WO2007111904A2 - C-met protein kinase inhibitors for the treatment of proliferative disorders - Google Patents

C-met protein kinase inhibitors for the treatment of proliferative disorders Download PDF

Info

Publication number
WO2007111904A2
WO2007111904A2 PCT/US2007/007016 US2007007016W WO2007111904A2 WO 2007111904 A2 WO2007111904 A2 WO 2007111904A2 US 2007007016 W US2007007016 W US 2007007016W WO 2007111904 A2 WO2007111904 A2 WO 2007111904A2
Authority
WO
WIPO (PCT)
Prior art keywords
aliphatic
nhr
pharmaceutically acceptable
compound according
acceptable salt
Prior art date
Application number
PCT/US2007/007016
Other languages
French (fr)
Other versions
WO2007111904A3 (en
Inventor
David J. Lauffer
Robert J. Davies
Dean Stamos
Alexander Aronov
David D. Deininger
Ronald Grey, Jr.
Jinwang Xu
Pan Li
Brian Ledford
Luc Farmer
Randy Scott Bethiel
Dylan Jacobs
Kira Mcginty
Original Assignee
Vertex Pharmaceuticals Incorporated
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
Priority to DE602007004092T priority Critical patent/DE602007004092D1/en
Priority to AT07753626T priority patent/ATE453635T1/en
Priority to NZ571933A priority patent/NZ571933A/en
Priority to CN2007800182084A priority patent/CN101479255B/en
Application filed by Vertex Pharmaceuticals Incorporated filed Critical Vertex Pharmaceuticals Incorporated
Priority to MX2008012096A priority patent/MX2008012096A/en
Priority to CA002646701A priority patent/CA2646701A1/en
Priority to EP07753626A priority patent/EP2004625B1/en
Priority to AU2007231577A priority patent/AU2007231577B2/en
Priority to JP2009501540A priority patent/JP2009533327A/en
Publication of WO2007111904A2 publication Critical patent/WO2007111904A2/en
Publication of WO2007111904A3 publication Critical patent/WO2007111904A3/en
Priority to IL194242A priority patent/IL194242A0/en
Priority to NO20084435A priority patent/NO20084435L/en
Priority to HK09107689.2A priority patent/HK1129890A1/en
Priority to US12/942,395 priority patent/US8232406B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic 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 three or more hetero 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
    • A61K31/435Heterocyclic 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
    • 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
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • 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
    • 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
    • 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
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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
    • C07D401/14Heterocyclic 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 three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D407/00Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00
    • C07D407/14Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing three or more hetero rings
    • 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
    • C07D409/04Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • 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
    • C07D451/00Heterocyclic compounds containing 8-azabicyclo [3.2.1] octane, 9-azabicyclo [3.3.1] nonane, or 3-oxa-9-azatricyclo [3.3.1.0<2,4>] nonane ring systems, e.g. tropane or granatane alkaloids, scopolamine; Cyclic acetals thereof
    • C07D451/02Heterocyclic compounds containing 8-azabicyclo [3.2.1] octane, 9-azabicyclo [3.3.1] nonane, or 3-oxa-9-azatricyclo [3.3.1.0<2,4>] nonane ring systems, e.g. tropane or granatane alkaloids, scopolamine; Cyclic acetals thereof containing not further condensed 8-azabicyclo [3.2.1] octane or 3-oxa-9-azatricyclo [3.3.1.0<2,4>] nonane ring systems, e.g. tropane; Cyclic acetals thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D451/00Heterocyclic compounds containing 8-azabicyclo [3.2.1] octane, 9-azabicyclo [3.3.1] nonane, or 3-oxa-9-azatricyclo [3.3.1.0<2,4>] nonane ring systems, e.g. tropane or granatane alkaloids, scopolamine; Cyclic acetals thereof
    • C07D451/02Heterocyclic compounds containing 8-azabicyclo [3.2.1] octane, 9-azabicyclo [3.3.1] nonane, or 3-oxa-9-azatricyclo [3.3.1.0<2,4>] nonane ring systems, e.g. tropane or granatane alkaloids, scopolamine; Cyclic acetals thereof containing not further condensed 8-azabicyclo [3.2.1] octane or 3-oxa-9-azatricyclo [3.3.1.0<2,4>] nonane ring systems, e.g. tropane; Cyclic acetals thereof
    • C07D451/04Heterocyclic compounds containing 8-azabicyclo [3.2.1] octane, 9-azabicyclo [3.3.1] nonane, or 3-oxa-9-azatricyclo [3.3.1.0<2,4>] nonane ring systems, e.g. tropane or granatane alkaloids, scopolamine; Cyclic acetals thereof containing not further condensed 8-azabicyclo [3.2.1] octane or 3-oxa-9-azatricyclo [3.3.1.0<2,4>] nonane ring systems, e.g. tropane; Cyclic acetals thereof with hetero atoms directly attached in position 3 of the 8-azabicyclo [3.2.1] octane or in position 7 of the 3-oxa-9-azatricyclo [3.3.1.0<2,4>] nonane ring system
    • C07D451/06Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D453/00Heterocyclic compounds containing quinuclidine or iso-quinuclidine ring systems, e.g. quinine alkaloids
    • C07D453/02Heterocyclic compounds containing quinuclidine or iso-quinuclidine ring systems, e.g. quinine alkaloids containing not further condensed quinuclidine ring systems
    • 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
    • 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
    • C07D471/08Bridged systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/08Bridged systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/99Enzyme inactivation by chemical treatment

Definitions

  • the present invention relates to compounds useful as inhibitors of c-MET.
  • the invention also provides pharmaceutically acceptable compositions comprising the compounds of the invention and methods of using the compositions in the treatment of various disorders.
  • Hepatocyte growth factor also known as scatter factor
  • HGF is a multifunctional growth factor that enhances transformation and tumor development by inducing mitogenesis and cell motility. Further, HGF promotes metastasis by stimulating cell motility and invasion through various signaling pathways.
  • HGF In order to produce cellular effects, HGF must bind to its receptor, c-MET, a receptor tyrosine kinase.
  • c-MET a widely expressed heterodimeric protein comprising of a 50 kilodalton (kDa) ⁇ -subunit and a 145 kDa alpha- subunit (Maggiora et al., J.
  • c-MET is overexpressed in a significant percentage of human cancers and is amplified during the transition between primary tumors and metastasis.
  • the various cancers in which c-MET overexpression is implicated include, but are not limited to, gastric adenocarcinoma, renal cancer, small cell lung carcinoma, colorectal cancer, prostate cancer, brain cancer, liver cancer, pancreatic cancer, and breast cancer.
  • c-MET is also implicated in atherosclerosis and lung fibrosis. Accordingly, there is a great need to develop compounds useful as inhibitors of c-MET protein kinase receptor.
  • the invention also provides pharmaceutical compositions that include a compound of formula I and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
  • the invention provides methods of treating or lessening the severity of a proliferative disease, condition, or disorder in a patient that includes the step of administering to the patient a therapeutically effective dose of a compound of formula I, or a pharmaceutical composition thereof.
  • compounds of the invention may optionally be substituted with one or more substituents, such as are illustrated generally above, or as exemplified by particular classes, subclasses, and species of the invention.
  • substituents such as are illustrated generally above, or as exemplified by particular classes, subclasses, and species of the invention.
  • phrase “optionally substituted” is used interchangeably with the phrase “substituted or unsubstituted.”
  • substituted refers to the replacement of one or more hydrogen radicals in a given structure with the radical of a specified substituent.
  • an optionally substituted group may have a substituent at each substitutable position of the group.
  • the substituent may be either the same or different at each position.
  • the term “optionally substituted” precedes a list, said term refers to all of the subsequent substitutable groups in that list. For example, if X is halogen; optionally substituted Ci -3 alkyl or phenyl; X may be either optionally substituted alkyl or optionally substituted phenyl. Likewise, if the term “optionally substituted” follows a list, said term also refers to all of the substitutable groups in the prior list unless otherwise indicated.
  • Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds.
  • stable refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, preferably, their recovery, purification, and use for one or more of the purposes disclosed herein.
  • a stable compound or chemically feasible compound is one that is not substantially altered when kept at a temperature of 40°C or less, in the absence of moisture or other chemically reactive conditions, for at least a week.
  • aliphatic or "aliphatic group,” as used herein, means a straight-chain (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation. Unless otherwise specified, aliphatic groups contain 1-20 carbon atoms. In some embodiments, aliphatic groups contain 1-10 carbon atoms. In other embodiments, aliphatic groups contain 1-8 carbon atoms. In still other embodiments, aliphatic groups contain 1-6 carbon atoms, and in yet other embodiments, aliphatic groups contain 1-4 carbon atoms.
  • Suitable aliphatic groups include, but are not limited to, linear or branched, substituted or unsubstituted alkyl, alkenyl, or alkynyl groups. Further examples of aliphatic groups include methyl, ethyl, propyl, butyl, isopropyl, isobutyl, vinyl, and sec-butyl.
  • alkyl and the prefix "alk-,” as used herein, are inclusive of both straight chain and branched saturated carbon chain.
  • alkylene represents a saturated divalent straight or branched chain hydrocarbon group and is exemplified by methylene, ethylene, isopropylene and the like.
  • alkylidene represents a divalent straight chain alkyl linking group.
  • alkenyl represents monovalent straight or branched chain hydrocarbon group containing one or more carbon-carbon double bonds.
  • alkynyl represents a monovalent straight or branched chain hydrocarbon group containing one or more carbon-carbon triple bonds.
  • cycloaliphatic refers to a monocyclic C 3 -C 8 hydrocarbon or bicyclic C 8 -Ci 2 hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule, and wherein any individual ring in said bicyclic ring system has 3-7 members.
  • Suitable cycloaliphatic groups include, but are not limited to, cycloalkyl, cycloalkenyl, and cycloalkynyl.
  • aliphatic groups include cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, and cycloheptenyl.
  • heterocycle refers to a monocyclic, bicyclic, or tricyclic ring system in which at least one ring in the system contains one or more heteroatoms, which is the same or different, and that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, and that has a single point of attachment to the rest of the molecule.
  • the "heterocycle,” “heterocyclyl,” “heterocycloaliphatic,” or “heterocyclic” group has three to fourteen ring members in which one or more ring members is a heteroatom independently selected from oxygen, sulfur, nitrogen, or phosphorus, and each ring in the system contains 3 to 8 ring members.
  • heterocyclic rings include, but are not limited to, the following monocycles: tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, tetrahydrothiophen-2-yl, tetrahydrothiophen-3-yl, 2-morpholino, 3-morpholino, 4-morpholino, 2-thiomorpholi ⁇ o, 3- thiomorpholino, 4-thiomorpholino, pyrrolidin-1-yl, pyrrolidin-2-yl, pyrrolidin-3-yl, tetrahydropiperazin-1-yl, tetrahydropiperazin-2-yl, tetrahydropiperazin-3-yl, piperidin-1-yl, piperidm-2-yl, piperidin-3-yl, piperidin-4-yl, pyrazolin-1-yl, pyrazolin-3-yl, pyrazolin-4-yl, pyrazolin
  • heteroatom means one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon, including any. oxidized form of nitrogen, sulfur, or phosphorus; the quaternized form of any basic nitrogen; or a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or NR + (as in N- substituted pyrrolidinyl).
  • alkoxy refers to an alkyl group, as previously defined, attached to the principal carbon chain through an oxygen (“alkoxy”) or sulfur (“thioalkyl”) atom.
  • haloalkyl means alkyl, alkenyl, or alkoxy, as the case may be, substituted with one or more halogen atoms.
  • halogen means F, Cl, Br, or I.
  • aralkoxy refers to monocyclic, bicyclic, and tricyclic carbocyclic ring systems having a total of six to fourteen ring members, wherein at least one ring in the system is aromatic, wherein each ring in the system contains 3 to 7 ring members and that has a single point of attachment to the rest of the molecule.
  • aryl may be used interchangeably with the term “aryl ring.” Examples of aryl rings would include phenyl, naphthyl, and anthracene.
  • heteroaryl used alone or as part of a larger moiety as in
  • heteroaryl refers to monocyclic, bicyclic, and tricyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic, at least one ring in the system contains one or more heteroatoms, wherein each ring in the system contains 3 to 7 ring members and that has a single point of attachment to the rest of the molecule.
  • heteroaryl may be used interchangeably with the term
  • heteroaryl ring or the term “heteroaromatic.”
  • Further examples of heteroaryl rings include the following monocycles: furanyl (e.g., furan-2-yl or furan-3-yl); imidazolyl (e.g., N- imidazolyl, imidazol-2-yl, imidazol-4-yl, or imidazol-5-yl); isoxazolyl (e.g., isoxazol-3-yl, isoxazol-4-yl, isoxazol-5-yl); oxazolyl (e.g., oxazol-2-yl, oxazol-4-yl, or oxazol-5-yl); pyrrolyl (e.g., N-pyrrolyl, pyrrol-2-yl, or pyrrol-3-yl); pyridinyl (e.g., pyrid-2-yl, pyrid-3-yl, or
  • an aryl (including aralkyl, aralkoxy, aryloxyalkyl and the like) or heteroaryl (including heteroaralkyl and heteroarylalkoxy and the like) group may contain one or more substituents. Suitable substituents on the unsaturated carbon atom of an aryl or heteroaryl group are selected from those listed in the definition of R 1 , R 2 , R 3 , R 4 , J M , J Q , or J R below.
  • Optional substituents on the aliphatic group of R° are selected from NH 2 , NH(Ci ⁇ aliphatic), N ⁇ M aliphatich, halogen, C M aliphatic, OH, O(Ci. 4 aliphatic), NO 2 , CN, CO 2 H, CO 2 (C ( ⁇ aliphatic), 0(1IaIoCi -4 aliphatic), or haloC]_4aliphatic, wherein each of the foregoing C M aliphatic groups of R° is unsubstituted.
  • an aliphatic, cycloaliphatic, heteroaliphatic group, or a non-aromatic heterocyclic ring may contain one or more substituents.
  • Optional substituents on the aliphatic group of R * are selected from NH 2 , NH(C (-4 aliphatic), N(Ci -4 aliphatic) 2 , halogen, Ci -4 aliphatic, OH, 0(Ci -4 aliphatic), NO 2 , CN, CO 2 H, CO 2 (C M aliphatic), O(halo C1.4 aliphatic), or halo(Ci_ 4 aliphatic), wherein each of the foregoing Cj. 4 aliphatic groups of R is unsubstituted.
  • Optional substituents on the aliphatic group or the phenyl ring of R + are selected from -NH 2 , -NH(C 1 . 4 aliphatic), -N(Ci -4 aliphatic) 2 , halogen, Ci -4 aliphatic, -OH, -0(Ci -4 aliphatic), -NO 2 , -CN, -C(O)OH, - C(O)O(C M aliphatic), -O(halo(Ci- 4 aliphatic)), or halo(Ci- 4 aliphatic), wherein each of the foregoing Ci ⁇ aliphatic groups of R + is unsubstituted.
  • two independent occurrences of R 0 may be taken together with the atom(s) to which each variable is bound to form a phenyl, 5-8-membered heterocyclyl, 5-8-membered heteroaryl, or a 3-8 membered cycloalkyl ring.
  • Exemplary rings that are formed when two independent occurrences of R° (or R + , or any other variable similarly defined herein) are taken together with the atom(s) to which each variable is bound include, but are not limited to the following: a) two independent occurrences of R° (or R + , or any other variable similarly defined herein) that are bound to the same atom and are taken together with that atom to form a ring, for example, N(R°) 2 , where both occurrences of R° are taken together with the nitrogen atom to form a piperidin-1-yl, piperazin-1-yl, or morpholin-4-yl group; and b) two independent occurrences of R° (or R , or any other variable similarly defined herein) that are bound to different atoms and are taken together with both of those atoms to form a ring, for
  • a methylene unit of the alkyl or aliphatic chain is optionally replaced with another atom or group.
  • the optional replacements form a chemically stable compound.
  • Optional atom or group replacements can occur both within the chain and at either end of the chain; i.e. both at the point of attachment and/or also at the terminal end. Two optional replacements can also be adjacent to each other within a chain so long as it results in a chemically stable compound.
  • the replacement atom is bound to an H on the terminal end. For example, if one methylene unit Of-CH 2 CH 2 CHa was optionally replaced with -O-, the resulting compound could be -OCH 2 CH 3 , -CH 2 OCH 3 , or -CH 2 CH 2 OH.
  • a bond drawn from a substituent to the center of one ring within a multiple-ring system represents substitution of the substituent at any substitutable position in any of the rings within the multiple ring system.
  • Figure a represents possible substitution in any of the positions shown in Figure b.
  • protecting group represent those groups intended to protect a functional group, such as, for example, an alcohol, amine, carboxyl, carbonyl, etc., against undesirable reactions during synthetic procedures. Commonly used protecting groups are disclosed in Greene and Wuts, Protective Groups in Organic Synthesis, 3 rd Edition (John Wiley & Sons, New York, 1999), which is incorporated herein by reference.
  • nitrogen protecting groups include acyl, aroyl, or carbamyl groups such as formyl, acetyl, propionyl, pivaloyl, t-butyl acetyl, 2-chloroacetyl, 2-bromoacetyl, trifluoroacetyl, trichloro acetyl, phthalyl, o-nitrophenoxyacetyl, ⁇ -chlorobutyryl, benzoyl, A- chlorobenzoyl, 4-bromobenzoyl, 4-nitrobenzoyl and chiral auxiliaries such as protected or unprotected D, L or D, L-amino acids such as alanine, leucine, phenylalanine and the like; sulfonyl groups such as benzenesulfonyl, p-toluenesulfonyl and the like; carbamate groups such as benzyloxycarbonyl, p-ch
  • Preferred N-protecting groups are formyl, acetyl, benzoyl, pivaloyl, t-butylacetyl, alanyl, phenylsulfonyl, benzyl, t-butyloxycarbonyl (Boc) and benzyloxycarbonyl (Cbz).
  • prodrug represents a compound that is transformed in vivo into a compound of formula I, I- A, I-B, I-C, I-D, or I-E, or a compound listed in Tables 1-5. Such a transformation can be affected, for example, by hydrolysis in blood or enzymatic transformation of the prodrug form to the parent form in blood or tissue.
  • Prodrugs of the compounds of the invention may be, for example, esters.
  • Esters that may be utilized as prodrugs in the present invention are phenyl esters, aliphatic (C 1 -C2 4 ) esters, acyloxymethyl esters, carbonates, carbamates, and amino acid esters.
  • a compound of the invention that contains an OH group may be acylated at this position in its prodrug form.
  • Other prodrug forms include phosphates, such as, for example those phosphates resulting from the phosphorylation of an OH group on the parent compound.
  • structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the (R) and (S) configurations for each asymmetric center, (Z) and (E) double bond isomers, and (Z) and (E) conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. [0030] Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention.
  • structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms.
  • compounds having the present structures except for the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13 C- or 14 C-enriched carbon are within the scope of this invention.
  • Such compounds are useful, for example, as analytical tools or probes in biological assays, or as c- MET inhibitors with improved therapeutic profile.
  • the invention features a compound having the formula:
  • Ring D is the selected from:
  • Ring B is a 5- or 6- membered aryl, cycloaliphatic, heteroaryl, or heterocyclyl ring, wherein said ring is optionally substituted with up to 4 occurrences of R B and said heteroaryl or heterocyclyl ring contains up to three heteroatoms selected from N, O, or S; each R B is independently selected from halogen, R B 1 , -CN, -CO 2 R 81 , -OC(O)R 81 ,
  • each R B1 is, independently, hydrogen or Ci -4 aliphatic, or two R BI together with the atom to which they are bound, form a 3-6 membered carbocycle optionally substituted with 0-2 occurrences of J R or a 3-6 membered heterocyclyl containing 1 -3 heteroatoms independently selected from N, O, or S and optionally substituted with 0-2 occurrences of J R on carbon and optionally substituted with J N on each substitutable ring nitrogen atom;
  • Q is C 6 - I o aryl or 5-10 membered heteroaryl wherein each Q is optionally substituted with up to 5 occurrences of J Q ;
  • U is N or CR 1 ;
  • V is N or CR 2 ;
  • U 1 is O, NR 5 , or S;
  • V I is O, NR 6 , or S;
  • R 1 is hydrogen, halogen, -CN, -NH 2 , -OH, C 1 - 2 haloalkyl, or selected from -NH(C M aliphatic), -N(C M aliphatic) 2 , C 3-4 cycloalkyl, -(Cj -2 aliphatic)-(C 3 ⁇ cycloalkyl), or Ci -4 aliphatic, each of which is optionally substituted with up to 2 occurrences of J R ;
  • R 2 is hydrogen, halogen, -CN, -NH 2 , -NH(C M aliphatic), -N(C M aliphatic) 2 , C1-2 haloalkyl, C3.4 cycloalkyl, or CM aliphatic;
  • R 3 is halogen or R A , wherein R A is C ⁇ -io aryl, 5-10 membered heteroaryl, 5-12 membered heterocyclyl, or C 3-S cycloaliphatic, each of which is optionally substituted with 0-3 occurrences of J M ;
  • R 4 is hydrogen, -CN, C M aliphatic, halogen or Ci -2 haloalkyl; each of R 5 , R 6 , and R 7 , is, independently, hydrogen or J N ; each J M is independently selected from halogen, -NO 2 , -CN, C M aliphatic, Ci -2 haloalkyl, -(CH 2 ) 0-2 CH(R') 2 , -OH, -OR', -(CR'" 2 ) q NH 2 , -(CR"' 2 ) q NHR ⁇ -(CR"' 2 ) q N(R') 2 , -(CR'" 2 ) q NHS(O) 2 R', -(CR'" 2 ) q NHC(O)R ⁇ -(CR'" 2 ) q NHC(O)OR ⁇ -(CR'" 2 ) q NHC(O)NH 2 , -(CR"' 2 )
  • each J N is independently selected from aliphatic, -(CR 5 " 2 ) q 'C 3-6 cycloalkyl, -(CR' ' ' 2 Vphenyl, -(CR' ' ' 2 ) q >C(O)C , ⁇ aliphatic; -(CR' ' ' 2 ) q .C(O)C l-2 haloalkyl; -C(O)O(C M alkyl), -(CR" ' 2 ) q -C(O)NH 2> -(CR"' 2 ) q' C(O)NH(C ⁇ aliphatic), -(CR'" 2 )q ' C(O)N(Ci- 4 aliphatic) 2 , or -S(O) 2 C M aliphatic 5 wherein q' is 0-2 and each aliphatic or cycloaliphatic is optionally substituted with up to 2 occurrences of J R
  • Z 1 is N or CR 4 ;
  • Z 2 is N or CH
  • Ring D is the selected from one of the 5-membered rings shown below:
  • each of R 5 and R 6 is hydrogen, Ci -2 haloalkyl, or selected from C 3-4 cycloalkyl, -(C i-2 aliphatic)-(C3- 4 cycloalkyl), or C] -4 aliphatic, each of which is optionally substituted with up to 2 occurrences of J R ;
  • R 3 is a C ⁇ -io aryl, 5-10 membered heteroaryl, 5-10 membered heterocyclyl, or C 3- S cycloaliphatic, each of which is optionally substituted with 0-3 occurrences of J M ; each J Q is, independently, selected from halogen, -NO 2 , -CN, Ci -4 aliphatic, Ci -4 haloalkyl, -OH, -OR", -NH 2 , -NHR", -N(R") 2 , -SH, -SR", -CO 2 H, -CO 2 R", -C(O)H, -C(O)R", -C(O)NH 2 , -C(O)NHR", -C(O)N(R") 2 , -C(O)N(OH)R", -C(0)N(OR")R", -C(O)N(OR”)H, -C(O)N(OH)H, -C(NOH)
  • each R'" is, independently, selected from hydrogen or Ci -4 aliphatic.
  • Ring D is selected from:
  • Ring D is selected from:
  • Ring D is
  • Ring D is selected from
  • Ring D is selected from
  • Ring D is selected from
  • Ring D is
  • Ring A is selected from
  • Ring A is
  • R 4 is hydrogen.
  • Q is
  • each J ⁇ is, independently, fluoro or chloro, such as, for example, when Q is
  • R 3 is R A , which is a C 6- ⁇ o aryl, a C 3-8 cycloaliphatic, or a monocyclic or bicyclic 5-10 membered heteroaryl or heterocyclyl containing 1-4 heteroatoms independently selected from N, O, or S, wherein said aryl, cycloaliphatic, heteroaryl, or heterocyclyl is optionally substituted with up to 3 occurrences of J M .
  • R A is phenyl optionally substituted with up to 3 occurrences of J M .
  • R A is a 5-6 membered heteroaryl optionally substituted with up to 3 occurrences of J M , such as, for example, an optionally substituted pyridyl, thienyl, thiazolyl, isothiazolyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, furanyl, isoxazolyl, or oxazolyl.
  • Further examples include lH-pyrazol-4-yl substituted at the 1- position with J M , thiophen-2-yl substituted at the 5-position with J M , thiophen-3-yl substituted at the 5-position with J M , furan-2-yl substituted at the 5-position with J M , furan-3- yl substituted at the 5-position with J M , l//-pyrrol-3-yl substituted at the 1 -position with J M , lH-l,2,3-triazol-4-yl substituted at the 1-position with J M , or thiazol-5-yl substituted at the 2- position with J M .
  • J M examples include those where J M is selected from phenyl, 5-8 membered heteroaryl, 5-10 membered heterocyclyl, or C 3 .scycloaliphatic; each optionally substituted with up to 3 occurrences of J N or J R . hi a further example, J M is an optionally substituted 5-
  • 10 membered heterocyclyl containing 1 or 2 nitrogen atoms such as, for example an optionally substituted piperidine, piperazine or pyrrolidine or an optionally substituted bicyclooctane or bicyclononane containing 1 or 2 nitrogen atoms.
  • R A is a Cs-io bicyclic heteroaryl selected from:
  • Ring E is a 5-membered heteroaryl ring with 1 to 2 heteroatoms selected from N, O, or S; and J E is hydrogen or J N .
  • Ring E is selected from thienyl, thiazolyl, pyrrolyl, imidazolyl, furanyl, or oxazolyl.
  • R A is
  • J F is selected from Ci -4 aliphatic, -C 0-2 aliphaticC ⁇ (R') 2 , -(CR" ' 2 )qNH 25 -(CR"' 2 ) q NHR ⁇ -(CR'" 2 ) q N(R') 2 , -(CR'" 2 ) q NS(O) 2 R', -(CR'" 2 ) q NHC(O)R', -(CR'" 2 ) q NHC(O)OR', -(CR'” 2 ) q NHC(O)NH 2 , -(CR'" 2 ) q NHC(O)NHR', -(CR' ' ' 2 ) q NHC(O)N(R') 2 , -(CR' ' ' 2 ) q NHC(NH)NH 2 , -(CR' ' ' 2 ) q NHC(NH)NHR' , -(CR
  • R A is
  • J F is -C 0-2 aliphaticCH(R') 2 , -(CR'” 2 ) q NH 2 , -(CR'" 2 ) q NHR' 5 -(CR'" 2 ) q N(R') 2j -C(O)R', -C(0)-(CH 2 )o-2CH(R') 2 , or optionally substituted heterocyclyl.
  • J M is selected from halogen, -NO 2 , -CN, C 1-4 aliphatic, C 1-2 haloalkyl, -OH, -OR', -(CR'" 2 ) q NH 2 , -(CR"' 2 ) q NHR ⁇ -(CR'" 2 ) q N(R') 2 , -(CR'" 2 ) q NS(O) 2 R', -(CR"' 2 ) q NHC(O)R ⁇ -(CR"' 2 ) q NHC(O)OR ⁇ -(CR' ' ' 2 ) q NHC(O)NH 2 , -(CR' ' ' 2 ) q NHC(O)NHR ⁇ -(CR' ' ' 2 ) q NHC(O)N(R') 2 , -(CR'" 2 ) q NHS(O) 2 NH 2
  • q is 1 or 2.
  • J M is not substituted on a ring position adjacent to
  • a substituted or unsubstituted C M or C, -6 aliphatic group of R 1 , R 2 , R 4 , R 5 , R 6 , J M , J N , J Q , J R , R', R", or R'" comprises two or more non-hydrogen atoms.
  • the invention features a compound having the formula:
  • I-A I B I C I-D or I E wherein said compound is selected from the compounds of Tables 1, 2, 3, 4, or 5, respectively.
  • the invention provides a pharmaceutical composition comprising a compound of any of the formulae or classes described herein.
  • the invention provides a pharmaceutical composition comprising a compound of Tables 1, 2, 3, 4, or 5.
  • the composition additionally comprises an additional therapeutic agent.
  • the invention provides a composition comprising a compound of this invention or a pharmaceutically acceptable derivative thereof and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
  • the amount of compound in a composition of this invention is such that is effective to measurably inhibit c-MET in a biological sample or in a patient.
  • the composition of this invention is formulated for administration to a patient in need of such composition.
  • the composition of this invention is formulated for oral administration to a patient.
  • patient means an animal, preferably a mammal, and most preferably a human.
  • a pharmaceutically acceptable derivative includes, but is not limited to, pharmaceutically acceptable prodrugs, salts, esters, salts of such esters, or any other adduct or derivative which upon administration to a patient in need is capable of providing, directly or indirectly, a compound as otherwise described herein, or a metabolite or residue thereof.
  • the term "pharmaceutically acceptable salt” refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like.
  • compositions of this invention include those derived from suitable inorganic and organic acids and bases.
  • Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid
  • organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate,
  • Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N + (Ci -4 alkyl) 4 salts.
  • This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein. Water or oil-soluble or dispersable products may be obtained by such quaternization.
  • Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
  • Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, C i- 8 sulfonate and aryl sulfonate.
  • compositions of the present invention additionally comprise a pharmaceutically acceptable carrier, adjuvant, or vehicle, which, as used herein, includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired.
  • a pharmaceutically acceptable carrier includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired.
  • Some examples of materials which can serve as pharmaceutically acceptable carriers include, but are not .limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, or potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, polyacrylates, waxes, polyethylene- polyoxypropylene-block polymers, wool fat, sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; ge
  • compositions of the present invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
  • parenteral as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intraocular, intrahepatic, intralesional and intracranial injection or infusion techniques.
  • the compositions are administered orally, intraperitoneally or intravenously.
  • Sterile injectable forms of the compositions of this invention may be aqueous or oleaginous suspension.
  • suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1 ,3-butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • Fatty acids such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions.
  • compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions.
  • carriers commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried cornstarch.
  • the pharmaceutically acceptable compositions of this invention may be administered in the form of suppositories for rectal administration. These can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug. Such materials include cocoa butter, beeswax and polyethylene glycols.
  • a suitable non-irritating excipient that is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug.
  • Such materials include cocoa butter, beeswax and polyethylene glycols.
  • the pharmaceutically acceptable compositions of this invention may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations are readily prepared for each of these areas or organs.
  • Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically- transdermal patches may also be used.
  • the pharmaceutically acceptable compositions may be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers.
  • Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water.
  • the pharmaceutically acceptable compositions can be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers.
  • Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • the pharmaceutically acceptable compositions may be formulated, e.g., as micronized suspensions in isotonic, pH adjusted sterile saline or other aqueous solution, or, preferably, as solutions in isotonic, pH adjusted sterile saline or other aqueous solution, either with or without a preservative such as benzylalkonium chloride.
  • the pharmaceutically acceptable compositions may be formulated in an ointment such as petrolatum.
  • the pharmaceutically acceptable compositions of this invention may also be administered by nasal aerosol or inhalation.
  • compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluoro carbons, and/or other conventional solubilizing or dispersing agents.
  • compositions of this invention are formulated for oral administration.
  • Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3 -butyl ene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents such as, for example, water or other solvents, so
  • Injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S. P. and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil can be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid are used in the preparation of injectables.
  • the injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
  • the rate of compound release can be controlled.
  • biodegradable polymers include poly(orthoesters) and poly( anhydrides).
  • Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues.
  • compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol
  • the dosage form may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner.
  • embedding compositions examples include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polethylene glycols and the like.
  • the active compounds can also be in micro-encapsulated form with one or more excipients as noted above.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art.
  • the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch.
  • inert diluent such as sucrose, lactose or starch.
  • Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose.
  • the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes.
  • Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches.
  • the active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required.
  • Ophthalmic formulation, eardrops, and eye drops are also contemplated as being within the scope of this invention.
  • the present invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body.
  • Such dosage forms can be made by dissolving or dispensing the compound in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the compound across the skin.
  • the rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
  • the compounds of the invention are preferably formulated in dosage unit form for ease of administration and uniformity of dosage.
  • dosage unit form refers to a physically discrete unit of agent appropriate for the patient to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific effective dose level for any particular patient or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts.
  • the amount of the compounds of the present invention that may be combined with the carrier materials to produce a composition in a single dosage form will vary depending upon the host treated, the particular mode of administration.
  • compositions should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of the inhibitor can be administered to a patient receiving these compositions.
  • additional therapeutic agents which are normally administered to treat or prevent that condition, may also be present in the compositions of this invention.
  • additional therapeutic agents that are normally administered to treat or prevent a particular disease, or condition, are known as "appropriate for the disease, or condition, being treated.” Examples of additional therapeutic agents are provided infra.
  • the amount of additional therapeutic agent present in the compositions of this invention will be no more than the amount that would normally be administered in a composition comprising that therapeutic agent as the only active agent.
  • the amount of additional therapeutic agent in the presently disclosed compositions will range from about 50% to 100% of the amount normally present in a composition comprising that agent as the only therapeutically active agent.
  • the invention relates to a method of inhibiting c- MET protein kinase activity in a biological sample comprising the step of contacting said biological sample with a compound of this invention, or a composition comprising said compound.
  • biological sample means a sample outside a living organism and includes, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof.
  • Inhibition of kinase activity in a biological sample is useful for a variety of purposes known to one of skill in the art. Examples of such purposes include, but are not limited to, biological specimen storage and biological assays.
  • the method of inhibiting kinase activity in a biological sample is limited to non- therapeutic methods.
  • the invention relates to a method of inhibiting c-MET kinase activity in a patient comprising the step of administering to said patient a compound of the present invention, or a composition comprising said compound.
  • c-MET-mediated disease or "c-MET-mediated condition” means any disease state or other deleterious condition in which c-MET is known to play a role.
  • c-MET-mediated disease or "c-MET-mediated condition” also mean those diseases or conditions that are alleviated by treatment with a c-MET inhibitor.
  • Such conditions include, without limitation, renal, gastric, colon, brain, breast, prostate, and lung cancer, glioblastoma, atherosclerosis, lung fibrosis, conditions associated with organ transplantation, allergic disorders, and autoimmune disorders.
  • the present invention features a method treating a proliferative disorder in a patient comprising the step of administering to the patient a therapeutically effective dose of any of the compounds or compositions of the invention.
  • the proliferative disorder is cancer, such as, for example, renal, gastric, colon, brain, breast, liver, prostate, and lung cancer, or a glioblastoma.
  • the present invention relates to a method of treating or lessening the severity of brain cancer in a patient in need thereof, comprising administering to said patient a compound of the present invention or composition thereof.
  • the proliferative disorder is polycythemia vera, essential thrombocythemia, chronic idiopathic myelofibrosis, myeloid metaplasia with myelofibrosis, chronic myeloid leukemia (CML), chronic myelomonocytic leukemia, chronic eosinophilic leukemia, hypereosinophilic syndrome, systematic mast cell disease, atypical CML, or juvenile myelomonocytic leukemia.
  • the proliferative disorder is atherosclerosis or lung fibrosis.
  • Another aspect of the present invention relates to a method of inhibiting tumor metastasis in a patient in need thereof, comprising administering to said patient a compound of the present invention or composition thereof.
  • additional therapeutic agents which are normally administered to treat that condition, may also be present in the compositions of this invention.
  • additional therapeutic agents that are normally administered to treat a particular disease, or condition are known as "appropriate for the disease, or condition, being treated”.
  • chemotherapeutic agents or other antiproliferative agents maybe combined with the compounds of this invention to treat proliferative diseases and cancer.
  • chemotherapeutic agents include, but are not limited to, alkylating agents, such as, for example, cyclophosphamide, lomustine, busulfan procarbazine, ifosfamide, altretamine, melphalan, estramustine phosphate, hexamethylmelamine, mechlorethamine, thiotepa, streptozocin, chlorambucil, temozolomide, dacarbazine, semustine, or carmustine; platinum agents, such as, for example, cisplatin, carboplatinum, oxaliplatin, ZD-0473 (AnorMED), spiroplatinum, lobaplatin (Aeterna), carboxyphthalatoplatinum, satraplatin (Johnson Matthey), tetraplatin BBR-
  • alkylating agents such
  • Those additional agents may be administered separately from the compound- containing composition, as part of a multiple dosage regimen. Alternatively, those agents may be part of a single dosage form, mixed together with the compound of this invention in a single composition. If administered as part of a multiple dosage regime, the two active agents maybe submitted simultaneously, sequentially or within a period of time from one another normally within five hours from one another.
  • compositions of this invention should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of a compound of formula I can be administered.
  • compositions that comprise an additional therapeutic agent that additional therapeutic agent and the compound of this invention may act synergistically. Therefore, the amount of additional therapeutic agent in such compositions will be less than that required in a monotherapy utilizing only that therapeutic agent. In such compositions a dosage of between 0.01 - 100 mg/kg body weight/day of the additional therapeutic agent can be administered.
  • the amount of additional therapeutic agent present in the compositions of this invention will be no more than the amount that would normally be administered in a composition comprising that therapeutic agent as the only active agent.
  • the amount of additional therapeutic agent in the presently disclosed compositions will range from about 50% to 100% of the amount normally present in a composition comprising that agent as the only therapeutically active agent.
  • the compounds of this invention, or pharmaceutical compositions thereof, may also be incorporated into compositions for coating an implantable medical device, such as prostheses, artificial valves, vascular grafts, stents and catheters.
  • an implantable medical device such as prostheses, artificial valves, vascular grafts, stents and catheters.
  • Vascular stents for example, have been used to overcome restenosis (re-narrowing of the vessel wall after injury).
  • patients using stents or other implantable devices risk clot formation or platelet activation. These unwanted effects may be prevented or mitigated by pre-coating the device with a pharmaceutically acceptable composition comprising a kinase inhibitor.
  • Suitable coatings and the general preparation of coated implantable devices are described in US Patents 6,099,562; 5,886,026; and 5,304,121.
  • the coatings are typically biocompatible polymeric materials such as a hydrogel polymer, polymethyldisiloxane, polycaprolactone, polyethylene glycol, polylactic acid, ethylene vinyl acetate, and mixtures thereof.
  • the coatings may optionally be farther covered by a suitable topcoat of fluorosilicone, polysaccarides, polyethylene glycol, phospholipids or combinations thereof to impart controlled release characteristics in the composition.
  • Implantable devices coated with a compound of this invention are another embodiment of the present invention.
  • J is used to represent an iodine atom. Lawesson's
  • R t (min) refers to the HPLC retention time, in minutes, associated with the compound. Unless otherwise indicated, the HPLC method utilized to obtain the reported retention time is as follows: column: Zorbax SB Cl 8 column, 3.0 x 150 mm; gradient: 10-90% acetonitrile/water (0.1%TFA), 5 minutes; flow rate: 1.0 mL/minute; and detection: 254 & 214 nm.
  • the compounds of this invention may be prepared by methods described herein or known to those skilled in the art for the preparation of analogous compounds.
  • the following non-limiting schemes and examples are presented to further exemplify the invention.
  • Physiochemical characterization of selected compounds of the invention is provided in Tables 6-10.
  • R ⁇ include optionally substituted C ⁇ -io aryls, 5-10 membered monocyclic or bicyclic heteroaryls, 5-10 membered monocyclic or bicyclic heterocyclyls, or 5-7 membered cycloaliphatics containing at least one point of unsaturation.
  • the Metal group can be, for example, -B(OAlkyl)2 or -B(OH) 2 (Suzuki reaction), -Mg-HaI (Kumada reaction), -Zn-HaI (Negishi reaction), -Sn(Alkyl) 3 (Stille reaction), -Si(Alkyl) 3 (Hiyama reaction), -Cu- HaI, -ZrCp2Cl, or -AlMe 2 .
  • the catalyst for the cross-coupling reaction can be, for example, a palladium catalyst/ligand system (such as, for example, Pd(PPb.
  • compound of formula I-a wherein Z 1 , Z 2 , Q and Ring D are as defined for a compound of formula I, can be transformed into a boronate or boronic acid of formula I-b, as shown in Scheme 2.
  • Subsequent reaction with an aryl, heteroaryl, or cycloalkenyl halide produces a compound of formula I-c (a compound of formula I, wherein R 3 is R A ).
  • 3-amino-iV-(2,3-dichlorophenyl)pyrazine- 2-carboxamide and 3-amino-Vv " -(3-chloro-2-fluorophenyl)pyrazme-2-carboxamide can be produced by reacting 3-aminopyrazine-2-carboxylic acid with 2,3-dichloroaniline and 3- chloro-2-fluoroaniline, respectively.
  • this procedure can be used to produce 2- amino- ⁇ f-(2,3-difluorophenyl)pyridine-3-carboxamide, 2-amino-iV " -(2,3- dichlorophenyl)pyridine-3-carboxarnide, and 2-amino--V-(3-chloro-2-fmorophenyl)pyridiine- 3-carboxamide by reacting 2-amino-pyridine-3-carboxylic acid with 2,3-difluoroaniline, 2,3- dichloroaniline, and 3-chloro-2-fluoroaniline, respectively.
  • the reaction was warmed to RT and maintained for 3 additional hours, followed by washing the mixture with 2N HCl, saturated NaCl, and saturated NaHCO 3 solution.
  • the organic extracts were dried over MgSO 4 , filtered, and concentrated in vacuo.
  • the resulting crude 2-fluoro-N-(2,3-difluorophenyl)pyridine-3-carboxamide was dissolved in NMP (20 mL) and reacted with excess £-butylamine at 80 0 C for 14 hours. After cooling to RT, the solution was poured into sat NaHCO 3 solution. The resulting precipitate was collected by filtration and washed with water.
  • N-tert-huty ⁇ -3- ⁇ -(2,3-dichlorophenyl)-lH-tetrazol-5-yl)pyridin-2-amine and i ⁇ " -fert-butyl-3-(l-(3-chloro-2-fluorophenyl)-lH-tetrazol-5-yl)pyridin-2-amine can be produced from 2-(fe ⁇ -butylamino)-iV-(2,3-dichlorophenyl)pvridine-3-carboxamide and 2- (/er ⁇ -butylamino)-N-(3-chloro-2-fluorophenyl)pyridine-3-carboxamide, respectively.
  • 5-bromo-3-(l-(2,3-dichlorophenyl)-lH r -tetrazol-5-yl)pyridin-2-amine and 5- bromo-3-(l-(3-chloro-2-fluorophenyl)-lH-tetrazol-5-yl)pyridin-2-amine can be produced from 3-(l-(2,3-dichlorophenyl)-lH-tetrazol-5-yl)pyridin-2-amine and 3-(l-(3-chloro-2- fluorophenyl)- lH-tetrazol-5-yl)pyridin-2 -amine, respectively.
  • 5-bromo-2-chloro-iV-(2,3-fluorophenyl)pyridine-3-carboxamide and 5-bromo-2- chloro-7V-(3-chloro-2-fluorophenyl)pyridine-3-carboxamide can be produced from reacting 5- bromo-2-chloropyridine-3-carbonyl chloride with 2,3 fluororoaniline and 3-chloro-2- fluoroaniline, respectively.
  • 5-bromo-2-chloro-3- (l-(2,3-difluorophenyl)-lH-imidazol-2-yl)pyridine and 5-bromo-2-chloro-3-(l-(3-chloro-2- fluorophenyl)-li/-imidazol-2-yl)pyridine can be produced from 5-bromo-2-chloro-N-(2,3- difluoro ⁇ henyl)pyridine-3-carboxamide and 5-bromo-2-chloro-N-(3-chloro-2- fluorophenyl)p yridine-3 -carboxamide, respectively.
  • N-(4-methoxybenzyl)-5-bromo-3- 1 -(2,3-difluorophenyl)-li ⁇ -imidazol-2-yl)pyridin-2-amine
  • N-(4-methoxybenzyl)-5- bromo-3-(l-(3-chloro-2-fluorophenyl)-lH ' -imidazol-2-yl)pyridin-2-amine can be produced from 5-bromo-2-chloro-3-(l-(2,3-difluorophenyl)-lH ' -imidazol-2-yl)pyridine and 5-bromo-2- chloro-3-(l-(3-chloro-2-fluorophenyl)-lH-imidazol-2-yl)pyridine, respectively
  • N-(4-methoxybenzyl)-5- bromo-3-(5-(2,3-difluorophenyl)-2-methyloxazol-4-yl)pyridin-2-amine and 7V-(4- methoxybenzyl)-5-bromo-3-(5-(3-chloro-2-fluorophenyl)-2-methyloxazol-4-yl)pyridin-2- amine can be produced from l-(5-bromo-2-chloropyridin-3-yl)-2-(2,3- difluorophenyl)ethanone and 1 -(5-bromo-2-chloropyridin-3-yl)-2-(3-chloro-2- fluorophenyl)ethanone, respectively.
  • 5-bromo-3-(5-(2,3-fluorophenyl)-l ,2,3-thiadiazol-4- yl)pyridin-2-amine and 5-bromo-3-(5-(3-chloro-2-fiuorophenyl)-l ,2,3-thiadiazol-4- yl)pyridi ⁇ -2 -amine can be produced from l-(5-bromo-2-chloropyridin-3-yl)-2-(2,3- difluorophenyl)ethanone and 1 -(5-bromo-2-chloropyridin-3-yl)-2-(3-chloro-2- fluorophenyl)ethanone, respectively.
  • 5-bromo-3-(5-(2,3-dichlorophenyl)-lH-tetrazol-l-yl)pyridin-2-amine and 5- bromo-3-(5-(3-chloro-2-fluorophenyl)-lH-tetrazol-l -yl)pyridin-2-amine can be produced from 2,3-dichlorobenzoyl chloride and 3-chloro-2-fluorobenzoyl chloride, respectively.
  • Example 18 1 -(5-Bromo-2-chloropyridin-3-yl)-2-(2,3-dichlorophenyl)-3- (dimethylamino)prop-2-en- 1 -one
  • the mixture was cooled to RT, concentrated to dryness under reduced pressure, diluted with water, extracted with ethyl acetate, washed with saturated aqueous NaHCO 3 solution, water, and brine solution, respectively, prior to drying over Na 2 SO4.
  • Example 20 ⁇ T-(4-Methoxybenzyl)-5-bromo-3-(4-(2,3-dichlorophenyl)-l -cyclohexyl- 1 H- pyrazol-3-yl)pyridin-2-amine and 7V-(4-methoxybenzyl)-5-bromo-3-(4-(2,3- dichlorophenyl)-l-cyclohexyl-li7-pyrazol-5-yl)pyridin-2-amine
  • reaction mixture was cooled to RT, evaporated to dryness, treated with saturated aqueous NaHCO 3 solution, and extracted with EtOAc. The combined organic, layer was washed with brine solution, dried over Na 2 SO 4 , and concentrated to obtain a mixture of iV-(4-methoxybenzyl)-5-bromo-3-(4-(2,3- dichlorophenyl)-l -cyclohexyl- 1 H-pyrazol-3 -yl)pyridin-2-amine and 7V-(4-methoxybenzyl)-5- bromo-3-(4-(2,3-dichlorophenyl)-l -cyclohexyl-lH-pyrazol-5-yl)pyridin-2-amine as brown oil (350 mg, 54.6% yield).
  • reaction mixture was concentrated in vacuo and the resulting residue was taken in saturated aq. NaHCO 3 solution (100 mL) and extracted with CHCl 3 (200 mL). The organics were washed with water (3 x 100 mL), brine solution (1 x 100 mL), and dried (Na 2 SO 14 ).
  • reaction mixture was concentrated in vacuo, the residue taken in THF (500 mL), and the mixture treated with solid NaHCO 3 (40 g, 472.2 mmol) at 0 °C. This was followed by addition of ethyl chloroformate (19.5 mL, 157 mmol) and saturated aq. NaHCO3 solution slowly until the gas evolution was minimal. The reaction mixture was stirred overnight and extracted with ethyl acetate.
  • Ethyl 4,5,7,8-tetrahydrothieno[3,2-d]azepine-6-carboxylate (16.72 mmol) was dissolved in CH 3 CN (150 mL) and NBS (4.74 g, 26.63 mmol) was added. The reaction was stirred at room temperature for 30 min, and poured into a solution OfNa 2 SO 3 (200 mL)/6N NaOH (5 mL). The aqueous layer was extracted with EtOAc (3 x 150 mL), dried over MgSO4, filtered, and evaporated.
  • reaction was quenched with ice, extracted with CH 2 Cl 2 (2 x 50 mL), washed with brine solution, dried (Na 2 SO 4 ), filtered, and concentrated to obtain tert- butyl 4-(thiophen-2-yl)piperidine-l-carboxylate, which was used as is for subsequent reactions without further purification.
  • N-Bromosuccinimide (4.9 g, 28.08 mmol) was added in portions to a solution of tert-butyl 4-(thiophen-2-yl)piperidine-l-carboxylate (5.0 g, 18.72 mmol) in MeCN at -10 0 C.
  • the reaction mixture was stirred for 30 minutes and treated with sat. aq. NaHCO 3 solution.
  • the mixture was extracted with CH 2 Cl 2 and the organics washed with water and brine solution, dried (Na 2 SO 4 ), filtered, and evaporated to obtain crude product.
  • this intermediate (20 g, 72 mmol) was suspended in pentane, filtered, washed with pentane, and the solid so obtained dried in vacuo and dissolved in EtOH (100 mL).
  • Thiourea (5.46 g, 72.0 mmol) was added and the reaction mixture heated for 4 hours at 70-75 0 C.
  • tert-Bv ⁇ yl 2-amino-6,7-dihydrothiazolo[5,4-c]pyridine-5(4H)-carboxylate (8.5 g, 33.33 mmol) was added dropwise over 2 minutes to a suspension of t-butyl nitrite and CuBr 2 mixture in C ⁇ 3CN at 0 0 C. The reaction mixture was slowly warmed to RT over 30 min and stirred for 16 hours at RT. The reaction mixture was concentrated to obtain residue that was dissolved in EtOAc, filtered through a CeliteTM plug, which was washed with EtOAc.
  • 3-(l-(2,3- chlorophenyl)-lH-tetrazol-5-yl)-5-(4 ) 4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyridin-2- amine and 3-(l -(3-chloro-2-fluorophenyl)-lH-tetrazol-5-yl)-5-(4,4,5,5-tetramethyl-l ,3,2- dioxaborolan-2-yl)pyridin-2-amine can be produced from 5-bromo-3-(l-(2,3-chlorophenyl)- lH-tetrazol-5-yl)pyridin-2-amine and 5-bromo-3-(l-(3-chloro-2-fluorophenyl)-lH-tetrazol-5- yl)pyridin-2-amine, respectively.
  • Example 40 l- ⁇ -S-C ⁇ -Amino-S-Cl ⁇ -difluorophenyO-lH-tetrazol-S-yOpyridin-S-yl)- 3,4-dihydro-2H-azepin-l(7H)-yl)-2,2,2-trifluoroethanone
  • N 3 N-Di(I, l-dimethylethoxycarbonyl)-3-(l-(2,3-difluorophenyl)-lH-tetrazol-5-yl)- 5-efhynyl ⁇ yridin-2-amine (59 mg, 119 ⁇ mol) was dissolved in methylene chloride (1 mL) and to this was added tris((l-benzyl-l#-l,2,3-triazol-4-yl)rnethyl)amine (TBTA, 20 mg, 38 ⁇ mol, see Henning et al., Organic Letters 9(1): 1-4; 2007), sodium ascorbate (100 mg, 505 ⁇ mol), water (1 mL), and methanol (2 mL).
  • tert-buty ⁇ A- azidopiperidine-1-carboxylate (1 mmol) (prepared from tert-butyl 4-aminopiperidine-l- carboxylate, via the method described by Alper et al., Tetrahedron Letters 37(34): 6029- 6032, 1996) in methylene chloride (2 mL).
  • methylene chloride 2 mL
  • the reaction was left at room temperature for 30 minutes and concentrated to remove the volatile organics.
  • the reaction was extracted with methylene chloride (2 x 10 mL) and saturated sodium bicarbonate (5 mL). The organics were dried over sodium sulfate, filtered and concentrated.
  • compounds of the invention were dissolved in DMSO to make 10 mM initial stock solutions. Serial dilutions in DMSO were then made to obtain the final solutions for the assay. A 1.5 ⁇ L aliquot of DMSO or inhibitor in DMSO was added to each well. The reaction was initiated by the addition Of 33 P-ATP and polyE4Y (obtained from Sigma). After 20 min, the reaction was quenched with 50 ⁇ L of 30% trichloroacetic acid (TCA) containing 4 mM ATP. The reaction mixture was transferred to the 0.66 mm GF filter plates (Corning) and washed three times with 5% TCA.
  • TCA trichloroacetic acid
  • Compounds having a Ki of less than or equal to 0.10 ⁇ M for the inhibition of c- MET include: I-A-5, 1-A-6, 1-A-7, 1-A-8, 1-A-9, 1-A-10, 1- A-11, I-A-12, 1-A-13, 1-A-19, I- A-22, 1-A-32, 1-A-46, 1-A-48, 1-A-50, I-A-51, I-A-52, 1-A-54, I-A-56, 1-A-57, 1-A-62, I-A- 63, 1-A-64, 1-A-65, l-A-66, l-A-67, I-A-79, I-A-81, 1-A-85, I-A-86, I-A-87, l-A-90, I-A-92, I-A-94, 1-A-95, I-A-96, 1-A-97, 1-A-98, 1-A-99, ⁇ -A-100, 1-A-104, 1-A-106, I-A-
  • Compounds having a Kj of greater than 0.10 ⁇ M and less than or equal to 1.0 ⁇ M the inhibition of c-MET include I-A-2, 1-A-3, 1-A-4, 1-A-15, I-A-16, 1-A-17, 1-A-18, 1-A-20, I-A-29, 1-A-30, 1-A-31, 1-A-33, 1-A-34, 1-A-39, 1-A-40, 1-A-41, 1-A-42, 1-A-53, 1-A-55, 1- A-68, 1-A-69, 1-A-70, 1-A-71, 1-A-72, 1-A-73, 1-A-74, 1-A-75, 1-A-76, 1-A-77, 1-A-78, 1-A- 82, 1-A-83, 1-A-84, 1-A-88, 1-A-89, 1-A-93, 1-A-101, 1-A-102, 1-A-103, 1-A-105, 1-A-107, 1- A-110, ⁇ -A-l li; I-

Abstract

The present invention relates to compounds useful of inhibitors of protein kinases. The invention also provides processes for preparing the compounds of this invention, pharmaceutically acceptable compositions comprising the compounds of the invention, and methods of using the compositions in the treatment of various disorders.

Description

C-MET PROTEIN KINASE INHIBITORS
TECHNICAL FIELD OF THE INVENTION
[00011 The present invention relates to compounds useful as inhibitors of c-MET. The invention also provides pharmaceutically acceptable compositions comprising the compounds of the invention and methods of using the compositions in the treatment of various disorders.
BACKGROUND OF THE INVENTION
[0002] Hepatocyte growth factor (HGF), also known as scatter factor, is a multifunctional growth factor that enhances transformation and tumor development by inducing mitogenesis and cell motility. Further, HGF promotes metastasis by stimulating cell motility and invasion through various signaling pathways. In order to produce cellular effects, HGF must bind to its receptor, c-MET, a receptor tyrosine kinase. c-MET, a widely expressed heterodimeric protein comprising of a 50 kilodalton (kDa) α-subunit and a 145 kDa alpha- subunit (Maggiora et al., J. Cell Physiol, 173:183-186, 1997), is overexpressed in a significant percentage of human cancers and is amplified during the transition between primary tumors and metastasis. The various cancers in which c-MET overexpression is implicated include, but are not limited to, gastric adenocarcinoma, renal cancer, small cell lung carcinoma, colorectal cancer, prostate cancer, brain cancer, liver cancer, pancreatic cancer, and breast cancer. c-MET is also implicated in atherosclerosis and lung fibrosis. Accordingly, there is a great need to develop compounds useful as inhibitors of c-MET protein kinase receptor.
SUMMARY OF THE INVENTION
[0003] It has been found that compounds of this invention, and pharmaceutically acceptable compositions thereof, are effective as inhibitors of c-MET. Accordingly, the invention features compounds having the formula:
Figure imgf000003_0001
or a pharmaceutically acceptable salt thereof, wherein Ring A, ring D, Z1, Z2, R3, and Q are as defined below.
[0004] The invention also provides pharmaceutical compositions that include a compound of formula I and a pharmaceutically acceptable carrier, adjuvant, or vehicle. In addition, the invention provides methods of treating or lessening the severity of a proliferative disease, condition, or disorder in a patient that includes the step of administering to the patient a therapeutically effective dose of a compound of formula I, or a pharmaceutical composition thereof.
DETAILED DESCRIPTION OF THE INVENTION Definitions and General Terminology
[0005] As used herein, the following definitions shall apply unless otherwise indicated. For purposes of this invention, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, and the Handbook of Chemistry and Physics, 75th Ed. 1994. Additionally, general principles of organic chemistry are described in "Organic Chemistry," Thomas Sorrell, University Science Books, Sausalito: 1999, and "March's Advanced Organic Chemistry," 5th Ed., Smith, M.B. and March, J., eds. John Wiley & Sons, New York: 2001, the entire contents of which are hereby incorporated by reference.
[0006] As described herein, compounds of the invention may optionally be substituted with one or more substituents, such as are illustrated generally above, or as exemplified by particular classes, subclasses, and species of the invention. It will be appreciated that the phrase "optionally substituted" is used interchangeably with the phrase "substituted or unsubstituted." In general, the term "substituted," whether preceded by the term "optionally" or not, refers to the replacement of one or more hydrogen radicals in a given structure with the radical of a specified substituent. Unless otherwise indicated, an optionally substituted group may have a substituent at each substitutable position of the group. When more than one position in a given structure can be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at each position. [0007] As described herein, when the term "optionally substituted" precedes a list, said term refers to all of the subsequent substitutable groups in that list. For example, if X is halogen; optionally substituted Ci-3 alkyl or phenyl; X may be either optionally substituted alkyl or optionally substituted phenyl. Likewise, if the term "optionally substituted" follows a list, said term also refers to all of the substitutable groups in the prior list unless otherwise indicated. For example: if X is halogen, Ci -3 alkyl, or phenyl, wherein X is optionally substituted by Jx, then both C1-3 alkyl and phenyl may be optionally substituted by Jx. As is apparent to one having ordinary skill in the art, groups such as H, halogen, NO2, CN, NH2, OH, or OCF3 would not be included because they are not substitutable groups. If a substituent radical or structure is not identified or defined as "optionally substituted," the substituent radical or structure is unsubstituted.
[0008] Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds. The term "stable," as used herein, refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, preferably, their recovery, purification, and use for one or more of the purposes disclosed herein. In some embodiments, a stable compound or chemically feasible compound is one that is not substantially altered when kept at a temperature of 40°C or less, in the absence of moisture or other chemically reactive conditions, for at least a week.
[0009] The term "aliphatic" or "aliphatic group," as used herein, means a straight-chain (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation. Unless otherwise specified, aliphatic groups contain 1-20 carbon atoms. In some embodiments, aliphatic groups contain 1-10 carbon atoms. In other embodiments, aliphatic groups contain 1-8 carbon atoms. In still other embodiments, aliphatic groups contain 1-6 carbon atoms, and in yet other embodiments, aliphatic groups contain 1-4 carbon atoms. Suitable aliphatic groups include, but are not limited to, linear or branched, substituted or unsubstituted alkyl, alkenyl, or alkynyl groups. Further examples of aliphatic groups include methyl, ethyl, propyl, butyl, isopropyl, isobutyl, vinyl, and sec-butyl. The terms "alkyl" and the prefix "alk-," as used herein, are inclusive of both straight chain and branched saturated carbon chain. The term "alkylene," as used herein, represents a saturated divalent straight or branched chain hydrocarbon group and is exemplified by methylene, ethylene, isopropylene and the like. The term "alkylidene," as used herein, represents a divalent straight chain alkyl linking group. The term "alkenyl," as used herein, represents monovalent straight or branched chain hydrocarbon group containing one or more carbon-carbon double bonds. The term "alkynyl," as used herein, represents a monovalent straight or branched chain hydrocarbon group containing one or more carbon-carbon triple bonds.
[0010] The term "cycloaliphatic" (or "carbocycle") refers to a monocyclic C3-C8 hydrocarbon or bicyclic C8-Ci2 hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule, and wherein any individual ring in said bicyclic ring system has 3-7 members. Suitable cycloaliphatic groups include, but are not limited to, cycloalkyl, cycloalkenyl, and cycloalkynyl. Further examples of aliphatic groups include cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, and cycloheptenyl. [0011] The term "heterocycle," "heterocyclyl," "heterocycloaliphatic," or "heterocyclic" as used herein refers to a monocyclic, bicyclic, or tricyclic ring system in which at least one ring in the system contains one or more heteroatoms, which is the same or different, and that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, and that has a single point of attachment to the rest of the molecule. In some embodiments, the "heterocycle," "heterocyclyl," "heterocycloaliphatic," or "heterocyclic" group has three to fourteen ring members in which one or more ring members is a heteroatom independently selected from oxygen, sulfur, nitrogen, or phosphorus, and each ring in the system contains 3 to 8 ring members.
[0012] Examples of heterocyclic rings include, but are not limited to, the following monocycles: tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, tetrahydrothiophen-2-yl, tetrahydrothiophen-3-yl, 2-morpholino, 3-morpholino, 4-morpholino, 2-thiomorpholiπo, 3- thiomorpholino, 4-thiomorpholino, pyrrolidin-1-yl, pyrrolidin-2-yl, pyrrolidin-3-yl, tetrahydropiperazin-1-yl, tetrahydropiperazin-2-yl, tetrahydropiperazin-3-yl, piperidin-1-yl, piperidm-2-yl, piperidin-3-yl, piperidin-4-yl, pyrazolin-1-yl, pyrazolin-3-yl, pyrazolin-4-yl, pyrazolin-5-yl, thiazolidin-2-yl, thiazolidin-3-yl, thiazolidin-4-yl, thiazolidin-5-yl, imidazolidin-1 -yl, ϊmidazolidin-2-yl, imidazolidin-4-yl, imidazolidin-5-yl; and the following bicycles: 3-li/-benzimidazol-2-one, 3-(l-alkyl)-benzimidazol-2-one, indolinyl, tetrahydroquinolinyl, tetrahydroisoquinoliπyl, benzothiolane, benzodithiane, and 1,3- dihydro-imidazol-2-one.
[0013] The term "heteroatom" means one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon, including any. oxidized form of nitrogen, sulfur, or phosphorus; the quaternized form of any basic nitrogen; or a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or NR+ (as in N- substituted pyrrolidinyl).
[0014] The term "unsaturated," as used herein, means that a moiety has one or more units of unsaturation.
[0015] The term "alkoxy," or "thioalkyl," as used herein, refers to an alkyl group, as previously defined, attached to the principal carbon chain through an oxygen ("alkoxy") or sulfur ("thioalkyl") atom.
[0016] The terms "haloalkyl," "haloalkenyl," and "haloalkoxy" mean alkyl, alkenyl, or alkoxy, as the case may be, substituted with one or more halogen atoms. The term "halogen" means F, Cl, Br, or I.
[0017] The term "aryl" used alone or as part of a larger moiety as in "aralkyl,"
"aralkoxy," or "aryloxyalkyl," refers to monocyclic, bicyclic, and tricyclic carbocyclic ring systems having a total of six to fourteen ring members, wherein at least one ring in the system is aromatic, wherein each ring in the system contains 3 to 7 ring members and that has a single point of attachment to the rest of the molecule. The term "aryl" may be used interchangeably with the term "aryl ring." Examples of aryl rings would include phenyl, naphthyl, and anthracene.
[0018] The term "heteroaryl," used alone or as part of a larger moiety as in
"heteroaralkyl," or "heteroarylalkoxy," refers to monocyclic, bicyclic, and tricyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic, at least one ring in the system contains one or more heteroatoms, wherein each ring in the system contains 3 to 7 ring members and that has a single point of attachment to the rest of the molecule. The term "heteroaryl" may be used interchangeably with the term
"heteroaryl ring" or the term "heteroaromatic." Further examples of heteroaryl rings include the following monocycles: furanyl (e.g., furan-2-yl or furan-3-yl); imidazolyl (e.g., N- imidazolyl, imidazol-2-yl, imidazol-4-yl, or imidazol-5-yl); isoxazolyl (e.g., isoxazol-3-yl, isoxazol-4-yl, isoxazol-5-yl); oxazolyl (e.g., oxazol-2-yl, oxazol-4-yl, or oxazol-5-yl); pyrrolyl (e.g., N-pyrrolyl, pyrrol-2-yl, or pyrrol-3-yl); pyridinyl (e.g., pyrid-2-yl, pyrid-3-yl, or pyrid-4-yl); pyrimidinyl (e.g., pyrimidin-2-yl, pyrimidin-4-yl, or pyrimidin-5-yl); pyridazinyl (e.g., pyridazin-3-yl, pyridazin-4-yl, pyridazin-5-yl, or pyridazin-6-yl); thiazolyl (e.g., thiazol-2-yl, thiazol-4-yl, or thiazol-5-yl); tetrazolyl (e.g., tetrazol-1-yl or tetrazol-5-yl); triazolyl (e.g., 2-triazolyl or 5-triazolyl), thienyl (e.g., thiophen-2-yl or thioρhen-3-yl); pyrazolyl (e.g., pyrazol-2-yl, pyrazol-3-yl, or pyrazol-4-yl); isothiazolyl; 1,2,3-oxadiazolyl; 1,2,5-oxadϊazolyl; 1,2,4-oxadiazolyl; 1,2,3-triazolyl; 1,2,3-thiadiazolyl; 1 ,3,4-thiadiazolyl; 1,2,5-thiadiazolyl; pyrazinyl; 1,3,5-triazinyl; and the following bicycles: benzimidazolyl; benzofuryl; benzothienyl; indolyl (e.g., 2-indolyl); purinyl; quinolinyl (e.g., 2-quinolinyl, 3-quinolinyl, or 4-quinolinyl); and isoquinolinyl (e.g., 1-isoquinolinyl, 3-isoquinolinyl, or 4- isoquinolinyl).
[0019] In some embodiments, an aryl (including aralkyl, aralkoxy, aryloxyalkyl and the like) or heteroaryl (including heteroaralkyl and heteroarylalkoxy and the like) group may contain one or more substituents. Suitable substituents on the unsaturated carbon atom of an aryl or heteroaryl group are selected from those listed in the definition of R1, R2, R3, R4, JM, JQ, or JR below. Other suitable substituents include: halogen; -R°; -OR°; -SR°; 1,2- methylenedioxy; 1,2-ethylenedioxy; phenyl (Ph) optionally substituted with R°; -O(Ph) optionally substituted with R°; -(CH2) I-2(Ph), optionally substituted with R0; -CH=CH(Ph), optionally substituted with R°; -NO2; -CN; -N(R°)2; -NR0C(O)R0; -NR0C(S)R0; -NR°C(O)N(R°)2; -NR°C(S)N(R°)2; -NR0CO2R0; -NR0NR0C(O)R0; -NR°NR°C(O)N(R°)2; -NR0NR0CO2R0; -C(O)C(O)R0; -C(O)CH2C(O)R0; -CO2R0; -C(O)R0; -C(S)R0; -C(O)N(R°)2; -C(S)N(R°)2; -OC(O)N(R°)2; -OC(O)R0; -C(O)N(OR0) R°; -C(NOR0) R°; -S(O)2R0; -S(O)2OR0; -S(O)2N(R°)2; -S(O)R0; -NRoS(0)2N(R0)2; -NR0S(O)2R0; -N(OR°)R°; -C(=NH)-N(R°)2; -(CH2)O-2NHC(O)R0; -L-R°; -L-N(R°)2; -L-SR°; -L-OR°; -L-(C3-10 cycloaliphatic), -L-(Ce- io aryl), -L-(5-10 membered heteroaryl), -L-(5-10 membered heterocyclyl), oxo, Ci^haloalkoxy, Ci-4haloalkyl, -L-NO2, -L-CN, -L-OH, -L-CF3; or two substituents, together with the intervening atoms to which they are bound, form a 5-7 membered saturated, unsaturated, or partially saturated ring, wherein L is a Ci-βalkylene group in which up to three methylene units are replaced by -NH-, -NR0-, -O-, -S-, -C(O)O-, -OC(O)-, -C(O)CO-, -C(O)-, -C(O)NH-, -C(O)NR0-, -C(=N-CN), -NHCO-, -NR0CO-, -NHC(O)O-, -NR0C(O)O-, -S(O)2NH-, -S(O)2NR0-, -NHS(O)2-, -NR0S(O)2-, -NHC(O)NH- , -NR0C(O)NH-, -NHC(O)NR0-, -NR0C(O)NR0, -OC(O)NH-, -OC(O)NR0-, -NHS(O)2NH-, -NR0S(O)2NH-, -NHS(O)2NR0-, -NR0S(O)2NR0-, -S(O)-, or -S(O)2-, and wherein each independent occurrence of R° is selected from hydrogen, optionally substituted Ci-6 aliphatic, an unsubstituted 5-8 membered heteroaryl or heterocyclic ring, phenyl, -O(Ph), or -CH2(Ph), or, two independent occurrences of R°, on the same substituent or different substituents, taken together with the atom(s) to which each R° group is bound, form a 5-8-membered heterocyclyl, aryl, or heteroaryl ring or a 3-8-membered cycloalkyl ring, wherein said heteroaryl or heterocyclyl ring has 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Optional substituents on the aliphatic group of R° are selected from NH2, NH(Ci ^aliphatic), N^Maliphatich, halogen, CMaliphatic, OH, O(Ci.4aliphatic), NO2, CN, CO2H, CO2(C (^aliphatic), 0(1IaIoCi-4 aliphatic), or haloC]_4aliphatic, wherein each of the foregoing CMaliphatic groups of R° is unsubstituted.
[0020] In some embodiments, an aliphatic, cycloaliphatic, heteroaliphatic group, or a non-aromatic heterocyclic ring may contain one or more substituents. In some instances two substituents, on the same atom or on different atoms, together with the intervening atoms to which they are bound, form a 5-7 membered saturated, unsaturated, or partially saturated ring containing 0-3 heteroatoms selected from N, O, or S. Suitable substituents on the saturated carbon of an aliphatic or heteroaliphatic group, or of a non-aromatic heterocyclic ring are selected from those listed above for the unsaturated carbon of an aryl or heteroaryl group and additionally include the following: =O, =S, =NNHR*, =NN(R*)2, =NNHC(O)R*, =NNHCO2(alkyl), =NNHS(O)2 (alkyl), or =NR*, where each R* is independently selected from hydrogen or an optionally substituted Ci-6 aliphatic, or two R* on the same nitrogen are taken together with the nitrogen to form a 5-8 membered heterocyclyl or heteroaryl ring having 1 -3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. Optional substituents on the aliphatic group of R* are selected from NH2, NH(C(-4 aliphatic), N(Ci-4 aliphatic)2, halogen, Ci-4 aliphatic, OH, 0(Ci-4 aliphatic), NO2, CN, CO2H, CO2(C M aliphatic), O(halo C1.4 aliphatic), or halo(Ci_4 aliphatic), wherein each of the foregoing Cj. 4aliphatic groups of R is unsubstituted.
[0021] In some embodiments, optional substituents on the nitrogen of a non-aromatic heterocyclic ring include -R+, -N(R+)2, -C(O)R+, -C(O)OR+, -C(O)C(O)R+, -C(O)CH2C(O)R+, -S(O)2R+, -S(O)2N(R+)2, -C(=S)N(R+)2, -C(=NH)-N(R+)2, or -NR+S(O)2R+; wherein R+ is hydrogen, an optionally substituted Cj-6 aliphatic, optionally substituted phenyl, optionally substituted -(D(Ph), optionally substituted -CH2(Ph), optionally substituted -(CH2)i-2(Ph); optionally substituted -CH=CH(Ph); or an unsubstituted 5-6 membered heteroaryl or heterocyclic ring having one to four heteroatoms independently selected from oxygen, nitrogen, or sulfur, or two independent occurrences of R+, on the same substituent or different substituents, taken together with the atom(s) to which each R+ group is bound, form a phenyl, 5-8-membered heterocyclyl, 5-8-membered heteroaryl, or a 3-8 membered cycloalkyl ring, wherein said heteroaryl or heterocyclyl ring has 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Optional substituents on the aliphatic group or the phenyl ring of R+ are selected from -NH2, -NH(C 1.4 aliphatic), -N(Ci-4 aliphatic)2, halogen, Ci-4 aliphatic, -OH, -0(Ci-4 aliphatic), -NO2, -CN, -C(O)OH, - C(O)O(CM aliphatic), -O(halo(Ci-4 aliphatic)), or halo(Ci-4 aliphatic), wherein each of the foregoing Ci ^aliphatic groups of R+ is unsubstituted.
10022] As detailed above, in some embodiments, two independent occurrences of R0 (or R+, or any other variable similarly defined herein), may be taken together with the atom(s) to which each variable is bound to form a phenyl, 5-8-membered heterocyclyl, 5-8-membered heteroaryl, or a 3-8 membered cycloalkyl ring. Exemplary rings that are formed when two independent occurrences of R° (or R+, or any other variable similarly defined herein) are taken together with the atom(s) to which each variable is bound include, but are not limited to the following: a) two independent occurrences of R° (or R+, or any other variable similarly defined herein) that are bound to the same atom and are taken together with that atom to form a ring, for example, N(R°)2, where both occurrences of R° are taken together with the nitrogen atom to form a piperidin-1-yl, piperazin-1-yl, or morpholin-4-yl group; and b) two independent occurrences of R° (or R , or any other variable similarly defined herein) that are bound to different atoms and are taken together with both of those atoms to form a ring, for
example where a phenyl group is substituted with two occurrences of OR
Figure imgf000009_0001
, these two occurrences of R° are taken together with the oxygen atoms to which they are
bound to form a fused 6-membered oxygen containing ring:
Figure imgf000009_0002
It will be appreciated that a variety of other rings can be formed when two independent occurrences of R0 (or R+, or any other variable similarly defined herein) are taken together with the atom(s) to which each variable is bound and that the examples detailed above are not intended to be limiting.
[0023] In some embodiments, a methylene unit of the alkyl or aliphatic chain is optionally replaced with another atom or group. Examples of such atoms or groups would include, but are not limited to, -NR0-, -O-, -S-, -C(O)O-, -OC(O)-, -C(O)CO-, -C(O)-, -C(O)NR0-, -C(=N-CN), -NR0CO-, -NR0C(O)O-, -S(O)2NR0-, -NR0S(O)2-, -NR0C(O)NR0-, -OC(O)NR0-, -NR0S(O)2NR0-, -S(O)-, or -S(O)2-, wherein R° is defined herein. Unless otherwise specified, the optional replacements form a chemically stable compound. Optional atom or group replacements can occur both within the chain and at either end of the chain; i.e. both at the point of attachment and/or also at the terminal end. Two optional replacements can also be adjacent to each other within a chain so long as it results in a chemically stable compound. Unless otherwise specified, if the replacement occurs at the terminal end, the replacement atom is bound to an H on the terminal end. For example, if one methylene unit Of-CH2CH2CHa was optionally replaced with -O-, the resulting compound could be -OCH2CH3, -CH2OCH3, or -CH2CH2OH.
[0024] As described herein, a bond drawn from a substituent to the center of one ring within a multiple-ring system (as shown below) represents substitution of the substituent at any substitutable position in any of the rings within the multiple ring system. For example, Figure a represents possible substitution in any of the positions shown in Figure b.
Figure imgf000010_0001
Figure a Figure b
[0025] This also applies to multiple ring systems fused to optional ring systems (which would be represented by dotted lines). For example, in Figure c, X is an optional substituent both for ring A and ring B.
Figure imgf000010_0002
Figure c [0026] If, however, two rings in a multiple ring system each have different substituents drawn from the center of each ring, then, unless otherwise specified, each substituent only represents substitution on the ring to which it is attached. For example, in Figure d, Y is an optionally substituent for ring A only, and X is an optional substituent for ring B only.
Figure imgf000011_0001
Figure d
[0027] The term "protecting group," as used herein, represent those groups intended to protect a functional group, such as, for example, an alcohol, amine, carboxyl, carbonyl, etc., against undesirable reactions during synthetic procedures. Commonly used protecting groups are disclosed in Greene and Wuts, Protective Groups in Organic Synthesis, 3rd Edition (John Wiley & Sons, New York, 1999), which is incorporated herein by reference. Examples of nitrogen protecting groups include acyl, aroyl, or carbamyl groups such as formyl, acetyl, propionyl, pivaloyl, t-butyl acetyl, 2-chloroacetyl, 2-bromoacetyl, trifluoroacetyl, trichloro acetyl, phthalyl, o-nitrophenoxyacetyl, α-chlorobutyryl, benzoyl, A- chlorobenzoyl, 4-bromobenzoyl, 4-nitrobenzoyl and chiral auxiliaries such as protected or unprotected D, L or D, L-amino acids such as alanine, leucine, phenylalanine and the like; sulfonyl groups such as benzenesulfonyl, p-toluenesulfonyl and the like; carbamate groups such as benzyloxycarbonyl, p-chlorobenzyloxycarbonyl, p-methoxybenzyloxycarbonyl, p- nitrobenzyloxycarbonyl, 2-nitrobenzyloxycarbonyl, p-bromobenzyloxycarbonyl, 3,4- dimethoxybenzyloxycarbonyl, "3,5-dimethoxybenzyloxycarbonyl, 2,4- dimethoxybenzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, 2-nitro-4,5- dimethoxybenzyloxycarbonyi, 3,4,5-trimethoxybenzyloxycarbonyl, 1 -(p-biphenylyl)- 1 - methyl ethoxycarbonyl, α,α-dimethyl-3,5-dimethoxybenzyloxycarbonyl, benzhydryloxycarbonyl, t-butyloxycarbonyl, diisopropylmethoxycarbonyl, isopropyloxycarbonyl, ethoxycarbonyl, methoxycarbonyl, allyloxycarbonyl, 2,2,2,- trichloroethoxycarbonyl, phenoxy carbonyl, 4-nitrophenoxy carbonyl, fluorenyl-9- methoxycarbonyl, cyclopentyloxycarbonyl, adamantyloxycarbonyl, cyclohexyloxycarbonyl, phenylthiocarbonyl and the like, arylalkyl groups such as benzyl, triphenylmethyl, benzyloxymethyl and the like and silyl groups such as trimethylsilyl and the like. Preferred N-protecting groups are formyl, acetyl, benzoyl, pivaloyl, t-butylacetyl, alanyl, phenylsulfonyl, benzyl, t-butyloxycarbonyl (Boc) and benzyloxycarbonyl (Cbz). [0028] The term "prodrug," as used herein, represents a compound that is transformed in vivo into a compound of formula I, I- A, I-B, I-C, I-D, or I-E, or a compound listed in Tables 1-5. Such a transformation can be affected, for example, by hydrolysis in blood or enzymatic transformation of the prodrug form to the parent form in blood or tissue. Prodrugs of the compounds of the invention may be, for example, esters. Esters that may be utilized as prodrugs in the present invention are phenyl esters, aliphatic (C1-C24) esters, acyloxymethyl esters, carbonates, carbamates, and amino acid esters. For example, a compound of the invention that contains an OH group may be acylated at this position in its prodrug form. Other prodrug forms include phosphates, such as, for example those phosphates resulting from the phosphorylation of an OH group on the parent compound. A thorough discussion of prodrugs is provided in T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems, Vol. 14 of the A.C.S. Symposium Series, Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, and Judkins et al, Synthetic Communications 26(23):4351-4367, 1996, each of which is incorporated herein by reference.
[0029] Unless otherwise stated, structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the (R) and (S) configurations for each asymmetric center, (Z) and (E) double bond isomers, and (Z) and (E) conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. [0030] Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention. Additionally, unless otherwise stated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures except for the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope of this invention. Such compounds are useful, for example, as analytical tools or probes in biological assays, or as c- MET inhibitors with improved therapeutic profile. Description of Compounds of the Invention
[0031] In a first aspect, the invention features a compound having the formula:
Figure imgf000013_0001
O), or a pharmaceutically acceptable salt thereof, wherein: Z1 is N or CR4; Z2 is N or CH; Ring D is the selected from:
Figure imgf000013_0002
Ring B is a 5- or 6- membered aryl, cycloaliphatic, heteroaryl, or heterocyclyl ring, wherein said ring is optionally substituted with up to 4 occurrences of RB and said heteroaryl or heterocyclyl ring contains up to three heteroatoms selected from N, O, or S; each RB is independently selected from halogen, RB 1, -CN, -CO2R81, -OC(O)R81,
-OC(O)N(R81), -NO2, -N(RB1)2, -NC(O)R81, -N(RB1)C(O)N(RB1)25 -SR81, -S(O)2R81, -S(O)2N(R8 ')2, or -S(O)R81, wherein each RB1 is, independently, hydrogen or Ci-4 aliphatic, or two RBI together with the atom to which they are bound, form a 3-6 membered carbocycle optionally substituted with 0-2 occurrences of JR or a 3-6 membered heterocyclyl containing 1 -3 heteroatoms independently selected from N, O, or S and optionally substituted with 0-2 occurrences of JR on carbon and optionally substituted with JN on each substitutable ring nitrogen atom;
Q is C6-Io aryl or 5-10 membered heteroaryl wherein each Q is optionally substituted with up to 5 occurrences of JQ;
U is N or CR1;
V is N or CR2;
U1 is O, NR5, or S;
VI is O, NR6, or S; R1 is hydrogen, halogen, -CN, -NH2, -OH, C1-2 haloalkyl, or selected from -NH(CM aliphatic), -N(CM aliphatic)2, C3-4 cycloalkyl, -(Cj-2 aliphatic)-(C3^ cycloalkyl), or Ci-4 aliphatic, each of which is optionally substituted with up to 2 occurrences of JR;
R2 is hydrogen, halogen, -CN, -NH2, -NH(CM aliphatic), -N(CM aliphatic)2, C1-2 haloalkyl, C3.4 cycloalkyl, or CM aliphatic;
R3 is halogen or RA, wherein RA is Cβ-io aryl, 5-10 membered heteroaryl, 5-12 membered heterocyclyl, or C3-S cycloaliphatic, each of which is optionally substituted with 0-3 occurrences of JM;
R4 is hydrogen, -CN, CM aliphatic, halogen or Ci-2 haloalkyl; each of R5, R6, and R7, is, independently, hydrogen or JN; each JM is independently selected from halogen, -NO2, -CN, CM aliphatic, Ci-2 haloalkyl, -(CH2)0-2CH(R')2, -OH, -OR', -(CR'"2)qNH2, -(CR"'2)qNHR\ -(CR"'2)qN(R')2, -(CR'"2)qNHS(O)2R', -(CR'"2)qNHC(O)R\ -(CR'"2)qNHC(O)OR\ -(CR'"2)qNHC(O)NH2, -(CR"'2)qNHC(O)NHR\ -(CR'"2)qNHC(O)N(R')2, -(CR" '2)qNHC(NH)NH2, -(CR" '2)qNHC(NH)NHR', -(CR" '2)qNHC(NR)N(R')2, -(CR'"2)qNHS(O)2NH2, -(CR'"2)qNHS(O)2NHR', -(CR'"2)qNHS(O)2N(R')2, -SH, -SR', -(CR"'2)qCO2H, -(CR'"2)qCO2R\ -C(O)H, -(CR'"2)qC(O)R\ -(CR" '2 )q-C(O)-(CH2)0- 2CH(R')2, -(CR' ' '2)q-C(0)-(CH2)o.2NHCH(R')2, -(CR' "2)q-C(O)-(CH2)0-2NR'CH(R')2, -(CR" '2)q-C(O)NH2, -(CR'"2)q-C(O)NHR'5 -(CR'"2)q-C(O)N(R')2, -(CR' ' '2)q-C(O)N(OH)R' , -(CR' ' '2)q-C(O)N(OR')R\ -(CR' ' '2)q-C(O)N(OR')H, -(CR' ' '2)q-C(O)N(OH)H, -(CR' ' '2)q-C(=NOH)R', -(CR' ' '2)q-C(=NOR')H, -(CR'"2)q-C(NOR')R', -(CR'"2)q-S(O)2R', -(CR'"2)q-S(O)2OH, -(CR'"2)q-S(O)2OR', -(CR" '2)q-S(O)2NH2, -(CR" '2)q-S(O)2NHR', -(CR"'2VS(O)2N(R')2. -(CR'"2)q-S(O)R', -(CR" '2)q-C(=NR')-NH2, -(CR" '2)q-C(=NR')-NHR', -(CR" '2VC(=NR')-N(R')2, -(CR'"2)q-C(=NH)-NH2, -(CR'"2)q-C(=NH)-NHR', -(CR'"2)q-C(=NH)-N(R')2, C6-io aryl, 5-10 membered heteroaryl, 5-10 membered heterocyclyl, or C3-S cycloaliphatic, wherein q is selected from 0-4; or two JM, together with the atom or atoms to which they are bound, form a 3-6 membered cycloaliphatic or heterocyclyl ring; wherein each of said cycloaliphatic or heterocyclyl is optionally substituted with up to 3 occurrences of JN or
JR; each JN is independently selected from
Figure imgf000014_0001
aliphatic, -(CR5"2)q'C3-6 cycloalkyl, -(CR' ' '2 Vphenyl, -(CR' ' '2)q>C(O)C , ^aliphatic; -(CR' ' '2)q.C(O)C l-2haloalkyl; -C(O)O(CMalkyl), -(CR" '2)q-C(O)NH2> -(CR"'2)q'C(O)NH(C^aliphatic), -(CR'"2)q'C(O)N(Ci-4aliphatic)2, or -S(O)2CMaliphatic5 wherein q' is 0-2 and each aliphatic or cycloaliphatic is optionally substituted with up to 2 occurrences of JR; each J^ is independently selected from halogen, -NO2, -CN, C1.4 aliphatic, CM haloalkyl, -OH, -OR", -NH2, -NHR", -N(R")2, -SH, -SR", -CO2H, -CO2R", -C(O)H, -C(O)R", -C(O)NH2, -C(O)NHR", -C(O)N(R")2, -C(O)N(OH)R", -C(O)N(OR")R", -C(O)N(OR")H, -C(O)N(OH)H, -C(NOH)R", -C(NOR")H, -C(NOR")R", -S(O)2R", -S(O)2OH, -S(O)2OR", -S(O)2NH2, -S(O)2NHR", -S(O)2N(R")2, -S(O)R", -C(-NR')-NH2, -C(=NR')-NHR', -C(=NR')-N(R')2, -C(=NH)-NH2, -C(=NH)-NHR", -C(=NH)-N(R")2, C6-i0aryl, 5-10 membered heteroaryl, 5-10 membered heterocyclyl, or C3-8 cycloaliphatic; each JR is independently selected from halogen, -NO2, -CN, Ci-4 aliphatic, C1-4 haloalkyl, C3-4 cycloalkyl, -OH, -NH2, -0(C]-4 aliphatic), -N(Ci-4 aliphatic)2, or -NH(Ci-4 aliphatic); each R' is independently selected from unsubstituted Ci-6 aliphatic; or two R' groups, together with the atom(s) to which they are bound, form a 3-6 membered cycloaliphatic or heterocyclyl, each optionally substituted with up to 2 occurrences of JR; each R" is independently selected from unsubstituted Ci-6 aliphatic; or two R" groups, together with the atom to which they are bound, form a 3-6 membered heterocyclyl, optionally substituted with up to 2 occurrences of JR; and each R'" is independently selected from hydrogen or Ci-4 aliphatic, or an R'" group and an R' group, together with the atoms to which they are bound, form a 3-6 membered cycloaliphatic or heterocyclyl, each optionally substituted with up to 2 occurrences of JR.
[0032] In one embodiment,
Z1 is N or CR4;
Z2 is N or CH;
Ring D is the selected from one of the 5-membered rings shown below:
Figure imgf000015_0001
each of R5 and R6 is hydrogen, Ci-2 haloalkyl, or selected from C3-4 cycloalkyl, -(C i-2 aliphatic)-(C3-4 cycloalkyl), or C]-4 aliphatic, each of which is optionally substituted with up to 2 occurrences of JR;
R3 is a Cβ-io aryl, 5-10 membered heteroaryl, 5-10 membered heterocyclyl, or C3-S cycloaliphatic, each of which is optionally substituted with 0-3 occurrences of JM; each JQ is, independently, selected from halogen, -NO2, -CN, Ci-4 aliphatic, Ci-4 haloalkyl, -OH, -OR", -NH2, -NHR", -N(R")2, -SH, -SR", -CO2H, -CO2R", -C(O)H, -C(O)R", -C(O)NH2, -C(O)NHR", -C(O)N(R")2, -C(O)N(OH)R", -C(0)N(OR")R", -C(O)N(OR")H, -C(O)N(OH)H, -C(NOH)R", -C(NOR")H, -C(NOR")R", -S(O)2R", -S(O)2OH, -S(O)2OR", -S(O)2NH2, -S(O)2NHR", -S(O)2N(R")2, -S(O)R", -C(=NR')- NH2, -C(=NR')-NHR', -C(=NR')-N(R')2, -C(=NH)-NH2, -C(=NH)-NHR", -C(=NH)- N(R")2, C6.joaryl, 5-10 membered heteroaryl, 5-10 membered heterocyclyl, or C3-8 cycloaliphatic; each JR is, independently, selected from halogen, -NO2, -CN, Ci-4 aliphatic, CM haloalkyl, C3-4 cycloalkyl, -OH, -NH2, -0(Ci-2 aliphatic), -N(Ci-2 aliphatic)2, or -NH(Ci-2 aliphatic); each JL is independently selected from halogen, -NO2, -CN, Q-4 aliphatic, Ci-4 haloalkyl, -OH, -NH2, -0(Ci-2 aliphatic), -N(Ci-2 aliphatic)2, or -NH(Ci-2 aliphatic); and each JM is, independently, selected from halogen, -NO2, -CN, Ci-4 aliphatic, Ci.2 haloalkyl, -OH, -OR', -(CR'"2)qNH2, -(CR'"2)qNHR', -(CR'"2)qN(R')2, -(CR"'2)qNS(O)2R\ -(CR'"2)qNHC(O)R', -(CR'"2)qNHC(O)OR', -(CR'"2)qNHC(O)NH2, -(CR" '2)qNHC(O)NHR' , -(CR' ' '2)qNHC(O)N(R')2, -(CR' ' '2)qNHS(O)2NH2 -(CR"'2)qNHS(O)2NHR\ -(CR'"2)qNHS(O)2N(R')2, -SH, -SR', -CO2H, -CO2R', -C(O)H, -C(O)R', -C(O)NH2, -C(O)NHR', -C(O)N(R')2, -C(O)N(OH)R', -C(O)N(OR')R', -C(O)N(OR')H, -C(O)N(OH)H, -C(=NOH)R\ -C(=NOR')H, -C(NOR')R', -S(O)2R', -S(O)2OH, -S(O)2OR', -S(O)2NH2, -S(O)2NHR', -S(O)2N(R')2, -S(O)R', -C(=NR')-NH2, -C(=NR')-NHR', -C(=NR')-N(R')2, -C(=NH)-NH2, -C(=NH)- NHR', -C(=NH)-N(R')2, C6-ιo aryl, 5-10 membered heteroaryl, 5-10 membered heterocyclyl, or C3-S cycloaliphatic, wherein q is selected from 0-4; each R' is, independently, selected from unsubstituted C1-6 aliphatic; or two R' groups, together with the atom to which they are bound, form a 3-6 membered heterocyclyl, optionally substituted with 0-2 occurrences of JR; each R" is, independently, selected from unsubstituted Cj.6 aliphatic; or two R" groups, together with the atom to which they are bound, form a 3-6 membered heterocyclyl, optionally substituted with 0-2 occurrences of JR; and each R'" is, independently, selected from hydrogen or Ci-4 aliphatic.
[0033] In one embodiment for compounds of the invention, Ring D is selected from:
Figure imgf000017_0001
[0034] In another embodiment, Ring D is selected from:
Figure imgf000017_0002
[0035J In a further embodiment, Ring D is
Figure imgf000017_0003
[0036] In an alternative embodiment, Ring D is selected from
Figure imgf000018_0001
[0037] In another embodiment, Ring D is selected from
Figure imgf000018_0002
[0038] In another embodiment, Ring D is selected from
Figure imgf000018_0003
[0039] In a further embodiment, Ring D is
Figure imgf000018_0004
[0040] In one embodiment, Ring A is selected from
Figure imgf000018_0005
[0041] In a further embodiment, Ring A is
Figure imgf000018_0006
[0042] Compounds of the invention include those of formulae II or III:
Figure imgf000019_0001
[0043] In one embodiment for compounds of the invention, R4 is hydrogen. [0044] In one embodiment for compounds of the invention, Q is
Figure imgf000019_0002
.
[0045] In a further embodiment, Q is
Figure imgf000019_0003
[0046] In yet a further embodiment, each J^ is, independently, fluoro or chloro, such as, for example, when Q is
Figure imgf000019_0004
[0047] In one embodiment for compounds of the invention, R3 is RA, which is a C6-ιo aryl, a C3-8 cycloaliphatic, or a monocyclic or bicyclic 5-10 membered heteroaryl or heterocyclyl containing 1-4 heteroatoms independently selected from N, O, or S, wherein said aryl, cycloaliphatic, heteroaryl, or heterocyclyl is optionally substituted with up to 3 occurrences of JM.
[0048] In another embodiment, RA is phenyl optionally substituted with up to 3 occurrences of JM. In another embodiment, RA is a 5-6 membered heteroaryl optionally substituted with up to 3 occurrences of JM, such as, for example, an optionally substituted pyridyl, thienyl, thiazolyl, isothiazolyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, furanyl, isoxazolyl, or oxazolyl. Further examples include lH-pyrazol-4-yl substituted at the 1- position with JM, thiophen-2-yl substituted at the 5-position with JM, thiophen-3-yl substituted at the 5-position with JM, furan-2-yl substituted at the 5-position with JM, furan-3- yl substituted at the 5-position with JM, l//-pyrrol-3-yl substituted at the 1 -position with JM, lH-l,2,3-triazol-4-yl substituted at the 1-position with JM, or thiazol-5-yl substituted at the 2- position with JM.
[0049] Examples of JM include those where JM is selected from phenyl, 5-8 membered heteroaryl, 5-10 membered heterocyclyl, or C3.scycloaliphatic; each optionally substituted with up to 3 occurrences of JN or JR. hi a further example, JM is an optionally substituted 5-
10 membered heterocyclyl containing 1 or 2 nitrogen atoms, such as, for example an optionally substituted piperidine, piperazine or pyrrolidine or an optionally substituted bicyclooctane or bicyclononane containing 1 or 2 nitrogen atoms.
[0050] In another embodiment, RA is a Cs-io bicyclic heteroaryl selected from:
Figure imgf000020_0001
or wherein
Ring E is a 5-membered heteroaryl ring with 1 to 2 heteroatoms selected from N, O, or S; and JE is hydrogen or JN.
[0051] In a further embodiment, Ring E is selected from thienyl, thiazolyl, pyrrolyl, imidazolyl, furanyl, or oxazolyl.
[0052] In another embodiment, RA is
Figure imgf000020_0002
[0053] In another embodiment,
Figure imgf000020_0003
from
Figure imgf000020_0004
wherein JF is selected from Ci-4 aliphatic, -C0-2aliphaticCΗ(R')2, -(CR" '2)qNH25 -(CR"'2)qNHR\ -(CR'"2)qN(R')2, -(CR'"2)qNS(O)2R', -(CR'"2)qNHC(O)R', -(CR'"2)qNHC(O)OR', -(CR'"2)qNHC(O)NH2, -(CR'"2)qNHC(O)NHR', -(CR' ' '2)qNHC(O)N(R')2, -(CR' ' '2)qNHC(NH)NH2, -(CR' ' '2)qNHC(NH)NHR' , -(CR' ' '2)qNHC(NH)N(R')2, -(CR' ' '2)qNHS(O)2NH2, -(CR' ' '2)qNHS(O)2NHR\ -(CR'"2)qNHS(O)2N(R')2, -CO2R', -C(O)H, -C(O)R', -C(O)-(CH2)0-2CH(R')2, -C(O)NH2, -C(O)NHR', -C(O)N(R')2, -S(O)2R', -S(O)2NH2, -S(O)2NHR', -S(O)2N(R')2, C6-10 aryl, 5-10 membered heteroaryl, 5-10 membered heterocyclyl, or C3.8 cycloaliphatic, wherein q is selected from 0-4 and said aryl, heteroaryl, heterocyclyl, or cycloaliphatic of JF is optionally substituted with halogen, -NO2, -CN, Ci-4 aliphatic, Ci-4 haloalkyl, C3-4 cycloalkyl, -OH, -NH2, -0(Ci-2 aliphatic), -N(Ci-2 aliphatic)2, or -NH(Ci-2 aliphatic). [0054] In a fiαrther embodiment, RA is
Figure imgf000021_0001
, wherein
JF is -C0-2aliphaticCH(R')2, -(CR'"2)qNH2, -(CR'"2)qNHR'5 -(CR'"2)qN(R')2j -C(O)R', -C(0)-(CH2)o-2CH(R')2, or optionally substituted heterocyclyl.
[0055] For any of the compounds of the invention, JM is selected from halogen, -NO2, -CN, C1-4 aliphatic, C1-2 haloalkyl, -OH, -OR', -(CR'"2)qNH2, -(CR"'2)qNHR\ -(CR'"2)qN(R')2, -(CR'"2)qNS(O)2R', -(CR"'2)qNHC(O)R\ -(CR"'2)qNHC(O)OR\ -(CR' ' '2)qNHC(O)NH2, -(CR' ' '2)qNHC(O)NHR\ -(CR' ' '2)qNHC(O)N(R')2, -(CR'"2)qNHS(O)2NH2, -(CR'"2)qNHS(O)2NHR', -(CR'"2)qNHS(O)2N(R')2, -SH, -SR', -CO2H, -CO2R', -C(O)H, -C(O)R', -C(O)NH2, -C(O)NHR', -C(O)N(R')2, -C(O)N(OH)R', -C(O)N(OR')R\ -C(O)N(OR')H, -C(O)N(OH)H, -C(=NOH)R', -C(=NOR')H, -C(NOR')R', -S(O)2R', -S(O)2OR', -S(O)2NH2, -S(O)2NHR', -S(O)2N(R')2, -S(O)R', -C(=NR')-NH2, -C(=NR')-NHR', -C(=NR')-N(R')2, -C(=NH)-NH2, -C(=NH)-NHR', or -C(=NH)-N(R')2. [0056] In another embodiment, JM is selected from
Figure imgf000021_0002
[0057] In another embodiment, q is 1 or 2.
[0058] In yet another embodiment, JM is not substituted on a ring position adjacent to
Ring A.
[0059] In one embodiment for compounds of the invention, wherein R3 is halogen, compounds of formula I are useful as intermediates for preparing compounds of formula I wherein R3 is RΛ.
[0060] In yet another embodiment for compounds of the invention, a substituted or unsubstituted CM or C,-6 aliphatic group of R1, R2, R4, R5, R6, JM, JN, JQ, JR, R', R", or R'" comprises two or more non-hydrogen atoms.
[0061] In another aspect, the invention features a compound having the formula:
Figure imgf000022_0001
I-A I B I C I-D or I E, wherein said compound is selected from the compounds of Tables 1, 2, 3, 4, or 5, respectively.
Table 1. Compounds of Formula 1-A
Figure imgf000022_0002
I A l I-A-2 I-A-3
Figure imgf000023_0001
I-A-7 I-A-8 I-A-9
Figure imgf000023_0002
I-A-10 I-A-l 1 I-A-12
Figure imgf000024_0001
I- A-13 I-A-14 I-A-15
Figure imgf000024_0002
I-A-16 I-A-17 I-A-18
Figure imgf000024_0003
Figure imgf000025_0001
I-A-25 I-A-26 I-A-27
Figure imgf000025_0002
Figure imgf000026_0001
Figure imgf000026_0002
Figure imgf000026_0003
I-A-37 I-A-38 I-A-39
Figure imgf000027_0001
I-A-40 I-A-41 I-A-42
Figure imgf000027_0002
I-A-43 I-A-44 I-A-45
Figure imgf000027_0003
Figure imgf000028_0001
Figure imgf000028_0002
I-A-55 I-A-56 I-A-57
Figure imgf000029_0001
I-A-58 ϊ-A-59 I-A-60
Figure imgf000029_0002
I-A-61 I-A-62 I-A-63
Figure imgf000029_0003
I-A-64 I-A-65 I-A-66
Figure imgf000030_0001
Figure imgf000030_0002
I-A-73 I-A-74 I-A-75
Figure imgf000031_0001
Figure imgf000031_0002
I-A-79 I-A-80 I-A-81
Figure imgf000031_0003
I-A-82 I-A-83 I-A-84
Figure imgf000032_0001
Figure imgf000032_0002
Figure imgf000032_0003
I-A-91 I-A-92 I-A-93
Figure imgf000033_0001
I-A-94 I-A-95 I-A-96
Figure imgf000033_0002
Figure imgf000034_0001
Figure imgf000034_0002
Figure imgf000034_0003
I-A-109 I-A-110 I- A-111
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000036_0002
I-A-124 I-A-125 I-A-126
Figure imgf000036_0003
I-A-127 I-A-128 I-A-129
Figure imgf000037_0001
Figure imgf000037_0002
I-A-133 I-A-134 I- A-135
Figure imgf000037_0003
I-A-136 I-A-137 I-A-138
Figure imgf000038_0001
I-A-142 I-A-143 I-A-144
Figure imgf000038_0002
Figure imgf000039_0001
I-A-148 I-A-149 I-A-150
Figure imgf000039_0002
Figure imgf000039_0003
Figure imgf000040_0001
I-A-160 I-A-161 I- A-162
Figure imgf000040_0002
Figure imgf000041_0001
I-A-166 I-A-167 I-A-168
Figure imgf000041_0002
Figure imgf000041_0003
Figure imgf000042_0001
Figure imgf000042_0002
I-A-178 I-A-179 I-A-l 80
Figure imgf000042_0003
Figure imgf000043_0001
I-A-184 I- A-185 I-A-186
Figure imgf000043_0002
Figure imgf000043_0003
Figure imgf000045_0001
I-A-208 I-A-209 I-A-210
Figure imgf000046_0001
I-A-211 I-A-212 I-A-213
Figure imgf000046_0002
Figure imgf000046_0003
Figure imgf000047_0001
Figure imgf000047_0002
Figure imgf000047_0003
Figure imgf000048_0001
Figure imgf000048_0002
Figure imgf000048_0003
Figure imgf000049_0001
Figure imgf000049_0002
Figure imgf000049_0003
Figure imgf000050_0001
Figure imgf000051_0001
I-A-262 I-A-263 I-A-264
Figure imgf000051_0002
Figure imgf000052_0001
I-A-271 I-A-272 I-A-273
Figure imgf000052_0002
I-A-274 I-A-275 I-A-276
Figure imgf000052_0003
I-A-277 I-A-278 I-A-279
Figure imgf000053_0001
Figure imgf000053_0002
Figure imgf000053_0003
Figure imgf000054_0001
I-A-289 I-A-290 I-A-291
Figure imgf000054_0002
Figure imgf000054_0003
Figure imgf000055_0001
Figure imgf000055_0002
Figure imgf000055_0003
Figure imgf000056_0001
Figure imgf000056_0002
Figure imgf000056_0003
Figure imgf000057_0001
Figure imgf000057_0002
I-A322 I-A-323 I-A-324
Figure imgf000057_0003
Figure imgf000058_0001
Figure imgf000058_0002
I-A-331 I-A-332 I-A-333
Figure imgf000058_0003
Figure imgf000059_0001
I-A-337 I-A-338 I-A-339
Figure imgf000059_0002
Figure imgf000059_0003
I-A-343 I-A-344 I-A-345
Figure imgf000060_0001
Figure imgf000060_0002
I-A-352 I-A-353 I-A-354
Figure imgf000061_0001
I-A-358 l-A-359 I-A-360
Figure imgf000061_0002
I-A-361 I-A-362 l-A-363
Figure imgf000062_0001
Figure imgf000062_0002
Figure imgf000063_0001
I-A-373 T-A-374 I-A-375
Figure imgf000063_0002
Figure imgf000063_0003
I-A-379 I-A-380 I-A-381
Figure imgf000064_0001
Figure imgf000064_0002
Figure imgf000065_0001
I-A-391 I-A-392 I-A-393
Figure imgf000065_0002
Figure imgf000066_0001
I-A-403 I-A-404 I-A-405
Figure imgf000066_0002
I-A-406 I-A-407 I-A-408
Figure imgf000067_0001
I-A-409 I-A-410 I-A-411
Figure imgf000067_0002
Figure imgf000067_0003
Figure imgf000068_0001
I-A-418 I-A-419 I-A-420
Figure imgf000068_0002
Figure imgf000068_0003
Figure imgf000069_0001
Figure imgf000069_0002
Figure imgf000069_0003
Figure imgf000070_0001
I-A-439 I-A-440 I-A-441
Figure imgf000070_0002
I-A-442 I-A-443 I-A-444
Figure imgf000071_0001
I-A-445 I-A-446 I-A-447
Figure imgf000071_0002
Figure imgf000071_0003
Figure imgf000072_0001
Figure imgf000072_0002
Figure imgf000073_0001
Figure imgf000073_0002
I-A-466 I-A-467 I-A-468
Figure imgf000073_0003
Figure imgf000074_0001
Figure imgf000074_0002
Figure imgf000075_0001
Figure imgf000075_0002
Figure imgf000075_0003
Figure imgf000076_0001
Figure imgf000076_0002
I-A-493 I-A-494 I-A-495
Figure imgf000076_0003
I-A-499 I-A-500 I-A-501
Figure imgf000077_0001
I-A-505 I-A-506 I-A-507
Figure imgf000077_0002
I-A-508 I-A-509 I-A-510
Figure imgf000078_0001
Figure imgf000078_0002
Figure imgf000078_0003
I-A-517 I-A-518 I-A-519
Figure imgf000079_0001
Figure imgf000079_0002
I-A-523 I-A-524 I-A-525
Figure imgf000079_0003
I-A-526 I-A-527 T-A-528
Figure imgf000080_0001
I-A-529 T-A-530 I-A-531
Figure imgf000080_0002
Figure imgf000080_0003
Figure imgf000081_0001
Figure imgf000081_0002
Figure imgf000081_0003
I-A-544 I-A-545 I-A-546
Figure imgf000082_0001
Figure imgf000082_0002
I-A-550 I-A-551 I-A-552
Figure imgf000082_0003
Figure imgf000082_0004
Figure imgf000083_0001
Figure imgf000083_0002
I-A-562 I-A-563 I-A-564
Figure imgf000083_0003
I-A-565 I-A-566 l-A-567
Figure imgf000083_0004
I-A-568 I-A-569 I-A-570
Figure imgf000084_0001
I-A-571 I-A-572 I-A-573
Figure imgf000084_0002
I-A-574 I-A-575 I-A-576
Figure imgf000084_0003
Figure imgf000085_0001
Figure imgf000085_0002
I-A-583 I-A-584 I-A-585
Figure imgf000085_0003
I-A-586 I-A-587 I-A-588
Figure imgf000086_0001
I-A-589 I-A-590 I-A-591
Figure imgf000086_0002
I-A-592 I-A-593 I-A-594
Figure imgf000086_0003
I-A-595 I-A-596 I-A-597
Figure imgf000087_0001
Figure imgf000087_0002
I-A-601 I-A-602 I-A-603
Figure imgf000087_0003
I-A-604 I-A-605 I-A-606
Figure imgf000088_0001
Figure imgf000088_0002
I-A-610 I-A-611 I-A-612
Figure imgf000088_0003
I-A-613 I-A-614 I-A-615
Figure imgf000089_0001
Figure imgf000089_0002
I-A-619 I-A-620 I-A-621
Figure imgf000089_0003
Figure imgf000090_0001
Figure imgf000090_0002
Figure imgf000090_0003
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000092_0002
Figure imgf000092_0003
Figure imgf000093_0001
Figure imgf000093_0002
I-A-655 I-A-656 I-A-657
Figure imgf000093_0003
I-A-658 I-A-659 I-A-660
Figure imgf000094_0001
Figure imgf000094_0002
Figure imgf000094_0003
Figure imgf000095_0001
Figure imgf000095_0002
I-A-673 I-A-674 I-A-675
Figure imgf000095_0003
T-A-676 I-A-677 I-A-678
Figure imgf000096_0001
I-A-679 I-A-680 I-A-681
Figure imgf000096_0002
I-A-682 I-A-683 I-A-684
Figure imgf000096_0003
I-A-685 I-A-686 I-A-687
Figure imgf000097_0001
Figure imgf000097_0002
I-A-691 I-A-692 I-A-693
Figure imgf000097_0003
Figure imgf000098_0001
I-A-697 I-A-698 I-A-699
Figure imgf000098_0002
I-A-700 I-A-701 I-A-702
Figure imgf000098_0003
I-A-703 I-A-704 I-A-705
Figure imgf000099_0001
I-A-706
Table 2. Compounds of Formula I-B
Figure imgf000099_0002
I-B-l I-B-2 I-B-3
Figure imgf000099_0003
I-B-4 I-B-5 I-B-6
Figure imgf000100_0001
I-B-10 I-B-l 1 I-B-12
Figure imgf000100_0002
Figure imgf000101_0001
IBl 6 IBl 7 I-B-18
Figure imgf000101_0002
Table 3. Compounds of Formula I-C
Figure imgf000102_0001
Figure imgf000102_0002
I-C-4 I-C-5 I-C-6
Figure imgf000102_0003
Figure imgf000103_0001
I-C-l 0 I-C-l 1
Table 4. Compounds of Formula I-D
Figure imgf000103_0002
I-D-l I-D-2 I-D-3
Figure imgf000104_0001
I-D-4 I-D-5 I-D-6
Figure imgf000104_0002
I-D-10 ID l 1 I-D-12
Figure imgf000105_0001
Figure imgf000106_0001
I-D-22 I-D-23
Table 5. Compounds of Formula I-E
Figure imgf000106_0002
Figure imgf000106_0003
I-E-4 I-E-5 Compositions, Formulations, and Administration of Compounds of the Invention [0062] In another aspect, the invention provides a pharmaceutical composition comprising a compound of any of the formulae or classes described herein. In a further embodiment, the invention provides a pharmaceutical composition comprising a compound of Tables 1, 2, 3, 4, or 5. hi a further embodiment, the composition additionally comprises an additional therapeutic agent.
[0063] According to another embodiment, the invention provides a composition comprising a compound of this invention or a pharmaceutically acceptable derivative thereof and a pharmaceutically acceptable carrier, adjuvant, or vehicle. In one embodiment, the amount of compound in a composition of this invention is such that is effective to measurably inhibit c-MET in a biological sample or in a patient. Preferably the composition of this invention is formulated for administration to a patient in need of such composition. Most preferably, the composition of this invention is formulated for oral administration to a patient.
[0064] The term "patient", as used herein, means an animal, preferably a mammal, and most preferably a human. \
[0065] It will also be appreciated that certain of the compounds of present invention can exist in free form for treatment, or where appropriate, as a pharmaceutically acceptable derivative thereof. According to the present invention, a pharmaceutically acceptable derivative includes, but is not limited to, pharmaceutically acceptable prodrugs, salts, esters, salts of such esters, or any other adduct or derivative which upon administration to a patient in need is capable of providing, directly or indirectly, a compound as otherwise described herein, or a metabolite or residue thereof.
[0066] As used herein, the term "pharmaceutically acceptable salt" refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like.
[0067] Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et al., describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 66:1-19, 1977, which is incorporated herein by reference. Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like. Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N+(Ci-4 alkyl)4 salts. This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein. Water or oil-soluble or dispersable products may be obtained by such quaternization. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, C i-8 sulfonate and aryl sulfonate.
[0068] As described above, the pharmaceutically acceptable compositions of the present invention additionally comprise a pharmaceutically acceptable carrier, adjuvant, or vehicle, which, as used herein, includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired. In Remington: The Science and Practice of Pharmacy, 21st edition, 2005, ed. D.B. Troy, Lippincott Williams & Wilkins, Philadelphia, and Encyclopedia of Pharmaceutical Technology, eds. J. Swarbrick and J. C. Boylan, 1988-1999, Marcel Dekker, New York, the contents of each of which is incorporated by reference herein, are disclosed various carriers used in formulating pharmaceutically acceptable compositions and known techniques for the preparation thereof. Except insofar as any conventional carrier medium is incompatible with the compounds of the invention, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutically acceptable composition, its use is contemplated to be within the scope of this invention.
[0069] Some examples of materials which can serve as pharmaceutically acceptable carriers include, but are not .limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, or potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, polyacrylates, waxes, polyethylene- polyoxypropylene-block polymers, wool fat, sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil; saffiower oil; sesame oil; olive oil; corn oil and soybean oil; glycols; such a propylene glycol or polyethylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator.
[0070] The compositions of the present invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. The term "parenteral" as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intraocular, intrahepatic, intralesional and intracranial injection or infusion techniques. Preferably, the compositions are administered orally, intraperitoneally or intravenously. Sterile injectable forms of the compositions of this invention may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1 ,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium.
[0071 J For this purpose, any bland fixed oil may be employed including synthetic mono- or diglycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions. Other commonly used surfactants, such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation. [0072] The pharmaceutically acceptable compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions. In the case of tablets for oral use, carriers commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried cornstarch. When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.
[0073] Alternatively, the pharmaceutically acceptable compositions of this invention may be administered in the form of suppositories for rectal administration. These can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug. Such materials include cocoa butter, beeswax and polyethylene glycols. [0074] The pharmaceutically acceptable compositions of this invention may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations are readily prepared for each of these areas or organs.
{0075] Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically- transdermal patches may also be used.
[0076] For topical applications, the pharmaceutically acceptable compositions may be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers. Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water. Alternatively, the pharmaceutically acceptable compositions can be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
[0077] For ophthalmic use, the pharmaceutically acceptable compositions may be formulated, e.g., as micronized suspensions in isotonic, pH adjusted sterile saline or other aqueous solution, or, preferably, as solutions in isotonic, pH adjusted sterile saline or other aqueous solution, either with or without a preservative such as benzylalkonium chloride. Alternatively, for ophthalmic uses, the pharmaceutically acceptable compositions may be formulated in an ointment such as petrolatum. The pharmaceutically acceptable compositions of this invention may also be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluoro carbons, and/or other conventional solubilizing or dispersing agents.
[0078] Most preferably, the pharmaceutically acceptable compositions of this invention are formulated for oral administration.
[0079] Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3 -butyl ene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
[0080] Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S. P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables.
[0081] The injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
[0082] In order to prolong the effect of a compound of the present invention, it is often desirable to slow the absorption of the compound from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the compound then depends upon its rate of dissolution that, in turn, may depend upon crystal size and crystalline form. Alternatively, dissolving or suspending the compound in an oil vehicle accomplishes delayed absorption of a parenterally administered compound form. Injectable depot forms are made by forming microencapsule matrices of the compound in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of compound to polymer and the nature of the particular polymer employed, the rate of compound release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly( anhydrides). Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues.
[0083] Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound. •
[0084] Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents. [0085] Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polethylene glycols and the like. [0086] The active compounds can also be in micro-encapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes.
[0087] Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required. Ophthalmic formulation, eardrops, and eye drops are also contemplated as being within the scope of this invention. Additionally, the present invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel. [0088] The compounds of the invention are preferably formulated in dosage unit form for ease of administration and uniformity of dosage. The expression "dosage unit form" as used herein refers to a physically discrete unit of agent appropriate for the patient to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific effective dose level for any particular patient or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts. [0089] The amount of the compounds of the present invention that may be combined with the carrier materials to produce a composition in a single dosage form will vary depending upon the host treated, the particular mode of administration. Preferably, the compositions should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of the inhibitor can be administered to a patient receiving these compositions. [0090] Depending upon the particular condition, or disease, to be treated or prevented, additional therapeutic agents, which are normally administered to treat or prevent that condition, may also be present in the compositions of this invention. As used herein, additional therapeutic agents that are normally administered to treat or prevent a particular disease, or condition, are known as "appropriate for the disease, or condition, being treated." Examples of additional therapeutic agents are provided infra.
[0091] The amount of additional therapeutic agent present in the compositions of this invention will be no more than the amount that would normally be administered in a composition comprising that therapeutic agent as the only active agent. Preferably the amount of additional therapeutic agent in the presently disclosed compositions will range from about 50% to 100% of the amount normally present in a composition comprising that agent as the only therapeutically active agent.
Uses of the Compounds and Compositions of the Invention
[0092] According to one embodiment, the invention relates to a method of inhibiting c- MET protein kinase activity in a biological sample comprising the step of contacting said biological sample with a compound of this invention, or a composition comprising said compound. The term "biological sample," as used herein, means a sample outside a living organism and includes, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof. Inhibition of kinase activity in a biological sample is useful for a variety of purposes known to one of skill in the art. Examples of such purposes include, but are not limited to, biological specimen storage and biological assays. In one embodiment, the method of inhibiting kinase activity in a biological sample is limited to non- therapeutic methods.
[0093] The teπn "c-MET" is synonymous with "c-Met," "cMet", "MET", "Met" or other designations known to one skilled in the art.
[0094] According to another embodiment, the invention relates to a method of inhibiting c-MET kinase activity in a patient comprising the step of administering to said patient a compound of the present invention, or a composition comprising said compound. [0095] The term "c-MET-mediated disease" or "c-MET-mediated condition", as used herein, means any disease state or other deleterious condition in which c-MET is known to play a role. The terms "c-MET-mediated disease" or "c-MET-mediated condition" also mean those diseases or conditions that are alleviated by treatment with a c-MET inhibitor. Such conditions include, without limitation, renal, gastric, colon, brain, breast, prostate, and lung cancer, glioblastoma, atherosclerosis, lung fibrosis, conditions associated with organ transplantation, allergic disorders, and autoimmune disorders.
[0096] In one aspect, the present invention features a method treating a proliferative disorder in a patient comprising the step of administering to the patient a therapeutically effective dose of any of the compounds or compositions of the invention. [0097] According to one embodiment, the proliferative disorder is cancer, such as, for example, renal, gastric, colon, brain, breast, liver, prostate, and lung cancer, or a glioblastoma.
[0098] In another embodiment, the present invention relates to a method of treating or lessening the severity of brain cancer in a patient in need thereof, comprising administering to said patient a compound of the present invention or composition thereof. [0099] In another embodiment, the proliferative disorder is polycythemia vera, essential thrombocythemia, chronic idiopathic myelofibrosis, myeloid metaplasia with myelofibrosis, chronic myeloid leukemia (CML), chronic myelomonocytic leukemia, chronic eosinophilic leukemia, hypereosinophilic syndrome, systematic mast cell disease, atypical CML, or juvenile myelomonocytic leukemia.
[00100] In another embodiment, the proliferative disorder is atherosclerosis or lung fibrosis. [00101] Another aspect of the present invention relates to a method of inhibiting tumor metastasis in a patient in need thereof, comprising administering to said patient a compound of the present invention or composition thereof.
[00102] Depending upon the particular condition, or disease, to be treated, additional therapeutic agents, which are normally administered to treat that condition, may also be present in the compositions of this invention. As used herein, additional therapeutic agents that are normally administered to treat a particular disease, or condition, are known as "appropriate for the disease, or condition, being treated".
[00103] In one embodiment, chemotherapeutic agents or other antiproliferative agents maybe combined with the compounds of this invention to treat proliferative diseases and cancer. Examples of known chemotherapeutic agents include, but are not limited to, alkylating agents, such as, for example, cyclophosphamide, lomustine, busulfan procarbazine, ifosfamide, altretamine, melphalan, estramustine phosphate, hexamethylmelamine, mechlorethamine, thiotepa, streptozocin, chlorambucil, temozolomide, dacarbazine, semustine, or carmustine; platinum agents, such as, for example, cisplatin, carboplatinum, oxaliplatin, ZD-0473 (AnorMED), spiroplatinum, lobaplatin (Aeterna), carboxyphthalatoplatinum, satraplatin (Johnson Matthey), tetraplatin BBR-3464, (Hoffinann- La Roche), ormiplatin, SM-11355 (Sumitomo), iproplatin, or AP-5280 (Access); antimetabolites, such as, for example, azacytidine, tomudex, gemcitabine, trimetrexate, capecitabine, deoxycoformycin, 5-fluorouracil, fludarabine, floxuridine, pentostatin, 2- chlorodeoxyadenosine, raltitrexed, 6-mercaptopurine, hydroxyurea, 6-thioguanine, decitabine (SuperGen), cytarabin, clofarabine (B ioen vision), 2-fluorodeoxy cytidine, irofulven (MGI Pharma), methotrexate, DMDC (Hoffmann-La Roche), idatrexate, or ethynylcytidine (Taiho); topoisomerase inhibitors, such as, for example, amsacrine, rubitecan (SuperGen), epirubicin, exatecan mesylate (Daiichi), etoposide, quinamed (ChemGenex), teniposide, mitoxantrone, gimatecan (Sigma-Tau), irinotecan (CPT-1 1), diflomotecan (Beaufour-Ipsen), 7-ethyl-lO-hydroxy-camptothecin, TAS- 103 (Taiho), topotecan, elsamitrucin (Spectrum), dexrazoxanet (TopoTarget), J-107088 (Merck & Co), pixantrone (Novuspharma), BNP-1350 (BioNumerik), rebeccamycin analogue (Exelixis), CKD-602 (Chong Kun Dang), BBR-3576 (Novuspharma), or KW-2170 (Kyowa Hakko); antitumor antibiotics, such as, for example, dactinomycin (actinomycin D), amonafide, doxorubicin (adriamycin), azonafide, deoxyrubicin, anthrapyrazole, valrubicin, oxantrazole, daunorubicin (daunomycin), losoxantrone, epirubicin, bleomycin, sulfate (blenoxane), therarubicin, bleomycinic acid, idarubicin, bleomycin A, rubidazone, bleomycin B, plicamycinp, mitomycin C, porfiromycin, MEN- 10755 (Menarini), cyanomoφholinodoxorubicin, GPX-100 (Gem Pharmaceuticals), or mitoxantrone (novantrone), antimitotic agents, such as, for example, paclitaxel, SB 408075 (GlaxoSmithKline), docetaxel, E7010 (Abbott), colchicines, PG-TXL (Cell Therapeutics), vinblastine, IDN 5109 (Bayer), vincristine A, 105972 (Abbott), vinorelbine, A 204197 (Abbott), vindesine, LU 223651 (BASF), dolastatin 10 (NCI), D 24851 (ASTAMedica), rhizoxin (Fujisawa), ER-86526 (Eisai), mivobulin (Warner-Lambert), combretastatin A4 (BMS), cemadotin (BASF), isohomohalichondrm-B (PharmaMar), RPR 109881 A (Aventis), ZD 6126 (AstraZeneca), TXD 258 (Aventis), PEG-paclitaxel (Enzon,) epothilone B (Novartis), AZ10992 (Asahi), T 900607 (Tularik), IDN-5109 (Indena), T 138067 (Tularik), AVLB (Prescient NeuroPharma), cryptophycin 52 (Eli Lilly), azaepothilone B (BMS), vinflunine (Fabre), BNP-7787 (BioNumerik), auristatin PE (Teikoku Hormone), CA-4 prodrug (OXiGENE), BMS 247550 (BMS), dolastatin-10 (NIH), BMS 184476 (BMS)3 CA-4 (OXiGENE), BMS 188797 (BMS), or taxoprexin (Protarga); aromatase inhibitors, such as, for example, aminoglutethimide, exemestane, letrozole, atamestane (BioMedicines), anastrazole, YM-511 (Yamanouchi), or formestane; thymidylate synthase inhibitors, such as, for example, pemetrexed (EH Lilly), nolatrexed (Eximias), ZD- 9331 (BTG), or CoFactor™ (BioKeys); DNA antagonists, such as, for example, trabectedin (PharmaMar), mafosfamide (Baxter International), glufosfamide (Baxter International), apaziquone (Spectrum Pharmaceuticals), albumin + 32P (Isotope Solutions), O6 benzyl guanine (Paligent), thymectacin (NewBiotics), or edotreotide (Novartis); farnesyltransferase inhibitors, such as, for example, arglabin (NuOncology Labs), tipifarnib (Johnson & Johnson), lonafarnib (Schering-Plough), perillyl alcohol (DOR BioPharma), or BAY-43- 9006 (Bayer); Pump inhibitors, such as, for example, CBT-I (CBA Pharma), zosuquidar trihydrochloride (Eli Lilly), tariquidar (Xenova), biricodar dicitrate (Vertex), or MS-209 (Schering AG); Histone acetyltransferase inhbitors, such as, for example, tacedinaline (Pfizer), pivaloyloxymethyl butyrate (Titan), SAHA (Aton Pharma), depsipeptide (Fujisawa), or MS-275 (Schering AG); Metalloproteinase inhibitors, such as, for example, Neovastat (Aeterna Laboratories), CMT-3 (CollaGenex), marimastat (British Biotech), or BMS-275291 (Celltech); ribonucleoside reductase inhibitors, such as, for example, gallium maltolate (Titan), tezacitabine (Aventis), triapine (Vion), or didox (Molecules for Health); TNF alpha agonists/antagonists, such as, for example, virulizin (Lorus Therapeutics), revimid (Celgene), CDC-394 (Celgene), entanercept (Immunex Corp.)* infliximab (Centocor, Inc.), or adalimumab (Abbott Laboratories); endothelin A receptor antagonists, such as, for example, atrasentan (Abbott) YM-598 (Yamanouchi) or ZD-4054 (AstraZeneca); retinoic acid receptor agonists, such as, for example, fenretinide (Johnson & Johnson) alitretinoin (Ligand) or LGD-1550 (Ligand); immuno- modulators, such as, for example, interferon dexosome therapy (Anosys), oncophage (Antigenics), pentrix (Australian Cancer Technology), GMK (Progenies), ISF- 154 (Tragen), adenocarcinoma vaccine (Biomira), cancer vaccine (Intercell), CTP-37 (AVI BioPharma), norelin (Biostar), IRX-2 (Immuno-Rx), BLP-25 (Biomira), PEP -005 (Peplin Biotech), MGV (Progenies), synchrovax vaccines (CTL Immuno), beta-alethine (Dovetail), melanoma vaccine (CTL Immuno), CLL therapy (Vasogen), orp21 RAS vaccine (GemVax); hormonal and antihormonal agents, such as, for example, estrogens, prednisone, conjugated estrogens, methylprednisolone, ethinyl estradiol, prednisolone, chlortrianisen, aminoglutethimide, idenestrol, leuprolide, hydroxyprogesterone caproate, goserelin, medroxyprogesterone, leuporelin, testosterone, bicalutamide, testosterone propionate, fluoxymesterone, flutamide, methyltestosterone, octreotide, diethylstilbestrol, nilutamide, megestrol, mitotane, tamoxifen, P-04 (Novogen), toremofine, 2-methoxyestradiol (EntreMed), dexamethasone, or arzoxifene (Eli Lilly); photodynamic agents, such as, for example, talaporfϊn (Light Sciences), Pd-bacteriopheophorbide (Yeda), Theralux (Theratechnologies), lutetium texaphyrin (Pharmacyclics), motexafin gadolinium (Pharmacyclics), or hypericin; and tyrosine kinase inhibitors, such as, for example, imatinib (Novartis), kahalide F (PharmaMar), leflunomide (Sugen/Phaπnacia), CEP-701 (Cephalon), ZDl 839 (AstraZeneca), CEP-751 (Cephalon), erlotinib (Oncogene Science), MLN518 (Millenium), canertinib (Pfizer), PKC412 (Novartis), squalamine (Genaera), phenoxodiol, SU5416 (Pharmacia), trastuzumab (Genentech), SU6668 (Pharmacia), C225 (ImClone), ZD4190 (AstraZeneca), rhu-Mab (Genentech), ZD6474 (AstraZeneca), MDX-H210 (Medarex), vatalanib (Novartis), 2C4 (Genentech), PKIl 66 (Novartis), MDX-447 (Medarex), GW2016 (GlaxoSmithKline), ABX-EGF (Abgenix), EKB-509 (Wyeth), IMC-ICl 1 (ImClone), or EKB-569 (Wyeth).
[00104] Those additional agents may be administered separately from the compound- containing composition, as part of a multiple dosage regimen. Alternatively, those agents may be part of a single dosage form, mixed together with the compound of this invention in a single composition. If administered as part of a multiple dosage regime, the two active agents maybe submitted simultaneously, sequentially or within a period of time from one another normally within five hours from one another.
[00105] The amount of both, the compound and the additional therapeutic agent (in those compositions which comprise an additional therapeutic agent as described above)) that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. Preferably, the compositions of this invention should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of a compound of formula I can be administered.
[00106] In those compositions that comprise an additional therapeutic agent, that additional therapeutic agent and the compound of this invention may act synergistically. Therefore, the amount of additional therapeutic agent in such compositions will be less than that required in a monotherapy utilizing only that therapeutic agent. In such compositions a dosage of between 0.01 - 100 mg/kg body weight/day of the additional therapeutic agent can be administered.
[00107] The amount of additional therapeutic agent present in the compositions of this invention will be no more than the amount that would normally be administered in a composition comprising that therapeutic agent as the only active agent. Preferably the amount of additional therapeutic agent in the presently disclosed compositions will range from about 50% to 100% of the amount normally present in a composition comprising that agent as the only therapeutically active agent.
[00108] The compounds of this invention, or pharmaceutical compositions thereof, may also be incorporated into compositions for coating an implantable medical device, such as prostheses, artificial valves, vascular grafts, stents and catheters. Vascular stents, for example, have been used to overcome restenosis (re-narrowing of the vessel wall after injury). However, patients using stents or other implantable devices risk clot formation or platelet activation. These unwanted effects may be prevented or mitigated by pre-coating the device with a pharmaceutically acceptable composition comprising a kinase inhibitor. Suitable coatings and the general preparation of coated implantable devices are described in US Patents 6,099,562; 5,886,026; and 5,304,121. The coatings are typically biocompatible polymeric materials such as a hydrogel polymer, polymethyldisiloxane, polycaprolactone, polyethylene glycol, polylactic acid, ethylene vinyl acetate, and mixtures thereof. The coatings may optionally be farther covered by a suitable topcoat of fluorosilicone, polysaccarides, polyethylene glycol, phospholipids or combinations thereof to impart controlled release characteristics in the composition. Implantable devices coated with a compound of this invention are another embodiment of the present invention. [00109] In order that the invention described herein may be more fully understood, the following examples are set forth. It should be understood that these examples are for illustrative purposes only and are not to be construed as limiting this invention in any manner.
Preparation of Compounds of the Invention
[00110] The following definitions describe terms and abbreviations used herein:
Ala alanine
ATP adenosine triphosphate
Boc t-butoxylcarbonyl
BSA bovine serum albumin
CDI carbonyl diimidazole
DCM dichloromethane
DIEA diisopropylethylamine
DMA dimethylacetamide
DMF dimethylformamide
DMSO methylsulfoxide
DTT dithiothreitol
EDC 1 -ethyl -3-(3 ' -dimethylaminopropyl)carbodiirnide hydrochloride
ES-MS electrospray mass spectrometry
Et2O ethyl ether
EtOAc ethyl acetate
EtOH ethyl alcohol
HBTU 0-(benzotriazol-l-yl)-l,l,3,3-tetramethyluronium hexafluorophosphate
HEPES 4-(2-hydroxyethyl)-l-piperazineethanesulfonic acid
HOBT hydroxy benzotriazole hydrate
HPLC high performance liquid chromatography
J In some structures, "J" is used to represent an iodine atom. Lawesson's
Reagent 2,4-bis(4-methoxyphenyl)- 1 ,3 -dithia-2,4-diphosphetane-2,4-disulfide
LC-MS liquid chromatography-mass spectrometry
LiHMDS lithium hexamethyldisilazide
Me methyl
MeOH methanol
NBS N-bromosuccinimide
NMP N-methylpyrrolidine
PdCl2(dppf) 1 , 1 '-bis(diphenylphosphino)ferrocene]dichloropalladium(II)
Ph phenyl
PyBOP benzotriazol- 1 -yl-oxytripyrrolidinophosphonium hexafluorophosphate
RT room temperature tBu tertiary butyl
TCA trichloroacetic acid
THF tetrahydrofuran
TEA triethylamine
TFA trifluoacetic acid
TsOH /7-toluenesulfonic acid
[00111] As used herein, other abbreviations, symbols and conventions are consistent with those used in the contemporary scientific literature. See, e.g., Janet S. Dodd, ed., The ACS
Style Guide: A Manual for Authors and Editors, 2nd Ed., Washington, D.C.: American
Chemical Society, 1997, herein incorporated in its entirety by reference.
[00112] As used herein, the term "Rt(min)" refers to the HPLC retention time, in minutes, associated with the compound. Unless otherwise indicated, the HPLC method utilized to obtain the reported retention time is as follows: column: Zorbax SB Cl 8 column, 3.0 x 150 mm; gradient: 10-90% acetonitrile/water (0.1%TFA), 5 minutes; flow rate: 1.0 mL/minute; and detection: 254 & 214 nm.
[00113] Purifications by reversed-phase HPLC were conducted on a Waters 20 x 100mm
YMC-P ack Pro Cl 8 column using a linear water/acetonitrile (0.1%TFA) gradient at a flow rate of 28 mL/minute. Beginning and final composition of the gradient varied for each compound between 10-40 and 50-90% acetonitrile, respectively. General Synthetic Procedures
[0001] In general, the compounds of this invention may be prepared by methods described herein or known to those skilled in the art for the preparation of analogous compounds. The following non-limiting schemes and examples are presented to further exemplify the invention. Physiochemical characterization of selected compounds of the invention is provided in Tables 6-10.
[00114] Compounds of the invention can, in general, be prepared as shown in Scheme 1. Accordingly, a compound of formula I-A-a, I-B-a, I-C-a, ϊ-D-a, or I-E-a, or a protected derivative thereof, wherein Z1, Z2, U, V, U1. V1, Q, and Ring B, are as defined for a compound or formula I, is reacted with intermediate RΛ-Metal in a catalyst-mediated cross coupling reaction to form a compound of formula I-A-b, I-B-b, I-C-b, I-D-b, or I-E-b, respectively. RA is as defined elsewhere herein or is a protected derivative thereof. Non- limiting examples of RΛ include optionally substituted Cβ-io aryls, 5-10 membered monocyclic or bicyclic heteroaryls, 5-10 membered monocyclic or bicyclic heterocyclyls, or 5-7 membered cycloaliphatics containing at least one point of unsaturation. The Metal group can be, for example, -B(OAlkyl)2 or -B(OH)2(Suzuki reaction), -Mg-HaI (Kumada reaction), -Zn-HaI (Negishi reaction), -Sn(Alkyl)3 (Stille reaction), -Si(Alkyl)3 (Hiyama reaction), -Cu- HaI, -ZrCp2Cl, or -AlMe2. The catalyst for the cross-coupling reaction can be, for example, a palladium catalyst/ligand system (such as, for example, Pd(PPb.3)4, Pd(PtBu3J4, Pd[P(MeXtBu3XU, PdCl2(PPh3)2, PdCl2(dppf)2, Pd2(dba)3BINAP, or Pd2(dba)3P(o-tol)3 (see Fu and Littke, Angew. Chem. Int. Ed. 41 :4176-4211, 2002; Nicolaou et al., Angew. Chem. Int. Ed. 44:4442-4489, 2005; or Hassen et al., Chemical Reviews 102(5):1359-1469, 2002). The reaction is usually performed in the presence of a base.
Seheme 1
Figure imgf000124_0001
[00115] Alternatively, compound of formula I-a, wherein Z1, Z2, Q and Ring D are as defined for a compound of formula I, can be transformed into a boronate or boronic acid of formula I-b, as shown in Scheme 2. Subsequent reaction with an aryl, heteroaryl, or cycloalkenyl halide produces a compound of formula I-c (a compound of formula I, wherein R3 is RA).
Scheme 2
Figure imgf000124_0002
Synthetic Examples
Example 1. 3-Amino-N-(2,3-difluorophenyl)pyrazine-2-carboxamide
Figure imgf000125_0001
PyBOP. DIEA
Figure imgf000125_0002
[00116] To a solution of S-aminopyrazine^-carboxylic acid (2.0 g, 14.38 mmol) in anhydrous DMF (20 mL) was added 2,3-difluoroaniline (2.2 g, 17.04 mmol) and DIEA (7.6 mL, 43.63 mmol). The mixture was stirred at room temperature while PyBOP (7.5 g, 14.41 mmol) was added. The stirring was continued for another 14 hours until HPLC detected no starting material. The reaction solution was then poured into saturated sodium bicarbonate solution. The crude product was collected by vacuum filtration and washed with water. After drying on vacuum pump for 24 hours, the off-white product (2.9 g, 80%) was used directly in the next reaction without further purification. LC-MS m/e= 250.8 (M+H); 1H- NMR (500 MHZ, DMSO-d6): 10.42 (s, IH), 8.33 (d, IH), 7.94 (d, IH), 7.72 (m, IH), 7.60 (br, 2H), 7.25 (m, 2H). Using the same procedure, 3-amino-iV-(2,3-dichlorophenyl)pyrazine- 2-carboxamide and 3-amino-Vv"-(3-chloro-2-fluorophenyl)pyrazme-2-carboxamide can be produced by reacting 3-aminopyrazine-2-carboxylic acid with 2,3-dichloroaniline and 3- chloro-2-fluoroaniline, respectively. Analogously, this procedure can be used to produce 2- amino-Λf-(2,3-difluorophenyl)pyridine-3-carboxamide, 2-amino-iV"-(2,3- dichlorophenyl)pyridine-3-carboxarnide, and 2-amino--V-(3-chloro-2-fmorophenyl)pyridiine- 3-carboxamide by reacting 2-amino-pyridine-3-carboxylic acid with 2,3-difluoroaniline, 2,3- dichloroaniline, and 3-chloro-2-fluoroaniline, respectively.
Example 2. 3-Amino-N-(2,3-dimαorophenyl)-ΛP-aminopyrazine-2-carboxamidine
1- Lawesson's Reagent 2. hydrazine
Figure imgf000125_0004
Figure imgf000125_0003
[00117] To a solution of 3-amino-N-(2,3-difluorophenyl) pyrazine-2-carboxamide (2.0 g, 8.0 mmol) in anhydrous 1,4-dioxane (50 mL) was added Lawesson's Reagent (2.3 g, 5.7 mmol). The solution was heated at 900C for 14 hours and cooled. The solvent was evaporated under vacuum, the residue was re-dissolved in ethanol (30 mL) and methylene chloride (30 mL), and hydrazine (2 inL) was added at RT. The mixture was then stirred for 3 hours and evaporated. The dark residue was poured into saturated NaHCO3 solution and extracted with ethyl acetate. The combined organic layers were dried over MgSO4, filtered, and removed under vacuum evaporation to give 3-arnino-7V-(2,3-difluoroρhenyl)-iV- aminopyrazine-2-carboxamidine (1.2 g, 57%), which was used directly in the next reaction without further purification. Using the same procedure, 3-amino-N-(2,3-dichlorophenyl)-iV- aminopyrazine-2-carboxamidine, 3-amino-N-(3-chloro-2-fluorophenyl)-iV1-aminopyrazine-2- carboxamidine, 2-amino-7V-(2,3-difluorophenyl)-N'-aminopyridine-3-carboxamidine, 2- amino-N-(2,3-dichlorophenyl)-iV-aminopyridine-3-carboxamidineJ and 2-amino-JV-(3- chloro^-fluorophenyty-ΛP-aminopyridine-S-carboxamidine can be produced from 3-amino- iV-(2,3-dichlorophenyl)pyrazine-2-carboxamide, S-amino-A^S -chloro-2- fluorophenyl)pyrazine-2-carboxamide, 2-amino-7V-(2,3-difluorophenyl)pyridine-3- carboxamide, 2-amino-N-(2,3-dichlorophenyl)pyridine-3-carboxamide, and 2-amino-N-(3- chloro-2-fluorophenyl)pyridine-3-carboxamide, respectively.
Example 3. 3-(4-(2,3-Difluorophenyl)-4H-l,2,4-triazol-3-yl)pyrazin-2-amine
Figure imgf000126_0001
[00118] 3-Amino-JV"-(2,3-difluorophenyl)-iV-aminopvrazine-2-carboxamidine (500 mg, 1.89 mmol) was dissolved in CH(OEt)3 (20 mL) and HCOOH (5 mL) was added slowly at RT. The solution was kept at RT for 30 min and carefully neutralized with saturated NaHCθ3 and 6N NaOH until a pH of 10 was achieved. The resulting mixture was extracted with ethyl acetate. The combined organic layers were dried over MgSθ4 and evaporated to produce 3-(4-(2,3-Difluorophenyl)-4H-l,2,4-triazol-3-yl)pyrazin-2 -amine as yellow solid (480 mg, 92%). A small amount of the crude product was purified by HPLC for characterization and the remainder used directly in the next reaction without further purification. LC-MS m/e= 274.8 (M+H). Using the same procedure, 3-(4-(2,3- dichloroρhenyl)-4//-l,2,4-triazol-3-yl)pyrazin-2-amine, 3-(4-(3-chloro-2-fluorophenyl)-4//- 1 ,2,4-triazol-3-yl)pyrazin-2-amine, 3-(4-(2,3-difluorophenyl)-4/7-l ,2,4-triazol-3-yl)pyridin- 2-amine, 3-(4-(2,3-dichlorophenyl)-4i-r-l,2,4-triazol-3-yl)pyridin-2-amine, and 3-(4-(3- chloro-2-fluorophenyl)-4H-l,2,4-triazol-3-yl)pyridin-2-amine can be produced from 3- amino-N-(2,3-dichlorophenyl)-N1-aminopyrazine-2-carboxamidine, 3-amino-JV-(3-chloro-2- fluorophenyl)-iV-aminopyrazine-2-carboxamidine, 2-ammo-JV-(2,3-difluorophenyl)-.V- aminopyridine-3-carboxamidine, 2-amino-N-(2,3-dichlorophenyl)-iV-aminopyridine-3- carboxamidine, and 2-amino-iV-(3-chloro-2-fluorophenyl)-iV-aminopyridine-3- j carboxamidine, respectively.
Example 4. 5-Bromo-3-(4-(2,3-difluorophenyl)-4H-l,2,4-triazol-3-yl)pyrazin-2-amine (compound I-A-l)
Figure imgf000127_0001
[00119] To a solution of 3-(4-(2,3-difluorophenyl)-4H-l,2,4-triazol-3-yl)pyrazin-2-amine (700 mg, 2.55 mmol) in dry CH3CN (20 mL) was added NBS (550 mg, 3.09 (mmol). The solution was stirred at RT for Ih and poured into saturated NaHCO3 solution. The precipitate was collected by vacuum filtration and washed with water (700 mg, 78%) to produce 5- bromo-3-(4-(2,3-difluorophenyl)-4H-l,2:>4-triazol-3-yl)pyrazin-2-amine [LC-MS m/e= 353/354.6 (M-HH)]. Using the same procedure, 5-bromo-3-(4-(2,3-dichlorophenyl)-4H-l,2,4- triazol-3-yl)pyrazin-2-amine, 5-bromo-3-(4-(3-chloro-2-fluorophenyl)-4H-l,2,4-triazol-3- yl)pyrazin-2-amine, 5-bromo-3-(4-(2,3-difluorophenyl)-4H-l,2,4-triazol-3-yl)pyridin-2- amine, 5-bromo-3-(4-(2,3-dichlorophenyl)-4i7-l,2,4-triazol-3-yl)pyridin-2 -amine, and 5- bromo-3-(4-(3-chloro-2-fluorophenyl)-4H-l ,2,4-triazol-3-yl)pyridin-2 -amine can be produced from 3-(4-(2,3-dichlorophenyl)-4H-l,2,4-triazol-3-yl)pyrazin-2-amine, 3-(4-(3- chloro-2-fluorophenyl)-4//-l,2,4-triazol-3-yl)pyrazin-2-amine, 3-(4-(2,3-difluorophenyl)-4i/- l,2,4-triazol-3-yl)pyridin-2-amine, 3-(4-(2,3-dichlorophenyl)-4H-l,2,4-triazol-3-yl)pyridin- 2-amine, and 3-(4-(3-chloro-2-fluorophenyl)-4H-l ,2,4-triazol-3-yl)pyridin-2-amine, respectively. Example 5. 2-(ter£-Butylamino)-Λf-(2,3-di:fluoroρhenyl)pyridme-3-carboxamide
Figure imgf000128_0001
[00120] A room temperature solution of 2-fluoronicotinic acid (1 g) in DCM (20 mL) was sequentially treated with DMF (0.2 mL) and oxalyl chloride (0.62 mL, 1 eq). The resulting solution was stirred at RT for 1 hour and monitored by HPLC (analyte quenced with methanol) until the consumption of starting material was complete. The reaction mixture was cooled to 00C and sequentially treated with 2,3-difluoroaniline (1.4 g, 1.5 eq) and 2 mL of triethylamine. The reaction was warmed to RT and maintained for 3 additional hours, followed by washing the mixture with 2N HCl, saturated NaCl, and saturated NaHCO3 solution. The organic extracts were dried over MgSO4, filtered, and concentrated in vacuo. The resulting crude 2-fluoro-N-(2,3-difluorophenyl)pyridine-3-carboxamide was dissolved in NMP (20 mL) and reacted with excess £-butylamine at 800C for 14 hours. After cooling to RT, the solution was poured into sat NaHCO3 solution. The resulting precipitate was collected by filtration and washed with water. The crude product, 2-(tert-butylamino)-N- (2,3-difiuorophenyl)pyridine-3-carboxamide, was dried in vacuo and used directly in the next reaction without further purification. Using the same procedure, 2-(tø-/-butylamino)-JV-(2,3- dichlorophenyl)pyridine-3-carboxamide and 2-(/ert-butylarnino)-j'V'-(3-chloro-2- fluorophenyl)pyridine-3-carboxamide can be produced from the reaction of 2-fluoronicotinic acid with 2,3-dichloroaniline and 3-chloro-2-fluoroaniline, respectively.
Example 6. N-tert-Bvtiy\-3-(\ -(2,3-difluorophenyl)-l/7-tetrazol-5-yl)pyridin-2-amine
Figure imgf000128_0002
[00121] Crude 2-(te^butylamino)-N-(2,3-difiuorophenyl)pyridine-3-carboxamide (1.5 g) was dissolved in dry toluene (24 mL), combined with Lawesson's reagent (1.4 g, 0.7 eq), heated at 90°C for 10 hours, then evaporated to near dryness. The residue was diluted DCM (20 mL) and EtOH (20 mL) and treated with NH2NH2. The mixture was stirred at RT for 2 hours then concentrated in vacuo. The residue was diluted with Et2θ and washed with sat NaHCθ3 three times. The Et2θ layer was washed with 6N HCl solution (2 x 20 mL), and the combined HCl extracts treated with NaNO2 (3 eq) in water at RT for 30 min. The resulting mixture was neutralized with 6N NaOH (to pH 7-8) and extracted with EtOAc. The combined extracts were dried over MgSO4, filtered, and concentrated in vacuo to afford crude N-tert-huty\-2>-(\ -(2,3-difluorophenyl)-l//-tetrazol-5-yl)pyridin-2-amine. Using the same procedure, N-tert-huty\-3-{\ -(2,3-dichlorophenyl)-lH-tetrazol-5-yl)pyridin-2-amine and iγ"-fert-butyl-3-(l-(3-chloro-2-fluorophenyl)-lH-tetrazol-5-yl)pyridin-2-amine can be produced from 2-(fe^-butylamino)-iV-(2,3-dichlorophenyl)pvridine-3-carboxamide and 2- (/erΛ-butylamino)-N-(3-chloro-2-fluorophenyl)pyridine-3-carboxamide, respectively.
Example 7. 3-(l-(2,3-Difluorophenyl)-lH-tetrazol-5-yl)pyridin-2-amine
Figure imgf000129_0001
[00122] Crude 7V-ter*-butyl-3-(l -(2,3-difluorophenyl)- 1 i^-tetrazol-S-ylJpyridin^-amine was dissolved in MeOH (10 mL), treated with 6N HCl (20 mL), and heated to reflux for 2 hours. The mixture was subsequently cooled to RT and neutralized with 6N NaOH. The resulting precipitate was collected, washed with water, and dried in vacuo to give 3-(l-(2,3- difluorophenyl)-lH-tetrazol-5-yl)ρyridin-2-amine as white solid. Using the same procedure, 3-(l -(2,3-dichlorophenyl)-l//-tetrazol-5-yl)pyridin-2-amine and 3-(l -(3-chloro-2- fluorophenyl)-l//-tetrazol-5-yl)pyridin-2-amine can be produced from iV-te/"?-butyl-3-(l-(2,3- dichlorophenyl)-l/7-tetrazol-5-yl)ρyridin-2-amine and N-rer?-butyl-3-(l-(3-chloro-2- fluorophenyl)- l/]r-tetrazol-5-yl)pyridin-2 -amine, respectively. Example 8. 5-Bromo-3-(l-(2,3-difluorophenyl)-l//-tetrazol-5-yl)pyridin-2-amine (compound I-A-61)
Figure imgf000130_0001
[00123] 3-(l-(2,3-Difluorophenyl)-lH-tetrazol-5-yl)pyridin-2-amine was stirred in CH3CN (15 mL) and treated with NBS (2 eq). The reaction mixture was maintained at room temperature for 30 min. The reaction was subsequently poured into sat NaHCO3 solution and treated sequentially with 5 mL Na2S2θ3 and 2 mL 6N NaOH. The solids were filtered, washed with water, and dried in vacuo to afford 5-bromo-3-(l-(2,3-difluorophenyl)-li/- tetrazol-5-yl)pyridin-2 -amine, which was purified by silica gel chromatography. Using the same procedure, 5-bromo-3-(l-(2,3-dichlorophenyl)-lHr-tetrazol-5-yl)pyridin-2-amine and 5- bromo-3-(l-(3-chloro-2-fluorophenyl)-lH-tetrazol-5-yl)pyridin-2-amine can be produced from 3-(l-(2,3-dichlorophenyl)-lH-tetrazol-5-yl)pyridin-2-amine and 3-(l-(3-chloro-2- fluorophenyl)- lH-tetrazol-5-yl)pyridin-2 -amine, respectively.
Example 9. 5-(3-Aminopyrazin-2-yl)-4-(2,3-difluorophenyl)-4H-l ,2,4-triazol-3-ol
Figure imgf000130_0002
[00124] 3-Amino-iV-(2,3-difluorophenyl)-iV-aminopyrazine-2-carboxamidine (500 mg, 1.89 mmol) was dissolved in dry THF (10 mL) and CDI (340 mg, 2.10 mmol) was added at RT. The solution was kept at RT for overnight. After the solvent was removed by evaporation, the residue was added to saturated NaHCO3 solution and filtered. After washing with water, the crude yellow solid product (500 mg, 91%) was obtained after drying under vacuum. LC-MS m/e= 290.8 (M+H). Using the same procedure, 5-(3-aminopyrazin-2-yl)-4- (2,3-dichlorophenyl)-4i/- 1 ,2,4-triazol-3-ol, 5-(3-aminopyrazin-2-yl)-4-(3-chloro-2- fluorophenyl)-4H-l,2,4-triazol-3-ol, 5-(2-aminopyridin-3-yl)-4-(2,3-difluorophenyl)-4H- l,2,4-triazol-3-ol, 5-(2-aminoρyridin-3-yl)-4-(2,3-dichlorophenyl)-4//-l,2,4-triazol-3-ol, and 5-(2-aminopyridin-3-yl)-4-(3-chloro-2-fiuorophenyl)-4H-l,2,4-triazol-3-ol can be produced from 3-amino-N-(2,3-dichlorophenyl)-7V-aminopyrazine-2-carboxamidine, 3-amino-JV-(3- chloro-2-fluorophenyl)-iV-aminopyrazine-2-carboxamidine, 2-amino-iV-(2,3-difluorophenyl)- iV-aminopyridme-3-carboxamidine, 2-amino-iV-(2,3-dichlorophenyl)-iV-aminopyridine-3- carboxamidine, and 2-amino-Λ^-(3-chloro-2-fluorophenyl)-7V-aminopyridine-3- carboxamidine, respectively.
Example 10. 5-(3-Amino-6-bromopyrazin-2-yl)-4-(2,3-difluorophenyl)-4H-l,2,4-triazol-3-ol (compound I-A-14)
Figure imgf000131_0001
100125] To a solution of 5-(3-aminopyrazin-2-yl)-4-(2,3-difluorophenyl)-4H-l ,2,4-triazol- 3-ol (400 mg, 1.38 mmol) in dry CH3CN (15 mL) was added NBS (300 mg, 1.68 mmol). The solution was stirred at RT for 3h and poured into saturated NaHCO3 solution. The aqueous solution was extracted with ethyl acetate (2 x 20 mL). The combined organic layers were dried over MgSO4, filtered, and evaporated. The crude foam product was used directly in the next reaction without further purification (350 mg, 69%). LC-MS m/e= 369/370.7 (M+H). Using the same procedure 5-(3-ammo-6-bromopyrazin-2-yl)-4-(2,3- dichlorophenyl)-4//- 1 ,2,4-triazol-3-ol, 5-(3-amino-6-bromopyrazin-2-yl)-4-(3-chloro-2- fluorophenyl)-4/^-l,2,4-triazol-3-ol, 5-(2-amino-5-bromopyridin-3-yl)-4-(2,3- di fluorophenyl)^//- 1 ,2,4-triazol-3-ol, 5-(2-amino-5-bromopyridin-3-yl)-4-(2,3- dichlorophenyl)-47/-l ,2,4-triazol-3-ol, and 5-(2-amino-5-bromopyridin-3-yl)-4-(3-chloro-2- fluorophenyl)-4//-l,2,4-triazol-3-ol can be produced from 5-(3-aminopyrazin-2-yl)-4-(2,3- dichlorophenyl)-4//-l,2,4-triazol-3-ol, 5-(3-aminopyrazin-2-yl)-4-(3-chloro-2-fluorophenyl)- 4H-l,2,4-triazol-3-ol, 5-(2-aminopyridin-3-yl)-4-(2,3-difluorophenyl)-4H-l,2,4-triazol-3-ol, 5-(2-aminopyridin-3-yl)-4-(2,3-dichlorophenyl)-4H-l,2,4-triazol-3-ol, and 5-(2- aminopyridin-3-yl)-4-(3-chloro-2-fluorophenyl)-4H-l,2,4-triazol-3-ol, respectively. Example 11. 5-Bromo-3-(4-(3-chloro-2-fluorophenyl)-5-methyl-4//-l,2,4-triazol-3- yl)pyridin-2-amine (compound I-A-437)
Figure imgf000132_0001
[00126] 2-amiπo-iV-(3-chloro-2-fluorophenyl)-Λ/'-aminopyridine-3-carboxamidine (500 mg, 1.79 mmol) was dissolved in ethyl acetate (20 mL) and 6N HCl (1 mL) was added at RT. The solution was kept at RT for 14 hours and evaporated to 90% dryness. The residue was poured into saturated NaHCO3 and an off-white solid was collected by filtration, which was dried in vacuo. The resulting 3-(4-(3-chloro-2-fluorophenyl)-5-methyl-4H-l,2,4-triazol-3- yl)pyridin-2-amine was used as is in the subsequent bromination reaction. [00127] 3-(4-(3-Chloro-2-fluoroρhenyl)-5-methyl-4H-l,2,4-triazol-3-yl)pyridin-2-amine was dissolved in CH3CN (15 mL) and NBS (320 mg, 1.80 mmol) was added. The reaction mixture was stirred at RT for 30 min, and then poured into saturated NaHCO3 solution. The precipitate was collected by filtration, washed with water, and purified by HPLC to 5-bromo- 3-(4-(3-chloro-2-fluorophenyl)-5-methyl-4H-l,2:>4-triazol-3-yl)pyridin-2-amine as a yellow solid (400 mg, 1.05 mmol). LC-MS m/e= 382.0 (M+H); 1H-NMR (300 MHz, DMSO) 8.06 (d, J = 2.4 Hz, IH), 7.89 - 7.84 (m, IH), 7.79 - 7.74 (m, IH), 7.49 (td, J = 8.2, 3.2 Hz, IH)3 7.12 (d, J = 2.4 Hz, IH), 6.92 (s, 2H), 3.30 (s, 3H). Using the same procedure, 5-bromo-3- (4-(2,3-difluorophenyl)-5-methyl-4H-l,2,4-triazol-3-yl)pyrazm-2-amine, 5-bromo-3-(4-(2,3- dichloroρhenyl)-5-methyl-4H- 1 ,2,4-triazol-3-yl)pyrazin-2-amine, 5-bromo-3-(4-(3-chloro-2- fluorophenyl)-5-methyl-4//- 1 ,2,4-triazol-3-yl)pyrazin-2-amine, 5-bromo-3-(4-(2,3- difluorophenyl)-5-methyl-4H-l ,2,4-triazol-3-yl)pyridin-2-amine5 and 3-(4-(2,3- dichlorophenyl)-5-methyl-4H-l,2,4-triazol-3-yl)pyridin-2-amine can be produced from 3- amino-Λ/-(2,3-difluorophenyl)-iV-aminopyrazine-2-carboxamidine- 3-amino-N-(2,3- dichlorophenyl)-Λ'τ-aminopyrazine-2-carboxamidine, 3 -amino-7V-(3 -chloro-2-fluorophenyl)- iV-aminopyrazine-2-carboxamidine, 2-amino-iV1(2,3-difluorophenyl)-Λ''-aminopyridine-3- carboxamidine, and 2-amino-N-(2,3-dichlorophenyl)-N-aminopyridine-3-carboxamidine, respectively. Example 12. 5-Bromo-2-chloro-N-(2,3-dichlorophenyl)pyridine-3-carboxamide
Figure imgf000133_0001
[00128] To a slurry of S-bromo^-chloronicotinic acid (2.063 g, 8.725 mmol) in methylene chloride (2OmL) was slowly added oxalyl chloride (1.11 g, 8.725 mmol) followed by the addition of dimethylformamide (5 drops). After 4 hours, the mixture was concentrated in vaccuo to provide 2.21 g of S-bromo^-chloropyridine-S-carbonyl chloride as a light brown solid, which was used as is in the next reaction.
[00129] To a solution of 2,3 dichloroaniline (3.875 g, 23.914 mmol) in diethyl ether (20 mL) was added 5-bromo-2-chloropyridine-3-carbonyl chloride (3.0 g, 11.96 mmol). The reaction was stirred overnight. The precipitated solids were collected and washed with diethyl ether to provide 3.6 g of 5-bromo-2-chloro-iV-(2,3-dichlorophenyl)pyridine-3- carboxamide as a cream colored solid [LC-MS m/e— 380.0 (M+H)]. Using the same procedure, 5-bromo-2-chloro-iV-(2,3-fluorophenyl)pyridine-3-carboxamide and 5-bromo-2- chloro-7V-(3-chloro-2-fluorophenyl)pyridine-3-carboxamide can be produced from reacting 5- bromo-2-chloropyridine-3-carbonyl chloride with 2,3 fluororoaniline and 3-chloro-2- fluoroaniline, respectively.
Example 13. 5-Bromo-2-chloro-3-(l-(2,3-dichlorophenyl)-l//:-imidazol-2-yl)pyridine
Figure imgf000133_0002
[00130] To a slurry of 5-bromo-2-chloro-iV-(253-dichlorophenyl)pyridine-3-carboxamide (0.75 g,1.97 mmol) in benzene was added phosphorous pentachloride (0.5 g, 2.365 mmol) and the mixture heated to reflux. After 1.5 hours., the resulting solution was concentrated to provide 0.76 g of iV-((5-bromo-2-chloropyridin-3-yl)chlorometliylene)-2,3- dichlorobenzenamine as an off white solid. This material was used directly in the next reaction as is. [00131] To a solution of 2,2-dimethoxyethanamine (0.053 g, 0.505 mmol) in anhydrous tetrahydrofliran (5 mL) at 0-5 0C was added theiV-((5-bromo-2-chloropyridin-3- yl)chloromethylene)-2,3-dichlorobenzenamine (0.1 g, 0.2526 mmol) as a solution in anhydrous tetrahydrofuran (5 mL). After stirring overnight, jt?-toluene sulfonic acid (0.05 g, 0.51 mmol) was added and the reaction mixture was stirred an additional 2 hours. After concentration to dryness the resulting solid purified by flash chromatography (0 to 30% ethyl acetate/methylene chloride) to provide 0.092 g of 5-bromo-2-chloro-3-(l-(2,3- dichlorophenyl)-lH-imidazol-2-yl)pyridine. Using the same procedure, 5-bromo-2-chloro-3- (l-(2,3-difluorophenyl)-lH-imidazol-2-yl)pyridine and 5-bromo-2-chloro-3-(l-(3-chloro-2- fluorophenyl)-li/-imidazol-2-yl)pyridine can be produced from 5-bromo-2-chloro-N-(2,3- difluoroρhenyl)pyridine-3-carboxamide and 5-bromo-2-chloro-N-(3-chloro-2- fluorophenyl)p yridine-3 -carboxamide, respectively.
Example 14. 5 N-(4-Methoxybenzyl)-5-bromo-3-(l-(2,3-dichlorophenyl)-lH-imidazol-2- yl)pyridin-2 -amine
Figure imgf000134_0001
[00132] To a solution of excess (4-methoxyphenyl)methanamine (0.2 mL) in dioxane (2 mL)was added 5-bromo-2-chloro-3-(l-(2,3-dichlorophenyl)-lH-imidazol-2-yl)pyridine (0.089 g, 0.222 mmol). The mixture was heated at 100 0C for 16 hours in a sealed reaction vessel. The mixture was then cooled, diluted with ethyl acetate, washed with saturated aqueous sodium bicarbonate, and the combined organics dried over sodium sulfate. After filtration and concentration, the residue was purified by flash chromatography (20% hexanes/methylene chloride to 25% ethyl acetate/methylene chloride to provide 0.065 g of N- (4-methoxybenzyl)-5-bromo-3-(l-(2,3-dichlorophenyl)-lH-imidazol-2-yl)pyridin-2-amine [LC-MS m/e= 505.0 (M+Η)]. Using the same procedure, N-(4-methoxybenzyl)-5-bromo-3- ( 1 -(2,3-difluorophenyl)-liϊ-imidazol-2-yl)pyridin-2-amine and N-(4-methoxybenzyl)-5- bromo-3-(l-(3-chloro-2-fluorophenyl)-lH'-imidazol-2-yl)pyridin-2-amine can be produced from 5-bromo-2-chloro-3-(l-(2,3-difluorophenyl)-lH'-imidazol-2-yl)pyridine and 5-bromo-2- chloro-3-(l-(3-chloro-2-fluorophenyl)-lH-imidazol-2-yl)pyridine, respectively
Example 15. N-(4-Methoxybenzyl)-5-bromo-3-(5-(2,3-dichlorophenyl)-2-methyloxazol-4- yl)pyridin-2-amine
Figure imgf000135_0001
[00133] To a solution of 1 -(5-bromo-2-chloropyridin-3-yl)-2-(2,3- dichlorophenyl)ethanone (0.315 g, 0.83 mmol) in TΗF (3 mL) was added ΗDNIB (0.585 g, 1.2 mmol, [hydroxy(2,4-dinitrobenzenesulfonyloxy)iodo]benzene, see Lee et al., Synlett, 10: 1563-1564, 2001) in 1 ,2-dichloroethane (3 mL). The reaction mixture was heated to reflux in a sealed tube for 2 hours, at which point a solution formed. The reaction was cooled to RT and acetamide (2 eq., 0.115 g, 1.9 mmol) was added. The reaction was then heated to reflux for 18 hours. After cooling, the mixture was dissolved in MeOH, adsorbed onto Celite™ and purified by silica gel chromatography ( 5 to 40% EtOAc/hexanes) to give 5-bromo-2-chloro- 3-(5-(2,3-dichlorophenyl)-2-methyloxazol-4-yl)pyridine as a colorless oil [0.084 g, 24% yield; LC-MS m/e= 418.8 (M+Η)].
[00134] To a solution of -bromo-2-chloro-3-(5-(2,3-dichlorophenyl)-2-methyloxazol-4- yl)pyridine (0.08 g, 0.19 mmol) in dioxane (2 mL) was added 4-methoxybenzyl amine (0.5 mL). The reaction mixture was heated to 12O0C in a sealed tube for 18 hours. After cooling, the mixture was diluted with EtOAc, washed with H2O (2 x 5 mL), and the aqueous phase back-extracted with EtOAc (2 x 5 mL). The combined organics were adsorbed onto Celite and purified by silica gel chromatography (5-40% EtOAc/hexanes) to give N-(4- methoxybenzyl)-5-bromo-3-(5-(2,3-dichlorophenyl)-2-methyloxazol-4-yl)pyridin-2-amine as a yellow solid (82 mg, 83% yield). Using the same procedure, N-(4-methoxybenzyl)-5- bromo-3-(5-(2,3-difluorophenyl)-2-methyloxazol-4-yl)pyridin-2-amine and 7V-(4- methoxybenzyl)-5-bromo-3-(5-(3-chloro-2-fluorophenyl)-2-methyloxazol-4-yl)pyridin-2- amine can be produced from l-(5-bromo-2-chloropyridin-3-yl)-2-(2,3- difluorophenyl)ethanone and 1 -(5-bromo-2-chloropyridin-3-yl)-2-(3-chloro-2- fluorophenyl)ethanone, respectively.
Example 16. 5-Bromo-3 -(5-(2,3 -dichlorophenyl)- 1 ,2,3-thiadiazol-4-yl)pyridin-2-amine (compound I-C-2)
Figure imgf000136_0001
[00135] To a solution of 1 -(5-bromo-2-chloroρyridin-3-yl)-2-(2,3- dichlorophenyl)ethanone (1.14 g, 3.03 mmol) in EtOH (40 mL) was added ethyl carbazate (0.95 g, 9.09 mmol) and TsOH (2 mg). The reaction mixture was heated for 3 hours, followed by evaporated of the volatiles to give ethyl-2-(l-(5-bromo-2-chloropyridin-3-yl)-2- (2,3-dichlorophenyl)ethylidene)hydrazinecarboxylate as a yellow oil, which was used directly in the next step.
[00136] A solution of ethyl-2-(l-(5-bromo-2-chloropyridin-3-yl)-2-(2,3- dichlorophenyl)ethylidene)hydrazinecarboxylate (3.03 mmol) was stirred in thionyl chloride (20 mL). The solution was heated from 00C to RT and stirred for 3.5 hours. The solvent was evaporated to give a yellow oil which was adsorbed onto Celite™ and purified by silica gel chromatography (5-40% EtOAc/hexanes) to give 5-bromo-2-chloro-3-(5-(2,3- dichlorophenyl)-l,2,3-thiadiazol-4-yl)pyridine as a yellow solid (640mg, 50% yield over 2 steps).
[00137] To a solution of 5-bromo-2-chloro-3-(5-(2,3-dichlorophenyl)-l,2,3-thiadiazol-4- yl)pyridine (0.37 g, 0.88 mmol) in dioxane (2 mL) was added 4-methoxybenzylamine (4 eq, 460 μL, 3.5 mmol). The solution was heated in a sealed tube at 120 0C for 18h. After cooling, the mixture was filtered and the filtrate was reduced in vacuo. The resulting oil was purified by silica gel chromatography (5 to 70% EtOAc/hexanes) to provide JV-(4- methoxybenzyl)-5-bromo-3-(5-(2,3-dichlorophenyl)-l,2,3-thiadiazol-4-yl)pyridin-2-amine as a yellow sticky solid (415mg, 90% yield)
[00138] To a solution of N-(4-methoxybenzyl)-5-bromo-3-(5-(253-dichlorophenyl)- 1,2,3- thiadiazol-4-yl)pyridin-2-amine (0.128 mmol) in CH2Cl2 (2 mL) was added TFA (3 mL). The mixture was heated at 40 0C for 20 hours then at RT for 3d. The volatiles were evaporated and the residue purified by preparative reversed-phase HPLC to afford 5-bromo- 3-(5-(2,3-dichlorophenyl)-l,2,3-thiadiazol-4-yl)pyridin-2-amine as a colorless oil (0.019 g, 37 % yield). Using the same procedure, 5-bromo-3-(5-(2,3-fluorophenyl)-l ,2,3-thiadiazol-4- yl)pyridin-2-amine and 5-bromo-3-(5-(3-chloro-2-fiuorophenyl)-l ,2,3-thiadiazol-4- yl)pyridiπ-2 -amine can be produced from l-(5-bromo-2-chloropyridin-3-yl)-2-(2,3- difluorophenyl)ethanone and 1 -(5-bromo-2-chloropyridin-3-yl)-2-(3-chloro-2- fluorophenyl)ethanone, respectively.
Example 17. 5-Bromo-3-(5-(2,3-difluorophenyl)-l//-tetrazol-l-yl)pyridin-2-amine
Figure imgf000137_0001
[00139] To a solution of 2-chloro-3-nitropyridine (5.0 g, 31.53 mmol) in anhydrous NMP (100 mL) was added /erf-butylamine (10 mL, 94.52 mmol). The solution was heated at 60 0C for 14 hours and cooled. The mixture was poured into IN HCl solution (400 mL) and extracted with EtOAc (3 x 150 mL). The combined organic layers were washed with saturated NaHCO3 solution and dried over MgSO4. After filtration, the solvent was evaporated under vacuum to afford 7V-ter/-butyl-3-nitropyridin-2-amine as dark brown syrup. [00140] To a solution of the 7V-tert-butyl-3-nitropyridin-2-amine in MeOH (200 mL), was added solid NH4Cl (16.4 g, 306.54 mmol) and zinc dust (10.0 g, 153 mmol). The suspension was heated under reflux for 3 hours and then cooled to RT. After filtration through Celite™, the solvent was removed by vacuum evaporation. The resulting black residue was taken up to EtOAc (300 mL) and filtered through Celite™ again to remove the remaining NH4Cl. The solvent was again evaporated and the residue was dried on the high vacuum oil pump overnight to yield 7V2-ter/-butylpyridine-2, 3 -diamine.
[00141] To a solution of N2-ter/-butylpyridine-253-diamine in dry DCM (150 mL) was added DIEA (17 mL, 95.5 mmol). The solution was cooled to 00C and carefully added 2,3- difluorobenzoyl chloride (8.35 g, 47.30 mmol). The reaction was allowed to warm up to RT and stirred for 1 hr. The mixture was then washed with water, saturated NaHCO3, and dried over MgSO4. Removal of the solvent by evaporation, the residue was purified by silica gel chromatography (5%-60% EtOAc/hexane) to yield iV-(2-(tert-butylamino)pyridin-3-yl)-2,3- difluorobenzamide (4.0 g, 13.10 mmol); LC-MS m/e= 306.1 (M+H).
[00142] To a solution of iV-(2-(ter?-butylamino)pyridin-3-yl)-2,3-difluorobenzamide (4.0 g, 13.10 mmol) in anhydrous 1,4-dioxane (60 mL) was added Lawesson's reagent (3.7 g, 9.15 mmol). The solution was heated at 800C for 40min and cooled to RT. To the cooled solution was added a solution of hydrazine (5 mL) in EtOH (60 mL). The mixture was stirred at RT for 14 hours and poured into saturated NaHCO3 solution (300 mL). The aqueous solution was extracted with EtOAc (3 x 150 mL), the combined organic layers were then reverse-extracted with 2N HCl solution (2 x 150 mL). The combined acidic layers were treated with NaNO2 (0.9 g, 13.1 mmol) at RT for 10 min. The reaction mixture was cooled to 00C and solid NaOH was added to a pH of 8. Filtration yielded 7V-tert-butyl-3-(5-(2,3- difluorophenyl)-l//-tetrazol-l-yl)pyridin-2-amine as a brownish solid; LC-MS m/e= 303.1 (M+H-N2).
[00143] To a solution of JV-ter^butyl-3-(5-(2,3-difluorophenyl)-l//-tetrazol-l -yl)pyridin- 2-amine (13.10 mmol) in CH3CN (150 mL) was added NBS (2.33 g, 13.10 mmol). The reaction was stirred at RT for 20 min and poured into a solution OfNa2SO3 (150 mL) and 6N NaOH (5 mL). The mixture was stirred for an additional 30 min and filtered. The resulting dark colored solid was collected and washed with water. To this solid was added MeOH (100 mL) and 6N HCl (100 mL). The mixture was heated under reflux for 4 hours and cooled to 00C. NaOH was added as a solid until a pH of 9 was achieved, at which time 5- bromo-3-(5-(2,3-difluorophenyl)-li7-tetrazol-l-yl)pyridin-2-amine was collected as a pinkish solid by filtration (3.8 g, 10.76 mmol); LC-MS m/e= 325.1 (M+H-N2). Using the same procedure, 5-bromo-3-(5-(2,3-dichlorophenyl)-lH-tetrazol-l-yl)pyridin-2-amine and 5- bromo-3-(5-(3-chloro-2-fluorophenyl)-lH-tetrazol-l -yl)pyridin-2-amine can be produced from 2,3-dichlorobenzoyl chloride and 3-chloro-2-fluorobenzoyl chloride, respectively.
Example 18. 1 -(5-Bromo-2-chloropyridin-3-yl)-2-(2,3-dichlorophenyl)-3- (dimethylamino)prop-2-en- 1 -one
Figure imgf000139_0001
[00144] A solution of l-(5-bromo-2-chloropyridin-3-yl)-2-(2,3-dichlorophenyl)ethanone in dry THF was slowly treated with DMF-DMA under N2 atmosphere for 45 min and stirred at RT until completion of the reaction. The reaction mixture was concentrated under reduced pressure to obtain l-(5-bromo-2-chloropyridin-3-yl)-2-(2,3-dichlorophenyl)-3- (dimethylamino)prop-2-en-l-one as semi solid. Washing the crude product with n-pentane and pet-ether resulted in a free flowing light brown solid (1.0 g, 58.8%) which was used as is in the next reaction without further purification.
Example 19. 5-Bromo-2-chloro-3-(4-(2,3-dichlorophenyl)-l-cyclohexyl-lH-pyrazol-3- yl)pyridine and 5-bromo-2-chloro-3-(4-(2,3-dichlorophenyl)-l-cyclohexyl-l//"-pyrazol-5- yl)pyridine
Figure imgf000139_0002
[00145] To a stirred solution of cyclohexyl hydrazine hydrochloride in toluene was added K2CO3. After stirring for 10 min at RT, the mixture was treated with l-(5-bromo-2- chloropyridin-3-yl)-2-(2,3-dichlorophenyl)-3-(dimethylamino)prop-2-en- 1 -one followed by treatment with acetic acid. The reaction mixture was stirred for 10 min at RT and heated to 80°C for 18 hr. The mixture was cooled to RT, concentrated to dryness under reduced pressure, diluted with water, extracted with ethyl acetate, washed with saturated aqueous NaHCO3 solution, water, and brine solution, respectively, prior to drying over Na2SO4. Filtration and evaporation of solvent gave crude mixture of 5-bromo-2-chloro-3-(4-(2,3- dichlorophenyl)- 1 -cyclohexyl- 1 H-pyrazol-3 -yl)pyridine and 5-bromo-2-chloro-3-(4-(2,3- dichlorophenyl)-l-cyclohexyl-lH-pyrazol-5-yl)pyridine as mixture of regioisomeric pyrazoles (700 mg , 52.2% yield; 80:20 as determined by LC-MS). The product mixture can be purified by column chromatography over silica gel 60-120 mesh size, 0 -» 2% EtOAc in pet ether to obtain both regioisomeric products.
Example 20. ΛT-(4-Methoxybenzyl)-5-bromo-3-(4-(2,3-dichlorophenyl)-l -cyclohexyl- 1 H- pyrazol-3-yl)pyridin-2-amine and 7V-(4-methoxybenzyl)-5-bromo-3-(4-(2,3- dichlorophenyl)-l-cyclohexyl-li7-pyrazol-5-yl)pyridin-2-amine
Figure imgf000140_0001
[00146] A solution of 5-bromo-2-chloro-3-(4-(2,3 -dichlorophenyl)-l -cyclohexyl- IH- pyrazol-3-yl)pyridine and 5-brorno-2-chloro-3-(4-(2,3-dichlorophenyl)-l -cyclohexyl- IH- pyrazol-5-yl)pyridine (about a 4/1 mixture) in 1,4-dioxane was treated with/?-methoxybenzyl amine at RT and the resulting mixture was heated to 120 °C for 24 hours until completion of reaction as monitored by LCMS. The reaction mixture was cooled to RT, evaporated to dryness, treated with saturated aqueous NaHCO3 solution, and extracted with EtOAc. The combined organic, layer was washed with brine solution, dried over Na2SO4, and concentrated to obtain a mixture of iV-(4-methoxybenzyl)-5-bromo-3-(4-(2,3- dichlorophenyl)-l -cyclohexyl- 1 H-pyrazol-3 -yl)pyridin-2-amine and 7V-(4-methoxybenzyl)-5- bromo-3-(4-(2,3-dichlorophenyl)-l -cyclohexyl-lH-pyrazol-5-yl)pyridin-2-amine as brown oil (350 mg, 54.6% yield). Each regioisomeric product was obtained in pure form after purification by silica gel column chromatography (60-120 mesh silica gel, 3%— >10% EtOAc/Pet ether). Example 21. 5-Bromo-3-(l-(2,3-difluorophenyl)-liir-imidazo[4,5-c]pyridin-2-yl)pyridm-2- amine
Figure imgf000141_0001
[00147] To a solution of 4-chloro-3-nitropyridine (3.0 g, 18.9 mmol) in dry ethanol as added 2,3-difluorobenzenamine (3.66 g, 28.3 mmol). The reaction mixture was refluxed for 30 min. After cooling and concentration in vacuo to give a brown solid, the crude product was taken up in water and the aqueous solution adjusted to a pH of 8-9 and the solution extracted with EtOAc (3 x 100 mL). The combined organics were washed with water (3 x 100 mL) and brine solution (1 x 100 mL), respectively, dried over Na2SO4, and evaporated in vacuo to give a light brown solid. The crude product was triturated with petroleum ether (3 x 30 mL) to afford iV*-(2,3-difluorophenyl)-3-nitropyridin-4-amine (3.7 g, 79% yield) as light yellow solid.
[00148] To a solution of 7V-(2,3-difluorophenyl)-3-nitropyridin-4-amine in 200 mL of methanol under an atmosphere of nitrogen was added 10% PoVC (0.75 g). The atmosphere was replaced with hydrogen gas and the reaction mixture stirred under balloon pressure for 14 hours at RT. The reaction mixture was filtered over Celite which was washed with methanol (3 x 30 mL). The filtrate was concentrated in vacuo to give a brown solid. The crude product was triturated with petroleum ether (3 x 50 mL) to afford //-(2,3- difluorophenyl)pyridine-3,4-diamine (4.0 g, 90% yield) as light brown solid; LC-MS: 222.2 (M+H).
[00149] To a solution of 2-fluoropyridine-3-carboxylic acid (1.41 g, 10.00 mmol) in anhydrous DMF (15 mL) was added HOBT (2.56 g, 16.74 mmol) and Et3N (2.0 g, 20.09 mmol) at RT. The mixture was cooled to 0 0C, treated with a solution ofN*-(2,3- difluorophenyl)pyridine-3,4-diamine in DMF (15 mL), stirred for 15 min at RT and EDC (3.2 g, 16.74 mmol) was added portion wise. The reaction mixture was warmed to RT and stirred for 16 hr. The reaction mixture was diluted with water (100 mL) and extracted with EtOAc (4 x 200 mL). The combined organics were washed with saturated aq. NaHCO3 solution (2 x 100 mL), water (2 x 100 mL), brine solution (100 mL), and dried (Na2SO4). Filtration and evaporation of the solvent in vacuo gave a brown solid. Chromatographic purification (silica gel, 1% methanol in chloroform) provided iV-(4-(2,3- difluorophenylamino)pyridin-3-yl)-2-fluoropyridine-3-carboxamide (1.05 g, 45% yield) as yellow solid; LC-MS: 345.2 (M+H).
[00150] To a solution of iV-(4-(2,3-difluorophenylamino)pyridin-3-yl)-2-fluoropyridine-3- carboxamide (4.0 g, 11.6 mmol) in NMP (30 mL) was added r-butylamine (15.0 mL, 145.3 mmol) at RT. The reaction mixture was slowly heated to 100 °C and stirred at this temperature for 48 hr. The reaction mixture was diluted with water (150 mL) and extracted with EtOAc (3 x 200 mL). The combined organics were washed with water (4 x 100 mL), brine solution (100 mL), and dried (Na2SO4). Filtration and evaporation of solvent in vacuo gave iV-(4-(2,3-difluorophenylamino)pyridin-3-yl)-2-(/'e/-r-butylamino)pyridine-3- carboxamide (4.1 g, 89% yield), which was used as is in subsequent reactions; LC-MS: 398.3 (M+H).
[00151] To a stirred solution of iV-(4-(2,3-difluorophenylamino)pyridin-3-yl)-2-(tør/- butylamino)pyridine-3-carboxamide (3.0 g, 7.55 mmol) in 1, 4-dioxane (50 mL) was added 2,4-bis(4-methoxyphenyl)-l ,3-dithia-2,4-diphosρhetane-2,4-disulfide (Lawesson's reagent, 4.5 g, 11.3 mmol) at room temperature. The reaction mixture was heated at reflux (110 0C) for 10 hr. The reaction mixture was concentrated in vacuo and the resulting residue was taken in saturated aq. NaHCO3 solution (100 mL) and extracted with CHCl3 (200 mL). The organics were washed with water (3 x 100 mL), brine solution (1 x 100 mL), and dried (Na2SO14). Filtration and evaporation of solvent in vacuo, followed by silica gel chromatography (0.5 % methanol/chloroform) gave N-tert-butyl-3-(l-(2,3-difluorophenyl)- lH-imidazo[4,5-c]pyridin-2-yl)pyridin-2 -amine (1.2 g, 41.9% yield) as a light yellow solid; LC-MS: 380.2 (M+H).
[00152] To a stirred solution of N-tert-bvΛy\-3-(l -(2,3-difiuorophenyl)-lH-imidazo[4,5- c]pyridin-2-yl)pyridin-2-amine (1.5 g, 3.95 rrimol) in methanol (7.0 mL) was added 6N aq. HCl at room temperature and the mixture heated at reflux for 5 hr. After cooling to 00C, the pΗ of the solution was adjusted to pΗ 8-9 with 2N aq. NaOH solution (150 mL). The resulting precipitate was filtered off and dissolved in EtOAc (300 mL). The combined organics were washed with water (3 x 100 mL), brine (100 mL), and dried (Na2SO4). Filtration and evaporation of solvent in vacuo gave a solid, which was triturated with petroleum ether (3 x 2OmL) to give 3-(l-(2,3-difluorophenyl)-lH-imidazo[4,5-c]pyridin-2- yl)pyridin-2-amine (0.9 g, 75% yield) as a light yellow solid; LC-MS: 324.0 (M+Η). [00153] A solution of 3-(l-(2,3-difluorophenyl)-lH'-imidazo[455-c]pyridin-2-yl)pyridin-2- amine (2.4 g, 7.43 mmol) in acetonitrile (70 mL) was treated with 7V-bromosuccinimide (1.45 g, 18.17 mmol) at 0 0C and the reaction mixture stirred for 30 min. reaction mixture was treated with saturated aq. NaHCO3 solution (150 mL), stirred for 30 min, and the resulting precipitate filtered and dissolved in CHCl3 (200 mL). The combined organics were washed with water (3 x 70 mL), brine (70 mL), and dried (Na2SO4). Filtration and evaporation of solvent in vacuo gave a solid, which was purified by silica gel chromatography (4% methanol/chloroform) to give 5-bromo-3-(l-(2,3-difluorophenyl)-l//-imidazo[4,5-c]pyridin- 2-yl)pyridin-2-amine (2.2 g, 75% yield) as a light yellow solid; LC-MS: 402.0, 404.0 (M+H).
Example 22. 5-Bromo-3-(5-(2,3-dichlorophenyl)thiazol-4-yl)pyridin-2-amine (compound I-C-7)
Figure imgf000143_0001
[00154] To a stirred solution of 1 -(5-bromo-2-chloropyridin-3-yl)-2-(2,3- dichlorophenyl)ethanone (1.01 g, 2.66 mmol) in 1 :1 methylene chloride/tetrahydrofuran (26 mL) was added bromine (136 μL, 2.66 mmol) and 3 mL acetic acid. The reaction mixture was stirred at room temperature overnight, at which point the orange color had dissipated. Concentration gave 2-bromo-l-(5-bromo-2-chloropyridin-3-yl)-2-(2,3- dichlorophenyl)ethanone as an orange oil (1.1 g, 95 %), which was used in the next reaction without further purification.
[00155] Formamide (528 μL, 13.3 mmol) and P2S5 (6.5 g, 14.63 mmol) were stirred in acetonitrile for 30 minutes, until the suspension became an unstirrable paste. The supernatant was decanted and added to an acetonitrile solution of 2-bromo-l-(5-bromo-2- chloropyridin-3-yl)-2-(2,3-dichlorophenyl)ethanone (0.609 g,l .33 mmol). The reaction mixture was stirred at room temperature overnight. The solids were removed by filtration, rinsed with methylene chloride, and the filtrate was concentrated and taken up in ethyl ether. The organics were filtered through silica, which was washed with diethyl ether. The filtrate was concentrated and the product obtained by crystallization from hexanes/ether to provide 5-bromo-2-chloro-3-(5-(2,3-dichlorophenyl)thiazol-4-yl)pyridine as a white solid [532 mg, 95 % yield; LC-MS, M+H = 419.0; 1H-NMR (300 MHz, CDCl3) 8.51 (IH, s), 8.36 (IH, d), 7.78 (IH, d), 7.42 (IH, dd), 7.01-7.11 (2H, m)].
[00156] A solution of 5-bromo-2-chloro-3-(5-(2,3-dichlorophenyl)thiazol-4-yl)pyridine (520 mg, 1.24 mmol) and/>-methoxybenzyl amine (373 mg, 2.72 mmol) in DME (12 mL) was stirred at 80 °C for 48 hours. The reaction mixture was concentrated and the residue purified by silica gel chromatography (0 - 25 % EtOAc/hexanes) to give N-(4- methoxybenzyl)-5-bromo-3-(5-(2,3-dichlorophenyl)thiazol-4-yl)pyridin-2-amine as a pale yellow solid [631 mg, 85 % yield; LC-MS, M+H = 519.9; 1H-NMR (300 MHz, CDCl3) 8.85 (1 H5 s), 8.00 (1 H, d, J = 2.3 Hz), 7.45 (IH, dd, J = 1.9, 7.6 Hz), 7.10-7.17 (4 H, m), 7.01 (1 H, d, J = 2.4 Hz)5 6.77 (1 H, t, J = 6.7 Hz)].
[00157] A solution of N-(4-methoxybenzyl)-5-bromo-3-(5-(2,3-dichlorophenyl)thiazol-4- yl)pyridin-2-amine (110 mg) and TFA (100 μL) in methylene chloride (2 mL) was stirred at 40 0C overnight. The reaction was concentrated and purified by reversed-phase HPLC to give 5-bromo-3-(5-(2,3-dichlorophenyl)thiazol-4-yl)pyridin-2-amine as a white solid [1.1 mg, 2 % yield; LC-MS M+H = 519.9; 1H-NMR (300 MHz, methanol-^) 9.17 (IH, d), 7.95 (IH, d), 7.60-7.63 (2H, m), 7.34-7.35 (I H, m), 7.27 (IH, dd)]. Example 23. 3-(5-(2>3-Dichlorophenyl)thiazol-4-yl)-5-(pyridin-3-yl)pyridin-2-amine (compound I-C-8)
Figure imgf000145_0001
[00158] A solution of N-(4-memoxybenzyl)-5-bromo-3-(5-(2,3-dichlorophenyl)thiazol-4- yl)pyridin-2-amine (50 mg, 0.116 mmol) in dimethoxyethane (0.5 mL) and water (0.5 mL) was degassed with nitrogen while adding NaHCO3 (30 mg, 0.347 mmol), and palladium tetrakis triphenylphosphine (13 mg, 0.012 mmol). 3-(4,4,5,5-Tetramethyl-l,3,2- dioxaborolan-2-yl)pyridine (28 mg, 174 mmol)was added under nitrogen and the mixture was heated to 120 0C overnight. The reaction mixture was concentrated and taken up in ethyl acetate, filtered through silica with ethyl acetate eluant, and the filtrate concentrated to provide iV-(4-methoxybenzyl)-3-(5-(2,3-dichlorophenyl)thiazol-4-yl)-5-(pyridin-3-yl)pyridin- 2-amine, which was used in the next reaction without further purification. [00159] A solution of N-(4-methoxybenzyl)-3-(5-(2,3-dichlorophenyl)thiazol-4-yl)-5- (pyridin-3-yl)pyridin-2-amine (24 mg, 0.046 mmol) in TFA (1 mL) was heated to reflux overnight. The solvent was removed in vacuo and the compound was purified by reversed- phase HPLC to provide 3-(5-(2,3-dichlorophenyl)thiazol-4-yl)-5-(pyridin-3-yl)pyridin-2- amine as a while solid [2.2 mg, 9 % yield; LC-MS M+H = 399.2; 1H-NMR (300 MHz, methanol-.**) 9.34 (IH, s), 8.57 (IH, dd), 8.41 (IH, d), 8.28 (IH, d), 7.79 (IH3 d), 7.71 (IH, dd), 7.58 (IH, dd), 7.57 (IH3 1), 7.46 (IH, t), 2.64 (3H, s)]. Example 24. tert-Butyl 2-bromo-4,5-dihydrothieno[2,3-c]pyridine-6(7//)-carboxylate
Figure imgf000146_0001
[00160] To tert-bntyl 4,5-dihydrothieno[2,3-c]pyridine-6(7Jyr)-carboxylate (2.0 g, 8.4 mmol) in chloroform (100 mL) at 0 0C was added dropwise bromine (430 μL, 8.4 mmol). The reaction was stirred 45 min with gradual warming to room temperature. The mixture was cooled to 0 0C5 and triethylamine (1.2 mL, 8.4 mmol) was added followed by άi-tert- butyl di carbonate (913 mg, 4.2 mmol). The reaction was stirred 1 hour, diluted with diehloromethane (50 mL), washed with 1.0 N HCl (1 x 50 mL), dried over MgSO4, and concentrated under reduced pressure. The resulting crude residue was purified via silica gel chromatography to afford 2 g (77% yield)
Figure imgf000146_0002
2-bromo-4,5-dihydrothieno[2,3- c]pyridine-6(7H)-carboxylate as a colorless oil; 1H NMR (CDCl3) δ 6.78 (s, IH), 4.50 (s, 2H), 3.67 (t, J= 5.7 Hz, 2H), 2.65 (t, J= 5.7 Hz, 2H), 1.50 (s, 9H); ES-MS: m/e = 261.9 (M- 55)+.
Example 25. /ert-Butyl 4-(5-bromothiazol-2-yl)piperidine-l-carboxylate
Figure imgf000146_0003
[00161] To a solution offers-butyl 4-thiocarbamoylpiperidine-l-carboxylate (1 g, 4.09 mmol) in acetone (5 mL) was added 2-chloroacetaldehyde (0.32 g, 4.08 mmol). The mixture was heated under reflux for 4 hours. Additional 2-chloroacetaldehyde (0.32 g, 4.08 mmol) was added and heating was continued for another 14 hours. The solvent was removed by evaporation and the crude product was purified by silica gel chromatography to give tert- butyl 4-(thiazol-2-yl)piperidine-l-carboxylateas an oil [530 mg (1.97 mmol); LC-MS = 213.1 (M+H); 1H NMR (300 MHz, CDC13) 7.74 (d, J = 3.3 Hz5 IH), 7.26 (d, J = 3.3 Hz3 IH), 4.23 (brd, 2H), 3.22 (m, IH), 2.91 (t, 2H), 2.14 (m, 2H), 1.77 (m, 2H), 1.48 (s, 9H)]. [00162] To a solution of tert-bvΛyl 4-(thiazol-2-yl)piperidine-l -carboxylate (530 mg, 1.97 mmol) in acetonitrile (10 mL) was added NBS (1.40 g, 7.86 mmol). The mixture was stirred at RT for 14 hours and heated at 50°C for 4 hours. The reaction mixture with some starting material recovered was poured into a solution OfNa2SO3 (30 mL) and 6N NaOH (2 mL). The aqueous layer was extracted with EtOAc, dried over MgSO4, and the combined organics concentrated in vacuo. The residue was purified by silica gel chromatography to provide tert-butyl 4-(5-bromothiazol-2-yl)piperidine-l -carboxylate as a yellow oil [210 mg (0.61 mmol); 1H NMR (300 MHz, CDCl3) 7.59 (s, IH)5 4.20 (brd, J = 12.9 Hz, 2H)5 3.13 (tt, J = 3.8, 11.5 Hz5 IH), 2.89 (t, J = 11.6 Hz5 2H)5 2.08 (d, J = 11.7 Hz, 2H), 1.72 (dq, J = 4.3, 11.9 Hz, 2H), 1.49 (s, 9H)].
Example 26. tert-Butyl 2-bromo-455,758-tetrahydrofuro[3,2-<5rjazepine-6-carboxylate
Figure imgf000147_0001
[00163] 2-(Furan-2-yl)-2-oxoacetic acid (15.0 g, 107 mmol) was dissolved in chloroform (420 mL) and treated with EtOH (165 mmol) and H2SO4 (1 mL). The reaction was heated to 65 °C overnight, at which point LC-MS indicated disappearance of starting material and product formation. The crude reaction was cooled and washed with sat. aq. NaHCO3 (3 x 100 mL) and brine (100 mL). The organics were concentrated to provide ethyl 2-(furan-2- yl)-2-oxoacetate (15.5 g5 64 %) as a brown oil, which was used without fiαrther purification. f 00164] Ethyl 2-(furan-2-yl)-2-oxoacetate (15.5 g, 92.1 mmol) in ethanol (500 mL) and water (25 mL) at 0 0C was treated with sodium borohydride (1.74 g, 46.05 mmol). The mixture was stirred for 15 minutes, at which point acetic acid (10 mL) was slowly added. After the cessation of gas evolution, water (100 mL) was carefully added, and the reaction mixture was concentrated. The residue was dissolved in methylene chloride and washed with brine. The crude product was dried and concentrated to give ethyl 2-(furan-2-yl)-2- hydroxyacetate as a pale brown viscous oil (11.05 g, 71 %) which was used without further purification.
[00165] A solution of ethyl 2-(furan-2-yl)-2-hydroxyacetate (11.05 g, 64.5 mmol), trimethylorthoacetate (29.8 mL, 387 mmol) and hexanoic acid (2 mL) in decalin (195 mL) is heated at 180 0C for 12 hours. The reaction is cooled and extracted with methanol to provide a mixture of diester and decalin. This mixture is dissolved in methanol (250 mL), cooled to 0 0C, treated with 2 M NaOH (150 mL) and stirred for 12 hours. The methanol is removed and the crude reaction is extracted with ether, and the basic layer is acidified with 6 N HCl and extracted with ethyl acetate. The organic layer is washed with brine, dried and concentrated to give 2,3-di(carboxymethyl)furan.
[00166] A solution of 2,3-di(carboxymethyl)furan (35 mmol) in THF (400 mL) is cooled to 00C and treated with BH3^THF (174 mmol) over 10 minutes, and stirred for an additional 20 minutes at 0 °C before warming to room temperature overnight. The crude reaction is poured over saturated aqueous NaHCO3 and extracted with ethyl acetate. The organic layer is dried and concentrated to give 2,3-di(2-hydroxyethyl)furan.
[00167] A solution of 2,3-di(2-hydroxyethyl)furan (23 mmol) in methylene chloride (114 mmol) is cooled to 0 0C and treated with triethylamine (69 mmol) followed by dropwise addition of methanesulfonyl chloride (50.4 mmol) over 10 minutes. After 1 hour the reaction mixture is transferred to a separatory funnel and extracted with cold water, 10 % citric acid, saturated aqueous NaHCO3, and brine. The organic layer is dried and concentrated twice with the aid of dioxane to produce 2,3-di(2-methanesulfonyloxy-ethyl)furan, which is used crude without further purification.
[00168] A solution of 2,3-di(2-methanesulfonyloxyethyl)furan (114 mmol) in dioxane (168 mL) is treated with postassium carbonate (337 mmol) and benzyl amine (70.1 mmol) and heated to 102 0C for 18 hours. The reaction is cooled, the precipitate removed by filtration, and the mother liquor concentrated to give a crude oil which is purified by silica chromatography (EtOAc/hexanes eluant) to provide 6-benzyl-5,6,7,8-tetrahydro-4i7- furo [3 ,2-d] azepine.
[00169] A solution of 6-benzyl-5,6,7,8-tetrahydro-4H-furo[3,2-cT]azepine (11.3 mmol) in methylene chloride (56 mL) is cooled to °C and treated with 1-chloroethyl chloroformate (ACE-Cl) (56.4 mmol). The reaction is warmed to RT for 1 hour, diluted with methylene chloride (100 mL), washed with NaHCO3 (50 mL), and extracted with methylene chloride (50 mL). The combined organic layers are washed with brine (50 mL), dried and concentrated to give an oily residue which was dissolved in methanol (150 mL) and refluxed for 1 hour. The solvent is removed in vacuo and the crude product is triturated in ether and filtered to give 5,6,7,8-tetrahydro-4iJ-furo[3,2-cT|azepme, which is used without further purification.
[00170] A solution of 5,6,7,8-tetrahydro-4#-furo[3,2-<J]azepine (2.88 mmol) in acetone (7.2 mL) and water (7.2 mL) is treated with NaHCO3 (5.76 mmol) and di-£-butyl dicarbonate (3.17 mmol) for one hour. The reaction is diluted with water (10 mL) and extracted with ethyl acetate (2 x 50 mL). The organic layer is dried, concentrated, and purified by silica chromatography (EtOAc/hexanes eluant) to give tø/t-butyl 4,5,7,8-tetrahydrofuro[3,2- c?]azepine-6-carboxylate.
[00171] A solution of tert-butyl 4,5,7,8-tetrahydrofuro[3,2-cT|azepme-6-carboxylate (0.21 mmol) in chloroform (0.5 mL) and acetic acid (0.5 mL) is treated with N-bromosuccinimide (0.21 mmol) at RT. After 1 hour, the reaction is poured over saturated aqueous NaHCO3, and extracted with ethyl acetate (2 x 5 mL). The organic layers are washed with brine, dried, and purified by silica chromatography (20 % ethyl acetate/hexanes) to give tør/-butyl 2-bromo- 4,5,7,8-tetrahydrofuro[3,2-cT]azepine-6-carboxylate. Example 27. Ethyl 2-bromo-4,5,7,84etrahydrothieno[3,2-d]azepine-6-carboxylate
Figure imgf000150_0001
Figure imgf000150_0003
Figure imgf000150_0002
[00172] To a solution of 2-(thiophen-2-yl)ethanamine (20 g, 157.4 mmol) in CH2Cl2 at 0 0C was added ethyl glyoxylate followed by acetic acid (4 mL). The reaction mixture stirred for 15 minutes followed by the addition OfNaBH(OAc)3 (40 g, 204.7 mmol) in portions. The reaction mixture was stirred for an additional 1 hour and 7 mL of acetic acid was added. The reaction was warmed to RT and stirred until complete consumption of 2-(thiophen-2- yl)ethanamine was observed. The reaction mixture was concentrated in vacuo, the residue taken in THF (500 mL), and the mixture treated with solid NaHCO3 (40 g, 472.2 mmol) at 0 °C. This was followed by addition of ethyl chloroformate (19.5 mL, 157 mmol) and saturated aq. NaHCO3 solution slowly until the gas evolution was minimal. The reaction mixture was stirred overnight and extracted with ethyl acetate. The combined organics were washed with brine solution and concentrated to obtain crude product, which was purified by silica gel chromatography to yield ethyl (ethoxycarbonyl)methyl2-(5-bromothiophen-2- yl)ethylcarbamate (15.0 g, 34% yield); ES-MS: 286.2 (M+H). [00173] To solution of ethyl (ethoxycarbonyl)methyl-2-(5-bromothiophen-2- yl)ethylcarbamate (30.0 g, 105.26 mmol) in ethanol at 0 0C was added dropwise 200 mL of IN NaOH. The reaction mixture was warmed to RT and stirred for 24 hours. The reaction mixture was extracted with Et2O to remove unreacted starting material and the aqueous layer acidified to pH = 1 with IN HCl. The aqueous solution was extracted with ethyl acetate (2 x 500 mL) and the combined organics were washed with brine solution, dried (Na2SO4), filtered, and evaporated to obtain crude product. 2-(JV-(Ethoxycarbonyl)-N-(2-(thiophen-2- yl)ethyl)amino)acetic acid (74% yield)was obtained as a colorless solid after washing the crude product with pentane; ES-MS: 258.2 (M+H).
[00174] 2-(iV-(Ethoxycarbonyl)-iV-(2-(thiophen-2-yl)ethyl)amino)acetic acid (14 g, 54.41 mmol) was dissolved in dry dichloromethane (DCM) (300 mL). To this suspension was added 0.1 mL of DMF, followed by the careful addition of oxayl chloride (10.4 g, 81.93 mmol). The reaction mixture was stirred at room temperature for 1 hour, at which time 0.5 mL of additional oxalyl chloride was added. The solvent was evaporated under vacuum to give 2-(N-(ethoxycarbonyl)-N-(2-(thiophen-2-yl)ethyl)amino)acetyl chloride. This acid chloride was re-dissolved in dry DCM (30OmL) and AlCl3 (18.1 g, 135.74 mmol) was added at room temperature. The reaction was kept at room temperature for 1 hour then quenched by the slow addition of ethanol (about 10 mL). The mixture was then poured into ice and stirred for lhr. The aqueous mixture was extracted with DCM (3 x 150 mL). The combined organic layers were dried over MgSO4, filtered, and evaporated to give a residue, which was purified by silica gel chromatography to produce ethyl 4,5,7,8-tetrahydro-4-oxothieno[3,2- cT]azepine-6-carboxylate (7.4 g, 30.92 mmol).
[00175] A suspension OfAlCl3 (6.7 g, 50.25 mmol) in dry DCM (60 mL) was cooled to 0 0C and BH3-^BuNH2 solid (8.7 g, 100 mmol) was added. After stirring at 0 0C for 5 min, a solution of ethyl 2-(4,5,7,8-tetrahydro-4-oxothieno[3,2-<i]azepm-6-yl)acetate (4 g, 16.72 mmol) in DCM was added. The reaction was stirred at room temperature for 14 hours, The reaction was monitored by TLC and, if necessary, more BH3^zBuNH2 was added to drive the reaction to completion. The mixture was carefully quenched by the addition of 2N HCl (gas evolution observed). When gas evolution stopped, more 2N HCl was added, and the mixture extracted with DCM (3 x 100 mL). The combined DCM layers were dried over MgSO4, filtered, and the filtrate evaporated under vacuum to afford ethyl 4,5,7,8- tetrahydrothieno[3,2-</]azepine-6-carboxylate as a crude white solid that was used directly without purification.
[00176] Ethyl 4,5,7,8-tetrahydrothieno[3,2-d]azepine-6-carboxylate (16.72 mmol) was dissolved in CH3CN (150 mL) and NBS (4.74 g, 26.63 mmol) was added. The reaction was stirred at room temperature for 30 min, and poured into a solution OfNa2SO3 (200 mL)/6N NaOH (5 mL). The aqueous layer was extracted with EtOAc (3 x 150 mL), dried over MgSO4, filtered, and evaporated. The residue was purified by silica gel chromatography to provide ethyl 2-(2-bromo-455,7,8-tetrahydrothieno[3,2-<|azepin-6-yl)acetate (3.1 g, 10.20 mmol).
Example 28. tert-Butyi 3-(4-iodo-lH-pyrazol-l-yl)azetidine-l-carboxylate
Figure imgf000152_0001
[00177] A solution of diphenylmethanamine (16.2 g, 88.5 mmol) and 2- (chloromethyl)oxirane (8.19 g, 88.5 mmol) in dry EtOH was stirred at RT for 48 hours and then heated at reflux for 48 hour. The reaction mixture was concentrated under reduced pressure and the residue was stirred in acetone (300 mL) for 30 min. The resulting off-white solid precipitate was filtered, washed with cold acetone (10OmL), and dried in vacuo to obtain l-benzhydrylazetidin-3-ol as white crystalline solid (11.0 g, 51% yield); LC-MS: 240.3 (M+H).
[00178] A suspension of l-benzhydrylazetidin-3-ol (11.0 g, 40 mmol) and 10% Pd/C (10 g) in ethanol (15OmL) was hydrogenated at 70 psi pressure for 18 hours. The reaction mixture was filtered through short Celite™ plug, washed with EtOH and the combined filtrates were concentrated to obtain crude product as pale yellow viscous liquid. The crude product was washed thoroughly with pet-ether to remove diphenylmethane and azetidin-3-ol was obtained as colorless solid (3.0 g, 90% yield); LC-MS: 74.2 (M+H). [00179] A solution of azetidin-3-ol (5.0 g, 68.5 mmol) and NaHCO3 (34.52 g, 410.9 mmol) in water at RT was treated with (Boc)2O (16.43 g, 75.34 mmol) and the reaction mixture stirred at RT until complete consumption of compound azetidin-3-ol was indicated by TLC (EtOAc/hexanes, 1:1). The reaction mixture was extracted with EtOAc (3 x 5OmL). The combined organics were washed with brine solution (5OmL), dried over Na2SO4, filtered, and concentrated in vacuo to obtain tert-bntyl 3-hydroxyazetidine-l-carboxylate (6.0 g, 51% yield); ES-MS: 172.11 (M+H).
[00180] To a solution of ter/-butyl 3-hydroxyazetidine-l-carboxylate (6.0 g, 34.68 mmol) in DCM was added by Et3N. The mixture was cooled to 0°C and methanesulfonyl chloride was slowly added. After addition was complete, the reaction mixture was warmed to RT and stirred until starting material was completely consumed as indicated by TLC (40% EtOAc/hexanes). The reaction mixture was filtered and CH2Cl2 was added to the filtrate (100 mL). The organics were washed with water (2 x 3OmL), brine solution (3OmL), dried (Na2SO4), filtered, and concentrated to obtain crude product, which was purified by silica gel chromatography (5% EtOAc/hexanes, then 15% EtOAc/hexanes). \-(tert- Butoxycarbonyl)azetidin-3-yl methanesulfonate was isolated as an off-while solid (2.2 g, 36% yield); ES-MS: 252.08 (M+H).
[00181] At 0°C, a solution of 4-iodopyrazole(3.0 g, 15.46 mmol) in DMF (40 mL) was treated with NaH (60% mineral oil dispersion, 1.11 g, 46.39 mmol) in portions. The mixture stirred for 30 minutes, followed by the slow addition of l-(fer?-butoxycarbonyl)azetidin-3-yl methanesulfonate (5.0 g, 20.1 mmol). The reaction mixture was warmed to RT and then heated at 1000C for 5 hours. The reaction mixture was poured into ice cold water, stirred for 30 minutes, and resulting precipitate collected by filtration and dried. The resulting residue was washed with pentane and dried to obtain ifer/-butyl 3-(4-iodo-lH-pyrazol-l-yl)azetidine- 1-carboxylate (3.3 g, 62 % yield); 1H-NMR (400 MHz, CDCl3): 7.58 (s, IH), 7.57 (s, 1 H), 5.07-5.0 (m, 1 H), 4.39-4.34 (m, 2H), 4.29-4.25 (dd, 2 H), 1.45 (s, 9H).
Example 29. endo- and exo- 3-(4-Iodo-pyrazol-l-yl)-8-aza-bicyclo[3.2.1]octane-8- carboxylic acid, tert-butyl ester
Figure imgf000154_0001
exo
[00182] 3-Oxo-8-aza-bicyclo[3.2. ljoctane-δ-carboxylic acid, tert-butyl ester (8 g, 35.5 mmol) was dissolved in 100 mL of ethanol. Sodium borohydride (2 g, 53.5 mmol) was added to the solution portionwise at room temperature. After stirring for 3 hours, the reaction was evaporated in vacuo to give clear viscous oil. The oil was dissolved in dichloromethane, washed with water and brine, dried over anhydrous sodium sulfate, filtered, and evaporated to afford 7.55 g of 3-hydroxy-8-aza-bicyclo[3.2.1]octane-8-carboxylic acid, tert-butyl ester as a white crystalline solid; 1H NMR (300 MHz5 DMSO-d6): 4.23 (dd, J = 2.7, 4.6 Hz, 1 H), 4.18 - 4.06 (m, 2 H), 2.17 - 2.06 (m, 1 H), 1.99 - 1.91 (m, 3 H), 1.72 - 1.50 (m, 5H), 1.47 (s, 9H).
[00183] 3-Hydroxy-8-aza-bicyclo[3.2.1]octane-8-carboxylic acid, tert-butyl ester (7.55 g, 33.2 mmol), triethylamine (5.1 mL, 37 mmol), and 4-dimethylaminopyridine (36 mg, 0.3 mmol) was dissolved in 100 mL of dichloromethane and cooled to 5 0C in an ice bath. Methanesulfonyl chloride (2.6 mL, 33.2 mmol) was added to the solution dropwise and the reaction warmed to room temperature and stirred at room temperature for 18 hours. The reaction was washed with water and brine, dried over anhydrous sodium sulfate, and the solvent removed to afford 3-methanesulfonyloxy-8-aza-bicyclo[3.2.1]octane-8-carboxylic acid, tert-butyl ester as a clear yellow oil (10.2 g); 1H NMR (300 MHz, DMSOd6): 5.09 - 5.01 (m, 1 H), 4.28 (s, 1 H), 4.22 (s, 1 H), 3.01 (s, 3 H), 2.20 - 1.97 (m, 6 H), 1.71 - 1.66 (m, 2 H), 1.46 (s, 9 H). [00184] Sodium hydride (60% in mineral oil) (1.52 g, 38 mmol) was added slowly to a cooled solution (0 0C) of 4-iodopyrazole (6.6 g, 34 mmol) in anhydrous DMF (75 mL). After stirring for 1 hour, a solution of (S-methanesulfonyloxy-S-aza-bicyclop^.lJoctane-S- carboxylic acid, tert-butyl ester (10.2 g, 34 mmol) in 25 mL of anhydrous DMF was added to the reaction. The reaction was heated to 100 0C for 18 hours. The reaction was poured into 50 mL of water and extracted with ethyl acetate. The combined ethyl acetate extracts were washed with water (2 x 50 mL) and brine (2 x 50 mL), dried over anhydrous sodium sulfate, and evaporated to give 12.82 g of product as a mixture of endo and exo isomers; 3.0 g which was purified by medium pressure chromatography (SiO2) eluting with a gradient (hexane- 10% ethyl acetate in hexane) over 30 minutes to afford 1.5 g of the front running spot (endo isomer) and 1.3 g of the 2nd spot {exo isomer); endo isomer - 1H NMR (300 MHz, DMSOd6): 7.58 (s, 1 H), 7.52 (s, 1 H), 7.26 (s, 1 H), 4.34 (q, J = 5.3 Hz, 1 H), 4.27 (s, 2 H), 2.44 (s, 4 H), 1.89 - 1.85 (m, 2 H), 1.60 - 1.53 (m, 2 H), 1.49 (s, 9H); exo isomer - H NMR (300 MHz, DMSOd6): 7.48 (d, J = 0.4 Hz, 1 H), 7.41 (s, 1 H)3 7.26 (s, 1 H), 4.68 (m, 1 H), 4.37 (br s, 2 H), 2.08-2.05 (m, 6 H), 1.79 - 1.75 (m, 2 H), 1.49 (s, 9 H).
Example 30. tert-Bxxty\ 4-(5-bromothiophen-2-yl)piperidine-l-carboxylate
3CΛCHJ
Figure imgf000155_0001
[00185] A solution of 2-bromothiophene in THF (25ml) was added drop wise to a suspension of Mg turnings in THF (100 mL). The mixture was stirred for 30 minutes, cooled to 00C, and a solution of tert-butyl 4-oxopiperidine-l-carboxylate (25.0 g, 125.4 mmol) in THF (25 mL) was added dropwise. The reaction mixture was slowly warmed to RT and stirred for 1 hour until complete consumption of 2-bromothiophene was indicated by TLC. The reaction was quenched with sat. aq. NH4Cl solution (50 mL), extracted with EtOAc (200 mL), washed with H2O (50 mL) and brine solution, dried (Na2SO4), filtered, and evaporated to afford crude product as light brown oil. Purification by silica gel chromatography (25% EtOAc/hexanes) yielded tert-butyl 4-hydroxy-4-(thiophen-2-yl)piperidine-l-carboxylate (14.O g, 40% yield).
[00186] To a solution of tert-butyl 4-hydroxy-4-(thiophen-2-yl)piperidine-l-carboxylate (6.0 g, 21.2 mmol) in 1 ,2-dichloroethane at 0°C was added ZnI2 (10.15 g, 31.8 mmol) in portions. The reaction mixture was stirred for 30 minutes, followed by the addition of NaCNBH3 (2.0 g, 31.8 mmol) in portions. The reaction mixture was slowly warmed to RT and stirred for 2 hours. The reaction was quenched with ice, extracted with CH2Cl2 (2 x 50 mL), washed with brine solution, dried (Na2SO4), filtered, and concentrated to obtain tert- butyl 4-(thiophen-2-yl)piperidine-l-carboxylate, which was used as is for subsequent reactions without further purification.
[00187] N-Bromosuccinimide (4.9 g, 28.08 mmol) was added in portions to a solution of tert-butyl 4-(thiophen-2-yl)piperidine-l-carboxylate (5.0 g, 18.72 mmol) in MeCN at -10 0C. The reaction mixture was stirred for 30 minutes and treated with sat. aq. NaHCO3 solution. The mixture was extracted with CH2Cl2 and the organics washed with water and brine solution, dried (Na2SO4), filtered, and evaporated to obtain crude product. Purification by silica gel chromatography (2% EtOAc/ pet ether) yielded tert-Butyl 4-(5-bromothiophen-2- yl)piperidine-l-carboxylate (1.5 g, 21 % from ter/-butyl 4-hydroxy-4-(thiophen-2- yl)ρiperidine-l-carboxylate); 1H-NMR (400 MHz, CHCl3): 6.85 (d, 1 H), 6.58 (d, 1 H)3 4.22- 4.10 (d, 2H), 3.90-3.78 (m, 3H), 1.98-1.90 (d, 2 H), 1.65-1.48 (m, 2 H), 1.45 (s, 9 H); ES- MS: 346.3, 348.3 (M+H).
Example 31. tert-Butyl 2-bromo-6,7-dihydrothiazolo[5,4-c]pyridine-5(4iϊ)-carboxylate
Figure imgf000156_0001
[00188] To a solution of compound tert-butyl 4-oxopiperidine-l-carboxylate (24.0 g, 120.6 mmol) in dry CHCl3 (500 mL) at 0-50C, was slowly added a solution OfBr2 in CHCl3 (100 mL) over 1.5 hours. The reaction mixture was warmed to RT and stirred for 3 hours. The reaction mixture was concentrated to obtain solid, which was thoroughly washed with Et2O. The resulting intermediate tert-butyl 3-bromo-4-oxopiperidine-l-carboxylate was used immediately in the next reaction. Accordingly, this intermediate (20 g, 72 mmol) was suspended in pentane, filtered, washed with pentane, and the solid so obtained dried in vacuo and dissolved in EtOH (100 mL). Thiourea (5.46 g, 72.0 mmol) was added and the reaction mixture heated for 4 hours at 70-75 0C. The reaction mixture was concentrated to yield a residue, which was treated with aq. Na2COs solution, adjusted to pH = 10, extracted with EtOAc, washed with water, washed with brine solution, dried (Na2SO4), filtered, and concentrated. The residue was washed with pentane to obtain /erϊ-butyl 2-amino-6,7- dihydrothiazolo[5,4-c]pyridine-5(4H)-carboxylate (8.7 g, 28% yield)
100189] tert-BvΛyl 2-amino-6,7-dihydrothiazolo[5,4-c]pyridine-5(4H)-carboxylate (8.5 g, 33.33 mmol) was added dropwise over 2 minutes to a suspension of t-butyl nitrite and CuBr2 mixture in CΗ3CN at 00C. The reaction mixture was slowly warmed to RT over 30 min and stirred for 16 hours at RT. The reaction mixture was concentrated to obtain residue that was dissolved in EtOAc, filtered through a Celite™ plug, which was washed with EtOAc. The organics were washed with water and brine solution, dried (Na2SO4), filtered, and concentrated to obtain crude product as yellow solid. Purification by washings with pet ether and pentane yielded tert-hxxXy\ 2-bromo-6,7-dihydrothiazolo[5,4-c]pyridine-5(4H)- carboxylate (4.1 g, 38% yield); 1H-NMR (400 MHz, CHCl3): 4.56 (s, 2H), 3.73 (t, 2H), 2.85 (brt, 2H), 1.48 (s, 9H).
Example 32. 4-Iodo-l-(2-(pyrrolidin-l-yl)ethyl)-lH-pyrazole
Figure imgf000157_0001
[00190] NaH (60% paraffin oil dispersion, 0.507 g, 21.13 mmol) was added portionwise to a solution of 4-iodopyrazole in DMF at 0°C. The reaction mixture was stirred for 1 hour, treated with solution of l-(2-chloroethyl)pyrrolidine (1.40 g, 10.56 mmol) in DMF, slowly warmed to RT and stirred for additional 16 hours. The reaction mixture was cooled to 00C and ice cold water was added, followed by extraction with EtOAc (2 x 40 mL). The combined organics were washed with water (3 x 40 mL), brine solution (40ml), dried (Na2SO4), filtered, and concentrated to obtain 4-iodo-l-(2-(pyrrolidm-l-yl)ethyl)-l/f- pyrazole, which was used as is for subsequent reactions without further purification; 1H- NMR (400 MHz, CHCl3): 7.60 (IH, s), 7. 52 (IH, s), 7.49 (IH, s), 4.25 (2H, t), 2.95 (2H, s), 2.57 (4H, m), 1.79 (4H, m); ES-MS: 292.0 (M+H).
Example 33. 3 -Bromo- 1 -(2-(pyrrolidin- 1 -yl)ethyl)- lH-pyrrole
Figure imgf000158_0001
[00191] A solution of 1 -(tert-butyldiisopropylsilyl)-l H-pyrrole (1.0 g, 4.48 mmol) in THF was added N-bromosuccinimde at -78 0C. The reaction mixture was slowly warmed to RT and stirred for 24 hours. The reaction mixture was concentrated to obtain dark black residue, which was suspended in CCU (100 mL) and stirred for 15 min and filtered. The filtrate was concentrated to obtain l-(ter^butyldiisopropylsilyl)-3-bromo-lH-pyrrole as oily liquid (1.0 g).
[00192] A solution of 1 -(tert-butyldiisopropylsilyl)-3-bromo- lH-pyrrole in THF at 0°C was treated with tetrabutylammonium fluoride.3H2θ (0.81 g, 2.58 mmol). The reaction mixture was stirred for 1 hour and concentrated in vacuo to obtain 3 -bromo- 1 H-pyrrole as an oil. The product was unstable upon standing and was used as is immediately in the next reaction.
[00193] To a solution of 3-bromo-lH-pyrrole (100 mg, 0.68 mmol) in DMF at 0 0C was added NaH (60% oil dispersion, 81 mg, 2.05 mmol). The reaction mixture was stirred for 1 hour, followed by the addition of l-(2-chloroethyl)pyrrolidine (108 mg, 0.82 mmol). The reaction mixture was for 6 hours at 100 0C. After cooling, the reaction mixture was treated with 20 mL of ice cold water, extracted with EtOAc (3 x 20 mL), washed with brine solution (20 mL), dried (Na2SC»4), filtered, and concentrated under vacuum at 48 0C to obtain crude product. Purification by silica gel chromatography yielded 3-bromo-l-(2-(pyrrolidin-l- yl)ethyl)-l H-pyrrole (50 mg, 31 % yield); 1H-NMR (400 MHz, CHCl3): 6.69 (IH, s), 6.59 (IH, s), 6.12 (IH, s), 3.95 (2H, t), 2.80 (2H, t), 2.51 (4H, m), 1.81 (4H, m). Example 34. l-(4-(4,4,5,5-Tetramethyl-l,3,2-dioxaborolan-2-yl)benzyl)-4-methyIpiperazine
Figure imgf000159_0001
[00194] To a solution of 2-(4-(bromomethyl)phenyl)-4,4,5,5-tetramethyl-l,3,2- dioxaborolane product (1.0 g, 3.37 mmol) in anhydrous ether (30 mL) was added N- methylpiperizine (1.0 g, 10.1 mmol). The solution was stirred at RT for 14 hours and filtered through Celite™. The filtrate was evaporated under vacuum to give a crude product that was used directly without further purification.
Example 35. tert-Butyl 3,4-dihydro-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2H- azepine- 1 (7H)-carboxylate
Figure imgf000159_0002
[00195] To a solution of tert-butyl 4-oxoazepane-l-carboxylate (3.0 g, 14 mmol, 1 eq.) in THF (20 mL) at -78°C was added a IN solution of LiHMDS (15.4 mL, 15.4 mmol, 1.1 eq.) dropwise under nitrogen. The mixture was stirred for 20 minutes, then a solution of (E)A - (?err-butoxycarbonyl)-2,5,6,7-tetrahydro-lH-azepin-4-yl trifluoromethanesulfonate (5.5 g, 15.4 mmol, 1.1 eq.) in THF (10 mL) was added. The mixture was warmed to 0°C and stirred for 3 hours. The reaction was concentrated, diluted with DCM, filtered through neutral alumina and the product was eluted with 9:1 hexanes/EtOAc. Concentration of the eluant in vacuo gave (Z)-tert-bxιtyl 3,4-dihydro-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2H- azepine-l(7H)-carboxylate (3.6 g, 10.4 mmol); 1H NMR (300 MHz, CDCl3): 5.9 (m, IH); 4.05-3.9 (m, 2H); 3.55 (m, 2H); 2.55 (m, 2H); 1.95 (m, 2H); 1.45 (s, 9H). This product was used without further purification and contained up to 33% of the (ϋT)-isomer. [00196] (Z)-?erf-butyl 3,4-dihydro-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2H'- azepine-l(7H)-carboxylate (3.6 g, 10.4 mmol, 1.0 eq.), bis(pinacolato)diboron (3.18 g, 12.5 mmol, 1.2 eq.) and potassium acetate (3.06 g, 21.2 mmol, 3 eq.) were combined and diluted with anhydrous 1,4-dioxane (100 mL). The mixture was degassed under nitrogen for 0.5 hours. PdCl2(PPh3)2 (0.73 g, 1.04 mmol, 0.1 eq.) was added and the mixture degassed with nitrogen for an additional 15 minutes. The reaction was stirred at 8O0C overnight, concentrated, diluted with ethyl acetate and filtered through floricil, which was eluted with 2:1 hexanes:EtOAc. After concentration in vacuo, the residue was purified by silica gel chromatography (EtOAc/hexanes) to provide (Z)-tert-hvι\y\ 3,4-dihydro-5-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)-2H-azepine-l(7H)-carboxylate (0.95 g, 2.95 mmol, 30% yield; 1H NMR (300 MHz, CDCl3): 6.5 (m, IH); 4.07-3.95 (m, 2H); 3.5 (m, 2H); 2.3 (m, 2H); 1.77 (m, 2H); 1.45 (s, 9H); 1.25 (s, 12H).
Example 36. 3-(l-(2,3-Difluorophenyl)-lH-tetrazol-5-yl)-5-(4,4,5,5-tetramethyl-l,3,2- dioxaborol an-2-yl)ρyridin-2-amine
Figure imgf000160_0001
[00197] To 1,4-dioxane (450 mL) was added (15 g, 42.5 mmol), bis(pinacolato)diboron (12.95 g, 51.0 mmol) and potassium acetate (12.5 g, 127.5 mmol). The mixture was degassed with a nitrogen stream for 20 min., followed by the addition of bis(triphenylphosphine)palladium(II) dichloride (3.0 g, 4.3 mmol). The reaction was stirred at 80 0C for 14 hours under inert atmosphere. After cooling to room temperature, the mixture was concentrated under reduced pressure and the resulting crude solid treated with ethyl acetate (~ 250 mL) and filtered. The filtrate was concentrated, treated with 50% ethyl acetate in hexanes (~ 200 mL), and the resulting precipitate was filtered and dried to yield 9.64 g of3-(l-(2,3-difluorophenyl)-lH-tetrazol-5-yl)-5-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)pyridin-2 -amine as a dark brown solid. The filtrate was concentrated again, treated with hexanes (200 mL), and filtered to provide an additional 6.2 g of product (93% overall yield) as a yellow solid: 1H NMR (CDCl3) δ 7.54-7.32 (m, 5H), 6.71 (s, 2H), 1.24 (s, 12H); ES-MS: m/e = 318 (M - 82)+. Using the same procedure, 3-(l-(2,3- chlorophenyl)-lH-tetrazol-5-yl)-5-(4)4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyridin-2- amine and 3-(l -(3-chloro-2-fluorophenyl)-lH-tetrazol-5-yl)-5-(4,4,5,5-tetramethyl-l ,3,2- dioxaborolan-2-yl)pyridin-2-amine can be produced from 5-bromo-3-(l-(2,3-chlorophenyl)- lH-tetrazol-5-yl)pyridin-2-amine and 5-bromo-3-(l-(3-chloro-2-fluorophenyl)-lH-tetrazol-5- yl)pyridin-2-amine, respectively.
Example 37. 3-(l-(2,3-Difluorophenyl)-lH-tetrazol-5-yl)-5-(4,5,6,7-tetrahydrothieno[2,3- c]pyridin-2-yl)pyridin-2-amine (compound I-A-492)
Figure imgf000161_0001
[00198] A mixture of 3-(l-(2,3-difluorophenyl)-lH-tetrazol-5-yl)-5-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)pyridin-2-amine (250 mg, 0.63 mmol), tert-buty\ 2-bromo-4,5- dihydrothieno[2,3-e]pyridine-6(7H)-carboxylate (198 mg, 0.63 mmol), and NaHCO3 (1.56 mL, saturated solution in H2O) in 7V,iV-dimethylforrnamide (8.3 mL) was degassed with a nitrogen stream for 20 min. To this mixture was added [l,l'-bis(diphenylphosphino)- ferrocenejpalladium (II) dichloride (46 mg, 0.06 mmol) and the reaction was stirred in a microwave for 10 min at 120 0C. The resulting crude mixture was diluted with ethyl acetate (30 mL) and filtered. The filtrate was washed with H2O (2 x 15 mL) and brine (1 x 15 mL), concentrated under reduced pressure, and purified via silica gel chromatography to provide ten-butyl 2-(6-amino-5-(l-(253-difluorophenyl)-lHr-tetrazol-5-yl)pyridin-3-yl)-4,5- dihydrothieno[2,3-c]pyridine-6(7H)-carboxylate. This material (137 mg, 0.27 mmol) was treated with HCl (4 mL, 4.0 N in dioxane) for 1 hr, and solvent was removed under reduced pressure. The residue was dissolved in MeOH (500 mL) and treated with cold diethyl ether (~ 15 mL). The resulting precipitate was collected and dried in vacuo to provide the hydrochloride salt of 3-(l-(2,3-difluorophenyl)-lH-tetrazol-5-yl)-5-(4,5,6,7- tetrahydrothieno[2,3-c]pyridin-2-yl)pyridin-2-amine as a bright yellow solid (103 mg, 37% yield over two steps); 1H NMR (DMSOd6): 9.50 (s, 2H), 8.44 (d, J= 2.4 Hz, IH), 7.85-7.46 (m, 4H)5 7.05 (s, IH), 4.29 (s, 2H), 3.39-3.25 (m, 2H), 2.85-2.81 (m, 2H); ES-MS: m/e = 412.2 (M+H).
Example 38. 3-(l-(2,3-Difluorophenyl)-l-Ef-tetrazol-5-yl)-5-(5,6,7,8-tetrahydro-4iϊ- thieno[3,2-έ/]azepin-2-yl)pyridin-2-amine (compound I-A-430)
Figure imgf000162_0001
[00199] A mixture of 3-(l -(2,3-difluorophenyl)-l H-tetrazol-5-yl)-5-(4 ,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)pyridin-2-amine (2.50 g, 6.3 mmol), ethyl 2-bromo-4,5,7,8- tetrahydrothieno[3,2-d]azepine-6-carboxylate (1.91 g, 06.3 mmol), and NaHCO3 (15.6 mL, saturated solution in H2O) in N,iV-dimethylformamide (83 mL) was degassed with a nitrogen stream for 20 min. To this mixture was added [1,1 '- bis(diphenylphosphino)ferrocene]palladium (II) dichloride (460 mg, 0.6 mmol) and the reaction was stirred in a microwave for 10 min at 120 0C. The resulting crude mixture was diluted with ethyl acetate (300 mL) and filtered. The filtrate was washed with H2O (2 x 150 mL) and brine (1 x 150 mL), concentrated under reduced pressure, and purified via silica gel chromatography to provide ethyl 2-(6-amino-5-(l-(2,3-difluorophenyl)-li/-tetrazol-5- yl)pyridin-3-yl)-4,5,7,8-tetrahydrothieno[3,2-</]azepine-6-carboxylate. [00200] Ethyl 2-(6-amino-5-(l -(2,3-difluorophenyl)-l//-tetrazol-5-yl)pyridin-3-yl)- 4,5,7,8-tetrahydrothieno[3,2-</jazepine-6-carboxylate (2 g, 4.02 mmol) was dissolved in dry CHCl3 (40 mL), followed by the addition of TMS-I (3.2 g, 16.0 mmol) at room temperature. The mixture was heated under reflux for 14 hours. After cooling, the reaction was carefully quenched by the addition of MeOH (10 mL), followed by IN NaOH (30 mL). The mixture was stirred for 30 minutes and then extracted with DCM. The combined DCM layers were dried over MgSO4 and evaporated. The residue was taken up in EtOAc (10 mL) and 4M HCl/dioxane solution was added. Isolation of the yellow precipitate provided 3-(l-(2,3- difluorophenyl)-l/-r-tetrazol-5-yl)-5-(5,6,7,8-tetrahydro-4H-thieno[3,2-ώrjazepin-2-yl)pyridin- 2-amine as the HCl salt (1.8 g, 3.9 mmol).
Example 39. tert-Bntyl 3-(6-amino-5-(l-(2,3-difluorophenyl)-lHr-tetrazol-5-yl)pyridin-3-yl)- 8-azabicyclo[3.2.1 ]oct-3-ene-8-carboxylate
Figure imgf000163_0001
[00201] A solution of tert-bvXyl 3-(trifluoromethylsulfonyloxy)-8-azabicyclo[3.2.1]oct-3- ene-8-carboxylate (50 mg, 140 μmol, 1.0 eq.) in DME (2 mL) was degassed with nitrogen for 15 minutes. NaHCO3 (1.2 mM solution, 350 μL, 420 μmol, 3 eq.) and 3-(l-(2,3- difluorophenyl)- l_H-tetrazol-5-yl)-5-(4,4,5.,5-tetramethyl-l ,3 ,2-dioxaborolan-2-yl)pyridin-2- amine (56 mg, 140 μmol, 1.0 eq.) were added and degassing was continued for an additional 30 minutes. PdCl2(dppf>2 (10 mg, 14 μmol, 0.1 eq.) was added and degassing was continued for an additional 15 minutes. The reaction was sealed and microwaved at 90 0C for 15 minutes. After cooling, the organic layer was concentrated, NaHCO3 was added, and the product was extracted with EtOAc. Silica gel chromatography (EtOAc/hexanes) provided /erf-butyl 3-(6-amino-5-(l-(2,3-difluorophenyl)-l//-tetrazol-5-yl)pyridin-3-yl)-8- azabicyclo[3.2.1]oct-3-ene-8-carboxylate (34 mg, 51% yield); 1H NMR (300 MHz, CDCl3): 8.25 (m, IH); 7.6-7.25 (m, 3H); 7.07 (s, IH); 6.4 (br s, 2H); 6.0 (m, IH); 4.38 (m, 2H); 2.65 (m, IH); 2.15 (m, IH); 2.14-1.5 (m, 4H); 1.44 (s, 9H). Example 40. l-^-S-Cό-Amino-S-Cl^^-difluorophenyO-lH-tetrazol-S-yOpyridin-S-yl)- 3,4-dihydro-2H-azepin-l(7H)-yl)-2,2,2-trifluoroethanone
Figure imgf000164_0001
[00202]
Figure imgf000164_0002
3,4-dihydro-5-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)-2H- azepine-l(7H)-carboxylate (510 mg, 1.44 mmol, 1.1 eq.) was dissolved in DME (14 mL) and degassed with nitrogen for 15 minutes. NaHCO3 (1.2 M solution, 3.25 mL, 3.9 mmol, 3 eq.) and 5-bromo-3-(l-(2,3-difluoroρhenyl)-lH-tetrazol-5-yl)pyridin-2 -amine (420 mg, 1.3 mmol, 1 eq.) were added and the mixture degassed with nitrogen for an additional 30 minutes. PdCl2(dppf)2 (95 mg, 0.13 mmol, 0.1 eq.) was added and the mixture degassed with nitrogen for an additional 15 minutes. The reaction was microwaved in a sealed tube for 20 min at 900C. To drive the reaction to completion, additional PdCl2(dppfh (95 mg, 0.13 mmol, 0.1 eq.) was added under an atmosphere of nitrogen and the reaction microwaved for an additional 20 min at 90 0C. The mixture was concentrated, NaHCO3 was added, and the product was extracted with ethyl acetate. The organics were filtered through floricil and purified by silica gel chromatography (EtOAc/hexanes) to provide (E)-tert-bvXy\ 5-(6-amino- 5-(l-(2,3-difluorophenyl)-l-H-tetrazol-5-yl)pyridin-3-yl)-3,4-dihydro-2H-a2epine-l(7H)- carboxylate (360 mg, 0.76 mmol, 53% yield); 1H NMR (300 MHz, CDCI3): 8.2 (m, IH); 7.5 (m, IH); 7.45-7.3 (m, 2H); 7.02 (s, IH); 6.4 (br s, 2H); 5.6 (m, IH); 3.8 (m, 2H); 3.55 (m, 2H); 2.2 (m, 2H); 1.7 (m, 2H) 1.45 (s, 9H).
[00203] (£)-ten-Butyl 5-(6-amino-5-(l-(2,3-difluorophenyl)-l/f-tetrazol-5-yl)pyridin-3- yl)-3,4-dihydro-2H-azepine-l(77ϊ)-carboxylate (14 mg, 29.9 μmol, 1 eq.) was diluted in 1:1 TFA:DCM (1.5 mL) and the mixture stirred at room temperature for 2 hours. The reaction was concentrated and purified by reversed-phase HPLC purification to give 3-(l-(2,3- difluorophenyl)-lH-tetrazol-5-yl)-5-((£)-2,5,6,7-tetrahydro-liy-azepin-4-yl)pyridin-2-amine in quantitative yield; 1H NMR (300 MHz, DMSO-d6): 9.0 (br s, 2H); 8.25 (m, IH); 7.87-7.77 (m, IH); 7.75-7.6 (m, 2H); 7.6-4.5 (m, IH); 5.85 (m, IH); 3.7 (m, 2H); 3.25 (m,2H); 2.5 (m, 2H); 1.78 (m, 2H).
[00204] 3-(l-(2,3-Difluorophenyl)-lJ:-'-tetrazol-5-yl)-5-((£1)-255)6,7-tetrahydro-lHr-azepin- 4-yl)pyridin-2-amine (37 rag, 100 μmol, 1 eq.) was diluted in DCM (1.5 mL). EDC (34.4 mg, 150 μmol, 1.5 eq.), trifluoroacetic acid (17.1 mg, 150 μmol, 1 eq.), and DIEA (53 μL, 300 μmol, 3 eq.) were added. The reaction was stirred at room temperature overnight then concentrated. The product was purified by reversed-phase HPLC purification to give 1-((E)- 5-(6-amino-5-(l-(2,3-difluorophenyl)-l-H-tetrazol-5-yl)pyridin-3-yl)-3,4-dihydro-2H-azepin- l(7H)-yl)-2,2,2-trifluoroethanone (13.6 mg, 30% yield); 1H NMR (300 MHz, DMSO-d6): 8.25 (m, IH); 7.87 (m, IH); 7.77 (m, IH); 7.55 (m, IH); 7.37 (m, IH); 5.95-5.7 (m, IH); 4.12 (m, 2H); 3.75-3.6 (m, 2H); 2.4 (m, 2H); 1.75 (m, 2H).
Example 41. 3-(l-(2,3-Difluoroρhenyl)-lH-tetrazol-5-yl)-5-(2-(piperidin-4-yl)thiazol-5- yl)pyridin-2-amine (compound I-A-549)
Figure imgf000165_0001
[00205] Using the same protocol used to prepare compound I-A-492, 3-(l-(2,3- difluorophenyl)-lHr-tetrazol-5-yl)-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyridin-2- amine and tert-butyl 4-(5-bromothiazol-2-yl)piperidine-l -carboxylate were reacted together in a PdCl2(dppf)2-mediated coupling to produce 3-(l-(2,3-difluorophenyl)-lΗ-tetrazol-5-yl)- 5-(2-(piperidin-4-yl)thiazol-5-yl)pyridin-2-amine; 1H NMR (DMSO-d6): 8.46 (d, J= 2.3 Hz, IH), 7.90-7.48 (m, 5H), 3.40-3.22 (m, 3H), 3.11-2.92 (m, 2H), 2.22-2.10 (m, 2H), 1.99-1.83 (m, 2H); ES- MS: m/e = 441.1 (M+H). Example 42. 3-(l -(2,3-Difluorophenyl)-l/-r-tetxazol-5-yl)-5-(4-((4-methylpiperazin-l - yl)methyl)phenyl)pyridin-2-amine (compound I-A-62)
Figure imgf000166_0001
[00206] Using the same protocol used to prepare compound I-A-492, 5-bromo-3-(l-(2,3- difluorophenyl)-lH-tetrazol-5-yl)pyridin-2-amine and l-(4-(4,4,5,5-tetramethyl-l ,3,2- dioxaborolan-2-yl)benzyl)-4-methylpiperazine were reacted together in a PdCl2(dppf)2- mediated coupling to produce 3-(l-(2,3-difluorophenyl)-lH-tetrazol-5-yl)-5-(2-(piperidin-4- yl)thiazol-5-yl)pyridin-2-amine; 1H NMR (DMSOd6): 8.46 (d, J= 2.3 Hz, IH)3 7.90-7.48 (m, 5H), 3.40-3.22 (m, 3H), 3.11-2.92 (m, 2H), 2.22-2.10 (m, 2H), 1.99-1.83 (m, 2H); ES- MS: m/e = 441.1 (M+H).
Example 43. 3-(l -(2,3-Difluorophenyl)-l#-tetrazol-5-yl)-5-(l -(piperidin-4-yl)-lH- 1 ,2,3- triazol-4-yl)pyridin-2-amine (compound I-A-483)
Figure imgf000166_0002
[00207] In a tube was placed N,N-di(l , 1 -dimethylethoxycarbonyl)-3~(l -(2,3- difluorophenyl)-lH-tetrazol-5-yl)-5-bromopyridin-2 -amine (2.3 g, 4.1 mmol) in diethylamine (10 mL) with ethynyltrimethylsilane (1.5 g, 15.3 mmol) and the reaction was deoxygenated with a stream of nitrogen gas. To the mixture was added copper(I) iodide (554 mg, 2.9 mmol). The reaction vessel was sealed and warmed to 500C to achieve dissolution. To the mixture was added PdCl2dρpf2»CH2Cl2 (190 mg, 0.06eq). The reaction vessel was sealed and the mixture sonicated at 5O0C for 10 minutes. The reaction was diluted with methylene chloride, filtered through Celite™, concentrated, and purified by silica gel chromatography (10% ethyl acetate/hexane to 50% ethyl acetate/hexane) to give JV,N-di( 1,1- dirnethylethoxycarbonyl)-3 -( 1 -(2,3 -difluorophenyl)- 1 H-tetrazol-5-yl)-5-(2- (trimethylsilyl)ethynyl)pyridin-2-amine (713 mg, 1.2 mmol, 30%) as a yellow solid; LC-MS: m/e = All .3 [M-Boc+Η].
[00208] NJV-Di(I, l-dimethylethoxycarbonyl)-3-(l-(2,3-difluorophenyl)-lH-tetrazol-5-yl)- 5-(2-(trimethylsilyl)ethynyl)pyridin-2-amine (713 mg, 1.2 mmol) was dissolved in methylene chloride (10 mL) and cooled to 00C. To this was added tetrabutylammonium fluoride hydrate (163 mg, 625 μmol) in methylene chloride (1 mL). The reaction was loaded directly onto silica and chromatographed (10% ethyl acetate/hexane to 50% ethyl acetate/hexane) to give NJV-di(l,l-dimethylethoxycarbonyl)-3-(l-(2,3-difluorophenyl)-lH-tetrazol-5-yl)-5- ethynylpyridin-2-amine (496 mg, 1.0 mmol, 83%) as a yellow solid; 1H ΝMR (CDCl3): 1.37(s, 18H), 3.35 (s, IH), 7.30 (m, IH), 7.40 (m, 2H), 7.89 (s, IH), 8.75 (s, IH). [00209] N3N-Di(I, l-dimethylethoxycarbonyl)-3-(l-(2,3-difluorophenyl)-lH-tetrazol-5-yl)- 5-efhynylρyridin-2-amine (59 mg, 119 μmol) was dissolved in methylene chloride (1 mL) and to this was added tris((l-benzyl-l#-l,2,3-triazol-4-yl)rnethyl)amine (TBTA, 20 mg, 38 μmol, see Henning et al., Organic Letters 9(1): 1-4; 2007), sodium ascorbate (100 mg, 505 μmol), water (1 mL), and methanol (2 mL). To the reaction mixture was added tert-buty\ A- azidopiperidine-1-carboxylate (1 mmol) (prepared from tert-butyl 4-aminopiperidine-l- carboxylate, via the method described by Alper et al., Tetrahedron Letters 37(34): 6029- 6032, 1996) in methylene chloride (2 mL). The reaction was left at room temperature for 30 minutes and concentrated to remove the volatile organics. The reaction was extracted with methylene chloride (2 x 10 mL) and saturated sodium bicarbonate (5 mL). The organics were dried over sodium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography (CH2Cl2 to 8% MeOH/ CH2Cl2) to give tert-butyl 4-(4-(6-NJV-di(l,l- dimethylethoxycarbonyl)amino-5-(l-(2,3-difluorophenyl)-lH-tetrazol-5-yl)pyridin-3-yl)-lϋ/:- l,2,3-triazol-l-yl)piperidine-l-carboxylate (57 mg, 79 μmol, 66% yield); 1H NMR (CDCl3): 1.37(s, 18H), 3.35 (s, IH), 7.30 (m, IH), 7.40 (m, 2H), 7.89 (s, IH), 8.75 (s, IH). LC-MS: m/e = 625.4 [M-Boc+H].
[00210] tert-Buiy\ 4-(4-(6-JVrΛT-di(l ,l~dimethylethoxycarbonyl)amino-5-(l-(2,3- difluorophenyl)-l//-tetrazol-5-yl)pyridin-3-yl)- IAT- 1 ,2,3-triazol- 1 -yl)piperidine-l - carboxylate (57 mg, 79 μmol) was treated with 4N hydrochloric acid in dioxane (1 mL) at room temperature, overnight. The reaction was concentrated and purified via reversed-phase HPLC (10% to 40% acetonitrile water with 0.1% TFA, over 10 minutes) to give 3-(l-(2,3- difluorophenyl)-lH4etrazol-5-yl)-5-(l-(piperidin-4-yl)-lH-l,2,3-triazol-4-yl)pyridin-2- amine (20.7 mg, 27 μmol, 34% yield); 1H NMR methanol-d4: 8.55 (s, IH); 8.3 (s, IH); 8.0 (s, IH); 7.6 (m, 2H); 7.5 (m, IH); 3.6 (m, 2H); 3.2 (m, 3H); 2.4 (m, 2H); LC-MS: m/e = 425.2 [M+H].
Table 6. Analytical Chararacterization Data for Compounds of Formula I-A (blank cells indicate that the test was not performed)
Figure imgf000168_0001
Figure imgf000169_0001
Figure imgf000170_0001
Figure imgf000171_0001
Figure imgf000172_0001
Figure imgf000173_0001
Figure imgf000174_0001
Figure imgf000175_0001
Figure imgf000176_0001
Figure imgf000177_0001
Figure imgf000178_0001
Figure imgf000179_0001
Figure imgf000180_0001
Figure imgf000181_0001
Figure imgf000182_0001
Figure imgf000183_0001
Figure imgf000184_0001
Figure imgf000185_0001
Figure imgf000186_0001
Figure imgf000187_0001
Figure imgf000188_0001
Figure imgf000189_0001
Figure imgf000190_0001
Figure imgf000191_0001
Figure imgf000192_0001
Figure imgf000193_0001
Figure imgf000194_0001
Figure imgf000195_0001
Figure imgf000196_0001
Figure imgf000197_0001
Figure imgf000198_0001
Figure imgf000199_0001
Figure imgf000200_0001
Figure imgf000201_0001
Figure imgf000202_0001
Figure imgf000203_0001
Figure imgf000204_0001
Figure imgf000205_0001
Figure imgf000206_0001
Figure imgf000207_0001
Figure imgf000208_0001
Figure imgf000209_0001
Figure imgf000210_0001
Figure imgf000211_0001
Figure imgf000212_0001
Figure imgf000213_0001
Figure imgf000214_0001
Figure imgf000215_0001
Figure imgf000216_0001
Figure imgf000217_0001
Figure imgf000218_0001
Figure imgf000219_0001
Figure imgf000220_0001
Figure imgf000221_0001
Figure imgf000222_0001
Figure imgf000223_0001
Figure imgf000224_0001
Figure imgf000225_0001
Figure imgf000226_0001
Figure imgf000227_0001
Figure imgf000228_0001
Table 7. Analytical Chararacterization Data for Compounds of Formula I-B
Figure imgf000228_0002
Figure imgf000229_0001
Figure imgf000230_0001
Table 8. Analytical Chararacterization Data for Compounds of Formula I-C (blank cells indicate that the test was not performed)
Figure imgf000230_0002
Figure imgf000231_0001
Table 9. Analytical Chararacterization Data for Compounds of Formula I-D (blank cells indicate that the test was not performed)
Figure imgf000231_0002
Figure imgf000232_0001
Figure imgf000233_0001
Table 10. Analytical Chararacterization Data for Compounds of Formula I-E
Figure imgf000234_0001
Biological assay of compounds of the invention Example 44. Kj Determination for the Inhibition of c-MET
10002] Compounds of the invention were screened for their ability to inhibit c-MET kinase activity using a standard radiometric assay. Briefly, in this kinase assay the transfer of the terminal P-phosphate in P-ATP to substrate polyE4Y is interrogated. The assay was carried out in 96-well plates to a final volume of 100 μL per well containing 1.0 nM c-Met, 100 mM HEPES (pH 7.5), 10 mM MgCl2, 25 mM NaCl, 0.01% BSA, 1 mM DTT, 0.5 mg/mL polyE4Y, and 35 μM ATP. Accordingly, compounds of the invention were dissolved in DMSO to make 10 mM initial stock solutions. Serial dilutions in DMSO were then made to obtain the final solutions for the assay. A 1.5 μL aliquot of DMSO or inhibitor in DMSO was added to each well. The reaction was initiated by the addition Of33P-ATP and polyE4Y (obtained from Sigma). After 20 min, the reaction was quenched with 50 μL of 30% trichloroacetic acid (TCA) containing 4 mM ATP. The reaction mixture was transferred to the 0.66 mm GF filter plates (Corning) and washed three times with 5% TCA. Following the addition of 50 μL of Ultimate Gold™ high efficiency scintillant (Packard Bioscience), the samples were counted in a Packard TopCoimt NXT Microplate Scintillation and Luminescence Counter (Packard BioScience). The Kj values were calculated using Microsoft Excel Solver macros to fit the data to the kinetic model for competitive tight- binding inhibition.
[00211] Compounds having a Ki of less than or equal to 0.10 μM for the inhibition of c- MET include: I-A-5, 1-A-6, 1-A-7, 1-A-8, 1-A-9, 1-A-10, 1- A-11, I-A-12, 1-A-13, 1-A-19, I- A-22, 1-A-32, 1-A-46, 1-A-48, 1-A-50, I-A-51, I-A-52, 1-A-54, I-A-56, 1-A-57, 1-A-62, I-A- 63, 1-A-64, 1-A-65, l-A-66, l-A-67, I-A-79, I-A-81, 1-A-85, I-A-86, I-A-87, l-A-90, I-A-92, I-A-94, 1-A-95, I-A-96, 1-A-97, 1-A-98, 1-A-99, ϊ-A-100, 1-A-104, 1-A-106, I-A-108, 1-A- 109, 1-A-l 18, 1-A-l 19, 1-A-120, 1-A-121, 1-A-122, I-A-123, 1-A-124, 1-A-125, 1-A-126, I- A-127, 1-A-128, 1-A-129, 1-A-130, 1-A-131, 1-A-132, 1-A-133, 1-A-134, 1-A-135, 1-A-136, I-A-137, I-A-138, 1-A-139, 1-A-141, 1-A-143, 1-A-144, 1-A-145, 1-A-146, 1-A-147, 1-A- 148, 1-A-149, 1-A-150, 1-A-151, 1-A-152, 1-A-153, 1-A-156, 1-A-157, 1-A-158, 1-A-159, I- A-165, 1-A-180, 1-A-181, 1-A-182, 1-A-183, 1-A-184, 1-A-186, 1-A-187, 1-A-188, 1-A-189, I-A-190, 1-A-191, 1-A-192, 1-A-193, 1-A-197, 1-A-198, 1-A-199, 1-A-200, 1-A-201, 1-A- 202, 1-A-203, 1-A-204, I-A-206, ϊ-A-207, l-A-208, 1-A-209, 1-A-210, 1-A-211, 1-A-212, I- A-213, 1-A-218, 1-A-219, 1-A-220. 1-A-221, 1-A-222, 1-A-223, 1-A-231, 1-A-232, 1-A-233, I-A-234, 1-A-235, 1-A-239, I-A-242, I-A-245, 1-A-247, 1-A-249, 1-A-250, 1-A-251, 1-A- 254, 1-A-256, 1-A-257, 1-A-258, 1-A-259, 1-A-261, I-A-262, 1-A-263, 1-A-264, 1-A-265, I- A-266, 1-A-267, 1-A-268, 1-A-272, 1-A-273, 1-A-276, 1-A-277, 1-A-280, 1-A-281, 1-A-283, I-A-286, 1-A-287, 1-A-290, 1-A-292, 1-A-295, 1-A-298, 1-A-299, 1-A-303, 1-A-305, I-A- 308, 1-A-309, 1-A-312, 1-A-313, 1-A-314, T-A-321 , 1-A-325, 1-A-328, 1-A-329, 1-A-331, I- A-332, 1-A-334, 1-A-335, ϊ-A-337, 1-A-338, 1-A-339, 1-A-340, I-A-341 , T-A-342, 1-A-343, I-A-344, 1-A-345, 1-A-346, 1-A-347, 1-A-348, 1-A-349, I-A-351, 1-A-354, 1-A-355, I-A- 360, 1-A-363, 1-A-366, 1-A-367, 1-A-368, 1-A-370, 1-A-371, 1-A-372, 1-A-376, 1-A-377, I- A-380, 1-A-381, 1-A-382, 1-A-385, 1-A-391, 1-A-393, 1-A-394, 1-A-396, 1-A-397, 1-A-398, I-A-399, 1-A-400, 1-A-401, 1-A-403, 1-A-404, 1-A-405, I-A-406, 1-A-407, 1-A-408, 1-A- 409, 1-A-410, 1-A-411, l-A-412, l-A-413, l-A-414, 1-A-415, 1-A-416, l-A-417. l-A-418, 1- A-419, 1-A-420, 1-A-422, 1-A-423, 1-A-425, 1-A-426, 1-A-427, 1-A-428, 1-A-429, 1-A-430, I-A-431, 1-A-432, 1-A-433, 1-A-434, 1-A-435, 1-A-436, 1-A-438, 1-A-440, 1-A-441, 1-A- 442, 1-A-443, 1-A-444, 1-A-445, 1-A-446, 1-A-447, 1-A-448, 1-A-449, 1-A-450, 1-A-451, 1- A-452, 1-A-453, 1-A-455, 1-A-456, 1-A-457, 1-A-458, 1-A-459, 1-A-460, 1-A-461, 1-A-462, I-A-463, 1-A-464, 1-A-466, 1-A-468, 1-A-469, 1-A-470, 1-A-471, 1-A-472, 1-A-473, 1-A- 474, I-A-475, 1-A-476, I-A-477, I-A-480, 1-A-481, 1-A-482, 1-A-483, I-A-484, 1-A-485, 1- A-486, I-A-487, I-A-491, 1-A-492, 1-A-497, 1-A-499, I-A-500, 1-A-501, 1-A-502, 1-A-503, I-A-504, 1-A-505, 1-A-506, I-A-507, 1-A-508, 1-A-510, 1-A-511, 1-A-512, 1-A-513, 1-A- 514, I-A-515, 1-A-516, l-A-518, 1-A-519, 1-A-520, I-A-521, l-A-522, l-A-523, 1-A-524, 1- A-526, 1-A-527, 1-A-528, 1-A-529, 1-A-530, 1-A-531, 1-A-532, 1-A-534, 1-A-535, 1-A-536, I-A-538, 1-A-540, 1-A-542, 1-A-584, 1-A-589, 1-A-599, 1-A-601, l-A-615, 1-A-616, 1-A- 619, 1-A-620, 1-A-622, 1-A-623, I-A-624, 1-A-626, 1-A-628, 1-A-629, 1-A-630, 1-A-631, I- A-635, 1-A-640, 1-A-642, I-A-650, 1-A-651, 1-A-652, 1-A-653, 1-A-655, 1-A-656, I-A-657, I-A-658, T-A-659, 1-A-660, 1-B-22, 1-C-3, 1-C-5, 1-C-6, 1-D-l, I-D-2, 1-D-3, 1-D-4, 1-D-5, 1- D-6, 1-D-7, I-D-8, I-D-9, I-D-10, 1-D-l 1, I-D-12, I-D-13, I-D-19, and I-D-23. [00212] Compounds having a Kj of greater than 0.10 μM and less than or equal to 1.0 μM the inhibition of c-MET include I-A-2, 1-A-3, 1-A-4, 1-A-15, I-A-16, 1-A-17, 1-A-18, 1-A-20, I-A-29, 1-A-30, 1-A-31, 1-A-33, 1-A-34, 1-A-39, 1-A-40, 1-A-41, 1-A-42, 1-A-53, 1-A-55, 1- A-68, 1-A-69, 1-A-70, 1-A-71, 1-A-72, 1-A-73, 1-A-74, 1-A-75, 1-A-76, 1-A-77, 1-A-78, 1-A- 82, 1-A-83, 1-A-84, 1-A-88, 1-A-89, 1-A-93, 1-A-101, 1-A-102, 1-A-103, 1-A-105, 1-A-107, 1- A-110, ϊ-A-l li; I-A-112, 1-A-l 13, 1-A-114, 1-A-116, 1-A-140, 1-A-142, 1-A-154, 1-A-155, I-A-160, 1-A-161, 1- A-162, 1-A-163, 1-A-179, 1-A-185, 1-A-194, 1-A-195, 1-A-196, 1-A- 205, 1-A-215, 1-A-224, 1-A-225, 1-A-228, 1-A-229, 1-A-236, 1-A-237, 1-A-240, 1-A-241, 1- A-248, 1-A-252, 1-A-255, 1-A-260, 1-A-269, l-A-270, 1-A-271, 1-A-274, 1-A-275, l-A-278, I-A-279, 1-A-282, 1-A-285, 1-A-288, 1-A-289, l-A-291, l-A-294, 1-A-296, l-A-301, 1-A- 304, 1-A-306, 1-A-310, T-A-315, T-A-316, 1-A-317, 1-A-318, T-A-319, 1-A-320, 1-A-324, 1- A-326, 1-A-327, 1-A-330, 1-A-333, 1-A-336, 1-A-350, 1-A-352, 1-A-353, 1-A-356, 1-A-357, I-A-358, 1-A-359, 1-A-362, 1-A-364, 1-A-365, 1-A-369, 1-A-383, 1-A-386, 1-A-387, 1-A- 388, 1-A-389, 1-A-390, 1-A-392, 1-A-395, 1-A-402, 1-A-421 , 1-A-424, 1-A-439, 1-A-454, 1- A-465, 1-A-467, 1-A-478, 1-A-479, 1-A-488, 1-A-490, 1-A-493, 1-A-494, 1-A-495, 1-A-496, I-A-509, 1-A-517, 1-A-525, 1-A-533, 1-A-537, 1-A-539, 1-A-541, 1-A-551, 1-A-554, 1-A- 557, 1-A-561, 1-A-562, 1-A-564, 1-A-565, 1-A-567, 1-A-568, 1-A-569, 1-A-570, 1-A-574, 1- A-575, 1-A-576, 1-A-577, 1-A-578, 1-A-579, 1-A-580, 1-A-581, 1-A-582, 1-A-583, 1-A-585, I-A-587, 1-A-588, 1-A-590, 1-A-591, 1-A-592, 1-A-593, 1-A-594, 1-A-595, 1-A-596, 1-A- 597, 1-A-598, 1-A-600, 1-A-602, 1-A-603, 1-A-604, 1-A-605, 1-A-606, 1-A-607, 1-A-608, 1- A-609, ϊ-A-610, 1-A-611, ϊ-A-612, 1-A-613, l-A-614, l-A-617, 1-A-618, l-A-621, l-A-625, I-A-627, 1-A-632, 1-A-633, 1-A-634, 1-A-636, I-A-637, I-A-638, 1-A-639, 1-A-641, 1-A- 643, 1-A-644, T-A-646, 1-A-647, 1-A-649, I-A-654, 1-B-l 1, 1-B-14, 1-B-16, 1-B-20, 1-B-21, I-C-l, I-C-4, 1-C-8, 1-C-ll, I-D-14, I-D-15, 1-D-16, 1-D-17, I-D-18, I-D-20, 1-D-21, 1-D- 22, 1-E-l , I-E-2, I-E-3, 1-E-4, and I-E-5.
[00213] All publications and patents cited in this specification are herein incorporated by reference as if each individual publication or patent were specifically and individually indicated to be incorporated by reference. Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be readily apparent to those of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope of the appended claims.

Claims

CLAIMS What is claimed is:
1. A compound having the formula:
Figure imgf000238_0001
(I), or a pharmaceutically acceptable salt thereof, wherein:
Z1 is N or CR4;
Z2 is N or CH;
Ring D is the selected from:
Figure imgf000238_0002
Ring B is a 5- or 6- membered aryl, cycloaliphatic, heteroaryl, or heterocyclyl ring, wherein said ring is optionally substituted with up to 4 occurrences of RB and said heteroaryl or heterocyclyl ring contains up to three heteroatoms selected from N, O, or S; each RB is independently selected from halogen, RB1, -CN, -CO2RB1, -OC(O)RB1,
-OC(O)N(R81), -NO2, -N(RBI)2, -NC(O)RB1, -N(RBI)C(O)N(RBI)2, -SRB1, -S(O)2R8', -S(O)2N(R8 ')2, or -S(O)R81, wherein each RBl is, independently, hydrogen or Q-4 aliphatic, or two RB1 together with the atom to which they are bound, form a 3-6 membered carbocycle optionally substituted with 0-2 occurrences of JR or a 3-6 membered heterocyclyl containing 1 -3 heteroatoms independently selected from N, O, or S and optionally substituted with 0-2 occurrences of JR on carbon and optionally substituted with JN on each substitutable ring nitrogen atom;
Q is C6-IO aryl or 5-10 membered heteroaryl wherein each Q is optionally substituted with up to 5 occurrences of J*"*;
U is N or CR1;
V is N or CR2; U1 is O5 NR5, or S;
V1 is O, NR6, or S;
R1 is hydrogen, halogen, -CN, -NHb5 -OH5 Ci-2 haloalkyl, or selected from -NH(Ci-4 aliphatic), -N(Cj-4 aliphatic)2, C3-4 cycloalkyl, -(Cj-2 aliphatic)-(C3-4 cycloalkyl), or Ci-4 aliphatic, each of which is optionally substituted with up to 2 occurrences of JR;
R2 is hydrogen, halogen, -CN, -NH2, -NH(Cj-4 aliphatic), -N(Ci-4 aliphatic)2, C1-2 haloalkyl, C3-4 cycloalkyl, or C1-4 aliphatic;
R3 is halogen or RA, wherein RΛ is C6.io aryl, 5-10 membered heteroaryl, 5-12 membered heterocyclyl, or C3-8 cycloaliphatic, each of which is optionally substituted with 0-3 occurrences of JM;
R4 is hydrogen, -CN, Cj-4 aliphatic, halogen, or Cj-2 haloalkyl; each of R5, R6, and R7, is, independently, hydrogen or JN; each JM is independently selected from halogen, -NO2, -CN, Ci-4 aliphatic, Ci-2 haloalkyl, -(CH2)o-2CH(R')2, -OH5 -OR', -(CR'"2)qNH2, -(CR'"2)qNHR', -(CR'"2)qN(R')2, -(CR'"2)qNHS(O)2R\ -(CR"'2)qNHC(O)R\ -(CR'"2)qNHC(O)OR\ -(CR' "2)qNHC(O)NH2, -(CR' ' '2)qNHC(O)NHR', -(CR'"2)qNHC(O)N(R')2, -(CR' ' '2)qNHC(NH)NH2, -(CR' ' '2)qNHC(NH)NHR', -(CR' ' '2)qNHC(NR)N(R')2, -(CR'"2)qNHS(O)2NH2, -(CR'"2)qNHS(O)2NHR'5 -(CR'"2)qNHS(O)2N(R')2, -SH5 -SR', -(CR'"2)qCO2H, -(CR'"2)qCO2R'5 -C(O)H, -(CR'"2)qC(O)R'5 -(CR" '2)q-C(0)-(CH2)o-2CH(R')25 -(CR" '2)q-C(O)-(CH2)0-2NHCH(R')2, -(CR'"2)q-C(0)-(CH2)o-2NR'CH(R')2, -(CR'"2)q-C(O)NH25 -(CR'"2)q-C(O)NHR', -(CR' ' '2)q-C(O)N(R')2, -(CR' ' '2)q-C(O)N(OH)R' , -(CR' ' '2)q-C(O)N(OR')R' , -(CR' "2)q-C(O)N(OR')H, -(CR' ' '2)q-C(O)N(OH)H, -(CR' ' '2)q-C(=NOH)R', -(CR'"2)q-C(=NOR')H, -(CR'"2)q-C(NOR')R'5 -(CR'"2)q-S(O)2R'5 -(CR'"2)q-S(O)2OH, -(CR'"2)q-S(O)2OR', -(CR'"2)q-S(O)2NH2, -(CR'"2)q-S(O)2NHR', -(CR'"2)q-S(O)2N(R')2, -(CR'"2)q-S(O)R\ -(CR'"2)q-C(=NR')-NH2, -(CR" '2)q-C(=NR')-NHR\ -(CR' ' '2)q-C(=NR')-N(R')2, -(CR' ' '2)q-C(=NH)-NH2, -(CR"'2)q-C(=NH)-NHR\ -(CR'"2)q-C(=NH)-N(R')2, C6-10 aryl, 5-10 membered heteroaryl, 5-10 membered heterocyclyl, or C3-8 cycloaliphatic, wherein q is selected from 0-4; or two JM, together with the atom or atoms to which they are bound, form a 3-6 membered cycloaliphatic or heterocyclyl ring; wherein each of said cycloaliphatic or heterocyclyl is optionally substituted with up to 3 occurrences of JN or JR; each JN is independently selected from -(CR"'2)q>Ci-4 aliphatic, -(CR'"2)q>C3-6 cycloalkyl, -(CR'"2)q'phenyl, -(CR"'2)q'C(O)CMaliphatic; -(CR"'2)q-C(O)Ci.2haloalkyl; -C(O)O(C,_4alkyl)5 -(CR") 2)q'C(O)NH2, -(CR"'2)q'C(O)NH(CMaliphatic), -(CR'"2)q'C(O)N(Ci-4aliphatic)2- or -S(O)2Ci ^aliphatic, wherein q' is 0-2 and each aliphatic or cycloaliphatic is optionally substituted with up to 2 occurrences of JR; each JQ is independently selected from halogen, -NO2, -CN, CM aliphatic, Ci-4 haloalkyl, -OH, -OR", -NH2, -NHR", -N(R")2, -SH, -SR", -CO2H, -CO2R", -C(O)H, -C(O)R", -C(O)NH2, -C(O)NHR", -C(O)N(R")2, -C(O)N(OH)R", -C(O)N(OR")R", -C(O)N(OR")H, -C(O)N(OH)H, -C(NOH)R", -C(NOR")H, -C(NOR")R", -S(O)2R", -S(O)2OH, -S(O)2OR", -S(O)2NH2, -S(O)2NHR", -S(O)2N(R")2, -S(O)R", -C(=NR')-NH2, -C(=NR')-NHR\ -C(=NR')-N(R')2, -C(=NH)-NH2, -C(=NH)-NHR", -C(=NH)-N(R")2, Cδ-ioaiyl, 5-10 membered heteroaryl, 5-10 membered heterocyclyl, or C3-8 cycloaliphatic; each JR is independently selected from halogen, -NO2, -CN, Ci-4 aliphatic, Ci-4 haloalkyl, C3-4 cycloalkyl, -OH, -NH2, -0(Ci-4 aliphatic), -N(Ci-4 aliphatic)2, or -NH(Ci-4 aliphatic); each R' is independently selected from unsubstituted Ci-6 aliphatic; or two R' groups, together with the atom(s) to which they are bound, form a 3-6 membered cycloaliphatic or heterocyclyl, each optionally substituted with up to 2 occurrences of JR; each R" is independently selected from unsubstituted Ci -β aliphatic; or two R" groups, together with the atom to which they are bound, form a 3-6 membered heterocyclyl, optionally substituted with up to 2 occurrences of JR; and each R'" is independently selected from hydrogen or Ci-4 aliphatic, or an R'" group and an R' group, together with the atoms to which they are bound, form a 3-6 membered cycloaliphatic or heterocyclyl, each optionally substituted with up to 2 occurrences of JR.
2. The compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein: Z1 is N or CR4; Z2 is N or CH; Ring D is the selected from one of the 5-membered rings shown below:
Figure imgf000241_0001
each of R5 and R6 is hydrogen, Ci-2 haloalkyl, or selected from C3-4 cycloalkyl,
-(Ci-2 aliphatic)-(C3-4 cycloalkyl), or C1-4 aliphatic, each of which is optionally substituted with up to 2 occurrences of JR;
R3 is a Ce-io aryl, 5-10 membered heteroaryl, 5-10 membered heterocyclyl, or C3-8 cycloaliphatic, each of which is optionally substituted with 0-3 occurrences of JM; each JQ is, independently, selected from halogen, -NO2, -CN, Ci-4 aliphatic, Ci-4 haloalkyl, -OH, -OR", -NH2, -NHR", -N(R")2, -SH, -SR", -CO2H, -CO2R", -C(O)H, -C(O)R", -C(O)NH2, -C(O)NHR", -C(O)N(R")2, -C(O)N(OH)R", -C(O)N(0R")R", -C(O)N(OR")H, -C(O)N(OH)H, -C(NOH)R", -C(NOR")H, -C(NOR")R", -S(O)2R", ' -S(O)2OH, -S(O)2OR", -S(O)2NH2, -S(O)2NHR", -S(O)2N(R")2, -S(O)R", -C(=NR')-NH2, -C(=NR')-NHR', -C(=NR')-N(R')2, -C(=NH)-NH2, -C(=NH)-NHR", -C(=NH)-N(R")2, C6-i0aryl, 5-10 membered heteroaryl, 5-10 membered heterocyclyl, or C3-8 cycloaliphatic; each JR is, independently, selected from halogen, -NO2, -CN, C1-4 aliphatic, C]-4 haloalkyl, C3-4 cycloalkyl, -OH, -NH2, -0(Ci-2 aliphatic), -N(C1-2 aliphatic)2, or -NH(Ci-2 aliphatic); each JM is, independently, selected from halogen, -NO2, -CN, Ci-4 aliphatic, Ci-2 haloalkyl, -OH, -OR', -(CR'"2)qNH2, -(CR'"2)qNHR'5 -(CR'"2)qN(R')2, -(CR'"2)qNS(O)2R', -(CR'"2)qNHC(O)R', -(CR'"2)qNHC(O)OR\ -(CR" '2)qNH C(O)NH2, -(CR'"2)qNHC(O)NHR', -(CR'"2)qNHC(O)N(R')2, -(CR'"2)qNHS(O)2NH2, -(CR"'2)qNHS(O)2NHR\ -(CR'"2)qNHS(O)2N(R')2, -SH, -SR', -CO2H, -CO2R', -C(O)H, -C(O)R', -C(O)NH2, -C(O)NHR', -C(O)N(R')2, -C(O)N(OH)R', -C(O)N(OR')R', -C(O)N(OR')H, -C(O)N(OH)H, -C(=N0H)R\ -C(=NOR')H, -C(NOR')R', -S(O)2R', -S(O)2OH, -S(O)2OR', -S(O)2NH2, -S(O)2NHR', -S(O)2N(R')2, -S(O)R', -C(=NR')-NH2, -C(=NR')-NHR', -C(=NR')-N(R')2, -C(=NH)-NH2, -C(=NH)- NHR', -C(=NH)-N(R')2, C6-io aryl, 5-10 membered heteroaryl, 5-10 membered heterocyclyl, or C^s cycloaliphatic, wherein q is selected from 0-4; each R' is, independently, selected from unsubstituted Ci-6 aliphatic; or two R' groups, together with the atom to which they are bound, form a 3-6 membered heterocyclyl, optionally substituted with 0-2 occurrences of JR; each R" is, independently, selected from unsubstituted Ci-6 aliphatic; or two R" groups, together with the atom to which they are bound, form a 3-6 membered heterocyclyl, optionally substituted with 0-2 occurrences of JR; and each R'" is, independently, selected from hydrogen or CM aliphatic.
3. The compound according to claim 1 , or a pharmaceutically acceptable salt thereof, wherein Ring D is selected from:
Figure imgf000242_0001
4. The compound according to claim 3, or a pharmaceutically acceptable salt thereof, wherein Ring D is selected from:
Figure imgf000242_0002
5. The compound according to claim 1 , or a pharmaceutically acceptable salt thereof, wherein Ring D is selected from:
Figure imgf000243_0001
6. The compound according to claim 5, or a pharmaceutically acceptable salt thereof, wherein Ring D is selected from
Figure imgf000243_0002
7. The compound according to claim 5, or a pharmaceutically acceptable salt thereof, wherein Ring D is
Figure imgf000243_0003
8. The compound according to claim 5, or a pharmaceutically acceptable salt thereof, wherein Ring D is selected from
Figure imgf000244_0001
9. The compound according to claim 8, or a pharmaceutically acceptable salt thereof, wherein Ring D is
Figure imgf000244_0002
10. The compound according to claim 8, or a pharmaceutically acceptable salt thereof, wherein Ring D is
Figure imgf000244_0003
11. The compound according to claim 1 , or a pharmaceutically acceptable salt thereof, wherein Ring D is
Figure imgf000244_0004
12. The compound according to claim 11 , or a pharmaceutically acceptable salt thereof, wherein Ring D is
Figure imgf000244_0005
13. The compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein Ring A is selected from
Figure imgf000245_0001
14. The compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein Ring A is
Figure imgf000245_0002
15. The compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein Ring A is
Figure imgf000245_0003
16. The compound according to claim 1, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000245_0004
17. The compound according to claim 1 , or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000246_0001
18. The compound according to claim 16 or 17, or a pharmaceutically acceptable salt thereof, wherein R4 is hydrogen.
19. The compound according to claim 1 , or a pharmaceutically acceptable salt thereof, wherein Q is
(JQ)θ-5 .
20. The compound according to any according to claims 1 to 19, or a pharmaceutically acceptable salt thereof, wherein Q is
Figure imgf000246_0002
21. The compound according to claim 20, or a pharmaceutically acceptable salt thereof, wherein each JQ is, independently, fluoro or chloro.
22. The compound according to claim 21 , or a pharmaceutically acceptable salt thereof, wherein Q is
Figure imgf000246_0003
23. The compound according to claim 1, wherein R3 is a C6-Io aryl, a C3-8 cycloaliphatic, or a monocyclic or bicyclic 5-10 membered heteroaryl or heterocyclyl containing 1-4 heteroatoms independently selected from N, O, or S, wherein said aryl, cycloaliphatic, heteroaryl, or heterocyclyl is optionally substituted with up to 3 occurrences ofJM.
24. The compound according to claim 1 , or a pharmaceutically acceptable salt thereof, wherein RA is phenyl optionally substituted with up to 3 occurrences of JM.
25. The compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein RA is a 5-6 membered heteroaryl optionally substituted with up to 3 occurrences of JM.
26. The compound according to claim 25, or a pharmaceutically acceptable salt thereof, wherein RA is selected from pyridyl, thienyl, thiazolyl, isothiazolyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, furanyl, isoxazolyl, or oxazolyl, each of which optionally substituted with up to 3 occurrences of JM.
27. The compound according to claim 26, wherein RA is selected from IH- pyrazol-4-yl substituted at the 1 -position with JM, thiophen-2-yl substituted at the 5-position with JM, thiophen-3-yl substituted at the 5-position with JM, furan-2-yl substituted at the 5- position with JM, furan-3-yi substituted at the 5-position with JM, lH-pyrrol-3-yl substituted at the 1 -position with JM, IH-1, 2,3 -triazol-4-yl substituted at the 1 -position with JM, or thiazol-5-yl substituted at the 2-position with JM.
28. The compound according to any according to claims 23 to 27, or a pharmaceutically acceptable salt thereof, wherein JM is selected from phenyl, 5-8 membered heteroaryl, 5-10 membered heterocyclyl, or C3-8cycloaliphatic. '
29. The compound according to claim 28, or a pharmaceutically acceptable salt thereof, wherein JM is selected from 5-10 membered heterocyclyl.
30. The compound according to claim 29, or a pharmaceutically acceptable salt thereof, wherein JM is a 5-10 membered heterocyclyl containing 1 or 2 nitrogen atoms.
31. The compound according to claim 30, or a pharmaceutically acceptable salt thereof, wherein JM is an optionally substituted piperidine, piperazine or pyrrolidine.
32. The compound according to claim 30, or a pharmaceutically acceptable salt thereof, wherein JM is an optionally substituted bicyclooctane or bicyclononane containing 1 or 2 nitrogen atoms.
33. The compound according to claim 23, or a pharmaceutically acceptable salt thereof, wherein RA is a Cs-iobicyclic heteroaryl selected from:
Figure imgf000248_0001
or , wherein
Ring E is a 5-membered heteroaryl ring with 1 to 2 heteroatoms selected from N, O, or S; and JE is hydrogen or JN.
34. The compound according to claim 33, or a pharmaceutically acceptable salt thereof, wherein Ring E is selected from thienyl, thiazolyl, pyrrolyl, imidazolyl, furanyl, or oxazolyl.
35. The compound according to claim 33, or a pharmaceutically acceptable salt thereof, wherein RA is
Figure imgf000248_0002
or , wherein X M i •s O or S
36. The compound according to claim 35, wherein XM is S.
37. The compound according to claim 23, or a pharmaceutically acceptable salt thereof, wherein RA is a 5-7 membered heterocyclyl selected from:
Figure imgf000249_0001
wherein JF is selected from Ci-4 aliphatic, -C0-2aliρhaticCH(R')2, -(CR'"2)qNH2, -(CR'"2)qNHR', -(CR'"2)qN(R')2, -(CR'"2)qNS(O)2R\ -(CR'"2)qNHC(O)R\ -(CR"'2)qNHC(O)OR\ -(CR'"2)qNHC(O)NH2, -(CR'"2)qNHC(O)NHR% -(CR' "2)qNHC(O)N(R')2, -(CR' ' '2)qNHC(NH)NH2, -(CR' ' '2)qNHC(NH)NHR' , -(CR' "2)qNHC(NH)N(R')2, -(CR' "2)qNHS(O)2NH2, -(CR' ' '2)qNHS(O)2NHR', -(CR'"2)qNHS(O)2N(R')25 -CO2R', -C(O)H, -C(O)R', -C(O)-(CH2)0.2CH(R')2, -C(O)NH2, -C(O)NHR', -C(O)N(R')2, -S(O)2R', -S(O)2NH2, -S(O)2NHR', -S(O)2N(R')2, C6.i0 aiyl, 5-10 membered heteroaryl, 5-10 membered heterocyclyl, or C3-8 cycloaliphatic, wherein q is selected from 0-4 and said aryl, heteroaryl, heterocyclyl, or cycloaliphatic of JF is optionally substituted with halogen, -NO2, -CN5 C1.4 aliphatic, Ci-4 haloalkyl, C3-4 cycloalkyl, -OH, - NH2, -O(Ci_2 aliphatic), -N(Ci-2 aliphatic)2, or -NH(Ci-2 aliphatic).
38. The compound according to claim 37, wherein RA is
Figure imgf000249_0002
, wherein
JF is -C0-2aliphaticCH(R')2, -(CR" '2)qNH2, -(CR"'2)qNHR\ -(CR'"2)qN(R')2, -C(O)R', -C(0)-(CH2)o-2CH(R')2, or optionally substituted heterocyclyl.
39. The compound according to any claims 23 to 38, or a pharmaceutically acceptable salt thereof, wherein JM is selected from halogen, -NO2, -CN, Ci-4 aliphatic, Ci-2 haloalkyl, -OH, -OR', -(CR'"2)qNH2, -(CR"'2)qNHR\ -(CR"'2)qN(R')2, - (CR"'2)qNS(O)2R\ -(CR'"2)qNHC(O)R', -(CR'"2)qNHC(O)OR', -(CR'"2)qNHC(O)NH2. -(CR'"2)qNHC(O)NHR', -(CR'"2)qNHC(O)N(R')2, -(CR' ' '2)qNHS(O)2NH2, -(CR'"2)qNHS(O)2NHR', -(CR'"2)qNHS(O)2N(R')2, -SH, -SR', -CO2H, -CO2R', -C(O)H, -C(O)R', -C(O)NH2, -C(O)NHR', -C(O)N(R')2, -C(O)N(OH)R', -C(O)N(OR')R\ -C(O)N(OR')H, -C(O)N(OH)H, -C(=NOH)R', -C(=NOR')H, -C(NOR')R\ -S(O)2R', -S(O)2OR', -S(O)2NH2, -S(O)2NHR', -S(O)2N(R')2, -S(O)R', -C(=NR')-NH2, -C(=NR')- NHR', -C(=NR')-N(R')2, -C(=NH)-NH2, -C(=NH)-NHR', or -C(=NH)-N(R')2.
40. The compound according to claim 39, or a pharmaceutically acceptable salt thereof, wherein JM is selected from halogen, -NO2, -CN, Ci ^aliphatic, C^haloalkyl, -OH, -OR', -NH2, -NHR', -N(R')2, -SH, -SR', -CO2H, -CO2R', -C(O)H, -C(O)R', -C(O)NH2, -C(O)NHR', -C(O)N(R')2, -C(O)N(OH)H, -C(O)N(OH)R', -C(O)N(OR')H, -C(O)N(OR')R\ -C(NOH)H, -C(NOR')H, -C(NOH)R', -C(NOR')R', -S(O)2R', -S(O)3R', -S(O)2NH2, -S(O)2NHR', -S(O)2N(R')2, -S(O)R', -C(=NR')-NH2, -C(=NR')-NHR', -C(=NR')-N(R')2, -C(=NH)-NH2, -C(=NH)-NHR\ or -C(=NH)-N(R')2.
41. The compound according to claim 40, or a pharmaceutically acceptable salt thereof, wherein JM is selected from halogen, NO2, CN, Ci-4 aliphatic, C1-2 haloalkyl, -OH, -OR', -NH2, -NHR', -N(R')2, -SH, or -SR'.
42. The compound according to claim 39, or a pharmaceutically acceptable salt thereof, wherein JM is selected from -(CR" '2)qNH2, -(CR'"2)qNHR5, -(CR'"2)qN(R')2, -(CR'"2)qNS(O)2R', -(CR'"2)qNHC(O)R', -(CR'"2)qNHC(O)OR', -(CR" > 2)qNHC(O)NH2, -(CR"'2)qNHC(O)NHR\ -(CR'"2)qNHC(O)N(R')2, -(CR'"2)qNHS(O)2NH2, - (CR'"2)qNHS(O)2NHR', or -(CR'"2)qNHS(O)2N(R')2.
43. The compound according to claim 42, or a pharmaceutically acceptable salt thereof, wherein JM is selected from
Figure imgf000251_0001
44. The compound according to claim 43, wherein q is 1 or 2.
45. The compound according to any of claims 23 to 44, or a pharmaceutically acceptable salt thereof, wherein JM is not substituted on a ring position adjacent to Ring A.
46. The compound according to claim 1 , wherein R3 is halogen.
47. The compound according to any of claims 1-17, 19-20, 23-27, or 33-44, or a pharmaceutically acceptable salt thereof, wherein said C1-4Or C1-6 aliphatic of R1, R2, R4, R5, R6, JM, JN, J^, JR, R', R", or R"' comprises two or more non-hydrogen atoms.
48. The compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein said compound has formula:
(I-A) and is listed in Table 1.
49. The compound according to claim 1 , or a pharmaceutically acceptable salt thereof, wherein said compound has formula:
Figure imgf000252_0001
(I-B) and is listed in Table 2.
50. The compound according to claim 1 , or a pharmaceutically acceptable salt thereof, wherein said compound has formula:
Figure imgf000252_0002
(I-C) and is listed in Table 3.
51. The compound according to claim 1 , or a pharmaceutically acceptable salt thereof, wherein said compound has formula:
Figure imgf000252_0003
(I-D) and is listed in Table 4.
52. The compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein said compound has formula:
Figure imgf000252_0004
(I-E) and is listed in Table 5.
53. A composition comprising an effective amount of a compound according to any of claims 1-17, 19, 23-27, 33-38, 46, or 48-52, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, adjuvant or vehicle.
54. The composition according to claim 53, wherein said compound is in an amount sufficient to detectably inhibit c-MET protein kinase activity.
55. The composition according to claim 53, additionally comprising a chemotherapeutic or anti-proliferative agent, an anti-inflammatory agent, an agent for treating atherosclerosis, an agent for treating lung fibrosis, an immunomodulatory or immunosuppressive agent, a neurotrophic factor, an agent for treating cardiovascular disease, an agent for treating conditions associated with organ transplantation, an agent for treating allergic disorders, an agent for treating destructive bone disorders, an agent for treating liver disease, an anti-viral agent, an agent for treating blood disorders, an agent for treating diabetes or an agent for treating immunodeficiency disorders.
56. Use of a compound according to any of claims 1-17, 19, 23-27, 33-38, 46, or 48-52, or a pharmaceutical composition comprising said compound, for treating or lessening the severity of a proliferative disorder in a patient.
57. Use of a compound according to any of claims 1-17, 19, 23-27, 33-38, 46, or 48-52, or a pharmaceutical composition comprising said compound, in the preparation of a medicament for treating or lessening the severity of a proliferative disorder in a patient.
58. The use according to claim 56, wherein said proliferative disorder is cancer.
59. The use according to claim 58, wherein said cancer is metastatic.
60. The use according to claim 56, wherein said disorder is a glioblastoma; a gastric carcinoma; or a cancer selected from colon, breast, prostate, brain, liver, pancreatic or lung cancer.
61. The use according to claim 56, wherein said disorder is gastric carcinoma.
62. The use according to claim 56, wherein said disorder is atherosclerosis or lung fibrosis.
63. A use of inhibiting c-MET protein kinase activity in a biological sample in vitro comprising contacting said biological sample with a compound according to any of claims 1-17, 19, 23-27, 33-38, 46, or 48-52, or a pharmaceutical composition comprising said compound.
PCT/US2007/007016 2006-03-22 2007-03-21 C-met protein kinase inhibitors for the treatment of proliferative disorders WO2007111904A2 (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
CA002646701A CA2646701A1 (en) 2006-03-22 2007-03-21 C-met protein kinase inhibitors for the treatment of proliferative disorders
NZ571933A NZ571933A (en) 2006-03-22 2007-03-21 C-Met protein kinase inhibitors for the treatment of proliferative disorders
CN2007800182084A CN101479255B (en) 2006-03-22 2007-03-21 C-MET protein kinase inhibitors for the treatment of proliferative disorders
AU2007231577A AU2007231577B2 (en) 2006-03-22 2007-03-21 c-MET protein kinase inhibitors for the treatment of proliferative disorders
MX2008012096A MX2008012096A (en) 2006-03-22 2007-03-21 C-met protein kinase inhibitors for the treatment of proliferative disorders.
AT07753626T ATE453635T1 (en) 2006-03-22 2007-03-21 C-MET PROTEIN KINASE INHIBITORS FOR THE TREATMENT OF PROLIFERATIVE DISEASES
EP07753626A EP2004625B1 (en) 2006-03-22 2007-03-21 C-met protein kinase inhibitors for the treatment of proliferative disorders
DE602007004092T DE602007004092D1 (en) 2006-03-22 2007-03-21 C-MET PROTEIN KINASE INHIBITOR FOR THE TREATMENT OF PROLIFERATIVE DISEASES
JP2009501540A JP2009533327A (en) 2006-03-22 2007-03-21 C-MET Kinase Inhibitors for Treating Proliferative Diseases
IL194242A IL194242A0 (en) 2006-03-22 2008-09-22 Heterocyclic compounds and pharmaceutical compositions containing the same
NO20084435A NO20084435L (en) 2006-03-22 2008-10-22 C-MET protein kinase inhibitors
HK09107689.2A HK1129890A1 (en) 2006-03-22 2009-08-21 C-met protein kinase inhibitors for the treatment of proliferative disorders
US12/942,395 US8232406B2 (en) 2006-03-22 2010-11-09 c-MET protein kinase inhibitors

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US78493706P 2006-03-22 2006-03-22
US60/784,937 2006-03-22
US87597306P 2006-12-20 2006-12-20
US60/875,973 2006-12-20

Publications (2)

Publication Number Publication Date
WO2007111904A2 true WO2007111904A2 (en) 2007-10-04
WO2007111904A3 WO2007111904A3 (en) 2007-12-13

Family

ID=38372386

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/007016 WO2007111904A2 (en) 2006-03-22 2007-03-21 C-met protein kinase inhibitors for the treatment of proliferative disorders

Country Status (20)

Country Link
US (2) US7872031B2 (en)
EP (1) EP2004625B1 (en)
JP (2) JP2009533327A (en)
KR (1) KR20090007347A (en)
CN (1) CN101479255B (en)
AR (1) AR060054A1 (en)
AT (1) ATE453635T1 (en)
AU (1) AU2007231577B2 (en)
CA (1) CA2646701A1 (en)
CL (1) CL2007000752A1 (en)
DE (1) DE602007004092D1 (en)
ES (1) ES2336625T3 (en)
HK (1) HK1129890A1 (en)
IL (1) IL194242A0 (en)
MX (1) MX2008012096A (en)
NO (1) NO20084435L (en)
NZ (1) NZ571933A (en)
RU (1) RU2008141761A (en)
TW (1) TW200806646A (en)
WO (1) WO2007111904A2 (en)

Cited By (75)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007139951A2 (en) * 2006-05-25 2007-12-06 Synta Pharmaceuticals Corp. Method for treating proliferative disorders associated with protooncogene products
WO2008074997A1 (en) 2006-12-21 2008-06-26 Cancer Research Technology Limited Pyridine benzamides and pyrazine benzamides used as pkd inhibitors
WO2009045992A2 (en) * 2007-10-03 2009-04-09 Vertex Pharmaceuticals Incorporated C-met protein kinase inhibitors
WO2009099982A1 (en) * 2008-02-04 2009-08-13 Osi Pharmaceuticals, Inc. 2-aminopyridine kinase inhibitors
WO2009139576A2 (en) * 2008-05-14 2009-11-19 한국화학연구원 Pyridine derivatives substituted with novel benzoxazoles or pharmaceutically acceptable salts thereof, preparation method thereof and pharmaceutical compositions containing the same as active ingredients for prevention and treatment of abnormal cell growth disease
EP2150255A1 (en) * 2007-05-10 2010-02-10 Smithkline Beecham Corporation Quinoxaline derivatives as p13 kinase inhibitors
WO2010048131A1 (en) 2008-10-21 2010-04-29 Vertex Pharmaceuticals Incorporated C-met protein kinase inhibitors
WO2010052448A2 (en) * 2008-11-05 2010-05-14 Ucb Pharma S.A. Fused pyrazine derivatives as kinase inhibitors
WO2010054398A1 (en) * 2008-11-10 2010-05-14 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
JP2010534634A (en) * 2007-07-26 2010-11-11 ノバルティス アーゲー Pyrimidine derivatives useful for the treatment of inflammatory or allergic conditions
DE102009033208A1 (en) 2009-07-15 2011-01-20 Merck Patent Gmbh aminopyridine derivatives
WO2011022473A1 (en) * 2009-08-19 2011-02-24 Ambit Biosciences Corporation Biaryl compounds and methods of use thereof
US20110172203A1 (en) * 2009-12-30 2011-07-14 Arqule, Inc. Substituted Imidazopyridinyl-Aminopyridine Compounds
US8017611B2 (en) 2007-10-25 2011-09-13 Astrazeneca Ab Pyridine and pyrazine derivatives -083
US8022206B2 (en) 2008-02-06 2011-09-20 OSI Pharmaceuticals, LLC Furo[3,2-C]pyridines
WO2011143422A1 (en) * 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated 2 -aminopyridine derivatives useful as inhibitors of atr kinase
WO2011143426A1 (en) * 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2011145035A1 (en) 2010-05-17 2011-11-24 Indian Incozen Therapeutics Pvt. Ltd. Novel 3,5-disubstitued-3h-imidazo[4,5-b]pyridine and 3,5- disubstitued -3h-[1,2,3]triazolo[4,5-b] pyridine compounds as modulators of protein kinases
JP2012512906A (en) * 2008-12-19 2012-06-07 バーテックス ファーマシューティカルズ インコーポレイテッド Pyrazine derivatives useful as inhibitors of ATR kinase
WO2013071088A1 (en) * 2011-11-09 2013-05-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2013071090A1 (en) * 2011-11-09 2013-05-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2013144737A2 (en) 2012-03-30 2013-10-03 Rhizen Pharmaceuticals Sa Novel 3,5-disubstitued-3h-imidazo[4,5-b]pyridine and 3,5- disubstitued -3h-[1,2,3]triazolo[4,5-b] pyridine compounds as modulators of c-met protein kinases
US8609688B2 (en) 2011-06-24 2013-12-17 Arqule, Inc. Substituted imidazopyridinyl-aminopyridine compounds
US8623869B2 (en) 2010-06-23 2014-01-07 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8765751B2 (en) 2011-09-30 2014-07-01 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
WO2014106612A1 (en) * 2013-01-02 2014-07-10 H. Lundbeck A/S Aminopyridine derived compounds as lrrk2 inhibitors
US8815854B2 (en) 2011-06-24 2014-08-26 Arqule, Inc. Substituted imidazopyridinyl compounds
US8822469B2 (en) 2011-06-22 2014-09-02 Vertex Pharmaceuticals Incorporated Pyrrolo[2,3-B]pyrazines useful as inhibitors of ATR kinase
US8841449B2 (en) 2011-11-09 2014-09-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8841450B2 (en) 2011-11-09 2014-09-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8846686B2 (en) 2011-09-30 2014-09-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8846918B2 (en) 2011-11-09 2014-09-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8853217B2 (en) 2011-09-30 2014-10-07 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8877759B2 (en) 2011-04-05 2014-11-04 Vertex Pharnaceuticals Incorporated Aminopyrazines as ATR kinase inhibitors
US8912198B2 (en) 2012-10-16 2014-12-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8962631B2 (en) 2010-05-12 2015-02-24 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8969356B2 (en) 2010-05-12 2015-03-03 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9035053B2 (en) 2011-09-30 2015-05-19 Vertex Pharmaceuticals Incorporated Processes for making compounds useful as inhibitors of ATR kinase
US9062008B2 (en) 2010-05-12 2015-06-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
EP2805946A4 (en) * 2012-01-17 2015-07-29 Shanghai Allist Pharmaceuticals Inc Amino heteroaryl compound, preparation method therefor and use thereof
US9096602B2 (en) 2011-06-22 2015-08-04 Vertex Pharmaceuticals Incorporated Substituted pyrrolo[2,3-B]pyrazines as ATR kinase inhibitors
WO2015148464A1 (en) * 2014-03-24 2015-10-01 Arqule Inc. Process of preparing 3-(3-(4-(1-aminocyclobutyl)phenyl)-5-phenyl-3h-imidazo[4,5-b]pyridin-2-yl)pyridin-2-amine
US9156831B2 (en) 2013-01-23 2015-10-13 Astrazeneca Ab Chemical compounds
WO2015164479A1 (en) * 2014-04-22 2015-10-29 Arqule, Inc. Salts and polymorphs of a substituted imidazopyridinyl-aminopyridine compound
US9205086B2 (en) 2010-04-19 2015-12-08 Synta Pharmaceuticals Corp. Cancer therapy using a combination of a Hsp90 inhibitory compounds and a EGFR inhibitor
US9309250B2 (en) 2011-06-22 2016-04-12 Vertex Pharmaceuticals Incorporated Substituted pyrrolo[2,3-b]pyrazines as ATR kinase inhibitors
US9334244B2 (en) 2010-05-12 2016-05-10 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9340546B2 (en) 2012-12-07 2016-05-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9403815B2 (en) 2010-06-24 2016-08-02 The Regents Of The University Of California Compounds and uses thereof in modulating levels of various amyloid beta peptide alloforms
US9439899B2 (en) 2011-11-02 2016-09-13 Synta Pharmaceuticals Corp. Cancer therapy using a combination of HSP90 inhibitors with topoisomerase I inhibitors
US9663519B2 (en) 2013-03-15 2017-05-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9670215B2 (en) 2014-06-05 2017-06-06 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
EP3122355A4 (en) * 2014-03-26 2017-08-09 The Brigham and Women's Hospital, Inc. Compositions and methods for inhibiting bmp
US9791456B2 (en) 2012-10-04 2017-10-17 Vertex Pharmaceuticals Incorporated Method for measuring ATR inhibition mediated increases in DNA damage
US9862711B2 (en) 2014-04-24 2018-01-09 Novartis Ag Pyrazine derivatives as phosphatidylinositol 3-kinase inhibitors
US9938263B2 (en) 2013-03-12 2018-04-10 The General Hospital Corporation Gamma-secretase modulators
EP3201187A4 (en) * 2014-10-01 2018-04-11 The Brigham and Women's Hospital, Inc. Compositions and methods for inhibiting bmp
US10004732B2 (en) 2014-04-24 2018-06-26 Novartis Ag Amino pyrazine derivatives as phosphatidylinositol 3-kinase inhibitors
US10017516B2 (en) 2013-03-14 2018-07-10 The Brigham And Women's Hospital, Inc. BMP inhibitors and methods of use thereof
US10112926B2 (en) 2014-04-24 2018-10-30 Novartis Ag Amino pyridine derivatives as phosphatidylinositol 3-kinase inhibitors
US10160760B2 (en) 2013-12-06 2018-12-25 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
WO2019038717A1 (en) 2017-08-23 2019-02-28 Novartis Ag 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
US10472346B2 (en) 2014-10-31 2019-11-12 The General Hospital Corporation Potent gamma-secretase modulators
US10478430B2 (en) 2012-04-05 2019-11-19 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase and combination therapies thereof
US10500193B2 (en) 2011-11-02 2019-12-10 Synta Pharmaceuticals Corporation Combination therapy of HSP90 inhibitors with platinum-containing agents
US10513521B2 (en) 2014-07-15 2019-12-24 The Brigham And Women's Hospital, Inc. Compositions and methods for inhibiting BMP
CN111116585A (en) * 2019-12-31 2020-05-08 北京鑫开元医药科技有限公司 Compound with c-MET kinase inhibitory activity, preparation method, composition and application
WO2020188015A1 (en) 2019-03-21 2020-09-24 Onxeo A dbait molecule in combination with kinase inhibitor for the treatment of cancer
US10813929B2 (en) 2011-09-30 2020-10-27 Vertex Pharmaceuticals Incorporated Treating cancer with ATR inhibitors
WO2021089791A1 (en) 2019-11-08 2021-05-14 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment of cancers that have acquired resistance to kinase inhibitors
US11046658B2 (en) 2018-07-02 2021-06-29 Incyte Corporation Aminopyrazine derivatives as PI3K-γ inhibitors
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use
US11179394B2 (en) 2014-06-17 2021-11-23 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of Chk1 and ATR inhibitors
US11464774B2 (en) 2015-09-30 2022-10-11 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of DNA damaging agents and ATR inhibitors
US11926616B2 (en) 2018-03-08 2024-03-12 Incyte Corporation Aminopyrazine diol compounds as PI3K-γ inhibitors

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5325783B2 (en) 2006-09-08 2013-10-23 エフ.ホフマン−ラ ロシュ アーゲー Benzotriazole kinase modulator
MX2012002596A (en) * 2009-09-03 2012-07-03 Allergan Inc Compounds as tyrosine kinase modulators.
CA2782601C (en) * 2009-12-18 2015-07-21 Mitsubishi Tanabe Pharma Corporation Novel antiplatelet agent
FR2974365B1 (en) * 2011-04-20 2017-08-25 Centre Nat De La Rech Scient (C N R S) 1,4-DISUBSTITUTED 1,2,3-TRIAZOLES, METHODS FOR PREPARING THEM AND DIAGNOSTIC AND THERAPEUTIC USES THEREOF
JP5959330B2 (en) * 2011-06-17 2016-08-02 田辺三菱製薬株式会社 New antiplatelet drugs
WO2012178038A1 (en) 2011-06-24 2012-12-27 The Broad Institute, Inc. Methods of treating cancer
EP2739617A4 (en) * 2011-07-27 2015-01-28 Nanjing Allgen Pharma Co Ltd Spirocyclic molecules as protein kinase inhibitors
EP2780010A1 (en) 2011-11-14 2014-09-24 Synta Pharmaceuticals Corp. Combination therapy of hsp90 inhibitors with braf inhibitors
WO2014055595A1 (en) 2012-10-05 2014-04-10 Merck Sharp & Dohme Corp. Indoline compounds as aldosterone synthase inhibitiors related applications
BR112016012146B1 (en) * 2013-11-27 2022-11-01 Signalchem Lifesciences Corporation AMINOPYRIDINE DERIVATIVE COMPOUNDS AS KINASE INHIBITORS OF THE TAM FAMILY, PHARMACEUTICAL COMPOSITION COMPRISING SAID COMPOUNDS AND THERAPEUTIC USE THEREOF
CN103804312B (en) * 2014-02-17 2016-04-20 四川百利药业有限责任公司 Aza cyclic cpds and its production and use
CN105315287A (en) * 2014-08-04 2016-02-10 杭州沙力医药科技有限公司 Synthetic method for mono-N-t-butyloxycarbonyl-2,6-bisazaspiro[3,3] heptane hydrochloride
MA41598A (en) * 2015-02-25 2018-01-02 Constellation Pharmaceuticals Inc PYRIDAZINE THERAPEUTIC COMPOUNDS AND THEIR USES
CN105061405A (en) * 2015-08-04 2015-11-18 合肥创新医药技术有限公司 Preparation method of fimasartan potassium salt hydrate
EP3728230A1 (en) * 2017-12-22 2020-10-28 Ravenna Pharmaceuticals, Inc. Aryl-bipyridine amine derivatives as phosphatidylinositol phosphate kinase inhibitors
CN110317190A (en) * 2018-03-28 2019-10-11 首都医科大学 A kind of application of triazole-ramification of carboxylic esters in field of medicaments
US20220127249A1 (en) * 2019-02-22 2022-04-28 Janssen Pharmaceutica Nv Crystalline form of 1-(1-oxo-1,2-dihydroisoquinolin-5-yl)-5-(trifluoromethyl)-n-(2-(trifluoromethyl)pyridin-4-yl)-1h-pyrazole-4-carboxamide monohydrate
CN112142716B (en) * 2020-10-29 2021-08-31 山东新时代药业有限公司 5-membered heteroaryl substituted pyrazine derivative and application thereof
CN112409337A (en) * 2020-12-21 2021-02-26 李寒 Substituted pyrazine derivative and application thereof
CN112341437A (en) * 2020-12-21 2021-02-09 李寒 Substituted pyrazine derivative and application thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005040345A2 (en) * 2003-09-24 2005-05-06 Vertex Pharmceuticals Incorporated 4-azole substituted imidazole compositions useful as inhibitors or c-met receptor tyrosine kinase
WO2005040154A1 (en) * 2003-10-17 2005-05-06 F. Hoffmann-La Roche Ag Novel imidazole derivatives and their use as pharmaceutical agents

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GEP20084341B (en) * 2003-02-26 2008-03-25 Sugen Inc Aminoheteroaryl compounds as protein kinase inhibitors
ATE463486T1 (en) * 2004-08-26 2010-04-15 Pfizer ENANTIOMER PURE AMINOHETEROARYL COMPOUNDS AS PROTEIN KINASE INHIBITORS

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005040345A2 (en) * 2003-09-24 2005-05-06 Vertex Pharmceuticals Incorporated 4-azole substituted imidazole compositions useful as inhibitors or c-met receptor tyrosine kinase
WO2005040154A1 (en) * 2003-10-17 2005-05-06 F. Hoffmann-La Roche Ag Novel imidazole derivatives and their use as pharmaceutical agents

Cited By (146)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007139951A3 (en) * 2006-05-25 2008-12-24 Synta Pharmaceuticals Corp Method for treating proliferative disorders associated with protooncogene products
US8969396B2 (en) 2006-05-25 2015-03-03 Synta Pharmaceuticals Corp. Method for treating a B-raf associated cancer with an Hsp90 inhibitor
WO2007139951A2 (en) * 2006-05-25 2007-12-06 Synta Pharmaceuticals Corp. Method for treating proliferative disorders associated with protooncogene products
WO2008074997A1 (en) 2006-12-21 2008-06-26 Cancer Research Technology Limited Pyridine benzamides and pyrazine benzamides used as pkd inhibitors
EP2150255A4 (en) * 2007-05-10 2011-10-05 Glaxosmithkline Llc Quinoxaline derivatives as p13 kinase inhibitors
EP2150255A1 (en) * 2007-05-10 2010-02-10 Smithkline Beecham Corporation Quinoxaline derivatives as p13 kinase inhibitors
JP2010534634A (en) * 2007-07-26 2010-11-11 ノバルティス アーゲー Pyrimidine derivatives useful for the treatment of inflammatory or allergic conditions
WO2009045992A2 (en) * 2007-10-03 2009-04-09 Vertex Pharmaceuticals Incorporated C-met protein kinase inhibitors
WO2009045992A3 (en) * 2007-10-03 2009-06-04 Vertex Pharma C-met protein kinase inhibitors
US8481524B2 (en) 2007-10-03 2013-07-09 Vertex Pharmaceuticals Incorporated c-MET protein kinase inhibitors
JP2010540649A (en) * 2007-10-03 2010-12-24 バーテックス ファーマシューティカルズ インコーポレイテッド Inhibitor of c-MET protein
US8017611B2 (en) 2007-10-25 2011-09-13 Astrazeneca Ab Pyridine and pyrazine derivatives -083
WO2009099982A1 (en) * 2008-02-04 2009-08-13 Osi Pharmaceuticals, Inc. 2-aminopyridine kinase inhibitors
US8178668B2 (en) 2008-02-04 2012-05-15 OSI Pharmaceuticals, LLC 2-aminopyridine kinase inhibitors
US8022206B2 (en) 2008-02-06 2011-09-20 OSI Pharmaceuticals, LLC Furo[3,2-C]pyridines
WO2009139576A3 (en) * 2008-05-14 2010-02-11 한국화학연구원 Pyridine derivatives substituted with novel benzoxazoles or pharmaceutically acceptable salts thereof, preparation method thereof and pharmaceutical compositions containing the same as active ingredients for prevention and treatment of abnormal cell growth disease
WO2009139576A2 (en) * 2008-05-14 2009-11-19 한국화학연구원 Pyridine derivatives substituted with novel benzoxazoles or pharmaceutically acceptable salts thereof, preparation method thereof and pharmaceutical compositions containing the same as active ingredients for prevention and treatment of abnormal cell growth disease
US8518938B2 (en) 2008-10-21 2013-08-27 Vertex Pharmaceuticals Incorporated c-MET protein kinase inhibitors
US20120165333A1 (en) * 2008-10-21 2012-06-28 Vertex Pharmaceuticals Incorporated C-met protein kinase inhibitors
WO2010048131A1 (en) 2008-10-21 2010-04-29 Vertex Pharmaceuticals Incorporated C-met protein kinase inhibitors
JP2012506381A (en) * 2008-10-21 2012-03-15 バーテックス ファーマシューティカルズ インコーポレイテッド c-MET protein kinase inhibitor
WO2010052448A3 (en) * 2008-11-05 2010-09-10 Ucb Pharma S.A. Fused pyrazine derivatives as kinase inhibitors
WO2010052448A2 (en) * 2008-11-05 2010-05-14 Ucb Pharma S.A. Fused pyrazine derivatives as kinase inhibitors
WO2010054398A1 (en) * 2008-11-10 2010-05-14 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
US8410112B2 (en) 2008-11-10 2013-04-02 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
AU2009313198B2 (en) * 2008-11-10 2016-03-24 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
CN102264721A (en) * 2008-11-10 2011-11-30 沃泰克斯药物股份有限公司 Compounds useful as inhibitors of ATR kinase
KR101712576B1 (en) * 2008-11-10 2017-03-06 버텍스 파마슈티칼스 인코포레이티드 Compounds useful as Inhibitors of ATR kinase
JP2012508260A (en) * 2008-11-10 2012-04-05 バーテックス ファーマシューティカルズ インコーポレイテッド Compounds useful as inhibitors of ATR kinase
JP2014240410A (en) * 2008-11-10 2014-12-25 バーテックス ファーマシューティカルズ インコーポレイテッドVertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
KR20110084527A (en) * 2008-11-10 2011-07-25 버텍스 파마슈티칼스 인코포레이티드 Compounds useful as inhibitors of atr kinase
JP2016216495A (en) * 2008-11-10 2016-12-22 バーテックス ファーマシューティカルズ インコーポレイテッドVertex Pharmaceuticals Incorporated Compound useful as inhibitor of atr kinase
JP2015024996A (en) * 2008-12-19 2015-02-05 バーテックス ファーマシューティカルズ インコーポレイテッドVertex Pharmaceuticals Incorporated Pyrazine compounds useful as inhibitors of atr kinase
US10479784B2 (en) 2008-12-19 2019-11-19 Vertex Pharmaceuticals Incorporated Substituted pyrazin-2-amines as inhibitors of ATR kinase
US8841308B2 (en) 2008-12-19 2014-09-23 Vertex Pharmaceuticals Incorporated Pyrazin-2-amines useful as inhibitors of ATR kinase
US9365557B2 (en) 2008-12-19 2016-06-14 Vertex Pharmaceuticals Incorporated Substituted pyrazin-2-amines as inhibitors of ATR kinase
US9701674B2 (en) 2008-12-19 2017-07-11 Vertex Pharmaceuticals Incorporated Substituted pyrazines as ATR kinase inhibitors
US10961232B2 (en) 2008-12-19 2021-03-30 Vertex Pharmaceuticals Incorporated Substituted pyrazines as ATR kinase inhibitors
JP2012512906A (en) * 2008-12-19 2012-06-07 バーテックス ファーマシューティカルズ インコーポレイテッド Pyrazine derivatives useful as inhibitors of ATR kinase
US20120115861A1 (en) * 2009-07-15 2012-05-10 Merck Patent Gesellschaft Mit Beschrankter Haftung Aminopyridine derivatives for treating tumors and inflammatory diseases
DE102009033208A1 (en) 2009-07-15 2011-01-20 Merck Patent Gmbh aminopyridine derivatives
JP2012532899A (en) * 2009-07-15 2012-12-20 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツング Aminopyridine derivatives for the treatment of tumors and inflammatory diseases
US8648201B2 (en) 2009-07-15 2014-02-11 Merck Patent Gmbh Aminopyridine derivatives for treating tumors and inflammatory diseases
AU2010272920B2 (en) * 2009-07-15 2016-05-05 Merck Patent Gmbh Aminopyridine derivatives for treating tumors and inflammatory diseases
WO2011006567A1 (en) 2009-07-15 2011-01-20 Merck Patent Gmbh Aminopyridine derivatives for treating tumors and inflammatory diseases
WO2011022473A1 (en) * 2009-08-19 2011-02-24 Ambit Biosciences Corporation Biaryl compounds and methods of use thereof
US20110172203A1 (en) * 2009-12-30 2011-07-14 Arqule, Inc. Substituted Imidazopyridinyl-Aminopyridine Compounds
AU2010339533B2 (en) * 2009-12-30 2015-05-07 Arqule, Inc. Substituted imidazopyridinyl-aminopyridine compounds
RU2619463C2 (en) * 2009-12-30 2017-05-16 Аркьюл, Инк. Substituted imidazopyridinyl-amino-pyridine compounds, useful for treatment of cancer
EP2519522A2 (en) * 2009-12-30 2012-11-07 ArQule, Inc. Substituted imidazopyridinyl-aminopyridine compounds
US8501770B2 (en) 2009-12-30 2013-08-06 Arqule, Inc. Substituted imidazopyridinyl-aminopyridine compounds
EP2519522A4 (en) * 2009-12-30 2013-03-13 Arqule Inc Substituted imidazopyridinyl-aminopyridine compounds
US9205086B2 (en) 2010-04-19 2015-12-08 Synta Pharmaceuticals Corp. Cancer therapy using a combination of a Hsp90 inhibitory compounds and a EGFR inhibitor
US9096584B2 (en) 2010-05-12 2015-08-04 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8969356B2 (en) 2010-05-12 2015-03-03 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9062008B2 (en) 2010-05-12 2015-06-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9334244B2 (en) 2010-05-12 2016-05-10 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9630956B2 (en) 2010-05-12 2017-04-25 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8962631B2 (en) 2010-05-12 2015-02-24 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
WO2011143422A1 (en) * 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated 2 -aminopyridine derivatives useful as inhibitors of atr kinase
WO2011143426A1 (en) * 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
US8481739B2 (en) 2010-05-17 2013-07-09 Incozen Therapeutics Pvt. Ltd. 3,5-Disubstituted-3H-imidazo[4,5-b]pyridine and 3,5-disubstituted-3H[1,2,3]triazolo [4,5-b] Pyridine Compounds as Modulators of protein kinases
US8912331B2 (en) 2010-05-17 2014-12-16 Rhizen Pharmaceuticals Sa 3,5-disubstituted-3H-imidazo[4,5-B]pyridine and 3,5-disubstituted-3H-[1,2,3]triazolo[4,5-B] pyridine compounds as modulators of protein kinases
WO2011145035A1 (en) 2010-05-17 2011-11-24 Indian Incozen Therapeutics Pvt. Ltd. Novel 3,5-disubstitued-3h-imidazo[4,5-b]pyridine and 3,5- disubstitued -3h-[1,2,3]triazolo[4,5-b] pyridine compounds as modulators of protein kinases
US10590129B2 (en) 2010-05-17 2020-03-17 Rhizen Pharmaceuticals Sa 3,5-disubstituted-3H-imidazo[4,5-B]pyridine and 3,5-disubstituted-3H-[1,2,3]triazolo[4,5-B] pyridine compounds as modulators of protein kinases
EP3450432A1 (en) 2010-05-17 2019-03-06 Incozen Therapeutics Pvt. Ltd. Novel 3,5-disubstitued-3h-imidazo[4,5-b]pyridine and 3,5- disubstitued - 3h-[1,2,3]triazolo[4,5-b] pyridine compounds as modulators of protein kinases
US10087182B2 (en) 2010-05-17 2018-10-02 Incozen Therapeutics Pvt. Ltd. 3,5-disubstituted-3H-imidazo[4,5-B]pyridine and 3,5-disubstituted-3H-[1,2,3]triazolo[4,5-B] pyridine compounds as modulators of protein kinases
US8623869B2 (en) 2010-06-23 2014-01-07 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9403815B2 (en) 2010-06-24 2016-08-02 The Regents Of The University Of California Compounds and uses thereof in modulating levels of various amyloid beta peptide alloforms
US8877759B2 (en) 2011-04-05 2014-11-04 Vertex Pharnaceuticals Incorporated Aminopyrazines as ATR kinase inhibitors
US9096602B2 (en) 2011-06-22 2015-08-04 Vertex Pharmaceuticals Incorporated Substituted pyrrolo[2,3-B]pyrazines as ATR kinase inhibitors
US9309250B2 (en) 2011-06-22 2016-04-12 Vertex Pharmaceuticals Incorporated Substituted pyrrolo[2,3-b]pyrazines as ATR kinase inhibitors
US8822469B2 (en) 2011-06-22 2014-09-02 Vertex Pharmaceuticals Incorporated Pyrrolo[2,3-B]pyrazines useful as inhibitors of ATR kinase
US8609688B2 (en) 2011-06-24 2013-12-17 Arqule, Inc. Substituted imidazopyridinyl-aminopyridine compounds
US8962619B2 (en) 2011-06-24 2015-02-24 Arqule, Inc. Substituted imidazopyridinyl-aminopyridine compounds
US8815854B2 (en) 2011-06-24 2014-08-26 Arqule, Inc. Substituted imidazopyridinyl compounds
US9035053B2 (en) 2011-09-30 2015-05-19 Vertex Pharmaceuticals Incorporated Processes for making compounds useful as inhibitors of ATR kinase
US9862709B2 (en) 2011-09-30 2018-01-09 Vertex Pharmaceuticals Incorporated Processes for making compounds useful as inhibitors of ATR kinase
US8765751B2 (en) 2011-09-30 2014-07-01 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10813929B2 (en) 2011-09-30 2020-10-27 Vertex Pharmaceuticals Incorporated Treating cancer with ATR inhibitors
US8846686B2 (en) 2011-09-30 2014-09-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10822331B2 (en) 2011-09-30 2020-11-03 Vertex Pharmaceuticals Incorporated Processes for preparing ATR inhibitors
US10208027B2 (en) 2011-09-30 2019-02-19 Vertex Pharmaceuticals Incorporated Processes for preparing ATR inhibitors
US8853217B2 (en) 2011-09-30 2014-10-07 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9439899B2 (en) 2011-11-02 2016-09-13 Synta Pharmaceuticals Corp. Cancer therapy using a combination of HSP90 inhibitors with topoisomerase I inhibitors
US10500193B2 (en) 2011-11-02 2019-12-10 Synta Pharmaceuticals Corporation Combination therapy of HSP90 inhibitors with platinum-containing agents
US8841450B2 (en) 2011-11-09 2014-09-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
WO2013071088A1 (en) * 2011-11-09 2013-05-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
US8841337B2 (en) 2011-11-09 2014-09-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8841449B2 (en) 2011-11-09 2014-09-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
WO2013071090A1 (en) * 2011-11-09 2013-05-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
US8846917B2 (en) 2011-11-09 2014-09-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8846918B2 (en) 2011-11-09 2014-09-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
EP2805946A4 (en) * 2012-01-17 2015-07-29 Shanghai Allist Pharmaceuticals Inc Amino heteroaryl compound, preparation method therefor and use thereof
WO2013144737A2 (en) 2012-03-30 2013-10-03 Rhizen Pharmaceuticals Sa Novel 3,5-disubstitued-3h-imidazo[4,5-b]pyridine and 3,5- disubstitued -3h-[1,2,3]triazolo[4,5-b] pyridine compounds as modulators of c-met protein kinases
US11066402B2 (en) 2012-03-30 2021-07-20 Rhizen Pharmaceuticals Sa 3,5-disubstituted-3H-imidazo[4,5-b]pyridine and 3,5-disubstituted-3H-[1,2,3]triazolo[4,5-B] pyridine compounds as modulators of c-Met protein kinases
US9815831B2 (en) 2012-03-30 2017-11-14 Rhizen Pharmaceuticals Sa 3,5-disubstituted-3H-imidazo[4,5-B]pyridine and 3,5-disubstituted-3H-[1,2,3]triazolo[4,5-B] pyridine compounds as modulators of c-Met protein kinases
US10478430B2 (en) 2012-04-05 2019-11-19 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase and combination therapies thereof
US11110086B2 (en) 2012-04-05 2021-09-07 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase and combination therapies thereof
US9791456B2 (en) 2012-10-04 2017-10-17 Vertex Pharmaceuticals Incorporated Method for measuring ATR inhibition mediated increases in DNA damage
US8912198B2 (en) 2012-10-16 2014-12-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9340546B2 (en) 2012-12-07 2016-05-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9650381B2 (en) 2012-12-07 2017-05-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10787452B2 (en) 2012-12-07 2020-09-29 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9718827B2 (en) 2012-12-07 2017-08-01 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10392391B2 (en) 2012-12-07 2019-08-27 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US11370798B2 (en) 2012-12-07 2022-06-28 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US11117900B2 (en) 2012-12-07 2021-09-14 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9393248B1 (en) 2013-01-02 2016-07-19 H. Lundbeck A/S Aminopyridine derived compounds as LRRK2 inhibitors
WO2014106612A1 (en) * 2013-01-02 2014-07-10 H. Lundbeck A/S Aminopyridine derived compounds as lrrk2 inhibitors
US9321748B2 (en) 2013-01-02 2016-04-26 H. Lundbeck A/S Aminopyridine derived compounds as LRRK2 inhibitors
US9657008B2 (en) 2013-01-23 2017-05-23 Astrazeneca Ab Chemical compounds
US9156831B2 (en) 2013-01-23 2015-10-13 Astrazeneca Ab Chemical compounds
US9938263B2 (en) 2013-03-12 2018-04-10 The General Hospital Corporation Gamma-secretase modulators
US10017516B2 (en) 2013-03-14 2018-07-10 The Brigham And Women's Hospital, Inc. BMP inhibitors and methods of use thereof
US9663519B2 (en) 2013-03-15 2017-05-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10815239B2 (en) 2013-12-06 2020-10-27 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10160760B2 (en) 2013-12-06 2018-12-25 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US11485739B2 (en) 2013-12-06 2022-11-01 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
RU2677478C2 (en) * 2014-03-24 2019-01-17 Аркьюл, Инк. Process of preparing 3-(3-(4-(1-aminocyclobutyl)phenyl)-5-phenyl-3h-imidazo[4,5-b]pyridin-2-yl)pyridin-2-amine
US9834556B2 (en) 2014-03-24 2017-12-05 Arqule, Inc. Process of preparing 3-(3-(4-aminocyclobutyl)phenyl)-5-phenyl-3H-imidazo[4,5-B]pyridin-2-YL)pyridin-2-amine
US9464083B2 (en) 2014-03-24 2016-10-11 Arqule Inc. Process of preparing 3-(3-(4-(1-aminocyclobutyl)phenyl)-5-phenyl-3H-imidazo[4,5-B]pyridin-2-yl)pyridin-2-amine
WO2015148464A1 (en) * 2014-03-24 2015-10-01 Arqule Inc. Process of preparing 3-(3-(4-(1-aminocyclobutyl)phenyl)-5-phenyl-3h-imidazo[4,5-b]pyridin-2-yl)pyridin-2-amine
EP3122355A4 (en) * 2014-03-26 2017-08-09 The Brigham and Women's Hospital, Inc. Compositions and methods for inhibiting bmp
RU2732125C2 (en) * 2014-04-22 2020-09-11 Аркьюл, Инк. Salts and polymorphs of substituted imidazopyridinyl-aminopyridine
WO2015164479A1 (en) * 2014-04-22 2015-10-29 Arqule, Inc. Salts and polymorphs of a substituted imidazopyridinyl-aminopyridine compound
US9422283B2 (en) 2014-04-22 2016-08-23 Arqule, Inc. Salts and polymorphs of a substituted imidazopyridinyl-aminopyridine compound
US10112926B2 (en) 2014-04-24 2018-10-30 Novartis Ag Amino pyridine derivatives as phosphatidylinositol 3-kinase inhibitors
US10004732B2 (en) 2014-04-24 2018-06-26 Novartis Ag Amino pyrazine derivatives as phosphatidylinositol 3-kinase inhibitors
US9862711B2 (en) 2014-04-24 2018-01-09 Novartis Ag Pyrazine derivatives as phosphatidylinositol 3-kinase inhibitors
US10800781B2 (en) 2014-06-05 2020-10-13 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9670215B2 (en) 2014-06-05 2017-06-06 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10093676B2 (en) 2014-06-05 2018-10-09 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US11179394B2 (en) 2014-06-17 2021-11-23 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of Chk1 and ATR inhibitors
US10513521B2 (en) 2014-07-15 2019-12-24 The Brigham And Women's Hospital, Inc. Compositions and methods for inhibiting BMP
EP3201187A4 (en) * 2014-10-01 2018-04-11 The Brigham and Women's Hospital, Inc. Compositions and methods for inhibiting bmp
US10472346B2 (en) 2014-10-31 2019-11-12 The General Hospital Corporation Potent gamma-secretase modulators
US11117884B2 (en) 2014-10-31 2021-09-14 The General Hospital Corporation Potent gamma-secretase modulators
US11464774B2 (en) 2015-09-30 2022-10-11 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of DNA damaging agents and ATR inhibitors
WO2019038717A1 (en) 2017-08-23 2019-02-28 Novartis Ag 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
US11926616B2 (en) 2018-03-08 2024-03-12 Incyte Corporation Aminopyrazine diol compounds as PI3K-γ inhibitors
US11046658B2 (en) 2018-07-02 2021-06-29 Incyte Corporation Aminopyrazine derivatives as PI3K-γ inhibitors
WO2020188015A1 (en) 2019-03-21 2020-09-24 Onxeo A dbait molecule in combination with kinase inhibitor for the treatment of cancer
WO2021089791A1 (en) 2019-11-08 2021-05-14 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment of cancers that have acquired resistance to kinase inhibitors
CN111116585A (en) * 2019-12-31 2020-05-08 北京鑫开元医药科技有限公司 Compound with c-MET kinase inhibitory activity, preparation method, composition and application
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use

Also Published As

Publication number Publication date
MX2008012096A (en) 2008-12-17
NO20084435L (en) 2008-11-28
KR20090007347A (en) 2009-01-16
CA2646701A1 (en) 2007-10-04
ES2336625T3 (en) 2010-04-14
NZ571933A (en) 2011-06-30
EP2004625B1 (en) 2009-12-30
US8232406B2 (en) 2012-07-31
AU2007231577A1 (en) 2007-10-04
CN101479255B (en) 2013-05-01
JP2014132024A (en) 2014-07-17
JP2009533327A (en) 2009-09-17
US20070254868A1 (en) 2007-11-01
RU2008141761A (en) 2010-04-27
WO2007111904A3 (en) 2007-12-13
AR060054A1 (en) 2008-05-21
CL2007000752A1 (en) 2008-02-08
CN101479255A (en) 2009-07-08
ATE453635T1 (en) 2010-01-15
US7872031B2 (en) 2011-01-18
IL194242A0 (en) 2009-08-03
TW200806646A (en) 2008-02-01
DE602007004092D1 (en) 2010-02-11
AU2007231577B2 (en) 2012-07-19
EP2004625A2 (en) 2008-12-24
HK1129890A1 (en) 2009-12-11
US20110059936A1 (en) 2011-03-10

Similar Documents

Publication Publication Date Title
EP2004625B1 (en) C-met protein kinase inhibitors for the treatment of proliferative disorders
US8518938B2 (en) c-MET protein kinase inhibitors
EP2205589B1 (en) C-met protein kinase inhibitors
AU2010336386B2 (en) Aminopyrimidine kinase inhibitors
EP2858983B1 (en) Tnf-alpha modulating benzimidazoles
JP2012519177A (en) Tricyclic pyrazolopyridine kinase inhibitor
EP2435443B1 (en) Aminopyrazole triazolothiadiazole inhibitors of c-met protien kinase
US8263777B2 (en) Aminopyrazole triazolothiadiazole inhibitor of c-Met protein kinase

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200780018208.4

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 2646701

Country of ref document: CA

Ref document number: 2007753626

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2009501540

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: MX/a/2008/012096

Country of ref document: MX

Ref document number: 194242

Country of ref document: IL

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2007231577

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 571933

Country of ref document: NZ

Ref document number: 4102/KOLNP/2008

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 1020087025755

Country of ref document: KR

ENP Entry into the national phase

Ref document number: 2008141761

Country of ref document: RU

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2007231577

Country of ref document: AU

Date of ref document: 20070321

Kind code of ref document: A