US20170319559A1 - 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
US20170319559A1
US20170319559A1 US15/656,990 US201715656990A US2017319559A1 US 20170319559 A1 US20170319559 A1 US 20170319559A1 US 201715656990 A US201715656990 A US 201715656990A US 2017319559 A1 US2017319559 A1 US 2017319559A1
Authority
US
United States
Prior art keywords
difluoro
phenyl
fluoro
sulfonylamino
amide
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
US15/656,990
Inventor
Guoxian Wu
Jiazhong Zhang
Yong-Liang Zhu
Chao Zhang
Prabha N. Ibrahim
Songyuan Shi
Wayne Spevak
Dean R. Artis
James Tsai
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=39745045&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20170319559(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 US15/656,990 priority Critical patent/US20170319559A1/en
Publication of US20170319559A1 publication Critical patent/US20170319559A1/en
Assigned to PLEXXIKON INC. reassignment PLEXXIKON INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SHI, SONGYUAN, TSAI, JAMES, ARTIS, DEAN R., IBRAHIM, PRABHA N., SPEVAK, WAYNE, WU, GUOXIAN, ZHANG, CHAO, ZHANG, JIAZHONG, ZHU, YONG-LIANG
Priority to US16/058,945 priority patent/US20190209536A1/en
Priority to US16/109,199 priority patent/US10426760B2/en
Priority to US17/372,330 priority patent/US20210346358A1/en
Priority to US17/372,346 priority patent/US20210353602A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • 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
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/433Thidiazoles
    • 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
    • A61K31/4353Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • 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
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4436Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a heterocyclic ring having sulfur as a ring hetero atom
    • 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
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • 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
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • 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
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • 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
    • A61K31/47Quinolines; Isoquinolines
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • 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
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/549Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame having two or more nitrogen atoms in the same ring, e.g. hydrochlorothiazide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • 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
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • 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
    • A61P11/02Nasal agents, e.g. decongestants
    • 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
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • 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
    • 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
    • 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
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • 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
    • A61P3/06Antihyperlipidemics
    • 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
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • 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
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • 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
    • 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
    • A61P9/08Vasodilators for multiple indications
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/72Nitrogen atoms
    • C07D213/74Amino or imino radicals substituted by hydrocarbon or substituted hydrocarbon radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/72Nitrogen atoms
    • C07D213/75Amino or imino radicals, acylated by carboxylic or carbonic acids, or by sulfur or nitrogen analogues thereof, e.g. carbamates
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/72Nitrogen atoms
    • C07D213/76Nitrogen atoms to which a second hetero atom is attached
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/81Amides; Imides
    • C07D213/82Amides; Imides in position 3
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/12Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/12Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D215/14Radicals substituted by oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/20Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/38Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/14Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D231/38Nitrogen atoms
    • C07D231/40Acylated on said nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/48Two nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/48Two nitrogen atoms
    • C07D239/49Two nitrogen atoms with an aralkyl radical, or substituted aralkyl radical, attached in position 5, e.g. trimethoprim
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D261/00Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings
    • C07D261/02Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings
    • C07D261/06Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings having two or more double bonds between ring members or between ring members and non-ring members
    • C07D261/10Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings having two or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D261/14Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/02Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings
    • C07D263/30Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D263/34Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D263/48Nitrogen atoms not forming part of a nitro radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D285/00Heterocyclic compounds containing rings having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by groups C07D275/00 - C07D283/00
    • C07D285/01Five-membered rings
    • C07D285/02Thiadiazoles; Hydrogenated thiadiazoles
    • C07D285/04Thiadiazoles; Hydrogenated thiadiazoles not condensed with other rings
    • C07D285/121,3,4-Thiadiazoles; Hydrogenated 1,3,4-thiadiazoles
    • C07D285/1251,3,4-Thiadiazoles; Hydrogenated 1,3,4-thiadiazoles with oxygen, sulfur or nitrogen atoms, directly attached to ring carbon atoms, the nitrogen atoms not forming part of a nitro radical
    • C07D285/135Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D285/00Heterocyclic compounds containing rings having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by groups C07D275/00 - C07D283/00
    • C07D285/15Six-membered rings
    • C07D285/16Thiadiazines; Hydrogenated thiadiazines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • 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

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.
  • Compounds are contemplated that are active on protein kinases in general, including, but not limited to, Ab1, 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, Mnk1,
  • 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:
  • the compound of Formula I has a structure according to the following sub-generic structure Formula Ia:
  • L 1 is a bond, —N(R 11 )—, —N(R 11 )—C(X)—, —N(R 11 )—S(O) 2 —, —N(R 11 )—C(NH)—, —N(R 11 )—C(X)—N(R 11 )—, or —N(R 11 )—S(O) 2 —N(R 11 )—, also —N(R 11 )—, —N(R 11 )—C(X)—, or —N(R 11 )—S(O) 2 —, also —N(R 11 )—C(O)—, wherein the left side (i.e.
  • L 1 is a bond, —N(R 11 )—, —N(R 11 )—C(X)—, —N(R 11 )—S(O) 2 —, —N(R 11 )—C(NH)—, —N(R 11 )—C(X)—N(R 11 )—, or —N(R 11 )—S(O) 2 —N(R 11 )—, also —N(R 11 )—, —N(R 11 )—C(X)—, or —N(R 11 )—S(O) 2 —, also —N(R 11 )—C(O)—, and each R 11 and R 4 are independently hydrogen or lower alkyl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group
  • L 1 is —C(X)—N(R 1 )—, —C(R 12 R 13 )—X—, —X—C(R 12 R 13 )—, —C(R 12 R 13 )—N(R 11 )—, or —N(R 11 )—C(R 12 R 13 )—, wherein the left side of L 1 is attached to Ar and the right side of L 1 is attached to the phenyl ring of Formula I or Ia.
  • L 1 is —C(X)—N(R 11 )—, —C(R 12 R 13 )—X—, —X—C(R 12 R 13 )—, —C(R 12 R 13 )—N(R 11 )—, or —N(R 11 )—C(R 12 R 13 )—, and each R 11 and R 4 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably each R 11 and R 4 are H.
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro.
  • R 2 is hydrogen, fluoro or chloro
  • each R 11 and R 4 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 2 is fluoro or chloro and each R 11 and R 4 are H.
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro
  • L 1 is a bond, —N(R 11 )—, —N(R 11 )—C(X)—, —N(R 11 )—S(O) 2 —, —N(R 11 )—C(NH)—, —N(R 11 )—C(X)—N(R 11 )—, or —N(R 11 )—S(O) 2 —N(R 11 )—, also —N(R 11 )—, —N(R 11 )—C(X)—, or —N(R 1 )—S(O) 2 —, also —N(R 1 )—C(O)—, wherein the left side (i.e.
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro;
  • L 1 is a bond, —N(R 11 )—, —N(R 11 )—C(X)—, —N(R 11 )—S(O) 2 —, —N(R 11 )—C(NH)—, —N(R 11 )—C(X)—N(R 11 )—, or —N(R 11 )—S(O) 2 —N(R 11 )—, also —N(R 11 )—, —N(R 11 )—C(X)—, or —N(R 11 )—S(O) 2 —, also —N(R 11 )—C(X)—, or —N(R 11 )—S(O) 2 —, also —N(R 11 )—C(O)—; and each R 11 and R 4 are independently hydrogen or lower alkyl, where
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro.
  • R 2 is hydrogen, fluoro or chloro
  • each R 11 and R 4 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 2 is fluoro or chloro and each R 11 and R 4 are H.
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro
  • L 1 is —C(X)—N(R 11 )—, —C(R 12 R 13 )—X—, —X—C(R 12 R 13 )—, —C(R 12 R 13 )—N(R 11 )—, or —N(R 11 )—C(R 12 R 13 )—, wherein the left side of L 1 is attached to Ar and the right side of L 1 is attached to the phenyl ring of Formula I or Ia.
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro;
  • L 1 is —C(X)—N(R 11 )—, —C(R 12 R 13 )—X—, —X—C(R 12 R 13 )—, —C(R 12 R 13 )—N(R 11 )—, or —N(R 11 )—C(R 12 R 13 )—; and each R 11 and R 4 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably each R 11 and R 4 are H.
  • the compound of Formula I has a structure according to the following sub-generic structure Formula Ib:
  • R 4 , R 11 , R 12 and R 13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 4 , R 11 , R 12 and R 13 are H.
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro.
  • R 2 is hydrogen, fluoro or chloro
  • R 4 , R 11 , R 12 and R 13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 2 is fluoro or chloro and R 4 , R 11 , R 12 and R 13 are H.
  • A is —C(O)—, and R 4 and R 11 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 4 and R 11 are H.
  • A is —C(O)—, and R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro.
  • A is —C(O)—
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro
  • R 4 and R 11 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 4 and R 11 are H.
  • A is —C(R 12 R 13 )—, and R 4 , R 11 , R 12 and R 13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 4 , R 11 , R 12 and R 13 are H.
  • A is —C(R 12 R 13 )—
  • R 12 and R 13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 12 and R 13 are H, and R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro.
  • A is —C(R 12 R 13 )—;
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro;
  • R 4 , R 11 , R 12 and R 13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 4 , R 11 , R 12 and R 13 are H.
  • the compound of Formula I has a structure according to the following sub-generic structure Formula Ic:
  • R 4 is hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 4 is H.
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro.
  • R 2 is hydrogen, fluoro or chloro and R 4 is hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 2 is fluoro or chloro and R 4 is H.
  • the compound of Formula I has a structure according to the following sub-generic structure Formula Id:
  • R 4 , R 11 , R 12 and R 13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 4 , R 11 , R 12 and R 13 are H.
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro.
  • R 2 is hydrogen, fluoro or chloro
  • R 4 , R 11 , R 12 and R 13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 2 is fluoro or chloro and R 4 , R 11 , R 12 and R 13 are H.
  • A is —C(O)—, and R 4 and R 11 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 4 and R 11 are H.
  • A is —C(O)—, and R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro.
  • A is —C(O)—
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro
  • R 4 and R 11 are hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 4 and R 11 are H.
  • A is —C(R 12 R 13 )—, and R 4 , R 11 , R 12 and R 13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 4 , R 11 , R 12 and R 13 are H.
  • A is —C(R 12 R 13 )—
  • R 12 and R 13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 12 and R 13 are H, and R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro.
  • A is —C(R 12 R 13 )—;
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro;
  • R 4 , R 11 , R 12 and R 13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 4 , R 11 , R 12 and R 13 are H.
  • the compound of Formula I has a structure according to the following sub-generic structure Formula Ie:
  • R 4 is hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 4 is H.
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro.
  • R 2 is hydrogen, fluoro or chloro and R 4 is hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 2 is fluoro or chloro and R 4 is H.
  • the compound of Formula I has a structure according to the following sub-generic structure Formula If:
  • L 3 is —C(X)—N(R 11 )—, —C(R 12 R 13 )—X—, —X—C(R 12 R 13 )—, —C(R 12 R 13 )—N(R 11 )—, or —N(R 11 )—C(R 12 R 13 )—; and m, Ar, R 1 , R 2 , R 3 , R 4 , R 11 , R 12 , R 13 , and X are as defined for Formula I.
  • L 3 is —C(O)—N(R 11 )—, —C(R 12 R 13 )—O—, —O—C(R 12 R 13 )—, —C(R 12 R 13 )—N(R 11 )—, or —N(R 11 )—C(R 12 R 13 )—, wherein the left side of L 3 is attached to Ar and the right side of L 3 is attached to the phenyl ring of Formula If.
  • L 3 is —C(O)—N(R 11 )—, —C(R 12 R 13 )—O—, —O—C(R 12 R 13 )—, —C(R 12 R 13 )—N(R 11 )—, or —N(R 11 )—C(R 12 R 13 )—
  • R 4 , R 11 , R 12 and R 13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 4 , R 11 , R 12 and R 13 are H.
  • the compound of Formula I has a structure according to the following sub-generic structure Formula Ig:
  • L 3 is —C(X)—N(R 11 )—, —C(R 12 R 13 )—X—, —X—C(R 12 R 13 )—, —C(R 12 R 13 )—N(R 11 )—, or —N(R 11 )—C(R 12 R 13 )—; and m, Ar, R 1 , R 2 , R 3 , R 4 , R 11 , R 12 , R 13 , X and L 2 are as defined for Formula I.
  • L 3 is —C(O)—N(R 11 )—, —C(R 12 R 13 )—O—, —O—C(R 12 R 13 )—, —C(R 12 R 13 )—N(R 11 )—, or —N(R 11 )—C(R 12 R 13 )—, wherein the left side of L 3 is attached to Ar and the right side of L 3 is attached to the phenyl ring of Formula Ig.
  • L 3 is —C(O)—N(R 11 )—, —C(R 12 R 13 )—O—, —O—C(R 12 R 13 )—, —C(R 12 R 13 )—N(R 11 )—, or —N(R 11 )—C(R 12 R 13 )—
  • R 4 , R 11 , R 12 and R 13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 4 , R 11 , R 12 and R 13 are H.
  • the compound of Formula I has a structure according to the following sub-generic structure Formula Ih:
  • R 4 , R 11 , R 12 and R 13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 4 , R 11 , R 12 and R 13 are H.
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro.
  • R 2 is hydrogen, fluoro or chloro
  • R 4 , R 11 , R 12 and R 13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 2 is fluoro or chloro and R 4 , R 11 , R 12 and R 13 are H.
  • A is —C(O)—, and R 4 and R 11 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 4 and R 11 are H.
  • A is —C(O)—, and R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro.
  • A is —C(O)—
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro
  • R 4 and R 11 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 4 and R 11 are H.
  • A is —C(R 12 R 13 )—, and R 4 , R 11 , R 12 and R 13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 4 , R 11 , R 12 and R 13 are H.
  • A is —C(R 12 R 13 )—
  • R 12 and R 13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 12 and R 13 are H, and R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro.
  • A is —C(R 12 R 13 )—;
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro;
  • R 4 , R 11 , R 12 and R 13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 4 , R 11 , R 12 and R 13 are H.
  • the compound of Formula I has a structure according to the following sub-generic structure Formula Ii:
  • R 4 , R 11 , R 12 and R 13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 4 , R 11 , R 12 and R 13 are H.
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro.
  • R 2 is hydrogen, fluoro or chloro
  • R 4 , R 11 , R 12 and R 13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 2 is fluoro or chloro and R 4 , R 11 , R 12 and R 13 are H.
  • A is —C(O)—, and R 4 and R 11 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 4 and R 11 are H.
  • A is —C(O)—, and R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro.
  • A is —C(O)—
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro
  • R 4 and R 11 are hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 4 and R 11 are H.
  • A is —C(R 12 R 13 )—, and R 4 , R 11 , R 12 and R 13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 4 , R 11 , R 12 and R 13 are H.
  • A is —C(R 12 R 13 )—
  • R 12 and R 13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 12 and R 13 are H, and R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro.
  • A is —C(R 12 R 13 )—;
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro;
  • R 4 , R 11 , R 12 and R 13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 4 , R 11 , R 12 and R 13 are H.
  • the compound of Formula I has a structure according to the following sub-generic structure Formula Ij:
  • L 1 is a bond, —N(R 11 )—, —N(R 11 )—C(X)—, —N(R 11 )—S(O) 2 —, —N(R 11 )—C(NH)—, —N(R 11 )—C(X)—N(R 11 )—, or —N(R 11 )—S(O) 2 —N(R 11 )—, also —N(R 11 )—, —N(R 11 )—C(X)—, or —N(R 11 )—S(O) 2 —, also —N(R 11 )—C(O)—, wherein the left side (i.e.
  • L 1 is a bond, —N(R 11 )—, —N(R 11 )—C(X)—, —N(R 11 )—S(O) 2 —, —N(R 11 )—C(NH)—, —N(R 11 )—C(X)—N(R 11 )—, or —N(R 11 )—S(O) 2 —N(R 11 )—, also —N(R 11 )—, —N(R 1 )—C(X)—, or —N(R 11 )—S(O) 2 —, also —N(R 11 )—C(O)—, and each R 11 and R 4 are independently hydrogen or lower alkyl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting
  • L 1 is —C(X)—N(R 11 )—, —C(R 12 R 13 )—X—, —X—C(R 12 R 13 )—, —C(R 12 R 13 )—N(R 11 )—, or —N(R 11 )—C(R 12 R 13 )—, wherein the left side of L 1 is attached to Ar and the right side of L 1 is attached to the phenyl ring of Formula Ij.
  • L 1 is —C(X)—N(R 11 )—, —C(R 12 R 13 )—X—, —X—C(R 12 R 13 )—, —C(R 12 R 13 )—N(R 11 )—, or —N(R 11 )—C(R 12 R 13 )—, and each R 11 and R 4 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably each R 11 and R 4 are H.
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro.
  • R 2 is hydrogen, fluoro or chloro
  • each R 11 and R 4 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 2 is fluoro or chloro and each R 11 and R 4 are H.
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro
  • L 1 is a bond, —N(R 11 )—, —N(R 1 )—C(X)—, —N(R 11 )—S(O) 2 —, —N(R 11 )—C(NH)—, —N(R 11 )—C(X)—N(R 11 )—, or —N(R 11 )—S(O) 2 —N(R 11 )—, also —N(R 11 )—, —N(R 11 )—C(X)—, or —N(R 11 )—S(O) 2 —, also —N(R 11 )—C(O)—, wherein the left side (i.e.
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro;
  • L 1 is a bond, —N(R 11 )—, —N(R 11 )—C(X)—, —N(R 11 )—S(O) 2 —, —N(R 11 )—C(NH)—, —N(R 11 )—C(X)—N(R 11 )—, or —N(R 11 )—S(O) 2 —N(R 11 )—, also —N(R 11 )—, —N(R 1 )—C(X)—, or —N(R 11 )—S(O) 2 —, also —N(R 11 )—C(O)—; and each R 11 and R 4 are independently hydrogen or lower alkyl, wherein lower
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro.
  • R 2 is hydrogen, fluoro or chloro
  • each R 11 and R 4 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R 2 is fluoro or chloro and each R 11 and R 4 are H.
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro
  • L 1 is —C(X)—N(R 11 )—, —C(R 12 R 13 )—X—, —X—C(R 12 R 13 )—, —C(R 12 R 13 )—N(R 11 )—, or —N(R 11 )—C(R 12 R 13 )—, wherein the left side of L 1 is attached to Ar and the right side of L 1 is attached to the phenyl ring of Formula Ij.
  • R 2 is hydrogen, fluoro or chloro, preferably fluoro or chloro;
  • L 1 is —C(X)—N(R 11 )—, —C(R 12 R 13 )—X—, —X—C(R 12 R 13 )—, —C(R 12 R 13 )—N(R 11 )—, or —N(R 11 )—C(R 12 R 13 )—; and each R 11 and R 4 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably each R 11 and R 4 are H.
  • R 22 is mono-alkylamino, di-alkylamino, or optionally substituted heterocycloalkyl, preferably wherein heterocycloalkyl is a 5 or 6 membered nitrogen containing heterocycloalkyl, wherein a nitrogen of the heterocycloalkyl is bound to the S(O) 2 of Formula Ij.
  • R 22 is mono-alkylamino, di-alkylamino or 5 or 6 membered nitrogen containing heterocycloalkyl, wherein the heterocycloalkyl is substituted with one or more substituents selected from the group consisting of fluoro, —OH, —NH 2 , lower alkyl, lower alkoxy, lower alkylthio, mono-alkylamino, di-alkylamino, and cycloalkylamino, wherein lower alkyl or the alkyl chain(s) of lower alkoxy, lower alkylthio, mono-alkylamino, or di-alkylamino are optionally substituted with one or more substituents selected from the group consisting of fluoro, —OH, —NH 2 , lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino and cycloalky
  • Ar is monocyclic or bicyclic nitrogen containing heteroaryl. In some embodiments, Ar is selected from the group consisting of
  • Ar is selected from the group consisting of
  • Ar is selected from the group consisting of
  • Ar is selected from the group consisting of
  • p 0, 1, 2 or 3;
  • R 16 substituting at any available CH or NH, is as defined in paragraph [0041].
  • Ar is selected from the group consisting of
  • R 1 is selected from the group consisting of
  • R 3 is optionally substituted lower alkyl or optionally substituted C 3-6 cycloalkyl.
  • R 3 is lower alkyl or C 3-6 cycloalkyl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, —OH, —NH 2 , lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, fluoro substituted mono-alkylamino, di-alkylamino, fluoro substituted di-alkylamino, cycloalkylamino, and C 3-5 cycloalkyl, and wherein C 3-6 cycloalkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, —OH, —NH 2 , lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, fluoro substituted mono-alkyla
  • R 3 is lower alkyl or C 3-6 cycloalkyl, wherein lower alkyl or C 3-6 cycloalkyl are optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, and fluoro substituted lower alkylthio. In some embodiments, R 3 is optionally fluoro substituted lower alkyl or optionally fluoro substituted C 3-6 cycloalkyl.
  • R 3 is lower alkyl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, —OH, —NH 2 , lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, fluoro substituted mono-alkylamino, di-alkylamino, fluoro substituted di-alkylamino, cycloalkylamino, also one or more substituents selected from the group consisting of fluoro, —OH, —NH 2 , lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, fluoro substituted mono-alkylamino, di-alkylamino, and fluoro substituted di-alkylamino, also one or more substituents selected from the group consisting of fluoro
  • R 3 is optionally substituted phenyl, also phenyl mono-substituted at the para position, also phenyl mono-substituted at the meta position.
  • R 3 is phenyl optionally substituted with one or more substituents selected from the group consisting of halogen, —CN, —NO 2 , —OH, —NH 2 , lower alkyl, lower alkoxy, lower alkylthio, mono-alkylamino, di-alkylamino, and cycloalkylamino, wherein lower alkyl or the alkyl chain(s) of lower alkoxy, lower alkylthio, mono-alkylamino, or di-alkylamino are optionally substituted with one or more substituents selected from the group consisting of fluoro, —OH, —NH 2 , lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, di-alkylamino and cycloalkylamino, preferably wherein the phenyl is mono-substituted at either the para or
  • R 3 is phenyl optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, and fluoro substituted lower alkylthio, preferably wherein the phenyl is mono-substituted at either the para or meta position.
  • the compound is selected from the group consisting of:
  • a compound or group of compounds includes pharmaceutically acceptable salts of such compound(s), pharmaceutically acceptable formulations of such compound(s), prodrug(s), and all stereoisomers thereof.
  • a compound of Formula I includes all sub-embodiments thereof (e.g. including Formula Ia, Ib, Ic, Id, Ie, If, Ig, Ih, Ii, and Ij and all embodiments as described above).
  • 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 compound(s) of Formula I.
  • treat refers to the administration of material, e.g., one or more compound(s) of Formula I, 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, and/or 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 modulators, including compounds described herein, provides a therapeutic benefit to the subject suffering from or at risk of the disease or condition.
  • the protein kinase modulator is an inhibitor of the protein kinase.
  • the method involves administering to the subject an effective amount of one or more compound(s) of Formula I 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(s) of Formula I.
  • 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(s) of Formula I.
  • Raf protein kinase mediated disease or condition refers 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, mutations of A-Raf, 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/T529I.
  • 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/T529I mutant B-Raf.
  • the Raf protein kinase mediated disease or condition includes a disease or condition for which Raf modulation provides a therapeutic benefit, e.g. wherein treatment with Raf modulators, including compounds described herein, provides a therapeutic benefit to the subject suffering from or at risk of the disease or condition.
  • the Raf modulator is a Raf inhibitor.
  • the method involves administering to the subject an effective amount of a compound of Formula I 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 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 Formula I 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 Ab1, 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, Ja
  • a compound of Formula I 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 Ab1, Akt1, 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, MAP2
  • a compound of Formula I 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 Ab1, 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, MAP4K4, MAPKAPK2, Met, p38, PDGFRB, Pim1, PKC theta, Pyk2, Ret, Src, Stk6, TrkA, TrkB, Yes, and Zap70, including any mutations thereof.
  • a compound of Formula I 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 Ab1, A-Raf, B-Raf, Btk, c-Raf-1, EGFR, EphB2, Erk2, Fak, Fms, Irak4, Jnk1, Jnk2, Jnk3, Kit, Lck, Lyn, MAP2K1, MAP4K4, MAPKAPK2, Met, p38, Pim1, PKC theta, Pyk2, Src, Stk6, TrkA, TrkB, Yes, and Zap70, including any mutations thereof.
  • a compound of Formula I 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 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 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 A-Raf, B-Raf, c-Raf-1, B-Raf V600E mutant, or B-Raf V600E/T529I mutant.
  • a compound of Formula I will selectively inhibit one Raf kinase relative to one or more other Raf kinases.
  • the compound of Formula I 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 of Formula I 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 of Formula I 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.
  • 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(s) of Formula I and at least one pharmaceutically acceptable carrier, excipient, and/or diluent, including combinations of any two or more compounds of Formula I.
  • the composition can further include a plurality of different pharmacologically active compounds, which can include a plurality of compounds of Formula I.
  • the composition can include one or more compounds of Formula I 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 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 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 Ab1, 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, Mnk
  • 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(s) of Formula I.
  • methods for treating a disease or condition mediated by a protein kinase selected from the group consisting of Ab1, 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
  • the invention provides methods for treating a disease or condition mediated by a protein kinase selected from the group consisting of Ab1, Akt1, 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 Ab1, 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, MAP4K4, MAPKAPK2, Met, p38, PDGFRB, Pim1, PKC theta, Pyk2, Ret, Src, Stk6, TrkA, TrkB, Yes, and Zap70, including any mutations thereof, by administering to the subject an effective amount of a composition including one or more compound(s) of Formula I.
  • a protein kinase selected from the group consisting of Ab1, A-Raf, B-Raf, Btk, c-Raf-1, EGFR,
  • the invention provides methods for treating a disease or condition mediated by a protein kinase selected from the group consisting of Ab1, A-Raf, B-Raf, Btk, c-Raf-1, EGFR, EphB2, Erk2, Fak, Fms, Irak4, Jnk1, Jnk2, Jnk3, Kit, Lck, Lyn, MAP2K1, MAP4K4, MAPKAPK2, Met, p38, Pim1, PKC theta, Pyk2, Src, Stk6, TrkA, TrkB, Yes, and Zap70, including any mutations thereof, by administering to the subject an effective amount of a composition including one or more compound(s) of Formula I.
  • a protein kinase selected from the group consisting of Ab1, A-Raf, B-Raf, Btk, c-Raf-1, EGFR, EphB2, Erk2, Fak, Fms, Irak4, Jnk1, Jnk2,
  • 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, including any mutations thereof, by administering to the subject an effective amount of a composition including one or more compound(s) of Formula I.
  • 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, Jn
  • 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(s) of Formula I.
  • 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(s) of Formula I 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/T529I mutant by administering to the subject an effective amount of a composition including one or more compound(s) of Formula I 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(s) of Formula I, 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, Q-ray, or electron, proton, neutron, or D 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(s) of Formula I 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; an antibiotic, including,
  • the method of treating a cancer involves administering to the subject an effective amount of a composition including one or more compound(s) of Formula I 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(s) of Formula I, a prodrug of such compound, a pharmaceutically acceptable salt of such compound or prodrug, or a pharmaceutically acceptable formulation 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(s) of Formula I, a prodrug of such compound, a pharmaceutically acceptable salt of such compound or prodrug, or a pharmaceutically acceptable formulation 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 leukemia, acute lymphocytic leukemia, chronic myeloid leukemia, chronic lymphocytic leukemia, multiple myeloma, melanoma, mastocytosis, gliomas, glioblastoma, astrocytoma, neuroblastoma, sarcomas (e.g.
  • neurologic diseases including, but not limited to, cerebrovascular ischemia, multi-infarct dementia
  • 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 (e.g. ulcerative colitis, Crohn's disease), systemic lupus erythematosis, Sjogren's Syndrome, Wegener's granulomatosis, 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 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
  • compounds of Formula I can be used in the preparation of a medicament for the treatment of a 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 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
  • 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 (e.g. ulcerative colitis, Crohn's disease), systemic lupus erythematosis, Sjogren's Syndrome, Wegener's granulomatosis, 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 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
  • 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.
  • 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; 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 chronic obstructive pulmonary disease
  • ARDS acute respiratory distress
  • compounds of Formula I 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.
  • 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; 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, but
  • the compounds of Formula I 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 chemically feasible and 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.
  • an “optionally substituted lower alkyl” denotes lower alkyl that is optionally 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 , —N(H)—C(O)—NH 2 , —N(H)—C(S)—NH 2 , —N(H)—S(O) 2 —NH 2 , —C(NH)—NH 2 , —O—R o , —S—R o , —O—C(O)—R o , —O—
  • substitutions include subsets of these substitutions, such as are indicated herein, for example, in the description of compounds of Formula I, 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. It is understood that substitutions are chemically feasible and attached at any available atom to provide 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.
  • the straight chain or branched lower alkenyl group is chemically feasible and attached at any available point to provide a stable compound. Examples of lower alkenyl groups include ethenyl, propenyl, isopropenyl, butenyl, and the like.
  • an “optionally substituted lower alkenyl” denotes lower alkenyl that is optionally 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 , —N(H)—C(O)—NH 2 , —N(H)—C(S)—NH 2 , —N(H)—S(O) 2 —NH 2 , —C(NH)—NH 2 , —O—R o , —S—R o , —O—C(O)—R o , —
  • substitutions include subsets of these substitutions, such as are indicated herein, for example, in the description of compounds of Formula I, 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.
  • C 3-6 alkenyl denotes lower alkenyl containing 3-6 carbon atoms.
  • An “optionally substituted C 3-6 alkenyl” denotes optionally substituted lower alkenyl containing 3-6 carbon atoms. It is understood that substitutions are chemically feasible and attached at any available atom to provide a stable compound.
  • “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.
  • the straight chain or branched lower alkynyl group is chemically feasible and attached at any available point to provide a stable compound. Examples of alkynyl groups include ethynyl, propynyl, butynyl, and the like.
  • an “optionally substituted lower alkynyl” denotes lower alkynyl that is optionally 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 , —N(H)—C(O)—NH 2 , —N(H)—C(S)—NH 2 , —N(H)—S(O) 2 —NH 2 , —C(NH)—NH 2 , —O—R o , —S—R o , —O—C(O)—R o ,
  • substitutions include subsets of these substitutions, such as are indicated herein, for example, in the description of compounds of Formula I, 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.
  • C 3-6 alkynyl denotes lower alkynyl containing 3-6 carbon atoms.
  • An “optionally substituted C 3-6 alkynyl” denotes optionally substituted lower alkynyl containing 3-6 carbon atoms. It is understood that substitutions are chemically feasible and attached at any available atom to provide a stable compound.
  • 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.
  • An “optionally substituted cycloalkyl” is a cycloalkyl that is optionally 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 , —N(H)—C(O)—NH 2 , —N(H)—C(S)—NH 2 , —N(H)—S(O) 2 —NH 2 , —C(NH)—NH 2 , —O—R o , —S—R o , —O—C(O)—R o ,
  • C 3-6 cycloalkyl denotes cycloalkyl containing 3-6 carbon atoms.
  • C 3-5 cycloalkyl denotes cycloalkyl containing 3-5 carbon atoms. It is understood that substitutions are chemically feasible and attached at any available atom to provide a stable compound.
  • 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.
  • “Nitrogen containing heterocycloalkyl” refers to heterocycloalkyl wherein at least one heteroatom is N.
  • An “optionally substituted heterocycloalkyl” is a heterocycloalkyl that is optionally 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 , —N(H)—C(O)—NH 2 , —N(H)—C(S)—NH 2 , —N(H)—S(O) 2 —NH 2 , —C(NH)—NH 2 , —O—R o , —S—R o , —O—C(O)—R o ,
  • 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.
  • an “optionally substituted aryl” is an aryl that is optionally 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 , —N(H)—C(O)—NH 2 , —N(H)—C(S)—NH 2 , —N(H)—S(O) 2 —NH 2 , —C(NH)—NH 2 , —O—R o , —S—R o , —O—C(O)—R o , —O—C(S
  • 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.
  • Neitrogen containing heteroaryl refers to heteroaryl wherein at least one heteroatom is N. In some instances, for example when R groups of a nitrogen combine with the nitrogen to form a 5 or 7 membered nitrogen containing heteroaryl, any heteroatoms in such 5 or 7 membered heteroaryl are N.
  • An “optionally substituted heteroaryl” is a heteroaryl that is optionally 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 , —N(H)—C(O)—NH 2 , —N(H)—C(S)—NH 2 , —N(H)—S(O) 2 —NH 2 , —C(NH)—NH 2 , —O—R o , —S—R o , —O—C(O)—R o , —O—C(
  • R o , R p , R c , 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:
  • all occurrences of optionally substituted lower alkyl, optionally substituted lower alkenyl, optionally substituted C 3-6 alkenyl, optionally substituted lower alkynyl, or optionally substituted C 3-6 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, —O—R 1a , —S—R 1a , —N(R 1a )—R 1a , —O—C(O)—R 1a , —O—C(S)—R 1a , —C(O)—R 1a , —C(O)—R 1a , —C(S)—R 1a , —C(O)—O—R 1a , —C(S)—O—R 1a , —C(O)—N(R 1a )—R 1a , —C(S)—N
  • all occurrences of optionally substituted lower alkyl, optionally substituted lower alkenyl, optionally substituted C 3-6 alkenyl, optionally substituted lower alkynyl, or optionally substituted C 3-6 alkynyl are optionally substituted with one or more, also 1, 2 or 3 groups or substituents selected from the group consisting of fluoro, —CN, —O—R 1a , —S—R 1a , —N(R 1a )—R 1a , —C(O)—R 1a , —C(S)—R 1a , —C(O)—O—R 1a , —C(O)—N(R 1a )—R 1a , —C(S)—N(R 1a )—R 1a , —S(O) 2 —N(R 1a )—R 1a , —N(R 1a )—C(O)—R 1a , —N(R 1
  • “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, including descriptions of substituted cycloalkyl, heterocycloalkyl, aryl and heteroaryl, attached at any available atom to provide 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. It is understood that substitutions on alkoxy are chemically feasible and attached at any available atom to provide a stable compound.
  • 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, including descriptions of substituted cycloalkyl, heterocycloalkyl, aryl and heteroaryl, attached at any available atom to provide a stable compound.
  • 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. It is understood that substitutions on alkylthio are chemically feasible and attached at any available atom to provide a stable compound.
  • 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 cc , where R bb 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. It is understood that when mono-alkylamino, di-alkylamino, or cycloalkylamino are substituents on other moieties, these are chemically feasible and attached at any available atom to provide a stable compound.
  • 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.
  • inhibitor refers to a modulator that decreases the activity of the target biomolecule.
  • 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.
  • 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 i.e. selectivity
  • 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, 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 Ab1, 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, L
  • 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. Specific target protein kinases contemplated by the present invention are described in the art, including, without limitation, protein kinases 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, including all specifications, figures, and tables, and for all purposes, as well as 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), Cl (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 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).
  • 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
  • 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
  • invention compounds may exist in a number of different forms or derivatives, all within the scope of the present invention.
  • Alternative forms or derivatives include, for example, (a) prodrugs, and active metabolites (b) tautomers, isomers (including stereoisomers and regioisomers), and racemic mixtures (c) pharmaceutically acceptable salts and formulations and (d) solid forms, including different crystal forms, polymorphic or amorphous solids, including hydrates and solvates thereof, and other forms.
  • the invention also includes prodrugs (generally pharmaceutically acceptable prodrugs), active metabolic derivatives (active metabolites), and their pharmaceutically acceptable salts.
  • Prodrugs are compounds or pharmaceutically acceptable salts thereof which, when metabolized under physiological conditions or when converted by solvolysis, yield the desired active compound.
  • Prodrugs include, without limitation, esters, amides, carbamates, carbonates, ureides, solvates, or hydrates of the active compound.
  • the prodrug is inactive, or less active than the active compound, but may provide one or more advantageous handling, administration, and/or metabolic properties.
  • some prodrugs are esters of the active compound; during metabolysis, the ester group is cleaved to yield the active drug.
  • Esters include, for example, esters of a carboxylic acid group, or S-acyl or O-acyl derivatives of thiol, alcohol, or phenol groups.
  • prodrugs are activated enzymatically to yield the active compound, or a compound which, upon further chemical reaction, yields the active compound.
  • Prodrugs may proceed from prodrug form to active form in a single step or may have one or more intermediate forms which may themselves have activity or may be inactive.
  • bioprecursor prodrugs can be conceptually divided into two non-exclusive categories, bioprecursor prodrugs and carrier prodrugs.
  • bioprecursor prodrugs are compounds that are inactive or have low activity compared to the corresponding active drug compound, that contain one or more protective groups and are converted to an active form by metabolism or solvolysis. Both the active drug form and any released metabolic products should have acceptably low toxicity.
  • the formation of active drug compound involves a metabolic process or reaction that is one of the follow types:
  • Oxidative reactions are exemplified without limitation to reactions such as oxidation of alcohol, carbonyl, and acid functionalities, hydroxylation of aliphatic carbons, hydroxylation of alicyclic carbon atoms, oxidation of aromatic carbon atoms, oxidation of carbon-carbon double bonds, oxidation of nitrogen-containing functional groups, oxidation of silicon, phosphorus, arsenic, and sulfur, oxidative N-dealkylation, oxidative O- and S-dealkylation, oxidative deamination, as well as other oxidative reactions.
  • Reductive reactions are exemplified without limitation to reactions such as reduction of carbonyl functionalitites, reduction of alcohol functionalities and carbon-carbon double bonds, reduction of nitrogen-containing functional groups, and other reduction reactions.
  • Reactions without change in the state of oxidation are exemplified without limitation to reactions such as hydrolysis of esters and ethers, hydrolytic cleavage of carbon-nitrogen single bonds, hydrolytic cleavage of non-aromatic heterocycles, hydration and dehydration at multiple bonds, new atomic linkages resulting from dehydration reactions, hydrolytic dehalogenation, removal of hydrogen halide molecule, and other such reactions.
  • Carrier prodrugs are drug compounds that contain a transport moiety, e.g., that improves uptake and/or localized delivery to a site(s) of action.
  • a transport moiety e.g., that improves uptake and/or localized delivery to a site(s) of action.
  • the linkage between the drug moiety and the transport moiety is a covalent bond
  • the prodrug is inactive or less active than the drug compound
  • the prodrug and any release transport moiety are acceptably non-toxic.
  • the transport moiety is intended to enhance uptake
  • the release of the transport moiety should be rapid.
  • it is desirable to utilize a moiety that provides slow release e.g., certain polymers or other moieties, such as cyclodextrins.
  • carrier prodrugs are often advantageous for orally administered drugs.
  • the transport moiety provides targeted delivery of the drug, for example the drug maybe conjugated to an antibody or antibody fragment.
  • Carrier prodrugs can, for example, be used to improve one or more of the following properties: increased lipophilicity, increased duration of pharmacological effects, increased site-specificity, decreased toxicity and adverse reactions, and/or improvement in drug formulation (e.g., stability, water solubility, suppression of an undesirable organoleptic or physiochemical property).
  • lipophilicity can be increased by esterification of hydroxyl groups with lipophilic carboxylic acids, or of carboxylic acid groups with alcohols, e.g., aliphatic alcohols. Wermuth, supra.
  • Metabolites e.g., active metabolites
  • prodrugs as described above, e.g., bioprecursor prodrugs.
  • metabolites are pharmacologically active compounds or compounds that further metabolize to pharmacologically active compounds that are derivatives resulting from metabolic processes in the body of a subject.
  • active metabolites are such pharmacologically active derivative compounds.
  • the prodrug compound is generally inactive or of lower activity than the metabolic product.
  • the parent compound may be either an active compound or may be an inactive prodrug.
  • one or more alkoxy groups can be metabolized to hydroxyl groups while retaining pharmacologic activity and/or carboxyl groups can be esterified, e.g., glucuronidation.
  • carboxyl groups can be esterified, e.g., glucuronidation.
  • there can be more than one metabolite where an intermediate metabolite(s) is further metabolized to provide an active metabolite.
  • a derivative compound resulting from metabolic glucuronidation may be inactive or of low activity, and can be further metabolized to provide an active metabolite.
  • Metabolites of a compound may be identified using routine techniques known in the art, and their activities determined using tests such as those described herein. See, e.g., Bertolini et al., 1997 , J. Med. Chem., 40:2011-2016; Shan et al., 1997 , J Pharm Sci 86(7):756-757; Bagshawe, 1995 , Drug Dev. Res., 34:220-230; Wermuth, supra.
  • some of the compounds according to the present invention may exist as stereoisomers, i.e. having the same atomic connectivity of covalently bonded atoms yet differing in the spatial orientation of the atoms.
  • compounds may be optical stereoisomers, which contain one or more chiral centers, and therefore, may exist in two or more stereoisomeric forms (e.g. enantiomers or diastereomers).
  • stereoisomers i.e., essentially free of other stereoisomers
  • racemates i.e., essentially free of other stereoisomers
  • stereoisomers include geometric isomers, such as cis- or trans-orientation of substituents on adjacent carbons of a double bond. All such single stereoisomers, racemates and mixtures thereof are intended to be within the scope of the present invention. Unless specified to the contrary, all such steroisomeric forms are included within the formulae provided herein.
  • a chiral compound of the present invention is in a form that contains at least 80% of a single isomer (60% enantiomeric excess (“e.e.”) or diastereomeric excess (“d.e.”)), or at least 85% (70% e.e. or d.e.), 90% (80% e.e. or d.e.), 95% (90% e.e. or d.e.), 97.5% (95% e.e. or d.e.), or 99% (98% e.e. or d.e.).
  • 60% enantiomeric excess (“e.e.”) or diastereomeric excess (“d.e.”) or at least 85% (70% e.e. or d.e.), 90% (80% e.e. or d.e.), 95% (90% e.e. or d.e.), 97.5% (95% e.e. or d.e.), or 99% (98% e.e. or d.e
  • an optically pure compound having one chiral center is one that consists essentially of one of the two possible enantiomers (i.e., is enantiomerically pure), and an optically pure compound having more than one chiral center is one that is both diastereomerically pure and enantiomerically pure.
  • the compound is present in optically pure form, such optically pure form being prepared and/or isolated by methods known in the art (e.g. by recrystallization techniques, chiral synthetic techniques (including synthesis from optically pure starting materials), and chromatographic separation using a chiral column.
  • compounds of Formula I can be in the form of pharmaceutically acceptable salts, or can be formulated as pharmaceutically acceptable salts.
  • Contemplated pharmaceutically acceptable salt forms include, without limitation, mono, bis, tris, tetrakis, and so on.
  • Pharmaceutically acceptable salts are non-toxic in the amounts and concentrations at which they are administered. The preparation of such salts can facilitate the pharmacological use by altering the physical characteristics of a compound without preventing it from exerting its physiological effect. Useful alterations in physical properties include lowering the melting point to facilitate transmucosal administration and increasing the solubility to facilitate administering higher concentrations of the drug.
  • a compound of the invention may possess a sufficiently acidic, a sufficiently basic, or both functional groups, and accordingly react with any of a number of inorganic or organic bases, and inorganic and organic acids, to form a pharmaceutically acceptable salt.
  • Pharmaceutically acceptable salts include acid addition salts such as those containing chloride, bromide, iodide, hydrochloride, acetate, phenylacetate, acrylate, ascorbate, aspartate, benzoate, 2-phenoxybenzoate, 2-acetoxybenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, methylbenzoate, bicarbonate, butyne-1,4 dioate, hexyne-1,6-dioate, caproate, caprylate, chlorobenzoate, cinnamate, citrate, decanoate, formate, fumarate, glycolate, gluconate, glucarate, glucuronate, glucose-6-phosphate, glutamate, heptanoate, hexanoate, isethionate, isobutyrate, gamma-hydroxybutyrate, phenylbutyrate, lactate, malate, maleate, hydroxymaleate, methyl
  • pharmaceutically acceptable salts also include basic addition salts such as those containing benzathine, chloroprocaine, choline, ethanolamine, diethanolamine, triethanolamine, t-butylamine, dicyclohexylamine, ethylenediamine, N,N′-dibenzylethylenediamine, meglumine, hydroxyethylpyrrolidine, piperidine, morpholine, piperazine, procaine, aluminum, calcium, copper, iron, lithium, magnesium, manganese, potassium, sodium, zinc, ammonium, and mono-, di-, or tri-alkylamines, or salts derived from amino acids such as L-histidine, L-glycine, L-lysine, and L-arginine.
  • bases addition salts such as those containing benzathine, chloroprocaine, choline, ethanolamine, diethanolamine, triethanolamine, t-butylamine, dicyclohexylamine, ethylenediamine, N,
  • salts can be prepared by standard techniques.
  • the free-base form of a compound can be dissolved in a suitable solvent, such as an aqueous or aqueous-alcohol solution containing the appropriate acid and then isolated by evaporating the solution.
  • a salt can be prepared by reacting the free base and acid in an organic solvent. If the particular compound is an acid, the desired pharmaceutically acceptable salt may be prepared by any suitable method, for example, treatment of the free acid with an appropriate inorganic or organic base.
  • the pharmaceutically acceptable salt of the different compounds may be present as a complex.
  • complexes include 8-chlorotheophylline complex (analogous to, e.g., dimenhydrinate: diphenhydramine 8-chlorotheophylline (1:1) complex; Dramamine) and various cyclodextrin inclusion complexes.
  • the compounds and salts may exist in different crystal or polymorphic forms, or may be formulated as co-crystals, or may be in an amorphous form, or may be any combination thereof (e.g. partially crystalline, partially amorphous, or mixtures of polymorphs) all of which are intended to be within the scope of the present invention and specified formulae.
  • salts are formed by acid/base addition, i.e.
  • co-crystals are a new chemical species that is formed between neutral compounds, resulting in the compound and an additional molecular species in the same crystal structure.
  • the formulae are intended to cover hydrated or solvated as well as unhydrated or unsolvated forms of the identified structures.
  • the indicated structures include both hydrated and non-hydrated forms.
  • Other examples of solvates include the structures in combination with a suitable solvent, such as isopropanol, ethanol, methanol, dimethylsulfoxide, ethyl acetate, acetic acid, or ethanolamine.
  • Compounds of Formula I can be administered by different routes, including injection (i.e. parenteral, including intravenous, intraperitoneal, subcutaneous, and intramuscular), oral, transdermal, transmucosal, rectal, or inhalant. Such dosage forms should allow the compound to reach target cells. Other factors are well known in the art, and include considerations such as toxicity and dosage forms that retard the compound or composition from exerting its effects. Techniques and formulations generally may be found in Remington: The Science and Practice of Pharmacy, 21 st edition, Lippincott, Williams and Wilkins, Philadelphia, Pa., 2005 (hereby incorporated by reference herein).
  • compositions will comprise carriers or excipients, which may be chosen to facilitate administration of the compound by a particular route.
  • carriers include calcium carbonate, calcium phosphate, various sugars such as lactose, glucose, or sucrose, types of starch, cellulose derivatives, gelatin, lipids, liposomes, nanoparticles, and the like.
  • Carriers also include physiologically compatible liquids as solvents or for suspensions, including, for example, sterile solutions of water for injection (WFI), saline solution, dextrose solution, Hank's solution, Ringer's solution, vegetable oils, mineral oils, animal oils, polyethylene glycols, liquid paraffin, and the like.
  • WFI water for injection
  • oral administration may be used.
  • Pharmaceutical preparations for oral use can be formulated into conventional oral dosage forms such as capsules, tablets, and liquid preparations such as syrups, elixirs, and concentrated drops.
  • Compounds of Formula I may be combined with solid excipients, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain, for example, tablets, coated tablets, hard capsules, soft capsules, solutions (e.g. aqueous, alcoholic, or oily solutions) and the like.
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, glucose, sucrose, mannitol, or sorbitol; cellulose preparations, for example, corn starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose (CMC), and/or polyvinylpyrrolidone (PVP: povidone); oily excipients, including vegetable and animal oils, such as sunflower oil, olive oil, or codliver oil.
  • fillers such as sugars, including lactose, glucose, sucrose, mannitol, or sorbitol
  • cellulose preparations for example, corn starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose (CMC), and/or polyvinylpyrrolidone (PVP: povid
  • the oral dosage formulations may also contain disintegrating agents, such as the cross-linked polyvinylpyrrolidone, agar, or alginic acid, or a salt thereof such as sodium alginate; a lubricant, such as talc or magnesium stearate; a plasticizer, such as glycerol or sorbitol; a sweetening such as sucrose, fructose, lactose, or aspartame; a natural or artificial flavoring agent, such as peppermint, oil of wintergreen, or cherry flavoring; or dye-stuffs or pigments, which may be used for identification or characterization of different doses or combinations. Also provided are dragee cores with suitable coatings.
  • concentrated sugar solutions may be used, which may optionally contain, for example, gum arabic, talc, poly-vinylpyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • compositions that can be used orally include push-fit capsules made of gelatin (“gelcaps”), as well as soft, sealed capsules made of gelatin, and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • injection parenteral administration
  • Compounds of Formula I for injection may be formulated in sterile liquid solutions, preferably in physiologically compatible buffers or solutions, such as saline solution, Hank's solution, or Ringer's solution.
  • Dispersions may also be prepared in non-aqueous solutions, such as glycerol, propylene glycol, ethanol, liquid polyethylene glycols, triacetin, and vegetable oils. Solutions may also contain a preservative, such as methylparaben, propylparaben, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • the compounds may be formulated in solid form, including, for example, lyophilized forms, and redissolved or suspended prior to use.
  • transmucosal, topical or transdermal administration may be used.
  • penetrants appropriate to the barrier to be permeated are used.
  • penetrants are generally known in the art, and include, for example, for transmucosal administration, bile salts and fusidic acid derivatives.
  • detergents may be used to facilitate permeation.
  • Transmucosal administration for example, may be through nasal sprays or suppositories (rectal or vaginal).
  • Compositions of compounds of Formula I for topical administration may be formulated as oils, creams, lotions, ointments, and the like by choice of appropriate carriers known in the art.
  • Suitable carriers include vegetable or mineral oils, white petrolatum (white soft paraffin), branched chain fats or oils, animal fats and high molecular weight alcohol (greater than C 12 ). In some embodiments, carriers are selected such that the active ingredient is soluble. Emulsifiers, stabilizers, humectants and antioxidants may also be included as well as agents imparting color or fragrance, if desired. Creams for topical application are preferably formulated from a mixture of mineral oil, self-emulsifying beeswax and water in which mixture the active ingredient, dissolved in a small amount of solvent (e.g., an oil), is admixed.
  • solvent e.g., an oil
  • administration by transdermal means may comprise a transdermal patch or dressing such as a bandage impregnated with an active ingredient and optionally one or more carriers or diluents known in the art.
  • a transdermal patch or dressing such as a bandage impregnated with an active ingredient and optionally one or more carriers or diluents known in the art.
  • the dosage administration will be continuous rather than intermittent throughout the dosage regimen.
  • compounds are administered as inhalants.
  • Compounds of Formula I may be formulated as dry powder or a suitable solution, suspension, or aerosol.
  • Powders and solutions may be formulated with suitable additives known in the art.
  • powders may include a suitable powder base such as lactose or starch, and solutions may comprise propylene glycol, sterile water, ethanol, sodium chloride and other additives, such as acid, alkali and buffer salts.
  • Such solutions or suspensions may be administered by inhaling via spray, pump, atomizer, or nebulizer, and the like.
  • the compounds of Formula I may also be used in combination with other inhaled therapies, for example corticosteroids such as fluticasone proprionate, beclomethasone dipropionate, triamcinolone acetonide, budesonide, and mometasone furoate; beta agonists such as albuterol, salmeterol, and formoterol; anticholinergic agents such as ipratroprium bromide or tiotropium; vasodilators such as treprostinal and iloprost; enzymes such as DNAase; therapeutic proteins; immunoglobulin antibodies; an oligonucleotide, such as single or double stranded DNA or RNA, siRNA; antibiotics such as tobramycin; muscarinic receptor antagonists; leukotriene antagonists; cytokine antagonists; protease inhibitors; cromolyn sodium; nedocril sodium; and sodium cromoglycate.
  • corticosteroids such as
  • the amounts of various compounds to be administered can be determined by standard procedures taking into account factors such as the compound activity (in vitro, e.g. the compound IC 50 vs. target, or in vivo activity in animal efficacy models), pharmacokinetic results in animal models (e.g. biological half-life or bioavailability), the age, size, and weight of the subject, and the disorder associated with the subject. The importance of these and other factors are well known to those of ordinary skill in the art. Generally, a dose will be in the range of about 0.01 to 50 mg/kg, also about 0.1 to 20 mg/kg of the subject being treated. Multiple doses may be used.
  • the compounds of Formula I may also be used in combination with other therapies for treating the same disease.
  • Such combination use includes administration of the compounds and one or more other therapeutics at different times, or co-administration of the compound and one or more other therapies.
  • dosage may be modified for one or more of the compounds of the invention or other therapeutics used in combination, e.g., reduction in the amount dosed relative to a compound or therapy used alone, by methods well known to those of ordinary skill in the art.
  • use in combination includes use with other therapies, drugs, medical procedures etc., where the other therapy or procedure may be administered at different times (e.g. within a short time, such as within hours (e.g. 1, 2, 3, 4-24 hours), or within a longer time (e.g. 1-2 days, 2-4 days, 4-7 days, 1-4 weeks)) than a compound of Formula I, or at the same time as a compound of Formula I.
  • Use in combination also includes use with a therapy or medical procedure that is administered once or infrequently, such as surgery, along with a compound of Formula I administered within a short time or longer time before or after the other therapy or procedure.
  • the present invention provides for delivery of a compound of Formula I and one or more other drug therapeutics delivered by a different route of administration or by the same route of administration.
  • the use in combination for any route of administration includes delivery of a compound of Formula I and one or more other drug therapeutics delivered by the same route of administration together in any formulation, including formulations where the two compounds are chemically linked in such a way that they maintain their therapeutic activity when administered.
  • the other drug therapy may be co-administered with a compound of Formula I.
  • Use in combination by co-administration includes administration of co-formulations or formulations of chemically joined compounds, or administration of two or more compounds in separate formulations within a short time of each other (e.g.
  • Co-administration of separate formulations includes co-administration by delivery via one device, for example the same inhalant device, the same syringe, etc., or administration from separate devices within a short time of each other.
  • Co-formulations of a compound of Formula I and one or more additional drug therapies delivered by the same route includes preparation of the materials together such that they can be administered by one device, including the separate compounds combined in one formulation, or compounds that are modified such that they are chemically joined, yet still maintain their biological activity.
  • Such chemically joined compounds may have a linkage that is substantially maintained in vivo, or the linkage may break down in vivo, separating the two active components.
  • 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.
  • Compound 1 (R 2 and R 4 as defined in paragraph [0004]) is dissolved in an anhydrous solvent (e.g. tetrahydrofuran) under nitrogen atmosphere.
  • anhydrous solvent e.g. tetrahydrofuran
  • the solution is cooled down with the aid of a dry ice and acetone bath.
  • a base e.g. n-butyllithium
  • 1, 2-bis(chlorodimethylsilyl)ethane at low temperature (typically below ⁇ 70° C.).
  • the resulting mixture is stirred at low temperature for 1-2 hours.
  • a base e.g. n-butyllithium
  • ethyl chloroformate e.g. n-butyllithium
  • the resulting mixture is allowed to warm to room temperature and then stirred at room temperature for 1-3 days.
  • the reaction mixture is quenched by an acid solution, stirred at room temperature for a couple of hours, and then basified.
  • the mixture is extracted with an organic solvent (e.g. dichloromethane or ethyl acetate).
  • the desired compound 2 is purified by chromatography.
  • a base e.g. lithium hydroxide or sodium hydroxide
  • the resulting suspension is stirred in a heated oil bath for over 10 hours.
  • the reaction mixture is cooled down to room temperature and then acidified with an acid solution such as concentrated hydrochloric acid.
  • the aqueous layer is separated and extracted with an appropriate organic solvent (e.g. ethyl acetate).
  • the desired compound 4 is purified by chromatography.
  • Compound 7 (R 2 as defined in paragraph [0005]) is dissolved in an appropriate solvent (e.g. methanol). To this solution is added catalyst (e.g. palladium on carbon). The suspension is then placed under a hydrogen atmosphere and shaken at room temperature for over 12 hours. The catalyst is removed by filtration on a pad of celite and washed with an appropriate solvent (e.g. methanol). The filtrate is concentrated under reduced pressure to give compound 8, which is used in the next step without further purification.
  • an appropriate solvent e.g. methanol
  • a base e.g. sodium hydride
  • an appropriate alkylating agent e.g. halide
  • the reaction mixture is stirred at room temperature or heated in an oil bath as necessary, for a few hours.
  • the reaction mixture is then poured into water.
  • the organic layer is collected and the aqueous layer is extracted with an appropriate organic solvent (e.g. ethyl acetate or dichloromethane).
  • the organic solvents are then combined.
  • the desired compound 10 (R 4 as defined in paragraph [0005]) is purified by chromatography.
  • a mixture of compound 10, an appropriate boronic acid 11 (Ar, m and R 1 as defined in paragraph [0004]), and a catalyst (e.g. tetrakis(triphenylphosphine)palladium) in a mixture of base (e.g. aqueous solution of potassium carbonate) and an appropriate organic solvent (e.g. acetonitrile) is heated in an oil bath or is irradiated in a microwave system at over 100° C. for an appropriate time depending on starting materials.
  • the reaction mixture is poured into water and then extracted with an appropriate organic solvent (e.g. dichloromethane or ethyl acetate). The organic solvents are then combined.
  • the desired compound of Formula Ic (L 2 is S(O) 2 ) or Id is purified by chromatography.
  • Compound 13 (LG is a suitable leaving group) is prepared by converting compound 12 into a mesylate or triflate by reacting with the corresponding sulfonyl chloride in an appropriate organic solvent. It can also be converted into the corresponding bromide by reacting with an appropriate agent (e.g. phosphorous tribromide) in the presence of an appropriate base (e.g. pyridine).
  • an appropriate agent e.g. phosphorous tribromide
  • an appropriate base e.g. pyridine
  • Step 3 Synthesis of Compound of Formula I where L 1 is —CH 2 NR 11 —
  • amine 6 is added to a mixture of compound 13 and a base (e.g. cesium carbonate) in an appropriate organic solvent (e.g. acetonitrile).
  • an appropriate organic solvent e.g. acetonitrile
  • the reaction mixture is stirred at room temperature, or heated in an oil bath if necessary, for 2-24 hours.
  • the reaction mixture is poured into water and then extracted with an appropriate organic solvent (e.g. dichloromethane or ethyl acetate). The organic solvents are then combined.
  • the desired compound of Formula I is purified by chromatography.
  • Step 3 Synthesis of Compound of Formula I where L 1 is —CH 2 NR 11 —
  • amine 6 Ar, m, R 1 and R 11 as defined in paragraph [0005]
  • reducing agent e.g. triethylsilane and trifluoroacetic acid
  • the reaction mixture is heated in an oil bath for 2-24 hours.
  • the reaction mixture is concentrated, poured into water and then extracted with an appropriate organic solvent (e.g. dichloromethane or ethyl acetate).
  • an appropriate organic solvent e.g. dichloromethane or ethyl acetate.
  • the organic layers are then combined.
  • the desired compound of Formula I is purified by chromatography.
  • compounds of Formula I where L 1 is —CH 2 NR 11 — may be prepared by reduction of compounds of Formula Ib or Id wherein A is —C(O)— (e.g. prepared as described in Example 1) with an appropriate reducing agent (e.g. borane or diisobutylaluminum hydride).
  • an appropriate reducing agent e.g. borane or diisobutylaluminum hydride.
  • 1,2-Bis(chlorodimethylsilyl)ethane (24.80 g) was dissolved in 80 mL of anhydrous tetrahydrofuran and slowly added dropwise to the reaction mixture while maintaining the temperature below ⁇ 70° C.
  • the resulting mixture was stirred at ⁇ 78° C. for 1 hour, then n-butyllithium (2.5 M, 45 mL) was slowly added dropwise maintaining the temperature below ⁇ 70° C.
  • the mixture was then stirred for 30 minutes at ⁇ 78° C., then warmed up to 15° C. over 1 hour.
  • the reaction mixture was cooled down to ⁇ 78° C.
  • n-butyllithium 2.5 M, 50 mL was slowly added dropwise maintaining the temperature below ⁇ 70° C.
  • the mixture was stirred at ⁇ 70° C. for 90 minutes, then 13.40 mL of ethylchloroformate was slowly added dropwise maintaining the temperature below ⁇ 70° C.
  • the reaction mixture was slowly warmed to room temperature and stirred at room temperature for 64 hours.
  • the reaction was quenched by careful addition of a solution of 50 mL of concentrated hydrochloric acid in 160 mL of water while cooling with an ice/water bath.
  • the mixture was stirred at room temperature for 2 hours, then made basic by addition of potassium carbonate.
  • the mixture was extracted with 3 ⁇ 100 mL of ethyl acetate and the combined organic extracts were washed with 50 mL of brine and dried with magnesium sulfate. After removal of the solvent, the residue was purified with silica gel column chromatography eluting with ethyl acetate in hexane to provide the desired compound (15, 17 g, 72%).
  • 3-Amino-6-chloro-2-fluorobenzoic acid ethyl ester (15, 17 g) was dissolved in 785 mL of dichloromethane, to which 13.2 mL of pyridine was added, followed by propane-1-sulfonylchloride (16, 12.8 g). The reaction mixture was stirred at room temperature for 18 hours, then poured into 400 mL of water. The organic layer was separated and the aqueous layer was extracted with 200 mL of dichlormethane. The combined organic extracts were dried over magnesium sulfate to give an orange oil (41 g). Trituration in 150 mL of diethylether removed the pyridine salt as a white solid.
  • 6-Chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoic acid 18 was suspended in anhydrous dichloromethane (30 mL/g). Dimethylformamide (2 drops) was added and the suspension cooled in an ice/water bath. Oxalyl chloride (5 eq) was slowly added dropwise. The bath was then removed and the reaction mixture stirred at room temperature for 2 to 3 hours and the solids slowly disappeared. Dichloromethane and excess oxalyl chloride were removed under reduced pressure and the residue was used without further purification in the next step.
  • 6-Bromo-pyridin-3-ylamine (20, 3.16 g, 18.26 mmol) was dissolved in 55 mL of anhydrous tetrahydrofuran. Triethylamine (1.85 g, 2.55 mL, 18.26 mmol) was added and the mixture cooled in an ice/salt bath. A solution of 6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoyl chloride (19, 3.8 g, 12.17 mmol) in 55 mL of anhydrous tetrahydrofuran was slowly added dropwise to the reaction mixture.
  • Step 6 Preparation 6-chloro-2-fluoro-N-[6-(5-methyl-thiazol-2-ylamino)-pyridin-3-yl]-3-(propane-1-sulfonylamino)-benzamide (P-0011)
  • N-(6-bromo-pyridin-3-yl)-6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzamide (21, 250 mg, 0.555 mmol) was placed in a microwave vial along with palladium acetate (12.5 mg, 0.055 mmol), BINAP (69 mg, 0.111 mmol), potassium tert-butoxide (124 mg, 1.11 mmol) and 2-amino-5-methyl-thiazole (22, 190 mg, 1.66 mmol). Dimethylformamide (2.5 mL) was added and the vial sealed. The mixture was then heated at 150° C. in a microwave for 3 hours.
  • 2-Bromo-5-nitro-pyridine (23, 400 mg) was placed in a microwave vial. Isopropylamine (24, 3 mL) was added and the vial sealed. The mixture was then heated at 120° C. for 30 minutes using a Biotage Initiator EXP microwave. The crude mixture was then absorbed on a column and purified by silica gel chromatography. The fractions containing the desired compound were combined and concentrated to provide the desired compound as a yellow solid.
  • the 6-isopropylamino-3-nitro pyridine 25 was dissolved in methanol (35 mL/g). Palladium on carbon catalyst (10%, wet, ⁇ 100 mg) was added and the suspension was placed under a hydrogen atmosphere at room temperature overnight ( ⁇ 18 hours). The catalyst was removed by filtration on a pad of celite and washed with 2 ⁇ 10 mL of methanol. The filtrate was concentrated under reduced pressure to provide the desired compound, which was used without further purification in the next step.
  • Step 3 Preparation of 6-chloro-2-fluoro-N-(6-isopropylamino-pyridin-3-yl)-3-(propane-1-sulfonylamino)-benzamide (P-0005)
  • N*2*-isopropyl-pyridine-2,5-diamine (26, 155 mg, 1.01 mmol) was dissolved in 4 mL of anhydrous tetrahydrofuran. Triethylamine (103 mg, 142 ⁇ L, 1.01 mmol) was added and the mixture cooled in an ice/salt bath. 6-Chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoyl chloride (19, 200 mg, 0.68 mmol) in 4 mL of anhydrous tetrahydrofuran was then slowly added dropwise.
  • N-(2-Acetylamino-pyrimidin-5-yl)-6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzamide P-0004 was synthesized in three steps from 2-amino-5-nitropyrimidine 27 as shown in Scheme 6.
  • 2-Amino-5-nitropyrimidine (27, 500 mg) was suspended in 5 mL of acetic anhydride. The mixture was heated at 160° C. for two hours, then cooled to room temperature. The solids were filtered and washed with 5 mL of water, then suspended in 10 mL of water and the pH was brought to 8-9 by addition of 25% ammonium hydroxide solution. The solids were filtered, washed with 2 ⁇ 10 mL cold water and recrystallized from ethyl acetate to provide the desired compound as beige needles (28, 382 mg, 58%).
  • N-(5—Nitro-pyrimidin-2-yl)-acetamide (28, 620 mg) was suspended in 31 mL of methanol.
  • Palladium on carbon catalyst (10%, wet, 60 mg) was added and the suspension was placed under hydrogen atmosphere for 17 hours.
  • the catalyst was filtered off on a pad of celite and washed with 2 ⁇ 30 mL of methanol. The filtrate was concentrated under reduced pressure to provide the desired compound as pale yellow needles (29, 520 mg, 100%).
  • N-(5-amino-pyrimidin-2-yl)-acetamide (29, 154 mg, 1.01 mmol) was dissolved in 3 mL of anhydrous tetrahydrofuran. Triethylamine (103 mg, 142 ⁇ L, 1.01 mmol) was added and the mixture cooled in an ice/salt bath. 6-Chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoyl chloride (19, 200 mg, 0.68 mmol) in 3 mL of anhydrous tetrahydrofuran was then slowly added dropwise.
  • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(propane-1-sulfonylamino)-benzamide P-0008 was synthesized in three steps from 2,6-difluoro-3-nitro-benzoic acid 30 and N-(5-amino-pyridin-2-yl)-acetamide 31 as shown in Scheme 7.
  • 2,6-difluoro-3-nitro-benzoic acid (30, 500 mg) was dissolved in 15 mL of anhydrous dichloromethane. N,N-Dimethylformamide (1 drop) was then added and the mixture cooled to 5° C. in an ice/water bath. Oxalyl chloride (1.1 mL, 5 eq) was slowly added dropwise.
  • reaction mixture was stirred at room temperature for two hours, then concentrated under reduced pressure to give a yellow solid residue, which was dissolved in 5 mL of anhydrous tetrahydrofuran and slowly added dropwise to a solution of N-(5-amino-pyridin-2-yl)-acetamide (31, 558 mg, 1.5 eq) and triethylamine (0.52 mL) in 10 mL of anhydrous tetrahydrofuran. The resulting suspension was stirred at room temperature overnight.
  • the mixture was diluted with 50 mL of ethyl acetate, washed with 2 ⁇ 25 mL of water, then 25 mL brine, dried over magnesium sulfate and concentrated to provide the crude desired compound as a brown solid (32, 980 mg, 84%), which was used in the next step without further purification.
  • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-nitro-benzamide (32, 950 mg) was suspended in 15 mL of methanol. Palladium on carbon catalyst (10%, wet, 100 mg) was added and the suspension was placed under hydrogen atmosphere for 17 hours. The catalyst was filtered off on a pad of celite and washed with 2 ⁇ 20 mL of methanol. The filtrate was concentrated under reduced pressure to provide the desired compound as a black solid (33, 750 mg, 86%).
  • Step 3 Preparation of N-(6-acetylamino-pyridin-3-yl)-2,6-difluoro-3-(propane-1-sulfonylamino)-benzamide (P-0008)
  • Example 8 Synthesis of pyrrolidine-1-carboxylic acid ⁇ 5-[6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoylamino]-pyridin-2-yl ⁇ -amide P-0006
  • 2-Amino-5-nitro-pyridine 34, 500 mg was dissolved in 5 mL of anhydrous tetrahydrofuran.
  • N-Methyl morpholine (436 mg, 1.2 eq) was added and the mixture cooled to ⁇ 10° C. in an ice/salt acetone bath.
  • a solution of isopropenyl chloroformate (520 mg) in 5 mL of tetrahydrofuran was then slowly added dropwise while maintaining the temperature below ⁇ 10° C. The reaction mixture was stirred at room temperature overnight, then diluted with 25 mL of ethyl acetate and 20 mL of water.
  • the aqueous layer was separated and extracted with 2 ⁇ 25 mL of ethyl acetate.
  • the combined organic extracts were washed with 25 mL of half saturated brine and dried over magnesium sulfate. After removal of the solvent, the residue was purified with silica gel column chromatography eluting with ethyl acetate in hexane to provide the desired compound as a white solid (35, 0.52 g, 65%).
  • Pyrrolidine-1-carboxylic acid (5-nitro-pyridin-2-yl)-amide (37, 115 mg) was dissolved in 10 mL of methanol. Palladium on carbon catalyst was added and the suspension placed under a hydrogen atmosphere for 64 hours. The catalyst was filtered over a pad of celite and washed with 2 ⁇ 10 mL of methanol. The filtrate was concentrated under reduced pressure to provide the desired compound as a grey solid (38, 0.1 g, 100%).
  • Step 4 Preparation of pyrrolidine-1-carboxylic acid ⁇ 5-[6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoylamino]-pyridin-2-yl ⁇ -amide (P-0006)
  • Pyrrolidine-1-carboxylic acid (5-amino-pyridin-2-yl)-amide (38, 100 mg, 0.48 mmol) was dissolved in 2 mL of anhydrous tetrahydrofuran.
  • Triethylamine 49 mg, 67 ⁇ L, 0.48 mmol
  • 6-Chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoyl chloride (19, 100 mg, 0.34 mmol) in 1 mL of anhydrous tetrahydrofuran was then slowly added dropwise.
  • 3-Aminoquinoline (39, 172 mg, 1.19 mmol) was dissolved in 5 mL of anhydrous tetrahydrofuran. Triethylamine (120 mg, 170 ⁇ L, 1.19 mmol) was added and the mixture cooled in an ice/salt bath. 6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoyl chloride (19, 250 mg, 0.80 mmol) in 5 mL of anhydrous tetrahydrofuran was slowly added dropwise.
  • Step 3 Preparation of propane-1-sulfonic acid ⁇ 2,4-difluoro-3-[(5-methyl-isoxazol-3-ylamino)-methyl]-phenyl ⁇ -amide (P-0019)
  • Example 12 Synthesis of quinoline-3-carboxylic acid [2,6-difluoro-3-(propane-1-sulfonylamino)-phenyl]-amide P-0024 and propane-1-sulfonic acid ⁇ 2,4-difluoro-3-[(quinolin-3-ylmethyl)-amino]-phenyl ⁇ -amide P-0025
  • Step 3a Preparation of quinoline-3-carboxylic acid [2,6-difluoro-3-(propane-1-sulfonylamino)-phenyl]-amide (P-0024)
  • Step 3b Preparation of propane-1-sulfonic acid ⁇ 2,4-difluoro-3-[(quinolin-3-ylmethyl)-amino]-phenyl ⁇ -amide (P-0025)
  • Additional compounds may be prepared following the protocol of Scheme 13, optionally replacing propane-1-sulfonic acid (2,4-difluoro-3-formyl-phenyl)-amide 43 with N-(2,4-difluoro-3-formyl-phenyl)-4-trifluoromethyl-benzenesulfonamide (prepared following the protocol of Scheme 11, Example 11, steps 1 and 2 using 4-trifluoromethyl-benzenesulfonyl chloride in Step 1 in place of propane-1-sulfonyl chloride 16) in Step 1 and 3-hydroxyquinoline 51 with a suitable alcohol in Step 3.
  • the following compounds may be prepared by this method:
  • Additional compounds may be synthesized in six steps according to the following Scheme 15 or in four steps according the following Scheme 16.
  • the purified Boc-protected amine is dissolved in dichloromethane, trifluoroacetic acid is added and the reaction stirred at room temperature, monitoring by TLC for the disappearance of starting material.
  • the completed reaction is neutralised by pouring into a cooled saturated solution of sodium hydrogen carbonate and then extracted 3 ⁇ into dichloromethane. The combined organic extracts are washed with brine, dried over magnesium sulfate, filtered and concentrated in vacuo to provide the desired compound.
  • carboxylic acid 55 (R a is optionally substituted heteroaryl), anhydrous tetrahydrofuran, 1 drop anhydrous dimethylformamide, and oxalyl chloride are added under nitrogen.
  • the reaction mixture is allowed to stir at room temperature for 1.5 hours, then concentrated to dryness.
  • the resulting residue is diluted with anhydrous tetrahydrofuran, then triethylamine and (3-amino-2,4-difluoro-phenyl)-carbamic acid methyl ester 54 are added and allowed to stir at room temperature overnight.
  • the reaction is diluted with water extracted 3 ⁇ into ethyl acetate.
  • the organic extracts are dried over magnesium sulfate, filtered and concentrated in vacuo to provide the desired compound as a crude solid, which is purified by silica gel column chromatography (hexane: ethyl acetate gradient).
  • reaction is allowed to stir at 60° C. and monitored by TLC. When complete, reaction is extracted with 1N aqueous HCl and ethyl acetate. The organic layer is dried over anhydrous magnesium sulfate, filtered and volatile solvents removed to provide the desired compound as a crude solid.
  • 2,6-Difluoro-3-nitrobenzoic acid 60 is converted to 2,6-difluoro-3-nitro-phenylamine 61 following the methods described in Scheme 15, Step 3.
  • 2,6-Difluoro-3-nitro-phenylamine 61 is reacted with compound 55 following the methods described in Scheme 15, Step 4 to provide the desired compound 62.
  • Assays for the activity of kinases including, but not limited to, Fms, Kit, B-Raf, B-Raf V600E, B-Raf V600E/T529I and c-Raf-1 are known in the art, for example as described in US Patent Publication Number US20070032519 and U.S. patent application Ser. No. 11/473,347 (see also, PCT publication WO2007002433), the disclosures of which are hereby incorporated by reference in their entireties including all specifications, figures, and tables, and for all purposes.
  • c-Raf-1 P-0001, P-0002, P-0004, P-0008, P-0009, P-0011, P-0013, P-0015, P-0016, P-0017, P-0020, P-0021, P-0027
  • TrkA P-0002, P-0011
  • Compounds of the invention 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.
  • 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

  • This application is a continuation application of U.S. application Ser. No. 15/260,042, filed Sep. 8, 2016, which is a continuation application of U.S. application Ser. No. 15/048,851, filed Feb. 19, 2016, which is a continuation of U.S. application Ser. No. 13/926,959, filed Jun. 25, 2013, which is a continuation of U.S. application Ser. No. 13/866,353, filed Apr. 19, 2013, which is a continuation application of Ser. No. 12/669,450, filed Jan. 15, 2010, which application is a National Phase application under 35 U.S.C. §371 of PCT/US2008/070124, filed Jul. 16, 2008, which claims the benefit under 35 U.S.C. §119(e) from U.S. Application No. 60/959,907, filed Jul. 17, 2007, which applications are hereby incorporated by reference in their entirety.
  • FIELD OF THE INVENTION Background 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.
  • SUMMARY OF THE INVENTION
  • Compounds are contemplated that are active on protein kinases in general, including, but not limited to, Ab1, 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/or Zap70, including any mutations of these kinases. In some aspects, the compounds are active on Raf protein kinases including A-Raf, B-Raf and/or c-Raf-1, including any mutations thereof. In some aspects, compounds are of Formula I 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 US20170319559A1-20171109-C00001
  • or a salt, a prodrug, a tautomer or an isomer thereof,
    wherein:
      • Ar is optionally substituted heteroaryl;
      • R1 at each occurrence is independently 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, —NO2, —CN, —O—R5, —N(R5)—R6, —C(X)—N(R5)—R6, —C(X)—R7, —S(O)2—N(R5)—R6, —S(O)n—R7, —O—C(X)—R7, —C(X)—O—R5, —C(NH)—N(R8)—R9, —N(R5)—C(X)—R7, —N(R5)—S(O)2—R7, —N(R5)—C(X)—N(R5)—R6, and —N(R5)—S(O)2—N(R5)—R6;
      • m is 0, 1, 2, 3, 4 or 5;
      • n is 0, 1 or 2;
      • R2 is hydrogen, lower alkyl or halogen;
      • L2 is selected from the group consisting of —S(O)2—, —C(X)—, —C(X)—N(R10)—, and —S(O)2—N(R10)—;
      • R3 is optionally substituted lower alkyl, optionally substituted C3-6 cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl or optionally substituted heteroaryl;
      • L1 is selected from the group consisting of a bond, —N(R11)—, —O—, —S—, —C(X)—, —C(R12R13)—X—, —X—C(R12R13)—, —C(R12R13)—N(R11)—, —N(R11)—C(R12R13)—, —O—C(X)—, —C(X)—O—, —C(X)—N(R11)—, —N(R11)—C(X)—, —S(O)—, —S(O)2—, —S(O)2—N(R11)—, —N(R11)—S(O)2—, —C(NH)—N(R11)—, —N(R11)—C(NH)—, —N(R11)—C(X)—N(R11)—, and —N(R11)—S(O)2—N(R11)—;
      • X is O or S;
      • R4, R10 and each R11 are independently hydrogen or lower alkyl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, —OH, —NH2, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, fluoro substituted mono-alkylamino, di-alkylamino, fluoro substituted di-alkylamino, and —NR14R15;
      • R5, R6, R8, and R9 at each occurrence are independently selected from the group consisting of hydrogen, optionally substituted lower alkyl, optionally substituted C3-6 alkenyl, optionally substituted C3-6 alkynyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl, or
      • R8 and R9 combine with the nitrogen to which they are attached to form a 5-7 membered optionally substituted nitrogen containing heterocycloalkyl or a 5 or 7 membered optionally substituted nitrogen containing heteroaryl;
      • R7 at each occurrence is independently selected from the group consisting of optionally substituted lower alkyl, optionally substituted C3-6 alkenyl, optionally substituted C3-6 alkynyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl;
      • R12 and R13 are independently selected from the group consisting of hydrogen, fluoro, —OH, —NH2, lower alkyl, lower alkoxy, lower alkylthio, mono-alkylamino, di-alkylamino, and —NR14R15, wherein the alkyl chain(s) of lower alkyl, lower alkoxy, lower alkylthio, mono-alkylamino, or di-alkylamino are optionally substituted with one or more 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 cycloalkylamino; or
      • R12 and R13 combine with the carbon to which they are attached to form a 3-7 membered monocyclic cycloalkyl or 5-7 membered monocyclic heterocycloalkyl, wherein the monocyclic cycloalkyl or monocyclic heterocycloalkyl are optionally substituted with one or more substituents selected from the group consisting of halogen, —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; and
      • R14 and R15 at each occurrence independently combine with the nitrogen to which they are attached to form a 5-7 membered heterocycloalkyl or 5-7 membered heterocycloalkyl substituted with one or more substituents selected from the group consisting of fluoro, —OH, —NH2, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, and fluoro substituted lower alkylthio,
        provided, however, that when L1 is a bond, —NR11—, —O—, —S—, —C(X)—, —S(O)—, or —S(O)2—, Ar is not 1H-pyrrolo[2,3-b]pyridine-3-yl, 1H-pyrazolo[3,4-b]pyridine-3-yl, 5H-pyrrolo[2,3-b]pyrazine-7-yl, 7H-pyrrolo[2,3-d]pyrimidine-5-yl, or 7H-pyrrolo[2,3-c]pyridazine-5-yl, i.e. is not
  • Figure US20170319559A1-20171109-C00002
  • wherein
  • Figure US20170319559A1-20171109-C00003
  • indicates the attachment point to L1.
  • In some embodiments, the compound of Formula I has a structure according to the following sub-generic structure Formula Ia:
  • Figure US20170319559A1-20171109-C00004
  • or a salt, a prodrug, a tautomer or an isomer thereof,
    wherein m, Ar, R1, R2, R3, R4, and L1 are as defined for Formula I.
  • In some embodiments of compounds of Formula I or Ia, L1 is a bond, —N(R11)—, —N(R11)—C(X)—, —N(R11)—S(O)2—, —N(R11)—C(NH)—, —N(R11)—C(X)—N(R11)—, or —N(R11)—S(O)2—N(R11)—, also —N(R11)—, —N(R11)—C(X)—, or —N(R11)—S(O)2—, also —N(R11)—C(O)—, wherein the left side (i.e. —N(R11)—) of L1 is attached to Ar and the right side of L1 is attached to the phenyl ring of Formula I or Ia. In some embodiments, L1 is a bond, —N(R11)—, —N(R11)—C(X)—, —N(R11)—S(O)2—, —N(R11)—C(NH)—, —N(R11)—C(X)—N(R11)—, or —N(R11)—S(O)2—N(R11)—, also —N(R11)—, —N(R11)—C(X)—, or —N(R11)—S(O)2—, also —N(R11)—C(O)—, and each R11 and R4 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably each R11 and R4 are H.
  • In some embodiments of compounds of Formula I or Ia, L1 is —C(X)—N(R1)—, —C(R12R13)—X—, —X—C(R12R13)—, —C(R12R13)—N(R11)—, or —N(R11)—C(R12R13)—, wherein the left side of L1 is attached to Ar and the right side of L1 is attached to the phenyl ring of Formula I or Ia. In some embodiments, L1 is —C(X)—N(R11)—, —C(R12R13)—X—, —X—C(R12R13)—, —C(R12R13)—N(R11)—, or —N(R11)—C(R12R13)—, and each R11 and R4 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably each R11 and R4 are H.
  • In some embodiments of compounds of Formula I or Ia, R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro. In some embodiments, R2 is hydrogen, fluoro or chloro, each R11 and R4 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R2 is fluoro or chloro and each R11 and R4 are H. In some embodiments, R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro, and L1 is a bond, —N(R11)—, —N(R11)—C(X)—, —N(R11)—S(O)2—, —N(R11)—C(NH)—, —N(R11)—C(X)—N(R11)—, or —N(R11)—S(O)2—N(R11)—, also —N(R11)—, —N(R11)—C(X)—, or —N(R1)—S(O)2—, also —N(R1)—C(O)—, wherein the left side (i.e. —N(R11)—) of L1 is attached to Ar and the right side of L1 is attached to the phenyl ring of Formula I or Ia. In some embodiments, R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro; L1 is a bond, —N(R11)—, —N(R11)—C(X)—, —N(R11)—S(O)2—, —N(R11)—C(NH)—, —N(R11)—C(X)—N(R11)—, or —N(R11)—S(O)2—N(R11)—, also —N(R11)—, —N(R11)—C(X)—, or —N(R11)—S(O)2—, also —N(R11)—C(O)—; and each R11 and R4 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably each R11 and R4 are H.
  • In some embodiments of compounds of Formula I or Ia, R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro. In some embodiments, R2 is hydrogen, fluoro or chloro, each R11 and R4 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R2 is fluoro or chloro and each R11 and R4 are H. In some embodiments, R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro, and L1 is —C(X)—N(R11)—, —C(R12R13)—X—, —X—C(R12R13)—, —C(R12R13)—N(R11)—, or —N(R11)—C(R12R13)—, wherein the left side of L1 is attached to Ar and the right side of L1 is attached to the phenyl ring of Formula I or Ia. In some embodiments, R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro; L1 is —C(X)—N(R11)—, —C(R12R13)—X—, —X—C(R12R13)—, —C(R12R13)—N(R11)—, or —N(R11)—C(R12R13)—; and each R11 and R4 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably each R11 and R4 are H.
  • In some embodiments, the compound of Formula I has a structure according to the following sub-generic structure Formula Ib:
  • Figure US20170319559A1-20171109-C00005
  • or a salt, a prodrug, a tautomer or an isomer thereof,
    wherein A is —C(O)— or —C(R12R13)—; and
    m, Ar, R1, R2, R3, R4, R11, R12 and R13 are as defined for Formula I.
  • In some embodiments of compounds of Formula Ib, R4, R11, R12 and R13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R4, R11, R12 and R13 are H. In some embodiments, R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro. In some embodiments, R2 is hydrogen, fluoro or chloro, and R4, R11, R12 and R13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R2 is fluoro or chloro and R4, R11, R12 and R13 are H.
  • In some embodiments of compounds of Formula Ib, A is —C(O)—, and R4 and R11 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R4 and R11 are H. In some embodiments, A is —C(O)—, and R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro. In some embodiments, A is —C(O)—, R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro; and R4 and R11 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R4 and R11 are H.
  • In some embodiments of compounds of Formula Ib, A is —C(R12R13)—, and R4, R11, R12 and R13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R4, R11, R12 and R13 are H. In some embodiments, A is —C(R12R13)—, R12 and R13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R12 and R13 are H, and R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro. In some embodiments, A is —C(R12R13)—; R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro; and R4, R11, R12 and R13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R4, R11, R12 and R13 are H.
  • In some embodiments, the compound of Formula I has a structure according to the following sub-generic structure Formula Ic:
  • Figure US20170319559A1-20171109-C00006
  • or a salt, a prodrug, a tautomer or an isomer thereof,
    wherein m, Ar, R1, R2, R3, and R4, are as defined for Formula I.
  • In some embodiments of compounds of Formula Ic, R4 is hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R4 is H. In some embodiments, R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro. In some embodiments, R2 is hydrogen, fluoro or chloro and R4 is hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R2 is fluoro or chloro and R4 is H.
  • In some embodiments, the compound of Formula I has a structure according to the following sub-generic structure Formula Id:
  • Figure US20170319559A1-20171109-C00007
  • or a salt, a prodrug, a tautomer or an isomer thereof,
    wherein A is —C(O)— or —C(R12R13)—; and
    m, Ar, R1, R2, R3, R4, R11, R12, R13 and L2 are as defined for Formula I.
  • In some embodiments of compounds of Formula Id, R4, R11, R12 and R13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R4, R11, R12 and R13 are H. In some embodiments, R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro. In some embodiments, R2 is hydrogen, fluoro or chloro, and R4, R11, R12 and R13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R2 is fluoro or chloro and R4, R11, R12 and R13 are H.
  • In some embodiments of compounds of Formula Id, A is —C(O)—, and R4 and R11 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R4 and R11 are H. In some embodiments, A is —C(O)—, and R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro. In some embodiments, A is —C(O)—, R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro; and R4 and R11 are hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R4 and R11 are H.
  • In some embodiments of compounds of Formula Id, A is —C(R12R13)—, and R4, R11, R12 and R13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R4, R11, R12 and R13 are H. In some embodiments, A is —C(R12R13)—, R12 and R13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R12 and R13 are H, and R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro. In some embodiments, A is —C(R12R13)—; R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro; and R4, R11, R12 and R13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R4, R11, R12 and R13 are H.
  • In some embodiments, the compound of Formula I has a structure according to the following sub-generic structure Formula Ie:
  • Figure US20170319559A1-20171109-C00008
  • or a salt, a prodrug, a tautomer or an isomer thereof,
    wherein m, Ar, R1, R2, R3, R4, and L2 are as defined for Formula I.
  • In some embodiments of compounds of Formula Ie, R4 is hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R4 is H. In some embodiments, R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro. In some embodiments, R2 is hydrogen, fluoro or chloro and R4 is hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R2 is fluoro or chloro and R4 is H.
  • In some embodiments, the compound of Formula I has a structure according to the following sub-generic structure Formula If:
  • Figure US20170319559A1-20171109-C00009
  • or a salt, a prodrug, a tautomer or an isomer thereof,
  • wherein:
  • L3 is —C(X)—N(R11)—, —C(R12R13)—X—, —X—C(R12R13)—, —C(R12R13)—N(R11)—, or —N(R11)—C(R12R13)—;
    and
    m, Ar, R1, R2, R3, R4, R11, R12, R13, and X are as defined for Formula I.
  • In some embodiments of compounds of Formula If, L3 is —C(O)—N(R11)—, —C(R12R13)—O—, —O—C(R12R13)—, —C(R12R13)—N(R11)—, or —N(R11)—C(R12R13)—, wherein the left side of L3 is attached to Ar and the right side of L3 is attached to the phenyl ring of Formula If. In some embodiments, L3 is —C(O)—N(R11)—, —C(R12R13)—O—, —O—C(R12R13)—, —C(R12R13)—N(R11)—, or —N(R11)—C(R12R13)—, and R4, R11, R12 and R13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R4, R11, R12 and R13 are H.
  • In some embodiments, the compound of Formula I has a structure according to the following sub-generic structure Formula Ig:
  • Figure US20170319559A1-20171109-C00010
  • or a salt, a prodrug, a tautomer or an isomer thereof,
  • wherein:
  • L3 is —C(X)—N(R11)—, —C(R12R13)—X—, —X—C(R12R13)—, —C(R12R13)—N(R11)—, or —N(R11)—C(R12R13)—;
    and
    m, Ar, R1, R2, R3, R4, R11, R12, R13, X and L2 are as defined for Formula I.
  • In some embodiments of compounds of Formula Ig, L3 is —C(O)—N(R11)—, —C(R12R13)—O—, —O—C(R12R13)—, —C(R12R13)—N(R11)—, or —N(R11)—C(R12R13)—, wherein the left side of L3 is attached to Ar and the right side of L3 is attached to the phenyl ring of Formula Ig. In some embodiments, L3 is —C(O)—N(R11)—, —C(R12R13)—O—, —O—C(R12R13)—, —C(R12R13)—N(R11)—, or —N(R11)—C(R12R13)—, and R4, R11, R12 and R13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R4, R11, R12 and R13 are H.
  • In some embodiments, the compound of Formula I has a structure according to the following sub-generic structure Formula Ih:
  • Figure US20170319559A1-20171109-C00011
  • or a salt, a prodrug, a tautomer or an isomer thereof,
    wherein A is —C(O)— or —C(R12R13)—; and
    m, Ar, R1, R2, R3, R4, R11, R12 and R13 are as defined for Formula I.
  • In some embodiments of compounds of Formula Ih, R4, R11, R12 and R13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R4, R11, R12 and R13 are H. In some embodiments, R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro. In some embodiments, R2 is hydrogen, fluoro or chloro, and R4, R11, R12 and R13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R2 is fluoro or chloro and R4, R11, R12 and R13 are H.
  • In some embodiments of compounds of Formula Ih, A is —C(O)—, and R4 and R11 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R4 and R11 are H. In some embodiments, A is —C(O)—, and R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro. In some embodiments, A is —C(O)—, R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro; and R4 and R11 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R4 and R11 are H.
  • In some embodiments of compounds of Formula Ih, A is —C(R12R13)—, and R4, R11, R12 and R13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R4, R11, R12 and R13 are H. In some embodiments, A is —C(R12R13)—, R12 and R13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R12 and R13 are H, and R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro. In some embodiments, A is —C(R12R13)—; R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro; and R4, R11, R12 and R13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R4, R11, R12 and R13 are H.
  • In some embodiments, the compound of Formula I has a structure according to the following sub-generic structure Formula Ii:
  • Figure US20170319559A1-20171109-C00012
  • or a salt, a prodrug, a tautomer or an isomer thereof,
    wherein A is —C(O)— or —C(R12R13)—; and
    m, Ar, R1, R2, R3, R4, R11, R12, R13 and L2 are as defined for Formula I.
  • In some embodiments of compounds of Formula Ii, R4, R11, R12 and R13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R4, R11, R12 and R13 are H. In some embodiments, R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro. In some embodiments, R2 is hydrogen, fluoro or chloro, and R4, R11, R12 and R13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R2 is fluoro or chloro and R4, R11, R12 and R13 are H.
  • In some embodiments of compounds of Formula Ii, A is —C(O)—, and R4 and R11 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R4 and R11 are H. In some embodiments, A is —C(O)—, and R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro. In some embodiments, A is —C(O)—, R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro; and R4 and R11 are hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R4 and R11 are H.
  • In some embodiments of compounds of Formula Ii, A is —C(R12R13)—, and R4, R11, R12 and R13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R4, R11, R12 and R13 are H. In some embodiments, A is —C(R12R13)—, R12 and R13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R12 and R13 are H, and R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro. In some embodiments, A is —C(R12R13)—; R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro; and R4, R11, R12 and R13 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R4, R11, R12 and R13 are H.
  • In some embodiments, the compound of Formula I has a structure according to the following sub-generic structure Formula Ij:
  • Figure US20170319559A1-20171109-C00013
  • or a salt, a prodrug, a tautomer or an isomer thereof,
    wherein:
      • m, Ar, R1, R2, R4, and L1 are as defined for Formula I; and
      • R22 is selected from the group consisting of mono-alkylamino, di-alkylamino, optionally substituted lower alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl, wherein the alkyl chain(s) of mono-alkylamino or di-alkylamino are independently optionally substituted with one or more 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 cycloalkylamino.
  • In some embodiments of compounds of Formula Ij, L1 is a bond, —N(R11)—, —N(R11)—C(X)—, —N(R11)—S(O)2—, —N(R11)—C(NH)—, —N(R11)—C(X)—N(R11)—, or —N(R11)—S(O)2—N(R11)—, also —N(R11)—, —N(R11)—C(X)—, or —N(R11)—S(O)2—, also —N(R11)—C(O)—, wherein the left side (i.e. —N(R11)—) of L1 is attached to Ar and the right side of L1 is attached to the phenyl ring of Formula Ij. In some embodiments, L1 is a bond, —N(R11)—, —N(R11)—C(X)—, —N(R11)—S(O)2—, —N(R11)—C(NH)—, —N(R11)—C(X)—N(R11)—, or —N(R11)—S(O)2—N(R11)—, also —N(R11)—, —N(R1)—C(X)—, or —N(R11)—S(O)2—, also —N(R11)—C(O)—, and each R11 and R4 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably each R11 and R4 are H.
  • In some embodiments of compounds of Formula Ij, L1 is —C(X)—N(R11)—, —C(R12R13)—X—, —X—C(R12R13)—, —C(R12R13)—N(R11)—, or —N(R11)—C(R12R13)—, wherein the left side of L1 is attached to Ar and the right side of L1 is attached to the phenyl ring of Formula Ij. In some embodiments, L1 is —C(X)—N(R11)—, —C(R12R13)—X—, —X—C(R12R13)—, —C(R12R13)—N(R11)—, or —N(R11)—C(R12R13)—, and each R11 and R4 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably each R11 and R4 are H.
  • In some embodiments of compounds of Formula Ij, R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro. In some embodiments, R2 is hydrogen, fluoro or chloro, each R11 and R4 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R2 is fluoro or chloro and each R11 and R4 are H. In some embodiments, R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro, and L1 is a bond, —N(R11)—, —N(R1)—C(X)—, —N(R11)—S(O)2—, —N(R11)—C(NH)—, —N(R11)—C(X)—N(R11)—, or —N(R11)—S(O)2—N(R11)—, also —N(R11)—, —N(R11)—C(X)—, or —N(R11)—S(O)2—, also —N(R11)—C(O)—, wherein the left side (i.e. —N(R11)—) of L1 is attached to Ar and the right side of L1 is attached to the phenyl ring of Formula Ij. In some embodiments, R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro; L1 is a bond, —N(R11)—, —N(R11)—C(X)—, —N(R11)—S(O)2—, —N(R11)—C(NH)—, —N(R11)—C(X)—N(R11)—, or —N(R11)—S(O)2—N(R11)—, also —N(R11)—, —N(R1)—C(X)—, or —N(R11)—S(O)2—, also —N(R11)—C(O)—; and each R11 and R4 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably each R11 and R4 are H.
  • In some embodiments of compounds of Formula Ij, R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro. In some embodiments, R2 is hydrogen, fluoro or chloro, each R11 and R4 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably R2 is fluoro or chloro and each R11 and R4 are H. In some embodiments, R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro, and L1 is —C(X)—N(R11)—, —C(R12R13)—X—, —X—C(R12R13)—, —C(R12R13)—N(R11)—, or —N(R11)—C(R12R13)—, wherein the left side of L1 is attached to Ar and the right side of L1 is attached to the phenyl ring of Formula Ij. In some embodiments, R2 is hydrogen, fluoro or chloro, preferably fluoro or chloro; L1 is —C(X)—N(R11)—, —C(R12R13)—X—, —X—C(R12R13)—, —C(R12R13)—N(R11)—, or —N(R11)—C(R12R13)—; and each R11 and R4 are independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio, preferably each R11 and R4 are H.
  • In some embodiments of compounds of Formula Ij, further to any of the above embodiments of Formula Ij, R22 is mono-alkylamino, di-alkylamino, or optionally substituted heterocycloalkyl, preferably wherein heterocycloalkyl is a 5 or 6 membered nitrogen containing heterocycloalkyl, wherein a nitrogen of the heterocycloalkyl is bound to the S(O)2 of Formula Ij. In some embodiments, R22 is mono-alkylamino, di-alkylamino or 5 or 6 membered nitrogen containing heterocycloalkyl, wherein the heterocycloalkyl is substituted with one or more substituents selected from the group consisting of fluoro, —OH, —NH2, lower alkyl, lower alkoxy, lower alkylthio, mono-alkylamino, di-alkylamino, and cycloalkylamino, wherein lower alkyl or the alkyl chain(s) of lower alkoxy, lower alkylthio, mono-alkylamino, or di-alkylamino are optionally substituted with one or more 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 cycloalkylamino, preferably wherein the 5 or 6 membered nitrogen containing heterocycloalkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, and fluoro substituted lower alkylthio.
  • In some embodiments of compounds of Formula I, Ia, Ib, Ic, Id, Ie, If, Ig, Ih, Ii, or Ij, further to any of the above embodiments of Formula I, Ia, Ib, Ic, Id, Ie, If, Ig, Ih, Ii or Ij, Ar is monocyclic or bicyclic nitrogen containing heteroaryl. In some embodiments, Ar is selected from the group consisting of
  • Figure US20170319559A1-20171109-C00014
    Figure US20170319559A1-20171109-C00015
  • wherein
  • Figure US20170319559A1-20171109-C00016
  • indicates the attachment point of the Ar ring to L1 in Formula I, Ia or Ij, to L3 in Formula If or Ig, to the nitrogen of Ar—N— in Formula Ib or Id, to A of Formula Ih or Ii, or to the phenyl ring of Formula Ic or Ie, and wherein:
      • T, U, X, and Z at each occurrence are independently N or CH, provided, however, that at least one, and no more than 2 of X in any ring is N, no more than 2 of U in any ring is N, at least one T is N and no more than two of T within any six-membered ring is N, and at least one Z is N and no more than 2 of Z within any bicyclic ring is N;
      • Y is NH, O, or S;
      • W at each occurrence is independently N or CH;
      • V is O, S, or NH, provided, however, that when V is O or S, at least one of U in any ring or W in any ring is N; and
      • any R1 is bound to Ar at any available NH or CH, preferably any R1 is independently R16, wherein R16 at each occurrence is independently selected from the group consisting of —OH, —NH2, —CN, —NO2, —C(O)—OH, —S(O)2—NH2, —C(O)—NH2, —O—R17, —S—R17, —N(R19)—R17, —N(R19)—C(O)—R17, —N(R19)—S(O)2—R17, —S(O)2—R17, —C(O)—R17, —C(O)—O—R17, —C(O)—N(R19)—R17, —S(O)2—N(R19)—R17, 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 R16, or as substituents of lower alkyl, are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —C(O)—OH, —S(O)2—NH2, —C(O)—NH2, —O—R18, —S—R18, —N(R19)—R18, —N(R19)—C(O)—R18, —N(R19)—S(O)2—R18, —S(O)2—R18, —C(O)—R18, —C(O)—O—R18, —C(O)—N(R19)—R18, —S(O)2—N(R19)—R18, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino;
      • R17 at each occurrence is independently 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 R17 or as substituents of lower alkyl are optionally substituted with one or more substituents selected from the group consisting of —OH, —NH2, —CN, —NO2, —C(O)—OH, —S(O)2—NH2, —C(O)—NH2, —O—R18, —S—R18, —N(R19)—R18, —N(R19)—C(O)—R18, —N(R19)—S(O)2—R18, —S(O)2—R18, —C(O)—R18, —C(O)—O—R18, —C(O)—N(R19)—R18, —S(O)2—N(R19)—R18, halogen, lower alkyl, fluoro substituted lower alkyl, and cycloalkylamino;
      • R18 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
      • R19 at each occurrence is independently hydrogen or lower alkyl, 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, and fluoro substituted lower alkylthio.
  • In some embodiments of compounds of Formula I, Ia, Ib, Id, If, Ig, Ih, Ii, or Ij, further to any of the above embodiments of Formula I, Ia, Ib, Id, If, Ig, Ih, Ii, or Ij, when L1 is other than a bond, Ar is selected from the group consisting of
  • Figure US20170319559A1-20171109-C00017
    Figure US20170319559A1-20171109-C00018
  • wherein
  • Figure US20170319559A1-20171109-C00019
  • indicates the attachment point of the Ar ring to L1 in Formula I, Ia or Ij, to L3 in Formula If or Ig, to the nitrogen of Ar—N— in Formula Ib or Id, or to A of Formula Ih or Ii, and wherein:
      • T1, U1, W1, X1, and Z1 at each occurrence are independently N or CH, provided, however, that at most 1 of T1, U1, X1, and Z1 is N;
      • T2 at each occurrence is independently N or CH, provided, however, that at most 2 of T2 are N;
      • Y and V1 are O, S, or NH;
      • any R1 is bound to Ar at any available NH or CH, preferably any R1 is independently R16, as defined in paragraph [0041], preferably wherein R16 at each occurrence is independently selected from the group consisting of halogen, —OH, —NH2, —CN, lower alkyl, lower alkoxy, lower alkylthio, mono-alkylamino, di-alkylamino, and —NR20R21, wherein lower alkyl and the alkyl chain(s) of lower alkoxy, lower alkylthio, mono-alkylamino or di-alkylamino are optionally substituted with one or more, preferably 1, 2, or 3 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, or cycloalkylamino; and
      • R20 and R21 at each occurrence independently combine with the nitrogen to which they are attached to form a 5-7 membered heterocycloalkyl or 5-7 membered heterocycloalkyl substituted with one or more substituents selected from the group consisting of fluoro, —OH, —NH2, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, and fluoro substituted lower alkylthio.
  • In some embodiments of compounds of Formula I, Ia, Ib, Id, If, Ig, Ih, Ii, or Ij, further to any of the above embodiments of Formula I, Ia, Ib, Id, If, Ig, Ih, Ii, or Ij, when L1 is other than a bond, Ar is selected from the group consisting of
  • Figure US20170319559A1-20171109-C00020
    Figure US20170319559A1-20171109-C00021
  • wherein
  • Figure US20170319559A1-20171109-C00022
  • indicates the attachment point of the Ar ring to L1 in Formula I, Ia or Ij, to L3 in Formula If or Ig, to the nitrogen of Ar—N— in Formula Ib or Id, or to A of Formula Ih or Ii, and wherein:
      • W1 at each occurrence is independently N or CH;
      • Y and V1 are O, S, or NH;
      • p is 0, 1, 2 or 3; and
      • R16, substituting at any available CH or NH, is as defined in paragraph [0041], preferably wherein R16 at each occurrence is independently selected from the group consisting of halogen, —OH, —NH2, —CN, lower alkyl, lower alkoxy, lower alkylthio, mono-alkylamino, di-alkylamino, and —NR20R21, wherein lower alkyl and the alkyl chain(s) of lower alkoxy, lower alkylthio, mono-alkylamino or di-alkylamino are optionally substituted with one or more, preferably 1, 2, or 3 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, or cycloalkylamino, wherein R20 and R21 are as defined in paragraph [0042].
  • In some embodiments of compounds of Formula I, Ia, Ib, Id, If, Ig, Ih, Ii, or Ij, further to any of the above embodiments of Formula I, Ia, Ib, Id, If, Ig, Ih, Ii, or Ij, when L1 is other than a bond, Ar is selected from the group consisting of
  • Figure US20170319559A1-20171109-C00023
  • wherein
  • Figure US20170319559A1-20171109-C00024
  • indicates the attachment point of the Ar ring to L1 in Formula I, Ia or Ij, to L3 in Formula If or Ig, to the nitrogen of Ar—N— in Formula Ib or Id, or to A of Formula Ih or Ii, and wherein:
  • p is 0, 1, 2 or 3; and
  • R16, substituting at any available CH or NH, is as defined in paragraph [0041].
  • In some embodiments of compounds of Formula Ic or Ie, further to any of the above embodiments of Formula Ic or Ie, Ar is selected from the group consisting of
  • Figure US20170319559A1-20171109-C00025
  • wherein
  • Figure US20170319559A1-20171109-C00026
  • indicates the attachment point of the Ar ring to the phenyl ring of Formula Ic or Ie, and wherein:
      • T1, U1, W1 and Z1 at each occurrence is independently N or CH, provided, however, that at most 1 of T1, U1, and Z1 is N;
      • T2 is N or CH, provided, however, that no more than 2 of T2 are N;
      • V1 is O, S, or NH; and
      • any R1 is bound to Ar at any available NH or CH, preferably any R1 is independently R16, as defined in paragraph [0041], preferably wherein R16 at each occurrence is independently selected from the group consisting of halogen, —OH, —NH2, —CN, lower alkyl, lower alkoxy, lower alkylthio, mono-alkylamino, di-alkylamino, and —NR20R21, wherein lower alkyl and the alkyl chain(s) of lower alkoxy, lower alkylthio, mono-alkylamino or di-alkylamino are optionally substituted with one or more, preferably 1, 2, or 3 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, or cycloalkylamino, wherein R20 and R21 are as defined in paragraph [0042].
  • In some embodiments of compounds of Formula Ic or Ie, further to any of the above embodiments of Formula Ic or Ie, R1 is selected from the group consisting of
  • Figure US20170319559A1-20171109-C00027
  • wherein
  • Figure US20170319559A1-20171109-C00028
  • indicates the attachment point of the Ar ring to the phenyl ring of Formula Ic or Ie, and wherein:
      • W1 is N or CH;
      • V1 is O, S, or NH;
      • p is 0, 1, 2 or 3; and
      • R16, substituting at any available CH or NH, is as defined in paragraph [0041], preferably wherein R16 at each occurrence is independently selected from the group consisting of halogen, —OH, —NH2, —CN, lower alkyl, lower alkoxy, lower alkylthio, mono-alkylamino, di-alkylamino, and —NR20R21, wherein lower alkyl and the alkyl chain(s) of lower alkoxy, lower alkylthio, mono-alkylamino or di-alkylamino are optionally substituted with one or more, preferably 1, 2, or 3 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, or cycloalkylamino, wherein R20 and R21 are as defined in paragraph [0042].
  • In some embodiments of compounds of Formula I, Ia, Ib, Ic, Id, Ie, If, Ig, Ih, or Ii, further to any of the above embodiments of Formula I, Ia, Ib, Ic, Id, Ie, If, Ig, Ih, or Ii, R3 is optionally substituted lower alkyl or optionally substituted C3-6 cycloalkyl. In some embodiments, R3 is lower alkyl or C3-6 cycloalkyl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, —OH, —NH2, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, fluoro substituted mono-alkylamino, di-alkylamino, fluoro substituted di-alkylamino, cycloalkylamino, and C3-5 cycloalkyl, and wherein C3-6 cycloalkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, —OH, —NH2, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, fluoro substituted mono-alkylamino, di-alkylamino, and fluoro substituted di-alkylamino. In some embodiments, R3 is lower alkyl or C3-6 cycloalkyl, wherein lower alkyl or C3-6 cycloalkyl are optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, and fluoro substituted lower alkylthio. In some embodiments, R3 is optionally fluoro substituted lower alkyl or optionally fluoro substituted C3-6 cycloalkyl. In some embodiments, R3 is lower alkyl, wherein lower alkyl is optionally substituted with one or more substituents selected from the group consisting of fluoro, —OH, —NH2, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, fluoro substituted mono-alkylamino, di-alkylamino, fluoro substituted di-alkylamino, cycloalkylamino, also one or more substituents selected from the group consisting of fluoro, —OH, —NH2, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, fluoro substituted lower alkylthio, mono-alkylamino, fluoro substituted mono-alkylamino, di-alkylamino, and fluoro substituted di-alkylamino, also one or more substituents selected from the group consisting of fluoro, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, and fluoro substituted lower alkylthio. In some embodiments, R3 is optionally fluoro substituted lower alkyl.
  • In some embodiments of compounds of Formula I, Ia, Ib, Ic, Id, Ie, If, Ig, Ih, or Ii, further to any of the above embodiments of Formula I, Ia, Ib, Ic, Id, Ie, If, Ig, Ih, or Ii, R3 is optionally substituted phenyl, also phenyl mono-substituted at the para position, also phenyl mono-substituted at the meta position. In some embodiments, R3 is phenyl optionally substituted with one or more substituents selected from the group consisting of halogen, —CN, —NO2, —OH, —NH2, lower alkyl, lower alkoxy, lower alkylthio, mono-alkylamino, di-alkylamino, and cycloalkylamino, wherein lower alkyl or the alkyl chain(s) of lower alkoxy, lower alkylthio, mono-alkylamino, or di-alkylamino are optionally substituted with one or more 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 cycloalkylamino, preferably wherein the phenyl is mono-substituted at either the para or meta position. In some embodiments, R3 is phenyl optionally substituted with one or more substituents selected from the group consisting of fluoro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, lower alkylthio, and fluoro substituted lower alkylthio, preferably wherein the phenyl is mono-substituted at either the para or meta position.
  • In one embodiment of compounds of Formula I, the compound is selected from the group consisting of:
    • 6-Chloro-2-fluoro-3-(propane-1-sulfonylamino)-N-pyridin-3-yl-benzamide (P-0001),
    • N-(6-Acetylamino-pyridin-3-yl)-6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzamide (P-0002),
    • 6-Chloro-2-fluoro-N-(6-methoxy-pyridin-3-yl)-3-(propane-1-sulfonylamino)-benzamide (P-0003),
    • N-(2-Acetylamino-pyrimidin-5-yl)-6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzamide (P-0004),
    • 6-Chloro-2-fluoro-N-(6-isopropylamino-pyridin-3-yl)-3-(propane-1-sulfonylamino)-benzamide (P-0005),
    • Pyrrolidine-1-carboxylic acid {5-[6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoylamino]-pyridin-2-yl}-amide (P-0006),
    • 6-Chloro-N-(3,5-dimethyl-isoxazol-4-yl)-2-fluoro-3-(propane-1-sulfonylamino)-benzamide (P-0007),
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(propane-1-sulfonylamino)-benzamide (P-0008),
    • 6-Chloro-N-(6-cyclopentylamino-pyridin-3-yl)-2-fluoro-3-(propane-1-sulfonylamino)-benzamide (P-0009),
    • 6-Chloro-N-[5-(4-chloro-phenyl)-2H-pyrazol-3-yl]-2-fluoro-3-(propane-1-sulfonylamino)-benzamide (P-0010),
    • 6-Chloro-2-fluoro-N-[6-(5-methyl-thiazol-2-ylamino)-pyridin-3-yl]-3-(propane-1-sulfonylamino)-benzamide (P-0011),
    • 6-Chloro-N-[5-(4-chloro-benzyl)-[1,3,4]thiadiazol-2-yl]-2-fluoro-3-(propane-1-sulfonylamino)-benzamide (P-0012),
    • 6-Chloro-2-fluoro-3-(propane-1-sulfonylamino)-N-quinolin-3-yl-benzamide (P-0013),
    • [2-[6-Chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoylamino]-4H-[1,3,4]thiadiazin-(5E)-ylidene]-acetic acid ethyl ester (P-0014),
    • 6-Chloro-N-(6-cyclopropylamino-pyridin-3-yl)-2-fluoro-3-(propane-1-sulfonylamino)-benzamide (P-0015),
    • 6-Chloro-2-fluoro-3-(propane-1-sulfonylamino)-N-{6-[(thiophen-2-ylmethyl)-amino]-pyridin-3-yl}-benzamide (P-0016),
    • N-(6-Benzylamino-pyridin-3-yl)-6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzamide (P-0017),
    • 6-Chloro-2-fluoro-N-imidazo[1,2-a]pyridin-3-yl-3-(propane-1-sulfonylamino)-benzamide (P-0018),
    • Propane-1-sulfonic acid {2,4-difluoro-3-[(5-methyl-isoxazol-3-ylamino)-methyl]-phenyl}-amide (P-0019),
    • N-{5-[2,6-Difluoro-3-(propane-1-sulfonylamino)-benzylamino]-pyridin-2-yl}-acetamide (P-0020),
    • Propane-1-sulfonic acid [2,4-difluoro-3-(quinolin-3-ylaminomethyl)-phenyl]-amide (P-0021),
    • Propane-1-sulfonic acid {3-[(6-chloro-pyridin-3-ylamino)-methyl]-2,4-difluoro-phenyl}-amide (P-0022),
    • Propane-1-sulfonic acid {2,4-difluoro-3-[(6-methoxy-pyridin-3-ylamino)-methyl]-phenyl}-amide (P-0023),
    • Quinoline-3-carboxylic acid [2,6-difluoro-3-(propane-1-sulfonylamino)-phenyl]-amide (P-0024),
    • Propane-1-sulfonic acid {2,4-difluoro-3-[(quinolin-3-ylmethyl)-amino]-phenyl}-amide (P-0025),
    • Propane-1-sulfonic acid [2,4-difluoro-3-(quinolin-3-yloxymethyl)-phenyl]-amide (P-0026),
    • 2,6-Difluoro-3-(propane-1-sulfonylamino)-N-quinolin-3-yl-benzamide (P-0027),
    • 6-Acetylamino-N-[2,6-difluoro-3-(2-fluoro-benzenesulfonylamino)-phenyl]-nicotinamide (P-0028),
    • 6-Acetylamino-N-[2,6-difluoro-3-(3-fluoro-benzenesulfonylamino)-phenyl]-nicotinamide (P-0029),
    • 6-Acetylamino-N-[3-(2,6-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-nicotinamide (P-0030),
    • 6-Acetylamino-N-[3-(2,4-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-nicotinamide (P-0031),
    • 6-Acetylamino-N-[3-(2,5-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-nicotinamide (P-0032),
    • 6-Acetylamino-N-[2,6-difluoro-3-(3-fluoro-4-methoxy-benzenesulfonylamino)-phenyl]-nicotinamide (P-0033),
    • 6-Acetylamino-N-[2,6-difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-phenyl]-nicotinamide (P-0034),
    • 6-Acetylamino-N-[3-(4-difluoromethoxy-benzenesulfonylamino)-2,6-difluoro-phenyl]-nicotinamide (P-0035),
    • 6-Acetylamino-N-[3-(3-difluoromethoxy-benzenesulfonylamino)-2,6-difluoro-phenyl]-nicotinamide (P-0036),
    • 6-Acetylamino-N-[2,6-difluoro-3-(4-isopropyl-benzenesulfonylamino)-phenyl]-nicotinamide (P-0037),
    • 6-Acetylamino-N-[3-(4-tert-butyl-benzenesulfonylamino)-2,6-difluoro-phenyl]-nicotinamide (P-0038),
    • 6-Acetylamino-N-[2,6-difluoro-3-(4-propyl-benzenesulfonylamino)-phenyl]-nicotinamide (P-0039),
    • 6-Acetylamino-N-[2,6-difluoro-3-(pyridine-2-sulfonylamino)-phenyl]-nicotinamide (P-0040),
    • 6-Acetylamino-N-[2,6-difluoro-3-(pyridine-3-sulfonylamino)-phenyl]-nicotinamide (P-0041),
    • 6-Acetylamino-N-[2,6-difluoro-3-(dimethylaminosulfonylamino)-phenyl]-nicotinamide (P-0042),
    • 6-Acetylamino-N-[2,6-difluoro-3-(piperidine-1-sulfonylamino)-phenyl]-nicotinamide (P-0043),
    • 6-Acetylamino-N-[2,6-difluoro-3-(morpholine-4-sulfonylamino)-phenyl]-nicotinamide (P-0044),
    • 6-Acetylamino-N-[2,6-difluoro-3-(tetrahydro-pyran-4-sulfonylamino)-phenyl]-nicotinamide (P-0045),
    • 6-Acetylamino-N-(3-cyclopentanesulfonylamino-2,6-difluoro-phenyl)-nicotinamide (P-0046),
    • 6-Acetylamino-N-[2,6-difluoro-3-(pyrrolidine-1-sulfonylamino)-phenyl]-nicotinamide (P-0047),
    • 6-Acetylamino-N-[2,6-difluoro-3-(3,3,3-trifluoro-propane-1-sulfonylamino)-phenyl]-nicotinamide (P-0048),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(2-fluoro-benzenesulfonylamino)-phenyl]-amide (P-0049),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(3-fluoro-benzenesulfonylamino)-phenyl]-amide (P-0050),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [3-(2,6-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide (P-0051),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [3-(2,4-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide (P-0052),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [3-(2,5-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide (P-0053),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(3-fluoro-4-methoxy-benzenesulfonylamino)-phenyl]-amide (P-0054),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-phenyl]-amide (P-0055),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [3-(4-difluoromethoxy-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide (P-0056),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [3-(3-difluoromethoxy-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide (P-0057),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(4-isopropyl-benzenesulfonylamino)-phenyl]-amide (P-0058),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [3-(4-tert-butyl-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide (P-0059),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(4-propyl-benzenesulfonylamino)-phenyl]-amide (P-0060),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(pyridine-2-sulfonylamino)-phenyl]-amide (P-0061),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(pyridine-3-sulfonylamino)-phenyl]-amide (P-0062),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(dimethylaminosulfonylamino)-phenyl]-amide (P-0063),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(piperidine-1-sulfonylamino)-phenyl]-amide (P-0064),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(morpholine-4-sulfonylamino)-phenyl]-amide (P-0065),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(tetrahydro-pyran-4-sulfonylamino)-phenyl]-amide (P-0066),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid (3-cyclopentanesulfonylamino-2,6-difluoro-phenyl)-amide (P-0067),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(pyrrolidine-1-sulfonylamino)-phenyl]-amide (P-0068),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(3,3,3-trifluoro-propane-1-sulfonylamino)-phenyl]-amide (P-0069),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(2-fluoro-benzenesulfonylamino)-phenyl]-amide (P-0070),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(3-fluoro-benzenesulfonylamino)-phenyl]-amide (P-0071),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [3-(2,6-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide (P-0072),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [3-(2,4-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide (P-0073),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [3-(2,5-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide (P-0074),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(3-fluoro-4-methoxy-benzenesulfonylamino)-phenyl]-amide (P-0075),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-phenyl]-amide (P-0076),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [3-(4-difluoromethoxy-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide (P-0077),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [3-(3-difluoromethoxy-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide (P-0078),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(4-isopropyl-benzenesulfonylamino)-phenyl]-amide (P-0079),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [3-(4-tert-butyl-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide (P-0080),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(4-propyl-benzenesulfonylamino)-phenyl]-amide (P-0081),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(pyridine-2-sulfonylamino)-phenyl]-amide (P-0082),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(pyridine-3-sulfonylamino)-phenyl]-amide (P-0083),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(dimethylaminosulfonylamino)-phenyl]-amide (P-0084),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(piperidine-1-sulfonylamino)-phenyl]-amide (P-0085),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(morpholine-4-sulfonylamino)-phenyl]-amide (P-0086),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(tetrahydro-pyran-4-sulfonylamino)-phenyl]-amide (P-0087),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid (3-cyclopentanesulfonylamino-2,6-difluoro-phenyl)-amide (P-0088),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(pyrrolidine-1-sulfonylamino)-phenyl]-amide (P-0089),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(3,3,3-trifluoro-propane-1-sulfonylamino)-phenyl]-amide (P-0090),
    • N-{5-[2,6-Difluoro-3-(propane-1-sulfonylamino)-benzyloxy]-pyridin-2-yl}-acetamide (P-0091),
    • Propane-1-sulfonic acid [3-(2-amino-pyridin-3-yloxymethyl)-2,4-difluoro-phenyl]-amide (P-0092),
    • Propane-1-sulfonic acid [2,4-difluoro-3-(1H-pyrrolo[2,3-b]pyridin-5-yloxymethyl)-phenyl]-amide (P-0093),
    • N-{5-[2,6-Difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzyloxy]-pyridin-2-yl}-acetamide (P-0094),
    • N-[3-(2-Amino-pyridin-3-yloxymethyl)-2,4-difluoro-phenyl]-4-trifluoromethyl-benzenesulfonamide (P-0095),
    • N-[2,4-Difluoro-3-(1H-pyrrolo[2,3-b]pyridin-5-yloxymethyl)-phenyl]-4-trifluoromethyl-benzenesulfonamide (P-0096),
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(2-fluoro-benzenesulfonylamino)-benzamide (P-0097),
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(3-fluoro-benzenesulfonylamino)-benzamide (P-0098),
    • N-(6-Acetylamino-pyridin-3-yl)-3-(2,6-difluoro-benzenesulfonylamino)-2,6-difluoro-benzamide (P-0099),
    • N-(6-Acetylamino-pyridin-3-yl)-3-(2,4-difluoro-benzenesulfonylamino)-2,6-difluoro-benzamide (P-0100),
    • N-(6-Acetylamino-pyridin-3-yl)-3-(2,5-difluoro-benzenesulfonylamino)-2,6-difluoro-benzamide (P-0101),
    • 6-Acetylamino-N-[2,6-difluoro-3-(3-fluoro-4-methoxy-benzenesulfonylamino)-phenyl]-nicotinamide (P-0102),
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzamide (P-0103),
    • N-(6-Acetylamino-pyridin-3-yl)-3-(4-difluoromethoxy-benzenesulfonylamino)-2,6-difluoro-benzamide (P-0104),
    • N-(6-Acetylamino-pyridin-3-yl)-3-(3-difluoromethoxy-benzenesulfonylamino)-2,6-difluoro-benzamide (P-0105),
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(4-isopropyl-benzenesulfonylamino)-benzamide (P-0106),
    • N-(6-Acetylamino-pyridin-3-yl)-3-(4-tert-butyl-benzenesulfonylamino)-2,6-difluoro-benzamide (P-0107),
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(4-propyl-benzenesulfonylamino)-benzamide (P-0108),
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(pyridine-2-sulfonylamino)-benzamide (P-0109),
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(pyridine-3-sulfonylamino)-benzamide (P-0110),
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(dimethylaminosulfonylamino)-benzamide (P-0111),
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(piperidine-1-sulfonylamino)-benzamide (P-0112),
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(morpholine-4-sulfonylamino)-benzamide (P-0113),
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(tetrahydro-pyran-4-sulfonylamino)-benzamide (P-0114),
    • N-(6-Acetylamino-pyridin-3-yl)-3-cyclopentanesulfonylamino-2,6-difluoro-benzamide (P-0115),
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(pyrrolidine-1-sulfonylamino)-benzamide (P-0116),
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(3,3,3-trifluoro-propane-1-sulfonylamino)-benzamide (P-0117),
    • 6-Acetylamino-N-[2,6-difluoro-3-(propane-1-sulfonylamino)-phenyl]-nicotinamide (P-0118),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(propane-1-sulfonylamino)-phenyl]-amide (P-0119),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(propane-1-sulfonylamino)-phenyl]-amide (P-0120), and
      any salt, prodrug, tautomer, or isomer thereof.
  • 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), pharmaceutically acceptable formulations of such compound(s), prodrug(s), and all stereoisomers thereof. In reference to compositions, kits, methods of use, etc. of compounds of Formula I described herein, it is understood (unless indicated otherwise) that a compound of Formula I includes all sub-embodiments thereof (e.g. including Formula Ia, Ib, Ic, Id, Ie, If, Ig, Ih, Ii, and Ij and all embodiments as described above).
  • 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 compound(s) of Formula I. The terms “treat,” “therapy,” and like terms refer to the administration of material, e.g., one or more compound(s) of Formula I, 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, and/or 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 modulators, 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 protein kinase modulator is an inhibitor of the protein kinase. In one aspect, the method involves administering to the subject an effective amount of one or more compound(s) of Formula I 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(s) of Formula I.
  • 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(s) of Formula I. 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, mutations of A-Raf, 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/T529I. 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/T529I mutant B-Raf. The Raf protein kinase mediated disease or condition includes a disease or condition for which Raf modulation provides a therapeutic benefit, e.g. wherein treatment with Raf modulators, 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 Raf modulator is a Raf inhibitor. In one aspect, the method involves administering to the subject an effective amount of a compound of Formula I 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 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 Formula I 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 Ab1, 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 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 Ab1, Akt1, 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 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 Ab1, 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, MAP4K4, MAPKAPK2, Met, p38, PDGFRB, Pim1, PKC theta, Pyk2, Ret, Src, Stk6, TrkA, TrkB, Yes, and Zap70, including any mutations thereof.
  • In some embodiments, a compound of Formula I 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 Ab1, A-Raf, B-Raf, Btk, c-Raf-1, EGFR, EphB2, Erk2, Fak, Fms, Irak4, Jnk1, Jnk2, Jnk3, Kit, Lck, Lyn, MAP2K1, MAP4K4, MAPKAPK2, Met, p38, Pim1, PKC theta, Pyk2, Src, Stk6, TrkA, TrkB, Yes, and Zap70, including any mutations thereof.
  • In some embodiments, a compound of Formula I 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 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 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 A-Raf, B-Raf, c-Raf-1, B-Raf V600E mutant, or B-Raf V600E/T529I mutant. In some embodiments, a compound of Formula I will selectively inhibit one Raf kinase relative to one or more other Raf kinases. In some embodiments, the compound of Formula I 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 of Formula I 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 of Formula I 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. 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(s) of Formula I and at least one pharmaceutically acceptable carrier, excipient, and/or diluent, including combinations of any two or more compounds of Formula I. The composition can further include a plurality of different pharmacologically active compounds, which can include a plurality of compounds of Formula I. In another aspect, the composition can include one or more compounds of Formula I 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 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 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 Ab1, 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, by contacting the protein kinase with an effective amount of one or more compound(s) of Formula I.
  • 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(s) of Formula I.
  • In one aspect, methods are provided for treating a disease or condition mediated by a protein kinase selected from the group consisting of Ab1, 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, ROCK1, ROCK2, Ron, Src, Stk6, Syk, TEC, Tie2, TrkA, TrkB, Yes, and Zap70, including any mutations thereof, by administering to the subject an effective amount of a composition including one or more compound(s) of Formula I.
  • In one aspect, the invention provides methods for treating a disease or condition mediated by a protein kinase selected from the group consisting of Ab1, Akt1, 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, by administering to the subject an effective amount of a composition including one or more compound(s) of Formula I.
  • In one aspect, the invention provides methods for treating a disease or condition mediated by a protein kinase selected from the group consisting of Ab1, 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, MAP4K4, MAPKAPK2, Met, p38, PDGFRB, Pim1, PKC theta, Pyk2, Ret, Src, Stk6, TrkA, TrkB, Yes, and Zap70, including any mutations thereof, by administering to the subject an effective amount of a composition including one or more compound(s) of Formula I.
  • In one aspect, the invention provides methods for treating a disease or condition mediated by a protein kinase selected from the group consisting of Ab1, A-Raf, B-Raf, Btk, c-Raf-1, EGFR, EphB2, Erk2, Fak, Fms, Irak4, Jnk1, Jnk2, Jnk3, Kit, Lck, Lyn, MAP2K1, MAP4K4, MAPKAPK2, Met, p38, Pim1, PKC theta, Pyk2, Src, Stk6, TrkA, TrkB, Yes, and Zap70, including any mutations thereof, by administering to the subject an effective amount of a composition including one or more compound(s) of Formula I.
  • 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, including any mutations thereof, by administering to the subject an effective amount of a composition including one or more compound(s) of Formula I.
  • 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(s) of Formula I. 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(s) of Formula I 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/T529I mutant by administering to the subject an effective amount of a composition including one or more compound(s) of Formula I 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(s) of Formula I, 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, Q-ray, or electron, proton, neutron, or D 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, and 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(s) of Formula I 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-hydroxy-staurosporine), 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-□, 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(s) of Formula I 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(s) of Formula I, a prodrug of such compound, a pharmaceutically acceptable salt of such compound or prodrug, or a pharmaceutically acceptable formulation 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(s) of Formula I, a prodrug of such compound, a pharmaceutically acceptable salt of such compound or prodrug, or a pharmaceutically acceptable formulation 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, 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 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 (e.g. ulcerative colitis, Crohn's disease), systemic lupus erythematosis, Sjogren's Syndrome, Wegener's granulomatosis, 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 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, 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 some aspects, compounds of Formula I can be used in the preparation of a medicament for the treatment of a 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 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 (e.g. ulcerative colitis, Crohn's disease), systemic lupus erythematosis, Sjogren's Syndrome, Wegener's granulomatosis, 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 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, 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, 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. 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; 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 some aspects, compounds of Formula I 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; 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 with kinase activity IC50 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:
    • Ab1, 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; 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 rheumatoid arthritis, systemic lupus erythematosis (SLE), and transplant rejection, inflammatory diseases including inflammatory bowel disease (e.g. ulcerative colitis, Crohn's disease), chronic obstructive pulmonary disease (COPD), emphysema, and atherosclerosis, metabolic disorders, including Type I diabetes, Type II diabetes, insulin resistance, hyperglycemia, and lipolysis, disorders of bone structure, mineralization and bone formation and resorption, including osteoporosis, increased risk of fracture, Paget's disease, hypercalcemia, and metastasis of cancer to bone, kidney diseases, including nephritis (e.g. glomerulonephritis, interstitial nephritis, Lupus nephritis), tubular necrosis, diabetes-associated renal complications (e.g. diabetic nephropathy), and hypertrophy, disorders of the central nervous system, including multiple sclerosis, stroke, Alzheimer's disease and Parkinson's disease; inflammatory and chronic pain, including bone pain; and cancers, including multiple myeloma, acute myeloid leukemia, chronic myeloid leukemia (CML), prostate cancer, breast cancer, ovarian cancer, and metastasis of tumors to other tissues;
    • 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, inflammatory bowel disease (e.g. ulcerative colitis, Crohn's disease), 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 disease (e.g. ulcerative colitis, Crohn's disease), systemic lupus erythematosis, 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 (e.g. 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 disease (e.g. ulcerative colitis, 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, pancreatic, 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 disease (e.g. ulcerative colitis, Crohn's disease), 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 disease (e.g. ulcerative colitis, Crohn's disease) and type I diabetes;
    • Lyn, related to cancers such as glioblastoma.
    • 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, and inflammatory bowel disease (e.g. ulcerative colitis, 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, impulsively, 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, rheumatoid 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 of the Invention 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 chemically feasible and 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. An “optionally substituted lower alkyl” denotes lower alkyl that is optionally 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)2—NH2, —N(H)—C(O)—NH2, —N(H)—C(S)—NH2, —N(H)—S(O)2—NH2, —C(NH)—NH2, —O—Ro, —S—Ro, —O—C(O)—Ro, —O—C(S)—Ro, —C(O)—Ro, —C(S)—Ro, —C(O)—O—Ro, —C(S)—O—Ro, —S(O)—Ro, —S(O)2—Ro, —C(O)—N(H)—Ro, —C(S)—N(H)—Ro, —C(O)—N(Ro)—Ro, —C(S)—N(Ro)—Ro, —S(O)2—N(H)—Ro, —S(O)2—N(Ro)—Ro, —C(NH)—N(H)—Ro, —C(NH)—N(Ro)—Ro, —N(H)—C(O)—Ro, —N(H)—C(S)—Ro, —N(Ro)—C(O)—Ro, —N(Ro)—C(S)—Ro, —N(H)—S(O)2—Ro, —N(Ro)—S(O)2—Ro, —N(H)—C(O)—N(H)—Ro, —N(H)—C(S)—N(H)—Ro, —N(Ro)—C(O)—NH2, —N(Ro)—C(S)—NH2, —N(Ro)—C(O)—N(H)—Ro, —N(Ro)—C(S)—N(H)—Ro, —N(H)—C(O)—N(Ro)—Ro, —N(H)—C(S)—N(Ro)—Ro, —N(Ro)—C(O)—N(Ro)—Ro, —N(Ro)—C(S)—N(Ro)—Ro, —N(H)—S(O)2—N(H)—Ro, —N(Ro)—S(O)2—NH2, —N(Ro)—S(O)2—N(H)—Ro, —N(H)—S(O)2—N(Ro)—Ro, —N(Ro)—S(O)2—N(Ro)—Ro, —N(H)—Ro, —N(Ro)—Ro, —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, 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. It is understood that substitutions are chemically feasible and attached at any available atom to provide 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. The straight chain or branched lower alkenyl group is chemically feasible and attached at any available point to provide a stable compound. Examples of lower alkenyl groups include ethenyl, propenyl, isopropenyl, butenyl, and the like. An “optionally substituted lower alkenyl” denotes lower alkenyl that is optionally 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)2—NH2, —N(H)—C(O)—NH2, —N(H)—C(S)—NH2, —N(H)—S(O)2—NH2, —C(NH)—NH2, —O—Ro, —S—Ro, —O—C(O)—Ro, —O—C(S)—Ro, —C(O)—Ro, —C(S)—Ro, —C(O)—O—Ro, —C(S)—O—Ro, —S(O)—Ro, —S(O)2—Ro, —C(O)—N(H)—Ro, —C(S)—N(H)—Ro, —C(O)—N(Ro)—Ro, —C(S)—N(Ro)—Ro, —S(O)2—N(H)—Ro, —S(O)2—N(Ro)—Ro, —C(NH)—N(H)—Ro, —C(NH)—N(Rp)—Rc, —N(H)—C(O)—Ro, —N(H)—C(S)—Ro, —N(Ro)—C(O)—Ro, —N(Ro)—C(S)—Ro, —N(H)—S(O)2—Ro, —N(Ro)—S(O)2—Ro, —N(H)—C(O)—N(H)—Ro, —N(H)—C(S)—N(H)—Ro, —N(Ro)—C(O)—NH2, —N(Ro)—C(S)—NH2, —N(Ro)—C(O)—N(H)—Ro, —N(Ro)—C(S)—N(H)—Ro, —N(H)—C(O)—N(Ro)—Ro, —N(H)—C(S)—N(Ro)—Ro, —N(Ro)—C(O)—N(Ro)—Ro, —N(Ro)—C(S)—N(Ro)—Ro, —N(H)—S(O)2—N(H)—Ro, —N(Ro)—S(O)2—NH2, —N(Ro)—S(O)2—N(H)—Ro, —N(H)—S(O)2—N(Ro)—Ro, —N(Ro)—S(O)2—N(Ro)—Ro, —N(H)—Ro, —N(Ro)—Ro, —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, 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. “C3-6 alkenyl” denotes lower alkenyl containing 3-6 carbon atoms. An “optionally substituted C3-6 alkenyl” denotes optionally substituted lower alkenyl containing 3-6 carbon atoms. It is understood that substitutions are chemically feasible and attached at any available atom to provide a stable compound.
  • “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. The straight chain or branched lower alkynyl group is chemically feasible and attached at any available point to provide a stable compound. Examples of alkynyl groups include ethynyl, propynyl, butynyl, and the like. An “optionally substituted lower alkynyl” denotes lower alkynyl that is optionally 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)2—NH2, —N(H)—C(O)—NH2, —N(H)—C(S)—NH2, —N(H)—S(O)2—NH2, —C(NH)—NH2, —O—Ro, —S—Ro, —O—C(O)—Ro, —O—C(S)—Ro, —C(O)—Ro, —C(S)—Ro, —C(O)—O—Ro, —C(S)—O—Ro, —S(O)—Ro, —S(O)2—Ro, —C(O)—N(H)—Ro, —C(S)—N(H)—Ro, —C(O)—N(Ro)—Ro, —C(S)—N(Ro)—Ro, —S(O)2—N(H)—Ro, —S(O)2—N(Ro)—Ro, —C(NH)—N(H)—Ro, —C(NH)—N(RP)—Rc, —N(H)—C(O)—Ro, —N(H)—C(S)—Ro, —N(Ro)—C(O)—Ro, —N(Ro)—C(S)—Ro, —N(H)—S(O)2—Ro, —N(Ro)—S(O)2—Ro, —N(H)—C(O)—N(H)—Ro, —N(H)—C(S)—N(H)—Ro, —N(Ro)—C(O)—NH2, —N(Ro)—C(S)—NH2, —N(Ro)—C(O)—N(H)—Ro, —N(Ro)—C(S)—N(H)—Ro, —N(H)—C(O)—N(Ro)—Ro, —N(H)—C(S)—N(Ro)—Ro, —N(Ro)—C(O)—N(Ro)—Ro, —N(Ro)—C(S)—N(Ro)—Ro, —N(H)—S(O)2—N(H)—Ro, —N(Ro)—S(O)2—NH2, —N(Ro)—S(O)2—N(H)—Ro, —N(H)—S(O)2—N(Ro)—Ro, —N(Ro)—S(O)2—N(Ro)—Ro, —N(H)—Ro, —N(Ro)—Ro, —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, 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. “C3-6 alkynyl” denotes lower alkynyl containing 3-6 carbon atoms. An “optionally substituted C3-6 alkynyl” denotes optionally substituted lower alkynyl containing 3-6 carbon atoms. It is understood that substitutions are chemically feasible and attached at any available atom to provide a stable compound.
  • “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. An “optionally substituted cycloalkyl” is a cycloalkyl that is optionally 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)2—NH2, —N(H)—C(O)—NH2, —N(H)—C(S)—NH2, —N(H)—S(O)2—NH2, —C(NH)—NH2, —O—Ro, —S—Ro, —O—C(O)—Ro, —O—C(S)—Ro, —C(O)—Ro, —C(S)—Ro, —C(O)—O—Ro, —C(S)—O—Ro, —S(O)—Ro, —S(O)2—Ro, —C(O)—N(H)—Ro, —C(S)—N(H)—Ro, —C(O)—N(Ro)—Ro, —C(S)—N(Ro)—Ro, —S(O)2—N(H)—Ro, —S(O)2—N(Ro)—Ro, —C(NH)—N(H)—Ro, —C(NH)—N(Rp)—Rc, —N(H)—C(O)—Ro, —N(H)—C(S)—Ro, —N(Ro)—C(O)—Ro, —N(Ro)—C(S)—Ro, —N(H)—S(O)2—Ro, —N(Ro)—S(O)2—Ro, —N(H)—C(O)—N(H)—Ro, —N(H)—C(S)—N(H)—Ro, —N(Ro)—C(O)—NH2, —N(Ro)—C(S)—NH2, —N(Ro)—C(O)—N(H)—Ro, —N(Ro)—C(S)—N(H)—Ro, —N(H)—C(O)—N(R)—Ro, —N(H)—C(S)—N(Ro)—Ro, —N(Ro)—C(O)—N(Ro)—Ro, —N(Ro)—C(S)—N(Ro)—Ro, —N(H)—S(O)2—N(H)—Ro, —N(Ro)—S(O)2—NH2, —N(Ro)—S(O)2—N(H)—Ro, —N(H)—S(O)2—N(Ro)—Ro, —N(Ro)—S(O)2—N(Ro)—Ro, —N(H)—Ro, —N(Ro)—Ro, —Rd, —Re, —Rf, and —Rg. “C3-6 cycloalkyl” denotes cycloalkyl containing 3-6 carbon atoms. “C3-5 cycloalkyl” denotes cycloalkyl containing 3-5 carbon atoms. It is understood that substitutions are chemically feasible and attached at any available atom to provide a stable compound.
  • “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. “Nitrogen containing heterocycloalkyl” refers to heterocycloalkyl wherein at least one heteroatom is N. An “optionally substituted heterocycloalkyl” is a heterocycloalkyl that is optionally 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)2—NH2, —N(H)—C(O)—NH2, —N(H)—C(S)—NH2, —N(H)—S(O)2—NH2, —C(NH)—NH2, —O—Ro, —S—Ro, —O—C(O)—Ro, —O—C(S)—Ro, —C(O)—Ro, —C(S)—Ro, —C(O)—O—Ro, —C(S)—O—Ro, —S(O)—Ro, —S(O)2—Ro, —C(O)—N(H)—Ro, —C(S)—N(H)—Ro, —C(O)—N(Ro)—Ro, —C(S)—N(Ro)—Ro, —S(O)2—N(H)—Ro, —S(O)2—N(Ro)—Ro, —C(NH)—N(H)—Ro, —C(NH)—N(RP)—Rc, —N(H)—C(O)—Ro, —N(H)—C(S)—Ro, —N(Ro)—C(O)—Ro, —N(Ro)—C(S)—Ro, —N(H)—S(O)2—Ro, —N(Ro)—S(O)2—Ro, —N(H)—C(O)—N(H)—Ro, —N(H)—C(S)—N(H)—Ro, —N(Ro)—C(O)—NH2, —N(Ro)—C(S)—NH2, —N(Ro)—C(O)—N(H)—Ro, —N(Ro)—C(S)—N(H)—Ro, —N(H)—C(O)—N(Ro)—Ro, —N(H)—C(S)—N(Ro)—Ro, —N(Ro)—C(O)—N(Ro)—Ro, —N(Ro)—C(S)—N(Ro)—Ro, —N(H)—S(O)2—N(H)—Ro, —N(Ro)—S(O)2—NH2, —N(Ro)—S(O)2—N(H)—Ro, —N(H)—S(O)2—N(Ro)—Ro, —N(Ro)—S(O)2—N(Ro)—Ro, —N(H)—Ro, —N(Ro)—Ro, —Rd, —Re, —Rf, and —Rg. It is understood that substitutions are chemically feasible and attached at any available atom to provide a stable compound.
  • “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. An “optionally substituted aryl” is an aryl that is optionally 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)2—NH2, —N(H)—C(O)—NH2, —N(H)—C(S)—NH2, —N(H)—S(O)2—NH2, —C(NH)—NH2, —O—Ro, —S—Ro, —O—C(O)—Ro, —O—C(S)—Ro, —C(O)—Ro, —C(S)—Ro, —C(O)—O—Ro, —C(S)—O—Ro, —S(O)—Ro, —S(O)2—Ro, —C(O)—N(H)—Ro, —C(S)—N(H)—Ro, —C(O)—N(Ro)—Ro, —C(S)—N(Ro)—Ro, —S(O)2—N(H)—Ro, —S(O)2—N(R)—R, —C(NH)—N(H)—Ro, —C(NH)—N(RP)—Rc, —N(H)—C(O)—Ro, —N(H)—C(S)—Ro, —N(Ro)—C(O)—Ro, —N(Ro)—C(S)—Ro, —N(H)—S(O)2—Ro, —N(Ro)—S(O)2—Ro, —N(H)—C(O)—N(H)—Ro, —N(H)—C(S)—N(H)—Ro, —N(Ro)—C(O)—NH2, —N(Ro)—C(S)—NH2, —N(Ro)—C(O)—N(H)—Ro, —N(Ro)—C(S)—N(H)—Ro, —N(H)—C(O)—N(Ro)—Ro, —N(H)—C(S)—N(Ro)—Ro, —N(Ro)—C(O)—N(Ro)—Ro, —N(Ro)—C(S)—N(Ro)—Ro, —N(H)—S(O)2—N(H)—Ro, —N(Ro)—S(O)2—NH2, —N(Ro)—S(O)2—N(H)—Ro, —N(H)—S(O)2—N(Ro)—Ro, —N(Ro)—S(O)2—N(Ro)—Ro, —N(H)—Ro, —N(Ro)—Ro, —Rd, —Re, —Rf, and —Rg. It is understood that substitutions are chemically feasible and attached at any available atom to provide a stable compound.
  • “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 at least one heteroatom is N. In some instances, for example when R groups of a nitrogen combine with the nitrogen to form a 5 or 7 membered nitrogen containing heteroaryl, any heteroatoms in such 5 or 7 membered heteroaryl are N. An “optionally substituted heteroaryl” is a heteroaryl that is optionally 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)2—NH2, —N(H)—C(O)—NH2, —N(H)—C(S)—NH2, —N(H)—S(O)2—NH2, —C(NH)—NH2, —O—Ro, —S—Ro, —O—C(O)—Ro, —O—C(S)—Ro, —C(O)—Ro, —C(S)—Ro, —C(O)—O—Ro, —C(S)—O—Ro, —S(O)—Ro, —S(O)2—Ro, —C(O)—N(H)—Ro, —C(S)—N(H)—Ro, —C(O)—N(Ro)—Ro, —C(S)—N(Ro)—Ro, —S(O)2—N(H)—Ro, —S(O)2—N(Ro)—Ro, —C(NH)—N(H)—Ro, —C(NH)—N(Rp)—Rc, —N(H)—C(O)—Ro, —N(H)—C(S)—Ro, —N(Ro)—C(O)—Ro, —N(Ro)—C(S)—Ro, —N(H)—S(O)2—Ro, —N(Ro)—S(O)2—Ro, —N(H)—C(O)—N(H)—Ro, —N(H)—C(S)—N(H)—Ro, —N(Ro)—C(O)—NH2, —N(Ro)—C(S)—NH2, —N(Ro)—C(O)—N(H)—Ro, —N(Ro)—C(S)—N(H)—Ro, —N(H)—C(O)—N(Ro)—Ro, —N(H)—C(S)—N(Ro)—Ro, —N(Ro)—C(O)—N(Ro)—Ro, —N(Ro)—C(S)—N(Ro)—Ro, —N(H)—S(O)2—N(H)—Ro, —N(Ro)—S(O)2—NH2, —N(Ro)—S(O)2—N(H)—Ro, —N(H)—S(O)2—N(Ro)—Ro, —N(Ro)—S(O)2—N(Ro)—Ro, —N(H)—Ro, —N(Ro)—Ro, —Rd, —Re, —Rf, and —Rg. It is understood that substitutions are chemically feasible and attached at any available atom to provide a stable compound.
  • The variables Ro, Rp, Rc, 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, —O—Ru, —S—Ru, —N(H)—Ru, —N(Ru)—Ru, —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)2—NH2, —N(H)—C(O)—NH2, —N(H)—C(S)—NH2, —N(H)—S(O)2—NH2, —C(NH)—NH2, —O—Rk, —S—Rk, —O—C(O)—Rk, —O—C(S)—Rk, —C(O)—Rk, —C(S)—Rk, —C(O)—O—Rk, —C(S)—O—Rk, —S(O)—Rk, —S(O)2—Rk, —C(O)—N(H)—Rk, —C(S)—N(H)—Rk, —C(O)—N(Rk)—Rk, —C(S)—N(Rk)—Rk, —S(O)2—N(H)—Rk, —S(O)2—N(Rk)—Rk, —C(NH)—N(H)—Rk, —C(NH)—N(Rm)—Rn, —N(H)—C(O)—Rk, —N(H)—C(S)—Rk, —N(Rk)—C(O)—Rk, —N(Rk)—C(S)—Rk, —N(H)—S(O)2—Rk, —N(Rk)—S(O)2—Rk, —N(H)—C(O)—N(H)—Rk, —N(H)—C(S)—N(H)—Rk, —N(Rk)—C(O)—NH2, —N(Rk)—C(S)—NH2, —N(Rk)—C(O)—N(H)—Rk, —N(Rk)—C(S)—N(H)—Rk, —N(H)—C(O)—N(Rk)—Rk, —N(H)—C(S)—N(Rk)—Rk, —N(Rk)—C(O)—N(Rk)—Rk, —N(Rk)—C(S)—N(Rk)—Rk, —N(H)—S(O)2—N(H)—Rk, —N(Rk)—S(O)2—NH2, —N(Rk)—S(O)2—N(H)—Rk, —N(H)—S(O)2—N(Rk)—Rk, —N(Rk)—S(O)2—N(Rk)—Rk, —N(H)—Rk, —N(Rk)—Rk, —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)2—NH2, —N(H)—C(O)—NH2, —N(H)—C(S)—NH2, —N(H)—S(O)2—NH2, —C(NH)—NH2, —O—Rk, —S—Rk, —O—C(O)—Rk, —O—C(S)—Rk, —C(O)—Rk, —C(S)—Rk, —C(O)—O—Rk, —C(S)—O—Rk, —S(O)—Rk, —S(O)2—Rk, —C(O)—N(H)—Rk, —C(S)—N(H)—Rk, —C(O)—N(Rk)—Rk, —C(S)—N(Rk)—Rk, —S(O)2—N(H)—Rk, —S(O)2—N(Rk)—Rk, —C(NH)—N(H)—Rk, —C(NH)—N(Rm)—Rn, —N(H)—C(O)—Rk, —N(H)—C(S)—Rk, —N(Rk)—C(O)—Rk, —N(Rk)—C(S)—Rk, —N(H)—S(O)2—Rk, —N(Rk)—S(O)2—Rk, —N(H)—C(O)—N(H)—Rk, —N(H)—C(S)—N(H)—Rk, —N(Rk)—C(O)—NH2, —N(Rk)—C(S)—NH2, —N(Rk)—C(O)—N(H)—Rk, —N(Rk)—C(S)—N(H)—Rk, —N(H)—C(O)—N(Rk)—Rk, —N(H)—C(S)—N(Rk)—Rk, —N(Rk)—C(O)—N(Rk)—Rk, —N(Rk)—C(S)—N(Rk)—Rk, —N(H)—S(O)2—N(H)—Rk, —N(Rk)—S(O)2—NH2, —N(Rk)—S(O)2—N(H)—Rk, —N(H)—S(O)2—N(Rk)—Rk, —N(Rk)—S(O)2—N(Rk)—Rk, —N(H)—Rk, —N(Rk)—Rk, —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)2—NH2, —N(H)—C(O)—NH2, —N(H)—C(S)—NH2, —N(H)—S(O)2—NH2, —C(NH)—NH2, —O—Rk, —S—Rk, —O—C(O)—Rk, —O—C(S)—Rk, —C(O)—Rk, —C(S)—Rk, —C(O)—O—Rk, —C(S)—O—Rk, —S(O)—Rk, —S(O)2—Rk, —C(O)—N(H)—Rk, —C(S)—N(H)—Rk, —C(O)—N(Rk)—Rk, —C(S)—N(Rk)—Rk, —S(O)2—N(H)—Rk, —S(O)2—N(Rk)—Rk, —C(NH)—N(H)—Rk, —C(NH)—N(Rm)—Rn, —N(H)—C(O)—Rk, —N(H)—C(S)—Rk, —N(Rk)—C(O)—Rk, —N(Rk)—C(S)—Rk, —N(H)—S(O)2—Rk, —N(Rk)—S(O)2—Rk, —N(H)—C(O)—N(H)—Rk, —N(H)—C(S)—N(H)—Rk, —N(Rk)—C(O)—NH2, —N(Rk)—C(S)—NH2, —N(Rk)—C(O)—N(H)—Rk, —N(Rk)—C(S)—N(H)—Rk, —N(H)—C(O)—N(Rk)—Rk, —N(H)—C(S)—N(Rk)—Rk, —N(Rk)—C(O)—N(Rk)—Rk, —N(Rk)—C(S)—N(Rk)—Rk, —N(H)—S(O)2—N(H)—Rk, —N(Rk)—S(O)2—NH2, —N(Rk)—S(O)2—N(H)—Rk, —N(H)—S(O)2—N(Rk)—Rk, —N(Rk)—S(O)2—N(Rk)—Rk, —N(H)—Rk, —N(Rk)—Rk, —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)2—NH2, —N(H)—C(O)—NH2, —N(H)—C(S)—NH2, —N(H)—S(O)2—NH2, —C(NH)—NH2, —O—Rk, —S—Rk, —O—C(O)—Rk, —O—C(S)—Rk, —C(O)—Rk, —C(S)—Rk, —C(O)—O—Rk, —C(S)—O—Rk, —S(O)—Rk, —S(O)2—Rk, —C(O)—N(H)—Rk, —C(S)—N(H)—Rk, —C(O)—N(Rk)—Rk, —C(S)—N(Rk)—Rk, —S(O)2—N(H)—Rk, —S(O)2—N(Rk)—Rk, —C(NH)—N(H)—Rk, —C(NH)—N(Rm)—Rn, —N(H)—C(O)—Rk, —N(H)—C(S)—Rk, —N(Rk)—C(O)—Rk, —N(Rk)—C(S)—Rk, —N(H)—S(O)2—Rk, —N(Rk)—S(O)2—Rk, —N(H)—C(O)—N(H)—Rk, —N(H)—C(S)—N(H)—Rk, —N(Rk)—C(O)—NH2, —N(Rk)—C(S)—NH2, —N(Rk)—C(O)—N(H)—Rk, —N(Rk)—C(S)—N(H)—Rk, —N(H)—C(O)—N(Rk)—Rk, —N(H)—C(S)—N(Rk)—Rk, —N(Rk)—C(O)—N(Rk)—Rk, —N(Rk)—C(S)—N(Rk)—Rk, —N(H)—S(O)2—N(H)—Rk, —N(Rk)—S(O)2—NH2, —N(Rk)—S(O)2—N(H)—Rk, —N(H)—S(O)2—N(Rk)—Rk, —N(Rk)—S(O)2—N(Rk)—Rk, —N(H)—Rk, —N(Rk)—Rk, —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 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, O—Ru, —S—Ru, —N(H)—Ru, —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)2—NH2, —N(H)—C(O)—NH2, —N(H)—C(S)—NH2, —N(H)—S(O)2—NH2, —C(NH)—NH2, —O—Rr, —S—Rr, —O—C(O)—Rr, —O—C(S)—Rr, —C(O)—Rr, —C(S)—Rr, —C(O)—O—Rr, —C(S)—O—Rr, —S(O)—Rr, —S(O)2—Rr, —C(O)—N(H)—Rr, —C(S)—N(H)—Rr, —C(O)—N(Rr)—Rr, —C(S)—N(Rr)—Rr, —S(O)2—N(H)—Rr, —S(O)2—N(Rr)—Rr, —C(NH)—N(H)—Rr, —C(NH)—N(Rs)—Rt, —N(H)—C(O)—Rr, —N(H)—C(S)—Rr, —N(Rr)—C(O)—Rr, —N(Rr)—C(S)—Rr, —N(H)—S(O)2—Rr, —N(Rr)—S(O)2—Rr, —N(H)—C(O)—N(H)—Rr, —N(H)—C(S)—N(H)—Rr, —N(Rr)—C(O)—NH2, —N(Rr)—C(S)—NH2, —N(Rr)—C(O)—N(H)—Rr, —N(Rr)—C(S)—N(H)—Rr, —N(H)—C(O)—N(Rr)—Rr, —N(H)—C(S)—N(Rr)—Rr, —N(Rr)—C(O)—N(Rr)—Rr, —N(Rr)—C(S)—N(Rr)—Rr, —N(H)—S(O)2—N(H)—Rr, —N(Rr)—S(O)2—NH2, —N(Rr)—S(O)2—N(H)—Rr, —N(H)—S(O)2—N(Rr)—Rr, —N(Rr)—S(O)2—N(Rr)—Rr, —N(H)—Rr, —N(Rr)—Rr, —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)2—NH2, —N(H)—C(O)—NH2, —N(H)—C(S)—NH2, —N(H)—S(O)2—NH2, —C(NH)—NH2, —O—Rr, —S—Rr, —O—C(O)—Rr, —O—C(S)—Rr, —C(O)—Rr, —C(S)—Rr, —C(O)—O—Rr, —C(S)—O—Rr, —S(O)—Rr, —S(O)2—Rr, —C(O)—N(H)—Rr, —C(S)—N(H)—Rr, —C(O)—N(Rr)—Rr, —C(S)—N(Rr)—Rr, —S(O)2—N(H)—Rr, —S(O)2—N(Rr)—Rr, —C(NH)—N(H)—Rr, —C(NH)—N(Rs)—Rt, —N(H)—C(O)—Rr, —N(H)—C(S)—Rr, —N(Rr)—C(O)—Rr, —N(Rr)—C(S)—Rr, —N(H)—S(O)2—Rr, —N(Rr)—S(O)2—Rr, —N(H)—C(O)—N(H)—Rr, —N(H)—C(S)—N(H)—Rr, —N(Rr)—C(O)—NH2, —N(Rr)—C(S)—NH2, —N(Rr)—C(O)—N(H)—Rr, —N(Rr)—C(S)—N(H)—Rr, —N(H)—C(O)—N(Rr)—Rr, —N(H)—C(S)—N(Rr)—Rr, —N(Rr)—C(O)—N(Rr)—Rr, —N(Rr)—C(S)—N(Rr)—Rr, —N(H)—S(O)2—N(H)—Rr, —N(Rr)—S(O)2—NH2, —N(Rr)—S(O)2—N(H)—Rr, —N(H)—S(O)2—N(Rr)—Rr, —N(Rr)—S(O)2—N(Rr)—Rr, —N(H)—Rr, —N(Rr)—r, 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)2—NH2, —N(H)—C(O)—NH2, —N(H)—C(S)—NH2, —N(H)—S(O)2—NH2, —C(NH)—NH2, —O—Rr, —S—Rr, —O—C(O)—Rr, —O—C(S)—Rr, —C(O)—Rr, —C(S)—Rr, —C(O)—O—Rr, —C(S)—O—Rr, —S(O)—Rr, —S(O)2—Rr, —C(O)—N(H)—Rr, —C(S)—N(H)—Rr, —C(O)—N(Rr)—Rr, —C(S)—N(Rr)—Rr, —S(O)2—N(H)—Rr, —S(O)2—N(Rr)—Rr, —C(NH)—N(H)—Rr, —C(NH)—N(Rs)—Rt, —N(H)—C(O)—Rr, —N(H)—C(S)—Rr, —N(Rr)—C(O)—Rr, —N(Rr)—C(S)—Rr, —N(H)—S(O)2—Rr, —N(Rr)—S(O)2—Rr, —N(H)—C(O)—N(H)—Rr, —N(H)—C(S)—N(H)—Rr, —N(Rr)—C(O)—NH2, —N(Rr)—C(S)—NH2, —N(Rr)—C(O)—N(H)—Rr, —N(Rr)—C(S)—N(H)—Rr, —N(H)—C(O)—N(Rr)—Rr, —N(H)—C(S)—N(Rr)—Rr, —N(Rr)—C(O)—N(Rr)—Rr, —N(Rr)—C(S)—N(Rr)—Rr, —N(H)—S(O)2—N(H)—Rr, —N(Rr)—S(O)2—NH2, —N(Rr)—S(O)2—N(H)—Rr, —N(H)—S(O)2—N(Rr)—Rr, —N(Rr)—S(O)2—N(Rr)—Rr, —N(H)—Rr, —N(Rr)—Rr, 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, 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, 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 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, O—Ru, —S—Ru, —N(H)—Ru, —N(Ru)—Ru, —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, 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, 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 lower alkenyl, optionally substituted C3-6 alkenyl, optionally substituted lower alkynyl, or optionally substituted C3-6 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, —O—R1a, —S—R1a, —N(R1a)—R1a, —O—C(O)—R1a, —O—C(S)—R1a, —C(O)—R1a, —C(S)—R1a, —C(O)—O—R1a, —C(S)—O—R1a, —C(O)—N(R1a)—R1a, —C(S)—N(R1a)—R1a, —S(O)2—N(R1a)—R1a, —C(NH)—N(R1a)—R1a, —N(R1a)—C(O)—R1a, —N(R1a)—C(S)—R1a, —N(R1a)—S(O)2—R1a, —N(R1a)—C(O)—N(R1a)—R1a, —N(R1a)—C(S)—N(R1a)—R1a, —N(R1a)—S(O)2—N(R1a)—R1a, —S(O)—R1a, —S(O)2—R1a, 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, —O—R1a, —S—R1a, —N(R1a)—R1a, —O—C(O)—R1a, —O—C(S)—R1a, —C(O)—R1a, —C(S)—R1a, —C(O)—O—R1a, —C(S)—O—R1a, —C(O)—N(R1a)—R1a, —C(S)—N(R1a)—R1a, —S(O)2—N(R1a)—R1a, —C(NH)—N(R1a)—R1a, —N(R1a)—C(O)—R1a, —N(R1a)—C(S)—R1a, —N(R1a)—S(O)2—R1a, —N(R1a)—C(O)—N(R1a)—R1a, —N(R1a)—C(S)—N(R1a)—R1a, —N(R1a)—S(O)2—N(R1a)—R1a, —S(O)—R1a, —S(O)2—R1a, —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 heteroaryl, 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, —O—R1a, —S—R1a, —N(R1a)—R1a, —O—C(O)—R1a, —O—C(S)—R1a, —C(O)—R1a, —C(S)—R1a, —C(O)—O—R1a, —C(S)—O—R1a, —C(O)—N(R1a)—R1a, —C(S)—N(R1a)—R1a, —S(O)2—N(R1a)—R1a, —C(NH)—N(R1a)—R1a, —N(R1a)—C(O)—R1a, —N(R1a)—C(S)—R1a, —N(R1a)—S(O)2—R1a, —N(R1a)—C(O)—N(R1a)—Ra, —N(R1a)—C(S)—N(R1a)—Ra, —N(R1a)—S(O)2—N(R1a)—R1a, —S(O)—R1a, —S(O)2—R1a, —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, —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 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 lower alkenyl, optionally substituted C3-6 alkenyl, optionally substituted lower alkynyl, or optionally substituted C3-6 alkynyl are optionally substituted with one or more, also 1, 2 or 3 groups or substituents selected from the group consisting of fluoro, —CN, —O—R1a, —S—R1a, —N(R1a)—R1a, —C(O)—R1a, —C(S)—R1a, —C(O)—O—R1a, —C(O)—N(R1a)—R1a, —C(S)—N(R1a)—R1a, —S(O)2—N(R1a)—R1a, —N(R1a)—C(O)—R1a, —N(R1a)—C(S)—R1a, —N(R1a)—S(O)2—R1a, —S(O)—R1a, —S(O)2—R1a, 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, —O—R1a, —S—R1a, —N(R1a)—R1a, —C(O)—R1a, —C(S)—R1a, —C(O)—O—R1a, —C(O)—N(R1a)—R1a, —C(S)—N(R1a)—R1a, —S(O)2—N(R1a)—R1a, —N(R1a)—C(O)—R1a, —N(R1a)—C(S)—R1a, —N(R1a)—S(O)2—R1a, —S(O)—R1a, —S(O)2—R1a, —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 heteroaryl, 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, —O—R1a, —S—R1a, —N(R1a)—R1a, —C(O)—R1a, —C(S)—R1a, —C(O)—O—R1a, —C(O)—N(R1a)—R1a, —C(S)—N(R1a)—R1a, —S(O)2—N(R1a)—R1a, —N(R1a)—C(O)—R1a, —N(R1a)—C(S)—R1a, —N(R1a)—S(O)2—R1a, —S(O)—R1a, —S(O)2—R1a, —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, —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 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, including descriptions of substituted cycloalkyl, heterocycloalkyl, aryl and heteroaryl, attached at any available atom to provide 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. It is understood that substitutions on alkoxy are chemically feasible and attached at any available atom to provide a stable compound.
  • “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, including descriptions of substituted cycloalkyl, heterocycloalkyl, aryl and heteroaryl, attached at any available atom to provide 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. It is understood that substitutions on alkylthio are chemically feasible and attached at any available atom to provide a stable compound.
  • “Amino” or “amine” denotes the group —NH2. “Mono-alkylamino” denotes the group —NHRbb where Rbb is lower alkyl. “Di-alkylamino” denotes the group —NRbbRcc, where Rbb 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. It is understood that when mono-alkylamino, di-alkylamino, or cycloalkylamino are substituents on other moieties, these are chemically feasible and attached at any available atom to provide a stable compound.
  • 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. The term “inhibitor” refers to a modulator that decreases the activity of the target biomolecule. 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.
  • 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 (i.e. selectivity) 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, 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 Ab1, 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, and any mutations thereof, 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. Specific target protein kinases contemplated by the present invention are described in the art, including, without limitation, protein kinases 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, including all specifications, figures, and tables, and for all purposes, as well as the following:
  • 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), Cl (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 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).
  • Raf inhibitors (A-Raf and/or B-Raf and/or c-Raf-1) 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.
  • 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.
  • 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, include, for example, (a) prodrugs, and active metabolites (b) tautomers, isomers (including stereoisomers and regioisomers), and racemic mixtures (c) pharmaceutically acceptable salts and formulations and (d) solid forms, including different crystal forms, polymorphic or amorphous solids, including hydrates and solvates thereof, and other forms.
  • (a) Prodrugs and Metabolites
  • In addition to the present formulae and compounds described herein, the invention also includes prodrugs (generally pharmaceutically acceptable prodrugs), active metabolic derivatives (active metabolites), and their pharmaceutically acceptable salts.
  • Prodrugs are compounds or pharmaceutically acceptable salts thereof which, when metabolized under physiological conditions or when converted by solvolysis, yield the desired active compound. Prodrugs include, without limitation, esters, amides, carbamates, carbonates, ureides, solvates, or hydrates of the active compound. Typically, the prodrug is inactive, or less active than the active compound, but may provide one or more advantageous handling, administration, and/or metabolic properties. For example, some prodrugs are esters of the active compound; during metabolysis, the ester group is cleaved to yield the active drug. Esters include, for example, esters of a carboxylic acid group, or S-acyl or O-acyl derivatives of thiol, alcohol, or phenol groups. In this context, a common example is an alkyl ester of a carboxylic acid. Some prodrugs are activated enzymatically to yield the active compound, or a compound which, upon further chemical reaction, yields the active compound. Prodrugs may proceed from prodrug form to active form in a single step or may have one or more intermediate forms which may themselves have activity or may be inactive.
  • As described in The Practice of Medicinal Chemistry, Ch. 31-32 (Ed. Wermuth, Academic Press, San Diego, Calif., 2001), prodrugs can be conceptually divided into two non-exclusive categories, bioprecursor prodrugs and carrier prodrugs. Generally, bioprecursor prodrugs are compounds that are inactive or have low activity compared to the corresponding active drug compound, that contain one or more protective groups and are converted to an active form by metabolism or solvolysis. Both the active drug form and any released metabolic products should have acceptably low toxicity. Typically, the formation of active drug compound involves a metabolic process or reaction that is one of the follow types:
  • Oxidative reactions: Oxidative reactions are exemplified without limitation to reactions such as oxidation of alcohol, carbonyl, and acid functionalities, hydroxylation of aliphatic carbons, hydroxylation of alicyclic carbon atoms, oxidation of aromatic carbon atoms, oxidation of carbon-carbon double bonds, oxidation of nitrogen-containing functional groups, oxidation of silicon, phosphorus, arsenic, and sulfur, oxidative N-dealkylation, oxidative O- and S-dealkylation, oxidative deamination, as well as other oxidative reactions.
  • Reductive reactions: Reductive reactions are exemplified without limitation to reactions such as reduction of carbonyl functionalitites, reduction of alcohol functionalities and carbon-carbon double bonds, reduction of nitrogen-containing functional groups, and other reduction reactions.
  • Reactions without change in the oxidation state: Reactions without change in the state of oxidation are exemplified without limitation to reactions such as hydrolysis of esters and ethers, hydrolytic cleavage of carbon-nitrogen single bonds, hydrolytic cleavage of non-aromatic heterocycles, hydration and dehydration at multiple bonds, new atomic linkages resulting from dehydration reactions, hydrolytic dehalogenation, removal of hydrogen halide molecule, and other such reactions.
  • Carrier prodrugs are drug compounds that contain a transport moiety, e.g., that improves uptake and/or localized delivery to a site(s) of action. Desirably for such a carrier prodrug, the linkage between the drug moiety and the transport moiety is a covalent bond, the prodrug is inactive or less active than the drug compound, the prodrug and any release transport moiety are acceptably non-toxic. For prodrugs where the transport moiety is intended to enhance uptake, typically the release of the transport moiety should be rapid. In other cases, it is desirable to utilize a moiety that provides slow release, e.g., certain polymers or other moieties, such as cyclodextrins. (See, e.g., Cheng et al., U.S. Patent Publ. No. 20040077595, application Ser. No. 10/656,838, incorporated herein by reference.) Such carrier prodrugs are often advantageous for orally administered drugs. In some instances, the transport moiety provides targeted delivery of the drug, for example the drug maybe conjugated to an antibody or antibody fragment. Carrier prodrugs can, for example, be used to improve one or more of the following properties: increased lipophilicity, increased duration of pharmacological effects, increased site-specificity, decreased toxicity and adverse reactions, and/or improvement in drug formulation (e.g., stability, water solubility, suppression of an undesirable organoleptic or physiochemical property). For example, lipophilicity can be increased by esterification of hydroxyl groups with lipophilic carboxylic acids, or of carboxylic acid groups with alcohols, e.g., aliphatic alcohols. Wermuth, supra.
  • Metabolites, e.g., active metabolites, overlap with prodrugs as described above, e.g., bioprecursor prodrugs. Thus, such metabolites are pharmacologically active compounds or compounds that further metabolize to pharmacologically active compounds that are derivatives resulting from metabolic processes in the body of a subject. Of these, active metabolites are such pharmacologically active derivative compounds. For prodrugs, the prodrug compound is generally inactive or of lower activity than the metabolic product. For active metabolites, the parent compound may be either an active compound or may be an inactive prodrug. For example, in some compounds, one or more alkoxy groups can be metabolized to hydroxyl groups while retaining pharmacologic activity and/or carboxyl groups can be esterified, e.g., glucuronidation. In some cases, there can be more than one metabolite, where an intermediate metabolite(s) is further metabolized to provide an active metabolite. For example, in some cases a derivative compound resulting from metabolic glucuronidation may be inactive or of low activity, and can be further metabolized to provide an active metabolite.
  • Metabolites of a compound may be identified using routine techniques known in the art, and their activities determined using tests such as those described herein. See, e.g., Bertolini et al., 1997, J. Med. Chem., 40:2011-2016; Shan et al., 1997, J Pharm Sci 86(7):756-757; Bagshawe, 1995, Drug Dev. Res., 34:220-230; Wermuth, supra.
  • (b) Tautomers, Stereoisomers, and Regioisomers
  • It is understood that some compounds may exhibit tautomerism. In such cases, the formulae provided herein expressly depict only one of the possible tautomeric forms. It is therefore to be understood that the formulae provided herein are intended to represent any tautomeric form of the depicted compounds and are not to be limited merely to the specific tautomeric form depicted by the drawings of the formulae.
  • Likewise, some of the compounds according to the present invention may exist as stereoisomers, i.e. having the same atomic connectivity of covalently bonded atoms yet differing in the spatial orientation of the atoms. For example, compounds may be optical stereoisomers, which contain one or more chiral centers, and therefore, may exist in two or more stereoisomeric forms (e.g. enantiomers or diastereomers). Thus, such compounds may be present as single stereoisomers (i.e., essentially free of other stereoisomers), racemates, and/or mixtures of enantiomers and/or diastereomers. As another example, stereoisomers include geometric isomers, such as cis- or trans-orientation of substituents on adjacent carbons of a double bond. All such single stereoisomers, racemates and mixtures thereof are intended to be within the scope of the present invention. Unless specified to the contrary, all such steroisomeric forms are included within the formulae provided herein.
  • In some embodiments, a chiral compound of the present invention is in a form that contains at least 80% of a single isomer (60% enantiomeric excess (“e.e.”) or diastereomeric excess (“d.e.”)), or at least 85% (70% e.e. or d.e.), 90% (80% e.e. or d.e.), 95% (90% e.e. or d.e.), 97.5% (95% e.e. or d.e.), or 99% (98% e.e. or d.e.). As generally understood by those skilled in the art, an optically pure compound having one chiral center is one that consists essentially of one of the two possible enantiomers (i.e., is enantiomerically pure), and an optically pure compound having more than one chiral center is one that is both diastereomerically pure and enantiomerically pure. In some embodiments, the compound is present in optically pure form, such optically pure form being prepared and/or isolated by methods known in the art (e.g. by recrystallization techniques, chiral synthetic techniques (including synthesis from optically pure starting materials), and chromatographic separation using a chiral column.
  • (c) Pharmaceutically Acceptable Salts and Formulations
  • Unless specified to the contrary, specification of a compound herein includes pharmaceutically acceptable salts of such compound. Thus, compounds of Formula I can be in the form of pharmaceutically acceptable salts, or can be formulated as pharmaceutically acceptable salts. Contemplated pharmaceutically acceptable salt forms include, without limitation, mono, bis, tris, tetrakis, and so on. Pharmaceutically acceptable salts are non-toxic in the amounts and concentrations at which they are administered. The preparation of such salts can facilitate the pharmacological use by altering the physical characteristics of a compound without preventing it from exerting its physiological effect. Useful alterations in physical properties include lowering the melting point to facilitate transmucosal administration and increasing the solubility to facilitate administering higher concentrations of the drug. A compound of the invention may possess a sufficiently acidic, a sufficiently basic, or both functional groups, and accordingly react with any of a number of inorganic or organic bases, and inorganic and organic acids, to form a pharmaceutically acceptable salt.
  • Pharmaceutically acceptable salts include acid addition salts such as those containing chloride, bromide, iodide, hydrochloride, acetate, phenylacetate, acrylate, ascorbate, aspartate, benzoate, 2-phenoxybenzoate, 2-acetoxybenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, methylbenzoate, bicarbonate, butyne-1,4 dioate, hexyne-1,6-dioate, caproate, caprylate, chlorobenzoate, cinnamate, citrate, decanoate, formate, fumarate, glycolate, gluconate, glucarate, glucuronate, glucose-6-phosphate, glutamate, heptanoate, hexanoate, isethionate, isobutyrate, gamma-hydroxybutyrate, phenylbutyrate, lactate, malate, maleate, hydroxymaleate, methylmaleate, malonate, mandelate, nicotinate, nitrate, isonicotinate, octanoate, oleate, oxalate, pamoate, phosphate, monohydrogenphosphate, dihydrogenphosphate, orthophosphate, metaphosphate, pyrophosphate, 2-phosphoglycerate, 3-phosphoglycerate, phthalate, propionate, phenylpropionate, propiolate, pyruvate, quinate, salicylate, 4-aminosalicylate, sebacate, stearate, suberate, succinate, sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, sulfamate, sulfonate, benzenesulfonate (i.e. besylate), ethanesulfonate (i.e. esylate), ethane-1,2-disulfonate, 2-hydroxyethanesulfonate (i.e. isethionate), methanesulfonate (i.e. mesylate), naphthalene-1-sulfonate, naphthalene-2-sulfonate (i.e. napsylate), propanesulfonate, p-toluenesulfonate (i.e. tosylate), xylenesulfonates, cyclohexylsulfamate, tartrate, and trifluoroacetate. These pharmaceutically acceptable acid addition salts can be prepared using the appropriate corresponding acid.
  • When acidic functional groups, such as carboxylic acid or phenol are present, pharmaceutically acceptable salts also include basic addition salts such as those containing benzathine, chloroprocaine, choline, ethanolamine, diethanolamine, triethanolamine, t-butylamine, dicyclohexylamine, ethylenediamine, N,N′-dibenzylethylenediamine, meglumine, hydroxyethylpyrrolidine, piperidine, morpholine, piperazine, procaine, aluminum, calcium, copper, iron, lithium, magnesium, manganese, potassium, sodium, zinc, ammonium, and mono-, di-, or tri-alkylamines, or salts derived from amino acids such as L-histidine, L-glycine, L-lysine, and L-arginine. For example, see Remington's Pharmaceutical Sciences, 19th ed., Mack Publishing Co., Easton, Pa., Vol. 2, p. 1457, 1995. These pharmaceutically acceptable base addition salts can be prepared using the appropriate corresponding base.
  • Pharmaceutically acceptable salts can be prepared by standard techniques. For example, the free-base form of a compound can be dissolved in a suitable solvent, such as an aqueous or aqueous-alcohol solution containing the appropriate acid and then isolated by evaporating the solution. In another example, a salt can be prepared by reacting the free base and acid in an organic solvent. If the particular compound is an acid, the desired pharmaceutically acceptable salt may be prepared by any suitable method, for example, treatment of the free acid with an appropriate inorganic or organic base.
  • The pharmaceutically acceptable salt of the different compounds may be present as a complex. Examples of complexes include 8-chlorotheophylline complex (analogous to, e.g., dimenhydrinate: diphenhydramine 8-chlorotheophylline (1:1) complex; Dramamine) and various cyclodextrin inclusion complexes.
  • (d) Other Compound Forms
  • In the case of agents that are solids, it is understood by those skilled in the art that the compounds and salts may exist in different crystal or polymorphic forms, or may be formulated as co-crystals, or may be in an amorphous form, or may be any combination thereof (e.g. partially crystalline, partially amorphous, or mixtures of polymorphs) all of which are intended to be within the scope of the present invention and specified formulae. Whereas salts are formed by acid/base addition, i.e. a free base or free acid of the compound of interest forms an acid/base reaction with a corresponding addition base or addition acid, respectively, resulting in an ionic charge interaction, co-crystals are a new chemical species that is formed between neutral compounds, resulting in the compound and an additional molecular species in the same crystal structure.
  • Additionally, the formulae are intended to cover hydrated or solvated as well as unhydrated or unsolvated forms of the identified structures. For example, the indicated structures include both hydrated and non-hydrated forms. Other examples of solvates include the structures in combination with a suitable solvent, such as isopropanol, ethanol, methanol, dimethylsulfoxide, ethyl acetate, acetic acid, or ethanolamine.
  • 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. Compounds of Formula I can be administered by different routes, including injection (i.e. parenteral, including intravenous, intraperitoneal, subcutaneous, and intramuscular), oral, transdermal, transmucosal, rectal, or inhalant. Such dosage forms should allow the compound to reach target cells. Other factors are well known in the art, and include considerations such as toxicity and dosage forms that retard the compound or composition from exerting its effects. Techniques and formulations generally may be found in Remington: The Science and Practice of Pharmacy, 21st edition, Lippincott, Williams and Wilkins, Philadelphia, Pa., 2005 (hereby incorporated by reference herein).
  • In some embodiments, compositions will comprise carriers or excipients, which may be chosen to facilitate administration of the compound by a particular route. Examples of carriers include calcium carbonate, calcium phosphate, various sugars such as lactose, glucose, or sucrose, types of starch, cellulose derivatives, gelatin, lipids, liposomes, nanoparticles, and the like. Carriers also include physiologically compatible liquids as solvents or for suspensions, including, for example, sterile solutions of water for injection (WFI), saline solution, dextrose solution, Hank's solution, Ringer's solution, vegetable oils, mineral oils, animal oils, polyethylene glycols, liquid paraffin, and the like.
  • In some embodiments, oral administration may be used. Pharmaceutical preparations for oral use can be formulated into conventional oral dosage forms such as capsules, tablets, and liquid preparations such as syrups, elixirs, and concentrated drops. Compounds of Formula I may be combined with solid excipients, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain, for example, tablets, coated tablets, hard capsules, soft capsules, solutions (e.g. aqueous, alcoholic, or oily solutions) and the like. Suitable excipients are, in particular, fillers such as sugars, including lactose, glucose, sucrose, mannitol, or sorbitol; cellulose preparations, for example, corn starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose (CMC), and/or polyvinylpyrrolidone (PVP: povidone); oily excipients, including vegetable and animal oils, such as sunflower oil, olive oil, or codliver oil. The oral dosage formulations may also contain disintegrating agents, such as the cross-linked polyvinylpyrrolidone, agar, or alginic acid, or a salt thereof such as sodium alginate; a lubricant, such as talc or magnesium stearate; a plasticizer, such as glycerol or sorbitol; a sweetening such as sucrose, fructose, lactose, or aspartame; a natural or artificial flavoring agent, such as peppermint, oil of wintergreen, or cherry flavoring; or dye-stuffs or pigments, which may be used for identification or characterization of different doses or combinations. Also provided are dragee cores with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain, for example, gum arabic, talc, poly-vinylpyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Pharmaceutical preparations that can be used orally include push-fit capsules made of gelatin (“gelcaps”), as well as soft, sealed capsules made of gelatin, and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • In some embodiments, injection (parenteral administration) may be used, e.g., intramuscular, intravenous, intraperitoneal, and/or subcutaneous. Compounds of Formula I for injection may be formulated in sterile liquid solutions, preferably in physiologically compatible buffers or solutions, such as saline solution, Hank's solution, or Ringer's solution. Dispersions may also be prepared in non-aqueous solutions, such as glycerol, propylene glycol, ethanol, liquid polyethylene glycols, triacetin, and vegetable oils. Solutions may also contain a preservative, such as methylparaben, propylparaben, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In addition, the compounds may be formulated in solid form, including, for example, lyophilized forms, and redissolved or suspended prior to use.
  • In some embodiments, transmucosal, topical or transdermal administration may be used. In such formulations of compounds of Formula I, penetrants appropriate to the barrier to be permeated are used. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, bile salts and fusidic acid derivatives. In addition, detergents may be used to facilitate permeation. Transmucosal administration, for example, may be through nasal sprays or suppositories (rectal or vaginal). Compositions of compounds of Formula I for topical administration may be formulated as oils, creams, lotions, ointments, and the like by choice of appropriate carriers known in the art. Suitable carriers include vegetable or mineral oils, white petrolatum (white soft paraffin), branched chain fats or oils, animal fats and high molecular weight alcohol (greater than C12). In some embodiments, carriers are selected such that the active ingredient is soluble. Emulsifiers, stabilizers, humectants and antioxidants may also be included as well as agents imparting color or fragrance, if desired. Creams for topical application are preferably formulated from a mixture of mineral oil, self-emulsifying beeswax and water in which mixture the active ingredient, dissolved in a small amount of solvent (e.g., an oil), is admixed. Additionally, administration by transdermal means may comprise a transdermal patch or dressing such as a bandage impregnated with an active ingredient and optionally one or more carriers or diluents known in the art. To be administered in the form of a transdermal delivery system, the dosage administration will be continuous rather than intermittent throughout the dosage regimen.
  • In some embodiments, compounds are administered as inhalants. Compounds of Formula I may be formulated as dry powder or a suitable solution, suspension, or aerosol. Powders and solutions may be formulated with suitable additives known in the art. For example, powders may include a suitable powder base such as lactose or starch, and solutions may comprise propylene glycol, sterile water, ethanol, sodium chloride and other additives, such as acid, alkali and buffer salts. Such solutions or suspensions may be administered by inhaling via spray, pump, atomizer, or nebulizer, and the like. The compounds of Formula I may also be used in combination with other inhaled therapies, for example corticosteroids such as fluticasone proprionate, beclomethasone dipropionate, triamcinolone acetonide, budesonide, and mometasone furoate; beta agonists such as albuterol, salmeterol, and formoterol; anticholinergic agents such as ipratroprium bromide or tiotropium; vasodilators such as treprostinal and iloprost; enzymes such as DNAase; therapeutic proteins; immunoglobulin antibodies; an oligonucleotide, such as single or double stranded DNA or RNA, siRNA; antibiotics such as tobramycin; muscarinic receptor antagonists; leukotriene antagonists; cytokine antagonists; protease inhibitors; cromolyn sodium; nedocril sodium; and sodium cromoglycate.
  • The amounts of various compounds to be administered can be determined by standard procedures taking into account factors such as the compound activity (in vitro, e.g. the compound IC50 vs. target, or in vivo activity in animal efficacy models), pharmacokinetic results in animal models (e.g. biological half-life or bioavailability), the age, size, and weight of the subject, and the disorder associated with the subject. The importance of these and other factors are well known to those of ordinary skill in the art. Generally, a dose will be in the range of about 0.01 to 50 mg/kg, also about 0.1 to 20 mg/kg of the subject being treated. Multiple doses may be used.
  • The compounds of Formula I may also be used in combination with other therapies for treating the same disease. Such combination use includes administration of the compounds and one or more other therapeutics at different times, or co-administration of the compound and one or more other therapies. In some embodiments, dosage may be modified for one or more of the compounds of the invention or other therapeutics used in combination, e.g., reduction in the amount dosed relative to a compound or therapy used alone, by methods well known to those of ordinary skill in the art.
  • It is understood that use in combination includes use with other therapies, drugs, medical procedures etc., where the other therapy or procedure may be administered at different times (e.g. within a short time, such as within hours (e.g. 1, 2, 3, 4-24 hours), or within a longer time (e.g. 1-2 days, 2-4 days, 4-7 days, 1-4 weeks)) than a compound of Formula I, or at the same time as a compound of Formula I. Use in combination also includes use with a therapy or medical procedure that is administered once or infrequently, such as surgery, along with a compound of Formula I administered within a short time or longer time before or after the other therapy or procedure. In some embodiments, the present invention provides for delivery of a compound of Formula I and one or more other drug therapeutics delivered by a different route of administration or by the same route of administration. The use in combination for any route of administration includes delivery of a compound of Formula I and one or more other drug therapeutics delivered by the same route of administration together in any formulation, including formulations where the two compounds are chemically linked in such a way that they maintain their therapeutic activity when administered. In one aspect, the other drug therapy may be co-administered with a compound of Formula I. Use in combination by co-administration includes administration of co-formulations or formulations of chemically joined compounds, or administration of two or more compounds in separate formulations within a short time of each other (e.g. within an hour, 2 hours, 3 hours, up to 24 hours), administered by the same or different routes. Co-administration of separate formulations includes co-administration by delivery via one device, for example the same inhalant device, the same syringe, etc., or administration from separate devices within a short time of each other. Co-formulations of a compound of Formula I and one or more additional drug therapies delivered by the same route includes preparation of the materials together such that they can be administered by one device, including the separate compounds combined in one formulation, or compounds that are modified such that they are chemically joined, yet still maintain their biological activity. Such chemically joined compounds may have a linkage that is substantially maintained in vivo, or the linkage may break down in vivo, separating the two active components.
  • 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.
  • Example 1: Synthesis of Compound of Formula Ib or Id wherein A is —C(O)—
  • Compounds of Formula Ib or Id, as defined in paragraphs [0010] and [0016], respectively, wherein A is —C(O)—, can be prepared in five steps as described in Scheme 1.
  • Figure US20170319559A1-20171109-C00029
  • Step 1—Synthesis of Compound 2
  • Compound 1 (R2 and R4 as defined in paragraph [0004]) is dissolved in an anhydrous solvent (e.g. tetrahydrofuran) under nitrogen atmosphere. The solution is cooled down with the aid of a dry ice and acetone bath. To this solution is added a base (e.g. n-butyllithium), followed by 1, 2-bis(chlorodimethylsilyl)ethane at low temperature (typically below −70° C.). The resulting mixture is stirred at low temperature for 1-2 hours. To this solution is added a base (e.g. n-butyllithium), followed by ethyl chloroformate. The resulting mixture is allowed to warm to room temperature and then stirred at room temperature for 1-3 days. The reaction mixture is quenched by an acid solution, stirred at room temperature for a couple of hours, and then basified. The mixture is extracted with an organic solvent (e.g. dichloromethane or ethyl acetate). The desired compound 2 is purified by chromatography.
  • Step 2—Synthesis of Compound 3
  • To compound 2 in an organic solvent (e.g. dichloromethane) is added pyridine, followed by an appropriate acylating agent, isocyanate, or sulfonyl chloride such as propane-1-sulfonylchloride. The reaction mixture is stirred at room temperature for over 12 hours and the mixture is then poured into water. The organic layer is separated and the aqueous layer is extracted with an appropriate organic solvent (e.g. dichloromethane). The desired compound 3 (L2 and R3 as defined in paragraph [0005], or L2 is S(O)2 for Formula Ib) is purified by chromatography.
  • Step 3—Synthesis of Compound 4
  • To compound 3 in a solvent mixture (e.g. tetrahydrofuran and water) is added a base (e.g. lithium hydroxide or sodium hydroxide). The resulting suspension is stirred in a heated oil bath for over 10 hours. The reaction mixture is cooled down to room temperature and then acidified with an acid solution such as concentrated hydrochloric acid. The aqueous layer is separated and extracted with an appropriate organic solvent (e.g. ethyl acetate). The desired compound 4 is purified by chromatography.
  • Step 4—Synthesis of Compound 5
  • To a suspension of compound 4 in an anhydrous solvent (e.g. dichloromethane), cooled with an ice and water bath, oxalyl chloride is added slowly, followed by dimethylformamide. The reaction mixture is stirred at room temperature for a few hours. After removal of the solvent and excess oxalyl chloride, the residue is used in the next step without further purification.
  • Step 5—Synthesis of Compound of Formula Ib or Id
  • To an appropriate amine 6 (Ar, m, R1 and R11 as defined in paragraph [0005]) in an anhydrous solvent (e.g. tetrahydrofuran) is added a base (e.g. triethylamine). To this mixture, cooled with an ice and salt bath, a solution of compound 5 in an anhydrous solvent (e.g. tetrahydrofuran) is added slowly. The resulting mixture was stirred at room temperature for over 12 hours. The desired compound of Formula Ib (L2 is S(O)2) or Id is purified by chromatography.
  • Example 2: Synthesis of Compound of Formula Ic or Ie
  • Compounds of Formula Ic or Ie, as defined in paragraphs [0015] and [0021], respectively, can be prepared in four steps as described in Scheme 2.
  • Figure US20170319559A1-20171109-C00030
  • Step 1—Synthesis of Compound 8
  • Compound 7 (R2 as defined in paragraph [0005]) is dissolved in an appropriate solvent (e.g. methanol). To this solution is added catalyst (e.g. palladium on carbon). The suspension is then placed under a hydrogen atmosphere and shaken at room temperature for over 12 hours. The catalyst is removed by filtration on a pad of celite and washed with an appropriate solvent (e.g. methanol). The filtrate is concentrated under reduced pressure to give compound 8, which is used in the next step without further purification.
  • Step 2—Synthesis of Compound 9
  • To compound 8 in an organic solvent (e.g. dichloromethane) is added a base (e.g. pyridine) followed by an appropriate acylating agent, isocyanate, or sulfonyl chloride. The reaction mixture is stirred at room temperature for over 12 hours. The reaction mixture is then poured into water. The organic layer is collected and the aqueous layer is extracted with an appropriate organic solvent (e.g. dichloromethane). The organic solvents are then combined. The desired compound 9 (L2 and R3 as defined in paragraph [0005], or L2 is S(O)2 for Formula Ic) is purified by chromatography.
  • Step 3—Synthesis of Compound 10
  • To compound 9 in an organic solvent (e.g. tetrahydrofuran or dichloromethane) is added a base (e.g. sodium hydride) at low temperature, followed by an appropriate alkylating agent (e.g. halide). The reaction mixture is stirred at room temperature or heated in an oil bath as necessary, for a few hours. The reaction mixture is then poured into water. The organic layer is collected and the aqueous layer is extracted with an appropriate organic solvent (e.g. ethyl acetate or dichloromethane). The organic solvents are then combined. The desired compound 10 (R4 as defined in paragraph [0005]) is purified by chromatography.
  • Step 4—Synthesis of Compound of Formula Ic or Ie
  • A mixture of compound 10, an appropriate boronic acid 11 (Ar, m and R1 as defined in paragraph [0004]), and a catalyst (e.g. tetrakis(triphenylphosphine)palladium) in a mixture of base (e.g. aqueous solution of potassium carbonate) and an appropriate organic solvent (e.g. acetonitrile) is heated in an oil bath or is irradiated in a microwave system at over 100° C. for an appropriate time depending on starting materials. The reaction mixture is poured into water and then extracted with an appropriate organic solvent (e.g. dichloromethane or ethyl acetate). The organic solvents are then combined. The desired compound of Formula Ic (L2 is S(O)2) or Id is purified by chromatography.
  • Example 3: Synthesis of Compound of Formula I where L1 is —CH2NR11
  • Compounds of Formula I, as defined in paragraph [0005] where L1 is —CH2NR11—, can be prepared in three steps as described in Scheme 3—Method A, or one step as described in Scheme 3—Method B.
  • Figure US20170319559A1-20171109-C00031
  • Step 1—Synthesis of Compound 12
  • Compound 4 (prepared as described in Scheme 1, Step 3 in Example 1) is dissolved in an appropriate solvent (e.g. tetrahydrofuran). To this solution is added an appropriate reducing agent (e.g. lithium tetrahydroaluminate) at low temperature (typically below −30° C.). The reaction mixture is then stirred at room temperature for 2-24 hours. Sodium sulfate is added and the mixture is stirred at room temperature for 30 minutes. The mixture is filtered through a pad of celite and washed with an appropriate solvent (e.g. ethyl acetate). The filtrate is concentrated under reduced pressure to give compound 12, which is used in the next step without further purification.
  • Step 2—Synthesis of Compound 13
  • Compound 13 (LG is a suitable leaving group) is prepared by converting compound 12 into a mesylate or triflate by reacting with the corresponding sulfonyl chloride in an appropriate organic solvent. It can also be converted into the corresponding bromide by reacting with an appropriate agent (e.g. phosphorous tribromide) in the presence of an appropriate base (e.g. pyridine).
  • Step 3—Synthesis of Compound of Formula I where L1 is —CH2NR11
  • To a mixture of compound 13 and a base (e.g. cesium carbonate) in an appropriate organic solvent (e.g. acetonitrile), amine 6 is added. The reaction mixture is stirred at room temperature, or heated in an oil bath if necessary, for 2-24 hours. The reaction mixture is poured into water and then extracted with an appropriate organic solvent (e.g. dichloromethane or ethyl acetate). The organic solvents are then combined. The desired compound of Formula I is purified by chromatography.
  • Figure US20170319559A1-20171109-C00032
  • Step 1—Preparation of Compound 63
  • To substituted phenylamine 1 (R2 and R4 as defined in paragraph [0005]) in an appropriate solvent (e.g. tetrahydrofuran) are added a base (e.g. triethylamine) and acid halide (e.g. acid chloride or sulfonyl chloride) in an appropriate organic solvent under an atmosphere of nitrogen. The reaction is stirred at room temperature for 2-24 hours. The reaction mixture is then poured into an acid solution and extracted with an appropriate organic solvent (e.g. dichloromethane or ethyl acetate). The organic layers are combined. The desired compound 63 (L2 and R3 as defined in paragraph [0004]) is purified by crystallization or chromatography.
  • Step 2—Preparation of Compound 64
  • Compound 63 in an appropriate solvent (e.g. tetrahydrofuran) under an atmosphere of nitrogen is cooled in an acetone/dry ice bath. To this solution is added a base (e.g. lithium diisopropylamide) and then an appropriate reagent (e.g. N,N-dimethyl-formamide). The reaction mixture is stirred for 0.5 to 3 hours at low temperature (<50° C.) and then allowed to warm to room temperature. The reaction mixture is poured into water and extracted with an appropriate organic solvent (e.g. dichloromethane or ethyl acetate). The desired compound 64 is purified by chromatography.
  • Step 3—Synthesis of Compound of Formula I where L1 is —CH2NR11
  • To a mixture of compound 64 in an appropriate organic solvent (e.g. acetonitrile) is added amine 6 (Ar, m, R1 and R11 as defined in paragraph [0005]), and reducing agent (e.g. triethylsilane and trifluoroacetic acid). The reaction mixture is heated in an oil bath for 2-24 hours. The reaction mixture is concentrated, poured into water and then extracted with an appropriate organic solvent (e.g. dichloromethane or ethyl acetate). The organic layers are then combined. The desired compound of Formula I is purified by chromatography.
  • Alternatively, compounds of Formula I where L1 is —CH2NR11— may be prepared by reduction of compounds of Formula Ib or Id wherein A is —C(O)— (e.g. prepared as described in Example 1) with an appropriate reducing agent (e.g. borane or diisobutylaluminum hydride).
  • Example 4: Synthesis of 6-chloro-2-fluoro-N-[6-(5-methyl-thiazol-2-ylamino)-pyridin-3-yl]-3-(propane-1-sulfonylamino)-benzamide P-0011
  • 6-Chloro-2-fluoro-N-[6-(5-methyl-thiazol-2-ylamino)-pyridin-3-yl]-3-(propane-1-sulfonylamino)-benzamide P-0011 was synthesized in six steps from 4-chloro-2-fluoroaniline 14 as shown in Scheme 4.
  • Figure US20170319559A1-20171109-C00033
    Figure US20170319559A1-20171109-C00034
  • Step 1—Preparation 3-amino-6-chloro-2-fluorobenzoic acid ethyl ester (15)
  • 4-Chloro-2-fluoroaniline (14, 12 mL) was dissolved in 200 mL of anhydrous tetrahydrofuran under a nitrogen atmosphere in a 1 L 3-neck round bottom flask. The mixture was cooled to −78° C. (dry ice/acetone bath) and n-butyllithium (2.5 M, 45 mL) was slowly added dropwise, maintaining the temperature below −70° C. The mixture was stirred at −70° C. for 30 minutes. 1,2-Bis(chlorodimethylsilyl)ethane (24.80 g) was dissolved in 80 mL of anhydrous tetrahydrofuran and slowly added dropwise to the reaction mixture while maintaining the temperature below −70° C. The resulting mixture was stirred at −78° C. for 1 hour, then n-butyllithium (2.5 M, 45 mL) was slowly added dropwise maintaining the temperature below −70° C. The mixture was then stirred for 30 minutes at −78° C., then warmed up to 15° C. over 1 hour. The reaction mixture was cooled down to −78° C. and n-butyllithium (2.5 M, 50 mL) was slowly added dropwise maintaining the temperature below −70° C. The mixture was stirred at −70° C. for 90 minutes, then 13.40 mL of ethylchloroformate was slowly added dropwise maintaining the temperature below −70° C. The reaction mixture was slowly warmed to room temperature and stirred at room temperature for 64 hours. The reaction was quenched by careful addition of a solution of 50 mL of concentrated hydrochloric acid in 160 mL of water while cooling with an ice/water bath. The mixture was stirred at room temperature for 2 hours, then made basic by addition of potassium carbonate. The mixture was extracted with 3×100 mL of ethyl acetate and the combined organic extracts were washed with 50 mL of brine and dried with magnesium sulfate. After removal of the solvent, the residue was purified with silica gel column chromatography eluting with ethyl acetate in hexane to provide the desired compound (15, 17 g, 72%).
  • Step 2—Preparation 6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoic acid ethyl ester (17)
  • 3-Amino-6-chloro-2-fluorobenzoic acid ethyl ester (15, 17 g) was dissolved in 785 mL of dichloromethane, to which 13.2 mL of pyridine was added, followed by propane-1-sulfonylchloride (16, 12.8 g). The reaction mixture was stirred at room temperature for 18 hours, then poured into 400 mL of water. The organic layer was separated and the aqueous layer was extracted with 200 mL of dichlormethane. The combined organic extracts were dried over magnesium sulfate to give an orange oil (41 g). Trituration in 150 mL of diethylether removed the pyridine salt as a white solid. The ether filtrate was concentrated to give orange oil, which was purified with silica gel column chromatography eluting with ethyl acetate in hexane to provide the desired compound as a pale yellow solid (17, 20 g, 57%).
  • Step 3—Preparation 6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoic acid (18)
  • 6-Chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoic acid ethyl ester (17, 20 g) was dissolved in a mixture of 500 mL tetrahydrofuran and 150 mL water. Lithium hydroxide (12.95 g) was added and the resulting suspension was stirred at 90° C. for 17 hours. The mixture was cooled down to room temperature, then brought to pH=1 with concentrated hydrochloric acid (˜36 mL). The aqueous layer was separated and extracted with 3×400 mL of ethyl acetate. The combined organic extracts were dried over magnesium sulfate and concentrated to give a beige solid (25 g). The solid was triturated in 100 mL of diethyl ether for 30 minutes, filtered, washed with 50 mL of diethyl ether and dried to provide the desired compound as a white solid (18, 16 g, 87%).
  • Step 4—Preparation 6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoyl chloride (19)
  • 6-Chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoic acid 18 was suspended in anhydrous dichloromethane (30 mL/g). Dimethylformamide (2 drops) was added and the suspension cooled in an ice/water bath. Oxalyl chloride (5 eq) was slowly added dropwise. The bath was then removed and the reaction mixture stirred at room temperature for 2 to 3 hours and the solids slowly disappeared. Dichloromethane and excess oxalyl chloride were removed under reduced pressure and the residue was used without further purification in the next step.
  • Step 5—Preparation of N-(6-bromo-pyridin-3-yl)-6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzamide (21)
  • 6-Bromo-pyridin-3-ylamine (20, 3.16 g, 18.26 mmol) was dissolved in 55 mL of anhydrous tetrahydrofuran. Triethylamine (1.85 g, 2.55 mL, 18.26 mmol) was added and the mixture cooled in an ice/salt bath. A solution of 6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoyl chloride (19, 3.8 g, 12.17 mmol) in 55 mL of anhydrous tetrahydrofuran was slowly added dropwise to the reaction mixture. The resulting mixture was stirred at room temperature for 18 hours, then diluted with 180 mL of ethyl acetate, washed 2×70 mL with water and once with 110 mL brine, dried over magnesium sulfate and concentrated to give a brown residue. The residue was purified by silica gel flash chromatography, eluting with 1% methanol, to provide the desired compound as a yellow solid (21, 6.9 g, 66%).
  • Step 6—Preparation 6-chloro-2-fluoro-N-[6-(5-methyl-thiazol-2-ylamino)-pyridin-3-yl]-3-(propane-1-sulfonylamino)-benzamide (P-0011)
  • N-(6-bromo-pyridin-3-yl)-6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzamide (21, 250 mg, 0.555 mmol) was placed in a microwave vial along with palladium acetate (12.5 mg, 0.055 mmol), BINAP (69 mg, 0.111 mmol), potassium tert-butoxide (124 mg, 1.11 mmol) and 2-amino-5-methyl-thiazole (22, 190 mg, 1.66 mmol). Dimethylformamide (2.5 mL) was added and the vial sealed. The mixture was then heated at 150° C. in a microwave for 3 hours. The black mixture was diluted with 50 mL of ethyl acetate and washed with 15 mL of water, then 15 mL of 0.67 M hydrochloric acid solution, 15 mL of water and finally 15 mL of brine. The organic phase was dried over magnesium sulfate. After removal of the solvent, the residue was purified by preparative TLC and then HPLC to provide the desired compound as a white solid (P-0011, 10 mg). MS (ESI) [M+H+]+=483.8.
  • Example 5: Synthesis of 6-chloro-2-fluoro-N-(6-isopropylamino-pyridin-3-yl)-3-(propane-1-sulfonylamino)-benzamide P-0005
  • 6-Chloro-2-fluoro-N-(6-isopropylamino-pyridin-3-yl)-3-(propane-1-sulfonylamino)-benzamide P-0005 was synthesized in three steps from 2-bromo-5-nitro-pyridine 23 as shown in Scheme 5.
  • Figure US20170319559A1-20171109-C00035
  • Step 1—Preparation of 6-isopropylamino-3-nitro pyridine (25)
  • 2-Bromo-5-nitro-pyridine (23, 400 mg) was placed in a microwave vial. Isopropylamine (24, 3 mL) was added and the vial sealed. The mixture was then heated at 120° C. for 30 minutes using a Biotage Initiator EXP microwave. The crude mixture was then absorbed on a column and purified by silica gel chromatography. The fractions containing the desired compound were combined and concentrated to provide the desired compound as a yellow solid.
  • Step 2—Preparation of N*2*-isopropyl-pyridine-2,5-diamine (26)
  • The 6-isopropylamino-3-nitro pyridine 25 was dissolved in methanol (35 mL/g). Palladium on carbon catalyst (10%, wet, ˜100 mg) was added and the suspension was placed under a hydrogen atmosphere at room temperature overnight (˜18 hours). The catalyst was removed by filtration on a pad of celite and washed with 2×10 mL of methanol. The filtrate was concentrated under reduced pressure to provide the desired compound, which was used without further purification in the next step.
  • Step 3—Preparation of 6-chloro-2-fluoro-N-(6-isopropylamino-pyridin-3-yl)-3-(propane-1-sulfonylamino)-benzamide (P-0005)
  • N*2*-isopropyl-pyridine-2,5-diamine (26, 155 mg, 1.01 mmol) was dissolved in 4 mL of anhydrous tetrahydrofuran. Triethylamine (103 mg, 142 μL, 1.01 mmol) was added and the mixture cooled in an ice/salt bath. 6-Chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoyl chloride (19, 200 mg, 0.68 mmol) in 4 mL of anhydrous tetrahydrofuran was then slowly added dropwise. The resulting mixture was stirred at room temperature for 20 hours, then diluted with 30 mL of ethyl acetate, washed with 3×10 mL of water and 15 mL of brine. After removal of the solvent, the residue was purified by silica gel chromatography to provide the desired compound as a white solid (P-0005, 35 mg). MS (ESI) [M+H+]+=429.0.
  • 6-Chloro-N-(6-cyclopentylamino-pyridin-3-yl)-2-fluoro-3-(propane-1-sulfonylamino)-benzamide P-0009, 6-Chloro-N-(6-cyclopropylamino-pyridin-3-yl)-2-fluoro-3-(propane-1-sulfonylamino)-benzamide P-0015, 6-Chloro-2-fluoro-3-(propane-1-sulfonylamino)-N-{6-[(thiophen-2-ylmethyl)-amino]-pyridin-3-yl}-benzamide P-0016, and N-(6-Benzylamino-pyridin-3-yl)-6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzamide P-0017,
  • Figure US20170319559A1-20171109-C00036
  • were prepared following the protocol of Scheme 5, replacing isopropylamine 24 with cyclopentylamine, cyclopropylamine, thiophen-2-yl-methylamine and benzylamine, respectively, in Step 1. MS (ESI) [M+H+]+ P-0009=455.2, P-0015=427.0, P-0016=483.2 and P-0017=477.2.
  • Example 6: Synthesis of N-(2-acetylamino-pyrimidin-5-yl)-6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzamide P-0004
  • N-(2-Acetylamino-pyrimidin-5-yl)-6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzamide P-0004 was synthesized in three steps from 2-amino-5-nitropyrimidine 27 as shown in Scheme 6.
  • Figure US20170319559A1-20171109-C00037
  • Step 1—Preparation of N-(5-nitro-pyrimidin-2-yl)-acetamide (28)
  • 2-Amino-5-nitropyrimidine (27, 500 mg) was suspended in 5 mL of acetic anhydride. The mixture was heated at 160° C. for two hours, then cooled to room temperature. The solids were filtered and washed with 5 mL of water, then suspended in 10 mL of water and the pH was brought to 8-9 by addition of 25% ammonium hydroxide solution. The solids were filtered, washed with 2×10 mL cold water and recrystallized from ethyl acetate to provide the desired compound as beige needles (28, 382 mg, 58%).
  • Step 2—Preparation of N-(5-amino-pyrimidin-2-yl)-acetamide (29)
  • N-(5—Nitro-pyrimidin-2-yl)-acetamide (28, 620 mg) was suspended in 31 mL of methanol. Palladium on carbon catalyst (10%, wet, 60 mg) was added and the suspension was placed under hydrogen atmosphere for 17 hours. The catalyst was filtered off on a pad of celite and washed with 2×30 mL of methanol. The filtrate was concentrated under reduced pressure to provide the desired compound as pale yellow needles (29, 520 mg, 100%).
  • Step 3—Preparation of N-(2-acetylamino-pyrimidin-5-yl)-6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzamide P-0004
  • N-(5-amino-pyrimidin-2-yl)-acetamide (29, 154 mg, 1.01 mmol) was dissolved in 3 mL of anhydrous tetrahydrofuran. Triethylamine (103 mg, 142 μL, 1.01 mmol) was added and the mixture cooled in an ice/salt bath. 6-Chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoyl chloride (19, 200 mg, 0.68 mmol) in 3 mL of anhydrous tetrahydrofuran was then slowly added dropwise. The resulting mixture was stirred at room temperature for 18 hours, then diluted with 20 mL of ethyl acetate, washed with 2×10 mL of water and 10 mL of brine. The organic layer was dried over magnesium sulfate. After removal of the solvent, the residue was purified by silica gel chromatography to provide the desired compound as a white solid (P-0004, 55 mg, 19%). MS (ESI) [M+H+]+=430.2.
  • Example 7: Synthesis of N-(6-acetylamino-pyridin-3-yl)-2,6-difluoro-3-(propane-1-sulfonylamino)-benzamide P-0008
  • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(propane-1-sulfonylamino)-benzamide P-0008 was synthesized in three steps from 2,6-difluoro-3-nitro-benzoic acid 30 and N-(5-amino-pyridin-2-yl)-acetamide 31 as shown in Scheme 7.
  • Figure US20170319559A1-20171109-C00038
  • Step 1—Preparation of N-(6-acetylamino-pyridin-3-yl)-2,6-difluoro-3-nitro-benzamide (32)
  • 2,6-difluoro-3-nitro-benzoic acid (30, 500 mg) was dissolved in 15 mL of anhydrous dichloromethane. N,N-Dimethylformamide (1 drop) was then added and the mixture cooled to 5° C. in an ice/water bath. Oxalyl chloride (1.1 mL, 5 eq) was slowly added dropwise. The reaction mixture was stirred at room temperature for two hours, then concentrated under reduced pressure to give a yellow solid residue, which was dissolved in 5 mL of anhydrous tetrahydrofuran and slowly added dropwise to a solution of N-(5-amino-pyridin-2-yl)-acetamide (31, 558 mg, 1.5 eq) and triethylamine (0.52 mL) in 10 mL of anhydrous tetrahydrofuran. The resulting suspension was stirred at room temperature overnight. The mixture was diluted with 50 mL of ethyl acetate, washed with 2×25 mL of water, then 25 mL brine, dried over magnesium sulfate and concentrated to provide the crude desired compound as a brown solid (32, 980 mg, 84%), which was used in the next step without further purification.
  • Step 2—Preparation of N-(6-acetylamino-pyridin-3-yl)-3-amino-2,6-difluoro-benzamide (33)
  • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-nitro-benzamide (32, 950 mg) was suspended in 15 mL of methanol. Palladium on carbon catalyst (10%, wet, 100 mg) was added and the suspension was placed under hydrogen atmosphere for 17 hours. The catalyst was filtered off on a pad of celite and washed with 2×20 mL of methanol. The filtrate was concentrated under reduced pressure to provide the desired compound as a black solid (33, 750 mg, 86%).
  • Step 3—Preparation of N-(6-acetylamino-pyridin-3-yl)-2,6-difluoro-3-(propane-1-sulfonylamino)-benzamide (P-0008)
  • N-(6-Acetylamino-pyridin-3-yl)-3-amino-2,6-difluoro-benzamide (33, 700 mg) was dissolved in 35 mL of pyridine. 4-dimethyl-amino-pyridine (1 eq) was added followed by propane-1-sulfonyl chloride (16, 0.80 g, 2.3 eq). The resulting mixture was stirred at room temperature for 3 days, then at 70° C. for 18 hours. The pyridine was removed under reduced pressure and the residue purified by silica gel column chromatography eluting with ethyl acetate in hexanes to provide the desired compound as a yellow solid (P-0008, 10 mg, 1%). MS (ESI) [M+H+]+=412.9.
  • Additional compounds may be prepared following the protocol of Scheme 7, replacing propane-1-sulfonyl chloride 16 with a suitable sulfonyl chloride in Step 3. The following compounds may be prepared by this method:
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(2-fluoro-benzenesulfonylamino)-benzamide (P-0097),
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(3-fluoro-benzenesulfonylamino)-benzamide (P-0098),
    • N-(6-Acetylamino-pyridin-3-yl)-3-(2,6-difluoro-benzenesulfonylamino)-2,6-difluoro-benzamide (P-0099),
    • N-(6-Acetylamino-pyridin-3-yl)-3-(2,4-difluoro-benzenesulfonylamino)-2,6-difluoro-benzamide (P-0100),
    • N-(6-Acetylamino-pyridin-3-yl)-3-(2,5-difluoro-benzenesulfonylamino)-2,6-difluoro-benzamide (P-0101),
    • 6-Acetylamino-N-[2,6-difluoro-3-(3-fluoro-4-methoxy-benzenesulfonylamino)-phenyl]-nicotinamide (P-0102),
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzamide (P-0103),
    • N-(6-Acetylamino-pyridin-3-yl)-3-(4-difluoromethoxy-benzenesulfonylamino)-2,6-difluoro-benzamide (P-0104),
    • N-(6-Acetylamino-pyridin-3-yl)-3-(3-difluoromethoxy-benzenesulfonylamino)-2,6-difluoro-benzamide (P-0105),
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(4-isopropyl-benzenesulfonylamino)-benzamide (P-0106),
    • N-(6-Acetylamino-pyridin-3-yl)-3-(4-tert-butyl-benzenesulfonylamino)-2,6-difluoro-benzamide (P-0107),
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(4-propyl-benzenesulfonylamino)-benzamide (P-0108),
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(pyridine-2-sulfonylamino)-benzamide (P-0109),
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(pyridine-3-sulfonylamino)-benzamide (P-0110),
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(dimethylaminosulfonylamino)-benzamide (P-0111),
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(piperidine-1-sulfonylamino)-benzamide (P-0112),
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(morpholine-4-sulfonylamino)-benzamide (P-0113),
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(tetrahydro-pyran-4-sulfonylamino)-benzamide (P-0114),
    • N-(6-Acetylamino-pyridin-3-yl)-3-cyclopentanesulfonylamino-2,6-difluoro-benzamide (P-0115),
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(pyrrolidine-1-sulfonylamino)-benzamide (P-0116), and
    • N-(6-Acetylamino-pyridin-3-yl)-2,6-difluoro-3-(3,3,3-trifluoro-propane-1-sulfonylamino)-benzamide (P-0117).
      The following table indicates the compound number in Column 1, the sulfonyl chloride used in Step 3 in Column 2, and the resulting compound in Column 3:
  • Compound
    number Sulfonyl Chloride in Step 3 Compound
    P-0097
    Figure US20170319559A1-20171109-C00039
    Figure US20170319559A1-20171109-C00040
    P-0098
    Figure US20170319559A1-20171109-C00041
    Figure US20170319559A1-20171109-C00042
    P-0099
    Figure US20170319559A1-20171109-C00043
    Figure US20170319559A1-20171109-C00044
    P-0100
    Figure US20170319559A1-20171109-C00045
    Figure US20170319559A1-20171109-C00046
    P-0101
    Figure US20170319559A1-20171109-C00047
    Figure US20170319559A1-20171109-C00048
    P-0102
    Figure US20170319559A1-20171109-C00049
    Figure US20170319559A1-20171109-C00050
    P-0103
    Figure US20170319559A1-20171109-C00051
    Figure US20170319559A1-20171109-C00052
    P-0104
    Figure US20170319559A1-20171109-C00053
    Figure US20170319559A1-20171109-C00054
    P-0105
    Figure US20170319559A1-20171109-C00055
    Figure US20170319559A1-20171109-C00056
    P-0106
    Figure US20170319559A1-20171109-C00057
    Figure US20170319559A1-20171109-C00058
    P-0107
    Figure US20170319559A1-20171109-C00059
    Figure US20170319559A1-20171109-C00060
    P-0108
    Figure US20170319559A1-20171109-C00061
    Figure US20170319559A1-20171109-C00062
    P-0109
    Figure US20170319559A1-20171109-C00063
    Figure US20170319559A1-20171109-C00064
    P-0110
    Figure US20170319559A1-20171109-C00065
    Figure US20170319559A1-20171109-C00066
    P-0111
    Figure US20170319559A1-20171109-C00067
    Figure US20170319559A1-20171109-C00068
    P-0112
    Figure US20170319559A1-20171109-C00069
    Figure US20170319559A1-20171109-C00070
    P-0113
    Figure US20170319559A1-20171109-C00071
    Figure US20170319559A1-20171109-C00072
    P-0114
    Figure US20170319559A1-20171109-C00073
    Figure US20170319559A1-20171109-C00074
    P-0115
    Figure US20170319559A1-20171109-C00075
    Figure US20170319559A1-20171109-C00076
    P-0116
    Figure US20170319559A1-20171109-C00077
    Figure US20170319559A1-20171109-C00078
    P-0117
    Figure US20170319559A1-20171109-C00079
    Figure US20170319559A1-20171109-C00080
  • Example 8: Synthesis of pyrrolidine-1-carboxylic acid {5-[6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoylamino]-pyridin-2-yl}-amide P-0006
  • Pyrrolidine-1-carboxylic acid {5-[6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoylamino]-pyridin-2-yl}-amide P-0006 was synthesized in four steps from 2-amino-5-nitro-pyridine 34 as shown in Scheme 8.
  • Figure US20170319559A1-20171109-C00081
  • Step 1—Preparation (5-nitro-pyridin-2-yl)-carbamic acid isopropenyl ester (35)
  • 2-Amino-5-nitro-pyridine (34, 500 mg) was dissolved in 5 mL of anhydrous tetrahydrofuran. N-Methyl morpholine (436 mg, 1.2 eq) was added and the mixture cooled to −10° C. in an ice/salt acetone bath. A solution of isopropenyl chloroformate (520 mg) in 5 mL of tetrahydrofuran was then slowly added dropwise while maintaining the temperature below −10° C. The reaction mixture was stirred at room temperature overnight, then diluted with 25 mL of ethyl acetate and 20 mL of water. The aqueous layer was separated and extracted with 2×25 mL of ethyl acetate. The combined organic extracts were washed with 25 mL of half saturated brine and dried over magnesium sulfate. After removal of the solvent, the residue was purified with silica gel column chromatography eluting with ethyl acetate in hexane to provide the desired compound as a white solid (35, 0.52 g, 65%).
  • Step 2—Preparation of pyrrolidine-1-carboxylic acid (5-nitro-pyridin-2-yl)-amide (37)
  • (5—Nitro-pyridin-2-yl)-carbamic acid isopropenyl ester (35, 160 mg) was dissolved in 2 mL of anhydrous tetrahydrofuran. N-Methylpyrrolidine (6 mg, 0.1 eq) was added, followed by pyrrolidine (36, 51 mg, 1 eq). The mixture was stirred at room temperature overnight, forming a precipitate. The precipitate was filtered off, washed with 1 mL of tetrahydrofuran and dried to give a white solid. Additional compound was obtained by concentration of the filtrate under reduced pressure and trituration of the residue in diethyl ether (˜5 mL) to provide the desired compound as a beige solid (37, 0.12 g, 71%).
  • Step 3—Preparation of pyrrolidine-1-carboxylic acid (5-amino-pyridin-2-yl)-amide (38)
  • Pyrrolidine-1-carboxylic acid (5-nitro-pyridin-2-yl)-amide (37, 115 mg) was dissolved in 10 mL of methanol. Palladium on carbon catalyst was added and the suspension placed under a hydrogen atmosphere for 64 hours. The catalyst was filtered over a pad of celite and washed with 2×10 mL of methanol. The filtrate was concentrated under reduced pressure to provide the desired compound as a grey solid (38, 0.1 g, 100%).
  • Step 4—Preparation of pyrrolidine-1-carboxylic acid {5-[6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoylamino]-pyridin-2-yl}-amide (P-0006)
  • Pyrrolidine-1-carboxylic acid (5-amino-pyridin-2-yl)-amide (38, 100 mg, 0.48 mmol) was dissolved in 2 mL of anhydrous tetrahydrofuran. Triethylamine (49 mg, 67 μL, 0.48 mmol) was added and the mixture cooled in an ice/salt bath. 6-Chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoyl chloride (19, 100 mg, 0.34 mmol) in 1 mL of anhydrous tetrahydrofuran was then slowly added dropwise. The resulting mixture was stirred at room temperature for 60 hours, then diluted with 20 mL of ethyl acetate, washed with 2×10 mL of water and 10 mL of brine. The organic layer was dried over magnesium sulfate. After removal of solvent, the residue was purified by silica gel column chromatography eluting with ethyl acetate in hexanes to provide the desired compound as a white solid (P-0006, 5 mg, 3%). MS (ESI) [M+H+]+=484.0.
  • Example 9: Synthesis of 6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-N-quinolin-3-yl-benzamide P-0013
  • 6-Chloro-2-fluoro-3-(propane-1-sulfonylamino)-N-quinolin-3-yl-benzamide P-0013 was synthesized in one step from 3-aminoquinoline 39 as shown in Scheme 9.
  • Figure US20170319559A1-20171109-C00082
  • Step 1—Preparation of 6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-N-quinolin-3-yl-benzamide (P-0013)
  • 3-Aminoquinoline (39, 172 mg, 1.19 mmol) was dissolved in 5 mL of anhydrous tetrahydrofuran. Triethylamine (120 mg, 170 μL, 1.19 mmol) was added and the mixture cooled in an ice/salt bath. 6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoyl chloride (19, 250 mg, 0.80 mmol) in 5 mL of anhydrous tetrahydrofuran was slowly added dropwise. The resulting mixture was stirred at room temperature for 20 hours, then diluted with 30 mL of ethyl acetate, washed with 3×10 mL of water and 15 mL of brine. The organic layer was dried over magnesium sulfate and concentrated to give a pale yellow residue (400 mg), which was further triturated in ethyl acetate to provide the desired compound as a white solid (P-0013, 110 mg, 32%). MS (ESI) [M+H+]+=421.9.
  • N-(6-Acetylamino-pyridin-3-yl)-6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzamide P-0002, 6-Chloro-2-fluoro-N-(6-methoxy-pyridin-3-yl)-3-(propane-1-sulfonylamino)-benzamide P-0003, 6-Chloro-N-(3,5-dimethyl-isoxazol-4-yl)-2-fluoro-3-(propane-1-sulfonylamino)-benzamide P-0007, 6-chloro-N-[5-(4-chloro-phenyl)-2H-pyrazol-3-yl]-2-fluoro-3-(propane-1-sulfonylamino)-benzamide P-0010, 6-chloro-N-[5-(4-chloro-benzyl)-[1,3,4]thiadiazol-2-yl]-2-fluoro-3-(propane-1-sulfonylamino)-benzamide P-0012, [2-[6-Chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoylamino]-4H-[1,3,4]thiadiazin-(5E)-ylidene]-acetic acid ethyl ester P-0014, and 6-Chloro-2-fluoro-N-imidazo[1,2-a]pyridin-3-yl-3-(propane-1-sulfonylamino)-benzamide P-0018,
  • Figure US20170319559A1-20171109-C00083
    Figure US20170319559A1-20171109-C00084
  • were prepared following the protocol of Scheme 9, replacing 3-aminoquinoline 39 with N-(5-amino-pyridin-2-yl)-acetamide; 6-methoxy-pyridin-3-ylamine; 3,5-dimethyl-isoxazol-4-ylamine; 5-(4-chloro-phenyl)-2H-pyrazol-3-ylamine; 5-(4-chloro-benzyl)-[1,3,4]thiadiazol-2-ylamine; [2-amino-4H-[1,3,4]thiadiazin-(5E)-ylidene]-acetic acid ethyl ester; and imidazo[1,2-a]pyridin-3-ylamine, respectively. MS (ESI) [M+H+]+ P-0002=429.2, P-0003=402.2, P-0007=389.9, P-0010=471.2, P-0012=503.0, P-0014=478.9 and P-0018=411.0.
  • Example 10: Synthesis of 6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-N-pyridin-3-yl-benzamide P-0001
  • 6-Chloro-2-fluoro-3-(propane-1-sulfonylamino)-N-pyridin-3-yl-benzamide P-0001 was synthesized in two steps from 6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoic acid 18 as shown in Scheme 10.
  • Figure US20170319559A1-20171109-C00085
  • Step 1—Preparation of 6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoyl chloride (19)
  • To 6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoic acid (18, 502 mg, 1.70 mmol, prepared as in Step 3 of Scheme 4, Example 4) in 35 mL of dichloromethane, oxalyl chloride (5 mL, 2.0 M in dichloromethane) and N,N-dimethylformamide (100 μL, 0.001 mol) were added. The reaction mixture was stirred at room temperature for 2 hours. The reaction was concentrated to give compound 19, used without further purification.
  • Step 2—Preparation of 6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-N-pyridin-3-yl-benzamide (P-0001)
  • To 6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoyl chloride (19, 0.200 g, 0.64 mmol) in 10.0 mL of dichloromethane, pyridin-3-ylamine (40, 0.126 g, 1.34 mmol) and 4-dimethylaminopyridine (7.8 mg, 0.064 mmol) were added. The reaction was stirred at room temperature overnight. The reaction mixture 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 with silica gel column chromatography eluting 5% methanol in dichloromethane to provide the desired compound (P-0001, 0.15 g, 63%). MS (ESI) [M+H+]+=371.1.
  • Example 11: Synthesis of propane-1-sulfonic acid {2,4-difluoro-3-[(5-methyl-isoxazol-3-ylamino)-methyl]-phenyl}-amide P-0019
  • Propane-1-sulfonic acid {2,4-difluoro-3-[(5-methyl-isoxazol-3-ylamino)-methyl]-phenyl}-amide P-0019 was synthesized in three steps from 2,4-difluoro-phenylamine 41 as shown in Scheme 11.
  • Figure US20170319559A1-20171109-C00086
  • Step 1—Preparation of propane-1-sulfonic acid (2,4-difluoro-phenyl)-amide (42)
  • To 2,4-difluoro-phenylamine (41, 3.0 mL, 29.8 mmol) in 50 mL of tetrahydrofuran, triethylamine (9.13 mL, 65.5 mmol) and propane-1-sulfonyl chloride (16, 2.90 mL, 25.8 mmol) were added under an atmosphere of nitrogen. The reaction was stirred at room temperature overnight. The reaction was poured into 1 M HCl and extracted with ethyl acetate. The organic layer was washed with brine, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated to provide the desired compound (42, 2.0 g, 28%), which was used without further purification in the next step.
  • Step 2—Preparation of propane-1-sulfonic acid (2,4-difluoro-3-formyl-phenyl)-amide (43)
  • To propane-1-sulfonic acid (2,4-difluoro-phenyl)-amide (42, 1.5 g, 6.38 mmol) in 10 mL of tetrahydrofuran under an atmosphere of nitrogen, cooled in a −78° C. acetone/dry ice bath, lithium diisopropylamide (0.80 M in tetrahydrofuran, 24 mL, freshly prepared from n-butyllithium and diisopropylamine) was added. After 30 minutes, N,N-dimethyl-formamide (542 μL, 7.018 mmol) was added dropwise to the reaction. The reaction was stirred for 30 minutes at −78° C. and then allowed to warm to room temperature for 40 minutes. 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 5% ethyl acetate in hexane to give a light yellow solid (43, 300 mg, 18%). MS(ESI)[M−H+]=262.3.
  • Step 3—Preparation of propane-1-sulfonic acid {2,4-difluoro-3-[(5-methyl-isoxazol-3-ylamino)-methyl]-phenyl}-amide (P-0019)
  • To 5-methyl-isoxazol-3-ylamine (44, 0.13 g, 1.3 mmol) in 20 mL of acetonitrile, propane-1-sulfonic acid (2,4-difluoro-3-formyl-phenyl)-amide (43, 0.35 g, 1.3 mmol), triethylsilane (1 mL, 7 mmol) and trifluoroacetic acid (0.5 mL, 7 mmol) were added. The reaction mixture was stirred at 80° C. overnight. The reaction mixture was concentrated, then poured into aqueous potassium carbonate and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate. After removal of drying agent and then solvent, the residue was purified by silica gel column chromatography to provide the desired compound as a white solid (P-0019, 0.22 g, 48%). MS (ESI) [M−H+]=346.95.
  • N-{5-[2,6-Difluoro-3-(propane-1-sulfonylamino)-benzylamino]-pyridin-2-yl}-acetamide P-0020, propane-1-sulfonic acid [2,4-difluoro-3-(quinolin-3-ylaminomethyl)-phenyl]-amide P-0021, propane-1-sulfonic acid {3-[(6-chloro-pyridin-3-ylamino)-methyl]-2,4-difluoro-phenyl}-amide P-0022, and propane-1-sulfonic acid {2,4-difluoro-3-[(6-methoxy-pyridin-3-ylamino)-methyl]-phenyl}-amide P-0023,
  • Figure US20170319559A1-20171109-C00087
  • were prepared following the protocol of Scheme 11, replacing 5-methyl-isoxazol-3-ylamine 44 with N-(5-amino-pyridin-2-yl)-acetamide, quinolin-3-ylamine, 6-chloro-pyridin-3-ylamine, and 6-methoxy-pyridin-3-ylamine, respectively. MS (ESI) [M+H+]+ P-0020=399.35, P-0021=392.40, P-0022=376.95, and P-0023=372.55.
  • Example 12: Synthesis of quinoline-3-carboxylic acid [2,6-difluoro-3-(propane-1-sulfonylamino)-phenyl]-amide P-0024 and propane-1-sulfonic acid {2,4-difluoro-3-[(quinolin-3-ylmethyl)-amino]-phenyl}-amide P-0025
  • Quinoline-3-carboxylic acid [2,6-difluoro-3-(propane-1-sulfonylamino)-phenyl]-amide P-0024 and propane-1-sulfonic acid {2,4-difluoro-3-[(quinolin-3-ylmethyl)-amino]-phenyl}-amide P-0025 were synthesized in three steps from propane-1-sulfonic acid (2,4-difluoro-3-formyl-phenyl)-amide 43 as shown in Scheme 12.
  • Figure US20170319559A1-20171109-C00088
    Figure US20170319559A1-20171109-C00089
  • Step 1—Preparation of 2,6-difluoro-3-(propane-1-sulfonylamino)-benzoic acid (45)
  • To a reaction flask, propane-1-sulfonic acid (2,4-difluoro-3-formyl-phenyl)-amide (43, 3.00 g, 11.4 mmol) and oxone (9.10 g, 14.8 mmol) and 30 mL of anhydrous N,N-dimethylformamide were added under nitrogen. The mixture was stirred at room temperature overnight, then quenched with 250 mL of 1 M hydrochloric acid solution and extracted with 250 mL of ethyl acetate. The organic layers were washed with 3×100 mL of 1M hydrochloric acid solution and dried over magnesium sulfate. After removal of drying agent and solvent, the residue was dried in vacuo to provide the desired compound (45, 2.9 g, 91%).
  • Step 2—Preparation of propane-1-sulfonic acid (3-amino-2,4-difluoro-phenyl)-amide (46)
  • To a reaction flask, 2,6-difluoro-3-(propane-1-sulfonylamino)-benzoic acid (45, 2.88 g, 10.3 mmol), triethylamine (2.09 g, 20.6 mmol) and diphenylphosphoryl azide (3.21 g, 11.7 mmol) and 84 mL of anhydrous tert-butanol were added under nitrogen. The reaction mixture was heated in an oil bath at 105° C. overnight, then cooled to room temperature and diluted with ethyl acetate. The organic layer was washed with 3×250 mL of water, 250 mL of brine, dried over magnesium sulfate, filtered and concentrated in vacuo to provide 4.1 g of crude Boc-protected amine, which was purified by silica gel column chromatography using hexane: ethyl acetate as eluant to provide 3.3 g of the Boc-protected amine. This was dissolved in 50 mL of dichloromethane and 16 mL of trifluoroacetic acid was added and the reaction stirred at room temperature until there was no starting material by TLC. The reaction was neutralized by pouring into a cooled saturated solution of sodium bicarbonate and extracted into 3×150 mL of dichloromethane. The combined organic extracts were washed with 50 mL of brine, dried over magnesium sulfate, filtered and concentrated in vacuo to provide the desired compound (46, 1.94 g, 75%).
  • Step 3a—Preparation of quinoline-3-carboxylic acid [2,6-difluoro-3-(propane-1-sulfonylamino)-phenyl]-amide (P-0024)
  • To a reaction vessel, 3-quinoline carboxylic acid (47, 39.1 mg, 0.23 mmol), 1.5 mL of anhydrous tetrahydrofuran, 1 drop of anhydrous N,N-dimethylformamide and oxalyl chloride (86 mg, 0.68 mmol) were added under nitrogen. The reaction was stirred at room temperature for 1.5 hours, then concentrated to dryness and the residue was diluted with 2 mL of anhydrous tetrahydrofuran. To this solution, triethylamine (15.8 mg, 0.16 mmol) and propane-1-sulfonic acid (3-amino-2,4-difluoro-phenyl)-amide (46, 100 mg, 0.40 mmol) were added and the reaction was stirred at room temperature over weekend. The reaction mixture was diluted with 5 mL of water and extracted into 3×10 mL of ethyl acetate. The organic extracts were dried over magnesium sulfate, filtered and concentrated in vacuo, then purified by silica gel column chromatography (hexane:ethyl acetate gradient) to provide the desired compound (P-0024, 33 mg, 36%). MS (ESI) [M+H+]+=406.1.
  • Step 3b—Preparation of propane-1-sulfonic acid {2,4-difluoro-3-[(quinolin-3-ylmethyl)-amino]-phenyl}-amide (P-0025)
  • To a reaction vessel, propane-1-sulfonic acid (3-amino-2,4-difluoro-phenyl)-amide (46, 155 mg, 0.62 mmol), 2 mL of anhydrous acetonitrile, 3-quinoline carboxaldehyde (48, 100 mg, 0.64 mmol), trifluoroacetic acid (431 mg, 3.78 mmol) and triethylsilane (425 mg, 3.65 mmol) were added under nitrogen. The reaction was heated at 80° C. overnight, then cooled to room temperature and concentrated in vacuo, to which 10 mL of an aqueous solution of 10% potassium carbonate was added. This was extracted into 3×15 mL of ethyl acetate. The combined organic extracts were washed with 15 mL of brine, dried over magnesium sulfate, filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography (hexane:ethyl acetate gradient) to provide the desired compound (P-0025, 70 mg, 29%). MS (ESI) [M+H+]+=392.0.
  • Example 13: Synthesis of propane-1-sulfonic acid [2,4-difluoro-3-(quinolin-3-yloxymethyl)-phenyl]-amide P-0026
  • Propane-1-sulfonic acid [2,4-difluoro-3-(quinolin-3-yloxymethyl)-phenyl]-amide P-0026 was synthesized in three steps from propane-1-sulfonic acid (2,4-difluoro-3-formyl-phenyl)-amide 43 as shown in Scheme 13.
  • Figure US20170319559A1-20171109-C00090
  • Step 1—Preparation of propane-1-sulfonic acid (2,4-difluoro-3-hydroxymethyl-phenyl)-amide (49)
  • To a reaction vessel, propane-1-sulfonic acid (2,4-difluoro-3-formyl-phenyl)-amide (43, 1.00 g, 3.80 mmol), 20 mL of methanol, and sodium borohydride (0.29 g, 7.60 mmol) were added under nitrogen. The reaction was stirred at room temperature for 1 hour, then poured onto 50 mL of aqueous 10% sodium dihydrogenphosphate. The mixture was extracted with 3×50 mL of dichloromethane and the combined organic extracts were dried over magnesium sulfate, filtered and concentrated in vacuo to provide the desired compound (49, 0.97 g, 96%), used in the next step without further purification.
  • Step 2—Preparation of propane-1-sulfonic acid (3-bromomethyl-2,4-difluoro-phenyl)-amide (50)
  • To a reaction vessel under nitrogen containing polymer-supported triphenylphosphine (2.45 g, 4.41 mmol) in 5 mL of acetonitrile, bromine (0.70 g, 4.41 mmol) was added, followed by a solution of propane-1-sulfonic acid (2,4-difluoro-3-hydroxymethyl-phenyl)-amide (49, 0.97 g, 3.67 mmol) in 5 mL of acetonitrile. The reaction mixture was stirred at 60° C. for approximately 3 hours. The reaction mixture was filtered and the polymer washed with 5 mL of ethyl acetate. The filtrate and wash were concentrated in vacuo to provide the desired compound (50, 0.91 g, 76%), used in the next step without further purification.
  • Step 3—Preparation of propane-1-sulfonic acid [2,4-difluoro-3-(quinolin-3-yloxymethyl)-phenyl]-amide (P-0026)
  • To reaction vessel, 3-hydroxyquinoline (51, 442 mg, 3.05 mmol) and 5 mL of anhydrous N,N-dimethylformamide were added under nitrogen. Sodium hydride (60% dispersion in mineral oil, 183 mg, 4.57 mmol) was added in portions. The reaction was stirred at room temperature for 30 minutes, then propane-1-sulfonic acid (3-bromomethyl-2,4-difluoro-phenyl)-amide (50, 500 mg, 1.52 mmol) was added and the reaction stirred at room temperature overnight. The reaction mixture was neutralized with acetic acid and extracted into 3×20 mL of ethyl acetate. The combined organic extracts were dried over magnesium sulfate, filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography (hexane: ethyl acetate gradient), followed by second purification by silica gel column chromatography to provide the desired compound (P-0026, 50 mg, 8%). MS (ESI) [M+H+]+=393.0.
  • Additional compounds may be prepared following the protocol of Scheme 13, optionally replacing propane-1-sulfonic acid (2,4-difluoro-3-formyl-phenyl)-amide 43 with N-(2,4-difluoro-3-formyl-phenyl)-4-trifluoromethyl-benzenesulfonamide (prepared following the protocol of Scheme 11, Example 11, steps 1 and 2 using 4-trifluoromethyl-benzenesulfonyl chloride in Step 1 in place of propane-1-sulfonyl chloride 16) in Step 1 and 3-hydroxyquinoline 51 with a suitable alcohol in Step 3. The following compounds may be prepared by this method:
    • N-{5-[2,6-Difluoro-3-(propane-1-sulfonylamino)-benzyloxy]-pyridin-2-yl}-acetamide (P-0091),
    • Propane-1-sulfonic acid [3-(2-amino-pyridin-3-yloxymethyl)-2,4-difluoro-phenyl]-amide (P-0092),
    • Propane-1-sulfonic acid [2,4-difluoro-3-(1H-pyrrolo[2,3-b]pyridin-5-yloxymethyl)-phenyl]-amide (P-0093),
    • N-{5-[2,6-Difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzyloxy]-pyridin-2-yl}-acetamide (P-0094),
    • N-[3-(2-Amino-pyridin-3-yloxymethyl)-2,4-difluoro-phenyl]-4-trifluoromethyl-benzenesulfonamide (P-0095), and
    • N-[2,4-Difluoro-3-(1H-pyrrolo[2,3-b]pyridin-5-yloxymethyl)-phenyl]-4-trifluoromethyl-benzenesulfonamide (P-0096).
      The following table indicates the compound number in Column 1, the aldehyde used in Step 1 in Column 2, the alcohol used in Step 3 in Column 3, and the resulting compound in Column 4:
  • Com-
    pound
    num-
    ber Aldehyde in Step 1 Alcohol in Step 3 Compound
    P-0091
    Figure US20170319559A1-20171109-C00091
    Figure US20170319559A1-20171109-C00092
    Figure US20170319559A1-20171109-C00093
    P-0092
    Figure US20170319559A1-20171109-C00094
    Figure US20170319559A1-20171109-C00095
    Figure US20170319559A1-20171109-C00096
    P-0093
    Figure US20170319559A1-20171109-C00097
    Figure US20170319559A1-20171109-C00098
    Figure US20170319559A1-20171109-C00099
    P-0094
    Figure US20170319559A1-20171109-C00100
    Figure US20170319559A1-20171109-C00101
    Figure US20170319559A1-20171109-C00102
    P-0095
    Figure US20170319559A1-20171109-C00103
    Figure US20170319559A1-20171109-C00104
    Figure US20170319559A1-20171109-C00105
    P-0096
    Figure US20170319559A1-20171109-C00106
    Figure US20170319559A1-20171109-C00107
    Figure US20170319559A1-20171109-C00108
  • Example 14: Synthesis of 2,6-difluoro-3-(propane-1-sulfonylamino)-N-quinolin-3-yl-benzamide P-0027
  • 2,6-Difluoro-3-(propane-1-sulfonylamino)-N-quinolin-3-yl-benzamide P-0027 was synthesized in one step from 2,6-difluoro-3-(propane-1-sulfonylamino)-benzoic acid 45 as shown in Scheme 14.
  • Figure US20170319559A1-20171109-C00109
  • Step 1—Preparation of 2,6-difluoro-3-(propane-1-sulfonylamino)-N-quinolin-3-yl-benzamide (P-0027)
  • To reaction vessel, 2,6-difluoro-3-(propane-1-sulfonylamino)-benzoic acid (45, 250 mg, 0.90 mmol), 5 mL of anhydrous dichloromethane and anhydrous N,N-dimethylformamide (6.54 mg, 0.09 mmol) were added under nitrogen. The reaction mixture was cooled to 0° C. and oxalyl chloride (568 mg, 4.48 mmol) was added dropwise. This was stirred at room temperature for 3 hours, then concentrated in vacuo. The residue was diluted with 5 mL of anhydrous tetrahydrofuran and triethylamine (136 mg, 1.34 mmol) and 3-aminoquinoline (39, 194 mg, 1.34 mmol) were added. The reaction mixture was stirred at room temperature over the weekend, then concentrated in vacuo. The residue was purified by silica gel column chromatography (hexane:ethyl acetate gradient) to provide the desired compound (P-0027, 163 mg, 45%). MS (ESI) [M+H+]+=406.1.
  • Example 15: Synthesis of Additional Compounds
  • Additional compounds may be synthesized in six steps according to the following Scheme 15 or in four steps according the following Scheme 16.
  • Figure US20170319559A1-20171109-C00110
  • Step 1—Preparation of (2,4-difluoro-phenyl)-carbamic acid methyl ester (52)
  • To 2,4-difluoroaniline 41, potassium carbonate, and water, methyl chloroformate 51 is added slowly dropwise. The reaction is stirred at 0° C. and then allowed to come to room temperature. The reaction mixture is extracted with ethyl acetate and washed with diluted HCl (pH=2), twice with saturated sodium bicarbonate, twice with brine, and dried with magnesium sulfate. Removal of solvent provides the desired compound as a crude solid.
  • Step 2—Preparation of 2,6-difluoro-3-methoxycarbonylamino-benzoic acid (53)
  • To (2,4-difluoro-phenyl)-carbamic acid methyl ester 52 in tetrahydrofuran at −78° C., 2.5 eq. of lithium diisopropylamide is added. After 15 minutes, solid carbon dioxide is added and the reaction is allowed to warm to room temperature. The reaction mixture is extracted with ethyl acetate and washed with diluted HCl (pH=2). The desired compound is isolated by silica gel column chromatography.
  • Step 3—Preparation of (3-amino-2,4-difluoro-phenyl)-carbamic acid methyl ester (54)
  • To 2,6-difluoro-3-methoxycarbonylamino-benzoic acid 53, anhydrous tert-butanol, and triethylamine, diphenylphosphoryl azide is added. The action is heated in an oil bath at 105° C. overnight. The reaction is cooled to room temperature and diluted with ethyl acetate. The organic layer is washed 3× with water, then 1× with brine, dried over magnesium sulfate and filtered and concentrated in vacuo to provide crude Boc-protected amine, which is purified by silica gel column chromatography. The purified Boc-protected amine is dissolved in dichloromethane, trifluoroacetic acid is added and the reaction stirred at room temperature, monitoring by TLC for the disappearance of starting material. The completed reaction is neutralised by pouring into a cooled saturated solution of sodium hydrogen carbonate and then extracted 3× into dichloromethane. The combined organic extracts are washed with brine, dried over magnesium sulfate, filtered and concentrated in vacuo to provide the desired compound.
  • Step 4—Preparation of Compound 56
  • To a reaction vessel, carboxylic acid 55 (Ra is optionally substituted heteroaryl), anhydrous tetrahydrofuran, 1 drop anhydrous dimethylformamide, and oxalyl chloride are added under nitrogen. The reaction mixture is allowed to stir at room temperature for 1.5 hours, then concentrated to dryness. The resulting residue is diluted with anhydrous tetrahydrofuran, then triethylamine and (3-amino-2,4-difluoro-phenyl)-carbamic acid methyl ester 54 are added and allowed to stir at room temperature overnight. The reaction is diluted with water extracted 3× into ethyl acetate. The organic extracts are dried over magnesium sulfate, filtered and concentrated in vacuo to provide the desired compound as a crude solid, which is purified by silica gel column chromatography (hexane: ethyl acetate gradient).
  • Step 5—Preparation of Compound 57
  • To compound 56 in dioxane, an equal volume of 1 N lithium hydroxide is added. The reaction is allowed to stir at 60° C. and monitored by TLC. When complete, reaction is extracted with 1N aqueous HCl and ethyl acetate. The organic layer is dried over anhydrous magnesium sulfate, filtered and volatile solvents removed to provide the desired compound as a crude solid.
  • Step 6—Preparation of Compound 59
  • To compound 57, tetrahydrofuran is added, followed by addition of compound 58 (Rb is di-alkylamino, optionally substituted lower alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl or optionally substituted heteroaryl) as a solution in tetrahydrofuran, then adding pyridine. The reaction vial is allowed to stir at room temperature. After 23 hours, the reaction is poured into water and 1N aqueous HCl and extracted with ethyl acetate. The organic layer is washed with brine, dried over anhydrous magnesium sulfate and filtered. The filtrate is concentrated and purified by silica gel column chromatography (hexane: ethyl acetate gradient) to provide the desired compound.
  • Figure US20170319559A1-20171109-C00111
  • Step 1—Preparation of 2,6-difluoro-3-nitro-phenylamine (61)
  • 2,6-Difluoro-3-nitrobenzoic acid 60 is converted to 2,6-difluoro-3-nitro-phenylamine 61 following the methods described in Scheme 15, Step 3.
  • Step 2—Preparation of Compound 62
  • 2,6-Difluoro-3-nitro-phenylamine 61 is reacted with compound 55 following the methods described in Scheme 15, Step 4 to provide the desired compound 62.
  • Step 3—Preparation of Compound 57
  • To compound 62 in ethanol and tetrahydrofuran, ˜3 cc of raney nickle slurry in water is added. The reaction is placed in a parr hydrogenator under hydrogen at 35 psi and monitored by TLC until all starting material is consumed. The reaction is filtered and all volatile solvents are removed to provide the desired compound as a crude solid.
  • Step 4—Preparation of Compound 59
  • Compound 57 is reacted with compound 58 following the methods described in Scheme 15, Step 6 to provide the desired compound 59.
  • The following compounds may be made following the protocol of either Scheme 15 or Scheme 16:
    • 6-Acetylamino-N-[2,6-difluoro-3-(2-fluoro-benzenesulfonylamino)-phenyl]-nicotinamide (P-0028),
    • 6-Acetylamino-N-[2,6-difluoro-3-(3-fluoro-benzenesulfonylamino)-phenyl]-nicotinamide (P-0029),
    • 6-Acetylamino-N-[3-(2,6-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-nicotinamide (P-0030),
    • 6-Acetylamino-N-[3-(2,4-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-nicotinamide (P-0031),
    • 6-Acetylamino-N-[3-(2,5-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-nicotinamide (P-0032),
    • 6-Acetylamino-N-[2,6-difluoro-3-(3-fluoro-4-methoxy-benzenesulfonylamino)-phenyl]-nicotinamide (P-0033),
    • 6-Acetylamino-N-[2,6-difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-phenyl]-nicotinamide (P-0034),
    • 6-Acetylamino-N-[3-(4-difluoromethoxy-benzenesulfonylamino)-2,6-difluoro-phenyl]-nicotinamide (P-0035),
    • 6-Acetylamino-N-[3-(3-difluoromethoxy-benzenesulfonylamino)-2,6-difluoro-phenyl]-nicotinamide (P-0036),
    • 6-Acetylamino-N-[2,6-difluoro-3-(4-isopropyl-benzenesulfonylamino)-phenyl]-nicotinamide (P-0037),
    • 6-Acetylamino-N-[3-(4-tert-butyl-benzenesulfonylamino)-2,6-difluoro-phenyl]-nicotinamide (P-0038),
    • 6-Acetylamino-N-[2,6-difluoro-3-(4-propyl-benzenesulfonylamino)-phenyl]-nicotinamide (P-0039),
    • 6-Acetylamino-N-[2,6-difluoro-3-(pyridine-2-sulfonylamino)-phenyl]-nicotinamide (P-0040),
    • 6-Acetylamino-N-[2,6-difluoro-3-(pyridine-3-sulfonylamino)-phenyl]-nicotinamide (P-0041),
    • 6-Acetylamino-N-[2,6-difluoro-3-(dimethylaminosulfonylamino)-phenyl]-nicotinamide (P-0042),
    • 6-Acetylamino-N-[2,6-difluoro-3-(piperidine-1-sulfonylamino)-phenyl]-nicotinamide (P-0043),
    • 6-Acetylamino-N-[2,6-difluoro-3-(morpholine-4-sulfonylamino)-phenyl]-nicotinamide (P-0044),
    • 6-Acetylamino-N-[2,6-difluoro-3-(tetrahydro-pyran-4-sulfonylamino)-phenyl]-nicotinamide (P-0045),
    • 6-Acetylamino-N-(3-cyclopentanesulfonylamino-2,6-difluoro-phenyl)-nicotinamide (P-0046),
    • 6-Acetylamino-N-[2,6-difluoro-3-(pyrrolidine-1-sulfonylamino)-phenyl]-nicotinamide (P-0047),
    • 6-Acetylamino-N-[2,6-difluoro-3-(3,3,3-trifluoro-propane-1-sulfonylamino)-phenyl]-nicotinamide (P-0048),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(2-fluoro-benzenesulfonylamino)-phenyl]-amide (P-0049),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(3-fluoro-benzenesulfonylamino)-phenyl]-amide (P-0050),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [3-(2,6-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide (P-0051),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [3-(2,4-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide (P-0052),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [3-(2,5-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide (P-0053),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(3-fluoro-4-methoxy-benzenesulfonylamino)-phenyl]-amide (P-0054),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-phenyl]-amide (P-0055),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [3-(4-difluoromethoxy-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide (P-0056),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [3-(3-difluoromethoxy-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide (P-0057),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(4-isopropyl-benzenesulfonylamino)-phenyl]-amide (P-0058),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [3-(4-tert-butyl-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide (P-0059),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(4-propyl-benzenesulfonylamino)-phenyl]-amide (P-0060),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(pyridine-2-sulfonylamino)-phenyl]-amide (P-0061),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(pyridine-3-sulfonylamino)-phenyl]-amide (P-0062),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(dimethylaminosulfonylamino)-phenyl]-amide (P-0063),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(piperidine-1-sulfonylamino)-phenyl]-amide (P-0064),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(morpholine-4-sulfonylamino)-phenyl]-amide (P-0065),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(tetrahydro-pyran-4-sulfonylamino)-phenyl]-amide (P-0066),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid (3-cyclopentanesulfonylamino-2,6-difluoro-phenyl)-amide (P-0067),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(pyrrolidine-1-sulfonylamino)-phenyl]-amide (P-0068),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(3,3,3-trifluoro-propane-1-sulfonylamino)-phenyl]-amide (P-0069),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(2-fluoro-benzenesulfonylamino)-phenyl]-amide (P-0070),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(3-fluoro-benzenesulfonylamino)-phenyl]-amide (P-0071),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [3-(2,6-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide (P-0072),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [3-(2,4-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide (P-0073),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [3-(2,5-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide (P-0074),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(3-fluoro-4-methoxy-benzenesulfonylamino)-phenyl]-amide (P-0075),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-phenyl]-amide (P-0076),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [3-(4-difluoromethoxy-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide (P-0077),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [3-(3-difluoromethoxy-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide (P-0078),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(4-isopropyl-benzenesulfonylamino)-phenyl]-amide (P-0079),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [3-(4-tert-butyl-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide (P-0080),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(4-propyl-benzenesulfonylamino)-phenyl]-amide (P-0081),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(pyridine-2-sulfonylamino)-phenyl]-amide (P-0082),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(pyridine-3-sulfonylamino)-phenyl]-amide (P-0083),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(dimethylaminosulfonylamino)-phenyl]-amide (P-0084),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(piperidine-1-sulfonylamino)-phenyl]-amide (P-0085),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(morpholine-4-sulfonylamino)-phenyl]-amide (P-0086),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(tetrahydro-pyran-4-sulfonylamino)-phenyl]-amide (P-0087),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid (3-cyclopentanesulfonylamino-2,6-difluoro-phenyl)-amide (P-0088),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(pyrrolidine-1-sulfonylamino)-phenyl]-amide (P-0089),
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(3,3,3-trifluoro-propane-1-sulfonylamino)-phenyl]-amide (P-0090),
    • 6-Acetylamino-N-[2,6-difluoro-3-(propane-1-sulfonylamino)-phenyl]-nicotinamide (P-0118),
    • 1H-Pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(propane-1-sulfonylamino)-phenyl]-amide (P-0119), and
    • 3H-Imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(propane-1-sulfonylamino)-phenyl]-amide (P-0120).
  • These compounds are shown in the following table, where column 1 provides the compound number, column 2 the carboxylic acid compound 55 used in either Step 4 of Scheme 15 or Step 2 of Scheme 16, column 3 the sulfonyl chloride compound 58 used in either Step 6 of Scheme 15 or Step 4 of Scheme 16, and column 4 the resulting compound 59.
  • Com-
    pound
    num-
    ber Compound 55 Compound 58 Resulting Compound 59
    P-0028
    Figure US20170319559A1-20171109-C00112
    Figure US20170319559A1-20171109-C00113
    Figure US20170319559A1-20171109-C00114
    P-0029
    Figure US20170319559A1-20171109-C00115
    Figure US20170319559A1-20171109-C00116
    Figure US20170319559A1-20171109-C00117
    P-0030
    Figure US20170319559A1-20171109-C00118
    Figure US20170319559A1-20171109-C00119
    Figure US20170319559A1-20171109-C00120
    P-0031
    Figure US20170319559A1-20171109-C00121
    Figure US20170319559A1-20171109-C00122
    Figure US20170319559A1-20171109-C00123
    P-0032
    Figure US20170319559A1-20171109-C00124
    Figure US20170319559A1-20171109-C00125
    Figure US20170319559A1-20171109-C00126
    P-0033
    Figure US20170319559A1-20171109-C00127
    Figure US20170319559A1-20171109-C00128
    Figure US20170319559A1-20171109-C00129
    P-0034
    Figure US20170319559A1-20171109-C00130
    Figure US20170319559A1-20171109-C00131
    Figure US20170319559A1-20171109-C00132
    P-0035
    Figure US20170319559A1-20171109-C00133
    Figure US20170319559A1-20171109-C00134
    Figure US20170319559A1-20171109-C00135
    P-0036
    Figure US20170319559A1-20171109-C00136
    Figure US20170319559A1-20171109-C00137
    Figure US20170319559A1-20171109-C00138
    P-0037
    Figure US20170319559A1-20171109-C00139
    Figure US20170319559A1-20171109-C00140
    Figure US20170319559A1-20171109-C00141
    P-0038
    Figure US20170319559A1-20171109-C00142
    Figure US20170319559A1-20171109-C00143
    Figure US20170319559A1-20171109-C00144
    P-0039
    Figure US20170319559A1-20171109-C00145
    Figure US20170319559A1-20171109-C00146
    Figure US20170319559A1-20171109-C00147
    P-0040
    Figure US20170319559A1-20171109-C00148
    Figure US20170319559A1-20171109-C00149
    Figure US20170319559A1-20171109-C00150
    P-0041
    Figure US20170319559A1-20171109-C00151
    Figure US20170319559A1-20171109-C00152
    Figure US20170319559A1-20171109-C00153
    P-0042
    Figure US20170319559A1-20171109-C00154
    Figure US20170319559A1-20171109-C00155
    Figure US20170319559A1-20171109-C00156
    P-0043
    Figure US20170319559A1-20171109-C00157
    Figure US20170319559A1-20171109-C00158
    Figure US20170319559A1-20171109-C00159
    P-0044
    Figure US20170319559A1-20171109-C00160
    Figure US20170319559A1-20171109-C00161
    Figure US20170319559A1-20171109-C00162
    P-0045
    Figure US20170319559A1-20171109-C00163
    Figure US20170319559A1-20171109-C00164
    Figure US20170319559A1-20171109-C00165
    P-0046
    Figure US20170319559A1-20171109-C00166
    Figure US20170319559A1-20171109-C00167
    Figure US20170319559A1-20171109-C00168
    P-0047
    Figure US20170319559A1-20171109-C00169
    Figure US20170319559A1-20171109-C00170
    Figure US20170319559A1-20171109-C00171
    P-0048
    Figure US20170319559A1-20171109-C00172
    Figure US20170319559A1-20171109-C00173
    Figure US20170319559A1-20171109-C00174
    P-0118
    Figure US20170319559A1-20171109-C00175
    Figure US20170319559A1-20171109-C00176
    Figure US20170319559A1-20171109-C00177
    P-0049
    Figure US20170319559A1-20171109-C00178
    Figure US20170319559A1-20171109-C00179
    Figure US20170319559A1-20171109-C00180
    P-0050
    Figure US20170319559A1-20171109-C00181
    Figure US20170319559A1-20171109-C00182
    Figure US20170319559A1-20171109-C00183
    P-0051
    Figure US20170319559A1-20171109-C00184
    Figure US20170319559A1-20171109-C00185
    Figure US20170319559A1-20171109-C00186
    P-0052
    Figure US20170319559A1-20171109-C00187
    Figure US20170319559A1-20171109-C00188
    Figure US20170319559A1-20171109-C00189
    P-0053
    Figure US20170319559A1-20171109-C00190
    Figure US20170319559A1-20171109-C00191
    Figure US20170319559A1-20171109-C00192
    P-0054
    Figure US20170319559A1-20171109-C00193
    Figure US20170319559A1-20171109-C00194
    Figure US20170319559A1-20171109-C00195
    P-0055
    Figure US20170319559A1-20171109-C00196
    Figure US20170319559A1-20171109-C00197
    Figure US20170319559A1-20171109-C00198
    P-0056
    Figure US20170319559A1-20171109-C00199
    Figure US20170319559A1-20171109-C00200
    Figure US20170319559A1-20171109-C00201
    P-0057
    Figure US20170319559A1-20171109-C00202
    Figure US20170319559A1-20171109-C00203
    Figure US20170319559A1-20171109-C00204
    P-0058
    Figure US20170319559A1-20171109-C00205
    Figure US20170319559A1-20171109-C00206
    Figure US20170319559A1-20171109-C00207
    P-0059
    Figure US20170319559A1-20171109-C00208
    Figure US20170319559A1-20171109-C00209
    Figure US20170319559A1-20171109-C00210
    P-0060
    Figure US20170319559A1-20171109-C00211
    Figure US20170319559A1-20171109-C00212
    Figure US20170319559A1-20171109-C00213
    P-0061
    Figure US20170319559A1-20171109-C00214
    Figure US20170319559A1-20171109-C00215
    Figure US20170319559A1-20171109-C00216
    P-0062
    Figure US20170319559A1-20171109-C00217
    Figure US20170319559A1-20171109-C00218
    Figure US20170319559A1-20171109-C00219
    P-0063
    Figure US20170319559A1-20171109-C00220
    Figure US20170319559A1-20171109-C00221
    Figure US20170319559A1-20171109-C00222
    P-0064
    Figure US20170319559A1-20171109-C00223
    Figure US20170319559A1-20171109-C00224
    Figure US20170319559A1-20171109-C00225
    P-0065
    Figure US20170319559A1-20171109-C00226
    Figure US20170319559A1-20171109-C00227
    Figure US20170319559A1-20171109-C00228
    P-0066
    Figure US20170319559A1-20171109-C00229
    Figure US20170319559A1-20171109-C00230
    Figure US20170319559A1-20171109-C00231
    P-0067
    Figure US20170319559A1-20171109-C00232
    Figure US20170319559A1-20171109-C00233
    Figure US20170319559A1-20171109-C00234
    P-0068
    Figure US20170319559A1-20171109-C00235
    Figure US20170319559A1-20171109-C00236
    Figure US20170319559A1-20171109-C00237
    P-0069
    Figure US20170319559A1-20171109-C00238
    Figure US20170319559A1-20171109-C00239
    Figure US20170319559A1-20171109-C00240
    P-0119
    Figure US20170319559A1-20171109-C00241
    Figure US20170319559A1-20171109-C00242
    Figure US20170319559A1-20171109-C00243
    P-0070
    Figure US20170319559A1-20171109-C00244
    Figure US20170319559A1-20171109-C00245
    Figure US20170319559A1-20171109-C00246
    P-0071
    Figure US20170319559A1-20171109-C00247
    Figure US20170319559A1-20171109-C00248
    Figure US20170319559A1-20171109-C00249
    P-0072
    Figure US20170319559A1-20171109-C00250
    Figure US20170319559A1-20171109-C00251
    Figure US20170319559A1-20171109-C00252
    P-0073
    Figure US20170319559A1-20171109-C00253
    Figure US20170319559A1-20171109-C00254
    Figure US20170319559A1-20171109-C00255
    P-0074
    Figure US20170319559A1-20171109-C00256
    Figure US20170319559A1-20171109-C00257
    Figure US20170319559A1-20171109-C00258
    P-0075
    Figure US20170319559A1-20171109-C00259
    Figure US20170319559A1-20171109-C00260
    Figure US20170319559A1-20171109-C00261
    P-0076
    Figure US20170319559A1-20171109-C00262
    Figure US20170319559A1-20171109-C00263
    Figure US20170319559A1-20171109-C00264
    P-0077
    Figure US20170319559A1-20171109-C00265
    Figure US20170319559A1-20171109-C00266
    Figure US20170319559A1-20171109-C00267
    P-0078
    Figure US20170319559A1-20171109-C00268
    Figure US20170319559A1-20171109-C00269
    Figure US20170319559A1-20171109-C00270
    P-0079
    Figure US20170319559A1-20171109-C00271
    Figure US20170319559A1-20171109-C00272
    Figure US20170319559A1-20171109-C00273
    P-0080
    Figure US20170319559A1-20171109-C00274
    Figure US20170319559A1-20171109-C00275
    Figure US20170319559A1-20171109-C00276
    P-0081
    Figure US20170319559A1-20171109-C00277
    Figure US20170319559A1-20171109-C00278
    Figure US20170319559A1-20171109-C00279
    P-0082
    Figure US20170319559A1-20171109-C00280
    Figure US20170319559A1-20171109-C00281
    Figure US20170319559A1-20171109-C00282
    P-0083
    Figure US20170319559A1-20171109-C00283
    Figure US20170319559A1-20171109-C00284
    Figure US20170319559A1-20171109-C00285
    P-0084
    Figure US20170319559A1-20171109-C00286
    Figure US20170319559A1-20171109-C00287
    Figure US20170319559A1-20171109-C00288
    P-0085
    Figure US20170319559A1-20171109-C00289
    Figure US20170319559A1-20171109-C00290
    Figure US20170319559A1-20171109-C00291
    P-0086
    Figure US20170319559A1-20171109-C00292
    Figure US20170319559A1-20171109-C00293
    Figure US20170319559A1-20171109-C00294
    P-0087
    Figure US20170319559A1-20171109-C00295
    Figure US20170319559A1-20171109-C00296
    Figure US20170319559A1-20171109-C00297
    P-0088
    Figure US20170319559A1-20171109-C00298
    Figure US20170319559A1-20171109-C00299
    Figure US20170319559A1-20171109-C00300
    P-0089
    Figure US20170319559A1-20171109-C00301
    Figure US20170319559A1-20171109-C00302
    Figure US20170319559A1-20171109-C00303
    P-0090
    Figure US20170319559A1-20171109-C00304
    Figure US20170319559A1-20171109-C00305
    Figure US20170319559A1-20171109-C00306
    P-0120
    Figure US20170319559A1-20171109-C00307
    Figure US20170319559A1-20171109-C00308
    Figure US20170319559A1-20171109-C00309
  • Example 12: Kinase Activity Assays
  • Assays for the activity of kinases, including, but not limited to, Fms, Kit, B-Raf, B-Raf V600E, B-Raf V600E/T529I and c-Raf-1 are known in the art, for example as described in US Patent Publication Number US20070032519 and U.S. patent application Ser. No. 11/473,347 (see also, PCT publication WO2007002433), the disclosures of which are hereby incorporated by reference in their entireties including all specifications, figures, and tables, and for all purposes.
  • Representative compounds screened by at least one of the methods described above, or by similar methods, having IC50 of less than 10 μM under the test conditions employed are shown in tables 2a (A-Raf), 2b (B-Raf), 2c (B-Raf V600E), 2d (c-Raf-1), 2e (Brk), 2f (Btk), 2g (Csk), 2h (Fak), 2i (Fms), 2j (Kdr), 2k (Kit), 2l (Lck), 2m (Lyn), 2n (Src), 2o (TrkA), and 2p (Yes).
  • TABLE 2a
    Representative compounds with activity toward kinase A-Raf
    with IC50 ≦10 μM under the test conditions employed.
    A-Raf P-0001, P-0002, P-0004, P-0005, P-0006, P-0008, P-0009,
    P-0010, P-0011, P-0012, P-0013, P-0015, P-0016, P-0017,
    P-0020, P-0021, P-0025, P-0026, P-0027
  • TABLE 2b
    Representative compounds with activity toward kinase B-Raf
    with IC50 ≦10 μM under the test conditions employed.
    B-Raf P-0002, P-0004, P-0008, P-0011, P-0013, P-0015, P-0016,
    P-0017, P-0018, P-0027
  • TABLE 2c
    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-0004, P-0005, P-0008, P-0011, P-0013,
    V600E P-0015, P-0016, P-0017, P-0018, P-0020, P-0021, P-0025,
    P-0026, P-0027
  • TABLE 2d
    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-0004, P-0008, P-0009, P-0011, P-0013,
    P-0015, P-0016, P-0017, P-0020, P-0021, P-0027
  • TABLE 2e
    Representative compounds with activity toward kinase Brk
    with IC50 ≦10 μM under the test conditions employed.
    Brk: P-0002
  • TABLE 2f
    Representative compounds with activity toward kinase Btk
    with IC50 ≦10 μM under the test conditions employed.
    Btk: P-0011
  • TABLE 2g
    Representative compounds with activity toward kinase Csk
    with IC50 ≦10 μM under the test conditions employed.
    Csk: P-0002
  • TABLE 2h
    Representative compounds with activity toward kinase Fak
    with IC50 ≦10 μM under the test conditions employed.
    Fak: P-0002
  • TABLE 2i
    Representative compounds with activity toward kinase Fms
    with IC50 ≦10 μM under the test conditions employed.
    Fms: P-0010, P-0026
  • TABLE 2j
    Representative compounds with activity toward kinase Kdr
    with IC50 ≦10 μM under the test conditions employed.
    Kdr: P-0011
  • TABLE 2k
    Representative compounds with activity toward kinase Kit
    with IC50 ≦10 μM under the test conditions employed.
    Kit: P-0011
  • TABLE 2l
    Representative compounds with activity toward kinase Lck
    with IC50 ≦10 μM under the test conditions employed.
    Lck: P-0002
  • TABLE 2m
    Representative compounds with activity toward kinase Lyn
    with IC50 ≦10 μM under the test conditions employed.
    Lyn: P-0002
  • TABLE 2n
    Representative compounds with activity toward kinase Src
    with IC50 ≦10 μM under the test conditions employed.
    Src: P-0002, P-0011
  • TABLE 2o
    Representative compounds with activity toward kinase TrkA
    with IC50 ≦10 μM under the test conditions employed.
    TrkA: P-0002, P-0011
  • TABLE 2p
    Representative compounds with activity toward kinase Yes
    with IC50 ≦10 μM under the test conditions employed.
    Yes: P-0002
  • Example 13: 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, 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. Such assays are known in the art, for example, as described in U.S. patent application Ser. No. 11/473,347.
  • Additional examples of certain methods contemplated by the present invention may be found in the following applications: U.S. Patent Publ. No. 2006/058339; U.S. Patent Publ. No. 2006/058340; U.S. Patent Publ. No. 2007/0032519; and U.S. patent application Ser. No. 11/473,347, filed Jun. 21, 2006 (Equivalent to PCT published as WO 2007/002433), 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.
  • 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 (11)

What is claimed is:
1. A method for treating a subject suffering from or at risk of a Raf mediated disease or condition, comprising administering to the subject an effective amount of a compound selected from the group consisting of:
6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-N-pyridin-3-yl-benzamide,
N-(6-acetylamino-pyridin-3-yl)-6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzamide,
6-chloro-2-fluoro-N-(6-methoxy-pyridin-3-yl)-3-(propane-1-sulfonylamino)-benzamide,
N-(2-acetylamino-pyrimidin-5-yl)-6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzamide,
6-chloro-2-fluoro-N-(6-isopropylamino-pyridin-3-yl)-3-(propane-1-sulfonylamino)-benzamide,
pyrrolidine-1-carboxylic acid {5-[6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoylamino]-pyridin-2-yl}-amide,
6-chloro-N-(3,5-dimethyl-isoxazol-4-yl)-2-fluoro-3-(propane-1-sulfonylamino)-benzamide,
N-(6-acetylamino-pyridin-3-yl)-2,6-difluoro-3-(propane-1-sulfonylamino)-benzamide,
6-chloro-N-(6-cyclopentylamino-pyridin-3-yl)-2-fluoro-3-(propane-1-sulfonylamino)-benzamide,
6-chloro-N-[5-(4-chloro-phenyl)-2H-pyrazol-3-yl]-2-fluoro-3-(propane-1-sulfonylamino)-benzamide,
6-chloro-2-fluoro-N-[6-(5-methyl-thiazol-2-ylamino)-pyridin-3-yl]-3-(propane-1-sulfonylamino)-benzamide,
6-chloro-N-[5-(4-chloro-benzyl)-[1,3,4]thiadiazol-2-yl]-2-fluoro-3-(propane-1-sulfonylamino)-benzamide,
6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-N-quinolin-3-yl-benzamide,
[2-[6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzoylamino]-4H-[1,3,4]thiadiazin-(5E)-ylidene]-acetic acid ethyl ester,
6-chloro-N-(6-cyclopropylamino-pyridin-3-yl)-2-fluoro-3-(propane-1-sulfonylamino)-benzamide,
6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-N-{6-[(thiophen-2-ylmethyl)-amino]-pyridin-3-yl}-benzamide,
N-(6-benzylamino-pyridin-3-yl)-6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzamide,
6-chloro-2-fluoro-N-imidazo[1,2-a]pyridin-3-yl-3-(propane-1-sulfonylamino)-benzamide, propane-1-sulfonic acid {2,4-difluoro-3-[(5-methyl-isoxazol-3-ylamino)-methyl]-phenyl}-amide,
N-{5-[2,6-difluoro-3-(propane-1-sulfonylamino)-benzylamino]-pyridin-2-yl}-acetamide,
propane-1-sulfonic acid [2,4-difluoro-3-(quinolin-3-ylaminomethyl)-phenyl]-amide,
propane-1-sulfonic acid {3-[(6-chloro-pyridin-3-ylamino)-methyl]-2,4-difluoro-phenyl}-amide,
propane-1-sulfonic acid {2,4-difluoro-3-[(6-methoxy-pyridin-3-ylamino)-methyl]-phenyl}-amide,
quinoline-3-carboxylic acid [2,6-difluoro-3-(propane-1-sulfonylamino)-phenyl]-amide,
propane-1-sulfonic acid {2,4-difluoro-3-[(quinolin-3-ylmethyl)-amino]-phenyl}-amide,
propane-1-sulfonic acid [2,4-difluoro-3-(quinolin-3-yloxymethyl)-phenyl]-amide,
2,6-difluoro-3-(propane-1-sulfonylamino)-N-quinolin-3-yl-benzamide,
6-acetylamino-N-[2,6-difluoro-3-(2-fluoro-benzenesulfonylamino)-phenyl]-nicotinamide,
6-acetylamino-N-[2,6-difluoro-3-(3-fluoro-benzenesulfonylamino)-phenyl]-nicotinamide,
6-acetylamino-N-[3-(2,6-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-nicotinamide,
6-acetylamino-N-[3-(2,4-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-nicotinamide,
6-acetylamino-N-[3-(2,5-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-nicotinamide,
6-acetylamino-N-[2,6-difluoro-3-(3-fluoro-4-methoxy-benzenesulfonylamino)-phenyl]-nicotinamide,
6-acetylamino-N-[2,6-difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-phenyl]-nicotinamide,
6-acetylamino-N-[3-(4-difluoromethoxy-benzenesulfonylamino)-2,6-difluoro-phenyl]-nicotinamide,
6-acetylamino-N-[3-(3-difluoromethoxy-benzenesulfonylamino)-2,6-difluoro-phenyl]-nicotinamide,
6-acetylamino-N-[2,6-difluoro-3-(4-isopropyl-benzenesulfonylamino)-phenyl]-nicotinamide,
6-acetylamino-N-[3-(4-tert-butyl-benzenesulfonylamino)-2,6-difluoro-phenyl]-nicotinamide,
6-acetylamino-N-[2,6-difluoro-3-(4-propyl-benzenesulfonylamino)-phenyl]-nicotinamide,
6-acetylamino-N-[2,6-difluoro-3-(pyridine-2-sulfonylamino)-phenyl]-nicotinamide,
6-acetylamino-N-[2,6-difluoro-3-(pyridine-3-sulfonylamino)-phenyl]-nicotinamide,
6-acetylamino-N-[2,6-difluoro-3-(dimethylaminosulfonylamino)-phenyl]-nicotinamide,
6-acetylamino-N-[2,6-difluoro-3-(piperidine-1-sulfonylamino)-phenyl]-nicotinamide,
6-acetylamino-N-[2,6-difluoro-3-(morpholine-4-sulfonylamino)-phenyl]-nicotinamide,
6-acetylamino-N-[2,6-difluoro-3-(tetrahydro-pyran-4-sulfonylamino)-phenyl]-nicotinamide,
6-acetylamino-N-(3-cyclopentanesulfonylamino-2,6-difluoro-phenyl)-nicotinamide,
6-acetylamino-N-[2,6-difluoro-3-(pyrrolidine-1-sulfonylamino)-phenyl]-nicotinamide,
6-acetylamino-N-[2,6-difluoro-3-(3,3,3-trifluoro-propane-1-sulfonylamino)-phenyl]-nicotinamide,
1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(2-fluoro-benzenesulfonylamino)-phenyl]-amide,
1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(3-fluoro-benzenesulfonylamino)-phenyl]-amide,
1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid [3-(2,6-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide,
1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid [3-(2,4-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide,
1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid [3-(2,5-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide,
1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(3-fluoro-4-methoxy-benzenesulfonylamino)-phenyl]-amide,
1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-phenyl]-amide,
1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid [3-(4-difluoromethoxy-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide,
1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid [3-(3-difluoromethoxy-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide,
1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(4-isopropyl-benzenesulfonylamino)-phenyl]-amide,
1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid [3-(4-tert-butyl-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide,
1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(4-propyl-benzenesulfonylamino)-phenyl]-amide,
1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(pyridine-2-sulfonylamino)-phenyl]-amide,
1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(pyridine-3-sulfonylamino)-phenyl]-amide,
1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(dimethylaminosulfonylamino)-phenyl]-amide,
1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(piperidine-1-sulfonylamino)-phenyl]-amide,
1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(morpholine-4-sulfonylamino)-phenyl]-amide,
1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(tetrahydro-pyran-4-sulfonylamino)-phenyl]-amide,
1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid (3-cyclopentanesulfonylamino-2,6-difluoro-phenyl)-amide,
1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(pyrrolidine-1-sulfonylamino)-phenyl]-amide,
1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(3,3,3-trifluoro-propane-1-sulfonylamino)-phenyl]-amide,
3H-imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(2-fluoro-benzenesulfonylamino)-phenyl]-amide,
3H-imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(3-fluoro-benzenesulfonylamino)-phenyl]-amide,
3H-imidazo[4,5-b]pyridine-6-carboxylic acid [3-(2,6-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide,
3H-imidazo[4,5-b]pyridine-6-carboxylic acid [3-(2,4-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide,
3H-imidazo[4,5-b]pyridine-6-carboxylic acid [3-(2,5-difluoro-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide,
3H-imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(3-fluoro-4-methoxy-benzenesulfonylamino)-phenyl]-amide,
3H-imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-phenyl]-amide,
3H-imidazo[4,5-b]pyridine-6-carboxylic acid [3-(4-difluoromethoxy-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide,
3H-imidazo[4,5-b]pyridine-6-carboxylic acid [3-(3-difluoromethoxy-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide,
3H-imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(4-isopropyl-benzenesulfonylamino)-phenyl]-amide,
3H-imidazo[4,5-b]pyridine-6-carboxylic acid [3-(4-tert-butyl-benzenesulfonylamino)-2,6-difluoro-phenyl]-amide,
3H-imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(4-propyl-benzenesulfonylamino)-phenyl]-amide,
3H-imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(pyridine-2-sulfonylamino)-phenyl]-amide,
3H-imidazo [4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(pyridine-3-sulfonylamino)-phenyl]-amide,
3H-imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(dimethylaminosulfonylamino)-phenyl]-amide,
3H-imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(piperidine-1-sulfonylamino)-phenyl]-amide,
3H-imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(morpholine-4-sulfonylamino)-phenyl]-amide,
3H-imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(tetrahydro-pyran-4-sulfonylamino)-phenyl]-amide,
3H-imidazo[4,5-b]pyridine-6-carboxylic acid (3-cyclopentanesulfonylamino-2,6-difluoro-phenyl)-amide,
3H-imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(pyrrolidine-1-sulfonylamino)-phenyl]-amide,
3H-imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(3,3,3-trifluoro-propane-1-sulfonylamino)-phenyl]-amide,
N-{5-[2,6-difluoro-3-(propane-1-sulfonyl amino)-benzyloxy]-pyridin-2-yl}-acetamide,
propane-1-sulfonic acid [3-(2-amino-pyridin-3-yloxymethyl)-2,4-difluoro-phenyl]-amide,
propane-1-sulfonic acid [2,4-difluoro-3-(1H-pyrrolo[2,3-b]pyridin-5-yloxymethyl)-phenyl]-amide,
N-{5-[2,6-difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzyloxy]-pyridin-2-yl}-acetamide,
N-[3-(2-amino-pyridin-3-yloxymethyl)-2,4-difluoro-phenyl]-4-trifluoromethyl-benzenesulfonamide,
N-[2,4-difluoro-3-(1H-pyrrolo[2,3-b]pyridin-5-yloxymethyl)-phenyl]-4-trifluoromethyl-benzenesulfonamide,
N-(6-acetylamino-pyridin-3-yl)-2,6-difluoro-3-(2-fluoro-benzenesulfonylamino)-benzamide,
N-(6-acetylamino-pyridin-3-yl)-2,6-difluoro-3-(3-fluoro-benzenesulfonylamino)-benzamide,
N-(6-acetylamino-pyridin-3-yl)-3-(2,6-difluoro-benzenesulfonylamino)-2,6-difluoro-benzamide, N-(6-acetylamino-pyridin-3-yl)-3-(2,4-difluoro-benzenesulfonylamino)-2,6-difluoro-benzamide, N-(6-acetylamino-pyridin-3-yl)-3-(2,5-difluoro-benzenesulfonylamino)-2,6-difluoro-benzamide, 6-acetylamino-N-[2,6-difluoro-3-(3-fluoro-4-methoxy-benzenesulfonylamino)-phenyl]-nicotinamide,
N-(6-acetylamino-pyridin-3-yl)-2,6-difluoro-3-(4-trifluoromethyl-benzenesulfonylamino)-benzamide,
N-(6-acetylamino-pyridin-3-yl)-3-(4-difluoromethoxy-benzenesulfonylamino)-2,6-difluoro-benzamide,
N-(6-acetylamino-pyridin-3-yl)-3-(3-difluoromethoxy-benzenesulfonylamino)-2,6-difluoro-benzamide,
N-(6-acetylamino-pyridin-3-yl)-2,6-difluoro-3-(4-isopropyl-benzenesulfonylamino)-benzamide, N-(6-acetylamino-pyridin-3-yl)-3-(4-tert-butyl-benzenesulfonylamino)-2,6-difluoro-benzamide, N-(6-acetylamino-pyridin-3-yl)-2,6-difluoro-3-(4-propyl-benzenesulfonylamino)-benzamide,
N-(6-acetylamino-pyridin-3-yl)-2,6-difluoro-3-(pyridine-2-sulfonylamino)-benzamide,
N-(6-acetylamino-pyridin-3-yl)-2,6-difluoro-3-(pyridine-3-sulfonylamino)-benzamide,
N-(6-acetylamino-pyridin-3-yl)-2,6-difluoro-3-(dimethylaminosulfonylamino)-benzamide,
N-(6-acetylamino-pyridin-3-yl)-2,6-difluoro-3-(piperidine-1-sulfonylamino)-benzamide,
N-(6-acetylamino-pyridin-3-yl)-2,6-difluoro-3-(morpholine-4-sulfonylamino)-benzamide,
N-(6-acetylamino-pyridin-3-yl)-2,6-difluoro-3-(tetrahydro-pyran-4-sulfonylamino)-benzamide,
N-(6-acetylamino-pyridin-3-yl)-3-cyclopentanesulfonylamino-2,6-difluoro-benzamide,
N-(6-acetylamino-pyridin-3-yl)-2,6-difluoro-3-(pyrrolidine-1-sulfonylamino)-benzamide,
N-(6-acetylamino-pyridin-3-yl)-2,6-difluoro-3-(3,3,3-trifluoro-propane-1-sulfonylamino)-benzamide,
6-acetylamino-N-[2,6-difluoro-3-(propane-1-sulfonylamino)-phenyl]-nicotinamide,
1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid [2,6-difluoro-3-(propane-1-sulfonylamino)-phenyl]-amide, and
3H-imidazo[4,5-b]pyridine-6-carboxylic acid [2,6-difluoro-3-(propane-1-sulfonylamino)-phenyl]-amide,
or a pharmaceutically acceptable salt of any of the above compounds.
2. A method for treating a subject suffering from or at risk of a B-Raf V600E mediated disease or condition, comprising administering to the subject an effective amount of a compound selected from the group consisting of:
6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-N-pyridin-3-yl-benzamide,
N-(6-acetylamino-pyridin-3-yl)-6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzamide,
N-(2-acetylamino-pyrimidin-5-yl)-6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzamide,
6-chloro-2-fluoro-N-(6-isopropylamino-pyridin-3-yl)-3-(propane-1-sulfonylamino)-benzamide,
N-(6-acetylamino-pyridin-3-yl)-2,6-difluoro-3-(propane-1-sulfonylamino)-benzamide,
6-chloro-2-fluoro-N-[6-(5-methyl-thiazol-2-ylamino)-pyridin-3-yl]-3-(propane-1-sulfonylamino)-benzamide,
6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-N-quinolin-3-yl-benzamide,
6-chloro-N-(6-cyclopropylamino-pyridin-3-yl)-2-fluoro-3-(propane-1-sulfonylamino)-benzamide,
6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-N-{6-[(thiophen-2-ylmethyl)-amino]-pyridin-3-yl}-benzamide,
N-(6-benzylamino-pyridin-3-yl)-6-chloro-2-fluoro-3-(propane-1-sulfonylamino)-benzamide,
6-chloro-2-fluoro-N-imidazo[1,2-a]pyridin-3-yl-3-(propane-1-sulfonylamino)-benzamide,
N-{5-[2,6-difluoro-3-(propane-1-sulfonylamino)-benzylamino]-pyridin-2-yl}-acetamide,
propane-1-sulfonic acid [2,4-difluoro-3-(quinolin-3-ylaminomethyl)-phenyl]-amide,
propane-1-sulfonic acid {2,4-difluoro-3-[(quinolin-3-ylmethyl)-amino]-phenyl}-amide,
propane-1-sulfonic acid [2,4-difluoro-3-(quinolin-3-yloxymethyl)-phenyl]-amide, and
2,6-difluoro-3-(propane-1-sulfonylamino)-N-quinolin-3-yl-benzamide,
or a pharmaceutically acceptable salt of any of the above compounds.
3. The method according to claim 1, wherein the Raf mediated disease or condition is a neurological disease selected from the group consisting of multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease and Parkinson's disease.
4. The method according to claim 1, wherein the Raf mediated disease or condition is a neoplastic disease selected from the group consisting of melanoma, glioma, sarcoma, colorectal cancer, lung cancer, breast cancer, pancreatic cancer, thyroid cancer, renal cancer, ovarian cancer, histiocytic lymphoma, neurofibromatosis, acute myeloid leukemia, myelodysplastic syndrome, leukemia, tumor angiogenesis, medullary thyroid cancer, carcinoid, small cell lung cancer and pheochromocytoma.
5. The method according to claim 1, wherein the Raf mediated disease or condition is a pain of neuropathic or inflammatory origin selected from the group consisting of acute pain, chronic pain, cancer-related pain and migraine.
6. The method according to claim 1, wherein the Raf mediated disease or condition is a cardiovascular disease selected from the group consisting of heart failure, ischemic stroke, cardiac hypertrophy, thrombosis, atherosclerosis and reperfusion injury.
7. The method according to claim 1, wherein the Raf mediated disease or condition is an inflammatory disease selected from the group consisting of ipsoriasis, arthritis and autoimmune diseases and conditions, osteoarthritis, endometriosis, scarring, vascular restenosis, fibrotic disorders, rheumatoid arthritis and inflammatory bowel disease.
8. The method according to claim 1, wherein the Raf mediated disease or condition is a renal or prostatic disease selected from the group consisting of diabetic nephropathy, polycystic kidney disease, nephrosclerosis, glomerulonephritis and prostate hyperplasia.
9. The method according to claim 1, wherein the Raf mediated disease or condition is a pulmonary disease selected from the group consisting of chronic obstructive pulmonary disease and acute respiratory distress syndrome.
10. The method according to claim 1, wherein the Raf mediated disease or condition is a genetic developmental disease selected from the group consisting of Noonan's syndrome, Costello syndrome, LEOPARD syndrome, cardio-faciocutaneous syndrome, and neural crest syndrome abnormalities causing cardiovascular, skeletal, intestinal, skin, hair and endocrine disease.
11. The method according to claim 2, wherein the B-Raf V600E mediated disease or condition is malignant melanoma.
US15/656,990 2007-07-17 2017-07-21 Compounds and methods for kinase modulation, and indications therefor Abandoned US20170319559A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
US15/656,990 US20170319559A1 (en) 2007-07-17 2017-07-21 Compounds and methods for kinase modulation, and indications therefor
US16/058,945 US20190209536A1 (en) 2007-07-17 2018-08-08 Compounds and methods for kinase modulation, and indications therefor
US16/109,199 US10426760B2 (en) 2007-07-17 2018-08-22 Compounds and methods for kinase modulation, and indications therefor
US17/372,330 US20210346358A1 (en) 2007-07-17 2021-07-09 Compounds and methods for kinase modulation, and indications therefor
US17/372,346 US20210353602A1 (en) 2007-07-17 2021-07-09 Compounds and methods for kinase modulation, and indications therefor

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US95990707P 2007-07-17 2007-07-17
PCT/US2008/070124 WO2009012283A1 (en) 2007-07-17 2008-07-16 Compounds and methods for kinase modulation, and indications therefor
US66945010A 2010-01-15 2010-01-15
US13/866,353 US20130237531A1 (en) 2007-07-17 2013-04-19 Compounds and methods for kinase modulation, and indications therefor
US13/926,959 US20140038948A1 (en) 2007-07-17 2013-06-25 Compounds and methods for kinase modulation, and indications therefor
US15/048,851 US9469640B2 (en) 2007-07-17 2016-02-19 Compounds and methods for kinase modulation, and indications therefor
US15/260,042 US9844539B2 (en) 2007-07-17 2016-09-08 Compounds and methods for kinase modulation, and indications therefor
US15/656,990 US20170319559A1 (en) 2007-07-17 2017-07-21 Compounds and methods for kinase modulation, and indications therefor

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/260,042 Continuation US9844539B2 (en) 2007-07-17 2016-09-08 Compounds and methods for kinase modulation, and indications therefor

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US16/058,945 Continuation US20190209536A1 (en) 2007-07-17 2018-08-08 Compounds and methods for kinase modulation, and indications therefor
US16/109,199 Continuation US10426760B2 (en) 2007-07-17 2018-08-22 Compounds and methods for kinase modulation, and indications therefor

Publications (1)

Publication Number Publication Date
US20170319559A1 true US20170319559A1 (en) 2017-11-09

Family

ID=39745045

Family Applications (11)

Application Number Title Priority Date Filing Date
US12/669,450 Abandoned US20100190777A1 (en) 2007-07-17 2008-07-16 Compounds and methods for kinase modulation, and indications therefor
US13/866,353 Abandoned US20130237531A1 (en) 2007-07-17 2013-04-19 Compounds and methods for kinase modulation, and indications therefor
US13/926,959 Abandoned US20140038948A1 (en) 2007-07-17 2013-06-25 Compounds and methods for kinase modulation, and indications therefor
US14/245,770 Abandoned US20140303187A1 (en) 2007-07-17 2014-04-04 Compounds and methods for kinase modulation, and indications therefor
US15/048,851 Active US9469640B2 (en) 2007-07-17 2016-02-19 Compounds and methods for kinase modulation, and indications therefor
US15/260,042 Active US9844539B2 (en) 2007-07-17 2016-09-08 Compounds and methods for kinase modulation, and indications therefor
US15/656,990 Abandoned US20170319559A1 (en) 2007-07-17 2017-07-21 Compounds and methods for kinase modulation, and indications therefor
US16/058,945 Abandoned US20190209536A1 (en) 2007-07-17 2018-08-08 Compounds and methods for kinase modulation, and indications therefor
US16/109,199 Active US10426760B2 (en) 2007-07-17 2018-08-22 Compounds and methods for kinase modulation, and indications therefor
US17/372,346 Abandoned US20210353602A1 (en) 2007-07-17 2021-07-09 Compounds and methods for kinase modulation, and indications therefor
US17/372,330 Abandoned US20210346358A1 (en) 2007-07-17 2021-07-09 Compounds and methods for kinase modulation, and indications therefor

Family Applications Before (6)

Application Number Title Priority Date Filing Date
US12/669,450 Abandoned US20100190777A1 (en) 2007-07-17 2008-07-16 Compounds and methods for kinase modulation, and indications therefor
US13/866,353 Abandoned US20130237531A1 (en) 2007-07-17 2013-04-19 Compounds and methods for kinase modulation, and indications therefor
US13/926,959 Abandoned US20140038948A1 (en) 2007-07-17 2013-06-25 Compounds and methods for kinase modulation, and indications therefor
US14/245,770 Abandoned US20140303187A1 (en) 2007-07-17 2014-04-04 Compounds and methods for kinase modulation, and indications therefor
US15/048,851 Active US9469640B2 (en) 2007-07-17 2016-02-19 Compounds and methods for kinase modulation, and indications therefor
US15/260,042 Active US9844539B2 (en) 2007-07-17 2016-09-08 Compounds and methods for kinase modulation, and indications therefor

Family Applications After (4)

Application Number Title Priority Date Filing Date
US16/058,945 Abandoned US20190209536A1 (en) 2007-07-17 2018-08-08 Compounds and methods for kinase modulation, and indications therefor
US16/109,199 Active US10426760B2 (en) 2007-07-17 2018-08-22 Compounds and methods for kinase modulation, and indications therefor
US17/372,346 Abandoned US20210353602A1 (en) 2007-07-17 2021-07-09 Compounds and methods for kinase modulation, and indications therefor
US17/372,330 Abandoned US20210346358A1 (en) 2007-07-17 2021-07-09 Compounds and methods for kinase modulation, and indications therefor

Country Status (12)

Country Link
US (11) US20100190777A1 (en)
EP (1) EP2170830B1 (en)
JP (5) JP2010533729A (en)
CN (1) CN101808994B (en)
AU (1) AU2008276063B2 (en)
BR (1) BRPI0814423B1 (en)
CA (1) CA2695004C (en)
IL (2) IL203331A (en)
MX (1) MX2010000617A (en)
NZ (1) NZ582772A (en)
SG (1) SG183036A1 (en)
WO (1) WO2009012283A1 (en)

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9994567B2 (en) 2015-05-22 2018-06-12 Plexxikon Inc. Synthesis of heterocyclic compounds
US10123998B2 (en) 2012-03-19 2018-11-13 Plexxikon Inc. Kinase modulation, and indications therefor
US10160747B2 (en) 2016-03-16 2018-12-25 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10189833B2 (en) 2015-05-06 2019-01-29 Plexxikon Inc. Solid forms of a compound modulating kinases
US10301280B2 (en) 2012-12-21 2019-05-28 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10399975B2 (en) 2015-05-06 2019-09-03 Plexxikon Inc. Synthesis of a compound that modulates kinases
US10421761B2 (en) 2013-05-30 2019-09-24 Plexxikon Inc. Compounds for kinase modulation, and indications therefor
US10428067B2 (en) 2017-06-07 2019-10-01 Plexxikon Inc. Compounds and methods for kinase modulation
US10426760B2 (en) 2007-07-17 2019-10-01 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10435404B2 (en) 2017-07-25 2019-10-08 Plexxikon Inc. Formulations of a compound modulating kinases
US10501460B2 (en) 2013-03-15 2019-12-10 Plexxikon Inc. Heterocyclic compounds and uses thereof
US10508085B2 (en) 2016-09-22 2019-12-17 Plexxikon Inc. Compounds and methods for IDO and TDO modulation, and indications therefor
US10519177B2 (en) 2013-03-15 2019-12-31 Plexxikon Inc. Heterocyclic compounds and uses thereof
US10577366B2 (en) 2017-03-20 2020-03-03 Plexxikon Inc. Crystalline forms of a compound that inhibits bromodomain
US10647716B2 (en) 2015-09-21 2020-05-12 Plexxikon Inc. Heterocyclic compounds and uses thereof
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
WO2020176579A1 (en) * 2019-02-26 2020-09-03 Aerie Pharmaceuticals, Inc. Thienyl cyclopropyl-amino-isoquinolinyl amide compounds
US10829484B2 (en) 2015-07-28 2020-11-10 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
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
US11337976B2 (en) 2011-02-07 2022-05-24 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor

Families Citing this family (119)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008063888A2 (en) 2006-11-22 2008-05-29 Plexxikon, Inc. Compounds modulating c-fms and/or c-kit activity and uses therefor
CL2008001943A1 (en) 2007-07-02 2009-09-11 Boehringer Ingelheim Int Compounds derived from phenyl-triazole, inhibitors of specific signal enzymes that participate in the control of cell proliferation; pharmaceutical composition comprising said compounds; and its use to treat cancer, infections, inflammatory and autoimmune diseases.
JP2011513329A (en) * 2008-02-29 2011-04-28 アレイ バイオファーマ、インコーポレイテッド Imidazo [4,5-b] pyridine derivatives used as RAF inhibitory compounds
TW200940540A (en) 2008-02-29 2009-10-01 Array Biopharma Inc RAF inhibitor compounds and methods of use thereof
CN102149712A (en) 2008-02-29 2011-08-10 阵列生物制药公司 Pyrazole [3, 4-b] pyridine Raf inhibitors
WO2009111280A1 (en) 2008-02-29 2009-09-11 Array Biopharma Inc. N- (6-aminopyridin-3-yl) -3- (sulfonamido) benzamide derivatives as b-raf inhibitors for the treatment of cancer
UA103319C2 (en) 2008-05-06 2013-10-10 Глаксосмитклайн Ллк Thiazole- and oxazole-benzene sulfonamide compounds
US8158636B2 (en) 2008-05-19 2012-04-17 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
EP2303861B1 (en) * 2008-06-17 2014-04-23 AstraZeneca AB Pyridine compounds
AR073501A1 (en) 2008-09-08 2010-11-10 Boehringer Ingelheim Int PYRIMID DERIVATIVES [5,4-D] PYRIMIDINE INHIBITORS OF THYROSINOQUINASE
EP2350052B1 (en) 2008-09-29 2014-08-13 Boehringer Ingelheim International GmbH Antiproliferative compounds
CA2752265A1 (en) 2009-02-17 2010-08-26 Boehringer Ingelheim International Gmbh Pyrimido [5,4-d] pyrimidine derivatives for the inhibition of tyrosine kinases
MX349923B (en) 2009-04-03 2017-08-21 Hoffmann La Roche Propane- i-sulfonic acid {3- [5- (4 -chloro-phenyl) -1h-pyrrolo [2, 3-b] pyridine-3-carbonyl] -2, 4-difluoro-pheny l } -amide compositions and uses thereof.
TW201041888A (en) * 2009-05-06 2010-12-01 Plexxikon Inc Compounds and methods for kinase modulation, and indications therefor
AR077033A1 (en) 2009-06-11 2011-07-27 Hoffmann La Roche INHIBITING COMPOUNDS OF JANUS KINASES AND THEIR USE IN THE TREATMENT OF IMMUNOLOGICAL DISEASES
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
US20120157439A1 (en) 2009-08-28 2012-06-21 Genentech, Inc. Raf inhibitor compounds and methods of use thereof
JP2013503194A (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 1H-pyrazolo [3,4-B] pyridine compounds for inhibiting Raf kinase
SG178900A1 (en) 2009-08-28 2012-04-27 Genentech Inc Raf inhibitor compounds and methods of use thereof
CA2771893A1 (en) * 2009-08-28 2011-03-03 Array Biopharma Inc. Raf inhibitor compounds and methods of use thereof
WO2011025938A2 (en) 2009-08-28 2011-03-03 Array Biopharma Inc. Raf inhibitor compounds and methods of use thereof
CA2771895A1 (en) 2009-08-28 2011-03-03 Array Biopharma Inc. Raf inhibitor compounds and methods of use thereof
JP2013510166A (en) 2009-11-06 2013-03-21 プレキシコン インコーポレーテッド Compounds, methods and applications for kinase regulation
WO2011060216A1 (en) 2009-11-12 2011-05-19 Concert Pharmaceuticals Inc. Substituted azaindoles
AU2010321883A1 (en) * 2009-11-18 2012-05-31 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
WO2011079133A2 (en) 2009-12-23 2011-06-30 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
WO2011117382A1 (en) 2010-03-26 2011-09-29 Boehringer Ingelheim International Gmbh Pyridyltriazoles
TWI619713B (en) 2010-04-21 2018-04-01 普雷辛肯公司 Compounds and methods for kinase modulation, and indications therefor
CA2803055A1 (en) * 2010-06-25 2011-12-29 Novartis Ag Heteroaryl compounds and compositions as protein kinase inhibitors
EP2616467A1 (en) 2010-09-13 2013-07-24 Concert Pharmaceuticals Inc. Substituted azaindoles
US8710055B2 (en) 2010-12-21 2014-04-29 Boehringer Ingelheim International Gmbh Triazolylphenyl sulfonamides as serine/threonine kinase inhibitors
US20130023531A1 (en) 2011-01-27 2013-01-24 Boehringer Ingelheim International Gmbh Pyrimido[5,4-d]pyrimidylamino phenyl sulfonamides as serine/threonine kinase inhibitors
US8889684B2 (en) 2011-02-02 2014-11-18 Boehringer Ingelheim International Gmbh Azaindolylphenyl sulfonamides as serine/threonine kinase inhibitors
US9199979B2 (en) 2011-02-24 2015-12-01 Nerviano Medical Sciences S.R.L. Thiazolylphenyl-benzenesulfonamido derivatives as kinase inhibitors
WO2012118492A1 (en) * 2011-03-01 2012-09-07 Array Biopharma Inc. Heterocyclic sulfonamides as raf inhibitors
BR112013025792A2 (en) 2011-04-05 2018-04-24 Pfizer Ltd PYRROID-RELATED KINASE INHIBITORS [2,3-D] PYRIMIDINE TROPOMISINE
AU2012255275B2 (en) 2011-05-17 2016-01-28 Plexxikon Inc. Kinase modulation and indications therefor
BR112013029758A2 (en) 2011-05-20 2018-10-09 Capital, Business Y Gestion De Finanzas S.L. pharmaceutical composition
AU2012282229B2 (en) 2011-07-08 2015-05-07 Novartis Ag Novel pyrrolo pyrimidine derivatives
WO2013029043A1 (en) * 2011-08-25 2013-02-28 Reaction Biology Corp. Selective kinase inhibitors
ES2725790T3 (en) * 2011-08-26 2019-09-27 Neupharma Inc Some chemical entities, compositions, and methods
US8883819B2 (en) 2011-09-01 2014-11-11 Irm Llc Bicyclic heterocycle derivatives for the treatment of pulmonary arterial hypertension
CN108794411B (en) * 2011-09-14 2022-06-07 润新生物公司 Certain chemical entities, compositions, and methods
US9249110B2 (en) 2011-09-21 2016-02-02 Neupharma, Inc. Substituted quinoxalines as B-raf kinase inhibitors
US9249111B2 (en) 2011-09-30 2016-02-02 Neupharma, Inc. Substituted quinoxalines as B-RAF kinase inhibitors
JP5868521B2 (en) 2011-12-31 2016-02-24 ベイジーン リミテッド Condensed tricyclic compounds as Raf kinase inhibitors
WO2013112950A2 (en) 2012-01-25 2013-08-01 Neupharma, Inc. Certain chemical entities, compositions, and methods
CA2866324C (en) 2012-03-13 2019-01-15 The Chinese University Of Hong Kong Methods for analyzing massively parallel sequencing data for noninvasive prenatal diagnosis
US9150570B2 (en) 2012-05-31 2015-10-06 Plexxikon Inc. Synthesis of heterocyclic compounds
AU2013312477B2 (en) 2012-09-06 2018-05-31 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9688635B2 (en) 2012-09-24 2017-06-27 Neupharma, Inc. Certain chemical entities, compositions, and methods
JP2015531393A (en) 2012-10-04 2015-11-02 ファイザー・リミテッドPfizer Limited Tropomyosin-related kinase inhibitor
EP2912036A1 (en) 2012-10-04 2015-09-02 Pfizer Limited Pyrrolo[3,2-c]pyridine tropomyosin-related kinase inhibitors
WO2014053967A1 (en) 2012-10-04 2014-04-10 Pfizer Limited Pyrrolo[2,3-d]pyrimidine tropomyosin-related kinase inhibitors
US9725421B2 (en) 2012-11-12 2017-08-08 Neupharma, Inc. Substituted quinoxalines as B-raf kinase inhibitors
CN103834003B (en) * 2012-11-26 2019-02-19 杨子剑 Noval chemical compound and preparation method and purposes containing Crizotinib structure
CN103848985B (en) * 2012-11-30 2018-03-02 杨子剑 Noval chemical compound and preparation method and purposes containing Sutent analog structure
FR3000492B1 (en) 2012-12-28 2015-09-11 Oribase Pharma NOVEL AZAINDOLE DERIVATIVES AS MULTIKINASE INHIBITORS
FR3000494B1 (en) * 2012-12-28 2015-08-21 Oribase Pharma NOVEL AZAINDOLE DERIVATIVES AS INHIBITORS OF PROTEIN KINASES
FR3000493A1 (en) * 2012-12-28 2014-07-04 Oribase Pharma NEW INHIBITORS OF PROTEIN KINASES
AR094411A1 (en) 2013-01-10 2015-07-29 Gruenenthal Gmbh CARBOXAMIDS BASED ON PIRAZOLILO II
MX2015008919A (en) 2013-01-10 2015-10-22 Gruenenthal Gmbh Pyrazolyl-based carboxamides i as crac channel inhibitors.
US9073921B2 (en) 2013-03-01 2015-07-07 Novartis Ag Salt forms of bicyclic heterocyclic derivatives
US9242969B2 (en) 2013-03-14 2016-01-26 Novartis Ag Biaryl amide compounds as kinase inhibitors
KR101663864B1 (en) 2013-04-19 2016-10-07 영남대학교 산학협력단 Pharmaceutical composition for treating or preventing inflammatory bowel disease comprising amidopyridinol derivative or a pharmaceutically acceptable salt
KR102272606B1 (en) 2013-06-28 2021-07-05 베이진 엘티디 Fused tricyclic urea compounds as raf kinase and/or raf kinase dimer inhibitors
JP6380862B2 (en) 2013-06-28 2018-08-29 ベイジーン リミテッド Condensed tricyclic amide compounds as inhibitors of multiple kinases
CA2934010A1 (en) 2013-12-20 2015-06-25 Pfizer Limited N-acylpiperidine ether tropomyosin-related kinase inhibitors
WO2015134536A1 (en) 2014-03-04 2015-09-11 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US20170197939A1 (en) 2014-04-15 2017-07-13 Pfizer Inc. Tropomyosin-Related Kinase Inhibitors Containing Both A 1H-Pyrazole And A Pyrimidine Moiety
WO2015170218A1 (en) 2014-05-07 2015-11-12 Pfizer Inc. Tropomyosin-related kinase inhibitors
EP3169670B1 (en) * 2014-07-16 2021-10-20 Development Center for Biotechnology N-(3-(quinoxalin-6-ylamino)phenyl)alkane-sulfonamide derivatives as b-raf kinase inhibitors for treating cancer
WO2016009296A1 (en) 2014-07-16 2016-01-21 Pfizer Inc. N-acylpiperidine ether tropomyosin-related kinase inhibitors
WO2016020784A1 (en) 2014-08-05 2016-02-11 Pfizer Inc. N-acylpyrrolidine ether tropomyosin-related kinase inhibitors
UY36294A (en) 2014-09-12 2016-04-29 Novartis Ag COMPOUNDS AND COMPOSITIONS AS QUINASA INHIBITORS
AU2015318233B2 (en) 2014-09-15 2020-03-12 Plexxikon Inc. Heterocyclic compounds and uses thereof
WO2016164641A1 (en) 2015-04-08 2016-10-13 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
JP2018515570A (en) 2015-05-22 2018-06-14 プレキシコン インコーポレーテッドPlexxikon Inc. PLX-8394 or PLX-7904 for use in the treatment of BRAF-V600 related diseases
EA035049B1 (en) 2015-07-16 2020-04-22 Аррэй Байофарма Инк. SUBSTITUTED PYRAZOLO[1,5-a]PYRIDINE COMPOUNDS AS RET KINASE INHIBITORS
GB201514021D0 (en) 2015-08-07 2015-09-23 Arner Elias Set Jeno Novel Pyridines and their use in the treatment of cancer
CN108697708A (en) 2015-10-26 2018-10-23 洛克索肿瘤学股份有限公司 Point mutation in TRK inhibitor resistant cancers and method related to this
US20180346418A1 (en) * 2015-11-25 2018-12-06 Centaurus Biopharma Co., Ltd. Kinase activity regulating compound intermediates preparation method
JP6862468B2 (en) 2015-12-07 2021-04-21 プレキシコン インコーポレーテッドPlexxikon Inc. Compounds and methods for kinase regulation and instructions for them
MA44612A (en) 2016-04-04 2019-02-13 Loxo Oncology Inc PEDIATRIC CANCER TREATMENT METHODS
US10045991B2 (en) 2016-04-04 2018-08-14 Loxo Oncology, Inc. Methods of treating pediatric cancers
TN2018000335A1 (en) 2016-04-04 2020-01-16 Loxo Oncology Inc Liquid formulations of (s)-n-(5-((r)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide
CN110049987B (en) 2016-05-18 2022-02-18 阵列生物制药公司 Process for preparing compounds and salts thereof
CN113135920A (en) 2016-06-30 2021-07-20 株式会社大熊制药 Pyrazolopyrimidine derivatives as kinase inhibitors
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
WO2018136663A1 (en) 2017-01-18 2018-07-26 Array Biopharma, Inc. Ret inhibitors
EP3571203B1 (en) 2017-01-18 2023-06-07 Array BioPharma Inc. Substituted pyrazolo[1,5-a]pyrazine compounds as ret kinase inhibitors
CN110382480A (en) 2017-02-07 2019-10-25 欧比力克治疗公司 The pyridine and its purposes in cancer treatment that heterocyclyl sulfonyl replaces
MX2019009351A (en) 2017-02-07 2019-09-19 Oblique Therapeutics Ab Heteroarylsulfonyl-substituted pyridines and their use in the treatment of cancer.
KR20190115011A (en) 2017-02-07 2019-10-10 오블리크 세러퓨틱스 에이비 Sulfinylpyridine and its use in the treatment of cancer
RU2019128063A (en) 2017-02-07 2021-03-09 Облик Терапьютикс Аб HYDROCARBONSULPHONYL SUBSTITUTED PYRIDINS AND THEIR APPLICATION IN THE TREATMENT OF CANCER
JOP20190213A1 (en) 2017-03-16 2019-09-16 Array Biopharma Inc Macrocyclic compounds as ros1 kinase inhibitors
MX2020003126A (en) 2017-09-20 2020-10-01 Abm Therapeutics Corp Cyclic iminopyrimidine derivatives as kinase inhibitors.
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
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
JP6997876B2 (en) 2018-01-18 2022-02-04 アレイ バイオファーマ インコーポレイテッド Substituted pyrazolyl [4,3-C] pyridine compound as a RET kinase inhibitor
TWI802635B (en) 2018-01-18 2023-05-21 美商亞雷生物製藥股份有限公司 Substituted pyrrolo[2,3-d]pyrimidines compounds as ret kinase inhibitors
US11524963B2 (en) 2018-01-18 2022-12-13 Array Biopharma Inc. Substituted pyrazolo[3,4-d]pyrimidines as RET kinase inhibitors
CA3224949A1 (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
CA3111984A1 (en) 2018-09-10 2020-03-19 Array Biopharma Inc. Fused heterocyclic compounds as ret kinase inhibitors
EP3898626A1 (en) 2018-12-19 2021-10-27 Array Biopharma, Inc. Substituted pyrazolo[1,5-a]pyridine compounds as inhibitors of fgfr tyrosine kinases
WO2020131674A1 (en) 2018-12-19 2020-06-25 Array Biopharma Inc. 7-((3,5-dimethoxyphenyl)amino)quinoxaline derivatives as fgfr inhibitors for treating cancer
CN113710668A (en) 2019-04-09 2021-11-26 普莱希科公司 Condensed azines for EP300 or CBP modulation and indications therefor
TWI817018B (en) 2019-06-28 2023-10-01 美商艾瑞生藥股份有限公司 Compounds for the treatment of braf-associated diseases and disorders
CN114555595B (en) * 2019-08-30 2023-12-26 Tsd生命科学有限公司 Imidazopyridine derivative and pharmaceutical composition containing same as active ingredient
KR20220123023A (en) 2019-12-27 2022-09-05 슈뢰딩거, 인크. Cyclic compounds and methods of use thereof
CA3177522A1 (en) 2020-04-23 2021-10-28 Opna Immuno-Oncology Sa Compounds and methods for cd73 modulation and indications therefor
TW202227068A (en) 2020-08-21 2022-07-16 美商普雷辛肯公司 Combinational drug therapies
WO2022055963A1 (en) 2020-09-10 2022-03-17 Schrödinger, Inc. Heterocyclic pericondensed cdc7 kinase inhibitors for the treatment of cancer
JP2023554062A (en) * 2020-12-18 2023-12-26 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Novel aryl-pyrido-pyrimidin-one derivatives
WO2022164789A1 (en) 2021-01-26 2022-08-04 Schrödinger, Inc. Tricyclic compounds useful in the treatment of cancer, autoimmune and inflammatory disoders
TW202300150A (en) 2021-03-18 2023-01-01 美商薛定諤公司 Cyclic compounds and methods of using same

Family Cites Families (340)

* 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
FR1579473A (en) 1966-10-21 1969-08-29
IL46853A0 (en) 1974-03-20 1975-05-22 Bayer Ag Novel alkoxycarbonylphenylureas,their preparation and their use as herbicides
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
GB1573212A (en) 1976-04-15 1980-08-20 Technicon Instr Immunoassay for gentamicin
US4664504A (en) 1983-01-20 1987-05-12 Tokyo Shibaura Denki Kabushiki Kaisha Image forming apparatus
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
AU567140B2 (en) 1984-01-06 1987-11-12 Shionogi & Co., Ltd. Sulphonamido-benzamide derivatives
EP0154734B1 (en) 1984-03-15 1990-08-29 Immunex Corporation Immediate ligand detection assay, a test kit and its formation
IT1196133B (en) 1984-06-06 1988-11-10 Ausonia Farma Srl FURANIC DERIVATIVES WITH ANTI-ULCER ACTIVITY
US4714693A (en) 1986-04-03 1987-12-22 Uop Inc. Method of making a catalyst composition comprising uniform size metal components on carrier
DE3642315A1 (en) * 1986-12-11 1988-06-23 Boehringer Mannheim Gmbh NEW PYRROLOBENZIMIDAZOLES, PROCESS FOR THEIR PRODUCTION AND MEDICINAL PRODUCTS
US5688655A (en) 1988-02-10 1997-11-18 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
US5700637A (en) 1988-05-03 1997-12-23 Isis Innovation Limited Apparatus and method for analyzing polynucleotide sequences and method of generating oligonucleotide arrays
US5658775A (en) 1988-05-17 1997-08-19 Sloan-Kettering Institute For Cancer Research Double copy retroviral vector
JP2528706B2 (en) 1988-05-30 1996-08-28 ゼリア新薬工業株式会社 Pharmaceutical composition of dihydropyridine compound
JP3082204B2 (en) 1988-09-01 2000-08-28 ホワイトヘッド・インスティチュート・フォー・バイオメディカル・リサーチ Recombinant retrovirus with an amphotropic and ecotropic host range
US5703055A (en) 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
US5527681A (en) 1989-06-07 1996-06-18 Affymax Technologies N.V. Immobilized molecular synthesis of systematically substituted compounds
US5744101A (en) 1989-06-07 1998-04-28 Affymax Technologies N.V. Photolabile nucleoside protecting groups
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
US5800992A (en) 1989-06-07 1998-09-01 Fodor; Stephen P.A. Method of detecting nucleic acids
WO1991018088A1 (en) 1990-05-23 1991-11-28 The United States Of America, Represented By The Secretary, United States Department Of Commerce Adeno-associated virus (aav)-based eucaryotic vectors
DE4022414A1 (en) 1990-07-13 1992-01-16 Bayer Ag SUBSTITUTED PYRROLO-PYRIDINE
ATE159426T1 (en) 1991-04-16 1997-11-15 Nippon Shinyaku Co Ltd METHOD FOR PRODUCING A SOLID DISPERSION
CA2113990A1 (en) 1991-07-26 1993-02-18 Frederick L. Moolten Cancer therapy utilizing malignant cells
US5632957A (en) 1993-11-01 1997-05-27 Nanogen Molecular biological diagnostic systems including electrodes
GB9127531D0 (en) 1991-12-31 1992-02-19 Fujisawa Pharmaceutical Co Heterocyclic compound
FR2687402B1 (en) 1992-02-14 1995-06-30 Lipha NOVEL AZAINDOLES, METHODS OF PREPARATION AND MEDICAMENTS CONTAINING THEM.
JPH05236997A (en) 1992-02-28 1993-09-17 Hitachi Ltd Chip for catching polynucleotide
JPH06135946A (en) 1992-10-30 1994-05-17 Otsuka Pharmaceut Co Ltd Pyrazine derivative
AU686115B2 (en) 1992-11-02 1998-02-05 Fujisawa Pharmaceutical Co., Ltd. Imidazo (I,2-a) pyridine derivatives as bradykinin antagonists, pharmaceuticals and processes for their preparation
GB9226855D0 (en) 1992-12-23 1993-02-17 Erba Carlo Spa Vinylene-azaindole derivatives and process for their preparation
US5576319A (en) 1993-03-01 1996-11-19 Merck, Sharp & Dohme Ltd. Pyrrolo-pyridine derivatives
JPH09500357A (en) 1993-03-01 1997-01-14 メルク シヤープ エンド ドーム リミテツド Pyrrolo-pyridine derivative
US5700809A (en) 1993-03-01 1997-12-23 Merck Sharp & Dohme, Ltd. Pyrrolo-pyridine derivatives
CZ224195A3 (en) 1993-03-01 1996-03-13 Merck Sharp & Dohme The use of pyrrolopyridine derivatives for preparing pharmaceutical preparations
ES2204921T3 (en) 1993-05-27 2004-05-01 Selectide Corporation SOLID PHASE LIBRARIES CODED, TOPOLOGICALLY SECREGATED.
US5840485A (en) 1993-05-27 1998-11-24 Selectide Corporation Topologically segregated, encoded solid phase libraries
IT1265057B1 (en) 1993-08-05 1996-10-28 Dompe Spa TROPIL 7-AZAINDOLIL-3-CARBOXYAMIDE
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
GB9319297D0 (en) 1993-09-17 1993-11-03 Wellcome Found Indole derivatives
US6045996A (en) 1993-10-26 2000-04-04 Affymetrix, Inc. Hybridization assays on oligonucleotide arrays
US5965452A (en) 1996-07-09 1999-10-12 Nanogen, Inc. Multiplexed active biologic array
US6468742B2 (en) 1993-11-01 2002-10-22 Nanogen, Inc. Methods for determination of single nucleic acid polymorphisms using bioelectronic microchip
US5486525A (en) 1993-12-16 1996-01-23 Abbott Laboratories Platelet activating factor antagonists: imidazopyridine indoles
EP1195372A1 (en) 1994-04-18 2002-04-10 Mitsubishi Pharma Corporation N-heterocyclic substituted benzamide derivatives with antihypertensive activity
GB9408577D0 (en) * 1994-04-29 1994-06-22 Fujisawa Pharmaceutical Co New compound
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
GB9412719D0 (en) 1994-06-24 1994-08-17 Erba Carlo Spa Substituted azaindolylidene compounds and process for their preparation
US5763198A (en) 1994-07-22 1998-06-09 Sugen, Inc. Screening assays for compounds
GB9416162D0 (en) 1994-08-10 1994-09-28 Merck Sharp & Dohme Therapeutic agents
GB9416189D0 (en) 1994-08-10 1994-09-28 Merck Sharp & Dohme Therapeutic agents
AU699196B2 (en) 1994-08-10 1998-11-26 Merck Sharp & Dohme Limited Tetrahydropyridinylmethyl derivatives of pyrrolo 2,3-b pyridine
GB9420521D0 (en) 1994-10-12 1994-11-30 Smithkline Beecham Plc Novel 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
US5837815A (en) 1994-12-15 1998-11-17 Sugen, Inc. PYK2 related polypeptide products
NL9500022A (en) * 1995-01-05 1996-08-01 Ihc Syst Bv Device for the magnetic-inductive measurement of the amount of mixture flowing through a pipeline.
GB2298199A (en) 1995-02-21 1996-08-28 Merck Sharp & Dohme Synthesis of azaindoles
GB9503400D0 (en) 1995-02-21 1995-04-12 Merck Sharp & Dohme Therpeutic agents
US5959098A (en) 1996-04-17 1999-09-28 Affymetrix, Inc. Substrate preparation process
US6117681A (en) 1995-03-29 2000-09-12 Bavarian Nordic Research Inst. A/S Pseudotyped retroviral particles
EP2163546B1 (en) 1995-03-30 2016-06-01 Pfizer Products Inc. Quinazoline derivatives
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
GB9507291D0 (en) 1995-04-07 1995-05-31 Merck Sharp & Dohme Therapeutic agents
GB9511220D0 (en) 1995-06-02 1995-07-26 Glaxo Group Ltd Solid dispersions
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
WO1997003967A1 (en) 1995-07-22 1997-02-06 Rhone-Poulenc Rorer Limited Substituted aromatic compounds and their pharmaceutical use
US5866411A (en) 1995-09-08 1999-02-02 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
US5804585A (en) 1996-04-15 1998-09-08 Texas Biotechnology Corporation Thieno-pyridine sulfonamides derivatives thereof and related compounds that modulate the activity of endothelin
US5725838A (en) 1996-05-31 1998-03-10 Resolution Pharmaceuticals, Inc. Radiolabeled D4 receptor ligands
AU3568897A (en) 1996-06-07 1998-01-05 Eos Biotechnology, Inc. Immobilised linear oligonucleotide arrays
WO1997049703A2 (en) 1996-06-25 1997-12-31 Takeda Chemical Industries, Ltd. Oxazolone derivatives and their use as anti-helicobacter pylori agents
DK0956865T4 (en) 1996-08-12 2010-11-22 Mitsubishi Tanabe Pharma Corp Medications comprising Rho kinase inhibitor
JPH10130269A (en) 1996-09-04 1998-05-19 Nippon Chemiphar Co Ltd Carboline derivative
JPH1087629A (en) 1996-09-18 1998-04-07 Fujisawa Pharmaceut Co Ltd New isoquinoline derivative, and its medicinal use
HUP9903330A2 (en) 1996-11-19 2000-03-28 Amgen Inc. Aryl and heteroaryl substituted fused pyrrole derivatives, as antiinflammatory agents containing the same
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
US7863444B2 (en) 1997-03-19 2011-01-04 Abbott Laboratories 4-aminopyrrolopyrimidines as kinase inhibitors
US5977131A (en) 1997-04-09 1999-11-02 Pfizer Inc. Azaindole-ethylamine derivatives as nicotinic acetylcholine receptor binding agents
WO1998047899A1 (en) 1997-04-24 1998-10-29 Ortho-Mcneil Corporation, Inc. Substituted pyrrolopyridines useful in the treatment of inflammatory diseases
US6096718A (en) 1997-06-05 2000-08-01 Gene Targeting Corp. Tissue specific adenovirus vectors for breast cancer treatment
SG72827A1 (en) 1997-06-23 2000-05-23 Hoffmann La Roche Phenyl-and aminophenyl-alkylsulfonamide and urea derivatives
AU8096798A (en) 1997-06-27 1999-01-19 Resolution Pharmaceuticals Inc. Dopamine d4 receptor ligands
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
US6826296B2 (en) 1997-07-25 2004-11-30 Affymetrix, Inc. Method and system for providing a probe array chip design database
EP0901786B1 (en) 1997-08-11 2007-06-13 Pfizer Products Inc. Solid pharmaceutical dispersions with enhanced bioavailability
US6161776A (en) 1997-08-12 2000-12-19 Nibco Inc. Multi-layered, porous mat turf irrigation apparatus and method
CA2300940A1 (en) 1997-08-15 1999-02-25 Hyseq, Inc. Methods and compositions for detection or quantification of nucleic acid species
CA2301539C (en) 1997-09-11 2003-06-17 Genovations, Inc. 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
US6465178B2 (en) 1997-09-30 2002-10-15 Surmodics, Inc. Target molecule attachment to surfaces
US6268387B1 (en) 1997-12-23 2001-07-31 Warner-Lambert Company Thiourea and benzamide compounds, compositions and methods of treating or preventing inflammatory diseases and atherosclerosis
CA2326140A1 (en) 1998-04-02 1999-10-14 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
JP2002510684A (en) 1998-04-02 2002-04-09 メルク エンド カムパニー インコーポレーテッド Gonadotropin-releasing hormone antagonist
WO1999051231A1 (en) 1998-04-02 1999-10-14 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
CA2326143A1 (en) 1998-04-02 1999-10-14 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
AU3210799A (en) 1998-04-02 1999-10-25 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
WO1999051234A1 (en) 1998-04-02 1999-10-14 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6537749B2 (en) 1998-04-03 2003-03-25 Phylos, Inc. Addressable protein 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
WO1999063929A2 (en) 1998-06-08 1999-12-16 Advanced Medicine, Inc. Multibinding inhibitors of microsomal triglyceride transferase protein
US6113913A (en) 1998-06-26 2000-09-05 Genvec, Inc. Recombinant adenovirus
EP1459743B9 (en) 1998-08-17 2012-08-01 Senju Pharmaceutical Co., Ltd. Agent for prophylaxis and treatment of glaucoma
IL141724A0 (en) 1998-08-28 2002-03-10 Scios Inc INHIBITORS OF P38-α KINASE
DE69912618T2 (en) 1998-08-28 2004-10-28 Astrazeneca Ab NEW THIENO [2,3-d] PYRIMIDINDIONE, METHOD FOR THE PRODUCTION THEREOF AND THEIR USE IN THERAPY
CN1326457A (en) 1998-09-18 2001-12-12 巴斯福股份公司 4-aminopyrrolopyrimidines as kinase inhibitors
KR20010085824A (en) 1998-09-18 2001-09-07 스타르크, 카르크 Pyrrolopyrimidines as protein kinase inhibitors
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
IT1303759B1 (en) 1998-11-17 2001-02-23 Dompe Spa IMPROVED PROCEDURE FOR THE PREPARATION OF 7-AZAINDOLYL-3-CARBOXYLIC ACID.
US6277489B1 (en) 1998-12-04 2001-08-21 The Regents Of The University Of California Support for high performance affinity chromatography and other uses
US20010001449A1 (en) 1998-12-30 2001-05-24 Thomas R. Kiliany Low-pressure hydrocracking process
IL144144A0 (en) 1999-01-13 2002-05-23 Bayer Ag Omega-carboxy aryl substituted diphenyl ureas as p38 kinase inhibitors
CA2366260A1 (en) 1999-03-05 2000-09-14 Masahiro Imoto Heterocyclic compounds having effect of activating .alpha.4.beta.2 nicotinic acetylcholine receptors
PT1163237E (en) 1999-03-17 2004-08-31 Astrazeneca Ab AMIDA DERIVATIVES
AR028475A1 (en) 1999-04-22 2003-05-14 Wyeth Corp DERIVATIVES OF AZAINDOL AND USE OF THE SAME FOR THE MANUFACTURE OF A MEDICINAL PRODUCT FOR THE TREATMENT OF DEPRESSION.
US6653309B1 (en) * 1999-04-26 2003-11-25 Vertex Pharmaceuticals Incorporated Inhibitors of IMPDH enzyme technical field of the invention
US6221653B1 (en) 1999-04-27 2001-04-24 Agilent Technologies, Inc. Method of performing array-based hybridization assays using thermal inkjet deposition of sample fluids
FR2793793B1 (en) 1999-05-19 2004-02-27 Adir NOVEL SUBSTITUTED DIMERIC DERIVATIVES, PROCESS FOR PREPARING THEM AND PHARMACEUTICAL COMPOSITIONS CONTAINING SAME
US6492406B1 (en) 1999-05-21 2002-12-10 Astrazeneca Ab Pharmaceutically active compounds
TWI234557B (en) 1999-05-26 2005-06-21 Telik Inc Novel naphthalene ureas as glucose uptake enhancers
JP2003501429A (en) 1999-06-03 2003-01-14 クノール・ゲー・エム・ベー・ハー Benzothiazinone and benzoxazinone compounds
US6653151B2 (en) 1999-07-30 2003-11-25 Large Scale Proteomics Corporation Dry deposition of materials for microarrays using matrix displacement
TR200200928T2 (en) 1999-07-30 2002-09-23 Abbott Gmbh & Co.Kg 2-pyrazoline-5-on compositions
SK3812002A3 (en) 1999-09-17 2003-09-11 Abbott Gmbh & Co Kg Pyrazolopyrimidines as therapeutic agents
WO2001024236A1 (en) 1999-09-27 2001-04-05 Infineon Technologies North America Corp. Semiconductor structures having a capacitor and manufacturing methods
GB9924962D0 (en) 1999-10-21 1999-12-22 Mrc Collaborative Centre Allosteric sites on muscarinic receptors
EP1233769B1 (en) 1999-11-22 2007-12-26 SmithKline Beecham plc Imidazole derivatives
US20010008765A1 (en) 1999-12-06 2001-07-19 Fuji Photo Film Co., Ltd. DNA chip and reactive solid carrier
CO5271715A1 (en) 1999-12-21 2003-04-30 Sugen Inc 7-AZA-INDOLIN-2-WAVES SUBSTITUTED IN 4 AND ITS USE AS PROTEIUNA QUINASA INHIBITORS
AU784266B2 (en) 1999-12-22 2006-03-02 Sugen, Inc. Indolinone derivatives for modulation of c-kit tyrosine kinase
FR2805259B1 (en) 2000-02-17 2002-03-29 Inst Nat Sante Rech Med NOVEL N-MERCAPTOACYL AMINOACID DERIVATIVES, PROCESS FOR THEIR PREPARATION AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
US20020061892A1 (en) 2000-02-22 2002-05-23 Tao Wang Antiviral azaindole derivatives
EP1263753B1 (en) 2000-03-06 2004-05-06 SmithKline Beecham plc Imidazol derivatives as raf kinase inhibitors
GB0005357D0 (en) 2000-03-06 2000-04-26 Smithkline Beecham Plc Compounds
JP2001278886A (en) 2000-03-28 2001-10-10 Dai Ichi Seiyaku Co Ltd Benzoxazine derivative and medicament containing the same
MXPA02009543A (en) 2000-03-29 2005-08-26 Abbott Gmbh & Co Kg Method of identifying inhibitors of tie 2.
GB0007934D0 (en) 2000-03-31 2000-05-17 Darwin Discovery Ltd Chemical compounds
US6335342B1 (en) 2000-06-19 2002-01-01 Pharmacia & Upjohn S.P.A. Azaindole derivatives, process for their preparation, and their use as antitumor agents
ES2247138T3 (en) 2000-06-26 2006-03-01 Lilly Icos Llc DERIVATIVES OF PIRAZINDIONA CONDENSED AS PDE5 INHIBITORS.
CN1518543A (en) 2000-08-31 2004-08-04 �Ʒ� Pyrazole derivatives and their use as protein kinase inhibitors
WO2002024680A1 (en) 2000-09-21 2002-03-28 Smithkline Beecham P.L.C. Imidazole derivatives as raf kinase inhibitors
US6878714B2 (en) 2001-01-12 2005-04-12 Amgen Inc. Substituted alkylamine derivatives and methods of use
IL157389A0 (en) 2001-02-27 2004-05-12 Astrazeneca Ab A pharmaceutical formulation comprising bicalutamide and an enteric polymer and use thereof as a medicament
AU2002303145A1 (en) 2001-03-23 2002-10-08 Smithkline Beecham Corporation Compounds useful as kinase inhibitors for the treatment of hyperproliferative diseases
JP4204326B2 (en) 2001-04-11 2009-01-07 千寿製薬株式会社 Visual function disorder improving agent
AU2002258971A1 (en) 2001-04-24 2002-11-05 Wyeth Antidepressant azaheterocyclylmethyl derivatives of 2,3-dihydro-1,4-benzodioxan
GB0112348D0 (en) 2001-05-19 2001-07-11 Smithkline Beecham Plc Compounds
GB0114417D0 (en) 2001-06-13 2001-08-08 Boc Group Plc Lubricating systems for regenerative vacuum pumps
WO2002102783A1 (en) 2001-06-19 2002-12-27 Merck & Co., Inc. Tyrosine kinase inhibitors
US7291639B2 (en) 2001-06-20 2007-11-06 Wyeth Aryloxy-acetic acid compounds useful as inhibitors of plasminogen activator inhibitor-1 (PAI-1)
GB0115109D0 (en) 2001-06-21 2001-08-15 Aventis Pharma Ltd Chemical compounds
SE0102300D0 (en) 2001-06-26 2001-06-26 Astrazeneca Ab Compounds
US20030091974A1 (en) 2001-06-29 2003-05-15 Alain Moussy Method for screening compounds capable of depleting mast cells
SE0102439D0 (en) 2001-07-05 2001-07-05 Astrazeneca Ab New compounds
UA75425C2 (en) 2001-07-09 2006-04-17 Piperazine oxime derivatives with antagonistic activity to nk-1 receptor, use thereof, a pharmaceutical composition based thereon, a method for producing and a method for producing intermediary compounds
GB0117583D0 (en) 2001-07-19 2001-09-12 Astrazeneca Ab Novel compounds
US6858860B2 (en) 2001-07-24 2005-02-22 Seiko Epson Corporation Apparatus and method for measuring natural period of liquid
GB0118479D0 (en) 2001-07-28 2001-09-19 Astrazeneca Ab Novel compounds
JP2003073357A (en) 2001-09-03 2003-03-12 Mitsubishi Pharma Corp Rho KINASE INHIBITOR COMPRISING AMIDE COMPOUND
GB0121490D0 (en) 2001-09-05 2001-10-24 Smithkline Beecham Plc Ciompounds
US20040192689A1 (en) 2001-09-05 2004-09-30 Dean David Kenneth Heterocycle-carboxamide derivatives as raf kinase inhibitors
WO2003022832A1 (en) 2001-09-05 2003-03-20 Smithkline Beecham P.L.C. Pyridylfurans and pyrroles as raf kinase inhibitors
ES2309207T3 (en) 2001-09-05 2008-12-16 Smithkline Beecham Plc FURANIC DERIVATIVES REPLACED WITH PIRIDINE AS INHIBITORS OF QUINASA RAF.
GB0121494D0 (en) 2001-09-05 2001-10-24 Smithkline Beecham Plc Compounds
GB0121488D0 (en) 2001-09-05 2001-10-24 Smithkline Beecham Plc Compounds
AU2002334355A1 (en) 2001-09-06 2003-03-18 Prochon Biotech Ltd. Protein tyrosine kinase inhibitors
WO2003028724A1 (en) 2001-10-04 2003-04-10 Smithkline Beecham Corporation Chk1 kinase inhibitors
JPWO2003037862A1 (en) 2001-10-30 2005-02-17 日本新薬株式会社 Amide derivatives and pharmaceuticals
US20030119839A1 (en) 2001-12-13 2003-06-26 Nan-Horng Lin Protein kinase inhibitors
PL371486A1 (en) 2002-02-01 2005-06-13 Astrazeneca Ab Quinazoline compounds
US20030236277A1 (en) 2002-02-14 2003-12-25 Kadow John F. Indole, azaindole and related heterocyclic pyrrolidine derivatives
US20040171062A1 (en) 2002-02-28 2004-09-02 Plexxikon, Inc. Methods for the design of molecular scaffolds and ligands
US6884889B2 (en) 2002-03-25 2005-04-26 Bristol-Myers Squibb Co. Processes for the preparation of antiviral 7-azaindole derivatives
ATE457312T1 (en) 2002-03-28 2010-02-15 Eisai R&D Man Co Ltd AZAINDOLES AS INHIBITORS OF C-JUN N-TERMINAL KINASES
WO2003082868A1 (en) 2002-03-28 2003-10-09 Eisai Co., Ltd. 7-azaindoles as inhibitors of c-jun n-terminal kinases for the treatment of neurodegenerative disorders
EP1495016A2 (en) 2002-04-09 2005-01-12 Astex Technology Limited Heterocyclic compounds and their use as modulators of p38 map kinase
UA78999C2 (en) 2002-06-04 2007-05-10 Wyeth Corp 1-(aminoalkyl)-3-sulfonylazaindoles as ligands of 5-hydroxytryptamine-6
WO2004005283A1 (en) 2002-07-09 2004-01-15 Vertex Pharmaceuticals Incorporated Imidazoles, oxazoles and thiazoles with protein kinase inhibiting activities
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
EP1388341A1 (en) 2002-08-07 2004-02-11 Aventis Pharma Deutschland GmbH Acylamino-substituted heteroaromatic compounds and their use as pharmaceuticals
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
CN104383554B (en) * 2002-09-06 2018-06-08 天蓝制药公司 For transmitting the polymer based on cyclodextrin of therapeutic agent
EP1558751A4 (en) 2002-09-16 2007-08-22 Plexxikon Inc Crystal structure of pim-1 kinase
US7183241B2 (en) 2002-10-15 2007-02-27 Exxonmobil Research And Engineering Company Long life lubricating oil composition with very low phosphorus content
SE0203654D0 (en) 2002-12-09 2002-12-09 Astrazeneca Ab New compounds
DE60323133D1 (en) 2002-12-13 2008-10-02 Smithkline Beecham Corp CYCLOHEXYL COMPOUNDS AS CCR5 ANTAGONISTS
UA80171C2 (en) 2002-12-19 2007-08-27 Pfizer Prod Inc Pyrrolopyrimidine derivatives
US7696225B2 (en) 2003-01-06 2010-04-13 Osi Pharmaceuticals, Inc. (2-carboxamido)(3-Amino) thiophene 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
US20050085463A1 (en) 2003-01-23 2005-04-21 Weiner David M. Use of N-desmethylclozapine to treat human neuropsychiatric disease
CN100594023C (en) 2003-02-03 2010-03-17 诺瓦提斯公司 Pharmaceutical formulation
US20050048573A1 (en) 2003-02-03 2005-03-03 Plexxikon, Inc. PDE5A crystal structure and uses
BRPI0407456A (en) * 2003-02-12 2006-01-31 Pfizer 2- (Sulfo-phenyl) -aminothiazole antiproliferative derivatives and pharmaceutical compositions, and methods for their use
WO2004074286A1 (en) 2003-02-14 2004-09-02 Wyeth Heterocyclyl-3-sulfonylazaindole or -azaindazole derivatives as 5-hydroxytryptamine-6 ligands
DK2476667T3 (en) * 2003-02-26 2014-09-15 Sugen Inc Amino heteroaryl compounds as protein kinase inhibitors
EP1627045A2 (en) 2003-02-28 2006-02-22 Plexxikon, Inc. Pyk2 crystal structure and uses
WO2004078756A2 (en) 2003-03-06 2004-09-16 Eisai Co., Ltd. Jnk inhibitors
US7612086B2 (en) 2003-05-16 2009-11-03 Eisai R & D Management Co. Ltd. JNK inhibitors
WO2005002673A1 (en) * 2003-07-03 2005-01-13 Astex Therapeutics Limited Raf kinase inhibitors
US20050079548A1 (en) 2003-07-07 2005-04-14 Plexxikon, Inc. Ligand development using PDE4B crystal structures
WO2005009958A1 (en) 2003-07-17 2005-02-03 Plexxikon, Inc. Ppar active compounds
US7348338B2 (en) 2003-07-17 2008-03-25 Plexxikon, Inc. PPAR active compounds
CL2004001884A1 (en) 2003-08-04 2005-06-03 Pfizer Prod Inc DRYING PROCEDURE FOR SPRAYING FOR THE FORMATION OF SOLID DISPERSIONS AMORPHES OF A PHARMACO AND POLYMERS.
US7504401B2 (en) 2003-08-29 2009-03-17 Locus Pharmaceuticals, Inc. Anti-cancer agents and uses thereof
AR045595A1 (en) 2003-09-04 2005-11-02 Vertex Pharma USEFUL COMPOSITIONS AS INHIBITORS OF KINASE PROTEINS
WO2005044181A2 (en) 2003-09-09 2005-05-19 Temple University-Of The Commonwealth System Of Higher Education Protection of tissues and cells from cytotoxic effects of ionizing radiation by abl inhibitors
EP1678147B1 (en) 2003-09-15 2012-08-08 Lead Discovery Center GmbH Pharmaceutically active 4,6-disubstituted aminopyrimidine derivatives as modulators of protein kinases
WO2005028624A2 (en) 2003-09-15 2005-03-31 Plexxikon, Inc. Molecular scaffolds for kinase ligand development
WO2005030128A2 (en) 2003-09-23 2005-04-07 Merck & Co., Inc. Pyrazole modulators of metabotropic glutamate receptors
US7449582B2 (en) * 2003-10-08 2008-11-11 Irm Llc Compounds and compositions as protein kinase inhibitors
DE10357510A1 (en) 2003-12-09 2005-07-07 Bayer Healthcare Ag Heteroaryl-substituted benzenes
PL1696920T3 (en) 2003-12-19 2015-03-31 Plexxikon Inc Compounds and methods for development of ret modulators
US20070066641A1 (en) 2003-12-19 2007-03-22 Prabha Ibrahim Compounds and methods for development of RET modulators
US7517970B2 (en) 2003-12-19 2009-04-14 Plexxikon, Inc. Nucleic acids encoding kinase and phosphatase enzymes, expression vectors and cells containing same
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
GB0330043D0 (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 comprising them
EP2119771B1 (en) 2003-12-24 2018-08-22 DuPont Nutrition Biosciences ApS Proteins
US20050222171A1 (en) 2004-01-22 2005-10-06 Guido Bold Organic compounds
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
EP1722789A2 (en) 2004-03-08 2006-11-22 Amgen Inc. Therapeutic modulation of ppar (gamma) activity
KR20050091462A (en) 2004-03-12 2005-09-15 한국과학기술연구원 Furopyrimidine compound and ddr2 tyrosine kinase activity inhibitor comprising the same
JP2007223901A (en) * 2004-03-24 2007-09-06 Takeda Chem Ind Ltd Heterocyclic compound and use thereof
MXPA06011327A (en) 2004-03-30 2006-12-15 Vertex Pharma Azaindoles useful as inhibitors of jak and other protein kinases.
CN101014597A (en) 2004-04-02 2007-08-08 沃泰克斯药物股份有限公司 Azaindoles useful as inhibitors of rock and other protein kinases
WO2006078287A2 (en) 2004-05-06 2006-07-27 Plexxikon, Inc. Pde4b inhibitors and uses therefor
WO2005115363A2 (en) 2004-05-25 2005-12-08 Yale University Method for treating skeletal disorders resulting from fgfr malfunction
US20090105218A1 (en) 2004-05-29 2009-04-23 7Tm Pharma A/S CRTH2 Receptor Ligands For Therapeutic Use
WO2006009755A2 (en) 2004-06-17 2006-01-26 Plexxikon, Inc. Azaindoles modulating c-kit activity and uses therefor
US7498342B2 (en) * 2004-06-17 2009-03-03 Plexxikon, Inc. Compounds modulating c-kit activity
CA2572058A1 (en) 2004-06-30 2006-01-12 Vertex Pharmaceuticals Incorporated Azaindoles useful as inhibitors of protein kinases
US7140816B2 (en) 2004-07-20 2006-11-28 H&S Tool, Inc. Multi-functional tube milling head
US7626021B2 (en) 2004-07-27 2009-12-01 Sgx Pharmaceuticals, Inc. Fused ring heterocycle kinase modulators
US7709645B2 (en) 2004-07-27 2010-05-04 Sgx Pharmaceuticals, Inc. Pyrrolo-pyridine kinase modulators
MX2007001127A (en) 2004-07-27 2007-07-11 Sgx Pharmaceuticals Inc Pyrrolo-pyridine kinase modulators.
US7361764B2 (en) 2004-07-27 2008-04-22 Sgx Pharmaceuticals, Inc. Pyrrolo-pyridine kinase modulators
MX2007001126A (en) 2004-07-27 2007-09-25 Sgx Pharmaceuticals Inc Fused ring heterocycle kinase modulators.
US20060024361A1 (en) 2004-07-28 2006-02-02 Isa Odidi Disintegrant assisted controlled release technology
EP1789393A2 (en) 2004-07-30 2007-05-30 GPC Biotech AG Pyridinylamines
CA2583428A1 (en) 2004-09-03 2006-03-09 Plexxikon, Inc. Bicyclic heteroaryl pde4b inhibitors
BRPI0517619A (en) * 2004-11-10 2008-10-14 Cgi Pharmaceuticals Inc imidazo [1,2-a] pyrazin-8-ylamine chemical entities, their pharmaceutical compositions, use of said compounds in the preparation of a medicament, process of preparing a medicament and methods of using said compounds
US20060135540A1 (en) 2004-11-30 2006-06-22 Jack Lin PPAR active compounds
JP2008521829A (en) 2004-11-30 2008-06-26 プレキシコン,インコーポレーテッド PPAR active compounds
JP4954086B2 (en) 2004-12-08 2012-06-13 グラクソスミスクライン・リミテッド・ライアビリティ・カンパニー 1H-pyrrolo [2,3-B] pyridine
US20060160135A1 (en) 2004-12-08 2006-07-20 Weiru Wang SF-1 and LRH-1 modulator development
TW200716551A (en) * 2005-03-10 2007-05-01 Cgi Pharmaceuticals Inc Certain substituted amides, method of making, and method of use thereof
CA2605738C (en) 2005-04-25 2013-10-01 Merck Patent Gesellschaft Mit Beschraenkter Haftung Novel azaheterocyclic compounds as kinase inhibitors
FR2884821B1 (en) 2005-04-26 2007-07-06 Aventis Pharma Sa SUBSTITUTED PYRROLOPYRIDINES, COMPOSITIONS CONTAINING SAME, METHOD OF MANUFACTURE AND USE
WO2006124874A2 (en) 2005-05-12 2006-11-23 Kalypsys, Inc. Inhibitors of b-raf kinase
NZ563444A (en) 2005-05-17 2011-04-29 Plexxikon Inc Pyrrolo(2,3-b)pyridine derivatives as protein kinase inhibitors
RU2435769C2 (en) 2005-05-20 2011-12-10 Вертекс Фармасьютикалз Инкорпорейтед Pyrrolopyridines effective as proteinkinase inhibitors
NZ565245A (en) * 2005-06-21 2010-07-30 Mitsui Chemicals Inc Amide derivative and pesticide containing such compound
DK1893612T3 (en) 2005-06-22 2011-11-21 Plexxikon Inc Pyrrole [2,3-B] pyridine derivatives as protein kinase inhibitors
DE102005029382B3 (en) 2005-06-24 2006-12-21 Sanofi-Aventis Deutschland Gmbh New alkoxymethylyl substituted benzoic acid derivatives are peroxisome proliferation activated receptor agonists used for treating and/or preventing e.g. diabetes mellitus, dyslipidemia and disturbances in fatty acid metabolism
GB0516156D0 (en) 2005-08-05 2005-09-14 Eisai London Res Lab Ltd JNK inhibitors
KR20080033526A (en) * 2005-08-09 2008-04-16 아이알엠 엘엘씨 Compounds and compositions as protein kinase inhibitors
US7754717B2 (en) * 2005-08-15 2010-07-13 Amgen Inc. Bis-aryl amide compounds and methods of use
AU2006287521A1 (en) 2005-09-07 2007-03-15 Plexxikon, Inc. PPARactive compounds
EP1943245A2 (en) 2005-09-07 2008-07-16 Plexxikon, Inc. 1,3-disubstituted indole derivatives for use as ppar modulators
CA2621275A1 (en) 2005-09-07 2007-03-15 Plexxikon, Inc. 1 , 4 and 1 , 5-disubstituted indole derivatives for use as ppar active compounds
WO2007109604A2 (en) 2006-03-20 2007-09-27 Vertex Pharmaceuticals Incorporated Pharmaceutical compositions
US7963673B2 (en) 2006-05-30 2011-06-21 Finn Bruce L Versatile illumination system
LT2848610T (en) * 2006-11-15 2017-11-10 Ym Biosciences Australia Pty Ltd Inhibitors of kinase activity
WO2008063888A2 (en) 2006-11-22 2008-05-29 Plexxikon, Inc. Compounds modulating c-fms and/or c-kit activity and uses therefor
WO2008079909A1 (en) 2006-12-21 2008-07-03 Plexxikon, Inc. Pyrrolo [2,3-b] pyridines as kinase modulators
EP2094701A2 (en) 2006-12-21 2009-09-02 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
PE20121126A1 (en) 2006-12-21 2012-08-24 Plexxikon Inc PIRROLO [2,3-B] PYRIDINES COMPOUNDS AS KINASE MODULATORS
BRPI0808196A2 (en) 2007-03-08 2014-07-08 Plexxikon Inc PPAR ACTIVE COMPOUNDS
PE20090159A1 (en) 2007-03-08 2009-02-21 Plexxikon Inc INDOL-PROPIONIC ACID DERIVED COMPOUNDS AS PPARs MODULATORS
JP2010526848A (en) 2007-05-11 2010-08-05 エフ.ホフマン−ラ ロシュ アーゲー Pharmaceutical composition for poorly soluble drugs
TW200908968A (en) 2007-05-29 2009-03-01 Sgx Pharmaceuticals Inc Substituted pyrrolopyridines and pyrazolopyridines as kinase modulators
US20100190777A1 (en) 2007-07-17 2010-07-29 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
EA021645B1 (en) 2007-07-23 2015-08-31 Фарматен С.А. Solid dosage form as a tablet and method for the preparation thereof
TW200940540A (en) 2008-02-29 2009-10-01 Array Biopharma Inc RAF inhibitor compounds and methods of use thereof
JP2011513329A (en) 2008-02-29 2011-04-28 アレイ バイオファーマ、インコーポレイテッド Imidazo [4,5-b] pyridine derivatives used as RAF inhibitory compounds
WO2009111280A1 (en) 2008-02-29 2009-09-11 Array Biopharma Inc. N- (6-aminopyridin-3-yl) -3- (sulfonamido) benzamide derivatives as b-raf inhibitors for the treatment of cancer
CN102149712A (en) 2008-02-29 2011-08-10 阵列生物制药公司 Pyrazole [3, 4-b] pyridine Raf inhibitors
UA103319C2 (en) * 2008-05-06 2013-10-10 Глаксосмитклайн Ллк Thiazole- and oxazole-benzene sulfonamide compounds
PE20091846A1 (en) 2008-05-19 2009-12-16 Plexxikon Inc PIRROLO [2,3-d] -PYRIMIDINE DERIVATIVES AS KINE MODULATORS
US8158636B2 (en) 2008-05-19 2012-04-17 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US8110576B2 (en) 2008-06-10 2012-02-07 Plexxikon Inc. Substituted pyrrolo[2,3b]pyrazines and methods for treatment of raf protein kinase-mediated indications
JP5767965B2 (en) 2008-06-10 2015-08-26 プレキシコン インコーポレーテッドPlexxikon Inc. 5H-pyrrolo [2,3-B] pyrazine derivatives that regulate kinases and indications thereof
RU2011141123A (en) 2009-03-11 2013-04-20 Плексксикон, Инк. PYRROLO [2,3-b] PYRIDINE DERIVATIVES AS RAF KINASE INHIBITORS
WO2010104973A1 (en) 2009-03-11 2010-09-16 Plexxikon, Inc. Pyrrolo [2, 3-b] pyridine derivatives for the inhibition of raf kinases
EP2406234A1 (en) 2009-03-12 2012-01-18 GlaxoSmithKline LLC Thiazole sulfonamide and oxazole sulfonamide kinase inhibitors
WO2010111527A1 (en) 2009-03-26 2010-09-30 Plexxikon, Inc. Pyrazolo [ 3, 4 -b] pyridines as kinase inhibitors and their medical use
MX349923B (en) 2009-04-03 2017-08-21 Hoffmann La Roche Propane- i-sulfonic acid {3- [5- (4 -chloro-phenyl) -1h-pyrrolo [2, 3-b] pyridine-3-carbonyl] -2, 4-difluoro-pheny l } -amide compositions and uses thereof.
SG175877A1 (en) 2009-05-04 2011-12-29 Plexxikon Inc Compounds and methods for inhibition of renin, and indications therefor
TW201041888A (en) 2009-05-06 2010-12-01 Plexxikon Inc Compounds and methods for kinase modulation, and indications therefor
JP2013510166A (en) 2009-11-06 2013-03-21 プレキシコン インコーポレーテッド Compounds, methods and applications for kinase regulation
AU2010321883A1 (en) 2009-11-18 2012-05-31 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
WO2011079133A2 (en) 2009-12-23 2011-06-30 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
TWI619713B (en) 2010-04-21 2018-04-01 普雷辛肯公司 Compounds and methods for kinase modulation, and indications therefor
US8642606B2 (en) 2010-09-29 2014-02-04 Plexxikon Inc. ZAP-70 active compounds
PL2672967T3 (en) 2011-02-07 2019-04-30 Plexxikon Inc Compounds and methods for kinase modulation, and indications therefor
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
AU2012255275B2 (en) 2011-05-17 2016-01-28 Plexxikon Inc. Kinase modulation and indications therefor
US9358235B2 (en) 2012-03-19 2016-06-07 Plexxikon Inc. Kinase modulation, and indications therefor
US9150570B2 (en) 2012-05-31 2015-10-06 Plexxikon Inc. Synthesis of heterocyclic compounds
AU2013312477B2 (en) 2012-09-06 2018-05-31 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
WO2014100620A2 (en) 2012-12-21 2014-06-26 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
RU2680100C9 (en) 2013-03-15 2019-04-18 Плексксикон Инк. Heterocyclic compounds and uses thereof
US20140303121A1 (en) 2013-03-15 2014-10-09 Plexxikon Inc. Heterocyclic compounds and uses thereof
JP2016520131A (en) 2013-05-30 2016-07-11 プレキシコン インコーポレーテッドPlexxikon Inc. Compounds for kinase regulation and their adaptation
JP2015073357A (en) 2013-10-02 2015-04-16 日産自動車株式会社 Molding device and molding method of stator assembly
WO2015134536A1 (en) 2014-03-04 2015-09-11 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
AU2015318233B2 (en) 2014-09-15 2020-03-12 Plexxikon Inc. Heterocyclic compounds and uses thereof
WO2016164641A1 (en) 2015-04-08 2016-10-13 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
MY192352A (en) 2015-05-06 2022-08-17 Plexxikon Inc Solid forms of a compound modulating kinases
ES2923943T3 (en) 2015-05-06 2022-10-03 Plexxikon Inc Synthesis of a compound that modulates kinases
CA2986735A1 (en) 2015-05-22 2016-12-01 Plexxikon Inc. Solid forms of a compound for modulating kinases
JP2018515570A (en) 2015-05-22 2018-06-14 プレキシコン インコーポレーテッドPlexxikon Inc. PLX-8394 or PLX-7904 for use in the treatment of BRAF-V600 related diseases
US10829484B2 (en) 2015-07-28 2020-11-10 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
CN108137585B (en) 2015-09-21 2021-10-22 普莱希科公司 Heterocyclic compounds and their use
JP6862468B2 (en) 2015-12-07 2021-04-21 プレキシコン インコーポレーテッドPlexxikon Inc. Compounds and methods for kinase regulation and instructions for them
US10160747B2 (en) 2016-03-16 2018-12-25 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
TW201815766A (en) 2016-09-22 2018-05-01 美商普雷辛肯公司 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
AU2018237047A1 (en) 2017-03-20 2019-10-17 Plexxikon Inc. Crystalline forms of 4-(1-(1,1-di(pyridin-2-yl)ethyl)-6-(3,5-dimethylisoxazol-4-yl)-1H- pyrrolo[3,2-b]pyridin-3-yl)benzoic acid that inhibits bromodomain
US10428067B2 (en) 2017-06-07 2019-10-01 Plexxikon Inc. Compounds and methods for kinase modulation
CN110944670B (en) 2017-07-25 2023-03-10 普莱希科公司 Formulations of compounds for modulating kinases
EP3694855A1 (en) 2017-10-13 2020-08-19 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

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10426760B2 (en) 2007-07-17 2019-10-01 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
US10123998B2 (en) 2012-03-19 2018-11-13 Plexxikon Inc. Kinase modulation, and indications therefor
US10301280B2 (en) 2012-12-21 2019-05-28 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10519177B2 (en) 2013-03-15 2019-12-31 Plexxikon Inc. Heterocyclic compounds and uses thereof
US10501460B2 (en) 2013-03-15 2019-12-10 Plexxikon Inc. Heterocyclic compounds and uses thereof
US10421761B2 (en) 2013-05-30 2019-09-24 Plexxikon Inc. Compounds for kinase modulation, and indications therefor
US10189833B2 (en) 2015-05-06 2019-01-29 Plexxikon Inc. Solid forms of a compound modulating kinases
US10399975B2 (en) 2015-05-06 2019-09-03 Plexxikon Inc. Synthesis of a compound that modulates kinases
US10730876B2 (en) 2015-05-06 2020-08-04 Plexxikon Inc. Synthesis of a compound that modulates kinases
US10584122B2 (en) 2015-05-06 2020-03-10 Plexxikon Inc. Solid forms of a compound modulating kinases
US9994567B2 (en) 2015-05-22 2018-06-12 Plexxikon Inc. Synthesis of heterocyclic compounds
US10316032B2 (en) 2015-05-22 2019-06-11 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
US10647716B2 (en) 2015-09-21 2020-05-12 Plexxikon Inc. Heterocyclic compounds and uses thereof
US10899761B2 (en) 2015-09-21 2021-01-26 Plexxikon Inc. Heterocyclic compounds and uses thereof
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
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
US10961240B2 (en) 2017-07-25 2021-03-30 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
WO2020176579A1 (en) * 2019-02-26 2020-09-03 Aerie Pharmaceuticals, Inc. Thienyl cyclopropyl-amino-isoquinolinyl amide compounds

Also Published As

Publication number Publication date
NZ582772A (en) 2012-06-29
JP6456900B2 (en) 2019-01-23
US20210346358A1 (en) 2021-11-11
US20170056382A1 (en) 2017-03-02
IL203331A (en) 2015-08-31
IL239959A (en) 2017-12-31
EP2170830B1 (en) 2014-10-15
US20140303187A1 (en) 2014-10-09
JP2015007100A (en) 2015-01-15
IL239959A0 (en) 2015-08-31
US20210353602A1 (en) 2021-11-18
RU2010105549A (en) 2011-08-27
JP2019218387A (en) 2019-12-26
US20190209536A1 (en) 2019-07-11
CA2695004A1 (en) 2009-01-22
JP2010533729A (en) 2010-10-28
JP2018104456A (en) 2018-07-05
WO2009012283A1 (en) 2009-01-22
JP2017061517A (en) 2017-03-30
AU2008276063A1 (en) 2009-01-22
SG183036A1 (en) 2012-08-30
CA2695004C (en) 2016-01-19
EP2170830A1 (en) 2010-04-07
US20190175567A1 (en) 2019-06-13
US20140038948A1 (en) 2014-02-06
US20160168146A1 (en) 2016-06-16
US20100190777A1 (en) 2010-07-29
US9844539B2 (en) 2017-12-19
US10426760B2 (en) 2019-10-01
CA2695004E (en) 2009-01-22
US9469640B2 (en) 2016-10-18
BRPI0814423A2 (en) 2021-06-15
MX2010000617A (en) 2010-05-17
CN101808994B (en) 2013-05-15
BRPI0814423B1 (en) 2022-04-19
AU2008276063B2 (en) 2013-11-28
US20130237531A1 (en) 2013-09-12
CN101808994A (en) 2010-08-18

Similar Documents

Publication Publication Date Title
US10426760B2 (en) Compounds and methods for kinase modulation, and indications therefor
US8153641B2 (en) Compounds and methods for kinase modulation, and indications therefor
US8110576B2 (en) Substituted pyrrolo[2,3b]pyrazines and methods for treatment of raf protein kinase-mediated indications
US8198273B2 (en) Compounds and methods for kinase modulation, and indications therefor
US9440969B2 (en) Compounds and methods for kinase modulation, and indications therefor
US8901301B2 (en) Pyrrolo[2,3-]pyridine kinase inhibitors
US8158636B2 (en) Compounds and methods for kinase modulation, and indications therefor
US20110092538A1 (en) Compounds and methods for kinase modulation, and indications therefor
WO2010111527A1 (en) Pyrazolo [ 3, 4 -b] pyridines as kinase inhibitors and their medical use
RU2487121C2 (en) Compounds and methods for kinase modulation and indications for use of said compounds and methods

Legal Events

Date Code Title Description
AS Assignment

Owner name: PLEXXIKON INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:WU, GUOXIAN;ZHANG, JIAZHONG;ZHU, YONG-LIANG;AND OTHERS;SIGNING DATES FROM 20080724 TO 20080805;REEL/FRAME:046270/0188

STCB Information on status: application discontinuation

Free format text: EXPRESSLY ABANDONED -- DURING PUBLICATION PROCESS