0031, P-0032, P-0033, P-0034, P-0035, P-0036
  • c-Raf-1 P-0001, P-0002, P-0003, P-0005, P-0006, P-0007, P-0008, P-0009, P-0011, P-0013, P-0014, P-0015, P-0016, P-0017, P-0018, P-0019, P-0020, P-0021, P-0022, P-0023, P-0025, P-0030, P-0031
  • Jnk1 P-0019, P-0020, P-0025
  • Kdr P-0001, P-0002, P-0003, P-0004, P-0005, P-0008, P-0010, P-0011, P-0013, P-0014, P-0016, P-0018, P-0019, P-0020, P-0021, P-0022, P-0023, P-0024, P-0025, P-0026, P-0027, P-0028, P-0029, P-0032, P-0034
  • Kit P-0001, P-0002, P-0003, P-0005, P-0006, P-0007, P-0008, P-0009, P-0010, P-0011, P-0013, P-0014, P-0015, P-0016, P-0017, P-0018, P-0019, P-0020, P-0021, P-0022, P-0023, P-0024, P-0025, P-0026, P-0027, P-0028, P-0029, P-0032, P-0035
  • Compounds of the invention such as compounds of Formula I, Formula II, or Formula III, in combination with a standard chemotherapeutic agent, such as 5-fluorouracil, carboplatin, dacarbazine, gefitinib, oxaliplatin, paclitaxel, SN-38, temozolomide, or vinblastine, can be assessed for their effectiveness in killing human tumor cells.
  • a standard chemotherapeutic agent such as 5-fluorouracil, carboplatin, dacarbazine, gefitinib, oxaliplatin, paclitaxel, SN-38, temozolomide, or vinblastine
  • Human tumor cell lines such as A-375 (malignant melanoma), SK-MEL-2 (malignant melanoma, skin metastasis), COLO 205 (colorectal adenocarcinoma, ascites metastasis) or SW-620 (colorectal adenocarcinoma, lymph node metastasis) can be treated with a compound of Formula I, Formula II, or Formula III alone, or in combination with one of the above-mentioned chemotherapeutic agents.
  • A-375 malignant melanoma
  • SK-MEL-2 malignant melanoma, skin metastasis
  • COLO 205 colonrectal adenocarcinoma, ascites metastasis
  • SW-620 colonrectal adenocarcinoma, lymph node metastasis
  • Tumor cells are grown as a monolayer at 37° C. in a humidified atmosphere (5% CO 2 , 95% air).
  • Cells are grown in a suitable culture medium, e.g. RPMI 1640 (Ref BE12-702F, Cambrex, Verviers, Belgium) containing 2 mM L-glutamine and supplemented with 10% fetal bovine serum (Ref DE14-801E, Cambrex).
  • RPMI 1640 Ref BE12-702F, Cambrex, Verviers, Belgium
  • the tumor cells are detached from the culture flask by a 5-minute treatment with trypsin-versene (Ref 02-007E, Cambrex), diluted in Hanks' medium without calcium or magnesium (Ref BE10-543F, Cambrex). Trypsin treatment is neutralized by culture medium addition.
  • the cells are counted in a hemocytometer and their viability assessed by 0.25% trypan blue exclusion.
  • the cell lines are checked for mycoplasma contamination with the Mycotect assay kit (Ref 15672-017, Invitrogen, Cergy-Pontoise, France) in accordance with the manufacturer's instructions.
  • the mycoplasma test is assayed from the culture supernatants of the cell lines and compared to negative and positive controls.
  • the tumor cells (10,000 per well) are plated in 96-well flat-bottom microtitration plates (Ref 055260, Nunc, Dutscher, Brumath, France) and incubated at 37° C. for 24 hours before treatment in 100 ⁇ l of drug-free culture medium supplemented with 10% FBS.
  • the tumor cells are incubated in a 200 ⁇ l final volume of RPMI 1640 supplemented with 10% FBS and containing either a compound of Formula I, Formula II, or Formula III, or one of 5-fluorouracil, carboplatin, dacarbazine, gefitinib, oxaliplatin, paclitaxel, SN-38, temozolomide, or vinblastine.
  • the compounds are tested in a suitable concentration range, such as 10 ⁇ 8 to 10 ⁇ 3 M for a compound of Formula I, Formula II, or Formula III, 5-fluorouracil, dacarbazine or gefitinib, 10 ⁇ 9 to 10 ⁇ 4 M for carboplatin, oxaliplatin, or temozolomide, 10 ⁇ 11 to 10 ⁇ 6 M for paclitaxel or SN-38, and 10 ⁇ 15 to 10 ⁇ 10 M for vinblastine.
  • Compounds of Formula I, Formula II, or Formula III are dissolved in DMSO and diluted with culture medium to the desired concentrations.
  • 5-fluorouracil 50 mg/ml, Dakota Pharm, LePlessis Robinson, France
  • carboplatin 10 mg/ml, Aguettant, Lyon, France
  • paclitaxel 6 mg/ml, Bristol-Myers Squibb SpA, Rueil Malmaison, France
  • dacarbazine Sigma, Saint Quentin Fallavier, France
  • vinblastine Lily France S.A., Saint Cloud, France
  • Gefitinib is dissolved in a mixed solution of RPMI 1640 and DMSO and diluted with culture medium to the desired concentrations (maximum final DMSO of 0.1% v/v).
  • SN-38 (LKT Laboratories, Inc., St. Paul, Minn.) is dissolved in DMSO and diluted with culture medium to the desired concentrations (maximum final DMSO of 0.1% v/v).
  • Temozolomide (LKT Laboratories, Inc., St. Paul, Minn.) is dissolved in water for injection and diluted with culture medium to the desired concentrations. Cells are incubated for 96 hours in the presence of test substances at 37° C. under 5% CO 2 . At the end of treatments, the cytotoxic activity is evaluated by an MTT assay.
  • MTT MTT assay
  • 20 ⁇ l of a 5 mg/ml solution 0.22 ⁇ m filtered tetrazolium reagent (MTT, Ref M2128, Sigma) in Phosphate Buffered Saline (PBS, Ref BE17-517Q, Cambrex) is added in each well.
  • Culture plates are incubated for 2 h at 37° C.
  • the resulting supernatant is removed and formazan crystals dissolved with 200 ⁇ l of DMSO per well.
  • Absorbency (OD) is measured at 570 nm in each well using VICTOR 3 TM 1420 multilabeled counter (Wallac, PerkinElmer, Courtaboeuf, France).
  • the IC 50 for each compound on each cell line is determined from the OD measurements of each sample.
  • the dose response inhibition of cell proliferation is expressed as:
  • IC ( OD of drug exposed cells/ OD of drug free wells) ⁇ 100.
  • the mean of multiple measurements for each concentration is plotted vs. the drug concentration.
  • the dose-response curves are plotted using XLFit 3 (IDBS, United Kingdom).
  • the IC 50 (drug concentration to obtain 50% inhibition of cell proliferation) determination values are calculated using the XLFit 3 from semi-log curves.
  • the IC 50 value determined for each compound in each cell line is used to determine the concentration of a compound of Formula I, Formula II, or Formula III, and of the standard chemotherapeutic to be used in combination.
  • the cells are treated with a combination of five concentrations of a compound of Formula I, Formula II, or Formula III and five concentrations of one of 5-fluorouracil, carboplatin, dacarbazine, gefitinib, oxaliplatin, paclitaxel, SN-38, temozolomide, or vinblastine, based on the IC 50 results.
  • the compounds and cells are treated per the IC 50 determination described above and assayed by the MTT assay.
  • the results are assessed to determine whether the combination is synergistic or antagonistic.
  • the compound interactions are calculated by multiple drug effect analysis and are performed by the median equation principle according to the methodology described by Chou and Talalay (Adv. Enzyme Regul. 1984, 22: 27-55).
  • the combination index (CI) will be calculated by the Chou et al. equation (Adv. Enzyme Regul. 1984, 22: 27-55; Encyclopaedia of human biology, Academic Press, 1991, 2: 371-9; Synergism and Antagonism in Chemotherapy, Academic Press, 1991, 61-102) which takes into account both the potency (D m or IC 50 ) and the shape of the dose-effect curve (the m value).
  • the general equation for the CI of the two compounds is given by:
  • (D X ) 1 and (D X ) 2 in the denominators are the doses (or concentrations) for compound 1 and compound 2 alone which demonstrate x % of inhibition
  • (D) 1 and (D) 2 in the numerators are doses of both compounds (1 and 2) in combination that also inhibit x % (iso-effective).
  • the (D X ) 1 and (D X ) 2 can be calculated from the median-effect equation of Chou et al. (J. Natl. Cancer Inst. 1994, 86: 1517-24):
  • CI will be calculated with CalcuSyn software (Biosoft, UK) from the mean affected fraction at each drug ratio concentration.
  • any of the terms “comprising”, “consisting essentially of” and “consisting of” may be replaced with either of the other two terms.
  • the invention also includes another embodiment wherein one of these terms is replaced with another of these terms.
  • the terms have their established meaning.
  • one embodiment may encompass a method “comprising” a series of steps, another embodiment would encompass a method “consisting essentially of” the same steps, and a third embodiment would encompass a method “consisting of” the same steps.

Abstract

Compounds active on protein kinases are described, as well as methods of using such compounds to treat diseases and conditions associated with aberrant activity of protein kinases.

Description

    RELATED PATENT APPLICATIONS
  • This application is a divisional application of U.S. patent application Ser. No. 11/960,590, “Compounds and Methods for Kinase Modulation, and Indications Therefor”, filed Dec. 19, 2007, which claims priority to U.S. Provisional App. No. 60/876,953, entitled “Compounds and Methods for Kinase Modulation, and Indications Therefor”, filed Dec. 21, 2006, and is related to U.S. patent application Ser. No. 11/473,347, entitled “Compounds and Methods for Kinase Modulation, and Indications Therefor”, filed Jun. 21, 2006, which claims the benefit of U.S. Provisional App. No. 60/731,528, entitled “Compounds and Methods for Kinase Modulation, and Indications Therefor”, filed Oct. 28, 2005, and U.S. Provisional App. No. 60/692,960, entitled “Compounds and Methods for Kinase Modulation, and Indications Therefor”, filed Jun. 22, 2005, all of which are incorporated herein by reference in their entireties and for all purposes.
  • FIELD OF THE INVENTION
  • The present invention relates to kinases and compounds which modulate kinases, and uses therefor. Particular embodiments contemplate disease indications which are amenable to treatment by modulation of kinase activity by the compounds of the present invention.
  • BACKGROUND OF THE INVENTION
  • The information provided herein is intended solely to assist the understanding of the reader. None of the information provided nor references cited is admitted to be prior art to the present invention. Each of the references cited herein is incorporated in its entirety.
  • Receptor protein kinases regulate key signal transduction cascades that control or are involved in the control of a plethora of physiological functions including cellular growth and proliferation, cell differentiation, cellular development, cell division, cell adhesion, stress response, short-range contact-mediated axonal guidance, transcription regulation, aberrant mitogenesis, angiogenesis, abnormal endothelial cell-cell or cell-matrix interactions during vascular development, inflammation, lymphohematopoietic stem cell activity, protective immunity against specific bacteria, allergic asthma, aberrant tissue-specific responses to the activation of the JNK signal transduction pathway, cell transformation, memory, apoptosis, competitive activity-dependent synapse modification at the neuromuscular synapse, immunological mediation of disease, and calcium regulation.
  • Specific disease states associated with aberrant regulation of protein kinases include, for example without limitation, acrocephalo-syndactyl type I, acute myeloid leukemia, AIDS-induced non-Hodgkin's lymphoma, Alzheimer's disease, amyotrophic lateral sclerosis, arthritis, asthma, atherosclerosis, atopic dermatitis, autoimmune diseases, bacterial infection, bladder cancer, cancer of the breast, cancer of the central nervous system, cancer of the colon, cancer of the endometrium, cancer of the fallopian tube, cancer of the gastrointestinal tract, cancer of the ovary, heart failure, chronic myeloid leukemia, colon carcinoma, colorectal cancer, chronic obstructive pulmonary disease (COPD), Crouzon Syndrome, diabetes, diabetic nephropathy, emphysema, endometriosis, epidermoid cancer, fibrotic disorders, gastrointestinal stromal tumor (GIST), glomerulonephritis, Graves' disease, head injury, hepatocellular carcinoma, Hirschsprung's disease, human gliomas, immunodeficiency diseases, inflammatory disorders, ischemic stroke, Jackson-Weiss syndrome, leiomyosarcoma, leukemias, lupus nephritis, malignant melanoma, malignant nephrosclerosis, mastocytosis, mast cell tumors, melanoma of the colon, MEN2 syndromes, metabolic disorders, migraine, multiple sclerosis, myeloproliferative disorders, nephritis, neurodegenerative diseases, neurotraumatic diseases, non small cell lung cancer, organ transplant rejection, osteoporosis, pain, Parkinson's disease, Pfeiffer Syndrome, polycystic kidney disease, primary lymphoedema, prostate cancer, psoriasis, vascular restenosis, rheumatoid arthritis, dermal and tissue scarring, selective T-cell defect (STD), severe combined immunodeficiency (SCID), small cell lung cancer, spinal cord injury, squamous cell carcinoma, systemic lupus erythematosis, testicular cancer, thrombotic microangiopathy syndromes, Wegener's granulomatosis, X-linked agammaglobulinemia, viral infection, diabetic retinopathy, alopecia, erectile dysfunction, macular degeneration, chronic lymphocytic leukemia (CLL), myelodysplastic syndrome (MDS), neurofibromatosis, and tuberous sclerosis.
  • This application is related to the following published patent applications: WO 2004024895, US 20040142864, WO 2004078923, US 20050170431, WO 2005028624, US 20050164300, and WO 2005062795, each of which are hereby incorporated by reference herein in their entireties including all specifications, figures, and tables, and for all purposes.
  • SUMMARY OF THE INVENTION
  • Compounds are contemplated that are active on protein kinases in general, including, but not limited to, Abl, Akt1, Akt2, Akt3, ALK, Alk5, A-Raf, B-Raf, Brk, Btk, Cdk2, CDK4, CDK5, CDK6, CHK1, c-Raf-1, Csk, EGFR, EphA1, EphA2, EphB2, EphB4, Erk2, Fak, FGFR1, FGFR2, FGFR3, FGFR4, Flt1, Flt3, Flt4, Fms, Frk, Fyn, Gsk3a, Gsk3β, HCK, Her2/Erbb2, Her4/Erbb4, IGF1R, IKK beta, Irak4, Itk, Jak1, Jak2, Jak3, Jnk1, Jnk2, Jnk3, Kdr, Kit, Lck, Lyn, MAP2K1, MAP2K2, MAP4K4, MAPKAPK2, Met, Mnk1, MLK1, p38, PDGFRA, PDGFRB, PDPK1, Pim1, Pim2, Pim3, PKC alpha, PKC beta, PKC theta, Plk1, Pyk2, Ret, ROCK1, ROCK2, Ron, Src, Stk6, Syk, TEC, Tie2, TrkA, TrkB, Yes, and/or Zap70, including any mutations of these kinases. In some aspects, the compounds are active on protein kinases including A-Raf, B-Raf and/or c-Raf-1, including any mutations thereof. In some aspects, compounds are of Formula I, Formula II, or Formula III, as described below.
  • Also contemplated in accordance with the present invention are methods for the use of the above-described compounds in treating diseases and conditions associated with regulation of the activity of the above-described kinases. Thus, the use of compounds for therapeutic methods involving modulation of protein kinases are provided, as well as compounds that can be used for therapeutic methods involving modulation of protein kinases.
  • In some embodiments, compounds have the structure according to the following Formula I:
  • Figure US20110092538A1-20110421-C00001
  • all salts, prodrugs, tautomers and isomers thereof,
  • wherein:
      • R1 is selected from the group consisting of hydrogen, halogen, optionally substituted lower alkyl, optionally substituted lower alkenyl, optionally substituted lower alkynyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, —OH, —NH2, —CN, —NO2, —C(O)OH, —S(O)2NH2, —C(O)NH2, —C(S)NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —OR4, —SR4, —NR5R4, —C(O)R4, —C(S)R4, —C(O)OR4, —C(O)NR5R4, —C(S)NR5R4, —S(O)2NR5R4, —NR5C(O)R4, —NR5C(S)R4, —NR5S(O)2R4, —NR5C(O)NH2, —NR5C(O)NR5R4, —NR5C(S)NH2, —NR5C(S)NR5R4, —NR5S(O)2NH2, —NR5S(O)2NR5R4, —S(O)R4, and —S(O)2R4;
      • R2 is selected from the group consisting of halogen, optionally substituted lower alkyl, optionally substituted lower alkenyl, optionally substituted lower alkynyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, —OH, —NH2, —CN, —NO2, —C(O)OH, —S(O)2NH2, —C(O)NH2, —C(S)NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —OR4, —SR4, —NR5R4, —C(O)R4, —C(S)R4, —C(O)OR4, —C(O)NR5R4, —C(S)NR5R4, —S(O)2NR5R4, —NR5C(O)R4, —NR5C(S)R4, —NR5S(O)2R4, —NR5C(O)NH2, —NR5C(O)NR5R4, —NR5C(S)NH2, —NR5C(S)NR5R4, —NR5S(O)2NH2, —NR5S(O)2NR5R4, —S(O)R4, and —S(O)2R4;
      • R3 is selected from the group consisting of hydrogen, fluoro and chloro;
      • R4 is selected from the group consisting of optionally substituted lower alkyl, optionally substituted lower alkenyl, provided, however, that when R4 is optionally substituted lower alkenyl, no alkene carbon thereof is bound to N, S, O, S(O), S(O)2, C(O) or C(S) of —OR4, —SR4, —NR5R4, —C(O)R4, —C(S)R4, —C(O)OR4, —C(O)NR5R4, —C(S)NR5R4, —S(O)2NR5R4, —NR5C(O)R4, —NR5C(S)R4, —NR5S(O)2R4, —NR5C(O)NH2, —NR5C(O)NR5R4, —NR5C(S)NH2, —NR5C(S)NR5R4, —NR5S(O)2NH2, —NR5S(O)2NR5R4, —S(O)R4, or —S(O)2R4, optionally substituted lower alkynyl, provided, however, that when R4 is optionally substituted lower alkynyl, no alkyne carbon thereof is bound to N, S, O, S(O), S(O)2, C(O) or C(S) of —OR4, —SR4, —NR5R4, —C(O)R4, —C(S)R4, —C(O)OR4, —C(O)NR5R4, —C(S)NR5R4, —S(O)2NR5R4, —NR5C(O)R4, —NR5C(S)R4, —NR5S(O)2R4, —NR5C(O)NH2, —NR5C(O)NR5R4, —NR5C(S)NH2, —NR5C(S)NR5R4, —NR5S(O)2NH2, —NR5S(O)2NR5R4, —S(O)R4, or —S(O)2R4, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; and
      • R5 is selected from the group consisting of hydrogen and optionally substituted lower alkyl, provided, however, the compound is not
  • Figure US20110092538A1-20110421-C00002
    Figure US20110092538A1-20110421-C00003
    Figure US20110092538A1-20110421-C00004
    Figure US20110092538A1-20110421-C00005
    Figure US20110092538A1-20110421-C00006
  • In some embodiments of compounds of Formula I, R1 is selected from the group consisting of hydrogen, halogen, optionally substituted lower alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, —CN, —OR4, —SR4, —NR5R4, —C(O)R4, —C(O)NR5R4, —S(O)2NR5R4, —NR5C(O)R4, —NR5S(O)2R4, —S(O)R4, and —S(O)2R4, and R2 is selected from the group consisting of halogen, optionally substituted lower alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, —CN, —OR4, —SR4, —NR5R4, —C(O)R4, —C(O)NR5R4, —S(O)2NR5R4, —NR5C(O)R4, —NR5S(O)2R4, —S(O)R4, and —S(O)2R4.
  • In some embodiments of compounds of Formula I, R1 is selected from the group consisting of hydrogen, halogen, lower alkyl, lower alkenyl, lower alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, —OH, —NH2, —CN, —NO2, —C(O)OH, —S(O)2NH2, —C(O)NH2, —C(S)NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —OR7, —SR7, —NR8R7, —C(O)R7, —C(S)R7, —C(O)OR7, —C(O)NR8R7, —C(S)NR8R7, —S(O)2NR8R7, —NR8C(O)R7, —NR8C(S)R7, —NR8S(O)2R7, —NR8C(O)NH2, —NR8C(O)NR8R7, —NR8C(S)NH2, —NR8C(S)NR8R7, —NR8S(O)2NH2, —NR8S(O)2NR8R7, —S(O)R7, and —S(O)2R7, wherein lower alkyl, lower alkenyl or lower alkynyl are optionally substituted with one or more substituents selected from the group consisting of fluoro, —OH, —NH2, C(O)OH, —C(O)NH2, —OR7, —NR8R7, —C(O)OR7, —C(O)NR8R7, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as R1 or as substituents of lower alkyl, lower alkenyl or lower alkynyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —S(O)2NH2, —C(O)NH2, —OR9, —SR9, —NR8R9, —NR8C(O)R9, —NR8S(O)2R9, —S(O)2R9, —S(O)2NR8R9, —C(O)R9, —C(O)NR8R9, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino; and R2 is selected from the group consisting of halogen, lower alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, —OH, —NH2, —CN, —NO2, —C(O)OH, —S(O)2NH2, —C(O)NH2, —C(S)NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —OR7, —SR7, —NR8R7, —C(O)R7, —C(S)R7, —C(O)OR7, —C(O)NR8R7, —C(S)NR8R7, —S(O)2NR8R7, —NR8C(O)R7, —NR8C(S)R7, —NR8S(O)2R7, —NR8C(O)NH2, —NR8C(O)NR8R7, —NR8C(S)NH2, —NR8C(S)NR8R7, —NR8S(O)2NH2, —NR8S(O)2NR8R7, —S(O)R7, and —S(O)2R7, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, —OR7, —NR8R7, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as R2 or as substituents of lower alkyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —S(O)2NH2, —C(O)NH2, —OR9, —SR9, —NR8R9, —NR8C(O)R9, —NR8S(O)2R9, —S(O)2R9, —S(O)2NR8R9, —C(O)R9, —C(O)NR8R9, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino; wherein R7 is selected from the group consisting of lower alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as R7 or as substituents of lower alkyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —S(O)2NH2, —C(O)NH2, —OR9, —SR9, —NR8R9, —NR8C(O)R9, —NR8S(O)2R9, —S(O)2R9, —S(O)2NR8R9, —C(O)R9, —C(O)NR8R9, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino; R8 at each occurrence is independently hydrogen or lower alkyl; and R9 at each occurrence is independently selected from the group consisting of lower alkyl, heterocycloalkyl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, and cycloalkylamino, and heterocycloalkyl and heteroaryl are optionally substituted with one or more substituents selected from the group consisting of halogen, —CN, lower alkyl, fluoro substituted lower alkyl, lower alkoxy and fluoro substituted lower alkoxy.
  • In some embodiments of compounds of Formula I, R1 is selected from the group consisting of hydrogen, —CN, —OR7, —SR7, —NR8R7, —NR8C(O)R7, —NR8S(O)2R7, —C(O)NR8R7, —C(O)R7, —S(O)2NR8R7, —S(O)R7, —S(O)2R7, halogen, lower alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as R1 or as substituents of lower alkyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —S(O)2NH2, —C(O)NH2, —OR9, —SR9, —NR8R9, —NR8C(O)R9, —NR8S(O)2R9, —S(O)2R9, —S(O)2NR8R9, —C(O)R9, —C(O)NR8R9, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino; and R2 is selected from the group consisting of —CN, —OR7, —SR7, —NR8R7, —NR8C(O)R7, —NR8S(O)2R7, —C(O)NR8R7, —C(O)R7, —S(O)2NR8R7, —S(O)R7, —S(O)2R7, halogen, lower alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, and wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as R2 or as a substituent of lower alkyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —S(O)2NH2, —C(O)NH2, —OR9, —SR9, —NR8R9, —NR8C(O)R9, —NR8S(O)2R9, —S(O)2R9, —S(O)2NR8R9, —C(O)R9, —C(O)NR8R9, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino; wherein R7, R8 and R9 are as defined in paragraph [0011].
  • In some embodiments of compounds of Formula I, R1 is hydrogen, —CN, —NR8R7, —OR7, —S(O)2R7, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, cycloalkyl, heterocycloalkyl, aryl or heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl or heteroaryl are optionally substituted with one or more substituents selected from the group consisting of halogen, lower alkyl, fluoro substituted lower alkyl, —NR8R7, —OR7 and —S(O)2R7, and R2 is —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, —NR8R7, —OR7 or —S(O)2R7; wherein R7 and R8 are as defined in paragraph [0011].
  • In some embodiments of compounds of Formula I, R1 is hydrogen, —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower alkoxy substituted C2-6 alkoxy, aryl or heteroaryl, wherein aryl or heteroaryl are optionally substituted with one or more substituents selected from the group consisting of fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, mono-alkylamino, di-alkylamino, and cycloalkylamino, and R2 is —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, mono-alkylamino, di-alkylamino, or cycloalkylamino.
  • In one embodiment of compounds of Formula I, the compound is selected from the group consisting of:
    • 4-Butoxy-N-[3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-benzenesulfonamide (P-0007),
    • N-[3-(5-Chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-4-pyrazol-1-yl-benzenesulfonamide (P-0008),
    • N-[3-(5-Chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-4-isopropoxy-benzenesulfonamide (P-0011),
    • 4-tert-Butyl-N-[3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-benzenesulfonamide (P-0014),
    • N-[3-(5-Chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-4-propyl-benzenesulfonamide (P-0015),
    • N-{2,4-Difluoro-3-[5-(2-methoxy-ethoxy)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-isopropyl-benzenesulfonamide (P-0018),
    • N-{2,4-Difluoro-3-[5-(4-methyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-isopropyl-benzenesulfonamide (P-0019),
    • 4-Difluoromethoxy-N-{2,4-difluoro-3-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-benzenesulfonamide (P-0020),
    • N-{2,4-Difluoro-3-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-propyl-benzenesulfonamide (P-0021),
    • N-{2,4-Difluoro-3-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-isopropyl-benzenesulfonamide (P-0022),
    • N-{2,4-Difluoro-3-[5-(5-methyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-propyl-benzenesulfonamide (P-0023),
    • N-{2,4-Difluoro-3-[5-(5-methyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-trifluoromethyl-benzenesulfonamide (P-0025),
    • N-{2,4-Difluoro-3-[5-(1-methyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-propyl-benzenesulfonamide (P-0026),
    • N-{3-[5-(1,5-Dimethyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-2,4-difluoro-phenyl}-4-propyl-benzenesulfonamide (P-0027),
    • N-[2,4-Difluoro-3-(5-methyl-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-phenyl]-4-trifluoromethyl-benzenesulfonamide (P-0030),
    • N-[3-(5-Cyano-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-4-trifluoromethyl-benzenesulfonamide (P-0031),
    • (E)-3-{3-[2,6-Difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl}-acrylic acid methyl ester (P-0032),
    • 3-{3-[2,6-Difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl}-propionic acid methyl ester (P-0033),
    • 3-{3-[2,6-Difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl}-propionic acid (P-0034),
    • 3-{3-[2,6-Difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl}-N-ethyl-propionamide (P-0035) and
      all salts, prodrugs, tautomers, and isomers thereof.
  • In some embodiments, compounds have the structure according to the following Formula II:
  • Figure US20110092538A1-20110421-C00007
  • all salts, prodrugs, tautomers and isomers thereof,
  • wherein:
      • R2a is as defined for R2 for Formula I; and
      • R1 and R3 are as defined for Formula I, provided, however, the compound is not
  • Figure US20110092538A1-20110421-C00008
    Figure US20110092538A1-20110421-C00009
    Figure US20110092538A1-20110421-C00010
  • In some embodiments of compounds of Formula II, R1 is selected from the group consisting of hydrogen, halogen, optionally substituted lower alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, —CN, —OR4, —SR4, —NR5R4, —C(O)R4, —C(O)NR5R4, —S(O)2NR5R4, —NR5C(O)R4, —NR5S(O)2R4, —S(O)R4, and —S(O)2R4, and R2a is selected from the group consisting of halogen, optionally substituted lower alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, —CN, —OR4, —SR4, —NR5R4, —C(O)R4, —C(O)NR5R4, —S(O)2NR5R4, —NR5C(O)R4, —NR5S(O)2R4, —S(O)R4, and —S(O)2R4.
  • In some embodiments of compounds of Formula II, R1 is selected from the group consisting of hydrogen, halogen, lower alkyl, lower alkenyl, lower alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, —OH, —NH2, —CN, —NO2, —C(O)OH, —S(O)2NH2, —C(O)NH2, —C(S)NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —OR7, —SR7, —NR8R7, —C(O)R7, —C(S)R7, —C(O)OR7, —C(O)NR8R7, —C(S)NR8R7, —S(O)2NR8R7, —NR8C(O)R7, —NR8C(S)R7, —NR8S(O)2R7, —NR8C(O)NH2, —NR8C(O)NR8R7, —NR8C(S)NH2, —NR8C(S)NR8R7, —NR8S(O)2NH2, —NR8S(O)2NR8R7, —S(O)R7, and —S(O)2R7, wherein lower alkyl, lower alkenyl or lower alkynyl are optionally substituted with one or more substituents selected from the group consisting of fluoro, —OH, —NH2, —C(O)OH, —C(O)NH2, —OR7, —NR8R7, —C(O)OR7, —C(O)NR8R7, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as R1 or as substituents of lower alkyl, lower alkenyl or lower alkynyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —S(O)2NH2, —C(O)NH2, —OR9, —SR9, —NR8R9, —NR8C(O)R9, —NR8S(O)2R9, —S(O)2R9, —S(O)2NR8R9, —C(O)R9, —C(O)NR8R9, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino; and R2a is selected from the group consisting of halogen, lower alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, —OH, —NH2, —CN, —NO2, —C(O)OH, —S(O)2NH2, —C(O)NH2, —C(S)NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —OR7, —SR7, —NR8R7, —C(O)R7, —C(S)R7, —C(O)OR7, —C(O)NR8R7, —C(S)NR8R7, —S(O)2NR8R7, —NR8C(O)R7, —NR8C(S)R7, —NR8S(O)2R7, —NR8C(O)NH2, —NR8C(O)NR8R7, —NR8C(S)NH2, —NR8C(S)NR8R7, —NR8S(O)2NH2, —NR8S(O)2NR8R7, —S(O)R7, and —S(O)2R7, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, —OR7, —NR8R7, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as R2a or as substituents of lower alkyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —S(O)2NH2, —C(O)NH2, —OR9, —SR9, —NR8R9, —NR8C(O)R9, —NR8S(O)2R9, —S(O)2R9, —S(O)2NR8R9, —C(O)R9, —C(O)NR8R9, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino; wherein R7 is selected from the group consisting of lower alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as R7 or as substituents of lower alkyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —S(O)2NH2, —C(O)NH2, —OR9, —SR9, —NR8R9, —NR8C(O)R9, —NR8S(O)2R9, —S(O)2R9, —S(O)2NR8R9, —C(O)R9, —C(O)NR8R9, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino; R8 at each occurrence is independently hydrogen or lower alkyl; and R9 at each occurrence is independently selected from the group consisting of lower alkyl, heterocycloalkyl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, and cycloalkylamino; and heterocycloalkyl and heteroaryl are optionally substituted with one or more substituents selected from the group consisting of halogen, —CN, lower alkyl, fluoro substituted lower alkyl, lower alkoxy and fluoro substituted lower alkoxy.
  • In some embodiments of compounds of Formula II, R1 is selected from the group consisting of hydrogen, —CN, —OR7, —SR7, —NR8R7, —NR8C(O)R7, —NR8S(O)2R7, —C(O)NR8R7, —C(O)R7, —S(O)2NR8R7, —S(O)R7, —S(O)2R7, halogen, lower alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as R1 or as substituents of lower alkyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —S(O)2NH2, —C(O)NH2, —OR9, —SR9, —NR8R9, —NR8C(O)R9, —NR8S(O)2R9, —S(O)2R9, —S(O)2NR8R9, —C(O)R9, —C(O)NR8R9, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino; and R2a is selected from the group consisting of —CN, —OR7, —SR7, —NR8R7, —NR8C(O)R7, —NR8S(O)2R7, —C(O)NR8R7, —C(O)R7, —S(O)2NR8R7, —S(O)R7, —S(O)2R7, halogen, lower alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, and wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as R2a or as a substituent of lower alkyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —S(O)2NH2, —C(O)NH2, —OR9, —SR9, —NR8R9, —NR8C(O)R9, —NR8S(O)2R9, —S(O)2R9, —S(O)2NR8R9, —C(O)R9, —C(O)NR8R9, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino; wherein R7, R8 and R9 are as defined for paragraph [0011].
  • In some embodiments of compounds of Formula II, R1 is hydrogen, —CN, —NR8R7, —OR7, —S(O)2R7, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, cycloalkyl, heterocycloalkyl, aryl or heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl or heteroaryl are optionally substituted with one or more substituents selected from the group consisting of halogen, lower alkyl, fluoro substituted lower alkyl, —NR8R7, —OR7 and —S(O)2R7, and R2a is —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, —NR8R7, —OR7 or —S(O)2R7; wherein R7 and R8 are as defined in paragraph [0011].
  • In some embodiments of compounds of Formula II, R1 is hydrogen, —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower alkoxy substituted C2-6 alkoxy, aryl or heteroaryl, wherein aryl or heteroaryl are optionally substituted with one or more substituents selected from the group consisting of fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, mono-alkylamino, di-alkylamino, and cycloalkylamino, and R2a is —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, mono-alkylamino, di-alkylamino, or cycloalkylamino.
  • In one embodiment of compounds of Formula II, the compound is selected from the group consisting of:
    • 3-[3-(5-Chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenylsulfamoyl]-benzoic acid (P-0004),
    • N-[3-(5-Chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-3-difluoromethoxy-benzenesulfonamide (P-0016),
    • 3-Difluoromethoxy-N-{2,4-difluoro-3-[5-(5-methyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-benzenesulfonamide (P-0024) and
      all salts, prodrugs, tautomers, and isomers thereof.
  • In some embodiments, compounds have the structure according to the following Formula III:
  • Figure US20110092538A1-20110421-C00011
  • all salts, prodrugs, tautomers and isomers thereof,
  • wherein:
      • R1 and R3 are as defined for Formula I; and
      • R6 is optionally substituted heteroaryl, provided, however, the compound is not
  • Figure US20110092538A1-20110421-C00012
    Figure US20110092538A1-20110421-C00013
    Figure US20110092538A1-20110421-C00014
  • In some embodiments of compounds of Formula III, R1 is selected from the group consisting of hydrogen, halogen, optionally substituted lower alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, —CN, —OR4, —SR4, —NR5R4, —C(O)R4, —C(O)NR5R4, —S(O)2NR5R4, —NR5C(O)R4, —NR5S(O)2R4, —S(O)R4, and —S(O)2R4, and R6 is heteroaryl optionally substituted with one or more substituents selected from the group consisting of halogen, optionally substituted lower alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, —CN, —OR4, —SR4, —NR5R4, —C(O)R4, —C(O)NR5R4, —S(O)2NR5R4, —NR5C(O)R4, —NR5S(O)2R4, —S(O)R4, and —S(O)2R4, where R4 and R5 are as defined for Formula I.
  • In some embodiments of compounds of Formula III, R1 is selected from the group consisting of hydrogen, halogen, lower alkyl, lower alkenyl, lower alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, —OH, —NH2, —CN, —NO2, —C(O)OH, —S(O)2NH2, —C(O)NH2, —C(S)NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —OR7, —SR7, —NR8R7, —C(O)R7, —C(S)R7, —C(O)OR7, —C(O)NR8R7, —C(S)NR8R7, —S(O)2NR8R7, —NR8C(O)R7, —NR8C(S)R7, —NR8S(O)2R7, —NR8C(O)NH2, —NR8C(O)NR8R7, —NR8C(S)NH2, —NR8C(S)NR8R7, —NR8S(O)2NH2, —NR8S(O)2NR8R7, —S(O)R7, and —S(O)2R7, wherein lower alkyl, lower alkenyl or lower alkynyl are optionally substituted with one or more substituents selected from the group consisting of fluoro, —OH, —NH2, C(O)OH, —C(O)NH2, —OR7, —NR8R7, —C(O)OR7, —C(O)NR8R7, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as R1 or as substituents of lower alkyl, lower alkenyl or lower alkynyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —S(O)2NH2, —C(O)NH2, —OR9, —SR9, —NR8R9, —NR8C(O)R9, —NR8S(O)2R9, —S(O)2R9, —S(O)2NR8R9, —C(O)R9, —C(O)NR8R9, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino; and R6 is heteroaryl optionally substituted with one or more substituents selected from the group consisting of halogen, lower alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, —OH, —NH2, —CN, —NO2, —C(O)OH, —S(O)2NH2, —C(O)NH2, —C(S)NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —OR7, —SR7, —NR8R7, —C(O)R7, —C(S)R7, —C(O)OR7, —C(O)NR8R7, —C(S)NR8R7, —S(O)2NR8R7, —NR8C(O)R7, —NR8C(S)R7, —NR8S(O)2R7, —NR8C(O)NH2, —NR8C(O)NR8R7, —NR8C(S)NH2, —NR8C(S)NR8R7, —NR8S(O)2NH2, —NR8S(O)2NR8R7, —S(O)R7, and —S(O)2R7, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, —OR7, —NR8R7, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as a substituent of R6 or as a substituent of lower alkyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —S(O)2NH2, —C(O)NH2, —OR9, —SR9, —NR8R9, —NR8C(O)R9, —NR8S(O)2R9, —S(O)2R9, —S(O)2NR8R9, —C(O)R9, —C(O)NR8R9, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino; wherein R7 is selected from the group consisting of lower alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as R7 or as substituents of lower alkyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —S(O)2NH2, —C(O)NH2, —OR9, —SR9, —NR8R9, —NR8C(O)R9, —NR8S(O)2R9, —S(O)2R9, —S(O)2NR8R9, —C(O)R9, —C(O)NR8R9, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino; R8 at each occurrence is independently hydrogen or lower alkyl; and R9 at each occurrence is independently selected from the group consisting of lower alkyl, heterocycloalkyl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, and cycloalkylamino, and heterocycloalkyl and heteroaryl are optionally substituted with one or more substituents selected from the group consisting of halogen, —CN, lower alkyl, fluoro substituted lower alkyl, lower alkoxy and fluoro substituted lower alkoxy.
  • In some embodiments of compounds of Formula III, R1 is selected from the group consisting of hydrogen, —CN, —OR7, —SR7, —NR8R7, —NR8C(O)R7, —NR8S(O)2R7, —C(O)NR8R7, —C(O)R7, —S(O)2NR8R7, —S(O)R7, —S(O)2R7, halogen, lower alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as R1 or as substituents of lower alkyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —S(O)2NH2, —C(O)NH2, —OR9, —SR9, —NR8R9, —NR8C(O)R9, —NR8S(O)2R9, —S(O)2R9, —S(O)2NR8R9, —C(O)R9, —C(O)NR8R9, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino; and R6 is heteroaryl optionally substituted with one or more substituents selected from the group consisting of —CN, —OR7, —SR7, —NR8R7, —NR8C(O)R7, —NR8S(O)2R7, —C(O)NR8R7, —C(O)R7, —S(O)2NR8R7, —S(O)R7, —S(O)2R7, halogen, lower alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, and wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as a substituent of R6 or as a substituent of lower alkyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —S(O)2NH2, —C(O)NH2, —OR9, —SR9, —NR8R9, —NR8C(O)R9, —NR8S(O)2R9, —S(O)2R9, —S(O)2NR8R9, —C(O)R9, —C(O)NR8R9, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino; wherein R7, R8 and R9 are as defined for paragraph [0011].
  • In some embodiments of compounds of Formula III, R1 is hydrogen, —CN, —NR8R7, —OR7, —S(O)2R7, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, cycloalkyl, heterocycloalkyl, aryl or heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl or heteroaryl are optionally substituted with one or more substituents selected from the group consisting of halogen, lower alkyl, fluoro substituted lower alkyl, —NR8R7, —OR7 and —S(O)2R7, and R6 is heteroaryl optionally substituted with one or more of —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, —NR8R7, —OR7 or —S(O)2R7; wherein R7 and R8 are as defined in paragraph [0011].
  • In some embodiments of compounds of Formula III, R1 is hydrogen, —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower alkoxy substituted C2-6 alkoxy, aryl or heteroaryl, wherein aryl or heteroaryl are optionally substituted with one or more substituents selected from the group consisting of fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, mono-alkylamino, di-alkylamino, and cycloalkylamino, and R6 is heteroaryl optionally substituted with one or more of —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, mono-alkylamino, di-alkylamino, or cycloalkylamino.
  • In one embodiment of compounds of Formula III, the compound is selected from the group consisting of:
    • Benzo[b]thiophene-3-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide (P-0001),
    • 5-Methyl-2-trifluoromethyl-furan-3-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide (P-0002),
    • 5-Oxazol-5-yl-thiophene-2-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide (P-0003),
    • 2-Oxo-2H-chromene-6-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide (P-0005),
    • 5-Isoxazol-5-yl-thiophene-2-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide (P-0006),
    • Benzothiazole-6-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide (P-0009),
    • 1-Methyl-3-trifluoromethyl-1H-pyrazole-4-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide (P-0010),
    • Benzo[1,2,5]thiadiazoe-5-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide (P-0013),
    • 5-Methyl-benzo[b]thiophene-2-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide (P-0017),
    • 5-Methyl-thiophene-2-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide (P-0028),
    • 1-Methyl-1H-pyrazole-3-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide (P-0029),
    • Pyridine-2-sulfonic acid [2,4-difluoro-3-(5-methoxy-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-phenyl]-amide (P-0036) and
      all salts, prodrugs, tautomers, and isomers thereof.
  • In some embodiments of the above compounds, compounds are excluded where N (except where N is a heteroaryl ring atom), O, or S is bound to a carbon that is also bound to N (except where N is a heteroaryl ring atom), O, or S; or where N (except where N is a heteroaryl ring atom), O, C(S), C(O), or S(O)n (n is 0-2) is bound to an alkene carbon of an alkenyl group or bound to an alkyne carbon of an alkynyl group; accordingly, in some embodiments compounds which include linkages such as the following are excluded from the compounds provided: —NR—CH2—NR—, —O—CH2—NR—, —S—CH2—NR—, —NR—CH2—O—, —O—CH2—O—, —S—CH2—O—, —NR—CH2—S—, —O—CH2—S—, —S—CH2—S—, —NR—CH═CH—, —CH═CH—NR—, —NR—C≡C—, —C≡C—NR—, —O—CH═CH—, —CH═CH—O—, —O—C≡C—, —C≡C—O—, —S(O)0-2—CH═CH—, —CH═CH—S(O)0-2—, —S(O)0-2—C≡C—, —C≡C—S(O)0-2—, —C(O)—CH═CH—, —CH═CH—C(O)—, —C≡C—C(O)—, or —C(O)—C≡C—, —C(S)—CH═CH—, —CH═CH—C(S)—, —C≡C—C(S)—, or —C(S)—C≡C—.
  • In reference to compounds herein, unless clearly indicated to the contrary, specification of a compound or group of compounds includes pharmaceutically acceptable salts of such compound(s), prodrug(s), and all stereoisomers thereof. In reference to compositions, kits, methods of use, etc. of compounds of Formula I, Formula II, or Formula III described herein, it is understood (unless indicated otherwise) that a compound of Formula I includes all sub-embodiments thereof, a compound of Formula II includes all sub-embodiments thereof, and a compound of Formula III includes all sub-embodiments thereof.
  • In one aspect, methods are provided for treating a protein kinase mediated disease or condition in an animal subject, wherein the method involves administering to the subject an effective amount of one or more compounds of Formula I, Formula II, or Formula III. The terms “treat,” “therapy,” and like terms refer to the administration of material, e.g., one or more compound of Formula I, Formula II, or Formula III, in an amount effective to prevent, alleviate, or ameliorate one or more symptoms of a disease or condition, i.e., indication, and/or to prolong the survival of the subject being treated. The term “protein kinase mediated disease or condition” refers to a disease or condition in which the biological function of a protein kinase affects the development, course, and/or symptoms of the disease or condition, and/or in which modulation of the protein kinase alters the development, course, and/or symptoms of the disease or condition. A protein kinase mediated disease or condition includes a disease or condition for which modulation provides a therapeutic benefit, e.g. wherein treatment with protein kinase inhibitors, including compounds described herein, provides a therapeutic benefit to the subject suffering from or at risk of the disease or condition. In one aspect, the method involves administering to the subject an effective amount of a compound of Formula I, Formula II, or Formula III in combination with one or more other therapies for the disease or condition.
  • In one aspect, methods are provided for treating a protein kinase mediated disease or condition in an animal subject, wherein the method involves administering to the subject an effective amount of any one or more compound of Formula I, Formula II, or Formula III.
  • In one aspect, the invention provides methods for treating a Raf protein kinase mediated disease or condition in an animal subject, wherein the method involves administering to the subject an effective amount of one or more compound of Formula I, Formula II, or Formula III. The terms “Raf protein kinase mediated disease or condition,” “Raf mediated disease or condition,” and the like refer to a disease or condition in which the biological function of a Raf kinase, including any mutations thereof, affects the development, course, and/or symptoms of the disease or condition, and/or in which modulation of the Raf protein kinase alters the development, course, and/or symptoms of the disease or condition. The Raf protein kinase includes, but is not limited to, A-Raf, A-Raf mutations, B-Raf, mutations of B-Raf, c-Raf-1 and mutations of c-Raf-1. In some embodiments, the Raf protein kinase is B-Raf mutation V600E. In some embodiments, the Raf protein kinase is B-Raf mutation V600E/T5291. In some embodiments, the disease or condition is a cancer that is amenable to treatment by an inhibitor of the V600E mutant B-Raf. In some embodiments, the disease or condition is a cancer that is amenable to treatment by an inhibitor of the V600E/T5291 mutant B-Raf. The Raf protein kinase mediated disease or condition includes a disease or condition for which Raf inhibition provides a therapeutic benefit, e.g. wherein treatment with Raf inhibitors, including compounds described herein, provides a therapeutic benefit to the subject suffering from or at risk of the disease or condition. In one aspect, the method involves administering to the subject an effective amount of a compound of Formula I, Formula II, or Formula III in combination with one or more other therapies for the disease or condition. Similarly, the terms “A-Raf, B-Raf or c-Raf-1 protein kinase mediated disease or condition,” “A-Raf, B-Raf or c-Raf-1 mediated disease or condition,” and the like refer to a disease or condition in which the biological function of an A-Raf, B-Raf or c-Raf-1 kinase, respectively, including any mutations thereof, affects the development, course and/or symptoms of the disease or condition, and/or in which modulation of the A-Raf, B-Raf or c-Raf-1 protein kinase, respectively, alters the development, course, and/or symptoms of the disease or condition.
  • In some embodiments, a compound of Formula I, Formula II, or Formula III will have an IC50 of less than 500 nm, less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 1 nM as determined in a generally accepted kinase activity assay. In some embodiments, a compound of any of Formula I, Formula II, or Formula III will have an IC50 of less than 500 nm, less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 1 nM with respect to at least one kinase selected from the group consisting of Abl, Akt1, Akt2, Akt3, ALK, Alk5, A-Raf, B-Raf, Brk, Btk, Cdk2, CDK4, CDK5, CDK6, CHK1, c-Raf-1, Csk, EGFR, EphA1, EphA2, EphB2, EphB4, Erk2, Fak, FGFR1, FGFR2, FGFR3, FGFR4, Flt1, Flt3, Flt4, Fms, Frk, Fyn, Gsk3α, Gsk3β, HCK, Her2/Erbb2, Her4/Erbb4, IGF1R, IKK beta, Irak4, Itk, Jak1, Jak2, Jak3, Jnk1, Jnk2, Jnk3, Kdr, Kit, Lck, Lyn, MAP2K1, MAP2K2, MAP4K4, MAPKAPK2, Met, Mnk1, MLK1, p38, PDGFRA, PDGFRB, PDPK1, Pim1, Pim2, Pim3, PKC alpha, PKC beta, PKC theta, Plk1, Pyk2, Ret, ROCK1, ROCK2, Ron, Src, Stk6, Syk, TEC, Tie2, TrkA, TrkB, Yes, and Zap70, including any mutations thereof.
  • In some embodiments, a compound of Formula I, Formula II, or Formula III will have an IC50 of less than 500 nm, less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 1 nM with respect to at least one kinase selected from the group consisting of Abl, Akt 1, Akt2, Akt3, ALK, Alk5, A-Raf, B-Raf, Btk, Cdk2, CDK4, CDK5, CDK6, CHK1, c-Raf-1, Csk, EGFR, EphA1, EphA2, EphB2, EphB4, Erk2, Fak, Fms, Fyn, Gsk3α, Gsk3β, HCK, Her2/Erbb2, Her4/Erbb4, IGF1R, IKK beta, Irak4, Itk, Jak1, Jak2, Jak3, Jnk1, Jnk2, Jnk3, Kit, Lck, Lyn, MAP2K1, MAP2K2, MAP4K4, MAPKAPK2, Met, Mnk1, MLK1, p38, PDPK1, Pim1, Pim2, Pim3, PKC alpha, PKC beta, PKC theta, Plk1, Pyk2, Ron, Src, Stk6, Syk, TEC, Tie2, TrkA, TrkB, Yes, and Zap70, including any mutations thereof.
  • In some embodiments, a compound of Formula I, Formula II, or Formula III will have an IC50 of less than 500 nm, less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 1 nM with respect to at least one kinase selected from the group consisting of Abl, A-Raf, B-Raf, Btk, c-Raf-1, EGFR, EphB2, Erk2, Fak, FGFR1, Flt1, Flt3, Flt4, Fms, Irak4, Jnk1, Jnk2, Jnk3, Kdr, Kit, Lck, Lyn, MAP2K1, MAPKAP kinase 2, Met, p38, PDGFRB, Pim1, PKC theta, Pyk2, Ret, Src, Stk6, Yes, and Zap70, including any mutations thereof.
  • In some embodiments, a compound of Formula I, Formula II, or Formula III will have an IC50 of less than 500 nm, less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 1 nM with respect to at least one kinase selected from the group consisting of Abl, A-Raf, B-Raf, Btk, c-Raf-1, EGFR, EphB2, Erk2, Fak, Fms, Irak4, Jnk1, Jnk2, Jnk3, Kit, Lck, Lyn, MAP2K1, MAPKAP kinase 2, Met, p38, Pim1, PKC theta, Pyk2, Src, Stk6, Yes, and Zap70, including any mutations thereof.
  • In some embodiments, a compound of Formula I, Formula II, or Formula III will have an IC50 of less than 500 nm, less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 1 nM with respect to at least one kinase selected from the group consisting of A-Raf, B-Raf, B-Raf V600E mutant, B-Raf V600E/T529I mutant, c-Raf-1, Fak, FGFR1, FGFR2, FGFR3, FGFR4, Jnk1, Jnk2, Jnk3, Lck, Lyn, Met, Pim1, Pim2, Pim3, Pyk2, Kdr, Src and Ret, including any mutations thereof.
  • In some embodiments, a compound of Formula I, Formula II, or Formula III is an inhibitor of a Raf kinase and has an IC50 of less than 500 nm, less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 1 nM as determined in a generally accepted Raf kinase activity assay. In some embodiments, a compound of Formula I, Formula II, or Formula III will have an IC50 of less than 500 nm, less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 1 nM with respect to B-Raf, c-Raf-1, B-Raf V600E mutant, or B-Raf V600E/T529I mutant. In some embodiments, a compound of Formula I, Formula II, or Formula III will selectively inhibit one Raf kinase relative to one or more other Raf kinases. In some embodiments, the compound of Formula I, Formula II, or Formula III will selectively inhibit a mutation of the Raf kinase relative to the wild type kinase, for example B-Raf V600E mutant relative to wild type B-Raf.
  • Further to any of the above mentioned embodiments, a compound will also inhibit the effects of a mutation of the kinase, including, but not limited to, a mutation that is related to a disease state, such as a cancer. For example, B-Raf V600E mutant is present in a high percentage of some cancers, such as melanoma, and compounds will inhibit the kinase activity of this mutant.
  • Further to any of the above embodiments, a compound may selectively inhibit one kinase relative to one or more other kinases, where preferably inhibition is selective with respect to any of the other kinases, whether a kinase discussed herein, or other kinases. In some embodiments, the compound may selectively inhibit the effects of a mutation of the kinase relative to the wild type kinase, for example B-Raf V600E mutant relative to wild type B-Raf. In some embodiments, the compound may selectively inhibit Fms relative to Kit. Selective inhibition of one kinase relative to another is such that the IC50 for the one kinase may be at least about 2-fold, also 5-fold, also 10-fold, also 20-fold, also 50-fold, or at least about 100-fold less than the IC50 for any of the other kinases as determined in a generally accepted kinase activity assay.
  • In another aspect, compositions are provided that include a therapeutically effective amount of one or more compound of Formula I, Formula II, or Formula III and at least one pharmaceutically acceptable carrier, excipient, and/or diluent, including combinations of any two or more compounds of Formula I, Formula II or Formula III. The composition can further include a plurality of different pharmacologically active compounds, which can include a plurality of compounds of Formula I, Formula II, or Formula III. In another aspect, the composition can include one or more compounds of Formula I, Formula II, or Formula III along with one or more compounds that are therapeutically effective for the same disease indication. In one aspect, the composition includes one or more compounds of Formula I, Formula II, or Formula III along with one or more compounds that are therapeutically effective for the same disease indication, wherein the compounds have a synergistic effect on the disease indication. In one aspect, the composition includes one or more compounds of Formula I, Formula II, or Formula III effective in treating a cancer and one or more other compounds that are effective in treating the cancer, further wherein the compounds are synergistically effective in treating the cancer.
  • In another aspect, methods are provided for modulating the activity of a protein kinase selected from the group consisting of Abl, Akt1, Akt2, Akt3, ALK, Alk5, A-Raf, B-Raf, Brk, Btk, Cdk2, CDK4, CDK5, CDK6, CHK1, c-Raf-1, Csk, EGFR, EphA1, EphA2, EphB2, EphB4, Erk2, Fak, FGFR1, FGFR2, FGFR3, FGFR4, Flt1, Flt3, Flt4, Fms, Frk, Fyn, Gsk3α, Gsk3β, HCK, Her2/Erbb2, Her4/Erbb4, IGF1R, IKK beta, Irak4, Itk, Jak1, Jak2, Jak3, Jnk1, Jnk2, Jnk3, Kdr, Kit, Lck, Lyn, MAP2K1, MAP2K2, MAP4K4, MAPKAPK2, Met, Mnk1, MLK1, p38, PDGFRA, PDGFRB, PDPK1, Pim1, Pim2, Pim3, PKC alpha, PKC beta, PKC theta, Plk1, Pyk2, Ret, ROCK1, ROCK2, Ron, Src, Stk6, Syk, TEC, Tie2, TrkA, TrkB, Yes, and Zap70 by contacting the protein kinase with an effective amount of one or more compound of Formula I, Formula II, or Formula III.
  • In another aspect, methods are provided for treating a protein kinase mediated disease or condition in an animal subject, wherein the method involves administering to the subject an effective amount of a composition including one or more compound of Formula I, Formula II, or Formula III.
  • In one aspect, methods are provided for treating a disease or condition mediated by a protein kinase selected from the group consisting of Abl, Akt 1, Akt2, Akt3, ALK, Alk5, A-Raf, B-Raf, Btk, Cdk2, CDK4, CDK5, CDK6, CHK1, c-Raf-1, Csk, EGFR, EphA1, EphA2, EphB2, EphB4, Erk2, Fak, FGFR1, FGFR2, FGFR3, FGFR4, Flt1, Flt3, Flt4, Fms, Fyn, Gsk3α, Gsk3β, HCK, Her2/Erbb2, Her4/Erbb4, IGF1R, IKK beta, Irak4, Itk, Jak1, Jak2, Jak3, Jnk1, Jnk2, Jnk3, Kdr, Kit, Lck, Lyn, MAP2K1, MAP2K2, MAP4K4, MAPKAPK2, Met, Mnk1, MLK1, p38, PDGFRA, PDGFRB, PDPK1, Pim1, Pim2, Pim3, PKC alpha, PKC beta, PKC theta, Plk1, Pyk2, Ret, ROCK1, ROCK2, Ron, Src, Stk6, Syk, TEC, Tie2, TrkA, TrkB, Yes, and Zap70 by administering to the subject an effective amount of a composition including one or more compound of Formula I, Formula II, or Formula III.
  • In one aspect, the invention provides methods for treating a disease or condition mediated by a protein kinase selected from the group consisting of Abl, Akt 1, Akt2, Akt3, ALK, Alk5, A-Raf, B-Raf, Btk, Cdk2, CDK4, CDK5, CDK6, CHK1, c-Raf-1, Csk, EGFR, EphA1, EphA2, EphB2, EphB4, Erk2, Fak, Fms, Fyn, Gsk3α, Gsk3β, HCK, Her2/Erbb2, Her4/Erbb4, IGF1R, IKK beta, Irak4, Itk, Jak1, Jak2, Jak3, Jnk1, Jnk2, Jnk3, Kit, Lck, Lyn, MAP2K1, MAP2K2, MAP4K4, MAPKAPK2, Met, Mnk1, MLK1, p38, PDPK1, Pim1, Pim2, Pim3, PKC alpha, PKC beta, PKC theta, Plk1, Pyk2, Ron, Src, Stk6, Syk, TEC, Tie2, TrkA, TrkB, Yes, and Zap70 by administering to the subject an effective amount of a composition including one or more compound of Formula I, Formula II, or Formula III.
  • In one aspect, the invention provides methods for treating a disease or condition mediated by a protein kinase selected from the group consisting of Abl, A-Raf, B-Raf, Btk, c-Raf-1, EGFR, EphB2, Erk2, Fak, FGFR1, Flt1, Flt3, Flt4, Fms, Irak4, Jnk1, Jnk2, Jnk3, Kdr, Kit, Lck, Lyn, MAP2K1, MAPKAPK2, Met, p38, PDGFRB, Pim1, PKC theta, Pyk2, Ret, Src, Stk6, Yes, and Zap70 by administering to the subject an effective amount of a composition including one or more compound of Formula I, Formula II, or Formula III.
  • In one aspect, the invention provides methods for treating a disease or condition mediated by a protein kinase selected from the group consisting of Abl, A-Raf, B-Raf, Btk, c-Raf-1, EGFR, EphB2, Erk2, Fak, Fms, Irak4, Jnk1, Jnk2, Jnk3, Kit, Lck, Lyn, MAP2K1, MAPKAPK2, Met, p38, Pim1, PKC theta, Pyk2, Src, Stk6, Yes, and Zap70 by administering to the subject an effective amount of a composition including one or more compound of Formula I, Formula II, or Formula III.
  • In one aspect, the invention provides methods for treating a disease or condition mediated by a protein kinase selected from the group consisting of A-Raf, B-Raf, B-Raf V600E mutant, B-Raf V600E/T529I mutant, c-Raf-1, Fak, FGFR1, FGFR2, FGFR3, FGFR4, Jnk1, Jnk2, Jnk3, Lck, Lyn, Met, Pim1, Pim2, Pim3, Pyk2, Kdr, Src and Ret by administering to the subject an effective amount of a composition including one or more compound of Formula I, Formula II, or Formula III.
  • In one aspect, the invention provides methods for treating a disease or condition mediated by A-Raf, B-Raf, c-Raf-1, B-Raf V600E mutant, or B-Raf V600E/T529I mutant by administering to the subject an effective amount of a composition including one or more compound of Formula I, Formula II or Formula III. In one aspect, the invention provides methods for treating a disease or condition mediated by A-Raf, B-Raf, c-Raf-1, B-Raf V600E mutant, or B-Raf V600E/T5291 mutant by administering to the subject an effective amount of a composition including one or more compound of Formula I, Formula II, or Formula III in combination with one or more other suitable therapies for treating the disease. In one aspect, the invention provides methods for treating a cancer mediated by B-Raf V600E mutant or B-Raf V600E/T5291 mutant by administering to the subject an effective amount of a composition including one or more compound of Formula I, Formula II, or Formula III in combination with one or more suitable anticancer therapies, such as one or more chemotherapeutic drugs.
  • In one aspect, the invention provides a method of treating a cancer by administering to the subject an effective amount of a composition including one or more compound of Formula I, Formula II, or Formula III, in combination with one or more other therapies or medical procedures effective in treating the cancer. Other therapies or medical procedures include suitable anticancer therapy (e.g. drug therapy, vaccine therapy, gene therapy, photodynamic therapy) or medical procedure (e.g. surgery, radiation treatment, hyperthermia heating, bone marrow or stem cell transplant). In one aspect, the one or more suitable anticancer therapies or medical procedures is selected from treatment with a chemotherapeutic agent (e.g. chemotherapeutic drug), radiation treatment (e.g. x-ray, y-ray, or electron, proton, neutron, or a particle beam), hyperthermia heating (e.g. microwave, ultrasound, radiofrequency ablation), Vaccine therapy (e.g. AFP gene hepatocellular carcinoma vaccine, AFP adenoviral vector vaccine, AG-858, allogeneic GM-CSF-secretion breast cancer vaccine, dendritic cell peptide vaccines), gene therapy (e.g. Ad5CMV-p53 vector, adenovector encoding MDA7, adenovirus 5-tumor necrosis factor alpha), photodynamic therapy (e.g. aminolevulinic acid, motexafin lutetium), surgery, or bone marrow and stem cell transplantation.
  • In a preferred embodiment, the invention provides a method of treating a cancer by administering to the subject an effective amount of a composition including one or more compound of Formula I, Formula II, or Formula III, in combination with one or more suitable chemotherapeutic agents. In one aspect, the one or more suitable chemotherapeutic agents is selected from an alkylating agent, including, but not limited to, adozelesin, altretamine, bizelesin, busulfan, carboplatin, carboquone, carmustine, chlorambucil, cisplatin, cyclophosphamide, dacarbazine, estramustine, fotemustine, hepsulfam, ifosfamide, improsulfan, irofulven, lomustine, mechlorethamine, melphalan, oxaliplatin, piposulfan, semustine, streptozocin, temozolomide, thiotepa, and treosulfan; an antibiotic, including, but not limited to, bleomycin, dactinomycin, daunorubicin, doxorubicin, epirubicin, idarubicin, menogaril, mitomycin, mitoxantrone, neocarzinostatin, pentostatin, and plicamycin; an antimetabolite, including, but not limited to, azacitidine, capecitabine, cladribine, clofarabine, cytarabine, decitabine, floxuridine, fludarabine, 5-fluorouracil, ftorafur, gemcitabine, hydroxyurea, mercaptopurine, methotrexate, nelarabine, pemetrexed, raltitrexed, thioguanine, and trimetrexate; an immunotherapy, including, but not limited to, alemtuzumab, bevacizumab, cetuximab, galiximab, gemtuzumab, panitumumab, pertuzumab, rituximab, tositumomab, trastuzumab, and 90 Y ibritumomab tiuxetan; a hormone or hormone antagonist, including, but not limited to, anastrozole, androgens, buserelin, diethylstilbestrol, exemestane, flutamide, fulvestrant, goserelin, idoxifene, letrozole, leuprolide, magestrol, raloxifene, tamoxifen, and toremifene; a taxane, including, but not limited to, DJ-927, docetaxel, TPI 287, paclitaxel and DHA-paclitaxel; a retinoid, including, but not limited to, alitretinoin, bexarotene, fenretinide, isotretinoin, and tretinoin; an alkaloid, including, but not limited to, etoposide, homoharringtonine, teniposide, vinblastine, vincristine, vindesine, and vinorelbine; an antiangiogenic agent, including, but not limited to, AE-941 (GW786034, Neovastat), ABT-510, 2-methoxyestradiol, lenalidomide, and thalidomide; a topoisomerase inhibitor, including, but not limited to, amsacrine, edotecarin, exatecan, irinotecan (also active metabolite SN-38 (7-ethyl-10-hydroxy-camptothecin)), rubitecan, topotecan, and 9-aminocamptothecin; a kinase inhibitor, including, but not limited to, erlotinib, gefitinib, flavopiridol, imatinib mesylate, lapatinib, sorafenib, sunitinib malate, AEE-788, AG-013736, AMG 706, AMN107, BMS-354825, BMS-599626, UCN-01 (7-hydroxystaurosporine), and vatalanib; a targeted signal transduction inhibitor including, but not limited to bortezomib, geldanamycin, and rapamycin; a biological response modifier, including, but not limited to, imiquimod, interferon-α, and interleukin-2; and other chemotherapeutics, including, but not limited to 3-AP (3-amino-2-carboxyaldehyde thiosemicarbazone), aminoglutethimide, asparaginase, bryostatin-1, cilengitide, E7389, ixabepilone, procarbazine, sulindac, temsirolimus, tipifarnib. Preferably, the method of treating a cancer involves administering to the subject an effective amount of a composition including one or more compound of Formula I, Formula II, or Formula III in combination with a chemotherapeutic agent selected from 5-fluorouracil, carboplatin, dacarbazine, gefitinib, oxaliplatin, paclitaxel, SN-38, temozolomide, vinblastine, bevacizumab, cetuximab, or erlotinib.
  • In another aspect, the invention provides a method of treating or prophylaxis of a disease or condition in a mammal, by administering to the mammal a therapeutically effective amount of one or more compound of Formula I, Formula II, or Formula III, a prodrug of such compound, or a pharmaceutically acceptable salt of such compound or prodrug. The compound can be alone or can be part of a composition. In another aspect, the invention provides a method of treating or prophylaxis of a disease or condition in a mammal, by administering to the mammal a therapeutically effective amount of one or more compound of Formula I, Formula II, or Formula III, a prodrug of such compound, or a pharmaceutically acceptable salt of such compound or prodrug in combination with one or more other suitable therapies for the disease or condition.
  • In a related aspect, the invention provides kits that include a composition as described herein. In some embodiments, the composition is packaged, e.g., in a vial, bottle, flask, which may be further packaged, e.g., within a box, envelope, or bag; the composition is approved by the U.S. Food and Drug Administration or similar regulatory agency for administration to a mammal, e.g., a human; the composition is approved for administration to a mammal, e.g., a human, for a protein kinase mediated disease or condition; the invention kit includes written instructions for use and/or other indication that the composition is suitable or approved for administration to a mammal, e.g., a human, for a protein kinase-mediated disease or condition; and the composition is packaged in unit dose or single dose form, e.g., single dose pills, capsules, or the like.
  • In aspects involving treatment or prophylaxis of a disease or condition with the compounds of Formula I, Formula II, or Formula III, the disease or condition is, for example without limitation, neurologic diseases, including, but not limited to, cerebrovascular ischemia, multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease (AD), Parkinson's disease, amyotrophic lateral sclerosis, dementia, senile chorea, and Huntington's disease; neoplastic diseases and associated complications, including, but not limited to, chemotherapy-induced hypoxia, gastrointestinal stromal tumors (GISTs), prostate tumors, mast cell tumors (including canine mast cell tumors), acute myeloid myelofibrosis, leukemia, acute lymphocytic leukemia, chronic myeloid leukemia, chronic lymphocytic leukemia, multiple myeloma, melanoma, mastocytosis, gliomas, glioblastoma, astrocytoma, neuroblastoma, sarcomas (e.g. sarcomas of neuroectodermal origin, leiomyosarcoma), carcinomas (e.g. lung, breast, pancreatic, colon, hepatocellular, renal, female genital tract, squamous cell, carcinoma in situ), lymphoma (e.g. histiocytic lymphoma, non-Hodgkin's lymphoma), MEN2 syndromes, neurofibromatosis (including Schwann cell neoplasia), myelodysplastic syndrome, leukemia, tumor angiogenesis, cancers of the thyroid, liver, bone, skin, brain, central nervous system, pancreas, lung (e.g. small cell lung cancer, non small cell lung cancer), breast, colon, bladder, prostate, gastrointestinal tract, endometrium, fallopian tube, testes and ovary, and metastasis of tumors to other tissues; pain of neuropathic or inflammatory origin, including, but not limited to, acute pain, chronic pain, bone pain, cancer-related pain and migraine; cardiovascular diseases, including, but not limited to, heart failure, ischemic stroke, cardiac hypertrophy, thrombosis (e.g. thrombotic microangiopathy syndromes), atherosclerosis, reperfusion injury and ischemia (e.g. cerebrovascular ischemia, liver ischemia); inflammation including, but not limited to, age-related macular degeneration, rheumatoid arthritis, allergic rhinitis, inflammatory bowel disease (IBD), ulcerative colitis, Crohn's disease, systemic lupus erythematosis, Sjogren's Syndrome, Wegener's granulomatosis, Kawasaki's Disease, hemophagocytic syndrome (macrophage activation syndrome), multicentric reticulohistiocytosis, psoriasis, scleroderma, chronic thyroiditis, Grave's disease, myasthenia gravis, multiple sclerosis, osteoarthritis, endometriosis, scarring (e.g. dermal, tissue), vascular restenosis, fibrotic disorders, hypereosinophilia, CNS inflammation, pancreatitis, nephritis, atopic dermatitis, and hepatitis; immunodeficiency diseases, including, but not limited to, severe combined immunodeficiency (SCID), organ transplant rejection, and graft versus host disease; renal or prostatic diseases, including, but not limited to, diabetic nephropathy, polycystic kidney disease, nephrosclerosis, glomerulonephritis, interstitial nephritis, Lupus nephritis, prostate hyperplasia, chronic renal failure, tubular necrosis, diabetes-associated renal complications, and renal hypertrophy; metabolic diseases, including, but not limited to, type 1 diabetes, type 2 diabetes, metabolic syndrome, obesity, hepatic steatosis, insulin resistance, hyperglycemia, lipolysis and obesity; infection, including, but not limited to, Helicobacter pylori, Hepatitis and Influenza viruses, fever, and sepsis; pulmonary diseases, including, but not limited to, chronic obstructive pulmonary disease (COPD), acute respiratory distress syndrome (ARDS), asthma, allergy, bronchitis, emphysema, and pulmonary fibrosis; genetic developmental diseases, including, but not limited to, Noonan's syndrome, Crouzon syndrome, acrocephalo-syndactyly type I, Pfeiffer's syndrome, Jackson-Weiss syndrome, Costello syndrome, (faciocutaneoskeletal syndrome), LEOPARD syndrome, cardio-faciocutaneous syndrome (CFC) and neural crest syndrome abnormalities causing cardiovascular, skeletal, intestinal, skin, hair and endocrine diseases; disorders of bone structure, mineralization and bone reformation and resorption, including, but not limited to, osteoporosis, increased risk of fracture, Paget's disease, hypercalcemia, osteomyelitis, peri-prosthetic or wear-debris-mediated osteolysis, and metastatis of cancer to bone; Grave's disease; Hirschsprung's disease; lymphoedema; selective T-cell defect (STD); X-linked agammaglobulinemia; diabetic retinopathy; alopecia; erectile dysfunction; tuberous sclerosis, and diseases associated with muscle regeneration or degeneration, including, but not limited to, sarcopenia, muscular dystrophies (including, but not limited to, Duchenne, Becker, Emery-Dreifuss, Limb-Girdle, Facioscapulohumeral, Myotonic, Oculopharyngeal, Distal and Congenital Muscular Dystrophies), motor neuron diseases (including, but not limited to, amyotrophic lateral sclerosis, infantile progressive spinal muscular atrophy, intermediate spinal muscular atrophy, juvenile spinal muscular atrophy, spinal bulbar muscular atrophy, and adult spinal muscular atrophy), inflammatory myopathies (including, but not limited to, dermatomyositis, polymyositis, and inclusion body myositis), diseases of the neuromuscular junction (including, but not limited to, myasthenia gravis, Lambert-Eaton syndrome, and congenital myasthenic syndrome), myopathies due to endocrine abnormalities (including, but not limited to, hyperthyroid myopathy and hypothyroid myopathy) diseases of peripheral nerve (including, but not limited to, Charcot-Marie-Tooth disease, Dejerine-Sottas disease, and Friedreich's ataxia), other myopathies (including, but not limited to, myotonia congenita, paramyotonia congenita, central core disease, nemaline myopathy, myotubular myopathy, and periodic paralysis), and metabolic diseases of muscle (including, but not limited to, phosphorylase deficiency, acid maltase deficiency, phosphofructokinase deficiency, debrancher enzyme deficiency, mitochondrial myopathy, carnitine deficiency, carnitine palmatyl transferase deficiency, phosphoglycerate kinase deficiency, phosphoglycerate mutase deficiency, lactate dehydrogenase deficiency, and myoadenylate deaminase deficiency).
  • In aspects involving treatment or prophylaxis of a disease or condition with the compounds of Formula I, Formula II, or Formula III, the invention provides methods for treating an A-Raf-mediated, B-Raf-mediated and/or c-Raf-1-mediated disease or condition in an animal subject (e.g. a mammal such as a human, other primates, sports animals, animals of commercial interest such as cattle, farm animals such as horses, or pets such as dogs and cats), e.g., a disease or condition characterized by abnormal A-Raf, B-Raf, and/or c-Raf-1 activity (e.g. kinase activity). Invention methods involve administering to the subject suffering from or at risk of an A-Raf-mediated, B-Raf-mediated and/or c-Raf-1-mediated disease or condition an effective amount of compound of Formula I, Formula II or Formula III. In one embodiment, the A-Raf-mediated, B-Raf-mediated, and/or c-Raf-1-mediated disease is selected from the group consisting of neurologic diseases, including, but not limited to, multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease (AD), Parkinson's disease; neoplastic diseases including, but not limited to, melanoma, glioma, sarcoma, carcinoma (e.g. colorectal, lung, breast, pancreatic, thyroid, renal, ovarian), lymphoma (e.g. histiocytic lymphoma) neurofibromatosis, acute myeloid leukemia, myelodysplastic syndrome, leukemia, tumor angiogenesis, neuroendocrine tumors such as medullary thyroid cancer, carcinoid, small cell lung cancer and pheochromocytoma; pain of neuropathic or inflammatory origin, including, but not limited to, acute pain, chronic pain, cancer-related pain, and migraine; cardiovascular diseases including, but not limited to, heart failure, ischemic stroke, cardiac hypertrophy, thrombosis (e.g. thrombotic microangiopathy syndromes), atherosclerosis, and reperfusion injury; inflammation including, but not limited to, psoriasis, arthritis and autoimmune diseases and conditions, osteoarthritis, endometriosis, scarring, vascular restenosis, fibrotic disorders, rheumatoid arthritis, inflammatory bowel disease (IBD); immunodeficiency diseases, including, but not limited to, organ transplant rejection, graft versus host disease; renal or prostatic diseases, including, but not limited to, diabetic nephropathy, polycystic kidney disease, nephrosclerosis, glomerulonephritis, prostate hyperplasia; metabolic disorders, including, but not limited to, obesity; infection, including, but not limited to Helicobacter pylori, Hepatitis and Influenza viruses, fever, and sepsis; pulmonary diseases including, but not limited to, chronic obstructive pulmonary disease (COPD) and acute respiratory distress syndrome (ARDS); genetic developmental diseases, including, but not limited to, Noonan's syndrome, Costello syndrome, (faciocutaneoskeletal syndrome), LEOPARD syndrome, cardio-faciocutaneous syndrome (CFC), and neural crest syndrome abnormalities causing cardiovascular, skeletal, intestinal, skin, hair and endocrine diseases; and diseases associated with muscle regeneration or degeneration, including, but not limited to, sarcopenia, muscular dystrophies (including, but not limited to, Duchenne, Becker, Emery-Dreifuss, Limb-Girdle, Facioscapulohumeral, Myotonic, Oculopharyngeal, Distal and Congenital Muscular Dystrophies), motor neuron diseases (including, but not limited to, amyotrophic lateral sclerosis, infantile progressive spinal muscular atrophy, intermediate spinal muscular atrophy, juvenile spinal muscular atrophy, spinal bulbar muscular atrophy, and adult spinal muscular atrophy), inflammatory myopathies (including, but not limited to, dermatomyositis, polymyositis, and inclusion body myositis), diseases of the neuromuscular junction (including, but not limited to, myasthenia gravis, Lambert-Eaton syndrome, and congenital myasthenic syndrome), myopathies due to endocrine abnormalities (including, but not limited to, hyperthyroid myopathy and hypothyroid myopathy) diseases of peripheral nerve (including, but not limited to, Charcot-Marie-Tooth disease, Dejerine-Sottas disease, and Friedreich's ataxia), other myopathies (including, but not limited to, myotonia congenita, paramyotonia congenita, central core disease, nemaline myopathy, myotubular myopathy, and periodic paralysis), and metabolic diseases of muscle (including, but not limited to, phosphorylase deficiency, acid maltase deficiency, phosphofructokinase deficiency, debrancher enzyme deficiency, mitochondrial myopathy, carnitine deficiency, carnitine palmatyl transferase deficiency, phosphoglycerate kinase deficiency, phosphoglycerate mutase deficiency, lactate dehydrogenase deficiency, and myoadenylate deaminase deficiency).
  • In a related aspect, compounds of Formula I, Formula II, or Formula III, can be used in the preparation of a medicament for the treatment of an A-Raf-mediated, B-Raf-mediated or c-Raf-1-mediated disease or condition selected from the group consisting of neurologic diseases, including, but not limited to, multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease (AD), Parkinson's disease; neoplastic diseases including, but not limited to, melanoma, glioma, sarcoma, carcinoma (e.g. colorectal, lung, breast, pancreatic, thyroid, renal, ovarian), lymphoma (e.g. histiocytic lymphoma) neurofibromatosis, acute myeloid leukemia, myelodysplastic syndrome, leukemia, tumor angiogenesis, neuroendocrine tumors such as medullary thyroid cancer, carcinoid, small cell lung cancer and pheochromocytoma; pain of neuropathic or inflammatory origin, including, but not limited to, acute pain, chronic pain, cancer-related pain, and migraine; cardiovascular diseases, including, but not limited to, heart failure, ischemic stroke, cardiac hypertrophy, thrombosis (e.g. thrombotic microangiopathy syndromes), atherosclerosis, and reperfusion injury; inflammation including, but not limited to, psoriasis, arthritis and autoimmune diseases and conditions, osteoarthritis, endometriosis, scarring, vascular restenosis, fibrotic disorders, rheumatoid arthritis, inflammatory bowel disease (IBD); immunodeficiency diseases, including, but not limited to, organ transplant rejection, graft versus host disease; renal or prostatic diseases, including, but not limited to, diabetic nephropathy, polycystic kidney disease, nephrosclerosis, glomerulonephritis, prostate hyperplasia; metabolic disorders, including, but not limited to, obesity; infection, including, but not limited to, Helicobacter pylori, Hepatitis and Influenza viruses, fever, and sepsis; pulmonary diseases, including, but not limited to, chronic obstructive pulmonary disease (COPD) and acute respiratory distress syndrome (ARDS); genetic developmental diseases, including, but not limited to, Noonan's syndrome, Costello syndrome, (faciocutaneoskeletal syndrome), LEOPARD syndrome, cardio-faciocutaneous syndrome (CFC), and neural crest syndrome abnormalities causing cardiovascular, skeletal, intestinal, skin, hair and endocrine diseases; and diseases associated with muscle regeneration or degeneration, including, but not limited to, sarcopenia, muscular dystrophies (including, but not limited to, Duchenne, Becker, Emery-Dreifuss, Limb-Girdle, Facioscapulohumeral, Myotonic, Oculopharyngeal, Distal and Congenital Muscular Dystrophies), motor neuron diseases (including, but not limited to, amyotrophic lateral sclerosis, infantile progressive spinal muscular atrophy, intermediate spinal muscular atrophy, juvenile spinal muscular atrophy, spinal bulbar muscular atrophy, and adult spinal muscular atrophy), inflammatory myopathies (including, but not limited to, dermatomyositis, polymyositis, and inclusion body myositis), diseases of the neuromuscular junction (including, but not limited to, myasthenia gravis, Lambert-Eaton syndrome, and congenital myasthenic syndrome), myopathies due to endocrine abnormalities (including, but not limited to, hyperthyroid myopathy and hypothyroid myopathy) diseases of peripheral nerve (including, but not limited to, Charcot-Marie-Tooth disease, Dejerine-Sottas disease, and Friedreich's ataxia), other myopathies (including, but not limited to, myotonia congenita, paramyotonia congenita, central core disease, nemaline myopathy, myotubular myopathy, and periodic paralysis), and metabolic diseases of muscle (including, but not limited to, phosphorylase deficiency, acid maltase deficiency, phosphofructokinase deficiency, debrancher enzyme deficiency, mitochondrial myopathy, carnitine deficiency, carnitine palmatyl transferase deficiency, phosphoglycerate kinase deficiency, phosphoglycerate mutase deficiency, lactate dehydrogenase deficiency, and myoadenylate deaminase deficiency).
  • The compounds of Formula I, Formula II, or Formula III with kinase activity IC50 less than 10 μM as determined in a standard assay described herein can be used to treat protein kinase mediated diseases and conditions related to the following protein kinases, including any mutations thereof, for example without limitation:
      • Abl, related to chronic myeloid leukemia (CML), acute lymphoblastic leukemia (ALL) and acute myelogenous leukemia (AML);
      • Akt1, related to gastric, prostate, colorectal, ovarian, pancreatic and breast cancer, glioblastoma and leukemia, as well as schizophrenia and bipolar disorders, and also use in combination with other chemotherapeutic drugs;
      • Akt2, related to hyperglycemia due to peripheral insulin resistance and nonsuppressible hepatic glucose production accompanied by inadequate compensatory hyperinsulinemia, also related to pancreatic, ovarian and breast cancer;
      • Akt3, related to melanoma, prostate and breast cancer;
      • ALK, related to non-Hodgkin lymphomas such as diffuse large B-cell lymphoma and anaplastic large cell lymphoma;
      • Alk5, related to pancreatic and biliary cancers, and cutaneous T-cell lymphoma;
      • A-Raf, related to neurologic diseases such as multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease (AD), Parkinson's disease; neoplastic diseases including, but not limited to, melanoma, glioma, sarcoma, carcinoma (e.g. colorectal, lung, breast, pancreatic, thyroid, renal, ovarian), lymphoma (e.g. histiocytic lymphoma), neurofibromatosis, myelodysplastic syndrome, leukemia, tumor angiogenesis; pain of neuropathic or inflammatory origin, including acute pain, chronic pain, cancer-related pain and migraine; and diseases associated with muscle regeneration or degeneration, including, but not limited to, vascular restenosis, sarcopenia, muscular dystrophies (including, but not limited to, Duchenne, Becker, Emery-Dreifuss, Limb-Girdle, Facioscapulohumeral, Myotonic, Oculopharyngeal, Distal and Congenital Muscular Dystrophies), motor neuron diseases (including, but not limited to, amyotrophic lateral sclerosis, infantile progressive spinal muscular atrophy, intermediate spinal muscular atrophy, juvenile spinal muscular atrophy, spinal bulbar muscular atrophy, and adult spinal muscular atrophy), inflammatory myopathies (including, but not limited to, dermatomyositis, polymyositis, and inclusion body myositis), diseases of the neuromuscular junction (including, but not limited to, myasthenia gravis, Lambert-Eaton syndrome, and congenital myasthenic syndrome), myopathies due to endocrine abnormalities (including, but not limited to, hyperthyroid myopathy and hypothyroid myopathy) diseases of peripheral nerve (including, but not limited to, Charcot-Marie-Tooth disease, Dejerine-Sottas disease, and Friedreich's ataxia), other myopathies (including, but not limited to, myotonia congenita, paramyotonia congenita, central core disease, nemaline myopathy, myotubular myopathy, and periodic paralysis), and metabolic diseases of muscle (including, but not limited to, phosphorylase deficiency, acid maltase deficiency, phosphofructokinase deficiency, debrancher enzyme deficiency, mitochondrial myopathy, carnitine deficiency, carnitine palmatyl transferase deficiency, phosphoglycerate kinase deficiency, phosphoglycerate mutase deficiency, lactate dehydrogenase deficiency, and myoadenylate deaminase deficiency).
      • B-Raf or c-Raf-1, related to neurologic diseases, including, but not limited to, as multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease (AD), Parkinson's disease; neoplastic diseases including, but not limited to, melanoma, glioma, sarcoma, carcinoma (e.g. colorectal, lung, breast, pancreatic, thyroid, renal, ovarian), lymphoma (e.g. histiocytic lymphoma) neurofibromatosis, acute myeloid leukemia, myelodysplastic syndrome, leukemia, tumor angiogenesis, neuroendocrine tumors such as medullary thyroid cancer, carcinoid, small cell lung cancer and pheochromocytoma; pain of neuropathic or inflammatory origin, including, but not limited to, acute pain, chronic pain, cancer-related pain, and migraine; cardiovascular diseases including, but not limited to, heart failure, ischemic stroke, cardiac hypertrophy, thrombosis (e.g. thrombotic microangiopathy syndromes), atherosclerosis, reperfusion injury; inflammation including, but not limited to, psoriasis, arthritis and autoimmune diseases and conditions, osteoarthritis, endometriosis, scarring, vascular restenosis, fibrotic disorders, rheumatoid arthritis, inflammatory bowel disease (IBD); immunodeficiency diseases, including, but not limited to, organ transplant rejection, graft versus host disease; renal or prostatic diseases, including, but not limited to, diabetic nephropathy, polycystic kidney disease, nephrosclerosis, glomerulonephritis, prostate hyperplasia; metabolic disorders, including, but not limited to, obesity; infection, including, but not limited to, Helicobacter pylori, Hepatitis and Influenza viruses, fever, and sepsis; pulmonary diseases including, but not limited to, chronic obstructive pulmonary disease (COPD) and acute respiratory distress syndrome (ARDS); genetic developmental diseases, including, but not limited to, Noonan's syndrome, Costello syndrome, (faciocutaneoskeletal syndrome), LEOPARD syndrome, cardio-faciocutaneous syndrome (CFC), and neural crest syndrome abnormalities causing cardiovascular, skeletal, intestinal, skin, hair and endocrine diseases;
      • Brk, related to breast and colon cancer, and head and neck squamous cell carcinoma;
      • Btk, related to X-linked agammaglobulinemia, acute lymphocytic leukemia, autoimmune diseases such as multiple sclerosis, systemic lupus erythematosis, rheumatoid arthritis, and Graves' disease, immune suppression in organ transplant, and drug sensitivity of B-lineage cells;
      • Cdk2, related to prostate, breast, colorectal and ovarian cancer;
      • Cdk4, related to glioblastoma (e.g. glioblastoma multiforme), anaplastic astrocytoma, and breast cancer;
      • Cdk5, related to Alzheimer's disease, amyotrophic lateral sclerosis and Lewy body disease;
      • Cdk6, related to glioblastoma multiforme, non-Hodgkin's lymphoma, splenic marginal zone lymphoma, T-cell lymphoblastic lymphoma (T-LBL) and T-cell acute lymphoblastic leukemia (T-ALL);
      • CHK1, related to DNA damage repair, sensitizes cells to chemotherapeutic agents;
      • Csk, related to colon and pancreatic carcinomas and autoimmune pathology such as type 1 diabetes, rheumatoid arthritis and systemic lupus erythematosus;
      • EGFR, related to breast, colorectal, bladder, prostate and non small cell lung cancer, squamous cell carcinomas of the head and neck cancer, oral cavity, and esophagus, and glioblastoma multiforme;
      • EphA1, related to head and neck squamous cell carcinoma, hepatoma and lung cancer;
      • EphA2, related to aberrant short-range contact-mediated axonal guidance, bladder, breast, prostate, colon, skin, cervical, ovarian, pancreatic and lung cancers, and metastatic melanoma;
      • EphB2, related to angiogenesis disorder (e.g. ocular angiogenesis disease such as retinopathy), and cancer (e.g. glioblastoma, breast and liver cancer);
      • EphB4, related to colorectal cancer (CRC), head and neck squamous cell carcinoma, and tumours of the prostate, breast, endometrium, and bladder;
      • Erk2, related to aberrant proliferation, differentiation, transcription regulation and development, and may be useful in treating inflammation, for example inflammation associated with Lyme neuroborreliosis, and in treating cancers, such as gastric cancer;
      • Fak, related to colon and breast tumors, and is also related to esophageal squamous cell carcinoma, melanoma, anaplastic astrocytoma, glioblastoma, ductal carcinoma in situ, prostate and hepatocellular carcinoma, and tumor metastases, and may also provide synergistic effects when used with other chemotherapeutic drugs;
      • FGFR1, related to 8p11 myeloproliferative syndrome;
      • FGFR2, related to Crouzon Syndrome, Jackson-Weiss Syndrome, Apert Syndrome, craniosynostosis, Pfeiffer Syndrome, acrocephalo syndactyly type V, and Beare-Stevenson Cutis Gyrata Syndrome;
      • FGFR3, related to angiogenesis, wound healing, achondroplasia, Muenke craniosynostosis, Crouzon syndrome, acanthosis nigricans, thanatophoric dysplasia, bladder carcinomas, and multiple myeloma;
      • FGFR4, related to cancer of the breast, lung, colon, medullary thyroid, pancreas, ovary, prostate, endometrium, and fallopian tube, head and neck squamous cell carcinomas and leiomyosarcoma;
      • Flt1, related to non-small cell lung carcinoma, prostate carcinoma, and colorectal cancer;
      • Flt3, related to acute myeloid leukemia, myelodysplastic syndrome, acute lymphoblastic leukemia;
      • Flt4, related to primary lymphoedema;
      • Fms, related to immune disorders, including, but not limited to, rheumatoid arthritis, systemic lupus erythematosis (SLE), and transplant rejection; inflammatory diseases including, but not limited to, osteoarthritis, inflammatory bowel syndrome, ulcerative colitis, Crohn's disease, chronic obstructive pulmonary disease (COPD), emphysema, Kawasaki's Disease, hemophagocytic syndrome (macrophage activation syndrome), multicentric reticulohistiocytosis, and atherosclerosis; metabolic disorders, including, but not limited to, Type I diabetes, Type II diabetes, insulin resistance, hyperglycemia, obesity, and lipolysis; disorders of bone structure, mineralization and bone formation and resorption, including, but not limited to, osteoporosis, increased risk of fracture, Paget's disease, hypercalcemia, infection-mediated osteolysis (e.g. osteomyelitis), peri-prosthetic or wear-debris-mediated osteolysis, and metastasis of cancer to bone; kidney and genitourinary diseases, including, but not limited to, endometriosis, nephritis (e.g. glomerulonephritis, interstitial nephritis, Lupus nephritis), tubular necrosis, diabetes-associated renal complications (e.g. diabetic nephropathy), and renal hypertrophy; disorders of the central nervous system, including, but not limited to, multiple sclerosis, stroke, Alzheimer's disease and Parkinson's disease; inflammatory and chronic pain, including, but not limited to, bone pain; and cancers, including, but not limited to, multiple myeloma, acute myeloid leukemia (AML), chronic myeloid leukemia (CML), prostate cancer, breast cancer, ovarian cancer, melanoma, glioblastoma multiforme, metastasis of tumors to other tissues, and other chronic myeloproliferative diseases such as myelofibrosis;
      • Frk, related to acute myeloid leukemia and type 1 diabetes;
      • Fyn, related to Alzheimer's disease, schizophrenia and prevention of metastases, e.g. in melanoma and squamous cell carcinoma;
      • GSK3 (Gsk3α and/or Gsk3β), related to CNS disorders such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, diabetes type II, bipolar disorders, stroke, cancer, chronic inflammatory disease, leucopenia, schizophrenia, chronic pain, neuropathic pain, and traumatic head injury;
      • HCK, related to chronic myelogenous leukemia and acute lymphocytic leukemia;
      • Her2/Erbb2, related to prostate and breast cancer;
      • Her4/Erbb4, related to childhood medulloblastoma;
      • IGF1R, related to prostate cancer, hepatocellular carcinoma;
      • IKK beta, related to leukemia of T-cells, necrosis, insulin resistance, and malignant neoplasms;
      • Irak4, related to bacterial infections, immunodeficiency syndrome, Crohn's disease, ulcerative colitis, asthma, chronic bronchitis, cardio hypertrophy, and kidney hypertension;
      • Itk, related to allergic asthma;
      • Jak1, related to Hepatitis C virus infection;
      • Jak2, related to myeloproliferative disorders such as polycythaemia vera, myelofibrosis, essential thrombocythemia, myeloid metaplasia and leukemias, including acute lymphoblastic leukemia, chronic neutrophilic leukemia, juvenile myelomonocytic leukemia, CMML, Philadelphia chromosome-negative CML, megakaryocytic leukemia, and acute erythroid leukemia;
      • Jak3, related to X-linked severe combined immunodeficiency, myeloproliferative disorders, transplant rejection and autoimmune diseases such as rheumatoid arthritis, inflammatory bowel syndrome, Crohn's disease, systemic lupus erythematosis, ulcerative colitis, psoriasis and multiple sclerosis;
      • Jnk (Jnk1, Jnk2, Jnk3), related to metabolic diseases including type 1 diabetes, type 2 diabetes, metabolic syndrome, obesity, and hepatic steatosis; cardiovascular diseases such as atherosclerosis, ischemia (e.g. cerebrovascular ischemia, liver ischemia), reperfusion injury, cardiac hypertrophy; renal diseases such as chronic renal failure; neoplastic diseases and associated complications, including chemotherapy-induced hypoxia, prostate tumors, myeloid leukemia and cancers of the liver, bone, skin, brain, pancreas, lung breast, colon, prostate and ovary; transplant rejection; pain of neuropathic or inflammatory origin including acute and chronic pain; inflammatory and autoimmune diseases including age-related macular degeneration, rheumatoid arthritis, inflammatory bowel disease, ulcerative colitis, Crohn's disease, systemic lupus erythematosis, Sjogren's Syndrome, psoriasis, scleroderma, chronic thyroiditis, Grave's disease, myasthenia gravis, and multiple sclerosis, and inflammation in other organs including CNS inflammation, pancreatitis, nephritis, atopic dermatitis, and hepatitis; airway inflammatory diseases such as asthma, allergy, bronchitis, pulmonary fibrosis, chronic obstructive pulmonary disease; neurologic diseases such as stroke, cerebrovascular ischemia, neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, dementia, senile chorea, head and spinal cord trauma, and Huntington's disease. More particularly, Jnk1 is related to type 1 diabetes, type 2 diabetes, metabolic syndrome, obesity and hepatic steatosis, Jnk2 is related to atherosclerosis, and Jnk3 is related to inflammatory diseases including autoimmune diseases such as rheumatoid arthritis, inflammatory bowel syndrome, Crohn's disease, systemic lupus erythematosis, Sjogren's Syndrome, psoriasis and multiple sclerosis, airway inflammatory diseases such as asthma, allergy, pulmonary fibrosis, and chronic obstructive pulmonary disease, and inflammation in other organs, such as CNS inflammation, pancreatitis, nephritis, and hepatitis; neurologic diseases such as stroke, cerebrovascular ischemia, and neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, and Huntington's disease; and neoplastic diseases such as prostate tumors and myeloid leukemia;
      • Kdr, related to anti-angiogenesis for treating solid tumor growth (e.g. ovarian, lung, breast, prancreatic, prostate, colon, gastrointestinal stromal tumor, non small cell lung cancer, and epidermoid cancer), metastasis, psoriasis, rheumatoid arthritis, diabetic retinopathy and age related macular degeneration;
      • Kit, related to malignancies, including mast cell tumors, small cell lung cancer, testicular cancer, gastrointestinal stromal tumors (GISTs), glioblastoma, astrocytoma, neuroblastoma, carcinomas of the female genital tract, sarcomas of neuroectodermal origin, colorectal carcinoma, carcinoma in situ, Schwann cell neoplasia associated with neurofibromatosis, acute myelocytic leukemia, acute lymphocytic leukemia, chronic myelogenous leukemia, mastocytosis, melanoma, and canine mast cell tumors, and inflammatory diseases, including asthma, rheumatoid arthritis, allergic rhinitis, multiple sclerosis, inflammatory bowel syndrome, transplant rejection, and hypereosinophilia;
      • Lck, related to acute lymphoblastic leukemia, T-cell lymphoma, lymphopenia, renal carcinoma, colon carcinoma, severe combined immunodeficiency, multiple sclerosis, inflammatory bowel and type I diabetes;
      • Lyn, related to
      • MAP2K1, related to acute myeloid leukemia, breast, ovarian and liver cancer;
      • MAP2K2, related to cancer and inflammation;
      • MAP4K4, related to metabolic indications, including re-sensitizing fat and muscle cells to insulin, ameliorating the pathology in adipocytes, ameliorating the pathology in muscle cells, metabolic syndrome, and type II diabetes; a broad range of oncology indications, including blocking the migration, invasion and metastasis in many different tumor types; and T-cell mediated autoimmune diseases;
      • MAPKAPK2, cancer (e.g. prostate, breast), stroke, menengitis, and inflammatory disorders;
      • Met, related to kidney, breast, bladder, non-small-cell lung, colorectal, and bladder cancers, and hepatocellular carcinoma;
      • Mnk1, related to conditions associated with heat shock, nutrient deprivation, oxidative or osmotic stress, and infection of mammalian cells (e.g. with viruses such as adenovirus (Ad) or influenza virus), and autoimmune diseases;
      • MLK1, related to neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease, and inflammatory disorders;
      • p38, related to acute coronary syndrome, stroke, atherosclerosis, and inflammatory autoimmune diseases such as rheumatoid arthritis, inflammatory bowel disease, and Crohn's disease;
      • PDGFR (PDGFRA, PDGFRB), related to idiopathic hypereosinophilic syndrome, chronic eosinophilic leukemia, glioma, gastrointestinal stromal tumors (GISTs), juvenile myelomonocytic leukemia, metastatic medulloblastoma, atherogenesis, and restenosis. More particularly, PDGFRA related to idiopathic hypereosinophilic syndrome, chronic eosinophilic leukemia, glioma, gastrointestinal stromal tumors (GISTs), juvenile myelomonocytic leukemia, metastatic medulloblastoma, atherogenesis, and restenosis, and PDGFRB related to idiopathic hypereosinophilic syndrome, chronic eosinophilic leukemia, juvenile myelomonocytic leukemia, and metastatic medulloblastoma;
      • PDPK1, related to cancer and diabetes;
      • Pim1, related to cancers such as hematopoietic (e.g. acute myeloid and acute lymphoid leukemias) and prostate cancers, and non-Hodgkin's lymphomas;
      • Pim2, related to lymphomas;
      • Pim3, related to hepatocellular carcinoma;
      • PKC alpha, related to pituitary tumors and prefrontal cortical dysfunction such as distractibility, impaired judgment, impulsivity, and thought disorder, also may be used to sensitize chemotherapy in breast, colon, and non small cell lung cancers;
      • PKC beta, related to diabetic retinopathy;
      • PKC-theta, related to insulin resistance, T-cell lymphoma;
      • Plk1, related to cancers (e.g. lymphoma of the thyroid, non-Hodgkin's lymphomas, colorectal cancers, leukemias and melanoma), also useful as sensitizer in chemotherapy;
      • Pyk2, related to inflammation (e.g. osteoporosis, polycystic kidney disease, rheumatoid arthritis and inflammatory bowel disease), CNS disease (e.g. Parkinson's disease and Alzheimer's disease), stroke and cancers (e.g. gliomas, breast cancer, and pancreatic cancer);
      • Ret, related to cancer of the thyroid, neuroblastoma, familial medullary thyroid carcinoma (FMTC), multiple endocrine neoplasia type IIA and IIB (MEN2A, MEN2B), and neurodegenerative disorders (e.g. Hirschsprung's disease, Parkinson's disease, Alzheimer's disease, and amyotrophic lateral sclerosis);
      • ROCK (ROCK-1, ROCK-2), related to cancers (e.g. ovarian cancer, hepatocellular carcinoma, pancreatic cancer), ocular disease (e.g. glaucoma), cardiac hypertrophy, improved renal perfusion, transplant rejection, and acute respiratory distress syndrome;
      • Ron, related to cancer and inflammation;
      • Src, related to cancer and osteoporosis;
      • Stk6, related to gastric, bladder, breast, lung, CNS, ovarian, kidney, colon, prostate, pancreas, and cervical cancers, melanoma, leukemia, and neuroblastoma;
      • Syk, related to lymphomas (e.g. mantle cell lymphoma);
      • TEC, related to sepsis, septic shock, inflammation, rheumatoid arthritis, Crohn's disease, irritable bowel disease (IBD), and ulcerative colitis;
      • Tie2 (TEK), related to cancer, arthritis (e.g. rheumatoid arthritis), and atherosclerosis;
      • TrkA, related to pain (e.g. chronic pain, neuropathic pain), cancer (e.g. prostate cancer, lung cancer, pancreatic cancer), allergic disorders (e.g. asthma), arthritis, diabetic retinopathy, macular degeneration and psoriasis;
      • TrkB, related to obesity, hyperphagia, developmental delays, cancer (e.g. prostate cancer, lung cancer, Wilms tumors, neuroblastoma, pancreatic cancer), various neuropathies (e.g. stroke, multiple sclerosis, transverse myelitis, and encephalitis), and diabetes.
      • Yes, related to various cancers including esophageal squamous cell carcinoma; and
      • Zap70, related to AIDS, systemic lupus erythematosus, myasthenia gravis, atherosclerosis, rejection of transplanted organs or tissues, allograft rejection including acute and chronic allograft rejection, graft versus host disease, rheumathoid arthritis, psoriasis, systemic sclerosis, atopic dermatitis, eczematous dermatitis, alopecia, and inflammation of the nasal mucus membrane, including all forms of rhinitis.
  • Additional aspects and embodiments will be apparent from the following Detailed Description and from the claims.
  • DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
  • As used herein the following definitions apply unless clearly indicated otherwise:
  • “Halogen” refer to all halogens, that is, chloro (Cl), fluoro (F), bromo (Br), or iodo (I).
  • “Hydroxyl” or “hydroxy” refer to the group —OH.
  • “Thiol” refers to the group —SH.
  • “Lower alkyl” alone or in combination means an alkane-derived radical containing from 1 to 6 carbon atoms (unless specifically defined) that includes a straight chain alkyl or branched alkyl. The straight chain or branched alkyl group is attached at any available point to produce a stable compound. In many embodiments, a lower alkyl is a straight or branched alkyl group containing from 1-6, 1-4, or 1-2, carbon atoms, such as methyl, ethyl, propyl, isopropyl, butyl, t-butyl, and the like. A “substituted lower alkyl” denotes lower alkyl that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, attached at any available atom to produce a stable compound, wherein the substituents are selected from the group consisting of —F, —OH, —NH2, —NO2, —CN, —C(O)OH, —C(S)OH, —C(O)NH2, —C(S)NH2, —S(O)2NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —C(NH)NH2, —ORo, —SRo, —OC(O)Ro, —OC(S)Ro, —C(O)Ro, —C(S)Ro, —C(O)ORo, —C(S)ORo, —S(O)Ro, —S(O)2Ro, —C(O)NHRo, —C(S)NHRo, —C(O)NRoRo, —C(S)NRoRo, —S(O)2NHRo, —S(O)2NRoRo, —C(NH)NHRo, —C(NH)NRpRc, —NHC(O)Ro, —NHC(S)Ro, —NRoC(O)Ro, —NRoC(S)Ro, —NHS(O)2Ro, —NRoS(O)2Ro, —NHC(O)NHRo, —NHC(S)NHRo, —NRoC(O)NH2, —NRoC(S)NH2, —NRoC(O)NHRo, —NRoC(S)NHRo, —NHC(O)NRoRo, —NHC(S)NRoRo, —NRoC(O)NRoRo, —NRoC(S)NRoRo, —NHS(O)2NHRo, —NRoS(O)2NH2, —NRoS(O)2NHRo, —NHS(O)2NRoRo, —NRoS(O)2NRoRo, —NHRo, —NRoRo, —Re, —Rf, and —Rg. Furthermore, possible substitutions include subsets of these substitutions, such as are indicated herein, for example, in the description of compounds of Formula I, Formula II, or Formula III, attached at any available atom to produce a stable compound. For example “fluoro substituted lower alkyl” denotes a lower alkyl group substituted with one or more fluoro atoms, such as perfluoroalkyl, where preferably the lower alkyl is substituted with 1, 2, 3, 4 or 5 fluoro atoms, also 1, 2, or 3 fluoro atoms. While it is understood that substitutions are attached at any available atom to produce a stable compound, when optionally substituted alkyl is an R group of a moiety such as —OR (e.g. alkoxy), —SR (e.g. thioalkyl), —NHR (e.g. alkylamino), —C(O)NHR, and the like, substitution of the alkyl R group is such that substitution of the alkyl carbon bound to any O, S, or N of the moiety (except where N is a heteroaryl ring atom) excludes substituents that would result in any O, S, or N of the substituent (except where N is a heteroaryl ring atom) being bound to the alkyl carbon bound to any O, S, or N of the moiety. “C2-6 alkyl” denotes lower alkyl containing 2-6 carbon atoms. A “substituted C2-6 alkyl” denotes optionally substituted lower alkyl containing 2-6 carbon atoms. A “substituted methyl” denotes methyl that is independently substituted, unless indicated otherwise, with 1, 2, or 3 substituents, wherein the substituents are selected as per optionally substituted lower alkyl.
  • “C1-3 alkylene” refers to a divalent alkane-derived radical containing 1-3 carbon atoms, straight chain or branched, from which two hydrogen atoms are taken from the same carbon atom or from different carbon atoms. C1-3 alkylene includes methylene —CH2—, ethylene —CH2CH2—, propylene —CH2CH2CH2—, and isopropylene —CH(CH3)CH2— or —CH2CH(CH3)—. C1-3 alkylene substituted with one or more substituents indicates C1-3 alkylene that is independently substituted, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents as indicated, attached at any available atom to produce a stable compound.
  • “Lower alkenyl” alone or in combination means a straight or branched hydrocarbon containing 2-6 carbon atoms (unless specifically defined) and at least one, preferably 1-3, more preferably 1-2, most preferably one, carbon to carbon double bond. Carbon to carbon double bonds may be either contained within a straight chain or branched portion. Examples of lower alkenyl groups include ethenyl, propenyl, isopropenyl, butenyl, and the like. A “substituted lower alkenyl” denotes lower alkenyl that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, attached at any available atom to produce a stable compound, wherein the substituents are selected from the group consisting of —F, —OH, —NH2, —NO2, —CN, —C(O)OH, —C(S)OH, —C(O)NH2, —C(S)NH2, —S(O)2NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —C(NH)NH2, —ORo, —SRo, —OC(O)Ro, —OC(S)Ro, —C(O)Ro, —C(S)Ro, —C(O)ORo, —C(S)ORo, —S(O)Ro, —S(O)2Ro, —C(O)NHRo, —C(S)NHRo, —C(O)NRoRo, —C(S)NRoRo, —S(O)2NHRo, —S(O)2NRoRo, —C(NH)NHRo, —C(NH)NRpRc, —NHC(O)Ro, —NHC(S)Ro, —NRoC(O)Ro, —NRoC(S)Ro, —NHS(O)2Ro, —NRoS(O)2Ro, —NHC(O)NHRo, —NHC(S)NHRo, —NRoC(O)NH2, —NRoC(S)NH2, —NRoC(O)NHR °, —NRoC(S)NHRo, —NHC(O)NRoRo, —NHC(S)NRoRo, —NRoC(O)NRoRo, —NRoC(S)NR ° Ro, —NHS(O)2NHRo, —NRoS(O)2NH2, —NRoS(O)2NHRo, —NHS(O)2NRoRo, —NRoS(O)2NRoRo, —NHRo, —NRoRo, —Rd, —Rf, and —Rg. Further, possible substitutions include subsets of these substitutions, such as are indicated herein, for example, in the description of compounds of Formula I, Formula II, or Formula III, attached at any available atom to produce a stable compound. For example “fluoro substituted lower alkenyl” denotes a lower alkenyl group substituted with one or more fluoro atoms, where preferably the lower alkenyl is substituted with 1, 2, 3, 4 or 5 fluoro atoms, also 1, 2, or 3 fluoro atoms. While it is understood that substitutions are attached at any available atom to produce a stable compound, substitution of alkenyl groups are such that —F, —C(O)—, —C(S)—, —C(NH)—, —S(O)—, —S(O)2—, —O—, —S—, or N (except where N is a heteroaryl ring atom), are not bound to an alkene carbon thereof. Further, where alkenyl is a substituent of another moiety or an R group of a moiety such as —OR, —NHR, —C(O)R, and the like, substitution of the moiety is such that any —C(O)—, —C(S)—, —S(O)—, —S(O)2—, —O—, —S—, or N thereof (except where N is a heteroaryl ring atom) are not bound to an alkene carbon of the alkenyl substituent or R group. Further, where alkenyl is a substituent of another moiety or an R group of a moiety such as —OR, —NHR, —C(O)NHR, and the like, substitution of the alkenyl R group is such that substitution of the alkenyl carbon bound to any O, S, or N of the moiety (except where N is a heteroaryl ring atom) excludes substituents that would result in any O, S, or N of the substituent (except where N is a heteroaryl ring atom) being bound to the alkenyl carbon bound to any O, S, or N of the moiety. An “alkenyl carbon” refers to any carbon within an alkenyl group, whether saturated or part of the carbon to carbon double bond. An “alkene carbon” refers to a carbon within an alkenyl group that is part of a carbon to carbon double bond.
  • “Lower alkynyl” alone or in combination means a straight or branched hydrocarbon containing 2-6 carbon atoms (unless specifically defined) containing at least one, preferably one, carbon to carbon triple bond. Examples of alkynyl groups include ethynyl, propynyl, butynyl, and the like. A “substituted lower alkynyl” denotes lower alkynyl that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, attached at any available atom to produce a stable compound, wherein the substituents are selected from the group consisting of —F, —OH, —NH2, —NO2, —CN, —C(O)OH, —C(S)OH, —C(O)NH2, —C(S)NH2, —S(O)2NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —C(NH)NH2, —ORo, —SRo, —OC(O)Ro, —OC(S)Ro, —C(O)Ro, —C(S)Ro, —C(O)ORo, —C(S)ORo, —S(O)Ro, —S(O)2Ro, —C(O)NHRo, —C(S)NHRo, —C(O)NRoRo, —C(S)NRoRo, —S(O)2NHRo, —S(O)2NRoRo, —C(NH)NHRo, —C(NH)NRpRc, —NHC(O)Ro, —NHC(S)Ro, —NRoC(O)Ro, —NRoC(S)Ro, —NHS(O)2Ro, —NRoS(O)2Ro, —NHC(O)NHRo, —NHC(S)NHRo, —NRoC(O)NH2, —NRoC(S)NH2, —NRoC(O)NHRo, —NRoC(S)NHRo, —NHC(O)NRoRo, —NHC(S)NRoRo, —NRoC(O)NRoRo, —NRoC(S)NRoRo, —NHS(O)2NHRo, —NRoS(O)2NH2, —NRoS(O)2NHRo, —NHS(O)2NRoRo, —NRoS(O)2NRoRo, —NHRo, —NRoRo, —Rd, —Re, and —Rg. Further, possible substitutions include subsets of these substitutions, such as are indicated herein, for example, in the description of compounds of Formula I, Formula II, or Formula III, attached at any available atom to produce a stable compound. For example “fluoro substituted lower alkynyl” denotes a lower alkynyl group substituted with one or more fluoro atoms, where preferably the lower alkynyl is substituted with 1, 2, 3, 4 or 5 fluoro atoms, also 1, 2, or 3 fluoro atoms. While it is understood that substitutions are attached at any available atom to produce a stable compound, substitution of alkynyl groups are such that —F, —C(O)—, —C(S)—, —C(NH)—, —S(O)—, —S(O)2—, —O—, —S—, or N (except where N is a heteroaryl ring atom) are not bound to an alkyne carbon thereof. Further, where alkynyl is a substituent of another moiety or an R group of a moiety such as —OR—, —NHR—, —C(O)R, and the like, substitution of the moiety is such that any —C(O)—, —C(S)—, —S(O)—, —S(O)2—, —O—, —S—, or N thereof (except where N is a heteroaryl ring atom) are not bound to an alkyne carbon of the alkynyl substituent or R group. Further, where alkynyl is a substituent of another moiety or an R group of a moiety such as —OR, —NHR, —C(O)NHR, and the like, substitution of the alkynyl R group is such that substitution of the alkynyl carbon bound to any O, S, or N of the moiety (except where N is a heteroaryl ring atom) excludes substituents that would result in any O, S, or N of the substituent (except where N is a heteroaryl ring atom) being bound to the alkynyl carbon bound to any O, S, or N of the moiety. An “alkynyl carbon” refers to any carbon within an alkynyl group, whether saturated or part of the carbon to carbon triple bond. An “alkyne carbon” refers to a carbon within an alkynyl group that is part of a carbon to carbon triple bond.
  • “Cycloalkyl” refers to saturated or unsaturated, non-aromatic monocyclic, bicyclic or tricyclic carbon ring systems of 3-10, also 3-8, more preferably 3-6, ring members per ring, such as cyclopropyl, cyclopentyl, cyclohexyl, adamantyl, and the like. A “substituted cycloalkyl” is a cycloalkyl that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, attached at any available atom to produce a stable compound, wherein the substituents are selected from the group consisting of halogen, —OH, —NH2, —NO2, —CN, —C(O)OH, —C(S)OH, —C(O)NH2, —C(S)NH2, —S(O)2NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —C(NH)NH2, —ORo, —SRo, —OC(O)Ro, —OC(S)Ro, —C(O)Ro, —C(S)Ro, —C(O)ORo, —C(S)ORo, —S(O)Ro, —S(O)2Ro, —C(O)NHRo, —C(S)NHRo, —C(O)NRoRo, —C(S)NRoRo, —S(O)2NHRo, —S(O)2NRoRo, —C(NH)NHRo, —C(NH)NRoRo, —NHC(O)Ro, —NHC(S)Ro, —NRoC(O)Ro, —NRoC(S)Ro, —NHS(O)2Ro, —NRoS(O)2Ro, —NHC(O)NHRo, —NHC(S)NHRo, —NRoC(O)NH2, —NRoC(S)NH2, —NRoC(O)NHRo, —NRoC(S)NHRo, —NHC(O)NRoRo, —NHC(S)NRoRo, —NRoC(O)NRoRo, —NRoC(S)NRoRo, —NHS(O)2NHRo, —NRoS(O)2NH2, —NRoS(O)2NHRo, —NHS(O)2NRoRo, —NRoS(O)2NRoRo, —NHRo, —NRoRo, —Rd, —Re, —Rf, and —Rg.
  • “Heterocycloalkyl” refers to a saturated or unsaturated non-aromatic cycloalkyl group having from 5 to 10 atoms in which from 1 to 3 carbon atoms in the ring are replaced by heteroatoms of O, S or N, and are optionally fused with benzo or heteroaryl of 5-6 ring members. Heterocycloalkyl is also intended to include oxidized S or N, such as sulfinyl, sulfonyl and N-oxide of a tertiary ring nitrogen. Heterocycloalkyl is also intended to include compounds in which a ring carbon may be oxo substituted, i.e. the ring carbon is a carbonyl group, such as lactones and lactams. The point of attachment of the heterocycloalkyl ring is at a carbon or nitrogen atom such that a stable ring is retained. Examples of heterocycloalkyl groups include, but are not limited to, morpholino, tetrahydrofuranyl, dihydropyridinyl, piperidinyl, pyrrolidinyl, pyrrolidonyl, piperazinyl, dihydrobenzofuryl, and dihydroindolyl. A “substituted heterocycloalkyl” is a heterocycloalkyl that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, attached at any available atom to produce a stable compound, wherein the substituents are selected from the group consisting of halogen, —OH, —NH2, —NO2, —CN, —C(O)OH, —C(S)OH, —C(O)NH2, —C(S)NH2, —S(O)2NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —C(NH)NH2, —ORo, —SRo, —OC(O)Ro, —OC(S)Ro, —C(O)Ro, —C(S)Ro, —C(O)ORo, —C(S)ORo, —S(O)Ro, —S(O)2Ro, —C(O)NHRo, —C(S)NHRo, —C(O)NRoRo, —C(S)NRoRo, —S(O)2NHRo, —S(O)2NRoRo, —C(NH)NHRo, —C(NH)NRpRc, —NHC(O)Ro, —NHC(S)Ro, —NRoC(O)Ro, —NRoC(S)Ro, —NHS(O)2Ro, —NRoS(O)2Ro, —NHC(O)NHRo, —NHC(S)NHRo, —NRoC(O)NH2, —NRoC(S)NH2, —NRoC(O)NHRo, —NRoC(S)NHRo, —NHC(O)NRoRo, —NHC(S)NRoRo, —NRoC(O)NRoRo, —NRoC(S)NRoRo, —NHS(O)2NHRo, —NRoS(O)2NH2, —NRoS(O)2NHRo, —NHS(O)2NRoRo, —NRoS(O)2NRoRo, —NHRo, —NRoRo, —Rd, —Re, —Rf, and —Rg.
  • “Aryl” alone or in combination refers to a monocyclic or bicyclic ring system containing aromatic hydrocarbons such as phenyl or naphthyl, which may be optionally fused with a cycloalkyl of preferably 5-7, more preferably 5-6, ring members. “Arylene” is a divalent aryl. A “substituted aryl” is an aryl that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, attached at any available atom to produce a stable compound, wherein the substituents are selected from the group consisting of halogen, —OH, —NH2, —NO2, —CN, —C(O)OH, —C(S)OH, —C(O)NH2, —C(S)NH2, —S(O)2NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —C(NH)NH2, —ORo, —SRo, —OC(O)Ro, —OC(S)Ro, —C(O)Ro, —C(S)Ro, —C(O)ORo, —C(S)ORo, —S(O)Ro, —S(O)2Ro, —C(O)NHRo, —C(S)NHRo, —C(O)NRoRo, —C(S)NRoRo, —S(O)2NHRo, —S(O)2NRoRo, —C(NH)NHRo, —C(NH)NRpRc, —NHC(O)Ro, —NHC(S)Ro, —NRoC(O)Ro, —NRoC(S)Ro, —NHS(O)2Ro, —NRoS(O)2Ro, —NHC(O)NHRo, —NHC(S)NHRo, —NRoC(O)NH2, —NRoC(S)NH2, —NRoC(O)NHRo, —NRoC(S)NHRo, —NHC(O)NRoRo, —NHC(S)NRoRo, —NRoC(O)NRoRo, —NRoC(S)NRoRo, —NHS(O)2NHRo, —NRoS(O)2NH2, —NRoS(O)2NHRo, —NHS(O)2NRoRo, —NRoS(O)2NRoRo, —NHRo, —NRoRo, —Rd, —Re, —Rf, and —Rg. A “substituted arylene” is a divalent substituted aryl.
  • “Heteroaryl” alone or in combination refers to a monocyclic aromatic ring structure containing 5 or 6 ring atoms, or a bicyclic aromatic group having 8 to 10 atoms, containing one or more, preferably 1-4, more preferably 1-3, even more preferably 1-2, heteroatoms independently selected from the group consisting of O, S, and N. Heteroaryl is also intended to include oxidized S or N, such as sulfinyl, sulfonyl and N-oxide of a tertiary ring nitrogen. A carbon or nitrogen atom is the point of attachment of the heteroaryl ring structure such that a stable compound is produced. Examples of heteroaryl groups include, but are not limited to, pyridinyl, pyridazinyl, pyrazinyl, quinaoxalyl, indolizinyl, benzo[b]thienyl, quinazolinyl, purinyl, indolyl, quinolinyl, pyrimidinyl, pyrrolyl, pyrazolyl, oxazolyl, thiazolyl, thienyl, isoxazolyl, oxathiadiazolyl, isothiazolyl, tetrazolyl, imidazolyl, triazolyl, furanyl, benzofuryl, and indolyl. “Nitrogen containing heteroaryl” refers to heteroaryl wherein any heteroatoms are N. “Heteroarylene” is a divalent heteroaryl. A “substituted heteroaryl” is a heteroaryl that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, attached at any available atom to produce a stable compound, wherein the substituents are selected from the group consisting of halogen, —OH, —NH2, —NO2, —CN, —C(O)OH, —C(S)OH, —C(O)NH2, —C(S)NH2, —S(O)2NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —C(NH)NH2, —ORo, —SRo, —OC(O)Ro, —OC(S)Ro, —C(O)Ro, —C(S)Ro, —C(O)ORo, —C(S)ORo, —S(O)Ro, —S(O)2Ro, —C(O)NHRo, —C(S)NHRo, —C(O)NRoRo, —C(S)NRoRo, —S(O)2NHRo, —S(O)2NRoRo, —C(NH)NHRo, —C(NH)NRpRc, —NHC(O)Ro, —NHC(S)Ro, —NRoC(O)Ro, —NRoC(S)Ro, —NHS(O)2Ro, —NRoS(O)2Ro, —NHC(O)NHRo, —NHC(S)NHRo, —NRoC(O)NH2, —NRoC(S)NH2, —NRoC(O)NHRo, —NRoC(S)NHRo, —NHC(O)NRoRo, —NHC(S)NRoRo, —NRoC(O)NRoRo, —NRoC(S)NRoRo, —NHS(O)2NHRo, —NRoS(O)2NH2, —NRoS(O)2NHRo, —NHS(O)2NRoRo, —NRoS(O)2NRoRo, —NHRo, —NRoRo, —Rd, —Re, —Rf, and —Rg. “Substituted heteroarylene” is a divalent substituted heteroaryl.
  • The variables Ro, Rp, Re, Rd, Re, Rf and Rg as used in the description of optional substituents for alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl are defined as follows:
  • each Ro, Rp, and Rc are independently selected from the group consisting of Rd, Re, Rf, and Rg, or Rp and Rc combine with the nitrogen to which they are attached to form a 5-7 membered heterocycloalkyl or a 5 or 7 membered nitrogen containing heteroaryl, wherein the 5-7 membered heterocycloalkyl or 5 or 7 membered nitrogen containing heteroaryl are optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents selected from the group consisting of halogen, —NO2, —CN, —OH, —NH2, —ORu, —SRu, —NHRu, —NRuRu, —Rx, and —Ry;
    each Rd is independently lower alkyl, wherein lower alkyl is optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2 or 3 substituents selected from the group consisting of fluoro, —OH, —NH2, —NO2, —CN, —C(O)OH, —C(S)OH, —C(O)NH2, —C(S)NH2, —S(O)2NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —C(NH)NH2, —ORk, —SRk, —OC(O)Rk, —OC(S)Rk, —C(O)Rk, —C(S)Rk, —C(O)ORk, —C(S)ORk, —S(O)Rk, —S(O)2Rk, —C(O)NHRk, —C(S)NHRk, —C(O)NRkRk, —C(S)NRkRk, —S(O)2NHRk, —S(O)2NRkRk, —C(NH)NHRk, —C(NH)NRmRn, —NHC(O)Rk, —NHC(S)Rk, —NRkC(O)Rk, —NRkC(S)Rk, —NHS(O)2Rk, —NRkS(O)2Rk, —NHC(O)NHRk, —NHC(S)NHRk, —NRkC(O)NH2, —NRkC(S)NH2, —NRkC(O)NHRk, —NRkC(S)NHRk, —NHC(O)NRkRk, —NHC(S)NRkRk, —NRkC(O)NH2, —NRkC(S)NRkRk, —NHS(O)2NHRk, —NRkS(O)2NH2, —NRkS(O)2NHRk, —NHS(O)2NRkRk, —NRkS(O)2NRkRk, —NHRk, —NRkRk, —Ri, and —Rj;
    each Re is independently lower alkenyl, wherein lower alkenyl is optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2 or 3 substituents selected from the group consisting of fluoro, —OH, —NH2, —NO2, —CN, —C(O)OH, —C(S)OH, —C(O)NH2, —C(S)NH2, —S(O)2NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —C(NH)NH2, —ORk, —SRk, —OC(O)Rk, —OC(S)Rk, —C(O)Rk, —C(S)Rk, —C(O)ORk, —C(S)ORk, —S(O)Rk, —S(O)2Rk, —C(O)NHRk, —C(S)NHRk, —C(O)NRkRk, —C(S)NRkRk, —S(O)2NHRk, —S(O)2NRkRk, —C(NH)NHRk, —C(NH)NRmRn, —NHC(O)Rk, —NHC(S)Rk, —NRkC(O)Rk, —NRkC(S)Rk, —NHS(O)2Rk, —NRkS(O)2Rk, —NHC(O)NHRk, —NHC(S)NHRk, —NRkC(O)NH2, —NRkC(S)NH2, —NRkC(O)NHRk, —NRkC(S)NHRk, —NHC(O)NRkRk, —NHC(S)NRkRk, —NRkC(O)NRkRk, —NRkC(S)NRkRk, —NHS(O)2NHRk, —NRkS(O)2NH2, —NRkS(O)2NHRk, —NHS(O)2NRkRk, —NRkS(O)2NRkRk, —NHRk, —NRkRk, —Rh, and —Rj;
    each Rf is independently lower alkynyl, wherein lower alkynyl is optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2 or 3 substituents selected from the group consisting of fluoro, —OH, —NH2, —NO2, —CN, —C(O)OH, —C(S)OH, —C(O)NH2, —C(S)NH2, —S(O)2NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —C(NH)NH2, —ORk, —SRk, —OC(O)Rk, —OC(S)Rk, —C(O)Rk, —C(S)Rk, —C(O)ORk, —C(S)ORk, —S(O)Rk, —S(O)2Rk, —C(O)NHRk, —C(S)NHRk, —C(O)NRkRk, —C(S)NRkRk, —S(O)2NHRk, —S(O)2NRkRk, —C(NH)NHRk, —C(NH)NRmRn, —NHC(O)Rk, —NHC(S)Rk, —NRkC(O)Rk, —NRkC(S)Rk, —NHS(O)2Rk, —NRkS(O)2Rk, —NHC(O)NHRk, —NHC(S)NHRk, —NRkC(O)NH2, —NRkC(S)NH2, —NRkC(O)NHRk, —NRkC(S)NHRk, —NHC(O)NRkRk, —NHC(S)NRkRk, —NRkRk, —NRkC(S)NRkRk, —NHS(O)2NHRk, —NRkS(O)2NH2, —NRkS(O)2NHRk, —NHS(O)2NRkRk, —NRkS(O)2NRkRk, —NHRk, —NRkRk, —Rh, and —Rj;
    each Rg is independently selected from the group consisting of cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl are optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2 or 3 substituents selected from the group consisting of halogen, —OH, —NH2, —NO2, —CN, —C(O)OH, —C(S)OH, —C(O)NH2, —C(S)NH2, —S(O)2NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —C(NH)NH2, —ORk, —SRk, —OC(O)Rk, —OC(S)Rk, —C(O)Rk, —C(S)Rk, —C(O)ORk, —C(S)ORk, —S(O)Rk, —S(O)2Rk, —C(O)NHRk, —C(S)NHRk, —C(O)NRkRk, —C(S)NRkRk, —S(O)2NHRk, —S(O)2NRkRk, —C(NH)NHRk, —C(NH)NRmRn, —NHC(O)Rk, —NHC(S)Rk, —NRkC(O)Rk, —NRkC(S)Rk, —NHS(O)2Rk, —NRkS(O)2Rk, —NHC(O)NHRk, —NHC(S)NHRk, —NRkC(O)NH2, —NRkC(S)NH2, —NRkC(O)NHRk, —NRkC(S)NHRk, —NHC(O)NRkRk, —NHC(S)NRkRk, —NRkC(O)NRkRk, —NRkC(S)NRkRk, —NHS(O)2NHRk, —NRkS(O)2NH2, —NRkS(O)2NHRk, —NHS(O)2NRkRk, —NRkS(O)2NRkRk, —NHRk, —NRkRk, —Rh, —Ri, and —Rj;
      • wherein Rk, Rm, and Rn at each occurrence are independently selected from the group consisting of Rh, Ri, and Rj, or Rm and Rn combine with the nitrogen to which they are attached form a 5-7 membered heterocycloalkyl or a 5 or 7 membered nitrogen containing heteroaryl, wherein the 5-7 membered heterocycloalkyl or 5 or 7 membered nitrogen containing heteroaryl are optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents selected from the group consisting of halogen, —NO2, —CN, —OH, —NH2, ORu, —SRu, —NHRu, —NRuRu, —Rx, and —Ry;
      • wherein each Rh is independently lower alkyl optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents selected from the group consisting of fluoro, —OH, —NH2, —NO2, —CN, —C(O)OH, —C(S)OH, —C(O)NH2, —C(S)NH2, —S(O)2NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —C(NH)NH2, —ORr, —SRr, —OC(O)Rr, —OC(S)Rr, —C(O)Rr, —C(S)Rr, —C(O)ORr, —C(S)ORr, —S(O)Rr, —S(O)2Rr, —C(O)NHRr, —C(S)NHRr, —C(O)NRrRr, —C(S)NRrRr, —S(O)2NHRr, —S(O)2NRrRr, —C(NH)NHRr, —C(NH)NRsRt, —NHC(O)Rr, —NRC(S)Rr, —NRrC(O)Rr, —NRrC(S)Rr, —NHS(O)2Rr, —NRrS(O)2Rr, —NHC(O)NHRr, —NHC(S)NHRr, —NRrC(O)NH2, —NRrC(S)NH2, —NRrC(O)NHRr, —NRrC(S)NHRr, —NHC(O)NRrRr, —NHC(S)NRrRr, —NRrC(O)NRrRr, —NRrC(S)NRrRr, —NHS(O)2NHRr, —NRrS(O)2NH2, —NRrS(O)2NHRr, —NHS(O)2NRrRr, —NRrS(O)2NRrRr, —NHRr, —NRrRr, —Ri, and —Rj;
      • wherein each Ri is independently selected from the group consisting of lower alkenyl and lower alkynyl, wherein lower alkenyl or lower alkynyl are optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2 or 3 substituents selected from the group consisting of fluoro, —OH, —NH2, —NO2, —CN, —C(O)OH, —C(S)OH, —C(O)NH2, —C(S)NH2, —S(O)2NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —C(NH)NH2, —ORr, —SRr, —OC(O)Rr, —OC(S)Rr, —C(O)Rr, —C(S)Rr, —C(O)ORr, —C(S)ORr, —S(O)Rr, —S(O)2Rr, —C(O)NHRr, —C(S)NHRr, —C(O)NRrRr, —C(S)NRrRr, —S(O)2NHRr, —S(O)2NRrRr, —C(NH)NHRr, —C(NH)NRsRt, —NHC(O)Rr, —NRC(S)Rr, —NRrC(O)Rr, —NRrC(S)Rr, —NHS(O)2Rr, —NRrS(O)2Rr, —NHC(O)NHRr, —NHC(S)NHRr, —NRrC(O)NH2, —NRrC(S)NH2, —NRrC(O)NHRr, —NRrC(S)NHRr, —NHC(O)NRrRr, —NHC(S)NRrRr, —NRrC(O)NRrRr, —NRrC(S)NRrRr, —NHS(O)2NHRr, —NRrS(O)2NH2, —NRrS(O)2NHRr, —NHS(O)2NRrRr, —NRrS(O)2NRrRr, —NHRr, —NRrRr, and —Rj;
      • wherein each Rj is independently selected from the group consisting of cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl are optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2 or 3 substituents selected from the group consisting of halogen, —OH, —NH2, —NO2, —CN, —C(O)OH, —C(S)OH, —C(O)NH2, —C(S)NH2, —S(O)2NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —C(NH)NH2, —ORr, —SRr, —OC(O)Rr, —OC(S)Rr, —C(O)Rr, —C(S)Rr, —C(O)ORr, —C(S)ORr, —S(O)Rr, —S(O)2Rr, —C(O)NHRr, —C(S)NHRr, —C(O)NRrRr, —C(S)NRrRr, —S(O)2NHRr, —S(O)2NRrRr, —C(NH)NHRr, —C(NH)NRsRt, —NHC(O)Rr, —NRC(S)Rr, —NRrC(O)Rr, —NRrC(S)Rr, —NHS(O)2Rr, —NRrS(O)2Rr, —NHC(O)NHRr, —NHC(S)NHRr, —NRrC(O)NH2, —NRrC(S)NH2, —NRrC(O)NHRr, —NRrC(S)NHRr, —NHC(O)NRrRr, —NHC(S)NRrRr, —NRrC(O)NRrRr, —NRrC(S)NRrRr, —NHS(O)2NHRr, —NRrS(O)2NH2, —NRrS(O)2NHRr, —NHS(O)2NRrRr, —NRrS(O)2NRrRr, —NHRr, —NRrRr, cycloalkylamino, and —Rx;
      • wherein each Rr, Rs, and Rt at each occurrence are independently selected from the group consisting of lower alkyl, C3-6 alkenyl, C3-6 alkynyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein lower alkyl is optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents selected from the group consisting of —Ry, fluoro, —OH, —NH2, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, and cycloalkylamino, provided, however, that any substitution of the lower alkyl carbon bound to any O, S, or N, of —ORr, —SRr, —C(O)ORr, —C(S)ORr, —C(O)NHRr, —C(S)NHRr, —C(O)NRrRr, —C(S)NRrRr, —S(O)2NHRr, —S(O)2NRrRr, —C(NH)NHRr, —NRrC(O)Rr, —NRrC(S)Rr, —NRrS(O)2Rr, —NHC(O)NHRr, —NHC(S)NHRr, —NRrC(O)NH2, —NRrC(S)NH2, —NRrC(O)NHRr, —NRrC(S)NHRr, —NHC(O)NRrRr, —NHC(S)NRrRr, —NRrC(O)NRrRr, —NRrC(S)NRrRr, —NHS(O)2NHRr, —NRrS(O)2NH2, —NRrS(O)2NHRr, —NHS(O)2NRrRr, —NRrS(O)2NRrRr, —NHRr, or —NRrRr is selected from the group consisting of fluoro and —RY, and wherein C3-6 alkenyl or C3-6 alkynyl are optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents selected from the group consisting of —Ry, fluoro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, and cycloalkylamino, provided, however, that any substitution of the C3-6 alkenyl or C3-6 alkynyl carbon bound to any O, S, or N, of —ORr, —SW, —C(O)ORr, —C(S)ORr, —C(O)NHRr, —C(S)NHRr, —C(O)NRrRr, —C(S)NRrRr, —S(O)2NHRr, —S(O)2NRrRr, —C(NH)NHRr, —NRrC(O)Rr, —NRrC(S)Rr, —NRrS(O)O, —NHC(O)NHRr, —NHC(S)NHRr, —NRrC(O)NH2, —NRrC(S)NH2, —NRrC(O)NHRr, —NRrC(S)NHRr, —NHC(O)NRrRr, —NHC(S)NRrRr, —NRrC(O)NRrRr, —NRrC(S)NRrRr, —NHS(O)2NHRr, —NRrS(O)2NH2, —NRrS(O)2NHRr, —NHS(O)2NRrRr, —NRrS(O)2NRrRr, —NHRr, or —NRrRr is selected from the group consisting of fluoro, lower alkyl, fluoro substituted lower alkyl, and —Ry, and wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl are optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents selected from the group consisting of halogen, —OH, —NH2, —NO2, —CN, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, and cycloalkylamino, or Rs and Rt combine with the nitrogen to which they are attached form a 5-7 membered heterocycloalkyl or a 5 or 7 membered nitrogen containing heteroaryl, wherein the 5-7 membered heterocycloalkyl or 5 or 7 membered nitrogen containing heteroaryl are optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents selected from the group consisting of halogen, —NO2, —CN, —OH, —NH2, ORu, —SRu, —NHRu, —NRuZu, —Rx, and —Ry;
      • wherein each Ru is independently selected from the group consisting of lower alkyl, C3-6 alkenyl, C3-6 alkynyl, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein lower alkyl is optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents selected from the group consisting of —Ry, fluoro, —OH, —NH2, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, and cycloalkylamino, provided, however, that any substitution of the lower alkyl carbon bound to the O of —ORu, S of —SRu, or N of —NHRu is fluoro or —Ry, and wherein C3-6 alkenyl or C3-6 alkynyl are optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents selected from the group consisting of —Ry, fluoro, —OH, —NH2, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, and cycloalkylamino, provided, however, that any substitution of the C3-6 alkenyl or C3-6 alkynyl carbon bound to the O of —ORu, S of —SRu, or N of —NHRu is fluoro, lower alkyl, fluoro substituted lower alkyl, or —Ry, and wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl are optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents selected from the group consisting of halogen, —OH, —NH2, —NO2, —CN, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, and cycloalkylamino;
      • wherein each Rx is selected from the group consisting of lower alkyl, lower alkenyl and lower alkynyl, wherein lower alkyl is optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents selected from the group consisting of —Ry, fluoro, —OH, —NH2, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, and cycloalkylamino, and wherein lower alkenyl or lower alkynyl are optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents selected from the group consisting of —Ry, fluoro, —OH, —NH2, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, and cycloalkylamino;
      • wherein each Ry is selected from the group consisting of cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl are optionally substituted with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents selected from the group consisting of halogen, —OH, —NH2, —NO2, —CN, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, and cycloalkylamino.
  • In some embodiments, all occurrences of optionally substituted lower alkyl, optionally substituted C2-6 alkyl, optionally substituted lower alkenyl, or optionally substituted lower alkynyl are optionally substituted with one or more, also 1, 2 or 3 groups or substituents selected from the group consisting of fluoro, —NO2, —CN, —OR1a, —SR1a, —NR1aR1a, —OC(O)R1a, —OC(S)R1a, —C(O)R1a, —C(S)R1a, —C(O)OR1a, —C(S)OR1a, —C(O)NR1aR1a, —C(S)NR1aR1a, —S(O)2NR1aR1a, —C(NH)NR1aR1a, —NR1aC(O)R1a, —NR1aC(S)R1a, —NR1aS(O)2R1a, —NR1aC(O) NR1a, —NR1aC(S)NR1aR1a, —NR1aS(O)2NR1aR1a, —S(O)R1a, —S(O)2R1a, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl and heteroaryl are optionally substituted with one or more, also 1, 2 or 3 groups or substituents selected from the group consisting of halogen, —NO2, —CN, —ORla, —SR1a, —NR1aR1a, —OC(O)R1a, —OC(S)R1a, —C(O)R1a, —C(S)R1a, —C(O)OR1a, —C(S)OR1a, —C(O)NR1aR1a, —C(S)NR1aR1a, —S(O)2NR1aR1a, —C(NH)NR1aR1a, —NR1aC(O)R1aNR1aC(S)R1a, —NR1aS(O)2R1a, —NR1aC(O)NR1aR1a, —NR1aC(S)NR1aNR1aS(O)2NR1aR1aS(O)R1aS(O)2R1a, —R1b, and lower alkyl optionally substituted with one or more, also 1, 2 or 3 groups or substituents selected from the group consisting of fluoro, —OH, —NH2, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, and —R1b, and all occurrences of optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted 5-7 membered heterocycloalkyl, optionally substituted aryl, optionally substituted arylene, optionally substituted heteroaryl, optionally substituted heteroarylene, or optionally substituted 5 or 7 membered nitrogen containing heteroaryl are optionally substituted with one or more, also 1, 2, or 3 groups or substituents selected from the group consisting of halogen, —NO2, —CN, —OR1a, —SR1a, —NR1aR1a, —OC(O)R1a, —OC(S)R1a, —C(O)R1a, —C(S)R1a, —C(O)OR1a, —C(S)OR1a, —C(O)NR1a, —C(S)NR1aR1a, —S(O)2NR1aR1a, —C(NH)NR1aR1a, —NR1aC(O)R1a, —NR1aC(S)R1a, —NR1aS(O)2R1a, —NR1aC(O)NR1aR1a, —NR1aC(S)NR1aR1a, —NR1aS(O)2NR1aR1a, —S(O)R1a, —S(O)2R1a, —R1b, and lower alkyl optionally substituted with one or more, also 1, 2 or 3 groups or substituents selected from the group consisting of fluoro, —OH, —NH2, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, and —R1b, wherein R1a is selected from the group consisting of hydrogen, provided, however, that hydrogen is not bound to any of C(S), C(O), S(O), or S(O)2 of —OC(O)R1a, —OC(S)R1a, —C(O)R1a, —C(S)R1a, —NR1aC(O)R1a, —NR1aC(S)R1a, —NR1aS(O)2R1a, —S(O)R1a, or —S(O)2R1a, —R1b, and lower alkyl optionally substituted with one or more, also 1, 2 or 3 groups or substituents selected from the group consisting of fluoro, —OH, —NH2, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, and —R1b, provided, however, that any substitution of the alkyl carbon bound to O, S, or N of −OR1a, —SR1a, —NR1aR1a, —C(O)OR1a, —C(S)OR1a, —C(O)NR1aR1a, —C(S)NR1aR1a, —S(O)2NR1aR1a, —C(NH)NR1aR1a, —NR1aC(O)R1a, —NR1aC(S)R1a, —NR1aS(O)2R1a, —NR1aC(O)NR1aR1a(S)NR1aR1a, or —NR1aS(O)2NR1aR1a, is fluoro or —R1b, and wherein —R1b is selected from the group consisting of cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl and heteroaryl are optionally substituted with one or more, also 1, 2 or 3 groups or substituents selected from the group consisting of halogen, —CN, —OH, —NH2, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, and cycloalkylamino.
  • In some embodiments, all occurrences of optionally substituted lower alkyl, optionally substituted C2-6 alkyl, optionally substituted lower alkenyl, or optionally substituted lower alkynyl are optionally substituted with one or more, also 1, 2 or 3 groups or substituents selected from the group consisting of fluoro, —CN, —OR1a, —SR1a, —NR1aR1a, —C(O)R1a, —C(S)R1a, —C(O)OR1a, —C(O)NR1aR1a, —C(S)NR1aR1a, —S(O)2NR1aR1a, —NR1aC(O)R1a, —NR1aC(S)R1a, —NR1aS(O)2R1a, —S(O)R1a, —S(O)2R1a, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl and heteroaryl are optionally substituted with one or more, also 1, 2 or 3 groups or substituents selected from the group consisting of halogen, —CN, —OR1a, —SR1a, —NR1aR1a, —C(O)R1a, —C(S)R1a, —C(O)OR1a, —C(O)NR1aR1a, —C(S)NR1aR1a, —S(O)2NR1aR1a, —NR1aC(O)R1a, —NR1aC(S)R1a, —NR1aS(O)2R1a, —S(O)R1a, —S(O)2R1a, —R1b, and lower alkyl optionally substituted with one or more, also 1, 2 or 3 groups or substituents selected from the group consisting of fluoro, —OH, —NH2, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, and —R1b, and all occurrences of optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted 5-7 membered heterocycloalkyl, optionally substituted aryl, optionally substituted arylene, optionally substituted heteroaryl, optionally substituted heteroarylene, or optionally substituted 5 or 7 membered nitrogen containing heteroaryl are optionally substituted with one or more, also 1, 2, or 3 groups or substituents selected from the group consisting of halogen, —CN, —OR1a, —SR1a, —NR1aR1a, —C(O)R1a, —C(S)R1a, —C(O)OR1a, —C(O)NR1aR1a, —C(S)NR1aR1a, —S(O)2NR1aR1a, —NR1aC(O)R1a, —NR1aC(S)R1a, —NR1aS(O)2R1a, —S(O)R1a, —S(O)2R1a, —R1b, and lower alkyl optionally substituted with one or more, also 1, 2 or 3 groups or substituents selected from the group consisting of fluoro, —OH, —NH2, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, and —R1b, wherein R1a is selected from the group consisting of hydrogen, provided, however, that hydrogen is not bound to any of C(S), C(O), S(O), or S(O)2 of —C(O)R1a, —C(S)R1a, —NR1aC(O)R1a, —NR1aC(S)R1a, —NR1aS(O)2R1a, —S(O)R1a, or —S(O)2R1a, —R1b, and lower alkyl optionally substituted with one or more, also 1, 2 or 3 groups or substituents selected from the group consisting of fluoro, —OH, —NH2, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, and —R1b, provided, however, that any substitution of the alkyl carbon bound to O, S, or N of —OR1a, —SR1a, —NR1aR1a, —C(O)OR1a, —C(O)NR1aR1a, —C(S)NR1aR1a, —S(O)2NR1aR1a, —NR1aC(O)R1a, —NR1aC(S)R1a, or —NR1aS(O)2R1a, is fluoro or —R1b, and wherein —R1b is selected from the group consisting of cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl and heteroaryl are optionally substituted with one or more, also 1, 2 or 3 groups or substituents selected from the group consisting of halogen, —CN, —OH, —NH2, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, and cycloalkylamino.
  • “Lower alkoxy” denotes the group —ORz, where Rz is lower alkyl. “Substituted lower alkoxy” denotes lower alkoxy in which Rz is lower alkyl substituted with one or more substituents as indicated herein, for example, in the description of compounds of Formula I, Formula II, or Formula III, including descriptions of substituted cycloalkyl, cycloheteroalkyl, aryl and heteroaryl, attached at any available atom to produce a stable compound. Preferably, substitution of lower alkoxy is with 1, 2, 3, 4, or 5 substituents, also 1, 2, or 3 substituents. For example “fluoro substituted lower alkoxy” denotes lower alkoxy in which the lower alkyl is substituted with one or more fluoro atoms, where preferably the lower alkoxy is substituted with 1, 2, 3, 4 or 5 fluoro atoms, also 1, 2, or 3 fluoro atoms. While it is understood that substitutions on alkoxy are attached at any available atom to produce a stable compound, substitution of alkoxy is such that O, S, or N (except where N is a heteroaryl ring atom), are not bound to the alkyl carbon bound to the alkoxy O. Further, where alkoxy is described as a substituent of another moiety, the alkoxy oxygen is not bound to a carbon atom that is bound to an O, S, or N of the other moiety (except where N is a heteroaryl ring atom), or to an alkene or alkyne carbon of the other moiety.
  • “Lower alkylthio” denotes the group —SRaa, where Raa is lower alkyl. “Substituted lower alkylthio” denotes lower alkylthio in which Raa is lower alkyl substituted with one or more substituents as indicated herein, for example, in the description of compounds of Formula I, Formula II, or Formula III, including descriptions of substituted cycloalkyl, cycloheteroalkyl, aryl and heteroaryl, attached at any available atom to produce a stable compound. Preferably, substitution of lower alkylthio is with 1, 2, 3, 4, or 5 substituents, also 1, 2, or 3 substituents. For example “fluoro substituted lower alkylthio” denotes lower alkylthio in which the lower alkyl is substituted with one or more fluoro atoms, where preferably the lower alkylthio is substituted with 1, 2, 3, 4 or 5 fluoro atoms, also 1, 2, or 3 fluoro atoms. While it is understood that substitutions on alkylthio are attached at any available atom to produce a stable compound, substitution of alkylthio is such that O, S, or N (except where N is a heteroaryl ring atom), are not bound to the alkyl carbon bound to the alkylthio S. Further, where alkylthio is described as a substituent of another moiety, the alkylthio sulfur is not bound to a carbon atom that is bound to an O, S, or N of the other moiety (except where N is a heteroaryl ring atom), or to an alkene or alkyne carbon of the other moiety.
  • “Amino” or “amine” denotes the group —NH2. “Mono-alkylamino” denotes the group —NHRbb where Rbb is lower alkyl. “Di-alkylamino” denotes the group —NRbbRee, where Rdd and Rcc are independently lower alkyl. “Cycloalkylamino” denotes the group —NRddRee, where Rdd and Ree combine with the nitrogen to form a 5-7 membered heterocycloalkyl, where the heterocycloalkyl may contain an additional heteroatom within the ring, such as O, N, or S, and may also be further substituted with lower alkyl. Examples of 5-7 membered heterocycloalkyl include, but are not limited to, piperidine, piperazine, 4-methylpiperazine, morpholine, and thiomorpholine. While it is understood that when mono-alkylamino, di-alkylamino, or cycloalkylamino are substituents on other moieties that are attached at any available atom to produce a stable compound, the nitrogen of mono-alkylamino, di-alkylamino, or cycloalkylamino as substituents is not bound to a carbon atom that is bound to an O, S, or N of the other moiety.
  • As used herein, the term “composition” refers to a formulation suitable for administration to an intended animal subject for therapeutic purposes that contains at least one pharmaceutically active compound and at least one pharmaceutically acceptable carrier or excipient.
  • The term “pharmaceutically acceptable” indicates that the indicated material does not have properties that would cause a reasonably prudent medical practitioner to avoid administration of the material to a patient, taking into consideration the disease or conditions to be treated and the respective route of administration. For example, it is commonly required that such a material be essentially sterile, e.g., for injectibles.
  • In the present context, the term “therapeutically effective” or “effective amount” indicates that the materials or amount of material is effective to prevent, alleviate, or ameliorate one or more symptoms of a disease or medical condition, and/or to prolong the survival of the subject being treated.
  • In the present context, the terms “synergistically effective” or “synergistic effect” indicate that two or more compounds that are therapeutically effective, when used in combination, provide improved therapeutic effects greater than the additive effect that would be expected based on the effect of each compound used by itself.
  • As used herein, the terms “ligand” and “modulator” are used equivalently to refer to a compound that changes (i.e., increases or decreases) the activity of a target biomolecule, e.g., an enzyme such as a kinase. Generally a ligand or modulator will be a small molecule, where “small molecule refers to a compound with a molecular weight of 1500 daltons or less, or preferably 1000 daltons or less, 800 daltons or less, or 600 daltons or less. Thus, an “improved ligand” is one that possesses better pharmacological and/or pharmacokinetic properties than a reference compound, where “better” can be defined by one skilled in the relevant art for a particular biological system or therapeutic use.
  • In the context of compounds binding to a target, the terms “greater affinity” and “selective” indicates that the compound binds more tightly than a reference compound, or than the same compound in a reference condition, i.e., with a lower dissociation constant. In some embodiments, the greater affinity is at least 2, 3, 4, 5, 8, 10, 50, 100, 200, 400, 500, 1000, or 10,000-fold greater affinity.
  • As used herein in connection with compounds of the invention, the term “synthesizing” and like terms means chemical synthesis from one or more precursor materials.
  • By “assaying” is meant the creation of experimental conditions and the gathering of data regarding a particular result of the experimental conditions. For example, enzymes can be assayed based on their ability to act upon a detectable substrate. A compound or ligand can be assayed based on its ability to bind to a particular target molecule or molecules.
  • As used herein, the term “modulating” or “modulate” refers to an effect of altering a biological activity, especially a biological activity associated with a particular biomolecule such as a protein kinase. For example, an agonist or antagonist of a particular biomolecule modulates the activity of that biomolecule, e.g., an enzyme, by either increasing (e.g. agonist, activator), or decreasing (e.g. antagonist, inhibitor) the activity of the biomolecule, such as an enzyme. Such activity is typically indicated in terms of an inhibitory concentration (IC50) or excitation concentration (EC50) of the compound for an inhibitor or activator, respectively, with respect to, for example, an enzyme.
  • In the context of the use, testing, or screening of compounds that are or may be modulators, the term “contacting” means that the compound(s) are caused to be in sufficient proximity to a particular molecule, complex, cell, tissue, organism, or other specified material that potential binding interactions and/or chemical reaction between the compound and other specified material can occur.
  • As used herein in connection with amino acid or nucleic acid sequence, the term “isolate” indicates that the sequence is separated from at least a portion of the amino acid and/or nucleic acid sequences with which it would normally be associated.
  • In connection with amino acid or nucleic sequences, the term “purified” indicates that the subject molecule constitutes a significantly greater proportion of the biomolecules in a composition than the proportion observed in a prior composition, e.g., in a cell culture. The greater proportion can be 2-fold, 5-fold, 10-fold, or more than 10-fold, with respect to the proportion found in the prior composition.
  • The present invention concerns compounds of Formula I, and all sub-generic formulae, compounds of Formula II and all sub-generic formulae, and compounds of Formula III and all sub-generic formulae that are modulators of protein kinases, for example without limitation, the compounds are modulators of at least one of the kinases selected from the group consisting of Abl, Akt1, Akt2, Akt3, ALK, Alk5, B-Raf, Brk, Btk, Cdk2, CDK4, CDK5, CDK6, CHK1, c-Raf-1, Csk, EGFR, EphA1, EphA2, EphB2, EphB4, Erk2, Fak, FGFR1, FGFR2, FGFR3, FGFR4, Flt1, Flt3, Flt4, Fms, Frk, Fyn, Gsk3α, Gsk3β, HCK, Her2/Erbb2, Her4/Erbb4, IGF1R, IKK beta, Irak4, Itk, Jak1, Jak2, Jak3, Jnk1, Jnk2, Jnk3, Kdr, Kit, Lck, Lyn, MAP2K1, MAP2K2, MAP4K4, MAPKAPK2, Met, Mnk1, MLK1, p38, PDGFRA, PDGFRB, PDPK1, Pim1, Pim2, Pim3, PKC alpha, PKC beta, PKC theta, Plk1, Pyk2, Ret, ROCK1, ROCK2, Ron, Src, Stk6, Syk, TEC, Tie2, TrkA, TrkB, Yes, and Zap70, and the use of such compounds in the treatment of diseases or conditions.
  • Kinase Targets and Indications of the Invention
  • Protein kinases play key roles in propagating biochemical signals in diverse biological pathways. More than 500 kinases have been described, and specific kinases have been implicated in a wide range of diseases or conditions (i.e., indications), including for example without limitation, cancer, cardiovascular disease, inflammatory disease, neurological disease, and other diseases. As such, kinases represent important control points for small molecule therapeutic intervention. Description of specific target protein kinases contemplated by the present invention may be found, for example, in U.S. patent application Ser. No. 11/473,347 (PCT publication WO2007002433), the disclosure of which is hereby incorporated by reference in its entirety, in addition to the following:
  • Exemplary Diseases Associated with Raf Kinases
  • A-Raf: Target kinase A-Raf (i.e., v-raf murine sarcoma 3611 viral oncogene homolog 1) is a 67.6 kDa serine/threonine kinase encoded by chromosome Xp11.4-p11.2 (symbol: ARAF). The mature protein comprises RBD (i.e., Ras binding domain) and phorbol-ester/DAG-type zinc finger domain and is involved in the transduction of mitogenic signals from the cell membrane to the nucleus. A-Raf inhibitors may be useful in treating neurologic diseases such as multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease (AD), Parkinson's disease; neoplastic diseases including, but not limited to, melanoma, glioma, sarcoma, carcinoma (e.g. colorectal, lung, breast, pancreatic, thyroid, renal, ovarian), lymphoma (e.g. histiocytic lymphoma), neurofibromatosis, myelodysplastic syndrome, leukemia, tumor angiogenesis; pain of neuropathic or inflammatory origin, including acute pain, chronic pain, cancer-related pain and migraine; and diseases associated with muscle regeneration or degeneration, including, but not limited to, vascular restenosis, sarcopenia, muscular dystrophies (including, but not limited to, Duchenne, Becker, Emery-Dreifuss, Limb-Girdle, Facioscapulohumeral, Myotonic, Oculopharyngeal, Distal and Congenital Muscular Dystrophies), motor neuron diseases (including, but not limited to, amyotrophic lateral sclerosis, infantile progressive spinal muscular atrophy, intermediate spinal muscular atrophy, juvenile spinal muscular atrophy, spinal bulbar muscular atrophy, and adult spinal muscular atrophy), inflammatory myopathies (including, but not limited to, dermatomyositis, polymyositis, and inclusion body myositis), diseases of the neuromuscular junction (including, but not limited to, myasthenia gravis, Lambert-Eaton syndrome, and congenital myasthenic syndrome), myopathies due to endocrine abnormalities (including, but not limited to, hyperthyroid myopathy and hypothyroid myopathy) diseases of peripheral nerve (including, but not limited to, Charcot-Marie-Tooth disease, Dejerine-Sottas disease, and Friedreich's ataxia), other myopathies (including, but not limited to, myotonia congenita, paramyotonia congenita, central core disease, nemaline myopathy, myotubular myopathy, and periodic paralysis), and metabolic diseases of muscle (including, but not limited to, phosphorylase deficiency, acid maltase deficiency, phosphofructokinase deficiency, debrancher enzyme deficiency, mitochondrial myopathy, carnitine deficiency, carnitine palmatyl transferase deficiency, phosphoglycerate kinase deficiency, phosphoglycerate mutase deficiency, lactate dehydrogenase deficiency, and myoadenylate deaminase deficiency).
  • B-Raf: Target kinase B-Raf (i.e., v-raf murine sarcoma viral oncogene homolog B1) is a 84.4 kDa serine/threonine kinase encoded by chromosome 7q34 (symbol: BRAF). The mature protein comprises RBD (i.e., Ras binding domain), C1 (i.e., protein kinase C conserved region 1) and STK (i.e., serine/threonine kinase) domains.
  • Target kinase B-Raf is involved in the transduction of mitogenic signals from the cell membrane to the nucleus and may play a role in the postsynaptic responses of hippocampal neurons. As such, genes of the RAF family encode kinases that are regulated by Ras and mediate cellular responses to growth signals. Indeed, B-Raf kinase is a key component of the RAS->Raf->MEK->ERK/MAP kinase signaling pathway, which plays a fundamental role in the regulation of cell growth, division and proliferation, and, when constitutively activated, causes tumorigenesis. Among several isoforms of Raf kinase, the B-type, or B-Raf, is the strongest activator of the downstream MAP kinase signaling.
  • The BRAF gene is frequently mutated in a variety of human tumors, especially in malignant melanoma and colon carcinoma. The most common reported mutation was a missense thymine (T) to adenine (A) transversion at nucleotide 1796 (T1796A; amino acid change in the B-Raf protein is Val<600> to Glu<600>) observed in 80% of malignant melanoma tumors. Functional analysis reveals that this transversion is the only detected mutation that causes constitutive activation of B-Raf kinase activity, independent of RAS activation, by converting B-Raf into a dominant transforming protein. Based on precedents, human tumors develop resistance to kinase inhibitors by mutating a specific amino acid in the catalytic domain as the “gatekeeper”. (Balak, et. al., Clin Cancer Res. 2006, 12:6494-501). Mutation of Thr-529 in BRAF to Ile is thus anticipated as a mechanism of resistance to BRAF inhibitors, and this can be envisioned as a transition in codon 529 from ACC to ATC.
  • Niihori et al., report that in 43 individuals with cardio-facio-cutaneous (CFC) syndrome, they identified two heterozygous KRAS mutations in three individuals and eight BRAF mutations in 16 individuals, suggesting that dysregulation of the RAS-RAF-ERK pathway is a common molecular basis for the three related disorders (Niihori et al., Nat. Genet. 2006, 38(3):294-6).
  • c-Raf-1: Target kinase c-Raf-1 (i.e., v-raf murine sarcoma viral oncogene homolog 1) is a 73.0 kDa STK encoded by chromosome 3p25 (symbol: RAF1). c-Raf-1 can be targeted to to the mitochondria by BCL2 (i.e., oncogene B-cell leukemia 2) which is a regulator of apoptotic cell death. Active c-Raf-1 improves BCL2-mediated resistance to apoptosis, and c-Raf-1 phosphorylates BAD (i.e., BCL2-binding protein). c-Raf-1 is implicated in carcinomas, including colorectal, ovarian, lung and renal cell carcinoma. C-Raf-1 is also implicated as an important mediator of tumor angiogenesis (Hood, J. D. et al., 2002, Science 296, 2404). C-Raf-1 inhibitors may also be useful for the treatment of acute myeloid leukemia and myelodysplastic syndromes (Crump, Curr Pharm Des 2002, 8(25):2243-8). Raf-1 activators may be useful as treatment for neuroendocrine tumors, such as medullary thyroid cancer, carcinoid, small cell lung cancer and pheochromocytoma (Kunnimalaiyaan et al., Anticancer Drugs 2006, 17(2):139-42).
  • B-Raf and/or C-Raf inhibitors may be useful in treating A-Raf-mediated, B-Raf-mediated or c-Raf-1-mediated disease or condition selected from the group consisting of neurologic diseases, including, but not limited to, multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease (AD), Parkinson's disease; neoplastic diseases including, but not limited to, melanoma, glioma, sarcoma, carcinoma (e.g. colorectal, lung, breast, pancreatic, thyroid, renal, ovarian), lymphoma (e.g. histiocytic lymphoma) neurofibromatosis, acute myeloid leukemia, myelodysplastic syndrome, leukemia, tumor angiogenesis, neuroendocrine tumors such as medullary thyroid cancer, carcinoid, small cell lung cancer and pheochromocytoma; pain of neuropathic or inflammatory origin, including, but not limited to, acute pain, chronic pain, cancer-related pain, and migraine; cardiovascular diseases, including, but not limited to, heart failure, ischemic stroke, cardiac hypertrophy, thrombosis (e.g. thrombotic microangiopathy syndromes), atherosclerosis, and reperfusion injury; inflammation including, but not limited to, psoriasis, arthritis and autoimmune diseases and conditions, osteoarthritis, endometriosis, scarring, vascular restenosis, fibrotic disorders, rheumatoid arthritis, inflammatory bowel disease (IBD); immunodeficiency diseases, including, but not limited to, organ transplant rejection, graft versus host disease; renal or prostatic diseases, including, but not limited to, diabetic nephropathy, polycystic kidney disease, nephrosclerosis, glomerulonephritis, prostate hyperplasia; metabolic disorders, including, but not limited to, obesity; infection, including, but not limited to, Helicobacter pylori, Hepatitis and Influenza viruses, fever, and sepsis; pulmonary diseases, including, but not limited to, chronic obstructive pulmonary disease (COPD) and acute respiratory distress syndrome (ARDS); genetic developmental diseases, including, but not limited to, Noonan's syndrome, Costello syndrome, (faciocutaneoskeletal syndrome), LEOPARD syndrome, cardio-faciocutaneous syndrome (CFC), and neural crest syndrome abnormalities causing cardiovascular, skeletal, intestinal, skin, hair and endocrine diseases.
  • Exemplary Diseases Associated with c-Kit.
  • The compounds described herein are useful for treating disorders related to c-kit e.g., diseases related to unregulated kinase signal transduction, including cell proliferative disorders, fibrotic disorders and metabolic disorders, among others. As described in more detail below and in Lipson et al., U.S. 20040002534 (U.S. application Ser. No. 10/600,868, filed Jun. 23, 2003) which is incorporated herein by reference in its entirety, cell proliferative disorders which can be treated by the present invention include cancers, and mast cell proliferative disorders.
  • The presence of c-kit has also been associated with a number of different types of cancers. In addition, the association between abnormalities in c-kit and disease are not restricted to cancer. As such, c-kit has been associated with malignancies, including, but not limited to, mast cell tumors, small cell lung cancer, testicular cancer, gastrointestinal stromal tumors (GISTs), glioblastoma, astrocytoma, neuroblastoma, carcinomas of the female genital tract, sarcomas of neuroectodermal origin, colorectal carcinoma, carcinoma in situ, Schwann cell neoplasia associated with neurofibromatosis, acute myelocytic leukemia, acute lymphocytic leukemia, chronic myelogenous leukemia, mastocytosis, melanoma, and canine mast cell tumors, and inflammatory diseases, including, but not limited to, asthma, rheumatoid arthritis, allergic rhinitis, multiple sclerosis, inflammatory bowel syndrome, transplant rejection, and hypereosinophilia.
  • Exemplary Diseases Associated with c-FMS
  • The presence of c-fins has been associated with a number of different types of diseases. As such, c-fins has been associated with immune disorders, including rheumatoid arthritis, systemic lupus erythematosis (SLE), and transplant rejection; inflammatory diseases including, but not limited to, osteoarthritis, inflammatory bowel syndrome, ulcerative colitis, Crohn's disease, chronic obstructive pulmonary disease (COPD), emphysema, Kawasaki's Disease, hemophagocytic syndrome (macrophage activation syndrome), multicentric reticulohistiocytosis, and atherosclerosis; metabolic disorders, including, but not limited to, Type I diabetes, Type II diabetes, insulin resistance, hyperglycemia, obesity, and lipolysis; disorders of bone structure, mineralization and bone formation and resorption, including, but not limited to, osteoporosis, increased risk of fracture, Paget's disease, hypercalcemia, infection-mediated osteolysis (e.g. osteomyelitis), peri-prosthetic or wear-debris-mediated osteolysis, and metastasis of cancer to bone; kidney and genitourinary diseases, including, but not limited to, endometriosis, nephritis (e.g. glomerulonephritis, interstitial nephritis, Lupus nephritis), tubular necrosis, diabetes-associated renal complications (e.g. diabetic nephropathy), and renal hypertrophy; disorders of the central nervous system, including, but not limited to, multiple sclerosis, stroke, Alzheimer's disease and Parkinson's disease; inflammatory and chronic pain, including, but not limited to, bone pain; and cancers, including, but not limited to, multiple myeloma, acute myeloid leukemia (AML), chronic myeloid leukemia (CML), prostate cancer, breast cancer, ovarian cancer, melanoma, glioblastoma multiforme, metastasis of tumors to other tissues, and other chronic myeloproliferative diseases such as myelofibrosis.
  • Exemplary Diseases Associated with TrkA and TrkB
  • TrkA: Target kinase TrkA (i.e., neurotrophic tyrosine kinase, receptor, type 1) is a 140 kDa tyrosine kinase encoded by chromosome 1q21-q22 (symbol: NTRK1). TrkA inhibitors may be useful in treating pain (e.g. chronic pain, neuropathic pain), cancer (e.g. prostate cancer, lung cancer, myeloid leukemia, pancreatic cancer), allergic disorders (e.g. asthma), arthritis, diabetic retinopathy, macular degeneration and psoriasis.
  • TrkA is a plasma member receptor composed of an extracellular domain (responsible for high affinity binding to nerve growth factor, NGF), a transmembrane segment and an intracellular protein tyrosine kinase domain (responsible to transmit the NGF signal to initiate and coordinate neuronal responses). NGF binding induces TrkA clustering on the membrane and activates the kinase. The kinase initiates a cascade of protein phosphorylation events through multiple pathways including SHC/Ras/MAPK, PI3K and PLCg1. A TrkA kinase inhibitor would not prevent NGF/TrkA binding, but could prevent down-stream signal transduction.
  • Nerve Growth Factor (NGF) is produced by a number of tissues and inflammatory cells during tissue injury and host immune response. It initiates and maintains hypersensitivity to incoming stimulus (hyperalgesia) and the perception of non-noxious stimuli (allodynia). Through its high-affinity receptor TrkA, NGF increases the excitation state of sensory neurons leading to the central nervous system (peripheral sensitization), and increases transmitter release from the dorsal spinal cord (central sensitization). In clinical trials, a single NGF subcutaneous injection generated local hyperalgesia persisting up to 7 weeks. At doses above 0.1 microgram/kg, NGF caused muscle pain that varied from mild to moderate, primarily in the bulbar and truncal musculature. Intravenous NGF produced earlier and more pronounced systemic effects (Petty et al, 1994, Ann Neurol. 36: 244-6). Conversely, TrkA kinase inhibitors could be used to treat diseases of enhanced states of nociception.
  • In Complete Freund's Adjuvant (CFA)-induced hind-paw inflammation, spinal nerve ligation and streptozoticin-induced neuropathic pain models, a single intraperitoneal injection of anti-NGF reversed established tactile allodynia from day 3 to day 7 following treatment. In the mouse CCI model, anti-NGF reversed tactile allodynia when administered 2 weeks after surgery. Repeated administration of this antibody to CCI mice for 3 weeks produced a sustained reversal of tactile allodynia (Wild et al, 2007, J. Pharmacol. Exp. Ther. 322:282-287).
  • Prostate tumors that have metastasized to bone frequently induce bone pain which can be difficult to fully control as it seems to be driven simultaneously by inflammatory, neuropathic, and tumorigenic mechanisms. Anti-NGF produced a significant reduction in both early and late stage bone cancer pain-related behaviors. This therapy did not influence tumor-induced bone remodeling, osteoblast proliferation, osteoclastogenesis, tumor growth, or markers of sensory or sympathetic innervation in the skin or bone. All nerve fibers that innervate the bone express TrkA and p′75, and these are the receptors through which NGF sensitizes and/or activates nociceptors (Halvorson et al, 2005, Cancer Res. 65:9426-35).
  • In patients with mild asthma due to exposure to cat allergen, NGF expression was strongly induced in epithelial cells, fibroblasts, blood vessels, and a few infiltrating cells. TrkA mRNA and protein levels in bronchial biopsies were increased significantly after allergen exposure in infiltrating mast cells before the onset of symptoms (Kassel et al, 2001, Clin Exp Allergy 31:1432-40).
  • The late phase reaction in asthma following allergen provocation is dominated by an influx of activated eosinophils into the bronchial lumen, which correlates with the release of eosinophilic products into the airways to increase disease severity. The viability and activation of eosinophils from patients with mild asthma were significantly enhanced after NGF stimulation. Addition of neutralizing anti-NGF antibodies ex vivo abrogated the effects (Nassentein et al, 2003, J Exp Med 198:455-467). TrkA kinase inhibitors could decrease this paracrine loop between the respiratory tract and infiltrating mast cells as well as endobronchial eosinophils, and thus be useful for the treatment of asthma and other allergic disorders.
  • TrkB: Target kinase TrkB (i.e., neurotrophic tyrosine kinase, receptor, type 2) is a 145 kDa tyrosine kinase encoded by chromosome 9q22.1 (symbol: NTRK2). TrkB inhibitors may be useful in treating various cancers and their metastases (e.g. prostate cancer, lung cancer, Wilms tumors, neuroblastoma, and pancreatic cancer), and various neuropathies (e.g. stroke, multiple sclerosis, transverse myelitis, and encephalitis).
  • In clinical trials with recombinant BDNF, paresthesia was developed at the site of subcutaneous injection (Coulie et al, 2000, Gastroenterology 119:41-50). Intrathecal infusion of BDNF in humans also induced paresthesia and warmth as side effects (Ochs et al, 2000, Amyotroph Lateral Scler Other Motor Neuron Disord. 1:201-6). Chronic paresthesia is often a symptom of an underlying neurological disease or traumatic nerve damage. Paresthesia can be caused by disorders affecting the central nervous system, such as stroke and transient ischemic attacks (mini-strokes), multiple sclerosis, transverse myelitis, and encephalitis. Since BDNF binds to TrkB specifically with high affinity these neuropath effects are mediated through TrkB signaling. Thus Trkb kinase inhibitors could be used to treat certain patients with neuropathy.
  • BDNF is known to act at the synapses between primary sensory and spinal dorsal horn neurons to affect pain transmission during inflammation. The primary afferent is the only source of BDNF in the spinal cord, and it is up-regulated in the dorsal root ganglion (DRG) by peripheral NGF a few days after inflammation, and is transported and released into the superficial dorsal horn in an activity-dependent manner. TrkB expression in the dorsal horn also increases for a few days after inflammation. These findings suggest that BDNF may act during the restricted period in the early phase of inflammation. Through TrkB, BDNF activates two distinct channels: (1) transient receptor potential canonicals (TRPC3), which produces a slow response by opening of a non-selective cation channel; and (2) Na+ channel, which mediates a rapid depolarization in the hippocampus. These channels have been strongly associated with inflammatory pain. Anti-BDNF significantly increased the withdrawal threshold in CFA-treated rats, a model of inflammatory pain. Since the swelling at the site of CFA injection was not affected by antiserum, the residual component might be due to peripheral sensitization (Matayoshi et al, 2005, J. Physiol. 569:685-95).
  • In patients with neuroblastomas, co-expression of TrkB and BDNF, co-expression of TrkB with N-Myc amplification, and expression of truncated TrkB are found to be associated with poorer clinical outcome (Nakagawara et al, 1994, Mol Cell Biol. 14:759-767). Co-expression of TrkB with its ligand BDNF could generate a positive feedback loop through autocrine and paracrine loops. Also TrkB truncations found in these tumors generate activated forms of the intracellular protein tyrosine kinase. The constitutively active TrkB signals through multiple pathways to promote cancer initiation, progression and metastasis. These truncated TrkB kinases were also found in hepatocellular carcinoma (Yang et al, 2005, Cancer. Res 65:219-225). Thus TrkB inhibitors could be used to treat a sub-population of cancer patients with an activated TrkB pathway.
  • In patients with pancreatic cancer, TrkB expression is correlated with perineural invasion, positive retroperitoneal margin, and shorter latency to development of liver metastasis (Sclabas et al, 2005, Clin. Cancer. Res V11:440-449). Mechanistically, TrkB activates the PI3K pathway to suppress anoikis (apoptosis resulting from loss of cell-matrix interactions) which is one of the physiological barriers to metastasis. TrkB kinase inhibition could break down resistance to anoikis of metastasizing tumors (Douma et al, 2004, Nature 430:1034-9). Therefore, TrkB inhibitors could have utility in a broad range of tumor types.
  • Exemplary Diseases Associated with MAPK4K
  • MAP4K4: Target kinase MAP4K4 (i.e., Mitogen-activated protein kinase kinase 4, aka Hematopoietic progenitor kinase/Germinal center kinase-like Kinase) is a 130 kDa serine/threonine kinase encoded by chromosome 2q11.2-q12 (symbol: MAP4K4) and is also known as HGK. It is a member of the human STE20/mitogen-activated protein kinase kinase kinase kinase (MAP4K) family of serine/threonine kinases and is the human ortholog of mouse NIK (Nck-interacting kinase). The N-terminus of the mature HGK protein has a catalytic kinase domain that shares 47% and 48% amino acid sequence identity to the catalytic domain of Hematopoietic progenitor kinase 1 (HPK1) and Germinal center kinase (GCK), respectively. Yao et al. (J. Biol. Chem. 274: 2118-2125, 1999) identified 2 HGK isoforms, one of which has no proline-rich domains, and another, longer variant that contains such domains and appears to be expressed in brain only. Northern blot analysis revealed expression of 3 HGK transcripts of approximately 4.6, 6.5, and 8.5 kb in heart, brain, skeletal muscle, pancreas, placenta, liver, lung, and kidney. By Western blot analysis with a polyclonal antibody, Yao et al. (J. Biol. Chem. 274: 2118-2125, 1999) found that the 130-kD protein is expressed in multiple cell lines.
  • Expression of HGK in transfected cell lines resulted in strong JNK activation and, in turn, c-jun transcriptional activity (Yao et al. J. Biol. Chem. 274: 2118-2125, 1999). HGK-induced JNK activation was inhibited by dominant-negative MAP2K4, MAP2K7, and TAK1 mutants. TNF-alpha also stimulated HGK kinase activity. HGK was identified as a putative effect of Rap2 to activate JNK (Machida et al. J. Biol. Chem. 279: 15711-15714, 2004). This link establishes HGK as a potential target for a range of metabolic indications, since the JNK pathway clearly antagonizes insulin signaling. An HGK inhibitor could re-sensitize fat and muscle cells to insulin.
  • HGK is found to be broadly expressed in human tumor cells and can modulate cellular transformation, invasion, and adhesion (Wright et al. Mol. Cell. Biol. 23: 2068-2082, 2003). Wright et al showed HGK to be highly expressed in most tumor cell lines relative to normal tissue. An active role for this kinase in transformation was suggested by an inhibition of H-Ras(V12)-induced focus formation by expression of inactive, dominant-negative mutants of HGK in both fibroblast and epithelial cell lines. Expression of an inactive mutant of HGK also inhibited the anchorage-independent growth of cells yet had no effect on proliferation in monolayer culture. Expression of HGK mutants modulated integrin receptor expression and had a striking effect on hepatocyte growth factor-stimulated epithelial cell invasion. Together, these results suggest an important role for HGK in cell transformation and invasiveness. More recently, a small interfering RNA screen for modulators of tumor cell motility identifies MAP4K4 as a promigratory kinase (Collins et al. Proc. Natl. Acad. Sci. USA, 103: 3775-3780, 2006). Collins et al. showed that the knockdown of the HGK transcript inhibited the migration of multiple carcinoma cell lines, indicating a broad role in cell motility, and potently suppressed the invasion of SKOV-3 cells in vitro. The effect of HGK on cellular migration was found to be mediated through JNK kinase, independent of AP1 activation and downstream transcription. Accordingly, small molecule inhibition of c-Jun N-terminal kinase suppressed SKOV-3 cell migration, underscoring the potential therapeutic utility of mitogen-activated protein kinase pathway inhibition in cancer progression (Collins et al. Proc. Natl. Acad. Sci. USA, 103: 3775-3780, 2006). These studies strongly support HGK as a target in a broad range of oncology indications. In particular, an HGK inhibitor could have utility in blocking the migration, invasion and metastasis in many different tumor types.
  • Activation of T-cells by antigens initiates a complex series of signal-transduction events that are critical for immune responses. Mack et al. (Immunol Lett. 96, 129-145, 2005) developed a genetic screen to survey the functional roles of kinases in antigen mediated T-cell activation and identified 19 protein kinases that were previously implicated in T-cell signaling processes and 12 kinases that were not previously linked to T-cell activation, including HGK. siRNA studies showed a role for HGK in antigen mediated T-cell responses in Jurkat and primary T-cells. In addition, by analyzing multiple promoter elements using reporter assays, Mack et al. have shown that MAP4K4 is implicated in the activation of the TNF-alpha promoter. Therefore, inhibition of HGK could have broad therapeutic utility for T-cell-mediated autoimmune diseases.
  • Insulin-regulated glucose transporter GLUT4 is a key modulator of whole body glucose homeostasis, and its selective loss in adipose tissue or skeletal muscle causes insulin resistance and diabetes. Using an RNA interference-based screen, Tang et al. (Proc Natl Acad Sci USA. 103:2087-2092, 2006) found 4 negative regulators of insulin-responsive glucose transport in mouse adipocytes: Pctk1, Pftk1, Ikbka (CHUK), and HGK. HGK suppressed expression of adipogenic transcription factors, C/EBPA, C/EBPB, and PPARG, and it suppressed surface expression of GLUT4 (SLC2A4), resulting in attenuated membrane hexose transport activity. RNA interference-mediated depletion of HGK early in differentiation enhanced adipogenesis and triglyceride deposition; in fully differentiated adipocytes, loss of HGK upregulated GLUT4 expression. Conversely, conditions that inhibited adipogenesis, such as TNF-alpha treatment or PPARG depletion, markedly upregulated HGK. Tang et al. (Proc Natl Acad Sci USA. 103:2087-2092, 2006) concluded that MAP4K4-dependent signaling inhibited PPARG-responsive gene expression, adipogenesis, and insulin-stimulated glucose transport. Furthermore, TNF-alpha signaling to down-regulate GLUT4 is impaired in the absence of HGK, indicating that HGK expression is required for optimal TNF-alpha action. This study further supports HGK as a target in metabolic disease, and suggests a role for HGK inhibition in ameliorating the pathology in adipocytes.
  • In a separate study (Bouzakri and Zierath J. Biol. Chem. 282:7783-7789, 2007), using small interfering RNA (siRNA) to suppress the expression of HGK protein 85% in primary human skeletal muscle cells, TNF-alpha-induced insulin resistance on glucose uptake was completely prevented. HGK silencing inhibited TNF-alpha-induced negative signaling inputs by preventing excessive JNK and ERK-1/2 phosphorylation, as well as IRS-1 serine phosphorylation. These results highlight the HGK/JNK/ERK/IRS module in the negative regulation of insulin signaling to glucose transport in response to TNF-alpha. Depletion of HGK also prevented TNF-alpha-induced insulin resistance on AKT and the AKT substrate 160 (AS160), providing evidence that appropriate insulin signaling inputs for glucose metabolism were rescued. The authors suggested that strategies to inhibit HGK may be efficacious in the prevention of TNF-alpha-induced inhibitory signals that cause skeletal muscle insulin resistance on glucose metabolism in humans. Moreover, in myotubes from insulin-resistant type II diabetic patients, siRNA against HGK restored insulin action on glucose uptake to levels observed in healthy subjects. This study further supports HGK as a target in metabolic diseases such as type II diabetes, and suggests a role for HGK inhibition in ameliorating the pathology in muscle cells.
  • HGK inhibitors may be useful in treating metabolic indications, including re-sensitizing fat and muscle cells to insulin, ameliorating the pathology in adipocytes, ameliorating the pathology in muscle cells, metabolic syndrome and type II diabetes; a broad range of oncology indications, including blocking the migration, invasion and metastasis in many different tumor types; and T-cell mediated autoimmune diseases.
  • Kinase Activity Assays
  • A number of different assays for kinase activity can be utilized for assaying for active modulators and/or determining specificity of a modulator for a particular kinase or group or kinases. In addition to the assay mentioned in the Examples below, one of ordinary skill in the art will know of other assays that can be utilized and can modify an assay for a particular application. For example, numerous papers concerning kinases describe assays that can be used.
  • Additional alternative assays can employ binding determinations. For example, this sort of assay can be formatted either in a fluorescence resonance energy transfer (FRET) format, or using an AlphaScreen (amplified luminescent proximity homogeneous assay) format by varying the donor and acceptor reagents that are attached to streptavidin or the phosphor-specific antibody.
  • Organic Synthetic Techniques
  • A wide array of organic synthetic techniques exist in the art to facilitate the construction of potential modulators. Many of these organic synthetic methods are described in detail in standard reference sources utilized by those skilled in the art. One example of such a reference is March, 1994, Advanced Organic Chemistry; Reactions, Mechanisms and Structure, New York, McGraw Hill. Thus, the techniques useful to synthesize a potential modulator of kinase function are readily available to those skilled in the art of organic chemical synthesis.
  • Regarding the synthetic examples described herein, solvents include polar and non-polar solvents known to those of skill in the art, including polar aprotic and polar protic solvents. Polar solvents include, without limitation, protic solvents such as methanol, ethanol, isopropyl alcohol, t-butanol, n-butanol, acetic acid, formic acid or water, or aprotic solvents such as tetrahydrofuran (THF), acetonitrile, dioxane, methylene chloride, dimethylsulfoxide (DMSO), acetone, N,N-dimethylformamide (DMF), N,N-dimethylacetamide (DMA), ethyl acetate, 1,2-dimethoxyethane, 1,2-dichloroethane, chloroform, 1,2-dichloroethane, or pyridine. Polar solvents include a mixture of water with any of the above, or a mixture of any two or more of the above. Apolar solvents include, without limitation, toluene, benzene, chlorobenzene, xylenes and hexanes.
  • Regarding the synthetic examples described herein, reducing agent includes, without limitation, a reducing agent such as catalytic reducing agents using hydrogen and transition metal catalysts such as palladium, platinum, rhodium, etc. (e.g. Pt/acetic acid/H2); a mixture of trifluoroacetic acid and triethylsilane, borane tetrahydrofuran complex, diborane, borane dimethylsulfide complex, and a combination of sodium borohydride and boron trifluoride; metals such as reduced iron, zinc powder, magnesium etc.; metal hydrogen complex compounds such as alkali metal borohydrides (for example, potassium borohydride, sodium borohydride, lithium borohydride, zinc borohydride, sodium triacetoxyborohydride, etc.), aluminum lithium hydride, etc.; metal hydrides such as sodium hydride, etc.; organic tin compounds (triphenyltin hydride, etc.); and metal salts such as nickel compounds, zinc compounds, tin compounds (for example tin(II) chloride), and samarium iodide/pivalic acid/hexamethylphorphoric triamide.
  • Regarding the synthetic examples described herein, oxidizing agent includes, without limitation, an oxidizing agent such as Dess-Martin reagent, TEMPO (2,2,6,6-tetramethylpiperidine-N-oxide), DDQ (2,3-Dichloro-5,6-dicyano-1,4-benzoquinone), PDC (pyridinium dichromate), PCC (pyridinium chlorochromate), Pyridine.SO3, Chromium trioxide, p-nitroperbenzoic acid, magnesium monoperoxyphthalate, sodium periodate, potassium periodate, hydrogen peroxide, urea peroxide, alkali metal bromates, cumene hydroperoxide, tert-butyl peroxide, peracids such as performic acid, peracetic acid, pertrifluoroacetic acid, perbenzoic acid, m-chloroperbenzoic acid, o-carboxyperbenzoic acid and the like; sodium metaperiodate, bichromic acid; bichromates such as sodium bichromate, potassium bichromate; permanganic acid; permanganates such as potassium permanganate, sodium permanganate; and lead salts such as lead tetraacetate.
  • Regarding the synthetic examples described herein, a nitrogen protecting group is a chemical group covalently bound to a nitrogen atom of a compound that is used to protect the nitrogen from reaction during a synthetic step. The nitrogen protecting group may be added to a compound and removed in a subsequent step by methods known to those of skill in the art. Nitrogen protecting groups include, without limitation, carbamates, amides, N-sulfonyl derivatives, groups of formula —C(O)OR, wherein R is, for example, methyl, ethyl, t-butyl, benzyl, phenylethyl, CH2═CHCH2—, and the like, groups of the formula —C(O)R′, wherein R′ is, for example, methyl, phenyl, trifluoromethyl, and the like, groups of the formula —SO2R″, wherein R″ is, for example, tolyl, phenyl, trifluoromethyl, 2,2,5,7,8-pentamethylchroman-6-yl, 2,3,6-trimethyl-4-methoxyphenyl, and the like, and silanyl containing groups, such as 2-trimethylsilylethoxymethyl, t-butyldimethylsilyl, triisopropylsilyl, and the like. Other suitable nitrogen protecting groups may be found in texts such as T. W. Greene & P. G. M. Wuts, Protective Groups in Organic Synthesis, John Wiley & Sons, 1991.
  • Alternative Compound Forms or Derivatives
  • Compounds contemplated herein are described with reference to both generic formulae and specific compounds. In addition, invention compounds may exist in a number of different forms or derivatives, all within the scope of the present invention. Alternative forms or derivatives, such as (a) Isomers, Prodrugs, and Active Metabolites (b) Tautomers, Stereoisomers, Regioisomers, and Solvated Forms (c) Prodrugs and Metabolites (d) Pharmaceutically acceptable salts and (e) Polymorphic forms, are described, for example, in U.S. patent application Ser. No. 11/473,347 (see also, PCT publication WO2007002433), the disclosure of which is hereby incorporated by reference in its entirety.
  • Administration
  • The methods and compounds will typically be used in therapy for human subjects. However, they may also be used to treat similar or identical indications in other animal subjects. In this context, the terms “subject,” “animal subject,” and the like refer to human and non-human vertebrates, e.g. mammals, such as non-human primates, sports and commercial animals, e.g., equines, bovines, porcines, ovines, rodents, and pets, e.g., canines and felines. A description of possible methods and routes of administration may be found, for example, in U.S. patent application Ser. No. 11/473,347 (see also, PCT publication WO2007002433), the disclosure of which is hereby incorporated by reference in its entirety.
  • EXAMPLES
  • Examples related to the present invention are described below. In most cases, alternative techniques can be used. The examples are intended to be illustrative and are not limiting or restrictive to the scope of the invention. In some examples, the mass spectrometry result indicated for a compound may have more than one value due to the isotope distribution of an atom in the molecule, such as a compound having a bromo or chloro substituent.
  • Unless specifically indicated otherwise, the Formula enumeration and R group enumeration used in the following examples is not related to such enumeration in other sections of this application. The reagents and solvents used in these examples can be readily substituted with appropriate alternatives as are known in the art and isolation of products is readily achieved by methods known in the art, including, but not limited to, extraction, crystallization, and chromatographic methods. In addition to the following Examples, exemplary methods for synthesis of compounds of the present invention may be found in U.S. patent application Ser. No. 11/473,347 (see also, PCT publication WO2007002433), the disclosure of which is hereby incorporated by reference in its entirety. The 1H-pyrrolo[2,3-b]pyridine core of compounds described in the examples may also be referred to as 7-azaindole in the examples.
  • Example 1 Synthesis of Compounds of Formula X
  • Figure US20110092538A1-20110421-C00015
  • Step 1—Preparation of Compounds of Formula Xc and Xd
  • To a compound of Formula Xa (R1 is as defined in paragraph [0009]) and a compound of Formula Xb (Y is consistent with compounds of Formula I, Formula II, or Formula III, e.g. Y is:
  • Figure US20110092538A1-20110421-C00016
  • where R2 and R3 are as defined in paragraph [0009], and R6 is as defined in paragraph [0023], where
    Figure US20110092538A1-20110421-P00001
    indicates the attachment point to the carbonyl carbon) is added an appropriate solvent (e.g. methanol) followed by an appropriate base (e.g. potassium hydroxide, sodium methoxide). The reaction is typically allowed to stir at room temperature overnight. Isolation by conventional means (e.g. extraction, washing and filtering) affords a mixture containing compound of Formula Xc, which may be isolated by silica gel chromatography if desired.
  • Step 2—Preparation of Compounds of Formula X
  • To a compound of Formula Xc in an appropriate solvent (e.g. tetrahydrofuran) is added an oxidizing agent (e.g. Dess-Martin periodane, TEMPO, DDQ). Typically, the reaction is allowed to stir at room temperature for 20 minutes. Isolation by conventional means (e.g. extraction and silica gel column chromatography) affords compounds of Formula X.
  • Example 2 Synthesis of Compounds of Formula X
  • Figure US20110092538A1-20110421-C00017
  • Step-1—Synthesis of Compound of Formula X
  • Compound of Formula X is synthesized by reacting a compound of Formula Xa (see Example 1) with a compound of Formula Xd (Y is as defined in Example 1), e.g. benzoyl chloride, in the presence of a Lewis acid (e.g. aluminum trichloride) in an inert solvent (e.g. dichloromethane) under an inert atmosphere (e.g. argon) at room temperature or with heating up to reflux for 1-18 hours. The desired compound X is isolated by extraction and silica gel column chromatography.
  • Example 3 Synthesis of Compounds of Formula X
  • Figure US20110092538A1-20110421-C00018
  • Step-1—Synthesis of Compound Xe
  • Compound of Formula Xe can be synthesized by reacting a compound of Formula Xa (see Example 1) with hexamethyltetramine and acetic acid in water with heating to reflux for two hours. After cooling, the desired compound precipitates and may be collected by filtration.
  • Step-2—Synthesis of Compound of Formula Xf
  • Compound of Formula Xf, where P is a nitrogen protecting group, is synthesized by reacting a compound Xe with an appropriate reagent to introduce a protecting group (e.g. triisopropylsilylchloride) and a base (e.g. sodium hydride) in a solvent (e.g. tetrahydrofuran) typically at room temperature for 8-12 hours. The desired compound is isolated by conventional means (e.g. extraction).
  • Step-3—Synthesis of Compound of Formula Xh
  • Compound of Formula Xh is synthesized by reacting a compound of Formula Xf in a solvent (e.g. tetrahydrofuran) with an organolithium reagent (e.g. phenyl lithium) in a solvent (e.g. tetrahydrofuran) under an inert atmosphere, cooled to −78° C. An appropriate organolithium reagent can also be prepared by reacting compounds of Formula Xg, where Y1 is
  • Figure US20110092538A1-20110421-C00019
  • or —R6 (
    Figure US20110092538A1-20110421-P00001
    indicates attachment point to the sulfone), where R2, R3 and R6 are as defined in Example 1, with an organolithium reagent (e.g. butyllithium) in a solvent (e.g. tetrahydrofuran) under an inert atmosphere, cooled to −78° C. The reaction is typically allowed to warm to room temperature and stirred for 30 minutes. The desired compound is isolated by conventional means (e.g. extraction).
  • Step-4—Synthesis of an Intermediate of Compound of Formula X
  • An intermediate of compound of Formula X is synthesized by reacting a compound of Formula Xh with an appropriate reagent to remove the protecting group, P, (e.g. tetra-n-butyl ammonium fluoride) in an appropriate solvent (e.g. tetrahydrofuran). The final product is isolated by standard procedures (e.g. extraction).
  • Step-5—Synthesis of Compound of Formula X
  • Compound of Formula X is synthesized by reacting the intermediate from Step 4 with an oxidizing agent (e.g. Dess-Martin periodane, TEMPO) in an aprotic solvent (e.g. tetrahydrofuran) typically at room temperature for 20 minutes. The desired compound is isolated by conventional means (e.g. extraction and silica gel chromatography).
  • Example 4 Synthesis of phenyl-(-1H-pyrrolo[2,3-b]pyridine-5-yl)-amine 4
  • Phenyl-(-1H-pyrrolo[2,3-b]pyridine-5-yl)-amine 1 was synthesized in three steps from 5-bromo-7-azaindole 1 as shown in Scheme 1.
  • Figure US20110092538A1-20110421-C00020
  • Step 1—Preparation of 5-bromo-1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridine (2)
  • To 5-bromo-7-azaindole (1, 1.5 g, 7.6 mmol) in N,N-dimethylformamide (20 mL) were added sodium hydride (60% in mineral oil, 0.27 g, 11.0 mmol) and trriisopropylsilyl chloride (2.6 mL, 12.0 mmol), under an atmosphere of nitrogen. The reaction was stirred for 2 hours at room temperature. The reaction was poured into water and extracted with ethyl acetate. The organic layer was washed with brine, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated and purified by silica gel column chromatography eluting with 10% ethyl acetate in hexane to give the compound (2, 1.6 g, 59%). MS (ESI)[M+H+]+=352.3.
  • Step 2—Preparation of 5-phenyl-(1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridine-5-yl)-amine (3)
  • To 5-bromo-1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridine (2, 0.10 g, 0.3 mmol) in toluene (5 mL) were added aniline (0.04 mL, 0.42 mmol), sodium tert-butoxide (0.15 g, 1.56 mmol), tris(dibenzyllideneacetone)dipalladium(0) (9.2 mg, 0.01 mmol) and (S)-(−)-2,2′-bis(diphenylphosphino)-1,1′-binaphthyl (6.3 mg, 0.01 mmol). The reaction was heated to 160° C. for 10 minutes in a CEM Discover microwave instrument. The reaction was concentrated and purified by silica gel column chromatography eluting with 3% ethyl acetate in hexane to give the compound (3, 40 mg, 40%). MS (ESI)[M+H+]+=366.6.
  • Step 3—Preparation of phenyl-(-1H-pyrrolo[2,3-b]pyridine-5-yl)-amine (4)
  • To 5-phenyl-(1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridine-5-yl)-amine (3, 0.14 g, mmol) in tetrahydrofuran (3.0 mL) was added tetra-n-butylammonium fluoride (0.197 g, 0.76 mmol). The reaction was stirred for 1 hour at room temperature. The reaction was concentrated and purified by silica gel column chromatography eluting with 3% ethyl acetate in hexane to give the compound (4, 60 mg, 76%). MS (ESI)[M+H+]+=210.3.
  • Example 5 Synthesis of 5-chloro-1H-pyrrolo[2,3-b]pyridine 6
  • 5-Chloro-1H-pyrrolo[2,3-b]pyridine 6 was synthesized in two steps from 5-bromo-1-triisopropylsilyl-7-azaindole 2 as shown in Scheme 2.
  • Figure US20110092538A1-20110421-C00021
  • Step 1—Preparation 5-chloro-1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridine (5)
  • To 5-bromo-1-triisopropylsilyl-7-azaindole (2, 1.60 g, 4.53 mmol, prepared as described in Example 4) in tetrahydrofuran (50.0 mL), under an atmosphere of nitrogen at −78° C., was added tert-butyllithium (1.70 M in hexane, 6.12 mL). The reaction was stirred for 1 hour, followed by addition of hexachloroethane (1.29 g, 5.43 mmol). The reaction was stirred for 3 hours, poured into water, and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated to give the crude compound (5, 1.60 g). MS (ESI)[M+H+]+=309.3.
  • Step 2—Preparation 5-chloro-1H-pyrrolo[2,3-b]pyridine (6)
  • To 5-chloro-1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridine (5, 1.40 g, 4.53 mmol) in tetrahydrofuran (15 mL) was added tetra-n-butylammonium fluoride (1.42 g, 5.43 mmol). The reaction mixture was stirred at room temperature for 10 minutes. The reaction mixture was concentrated and isolated by silica gel column chromatography eluting with 30% ethyl acetate in hexane to give the compound (6, 0.40 g. 58% over 2 steps). MS (ESI)[M−H+]=153.1.
  • 5-Fluoro-1H-pyrrolo[2,3-b]pyridine 7
  • Figure US20110092538A1-20110421-C00022
  • was prepared using the protocol of Scheme 2, substituting hexachloroethane with N-fluoro-N-(phenylsulfonyl)-benzenesulfonamide in Step 1. MS (ESI) [M+H+]+=137.1.
  • Example 6 Synthesis of 1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridine-3-carbaldehyde 10
  • 1-Triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridine-3-carbaldehyde 10 was synthesized in two steps from 7-azaindole 8 as described in Scheme 3.
  • Figure US20110092538A1-20110421-C00023
  • Step 1—Preparation of 1H-pyrrolo[2,3-b]pyridine-3-carbaldehyde (9)
  • To 1H-Pyrrolo[2,3-b]pyridine (8, 16.0 g, 135 mmol) in water (110 mL), were added hexamethylenetetramine (26.0 g, 185 mmol), and acetic acid (55.0 mL, 967 mmol). The reaction was refluxed for 12 hours. Water (329 mL) was added and the reaction was cooled to room temperature. The reaction was filtrated and washed with water to give the compound (9, 15.0 g, 76%). MS (ESI)[M+H+]+=147.
  • Step 2—Preparation of 1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridine-3-carbaldehyde (10)
  • To 1H-Pyrrolo[2,3-b]pyridine-3-carbaldehyde (9, 4.05 g, 27.71 mmol) in tetrahydrofuran (30.0 mL) were added sodium hydride (60% in mineral oil, 1.5 g, 38 mmol) and trriisopropylsilyl chloride (8.0 mL, 38 mmol) under an atmosphere of nitrogen. The reaction was stirred for 2 hours at room temperature. The reaction was poured into water and extracted with ethyl acetate. The organic layer was washed with brine, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated and purified by silica gel column chromatography eluting with 10% ethyl acetate in hexane to give the compound (10, 3.0 g, 36%). MS (ESI)[M+H+]+=303.
  • Example 7 Synthesis of 5-isopropyl-1H-pyrrolo[2,3-b]pyridine 13
  • 5-Isopropyl-1H-pyrrolo[2,3-b]pyridine 13 was synthesized in three steps from 5-bromo-1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridine 2 as described in Scheme 4.
  • Figure US20110092538A1-20110421-C00024
  • Step 1—Preparation of 2-(1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridin-5-yl)-propan-2-ol (11)
  • To 5-bromo-1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridine (2, 2.0 g, 5.66 mmol, prepared as described in Example 1) in tetrahydrofuran (20.0 mL), cooled in a −78° C. acetone/dry ice bath, under an atmosphere of nitrogen, was added tert-butyllithium (1.7 M in tetrahydrofuran, 7.3 mL, 12 mmol) dropwise. After 20 minutes, acetone (0.830 mL, 11 mmol) was added dropwise to the reaction. The reaction was stirred for 30 minutes at −78° C. and then allowed to reach room temperature. The reaction was poured into water and extracted with ethyl acetate. The organic layer was washed with brine, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated and purified by silica gel column chromatography eluting with 10% ethyl acetate in hexane to give the compound (11, 1.30 g, 69%). MS (ESI)[M+H+]+=333.
  • Step 2—Preparation of 5-isopropenyl-1H-pyrrolo[2,3-b]pyridine (12)
  • To 2-(1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridin-5-yl)-propan-2-ol (11, 0.500 g, 1.5 mmol) in acetonitrile (10.0 mL) were added triethylsilane (1.00 mL, 6.3 mmol) and trifluoroacetic acid (0.50 mL, 6.5 mmol) under an atmosphere of nitrogen. The reaction was refluxed for 3 hours, then cooled down to room temperature. The reaction was poured into water and extracted with ethyl acetate. The organic layer was washed with brine, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated and purified by silica gel column chromatography eluting with 50% ethyl acetate in hexane to give the compound (12, 0.200 g, 84%). MS (ESI)[M+H+]+=159.
  • Step 3—Preparation of 5-isopropyl-1H-pyrrolo[2,3-b]pyridine (13)
  • To 5-isopropenyl-1H-pyrrolo[2,3-b]pyridine (12, 0.080 g, 0.501 mmol) in tetrahydrofuran (5.0 mL) was added 20% palladium hydroxide on carbon (5.0 mg). The reaction was stirred under hydrogen at 40 psi for 30 minutes. The reaction mixture was filtered and concentrated to give the compound (13, 0.078 g, 96%). MS (ESI)[M+H+]+=161.
  • Example 8 Synthesis of 5-methyl-1H-pyrrolo[2,3-b]pyridine 15
  • 5-Methyl-1H-pyrrolo[2,3-b]pyridine 15 was synthesized in two steps from 5-bromo-1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridine 2 as described in Scheme 5.
  • Figure US20110092538A1-20110421-C00025
  • Step 1—Preparation of 5-methyl-1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridine (14)
  • To PdCl2(dppf) (0.04 g, 0.05 mmol) in toluene (10.0 mL) under an atmosphere of nitrogen were added 5-bromo-1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridine (2, 0.3 g, 0.8 mmol, prepared as described in Example 1, 1.0 mL in toluene) and methylmagnesium bromide (1.0 M in tetrahydrofuran, 3.0 mL, 3.0 mmol). The reaction was stirred 90° C. for 2 hours and then allowed to reach to room temperature. The reaction was poured into citric acid (0.1 M in water) and extracted with ethyl acetate. The organic layer was washed with brine, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated and purified by silica gel column chromatography eluting with 50% ethyl acetate in hexane to give the compound (14, 0.16 g, 60.0%). MS (ESI)[M+H+]+=289.4.
  • Step 2—Preparation of 5-methyl-1H-pyrrolo[2,3-b]pyridine (15)
  • To 5-Methyl-1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridine (14, 0.160 g, 0.55 mmol) in tetrahydrofuran (3.0 mL) was added tetra-n-butylammonium fluoride (0.145 g, 0.55 mmol). The reaction was stirred for 1 hour at room temperature. The reaction was poured into water and extracted with ethyl acetate. The organic layer was washed with brine, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated and purified by silica gel column chromatography eluting with 3% methanol in dichloromethane to provide light yellow solid (15, 0.07 g, 95%). MS (ESI)[M+H+]+=133.2.
  • 5-Methyl-1H-pyrrolo[2,3-b]pyridine 16
  • Figure US20110092538A1-20110421-C00026
  • was prepared following the protocol of Scheme 5, substituting methylmagnesium bromide with ethylmagnesium bromide in Step 1.
  • Example 9 Synthesis of 5-methoxy-1H-pyrrolo[2,3-b]pyridine 17 and Related Compounds
  • 5-Methoxy-1H-pyrrolo[2,3-b]pyridine 17 was synthesized in one step from 5-bromo-1H-pyrrolo[2,3-b]pyridine 1 as described in Scheme 6.
  • Figure US20110092538A1-20110421-C00027
  • Step 1—Preparation of 5-methoxy-1H-pyrrolo[2,3-b]pyridine (17)
  • To 5-bromo-7-azaindole (1, 500.0 mg, 2.53 mmol) in N,N-dimethylformamide (8 mL) were added copper(I) iodide (966 mg, 5.08 mmol) and sodium methoxide in methanol (3 M, 5 mL). The reaction was stirred overnight at 120° C. under an atmosphere of Argon. The reaction was poured into water, and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate, filtered. The filtrate was concentrated and purified with silica gel column chromatograph eluting with 20% ethyl acetate in hexane to give white solid (17, 140 mg, 28%). MS (ESI)[M+H+]+=149.1. In an alternative method, 2.3 g (11.7 mmol) 5-bromo-7-azaindole (1, 2.3 g, 11.7 mmol) was dissolved in 75 mL N,N-dimethylformamide and 50 mL methanol (50 mL), adding sodium methoxide (32 g, 0.6 mol) and copper-(I) bromide (3.2 g, 22.4 mmol) at room temperature. The reaction was stirred for three hours at 100° C. under an atmosphere of argon. The mixture was diluted with ethyl acetate and poured into a solution of ammonium chloride:ammonium hydroxide (4:1). The organic layer was extracted with ammonium chloride:ammonium hydroxide (4:1), washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated. The desired compound was isolated by silica gel column chromatography eluting with 30% to 70% ethyl acetate in hexanes to give a yellow solid (17, 0.27 g, 15.6%). MS (ESI) [M+H+]+=149.2.
  • 5-Ethoxy-1H-pyrrolo[2,3-b]pyridine 18
  • Figure US20110092538A1-20110421-C00028
  • was prepared using the protocol of Scheme 6, substituting methanol with ethanol and sodium methoxide with sodium ethoxide.
  • 5-(2-Methoxy-ethoxy)-1H-pyrrolo[2,3-b]pyridine 19
  • Figure US20110092538A1-20110421-C00029
  • was prepared using the protocol of Scheme 6, substituting methanol with 2-methoxy-ethanol and sodium methoxide with sodium 2-methoxy-ethoxide (prepared from 2-methoxy-ethanol and sodium hydride). MS (ESI) [M+H+]+=193.3.
  • Diethyl-[2-(1H-pyrrolo[2,3-b]pyridin-5-yloxy)-ethyl]-amine 20
  • Figure US20110092538A1-20110421-C00030
  • was prepared using the protocol of Scheme 6, substituting methanol with 2-diethylamino-ethanol and sodium methoxide with sodium 2-diethylamino-ethoxide (prepared from 2 2-diethylamino-ethanol and sodium hydride). MS (ESI) [M+H+]+=234.5.
  • Example 10 Synthesis of 5-pyridin-3-yl-1H-pyrrolo[2,3-b]pyridine 22
  • 5-Pyridin-3-yl-1H-pyrrolo[2,3-b]pyridine 22 was synthesized in one step from 5-bromo-1H-pyrrolo[2,3-b]pyridine 1 as described in Scheme 7.
  • Figure US20110092538A1-20110421-C00031
  • Step 1—Preparation of 5-pyridin-3-yl-1H-pyrrolo[2,3-b]pyridine (22)
  • To 5-bromo-7-azaindole (1, 1.00 g, 5.08 mmol) in water (13.0 mL) and acetonitrile (36 mL) were added pyridine-3-boronic acid (21, 1.0 g, 8.1 mmol), potassium carbonate (1.79 g, 0.0130 mol) and Tetrakis(triphenylphosphine)palladium(0) (50.0 mg, 0.043 mmol) under an atmosphere of nitrogen. The reaction mixture was heated to 170° C. overnight. The reaction mixture was poured into water and extracted with ethyl acetate. The organic layer was washed with brine, dried over sodium sulfate, and concentrated. The residue was purified with silica gel column chromatography eluting with 25% ethyl acetate in hexane to provide a light yellow solid (22, 820 mg, 82%). MS (ESI)[M+H+]+=196.1.
  • Additional compounds were prepared following the protocol of Scheme 7, either by substituting pyridine-3-boronic acid with an appropriate boronic acid or by substituting the 5-bromo-7-azaindole with 5-(4,4,5,5-Tetramethyl-[1,3,2]dioxaborolan-2-yl)-1H-pyrrolo[2,3-b]pyridine and reacting with a suitable aryl or heteroaryl halide (i.e. coupling with the boronic acid ester on the azaindole, and the halide on the group to be coupled to the 5-position of the azaindole). The following compounds were prepared by this procedure:
    • 5-(4-Chloro-phenyl)-1H-pyrrolo[2,3-b]pyridine (23),
    • 5-(4-Fluoro-phenyl)-1H-pyrrolo[2,3-b]pyridine (24),
    • 5-Phenyl-1H-pyrrolo[2,3-b]pyridine (25),
    • 5-(6-Methoxy-pyridin-3-yl)-1H-pyrrolo[2,3-b]pyridine (26),
    • 5-(2-Methoxy-pyrimidin-5-yl)-1H-pyrrolo[2,3-b]pyridine (27),
    • 5-Pyridin-4-yl-1H-pyrrolo[2,3-b]pyridine (28),
    • 4-(1H-Pyrrolo[2,3-b]pyridin-5-yl)-benzenesulfonamide (29),
    • 3-(1H-Pyrrolo[2,3-b]pyridin-5-yl)-benzenesulfonamide (30),
    • 5-Pyrimidin-5-yl-1H-pyrrolo[2,3-b]pyridine (31),
    • 5-(3-Methanesulfonyl-phenyl)-1H-pyrrolo[2,3-b]pyridine (32),
    • 3-(1H-Pyrrolo[2,3-b]pyridin-5-yl)-benzamide (33),
    • 5-(5-Methyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine (34),
    • 5-(1-Methyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine (35), and
    • 5-(1,5-Dimethyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine (59).
      The following table indicates either 5-bromo-7-azaindole or 5-(4,4,5,5-Tetramethyl-[1,3,2]dioxaborolan-2-yl)-1H-pyrrolo[2,3-b]pyridine starting material (column 1) and the appropriate reagent to be coupled to the 5 position of the azaindole (column 2) to afford the resulting compound (column 3), with the observed mass given in column 4
  • MS(ESI)
    Reagent coupled to [M + H+]+
    Starting azaindole 5 position Compound observed
    Figure US20110092538A1-20110421-C00032
    Figure US20110092538A1-20110421-C00033
    Figure US20110092538A1-20110421-C00034
    229.1
    Figure US20110092538A1-20110421-C00035
    Figure US20110092538A1-20110421-C00036
    Figure US20110092538A1-20110421-C00037
    213.1
    Figure US20110092538A1-20110421-C00038
    Figure US20110092538A1-20110421-C00039
    Figure US20110092538A1-20110421-C00040
    195.2
    Figure US20110092538A1-20110421-C00041
    Figure US20110092538A1-20110421-C00042
    Figure US20110092538A1-20110421-C00043
    226.2
    Figure US20110092538A1-20110421-C00044
    Figure US20110092538A1-20110421-C00045
    Figure US20110092538A1-20110421-C00046
    227.2
    Figure US20110092538A1-20110421-C00047
    Figure US20110092538A1-20110421-C00048
    Figure US20110092538A1-20110421-C00049
    196.2
    Figure US20110092538A1-20110421-C00050
    Figure US20110092538A1-20110421-C00051
    Figure US20110092538A1-20110421-C00052
    274.1
    Figure US20110092538A1-20110421-C00053
    Figure US20110092538A1-20110421-C00054
    Figure US20110092538A1-20110421-C00055
    274.1
    Figure US20110092538A1-20110421-C00056
    Figure US20110092538A1-20110421-C00057
    Figure US20110092538A1-20110421-C00058
    197.2
    Figure US20110092538A1-20110421-C00059
    Figure US20110092538A1-20110421-C00060
    Figure US20110092538A1-20110421-C00061
    273.1
    Figure US20110092538A1-20110421-C00062
    Figure US20110092538A1-20110421-C00063
    Figure US20110092538A1-20110421-C00064
    238.2
    Figure US20110092538A1-20110421-C00065
    Figure US20110092538A1-20110421-C00066
    Figure US20110092538A1-20110421-C00067
    199.2
    Figure US20110092538A1-20110421-C00068
    Figure US20110092538A1-20110421-C00069
    Figure US20110092538A1-20110421-C00070
    199.2
    Figure US20110092538A1-20110421-C00071
    Figure US20110092538A1-20110421-C00072
    Figure US20110092538A1-20110421-C00073
    213.2
  • Example 11 Synthesis of 3-Iodo-1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridine 37
  • 3-Iodo-1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridine 37 was synthesized in one step from 3-Iodo-1H-pyrrolo[2,3-b]pyridine 36 as shown in Scheme 8.
  • Figure US20110092538A1-20110421-C00074
  • Step 1—Preparation of 3-Iodo-1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridine (37)
  • 3-Iodo-1H-pyrrolo[2,3-b]pyridine 36 (2.00 g, 8.20 mmol) was dissolved in N,N-dimethylformamide (50 mL). Sodium hydride (60% dispersion in mineral oil, 390 mg, 9.8 mmol) was added. After 20 minutes, triisopropylsilyl chloride (1.74 mL, 8.20 mmol) was added dropwise. After 1.5 hours, the reaction was poured into water and extracted with ethyl acetate, washed with saturated sodium bicarbonate and brine. The organic portions were dried over anhydrous sodium sulfate and concentrated. Purification by silica gel chromatography, 0-25% gradient ethyl acetate/hexane gave compound 37 as a white solid (3.224 g, 98.2%). 1H-NMR was consistent with the desired compound.
  • Example 12 Synthesis of 1-(tert-Butyl-dimethyl-silanyl)-3-iodo-1H-pyrrolo[2,3-b]pyridine 38
  • 1-(tert-Butyl-dimethyl-silanyl)-3-iodo-1H-pyrrolo[2,3-b]pyridine 38 was synthesized in one step from 3-Iodo-1H-pyrrolo[2,3-b]pyridine 36 as shown in Scheme 9.
  • Figure US20110092538A1-20110421-C00075
  • Step 1—Preparation of 1-(tert-Butyl-dimethyl-silanyl)-3-iodo-1H-pyrrolo[2,3-b]pyridine (38)
  • 3-Iodo-1H-pyrrolo[2,3-b]pyridine 36 (1.11 g, 4.6 mmol) was dissolved in tetrahydrofuran (120 mL). Sodium hydride (60% dispersion in mineral oil, 0.13 g, 5.5 mmol) was added, followed by tert-butyldimethylsilyl chloride (0.85 g, 5.5 mmol). The reaction was stirred at room temperature overnight. The reaction was poured into water and extracted with ethyl acetate. The organic portion was washed with brine, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated and purified with silica gel column chromatography eluting with 30% ethyl acetate in hexane to give the compound as a white solid (38, 100 mg, 15%).
  • Example 13 Synthesis of 5-chloro-3-iodo-1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridine 40
  • 5-Chloro-3-iodo-1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridine 40 was synthesized in one step from 5-Chloro-3-iodo-1H-pyrrolo[2,3-b]pyridine 39 as shown in Scheme 10.
  • Figure US20110092538A1-20110421-C00076
  • Step 1—Preparation of 5-Chloro-3-iodo-1-triisopropylsilanyl-1H-pyrrolo[2,3-b]pyridine (40)
  • 5-Chloro-3-iodo-1H-pyrrolo[2,3-b]pyridine (39, 31.2 g, 0.112 mol) was dissolved in N-methylpyrrolidinone (800 mL) and NaH (60% dispersion, 4.93 g, 0.123 mol) was added at room temperature. The resulting mixture was stirred for 30 minutes. To this mixture was then added triisopropylsilylchloride (24.0 mL, 0.112 mol) and the resulting mixture was stirred for 2 hours. The reaction was quenched with water and extracted with ethyl acetate three times, washed by brine, dried, filtered, and concentrated in vacuo. The residue was subjected to silica gel flash chromatography (eluted by heptane to 5% ethyl acetate/heptane) to afford the desired compound (43 g, 88%) as a pale-yellow solid.
  • Example 14 Synthesis of 5-[4-(2-methoxyethoxy)-phenyl]-1H-pyrrolo[2,3-b]pyridine 44
  • 5-[4-(2-Methoxyethoxy)-phenyl]-1H-pyrrolo[2,3-b]pyridine 44 was synthesized in two steps from 4-bromophenol 41 as shown in Scheme 11.
  • Figure US20110092538A1-20110421-C00077
  • Step 1—Preparation 1-Bromo-4-(2-methoxy-ethoxy)-benzene (42)
  • To a solution of 4-bromophenol (41, 5.0 g, 28.9 mmol) in dimethylformamide (15 mL) were added potassium carbonate (4.40 g, 31.8 mmol) and 1-bromo-2-methoxyethane (5.00 g, 36.0 mmol) under an atmosphere of nitrogen. The reaction mixture was stirred at ambient temperature overnight and concentrated under reduced pressure. The residue was slurried in ethyl acetate (50 mL) and filtered. The filtrate was washed with saturated sodium bicarbonate solution, dried over magnesium sulfate and filtered. Silica gel column chromatography (0-10% ethyl acetate in hexanes) gave the desired compound as a colorless oil (42, 3.2 g, 48%).
  • Step 2—Preparation of 5-[4-(2-Methoxy-ethoxy)-phenyl]-1H-pyrrolo[2,3-b]pyridine (44)
  • To a solution of 5-(4,4,5,5,-tetramethyl-[1,3,2]dioxaborolan-2-yl)-1H-pyrrolol[2,3-b]pyridine (43, 1.1 g, 4.3 mmol) in tetrahydrofuran (40 mL) was added 1-bromo-4-(2-methoxy-ethoxy)-benzene (42, 1.50 g, 6.49 mmol) and tetrakis(triphenylphosphine)palladium (0) (0.25 g, 0.21 mmol). The reaction mixture was stirred with potassium carbonate solution (10 mL, 1.0 M) and warmed to reflux overnight. The biphasic reaction mixture was diluted with ethyl acetate (50 mL) and saturated sodium carbonate solution (20 mL). The organic layer was separated, washed with brine, dried over magnesium sulfate and purified by silica gel column chromatography (50-100% ethyl acetate in hexanes) to give the desired compound as a colorless solid (44, 782 mg, 67%). MS (ESI) [M+H+]+=267.4.
  • Example 15 Synthesis of 5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine 46
  • 5-(1-Methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine 46 was synthesized in one step from 5-bromo-1H-pyrrolo[2,3-b]pyridine 1 as shown in Scheme 12.
  • Figure US20110092538A1-20110421-C00078
  • Step 1—Preparation of 5-(1-Methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine (46)
  • To 5-bromo-7-azaindole (1, 1.04 g, 5.28 mmol) in 1.00 M potassium carbonate in water (15.8 mL) and tetrahydrofuran (50.0 mL) were added 1-methyl-4-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-1H-pyrazole (45, 1.65 g, 7.92 mmol), Tetrakis(triphenylphosphine)-palladium(0) (0.305 mg, 0.26 mmol) and tetra-n-butylammonium iodide (0.20 g, 0.53 mmol). The reaction mixture was stirred at 70° C. overnight. The reaction mixture was poured into water and the organic layer was washed with brine, dried over sodium sulfate, and concentrated. The residue was purified with silica gel column chromatography eluting with 25% ethyl acetate in hexane to provide a light yellow solid (46, 670 mg, 64.0%). MS (ESI)[M+H+]+=199.4.
  • Example 16 Preparation of propane-2-sulfonic acid [2,4-difluoro-3-(5-pyridin-3-yl-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-phenyl]-amide 51
  • Propane-2-sulfonic acid [2,4-difluoro-3-(5-pyridin-3-yl-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-phenyl]-amide 51 was synthesized in four steps from 2,4-difluorophenylamine 47 as shown in Scheme 13.
  • Figure US20110092538A1-20110421-C00079
  • Step 1—Preparation of propane-2-sulfonic acid (2,4-difluoro-phenyl)-amide (48)
  • To 2,4-difluoro-phenylamine (47, 4.0 mL, 40.0 mmol) in dichloromethane (50 mL) were added pyridine (3.37 mL, 42.3 mmol), propane-2-sulfonyl chloride (6.00 g, 42.3 mmol) and dimethylaminopyridine (0.20 g, 1.64 mmol) under an atmosphere of nitrogen. The reaction was stirred at 45° C. overnight. The reaction was poured into water and extracted with ethyl acetate. The organic layer was washed with brine, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated and purified by silica gel column chromatography 3% methanol in methylene chloride to give a white solid (48, 8.0 g, 85%). MS (ESI) [M−H+]=234.0.
  • Step 2—Preparation of propane-2-sulfonic acid (2,4-difluoro-3-formyl-phenyl)-amide (49)
  • To propane-2-sulfonic acid (2,4-difluoro-phenyl)-amide (48, 2.35 g, 9.95 mmol) in tetrahydrofuran (70 mL) under an atmosphere of nitrogen cooled with a dry ice/acetone bath was added 1.60 M of n-butyllithium (1.60 M in hexane, 6.53 mL, 10.45 mmol). The reaction was stirred for 40 minutes, and then another portion of n-butyllithium (1.60 M in hexane, 6.84 mL, 10.94 mmol). The reaction was stirred for 1 hour and N,N-dimethylformamide (0.92 mL, 11.9 mmol) was added. The reaction was allowed to warm to room temperature overnight. The reaction was poured into water extracted with ethyl acetate. The organic layer was washed with brine, dried over anhydrous sodium sulfate and filtrated. The filtrate was concentrated and purified by silica gel column chromatography (dichloromethane/methanol 5%) to give the compound (49, 1.4 g, 53.4%). MS (ESI) [M−H+]=263.4.
  • Step 3—Preparation of propane-2-sulfonic acid {2,4-difluoro-3-[hydroxy-(5-pyridin-3-yl-1H-pyrrolo[2,3-b]pyridin-3-yl)-methyl]-phenyl}-amide (50)
  • To propane-2-sulfonic acid (2,4-difluoro-3-formyl-phenyl)-amide (49, 220.0 mg, 0.83 mmol) in methanol (15 mL) was added 5-pyridin-3-yl-1H-pyrrolo[2,3-b]pyridine (22, 150.0 mg, 0.77 mmol, prepared as described in Example 10) and potassium hydroxide (537.0 mg, 9.6 mmol) under an atmosphere of nitrogen. The reaction was stirred at room temperature overnight. The reaction was poured into water and extracted with ethyl acetate. The organic layer was washed with brine, dried over sodium sulfate and filtrated. The filtrate was concentrated and purified by silica gel column chromatography eluting with 5% methanol in dichloromethane to give the compound (50, 160 mg, 45.3%). In this step, minor compound propane-2-sulfonic acid {2,4-difluoro-3-[methoxy-(5-pyridin-3-yl-1H-pyrrolo[2,3-b]pyridin-3-yl)-methyl]-phenyl}-amide was also formed and isolated. MS (ESI) [M+H+]+=460.1.
  • Step 4—Preparation of propane-2-sulfonic acid [2,4-difluoro-3-(5-pyridin-3-yl-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-phenyl]-amide (51)
  • To propane-2-sulfonic acid {2,4-difluoro-3-[hydroxy-(5-pyridin-3-yl-1H-pyrrolo[2,3-b]pyridin-3-yl)-methyl]-phenyl}-amide (50, 40.0 mg, 0.087 mmol) in tetrahydrofuran (10 mL) was added Dess-Martin periodane (48.0 mg, 0.11 mmol). The reaction was stirred at room temperature for 5 minutes. The reaction was poured into sodium thiosulfate and potassium carbonate solution and extracted with ethyl acetate. The organic layer was washed with brine, dried over anhydrous sodium sulfate and filtrated. The filtrate was concentrated and purified by silica gel column chromatography eluting with 5% methanol in methylene chloride to give the compound (51, 13.4 mg, 33.5%). MS (ESI) [M+H+]+=458.1.
  • N-{2,4-Difluoro-3-[5-(2-methoxy-ethoxy)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-isopropyl-benzenesulfonamide P-0018
  • Figure US20110092538A1-20110421-C00080
  • was prepared following the protocol of Scheme 13, substituting propane-2-sulfonyl chloride with 4-isopropyl-phenyl sulfonyl chloride in step 1 and 5-pyridin-3-yl-1H-pyrrolo[2,3-b]pyridine 22 with 5-(2-methoxy-ethoxy)-1H-pyrrolo[2,3-b]pyridine 19 (see Example 9) in step 3. MS (ESI) [M−H+]=530.
  • N-{2,4-Difluoro-3-[5-(4-methyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-isopropyl-benzenesulfonamide P-0019
  • Figure US20110092538A1-20110421-C00081
  • was prepared following the protocol of Scheme 13, substituting propane-2-sulfonyl chloride with 4-isopropyl-phenyl sulfonyl chloride in Step 1 and 5-pyridin-3-yl-1H-pyrrolo[2,3-b]pyridine 22 with 5-(5-methyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine 34 (see Example 10) in step 3. MS (ESI) [M−H+]=536.
  • N-{2,4-Difluoro-3-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-isopropyl-benzenesulfonamide P-0022
  • Figure US20110092538A1-20110421-C00082
  • was prepared following the protocol of Scheme 13, substituting propane-2-sulfonyl chloride with 4-isopropyl-phenyl sulfonyl chloride in Step 1 and 5-pyridin-3-yl-1H-pyrrolo[2,3-b]pyridine 22 with 5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine 46 (see Example 15) in step 3. MS (ESI) [M−H+]=536.
  • N-{2,4-Difluoro-3-[5-(5-methyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-trifluoromethyl-benzenesulfonamide P-0025
  • Figure US20110092538A1-20110421-C00083
  • was prepared following the protocol of Scheme 13, substituting propane-2-sulfonyl chloride with 4-trifluoromethyl-phenyl sulfonyl chloride in Step 1 and 5-pyridin-3-yl-1H-pyrrolo[2,3-b]pyridine 22 with 5-(5-methyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine 34 (see Example 10). MS (ESI) [M−H+]=562.
  • N-{2,4-Difluoro-3-[5-(1-methyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-propyl-benzenesulfonamide P-0026
  • Figure US20110092538A1-20110421-C00084
  • was prepared following the protocol of Scheme 13, substituting propane-2-sulfonyl chloride with propane sulfonyl chloride in Step 1 and 5-pyridin-3-yl-1H-pyrrolo[2,3-b]pyridine 22 with 5-(5-methyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine 35 (see Example 10). MS (ESI) [M−H+]=536.2.
  • N-{3-[5-(1,5-Dimethyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-2,4-difluoro-phenyl}-4-propyl-benzenesulfonamide P-0027
  • Figure US20110092538A1-20110421-C00085
  • was prepared following the protocol of Scheme 13, substituting propane-2-sulfonyl chloride with propane sulfonyl chloride in Step 1 and 5-pyridin-3-yl-1H-pyrrolo[2,3-b]pyridine 22 with 5-(1,5-dimethyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine (59) (see Example 10). MS (ESI) [M−H+]+=550.1.
  • Example 17 Synthesis of N-[3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-3,5-difluorobenzenesulfonamide 58
  • Compound P-1841 was synthesized in six steps from 2,4-difluoroaniline 47 as shown in Scheme 14.
  • Figure US20110092538A1-20110421-C00086
    Figure US20110092538A1-20110421-C00087
  • Step 1—Preparation of (2,4-difluoro-phenyl)-carbamic acid benzyl ester (52)
  • To 2,4-difluoroaniline (47, 7.0 mL, 0.070 mol) in 100 mL of dichloromethane was added pyridine (11 mL, 0.14 mol) and benzyl chloroformate (11.9 mL, 0.0834 mol). The reaction mixture was stirred at ambient temperature for 1.5 hours. The reaction mixture was concentrated under reduced pressure and the residue was partitioned between ethyl acetate and KHSO4 solution. The organic layer was dried (MgSO4), concentrated and crystallized from hexanes to give compound 52 (15.6 g, 85%).
  • Step 2—Preparation of (2,4-difluoro-3-formyl-phenyl)-carbamic acid benzyl ester (53)
  • Into a round bottom flask was added (2,4-difluoro-phenyl)-carbamic acid benzyl ester (52, 3.83 g, 14.5 mmol) in tetrahydrofuran (148 mL, 1.82 mol). The solution was chilled to −78° C. and n-butyllithium (1.60 M in hexane, 19.1 mL, 30.0 mmol) was added over 30 minutes followed by the addition of, N,N-dimethylformamide (1.12 mL, 14.5 mol). The reaction mixture was allowed to warm to ambient temperature and was stirred overnight. The reaction mixture was poured into water and extracted with ethyl acetate. The organic layer was washed with brine, dried over sodium sulfate, concentrated and crystallized from ether to give compound 53 (3.0 g, 71%).
  • Step 3—Preparation of {2,4-difluoro-3-[hydroxy-(1H-pyrrolo[2,3-b]pyridin-3-yl)-methyl]-phenyl}-carbamic acid benzyl ester (54)
  • Into a round bottom flask was added 5-chloro-1H-pyrrolo[2,3-b]pyridine (6, 0.524 g, 3.43 mmol, prepared as described in Example 5) in methanol (5.00 mL, 0.123 mol). Potassium hydroxide (0.800 g, 14.2 mmol) and (2,4-difluoro-3-formyl-phenyl)-carbamic acid benzyl ester (53, 1.02 g, 3.5 mmol) were added and the reaction mixture was stirred overnight. The reaction mixture was poured into 1N HCl and extracted with ethyl acetate. The organic layer was washed with brine, dried over sodium sulfate, concentrated and crystallized from ethyl acetate to give compound 54 (710 mg, 46%). MS (ESI)[M+H+]+=444.
  • Step 4—Preparation of [2,4-difluoro-3-(1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-phenyl]-carbamic acid benzyl ester (55)
  • Into a round bottom flask was added {2,4-difluoro-3-[hydroxy-(1H-pyrrolo[2,3-b]pyridin-3-yl)-methyl]-phenyl}-carbamic acid benzyl ester (54, 1.01 g, 2.28 mmol) in tetrahydrofuran (5.00 mL, 0.0616 mol). Dess-Martin periodinane (1.20 g, 2.89 mmol) was added in portions. The reaction mixture was stirred at ambient temperature for 10 minutes, then poured into water and extracted with ethyl acetate. The organic layer was washed with brine, dried over sodium sulfate, concentrated and purified by silica gel chromatography to give compound 55 (914 mg, 91%). MS (ESI)[M+H+]+=442.
  • Step 5—Preparation of (3-Amino-2,6-difluoro-phenyl)-(5-chloro-1H-pyrrolo[2,3-b]pyridin-3-yl)-methanone (56)
  • [2,4-difluoro-3-(1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-phenyl]-carbamic acid benzyl ester (55, 800 mg, 1.81 mmol) was added to 10 M NaOH (15.00 mL) and warmed to reflux overnight. The reaction mixture was diluted with 30 mL of water and was extracted with ethyl acetate to give compound 56 (450 mg, 81%).
  • Step 6—Preparation of N-[3-(5-chloro-1H-pyrrolo[2,3-]-pyridine-3-carbonyl)-2,4-difluoro-phenyl]-3,5-difluorobenzenesulfonamide (58)
  • Into a microwave reaction vessel were combined (3-amino-2,6-difluoro-phenyl)-(5-chloro-1H-pyrrolo[2,3-b]pyridin-3-yl)-methanone (56, 50 mg, 0.16 mmol), 3,5-difluorobenzenesulfonyl chloride (57, 103 mg, 0.49 mmol), pyridine (0.5 mL, 6.1820 mol) and tetrahydrofuran (3.0 mL,). The reaction was warmed in the CEM microwave at 300 watts, 130° C. for 10 minutes. The reaction mixture was partitioned between ethyl acetate and brine. The organic layer was collected, dried over Na2SO4, filtered and concentrated. The compound (58) was isolated using column chromatography (silica, hexane:ethyl acetate 70:30) to obtain 36 mg (46%) compound. MS=482.0.
  • Additional compounds were prepared following the protocol of Scheme 14, optionally substituting 5-chloro-1H-pyrrolo[2,3-b]pyridine 6 with an appropriate azaindole (see Examples 5, 10 or 15) in Step 3 and/or 3,5-difluorobenzenesulfonyl chloride 57 with an appropriate sulfonyl chloride in Step 6. The following compounds were prepared by this procedure:
    • Benzo[b]thiophene-3-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide (P-0001),
    • 5-Methyl-2-trifluoromethyl-furan-3-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide (P-0002),
    • 5-Oxazol-5-yl-thiophene-2-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide (P-0003),
    • 3-[3-(5-Chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenylsulfamoyl]-benzoic acid (P-0004),
    • 2-Oxo-2H-chromene-6-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide (P-0005),
    • 5-Isoxazol-5-yl-thiophene-2-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide (P-0006),
    • 4-Butoxy-N-[3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-benzenesulfonamide (P-0007),
    • N-[3-(5-Chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-4-pyrazol-1-yl-benzenesulfonamide (P-0008),
    • Benzothiazole-6-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide (P-0009),
    • 1-Methyl-3-trifluoromethyl-1H-pyrazole-4-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide (P-0010),
    • N-[3-(5-Chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-4-isopropoxy-benzenesulfonamide (P-0011),
    • Benzo[1,2,5]thiadiazoe-5-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide (P-0013),
    • 4-tert-Butyl-N-[3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-benzenesulfonamide (P-0014),
    • N-[3-(5-Chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-4-propyl-benzenesulfonamide (P-0015),
    • N-[3-(5-Chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-3-difluoromethoxy-benzenesulfonamide (P-0016),
    • 5-Methyl-benzo[b]thiophene-2-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide (P-0017),
    • 3-Difluoromethoxy-N-{2,4-difluoro-3-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-benzenesulfonamide (P-0020),
    • N-{2,4-Difluoro-3-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-propyl-benzenesulfonamide (P-0021),
    • N-{2,4-Difluoro-3-[5-(5-methyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-propyl-benzenesulfonamide (P-0023), and
    • 3-Difluoromethoxy-N-{2,4-difluoro-3-[5-(5-methyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-benzenesulfonamide (P-0024),
    • 5-Methyl-thiophene-2-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide (P-0028),
    • 1-Methyl-1H-pyrazole-3-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide (P-0029),
    • N-[2,4-Difluoro-3-(5-methyl-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-phenyl]-4-trifluoromethyl-benzenesulfonamide (P-0030),
    • N-[3-(5-Cyano-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-4-trifluoromethyl-benzenesulfonamide (P-0031),
    • (E)-3-{3-[2,6-Difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl}-acrylic acid methyl ester (P-0032), and
    • Pyridine-2-sulfonic acid [2,4-difluoro-3-(5-methoxy-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-phenyl]-amide (P-0036).
      The following table indicates the azaindole used in step 3 (column 2) and the sulfonyl chloride used in step 6 (column 3) to afford the target compound (column 4). The compound number is provided in column 1, with the observed mass given in column 5.
  • MS(ESI)
    [M + H+]+
    Azaindole Sulfonyl chloride Compound observed
    P-0001
    Figure US20110092538A1-20110421-C00088
    Figure US20110092538A1-20110421-C00089
    Figure US20110092538A1-20110421-C00090
    503.96
    P-0002
    Figure US20110092538A1-20110421-C00091
    Figure US20110092538A1-20110421-C00092
    Figure US20110092538A1-20110421-C00093
    P-0003
    Figure US20110092538A1-20110421-C00094
    Figure US20110092538A1-20110421-C00095
    Figure US20110092538A1-20110421-C00096
    520.3
    P-0004
    Figure US20110092538A1-20110421-C00097
    Figure US20110092538A1-20110421-C00098
    Figure US20110092538A1-20110421-C00099
    491.9
    P-0005
    Figure US20110092538A1-20110421-C00100
    Figure US20110092538A1-20110421-C00101
    Figure US20110092538A1-20110421-C00102
    515.9
    P-0006
    Figure US20110092538A1-20110421-C00103
    Figure US20110092538A1-20110421-C00104
    Figure US20110092538A1-20110421-C00105
    521.1
    P-0007
    Figure US20110092538A1-20110421-C00106
    Figure US20110092538A1-20110421-C00107
    Figure US20110092538A1-20110421-C00108
    519.9
    P-0008
    Figure US20110092538A1-20110421-C00109
    Figure US20110092538A1-20110421-C00110
    Figure US20110092538A1-20110421-C00111
    514.3
    P-0009
    Figure US20110092538A1-20110421-C00112
    Figure US20110092538A1-20110421-C00113
    Figure US20110092538A1-20110421-C00114
    505.1
    P-0010
    Figure US20110092538A1-20110421-C00115
    Figure US20110092538A1-20110421-C00116
    Figure US20110092538A1-20110421-C00117
    520.3
    P-0011
    Figure US20110092538A1-20110421-C00118
    Figure US20110092538A1-20110421-C00119
    Figure US20110092538A1-20110421-C00120
    505.9
    P-0013
    Figure US20110092538A1-20110421-C00121
    Figure US20110092538A1-20110421-C00122
    Figure US20110092538A1-20110421-C00123
    505.9
    P-0014
    Figure US20110092538A1-20110421-C00124
    Figure US20110092538A1-20110421-C00125
    Figure US20110092538A1-20110421-C00126
    503.9
    P-0015
    Figure US20110092538A1-20110421-C00127
    Figure US20110092538A1-20110421-C00128
    Figure US20110092538A1-20110421-C00129
    490.3
    P-0016
    Figure US20110092538A1-20110421-C00130
    Figure US20110092538A1-20110421-C00131
    Figure US20110092538A1-20110421-C00132
    514.3
    P-0017
    Figure US20110092538A1-20110421-C00133
    Figure US20110092538A1-20110421-C00134
    Figure US20110092538A1-20110421-C00135
    517.9
    P-0020
    Figure US20110092538A1-20110421-C00136
    Figure US20110092538A1-20110421-C00137
    Figure US20110092538A1-20110421-C00138
    560
    P-0021
    Figure US20110092538A1-20110421-C00139
    Figure US20110092538A1-20110421-C00140
    Figure US20110092538A1-20110421-C00141
    536
    P-0023
    Figure US20110092538A1-20110421-C00142
    Figure US20110092538A1-20110421-C00143
    Figure US20110092538A1-20110421-C00144
    536
    P-0024
    Figure US20110092538A1-20110421-C00145
    Figure US20110092538A1-20110421-C00146
    Figure US20110092538A1-20110421-C00147
    560
    P-0028
    Figure US20110092538A1-20110421-C00148
    Figure US20110092538A1-20110421-C00149
    Figure US20110092538A1-20110421-C00150
    469
    P-0029
    Figure US20110092538A1-20110421-C00151
    Figure US20110092538A1-20110421-C00152
    Figure US20110092538A1-20110421-C00153
    453
    P-0030
    Figure US20110092538A1-20110421-C00154
    Figure US20110092538A1-20110421-C00155
    Figure US20110092538A1-20110421-C00156
    496
    P-0031
    Figure US20110092538A1-20110421-C00157
    Figure US20110092538A1-20110421-C00158
    Figure US20110092538A1-20110421-C00159
    507
    P-0032
    Figure US20110092538A1-20110421-C00160
    Figure US20110092538A1-20110421-C00161
    Figure US20110092538A1-20110421-C00162
    566
    P-0036
    Figure US20110092538A1-20110421-C00163
    Figure US20110092538A1-20110421-C00164
    Figure US20110092538A1-20110421-C00165
    445
  • Example 18 Synthesis of 3-{3-[2,6-difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl}-N-ethyl-propionamide P-0035
  • 3-{3-[2,6-Difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl}-N-ethyl-propionamide P-0035 was synthesized in three steps from (E)-3-{3-[2,6-Difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl}-acrylic acid methyl ester P-0032 as shown in Scheme 15.
  • Figure US20110092538A1-20110421-C00166
  • Step 1—Preparation of 3-{3-[2,6-difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl}-propionic acid methyl ester (P-0033)
  • To (E)-3-{3-[2,6-difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl}-acrylic acid methyl ester (P-0032, 229 mg, 0.41 mmol, prepared as described in Example 17) was added methanol (15 mL) and tetrahydrofuran (5 mL). The solution was degassed with nitrogen for 5 minutes, followed by the addition of Pd/C (10% weight, 30 mg). The reaction mixture was allowed to stir under an atmosphere of hydrogen at room temperature for 18 hrs. After removing Pd/C and volatiles, the residue was purified through a quick silica plug eluted with ethyl acetate. Solvent was removed to provide an off-white solid (P-0033, 222 mg, 96%). MS (ESI) [M+H+]+=568.3.
  • Step 2—Preparation of 3-{3-[2,6-difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl}-propionic acid (P-0034)
  • To 3-{3-[2,6-difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl}-propionic acid methyl ester (P-0033, 215 mg, 0.38 mmol) dissolved in tetrahydrofuran (30 mL) was added 1 N lithium hydroxide solution (15 mL). The mixture was heated at 50° C. for 2 hrs, then acidified to pH˜1-2 with 6 N HCl and extracted with ethyl acetate. The organic layer was washed with water and brine, dried over anhydrous magnesium sulfate, filtered and concentrated to provide an off-white solid (P-0034, 202 mg, 96%). MS (ESI) [M+H+]+=554.0.
  • Step 3—Preparation of 3-{3-[2,6-difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl}-N-ethyl-propionamide (P-0035)
  • To 3-{3-[2,6-difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl}-propionic acid (P-0034, 42 mg, 0.076 mmol) dissolved in tetrahydrofuran (3 mL) was added a 2.0 M solution of ethylamine in tetrahydrofuran (110 μL, 0.23 mmol) and N-(3-dimethylaminopropyl)-N′-ethylcarbodiimide hydrochloride (29 mg, 0.15 mmol). The reaction mixture was stirred at room temperature for 16 hrs, then extracted with ethyl acetate and water. The organic layer was washed with water and brine, dried over anhydrous magnesium sulfate, filtered and concentrated. The desired compound was isolated by silica gel column chromatography (methanol/dichloromethane), with the eluting solvents removed via lyophilizing to provide a white fluffy solid (P-0035, 13 mg, 30%). MS (ESI) [M+H+]+=581.0.
  • Example 19 Kinase Activity Assays
  • Assays for the activity of kinases, including, but not limited to, B-Raf, B-Raf V600E, B-Raf V600E/T529I and c-Raf-1 are known in the art, for example as described in U.S. patent application Ser. No. 11/473,347 (see also, PCT publication WO2007002433), the disclosure of which is hereby incorporated by reference in its entirety.
  • Representative compounds screened by at least one of the methods described in U.S. patent application Ser. No. 11/473,347, or by similar methods, having IC50 of less than 10 μM under the test conditions employed are shown in tables 1a (B-Raf), 1b (B-Raf V600E), 1c (B-Raf V600E/T529I), 1d (Btk), 1e (c-Raf-1), 1f (Flt1), 1g (Fms), 1h (Jnk1), 1i (Kdr), 1j (Kit), 1k (Src), 1l (TEC), 1m (p38), and 1n (HGK).
  • TABLE 1a
    Representative compounds with activity toward kinase B-Raf
    with IC50 ≦ 10 μM under the test conditions employed.
    B-Raf P-0001, P-0002, P-0003, P-0005, P-0006, P-0007, P-0009,
    P-0011, P-0013, P-0014, P-0015, P-0016, P-0017, P-0018,
    P-0019, P-0020, P-0021, P-0022, P-0023, P-0024, P-0025,
    P-0026, P-0027, P-0028, P-0030, P-0031, P-0032, P-0033,
    P-0034, P-0035, P-0036
  • TABLE 1b
    Representative compounds with activity toward kinase B-Raf V600E
    with IC50 ≦ 10 μM under the test conditions employed.
    B-Raf P-0001, P-0002, P-0003, P-0005, P-0006, P-0007, P-0009,
    V600E P-0013, P-0014, P-0015, P-0016, P-0017, P-0019, P-0020,
    P-0021, P-0022, P-0023, P-0024, P-0025, P-0028, P-0029,
    P-0030, P-0031, P-0032, P-0033, P-0034, P-0035
  • TABLE 1c
    Representative compounds with activity toward kinase B-Raf
    V600E/T529I with IC50 ≦ 10 μM under the test conditions employed.
    B-Raf P-0003, P-0005, P-0006
    V600E
    T529I
  • TABLE 1d
    Representative compounds with activity toward kinase Btk with
    IC50 ≦ 10 μM under the test conditions employed.
    Btk: P-0013, P-0020, P-0021, P-0022, P-0023, P-0024
  • TABLE 1e
    Representative compounds with activity toward kinase c-Raf-1 with
    IC50 ≦ 10 μM under the test conditions employed.
    c-Raf-1: P-0001, P-0002, P-0003, P-0005, P-0006, P-0007, P-0008,
    P-0009, P-0011, P-0013, P-0014, P-0015, P-0016, P-0017,
    P-0018, P-0019, P-0020, P-0021, P-0022, P-0023, P-0025,
    P-0030, P-0031
  • TABLE 1f
    Representative compounds with activity toward kinase Flt1 with
    IC50 ≦ 10 μM under the test conditions employed.
    Flt1: P-0016, P-0020, P-0025, P-0026, P-0031
  • TABLE 1g
    Representative compounds with activity toward kinase Fms with
    IC50 ≦ 10 μM under the test conditions employed.
    Fms: P-0001, P-0002, P-0003, P-0005, P-0006, P-0009, P-0010,
    P-0011, P-0013, P-0014, P-0015, P-0016, P-0020, P-0021,
    P-0022, P-0023, P-0024, P-0027, P-0028, P-0035
  • TABLE 1h
    Representative compounds with activity toward kinase Jnk1 with
    IC50 ≦ 10 μM under the test conditions employed.
    Jnk1: P-0019, P-0020, P-0025
  • TABLE 1i
    Representative compounds with activity toward kinase Kdr with
    IC50 ≦ 10 μM under the test conditions employed.
    Kdr: P-0001, P-0002, P-0003, P-0004, P-0005, P-0008, P-0010,
    P-0011, P-0013, P-0014, P-0016, P-0018, P-0019, P-0020,
    P-0021, P-0022, P-0023, P-0024, P-0025, P-0026, P-0027,
    P-0028, P-0029, P-0032, P-0034
  • TABLE 1j
    Representative compounds with activity toward kinase Kit with
    IC50 ≦ 10 μM under the test conditions employed.
    Kit: P-0001, P-0002, P-0003, P-0005, P-0006, P-0007, P-0008, P-0009,
    P-0010, P-0011, P-0013, P-0014, P-0015, P-0016, P-0017, P-0018,
    P-0019, P-0020, P-0021, P-0022, P-0023, P-0024, P-0025, P-0026,
    P-0027, P-0028, P-0029, P-0032, P-0035
  • TABLE 1k
    Representative compounds with activity toward kinase Src with
    IC50 ≦ 10 μM under the test conditions employed.
    Src: P-0002, P-0003, P-0004, P-0005, P-0006, P-0008, P-0009, P-0010,
    P-0011, P-0013, P-0015, P-0016, P-0017, P-0018, P-0019, P-0020,
    P-0021, P-0022, P-0023, P-0024, P-0025, P-0026, P-0028, P-0029,
    P-0031, P-0036
  • TABLE 1l
    Representative compounds with activity toward kinase TEC with
    IC50 ≦ 10 μM under the test conditions employed.
    TEC: P-0016
  • TABLE 1m
    Representative compounds with activity toward kinase p38 with
    IC50 ≦ 10 μM under the test conditions employed.
    P38: P-0020, P-0028, P-0029
  • TABLE 1n
    Representative compounds with activity toward kinase HGK with
    IC50 ≦ 10 μM under the test conditions employed.
    HGK: P-0020
  • Example 19 Efficacy of Compounds in Combination with Standard-of-Care Chemotherapeutic Agents in Four Human Cancer Cell Lines
  • Compounds of the invention, such as compounds of Formula I, Formula II, or Formula III, in combination with a standard chemotherapeutic agent, such as 5-fluorouracil, carboplatin, dacarbazine, gefitinib, oxaliplatin, paclitaxel, SN-38, temozolomide, or vinblastine, can be assessed for their effectiveness in killing human tumor cells. Human tumor cell lines, such as A-375 (malignant melanoma), SK-MEL-2 (malignant melanoma, skin metastasis), COLO 205 (colorectal adenocarcinoma, ascites metastasis) or SW-620 (colorectal adenocarcinoma, lymph node metastasis) can be treated with a compound of Formula I, Formula II, or Formula III alone, or in combination with one of the above-mentioned chemotherapeutic agents.
  • Tumor cells are grown as a monolayer at 37° C. in a humidified atmosphere (5% CO2, 95% air). Cells are grown in a suitable culture medium, e.g. RPMI 1640 (Ref BE12-702F, Cambrex, Verviers, Belgium) containing 2 mM L-glutamine and supplemented with 10% fetal bovine serum (Ref DE14-801E, Cambrex). For experimental use, the tumor cells are detached from the culture flask by a 5-minute treatment with trypsin-versene (Ref 02-007E, Cambrex), diluted in Hanks' medium without calcium or magnesium (Ref BE10-543F, Cambrex). Trypsin treatment is neutralized by culture medium addition. The cells are counted in a hemocytometer and their viability assessed by 0.25% trypan blue exclusion.
  • The cell lines are checked for mycoplasma contamination with the Mycotect assay kit (Ref 15672-017, Invitrogen, Cergy-Pontoise, France) in accordance with the manufacturer's instructions. The mycoplasma test is assayed from the culture supernatants of the cell lines and compared to negative and positive controls.
  • The tumor cells (10,000 per well) are plated in 96-well flat-bottom microtitration plates (Ref 055260, Nunc, Dutscher, Brumath, France) and incubated at 37° C. for 24 hours before treatment in 100 μl of drug-free culture medium supplemented with 10% FBS. In order to assess the IC50 of each compound to be used for each cell line, the tumor cells are incubated in a 200 μl final volume of RPMI 1640 supplemented with 10% FBS and containing either a compound of Formula I, Formula II, or Formula III, or one of 5-fluorouracil, carboplatin, dacarbazine, gefitinib, oxaliplatin, paclitaxel, SN-38, temozolomide, or vinblastine. The compounds are tested in a suitable concentration range, such as 10−8 to 10−3 M for a compound of Formula I, Formula II, or Formula III, 5-fluorouracil, dacarbazine or gefitinib, 10−9 to 10−4 M for carboplatin, oxaliplatin, or temozolomide, 10−11 to 10−6M for paclitaxel or SN-38, and 10−15 to 10−10 M for vinblastine. Compounds of Formula I, Formula II, or Formula III are dissolved in DMSO and diluted with culture medium to the desired concentrations. 5-fluorouracil (50 mg/ml, Dakota Pharm, LePlessis Robinson, France), carboplatin (10 mg/ml, Aguettant, Lyon, France), and paclitaxel (6 mg/ml, Bristol-Myers Squibb SpA, Rueil Malmaison, France), are diluted with culture medium to the desired concentrations. Dacarbazine (Sigma, Saint Quentin Fallavier, France) and vinblastine (Lilly France S.A., Saint Cloud, France) are dissolved in NaCl 0.9% and diluted with culture medium to the desired concentrations. Gefitinib is dissolved in a mixed solution of RPMI 1640 and DMSO and diluted with culture medium to the desired concentrations (maximum final DMSO of 0.1% v/v). SN-38 (LKT Laboratories, Inc., St. Paul, Minn.) is dissolved in DMSO and diluted with culture medium to the desired concentrations (maximum final DMSO of 0.1% v/v). Temozolomide (LKT Laboratories, Inc., St. Paul, Minn.) is dissolved in water for injection and diluted with culture medium to the desired concentrations. Cells are incubated for 96 hours in the presence of test substances at 37° C. under 5% CO2. At the end of treatments, the cytotoxic activity is evaluated by an MTT assay.
  • For the MTT assay, at the end of the cells treatment, 20 μl of a 5 mg/ml solution 0.22 μm filtered tetrazolium reagent (MTT, Ref M2128, Sigma) in Phosphate Buffered Saline (PBS, Ref BE17-517Q, Cambrex), is added in each well. Culture plates are incubated for 2 h at 37° C. The resulting supernatant is removed and formazan crystals dissolved with 200 μl of DMSO per well. Absorbency (OD) is measured at 570 nm in each well using VICTOR3™ 1420 multilabeled counter (Wallac, PerkinElmer, Courtaboeuf, France).
  • The IC50 for each compound on each cell line is determined from the OD measurements of each sample. The dose response inhibition of cell proliferation is expressed as:

  • IC=(OD of drug exposed cells/OD of drug free wells)×100.
  • The mean of multiple measurements for each concentration is plotted vs. the drug concentration. The dose-response curves are plotted using XLFit 3 (IDBS, United Kingdom). The IC50 (drug concentration to obtain 50% inhibition of cell proliferation) determination values are calculated using the XLFit 3 from semi-log curves. The IC50 value determined for each compound in each cell line is used to determine the concentration of a compound of Formula I, Formula II, or Formula III, and of the standard chemotherapeutic to be used in combination.
  • The cells are treated with a combination of five concentrations of a compound of Formula I, Formula II, or Formula III and five concentrations of one of 5-fluorouracil, carboplatin, dacarbazine, gefitinib, oxaliplatin, paclitaxel, SN-38, temozolomide, or vinblastine, based on the IC50 results. The compounds and cells are treated per the IC50 determination described above and assayed by the MTT assay.
  • The results are assessed to determine whether the combination is synergistic or antagonistic. The compound interactions are calculated by multiple drug effect analysis and are performed by the median equation principle according to the methodology described by Chou and Talalay (Adv. Enzyme Regul. 1984, 22: 27-55).
  • The combination index (CI) will be calculated by the Chou et al. equation (Adv. Enzyme Regul. 1984, 22: 27-55; Encyclopaedia of human biology, Academic Press, 1991, 2: 371-9; Synergism and Antagonism in Chemotherapy, Academic Press, 1991, 61-102) which takes into account both the potency (Dm or IC50) and the shape of the dose-effect curve (the m value). The general equation for the CI of the two compounds is given by:
  • CI = ( D ) 1 ( D x ) 1 + ( D ) 2 ( D x ) 2 + ( D ) 1 ( D ) 2 ( D x ) 1 ( D x ) 2
  • where:
    (DX)1 and (DX)2 in the denominators are the doses (or concentrations) for compound 1 and compound 2 alone which demonstrate x % of inhibition, whereas (D)1 and (D)2 in the numerators are doses of both compounds (1 and 2) in combination that also inhibit x % (iso-effective). CI<1, =1, and >1 indicate synergism, additive effect and antagonism, respectively.
  • The (DX)1 and (DX)2 can be calculated from the median-effect equation of Chou et al. (J. Natl. Cancer Inst. 1994, 86: 1517-24):
  • D x = D m ( f a ( 1 - f a ) ) 1 / m
  • where:
    Dm is the median-effect dose that is obtained from the anti-log of x-intercept of the median-effect plot, x=log(D) versus y=log{fa/(1−fa)}, or Dm=10−(y-intercept)/m; and m is the slope of the median-effect plot and fa is the fraction of cells affected by the treatment.
    Each CI will be calculated with CalcuSyn software (Biosoft, UK) from the mean affected fraction at each drug ratio concentration.
  • Additional examples of certain methods contemplated by the present invention may be found in the following applications: U.S. Patent Publ. No. 2006/058339, application Ser. No. 11/154,287; U.S. Patent Publ. No. 2006/058340, application Ser. No. 11/154,988; U.S. Prov. App. No. 60/682,076, filed May 17, 2005; U.S. Prov. App. No. 60/682,058, filed May 17, 2005; U.S. Prov. App. No. 60/682,063, filed May 17, 2005; U.S. Prov. App. No. 60/682,051, filed May 17, 2005; U.S. Prov. App. No. 60/682,042, filed May 17, 2005; U.S. Prov. App. No. 60/692,750, filed Jun. 22, 2005; and U.S. Prov. App. No. 60/692,960, filed Jun. 22, 2005; U.S. Prov. App. No. 60/731,528, filed Oct. 28, 2005, U.S. patent application Ser. No. 11/435,381, filed May 16, 2006, and U.S. patent application Ser. No. 11/473,347, filed Jun. 21, 2006, each of which are hereby incorporated by reference herein in their entireties including all specifications, figures, and tables, and for all purposes.
  • All patents and other references cited in the specification are indicative of the level of skill of those skilled in the art to which the invention pertains, and are incorporated by reference in their entireties, including any tables and figures, to the same extent as if each reference had been incorporated by reference in its entirety individually.
  • One skilled in the art would readily appreciate that the present invention is well adapted to obtain the ends and advantages mentioned, as well as those inherent therein. The methods, variances, and compositions described herein as presently representative of preferred embodiments are exemplary and are not intended as limitations on the scope of the invention. Changes therein and other uses will occur to those skilled in the art, which are encompassed within the spirit of the invention, are defined by the scope of the claims.
  • It will be readily apparent to one skilled in the art that varying substitutions and modifications may be made to the invention disclosed herein without departing from the scope and spirit of the invention. For example, variations can be made to crystallization or co-crystallization conditions for Ret and Ret surrogate proteins and/or various kinase domain sequences can be used. Thus, such additional embodiments are within the scope of the present invention and the following claims.
  • The invention illustratively described herein suitably may be practiced in the absence of any element or elements, limitation or limitations which is not specifically disclosed herein. Thus, for example, in each instance herein any of the terms “comprising”, “consisting essentially of” and “consisting of” may be replaced with either of the other two terms. Thus, for an embodiment of the invention using one of the terms, the invention also includes another embodiment wherein one of these terms is replaced with another of these terms. In each embodiment, the terms have their established meaning. Thus, for example, one embodiment may encompass a method “comprising” a series of steps, another embodiment would encompass a method “consisting essentially of” the same steps, and a third embodiment would encompass a method “consisting of” the same steps. The terms and expressions which have been employed are used as terms of description and not of limitation, and there is no intention that in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions thereof, but it is recognized that various modifications are possible within the scope of the invention claimed. Thus, it should be understood that although the present invention has been specifically disclosed by preferred embodiments and optional features, modification and variation of the concepts herein disclosed may be resorted to by those skilled in the art, and that such modifications and variations are considered to be within the scope of this invention as defined by the appended claims.
  • In addition, where features or aspects of the invention are described in terms of Markush groups or other grouping of alternatives, those skilled in the art will recognize that the invention is also thereby described in terms of any individual member or subgroup of members of the Markush group or other group.
  • Also, unless indicated to the contrary, where various numerical values are provided for embodiments, additional embodiments are described by taking any 2 different values as the endpoints of a range. Such ranges are also within the scope of the described invention.
  • Thus, additional embodiments are within the scope of the invention and within the following claims.

Claims (28)

1. A method for treating a subject suffering from or at risk of an A-Raf-mediated, B-Raf-mediated and/or c-Raf-1-mediated disease or condition, said method comprising administering to the subject an effective amount of a compound of Formula I:
Figure US20110092538A1-20110421-C00167
or a pharmaceutically acceptable salt, a tautomer or an isomer thereof, wherein:
R1 is selected from the group consisting of hydrogen, halogen, optionally substituted lower alkyl, optionally substituted lower alkenyl, optionally substituted lower alkynyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, —OH, —NH2, —CN, —NO2, —C(O)OH, —S(O)2NH2, —C(O)NH2, —C(S)NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —OR4, —SR4, —NR5R4, —C(O)R4, —C(S)R4, —C(O)OR4, —C(O)NR5R4, —C(S)NR5R4, —S(O)2NR5R4, —NR5C(O)R4, —NR5C(S)R4, —NR5S(O)2R4, —NR5C(O)NH2, —NR5C(O)NR5R4, —NR5C(S)NH2, —NR5C(S)NR5R4, —NR5S(O)2NH2, —NR5S(O)2NR5R4, —S(O)R4, and —S(O)2R4;
R2 is selected from the group consisting of halogen, optionally substituted lower alkyl, optionally substituted lower alkenyl, optionally substituted lower alkynyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, —OH, —NH2, —CN, —NO2, —C(O)OH, —S(O)2NH2, —C(O)NH2, —C(S)NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —OR4, —SR4, —NR5R4, —C(O)R4, —C(S)R4, —C(O)OR4, —C(O)NR5R4, —C(S)NR5R4, —S(O)2NR5R4, —NR5C(O)R4, —NR5C(S)R4, —NR5S(O)2R4, —NR5C(O)NH2, —NR5C(O)NR5R4, —NR5C(S)NH2, —NR5C(S)NR5R4, —NR5S(O)2NH2, —NR5S(O)2NR5R4, —S(O)R4, and —S(O)2R4;
R3 is selected from the group consisting of hydrogen, fluoro and chloro;
R4 is selected from the group consisting of optionally substituted lower alkyl, optionally substituted lower alkenyl, provided, however, that when R4 is optionally substituted lower alkenyl, no alkene carbon thereof is bound to N, S, O, S(O), S(O)2, C(O) or C(S) of —OR4, —SW, —NR5R4, —C(O)R4, —C(S)R4, —C(O)OR4, —C(O)NR5R4, —C(S)NR5R4, —S(O)2NR5R4, —NR5C(O)R4, —NR5C(S)R4, —NR5S(O)2R4, —NR5C(O)NH2, —NR5C(O)NR5R4, —NR5C(S)NH2, —NR5C(S)NR5R4, —NR5S(O)2NH2, —NR5S(O)2NR5R4, —S(O)R4, or —S(O)2R4, optionally substituted lower alkynyl, provided, however, that when R4 is optionally substituted lower alkynyl, no alkyne carbon thereof is bound to N, S, O, S(O), S(O)2, C(O) or C(S) of —OR4, —SR4, —NR5R4, —C(O)R4, —C(S)R4, —C(O)OR4, —C(O)NR5R4, —C(S)NR5R4, —S(O)2NR5R4, —NR5C(O)R4, —NR5C(S)R4, —NR5S(O)2R4, —NR5C(O)NH2, —NR5C(O)NR5R4, —NR5C(S)NH2, —NR5C(S)NR5R4, —NR5S(O)2NH2, —NR5S(O)2NR5R4, —S(O)R4, or —S(O)2R4, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; and
R5 is selected from the group consisting of hydrogen and optionally substituted lower alkyl, provided, however, the compound is not
Figure US20110092538A1-20110421-C00168
Figure US20110092538A1-20110421-C00169
Figure US20110092538A1-20110421-C00170
Figure US20110092538A1-20110421-C00171
Figure US20110092538A1-20110421-C00172
2. The method of claim 1, wherein the compound is approved for administration to a human.
3. The method of claim 2, wherein the disease or condition is selected from the group consisting of multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease, Parkinson's disease, melanoma, glioma, sarcoma, cancer of the colon, lung, breast, pancreas, thyroid, ovary, liver, or kidney, medullary thyroid cancer, carcinoid, small cell lung cancer, pheochromocytoma, lymphoma, neurofibromatosis, myelodysplastic syndrome, leukemia, tumor angiogenesis, neuropathic pain, inflammatory pain, acute pain, chronic pain, cancer-related pain, migraine, heart failure, ischemic stroke, cardiac hypertrophy, thrombosis, atherosclerosis, reperfusion injury, psoriasis, arthritis, osteoarthritis, endometriosis, scarring, vascular restenosis, fibrotic disorders, rheumatoid arthritis, inflammatory bowel disease, immunodeficiency diseases, organ transplant rejection, graft versus host disease, diabetic nephropathy, polycystic kidney disease, nephrosclerosis, glomerulonephritis, prostate hyperplasia, obesity, Helicobacter pylori infection, Hepatitis infection, Influenza virus infection, fever, sepsis, chronic obstructive pulmonary disease, acute respiratory distress syndrome, Noonan's syndrome, Costello syndrome, LEOPARD syndrome, cardio-faciocutaneous syndrome, neural crest syndrome abnormalities causing cardiovascular, skeletal, intestinal, skin, hair and endocrine diseases, sarcopenia, muscular dystrophy, motor neuron diseases, inflammatory myopathies, diseases of the neuromuscular junction, myopathies due to endocrine abnormalities, diseases of peripheral nerve, myotonia congenita, paramyotonia congenita, central core disease, nemaline myopathy, myotubular myopathy, periodic paralysis and metabolic diseases of muscle.
4. The method of claim 3, wherein:
R1 is selected from the group consisting of hydrogen, halogen, lower alkyl, lower alkenyl, lower alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, —OH, —NH2, —CN, —NO2, —C(O)OH, —S(O)2NH2, —C(O)NH2, —C(S)NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —OR7, —SR7, —NR8R7, —C(O)R7, —C(S)R7, —C(O)OR7, —C(O)NR8R7, —C(S)NR8R7, —S(O)2NR8R7, —NR8C(O)R7, —NR8C(S)R7, —NR8S(O)2R7, —NR8C(O)NH2, —NR8C(O)NR8R7, —NR8C(S)NH2, —NR8C(S)NR8R7, —NR8S(O)2NH2, —NR8S(O)2NR8R7, —S(O)R7, and —S(O)2R7, wherein lower alkyl, lower alkenyl or lower alkynyl are optionally substituted with one or more substituents selected from fluoro, —OH, —NH2, C(O)OH, —C(O)NH2, —OR7, —NR8R7, —C(O)OR7, —C(O)NR8R7, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as R1 or as substituents of lower alkyl, lower alkenyl or lower alkynyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —S(O)2NH2, —C(O)NH2, —OR9, —SR9, —NHR9, —NR8R9, —NR8C(O)R9, —NR8S(O)2R9, —S(O)2R9, —S(O)2NR8R9, —C(O)R9, —C(O)NR8R9, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino;
R2 is selected from the group consisting of halogen, lower alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, —OH, —NH2, —CN, —NO2, —C(O)OH, —S(O)2NH2, —C(O)NH2, —C(S)NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —OR7, —SR7, —NR8R7, —C(O)R7, —C(S)R7, —C(O)OR7, —C(O)NR8R7, —C(S)NR8R7, —S(O)2NR8R7, —NR8C(O)R7, —NR8C(S)R7, —NR8S(O)2R7, —NR8C(O)NH2, —NR8C(O)NR8R7, —NR8C(S)NH2, —NR8C(S)NR8R7, —NR8S(O)2NH2, —NR8S(O)2NR8R7, —S(O)R7, and —S(O)2R7, wherein lower alkyl is optionally substituted with one or more substituents selected from fluoro, —OR7, —NR8R7, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as R2 or as substituents of lower alkyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —S(O)2NH2, —C(O)NH2, —OR9, —SR9, —NHR9, —NR8R9, —NR8C(O)R9, —NR8S(O)2R9, —S(O)2R9, —S(O)2NR8R9, —C(O)R9, —C(O)NR8R9, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino;
R7 is selected from the group consisting of lower alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as R7 or as substituents of lower alkyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —S(O)2NH2, —C(O)NH2, —OR9, —SR9, —NHR9, —NR8R9, —NR8C(O)R9, —NR8S(O)2R9, —S(O)2R9, —S(O)2NR8R9, —C(O)R9, —C(O)NR8R9, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino;
R8 at each occurrence is independently hydrogen or lower alkyl; and
R9 at each occurrence is independently selected from the group consisting of lower alkyl, heterocycloalkyl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, and cycloalkylamino, and heterocycloalkyl and heteroaryl are optionally substituted with one or more substituents selected from the group consisting of halogen, —CN, lower alkyl, fluoro substituted lower alkyl, lower alkoxy and fluoro substituted lower alkoxy.
5. The method of claim 4, wherein
R1 is selected from the group consisting of hydrogen, —CN, —OR7, —SR7, —NR8R7, —NR8C(O)R7, —NR8S(O)2R7, —C(O)NR8R7, —C(O)R7, —S(O)2NR8R7, —S(O)R7, —S(O)2R7, halogen, lower alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as R1 or as substituents of lower alkyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —S(O)2NH2, —C(O)NH2, —OR9, —SR9, —NHR9, —NR8R9, —NR8C(O)R9, —NR8S(O)2R9, —S(O)2R9, —S(O)2NR8R9, —C(O)R9, —C(O)NR8R9, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino; and
R2 is selected from the group consisting of —CN, —OR7, —SR7, —NR8R7, —NR8C(O)R7, —NR8S(O)2R7, —C(O)NR8R7, —C(O)R7, —S(O)2NR8R7, —S(O)R7, —S(O)2R7, halogen, lower alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, and wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as R2 or as a substituent of lower alkyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —S(O)2NH2, —C(O)NH2, —OR9, —SR9, —NHR9, —NR8R9, —NR8C(O)R9, —NR8S(O)2R9, —S(O)2R9, —S(O)2NR8R9, —C(O)R9, —C(O)NR8R9, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino.
6. The method of claim 5, wherein:
R1 is hydrogen, —CN, —NR8R7, —OR7, —S(O)2R7, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, cycloalkyl, heterocycloalkyl, aryl or heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl or heteroaryl are optionally substituted with one or more substituents selected from the group consisting of halogen, lower alkyl, fluoro substituted lower alkyl, —NR8R7, —OR7 and —S(O)2R7, and
R2 is —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, —NR8R7, —OR7 or —S(O)2R7.
7. The method of claim 6, wherein:
R1 is hydrogen, —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower alkoxy substituted C2-6alkoxy, aryl or heteroaryl, wherein aryl or heteroaryl are optionally substituted with one or more substituents selected from the group consisting of fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, mono-alkylamino, di-alkylamino, and cycloalkylamino, and
R2 is —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, mono-alkylamino, di-alkylamino, or cycloalkylamino.
8. The method of claim 3, wherein the compound is selected from the group consisting of:
4-Butoxy-N-[3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-benzenesulfonamide,
N-[3-(5-Chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-4-pyrazol-1-yl-benzenesulfonamide,
N-[3-(5-Chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-4-isopropoxy-benzenesulfonamide,
4-tert-Butyl-N-[3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-benzenesulfonamide,
N-[3-(5-Chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-4-propyl-benzenesulfonamide,
N-{2,4-Difluoro-3-[5-(2-methoxy-ethoxy)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-isopropyl-benzenesulfonamide,
N-{2,4-Difluoro-3-[5-(4-methyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-isopropyl-benzenesulfonamide,
4-Difluoromethoxy-N-{2,4-difluoro-3-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-benzenesulfonamide,
N-{2,4-Difluoro-3-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-propyl-benzenesulfonamide,
N-{2,4-Difluoro-3-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-isopropyl-benzenesulfonamide,
N-{2,4-Difluoro-3-[5-(5-methyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-propyl-benzenesulfonamide,
N-{2,4-Difluoro-3-[5-(5-methyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-trifluoromethyl-benzenesulfonamide,
N-{2,4-Difluoro-3-[5-(1-methyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-propyl-benzenesulfonamide,
N-{3-[5-(1,5-Dimethyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-2,4-difluoro-phenyl}-4-propyl-benzenesulfonamide,
N-[2,4-Difluoro-3-(5-methyl-1H-pyrrolo[2, 3-1)]pyridine-3-carbonyl)-phenyl]-4-trifluoromethyl-benzene sulfonamide,
N-[3-(5-Cyano-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-4-trifluoromethyl-benzene sulfonamide,
(E)-3-{3-[2,6-Difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl}-acrylic acid methyl ester,
3-{3-[2,6-Difluoro-3-(4-trifluoromethyl-benzene sulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl}-prop ionic acid methyl ester,
3-{3-[2,6-Difluoro-3-(4-trifluoromethyl-benzene sulfonylamino)-benzoyl]-1H-pyrrolo[2, 3-1)]pyridin-5-yl}-propionic acid,
3-{3-[2,6-Difluoro-3-(4-trifluoromethyl-benzene sulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl}-N-ethyl-propionamide; and
pharmaceutically acceptable salts, tautomers, and isomers thereof.
9. A method for treating a subject suffering from or at risk of an A-Raf-mediated, B-Raf-mediated and/or c-Raf-1-mediated disease or condition, said method comprising administering to the subject an effective amount of a compound of Formula II:
Figure US20110092538A1-20110421-C00173
or a pharmaceutically acceptable salt, a tautomer or an isomer thereof, wherein:
R1 is selected from the group consisting of hydrogen, halogen, optionally substituted lower alkyl, optionally substituted lower alkenyl, optionally substituted lower alkynyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, —OH, —NH2, —CN, —NO2, —C(O)OH, —S(O)2NH2, —C(O)NH2, —C(S)NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —OR7, —SR4, —NR5R4, —C(O)R4, —C(S)R4, —C(O)OR4, —C(O)NR5R4, —C(S)NR5R4, —S(O)2NR5R4, —NR5C(O)R4, —NR5C(S)R4, —NR5S(O)2R4, —NR5C(O)NH2, —NR5C(O)NR5R4, —NR5C(S)NH2, —NR5C(S)NR5R4, —NR5S(O)2NH2, —NR5S(O)2NR5R4, —S(O)R4, and —S(O)2R4;
R2a is selected from the group consisting of halogen, optionally substituted lower alkyl, optionally substituted lower alkenyl, optionally substituted lower alkynyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, —OH, —NH2, —CN, —NO2, —C(O)OH, —S(O)2NH2, —C(O)NH2, —C(S)NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —OR4, —SR4, —NR5R4, —C(O)R4, —C(S)R4, —C(O)OR4, —C(O)NR5R4, —C(S)NR5R4, —S(O)2NR5R4, —NR5C(O)R4, —NR5C(S)R4, —NR5S(O)2R4, —NR5C(O)NH2, —NR5C(O)NR5R4, —NR5C(S)NH2, —NR5C(S)NR5R4, —NR5S(O)2NH2, —NR5S(O)2NR5R4, —S(O)R4, and —S(O)2R4;
R3 is selected from the group consisting of hydrogen, fluoro and chloro;
R4 is selected from the group consisting of optionally substituted lower alkyl, optionally substituted lower alkenyl, provided, however, that when R4 is optionally substituted lower alkenyl, no alkene carbon thereof is bound to N, S, O, S(O), S(O)2, C(O) or C(S) of —OR4, —SR4, —NR5R4, —C(O)R4, —C(S)R4, —C(O)OR4, —C(O)NR5R4, —C(S)NR5R4, —S(O)2NR5R4, —NR5C(O)R4, —NR5C(S)R4, —NR5S(O)2R4, —NR5C(O)NH2, —NR5C(O)NR5R4, —NR5C(S)NH2, —NR5C(S)NR5R4, —NR5S(O)2NH2, —NR5S(O)2NR5R4, —S(O)R4, or —S(O)2R4, optionally substituted lower alkynyl, provided, however, that when R4 is optionally substituted lower alkynyl, no alkyne carbon thereof is bound to N, S, O, S(O), S(O)2, C(O) or C(S) of —OR4, —SR4, —NR5R4, —C(O)R4, —C(S)R4, —C(O)OR4, —C(O)NR5R4, —C(S)NR5R4, —S(O)2NR5R4, —NR5C(O)R4, —NR5C(S)R4, —NR5S(O)2R4, —NR5C(O)NH2, —NR5C(O)NR5R4, —NR5C(S)NH2, —NR5C(S)NR5R4, —NR5S(O)2NH2, —NR5S(O)2NR5R4, —S(O)R4, or —S(O)2R4, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; and
R5 is selected from the group consisting of hydrogen and optionally substituted lower alkyl, provided, however, the compound is not
Figure US20110092538A1-20110421-C00174
Figure US20110092538A1-20110421-C00175
Figure US20110092538A1-20110421-C00176
10. The method of claim 9, wherein the compound is approved for administration to a human.
11. The method of claim 10, wherein the disease or condition is selected from the group consisting of multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease, Parkinson's disease, melanoma, glioma, sarcoma, cancer of the colon, lung, breast, pancreas, thyroid, ovary, liver, or kidney, medullary thyroid cancer, carcinoid, small cell lung cancer, pheochromocytoma, lymphoma, neurofibromatosis, myelodysplastic syndrome, leukemia, tumor angiogenesis, neuropathic pain, inflammatory pain, acute pain, chronic pain, cancer-related pain, migraine, heart failure, ischemic stroke, cardiac hypertrophy, thrombosis, atherosclerosis, reperfusion injury, psoriasis, arthritis, osteoarthritis, endometriosis, scarring, vascular restenosis, fibrotic disorders, rheumatoid arthritis, inflammatory bowel disease, immunodeficiency diseases, organ transplant rejection, graft versus host disease, diabetic nephropathy, polycystic kidney disease, nephrosclerosis, glomerulonephritis, prostate hyperplasia, obesity, Helicobacter pylori infection, Hepatitis infection, Influenza virus infection, fever, sepsis, chronic obstructive pulmonary disease, acute respiratory distress syndrome, Noonan's syndrome, Costello syndrome, LEOPARD syndrome, cardio-faciocutaneous syndrome, neural crest syndrome abnormalities causing cardiovascular, skeletal, intestinal, skin, hair and endocrine diseases, sarcopenia, muscular dystrophy, motor neuron diseases, inflammatory myopathies, diseases of the neuromuscular junction, myopathies due to endocrine abnormalities, diseases of peripheral nerve, myotonia congenita, paramyotonia congenita, central core disease, nemaline myopathy, myotubular myopathy, periodic paralysis and metabolic diseases of muscle.
12. The method of claim 11, wherein:
R1 is selected from the group consisting of hydrogen, —CN, —OR7, —SR7, —NR8R7, —NR8C(O)R7, —NR8S(O)2R7, —C(O)NR8R7, —C(O)R7, —S(O)2NR8R7, —S(O)R7, —S(O)2R7, halogen, lower alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as R1 or as substituents of lower alkyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —S(O)2NH2, —C(O)NH2, —OR9, —SR9, —NHR9, —NR8R9, —NR8C(O)R9, —NR8S(O)2R9, —S(O)2R9, —S(O)2NR8R9, —C(O)R9, —C(O)NR8R9, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino; and
R2a is selected from the group consisting of —CN, —OR7, —SR7, —NR8R7, —NR8C(O)R7, —NR8S(O)2R7, —C(O)NR8R7, —C(O)R7, —S(O)2NR8R7, —S(O)R7, —S(O)2R7, halogen, lower alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, and wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as R2a or as a substituent of lower alkyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —S(O)2NH2, —C(O)NH2, —OR9, —SR9, —NHR9, —NR8R9, —NR8C(O)R9, —NR8S(O)2R9, —S(O)2R9, —S(O)2NR8R9, —C(O)R9, —C(O)NR8R9, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino.
13. The method of claim 12, wherein:
R1 is hydrogen, —CN, —NR8R7, —OR7, —S(O)2R7, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, cycloalkyl, heterocycloalkyl, aryl or heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl or heteroaryl are optionally substituted with one or more substituents selected from the group consisting of halogen, lower alkyl, fluoro substituted lower alkyl, —NR8R7, —OR7 and —S(O)2R7, and
R2a is —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, —NR8R7, —OR7 or —S(O)2R7.
14. The method of claim 13, wherein:
R1 is hydrogen, —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower alkoxy substituted C2-6alkoxy, aryl or heteroaryl, wherein aryl or heteroaryl are optionally substituted with one or more substituents selected from the group consisting of fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, mono-alkylamino, di-alkylamino, and cycloalkylamino, and
R2a is —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, mono-alkylamino, di-alkylamino, or cycloalkylamino.
15. The method of claim 11, wherein the compound is selected from the group consisting of:
3-[3-(5-Chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenylsulfamoyl]-benzoic acid,
N-[3-(5-Chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-3-difluoromethoxy-benzenesulfonamide,
3-Difluoromethoxy-N-{2,4-difluoro-3-[5-(5-methyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-benzenesulfonamide; and
pharmaceutically acceptable salts, tautomers, and isomers thereof.
16. A method for treating a subject suffering from or at risk of an A-Raf-mediated, B-Raf-mediated and/or c-Raf-1-mediated disease or condition, comprising administering to the subject an effective amount of a compound of Formula III:
Figure US20110092538A1-20110421-C00177
or a pharmaceutically acceptable salt, a tautomer or an isomer thereof, wherein:
R1 is selected from the group consisting of hydrogen, halogen, optionally substituted lower alkyl, optionally substituted lower alkenyl, optionally substituted lower alkynyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, —OH, —NH2, —CN, —NO2, —C(O)OH, —S(O)2NH2, —C(O)NH2, —C(S)NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —OR4, —SR4, —NR5R4, —C(O)R4, —C(S)R4, —C(O)OR4, —C(O)NR5R4, —C(S)NR5R4, —S(O)2NR5R4, —NR5C(O)R4, —NR5C(S)R4, —NR5S(O)2R4, —NR5C(O)NH2, —NR5C(O)NR5R4, —NR5C(S)NH2, —NR5C(S)NR5R4, —NR5S(O)2NH2, —NR5S(O)2NR5R4, —S(O)R4, and —S(O)2R4;
R3 is selected from the group consisting of hydrogen, fluoro and chloro;
R4 is selected from the group consisting of optionally substituted lower alkyl, optionally substituted lower alkenyl, provided, however, that when R4 is optionally substituted lower alkenyl, no alkene carbon thereof is bound to N, S, O, S(O), S(O)2, C(O) or C(S) of —OR4, —SW, —NR5R4, —C(O)R4, —C(S)R4, —C(O)OR4, —C(O)NR5R4, —C(S)NR5R4, —S(O)2NR5R4, —NR5C(O)R4, —NR5C(S)R4, —NR5S(O)2R4, —NR5C(O)NH2, —NR5C(O)NR5R4, —NR5C(S)NH2, —NR5C(S)NR5R4, —NR5S(O)2NH2, —NR5S(O)2NR5R4, —S(O)R4, or —S(O)2R4, optionally substituted lower alkynyl, provided, however, that when R4 is optionally substituted lower alkynyl, no alkyne carbon thereof is bound to N, S, O, S(O), S(O)2, C(O) or C(S) of —OR4, —SR7, —NR5R4, —C(O)R4, —C(S)R4, —C(O)OR4, —C(O)NR5R4, —C(S)NR5R4, —S(O)2NR5R4, —NR5C(O)R4, —NR5C(S)R4, —NR5S(O)2R4, —NR5C(O)NH2, —NR5C(O)NR5R4, —NR5C(S)NH2, —NR5C(S)NR5R4, —NR5S(O)2NH2, —NR5S(O)2NR5R4, —S(O)R4, or —S(O)2R4, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl;
R5 is selected from the group consisting of hydrogen and optionally substituted lower alkyl; and
R6 is optionally substituted heteroaryl, provided, however, the compound is not
Figure US20110092538A1-20110421-C00178
Figure US20110092538A1-20110421-C00179
Figure US20110092538A1-20110421-C00180
17. The method of claim 16, wherein the compound is approved for administration to a human.
18. The method of claim 17, wherein the disease or condition is selected from the group consisting of multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease, Parkinson's disease, melanoma, glioma, sarcoma, cancer of the colon, lung, breast, pancreas, thyroid, ovary, liver, or kidney, medullary thyroid cancer, carcinoid, small cell lung cancer, pheochromocytoma, lymphoma, neurofibromatosis, myelodysplastic syndrome, leukemia, tumor angiogenesis, neuropathic pain, inflammatory pain, acute pain, chronic pain, cancer-related pain, migraine, heart failure, ischemic stroke, cardiac hypertrophy, thrombosis, atherosclerosis, reperfusion injury, psoriasis, arthritis, osteoarthritis, endometriosis, scarring, vascular restenosis, fibrotic disorders, rheumatoid arthritis, inflammatory bowel disease, immunodeficiency diseases, organ transplant rejection, graft versus host disease, diabetic nephropathy, polycystic kidney disease, nephrosclerosis, glomerulonephritis, prostate hyperplasia, obesity, Helicobacter pylori infection, Hepatitis infection, Influenza virus infection, fever, sepsis, chronic obstructive pulmonary disease, acute respiratory distress syndrome, Noonan's syndrome, Costello syndrome, LEOPARD syndrome, cardio-faciocutaneous syndrome, neural crest syndrome abnormalities causing cardiovascular, skeletal, intestinal, skin, hair and endocrine diseases, sarcopenia, muscular dystrophy, motor neuron diseases, inflammatory myopathies, diseases of the neuromuscular junction, myopathies due to endocrine abnormalities, diseases of peripheral nerve, myotonia congenita, paramyotonia congenita, central core disease, nemaline myopathy, myotubular myopathy, periodic paralysis and metabolic diseases of muscle.
19. The method of claim 18, wherein
R1 is selected from the group consisting of hydrogen, halogen, lower alkyl, lower alkenyl, lower alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, —OH, —NH2, —CN, —NO2, —C(O)OH, —S(O)2NH2, —C(O)NH2, —C(S)NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —OR7, —SR7, —NR8R7, —C(O)R7, —C(S)R7, —C(O)OR7, —C(O)NR8R7, —C(S)NR8R7, —S(O)2NR8R7, —NR8C(O)R7, —NR8C(S)R7, —NR8S(O)2R7, —NR8C(O)NH2, —NR8C(O)NR8R7, —NR8C(S)NH2, —NR8C(S)NR8R7, —NR8S(O)2NH2, —NR8S(O)2NR8R7, —S(O)R7, and —S(O)2R7, wherein lower alkyl, lower alkenyl or lower alkynyl are optionally substituted with one or more substituents selected from fluoro, —OH, —NH2, C(O)OH, —C(O)NH2, —OR7, —NR8R7, —C(O)OR7, —C(O)NR8R7, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as R1 or as substituents of lower alkyl, lower alkenyl or lower alkynyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —S(O)2NH2, —C(O)NH2, —OR9, —SR9, —NHR9, —NR8R9, —NR8C(O)R9, —NR8S(O)2R9, —S(O)2R9, —S(O)2NR8R9, —C(O)R9, —C(O)NR8R9, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino;
R6 is heteroaryl optionally substituted with one or more substituents selected from the group consisting of halogen, lower alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, —OH, —NH2, —CN, —NO2, —C(O)OH, —S(O)2NH2, —C(O)NH2, —C(S)NH2, —NHC(O)NH2, —NHC(S)NH2, —NHS(O)2NH2, —OR7, —SR7, —NR8R7, —C(O)R7, —C(S)R7, —C(O)OR7, —C(O)NR8R7, —C(S)NR8R7, —S(O)2NR8R7, —NR8C(O)R7, —NR8C(S)R7, —NR8S(O)2R7, —NR8C(O)NH2, —NR8C(O)NR8R7, —NR8C(S)NH2, —NR8C(S)NR8R7, —NR8S(O)2NH2, —NR8S(O)2NR8R7, —S(O)R7, and —S(O)2R7, wherein lower alkyl is optionally substituted with one or more substituents selected from fluoro, —OR7, —NR8R7, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as a substituent of R6 or as a substituent of lower alkyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —S(O)2NH2, —C(O)NH2, —OR9, —SR9, —NHR9, —NR8R9, —NR8C(O)R9, —NR8S(O)2R9, —S(O)2R9, —S(O)2NR8R9, —C(O)R9, —C(O)NR8R9, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino;
R7 is selected from the group consisting of lower alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl, and heteroaryl as R7 or as substituents of lower alkyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —S(O)2NH2, —C(O)NH2, —OR9, —SR9, —NHR9, —NR8R9, —NR8C(O)R9, —NR8S(O)2R9, —S(O)2R9, —S(O)2NR8R9, —C(O)R9, —C(O)NR8R9, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino;
R8 at each occurrence is independently hydrogen or lower alkyl; and
R9 at each occurrence is independently selected from the group consisting of lower alkyl, heterocycloalkyl and heteroaryl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino, and cycloalkylamino, and heterocycloalkyl and heteroaryl are optionally substituted with one or more substituents selected from the group consisting of halogen, —CN, lower alkyl, fluoro substituted lower alkyl, lower alkoxy and fluoro substituted lower alkoxy.
20. The method of claim 18, wherein:
R1 is hydrogen, —CN, —NR8R7, —OR7, —S(O)2R7, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, cycloalkyl, heterocycloalkyl, aryl or heteroaryl, wherein cycloalkyl, heterocycloalkyl, aryl or heteroaryl are optionally substituted with one or more substituents selected from the group consisting of halogen, lower alkyl, fluoro substituted lower alkyl, —NR8R7, —OR7 and —S(O)2R7, and
R6 is heteroaryl optionally substituted with one or more of —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, —NR8R7, —OR7 or —S(O)2R7.
21. The method of claim 20, wherein:
R1 is hydrogen, —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower alkoxy substituted C2-6alkoxy, aryl or heteroaryl, wherein aryl or heteroaryl are optionally substituted with one or more substituents selected from the group consisting of fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, mono-alkylamino, di-alkylamino, and cycloalkylamino, and
R6 is heteroaryl optionally substituted with one or more of —CN, fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, mono-alkylamino, di-alkylamino, or cycloalkylamino.
22. The method of claim 18, wherein the compound is selected from the group consisting of:
Benzo[b]thiophene-3-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide,
5-Methyl-2-trifluoromethyl-furan-3-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide,
5-Oxazol-5-yl-thiophene-2-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide,
2-Oxo-2H-chromene-6-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide,
5-Isoxazol-5-yl-thiophene-2-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide,
Benzothiazole-6-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide,
1-Methyl-3-trifluoromethyl-1H-pyrazole-4-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide,
Benzo[1,2,5]thiadiazoe-5-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide,
5-Methyl-benzo[b]thiophene-2-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide,
5-Methyl-thiophene-2-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide,
1-Methyl-1H-pyrazole-3-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide,
Pyridine-2-sulfonic acid [2,4-difluoro-3-(5-methoxy-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-phenyl]-amide; and
pharmaceutically acceptable salts, tautomers, and isomers thereof.
23. A kit comprising a compound of Formula I according to claim 1, wherein the compound is approved for a medical indication selected from the group consisting of multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease, Parkinson's disease, melanoma, glioma, sarcoma, cancer of the colon, lung, breast, pancreas, thyroid, ovary, liver, or kidney, medullary thyroid cancer, carcinoid, small cell lung cancer, pheochromocytoma, lymphoma, neurofibromatosis, myelodysplastic syndrome, leukemia, tumor angiogenesis, neuropathic pain, inflammatory pain, acute pain, chronic pain, cancer-related pain, migraine, heart failure, ischemic stroke, cardiac hypertrophy, thrombosis, atherosclerosis, reperfusion injury, psoriasis, arthritis, osteoarthritis, endometriosis, scarring, vascular restenosis, fibrotic disorders, rheumatoid arthritis, inflammatory bowel disease, immunodeficiency diseases, organ transplant rejection, graft versus host disease, diabetic nephropathy, polycystic kidney disease, nephrosclerosis, glomerulonephritis, prostate hyperplasia, obesity, Helicobacter pylori infection, Hepatitis infection, Influenza virus infection, fever, sepsis, chronic obstructive pulmonary disease, acute respiratory distress syndrome, Noonan's syndrome, Costello syndrome, LEOPARD syndrome, cardio-faciocutaneous syndrome, neural crest syndrome abnormalities causing cardiovascular, skeletal, intestinal, skin, hair and endocrine diseases, sarcopenia, muscular dystrophy, motor neuron diseases, inflammatory myopathies, diseases of the neuromuscular junction, myopathies due to endocrine abnormalities, diseases of peripheral nerve, myotonia congenita, paramyotonia congenita, central core disease, nemaline myopathy, myotubular myopathy, periodic paralysis and metabolic diseases of muscle.
24. The kit of claim 23, wherein the compound is selected from the group consisting of:
4-Butoxy-N-[3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-benzenesulfonamide,
N-[3-(5-Chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-4-pyrazol-1-yl-benzenesulfonamide,
N-[3-(5-Chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-4-isopropoxy-benzenesulfonamide,
4-tert-Butyl-N-[3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-benzenesulfonamide,
N-[3-(5-Chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-4-propyl-benzenesulfonamide,
N-{2,4-Difluoro-3-[5-(2-methoxy-ethoxy)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-isopropyl-benzenesulfonamide,
N-{2,4-Difluoro-3-[5-(4-methyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-isopropyl-benzenesulfonamide,
4-Difluoromethoxy-N-{2,4-difluoro-3-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-benzenesulfonamide,
N-{2,4-Difluoro-3-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-propyl-benzenesulfonamide,
N-{2,4-Difluoro-3-[5-(1-methyl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-isopropyl-benzenesulfonamide,
N-{2,4-Difluoro-3-[5-(5-methyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-propyl-benzenesulfonamide,
N-{2,4-Difluoro-3-[5-(5-methyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-trifluoromethyl-benzenesulfonamide,
N-{2,4-Difluoro-3-[5-(1-methyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-4-propyl-benzenesulfonamide,
N-{3-[5-(1,5-Dimethyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-2,4-difluoro-phenyl}-4-propyl-benzenesulfonamide,
N-[2,4-Difluoro-3-(5-methyl-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-phenyl]-4-trifluoromethyl-benzene sulfonamide,
N-[3-(5-Cyano-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-4-trifluoromethyl-benzene sulfonamide,
(E)-3-{3-[2,6-Difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl}-acrylic acid methyl ester,
3-{3-[2,6-Difluoro-3-(4-trifluoromethyl-benzene sulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl}-prop ionic acid methyl ester,
3-{3-[2,6-Difluoro-3-(4-trifluoromethyl-benzene sulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl}-propionic acid,
3-{3-[2,6-Difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl}-N-ethyl-propionamide; and
pharmaceutically acceptable salts, tautomers, and isomers thereof.
25. A kit comprising a compound of Formula II according to claim 9, wherein the compound is approved for a medical indication selected from the group consisting of multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease, Parkinson's disease, melanoma, glioma, sarcoma, cancer of the colon, lung, breast, pancreas, thyroid, ovary, liver, or kidney, medullary thyroid cancer, carcinoid, small cell lung cancer, pheochromocytoma, lymphoma, neurofibromatosis, myelodysplastic syndrome, leukemia, tumor angiogenesis, neuropathic pain, inflammatory pain, acute pain, chronic pain, cancer-related pain, migraine, heart failure, ischemic stroke, cardiac hypertrophy, thrombosis, atherosclerosis, reperfusion injury, psoriasis, arthritis, osteoarthritis, endometriosis, scarring, vascular restenosis, fibrotic disorders, rheumatoid arthritis, inflammatory bowel disease, immunodeficiency diseases, organ transplant rejection, graft versus host disease, diabetic nephropathy, polycystic kidney disease, nephrosclerosis, glomerulonephritis, prostate hyperplasia, obesity, Helicobacter pylori infection, Hepatitis infection, Influenza virus infection, fever, sepsis, chronic obstructive pulmonary disease, acute respiratory distress syndrome, Noonan's syndrome, Costello syndrome, LEOPARD syndrome, cardio-faciocutaneous syndrome, neural crest syndrome abnormalities causing cardiovascular, skeletal, intestinal, skin, hair and endocrine diseases, sarcopenia, muscular dystrophy, motor neuron diseases, inflammatory myopathies, diseases of the neuromuscular junction, myopathies due to endocrine abnormalities, diseases of peripheral nerve, myotonia congenita, paramyotonia congenita, central core disease, nemaline myopathy, myotubular myopathy, periodic paralysis and metabolic diseases of muscle.
26. The kit of claim 25, wherein the compound is selected from the group consisting of:
3-[3-(5-Chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenylsulfamoyl]-benzoic acid,
N-[3-(5-Chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-3-difluoromethoxy-benzenesulfonamide,
3-Difluoromethoxy-N-{2,4-difluoro-3-[5-(5-methyl-1H-imidazol-2-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl}-benzenesulfonamide; and
pharmaceutically acceptable salts, tautomers, and isomers thereof.
27. A kit comprising a compound of Formula III according to claim 16, wherein the compound is approved for a medical indication selected from the group consisting of multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease, Parkinson's disease, melanoma, glioma, sarcoma, cancer of the colon, lung, breast, pancreas, thyroid, ovary, liver, or kidney, medullary thyroid cancer, carcinoid, small cell lung cancer, pheochromocytoma, lymphoma, neurofibromatosis, myelodysplastic syndrome, leukemia, tumor angiogenesis, neuropathic pain, inflammatory pain, acute pain, chronic pain, cancer-related pain, migraine, heart failure, ischemic stroke, cardiac hypertrophy, thrombosis, atherosclerosis, reperfusion injury, psoriasis, arthritis, osteoarthritis, endometriosis, scarring, vascular restenosis, fibrotic disorders, rheumatoid arthritis, inflammatory bowel disease, immunodeficiency diseases, organ transplant rejection, graft versus host disease, diabetic nephropathy, polycystic kidney disease, nephrosclerosis, glomerulonephritis, prostate hyperplasia, obesity, Helicobacter pylori infection, Hepatitis infection, Influenza virus infection, fever, sepsis, chronic obstructive pulmonary disease, acute respiratory distress syndrome, Noonan's syndrome, Costello syndrome, LEOPARD syndrome, cardio-faciocutaneous syndrome, neural crest syndrome abnormalities causing cardiovascular, skeletal, intestinal, skin, hair and endocrine diseases, sarcopenia, muscular dystrophy, motor neuron diseases, inflammatory myopathies, diseases of the neuromuscular junction, myopathies due to endocrine abnormalities, diseases of peripheral nerve, myotonia congenita, paramyotonia congenita, central core disease, nemaline myopathy, myotubular myopathy, periodic paralysis and metabolic diseases of muscle.
28. The kit of claim 27, wherein the compound is selected from the group consisting of:
Benzo[b]thiophene-3-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide,
5-Methyl-2-trifluoromethyl-furan-3-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide,
5-Oxazol-5-yl-thiophene-2-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide,
2-Oxo-2H-chromene-6-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide,
5-Isoxazol-5-yl-thiophene-2-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide,
Benzothiazole-6-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide,
1-Methyl-3-trifluoromethyl-1H-pyrazole-4-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide,
Benzo[1,2,5]thiadiazoe-5-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide,
5-Methyl-benzo[b]thiophene-2-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide,
5-Methyl-thiophene-2-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide,
1-Methyl-1H-pyrazole-3-sulfonic acid [3-(5-chloro-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide,
Pyridine-2-sulfonic acid [2,4-difluoro-3-(5-methoxy-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-phenyl]-amide; and
pharmaceutically acceptable salts, tautomers, and isomers thereof.
US12/981,427 2006-12-21 2010-12-29 Compounds and methods for kinase modulation, and indications therefor Abandoned US20110092538A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/981,427 US20110092538A1 (en) 2006-12-21 2010-12-29 Compounds and methods for kinase modulation, and indications therefor

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US87695306P 2006-12-21 2006-12-21
US11/960,590 US7863289B2 (en) 2006-12-21 2007-12-19 Compounds and methods for kinase modulation, and indications therefor
US12/981,427 US20110092538A1 (en) 2006-12-21 2010-12-29 Compounds and methods for kinase modulation, and indications therefor

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/960,590 Division US7863289B2 (en) 2006-12-21 2007-12-19 Compounds and methods for kinase modulation, and indications therefor

Publications (1)

Publication Number Publication Date
US20110092538A1 true US20110092538A1 (en) 2011-04-21

Family

ID=39281754

Family Applications (2)

Application Number Title Priority Date Filing Date
US11/960,590 Active 2028-12-09 US7863289B2 (en) 2006-12-21 2007-12-19 Compounds and methods for kinase modulation, and indications therefor
US12/981,427 Abandoned US20110092538A1 (en) 2006-12-21 2010-12-29 Compounds and methods for kinase modulation, and indications therefor

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US11/960,590 Active 2028-12-09 US7863289B2 (en) 2006-12-21 2007-12-19 Compounds and methods for kinase modulation, and indications therefor

Country Status (24)

Country Link
US (2) US7863289B2 (en)
EP (1) EP2097414A1 (en)
JP (1) JP2010514687A (en)
KR (1) KR20090095654A (en)
CN (1) CN101668757A (en)
AR (1) AR064476A1 (en)
AU (1) AU2007336906A1 (en)
BR (1) BRPI0722088A2 (en)
CA (1) CA2673000A1 (en)
CL (1) CL2007003797A1 (en)
CO (1) CO6190519A2 (en)
CR (1) CR10860A (en)
EC (1) ECSP099444A (en)
HN (1) HN2009001202A (en)
MA (1) MA31153B1 (en)
MX (1) MX2009006162A (en)
MY (1) MY148468A (en)
NO (1) NO20092251L (en)
PE (2) PE20081581A1 (en)
RU (1) RU2009122436A (en)
TW (1) TW200845975A (en)
UA (1) UA98629C2 (en)
UY (1) UY30816A1 (en)
WO (2) WO2008079903A1 (en)

Cited By (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140094611A1 (en) * 2012-05-31 2014-04-03 Plexxikon Inc. Synthesis of heterocyclic compounds
US9096593B2 (en) 2009-11-06 2015-08-04 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9169250B2 (en) 2006-11-22 2015-10-27 Plexxikon Inc. Compounds modulating c-fms and/or c-kit activity and uses therefor
US9260437B2 (en) 2008-05-19 2016-02-16 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9358235B2 (en) 2012-03-19 2016-06-07 Plexxikon Inc. Kinase modulation, and indications therefor
US9399639B2 (en) 2012-08-27 2016-07-26 Calitor Sciences, Llc Substituted azaindole compounds, salts, pharmaceutical compositions thereof and methods of use
US9440969B2 (en) 2009-12-23 2016-09-13 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9447089B2 (en) 2009-04-03 2016-09-20 Plexxikon Inc. Compositions and uses thereof
US9469640B2 (en) 2007-07-17 2016-10-18 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9550768B2 (en) 2011-05-17 2017-01-24 Plexxikon Inc. Kinase modulation and indications therefor
US9617267B2 (en) 2009-11-18 2017-04-11 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9624213B2 (en) 2011-02-07 2017-04-18 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9676748B2 (en) 2012-12-21 2017-06-13 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9682981B2 (en) 2010-04-21 2017-06-20 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9718847B2 (en) 2013-03-15 2017-08-01 Plexxikon Inc. Heterocyclic compounds and uses thereof
US9771363B2 (en) 2015-09-21 2017-09-26 Plexxikon Inc. Heterocyclic compounds and uses thereof
US9771369B2 (en) 2014-03-04 2017-09-26 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9822109B2 (en) 2013-03-15 2017-11-21 Plexxikon Inc. Heterocyclic compounds and uses thereof
US9856259B2 (en) 2014-09-15 2018-01-02 Plexxikon Inc. Heterocyclic compounds and uses thereof
US9873700B2 (en) 2013-05-30 2018-01-23 Plexxikon Inc. Compounds for kinase modulation, and indications therefor
US9938273B2 (en) 2015-12-07 2018-04-10 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10160747B2 (en) 2016-03-16 2018-12-25 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10160755B2 (en) 2015-04-08 2018-12-25 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10227357B2 (en) 2012-09-06 2019-03-12 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10428067B2 (en) 2017-06-07 2019-10-01 Plexxikon Inc. Compounds and methods for kinase modulation
US10435404B2 (en) 2017-07-25 2019-10-08 Plexxikon Inc. Formulations of a compound modulating kinases
US10508085B2 (en) 2016-09-22 2019-12-17 Plexxikon Inc. Compounds and methods for IDO and TDO modulation, and indications therefor
US10577366B2 (en) 2017-03-20 2020-03-03 Plexxikon Inc. Crystalline forms of a compound that inhibits bromodomain
US10703757B2 (en) 2016-12-23 2020-07-07 Plexxikon Inc. Compounds and methods for CDK8 modulation and indications therefor
US10717735B2 (en) 2017-10-13 2020-07-21 Plexxikon Inc. Solid forms of a compound for modulating kinases
US10829484B2 (en) 2015-07-28 2020-11-10 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
WO2021158936A1 (en) * 2020-02-05 2021-08-12 The Rockefeller University Pyrrolo [2,3-b]pyridine-3-carboxamide compositions and methods for ameliorating hearing loss
US11103505B2 (en) 2017-10-27 2021-08-31 Plexxikon Inc. Formulations of a compound modulating kinases
US11149011B2 (en) 2018-03-20 2021-10-19 Plexxikon Inc. Compounds and methods for IDO and TDO modulation, and indications therefor

Families Citing this family (138)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090118139A1 (en) * 2000-11-07 2009-05-07 Caliper Life Sciences, Inc. Microfluidic method and system for enzyme inhibition activity screening
CN1925855B (en) 2003-12-19 2010-06-16 普莱希科公司 Compounds and methods for development of Ret modulators
US7498342B2 (en) * 2004-06-17 2009-03-03 Plexxikon, Inc. Compounds modulating c-kit activity
JP2008545652A (en) * 2005-05-17 2008-12-18 プレキシコン,インコーポレーテッド Compounds that modulate c-kit and c-fms activity and uses thereof
NZ565255A (en) * 2005-06-22 2010-04-30 Plexxikon Inc Pyrrolo[2,3-b] pyridine derivatives as protein kinase inhibitors
WO2008080015A2 (en) 2006-12-21 2008-07-03 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
WO2008079909A1 (en) * 2006-12-21 2008-07-03 Plexxikon, Inc. Pyrrolo [2,3-b] pyridines as kinase modulators
PE20081581A1 (en) 2006-12-21 2008-11-12 Plexxikon Inc PIRROLO [2,3-b] PYRIDINES COMPOUNDS AS KINASE MODULATORS
WO2009099982A1 (en) * 2008-02-04 2009-08-13 Osi Pharmaceuticals, Inc. 2-aminopyridine kinase inhibitors
AR070317A1 (en) * 2008-02-06 2010-03-31 Osi Pharm Inc FURO (3,2-C) PIRIDINE AND HAVING (3,2-C) PIRIDINES
CN101952296B (en) 2008-02-25 2013-08-21 霍夫曼-拉罗奇有限公司 Pyrrolopyrazine kinase inhibitors
CA2712507A1 (en) 2008-02-25 2009-09-03 F. Hoffmann-La Roche Ag Pyrrolopyrazine kinase inhibitors
JP5485178B2 (en) * 2008-02-25 2014-05-07 エフ.ホフマン−ラ ロシュ アーゲー Pyrrolopyrazine kinase inhibitor
RU2503676C2 (en) * 2008-02-25 2014-01-10 Ф.Хоффманн-Ля Рош Аг Pyrrolopyrazine kinase inhibitors
EP2245033B1 (en) 2008-02-25 2011-08-31 F. Hoffmann-La Roche AG Pyrrolopyrazine kinase inhibitors
EP2265574A1 (en) * 2008-02-29 2010-12-29 Array Biopharma, Inc. N- (6-aminopyridin-3-yl) -3- (sulfonamido) benzamide derivatives as b-raf inhibitors for the treatment of cancer
CL2009000447A1 (en) * 2008-02-29 2010-01-04 Array Biopharma Inc Y Genentech Inc Compounds derived from (1h-pyrrolo {2,3-b} pyridin-5-yl) -sulfonamido-substituted benzamide; preparation procedure; pharmaceutical composition; and its use in the treatment of cancer, through the inhibition of raf.
KR20100117686A (en) * 2008-02-29 2010-11-03 어레이 바이오파마 인크. Pyrazole [3,4-b] pyridine raf inhibitors
CA2716947A1 (en) * 2008-02-29 2009-09-11 Array Biopharma Inc. Imidazo [4,5-b] pyridine derivatives used as raf inhibitors
WO2010019480A1 (en) * 2008-08-11 2010-02-18 The University Of Chicago Attenuated herpesviruses encoding a mek pathway polypeptide
RS53350B (en) 2008-09-22 2014-10-31 Array Biopharma, Inc. Substituted imidazo[1,2b] pyridazine compounds as trk kinase inhibitors
NZ593316A (en) 2008-11-10 2013-06-28 Vertex Pharma Pyrazine compounds useful as inhibitors of atr kinase
US8592448B2 (en) * 2008-11-20 2013-11-26 OSI Pharmaceuticals, LLC Substituted pyrrolo[2,3-b]-pyridines and -pyrazines
LT3354650T (en) 2008-12-19 2022-06-27 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
TW201036973A (en) * 2009-03-11 2010-10-16 Plexxikon Inc Compounds and uses thereof
WO2010104973A1 (en) * 2009-03-11 2010-09-16 Plexxikon, Inc. Pyrrolo [2, 3-b] pyridine derivatives for the inhibition of raf kinases
WO2010111527A1 (en) * 2009-03-26 2010-09-30 Plexxikon, Inc. Pyrazolo [ 3, 4 -b] pyridines as kinase inhibitors and their medical use
TW201041888A (en) * 2009-05-06 2010-12-01 Plexxikon Inc Compounds and methods for kinase modulation, and indications therefor
AR077468A1 (en) 2009-07-09 2011-08-31 Array Biopharma Inc PIRAZOLO COMPOUNDS (1,5-A) PYRIMIDINE SUBSTITUTED AS TRK-QUINASA INHIBITORS
US8329724B2 (en) 2009-08-03 2012-12-11 Hoffmann-La Roche Inc. Process for the manufacture of pharmaceutically active compounds
KR20120089459A (en) 2009-08-24 2012-08-10 에즈셉피온 파마슈티컬즈, 인코포레이티드 5,6-bicyclic heteroaryl-containing urea compounds as kinase inhibitors
JP2013503187A (en) 2009-08-28 2013-01-31 アレイ バイオファーマ、インコーポレイテッド Raf inhibitory compounds and methods of use thereof
EP2470532A1 (en) 2009-08-28 2012-07-04 Array Biopharma, Inc. 1h-pyrazolo [ 3, 4-b]pyridine compounds for inhibiting raf kinase
JP2013503189A (en) 2009-08-28 2013-01-31 アレイ バイオファーマ、インコーポレイテッド RAF inhibitory compounds and methods of use thereof
SG178853A1 (en) 2009-08-28 2012-04-27 Array Biopharma Inc Raf inhibitor compounds and methods of use thereof
CA2772074A1 (en) * 2009-08-28 2011-03-03 Array Biopharma Inc. Raf inhibitor compounds and methods of use thereof
CA2772575A1 (en) 2009-08-28 2011-03-03 Genentech, Inc. Raf inhibitor compounds and methods of use thereof
CN105153188B (en) * 2009-10-22 2018-06-01 法博太科制药有限公司 The fused ring analogs of antifibrotic agents
WO2011060216A1 (en) * 2009-11-12 2011-05-19 Concert Pharmaceuticals Inc. Substituted azaindoles
EP2538943B1 (en) * 2010-02-25 2016-03-30 Dana-Farber Cancer Institute, Inc. Braf mutations conferring resistance to braf inhibitors
US8518945B2 (en) 2010-03-22 2013-08-27 Hoffmann-La Roche Inc. Pyrrolopyrazine kinase inhibitors
US8481541B2 (en) 2010-03-22 2013-07-09 Hoffmann-La Roche Inc. Pyrrolopyrazine kinase inhibitors
WO2011143426A1 (en) 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
US9062008B2 (en) 2010-05-12 2015-06-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
JP2013526538A (en) 2010-05-12 2013-06-24 バーテックス ファーマシューティカルズ インコーポレイテッド Compounds useful as ATR kinase inhibitors
WO2011143425A2 (en) 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2011143419A1 (en) 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated Pyrazines useful as inhibitors of atr kinase
JP5856151B2 (en) 2010-05-12 2016-02-09 バーテックス ファーマシューティカルズ インコーポレイテッドVertex Pharmaceuticals Incorporated 2-Aminopyridine derivatives useful as ATR kinase inhibitors
AR081039A1 (en) 2010-05-14 2012-05-30 Osi Pharmaceuticals Llc QUINASA FUSIONED BICYCLE INHIBITORS
US20130072495A1 (en) 2010-05-14 2013-03-21 OSI Pharmaceuticals, LLC Fused bicyclic kinase inhibitors
TWI496785B (en) 2010-05-20 2015-08-21 Hoffmann La Roche Pyrrolopyrazine kinase inhibitors
WO2011163527A1 (en) 2010-06-23 2011-12-29 Vertex Pharmaceuticals Incorporated Pyrrolo- pyrazine derivatives useful as inhibitors of atr kinase
US8709419B2 (en) * 2010-08-17 2014-04-29 Hoffmann-La Roche, Inc. Combination therapy
EP2616467A1 (en) * 2010-09-13 2013-07-24 Concert Pharmaceuticals Inc. Substituted azaindoles
AR085279A1 (en) 2011-02-21 2013-09-18 Plexxikon Inc SOLID FORMS OF {3- [5- (4-CHLORINE-PHENYL) -1H-PIRROLO [2,3-B] PIRIDINA-3-CARBONIL] -2,4-DIFLUOR-PHENIL} -AMIDE OF PROPANE ACID-1- SULFONIC
WO2012118492A1 (en) * 2011-03-01 2012-09-07 Array Biopharma Inc. Heterocyclic sulfonamides as raf inhibitors
WO2012138938A1 (en) 2011-04-05 2012-10-11 Vertex Pharmaceuticals Incorporated Aminopyrazine compounds useful as inhibitors of tra kinase
WO2012158658A1 (en) 2011-05-16 2012-11-22 OSI Pharmaceuticals, LLC Fused bicyclic kinase inhibitors
JP2014522818A (en) 2011-06-22 2014-09-08 バーテックス ファーマシューティカルズ インコーポレイテッド Compounds useful as ATR kinase inhibitors
EP2723745A1 (en) 2011-06-22 2014-04-30 Vertex Pharmaceuticals Inc. Compounds useful as inhibitors of atr kinase
JP2014517079A (en) 2011-06-22 2014-07-17 バーテックス ファーマシューティカルズ インコーポレイテッド Compounds useful as ATR kinase inhibitors
EP2747767B8 (en) 2011-08-26 2019-07-17 Neupharma, Inc. Certain chemical entities, compositions, and methods
WO2013040515A1 (en) 2011-09-14 2013-03-21 Neupharma, Inc. Certain chemical entities, compositions, and methods
SG10201606774UA (en) 2011-09-30 2016-10-28 Vertex Pharma Processes for making compounds useful as inhibitors of atr kinase
MX2014003796A (en) 2011-09-30 2015-01-16 Vertex Pharma Compounds useful as inhibitors of atr kinase.
BR112014007690B1 (en) 2011-09-30 2022-10-04 Vertex Pharmaceuticals Incorporated Uses of ATR inhibitors in the treatment of pancreatic cancer and non-small cell lung cancer
EP2751088B1 (en) 2011-09-30 2016-04-13 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2013049720A1 (en) 2011-09-30 2013-04-04 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
GB2496135B (en) 2011-11-01 2015-03-18 Valirx Plc Inhibitors of the interaction between a Src family kinase and an androgen receptor or estradiol receptor for treatment of endometriosis
WO2013071093A1 (en) 2011-11-09 2013-05-16 Vertex Pharmaceuticals Incorporated Pyrazine compounds useful as inhibitors of atr kinase
EP2776422A1 (en) 2011-11-09 2014-09-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2013071088A1 (en) 2011-11-09 2013-05-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
US8846918B2 (en) 2011-11-09 2014-09-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
WO2013071085A1 (en) 2011-11-09 2013-05-16 Vertex Pharmaceuticals Incorporated Pyrazine compounds useful as inhibitors of atr kinase
DK2797888T3 (en) 2011-12-31 2016-09-19 Beigene Ltd Fused tricyclic compounds as RAF kinase inhibitors
EP2806874B1 (en) 2012-01-25 2017-11-15 Neupharma, Inc. Quinoxaline-oxy-phenyl derivatives as kinase inhibitors
MX353190B (en) 2012-01-30 2018-01-05 F Hoffmann La Roche Ag Star Isoquinoline and naphthyridine derivatives.
EP2833973B1 (en) 2012-04-05 2017-09-13 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase and combination therapies thereof
EP2855459B1 (en) 2012-05-31 2020-04-29 F.Hoffmann-La Roche Ag Aminoquinazoline and pyridopyrimidine derivatives
JO3407B1 (en) 2012-05-31 2019-10-20 Eisai R&D Man Co Ltd Tetrahydropyrazolopyrimidine Compounds
CN104812389B (en) 2012-09-24 2020-07-17 润新生物公司 Certain chemical entities, compositions, and methods
WO2014055756A1 (en) 2012-10-04 2014-04-10 Vertex Pharmaceuticals Incorporated Method for measuring atr inhibition mediated increases in dna damage
WO2014062604A1 (en) 2012-10-16 2014-04-24 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
US9725421B2 (en) 2012-11-12 2017-08-08 Neupharma, Inc. Substituted quinoxalines as B-raf kinase inhibitors
PL2941432T3 (en) 2012-12-07 2018-09-28 Vertex Pharmaceuticals Incorporated 2-amino-6-fluoro-n-(5-fluoro-4-(4-(4-(oxetan-3-yl)piperazine-1-carbonyl)piperidin-1-yl)pyridin-3-yl)pyrazolo[1,5alpha]pyrimidine-3-carboxamide as inhibitor of atr kinase
EP2970286A1 (en) 2013-03-15 2016-01-20 Vertex Pharmaceuticals Inc. Fused pyrazolopyrimidine derivatives useful as inhibitors of atr kinase
US9206188B2 (en) * 2013-04-18 2015-12-08 Arrien Pharmaceuticals Llc Substituted pyrrolo[2,3-b]pyridines as ITK and JAK inhibitors
GB2513615A (en) 2013-05-01 2014-11-05 Cancer Rec Tech Ltd Medical use
EP3013797B1 (en) 2013-06-28 2018-01-03 BeiGene, Ltd. Fused tricyclic amide compounds as multiple kinase inhibitors
JP6380861B2 (en) 2013-06-28 2018-08-29 ベイジーン リミテッド Condensed tricyclic urea compounds as Raf kinase and / or dimer inhibitors of Raf kinase
WO2015075749A1 (en) * 2013-11-22 2015-05-28 Laurus Labs Private Limited Novel processes for the preparation of vemurafenib
WO2015085132A1 (en) 2013-12-06 2015-06-11 Vertex Pharmaceuticals Incorporated 2-amino-6-fluoro-n-[5-fluoro-pyridin-3-yl]pyrazolo[1,5-a]pyrimidin-3-carboxamide compound useful as atr kinase inhibitor, its preparation, different solid forms and radiolabelled derivatives thereof
EA028614B1 (en) * 2014-05-22 2017-12-29 Общество С Ограниченной Ответственностью "Русские Фармацевтические Технологии" Selective inhibitors interfering with fibroblast growth factor receptor and frs2 interaction for the prevention and treatment of cancer
US9670215B2 (en) 2014-06-05 2017-06-06 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
WO2015195740A1 (en) 2014-06-17 2015-12-23 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of chk1 and atr inhibitors
JP6820854B2 (en) * 2015-01-18 2021-01-27 エスアールアイ インターナショナルSRI International MAP4K4 (HGK) inhibitor
PL3322706T3 (en) 2015-07-16 2021-07-19 Array Biopharma, Inc. Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
US11464774B2 (en) 2015-09-30 2022-10-11 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of DNA damaging agents and ATR inhibitors
MX2018005087A (en) 2015-10-26 2019-05-16 Loxo Oncology Inc Point mutations in trk inhibitor-resistant cancer and methods relating to the same.
UA125025C2 (en) 2016-04-04 2021-12-29 Локсо Онколоджі, Інк. Methods of treating pediatric cancers
KR102400423B1 (en) 2016-04-04 2022-05-19 록쏘 온콜로지, 인코포레이티드 (S)-N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-A]pyrimidine-3- Liquid formulation of yl)-3-hydroxypyrrolidine-1-carboxamide
US10045991B2 (en) 2016-04-04 2018-08-14 Loxo Oncology, Inc. Methods of treating pediatric cancers
MX2018012471A (en) 2016-04-15 2019-02-21 Genentech Inc Diagnostic and therapeutic methods for cancer.
PT3800189T (en) 2016-05-18 2023-07-27 Array Biopharma Inc Preparation of (s)-n-(5-((r)-2-(2,5-difluorophenyl)pyrrolidin-1-yl)pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide
TWI704148B (en) 2016-10-10 2020-09-11 美商亞雷生物製藥股份有限公司 Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
JOP20190077A1 (en) 2016-10-10 2019-04-09 Array Biopharma Inc Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
JOP20190092A1 (en) 2016-10-26 2019-04-25 Array Biopharma Inc PROCESS FOR THE PREPARATION OF PYRAZOLO[1,5-a]PYRIMIDINES AND SALTS THEREOF
EP3571203B1 (en) 2017-01-18 2023-06-07 Array BioPharma Inc. Substituted pyrazolo[1,5-a]pyrazine compounds as ret kinase inhibitors
WO2018136663A1 (en) 2017-01-18 2018-07-26 Array Biopharma, Inc. Ret inhibitors
JP7185631B2 (en) 2017-02-03 2022-12-07 サータ セラピューティクス プロプライエタリー リミテッド antifibrotic compound
JOP20190213A1 (en) 2017-03-16 2019-09-16 Array Biopharma Inc Macrocyclic compounds as ros1 kinase inhibitors
JP2020532982A (en) 2017-09-08 2020-11-19 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト How to diagnose and treat cancer
CN111247152A (en) 2017-09-20 2020-06-05 璧辰医药技术股份有限公司 Cyclic iminopyrimidine derivatives as kinase inhibitors
TWI791053B (en) 2017-10-10 2023-02-01 美商亞雷生物製藥股份有限公司 Crystalline forms of 6-(2-hydroxy-2-methylpropoxy)-4-(6-(6-((6-methoxypyridin-3-yl)methyl)-3,6-diazabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)pyrazolo[1,5-a]pyridine-3-carbonitrile and pharmaceutical composition thereof
TWI812649B (en) 2017-10-10 2023-08-21 美商絡速藥業公司 Formulations of 6-(2-hydroxy-2-methylpropoxy)-4-(6-(6-((6-methoxypyridin-3-yl)methyl)-3,6-diazabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)pyrazolo[1,5-a]pyridine-3-carbonitrile
TWI823890B (en) 2018-01-10 2023-12-01 美商克拉治療有限責任公司 Pharmaceutical compositions comprising phenylsulfonamides, and their therapeutic applications
WO2019143994A1 (en) 2018-01-18 2019-07-25 Array Biopharma Inc. Substituted pyrazolyl[4,3-c]pyridinecompounds as ret kinase inhibitors
US11603374B2 (en) 2018-01-18 2023-03-14 Array Biopharma Inc. Substituted pyrrolo[2,3-d]pyrimidines compounds as ret kinase inhibitors
CN111630054B (en) 2018-01-18 2023-05-09 奥瑞生物药品公司 Substituted pyrazolo [3,4-d ] pyrimidine compounds as RET kinase inhibitors
BR112020015072A2 (en) * 2018-01-31 2021-01-05 Heparegenix Gmbh PROTEIN KINASE MKK4 INHIBITORS TO PROMOTE LIVER REGENERATION, OR REDUCE, OR PREVENT HEPATOCYT DEATH
EP3752200A1 (en) 2018-02-13 2020-12-23 Vib Vzw Targeting minimal residual disease in cancer with rxr antagonists
US11731968B2 (en) 2018-06-21 2023-08-22 Heparegenix Gmbh Tricyclic protein kinase inhibitors for promoting liver regeneration or reducing or preventing hepatocyte death
JP7365393B2 (en) * 2018-07-16 2023-10-19 ヘパリジェニックス ゲーエムベーハー Protein kinase inhibitors for promoting liver regeneration or suppressing or preventing hepatocyte death
CA3108065A1 (en) 2018-07-31 2020-02-06 Loxo Oncology, Inc. Spray-dried dispersions, formulations, and polymorphs of (s)-5-amino-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1-(1,1,1-trifluoropropan-2-yl)-1h-pyrazole-4-carboxamide
CN112996794A (en) 2018-09-10 2021-06-18 阿雷生物药品公司 Fused heterocyclic compounds as RET kinase inhibitors
WO2020131627A1 (en) 2018-12-19 2020-06-25 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyridine compounds as inhibitors of fgfr tyrosine kinases
JP2022515197A (en) 2018-12-19 2022-02-17 アレイ バイオファーマ インコーポレイテッド 7-((3,5-dimethoxyphenyl) amino) quinoxaline derivative as an FGFR inhibitor for treating cancer
BR112022001488A2 (en) * 2019-07-29 2022-04-19 Heparegenix Gmbh Heteroaryl-substituted pyrazolo-pyridine protein kinase inhibitors to promote liver regeneration or reduction or prevention of hepatocyte death
IL294214A (en) 2019-12-27 2022-08-01 Schr?Dinger Inc Cyclic compounds and methods of using same
CA3161513A1 (en) 2020-01-28 2021-08-05 Irwin DAVIDSON Antisense oligonucleotide targeting linc00518 for treating melanoma
CN111662288B (en) * 2020-06-23 2022-06-14 徐州医科大学 Small molecule compound for inhibiting AKT and STAT3 activity and application thereof
WO2022055963A1 (en) 2020-09-10 2022-03-17 Schrödinger, Inc. Heterocyclic pericondensed cdc7 kinase inhibitors for the treatment of cancer
WO2022164789A1 (en) 2021-01-26 2022-08-04 Schrödinger, Inc. Tricyclic compounds useful in the treatment of cancer, autoimmune and inflammatory disoders
JP2024506602A (en) * 2021-02-04 2024-02-14 プレジデント アンド フェローズ オブ ハーバード カレッジ Selective ROCK2 inhibition for the treatment of edema and related conditions
TW202300150A (en) 2021-03-18 2023-01-01 美商薛定諤公司 Cyclic compounds and methods of using same
WO2024003350A1 (en) 2022-06-30 2024-01-04 Universite De Strasbourg Combination therapy for melanoma
CN115160315A (en) * 2022-07-28 2022-10-11 乐威医药(江苏)股份有限公司 7-azaindole derivatives and preparation method thereof
WO2024033381A1 (en) 2022-08-10 2024-02-15 Vib Vzw Inhibition of tcf4/itf2 in the treatment of cancer

Citations (90)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2234705A (en) * 1940-04-12 1941-03-11 Eastman Kodak Co Cellulose organic derivative composition containing esters of monoalkoxy benzoic acids
US2413258A (en) * 1942-07-07 1946-12-24 United Gas Improvement Co Polystyrene-type resins plasticized with high boiling fatty acid alkyl esters
US4150949A (en) * 1976-04-15 1979-04-24 Technicon Instruments Corporation Immunoassay for gentamicin
US4568649A (en) * 1983-02-22 1986-02-04 Immunex Corporation Immediate ligand detection assay
US4626513A (en) * 1983-11-10 1986-12-02 Massachusetts General Hospital Method and apparatus for ligand detection
US4634701A (en) * 1984-06-06 1987-01-06 Ausonia Farmaceutici S.R.L. Furan derivatives having anti-ulcer activity
US4727395A (en) * 1983-01-20 1988-02-23 Tokyo Shibaura Denki Kabushiki Kaisha Reverse developing image forming apparatus with small drum
US5120782A (en) * 1990-07-13 1992-06-09 Bayer Aktiengesellschaft Substituted pyrrolo-pyridines pharmaceuticals
US5143854A (en) * 1989-06-07 1992-09-01 Affymax Technologies N.V. Large scale photolithographic solid phase synthesis of polypeptides and receptor binding screening thereof
US5338849A (en) * 1992-02-14 1994-08-16 Lipha, Lyonnaise Industrielle Pharmaceutique Azaindoles, preparation processes and medicinal products containing them
US5426039A (en) * 1993-09-08 1995-06-20 Bio-Rad Laboratories, Inc. Direct molecular cloning of primer extended DNA containing an alkane diol
US5432177A (en) * 1993-03-01 1995-07-11 Merck Sharpe & Dohme Ltd. Pyrrolo-pyridine derivatives
US5434049A (en) * 1992-02-28 1995-07-18 Hitachi, Ltd. Separation of polynucleotides using supports having a plurality of electrode-containing cells
US5449614A (en) * 1988-09-01 1995-09-12 Whitehead Institue For Biomedical Research Recombinant retroviruses with amphotropic and ecotropic host ranges
US5486525A (en) * 1993-12-16 1996-01-23 Abbott Laboratories Platelet activating factor antagonists: imidazopyridine indoles
US5556752A (en) * 1994-10-24 1996-09-17 Affymetrix, Inc. Surface-bound, unimolecular, double-stranded DNA
US5576319A (en) * 1993-03-01 1996-11-19 Merck, Sharp & Dohme Ltd. Pyrrolo-pyridine derivatives
US5631236A (en) * 1993-08-26 1997-05-20 Baylor College Of Medicine Gene therapy for solid tumors, using a DNA sequence encoding HSV-Tk or VZV-Tk
US5632957A (en) * 1993-11-01 1997-05-27 Nanogen Molecular biological diagnostic systems including electrodes
US5658775A (en) * 1988-05-17 1997-08-19 Sloan-Kettering Institute For Cancer Research Double copy retroviral vector
US5681959A (en) * 1995-02-21 1997-10-28 Merck, Sharp & Dohme Ltd. Chemical synthesis of azaindoles
US5712285A (en) * 1995-04-07 1998-01-27 Merck, Sharp & Dohme, Ltd. Pyrrolo-pyridine derivatives
US5721118A (en) * 1995-10-31 1998-02-24 The Regents Of The University Of California, San Diego Mammalian artificial chromosomes and methods of using same
US5744305A (en) * 1989-06-07 1998-04-28 Affymetrix, Inc. Arrays of materials attached to a substrate
US5747276A (en) * 1995-09-15 1998-05-05 The Scripps Research Institute Screening methods for the identification of novel antibiotics
US5763198A (en) * 1994-07-22 1998-06-09 Sugen, Inc. Screening assays for compounds
US5770456A (en) * 1989-06-07 1998-06-23 Affymetrix, Inc. Cyclic nucleic acid and polypeptide arrays
US5800992A (en) * 1989-06-07 1998-09-01 Fodor; Stephen P.A. Method of detecting nucleic acids
US5807522A (en) * 1994-06-17 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods for fabricating microarrays of biological samples
US5830645A (en) * 1994-12-09 1998-11-03 The Regents Of The University Of California Comparative fluorescence hybridization to nucleic acid arrays
US5840485A (en) * 1993-05-27 1998-11-24 Selectide Corporation Topologically segregated, encoded solid phase libraries
US5856174A (en) * 1995-06-29 1999-01-05 Affymetrix, Inc. Integrated nucleic acid diagnostic device
US5877007A (en) * 1988-02-10 1999-03-02 Ict Pharmaceuticals, Inc. Method of screening for protein inhibitors and activators
US5959098A (en) * 1996-04-17 1999-09-28 Affymetrix, Inc. Substrate preparation process
US5965452A (en) * 1996-07-09 1999-10-12 Nanogen, Inc. Multiplexed active biologic array
US6013440A (en) * 1996-03-11 2000-01-11 Affymetrix, Inc. Nucleic acid affinity columns
US6022963A (en) * 1995-12-15 2000-02-08 Affymetrix, Inc. Synthesis of oligonucleotide arrays using photocleavable protecting groups
US6025155A (en) * 1996-04-10 2000-02-15 Chromos Molecular Systems, Inc. Artificial chromosomes, uses thereof and methods for preparing artificial chromosomes
US6045996A (en) * 1993-10-26 2000-04-04 Affymetrix, Inc. Hybridization assays on oligonucleotide arrays
US6048695A (en) * 1998-05-04 2000-04-11 Baylor College Of Medicine Chemically modified nucleic acids and methods for coupling nucleic acids to solid support
US6054270A (en) * 1988-05-03 2000-04-25 Oxford Gene Technology Limited Analying polynucleotide sequences
US6090912A (en) * 1993-05-27 2000-07-18 Selectide Corporation Topologically segregated, encoded solid phase libraries comprising linkers having an enzymatically susceptible bond
US6096718A (en) * 1997-06-05 2000-08-01 Gene Targeting Corp. Tissue specific adenovirus vectors for breast cancer treatment
US6107478A (en) * 1995-09-08 2000-08-22 Pedersen; Finn Skou Retroviral vector, a replication system for said vector and avian or mammalian cells transfected with said vector
US6110458A (en) * 1991-07-26 2000-08-29 University Of Rochester Cancer therapy utilizing ex vivo gene transduction
US6110456A (en) * 1995-06-07 2000-08-29 Yale University Oral delivery or adeno-associated viral vectors
US6113913A (en) * 1998-06-26 2000-09-05 Genvec, Inc. Recombinant adenovirus
US6117681A (en) * 1995-03-29 2000-09-12 Bavarian Nordic Research Inst. A/S Pseudotyped retroviral particles
US6178384B1 (en) * 1997-09-29 2001-01-23 The Trustees Of Columbia University In The City Of New York Method and apparatus for selecting a molecule based on conformational free energy
US6235769B1 (en) * 1997-07-03 2001-05-22 Sugen, Inc. Methods of preventing and treating neurological disorders with compounds that modulate the function of the C-RET receptor protein tyrosine kinase
US6243980B1 (en) * 1997-03-07 2001-06-12 Tropix, Inc. Protease inhibitor assay
US20010008765A1 (en) * 1999-12-06 2001-07-19 Fuji Photo Film Co., Ltd. DNA chip and reactive solid carrier
US20010012537A1 (en) * 1999-07-30 2001-08-09 Anderson Norman G. Dry deposition of materials for microarrays using matrix displacement
US20010014448A1 (en) * 1997-09-30 2001-08-16 Surmodics, Inc. Target molecule attachment to surfaces
US20010014449A1 (en) * 1993-11-01 2001-08-16 Michael I. Nerenberg Methods for determination of single nucleic acid polymorphisms using bioelectronic microchip
US6277489B1 (en) * 1998-12-04 2001-08-21 The Regents Of The University Of California Support for high performance affinity chromatography and other uses
US6277628B1 (en) * 1998-10-02 2001-08-21 Incyte Genomics, Inc. Linear microarrays
US20010016322A1 (en) * 1999-04-27 2001-08-23 Caren Michael P. Method of performing array-based hybridization assays using thermal inkjet deposition of sample fluids
US20010018642A1 (en) * 1997-07-25 2001-08-30 David Balaban Method and system for providing a probe array chip design database
US20010019827A1 (en) * 1997-09-11 2001-09-06 Dawson Elliott P. Method of making high density arrays
US6288234B1 (en) * 1998-06-08 2001-09-11 Advanced Medicine, Inc. Multibinding inhibitors of microsomal triglyceride transferase protein
US6294330B1 (en) * 1997-01-31 2001-09-25 Odyssey Pharmaceuticals Inc. Protein fragment complementation assays for the detection of biological or drug interactions
US6310074B1 (en) * 1999-05-19 2001-10-30 Adir Et Compagnie Substituted dimeric compounds
US6350786B1 (en) * 1998-09-22 2002-02-26 Hoffmann-La Roche Inc. Stable complexes of poorly soluble compounds in ionic polymers
US20030003004A1 (en) * 2001-06-13 2003-01-02 Stones Ian David Lubricating systems for regenerative vacuum pumps
US6545014B2 (en) * 1996-04-15 2003-04-08 Texas Biotechnology Corporation Method of treating glaucoma
US20040002534A1 (en) * 1999-12-22 2004-01-01 Sugen, Inc. Methods of modulating c-kit tyrosine protein kinase function with indolinone compounds
US20040077595A1 (en) * 2002-09-06 2004-04-22 Insert Therapeutics, Inc. Cyclodextrin-based polymers for therapeutics delivery
US20040142864A1 (en) * 2002-09-16 2004-07-22 Plexxikon, Inc. Crystal structure of PIM-1 kinase
US20040167030A1 (en) * 2003-02-14 2004-08-26 Wyeth Heterocyclyl-3-sulfonylazaindole or-azaindazole derivatives as 5-hydroxytryptamine-6 ligands
US6858860B2 (en) * 2001-07-24 2005-02-22 Seiko Epson Corporation Apparatus and method for measuring natural period of liquid
US20050085463A1 (en) * 2003-01-23 2005-04-21 Weiner David M. Use of N-desmethylclozapine to treat human neuropsychiatric disease
US6897207B2 (en) * 2001-06-21 2005-05-24 Aventis Pharmaceuticals Inc. Azaindoles
US20050154014A1 (en) * 2003-01-06 2005-07-14 Jason Bloxham (2-carboxamido)(3-amino) thiophene compounds
US20050164300A1 (en) * 2003-09-15 2005-07-28 Plexxikon, Inc. Molecular scaffolds for kinase ligand development
US20050170431A1 (en) * 2003-02-28 2005-08-04 Plexxikon, Inc. PYK2 crystal structure and uses
US20050256151A1 (en) * 2003-12-24 2005-11-17 Pharmacia Italia S.P.A. Pyrrolo[2,3-b]pyridine derivatives active as kinase inhibitors, process for their preparation and pharmaceutical compositions comprising them
US20060035898A1 (en) * 2004-07-27 2006-02-16 Structural Genomix, Inc. Fused ring heterocycle kinase modulators
US20060058340A1 (en) * 2004-06-17 2006-03-16 Ibrahim Prabha N Compounds modulating c-kit activity
US20060058339A1 (en) * 2004-06-17 2006-03-16 Ibrahim Prabha N Compounds modulating c-kit activity and uses therefor
US20070032519A1 (en) * 2005-05-17 2007-02-08 Chao Zhang Compounds modulating c-kit and c-fms activity and uses therefor
US20070049615A1 (en) * 2003-12-19 2007-03-01 Plexxikon, Inc. Compounds and methods for development of Ret modulators
US7361764B2 (en) * 2004-07-27 2008-04-22 Sgx Pharmaceuticals, Inc. Pyrrolo-pyridine kinase modulators
US7361763B2 (en) * 2004-07-27 2008-04-22 Sgx Pharmaceuticals, Inc. Pyrrolo-pyridine kinase modulators
US20080167338A1 (en) * 2006-12-21 2008-07-10 Wayne Spevak Compounds and methods for kinase modulation, and indications therefor
US20080188514A1 (en) * 2006-12-21 2008-08-07 Guoxian Wu Compounds and methods for kinase modulation, and indications therefor
US20090005356A1 (en) * 2007-05-29 2009-01-01 Sgx Pharmaceuticals, Inc. Substituted pyrrolopyridines and pyrazolopyridines as kinase modulators
US20090076046A1 (en) * 2006-11-22 2009-03-19 Plexxikon Inc Compounds modulating c-fms and/or c-kit activity and uses therefor
US20090143352A1 (en) * 2004-07-27 2009-06-04 Sgx Pharmaceuticals, Inc. Pyrrolo-pyridine kinase modulatiors
US7863288B2 (en) * 2005-06-22 2011-01-04 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor

Family Cites Families (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2413258A1 (en) 1974-03-20 1975-10-02 Bayer Ag Herbicidal N-(alkoxycarbonyl-phenyl)-N'-methyl-urea derivs - prepd by reacting alkoxycarbonyl-phenyl isocyanates with methylamines
EP0154734B1 (en) 1984-03-15 1990-08-29 Immunex Corporation Immediate ligand detection assay, a test kit and its formation
US5700637A (en) 1988-05-03 1997-12-23 Isis Innovation Limited Apparatus and method for analyzing polynucleotide sequences and method of generating oligonucleotide arrays
US5703055A (en) 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
AU7906691A (en) 1990-05-23 1991-12-10 United States of America, as represented by the Secretary, U.S. Department of Commerce, The Adeno-associated virus (aav)-based eucaryotic vectors
ES2134929T3 (en) 1993-03-01 1999-10-16 Merck Sharp & Dohme PIRROLO-PIRIDINA DERIVATIVES AS LINKS FOR DOPAMINE RECEPTORS.
GB9416189D0 (en) 1994-08-10 1994-09-28 Merck Sharp & Dohme Therapeutic agents
GB9416162D0 (en) 1994-08-10 1994-09-28 Merck Sharp & Dohme Therapeutic agents
GB9503400D0 (en) 1995-02-21 1995-04-12 Merck Sharp & Dohme Therpeutic agents
GB2299581A (en) 1995-04-07 1996-10-09 Merck Sharp & Dohme 3-(Tetrahydropyridin-1-yl-methyl)pyrrolo[2,3-b]pyridine derivatives as ligands for dopamine receptor subtypes
US5977131A (en) 1997-04-09 1999-11-02 Pfizer Inc. Azaindole-ethylamine derivatives as nicotinic acetylcholine receptor binding agents
US6161776A (en) 1997-08-12 2000-12-19 Nibco Inc. Multi-layered, porous mat turf irrigation apparatus and method
EP1267111A1 (en) 2001-06-15 2002-12-18 Dsm N.V. Pressurized fluid conduit
TW200403243A (en) 2002-07-18 2004-03-01 Wyeth Corp 1-Heterocyclylalkyl-3-sulfonylazaindole or-azaindazole derivatives as 5-hydroxytryptamine-6 ligands
TWI329112B (en) 2002-07-19 2010-08-21 Bristol Myers Squibb Co Novel inhibitors of kinases
US6878887B2 (en) 2002-08-07 2005-04-12 Matsushita Electric Industrial Co., Ltd. Anti-malfunction mechanism for variable output device
SE0202463D0 (en) 2002-08-14 2002-08-14 Astrazeneca Ab Novel compounds
SE0300119D0 (en) 2003-01-17 2003-01-17 Astrazeneca Ab Novel compounds
SE0300120D0 (en) 2003-01-17 2003-01-17 Astrazeneca Ab Novel compounds
DE10357510A1 (en) 2003-12-09 2005-07-07 Bayer Healthcare Ag Heteroaryl-substituted benzenes
GB0330042D0 (en) 2003-12-24 2004-01-28 Pharmacia Italia Spa Pyrrolo [2,3-b] pyridine derivatives active as kinase inhibitors process for their preparation and pharmaceutical compositions them
GB0403635D0 (en) 2004-02-18 2004-03-24 Devgen Nv Pyridinocarboxamides with improved activity as kinase inhibitors
GB0405055D0 (en) 2004-03-05 2004-04-07 Eisai London Res Lab Ltd JNK inhibitors
RS51830B (en) 2004-03-30 2012-02-29 Vertex Pharmaceuticals Incorporated Azaindoles useful as inhibitors of jak and other protein kinases
WO2006004984A1 (en) 2004-06-30 2006-01-12 Vertex Pharmaceuticals Incorporated Azaindoles useful as inhibitors of protein kinases
US7626021B2 (en) 2004-07-27 2009-12-01 Sgx Pharmaceuticals, Inc. Fused ring heterocycle kinase modulators
WO2006114180A1 (en) 2005-04-25 2006-11-02 Merck Patent Gmbh Novel aza- heterocycles serving as kinase inhibitors
CA2609126A1 (en) 2005-05-20 2006-11-30 Vertex Pharmaceuticals Incorporated Pyrrolopyridines useful as inhibitors of protein kinase
GB0516156D0 (en) 2005-08-05 2005-09-14 Eisai London Res Lab Ltd JNK inhibitors

Patent Citations (99)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2234705A (en) * 1940-04-12 1941-03-11 Eastman Kodak Co Cellulose organic derivative composition containing esters of monoalkoxy benzoic acids
US2413258A (en) * 1942-07-07 1946-12-24 United Gas Improvement Co Polystyrene-type resins plasticized with high boiling fatty acid alkyl esters
US4150949A (en) * 1976-04-15 1979-04-24 Technicon Instruments Corporation Immunoassay for gentamicin
US4727395A (en) * 1983-01-20 1988-02-23 Tokyo Shibaura Denki Kabushiki Kaisha Reverse developing image forming apparatus with small drum
US4568649A (en) * 1983-02-22 1986-02-04 Immunex Corporation Immediate ligand detection assay
US4626513A (en) * 1983-11-10 1986-12-02 Massachusetts General Hospital Method and apparatus for ligand detection
US4634701A (en) * 1984-06-06 1987-01-06 Ausonia Farmaceutici S.R.L. Furan derivatives having anti-ulcer activity
US5877007A (en) * 1988-02-10 1999-03-02 Ict Pharmaceuticals, Inc. Method of screening for protein inhibitors and activators
US6054270A (en) * 1988-05-03 2000-04-25 Oxford Gene Technology Limited Analying polynucleotide sequences
US5658775A (en) * 1988-05-17 1997-08-19 Sloan-Kettering Institute For Cancer Research Double copy retroviral vector
US5449614A (en) * 1988-09-01 1995-09-12 Whitehead Institue For Biomedical Research Recombinant retroviruses with amphotropic and ecotropic host ranges
US5143854A (en) * 1989-06-07 1992-09-01 Affymax Technologies N.V. Large scale photolithographic solid phase synthesis of polypeptides and receptor binding screening thereof
US6261776B1 (en) * 1989-06-07 2001-07-17 Affymetrix, Inc. Nucleic acid arrays
US5800992A (en) * 1989-06-07 1998-09-01 Fodor; Stephen P.A. Method of detecting nucleic acids
US5770456A (en) * 1989-06-07 1998-06-23 Affymetrix, Inc. Cyclic nucleic acid and polypeptide arrays
US5744305A (en) * 1989-06-07 1998-04-28 Affymetrix, Inc. Arrays of materials attached to a substrate
US5120782A (en) * 1990-07-13 1992-06-09 Bayer Aktiengesellschaft Substituted pyrrolo-pyridines pharmaceuticals
US6110458A (en) * 1991-07-26 2000-08-29 University Of Rochester Cancer therapy utilizing ex vivo gene transduction
US5338849A (en) * 1992-02-14 1994-08-16 Lipha, Lyonnaise Industrielle Pharmaceutique Azaindoles, preparation processes and medicinal products containing them
US5434049A (en) * 1992-02-28 1995-07-18 Hitachi, Ltd. Separation of polynucleotides using supports having a plurality of electrode-containing cells
US5432177A (en) * 1993-03-01 1995-07-11 Merck Sharpe & Dohme Ltd. Pyrrolo-pyridine derivatives
US5576319A (en) * 1993-03-01 1996-11-19 Merck, Sharp & Dohme Ltd. Pyrrolo-pyridine derivatives
US5840485A (en) * 1993-05-27 1998-11-24 Selectide Corporation Topologically segregated, encoded solid phase libraries
US6090912A (en) * 1993-05-27 2000-07-18 Selectide Corporation Topologically segregated, encoded solid phase libraries comprising linkers having an enzymatically susceptible bond
US5631236A (en) * 1993-08-26 1997-05-20 Baylor College Of Medicine Gene therapy for solid tumors, using a DNA sequence encoding HSV-Tk or VZV-Tk
US5426039A (en) * 1993-09-08 1995-06-20 Bio-Rad Laboratories, Inc. Direct molecular cloning of primer extended DNA containing an alkane diol
US6045996A (en) * 1993-10-26 2000-04-04 Affymetrix, Inc. Hybridization assays on oligonucleotide arrays
US20010014449A1 (en) * 1993-11-01 2001-08-16 Michael I. Nerenberg Methods for determination of single nucleic acid polymorphisms using bioelectronic microchip
US5632957A (en) * 1993-11-01 1997-05-27 Nanogen Molecular biological diagnostic systems including electrodes
US5486525A (en) * 1993-12-16 1996-01-23 Abbott Laboratories Platelet activating factor antagonists: imidazopyridine indoles
US5807522A (en) * 1994-06-17 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods for fabricating microarrays of biological samples
US5763198A (en) * 1994-07-22 1998-06-09 Sugen, Inc. Screening assays for compounds
US5556752A (en) * 1994-10-24 1996-09-17 Affymetrix, Inc. Surface-bound, unimolecular, double-stranded DNA
US5830645A (en) * 1994-12-09 1998-11-03 The Regents Of The University Of California Comparative fluorescence hybridization to nucleic acid arrays
US5681959A (en) * 1995-02-21 1997-10-28 Merck, Sharp & Dohme Ltd. Chemical synthesis of azaindoles
US6117681A (en) * 1995-03-29 2000-09-12 Bavarian Nordic Research Inst. A/S Pseudotyped retroviral particles
US5712285A (en) * 1995-04-07 1998-01-27 Merck, Sharp & Dohme, Ltd. Pyrrolo-pyridine derivatives
US6110456A (en) * 1995-06-07 2000-08-29 Yale University Oral delivery or adeno-associated viral vectors
US5856174A (en) * 1995-06-29 1999-01-05 Affymetrix, Inc. Integrated nucleic acid diagnostic device
US6107478A (en) * 1995-09-08 2000-08-22 Pedersen; Finn Skou Retroviral vector, a replication system for said vector and avian or mammalian cells transfected with said vector
US5747276A (en) * 1995-09-15 1998-05-05 The Scripps Research Institute Screening methods for the identification of novel antibiotics
US5721118A (en) * 1995-10-31 1998-02-24 The Regents Of The University Of California, San Diego Mammalian artificial chromosomes and methods of using same
US6022963A (en) * 1995-12-15 2000-02-08 Affymetrix, Inc. Synthesis of oligonucleotide arrays using photocleavable protecting groups
US6013440A (en) * 1996-03-11 2000-01-11 Affymetrix, Inc. Nucleic acid affinity columns
US6025155A (en) * 1996-04-10 2000-02-15 Chromos Molecular Systems, Inc. Artificial chromosomes, uses thereof and methods for preparing artificial chromosomes
US6545014B2 (en) * 1996-04-15 2003-04-08 Texas Biotechnology Corporation Method of treating glaucoma
US5959098A (en) * 1996-04-17 1999-09-28 Affymetrix, Inc. Substrate preparation process
US5965452A (en) * 1996-07-09 1999-10-12 Nanogen, Inc. Multiplexed active biologic array
US6258606B1 (en) * 1996-07-09 2001-07-10 Nanogen, Inc. Multiplexed active biologic array
US6294330B1 (en) * 1997-01-31 2001-09-25 Odyssey Pharmaceuticals Inc. Protein fragment complementation assays for the detection of biological or drug interactions
US6243980B1 (en) * 1997-03-07 2001-06-12 Tropix, Inc. Protease inhibitor assay
US6096718A (en) * 1997-06-05 2000-08-01 Gene Targeting Corp. Tissue specific adenovirus vectors for breast cancer treatment
US6235769B1 (en) * 1997-07-03 2001-05-22 Sugen, Inc. Methods of preventing and treating neurological disorders with compounds that modulate the function of the C-RET receptor protein tyrosine kinase
US20010018642A1 (en) * 1997-07-25 2001-08-30 David Balaban Method and system for providing a probe array chip design database
US20010019827A1 (en) * 1997-09-11 2001-09-06 Dawson Elliott P. Method of making high density arrays
US6178384B1 (en) * 1997-09-29 2001-01-23 The Trustees Of Columbia University In The City Of New York Method and apparatus for selecting a molecule based on conformational free energy
US20010014448A1 (en) * 1997-09-30 2001-08-16 Surmodics, Inc. Target molecule attachment to surfaces
US6048695A (en) * 1998-05-04 2000-04-11 Baylor College Of Medicine Chemically modified nucleic acids and methods for coupling nucleic acids to solid support
US6288234B1 (en) * 1998-06-08 2001-09-11 Advanced Medicine, Inc. Multibinding inhibitors of microsomal triglyceride transferase protein
US6113913A (en) * 1998-06-26 2000-09-05 Genvec, Inc. Recombinant adenovirus
US6350786B1 (en) * 1998-09-22 2002-02-26 Hoffmann-La Roche Inc. Stable complexes of poorly soluble compounds in ionic polymers
US6277628B1 (en) * 1998-10-02 2001-08-21 Incyte Genomics, Inc. Linear microarrays
US6277489B1 (en) * 1998-12-04 2001-08-21 The Regents Of The University Of California Support for high performance affinity chromatography and other uses
US20010016322A1 (en) * 1999-04-27 2001-08-23 Caren Michael P. Method of performing array-based hybridization assays using thermal inkjet deposition of sample fluids
US6310074B1 (en) * 1999-05-19 2001-10-30 Adir Et Compagnie Substituted dimeric compounds
US20010012537A1 (en) * 1999-07-30 2001-08-09 Anderson Norman G. Dry deposition of materials for microarrays using matrix displacement
US20010008765A1 (en) * 1999-12-06 2001-07-19 Fuji Photo Film Co., Ltd. DNA chip and reactive solid carrier
US20040002534A1 (en) * 1999-12-22 2004-01-01 Sugen, Inc. Methods of modulating c-kit tyrosine protein kinase function with indolinone compounds
US20030003004A1 (en) * 2001-06-13 2003-01-02 Stones Ian David Lubricating systems for regenerative vacuum pumps
US6897207B2 (en) * 2001-06-21 2005-05-24 Aventis Pharmaceuticals Inc. Azaindoles
US6858860B2 (en) * 2001-07-24 2005-02-22 Seiko Epson Corporation Apparatus and method for measuring natural period of liquid
US20040077595A1 (en) * 2002-09-06 2004-04-22 Insert Therapeutics, Inc. Cyclodextrin-based polymers for therapeutics delivery
US20040142864A1 (en) * 2002-09-16 2004-07-22 Plexxikon, Inc. Crystal structure of PIM-1 kinase
US20050154014A1 (en) * 2003-01-06 2005-07-14 Jason Bloxham (2-carboxamido)(3-amino) thiophene compounds
US20050085463A1 (en) * 2003-01-23 2005-04-21 Weiner David M. Use of N-desmethylclozapine to treat human neuropsychiatric disease
US20040167030A1 (en) * 2003-02-14 2004-08-26 Wyeth Heterocyclyl-3-sulfonylazaindole or-azaindazole derivatives as 5-hydroxytryptamine-6 ligands
US7259165B2 (en) * 2003-02-14 2007-08-21 Wyeth Heterocyclyl-3-sulfonylazaindole or-azaindazole derivatives as 5-hydroxytryptamine-6 ligands
US20050170431A1 (en) * 2003-02-28 2005-08-04 Plexxikon, Inc. PYK2 crystal structure and uses
US20050164300A1 (en) * 2003-09-15 2005-07-28 Plexxikon, Inc. Molecular scaffolds for kinase ligand development
US20070049615A1 (en) * 2003-12-19 2007-03-01 Plexxikon, Inc. Compounds and methods for development of Ret modulators
US7504509B2 (en) * 2003-12-19 2009-03-17 Plexxikon, Inc. Compounds and methods for development of Ret modulators
US20050256151A1 (en) * 2003-12-24 2005-11-17 Pharmacia Italia S.P.A. Pyrrolo[2,3-b]pyridine derivatives active as kinase inhibitors, process for their preparation and pharmaceutical compositions comprising them
US20060058340A1 (en) * 2004-06-17 2006-03-16 Ibrahim Prabha N Compounds modulating c-kit activity
US7498342B2 (en) * 2004-06-17 2009-03-03 Plexxikon, Inc. Compounds modulating c-kit activity
US20060058339A1 (en) * 2004-06-17 2006-03-16 Ibrahim Prabha N Compounds modulating c-kit activity and uses therefor
US7582637B2 (en) * 2004-07-27 2009-09-01 Sgx Pharmaceuticals, Inc. Pyrrolo-pyridine kinase modulators
US20060035898A1 (en) * 2004-07-27 2006-02-16 Structural Genomix, Inc. Fused ring heterocycle kinase modulators
US7601839B2 (en) * 2004-07-27 2009-10-13 Sgx Pharmaceuticals Inc. Pyrrolo-pyridine kinase modulators
US20090143352A1 (en) * 2004-07-27 2009-06-04 Sgx Pharmaceuticals, Inc. Pyrrolo-pyridine kinase modulatiors
US7452993B2 (en) * 2004-07-27 2008-11-18 Sgx Pharmaceuticals, Inc. Fused ring heterocycle kinase modulators
US7361763B2 (en) * 2004-07-27 2008-04-22 Sgx Pharmaceuticals, Inc. Pyrrolo-pyridine kinase modulators
US7361764B2 (en) * 2004-07-27 2008-04-22 Sgx Pharmaceuticals, Inc. Pyrrolo-pyridine kinase modulators
US20070032519A1 (en) * 2005-05-17 2007-02-08 Chao Zhang Compounds modulating c-kit and c-fms activity and uses therefor
US7863288B2 (en) * 2005-06-22 2011-01-04 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
US20090076046A1 (en) * 2006-11-22 2009-03-19 Plexxikon Inc Compounds modulating c-fms and/or c-kit activity and uses therefor
US20080188514A1 (en) * 2006-12-21 2008-08-07 Guoxian Wu Compounds and methods for kinase modulation, and indications therefor
US20080167338A1 (en) * 2006-12-21 2008-07-10 Wayne Spevak Compounds and methods for kinase modulation, and indications therefor
US7863289B2 (en) * 2006-12-21 2011-01-04 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
US20090005356A1 (en) * 2007-05-29 2009-01-01 Sgx Pharmaceuticals, Inc. Substituted pyrrolopyridines and pyrazolopyridines as kinase modulators

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Arthan et al., cancer letters, 297 (2010), pages 31-41. *
Roberts et al., Oncogene (2007), 26, pages 3291-3310. *

Cited By (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9169250B2 (en) 2006-11-22 2015-10-27 Plexxikon Inc. Compounds modulating c-fms and/or c-kit activity and uses therefor
US9487515B2 (en) 2006-11-22 2016-11-08 Plexxikon Inc. Compounds modulating c-fms and/or c-kit activity and uses therefor
US9844539B2 (en) 2007-07-17 2017-12-19 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10426760B2 (en) 2007-07-17 2019-10-01 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9469640B2 (en) 2007-07-17 2016-10-18 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9260437B2 (en) 2008-05-19 2016-02-16 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9447089B2 (en) 2009-04-03 2016-09-20 Plexxikon Inc. Compositions and uses thereof
US9663517B2 (en) 2009-04-03 2017-05-30 Plexxikon Inc. Compositions and uses thereof
US9096593B2 (en) 2009-11-06 2015-08-04 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9617267B2 (en) 2009-11-18 2017-04-11 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9440969B2 (en) 2009-12-23 2016-09-13 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9682981B2 (en) 2010-04-21 2017-06-20 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US11337976B2 (en) 2011-02-07 2022-05-24 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9624213B2 (en) 2011-02-07 2017-04-18 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9550768B2 (en) 2011-05-17 2017-01-24 Plexxikon Inc. Kinase modulation and indications therefor
US9358235B2 (en) 2012-03-19 2016-06-07 Plexxikon Inc. Kinase modulation, and indications therefor
US10123998B2 (en) 2012-03-19 2018-11-13 Plexxikon Inc. Kinase modulation, and indications therefor
US9730918B2 (en) 2012-03-19 2017-08-15 Plexxikon Inc. Kinase modulation, and indications therefor
US20140094611A1 (en) * 2012-05-31 2014-04-03 Plexxikon Inc. Synthesis of heterocyclic compounds
US9695169B2 (en) 2012-05-31 2017-07-04 Plexxikon Inc. Synthesis of heterocyclic compounds
US9150570B2 (en) * 2012-05-31 2015-10-06 Plexxikon Inc. Synthesis of heterocyclic compounds
US9399639B2 (en) 2012-08-27 2016-07-26 Calitor Sciences, Llc Substituted azaindole compounds, salts, pharmaceutical compositions thereof and methods of use
US10227357B2 (en) 2012-09-06 2019-03-12 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10301280B2 (en) 2012-12-21 2019-05-28 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9676748B2 (en) 2012-12-21 2017-06-13 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9822109B2 (en) 2013-03-15 2017-11-21 Plexxikon Inc. Heterocyclic compounds and uses thereof
US9718847B2 (en) 2013-03-15 2017-08-01 Plexxikon Inc. Heterocyclic compounds and uses thereof
US9873700B2 (en) 2013-05-30 2018-01-23 Plexxikon Inc. Compounds for kinase modulation, and indications therefor
US10421761B2 (en) 2013-05-30 2019-09-24 Plexxikon Inc. Compounds for kinase modulation, and indications therefor
US9771369B2 (en) 2014-03-04 2017-09-26 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9856259B2 (en) 2014-09-15 2018-01-02 Plexxikon Inc. Heterocyclic compounds and uses thereof
US10160755B2 (en) 2015-04-08 2018-12-25 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10829484B2 (en) 2015-07-28 2020-11-10 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10370374B2 (en) 2015-09-21 2019-08-06 Plexxikon Inc. Heterocyclic compounds and uses thereof
US10899761B2 (en) 2015-09-21 2021-01-26 Plexxikon Inc. Heterocyclic compounds and uses thereof
US9771363B2 (en) 2015-09-21 2017-09-26 Plexxikon Inc. Heterocyclic compounds and uses thereof
US10647716B2 (en) 2015-09-21 2020-05-12 Plexxikon Inc. Heterocyclic compounds and uses thereof
US9975894B2 (en) 2015-09-21 2018-05-22 Plexxikon Inc. Heterocyclic compounds and uses thereof
US9938273B2 (en) 2015-12-07 2018-04-10 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10160747B2 (en) 2016-03-16 2018-12-25 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10508085B2 (en) 2016-09-22 2019-12-17 Plexxikon Inc. Compounds and methods for IDO and TDO modulation, and indications therefor
US10703757B2 (en) 2016-12-23 2020-07-07 Plexxikon Inc. Compounds and methods for CDK8 modulation and indications therefor
US10577366B2 (en) 2017-03-20 2020-03-03 Plexxikon Inc. Crystalline forms of a compound that inhibits bromodomain
US10428067B2 (en) 2017-06-07 2019-10-01 Plexxikon Inc. Compounds and methods for kinase modulation
US10961240B2 (en) 2017-07-25 2021-03-30 Plexxikon Inc. Formulations of a compound modulating kinases
US10941142B2 (en) 2017-07-25 2021-03-09 Plexxikon Inc. Formulations of a compound modulating kinases
US10435404B2 (en) 2017-07-25 2019-10-08 Plexxikon Inc. Formulations of a compound modulating kinases
US10717735B2 (en) 2017-10-13 2020-07-21 Plexxikon Inc. Solid forms of a compound for modulating kinases
US11103505B2 (en) 2017-10-27 2021-08-31 Plexxikon Inc. Formulations of a compound modulating kinases
US11149011B2 (en) 2018-03-20 2021-10-19 Plexxikon Inc. Compounds and methods for IDO and TDO modulation, and indications therefor
WO2021158936A1 (en) * 2020-02-05 2021-08-12 The Rockefeller University Pyrrolo [2,3-b]pyridine-3-carboxamide compositions and methods for ameliorating hearing loss

Also Published As

Publication number Publication date
US7863289B2 (en) 2011-01-04
PE20081581A1 (en) 2008-11-12
NO20092251L (en) 2009-07-15
PE20121126A1 (en) 2012-08-24
MA31153B1 (en) 2010-02-01
CR10860A (en) 2009-06-30
KR20090095654A (en) 2009-09-09
CN101668757A (en) 2010-03-10
UY30816A1 (en) 2008-07-31
HN2009001202A (en) 2012-06-04
RU2009122436A (en) 2011-01-27
MY148468A (en) 2013-04-30
ECSP099444A (en) 2009-07-31
EP2097414A1 (en) 2009-09-09
MX2009006162A (en) 2009-06-19
US20080167338A1 (en) 2008-07-10
AU2007336906A1 (en) 2008-07-03
WO2008079903A1 (en) 2008-07-03
UA98629C2 (en) 2012-06-11
TW200845975A (en) 2008-12-01
AR064476A1 (en) 2009-04-01
CA2673000A1 (en) 2008-07-03
CO6190519A2 (en) 2010-08-19
WO2008079906A1 (en) 2008-07-03
BRPI0722088A2 (en) 2016-03-22
JP2010514687A (en) 2010-05-06
CL2007003797A1 (en) 2008-07-18

Similar Documents

Publication Publication Date Title
US7863289B2 (en) Compounds and methods for kinase modulation, and indications therefor
US8268858B2 (en) Compounds and methods for kinase modulation, and indications therefor
US10426760B2 (en) Compounds and methods for kinase modulation, and indications therefor
US7872018B2 (en) Compounds and methods for kinase modulation, and indications therefor
US9260437B2 (en) Compounds and methods for kinase modulation, and indications therefor
US8198273B2 (en) Compounds and methods for kinase modulation, and indications therefor
US8153641B2 (en) Compounds and methods for kinase modulation, and indications therefor
EP1893612B1 (en) Pyrrolo [2, 3-b]pyridine derivatives as protein kinase inhibitors

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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