WO2009158571A1 - Heteroaryl compounds and uses thereof - Google Patents

Heteroaryl compounds and uses thereof Download PDF

Info

Publication number
WO2009158571A1
WO2009158571A1 PCT/US2009/048784 US2009048784W WO2009158571A1 WO 2009158571 A1 WO2009158571 A1 WO 2009158571A1 US 2009048784 W US2009048784 W US 2009048784W WO 2009158571 A1 WO2009158571 A1 WO 2009158571A1
Authority
WO
WIPO (PCT)
Prior art keywords
ring
independently selected
nitrogen
oxygen
sulfur
Prior art date
Application number
PCT/US2009/048784
Other languages
French (fr)
Other versions
WO2009158571A8 (en
Inventor
Arthur F. Kluge
Russell C. Petter
Richland Wayne Tester
Lixin Qiao
Deqiang Niu
William Frederick Westlin
Juswinder Singh
Hormoz Mazdiyasni
Original Assignee
Avila Therapeutics And Uses Thereof
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 MX2015012477A priority Critical patent/MX360970B/en
Priority to KR1020187011709A priority patent/KR101955914B1/en
Priority to AU2009262068A priority patent/AU2009262068C1/en
Priority to BRPI0914682A priority patent/BRPI0914682B8/en
Priority to DK09771102.2T priority patent/DK2361248T3/en
Priority to CN2009801244104A priority patent/CN102083800A/en
Priority to CA2727455A priority patent/CA2727455C/en
Priority to NZ589843A priority patent/NZ589843A/en
Priority to MX2013013212A priority patent/MX357627B/en
Priority to MX2010014029A priority patent/MX2010014029A/en
Priority to KR1020167034131A priority patent/KR101892989B1/en
Priority to EP09771102.2A priority patent/EP2361248B1/en
Priority to ES09771102T priority patent/ES2711249T3/en
Priority to EP18195292.0A priority patent/EP3549934A1/en
Application filed by Avila Therapeutics And Uses Thereof filed Critical Avila Therapeutics And Uses Thereof
Priority to JP2011516699A priority patent/JP2011526299A/en
Priority to RU2010151355/04A priority patent/RU2536584C2/en
Publication of WO2009158571A1 publication Critical patent/WO2009158571A1/en
Publication of WO2009158571A8 publication Critical patent/WO2009158571A8/en
Priority to IL209969A priority patent/IL209969A/en
Priority to ZA2010/09216A priority patent/ZA201009216B/en
Priority to IL230290A priority patent/IL230290A/en
Priority to PH12015501484A priority patent/PH12015501484A1/en
Priority to IL250108A priority patent/IL250108A0/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/5381,4-Oxazines, e.g. morpholine ortho- or peri-condensed with carbocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • 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
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs 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
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/47One nitrogen atom and one oxygen or sulfur atom, e.g. cytosine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/48Two nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D403/12Heterocyclic 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 linked by a chain containing hetero atoms as chain links
    • 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
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D407/02Heterocyclic 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 two hetero rings
    • C07D407/12Heterocyclic 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 two hetero rings linked by a chain containing hetero atoms as chain links
    • 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
    • C07D413/12Heterocyclic 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 linked by a chain containing hetero atoms as chain links
    • 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
    • C07D417/14Heterocyclic 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
    • 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
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains three hetero rings
    • C07D487/18Bridged systems

Definitions

  • the present invention relates to compounds useful as inhibitors of protein kinases.
  • the invention also provides pharmaceutically acceptable compositions comprising compounds of the present invention and methods of using said compositions in the treatment of various disorders.
  • Protein kinases constitute a large family of structurally related enzymes that are responsible for the control of a variety of signal transduction processes within the cell. Protein kinases are thought to have evolved from a common ancestral gene due to the conservation of their structure and catalytic function. Almost all kinases contain a similar 250-300 amino acid catalytic domain. The kinases may be categorized into families by the substrates they phosphorylate (e.g., protein-tyrosine, protein-serine/threonine, lipids, etc.).
  • protein kinases mediate intracellular signaling by effecting a phosphoryl transfer from a nucleoside triphosphate to a protein acceptor that is involved in a signaling pathway. These phosphorylation events act as molecular on/off switches that can modulate or regulate the target protein biological function. These phosphorylation events are ultimately triggered in response to a variety of extracellular and other stimuli.
  • Examples of such stimuli include environmental and chemical stress signals (e.g., osmotic shock, heat shock, ultraviolet radiation, bacterial endotoxin, and H 2 O 2 ), cytokines (e.g., interleukin-1 (IL-I) and tumor necrosis factor ⁇ (TNF- ⁇ )), and growth factors (e.g., granulocyte macrophage-colony-stimulating factor (GM-CSF), and fibroblast growth factor (FGF)).
  • IL-I interleukin-1
  • TNF- ⁇ tumor necrosis factor ⁇
  • growth factors e.g., granulocyte macrophage-colony-stimulating factor (GM-CSF), and fibroblast growth factor (FGF)
  • An extracellular stimulus may affect one or more cellular responses related to cell growth, migration, differentiation, secretion of hormones, activation of transcription factors, muscle contraction, glucose metabolism, control of protein synthesis, and regulation of the cell cycle.
  • Compounds of the present invention are useful for treating a variety of diseases, disorders or conditions, associated with abnormal cellular responses triggered by protein kinase-mediated events. Such diseases, disorders, or conditions include those described herein.
  • Compounds provided by this invention are also useful for the study of kinases in biological and pathological phenomena; the study of intracellular signal transduction pathways mediated by such kinases; and the comparative evaluation of new kinase inhibitors.
  • Figure 1 depicts dose-response inhibition of phospho-plc gamma2 (p-plc gamma 2) with compound 1-2 in Ramos Cells; and the results of compound 1-2 in a "washout” experiment.
  • Figure 2 depicts dose-response inhibition of p-plc gamma2 with compound 1-4 in Ramos Cells; and the results of compound 1-4 in a "washout” experiment.
  • Figure 3 depicts dose response inhibition of p-plc gamma2 with compound 1-7 in Ramos cells; and the results of compound 1-7 in a "washout” experiment.
  • Figure 4 depicts dose response inhibition of p-plc gamma2 with compound 1-35 in Ramos cells.
  • Figure 5 depicts dose response inhibition of p-plc gamma2 with compound 1-38 in Ramos cells.
  • Figure 6 depicts MS analysis confirming covalent modification of TEC kinase at Cys449 by compound 1-2.
  • Figure 7 depicts MS analysis confirming covalent modification of TEC kinase at Cys449 by compound 1-4.
  • Figure 8 depicts MS analysis confirming covalent modification of TEC kinase at Cys449 by compound 1-7.
  • Figure 9 depicts the results of compound 1-2 in a "washout” experiment as compared to results of compound 1-4 and compound 1-7 in the same "washout” experiment in HCC827 cells containing EGFR deletion mutant.
  • Figure 10 depicts the results of compound 1-7 in a "washout" experiment as compared to results of an EGF control in A431 cells containing EGFR wild type.
  • Figure 11 depicts MS analysis confirming covalent modification of JAK-3 kinase at Cys909 by compound 1-7.
  • Figure 12 depicts dose-response inhibition of P-Stat5 with compound 1-2 in IL-2 stimulated
  • Figure 13 depicts dose-response inhibition of P-Stat5 with compound 1-4 in IL-2 stimulated
  • Figure 14 depicts dose-response inhibition of P-Stat5 with compound 1-7 in IL-2 stimulated
  • Figure 15 depicts MS analysis confirming covalent modification of BTK by compound 1-7.
  • Figure 16 depicts a Western blot showing BTK protein available to the probe compound 1-215 after treating with varying amounts of 1-7.
  • Figure 17 depicts quantitation of the Western blot results in Figure 16.
  • Figure 18 depicts a Western blot for a washout experiment with compound 1-7 and probe compound 1-215.
  • Figure 19 depicts quantitation of the Western blot results in Figure 18.
  • Figure 20 depicts an amino acid sequence for BTK (SEQ ID 1).
  • Figure 21 depicts an amino acid sequence for TEC (SEQ ID 2).
  • Figure 22 depicts an amino acid sequence for ITK (SEQ ID 3).
  • Figure 23 depicts an amino acid sequence for BMX (SEQ ID 4).
  • Figure 24 depicts an amino acid sequence for TXK (SEQ ID 5).
  • Figure 25 depicts an amino acid sequence for JAK3 (SEQ ID 6).
  • the present invention provides a compound of formula I-a or I-b:
  • Ring A is an optionally substituted group selected from phenyl, a 3-7 membered saturated or partially unsaturated carbocyclic ring, an 8-10 membered bicyclic saturated, partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • Ring B is an optionally substituted group selected from phenyl, a 3-7 membered saturated or partially unsaturated carbocyclic ring, an 8-10 membered bicyclic saturated, partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • R 1 is a warhead group;
  • R y is hydrogen, halogen, -CN, -CF 3 , Ci_4 aliphatic, Ci_4 haloaliphatic, -OR, -C(O)R,
  • each R group is independently hydrogen or an optionally substituted group selected from Ci_ 6 aliphatic, phenyl, a 4-7 membered heterocylic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • W 1 and W 2 are each independently a covalent bond or a bivalent Ci_ 3 alkylene chain wherein one methylene unit of W 1 or W 2 is optionally replaced by -NR 2 -, -N(R 2 )C(O)-, -C(O)N(R 2 )-, -N(R 2 )SO 2 -, -SO 2 N(R 2 )-, -O-, -C(O)-, -OC(O)-, -C(O)O-, -S-, -SO- or -SO 2 -;
  • R 2 is hydrogen,
  • R 2 and a substituent on Ring A are taken together with their intervening atoms to form a
  • R x and R v are independently selected from -R, halogen, -OR, -0(CH 2 ) q 0R, -CN, -NO 2 , -SO 2 R, -SO 2 N(R) 2 , -SOR, -C(O)R, -CO 2 R, -C(O)N(R) 2 , -NRC(O)R, -NRC(O)NR 2 , -NRSO 2 R, or -N(R) 2 , wherein q is 1-4; or:
  • R x and R 1 when concurrently present on Ring B are taken together with their intervening atoms to form a 5-7 membered saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with a warhead group and 0-3 groups independently selected from oxo, halogen, -CN, or Ci_6 aliphatic; or R v and R 1 when concurrently present on Ring A are taken together with their intervening atoms to form a 5-7 membered saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with a warhead group and 0-3 groups independently selected from oxo, halogen, -CN, or Ci_6 aliphatic.
  • 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, or a monocyclic hydrocarbon or bicyclic hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic (also referred to herein as “carbocycle” "cycloaliphatic” or “cycloalkyl”), that has a single point of attachment to the rest of the molecule.
  • aliphatic groups contain 1-6 aliphatic carbon atoms.
  • aliphatic groups contain 1-5 aliphatic carbon atoms. In other embodiments, aliphatic groups contain 1-4 aliphatic carbon atoms. In still other embodiments, aliphatic groups contain 1-3 aliphatic carbon atoms, and in yet other embodiments, aliphatic groups contain 1-2 aliphatic carbon atoms.
  • cycloaliphatic (or “carbocycle” or “cycloalkyl”) refers to a monocyclic C 3 -C 6 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.
  • Suitable aliphatic groups include, but are not limited to, linear or branched, substituted or unsubstituted alkyl, alkenyl, alkynyl groups and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl or (cycloalkyl)alkenyl.
  • the term "lower alkyl” refers to a Ci_ 4 straight or branched alkyl group.
  • Exemplary lower alkyl groups are methyl, ethyl, propyl, isopropyl, butyl, isobutyl, and tert-butyl.
  • lower haloalkyl refers to a Ci_ 4 straight or branched alkyl group that is substituted with one or more halogen atoms.
  • heteroatom means one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon (including, any oxidized form of nitrogen, sulfur, phosphorus, or silicon; 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)).
  • Ci_8 (or C 1-6 ) saturated or unsaturated, straight or branched, hydrocarbon chain
  • bivalent alkylene, alkenylene, and alkynylene chains that are straight or branched as defined herein.
  • alkylene refers to a bivalent alkyl group.
  • An "alkylene chain” is a polymethylene group, i.e., -(CH 2 ) n -, wherein n is a positive integer, preferably from 1 to 6, from
  • a substituted alkylene chain is a polymethylene group in which one or more methylene hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group.
  • alkenylene refers to a bivalent alkenyl group.
  • a substituted alkenylene chain is a polymethylene group containing at least one double bond in which one or more hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group.
  • cyclopropylenyl refers to a bivalent cyclopropyl group of
  • halogen means F, Cl, Br, or I.
  • aryl used alone or as part of a larger moiety as in “aralkyl”, “aralkoxy”, or
  • aryloxyalkyl refers to monocyclic and bicyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic and wherein each ring in the system contains three to seven ring members.
  • aryl may be used interchangeably with the term “aryl ring”.
  • aryl refers to an aromatic ring system which includes, but not limited to, phenyl, biphenyl, naphthyl, anthracyl and the like, which may bear one or more substituents.
  • aryl is a group in which an aromatic ring is fused to one or more non-aromatic rings, such as indanyl, phthalimidyl, naphthimidyl, phenanthridinyl, or tetrahydronaphthyl, and the like.
  • heteroaryl and “heteroar-”, used alone or as part of a larger moiety refer to groups having 5 to 10 ring atoms, preferably 5, 6, or 9 ring atoms; having 6, 10, or 14 ⁇ electrons shared in a cyclic array; and having, in addition to carbon atoms, from one to five heteroatoms.
  • heteroatom refers to nitrogen, oxygen, or sulfur, and includes any oxidized form of nitrogen or sulfur, and any quaternized form of a basic nitrogen.
  • Heteroaryl groups include, without limitation, thienyl, furanyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, purinyl, naphthyridinyl, and pteridinyl.
  • heteroaryl and “heteroar-”, as used herein, also include groups in which a heteroaromatic ring is fused to one or more aryl, cycloaliphatic, or heterocyclyl rings, where the radical or point of attachment is on the heteroaromatic ring.
  • Nonlimiting examples include indolyl, isoindolyl, benzothienyl, benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzthiazolyl, quinolyl, isoquinolyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4H-quinolizinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, and pyrido[2,3-b]-l,4-oxazin- 3(4 ⁇ )-one.
  • heteroaryl group may be mono- or bicyclic.
  • heteroaryl may be used interchangeably with the terms “heteroaryl ring”, “heteroaryl group”, or “heteroaromatic”, any of which terms include rings that are optionally substituted.
  • heteroarylkyl refers to an alkyl group substituted by a heteroaryl, wherein the alkyl and heteroaryl portions independently are optionally substituted.
  • heterocycle As used herein, the terms “heterocycle”, “heterocyclyl”, “heterocyclic radical”, and “heterocyclic ring” are used interchangeably and refer to a stable 5- to 7-membered monocyclic or 7-10-membered bicyclic heterocyclic moiety that is either saturated or partially unsaturated, and having, in addition to carbon atoms, one or more, preferably one to four, heteroatoms, as defined above.
  • nitrogen includes a substituted nitrogen.
  • the nitrogen may be N (as in 3,4- dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl), or + NR (as in TV-substituted pyrrolidinyl).
  • a heterocyclic ring can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure and any of the ring atoms can be optionally substituted.
  • saturated or partially unsaturated heterocyclic radicals include, without limitation, tetrahydrofuranyl, tetrahydrothiophenyl pyrrolidinyl, piperidinyl, pyrrolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, diazepinyl, oxazepinyl, thiazepinyl, morpholinyl, and quinuclidinyl.
  • heterocycle refers to an alkyl group substituted by a heterocyclyl, wherein the alkyl and heterocyclyl portions independently are optionally substituted.
  • partially unsaturated refers to a ring moiety that includes at least one double or triple bond.
  • partially unsaturated is intended to encompass rings having multiple sites of unsaturation, but is not intended to include aryl or heteroaryl moieties, as herein defined.
  • compounds of the invention may contain "optionally substituted” moieties.
  • substituted whether preceded by the term “optionally” or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent.
  • an “optionally substituted” group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position.
  • 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, in certain embodiments, their recovery, purification, and use for one or more of the purposes disclosed herein.
  • Suitable monovalent substituents on R 0 are independently halogen, -(CH 2 ) 0 2 R*, -(haloR*), -(CH 2 V 2 OH, -(CH 2 ) 0 2 OR*, -(CH 2 ) 0 2 CH(OR*) 2 ; -O(haloR'), -CN, -N 3 , -(CH 2 ) 0 2 C(O)R*, -(CH 2 ) 0 2 C(O)OH, -(CH 2 ) 0 2 C(O)OR*, -(CH 2 ) 0 2 SR*, -(CH 2 )o 2 SH, -(CH 2 )o 2 NH 2 , -(CH 2 ) 0 2 NHR*, -(CH 2 ) 0 2 NR* 2 , -NO 2 , -
  • Suitable divalent substituents that are bound to vicinal substitutable carbons of an "optionally substituted” group include: -O(CR 2 ) 2 _ 3 O-, wherein each independent occurrence of R is selected from hydrogen, C i_6 aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Suitable substituents on the aliphatic group of R * include halogen, -R*, -(haloR*), -OH, -OR*, -O(haloR'), -CN, -C(O)OH, -C(O)OR*, -NH 2 , -NHR*, -NR* 2 , or -NO 2 , wherein each R* is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently Ci_ 4 aliphatic, -CH 2 Ph, -O(CH 2 ) 0 iPh, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Suitable substituents on a substitutable nitrogen of an "optionally substituted" group include -S(O) 2 R 1 ; -S(O) 2 NR 1 Z, -C(S)NR 1 Z, -C(NH)NR 1 Z, or -N(R t )S(O) 2 R t ; wherein each R f is independently hydrogen, Ci_6 aliphatic which may be substituted as defined below, unsubstituted -OPh, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of taken together with their intervening atom(s) form an unsubstituted 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Suitable substituents on the aliphatic group of R ⁇ are independently halogen, -R*, -(haloR*), -OH, -OR*, -O(haloR'), -CN, -C(O)OH, -C(O)OR*, -NH 2 , -NHR*, -NR* 2 , or -NO 2 , wherein each R* is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently Ci_4 aliphatic, -CH 2 Ph, -0(CH 2 )o iPh, or a 5-6- membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • 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, and are commensurate with a reasonable benefit/risk ratio.
  • 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, 1977, 66, 1-19, 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.
  • 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 salts.
  • 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, loweralkyl sulfonate and aryl sulfonate.
  • 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. 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 including 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, as probes in biological assays, or as therapeutic agents in accordance with the present invention.
  • the R 1 group of formula I-a and I-b comprises one or more deuterium atoms.
  • the term "irreversible” or “irreversible inhibitor” refers to an inhibitor (i.e. a compound) that is able to be covalently bonded to a target protein kinase in a substantially non-reversible manner. That is, whereas a reversible inhibitor is able to bind to (but is generally unable to form a covalent bond) the target protein kinase, and therefore can become dissociated from the target protein kinase, an irreversible inhibitor will remain substantially bound to the target protein kinase once covalent bond formation has occurred. Irreversible inhibitors usually display time dependency, whereby the degree of inhibition increases with the time with which the inhibitor is in contact with the enzyme.
  • Such methods include, but are not limited to, enzyme kinetic analysis of the inhibition profile of the compound with the protein kinase target, the use of mass spectrometry of the protein drug target modified in the presence of the inhibitor compound, discontinuous exposure, also known as "washout," experiments, and the use of labeling, such as radiolabeled inhibitor, to show covalent modification of the enzyme, as well as other methods known to one of skill in the art.
  • warheads refers to a functional group present on a compound of the present invention wherein that functional group is capable of covalently binding to an amino acid residue (such as cysteine, lysine, histidine, or other residues capable of being covalently modified) present in the binding pocket of the target protein, thereby irreversibly inhibiting the protein.
  • an amino acid residue such as cysteine, lysine, histidine, or other residues capable of being covalently modified
  • an inhibitor is defined as a compound that binds to and /or inhibits the target protein kinase with measurable affinity.
  • an inhibitor has an IC 50 and/or binding constant of less about 50 ⁇ M, less than about 1 ⁇ M, less than about 500 nM, less than about 100 nM, or less than about 10 nM.
  • measurable affinity means a measurable change in at least one of ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3 activity between a sample comprising a compound of the present invention, or composition thereof, and at least one of ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3, and an equivalent sample comprising at least one of ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3, in the absence of said compound, or composition thereof.
  • the present invention provides a compound of formula I-a or I-b,
  • Ring A is an optionally substituted group selected from phenyl, a 3-7 membered saturated or partially unsaturated carbocyclic ring, an 8-10 membered bicyclic saturated, partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • Ring B is an optionally substituted group selected from phenyl, a 3-7 membered saturated or partially unsaturated carbocyclic ring, an 8-10 membered bicyclic saturated, partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • R 1 is -L-Y, wherein:
  • Y is hydrogen, Ci_6 aliphatic optionally substituted with oxo, halogen, or CN, or a 3-10 membered monocyclic or bicyclic, saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, and wherein said ring is substituted with at 1-4 groups independently selected from -Q-Z, oxo, NO 2 , halogen, CN, or Ci_6 aliphatic, wherein:
  • Q is a covalent bond or a bivalent Ci_ 6 saturated or unsaturated, straight or branched, hydrocarbon chain, wherein one or two methylene units of Q are optionally and independently replaced by -NR-, -S-, -O-, -C(O)-, -SO-, or -SO 2 -; and Z is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, or CN; R y is hydrogen, halogen, -CN, -CF 3 , Ci_ 4 aliphatic, Ci_ 4 haloaliphatic, -OR, -C(O)R, or -C(O)N(R) 2 ; each R group is independently hydrogen or an optionally substituted group selected from Ci_ 6 aliphatic, phenyl, a 4-7 membered heterocylic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered monocyclic heteroaryl ring having
  • R 2 and a substituent on Ring A are taken together with their intervening atoms to form a 4-6 membered partially unsaturated or aromatic fused ring; or R 2 and R y are taken together with their intervening atoms to form a 4-6 membered saturated, partially unsaturated, or aromatic fused ring; m and p are independently 0-4; and
  • R x and R v are independently selected from -R, halogen, -OR, -0(CH 2 ) q 0R, -CN, -NO 2 , -SO 2 R, -SO 2 N(R) 2 , -SOR, -C(O)R, -CO 2 R, -C(O)N(R) 2 , -NRC(O)R, -NRC(O)NR 2 , -NRSO 2 R, or -N(R) 2 , or:
  • R x and R 1 when concurrently present on Ring B are taken together with their intervening atoms to form a 5-7 membered saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with a warhead group and 0-3 groups independently selected from oxo, halogen, -CN, or Ci_6 aliphatic; or
  • R v and R 1 when concurrently present on Ring A are taken together with their intervening atoms to form a 5-7 membered saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with a warhead group and 0-3 groups independently selected from oxo, halogen, -CN, or Ci_ 6 aliphatic.
  • Ring A is an optionally substituted group selected from phenyl, a 3-7 membered saturated or partially unsaturated carbocyclic ring, an 8-10 membered bicyclic saturated, partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Ring A is an optionally substituted phenyl group. In some embodiments, Ring A is an optionally substituted naphthyl ring or a bicyclic 8-10 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In certain other embodiments, Ring A is an optionally substituted 3-7 membered carbocyclic ring. In yet other embodiments, Ring A is an optionally substituted 4-7 membered heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Ring A is substituted as defined herein.
  • Ring A is substituted with one, two, or three groups independently selected from halogen, R 0 , or -(CH 2 )O ⁇ tOR 0 , or -0(CH 2 )(MR 0 , wherein each R 0 is as defined herein.
  • Exemplary substituents on Ring A include Br, I, Cl, methyl, -CF 3 , -C ⁇ CH, -OCH 2 phenyl, -OCH 2 (fluorophenyl), or -OCH 2 pyridyl.
  • Exemplary Ring A groups are set forth in Table 1. Table 1. Exemplary Ring A Groups
  • Ring A is selected from i, ii, iv, v, vi, vii, ix, xiv, xvi, Hi, Ixiii,
  • Ring B is an optionally substituted group selected from phenyl, a 3-7 membered saturated or partially unsaturated carbocyclic ring, an 8-10 membered bicyclic saturated, partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Ring B is an optionally substituted phenyl group. In some embodiments, Ring B is an optionally substituted naphthyl ring or a bicyclic 8-10 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In certain other embodiments, Ring B is an optionally substituted 3-7 membered carbocyclic ring. In yet other embodiments, Ring B is an optionally substituted 4-7 membered heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Ring B is phenyl. In some embodiments, Ring B is a 6- membered heteroaryl ring having 1-3 nitrogens. In some embodiments, Ring B is a 5-membered heteroaryl ring having 1 or 2 or 3 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Ring B is a 5-6 membered saturated heterocyclic ring having 1 nitrogen. In some embodiments, Ring B is a 9-10 membered bicyclic partially saturated heteroaryl ring having 1-3 nitrogens. In some embodiments, Ring B is a 9-10 membered bicyclic partially saturated heteroaryl ring having 1 nitrogen. In some embodiments, Ring B is a 9-10 membered bicyclic partially saturated heteroaryl ring having 1 nitrogen and 1 oxygen.
  • Ring B is an optionally substituted group selected from phenyl, pyridyl, pyrazinyl, pyrimidinyl, imidazolyl, pyrrolidinyl, piperdinyl, indolinyl, indazolyl, and isoindolinyl.
  • Ring B groups are set forth in Table 4. Table 2. Ring B Groups
  • Ring B is selected from i, H, Hi, iv, v, ix, x, xi, xiii, xvi, xvii, jcijc, jcjc, jcjcv, jcjcvi, JCJCJCH, JCJCJCiV, JCJCJCV, jcjcjcviii, JCHi, xlvi, xlviii, I, Iviii, Ixiv, Ixxviii, Ixxxiii, Ixxxvi, xciv, c, ci, cii, ciii, civ, and cv.
  • the m moiety of formula I is 1, 2, 3 or 4.
  • m is 1. In other embodiments, m is 0.
  • the p moiety of formula I is 1, 2, 3 or 4. In some embodiments, p is 1. In other embodiments, p is 0.
  • each R x group of formula I is independently selected from -R, halogen, -OR, -O(CH 2 ) q OR, -CN, -NO 2 , -SO 2 R, -SO 2 N(R) 2 , -SOR, -C(O)R, -CO 2 R, -
  • each instance of R x is independently selected from -R, -OR, -
  • R x is lower alkyl, lower alkoxy, lower alkoxyalkoxy, or halogen.
  • exemplary R x groups include methyl, methoxy, methoxyethoxy and fluoro.
  • R x is hydrogen.
  • each R v group of formula I is independently selected from -R, halogen, -OR, -O(CH 2 ) q OR, -CN, -NO 2 , -SO 2 R, -SO 2 N(R) 2 , -SOR, -C(O)R, -CO 2 R, -
  • each instance of R v is independently selected from -R, -OR, -
  • R v is lower alkyl, lower alkoxy, lower alkoxyalkoxy, or halogen.
  • exemplary R v groups include methyl, methoxy, trifluoromethyl, methoxyethoxy, and chloro.
  • R v is hydrogen.
  • the q moiety is 1, 2, 3, or 4. In certain embodiments, q is 1.
  • q is 2.
  • R y is hydrogen, halogen, -CN, -CF 3 , Ci_4 aliphatic, Ci_4 haloaliphatic, -OR, -C(O)R, or -C(O)N(R) 2 , where R is as defined above and described herein.
  • R y is hydrogen, halogen, -CN, -CF 3 , lower alkyl or lower haloalkyl, - C ⁇ CR and cyclopropyl.
  • R y is -OR, -C(O)R, or -C(O)N(R) 2 .
  • R y is -OCH 3 .
  • R y is -C(O)CH 3 .
  • R y is -C(O)NHR.
  • R y is hydrogen.
  • R y is fluorine.
  • R y is methyl.
  • W 1 and W 2 are each independently a covalent bond or a bivalent Ci_ 3 alkylene chain wherein one methylene unit of W 1 or W 2 is optionally replaced by -NR 2 -, -N(R 2 )C(0)-, -C(O)N(R 2 )-, -N(R 2 )SO 2 -, -SO 2 N(R 2 )-, -0-, -C(O)-, -OC(O)-, -C(O)O-, -S-, -SO- or -SO 2 -.
  • W 1 and W 2 are the same. In some embodiments, W 1 and W 2 are different.
  • W 1 is a covalent bond.
  • W 1 is a bivalent Ci_ 3 alkylene chain wherein one methylene unit of W 1 is optionally replaced by -NR 2 -, -N(R 2 )C(0)-, -C(O)N(R 2 )-, -N(R 2 )SO 2 -, -SO 2 N(R 2 )-, -0-, -C(O)-, -OC(O)-, -C(O)O-, -S-, -SO- or -SO 2 -.
  • W 1 is -NR 2 -. In other embodiments, W 1 is -0-. In certain embodiments, W 1 is -NH-, -S-, or -0-. In some embodiments, W 1 is -CH 2 O-, -CH 2 S-, or -CH 2 NH-. In some aspects, W 1 is -OCH 2 -, -SCH 2 -, -NHCH 2 -, or -CH 2 CH 2 -.
  • W 2 is a covalent bond.
  • W 2 is a bivalent Ci_ 3 alkylene chain wherein one methylene unit of W 2 is optionally replaced by -NR 2 -, -N(R 2 )C(0)-, -C(O)N(R 2 )-, -N(R 2 )SO 2 -, -SO 2 N(R 2 )-, -0-, -C(O)-, -OC(O)-, -C(O)O-, -S-, -SO- or -SO 2 -.
  • W 2 is -NR 2 -. In other embodiments, W 2 is -0-. In certain embodiments, W 2 is -NH-, -S-, or -0-. In some embodiments, W 2 is -CH 2 O-, -CH 2 S-, or -CH 2 NH-. In some aspects, W 2 is -OCH 2 -, -SCH 2 -, -NHCH 2 -, or -CH 2 CH 2 -.
  • Ring B is phenyl, thus forming a compound of formula II-a or II-b:
  • Ring A, m, p, R x , R y , R V ,W ⁇ W 2 , and R 1 is as defined above and described in classes and subclasses above and herein.
  • Ring A is phenyl, thus forming a compound of formula III-a or III-b:
  • Ring A is phenyl and Ring B is phenyl, thus forming a compound of formula IV-a or IV-b:
  • each R 2 is independently hydrogen, optionally substituted Ci_6 aliphatic, or -C(O)R, or R 2 and a substituent on Ring A are taken together with their intervening atoms to form a 4-6 membered partially unsaturated or aromatic fused ring, or R 2 and R y are taken together with their intervening atoms to form a 4-6 membered saturated, partially unsaturated, or aromatic fused ring.
  • R 2 is hydrogen.
  • R 2 is -C(O)R, wherein R is an optionally substituted Ci_6 aliphatic group.
  • R 2 and a substituent on Ring A are taken together with their intervening atoms to form a 4-7 membered saturated or partially unsaturated ring, thus forming a compound of formula I-a-/ or I-b-/:
  • R 2 and R y are taken together with their intervening atoms to form a 4-7 partially unsaturated ring, thus forming a compound of formula I-a-// or I-b-//:
  • Y is hydrogen, Ci_6 aliphatic optionally substituted with oxo, halogen, NO 2 , or CN, or a 3-10 membered monocyclic or bicyclic, saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, and wherein said ring is substituted with 1-4 R e groups; and each R e is independently selected from -Q-Z, oxo, NO 2 , halogen, CN, a suitable leaving group, or a Ci_6 aliphatic optionally substituted with oxo, halogen, NO 2 , or CN, wherein: Q is a covalent bond or a bivalent Ci_6 saturated or unsaturated, straight or branched, hydrocarbon chain, wherein one or two methylene units of Q are optionally and independently replaced by -N(R)-, -S-, -0-, -C(O)-, -OC(O)-, -C
  • L is a bivalent Ci_g saturated or unsaturated, straight or branched, hydrocarbon chain. In certain embodiments, L is -CH 2 -.
  • L is a covalent bond, -CH 2 -, -NH-, -CH 2 NH-, -NHCH 2 -, -NHC(O)-, -NHC(O)CH 2 OC(O)-, -CH 2 NHC(O)-, -NHSO 2 -, -NHSO 2 CH 2 -,
  • L is a bivalent C 2 _ 8 straight or branched, hydrocarbon chain wherein L has at least one double bond and one or two additional methylene units of L are optionally and independently replaced by -NRC(O)-, -C(O)NR-, -N(R)SO 2 -, -SO 2 N(R)-, -S-, -S(O)-, -SO 2 -, -OC(O)-, -C(O)O-, cyclopropylene, -0-, -N(R)-, or -C(O)-.
  • L is a bivalent C 2 _8 straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by -C(O)-, -NRC(O)-, -C(O)NR-, -N(R)SO 2 -, -SO 2 N(R)-, -S-, -S(O)-, -SO 2 -, -OC(O)-, or -C(O)O-, and one or two additional methylene units of L are optionally and independently replaced by cyclopropylene, -O-, -N(R)-, or -C(O)-.
  • L is a bivalent C2-8 straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by
  • -C(O)- and one or two additional methylene units of L are optionally and independently replaced by cyclopropylene, -O-, -N(R)-, or -C(O)-.
  • L is a bivalent C2-8 straight or branched, hydrocarbon chain wherein L has at least one double bond.
  • L has at least one double bond.
  • One of ordinary skill in the art will recognize that such a double bond may exist within the hydrocarbon chain backbone or may be
  • L is a bivalent C2-8 straight or branched, hydrocarbon chain wherein L has at least one alkylidenyl double bond.
  • L is a bivalent C 2 - 8 straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by
  • L is a bivalent C 2 _8 straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by
  • L is a bivalent C 2 _s straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by
  • each R is independently hydrogen or optionally substituted Ci_6 aliphatic.
  • L is a bivalent C 2 _8 straight or branched, hydrocarbon chain wherein L has at least one triple bond.
  • L is a bivalent C 2 _g straight or branched, hydrocarbon chain wherein L has at least one triple bond and one or two additional methylene units of L are optionally and independently replaced by -NRC(O)-, -C(O)NR-, -S-,
  • L has at least one triple bond and at least one methylene unit of L is replaced by -N(R)-, -N(R)C(O)-, -C(O)-,
  • L is a bivalent C 2 _g straight or branched, hydrocarbon chain wherein one methylene unit of L is replaced by cyclopropylene and one or two additional methylene units of L are independently replaced by -C(O)-, -NRC(O)-, -C(O)NR-, -N(R)SO 2 -, or -SO 2 N(R)-.
  • Exemplary L groups include -NHC(O)-cyclopropylene-SO 2 - and -NHC(O)- cyclopropylene-.
  • Y is hydrogen, Ci_ 6 aliphatic optionally substituted with oxo, halogen, NO 2 , or CN, or a 3-10 membered monocyclic or bicyclic, saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, and wherein said ring is substituted with at 1-4 R e groups, each R e is independently selected from -Q-Z, oxo, NO 2 , halogen, CN, a suitable leaving group, or Ci_ 6 aliphatic, wherein
  • Q is a covalent bond or a bivalent Ci_6 saturated or unsaturated, straight or branched, hydrocarbon chain, wherein one or two methylene units of Q are optionally and independently replaced by -N(R)-, -S-, -0-, -C(O)-, -OC(O)-, -C(O)O-, -SO-, or -SO 2 -, -N(R)C(O)-, - C(O)N(R)-, -N(R)SO 2 -, or -SO 2 N(R)-; and, Z is hydrogen or Ci_ 6 aliphatic optionally substituted with oxo, halogen, NO 2 , or CN.
  • Y is hydrogen.
  • Y is Ci_6 aliphatic optionally substituted with oxo, halogen, NO 2 , or CN.
  • Y is C 2 _6 alkenyl optionally substituted with oxo, halogen, NO 2 , or CN.
  • Y is C 2 _ 6 alkynyl optionally substituted with oxo, halogen, NO 2 , or CN.
  • Y is C 2 _6 alkenyl.
  • Y is C 2 _4 alkynyl.
  • Y is Ci_6 alkyl substituted with oxo, halogen, NO 2 , or CN.
  • Y groups include -CH 2 F, -CH 2 Cl, -CH 2 CN, and -CH 2 NO 2 .
  • Y is a saturated 3-6 membered monocyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein Y is substituted with 1 -4 R e groups, wherein each R e is as defined above and described herein.
  • Y is a saturated 3-4 membered heterocyclic ring having 1 heteroatom selected from oxygen or nitrogen wherein said ring is substituted with 1-2 R e groups, wherein each R e is as defined above and described herein.
  • Exemplary such rings are epoxide and oxetane rings, wherein each ring is substituted with 1-2 R e groups, wherein each R e is as defined above and described herein.
  • Y is a saturated 5-6 membered heterocyclic ring having 1-2 heteroatom selected from oxygen or nitrogen wherein said ring is substituted with 1-4 R e groups, wherein each R e is as defined above and described herein.
  • Such rings include piperidine and pyrrolidine, wherein each ring is substituted with 1-4 R e groups, wherein each R e is as defined
  • Y is "2 , or
  • Y is a saturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 R e groups, wherein each R e is as defined above and described herein.
  • Y is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl, wherein each ring is substituted with 1-4 R e groups, wherein each R e is as defined above and described herein..
  • Y is , wherein R e is as defined above and described herein.
  • Y is cyclopropyl optionally substituted with halogen, CN or NO 2 .
  • Y is a partially unsaturated 3-6 membered monocyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 R e groups, wherein each R e is as defined above and described herein.
  • Y is a partially unsaturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 R e groups, wherein each R e is as defined above and described herein.
  • Y is cyclopropenyl, cyclobutenyl, cyclopentenyl, or cyclohexenyl wherein each ring is substituted with 1-4 R e groups, wherein each R e is as defined
  • Y is ** 1"2 , wherein each R e is as defined above and described herein.
  • Y is a partially unsaturated 4-6 membered heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 R e groups, wherein each R e is as defined above and described herein.
  • Y is selected from:
  • each R and R e is as defined above and described herein.
  • Y is a 6-membered aromatic ring having 0-2 nitrogens wherein said ring is substituted with 1-4 R e groups, wherein each R e group is as defined above and described herein.
  • Y is phenyl, pyridyl, or pyrimidinyl, wherein each ring is substituted with 1-4 R e groups, wherein each R e is as defined above and described herein.
  • Y is selected from:
  • each R e is as defined above and described herein.
  • Y is a 5-membered heteroaryl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-3 R e groups, wherein each R e group is as defined above and described herein.
  • Y is a 5 membered partially unsaturated or aryl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, wherein said ring is substituted with 1- 4 R e groups, wherein each R e group is as defined above and described herein.
  • rings are isoxazolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, pyrrolyl, furanyl, thienyl, triazole, thiadiazole, and oxadiazole, wherein each ring is substituted with 1-3 R e groups, wherein each R e group is as defined above and described herein.
  • Y is selected from:
  • each R and R e is as defined above and described herein.
  • Y is an 8-10 membered bicyclic, saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 R e groups, wherein R e is as defined above and described herein.
  • Y is a 9-10 membered bicyclic, partially unsaturated, or aryl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 R e groups, wherein R e is as defined above and described herein.
  • each R e group is independently selected from -Q-Z, oxo, NO 2 , halogen, CN, a suitable leaving group, or Ci_ 6 aliphatic optionally substituted with oxo, halogen, NO 2 , or CN, wherein Q is a covalent bond or a bivalent Ci_6 saturated or unsaturated, straight or branched, hydrocarbon chain, wherein one or two methylene units of Q are optionally and independently replaced by -N(R)-, -S-, -O-, -C(O)-, -OC(O)-, -C(O)O-, -SO-, or -SO 2 -, - N(R)C(O)-, -C(
  • R e is Ci_6 aliphatic optionally substituted with oxo, halogen, NO 2 , or CN. In other embodiments, R e is oxo, NO 2 , halogen, or CN.
  • R e is -Q-Z, wherein Q is a covalent bond and Z is hydrogen (i.e., R e is hydrogen).
  • R e is -Q-Z, wherein Q is a bivalent Ci_6 saturated or unsaturated, straight or branched, hydrocarbon chain, wherein one or two methylene units of Q are optionally and independently replaced by -NR-, -NRC(O)-, -C(O)NR-, -S-, -O-, -C(O)-, -SO-, or -SO 2 -.
  • Q is a bivalent C 2 _ 6 straight or branched, hydrocarbon chain having at least one double bond, wherein one or two methylene units of Q are optionally and independently replaced by -NR-, -NRC(O)-, -C(O)NR-, -S-, -0-, -C(O)-, -SO-, or -SO 2 -.
  • the Z moiety of the R e group is hydrogen.
  • R e is a suitable leaving group, ie a group that is subject to nucleophilic displacement.
  • a “suitable leaving” is a chemical group that is readily displaced by a desired incoming chemical moiety such as the thiol moiety of a cysteine of interest.
  • Suitable leaving groups are well known in the art, e.g., see, “Advanced Organic Chemistry,” Jerry March, 5 th Ed., pp. 351-357, John Wiley and Sons, N.Y.
  • Such leaving groups include, but are not limited to, halogen, alkoxy, sulphonyloxy, optionally substituted alkylsulphonyloxy, optionally substituted alkenylsulfonyloxy, optionally substituted arylsulfonyloxy, acyl, and diazonium moieties.
  • Suitable leaving groups include chloro, iodo, bromo, fluoro, acetoxy, methanesulfonyloxy (mesyloxy), tosyloxy, triflyloxy, nitro-phenylsulfonyloxy (nosyloxy), and bromo-phenylsulfonyloxy (brosyloxy).
  • L is a bivalent C 2 - 8 straight or branched, hydrocarbon chain wherein L has at least one double bond and one or two additional methylene units of L are optionally and independently replaced by -NRC(O)-, -C(O)NR-, -N(R)SO 2 -, -SO 2 N(R)-, -S-, -S(O)-, -SO 2 -, -OC(O)-, -C(O)O-, cyclopropylene, -0-, -N(R)-, or -C(O)- ; and Y is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO 2 , or CN; or
  • L is a bivalent C 2 -8 straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by -C(O)-, -NRC(O)-, -C(O)NR-, -N(R)SO 2 -, -SO 2 N(R)-, -S-, -S(O)-, -SO 2 -, -OC(O)-, or -C(O)O-, and one or two additional methylene units of L are optionally and independently replaced by cyclopropylene, -0-, -N(R)-, or -C(O)-; and Y is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO 2 , or CN; or
  • L is a bivalent C 2 _ 8 straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by -C(O)-, and one or two additional methylene units of L are optionally and independently replaced by cyclopropylene, -0-, -N(R)-, or -C(O)-; and Y is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO 2 , or CN; or
  • L is a bivalent C 2 _ 8 straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by -C(O)-; and Y is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO 2 , or CN; or
  • L is a bivalent C 2 -8 straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by -OC(O)-; and Y is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO 2 , or CN; or
  • Y is hydrogen or Ci_ 6 aliphatic optionally substituted with oxo, halogen, NO 2 , or CN; or
  • L is a bivalent C 2 _ 8 straight or branched, hydrocarbon chain wherein L has at least one alkylidenyl double bond and at least one methylene unit of L is replaced by -C(O)-, -NRC(O)-, -C(O)NR-, -N(R)SO 2 -, -SO 2 N(R)-, -S-, -S(O)-, -SO 2 -, -OC(O)-, or -C(O)O-, and one or two additional methylene units of L are optionally and independently replaced by cyclopropylene, -0-, -N(R)-, or -C(O)-; and Y is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO 2 , or CN; or
  • L is a bivalent C 2 _ 8 straight or branched, hydrocarbon chain wherein L has at least one triple bond and one or two additional methylene units of L are optionally and independently replaced by -NRC(O)-, -C(O)NR-, -N(R)SO 2 -, -SO 2 N(R)-, -S-, -S(O)-, -SO 2 -, -OC(O)-, or -C(O)O-, and Y is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO 2 , or CN; or
  • (k) L is a bivalent C 2 _ 8 straight or branched, hydrocarbon chain wherein one methylene unit of L is replaced by cyclopropylene and one or two additional methylene units of L are independently replaced by -NRC(O)-, -C(O)NR-, -N(R)SO 2 -, -SO 2 N(R)-, -S-, -S(O)-, -SO 2 -, -OC(O)-, or -C(O)O-; and Y is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO 2 , or CN; or
  • L is a covalent bond and Y is selected from:
  • R e is as defined above and described herein; or (v) a saturated 5-6 membered heterocyclic ring having 1-2 heteroatom selected from oxygen or nitrogen wherein said ring is substituted with 1-4 R e groups, wherein each
  • R e is as defined above and described herein; or
  • R e groups wherein each R e is as defined above and described herein; or ⁇ vii ⁇ ) a partially unsaturated 3-6 membered monocyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 R e groups, wherein each R e is as defined above and described herein; or (Jx) a partially unsaturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 R e groups, wherein each R e is as defined above and described herein; or
  • each R and R e is as defined above and described herein; or (xiii) a 6-membered aromatic ring having 0-2 nitrogens wherein said ring is substituted with 1-4 R e groups, wherein each R e group is as defined above and described herein; or N N
  • each R and R e is as defined above and described herein; or (pcvii) an 8-10 membered bicyclic, saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 R e groups, wherein R e is as defined above and described herein; (m) L is -C(O)- and Y is selected from:
  • each R, Q, Z, and R e is as defined above and described herein; or (vii) a saturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4
  • R e groups wherein each R e is as defined above and described herein; or (viii) a partially unsaturated 3-6 membered monocyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 R e groups, wherein each R e is as defined above and described herein; or (ix) a partially unsaturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 R e groups, wherein each R e is as defined above and described herein; or
  • (xz) a partially unsaturated 4-6 membered heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 R e groups, wherein each R e is as defined above and described herein; or
  • each R and R e is as defined above and described herein; or (pcvii) an 8-10 membered bicyclic, saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 R e groups, wherein R e is as defined above and described herein; (n) L is -N(R)C(O)- and Y is selected from:
  • R e is as defined above and described herein; or (v) a saturated 5-6 membered heterocyclic ring having 1-2 heteroatom selected from oxygen or nitrogen wherein said ring is substituted with 1-4 R e groups, wherein each
  • R e is as defined above and described herein; or (vi) wherein each R, Q, Z, and R e is as defined above and described herein; or (vii) a saturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4
  • R e groups wherein each R e is as defined above and described herein; or (viii) a partially unsaturated 3-6 membered monocyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 R e groups, wherein each R e is as defined above and described herein; or (ix) a partially unsaturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 R e groups, wherein each R e is as defined above and described herein; or
  • each R e is as defined above and described herein; or (xv) a 5-membered heteroaryl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-3 R e groups, wherein each R e group is as defined above and described herein; or
  • each R and R e is as defined above and described herein; or (xvii) an 8-10 membered bicyclic, saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 R e groups, wherein R e is as defined above and described herein;
  • L is a bivalent Ci_g saturated or unsaturated, straight or branched, hydrocarbon chain; and Y is selected from:
  • Ci_6 alkyl substituted with oxo, halogen, NO 2 , or CN (U) C 2 -6 alkenyl optionally substituted with oxo, halogen, NO 2 , or CN; or (in) C 2 - 6 alkynyl optionally substituted with oxo, halogen, NO 2 , or CN; or (Jv) a saturated 3-4 membered heterocyclic ring having 1 heteroatom selected from oxygen or nitrogen wherein said ring is substituted with 1-2 R e groups, wherein each
  • R e is as defined above and described herein; or (v) a saturated 5-6 membered heterocyclic ring having 1-2 heteroatom selected from oxygen or nitrogen wherein said ring is substituted with 1-4 R e groups, wherein each
  • R e is as defined above and described herein; or (vi) wherein each R, Q, Z, and R e is as defined above and described herein; or (vii) a saturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4
  • R e groups wherein each R e is as defined above and described herein; or (viii) a partially unsaturated 3-6 membered monocyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 R e groups, wherein each R e is as defined above and described herein; or (ix) a partially unsaturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 R e groups, wherein each R e is as defined above and described herein; or
  • each R e is as defined above and described herein; or (xv) a 5-membered heteroaryl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-3 R e groups, wherein each R e group is as defined above and described herein; or
  • each R and R e is as defined above and described herein; or (xvii) an 8-10 membered bicyclic, saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 R e groups, wherein R e is as defined above and described herein;
  • (p) L is a covalent bond, -CH 2 -, -NH-, -C(O)-, -CH 2 NH-, -NHCH 2 -, -NHC(O)-, -NHC(O)CH 2 OC(O)-, -CH 2 NHC(O)-, -NHSO 2 -, -NHSO 2 CH 2 -, -NHC(O)CH 2 OC(O)-, or -SO 2 NH-; and Y is selected from:
  • Ci_6 alkyl substituted with oxo, halogen, NO 2 , or CN (i) Ci_6 alkyl substituted with oxo, halogen, NO 2 , or CN; or (U) C 2 _ 6 alkenyl optionally substituted with oxo, halogen, NO 2 , or CN; or (Hi) C 2 _6 alkynyl optionally substituted with oxo, halogen, NO 2 , or CN; or (Jv) a saturated 3-4 membered heterocyclic ring having 1 heteroatom selected from oxygen or nitrogen wherein said ring is substituted with 1-2 R e groups, wherein each
  • R e is as defined above and described herein; or (v) a saturated 5-6 membered heterocyclic ring having 1-2 heteroatom selected from oxygen or nitrogen wherein said ring is substituted with 1-4 R e groups, wherein each
  • R e is as defined above and described herein; or (vi) wherein each R, Q, Z, and R e is as defined above and described herein; or (vii) a saturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4
  • R e groups wherein each R e is as defined above and described herein; or (viii) a partially unsaturated 3-6 membered monocyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 R e groups, wherein each R e is as defined above and described herein; or (ix) a partially unsaturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 R e groups, wherein each R e is as defined above and described herein; or
  • each R e is as defined above and described herein; or (xv) a 5-membered heteroaryl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-3 R e groups, wherein each R e group is as defined above and described herein; or
  • each R and R e is as defined above and described herein; or (pcvii) an 8-10 membered bicyclic, saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 R e groups, wherein R e is as defined above and described herein.
  • the Y group of formula Ia or Ib is selected from those set forth in Table 3, below, wherein each wavy line indicates the point of attachment to the rest of the molecule.
  • R 1 is selected from those set forth in Table 4, below, wherein each wavy line indicates the point of attachment to the rest of the molecule.
  • R 1 is a warhead group, or, when R 1 and R x form a ring, then -Q-Z is a warhead group.
  • R 1 groups i.e. warhead groups, are particularly suitable for covalently binding to a key cysteine residue in the binding domain of certain protein kinases.
  • Protein kinases having a cysteine residue in the binding domain are known to one of ordinary skill in the art and include ErbBl, ErbB2, and ErbB4, or a mutant thereof.
  • compounds of the present invention have a warhead group characterized in that inventive compounds target one or more of the following cysteine residues:
  • R 1 is characterized in that the -L-Y moiety is capable of covalently binding to a cysteine residue thereby irreversibly inhibiting the enzyme.
  • the cysteine residue is Cys797 of ErbBl, Cys805 of ErbB2 and Cys803 of ErbB4, or a mutant thereof, where the provided residue numbering is in accordance with Uniprot (code POO533 for ErbBl; code PO4626 for ErbB2, and Q 15303 for ErbB4).
  • the Cys of ErbBl (EGFR) is variably called 773 or 797 depending on whether the parent sequence contains the signal peptide or not.
  • the relevant cysteine residue of ErbBl may be described as Cys 773 or Cys 797 and these terms are used interchangeably.
  • R 1 groups include, but are not limited to, those described herein and depicted in Table 3, infra.
  • the R 1 warhead group can be in an ortho-, meta-, or para-position. In certain embodiments, the R 1 warhead group is in a meta-position of the phenyl ring relative to the rest of the molecule. [00114] In certain embodiments, R 1 is characterized in that the -L-Y moiety is capable of covalently binding to a cysteine residue of TEC, thereby irreversibly inhibiting the enzyme. In some embodiments, the cysteine residue is Cys 449.
  • R 1 is characterized in that the -L-Y moiety is capable of covalently binding to a cysteine residue of BTK, thereby irreversibly inhibiting the enzyme.
  • the cysteine residue is Cys 481.
  • R 1 is characterized in that the -L-Y moiety is capable of covalently binding to a cysteine residue of ITK, thereby irreversibly inhibiting the enzyme.
  • the cysteine residue is Cys 442.
  • R 1 is characterized in that the -L-Y moiety is capable of covalently binding to a cysteine residue of BMX, thereby irreversibly inhibiting the enzyme.
  • the cysteine residue is Cys 496.
  • R 1 is characterized in that the -L-Y moiety is capable of covalently binding to a cysteine residue of JAK3, thereby irreversibly inhibiting the enzyme.
  • the cysteine residue is Cys 909.
  • R 1 is characterized in that the -L-Y moiety is capable of covalently binding to a cysteine residue of TXK, thereby irreversibly inhibiting the enzyme.
  • the cysteine residue is Cys 350.
  • warhead groups as defined herein, are suitable for such covalent bonding.
  • R 1 groups include, but are not limited to, those described herein and depicted in Table 3, infra.
  • the present invention provides any compound depicted in
  • the present invention provides a compound selected from:
  • compounds of the present invention are irreversible inhibitors of at least one of ErbBl, ErbB2, ErbB3 and ErbB4, or a mutant thereof.
  • provided compounds are irreversible inhibitors of a TEC-kinase (e.g. BTK) and JAK3.
  • BTK TEC-kinase
  • JAK3 JAK3
  • such reversible compounds are useful as inhibitors of ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3, or a mutant thereof, and therefore useful for treating one or disorders as described herein.
  • An exemplary reversible compound of the present invention has the following structure.
  • 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 compositions of this invention is such that is effective to measurably inhibit a protein kinase, particularly at least one of ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3, or a mutant thereof, in a biological sample or in a patient.
  • the amount of compound in compositions of this invention is such that is effective to measurably inhibit at least one of ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3, or a mutant thereof, in a biological sample or in a patient.
  • a composition of this invention is formulated for administration to a patient in need of such composition.
  • a composition of this invention is formulated for oral administration to a patient.
  • patient means an animal, preferably a mammal, and most preferably a human.
  • compositions of this invention refers to a nontoxic carrier, adjuvant, or vehicle that does not destroy the pharmacological activity of the compound with which it is formulated.
  • Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the compositions of this invention 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, 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, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropy
  • a "pharmaceutically acceptable derivative” means any non-toxic salt, ester, salt of an ester or other derivative of a compound of this invention that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or an inhibitorily active metabolite or residue thereof.
  • the term "inhibitorily active metabolite or residue thereof means that a metabolite or residue thereof is also an inhibitor of at least one of ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3, or a mutant thereof.
  • 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, intraarticular, intra-synovial, intrasternal, intrathecal, 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. 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 nontoxic parenterally acceptable diluent or solvent, for example as a solution in 1,3-butanediol.
  • a nontoxic 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 di-glycerides.
  • 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.
  • 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.
  • 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.
  • compositions of this invention may be administered in the form of suppositories for rectal administration.
  • suppositories for rectal administration.
  • suppositories 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.
  • suitable non-irritating excipient include cocoa butter, beeswax and polyethylene glycols.
  • 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.
  • 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 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.
  • provided 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.
  • compositions may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or, preferably, as solutions in isotonic, pH adjusted sterile saline, either with or without a preservative such as benzylalkonium chloride.
  • the pharmaceutically acceptable compositions may be formulated in an ointment such as petrolatum.
  • 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, fluorocarbons, and/or other conventional solubilizing or dispersing agents. [00139] Most preferably, pharmaceutically acceptable compositions of this invention are formulated for oral administration. [00140] The amount of 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, provided 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.
  • a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated.
  • the amount of a compound of the present invention in the composition will also depend upon the particular compound in the composition.
  • Compounds and compositions described herein are generally useful for the inhibition of protein kinase activity of one or more enzymes.
  • Drug resistance is emerging as a significant challenge for targeted therapies.
  • drug resistance has been reported for Gleevec ® and Iressa ® , as well as several other kinase inhibitors in development.
  • drug resistance has been reported for the cKit and
  • PDGFR receptors It has been reported that irreversible inhibitors may be effective against drug resistant forms of protein kinases (Kwak, E. L., R. Sordella, et al. (2005). "Irreversible inhibitors of the EGF receptor may circumvent acquired resistance to gefitinib.” PNAS 102(21): 7665-
  • compounds of the present invention may be effective inhibitors of drug resistant forms of protein kinases.
  • clinical drug resistance refers to the loss of susceptibility of a drug target to drug treatment as a consequence of mutations in the drug target.
  • the term “resistance” refers to changes in the wild-type nucleic acid sequence coding a target protein, and/or the protein sequence of the target, which changes decrease or abolish the inhibitory effect of the inhibitor on the target protein.
  • kinases that are inhibited by the compounds and compositions described herein and against which the methods described herein are useful include ErbBl, ErbB2, ErbB3,
  • ErbB4 a TEC-kinase (including BTK, ITK, TEC, BMX and RLK), and/or JAK3, or a mutant thereof.
  • the activity of a compound utilized in this invention as an inhibitor of ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3, or a mutant thereof, may be assayed in vitro, in vivo or in a cell line.
  • In vitro assays include assays that determine inhibition of either the phosphorylation activity and/or the subsequent functional consequences, or ATPase activity of activated ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3, or a mutant thereof. Alternate in vitro assays quantitate the ability of the inhibitor to bind to ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3.
  • Inhibitor binding may be measured by radio labeling the inhibitor prior to binding, isolating the inhibitor/ErbBl, inhibitor/ErbB2, inhibitor/ErbB3, inhibitor/ErbB4, inhibitor/TEC-kinase (i.e., TEC, BTK, ITK, RLK and BMX), or inhibitor/JAK3 complex and determining the amount of radiolabel bound.
  • inhibitor binding may be determined by running a competition experiment where new inhibitors are incubated with ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3 bound to known radioligands.
  • Protein tyrosine kinases are a class of enzymes that catalyze the transfer of a phosphate group from ATP or GTP to a tyrosine residue located on a protein substrate. Receptor tyrosine kinases act to transmit signals from the outside of a cell to the inside by activating secondary messaging effectors via a phosphorylation event. A variety of cellular processes are promoted by these signals, including proliferation, carbohydrate utilization, protein synthesis, angiogenesis, cell growth, and cell survival. (a) ErbB Family
  • ErbB receptors a major family of receptor tyrosine kinases, are composed of an extracellular ligand binding domain, a single transmembrane domain, and an intracellular domain with tyrosine kinase activity.
  • the ErbB family comprises ErbBl (commonly known as EGFR), ErbB2 (commonly known as HER2 or neu), ErbB3 (commonly known as HER3), and ErbB4 (commonly known as HER4). More than 10 ligands (including EGF, TGF ⁇ , AR, BTC, EPR, HB-EGF, NRG-I, NRG-2, NRG-3, NRG-4) have been identified for the various receptor family members.
  • PBK phosphatidyl-inositol 3-kinase
  • mitogen-activated protein kinase pathways occur, leading to cell proliferation and survival (Lin, N. U.; Winer, E. P., Breast Cancer Res 6: 204-210, 2004).
  • Representative downstream gene products in the ErbB signal transduction pathway include She, Grb2, SOSl, Ras, Rafl, Mek, ERKl, ERK2, ERa, Akt, mTOR, FKHR, p27, Cyclin Dl, FasL, GSK-3, Bad, and STAT3.
  • ErbBl and/or ErbB2 amplification has also been implicated in squamous cell carcinomas, salivary gland carcinomas, ovarian carcinomas, and pancreatic cancers (Cooper, G. C. Oncogenes. 2 nd ed. Sudbury: Jones and Barlett, 1995; Zhang, Y., et al, Cancer Res 66 : 1025-32, 2006).
  • Overexpression of ErbB2 has potent transforming activity, likely due to its ability to cooperate with other ErbB receptors (Sherman, L., et al., Oncogene J_8: 6692-99, 1999).
  • ErbB signaling network is often a key component in the pathogenesis of breast cancer. Amplification of ErbB2 is associated with an aggressive tumor phenotype that is characterized by relatively rapid tumor growth, metastatic spread to visceral sites, and drug resistance. ErbB2 has been shown to be amplified in 20% of axillary node-negative ("ANN") breast cancer cases, and this amplification has been identified as an independent prognostic factor for risk of recurrence in ANN breast cancer. (Andrulis, LL. , et al., J Clin Oncol Jj5: 1340- 9, 1998).
  • ANN axillary node-negative
  • Targeted blockade of ErbB signaling with trastuzumab (Herceptin), a monoclonal antibody directed at ErbB2, has been shown to improve survival in women with ErbB2-positive, advanced breast cancer.
  • Other monoclonal antibodies directed against ErbB receptors include cetuximab (Erbitux) and panitumumab (Vectibix).
  • TKIs small molecule tyrosine kinase inhibitors
  • Iressa gefitinib
  • Tarceva erlotinib
  • TKIs have several advantages in that they are orally bioavailable, well-tolerated, and appear to be active against truncated forms of ErbB2 and EGFR receptors (e.g., EGFR vIII) in vitro.
  • TKIs small molecule TKIs may allow them to penetrate sanctuary sites such as the central nervous system.
  • the homology between kinase domains of ErbB receptors allows for development of TKIs that target more than one member of the ErbB family simultaneously, the advantages of which are described herein.
  • pan-ErbB regulators Agents that target two or more ErbB receptors are called pan-ErbB regulators.
  • ERRP is a pan-ErbB negative regulator that is expressed in most benign pancreatic ductal epithelium and islet cells. Tumors have been found to experience a progressive loss in ERRP expression. That Erbitux and Herceptin show success in a limited patient base (tumors having increased expression of EGFR or ErbB2) could be partly due to coexpression of multiple ErbB family members.
  • strategies that employ a dual ErbB approach seem to have greater antitumor activity than agents targeting a single ErbB receptor.
  • provided compounds inhibit one or more of ErbBl, ErbB2, ErbB3, and ErbB4. In some embodiments, provided compounds inhibit two or more of ErbBl, ErbB2, ErbB3, and ErbB4, or a mutant thereof, and are therefore pan-ErbB inhibitors.
  • pan-ErbB ErbB
  • Possible pan-ErbB approaches with small molecules include using combinations of agents that target individual ErbB receptors, using single agents that target multiple ErbB receptors, or using agents that interfere with ErbB receptor interactions (e.g., dimerization). Additional strategies include therapies utilizing a small molecule in combination with antibodies, or chemoprevention therapies (Lin, N. U.; Winer, E. P., Breast Cancer Res 6: 204-210, 2004).
  • pan-ErbB inhibition is CI- 1033, an irreversible pan- ErbB inhibitor that covalently binds to the ATP binding site of the intracellular kinase domain.
  • Another irreversible pan-ErbB receptor tyrosine kinase inhibitor is HKI-272, which inhibits the growth of tumor cells that express ErbB-1 (EGFR) and ErbB-2 (HER-2) in culture and xenografts, and has antitumor activity in HER-2-positive breast cancer (Andrulis, LL. , et al., J Clin Oncol 16: 1340-9, 1998). Irreversible inhibitors have demonstrated superior antitumor activity in comparison with reversible inhibitors.
  • Neurofibromatosis type I is a dominantly inherited human disease affecting one in 2500-3500 individuals.
  • organ systems are affected, including bones, skin, iris, and the central nervous system, as manifested in learning disabilities and gliomas.
  • a hallmark of NFl is the development of benign tumors of the peripheral nervous system (neurofibromas), which vary greatly in both number and size among patients.
  • Neurofibromas are heterogeneous tumors composed of Schwann cells, neurons, fibroblasts and other cells, w/ Schwann cells being the major (60-80%) cell type.
  • EGFR expression affects the growth of tumor cell lines derived from NFl patients under conditions where EGF is not the primary factor driving growth of the cells. These data suggest that EGFR may play an important role in NFl tumorigenesis and Schwann cell transformation (DeClue, J. E., et al, J Clin Invest 105: 1233-41, 2000).
  • Schwannomas are peripheral nerve tumors comprised almost entirely of Schwann- like cells, and typically have mutations in the neurofibromatosis type II (NF2) tumor suppressor gene.
  • NF2 neurofibromatosis type II
  • schwannoma cells Both normal human Schwann cells and schwannoma cells express neuregulin receptors (i.e., ErbB receptors), and schwannoma cells proliferate in response to neuregulin. It is possible that aberrant neuregulin production or response contributes to aberrant schwannoma cell proliferation (Pelton, P. D., et al., Oncogene 17: 2195-2209, 1998).
  • neuregulin receptors i.e., ErbB receptors
  • the NF2 tumor suppressor, Merlin is a membrane/cytoskeleton-associated protein implicated in the regulation of tyrosine kinase activity. Genetic interactions between a Merlin mutation and EGFR pathway mutations have been documented in Drosophila (LaJeunesse, D. R., et al, Genetics 158: 667-79, 2001). Other evidence suggests Merlin can inhibit EGFR internalization and signaling upon cell-cell contact by restraining the EGFR into a membrane compartment from which it can neither signal nor be internalized (McClatchey, A. L, et al., Genes and Development 19: 2265-77, 2005; Curto, M.
  • treatment refers to reversing, alleviating, delaying the onset of, or inhibiting the progress of a disease or disorder, or one or more symptoms thereof, as described herein.
  • treatment may be administered after one or more symptoms have developed.
  • treatment may be administered in the absence of symptoms.
  • treatment may be administered to a susceptible individual prior to the onset of symptoms (e.g., in light of a history of symptoms and/or in light of genetic or other susceptibility factors). Treatment may also be continued after symptoms have resolved, for example to prevent or delay their recurrence.
  • the present invention provides a method for treating an ErbBl -mediated, an ErbB2-mediated, an ErbB3- mediated, and/or ErbB4-mediated disorder comprising the step of administering to a patient in need thereof a compound of the present invention, or pharmaceutically acceptable composition thereof.
  • ErbBl -mediated means any disease or other deleterious condition in which one or more of ErbBl, ErbB2, ErbB3, and/or ErbB4, or a mutant thereof, are known to play a role.
  • another embodiment of the present invention relates to treating or lessening the severity of one or more diseases in which one or more of ErbBl, ErbB2, ErbB3, and/or ErbB4, or a mutant thereof, are known to play a role.
  • the present invention relates to a method of treating or lessening the severity of a disease or condition selected from a proliferative disorder, wherein said method comprises administering to a patient in need thereof a compound or composition according to the present invention.
  • the present invention provides a method for treating or lessening the severity of one or more disorders selected from a cancer.
  • the cancer is associated with a solid tumor.
  • the cancer is breast cancer, glioblastoma, lung cancer, cancer of the head and neck, colorectal cancer, bladder cancer, or non-small cell lung cancer.
  • the present invention provides a method for treating or lessening the severity of one or more disorders selected from squamous cell carcinoma, salivary gland carcinoma, ovarian carcinoma, or pancreatic cancer. [00171] In certain embodiments, the present invention provides a method for treating or lessening the severity of neurofibromatosis type I (NFl), neurofibromatosis type II (NF2) Schwann cell neoplasms (e.g. MPNST's), or Schwannomas. (b) TEC Family
  • TEC- kinases The TEC family of non-receptor tyrosine kinases, referred to herein as "TEC- kinases,” plays a central role in signaling through antigen-receptors such as the TCR, BCR and Fc receptors (reviewed in Miller A, et al. Current Opinion in Immunology 14; 331-340 (2002). TEC-kinases are essential for T cell activation. Three members of the family, Itk, RIk and, are activated downstream of antigen receptor engagement in T cells and transmit signals to downstream effectors, including PLC- ⁇ .
  • mice Combined deletion of Itk and RIk in mice leads to a profound inhibition of TCR responses including proliferation, cytokine production and immune responses to an intracellular parasite (Toxoplasma gondii) (Schaeffer et al., Science 284; 638- 641 (1999)).
  • Intracellular signalling following TCR engagement is effected in ITK/RLK deficient T cells; inositol triphosphate production, calcium mobilization and MAP kinase activation are all reduced.
  • Tec-kinases are also essential for B cell development and activation.
  • TEC-kinases include five family members, which are expressed primarily in hematopoietic cells: TEC, BTK, ITK (also known as TSK and EMT), RLK (also known as TXK), and BMX (also known as ETK). Additional related TEC-kinases have been found in Drosophila melanogaster, zebraf ⁇ sh (Danio reri ⁇ ), skate (Raja eglanteria), and sea urchin (Anthocidaris crassispina).
  • TEC-mediated condition means any disease or other deleterious condition in which TEC-kinases are known to play a role.
  • TEC-kinases Such conditions include those described herein and in Melcher, M et al., "The Role of TEC Family Kinases in Inflammatory Processes", Anti-Inflammatory & Anti-Allergy Agents in Medicinal Chemistry, Vol. 6, No. 1, pp. 61-69 (Feb. 2007). Accordingly, another embodiment of the present invention relates to treating or lessening the severity of one or more diseases in which TEC-kinases are known to play a role.
  • the present invention relates to a method of treating or lessening the severity of a disease or condition selected from autoimmune, inflammatory, proliferative, and hyperproliferative diseases and immunologically-mediated diseases including rejection of transplanted organs or tissues and Acquired Immunodeficiency Syndrome (AIDS)(also known as HIV), wherein said method comprises administering to a patient in need thereof a composition of the present invention.
  • a disease or condition selected from autoimmune, inflammatory, proliferative, and hyperproliferative diseases and immunologically-mediated diseases including rejection of transplanted organs or tissues and Acquired Immunodeficiency Syndrome (AIDS)(also known as HIV)
  • AIDS Acquired Immunodeficiency Syndrome
  • the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with TEC-kinases including diseases of the respiratory tract including, without limitation, reversible obstructive airways diseases including asthma, such as bronchial, allergic, intrinsic, extrinsic and dust asthma, particularly chronic or inveterate asthma (e.g., late asthma airways hyper- responsiveness) and bronchitis.
  • diseases of the respiratory tract including, without limitation, reversible obstructive airways diseases including asthma, such as bronchial, allergic, intrinsic, extrinsic and dust asthma, particularly chronic or inveterate asthma (e.g., late asthma airways hyper- responsiveness) and bronchitis.
  • the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with TEC-kinases including those conditions characterized by inflammation of the nasal mucus membrane, including acute rhinitis, allergic, atrophic rhinitis and chronic rhinitis including rhinitis caseosa, hypertrophic rhinitis, rhinitis purulenta, rhinitis sicca and rhinitis medicamentosa; membranous rhinitis including croupous, fibrinous and pseudomembranous rhinitis and scrofoulous rhinitis, seasonal rhinitis including rhinitis nervosa (hay fever) and vasomotor rhinitis, sarcoidosis, farmer's lung and related diseases, fibroid lung, and idiopathic interstitial pneumonia.
  • TEC-kinases including those conditions characterized by inflammation of the nasal mucus membrane, including acute rhinitis, allergic
  • the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with TEC-kinases including diseases of the bone and joints including, without limitation, rheumatoid arthritis, seronegative spondyloarthropathies (including ankylosing spondylitis, psoriatic arthritis and Reiter's disease), Behcet's disease, Sjogren's syndrome, systemic sclerosis, osteoporosis, bone cancer, and bone metastasis.
  • diseases of the bone and joints including, without limitation, rheumatoid arthritis, seronegative spondyloarthropathies (including ankylosing spondylitis, psoriatic arthritis and Reiter's disease), Behcet's disease, Sjogren's syndrome, systemic sclerosis, osteoporosis, bone cancer, and bone metastasis.
  • the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with TEC-kinases including diseases and disorders of the skin, including, without limitation, psoriasis, systemic sclerosis, atopical dermatitis, contact dermatitis and other eczematous dermatitis, seborrhoetic dermatitis, Lichen planus, pemphigus, bullous pemphigus, epidermolysis bullosa, urticaria, angiodermas, vasculitides, erythemas, cutaneous eosinophilias, uveitis, alopecia, areata and vernal conjunctivitis.
  • diseases and disorders of the skin including, without limitation, psoriasis, systemic sclerosis, atopical dermatitis, contact dermatitis and other eczematous dermatitis, seborrhoetic dermatitis, Lichen planus, pemph
  • the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with TEC-kinases including diseases and disorders of the gastrointestinal tract, including, without limitation, celiac disease, proctitis, eosinophilic gastro-enteritis, mastocytosis, pancreatitis, Crohn's disease, ulcerative colitis, food-related allergies which have effects remote from the gut, e. g. migraine, rhinitis and eczema.
  • diseases and disorders of the gastrointestinal tract including, without limitation, celiac disease, proctitis, eosinophilic gastro-enteritis, mastocytosis, pancreatitis, Crohn's disease, ulcerative colitis, food-related allergies which have effects remote from the gut, e. g. migraine, rhinitis and eczema.
  • the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with TEC-kinases including those diseases and disorders of other tissues and systemic disease, including, without limitation, multiple sclerosis, artherosclerosis, lupus erythematosus, systemic lupus erythematosus, Hashimoto's thyroiditis, myasthenia gravis, type I diabetes, nephrotic syndrome, eosinophilia fascitis, hyper IgE syndrome, lepromatous leprosy, sezary syndrome and idiopathic thrombocytopenia purpura, restenosis following angioplasty, tumours (for example leukemia, lymphomas, and prostate cancers), and artherosclerosis.
  • diseases and disorders of other tissues and systemic disease including, without limitation, multiple sclerosis, artherosclerosis, lupus erythematosus, systemic lupus erythematosus, Hashimoto's thyroiditis
  • the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with TEC-kinases including allograft rejection including, without limitation, acute and chronic allograft rejection following for example transplantation of kidney, heart, liver, lung, bone marrow, skin and cornea; and chronic graft versus host disease.
  • allograft rejection including, without limitation, acute and chronic allograft rejection following for example transplantation of kidney, heart, liver, lung, bone marrow, skin and cornea; and chronic graft versus host disease.
  • the present invention relates to a method of treating or lessening the severity of one or more of the diseases or conditions associated with TEC-kinases, as recited above, wherein said method comprises administering to a patient in need thereof a compound or composition according to the present invention.
  • Bmton's tyrosine kinase Bmton's tyrosine kinase
  • BTK Bruton's tyrosine kinase
  • BCR cell surface B-cell receptor
  • BTK is a key regulator of B-cell development, activation, signaling, and survival (Kurosaki, Curr Op Imm, 2000, 276-281; Schaeffer and Schwartzberg, Curr Op Imm 2000, 282- 288).
  • BTK plays a role in a number of other hematopoietic cell signaling pathways, e.g., Toll like receptor (TLR) and cytokine receptor-mediated TNF- ⁇ production in macrophages, IgE receptor (Fc epsilon RI) signaling in mast cells, inhibition of Fas/ APO-I apoptotic signaling in B-lineage lymphoid cells, and collagen-stimulated platelet aggregation.
  • TLR Toll like receptor
  • Fc epsilon RI IgE receptor
  • BTK also plays a crucial role in mast cell activation through the high-affinity IgE receptor (Fc epsilon RI). BTK deficient murine mast cells have reduced degranulation and decreased production of proinflammatory cytokines following Fc epsilon RI cross-linking (Kawakami et al. Journal of Leukocyte Biology 65: 286-290).
  • the present invention provides a method for treating a BTK-mediated disorder comprising the step of administering to a patient in need thereof a compound of the present invention, or pharmaceutically acceptable composition thereof.
  • the term "BTK-mediated" disorders or conditions as used herein means any disease or other deleterious condition in which BTK, or a mutant thereof, is known to play a role. Accordingly, another embodiment of the present invention relates to treating or lessening the severity of one or more diseases in which BTK, or a mutant thereof, is known to play a role. Specifically, the present invention relates to a method of treating or lessening the severity of a disease or condition selected from a proliferative disorder or an autoimmune disorder, wherein said method comprises administering to a patient in need thereof a compound or composition according to the present invention.
  • the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with BTK.
  • the disease or condition is an autoimmune disease, e.g., inflammatory bowel disease, arthritis, lupus, rheumatoid arthritis, psoriatic arthritis, osteoarthritis, Still's disease, juvenile arthritis, diabetes, myasthenia gravis, Hashimoto's thyroiditis, Ord's thyroiditis, Graves' disease, Sjogren's syndrome, multiple sclerosis, Guillain-Barre syndrome, acute disseminated encephalomyelitis, Addison's disease, opsoclonus-myoclonus syndrome, ankylosing spondylosis, antiphospholipid antibody syndrome, aplastic anemia, autoimmune hepatitis, celiac disease, Goodpasture's syndrome, idiopathic thrombocytopenic purpura, optic neuritis, s
  • an autoimmune disease e.g.
  • the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with BTK, wherein the disease or condition is selected from heteroimmune conditions or diseases, which include, but are not limited to graft versus host disease, transplantation, transfusion, anaphylaxis, allergies (e.g., allergies to plant pollens, latex, drugs, foods, insect poisons, animal hair, animal dander, dust mites, or cockroach calyx), type I hypersensitivity, allergic conjunctivitis, allergic rhinitis, and atopic dermatitis.
  • heteroimmune conditions or diseases include, but are not limited to graft versus host disease, transplantation, transfusion, anaphylaxis, allergies (e.g., allergies to plant pollens, latex, drugs, foods, insect poisons, animal hair, animal dander, dust mites, or cockroach calyx), type I hypersensitivity, allergic conjunctivitis, allergic rhinitis, and a
  • the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with BTK, wherein the disease or condition is selected from an inflammatory disease, e.g., asthma, appendicitis, blepharitis, bronchiolitis, bronchitis, bursitis, cervicitis, cholangitis, cholecystitis, colitis, conjunctivitis, cystitis, dacryo adenitis, dermatitis, dermatomyositis, encephalitis, endocarditis, endometritis, enteritis, enterocolitis, epicondylitis, epididymitis, fasciitis, f ⁇ brositis, gastritis, gastroenteritis, hepatitis, hidradenitis suppurativa, laryngitis, mastitis, meningitis, myelitis myocarditis, myositis
  • an inflammatory disease e
  • the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with BTK, wherein the disease or condition is selected from a cancer.
  • the cancer is a B-cell proliferative disorder, e.g., diffuse large B cell lymphoma, follicular lymphoma, chronic lymphocytic lymphoma, chronic lymphocytic leukemia, acute lymphocytic leukemia, B-cell prolymphocytic leukemia, lymphoplasmacytic lymphoma/Waldenstrom macroglobulinemia, splenic marginal zone lymphoma, multiple myeloma (also known as plasma cell myeloma), non- Hodgkin's lymphoma, Hodgkin's lymphoma, plasmacytoma, extranodal marginal zone B cell lymphoma, nodal marginal zone B cell lymphoma, mantle cell lymphoma, mediastinal (
  • the cancer is breast cancer, prostate cancer, or cancer of the mast cells (e.g., mastocytoma, mast cell leukemia, mast cell sarcoma, systemic mastocytosis).
  • the cancer is bone cancer.
  • the cancer is of other primary origin and metastasizes to the bone.
  • the present invention provides a method for treating or lessening the severity of one or more diseases or conditions associated with BTK including diseases of the bone and joints including, without limitation, rheumatoid arthritis, seronegative spondyloarthropathies (including ankylosing spondylitis, psoriatic arthritis and Reiter's disease), Behcet's disease, Sjogren's syndrome, systemic sclerosis, osteoporosis, bone cancer, and bone metastasis.
  • diseases of the bone and joints including, without limitation, rheumatoid arthritis, seronegative spondyloarthropathies (including ankylosing spondylitis, psoriatic arthritis and Reiter's disease), Behcet's disease, Sjogren's syndrome, systemic sclerosis, osteoporosis, bone cancer, and bone metastasis.
  • the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with BTK, wherein the disease or condition is selected from a thromboembolic disorder, e.g., myocardial infarct, angina pectoris, reocclusion after angioplasty, restenosis after angioplasty, reocclusion after aortocoronary bypass, restenosis after aortocoronary bypass, stroke, transitory ischemia, a peripheral arterial occlusive disorder, pulmonary embolism, or deep venous thrombosis.
  • a thromboembolic disorder e.g., myocardial infarct, angina pectoris, reocclusion after angioplasty, restenosis after angioplasty, reocclusion after aortocoronary bypass, restenosis after aortocoronary bypass, stroke, transitory ischemia, a peripheral arterial occlusive disorder, pulmonary embo
  • the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with BTK, including infectious and noninfectious inflammatory events and autoimmune and other inflammatory diseases.
  • autoimmune and inflammatory diseases, disorders, and syndromes include inflammatory pelvic disease, urethritis, skin sunburn, sinusitis, pneumonitis, encephalitis, meningitis, myocarditis, nephritis, osteomyelitis, myositis, hepatitis, gastritis, enteritis, dermatitis, gingivitis, appendicitis, pancreatitis, cholocystitus, agammaglobulinemia, psoriasis, allergy, Crohn's disease, irritable bowel syndrome, ulcerative colitis, Sjogren's disease, tissue graft rejection, hyperacute rejection of transplanted organs, asthma, allergic rhinitis, chronic obstructive pulmonary disease (COP
  • the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with BTK, selected from rheumatoid arthritis, multiple sclerosis, B-cell chronic lymphocytic leukemia, acute lymphocytic leukemia, hairy cell leukemia, non-Hodgkin's lymphoma, Hodgkin's lymphoma, multiple myeloma, bone cancer, bone metastasis, osteoporosis, irritable bowel syndrome, Crohn's disease, lupus and renal transplant.
  • ITK ITK
  • Interleukin-2 inducible T-cell kinase is expressed in T cells, mast cells and natural killer cells. It is activated in T cells upon stimulation of the T cell receptor (TCR), and in mast cells upon activation of the high affinity IgE receptor. Following receptor stimulation in T cells, Lck, a Src tyrosine kinase family member, phosphorylates Y511 in the kinase domain activation loop of ITK (S. D. Heyeck et al, 1997, J. Biol. Chem, 272, 25401-25408). Activated ITK, together with Zap-70 is required for phosphorylation and activation of PLC-gamma (S. C.
  • PLC-gamma catalyzes the formation of inositol 1,4,5-triphosphate and diacylglycerol, leading to calcium mobilization and PKC activation, respectively. These events activate numerous downstream pathways and lead ultimately to degranulation (mast cells) and cytokine gene expression (T cells) (Y. Kawakami et al., 1999, J. Leukocyte Biol, 65, 286-290).
  • ITK knockout mice The role of ITK in T cell activation has been confirmed in ITK knockout mice.
  • CD4 + T cells from ITK knockout mice have a diminished proliferative response in a mixed lymphocyte reaction or upon Con A or anti-CD3 stimulation.
  • T cells from ITK knockout mice produced little IL-2 upon TCR stimulation resulting in reduced proliferation of these cells.
  • ITK deficient CD4 + T cells produced reduced levels of cytokines including IL-4, IL-5 and IL- 13 upon stimulation of the TCR, even after priming with inducing conditions (D. J. Fowell, 1999, Immunity, 11, 399-409).
  • T cells play an important role in regulating the immune response (Powrie and Coffman, 1993, Immunology Today, 14, 270-274). Indeed, activation of T cells is often the initiating event in immunological disorders. Following activation of the TCR, there is an influx of calcium that is required for T cell activation. Upon activation, T cells produce cytokines, including IL-2, 4, 5, 9, 10, and 13 leading to T cell proliferation, differentiation, and effector function. Clinical studies with inhibitors of IL-2 have shown that interference with T cell activation and proliferation effectively suppresses immune response in vivo (Waldmann, 1993, Immunology Today, 14, 264-270).
  • agents that inhibit T lymphocyte activation and subsequent cytokine production are therapeutically useful for selectively suppressing the immune response in a patient in need of such immunosuppression.
  • Mast cells play a critical roll in asthma and allergic disorders by releasing proinflammatory mediators and cytokines.
  • Antigen-mediated aggregation of Fc.epsilon.RI the high- affinity receptor for IgE, results in activation of mast cells (D. B. Corry et al., 1999, Nature, 402, B 18-23). This triggers a series of signaling events resulting in the release of mediators, including histamine, proteases, leukotrienes and cytokines (J. R.
  • mice results in reduced T cell receptor (TCR)-induced proliferation and secretion of the cytokines IL-2, IL-4, IL-5, IL-10 and IFN-y (Schaeffer et al, Science 284; 638-641 (1999) ), Fowell et al, Immunity 11, 399-409 (1999), Schaeffer et al, Nature Immunology 2 (12): 1183-1188 (2001) )).
  • TCR T cell receptor
  • ITK Lung inflammation, eosinophil infiltration and mucus production are drastically reduced in ITK-/-mice in response to challenge with the allergen OVA (Mueller et al, Journal of Immunology 170: 5056-5063 (2003)). ITK has also been implicated in atopic dermatitis. This gene has been reported to be more highly expressed in peripheral blood T cells from patients with moderate and/or severe atopic dermatitis than in controls or patients with mild atopic dermatitis (Matsumoto et al, International Archives of Allergy and Immunology 129: 327-340 (2002)).
  • Splenocytes from RLK-/-mice secrete half the IL-2 produced by wild type animals in response to TCR engagement (Schaeffer et al, Science 284: 638-641 (1999)), while combined deletion of ITK and RLK in mice leads to a profound inhibition of TCR-induced responses including proliferation and production of the cytokines IL-2, IL-4, IL-5 and IFN -y (Schaeffer et al, Nature Immunology 2 (12): 1183-1188 (2001), Schaeffer et al, Science 284: 638-641 (1999)).
  • Intracellular signalling following TCR engagement is effected in ITK/RLK deficient T cells; inositol triphosphate production, calcium mobilization, MAP kinase activation, and activation of the transcription factors NFAT and AP-I are all reduced (Schaeffer et al, Science 284: 638-641 (1999), Schaeffer et al, Nature Immunology 2 (12): 1183-1188 (2001)).
  • the present invention provides a method for treating an ITK-mediated disorder comprising the step of administering to a patient in need thereof a compound of the present invention, or pharmaceutically acceptable composition thereof.
  • the term "ITK-mediated" disorders or conditions as used herein means any disease or other deleterious condition in which ITK, or a mutant thereof, is known to play a role. Accordingly, another embodiment of the present invention relates to treating or lessening the severity of one or more diseases in which ITK, or a mutant thereof, is known to play a role. Specifically, the present invention relates to a method of treating or lessening the severity of a disease or condition selected from a mast cell-mediated condition, a basophil- mediated disorder, an immune or allergic disorder, wherein said method comprises administering to a patient in need thereof a compound or composition according to the present invention.
  • the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with ITK, wherein the disease or condition is an immune disorder, including inflammatory diseases, autoimmune diseases, organ and bone marrow transplant rejection and other disorders associated with T cell- mediated immune response or mast cell-mediated immune response.
  • an immune disorder including inflammatory diseases, autoimmune diseases, organ and bone marrow transplant rejection and other disorders associated with T cell- mediated immune response or mast cell-mediated immune response.
  • the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with ITK, wherein the disease or condition is acute or chronic inflammation, an allergy, contact dermatitis, psoriasis, rheumatoid arthritis, multiple sclerosis, type 1 diabetes, inflammatory bowel disease, Guillain- Barre syndrome, Crohn's disease, ulcerative colitis, cancer, graft versus host disease (and other forms of organ or bone marrow transplant rejection) or lupus erythematosus.
  • the disease or condition is acute or chronic inflammation, an allergy, contact dermatitis, psoriasis, rheumatoid arthritis, multiple sclerosis, type 1 diabetes, inflammatory bowel disease, Guillain- Barre syndrome, Crohn's disease, ulcerative colitis, cancer, graft versus host disease (and other forms of organ or bone marrow transplant rejection) or lupus erythematosus.
  • the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with ITK, wherein the disease or condition is a mast cell driven conditions, a basophil-mediated disorder, reversible obstructive airway disease, asthma, rhinitis, chronic obstructive pulmonary disease (COPD), peripheral T-cell lymphomas or HIV [also known as Acquired Immunodeficiency Syndrome
  • the Janus kinases are a family of tyrosine kinases consisting of JAKl, JAK2,
  • JAK3 and TYK2 The JAKs play a critical role in cytokine signaling.
  • the down-stream substrates of the JAK family of kinases include the signal transducer and activator of transcription (STAT) proteins.
  • STAT signal transducer and activator of transcription
  • JAK/STAT signaling has been implicated in the mediation of many abnormal immune responses such as allergies, asthma, autoimmune diseases such as transplant rejection, rheumatoid arthritis, amyotrophic lateral sclerosis and multiple sclerosis as well as in solid and hematologic malignancies such as leukemias and lymphomas.
  • the pharmaceutical intervention in the JAK/STAT pathway has been reviewed [Frank, MoI. Med. 5 :
  • JAKl, JAK2, and TYK2 are ubiquitously expressed, while JAK3 is predominantly expressed in hematopoietic cells. JAK3 binds exclusively to the common cytokine receptor gamma chain (yc) and is activated by IL-2, IL-4, IL-7, IL-9, and IL-15.
  • yc common cytokine receptor gamma chain
  • IL-4-mediated STAT -phosphorylation has been implicated as the mechanism involved in early and late stages of rheumatoid arthritis (RA). Up-regulation of proinflammatory cytokines in RA synovium and synovial fluid is a characteristic of the disease. It has been demostrated that IL-4-mediated activation of IL-4/STAT pathway is mediated through the Janus kinases (JAK 1 & 3) and that IL-4-associated JAK kinases are expressed in the RA synovium
  • Familial amyotrophic lateral sclerosis is a fatal neurodegenerative disorder affecting about 10% of ALS patients.
  • the survival rates of FALS mice were increased upon treatment with a JAK3 specific inhibitor. This confirmed that JAK3 plays a role in FALS [Trieu, et al, Biochem. Biophys. Res. Commun. 267 : 22-25 (2000)].
  • STAT Signal transducer and activator of transcription
  • JAK3 specific compounds were shown to inhibit the clonogenic growth of JAK3 -expressing cell lines DAUDI, RAMOS, LCl ; 19, NALM-6, MOLT-3 and HL-60. Inhibition of JAK3 and
  • TYK 2 abrogated tyrosine phosphorylation of STAT3, and inhibited cell growth of mycosis fungoides, a form of cutaneous T cell lymphoma.
  • the invention provides a method for treating or lessening the severity of a JAK3 -mediated disease or condition in a patient comprising the step of administering to said patient a composition according to the present invention.
  • J AK3 -mediated disease means any disease or other deleterious condition in which a JAK3 kinase is known to play a role.
  • another embodiment of the present invention relates to treating or lessening the severity of one or more diseases in which JAK3 is known to play a role.
  • the present invention relates to a method of treating or lessening the severity of a disease or condition selected from immune responses such as allergic or type I hypersensitivity reactions, asthma, autoimmune diseases such as transplant rejection, graft versus host disease, rheumatoid arthritis, amyotrophic lateral sclerosis, and multiple sclerosis, neurodegenerative disorders such as familial amyotrophic lateral sclerosis (FALS), as well as in solid and hematologic malignancies such as leukemias and lymphomas, wherein said method comprises administering to a patient in need thereof a composition according to the present invention.
  • a disease or condition selected from immune responses such as allergic or type I hypersensitivity reactions, asthma, autoimmune diseases such as transplant rejection, graft versus host disease, rheumatoid arthritis, amyotrophic lateral sclerosis, and multiple sclerosis, neurodegenerative disorders such as familial amyotrophic lateral sclerosis (FALS), as well as in solid and hematologic malignancies such as
  • the compounds and compositions, according to the method of the present invention may be administered using any amount and any route of administration effective for treating or lessening the severity of cancer, an autoimmune disorder, a neurodegenerative or neurological disorder, schizophrenia, a bone-related disorder, liver disease, or a cardiac disorder.
  • the exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular agent, its mode of administration, and the like.
  • 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.
  • 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.
  • patient means an animal, preferably a mammal, and most preferably a human.
  • compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like, depending on the severity of the infection being treated.
  • the compounds of the invention may be administered orally or parenterally at dosage levels of about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect.
  • 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-butylene 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 commonly used in the art such as, for example, water or other solvents,
  • the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • 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.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.
  • 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.
  • a compound of the present invention 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, delayed absorption of a parenterally administered compound form is accomplished by dissolving or suspending the compound in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the compound in biodegradable polymers such as polylactide- polyglycolide.
  • 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.
  • 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, ear drops, 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.
  • the invention relates to a method of inhibiting 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 invention relates to a method of inhibiting ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3, or a mutant thereof, 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 invention relates to a method of irreversibly inhibiting ErbBl, ErbB2, ErbB3, ErbB4, a TEC- kinase, and/or JAK3,, or a mutant thereof, 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 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 protein kinase, or a protein kinase selected from ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3, or a mutant thereof, activity in a biological sample is useful for a variety of purposes that are known to one of skill in the art. Examples of such purposes include, but are not limited to, blood transfusion, organ transplantation, biological specimen storage, and biological assays.
  • Another embodiment of the present invention relates to a method of inhibiting protein 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.
  • the invention relates to a method of inhibiting one or more of ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3,, or a mutant thereof, activity in a patient comprising the step of administering to said patient a compound of the present invention, or a composition comprising said compound.
  • the invention relates to a method of irreversibly inhibiting one or more of ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3,, or a mutant thereof, activity in a patient comprising the step of administering to said patient a compound of the present invention, or a composition comprising said compound.
  • the present invention provides a method for treating a disorder mediated by one or more of ErbBl, ErbB2, ErbB3, ErbB4, a TEC- kinase, and/or JAK3, or a mutant thereof, in a patient in need thereof, comprising the step of administering to said patient a compound according to the present invention or pharmaceutically acceptable composition thereof.
  • a disorder mediated by one or more of ErbBl, ErbB2, ErbB3, ErbB4, a TEC- kinase, and/or JAK3, or a mutant thereof, in a patient in need thereof, comprising the step of administering to said patient a compound according to the present invention or pharmaceutically acceptable 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 include, but are not limited to, Adriamycin, dexamethasone, vincristine, cyclophosphamide, fluorouracil, topotecan, taxol, interferons, platinum derivatives, taxane (e.g., paclitaxel), vinca alkaloids (e.g., vinblastine), anthracyclines (e.g., doxorubicin), epipodophyllotoxins (e.g., etoposide), cisplatin, an mTOR inhibitor (e.g., a rapamycin), methotrexate, actinomycin D, dolastatin 10, colchicine, emetine, trimetrexate, metoprine, cyclosporine, daunorubicin, teniposide
  • chemotherapeutic agents include, but are not limited to, Adriamycin, dexamethasone, vincristine, cyclophosphamide, fluorouracil
  • a compound of the present invention is administered in combination with a biologic agent, such as Avastin or VECTIBIX.
  • a biologic agent such as Avastin or VECTIBIX.
  • compounds of the present invention, or a pharmaceutically acceptable composition thereof are administered in combination with an antiproliferative or chemotherapeutic agent selected from any one or more of abarelix, aldesleukin, alemtuzumab, alitretinoin, allopurinol, altretamine, amifostine, anastrozole, arsenic trioxide, asparaginase, azacitidine, BCG Live, bevacuzimab, fluorouracil, bexarotene, bleomycin, bortezomib, busulfan, calusterone, capecitabine, camptothecin, carboplatin, carmustine, celecoxib, cetuximab, chlorambucil, cladribine, clofarabine,
  • agents the inhibitors of this invention may also be combined with include, without limitation: treatments for Alzheimer's Disease such as donepezil hydrochloride (Aricept ® ) and rivastigmine (Exelon ® ); treatments for Parkinson's Disease such as L- DOPA/carbidopa, entacapone, ropinrole, pramipexole, bromocriptine, pergolide, trihexephendyl, and amantadine; agents for treating Multiple Sclerosis (MS) such as beta interferon (e.g., Avonex ® and Rebif ® ), glatiramer acetate (Copaxone ® ), and mitoxantrone; treatments for asthma such as albuterol and montelukast (Singulair ® ); agents for treating schizophrenia such as zyprexa, risperdal, seroquel, and haloperidol; anti-inflammatory agents such as corticosteroids, TNF
  • Those additional agents may be administered separately from an inventive compound-containing composition, as part of a multiple dosage regimen.
  • those agents may be part of a single dosage form, mixed together with a compound of this invention in a single composition. If administered as part of a multiple dosage regime, the two active agents may be submitted simultaneously, sequentially or within a period of time from one another normally within five hours from one another.
  • the term “combination,” “combined,” and related terms refers to the simultaneous or sequential administration of therapeutic agents in accordance with this invention.
  • a compound of the present invention may be administered with another therapeutic agent simultaneously or sequentially in separate unit dosage forms or together in a single unit dosage form.
  • the present invention provides a single unit dosage form comprising a provided compound, an additional therapeutic agent, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
  • compositions of this invention should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of an inventive can be administered.
  • compositions which 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 - 1,000 ⁇ g/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.
  • Implantable devices coated with a compound of this invention are another embodiment of the present invention. 5.
  • a compound of the present invention may be tethered to a detectable moiety to form a probe compound.
  • a probe compound of the invention comprises an irreversible protein kinase inhibitor of formula I-a or I-b, as described herein, a detectable moiety, and a tethering moiety that attaches the inhibitor to the detectable moiety.
  • probe compounds of the present invention comprise a provided compound of formula I-a or I-b tethered to a detectable moiety, R 1 , by a bivalent tethering moiety, -T-.
  • the tethering moiety may be attached to a compound of formula I-a or I- b via Ring A, Ring B, or R 1 .
  • R 1 is a bivalent warhead group denoted as R 1 .
  • a provided probe compound is selected from any of formula V-a, V-b, VI-a, VI-b, VII-a, or VII-b:
  • R 1 is a bivalent warhead group
  • T is a bivalent tethering moiety
  • R 1 is a detectable moiety.
  • R 1 is a detectable moiety selected from a primary label or a secondary label.
  • R 1 is a detectable moiety selected from a fluorescent label (e.g., a fluorescent dye or a fluorophore), a mass-tag, a chemiluminescent group, a chromophore, an electron dense group, or an energy transfer agent.
  • a fluorescent label e.g., a fluorescent dye or a fluorophore
  • mass-tag e.g., a chemiluminescent group, a chromophore, an electron dense group, or an energy transfer agent.
  • detectable moiety is used interchangeably with the term “label” and “reporter” and relates to any moiety capable of being detected, e.g., primary labels and secondary labels.
  • a presence of a detectable moiety can be measured using methods for quantifying (in absolute, approximate or relative terms) the detectable moiety in a system under study.
  • such methods are well known to one of ordinary skill in the art and include any methods that quantify a reporter moiety (e.g., a label, a dye, a photocrosslinker, a cytotoxic compound, a drug, an affinity label, a photoaffinity label, a reactive compound, an antibody or antibody fragment, a biomaterial, a nanoparticle, a spin label, a fluorophore, a metal- containing moiety, a radioactive moiety, quantum dot(s), a novel functional group, a group that covalently or noncovalently interacts with other molecules, a photocaged moiety, an actinic radiation excitable moiety, a ligand, a photoisomerizable moiety, biotin, a biotin analog (e.g., biotin sulfoxide), a moiety incorporating a heavy atom, a chemically cleavable group, a photocleavable group, a redox-active agent, an a reporter mo
  • Primary labels such as radioisotopes (e.g., tritium, 32 P, 33 P, 35 S, 14 C, 123 I, 124 I, 125 I, or 131 I), mass-tags including, but not limited to, stable isotopes (e.g., 13 C, 2 H, 17 O, 18 O, 15 N, 19 F, and 127 I), positron emitting isotopes (e.g., 11 C, 18 F, 13 N, 124 I, and 15 O), and fluorescent labels are signal generating reporter groups which can be detected without further modifications.
  • radioisotopes e.g., tritium, 32 P, 33 P, 35 S, 14 C, 123 I, 124 I, 125 I, or 131 I
  • mass-tags including, but not limited to, stable isotopes (e.g., 13 C, 2 H, 17 O, 18 O, 15 N, 19 F, and 127 I), positron emitting isotopes (e.g., 11
  • Detectable moities may be analyzed by methods including, but not limited to fluorescence, positron emission tomography, SPECT medical imaging, chemiluminescence, electron-spin resonance, ultraviolet/visible absorbance spectroscopy, mass spectrometry, nuclear magnetic resonance, magnetic resonance, flow cytometry, autoradiography, scintillation counting, phosphoimaging, and electrochemical methods.
  • secondary label refers to moieties such as biotin and various protein antigens that require the presence of a second intermediate for production of a detectable signal.
  • the secondary intermediate may include streptavidin-enzyme conjugates.
  • antigen labels secondary intermediates may include antibody-enzyme conjugates.
  • fluorescent label refers to moieties that absorb light energy at a defined excitation wavelength and emit light energy at a different wavelength.
  • fluorescent labels include, but are not limited to: Alexa Fluor dyes (Alexa Fluor 350, Alexa Fluor 488, Alexa Fluor 532, Alexa Fluor 546, Alexa Fluor 568, Alexa Fluor 594, Alexa Fluor 633, Alexa Fluor 660 and Alexa Fluor 680), AMCA, AMCA-S, BODIPY dyes (BODIPY FL, BODIPY R6G, BODIPY TMR, BODIPY TR, BODIPY 493/503, BODIPY 530/550, BODIPY 558/568, BODIPY 564/570, BODIPY 576/589, BODIPY 581/591, BODIPY 630/650, BODIPY 650/665), Carboxyr
  • Carboxytetramethylrhodamine (TAMRA), Texas Red, Texas Red-X, 5(6)-Carboxyfluorescein, 2,7-Dichlorofluorescein, N,N-Bis(2,4,6-trimethylphenyl)-3,4:9,10-perylenebis(dicarboximide, HPTS, Ethyl Eosin, DY-490XL MegaStokes, DY-485XL MegaStokes, Adirondack Green 520, ATTO 465, ATTO 488, ATTO 495, Y0Y0-l,5-FAM, BCECF, dichlorofluorescein, rhodamine 110, rhodamine 123, YO-PRO-I, SYTOX Green, Sodium Green, SYBR Green I, Alexa Fluor 500, FITC, Fluo-3, Fluo-4, fluoro-emerald, YoYo-I ssDNA, YoYo-I dsDNA, Yo
  • mass-tag refers to any moiety that is capable of being uniquely detected by virtue of its mass using mass spectrometry (MS) detection techniques.
  • mass-tags include electrophore release tags such as N-[3-[4'-[(p- Methoxytetrafluorobenzyl)oxy]phenyl]-3-methylglyceronyl]isonipecotic Acid, 4 ' -[2,3 ,5 ,6- Tetrafluoro-4-(pentafluorophenoxyl)]methyl acetophenone, and their derivatives.
  • mass-tags include, but are not limited to, nucleotides, dideoxynucleotides, oligonucleotides of varying length and base composition, oligopeptides, oligosaccharides, and other synthetic polymers of varying length and monomer composition.
  • nucleotides dideoxynucleotides
  • oligonucleotides of varying length and base composition oligopeptides, oligosaccharides
  • other synthetic polymers of varying length and monomer composition.
  • a large variety of organic molecules, both neutral and charged (biomolecules or synthetic compounds) of an appropriate mass range (100-2000 Daltons) may also be used as mass-tags.
  • Stable isotopes e.g., 13 C, 2 H, 17 O, 18 O, and 15 N
  • mass-tags may also be used as mass-tags.
  • chemiluminescent group refers to a group which emits light as a result of a chemical reaction without the addition of heat.
  • luminol 5-amino-2,3-dihydro-l,4-phthalazinedione
  • oxidants like hydrogen peroxide (H 2 O 2 ) in the presence of a base and a metal catalyst to produce an excited state product (3- aminophthalate, 3-APA).
  • chromophore refers to a molecule which absorbs light of visible wavelengths, UV wavelengths or IR wavelengths.
  • die refers to a soluble, coloring substance which contains a chromophore.
  • electrostatic dense group refers to a group which scatters electrons when irradiated with an electron beam.
  • groups include, but are not limited to, ammonium molybdate, bismuth subnitrate, cadmium iodide, carbohydrazide, ferric chloride hexahydrate, hexamethylene tetramine, indium trichloride anhydrous, lanthanum nitrate, lead acetate trihydrate, lead citrate trihydrate, lead nitrate, periodic acid, phosphomolybdic acid, phosphotungstic acid, potassium ferricyanide, potassium ferrocyanide, ruthenium red, silver nitrate, silver proteinate (Ag Assay: 8.0-8.5%) "Strong", silver tetraphenylporphin (S-TPPS), sodium chloroaurate, sodium tungstate, thallium nitrate, thiosemicarbazide (TSC), ur
  • the term "energy transfer agent,” as used herein, refers to a molecule which either donates or accepts energy from another molecule.
  • fluorescence resonance energy transfer FRET is a dipole-dipole coupling process by which the excited-state energy of a fluorescence donor molecule is non-radiatively transferred to an unexcited acceptor molecule which then fluorescently emits the donated energy at a longer wavelength.
  • moiety incorporating a heavy atom refers to a group which incorporates an ion of atom which is usually heavier than carbon.
  • such ions or atoms include, but are not limited to, silicon, tungsten, gold, lead, and uranium.
  • photoaffinity label refers to a label with a group, which, upon exposure to light, forms a linkage with a molecule for which the label has an affinity.
  • photocaged moiety refers to a group which, upon illumination at certain wavelengths, covalently or non-covalently binds other ions or molecules.
  • photoisomerizable moiety refers to a group wherein upon illumination with light changes from one isomeric form to another.
  • radioactive moiety refers to a group whose nuclei spontaneously give off nuclear radiation, such as alpha, beta, or gamma particles; wherein, alpha particles are helium nuclei, beta particles are electrons, and gamma particles are high energy photons.
  • spin label refers to molecules which contain an atom or a group of atoms exhibiting an unpaired electron spin (i.e. a stable paramagnetic group) that in some embodiments are detected by electron spin resonance spectroscopy and in other embodiments are attached to another molecule.
  • spin-label molecules include, but are not limited to, nitryl radicals and nitroxides, and in some embodiments are single spin-labels or double spin-labels.
  • quantum dots refers to colloidal semiconductor nanocrystals that in some embodiments are detected in the near-infrared and have extremely high quantum yields (i.e., very bright upon modest illumination).
  • a detectable moiety may be attached to a provided compound via a suitable substituent.
  • suitable substituent refers to a moiety that is capable of covalent attachment to a detectable moiety.
  • moieties are well known to one of ordinary skill in the art and include groups containing, e.g., a carboxylate moiety, an amino moiety, a thiol moiety, or a hydroxyl moiety, to name but a few. It will be appreciated that such moieties may be directly attached to a provided compound or via a tethering moiety, such as a bivalent saturated or unsaturated hydrocarbon chain.
  • detectable moieties are attached to a provided compound via click chemistry.
  • such moieties are attached via a 1,3-cycloaddition of an azide with an alkyne, optionally in the presence of a copper catalyst.
  • Methods of using click chemistry are known in the art and include those described by Rostovtsev et al. , Angew. Chem. Int. Ed. 2002, 41, 2596-99 and Sun et al., Bioconjugate Chem., 2006, JJ, 52-57.
  • a click ready inhibitor moiety is provided and reacted with a click ready -T-R 1 moiety.
  • click ready refers to a moiety containing an azide or alkyne for use in a click chemistry reaction.
  • the click ready inhibitor moiety comprises an azide.
  • the click ready -T-R 1 moiety comprises a strained cyclooctyne for use in a copper-free click chemistry reaction (for example, using methods described in Baskin et al., Proc. Natl. Acad. Sci. USA 2007, 104, 16793-16797).
  • the click ready inhibitor moiety is of one of the following formulae:
  • Ring A, Ring B, W 1 , W 2 , R y , R v , p, R x , and m are as defined above with respect to Formula I and described herein, and q is 1, 2, or 3.
  • exemplary click ready inhibitors include:
  • the click ready -T-R 1 moiety is of formula:
  • the detectable moiety, R 1 is selected from a label, a dye, a photocrosslinker, a cytotoxic compound, a drug, an affinity label, a photoaffmity label, a reactive compound, an antibody or antibody fragment, a biomatenal, a nanoparticle, a spin label, a fluorophore, a metal-contammg moiety, a radioactive moiety, quantum dot(s), a novel functional group, a group that covalently or noncovalently interacts with other molecules, a photocaged moiety, an actinic radiation excitable moiety, a ligand, a photoisomerizable moiety, biotm, a biotin analog (e g , biotm sulfoxide), a moiety incorporating a heavy atom, a chemically cleavable group, a photocleavable group, a redox-active agent, an isotopically labele
  • R 1 is biotm or an analog thereof In certain embodiments, R 1 is biotm In certain other embodiments, R 1 is biotm sulfoxide
  • R 1 is a fluorophore
  • the fluorophore is selected from Alexa Fluor dyes (Alexa Fluor 350, Alexa Fluor 488, Alexa Fluor 532, Alexa Fluor 546, Alexa Fluor 568, Alexa Fluor 594, Alexa Fluor 633, Alexa Fluor 660 and Alexa Fluor 680), AMCA, AMCA-S, BODIPY dyes (BODIPY FL, BODIPY R6G, BODIPY TMR, BODIPY TR, BODIPY 493/503, BODIPY 530/550, BODIPY 558/568, BODIPY 564/570, BODIPY 576/589, BODIPY 581/591, BODIPY 630/650, BODIPY 650/665), Carboxyrhodamine 6G, carboxy-X-rhodamine (ROX), Cascade Blue, Cascade Yellow, Coumarin 343,
  • a provided probe compound comprises a tethering moiety, -T-, that attaches the irreversible inhibitor to the detectable moiety.
  • tether or “tethering moiety” refers to any bivalent chemical spacer including, but not limited to, a covalent bond, a polymer, a water soluble polymer, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted heterocycloalkylalkyl, optionally substituted heterocycloalkylalkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkylalkenylalkyl, an optionally substituted amide moiety, an ether moiety, an ketone moiety, an ester moiety, an optionally substituted carbamate moiety, an optionally substituted
  • the tethering moiety, -T- is selected from a covalent bond, a polymer, a water soluble polymer, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkylalkyl, optionally substituted heterocycloalkylalkenyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted heterocycloalkylalkenylalkyl.
  • the tethering moiety is an optionally substituted heterocycle.
  • the heterocycle is selected from aziridine, oxirane, episulfide, azetidine, oxetane, pyrroline, tetrahydrofuran, tetrahydrothiophene, pyrrolidine, pyrazole, pyrrole, imidazole, triazole, tetrazole, oxazole, isoxazole, oxirene, thiazole, isothiazole, dithiolane, furan, thiophene, piperidine, tetrahydropyran, thiane, pyridine, pyran, thiapyrane, pyridazine, pyrimidine, pyrazine, piperazine, oxazine, thiazine, dithiane, and dioxane.
  • the heterocycle is piperazine.
  • the tethering moiety is optionally substituted with halogen, -CN, -OH, -NO 2 , alkyl, S(O), and S(O) 2 .
  • the water soluble polymer is a PEG group.
  • the tethering moiety provides sufficient spatial separation between the detectable moiety and the protein kinase inhibitor moiety. In further embodiments, the tethering moiety is stable. In yet a further embodiment, the tethering moiety does not substantially affect the response of the detectable moiety. In other embodiments, the tethering moiety provides chemical stability to the probe compound. In further embodiments, the tethering moiety provides sufficient solubility to the probe compound.
  • a tethering moiety, -T- such as a water soluble polymer is coupled at one end to a provided irreversible inhibitor and to a detectable moiety, R 1 , at the other end.
  • a water soluble polymer is coupled via a functional group or substituent of the provided irreversible inhibitor.
  • a water soluble polymer is coupled via a functional group or substituent of the reporter moiety.
  • examples of hydrophilic polymers for use in tethering moiety -T-, include, but are not limited to: polyalkyl ethers and alkoxy-capped analogs thereof (e.g., polyoxyethylene glycol, polyoxyethylene/propylene glycol, and methoxy or ethoxy-capped analogs thereof, polyoxyethylene glycol, the latter is also known as polyethylene glycol or PEG); polyvinylpyrrolidones; polyvinylalkyl ethers; polyoxazolines, polyalkyl oxazolines and polyhydroxyalkyl oxazolines; polyacrylamides, polyalkyl acrylamides, and polyhydroxyalkyl acrylamides (e.g., polyhydroxypropylmethacrylamide and derivatives thereof); polyhydroxyalkyl acrylates; polysialic acids and analogs thereof, hydrophilic peptide sequences; polysaccharides and their derivatives, including dextran and dextran derivatives
  • a water soluble polymer is any structural form including but not limited to linear, forked or branched.
  • multifunctional polymer derivatives include, but are not limited to, linear polymers having two termini, each terminus being bonded to a functional group which is the same or different.
  • a water polymer comprises a poly(ethylene glycol) moiety.
  • the molecular weight of the polymer is of a wide range, including but not limited to, between about 100 Da and about 100,000 Da or more.
  • the molecular weight of the polymer is between about 100 Da and about 100,000 Da, including but not limited to, about 100,000 Da, about 95,000 Da, about 90,000 Da, about 85,000 Da, about 80,000 Da, about 75,000 Da, about 70,000 Da, about 65,000 Da, about 60,000 Da, about 55,000 Da, about 50,000 Da, about 45,000 Da, about 40,000 Da, about 35,000 Da, 30,000 Da, about 25,000 Da, about 20,000 Da, about 15,000 Da, about 10,000 Da, about 9,000 Da, about 8,000 Da, about 7,000 Da, about 6,000 Da, about 5,000 Da, about 4,000 Da, about 3,000 Da, about 2,000 Da, about 1,000 Da, about 900 Da, about 800 Da, about 700 Da, about 600 Da, about 500 Da, about 400 Da, about 300 Da, about 200 Da, and about 100 Da.
  • the molecular weight of the polymer is between about 100 Da and 50,000 Da. In some embodiments, the molecular weight of the polymer is between about 100 Da and 40,000 Da. In some embodiments, the molecular weight of the polymer is between about 1,000 Da and 40,000 Da. In some embodiments, the molecular weight of the polymer is between about 5,000 Da and 40,000 Da. In some embodiments, the molecular weight of the polymer is between about 10,000 Da and 40,000 Da. In some embodiments, the poly(ethylene glycol) molecule is a branched polymer.
  • the molecular weight of the branched chain PEG is between about 1,000 Da and about 100,000 Da, including but not limited to, about 100,000 Da, about 95,000 Da, about 90,000 Da, about 85,000 Da, about 80,000 Da, about 75,000 Da, about 70,000 Da, about 65,000 Da, about 60,000 Da, about 55,000 Da, about 50,000 Da, about 45,000 Da, about 40,000 Da, about 35,000 Da, about 30,000 Da, about 25,000 Da, about 20,000 Da, about 15,000 Da, about 10,000 Da, about 9,000 Da, about 8,000 Da, about 7,000 Da, about 6,000 Da, about 5,000 Da, about 4,000 Da, about 3,000 Da, about 2,000 Da, and about 1,000 Da.
  • the molecular weight of a branched chain PEG is between about 1,000 Da and about 50,000 Da. In some embodiments, the molecular weight of a branched chain PEG is between about 1,000 Da and about 40,000 Da. In some embodiments, the molecular weight of a branched chain PEG is between about 5,000 Da and about 40,000 Da. In some embodiments, the molecular weight of a branched chain PEG is between about 5,000 Da and about 20,000 Da.
  • the foregoing list for substantially water soluble backbones is by no means exhaustive and is merely illustrative, and in some embodiments, polymeric materials having the qualities described above are suitable for use in methods and compositions described herein.
  • the resulting tethering moiety comprises the R 1 warhead group.
  • the phrase "comprises a warhead group” means that the tethering moiety formed by -R 1 -T- of formula V-a or V-b is either substituted with a warhead group or has such a warhead group incorporated within the tethering moiety.
  • the tethering moiety formed by -R 1 -T- may be substituted with an -L-Y warhead group, wherein such groups are as described herein.
  • the tethering moiety formed by -R 1 -T- has the appropriate features of a warhead group incorporated within the tethering moiety.
  • the tethering moiety formed by -R 1 -T- may include one or more units of unsaturation and optional substituents and/or heteroatoms which, in combination, result in a moiety that is capable of covalently modifying a protein kinase in accordance with the present invention.
  • Such -R 1 -T- tethering moiety are depicted below.
  • a methylene unit of an -R 1 -T- tethering moiety is replaced by a bivalent -L-Y'- moiety to provide a compound of formula V-a- «7 or V-b-M ⁇ :
  • a methylene unit of an -R 1 -T- tethering moiety is replaced by an -L(Y)- moiety to provide a compound of formula V-a-iv or V-b-/v:
  • Ring A, Ring B, m, p, R x , R y , W 2 , T, L, Y, and R 1 is as defined above and described in classes and subclasses herein.
  • a tethering moiety is substituted with an L-Y moiety to provide a compound of formula V-a-v or V-b-v:
  • Ring A, Ring B, m, p, R x , R y , W 2 , T, L, Y, and R 1 is as defined above and described in classes and subclasses herein.
  • the tethering moiety, -T- has one of the following structures:
  • the tethering moiety, -T- has the following structure:
  • the tethering moiety, -T- has the following structure: [00288] In certain other embodiments, the tethering moiety, -T-, has the following structure:
  • the tethering moiety, -T- has the following structure:
  • the tethering moiety, -T- has the following structure:
  • -T-R 1 is of the following structure:
  • -T-R 1 is of the following structure:
  • -T-R 1 is of the following structure:
  • VII-b is derived from any compound of Table 5.
  • the probe compound is one of the following structures:
  • a provided probe compound covalently modifies a phosphorylated conformation of a protein kinase.
  • the phosphorylated conformation of the protein kinase is either an active or inactive form of the protein kinase.
  • the phosphorylated conformation of the protein kinase is an active form of said kinase.
  • the probe compound is cell permeable.
  • the present invention provides a method for determining occupancy of a protein kinase by a provided irreversible inhibitor (i.e., a compound of formula I- a or I-b) in a patient, comprising providing one or more tissues, cell types, or a lysate thereof, obtained from a patient administered at least one dose of a compound of said irreversible inhibitor, contacting said tissue, cell type or lysate thereof with a probe compound (i.e., a compound of formula V-a, V-b, VI-a, VI-b, VII-a, or VII-b) to covalent modify at least one protein kinase present in said lysate, and measuring the amount of said protein kinase covalently modified by the probe compound to determine occupancy of said protein kinase by said compound of formula I-a or I-b as compared to occupancy of said protein kinase by said probe compound.
  • a probe compound i.e., a compound of formula V-a, V-b
  • the method further comprises the step of adjusting the dose of the compound of formula I-a or I-b to increase occupancy of the protein kinase. In certain other embodiments, the method further comprises the step of adjusting the dose of the compound of formula I-a or I-b to decrease occupancy of the protein kinase.
  • occupancy refers to the extent to which a protein kinase is modified by a provided covalent inhibitor compound.
  • a protein kinase is modified by a provided covalent inhibitor compound.
  • One of ordinary skill in the art would appreciate that it is desirable to administer the lowest dose possible to achieve the desired efficacious occupancy of the protein kinase.
  • the protein kinase to be modified is BTK. In other embodiments, the protein kinase to be modified is EGFR. In certain embodiments, the protein kinase is JAK. In certain other embodiments, the protein kinase is one or more of ErbBl, ErbB2, or ErbB4. In yet other embodiments, the protein kinase is TEC, ITK, or BMX.
  • the probe compound comprises the irreversible inhibitor for which occupancy is being determined.
  • the present invention provides a method for assessing the efficacy of a provided irreversible inhibitor in a mammal, comprising administering a provided irreversible inhibitor to the mammal, administering a provided probe compound to tissues or cells isolated from the mammal, or a lysate thereof, measuring the activity of the detectable moiety of the probe compound, and comparing the activity of the detectable moiety to a standard.
  • the present invention provides a method for assessing the pharmacodynamics of a provided irreversible inhibitor in a mammal, comprising administering a provided irreversible inhibitor to the mammal, administering a probe compound presented herein to one or more cell types, or a lysate thereof, isolated from the mammal, and measuring the activity of the detectable moiety of the probe compound at different time points following the administration of the inhibitor.
  • the present invention provides a method for in vitro labeling of a protein kinase comprising contacting said protein kinase with a probe compound described herein.
  • the contacting step comprises incubating the protein kinase with a probe compound presented herein.
  • the present invention provides a method for in vitro labeling of a protein kinase comprising contacting one or more cells or tissues, or a lysate thereof, expressing the protein kinase with a probe compound described herein.
  • the present invention provides a method for detecting a labeled protein kinase comprising separating proteins, the proteins comprising a protein kinase labeled by probe compound described herein, by electrophoresis and detecting the probe compound by fluorescence.
  • the present invention provides a method for assessing the pharmacodynamics of a provided irreversible inhibitor in vitro, comprising incubating the provided irreversible inhibitor with the target protein kinase, adding the probe compound presented herein to the target protein kinase, and determining the amount of target modified by the probe compound.
  • the probe compound is detected by binding to avidin, streptavidin, neutravidin, or captavidin.
  • the probe is detected by Western blot. In other embodiments, the probe is detected by ELISA. In certain embodiments, the probe is detected by flow cytometry.
  • the present invention provides a method for probing the kinome with irreversible inhibitors comprising incubating one or more cell types, or a lysate thereof, with a biotinylated probe compound to generate proteins modified with a biotin moiety, digesting the proteins, capturing with avidin or an analog thereof, and performing multidimensional LC-MS-MS to identify protein kinases modified by the probe compound and the adduction sites of said kinases.
  • the present invention provides a method for measuring protein synthesis in cells comprising incubating cells with an irreversible inhibitor of the target protein, forming lysates of the cells at specific time points, and incubating said cell lysates with an inventive probe compound to measure the appearance of free protein over an extended period of time.
  • the present invention provides a method for determining a dosing schedule in a mammal for maximizing occupancy of a target protein kinase comprising assaying a one or more cell types, or a lysate thereof, isolated from the mammal, (derived from, e.g., splenocytes, peripheral B cells, whole blood, lymph nodes, intestinal tissue, or other tissues) from a mammal administered a provided irreversible inhibitor of formula I-a or I-b, wherein the assaying step comprises contacting said one or more tissues, cell types, or a lysate thereof, with a provided probe compound and measuring the amount of protein kinase covalently modified by the probe compound.
  • Step-1 A) DIPEA, n-BuOH, 120 0 C, 30 min, MW; B) NMP, 200 0 C, 10 min, MW; C) NMP, 0 °C-30 min, rt-30 min. [00318] Step-1
  • Step-1 DIEA, n-BuOH, 110 0 C, 30 min, microwave; B) NMP, 200 0 C, 10 min, microwave; C) acryloyl chloride, NMP, 0 °C-30 min, rt-30 min.
  • Step-1 A) DIPEA, n-BuOH, 110 0 C, 30 min, MW; B) NMP, 200 0 C, 15 min, MW; C) acryloyl chloride, NMP, 0 °C-30 min, rt-30 min. [00334] Step-1
  • Step-1 A) n-butanol, DIPEA, 110 ° C, 45 min., M W;B ) NMP, 200°C, 10min., MW; C) oxal yl chloride, CH 3 CN, 1 ⁇ 2 h at 0 0 C, 2 h at 25 0 C, 5 min at 45 °C; D) NMP, 0 °C to 10 °C, 30 min. [00350] Step-1
  • Step-1 A) DIPEA, n-BuOH, 110 0 C, 30 min, MW; B) NMP, 200 0 C, 15 min, MW; C) acryloyl chloride, NMP, 0 °C-30 min, rt-30 min. [00360] Step-1
  • Step l A) DIPEA, n-butanol, 110 0 C, 1 h, MW; B) 1.5 N HCl, ethanol, 90 0 C, 30 min., MW; C) acryloyl chloride, NMP, 0 0 C, 30 min. [00380] Step l
  • Step S The procedure for synthesizing scaffold 7 is described in the experimental for Compound I- 11 herein. [00389] Step S
  • A') DPPA Benzyl alcohol, Et 3 N, toluene, 110 C, 12 h.
  • B' Pd(OH) 2 , Ammonium formate, EtOH, reflux, 6 h
  • Step-1 A) DIPEA, n-BuOH, 120 0 C, 30 min., MW; B) 1.5 N HCl, Ethanol, 100 0 C, 12 h; C) NMP, 0 0 C to rt, 1 h. [00413] Step-1
  • Step-1 A) Pd(OAC) 2 , BINAP, Cs 2 CO 3 , Toluene, 100 0 C, 16 h; B) NaH, CH 3 I, THF, 0 °C-30 min, rt-16 h.; C) Aniline, conc.HCl, Ethanol, 90 0 C, 60 min; D) H 2 , Pd/C, Ethanol, 16 h; E) acryloyl chloride, NMP, 0 0 C, 1 h. [00421] Step-1
  • Step-1 A) K 2 CO 3 , CH 3 CN, 65 U C, 8 h; B) Fe powder, NH 4 Cl, MeOH, 1,3- pheneylendiamine, DIPEA, n-BuOH, 120 0 C, 30 min, MW; D) Con.HCl, absolute ethanol, 110 0 C, 2 h; E) NMP, 0 0 C, 1 h. [00433] Step-1
  • Step-1 A) 2, DIPEA, THF, reflux; B) 4, Pd(OAc) 2 , X-Phos, CsCO 3 , dioxane, reflux, 12 h; C) TFA, DCM; D) 7, DIPEA, THF, -10 0 C.

Abstract

The present invention provides inhibitors of protein kinases of formula I-a and I-b, pharmaceutically acceptable compositions thereof, and methods of using the same.

Description

HETEROARYL COMPOUNDS AND USES THEREOF
TECHNICAL FIELD OF THE INVENTION
[0001] The present invention relates to compounds useful as inhibitors of protein kinases. The invention also provides pharmaceutically acceptable compositions comprising compounds of the present invention and methods of using said compositions in the treatment of various disorders.
BACKGROUND OF THE INVENTION
[0002] The search for new therapeutic agents has been greatly aided in recent years by a better understanding of the structure of enzymes and other biomolecules associated with diseases. One important class of enzymes that has been the subject of extensive study is protein kinases.
[0003] Protein kinases constitute a large family of structurally related enzymes that are responsible for the control of a variety of signal transduction processes within the cell. Protein kinases are thought to have evolved from a common ancestral gene due to the conservation of their structure and catalytic function. Almost all kinases contain a similar 250-300 amino acid catalytic domain. The kinases may be categorized into families by the substrates they phosphorylate (e.g., protein-tyrosine, protein-serine/threonine, lipids, etc.). [0004] In general, protein kinases mediate intracellular signaling by effecting a phosphoryl transfer from a nucleoside triphosphate to a protein acceptor that is involved in a signaling pathway. These phosphorylation events act as molecular on/off switches that can modulate or regulate the target protein biological function. These phosphorylation events are ultimately triggered in response to a variety of extracellular and other stimuli. Examples of such stimuli include environmental and chemical stress signals (e.g., osmotic shock, heat shock, ultraviolet radiation, bacterial endotoxin, and H2O2), cytokines (e.g., interleukin-1 (IL-I) and tumor necrosis factor α (TNF-α)), and growth factors (e.g., granulocyte macrophage-colony-stimulating factor (GM-CSF), and fibroblast growth factor (FGF)). An extracellular stimulus may affect one or more cellular responses related to cell growth, migration, differentiation, secretion of hormones, activation of transcription factors, muscle contraction, glucose metabolism, control of protein synthesis, and regulation of the cell cycle. [0005] Many diseases are associated with abnormal cellular responses triggered by protein kinase-mediated events as described above. These diseases include, but are not limited to, autoimmune diseases, inflammatory diseases, bone diseases, metabolic diseases, neurological and neurodegenerative diseases, cancer, cardiovascular diseases, allergies and asthma, Alzheimer's disease, and hormone-related diseases. Accordingly, there remains a need to find protein kinase inhibitors useful as therapeutic agents.
SUMMARY OF THE INVENTION
[0006] It has now been found that compounds of this invention, and pharmaceutically acceptable compositions thereof, are effective as inhibitors of one or more protein kinases. Such compounds have general formulae I-a and I-b:
Figure imgf000003_0001
I-a I-b or a pharmaceutically acceptable salt thereof, wherein Ring A, Ring B, m, p, Rx, Ry,
Figure imgf000003_0002
W2, and R1 are as defined herein.
[0007] Compounds of the present invention, and pharmaceutically acceptable compositions thereof, are useful for treating a variety of diseases, disorders or conditions, associated with abnormal cellular responses triggered by protein kinase-mediated events. Such diseases, disorders, or conditions include those described herein.
[0008] Compounds provided by this invention are also useful for the study of kinases in biological and pathological phenomena; the study of intracellular signal transduction pathways mediated by such kinases; and the comparative evaluation of new kinase inhibitors.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 depicts dose-response inhibition of phospho-plc gamma2 (p-plc gamma 2) with compound 1-2 in Ramos Cells; and the results of compound 1-2 in a "washout" experiment. Figure 2 depicts dose-response inhibition of p-plc gamma2 with compound 1-4 in Ramos Cells; and the results of compound 1-4 in a "washout" experiment. Figure 3 depicts dose response inhibition of p-plc gamma2 with compound 1-7 in Ramos cells; and the results of compound 1-7 in a "washout" experiment.
Figure 4 depicts dose response inhibition of p-plc gamma2 with compound 1-35 in Ramos cells.
Figure 5 depicts dose response inhibition of p-plc gamma2 with compound 1-38 in Ramos cells.
Figure 6 depicts MS analysis confirming covalent modification of TEC kinase at Cys449 by compound 1-2.
Figure 7 depicts MS analysis confirming covalent modification of TEC kinase at Cys449 by compound 1-4.
Figure 8 depicts MS analysis confirming covalent modification of TEC kinase at Cys449 by compound 1-7.
Figure 9 depicts the results of compound 1-2 in a "washout" experiment as compared to results of compound 1-4 and compound 1-7 in the same "washout" experiment in HCC827 cells containing EGFR deletion mutant.
Figure 10 depicts the results of compound 1-7 in a "washout" experiment as compared to results of an EGF control in A431 cells containing EGFR wild type.
Figure 11 depicts MS analysis confirming covalent modification of JAK-3 kinase at Cys909 by compound 1-7.
Figure 12 depicts dose-response inhibition of P-Stat5 with compound 1-2 in IL-2 stimulated
CTLL-2 cells; and dose-response inhibition of P-JAK-3 with compound 1-2 in IL-2 stimulated
CTLL-2 cells.
Figure 13 depicts dose-response inhibition of P-Stat5 with compound 1-4 in IL-2 stimulated
CTLL-2 cells; and dose-response inhibition of P-JAK-3 with compound 1-4 in IL-2 stimulated
CTLL-2 cells.
Figure 14 depicts dose-response inhibition of P-Stat5 with compound 1-7 in IL-2 stimulated
CTLL-2 cells.
Figure 15 depicts MS analysis confirming covalent modification of BTK by compound 1-7.
Figure 16 depicts a Western blot showing BTK protein available to the probe compound 1-215 after treating with varying amounts of 1-7.
Figure 17 depicts quantitation of the Western blot results in Figure 16.
Figure 18 depicts a Western blot for a washout experiment with compound 1-7 and probe compound 1-215. Figure 19 depicts quantitation of the Western blot results in Figure 18. Figure 20 depicts an amino acid sequence for BTK (SEQ ID 1). Figure 21 depicts an amino acid sequence for TEC (SEQ ID 2). Figure 22 depicts an amino acid sequence for ITK (SEQ ID 3). Figure 23 depicts an amino acid sequence for BMX (SEQ ID 4). Figure 24 depicts an amino acid sequence for TXK (SEQ ID 5). Figure 25 depicts an amino acid sequence for JAK3 (SEQ ID 6).
DETAILED DESCRIPTION OF CERTAIN EMBODIMENTS 1. General Description of Compounds of the Invention
[0009] In certain embodiments, the present invention provides a compound of formula I-a or I-b:
Figure imgf000005_0001
I-a I-b or a pharmaceutically acceptable salt thereof, wherein:
Ring A is an optionally substituted group selected from phenyl, a 3-7 membered saturated or partially unsaturated carbocyclic ring, an 8-10 membered bicyclic saturated, partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
Ring B is an optionally substituted group selected from phenyl, a 3-7 membered saturated or partially unsaturated carbocyclic ring, an 8-10 membered bicyclic saturated, partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur; R1 is a warhead group; Ry is hydrogen, halogen, -CN, -CF3, Ci_4 aliphatic, Ci_4 haloaliphatic, -OR, -C(O)R, or -
C(O)N(R)2; each R group is independently hydrogen or an optionally substituted group selected from Ci_6 aliphatic, phenyl, a 4-7 membered heterocylic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; W1 and W2 are each independently a covalent bond or a bivalent Ci_3 alkylene chain wherein one methylene unit of W1 or W2 is optionally replaced by -NR2-, -N(R2)C(O)-, -C(O)N(R2)-, -N(R2)SO2-, -SO2N(R2)-, -O-, -C(O)-, -OC(O)-, -C(O)O-, -S-, -SO- or -SO2-; R2 is hydrogen, optionally substituted Ci_6 aliphatic, or -C(O)R, or:
R2 and a substituent on Ring A are taken together with their intervening atoms to form a
4-6 membered saturated, partially unsaturated, or aromatic fused ring, or: R2 and Ry are taken together with their intervening atoms to form a 4-7 membered partially unsaturated or aromatic fused ring; m and p are independently 0-4; and
Rx and Rv are independently selected from -R, halogen, -OR, -0(CH2)q0R, -CN, -NO2, -SO2R, -SO2N(R)2, -SOR, -C(O)R, -CO2R, -C(O)N(R)2, -NRC(O)R, -NRC(O)NR2, -NRSO2R, or -N(R)2, wherein q is 1-4; or:
Rx and R1 when concurrently present on Ring B are taken together with their intervening atoms to form a 5-7 membered saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with a warhead group and 0-3 groups independently selected from oxo, halogen, -CN, or Ci_6 aliphatic; or Rv and R1 when concurrently present on Ring A are taken together with their intervening atoms to form a 5-7 membered saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with a warhead group and 0-3 groups independently selected from oxo, halogen, -CN, or Ci_6 aliphatic. 2. Compounds and Definitions
[0010] Compounds of this invention include those described generally above, and are further illustrated by the classes, subclasses, and species disclosed herein. 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, Handbook of Chemistry and Physics, 75th Ed. 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., Ed.: Smith, M.B. and March, J., John Wiley & Sons, New York: 2001, the entire contents of which are hereby incorporated by reference.
[0011] 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, or a monocyclic hydrocarbon or bicyclic hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic (also referred to herein as "carbocycle" "cycloaliphatic" or "cycloalkyl"), that has a single point of attachment to the rest of the molecule. Unless otherwise specified, aliphatic groups contain 1-6 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1-5 aliphatic carbon atoms. In other embodiments, aliphatic groups contain 1-4 aliphatic carbon atoms. In still other embodiments, aliphatic groups contain 1-3 aliphatic carbon atoms, and in yet other embodiments, aliphatic groups contain 1-2 aliphatic carbon atoms. In some embodiments, "cycloaliphatic" (or "carbocycle" or "cycloalkyl") refers to a monocyclic C3-C6 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. Suitable aliphatic groups include, but are not limited to, linear or branched, substituted or unsubstituted alkyl, alkenyl, alkynyl groups and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl or (cycloalkyl)alkenyl. [0012] The term "lower alkyl" refers to a Ci_4 straight or branched alkyl group. Exemplary lower alkyl groups are methyl, ethyl, propyl, isopropyl, butyl, isobutyl, and tert-butyl.
[0013] The term "lower haloalkyl" refers to a Ci_4 straight or branched alkyl group that is substituted with one or more halogen atoms.
[0014] The term "heteroatom" means one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon (including, any oxidized form of nitrogen, sulfur, phosphorus, or silicon; 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)).
[0015] The term "unsaturated", as used herein, means that a moiety has one or more units of unsaturation.
[0016] As used herein, the term "bivalent Ci_8 (or C1-6) saturated or unsaturated, straight or branched, hydrocarbon chain", refers to bivalent alkylene, alkenylene, and alkynylene chains that are straight or branched as defined herein.
[0017] The term "alkylene" refers to a bivalent alkyl group. An "alkylene chain" is a polymethylene group, i.e., -(CH2)n-, wherein n is a positive integer, preferably from 1 to 6, from
1 to 4, from 1 to 3, from 1 to 2, or from 2 to 3. A substituted alkylene chain is a polymethylene group in which one or more methylene hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group.
[0018] The term "alkenylene" refers to a bivalent alkenyl group. A substituted alkenylene chain is a polymethylene group containing at least one double bond in which one or more hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group.
[0019] As used herein, the term "cyclopropylenyl" refers to a bivalent cyclopropyl group of
the following structure:
Figure imgf000008_0001
[0020] The term "halogen" means F, Cl, Br, or I.
[0021] The term "aryl" used alone or as part of a larger moiety as in "aralkyl", "aralkoxy", or
"aryloxyalkyl", refers to monocyclic and bicyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic and wherein each ring in the system contains three to seven ring members. The term "aryl" may be used interchangeably with the term "aryl ring". In certain embodiments of the present invention, "aryl" refers to an aromatic ring system which includes, but not limited to, phenyl, biphenyl, naphthyl, anthracyl and the like, which may bear one or more substituents. Also included within the scope of the term "aryl", as it is used herein, is a group in which an aromatic ring is fused to one or more non-aromatic rings, such as indanyl, phthalimidyl, naphthimidyl, phenanthridinyl, or tetrahydronaphthyl, and the like.
[0022] The terms "heteroaryl" and "heteroar-", used alone or as part of a larger moiety, e.g., "heteroaralkyl", or "heteroaralkoxy", refer to groups having 5 to 10 ring atoms, preferably 5, 6, or 9 ring atoms; having 6, 10, or 14 π electrons shared in a cyclic array; and having, in addition to carbon atoms, from one to five heteroatoms. The term "heteroatom" refers to nitrogen, oxygen, or sulfur, and includes any oxidized form of nitrogen or sulfur, and any quaternized form of a basic nitrogen. Heteroaryl groups include, without limitation, thienyl, furanyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, purinyl, naphthyridinyl, and pteridinyl. The terms "heteroaryl" and "heteroar-", as used herein, also include groups in which a heteroaromatic ring is fused to one or more aryl, cycloaliphatic, or heterocyclyl rings, where the radical or point of attachment is on the heteroaromatic ring. Nonlimiting examples include indolyl, isoindolyl, benzothienyl, benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzthiazolyl, quinolyl, isoquinolyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4H-quinolizinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, and pyrido[2,3-b]-l,4-oxazin- 3(4Η)-one. A heteroaryl group may be mono- or bicyclic. The term "heteroaryl" may be used interchangeably with the terms "heteroaryl ring", "heteroaryl group", or "heteroaromatic", any of which terms include rings that are optionally substituted. The term "heteroaralkyl" refers to an alkyl group substituted by a heteroaryl, wherein the alkyl and heteroaryl portions independently are optionally substituted.
[0023] As used herein, the terms "heterocycle", "heterocyclyl", "heterocyclic radical", and "heterocyclic ring" are used interchangeably and refer to a stable 5- to 7-membered monocyclic or 7-10-membered bicyclic heterocyclic moiety that is either saturated or partially unsaturated, and having, in addition to carbon atoms, one or more, preferably one to four, heteroatoms, as defined above. When used in reference to a ring atom of a heterocycle, the term "nitrogen" includes a substituted nitrogen. As an example, in a saturated or partially unsaturated ring having 0-3 heteroatoms selected from oxygen, sulfur or nitrogen, the nitrogen may be N (as in 3,4- dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl), or +NR (as in TV-substituted pyrrolidinyl). [0024] A heterocyclic ring can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure and any of the ring atoms can be optionally substituted. Examples of such saturated or partially unsaturated heterocyclic radicals include, without limitation, tetrahydrofuranyl, tetrahydrothiophenyl pyrrolidinyl, piperidinyl, pyrrolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, diazepinyl, oxazepinyl, thiazepinyl, morpholinyl, and quinuclidinyl. The terms "heterocycle", "heterocyclyl", "heterocyclyl ring", "heterocyclic group", "heterocyclic moiety", and "heterocyclic radical", are used interchangeably herein, and also include groups in which a heterocyclyl ring is fused to one or more aryl, heteroaryl, or cycloaliphatic rings, such as indolinyl, 3H-indolyl, chromanyl, phenanthridinyl, or tetrahydroquinolinyl, where the radical or point of attachment is on the heterocyclyl ring. A heterocyclyl group may be mono- or bicyclic. The term "heterocyclylalkyl" refers to an alkyl group substituted by a heterocyclyl, wherein the alkyl and heterocyclyl portions independently are optionally substituted.
[0025] As used herein, the term "partially unsaturated" refers to a ring moiety that includes at least one double or triple bond. The term "partially unsaturated" is intended to encompass rings having multiple sites of unsaturation, but is not intended to include aryl or heteroaryl moieties, as herein defined.
[0026] As described herein, compounds of the invention may contain "optionally substituted" moieties. In general, the term "substituted", whether preceded by the term "optionally" or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent. Unless otherwise indicated, an "optionally substituted" group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position. 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, in certain embodiments, their recovery, purification, and use for one or more of the purposes disclosed herein.
[0027] Suitable monovalent substituents on a substitutable carbon atom of an "optionally substituted" group are independently halogen; -(CH2V4R0; -(CH2)0^OR°; -0(CH2V4R0, -O- (CH2V4C(O)OR0; -(CH2)0 4CH(OR°)2; -(CH2)0^SR°; -(CH2)0^Ph, which may be substituted with R°;
Figure imgf000011_0001
which may be substituted with R°; -CH=CHPh, which may be substituted with R°; -(CH2)0-4O(CH2)0-i-pyridyl which may be substituted with R°; -NO2; -CN; -N3; -(CH2)O 4N(R°)2; -(CH2)0 4N(R°)C(0)R°; -N(R°)C(S)R°; -(CH2)0 4N(R°)C(O)NR°2; -N(R°)C(S)NR°2; -(CH2)0 4N(R°)C(0)0R°; -N(R°)N(R°)C(O)R°; -N(R°)N(R°)C(0)NR°2; -N(R°)N(R°)C(0)0R°; -(CH2)0 4C(O)R°; -C(S)R0; -(CH2)0 4C(O)OR°; -(CH2)0 4C(O)SR°; -(CH2)o^C(0)OSiR°3; -(CH2)0 4OC(O)R°; -OC(O)(CH2)0 4SR°, SC(S)SR0; -(CH2)0 4SC(O)R°; -(CH2)o 4C(O)NR°2; -C(S)NR°2; -C(S)SR0; -SC(S)SR0, -(CH2)0 4OC(O)NR°2; -C(O)N(OR°)R°; -C(O)C(O)R0; -C(O)CH2C(O)R0; -C(N0R°)R°; -(CH2)0^SSR°; -(CH2)0 4S(O)2R°; -(CH2)O 4S(O)2OR0; -(CH2)0 4OS(O)2R°; -S(O)2NR°2; -(CH2)0 4S(O)R°; -N(R°)S(O)2NR°2; -N(R°)S(O)2R°; -N(0R°)R°; -C(NH)NR°2; -P(O)2R0; -P(O)R°2; -OP(O)R°2; -OP(O)(OR°)2; SiR°3; -(C1-4 straight or branched alkylene)O-N(R°)2; or -(C1-4 straight or branched alkylene)C(O)O-N(R°)2, wherein each R0 may be substituted as defined below and is independently hydrogen, Ci_6 aliphatic, -CH2Ph, -0(CH2ViPh, -CH2-(5-6 membered heteroaryl ring), or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R0, taken together with their intervening atom(s), form a 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, which may be substituted as defined below.
[0028] Suitable monovalent substituents on R0 (or the ring formed by taking two independent occurrences of R0 together with their intervening atoms), are independently halogen, -(CH2)0 2R*, -(haloR*), -(CH2V2OH, -(CH2)0 2OR*, -(CH2)0 2CH(OR*)2; -O(haloR'), -CN, -N3, -(CH2)0 2C(O)R*, -(CH2)0 2C(O)OH, -(CH2)0 2C(O)OR*, -(CH2)0 2SR*, -(CH2)o 2SH, -(CH2)o 2NH2, -(CH2)0 2NHR*, -(CH2)0 2NR*2, -NO2, -SiR*3, -OSiR*3, -C(O)SR*, -(C1-4 straight or branched alkylene)C(O)OR*, or -SSR* wherein each R* is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently selected from C1^t aliphatic, -CH2Ph, -0(CH2)o-iPh, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents on a saturated carbon atom of R0 include =0 and =S.
[0029] Suitable divalent substituents on a saturated carbon atom of an "optionally substituted" group include the following: =0, =S, =NNR* 2, =NNHC(O)R*, =NNHC(O)OR*, =NNHS(O)2R*, =NR*, =NOR*, -O(C(R* 2))2 3O-, or -S(C(R* 2))2_3S-, wherein each independent occurrence of R is selected from hydrogen, Ci_6 aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0- 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents that are bound to vicinal substitutable carbons of an "optionally substituted" group include: -O(CR 2)2_3O-, wherein each independent occurrence of R is selected from hydrogen, C i_6 aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[0030] Suitable substituents on the aliphatic group of R* include halogen, -R*, -(haloR*), -OH, -OR*, -O(haloR'), -CN, -C(O)OH, -C(O)OR*, -NH2, -NHR*, -NR*2, or -NO2, wherein each R* is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently Ci_4 aliphatic, -CH2Ph, -O(CH2)0 iPh, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[0031] Suitable substituents on a substitutable nitrogen of an "optionally substituted" group include
Figure imgf000012_0001
-S(O)2R1; -S(O)2NR1Z, -C(S)NR1Z, -C(NH)NR1Z, or -N(Rt)S(O)2Rt; wherein each Rf is independently hydrogen, Ci_6 aliphatic which may be substituted as defined below, unsubstituted -OPh, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of
Figure imgf000012_0002
taken together with their intervening atom(s) form an unsubstituted 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. [0032] Suitable substituents on the aliphatic group of R^ are independently halogen, -R*, -(haloR*), -OH, -OR*, -O(haloR'), -CN, -C(O)OH, -C(O)OR*, -NH2, -NHR*, -NR*2, or -NO2, wherein each R* is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently Ci_4 aliphatic, -CH2Ph, -0(CH2)o iPh, or a 5-6- membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[0033] 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, and are commensurate with a reasonable benefit/risk ratio. 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, 1977, 66, 1-19, 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, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like.
[0034] Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and
Figure imgf000013_0001
salts. 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, loweralkyl sulfonate and aryl sulfonate.
[0035] 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. 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 including 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, as probes in biological assays, or as therapeutic agents in accordance with the present invention. In some embodiments, the R1 group of formula I-a and I-b comprises one or more deuterium atoms.
[0036] As used herein, the term "irreversible" or "irreversible inhibitor" refers to an inhibitor (i.e. a compound) that is able to be covalently bonded to a target protein kinase in a substantially non-reversible manner. That is, whereas a reversible inhibitor is able to bind to (but is generally unable to form a covalent bond) the target protein kinase, and therefore can become dissociated from the target protein kinase, an irreversible inhibitor will remain substantially bound to the target protein kinase once covalent bond formation has occurred. Irreversible inhibitors usually display time dependency, whereby the degree of inhibition increases with the time with which the inhibitor is in contact with the enzyme. Methods for identifying if a compound is acting as an irreversible inhibitor are known to one of ordinary skill in the art. Such methods include, but are not limited to, enzyme kinetic analysis of the inhibition profile of the compound with the protein kinase target, the use of mass spectrometry of the protein drug target modified in the presence of the inhibitor compound, discontinuous exposure, also known as "washout," experiments, and the use of labeling, such as radiolabeled inhibitor, to show covalent modification of the enzyme, as well as other methods known to one of skill in the art. [0037] One of ordinary skill in the art will recognize that certain reactive functional groups can act as "warheads." As used herein, the term "warhead" or "warhead group" refers to a functional group present on a compound of the present invention wherein that functional group is capable of covalently binding to an amino acid residue (such as cysteine, lysine, histidine, or other residues capable of being covalently modified) present in the binding pocket of the target protein, thereby irreversibly inhibiting the protein. It will be appreciated that the -L-Y group, as defined and described herein, provides such warhead groups for covalently, and irreversibly, inhibiting the protein.
[0038] As used herein, the term "inhibitor" is defined as a compound that binds to and /or inhibits the target protein kinase with measurable affinity. In certain embodiments, an inhibitor has an IC50 and/or binding constant of less about 50 μM, less than about 1 μM, less than about 500 nM, less than about 100 nM, or less than about 10 nM.
[0039] The terms "measurable affinity" and "measurably inhibit," as used herein, means a measurable change in at least one of ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3 activity between a sample comprising a compound of the present invention, or composition thereof, and at least one of ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3, and an equivalent sample comprising at least one of ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3, in the absence of said compound, or composition thereof. 3. Description of Exemplary Compounds
[0040] According to one aspect, the present invention provides a compound of formula I-a or I-b,
Figure imgf000015_0001
I-a I-b or a pharmaceutically acceptable salt thereof, wherein:
Ring A is an optionally substituted group selected from phenyl, a 3-7 membered saturated or partially unsaturated carbocyclic ring, an 8-10 membered bicyclic saturated, partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
Ring B is an optionally substituted group selected from phenyl, a 3-7 membered saturated or partially unsaturated carbocyclic ring, an 8-10 membered bicyclic saturated, partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur; R1 is -L-Y, wherein:
L is a covalent bond or a bivalent Ci_8 saturated or unsaturated, straight or branched, hydrocarbon chain, wherein one, two, or three methylene units of L are optionally and independently replaced by cyclopropylene, -NR-, -N(R)C(O)-, -C(O)N(R)-, -N(R)SO2-, -SO2N(R)-, -O-, -C(O)-, -OC(O)-, -C(O)O-, -S-, -SO-, -SO2-, -C(=S)-, -C(=NR)-, -N=N-, or -CC=N2)-;
Y is hydrogen, Ci_6 aliphatic optionally substituted with oxo, halogen, or CN, or a 3-10 membered monocyclic or bicyclic, saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, and wherein said ring is substituted with at 1-4 groups independently selected from -Q-Z, oxo, NO2, halogen, CN, or Ci_6 aliphatic, wherein:
Q is a covalent bond or a bivalent Ci_6 saturated or unsaturated, straight or branched, hydrocarbon chain, wherein one or two methylene units of Q are optionally and independently replaced by -NR-, -S-, -O-, -C(O)-, -SO-, or -SO2-; and Z is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, or CN; Ry is hydrogen, halogen, -CN, -CF3, Ci_4 aliphatic, Ci_4 haloaliphatic, -OR, -C(O)R, or -C(O)N(R)2; each R group is independently hydrogen or an optionally substituted group selected from Ci_6 aliphatic, phenyl, a 4-7 membered heterocylic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; W1 and W2 are each independently a covalent bond or a bivalent Ci_3 alkylene chain wherein one methylene unit of W1 or W2 is optionally replaced by -NR2-, -N(R2)C(O)-, -C(O)N(R2)-, -N(R2)SO2-, -SO2N(R2)-, -0-, -C(O)-, -OC(O)-, -C(O)O-, -S-, -SO- or -SO2-; R2 is hydrogen, optionally substituted Ci_6 aliphatic, or -C(O)R, or:
R2 and a substituent on Ring A are taken together with their intervening atoms to form a 4-6 membered partially unsaturated or aromatic fused ring; or R2 and Ry are taken together with their intervening atoms to form a 4-6 membered saturated, partially unsaturated, or aromatic fused ring; m and p are independently 0-4; and
Rx and Rv are independently selected from -R, halogen, -OR, -0(CH2)q0R, -CN, -NO2, -SO2R, -SO2N(R)2, -SOR, -C(O)R, -CO2R, -C(O)N(R)2, -NRC(O)R, -NRC(O)NR2, -NRSO2R, or -N(R)2, or:
Rx and R1 when concurrently present on Ring B are taken together with their intervening atoms to form a 5-7 membered saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with a warhead group and 0-3 groups independently selected from oxo, halogen, -CN, or Ci_6 aliphatic; or
Rv and R1 when concurrently present on Ring A are taken together with their intervening atoms to form a 5-7 membered saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with a warhead group and 0-3 groups independently selected from oxo, halogen, -CN, or Ci_6 aliphatic.
[0041] As defined generally above, Ring A is an optionally substituted group selected from phenyl, a 3-7 membered saturated or partially unsaturated carbocyclic ring, an 8-10 membered bicyclic saturated, partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In certain embodiments, Ring A is an optionally substituted phenyl group. In some embodiments, Ring A is an optionally substituted naphthyl ring or a bicyclic 8-10 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In certain other embodiments, Ring A is an optionally substituted 3-7 membered carbocyclic ring. In yet other embodiments, Ring A is an optionally substituted 4-7 membered heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[0042] In certain embodiments, Ring A is substituted as defined herein. In some embodiments, Ring A is substituted with one, two, or three groups independently selected from halogen, R0, or -(CH2)O^tOR0, or -0(CH2)(MR0, wherein each R0 is as defined herein. Exemplary substituents on Ring A include Br, I, Cl, methyl, -CF3, -C≡CH, -OCH2phenyl, -OCH2(fluorophenyl), or -OCH2pyridyl. [0043] Exemplary Ring A groups are set forth in Table 1. Table 1. Exemplary Ring A Groups
Figure imgf000018_0001
Figure imgf000019_0001
Figure imgf000020_0001
Figure imgf000021_0001
wherein each R°, R+, and R1 is as defined above and described in classes and subclasses herein. [0044] In certain embodiments, Ring A is selected from i, ii, iv, v, vi, vii, ix, xiv, xvi, Hi, Ixiii,
Ixxi, Ixxiv, Ixxvi, Ixxviii, and taxi.
[0045] As defined generally above, Ring B is an optionally substituted group selected from phenyl, a 3-7 membered saturated or partially unsaturated carbocyclic ring, an 8-10 membered bicyclic saturated, partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In certain embodiments, Ring B is an optionally substituted phenyl group. In some embodiments, Ring B is an optionally substituted naphthyl ring or a bicyclic 8-10 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In certain other embodiments, Ring B is an optionally substituted 3-7 membered carbocyclic ring. In yet other embodiments, Ring B is an optionally substituted 4-7 membered heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[0046] In some embodiments, Ring B is phenyl. In some embodiments, Ring B is a 6- membered heteroaryl ring having 1-3 nitrogens. In some embodiments, Ring B is a 5-membered heteroaryl ring having 1 or 2 or 3 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[0047] In some embodiments, Ring B is a 5-6 membered saturated heterocyclic ring having 1 nitrogen. In some embodiments, Ring B is a 9-10 membered bicyclic partially saturated heteroaryl ring having 1-3 nitrogens. In some embodiments, Ring B is a 9-10 membered bicyclic partially saturated heteroaryl ring having 1 nitrogen. In some embodiments, Ring B is a 9-10 membered bicyclic partially saturated heteroaryl ring having 1 nitrogen and 1 oxygen.
[0048] In some embodiments, Ring B is an optionally substituted group selected from phenyl, pyridyl, pyrazinyl, pyrimidinyl, imidazolyl, pyrrolidinyl, piperdinyl, indolinyl, indazolyl, and isoindolinyl.
[0049] Exemplary Ring B groups are set forth in Table 4. Table 2. Ring B Groups
Figure imgf000023_0001
Figure imgf000024_0001
Figure imgf000025_0001
Figure imgf000026_0001
wherein each R1 and Rx is as defined above and described in classes and subclasses herein. [0050] In certain embodiments, Ring B is selected from i, H, Hi, iv, v, ix, x, xi, xiii, xvi, xvii, jcijc, jcjc, jcjcv, jcjcvi, JCJCJCH, JCJCJCiV, JCJCJCV, jcjcjcviii, JCHi, xlvi, xlviii, I, Iviii, Ixiv, Ixxviii, Ixxxiii, Ixxxvi, xciv, c, ci, cii, ciii, civ, and cv. [0051] In some embodiments, the m moiety of formula I is 1, 2, 3 or 4. In some embodiments, m is 1. In other embodiments, m is 0.
[0052] In some embodiments, the p moiety of formula I is 1, 2, 3 or 4. In some embodiments, p is 1. In other embodiments, p is 0.
[0053] As defined generally above, each Rx group of formula I is independently selected from -R, halogen, -OR, -O(CH2)qOR, -CN, -NO2, -SO2R, -SO2N(R)2, -SOR, -C(O)R, -CO2R, -
C(O)N(R)2, -NRC(O)R, -NRC(O)NR2, -NRSO2R, or -N(R)2, wherein q is 1-4, or Rx and R1 when concurrently present on Ring B are taken together with their intervening atoms to form a 5-
7 membered saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with a warhead group and 0-3 groups independently selected from oxo, halogen, CN, or Ci_6 aliphatic.
[0054] In some embodiments, each instance of Rx is independently selected from -R, -OR, -
O(CH2)qOR, or halogen. In certain embodiments, Rx is lower alkyl, lower alkoxy, lower alkoxyalkoxy, or halogen. Exemplary Rx groups include methyl, methoxy, methoxyethoxy and fluoro. In some embodiments, Rx is hydrogen.
[0055] As defined generally above, each Rv group of formula I is independently selected from -R, halogen, -OR, -O(CH2)qOR, -CN, -NO2, -SO2R, -SO2N(R)2, -SOR, -C(O)R, -CO2R, -
C(O)N(R)2, -NRC(O)R, -NRC(O)NR2, -NRSO2R, or -N(R)2, wherein q is 1-4, or Rv and R1 when concurrently present on Ring A are taken together with their intervening atoms to form a 5-
7 membered saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with a warhead group and 0-3 groups independently selected from oxo, halogen, CN, or Ci_6 aliphatic.
[0056] In some embodiments, each instance of Rv is independently selected from -R, -OR, -
O(CH2)qOR, or halogen. In certain embodiments, Rv is lower alkyl, lower alkoxy, lower alkoxyalkoxy, or halogen. Exemplary Rv groups include methyl, methoxy, trifluoromethyl, methoxyethoxy, and chloro. In some embodiments, Rv is hydrogen.
[0057] In some embodiments, the q moiety is 1, 2, 3, or 4. In certain embodiments, q is 1.
In certain other embodiments, q is 2.
[0058] As defined generally above, Ry is hydrogen, halogen, -CN, -CF3, Ci_4 aliphatic, Ci_4 haloaliphatic, -OR, -C(O)R, or -C(O)N(R)2, where R is as defined above and described herein.
In certain embodiments, Ry is hydrogen, halogen, -CN, -CF3, lower alkyl or lower haloalkyl, - C≡CR and cyclopropyl. In other embodiments, Ry is -OR, -C(O)R, or -C(O)N(R)2. In certain embodiments, Ry is -OCH3. In certain other embodiments, Ry is -C(O)CH3. In yet other embodiments, Ry is -C(O)NHR. In some embodiments, Ry is hydrogen. In certain embodiments, Ry is fluorine. In certain other embodiments, Ry is methyl.
[0059] As generally defined above, W1 and W2 are each independently a covalent bond or a bivalent Ci_3 alkylene chain wherein one methylene unit of W1 or W2 is optionally replaced by -NR2-, -N(R2)C(0)-, -C(O)N(R2)-, -N(R2)SO2-, -SO2N(R2)-, -0-, -C(O)-, -OC(O)-, -C(O)O-, -S-, -SO- or -SO2-. In certain embodiments, W1 and W2 are the same. In some embodiments, W1 and W2 are different.
[0060] In some embodiments, W1 is a covalent bond. In certain embodiments, W1 is a bivalent Ci_3 alkylene chain wherein one methylene unit of W1 is optionally replaced by -NR2-, -N(R2)C(0)-, -C(O)N(R2)-, -N(R2)SO2-, -SO2N(R2)-, -0-, -C(O)-, -OC(O)-, -C(O)O-, -S-, -SO- or -SO2-. In certain embodiments, W1 is -C(=0), -NR2-, -S-, or -0-. In some embodiments, W1 is -NR2-. In other embodiments, W1 is -0-. In certain embodiments, W1 is -NH-, -S-, or -0-. In some embodiments, W1 is -CH2O-, -CH2S-, or -CH2NH-. In some aspects, W1 is -OCH2-, -SCH2-, -NHCH2-, or -CH2CH2-.
[0061] In certain embodiments, W2 is a covalent bond. In some embodiments, W2 is a bivalent Ci_3 alkylene chain wherein one methylene unit of W2 is optionally replaced by -NR2-, -N(R2)C(0)-, -C(O)N(R2)-, -N(R2)SO2-, -SO2N(R2)-, -0-, -C(O)-, -OC(O)-, -C(O)O-, -S-, -SO- or -SO2-. In certain embodiments, W2 is -C(=0), -NR2-, -S-, or -0-. In some embodiments, W2 is -NR2-. In other embodiments, W2 is -0-. In certain embodiments, W2 is -NH-, -S-, or -0-. In some embodiments, W2 is -CH2O-, -CH2S-, or -CH2NH-. In some aspects, W2 is -OCH2-, -SCH2-, -NHCH2-, or -CH2CH2-.
[0062] In some embodiments, Ring B is phenyl, thus forming a compound of formula II-a or II-b:
Figure imgf000028_0001
II-a II-b or a pharmaceutically acceptable salt thereof, wherein each of Ring A, m, p, Rx, Ry, RV,W\ W2, and R1 is as defined above and described in classes and subclasses above and herein.
[0063] In certain embodiments, Ring A is phenyl, thus forming a compound of formula III-a or III-b:
Figure imgf000029_0001
or a pharmaceutically acceptable salt thereof, wherein each of Ring B, m, p, Rx, Ry, RV,W\ W2, and R1 is as defined above and described in classes and subclasses above and herein. [0064] In certain embodiments, Ring A is phenyl and Ring B is phenyl, thus forming a compound of formula IV-a or IV-b:
Figure imgf000029_0002
or a pharmaceutically acceptable salt thereof, wherein each of m, p, Rx, Ry, RV,W\ W2, and R1 is as defined above and described in classes and subclasses above and herein. [0065] As defined generally above, each R2 is independently hydrogen, optionally substituted Ci_6 aliphatic, or -C(O)R, or R2 and a substituent on Ring A are taken together with their intervening atoms to form a 4-6 membered partially unsaturated or aromatic fused ring, or R2 and Ry are taken together with their intervening atoms to form a 4-6 membered saturated, partially unsaturated, or aromatic fused ring. According to one aspect, R2 is hydrogen. According to another aspect, R2 is -C(O)R, wherein R is an optionally substituted Ci_6 aliphatic group. [0066] According to some aspects, R2 and a substituent on Ring A are taken together with their intervening atoms to form a 4-7 membered saturated or partially unsaturated ring, thus forming a compound of formula I-a-/ or I-b-/:
Figure imgf000030_0001
I-b-/ or a pharmaceutically acceptable salt thereof, wherein each of Ring A, R1, Rx, and m are as defined above and described in classes and subclasses above and herein.
[0067] Similar to the formation of compounds of formulae I-a-/ and I-b-/ above, it will be understood by one skilled in the art that compounds of formulae II-a, II-b, III-a, III-b, IV-a, and IV-b, will form corresponding compounds II-a-/, II-b-/, III-a-/, III-b-/, IV-a-/, and IV-b-/ when R2 and a substituent on Ring A are taken together with their intervening atoms to form a A-
7 membered saturated or partially unsaturated ring.
[0068] According to some aspects, R2 and Ry are taken together with their intervening atoms to form a 4-7 partially unsaturated ring, thus forming a compound of formula I-a-// or I-b-//:
Figure imgf000030_0002
I-b-// or a pharmaceutically acceptable salt thereof, wherein each of Ring A, R1, Rx, and m are as defined above and described in classes and subclasses above and herein.
[0069] Similar to the formation of compounds of formulae I-a-// and I-b-// above, it will be understood by one skilled in the art that compounds of formulae II-a, II-b, III-a, III-b, IV-a, and IV-b, will form corresponding compounds II-a-//, II-b-//, III-a-//, III-b-//, IV-a-//, and IV- b-// when R2 and Ry are taken together with their intervening atoms to form a 4-7 membered partially unsaturated ring. [0070] As defined generally above, the R1 group of formulae I and II is -L-Y, wherein:
L is a covalent bond or a bivalent Ci_g saturated or unsaturated, straight or branched, hydrocarbon chain, wherein one, two, or three methylene units of L are optionally and independently replaced by cyclopropylene, -NR-, -N(R)C(O)-, -C(O)N(R)-, -N(R)SO2-, -SO2N(R)-, -O-, -C(O)-, -OC(O)-, -C(O)O-, -S-, -SO-, -SO2-, -C(=S)-, -C(=NR)-, -N=N-, or -CC=N2)-;
Y is hydrogen, Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN, or a 3-10 membered monocyclic or bicyclic, saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, and wherein said ring is substituted with 1-4 Re groups; and each Re is independently selected from -Q-Z, oxo, NO2, halogen, CN, a suitable leaving group, or a Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN, wherein: Q is a covalent bond or a bivalent Ci_6 saturated or unsaturated, straight or branched, hydrocarbon chain, wherein one or two methylene units of Q are optionally and independently replaced by -N(R)-, -S-, -0-, -C(O)-, -OC(O)-, -C(O)O-, -SO-, or -SO2-, -N(R)C(O)-, -C(O)N(R)-, -N(R)SO2-, Or -SO2N(R)-; and Z is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN. [0071] In certain embodiments, L is a covalent bond.
[0072] In certain embodiments, L is a bivalent Ci_g saturated or unsaturated, straight or branched, hydrocarbon chain. In certain embodiments, L is -CH2-.
[0073] In certain embodiments, L is a covalent bond, -CH2-, -NH-, -CH2NH-, -NHCH2-, -NHC(O)-, -NHC(O)CH2OC(O)-, -CH2NHC(O)-, -NHSO2-, -NHSO2CH2-,
-NHC(O)CH2OC(O)-, or -SO2NH-.
[0074] In some embodiments, L is a bivalent C2_8 straight or branched, hydrocarbon chain wherein L has at least one double bond and one or two additional methylene units of L are optionally and independently replaced by -NRC(O)-, -C(O)NR-, -N(R)SO2-, -SO2N(R)-, -S-, -S(O)-, -SO2-, -OC(O)-, -C(O)O-, cyclopropylene, -0-, -N(R)-, or -C(O)-. [0075] In certain embodiments, L is a bivalent C2_8 straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by -C(O)-, -NRC(O)-, -C(O)NR-, -N(R)SO2-, -SO2N(R)-, -S-, -S(O)-, -SO2-, -OC(O)-, or -C(O)O-, and one or two additional methylene units of L are optionally and independently replaced by cyclopropylene, -O-, -N(R)-, or -C(O)-.
[0076] In some embodiments, L is a bivalent C2-8 straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by
-C(O)-, and one or two additional methylene units of L are optionally and independently replaced by cyclopropylene, -O-, -N(R)-, or -C(O)-.
[0077] As described above, in certain embodiments, L is a bivalent C2-8 straight or branched, hydrocarbon chain wherein L has at least one double bond. One of ordinary skill in the art will recognize that such a double bond may exist within the hydrocarbon chain backbone or may be
"exo" to the backbone chain and thus forming an alkylidene group. By way of example, such an
L group having an alkylidene branched chain includes -CH2C(=CH2)CH2-. Thus, in some embodiments, L is a bivalent C2-8 straight or branched, hydrocarbon chain wherein L has at least one alkylidenyl double bond. Exemplary L groups include -NHC(O)C(=CH2)CH2-.
[0078] In certain embodiments, L is a bivalent C2-8 straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by
-C(O)-. In certain embodiments, L is -C(O)CH=CH(CH3)-, -C(O)CH=CHCH2NH(CH3)-,
-C(O)CH=CH(CH3)-, -C(O)CH=CH-, -CH2C(O)CH=CH-, -CH2C(O)CH=CH(CH3)-,
-CH2CH2C(O)CH=CH-, -CH2CH2C(O)CH=CHCH2-, -CH2CH2C(O)CH=CHCH2NH(CH3)-, or
-CH2CH2C(O)CH=CH(CH3)-, or -CH(CH3)OC(O)CH=CH-.
[0079] In certain embodiments, L is a bivalent C2_8 straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by
-OC(O)-.
[0080] In some embodiments, L is a bivalent C2_s straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by
-NRC(O)-, -C(O)NR-, -N(R)SO2-, -SO2N(R)-, -S-, -S(O)-, -SO2-, -OC(O)-, Or -C(O)O-, and one or two additional methylene units of L are optionally and independently replaced by cyclopropylene, -0-, -N(R)-, or -C(O)-. In some embodiments, L is -CH2OC(O)CH=CHCH2-,
-CH2-OC(O)CH=CH-, or -CH(CH=CH2)OC(O)CH=CH-.
[0081] In certain embodiments, L is -NRC(O)CH=CH-, -NRC(O)CH=CHCH2N(CH3)-,
-NRC(O)CH=CHCH2O-, -CH2NRC(O)CH=CH-, -NRSO2CH=CH-, -NRSO2CH=CHCH2-,
-NRC(O)(C=N2)C(O)-, -NRC(O)CH=CHCH2N(CH3)-, -NRSO2CH=CH-, -NRSO2CH=CHCH2-, -NRC(O)CH=CHCH2O-, -NRC(O)C(=CH2)CH2-, -CH2NRC(O)-, -CH2NRC(O)CH=CH-,
-CH2CH2NRC(O)-, or -CH2NRC(O)cyclopropylene-, wherein each R is independently hydrogen or optionally substituted Ci_6 aliphatic.
[0082] In certain embodiments, L is -NHC(O)CH=CH-, -NHC(O)CH=CHCH2N(CH3)-,
-NHC(O)CH=CHCH2O-, -CH2NHC(O)CH=CH-, -NHSO2CH=CH-, -NHSO2CH=CHCH2-,
-NHC(O)(C=N2)C(O)-, -NHC(O)CH=CHCH2N(CH3)-, -NHSO2CH=CH-, -NHSO2CH=CHCH2-
, -NHC(O)CH=CHCH2O-, -NHC(O)C(=CH2)CH2-, -CH2NHC(O)-, -CH2NHC(O)CH=CH-,
-CH2CH2NHC(O)-, or -CH2NHC(O)cyclopropylene-.
[0083] In some embodiments, L is a bivalent C2_8 straight or branched, hydrocarbon chain wherein L has at least one triple bond. In certain embodiments, L is a bivalent C2_g straight or branched, hydrocarbon chain wherein L has at least one triple bond and one or two additional methylene units of L are optionally and independently replaced by -NRC(O)-, -C(O)NR-, -S-,
-S(O)-, -SO2-, -C(=S)-, -C(=NR)-, -0-, -N(R)-, or -C(O)-. In some embodiments, L has at least one triple bond and at least one methylene unit of L is replaced by -N(R)-, -N(R)C(O)-, -C(O)-,
-C(O)O-, or -OC(O)-, or -0-.
[0084] Exemplary L groups include -C≡C-, -C≡CCH2N(isopropyl)-, -NHC(O)C=CCH2CH2-,
-CH2-C=C-CH2-, -C=CCH2O-, -CH2C(O)C=C-, -C(O)C=C-, or -CH2OC(=O)C≡C-.
[0085] In certain embodiments, L is a bivalent C2_g straight or branched, hydrocarbon chain wherein one methylene unit of L is replaced by cyclopropylene and one or two additional methylene units of L are independently replaced by -C(O)-, -NRC(O)-, -C(O)NR-, -N(R)SO2-, or -SO2N(R)-. Exemplary L groups include -NHC(O)-cyclopropylene-SO2- and -NHC(O)- cyclopropylene-.
[0086] As defined generally above, Y is hydrogen, Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN, or a 3-10 membered monocyclic or bicyclic, saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, and wherein said ring is substituted with at 1-4 Re groups, each Re is independently selected from -Q-Z, oxo, NO2, halogen, CN, a suitable leaving group, or Ci_6 aliphatic, wherein
Q is a covalent bond or a bivalent Ci_6 saturated or unsaturated, straight or branched, hydrocarbon chain, wherein one or two methylene units of Q are optionally and independently replaced by -N(R)-, -S-, -0-, -C(O)-, -OC(O)-, -C(O)O-, -SO-, or -SO2-, -N(R)C(O)-, - C(O)N(R)-, -N(R)SO2-, or -SO2N(R)-; and, Z is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN. [0087] In certain embodiments, Y is hydrogen.
[0088] In certain embodiments, Y is Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN. In some embodiments, Y is C2_6 alkenyl optionally substituted with oxo, halogen, NO2, or CN. In other embodiments, Y is C2_6 alkynyl optionally substituted with oxo, halogen, NO2, or CN. In some embodiments, Y is C2_6 alkenyl. In other embodiments, Y is C2_4 alkynyl. [0089] In other embodiments, Y is Ci_6 alkyl substituted with oxo, halogen, NO2, or CN. Such Y groups include -CH2F, -CH2Cl, -CH2CN, and -CH2NO2.
[0090] In certain embodiments, Y is a saturated 3-6 membered monocyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein Y is substituted with 1 -4 Re groups, wherein each Re is as defined above and described herein. [0091] In some embodiments, Y is a saturated 3-4 membered heterocyclic ring having 1 heteroatom selected from oxygen or nitrogen wherein said ring is substituted with 1-2 Re groups, wherein each Re is as defined above and described herein. Exemplary such rings are epoxide and oxetane rings, wherein each ring is substituted with 1-2 Re groups, wherein each Re is as defined above and described herein.
[0092] In other embodiments, Y is a saturated 5-6 membered heterocyclic ring having 1-2 heteroatom selected from oxygen or nitrogen wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein. Such rings include piperidine and pyrrolidine, wherein each ring is substituted with 1-4 Re groups, wherein each Re is as defined
above and described herein. In certain embodiments, Y is
Figure imgf000034_0001
"2 , or
Figure imgf000034_0002
, wherein each R, Q, Z, and Re is as defined above and described herein. [0093] In some embodiments, Y is a saturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein. In certain embodiments, Y is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl, wherein each ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein.. In certain embodiments, Y is , wherein Re is as defined above and described herein.
Figure imgf000035_0004
In certain embodiments, Y is cyclopropyl optionally substituted with halogen, CN or NO2. [0094] In certain embodiments, Y is a partially unsaturated 3-6 membered monocyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein. [0095] In some embodiments, Y is a partially unsaturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein. In some embodiments, Y is cyclopropenyl, cyclobutenyl, cyclopentenyl, or cyclohexenyl wherein each ring is substituted with 1-4 Re groups, wherein each Re is as defined
above and described herein. In certain embodiments, Y is
Figure imgf000035_0001
** 1"2 , wherein each Re is as defined above and described herein.
[0096] In certain embodiments, Y is a partially unsaturated 4-6 membered heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein. In certain embodiments, Y is selected from:
Figure imgf000035_0002
wherein each R and Re is as defined above and described herein.
[0097] In certain embodiments, Y is a 6-membered aromatic ring having 0-2 nitrogens wherein said ring is substituted with 1-4 Re groups, wherein each Re group is as defined above and described herein. In certain embodiments, Y is phenyl, pyridyl, or pyrimidinyl, wherein each ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein.
[0098] In some embodiments, Y is selected from:
Figure imgf000035_0003
wherein each Re is as defined above and described herein.
[0099] In other embodiments, Y is a 5-membered heteroaryl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-3 Re groups, wherein each Re group is as defined above and described herein. In some embodiments, Y is a 5 membered partially unsaturated or aryl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, wherein said ring is substituted with 1- 4 Re groups, wherein each Re group is as defined above and described herein. Exemplary such rings are isoxazolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, pyrrolyl, furanyl, thienyl, triazole, thiadiazole, and oxadiazole, wherein each ring is substituted with 1-3 Re groups, wherein each Re group is as defined above and described herein. In certain embodiments, Y is selected from:
Figure imgf000036_0001
wherein each R and Re is as defined above and described herein.
[00100] In certain embodiments, Y is an 8-10 membered bicyclic, saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups, wherein Re is as defined above and described herein. According to another aspect, Y is a 9-10 membered bicyclic, partially unsaturated, or aryl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups, wherein Re is as defined above and described herein. Exemplary such bicyclic rings include 2,3-dihydrobenzo[d]isothiazole, wherein said ring is substituted with 1-4 Re groups, wherein Re is as defined above and described herein. [00101] As defined generally above, each Re group is independently selected from -Q-Z, oxo, NO2, halogen, CN, a suitable leaving group, or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN, wherein Q is a covalent bond or a bivalent Ci_6 saturated or unsaturated, straight or branched, hydrocarbon chain, wherein one or two methylene units of Q are optionally and independently replaced by -N(R)-, -S-, -O-, -C(O)-, -OC(O)-, -C(O)O-, -SO-, or -SO2-, - N(R)C(O)-, -C(O)N(R)-, -N(R)SO2-, or -SO2N(R)-; and Z is hydrogen or C1-6 aliphatic optionally substituted with oxo, halogen, NO2, or CN.
[00102] In certain embodiments, Re is Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN. In other embodiments, Re is oxo, NO2, halogen, or CN.
[00103] In some embodiments, Re is -Q-Z, wherein Q is a covalent bond and Z is hydrogen (i.e., Re is hydrogen). In other embodiments, Re is -Q-Z, wherein Q is a bivalent Ci_6 saturated or unsaturated, straight or branched, hydrocarbon chain, wherein one or two methylene units of Q are optionally and independently replaced by -NR-, -NRC(O)-, -C(O)NR-, -S-, -O-, -C(O)-, -SO-, or -SO2-. In other embodiments, Q is a bivalent C2_6 straight or branched, hydrocarbon chain having at least one double bond, wherein one or two methylene units of Q are optionally and independently replaced by -NR-, -NRC(O)-, -C(O)NR-, -S-, -0-, -C(O)-, -SO-, or -SO2-. In certain embodiments, the Z moiety of the Re group is hydrogen. In some embodiments, -Q-Z is -NHC(O)CH=CH2 or -C(O)CH=CH2.
[00104] In certain embodiments, each Re is independently selected from from oxo, NO2, CN, fluoro, chloro, -NHC(O)CH=CH2, -C(O)CH=CH2, -CH2CH=CH2, -C≡CH, -C(O)OCH2Cl, -C(O)OCH2F, -C(O)OCH2CN, -C(O)CH2Cl, -C(O)CH2F, -C(O)CH2CN, Or -CH2C(O)CH3. [00105] In certain embodiments, Re is a suitable leaving group, ie a group that is subject to nucleophilic displacement. A "suitable leaving" is a chemical group that is readily displaced by a desired incoming chemical moiety such as the thiol moiety of a cysteine of interest. Suitable leaving groups are well known in the art, e.g., see, "Advanced Organic Chemistry," Jerry March, 5th Ed., pp. 351-357, John Wiley and Sons, N.Y. Such leaving groups include, but are not limited to, halogen, alkoxy, sulphonyloxy, optionally substituted alkylsulphonyloxy, optionally substituted alkenylsulfonyloxy, optionally substituted arylsulfonyloxy, acyl, and diazonium moieties. Examples of suitable leaving groups include chloro, iodo, bromo, fluoro, acetoxy, methanesulfonyloxy (mesyloxy), tosyloxy, triflyloxy, nitro-phenylsulfonyloxy (nosyloxy), and bromo-phenylsulfonyloxy (brosyloxy). [00106] In certain embodiments, the following embodiments and combinations of -L-Y apply:
(a) L is a bivalent C2-8 straight or branched, hydrocarbon chain wherein L has at least one double bond and one or two additional methylene units of L are optionally and independently replaced by -NRC(O)-, -C(O)NR-, -N(R)SO2-, -SO2N(R)-, -S-, -S(O)-, -SO2-, -OC(O)-, -C(O)O-, cyclopropylene, -0-, -N(R)-, or -C(O)- ; and Y is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN; or
(b) L is a bivalent C2-8 straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by -C(O)-, -NRC(O)-, -C(O)NR-, -N(R)SO2-, -SO2N(R)-, -S-, -S(O)-, -SO2-, -OC(O)-, or -C(O)O-, and one or two additional methylene units of L are optionally and independently replaced by cyclopropylene, -0-, -N(R)-, or -C(O)-; and Y is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN; or
(c) L is a bivalent C2_8 straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by -C(O)-, and one or two additional methylene units of L are optionally and independently replaced by cyclopropylene, -0-, -N(R)-, or -C(O)-; and Y is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN; or
(d) L is a bivalent C2_8 straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by -C(O)-; and Y is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN; or
(e) L is a bivalent C2-8 straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by -OC(O)-; and Y is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN; or
(f) L is -NRC(O)CH=CH-, -NRC(O)CH=CHCH2N(CH3)-, -NRC(O)CH=CHCH2O-, -CH2NRC(O)CH=CH-, -NRSO2CH=CH-, -NRSO2CH=CHCH2-, -NRC(O)(C=N2)-, -NRC(O)(C=N2)C(O)-, -NRC(O)CH=CHCH2N(CH3)-, -NRSO2CH=CH-, -NRSO2CH=CHCH2-, -NRC(O)CH=CHCH2O-, -NRC(O)Q=CH2)CH2-, -CH2NRC(O)-, -CH2NRC(O)CH=CH-, -CH2CH2NRC(O)-, or -CH2NRC(O)cyclopropylene-; wherein R is H or optionally substituted Ci_6 aliphatic; and Y is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN; or (g) L is -NHC(O)CH=CH-, -NHC(O)CH=CHCH2N(CH3)-, -NHC(O)CH=CHCH2O-, -CH2NHC(O)CH=CH-, -NHSO2CH=CH-, -NHSO2CH=CHCH2-, -NHC(O)(C=N2)-, -NHC(O)(C=N2)C(O)-, -NHC(O)CH=CHCH2N(CH3)-, -NHSO2CH=CH-,
-NHSO2CH=CHCH2-, -NHC(O)CH=CHCH2O-, -NHC(O)C(=CH2)CH2-, -CH2NHC(O)-, -CH2NHC(O)CH=CH-, -CH2CH2NHC(O)-, or -CH2NHC(O)cyclopropylene-; and Y is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN; or
(h) L is a bivalent C2_8 straight or branched, hydrocarbon chain wherein L has at least one alkylidenyl double bond and at least one methylene unit of L is replaced by -C(O)-, -NRC(O)-, -C(O)NR-, -N(R)SO2-, -SO2N(R)-, -S-, -S(O)-, -SO2-, -OC(O)-, or -C(O)O-, and one or two additional methylene units of L are optionally and independently replaced by cyclopropylene, -0-, -N(R)-, or -C(O)-; and Y is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN; or
(i) L is a bivalent C2_8 straight or branched, hydrocarbon chain wherein L has at least one triple bond and one or two additional methylene units of L are optionally and independently replaced by -NRC(O)-, -C(O)NR-, -N(R)SO2-, -SO2N(R)-, -S-, -S(O)-, -SO2-, -OC(O)-, or -C(O)O-, and Y is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN; or
O) L is -C≡C-, -C≡CCH2N(isopropyl)-, -NHC(O)C=CCH2CH2-, -CH2-C=C-CH2-, -C=CCH2O-, -CH2C(O)C=C-, -C(O)C=C-, or -CH2OC(=O)C≡C-; and Y is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN; or
(k) L is a bivalent C2_8 straight or branched, hydrocarbon chain wherein one methylene unit of L is replaced by cyclopropylene and one or two additional methylene units of L are independently replaced by -NRC(O)-, -C(O)NR-, -N(R)SO2-, -SO2N(R)-, -S-, -S(O)-, -SO2-, -OC(O)-, or -C(O)O-; and Y is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN; or
(1) L is a covalent bond and Y is selected from:
(i) Ci_6 alkyl substituted with oxo, halogen, NO2, or CN;
(Ji) C2_6 alkenyl optionally substituted with oxo, halogen, NO2, or CN; or
(Hi) C2_6 alkynyl optionally substituted with oxo, halogen, NO2, or CN; or (Jv) a saturated 3-4 membered heterocyclic ring having 1 heteroatom selected from oxygen or nitrogen wherein said ring is substituted with 1-2 Re groups, wherein each
Re is as defined above and described herein; or (v) a saturated 5-6 membered heterocyclic ring having 1-2 heteroatom selected from oxygen or nitrogen wherein said ring is substituted with 1-4 Re groups, wherein each
Re is as defined above and described herein; or
(vz)
Figure imgf000040_0001
"2 , wherein each R, Q, Z, and Re is as defined above and described herein; or (vzϊ) a saturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4
Re groups, wherein each Re is as defined above and described herein; or {viiϊ) a partially unsaturated 3-6 membered monocyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or (Jx) a partially unsaturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or
(x)
Figure imgf000040_0002
, wherein each Re is as defined above and described herein; or
(xi) a partially unsaturated 4-6 membered heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or
Figure imgf000040_0003
wherein each R and Re is as defined above and described herein; or (xiii) a 6-membered aromatic ring having 0-2 nitrogens wherein said ring is substituted with 1-4 Re groups, wherein each Re group is as defined above and described herein; or N N
(xiv)
Figure imgf000041_0002
wherein each Re is as defined above and described herein; or
(xv) a 5-membered heteroaryl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-3 Re groups, wherein each Re group is as defined above and described herein; or
Figure imgf000041_0001
wherein each R and Re is as defined above and described herein; or (pcvii) an 8-10 membered bicyclic, saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups, wherein Re is as defined above and described herein; (m) L is -C(O)- and Y is selected from:
(J) Ci_6 alkyl substituted with oxo, halogen, NO2, or CN; or (Ji) C2-6 alkenyl optionally substituted with oxo, halogen, NO2, or CN; or (Hi) C2-6 alkynyl optionally substituted with oxo, halogen, NO2, or CN; or (Jv) a saturated 3-4 membered heterocyclic ring having 1 heteroatom selected from oxygen or nitrogen wherein said ring is substituted with 1-2 Re groups, wherein each Re is as defined above and described herein; or (v) a saturated 5-6 membered heterocyclic ring having 1-2 heteroatom selected from oxygen or nitrogen wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or
(vO
Figure imgf000042_0001
wherein each R, Q, Z, and Re is as defined above and described herein; or (vii) a saturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4
Re groups, wherein each Re is as defined above and described herein; or (viii) a partially unsaturated 3-6 membered monocyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or (ix) a partially unsaturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or
(x)
Figure imgf000042_0002
, wherein each Re is as defined above and described herein; or
(xz) a partially unsaturated 4-6 membered heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or
1,
(JCH)
Figure imgf000042_0003
wherein each R and Re is as defined above and described herein; or
(xiii) a 6-membered aromatic ring having 0-2 nitrogens wherein said ring is substituted with 1-4 Re groups, wherein each Re group is as defined above and described herein; or m
Figure imgf000043_0002
wherein each Re is as defined above and described herein; or
(xv) a 5-membered heteroaryl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-3 Re groups, wherein each Re group is as defined above and described herein; or
(xvi)
Figure imgf000043_0001
wherein each R and Re is as defined above and described herein; or (pcvii) an 8-10 membered bicyclic, saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups, wherein Re is as defined above and described herein; (n) L is -N(R)C(O)- and Y is selected from:
(i) Ci_6 alkyl substituted with oxo, halogen, NO2, or CN; or
(U) C2-6 alkenyl optionally substituted with oxo, halogen, NO2, or CN; or
(in) C2_6 alkynyl optionally substituted with oxo, halogen, NO2, or CN; or
(Jv) a saturated 3-4 membered heterocyclic ring having 1 heteroatom selected from oxygen or nitrogen wherein said ring is substituted with 1-2 Re groups, wherein each
Re is as defined above and described herein; or (v) a saturated 5-6 membered heterocyclic ring having 1-2 heteroatom selected from oxygen or nitrogen wherein said ring is substituted with 1-4 Re groups, wherein each
Re is as defined above and described herein; or (vi) wherein each R, Q, Z, and Re is as
Figure imgf000044_0001
defined above and described herein; or (vii) a saturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4
Re groups, wherein each Re is as defined above and described herein; or (viii) a partially unsaturated 3-6 membered monocyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or (ix) a partially unsaturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or
(x)
Figure imgf000044_0003
, wherein each Re is as defined above and described herein; or
(xi) a partially unsaturated 4-6 membered heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or
(xii)
Figure imgf000044_0004
wherein each R and Re is as defined above and described herein; or
(xiii) a 6-membered aromatic ring having 0-2 nitrogens wherein said ring is substituted with 1-4 Re groups, wherein each Re group is as defined above and described herein; or
,,,
Figure imgf000044_0002
wherein each Re is as defined above and described herein; or (xv) a 5-membered heteroaryl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-3 Re groups, wherein each Re group is as defined above and described herein; or
Figure imgf000045_0001
wherein each R and Re is as defined above and described herein; or (xvii) an 8-10 membered bicyclic, saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups, wherein Re is as defined above and described herein; L is a bivalent Ci_g saturated or unsaturated, straight or branched, hydrocarbon chain; and Y is selected from:
(i) Ci_6 alkyl substituted with oxo, halogen, NO2, or CN; (U) C2-6 alkenyl optionally substituted with oxo, halogen, NO2, or CN; or (in) C2-6 alkynyl optionally substituted with oxo, halogen, NO2, or CN; or (Jv) a saturated 3-4 membered heterocyclic ring having 1 heteroatom selected from oxygen or nitrogen wherein said ring is substituted with 1-2 Re groups, wherein each
Re is as defined above and described herein; or (v) a saturated 5-6 membered heterocyclic ring having 1-2 heteroatom selected from oxygen or nitrogen wherein said ring is substituted with 1-4 Re groups, wherein each
Re is as defined above and described herein; or (vi)
Figure imgf000046_0001
wherein each R, Q, Z, and Re is as defined above and described herein; or (vii) a saturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4
Re groups, wherein each Re is as defined above and described herein; or (viii) a partially unsaturated 3-6 membered monocyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or (ix) a partially unsaturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or
(x) , wherein each Re is as defined above and described herein; or
Figure imgf000046_0003
(xi) a partially unsaturated 4-6 membered heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or
(xii)
Figure imgf000046_0004
wherein each R and Re is as defined above and described herein; or
(xiii) a 6-membered aromatic ring having 0-2 nitrogens wherein said ring is substituted with 1-4 Re groups, wherein each Re group is as defined above and described herein; or
,,,
Figure imgf000046_0002
wherein each Re is as defined above and described herein; or (xv) a 5-membered heteroaryl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-3 Re groups, wherein each Re group is as defined above and described herein; or
Figure imgf000047_0001
wherein each R and Re is as defined above and described herein; or (xvii) an 8-10 membered bicyclic, saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups, wherein Re is as defined above and described herein;
(p) L is a covalent bond, -CH2-, -NH-, -C(O)-, -CH2NH-, -NHCH2-, -NHC(O)-, -NHC(O)CH2OC(O)-, -CH2NHC(O)-, -NHSO2-, -NHSO2CH2-, -NHC(O)CH2OC(O)-, or -SO2NH-; and Y is selected from:
(i) Ci_6 alkyl substituted with oxo, halogen, NO2, or CN; or (U) C2_6 alkenyl optionally substituted with oxo, halogen, NO2, or CN; or (Hi) C2_6 alkynyl optionally substituted with oxo, halogen, NO2, or CN; or (Jv) a saturated 3-4 membered heterocyclic ring having 1 heteroatom selected from oxygen or nitrogen wherein said ring is substituted with 1-2 Re groups, wherein each
Re is as defined above and described herein; or (v) a saturated 5-6 membered heterocyclic ring having 1-2 heteroatom selected from oxygen or nitrogen wherein said ring is substituted with 1-4 Re groups, wherein each
Re is as defined above and described herein; or (vi) wherein each R, Q, Z, and Re is as
Figure imgf000048_0001
defined above and described herein; or (vii) a saturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4
Re groups, wherein each Re is as defined above and described herein; or (viii) a partially unsaturated 3-6 membered monocyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or (ix) a partially unsaturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or
(x)
Figure imgf000048_0003
, wherein each Re is as defined above and described herein; or
(xi) a partially unsaturated 4-6 membered heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or
(jci'O
Figure imgf000048_0004
wherein each R and Re is as defined above and described herein; or
(xiii) a 6-membered aromatic ring having 0-2 nitrogens wherein said ring is substituted with 1-4 Re groups, wherein each Re group is as defined above and described herein; or
,,,
Figure imgf000048_0002
wherein each Re is as defined above and described herein; or (xv) a 5-membered heteroaryl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-3 Re groups, wherein each Re group is as defined above and described herein; or
Figure imgf000049_0001
wherein each R and Re is as defined above and described herein; or (pcvii) an 8-10 membered bicyclic, saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups, wherein Re is as defined above and described herein.
[00107] In certain embodiments, the Y group of formula Ia or Ib is selected from those set forth in Table 3, below, wherein each wavy line indicates the point of attachment to the rest of the molecule.
Figure imgf000049_0002
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
w eren eac s n epen en y a su a e eavng group, 2, , or oxo. [00108] In certain embodiments, R1 is -C≡CH, -C≡CCH2NH(isopropyl), -NHC(O)C≡CCH2CH3, -CH2-C≡C-CH3, -C≡CCH2OH, -CH2C(O)C≡CH, -C(O)C≡CH, or -CH2OC(=O)C≡CH. In some embodiments, R1 is selected from -NHC(O)CH=CH2, -NHC(O)CH=CHCH2N(CHS)2, or -CH2NHC(O)CH=CH2.
[00109] In certain embodiments, R1 is selected from those set forth in Table 4, below, wherein each wavy line indicates the point of attachment to the rest of the molecule.
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
# uuuuuu vvvvvv wwwwww or raxα
Figure imgf000057_0001
wherein each Re is independently a suitable leaving group, NO2, CN, or oxo. [00110] As defined generally above, R1 is a warhead group, or, when R1 and Rx form a ring, then -Q-Z is a warhead group. Without wishing to be bound by any particular theory, it is believed that such R1 groups, i.e. warhead groups, are particularly suitable for covalently binding to a key cysteine residue in the binding domain of certain protein kinases. Protein kinases having a cysteine residue in the binding domain are known to one of ordinary skill in the art and include ErbBl, ErbB2, and ErbB4, or a mutant thereof. In certain embodiments, compounds of the present invention have a warhead group characterized in that inventive compounds target one or more of the following cysteine residues:
ERBBl ITQLMPFGCLLDYVREH
ERBB2 VTQLMPYGCLLDHVREN
ERBB4 VTQLMPHGCLLEYVHEH
[00111] Thus, in some embodiments, R1 is characterized in that the -L-Y moiety is capable of covalently binding to a cysteine residue thereby irreversibly inhibiting the enzyme. In certain embodiments, the cysteine residue is Cys797 of ErbBl, Cys805 of ErbB2 and Cys803 of ErbB4, or a mutant thereof, where the provided residue numbering is in accordance with Uniprot (code POO533 for ErbBl; code PO4626 for ErbB2, and Q 15303 for ErbB4). It will be understood that the Cys of ErbBl (EGFR) is variably called 773 or 797 depending on whether the parent sequence contains the signal peptide or not. Thus, in accordance with the present invention, the relevant cysteine residue of ErbBl may be described as Cys 773 or Cys 797 and these terms are used interchangeably.
[00112] One of ordinary skill in the art will recognize that a variety of warhead groups, as defined herein, are suitable for such covalent bonding. Such R1 groups include, but are not limited to, those described herein and depicted in Table 3, infra.
[00113] As depicted in formulae I-a and I-b supra, the R1 warhead group can be in an ortho-, meta-, or para-position. In certain embodiments, the R1 warhead group is in a meta-position of the phenyl ring relative to the rest of the molecule. [00114] In certain embodiments, R1 is characterized in that the -L-Y moiety is capable of covalently binding to a cysteine residue of TEC, thereby irreversibly inhibiting the enzyme. In some embodiments, the cysteine residue is Cys 449.
[00115] In certain embodiments, R1 is characterized in that the -L-Y moiety is capable of covalently binding to a cysteine residue of BTK, thereby irreversibly inhibiting the enzyme. In some embodiments, the cysteine residue is Cys 481.
[00116] In certain embodiments, R1 is characterized in that the -L-Y moiety is capable of covalently binding to a cysteine residue of ITK, thereby irreversibly inhibiting the enzyme. In some embodiments, the cysteine residue is Cys 442.
[00117] In certain embodiments, R1 is characterized in that the -L-Y moiety is capable of covalently binding to a cysteine residue of BMX, thereby irreversibly inhibiting the enzyme. In some embodiments, the cysteine residue is Cys 496.
[00118] In certain embodiments, R1 is characterized in that the -L-Y moiety is capable of covalently binding to a cysteine residue of JAK3, thereby irreversibly inhibiting the enzyme. In some embodiments, the cysteine residue is Cys 909.
[00119] In certain embodiments, R1 is characterized in that the -L-Y moiety is capable of covalently binding to a cysteine residue of TXK, thereby irreversibly inhibiting the enzyme. In some embodiments, the cysteine residue is Cys 350.
[00120] One of ordinary skill in the art will recognize that a variety of warhead groups, as defined herein, are suitable for such covalent bonding. Such R1 groups include, but are not limited to, those described herein and depicted in Table 3, infra.
[00121] Exemplary compounds of the present invention are set forth in Table 5 below.
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0003
[00122] In certain embodiments, the present invention provides any compound depicted in
Table 5, above, or a pharmaceutically acceptable salt thereof.
[00123] In certain embodiments, the present invention provides a compound selected from:
Figure imgf000094_0001
1-7 1-4
Figure imgf000094_0002
1-96 1-182 1-190
Figure imgf000095_0001
1-342 or a pharmaceutically acceptable salt thereof.
[00124] As described herein, compounds of the present invention are irreversible inhibitors of at least one of ErbBl, ErbB2, ErbB3 and ErbB4, or a mutant thereof. In some embodiments, provided compounds are irreversible inhibitors of a TEC-kinase (e.g. BTK) and JAK3. One of ordinary skill in the art will recognize that certain compounds of the present invention are reversible inhibitors. In certain embodiments, such compounds are useful as assay comparator compounds. In other embodiments, such reversible compounds are useful as inhibitors of ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3, or a mutant thereof, and therefore useful for treating one or disorders as described herein. An exemplary reversible compound of the present invention has the following structure.
Figure imgf000095_0002
IR-7 or a pharmaceutically acceptable salt thereof. 4. Uses, Formulation and Administration Pharmaceutically Acceptable Compositions
[00125] 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. The amount of compound in compositions of this invention is such that is effective to measurably inhibit a protein kinase, particularly at least one of ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3, or a mutant thereof, in a biological sample or in a patient. In certain embodiments, the amount of compound in compositions of this invention is such that is effective to measurably inhibit at least one of ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3, or a mutant thereof, in a biological sample or in a patient. In certain embodiments, a composition of this invention is formulated for administration to a patient in need of such composition. In some embodiments, a composition of this invention is formulated for oral administration to a patient. [00126] The term "patient", as used herein, means an animal, preferably a mammal, and most preferably a human.
[00127] The term "pharmaceutically acceptable carrier, adjuvant, or vehicle" refers to a nontoxic carrier, adjuvant, or vehicle that does not destroy the pharmacological activity of the compound with which it is formulated. Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the compositions of this invention 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, 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, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat. [00128] A "pharmaceutically acceptable derivative" means any non-toxic salt, ester, salt of an ester or other derivative of a compound of this invention that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or an inhibitorily active metabolite or residue thereof.
[00129] As used herein, the term "inhibitorily active metabolite or residue thereof means that a metabolite or residue thereof is also an inhibitor of at least one of ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3, or a mutant thereof.
[00130] 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, intraarticular, intra-synovial, intrasternal, intrathecal, 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 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 and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium.
[00131] For this purpose, any bland fixed oil may be employed including synthetic mono- or di-glycerides. 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.
[00132] 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.
[00133] Alternatively, 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.
[00134] 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. [00135] 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.
[00136] For topical applications, provided 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 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, provided 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.
[00137] For ophthalmic use, provided pharmaceutically acceptable compositions may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or, preferably, as solutions in isotonic, pH adjusted sterile saline, 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.
[00138] 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, fluorocarbons, and/or other conventional solubilizing or dispersing agents. [00139] Most preferably, pharmaceutically acceptable compositions of this invention are formulated for oral administration. [00140] The amount of 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, provided 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.
[00141] It should also be understood that a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated. The amount of a compound of the present invention in the composition will also depend upon the particular compound in the composition.
Uses of Compounds and Pharmaceutically Acceptable Compositions
[00142] Compounds and compositions described herein are generally useful for the inhibition of protein kinase activity of one or more enzymes.
[00143] Drug resistance is emerging as a significant challenge for targeted therapies. For example, drug resistance has been reported for Gleevec® and Iressa®, as well as several other kinase inhibitors in development. In addition, drug resistance has been reported for the cKit and
PDGFR receptors. It has been reported that irreversible inhibitors may be effective against drug resistant forms of protein kinases (Kwak, E. L., R. Sordella, et al. (2005). "Irreversible inhibitors of the EGF receptor may circumvent acquired resistance to gefitinib." PNAS 102(21): 7665-
7670.) Without wishing to be bound by any particular theory, it is believed that compounds of the present invention may be effective inhibitors of drug resistant forms of protein kinases.
[00144] As used herein, the term "clinical drug resistance" refers to the loss of susceptibility of a drug target to drug treatment as a consequence of mutations in the drug target.
[00145] As used herein, the term "resistance" refers to changes in the wild-type nucleic acid sequence coding a target protein, and/or the protein sequence of the target, which changes decrease or abolish the inhibitory effect of the inhibitor on the target protein.
[00146] Examples of kinases that are inhibited by the compounds and compositions described herein and against which the methods described herein are useful include ErbBl, ErbB2, ErbB3,
ErbB4, a TEC-kinase (including BTK, ITK, TEC, BMX and RLK), and/or JAK3, or a mutant thereof. [00147] The activity of a compound utilized in this invention as an inhibitor of ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3, or a mutant thereof, may be assayed in vitro, in vivo or in a cell line. In vitro assays include assays that determine inhibition of either the phosphorylation activity and/or the subsequent functional consequences, or ATPase activity of activated ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3, or a mutant thereof. Alternate in vitro assays quantitate the ability of the inhibitor to bind to ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3. Inhibitor binding may be measured by radio labeling the inhibitor prior to binding, isolating the inhibitor/ErbBl, inhibitor/ErbB2, inhibitor/ErbB3, inhibitor/ErbB4, inhibitor/TEC-kinase (i.e., TEC, BTK, ITK, RLK and BMX), or inhibitor/JAK3 complex and determining the amount of radiolabel bound. Alternatively, inhibitor binding may be determined by running a competition experiment where new inhibitors are incubated with ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3 bound to known radioligands. Detailed conditions for assaying a compound utilized in this invention as an inhibitor of ErbB 1 , ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3, or a mutant thereof, are set forth in the Examples below.
[00148] Protein tyrosine kinases are a class of enzymes that catalyze the transfer of a phosphate group from ATP or GTP to a tyrosine residue located on a protein substrate. Receptor tyrosine kinases act to transmit signals from the outside of a cell to the inside by activating secondary messaging effectors via a phosphorylation event. A variety of cellular processes are promoted by these signals, including proliferation, carbohydrate utilization, protein synthesis, angiogenesis, cell growth, and cell survival. (a) ErbB Family
[00149] ErbB receptors, a major family of receptor tyrosine kinases, are composed of an extracellular ligand binding domain, a single transmembrane domain, and an intracellular domain with tyrosine kinase activity. The ErbB family comprises ErbBl (commonly known as EGFR), ErbB2 (commonly known as HER2 or neu), ErbB3 (commonly known as HER3), and ErbB4 (commonly known as HER4). More than 10 ligands (including EGF, TGFα, AR, BTC, EPR, HB-EGF, NRG-I, NRG-2, NRG-3, NRG-4) have been identified for the various receptor family members. Upon ligand binding the extracellular domain undergoes conformational change, allowing the formation of homodimers or heterodimers with other members of the ErbB family. Dimerization induces tyrosine phosphorylation of specific residues in the intracellular domain that serve as docking sites for adaptor proteins and downstream effectors. In some contexts, activation of phosphatidyl-inositol 3-kinase (PBK) and mitogen-activated protein kinase pathways occur, leading to cell proliferation and survival (Lin, N. U.; Winer, E. P., Breast Cancer Res 6: 204-210, 2004).
[00150] Interaction between family members is necessitated by deficiencies in ErbB2, which has no known ligand, and ErbB3, which is kinase dead. EGFR, ErbB3, and ErbB4 bind ligand to induce ErbB receptor homodimerization or heterodimerization, whereas ErbB2 functions as the preferred dimerization partner. The composition of the pairwise combinations is important for signal diversification, as dimer identity determines which downstream pathways are activated. Representative downstream gene products in the ErbB signal transduction pathway include She, Grb2, SOSl, Ras, Rafl, Mek, ERKl, ERK2, ERa, Akt, mTOR, FKHR, p27, Cyclin Dl, FasL, GSK-3, Bad, and STAT3.
[00151] There is strong precedent for involvement of the EGFR and other members of the ErbB family in human cancer because over 60% of all solid tumors overexpress at least one of these proteins or their ligands. Constitutively active, tumorigenic EGFR vIII, a mutant possessing a truncated extracellular domain, has been reported to be present in up to 78% of breast carcinomas and has also been found in glioblastomas. Overexpression of EGFR is commonly found in breast, lung, head and neck, bladder tumors, while ErbB2 expression is frequently elevated in human tumors of epithelial origin. Activating mutations in the tyrosine kinase domain have been identified in patients with non-small cell lung cancer (Lin, N. U.; Winer, E. P., Breast Cancer Res 6: 204-210, 2004). ErbBl and/or ErbB2 amplification has also been implicated in squamous cell carcinomas, salivary gland carcinomas, ovarian carcinomas, and pancreatic cancers (Cooper, G. C. Oncogenes. 2nd ed. Sudbury: Jones and Barlett, 1995; Zhang, Y., et al, Cancer Res 66 : 1025-32, 2006). Overexpression of ErbB2 has potent transforming activity, likely due to its ability to cooperate with other ErbB receptors (Sherman, L., et al., Oncogene J_8: 6692-99, 1999). In fact, some human cancers that overexpress both EGFR and ErbB2 have a poorer prognosis than cancers that overexpress either receptor alone. [00152] The ErbB signaling network is often a key component in the pathogenesis of breast cancer. Amplification of ErbB2 is associated with an aggressive tumor phenotype that is characterized by relatively rapid tumor growth, metastatic spread to visceral sites, and drug resistance. ErbB2 has been shown to be amplified in 20% of axillary node-negative ("ANN") breast cancer cases, and this amplification has been identified as an independent prognostic factor for risk of recurrence in ANN breast cancer. (Andrulis, LL. , et al., J Clin Oncol Jj5: 1340- 9, 1998).
[00153] Targeted blockade of ErbB signaling with trastuzumab (Herceptin), a monoclonal antibody directed at ErbB2, has been shown to improve survival in women with ErbB2-positive, advanced breast cancer. Other monoclonal antibodies directed against ErbB receptors include cetuximab (Erbitux) and panitumumab (Vectibix).
[00154] Several small molecule tyrosine kinase inhibitors (TKIs) have been found to act selectively upon ErbB family members. Notable examples include gefitinib (Iressa) and erlotinib (Tarceva), both of which target the EGFR. These small molecules compete with ATP for binding to the kinase domain of the receptor. Compared to monoclonal antibodies, TKIs have several advantages in that they are orally bioavailable, well-tolerated, and appear to be active against truncated forms of ErbB2 and EGFR receptors (e.g., EGFR vIII) in vitro. In addition, the small size of small molecule TKIs may allow them to penetrate sanctuary sites such as the central nervous system. Finally, the homology between kinase domains of ErbB receptors allows for development of TKIs that target more than one member of the ErbB family simultaneously, the advantages of which are described herein.
[00155] Although certain malignancies have been linked to the overexpression of individual receptors, efficient signal transduction relies on the coexpression of ErbB receptor family members. This cooperation of ErbB receptor family members in signal transduction and malignant transformation may limit the success of agents that target individual receptors in the treatment of cancer; a potential mechanism of resistance to agents targeting a single ErbB receptor is upregulation of other members of the receptor family (Britten, C. D., MoI Cancer Ther 3: 1335-42, 2004).
[00156] Agents that target two or more ErbB receptors are called pan-ErbB regulators. ERRP is a pan-ErbB negative regulator that is expressed in most benign pancreatic ductal epithelium and islet cells. Tumors have been found to experience a progressive loss in ERRP expression. That Erbitux and Herceptin show success in a limited patient base (tumors having increased expression of EGFR or ErbB2) could be partly due to coexpression of multiple ErbB family members. [00157] In both in vitro and in vivo models, strategies that employ a dual ErbB approach seem to have greater antitumor activity than agents targeting a single ErbB receptor. Thus, agents that target multiple members of ErbB family are likely to provide therapeutic benefit to a broader patient population (Zhang, Y., et al., Cancer Res 66: 1025-32, 2006). In certain embodiments, provided compounds inhibit one or more of ErbBl, ErbB2, ErbB3, and ErbB4. In some embodiments, provided compounds inhibit two or more of ErbBl, ErbB2, ErbB3, and ErbB4, or a mutant thereof, and are therefore pan-ErbB inhibitors.
[00158] Clearly, there is growing evidence to support the concurrent inhibition of two or more ErbB (i.e., pan-ErbB) receptors in cancer therapy. Possible pan-ErbB approaches with small molecules include using combinations of agents that target individual ErbB receptors, using single agents that target multiple ErbB receptors, or using agents that interfere with ErbB receptor interactions (e.g., dimerization). Additional strategies include therapies utilizing a small molecule in combination with antibodies, or chemoprevention therapies (Lin, N. U.; Winer, E. P., Breast Cancer Res 6: 204-210, 2004).
[00159] An example of small molecule pan-ErbB inhibition is CI- 1033, an irreversible pan- ErbB inhibitor that covalently binds to the ATP binding site of the intracellular kinase domain. Another irreversible pan-ErbB receptor tyrosine kinase inhibitor is HKI-272, which inhibits the growth of tumor cells that express ErbB-1 (EGFR) and ErbB-2 (HER-2) in culture and xenografts, and has antitumor activity in HER-2-positive breast cancer (Andrulis, LL. , et al., J Clin Oncol 16: 1340-9, 1998). Irreversible inhibitors have demonstrated superior antitumor activity in comparison with reversible inhibitors.
[00160] Neurofibromatosis type I (NFl) is a dominantly inherited human disease affecting one in 2500-3500 individuals. Several organ systems are affected, including bones, skin, iris, and the central nervous system, as manifested in learning disabilities and gliomas. A hallmark of NFl is the development of benign tumors of the peripheral nervous system (neurofibromas), which vary greatly in both number and size among patients. Neurofibromas are heterogeneous tumors composed of Schwann cells, neurons, fibroblasts and other cells, w/ Schwann cells being the major (60-80%) cell type.
[00161] Abberant expression of the EGFR is associated with tumor development in NFl and in animal models of NFl, suggesting a role in pathogenesis and representing a novel potential therapeutic target. EGFR expression affects the growth of tumor cell lines derived from NFl patients under conditions where EGF is not the primary factor driving growth of the cells. These data suggest that EGFR may play an important role in NFl tumorigenesis and Schwann cell transformation (DeClue, J. E., et al, J Clin Invest 105: 1233-41, 2000).
[00162] Patients with NFl develop aggressive Schwann cell neoplasmas known as malignant peripheral nerve sheath tumors (MPNSTs). Schwann cells are the major supportive cell population in the peripheral nervous system. Neoplastic Schwann cells within these neoplasms variably express the ErbB tyrosine kinases mediating NRG-I responses (ErbB2, ErbB3, ErbB4). Neuregulin-1 (NRG-I) proteins promote the differentiation, survival, and/or proliferation of many cell types in the developing nervous system, and overexpression of NRG-I in myelinating Schwann cells induces the formation of malignant peripheral nerve sheath tumors (MPNSTs) (Fallon, K. B., et al, J Neuro Oncol 66: 273-84, 2004).
[00163] Deregulation of Schwann cell growth is a primary defect driving the development of both benign neurofibromas and MPNST in neurofibromatosis type I (NFl) patients. Growth of MPNSTs and transformed mouse Schwann cells in vitro is highly EGF-dependent and can be blocked by EGFR inhibitors under conditions where EGF is the primary growth factor. Some human MPNST cell lines have been found to demonstrate constitutive ErbB phosphorylation. While treatment with ErbB inhibitors abolishes ErbB phosphorylation and reduces DNA synthesis in these lines, effective chemotherapeutic regimens for MPNST remain elusive (Stonecypher, M. S., et al., Oncogene 24: 5589-5605, 2005).
[00164] Schwannomas are peripheral nerve tumors comprised almost entirely of Schwann- like cells, and typically have mutations in the neurofibromatosis type II (NF2) tumor suppressor gene. Ninety percent of NF2 patients develop bilateral vestibular schwannomas and/or spinal schwannomas. Enlarging schwannomas can compress adjacent structures, resulting in deafness and other neurologic problems. Surgical removal of these tumors is difficult, often resulting in increased patient morbidity.
[00165] Both normal human Schwann cells and schwannoma cells express neuregulin receptors (i.e., ErbB receptors), and schwannoma cells proliferate in response to neuregulin. It is possible that aberrant neuregulin production or response contributes to aberrant schwannoma cell proliferation (Pelton, P. D., et al., Oncogene 17: 2195-2209, 1998).
[00166] The NF2 tumor suppressor, Merlin, is a membrane/cytoskeleton-associated protein implicated in the regulation of tyrosine kinase activity. Genetic interactions between a Merlin mutation and EGFR pathway mutations have been documented in Drosophila (LaJeunesse, D. R., et al, Genetics 158: 667-79, 2001). Other evidence suggests Merlin can inhibit EGFR internalization and signaling upon cell-cell contact by restraining the EGFR into a membrane compartment from which it can neither signal nor be internalized (McClatchey, A. L, et al., Genes and Development 19: 2265-77, 2005; Curto, M. C, et al., J Cell Biol 177: 893-903, 2007). [00167] As used herein, the terms "treatment," "treat," and "treating" refer to reversing, alleviating, delaying the onset of, or inhibiting the progress of a disease or disorder, or one or more symptoms thereof, as described herein. In some embodiments, treatment may be administered after one or more symptoms have developed. In other embodiments, treatment may be administered in the absence of symptoms. For example, treatment may be administered to a susceptible individual prior to the onset of symptoms (e.g., in light of a history of symptoms and/or in light of genetic or other susceptibility factors). Treatment may also be continued after symptoms have resolved, for example to prevent or delay their recurrence.
[00168] Provided compounds are inhibitors of one or more of ErbBl, ErbB2, ErbB3, and ErbB4 and are therefore useful for treating one or more disorders associated with activity of one of more of ErbBl, ErbB2, ErbB3, and ErbB4. Thus, in certain embodiments, the present invention provides a method for treating an ErbBl -mediated, an ErbB2-mediated, an ErbB3- mediated, and/or ErbB4-mediated disorder comprising the step of administering to a patient in need thereof a compound of the present invention, or pharmaceutically acceptable composition thereof.
[00169] As used herein, the terms "ErbBl -mediated", "ErbB2-mediated," "ErbB 3 -mediated," and/or "ErbB4-mediated" disorders or conditions as used herein means any disease or other deleterious condition in which one or more of ErbBl, ErbB2, ErbB3, and/or ErbB4, or a mutant thereof, are known to play a role. Accordingly, another embodiment of the present invention relates to treating or lessening the severity of one or more diseases in which one or more of ErbBl, ErbB2, ErbB3, and/or ErbB4, or a mutant thereof, are known to play a role. Specifically, the present invention relates to a method of treating or lessening the severity of a disease or condition selected from a proliferative disorder, wherein said method comprises administering to a patient in need thereof a compound or composition according to the present invention. [00170] In some embodiments, the present invention provides a method for treating or lessening the severity of one or more disorders selected from a cancer. In some embodiments, the cancer is associated with a solid tumor. In certain embodiments, the cancer is breast cancer, glioblastoma, lung cancer, cancer of the head and neck, colorectal cancer, bladder cancer, or non-small cell lung cancer. In some embodiments, the present invention provides a method for treating or lessening the severity of one or more disorders selected from squamous cell carcinoma, salivary gland carcinoma, ovarian carcinoma, or pancreatic cancer. [00171] In certain embodiments, the present invention provides a method for treating or lessening the severity of neurofibromatosis type I (NFl), neurofibromatosis type II (NF2) Schwann cell neoplasms (e.g. MPNST's), or Schwannomas. (b) TEC Family
[00172] The TEC family of non-receptor tyrosine kinases, referred to herein as "TEC- kinases," plays a central role in signaling through antigen-receptors such as the TCR, BCR and Fc receptors (reviewed in Miller A, et al. Current Opinion in Immunology 14; 331-340 (2002). TEC-kinases are essential for T cell activation. Three members of the family, Itk, RIk and, are activated downstream of antigen receptor engagement in T cells and transmit signals to downstream effectors, including PLC-γ. Combined deletion of Itk and RIk in mice leads to a profound inhibition of TCR responses including proliferation, cytokine production and immune responses to an intracellular parasite (Toxoplasma gondii) (Schaeffer et al., Science 284; 638- 641 (1999)). Intracellular signalling following TCR engagement is effected in ITK/RLK deficient T cells; inositol triphosphate production, calcium mobilization and MAP kinase activation are all reduced. Tec-kinases are also essential for B cell development and activation. [00173] TEC-kinases include five family members, which are expressed primarily in hematopoietic cells: TEC, BTK, ITK (also known as TSK and EMT), RLK (also known as TXK), and BMX (also known as ETK). Additional related TEC-kinases have been found in Drosophila melanogaster, zebrafϊsh (Danio reriό), skate (Raja eglanteria), and sea urchin (Anthocidaris crassispina).
[00174] Provided compounds are inhibitors of one of more TEC-kinases and are therefore useful for treating one or more disorders associated with activity of one or more TEC-kinases. Thus, in certain embodiments, the present invention provides a method for treating a TEC- mediated disorder comprising the step of administering to a patient in need thereof a compound of the present invention, or pharmaceutically acceptable composition thereof. [00175] The term "TEC-mediated condition", as used herein means any disease or other deleterious condition in which TEC-kinases are known to play a role. Such conditions include those described herein and in Melcher, M et al., "The Role of TEC Family Kinases in Inflammatory Processes", Anti-Inflammatory & Anti-Allergy Agents in Medicinal Chemistry, Vol. 6, No. 1, pp. 61-69 (Feb. 2007). Accordingly, another embodiment of the present invention relates to treating or lessening the severity of one or more diseases in which TEC-kinases are known to play a role. Specifically, the present invention relates to a method of treating or lessening the severity of a disease or condition selected from autoimmune, inflammatory, proliferative, and hyperproliferative diseases and immunologically-mediated diseases including rejection of transplanted organs or tissues and Acquired Immunodeficiency Syndrome (AIDS)(also known as HIV), wherein said method comprises administering to a patient in need thereof a composition of the present invention.
[00176] In some embodiments, the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with TEC-kinases including diseases of the respiratory tract including, without limitation, reversible obstructive airways diseases including asthma, such as bronchial, allergic, intrinsic, extrinsic and dust asthma, particularly chronic or inveterate asthma (e.g., late asthma airways hyper- responsiveness) and bronchitis. In some embodiments, the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with TEC-kinases including those conditions characterized by inflammation of the nasal mucus membrane, including acute rhinitis, allergic, atrophic rhinitis and chronic rhinitis including rhinitis caseosa, hypertrophic rhinitis, rhinitis purulenta, rhinitis sicca and rhinitis medicamentosa; membranous rhinitis including croupous, fibrinous and pseudomembranous rhinitis and scrofoulous rhinitis, seasonal rhinitis including rhinitis nervosa (hay fever) and vasomotor rhinitis, sarcoidosis, farmer's lung and related diseases, fibroid lung, and idiopathic interstitial pneumonia.
[00177] In some embodiments, the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with TEC-kinases including diseases of the bone and joints including, without limitation, rheumatoid arthritis, seronegative spondyloarthropathies (including ankylosing spondylitis, psoriatic arthritis and Reiter's disease), Behcet's disease, Sjogren's syndrome, systemic sclerosis, osteoporosis, bone cancer, and bone metastasis.
[00178] In some embodiments, the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with TEC-kinases including diseases and disorders of the skin, including, without limitation, psoriasis, systemic sclerosis, atopical dermatitis, contact dermatitis and other eczematous dermatitis, seborrhoetic dermatitis, Lichen planus, pemphigus, bullous pemphigus, epidermolysis bullosa, urticaria, angiodermas, vasculitides, erythemas, cutaneous eosinophilias, uveitis, alopecia, areata and vernal conjunctivitis.
[00179] In some embodiments, the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with TEC-kinases including diseases and disorders of the gastrointestinal tract, including, without limitation, celiac disease, proctitis, eosinophilic gastro-enteritis, mastocytosis, pancreatitis, Crohn's disease, ulcerative colitis, food-related allergies which have effects remote from the gut, e. g. migraine, rhinitis and eczema.
[00180] In some embodiments, the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with TEC-kinases including those diseases and disorders of other tissues and systemic disease, including, without limitation, multiple sclerosis, artherosclerosis, lupus erythematosus, systemic lupus erythematosus, Hashimoto's thyroiditis, myasthenia gravis, type I diabetes, nephrotic syndrome, eosinophilia fascitis, hyper IgE syndrome, lepromatous leprosy, sezary syndrome and idiopathic thrombocytopenia purpura, restenosis following angioplasty, tumours (for example leukemia, lymphomas, and prostate cancers), and artherosclerosis.
[00181] In some embodiments, the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with TEC-kinases including allograft rejection including, without limitation, acute and chronic allograft rejection following for example transplantation of kidney, heart, liver, lung, bone marrow, skin and cornea; and chronic graft versus host disease.
[00182] In some embodiments, the present invention relates to a method of treating or lessening the severity of one or more of the diseases or conditions associated with TEC-kinases, as recited above, wherein said method comprises administering to a patient in need thereof a compound or composition according to the present invention. (c) Bmton's tyrosine kinase (BTK)
[00183] Bruton's tyrosine kinase ("BTK"), a member of TEC-kinases, is a key signaling enzyme expressed in all hematopoietic cell types except T lymphocytes and natural killer cells. BTK plays an essential role in the B-cell signaling pathway linking cell surface B-cell receptor (BCR) stimulation to downstream intracellular responses.
[00184] BTK is a key regulator of B-cell development, activation, signaling, and survival (Kurosaki, Curr Op Imm, 2000, 276-281; Schaeffer and Schwartzberg, Curr Op Imm 2000, 282- 288). In addition, BTK plays a role in a number of other hematopoietic cell signaling pathways, e.g., Toll like receptor (TLR) and cytokine receptor-mediated TNF-α production in macrophages, IgE receptor (Fc epsilon RI) signaling in mast cells, inhibition of Fas/ APO-I apoptotic signaling in B-lineage lymphoid cells, and collagen-stimulated platelet aggregation. See, e.g., C. A. Jeffries, et al, (2003), Journal of Biological Chemistry 278:26258-26264; N. J. Horwood, et al, (2003), The Journal of Experimental Medicine 197: 1603- 1611 ; Iwaki et al. (2005), Journal of Biological Chemistry 280(48) :40261 -40270; Vassilev et al. (1999), Journal of Biological Chemistry 274(3): 1646-1656, and Quek et al. (1998), Current Biology 8(20): 1137-1140. [00185] Patients with mutations in BTK have a profound block in B cell development, resulting in the almost complete absence of mature B lymphocytes and plasma cells, severely reduced Ig levels and a profound inhibition of humoral response to recall antigens (reviewed in Vihinen et al Frontiers in Bioscience 5 : d917-928). Mice deficient in BTK also have a reduced number of peripheral B cells and greatly decreased serum levels of IgM and IgG3. BTK deletion in mice has a profound effect on B cell proliferation induced by anti-IgM, and inhibits immune responses to thymus-independent type II antigens (Ellmeier et al, J Exp Med 192: 1611-1623 (2000)). BTK also plays a crucial role in mast cell activation through the high-affinity IgE receptor (Fc epsilon RI). BTK deficient murine mast cells have reduced degranulation and decreased production of proinflammatory cytokines following Fc epsilon RI cross-linking (Kawakami et al. Journal of Leukocyte Biology 65: 286-290).
[00186] Provided compounds are inhibitors of BTK and are therefore useful for treating one or more disorders associated with activity of BTK. Thus, in some embodiments, the present invention provides a method for treating a BTK-mediated disorder comprising the step of administering to a patient in need thereof a compound of the present invention, or pharmaceutically acceptable composition thereof.
[00187] As used herein, the term "BTK-mediated" disorders or conditions as used herein means any disease or other deleterious condition in which BTK, or a mutant thereof, is known to play a role. Accordingly, another embodiment of the present invention relates to treating or lessening the severity of one or more diseases in which BTK, or a mutant thereof, is known to play a role. Specifically, the present invention relates to a method of treating or lessening the severity of a disease or condition selected from a proliferative disorder or an autoimmune disorder, wherein said method comprises administering to a patient in need thereof a compound or composition according to the present invention.
[00188] In some embodiments, the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with BTK. In some embodiments, the disease or condition is an autoimmune disease, e.g., inflammatory bowel disease, arthritis, lupus, rheumatoid arthritis, psoriatic arthritis, osteoarthritis, Still's disease, juvenile arthritis, diabetes, myasthenia gravis, Hashimoto's thyroiditis, Ord's thyroiditis, Graves' disease, Sjogren's syndrome, multiple sclerosis, Guillain-Barre syndrome, acute disseminated encephalomyelitis, Addison's disease, opsoclonus-myoclonus syndrome, ankylosing spondylosis, antiphospholipid antibody syndrome, aplastic anemia, autoimmune hepatitis, celiac disease, Goodpasture's syndrome, idiopathic thrombocytopenic purpura, optic neuritis, scleroderma, primary biliary cirrhosis, Reiter's syndrome, Takayasu's arteritis, temporal arteritis, warm autoimmune hemolytic anemia, Wegener's granulomatosis, psoriasis, alopecia universalis, Behcet's disease, chronic fatigue, dysautonomia, endometriosis, interstitial cystitis, neuromyotonia, scleroderma, or vulvodynia. In some embodiments, the disease or condition is a hyperproliferative disease or immunologically-mediated diseases including rejection of transplanted organs or tissues and Acquired Immunodeficiency Syndrome (AIDS, also known as HIV).
[00189] In some embodiments, the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with BTK, wherein the disease or condition is selected from heteroimmune conditions or diseases, which include, but are not limited to graft versus host disease, transplantation, transfusion, anaphylaxis, allergies (e.g., allergies to plant pollens, latex, drugs, foods, insect poisons, animal hair, animal dander, dust mites, or cockroach calyx), type I hypersensitivity, allergic conjunctivitis, allergic rhinitis, and atopic dermatitis.
[00190] In some embodiments, the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with BTK, wherein the disease or condition is selected from an inflammatory disease, e.g., asthma, appendicitis, blepharitis, bronchiolitis, bronchitis, bursitis, cervicitis, cholangitis, cholecystitis, colitis, conjunctivitis, cystitis, dacryo adenitis, dermatitis, dermatomyositis, encephalitis, endocarditis, endometritis, enteritis, enterocolitis, epicondylitis, epididymitis, fasciitis, fϊbrositis, gastritis, gastroenteritis, hepatitis, hidradenitis suppurativa, laryngitis, mastitis, meningitis, myelitis myocarditis, myositis, nephritis, oophoritis, orchitis, osteitis, otitis, pancreatitis, parotitis, pericarditis, peritonitis, pharyngitis, pleuritis, phlebitis, pneumonitis, pneumonia, proctitis, prostatitis, pyelonephritis, rhinitis, salpingitis, sinusitis, stomatitis, synovitis, tendonitis, tonsillitis, uveitis, vaginitis, vasculitis, or vulvitis.
[00191] In some embodiments, the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with BTK, wherein the disease or condition is selected from a cancer. In one embodiment, the cancer is a B-cell proliferative disorder, e.g., diffuse large B cell lymphoma, follicular lymphoma, chronic lymphocytic lymphoma, chronic lymphocytic leukemia, acute lymphocytic leukemia, B-cell prolymphocytic leukemia, lymphoplasmacytic lymphoma/Waldenstrom macroglobulinemia, splenic marginal zone lymphoma, multiple myeloma (also known as plasma cell myeloma), non- Hodgkin's lymphoma, Hodgkin's lymphoma, plasmacytoma, extranodal marginal zone B cell lymphoma, nodal marginal zone B cell lymphoma, mantle cell lymphoma, mediastinal (thymic) large B cell lymphoma, intravascular large B cell lymphoma, primary effusion lymphoma, Burkitt lymphoma/leukemia, or lymphomatoid granulomatosis. In some embodiments, the cancer is breast cancer, prostate cancer, or cancer of the mast cells (e.g., mastocytoma, mast cell leukemia, mast cell sarcoma, systemic mastocytosis). In one embodiment, the cancer is bone cancer. In another embodiment, the cancer is of other primary origin and metastasizes to the bone.
[00192] In some embodiments, the present invention provides a method for treating or lessening the severity of one or more diseases or conditions associated with BTK including diseases of the bone and joints including, without limitation, rheumatoid arthritis, seronegative spondyloarthropathies (including ankylosing spondylitis, psoriatic arthritis and Reiter's disease), Behcet's disease, Sjogren's syndrome, systemic sclerosis, osteoporosis, bone cancer, and bone metastasis.
[00193] In some embodiments, the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with BTK, wherein the disease or condition is selected from a thromboembolic disorder, e.g., myocardial infarct, angina pectoris, reocclusion after angioplasty, restenosis after angioplasty, reocclusion after aortocoronary bypass, restenosis after aortocoronary bypass, stroke, transitory ischemia, a peripheral arterial occlusive disorder, pulmonary embolism, or deep venous thrombosis. [00194] In some embodiments, the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with BTK, including infectious and noninfectious inflammatory events and autoimmune and other inflammatory diseases. These autoimmune and inflammatory diseases, disorders, and syndromes include inflammatory pelvic disease, urethritis, skin sunburn, sinusitis, pneumonitis, encephalitis, meningitis, myocarditis, nephritis, osteomyelitis, myositis, hepatitis, gastritis, enteritis, dermatitis, gingivitis, appendicitis, pancreatitis, cholocystitus, agammaglobulinemia, psoriasis, allergy, Crohn's disease, irritable bowel syndrome, ulcerative colitis, Sjogren's disease, tissue graft rejection, hyperacute rejection of transplanted organs, asthma, allergic rhinitis, chronic obstructive pulmonary disease (COPD), autoimmune polyglandular disease (also known as autoimmune polyglandular syndrome), autoimmune alopecia, pernicious anemia, glomerulonephritis, dermatomyositis, multiple sclerosis, scleroderma, vasculitis, autoimmune hemolytic and thrombocytopenic states, Goodpasture's syndrome, atherosclerosis, Addison's disease, Parkinson's disease, Alzheimer's disease, type I diabetes, septic shock, systemic lupus erythematosus (SLE), rheumatoid arthritis, psoriatic arthritis, juvenile arthritis, osteoarthritis, chronic idiopathic thrombocytopenic purpura, Waldenstrom macroglobulinemia, myasthenia gravis, Hashimoto's thyroiditis, atopic dermatitis, degenerative joint disease, vitiligo, autoimmune hypopituitarism, Guillain-Barre syndrome, Behcet's disease, scleraderma, mycosis fungoides, acute inflammatory responses (such as acute respiratory distress syndrome and ischemia/reperfusion injury), and Graves' disease.
[00195] In some embodiments, the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with BTK, selected from rheumatoid arthritis, multiple sclerosis, B-cell chronic lymphocytic leukemia, acute lymphocytic leukemia, hairy cell leukemia, non-Hodgkin's lymphoma, Hodgkin's lymphoma, multiple myeloma, bone cancer, bone metastasis, osteoporosis, irritable bowel syndrome, Crohn's disease, lupus and renal transplant. (d) ITK
[00196] Interleukin-2 inducible T-cell kinase ("ITK") is expressed in T cells, mast cells and natural killer cells. It is activated in T cells upon stimulation of the T cell receptor (TCR), and in mast cells upon activation of the high affinity IgE receptor. Following receptor stimulation in T cells, Lck, a Src tyrosine kinase family member, phosphorylates Y511 in the kinase domain activation loop of ITK (S. D. Heyeck et al, 1997, J. Biol. Chem, 272, 25401-25408). Activated ITK, together with Zap-70 is required for phosphorylation and activation of PLC-gamma (S. C. Bunnell et al., 2000, J. Biol. Chem., 275, 2219-2230). PLC-gamma catalyzes the formation of inositol 1,4,5-triphosphate and diacylglycerol, leading to calcium mobilization and PKC activation, respectively. These events activate numerous downstream pathways and lead ultimately to degranulation (mast cells) and cytokine gene expression (T cells) (Y. Kawakami et al., 1999, J. Leukocyte Biol, 65, 286-290).
[00197] The role of ITK in T cell activation has been confirmed in ITK knockout mice. CD4+ T cells from ITK knockout mice have a diminished proliferative response in a mixed lymphocyte reaction or upon Con A or anti-CD3 stimulation. (X. C. Liao and D. R. Littman, 1995, Immunity, 3, 757-769). Also, T cells from ITK knockout mice produced little IL-2 upon TCR stimulation resulting in reduced proliferation of these cells. In another study, ITK deficient CD4+ T cells produced reduced levels of cytokines including IL-4, IL-5 and IL- 13 upon stimulation of the TCR, even after priming with inducing conditions (D. J. Fowell, 1999, Immunity, 11, 399-409). [00198] The role of ITK in PLC-gamma activation and in calcium mobilization was also confirmed in the T cells of these knockout mice, which had severely impaired IP3 generation and no extracellular calcium influx upon TCR stimulation (K. Liu et al., 1998, J. Exp. Med. 187, 1721-1727). Such studies support a key role for ITK in activation of T cells and mast cells. Thus an inhibitor of ITK would be of therapeutic benefit in diseases mediated by inappropriate activation of these cells.
[00199] It has been well established that T cells play an important role in regulating the immune response (Powrie and Coffman, 1993, Immunology Today, 14, 270-274). Indeed, activation of T cells is often the initiating event in immunological disorders. Following activation of the TCR, there is an influx of calcium that is required for T cell activation. Upon activation, T cells produce cytokines, including IL-2, 4, 5, 9, 10, and 13 leading to T cell proliferation, differentiation, and effector function. Clinical studies with inhibitors of IL-2 have shown that interference with T cell activation and proliferation effectively suppresses immune response in vivo (Waldmann, 1993, Immunology Today, 14, 264-270). Accordingly, agents that inhibit T lymphocyte activation and subsequent cytokine production, are therapeutically useful for selectively suppressing the immune response in a patient in need of such immunosuppression. [00200] Mast cells play a critical roll in asthma and allergic disorders by releasing proinflammatory mediators and cytokines. Antigen-mediated aggregation of Fc.epsilon.RI, the high- affinity receptor for IgE, results in activation of mast cells (D. B. Corry et al., 1999, Nature, 402, B 18-23). This triggers a series of signaling events resulting in the release of mediators, including histamine, proteases, leukotrienes and cytokines (J. R. Gordon et al., 1990, Immunology Today, 11, 458-464.) These mediators cause increased vascular permeability, mucus production, bronchoconstriction, tissue degradation and inflammation thus playing key roles in the etiology and symptoms of asthma and allergic disorders.
[00201] Published data using ITK knockout mice suggests that in the absence of ITK function, increased numbers of memory T cells are generated (A. T. Miller et al., 2002 The Journal of Immunology, 168, 2163-2172). One strategy to improve vaccination methods is to increase the number of memory T cells generated (S. M. Kaech et al., Nature Reviews Immunology, 2, 251- 262). In addition, deletion of ITK in mice results in reduced T cell receptor (TCR)-induced proliferation and secretion of the cytokines IL-2, IL-4, IL-5, IL-10 and IFN-y (Schaeffer et al, Science 284; 638-641 (1999) ), Fowell et al, Immunity 11, 399-409 (1999), Schaeffer et al, Nature Immunology 2 (12): 1183-1188 (2001) )). The immunological symptoms of allergic asthma are attenuated in ITK-/-mice. Lung inflammation, eosinophil infiltration and mucus production are drastically reduced in ITK-/-mice in response to challenge with the allergen OVA (Mueller et al, Journal of Immunology 170: 5056-5063 (2003)). ITK has also been implicated in atopic dermatitis. This gene has been reported to be more highly expressed in peripheral blood T cells from patients with moderate and/or severe atopic dermatitis than in controls or patients with mild atopic dermatitis (Matsumoto et al, International Archives of Allergy and Immunology 129: 327-340 (2002)). [00202] Splenocytes from RLK-/-mice secrete half the IL-2 produced by wild type animals in response to TCR engagement (Schaeffer et al, Science 284: 638-641 (1999)), while combined deletion of ITK and RLK in mice leads to a profound inhibition of TCR-induced responses including proliferation and production of the cytokines IL-2, IL-4, IL-5 and IFN -y (Schaeffer et al, Nature Immunology 2 (12): 1183-1188 (2001), Schaeffer et al, Science 284: 638-641 (1999)). Intracellular signalling following TCR engagement is effected in ITK/RLK deficient T cells; inositol triphosphate production, calcium mobilization, MAP kinase activation, and activation of the transcription factors NFAT and AP-I are all reduced (Schaeffer et al, Science 284: 638-641 (1999), Schaeffer et al, Nature Immunology 2 (12): 1183-1188 (2001)).
[00203] Provided compounds are inhibitors of ITK and are therefore useful for treating one or more disorders associated with activity of ITK. Thus, in some embodiments, the present invention provides a method for treating an ITK-mediated disorder comprising the step of administering to a patient in need thereof a compound of the present invention, or pharmaceutically acceptable composition thereof.
[00204] As used herein, the term "ITK-mediated" disorders or conditions as used herein means any disease or other deleterious condition in which ITK, or a mutant thereof, is known to play a role. Accordingly, another embodiment of the present invention relates to treating or lessening the severity of one or more diseases in which ITK, or a mutant thereof, is known to play a role. Specifically, the present invention relates to a method of treating or lessening the severity of a disease or condition selected from a mast cell-mediated condition, a basophil- mediated disorder, an immune or allergic disorder, wherein said method comprises administering to a patient in need thereof a compound or composition according to the present invention. [00205] In some embodiments, the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with ITK, wherein the disease or condition is an immune disorder, including inflammatory diseases, autoimmune diseases, organ and bone marrow transplant rejection and other disorders associated with T cell- mediated immune response or mast cell-mediated immune response.
[00206] In certain embodiments, the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with ITK, wherein the disease or condition is acute or chronic inflammation, an allergy, contact dermatitis, psoriasis, rheumatoid arthritis, multiple sclerosis, type 1 diabetes, inflammatory bowel disease, Guillain- Barre syndrome, Crohn's disease, ulcerative colitis, cancer, graft versus host disease (and other forms of organ or bone marrow transplant rejection) or lupus erythematosus.
[00207] In certain embodiments, the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with ITK, wherein the disease or condition is a mast cell driven conditions, a basophil-mediated disorder, reversible obstructive airway disease, asthma, rhinitis, chronic obstructive pulmonary disease (COPD), peripheral T-cell lymphomas or HIV [also known as Acquired Immunodeficiency Syndrome
(AIDS)]. Such conditions include those described in Readinger, et al, PNAS 105: 6684-6689
(2008).
(e) JAK Family
[00208] The Janus kinases (JAK) are a family of tyrosine kinases consisting of JAKl, JAK2,
JAK3 and TYK2. The JAKs play a critical role in cytokine signaling. The down-stream substrates of the JAK family of kinases include the signal transducer and activator of transcription (STAT) proteins. JAK/STAT signaling has been implicated in the mediation of many abnormal immune responses such as allergies, asthma, autoimmune diseases such as transplant rejection, rheumatoid arthritis, amyotrophic lateral sclerosis and multiple sclerosis as well as in solid and hematologic malignancies such as leukemias and lymphomas. The pharmaceutical intervention in the JAK/STAT pathway has been reviewed [Frank, MoI. Med. 5 :
432-456 (1999) & Seidel, et al, Oncogene 19 : 2645-2656 (2000)].
[00209] JAKl, JAK2, and TYK2 are ubiquitously expressed, while JAK3 is predominantly expressed in hematopoietic cells. JAK3 binds exclusively to the common cytokine receptor gamma chain (yc) and is activated by IL-2, IL-4, IL-7, IL-9, and IL-15.
[00210] The proliferation and survival of murine mast cells induced by IL-4 and IL-9 have, in fact, been shown to be dependent on JAK3-and yc-signaling [Suzuki et al, Blood 96 : 2172-2180
(2000) ].
[00211] Cross-linking of the high-affinity immunoglobulin (Ig) E receptors of sensitized mast cells leads to a release of proinflammatory mediators, including a number of vasoactive cytokines resulting in acute allergic, or immediate (type I) hypersensitivity reactions [Gordon et al, Nature 346 : 274-276 (1990) & Galli, N. Engl. J. Med., 328 : 257- 265 (1993) ]. A crucial role for JAK3 in IgE receptor-mediated mast cell responses in vitro and in vivo has been established [Malaviya, et al, Biochem. Biophys. Res. Commun. 257 : 807-813 (1999) ]. In addition, the prevention of type I hypersensitivity reactions, including anaphylaxis, mediated by mast cell-activation through inhibition of JAK3 has also been reported [Malaviya et al, J. Biol.
Chem. 274 : 27028-27038 (1999) ]. Targeting mast cells with JAK3 inhibitors modulated mast cell degranulation in vitro and prevented IgE receptor/antigen-mediated anaphylactic reactions in vivo.
[00212] A recent study described the successful targeting of JAK3 for immune suppression and allograft acceptance. The study demonstrated a dose-dependent survival of buffalo heart allograft in Wistar Furth recipients upon administration of inhibitors of JAK3 indicating the possibility of regulating unwanted immune responses in graft versus host disease [Kirken,
Transpl. Proc. 33: 3268-3270 (2001)].
[00213] IL-4-mediated STAT -phosphorylation has been implicated as the mechanism involved in early and late stages of rheumatoid arthritis (RA). Up-regulation of proinflammatory cytokines in RA synovium and synovial fluid is a characteristic of the disease. It has been demostrated that IL-4-mediated activation of IL-4/STAT pathway is mediated through the Janus kinases (JAK 1 & 3) and that IL-4-associated JAK kinases are expressed in the RA synovium
[Muller-Ladner, et al, J. Immunol. 164 : 3894-3901 (2000)].
[00214] Familial amyotrophic lateral sclerosis (FALS) is a fatal neurodegenerative disorder affecting about 10% of ALS patients. The survival rates of FALS mice were increased upon treatment with a JAK3 specific inhibitor. This confirmed that JAK3 plays a role in FALS [Trieu, et al, Biochem. Biophys. Res. Commun. 267 : 22-25 (2000)].
[00215] Signal transducer and activator of transcription (STAT) proteins are activated by, among others, the JAK family kinases. Results form a recent study suggested the possibility of intervention in the JAK/STAT signaling pathway by targeting JAK family kinases with specific inhibitors for the treatment of leukemia [Sudbeck, et al., Clin. Cancer Res. 5 : 1569-1582 (1999)
]. JAK3 specific compounds were shown to inhibit the clonogenic growth of JAK3 -expressing cell lines DAUDI, RAMOS, LCl ; 19, NALM-6, MOLT-3 and HL-60. Inhibition of JAK3 and
TYK 2 abrogated tyrosine phosphorylation of STAT3, and inhibited cell growth of mycosis fungoides, a form of cutaneous T cell lymphoma.
[00216] According to another embodiment, the invention provides a method for treating or lessening the severity of a JAK3 -mediated disease or condition in a patient comprising the step of administering to said patient a composition according to the present invention. [00217] The term " J AK3 -mediated disease", as used herein means any disease or other deleterious condition in which a JAK3 kinase is known to play a role. Accordingly, another embodiment of the present invention relates to treating or lessening the severity of one or more diseases in which JAK3 is known to play a role. Specifically, the present invention relates to a method of treating or lessening the severity of a disease or condition selected from immune responses such as allergic or type I hypersensitivity reactions, asthma, autoimmune diseases such as transplant rejection, graft versus host disease, rheumatoid arthritis, amyotrophic lateral sclerosis, and multiple sclerosis, neurodegenerative disorders such as familial amyotrophic lateral sclerosis (FALS), as well as in solid and hematologic malignancies such as leukemias and lymphomas, wherein said method comprises administering to a patient in need thereof a composition according to the present invention.
[00218] The compounds and compositions, according to the method of the present invention, may be administered using any amount and any route of administration effective for treating or lessening the severity of cancer, an autoimmune disorder, a neurodegenerative or neurological disorder, schizophrenia, a bone-related disorder, liver disease, or a cardiac disorder. The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular agent, its mode of administration, and the like. 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. The term "patient", as used herein, means an animal, preferably a mammal, and most preferably a human. [00219] Pharmaceutically acceptable compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like, depending on the severity of the infection being treated. In certain embodiments, the compounds of the invention may be administered orally or parenterally at dosage levels of about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect.
[00220] 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-butylene 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. [00221] 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. [00222] 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. [00223] 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, delayed absorption of a parenterally administered compound form is accomplished by dissolving or suspending the compound in an oil vehicle. 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.
[00224] 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.
[00225] 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. [00226] 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.
[00227] 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. [00228] 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, ear drops, 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.
[00229] According to one embodiment, the invention relates to a method of inhibiting 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. [00230] According to another embodiment, the invention relates to a method of inhibiting ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3, or a mutant thereof, 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. In certain embodiments, the invention relates to a method of irreversibly inhibiting ErbBl, ErbB2, ErbB3, ErbB4, a TEC- kinase, and/or JAK3,, or a mutant thereof, 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.
[00231] The term "biological sample", as used herein, 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. [00232] Inhibition of protein kinase, or a protein kinase selected from ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3, or a mutant thereof, activity in a biological sample is useful for a variety of purposes that are known to one of skill in the art. Examples of such purposes include, but are not limited to, blood transfusion, organ transplantation, biological specimen storage, and biological assays.
[00233] Another embodiment of the present invention relates to a method of inhibiting protein 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.
[00234] According to another embodiment, the invention relates to a method of inhibiting one or more of ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3,, or a mutant thereof, activity in a patient comprising the step of administering to said patient a compound of the present invention, or a composition comprising said compound. According to certain embodiments, the invention relates to a method of irreversibly inhibiting one or more of ErbBl, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3,, or a mutant thereof, activity in a patient comprising the step of administering to said patient a compound of the present invention, or a composition comprising said compound. In other embodiments, the present invention provides a method for treating a disorder mediated by one or more of ErbBl, ErbB2, ErbB3, ErbB4, a TEC- kinase, and/or JAK3, or a mutant thereof, in a patient in need thereof, comprising the step of administering to said patient a compound according to the present invention or pharmaceutically acceptable composition thereof. Such disorders are described in detail herein. [00235] 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."
[00236] For example, compounds of the present invention, or a pharmaceutically acceptable composition thereof, are administered in combination with chemotherapeutic agents to treat proliferative diseases and cancer. Examples of known chemotherapeutic agents include, but are not limited to, Adriamycin, dexamethasone, vincristine, cyclophosphamide, fluorouracil, topotecan, taxol, interferons, platinum derivatives, taxane (e.g., paclitaxel), vinca alkaloids (e.g., vinblastine), anthracyclines (e.g., doxorubicin), epipodophyllotoxins (e.g., etoposide), cisplatin, an mTOR inhibitor (e.g., a rapamycin), methotrexate, actinomycin D, dolastatin 10, colchicine, emetine, trimetrexate, metoprine, cyclosporine, daunorubicin, teniposide, amphotericin, alkylating agents (e.g., chlorambucil), 5 -fluorouracil, campthothecin, cisplatin, metronidazole, and Gleevec™, among others. In other embodiments, a compound of the present invention is administered in combination with a biologic agent, such as Avastin or VECTIBIX. [00237] In certain embodiments, compounds of the present invention, or a pharmaceutically acceptable composition thereof, are administered in combination with an antiproliferative or chemotherapeutic agent selected from any one or more of abarelix, aldesleukin, alemtuzumab, alitretinoin, allopurinol, altretamine, amifostine, anastrozole, arsenic trioxide, asparaginase, azacitidine, BCG Live, bevacuzimab, fluorouracil, bexarotene, bleomycin, bortezomib, busulfan, calusterone, capecitabine, camptothecin, carboplatin, carmustine, celecoxib, cetuximab, chlorambucil, cladribine, clofarabine, cyclophosphamide, cytarabine, dactinomycin, darbepoetin alfa, daunorubicin, denileukin, dexrazoxane, docetaxel, doxorubicin (neutral), doxorubicin hydrochloride, dromostanolone propionate, epirubicin, epoetin alfa, erlotinib, estramustine, etoposide phosphate, etoposide, exemestane, filgrastim, floxuridine fludarabine, Mvestrant, gefitinib, gemcitabine, gemtuzumab, goserelin acetate, histrelin acetate, hydroxyurea, ibritumomab, idarubicin, ifosfamide, imatinib mesylate, interferon alfa-2a, interferon alfa-2b, irinotecan, lenalidomide, letrozole, leucovorin, leuprolide acetate, levamisole, lomustine, megestrol acetate, melphalan, mercaptopurine, 6-MP, mesna, methotrexate, methoxsalen, mitomycin C, mitotane, mitoxantrone, nandrolone, nelarabine, nofetumomab, oprelvekin, oxaliplatin, paclitaxel, palifermin, pamidronate, pegademase, pegaspargase, pegfilgrastim, pemetrexed disodium, pentostatin, pipobroman, plicamycin, porfimer sodium, procarbazine, quinacrine, rasburicase, rituximab, sargramostim, sorafenib, streptozocin, sunitinib maleate, talc, tamoxifen, temozolomide, teniposide, VM-26, testolactone, thioguanine, 6-TG, thiotepa, topotecan, toremifene, tositumomab, trastuzumab, tretinoin, ATRA, uracil mustard, valrubicin, vinblastine, vincristine, vinorelbine, zoledronate, or zoledronic acid.
[00238] Other examples of agents the inhibitors of this invention may also be combined with include, without limitation: treatments for Alzheimer's Disease such as donepezil hydrochloride (Aricept®) and rivastigmine (Exelon®); treatments for Parkinson's Disease such as L- DOPA/carbidopa, entacapone, ropinrole, pramipexole, bromocriptine, pergolide, trihexephendyl, and amantadine; agents for treating Multiple Sclerosis (MS) such as beta interferon (e.g., Avonex® and Rebif®), glatiramer acetate (Copaxone®), and mitoxantrone; treatments for asthma such as albuterol and montelukast (Singulair®); agents for treating schizophrenia such as zyprexa, risperdal, seroquel, and haloperidol; anti-inflammatory agents such as corticosteroids, TNF blockers, IL-I RA, azathioprine, cyclophosphamide, and sulfasalazine; immunomodulatory and immunosuppressive agents such as cyclosporin, tacrolimus, rapamycin, mycophenolate mofetil, interferons, corticosteroids, cyclophophamide, azathioprine, and sulfasalazine; neurotrophic factors such as acetylcholinesterase inhibitors, MAO inhibitors, interferons, anticonvulsants, ion channel blockers, riluzole, and anti-Parkinsonian agents; agents for treating cardiovascular disease such as beta-blockers, ACE inhibitors, diuretics, nitrates, calcium channel blockers, and statins; agents for treating liver disease such as corticosteroids, cholestyramine, interferons, and anti-viral agents; agents for treating blood disorders such as corticosteroids, antileukemic agents, and growth factors; and agents for treating immunodeficiency disorders such as gamma globulin. [00239] In certain embodiments, compounds of the present invention, or a pharmaceutically acceptable composition thereof, are administered in combination with a monoclonal antibody or an siRNA therapeutic.
[00240] Those additional agents may be administered separately from an inventive compound-containing composition, as part of a multiple dosage regimen. Alternatively, those agents may be part of a single dosage form, mixed together with a compound of this invention in a single composition. If administered as part of a multiple dosage regime, the two active agents may be submitted simultaneously, sequentially or within a period of time from one another normally within five hours from one another.
[00241] As used herein, the term "combination," "combined," and related terms refers to the simultaneous or sequential administration of therapeutic agents in accordance with this invention. For example, a compound of the present invention may be administered with another therapeutic agent simultaneously or sequentially in separate unit dosage forms or together in a single unit dosage form. Accordingly, the present invention provides a single unit dosage form comprising a provided compound, an additional therapeutic agent, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
[00242] The amount of both, an inventive compound and 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, compositions of this invention should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of an inventive can be administered.
[00243] In those compositions which 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 - 1,000 μg/kg body weight/day of the additional therapeutic agent can be administered. [00244] 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.
[00245] 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. Implantable devices coated with a compound of this invention are another embodiment of the present invention. 5. Probe Compounds
[00246] In certain aspects, a compound of the present invention may be tethered to a detectable moiety to form a probe compound. In one aspect, a probe compound of the invention comprises an irreversible protein kinase inhibitor of formula I-a or I-b, as described herein, a detectable moiety, and a tethering moiety that attaches the inhibitor to the detectable moiety. [00247] In some embodiments, such probe compounds of the present invention comprise a provided compound of formula I-a or I-b tethered to a detectable moiety, R1, by a bivalent tethering moiety, -T-. The tethering moiety may be attached to a compound of formula I-a or I- b via Ring A, Ring B, or R1. One of ordinary skill in the art will appreciate that when a tethering moiety is attached to R1, R1 is a bivalent warhead group denoted as R1 . In certain embodiments, a provided probe compound is selected from any of formula V-a, V-b, VI-a, VI-b, VII-a, or VII-b:
Figure imgf000126_0001
V-a V-b
Figure imgf000127_0001
VII-a VII-b, wherein each of Ring A, Ring B, R1, m, p, Rx, Ry, RV,W1, and W2 is as defined above with respect to formulae I-a and I-b, and described in classes and subclasses herein, R1 is a bivalent warhead group, T is a bivalent tethering moiety; and R1 is a detectable moiety. [00248] In some embodiments, R1 is a detectable moiety selected from a primary label or a secondary label. In certain embodiments, R1 is a detectable moiety selected from a fluorescent label (e.g., a fluorescent dye or a fluorophore), a mass-tag, a chemiluminescent group, a chromophore, an electron dense group, or an energy transfer agent.
[00249] As used herein, the term "detectable moiety" is used interchangeably with the term "label" and "reporter" and relates to any moiety capable of being detected, e.g., primary labels and secondary labels. A presence of a detectable moiety can be measured using methods for quantifying (in absolute, approximate or relative terms) the detectable moiety in a system under study. In some embodiments, such methods are well known to one of ordinary skill in the art and include any methods that quantify a reporter moiety (e.g., a label, a dye, a photocrosslinker, a cytotoxic compound, a drug, an affinity label, a photoaffinity label, a reactive compound, an antibody or antibody fragment, a biomaterial, a nanoparticle, a spin label, a fluorophore, a metal- containing moiety, a radioactive moiety, quantum dot(s), a novel functional group, a group that covalently or noncovalently interacts with other molecules, a photocaged moiety, an actinic radiation excitable moiety, a ligand, a photoisomerizable moiety, biotin, a biotin analog (e.g., biotin sulfoxide), a moiety incorporating a heavy atom, a chemically cleavable group, a photocleavable group, a redox-active agent, an isotopically labeled moiety, a biophysical probe, a phosphorescent group, a chemiluminescent group, an electron dense group, a magnetic group, an intercalating group, a chromophore, an energy transfer agent, a biologically active agent, a detectable label, and any combination of the above).
[00250] Primary labels, such as radioisotopes (e.g., tritium, 32P, 33P, 35S, 14C, 123I, 124I, 125I, or 131I), mass-tags including, but not limited to, stable isotopes (e.g., 13C, 2H, 17O, 18O, 15N, 19F, and 127I), positron emitting isotopes (e.g., 11C, 18F, 13N, 124I, and 15O), and fluorescent labels are signal generating reporter groups which can be detected without further modifications. Detectable moities may be analyzed by methods including, but not limited to fluorescence, positron emission tomography, SPECT medical imaging, chemiluminescence, electron-spin resonance, ultraviolet/visible absorbance spectroscopy, mass spectrometry, nuclear magnetic resonance, magnetic resonance, flow cytometry, autoradiography, scintillation counting, phosphoimaging, and electrochemical methods.
[00251] The term "secondary label" as used herein refers to moieties such as biotin and various protein antigens that require the presence of a second intermediate for production of a detectable signal. For biotin, the secondary intermediate may include streptavidin-enzyme conjugates. For antigen labels, secondary intermediates may include antibody-enzyme conjugates. Some fluorescent groups act as secondary labels because they transfer energy to another group in the process of nonradiative fluorescent resonance energy transfer (FRET), and the second group produces the detected signal.
[00252] The terms "fluorescent label", "fluorescent dye", and "fluorophore" as used herein refer to moieties that absorb light energy at a defined excitation wavelength and emit light energy at a different wavelength. Examples of fluorescent labels include, but are not limited to: Alexa Fluor dyes (Alexa Fluor 350, Alexa Fluor 488, Alexa Fluor 532, Alexa Fluor 546, Alexa Fluor 568, Alexa Fluor 594, Alexa Fluor 633, Alexa Fluor 660 and Alexa Fluor 680), AMCA, AMCA-S, BODIPY dyes (BODIPY FL, BODIPY R6G, BODIPY TMR, BODIPY TR, BODIPY 493/503, BODIPY 530/550, BODIPY 558/568, BODIPY 564/570, BODIPY 576/589, BODIPY 581/591, BODIPY 630/650, BODIPY 650/665), Carboxyrhodamine 6G, carboxy-X-rhodamine (ROX), Cascade Blue, Cascade Yellow, Coumarin 343, Cyanine dyes (Cy3, Cy5, Cy3.5, Cy5.5), Dansyl, Dapoxyl, Dialkylaminocoumarin, 4',5'-Dichloro-2',7'-dimethoxy-fluorescein, DM- NERF, Eosin, Erythrosin, Fluorescein, FAM, Hydroxycoumarin, IRDyes (IRD40, IRD 700, IRD 800), JOE, Lissamine rhodamine B, Marina Blue, Methoxycoumarin, Naphthofluorescein, Oregon Green 488, Oregon Green 500, Oregon Green 514, Pacific Blue, PyMPO, Pyrene, Rhodamine B, Rhodamine 6G, Rhodamine Green, Rhodamine Red, Rhodol Green, 2',4',5',7'- Tetra-bromosulfone-fluorescein, Tetramethyl-rhodamine (TMR),
Carboxytetramethylrhodamine (TAMRA), Texas Red, Texas Red-X, 5(6)-Carboxyfluorescein, 2,7-Dichlorofluorescein, N,N-Bis(2,4,6-trimethylphenyl)-3,4:9,10-perylenebis(dicarboximide, HPTS, Ethyl Eosin, DY-490XL MegaStokes, DY-485XL MegaStokes, Adirondack Green 520, ATTO 465, ATTO 488, ATTO 495, Y0Y0-l,5-FAM, BCECF, dichlorofluorescein, rhodamine 110, rhodamine 123, YO-PRO-I, SYTOX Green, Sodium Green, SYBR Green I, Alexa Fluor 500, FITC, Fluo-3, Fluo-4, fluoro-emerald, YoYo-I ssDNA, YoYo-I dsDNA, YoYo-I, SYTO RNASelect, Diversa Green-FP, Dragon Green, EvaGreen, Surf Green EX, Spectrum Green, NeuroTrace 500525, NBD-X, MitoTracker Green FM, LysoTracker Green DND-26, CBQCA, PA-GFP (post-activation), WEGFP (post-activation), F1ASH-CCXXCC, Azami Green monomeric, Azami Green, green fluorescent protein (GFP), EGFP (Campbell Tsien 2003), EGFP (Patterson 2001), Kaede Green, 7-Benzylamino-4-Nitrobenz-2-Oxa-l,3-Diazole, Bexl, Doxorubicin, Lumio Green, and SuperGlo GFP.
[00253] The term "mass-tag" as used herein refers to any moiety that is capable of being uniquely detected by virtue of its mass using mass spectrometry (MS) detection techniques. Examples of mass-tags include electrophore release tags such as N-[3-[4'-[(p- Methoxytetrafluorobenzyl)oxy]phenyl]-3-methylglyceronyl]isonipecotic Acid, 4 ' -[2,3 ,5 ,6- Tetrafluoro-4-(pentafluorophenoxyl)]methyl acetophenone, and their derivatives. The synthesis and utility of these mass-tags is described in United States Patents 4,650,750, 4,709,016, 5,360,8191, 5,516,931, 5,602,273, 5,604,104, 5,610,020, and 5,650,270. Other examples of mass-tags include, but are not limited to, nucleotides, dideoxynucleotides, oligonucleotides of varying length and base composition, oligopeptides, oligosaccharides, and other synthetic polymers of varying length and monomer composition. A large variety of organic molecules, both neutral and charged (biomolecules or synthetic compounds) of an appropriate mass range (100-2000 Daltons) may also be used as mass-tags. Stable isotopes (e.g., 13C, 2H, 17O, 18O, and 15N) may also be used as mass-tags.
[00254] The term "chemiluminescent group," as used herein, refers to a group which emits light as a result of a chemical reaction without the addition of heat. By way of example, luminol (5-amino-2,3-dihydro-l,4-phthalazinedione) reacts with oxidants like hydrogen peroxide (H2O2) in the presence of a base and a metal catalyst to produce an excited state product (3- aminophthalate, 3-APA).
[00255] The term "chromophore," as used herein, refers to a molecule which absorbs light of visible wavelengths, UV wavelengths or IR wavelengths.
[00256] The term "dye," as used herein, refers to a soluble, coloring substance which contains a chromophore.
[00257] The term "electron dense group," as used herein, refers to a group which scatters electrons when irradiated with an electron beam. Such groups include, but are not limited to, ammonium molybdate, bismuth subnitrate, cadmium iodide, carbohydrazide, ferric chloride hexahydrate, hexamethylene tetramine, indium trichloride anhydrous, lanthanum nitrate, lead acetate trihydrate, lead citrate trihydrate, lead nitrate, periodic acid, phosphomolybdic acid, phosphotungstic acid, potassium ferricyanide, potassium ferrocyanide, ruthenium red, silver nitrate, silver proteinate (Ag Assay: 8.0-8.5%) "Strong", silver tetraphenylporphin (S-TPPS), sodium chloroaurate, sodium tungstate, thallium nitrate, thiosemicarbazide (TSC), uranyl acetate, uranyl nitrate, and vanadyl sulfate.
[00258] The term "energy transfer agent," as used herein, refers to a molecule which either donates or accepts energy from another molecule. By way of example only, fluorescence resonance energy transfer (FRET) is a dipole-dipole coupling process by which the excited-state energy of a fluorescence donor molecule is non-radiatively transferred to an unexcited acceptor molecule which then fluorescently emits the donated energy at a longer wavelength. [00259] The term "moiety incorporating a heavy atom," as used herein, refers to a group which incorporates an ion of atom which is usually heavier than carbon. In some embodiments, such ions or atoms include, but are not limited to, silicon, tungsten, gold, lead, and uranium. [00260] The term "photoaffinity label," as used herein, refers to a label with a group, which, upon exposure to light, forms a linkage with a molecule for which the label has an affinity. [00261] The term "photocaged moiety," as used herein, refers to a group which, upon illumination at certain wavelengths, covalently or non-covalently binds other ions or molecules. [00262] The term "photoisomerizable moiety," as used herein, refers to a group wherein upon illumination with light changes from one isomeric form to another. [00263] The term "radioactive moiety," as used herein, refers to a group whose nuclei spontaneously give off nuclear radiation, such as alpha, beta, or gamma particles; wherein, alpha particles are helium nuclei, beta particles are electrons, and gamma particles are high energy photons.
[00264] The term "spin label," as used herein, refers to molecules which contain an atom or a group of atoms exhibiting an unpaired electron spin (i.e. a stable paramagnetic group) that in some embodiments are detected by electron spin resonance spectroscopy and in other embodiments are attached to another molecule. Such spin-label molecules include, but are not limited to, nitryl radicals and nitroxides, and in some embodiments are single spin-labels or double spin-labels.
[00265] The term "quantum dots," as used herein, refers to colloidal semiconductor nanocrystals that in some embodiments are detected in the near-infrared and have extremely high quantum yields (i.e., very bright upon modest illumination).
[00266] One of ordinary skill in the art will recognize that a detectable moiety may be attached to a provided compound via a suitable substituent. As used herein, the term "suitable substituent" refers to a moiety that is capable of covalent attachment to a detectable moiety. Such moieties are well known to one of ordinary skill in the art and include groups containing, e.g., a carboxylate moiety, an amino moiety, a thiol moiety, or a hydroxyl moiety, to name but a few. It will be appreciated that such moieties may be directly attached to a provided compound or via a tethering moiety, such as a bivalent saturated or unsaturated hydrocarbon chain. [00267] In some embodiments, detectable moieties are attached to a provided compound via click chemistry. In some embodiments, such moieties are attached via a 1,3-cycloaddition of an azide with an alkyne, optionally in the presence of a copper catalyst. Methods of using click chemistry are known in the art and include those described by Rostovtsev et al. , Angew. Chem. Int. Ed. 2002, 41, 2596-99 and Sun et al., Bioconjugate Chem., 2006, JJ, 52-57. In some embodiments, a click ready inhibitor moiety is provided and reacted with a click ready -T-R1 moiety. As used herein, "click ready" refers to a moiety containing an azide or alkyne for use in a click chemistry reaction. In some embodiments, the click ready inhibitor moiety comprises an azide. In certain embodiments, the click ready -T-R1 moiety comprises a strained cyclooctyne for use in a copper-free click chemistry reaction (for example, using methods described in Baskin et al., Proc. Natl. Acad. Sci. USA 2007, 104, 16793-16797). [00268] In certain embodiments, the click ready inhibitor moiety is of one of the following formulae:
Figure imgf000132_0001
wherein Ring A, Ring B, W1, W2, Ry, Rv, p, Rx, and m are as defined above with respect to Formula I and described herein, and q is 1, 2, or 3. [00269] Exemplary click ready inhibitors include:
Figure imgf000132_0002
[00270] In some embodiments, the click ready -T-R1 moiety is of formula:
Figure imgf000133_0001
[00271] An exemplary reaction in which a click ready inhibitor moiety and a click ready -T- R1 moiety are joined through a [2+3]-cycloaddition is as follows
Figure imgf000133_0002
[00272] In some embodiments, the detectable moiety, R1, is selected from a label, a dye, a photocrosslinker, a cytotoxic compound, a drug, an affinity label, a photoaffmity label, a reactive compound, an antibody or antibody fragment, a biomatenal, a nanoparticle, a spin label, a fluorophore, a metal-contammg moiety, a radioactive moiety, quantum dot(s), a novel functional group, a group that covalently or noncovalently interacts with other molecules, a photocaged moiety, an actinic radiation excitable moiety, a ligand, a photoisomerizable moiety, biotm, a biotin analog (e g , biotm sulfoxide), a moiety incorporating a heavy atom, a chemically cleavable group, a photocleavable group, a redox-active agent, an isotopically labeled moiety, a biophysical probe, a phosphorescent group, a chemilummescent group, an electron dense group, a magnetic group, an intercalating group, a chromophore, an energy transfer agent, a biologically active agent, a detectable label, or a combination thereof
[00273] In some embodiments, R1 is biotm or an analog thereof In certain embodiments, R1 is biotm In certain other embodiments, R1 is biotm sulfoxide
[00274] In another embodiment, R1 is a fluorophore In a further embodiment, the fluorophore is selected from Alexa Fluor dyes (Alexa Fluor 350, Alexa Fluor 488, Alexa Fluor 532, Alexa Fluor 546, Alexa Fluor 568, Alexa Fluor 594, Alexa Fluor 633, Alexa Fluor 660 and Alexa Fluor 680), AMCA, AMCA-S, BODIPY dyes (BODIPY FL, BODIPY R6G, BODIPY TMR, BODIPY TR, BODIPY 493/503, BODIPY 530/550, BODIPY 558/568, BODIPY 564/570, BODIPY 576/589, BODIPY 581/591, BODIPY 630/650, BODIPY 650/665), Carboxyrhodamine 6G, carboxy-X-rhodamine (ROX), Cascade Blue, Cascade Yellow, Coumarin 343, Cyanine dyes (Cy3, Cy5, Cy3.5, Cy5.5), Dansyl, Dapoxyl, Dialkylaminocoumarin, 4',5'-Dichloro-2',7'- dimethoxy-fluorescein, DM-NERF, Eosin, Erythrosin, Fluorescein, FAM, Hydroxycoumarin, IRDyes (IRD40, IRD 700, IRD 800), JOE, Lissamine rhodamine B, Marina Blue, Methoxycoumarin, Naphthofluorescein, Oregon Green 488, Oregon Green 500, Oregon Green 514, Pacific Blue, PyMPO, Pyrene, Rhodamine B, Rhodamine 6G, Rhodamine Green, Rhodamine Red, Rhodol Green, 2',4',5',7'-Tetra-bromosulfone-fluorescein, Tetramethyl- rhodamine (TMR), Carboxytetramethylrhodamine (TAMRA), Texas Red, Texas Red-X, 5(6)- Carboxyfluorescein, 2,7-Dichlorofluorescein, N,N-Bis(2,4,6-trimethylphenyl)-3,4:9,10- perylenebis(dicarboximide, HPTS, Ethyl Eosin, DY-490XL MegaStokes, DY-485XL MegaStokes, Adirondack Green 520, ATTO 465, ATTO 488, ATTO 495, Y0Y0-l,5-FAM, BCECF, dichlorofluorescein, rhodamine 110, rhodamine 123, YO-PRO-I, SYTOX Green, Sodium Green, SYBR Green I, Alexa Fluor 500, FITC, Fluo-3, Fluo-4, fluoro-emerald, YoYo-I ssDNA, YoYo-I dsDNA, YoYo-I, SYTO RNASelect, Diversa Green-FP, Dragon Green, EvaGreen, Surf Green EX, Spectrum Green, NeuroTrace 500525, NBD-X, MitoTracker Green FM, LysoTracker Green DND-26, CBQCA, PA-GFP (post-activation), WEGFP (post- activation), FIASH-CCXXCC, Azami Green monomeric, Azami Green, green fluorescent protein (GFP), EGFP (Campbell Tsien 2003), EGFP (Patterson 2001), Kaede Green, 7- Benzylamino-4-Nitrobenz-2-Oxa-l,3-Diazole, Bexl, Doxorubicin, Lumio Green, or SuperGlo GFP.
[00275] As described generally above, a provided probe compound comprises a tethering moiety, -T-, that attaches the irreversible inhibitor to the detectable moiety. As used herein, the term "tether" or "tethering moiety" refers to any bivalent chemical spacer including, but not limited to, a covalent bond, a polymer, a water soluble polymer, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted heterocycloalkylalkyl, optionally substituted heterocycloalkylalkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkylalkenylalkyl, an optionally substituted amide moiety, an ether moiety, an ketone moiety, an ester moiety, an optionally substituted carbamate moiety, an optionally substituted hydrazone moiety, an optionally substituted hydrazine moiety, an optionally substituted oxime moiety, a disulfide moiety, an optionally substituted imine moiety, an optionally substituted sulfonamide moiety, a sulfone moiety, a sulfoxide moiety, a thioether moiety, or any combination thereof.
[00276] In some embodiments, the tethering moiety, -T-, is selected from a covalent bond, a polymer, a water soluble polymer, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkylalkyl, optionally substituted heterocycloalkylalkenyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted heterocycloalkylalkenylalkyl. In some embodiments, the tethering moiety is an optionally substituted heterocycle. In other embodiments, the heterocycle is selected from aziridine, oxirane, episulfide, azetidine, oxetane, pyrroline, tetrahydrofuran, tetrahydrothiophene, pyrrolidine, pyrazole, pyrrole, imidazole, triazole, tetrazole, oxazole, isoxazole, oxirene, thiazole, isothiazole, dithiolane, furan, thiophene, piperidine, tetrahydropyran, thiane, pyridine, pyran, thiapyrane, pyridazine, pyrimidine, pyrazine, piperazine, oxazine, thiazine, dithiane, and dioxane. In some embodiments, the heterocycle is piperazine. In further embodiments, the tethering moiety is optionally substituted with halogen, -CN, -OH, -NO2, alkyl, S(O), and S(O)2. In other embodiments, the water soluble polymer is a PEG group.
[00277] In other embodiments, the tethering moiety provides sufficient spatial separation between the detectable moiety and the protein kinase inhibitor moiety. In further embodiments, the tethering moiety is stable. In yet a further embodiment, the tethering moiety does not substantially affect the response of the detectable moiety. In other embodiments, the tethering moiety provides chemical stability to the probe compound. In further embodiments, the tethering moiety provides sufficient solubility to the probe compound.
[00278] In some embodiments, a tethering moiety, -T-, such as a water soluble polymer is coupled at one end to a provided irreversible inhibitor and to a detectable moiety, R1, at the other end. In other embodiments, a water soluble polymer is coupled via a functional group or substituent of the provided irreversible inhibitor. In further embodiments, a water soluble polymer is coupled via a functional group or substituent of the reporter moiety. [00279] In some embodiments, examples of hydrophilic polymers, for use in tethering moiety -T-, include, but are not limited to: polyalkyl ethers and alkoxy-capped analogs thereof (e.g., polyoxyethylene glycol, polyoxyethylene/propylene glycol, and methoxy or ethoxy-capped analogs thereof, polyoxyethylene glycol, the latter is also known as polyethylene glycol or PEG); polyvinylpyrrolidones; polyvinylalkyl ethers; polyoxazolines, polyalkyl oxazolines and polyhydroxyalkyl oxazolines; polyacrylamides, polyalkyl acrylamides, and polyhydroxyalkyl acrylamides (e.g., polyhydroxypropylmethacrylamide and derivatives thereof); polyhydroxyalkyl acrylates; polysialic acids and analogs thereof, hydrophilic peptide sequences; polysaccharides and their derivatives, including dextran and dextran derivatives, e.g., carboxymethyldextran, dextran sulfates, aminodextran; cellulose and its derivatives, e.g., carboxymethyl cellulose, hydroxyalkyl celluloses; chitin and its derivatives, e.g., chitosan, succinyl chitosan, carboxymethylchitin, carboxymethylchitosan; hyaluronic acid and its derivatives; starches; alginates; chondroitin sulfate; albumin; pullulan and carboxymethyl pullulan; polyaminoacids and derivatives thereof, e.g., polyglutamic acids, polylysines, polyaspartic acids, polyaspartamides; maleic anhydride copolymers such as: styrene maleic anhydride copolymer, divinylethyl ether maleic anhydride copolymer; polyvinyl alcohols; copolymers thereof, terpolymers thereof, mixtures thereof, and derivatives of the foregoing. In other embodiments, a water soluble polymer is any structural form including but not limited to linear, forked or branched. In further embodiments, multifunctional polymer derivatives include, but are not limited to, linear polymers having two termini, each terminus being bonded to a functional group which is the same or different.
[00280] In some embodiments, a water polymer comprises a poly(ethylene glycol) moiety. In further embodiments, the molecular weight of the polymer is of a wide range, including but not limited to, between about 100 Da and about 100,000 Da or more. In yet further embodiments, the molecular weight of the polymer is between about 100 Da and about 100,000 Da, including but not limited to, about 100,000 Da, about 95,000 Da, about 90,000 Da, about 85,000 Da, about 80,000 Da, about 75,000 Da, about 70,000 Da, about 65,000 Da, about 60,000 Da, about 55,000 Da, about 50,000 Da, about 45,000 Da, about 40,000 Da, about 35,000 Da, 30,000 Da, about 25,000 Da, about 20,000 Da, about 15,000 Da, about 10,000 Da, about 9,000 Da, about 8,000 Da, about 7,000 Da, about 6,000 Da, about 5,000 Da, about 4,000 Da, about 3,000 Da, about 2,000 Da, about 1,000 Da, about 900 Da, about 800 Da, about 700 Da, about 600 Da, about 500 Da, about 400 Da, about 300 Da, about 200 Da, and about 100 Da. In some embodiments, the molecular weight of the polymer is between about 100 Da and 50,000 Da. In some embodiments, the molecular weight of the polymer is between about 100 Da and 40,000 Da. In some embodiments, the molecular weight of the polymer is between about 1,000 Da and 40,000 Da. In some embodiments, the molecular weight of the polymer is between about 5,000 Da and 40,000 Da. In some embodiments, the molecular weight of the polymer is between about 10,000 Da and 40,000 Da. In some embodiments, the poly(ethylene glycol) molecule is a branched polymer. In further embodiments, the molecular weight of the branched chain PEG is between about 1,000 Da and about 100,000 Da, including but not limited to, about 100,000 Da, about 95,000 Da, about 90,000 Da, about 85,000 Da, about 80,000 Da, about 75,000 Da, about 70,000 Da, about 65,000 Da, about 60,000 Da, about 55,000 Da, about 50,000 Da, about 45,000 Da, about 40,000 Da, about 35,000 Da, about 30,000 Da, about 25,000 Da, about 20,000 Da, about 15,000 Da, about 10,000 Da, about 9,000 Da, about 8,000 Da, about 7,000 Da, about 6,000 Da, about 5,000 Da, about 4,000 Da, about 3,000 Da, about 2,000 Da, and about 1,000 Da. In some embodiments, the molecular weight of a branched chain PEG is between about 1,000 Da and about 50,000 Da. In some embodiments, the molecular weight of a branched chain PEG is between about 1,000 Da and about 40,000 Da. In some embodiments, the molecular weight of a branched chain PEG is between about 5,000 Da and about 40,000 Da. In some embodiments, the molecular weight of a branched chain PEG is between about 5,000 Da and about 20,000 Da. The foregoing list for substantially water soluble backbones is by no means exhaustive and is merely illustrative, and in some embodiments, polymeric materials having the qualities described above are suitable for use in methods and compositions described herein.
[00281] One of ordinary skill in the art will appreciate that when -T-R1 is attached to a compound of formula I-a or I-b via the R1 warhead group, then the resulting tethering moiety comprises the R1 warhead group. As used herein, the phrase "comprises a warhead group" means that the tethering moiety formed by -R1 -T- of formula V-a or V-b is either substituted with a warhead group or has such a warhead group incorporated within the tethering moiety. For example, the tethering moiety formed by -R1 -T- may be substituted with an -L-Y warhead group, wherein such groups are as described herein. Alternatively, the tethering moiety formed by -R1 -T- has the appropriate features of a warhead group incorporated within the tethering moiety. For example, the tethering moiety formed by -R1 -T- may include one or more units of unsaturation and optional substituents and/or heteroatoms which, in combination, result in a moiety that is capable of covalently modifying a protein kinase in accordance with the present invention. Such -R1 -T- tethering moiety are depicted below.
[00282] In some embodiments, a methylene unit of an -R1 -T- tethering moiety is replaced by a bivalent -L-Y'- moiety to provide a compound of formula V-a-«7 or V-b-MΪ:
Figure imgf000138_0001
\ -a.-iii V-b-iϊϊ wherein each of Ring A, Ring B, m, p, Rx, Ry,
Figure imgf000138_0002
W2, T, L, Y', and R1 is as defined above and described in classes and subclasses herein and Y' is a bivalent version of the Y group defined above and described in classes and subclasses herein.
[00283] In some embodiments, a methylene unit of an -R1 -T- tethering moiety is replaced by an -L(Y)- moiety to provide a compound of formula V-a-iv or V-b-/v:
Figure imgf000138_0003
wherein each of Ring A, Ring B, m, p, Rx, Ry,
Figure imgf000138_0004
W2, T, L, Y, and R1 is as defined above and described in classes and subclasses herein.
[00284] In some embodiments, a tethering moiety is substituted with an L-Y moiety to provide a compound of formula V-a-v or V-b-v:
Figure imgf000139_0001
wherein each of Ring A, Ring B, m, p, Rx, Ry,
Figure imgf000139_0002
W2, T, L, Y, and R1 is as defined above and described in classes and subclasses herein.
[00285] In certain embodiments, the tethering moiety, -T-, has one of the following structures:
Figure imgf000139_0006
[00286] In some embodiments, the tethering moiety, -T-, has the following structure:
Figure imgf000139_0003
[00287] In other embodiments, the tethering moiety, -T-, has the following structure:
Figure imgf000139_0007
[00288] In certain other embodiments, the tethering moiety, -T-, has the following structure:
Figure imgf000139_0004
[00289] In yet other embodiments, the tethering moiety, -T-, has the following structure:
Figure imgf000139_0005
[00290] In some embodiments, the tethering moiety, -T-, has the following structure:
Figure imgf000140_0001
[00291] In some embodiments, -T-R1 is of the following structure:
Figure imgf000140_0002
[00292] In other embodiments, -T-R1 is of the following structure:
Figure imgf000140_0003
[00293] In certain embodiments, -T-R1 is of the following structure:
Figure imgf000140_0004
[00294] In some embodiments, a probe compound of formula V-a, V-b, VI-a, VI-b, VII-a, or
VII-b is derived from any compound of Table 5.
[00295] In certain embodiments, the probe compound is one of the following structures:
Figure imgf000141_0001
1-363
Figure imgf000142_0001
Figure imgf000143_0001
1-368.
[00296] It will be appreciated that many -T-R1 reagents are commercially available. For example, numerous biotinylating reagents are available from, e.g., Thermo Scientific having varying tether lengths. Such reagents include NHS-PEG4-Biotin and NHS-PEGi2-Biotin. [00297] In some embodiments, analogous probe structures to the ones exemplified above are prepared using click-ready inhibitor moieties and click-ready -T-R1 moieties, as described herein.
[00298] In some embodiments, a provided probe compound covalently modifies a phosphorylated conformation of a protein kinase. In one aspect, the phosphorylated conformation of the protein kinase is either an active or inactive form of the protein kinase. In certain embodiments, the phosphorylated conformation of the protein kinase is an active form of said kinase. In certain embodiments, the probe compound is cell permeable. [00299] In some embodiments, the present invention provides a method for determining occupancy of a protein kinase by a provided irreversible inhibitor (i.e., a compound of formula I- a or I-b) in a patient, comprising providing one or more tissues, cell types, or a lysate thereof, obtained from a patient administered at least one dose of a compound of said irreversible inhibitor, contacting said tissue, cell type or lysate thereof with a probe compound (i.e., a compound of formula V-a, V-b, VI-a, VI-b, VII-a, or VII-b) to covalent modify at least one protein kinase present in said lysate, and measuring the amount of said protein kinase covalently modified by the probe compound to determine occupancy of said protein kinase by said compound of formula I-a or I-b as compared to occupancy of said protein kinase by said probe compound. In certain embodiments, the method further comprises the step of adjusting the dose of the compound of formula I-a or I-b to increase occupancy of the protein kinase. In certain other embodiments, the method further comprises the step of adjusting the dose of the compound of formula I-a or I-b to decrease occupancy of the protein kinase.
[00300] As used herein, the terms "occupancy" or "occupy" refer to the extent to which a protein kinase is modified by a provided covalent inhibitor compound. One of ordinary skill in the art would appreciate that it is desirable to administer the lowest dose possible to achieve the desired efficacious occupancy of the protein kinase.
[00301] In some embodiments, the protein kinase to be modified is BTK. In other embodiments, the protein kinase to be modified is EGFR. In certain embodiments, the protein kinase is JAK. In certain other embodiments, the protein kinase is one or more of ErbBl, ErbB2, or ErbB4. In yet other embodiments, the protein kinase is TEC, ITK, or BMX.
[00302] In some embodiments, the probe compound comprises the irreversible inhibitor for which occupancy is being determined.
[00303] In some embodiments, the present invention provides a method for assessing the efficacy of a provided irreversible inhibitor in a mammal, comprising administering a provided irreversible inhibitor to the mammal, administering a provided probe compound to tissues or cells isolated from the mammal, or a lysate thereof, measuring the activity of the detectable moiety of the probe compound, and comparing the activity of the detectable moiety to a standard.
[00304] In other embodiments, the present invention provides a method for assessing the pharmacodynamics of a provided irreversible inhibitor in a mammal, comprising administering a provided irreversible inhibitor to the mammal, administering a probe compound presented herein to one or more cell types, or a lysate thereof, isolated from the mammal, and measuring the activity of the detectable moiety of the probe compound at different time points following the administration of the inhibitor.
[00305] In yet other embodiments, the present invention provides a method for in vitro labeling of a protein kinase comprising contacting said protein kinase with a probe compound described herein. In one embodiment, the contacting step comprises incubating the protein kinase with a probe compound presented herein.
[00306] In certain embodiments, the present invention provides a method for in vitro labeling of a protein kinase comprising contacting one or more cells or tissues, or a lysate thereof, expressing the protein kinase with a probe compound described herein.
[00307] In certain other embodiments, the present invention provides a method for detecting a labeled protein kinase comprising separating proteins, the proteins comprising a protein kinase labeled by probe compound described herein, by electrophoresis and detecting the probe compound by fluorescence.
[00308] In some embodiments, the present invention provides a method for assessing the pharmacodynamics of a provided irreversible inhibitor in vitro, comprising incubating the provided irreversible inhibitor with the target protein kinase, adding the probe compound presented herein to the target protein kinase, and determining the amount of target modified by the probe compound.
[00309] In certain embodiments, the probe compound is detected by binding to avidin, streptavidin, neutravidin, or captavidin.
[00310] In some embodiments, the probe is detected by Western blot. In other embodiments, the probe is detected by ELISA. In certain embodiments, the probe is detected by flow cytometry.
[00311] In other embodiments, the present invention provides a method for probing the kinome with irreversible inhibitors comprising incubating one or more cell types, or a lysate thereof, with a biotinylated probe compound to generate proteins modified with a biotin moiety, digesting the proteins, capturing with avidin or an analog thereof, and performing multidimensional LC-MS-MS to identify protein kinases modified by the probe compound and the adduction sites of said kinases.
[00312] In certain embodiments, the present invention provides a method for measuring protein synthesis in cells comprising incubating cells with an irreversible inhibitor of the target protein, forming lysates of the cells at specific time points, and incubating said cell lysates with an inventive probe compound to measure the appearance of free protein over an extended period of time. [00313] In other embodiments, the present invention provides a method for determining a dosing schedule in a mammal for maximizing occupancy of a target protein kinase comprising assaying a one or more cell types, or a lysate thereof, isolated from the mammal, (derived from, e.g., splenocytes, peripheral B cells, whole blood, lymph nodes, intestinal tissue, or other tissues) from a mammal administered a provided irreversible inhibitor of formula I-a or I-b, wherein the assaying step comprises contacting said one or more tissues, cell types, or a lysate thereof, with a provided probe compound and measuring the amount of protein kinase covalently modified by the probe compound.
EXEMPLIFICATION
[00314] As depicted in the Examples below, in certain exemplary embodiments, compounds are prepared according to the following general procedures. It will be appreciated that, although the general methods depict the synthesis of certain compounds of the present invention, the following general methods, and other methods known to one of ordinary skill in the art, can be applied to all compounds and subclasses and species of each of these compounds, as described herein.
[00315] Compound numbers utilized in the Examples below correspond to compound numbers set forth in Table 5, supra.
EXAMPLE 1
[00316] Preparation of N-(3-(5-methyl-2-(phenylamino)pyrimidin-4-ylamino)phenyl) acrylamide 1-7
Figure imgf000146_0001
1-7
[00317] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000147_0001
A) DIPEA, n-BuOH, 120 0C, 30 min, MW; B) NMP, 200 0C, 10 min, MW; C) NMP, 0 °C-30 min, rt-30 min. [00318] Step-1
Figure imgf000147_0002
[00319] A solution of 1 (2.0 g, 0.012 mol), 1,3-phenylenediamine (2.0 g, 0.018 mmol), DIPEA (2.33 g, 0.018 mol) in n-BuOH (20 mL) was subjected to microwave irradiation at 120 0C for 30 min. The reaction mixture was then quenched with water (100 mL), extracted with EtOAc (3x100 mL). The combined EtOAc extract was washed with water (100 mL), brine (100 mL), dried over Na2SO4 and concentrated under reduced pressure. The residue obtained was further purified column chromatography (SiO2, 60-120 mesh, EtOAc/CHCl3 : 15/85) gave 3 (1.3 g, 45 %) as a dark brown solid. [00320] Step-2
Figure imgf000147_0003
[00321] A solution of 3 (1.0 g, 4.27 mmol), 4 (1.5 g, 16.12 mmol) in NMP (10.0 mL) was subjected to microwave irradiation (200 0C, 10 min). The reaction mixture was cooled, diluted with water (100 mL) and extracted with EtOAc (3x100 mL). The combined ethyl acetate extract was washed with water (100 mL), brine (100 mL), dried over Na2SO4 and concentrated under reduced pressure gave a residue. The crude residue was further purified by column chromatography (SiO2, CHCl3/MeOH : 98/2) gave 5 ( 0.5 g, 40.3%) as a light brown solid. [00322] Step-3
Figure imgf000148_0001
1-7
[00323] To a stirred solution of 5 (200 mg, 0.68 mmol) in NMP (2.0 mL) at 0 0C was added acryloyl chloride (248 mg, 0.2.74 mmol) and the reaction mixture was stirred at 0 0C for 60 min.. The reaction mixture was then stirred with hexane for 1A h and then hexane was removed by decantation from the mixture and the residue was quenched with water (10 mL). The aqueous solution was basified with sat. NaHCO3 solution and then extracted with EtOAc (3x10 mL). The combined EtOAc extract was washed with water (10 mL), brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure. The residue obtained was further purified column chromatography (SiO2, 230-400, MeOH/ CHCl3 : 10/90) gave 1-7 (110 mg, 46.4%) as a brown solid. 1H NMR (DMSO-d6) δ ppm: 2.10 (s, 3H), 5.73 (dd, 1.88 & 10.42 Hz, IH), 6.24 (dd, J = 1.88 & 17 Hz, IH), 6.44 (dd, J = 10.08 & 16.92 Hz, IH), 6.78 (t, J= 7.36 Hz, IH), 7.06-7.11 (m, 2H), 7.26 (t, J= 8.08 Hz, IH), 7.38-7.40 (bm, 2H), 7.65 (d, J= 8.52 Hz, 2H), 7.88 (s, IH), 7.92 (s, IH), 8.37 (s, IH), 8.91 (s, IH), 10.09 (s, IH); LCMS: m/e 346.8 (M+l).
EXAMPLE 2 [00324] Preparation of N-(3-(4-(m-tolylamino)pyrimidin-2-ylamino)phenyl)acrylamide 1-1
Figure imgf000148_0002
1-1 [00325] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000149_0001
A) DIEA, n-BuOH, 110 0C, 30 min, microwave; B) NMP, 200 0C, 10 min, microwave; C) acryloyl chloride, NMP, 0 °C-30 min, rt-30 min. [00326] Step-1
Figure imgf000149_0002
[00327] A solution of 1 (0.5 g, 3.35 mmol), m-toluidine (0.36 g, 3.35 mmol), DIEA (0.65 g, 5.0 mmol) in n-BuOH (2.0 mL) was subjected to microwave irradiation at 110 0C for 30 min. The reaction mixture was then concentrated under reduced pressure, quenched with water (5 mL), extracted with EtOAc (3x20 mL). The combined EtOAc extract was washed with water (5 mL), brine (5 mL), dried over Na2SO4 and concentrated under reduced pressure. The residue obtained was further purified column chromatography (SiO2, 60-120 mesh, CHCVMeOH : 99/1) gave 3 (0.4 g, 54.2%) as a yellow solid. [00328] Step-2
Figure imgf000150_0001
[00329] A solution of 3 (0.2 g, 0.91 mmol), 4 (0.2 g, 1.8 mmol) in NMP (2.0 mL) was subjected to microwave irradiation (200 0C, 10 min). Then the reaction mixture was cooled, diluted with water (10 mL) and extracted with CH2Cl2 (3x15 mL). The combined CH2Cl2 extract was washed with water (5 mL), brine (5 mL), dried over Na2SO4 and concentrated under reduced pressure to get a residue. The crude residue was further purified by column chromatography (SiO2, CHCl3MeOH : 98/2) and gave 5 (0.14 g, 53%) as a light yellow solid. [00330] Step-3
Figure imgf000150_0002
[00331] To a stirred solution of 5 (0.075 g, 0.25 mmol) in NMP (1.0 mL) at 0 0C was added acryloyl chloride (0.19 g, 2.0 mmoL) and the reaction mixture was stirred at 0 0C for 30 min followed by stirring at rt for 30 min. The neat reaction mixture was subjected to purification by column chromatography (neutral Al2O3, CHCl3/MeOH : 98/2) gave 1-1 (0.04 g, 45%) as a white solid. 1H NMR (DMSO-d6) δ ppm: 2.56 (s, 3H), 5.71 (dd, J = 2.0 & 10.08 Hz, IH), 6.20-6.25 (m, 2H), 6.45 (dd, J= 10.12 & 17.00 Hz, IH), 6.78 (d, J= 7.52 Hz, IH), 7.12-7.19 (m, 2H), 7.31 (d, J = 8.44 Hz, IH), 7.46-7.53 (m, 3H), 7.87 (s, IH), 7.99 (d, J = 5.76 Hz, IH), 9.15 (s, IH), 9.24 (s, IH), 10.03 (s, IH); LCMS : m/e 346.4 (M+l). EXAMPLE 3
[00332] Preparation of N-(3 -(5 -methyl-4-(m-tolylamino)pyrimidin-2-ylamino)phenyl) acrylamide 1-2
Figure imgf000151_0001
1-2
[00333] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000151_0002
1-2
A) DIPEA, n-BuOH, 110 0C, 30 min, MW; B) NMP, 200 0C, 15 min, MW; C) acryloyl chloride, NMP, 0 °C-30 min, rt-30 min. [00334] Step-1
Figure imgf000151_0003
[00335] A solution of 1 (0.1 g, 0.613 mmol), 2 (0.066 g, 0.613 mmol), DIPEA (0.118 g, 0.919 mmol) in n-BuOH (2.0 mL) was subjected to microwave irradiation at 110 0C for 90 min. The reaction mixture was cooled, concentrated under reduced pressure and the residue obtained was further purified by column chromatography (SiO2, Methanol/chloroform mixtures) gave 3 (0.05 g, 34 %) as an off white solid. [00336] Step-2
Figure imgf000152_0001
[00337] A solution of 3 (0.05 g, 0.213 mmol), 4 (0.046 g, 0.427 mmol) in NMP (2.0 mL) was subjected to microwave irradiation (200 0C, 15 min). Then the reaction mixture was cooled, diluted with water (15 mL) and extracted with EtOAc (3x15 mL). The combined EtOAc extract was washed with water (10 mL), brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure gave a residue. The crude residue was further purified by column chromatography (SiO2, CHCl3/MeOH: 98/2) gave 5 (0.03 g, 46%) as a grey solid. [00338] Step-3
Figure imgf000152_0002
1-2
[00339] To a stirred solution of 5 (0.025 g, 0.082 mmol) in NMP (0.5 mL) at 0 0C was added acryloyl chloride (0.073 g, 0.821 mmol) and the reaction mixture was stirred at 0 0C for 30 min followed by stirring at rt for 30 min. The crude reaction mixture was passed through an alumina column (neutral Al2O3, chloroform/methanol mixtures) gave 1-2 (0.012 g, 41%) as a pale brown solid. 1H NMR (DMSO-d6) δ ppm: 2.10 (s, 3H), 2.27 (s, 3H), 5.72 (dd, J = 2 & 10.04 Hz, IH), 6.22 (dd, J = 1.96 & 16.92 Hz, IH), 6.45 (dd, J= 10.08 & 16.92 Hz, IH), 6.83 (d, J = 7.36 Hz, IH), 7.09 (t, J = 8.06 Hz, IH), 7.17 (t, J= 7.78 Hz, IH), 7.26 (d, J = 7.80 Hz, IH), 7.47 (d, J = 1.08 Hz, IH), 7.53 (s, IH), 7.58 (d, J = 8.60 Hz, IH), 7.78 (s, IH), 7.88 (s, IH), 8.15 (s, IH), 9.01 (s, IH), 9.99 (s, IH); LCMS: m/e 360.1 (M+l).
EXAMPLE 4
[00340] Preparation of N-(3-(5-fluoro-4-(m-tolylamino)pyrimidin-2-ylamino)phenyl) acrylamide 1-3
Figure imgf000153_0001
[00341] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000153_0002
A) n-butanol, DIPEA, 110 o υ,C, 45 min, MW; B) NMP, 200 0C, 10 min, MW; C) NMP, DMAP, 0 0C, 30 min. [00342]
Figure imgf000154_0001
[00343] To a solution of 1 (0.5 g, 3 mmol) in n-butanol (5.0 niL) was added 2 (0.64 g, 0.6 mmol), DIPEA (0.116 g, 0.8 mmol) and the reaction mixture was irradiated under microwave at 110 0C for 45 min. It was cooled, quenched with water (50 mL) and extracted with EtOAc (2x25 mL). The combined EtOAc extract was washed with water (25 mL), brine (25mL), dried over Na2SO4 and concentrated under reduced pressure gave 3 (0.45 g, 63%) which was taken for the next step without further purification. [00344] Step-2
Figure imgf000154_0002
[00345] A solution of 3 (0.45 g, 1.8 mmol) and 4 (0.41 g, 3.7 mmol) in NMP (4.5 mL) was subjected to microwave irradiation at 200 0C for 10 min. It was cooled, diluted with water (25 mL) and extracted with EtOAc (3x25 mL). The combined EtOAc extract was washed with water (2x25 mL), brine (25 mL), dried over Na2SO4 and concentrated under reduced pressure. The residue obtained was further purified by column chromatography (SiO2, 60-120, Chloroform/Ethyl acetate: 90/10) gave 5 (0.23 g, 41%) as a light yellow solid. [00346] Step-3
Figure imgf000154_0003
1-3 [00347] To a stirred solution of 5 ( 0.075 g, 0.24 mmol), in NMP(1.5 niL) at 0 0C under N2 atmosphere was added DMAP (0.059 g, 0.48 mmol) and Acryloyl chloride (0.064 g, 0.725 mmol) and the reaction mixture was kept at this temperature for 30 min. It was quenched with water (7.5 mL) and extracted with EtOAc (3x25 mL). The combined EtOAc extract was washed with 5% Citric acid (10 mL), water (2x10 mL), brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure. The crude residue was further purified by column chromatography (AI2O3, Chloroform/Methanol: 98/2) gave 1-3 (0.01 g, 11.3%) as an off white solid. 1H NMR (DMSO-d6) δ ppm: 2.27 (s, 3H), 5.72 (d, J= 9.84 Hz, IH), 6.22 (d, J= 16.92 Hz, IH), 6.44 (dd, J= 10.2 & 17.02 Hz, IH), 6.85 (d, J= 7.12 Hz, IH), 7.12-7.19 (m, 2H), 7.29 (d, J = 7.68 Hz, IH), 7.43 (d, J= 7.92 Hz, IH), 7.61-7.63 (m, 2H), 7.82 (s, IH), 8.08 (s, IH), 9.23 (bs, 2H), 10.03 (s, IH); LCMS: m/e 364.2 (M+l).
EXAMPLE 5
[00348] Preparation of (E)-4-(dimethylamino)-N-(3-(5-fluoro-4-(m-tolylamino)pyrimidin-2- ylamino)phenyl)but-2-enamide 1-4
Figure imgf000155_0001
1-4
[00349] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000156_0001
A) n-butanol, DIPEA, 110 ° C, 45 min., M W;B ) NMP, 200°C, 10min., MW; C) oxal yl chloride, CH3CN, ½ h at 0 0C, 2 h at 25 0C, 5 min at 45 °C; D) NMP, 0 °C to 10 °C, 30 min. [00350] Step-1
Figure imgf000156_0002
[00351] A solution of 1 (0.5 g, 3.0 mmol), 2 (0.32 g, 3.0 mmol) in n-butanol (5.0 mL) was subjected to microwave irradiation (110 0C, 45 min). It was cooled, quenched with water (50 mL) and extracted with EtOAc (2x25 mL). The combined EtOAc extract was washed with water (25 mL), brine (25 mL), dried over Na2SO4 and concentrated under reduced pressure gave 3 (0.45 g, 63%) which was taken for next step without further purification. [00352] Step-2
Figure imgf000156_0003
[00353] A solution of 3 (0.45 g, 1.8 mmol), 4 (0.41 g, 3.7 mmol) in NMP (4.5 niL) was subjected to microwave irradiation (200 0C, 10 min). It was cooled, diluted with water (25 mL) and extracted with EtOAc (3x25 mL). The combined ethyl acetate extract was washed with water (2x25 mL), brine (25 mL), dried over Na2SO4 and concentrated under reduced pressure. The residue was further purified by column chromatography (SiO2, Chloroform/Ethyl acetate: 90/10) gave 5 (0.23 g, 41%) as a light yellow solid.
[00354] Step-3a
Figure imgf000157_0001
[00355] To a stirred solution of 6 (0.13 g, 0.80 mmol) in CH3CN (1.0 mL) was added oxalyl chloride (0.122 g, 0.96 mmol) at 0 0C. The reaction mixture was allowed to stir at 0 0C for 1A h and then at RT for 2 h. Finally it was heated at 45 0C for 5 min, cooled and the reaction mixture was taken for next step without further purification. [00356] Step-3
Figure imgf000157_0002
[00357] To a stirred solution of 5 (0.05 g, 0.16 mmol) in NMP (1.0 mL) was added 7 at 0 0C. The reaction mixture was stirred at 0 0C for 30 min and at 10 0C for 30 min. It was quenched with sat. Sodium bicarbonate soln. (5 mL) and extracted with CH2Cl2 (3x5 mL). The combined organic extract was washed with water (1 mL), brine (1 mL) and dried over Na2SO4. Concentration under reduced pressure followed by purification by column chromatography (SiO2, 230-400, CHCls/MeOH, 95/5) gave 1-4 (0.02 g, 29.4%) as a white solid. 1H NMR (DMSO-de) δ ppm: 2.21 (s, 6H), 2.28 (s, 3H), 3.08 (bd, J= 5.6 Hz, 2H), 6.29 (d, J= 15.60 Hz, IH), 6.67-6.74 (m, IH), 6.86 (d, J= 7.20 Hz, IH), 7.12-7.20 (m, 2H), 7.27 (d, J= 8.00 Hz, IH), 7.43 (d, J= 8.00 Hz, IH), 7.62-7.64 (m, 2H), 7.82 (s, IH), 8.08 (d, J= 3.6 Hz, IH), 9.23 (s, IH), 9.24 (s, IH), 9.96 (s, IH); LCMS: m/e 421.2 (M+l). EXAMPLE 6
[00358] Preparation of N-(3 -(5 -methyl-4-(phenylamino)pyrimidin-2-ylamino)phenyl) acrylamide 1-5
Figure imgf000158_0001
1-5
[00359] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000158_0002
I-5
A) DIPEA, n-BuOH, 110 0C, 30 min, MW; B) NMP, 200 0C, 15 min, MW; C) acryloyl chloride, NMP, 0 °C-30 min, rt-30 min. [00360] Step-1
Figure imgf000158_0003
[00361] A solution of 1 (0.1 g, 0.613 mmol), 2 (0.114 g, 1.226 mmol), DIPEA (0.118 g, 0.919 mmol) in n-BuOH (2.0 mL) was subjected to microwave irradiation at 110 0C for 90 min. The reaction mixture was cooled, concentrated under reduced pressure and the residue was further purified by column chromatography (SiO2, 60-120, Methanol/chloroform: 1/9) gave 3 (0.08 g, 59 %) as a white solid [00362] Step-2
Figure imgf000159_0001
[00363] A solution of 3 (0.08 g, 0.364 mmol), 4 (0.059 g, 0.546 mmol) in NMP (2.0 ml) was subjected to microwave irradiation (200 0C, 15 min). The reaction mixture was cooled, diluted with water (15 mL) and extracted with EtOAc (3x15 mL). The combined ethyl acetate extract was washed with water (10 mL), brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure gave a residue. The crude residue was further purified by column chromatography (SiO2, 60-120, CHCl3/MeOH: 98/2) gave 5(0.06 g, 60%) as a light grey solid. 1H NMR (DMSO-de) δ ppm: 2.09 (s, 3H), 4.74 (s, 2H), 6.09-6.11 (m, IH), 6.77-6.85 (m, 2H), 6.91 (t, J = 1.72 Hz, IH), 7.02 (t, J = 7.36 Hz, IH), 7.31 (t, J = 7.52 Hz, 2H), 7.75 (d, J = 7.68 Hz, 2H), 7.84 (s, IH), 8.18 (s, IH), 8.65 (s, IH); LCMS: m/e 293.2 (M+l). [00364] Step-3
Figure imgf000159_0002
1-5
[00365] To a stirred solution of 5 (60 mg, 0.205 mmol) in NMP (2.0 mL) at 0 0C was added acryloyl chloride (0.148 g, 1.64 mmol) and the reaction mixture was stirred at 0 0C for 30 min.. The neat reaction mixture was passed through an alumina column (neutral Al2O3, chloroform/methanol, 99/1) gave 1-5 (0.013 g, 18.5%) as an off-white solid. 1H NMR (DMSO- de) δ ppm: 2.11 (s, 3H), 5.72 (dd, J= 1.92 & 10.04 Hz, IH), 6.22 (dd, J= 1.92 & 16.92 Hz, IH), 6.45 (dd, J = 9.32 & 16.92 Hz, IH), 7.00 (t, J = 7.28 Hz, IH), 7.09 (t, J = 8.04 Hz, IH), 7.23- 7.30 (m, 3H), 7.43 (d, J = 8.04 Hz, IH), 7.75-7.77 (m, 2H), 7.83 (s, IH), 7.88 (s, IH), 8.22 (s, IH), 9.00 (s, IH), 9.99 (s, IH); LCMS: m/e 346 (M+l).
EXAMPLE 7
[00366] Preparation of N-(4-methyl-3-(5-methyl-4-(m-tolylamino)pyrimidin-2-ylamino) phenyl)acrylamide 1-8
Figure imgf000160_0001
1-8
[00367] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000160_0002
8
A) DIPEA, n-BuOH, 120 0C, 60 min., MW; A') (BOC)2O, MeOH, -10 0C, 4 h; B) Pd(OAc)2, BINAP, Cs2CO3, toluene, 110 0C, 12 h; C) TFA, CH2Cl2, 0 °C-30 min, rt-2 h; D) acryloyl chloride, NMP, 0 °C-30 min, rt-30 min. [00368]
Figure imgf000161_0001
[00369] To a stirred solution of A (5 g, 0.04 mmol) in MeOH (75 niL) was added (BOC)2O (11.59 g, 0.050 mmol), slowly at -10 0C. The reaction was stirred at this temperature for 4 h and then reaction mixture was concentrated under reduced pressure. The residue obtained was taken in EtOAc (300 mL). It was washed with water (25 mL), brine (25 mL) and dried over Na2SO4. Filtration followed by concentration under reduced pressure offered 4 (2.5 g, 27%) as an off- white solid. [00370] Step l
Figure imgf000161_0002
[00371] A solution of 1 (0.5 g, 3.06 mmol), 2 (0.39 g, 3.06 mmol), DIPEA (0.59 g, 4.5 mmol) in n-BuOH (5 mL) was subjected to microwave irradiation (120 0C, 30 min). The reaction mixture was cooled, solvents removed under reduced pressure and the residue obtained was quenched with water (5 mL). It was extracted with EtOAc (3x20 mL) and the combined EtOAc layer was washed with water (5 mL), brine (5 mL) and dried over Na2SO4. Filtration followed by concentration under reduced pressure offered a residue which was further purified by column chromatography (SiO2, 60-120, CHCl3/MeOH : 9/1) gave 3 (0.35 g, 49%) as an off-white solid. [00372] Step 2
Figure imgf000161_0003
[00373] A solution of 3 (0.1 g, 0.43 mmol), 4 (0.14 g, 0.64 mmol), Pd(OAc)2 (10 mg, 0.043 mmol), BINAP (0.013 g, 0.021 mmol) and Cs2CO3 (0.2 g, 1.06 mmol) in degassed toluene (toluene was purged with N2 for 15 min) was refluxed for 12 h under N2 atmosphere. The reaction mixture was cooled and passed through a short bed of celite®. The filtrate was diluted with EtOAc (25 mL) and washed with water (5 mL), brine (5 mL) and dried over Na2SO4. Filtration followed by concentration under reduced pressure offered a residue which was further purified by column chromatography (SiO2, 60-120, CHCl3/MeOH : 9/1) gave 5 (40 mg, 22%) as an off-white solid. [00374] Step-3
Figure imgf000162_0001
[00375] To a stirred solution of 5 (0.04 g, 0.095 mmol) in dry CH2Cl2 (2 mL) at 0 C was added CF3COOH (0.2 mL, 5 vol) and the reaction mixture was kept at this temperature for 30 min. It was allowed to come to rt and stir at this temperature for 2 h. It was quenched with ice- cooled water (2 mL), basified with sodium carbonate solution and extracted with EtOAc (2x10 mL). The combined EtOAc extract was washed with water (2 mL), brine (2 mL) and dried over Na2SO4. Filtration followed by concentration under reduced pressure offered 6 (22 mg, 73%) as a light brown solid. [00376] Step-4
Figure imgf000162_0002
1-8
[00377] To a stirred solution of 6 (0.2 g, 0.63 mmol) in NMP (4 mL) at 0 0C was added acryloyl chloride (0.12 g, 1.25 mmol). The reaction was kept at this temperature for 30 min and then at rt for 30 min. It was quenched with ice-cooled water (2 mL) and extracted with EtOAc (2x10 mL). The combined EtOAc extract was washed with water (2 mL), brine (2 mL) and dried over Na2SO4. Filtration followed by concentration under reduced pressure offered a residue which was further purified by column chromatography (SiO2, 230-400, CHCl3/MeOH : 9/1) gave 1-8 (10 mg, 4%) as a white solid. 1H NMR (DMSO-d6) δ ppm: 2.07 (s, 3H), 2.13 (s, 6H), 5.70 (dd, J = 1.92 & 10.08 Hz, IH), 6.20 (dd, J = 1.96 & 16.88 Hz, IH), 6.41 (dd, J = 10.16 & 16.96 Hz, IH), 6.69 (d, J = 7.36 Hz, IH), 6.98 (t, J = 7.76 Hz, IH), 7.11 (d, J = 8.24 Hz, IH), 7.41 (q, J = 9.92 Hz, IH), 7.49-7.51 (m, 2H), 7.73 (s, IH), 7.80 (s, IH), 7.97 (s, IH), 8.16 (s, IH), 10.00 (s, IH); LCMS : m/e 374 (M+ 1).
EXAMPLE 8
[00378] Preparation of N-(3 -(4-(3 -bromophenylamino)-5 -methylpyrimidin-2-ylamino)pheny 1) acrylamide 1-9
Figure imgf000163_0001
1-9
[00379] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000164_0001
I-9
A) DIPEA, n-butanol, 110 0C, 1 h, MW; B) 1.5 N HCl, ethanol, 90 0C, 30 min., MW; C) acryloyl chloride, NMP, 0 0C, 30 min. [00380] Step l
Figure imgf000164_0002
[00381] A solution of 1 (0.5 g, 3.06 mmol), 2 (0.53 g, 3.06 mmol) and DIPEA (0.80 mL, 4.06 mmol) in n-butanol (5 mL) was subjected to microwave irradiation (110 0C, 1 h). The reaction mixture was cooled and concentrated under reduced pressure gave a residue. The residue taken in EtOAc (5 mL) and washed with NaHCO3 solution (2 mL), water (2 mL) and with brine solution (2 mL). Drying over Na2SO4 followed by concentration under reduced pressure offered crude 3 which was further purified by column chromatography (SiO2, 60-120, chloroform/methanol, 9/1) gave 3 (0.125 g, 13%) as a brown solid. [00382]
Figure imgf000165_0001
[00383] To a solution of 3 (0.15 g, 0.5 mmol) in EtOH (3 mL)was added 4 (0.081 g, 0.75 mmol) followed by 1.5 N HCl (0.055 g, 1.5 mmol). The reaction mixture was subjected to microwave irradiation (90 0C, 30 min), cooled and concentrated under reduced pressure. The residue obtained was taken in EtOAc (5 mL) and washed with NaHCO3 solution (2 mL), water (2 mL), and brine (2 mL). It was dried over Na2SO4, filtered and concentrated under reduced pressure gave crude 5. It was further purified by column chromatography (SiO2, 60-120, chloroform/methanol, 9/1) gave 5 (0.06 g, 32%) as a light brown solid. [00384] Step 3
Figure imgf000165_0002
1-9
[00385] To a stirred solution of 5 (0.06 g, 0.16 mmol) in NMP (1 mL) was added acryloyl chloride (0.117 g, 1.29 mmol) at 0 0C. The reaction mixture was allowed to stir at this temperature for 30 min and then taken in dichloromethane (2 mL). It was washed with NaHCO3 solution (1 mL), water (1 mL) and with brine solution (1 mL). It was dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was further purified by column chromatography (SiO2, 60-120, chloroform/methanol, 9/1) gave 1-9 (0.016 g, 23%) as a pale brown solid. 1H NMR (DMSO-d6) δ ppm: 2.16 (s, 3H), 5.75 (dd, J = 1.72 & 10 Hz, IH), 6.23 (dd, J = 1.76 & 16.88, Hz, IH), 6.45 (dd, J = 10.08 & 16.92 Hz, IH), 7.22-7.34 (m, 4H), 7.38 (d, J = 8.00 Hz, IH), 7.63 (d, J = 8.08 Hz, IH), 7.77 (s, IH), 7.82 (s, IH), 7.93 (s, IH), 9.68 (s, IH), 10.26 (s, IH), 10.34 (s, IH); LCMS: m/e 426 (M+l). EXAMPLE 9
[00386] Preparation of 3-(4-(2-(cyclopropylsulfonyl)-l,2,3,4-tetrahydroisoquinolin-6- ylamino)-5 -methylpyrimidin-2-ylamino)benzenesulfonamide I- 10
Figure imgf000166_0001
1-10
[00387] The title compound was prepared according to the schemes, steps and intermediates described below.
Z J
Figure imgf000166_0002
A') DPPA, Benzyl alcohol, Et3N, toluene, 110 0C, 12 h.; B') Pd(OH)2, Ammonium formate, EtOH, reflux, 6 h; A) DIPEA, n-BuOH, 120 0C, 1 h., MW; B) 1.5 N HCl, EtOH, reflux 12 h.; C) Cyclopropylsulphonyl chloride, DIPEA, THF, rt, 12 h. [00388] Steps 1-4 The procedure for synthesizing scaffold 7 is described in the experimental for Compound I- 11 herein. [00389] Step S
Figure imgf000167_0001
1-10
[00390] To a stirred solution of 7 (0.05 g, 0.0121 mmol) in THF (4 mL) at 0 0C, was added DIPEA (0.023 g, 0.182 mmol) followed by cyclopropylsulphonyl chloride (0.031 g, 0.182 mmol) under N2 atmosphere. The reaction mixture was allowed to come to rt and maintained at this temperature for 12 h. It was taken in EtOAc (10 mL), washed with water (5 mL), brine (5 mL) and dried over Na2SO4. Filtration followed by concentration under reduced pressure offered a residue which was further purified by column chromatography (SiO2, 60-120, pet ether/ethyl acetate, 6/4) gave MO (0.035 g, 56%) as a yellow solid. 1H NMR (DMSO-d6) δ ppm: 0.97- 0.1.00 (m, 4H), 2.12 (s, 3H), 2.60-2.66 (m, IH), 2.90 (t, J= 5.2 Hz, 2H), 3.52 (t, J= 6 Hz, 2H), 4.42 (s, 2H), 7.16 (d, J = 8.4 Hz, IH), 7.27 (s, 2H), 7.31-7.35 (m, 2H), 7.53 (s, IH), 7.59 (d, J = 8.4 Hz, IH), 7.92 (s, IH), 8.03-8.04 (m, 2H), 8.45 (s, IH), 9.40 ( s, IH); LCMS: m/e 515 (M+l).
EXAMPLE 10
[00391] Preparation of 3-(4-(2-(2-chloroacetyl)-l,2,3,4-tetrahydroisoquinolin-6-ylamino)-5- methylpyrimidin-2-ylamino)benzenesulfonamide 1-11
Figure imgf000167_0002
1-11 [00392] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000168_0001
1-11
A') DPPA, Benzyl alcohol, Et3N, toluene, 110 C, 12 h.; B') Pd(OH)2, Ammonium formate, EtOH, reflux, 6 h; A) DIPEA, n-Bu0H, 120 0C, 1 h., MW; B) 1.5 N HCl, EtOH, reflux 12 h.; C) Cl-CH2-COCl, Et3N, THF, rt, 12 h. [00393] Step-1
Figure imgf000168_0002
[00394] To a stirred solution of 1 (1.5 g, 5.4 mmol) in toluene (15 mL) was added DPPA (2.17 g, 8.11 mmol), Et3N (1.05 mL, 8.11 mmol) and benzyl alcohol (0.876 g, 8.11 mmol) under N2. The reaction mixture was allowed to reflux for 12 h, cooled and diluted with ethyl acetate (100 mL). It was washed with water (5 mL), brine solution (5 mL) and dried over Na2SO4. It was filtered and concentrated under reduced pressure and the residue was purified by column chromatography (SiO2, 60-120, chloroform/methanol, 9/1) gave 2 (2.0 g, 97%) as a white solid. [00395]
Figure imgf000169_0001
[00396] To a stirred solution of 2 (2.2 g, 5.75 mmol) in EtOH (25 mL) was added ammonium formate (3.68 g, 57.5 mmol) and the reaction mixture was refluxed for 6 h. It was cooled, filtered though a celite® bed and filtrate was concentrated under reduced pressure gave 3 (1.3 g, 91%) as a dark brown oil which was used without further purification.
[00397] Step-3
Figure imgf000169_0002
5 [00398] A solution of 3 (1.4 g, 5.56 mmol), 4 (0.912 g, 5.56 mmol) and DIPEA (1.077 g, 8.3 mmol) in n-BuOH (15 mL) was subjected to microwave irradiation at 120 0C for 45 min. The reaction mixture was cooled and concentrated under reduced pressure. The residue was taken in ethyl acetate (20 mL) and washed with water (5 mL) and brine (5 mL). Drying over Na2SO4 followed by concentration under reduced pressure offered a residue which was purified by column chromatography (SiO2, 60-120, chloroform/methanol, 9/1) gave 5 (1.1 g, 52%) as a cream colored solid. [00399] Step-4
Figure imgf000169_0003
[00400] To a stirred solution of 5 (0.25 g, 0.66 mmol) in ethanol (5 mL) was added 6 (0.126 g,
0.73 mmol) and catalytic amount of aq.HCl and the reaction mixture was refluxed for 12 h at 100 0C. It was cooled, the solid precipitated was filtered and washed with diethyl ether and dried under high vacuum gave 7 (0.24 g, 82%) as a light yellow solid. [00401]
Figure imgf000170_0001
[00402] To a stirred solution of 7 (0.2 g, 0.487 mmol) in NMP (5 mL) was added Et3N (0.094 g, 0.731 mmol). The solution was cooled to 0 0C and chloroacetylchloride (0.082 g, 0.731 mmol) was added to it. The reaction mixture was allowed to come to rt and stir at this temperature for 12 h. It was quenched with ice cooled water (2 mL) and extracted with ethyl acetate (3x5 mL). The combined ethyl acetate extract was washed with brine solution (2 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue obtained was further purified by column chromatography (SiO2, 60-120, chloroform/methanol, 9/1) gave 1-11 (0.038 g, 16%) as a light yellow solid. 1H NMR (DMSO-d6) δ ppm: 2.11 (s, 3H), 2.77-2.89 (m, 2H), 3.70-3.72 (m, 2H), 4.49 (d, J= 2.92 Hz, 2H), 4.63 (d, J= 23.56 Hz, 2H), 7.15-7.17 (m, IH), 7.24 (s, 2H), 7.30-7.32 (m, 2H), 7.50-7.65 (m, 2H), 7.91 (s, IH), 8.04-8.05 (m, 2H), 8.27 (s, IH), 9.31 (s, IH), .LCMS: m/e 486.8 (MH+).
EXAMPLE 11
[00403] Preparation of N-(3 -(5 -methyl-4-(4-phenoxyphenylamino)pyrimidin-2-ylamino) phenyl)acrylamide 1-23
Figure imgf000170_0002
[00404] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000171_0001
C Λ) Acryloyl chloride 0 0C, rt, 1 h.
[00405] Step-1
Figure imgf000171_0002
[00406] A solution of 1 (0.2 g, 1.2 mmol), 2 (0.12 g, 0.95 mmol) and DIPEA (0.23 g, 1.78 mmol) in n-BuOH (2 rnL) was subjected to microwave irradiation (100 0C for 1 h). Then the reaction mixture was cooled, concentrated under reduced pressure and the residue was taken in EtOAc (5 mL). It was washed with NaHCO3 solution (2 mL), water (2 mL), brine (2 mL) and then dried over anhydrous Na2SO4. Concentrated under reduced pressure followed with purification by column chromatography (SiO2, 60-120, chloroform/methanol, 9/1) gave 3 (0.11 g, 28.9%) as a light brown solid.
[00407] Step-2
Figure imgf000171_0003
[00408] To a solution 3 (0.11 g, 0.3 mmol), 4 (0.114 g, 1.05 mmol) in n-butanol (1 mL) was added cone. HCl (1 drop) and the mixture was subjected to microwave irradiation (165 0C for 10 min). The reaction mixture was cooled, concentrated under reduced pressure and the residue was taken in EtOAc (5 mL). It was washed with NaHCO3 solution (2 mL), water (2 mL) and brine (2 niL). Drying over Na2SO4 followed by concentration under reduced pressure offered residue which was purified by column chromatography (SiO2, 60-120, chloroform/methanol, 9/1) gave 5 (0.08 g, 65%) as a brown solid.
[00409]
Figure imgf000172_0001
[00410] To a stirred solution 5 (0.015 g, 0.03 mmol) in NMP (1 mL) was added acryloyl chloride (0.005 g, 0.05 mmol) at 0 0C. The reaction mixture was allowed to come to rt and kept at this temperature for 1 h. It was diluted with dichloromethane (2 mL) and washed with NaHCO3 solution (1 mL), water (1 mL) and brine (1 mL). Drying over Na2SO4 followed by concentration under reduced pressure offered a residue which was further purified by column chromatography (SiO2, 60-120, chloroform/methanol, 9/1) gave 1-23 (0.004 g, 23%) as a brown solid. 400 MHz, MeOD: δ 2.14 (s, 3H), 5.71 (d, J= 11.20 Hz, IH), 6.30-6.44 (m, 2H), 6.94-6.99 (m, 4H), 7.07-7.15 (m, 2H), 7.22 (d, J = 7.2 Hz, IH), 7.34-7.36 (m, 3H), 7.63 (d, J = 8.8 Hz, 2H), 7.79 (s, 2H); LCMS: m/e 437 (M+l).
EXAMPLE 12
[00411] Preparation of N-(3-(5-methyl-2-(3-sulfamoylphenylamino)pyrimidin-4- ylamino)phenyl)acrylamide 1-33
Figure imgf000172_0002
1-33
[00412] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000173_0001
A) DIPEA, n-BuOH, 120 0C, 30 min., MW; B) 1.5 N HCl, Ethanol, 100 0C, 12 h; C) NMP, 0 0C to rt, 1 h. [00413] Step-1
Figure imgf000173_0002
1 [00414] A solution of 1 (0.5 g, 3.06 mmol), 1 (0.49 g, 4.59 mmol) and DIPEA (0.59 g, 4.59 mmol) in n-butanol (8 niL) was subjected to microwave irradiation (120 0C, 30 min). It was cooled, quenched with water (5 mL) and extracted with ethyl acetate (3x20 mL). The combined ethyl acetate layer was washed with brine solution (5 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was further purified by column chromatography (SiO2, 60-120, chloroform/ethyl acetate, 9/1) gave 1 (0.25 g, 34.77%) as a light brown solid. [00415] Step-2
Figure imgf000173_0003
[00416] To a stirred solution of 3 (0.1 g, 0.48 mmol), in ethanol (2 mL) was added 4 (0.070 g, 0.42 mmol) and catalytic amount of 1.5 N HCl (3 drops), then heated to 100 0C, for 12 h. Reaction mixture then cooled, solid separated, which was filtered and washed with ether 5 (0.1 g as crude), which was taken to next step as such. [00417] Step-3
Figure imgf000174_0001
1-33
[00418] To a stirred solution of 5 (0.1 g, 0.27 mmol) in NMP (2 rnL) was added acryloyl chloride (0.037 g, 0.425 mmol) at 0 0C, this was then stirred at room temperature for 1 h, then the reaction mixture was quenched with water (4 mL) and basified with NaHCO3, this was then extracted with ethyl acetate (5 mL), combined organic layer washed with brine solution (1 mL), dried over anhydrous Na2SO4, filtered then concentrated, Crude then purified using preparative HPLC yields 1-33 (0.07 g, 6%) as an off white solid. 1H NMR (MeOD) δ ppm: 2.17 (s, 3H), 5.78 (dd, J= 2.36 & 9.52 Hz, IH), 6.34-6.48 (m, 2H), 7.26-7.43 (m, 5H), 7.87 (s, IH), 7.96-8.03 (m, 3H); LCMS: m/e 425 (M+ 1).
EXAMPLE 13
[00419] Preparation of N-(3 -(methyl(5 -methyl-2-(phenylamino)pyrimidin-4-yl)amino) phenyl)acrylamide 1-34
Figure imgf000174_0002
[00420] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000175_0001
I-34
A) Pd(OAC)2, BINAP, Cs2CO3, Toluene, 100 0C, 16 h; B) NaH, CH3I, THF, 0 °C-30 min, rt-16 h.; C) Aniline, conc.HCl, Ethanol, 90 0C, 60 min; D) H2, Pd/C, Ethanol, 16 h; E) acryloyl chloride, NMP, 0 0C, 1 h. [00421] Step-1
Figure imgf000175_0002
[00422] To a stirred solution of 2 (1.0 g, 6.0 mmol), in Toluene (30.0 mL) was added 1 (0.84 g, 6.0 mmol), BINAP (0.186 g, 0.3 mmol), Cs2CO3 (4.87 g, 15.0 mmol). The reaction mixture was degassed by purging N2 for 15 min. Pd(OAc)2 (0.134 g, 0.6 mmol) was then added to the reaction mixture and the reaction mixture was heated at 100 0C for 16 h under N2 atmosphere. It was then cooled, diluted with Ethyl acetate (30 mL) and filtered through celite®. Filtrate was washed with water (2x25 mL), brine (25 mL), dried over Na2SO4 and concentrated under reduced pressure. The residue obtained was further purified column chromatography (SiO2, 60- 120 mesh, Ethylacetete/hexane: 10/90) gave a solid which was washed with ether gave 3 (0.6 g, 37%) as a light yellow solid. [00423]
Figure imgf000176_0001
[00424] To a stirred mixture of NaH (0.1 g, 2.5 mmol, 60% dispersion in paraffin oil) in dry THF (10.0 rnL ) was added 3 (0.5 g, 1.89 mmol) at 0 0C, and the reaction mixture was stirred at this temperature for 30 min. CH3I (0.305 g, 2.15 mmol) was added to it and the reaction was allowed to come to rt and stir at this temperature for 16 h. The reaction mixture was diluted with water (25 mL) and extracted with EtOAc (3x25 mL). The combined EtOAc extract was washed with water (25 mL), brine (25 mL), dried over Na2SO4 and concentrated under reduced pressure gave a residue. The crude residue was further purified by column chromatography (SiO2, CHCl3/MeOH: 99/1) gave 4 (0.12 g, 22.7%) as a light yellow solid. [00425] Step-3
Figure imgf000176_0002
[00426] To a solution of 4 (120 mg, 0.431 mmol) in EtOH (2 mL) was added Conc.HCl (0.044 g, 1.2 mmol) and Aniline (0.16 g, 1.72 mmol) and the reaction mixture was heated in a sealed pressure tube at 90 0C for 1 h. The reaction mixture was cooled, solvents removed by concentration under reduced pressure and the residue obtained was diluted with 10% NaHCO3 (10.0 mL). It was extracted with EtOAc (3x15 mL) and the combined EtOAc extract was washed with water (15 mL), brine (15 mL), dried over Na2SO4. Concentration under reduced pressure offered a residue which was further purified by column chromatography (SiO2, CHCl3/MeOH: 99/1) gave 5 (0.11 g, 76%) as a light yellow solid. [00427]
Figure imgf000177_0001
[00428] A solution of 5 (0.110 g, 0.328 mmol), in Ethanol (50 mL)) was added 10% Palladium on charcoal (0.022 g) and the reaction mixture was stirred under H2 atmosphere (1.5 Kg) at rt for 16 h. It was filtered through celite® and concentrated under reduced pressure gave a residue. The residue was purified by column chromatography (SiO2, 60-120, methanol/chloroform: 1/99) gave 6 (0.07 g, 69.9%) as a colorless viscous liquid. [00429] Step-5
Figure imgf000177_0002
1-34
[00430] To a stirred solution of 6 (0.070 g, 0.23 mmol) in NMP (1.5 mL) at 0 0C was added acryloyl chloride (0.083 g, 0.916 mmol) and the reaction mixture was stirred at 0 0C for 1 h. It was quenched with 10% sodium bicarbonate solution (15 mL) and the solid precipitated out was filtered, washed with cold water (5 mL), hexane (5 mL). The solid was dried for 2 h under reduced pressure gave 1-34 (0.033 g, 40%) as a pale yellow sold. 1H NMR (DMSO-dβ) δ ppm: δ 1.47 (s, 3H), 3.45 (s, 3H), 5.74 (dd, J= Hz, IH), 6.22 (dd, J= 2.0 & 16.98 Hz, IH), 6.38 (dd, J = 10 & 16.94 Hz, IH), 6.85-6.91 (m, 2H), 7.21-7.25 (m, 2H), 7.32 (t, J = 8.02 Hz, IH), 7.43-7.47 (m, 2H), 7.77-7.79 (m, 2H), 7.90 (s, IH), 9.22 (s, IH), 10.18 (s, IH); LCMS: m/e 360.8 (M+l). EXAMPLE 14
[00431] Preparation of N-(3-(5-methyl-2-(3-(prop-2-ynyloxy)phenylamino)pyrimidin-4- ylamino)phenyl) acrylamide 1-35
Figure imgf000178_0001
1-35
[00432] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000178_0002
A) K2CO3, CH3CN, 65 UC, 8 h; B) Fe powder, NH4Cl, MeOH,
Figure imgf000178_0003
1,3- pheneylendiamine, DIPEA, n-BuOH, 120 0C, 30 min, MW; D) Con.HCl, absolute ethanol, 110 0C, 2 h; E) NMP, 0 0C, 1 h. [00433] Step-1
Figure imgf000178_0004
[00434] To a stirred solution of Ia (4 g, 0.0287 mol) and K2CO3 (5.6 g, 0.0574 mol) in CH3CN (15 rnL) was added propargyl bromide (4.1 g, 0.0345 mol) and the resulting mixture was allowed to reflux for 8 h. The reaction mixture was then cooled, quenched with water and extracted with EtOAc (3x50 mL). The combined EtOAc extract was washed with water (20 mL), brine (20 mL) and dried over Na2SO4. Filtration followed by concentration under reduced pressure furnished 2b as a brownish solid which was used without further purification. [00435] Step-2
Figure imgf000179_0001
[00436] To a stirred solution of 2b in a mixture of methanol (30 mL) and water (30 mL) was added, NH4Cl (10.3 g, 0.194 mol) and iron powder (6.8 g, 0.121 mol) respectively. Resulting mixture was refluxed at 80 0C for 4 h. Reaction mixture was cooled, diluted with methanol and filtered through a pad of celite®. The filtrate was concentrated under reduced pressure and the residue was taken in EtOAc. It was washed with water, brine, dried over Na2SO4 and concentrated under reduced pressure gave a residue. The residue was further purified by column chromatography (SiO2, 60-120, gravity column chromatography, the expected product was eluted with CHCl3/MeOH : 96/4) gave 3 (3.2 g, 91%) as a brownish solid. [00437] Step-3
Figure imgf000179_0002
2
[00438] A solution of 2, 4-dichloro-5 -methyl pyrimidine 1 (0.3 g, 0.0018 mol), 1,3-phenylene diamine (0.24 g, 0022 mol), DIPEA (0.35 g, 0.0027 mol) in n-BuOH (3 mL) was subjected to microwave irradiation (120 0C, 30 min). The reaction mixture was cooled, quenched with water (15 mL) and extracted with EtOAc (3x15 mL). The combined EtOAc extract was washed with water (20 mL), brine (20 mL), dried over Na2SO4 and concentrated under reduced pressure. The residue obtained was further purified column chromatography (SiO2, 60-120) gave 2 (0.15 g, 35%) as a brownish solid. [00439]
Figure imgf000180_0001
2 (0.15 g, 0.006 mol) and 3 (0.37 g, 0.0025 mol) were taken in a pressure tube and to it were added abs. EtOH (3 mL) followed by cone. HCl (0.04 g, 0.0012 mol). The tube was tightly screw fitted and was heated at 120 0C for 2 h. The reaction mixture was then cooled, solvents removed under reduced pressure and residue obtained was taken in EtOAc (10 mL). It was washed with water (4 mL), NaHCO3 (4 mL) and brine (5 mL). Drying over Na2SO4 followed by concentration under reduced pressure offered a residue which was further purified by column chromatography (SiO2, 60-120, gravity column chromatography, expected compound getting eluted in CHCl3/MeOH : 94/6) gave 4 (125 mg, 56%) as a light brown solid. [00440] Step-5
Figure imgf000180_0002
1-35 [00441] To a stirred solution of 4 (0.1 g, 0.002 mol) in NMP (8 mL) was added acryloyl chloride (0.1 g, 0.001 mol) drop wise at 0 0C. The reaction was kept at this temperature for 10 min and then allowed to come to rt and stir at this temperature for 1.5 h. It was then quenched with 10% sodium bicarbonate solution (8 mL) and extracted with EtOAc (2x15 mL). The combined EtOAc extract was washed with water (10 mL), brine (10 mL) dried over Na2SO4 and concentrated under reduced pressure. The residue obtained was purified by column chromatography (SiO2, 60-120, gravity column chromatography, expected compound getting eluted in CHCl3/MeOH : 90/10) gave 1-35 (20 mg, 18%) as an off-white solid. 1H NMR (DMSO-de) δ ppm: 2.11 (s, 3H), 3.51 (s, IH), 4.61 (s, 2H), 5.74 (d, J = 9.08 Hz, IH), 6.25 (d, J = 15.84 Hz, IH), 6.45 (s, 2H), 7.02 (s, IH), 7.27-7.45 (m, 5H), 7.91 (d, J = 8.84 Hz, 2H), 8.36 (s, IH), 8.93 (s, IH), 10.09 (s, IH), LCMS: m/e 400 (M+l). EXAMPLE 15
[00442] Preparation of (E)-4-(dimethylamino)-N-(3-(5-methyl-2-(phenylamino)pyrimidin-4- ylamino) phenyl)but-2-enamide 1-38
Figure imgf000181_0001
1-38
[00443] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000181_0002
A) DIEA, n-BuOH, 120 0C, 30 min, MW; B) NMP, 200 0C, 10 min, MW; C) ) oxalyl chloride, CH3CN, 30 min at 0 0C, 2 h at 25 0C, 5 min at 45 0C, D) NMP, 0 0C, 1 h.
[00444] Step-1
Figure imgf000181_0003
[00445] A solution of 1 (2.0 g, 12 mmol), 2 (2.0 g, 18 mmol), DIPEA (2.33 g, 18 mmol) in n- BuOH (20.0 mL) was subjected to microwave irradiation at 120 0C for 30 min. The reaction mixture was then quenched with water (100 mL), extracted with EtOAc (3x100 mL). The combined EtOAc extract was washed with water (100 niL), brine (100 niL), dried over Na2SO4 and concentrated under reduced pressure. The residue obtained was further purified column chromatography (SiO2, 60-120 mesh, EtOAc/CHCl3: 15/85) gave 3 (1.3 g, 45 %) as a dark brown solid.
[00446] Step-2
Figure imgf000182_0001
5
[00447] A solution of 3 (1.0 g, 4.27 mmol), 4 (1.5 g, 16.12 mmol) in NMP (10 mL) was subjected to microwave irradiation (200 0C, 10 min). Then the reaction mixture was cooled, diluted with water (100 mL) and extracted with EtOAc (3x100 mL). The combined EtOAc extract was washed with water (100 mL), brine (100 mL), dried over Na2SO4 and concentrated under reduced pressure gave a residue. The crude residue was further purified by column chromatography (SiO2, 60-120, CHCl3/MeOH : 98/2) gave 5 (0.5 g, 40.3%) as a light brown solid.
[00448] Step-2'
Figure imgf000182_0002
[00449] To a stirred solution of 6' (70 mg, 0.42 mmol) in CH3CN (1.0 mL) was added oxalyl chloride (80 mg, 0.62 mmol) at 0 0C. The reaction mixture was allowed to stir at 0 0C for 1A h and then at rt for 2 h. Finally it was heated at 45 0C for 5 min, cooled and the reaction mixture was taken for the next step without further purification.
[00450]
Figure imgf000183_0001
1-38
[00451] To a stirred solution of 5 (75 mg, 0.12 mmol) in NMP (1 niL) was added 6 at 0 0C. The reaction mixture was stirred at 0 0C for 1 h, quenched with cold water (5 mL), basifϊed with Et3N and extracted with CH2Cl2 (3x10 mL). The combined organic extract was washed with water (5 mL), brine (5 mL) and dried over Na2SO4. Concentration under reduced pressure followed by purification over silica gel (60-120) using 5% methanol in chloroform gave crude compound (20 mg) as a brown gummy solid, which was again taken into dichloromethane and stirred with 10% bicarbonate solution for 30 min, dichloromethane layer separated, dried over Na2SO4 and concentrated to give 1-38 (8 mg, 17%) as a brown solid. 1H NMR (DMSO-d6) δ ppm: 2.15 (s, 3H), 2.32 (s, 6H), 3.21 (d, J= 5.76 Hz, 2H), 6.27 (d, J= 15.36 Hz, IH), 6.84-6.93 (m, 2H), 7.14 (t, J = 7.52 Hz, 2H), 7.27-7.33 (m, 2H), 7.44 (dd, J = 2.04 Hz & 5.08 Hz, IH), 7.53 (d, J= 7.72 Hz, 2H), 7.80 (s, IH), 8.00 (s, IH); LCMS: m/e 402.8 (M+l).
EXAMPLE 16
[00452] Preparation of N-(4-(5 -methy l-2-(phenylamino)pyrimidin-4-ylamino)pheny 1) acrylamide 1-39
Figure imgf000183_0002
1-39 [00453] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000184_0001
A) DIPEA, n-BuOH, 110 °C, 45 min, MW; B) Cone. HCl, n-BuOH, 150uC, 10 min, MW; C) Acryloyl chloride, NMP, 0 °C-30 min, rt-2 h. [00454] Step-l
Figure imgf000184_0002
[00455] A solution of 1 (0.4 g, 2.4 mmol), 2 (0.3 g, 2.6 mmol), DIPEA (0.46 g, 3.6 mmol) in n-BuOH (10 mL) was subjected to microwave irradiation (110 0C, 45 min). The reaction mixture was cooled, quenched with water (20 mL) and extracted with EtOAc (3x15 mL). The combined EtOAc extract was washed with water (20 mL), brine (20 mL), dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, 60-120, CHCl3/MeOH : 99/1) gave 3 (350 mg, 62%) as an off-white solid.
[00456] Step-2
Figure imgf000184_0003
[00457] A solution of 3 (0.2 g, 0.8 mmol), 4 (0.63 g, 6.8 mmol) and con.HCl (0.03 g, 0.8 mmol) in n-BuOH (10 mL) was subjected to microwave irradiation (150 0C, 10 min). Then the reaction mixture was cooled, diluted with water (10 mL), basified with 10% sodium bicarbonate solution and extracted with EtOAc (3x15 mL). The combined EtOAc extract was washed with water (15 mL), brine (15 mL), dried over Na2SO4 and concentrated under reduced pressure. The crude residue was purified by column chromatography (SiO2, 60-120, CHCb/MeOH : 97/3) gave 5 (110 mg, 47%) as a brown colored gummy solid. [00458] Step-3
Figure imgf000185_0001
[00459] To a stirred solution of 5 (0.06 g, 0.2 mmol) in NMP (2 mL) was added acryloyl chloride (0.03 g, 0.3 mmol) at 0 0C. It was allowed to stir at the same temperature for 20 min and then at rt for 2 h. The reaction mixture was quenched with water, basified with 10% sodium bicarbonate solution and extracted with EtOAc (3x10 mL). The combined EtOAc layer was washed with water (10 mL), brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, 60-120) and finally by preparative HPLC gave 1-39 (10 mg, 16%) as an off-white solid. 1H NMR (DMSO-d6) δ ppm: 2.10 (s, 3H), 5.71-5.76 (m, IH), 6.25 (dd, J 2.04 & 16.96 Hz, IH), 6.45 (dd, J = 10.08 & 16.92 Hz, IH), 6.84 (t, J = 7.30 Hz, IH), 7.14-7.18 (m, 2H), 7.62-7.68 (m, 6H), 7.86 (s, IH), 8.26 (s, IH), 8.94 (s, IH), 10.11 (s, IH), LCMS: m/e 346 (M+l).
EXAMPLE 17
[00460] Preparation of N-(3 -(5 -methy l-2-(phenylamino)pyrimidin-4-ylamino)pheny 1) propionamide IR-7
Figure imgf000185_0002
IR-7 [00461] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000186_0001
A) DIPEA, n-BuOH, 120 0C, 30 min, MW; B) NMP, 200 0C, 10 min, MW; C) 6, NMP, 0 0C, 60 min.
[00462] Step-1
Figure imgf000186_0002
[00463] A solution of 1 (2.0 g, 12 mmol), 2 (2.0 g, 18 mmol), DIPEA (2.33 g, 18 mmol) in n- BuOH (20.0 mL) was subjected to microwave irradiation at 120 0C for 30 min. The reaction mixture was then quenched with water (100 mL), extracted with EtOAc (3x100 mL). The combined EtOAc extract was washed with water (100 mL), brine (100 mL), dried over Na2SO4 and concentrated under reduced pressure. The residue obtained was further purified column chromatography (SiO2, 60-120 mesh, EtOAc/CHCl3 : 15/85) gave 3 (1.3 g, 45%) as a dark brown solid. [00464] Step-2
Figure imgf000186_0003
[00465] A solution of 3 (1.0 g, 4.27 mmol), 4 (1.5 g, 16.12 mmol) in NMP (10 mL) was subjected to microwave irradiation (200 0C, 10 min). Then the reaction mixture was cooled, diluted with water (100 mL) and extracted with EtOAc (3x100 mL). The combined EtOAc extract was washed with water (100 mL), brine (100 mL), dried over Na2SO4 and concentrated under reduced pressure gave a residue. The crude residue was further purified by column chromatography (SiO2, CHCl3/MeOH : 98/2) gave 5 ( 0.5 g, 40.3%) as a light brown solid. [00466] Step-3
Figure imgf000187_0001
[00467] To a stirred solution of 5 (75 mg, 0.25 mmol) in NMP (1.0 mL) at 0 0C was added propanoyl chloride (6) (72 mg, 0.75 mmol) and the reaction mixture was stirred at 0 0C for 60 min. The reaction mixture was then quenched with water (5 mL), basified with Et3N and extracted with EtOAc (3x10 mL). The combined EtOAc extract was washed with water (10 mL), brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure. The residue obtained was further purified column chromatography (SiO2, 230-400, methanol/chloroform : 2/98) gave IR-7 (0.025 g, 28.73%) as an off white solid. 1H NMR (DMSO-d6) δ ppm: 1.08 (t, J = 7.6 Hz, 3H), 2.11 (s, 3H), 2.31 (q, J = 7.6 Hz, 2H), 6.81 (t, J = 7.2 Hz, IH), 7.11 (t, J = 8 Hz, 2H), 7.21-7.25 (m, IH), 7.31 (d, J = 8.40 Hz, IH), 7.36 (d, J = 8.00 Hz, IH), 7.66 (d, J = 8.40 Hz, 2H), 7.86 (s, IH), 7.89 (s, IH), 8.35 (s, IH), 8.93 (s, IH), 9.81 (s, IH); LCMS: m/e 348.3 (M+l). EXAMPLE 18
[00468] Preparation of N-(4-methyl-3 -(4-(pyridin-3 -yl)pyrimidin-2-ylamino)phenyl) acrylamide 1-56
Figure imgf000188_0001
1-56
[00469] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000188_0002
1 1-56
A) acryloyl chloride, Et3N, DMF, rt, 12 h [00470] Step-1
Figure imgf000188_0003
1-56
[00471] To a stirred solution of 1 (0.15 g, 0.54 mmol) and Et3N (0.11 g, 1.08 mmol) in DMF (1 mL) at 0 0C was added acryloyl chloride (0.09 g, 1.08 mmol), drop-wise, under N2 atmosphere. The reaction mixture was allowed to come to rt and stirred further 12 h. It was then quenched with ice-cold water (2 mL) and extracted with EtOAc (2x15 mL). The combined EtOAc extract was washed with brine (2 mL), dried over Na2SO4 and concentrated under reduced pressure to get a crude residue. The residue was further purified by preparative HPLC and gave 1-56 (0.060 g, 33%) as a pale yellow solid. 1H NMR (DMSO-d6) δ ppm: 2.19 (s, 3H), 5.72 (dd, J= 2 & 10.08 Hz, IH), 6.22 (dd, J= 2 & 16.92 Hz, IH), 6.45 (dd, J = 10 & 17 Hz, IH), 7.16 (d, J = 8.36 Hz, IH), 7.32 (dd, J = 1.92 & 8.16 Hz, IH), 7.42 (d, J = 5.12 Hz, IH), 7.50- 7.53 (m, IH), 7.95 (d, J= 1.68 Hz, IH), 8.45 (dd, J= 6.16 & 8.16 Hz, IH), 8.49 (d, J= 5.16 Hz, IH), 8.68 (dd, J = 1.56 & 4.76 Hz, IH), 8.95 (s, IH), 9.25 (d, J = 1.56 Hz, IH), 10.08 (s, IH); LCMS: m/e 332.4 (M+l).
EXAMPLE 19 [00472] General method for preparing compounds having an enone-containing warhead, e. g. , 3 -methyl- 1 -(3 -(5 -methyl-2-(phenylamino)pyrimidin-4-ylamino)phenyl)but-2-en- 1 -one 1-47
Figure imgf000189_0001
1-47
[00473] The title compound is prepared according to the schemes, steps and intermediates described below. It is also appreciated by one skilled in the art that 1-47 is an exemplary compounds having enone-containing warheads, and that other compounds having enone- containing warheads can be synthesized in a substantially similar manner according to the schemes, steps and corresponding intermediates described below.
Figure imgf000190_0001
[00474] Compounds 1 and 2 are coupled in the presence of triethylamine to yield compound 3. Compound 3 is treated with analine at elevated temperature to yield compound 4. Saponification of Compound 4 with potassium hydroxide yields acid compound 5, which is coupled to N-O-dimethylhydroxylamine using EDC to yield compound 6. Treatment of Compound 6 at low temperature yields exemplary compound 1-47.
EXAMPLE 20
[00475] Preparation of N-(3-(5-fluoro-2-(4-(2-methoxyethoxy)phenylamino)pyrimidin-4- ylamino)phenyl)acrylamide 1-182
Figure imgf000191_0001
1-182
[00476] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000191_0002
1-182
A) 2, DIPEA, THF, reflux; B) 4, t-amyl alcohol, HOAc, reflux; C) TFA, DCM; D) 7, DIPEA, THF, -10 0C. [00477]
Figure imgf000192_0001
[00478] 1 (800mg, 4.8mmoL), 2 (996mg, 4.8mmoL) and Hunig's base (948uL, 5.75mmoL) were dissolved in THF (2OmL). The reaction mixture was heated at reflux overnight. After cooling, partitioned between water/brine (10 mL), agitated and separated the layers. Dried organic phase over sodium sulfate, and the solvent was removed via rotary evaporation. Titration with EtOAc and Heptane gave after filtration a white solid, Ig. LC/MS (RT = 2.03/(M+l)) 339.1.
[00479]
Figure imgf000192_0002
[00480] 3 (800mg, 2.37mmoL) and 4 (576mg, 2.84mmoL) were suspended in tert-amyl alcohol (14 mL) and acetic acid (5 drops). Heated to reflux for 4h. After cooling, solvent was removed via rotary evaporation. The dark oil was partitioned between water/brine and THF (10 mL each), agitated, and separated layers and dried organic phase over sodium sulfate. The solvent was removed via rotary evaporation to afford a purple solid, 0.55 g. LC/MS (RT = 2.997/(MH-I)) 470.2. Additional 150 mg of product minus the (BOC) protecting group crystallized from the aqueous layer. [00481]
Figure imgf000193_0001
6
[00482] To a solution of 6 (550 mg, 1.17 mmol) in DCM (20 mL) was added TFA (2 niL). Stirred for 30 min at rt for 4h; removed solvent via rotary evaporation and partitioned oil with cold (0 0C) saturated sodium bicarbonate (10 mL) and EtOAc (10 mL), agitated and separated layers. Organic phase was dried over sodium sulfate and the solvent was removed via rotary evaporation to give a dark oil. Flash chromatography using 20%- 100% Heptane/EtOAc gradient using combifiash system gave 309 mg of a light pink solid. LC/MS (RT = 2.78/(M+l)) 370.2. [00483] Step-4
Figure imgf000193_0002
1-182
[00484] A solution of 6 (309 mg, 0.84 mmol) in THF (10 mL) was cooled in a water/ice- MeOH bath (-10 0C). To this was added 7 (71 μL, 0.88 mmoL), stirred for 10 min, then added Hunig's base (145uL, 0.88mmoL), and stirred for 10 min. Partitioned between water/brine (10 niL), agitated and separated the layers. Dried organic phase over sodium sulfate. The solvent was removed via rotary evaporation and triturated with diethyl ether to afford after filtration 285 mg (80%) of an off-white solid. LC/MS (RT = 2.79/(M + H)) 424.2.
EXAMPLE 21
[00485] Preparation of N-(3-(2-(3-chloro-4-(pyridin-2-ylmethoxy)phenylamino)-5- fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-86
Figure imgf000194_0001
1-86
[00486] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 3-chloro-4-(pyridine-2-ylmethoxy)aniline in the place of 4 in Step 2. LC/MS (RT = 2.87/(M + H)) 491.1.
EXAMPLE 22
[00487] Preparation of N-(3-(5-fluoro-2-(4-(2-(2-oxopyrrolidin-l- yl)ethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-92
Figure imgf000195_0001
1-92
[00488] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using l-(2-(4-aminophenoxy)ethyl)pyrrolidin-2-one in the place of 4 in Step 2. LC/MS (RT = 2.718/(M + H)) 477.1.
EXAMPLE 23
[00489] Preparation of N-(3-(5-fluoro-2-(4-(l-hydroxy-2-methylpropan-2- yloxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-93
Figure imgf000195_0002
1-93 [00490] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 2-(4-aminophenoxy)-2-methylpropan-l-ol in the place of 4 in Step 2. LC/MS (RT = 2.724/(M + H)) 438.1.
EXAMPLE 24
[00491] Preparation of Λ/-(3-(5-fluoro-2-(6-isopropoxypyridin-3-ylamino)pyrimidin-4- ylamino)phenyl)acrylamide 1-172
Figure imgf000196_0001
1-172
[00492] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 6-isopropoxypyridin-3 -amine in the place of 4 in Step 2. LC/MS (RT = 2.878/(M + H)) 409.2.
EXAMPLE 25
[00493] Preparation of Λ/-(3-(5-fluoro-2-(2-oxoindolin-5-ylamino)pyrimidin-4- ylamino)phenyl)acrylamide 1-181
Figure imgf000196_0002
1-181 [00494] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 5-aminoindolin-2-one in the place of 4 in Step 2. LC/MS (RT = 2.617/(M + H)) 405.1.
EXAMPLE 26
[00495] Preparation of iV-(2-chlσro-5-(5-fluoio-2-(4-(2- methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-108
Figure imgf000197_0001
1-108
[00496] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using tert-butyl 5-amino-2-chlorophenylcarbamate in the place of 2 in Step 1. LC/MS (RT = 2.852/(M + H)) 458.1.
EXAMPLE 27
[00497] Preparation of N-(2-chloro-5-(5-fluoro-2-(6-isopropoxypyridin-3-ylamino)pyrimidin- 4-ylamino)phenyl)acrylamide 1-107
Figure imgf000198_0001
1-107
[00498] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using tert-butyl 5-amino-2-fluorophenylcarbamate in the place of 2 in Step 1 and 6-isopropoxypyridin-3 -amine in the place of 4 in Step 2. LC/MS (RT = 2.938/(M + H)) 443.1.
EXAMPLE 28
[00499] Preparation of iV-(2-fluoio-5-(5-fluoio-2-(4-(2- methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-87
Figure imgf000198_0002
1-87 [00500] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using tert-butyl 5-amino-2-fluorophenylcarbamate in the place of 2 in Step 1. LC/MS (RT = 2.797/(M + H)) 442.0.
EXAMPLE 29
[00501] Preparation of N-(3-(5-fiuoro-2-(4-((l-methylpiperidin-4- yl)methoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-90
Figure imgf000199_0001
1-90
[00502] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000200_0001
A) 2, DIPEA, THF, reflux; B) 4, Pd(OAc)2, X-Phos, CsCO3, dioxane, reflux, 12 h; C) TFA, DCM; D) 7, DIPEA, THF, -10 0C. [00503] Step-1
Figure imgf000200_0002
[00504] 1 (800mg, 4.8mmoL), 2 (996mg, 4.8mmoL) and Hunig's base (948uL, 5.75mmoL) were dissolved in THF (2OmL). The reaction mixture was heated at reflux overnight. After cooling, partitioned with water/brine (10 mL), agitated and separated the layers. Dried organic phase over sodium sulfate and the solvent was removed via rotary evaporation. Titration with EtOAc and Heptane gave after filtration a white solid, 1 g. LC/MS (RT = 2.03/(M+l)) 339.1. [00505] Step-2
Figure imgf000201_0001
5
[00506] 3 (205 mg, 0.61 mmoL) and 4 (150 mg, 0.73 mmoL) was dissolved in dioxane (4 niL). Degassed the solution for 1 min. Palladium acetate (20mg, 5 moL%), X-Phos ligand (35 mg, 10 moL%) and CSCO3 (325 mg, 1.2 mmoL) were added in that order. Degassed the suspension for 1 min and under argon atmosphere the mixture was heated to reflux for 12 h. After cooling, solvent was removed via rotary evaporation. The dark oil was partitioned between water/brine and EtOAc (5 mL each), agitated, filtered off precipitate and separated layers of the filtrate. Dried organic phase over sodium sulfate. The solvent was removed via rotary evaporation to give a dark oil. Flash chromatography using 0-30% gradient of Heptane/EtOAc afforded light yellow oil. LC/MS (RT = 3.043/(M+l)) 523.2. [00507] Step-3
Figure imgf000201_0002
[00508] To a solution of 5 (144 mg, 0.27 mmol) in DCM (10 mL) was added TFA (1 niL). Stirred for 30 min at rt for 12 h; removed solvent via rotary evaporation and partitioned oil with cold (0 0C) saturated sodium bicarbonate (5 mL) and EtOAc (5 mL), agitated and separated layers. Organic phase was dried over sodium sulfate and the solvent was removed via rotary evaporation to give light yellow foam. LC/MS (RT = 2.723/(M+l)) 423.1. [00509] Step-4
Figure imgf000202_0001
1-90
[00510] A solution of 6 (105 mg, 0.25 mmol) in THF (3 mL) was cooled in water/ice-MeOH bath (-10 0C). To this was added 7 (21 μL, 0.26 mmoL), stirred for 10 min, then added Hunig's base (51 μL, 0.26 mmoL), and stirred for 10 min. Partitioned with water/brine (5 mL), agitated and separated the layers. Dried organic phase over sodium sulfate. The solvent was removed via rotary evaporation afford a light yellow foam. LC/MS (RT = 2.726/(M + H)) 477.1.
EXAMPLE 30
[00511] Preparation of Λ/-(3-(5-fluoro-2-(6-((2-methoxyethyl)(methyl)amino)pyridin-3- ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-77
Figure imgf000203_0001
1-77
[00512] The title compound was prepared according to the schemes, steps and intermediates described in Example 29, by using N2-(2-methoxyethyl)-N2-methylpyridine-2,5-diamine in the place of 4 in Step 2. LC/MS (RT = 2.739/(M + H)) 438.1.
EXAMPLE 31
[00513] Preparation of l-(6-(5-fluoro-2-(6-methoxypyridin-3-ylamino)pyrimidin-4-ylamino)- 2H-benzo [b] [ 1 ,4]oxazin-4(3H)-yl)prop-2-en- 1 -one I- 194
Figure imgf000203_0002
[00514] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000204_0001
A) 2, DIPEA, THF, reflux; B) 4, HOAc, tert-amyl alcohol, reflux, 12 h; C) TFA, DCM; D) 7, DIPEA, DCM, NMP, -10 0C.
[00515] Step-1
Figure imgf000204_0002
[00516] 1 (186 mg, 1.1 mmoL), 2 (280mg, l.lmmoL) and Hunig's base (220 μL, 1.3 mmoL) were dissolved in THF (6 rnL). The reaction mixture was heated at reflux overnight. After cooling, partitioned with water/brine (6mL), agitated and separated the layers. Dried organic phase over sodium sulfate and the solvent was removed via rotary evaporation to give a tan solid. LC/MS (RT = 3.008/(M+l)) 381.1. [00517]
Figure imgf000205_0001
[00518] 3 (215 mg, 0.56 mmoL) and 4 (83 mg, 0.66 mmoL) was suspended in tert-amyl alcohol (6 niL) and acetic acid (3 drops). Heated to reflux for 12 h. After cooling, solvent was removed via rotary evaporation. The dark oil was partitioned between water/brine and EtOAc (5 rnL each), agitated, and separated layers and dried organic phase over sodium sulfate. The solvent was removed via rotary evaporation to afford an oil. Flash chromatography using 30- 70% gradient of heptane/ethyl acetate on combiflash system gave a tan solid. LC/MS (RT = 2.011/(M+l)) 469.2. [00519] Step-3
Figure imgf000205_0002
[00520] To a solution of 5 (200 mg, 0.43 mmol) in DCM (10 mL) was added TFA (1 mL). Stir for 30 min at rt for 12 h; removed solvent via rotary evaporation and partitioned oil between cold (0 0C) saturated sodium bicarbonate (5 rnL) and EtOAc (5 mL), agitated and separated layers. Organic phase was dried over sodium sulfate and the solvent was removed via rotary evaporation to give a pink solid. LC/MS (RT = 2.782/(M+l)) 369.1. [00521] Step-4
Figure imgf000206_0001
1-194
[00522] A solution of 6 (150 mg, 0.41 mmol) in DCM (2 mL) and NMP (0.5 mL) was cooled in water/ice-MeOH bath (-10 0C). To this was added 7 (34 μL, 0.43 mmoL), stirred for lOmin, then added Hunig's base (70 μL, 0.43 mmoL), and stirred for 10 min. Partitioned between water/brine (5 mL), agitated and separated the layers. Dried organic phase over sodium sulfate. Purified directly via flash chromatography using 20-80% gradient of heptane/ethyl acetate to give a pink solid. LC/MS (RT = 2.8/(M + H)) 423.1.
EXAMPLE 32 [00523] Preparation of l-(6-(5-fluoro-2-(4-(2-methoxyethoxy)phenylamino)pyrimidin-4- ylamino)-2H-benzo[b][ 1 ,4]oxazin-4(3H)-yl)prop-2-en- 1 -one 1-141
Figure imgf000207_0001
1-141
[00524] The title compound was prepared according to the schemes, steps and intermediates described in Example 31, by using 4-(2-methoxyethoxy)aniline in the place of 4 in Step 2. LC/MS (RT = 2.845/(M + H)) 466.2.
EXAMPLE 33
[00525] Preparation of l-(6-(5-fluoro-2-(6-methoxypyridin-3-ylamino)pyrimidin-4- ylamino)indolin- 1 -yl)prop-2-en- 1 -one 1-166
Figure imgf000207_0002
1-166 [00526] The title compound was prepared according to the schemes, steps and intermediates described in Example 31, by using tert-butyl 6-aminoindoline-l-carboxylate in the place of 2 in Step 1. LC/MS (RT = 2.825/(M + H)) 407.1.
EXAMPLE 34
[00527] Preparation of l-(5-(5-fluoro-2-(6-methoxypyridin-3-ylamino)pyrimidin-4- ylamino)isoindolin-2-yl)prop-2-en-l-one 1-165
Figure imgf000208_0001
[00528] The title compound was prepared according to the schemes, steps and intermediates described in Example 31, by using tert-butyl 5-aminoisoindoline-l-carboxylate in the place of 2 in Step 1. LC/MS (RT = 2.751/(M + H)) 407.1.
EXAMPLE 35
[00529] Preparation of l-(6-(4-(3-chlorophenylamino)-5-fluoropyrimidin-2-ylamino)-2H- benzo[b] [1 ,4]oxazin-4(3H)-yl)prop-2-en- 1 -one 1-149
Figure imgf000208_0002
1-149 [00530] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000209_0001
A) 2, DIPEA, THF, reflux; B) 4, HOAc, tert-amyl alcohol, reflux, 12 h; C) TFA, DCM; D) 7, DIPEA, THF, -10 0C. [00531] Step-1
Figure imgf000209_0002
3
[00532] 1 (484 mg, 2.9 mmoL), 2 (305 mg, 2.9 mmoL) and Hunig's base (526 μL, 3.5 mmoL) were dissolved in THF (10 mL). The reaction mixture was heated at reflux overnight. After cooling, partitioned between water/brine (10 mL), agitated and separated the layers. Dried organic phase over sodium sulfate and the solvent was removed via rotary evaporation. Flash chromatography using a gradient of 0-30% heptane/ethyl acetate on combiflash system gave a white solid. LC/MS (RT = 2.03/(M+l)) 339.1. [00533] Step-2
Figure imgf000210_0001
[00534] 3 (150 mg, 0.58 mmoL) and 4 (175 mg, 0.7 mmoL) were suspended in tert-amyl alcohol (8 mL) and acetic acid (3 drops). Heated to reflux for 12h. After cooling, solvent was removed via rotary evaporation. The dark oil was partitioned between water/brine and EtOAC (5 mL each), agitated, and separated layers and dried organic phase over sodium sulfate. The solvent was removed via rotary evaporation to afford a dark oil. Flash chromatography using a gradient of 0-25% heptane/ethyl acetate on combiflash system gave a white solid. LC/MS (RT = 2.997/(M+l)) 470.2. [00535] Step-3
Figure imgf000210_0002
[00536] To a solution of 5 (180 mg, 0.38 mmol) in DCM (10 mL) was added TFA (1 mL). Stirred for 30 min at rt for 4h; removed solvent via rotary evaporation and partitioned oil between cold (0 0C) saturated sodium bicarbonate (5 mL) and EtOAc (5 mL), agitated and separated layers. Organic phase was dried over sodium sulfate and the solvent was removed via rotary evaporation to give light yellow solid. LC/MS (RT = 2.723/(M+l)) 423.1. [00537]
Figure imgf000211_0001
1-149
[00538] A solution of 6 (150 mg, 0.4 mmol) in THF (3 niL) was cooled in water/ice-MeOH bath (-10 0C). To this was added 7 (34 μL, 0.42 mmoL), stirred for 10 min, then added Hunig's base (70 μL, 0.42 mmoL), and stirred for lOmin. Partitioned between water/brine (5mL), agitated and separated the layers. Dried organic phase over sodium sulfate. The solvent was removed via rotary evaporation to afford a light yellow solid. Flash chromatography using gradient of 10-50% heptane/ethyl acetate on combiflash system gave a white solid. LC/MS (RT = 2.945/(M + H)) 426.
EXAMPLE 36
[00539] Preparation of 5-(2-(4-acryloyl-3,4-dihydro-2H-benzo[b][l,4]oxazin-6-ylamino)-5- fluoropyrimidin-4-ylamino)indolin-2-one 1-130
Figure imgf000211_0002
1-130
[00540] The title compound was prepared according to the schemes, steps and intermediates described in Example 35, by using 5-aminoindolin-2-one in the place of 2 in Step 1. LC/MS (RT = 2.673/(M + H)) 447.1. EXAMPLE 37
[00541] Preparation of 4-(3-acrylamidophenylamino)-2-(phenylamino)pyrimidine-5- carboxamide 1-230
Figure imgf000212_0001
1-230
[00542] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000212_0002
A) 2, NEt3, DCM, 0 0C to rt; B) 4, DIPEA, THF, rt, 12 h; C) 6, DIPEA, t-amyl alcohol, reflux, 4 h; D) TFA, DCM, rt; E) 7, NEt3, THF, 0 0C; F) TFA, TfOH, DCM, rt. [00543]
Figure imgf000213_0001
[00544] 1 (500 mg, 2.4 mmoL, prepared from 2,4-dihydroxypyrimidine-5-carboxylic acid according to J. Med. Chem. 50: 591 (2007) and US 2007/0072851) was dissolved in DCM (10 rnL) and chilled in an ice/water bath (0 0C). 2 (309 μL, 2.4 mmoL) was added and the mixture stirred for 10 min. Triethylamine (365 μL, 2.6 mmol) was added and the mixture was allowed to warm to rt and stir for 30min. The solvent was reduced in volume via rotary evaporation and directly purified by flash chromatography using a gradient of 0-30% heptane/ethyl acetate on combiflash system to give a white solid. LC/MS (RT = 2.789/(M+l)) 312. [00545] Step-2
Figure imgf000213_0002
[00546] 3 (170 mg, 0.55 mmoL), 4 (113 mg, 0.55 mmoL) and Hunig's base (108 μL, 0.65 mmoL) were dissolved in THF (6 mL). Stirred at rt for 12 h. Partitioned between water/brine, agitated, and separated layers and dried organic phase over sodium sulfate. The solvent was removed via rotary evaporation to afford after titration with EtOAc a white solid. LC/MS (RT = 3.123/(M+1)) 484. [00547] Step-3
Figure imgf000213_0003
[00548] 5 (230 mg, 0.48 mmol), 6 (126 μL, 1.4 mmoL) and Hunig's base (94 μL, 0.57 mmoL) is dissolved in t-amyl alcohol (6 mL). Heat to reflux for 4 h, cool and water was added to the solid mass. Agitated, filtered and dried to give a white solid. LC/MS (RT = 3.182/(M+1))
541.2.
[00549] Step-4
Figure imgf000214_0001
8
[00550] 7 (180 mg, 0.33 mmol) was suspended in DCM (10 mL) and treated with TFA (1 mL). Stirred overnight at rt. Diluted with DCM (40 mL) and washed with NaOH (IN, 25mL). Agitated, precipitate formed, filtered and dry to give a white solid. LC/MS (RT = 2.934/(M+l)) 441.1. [00551] Step-5
Figure imgf000214_0002
1-231
[00552] A suspension of 8 (130 mg, 0.29 mmol) in THF (6 mL) was cooled in water/ice (0 0C). To this was added 9 (25 μL (plus additional 5 μL), 0.38 mmoL (total)), then added triethyl amine (43 μL (plus additional 11 μL), 0.38 mmoL(total)), and stirred for a total time of 1 h. Water was added, agitated, filtered off remaining precipitate and discarded. The filtrate was dried over sodium sulfate. The solvent was removed via rotary evaporation to afford a yellow solid. Flash chromatography using a gradient of 0-25% heptane/ethyl acetate on combiflash system gave a white solid. LC/MS (RT = 2.964/(M + H)) 495.1. [00553]
Figure imgf000215_0001
1-230
[00554] To a suspension of 1-231 (30 mg, 0.061 mmol) in DCM (4 niL) was added TFA (200 μL) and triflic acid (68 μL, 0.61mmoL). Stirred at it 1 h. Removed solvent under reduce pressure via rotary evaporation and partitioned with cold (0 0C) saturated sodium bicarbonate (10 mL) and EtOAc (10 mL), agitated and separated layers. Dried organic layer over sodium sulfate and the solvent was removed via rotary evaporation to afford after titration with diethyl ether a white solid. LC/MS (RT = 2.715/(M + H)) 375.1.
EXAMPLE 38
[00555] Preparation of 4-(3-acrylamidophenylamino)-N-phenyl-2-(phenylamino)pyrimidine- 5-carboxamide 1-222
Figure imgf000215_0002
1-222
[00556] The title compound was prepared according to the schemes, steps and intermediates described in Example 37, by using aniline in the place of 2 in Step 1 and omitting Step 6. LC/MS (RT = 2.991/(M + H)) 451.2. EXAMPLE 39
[00557] Preparation of 4-(3-acrylamidophenylamino)-N-cyclopropyl-2-
(phenylamino)pyrimidine-5 -carboxamide 1-221
Figure imgf000216_0001
1-221
[00558] The title compound was prepared according to the schemes, steps and intermediates described in Example 37, by using cyclopropylamine in the place of 2 in Step 1 and omitting Step 6. LC/MS (RT = 2.838/(M + H)) 415.2.
EXAMPLE 40
[00559] Preparation of 4-(3-acrylamidophenylamino)-2-(3-methoxyphenylamino)pyrimidine- 5 -carboxamide 1-210
Figure imgf000216_0002
1-210
[00560] The title compound was prepared according to the schemes, steps and intermediates described in Example 37, by using 3 -methoxy aniline in the place of 6 in Step 3. LC/MS (RT = 2.743/(M + H)) 405.1. EXAMPLE 41
[00561] Preparation of 4-(3 -acrylamidophenylamino)-2-(6-methoxypyridin-3 - ylamino)pyrimidine-5 -carboxamide 1-209
Figure imgf000217_0001
1-209
[00562] The title compound was prepared according to the schemes, steps and intermediates described in Example 37, by using 6-methoxypyridin-3 -amine in the place of 6 in Step 3. LC/MS (RT = 2.657/(M + H)) 406.2.
EXAMPLE 42
[00563] Preparation of l-{6-[5-Acetyl-2-(6-methoxy-pyridin-3-ylamino)-pyrimidin-4- ylamino]-2,3-dihydro-benzo[l,4]oxazin-4-yl}-propenone 1-170
Figure imgf000217_0002
[00564] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000218_0001
A) 2, DIPEA, THF, 70 0C, 16 h; B) (a) 4, PdCl2(PPh3)2, DMF, 70 0C; (b) IN HCl, acetone, 60 0C, 15 min; C) HCl/dioxane, DCM; D) 7, DIPEA, NMP, DCM, -20 0C to rt; E) 9, pTsOH, dioxane, 100 0C, 15 min. [00565] Step-1
Figure imgf000218_0002
3
[00566] A mixture of 499 mg of 1 (2.19 mmol), 547 mg of 2 (2.19 mmol), and 500 uL of N,N-diisopropylethylamine in 20 mL of anhydrous tetrahydrofuran was heated at 70 0C overnight. After cooling down, the reaction mixture was concentrated, and subject to aqueous workup with 50 mL of EtOAc, 20 mL of sodium bicarbonate solution, brine, and dried over anhydrous sodium sulfate. After filtration and concentration, the residue was passed through a short silica cartridge, eluted with heptanes/EtOAc (v/v 3/1), giving 815 mg of a slight yellow solid (84%). LC-MS: m/z 441.0 (ES+), 439.0 (ES-). [00567]
Figure imgf000219_0001
5
[00568] A mixture of intermediate 3 (815 mg, 1.85 mmol), 4 (740 mg, 1.1 equiv.), 27 mg of dichlorobis(triphenylphosphine)palladium (II) (2% mol) in 6 mL of anhydrous DMF was purged with nitrogen for 30 min. The reaction mixture was then heated at 70 0C overnight. LC-MS showed 70% conversion. After cooling down, 30 mL of ethyl acetate and 760 mg of potassium fluoride in 5 mL of water was added, and the mixture was stirred at rt for at least 2 hr. The white precipitate was filtered out, and the organic layer was separated, washed with water, brine, and dried over anhydrous sodium sulfate.
[00569] After filtration and concentration, the residue was dissolved in 20 mL of acetone, followed by additon of 3 mL of 1.0 N aqueous HCl solution. The mixture was heated at 60 0C for 15 min, and concentrated under reduced pressure. Normal workup was done using 50 mL of EtOAc, 10 mL of saturated sodium bicarbonate solution, brine, anhydrous sodium sulfate. After concentration, the residue was purified by flash column chromatography on silica gel, giving 405 mg of yellow solid (70% based on consumed starting material), also recovering intermediate 3 183 mg. LC-MS: m/z 405.1 (ES+), 403.1 (ES-). [00570] Step-3
Figure imgf000219_0002
6
[00571] To a mixture of 1.28 g of intermediate 1-2 in 10 mL of dichloromethane, was added 10 mL of 4.0 N HCl in dioxane. After stirring at rt overnight, the solvent was removed, and the residue was dried in vacuum. LC-MS: m/z 305.1 (ES+), 303.1 (ES-). [00572]
Figure imgf000220_0001
8
[00573] Under N2, to a mixture of the intermediate 6 obtained above, 1 mL of DIPEA in 10 mL of NMP and 10 mL of dichloromethane at -20 0C, was added 275 uL of 7 (1.1 equiv). The reaction was continued for 5 min, then quenched with 1 mL of isopropyl alcohol. The reaction mixture was warmed up to rt, and extracted with 100 mL of EtOAc, washed with water 10 mL x 2, brine, dried over sodium sulfate. After filtration and concentration, the residue was purified by flash column chromatography with eluent heptanes/EtOAc (v/v 2/3), giving yellow solid 1-3 450 mg (40 %). LC-MS: m/z 359.1 (ES+), 357.1 (ES-). [00574] Step-5
Figure imgf000220_0002
1-170
[00575] The mixture of 30 mg intermediate 8 (84 μmol) and 13 mg of 9 (1.2 equiv) in 1 mL of 0.08 M p-TsOH dioxane solution was heated at 100 0C for 15 min. After cooling down, the reaction mixture was subject to regular work up with 50 mL of EtOAc, aqueous sodium bicarbonate, brine, and dried over anhydrous sodium sulfate. After concentration, the residue was purified by column chromatography on silica gel with heptane/EtOAc (v/v 1/4) as eluent, giving 22.8 mg pale white solid (61%). LC-MS: m/z = 447.1 (ES+), 445.2 (ES-). EXAMPLE 43
[00576] Preparation of l-{6-[5-Acetyl-2-(4-morpholin-4-yl-phenylamino)-pyrimidin-4- ylamino]-2,3-dihydro-benzo[l,4]oxazin-4-yl}-propenone 1-169
Figure imgf000221_0001
1-169
[00577] The title compound was prepared according to the schemes, steps and intermediates described in Example 42, by using 4-morpholin-4-yl-phenylamine in the place of 9 in Step 5. LC-MS: m/z 501.1 (ES+), 499.2 (ES-).
EXAMPLE 44
[00578] Preparation of l-{6-[5-Acetyl-2-(6-morpholin-4-yl-pyridin-3-ylamino)-pyrimidin-4- ylamino]-2,3-dihydro-benzo[l,4]oxazin-4-yl}-propenone 1-168
Figure imgf000221_0002
1-168
[00579] The title compound was prepared according to the schemes, steps and intermediates described in Example 42, by using 3-amino-[6-morpholin-4-yl]-pyridine in the place of 9 in Step 5. LC-MS: m/z 502.2 (ES+), 500.3 (ES-). EXAMPLE 45
[00580] Preparation of l-{6-[5-Acetyl-2-(l-methyl-lH-indazol-6-ylamino)-pyrimidin-4- ylamino]-2,3-dihydro-benzo[l,4]oxazin-4-yl}-propenone 1-154
Figure imgf000222_0001
1-154
[00581] The title compound was prepared according to the schemes, steps and intermediates described in Example 42, by using 1 -methyl- lH-indazol-6-ylamine in the place of 9 in Step 5. LC-MS: m/z 470.1 (ES+), 468.1 (ES-).
EXAMPLE 46
[00582] Preparation of l-{6-[5-Acetyl-2-(lH-indazol-6-ylamino)-pyrimidin-4-ylamino]-2,3- dihydro-benzo[l,4]oxazin-4-yl}-propenone 1-153
Figure imgf000222_0002
1-153
[00583] The title compound was prepared according to the schemes, steps and intermediates described in Example 42, by using lH-indazole-6-ylamine in the place of 9 in Step 5. LC-MS: m/z 456.1 (ES+), 454.2 (ES-). EXAMPLE 47
[00584] Preparation of l-{4-[5-Acetyl-4-(4-acryloyl-3,4-dihydro-2H-benzo[l,4]oxazin-6- ylamino)-pyrimidin-2-ylamino]-phenyl} -pyrrolidin-2-one 1-152
Figure imgf000223_0001
1-152
[00585] The title compound was prepared according to the schemes, steps and intermediates described in Example 42, by using l-(4-amino-phenyl)-pyrrolidin-2-one in the place of 9 in Step 5. LC-MS: m/z 456.1 (ES+), 454.2 (ES-).
EXAMPLE 48
[00586] Preparation of l-(6-{5-Acetyl-2-[4-(2-methoxy-ethoxy)-phenylamino]-pyrimidin-4- ylamino} -2,3-dihydro-benzo[ 1 ,4]oxazin-4-yl)-propenone 1-150
Figure imgf000223_0002
1-150
[00587] The title compound was prepared according to the schemes, steps and intermediates described in Example 42, by using 4-(2-methoxy-ethoxy)-phenylamine in the place of 9 in Step 5. LC-MS: m/z 490.2 (ES+), 488.3 (ES-). EXAMPLE 49
[00588] Preparation of 5-[5-Acetyl-4-(4-acryloyl-3,4-dihydro-2H-benzo[l,4]oxazin-6- ylamino)-pyrimidin-2-ylamino]- 1 ,3-dihydro-indol-2-one 1-129
1-129
[00589] The title compound was prepared according to the schemes, steps and intermediates described in Example 42, by using 5-amino-l,3-dihydro-indol-2-one in the place of 9 in Step 5. LC-MS: m/z 471.1 (ES+), 469.2 (ES-).
EXAMPLE 50
[00590] Preparation of l-(6-{5-Acetyl-2-[6-(2-hydroxy-ethoxy)-pyridin-3-ylamino]- pyrimidin-4-ylamino}-2,3-dihydro-benzo[l,4]oxazin-4-yl)-propenone 1-128
Figure imgf000224_0002
1-128
[00591] The title compound was prepared according to the schemes, steps and intermediates described in Example 42, by using 2-(5-amino-pyridin-2-yloxy)-ethanol in the place of 9 in Step 5. LC-MS: m/z 477.1 (ES+), 475.2 (ES-). EXAMPLE 51
[00592] Preparation of N-{3-[5-Acetyl-2-(6-methoxy-pyridin-3-ylamino)-pyrimidin-4- ylamino]-phenyl}-acrylamide 1-189
Figure imgf000225_0001
1-189
[00593] The title compound was prepared according to the schemes, steps and intermediates described in Example 42, by using tert-butyl 3-aminophenylcarbamate in the place of 2 in Step 1 and 5-amino-2-methoxypyridine in the place of 9 in Step 5. LC-MS: m/z 405.1 (ES+), 403.2 (ES-).
EXAMPLE 52
[00594] Preparation of N- {3-[5-Acetyl-2-(6-methoxy-pyridin-3-ylamino)-pyrimidin-4-yloxy]- phenyl}-acrylamide 1-188
Figure imgf000225_0002
1-188
[00595] The title compound was prepared according to the schemes, steps and intermediates described in Example 42, by using tert-butyl 3-hydroxyphenylcarbamate in the place of 2 in Step 1 and 5-amino-2-methoxypyridine in the place of 9 in Step 5. LC-MS: m/z 406.2 (ES+), 404.1 (ES-). EXAMPLE 53
[00596] Preparation of l-{3-[5-Acetyl-2-(6-methoxy-pyridin-3-ylamino)-pyrimidin-4- ylamino]-azetidin- 1 -yl} -propenone 1-187
Figure imgf000226_0001
1-187
[00597] The title compound was prepared according to the schemes, steps and intermediates described in Example 42, by using 3-amino-N-Boc-azetidine in the place of 2 in Step 1 and 5- amino-2-methoxypyridine in the place of 9 in Step 5. LC-MS: m/z 369.1 (ES+), 367.2 (ES-).
EXAMPLE 54
[00598] Preparation of N-(3-{5-Acetyl-2-[4-(2-methoxy-ethoxy)-phenylamino]-pyrimidin-4- ylamino}-phenyl)-acrylamide 1-124
Figure imgf000226_0002
1-124
[00599] The title compound was prepared according to the schemes, steps and intermediates described in Example 42, by using tert-butyl 3-aminophenylcarbamate in the place of 2 in Step 1 and 4-(2-methoxy-ethoxy)-phenylamine in the place of 9 in Step 5. LC-MS: m/z 448.2 (ES+), 446.3 (ES-). EXAMPLE 55
[00600] Preparation of N-(3-{5-Acetyl-2-[6-(2-methoxy-ethoxy)-pyridin-3-ylamino]- pyrimidin-4-ylamino} -phenyl)-acrylamide 1-122
Figure imgf000227_0001
1-122
[00601] The title compound was prepared according to the schemes, steps and intermediates described in Example 42, by using tert-butyl 3-aminophenylcarbamate in the place of 2 in Step 1 and 6-(2-Methoxy-ethoxy)-pyridin-3-ylamine in the place of 9 in Step 5. LC-MS: m/z 449.2 (ES+), 447.1 (ES-).
EXAMPLE 56
[00602] Preparation of N-(3-{5-Acetyl-2-[6-(2-hydroxy-ethoxy)-pyridin-3-ylamino]- pyrimidin-4-ylamino} -phenyl)-acrylamide 1-121
Figure imgf000227_0002
1-121
[00603] The title compound was prepared according to the schemes, steps and intermediates described in Example 42, by using tert-butyl 3-aminophenylcarbamate in the place of 2 in Step 1 and 2-(5-Amino-pyridin-2-yloxy)-ethanol in the place of 9 in Step 5. LC-MS: m/z 435.1 (ES+), 433.2 (ES-). EXAMPLE 57
[00604] Preparation of 4-(3-acrylamidophenoxy)-2-(3-methoxyphenylamino)-pyrimidine-5- carboxylic acid phenylamide 1-200
Figure imgf000228_0001
1-200
[00605] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000229_0001
A B
Figure imgf000229_0002
A) 2, NaH, THF, O 0C; B) NaOH, THF, MeOH; C) 5, TBTU, DIPEA, CH3CN, O 0C; D) MCPBA, CH2Cl2, O 0C; E) 8, 50 0C, 3 h; F) TFA, CH2Cl2; G) 11, DIPEA, CH2Cl2 [00606] Step l
Figure imgf000229_0003
3
[00607] To a stirred solution of (3-hydroxyphenyl)carbamic acid tert-butyl ester 2 (1.79 g,
8.59 mmol) at O 0C was added a suspension of sodium hydride (60% dispersion in mineral oil) (0.34 g, 8.9 mmol) in anhdyrous THF (30 mL). The mixture was stirred at 0 0C for 20 minutes. The phenoxide solution was then added dropwise at 0 0C to a solution of 4-chloro-(2- methylsulfanyl)pyrimidine-5-carboxylic acid ethyl ester 1 (2 g, 8.59 mmol) in THF (20 mL). The reaction mixture was stirred at 0 0C for 2 hours. The reaction mixture was diluted with ethyl acetate (150 mL) and washed with water (50 mL) and then brine (50 mL). The organic layer was dried over sodium sulphate, filtered and concentrated in vacuo. The crude product was washed with CH2Cl2:hexane (1 :9) to afford the title compound 3 as a white solid (2.43 g, 70%). [00608] Step-2
Figure imgf000230_0001
4 [00609] To a stirred solution of 4-(3-tert-butoxycarbonylaminophenoxy)-2-(methylsulfanyl- pyrimidine)-5-carboxylic acid ethyl ester 3 (2 g, 4.93 mmol) in THF (60 mL), was added methanol (60 mL) at -10 0C, followed by aqueous sodium hydroxide (0.3 g, 30 mL water, 7.5 mmol). The reaction mixture was allowed warm to room temperature and was stirred for 1 hour. The reaction mixture was diluted with water (50 mL), acidified with citric acid and the resulting solid was collected by filtration and washed with ice cold water (50 mL) to yield 4 as a white solid. (1.52 g, 82%). [00610] Step-3
Figure imgf000230_0002
[00611] To a stirred solution of 4-(3-ter£-butoxycarbonylaminophenoxy)-2- (methylsulfanyl)pyrimidine-5-carboxylic acid 4 (2.0 g, 5.29 mmol) and TBTU (2.55 g, 7.94 mmol) in acetonitrile (30 mL) at 00C was added DIPEA (1.36g, 10.6 mmol) followed by aniline 5 (0.60 g, 6.35 mmol). The reaction was stirred at room temperature for 2 hours. After completion of the reaction the reaction mixture was poured into ice cold water (100 mL) and the white solid obtained was collected by filtration and washed with ice cold water (20 mL), dried under in vacuo to afford the title compound 6 (1.79 g, 75%). [00612]
Figure imgf000231_0001
[00613] To a stirred solution of [3-(2-methylsulfanyl-5-phenylcarbamoylpyrimidin-4-yloxy)- phenylj-carbamic acid tert-butyl ester 6 (1.5 g, 3.31 mmol) in CH2Cl2 at 0 °C was added a solution m-CPBA (70%, 1.62 g, 2 eq) in CH2Cl2 (10 mL). The reaction mixture was allowed to warm to room temperature and was stirred for 12 h. The reaction was quenched with saturated aqueous NaHCO3 and the whole was extracted with EtOAc. The organic layer was washed with brine, dried over Na2SO4, filtered, and concentrated in vacuo. The crude product was washed with CH2Cl2:hexane (1 :9) to afford the title compound 7 as a white solid (1.16 g, 73%). [00614] Step-5
Figure imgf000231_0002
[00615] Excess 3 -methoxy aniline (8) (2 mL) was added to solid [3-(2-methanesulfonyl-5- phenylcarbamoyl-pyrimidin-4-yloxy)-phenyl]-carbamic acid tert-butyi ester 7 (0.5 g, 1.03 mmol) and the resulting mixture was heated to 50 0C under an argon atmosphere for 3 hours. The reaction mixture was cooled to room temperature and diluted with ethyl acetate/hexane (1 :1, 20 mL) and the resulting precipitate filtered and washed with ethyl acetate/hexane (1 :1, 10 mLl) to afford the desired product 9 as a white solid (0.40 g, 75% yield). [00616] Step-6
Figure imgf000231_0003
[00617] To a solution of {3-[2-(3-methoxy-phenylamino)-5-phenylcarbamoyl-pyrimidin-4- yloxy]-phenyl}-carbamic acid tert-butyl ester 9 (0.3 g, 0.56 mmol) in CH2Cl2 (10 rnL) was added trifluoroacetic acid (2 rnL) and the mixture was stirred at room temperature for 1 hour. Solvents were removed under reduced pressure and the residue was dissolved in CH2Cl2, washed with 10% aqueous NaHCO3 solution, dried (Na2SO4), filtered, and evaporated under reduced pressure to provide the free amine 10 as white solid. [00618] Step-7
Figure imgf000232_0001
1-200
[00619] To a stirred solution of amine 10 (0.24 g, 0.56 mmol) in dichloromethane (2OmL) under argon atmosphere cooled to -70 0C was added DIPEA (0.072 g, 0.56 mmol) followed by drop wise addition of acryloyl chloride (0.050 g, 0.56 mmol). The resulting mixture was stirred at -70 0C for 5 minutes, and the reaction mixture diluted with CH2Cl2 (50 mL) and then was washed with saturated aqueous NaCl solution (10 mL). The organic layer was dried (Na2SO4), filtered and evaporated under reduced pressure. The residue was purified by flash chromatography on silica gel using (MeOH-CHCl3 5:95) as eluent to provide the target compound 11 (0.094 g, 35%) as white solid: 1H NMR (200 MHz, DMF-d7) δ 8.9 (s, IH), 8.10- 7.70 (m, 6H), 7.60-7.10 (m, 6H),6.60 (m, 2H) 6.40 (dd, IH, J= 8.0, 2.0 Hz ), 5.80 (m, 2H), 3.70 (s, 3H).
EXAMPLE 58
[00620] Preparation of 4-(3-acrylamidophenoxy)-2-(6-methoxypyridin-3-ylamino)- pyrimidine-5-carboxylic acid phenylamide 1-159
Figure imgf000232_0002
1-159 [00621] The title compound was prepared according to the schemes, steps and intermediates described in Example 57 by using 6-methoxy-3-aminopyridine in place of 8 in Step 5. 1H NMR (200 MHz, DMSO-d6 δ 8.90 (s, IH), 8.20 (brs, IH), 7.90-7.60 (m, 4H), 7.45(m, 4H), 7.10 (m, 2H), 6.50 (m, IH), 6.20 (m, 2H), 5.90 (dd, J= 8.0, 2.0 Hz, IH), 3.90 (s, 3H).
EXAMPLE 59
[00622] Preparation of 4-(3-acrylamidophenoxy)-2-(3-methoxyphenylamino)-pyrimidine-5- carboxylic acid cyclopropylamide 1-177
Figure imgf000233_0001
1-177
[00623] The title compound was prepared according to the schemes, steps and intermediates described in Example 57 by using cyclopropylamine in place of 5 in Step 3. 1H NMR (200 MHz, CD3OD) δ 9.0 (s, IH), 7.90 (brs, IH), 7.50 (m, 3H), 7.0 (m, 4H), 6.50 (m, IH), 6.40 (d, J = 8.0 Hz, 2H), 5.80 (dd, J= 8.2, 3.0 Hz, IH), 3.60 (s, 3H), 0.90 (m, 2H), 0.62 (m, 2H).
EXAMPLE 60
[00624] Preparation of 4-(3-acrylamidophenoxy)-2-(6-methoxypyridin-3-ylamino)- pyrimidine-5-carboxylic acid cyclopropylamide 1-176
Figure imgf000233_0002
1-176
[00625] The title compound was prepared according to the schemes, steps and intermediates described in Example 57 by using cyclopropylamine in place of 5 in Step 3 and 6-methoxy-3- aminopyridine in place of 8 in Step 5. 1H NMR (200 MHz, CD3OD) δ 8.90 (s, IH), 7.95 (brs, IH), 7.90-7.82 (m, 3H), 7.40 (m, 3H), 6.98 (d, J = 6.0 Hz, IH), 6.42 (m, 2H), 5.90 (dd, J = 8.0, 2.0 Hz, IH), 3.90 (s, 3H), 0.95 (m, 2H), 0.83 (m, 2H).
EXAMPLE 61
[00626] Preparation of 4-(3-acrylamidophenoxy)-2-(3-methoxyphenylamino)pyrimidine-5- carboxylic acid amide 1-178
Figure imgf000234_0001
1-178
[00627] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000235_0001
1-178
A) 2, NaH, THF, O 0C; B) LiOH, THF, H2O; C) 5, TBTU, DIPEA, CH3CN; D) MCPBA, CHCl3, O 0C; E) 8, DMA, 90 0C, 24 h; F) 4N HCl, dioxane; G) 11, CH2Cl2; H) triflic acid, TFA, CH2Cl2 [00628]
Figure imgf000236_0001
3
[00629] Step 1 was carried out in a manner similar to Step 1 in Example 57. [00630] Step-2
Figure imgf000236_0002
4
[00631] Saponification of 3 (4.58 g, 11.3 mmol) by LiOH (500 mg, 20 mmol) in 80 mL THF/H2O (1 : 1) and usual workup with 1 N HCl gave free acid 4. [00632] Step-3
Figure imgf000236_0003
[00633] Acid 4 was directly mixed with 4-methoxybenzylamine (1.55 g, 11.3 mmol), TBTU (5.4 g, 16.8 mmol) and DIEA (2.4 mL, 13.4 mmol) in 100 mL MeCN at room temperature. The reaction mixture was run overnight to give 6 as a white solid (4.2 g, 8.5 mmol) after flash chromatography (EtOAc-hexane). [00634] Step-4
Figure imgf000236_0004
[00635] Step 4 was run in a manner similar to Step 4 in Example 57 with CHCI3 being substituted for CH2Cl2 as the solvent. [00636] Step-5
Figure imgf000237_0001
[00637] The 2-methylsulfone of 7 (1.0 g, 1.9 mmol) was mixed with 3 -methoxy aniline (420 mg, 3.4 mmol) in DMA and the mixture was heated at 90 0C for 24 hours. Workup was done in a manner similar to that for Step-5 in Example 57 to give 9 (300 mg, 0.52 mmol). [00638] Steps-6, 7, and 8
Figure imgf000237_0002
1-178
[00639] The Boc group was removed from 9 by treatment with 4 N HCl in dioxane. The product (300 mg, 0.52 mmol) was treated immediately with acryloyl chloride (43 μL, 0.52 mmol) in 15 mL DCM at -40 0C. This intermediate was purified by flash chromatography (MeOH-DCM) and was reacted with triflic acid and TFA in DCM to provide the crude benzylamine 11 (120 mg, 0.228 mmol). This intermediate(120 mg, 0.228 mmol) was converted to 1-178 using triflic acid (305 μL, 3.44 mmol) in TFA/DCM (5 mL, 1 :1) at room temperature to provide ~ 35 mg final compound 1-178 as grey powder after purification via column chromatography (16% yield for three steps). MS: m/z = 405. EXAMPLE 62
[00640] Preparation of tert-butyl 3-(3-(4-(3-acrylamidophenylamino)-5-methylpyrimidin-2- ylamino)phenoxy)propylcarbamate 1-45
Figure imgf000238_0001
1-45
[00641] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000239_0001
A) 1, methanesulfonyl chloride, CH2Cl2, Et3N, rt, 1 h; B) 3, K2CO3, DMF, 60 0C; C) H2, Pd/C, EtOH, rt, 16 hr; D) 6, 7, Pd(OAc)2, BINAP, Cs2CO3, toluene, 100 0C, 16 hr; E) 5, AcOH, EtOH, 90 0C, 16 hr; F) H2, Pd/C, EtOH, rt, 16 hr; G) 11, NMP, O 0C, 15 min 642] Step-1
Figure imgf000239_0002
[00643] To a stirring solution of 1 (1.0 g, 5.7 mmol) in dichloromethane (20.0 rnL) was added Et3N (1.15 g, 11.41 mmol) and methanesulfonyl chloride (0.98 g, 8.56 mmol). The reaction mixture was stirred under nitrogen atmosphere at rt for 60 min. It was quenched with water (20 mL) and extracted with EtOAc (2 x 50 mL). The combined EtOAc extract was washed with 10% NaHCO3 soln. (25 mL), water (25 mL), brine (25 mL), dried over Na2SO4 and concentrated under reduced pressure to get 2 (1.36 g, 94%) as a colorless viscous liquid. It was used in the next step without further purification. [00644] Step-2
Figure imgf000240_0001
4
[00645] To a stirring solution of 2 (0.749 g, 5.39 mmol) and K2CO3 (0.99 g, 7.19 mmol) in dry DMF (20 mL) was added 3 (1.36 g, 5.39 mmol) and the reaction mixture was heated at 60 0C for 16 h under nitrogen atmosphere. It was cooled, concentrated under reduced pressure and the residue was taken in ethyl acetate (25 mL). The ethyl acetate soln. was washed with water (2x10 mL), brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure to get 4 (1.2 g, 75%) as a yellowish viscous liquid. It was used in the next step without further purification. [00646] Step-3
Figure imgf000240_0002
5
[00647] To a solution of 4 (1.20 g, 4.05 mmol) in ethanol (25 mL)) was added Pd/C (0.12 g, 10% w/w) and the reaction mixture was allowed to stir under H2 atmosphere (1.5 Kg hydrogen pressure) at rt for 16 h. The reaction mixture was filtered through a pad of Celite® and concentrated under reduced pressure to get 5 (0.95 g, 88%) as a brownish viscous oil. It was used in the next step without further purification. [00648]
Figure imgf000241_0001
[00649] To a solution of 6 (1.69 g, 12.26 mmol), in toluene (50.0 niL) was added 7 (2.0 g, 12.26 mmol), BINAP (0.3 g, 0.49 mmol), cesium carbonate (7.9 g, 24.5 mmol). The solution was degassed (by purging N2 for 15 min) and to it was added Pd(OAc)2 (0.054 g, 0.25 mmol). The reaction mixture was stirred at 100 0C for 16 h under nitrogen atmosphere. It was cooled, diluted with ethyl acetate (100 mL) and filtered through Celite®. The filtrate was washed with water (2 x 25 mL), brine (25 mL), dried over Na2SO4 and concentrated under reduced pressure. The residue obtained was further purified by column chromatography (SiO2, 60-120 mesh, Ethylacetete/hexane: 15/85). The solid obtained after evaporating the required fractions was washed with diethyl ether and dried under high vacuum to get 8 (1.2 g, 37%) as a light yellow solid. [00650] Step-5
Figure imgf000241_0002
[00651] To a solution of 8 (0.5 g, 1.89 mmol) and 5 (0.805 g, 3.0 mmol) in ethanol (10.0 mL) was added glacial acetic acid (0.056 g, 0.95 mmol), and the reaction mixture was stirred in a sealed tube for 16 h at 90 0C. The reaction mixture was cooled, concentrated under reduced pressure. The residue was quenched with 10% sodium bicarbonate so In. (10.0 mL) and extracted with ethyl acetate (3x15 mL). The combined ethyl acetate extract was washed with water (15 mL), brine (15 mL), dried over Na2SO4 and concentrated under reduced pressure to get a residue. The crude residue was further purified by column chromatography (SiO2, EtOAc/Hexane: 50/50) to get 9 (0.57 g, 61%) as a light yellow solid. [00652]
Figure imgf000242_0001
[00653] To a solution of 9 (0.56 g, 1.13 mmol) in ethanol (25 niL)) was added 10% Pd/C (0.068 g) and the reaction mixture was allowed to stir under H2 atmosphere (1.5 Kg hydrogen pressure) at rt for 16 h. The reaction mixture was filtered through a pad of celite® and concentrated under reduced pressure to get 10 (0.45 g, 85%) as a brownish solid. It was used in the next step without further purification. [00654] Step-7
Figure imgf000242_0002
1-45
[00655] To a stirred solution of 10 (0.25 g, 0.5382 mmol) in NMP (2.5 mL) at 0 0C was added acryloyl chloride (0.073 g, 0.807 mmol) and the reaction mixture was stirred at 0 0C for 15 min The reaction mixture was added drop wise to a cold, stirring solution of 10% NaHCO3. After complete addition the solution was stirred for another 30 min at 0 0C, and then filtered through a Buchner funnel to isolate the precipitated solid. The solid was washed with cold water and hexane. It was dissolved in methanol: dichloromethane (50:50, 10 mL) and concentrated under reduced pressure. The residue obtained was suspended in cold water (50 mL), Et3N was added to it and it was extracted with ethyl acetate (2 x 100 mL). The combined ethyl acetate extract was washed with water (50 mL), brine (50 mL), dried over Na2SO4 and concentrated under reduced pressure to get 1-45 (0.100 g, 35.8%) as an off-white solid. 1H NMR (DMSO-d6) δ ppm: 1.37 (s, 9H), 1.70-1.80 (m, 2H), 2.10 (s, 3H), 3.00-3.06 (m, 2H), 3.79 (t, J = 6.24 Hz, 2H), 5.74 (d, J = 11.92 Hz, IH), 6.24 (dd, J = 1.84 & 15.16 Hz, IH), 6.35-6.47 (m, 2H), 6.80-6.90 (bs, IH), 6.97 (t, J = 8.28 Hz, IH), 7.23-7.27 (m, 2H), 7.31 (s, IH), 7.37 (d, J = 8.2 Hz, IH), 7.46 (d, J = 7.48 Hz, IH), 7.90-7.90-7.91 (m, 2H), 8.36 (s, IH), 8.87 (s, IH), 10.07 (s, IH); LCMS: m/e 519 (M+l).
EXAMPLE 63
[00656] Preparation of tert-butyl 3-(3-(4-(3-acrylamidophenylamino)-5-fluoropyrimidin-2- ylamino)phenoxy)propylcarbamate I- 183
Figure imgf000243_0001
1-183
[00657] The title compound was prepared according to the schemes, steps and intermediates described below.
step-3'
Figure imgf000243_0002
A) methanesulfonyl chloride, CH2Cl2, Et3N, rt, 1 h; B) K2CO3, DMF, 60 0C, 16 h; C) Pd-C, H2, ethanol, rt, 16 h. [00658] Step 1'
Figure imgf000243_0003
[00659] To a stired solution of 2' (4.0 g, 22.8 mmol) in dichloromethane (80.0 mL) was added Et3N (4.6 g, 45.5 mmol) and methanesulfonyl chloride (3.92 g, 34.2 mmol), and the reaction mixture was stirred under nitrogen atmosphere at RT for 60 min. The reaction was quenched with water (50 mL) and extracted with EtOAc (2x100 mL). The combined extracts were washed with 10% NaHCO3 solution (50 mL), water (50 mL), and brine (50 mL), dried over Na2SO4, and concentrated under reduced pressure to give 2 (5.5 g, 95.2%) as a light yellow viscous liquid. Compound 2 was used in the next step without further purification. [00660] Step V
Figure imgf000244_0001
[00661] To a stirred solution of 1 (2.3 g, 16.5 mmol) and K2CO3 (4.6 g, 33.3 mmol) in dry DMF (100 mL) was added 2 (5.5 g, 21.7 mmol), and the reaction mixture was heated at 60 0C for 16 h under nitrogen atmosphere. The reaction was cooled, quenched with water (250ml), and extracted with EtOAc (2x100 mL). The combined extracts were washed with 10% NaHCO3 solution(100 mL), water (3x100 mL), and brine (100 mL), dried over Na2SO4, and concentrated under reduced pressure to give 3 (4.0 g, 81.6%) as a light yellow viscous liquid. Compound 3 was used in the next step without further purification. [00662] Step 3'
Figure imgf000244_0002
[00663] To a solution of 3 (4.0 g, 13.4 mmol) in ethanol (50 mL) was added Pd/C (0.8 g, 10% w/w), and the reaction mixture was allowed to stir under H2 atmosphere (1.5 Kg hydrogen pressure) at rt for 16 h. The reaction mixture was filtered through a pad of Celite® and concentrated under reduced pressure to give 4 (3.3 g, 91.9%) as a brownish viscous oil. Compound 4 was used in the next step without further purification.
Figure imgf000245_0001
1-183
[00664]
Figure imgf000245_0002
[00665] A pressure tube was charged with 2 (10.0 g, 0.072 mol), 1 (24.1 g, 0.145 mol), n- BuOH (100 niL) and DIPEA (13.9 g, 0.108 mol), and the contents were stirred at 120 0C for 2 h. The reaction mixture was cooled, and the precipitated solid was isolated by filtration through a Buchner funnel, washed with cold hexane and dried to give 3 (12.5 g, 64%) as a yellow solid. Compound 3 was used in the next step without further purifications. [00666] Step 2
Figure imgf000245_0003
[00667] To a solution of 3 (1.5 g, 5.58 mmol) and 4 (1.48 g, 5.58 mmol) in ethanol (30.0 mL) was added glacial acetic acid (0.167 g, 2.79 mmol), and the reaction mixture was stirred in a pressure tube at 90 0C for 48 h. The reaction mixture was cooled and concentrated under reduced pressure; the residue was quenched with 10% sodium bicarbonate solution (20.0 rnL) and extracted with ethyl acetate (2x50 mL). The combined extracts were washed with water (25 mL) and brine (25 mL), dried over Na2SO4, and concentrated under reduced pressure to give crude 5. The crude residue was purified by column chromatography (neutral AI2O3, MeOH/Chloroform: 0.5/99.5) to give 5 (1.4 g, 50.3%) as a brown solid. [00668] Step 3
Figure imgf000246_0001
[00669] To a solution of 5 (1.4 g, 2.8 mmol) in ethanol (50 mL)) was added 10% Pd/C (0.28 g, 10% w/w) and the reaction mixture was allowed to stir under H2 atmosphere (1.5 Kg hydrogen pressure) at rt for 16 h. The reaction mixture was filtered through a pad of Celite® and concentrated under reduced pressure to give a residue. The crude residue was further purified by column chromatography (neutral Al2O3, MeOH/Chloroform: 0.5/99.5) to give a solid which was washed with dichloromethane/hexane mixtures to give 6 (0.7 g, 53.4%) as a pale brown solid. [00670] Step 4
Figure imgf000246_0002
1-183
[00671] tert-Butyl 3-(3-(4-(3-acrylamidophenylamino)-5-fluoropyrimidin-2- ylamino)phenoxy)propylcarbamate. To a stirred solution of 6 (0.25 g, 0.533 mmol) and potassium carbonate (0.138 g, 1.02 mmol) in NMP (2.5 mL) at 0 0C was added acryloyl chloride (0.060 g, 0.665 mmol), and the reaction mixture was stirred at 0 0C for 30 min. The reaction mixture was added dropwise to a cold, stirring solution of 10% NaHCO3 and stirred at the same temperature (0 0C) for 30 min. A white solid precipitated out and was isolated by filtration through a Buchner funnel. The solid was washed with cold water and hexane and dissolved in mixture of methanol/dichloromethane (50:50, 10 mL) and concentrated under reduced pressure. The residue obtained was suspended in cold water (25 mL), Et3N was added, and it was extracted with ethyl acetate (2x50 mL). The combined extracts were washed with water (50 mL), brine (50 mL), dried over Na2SO4 and concentrated under reduced pressure to give 1-183 (0.255 g, 91.4%) as light yellow solid. 1H NMR (DMSO-d6) δ ppm: 1.36 (s, 9H), 1.78 (quin, J = 6.4 Hz, 2H), 3.01-3.06 (m, 2H), 3.83 (t, J = 6.12 Hz, 2H), 5.74 (dd, J = 1.4 & 10.04 Hz, IH), 6.24 (d, J = 16.84 Hz, IH), 6.41-6.48 (m, 2H), 6.88 (s, IH), 7.03 (t, J = 8.24 Hz, IH), 7.23-7.31 (m, 3H), 7.41 (d, J= 8.28 Hz, IH), 7.56 (d, J= 7.96 Hz, IH), 7.90 (s, IH), 8.11 (d, J= 3.56 Hz, IH), 9.11 (s, IH), 9.43 (s, IH), 10.10 (s, IH); LCMS : m/e 523.1 (M+l).
EXAMPLE 64
[00672] Preparation of tert-butyl 3-(4-(4-(3-acrylamidophenylamino)-5-fluoropyrimidin-2- ylamino)phenoxy)propylcarbamate 1-198
Figure imgf000247_0001
1-198
[00673] The title compound was prepared according to the schemes, steps and intermediates described in Example 63 by using tert-butyl 3-(4aminophenoxy)propylcarbamate in place of 4 in Step-2. 1H NMR (DMSO-d6) δ ppm: 1.37 (s, 9H), 1.78 (quin, J= 6.36 Hz, 2H), 3.05 (q, J= 6.24 Hz, 2H), 3.86 (t, J= 6.2 Hz, 2H), 5.75 (dd, J= 1.92 & 10.04 Hz, IH), 6.24 (dd, J= 1.92 & 16.92 Hz, IH), 6.45 (dd, J = 10.08 & 16.92 Hz, IH), 6.72 (d, J = 9 Hz, 2H), 6.89 (t, J = 5.4 Hz, IH), 7.26 (t, J = 8.08 Hz, IH), 7.40 (d, J= 8.12 Hz, IH), 7.48-7.52 (m, 3H), 7.92 (s, IH), 8.05 (d, J = 3.72 Hz,lH), 8.95 (s, IH), 9.36 (s, IH), 10.12 (s, IH); LCMS : m/e 523.2 (M+l). [00674] The intermediate tert-butyl 3-(4aminophenoxy)propylcarbamate was prepared by the scheme shown below.
Figure imgf000248_0001
A) NaH, THF, rt, 16 h; B) H2, Pd/C, EtOH, rt, 16 hr [00675] Step-1
[00676] To a stirring solution of 1 (1.7 g, 9.7 mmol) in dry THF (40 rnL) was added NaH (0.72 g, 18.0 mmol, 60% dispersion in paraffin oil) at 0 0C and the reaction mixture was stirred at rt for 15 min under nitrogen atmosphere. To it was added 2 (2.0 g, 13.87 mmol) and the reaction mixture was stirred at rt for 16 h. It was quenched with cold water (20 mL), and extracted with ethyl acetate (25 mL). The ethyl acetate extract was washed with water (2x10 mL), brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure to get an oily liquid which was triturated with hexane to get 3 (2.Og, 69.5%) as a yellow crystalline solid. [00677] Step-2
[00678] To a solution of 3 (2.0 g, 6.749 mmol) in ethanol (30 mL)) was added 10% Pd/C (0.4 g, 20% w/w) and the reaction mixture was allowed to stir under H2 atmosphere (1.5 Kg hydrogen pressure) at rt for 16 h. The reaction mixture was filtered through a pad of celite® and concentrated under reduced pressure to get 4 (1.6 g, 89.3%) as a pinkish viscous oil. It was used in the next step without further purification.
EXAMPLE 65
[00679] Preparation of 4-(3acrylamidophenylamino)-5-fluoro-2-(3,4- dimethoxyphenylamino)-pyrimidine 1-134
Figure imgf000249_0001
1-134
[00680] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 3,4-dimethoxyaniline in place of 4 in Step-2. 1H NMR (200 MHz, CD3OD) δ 8.50 (s, IH), 7.80 (d, J= 6.5 Hz, IH), 7.70-7.66 (m, 2H), 7.20 (m, IH), 7.0 (m, 2H), 6.41 (m, 2H), 5.92 (dd, J= 8.0, 2.0 Hz, IH), 3.89 (s, 6H).
EXAMPLE 66
[00681] Preparation of 4-(3-acrylamidophenylamino)-5-fluoro-2-(3,4,5- trimethoxyphenylamino)-pyrimidine 1-133
Figure imgf000249_0002
1-133
[00682] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 3,4,5-trimethoxyaniline in place of 4 in Step-2. 1H NMR (200 MHz, CD3OD) δ 8.10 (s, IH), 8.0 (d, J = 6.0 Hz, IH), 7.50 (m, 2H), 7.30 (m, IH), 7.0 (m, 2H), 6.45 (m, 2H), 5.90 (dd, J= 8.0, 2.0 Hz, IH), 3.90 (s, 3H), 3.89 (s, 9H). EXAMPLE 67
[00683] Preparation of 4-(3-acrylamidophenylamino)-5-fluoro-2-(3-
(hydroxymethyl)phenylamino)-pyrimidine 1-145
Figure imgf000250_0001
1-145
[00684] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 3-hydroxymethylaniline in place of 4 in Step-2. 1H NMR (DMSO-de) δ ppm: 4.38 (d, J = 5.6 Hz, 2H), 5.07 (t, J = 5.68 Hz, IH), 5.75 (d, J = 10.84 Hz, IH), 6.24 (dd, J = 16.96 Hz, IH), 6.44 (dt, J = 10.04 & 17.0 Hz, IH), 6.83 (d, J = 7.4 Hz, IH), 7.10 (t, J= 7.72 Hz, IH), 7.28 (t, J= 8.16 Hz, IH), 7.40 (d, J= 8.08 Hz, IH), 7.55-7.59 (m, 3H), 7.92 (s, IH), 8.09 (d, J= 3.6 Hz, IH), 9.11 (s, IH), 9.40 (s, IH), 10.1 (s, IH); LCMS : m/e 378.0 (M+l).
EXAMPLE 68
[00685] Preparation of 4-(3-acrylamidophenylamino)-5-fluoro-2-(3-(3-(2-oxopyrrolidin-l- yl)propoxy)phenylamino)-pyrimidine 1-144
Figure imgf000250_0002
1-144
[00686] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 3-(2-oxopyrrolidin-l-yl)propoxyaniline in place of 4 in Step- 2. 1H NMR (DMSO-d6) δ ppm: 1.8-1.94 (m, 4H), 2.18 (q, J = 8.08 Hz, 2H), 3.26-3.40 (m, 4H), 3.80 (t, J= 6 Hz, 2H), 5.74 (d, J= 10.72 Hz, IH), 6.24 (d, J= 15.64 Hz, IH), 6.41-6.80 (m, 2H), 7.04 (t, J= 8.16 Hz, IH), 7.22-7.29 (m, 2H), 7.33 (s, IH), 7.42 (d, J= 8.08 Hz, IH), 7.55 (d, J = 7.52 Hz, IH), 7.91 (s, IH), 8.11 (d, J = 3.48 Hz, IH), 9.13 (s, IH), 9.43 (s, IH), 10.11 (s, IH); LCMS : m/e 49l (M+ 1).
EXAMPLE 69
[00687] Preparation of 4-(3-acrylamidophenylamino)-5-fluoro-2-(3-(3-
(methy lsulfony l)propoxy)pheny lamino)-pyrimidine 1-138
Figure imgf000251_0001
1-138
[00688] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 3-(3-(methylsulfonyl)propoxyaniline in place of 4 in Step-2. 1H NMR (DMSO-de) δ ppm: 2.05-2.15 (m, 2H), 3.0 (s, 3H), 3.22 (t, J = 7.76 Hz, 2H), 3.93 (t, J = 6.08 Hz, 2H), 5.74 (dd, J = 1.88 & 10 Hz, IH), 6.25 (dd, J= 1.8 & 16.88 Hz, IH), 6.44 (dd, J = 10.16 & 16.84 Hz, 2H), 7.05 (t, J= 8.16 Hz, IH), 7.24-7.30 (m, 2H), 7.35 (s, IH), 7.42 (d, J = 8.2 Hz, IH), 7.55 (d, J= 8 Hz, IH), 7.90 (s, IH), 8.11 (d, J = 3.6 Hz, IH), 9.14 (s, IH), 9.43 (s, IH), 10.10 (s, IH); LCMS : m/e 484 (M+l).
[00689] The intermediate 3-(3-(methylsulfonyl)propoxyaniline was prepared by the scheme shown below.
Figure imgf000251_0002
A) DEAD, Ph3P, Et3N, THF, rt, 1 hr; B) MCPBA, CH2Cl2, rt, 30 min; C) TFA, CH2Cl2, rt, 1 hr [00690]
Figure imgf000252_0001
[00691] To a stirred solution of 2 (1.1 g, 10.3 mmol) in THF (20 niL) were added 1 (2.18 g, 10.3 mmol), PPh3 (2.98 g, 11.3 mmol) and Et3N (1.68 g, 15 mmol) under N2 atmosphere. The reaction mixture was cooled to 0 0C and to it was added DEAD (1.98 g, 11.3 mmol). The reaction mixture was allowed to come to rt and stirred for 1 h. It was quenched with water, extracted with ethyl aceate (3x25 mL) and the combined EtOAc extract was washed with water and brine solution (5 mL each). The residue obtained after concentration under reduced pressure was purified by column chromatography (SiO2, 60-120, pet ether/ethyl acetate, 8/2) to get 3 (2 g 60.6%) as a white solid. [00692] Step-2
Figure imgf000252_0002
[00693] To a stirred solution of 3 (2 g, 6.7 mmol) in CH2Cl2 (25 mL) was added m-CPBA (4.13 g, 26.7 mmol) at -10 0C. The reaction mixture was allowed to come to rt and stirred for 30 min. It was quenched with Na2CO3 solution (10 mL), extracted with CH2Cl2 (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was further purified by column chromatography (SiO2, 60-120, chloroform/methanol 9/1) to get 4 (1.05 g, 68.8%) as a yellow oil. [00694] Step-3
Figure imgf000252_0003
[00695] To a stirred solution of 4 (0.75 g, 2.2 mmol) in CH2Cl2 (7.5 mL) was added TFA (3 vol.) at 0 0C. The reaction mixture was allowed to come to rt and stirred further at it for 1 h. It was concentrated under reduced pressure, basified with NaHCO3 solution (5 mL) and extracted with CH2Cl2 (3x10 mL). The combined organic extract was washed with water (2 mL) and brine solution (2 mL). Drying over Na2SO4 followed by filtration and concentration under reduced pressure offered 5 (500 mg, 96%) as brown solid. EXAMPLE 70
[00696] Preparation of N-(3-(5-fluoro-2-(3-(2-hydroxyethoxy)phenylamino)pyrimidin-4- ylamino)phenyl)acrylamide 1-105
Figure imgf000253_0001
1-105
[00697] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 3-(2-hydroxy)ethoxyaniline in place of 4 in Step-2. 1H NMR (DMSO-de) δ ppm: 3.67 (dd, J = 4.5 & 10 Hz, 2H), 3.85-3.87 (m, 2H), 4.83 (t, J = 5.6 Hz, IH), 5.75 (bd, J = IO Hz, IH), 6.25 (d, J = 15.6 Hz, IH), 6.42-6.46 (m, 2H), 7.05 (t, J = 8.4 Hz, IH), 7.26 (d, J = 8 Hz, IH), 7.30 (d, J = 8 Hz, IH), 7.33 (s, IH), 7.41 (d, J = 8 Hz, IH), 7.57 (d, J = 8 Hz, IH), 7.92 (s, IH), 8.11 (d, J = 3.6 Hz, IH), 9.11 (s, IH), 9.42 (s, IH), 10.11 (s, IH); LCMS : m/e 409.9 (M+l).
[00698] The intermediate 3-(2-hydroxy)ethoxyaniline was prepared by the scheme shown below.
Br-CH2COOEt 2
Figure imgf000253_0002
A) K2CO3, DMF, 70 0C, 12 h; B) Pd-C, H2, ethanol, rt, 10 h; C) IM LAH solution, THF, -15 0C, 45 min. [00699] Step-1
Figure imgf000253_0003
[00700] To a stirring solution of 1 (2.0 g, 14.37 mmol) and K2CO3 (3.95 g, 28.6 mmol) in dry DMF (15 rnL) was added 2 (2.88 g, 17.25 mmol) and the reaction was stirred at rt 70 0C for 12 h under nitrogen atmosphere. The reaction mixture was cooled, concentrated under reduced pressure and the residue was diluted with ethyl acetate (50 mL). It was washed with water (2x10 mL), brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure to get 3 (2.5g, 78%) as a light brown liquid. It was used in the next step without further purification. [00701] Step-2
Figure imgf000254_0001
[00702] To a solution of 3 (2.0 g, 8.88 mmol) in ethanol (20 mL)) was added Pd/C (0.2 g, 10% w/w) and the reaction mixture was allowed to stir under H2 atmosphere (1.0 Kg hydrogen pressure) at rt for 10 h. The reaction mixture was filtered through a pad of Celite® and concentrated under reduced pressure to get 4 (1.6 g, 94%) as a light brown liquid. It was used in the next step without further purification. [00703] Step-3
Figure imgf000254_0002
5
[00704] To a stirring solution of 4 (1.2 g, 6.14 mmol) in dry THF (12 mL)) was added lithium aluminum hydride (9.2 mL, 9.20 mmol, 1.0 M soln. in THF) at -15 0C, under N2 atmosphere. The reaction mixture was allowed to come to rt and stirred at it for 45 min. The reaction mixture was quenched with saturated ammonium chloride solution and was filtered through a pad of celite® and extracted with EtOAc (2 x 20 mL).The combined organic layer was washed with brine (10 mL) and concentrated under reduced pressure to get A (0.9 g, 95%) as a dark brown liquid. It was used in the next step without further purification. EXAMPLE 71
[00705] Preparation of N-(3-(5-fluoro-2-(3-(2-hydroxy-2- methylpropoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-118
Figure imgf000255_0001
1-118
[00706] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000255_0002
1-118
A) DIPEA, n-BuOH, 110 0C, 16 h; B) Pd(OAc)2, BINAP, Cs2CO3, toluene, 100 0C, 16 h; C) MeMgBr (3M solution in ether), THF, -78 0C, 3 h; D) TFA, CH2Cl2, rt, 3 h.
E) K2CO3, NMP, rt, 45 min. [00707]
Figure imgf000256_0001
[00708] Compound 3 was prepared according to the schemes, steps and intermediates described in Example 20. [00709] Step-2
Figure imgf000256_0002
[00710] A solution of 4 (0.7 g, 3.5 mmol), 3 (1.45g, 4.3 mmol), Pd(OAc)2 (0.03 g, 0.14 mmol), BINAP (0.13 g, 0..21 mmol) and Cs2CO3 (2.8 g, 8.7 mmol) in degassed toluene (30 mL) (toluene was purged with N2 for 30 min) was heated at 100 0C for 16 h under N2 atmosphere. The reaction mixture was cooled, diluted with EtOAc (15 mL) and washed with water (10 mL), brine (10 mL) and dried over Na2SO4. Filtration followed by concentration under reduced pressure offered a residue which was further purified by column chromatography (SiO2, 60-120, pet ether/ethyl acetate, 6/4) to get 5 (700 mg, 40%) as a white solid. [00711] Step-3
[00712] To a stirred solution of 5 (0.4 g, 0.8 mmol) in THF (10 mL) was added Methyl magnesium bromide ((3 M solution in ether, 1.6 mL, 4.8 mmol) at -78 0C. The reaction mixture was allowed to warm to -30 0C over 3 h, cooled again to -78 0C and quenched with saturated ammonium chloride solution (5 mL). The mixture was filtered through Celite® and filtrate was concentrated under reduced pressure to afford 6 as a pale yellow solid (300 mg, 78%) which was taken for next step without further purification. [00713] Step-4
Figure imgf000257_0001
[00714] To a stirred solution of 6 (0.2 g, 0.4 mmol) in CH2Cl2 (7.5 mL) was added TFA (3 vol.) at 0 0C. The reaction mixture was allowed to come to rt and stirred further at it for 3 h. It was concentrated under reduced pressure, basified with NaHCOs solution (5 mL) and extracted with CH2Cl2 (3x10 mL). The combined organic extract was washed with water (2 mL) and brine solution (2 mL). Drying over Na2SO4 followed by filtration and concentration under reduced pressure afforded a residue which was further purified by column chromatography (SiO2, 60- 120, pet ether/ethyl acetate, 6/4) to get 7 (130 mg, 86%) as a white solid. [00715] Step-5
Figure imgf000257_0002
1-118
[00716] To a stirred solution of 7 (0.08 g, 0.2 mmol) and potassium carbonate (0.11 g, 0.8 mmol) in NMP (1 mL) at 0 0C was added 8 (0.023 g, 0.22 mmol) and the reaction mixture was stirred at 0 0C for 45 min The reaction mixture was added drop wise to a cold, stirring solution of 10% NaHCO3 and stirred at the same temperature (0 0C) for 30 min. A solid precipitated out which was isolated by filtration through a Buchner funnel. The solid was washed with cold water, hexane and dissolved in a mixture of methanol/dichloromethane (50:50, 5 mL) and concentrated under reduced pressure. The residue obtained was suspended in cold water (10 mL), Et3N was added to it and it was extracted with ethyl acetate (2x5 mL). The combined ethyl acetate extract was washed with water (2 mL), brine (2 rnL), dried over Na2SO4 and concentrated under reduced pressure. The residue obtained was further purified by column chromatography (SiO2, 60-120, pet ether/ethyl acetate, 5/5) to get 1-118 (35 mg, 38%) as a white solid. 1H NMR (CD3OD) δ ppm: 1.27 (s, 6H), 3.67 (s, 2H), 5.76 (dd, J = 2.4 & 9.6 Hz, IH), 6.34 (dd, J = 2 & 16.8 Hz, IH), 6.42 (dd, J= 9.6 & 16.8 Hz, IH), 6.54 (td, J= 2 & 7.2 Hz, IH), 7.07-7.12 (m, 2H), 7.27-7.31 (m, 2H), 7.40 (d, J= 8 Hz, IH), 7.45 (d, J= 8 Hz, IH), 7.92 (d, J= 4 Hz, IH), 8.07 (d, J= 2 Hz, IH); LCMS : m/e 436.2 (M-I).
EXAMPLE 72
[00717] Preparation of N-(3-(5-fluoro-2-(3-(2-morpholino-2- oxoethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide I- 110
Figure imgf000258_0001
1-110
[00718] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 4-[(3-aminophenoxy)acetyl]-morpholine in place of 4 in Step- 2. 1H NMR (DMSO-de) δ ppm: 3.4-3.5 (bm, 4H), 3.5-3.6 (bm, 4H), 4.69 (s, 2H), 5.75 (dd, J= 2 & 10 Hz, IH), 6.25 (dd, J= 2 & 17.2 Hz, IH), 6.42-6.49 (m, 2H), 7.05 (t, J= 8 Hz, IH), 7.29 (t, J= 8 Hz, 3H), 7.41 (d, J= 8 Hz, IH), 7.57 (d, J= 8.8 Hz, IH), 7.91 (s, IH), 8.12 (d, J= 3.6 Hz, IH), 9.15 (s, IH), 9.45 (s, IH), 10.12 (s, IH); LCMS : m/e 491.0 (M-2).
[00719] The intermediate 4-[(3-aminophenoxy)acetyl]-morpholine was prepared by the scheme shown below.
Figure imgf000259_0001
Figure imgf000259_0002
A) LiOH, THF, MeOH, H2O, rt, 4 h; B) SOCl2, 85 0C, morpholine, O 0C, 30 min; C) Pd-C, H2, ethyl acetate, rt, 2 h. [00720] Step-1
Figure imgf000259_0003
[00721] To a stirred solution of 1 (1.0 g, 4.44 mmol) in methanol/THF/water : 5 mL/ 5 mL/5 rnL was added LiOH monohydrate (0.75 g, 17.76 mmol) and the reaction mixture was stirred at rt for 4 h. It was concentrated under reduced pressure, the residue was diluted with water (10 mL), acidified with 1.0 N HCl (PH ~5-6) and extracted with ether (2x20 mL). The combined ether extract was washed with water (20 mL), brine (20 mL), dried over Na2SO4 and concentrated under reduced pressure to get 2 (0.8 g, 91.43%) as an off-white solid. [00722] Step-2
Figure imgf000259_0004
[00723] Thionyl chloride (2.0 ml, 27.56 mmol) was added to 2 (0.2 g, 1.014 mmol) under nitrogen atmosphere. A drop of N5N Dimethylformamide was added to the mixture and the contents were stirred at 85 0C for 2 h. After cooling to rt thionyl chloride was removed by concentration under reduced pressure. The residue was cooled to 0 0C, morpholine (0.5 g, 5.74 mmol) was added to it in small portions and the reaction mixture was stirred at 0 0C for 30 min. The reaction mixture was allowed to come to rt and stirred at it for 30 min, cooled and quenched with water (10 mL). The contents were extracted with ether (2x10 mL) and the combined ether extract was washed with water (5 mL), brine (5 mL), dried over Na2SO4 and concentrated under reduced pressure to get 3 (0.180 g, 66.67%) as a yellow solid. [00724]
Figure imgf000260_0001
[00725] To a solution of 3 (0.180 g, 0.676 mmol) in ethyl acetate (10 niL)) was added Pd/C (0.036 g, 20% w/w) and the reaction mixture was allowed to stir under H2 atmosphere (1.0 Kg hydrogen pressure) at rt for 2 h. The reaction mixture was filtered through a pad of celite® and concentrated under reduced pressure to get A (0.14 g, 87.67%) as an off-white solid. It was used in the next step without further purifications.
EXAMPLE 73
[00726] Preparation of N-(3-(5-fluoro-2-(3-(l-hydroxy-2-methylpropan-2- yloxy)phenylamino)pyrimidin-4-ylamino)pheny l)acrylamide 1-91
Figure imgf000260_0002
1-91
[00727] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 3-(l-hydroxy-2-methylpropan-2-yloxy)aniline in place of 4 in Step-2. 1H NMR (DMSO-d6) δ ppm: 1.16 (s, 6H), 3.32-3.35 (m, 2H), 4.81 (t, J = 5.74 Hz, IH), 5.74 (dd, J = 1.84 & 10.04 Hz, IH), 6.24 (dd, J = 1.88 & 16.96 Hz, IH), 6.44 (dd, J = 10.12 & 16.96 Hz, IH), 6.50 (dd, J = 2.12 & 7.96 Hz, IH), 7.02 (t, J = 8.12 Hz, IH), 7.26-7.30 (m, 2H), 7.41 (d, J = 8.16 Hz, IH), 7.48 (d, J = 8.24 Hz, IH), 7.57 (d, J = 8.12 Hz, IH), 7.92 (s, IH), 8.09 (d, J = 3.6 Hz, IH), 9.07 (s, IH), 9.41 (s, IH), 10.09 (s, IH); LCMS : m/e 438.0 (M+l). [00728] The intermediate 3-(l-hydroxy-2-methylpropan-2-yloxy)aniline was prepared by the scheme shown below.
Figure imgf000261_0001
A) K2CO3, DMF, 16 h, rt; B) Pd/C, ethanol, 5 h, rt; C) LAH (IM in THF solution), O 0C to rt, 2 h. [00729] Step-1
Figure imgf000261_0002
[00730] To a solution of 1 (0.5 g 3.59 mmol) and 2 (0.84 g 4.316 mmol) in DMF was added K2CO3 (0.99 g, 7.194 mmol). After stirring at rt for 16 h, reaction mixture was concentrated under reduced pressure. The residue was diluted with ethyl acetate (10 mL) and washed with 10% NaOH solution (5 mL), water (5 mL) and brine solution (5 mL). Drying over Na2SO4, followed by concentration under reduced pressure gave 3 as red brown liquid (0.5 g, 52%). [00731] Step-2
Figure imgf000261_0003
[00732] To a stirred solution of 3 (0.45 g, 1.77 mmol) in ethanol (5 mL) was added Pd/C (45 mg) and the reaction mixture was hydrogenated (bladder pressure, ~1.5 Kg) for 5 h. The reaction mixture was passed through a celite® bed and concentrated under vacuum to get 4 (0.35 g, 88%) as a colorless liquid. [00733] Step-3
Figure imgf000261_0004
[00734] To a stirred solution of 4 (0.25 g, 1.15 mmol) in THF (5 niL) under N2 was added LAH (3.45 rnL, 3.35 mmol, IM solution in THF,) at 0 0C. The reaction mixture was allowed to come to rt and stirred at it for 2 h. It was carefully quenched with saturated Na2SO4 solution (2 mL), filtered and concentrated. The residue was further purified by column chromatography (SiO2, 60-120, pet ether/ethyl acetate, 6/4) to give 5 as a light brown liquid (0.15 g, 71%).
EXAMPLE 74
[00735] Preparation of N-(3-(5-fluoro-2-(3-(2-(2-oxopyrrolidin-l- yl)ethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide I- 164
Figure imgf000262_0001
1-164
[00736] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 3-(2-(2-oxopyrrolidin-l-yl)ethoxy)aniline in place of 4 in Step-2. 1H NMR (DMSO-d6) δ ppm: 1.89 (quin, J= 7.6 Hz, 2H), 2.21 (t, J= 8 Hz, 2H), 3.40 (t, J = 6.8 Hz, 2H), 3.50 (t, J = 5.6 Hz, 2H), 3.93 (t, J = 5.2 Hz, 2H), 5.75 (dd, J = 2 & 10 Hz, IH), 6.25 (dd, J = 2 & 16.84 Hz, IH), 6.42-6.49 (m, 2H), 7.05 (t, J = 8.4 Hz, IH), 7.28 (t, J = 8 Hz, 2H), 7.33 (s, IH), 7.43 (d, J= 8 Hz, IH), 7.57 (d, J= 8 Hz, IH), 7.92 (s, IH), 8.12 (d, J= 3.6 Hz, IH), 9.15 (s, IH), 9.45 (s, IH), 10.13 (s, IH); LCMS : m/e 475 (M-2).
EXAMPLE 75
[00737] Preparation of N-(3-(5-fluoro-2-(6-(3-(methylsulfonyl)propoxy)pyridin-3- ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-80
Figure imgf000262_0002
1-80 [00738] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 3-amino-6-(3-(methylsulfonyl)propoxy)pyridine in place of 4 in Step-2. 1H NMR (DMSO-d6) δ ppm: 2.05-2.20 (m, 2H), 3.00 (s, 3H), 3.24 (t, J = 7.46 Hz, 2H), 4.27 (t, J = 6.32 Hz, 2H), 5.75 (dd, J = 1.76 & 10 Hz, IH), 6.25 (dd, J = 1.8 & 16.96 Hz, IH), 6.45 (dd, J = 10.04 & 16.92 Hz, IH), 6.65 (d, J = 8.88 Hz, IH), 7.27 (t, J = 8.08 Hz, IH), 7.39 (d, J = 8.08 Hz, IH), 7.49 (d, J = 8 Hz, IH), 7.92 (s, IH), 7.99 (dd, J = 2.6 & 8.76 Hz, IH), 8.07 (d, J = 3.64 Hz, IH), 8.31 (d, J = 2.28 Hz, IH), 9.10 (s, IH), 10.11 (s, IH); LCMS : m/e 486.9 (M+l).
EXAMPLE 76
[00739] Preparation of N-(3-(2-(6-cyclobutoxypyridin-3-ylamino)-5-fluoropyrimidin-4- ylamino)phenyl)acrylamide 1-79
Figure imgf000263_0001
1-79
[00740] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 3-amino-6-cyclobutoxypyridine in place of 4 in Step-2. 1H NMR (DMSO-d6) δ ppm: 1.57-1.66 (m, IH), 1.71-1.78 (m, IH), 1.94-2.04 (m, 2H), 2.32-2.38 (m, 2H), 4.95-5.05 (m, IH), 5.73-5.76 (m, IH), 6.25 (dd, J = 1.92 & 16.92 Hz, IH), 6.45 (dd, J = 10.08 & 16.92 Hz, IH), 6.58 (d, J = 8.84 Hz, IH), 7.25 (t, J = 8.04 Hz, IH), 7.39 (d, J = 7.84 Hz, IH), 7.45-7.55 (m, IH), 7.90 (s, IH), 7.94 (dd, J = 2.72 & 8.88 Hz, IH), 8.06 (d, J = 3.68 Hz, IH), 8.27 (d, J = 2.6 Hz, IH), 9.04 (s, IH), 9.41 (s, IH), 10.1 (s, IH); LCMS : m/e 421.2 (M+l). EXAMPLE 77
[00741] Preparation of N-(3-(5-fluoro-2-(6-((l-methylpiperidin-4-yl)methoxy)pyridin-3- ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-78
Figure imgf000264_0001
1-78
[00742] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 3-amino-6-(l-methylpiperidin-4-yl)methoxypyridine in place of 4 in Step-2. 1H NMR (DMSO-d6) δ ppm: 1.23-1.27 (m, 3H), 1.65-1.69 (m, 2H), 1.83 (t, J = 11.72 Hz, 2H), 2.14 (s, 3H), 2.75 (d, J = 11.24 Hz, 2H), 4.0 (d, J = 6.2 Hz, 2H), 5.74 (dd, J = 2 & 10.04 Hz, IH), 6.24 (dd, J = 1.96 & 16.92 Hz, IH), 6.45 (dd, J = 10.08 & 16.92 Hz, IH), 6.62 (d, J = 8.88 Hz, IH), 7.27 (t, J = 8.08 Hz, IH), 7.40 (d, J = 8.88 Hz, IH), 7.47 (d, J = 7.6 Hz, IH), 7.92 (s, IH), 7.97 (dd, J = 2.76 & 8.92 Hz, IH), 8.07 (d, J = 3.72 Hz, IH), 8.28 (d, J = 2.64 Hz, IH), 9.07 (s, IH), 9.41 (s, IH), 10.11 (s, IH); LCMS : m/e 478.0 (M+l).
EXAMPLE 77
[00743] Preparation of N-(3-(5-fluoro-2-(4-chloro-3-methoxyphenylamino)pyrimidin-4- ylamino)phenyl)acrylamide 1-74
Figure imgf000264_0002
1-74
[00744] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 4-chloro-3-methoxyaniline in place of 4 in Step-2. 1H NMR (DMSO-de) δ ppm: 3.64 (s, 3H), 5.74 (dd, J = 2.12 & 9.96 Hz, IH), 6.24 (dd, J = 1.84 & 17 Hz, IH), 6.44 (dd, J = 10 & 16.84 Hz, IH), 7.13 (s, IH), 7.28 (t, J = 8.08 Hz, IH), 7.36 (dd, J = 2.12 & 8.68 Hz, IH), 7.40 (d, J = 7.64 Hz, IH), 7.45 (d, J = 2.04 Hz, IH), 7.50 (d, J = 8.12 Hz, IH), 7.91 (s, IH), 8.13 (d, J = 3.52 Hz, IH), 9.28 (s, IH), 9.48 (s, IH), 10.12 (s, IH); LCMS : m/e 414.0 (M+l).
EXAMPLE 78
[00745] Preparation of N-(3-(5-fluoro-2-(4-(2-hydroxy-2- methylpropoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-73
Figure imgf000265_0001
1-73
[00746] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 4-(2-hydroxy-2-methylpropoxy)aniline in place of 4 in Step-2. 1H NMR (MeOD) δ ppm: 1.33 (s, 6H), 3.75 (s, 2H), 5.80 (dd, J = 3.28 & 10.64 Hz, IH), 6.39 (dd, J = 2.24 & 16.96 Hz, IH), 6.47 (dd, J = 9.6 & 16.96 Hz, IH), 6.84 (td, J = 3.48 & 9.0 Hz, 2H), 7.30 (t, J = 7.72 Hz, IH), 7.41-7.50 (m, 4H), 7.89 (d, J = 3.88 Hz, IH), 8.09 (s, IH); LCMS : m/e 438 (M+l).
EXAMPLE 79
[00747] Preparation of N-(3-(5-fluoro-2-(6-(l,l-dioxidothiomorpholin-4-yl) pyridin-3- ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-72
Figure imgf000265_0002
1-72 [00748] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 3-amino-6-(l,l-dioxidothiomorpholin-4-yl) pyridine in place of 4 in Step-2. 1H NMR (DMS0-d6) δ ppm: 3.00-3.15 (bm, 4H), 3.90-4.10 (bm, 4H), 5.76 (dd, J = 1.64 & 10.04 Hz, IH), 6.26 (dd, J = 1.72 & 16.92 Hz, IH), 6.46 (dd, J = 10.04 & 16.88 Hz, IH), 6.87 (d, J = 9.04 Hz, IH), 7.20 (t, J = 8.04 Hz, IH), 7.39 (d, J = 8.24 Hz, IH), 7.50 (d, J = 7.68 Hz, IH), 7.90-7.93 (m, 2H), 8.06 (d, J = 3.6 Hz, IH), 8.35 (d, J = 2.4 Hz, IH), 9.0 (s, IH), 9.40 (s, IH), 10.12 (s, IH); LCMS : m/e 484 (M+l).
EXAMPLE 80
[00749] Preparation of N-(3-(5-fluoro-2-(6-(2-(2-oxopyrrolidin-l-yl)ethoxy)pyridin-3- ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-70
Figure imgf000266_0001
1-70
[00750] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 3-amino-6-(2-(2-oxopyrrolidin-l-yl)ethoxy)pyridine in place of 4 in Step-2. 1H NMR (DMSO-d6) δ ppm: 1.90 (quintet, J = 7.6 Hz, 2H), 2.19 (t, J = 8.04 Hz, 2H), 3.41 (t, J = 6.88 Hz, 2H), 3.50 (t, J = 5.36 Hz, 2H), 4.27 (t, J = 5.48 Hz, 2H), 5.75 (d, J = 10.92 Hz, IH), 6.25 (d, J = 17.04 Hz, IH), 6.45 (dd, J = 10.12 & 16.84 Hz, IH), 6.63 (d, J = 8.96 Hz, IH), 7.27 (t, J = 8.04 Hz, IH), 7.39 (d, J = 7.56 Hz, IH), 7.47 (d, J = 7.32 Hz, IH), 7.92 (s, IH), 7.98 (dd, J = 2.36 & 8.84 Hz, IH), 8.08 (d, J = 3.3 Hz, IH), 8.31 (d, J = 2.24 Hz, IH), 9.10 (s, IH), 9.44 (s, IH), 10.11 (s, IH); LCMS : m/e 478.0 (M+l).
EXAMPLE 81
[00751] Preparation of (R)-N-(3-(5-fluoro-2-(6-(tetrahydrofuran-3-yloxy)pyridin-3- ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-69
Figure imgf000267_0001
1-69
[00752] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using (R)-3-amino-6-(tetrahydrofuran-3-yloxy)pyridine in place of 4 in Step-2. 1H NMR (DMSO-d6) δ ppm: 1.91-1.99 (m, IH), 2.14-2.23 (m, IH), 3.70-3.77 (m, 2H), 3.81 (dd, J = 7.90 & 15.48 Hz, IH), 3.88 (dd, J = 4.76 & 10.16 Hz, IH), 5.38 (t, J = 4.68 Hz, IH), 5.75 (dd, J = 1.72 & 10.08 Hz, IH), 6.24 (d, J = 16.92 Hz, IH), 6.45 (dd, J = 10.16 & 16.88 Hz, IH), 6.63 (d, J = 8.84 Hz, IH), 7.26 (d, J = 7.64 Hz, IH), 7.39 (d, J-= 7.92 Hz, IH), 7.46 (d, J = 7.64 Hz, IH), 7.92 (s, IH), 7.97 (dd, J = 2.6 & 8.83 Hz, IH), 8.07 (d, J = 3.6 Hz, IH), 8.32 (d, J = 2.48 Hz, IH), 9.08 (s, IH), 9.42 (s, IH), 10.10 (s, IH); LCMS : m/e 437.2 (M+l).
EXAMPLE 82
[00753] Preparation of N-(3-(2-(4-chloro-3-(3-(methylsulfonyl)propoxy)phenylamino)-5- fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-55
Figure imgf000267_0002
1-55
[00754] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 4-chloro-3-(3-(methylsulfonyl)propoxy)aniline in place of 4 in Step-2. 1H NMR (DMSO-d6) δ ppm: 2.07-2.14 (m, 2H), 3.0 (s, 3H), 3.22 (t, J = 7.72 Hz, 2H), 3.90 (t, J = 6.08 Hz, 2H), 5.75 (dd, J = 1.88 & 10.08 Hz, IH), 6.24 (dd, J = 1.84 & 16.92 Hz, IH), 6.44 (dd, J = 10.12 & 16.96 Hz, IH), 7.15 (d, J = 8.72 Hz, IH), 7.30 (t, J = 8.08 Hz, IH), 7.35 (dd, J = 22 SL 8.8 Hz, IH), 7.43 (d, J = 8 Hz, IH), 7.45-7.55 (m, 2H), 7.91 (s, IH), 8.14 (d, J = 3.56 Hz, IH), 9.31 (s, IH), 9.49 (s, IH), 10.14 (s, IH); LCMS : m/e 520.0 (M+ 1).
EXAMPLE 83
[00755] Preparation of N-(3-(2-(3-fluoro-4-(2-methoxyethoxy)phenylamino)-5- fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-96
Figure imgf000268_0001
1-96
[00756] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 3-fluoro-4-(2-methoxyethoxy)aniline in place of 4 in Step-2. 1H NMR (DMSO, 400 MHz) δ 10.13 (s, IH), 9.43 (s, IH), 9.18 (s, IH), 8.09 (d, IH, J = 3.68 Hz), 7.92 (s, IH), 7.65 (dd, IH, J = 2.3, 14.2 Hz), 7.47 (d, IH, J = 8.24 Hz), 7.41 (d, IH, J = 8.28 Hz), 7.27 (t, 2H, J = 8.0 Hz), 6.94 (t, IH, J = 9.4 Hz), 6.44 (dd, IH, J = 16.96, 10.1 Hz), 6.23 (dd, IH, J =1.84, 16.96 Hz), 5.73 (dd, IH, J=I.4, 10.1 Hz), 4.04 (m, 2H), 3.61 (m, 2H), 3.29 (s, 3H). MS m/z: 442.0 (M+H+).
EXAMPLE 84
[00757] Preparation of N-(3-(2-(4-tert-butoxycarbonyl-2,3-dihydrobenzo[l,4]oxazin-6- yl)amino-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-175
Figure imgf000269_0001
1-175
[00758] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 6-amino-4-tert-butoxycarbonyl-2,3-dihydrobenzo[l,4]oxaxine in place of 4 in Step-2. MS m/z: 507.1 (M+H+).
EXAMPLE 85
[00759] Preparation of N-(3-(2-(4-tert-butoxycarbonyl-2,3-dihydrobenzo[l,4]oxazin-6- yl)amino-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-174
Figure imgf000269_0002
1-174
[00760] The title compound was prepared by treating the product of Example 84 with 4N HCl in dioxane at rt for 1 hr followed by removal of solvents in vacuo. MS m/z: 407.1 (M+H +). EXAMPLE 86
[00761] Preparation of N-(3-(2-(4-trifluoroacetyl-2,3-dihydrobenzo[l,4]oxazin-6-yl)amino-5- fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-143
Figure imgf000270_0001
1-143
[00762] The title compound was prepared by treating the product of Example 85 with trifluoroacetic anhydride at rt for 1 hr followed by removal of solvents in vacuo. MS m/z: 503.1 (M+H +).
EXAMPLE 87
[00763] Preparation of N-(3-(2-(4-methylsulfonyl-2,3-dihydrobenzo[l,4]oxazin-6-yl)amino-5- fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-140
Figure imgf000270_0002
1-140
[00764] The title compound was prepared by treating the product of Example 85 with mesyl chloride Et3N in CH2Cl2 at 0 0C for 30 min, followed by washing with aqueous NaHCO3, drying over Na2SO4 and removal of solvents in vacuo. MS m/z: 485.1 (M+H +). EXAMPLE 88
[00765] Preparation of N-(3-(2-(4-methyl-2,3-dihydrobenzo[l,4]oxazin-6-yl)amino-5- fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-126
Figure imgf000271_0001
1-126
[00766] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 6-amino-4-methyl-2,3-dihydrobenzo[l,4]oxazine in place of 4 in Step-2. MS m/z: 421.1 (M+H+).
EXAMPLE 89
[00767] Preparation of N-(3-(2-(4-acetyl-2,3-dihydrobenzo[l,4]oxazin-6-yl)amino-5- fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-112
Figure imgf000271_0002
1-112
[00768] The title compound was prepared by treating the product of Example 85 with acetic anhydride and pyridine in CH2Cl2 at rt for 1 hr, followed by washing with IN HCl, then with aqueous NaHCO3, drying over Na2SO4 and removal of solvents in vacuo. MS m/z: 449.1 (M+H EXAMPLE 90
[00769] Preparation of N-(3-(2-(l-tert-butoxycarbonyl-lH-indazol-5-yl)amino)-5- fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-151
Figure imgf000272_0001
1-151
[00770] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 5-amino-N-(tert-butoxycarbonyl)-lH-indazole in place of 4 in Step-2. MS m/z: 490.2 (M+H+).
EXAMPLE 91
[00771] Preparation of N-(3-(2-(lH-indazol-5-yl)amino)-5-fluoropyrimidin-4- ylamino)phenyl)acrylamide 1-156
Figure imgf000272_0002
1-156
[00772] The title compound was prepared by treating the product of Example 90 with 4N HCl in dioxane at rt for 1 hr followed by removal of solvents in vacuo. MS m/z: 390.1 (M+H+). EXAMPLE 92
[00773] Preparation of N-(3 -(2-(I -methyl- lH-indazol-5 -yl)amino)-5-fluoropyrimidin-4- ylamino)phenyl)acrylamide 1-155
Figure imgf000273_0001
1-155
[00774] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 5 -amino- 1 -methyl- lH-indazole in place of 4 in Step-2. MS m/z: 404.2 (M+H+).
EXAMPLE 93
[00775] Preparation of N-(3-(5-fluoro-2-(3-sulfamoylphenylamino)pyrimidin-4- ylamino)phenyl)acrylamide 1-160
Figure imgf000273_0002
1-160
[00776] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000274_0001
1-160
A) DIPEA, n-butanol, 120 0C, 2 h, pressure tube; B) AcOH, ethanol, 90 0C, 16 h; C) Pd-C, H2, ethanol, rt, 3 h; D) acryloyl chloride, K2CO3, NMP, 0 0C, 60 min. [00777] Step-1
Figure imgf000274_0002
[00778] A pressure tube was charged with 2 (10.0 g, 0.072 mol), 1 (24.1 g, 0.145 mol), n- BuOH (100 niL) and DIPEA (13.9 g, 0.108 mol) and the contents were stirred at 120 0C for 2 h. The reaction mixture was cooled, the precipitated solid was isolated by filtration through a Buchner funnel, washed with cold hexane and dried to get 3 (12.5 g, 64%) as a yellow solid. It was used in the next step without further purification. [00779]
Figure imgf000275_0001
[00780] To a solution of 3 (0.25 g, 0.93 mmol) and 4 (0.16 g, 0.93 mmol) in ethanol (2.5 niL) was added glacial acetic acid (0.083 g, 1.39 mmol), and the reaction mixture was stirred in a pressure tube at 90 0C for 16 h. It was cooled, the precipitated solid was isolated by filtration through a Buchner funnel, washed with cold ether and dried to get 5 (0.245 g, 65%) as brown solid. It was used in the next step without further purification. [00781] Step-3
Figure imgf000275_0002
[00782] To a solution of 5 (0.1 g, 0.24 mmol in methanol (4 mL)) was added 10% Pd/C (0.2 g, 20% w/w) and the reaction mixture was allowed to stir under H2 atmosphere (1.5 Kg hydrogen pressure) at rt for 3 h. The reaction mixture was filtered through a pad of Celite® and concentrated under reduced pressure to get 6 (0.076 g, 82%) as a brown solid. It was used in the next step without further purification. [00783] Step-4
Figure imgf000275_0003
1-160
[00784] To a stirred solution of 6 (0.07 g, 0.18 mmol) and potassium carbonate (0.051 g, 0.37 mmol) in NMP (0.7 mL) at 0 0C was added acryloyl chloride (0.021 g, 0.23 mmol) and the reaction mixture was stirred at 0 0C for 60 min The reaction mixture was added drop wise to a cold, stirring solution of 10% NaHCO3 and kept at the same temperature (0 0C) for 30 min. A solid precipitated out which was isolated by filtration through a Buchner funnel. The solid was washed with cold water and hexane and dissolved in mixture of methanol/dichloromethane (50:50, 5 mL) and concentrated under reduced pressure. The residue obtained was suspended in cold water (10 mL), Et3N was added to it and it was extracted with ethyl acetate (2x10 mL). The combined ethyl acetate extract was washed with water (5 mL), brine (5 mL), dried over Na2SO4 and concentrated under reduced pressure get a residue. The crude residue was further purified by column chromatography (neutral Al2O3, MeOH/chloroform: 3/97) to get 1-160 (0.028 g, 35%) as light brown solid. 1H NMR (DMSO-d6) δ ppm: 5.75 (dd, J= 1.68 & 10.24 Hz, IH), 6.25 (dd, J = 1.8 & 17 Hz, IH), 6.43 (dd, J= 10 & 16.92 Hz, IH), 7.27-7.35 (m, 5H), 7.40 (d, J = 8 Hz, IH), 7.60 (d, J = 8.16 Hz, IH), 7.92 (s, IH), 7.95-8.05 (m, IH), 8.07 (s, IH), 8.14 (d, J = 3.52 Hz, IH), 9.50 (s, 2H), 10.12 (s, IH); LCMS : m/e 428.9 (M+ 1).
EXAMPLE 94
[00785] Preparation of N-(3-(5-cyano-2-(4-(2-methoxyethoxy)phenylamino)pyrimidin-4- ylamino)phenyl)acrylamide 1-109
Figure imgf000276_0001
1-109
[00786] The title compound was prepared according to the steps and intermediates as described below.
Figure imgf000277_0001
6 1-109
A) DMA, K2CO3, rt, 10 h, pressure tube; B) PTSA, dioxane, 100 0C, 2 h; C) Zn(CN)2, Ph3P, DMF, 120 0C, 12 h; D) 4N HCl, dioxane, rt, 1 hr; then acryloyl chloride, Et3N, DCM, -10 0C, 10 min. [00787] Step-1
Figure imgf000277_0002
[00788] To a solution of 5-bromo-2,4-dichloropyrimidine (0.45 g, 2.0 mmol) and tert-butyl 3- aminophenylcarbamate (0.44 g, 2.1 mmol) in DMA (3 mL) was added K2CO3 (0.55 g, 4.0 mmol). The suspention was stirred for 10 hours. Water (10 mL) was added and the precipitate was collected by filtration. The solid was washed with ether and dried to yield 0.8 g of compound 3. MS: m/e=399.1, 401.2 (M+ 1). [00789]
Figure imgf000278_0001
[00790] To a solution of compound 3 (400 mg, 1.0 mmol) and 4-(2-methoxyethoxy)aniline (0.2 g, 1.2 mmol) in 8 ml dioxane was added 4-methylbenzenesulfonic acid monohydrate (0.15g, 0.8 mmol). The mixture was stirred at 100 0C for two hours. The solvent was evaporated. The residue was dissolved in 30 ml ethyl acetate and washed with NaHCO3 aqueous solution, water and brine. The organic layer was separated and dried over Na2SO4. After removal of solvent, the crude product was subject to chromatography on silica gel (hexane:EtOAc = 1 :1). 0.40 g of the title compound 5 was obtained: MS m/z: 530.1, 532.1(M+H+). [00791] Step-3
Figure imgf000278_0002
[00792] To a suspension of Zn(CN)2 (0.24g, 2.0 mmol), Pd(PPh3)4 (60 mg, 0.05 mmol) in 3 ml DMF was added to 5 (0.25 g, 0.5 mmol). The mixture was degassed and sealed under argon, and heated at 120 0C for 12 hours. Water (10 ml) was added and the precipitate was collected by filtration. The solid was washed with ether and dried to yield 0.2 g of compound 6. MS: m/e=477.1 (M+l). [00793]
Figure imgf000279_0001
1-109
[00794] Compound 6 (0.10 g, 0.21 mmol) was dissolved in 4 N HCl (2 niL) in dioxane. The mixture was stirred at rt for 1 hour. After removal of solvents, a 5-mL portion of DCM was poured in followed by evaporation to dryness. This process of DCM addition followed by evaporation was repeated three times to give a residue solid which was used directly for the next step: MS m/z: 377.0 (M+H+).
[00795] To a solution of the intermediate obtained above, triethylamine (0.1 ml, 0.8 mmol) in 2 ml dichloromethane was added acryloyl chloride (19 mg, 0.21 mmol) at -10 0C. The reaction was stirred for 10 minutes at -10 0C and was quenched by NaHCO3 aqueous solution. Ethyl acetate (10 mL) was added and washed with NaHCO3 aqueous solution, water and brine. The organic layer was separated and dried over Na2SO4. After removal of solvent, the crude product was subject to chromatography on silica gel (hexane:EtOAc = 1 :2) to give 30 mg of the title compound. MS m/z: 431.1 (M+H+).
EXAMPLE 95
[00796] Preparation of N-(3-(5-cyano-2-(6-methoxypyridin-3-ylamino)pyrimidin-4- ylamino)phenyl)acrylamide 1-173
Figure imgf000280_0001
1-173
[00797] The title compound was prepared according to the schemes, steps and intermediates described in Example 94 by using 3-amino-6-methoxypyridine in place of 4 in Step-2. MS m/z:
388.2 (M+H +).
EXAMPLE 96
[00798] Preparation of N-(3-(5-cyclopropyl-2-(4-(2-methoxyethoxy)phenylamino)pyrimidin- 4-ylamino)phenyl)acrylamide 1-139
Figure imgf000280_0002
1-139
[00799] The title compound was prepared according to the steps and intermediates as described below.
Figure imgf000281_0001
A) Potassium cyclopropyltrifluoroborate, Pd(OAc)2, Xanphos, Cs2(CO3), toluene, 100 0C, 12 h;
B) PTSA, dioxane, 100 0C, 2 h; C) Zn(CN)2, Ph3P, DMF, 120 0C, 12 h; D) 4N HCl, dioxane, rt, 1 hr; then acryloyl chloride, Et3N, DCM, -10 0C, 10 min.
[00800] Step-1
Figure imgf000281_0002
[00801] Potassium cyclopropyltrifluoroborate (0.4g, 3.0 mmol), compound 1 (l.Og, 2.5 mmol), palladium acetate (34 mg, 0.15 mmol), Xanphos (0.17 g, 0.3 mmol) and Cs2CO3 (2.4g, 7.5 mmol) were suspended in 25 ml toluene and 5ml water. The mixture was degassed, sealed under argon and heated at 100 0C for 12 hours. 50 ml ethyl acetate was added and washed with NaHCO3 aqueous solution, water and brine. The organic layer was separated and dried over Na2SO4. After removal of solvent, the crude product was subject to chromatography on silica gel (hexane:EtOAc = 3:2). 0.54 g of the title compound 2 was obtained: MS m/z: 361.2 (M+H+). [00802]
Figure imgf000282_0001
[00803] Compound 4 was prepared from compound 2 and 3 following the procedure described in Step-2 of Example 94. MS m/z: 492.2 (M+H+). [00804] Step-3
Figure imgf000282_0002
1-139
[00805] The title compound 1-139 was prepared from compound 4 following the procedure described in Step-4 of Example 94. MS m/z: 446.1 (M+H+).
EXAMPLE 97
[00806] Preparation of N-(3-(5-cyclopropyl-2-(6-methoxypyridin-3-ylamino)pyrimidin-4- ylamino)phenyl)acrylamide 1-167
Figure imgf000282_0003
1-167 [00807] The title compound was prepared according to the schemes, steps and intermediates described in Example 96 by using 3-amino-6-methoxypyridine in place of 3 in Step-2. MS m/z: 403.2 (M+H +).
EXAMPLE 98
[00808] Preparation of N-(3-(5-fluoro-2-(3-(3-(2-oxopyrrolidin-l- yl)propoxy)phenylamino)pyrimidin-4-yloxy)phenyl)acrylamide I- 162
Figure imgf000283_0001
1-162
[00809] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000283_0002
1
Figure imgf000283_0003
A) DIPEA, n-BuOH, 110 0C, 16 h; B) Pd(OAc)2, BINAP, Cs2CO3, toluene, 100 0C, 16 h; C) TFA, CH2Cl2, rt. 2 h; D) K2CO3, NMP, rt, 45 min. [00810]
Figure imgf000284_0001
[00811] A pressure tube was charged with 2 (2.0 g, 9.61 mmol), 1 (3.21 g, 19.23 mmol), n- BuOH (30 rnL) and DIPEA (1.86 g, 14.42 mmol) and the contents were stirred at 110 0C for 16 h. The reaction mixture was cooled, concentrated under reduced pressure, quenched with water (30 mL) and extracted with ethyl acetate (2 x 30 mL). The combined ethyl acetate extract was washed with water (20 mL), brine (20 mL), dried over Na2SO4 and concentrated under reduced pressure to get a residue. It was triturated with hexane to get 3 (2.5 g, 96%) as a yellow solid. [00812] Step-2
Figure imgf000284_0002
[00813] To a solution of 3 (0.36 g, 1.1 mmol) in toluene (15 mL) was added 3-(3-(2- oxopyrrolidin-l-yl)propoxyaniline 4 (0.25 g, 1.1 mmol) followed by BINAP (0.031 g, 0.05 mmol), palladium acetate (0.0022 g, 0.01 mmol), and Cs2CO3 (0.82 g, 2.5 mmol). The reaction mixture was stirred and N2 was bubbled into it for 15 min. It was heated at 100 0C for 8 h under N2 atmosphere. The reaction mixture was cooled to room temperature, diluted with ethyl acetate (30 mL), washed with water (15 mL), brine (15 mL), and dried over Na2SO4. Concentration under reduced pressure offered a residue which was purified by column chromatography (SiO2, 60-120, product getting eluted in 3% methanol/chloroform: 3/97) to get 5 (0.3 g, 60%) as yellow solid. [00814]
Figure imgf000285_0001
[00815] To a stirred solution of 5 (0.25 g, 0.46 mmol) in CH2Cl2 (10 niL) was added TFA (1.0 rnL) at 0 0C under nitrogen atmosphere. The reaction mixture was allowed to come to rt and stirred at this temperature for 2 h. Crude reaction mixture was poured into ice cold water (10 mL), basified with sodium bicarbonate solution and extracted with ethyl acetate (3x15 mL). The combined ethyl acetate extract was washed with water (15 mL), brine (10 mL), dried over Na2SO4, and concentrated under reduced pressure to get 6 (0.130 g, 65%) as a yellow solid. It was used in the next step without further purifications [00816] Step-4
Figure imgf000285_0002
1-162
[00817] To a stirred solution of 6 (0.08 g, 0.18 mmol) and potassium carbonate (0.124 g, 0.9 mmol) in NMP (1.2 mL) at 0 0C was added acryloyl chloride (0.020 g, 0.22 mmol) and the reaction mixture was stirred at 0 0C for 45 min The reaction mixture was added drop wise to a cold, stirring solution of 10% NaHCO3 and stirred at the same temperature (0 0C) for 30 min. A solid precipitated out which was isolated by filtration through a Buchner funnel. The solid was washed with cold water, hexane and dissolved in a mixture of methanol/dichloromethane (50:50, 5 mL) and concentrated under reduced pressure. The residue obtained was suspended in cold water (10 mL), Et3N was added to it and it was extracted with ethyl acetate (2 x 10 mL). The combined ethyl acetate extract was washed with water (5 mL), brine (5 mL), dried over Na2SO4 and concentrated under reduced pressure to get 1-162 (0.050 mg, 56%). 1H NMR (DMSO-d6) δ ppm: 1.81-1.91 (m, 4H), 2.19 (t, J= 7.84 Hz, 2H), 3.26-3.35 (m, 4H), 3.73 (t, J = 6.04 Hz, 2H), 5.76 (dd, J= 1.92 & 10.04 Hz, IH), 6.25 (dd, J = 1.88 & 16.9 Hz, IH), 6.38-6.45 (m, 2H), 6.93 (t, J= 8.12 Hz, IH), 7.02-7.04 (m, 2H), 7.11 (s, IH), 7.43 (t, J= 8.16 Hz, IH), 7.55 (d, J= 8.24 Hz, IH), 7.68 (d, J= 1.8 Hz, IH), 8.56 (d, J= 2.88 Hz, IH), 9.56 (s, IH), 10.34 (s, IH); LCMS : m/e 490.0 (M-2).
[00818] The intermediate 3-(3-(2-oxopyrrolidin-l-yl)propoxyaniline 4 was prepared according to the scheme shown below.
Figure imgf000286_0001
A) NaH, DMF, rt, 16 h; B) SnCl2, Cone. HCl, 50 0C, 2 h. [00819] Step-1
Figure imgf000286_0002
[00820] To a stirred solution of NaH (1.0 g, 20.94 mmol) in DMF (10 mL) was added 1 (2.0 g, 13.96 mmol) at 0 0C. The reaction mixture was allowed to come to rt and stirred at it for 30 mins. To the reaction mixture was added 2 (1.96 g, 13.96 mmol), slowly and the reaction mixture was allowed to stir at rt for 16 h. The reaction mixture was concentrated under reduced pressure and the residue was diluted with ethyl acetate (20 mL). It was washed with water (2 x 5 mL), brine (5 mL) and dried over Na2SO4. Filtration followed by concentration under reduced pressure offered crude 3 (2 g, 55.5%) which was used in the next step without further purification. [00821] Step-2
Figure imgf000286_0003
[00822] To a stirred solution of 3 (2 g, 7.57 mmol) in cone. HCl (20 niL) was added SnCl2 (7.5 g, 34.06 mmol) in small portions. The reaction mixture was stirred at 50 0C for 2 h, cooled and basifϊed with NaHCOs. It was extracted with ethyl acetate (3 x 25 mL), washed with water (5 mL), brine solution (5 mL) and dried over anhydrous Na2SO4. Filtration followed by concentration under reduced pressure gave 4 (1.65 g, 93%) as dark brown solid which was used as such in the next step.
EXAMPLE 99
[00823] Preparation of N-(4-(5-fluoro-2-(3-(2-(2-oxopyrrolidin-l- yl)ethoxy)phenylamino)pyrimidin-4-yloxy)benzyl)-N-methylacrylamide 1-146
Figure imgf000287_0001
1-146
[00824] The title compound was prepared according to the schemes, steps and intermediates described in Example 98 by using (4-hydroxybenzyl)(methyl)carbamic acid tert-butyl ester in place of 2 in Step-1 and 3-(2-(2-oxopyrrolidin-l-yl)ethoxyaniline in place of 4 in Step-2. 1H NMR (CDCl3) δ ppm: 2.03 (quin, J = 7.4 Hz, 2H), 2.39 (t, J = 8 Hz, 2H), 3.07 & 3.06 (s, together 3H), 3.57 (t, J = 6.96 Hz, 2H), 3.66 (t, J = 5.08 Hz, 2H), 4.03-4.04 (bd, J = 4.96 Hz, 2H), 4.67 & 4.72 (s, together 2H), 5.70-5.85 (m, IH), 6.43 (d, J = 16.72 Hz, IH), 6.50 (d, J = 5.72 Hz, IH), 6.60-6.75 (m, IH), 6.89-6.96 (m, 2H), 7.06-7.08 (m, 2H), 7.18-7.30 (m, 2H), 7.37 (d, J= 8.44 Hz, IH), 8.21 (d, J= 2.36 Hz, IH); LCMS : m/e 506.2 (M+l). EXAMPLE 100
[00825] Preparation of N-(4-(5-fluoro-2-(3-(3-
(methylsulfonyl)propoxy)phenylamino)pyrimidin-4-yloxy)benzyl)-N-methylacrylamide 1-136
Figure imgf000288_0001
1-136
[00826] The title compound was prepared according to the schemes, steps and intermediates described in Example 98 by using (4-hydroxybenzyl)(methyl)carbamic acid tert-butyl ester in place of 2 in Step-1 and 3-(3-(3-methylsulfonyl)propoxyaniline in place of 4 in Step-2. 1H NMR (CDCl3) δ ppm: 2.25-2.40 (m, 2H), 2.97 (s, 3H), 3.07 (s, 3H), 3.20-3.30 (m, 2H), 3.98-4.05 (m, 2H), 4.67 (s, IH), 4.72 (s, IH), 5.7-5.82 (m, IH), 6.43 (dd, J= 1.96 & 16.96 Hz, IH), 6.49-6.53 (m, IH), 6.6-6.75 (m, IH), 6.85-7.00 (m, 2H), 7.05-7.15 (m, 2H), 7.18-7.25 (m, 2H), 7.36 (d, J = 8.36 Hz, IH), 8.21 (bd, J= 2.52 Hz, IH); LCMS : m/e 515.0 (M+l).
EXAMPLE 101
[00827] Preparation of N-(4-(5-fluoro-2-(6-methoxypyridin-3-ylamino)pyrimidin-4- yloxy)benzyl)-N-methylacrylamide 1-117
Figure imgf000288_0002
1-117
[00828] The title compound was prepared according to the schemes, steps and intermediates described in Example 98 by using (4-hydroxybenzyl)(methyl)carbamic acid tert-butyl ester in place of 2 in Step-1 and 6-methoxy-3-aminopyridine in place of 4 in Step-2. 1H NMR (DMSO- d6) δ ppm: 2.92 & 3.07 (s, together 3H), 3.76 (s, 3H), 4.62 & 4.74 (s, together 2H), 5.69 & 5.75 (dd, J= 1.6 & 10.4 Hz, together IH), 6.20 (dd, J= 1.2 & 16.4 Hz, IH), 6.56 (d, J= 8.8 Hz, IH), 6.82-6.90 (m, IH), 7.28-7.35 (m, 4H), 7.71 (bd, J = 7.6 Hz, IH), 8.14 (s, IH), 8.44 (bd, J = 2.8 Hz, IH), 9.48 (s, IH); LCMS : m/e 410 (M+ 1).
EXAMPLE 102
[00829] Preparation of N-(4-(5-fiuoro-2-(3-(3-(2-oxopyrrolidin-l- yl)propoxy)phenylamino)pyrimidin-4-yloxy)benzyl)-N-methylacrylamide 1-111
Figure imgf000289_0001
1-111
[00830] The title compound was prepared according to the schemes, steps and intermediates described in Example 98 by using (4-hydroxybenzyl)(methyl)carbamic acid tert-butyl ester in place of 2 in Step-1 and 3-(3-(2-oxopyrrolidin-l-yl)propoxy)aniline in place of 4 in Step-2. 1H NMR (DMSO-de) δ ppm: 1.80-2.6 (m, 4H), 2.20 (t, J = 7.6 Hz, 2H), 2.92 & 3.06 (s, together 3H), 3.20-3.40 (m, 4H), 3.75-3.90 (m, 2H), 4.62 & 4.73 (s, together 2H), 5.65-5.77 (m, IH), 6.20 (dd, J= 2.4 & 16.8 Hz, IH), 6.24 (bd, J= 8 Hz, IH), 6.86 (dd, J= 10.4 & 16.8 Hz, IH), 6.93 (t, J = 8 Hz, IH), 7.08 (t, J = 8 Hz, 2H), 7.28-7.36 (m, 4H), 8.48 (d, J = 2.8 Hz, IH), 9.51 (s, IH); LCMS : m/e 520.2 (M+l).
EXAMPLE 103
[00831] Preparation of N-(3-(5-fluoro-2-(3-(2-(2-oxopyrrolidin-l- yl)ethoxy)phenylamino)pyrimidin-4-yloxy)phenyl)acrylamide I- 184
Figure imgf000289_0002
1-184 [00832] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000290_0001
Figure imgf000290_0002
A) ) K2CO3, DMF, rt, 16 h; B) Pd(OAc)2, BINAP, Cs2CO3, toluene, 100 0C, 8 h; C) Pd-C, H2, methanol, rt, 16 h. D) ) acryloyl chloride, K2CO3, NMP, O 0C, 30 min. [00833] Step-1
Figure imgf000290_0003
[00834] To a stirring solution of 1 (24 g, 143.7 mmol) and K2CO3 (20 g, 143.6 mmol) in dry DMF (300 mL) was added 2 (1O g, 71.8 mmol) and the reaction mixture was stirred at rt for 16 h under nitrogen atmosphere. It was cooled and quenched with water (600 mL). A white solid precipitated out which was isolated by filtration through Buchner funnel and vacuum dried to get 3 (13 g, 68%) as a white solid. [00835]
Figure imgf000291_0001
5
[00836] To a solution of 3 (0.9 g, 3.3 mmol) in toluene (30 niL) was added 4 (950 mg, 4.3 mmol) followed by BINAP (0.12 g, 0.19 mmol), palladium acetate (0.02 g, 0.09 mmol), and CS2CO3 (2.7 g, 8.2 mmol). The reaction mixture was stirred and N2 was bubbled into it for 15 min. It was then heated at 100 0C for 8 h under N2 atmosphere. The reaction mixture was cooled to room temperature, diluted with ethyl acetate (60 mL), washed with water (35 mL), brine (35 mL), and dried over Na2SO4. Concentration under reduced pressure offered a residue which was purified by column chromatography (SiO2, 60-120, product getting eluted in methanol/chloroform: 8/92) to get 5 (0.50 g, 33%) as a white solid. [00837] Step-3
Figure imgf000291_0002
[00838] To a solution of 5 (0.5 g, 1.1 mmol) in methanol (50 mL)) was added 10% Pd/C (0.05 g, 10% w/w) and the reaction mixture was allowed to stir under H2 atmosphere (1.5 Kg hydrogen pressure) at rt for 16 h. The reaction mixture was filtered through a pad of celite and concentrated under reduced pressure to get 6 (0.3 g, 65%) as a colorless viscous liquid. [00839] Step-4
Figure imgf000291_0003
1-184 [00840] To a stirred solution of 6 (0.21 g, 0.5 mmol) and potassium carbonate (0.27 g, 2.0 mmol) in NMP (2.5 rnL) at 0 0C was added acryloyl chloride (0.053 g, 0.6 mmol) and the reaction mixture was stirred at 0 0C for 30 min The reaction mixture was added drop wise to a cold, stirring solution of 10% NaHCO3 and stirred at the same temperature (0 0C) for 30 min. A white solid precipitated out which was isolated by filtration through a Buchner funnel. The solid was washed with cold water and hexane and dissolved in mixture of methanol/dichloromethane (50:50, 10 mL) and concentrated under reduced pressure. The residue obtained was suspended in cold water (25 mL), Et3N was added to it and it was extracted with ethyl acetate (2 x 50 mL). The combined ethyl acetate extract was washed with water (50 mL), brine (50 mL), dried over Na2SO4 and concentrated under reduced pressure to get 1-184 (0.150 g, 65%) as white solid. 1H NMR (DMSO-d6) δ ppm: 1.89 (quin, J = 7.2 Hz, 2H), 2.21 (t, J = 7.6 Hz, 2H), 3.39 (t, J = 7.2 Hz, 2H), 3.49 (t, J= 5.2 Hz, 2H), 3.87 (t, J= 5.6 Hz, 2H), 5.77 (dd, J= 1.6 & 10.4 Hz, IH), 6.26 (dd, J= 1.6 & 17.2 Hz, IH), 6.39-6.46 (m, 2H), 6.95 (t, J= 8.4 Hz, IH), 7.03-7.12 (m, 3H), 7.44 (t, J = 8.4 Hz, IH), 7.56 (d, J = 8.4 Hz, IH), 7.70 (s, IH), 8.51 (d, J = 2.8 Hz, IH), 9.56 (s, IH), 10.35 (s, IH); LCMS : m/e 478 (M+ 1).
[00841] The intermediate 3-(2-(2-oxopyrrolidin-l-yl)ethoxyaniline 4 was prepared according to the scheme shown below.
Figure imgf000292_0001
A) NaH, THF, rt,16 h; B) Pd-C, H2, methanol, rt, 16 h. [00842] Step-1
Figure imgf000292_0002
[00843] To a stirring solution of NaH (3.4 g, 141.6 mmol, 60% dispersion in paraffin oil) in dry THF (50 mL) was added 1 (6 g, 46.0 mmol) at 0 0C and the reaction mixture was stirred at rt for 15 min under nitrogen atmosphere. To it was added a solution of 2 (5.0 g, 35.4 mmol) in THF (10 mL) and the reaction mixture was stirred at rt for 16 h. It was quenched with cold water (40 mL), and extracted with ethyl acetate (35 mL). The ethyl acetate extract was washed with water (2x25 niL), brine (25 mL), dried over Na2SO4 and concentration under reduced pressure to get a residue which was purified by column chromatography (SiO2, 60-120, product getting eluted in methanol/chloroform: 10/90) to get 3 (2.5 g, 30%) as a brownish liquid. [00844] Step-2
Figure imgf000293_0001
[00845] To a solution of 3 (2.2 g, 8.8 mmol) in methanol (50 mL)) was added 10% Pd/C (0. 22 g, 10% w/w) and the reaction mixture was allowed to stir under H2 atmosphere (1.5 Kg hydrogen pressure) at rt for 16 h. The reaction mixture was filtered through a pad of celite® and concentrated under reduced pressure to get 4 (1.7 g, 89%) as a yellowish liquid. It was used in the next step without further purification.
EXAMPLE 104
[00846] Preparation of N-(3 -(2-(6-methoxypyridin-3 -ylamino)-5 -methylpyrimidin-4- yloxy)phenyl)acrylamide 1-186
Figure imgf000293_0002
1-186
[00847] The title compound was prepared according to the schemes, steps and intermediates described in Example 103 by using 2,4-dichloro-5-methylpyrimidine in place of 1 in Step-1 and 6-methoxy-3-aminopyridine in place of 4 in Step-2. 1H NMR (DMSO-dβ) δ ppm: 2.16 (s, 3H), 3.73 (s, 3H), 5.76 (dd, J= 1.92 & 10.04 Hz, IH), 6.24 (dd, J= 1.92 & 16.92 Hz, IH), 6.39-6.49 (m, 2H), 6.92 (dd, J = 1.48 & 8 Hz, IH), 7.40 (t, J = 8.08 Hz, IH), 7.49 (d, J = 8.16 Hz, IH), 7.64 (d, J = 1.84 Hz, IH), 7.77-7.79 (m, IH), 8.17 (bs, IH), 8.20 (s, IH), 9.27 (s, IH), 10.29 (s, IH); LCMS : m/e 378 (M+l). EXAMPLE 105
[00848] Preparation of N-(3-(5-methyl-2-(phenylamino)pyrimidin-4-yloxy)phenyl)acrylamide
1-248
Figure imgf000294_0001
1-248
[00849] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000294_0002
5 1-248
A) K2CO3, DMF, rt, 24 h; A') (Boc)2O, THF, 60 0C, 2 h; B) aniline, cone. HCl, EtOH, 80 0C, 1 h; C) TFA, CH2Cl2, 0 0C to rt, 1Z2 h; D) acryloyl chloride, NMP, 0 0C, 10 min. [00850] Step-1
Figure imgf000295_0001
3
[00851] To a stirring solution of 2 (100 mg, 0.48 mmol) and K2CO3 (99.2 mg, 0.717 mmol) in dry DMF (5 mL) was added 1 (78 mg, 0.478 mmol) and the reaction was continued at rt for 24 h under nitrogen atmosphere. The reaction mixture was concentrated under reduced pressure and the residue was diluted with ethyl acetate (10 mL). It was washed with water (2 x 5 mL), brine (5 mL), dried over Na2SO4 and concentrated under reduced pressure to get 3 (120 mg, 75%) as a white solid. It was used for next step without further purification.
[00852] Step-2
Figure imgf000295_0002
[00853] A pressure tube was charged with 3 (75 mg, 0.224 mmol), cone. HCl (40 mg, 0.4 mmol), aniline (83 mg, 0.89 mmol) and ethanol (2.0 mL). The tube was screw capped and the contents were stirred at 80 0C for 60 min. The reaction mixture was cooled, concentrated under reduced pressure and the residue was quenched with water (5.0 mL). It was basified with 10% NaHCO3 so In. and extracted with Ethyl acetate (3x10 mL). The combined EtOAc layer was washed with water (2x5 mL), brine (5 mL), dried over Na2SO4 and concentrated under reduced pressure. The residue obtained was further purified by column chromatography (SiO2, 60-120 mesh, EtoAc/Hexane: 50/50) to get 4 (0.04 g, 45.9%) as an off-white sold. [00854]
Figure imgf000296_0001
[00855] To a stirring solution of 4 (160 mg, 0.40 mmol) in dichloromethane (4.0 rnL) was added at 0 0C, trifluoroacetic acid (0.8 rnL). Stirring was continued at the same temperature for 30 min after which the reaction mixture was concentrated under reduced pressure and the residue was dissolved in water (5.0 mL), basified with 10% NaHCO3 solution and extracted with dichloromethane (2 x 5 mL). The dichloromethane extract was washed with water (5 mL), brine (5 mL), dried over Na2SO4 and concentrated under reduced pressure to get 5 (110 mg, 93.2%) as an off white solid. It was used for next step without further purification. [00856] Step-4
Figure imgf000296_0002
[00857] To a stirred solution of 5 (75 mg, 0.256 mmol) in NMP (0.8 mL) at 0 0C was added acryloyl chloride (34.8 mg, 0.38 mmol) and the reaction mixture was stirred at 0 0C for 10 min. The reaction mixture was quenched with water (4.0 mL), basified with 10% NaHCO3 soln. and extracted with dichloromethane (2x5 mL). The dichloromethane extract was washed with water (5 mL), brine (5 mL), dried over Na2SO4 and concentrated under reduced pressure. The residue obtained was further purified column chromatography (SiO2, 60-120 mesh, CHCl3ZMeOH: 99/1) to get 1-248 (0.035 g, 39.6%) as a white colored solid. 1H NMR (DMSO-d6) δ ppm: 2.15 (s, 3H), 5.75 (dd, J = 1.92 & 10.04 Hz, IH), 6.24 (dd, J = 1.96 & 16.96 Hz, IH), 6.41 (dd, J = 10.6 & 17 Hz, IH), 6.78-6.8 (m, IH), 6.94 (dd, J = 1.44 & 8.04 Hz, IH), 7.00 (t, J = 7.52 Hz, 2H), 7.40- 7.44 (m, 3H), 7.53 (d, J = 8.24 Hz, IH), 7.6 (t, J = 2 Hz, IH), 8.23 (d, J = 1.04 Hz, IH), 9.36 (s, IH), 10.30 (s, IH); LCMS: m/e 346.8 (M+l).
EXAMPLE 106
[00858] Preparation of l-(4-(5-fluoro-2-(phenylamino)pyrimidin-4-ylamino)piperidin-l- yl)prop-2-en-l-one 1-229
Figure imgf000297_0001
1-229
[00859] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using l-tert-butyloxycarbonyl-4-aminopiperidine in place of 2 in Step-1 and aniline in place of 4 in Step-2. 1H NMR (DMSO-d6) δ ppm: 1.35-1.50 (m, 2H), 1.90- 2.05 (m, 2H), 2.7-2.85 (m, IH), 3.10-3.20 (m, IH), 4.11-4.15 (m, 2H), 4.46 (bd, J = 13.72 Hz, IH), 5.67 (dd, J = 2.44 & 10.4 Hz, IH), 6.10 (dd, J = 2.44 & 16.6 Hz, IH), 6.82-6.88 (m, 2H), 7.22 (t, J = 7.44 Hz, 2H), 7.35 (d, J= 7.56 Hz, IH), 7.70 (d, J = 7.72 Hz, 2H), 7.87 (d, i = 3.76 Hz, IH), 9.07 (s, IH); LCMS : m/e 341.383 (M+l).
EXAMPLE 107
[00860] Preparation of 2-((3-(5-fluoro-2-(4-(2-methoxyethoxy)phenylamino)pyrimidin-4- ylamino)phenyl)(hydroxy)methyl)acrylonitrile 1-71
Figure imgf000297_0002
1-71 [00861] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000298_0001
1-71
A) 2, DIPEA, n-BuOH, 120 0C, 12 h; B) cone. HCl, ethanol, 100 0C, 5 h; C) LiOH, MeOH/THF/H2O, rt, 6 h; D) Me-NH-OMe.HCl, EDCLHCl, HOBT, DIPEA, DMF, rt, 8 h; E) LAH (1.0 M soln. in THF), -78 0C, 30 min; F) DABCO, 1 ,4-dioxan/water, rt, 48 h. [00862]
Figure imgf000299_0001
[00863] A solution of 1 (0.50 g, 2.99 mmol), 2 (0.45 g, 2.99 mmol) and DIPEA (0.57 g, 4.48 mmol) in n-butanol (5.0 niL) was heated in a pressure tube (120 0C, 16 h). It was cooled, quenched with water (5 mL) and extracted with EtOAc (2x5 mL). The combined EtOAc extract was washed with water (2 mL), brine (2 mL), dried over Na2SO4 and concentrated under reduced pressure to afford 3 (0.70 g, 83.3%) as an off-white solid. [00864] Step-2
Figure imgf000299_0002
[00865] A solution of 3 (0.5 g, 1.77 mmol) and 4 (0.29 g, 1.77 mmol) in ethanol (2.5 mL) was taken in a pressure tube and acetic acid (0.1 mL) was added to it. The tube was tightly closed and the contents were stirred at 100 0C for 5 h. The reaction mixture was cooled, ethanol was removed under reduced pressure and the residue was taken in ethyl acetate (50 mL). It was washed with NaHCO3 solution (5 mL), brine (5 mL), dried over Na2SO4 and concentrated under reduced pressure. The solid precipitated was isolated by filtration. It was dried under vacuum to get 5 (0.6 g, 80%). [00866]
Figure imgf000300_0001
[00867] To a stirred solution of 5 (0.6 g, 1.4 mmol) in methanol/THF/water: 6 mL/6 mL/3 rnL was added LiOH (0.298 g, 7 mmol) and the reaction mixture was stirred at rt for 2 h. It was concentrated under reduced pressure; residue was diluted with water (2 mL) and extracted with diethyl ether (5 mL). The aqueous layer was separated and acidified with 1.5 N HCl (pH ~4-5), concentrated and dried under vacuum to get 6 (0.4 g, 70%) as a white solid which was taken for next step without further purification. [00868] Step-4
Figure imgf000300_0002
[00869] To a stirred solution of 6 (0.4 g, 1 mmol) in DMF (3 mL) were added MeNH- OMe.HCl (0.102 g, 0.1 mmol), EDCLHCl (0.003g, 1.5 mmol), HOBT (71 mg, 0.5 mmol) and DIPEA (0.204 g, 1.5mmol). The reaction mixture was stirred at room temperature for 8 h and quenched with water and extracted with EtOAc (2 x 5 mL). The combined organic layer was washed with brine, dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to get 7 (0.4 g, 90.9%) as a white solid. [00870]
Figure imgf000301_0001
8
[00871] To a stirred solution of 7 (0.4 g, 0.9 mmol) in THF (10 mL) was added LAH (1.8 niL, 1.8 mmol) at -78 0C. The reaction mixture was allowed to stir at the same temperature for 30 mins after which it was quenched with Na2SO4 solution (2 mL) and extracted with ethyl acetate (10 mL). The ethyl acetate layer was separated and washed with water (2 mL), brine solution (2 mL) and dried over anhydrous Na2SO4. Filtration followed by concentration under reduced pressure offered a residue which was purified by column chromatography (SiO2, 60-120, pet ether/ethyl acetate 7/3) to get 8 (200 mg, 58%) as a yellow solid. [00872] Step-6
Figure imgf000301_0002
1-71
[00873] To a stirred solution of 8 (200 mg, 0.523 mmol) and 9 (69 mg, 1.3 mmol) in 1,4- dioxane/H2O (1.4 mL/0.6 mL) was added DABCO (50 mg, 0.2523 mmol) at rt. Stirring was continued at room temperature for 48 h after which the reaction mixture was concentrated under reduced pressure. The residue obtained was further purified by column chromatography (SiO2, pet ether/ethyl acetate, 6/4) to get 1-71 as greenish gummy material (0.05 g, 22.70Zo)-1H NMR (DMSO-d6) δ ppm: 3.48 (s, 3H), 3.77 (t, J = 4.4 Hz, 2H), 4.11-4.16 (m, 2H), 5.11 (s, IH), 5.99 (s, IH), 6.06 (s, IH), 6.85 (s, IH), 6.91 (d, J = 8.84 Hz, 2H), 7.15 (d, J = 7.44 Hz, IH), 7.30- 7.40 (m, ; LCMS : m/e (M+l).
EXAMPLE 108
[00874] Preparation of 2-((4-(5-fluoro-2-(phenylamino)pyrimidin-4- ylamino)phenyl)(hydroxy)methyl)acrylonitrile 1-161
Figure imgf000302_0001
1-161
[00875] The title compound was prepared according to the schemes, steps and intermediates described in Example 107 by using aniline in place of 4 in Step-2. 1H NMR (CDCI3) δ ppm: 5.32 (s, IH), 6.07 (d, J= 0.8 Hz, IH), 6.15 (d, J= 1.6 Hz, IH), 6.84 (d, J= 2.8 Hz, IH), 7.03-7.06 (m, 2H), 7.29 (t, J = 1.6 Hz, 2H), 7.38 (d, J = 8.44 Hz, 2H), 7.52 (d, J = 8.8 Hz, 2H), 7.67 (dd, J = 1.6 & 6.4 Hz, 2H), 7.96 (d, J= 3.2 Hz, IH); LCMS : m/e 361.8 (M+l).
EXAMPLE 109
[00876] Preparation of 2-((4-(5-fluoro-2-(3-trifluoromethoxyphenylamino)pyrimidin-4- ylamino)phenyl)(hydroxy)methyl)acrylonitrile I- 163
Figure imgf000302_0002
1-163
[00877] The title compound was prepared according to the schemes, steps and intermediates described in Example 107 by using 3 -trifluoromethoxy aniline in place of 4 in Step-2. 1H NMR (DMSO-de) δ ppm: 5.29 (d, J= 3.8 Hz, IH), 6.13 (s, IH), 6.19 (s, IH), 6.31 (dd, J= 3.8 Hz, IH), 6.83 (d, J = 7.76 Hz, IH), 7.11 (d, J = 7.8 Hz, IH), 7.30-7.37 (m, 2H), 7.61-7.63 (m, 2H), 7.81 (s, IH), 7.90 (d, J = 7.4 Hz, IH), 8.16 (dd, J = 1.44 & 3.56 Hz, IH), 9.45 (s, IH), 9.53 (s, IH); LCMS : m/e 446 (M+ 1).
EXAMPLE 110
[00878] Preparation of N-(3-(5-fluoro-2-(3-(3-
(methylsulfony l)propoxy)phenylamino)pyrimidin-4-yloxy)phenyl)acrylamide 1-116
Figure imgf000303_0001
1-116
[00879] The title compound was prepared according to the schemes, steps and intermediates described in Example 98 by using 3-(3-methylsulfonyl)propoxyaniline in place of 4 in Step-2. 1H NMR (DMSO-de) δ ppm: 2.02-2.15 (m, 2H), 3.01 (s, 3H), 3.22 (t, J= 7.56 Hz, 2H), 3.88 (t, J = 6.12 Hz, 2H), 5.77 (dd, J= 1.84 & 10.12 Hz, IH), 6.25 (dd, J= 1.72 & 16.88 Hz, IH), 6.43 (d, J= 9.96 & 16.76 Hz, 2H), 6.95 (t, J= 8.12 Hz, IH), 7.06 (t, J= 7.48 Hz, 2H), 7.13 (s, IH), 7.44 (t, J = 8.12 Hz, IH), 7.56 (d, J = 8.44 Hz, IH), 7.68 (s, IH), 8.50 (d, J = 2.84 Hz, IH), 9.57 (s, IH), 10.34 (s, IH); LCMS : m/e 487.0 (M+2).
EXAMPLE 111
[00880] Preparation of N-(3-(5-cyclopropyl-2-(4-(2-methoxyethoxy)phenylamino)pyrimidin- 4-yloxy)phenyl)acrylamide 1-131
Figure imgf000303_0002
1-131 [00881] The title compound was prepared according to the steps and intermediates as described below.
Figure imgf000304_0001
1-131
A) K2CO3, DMA, rt, 5 h; B) PTSA, dioxane, 100 0C, 2 h; C) potassium cyclopropyltrifluoroborate, Pd(OAc)2, Xanphos, Cs2CO3, toluene, 100 0C, 12 h; D) 4N HCL, dioxane, rt, 1 hr; then acryoyl chloride, Et3N, DCM, -10 0C, 10 min [00882] Step-1
Figure imgf000304_0002
[00883] To a solution of 5-bromo-2,4-dichloropyrimidine (0.68g, 3.0 mmol) and tert-butyl 3- hydroxyphenylcarbamate (0.65g, 3.1 mmol) in DMA (4 mL) was added K2CO3 (0.83g, 6.0 mmol). The suspension was stirred for 5 hours. Water (15 ml) was added and the precipitate was collected by filtration. The solid was washed with ether and dried to yield 1.2 g of compound 3. MS: m/e=400.2, 402.2 (M+ 1). [00884]
Figure imgf000305_0001
[00885] To a solution of compound 3 (200 mg, 0.5 mmol) and 4-(2-methoxyethoxy)aniline (0.1 g, 0.6 mmol) in 5 ml dioxane was added 4-methylbenzenesulfonic acid monohydrate (0.08 g, 0.4 mmol). The mixture was stirred at 1000C for two hours. The solvent was evaporated. The residue was dissolved in 20 ml ethyl acetate and washed with NaHCO3 aqueous solution, water and brine. The organic layer was separated and dried over Na2SO4. After removal of solvent, the crude product was subject to chromatography on silica gel (hexane:EtOAc = 1 :1). 0.10 g of compound 5 was obtained: MS m/z: 531.1, 531.0 (M+H+). [00886] Step-3
Figure imgf000305_0002
[00887] Potassium cyclopropyltrifluoroborate (36 mg, 0.25 mmol), compound 5 (0.10 g, 0.19 mmol), palladium acetate (3.4 mg, 0.015 mmol), Xantphos (17.5 mg, 0.03 mmol) and Cs2CO3 (186 mg, 0.57 mmol) were suspended in 5 mL toluene and 1 mL water. The mixture was degassed, sealed under argon and heated at 100 0C for 12 hours. 20 mL ethyl acetate was added and washed with NaHCO3 aqueous solution, water and brine. The organic layer was separated and dried over Na2SO4. After removal of solvent, the crude product was subject to chromatography on silica gel (hexane:EtOAc = 1 :1). 50 mg of compound 6 was obtained: MS m/z: 493.2 (M+H+). [00888]
Figure imgf000306_0001
1-131
[00889] The title compound was prepared from compound 6 following the procedure described in Example 96. MS m/z: 447.1 (M+H+).
EXAMPLE 112
[00890] Preparation of l-(4-(5-fluoro-2-(3-(2-dimethylaminoethoxy)phenylamino)pyrimidin- 4-ylamino)phenyl)-2-methylprop-2-en- 1 -one 1-207
Figure imgf000306_0002
1-207
[00891] The title compound was prepared according to the schemes steps and intermediates described below.
Figure imgf000307_0001
A) 2, DIPEA, n-BuOH, 90 0C, 12 h; B) 4, Pd(OAc)2, BINAP, Cs2CO3, toluene, 110 0C, 16 h; C) LiOH, MeOH/THF/H2O, rt, 6 h; D) MeNHOMe.HCl, EDCLHCl, HOBT, DIPEA, DMF, rt, 3 h; E) 8, THF, O 0C to rt, 2 h. [00892] Step-1
Figure imgf000307_0002
[00893] A solution of 1 (4 g, 23.9 mmol), 2 (3.6 g, 23.7 mmol) and DIPEA (4.6 g, 35.58 mmol) in n-butanol (40 mL) was heated in a pressure tube (90 0C, 12 h). It was cooled, quenched with water (5 mL) and extracted with EtOAc (2 x 5 mL). The combined EtOAc extract was washed with water (60 mL), brine (40 mL), dried over Na2SO4 and concentrated under reduced pressure to afford 3 (5.5 g, 82 %) as an off- white solid. [00894]
Figure imgf000308_0001
[00895] A solution of 4 (0.319 g, 1.76 mmol), 3 (0.5 g, 1.76 mmol), Pd(OAc)2 (0.039 g, 0.17 mmol), BINAP (0.055 g, 0.08 mmol) and Cs2CO3 (1.44 g, 4.42 mmol) in degassed toluene (toluene was purged with N2 for 30 min) was heated for 16 h at 110 0C under N2 atmosphere. The reaction mixture was cooled, diluted with EtOAc (25 mL) and washed with water (5 mL), brine (2 mL) and dried over Na2SO4. Filtration followed by concentration under reduced pressure offered a residue which was further purified by column chromatography (SiO2, 60-120, chloroform/methanol, 9/1) to get 4 (0.63 g, 84%) as yellow solid. [00896] Step-3
Figure imgf000308_0002
[00897] To a stirred solution of 5 (0.3 g, 0.70 mmol) in methanol/THF/water : 1 mL/1 mL/0.5 mL was added LiOH (0.147 g, 3.52 mmol) and the reaction mixture was stirred at rt for 6 h. It was concentrated under reduced pressure; residue was diluted with water (2 mL) and extracted with diethyl ether (5 mL). The aqueous layer was separated and acidified with 1.5 N HCl (pH ~4-5) which was concentrated as such and dried under vacuum to get 6 (0.31 g, crude) as yellow gummy solid which was taken for next step without further purification. [00898]
Figure imgf000309_0001
[00899] To a stirred solution of 6 (0.29 g, 0.70 mmol) in DMF (3 niL) were added MeNH- OMe.HCl (0.068 g, 0.70 mmol), EDCLHCl (0.202 g, 1.05 mmol), HOBT (0.047 g, 0.35 mmol) and DIPEA (0.136 g, 1.05 mmol). The reaction mixture was stirred at room temperature for 3 h, quenched with water and extracted with EtOAc (2x5 mL). The combined organic layer was washed with brine, dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was further purified by column chromatography (SiO2, 60-120, methanol/chloroform : 20/80) to get 7 (0.061 g, 19%) as gummy yellow solid. [00900] Step-5
Figure imgf000309_0002
1-207
[00901] To a stirred solution of 7 (100 mg, 0.22 mmol) in THF (1 mL) at 0 0C was added 8 (17.6 mL, 8.80 mmol). The reaction mixture was allowed to stir at room temperature for 2 h. It was quenched with saturated NH4Cl solution (0.5 mL) and extracted with EtOAc (2x3 mL). The combined organic layer was washed with brine, dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to get a white solid. It was further purified by column chromatography (SiO2, 60-120, product getting eluted in 20% methanol/chloroform) to get 1-207 (9 mg. 9%) as gummy yellow solid. 1H NMR (DMSO-d6) δ ppm: 1.90 (s, 3H), 2.01 (s, 3H), 2.19 (s, 6H), 2.57 (t, J = 5.64 Hz, 2H), 3.87 (t, J = 5.84 Hz, 2H), 6.45 (dd, J = 1.64 & 8.08 Hz, IH), 6.82 (s, IH), 7.04 (t, J = 8.16 Hz, IH), 7.18 (d, J = 8.16 Hz, IH), 7.33 (s, IH), 7.47 (t, J = 7.88 Hz, 1H),7.62 (d, J = 7.72 Hz, IH), 8.13-8.16 (m, 3H), 9.24 (s, IH), 9.56 (s, IH); LCMS : m/e 450.1 (M+l).
EXAMPLE 113
[00902] Preparation of 1 -(4-(5 -fluoro-2-(phenylamino)pyrimidin-4-ylamino)phenyl)-3 - methylbut-2-en-l-one 1-206
Figure imgf000310_0001
1-206
[00903] The title compound was prepared according to the schemes, steps and intermediates described in Example 112 by using methyl 4-aminobenzoate in place of 2 in step-1 and aniline in place of 4 in step-2. 1H NMR (DMSO-d6) δ ppm : 1.98 (s, 3H), 2.12 (s, 3H), 6.90-7.00 (m, 2H), 7.20-7.30 (m, 2H), 7.65 (d, J = 8.16 Hz, 2H), 7.90 (d, J = 8.56 Hz, 2H), 7.98 (d, J = 8.68 Hz, 2H), 8.18 (bs, IH), 9.31 (s, IH), 9.68 (s, IH); LCMS : m/e 363.0 (M+l).
EXAMPLE 114
[00904] Preparation of l-(3-(5-fluoro-2-(3-(prop-2-ynyloxy)phenylamino)pyrimidin-4- ylamino)phenyl)-3 -methylbut-2-en- 1 -one 1-211
Figure imgf000310_0002
1-211
[00905] The title compound was prepared according to the schemes, steps and intermediates described in Example 112 by using 3-prop-2-ynyloxyaniline in place of 4 in step-2. 1H NMR (CD3OD) δ ppm: 2.0 (d, J= 1 Hz, 3H), 2.21 (d, J= 1.04 Hz, 3H), 2.94-2.96 (d, J = 2.44 Hz, IH), 4.59 (d, J = 2.36 Hz, 2H), 6.79-6.81 (m, 2H), 7.03 (dd, J= 3.12 & 8.04 Hz, IH), 7.14 (t, J= 2.2 Hz, IH), 7.23 (t, J= 8.12 Hz, IH), 7.54 (t, J= 7.92 Hz, IH), 7.83 (d, J= 7.96 Hz, IH), 7.86 (dd, J= 2.08 & 8.08 Hz, IH), 8.03 (d, J= 4.96 Hz, IH), 8.21 (t, J= 1.88 Hz, IH); LCMS : m/e 417.0 (M+l).
EXAMPLE 115
[00906] Preparation of l-(3-(5-fluoro-2-(3-(trifluoromethoxy)phenylamino)pyrimidin-4- ylamino)phenyl)-3-methylbut-2-en- 1 -one 1-223
Figure imgf000311_0001
1-223
[00907] The title compound was prepared according to the schemes, steps and intermediates described in Example 112 by using 3 -trifluoromethoxy aniline in place of 4 in step-2. 1H NMR (CDCl3) δ ppm: 2.0 (d, J = 1.08 Hz, 3H), 2.24 (d, J = 1.04 Hz, 3H), 6.74 (t, J = 1.24 Hz, IH), 6.85 (dd, J = 1.08 & 7.0 Hz, IH), 6.90 (s, IH), 7.08 (s, IH), 7.24-7.28 (m, IH), 7.35 (td, J = 1.2 & 7.44 Hz, IH), 7.49 (t, J = 7.88 Hz, IH), 7.63 (s, IH), 7.72 (td, J = 1.04 & 7.76 Hz, 1H),7.9O- 7.92 (m, IH), 8.00-8.05 (m, 2H); LCMS : m/e 447 (M+l).
EXAMPLE 116
[00908] Preparation of l-(3-(5-fluoro-2-(3-(trifluoromethoxy)phenylamino)pyrimidin-4- ylamino)phenyl)-2-methylprop-2-en-l-one 1-199
Figure imgf000312_0001
1-199
[00909] The title compound was prepared according to the schemes, steps and intermediates described in Example 112 by using 3 -trifluoromethoxy aniline in place of 4 in step-2 and isopropenylmagnesium bromide in place of 8 in step-5. 1H NMR (DMSO-dβ) δ ppm: 1.97 (s, 3H), 5.6 (s, IH), 6.0 (d, J = 0.96 Hz, IH), 6.82 (d, J = 8.08 Hz, IH), 7.27 (t, J = 8.2 Hz, IH), 7.41 (dd, J= 1.12 & 7.56 Hz, IH), 7.48 (t, J= 7.76 Hz, IH), 7.60 (dd, J= 1.28 & 7.88 Hz, IH), 7.78 (s, IH), 7.90 (d, J= 1.64 Hz, IH), 8.15 (d, J= 8 Hz, IH), 8.19 (d, J= 3.64 Hz, IH), 9.55 (s, IH), 9.65 (s, IH); LCMS : m/e 433 (M+ 1).
EXAMPLE 117
[00910] Preparation of 1 -(4-(5 -fluoro-2-(phenylamino)pyrimidin-4-ylamino)phenyl)-2- methylprop-2-en-l-one 1-185
Figure imgf000312_0002
1-185
[00911] The title compound was prepared according to the schemes, steps and intermediates described in Example 112 by using methyl 4-aminobenzoate in plave of 2 in step-1, aniline in place of 4 in step-2 and isopropenylmagnesium bromide in place of 8 in step-5. 1H NMR (DMSO-de) δ ppm: 1.99 (s, 3H), 5.54 (s, IH), 1.01 (s, IH), 6.93 (t, J= 7.36 Hz, IH), 7.24 (t, J = 7.52 Hz, 2H), 7.66-7.72 (m, 4H), 8.01 (d, J = 8.72 Hz, 2H), 8.19 (d, J = 3.6 Hz, IH), 9.32 (s, IH), 9.72 (s, IH); LCMS : m/e 348.8 (M+l).
EXAMPLE 118
[00912] Preparation of N-(3-(2-(3-(2-(dimethylamino)ethoxy)phenylamino)-5- fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-233
Figure imgf000313_0001
1-233
[00913] The title compound was prepared according to the steps, schemes and intermediates described in Example 20 by using 3-(2-dimethylaminoethoxy)aniline in place of 4 in step-2. 1H NMR (CD3OD) δ ppm: 2.31 (s, 6H), 2.76 (t, J= 5.6 Hz, 2H), 3.97 (t, J= 5.2 Hz, 2H), 5.78 (dd, J = 2 & 9.2 Hz, IH), 6.38-6.42 (m, 2H), 6.52-6.55 (m, IH), 7.1-7.11 (m 2H), 7.30 (t, J = 8.0 Hz, IH), 7.36 (s, IH), 7.42-7.48 (m, 2H), 7.94 (d, J = 3.6 Hz, IH), 8.05 (s, IH); LCMS : m/e 437 (M+l).
EXAMPLE 119
[00914] Preparation of N-(3-(5-fluoro-4-(4-phenoxyphenoxy)pyrimidin-2- ylamino)phenyl)acrylamide 1-130
Figure imgf000313_0002
[00915] The title compound was prepared according to the steps, schemes and intermediates described in Example 11 by using 5-fluoro-2,4-dichloropyrimidine in place of 1 in step-1. 1H NMR (DMSO-d6) δ ppm: 5.71 (dd, J= 1.6 & 10 Hz, IH), 6.22 (dd, J= 1.6 & 16.8 Hz, IH), 6.44 (dd, J= 10.4 & 17.2 Hz, IH), 6.98-7.05 (m, 3H), 7.1-7.12 (m, 2H), 7.17 (t, J= 7.2 Hz, IH), 7.24 (t, J = 7.6 Hz, 2H), 7.35-7.37 (m, 2H), 7.42 (t, J = 8.4 Hz, 2H), 7.71 (s, IH), 8.5 (s, IH), 9.6 (s, IH), 10.05 (s, IH); LCMS : m/e 443.0 (M+l).
EXAMPLE 120
[00916] Preparation of (S)-N-(3-(5-fluoro-2-(4-(tetrahydrofuran-3- yloxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-43
Figure imgf000314_0001
[00917] The title compound was prepared according to the steps, schemes and intermediates described in Example 20 by using (S)-4-(tetrahydrofuran-3-yloxyaniline in place of 4 in step-2. 1H NMR (DMSO-J6, 500 MHz): δ 10.10 (s, IH), 9.35 (s, IH), 8.95 (s, IH), 8.05 (d, J = 4.0 Hz, IH), 7.92 (s, IH), 7.52 (d, J = 9.0 Hz, 2H), 7.47 (d, J = 7.5 Hz, IH), 7.41 (d, J = 8.5 Hz, IH), 7.27 (t, J = 8.0 Hz, IH), 6.72 (d, J = 9.0 Hz, 2H), 6.45 (dd, J = 1.5, 17.0 Hz, IH), 6.25 (dd, J = 1.1, 16.5 Hz, IH), 5.75 (dd, J= 1.1, 10.0 Hz, IH), 4.93 - 4.84 (m, IH), 3.88 - 3.72 (m, 4H), 2.20 - 2.10 (m, IH), 1.97 - 1.90 (m, IH). MS m/e = 436 [M++l]
EXAMPLE 121
[00918] Preparation of (R)-N-(3-(5-fluoro-2-(4-(tetrahydrofuran-3- yloxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-46
Figure imgf000315_0001
1-46
[00919] The title compound was prepared according to the steps, schemes and intermediates described in Example 20 by using (R)-4-(tetrahydrofuran-3-yloxyaniline in place of 4 in step-2. 1H NMR (DMSO-J6, 500 MHz): δ 10.10 (s, IH), 9.35 (s, IH), 8.95 (s, IH), 8.05 (d, J = 4.0 Hz, IH), 7.92 (s, IH), 7.52 (d, J = 9.0 Hz, 2H), 7.47 (d, J = 7.5 Hz, IH), 7.41 (d, J = 8.5 Hz, IH), 7.27 (t, J = 8.0 Hz, IH), 6.72 (d, J = 9.0 Hz, 2H), 6.45 (dd, J = 1.5, 17.0 Hz, IH), 6.25 (dd, J = 1.1, 16.5 Hz, IH), 5.75 (dd, J= 1.1, 10.0 Hz, IH), 4.93 - 4.84 (m, IH), 3.88 - 3.72 (m, 4H), 2.22 - 2.14 (m, IH), 1.97 - 1.90 (m, IH). MS: m/e = 436 [M++l]
EXAMPLE 122
[00920] Preparation of N-(3-(5-fiuoro-2-(3-((l-methylpiperidin-3- yl)methoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-acrylamide I- 76)
Figure imgf000315_0002
1- 76
[00921] The title compound was prepared according to the steps, schemes and intermediates described in Example 20 by using 3-(l-methylpiperidin-3-yl)methoxyaniline in place of 4 in step-2. 1H NMR (DMSO-J6, 500 MHz): δ 10.08 (s, IH), 9.41 (s, IH), 9.09 (s, IH), 8.11 (d, J = 3.5 Hz, IH), 7.90 (s, IH), 7.57 (d, J = 8.0 Hz, IH), 7.41 (d, J= 8.0 Hz, IH), 7.32 (s, IH), 7.30 - 7.20 (m, 2H), 7.03 (t, J= 8.0 Hz, IH), 6.50 - 6.40 (m, 2H), 6.24 (dd, J= 1.5, 17.0 Hz, IH), 5.74 (dd, J = 2.0, 10.5 Hz, IH), 3.75 - 3.65 (m, 2H), 2.73 (d, J = 10 Hz, IH), 2.60 (d, J = 10.5 Hz, IH), 2.13 (s, 3H), 1.98 - 1.83 (m, 2H), 1.75 - 1.58 (m, 3H), 1.55 - 1.45 (m, IH), 1.05 - 0.95 (m, IH). MS: m/e = 477 (M++l).
EXAMPLE 123
[00922] Preparation of N-(3-(5-fluoro-2-(3-((l-methylpiperidin-4- yl)methoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-acrylamide 1-82
Figure imgf000316_0001
1-82
[00923] The title compound was prepared according to the steps, schemes and intermediates described in Example 20 by using 3-(l-methylpiperidin-4-yl)methoxyaniline in place of 4 in step-2. 1H-NMR (CDC13+DMSO-D6, 500 MHz): δ 9.04 (bs, IH), 8.30 (s, IH), 7.96 (s, IH), 7.75 (s, IH), 7.63 (s, IH), 7.59 (d, J = 7.0 Hz, IH), 7.38 - 7.33 (m, 2H), 7.27 (t, J = 8.5 Hz, IH), 7.16 (t, J = 8 Hz, IH), 7.09 (d, J = 8.5 Hz, IH), 6.52 (d, J = 7.0 Hz, IH), 6.51 - 6.38 (m, 2H), 5.73 (dd, J = 2.0, 9.0 Hz, IH), 3.77 (d, J = 6.0 Hz, 2H), 2.90 - 2.84 (m, 2H), 2.28 (s, 3H), 1.95 (t, J = 10.0 Hz, IH), 1.85 - 1.74 (m, 2H), 1.45 - 1.32 (m, 2H), 1.28 - 1.25 (m, 2H). MS: m/e = 477 (M++l). EXAMPLE 124
[00924] Preparation of N-(3-(2-(3-(4-(2-hydroxyethyl)piperazin-l-yl)phenylamino)-5- fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-83
Figure imgf000317_0001
1-83
[00925] The title compound was prepared according to the steps, schemes and intermediates described in Example 20 by using 3-(4-(2-t-butyldimethylsilyloxyethyl)piperazin-l-ylaniline in place of 4 in step-2 and deprotecting the TBS ether with TFA in DCM as a step-5. 1H NMR (DMSO-J6, 500 MHz): δ 8.99 (s, IH), 8.85 (s, IH), 8.04 (d, J= 4.0 Hz, IH), 7.28 - 7.19 (m, 2H), 7.08 - 7.00 (m, 2H), 6.94 (t, J= 3.5 Hz, 2H), 6.48 (dd, J= 2.0, 8.0 Hz, IH), 6.35 - 6.31 (m, IH), 4.94 (s, 2H), 3.71 (t, J = 6.0 Hz, 2H), 3.02 (t, J = 4.5 Hz, 4H), 2.57 - 2.50 (m, 4H), 2.46 (t, J = 6.0 Hz, 2H), 0.87 (s, 9H), 0.05 (s, 6H). MS: m/e = 538 (M++l).
EXAMPLE 125
[00926] Preparation of N-(4-(5-fluoro-2-(3-methoxyphenylamino)pyrimidin-4- ylamino)benzyl)-N-methylacrylamide 1-113
Figure imgf000317_0002
1-113
[00927] The title compound was prepared according to the steps, schemes and intermediates described in Example 20 by using 4-(N-methyl-N-tert-butyloxycarbonylamino)methylaniline in place of 2 in step-1 and 3 -methoxy aniline in place of 4 in step-2. 1H-NMR (DMSO-dβ, 200 MHz): δ 9.36 (bs, IH), 9.16 (bs, IH), 8.10 (d, J = 3.4 Hz, IH), 7.83 - 7.70 (m, 2H), 7.34 (bs, IH), 7.26 - 7.01 (m, 4H), 6.86 - 6.73 (m, IH), 6.48 (d, J = 8.0 Hz, IH), 6.21 (dd, J = 16.4, 2.2 Hz, IH), 5.76- 5.64 (m, IH), 4.64 - 4.53 (two s, 2H), 3.65 (s, 3H), 3.00 - 2.88 (two s, 3H). MS: ml Q = 408.2 [M++l].
EXAMPLE 126
[00928] Preparation of N-(4-(5-fluoro-2-(6-methoxypyridin-3-ylamino)pyrimidin-4- ylamino)benzyl)-N-methylacrylamide 1-114
Figure imgf000318_0001
1-114
[00929] The title compound was prepared according to the steps, schemes and intermediates described in Example 20 by using 4-(N-methyl-N-tert-butyloxycarbonylamino)methylaniline in place of 2 in step-1 and 6-methoxy-3-aminopyridine in place of 4 in step-2. 1H-NMR (DMSO- D6, 200 MHz): δ 9.36 (bs, IH), 9.09 (bs, IH), 8.32 (bs, IH), 8.06 (dd, J = 3.8 Hz, IH), 7.97- 7.92 (m, IH), 7.76- 7.68 (m, 2H), 7.20 - 7.08 (m, 2H), 6.87 - 6.75 (m, IH), 6.72 (d, J = 8.4 Hz, IH), 6.22 (dd, J = 19.4, 2.6 Hz, IH), 5.74 - 5.65 (m, IH), 4.64 - 4.53 (two s, 2H), 3.78 (s, 3H), 3.00 - 2.88 (two s, 3H). MS: m/e = 409 (M++l).
EXAMPLE 127
[00930] Preparation of N-(3-(5-fluoro-2-(3-methyoxyphenylamino)pyrimidin-4- ylamino)benzyl)-N-methylacrylamide 1-115
Figure imgf000318_0002
[00931] The title compound was prepared according to the steps, schemes and intermediates described in Example 20 by using 3-(N-methyl-N-tert-butyloxycarbonylamino)methylaniline in place of 2 in step-1 and 3 -methoxy aniline in place of 4 in step-2. 1H-NMR (DMSO-J6, 200 MHz): δ 9.50 - 9.30 (m, IH), 9.15 - 8.96 (m, IH), 8.15 (bs, IH), 7.82 - 7.59 (m, 2H), 7.45 - 7.00 (m, 4H), 6.97 - 6.65 (m, 2H), 6.55 - 6.45 (m, IH), 6.26 - 6.12 (m, IH), 5.78 - 5.60 (m, IH), 4.68 (s, IH), 4.55 & 3.75 (two s, 3H), 2.90 & 3.00 (two s, 3H). MS: m/e = 408.2 [M++l].
EXAMPLE 128
[00932] Preparation of N-(3-(5-fluoro-2-(3-(4-(2-hydroxyethyl)piperazin-l- yl)phenylamino)pyrimidin-4-yloxy) phenyl)acrylamide 1-84
Figure imgf000319_0001
1-84
[00933] The title compound was prepared according to the steps, schemes and intermediates described in Example 98 by using 3-(4-(2-t-butyldimethylsilyloxyethyl)piperazin-l-ylaniline in place of 4 in step-2 and deprotecting the TBS ether with TFA in DCM as a step-5. 1H-NMR (DMSO-D6, 500 MHz): δ 8.19 (s, IH), 7.75 - 7.70 (m, 2H), 7.42 - 7.35 (m, 2H), 7.12 - 7.05 (m, 2H), 6.97 (dd, J = 2.0, 10.5 Hz, IH), 6.93 (s, IH), 6.72 (d, J = 6.5 Hz, IH), 6.57 - 6.54 (m, IH), 6.44 (d, J = 17.0 Hz, IH), 6.29 - 6. 20 (m, IH), 5.78 (d, J = 10.0 Hz, IH), 3.68 (t, J = 5.5 Hz, 2H), 3.00 - 2.94 (m, 4H), 2.63 - 2.56 (m, 6H). MS: m/e = 479 (M++l). EXAMPLE 129
[00934] Preparation of N-(3-(5-fiuoro-2-(3-((l-methylpiperidin-3- yl)methoxy)phenylamino)pyrimidin-4-yloxy)phenyl)acrylamide 1-81
Figure imgf000320_0001
1-81
[00935] The title compound was prepared according to the steps, schemes and intermediates described in Example 98 by using 3-(l-methylpiperidin-3-yl)methoxyaniline in place of 4 in step-2. 1H-NMR (CDCl3, 500 MHz): δ 8.19 (d, J= 2.5 Hz, IH), 7.82 - 7.75 (m, 2H), 7.42 - 7.36 (m, 2H), 7.08 - 7.02 (m, 3H), 6.99 (d, J = 7.0 Hz, IH), 6.91 (s, IH), 6.79 (d, J = 7.5 Hz, IH), 6.48 - 6.44 (m, IH), 6.42 (s, IH), 6.29 - 6.23 (m, IH), 5.77 (d, J = 10 Hz, IH), 3.67 - 3.62 (m, 2H), 2.95 - 2.91 (m, IH), 2.82 - 2.76 (m, IH), 2.28 (s, 3H), 2.11 - 2.05 (m, IH), 1.98 - 1.92 (m, IH), 1.79 - 1.70 (m, 3H), 1.11 - 1.04 (m, IH). MS: m/e = 478 (M++l).
EXAMPLE 130
[00936] Preparation of N-(3-(5-fiuoro-2-(3-((l-methylpiperidin-4- yl)methoxy)phenylamino)pyrimidin-4-yloxy)phenyl)-acrylamide 1-75
Figure imgf000320_0002
1-75 [00937] The title compound was prepared according to the steps, schemes and intermediates described in Example 98 by using 3-(l-methylpiperidin-4-yl)methoxyaniline in place of 4 in step-2. 1H-NMR (DMSO-D6, 500 MHz): δ 10.3 l(s, IH), 9.50 (s, IH), 8.50 (s, IH), 7.67 (s, IH), 7.55 (d, J = 8.0 Hz, IH), 7.42 (t, J = 8.0 Hz, IH), 7.10 (s, IH), 7.04 (d, J = 7.0 Hz, 2H ), 6.94 (t, J = 8.0 Hz, IH ), 6.45 - 6.39 (m, 2H), 6.27 (d, J = 15.0 Hz, IH ), 5.78 (dd, J = 2.0, 10.5 Hz, IH), 3.64 (d, J = 6.0 Hz, 2H), 2.75 (d, J = 6.5 Hz, 2H), 2.14 (s, 3H), 1.83 (t, J = 10.5 Hz, 2H), 1.66 - 1.64 (m, 3H), 1.25 - 1.23 (m, 2H). MS: m/e = 478 (M++l).
EXAMPLE 131
[00938] Preparation of N-(3-(5-cyano-2-(phenylamino)pyrimidin-4- ylamino)phenyl)acrylamide 1-157
Figure imgf000321_0001
1-157
[00939] The title compound was prepared according to the schemes, steps and intermediates described in Example 94, by using 2,4-dichloro-5-cyanopyrimidine in the place of 4 in Step 2. MS 379.1 (M+Na).
EXAMPLE 132
[00940] Preparation of N-(3-(5-fluoro-2-(3-(trifluoromethoxy)phenylamino)pyrimidin-4- ylamino)phenyl)acrylamide 1-244
Figure imgf000321_0002
1-244 [00941] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 3-(trifluoromethoxy)aniline in the place of 4 in Step 2. MS 434.1 (M+l).
EXAMPLE 133
[00942] Preparation of N-(3-(5-fluoro-2-(pyridin-3-ylamino)pyrimidin-4- ylamino)phenyl)acrylamide 1-234
Figure imgf000322_0001
1-234
[00943] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 3-aminopyridine in the place of 4 in Step 2. MS 351.1 (M+l).
EXAMPLE 134
[00944] Preparation of N-(3-(5-fluoro-2-(4-fluorophenylamino)pyrimidin-4- ylamino)phenyl)acrylamide I- 247
Figure imgf000322_0002
1- 247
[00945] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 4-fluoroaniline in the place of 4 in Step 2. MS 368.1 (M+l) EXAMPLE 135
[00946] Preparation of N-(3-(5-fluoro-2-(3-(3-morpholinopropoxy)phenylamino)pyrimidin-
4-ylamino)phenyl)acrylamide 1-208
Figure imgf000323_0001
1- 208
[00947] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 3-(3-morpholinopropoxy)aniline in the place of 4 in Step 2. MS 515.3 (M+Na).
EXAMPLE 136
[00948] Preparation of N-(3-(2-(3-(3-(lH-imidazol-l-yl)propoxy)phenylamino)-5- fluoropyrimidin-4-ylamino)phenyl)acrylamide I- 204
Figure imgf000323_0002
1- 204
[00949] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 3-(3-(lH-imidazol-l-yl)propoxy)aniline in the place of 4 in Step 2. MS 474.3 (M+Na). EXAMPLE 137
[00950] Preparation of N-(3-(2-(l-acetylpiperidin-3-ylamino)-5-fluoropyrimidin-4- ylamino)phenyl)acrylamide 1-238
Figure imgf000324_0001
1-238
[00951] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using l-(3-aminopiperidin-l-yl)ethanone in the place of 4 in Step 2. MS 421.1 (M+Na).
EXAMPLE 138
[00952] Preparation of N-(3-(5-fluoro-2-(phenylamino)pyrimidin-4-yloxy)phenyl)acrylamide 1-228
Figure imgf000324_0002
1-228
[00953] The title compound was prepared according to the schemes, steps and intermediates described in Example 98, by using aniline in the place of 4 in Step 2. MS 351.3 (M+l). EXAMPLE 139
[00954] Preparation of N-(3-(5-fluoro-2-(6-methoxypyridin-3-ylamino)pyrimidin-4- ylamino)phenyl)acrylamide 1-243
Figure imgf000325_0001
1-243
[00955] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 6-methoxypyridin-3 -amine in the place of 4 in Step 2. MS 381.1 (M+l).
EXAMPLE 140
[00956] Preparation of N-(3-(5-methoxy-2-(3-methoxyphenylamino)pyrimidin-4- ylamino)phenyl)acrylamide. 1-158
Figure imgf000325_0002
1-158
[00957] The title compound was prepared according to the schemes, steps and intermediates described in Example 1, by using 5-methoxy-2,4-dichloropyrimidine in the place of 1 in Step 1 and 3 -methoxy aniline in place of 4 in step 2. MS 392.3 (M+l).
EXAMPLE 141
[00958] Preparation of N-(3-(5-methoxy-2-(6-methoxypyridin-3-ylamino)pyrimidin-4- ylamino)phenyl)acrylamide 1-192
Figure imgf000326_0001
1-192
[00959] The title compound was prepared according to the schemes, steps and intermediates described in Example 1, by using 5-methoxy-2,4-dichloropyrimidine in the place of 1 in Step 1 and 5-amino-2-methoxypyridine in place of 4 in step 2. MS 393.3 (M+l).
EXAMPLE 142
[00960] Preparation of N-(3 -(5 -fluoro-2-(6-methoxypyridin-3 -ylamino)pyrimidin-4- yloxy)phenyl)acrylamide 1-222
Figure imgf000326_0002
1-222
[00961] The title compound was prepared according to the schemes, steps and intermediates described in Example 98, by using 3-amino-6-methoxypyridine in the place of 4 in Step 2. MS 382.3 (M+l).
EXAMPLE 143
[00962] Preparation of 4-(3-acrylamidophenylamino)-N-tert-butyl-2-(6-methoxypyridin-3- ylamino)pyrimidine-5 -carboxamide 1-216
Figure imgf000327_0001
1-216
[00963] The title compound was prepared according to the schemes, steps and intermediates described in Example 37, by using tert-butylamine in the place of 2 in Step-1 and omitting Step- 6. MS 484.3 (M+Na).
EXAMPLE 144
[00964] Preparation of (R)-l-(3-(5-fluoro-2-(6-methoxypyridin-3-ylamino)pyrimidin-4- ylamino)piperidin- 1 -yl)prop-2-en- 1 -one 1-202
Figure imgf000327_0002
1-202
[00965] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using (R)-tert-butyl 3-aminopiperidine-l-carboxylate in the place of 2 in Step 1 and 3-amino-6-methoxypyridine in place of 4 in step 2. MS 395.3 (M+Na). EXAMPLE 145
[00966] Preparation of (R)-l-(3-(5-fluoro-2-(3-methoxyphenylamino)pyrimidin-4- ylamino)piperidin- 1 -yl)prop-2-en- 1 -one 1-195
Figure imgf000328_0001
1-195
[00967] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using (R)-tert-butyl 3-aminopiperidine-l-carboxylate in the place of 2 in Step 1 and 3 -methoxy aniline in place of 4 in step 2. MS 394.3 (M+Na).
EXAMPLE 146
[00968] Preparation of (S)-l-(3-(5-fluoro-2-(6-methoxypyridin-3-ylamino)pyrimidin-4- ylamino)piperidin- 1 -yl)prop-2-en- 1 -one 1-197
Figure imgf000328_0002
1-197
[00969] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using (S)-tert-butyl 3-aminopiperidine-l-carboxylate in the place of 2 in Step 1 and 3-amino-6-methoxypyridine in place of 4 in step 2. MS 373.3 (M+l). EXAMPLE 147
[00970] Preparation of (S)-l-(3-(5-fluoro-2-(3-methoxyphenylamino)pyrimidin-4- ylamino)piperidin- 1 -yl)prop-2-en- 1 -one 1-196
Figure imgf000329_0001
1-196
[00971] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using (S)-tert-butyl 3-aminopiperidine-l-carboxylate in the place of 2 in Step 1 and 3 -methoxy aniline in place of 4 in step 2. MS 372.3 (M+l).
EXAMPLE 148
[00972] Preparation of (R)-l-(3-(5-fluoro-2-(6-methoxypyridin-3-ylamino)pyrimidin-4- yloxy)piperidin- 1 -yl)prop-2-en- 1 -one 1-180
Figure imgf000329_0002
1-180
[00973] The title compound was prepared according to the schemes, steps and intermediates described in Example 98, by using (R)-tert-butyl 3-hydroxypiperidine-l-carboxylate in the place of 2 in Step 1 and 3-amino-6-methoxypyridine in place of 4 in step 2. MS 374.3 (M+l). EXAMPLE 149
[00974] Preparation of (R)-l-(3-(5-fluoro-2-(3-methoxyphenylamino)pyrimidin-4- yloxy)piperidin- 1 -yl)prop-2-en- 1 -one 1-190
Figure imgf000330_0001
1-190
[00975] The title compound was prepared according to the schemes, steps and intermediates described in Example 98, by using (R)-tert-butyl 3-hydroxypiperidine-l-carboxylate in the place of 2 in Step 1 and 3 -methoxy aniline in place of 4 in step 2. MS 395.3 (M+Na).
EXAMPLE 150
[00976] Preparation of (S)-l-(3-(5-fluoro-2-(6-methoxypyridin-3-ylamino)pyrimidin-4- yloxy)piperidin- 1 -yl)prop-2-en- 1 -one 1-193
Figure imgf000330_0002
1-193
[00977] The title compound was prepared according to the schemes, steps and intermediates described in Example 98, by using (S)-tert-butyl 3-hydroxypiperidine-l-carboxylate in the place of 2 in Step 1 and 3-amino-6-methoxypyridine in place of 4 in step 2. MS 396.3 (M+Na). EXAMPLE 151
[00978] Preparation of (S)-l-(3-(5-fluoro-2-(3-methoxyphenylamino)pyrimidin-4- yloxy)piperidin- 1 -yl)prop-2-en- 1 -one 1-179
Figure imgf000331_0001
1-179
[00979] The title compound was prepared according to the schemes, steps and intermediates described in Example 98, by using (S)-tert-butyl 3-hydroxypiperidine-l-carboxylate in the place of 2 in Step 1 and 3 -methoxy aniline in place of 4 in step 2. MS 395.3 (M+Na).
EXAMPLE 152
[00980] Preparation of l-(3-(5-fluoro-2-(6-methoxypyridin-3-ylamino)pyrimidin-4- ylamino)pyrrolidin- 1 -yl)prop-2-en- 1 -one 1-203
Figure imgf000331_0002
1-203
[00981] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using tert-butyl 3-aminopyrrolidine-l-carboxylatein the place of 2 in Step 1 and 3-amino-6-methoxypyridine in place of 4 in step 2. MS 381.3 (M+Na). EXAMPLE 153
[00982] Preparation of l-(3-(5-fluoro-2-(3-methoxyphenylamino)pyrimidin-4- ylamino)pyrrolidin- 1 -yl)prop-2-en- 1 -one 1-201
Figure imgf000332_0001
1-201
[00983] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using tert-butyl 3-aminopyrrolidine-l-carboxylate in the place of 2 in Step 1 and 3 -methoxy aniline in place of 4 in step 2. MS 358.3 (M+l).
EXAMPLE 154
[00984] Preparation of (R)-l-(3-(5-fluoro-2-(3-methoxyphenylamino)pyrimidin-4- ylthio)piperidin- 1 -yl)prop-2-en- 1 -one 1-137
Figure imgf000332_0002
1-137
[00985] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using (S)-tert-butyl 3-mercaptopiperidine-l-carboxylate in the place of 2 in Step 1 and 3 -methoxy aniline in place of 4 in step 2. MS 411.1 (M+Na). EXAMPLE 155
[00986] Preparation of (R)-l-(3-(2-(3-chlorophenylamino)-5-fluoropyrimidin-4- ylamino)piperidin- 1 -yl)prop-2-en- 1 -one 1-147
Figure imgf000333_0001
1-147
[00987] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using (S)-tert-butyl 3-aminopiperidine-l-carboxylate in the place of 2 in Step 1 and 3-chloroaniline in place of 4 in step 2. MS 376.1 (M+l).
EXAMPLE 156
[00988] Preparation of (R)-l-(3-(5-fluoro-2-(3-(2-morpholinoethoxy)phenylamino)pyrimidin- 4-ylamino)piperidin- 1 -yl)prop-2-en- 1 -one 1-135
Figure imgf000333_0002
1-135
[00989] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using (S)-tert-butyl 3-aminopiperidine-l-carboxylate in the place of 2 in Step 1 and 3-(2-morpholinoethoxy)aniline in place of 4 in step 2. MS 471.3 (M+l). EXAMPLE 157
[00990] Preparation of (E)-4-(dimethylamino)-N-(3-(5-fluoro-2-(6-methoxypyridin-3- ylamino)pyrimidin-4-ylamino)phenyl)but-2-enamide 1-125
Figure imgf000334_0001
1-125
[00991] The title compound was prepared according to the schemes, steps and intermediates described in Example 139, by using (E)-4-(dimethylamino)but-2-enoyl chloride in the place of 7 in Step 4. MS 460.1 (M+Na).
EXAMPLE 158
[00992] Preparation of 2-((lH-pyrazol-l-yl)methyl)-N-(3-(5-fluoro-4-(m- tolylamino)pyrimidin-2-ylamino)phenyl)acrylamide. 1-98
Figure imgf000334_0002
1-98
[00993] The title compound was prepared according to the schemes, steps and intermediates described in Example 4, by using 2-((lH-pyrazol-l-yl)methyl)acryloyl chloride in the place of 6 in Step 3. MS 466.1 (M+Na).
EXAMPLE 159
[00994] Preparation of (E)-4-(azetidin-l-yl)-N-(3-(5-fluoro-4-(m-tolylamino)pyrimidin-2- ylamino)phenyl)but-2-enamide 1-123
Figure imgf000335_0001
1-123
[00995] The title compound was prepared according to the schemes, steps and intermediates described in Example 4, by using (E)-4-(azetidin-l-yl)but-2-enoyl chloride in the place of 6 in Step 3. MS 455.1 (M+Na).
EXAMPLE 160
[00996] Preparation of (E)-N-(3-(5-fluoro-4-(m-tolylamino)pyrimidin-2-ylamino)phenyl)-4- morpholinobut-2-enamide 1-102
Figure imgf000335_0002
1-102
[00997] The title compound was prepared according to the schemes, steps and intermediates described in Example Example 4, by using (E)-4-(morpholin-4-yl)but-2-enoyl chloride in the place of 6 in Step 3. MS 485.3 (M+Na).
EXAMPLE 161
[00998] Preparation of (E)-4-((lS,4S)-2,5-diazabicyclo[2.2.1]heptan-2-yl)-N-(3-(5-fiuoro-4- (m-tolylamino)pyrimidin-2-ylamino)phenyl)but-2-enamide 1-101
Figure imgf000336_0001
1-101
[00999] The title compound was prepared according to the schemes, steps and intermediates described in Example 4, by using (E)-4-((lS,4S)-2,5-diazabicyclo[2.2.1]heptan-2-yl)but-2-enoyl chloride in the place of 6 in Step 3. MS 496.1 (M+Na).
EXAMPLE 162
[001000] Preparation of (E)-N-(3-(5-fluoro-4-(m-tolylamino)pyrimidin-2- ylamino)phenyl)-4-((2-methoxyethyl)(methyl)amino)but-2-enamide 1-120
Figure imgf000336_0002
1-120
[001001] The title compound was prepared according to the schemes, steps and intermediates described in Example 4, by using (E)-4-((2-methoxyethyl)(methyl)amino)but-2- enoyl chloride in the place of 6 in Step 3. MS 487.3 (M+Na).
EXAMPLE 163
[001002] Preparation of (S,E)-N-(3-(5-fluoro-4-(m-tolylamino)pyrimidin-2- ylamino)phenyl)-4-(3 -hydroxypyrrolidin- 1 -yl)but-2-enamide 1-99
Figure imgf000337_0001
1-99
[001003] The title compound was prepared according to the schemes, steps and intermediates described in Example 4, by using (S,E)-4-(3-hydroxypyrrolidin-l-yl)but-2-enoyl chloride in the place of 6 in Step 3. MS 485.3 (M+Na)
EXAMPLE 164
[001004] Preparation of (R,E)-N-(3-(5-fluoro-4-(m-tolylamino)pyrimidin-2- ylamino)phenyl)-4-(3 -hydroxypyrrolidin- 1 -yl)but-2-enamide I- 104 H
Figure imgf000337_0002
1-104
[001005] The title compound was prepared according to the schemes, steps and intermediates described in Example 4, by using (R,E)-4-(3 -hydroxypyrrolidin- l-yl)but-2-enoyl in the place of 6 in Step 3. MS 485.3 (M+Na). EXAMPLE 165
[001006] Preparation of (E)-N-(3-(5-fluoro-4-(m-tolylamino)pyrimidin-2-ylamino)phenyl)- 4-(lH-pyrazol-l-yl)but-2-enamide I-100
Figure imgf000338_0001
1-100
[001007] The title compound was prepared according to the schemes, steps and intermediates described in Example 4, by using (E)-4-(lH-imidazol-l-yl)but-2-enoyl chloride in the place of 6 in Step 3. MS 466.1 (M+Na).
EXAMPLE 166
[001008] Preparation of (R,E)-N-(3-(5-fluoro-2-(4-(2- methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-4-(3 -hydroxypyrrolidin- 1 -yl)but-2- enamide 1-89
Figure imgf000338_0002
1-89
[001009] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using (R,E)-4-(3 -hydroxypyrrolidin- l-yl)but-2-enoyl chloride in the place of 7 in Step 4. MS 545.3 (M+Na). EXAMPLE 167
[001010] Preparation of (S,E)-N-(3-(5-fluoro-2-(4-(2- methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-4-(3 -hydroxypyrrolidin- 1 -yl)but-2- enamide 1-88
Figure imgf000339_0001
1-88
[001011] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using (S,E)-4-(3-hydroxypyrrolidin-l-yl)but-2-enoyl chloride in the place of 7 in Step 4. MS 545.3 (M+Na).
EXAMPLE 168
[001012] Preparation of 2-((lH-pyrazol-l-yl)methyl)-N-(3-(5-fiuoro-2-(4-(2- methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-85
Figure imgf000339_0002
1-85 [001013] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 2-((lH-pyrazol-l-yl)methyl)acryloyl chloride in the place of 7 in Step 4. MS 526.1 (M+Na).
EXAMPLE 169
[001014] Preparation of N-(3-(5-fluoro-2-(phenylamino)pyrimidin-4- ylamino)phenyl)acrylamide 1-28
Figure imgf000340_0001
1-28
[001015] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using aniline in the place of 4 in Step 2. MS 372.1 (M+Na).
EXAMPLE 170
[001016] Preparation of (E)-4-((3R,5S)-3,5-dimethylpiperazin-l-yl)-N-(3-(5-fluoro-4-(m- tolylamino)pyrimidin-2-ylamino)phenyl)but-2-enamide 1-119
Figure imgf000340_0002
1-119
[001017] The title compound was prepared according to the schemes, steps and intermediates described in Example 4, by using (E)-4-((3R,5S)-3,5-dimethylpiperazin-l-yl)but- 2-enoyl chloride in the place of 6 in Step 3. MS 512.3 (M+Na). EXAMPLE 171
[001018] Preparation of l-(3-(5-methyl-2-(3-aminosulfonylphenylamino)pyrimidin-4- ylamino)phenyl)-3 -methylbut-2-en- 1 -one 1-224
Figure imgf000341_0001
1-224
[001019] The title compound was prepared according to the schemes, steps and intermediates described in Example 112 using 2,4-dichloro-5-methylpyrimine in place of 1 in step-1 and 3-aminobenzenesulfonamide in place of 4 in step-2. 1H NMR (DMSO-dβ) δ ppm: 1.97 (s, 3H), 2.14 (s, 6H), 6.88 (s, IH), 7.25-7.30 (m, 4H), 7.47 (t, J= 7.92 Hz, IH), 7.62 (d, J= 7.72 Hz. IH), 7.96 (s, IH), 8.0-8.07 (m, 3H), 8.20 (t, J = 7.36 Hz, IH), 8.55 (s, IH), 9.37 (s, IH); LCMS : m/e 438 (M+l).
EXAMPLE 172
[001020] Preparation of N-(3-acrylamidophenyl)-N-(5-cyano-2-(6-methoxypyridin-3- ylamino)pyrimidin-4-yl)acrylamide 1-171
Figure imgf000341_0002
1-171
[001021] The title compound was prepared according to the schemes, steps and intermediates described in Example 95 using excess acroyl chloride in step-4. MS m/z: 442.1 (M+H +). EXAMPLE 173
[001022] Preparation of N-3-(N-methyl-N-(5-fiuoro-2-(4-methyl-3,4-dihydro-2H- benzo[b][l,4]oxazin-6-ylamino)pyrimidin-4-yl)aminophenylacrylamide 1-127
Figure imgf000342_0001
1-127
[001023] The title compound was prepared by treating the product of Example 88 with excess formaldehyde and NaBH3CN (2 equiv.) in acetonitrile and acetic acid (4:1). MS m/z: 435.1 (M+H +).
EXAMPLE 174
[001024] Preparation of N-(3-(5-methyl-2-(phenylamino)pyrimidin-4- ylamino)benzyl)acrylamide 1-205
Figure imgf000342_0002
1-205
[001025] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 using 2,4-dichloro-5-methylpyrimidine in place of 1 and 3-(tert-butoxycarbonylamino)methylaniline in place of 2 in step-1, and aniline in place of 4 on step-2. 1H-NMR (CDCl3, 500 MHz): δ 7.91 (s, IH), 7.77 (s, IH), 7.57 (d, J = 9.0 Hz, 2H), 7.41 (d, J = 8.0 Hz, IH), 7.36 - 7.26 (m, 2H), 7.13 - 6.96 (m, 4H), 6.36 - 6.25 (m, 2H), 5.97 (dd, J = 10.5, 17.0 Hz, IH), 5.78 (bs, IH), 5.63 (d, J = 10.5 Hz, IH), 4.51 (d, J = 6.0 Hz, 2H), 2.12 (s, 3H). MS: m/e = 360 (M++l). EXAMPLE 175
[001026] Preparation of (E)-3-(5-methyl-2-(phenylamino) pyrimidin-4-ylamino) benzyl but-2-enoate 1-246
Figure imgf000343_0001
1-246
[001027] The title compound was prepared according to the schemes, steps, and intermediates described below.
Figure imgf000343_0002
I-246
A) 2, Xanthophos, Pd2(dba)3, Cs2CO3, CH3CN, 90 0C, 12 hr; B) 4, t-BuOH, 90 0C, 4 hr; C) 6, TEA, DCM , -30 0C, 5 min [001028] Step-1
Figure imgf000343_0003
3 [001029] To a stirred solution of 1 (0.34 g, 2.08 mmol) in acetonitrile (5 rnL) were added
Cs2CO3 (1.09 g, 3.35 mmol), Xanthophos (0.024 g, 0.041 mmol), Pd2(dba)3 (38 mg, 0.04 mmol) and 2 (0.5 g, 2.1 mmol) at RT under N2 atmosphere. Argon gas was purged in to the reaction mixture for 1 h and stirred at 90 0C for 12 h. The progress of the reaction was monitored by TLC. The reaction mixture was filtered through a pad of celite, and the filtrate was concentrated under reduced pressure. The crude compound was purified by silica gel column chromatography to afford 3 (0.56 g, 29.16%) as light yellow liquid. 1H-NMR (CDCl3, 500 MHz): δ 8.0 (s, IH), 7.55 (s, IH), 7.51 (d, J = 8.0 Hz, IH), (t, J = 7.5 Hz, IH), (d, J = 7.5 Hz, IH), 6.48 (s, IH), 4.76 (s, 2H), 2.18 (s, 3H), 0.95 (s, 9H), 0.10 (s, 6H). MS: m/e = 364 [M++l]. [001030] Step-2
Figure imgf000344_0001
[001031] To a stirred solution of 3 (0.07 g, 0.19 mmol) in t-BuOH (1.5 mL) was added aniline (4) (0.018 g, 0.19 mmol) at room temperature. The reaction mixture was heated up to 90 0C and stirred for 4 h at the same temperature. The progress of the reaction was monitored by TLC. After the completion of starting materials, the volatiles were removed under reduced pressure to give 5 (0.033 g, 55.9%) as light yellow solid. 1H-NMR (DMSO-d6, 500 MHz): δ 10.45 (s, IH), 9.82 (s, IH), 7.91 (s, IH), 7.58-7.01 (m, 9H), 4.50 (s, 2H), 2.16 (s, 3H). MS: m/e = 307 [M++l]. [001032] Step-3
Figure imgf000344_0002
1-246
[001033] To a stirred solution of 5 (0.5 g, 1.63 mmol) in DCM (5 mL) was added 6 (0.18 g,
1.72 mmol) followed by TEA (0.66 mL, 4.78 mmol) at -30 0C under N2 atmosphere. The reaction mixture was stirred for 5 minutes at -30 0C and the progress of the reaction was monitored by TLC. After the completion of reaction, quenched with water and extracted with DCM (2 x 50 mL). The organic layer was separated, dried over anhydrous Na2SO4 and concentrated under reduced pressure. The crude material was purified by silica gel column chromatography to give 50 mg of an isomeric mixture of the title compound. This mixture in DCM (2 mL) was treated with DBU (0.02 g, 0.127 mmol) at room temperature. The reaction mixture was stirred for 2 h at room temperature, quenched with water and extracted with DCM (2 x 10 mL). The organic layer was separated, dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford 1-246 (0.05 g, 10%) as light yellow solid. 1H-NMR (CDCl3, 500 MHz): δ 7.87 (s, IH), 7.65 (s, IH), 7.58-7.51 (m, 3H), 7.34 (t, J= 7.5 Hz, IH), 7.30-7.23 (m, 3H), 7.13 (d, J = 7.5 Hz, IH), 7.08-6.96 (m, 2H), 6.40 (s, IH), 5.87 (dd, J = 1.5, 15.5 Hz, IH), 5.16 (s, 2H), 2.13 (s, 3H), 1.87 (dd, J= 2.0, 7.0 Hz, 3H). 13C-NMR (CDCl3, 125 MHz): δ 166.3, 159.1, 158.4, 155.4, 145.3, 139.9, 138.9, 137.0, 128.9, 128.7, 123.2, 122.4, 121.9, 121.2, 121.0, 119.3, 105.2, 65.7, 17.9, 13.2. MS: m/e = 375 [M++l].
EXAMPLE 176
[001034] Preparation of (E)-4-(5-methyl-2-(phenylamino) pyrimidin-4-ylamino) benzyl but-2-enoate 1-60
Figure imgf000345_0001
1-60
[001035] The title compound was prepared according to the schemes, steps and intermediates described in Example 175 using 4-((tert-butyldimethylsilyloxy) methyl) aniline in place of 2 in step-1. 1H-NMR (CDCl3, 500 MHz): δ 8.19 (bs, IH), 7.81 (s, IH), 7.56 (d, J = 8.5 Hz, 2H), 7.53 (d, J= 7.5 Hz, 2H), 7.42-7.36 (m, 3H), 7.28-7.22 (m, IH), 7.08-6.98 (m, 2H), 6.54 (s, IH), 5.89 (dd, J = 12.5, 14.0 Hz, IH), 5.17 (s, 2H), 2.15 (s, 3H), 1.89 (dd, J = 1.5, 7.0 Hz, 3H). MS: m/e = 375 [M++!]. EXAMPLE 177
[001036] Preparation of N-methyl-N-(3-(5-methyl-2-(phenylamino)pyrimidin-4- ylamino)benzyl)acrylamide 1-220
Figure imgf000346_0001
1-220
[001037] The title compound was prepared according to the schemes, steps, and intermediates described below.
Figure imgf000346_0002
1-220
A) 2, Xanthophos, Pd2(dba)3, Cs2CO3, CH3CN, 100 0C, 12 hr; B) 4, t-BuOH, 90 0C, 4 hr; C) 10 N HCl, DCM , rt, 30 min: D) 7, TEA, DCM, -10 0C, 10 min. [001038] Step-1
Figure imgf000347_0001
[001039] To a stirred solution of 1 (2.6 g, 15.7 mmol) in acetonitrile (26.7 rnL) was added 2
(2.67 g, 11.3 mmol), Pd2 (dba)3 (0.31 g, 0.33 mmol), Xanthophos (0.52 g, 0.89 mmol) and Cs2CO3 (6.6 g, 20.0 mmol). The reaction mixture was then degassed by purging argon for Ih and further heated to 100 0C for 12 h. After the completion of the reaction (monitored by TLC), the reaction mixture was filtered through celite bed and the filtrate was concentrated under reduced pressure. The resulting crude material was purified by column chromatography (60-120 mesh silica gel; 20% ethyl acetate/Hexane) to afford 3 (2.32 g, 56.71%) as light brown solid. 1H-NMR (CDCl3, 500 MHz): 8.01 (s, IH), 7.56 (d, J = 7.5 Hz, IH), 7.43 (s, IH), 7.35 (t, J = 7.0 Hz, IH), 7.05 (s, IH), 6.80 (bs, IH), 4.45 (s, 2H), 2.87 (s, 3H), 2.30 (s, 3H), 1.48 (s, 9H). [001040] Step-2
Figure imgf000347_0002
[001041] To a stirred solution of 3 (2.32 g, 6.0 mmol) in t-BuOH (11.6 mL) was added 4
(0.65 g, 6.9 mmol) at RT and the reaction mixture was further heated at reflux for 48 h. The progress of the reaction was monitored by TLC. After the completion of the reaction, t-BuOH was concentrated under reduced pressure to dryness to give 5 (2.3 g, 85.82%) as light yellow solid. 1H-NMR (DMSO-J6, 500 MHz): δ 10.32 (s, IH), 9.78 (s, IH), 7.91 (s, IH), 7.50 (d, J = 8.0 Hz, IH), 7.45 - 7.30 (m, 4H), 7.24 (t, J = 7.0 Hz, 2H), 7.15 - 7.06 (m, 2H), 4.36 (s, 2H), 2.72 (s, 3H), 2.17 (s, 3H), 1.41, 1.34 (two s, 9H). MS: m/e = 420 (M++l). [001042] Step-3
Figure imgf000348_0001
[001043] To a stirred solution of 5 (0.05 mg, 0.11 mmol) in DCM (5 niL) was added 37%
HCl (1.0 rnL) and stirred at RT for 30 min. After the completion of the reaction (monitored by TLC), volatiles were removed under reduced pressure. The aqueous layer was cooled to 0 0C, basifϊed up to pH ~ 8-9 with 10% NaOH solution and extracted with DCM (50 mL). The organic portion was separated, washed with water, brine, dried over anhydrous Na2SO4 and evaporated under reduced pressure to afford 6 (0.015 g, 48.36%) as light yellow solid. 1H-NMR (CDCl3, 500 MHz): δ 7.95 - 7.85 (m, 2H), 7.68 -7.60 (m, 3H), 7.42 (d, J = 8.0 Hz, IH), 7.32 - 7.20 (m, 4H), 7.05 (d, J = 7.5 Hz, IH), 7.00 - 6.95 (m, IH), 6.40 (s, IH), 3.84 (s, 2H), 2.48 (s, 3H), 2.06 (s, 3H). MS: m/e = 320 (M++l). [001044] Step-4
Figure imgf000348_0002
1-220
[001045] To a stirred solution of 6 (0.3 g, 0.94 mmol) in DCM (12 mL) was added TEA
(0.10 g, 0.99 mmol) and 7 (0.08 g, 0.88 mmol) dropwise over a period of 5 min at -10 0C under inert atmosphere. The reaction mixture was then stirred at -10 0C for 5-10 min. After the completion of the reaction (monitored by TLC), the reaction mixture was quenched with cold water (5 mL) and extracted with DCM (2 x 50 mL). The DCM layer was washed with water, brine, dried over anhydrous Na2SO4 and concentrated under reduced pressure. The crude material was purified by column chromatography (60-120 mesh silica gel; 30% Ethyl acetate/Hexane) to afford 1-220 (0.15 g, 42.85%) as off white solid. 1H-NMR (DMSO-J6, 500 MHz, at 8O0C): δ 8.48 (bs, IH), 8.07 (s, IH), 7.88 (s, IH), 7.69 - 7.58 (m, 4H), 7.28 (t, J = 8.0 Hz, IH), 7.16 (t, J = 8.0 Hz, 2H), 6.91 - 6.85 (m, 2H), 6.75 (dd, J = 2.5, 15.3 Hz, IH), 6.13 (d, J = 15.0 Hz, IH), 5.66 (d, J = 7.5 Hz, IH), 4.60 (s, 2H), 2.95 (s, 3H), 2.12 (s, 3H). MS: m/e = 374 (M++l).
EXAMPLE 178
[001046] Preparation of N-methyl-N-(4-(5-methyl-2-(phenylamino)pyrimidin-4- ylamino)benzyl)acrylamide 1-219
Figure imgf000349_0001
1-219
[001047] The title compound was prepared according to the schemes, steps and intermediates described in Example 177 using tert-butyl-4-aminobenzyl(methyl)-carbamate in place of 2 in step-1. 1H NMR (DMSO-J6, 500 MHz at 8O0C): δ 8.55 (s, IH), 8.03 (s, IH), 7.86 (s, IH), 7.65 (d, J = 8.0 Hz, 2H), 7.62 (d, J = 8.0 Hz, 2H), 7.20 - 7.13 (m, 4H), 6.86 - 6.75 (m, 2H), 6.14 (dd, J= 2.5, 17.0 Hz, IH), 5.67 (d, J= 15.0 Hz, IH), 4.58 (s, 2H), 2.96 (s, 3H), 2.10 (s, 3H). MS: m/e = 374 (M++l).
EXAMPLE 179
[001048] Preparation of N-(5-(5-acetyl-4-(4-acryloyl-3,4-dihydro-2H-benzo[b][l,4]oxazin-
6-ylamino)pyrimidin-2-ylamino)pyridin-2-yl)-2,2,2-trifluoro-N-methylacetamide 1-142
Figure imgf000349_0002
1-142 [001049] The title compound was prepared according to the schemes, steps and intermediates described in Example 42 using 5-amino-2(2,2,2-trifluoroacetamido)pyridine in place of 9 in step-5. LC-MS: m/z 542.2 (ES+), 540.2.2 (ES-).
EXAMPLE 180
[001050] Preparation of l-(6-(5-fluoro-2-(4-(2-methoxyethoxy)phenylamino)pyrimidin-4- ylamino)indolin- 1 -yl)prop-2-en- 1 -one 1-94
Figure imgf000350_0001
1-94
[001051] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 using N-Boc-6-aminoindoline in place of 2 in step-1. LC- MS: m/z 450.1 (ES+), 448.1 (ES-).
EXAMPLE 181
[001052] Preparation of N-(3-(5-fluoro-2-(6-(2-methoxyethoxy)pyridine-3- ylamino)pyrimidin-4-ylamino)phenyl)acrylamide I- 103
Figure imgf000350_0002
1-103
[001053] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 3-amino-6-(2-methoxyethoxy)pyridine in place of 4 in Step-2. 1H NMR (CDCl3 + trace of DMSO-d6) δ ppm: 3.44 (s, 3H), 3.75 (t, J = 4.4 Hz, 2H), 4.43 (t, J= 4.4 Hz, 2H), 5.81 (dd, J= 1.8 & 9.6 Hz, IH), 6.45 (m, IH), 6.80 (m, 3H), 7.17 (m, IH), 7.29 (m, IH), 7.43 (m, IH), 7.49 (m, IH), 7.60 (m, IH), 7.80 (dd, J = 2.8 & 9.2 Hz, IH), 7.94 (d, J = 3.1 Hz, IH), 8.14 (s, IH), 8.32 (d, J= 2.9 Hz, IH); LCMS : m/e 425.1 (M+l).
EXAMPLE 182
[001054] Preparation of N-(3-(5-fluoro-2-(4-(3- methylsulfonylpropoxy)phenyl)aminopyrimidin-4-ylamino)phenyl)acrylamide 1-97
Figure imgf000351_0001
1-97
[001055] The title compound was prepared as a TFA salt according to the schemes, steps and intermediates described in Example 20 by using 4-(3-methylsulfonylpropoxy)aniline in place of 4 in Step-2. 1H NMR (CDCl3 + trace of DMSO-d6) δ ppm: 1.95 (m, 2H), 2.67 (s, 3H), 2.98 (m, 5H), 3.74 (t, J = 6.0 Hz, 2H), 5.45 (dd, J = 4.1 & 7.3 Hz, IH), 6.07 (m, 2H), 6.48 (d, J = 8.2 Hz, IH), 6.77 (m, 4H), 7.09 (d, J= 7.4 Hz, IH), 7.51 (d, J= 4.1 Hz, IH), 7.70 (br, IH); LCMS : m/e 486.1 (M+l).
EXAMPLE 183
[001056] Preparation of N-(3-(5-fluoro-2-(6-(trideutereomethoxy)pyridine-3- ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-95
Figure imgf000351_0002
1-95
[001057] The title compound was prepared as a TFA salt according to the schemes, steps and intermediates described in Example 20 by using 6-(trideuteriomethoxy)pyridin-3 -amine in place of 4 in Step-2. 1H NMR (CDCl3 + trace of DMSO-d6) δ ppm: 5.78 (dd, J = 3.7 & 7.8 Hz, IH), 6.40 (m, 2H), 6.71 (d, J = 8.7 Hz, IH), 7.3 (m, 3H), 7.75 (dd, J = 2.1 SL 8.7 Hz, IH), 7.82 (d, J = 4.6 Hz, IH), 7.95 (s, IH), 8.33 (d, J = 2.3 Hz, IH); LCMS : m/e 384.1 (M+l). [001058] The intermediate 6-(trideutratedmethoxy)pyridin-3 -amine was prepared by the scheme shown below.
Figure imgf000352_0001
A) NaH, CD3OD, rt; BH3 NMe3, Pd(OH)2
[001059] Step l
Figure imgf000352_0002
[001060] To NaH (60%, 0.30 g) in 5 mL of CD3OD at 0 0C was added 2-chloro-5- nitropyridine (1.0 g). The mixture was stirred at rt overnight. To this mixture were added BH3-NMe3 (550 mg) and Pd(OH)2 (100 mg). The resulting mixture was refluxed for 2 h. After cooling down, the mixture was concentrated and purified using silica gel chromatography to give the desired 6-(trideuteratedmethoxy)pyridin-3-amine (130 mg). 1H NMR (CDCl3) δ ppm: 3.30 (br, 2H), 6.60 (d, J = 8.7 Hz, IH), 7.03 (dd, J = 3.2 & 8.7 Hz, IH), 7.66 (d, J = 3.2 Hz, IH).
EXAMPLE 184
[001061] Preparation of N-(3-(5-fluoro-2(3,4,5-trimethoxyphenylamino)pyrimidin-4- yloxy)phenyl)acrylamide 1-148
Figure imgf000352_0003
1-148
[001062] The title compound was prepared according to the schemes, steps and intermediates described in Example 98 by using 3,4,5-trimethoxyaniline in place of 4 in Step-2. MS: m/e 441 [M+l]. EXAMPLE 185
[001063] Preparation of 3 -methyl- 1 -(3 -(5 -methyl-2-(phenylamino)pyrimidin-4- ylamino)phenyl)but-2-en- 1 -one 1-232
Figure imgf000353_0001
1-232
[001064] The title compound was prepared according to the schemes, steps and intermediates described in Example 112 using 2,4-dichloro-5-methylpyrimidine in place of 1 in step-1 and aniline in place of 4 in step-2. 1H NMR (CDCl3) δ ppm: 1.97 (s, 3H), 2.15 (s, 3H), 2.22 (s, 3H), 6.47 (s, IH), 6.71 (s, IH), 6.97 (t, J = 9.8 Hz, IH), 7.17 (s, IH), 7.24 (t, J = 10.36 Hz, IH), 7.27 (s, IH), 7.44 (t, J = 10.64 Hz, IH), 7.53 (d, J= 10.48 Hz, 2H), 7.68 (d, J = 10.28 Hz, IH), 7.94 (d, J= 10 Hz, IH), 7.98 (s, IH); LCMS : m/e 359 (M+l).
EXAMPLE 186
[001065] Preparation of l-(3-(5-methyl-2-phenylamino)pyrimidin-4-ylamino(piperidin-l- yl)prop-2-en-one 1-27
Figure imgf000353_0002
1-27
[001066] The title compound was prepared according to the schemes, steps and intermediates described in Example 1 using l-tert-butoxycarbonyl-3-aminopiperidine in place of 1 in step-1. 1H NMR (DMSO-d6) δ ppm: 1.30-1.50 (m, IH), 1.55-1.75 (m, IH), 1.75-1.90 (m, IH), 1.92 (s, 3H), 1.95-2.05 (m, IH), 2.75-3.31 (m, 2H), 3.99-4.09 (m, 2H), 4.10-4.15 & 4.40- 4.47 (m, IH), 5.49 & 5.70 (d, J = 10.8 Hz & d, J = 9.2 Hz respectively, together IH), 6.02 & 6.13 (d, J= 17.6 Hz & d, J= 16.8 Hz respectively, together IH), 6.25-6.40 (m, IH), 6.63 & 6.80- 6.90 (dd, J= 10.8, 16.8 Hz & m respectively, together IH), 6.75-6.85 (m, IH), 7.15 (t, J= 8 Hz, 2H), 7.69 (bs, 3H), 8.81 (s, IH); LCMS: m/e 337.8 (M+ 1).
EXAMPLE 187
[001067] Preparation of 3-(4-(2-acryloyl-l,2,3,4-tetrahydroisoquinolin-6-ylamino)-5- methylpyrimidin-2-ylamino)benzenesulfonamide 1-40
Figure imgf000354_0001
1-40
[001068] The title compound was prepared according to the schemes, steps and intermediates described in Example 1 using 6-amino-2-tert-butoxycarbonyl-l,2,3,4- tetrahydroisoquinoline in place of 1 in step-1. 1H NMR (DMSO-d6) δ ppm: 2.10 (s, 3H), 2.80- 2.83 (m, 2H), 3.75-3.90 (m, 2H), 4.66 (s, IH), 4.76 (s, IH), 5.71-5.74 (m, IH), 6.16 (dd, J = 2.32 & 16.76 Hz, IH), 6.87-6.91 (m, IH), 7.13-7.18 (m, IH), 7.25-7.31 (m, 4H), 7.53-7.57 (m, 2H), 7.90 (s, IH), 8.05 (s, 2H), 8.28 (s, IH), 9.31 (s, IH); LCMS: m/e 464.8 (M+l).
EXAMPLE 188
[001069] Preparation of (S)-N-(3-(5-fluoro-2-(tetrahydrofuran-3-yloxy)pyridine-3- ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-54
Figure imgf000354_0002
1-54 [001070] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 using (S)-3-amino-6-(tetrahydrofuran-3-yloxy)pyridine in place of 4 in step-2. MS: m/e = 437 [M+ 1].
EXAMPLE 189
[001071] Preparation of N-(3-(5-trifluoromethyl-2-(phenylamino)pyrimidin-4- ylamino)phenyl)acrylamide 1-245
Figure imgf000355_0001
1-245
[001072] The title compound was prepared according to the schemes, steps, and intermediates described below.
Figure imgf000356_0001
A) 2, ZnCl2, DCE, t-BuOH (1 :1) , O 0C, 30 min; B) 4, DMF, DIEPA, 70 0C, 16 hr; C) TFA, DCM, rt, 1 hr; D) 7, TEA, DCM. [001073] Step-1
Figure imgf000356_0002
[001074] To a cold (0 0C) solution of 1 (2 g, 9.2 mmol) in 80 mL of a 1 :1 mixture of tBuOH/DCE was added zinc chloride (11 mL of a 1 M solution in ether, 1.2 eq). After one hour, 2 (0.858g, 9.2 mmol) was added followed by dropwise addition of triethylamine (1.03 g; 1.1 eq) in 10 mL of DCE/t-BuOH. After stirring for 30 minutes, the solvents were removed under reduced pressure and the residue was dissolved in ethyl acetate (50 mL) and washed with brine (10 mL). The organic layer was dried over sodium sulphate, filtered and concentrated in vacuo. The desired product 3 was obtained as a white solid following recrystalization from EtOAc/Hexane (1 :9), (2g, 80%). [001075] Step-2
Figure imgf000357_0001
[001076] To a solution of 3 (0.5 g, 1.82 mmol) and 4 (0.38 g, 1.83 mmol) in DMF (10 mL) was added DIPEA (0.283 g, 2.192 mmol) and the mixture was heated to 60 0C under an argon atmosphere for 16 h. The solvent was distilled off and the residue was dissolved in ethyl acetate (50 mL) and washed with brine (10 mL). The organic layer was dried over sodium sulphate, filtered and concentrated in vacuo. The crude mixture was purified by flash column chromatography (eluent: EtOAc/hexane 1 :1) to afford 5 as a white solid (0.48 g, 60%). [001077] Step-3
Figure imgf000357_0002
[001078] To a solution of 6 (0.25 g, 0.63 mmol) in CH2Cl2 (10 mL) was added trifluoroacetic acid (2 mL) and the mixture was stirred at room temperature for 1 hour. Solvents were removed under reduced pressure and the residue was dissolved in CH2Cl2, washed with 10% aqueous NaHCO3 solution, dried (Na2SO4), filtered, and evaporated under reduced pressure to provide the free amine as white solid.
[001079] Step-4
Figure imgf000357_0003
[001080] To a stirred solution of 6 (0.2 g) in DCM (20 rnL) under argon atmosphere cooled to -40 0C was added triethylamine followed by dropwise addition of 7 (0.069 g, 0.686 mmol). The resulting mixture was stirred at -40 0C for 10 min. The reaction mixture was diluted with DCM (50 mL) and washed with brine (10 mL). The organic layer was dried over sodium sulfate, filtered, and evaporated under reduced pressure. The residue was purified by flash chromatography on silica gel using (MeOH- EtOAc 5:95) as eluent to provide the target compound 1-245. 1H NMR (200 MHz, CD3OD) δ 8.25 (s, IH), 7.80 (s, IH), 7.60-7.05 (m, 7H), 6.90 (m, IH), 6.35 (m, 2H), 5.75 (dd, J= 8.0, 2.0 Hz, IH).
EXAMPLE 190
[001081] Preparation of N-(3-(5-trifluoromethyl-2-(3-methoxyphenylamino)pyrimidin-4- ylamino)phenyl)acrylamide 1-242
Figure imgf000358_0001
1-242
[001082] The title compound was prepared according to the schemes, steps and intermediates described in Example 189 using 3 -methoxy aniline in place of 2 in step-1. 1H NMR (200 MHz, CD3OD) δ 8.31 (s, IH), 7.84 (s, IH), 7.59 (m, IH), 7.37-7.09 (m, 5H) 6.53 (m, IH), 6.41 (m, 2H), 5.79 (dd, J= 8.0, 2.0, Hz, IH), 3.66 (s, 3H).
EXAMPLE 191
[001083] Preparation of N-(4-(5-trifluoromethyl-2-(3-methoxyphenylamino)pyrimidin-4- ylamino)phenyl)acrylamide 1-236
Figure imgf000358_0002
1-236 [001084] The title compound was prepared according to the schemes, steps and intermediates described in Example 189 using 3 -methoxy aniline in place of 2 in step-1 and A- amino-N-tert-butoxycarbonylaniline in place of 4 in step-2. 1H NMR (200 MHz, CD3OD) δ 8.27 (s, IH), 7.70 (d, J = 6.0Hz) IH), 7.46 (d, J = 6.0Hz, IH), 7.09 (brs, IH), 7.07 (m, 2H) 6.51 (m, IH), 6.44 (m, 2H), 5.80 (dd, J= 8.0, 2.0 Hz, IH), 3.56 (s, 3H).
EXAMPLE 192
[001085] Preparation of N-(4-(5-trifluoromethyl-2-(3-methoxyphenylamino)pyrimidin-4- ylamino)phenyl)methylacrylamide 1-235
Figure imgf000359_0001
1-235
[001086] The title compound was prepared according to the schemes, steps and intermediates described in Example 189 using 3 -methoxy aniline in place of 2 in step-1 and A- aminophenylmethyl-N-tert-butoxycarbonylamine in place of 4 in step-2. 1H NMR (200 MHz, CD3OD) δ 8.30 (s, IH), 7.49 (d, J= 8.0 Hz, IH), 7.37 (d, J= 8.0 Hz, IH), 7.10 (m, 3H), 6.60 (m, IH) 6.34 (m, 2H), 5.75 (dd, J= 8.0, 2.0 Hz, IH), 4.51(s, 2H), 3.68 (s, 3H).
EXAMPLE 193
[001087] Preparation of N-(4-chloro-3-(5-trifluoromethyl-2-(3- methoxyphenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-227
Figure imgf000360_0001
1-227
[001088] The title compound was prepared according to the schemes, steps and intermediates described in Example 189 using 3 -methoxy aniline in place of 2 in step-1 and N- tert-butoxycarbony-3-amino-6-chloroaniline in place of 4 in step-2. 1H NMR (200 MHz, CD3OD) δ 8.33 (s, IH), δ 8.08 (s, IH), 7.45 (m, 2H), 7.21-7.07 (m, 3H), 6.60-6.36 (m, 3H), 5.84 (dd, J= 8.0, 2.0 Hz, IH), 3.71 (s, 3H).
EXAMPLE 194
[001089] Preparation of N-(3-(5-trifluoromethyl-2-(3-methoxyphenylamino)pyrimidin-4- ylamino)phenyl)methylacrylamide 1-226
Figure imgf000360_0002
1-226
[001090] The title compound was prepared according to the schemes, steps and intermediates described in Example 189 using 3 -methoxy aniline in place of 2 in step-1 and 3- aminophenylmethyl-N-tert-butoxycarbonylamine in place of 4 in step-2. 1H NMR (200 MHz, CD3OD) δ 8.31 (s, IH), 7.71-7.33 (m, 3H), 7.21-7.08 (m, 4H), 6.57 (m, IH), 6.26 (d, J = 4Hz, 2H), 5.69 (dd, J= 8.0, 2.0 Hz, IH), 4.47 (s, 2H), 3.67 (s, 3H).
EXAMPLE 195
[001091] Preparation of N-(4-(5-trifluoromethyl-2-(6-methoxypyridin-3- ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-218
Figure imgf000361_0001
1-218
[001092] The title compound was prepared according to the schemes, steps and intermediates described in Example 189 using 3-amino-6-methoxypyridine in place of 2 in step- 1. 1H NMR (200 MHz, CD3OD) δ 8.21 (s, IH), 7.90-7.78 (m, 3H), 7.48 (m, 2H), 7.30 (m, 2H), 6.40 (m, 2H), 5.75 (dd, J= 8.0, 2.0 Hz, IH), 3.81 (s, 3H).
EXAMPLE 196
[001093] Preparation of N-(4-(5-trifluoromethyl-2-(5-methoxypyridin-3- ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-214
Figure imgf000361_0002
1-214
[001094] The title compound was prepared according to the schemes, steps and intermediates described in Example 189 using 3-amino-5-methoxypyridine in place of 2 in step-1 and 4-amino-N-tert-butoxycarbonylaniline in place of 4 in step-2. MS: m/e = 431 [M+ 1]. EXAMPLE 197
[001095] Preparation of l-(3-(5-trifluoromethyl-2-(3-methoxyphenylamino)pyrimidin-4- ylamino)phenyl)-3-methyl-but-2-en- 1 -one 1-225
Figure imgf000362_0001
1-225
[001096] The title compound was prepared according to the schemes, steps and intermediates described in Example 189 using 3 -methoxy aniline in place of 2 in step-1 and l-(3- aminophenyl)-3-methylbut-2-en-l-one in place of 4 in step-2. 1H NMR (200 MHz, CDCl3) δ 8.33 (s, IH), δ 8.38 (s, IH), 7.99-7.77 (m, 4H), 7.50 (m, 2H), 7.20-7.01 (m, 4H), 6.68-6.60(m, 2H), 3.68 (s, 3H), 2.25 (s, 3H), 1.99 (s, 3H).
[001097] l-(3-Aminophenyl)-3-methylbut-2-en-l-one was prepared according to the scheme, steps, and intermediates described below.
Figure imgf000362_0002
A) BoC2O, NEt3, DMAP, DCM; B) NHMe(OMe)-HCl, TBTU, DCM, 0 0C to rt; C) 4, THF, 0 0C to rt; D) TFA, DCM. [001098] Step-1
Figure imgf000362_0003
[001099] Di-tert-butyldicarbonate (6.54 g, 30 mmol, 1.5 eq.) was added to a solution of 1
(2.70 g, 20 mmol) in CH2Cl2 (100 mL) containing Et3N (3.4 mL, 24 mmol, 1.2 eq.) and DMAP (122 mg, 1.0 mmol, 5 mol%). The mixture was stirred overnight under a CaCl2 drying tube. The solvents were evaporated and the residue was partitioned between ether (50 mL) and water (50 mL). The aqueous phase was extracted with ether then acidified to pH 3 with IN HCl and extracted with EtOAc (2 X 50 mL). The combined organic layers were washed with water and brine and dried over NaSO4. Concentration afforded the crude product which was recrystallized from EtOAc/hexanes to give 2 (2.92 g, 62%). [001100] Step-2
Figure imgf000363_0001
[001101] To a mixture of 2 (1.5 g, 6.33 mmol), N-methoxy-N-methylamine hydrochloride
(614 mg, 6.33 mmol), and TBTU (2.05 g, 6.33 mmol) in DCM (30 mL) at 0 0C was added NEt3 (2.7 mL, 19 mmol, 3 eq.). The mixture was stirred for 30 min at 0 0C then at room temperature for 2 h whereupon HPLC analysis indicated the reaction to be complete. The reaction mixture was poured into 150 mL of cold water and the product precipitated as a white solid which was collected and washed with cold water. The product was dried in a vacuum oven overnight to give 3 (1.34 g, 75%) as a white solid. [001102] Step-3
Figure imgf000363_0002
[001103] To a solution of 3 (546 mg, 1.95 mmol) in THF (3 mL) at 0 0C under Ar was added dropwise 4 (0.5 M in THF, 9.75 mL, 4.9 mmol, 2.5 eq.). The reaction mixture was stirred at 0 oC for 30 min then the cooling bath was removed and the reaction was stirred at rt for 2 h. The reaction mixture was cooled to 0 0C and quenched with 5% citric acid solution. After dilution with water (10 mL) the aqueous phase was extracted with ether (2 X 15 mL) and the combined organic layers were washed with water and brine and dried over NaSO4. Concentration afforded 5 (82%) as a yellow solid which was sufficiently pure to be used directly in the next step. [001104] Step-4
Figure imgf000364_0001
[001105] A sample of 400 mg of 5 was treated with 5 rnL 1 :3 CH2Cl2 : trifluoroacetic acid and the resulting solution stirred at room temperature for 15 minutes. The solvents were removed in vacuo and the residue redissolved in CH2Cl2 and re-evaporated three times. The residue was again taken up in CH2Cl2 and the solution washed with saturated sodium bicarbonate solution. The CH2Cl2 layer was dried over sodium sulphate, filtered and evaporated to afford 6 as a white solid that was used directly in the next reaction.
EXAMPLE 198
[001106] Preparation of l-(3-(2-(3 -Methoxy-phenylamino)-5 -trifluoromethyl-pyrimidin-4- ylamino)-cyclohexyl)-3-methyl-but-2-en- 1 -one 1-213
Figure imgf000364_0002
1-213
[001107] The title compound was prepared according to the schemes, steps and intermediates described in Example 189 using 3 -methoxy aniline in place of 2 in step-1 and (d,l)- cώ-l-(3-Amino-cyclohexyl)-3-methyl-but-2-en-l-one in place of 4 in step-2. 1H NMR (200 MHz, CDCl3) δ 8.07 (s, IH), 7.33 (s, IH), 7.19-7.0 (m, 3H), 6.54 (d, J = 2.7 Hz, IH), 6.02 (s, IH), 4.04 (m, IH), 3.75 (s, 3H), 2.50 (m, IH), 2.10 (m, IH), 1.89-1.15 (m, 14H). [001108] (D,L)-cώ-l-(3-Amino-cyclohexyl)-3-methyl-but-2-en-l-one was prepared according to the scheme, steps, and intermediates described below.
Figure imgf000365_0001
A) BoC2O, Na2CO3, acetone, H2O; B) NHMe(OMe)-HCl, TBTU, DCM, 0 0C to rt; C) 4, THF, O 0C to rt; D) TFA, DCM.
[001109] Step-1
Figure imgf000365_0002
2
[001110] To a stirred solution of (±)-l (4.05 g, 28.2 mmol) in water (150 rnL) containing
Na2CO3 (3.0 g, 28.2 mmol) and acetone (100 mL) was added BOC2O (7.4 g, 33.8 mmol, 1.2 eq) and the mixture was stirred at 250C overnight. The acetone was stripped and the aqueous layer was extracted with ether (2X). The aqueous layer was acidified to pH 3 and the precipitated product was collected and washed with water. The product was dried in a vacuum oven overnight to give 2 (5.90 g, 85%) as a white solid.
[001111] Step-2
Figure imgf000365_0003
3
[001112] To a stirred solution of 2 (2.45 g, 10.1 mmol), TBTU (3.44 g, 10.6 mmol, 1.05 eq) and Λ/-methyl-Λ/-methoxyamine hydrochloride (1.03 g, 10.6 mmol, 1.05 eq) in CH2Cl2 (40 mL) at 0 0C was added triethylamine (4.25 mL, 30.3 mmol, 3 eq). The mixture was stirred at 0 0C for 20 min and the bath was removed and stirring was continued for 3 h at 250C. After quenching with water, the CH2Cl2 was stripped and the residue was partitioned between ether and water. The aqueous phase was extracted with ether and the combined organic layers were washed with water and brine and dried over MgSO4. Evaporation of the solvents gave 3 (2.37 g, 82%) as a white solid.
[001113] Step-3
Figure imgf000366_0001
[001114] To a solution of 3 (1.23 g, 4.32 mmol) in THF (20 niL) at 0 0C under argon was added dropwise 4 (27 mL, 0.5 M in THF, 10.8 mmol, 2.5 eq). After the addition was complete the mixture was stirred at 0 0C for 30 min, then at rt for 1 h. The reaction mixture was cooled to 0 0C then quenched with 5% citric acid solution (5 mL). After dilution with water the mixture was extracted with ether (2X) and the combined organic layers were washed with water and brine and dried over NaSO4. Evaporation left an orange residue which was chromatographed on silica gel eluting with 20% EtOAc in hexanes to give 5 (600 mg, 56%) as a light yellow solid.
[001115] Step-4
Figure imgf000366_0002
[001116] A sample of 500 mg of 5 was treated with 6 mL 1 :3 CH2Cl2 : trifluoroacetic acid and the resulting solution stirred at room temperature for 15 minutes. The solvents were removed in vacuo and the residue redissolved in CH2Cl2 and re-evaporated three times. The residue was again taken up in CH2Cl2 and the solution washed with saturated sodium bicarbonate solution. The CH2Cl2 layer was dried over sodium sulphate, filtered and evaporated to afford 6 as a white solid that was used directly without purification.
EXAMPLE 199
[001117] Preparation of l-(5-(5-trifluoromethyl-2-(3-methoxyphenylamino)pyrimidin-4- yl)amino- 1 ,3-dihydroisoindol-2-yl)2-propen- 1 -one 1-132
Figure imgf000367_0001
1-132
[001118] The title compound was prepared according to the schemes, steps and intermediates described in Example 189 using 3 -methoxy aniline in place of 2 in step-1 and 2-(N- tert-butoxycarbonyl)-5-aminoisoindoline in place of 4 in step-2. MS m/e =456 [M+l].
EXAMPLE 200
[001119] Preparation of 3-(2-(2-acryloylisoindolin-5-ylamino)-5-fluoropyrimidin-4- ylamino)benzonitrile 1-106
Figure imgf000367_0002
1-106
[001120] The title compound was prepared according to the schemes, steps and intermediates described in Example 2 using 5-fluoro-2,4-dichloropyrimidine in place of 1 and 3- aminobenzonitrile in place of 2 in step-1 and 2-(tert-butoxycarbonyl-5-aminoisoindoline in place of 4 in step-2. LC/MS (RT = 2.82/(M + H)) 401.1 EXAMPLE 201
[001121] Preparation of N-(3-(5-fluoro-4-((6-(trifluoromethyl)pyridin-3- yl)methylamino)pyrimidin-2-ylamino)phenyl)acrylamide 1-53
Figure imgf000368_0001
1-53
[001122] The title compound was prepared according to the schemes, steps and intermediates described in Example 2 using 5-fluoro-2,4-dichloropyrimidine in place of 1 and 3- aminomethyl-6-trifluoromethylpyridine in place of 2 in step-1. LC/MS (RT = 2.805/(M + H)) 433.0
EXAMPLE 202
[001123] Preparation of N-(3-(4-((2,3-dihydrobenzofuran-5-yl)methylamino)-5- fluoropyrimidin-2-ylamino)phenyl)acrylamide 1-6
Figure imgf000368_0002
1-6
[001124] The title compound was prepared according to the schemes, steps and intermediates described in Example 2 using 5-fluoro-2,4-dichloropyrimidine in place of 1 and 3- aminomethyl-2,3-dihydrobenzofuran in place of 2 in step-1. LC/MS (RT = 2.815/(M + H)) 406.2 EXAMPLE 203
[001125] Preparation of N-(3-(5-fluoro-2-(4-methoxybenzylamino)pyrimidin-4- ylamino)phenyl)acrylamide 1-241
Figure imgf000369_0001
1-241
[001126] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 using 4-methoxybenzylamine in place of 4 in step-2. LC/MS (RT = 2.801/(M + H)) 394.2
EXAMPLE 204
[001127] Preparation of N1-(3-(3-(4-(3-acrylamidophenylamino)-5-methylpyrimidin-2- ylamino)phenoxy)propyl)-N5-(15-oxo-19-((3aR,4R,6aS)-2-oxohexahydro-lH-thieno[3,4- d]imidazol-4-yl)-4,7,10-trioxa-14-azanonadecyl)glutaramide 1-215
Figure imgf000369_0002
1-215 [001128] The title compound was prepared according to the schemes steps and intermediates described below.
Figure imgf000370_0001
A) TFA, DCM; B) N-Biotinyl-NH-(PEG)2-COOH-DIPEA, HOBt, EDC, NMM, DMF. [001129] Step-1
Figure imgf000370_0002
[001130] 1-45 (97 mg, 0.19 mmol; synthesis of 1-45 provided in Example 62) was dissolved in DCM (10 mL). Trifluoroacetic acid (200 μL) was added and allow to stir at rt for 24 hr. The solvent was removed via rotary evaporation to give a tan-brown foam (130mg) which was used without purification in the next reaction. LC/MS (RT = 2.63/(MH+) 419.2) [001131] Step-2
Figure imgf000371_0001
1-215
1 (80 mg, 0.15 mmoL) was dissolved in DMF (2 mL). To the mxiture was added N-Biotinyl- NH-(PEG)2-COOH-DIPEA (114 mg, 0.16 mmol) and HOBt (25 mg, 0.16 mmoL (89%)), and the mixture was cooled in an ice-water bath. EDC (32 mg, 0.16mmoL) was added, followed by N- methylmorpholine (50 μL, 0.45 mmoL). The mixture was allowed to warm to room temperature and continue to stir for 30 min. Direct purification by flash chromatography using 10% gradient of MeOH in DCM gave 40 mg of 1-215 as a yellow film. LC/MS (RT = 2.654/(MH+) 961.3).
EXAMPLE 205
[001132] Preparation of N-(3-(3-(4-(3-acrylamidophenylamino)-5-methylpyrimidin-2- ylamino)phenoxy)propyl)-5-((3aS,4S,6aR)-2-oxohexahydro-lH-thieno[3,4-d]imidazol-4- yl)pentanamide 1-237
Figure imgf000372_0001
1-237
[001133] The title compound was prepared according to the schemes, steps and intermediates described in Example 204 using D-(+)-biotin in place of N-Biotinyl-NH-(PEG)2- COOH-DIPEA in step-2. LC/MS (RT = 2.686/(M + H)) 645.2
EXAMPLE 206
[001134] Preparation of (R)-N-(3-(5-fluoro-2-(3-fluoro-4-(tetrahydrofuran-3- yloxy)phenylamino)pyrimidin-4-ylamino)pheny l)acrylamide 1-316
Figure imgf000372_0002
1-316
[001135] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using (R)-3-fluoro-4-(tetrahydrofuran-3-yloxyaniline in place of 4 in step 2. 1H NMR (DMSO-d6) δ ppm: 1.85-2.00 (m, IH), 2.20 (m, IH), 3.70-3.90 (m, 4H), 4.90 (s, IH), 5.73 (dd, J = 1.56 & 10.04 Hz, IH), 6.23 (dd, J = 1.76 & 17.00 Hz, IH), 6.44 (dd, J = 10.08 & 16.88 Hz, IH), 7.28 (t, J = 8.04 Hz, IH), 7.40-7.47 (m, 2H), 7.67-7.71 (m, 2H), 7.68 (dd, J = 1.96 & 14.08 Hz, IH), 7.92 (s, IH), 8.1 (d, J = 3.64 Hz, IH), 9.21 (s, IH), 9.44 (s, IH), 10.12 (s, IH); LCMS : m/e 452.0 (M-I).
EXAMPLE 207
[001136] Preparation of l-(4-(5-fiuoro-2-(3-fluoro-4-(2- methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-3-methylbut-2-en- 1 -one 1-325
Figure imgf000373_0001
1-325
[001137] The title compound was prepared according to the schemes, steps and intermediates described in Example 112, by using methyl 4-aminobenzoate in place of 2 in step 1 and 3-fluoro-4-(2-methoxyethoxy)aniline in place of 4 in step 2. 1H NMR (DMSO-dβ) δ ppm: 2.01 (s, 3H), 2.15 (d, J = 0.72 Hz, 3H), 3.31 (s, 3H), 3.66 (dd, J = 3.64 & 4.56 Hz, 2H), 4.11 (dd, J = 4.44 & 6.12 Hz, 2H), 6.93 (s, IH), 7.08 (t, J = 9.44 Hz, IH), 7.27 (d, J = 8.88 Hz, IH), 7.74 (dd, J = 2.44 & 14.24 Hz, IH), 7.93 (d, J = 8.96 Hz, 2H), 7.98 (d, J = 8.92 Hz, 2H), 8.20 (d, J = 3.64 Hz, IH), 9.35 (s, IH), 9.73 (s, IH); LCMS : m/e 455 (M+ 1).
EXAMPLE 208
[001138] Preparation of l-(3-(5-fiuoro-2-(3-fluoro-4-(2- methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-3-methylbut-2-en- 1 -one 1-323
Figure imgf000374_0001
1-325
[001139] The title compound was prepared according to the schemes, steps and intermediates described in Example 112, by using 2-(3-fluoro-4-(2-methoxyethoxy)aniline in place of 4 in step 2. 1H NMR (DMSO-d6) δ ppm: 1.95 (s, 3H), 2.14 (s, 3H), 3.31 (s, 3H), 3.63 (t, J = 4.64 Hz, 2H), 4.06 (t, J = 4.36 Hz, 2H), 6.85 (bs, IH), 6.97 (t, J = 9.52 Hz, IH), 7.26 (bd, J = 8.32 Hz, IH), 7.48 (t, J = 7.92 Hz, IH), 7.62-7.67 (m, 2H), 8.08 (bd, J = 7.04 Hz, IH), 8.15- 8.16 (m, 2H), 9.29 (s, IH), 9.58 (s, IH); LCMS : m/e 455 (M+l).
EXAMPLE 209
[001140] Preparation of l-(4-(5-fiuoro-2-(3-fluoro-4-(2- methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-2-methylprop-2-en- 1 -one 1-324
Figure imgf000374_0002
1-324
[001141] The title compound was prepared according to the schemes, steps and intermediates described in Example 112, by using methyl 4-aminobenzoate in place of 2 in step 1, 3-fluoro-4-(2-methoxyethoxy)aniline in place of 4 in step 2 and isopropenylmagnesium bromide in place of 8 in step 5. 1U NMR (DMSO-d6) δ ppm: 2.0 (s, 3H), 3.32 (s, 3H), 3.66 (t, J = 4.36 Hz, 2H), 4.11 (t, J = 4.44 Hz, 2H), 5.55 (s, IH), 5.94 (s, IH), 7.07 (t, J = 9.32 Hz, IH), 7.26 (t, J = 9.4 Hz, IH), 7.72-7.76 (m, 3H), 7.99 (d, J = 8.44 Hz, 2H), 8.21 (d, J = 3.56 Hz, IH), 9.38 (s, IH), 9.75 (s, IH); LCMS : m/e 441.2 (M+ 1).
EXAMPLE 210
[001142] Preparation of l-(4-(5-fluoro-2-(3-fluoro-4-(2- methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-3 -methylbut-3 -en-2-one 1-329
Figure imgf000375_0001
1-329
[001143] The title compound was prepared according to the schemes, steps and intermediates described in Example 112, by using ethyl 4-aminophenylacetate in place of 2 in step 1, 3-fluoro-4-(2-methoxyethoxy)aniline in place of 4 in step 2 and isopropenylmagnesium bromide in place of 8 in step 5. 1H NMR (DMSO-d6) δ ppm: 1.79 (s, 3H), 3.30 (s, 3H), 3.62-3.65 (m, 2H), 4.06 (s, 2H), 4.08-4.10 (m, 2H), 5.95 (d, J = 1 Hz, IH), 6.27 (s, IH), 7.01 (t, J = 9.44 Hz, IH), 7.15 (d, J = 8.52 Hz, 2H), 7.28 (d, J = 8.88 Hz, IH), 7.64-7.70 (m, 3H), 8.08 (d, J = 3.72 Hz, IH), 9.19 (s, IH), 9.33 (s, IH); LCMS : m/e 455.3 (M+l).
EXAMPLE 211
[001144] Preparation of l-(4-(5-fluoro-2-(3-fluoro-4-(2- methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-4-methylpent-3 -en-2-one 1-331
Figure imgf000375_0002
1-331
[001145] The title compound was prepared according to the schemes, steps and intermediates described in Example 112, by using ethyl 4-aminophenylacetate in place of 2 in step 1, 3-fluoro-4-(2-methoxyethoxy)aniline in place of 4 in step 2. 1H NMR (DMSO-dβ) δ ppm: 1.84 (d, J = 1 Hz, 3H), 2.05 (d, J = 0.92 Hz, 3H), 3.30 (s, 3H), 3.62-3.64 (m, 2H), 3.68 (s, 2H), 4.07-4.09 (m, 2H), 6.21 (t, J = 1.2 Hz, IH), 7.01 (t, J = 8.68 Hz, IH), 7.16 (d, J = 8.48 Hz, 2H), 7.26 (d, J = 8.96 Hz, IH), 7.65-7.72 (m, 3H), 8.08 (d, J = 3.72 Hz, IH), 9.20 (s, IH), 9.34 (s, IH); LCMS : m/e 469.3 (M+l).
EXAMPLE 212
[001146] Preparation of l-(3-(5-fiuoro-2-(3-fluoro-4-(2- methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-2-methylprop-2-en- 1 -one 1-322
Figure imgf000376_0001
1-322
[001147] The title compound was prepared according to the schemes, steps and intermediates described in Example 112, by using l,3-fluoro-4-(2-methoxyethoxy)aniline in place of 4 in step 2 and isopropenylmagnesium bromide in place of 8 in step 5. 1H NMR (DMSO-de) δ ppm: 1.96 (s, 3H), 3.30 (s, 3H), 3.63 (t, J = 4.6 Hz, 2H), 4.07 (t, J = 4.36 Hz, 2H), 5.62 (s, IH), 6.00 (s, IH), 6.99 (t, J = 9.24 Hz, IH), 7.26 (d, J = 8.92 Hz, IH), 7.39 (d, J = 7.56 Hz, IH), 7.46 (t, J = 7.72 Hz, IH), 7.62 (bd, J = 14.4 Hz, IH), 7.93 (s, IH), 8.12-8.14 (m, 2H), 9.25 (s, IH), 9.58 (s, IH); LCMS : m/e 441.2 (M+l).
EXAMPLE 213
[001148] Preparation of l-(3-(5-fiuoro-2-(3-fluoro-4-(2- methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-3 -methylbut-3 -en-2-one 1-328
Figure imgf000377_0001
1-328
[001149] The title compound was prepared according to the schemes, steps and intermediates described in Example 112, by using ethyl 3-aminophenylacetate in place of 2 in step 1, 3-fluoro-4-(2-methoxyethoxy)aniline in place of 4 in step 2 and isopropenylmagnesium bromide in place of 8 in step 5. 1H NMR (DMSO-d6) δ ppm: 1.8 (s, 3H), 3.31 (s, 3H), 3.64 (t, J = 4.56 Hz, 2H), 4.06 (s, 2H), 4.09 (t, J = 4.37 Hz, 2H), 5.95 (s, IH), 6.23 (s, IH), 6.92 (d, J = 7.52 Hz, IH), 7.02 (t, J = 9.4 Hz, IH), 7.27 (t, J = 7.8 Hz, 2H), 7.50 (s, IH), 7.66-7.72 (m, 2H), 8.10 (d, J = 3.56 Hz, IH), 9.21 (s, IH), 9.36 (s, IH); LCMS : m/e 455.1 (M+l).
EXAMPLE 214
[001150] Preparation of 2-((3-(5-fluoro-2-(3-fluoro-4-(2- methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)(hydroxy)methyl)acrylonitrile 1-326
Figure imgf000377_0002
1-326
[001151] The title compound was prepared according to the schemes, steps and intermediates described in Example 107, by using 3-fluoro-4-(2-methoxyethoxy)aniline in place of 4 in step 2. 1H NMR (DMS0-d6) δ ppm: 3.30 (s, 3H), 3.62-3.65 (m, 2H), 4.09 (t, J = 4.6 Hz, 2H), 5.29 (d, J = 3.84 Hz, IH), 6.13 (s, IH), 6.19 (s, IH), 6.31 (d, J = 4.04 Hz, IH), 7.03 (t, J = 9.24 Hz, IH), 7.10 (d, J = 7.44 Hz, IH), 7.28 (d, J = 8.72 Hz, IH), 7.36 (t, J = 7.8 Hz, IH), 7.62 (s, IH), 7.66 (dd, J = 2.32 & 14.44 Hz, IH), 7.89 (d, J = 8.2 Hz, IH), 8.10 (d, J = 3.68 Hz, IH), 9.14 (s, IH), 9.45 (s, IH); LCMS : m/e 454 (M+l).
EXAMPLE 215
[001152] Preparation of 2-((4-(5-fiuoro-2-(3-fluoro-4-(2- methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)(hydroxy)methyl)acrylonitrile 1-327
Figure imgf000378_0001
1-327
[001153] The title compound was prepared according to the schemes, steps and intermediates described in Example 107, by using methyl 4-aminobenzoate in place of 2 in step 1 and 3-fluoro-4-(2-methoxyethoxy)aniline in place of 4 in step 2. 1H NMR (DMSO-dβ) δ ppm: 3.30 (s, 3H), 3.64 (dd, J = 2.96 & 4.56 Hz, 2H), 4.08 (t, J = 4.48 Hz, 2H), 5.29 (d, J = 3.8 Hz, IH), 6.11 (s, IH), 6.21 (s, IH), 6.24 (d, J = 4.12 Hz, IH), 7.01 (t, J = 9.4 Hz, IH), 7.29-7.34 (m, 3H), 7.68 (dd, J = 2.2 & 14.16 Hz, IH), 7.77 (d, J = 8.52 Hz, 2H), 8.11 (d, J = 3.68 Hz, IH), 9.23 (s, IH), 9.42 (s, IH); LCMS : m/e 454.0 (M+l).
EXAMPLE 216
[001154] Preparation of N-(3-(2-(4-chloro-3-(2-hydroxy-2-methylpropoxy)phenylamino)-
5 -fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-249
Figure imgf000379_0001
1-249
[001155] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 4-chloro-3-(2-hydroxy-2- methylpropoxy)aniline in place of 4 in step 2. 1H NMR (DMSO-dβ) δ ppm: 1.20 (s, 6H), 3.61 (s, 2H), 4.61 (s, IH), 5.75 (d, J = 11.4 Hz, IH), 6.24 (d, J = 18.36 Hz, IH), 6.44 (dd, J = 10.32 & 17.08 Hz, IH), 7.13 (d, J = 8.64 Hz, IH), 7.28 (t, J = 8 Hz, IH), 7.37-7.44 (m, 3H), 7.55 (d, J = 7.08 Hz, IH), 7.93 (s, IH), 8.12 (d, J = 3.44 Hz, IH), 9.23 (s, IH), 9.47 (s, IH), 10.11 (s, IH); LCMS : m/e 472.0 (M+ 1).
EXAMPLE 217
[001156] Preparation of N-(3-(5-fluoro-2-(3-fluoro-4-(2-hydroxy-2- methylpropoxy)pheny lamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-315
Figure imgf000379_0002
1-315
[001157] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 3-fluoro-4-(2-hydroxy-2-methylpropoxy)aniline in place of 4 in step 2. 1H NMR (DMSO-d6) δ ppm: 1.19 (s, 6H), 3.67 (s, 2H), 4.62 (s, IH), 5.75 (d, J = 10.4 Hz, IH), 6.25 (d, J = 17.2 Hz, IH), 6.45 (dd, J = 10 & 16.8 Hz, IH), 6.94 (t, J = 9.2 Hz, IH), 7.29 (t, J = 8 Hz, 2H), 7.43 (d, J = 8.4 Hz, IH), 7.49 (d, J = 7.6 Hz, IH), 7.67 (d, J = 13.6 Hz, IH), 7.94 (s, IH), 8.11 (d, J = 3.6 Hz, IH), 9.19 (s, IH), 9.45 (s, IH), 10.14 (s, IH); LCMS : m/e 456 (M-I).
EXAMPLE 218
[001158] Preparation of N-(3-(5-fluoro-2-(3-fluoro-4-(l-hydroxypropan-2- yloxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-333
Figure imgf000380_0001
1-333
[001159] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 3-fluoro-4-(2-hydroxy-l-methylethoxy)aniline in place of 4 in step 2. 1H NMR (DMSO-d6) δ ppm: 1.16 (d, J = 6.12 Hz, 3H), 3.40-3.46 (m, IH), 3.50-3.56 (m, IH), 4.22 (sextet, J = 5.6 Hz, IH), 4.84 (t, J = 5.68 Hz, IH), 5.75 (dd, J = 1.96 & 10.08 Hz, IH), 6.25 (dd, J = 1.92 & 16.92 Hz, IH), 6.46 (dd, J = 10.08 & 16.92 Hz, IH), 6.98 (t, J = 9.32 Hz, IH), 7.26-7.31 (m, 2H), 7.43 (d, J = 8.76 Hz, IH), 7.49 (d, J = 8 Hz, IH), 7.68 (dd, J = 2.44 & 14.28 Hz, IH), 7.95 (s, IH), 8.11 (d, J = 3.68 Hz, IH), 9.23 (s, IH), 9.46 (s, IH), 10.17 (s, IH); LCMS : m/e 442.2 (M+l).
EXAMPLE 219
[001160] Preparation of N-(3-(2-(4-(2,3-dihydroxypropoxy)-3-fluorophenylamino)-5- fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-334
Figure imgf000380_0002
1-334
[001161] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 4-(2,3-dihydroxypropoxy)-3-fluoroaniline in place of 4 in step 2. 1H NMR (DMS0-d6) δ ppm: 3.42 (t, J = 5.6 Hz, 2H), 3.7-3.8 (m, IH), 3.8- 3.9 (m, IH), 3.94 (dd, J = 4.36 & 9.92 Hz, IH), 4.65 (t, J = 5.64 Hz, IH), 4.93(d, J = 5.08 Hz, IH), 5.7-5.8 (m, IH), 6.24 (dd, J = 1.64 & 16.84 Hz, IH), 6.44 (dd, J = 10 & 16.96 Hz, IH), 6.94 (t, J = 9.32 Hz, IH), 7.28 (t, J = 7.96 Hz, 2H), 7.40 (d, J = 8.28 Hz, IH), 7.49 (d, J = 7.44 Hz, IH), 7.66 (d, J = 14.24 Hz, IH), 7.92 (s, IH), 8.09 (d, J = 3.6 Hz, IH), 9.17 (s, IH), 9.45 (s, IH), 10.15 (s, IH); LCMS : m/e 456 (M-I).
EXAMPLE 220
[001162] Preparation of N-(3-(2-(4-chloro-3-(l-hydroxy-2-methylpropan-2- yloxy)phenylamino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-336
Figure imgf000381_0001
1-336
[001163] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 4-chloro-3-(l-hydroxy-2-methylpropan-2- yloxy)aniline in place of 4 in step 2. 1H NMR (DMSO-d6) δ ppm: 1.22 (s, 6H), 3.47 (d, J = 5.88 Hz, 2H), 4.88 (t, J = 5.84 Hz, IH), 5.75 (dd, J = 3.24 & 10 Hz, IH), 6.25 (dd, J = 2 & 16.92 Hz, IH), 6.46 (dd, J = 10.12 & 17.08 Hz, IH), 7.15 (d, J = 8.8 Hz, IH), 7.31 (t, J = 8.2 Hz, IH), 7.40-7.45 (m, IH), 7.51-7.60 (m, 3H), 7.93 (s, IH), 8.13 (d, J = 3.56 Hz, IH), 9.24 (s, IH), 9.47 (s, IH), 10.12 (s, IH); LCMS : m/e 472.2 (M+ 1).
EXAMPLE 221
[001164] Preparation of N-(3-(2-(4-chloro-3-(l-hydroxypropan-2-yloxy)phenylamino)-5- fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-337
Figure imgf000381_0002
1-337 [001165] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 4-chloro-3-(l-hydroxypropan-2-yloxy)aniline in place of 4 in step 2. 1H NMR (DMSO-d6) δ ppm: 1.18 (d, J = 6.12 Hz, 3H), 3.40-3.47 (m, IH), 3.50-3.56 (m, IH), 4.20-4.30 (m, IH), 4.82 (t, J = 5.6 Hz, IH), 5.75 (dd, J = 1.88 & 10.08 Hz, IH), 6.25 (dd, J = 1.92 & 16.92 Hz, IH), 6.45 (dd, J = 10.08 & 16.92 Hz, IH), 7.12 (d, J = 8.76 Hz, IH), 7.29 (t, J = 8.08 Hz, IH), 7.40-7.44 (m, 3H), 7.52 (d, J = 8.44 Hz, IH), 7.91 (s, IH), 8.12 (d, J = 3.64 Hz, IH), 9.21 (s, IH), 9.45 (s, IH), 10.12 (s, IH); LCMS : m/e 458.0 (M+l).
EXAMPLE 222
[001166] Preparation of N-(3-(2-(4-(2,3-dihydroxypropoxy)phenylamino)-5- fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-335
Figure imgf000382_0001
1-335
[001167] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 4-(l-hydroxypropan-2-yloxy)aniline in place of 4 in step 2. 1H NMR (DMSO-d6) δ ppm: 3.43 (dd, J = 0.64 & 6.84 Hz, 2H), 3.70-3.80 (m, 2H), 3.85-3.95 (m, IH), 4.62 (t, J = 5.6 Hz, IH), 4.88 (d, J = 4.76 Hz, IH), 5.75 (dd, J = 1.76 & 10.08 Hz, IH), 6.25 (dd, J = 1.72 & 16.92 Hz, IH), 6.45 (dd, J = 10.08 & 16.88 Hz, IH), 6.74 (d, J = 9 Hz, 2H), 7.27 (t, J = 8.08 Hz, IH), 7.39 (d, J = 8.04 Hz, IH), 7.38-7.53 (m, 3H), 7.93 (s, IH), 8.05 (d, J = 3.68 Hz, IH), 8.93 (s, IH), 9.35 (s, IH), 10.11 (s, IH); LCMS : m/e 440.3 (M+l). EXAMPLE 223
[001168] Preparation of (R)-N-(3-(2-(4-chloro-3-(tetrahydrofuran-3-yloxy)phenylamino)-5- fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-341
Figure imgf000383_0001
1-341
[001169] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using (R)- 4-chloro-3-(tetrahydrofuran-3- yloxy)aniline in place of 4 in step 2. 1H NMR (DMSO-d6) δ ppm: 1.85-1.95 (m, IH), 2.0-2.15 (m, IH), 3.60-3.70 (m, IH), 3.73-3.83 (m, 3H), 4.68 (s, IH), 5.74 (dt, J = 1.92 & 10.0 Hz, IH), 6.23 (dd, J = 1.88 & 16.92 Hz, IH), 6.43 (dd, J = 10.12 & 16.96 Hz, IH), 7.14 (d, J = 8.72 Hz, IH), 7.29 (t, J = 8.08 Hz, IH), 7.33 (dd, J = 4.16 & 8.76 Hz, IH), 7.41-7.47 (m, 3H), 7.90 (s, IH), 8.13 (d, J = 3.56 Hz, IH), 9.28 (s, IH), 9.47 (s, IH), 10.13 (s, IH); LCMS : m/e 469.8 (M+l).
EXAMPLE 224
[001170] Preparation of N-(3-(5-fluoro-2-(3-fluoro-4-(l-hydroxy-2-methylpropan-2- yloxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-332
Figure imgf000383_0002
1-332
[001171] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 3-fluoro-4-(l-hydroxy-2-methylpropan-2- yloxy)aniline in place of 4 in step 2. 1H NMR (DMSO-d6) δ ppm: 1.13 (s, 6H), 3.36 (d, J = 5.84 Hz, 2H), 4.86 (t, J = 5.84 Hz, IH), 5.73 (dd, J = 1.96 & 10.04 Hz, IH), 6.24 (dd, J = 1.96 & 16.96 Hz, IH), 6.44 (dd, J = 10.08 & 16.92 Hz, IH), 6.95 (t, J = 9.16 Hz, IH), 7.23 (dd, J = 1.64 & 8.96 Hz, IH), 7.28 (t, J = 8.12 Hz, IH), 7.43 (d, J = 8.8 Hz, IH), 7.48 (d, J = 7.92 Hz, IH), 7.70 (dd, J = 2.48 & 13.84 Hz, IH), 7.92 (s, IH), 8.11 (d, J = 3.68 Hz, IH), 9.25 (s, IH), 9.45 (s, IH), 10.10 (s, IH); LCMS: m/e 456.2 (M+l).
EXAMPLE 225
[001172] Preparation of N-(3-(2-(3-(2,3-dihydroxypropoxy)phenylamino)-5- fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-339
Figure imgf000384_0001
1-339
[001173] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 3-(2,3-dihydroxypropoxy)aniline in place of 4 in step 2. 1H NMR (MeOD) δ ppm: 3.59-3.69 (m, 2H), 3.88-3.98 (m, 3H), 5.78 (dd, J = 2.16 & 9.6 Hz, IH), 6.36 (dd, J = 2.24 & 17.04 Hz, IH), 6.44 (dd, J = 9.56 & 16.96 Hz, IH), 6.54-6.57 (m, IH), 7.08-7.12 (m, 2H), 7.32 (t, J = 7.92 Hz, 2H), 7.44 (dd, J = 7.88 & 13.4 Hz, 2H), 7.94 (d, J = 3.8 Hz, IH), 8.09 (s, IH); LCMS : m/e 440.1 (M+l).
EXAMPLE 226
[001174] Preparation of N-(4-(5-fiuoro-2-(3-fluoro-4-(2- methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-351
Figure imgf000384_0002
1-351 [001175] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using tert-butoxycarbonylamino-4-aminoaniline in place of 2 in step 1 and 3-fluoro-4-(2-methoxyethoxy)aniline in place of 4 in step 2. 1H NMR (DMSO-de) δ ppm: 3.30 (s, 3H), 3.63 (t, J = 4.6 Hz, 2H), 4.08 (t, J = 4.48 Hz, 2H), 5.74 (dd, J = 2 & 10.08 Hz, IH), 6.25 (dd, J = 1.96 & 16.92 Hz, IH), 6.44 (dd, J = 10.04 & 16.96 Hz, IH), 7.02 (t, J = 9.48 Hz, IH), 7.23 (bd, J = 7.44 Hz, IH), 7.64 (d, J = 9 Hz, 2H), 7.70-7.74 (m, 3H), 8.07 (d, J = 3.72 Hz, IH), 9.19 (s, IH), 9.34 (s, IH), 10.13 (s, IH); LCMS : m/e 442.0 (M+l).
EXAMPLE 227
[001176] Preparation of 2-((3-(5-fluoro-2-(6-(2-hydroxy-2-methylpropoxy)pyridin-3- ylamino)pyrimidin-4-ylamino)phenyl)(hydroxy)methyl)acrylonitrile 1-312
Figure imgf000385_0001
1-312
[001177] The title compound was prepared according to the schemes, steps and intermediates described in Example 107, by using 3-amino-6-(2-hydroxy-2- methylpropoxy)pyridine in place of 4 in step 2. 1H NMR (DMSO-d6) δ ppm: 1.18 (s, 6H), 3.98 (s, 2H), 4.61 (s, IH), 5.31 (d, J = 3.88 Hz, IH), 6.13 (s, IH), 6.19 (s, IH), 6.32 (d, J = 4 Hz, IH),
6.75 (d, J = 8.88 Hz, IH), 7.10 (d, J = 7.72 Hz, IH), 7.33 (t, J = 7.84 Hz, IH), 7.68 (s, IH), 7.84 (d, J = 7.52 Hz, IH), 7.96 (dd, J = 2.72 & 8.88 Hz, IH), 8.08 (d, J = 3.64 Hz, IH), 8.33 (d, J =
1.76 Hz, IH), 9.06 (s, IH), 9.44 (s, IH); LCMS : m/e 451 (M+l). EXAMPLE 228
[001178] Preparation of 4-(4-(4-(3-acrylamidophenylamino)-5-fluoropyrimidin-2- ylamino)phenoxy)-N-methylpicolinamide 1-342
Figure imgf000386_0001
1-342
[001179] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 4-(4-aminophenoxy)-N-methylpicolinamide in place of 4 in step 2. LC/MS (M + H) 500.2
EXAMPLE 229
[001180] Preparation of (R)- 1 -(3 -(3 -fluoro-4-(2-methoxyethoxy)pheny lamino)pyrimidin-4- ylamino)piperidin- 1 -yl)prop-2-en- 1 -one 1-344
Figure imgf000386_0002
1-344
[001181] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using (R)-l-tert-butoxycarbonyl-3-aminopiperidine in place of 2 in step 1 and 3-fluoro-4-(2-methoxyethoxy)aniline in place of 4 in step 2. LC/MS (M + H) 434.1. EXAMPLE 230
[001182] Preparation of (R)- 1 -(3 -(4-(2-methoxyethoxy)phenylamino)pyrimidin-4- ylamino)piperidin- 1 -yl)prop-2-en- 1 -one 1-345
Figure imgf000387_0001
1-345
[001183] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using (R)-l-tert-butoxycarbonyl-3-aminopiperidine in place of 2 in step 1 and 4-(2-methoxyethoxy)aniline in place of 4 in step 2. LC/MS (M + H) 416.2
EXAMPLE 231
[001184] Preparation of 4-(4-(4-(3 -acrylamidophenylamino)-5 -fluoropyrimidin-2- ylamino)phenoxy) pyridine 1-346
Figure imgf000387_0002
1-346
[001185] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 4-(4-aminophenoxy)pyridine in place of 4 in step 2. LC/MS (RT = 2.802/(M + H)) 500.2 EXAMPLE 232
[001186] Preparation of l-((R)-3-(5-fluoro-2-(4-((S)-tetrahydrofuran-3- yloxy)phenylamino)pyrimidin-4-ylamino)piperidin- 1 -yl)prop-2-en- 1 -one 1-347
Figure imgf000388_0001
1-347
[001187] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using (R)-l-tert-butoxycarbonyl-3-aminopiperidine in place of 2 in step 1 and 4-(S)-(tetrahydrofuran-3-yloxy)aniline in place of 4 in step 2. LC/MS (M + H) 428.3.
EXAMPLE 233
[001188] Preparation of l-((R)-3-(5-fluoro-2-(4-((R)-tetrahydrofuran-3- yloxy)phenylamino)pyrimidin-4-ylamino)piperidin- 1 -yl)prop-2-en- 1 -one 1-348
Figure imgf000388_0002
1-348
[001189] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using (R)-l-tert-butoxycarbonyl-3-aminopiperidine in place of 2 in step 1 and 4-(R)-(tetrahydrofuran-3-yloxy)aniline in place of 4 in step 2. LC/MS (M + H) 428.3. EXAMPLE 234
[001190] Preparation of N-(3-(2-(2,3-dihydrobenzo[6]l,4]dioxin-6-ylamino)-5- fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-349
Figure imgf000389_0001
1-349
[001191] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 6-amino-2,3-dihydrobenzo[δ]l,4]dioxane in place of 4 in step 2. LC/MS (M + H) 408.
EXAMPLE 235
[001192] Preparation of l-(6-(4-(3-chloro-4-(pyridine-2-ylmethoxy)phenylamino)-5- fluoropyrimidin-2-ylamino)-2H-benzo[δ][l,4]oxazin-4(3H)-yl)prop-2-en-l-one 1-343
Figure imgf000389_0002
1-343
[001193] The title compound was prepared according to the schemes, steps and intermediates described in Example 35, using 3-chloro-4-(pyridine-2-ylmethoxy)aniline in place of 2 in step 1. LC/MS (M + H) 533.1. EXAMPLE 236
[001194] Preparation of N-(3-(5-cyano-2-(3-fluoro-4-(2- methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-350
Figure imgf000390_0001
1-350
[001195] The title compound was prepared according to the schemes, steps and intermediates described in Example 94, by using 3-fluoro-4-(2-methoxyethoxy)aniline for 4 in step 2. LC/MS (M + H) 449.1
EXAMPLE 237
[001196] Preparation of N-(3-(5-trifiuoromethyl-2-(3-fiuoro-4-(2- methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-352
Figure imgf000390_0002
1-352
[001197] The title compound was prepared according to the schemes, steps and intermediates described in Example 189 using 3-fluoro-4-(2-methoxyethoxy)aniline for 2 in step 1. LC/MS (M + H) 492.1 EXAMPLE 238
[001198] Preparation of N-(3-(2-(4-chloro-3-(2-methoxyethoxy)phenylamino)-5- fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-321
Figure imgf000391_0001
1-321
[001199] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 4-chloro-3-(2-methoxyethoxy)aniline in place of 4 in step 2. 1H NMR (DMSO-d6) δ ppm: 3.30 (s, 3H), 3.60 (t, J = 4.56 Hz, 2H), 3.88 (t, J = 3.48 Hz, 2H), 5.74 (dd, J = 4.36 & 10.0 Hz, IH), 6.24 (dd, J = 1.8 & 16.88 Hz, IH), 6.44 (dd, J = 4.36 & 10.0 Hz, IH), 7.13 (d, J = 8.72 Hz, IH), 7.28 (t, J = 8.04 Hz, IH), 7.33 (dd, J = 2.16 & 8.8 Hz, IH), 7.41 (d, J = 7.96 Hz, IH), 7.47-7.49 (m, 2H), 7.84 (s, IH), 8.13 (d, J = 3.6 Hz, IH), 9.27 (s, IH), 9.47 (s, IH), 10.12 (s, IH); LCMS : m/e 458.0 (M+l).
EXAMPLE 239
[001200] Preparation of N-(3-(5-fluoro-2-(6-(2-hydroxy-2-methylpropoxy)pyridin-3- ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-313
Figure imgf000391_0002
1-313
[001201] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 3-amino-6-(2-hydroxy-2- methylpropoxy)pyridine in place of 4 in step 2. 1H NMR (DMSO-d6) δ ppm: 1.16 (s, 6H), 3.93 (s, 2H), 4.57 (s, IH), 5.74 (dd, J = 1.68 & 10.04 Hz, IH), 6.24 (dd, J = 1.84 & 16.92 Hz, IH), 6.45 (dd, J = 10.04 & 16.88 Hz, IH), 6.65 (d, J = 8.88 Hz, IH), 7.26 (t, J = 8.04 Hz, IH), 7.39 (d, J = 8 Hz, IH), 7.48 (d, J = 7.8 Hz, IH), 7.91 (s, IH), 7.99 (dd, J = 2.72 & 8.92 Hz, IH), 8.07 (d, J = 3.68 Hz, IH), 8.27 (d, J = 2.52 Hz, IH), 9.06 (s, IH), 9.41 (s, IH), 10.1 (s, IH); LCMS : m/e 439.0 (M+ 1).
EXAMPLE 240
[001202] Preparation of N-(3-(5-fluoro-2-(3-fluoro-4-(3-
(methylsulfony l)propoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-318
Figure imgf000392_0001
1-318
[001203] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 3-fluoro-4-(3-(methylsulfonyl)propoxy)aniline in place of 4 in step 2. 1H NMR (DMSO-d6) δ ppm: 2.05-2.15 (m, 2H), 3.01 (s, 3H), 3.24 (t, J = 7.56 Hz, 2H), 4.05 (t, J = 6.12 Hz, 2H), 5.74 (dd, J = 1.84 & 9.72 Hz, IH), 6.24 (dd, J = 1.72 & 16.96 Hz, IH), 6.44 (dd, J = 10 & 16.84 Hz, IH), 6.96 (t, J = 9.36 Hz, IH), 7.28 (t, J = 8.04 Hz, 2H), 7.40 (d, J = 7.8 Hz, IH), 7.48 (d, J = 8.32 Hz, IH), 7.69 (dd, J = 2.2 & 14.4 Hz, IH), 7.91 (s, IH), 8.10 (d, J = 3.64 Hz, IH), 9.20 (s, IH), 9.44 (s, IH), 10.12 (s, IH); LCMS : m/e 504.2 (M+l).
EXAMPLE 241
[001204] Preparation of l-(3-(5-fiuoro-2-(3-fluoro-4-(2- methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-4-methylpent-3-en-2-one 1-330
Figure imgf000393_0001
1-330
[001205] The title compound was prepared according to the schemes, steps and intermediates described in Example 112, by using ethyl 4-aminomethylbenzoate in place of 2 in step 1 and 3-fluoro-4-(2-methoxyethoxy)aniline in place of 4 in step 2. 1H NMR (DMSO-dβ) δ ppm: 1.83 (s, 3H), 2.05 (s, 3H), 3.31 (s, 3H), 3.64 (t, J = 4.56 Hz, 2H), 3.69 (s, 2H), 4.08 (t, J = 4.4 Hz, 2H), 6.18 (s, IH), 6.92 (d, J = 7.44 Hz, IH), 7.01 (t, J = 9.36 Hz, IH), 7.27 (t, J = 7.84 Hz, 2H), 7.51 (s, IH), 7.64-7.71 (m, 2H), 8.09 (d, J = 3.64 Hz, IH), 9.19 (s, IH), 9.34 (s, IH); LCMS : m/e 469.1 (M+ 1).
EXAMPLE 242
[001206] Preparation of N-(3-(2-(4-chloro-3-(2,3-dihydroxypropoxy)phenylamino)-5- fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-353
Figure imgf000393_0002
1-353
[001207] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000394_0001
A) Pd(OAc)2, BINAP, Cs2CO3, toluene, 110 0C 16 h; B) TFA, CH2Cl2, rt, 2 h; C) acryloyl chloride, K2CO3, NMP, rt, 45 min. [001208] Step-1
Figure imgf000394_0002
[001209] A solution of 2 (200 mg, 0.77 mmol), 1 (262 mg, 0.77 mmol), Pd(OAc)2 (17.3 mg, 0.07 mmol), BINAP (24 mg, 0.038 mmol) and Cs2CO3 (630 mg, 1.9 mmol) in degassed toluene (toluene was purged with N2 for 30 min) was heated for 16 h at 100 0C under N2 atmosphere. The reaction mixture was cooled, diluted with EtOAc (15 mL) and filtered through Celite®. The filtrate was washed with water (5 mL) and brine (3 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give 3 (0.3 g, 69 %) as a yellow solid. [001210]
Figure imgf000395_0001
[001211] To a stirred solution of 3 (300 mg, 0.5 mmol) in dry CH2Cl2 (6 mL) at 0 0C was added CF3COOH (3 mL), and the reaction mixture was kept at this temperature for 30 min. The reaction was allowed to come to rt and stirred at this temperature for 3 h. The reaction mixture was concentrated under reduced pressure, and the residue was quenched with water (5 mL), basifϊed with NaCO3 solution, and extracted with ethyl acetate (2x10 mL). The combined extracts were washed with water (5 mL) and brine (5 mL), dried over Na2SO4, and concentrated under reduced pressure to get 4 (200 mg, 88 %) as a yellow solid. [001212] Step-3
Figure imgf000395_0002
[001213] To a stirred solution of 4 (240 mg, 0.5 mmol), in NMP (1.5 mL) at 0 0C was added potassium carbonate (780 mg, 5.7 mmol) and acryloyl chloride (57 mg, 0.5 mmol), and the reaction mixture was stirred at 0 0C for 3 h. The reaction mixture was further stirred at rt for 30 min and quenched by dropwise addition to a cold, stirring solution of 10% NaHCO3 and stirred at 0 0C for 30 min. A solid precipitated out and was isolated by filtration through a Buchner funnel. The solid was washed with cold water, dissolved in EtOAc (20 mL) and was basified by using triethylamine and washed with water (2 mL), brine (1 mL), dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by preparative HPLC to give the title compound (45 mg, 15.5 %) as a white solid. 1U NMR (DMSO-d6) δ ppm: 3.43-3.50 (m, 2H), 3.78-3.85 (m, 2H), 3.89-3.92 (m, IH), 4.65 (t, J = 5.6 Hz, IH), 4.93 (d, J = 4.8 Hz, IH), 5.76 (dd, J = 1.92 & 10.04 Hz, IH), 6.26 (dd, J = 1.92 & 16.92 Hz, IH), 6.46 (dd, J = 10.08 & 16.92 Hz, IH), 7.14 (d, J= 8.72 Hz, IH), 7.30 (t, J= 8.08 Hz, IH), 7.39-7.43 (m, 2H), 7.46 (dd, J= 2.2 & 8.72 Hz, IH), 7.56 (d, J= 8.04 Hz, IH), 7.94 (s, IH), 8.14 (d, J= 3.6 Hz, IH), 9.24 (s, IH), 9.48 (s, IH), 10.13 (s, IH); LCMS : m/e 473.8 (M+). [001214] Synthesis of intermediate 2
Figure imgf000396_0001
A) DIAD, PPh3, Et3N, dry THF, rt, 1 h; B) H2, Ra Ni, methanol, 2 h. [001215] Step-1
Figure imgf000396_0002
[001216] To a stirred solution of 2' (0.640 g, 3.7 mmol) in THF (20 niL) were added 1' (0.5 g, 3.7 mmol), PPh3 (1.09 g, 4.1 mmol) and Et3N (0.73 g, 5.6 mmol) under N2 atmosphere. The reaction mixture was cooled to 0 0C and DIAD (0.84 g, 4.1 mmol) was added. The reaction mixture was allowed to come to rt and stir for 1 h. The reaction was quenched with water, extracted with ethyl acetate (3x10 mL), and the combined extracts were washed with water and brine solution (5 mL each). The residue obtained after concentration under reduced pressure was purified by column chromatography (SiO2, 60-120, pet ether/ethyl acetate, 9/1) to give 3' (0.6 g, 60 %) as a white solid. [001217] Step-2
Figure imgf000397_0001
[001218] To a solution of 3' (0.3 g, 1.04 mmol) in methanol was added Raney Ni (60 mg,
20% w/w) under N2, and the reaction mixture was kept under H2 atmosphere (bladder pressure) for 16 h. The reaction mixture was filtered through a bed of Celite®, and the filtrate was concentrated under reduced pressure. The residue was diluted with 1.5 N HCl (2 mL) and washed with ethyl acetate (5 mL) to remove organic impurities. The aqueous layer was basified with NaHCO3 solution (5 mL), extracted with ethyl acetate, washed with water (2 mL) and brine (2 mL), and dried over anhydrous Na2SO4. Filtration followed by concentration under reduced pressure gave 2 (0.2 g, 76.9 %) as a brown liquid.
EXAMPLE 243
[001219] Preparation of (S)-N-(3-(2-(4-chloro-3-(tetrahydrofuran-3-yloxy)phenylamino)-5- fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-354
Figure imgf000397_0002
1-354
[001220] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using (S)- 4-chloro-3-(tetrahydrofuran-3- yloxy)aniline in place of 4 in step 2. LCMS : m/e 469.8 (M+ 1). EXAMPLE 244
[001221] Preparation of (N-(3-(5-fluoro-2-(3-fluoro-4-(((2S,4R)-4-hydroxypyrrolidin-2- yl)methoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-355
Figure imgf000398_0001
1-355
[001222] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000398_0002
A) Pd(OAc)2, BINAP, Cs2CO3, toluene, 110 0C, 6 h; B) TFA, CH2Cl2, rt, 1 h; C) (Boc)2O, 30 min then acryloyl chloride, K2CO3, NMP, O 0C, 90 min; D) HF (49% aq. Solution), CH3CN, rt, 2 h; E) TFA, DCM, rt, 2 h. [001223]
Figure imgf000399_0001
[001224] A solution of 2 (0.50 g, 1.13 mmol), 1 (0.30g, 1.13 mmol), Pd(OAc)2 (0.0025 g, 0.1 mmol), BINAP (0.0035 g, 0.05 mmol) and Cs2CO3 (0.92 g, 2.8 mmol) in degassed toluene (toluene was purged with N2 for 30 min) was heated at 110 0C for 16 h under N2 atmosphere. The reaction mixture was cooled, diluted with EtOAc (20 mL), washed with water (10 mL), brine (10 mL) and dried over Na2SO4. Filtration followed by concentration under reduced pressure offered a residue which was further washed with hexane to give 3 (0.3 g, 42.8%) as a yellow solid.
[001225]
Figure imgf000399_0002
[001226] To a solution of 3 (0.3 g, 0.44 mmol) in methanol (5 mL)) was added Pd/C (0.030 g, 10% w/w) and the reaction mixture was allowed to stir under H2 atmosphere (balloon) at rt for 16 h. The reaction mixture was filtered through a pad of Celite® and was concentrated under reduced pressure to give 4 (0.19 g, 67.6%) as a yellow solid. [001227] Step-3
Figure imgf000399_0003
[001228] To a stirred solution of 4 (0.1 g, 0.15 mmol) in NMP (1.0 mL) at rt was added
Boc anhydride (0.046 g, 0.212 mmol) and the reaction mixture was stirred at rt for 60 min. It was then cooled to 0 0C and to it was added K2CO3 (0.107 g, 0.77 mmol), acryloyl chloride (0.016 g, 0.18 mmol) and the reaction mixture was stirred at 0 0C for 90 min. The reaction mixture was added drop wise, to a cold, stirring solution of 10% NaHCO3. After the addition was over, the solution was stirred for another 30 min at 0 0C, and the solid was isolated by filtration through a Buchner funnel. The solid was washed with cold water, hexane and was dissolved in methanol: dichloromethane (50:50, 10 mL) and concentrated under reduced pressure. The residue obtained was suspended in cold water (5 mL), Et3N was added to it and it was extracted with ethyl acetate (2x10 mL). The combined ethyl acetate extract was washed with water (5 mL), brine (5 mL), dried over Na2SO4 and concentrated under reduced pressure. The residue was further purified by column chromatography (SiO2, methanol/chloroform: 4/96) to give 5 (0.075 g, 71.4%) as yellow solid. [001229] Step-4
Figure imgf000400_0001
[001230] To a solution of 5 (15 mg, 0.02 mmol) in acetonitrile was added HF (49% aq.
Solution, 0.0048 mL, 0.024 mmol) at 0 0C. The reaction mixture stirred at rt for 2 h, was extracted with ethyl acetate (2 mL), was washed with water (1 mL), and was dried over Na2SO4 and filtered. The filtrate was concentrated under reduced pressure. The residue showed 60% purity by LCMS and was used in the next step without further purification. [001231] Step-5
Figure imgf000400_0002
[001232] To a stirred solution of 6 (0.008 g, 0.013 mmol) in CH2Cl2 (0.024 mL) was added
TFA (0.016 mL) at 0 0C. The reaction mixture was allowed to come to rt and was stirred for additional 2 h. It was then concentrated and was stirred with cold 10% NaHCO3 (1.0 mL). It was extracted with EtOAc (2x2 rnL) and the combined EtOAc extract was washed with brine (1 niL), was dried over Na2SO4 and was concentrated under reduced pressure. The crude residue was further purified by column chromatography (SiO2, methanol/chloroform: 2/98) and was then purified by preparative TLC to give the title compound (2 mg, 81% purity by HPLC, and 79% purity by LCMS) as a white solid. LCMS : m/e 483 (M+).
[001233] Compound 2 was prepared according to the schemes, steps and intermediates described below.
Figure imgf000401_0001
A) MeOH, SOCl2, reflux, 5 h; B) (Boc)2O, Et3N, CH2Cl2, rt, 5 h; C) TBDMS-Cl, imidazole, DMF, rt, 16 h; D) LAH solution (IM in THF), -20 0C, 20 min; E) DIAD, PPh3, Et3N, THF, 16 h; F) H2, Pd/C, methanol, rt, 16 h. [001234] Step-1
Figure imgf000401_0002
[001235] To a stirred solution of 1' (2 g, 15.26 mmol) was added a solution prepared by adding thionyl choride (2 mL) to methanol (20 mL). The reaction mixture was heated at reflux for 5 h. After completion of the reaction, methanol was removed under reduced pressure to give 2' as colorless salt (3.0 g) it was used as such in the next reaction. [001236] Step-2
Figure imgf000402_0001
31
[001237] To a stirred solution of V (3.0 g, 12.24 mmol) in DCM (30 mL) was added Et3N
(1.85 g, 18.31 mmol) and Boc anhydride (2.92 g, 13.46 mmol). Stirring was continued at rt for 5 h after which the reaction was quenched with water. The organic layer was separated, dried and concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, 60-120, 100% ethyl acetate) to give 3' (3.2 g, 64%) as a white solid. [001238] Step-3
Figure imgf000402_0003
[001239] To a stirred solution of 3' (3 g, 12.24 mmol) in DMF (30 mL) was added imidazole (1.2 g, 18.36 mmol) followed by TBDMS chloride (1.84 g, 12.24 g). Stirring was continued for 16 h. The reaction mixture was diluted with ethyl acetate (50 mL) and the ethyl acetate layer was separated. It was washed with water (5 mL), brine solution (5 mL) and dried over Na2SO4. Filtration followed by concentration under reduced pressure offered a residue which was purified by column chromatography (SiO2, 60-120, petroleum ether/ ethyl acetate : 6/4) to give 4' (3.2 g, 80%) as a colorless liquid. [001240] Step-4
Figure imgf000402_0002
[001241] To a stirred solution of 4' (0.5 g, 1.39 mmol) in THF (5 mL) was added LAH
(1.39 mL, IM solution, 1.39 mmol) at -20 0C. The reaction was continued at the same temperature for 15 min after which it was quenched with Na2SO4 solution. The reaction mass was filtered through celite® and filtrate was concentrated under reduced pressure. The residue was diluted with ethyl acetate (10 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced under reduced pressure to give 5' (0.3 g, 65%) as a colorless liquid. [001242] Step-5
Figure imgf000403_0001
[001243] To a stirred solution of 5' (0.1 g, 0.3 mmol) in THF (6 mL) were added 6' (0.047 g, 0.3 mmol), PPh3 (0.16 g, 0.64 mmol) and Et3N (0.048 g, 0.48 mmol) under N2 atmosphere. The reaction mixture was cooled to 0 0C and to it was added DIAD (0.094 g, 0.48 mmol). The reaction mixture was allowed to come to rt and stirred at it for 1 h. It was quenched with water, was extracted with ethyl acetate (2x5 mL) and the combined ethyl acetate extract was washed with water and brine solution (5 mL each). The residue obtained after concentration under reduced pressure was purified by column chromatography (SiO2, 60-120, pet ether/ethyl acetate, 9/1) to give T (0.120 g, 85%) as a yellow solid [001244] Step-6 TBDMS
Figure imgf000403_0002
2
[001245] To a solution of T (0.1 g, 0.21 mmol) in methanol (5 mL)) was added Pd/C
(0.010 g, 10% w/w) and the reaction mixture was allowed to stir under H2 atmosphere (bladder) at rt for 16 h. The reaction mixture was filtered through a pad of celite® and concentrated under reduced pressure to give 2 (0.085 g, 91%) as a brownish viscous oil. It was used in the next step without further purification. EXAMPLE 245
[001246] Preparation of tert-butyl 2-(2-(4-(4-(3-acrylamidophenylamino)-5- fluoropyrimidin-2-ylamino)-2-fluorophenoxy)ethoxy)ethylcarbamate 1-356
Figure imgf000404_0001
1-356
[001247] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000404_0002
Figure imgf000404_0003
A) Pd(OAc)2, BINAP, Cs2CO3, toluene, 110 0C, 6 h; B) TFA, CH2Cl2, RT 1 h; C) (Boc)2O, 30 min then acryloyl chloride, K2CO3, NMP, 0 0C, 90 min. [001248] Step-1
Figure imgf000404_0004
[001249] A solution of 2 (0.050 g, 0.159 mmol), 1 (0.053 g, 0.159 mmol), Pd(OAc)2 (0.0035 g, 0.01590 mmol), BINAP (0.0049 g, 0.0079 mmol) and Cs2CO3 (0.129 g, 0.3975mmol) in degassed toluene (toluene was purged with N2 for 30 min) was heated at 110 0C for 16 h under N2 atmosphere. The reaction mixture was cooled, diluted with EtOAc (20 mL), washed with water (10 mL), brine (10 mL) and dried over Na2SO4. Filtration followed by concentration under reduced pressure offered a residue which was further washed with hexane to give 3 (0.049 g, 50%) as a brown solid. [001250] Step-2
Figure imgf000405_0001
[001251] To a stirred solution of 3 (0.047 g, 0.0762 mmol) in dry CH2Cl2 (3 mL) at 0 0C was added CF3COOH (1.0 mL) and the reaction mixture was stirred at 0 0C for 30 min. The reaction was allowed to come to rt and stirred at it for 1 h. It was concentrated under reduced pressure and the residue was quenched with NaHCO3 solution (3 mL). The contents were extracted with ethyl acetate (3x10 mL) and the combined EtOAc extract was washed with water (10 mL) followed by 10% citric acid solution (3x10 mL). The combined citric acid extract was basifϊed with 10% NaOH solution and extracted with EtOAc (3x25 mL). The EtOAc extract was washed with water (20 mL), brine (10 mL) and dried over Na2SO4. to get 4 (0.028 g, 88%) as a light yellow solid.
[001252] Step-3
NHBoc
Figure imgf000405_0002
1-356
[001253] To a stirred solution of 4 (0.028 g, 0.06731 mmol) in NMP (1.0 mL) at rt was added (Boc)2O (0.016 g, 0.07404 mmol) and the reaction mixture was stirred at rt for 30 min. It was cooled to 0 0C and to it was added K2CO3 (0.051 g, 0.372 mmol) and acryloyl chloride (0.0067 g, 0.07404 mmol) and the reaction mixture was stirred at 0 0C for 30 min. The reaction mixture was added dropwise, to a cold, stirring solution of 10% NaHCO3. After the addition was over, the solution was stirred for another 30 min at 0 0C, and the solid was isolated by filtration through a Buchner funnel. The solid was washed with cold water, hexane and was dissolved in methanol: dichloromethane (50:50, 5 mL) and concentrated under reduced pressure. The residue obtained was suspended in cold water (3 mL), Et3N was added to it and it was extracted with ethyl acetate (2x5 mL). The combined ethyl acetate extract was washed with water (5 mL), brine (5 mL), dried over Na2SO4 and concentrated under reduced pressure to give the title compound (0.016 g, 42 %) as a grey solid. 1H NMR (DMSO-d6) δ ppm: 1.37 (s, 9H), 3.09 (d, J = 5.5 Hz, 2H), 3.43 (d, J= 5.84 Hz, 2H), 3.69 (s, 2H), 4.05 (s, 2H), 5.75 (d, J= 11.12 Hz, IH), 6.25 (d, J = 16.76 Hz, IH), 6.46 (dd, J = 10.12 & 16.84 Hz, IH), 6.81 (s, IH), 6.96 (t, J = 9.12 Hz, IH), 7.24-7.31 (m, 2H), 7.43 (d, J= 7.96 Hz, IH), 7.49 (d, J= 7.56 Hz, IH), 7.68 (d, J= 14 Hz, IH), 7.94 (s, IH), 8.11 (d, J = 3.24 Hz, IH), 9.21 (s, IH), 9.46 (s, IH), 10.15 (s, IH); LCMS : m/e 571.1 (M+l).
[001254] The intermediate 2 was prepared according to the schemes, steps and intermediates described below.
Figure imgf000406_0001
Figure imgf000406_0002
41 2
A) (BoC)2O, aq. NaOH, rt, 16 h; B) DIAD, PPh3, Et3N, dry THF, rt, 1 h; C) H2, Pd/C, ethanol, rt, 16 h. [001255]
Figure imgf000407_0003
[001256] To a solution of NaOH (0.76 g, 0.019 mmol) in water (9.6 niL) at rt was added 1'
(2.0 g, 19.022 mmol) and the reaction was stirred for 30 min. A solution of Boc-anhydride (4.561 g, 20.92 mmol) in THF (12.0 mL) was added dropwise over 5 min to it. The reaction mixture was stirred at rt for 16 h. It was concentrated under reduced pressure, diluted with water (20 mL) and extracted with EtOAc (4x50 mL). The combined EtOAc extract was washed with water (50 mL), brine (50 mL), dried over Na2SO4 to give V (3.2 g, 82%) as a viscous oil. [001257] Step-2
Figure imgf000407_0001
[001258] To a stirred solution of 2' (0.38 g, 1.851 mmol) in THF (6 mL) were added 3'
(0.29 g, 1.851 mmol), PPh3 (0.534 g, 2.0361 mmol) and Et3N (0.280 g, 2.776 mmol) under N2 atmosphere. The reaction mixture was cooled to 0 0C and to it was added DIAD (0.411 g, 2.0361 mmol). The reaction mixture was allowed to come to rt and stirred at it for 1 h. It was quenched with water, extracted with ethyl acetate (3x5 mL) and the combined ethyl acetate extract was washed with water and brine solution (5 mL each). The residue obtained after concentration under reduced pressure was purified by column chromatography (SiO2, 60-120, pet ether/ethyl acetate, 9/1) to give 4' (0.360 g, crude) as a yellow solid [001259] Step-3
Figure imgf000407_0002
[001260] To a solution of 4' (0.360 g, 1.0456 mmol) in ethanol (10 mL)) was added Pd/C
(0.072 g, 20% w/w) and the reaction mixture was allowed to stir under H2 atmosphere (1.5 Kg hydrogen pressure) at rt for 16 h. The reaction mixture was filtered through a pad of celite® and concentrated under reduced pressure to give 2 (0.28 g, 85%) as a brownish viscous oil. It was used in the next step without further purification. EXAMPLE 246
[001261] Preparation of N1-(2-(2-(4-(4-(3-acrylamidophenylamino)-5-fluoropyrimidin-2- ylamino)-2-fluorophenoxy)ethoxy)ethyl)-N5-(15-oxo-18-((3aR,4R,6aS)-2-oxohexahydro-lH- thieno[3,4-d]imidazol-4-yl)-4,7,10-trioxa-14-azaoctadecyl)glutaramide 1-362
Figure imgf000408_0001
[001262] The title compound was prepared according to the schemes, steps and intermediates described in Example 204, by using tert-butyl 2-(2-(4-(4-(3- acrylamidophenylamino)-5-fluoropyrimidin-2-ylamino)-2-fluorophenoxy)ethoxy)ethylcarbamate (1-356, described in Example 245) in place of 1-45 in step 1. 1U NMR (DMSO-d6) δ ppm: 10.1 (s, IH), 9.87 (s, IH), 9.52 (s, IH), 8.09 (d, J= 4.1 Hz, IH), 7.86 (s, IH), 7.78 (t, J= 5.5 Hz, IH), 7.66 (m, 3H), 7.48 (dd, J= 2.3 & 13.8 Hz, IH), 7.33 (m, 2H), 7.21 (t, J= 7.8 Hz, IH), 7.11 (d, J = 9.2 Hz, IH), 6.90 (t, J= 9.2 Hz, IH), 6.34 (m, 2H), 6.14 (dd, J= 2.3 & 17.0 Hz, IH), 5.66 (dd, J= 2.3 & 17.0 Hz, IH), 4.20 (dd, J= 5.0 & 7.3 Hz, IH), 3.99 (m, 3H), 3.61 (m, 2H), 3.12 (q, J = 6.0 Hz, 2H), 2.97 (m, 9H), 2.72 (m, 2H), 2.46 (m, 2H), 1.95 (m, 9H), 1.1-1.6 (m, 18H); LCMS : m/e 1013. (M+ 1).
EXAMPLE 247
[001263] Preparation of N-(3-(5-fluoro-2-(3-fluoro-4-(2-(2-methoxyethoxy)ethoxy)- phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-359
Figure imgf000408_0002
[001264] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000409_0001
A) DIAD, PPh3, Et3N, dry THF, rt, 1 h; B) H2, Pd/C, methanol, rt, 16 h; C) Pd(OAc)2, BINAP, Cs2CO3, toluene, 110 0C, 6 h; D) TFA, CH2Cl2, rt, 1 h; E) (BOC)2O, 30 min, then K2CO3, NMP, O 0C, 15 min.
[001265]
Figure imgf000409_0002
[001266] To a stirred solution of 1 (0.5 g, 3.18 mmol) in THF (10 mL) were added 2 (0.38 g, 3.18 mmol), PPh3 (0.91 g, 3.498 mmol) and Et3N (0.48 g, 4.776 mmol) under N2 atmosphere. The reaction mixture was cooled to 0 0C and to it was added DIAD (0.707 g, 3.5 mmol). The reaction mixture was allowed to come to rt and stirred at it for 1 h. It was quenched with water, extracted with ethyl acetate (3x5 mL) and the combined EtOAc extract was washed with water and brine solution (5 mL each). The residue obtained after concentration under reduced pressure was purified by column chromatography (SiO2, 60-120, pet ether/ethyl acetate, 7/3) to give 3 (0.61 g, 65%) as a white solid. [001267] Step-2
Figure imgf000410_0001
[001268] To a solution of 3 (0.6 g, 2.31 mmol) in ethanol (20 mL)) was added Pd/C (0.060 g, 10% w/w) and the reaction mixture was allowed to stir under H2 atmosphere (bladder pressure) at rt for 16 h. The reaction mixture was filtered through a pad of Celite® and concentrated under reduced pressure to give 4 (0.375 g, 70.7%) as a brownish viscous oil. [001269] Step-3
Figure imgf000410_0002
[001270] A solution of 4 (0.275 g, 1.19 mmol), 5 (0.403 g, 1.19 mmol), prepared according to Step-1 of Example 20, Pd(OAc)2 (0.0026 g, 0.11 mmol), BINAP (0.0037 g, 0.059 mmol) and CS2CO3 (0.969 g, 2.95 mmol) in degassed toluene (toluene was purged with N2 for 30 min) was heated at 110 0C for 16 h under N2 atmosphere. The reaction mixture was cooled, diluted with EtOAc (20 mL), washed with water (10 mL), brine (10 mL) and dried over Na2SO4. Filtration followed by concentration under reduced pressure offered a residue which was further purified by column chromatography (SiO2, 60-120, pet ether/ethyl acetate 5/5) to give 6 (0.350 g, 55%) as a yellow solid. [001271] Step-4
Figure imgf000410_0003
[001272] To a stirred solution of 6 (0.3 g, 0.56 mmol) in dry CH2Cl2 (3 mL) at 0 0C was added CF3COOH (1.0 mL) and the reaction mixture was stirred at 0 0C for 30 min. The reaction was allowed to come to rt and stirred at it for 1 h. It was concentrated under reduced pressure and the residue was quenched with NaHCO3 solution (3 mL) and extracted with EtOAc (3x25 rnL). The combined EtOAc extract was washed with water (20 mL), brine (10 mL) and dried over Na2SO4 to give 7 (0.15 g, 62.5%) as a light brown viscous liquid. [001273] Step-5
Figure imgf000411_0001
1-359
[001274] To a cooled solution of 7 (0.1 g, 0.23 mmol) in NMP (1.0 mL) at about 0 0C was added K2CO3 (0.15 g, 1.1 mmol), acryloyl chloride (0.0022 g, 0.25 mmol) and the reaction mixture was stirred at 0 0C for 30 min. The reaction mixture was added dropwise to a cold, stirring solution of 10% NaHCO3. After the addition was over, the solution was stirred for another 30 min at 0 0C, and the solid was isolated by filtration through a Buchner funnel. The solid was washed with cold water, hexane and was dissolved in methanol: dichloromethane (50:50, 25 mL) and concentrated under reduced pressure. The residue obtained was suspended in cold water (3 mL), Et3N was added to it and it was extracted with ethyl acetate (2x5 mL). The combined ethyl acetate extract was washed with water (5 mL), brine (5 mL), dried over Na2SO4 and concentrated under reduced pressure to give the title compound (0.055 g, 50%) as yellow solid. 1H NMR (DMSO-d6) δ ppm: 3.24 (s, 3H), 3.44 (t, J = 4.88 Hz, 2H), 3.57 (t, J = 4.04 Hz, 2H), 3.69 (t, J= 4.24 Hz, 2H), 4.04 (t, J= 4.04 Hz, 2H), 5.73 (d, J= 10.12 Hz, IH), 6.23 (d, J = 16.8 Hz, IH), 6.45 (dd, J= 10.12 & 16.92 Hz, IH), 6.95 (t, J = 9.4 Hz, IH), 7.28-7.30 (m, 2H), 7.42 (d, J = 8.04 Hz, IH), 7.48 (d, J = 7.48 Hz, IH), 7.67 (d, J = 14.36 Hz, IH), 7.93 (s, IH), 8.10 (d, J= 3.44 Hz, IH), 9.20 (s, IH), 9.44 (s, IH), 10.14 (s, IH); LCMS : m/e 486.1 (M+l).
EXAMPLE 248
[001275] Preparation of (S)-N-(3-(2-(4-chloro-3-(l-hydroxypropan-2-yloxy)phenylamino)- 5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-357
Figure imgf000412_0001
[001276] The title compound was prepared according to the schemes, steps and intermediates described below.
Figure imgf000412_0002
A) TBDMSCl, imidazole, CH2Cl2, O 0C, 2 h; B) DIAD, PPh3, Et3N, dry THF, rt, 1 h; C) H2, Raney Ni, MeOH, 2 h; D) Pd(OAc)2, BINAP, Cs2CO3, toluene, 110 0C, 6 h; E) TFA, CH2Cl2, rt, 1 h; F) (BOC)2O, 30 min, then K2CO3, NMP, O 0C, 15 min. [001277] Step-1
Figure imgf000413_0003
[001278] To stirred solution of 1 (1 g, 13.1 mmol) in DCM was added at 0 0C, imidazole
(0.875 g, 13.1 mmol) and tert-butyldimethylsilyl chloride (1.98 g, 13.1 mmol). The same temperature was maintained for 2 h, and then the reaction mixture was filtered and concentrated. The residue was purified by column chromatography (neutral alumina, pet ether/ethyl acetate 7/3) to give 2 (1.4 g, 56%) as a colorless liquid. [001279] Step-2
Figure imgf000413_0001
4
[001280] To a stirred solution of 2 (1.5 g, 7.89 mmol) in THF (15 mL) were added 3 (1.36 g, 7.89 mmol), PPh3 (2.27 g, 8.6 mmol) and Et3N (1.19 g, 11.1 mmol) under N2 atmosphere. The reaction mixture was cooled to 0 0C and to it was added DIAD (1.75 g, 8.6 mmol). The reaction mixture was allowed to come to rt and stirred at it for 1 h. It was quenched with water, extracted with ethyl acetate (3x5 mL) and the combined EtOAc extract was washed with water and brine solution (5 mL each). The residue obtained after concentration under reduced pressure was purified by column chromatography (SiO2, 60-120, pet ether/ethyl acetate, 7/3) to give 4 (2.1 g, 76.9%) as a yellow oil. [001281] Step-3
Figure imgf000413_0002
5
[001282] To a solution of 4 (2 g, 5.7 mmol) in methanol (20 mL)) was added Raney Ni (3 g). The reaction mixture was allowed to stir under H2 atmosphere (bladder pressure) at room temperature for 2 h. The reaction mixture was filtered through a pad of Celite® and concentrated under reduced pressure and the residue was purified by column chromatography (neutral alumina, pet ether/ ethyl acetate, 8/2) to give 5 (1.4 g, 77%) as a brownish viscous oil. [001283] Step-4
Figure imgf000414_0001
[001284] A solution of 6 (0.2 g, 0.63 mmol), prepared according to Step-1 of Example 20, 1 (0.213. g, 0.63 mmol), Pd(OAc)2 (0.014 g, 0.063 mmol), BINAP (0.0019 g, 0.031 mmol) and CS2CO3 (0.511 g, 1.5 mmol) in degassed toluene (toluene was purged with N2 for 30 min) was heated at 110 0C for 16 h under N2 atmosphere. The reaction mixture was cooled, diluted with EtOAc (20 mL), washed with water (10 mL), brine (10 mL) and dried over Na2SO4. Filtration followed by concentration under reduced pressure offered a residue which was further purified using column chromatography (SiO2, 60-120, pet ether/ethyl acetate 7/3) to give 7 (0.15 g, 38.4%) as a yellow solid. [001285] Step-5
Figure imgf000414_0002
[001286] To a stirred solution of 7 (0.15 g, 0.24 mmol) in dry CH2Cl2 (5 mL) at 0 0C was added CF3COOH (1.5 mL) and the reaction mixture was stirred at 0 0C for 30 min. The reaction was allowed to come to rt and stirred at it for 1 h. It was concentrated under reduced pressure and the residue was quenched with NaHCOs solution (3 mL) and extracted with EtOAc (3x25 mL). The combined EtOAc extract was washed with water (20 mL), brine (10 mL) and dried over Na2SO4 and concentrated under reduced pressure to give 8 (0.085 g, 86.7%) as a white solid. [001287] Step-6
Figure imgf000415_0001
1-357
[001288] A stirred solution of 8 (0.085 g, 0.21 mmol) in NMP (2.0 niL) was cooled to 0 0C and to it was added K2CO3 (0.29 g, 2.1 mmol) and acryloyl chloride (1 M solution in THF, 0.21 mL, 0.21 mmol) and the reaction mixture was stirred at 0 0C for 30 min. The reaction mixture was added dropwise to a cold, stirring solution of 10% NaHCO3. After the addition was over, the solution was stirred for another 30 min at 0 0C, and the solid was isolated by filtration through a Buchner funnel. The solid was washed with cold water, hexane and was dissolved in methanol: dichloromethane (50:50, 25 mL) and concentrated under reduced pressure. The residue obtained was suspended in cold water (3 mL), Et3N was added to it and it was extracted with ethyl acetate (2x5 mL). The combined ethyl acetate extract was washed with water (5 mL), brine (5 mL), dried over Na2SO4 and concentrated under reduced pressure to give the title compound (65 mg, 67%) as yellow solid. 1H NMR (DMSO-d6) δ ppm: 1.18 (d, J = 6.12 Hz, 3H), 3.40-3.47 (m, IH), 3.50-3.56 (m, IH), 4.20-4.30 (m, IH), 4.82 (t, J = 5.6 Hz, IH), 5.75 (dd, J = 1.88 & 10.08 Hz, IH), 6.25 (dd, J = 1.92 & 16.92 Hz, IH), 6.45 (dd, J = 10.08 & 16.92 Hz, IH), 7.12 (d, J = 8.76 Hz, IH), 7.29 (t, J = 8.08 Hz, IH), 7.40-7.44 (m, 3H), 7.52 (d, J = 8.44 Hz, IH), 7.91 (s, IH), 8.12 (d, J = 3.64 Hz, IH), 9.21 (s, IH), 9.45 (s, IH), 10.12 (s, IH); LCMS : m/e 458.0 (M+l).
E EXXAAMMPPLLEE 2 Z449?
[001289] Preparation of (R)-N-(3-(2-(4-chloro-3-(l-hydroxypropan-2-yloxy)phenylamino)-
5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-358
Figure imgf000416_0001
1-358
[001290] The title compound was prepared according to the schemes, steps and intermediates described in Example 248 by using (R)-propane-l,2-diol in place of 1 in Step-1. 1H NMR (DMSO-d6) δ ppm: 1.18 (d, J = 6.12 Hz, 3H), 3.40-3.47 (m, IH), 3.50-3.56 (m, IH), 4.20-4.30 (m, IH), 4.82 (t, J = 5.6 Hz, IH), 5.75 (dd, J = 1.88 & 10.08 Hz, IH), 6.25 (dd, J = 1.92 & 16.92 Hz, IH), 6.45 (dd, J = 10.08 & 16.92 Hz, IH), 7.12 (d, J = 8.76 Hz, IH), 7.29 (t, J = 8.08 Hz, IH), 7.40-7.44 (m, 3H), 7.52 (d, J = 8.44 Hz, IH), 7.91 (s, IH), 8.12 (d, J = 3.64 Hz, IH), 9.21 (s, IH), 9.45 (s, IH), 10.12 (s, IH); LCMS : m/e 458.0 (M+l).
EXAMPLE 250
[001291] Preparation of (E)-4-(dimethylamino)-N-(3-(5-methyl-4-(m- tolylamino)pyrimidin-2-ylamino)phenyl)but-2-enamide 1-360
Figure imgf000416_0002
[001292] The title compound was prepared according to the schemes, steps and intermediates described in Example 3 by using (E)-4-(dimethylamino)but-2-enoyl chloride place of acryloyl chloride in Step-3. 1H NMR (DMSO-d6) δ ppm: 7.91 (s, IH), 7.85 (s, IH), 7.52 (d, J = 6.4 Hz, IH), 7.45 (d, J= 7.8 Hz, IH), 7.34 (s, IH), 7.31-7.26 (m, IH), 7.21 (dd, J= 8.2, 8.0 Hz, IH), 7.01-6.92 (m, 4H); 6.27 (s, IH), 6.06 (d, J = 15.1 Hz, IH), 3.14 (d, J = 5.5 Hz, 2H), 2.37 (s, 3H), 2.31 (s, 6H), 2.13 (s, 3H); LCMS m/z All (M+l). Biological Examples
[001293] Described below are assays used to measure the biological activity of provided compounds as inhibitors of BTK, TEC, ITK, BMX, ErbBl (EGFR), ErbB2, ErbB4, and JAK3.
EXAMPLE 251 Omnia Assay Protocol for Potency Assessment Against BTK
[001294] Below describes the protocol using EGFR-WT and EGFR-T790M/L858R and the protocol BTK-optimized reagent conditions then follow.
[001295] The mechanics of the assay platform are best described by the vendor (Invitrogen,
Carlsbad, CA) on their website at the following URL: www.invitrogen.com/content.cfm?pageid=l 1338 or www.invitrogen.com/site/us/en/home/Products-and-Services/Applications/Drug- Discovery/Target-and-Lead-Identification-and-Validation/KinaseBiology/KB- Misc/Biochemical-Assays/Omnia-Kinase- Assays.html.
[001296] Briefly, 1OX stocks of EGFR-WT (PV3872) from Invitrogen and EGFR-
T790M/L858R (40350) from BPS Bioscience, San Diego, CA, 1.13X ATP (ASOOlA) and appropriate Tyr-Sox conjugated peptide substrates (KCZlOOl) were prepared in IX kinase reaction buffer consisting of 20 mM Tris, pH 7.5, 5 mM MgCl2, 1 mM EGTA, 5 mM β- glycerophosphate, 5% glycerol (1OX stock, KB002A) and 0.2 mM DTT (DSOOlA). 5 μL of each enzyme were pre-incubated in a Corning (#3574) 384-well, white, non-binding surface microtiter plate (Corning, NY) for 30 min. at 270C with a 0.5 μL volume of 50% DMSO and serially diluted compounds prepared in 50% DMSO. Kinase reactions were started with the addition of 45 μL of the ATP/Tyr-Sox peptide substrate mix and monitored every 30-90 seconds for 60 minutes at λeX360/λem485 in a Synergy4 plate reader from BioTek (Winooski, VT). At the conclusion of each assay, progress curves from each well were examined for linear reaction kinetics and fit statistics (R2, 95% confidence interval, absolute sum of squares). Initial velocity (0 minutes to ~30 minutes) from each reaction was determined from the slope of a plot of relative fluorescence units vs time (minutes) and then plotted against inhibitor concentration to estimate IC50 from log[Inhibitor] vs Response, Variable Slope model in GraphPad Prism from GraphPad Software (San Diego, CA). [001297] The modified BTK-optimized reagent conditions for the above protocol are: [BTK] = 5 nM, [ATP] = 40 niM, [Y5-Sox] = 10 mM (ATP KMapp ~ 36 niM).
EXAMPLE 252
[001298] Table 6 shows the activity of selected compounds of this invention in the BTK inhibition assay. The compound numbers correspond to the compound numbers in Table 5. Compounds having an activity designated as "A" provided an IC50 <10 nM; compounds having an activity designated as "B" provided an IC50 10-100 nM; compounds having an activity designated as "C" provided an IC50 of 100-1000 nM; compounds having an activity designated as "D" provided an IC50 of 1000-10,000 nM; and compounds having an activity designated as "E" provided an IC50 >10,000 nM.
Figure imgf000418_0001
Figure imgf000419_0001
Figure imgf000420_0001
Figure imgf000421_0001
Figure imgf000422_0001
Figure imgf000423_0001
EXAMPLE 253 BTK Ramos Cellular Assay
[001299] Compounds 1-2, 1-4, and 1-7 were assayed in Ramos human Burkitt lymphoma cells. Ramos cells were grown in suspension in T225 flasks, spun down, resuspended in 50 ml serum-free media and incubated for 1 hour. Compound was added to Ramos cells in serum free media to a final concentration of 1, 0.1, 0.01, or 0.001 μM. Ramos cells were incubated with compound for 1 hour, washed again and resuspended in lOOul serum- free media. Cells were then stimulated with lμg of goat F(ab')2 Anti-Human IgM and incubated on ice for 10 minutes to activate B cell receptor signaling pathways. After 10 minutes, the cells were washed once with PBS and then lysed on ice with Invitrogen Cell Extraction buffer. 16 μg total protein from lysates were loaded on gel and blots were probed for phosphorylation of the BTK substrate PLCγ2. Dose response inhibition of BTK signaling in Ramos cells is depicted in Figures 1, 2, 3, 4 and 5.
[001300] Table 7 shows the activity of selected compounds of this invention in the BTK
Ramos cellular inhibition assay. The compound numbers correspond to the compound numbers in Table 5. Compounds having an activity designated as "A" provided an IC50 <10 nM; compounds having an activity designated as "B" provided an IC50 10-100 nM; compounds having an activity designated as "C" provided an IC50 of 100-1000 nM; compounds having an activity designated as "D" provided an IC50 of 1000-10,000 nM; and compounds having an activity designated as "E" provided an IC50 >10,000 nM.
Figure imgf000424_0001
Figure imgf000425_0001
Figure imgf000426_0001
Figure imgf000427_0001
EXAMPLE 254
Washout Experiment with Ramos cells
[001301] Ramos cells were serum starved for one hour in RPMI media +1% glutamine at
37°C. After starvation, Ramos cells were treated with 10OnM compound diluted in serum free RPMI media for 1 hour. After compound treatment, the media was removed and cells were washed with compound-free media. Subsequently, Ramos cells were washed every 2 hours and resuspended in fresh compound-free media. Cells were collected at specified timepoints, treated with lug anti-human IgM (Southern Biotech cat # 2022-01) for 10 minutes on ice to induce BCR signaling and then washed in PBS. Ramos cells were then lysed in Cell Extraction Buffer (Invitrogen FNNOOl 1) supplemented with Roche complete protease inhibitor tablets (Roche 11697498001) and phosphatase inhibitors (Roche 04 906 837 001) and 18ug total protein lysate was loaded in each lane. Inhibition of BTK kinase activity was assayed by measuring its substrate (PLCγ2) phosphorylation by western blot with phospho-specific antibodies from Cell Signaling Technologies cat# 3871. The results of this experiment with compounds 1-2, 1-4 and I- 7 are depicted in Figures 1, 2 and 3.
[001302] Table 8 provides data for selected compounds in the Ramos washout assay.
Figure imgf000427_0002
Figure imgf000428_0001
EXAMPLE 255 Mass Spectrometry for BTK
[001303] Intact BTK was incubated for 1 hr at a 1OX fold excess of 1-7 to protein. Aliquots
(2 μl) of the samples were diluted with 10 μl of 0.1% TFA prior to micro C4 ZipTipping directly onto the MALDI target using Sinapinic acid as the desorption matrix (10 mg/ml in 0.1% TFA:Acetonitrile 20:80). See Figure 15. Top panel shows the mass spec trace of the intact BTK protein (m/z 81,032 Da). The bottom panel shows mass spec trace when BTK was incubated with 1-7 (mw=345.4). The centroid mass (m/z= 81,403 Da) shows a positive shift of about 371.1 Da indicating complete modification of BTK by 1-7. Other compounds that completely modify BTK include 1-96, 1-71, 1-149, 1-161, 1-163, 1-182, 1-195, 1-207, 1-219, and 1-244.
EXAMPLE 256 Human primary B cell proliferation assay
[001304] Human naϊve B cells were purified from 100 mL whole blood using a MACS purification kit designed to isolate CD 19+, IgD+ cells by negative selection. Purified naϊve B cells were resuspended in RPMI complete and stimulated with 5μg/ml α-IgM for 72 hours. 3H- Thymidine was included in the culture media for the final 16h, cells were harvested and 3H incorporation measured. Inhibition of B cell proliferation correlates with inhibition of BTK substrate phosphorylation after α-IgM stimulation. Importantly, a molecule with the same scaffold as 1-7 but biochemically inactive against BTK, IR-7, is not active in the naϊve B cell proliferation assay.
Figure imgf000428_0002
EXAMPLE 257 B cell lymphoma proliferation assay
[001305] Provided compounds inhibit proliferation of various B cell lymphoma cell lines, as shown in Table 10. The compound numbers correspond to the compound numbers in Table 5. Compounds having an activity designated as "A" provided an EC50 <0.1 μM; compounds having an activity designated as "B" provided an EC50 0.1-1 μM; compounds having an activity designated as "C" provided an EC50 of 1-10 μM; and compounds having an activity designated as "D" provided an EC50 of >10 μM.
Figure imgf000429_0001
Figure imgf000430_0001
Figure imgf000431_0001
EXAMPLE 258 In vivo Thymus-Independent (TI-2) B cell Activation
[001306] C57/B6 mice were dosed daily with 100 mg/kg of the appropriate compound on day 0 through 5. Mice were immunized once with 25 μg TNP-Ficoll on day 1, serum was collected on day 6 and analyzed for circulating α-TNP IgM (1 :1600 serum dilution) and IgG3 (1 :200 serum dilution) antibody production by ELISA. Results represent the average of 10 mice per treatment group and are given in Table 11 as % inhibition of TI-2 independent B cell activation.
Figure imgf000431_0002
EXAMPLE 259 Collagen Antibody Induced Arthritis Model
[001307] On day 0 baseline footpad measurements were made and animals were distributed to the experimental groups in such a way as to generate groups with no significant differences between the groups. Each animal was then inoculated intravenously with 2 mg Arthritomab monoclonal antibody cocktail. Treatment with test agents began at this time. On day 6, each animal was injected intraperitoneally with 50 μg LPS in 200 μl of sterile PBS. Footpad measurements and clinical scoring were conducted on days 6, 7, 8, 9, 10, 11, 12, 14, 18, and 21. Table 12 shows the results.
Figure imgf000432_0001
EXAMPLE 260 PG-PS Arthritis Model
[001308] On day 0, female Lewis rats received an intraperitoneal (IP) bolus of peptidoglycan-polysaccharide (PG-PS) in an amount of 15μg/g rat body weight. Baseline control rats received an IP bolus of PBS. The vehicle and treatment groups were dosed via oral gavage just prior to PG-PS administration. Treatment with vehicle and compound continued each day through day 22. Maximal lateral ankle width measurements of both rear limbs were collected with a caliper throughout the study. On day 23, study was terminated and the final change in ankle swelling was calculated and compared to vehicle controls. Table 13 shows the results for two compounds (n = number of experiments).
Figure imgf000432_0002
EXAMPLE 261 Mass Spectrometry for TEC Kinase (Compound 1-2)
[001309] TEC kinase (45 pmols; Invitrogen) was incubated with (1-2) (450 pmols) for 3 hrs at 1OX excess prior to tryptic digestion. Iodoacetamide was used as the alkylating agent after compound incubation. A control sample (45 pmols) was also prepared which did not have the addition of (1-2). For tryptic digests a 5ul aliquot (7.5 pmols) was diluted with 15 ul of 0.1%
TFA prior to micro Cl 8 Zip Tipping directly onto the MALDI target using alpha cyano-4- hydroxy cinnamic acid as the matrix (5mg/ml in 0.1% TFA:Acetonitrile 50:50).
[001310] As depicted in Figure 6, the expected peptide (GCLLNFLR) to be modified was immediately evident after reaction with 1-2 (MI mass of 359.17 Da) at MH+ of 1294.72. The peptide was also quite evident in the control sample as modified by Iodacetamide at MH+ of
992.56. Interestingly the iodoacetamide modified peptide was not evident in the digest reacted with compound 1-2 indicating that the reaction was complete. There was no evidence of other modified peptides.
[001311] Evidence of compound 1-2 was observed at MH+ of 360.17 in the low mass range of the spectra. The fragmentation spectra of the 360.17 peak did show diagnostic fragments that were apparent in the PSD spectra of the modified peptide at 1294.72 (See Figure 6).
[001312] To further verify the presence of the modified peptides, both the iodoacetamide labeled (992.56) and 1-2 labeled (1294.72) were subjected to PSD (MS/MS) analsysis. After a database search of the NCBI nr Homo sapien database using Mascot MS/MS Ion Search program the top match was the expected peptide in both cases.
Instrumental:
[001313] For tryptic digests the instrument was set in Reflectron mode with a pulsed extraction setting of 2200. Calibration was done using the Laser Biolabs Pep Mix standard
(1046.54, 1296.69, 1672.92, 2093.09, 2465.20). For CID/PSD analysis the peptide was selected using cursors to set ion gate timing and fragmentation occurred at a laser power about 20% higher and He was used as the collision gas for CID. Calibration for fragments was done using the P14R fragmentation calibration for the Curved field Reflectron. EXAMPLE 262
Mass Spectrometry for TEC Kinase (Compound 1-4)
[001314] TEC kinase (45 pmols; Invitrogen) was incubated with (1-4) (450 pmols) for 3hrs at 1OX excess prior to tryptic digestion. Iodoacetamide was used as the alkylating agent after compound incubation. A control sample (45 pmols) was also prepared which did not have the addition of (1-4). For tryptic digests a 5ul aliquot (7.5 pmols) was diluted with 15 ul of 0.1% TFA prior to micro Cl 8 Zip Tipping directly onto the MALDI target using alpha cyano-4- hydroxy cinnamic acid as the matrix (5mg/ml in 0.1%TFA:Acetonitrile 50:50). [001315] As depicted in Figure 7, the expected peptide (GCLLNFLR) to be modified was immediately evident at MH+ of 1355.72. This is the mass to be expected when compound 1-4, with an adduct mass of 420.21, is added to the peptide mass of 935.51. The peptide was also quite evident in the control sample as modified by Iodacetamide at MH+ of 992.56. Interestingly the iodoacetamide modified peptide was not evident in the digest reacted with compound 1-4 indicating that the reaction was complete. There was no evidence of other modified peptides.
[001316] Evidence of compound 1-4 was observed at MH+ of 421.35 in the low mass range of the spectra. The fragmentation spectra of the 421.35 peak did reveal two prominent peaks that were apparent in the PSD spectra of the modified peptide at 1355.72 (See Figure 7). [001317] To further verify the presence of the modified peptide with compound 1-4, the peptide at MH+ of 1355.72 was subjected to PSD (MS/MS) analsysis. Because of the low intensity of fragments, a database correlation was not possible. However, diagnostic fragments from the 1-4 molecule itself provided confidnece in the identification. Diagnostic fragments at MH+ of 376.38 and 421.83 are from 1-4. Instrumental: .
[001318] For tryptic digests the instrument was set in Reflectron mode with a pulsed extraction setting of 1800. Calibration was done using the Laser Biolabs Pep Mix standard (1046.54, 1296.69, 1672.92, 2093.09, 2465.20). For CID/PSD analysis the peptide was selected using cursors to set ion gate timing and fragmentation occurred at a laser power about 20% higher and He was used as the collision gas for CID. Calibration for fragments was done using the P14R fragmentation calibration for the Curved field Reflectron. EXAMPLE 263
Mass Spectrometry for TEC Kinase (Compound 1-7)
[001319] TEC kinase (45 pmols; Invitrogen) was incubated with (1-7) (450 pmols) for 3hrs at 1OX excess prior to tryptic digestion. Iodoacetamide was used as the alkylating agent after compound incubation. A control sample (45 pmols) was also prepared which did not have the addition of (1-7). For tryptic digests a 5ul aliquot (7.5 pmols) was diluted with 15 ul of 0.1% TFA prior to micro Cl 8 Zip Tipping directly onto the MALDI target using alpha cyano-4- hydroxy cinnamic acid as the matrix (5mg/ml in 0.1%TFA:Acetonitrile 50:50). [001320] As depicted in Figure 8, the expected peptide (GCLLNFLR) to be modified was immediately evident at MH+ of 1280.73. This is the mass to be expected when compound 1-7, with an adduct mass of 345.16, is added to the peptide mass of 935.51. The peptide was also quite evident in the control sample as modified by Iodacetamide at MH+ of 992.56. Interestingly the iodoacetamide modified peptide was not evident in the digest reacted with compound 1-7 indicating that the reaction was complete. There was no evidence of any other modified peptide at MH+ of 1985.93 (TIDEL VECEETFGR).
[001321] Evidence of compound 1-7 was observed at MH+ of 346.32 in the low mass range of the spectra. The fragmentation spectra of the 346.32 peak did show many diagnostic fragments that were apparent in the PSD spectra of the two modified peptides (See Figure 8). [001322] To further verify the presence of the modified peptides with compound 1-7, the peptides at MH+ of 1280.73 and 1985.93 were subjected to PSD (MS/MS) analsysis. A correlational analysis with the homosapien database identified the correct peptide modified by I- 7.
Instrumental:
[001323] For tryptic digests the instrument was set in Reflectron mode with a pulsed extraction setting of 2200. Calibration was done using the Laser Biolabs Pep Mix standard (1046.54, 1296.69, 1672.92, 2093.09, 2465.20). For CID/PSD analysis the peptide was selected using cursors to set ion gate timing and fragmentation occurred at a laser power about 20% higher and He was used as the collision gas for CID. Calibration for fragments was done using the P14R fragmentation calibration for the Curved field Reflectron. EXAMPLE 264 Omnia Assay Protocol for Potency Assessment Against Active Forms of ITK Kinase
[001324] This example describes continuous-read kinase assays to measure inherent potency of compound against active forms of ITK enzymes as described in Example 251 above except that the modified ITK-optimized reagent conditions are:
[ITK] = 10 nM, [ATP] = 25 μM, [Y6-Sox] = 10 μM (ATP KMaPP = 33 μM).
EXAMPLE 265
[001325] Table 14 shows the activity of selected compounds of this invention in the ITK inhibition assay. The compound numbers correspond to the compound numbers in Table 5. Compounds having an activity designated as "A" provided an IC50 <10 nM; compounds having an activity designated as "B" provided an IC50 10-100 nM; compounds having an activity designated as "C" provided an IC50 of 100-1000 nM; compounds having an activity designated as "D" provided an IC50 of 1000-10,000 nM; and compounds having an activity designated as "E" provided an IC50 >10,000 nM.
Figure imgf000436_0001
Figure imgf000437_0001
Figure imgf000438_0001
Figure imgf000439_0001
EXAMPLE 266 Omnia Assay Protocol for Potency Assessment Against Active Forms of BMX Kinase
[001326] This example describes continuous-read kinase assays to measure inherent potency of compound against active forms of BMX enzymes as described in Example 251 above except that the modified BMX-optimized reagent conditions are: [BMX] = 2.5 nM, [ATP] = 100 μM, [Y5-Sox] = 7.5 μM (ATP KMaPP = 107 μM).
EXAMPLE 267
[001327] Table 15 shows the activity of selected compounds of this invention in the BMX inhibition assay. The compound numbers correspond to the compound numbers in Table 5. Compounds having an activity designated as "A" provided an IC50 <10 nM; compounds having an activity designated as "B" provided an IC50 10-100 nM; compounds having an activity designated as "C" provided an IC50 of 100-1000 nM; compounds having an activity designated as "D" provided an IC50 of 1000-10,000 nM; and compounds having an activity designated as "E" provided an IC50 >10,000 nM.
Figure imgf000440_0001
EXAMPLE 268
Cloning, Expression and Purification of EGFR-WT and EGFR C797S Mutant Using Baculovirus and Insect Cells (i) Subclonins of EGFR-WT and mutant kinase domains
[001328] Amino acids 696 to 1022 of the EGFR-WT kinase domain (NM 005228, NP 005219.2) was subcloned into the Ncol and HindIII sites of the pFastHTa vector (Invitrogen, Carlsbad, CA). To make the EGFR-mutant protein, the cysteine at position 797 was changed to a serine using the Stratagene QuikChange kit (Stratagene, Cedar Creek, TX), according to manufacturer's instructions. (H) Expression
[001329] Pl baculovirus stocks were generated in SF9 cells via Blue Sky Biotech's suspension transfection protocol (Worcester, MA). Expression analysis was conducted in 125 ml culture of SF21 insect cells ((grown in SF900I SFM (Invitrogen cat # 10902-088), supplemented with 10mg/L gentamicin (Invitrogen, Carlsbad, CA, cat# 15710-064)) using a viral load of 0.1ml of virus per 100 ml of cell suspension. Expression was optimized using Blue Sky Biotech's Infection Kinetics Monitoring system (Worcester, MA). (Hi) Purification
[001330] Infected insect cells were pelleted. Cell pellets were resuspended in Blue Sky Biotech's lysis buffer (Worcester, MA, IX WX; solubilization buffer, containing a protease inhibitor cocktail of leupeptin, pepstatin, PMSF, aprotinin and EDTA) at a ratio of 10 ml per gram of wet cell paste. Cells were lysed by sonication and the lysate was clarified by centrifugation at 9,000 RPM for 30 minutes in a GSA rotor. 500 μl bed volume of NiNTA resin (Qiagen, Valencia, CA) was added to the supernatants and batch bound for two hours with constant agitation. The material was transferred by gravity into an empty 2ml column. The column was washed with 2 ml of wash buffer (Blue Sky Biotech, Worcester, MA, IX WX, 25mM imidazole ).The protein was eluted with IX WX + imidazole at varying concentrations: Elution 1 : 75 mM imidazole ( 2 fractions, 1 column volume); Elution 2 : 150 mM imidazole (2 fractions, 1 column volume); Elution 3 : 300 mM imidazole (2 fractions, 1 column volume). All the elution fractions were analyzed by SDS page followed by Coomassie staining and Western Blotting using anti-penta-his antibody (Qiagen, Valencia, CA). The carboxy-terminal six- histidine "tag" was removed from some of the purified protein using AcTEV Protease kit ( Invitrogen, Carlsbad, CA, Cat# 12575-015), following manufacturer's instructions. All the samples (pre- and post- Tev cut) were analyzed by SDS page followed by Coomassie staining and Western Blotting, as described above.
EXAMPLE 269 Mass Spectrometry for EGFR
[001331] EGFR wild type and EGFR (mutant C797S) is incubated with 10-fold excess of test compound for 1 hr and 3 hrs. 1 μl aliquots of the samples (total volume 5-8 ul) are diluted with 10 ul of 0.1% TFA prior to micro C4 ZipTipping directly onto the MALDI target using Sinapinic acid as the desorption matrix (10 mg/ml in 0.1%TFA:Acetonitrile 50:50). Intact mass measurement reveals that the wild type has a nominal mass of about 37557 and the mutant slightly lower at 37500. Reactivity is only observed for the wild type EGFR with a new peak appearing at a mass consistent with a single site covalent modification with test compound which has a mass of 410 Da.
EXAMPLE 270
Omnia Assay Protocol for Potency Assessment Against EGFR (WT) and EGFR (T790M/L858R) Active Enzymes
[001332] The Omnia Assay Protocol for potency assessment against EGFR is performed as described in Example 251 above except that the EGFR-WT- and EGFR T790M/L858R-modifϊed optimized reagent conditions are:
[001333] [EGFR-WT] = 5 nM, [ATP] = 15 mM, [Y12-Sox] = 5 mM (ATP KMapp ~ 12 mM); and [EGFR-T790M/L858R] = 3 nM, [ATP] = 50 mM, [Y12-Sox] = 5 mM (ATP KMapp ~ 45 mM).
EXAMPLE 271
[001334] Tables 16 and 17 show the activity of selected compounds of this invention in the
EGFR inhibition assay. Table 16 shows wild-type EGFR data; Table 17 shows data for two EGFR mutants. The compound numbers correspond to the compound numbers in Table 5. Compounds having an activity designated as "A" provided an IC50 <10 nM; compounds having an activity designated as "B" provided an IC50 10-100 nM; compounds having an activity designated as "C" provided an IC50 of 100-1000 nM; compounds having an activity designated as "D" provided an IC50 of 1000-10,000 nM; and compounds having an activity designated as "E" provided an IC50 >10,000 nM.
Figure imgf000443_0001
Figure imgf000444_0001
Figure imgf000445_0001
Figure imgf000446_0001
Figure imgf000447_0001
Figure imgf000448_0001
Figure imgf000448_0002
Figure imgf000449_0001
EXAMPLE 272 Cellular Assays for EGFR Activity
[001335] Compounds were assayed in A431 human epidermoid carcinoma cells using a method substantially similar to that described in Fry, et al., Proc. Natl. Acad. Sci. USA VoI 95, pp 12022-12027, 1998. Specifically, A431 human epidermoid carcinoma cells were grown in 6- well plates to 90% confluence and then incubated in serum-free media for 18 hr. Duplicate sets of cells were treated with 1 μM designated compound for 2, 5, 10, 30, or 60 min. Cells were washed free of the compound with warmed serum-free medium, incubated for 2 hr, washed again, incubated another 2 hr, washed again, and then incubated another 2 hr washed again and incubated for additional 2 hr and then stimulated with 100 ng/ml EGF for 5 min. Extracts were made as described Fry, et al.
[001336] Compounds were assayed in A431 human epidermoid carcinoma cells using a method substantially similar to that described in Fry, et al. Specifically, A431 human epidermoid carcinoma cells were grown in 6-well plates to 90% confluence and then incubated in serum- free media for 18 hr. Cells were then treated with 10, 1, 0.1, 0.01, or 0.00 lμM test compound for 1 hr. Cells were then stimulated with 100 ng/ml EGF for 5 min, and extracts were made as described in Fry, et al. 20ug total protein from lysates were loaded on gel and blots were probed for either EGFR phosphorylation or p42/p44 Erk phosphorylation.
EXAMPLE 273 Washout Experiment for EGFR Activity
[001337] A431 human epidermoid carcinoma cells were grown in 6-well plates to '90% confluence and then incubated in serum- free media for 18 hr. Duplicate sets of cells were treated with 1 μM designated compound for 1 hr. One set of cells was then stimulated with 100 ng/ml EGF for 5 min, and extracts were made as described. The other set of cells was washed free of compound 1-7 with warmed compound-free medium, incubated for 2 hr, washed again, incubated another 2 hr, washed again, and then incubated another 2 hr washed again and incubated for additional 2 hr and then stimulated with EGF. The results of this experiment with compound 1-7 are depicted in Figure 10.
EXAMPLE 274 Washout Experiment in HCC827 cells containing EGFR deletion mutantHCC827
[001338] Cells (ATCC, Manassas,VA) were plated in Growth Media (RPMI 1640) supplemented with 10% FBS,10uM HEPES, 2mM 1-glutamine, ImM NaPyruvate and pen/strep (Invitrogen, Carlsbad, CA) at a density of 2.5 x 10 cells per well in 6 well tissue culture plates. Twenty four hours later the cells were washed 2 x with PBS then serum starved overnight in Basal Media (Growth Media without FBS).
[001339] The following morning the media was removed and 2 ml fresh Basal Media containing IuM compound in 0.1% DMSO was added to duplicate wells. At 1 hour, one well of cells was treated with 100 ng/ml of EGF for 5 minutes, rinsed with PBS, then lysed by scraping into 75 ul of Cell Extraction Buffer (Invitrogen, Carlsbad, CA) plus PhosSTOP Phosphatase Inhibitor and Complete Protease Inhibitor (Roche, Indianapolis, IN) for the Oh time point. The compound was removed from the second set of wells and they were washed 2X with Basal Media. The cells were washed with Basal Media every 2 hours until 8 hours when they were treated with EGF and lysed as at the 0 h time point.
[001340] Lysate protein concentrations were determined by BCA Assay (Pierce, Rockford,
IL) and 10 ug of each lysate was separated by 4-12% gradient SDS-PAGE (Invitrogen), transferred to Immobilon-FL membrane (Millipore) and probed with rabbit anti-Phospho-EGFR (TyrlO68) (Zymed-now Invitrogen) and mouse anti-EGFR (Cell Signaling Technologies, Danvers, MA) antibodies. Phospho-protein signals were quantitated using Odyssey Infrared Imagning (Li-Cor Biosciences, Lincoln, Nebraska). The results of this experiment are depicted in Figure 9 where it shows compound 1-2 compared to results of compound 1-4 and compound I- 7 in the same "washout" experiment. EXAMPLE 275
Mass Spectrometry for ERBB4
[001341] Erbb4 kinase domain (Upstate) was incubated with compound for 60 minutes at
10-fold excess of compound 1-4 and 1-11 to protein. 1 μl aliquots of the samples (total volume of 4.24ul) were diluted with 10 μl of 0.1% TFA prior to micro C4 ZipTipping directly onto the MALDI target using Sinapinic acid as the desorption matrix (10 mg/ml in 0.1%TFA:Acetonitrile 50:50). For intact protein mass measurement the instrument was set in Linear mode using a pulsed extraction setting of 16,952 for the myoglobin standard used to calibrate the instrument (Shimadzu Axima TOF2).
[001342] Intact ErbB4 protein occurs at MH+ of 35850 with corresponding sinapinic
(matrix) adducts occurring about 200 Da higher. A stochiometric incorporation of the test compound (1-4 and 1-11) (Mw of 410 Da) produced a new mass peak which is approximately 410 Da higher (MH+ of 36260). This is consistent with covalent modification of ErbB4 with compounds 1-4 and 1-11.
EXAMPLE 276
ErbBl, ErbB2 and/or ErbB4 Kinase Inhibition
[001343] Compounds of the present invention were assayed as inhibitors of one or more of
ErbBl, ErbB2, and/or ErbB4 in a manner substantially similar to the method described by Invitrogen Corp (Invitrogen Corporation, 1600 Faraday Avenue, Carlsbad, California, CA; http://www.invitrogen. com/do wnloads/Z-LYTE_Brochure_ 1205.pdf) using the Z'-LYTE™ biochemical assay procedure or similar biochemical assay. The Z'-LYTE™ biochemical assay employs a fluorescence-based, coupled-enzyme format and is based on the differential sensitivity of phosphorylated and non-phosphorylated peptides to proteolytic cleavage. Using this assay, Compound 1-56 was found to inhibit ERBBl with an IC50 of 2,233 nM. Using this assay, Compound 1-56 was found to inhibit ERBB4 (HER4) with an IC50 of 2,165 nM.
EXAMPLE 277
Mass Spectrometry for Janus-3 Kinase ( JAK3)
[001344] JAK3 kinase (33 pmols; Invitrogen) was incubated with (1-7) (327 pmols) for
3hrs at 1OX excess prior to tryptic digestion. Iodoacetamide was used as the alkylating agent after compound incubation. For tryptic digests a 5ul aliquot (5.5 pmols) was diluted with 15 ul of 0.1% TFA prior to micro Cl 8 Zip Tipping directly onto the MALDI target using alpha cyano-
4-hydroxy cinnamic acid as the matrix (5mg/ml in 0.1%TFA:Acetonitrile 50:50).
[001345] As depicted in Figure 11, the expected peptide (LVMEYLPSGCLR) to be modified was immediately evident as the largest peak at MH+ of 1725.88. This is the mass to be expected when compound 1-7, with an adduct mass of 345.16, is added to the peptide mass of
1380.70. Interestingly the iodoacetamide modified peptide was not evident at MH+ of 1437.73 in the digest reacted with compound 1-7 indicating that the reaction was not entirely complete.
There was also evidence for a number of other modified peptides, however, their signals were low.
[001346] Evidence of compound 1-7 was observed at MH+ of 346.12 in the low mass range of the spectra. The fragmentation spectra of the 346.12 peak did not show diagnostic fragments that were apparent in the PSD spectra of the modified peptides (See Figure 11).
[001347] To further verify the presence of the modified peptides with compound 1-7, the peptides at MH+ of 1725.88 and 1118.55 were subjected to PSD (MS/MS) analsysis. A correlational analysis with the homosapien database identified the correct peptides as being modified by 1-7. Compound 1-11 was also tested using the same procedure and showed measurable modification.
Instrumental:
[001348] For tryptic digests the instrument was set in Reflectron mode with a pulsed extraction setting of 2200. Calibration was done using the Laser Biolabs Pep Mix standard
(1046.54, 1296.69, 1672.92, 2093.09, 2465.20). For CID/PSD analysis the peptide was selected using cursors to set ion gate timing and fragmentation occurred at a laser power about 20% higher and He was used as the collision gas for CID. Calibration for fragments was done using the P14R fragmentation calibration for the Curved field Reflectron.
EXAMPLE 278 Omnia Assay Protocol for Potency Assessment Against the Active Form of JAK3:
[001349] The Omnia Assay Protocol for potency assessment against JAK3 was performed in a substantially similar manner as that described in Example 251 above except that the modified J AK3 -optimized reagent conditions were:
[JAK3] = 5 nM, [ATP] = 5 μM, [Y12-Sox] = 5 μM (ATP KMapp ~5 μM). EXAMPLE 279
[001350] Table 18 shows the activity of selected compounds of this invention in the JAK3 inhibition assay. The compound numbers correspond to the compound numbers in Table 5. Compounds having an activity designated as "A" provided an IC50 <10 nM; compounds having an activity designated as "B" provided an IC50 10-100 nM; compounds having an activity designated as "C" provided an IC50 of 100-1000 nM; compounds having an activity designated as "D" provided an IC50 of 1000-10,000 nM; and compounds having an activity designated as "E" provided an IC50 >10,000 nM.
Figure imgf000453_0001
Figure imgf000454_0001
EXAMPLE 280 JAK3 Cellular Assay Protocol in CTLL2 Cells
[001351] Compounds 1-2, 1-4 and 1-7 were tested in the following protocol.
CTLL2: murine lymphoma cell line ATCC: TIB-214. 5xlO6 cells/sample were IL-2 starved in RPMI- 1640 media for 2 hours. Designated samples were then treated with compound for 90 minutes. Samples, except DMSO control were then stimulated with 10OnM IL-2 for 10 minutes. Samples were lysed and subjected to Western Analysis. The results are displayed in Figure 12, Figure 13 and Figure 14.
EXAMPLE 281 BTK occupancy in Ramos cells with 1-7 and 1-215 using strep tavidin beads
[001352] Ramos cells were incubated with 0.1, 0.05, 0.01, or 0.001 μM 1-7 in serum free media for 1 hour at 37 0C. The cells were pelleted by centrifugation and lysed in Cell Extraction buffer (Invitrogen) for 10 minutes on ice, centrifuged (10 minutes at 14,000 rpm) and the supernatant was collected. Cell lysates were incubated with 1 μM 1-215 for 1 hour at room temperature, then incubated with streptavidin-coupled agarose beads (ThermoFisher) overnight at 4 0C. The beads were washed three times with lysis buffer and the bound proteins were boiled off the beads at 95 0C for 5 minutes in 4X LDS Sample Buffer. The amount of BTK associated with the probe 1-215 was assessed by BTK western blot. All values were normalized to the DMSO-treated sample which is set to 100%. Figure 16 shows the western blot; Figure 17 shows quantitation of Figure 16 demonstrating unoccupied BTK protein is available to the probe 1-215 when the cells have been exposed to low concentrations (10 nM, 1 nM) of 1-7 but at higher concentrations of 1-7 the BTK protein is fully occupied and cannot interact with 1-215. EXAMPLE 282 Washout Experiment with 1-7 and probe compound 1-215
[001353] Ramos cells were incubated with 0.1 μM 1-7 or a reversible BTK inhibitor control compound in serum free media for 1 hour at 37 0C. The cells were then washed in compound- free media and lysed 0, 4, 6, or 8 hours after compound removal. Cell lysates were incubated with 1 μM 1-215 for 1 hour at room temperature, then overnight at 4 0C with streptavidin- coupled agarose beads. Protein was boiled off the beads and BTK association was assessed by western blot. Figure 18 shows the Western blot; Figure 19 shows the quantitation of Figure 18 and demonstrates all the BTK protein remains occupied by 1-7 for over 8 hours. This suggests that the timeframe for re-synthesis of detectable BTK protein in Ramos cells is greater than 8 hours. In contrast, with the reversible inhibitor control, 45% of BTK protein is unbound and available to the probe at 0 hours and by 4 hours 100% of BTK protein is unbound and available to bind the probe. All samples were normalized to the DMSO-treated cells harvested at 0 hours.
EXAMPLE 283 Measuring BTK occupancy from in vitro samples by ELISA
[001354] In order to determine the amount of free BTK in cell or tissue lysates, an ELISA protocol was employed that utilizes a biotinylated probe compound that binds only to free, unoccupied BTK. The conjugated biotin is captured on a streptavidin-coated ELISA plate and detected with a mouse anti-BTK antibody (Becton Dickinson, Franklin Lakes, NJ, USA) and a secondary goat anti-mouse HRP antibody (Zymed, South San Francisco, CA, USA). [001355] All samples were prepared with equal concentrations of Biorad lysis buffer (Hercules, CA, USA), 0.5% bovine serum albumin in PBS with 0.05% Tween-20 to give a final concentration of 1 μM 1-215. Samples were incubated in a mixing plate for 1 hr at room temperature while shaking to allow probe compound 1-215 to bind to free BTK. After incubation with 1-215, samples were added to a washed, streptavidin-coated ELISA plate (Pierce, Rockford, IL, USA) and incubated for 1 hr at room temperature while shaking. The plate was then washed with PBS containing 0.05% Tween-20 using an automatic plate washer. Anti-BTK antibody was prepared at 1 : 1000 dilution in 0.5% BSA in PBS (0.05% Tween-20) and added to the ELISA plate. The plate was incubated for 1 hr at room temperature while shaking. The plate was washed as described above and the secondary HRP antibody was prepared at 1 : 5000 dilution in 0.5% BSA in PBS (0.05% Tween-20). The plate was incubated and washed as described above. TMB was added to the plate, and OD650 was monitored until reaching 1 OD unit. The reaction was then stopped with addition of H2SO4. The plate was analyzed using Gen 5 software, and a 4 Parameter Logistic curve was employed to quantitate samples. Recombinant BTK (Invitrogen, Carlsbad, CA, USA) was used for the standard curve.
[001356] Table 19 shows results with Ramos cells reported as concentration at which >50% or >90% of BTK is occupied. A concentration designated as "A" is greater than 1 nM; a concentration designated as "B" is greater than 10 nM; and a concentration designated as "C" is greater than 50 nM.
Figure imgf000456_0001
EXAMPLE 284 Human primary B cell covalent probe occupancy in vitro
[001357] Human primary B cells were isolated as described in Example 256, then resuspended in RPMI media (10% serum). The compound to be analyzed was added at a 1 : 1000 dilution to media. Cells were incubated with compound in a tissue culture incubator for 1 h at 37 0C. After incubation, the cells were pelleted, washed with IX PBS, and lysed on ice for 45 min with occasional agitation. Samples were spun in a chilled microcentrifuge for 30 min at 14,000 rpm and the supernatant was isolated. The supernatant was analyzed as described in Example 283 using 1-215. 1-96 and 1-182 occupied at least 50% of BTK at concentrations greater than 10 nM.
EXAMPLE 285 Dog primary B cell covalent probe occupancy in vitro
[001358] Canine whole blood (30 mL) was diluted to 50 mL total with IX PBS and layered on top of Histopaque-1077 (Sigma Aldrich). The whole blood-Histopaque was spun at 400 x g for 30 min in a Beckman centrifuge with no brake. Peripheral blood mononuclear cells (PBMCs) were collected and pelleted at 400 x g for 15 min. Red blood cells (RBCs) were lysed with 2.5 niL RBC lysis buffer (Boston Bioproducts) and the remaining PBMCs were washed 3 times in IX PBS at 250 x g. PBMCs were treated with compound at a 1 :1000 dilution for one hour at 37 0C, washed with PBS and lysed on ice for 45 minutes. The lysate was centrifuged for 30 minutes at 14,000 x g and the supernatant collected. The supernatant was analyzed as described in Example 283 using 1-215. 1-96 occupied at least 50% of BTK at concentrations greater than 10 nM.
EXAMPLE 286 Measuring BTK occupancy from in vivo samples by ELISA
[001359] Rats were dosed orally with 30 mg/kg of compound and spleens were harvested either 2 or 24 hours after compound treatment. Rat spleens were disrupted between two microscope slides coated with frosted glass to recover single cell suspensions. Red blood cells were lysed by incubating with RBC lysis buffer (Boston BioProducts) for 2 minutes at room temperature, the cells were then resuspended in RPMI complete media and pelleted by centrifugation. Rat B cells were isolated by positive selection with B220+ antibody-magnetic bead conjugates, purified by MACS column and lysed in Bio-Rad lysis buffer at a concentration of 10 million cells/100 μl. Lysates were analyzed employing the biotinylated probe compound I- 215 in an ELISA protocol as described in detail in Example 278. Table 20 shows the results.
Figure imgf000457_0001
EXAMPLE 286 Proteomics analysis
[001360] Proteins that are covalently bound to 1-215 in a cell lysate are identified using mass spectrometry. Cell lysate is incubated with 1 μM 1-215 for 1 hour at room temperature, followed by the addition of streptavidin-coupled agarose beads. Mass spectrometry is used to identify proteins other than BTK. These are potential "off-target" interactions. [001361] While a number of embodiments of this invention are described herein, it is apparent that the basic examples may be altered to provide other embodiments that utilize the compounds and methods of this invention. Therefore, it will be appreciated that the scope of this invention is to be defined by the appended claims rather than by the specific embodiments that have been represented by way of example.

Claims

CLAIMSWe claim:
1. A compound of formula I-a or I-b:
Figure imgf000459_0001
I-a I-b or a pharmaceutically acceptable salt thereof, wherein:
Ring A is an optionally substituted group selected from phenyl, a 3-7 membered saturated or partially unsaturated carbocyclic ring, an 8-10 membered bicyclic saturated, partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
Ring B is an optionally substituted group selected from phenyl, a 3-7 membered saturated or partially unsaturated carbocyclic ring, an 8-10 membered bicyclic saturated, partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
R1 is a warhead group; Ry is hydrogen, halogen, -CN, -CF3, Ci_4 aliphatic, Ci_4 haloaliphatic, -OR, -C(O)R, or
-C(O)N(R)2; each R group is independently hydrogen or an optionally substituted group selected from Ci_6 aliphatic, phenyl, a 4-7 membered heterocylic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; W1 and W2 are each independently a covalent bond or a bivalent Ci_3 alkylene chain wherein one methylene unit of W1 or W2 is optionally replaced by -NR2-, -N(R2)C(0)-, -C(O)N(R2)-, -N(R2)SO2-, -SO2N(R2)-, -0-, -C(O)-, -OC(O)-, -C(O)O-, -S-, -SO- or -SO2-; R2 is hydrogen, optionally substituted Ci_6 aliphatic, or -C(O)R, or:
R2 and a substituent on Ring A are taken together with their intervening atoms to form a 4-6 membered partially unsaturated or aromatic fused ring; or R2 and Ry are taken together with their intervening atoms to form a 4-6 membered saturated, partially unsaturated, or aromatic fused ring; m and p are independently 0-4; and
Rx and Rv are independently selected from -R, halogen, -OR, -0(CH2)q0R, -CN, -NO2, -SO2R, -SO2N(R)2, -SOR, -C(O)R, -CO2R, -C(O)N(R)2, -NRC(O)R, -NRC(O)N(R)2, -NRSO2R, or -N(R)2, wherein q is 1-4; or:
Rx and R1 when concurrently present on Ring B are taken together with their intervening atoms to form a 5-7 membered saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with a warhead group and 0-3 groups independently selected from oxo, halogen, CN, or Ci_6 aliphatic; or
Rv and R1 when concurrently present on Ring A are taken together with their intervening atoms to form a 5-7 membered saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with a warhead group and 0-3 groups independently selected from oxo, halogen, CN, or Ci_6 aliphatic.
2. The compound according to claim 1, wherein Ring A is selected from:
Figure imgf000461_0001
Figure imgf000462_0001
Figure imgf000463_0001
Figure imgf000464_0001
3. The compound according to claim 1, wherein Ring A is selected from i, H, iv, v, vi, vii, ix, xiv, xvi, Hi, Ixiii, Ixxi, Ixxiv, Ixxvi, Ixxviii, and Ixxxi.
4. The compound according to claim 1, wherein Ring B is selected from:
Figure imgf000464_0002
Figure imgf000465_0001
Figure imgf000466_0001
Figure imgf000467_0001
Figure imgf000468_0001
5. The compound according to claim 1, wherein Ring B is selected from i, H, Hi, iv, v, ix, x, jci, JCiVi, jcvi, jcvi'i, jcijc, jcjc, jcjcv, jcjcvi, JCJCJCiV, jcjcjciv, JCJCJCV, jcjcjcviVi, jcftV, xlvi, xlviii, I, Iviii, Ixiv, Ixxviii, Ixxxiii, Ixxxvi, xciv, c, ci, cii, ciii, civ, and cv.
6. The compound according to claim 1, wherein W1 and W2 are each a bivalent Ci_3 alkylene chain wherein one methylene unit of W1 or W2 is optionally replaced by -NR2-, - N(R2)C(0)-, -C(O)N(R2)-, -N(R2)SO2-, -SO2N(R2)-, -0-, -C(O)-, -OC(O)-, -C(O)O-, -S-, -SO- or -SO2-.
7. The compound according to claim 1, wherein W1 and W2 are each independently - C(=0), -NR2-, -S-, or -O-
8. The compound according to claim 1, wherein said compound is of formula II-a or II-b:
Figure imgf000469_0001
II-a II-b or a pharmaceutically acceptable salt thereof.
9. The compound according to claim 1, wherein said compound is of formula III-a or III-b:
Figure imgf000469_0002
III-a III-b or a pharmaceutically acceptable salt thereof.
10. The compound according to claim 12 or claim 13, wherein said compound is of formula IV-a or IV-b:
Figure imgf000469_0003
IV-a IV-b or a pharmaceutically acceptable salt thereof.
11. The compound according to claim 1 , wherein R1 is -L-Y, wherein:
L is a bivalent C2-8 straight or branched, hydrocarbon chain wherein L has at least one double bond and one or two additional methylene units of L are optionally and independently replaced by -NRC(O)-, -C(O)NR-, -N(R)SO2-, -SO2N(R)-, -S-, -S(O)-, -SO2-, -OC(O)-, -C(O)O-, cyclopropylene, -0-, -N(R)-, or -C(O)-;
Y is hydrogen, Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN, or a 3-10 membered monocyclic or bicyclic, saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, and wherein said ring is substituted with 1-4 Re groups; and each Re is independently selected from -Q-Z, oxo, NO2, halogen, CN, a suitable leaving group, or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN, wherein:
Q is a covalent bond or a bivalent Ci_6 saturated or unsaturated, straight or branched, hydrocarbon chain, wherein one or two methylene units of Q are optionally and independently replaced by -N(R)-, -S-, -0-, -C(O)-, -OC(O)-, -C(O)O-, -SO-, or -SO2-, -N(R)C(O)-, -C(O)N(R)-, -N(R)SO2-, or -SO2N(R)-; and Z is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN.
12. The compound according to claim 11, wherein:
L is a bivalent C2_8 straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by -C(O)-, -NRC(O)-, -C(O)NR-, -N(R)SO2-, -SO2N(R)-, -S-, -S(O)-, -SO2-, -OC(O)-, or -C(O)O-, and one or two additional methylene units of L are optionally and independently replaced by cyclopropylene, -0-, -N(R)-, or -C(O)-; and
Y is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN.
13. The compound according to claim 12, wherein L is a bivalent C2_g straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by -C(O)-, and one or two additional methylene units of L are optionally and independently replaced by cyclopropylene, -0-, -N(R)-, or -C(O)-.
14. The compound according to claim 12, wherein L is a bivalent C2_8 straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by -OC(O)-.
15. The compound according to claim 12, wherein L is -NRC(O)CH=CH-, -NRC(O)CH=CHCH2N(CH3)-, -NRC(O)CH=CHCH2O-, -CH2NRC(O)CH=CH-, -NRSO2CH=CH-, -NRSO2CH=CHCH2-, -NRC(O)(C=N2)-, -NRC(O)(C=N2)C(O)-, -NRC(O)CH=CHCH2N(CH3)-, -NRSO2CH=CH-, -NRSO2CH=CHCH2-, -NRC(O)CH=CHCH2O-, -NRC(O)Q=CH2)CH2-, -CH2NRC(O)-, -CH2NRC(O)CH=CH-, -CH2CH2NRC(O)-, or -CH2NRC(O)cyclopropylene-; wherein R is H or optionally substituted Ci_6 aliphatic; and Y is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN.
16. The compound according to claim 15, wherein L is -NHC(O)CH=CH-, -NHC(O)CH=CHCH2N(CH3)-, -NHC(O)CH=CHCH2O-, -CH2NHC(O)CH=CH-, -NHSO2CH=CH-, -NHSO2CH=CHCH2-, -NHC(O)(C=N2)-, -NHC(O)(C=N2)C(O)-, -NHC(O)CH=CHCH2N(CH3)-, -NHSO2CH=CH-, -NHSO2CH=CHCH2-, -NHC(O)CH=CHCH2O-, -NHC(O)C(=CH2)CH2-, -CH2NHC(O)-, -CH2NHC(O)CH=CH-, -CH2CH2NHC(O)-, or -CH2NHC(O)cyclopropylene-.
17. The compound according to claim 12, wherein L is a bivalent C2_8 straight or branched, hydrocarbon chain wherein L has at least one alkylidenyl double bond and at least one methylene unit of L is replaced by -C(O)-, -NRC(O)-, -C(O)NR-, -N(R)SO2-, -SO2N(R)-, -S-, -S(O)-, -SO2- , -OC(O)-, or -C(O)O-, and one or two additional methylene units of L are optionally and independently replaced by cyclopropylene, -0-, -N(R)-, or -C(O)-.
18. The compound according to claim 1, wherein R1 is -L-Y, wherein:
L is a bivalent C2_8 straight or branched, hydrocarbon chain wherein L has at least one triple bond and one or two additional methylene units of L are optionally and independently replaced by -NRC(O)-, -C(O)NR-, -N(R)SO2-, -SO2N(R)-, -S-, -S(O)-, -SO2-, -OC(O)-, or -C(O)O-,
Y is hydrogen, Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN, or a 3-10 membered monocyclic or bicyclic, saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, and wherein said ring is substituted with 1-4 Re groups; and each Re is independently selected from -Q-Z, oxo, NO2, halogen, CN, a suitable leaving group, or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN, wherein:
Q is a covalent bond or a bivalent Ci_6 saturated or unsaturated, straight or branched, hydrocarbon chain, wherein one or two methylene units of Q are optionally and independently replaced by -N(R)-, -S-, -0-, -C(O)-, -OC(O)-, -C(O)O-, -SO-, or -SO2-, -N(R)C(O)-, -C(O)N(R)-, -N(R)SO2-, or -SO2N(R)-; and Z is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN.
19. The compound according to claim 18, wherein Y is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN.
20. The compound according to claim 19, wherein L is -C≡C-, -C≡CCH2N(isopropyl)-, -NHC(O)C≡CCH2CH2-, -CH2-C≡C-CH2-, -C≡CCH2O-, -CH2C(O)C≡C-, -C(O)C≡C-, or -CH2OC(=O)C≡C-.
21. The compound according to claim 1, wherein R1 is -L-Y, wherein:
L is a bivalent C2_8 straight or branched, hydrocarbon chain wherein one methylene unit of L is replaced by cyclopropylene and one or two additional methylene units of L are independently replaced by -NRC(O)-, -C(O)NR-, -N(R)SO2-, -SO2N(R)-, -S-, -S(O)-, -SO2-, -OC(O)-, or -C(O)O-;
Y is hydrogen, Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN, or a 3-10 membered monocyclic or bicyclic, saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, and wherein said ring is substituted with 1-4 Re groups; and each Re is independently selected from -Q-Z, oxo, NO2, halogen, CN, a suitable leaving group, or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN, wherein:
Q is a covalent bond or a bivalent Ci_6 saturated or unsaturated, straight or branched, hydrocarbon chain, wherein one or two methylene units of Q are optionally and independently replaced by -N(R)-, -S-, -0-, -C(O)-, -OC(O)-, -C(O)O-, -SO-, or -SO2-, -N(R)C(O)-, -C(O)N(R)-, -N(R)SO2-, or -SO2N(R)-; and Z is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN.
22. The compound according to claim 21, wherein Y is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN.
23. The compound according to claim 1, wherein R1 is -L-Y, wherein:
L is a covalent bond, -C(O)-, -N(R)C(O)-, or a bivalent Ci_8 saturated or unsaturated, straight or branched, hydrocarbon chain; and Y is selected from the following (i) through (xvii):
(i) Ci_6 alkyl substituted with oxo, halogen, NO2, or CN;
(U) C2-6 alkenyl optionally substituted with oxo, halogen, NO2, or CN; or
(in) C2-6 alkynyl optionally substituted with oxo, halogen, NO2, or CN; or
(Jv) a saturated 3-4 membered heterocyclic ring having 1 heteroatom selected from oxygen or nitrogen wherein said ring is substituted with 1-2 Re groups; or (v) a saturated 5-6 membered heterocyclic ring having 1-2 heteroatom selected from oxygen or nitrogen wherein said ring is substituted with 1-4 Re groups; or
(vi)
Figure imgf000473_0001
, wherein each R, Q, Z; or
(vii) a saturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4
Re groups; or (yiii) a partially unsaturated 3-6 membered monocyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups; or (ix) a partially unsaturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 Re groups;
Figure imgf000473_0002
(xi) a partially unsaturated 4-6 membered heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups; or
Figure imgf000474_0001
(xzzz) a 6-membered aromatic ring having 0-2 nitrogens wherein said ring is substituted with 1-4 Re groups; or
Figure imgf000474_0002
wherein each Re is as defined above and described herein; or
(xv) a 5-membered heteroaryl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-3 Re groups; or
Figure imgf000474_0003
(xvzϊ) an 8-10 membered bicyclic, saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups; wherein: each R group is independently hydrogen or an optionally substituted group selected from Ci_6 aliphatic, phenyl, a 4-7 membered heterocylic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; each Re is independently selected from -Q-Z, oxo, NO2, halogen, CN, a suitable leaving group, or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN, wherein:
Q is a covalent bond or a bivalent Ci_6 saturated or unsaturated, straight or branched, hydrocarbon chain, wherein one or two methylene units of Q are optionally and independently replaced by -N(R)-, -S-, -0-, -C(O)-, -OC(O)-, -C(O)O-, -SO-, or -SO2-, -N(R)C(O)-, -C(O)N(R)-, -N(R)SO2-, or -SO2N(R)-; and Z is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN.
24. The compound according to claim 23, wherein L is a covalent bond, -CH2-, -NH-, -C(O)- , -CH2NH-, -NHCH2-, -NHC(O)-, -NHC(O)CH2OC(O)-, -CH2NHC(O)-, -NHSO2-, -NHSO2CH2-, -NHC(O)CH2OC(O)-, or -SO2NH-.
25. The compound according to claim 24, wherein L is a covalent bond.
26. The compound according to any of claims 23, 24, and 25, wherein Y is selected from:
Figure imgf000475_0001
Figure imgf000476_0001
Figure imgf000477_0001
Figure imgf000478_0001
wherein each Re is independently selected from halogen.
27. The compound according to claim 1 , wherein R1 is selected from:
Figure imgf000478_0002
Figure imgf000479_0001
Figure imgf000480_0001
Figure imgf000481_0001
Figure imgf000482_0001
wherein each Re is independently a suitable leaving group, NO2, CN, or oxo.
28. A compound selected from the group consisting of:
Figure imgf000483_0001
Figure imgf000484_0001
Figure imgf000485_0001
Figure imgf000486_0001
Figure imgf000487_0001
Figure imgf000488_0001
Figure imgf000489_0001
Figure imgf000490_0001
Figure imgf000491_0001
Figure imgf000492_0001
Figure imgf000493_0001
Figure imgf000494_0001
Figure imgf000495_0001
Figure imgf000496_0001
Figure imgf000497_0001
Figure imgf000498_0001
Figure imgf000499_0001
Figure imgf000500_0001
Figure imgf000501_0001
Figure imgf000502_0001
Figure imgf000503_0001
Figure imgf000504_0001
Figure imgf000505_0001
Figure imgf000506_0001
Figure imgf000507_0001
Figure imgf000508_0001
Figure imgf000509_0001
Figure imgf000510_0001
Figure imgf000511_0001
Figure imgf000512_0001
Figure imgf000513_0001
Figure imgf000514_0001
Figure imgf000515_0001
Figure imgf000516_0001
1-360 1-361, or a pharmaceutically acceptable salt thereof.
29. The compound according to claim 28 having the structure:
Figure imgf000516_0002
1-7 1-4
Figure imgf000516_0003
1-96 1-182 1-190
Figure imgf000517_0001
or a pharmaceutically acceptable salt thereof.
30. A composition comprising a compound according to claim 1, and a pharmaceutically acceptable adjuvant, carrier, or vehicle.
31. The composition according to claim 30, in combination with an additional therapeutic agent.
32. The composition according to claim 31, wherein the additional therapeutic agent is a chemotherapeutic agent.
33. A method for inhibiting BTK, or a mutant thereof, activity in a patient or in a biological sample comprising the step of administering to said patient or contacting said biological sample with a compound according to claim 1.
34. The method according to claim 33, wherein the BTK, or a mutant thereof, activity is inhibited irreversibly.
35. The method according to claim 34, wherein the BTK, or a mutant thereof, activity is inhibited irreversibly by covalently modifying Cys 481 of BTK.
36. A method for treating a BTK-mediated disorder in a patient in need thereof, comprising the step of administering to said patient a compound according to claim 1.
37. The method according to claim 36, wherein the disorder is an autoimmune disease, a heteroimmune disease, an inflammatory disease, a cancer, a disease of the bone and joints, or a thromboembolic disorder.
38. The method according to claim 37, wherein the disorder is selected from rheumatoid arthritis, multiple sclerosis, B-cell chronic lymphocytic leukemia, acute lymphocytic leukemia, hairy cell leukemia, non-Hodgkin's lymphoma, Hodgkin's lymphoma, multiple myeloma, bone cancer, bone metastasis, osteoporosis, irritable bowel syndrome, Crohn's disease, lupus, or disorders associated with renal transplant.
39. A method for inhibiting one or more TEC-kinases, or a mutant thereof, activity in a patient or in a biological sample comprising the step of administering to said patient or contacting said biological sample with a compound according to claim 1.
40. The method according to claim 39, wherein the TEC-kinase, or a mutant thereof, activity is inhibited irreversibly.
41. The method according to claim 40 wherein the TEC-kinase is selected from one or more of TEC, ITK or BMX.
42. The method according to claim 41, where the one or more of TEC, ITK or BMX activity is inhibited irreversibly by colvalently modifying Cys 449 of TEC, Cys 442 of ITK or Cys 496 of BMX.
43. A method for treating a TEC-kinase-mediated disorder in a patient in need thereof, comprising the step of administering to said patient a compound according to claim 1.
44. The method according to claim 43, wherein the disorder is an autoimmune disorder, an inflammatory disorder, a proliferative disorder, a hyperproliferative disease, an immunological- mediated diseases, a disease of the respiratory tract, a disease of the bone and joints, a skin disorder, a gastrointestinal disorder, a systemic disease, or allograft rejection.
45. A method for inhibiting one or more of ErbBl, ErbB2, ErbB3, or ErbB4, or a mutant thereof, activity in a patient or in a biological sample comprising the step of administering to said patient or contacting said biological sample with a compound according to claim 1.
46. The method according to claim 45, wherein the one or more of ErbBl, ErbB2, or ErbB4, or a mutant thereof, activity is inhibited irreversibly.
47. The method according to claim 46, wherein the one or more of ErbBl, ErbB2, or ErbB4, or a mutant thereof, activity is inhibited irreversibly by covalently modifying Cys797 of ErbBl, Cys805 of ErbB2 or Cys803 of ErbB4.
48. A method for treating an ErbBl-, ErbB2-, ErbB3-, or ErbB4-mediated disorder in a patient in need thereof, comprising the step of administering to said patient a compound according to claim 1.
49. The method according to claim 48, wherein the disorder is a carcinoma selected from breast cancer, glioblastoma, lung cancer, cancer of the head and neck, colorectal cancer, bladder cancer, non-small cell lung cancer, squamous cell carcinoma, salivary gland carcinoma, ovarian carcinoma, or pancreatic cancer.
50. The method according to claim 49, wherein the disorder is selected from neurofibromatosis type I (NFl), neurofibromatosis type II (NF2) Schwann cell neoplasms (e.g. MPNST's), or a Schwannoma.
51. A method for inhibiting JAK3, or a mutant thereof, activity in a patient or in a biological sample comprising the step of administering to said patient or contacting said biological sample with a compound according to claim 1.
52. The method according to claim 51, wherein the JAK3, or a mutant thereof, activity is inhibited irreversibly.
53. The method according to claim 52, wherein the JAK3, or a mutant thereof, activity is inhibited irreversibly by covalently modifying Cys 909 of JAK3.
54. A method for treating a JAK3-mediated disorder in a patient in need thereof, comprising the step of administering to said patient a compound according to claim 1.
55. The method according to claim 54, wherein the disorder is selected from an autoimmune disorder, an inflammatory disorder, a neurodegenerative disorder, or a solid or hematologic malignancy.
56. A compound of formula V-a or V-b:
Figure imgf000520_0001
V-a V-b. wherein:
Ring A is an optionally substituted group selected from phenyl, a 3-7 membered saturated or partially unsaturated carbocyclic ring, an 8-10 membered bicyclic saturated, partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
Ring B is an optionally substituted group selected from phenyl, a 3-7 membered saturated or partially unsaturated carbocyclic ring, an 8-10 membered bicyclic saturated, partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur; R1 is a bivalent warhead group; Ry is hydrogen, halogen, CN, CF3, Ci_4 aliphatic, Ci_4 haloaliphatic, -OR, -C(O)R, or
-C(O)N(R)2; each R group is independently hydrogen or an optionally substituted group selected from Ci_6 aliphatic, phenyl, a 4-7 membered heterocylic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; W1 and W2 are each independently a covalent bond or a bivalent Ci_3 alkylene chain wherein one methylene unit of W1 or W2 is optionally replaced by -NR2-, -N(R2)C(O)-, -C(O)N(R2)-, -N(R2)SO2-, -SO2N(R2)-, -O-, -C(O)-, -OC(O)-, -C(O)O-, -S-, -SO- or -SO2-; R2 is hydrogen, optionally substituted Ci_6 aliphatic, or -C(O)R, or:
R2 and a substituent on Ring A are taken together with their intervening atoms to form a 4-6 membered partially unsaturated or aromatic fused ring; or R2 and Ry are taken together with their intervening atoms to form a 4-6 membered saturated, partially unsaturated, or aromatic fused ring; m and p are independently 0-4;
Rx and Rv are independently selected from -R, halogen, -OR, -0(CH2)q0R, -CN, -NO2, -SO2R, -SO2N(R)2, -SOR, -C(O)R, -CO2R, -C(O)N(R)2, -NRC(O)R, -NRC(O)N(R)2, -NRSO2R, or -N(R)2, wherein q is 1-4; or:
Rx and R1 when concurrently present on Ring B are taken together with their intervening atoms to form a 5-7 membered saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with a warhead group and 0-3 groups independently selected from oxo, halogen, CN, or Ci_6 aliphatic; or
Rv and R1 when concurrently present on Ring A are taken together with their intervening atoms to form a 5-7 membered saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with a warhead group and 0-3 groups independently selected from oxo, halogen, CN, or Ci_6 aliphatic;
T is a bivalent tethering moiety; and
R1 is a detectable moiety.
57. A compound of formula VI-a, VI-b, VII-a, or VII-b:
Figure imgf000522_0001
VII-a VII-b, wherein:
Ring A is an optionally substituted group selected from phenyl, a 3-7 membered saturated or partially unsaturated carbocyclic ring, an 8-10 membered bicyclic saturated, partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
Ring B is an optionally substituted group selected from phenyl, a 3-7 membered saturated or partially unsaturated carbocyclic ring, an 8-10 membered bicyclic saturated, partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur; R1 is a warhead group; Ry is hydrogen, halogen, CN, CF3, Ci_4 aliphatic, Ci_4 haloaliphatic, -OR, -C(O)R, or
-C(O)N(R)2; each R group is independently hydrogen or an optionally substituted group selected from Ci_6 aliphatic, phenyl, a 4-7 membered heterocylic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; W1 and W2 are each independently a covalent bond or a bivalent Ci_3 alkylene chain wherein one methylene unit of W1 or W2 is optionally replaced by -NR2-, -N(R2)C(O)-, -C(O)N(R2)-, -N(R2)SO2-, -SO2N(R2)-, -O-, -C(O)-, -OC(O)-, -C(O)O-, -S-, -SO- or -SO2-; R2 is hydrogen, optionally substituted Ci_6 aliphatic, or -C(O)R, or:
R2 and a substituent on Ring A are taken together with their intervening atoms to form a 4-6 membered partially unsaturated or aromatic fused ring; or R2 and Ry are taken together with their intervening atoms to form a 4-6 membered saturated, partially unsaturated, or aromatic fused ring; m and p are independently 0-4;
Rx and Rv are independently selected from -R, halogen, -OR, -0(CH2)q0R, -CN, -NO2, -SO2R, -SO2N(R)2, -SOR, -C(O)R, -CO2R, -C(O)N(R)2, -NRC(O)R, -NRC(O)N(R)2, -NRSO2R, or -N(R)2, wherein q is 1-4; or:
Rx and R1 when concurrently present on Ring B are taken together with their intervening atoms to form a 5-7 membered saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with a warhead group and 0-3 groups independently selected from oxo, halogen, CN, or Ci_6 aliphatic; or Rv and R1 when concurrently present on Ring A are taken together with their intervening atoms to form a 5-7 membered saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with a warhead group and 0-3 groups independently selected from oxo, halogen, CN, or Ci_6 aliphatic;
T is a bivalent tethering moiety; and
R1 is a detectable moiety.
58. The compound of claim 56 or 57, wherein T is selected from:
Figure imgf000524_0001
59. The compound of claim 56 or 57, wherein R1 is biotin.
60. The compound of claim 56 or 57, wherein R1 is biotin sulfoxide.
61. The compound of claim 56 or 57, wherein R1 is a radioisotope.
62. The compound of claim 56 or 57, wherein R1 is a fluorescent label.
63. The compound of claim 56 having the structure:
Figure imgf000525_0001
1-363
Figure imgf000526_0001
Figure imgf000527_0001
or 1-368.
64. A method comprising the steps of:
(a) providing one or more tissues, cell types, or a lysate thereof, obtained from a patient administered at least one dose of a compound of formula I-a or I-b:
Figure imgf000527_0002
or a pharmaceutically acceptable salt thereof, wherein:
Ring A is an optionally substituted group selected from phenyl, a 3-7 membered saturated or partially unsaturated carbocyclic ring, an 8-10 membered bicyclic saturated, partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
Ring B is an optionally substituted group selected from phenyl, a 3-7 membered saturated or partially unsaturated carbocyclic ring, an 8-10 membered bicyclic saturated, partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
R1 is a warhead group;
Ry is hydrogen, halogen, -CN, lower alkyl, Ci_4 haloaliphatic, -OR, -C(O)R, or -C(O)N(R)2; each R group is independently hydrogen or an optionally substituted group selected from C1. 6 aliphatic, phenyl, a 4-7 membered heterocylic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
W1 and W2 are each independently a covalent bond or a bivalent Ci_3 alkylene chain wherein one methylene unit of W1 or W2 is optionally replaced by -NR2-, -N(R2)C(O)-, - C(O)N(R2)-, -N(R2)SO2-, -SO2N(R2)-, -O-, -C(O)-, -OC(O)-, -C(O)O-, -S-, -SO- or -SO2-;
R2 is hydrogen, optionally substituted Ci_6 aliphatic, or -C(O)R, or:
R2 and a substituent on Ring A are taken together with their intervening atoms to form a 4-6 membered saturated, partially unsaturated, or aromatic fused ring, or: R2 and Ry are taken together with their intervening atoms to form a 4-7 membered partially unsaturated or aromatic fused ring; m and p are independently 0-4; and
Rx and Rv are independently selected from -R, halogen, -OR, -0(CH2)q0R, -CN, -NO2, -SO2R, -SO2N(R)2, -SOR, -C(O)R, -CO2R, -C(O)N(R)2, -NRC(O)R, -NRC(O)NR2, - NRSO2R, or -N(R)2, wherein q is 1-4; or: Rx and R1 when concurrently present on Ring B are taken together with their intervening atoms to form a 5-7 membered saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with a warhead group and 0-3 groups independently selected from oxo, halogen, -CN, or Ci_6 aliphatic; or
Rv and R1 when concurrently present on Ring A are taken together with their intervening atoms to form a 5-7 membered saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with a warhead group and 0-3 groups independently selected from oxo, halogen, -CN, or Ci_6 aliphatic,
(b) contacting said tissue, cell type, or a lysate thereof, with a different compound of formula I-a or I-b, tethered to a detectable moiety to form a probe compound, to covalently modify at least one protein kinase present in said tissue, cell type, or a lysate thereof; and
(c) measuring the amount of said protein kinase covalently modified by the probe compound to determine occupancy of said protein kinase by said compound of formula I-a or I-b as compared to occupancy of said protein kinase by said probe compound.
65. The method of claim 64, further comprising the step of adjusting the dose of the compound of formula I-a or I-b to increase occupancy of the protein kinase.
66. The method of claim 64, further comprising the step of adjusting the dose of the compound of formula I-a or I-b to decrease occupancy of the protein kinase.
67. The method of claim 64, wherein the measuring step is carried out by one of the following: flow cytometry, Western blot, or ELISA.
PCT/US2009/048784 2008-06-27 2009-06-26 Heteroaryl compounds and uses thereof WO2009158571A1 (en)

Priority Applications (21)

Application Number Priority Date Filing Date Title
RU2010151355/04A RU2536584C2 (en) 2008-06-27 2009-06-26 Heteroaryl compounds and using them
AU2009262068A AU2009262068C1 (en) 2008-06-27 2009-06-26 Heteroaryl compounds and uses thereof
BRPI0914682A BRPI0914682B8 (en) 2008-06-27 2009-06-26 heteroaryl compounds and compositions comprising said compounds
DK09771102.2T DK2361248T3 (en) 2008-06-27 2009-06-26 Heteroberl compounds and uses thereof
CN2009801244104A CN102083800A (en) 2008-06-27 2009-06-26 Heteroaryl compounds and uses thereof
CA2727455A CA2727455C (en) 2008-06-27 2009-06-26 Heteroaryl compounds and uses thereof
NZ589843A NZ589843A (en) 2008-06-27 2009-06-26 Pyrimidine heteroaryl compounds and uses thereof as protein kinase inhibitors
MX2013013212A MX357627B (en) 2008-06-27 2009-06-26 Heteroaryl compounds and uses thereof.
MX2010014029A MX2010014029A (en) 2008-06-27 2009-06-26 Heteroaryl compounds and uses thereof.
KR1020167034131A KR101892989B1 (en) 2008-06-27 2009-06-26 Heteroaryl compounds and uses thereof
EP09771102.2A EP2361248B1 (en) 2008-06-27 2009-06-26 Heteroaryl compounds and uses thereof
MX2015012477A MX360970B (en) 2008-06-27 2009-06-26 Heteroaryl compounds and uses thereof.
EP18195292.0A EP3549934A1 (en) 2008-06-27 2009-06-26 Heteroaryl compounds and uses thereof
ES09771102T ES2711249T3 (en) 2008-06-27 2009-06-26 Heteroaryl compounds and uses thereof
JP2011516699A JP2011526299A (en) 2008-06-27 2009-06-26 Heteroaryl compounds and their use
KR1020187011709A KR101955914B1 (en) 2008-06-27 2009-06-26 Heteroaryl compounds and uses thereof
IL209969A IL209969A (en) 2008-06-27 2010-12-13 Heteroaryl compounds and uses thereof
ZA2010/09216A ZA201009216B (en) 2008-06-27 2010-12-22 Heteroaryl compounds and uses thereof
IL230290A IL230290A (en) 2008-06-27 2014-01-02 Heteroaryl compounds and uses thereof
PH12015501484A PH12015501484A1 (en) 2008-06-27 2015-06-26 Heteroaryl compounds and uses thereof
IL250108A IL250108A0 (en) 2008-06-27 2017-01-15 Heteroaryl compounds and uses thereof

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US7645008P 2008-06-27 2008-06-27
US61/076,450 2008-06-27
US14838809P 2009-01-29 2009-01-29
US61/148,388 2009-01-29
US17087409P 2009-04-20 2009-04-20
US61/170,874 2009-04-20

Publications (2)

Publication Number Publication Date
WO2009158571A1 true WO2009158571A1 (en) 2009-12-30
WO2009158571A8 WO2009158571A8 (en) 2010-02-25

Family

ID=41228450

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/048784 WO2009158571A1 (en) 2008-06-27 2009-06-26 Heteroaryl compounds and uses thereof

Country Status (19)

Country Link
US (6) US8450335B2 (en)
EP (2) EP3549934A1 (en)
JP (4) JP2011526299A (en)
KR (3) KR20110025224A (en)
CN (2) CN108047142B (en)
AU (1) AU2009262068C1 (en)
BR (1) BRPI0914682B8 (en)
CA (3) CA3031835C (en)
DK (1) DK2361248T3 (en)
ES (1) ES2711249T3 (en)
IL (3) IL209969A (en)
MX (3) MX360970B (en)
NZ (3) NZ603525A (en)
PH (1) PH12015501484A1 (en)
RU (2) RU2536584C2 (en)
SG (1) SG10201510696RA (en)
TW (3) TWI458721B (en)
WO (1) WO2009158571A1 (en)
ZA (1) ZA201009216B (en)

Cited By (178)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010144647A1 (en) * 2009-06-12 2010-12-16 Bristol-Myers Squibb Company Nicotinamide compounds useful as kinase modulators
WO2011018517A1 (en) 2009-08-14 2011-02-17 Boehringer Ingelheim International Gmbh Regioselective preparation of 2-amino-5-trifluoromethylpyrimidine derivatives
WO2011018518A1 (en) 2009-08-14 2011-02-17 Boehringer Ingelheim International Gmbh Regioselective preparation of 2 -amino-5-trifluoromethylpyrimidine derivatives
WO2011068898A1 (en) * 2009-12-01 2011-06-09 Rigel Pharmaceuticals, Inc. Protein kinase c inhibitors and uses thereof
KR20110139653A (en) * 2010-06-23 2011-12-29 한미홀딩스 주식회사 Novel fused pyrimidine derivatives for inhibition of tyrosine kinase activity
EP2440559A2 (en) 2009-05-05 2012-04-18 Dana-Farber Cancer Institute, Inc. Egfr inhibitors and methods of treating disorders
JP2012515724A (en) * 2009-01-21 2012-07-12 ライジェル ファーマシューティカルズ, インコーポレイテッド N2- (3-pyridyl or phenyl) -N4- (4-piperidyl) -2,4-pyrimidinediamine derivatives useful for the treatment of inflammatory diseases, autoimmune diseases or proliferative diseases
EP2519664A1 (en) * 2009-12-30 2012-11-07 Avila Therapeutics, Inc. Ligand-directed covalent modification of protein
WO2013014448A1 (en) * 2011-07-27 2013-01-31 Astrazeneca Ab 2 - (2, 4, 5 - substituted -anilino) pyrimidine derivatives as egfr modulators useful for treating cancer
WO2012170976A3 (en) * 2011-06-10 2013-04-11 Merck Patent Gmbh Compositions and methods for the production of pyrimidine and pyridine compounds with btk inhibitory activity
JP2013523891A (en) * 2010-04-13 2013-06-17 ライジェル ファーマシューティカルズ, インコーポレイテッド 2,4-pyrimidinediamine compounds and prodrugs thereof and uses thereof
US8466155B2 (en) 2009-10-02 2013-06-18 Boehringer Ingelheim International Gmbh Pyrimidines
US8476284B2 (en) 2006-09-22 2013-07-02 Pharmacyclics, Inc. Inhibitors of Bruton's tyrosine kinase
WO2013108754A1 (en) 2012-01-17 2013-07-25 アステラス製薬株式会社 Pyrazine carboxamide compound
CN103269704A (en) * 2010-11-01 2013-08-28 西建阿维拉米斯研究公司 Heterocyclic compounds and uses thereof
EP2635285A1 (en) * 2010-11-01 2013-09-11 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
US8563568B2 (en) 2010-08-10 2013-10-22 Celgene Avilomics Research, Inc. Besylate salt of a BTK inhibitor
WO2013157540A1 (en) * 2012-04-17 2013-10-24 富士フイルム株式会社 Nitrogen-containing heterocyclic compound or salt thereof
JP2013539795A (en) * 2010-10-14 2013-10-28 アリアド・ファーマシューティカルズ・インコーポレイテッド Method for inhibiting cell proliferation of EGFR-activated cancer
JP2013542256A (en) * 2010-11-10 2013-11-21 セルジーン アヴィロミクス リサーチ, インコーポレイテッド Mutant selective EGFR inhibitor and use thereof
US8609679B2 (en) 2008-06-27 2013-12-17 Celgene Avilomics Research, Inc. 2,4-diaminopyrimidines useful as kinase inhibitors
WO2014011900A3 (en) * 2012-07-11 2014-02-27 Blueprint Medicines Inhibitors of the fibroblast growth factor receptor
US8685988B2 (en) 2012-08-06 2014-04-01 Acea Biosciences, Inc. EGFR modulators and uses thereof
US8697715B2 (en) 2012-03-01 2014-04-15 Array Biopharma, Inc. Serine/threonine kinase inhibitors
US8710222B2 (en) 2008-06-27 2014-04-29 Celgene Avilomics Research, Inc. 2,4-disubstituted pyrimidines useful as kinase inhibitors
US8754090B2 (en) 2010-06-03 2014-06-17 Pharmacyclics, Inc. Use of inhibitors of bruton's tyrosine kinase (Btk)
JP2014514348A (en) * 2011-05-04 2014-06-19 アリアド・ファーマシューティカルズ・インコーポレイテッド Compound for inhibiting cell proliferation of EGFR-activated cancer
US8785464B2 (en) 2008-11-24 2014-07-22 Boehringer Ingelheim International Gmbh Pyrimidine derivatives that inhibit FAK/PTK2
US20140228322A1 (en) * 2013-02-08 2014-08-14 Celgene Avilomics Research, Inc. Erk inhibitors and uses thereof
US8809273B2 (en) 2007-03-28 2014-08-19 Pharmacyclics, Inc. Inhibitors of Bruton's tyrosine kinase
WO2014134308A1 (en) * 2013-03-01 2014-09-04 Amgen Inc. Substituted 7-oxo-pyrido [2, 3-d] pyrimidines and their use for the treatment of egfr / erbb2 related disorders
US8846689B2 (en) 2008-11-24 2014-09-30 Boehringer Ingelheim International Gmbh Substituted pyrimidines for the treatment of diseases such as cancer
CN104130265A (en) * 2014-04-29 2014-11-05 苏州景泓生物技术有限公司 Spiral ring or bridged ring containing pyrimidine compound
US8883803B2 (en) 2008-07-16 2014-11-11 Pharmacyclics, Inc. Inhibitors of Bruton's tyrosine kinase for the treatment of solid tumors
WO2014188173A1 (en) 2013-05-20 2014-11-27 Redx Pharma Limited Pyrazolopyrimidine derivatives useful as inhibitors of bruton's tyrosine kinase
WO2015003658A1 (en) 2013-07-11 2015-01-15 Betta Pharmaceuticals Co., Ltd Protein tyrosine kinase modulators and methods of use
WO2015019365A1 (en) 2013-08-07 2015-02-12 Cadila Healthcare Limited N-cyanomethylamides as inhibitors of janus kinase
WO2015022926A1 (en) 2013-08-12 2015-02-19 大鵬薬品工業株式会社 Novel fused pyrimidine compound or salt thereof
US9012462B2 (en) 2008-05-21 2015-04-21 Ariad Pharmaceuticals, Inc. Phosphorous derivatives as kinase inhibitors
WO2015048662A3 (en) * 2013-09-30 2015-05-07 X-Rx Discovery, Inc. Substituted nicotinimide inhibitors of btk and their preparation and use in the treatment of cancer, inflammation and autoimmune disease
US20150133457A1 (en) * 2012-01-13 2015-05-14 Acea Biosciences Inc. Heterocyclic compounds and uses thereof
WO2015084998A1 (en) * 2013-12-05 2015-06-11 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase
US9056839B2 (en) 2012-03-15 2015-06-16 Celgene Avilomics Research, Inc. Solid forms of an epidermal growth factor receptor kinase inhibitor
WO2015061247A3 (en) * 2013-10-21 2015-07-23 Merck Patent Gmbh Heteroaryl compounds as btk inhibitors and uses thereof
US9108927B2 (en) 2012-03-15 2015-08-18 Celgene Avilomics Research, Inc. Salts of an epidermal growth factor receptor kinase inhibitor
AU2013204962B2 (en) * 2011-07-27 2015-09-03 Astrazeneca Ab Polymorphic form of a mesylate salt of n-(2-{2-dimethylaminoethyl-methylamino}-4-methoxy-5-{[4-(1-methylindol-3-yl)pyrimidin-2-yl]amino}phenyl)prop-2-enamide
US9126944B2 (en) 2013-02-28 2015-09-08 Bristol-Myers Squibb Company Phenylpyrazole derivatives as potent ROCK1 and ROCK2 inhibitors
US9126950B2 (en) 2012-12-21 2015-09-08 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
US9133187B2 (en) 2011-02-28 2015-09-15 Array Biopharma Inc. Serine/threonine kinase inhibitors
US9139534B2 (en) 2011-04-22 2015-09-22 Signal Pharmaceuticals, Llc Substituted diaminocarboxamide and diaminocarbonitrile pyrimidines, compositions thereof, and methods of treatment therewith
WO2015151006A1 (en) 2014-03-29 2015-10-08 Lupin Limited Substituted purine compounds as btk inhibitors
WO2015170266A1 (en) 2014-05-07 2015-11-12 Lupin Limited Substituted pyrimidine compounds as btk inhibitors
US9187462B2 (en) 2011-08-04 2015-11-17 Array Biopharma Inc. Substituted quinazolines as serine/threonine kinase inhibitors
WO2016014904A1 (en) * 2014-07-24 2016-01-28 Beta Pharma, Inc. 2-h-indazole derivatives as cyclin-dependent kinase (cdk) inhibitors and therapeutic uses thereof
US9273077B2 (en) 2008-05-21 2016-03-01 Ariad Pharmaceuticals, Inc. Phosphorus derivatives as kinase inhibitors
US9296753B2 (en) 2012-06-04 2016-03-29 Pharmacyclics Llc Crystalline forms of a Bruton's tyrosine kinase inhibitor
US9321786B2 (en) 2013-03-15 2016-04-26 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
AU2014202057B2 (en) * 2010-06-23 2016-05-05 Hanmi Science Co., Ltd. Novel Fused Pyrimidine Derivatives for Inhibition of Tyrosine Kinase Activity
US9346801B2 (en) 2013-03-01 2016-05-24 Amgen Inc. Substituted 7-oxo-pyrido[2,3-d]pyrimidines and methods of use
EP2922546A4 (en) * 2012-11-20 2016-06-08 Celgene Avilomics Res Inc Methods of treating a disease or disorder associated with bruton's tyrosine kinase
US9364476B2 (en) 2011-10-28 2016-06-14 Celgene Avilomics Research, Inc. Methods of treating a Bruton's Tyrosine Kinase disease or disorder
US9365524B2 (en) 2014-01-30 2016-06-14 Signal Pharmaceuticals, Llc Solid forms of 2-(tert-butylamino)-4-((1R,3R,4R)-3-hydroxy-4-methylcyclohexylamino)-pyrimidine-5-carboxamide, compositions thereof and methods of their use
US9388171B2 (en) 2012-08-27 2016-07-12 Genetech, Inc. Serine/threonine kinase inhibitors
US9415050B2 (en) 2013-08-12 2016-08-16 Pharmacyclics Llc Methods for the treatment of HER2 amplified cancer
US9415049B2 (en) 2013-12-20 2016-08-16 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
US9421208B2 (en) 2013-08-02 2016-08-23 Pharmacyclics Llc Methods for the treatment of solid tumors
US9434700B2 (en) 2013-10-25 2016-09-06 Neil Bifulco, JR. Inhibitors of the fibroblast growth factor receptor
US9447106B2 (en) 2013-04-25 2016-09-20 Beigene, Ltd. Substituted pyrazolo[1,5-a]pyrimidines as bruton's tyrosine kinase modulators
EP2968327A4 (en) * 2013-03-14 2016-09-28 Celgene Avilomics Res Inc Heteroaryl compounds and uses thereof
WO2016174183A1 (en) 2015-04-30 2016-11-03 Bayer Pharma Aktiengesellschaft Combinations of inhibitors of irak4 with inhibitors of btk
US9492471B2 (en) 2013-08-27 2016-11-15 Celgene Avilomics Research, Inc. Methods of treating a disease or disorder associated with Bruton'S Tyrosine Kinase
US9513297B2 (en) 2014-12-16 2016-12-06 Signal Pharmaceuticals, Llc Methods for measurement of inhibition of c-Jun N-terminal kinase in skin
US9533991B2 (en) 2014-08-01 2017-01-03 Pharmacyclics Llc Inhibitors of Bruton's tyrosine kinase
US9540385B2 (en) 2012-11-15 2017-01-10 Pharmacyclics Llc Pyrrolopyrimidine compounds as kinase inhibitors
US9545407B2 (en) 2014-08-07 2017-01-17 Pharmacyclics Llc Formulations of a bruton's tyrosine kinase inhibitor
US9556426B2 (en) 2009-09-16 2017-01-31 Celgene Avilomics Research, Inc. Protein kinase conjugates and inhibitors
JP2017036268A (en) * 2010-08-18 2017-02-16 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Merck Patent Gesellschaft mit beschraenkter Haftung Pyrimidine derivative as fak inhibitor
US9586965B2 (en) 2012-01-13 2017-03-07 Acea Biosciences Inc. Pyrrolo[2,3-d]pyrimidine compounds as inhibitors of protein kinases
WO2017040617A1 (en) 2015-08-31 2017-03-09 Pharmacyclics Llc Btk inhibitor combinations for treating multiple myeloma
WO2017036263A1 (en) * 2015-08-31 2017-03-09 广州科擎新药开发有限公司 2, 4-di-(nitrogen containing group) substituted pyrimidine compound and preparation method and use thereof
US9611283B1 (en) 2013-04-10 2017-04-04 Ariad Pharmaceuticals, Inc. Methods for inhibiting cell proliferation in ALK-driven cancers
AU2015261672B2 (en) * 2011-07-27 2017-04-27 Astrazeneca Ab 2 - (2, 4, 5 - substituted -anilino) pyrimidine derivatives as egfr modulators useful for treating cancer
CN106687457A (en) * 2014-05-13 2017-05-17 阿里亚德医药股份有限公司 Heteroaryl compounds for kinase inhibition
US9655857B2 (en) 2015-03-03 2017-05-23 Pharmacyclics Llc Pharmaceutical formulations of a Bruton's tyrosine kinase inhibitor
US9663524B2 (en) 2013-03-15 2017-05-30 Celgene Car Llc Substituted pyrido[2,3-d]pyrimidines as protein kinase inhibitors
US9695165B2 (en) 2014-01-15 2017-07-04 Blueprint Medicines Corporation Inhibitors of the fibroblast growth factor receptor
EP3195865A1 (en) 2016-01-25 2017-07-26 Bayer Pharma Aktiengesellschaft Combinations of inhibitors of irak4 with inhibitors of btk
US9730931B2 (en) 2013-10-18 2017-08-15 Eisai R&D Management Co., Ltd. Pyrimidine FGFR4 inhibitors
US9782412B2 (en) 2015-01-30 2017-10-10 Taiho Pharmaceutical Co., Ltd. Preventive and/or therapeutic agent of immune disease
US9796685B2 (en) 2014-12-16 2017-10-24 Signal Pharmaceuticals, Llc Formulations of 2-(tert-butylamino)-4-((1R,3R,4R)-3-hydroxy-4-Methylcyclohexylamino)-pyrimidine-5-carboxamide
US9815813B2 (en) 2014-01-17 2017-11-14 Novartis Ag 1-(triazin-3-yl/pyridazin-3-yl)-piper(-azine)idine derivatives and compositions therefor for inhibiting the activity of SHP2
US9828345B2 (en) 2013-02-28 2017-11-28 Bristol-Myers Squibb Company Phenylpyrazole derivatives as potent ROCK1 and ROCK2 inhibitors
KR20170131341A (en) * 2014-11-24 2017-11-29 상하이 인스티튜트 오브 마테리아 메디카 차이니즈 아카데미 오브 싸이언시즈 2-aminopyrimidine compound and pharmaceutical composition and use thereof
US9834571B2 (en) 2012-05-05 2017-12-05 Ariad Pharmaceuticals, Inc. Compounds for inhibiting cell proliferation in EGFR-driven cancers
US9850233B2 (en) 2013-03-14 2017-12-26 Respivert Limited Kinase inhibitors
EP3144292A4 (en) * 2014-04-14 2018-01-17 Shanghai Haiyan Pharmaceutical Technology Co., Ltd 2,3,4,6-tetra-substituted benzene-1,5-diamine derivatives, preparation method therefor and medicinal use thereof
WO2018019252A1 (en) * 2016-07-26 2018-02-01 Jacobio Pharmaceuticals Co., Ltd. Novel fused pyridine derivatives useful as fak/aurora kinase inhibitors
WO2018019204A1 (en) * 2016-07-26 2018-02-01 深圳市塔吉瑞生物医药有限公司 Amino pyrimidine compound for inhibiting protein tyrosine kinase activity
US9885086B2 (en) 2014-03-20 2018-02-06 Pharmacyclics Llc Phospholipase C gamma 2 and resistance associated mutations
AU2016210613B2 (en) * 2010-11-01 2018-03-01 Celgene Car Llc Heterocyclic compounds and uses thereof
US9908889B2 (en) 2015-01-30 2018-03-06 Taiho Pharmaceutical Co., Ltd. Salt of fused pyrimidine compound and crystal thereof
EP3157916A4 (en) * 2014-06-19 2018-03-07 ARIAD Pharmaceuticals, Inc. Heteroaryl compounds for kinase inhibition
WO2018053437A1 (en) 2016-09-19 2018-03-22 Mei Pharma, Inc. Combination therapy
WO2018085731A2 (en) 2016-11-03 2018-05-11 Juno Therapeutics, Inc. Combination therapy of a t cell therapy and a btk inhibitor
US9975897B2 (en) 2014-06-11 2018-05-22 Loxo Oncology, Inc. Pyrazolopyrimidine derivatives useful as inhibitors of Bruton's tyrosine kinase
US10005760B2 (en) 2014-08-13 2018-06-26 Celgene Car Llc Forms and compositions of an ERK inhibitor
US10012650B2 (en) * 2014-12-11 2018-07-03 Merck Patent Gmbh Assays for BTK inhibitors
US10065966B2 (en) 2013-03-15 2018-09-04 Celgene Car Llc Substituted pyrido[2,3-d]pyrimidines as inhibitors of protein kinases
US10077276B2 (en) 2014-01-17 2018-09-18 Novartis Ag N-azaspirocycloalkane substituted N-heteroaryl compounds and compositions for inhibiting the activity of SHP2
US10093646B2 (en) 2014-01-17 2018-10-09 Novartis Ag 1-pyridazin-/triazin-3-yl-piper(-azine)/idine/pyrolidine derivatives and compositions thereof for inhibiting the activity of SHP2
AU2014287016B2 (en) * 2013-07-11 2018-11-01 Acea Biosciences Inc. Pyrimidine derivatives as kinase inhibitors
US10189829B2 (en) 2017-04-14 2019-01-29 Biogen Ma Inc. Inhibiting agents for Bruton's tyrosine kinase
US10202356B2 (en) 2013-03-14 2019-02-12 Tolero Pharmaceuticals, Inc. JAK2 and ALK2 inhibitors and methods for their use
EP3354647A4 (en) * 2015-09-25 2019-04-03 Zhejiang Bossan Pharmaceutical Co. Ltd. Egfr kinase inhibitor and preparation method and use thereof
US10252981B2 (en) 2015-07-24 2019-04-09 Celgene Corporation Methods of synthesis of (1R,2R,5R)-5-amino-2-methylcyclohexanol hydrochloride and intermediates useful therein
US10287266B2 (en) 2015-06-19 2019-05-14 Novartis Ag Compounds and compositions for inhibiting the activity of SHP2
US10308660B2 (en) 2015-06-19 2019-06-04 Novartis Ag Compounds and compositions for inhibiting the activity of SHP2
US10342780B2 (en) 2015-12-16 2019-07-09 Loxo Oncology, Inc. Compounds useful as kinase inhibitors
US10399989B2 (en) 2015-09-16 2019-09-03 Loxo Oncology, Inc. Pyrazolopyrimidine derivatives as BTK inhibitors for the treatment of cancer
WO2019177374A1 (en) * 2018-03-13 2019-09-19 포로노이바이오 주식회사 2, 4, 5-substituted pyrimidine derivative, preparation method therefor, and pharmaceutical composition comprising same as effective ingredient for prevention or treatment of cancer or inflammatory disease
EP3550031A1 (en) 2012-07-24 2019-10-09 Pharmacyclics, LLC Mutations associated with resistance to inhibitors of bruton's tyrosine kinase (btk)
WO2019200254A1 (en) 2018-04-13 2019-10-17 Tolero Pharmaceuticals, Inc. Pim kinase inhibitors for treatment of myeloproliferative neoplasms and fibrosis associated with cancer
US10450269B1 (en) 2013-11-18 2019-10-22 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
US10457669B2 (en) 2015-10-21 2019-10-29 Otsuka Pharmaceutical Co., Ltd. Benzolactam compounds as protein kinase inhibitors
WO2019208805A1 (en) 2018-04-27 2019-10-31 小野薬品工業株式会社 PREVENTIVE AND/OR THERAPEUTIC AGENT FOR AUTOIMMUNE DISEASE COMPRISING COMPOUND HAVING Btk INHIBITORY ACTIVITY AS ACTIVE INGREDIENT
WO2019213184A1 (en) 2018-05-03 2019-11-07 Juno Therapeutics, Inc. Combination therapy of a chimeric antigen receptor (car) t cell therapy and a kinase inhibitor
US10533011B2 (en) 2015-10-09 2020-01-14 ACEA Therapeutics, Inc. Pharmaceutical salts, physical forms, and compositions of pyrrolopyrimidine kinase inhibitors, and methods of making same
US10562888B2 (en) 2015-04-14 2020-02-18 Eisai R&D Management Co., Ltd. Crystalline FGFR4 inhibitor compound and uses thereof
US10596174B2 (en) 2012-01-13 2020-03-24 ACEA Therapeutics, Inc. Pyrrolopyrimidine compounds as inhibitors of protein kinases
US10676434B2 (en) 2014-10-24 2020-06-09 Bristol-Myers Squibb Company Carbazole derivatives
US10689351B2 (en) 2015-01-29 2020-06-23 Signal Pharmaceuticals, Llc Isotopologues of 2-(tert butylamino)-4-((1R,3R,4R)-3-hydroxy-4-methylcyclohexylamino)-pyrimidine-5-carboxamide
US10766884B2 (en) 2018-04-26 2020-09-08 Pfizer Inc. Cyclin dependent kinase inhibitors
CN111747934A (en) * 2018-01-31 2020-10-09 迪哲(江苏)医药有限公司 ERBB/BTK inhibitors
EA036521B1 (en) * 2012-01-27 2020-11-19 Астразенека Аб 2-(2,4,5-substituted-anilino)pyrimidine derivatives as egfr modulators useful for treating cancer
US10875864B2 (en) 2011-07-21 2020-12-29 Sumitomo Dainippon Pharma Oncology, Inc. Substituted imidazo[1,2-B]pyridazines as protein kinase inhibitors
US10927117B2 (en) 2016-08-16 2021-02-23 Beigene Switzerland Gmbh Crystalline form of (S)-7-(1-acryloylpiperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetra-hydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
US10934285B2 (en) 2016-06-14 2021-03-02 Novartis Ag Compounds and compositions for inhibiting the activity of SHP2
EP3640248A4 (en) * 2017-06-13 2021-03-31 Beijing Adamadle Biotechnology Limited Liability Company Aminopyrimidine compound, preparation method therefor and use thereof
US10975080B2 (en) 2015-06-19 2021-04-13 Novartis Ag Compounds and compositions for inhibiting the activity of SHP2
US11001569B2 (en) 2016-01-22 2021-05-11 Janssen Pharmaceutica Nv 6-membered heteroaromatic substituted cyanoindoline derivatives as NIK inhibitors
US11040038B2 (en) 2018-07-26 2021-06-22 Sumitomo Dainippon Pharma Oncology, Inc. Methods for treating diseases associated with abnormal ACVR1 expression and ACVR1 inhibitors for use in the same
US11053195B2 (en) 2013-03-15 2021-07-06 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
US11066404B2 (en) 2018-10-11 2021-07-20 Incyte Corporation Dihydropyrido[2,3-d]pyrimidinone compounds as CDK2 inhibitors
US11098030B2 (en) 2016-05-26 2021-08-24 Recurium Ip Holdings, Llc EGFR inhibitor compounds
US11136311B2 (en) 2016-06-30 2021-10-05 Janssen Pharmaceutica Nv Heteroaromatic derivatives as NIK inhibitors
US11142518B2 (en) 2017-04-20 2021-10-12 Otsuka Pharmaceutical Co., Ltd. 6-pyrimidin-isoindole derivative as ERK1/2 inhibitor
US11180487B2 (en) 2016-01-22 2021-11-23 Janssen Pharmaceutica Nv Substituted cyanoindoline derivatives as NIK inhibitors
US11186589B2 (en) 2016-06-30 2021-11-30 Janssen Pharmaceutica Nv Cyanoindoline derivatives as NIK inhibitors
US11186637B2 (en) 2013-09-13 2021-11-30 Beigene Switzerland Gmbh Anti-PD1 antibodies and their use as therapeutics and diagnostics
US11247987B2 (en) 2017-10-06 2022-02-15 Forma Therapeutics, Inc. Inhibiting ubiquitin specific peptidase 30
US11248003B2 (en) 2017-12-07 2022-02-15 Oncobix Co., Ltd. Pyrimidine derivative having effect of inhibiting cancer cell growth and pharmaceutical composition containing same
WO2022094354A1 (en) * 2020-10-30 2022-05-05 Lengo Therapeutics, Inc. Pyrimidine compounds, compositions, and medicinal applications thereof
US11351168B1 (en) 2008-06-27 2022-06-07 Celgene Car Llc 2,4-disubstituted pyrimidines useful as kinase inhibitors
US11357769B2 (en) 2016-05-10 2022-06-14 Eisai R&D Management Co., Ltd. Drug combinations for reducing cell viability and/or cell proliferation
US11377449B2 (en) 2017-08-12 2022-07-05 Beigene, Ltd. BTK inhibitors with improved dual selectivity
WO2022094172A3 (en) * 2020-10-30 2022-07-07 Newave Pharmaceutical Inc. Inhibitors of btk
US11384083B2 (en) 2019-02-15 2022-07-12 Incyte Corporation Substituted spiro[cyclopropane-1,5′-pyrrolo[2,3-d]pyrimidin]-6′(7′h)-ones as CDK2 inhibitors
US11427567B2 (en) 2019-08-14 2022-08-30 Incyte Corporation Imidazolyl pyrimidinylamine compounds as CDK2 inhibitors
US11427558B1 (en) 2019-07-11 2022-08-30 ESCAPE Bio, Inc. Indazoles and azaindazoles as LRRK2 inhibitors
US11440914B2 (en) 2019-05-01 2022-09-13 Incyte Corporation Tricyclic amine compounds as CDK2 inhibitors
US11447494B2 (en) 2019-05-01 2022-09-20 Incyte Corporation Tricyclic amine compounds as CDK2 inhibitors
US11471456B2 (en) 2019-02-12 2022-10-18 Sumitomo Pharma Oncology, Inc. Formulations comprising heterocyclic protein kinase inhibitors
US11472791B2 (en) 2019-03-05 2022-10-18 Incyte Corporation Pyrazolyl pyrimidinylamine compounds as CDK2 inhibitors
US11498922B2 (en) 2017-04-07 2022-11-15 ACEA Therapeutics, Inc. Pharmaceutical composition comprising N-(3-((2-((3-fluoro-4-(4-methylpiperazin-1-yl phenyl)amino)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)oxy)phenylacrylamide
US11501284B2 (en) 2018-02-19 2022-11-15 Newave Pharmaceutical Inc. Substituted pyrazines as inhibitors of BTK, and mutants thereof
US11512132B2 (en) 2014-07-03 2022-11-29 Beigene, Ltd. Anti-PD-L1 antibodies and their use as therapeutics and diagnostics
US11535618B2 (en) 2018-10-05 2022-12-27 Forma Therapeutics, Inc. Fused pyrrolines which act as ubiquitin-specific protease 30 (USP30) inhibitors
US11534431B2 (en) 2016-07-05 2022-12-27 Beigene Switzerland Gmbh Combination of a PD-1 antagonist and a RAF inhibitor for treating cancer
EP4115886A1 (en) 2013-10-25 2023-01-11 Pharmacyclics LLC Methods of treating and preventing graft versus host disease
US11555038B2 (en) 2017-01-25 2023-01-17 Beigene, Ltd. Crystalline forms of (S)-7-(1-(but-2-ynoyl)piperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
US11597768B2 (en) 2017-06-26 2023-03-07 Beigene, Ltd. Immunotherapy for hepatocellular carcinoma
WO2023076167A1 (en) * 2021-10-25 2023-05-04 Newave Pharmaceutical Inc. Inhibitor of btk and mutants thereof
US11701357B2 (en) 2016-08-19 2023-07-18 Beigene Switzerland Gmbh Treatment of B cell cancers using a combination comprising Btk inhibitors
US11786531B1 (en) 2022-06-08 2023-10-17 Beigene Switzerland Gmbh Methods of treating B-cell proliferative disorder
US11786529B2 (en) 2017-11-29 2023-10-17 Beigene Switzerland Gmbh Treatment of indolent or aggressive B-cell lymphomas using a combination comprising BTK inhibitors
WO2023220655A1 (en) 2022-05-11 2023-11-16 Celgene Corporation Methods to overcome drug resistance by re-sensitizing cancer cells to treatment with a prior therapy via treatment with a t cell therapy
US11851426B2 (en) 2019-10-11 2023-12-26 Incyte Corporation Bicyclic amines as CDK2 inhibitors
US11919904B2 (en) 2019-03-29 2024-03-05 Incyte Corporation Sulfonylamide compounds as CDK2 inhibitors

Families Citing this family (149)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7989465B2 (en) 2007-10-19 2011-08-02 Avila Therapeutics, Inc. 4,6-disubstituted pyrimidines useful as kinase inhibitors
CA2702674C (en) 2007-10-19 2016-05-03 Avila Therapeutics, Inc. Heteroaryl compounds and uses thereof
JP5815411B2 (en) * 2008-12-30 2015-11-17 ライジェル ファーマシューティカルズ, インコーポレイテッド Pyrimidinediamine kinase inhibitor
TW201040162A (en) * 2009-05-06 2010-11-16 Portola Pharm Inc Inhibitors of JAK
JP5607241B2 (en) 2010-05-21 2014-10-15 ケミリア・エービー New pyrimidine derivatives
MX2013010898A (en) 2011-03-24 2013-12-06 Chemilia Ab Novel pyrimidine derivatives.
WO2012135641A2 (en) 2011-03-30 2012-10-04 H. Lee Moffitt Cancer Center And Research Institute Aurora kinase inhibitors and methods of making and using thereof
WO2012148994A1 (en) * 2011-04-25 2012-11-01 Usher Iii Initiative Pyrazolopyridazines and methods for treating retinal-degenerative diseases and hearing loss associated with usher syndrome
US8841301B2 (en) 2011-09-26 2014-09-23 Bristol-Myers Squibb Company Selective NR2B antagonists
US8722670B2 (en) 2011-09-30 2014-05-13 Bristol-Myers Squibb Company Selective NR2B antagonists
US9782406B2 (en) 2011-10-25 2017-10-10 Peking University Shenzhen Graduate School Kinase inhibitor and method for treatment of related diseases
CN103073508B (en) * 2011-10-25 2016-06-01 北京大学深圳研究生院 The method of inhibitors of kinases and treatment relevant disease
AU2012321091B2 (en) * 2011-10-28 2016-05-12 Celgene Avilomics Research, Inc. Methods of treating a Bruton's Tyrosine Kinase disease or disorder
WO2013173518A1 (en) 2012-05-16 2013-11-21 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase
IN2014DN09610A (en) * 2012-05-22 2015-07-31 Univ North Carolina
WO2014039452A1 (en) * 2012-09-04 2014-03-13 Celgene Avilomics Research, Inc. Methods of treating a bruton's tyrosine kinase disease or disorder
WO2014040555A1 (en) * 2012-09-12 2014-03-20 山东亨利医药科技有限责任公司 Nitrogen-containing heteroaromatic ring derivative as tyrosine kinase inhibitor
CA2887435A1 (en) * 2012-10-04 2014-04-10 University Of Utah Research Foundation Substituted n-(3-(pyrimidin-4-yl)phenyl)acrylamide analogs as tyrosine receptor kinase btk inhibitors
BR112015008042A2 (en) * 2012-10-11 2017-07-04 Pharmacyclics Inc complementary diagnoses for tec family kinase inhibitor therapy
US9227976B2 (en) 2012-10-25 2016-01-05 Usher Iii Initiative, Inc. Pyrazolopyridazines and methods for treating retinal-degenerative diseases and hearing loss associated with usher syndrome
CA2889537C (en) * 2012-10-25 2017-12-12 Usher Iii Initiative, Inc. Pyrazolopyridazines and methods for treating retinal-degenerative diseases and hearing loss associated with usher syndrome
US8765762B2 (en) 2012-10-25 2014-07-01 Usher III, Initiative, Inc. Pyrazolopyridazines and methods for treating retinal-degerative diseases and hearing loss associated with usher syndrome
WO2014072419A1 (en) 2012-11-08 2014-05-15 Universiteit Antwerpen Novel anti-hiv compounds
US20140140991A1 (en) * 2012-11-20 2014-05-22 Celgene Avilomics Research, Inc. Methods of treating a disease or disorder associated with bruton's tyrosine kinase
WO2014089913A1 (en) * 2012-12-12 2014-06-19 山东亨利医药科技有限责任公司 Bicyclic compound functioning as tyrosine kinase inhibitor
CN110746424A (en) 2013-03-15 2020-02-04 西建卡尔有限责任公司 MK2 inhibitors and uses thereof
CA2909579A1 (en) 2013-04-17 2014-10-23 Signal Pharmaceuticals, Llc Combination therapy comprising a tor kinase inhibitor and n-(3-(5-fluoro-2-(4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide for treating cancer
CA2909625C (en) 2013-04-17 2021-06-01 Signal Pharmaceuticals, Llc Combination therapy comprising a tor kinase inhibitor and a 5-substituted quinazolinone compound for treating cancer
JP2016521280A (en) * 2013-05-03 2016-07-21 セルジーン コーポレイション How to treat cancer with combination therapy
CN105377835B (en) * 2013-07-11 2018-08-17 贝达药业股份有限公司 Tyrosine protein kinase conditioning agent and its application process
US10273224B2 (en) 2013-08-22 2019-04-30 Jubilant Biosys Limited Substituted pyrimidine compounds, compositions and medicinal applications thereof
JP6458039B2 (en) * 2013-09-18 2019-01-23 北京韓美薬品有限公司 Compound that suppresses the activity of BTK and / or JAK3 kinase
WO2015066696A1 (en) * 2013-11-04 2015-05-07 Forum Pharmaceuticals Inc. Fused morphlinopyrimidines and methods of use thereof
US20150139979A1 (en) * 2013-11-19 2015-05-21 Celgene Avilomics Research, Inc. Methods of treating a disease or disorder associated with bruton's tyrosine kinase
BR112016012794A2 (en) 2013-12-05 2017-08-08 Acerta Pharma Bv THERAPEUTIC COMBINATION OF A PI3K INHIBITOR AND A BTK INHIBITOR
JP6879740B2 (en) * 2013-12-13 2021-06-02 ダナ−ファーバー キャンサー インスティテュート, インコーポレイテッド How to Treat Lymphatic Plasma Cell Lymphoma
CN109045032A (en) 2014-01-01 2018-12-21 麦迪威森技术有限责任公司 Aminopyridines and application method
CN104860941B (en) 2014-02-25 2017-03-22 上海海雁医药科技有限公司 2,4-disubstituted phenyl-1,5-diamine derivatives and use thereof, and pharmaceutical composition and medicinal composition prepared from 2,4-disubstituted phenyl-1,5-diamine derivative
CN104860891B (en) * 2014-02-25 2017-06-30 上海海雁医药科技有限公司 Fragrant amino pyrimidines and its application and pharmaceutical composition prepared therefrom and Pharmaceutical composition
WO2015134805A1 (en) * 2014-03-07 2015-09-11 Celgene Avilomics Research, Inc. Methods of treating a bruton's tyrosine kinase disease or disorder
CN104892527A (en) * 2014-03-07 2015-09-09 山东亨利医药科技有限责任公司 Optical isomers used as tyrosine kinase inhibitors
US9403801B2 (en) 2014-03-28 2016-08-02 Calitor Sciences, Llc Substituted heteroaryl compounds and methods of use
WO2015181633A2 (en) 2014-04-11 2015-12-03 Acerta Pharma B.V. Methods of blocking the cxcr-4/sdf-1 signaling pathway with inhibitors of bruton's tyrosine kinase
WO2015185998A2 (en) 2014-04-11 2015-12-10 Acerta Pharma B.V. Methods of blocking the cxcr-4/sdf-1 signaling pathway with inhibitors of bone marrow x kinase
JP2017514805A (en) * 2014-04-16 2017-06-08 シグナル ファーマシューティカルズ,エルエルシー Methods of treating cancer using TOR kinase inhibitor combination therapy
WO2015158310A1 (en) * 2014-04-18 2015-10-22 山东轩竹医药科技有限公司 Tyrosine kinase inhibitor and uses thereof
AU2015266552A1 (en) * 2014-05-28 2016-12-01 Astellas Pharma Inc. Pharmaceutical composition comprising pyrazine carboxamide compound as active ingredient
CN104086551B (en) * 2014-06-06 2016-09-21 人福医药集团股份公司 Compound and its production and use
CN111892579B (en) * 2014-06-12 2023-07-25 上海艾力斯医药科技股份有限公司 Kinase inhibitors
TW201618783A (en) 2014-08-07 2016-06-01 艾森塔製藥公司 Methods of treating cancers, immune and autoimmune diseases, and inflammatory diseases based on BTK occupancy and BTK resynthesis rate
TW201618774A (en) 2014-08-11 2016-06-01 艾森塔製藥公司 Methods of using BTK inhibitors to treat solid tumors and other diseases through modulation of the tumor microenvironment
PL3179992T3 (en) 2014-08-11 2022-08-16 Acerta Pharma B.V. Therapeutic combinations of a btk inhibitor, a pd-1 inhibitor and/or a pd-l1 inhibitor
PL3179991T3 (en) 2014-08-11 2022-02-14 Acerta Pharma B.V. Therapeutic combinations of a btk inhibitor and a bcl-2 inhibitor
TW201618773A (en) 2014-08-11 2016-06-01 艾森塔製藥公司 Therapeutic combinations of a BTK inhibitor, a PI3K inhibitor, a JAK-2 inhibitor, and/or a CDK4/6 inhibitor
WO2016025651A1 (en) * 2014-08-13 2016-02-18 Celgene Avilomics Research, Inc. Combinations of an erk inhibitor and a tor inhibitor and related methods
WO2016025640A1 (en) * 2014-08-13 2016-02-18 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
WO2016025650A1 (en) * 2014-08-13 2016-02-18 Celgene Avilomics Research, Inc. Combinations of an erk inhibitor and a cdk4/6 inhibitor and related methods
WO2016025621A1 (en) * 2014-08-13 2016-02-18 Celgene Avilomics Research, Inc. Methods of treatment using an erk inhibitor
WO2016025567A1 (en) * 2014-08-13 2016-02-18 Celgene Avilomics Research, Inc. Oral formulations and uses thereof
CN106458969A (en) * 2014-08-25 2017-02-22 四川海思科制药有限公司 (substituted phenyl) (substituted pyrimidine) amino derivative, preparation method therefor, and medication use
CN105399685B (en) * 2014-09-16 2018-05-22 深圳微芯生物科技有限责任公司 The alternatively preparation method and applications of the heteroaromatic compounds of property JAK3 and/or JAK1 kinase inhibitors
DK3193611T3 (en) 2014-09-17 2021-05-10 Celgene Car Llc MK2 INHIBITORS AND USES THEREOF
MX2017004234A (en) * 2014-10-11 2017-06-08 Shanghai Hansoh Biomedical Co Ltd Egfr inhibitor, and preparation and application thereof.
RS61865B1 (en) * 2014-10-13 2021-06-30 Yuhan Corp Compounds and compositions for modulating egfr mutant kinase activities
US10059689B2 (en) 2014-10-14 2018-08-28 Calitor Sciences, Llc Substituted heteroaryl compounds and methods of use
CN111170998B (en) 2014-11-05 2023-04-11 益方生物科技(上海)股份有限公司 Pyrimidine or pyridine compound, preparation method and medical application thereof
WO2016087994A1 (en) 2014-12-05 2016-06-09 Acerta Pharma B.V. Btk inhibitors to treat solid tumors through modulation of the tumor microenvironment
US10266517B2 (en) 2014-12-23 2019-04-23 Dana-Farber Cancer Institute, Inc. Pyrimidines as EGFR inhibitors and methods of treating disorders
CN104788427B (en) * 2015-02-05 2017-05-31 上海泓博智源医药股份有限公司 3 (2 pyrimdinyl-amino) phenylacryloyl amine compounds and its application
CN106146468B (en) * 2015-04-17 2020-12-01 杭州雷索药业有限公司 Pyridone protein kinase inhibitors
JP2018104290A (en) * 2015-04-28 2018-07-05 アステラス製薬株式会社 Pharmaceutical composition containing pyrazinecarboxamide compound as active ingredient
CN104860890B (en) * 2015-04-29 2018-03-13 上海泓博智源医药股份有限公司 T790M mutant egfs R inhibitor and its application in antineoplastic is prepared
WO2016183278A1 (en) * 2015-05-13 2016-11-17 Ariad Pharmaceuticals, Inc. Heteroaryl compounds for kinase inhibition
WO2017033113A1 (en) 2015-08-21 2017-03-02 Acerta Pharma B.V. Therapeutic combinations of a mek inhibitor and a btk inhibitor
CN106478605A (en) * 2015-09-02 2017-03-08 上海页岩科技有限公司 Pyrimidines, its preparation method and medical usage
EP3347097B1 (en) 2015-09-11 2021-02-24 Sunshine Lake Pharma Co., Ltd. Substituted aminopyrimidine derivatives as modulators of the kinases jak, flt3 and aurora
MA44909A (en) 2015-09-15 2018-07-25 Acerta Pharma Bv THERAPEUTIC ASSOCIATION OF A CD19 INHIBITOR AND A BTK INHIBITOR
US20190022092A1 (en) 2015-09-15 2019-01-24 Acerta Pharma B.V. Therapeutic Combinations of a BTK Inhibitor and a GITR Binding Molecule, a 4-1BB Agonist, or an OX40 Agonist
WO2017063103A1 (en) * 2015-10-12 2017-04-20 Peking University Shenzhen Graduate School Novel inhibitors and probes for kinases and uses thereof
CN108137544B (en) * 2015-12-10 2022-01-04 深圳市塔吉瑞生物医药有限公司 Aminopyrimidines useful for inhibiting protein tyrosine kinase activity
CN106928150B (en) * 2015-12-31 2020-07-31 恩瑞生物医药科技(上海)有限公司 Acrylamide aniline derivative and pharmaceutical application thereof
CN109328059B (en) 2016-01-07 2021-08-17 Cs制药技术有限公司 Selective inhibitors of clinically important mutants of EGFR tyrosine kinase
WO2017122175A1 (en) 2016-01-13 2017-07-20 Acerta Pharma B.V. Therapeutic combinations of an antifolate and a btk inhibitor
CN106995437A (en) * 2016-01-22 2017-08-01 齐鲁制药有限公司 Substituted indole or indazole pyrimidine derivatives and its production and use
CN107043368B (en) * 2016-02-05 2020-07-31 齐鲁制药有限公司 Crystals of arylamine pyrimidine compounds and salts thereof
CN107098887B (en) * 2016-02-22 2019-08-09 复旦大学 Pyrimidines
CN105968056A (en) * 2016-05-28 2016-09-28 大连医科大学 Diarylpyrimidine compound, composition and application
CN106349331B (en) * 2016-08-23 2020-05-15 国家纳米科学中心 Bipyrene-based pH response self-assembly polypeptide nano material and preparation method and application thereof
US20180072718A1 (en) 2016-09-09 2018-03-15 Incyte Corporation Pyrazolopyridine compounds and uses thereof
TW201811799A (en) 2016-09-09 2018-04-01 美商英塞特公司 Pyrazolopyrimidine compounds and uses thereof
KR102507967B1 (en) 2016-09-09 2023-03-09 인사이트 코포레이션 Pyrazolopyridine derivatives as HPK1 modulators and their use to treat cancer
EP3523289A1 (en) * 2016-10-07 2019-08-14 Araxes Pharma LLC Heterocyclic compounds as inhibitors of ras and methods of use thereof
CN106565782B (en) * 2016-10-10 2018-04-06 大连医科大学 Phosphoryl pyrimidines, composition and purposes
CN106496196B (en) * 2016-10-20 2019-07-02 南京雷科星生物技术有限公司 A kind of quinazoline, Pyridopyrimidine or double and pyrimidine derivatives egf inhibitor and preparation method thereof and purposes
CN106588885B (en) * 2016-11-10 2019-03-19 浙江大学 2- replaces aromatic ring-pyridine derivatives and preparation and application
CN106749042B (en) * 2016-11-16 2019-01-22 大连医科大学 Sulfoamido pyrimidines, composition and purposes
CN106565614A (en) * 2016-11-16 2017-04-19 大连医科大学 Diphenylaminopyrimidine compound, composition and application
US10316021B2 (en) 2016-11-28 2019-06-11 Pfizer Inc. Heteroarylphenoxy benzamide kappa opioid ligands
EP3548046A2 (en) 2016-12-03 2019-10-09 Juno Therapeutics, Inc. Methods and compositions for use of therapeutic t cells in combination with kinase inhibitors
JP6919922B2 (en) * 2016-12-19 2021-08-18 アビスコ セラピューティクス カンパニー リミテッド FGFR4 inhibitor, its manufacturing method and pharmaceutical application
CN106632273A (en) * 2016-12-29 2017-05-10 大连医科大学 Pyrimidine compound containing azole heterocycle, composition and applications of pyrimidine compound and combination
WO2018134786A1 (en) 2017-01-19 2018-07-26 Acerta Pharma B.V. Compositions and methods for the assessment of drug target occupancy for bruton's tyrosine kinase
US20180228786A1 (en) 2017-02-15 2018-08-16 Incyte Corporation Pyrazolopyridine compounds and uses thereof
CN108498477A (en) * 2017-02-27 2018-09-07 江苏奥赛康药业股份有限公司 A kind of Pharmaceutical composition and preparation method thereof of 2- amino-metadiazine compounds
AR111469A1 (en) * 2017-04-21 2019-07-17 Yuhan Corp COME OUT OF AN AMINOPIRIDINE DERIVATIVE COMPOUND, A CRYSTAL FORM OF THE SAME, AND A PROCESS TO PREPARE THE SAME
CN107200713A (en) * 2017-06-09 2017-09-26 大连医科大学附属第医院 Antitumoral compounds and preparation method thereof and purposes
ES2959860T3 (en) 2017-06-22 2024-02-28 Celgene Corp Treatment of hepatocellular carcinoma characterized by hepatitis B virus infection
EP3648753A4 (en) * 2017-07-05 2021-03-17 CS Pharmatech Limited Selective inhibitors of clinically important mutants of the egfr tyrosine kinase
AU2018308164B2 (en) * 2017-07-28 2021-12-09 Yuhan Corporation Intermediates useful for the synthesis of a selective inhibitor against protein kinase and processes for preparing the same
HUE063533T2 (en) 2017-07-28 2024-01-28 Yuhan Corp Process for preparing n-(5-((4-(4-((dimethylamino)methyl)-3-phenyl-1h-pyrazol-1-yl)pyrimidin-2-yl)amino)-4-methoxy-2-morpholinophenyl)acrylamide by reacting the corresponding amine with a 3-halo-propionyl chloride
KR20200041954A (en) * 2017-08-18 2020-04-22 북경한미약품 유한공사 Compounds, pharmaceutical compositions thereof, and uses and applications thereof
KR102383561B1 (en) * 2017-09-07 2022-04-06 한국화학연구원 Tetrahydroisoquinoline substituted pyrimidine derivative, optical isomer thereof, or pharmaceutically acceptable salts thereof, and composition comprising its same for preventing or treating of cancer
CN107721991B (en) * 2017-11-17 2019-10-18 南方医科大学中西医结合医院 A kind of 6- (pyrimidine-4-yl) -1H- indazole derivative and its preparation method and application
JP7254076B2 (en) 2017-11-19 2023-04-07 サンシャイン・レイク・ファーマ・カンパニー・リミテッド Substituted heteroaryl compounds and methods of use
CN107964027A (en) * 2017-12-07 2018-04-27 大连医科大学 Phosphoryl pyrimidine anti-tumor compounds and preparation method thereof and purposes
CN108047132A (en) * 2017-12-07 2018-05-18 大连医科大学 Diphenylamines yl pyridines anti-tumor compounds and preparation method thereof and purposes
US11384069B2 (en) * 2018-01-16 2022-07-12 Shenzhen Targetrx, Inc. Diphenylaminopyrimidine compound for inhibiting kinase activity
CR20200421A (en) 2018-02-20 2021-01-26 Incyte Corp N-(phenyl)-2-(phenyl)pyrimidine-4-carboxamide derivatives and related compounds as hpk1 inhibitors for treating cancer
US10745388B2 (en) 2018-02-20 2020-08-18 Incyte Corporation Indazole compounds and uses thereof
WO2019164847A1 (en) 2018-02-20 2019-08-29 Incyte Corporation Indazole compounds and uses thereof
CN108610295B (en) * 2018-04-04 2023-01-10 大连医科大学附属第一医院 Pyrimidines, compositions and their use in the treatment of lymphoma leukemia
BR112020020246A8 (en) 2018-04-05 2022-10-18 Sumitomo Dainippon Pharma Oncology Inc AXL KINASE INHIBITORS AND THEIR USE
US11299473B2 (en) 2018-04-13 2022-04-12 Incyte Corporation Benzimidazole and indole compounds and uses thereof
CN108658874B (en) * 2018-07-17 2020-04-14 大连医科大学 Thiopyrimidine heterocyclic antitumor compound and preparation method and application thereof
CA3224945A1 (en) 2018-07-31 2020-02-06 Loxo Oncology, Inc. Spray-dried dispersions, formulations, and polymorphs of (s)-5-amino-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1-(1,1,1-trifluoropropan-2-yl)-1h-pyrazole-4-carboxamide
US10899755B2 (en) 2018-08-08 2021-01-26 Incyte Corporation Benzothiazole compounds and uses thereof
JP7399968B2 (en) 2018-09-25 2023-12-18 インサイト・コーポレイション Pyrazolo[4,3-D]pyrimidine compounds as ALK2 and/or FGFR modulators
CN111170986A (en) * 2018-11-13 2020-05-19 北京睿熙生物科技有限公司 Inhibitors of bruton's tyrosine kinase
CN109593064B (en) * 2018-11-28 2020-08-11 北京博远精准医疗科技有限公司 Dithiocarbamate compounds as BTK inhibitors
CN111233774B (en) * 2018-11-28 2023-04-14 鲁南制药集团股份有限公司 Amino pyrimidine compound
WO2020131674A1 (en) 2018-12-19 2020-06-25 Array Biopharma Inc. 7-((3,5-dimethoxyphenyl)amino)quinoxaline derivatives as fgfr inhibitors for treating cancer
WO2020131627A1 (en) 2018-12-19 2020-06-25 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyridine compounds as inhibitors of fgfr tyrosine kinases
CN109776495A (en) * 2019-01-31 2019-05-21 李佳睿 Antitumoral compounds and preparation method thereof and purposes
MX2021012824A (en) 2019-05-01 2022-10-19 Clexio Biosciences Ltd Methods of treating pruritus.
CA3139708A1 (en) 2019-05-10 2020-11-19 Deciphera Pharmaceuticals, Llc Phenylaminopyrimidine amide autophagy inhibitors and methods of use thereof
AU2020274011B2 (en) 2019-05-10 2024-02-15 Deciphera Pharmaceuticals, Llc Heteroarylaminopyrimidine amide autophagy inhibitors and methods of use thereof
MA56191A (en) 2019-06-17 2022-04-20 Deciphera Pharmaceuticals Llc AMINOPYRIMIDINE AMIDE AUTOPHAGY INHIBITORS AND METHODS OF USE THEREOF
CN110407812B (en) * 2019-07-31 2020-08-25 武汉工程大学 Indazole piperidine pyrimidine derivative and preparation method and application thereof
CN114450276A (en) 2019-08-06 2022-05-06 因赛特公司 Solid forms of HPK1 inhibitor
KR102168179B1 (en) * 2019-10-31 2020-10-20 주식회사 온코빅스 Novel pyrimidine derivative showing growth inhibition of cancer cell and pharmaceutical composition comprising the same
WO2022059996A1 (en) * 2020-09-15 2022-03-24 서울대학교 기술지주 주식회사 Blood circulation micro in-vitro corpuscle-mediated cancer treatment composition
KR20230107207A (en) * 2020-09-22 2023-07-14 베이진 엘티디 Indoline compounds and derivatives as EGFR inhibitors
WO2022071772A1 (en) * 2020-09-29 2022-04-07 (주)메디톡스 Protein kinase inhibitor and use thereof
EP4279486A1 (en) * 2021-01-11 2023-11-22 Guangzhou Salustier Biosciences Co., Ltd. 2-aminopyrimidine compound and pharmaceutical composition thereof and application thereof
EP4313023A1 (en) 2021-04-02 2024-02-07 Biogen MA Inc. Combination treatment methods of multiple sclerosis
WO2022216097A1 (en) * 2021-04-08 2022-10-13 주식회사 스탠다임 Novel lrrk2 inhibitor
WO2022253333A1 (en) * 2021-06-02 2022-12-08 南京明德新药研发有限公司 Amide compounds and application thereof
CN114181161B (en) * 2021-12-28 2024-02-27 南通大学 (2- ((substituted oxy) phenyl) amino) pyrimidin-4-yl) aminobenzoyl derivative and preparation method and application thereof
CN115448906A (en) * 2022-09-26 2022-12-09 深圳大学 2-anilinopyrimidine derivative and preparation method and application thereof

Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997019065A1 (en) * 1995-11-20 1997-05-29 Celltech Therapeutics Limited Substituted 2-anilinopyrimidines useful as protein kinase inhibitors
EP1054004A1 (en) * 1997-12-15 2000-11-22 Yamanouchi Pharmaceutical Co. Ltd. Novel pyrimidine-5-carboxamide derivatives
WO2001064654A1 (en) * 2000-03-01 2001-09-07 Astrazeneca Ab Pyrimidine compounds
WO2005026130A1 (en) * 2003-09-18 2005-03-24 Novartis Ag 2,4-di (phenylamino) pyrimidines useful in the treatment of proliferative disorders
WO2005026158A1 (en) * 2003-09-16 2005-03-24 Novartis Ag 2,4 di (hetero) -arylamino-pyrimidine derivatives as zap-70 and/or syk inhibitors
WO2006021544A1 (en) * 2004-08-20 2006-03-02 Boehringer Ingelheim International Gmbh 2,4-di(aminophenyl) pyrimidines as plk inhibitors
WO2006074057A2 (en) * 2004-12-30 2006-07-13 Exelixis, Inc. Pyrimidine derivatives as kinase modulators and method of use
WO2006101977A2 (en) * 2005-03-16 2006-09-28 Targegen, Inc. Pyrimidine compounds and methods of use
WO2006108487A1 (en) * 2005-04-12 2006-10-19 Merck Patent Gmbh (lh-ind0l-7-yl)-( (pyrimidin-2 -yl-amino) methanone derivatives and related compounds as igf-rl inhibitors for treating cancer
WO2006129100A1 (en) * 2005-06-03 2006-12-07 Glaxo Group Limited Novel compounds
WO2006133426A2 (en) * 2005-06-08 2006-12-14 Rigel Pharmaceuticals, Inc. Compositions and methods for inhibition of the jak pathway
WO2007056151A2 (en) * 2005-11-03 2007-05-18 Irm Llc Protein kinase inhbitors
WO2007085833A2 (en) * 2006-01-26 2007-08-02 Astrazeneca Ab Pyrimidine derivatives
WO2007120339A1 (en) * 2005-12-19 2007-10-25 Genentech, Inc. Pyrimidine kinase inhibitors
WO2008049123A2 (en) * 2006-10-19 2008-04-24 Rigel Pharmaceuticals, Inc. 2,4-pyrimidinediamine derivatives as inhibitors of jak kinases for the treatment of autoimmune diseases
WO2008073687A2 (en) * 2006-12-08 2008-06-19 Irm Llc Compounds and compositions as protein kinase inhibitors
WO2008074515A1 (en) * 2006-12-20 2008-06-26 Bayer Schering Pharma Aktiengesellschaft Novel hetaryl-phenylene-diamine pyrimidine as protein kinase inhibitors
WO2008079719A1 (en) * 2006-12-19 2008-07-03 Genentech, Inc. Pyrimidine kinase inhibitors
WO2008092199A1 (en) * 2007-01-31 2008-08-07 Cytopia Research Pty Ltd Thiopyrimidine-based compounds and uses thereof
WO2008107096A1 (en) * 2007-03-02 2008-09-12 Bayer Cropscience Ag Diaminopyrimidines as fungicides
WO2009115267A2 (en) * 2008-03-20 2009-09-24 Bayer Cropscience Ag Diaminopyrimidines as plant protection agents

Family Cites Families (151)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US751444A (en) * 1904-02-09 Process of making pigments
US4650750A (en) 1982-02-01 1987-03-17 Giese Roger W Method of chemical analysis employing molecular release tag compounds
US5650270A (en) 1982-02-01 1997-07-22 Northeastern University Molecular analytical release tags and their use in chemical analysis
US4709016A (en) 1982-02-01 1987-11-24 Northeastern University Molecular analytical release tags and their use in chemical analysis
US5516931A (en) 1982-02-01 1996-05-14 Northeastern University Release tag compounds producing ketone signal groups
GB8608335D0 (en) 1986-04-04 1986-05-08 Pfizer Ltd Pharmaceutically acceptable salts
JPH0741461A (en) 1993-05-27 1995-02-10 Eisai Co Ltd Sulfonic acid ester derivative
DE69630214T2 (en) 1995-03-10 2004-07-15 Berlex Laboratories, Inc., Richmond BENZAMIDINE DERIVATIVES, THEIR PRODUCTION AND THEIR USE AS ANTI-COAGULANTS
GB9619284D0 (en) 1996-09-16 1996-10-30 Celltech Therapeutics Ltd Chemical compounds
GB9622363D0 (en) 1996-10-28 1997-01-08 Celltech Therapeutics Ltd Chemical compounds
US6303652B1 (en) 1998-08-21 2001-10-16 Hughes Institute BTK inhibitors and methods for their identification and use
ES2274634T3 (en) 1998-08-29 2007-05-16 Astrazeneca Ab PIRIMIDINE COMPOUNDS.
JP3635238B2 (en) 1998-11-10 2005-04-06 ジヤンセン・フアーマシユーチカ・ナームローゼ・フエンノートシヤツプ Pyrimidines that inhibit HIV replication
US6262088B1 (en) 1998-11-19 2001-07-17 Berlex Laboratories, Inc. Polyhydroxylated monocyclic N-heterocyclic derivatives as anti-coagulants
US6127376A (en) 1998-12-04 2000-10-03 Berlex Laboratories, Inc. Aryl and heterocyclyl substituted pyrimidine derivatives as anti-coagulants
GB9828511D0 (en) 1998-12-24 1999-02-17 Zeneca Ltd Chemical compounds
US6495558B1 (en) 1999-01-22 2002-12-17 Amgen Inc. Kinase inhibitors
AU2871900A (en) 1999-02-04 2000-08-25 Millennium Pharmaceuticals, Inc. G-protein coupled heptahelical receptor binding compounds and methods of use thereof
GB9914258D0 (en) 1999-06-18 1999-08-18 Celltech Therapeutics Ltd Chemical compounds
PL352476A1 (en) 1999-06-29 2003-08-25 Egyt Gyogyszervegyeszeti Gyar Novel derivatives of piperazinylalkylthiopyrimidine, parmacological compositions containing tese compounds and method of obtaining active substance
AU2457201A (en) 1999-12-28 2001-07-09 Bristol-Myers Squibb Company Cytokine, especially tnf-alpha, inhibitors
HUP0301117A3 (en) * 2000-02-17 2004-01-28 Amgen Inc Thousand Oaks Imidazole derivatives kinase inhibitors, their use, process for their preparation and pharmaceutical compositions containing them
GB0004887D0 (en) 2000-03-01 2000-04-19 Astrazeneca Uk Ltd Chemical compounds
GB0004890D0 (en) 2000-03-01 2000-04-19 Astrazeneca Uk Ltd Chemical compounds
ES2559273T3 (en) 2000-05-08 2016-02-11 Janssen Pharmaceutica Nv Pyrimidine prodrugs that inhibit HIV replication
GB0016877D0 (en) * 2000-07-11 2000-08-30 Astrazeneca Ab Chemical compounds
US7427689B2 (en) 2000-07-28 2008-09-23 Georgetown University ErbB-2 selective small molecule kinase inhibitors
US6881737B2 (en) 2001-04-11 2005-04-19 Amgen Inc. Substituted triazinyl acrylamide derivatives and methods of use
JO3429B1 (en) * 2001-08-13 2019-10-20 Janssen Pharmaceutica Nv Hiv inhibiting pyrimidines derivatives
US6939874B2 (en) 2001-08-22 2005-09-06 Amgen Inc. Substituted pyrimidinyl derivatives and methods of use
WO2003030909A1 (en) 2001-09-25 2003-04-17 Bayer Pharmaceuticals Corporation 2- and 4-aminopyrimidines n-substtituded by a bicyclic ring for use as kinase inhibitors in the treatment of cancer
JP2005507423A (en) 2001-11-01 2005-03-17 ジヤンセン・フアーマシユーチカ・ナームローゼ・フエンノートシヤツプ Heteroarylamines as glycogen synthase kinase 3 beta inhibitors (GSK3 inhibitors)
TWI329105B (en) 2002-02-01 2010-08-21 Rigel Pharmaceuticals Inc 2,4-pyrimidinediamine compounds and their uses
MXPA04007637A (en) 2002-02-08 2004-11-10 Smithkline Beecham Corp Pyrimidine compounds.
GB0206215D0 (en) 2002-03-15 2002-05-01 Novartis Ag Organic compounds
WO2003081210A2 (en) 2002-03-21 2003-10-02 Sunesis Pharmaceuticals, Inc. Identification of kinase inhibitors
US20040002395A1 (en) 2002-06-27 2004-01-01 Poynor Raymond L. Bridge weight for metal wood golf club
CN103169708B (en) 2002-07-29 2018-02-02 里格尔药品股份有限公司 The method that autoimmune disease is treated or prevented with 2,4 pyrimidinediamine compounds
CA2439440A1 (en) 2002-09-05 2004-03-05 Emory University Treatment of tuberous sclerosis associated neoplasms
AU2002951853A0 (en) 2002-10-04 2002-10-24 Commonwealth Scientific And Industrial Research Organisation Crystal structure of erbb2 and uses thereof
AU2002350719A1 (en) 2002-11-29 2004-06-23 Janssen Pharmaceutica N.V. Pharmaceutical compositions comprising a basic respectively acidic drug compound, a surfactant and a physiologically tolerable water-soluble acid respectively base
UA80767C2 (en) 2002-12-20 2007-10-25 Pfizer Prod Inc Pyrimidine derivatives for the treatment of abnormal cell growth
CL2004000192A1 (en) 2003-02-07 2005-03-18 Janssen Pharmaceutica Nv USE OF COMPOUNDS DERIVED FROM 2-AMINOPIRIMIDINE TO PREPARE A MEDICINAL PRODUCT FOR THE PREVENTION OF HIV INFECTION SEXUAL RELATIONS OR AN INTIMATE COUPLE CONTACT.
ES2325440T3 (en) * 2003-02-20 2009-09-04 Smithkline Beecham Corporation PIRIMIDINE COMPOUNDS.
RU2400477C2 (en) * 2003-03-14 2010-09-27 Новартис Аг 2,4-di(phenylamino)pyrimidines, applied in treatment of neoplastic diseases, inflammatory disorders and immune system disorders
GB0305929D0 (en) 2003-03-14 2003-04-23 Novartis Ag Organic compounds
US20050014753A1 (en) 2003-04-04 2005-01-20 Irm Llc Novel compounds and compositions as protein kinase inhibitors
US20050043233A1 (en) 2003-04-29 2005-02-24 Boehringer Ingelheim International Gmbh Combinations for the treatment of diseases involving cell proliferation, migration or apoptosis of myeloma cells or angiogenesis
US7504396B2 (en) * 2003-06-24 2009-03-17 Amgen Inc. Substituted heterocyclic compounds and methods of use
PL1656372T3 (en) 2003-07-30 2013-08-30 Rigel Pharmaceuticals Inc 2,4-pyrimidinediamine compounds for use in the treatment or prevention of autoimmune diseases
SI1663242T1 (en) 2003-08-07 2011-09-30 Rigel Pharmaceuticals Inc 2,4-pyrimidinediamine compounds and uses as anti-proliferative agents
ES2380206T3 (en) 2003-08-15 2012-05-09 Novartis Ag 2,4-Pyrimidinediamines useful for the treatment of neoplastic diseases, inflammatory and immune system disorders
EP1694652A1 (en) 2003-12-19 2006-08-30 Rigel Pharmaceuticals, Inc. Stereoisomers and stereoisomeric mixtures of 1-(2,4-pyrimidinediamino)-2-cyclopentanecarboxamide synthetic intermediates
PT1704145E (en) 2004-01-12 2012-09-04 Ym Biosciences Australia Pty Selective kinase inhibitors
UA85698C2 (en) 2004-01-16 2009-02-25 Уайет Quinoline intermediates of receptor tyrosine kinase inhibitors and the synthesis thereof
WO2005092836A1 (en) 2004-03-15 2005-10-06 Eli Lilly And Company Opioid receptor antagonists
US7558717B2 (en) 2004-04-28 2009-07-07 Vertex Pharmaceuticals Incorporated Crystal structure of human JAK3 kinase domain complex and binding pockets thereof
JP4812763B2 (en) 2004-05-18 2011-11-09 ライジェル ファーマシューティカルズ, インコーポレイテッド Cycloalkyl-substituted pyrimidinediamine compounds and uses thereof
EP1598343A1 (en) 2004-05-19 2005-11-23 Boehringer Ingelheim International GmbH 2-Arylaminopyrimidine derivatives as PLK inhibitors
ES2564127T5 (en) 2004-06-04 2020-03-26 Genentech Inc EGFR mutations
GB0419161D0 (en) 2004-08-27 2004-09-29 Novartis Ag Organic compounds
WO2006034473A2 (en) 2004-09-23 2006-03-30 Reddy Us Therapeutics, Inc. Novel pyrimidine compounds, process for their preparation and compositions containing them
RU2403244C2 (en) 2004-09-30 2010-11-10 Тиботек Фармасьютикалз Лтд. Hiv-inhibiting 5-carbo- or heterocyclic substituted pyrimidines
WO2006044457A1 (en) 2004-10-13 2006-04-27 Wyeth N-benzenesulfonyl substituted anilino-pyrimidine analogs
WO2006045066A2 (en) 2004-10-20 2006-04-27 Proteolix, Inc. Labeled compounds for proteasome inhibition
WO2006053109A1 (en) 2004-11-10 2006-05-18 Synta Pharmaceuticals Corp. Heteroaryl compounds
GB2420559B (en) 2004-11-15 2008-08-06 Rigel Pharmaceuticals Inc Stereoisomerically enriched 3-aminocarbonyl bicycloheptene pyrimidinediamine compounds and their uses
CA2584295C (en) 2004-11-24 2014-08-26 Rigel Pharmaceuticals, Inc. Spiro-2, 4-pyrimidinediamine compounds and their uses
JP2008527007A (en) * 2005-01-14 2008-07-24 ミレニアム・ファーマシューティカルズ・インコーポレイテッド Cinnamide and hydrocinnamide derivatives having Raf-kinase inhibitory activity
DE602006010979D1 (en) 2005-01-19 2010-01-21 Rigel Pharmaceuticals Inc PRODRUGS FROM 2,4-PYRIMIDIN DIAMIN COMPOUNDS AND THEIR USES
CN101193905B (en) 2005-02-11 2014-06-25 纪念斯隆-凯特林癌症中心 Methods and compositions for detecting a drug resistant EGFR mutant
US20060270694A1 (en) * 2005-05-03 2006-11-30 Rigel Pharmaceuticals, Inc. JAK kinase inhibitors and their uses
WO2006124874A2 (en) 2005-05-12 2006-11-23 Kalypsys, Inc. Inhibitors of b-raf kinase
WO2006128129A2 (en) 2005-05-26 2006-11-30 Synta Pharmaceuticals Corp. Method for treating cancer
US20070032493A1 (en) 2005-05-26 2007-02-08 Synta Pharmaceuticals Corp. Method for treating B cell regulated autoimmune disorders
US20070203161A1 (en) 2006-02-24 2007-08-30 Rigel Pharmaceuticals, Inc. Compositions and methods for inhibition of the jak pathway
WO2007024680A1 (en) 2005-08-22 2007-03-01 Amgen Inc. Pyrazolopyridine and pyrazolopyrimidine compounds useful as kinase enzymes modulators
AU2006304875B2 (en) 2005-10-21 2013-01-17 Exelixis, Inc. Pyrazolo-pyrimidines as casein kinase II (CK2) modulators
RU2448959C2 (en) 2005-11-01 2012-04-27 Таргеджен, Инк. Bi-aryl-metha-pyrimidine kinase inhibitors
US7713987B2 (en) 2005-12-06 2010-05-11 Rigel Pharmaceuticals, Inc. Pyrimidine-2,4-diamines and their uses
KR20080110998A (en) 2006-01-30 2008-12-22 엑셀리시스, 인코포레이티드 4-aryl-2-mino-pyrimidines or 4-aryl-2-aminoalkyl-pyrimidines as jak-2 modulators and pharmaceutical compositions containing them
WO2007120980A2 (en) 2006-02-17 2007-10-25 Rigel Pharmaceuticals, Inc. 2,4-pyrimidinediamine compounds for treating or preventing autoimmune diseases
WO2007098507A2 (en) 2006-02-24 2007-08-30 Rigel Pharmaceuticals, Inc. Compositions and methods for inhibition of the jak pathway
RU2480464C2 (en) 2006-03-30 2013-04-27 Тиботек Фармасьютикалз Лтд. Hiv-inhibiting 5-amido-substituted pyrimidines
ES2353725T3 (en) 2006-03-30 2011-03-04 Tibotec Pharmaceuticals PYRIMIDINES REPLACED WITH 5- (HYDROXIMETHYLENE AND AMYNOMETHYLENE) THAT INHIBIT HIV.
GB0608386D0 (en) 2006-04-27 2006-06-07 Senexis Ltd Compounds
CA2651732C (en) 2006-05-18 2014-10-14 Mannkind Corporation Intracellular kinase inhibitors
DE102006027156A1 (en) 2006-06-08 2007-12-13 Bayer Schering Pharma Ag New sulfimide compounds are protein kinase inhibitors useful to treat e.g. cancer, Hodgkin's lymphoma, Kaposi's sarcoma, cardiovascular disease, Crohn's disease, endometriosis and hemangioma
CA2656290A1 (en) 2006-07-05 2008-01-10 Exelixis, Inc. Methods of using igf1r and abl kinase modulators
AU2007276369A1 (en) 2006-07-21 2008-01-24 Novartis Ag 2, 4 -di (arylaminio) -pyrimidine-5-carboxamide compounds as JAK kinases inhibitors
DE102006041382A1 (en) 2006-08-29 2008-03-20 Bayer Schering Pharma Ag Carbamoyl sulfoximides as protein kinase inhibitors
MX347525B (en) 2006-09-22 2017-04-27 Pharmacyclics Llc Inhibitors of bruton's tyrosine kinase.
CA2668286C (en) 2006-11-03 2014-09-16 Pharmacyclics, Inc. Bruton's tyrosine kinase activity probe and method of using
DK2078026T3 (en) 2006-11-21 2012-04-30 Rigel Pharmaceuticals Inc PRODRUG SALTS OF 2,4-PYRIMIDINE DIAMINE COMPOUNDS AND APPLICATIONS THEREOF
CN106045965A (en) 2006-12-29 2016-10-26 爱尔兰詹森科学公司 HIV inhibiting 5,6-substituted pyrimidines
US9006243B2 (en) 2006-12-29 2015-04-14 Janssen R&D Ireland HIV inhibiting 6-substituted pyrimidines
PE20081636A1 (en) 2007-01-26 2009-01-10 Smithkline Beecham Corp ANTHRANILAMIDE INHIBITORS FOR AURORA KINASE
JP4221470B2 (en) 2007-02-01 2009-02-12 トヨタ自動車株式会社 Electric vehicle control apparatus and electric vehicle control method
CN101677554A (en) 2007-03-20 2010-03-24 史密丝克莱恩比彻姆公司 Chemical compounds
CN101686675A (en) 2007-03-20 2010-03-31 史密丝克莱恩比彻姆公司 Compound
US7947698B2 (en) 2007-03-23 2011-05-24 Rigel Pharmaceuticals, Inc. Compositions and methods for inhibition of the JAK pathway
WO2008118823A2 (en) 2007-03-26 2008-10-02 Rigel Pharmaceuticals, Inc. Compositions and methods for inhibition of the jak pathway
US20120065201A1 (en) 2007-03-28 2012-03-15 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase
US20120101113A1 (en) 2007-03-28 2012-04-26 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase
US8242271B2 (en) 2007-06-04 2012-08-14 Avila Therapeutics, Inc. Heterocyclic compounds and uses thereof
US8993585B2 (en) * 2007-07-17 2015-03-31 Rigel Pharmaceuticals, Inc. Cyclic amine substituted pyrimidinediamines as PKC inhibitors
WO2009017838A2 (en) 2007-08-01 2009-02-05 Exelixis, Inc. Combinations of jak-2 inhibitors and other agents
WO2009029682A1 (en) 2007-08-28 2009-03-05 Rigel Pharmaceuticals, Inc. Combination therapy with syk kinase inhibitor
US8440681B2 (en) 2007-08-28 2013-05-14 Irm Llc 2-biphenylamino-4-aminopyrimidine derivatives as kinase inhibitors
WO2009032703A1 (en) 2007-08-28 2009-03-12 Irm Llc 2- (het) arylamino-6-aminopyridine derivatives and fused forms thereof as anaplastic lymphoma kinase inhibitors
CA2702674C (en) 2007-10-19 2016-05-03 Avila Therapeutics, Inc. Heteroaryl compounds and uses thereof
US7989465B2 (en) 2007-10-19 2011-08-02 Avila Therapeutics, Inc. 4,6-disubstituted pyrimidines useful as kinase inhibitors
CA2710118A1 (en) 2007-12-20 2009-07-02 Cellzome Limited Sulfamides as zap-70 inhibitors
SI2252300T1 (en) 2008-02-22 2017-04-26 Rigel Pharmaceuticals, Inc. Use of 2,4-pyrimidinediamines for the treatment of atherosclerosis
EP2276747A1 (en) 2008-03-11 2011-01-26 Cellzome Limited Sulfonamides as zap-70 inhibitors
US8205348B2 (en) 2008-03-19 2012-06-26 Zashiki-Warashi Manufacturing Inc. Tile spacer and holder therefor
WO2009127642A2 (en) 2008-04-15 2009-10-22 Cellzome Limited Use of lrrk2 inhibitors for neurodegenerative diseases
CA2728893C (en) 2008-04-16 2017-03-14 Portola Pharmaceuticals, Inc. Inhibitors of syk protein kinase
BRPI0910560B8 (en) 2008-04-16 2023-03-21 Portola Pharm Inc 2,6-DIAMINO-PYRIMIDIN-5-YL-CARBOXAMIDE COMPOUNDS AS INHIBITORS OF SYK OR JAK KINASES, COMPOSITION, METHOD FOR INHIBITING SYK, JAK KINASE OR A TRANSDUCTION PATHWAY SIGNAL MEDIATED AT LEAST BY SYK OR JAK ACTIVITY, AND KIT
US8138339B2 (en) * 2008-04-16 2012-03-20 Portola Pharmaceuticals, Inc. Inhibitors of protein kinases
WO2009132202A2 (en) * 2008-04-24 2009-10-29 Incyte Corporation Macrocyclic compounds and their use as kinase inhibitors
KR101860057B1 (en) 2008-05-21 2018-05-21 어리어드 파마슈티칼스, 인코포레이티드 Phosphorous derivatives as kinase inhibitors
RU2536584C2 (en) * 2008-06-27 2014-12-27 Авила Терапьютикс, Инк. Heteroaryl compounds and using them
US8338439B2 (en) 2008-06-27 2012-12-25 Celgene Avilomics Research, Inc. 2,4-disubstituted pyrimidines useful as kinase inhibitors
MX2011000661A (en) 2008-07-16 2011-05-25 Pharmacyclics Inc Inhibitors of bruton's tyrosine kinase for the treatment of solid tumors.
CN102203070A (en) 2008-09-03 2011-09-28 拜尔农作物科学股份公司 Alkoxy-substituted and alkylthio-substituted anilinopyrimidines
US20110245156A1 (en) 2008-12-09 2011-10-06 Cytokine Pharmasciences, Inc. Novel antiviral compounds, compositions, and methods of use
CA3018087C (en) 2009-01-15 2019-09-24 F. Hoffmann-La Roche Ag Antibodies against phosphorylated tyrosines on erythropoietin receptor (epor)
CA2757671A1 (en) 2009-04-03 2010-10-07 Cellzome Ag Methods for the identification of kinase interacting molecules and for the purification of kinase proteins
JP5918693B2 (en) 2009-05-05 2016-05-18 ダナ ファーバー キャンサー インスティテュート インコーポレイテッド EGFR inhibitor and disease treatment method
DK2428508T3 (en) 2009-05-08 2016-02-01 Astellas Pharma Inc Diamino heterocyclic carboxamide COMPOUND
US20120165332A1 (en) 2009-06-18 2012-06-28 Cellzome Limited Sulfonamides and sulfamides as zap-70 inhibitors
US7718662B1 (en) 2009-10-12 2010-05-18 Pharmacyclics, Inc. Pyrazolo-pyrimidine inhibitors of bruton's tyrosine kinase
WO2011079231A1 (en) 2009-12-23 2011-06-30 Gatekeeper Pharmaceutical, Inc. Compounds that modulate egfr activity and methods for treating or preventing conditions therewith
WO2011140338A1 (en) 2010-05-05 2011-11-10 Gatekeeper Pharmaceuticals, Inc. Compounds that modulate egfr activity and methods for treating or preventing conditions therewith
AU2011261185A1 (en) 2010-06-03 2013-01-10 Pharmacyclics, Inc. The use of inhibitors of Bruton's tyrosine kinase (Btk)
US20120071497A1 (en) 2010-06-03 2012-03-22 Pharmacyclics, Inc. Methods of treating abc-dlbcl using inhibitors of bruton's tyrosine kinase
WO2011153553A2 (en) 2010-06-04 2011-12-08 The Regents Of The University Of California Methods and compositions for kinase inhibition
SG187796A1 (en) 2010-08-10 2013-03-28 Celgene Avilomics Res Inc Besylate salt of a btk inhibitor
JP5956999B2 (en) 2010-11-01 2016-07-27 セルジーン アヴィロミクス リサーチ, インコーポレイテッド Heteroaryl compounds and uses thereof
MY181898A (en) 2010-11-01 2021-01-12 Celgene Car Llc Heterocyclic compounds and uses thereof
US20130317029A1 (en) 2010-11-01 2013-11-28 Portola Pharmaceuticals, Inc. Oxypyrimidines as syk modulators
US8796255B2 (en) 2010-11-10 2014-08-05 Celgene Avilomics Research, Inc Mutant-selective EGFR inhibitors and uses thereof
CN102558149A (en) 2010-12-29 2012-07-11 中国医学科学院药物研究所 Pyrimidine derivative, preparation method thereof, medicinal composition and application thereof
EA201391626A1 (en) 2011-05-04 2014-03-31 Ариад Фармасьютикалз, Инк. COMPOUNDS FOR INHIBITING CELL PROLIFERATION IN EGFR-STIMULATED CANCER TYPES
KR102052670B1 (en) 2011-05-17 2019-12-06 더 리젠츠 오브 더 유니버시티 오브 캘리포니아 Kinase inhibitors
SG10201702913XA (en) 2011-10-19 2017-06-29 Pharmacyclics Llc Use of inhibitors of bruton's tyrosine kinase (btk)
CN103159742B (en) 2011-12-16 2015-08-12 北京韩美药品有限公司 5-chloropyrimide compounds and the application as EGFR tyrosine kinase inhibitor thereof
KR20150080592A (en) 2012-11-02 2015-07-09 파마시클릭스, 인코포레이티드 Tec family kinase inhibitor adjuvant therapy
WO2015017812A1 (en) 2013-08-02 2015-02-05 Pharmacyclics, Inc. Methods for the treatment of solid tumors
KR102257121B1 (en) * 2013-08-20 2021-05-27 삼성전자주식회사 Method and system for dual role handling in a wireless environment

Patent Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997019065A1 (en) * 1995-11-20 1997-05-29 Celltech Therapeutics Limited Substituted 2-anilinopyrimidines useful as protein kinase inhibitors
EP1054004A1 (en) * 1997-12-15 2000-11-22 Yamanouchi Pharmaceutical Co. Ltd. Novel pyrimidine-5-carboxamide derivatives
WO2001064654A1 (en) * 2000-03-01 2001-09-07 Astrazeneca Ab Pyrimidine compounds
WO2005026158A1 (en) * 2003-09-16 2005-03-24 Novartis Ag 2,4 di (hetero) -arylamino-pyrimidine derivatives as zap-70 and/or syk inhibitors
WO2005026130A1 (en) * 2003-09-18 2005-03-24 Novartis Ag 2,4-di (phenylamino) pyrimidines useful in the treatment of proliferative disorders
WO2006021544A1 (en) * 2004-08-20 2006-03-02 Boehringer Ingelheim International Gmbh 2,4-di(aminophenyl) pyrimidines as plk inhibitors
WO2006074057A2 (en) * 2004-12-30 2006-07-13 Exelixis, Inc. Pyrimidine derivatives as kinase modulators and method of use
WO2006101977A2 (en) * 2005-03-16 2006-09-28 Targegen, Inc. Pyrimidine compounds and methods of use
WO2006108487A1 (en) * 2005-04-12 2006-10-19 Merck Patent Gmbh (lh-ind0l-7-yl)-( (pyrimidin-2 -yl-amino) methanone derivatives and related compounds as igf-rl inhibitors for treating cancer
WO2006129100A1 (en) * 2005-06-03 2006-12-07 Glaxo Group Limited Novel compounds
WO2006133426A2 (en) * 2005-06-08 2006-12-14 Rigel Pharmaceuticals, Inc. Compositions and methods for inhibition of the jak pathway
WO2007056151A2 (en) * 2005-11-03 2007-05-18 Irm Llc Protein kinase inhbitors
WO2007120339A1 (en) * 2005-12-19 2007-10-25 Genentech, Inc. Pyrimidine kinase inhibitors
WO2007085833A2 (en) * 2006-01-26 2007-08-02 Astrazeneca Ab Pyrimidine derivatives
WO2008049123A2 (en) * 2006-10-19 2008-04-24 Rigel Pharmaceuticals, Inc. 2,4-pyrimidinediamine derivatives as inhibitors of jak kinases for the treatment of autoimmune diseases
WO2008073687A2 (en) * 2006-12-08 2008-06-19 Irm Llc Compounds and compositions as protein kinase inhibitors
WO2008079719A1 (en) * 2006-12-19 2008-07-03 Genentech, Inc. Pyrimidine kinase inhibitors
WO2008074515A1 (en) * 2006-12-20 2008-06-26 Bayer Schering Pharma Aktiengesellschaft Novel hetaryl-phenylene-diamine pyrimidine as protein kinase inhibitors
WO2008092199A1 (en) * 2007-01-31 2008-08-07 Cytopia Research Pty Ltd Thiopyrimidine-based compounds and uses thereof
WO2008107096A1 (en) * 2007-03-02 2008-09-12 Bayer Cropscience Ag Diaminopyrimidines as fungicides
WO2009115267A2 (en) * 2008-03-20 2009-09-24 Bayer Cropscience Ag Diaminopyrimidines as plant protection agents

Cited By (476)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9133198B2 (en) 2006-09-22 2015-09-15 Pharmacyclics Llc Inhibitors of bruton'S tyrosine kinase
US8691546B2 (en) 2006-09-22 2014-04-08 Pharmacyclics, Inc. Inhibitors of Bruton's tyrosine kinase
US8759516B2 (en) 2006-09-22 2014-06-24 Pharmacyclics, Inc. Inhibitors of Bruton's tyrosine kinase
US8563563B2 (en) 2006-09-22 2013-10-22 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase
US9133201B2 (en) 2006-09-22 2015-09-15 Pharmacyclics, Inc. Inhibitors of Bruton's tyrosine kinase
US8748439B2 (en) 2006-09-22 2014-06-10 Pharmacyclics, Inc. Inhibitors of Bruton's tyrosine kinase
US8748438B2 (en) 2006-09-22 2014-06-10 Pharmacyclics, Inc. Inhibitors of Bruton's tyrosine kinase
US9409911B2 (en) 2006-09-22 2016-08-09 Pharmacyclics Llc Inhibitors of bruton's tyrosine kinase
US8741908B2 (en) 2006-09-22 2014-06-03 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase
US8735403B2 (en) 2006-09-22 2014-05-27 Pharmacyclics, Inc. Inhibitors of Bruton's tyrosine kinase
US8735404B2 (en) 2006-09-22 2014-05-27 Pharmacyclics, Inc. Inhibitors of Bruton's tyrosine kinase
US9266893B2 (en) 2006-09-22 2016-02-23 Pharmacyclics Llc Inhibitors of Bruton's tyrosine kinase
US9212185B2 (en) 2006-09-22 2015-12-15 Pharmacyclics Llc Inhibitors of Bruton's tyrosine kinase
US8883435B2 (en) 2006-09-22 2014-11-11 Pharmacyclics, Inc. Inhibitors of Bruton's tyrosine kinase
US8476284B2 (en) 2006-09-22 2013-07-02 Pharmacyclics, Inc. Inhibitors of Bruton's tyrosine kinase
US9206189B2 (en) 2006-09-22 2015-12-08 Pharmacyclics Llc Inhibitors of bruton's tyrosine kinase
US9193735B2 (en) 2006-09-22 2015-11-24 Pharmacyclics Llc Inhibitors of Bruton's tyrosine kinase
US8497277B2 (en) 2006-09-22 2013-07-30 Pharmacyclics, Inc. Inhibitors of Bruton's tyrosine kinase
US8501751B2 (en) 2006-09-22 2013-08-06 Pharmacyclics, Inc. Inhibitors of Bruton's tyrosine kinase
US9181257B2 (en) 2006-09-22 2015-11-10 Pharmacyclics Llc Inhibitors of Bruton's tyrosine kinase
US8703780B2 (en) 2006-09-22 2014-04-22 Pharmacyclics, Inc. Inhibitors of Bruton's tyrosine kinase
US8697711B2 (en) 2006-09-22 2014-04-15 Pharmacyclics, Inc. Inhibitors of bruton'S tyrosine kinase
US8552010B2 (en) 2006-09-22 2013-10-08 Pharmacyclics, Inc. Inhibitors of Bruton'S tyrosine kinase
US9133202B2 (en) 2006-09-22 2015-09-15 Pharmacyclics, Inc. Inhibitors of Bruton's tyrosine kinase
US8754091B2 (en) 2006-09-22 2014-06-17 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase
US8952015B2 (en) 2006-09-22 2015-02-10 Pharmacyclics, Inc. Inhibitors of Bruton's tyrosine kinase
US8957079B2 (en) 2006-09-22 2015-02-17 Pharmacyclics, Inc. Inhibitors of Bruton's tyrosine kinase
US9127012B2 (en) 2006-09-22 2015-09-08 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase
US8975266B2 (en) 2006-09-22 2015-03-10 Pharmacyclics, Inc. Inhibitors of Bruton's tyrosine kinase
US8658653B2 (en) 2006-09-22 2014-02-25 Pharmacyclics, Inc. Inhibitors of Bruton's tyrosine kinase
US8940750B2 (en) 2007-03-28 2015-01-27 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase
US9139591B2 (en) 2007-03-28 2015-09-22 Pharmacyclics Llc Inhibitors of bruton's tyrosine kinase
US9079908B2 (en) 2007-03-28 2015-07-14 Pharmacyclics, Inc. Inhibitors of Bruton'S tyrosine kinase
US9181263B2 (en) 2007-03-28 2015-11-10 Pharmacyclics Llc Inhibitors of bruton's tyrosine kinase
US8809273B2 (en) 2007-03-28 2014-08-19 Pharmacyclics, Inc. Inhibitors of Bruton's tyrosine kinase
US9556182B2 (en) 2007-03-28 2017-01-31 Pharmacylics LLC Inhibitors of Bruton's tyrosine kinase
US9012462B2 (en) 2008-05-21 2015-04-21 Ariad Pharmaceuticals, Inc. Phosphorous derivatives as kinase inhibitors
US9273077B2 (en) 2008-05-21 2016-03-01 Ariad Pharmaceuticals, Inc. Phosphorus derivatives as kinase inhibitors
US9296737B2 (en) 2008-06-27 2016-03-29 Celgene Avilomics Research, Inc. Substituted 2,4-diaminopyrimidines as kinase inhibitors
US8609679B2 (en) 2008-06-27 2013-12-17 Celgene Avilomics Research, Inc. 2,4-diaminopyrimidines useful as kinase inhibitors
US10010548B2 (en) 2008-06-27 2018-07-03 Celgene Car Llc 2,4-disubstituted pyrimidines useful as kinase inhibitors
US8710222B2 (en) 2008-06-27 2014-04-29 Celgene Avilomics Research, Inc. 2,4-disubstituted pyrimidines useful as kinase inhibitors
US9212181B2 (en) 2008-06-27 2015-12-15 Celgene Avilomics Research, Inc. Substituted 2,4-diaminopyrimidines as kinase inhibitors
US9987276B2 (en) 2008-06-27 2018-06-05 Celgene Car Llc Substituted 2,4-diaminopyrimidines as kinase inhibitors
US10596172B2 (en) 2008-06-27 2020-03-24 Celgene Car Llc 2,4-disubstituted pyrimidines useful as kinase inhibitors
US11351168B1 (en) 2008-06-27 2022-06-07 Celgene Car Llc 2,4-disubstituted pyrimidines useful as kinase inhibitors
US9409921B2 (en) 2008-06-27 2016-08-09 Celgene Avilomics Research, Inc. 2,4-disubstituted pyrimidines as kinase inhibitors
US10828300B2 (en) 2008-06-27 2020-11-10 Celgene Car Llc Substituted 2,4-diaminopyrimidines as kinase inhibitors
US8883803B2 (en) 2008-07-16 2014-11-11 Pharmacyclics, Inc. Inhibitors of Bruton's tyrosine kinase for the treatment of solid tumors
US9278100B2 (en) 2008-07-16 2016-03-08 Pharmacyclics Llc Inhibitors of bruton's tyrosine kinase for the treatment of solid tumors
US9795605B2 (en) 2008-07-16 2017-10-24 Pharmacyclics Llc Inhibitors of Bruton's tyrosine kinase for the treatment of solid tumors
US9107924B2 (en) 2008-07-16 2015-08-18 Pharmacyclics, Inc. Inhibitors of Bruton'S tyrosine kinase for the treatment of solid tumors
US8846689B2 (en) 2008-11-24 2014-09-30 Boehringer Ingelheim International Gmbh Substituted pyrimidines for the treatment of diseases such as cancer
US8785464B2 (en) 2008-11-24 2014-07-22 Boehringer Ingelheim International Gmbh Pyrimidine derivatives that inhibit FAK/PTK2
US9676762B2 (en) 2008-11-24 2017-06-13 Boehringer Ingelheim International Gmbh Pyrimidine compounds containing seven-membered fused ring systems
JP2012515724A (en) * 2009-01-21 2012-07-12 ライジェル ファーマシューティカルズ, インコーポレイテッド N2- (3-pyridyl or phenyl) -N4- (4-piperidyl) -2,4-pyrimidinediamine derivatives useful for the treatment of inflammatory diseases, autoimmune diseases or proliferative diseases
US9149475B2 (en) 2009-01-21 2015-10-06 Rigel Pharmaceuticals, Inc. Protein kinase C inhibitors and uses thereof
EP2440559A2 (en) 2009-05-05 2012-04-18 Dana-Farber Cancer Institute, Inc. Egfr inhibitors and methods of treating disorders
US9908884B2 (en) 2009-05-05 2018-03-06 Dana-Farber Cancer Institute, Inc. EGFR inhibitors and methods of treating disorders
US8586751B2 (en) 2009-06-12 2013-11-19 Bristol-Myers Squibb Company Nicotinamide compounds useful as kinase modulators
WO2010144647A1 (en) * 2009-06-12 2010-12-16 Bristol-Myers Squibb Company Nicotinamide compounds useful as kinase modulators
WO2011018517A1 (en) 2009-08-14 2011-02-17 Boehringer Ingelheim International Gmbh Regioselective preparation of 2-amino-5-trifluoromethylpyrimidine derivatives
WO2011018518A1 (en) 2009-08-14 2011-02-17 Boehringer Ingelheim International Gmbh Regioselective preparation of 2 -amino-5-trifluoromethylpyrimidine derivatives
US10662195B2 (en) 2009-09-16 2020-05-26 Celgene Car Llc Protein kinase conjugates and inhibitors
US9556426B2 (en) 2009-09-16 2017-01-31 Celgene Avilomics Research, Inc. Protein kinase conjugates and inhibitors
US8466155B2 (en) 2009-10-02 2013-06-18 Boehringer Ingelheim International Gmbh Pyrimidines
WO2011068898A1 (en) * 2009-12-01 2011-06-09 Rigel Pharmaceuticals, Inc. Protein kinase c inhibitors and uses thereof
US9732070B2 (en) 2009-12-01 2017-08-15 Rigel Pharmaceuticals, Inc. Protein kinase C inhibitors and uses thereof
AU2010343055B2 (en) * 2009-12-29 2016-11-10 Celgene Car Llc Heteroaryl compounds and uses thereof
EP2519664A1 (en) * 2009-12-30 2012-11-07 Avila Therapeutics, Inc. Ligand-directed covalent modification of protein
US11542492B2 (en) 2009-12-30 2023-01-03 Celgene Car Llc Ligand-directed covalent modification of protein
EP2519664A4 (en) * 2009-12-30 2014-03-12 Avila Therapeutics Inc Ligand-directed covalent modification of protein
JP2013523891A (en) * 2010-04-13 2013-06-17 ライジェル ファーマシューティカルズ, インコーポレイテッド 2,4-pyrimidinediamine compounds and prodrugs thereof and uses thereof
US10478439B2 (en) 2010-06-03 2019-11-19 Pharmacyclics Llc Use of inhibitors of bruton's tyrosine kinase (Btk)
US9801883B2 (en) 2010-06-03 2017-10-31 Pharmacyclics Llc Use of inhibitors of bruton's tyrosine kinase (Btk)
US10004746B2 (en) 2010-06-03 2018-06-26 Pharmacyclics Llc Use of inhibitors of Bruton's tyrosine kinase (Btk)
US10004745B2 (en) 2010-06-03 2018-06-26 Pharmacyclics Llc Use of inhibitors of Bruton'S tyrosine kinase (Btk)
US10751342B2 (en) 2010-06-03 2020-08-25 Pharmacyclics Llc Use of inhibitors of Bruton's tyrosine kinase (Btk)
US9801881B2 (en) 2010-06-03 2017-10-31 Pharmacyclics Llc Use of inhibitors of bruton's tyrosine kinase (BTK)
US9125889B2 (en) 2010-06-03 2015-09-08 Pharmacyclics, Inc. Use of inhibitors of Bruton's tyrosine kinase (Btk)
US10016435B2 (en) 2010-06-03 2018-07-10 Pharmacyclics Llc Use of inhibitors of Bruton's tyrosine kinase (Btk)
US8999999B2 (en) 2010-06-03 2015-04-07 Pharmacyclics, Inc. Use of inhibitors of Bruton's tyrosine kinase (Btk)
US10653696B2 (en) 2010-06-03 2020-05-19 Pharmacyclics Llc Use of inhibitors of bruton's tyrosine kinase (BTK)
US11672803B2 (en) 2010-06-03 2023-06-13 Pharmacyclics Llc Use of inhibitors of Brutons tyrosine kinase (Btk)
US8754090B2 (en) 2010-06-03 2014-06-17 Pharmacyclics, Inc. Use of inhibitors of bruton's tyrosine kinase (Btk)
US9814721B2 (en) 2010-06-03 2017-11-14 Pharmacyclics Llc Use of inhibitors of bruton'S tyrosine kinase (BTK)
KR20140060473A (en) * 2010-06-23 2014-05-20 한미사이언스 주식회사 Novel fused pyrimidine derivatives for inhibition of tyrosine kinase activity
KR101587506B1 (en) 2010-06-23 2016-01-25 한미사이언스 주식회사 Novel fused pyrimidine derivatives for inhibition of tyrosine kinase activity
US9345719B2 (en) 2010-06-23 2016-05-24 Hanmi Science Co., Ltd. Fused pyrimidine derivatives for inhibition of tyrosine kinase activity
AU2014202057B2 (en) * 2010-06-23 2016-05-05 Hanmi Science Co., Ltd. Novel Fused Pyrimidine Derivatives for Inhibition of Tyrosine Kinase Activity
USRE46511E1 (en) 2010-06-23 2017-08-15 Hanmi Science Co., Ltd. Fused pyrimidine derivatives for inhibition of tyrosine kinase activity
CN102947316A (en) * 2010-06-23 2013-02-27 韩美科学株式会社 Novel fused pyrimidine derivatives for inhibition of tyrosine kinase activity
AU2011269989B2 (en) * 2010-06-23 2014-12-11 Hanmi Science Co., Ltd. Novel fused pyrimidine derivatives for inhibition of tyrosine kinase activity
EP2975042A1 (en) * 2010-06-23 2016-01-20 Boehringer Ingelheim International GmbH Novel fused pyrimidine derivatives for inhibition of tyrosine kinase activity
EP2585470A4 (en) * 2010-06-23 2014-01-01 Hanmi Science Co Ltd Novel fused pyrimidine derivatives for inhibition of tyrosine kinase activity
EP2585470A2 (en) * 2010-06-23 2013-05-01 Hanmi Science Co., Ltd. Novel fused pyrimidine derivatives for inhd3ition of tyrosine kinase activity
JP2014159473A (en) * 2010-06-23 2014-09-04 Hanmi Science Co Ltd Novel fused pyrimidine derivatives with tyrosine kinase activity inhibitory action
JP2013529630A (en) * 2010-06-23 2013-07-22 ハンミ・サイエンス・カンパニー・リミテッド Novel condensed pyrimidine derivatives having tyrosine kinase activity inhibitory action
KR101589114B1 (en) 2010-06-23 2016-01-29 한미사이언스 주식회사 Novel fused pyrimidine derivatives for inhibition of tyrosine kinase activity
TWI513701B (en) * 2010-06-23 2015-12-21 Hanmi Science Co Ltd Novel fused pyrimidine derivatives for inhibition of tyrosine kinase activity
CN102947316B (en) * 2010-06-23 2016-08-10 韩美科学株式会社 For suppressing the novel fused pyrimidine derivatives of tyrosine kinase activity
KR20110139653A (en) * 2010-06-23 2011-12-29 한미홀딩스 주식회사 Novel fused pyrimidine derivatives for inhibition of tyrosine kinase activity
US8957065B2 (en) 2010-06-23 2015-02-17 Hanmi Science Co., Ltd Fused pyrimidine derivatives for inhibition of tyrosine kinase activity
CN105566229A (en) * 2010-08-10 2016-05-11 西建阿维拉米斯研究公司 Besylate salt of a BTK inhibitor, application and preparation method thereof
US8563568B2 (en) 2010-08-10 2013-10-22 Celgene Avilomics Research, Inc. Besylate salt of a BTK inhibitor
US9604936B2 (en) 2010-08-10 2017-03-28 Celgene Car Llc Besylate salt of a BTK inhibitor
JP2017036268A (en) * 2010-08-18 2017-02-16 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Merck Patent Gesellschaft mit beschraenkter Haftung Pyrimidine derivative as fak inhibitor
JP2013539795A (en) * 2010-10-14 2013-10-28 アリアド・ファーマシューティカルズ・インコーポレイテッド Method for inhibiting cell proliferation of EGFR-activated cancer
US9867824B2 (en) 2010-11-01 2018-01-16 Celgene Car Llc Heterocyclic compounds and uses thereof
US9765038B2 (en) 2010-11-01 2017-09-19 Celgene Car Llc Heteroaryl compounds and uses thereof
EP2635285A4 (en) * 2010-11-01 2014-07-23 Celgene Avilomics Res Inc Heteroaryl compounds and uses thereof
JP2016185975A (en) * 2010-11-01 2016-10-27 セルジーン アヴィロミクス リサーチ, インコーポレイテッド Heterocyclic compound and use thereof
EP2635284A4 (en) * 2010-11-01 2014-07-09 Celgene Avilomics Res Inc Heterocyclic compounds and uses thereof
EP2635285A1 (en) * 2010-11-01 2013-09-11 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
CN103269704A (en) * 2010-11-01 2013-08-28 西建阿维拉米斯研究公司 Heterocyclic compounds and uses thereof
KR20130133202A (en) * 2010-11-01 2013-12-06 셀진 아빌로믹스 리서치, 인코포레이티드 Heterocyclic compounds and uses thereof
US9375431B2 (en) 2010-11-01 2016-06-28 Celgene Avilomics Research, Inc. 2,4-disubstituted pyrimidine compounds useful as kinase inhibtors
JP2013544807A (en) * 2010-11-01 2013-12-19 セルジーン アヴィロミクス リサーチ, インコーポレイテッド Heteroaryl compounds and uses thereof
US10434101B2 (en) 2010-11-01 2019-10-08 Celgene Car Llc Heterocyclic compounds and uses thereof
EP2635284A1 (en) * 2010-11-01 2013-09-11 Celgene Avilomics Research, Inc. Heterocyclic compounds and uses thereof
US8975249B2 (en) 2010-11-01 2015-03-10 Celgene Avilomics Research, Inc. Heterocyclic compounds and uses thereof
US11096942B2 (en) 2010-11-01 2021-08-24 Celgene Car Llc Heterocyclic compounds and uses thereof
KR101954593B1 (en) * 2010-11-01 2019-03-06 셀젠 카르 엘엘씨 Heterocyclic compounds and uses thereof
AU2016210613B2 (en) * 2010-11-01 2018-03-01 Celgene Car Llc Heterocyclic compounds and uses thereof
US9238629B2 (en) 2010-11-01 2016-01-19 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
US10081606B2 (en) 2010-11-01 2018-09-25 Celgene Car Llc Heteroaryl compounds and uses thereof
TWI632134B (en) * 2010-11-01 2018-08-11 阿維拉製藥公司 Heterocyclic compounds and uses thereof
US9868723B2 (en) 2010-11-10 2018-01-16 Celgene Car Llc Mutant-selective EGFR inhibitors and uses thereof
JP2013542256A (en) * 2010-11-10 2013-11-21 セルジーン アヴィロミクス リサーチ, インコーポレイテッド Mutant selective EGFR inhibitor and use thereof
US9409887B2 (en) 2010-11-10 2016-08-09 Celgene Avilomics Research, Inc. Mutant-selective EGFR inhibitors and uses thereof
US9133187B2 (en) 2011-02-28 2015-09-15 Array Biopharma Inc. Serine/threonine kinase inhibitors
US10266500B2 (en) 2011-04-22 2019-04-23 Signal Pharmaceuticals, Llc Substituted diaminocarboxamide and diaminocarbonitrile pyrimidines, compositions thereof, and methods of treatment therewith
US10919865B2 (en) 2011-04-22 2021-02-16 Signal Pharmaceuticals, Llc Substituted diaminocarboxamide and diaminocarbonitrile pyrimidines, compositions thereof, and methods of treatment therewith
US9139534B2 (en) 2011-04-22 2015-09-22 Signal Pharmaceuticals, Llc Substituted diaminocarboxamide and diaminocarbonitrile pyrimidines, compositions thereof, and methods of treatment therewith
US10040770B2 (en) 2011-04-22 2018-08-07 Signal Pharmaceuticals, Llc Substituted diaminocarboxamide and diaminocarbonitrile pyrimidines, compositions thereof, and methods of treatment therewith
US9701643B2 (en) 2011-04-22 2017-07-11 Signal Pharmaceuticals, Llc Substituted diaminocarboxamide and diaminocarbonitrile pyrimidines, compositions thereof, and methods of treatment therewith
US11325890B2 (en) 2011-04-22 2022-05-10 Signal Pharmaceuticals, Llc Substituted diaminocarboxamide and diaminocarbonitrile pyrimidines, compositions thereof, and methods of treatment therewith
JP2014514348A (en) * 2011-05-04 2014-06-19 アリアド・ファーマシューティカルズ・インコーポレイテッド Compound for inhibiting cell proliferation of EGFR-activated cancer
US9834518B2 (en) 2011-05-04 2017-12-05 Ariad Pharmaceuticals, Inc. Compounds for inhibiting cell proliferation in EGFR-driven cancers
US9073947B2 (en) 2011-06-10 2015-07-07 Merck Patent Gmbh Compositions and methods for the production of pyrimidine and pyridine compounds with BTK inhibitory activity
JP2018115165A (en) * 2011-06-10 2018-07-26 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Merck Patent Gesellschaft mit beschraenkter Haftung Compositions and methods for production of pyrimidine and pyridine compounds with btk inhibitory activity
WO2012170976A3 (en) * 2011-06-10 2013-04-11 Merck Patent Gmbh Compositions and methods for the production of pyrimidine and pyridine compounds with btk inhibitory activity
US9580449B2 (en) 2011-06-10 2017-02-28 Merck Patent Gmbh Compositions and methods for the production of pyrimidine and pyridine compounds with BTK inhibitory activity
EA031153B1 (en) * 2011-06-10 2018-11-30 Мерк Патент Гмбх Use of pyrimidine compounds with btk inhibitory activity
JP2014517016A (en) * 2011-06-10 2014-07-17 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 Pyrimidine having BTK inhibitory activity and composition of pyrimidine compound and method for producing the same
US10016448B2 (en) 2011-06-10 2018-07-10 Merck Patent Gmbh Compositions and methods for the production of pyrimidine and pyridine compounds with BTK inhibitory activity
JP2016185953A (en) * 2011-06-10 2016-10-27 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツングMerck Patent Gesellschaft mit beschraenkter Haftung Compositions and methods for the production of pyrimidine and pyridine compounds with btk inhibitory activity
US10413562B2 (en) 2011-06-10 2019-09-17 Merck Patent Gmbh Compositions and methods for the production of pyrimidine and pyridine compounds with BTK inhibitory activity
JP2020073593A (en) * 2011-06-10 2020-05-14 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Merck Patent Gesellschaft mit beschraenkter Haftung Compositions and methods for production of pyrimidine and pyridine compounds with btk inhibitory activity
EA027584B1 (en) * 2011-06-10 2017-08-31 Мерк Патент Гмбх Compositions and methods for the production of pyrimidine and pyridine compounds with btk inhibitory activity
US10875864B2 (en) 2011-07-21 2020-12-29 Sumitomo Dainippon Pharma Oncology, Inc. Substituted imidazo[1,2-B]pyridazines as protein kinase inhibitors
EP3686194A1 (en) * 2011-07-27 2020-07-29 Astrazeneca AB 2-(2,4,5-substituted-anilino)pyrimidine compounds
US10017493B2 (en) 2011-07-27 2018-07-10 Astrazeneca Ab 2-(2,4,5-substituted-anilino)pyrimidine compounds
TWI583386B (en) * 2011-07-27 2017-05-21 阿斯特捷利康公司 2-(2,4,5-substituted-anilino)pyrimidine compounds
TWI465445B (en) * 2011-07-27 2014-12-21 Astrazeneca Ab 2-(2,4,5-substituted-anilino)pyrimidine compounds
AU2015261672B2 (en) * 2011-07-27 2017-04-27 Astrazeneca Ab 2 - (2, 4, 5 - substituted -anilino) pyrimidine derivatives as egfr modulators useful for treating cancer
EA029488B1 (en) * 2011-07-27 2018-04-30 Астразенека Аб 2-(2,4,5-substituted-anilino)pyrimidine derivatives as egfr modulators useful for treating cancer
US10858336B2 (en) 2011-07-27 2020-12-08 Astazeneca Ab 2-(2,4,5-substituted-anilino)pyrimidine compounds
AU2013204962C1 (en) * 2011-07-27 2016-03-10 Astrazeneca Ab Polymorphic form of a mesylate salt of n-(2-{2-dimethylaminoethyl-methylamino}-4-methoxy-5-{[4-(1-methylindol-3-yl)pyrimidin-2-yl]amino}phenyl)prop-2-enamide
EP4119551A1 (en) * 2011-07-27 2023-01-18 Astrazeneca AB 2-(2,4,5-substituted-anilino)pyrimidine compounds
EA024421B1 (en) * 2011-07-27 2016-09-30 Астразенека Аб N-(2-{2-dimethylaminoethyl-methylamino}-4-methoxy-5-{[4-(1-methylindol-3-yl)pyrimidin-2-yl]amino}phenyl)prop-2-enamide and pharmaceutically acceptable salts thereof as egfr modulators useful for treating cancer
WO2013014448A1 (en) * 2011-07-27 2013-01-31 Astrazeneca Ab 2 - (2, 4, 5 - substituted -anilino) pyrimidine derivatives as egfr modulators useful for treating cancer
EP3009431A1 (en) * 2011-07-27 2016-04-20 Astrazeneca AB 2-(2,4,5-substituted-anilino)pyrimidine derivatives as egfr modulators useful for treating cancer
EP3686193A1 (en) * 2011-07-27 2020-07-29 Astrazeneca AB 2-(2,4,5-substituted-anilino)pyrimidine compounds
AU2012288626C1 (en) * 2011-07-27 2015-09-17 Astrazeneca Ab 2 - (2, 4, 5 - substituted -anilino) pyrimidine derivatives as EGFR modulators useful for treating cancer
TWI555743B (en) * 2011-07-27 2016-11-01 阿斯特捷利康公司 2-(2,4,5-substituted-anilino)pyrimidine compounds
AU2012288626B2 (en) * 2011-07-27 2015-05-07 Astrazeneca Ab 2 - (2, 4, 5 - substituted -anilino) pyrimidine derivatives as EGFR modulators useful for treating cancer
EP3333161A1 (en) * 2011-07-27 2018-06-13 Astrazeneca AB 2-(2,4,5-substituted-anilino)pyrimidine derivatives as egfr modulators useful for treating cancer
EA033733B1 (en) * 2011-07-27 2019-11-20 Astrazeneca Ab 2-(2,4,5-substituted-anilino)pyrimidine derivatives as egfr modulators useful for treating cancer and intermediates for manufacture thereof
AU2013204962B2 (en) * 2011-07-27 2015-09-03 Astrazeneca Ab Polymorphic form of a mesylate salt of n-(2-{2-dimethylaminoethyl-methylamino}-4-methoxy-5-{[4-(1-methylindol-3-yl)pyrimidin-2-yl]amino}phenyl)prop-2-enamide
US8946235B2 (en) 2011-07-27 2015-02-03 Astrazeneca Ab 2-(2,4,5-substituted-anilino) pyrimidine compounds
US11524951B2 (en) 2011-07-27 2022-12-13 Astrazeneca Ab 2-(2,4,5-substituted-anilino)pyrimidine compounds
EP3009431B1 (en) 2011-07-27 2017-10-25 Astrazeneca AB 2-(2,4,5-substituted-anilino)pyrimidine derivatives as egfr modulators useful for treating cancer
JP2014139226A (en) * 2011-07-27 2014-07-31 Astrazeneca Ab 2-(2,4,5-substituted-anilino)pyrimidine compound
KR101422619B1 (en) 2011-07-27 2014-07-24 아스트라제네카 아베 2-(2,4,5-substituted-anilino)pyrimidine derivatives as egfr modulators useful f0r treating cancer
EP2736895B1 (en) 2011-07-27 2016-01-06 Astrazeneca AB 2-(2,4,5-substituted-anilino) pyrimidine derivatives as egfr modulators useful for treating cancer
EP4086246A1 (en) * 2011-07-27 2022-11-09 AstraZeneca AB 2-(2,4,5-substituted-anilino)pyrimidine compounds as egfr modulators
US9732058B2 (en) 2011-07-27 2017-08-15 Astrazeneca Ab 2-(2,4,5-substituted-anilino)pyrimidine compounds
US9187462B2 (en) 2011-08-04 2015-11-17 Array Biopharma Inc. Substituted quinazolines as serine/threonine kinase inhibitors
US9364476B2 (en) 2011-10-28 2016-06-14 Celgene Avilomics Research, Inc. Methods of treating a Bruton's Tyrosine Kinase disease or disorder
US9763949B2 (en) 2012-01-13 2017-09-19 Acea Biosciences Inc. EGFR modulators and uses thereof
US9586965B2 (en) 2012-01-13 2017-03-07 Acea Biosciences Inc. Pyrrolo[2,3-d]pyrimidine compounds as inhibitors of protein kinases
US20150133457A1 (en) * 2012-01-13 2015-05-14 Acea Biosciences Inc. Heterocyclic compounds and uses thereof
US11612602B2 (en) 2012-01-13 2023-03-28 ACEA Therapeutics, Inc. Heterocyclic compounds and uses as anticancer agents
US10799504B2 (en) 2012-01-13 2020-10-13 ACEA Therapeutics, Inc. Heterocyclic compounds and uses as anticancer agents
US9034885B2 (en) 2012-01-13 2015-05-19 Acea Biosciences Inc. EGFR modulators and uses thereof
US9920074B2 (en) 2012-01-13 2018-03-20 Acea Biosciences Inc. Heterocyclic compounds and uses thereof
US9464089B2 (en) 2012-01-13 2016-10-11 Acea Biosciences Inc. Heterocyclic compounds and uses thereof
US10596174B2 (en) 2012-01-13 2020-03-24 ACEA Therapeutics, Inc. Pyrrolopyrimidine compounds as inhibitors of protein kinases
US9085540B2 (en) 2012-01-17 2015-07-21 Astellas Pharma Inc. Pyrazinecarboxamide compound
KR20140114404A (en) 2012-01-17 2014-09-26 아스테라스 세이야쿠 가부시키가이샤 Pyrazine carboxamide compound
WO2013108754A1 (en) 2012-01-17 2013-07-25 アステラス製薬株式会社 Pyrazine carboxamide compound
EA036521B1 (en) * 2012-01-27 2020-11-19 Астразенека Аб 2-(2,4,5-substituted-anilino)pyrimidine derivatives as egfr modulators useful for treating cancer
US9708290B2 (en) 2012-03-01 2017-07-18 Array Biopharma Inc. Serine/threonine kinase inhibitors
US9259470B2 (en) 2012-03-01 2016-02-16 Array Biopharma Inc. Serine/threonine kinase inhibitors
US8697715B2 (en) 2012-03-01 2014-04-15 Array Biopharma, Inc. Serine/threonine kinase inhibitors
US10519126B2 (en) 2012-03-01 2019-12-31 Array Biopharma Inc. Serine/threonine kinase inhibitors
US10570099B2 (en) 2012-03-15 2020-02-25 Celgene Car Llc Salts of an epidermal growth factor receptor kinase inhibitor
CN108658873B (en) * 2012-03-15 2021-09-14 西建卡尔有限责任公司 Solid forms of an epidermal growth factor receptor kinase inhibitor
RU2711077C2 (en) * 2012-03-15 2020-01-15 Селджен Кар Ллс Salts of epidermal growth factor receptor kinase
US9056839B2 (en) 2012-03-15 2015-06-16 Celgene Avilomics Research, Inc. Solid forms of an epidermal growth factor receptor kinase inhibitor
US9108927B2 (en) 2012-03-15 2015-08-18 Celgene Avilomics Research, Inc. Salts of an epidermal growth factor receptor kinase inhibitor
US10004741B2 (en) 2012-03-15 2018-06-26 Celgene Car Llc Solid forms of an epidermal growth factor receptor kinase inhibitor
US9540335B2 (en) 2012-03-15 2017-01-10 Celgene Avilomics Research, Inc. Salts of an epidermal growth factor receptor kinase inhibitor
US10005738B2 (en) 2012-03-15 2018-06-26 Celgene Car Llc Salts of an epidermal growth factor receptor kinase inhibitor
CN108658873A (en) * 2012-03-15 2018-10-16 西建卡尔有限责任公司 The solid form of epidermal growth factor receptor kinase inhibitor
US9539255B2 (en) 2012-03-15 2017-01-10 Celgene Avilomics Research, Inc. Solid forms of an epidermal growth factor receptor kinase inhibitor
US11292772B2 (en) 2012-03-15 2022-04-05 Celgene Car Llc Salts of an epidermal growth factor receptor kinase inhibitor
RU2673077C2 (en) * 2012-03-15 2018-11-22 Селджен Авиломикс Рисерч, Инк. Solid forms of epidermal growth factor receptor kinase inhibitor
US10946016B2 (en) 2012-03-15 2021-03-16 Celgene Car Llc Solid forms of an epidermal growth factor receptor kinase inhibitor
RU2711077C9 (en) * 2012-03-15 2020-08-11 Селджен Кар Ллс Salts of epidermal growth factor receptor kinase
WO2013157540A1 (en) * 2012-04-17 2013-10-24 富士フイルム株式会社 Nitrogen-containing heterocyclic compound or salt thereof
AU2013250378B2 (en) * 2012-04-17 2016-01-14 Fujifilm Corporation Nitrogen-containing heterocyclic compound or salt thereof
JP5736507B2 (en) * 2012-04-17 2015-06-17 富士フイルム株式会社 Nitrogen-containing heterocyclic compound or salt thereof
US9834571B2 (en) 2012-05-05 2017-12-05 Ariad Pharmaceuticals, Inc. Compounds for inhibiting cell proliferation in EGFR-driven cancers
US10266540B2 (en) 2012-06-04 2019-04-23 Pharmacyclics Llc Crystalline forms of a Bruton's tyrosine kinase inhibitor
US9828383B1 (en) 2012-06-04 2017-11-28 Pharmacyclic s LLC Crystalline forms of a bruton's tyrosine kinase inhibitor
US9296753B2 (en) 2012-06-04 2016-03-29 Pharmacyclics Llc Crystalline forms of a Bruton's tyrosine kinase inhibitor
US10961251B1 (en) 2012-06-04 2021-03-30 Pharmacyclics Llc Crystalline forms of a Bruton's tyrosine kinase inhibitor
US10294231B2 (en) 2012-06-04 2019-05-21 Pharmacyclics Llc Crystalline forms of a Bruton's tyrosine kinase inhibitor
US10065968B2 (en) 2012-06-04 2018-09-04 Pharmacyclics Llc Crystalline forms of a bruton's tyrosine kinase inhibitor
US10752634B2 (en) 2012-06-04 2020-08-25 Pharmacyclics Llc Crystalline forms of a brutons tyrosine kinase inhibitor
US9713617B2 (en) 2012-06-04 2017-07-25 Pharmacyclics Llc Crystalline forms of a Bruton's tyrosine kinase inhibitor
US10106548B2 (en) 2012-06-04 2018-10-23 Pharmacyclics Llc Crystalline forms of a Bruton's tyrosine kinase inhibitor
US10294232B2 (en) 2012-06-04 2019-05-21 Pharmacyclics Llc Crystalline forms of a Bruton's tyrosine kinase inhibitor
US10125140B1 (en) 2012-06-04 2018-11-13 Pharmacyclics Llc Crystalline forms of a bruton's tyrosine kinase inhibitor
US9725455B1 (en) 2012-06-04 2017-08-08 Pharmacyclics Llc Crystalline forms of a bruton's tyrosine kinase inhibitor
US9540382B2 (en) 2012-06-04 2017-01-10 Pharmacyclics Llc Crystalline forms of a Bruton's tyrosine kinase inhibitor
US10196436B2 (en) 2012-07-11 2019-02-05 Blueprint Medicines Corporation Inhibitors of the fibroblast growth factor receptor
CN104540809A (en) * 2012-07-11 2015-04-22 蓝印药品公司 Inhibitors of the fibroblast growth factor receptor
US9126951B2 (en) 2012-07-11 2015-09-08 Blueprint Medicines Corporation Inhibitors of the fibroblast growth factor receptor
US8802697B2 (en) 2012-07-11 2014-08-12 Blueprint Medicines Corporation Inhibitors of the fibroblast growth factor receptor
US9340514B2 (en) 2012-07-11 2016-05-17 Blueprint Medicines Corporation Inhibitors of the fibroblast growth factor receptor
WO2014011900A3 (en) * 2012-07-11 2014-02-27 Blueprint Medicines Inhibitors of the fibroblast growth factor receptor
RU2679130C2 (en) * 2012-07-11 2019-02-06 Блюпринт Медсинс Корпорейшн Inhibitors of fibroblast growth factor receptor
US10954567B2 (en) 2012-07-24 2021-03-23 Pharmacyclics Llc Mutations associated with resistance to inhibitors of Bruton's Tyrosine Kinase (BTK)
EP3550031A1 (en) 2012-07-24 2019-10-09 Pharmacyclics, LLC Mutations associated with resistance to inhibitors of bruton's tyrosine kinase (btk)
US11007197B2 (en) 2012-08-06 2021-05-18 ACEA Therapeutics, Inc. EGFR modulators and uses thereof
US8685988B2 (en) 2012-08-06 2014-04-01 Acea Biosciences, Inc. EGFR modulators and uses thereof
US10449196B2 (en) 2012-08-06 2019-10-22 ACEA Therapeutics, Inc. EGFR modulators and uses thereof
US9388171B2 (en) 2012-08-27 2016-07-12 Genetech, Inc. Serine/threonine kinase inhibitors
US9540385B2 (en) 2012-11-15 2017-01-10 Pharmacyclics Llc Pyrrolopyrimidine compounds as kinase inhibitors
EP2922546A4 (en) * 2012-11-20 2016-06-08 Celgene Avilomics Res Inc Methods of treating a disease or disorder associated with bruton's tyrosine kinase
US9549927B2 (en) 2012-12-21 2017-01-24 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
US9126950B2 (en) 2012-12-21 2015-09-08 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
EP2935226A4 (en) * 2012-12-21 2016-11-02 Celgene Avilomics Res Inc Heteroaryl compounds and uses thereof
US20140228322A1 (en) * 2013-02-08 2014-08-14 Celgene Avilomics Research, Inc. Erk inhibitors and uses thereof
EP2953457A4 (en) * 2013-02-08 2016-07-06 Celgene Avilomics Res Inc Erk inhibitors and uses thereof
US9145387B2 (en) * 2013-02-08 2015-09-29 Celgene Avilomics Research, Inc. ERK inhibitors and uses thereof
US9796700B2 (en) 2013-02-08 2017-10-24 Celgene Car Llc ERK inhibitors and uses thereof
US9561228B2 (en) 2013-02-08 2017-02-07 Celgene Avilomics Research, Inc. ERK inhibitors and uses thereof
US9980964B2 (en) 2013-02-08 2018-05-29 Celgene Car Llc ERK inhibitors and uses thereof
US9504686B2 (en) 2013-02-08 2016-11-29 Celgene Avilomics Research, Inc. ERK inhibitors and uses thereof
US9828345B2 (en) 2013-02-28 2017-11-28 Bristol-Myers Squibb Company Phenylpyrazole derivatives as potent ROCK1 and ROCK2 inhibitors
US9458110B2 (en) 2013-02-28 2016-10-04 Bristol-Myers Squibb Company Phenylpyrazole derivatives as potent ROCK1 and ROCK2 inhibitors
US9126944B2 (en) 2013-02-28 2015-09-08 Bristol-Myers Squibb Company Phenylpyrazole derivatives as potent ROCK1 and ROCK2 inhibitors
WO2014134308A1 (en) * 2013-03-01 2014-09-04 Amgen Inc. Substituted 7-oxo-pyrido [2, 3-d] pyrimidines and their use for the treatment of egfr / erbb2 related disorders
US9346801B2 (en) 2013-03-01 2016-05-24 Amgen Inc. Substituted 7-oxo-pyrido[2,3-d]pyrimidines and methods of use
US10752594B2 (en) 2013-03-14 2020-08-25 Sumitomo Dainippon Pharma Oncology, Inc. JAK1 and ALK2 inhibitors and methods for their use
US9850233B2 (en) 2013-03-14 2017-12-26 Respivert Limited Kinase inhibitors
EP2968327A4 (en) * 2013-03-14 2016-09-28 Celgene Avilomics Res Inc Heteroaryl compounds and uses thereof
US10202356B2 (en) 2013-03-14 2019-02-12 Tolero Pharmaceuticals, Inc. JAK2 and ALK2 inhibitors and methods for their use
US10301288B2 (en) 2013-03-14 2019-05-28 Topivert Pharma Limited Kinase inhibitors
US10618902B2 (en) 2013-03-15 2020-04-14 Celgene Car Llc Substituted pyrido[2,3-d]pyrimidines as inhibitors of protein kinases
US10189794B2 (en) 2013-03-15 2019-01-29 Celgene Car Llc Heteroaryl compounds and uses thereof
US9695132B2 (en) 2013-03-15 2017-07-04 Celgene Car Llc Heteroaryl compounds and uses thereof
US9663524B2 (en) 2013-03-15 2017-05-30 Celgene Car Llc Substituted pyrido[2,3-d]pyrimidines as protein kinase inhibitors
US11053195B2 (en) 2013-03-15 2021-07-06 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
US9321786B2 (en) 2013-03-15 2016-04-26 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
US10065966B2 (en) 2013-03-15 2018-09-04 Celgene Car Llc Substituted pyrido[2,3-d]pyrimidines as inhibitors of protein kinases
US10774052B2 (en) 2013-03-15 2020-09-15 Celgene Car Llc Heteroaryl compounds and uses thereof
US9611283B1 (en) 2013-04-10 2017-04-04 Ariad Pharmaceuticals, Inc. Methods for inhibiting cell proliferation in ALK-driven cancers
US9556188B2 (en) 2013-04-25 2017-01-31 Beigene, Ltd. Substituted imidazo[1,2-b]pyrazoles as bruton'S tyrosine kinase modulators
US11142528B2 (en) 2013-04-25 2021-10-12 Beigene Switzerland Gmbh Substituted pyrazolo[1,5-a]pyrimidines as Bruton's tyrosine kinase modulators
US9447106B2 (en) 2013-04-25 2016-09-20 Beigene, Ltd. Substituted pyrazolo[1,5-a]pyrimidines as bruton's tyrosine kinase modulators
US10570139B2 (en) 2013-04-25 2020-02-25 Beigene Switzerland Gmbh Substituted pyrazolo[1,5-a]pyrimidines as Bruton's tyrosine kinase modulators
US10005782B2 (en) 2013-04-25 2018-06-26 Beigene, Ltd. Substituted pyrazolo[1,5-a]pyrimidines as bruton's tyrosine kinase modulators
WO2014188173A1 (en) 2013-05-20 2014-11-27 Redx Pharma Limited Pyrazolopyrimidine derivatives useful as inhibitors of bruton's tyrosine kinase
EP3019489A4 (en) * 2013-07-11 2016-12-28 Betta Pharmaceuticals Co Ltd Protein tyrosine kinase modulators and methods of use
US10059688B2 (en) 2013-07-11 2018-08-28 Betta Pharmaceuticals Co., Ltd. Protein tyrosine kinase modulators and methods of use
US9783524B2 (en) 2013-07-11 2017-10-10 Betta Pharmaceuticals Co., Ltd. Protein tyrosine kinase modulators and methods of use
US10562918B2 (en) 2013-07-11 2020-02-18 ACEA Therapeutics, Inc. Heterocyclic compounds and uses thereof
AU2014287016B2 (en) * 2013-07-11 2018-11-01 Acea Biosciences Inc. Pyrimidine derivatives as kinase inhibitors
RU2656591C2 (en) * 2013-07-11 2018-06-06 Бетта Фармасьютикалз Ко., Лтд Protein tyrosine kinase modulators and methods of use
WO2015003658A1 (en) 2013-07-11 2015-01-15 Betta Pharmaceuticals Co., Ltd Protein tyrosine kinase modulators and methods of use
RU2677653C2 (en) * 2013-07-11 2019-01-18 Ацея Байосайенсиз Инк. Pyrimidine derivatives as kinase inhibitors
US9421208B2 (en) 2013-08-02 2016-08-23 Pharmacyclics Llc Methods for the treatment of solid tumors
WO2015019365A1 (en) 2013-08-07 2015-02-12 Cadila Healthcare Limited N-cyanomethylamides as inhibitors of janus kinase
US9556148B2 (en) 2013-08-07 2017-01-31 Cadila Healthcare Limited N-cyanomethylamides as inhibitors of janus kinase
US9580432B2 (en) 2013-08-12 2017-02-28 Taiho Pharmaceutical Co., Ltd. Fused pyrimidine compound or salt thereof
US9724349B2 (en) 2013-08-12 2017-08-08 Pharmacyclics Llc Methods for the treatment of HER2 amplified cancer
WO2015022926A1 (en) 2013-08-12 2015-02-19 大鵬薬品工業株式会社 Novel fused pyrimidine compound or salt thereof
US10016434B2 (en) 2013-08-12 2018-07-10 Pharmacyclics Llc Methods for the treatment of HER2 amplified cancer
US9415050B2 (en) 2013-08-12 2016-08-16 Pharmacyclics Llc Methods for the treatment of HER2 amplified cancer
US9492471B2 (en) 2013-08-27 2016-11-15 Celgene Avilomics Research, Inc. Methods of treating a disease or disorder associated with Bruton'S Tyrosine Kinase
US11186637B2 (en) 2013-09-13 2021-11-30 Beigene Switzerland Gmbh Anti-PD1 antibodies and their use as therapeutics and diagnostics
US11673951B2 (en) 2013-09-13 2023-06-13 Beigene Switzerland Gmbh Anti-PD1 antibodies and their use as therapeutics and diagnostics
US10301297B2 (en) 2013-09-30 2019-05-28 Guangzhou Innocare Pharma Tech Co., Ltd. Substituted nicotinimide inhibitors of BTK and their preparation and use in the treatment of cancer, inflammation and autoimmune disease
WO2015048662A3 (en) * 2013-09-30 2015-05-07 X-Rx Discovery, Inc. Substituted nicotinimide inhibitors of btk and their preparation and use in the treatment of cancer, inflammation and autoimmune disease
AU2014324595B2 (en) * 2013-09-30 2018-12-20 Beijing Innocare Pharma Tech Co., Ltd Substituted nicotinimide inhibitors of BTK and their preparation and use in the treatment of cancer, inflammation and autoimmune disease
US11345695B2 (en) 2013-09-30 2022-05-31 Guangzhou Innocare Pharma Tech Co., Ltd. Substituted nicotinimide inhibitors of BTK and their preparation and use in the treatment of cancer, inflammation and autoimmune disease
US11840513B2 (en) 2013-09-30 2023-12-12 Guangzhou Innocare Pharma Tech Co., Ltd Substituted nicotinimide inhibitors of BTK for treating cancer
RU2677884C2 (en) * 2013-09-30 2019-01-22 Гуанчжоу Иннокэа Фарма Тек Ко., Лтд. Substituted nicotinimide inhibitors of btk, their preparation and use in treatment of cancer, inflammation and autoimmune diseases
US9951056B2 (en) 2013-09-30 2018-04-24 Beijing Innocare Pharma Tech Co., Ltd. Substituted nicotinamide inhibitors of BTK and their preparation and use in the treatment of cancer, inflammation and autoimmune disease
US10537571B2 (en) 2013-10-18 2020-01-21 Eisai R&D Management Co., Ltd. Pyrimidine FGFR4 inhibitors
US10912774B2 (en) 2013-10-18 2021-02-09 Eisai R&D Management Co., Ltd. Pyrimidine FGFR4 inhibitors
US9730931B2 (en) 2013-10-18 2017-08-15 Eisai R&D Management Co., Ltd. Pyrimidine FGFR4 inhibitors
WO2015061247A3 (en) * 2013-10-21 2015-07-23 Merck Patent Gmbh Heteroaryl compounds as btk inhibitors and uses thereof
US10329270B2 (en) 2013-10-21 2019-06-25 Merck Patent Gmbh Heteroaryl compounds as BTK inhibitors and uses thereof
US9434700B2 (en) 2013-10-25 2016-09-06 Neil Bifulco, JR. Inhibitors of the fibroblast growth factor receptor
US10875837B2 (en) 2013-10-25 2020-12-29 Blueprint Medicines Corporation Inhibitors of the fibroblast growth factor receptor
EP4115886A1 (en) 2013-10-25 2023-01-11 Pharmacyclics LLC Methods of treating and preventing graft versus host disease
US10221154B2 (en) 2013-10-25 2019-03-05 Blueprint Medicines Corporation Inhibitors of the fibroblast growth factor receptor
US10450269B1 (en) 2013-11-18 2019-10-22 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
WO2015084998A1 (en) * 2013-12-05 2015-06-11 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase
US9382246B2 (en) 2013-12-05 2016-07-05 Pharmacyclics Llc Inhibitors of Bruton's tyrosine kinase
US9656988B2 (en) 2013-12-05 2017-05-23 Pharmacyclics Llc Inhibitors of Bruton's tyrosine kinase
US9415049B2 (en) 2013-12-20 2016-08-16 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
US10000490B2 (en) 2014-01-15 2018-06-19 Blueprint Medicines Corporation Inhibitors of the fibroblast growth factor receptor
US9695165B2 (en) 2014-01-15 2017-07-04 Blueprint Medicines Corporation Inhibitors of the fibroblast growth factor receptor
US10301278B2 (en) 2014-01-17 2019-05-28 Novartis Ag 1-(triazin-3-yl/pyridazin-3-yl)-piper(-azine)idine derivatives and compositions therefor for inhibiting the activity of SHP2
US11401259B2 (en) 2014-01-17 2022-08-02 Novartis Ag 1-Pyridazin-/triazin-3-yl-piper(-azine)/idine/pyrolidine derivatives and compositions thereof for inhibiting the activity of SHP2
US10336774B2 (en) 2014-01-17 2019-07-02 Novartis Ag N-azaspirocycloalkane substituted N-heteroaryl compounds and compositions for inhibiting the activity of SHP2
US10093646B2 (en) 2014-01-17 2018-10-09 Novartis Ag 1-pyridazin-/triazin-3-yl-piper(-azine)/idine/pyrolidine derivatives and compositions thereof for inhibiting the activity of SHP2
US10077276B2 (en) 2014-01-17 2018-09-18 Novartis Ag N-azaspirocycloalkane substituted N-heteroaryl compounds and compositions for inhibiting the activity of SHP2
US10968235B2 (en) 2014-01-17 2021-04-06 Novartis Ag N-azaspirocycloalkane substituted N-heteroaryl compounds and compositions for inhibiting the activity of SHP2
US9815813B2 (en) 2014-01-17 2017-11-14 Novartis Ag 1-(triazin-3-yl/pyridazin-3-yl)-piper(-azine)idine derivatives and compositions therefor for inhibiting the activity of SHP2
US10774065B2 (en) 2014-01-17 2020-09-15 Novartis Ag 1-pyridazin-/triazin-3-yl-piper(-azine)/idine/pyrolidine derivatives and compositions thereof for inhibiting the activity of SHP2
US9365524B2 (en) 2014-01-30 2016-06-14 Signal Pharmaceuticals, Llc Solid forms of 2-(tert-butylamino)-4-((1R,3R,4R)-3-hydroxy-4-methylcyclohexylamino)-pyrimidine-5-carboxamide, compositions thereof and methods of their use
US10226461B2 (en) 2014-01-30 2019-03-12 Signal Pharmaceuticals, Llc Solid forms of 2-(tert-butylamino)-4-((1R,3R,4R)-3-hydroxy-4-methylcyclohexylamino)-pyrimidine-5-carboxamide, compositions thereof and methods of their use
US9814713B2 (en) 2014-01-30 2017-11-14 Signal Pharmaceuticals, Llc Solid forms of 2-(tert-butylamino)-4-((1R,3R,4R)-3-hydroxy-4-methylcyclohexylamino)-pyrimidine-5-carboxamide, compositions thereof and methods of their use
US11241430B2 (en) * 2014-01-30 2022-02-08 Signal Pharmaceuticals, Llc Solid forms of 2-(tert-butylamino)-4-((1R,3R,4R)-3-hydroxy-4-methylcyclohexylamino)-pyrimidine-5-carboxamide, compositions thereof and methods of their use
US10517873B2 (en) 2014-01-30 2019-12-31 Signal Pharmaceuticals, Llc Solid forms of 2-(tert-butylamino)-4-((1R,3R,4R)-3-hydroxy-4-methylcyclohexylamino)-pyrimidine-5-carboxamide, compositions thereof and methods of their use
US9885086B2 (en) 2014-03-20 2018-02-06 Pharmacyclics Llc Phospholipase C gamma 2 and resistance associated mutations
WO2015151006A1 (en) 2014-03-29 2015-10-08 Lupin Limited Substituted purine compounds as btk inhibitors
EP3144292A4 (en) * 2014-04-14 2018-01-17 Shanghai Haiyan Pharmaceutical Technology Co., Ltd 2,3,4,6-tetra-substituted benzene-1,5-diamine derivatives, preparation method therefor and medicinal use thereof
CN104130265A (en) * 2014-04-29 2014-11-05 苏州景泓生物技术有限公司 Spiral ring or bridged ring containing pyrimidine compound
CN104130265B (en) * 2014-04-29 2017-01-25 苏州景泓生物技术有限公司 Spiral ring or bridged ring containing pyrimidine compound
WO2015170266A1 (en) 2014-05-07 2015-11-12 Lupin Limited Substituted pyrimidine compounds as btk inhibitors
CN106687457A (en) * 2014-05-13 2017-05-17 阿里亚德医药股份有限公司 Heteroaryl compounds for kinase inhibition
CN106687457B (en) * 2014-05-13 2020-01-10 阿里亚德医药股份有限公司 Heteroaryl compounds for kinase inhibition
US9975897B2 (en) 2014-06-11 2018-05-22 Loxo Oncology, Inc. Pyrazolopyrimidine derivatives useful as inhibitors of Bruton's tyrosine kinase
US10538524B2 (en) 2014-06-11 2020-01-21 Loxo Oncology Inc. Pyrazolopyrimidine derivatives useful as inhibitors of Bruton's tyrosine kinase
JP2019194217A (en) * 2014-06-19 2019-11-07 アリアド ファーマシューティカルズ, インコーポレイテッド Heteroaryl compounds for kinase inhibition
EP3409669A1 (en) * 2014-06-19 2018-12-05 ARIAD Pharmaceuticals, Inc. Heteroaryl compounds for kinase inhibition
EP3157916A4 (en) * 2014-06-19 2018-03-07 ARIAD Pharmaceuticals, Inc. Heteroaryl compounds for kinase inhibition
AU2015277786B2 (en) * 2014-06-19 2019-04-18 Takeda Pharmaceutical Company Limited Heteroaryl compounds for kinase inhibition
US10227342B2 (en) 2014-06-19 2019-03-12 Ariad Pharmaceuticals, Inc. Heteroaryl compounds for kinase inhibition
US11512132B2 (en) 2014-07-03 2022-11-29 Beigene, Ltd. Anti-PD-L1 antibodies and their use as therapeutics and diagnostics
EA032693B1 (en) * 2014-07-24 2019-07-31 Бета Фарма, Инк. 2h-indazole derivatives as cyclin-dependent kinase (cdk) inhibitors and therapeutic uses thereof
US10239864B2 (en) 2014-07-24 2019-03-26 Beta Pharma, Inc. 2-H-indazole derivatives as cyclin-dependent kinase (CDK) inhibitors and therapeutic uses thereof
WO2016014904A1 (en) * 2014-07-24 2016-01-28 Beta Pharma, Inc. 2-h-indazole derivatives as cyclin-dependent kinase (cdk) inhibitors and therapeutic uses thereof
US9878994B2 (en) 2014-07-24 2018-01-30 Beta Pharma Inc. 2-H-indazole derivatives as cyclin-dependent kinase (CDK) inhibitors and therapeutic uses thereof
AU2015292425B2 (en) * 2014-07-24 2018-12-20 Beta Pharma, Inc. 2-H-indazole derivatives as cyclin-dependent kinase (CDK) inhibitors and therapeutic uses thereof
US9533991B2 (en) 2014-08-01 2017-01-03 Pharmacyclics Llc Inhibitors of Bruton's tyrosine kinase
US9545407B2 (en) 2014-08-07 2017-01-17 Pharmacyclics Llc Formulations of a bruton's tyrosine kinase inhibitor
US20180028537A1 (en) 2014-08-07 2018-02-01 Pharmacyclics Llc Novel Formulations of a Bruton's Tyrosine Kinase Inhibitor
US10005760B2 (en) 2014-08-13 2018-06-26 Celgene Car Llc Forms and compositions of an ERK inhibitor
US10202364B2 (en) 2014-08-13 2019-02-12 Celgene Car Llc Forms and compositions of an ERK inhibitor
US10676434B2 (en) 2014-10-24 2020-06-09 Bristol-Myers Squibb Company Carbazole derivatives
US11053197B2 (en) 2014-10-24 2021-07-06 Bristol-Myers Squibb Company Carbazole derivatives
RU2704129C2 (en) * 2014-11-24 2019-10-24 Шангхаи Институте Оф Материа Медика, Чайнесе Академи Оф Сайнсес 2-aminopyrimidine compound and pharmaceutical composition and use of said compound
US10059694B2 (en) 2014-11-24 2018-08-28 Shanghai Institute Of Materia Medica, Chinese Academy Of Sciences 2-aminopyrimidine compound and pharmaceutical composition and use thereof
KR20170131341A (en) * 2014-11-24 2017-11-29 상하이 인스티튜트 오브 마테리아 메디카 차이니즈 아카데미 오브 싸이언시즈 2-aminopyrimidine compound and pharmaceutical composition and use thereof
KR101941929B1 (en) 2014-11-24 2019-01-24 상하이 인스티튜트 오브 마테리아 메디카 차이니즈 아카데미 오브 싸이언시즈 2-aminopyrimidine compound and pharmaceutical composition and use thereof
EP3225619A4 (en) * 2014-11-24 2018-05-23 Shanghai Institute Of Materia Medica Chinese Academy of Sciences 2-aminopyrimidine compound and pharmaceutical composition and use thereof
US10012650B2 (en) * 2014-12-11 2018-07-03 Merck Patent Gmbh Assays for BTK inhibitors
US9513297B2 (en) 2014-12-16 2016-12-06 Signal Pharmaceuticals, Llc Methods for measurement of inhibition of c-Jun N-terminal kinase in skin
US9796685B2 (en) 2014-12-16 2017-10-24 Signal Pharmaceuticals, Llc Formulations of 2-(tert-butylamino)-4-((1R,3R,4R)-3-hydroxy-4-Methylcyclohexylamino)-pyrimidine-5-carboxamide
US10590089B2 (en) 2014-12-16 2020-03-17 Signal Pharmaceuticals, Llc Formulations of 2-(tert-butylamino)-4-((1R,3R,4R)-3-hydroxy-4-methylcyclohexylamino)-pyrimidine-5-carboxamide
US10197579B2 (en) 2014-12-16 2019-02-05 Signal Pharmaceuticals, Llc Methods for measurement of inhibition of c-Jun N-terminal kinase in skin
US10131639B2 (en) 2014-12-16 2018-11-20 Signal Pharmaceuticals, Llc Formulations of 2-(tert-butylamino)-4-((1R,3R,4R)-3-hydroxy-4methylcyclohexylamino)-pyrimidine-5-carboxamide
US10975039B2 (en) 2015-01-29 2021-04-13 Signal Pharmaceuticals, Llc Isotopologues of 2-(tert-butylamino)-4-((1R,3R,4R)-3-hydroxy-4-methylcyclohexylamino)-pyrimidine-5-carboxamide
US10689351B2 (en) 2015-01-29 2020-06-23 Signal Pharmaceuticals, Llc Isotopologues of 2-(tert butylamino)-4-((1R,3R,4R)-3-hydroxy-4-methylcyclohexylamino)-pyrimidine-5-carboxamide
US11492332B2 (en) 2015-01-29 2022-11-08 Signal Pharmaceuticals, Llc Isotopologues of 2-(tert-butylamino)-4-((1R,3R,4R)-3-hydroxy-4-methylcyclohexylamino)-pyrimidine-5-carboxamide
US10280174B2 (en) 2015-01-30 2019-05-07 Taiho Pharmaceutical Co., Ltd. Salt of fused pyrimidine compound and crystal thereof
US9908889B2 (en) 2015-01-30 2018-03-06 Taiho Pharmaceutical Co., Ltd. Salt of fused pyrimidine compound and crystal thereof
US9782412B2 (en) 2015-01-30 2017-10-10 Taiho Pharmaceutical Co., Ltd. Preventive and/or therapeutic agent of immune disease
US10213386B2 (en) 2015-03-03 2019-02-26 Pharmacyclics Llc Pharmaceutical formulations of a Bruton's tyrosine kinase inhibitor
US10010507B1 (en) 2015-03-03 2018-07-03 Pharmacyclics Llc Pharmaceutical formulations of a bruton's tyrosine kinase inhibitor
US10828259B2 (en) 2015-03-03 2020-11-10 Pharmacyclics Llc Pharmaceutical formulations of a Bruton's tyrosine kinase inhibitor
US9655857B2 (en) 2015-03-03 2017-05-23 Pharmacyclics Llc Pharmaceutical formulations of a Bruton's tyrosine kinase inhibitor
US11498916B2 (en) 2015-04-14 2022-11-15 Eisai R&D Management Co., Ltd. Crystalline FGFR4 inhibitor compound and uses thereof
US10562888B2 (en) 2015-04-14 2020-02-18 Eisai R&D Management Co., Ltd. Crystalline FGFR4 inhibitor compound and uses thereof
WO2016174183A1 (en) 2015-04-30 2016-11-03 Bayer Pharma Aktiengesellschaft Combinations of inhibitors of irak4 with inhibitors of btk
US10975080B2 (en) 2015-06-19 2021-04-13 Novartis Ag Compounds and compositions for inhibiting the activity of SHP2
US10308660B2 (en) 2015-06-19 2019-06-04 Novartis Ag Compounds and compositions for inhibiting the activity of SHP2
US10287266B2 (en) 2015-06-19 2019-05-14 Novartis Ag Compounds and compositions for inhibiting the activity of SHP2
US10774033B2 (en) 2015-07-24 2020-09-15 Celgene Corporation Methods of synthesis of (1R, 2R, 5R)-5-amino-2-methylcyclohexanol hydrochloride and intermediates useful therein
US10252981B2 (en) 2015-07-24 2019-04-09 Celgene Corporation Methods of synthesis of (1R,2R,5R)-5-amino-2-methylcyclohexanol hydrochloride and intermediates useful therein
US11192847B2 (en) 2015-07-24 2021-12-07 Celgene Corporation Methods of synthesis of (1R,2R,5R)-5-amino-2-methylcyclohexanol hydrochloride and intermediates useful therein
US11780801B2 (en) 2015-07-24 2023-10-10 Celgene Corporation Methods of synthesis of (1R,2R,5R)-5-amino-2-methyl-cyclohexanol hydrochloride and intermediates useful therein
WO2017036263A1 (en) * 2015-08-31 2017-03-09 广州科擎新药开发有限公司 2, 4-di-(nitrogen containing group) substituted pyrimidine compound and preparation method and use thereof
WO2017040617A1 (en) 2015-08-31 2017-03-09 Pharmacyclics Llc Btk inhibitor combinations for treating multiple myeloma
AU2016314898B2 (en) * 2015-08-31 2019-06-27 Bebetter Med Inc. 2,4-bis(nitrogen-containing group)-substituted pyrimidine compound, preparation method and use thereof
US11267802B2 (en) 2015-08-31 2022-03-08 Bebetter Med Inc. 2,4-bis(nitrogen-containing group)-substituted pyrimidine compound, preparation method and use thereof
US10399989B2 (en) 2015-09-16 2019-09-03 Loxo Oncology, Inc. Pyrazolopyrimidine derivatives as BTK inhibitors for the treatment of cancer
US10399990B2 (en) 2015-09-16 2019-09-03 Loxo Oncology, Inc. Pyrazolopyrimidine derivatives as BTK inhibitors for the treatment of cancer
US10611766B2 (en) 2015-09-16 2020-04-07 Loxo Oncology Inc. Pyrazolopyrimidine derivatives as BTK inhibitors for the treatment of cancer
EP3354647A4 (en) * 2015-09-25 2019-04-03 Zhejiang Bossan Pharmaceutical Co. Ltd. Egfr kinase inhibitor and preparation method and use thereof
AU2016327857B2 (en) * 2015-09-25 2019-10-03 Zhejiang Bossan Pharmaceutical Co. Ltd. EGFR kinase inhibitor and preparation method and use thereof
US10533011B2 (en) 2015-10-09 2020-01-14 ACEA Therapeutics, Inc. Pharmaceutical salts, physical forms, and compositions of pyrrolopyrimidine kinase inhibitors, and methods of making same
US10457669B2 (en) 2015-10-21 2019-10-29 Otsuka Pharmaceutical Co., Ltd. Benzolactam compounds as protein kinase inhibitors
US11001575B1 (en) 2015-10-21 2021-05-11 Otsuka Pharmaceutical Co., Ltd. Benzolactam compounds as protein kinase inhibitors
US10695323B2 (en) 2015-12-16 2020-06-30 Loxo Oncology, Inc. Compounds useful as kinase inhibitors
US11471441B2 (en) 2015-12-16 2022-10-18 Loxo Oncology Inc. Compounds useful as kinase inhibitors
US10342780B2 (en) 2015-12-16 2019-07-09 Loxo Oncology, Inc. Compounds useful as kinase inhibitors
US10918622B2 (en) 2015-12-16 2021-02-16 Loxo Oncology, Inc. Compounds useful as kinase inhibitors
US10464905B2 (en) 2015-12-16 2019-11-05 Loxo Oncology Inc. Compounds useful as kinase inhibitors
US11826351B2 (en) 2015-12-16 2023-11-28 Loxo Oncology Inc. Compounds useful as kinase inhibitors
US11001569B2 (en) 2016-01-22 2021-05-11 Janssen Pharmaceutica Nv 6-membered heteroaromatic substituted cyanoindoline derivatives as NIK inhibitors
US11180487B2 (en) 2016-01-22 2021-11-23 Janssen Pharmaceutica Nv Substituted cyanoindoline derivatives as NIK inhibitors
EP3195865A1 (en) 2016-01-25 2017-07-26 Bayer Pharma Aktiengesellschaft Combinations of inhibitors of irak4 with inhibitors of btk
US11357769B2 (en) 2016-05-10 2022-06-14 Eisai R&D Management Co., Ltd. Drug combinations for reducing cell viability and/or cell proliferation
US11098030B2 (en) 2016-05-26 2021-08-24 Recurium Ip Holdings, Llc EGFR inhibitor compounds
US10934285B2 (en) 2016-06-14 2021-03-02 Novartis Ag Compounds and compositions for inhibiting the activity of SHP2
US11905283B2 (en) 2016-06-14 2024-02-20 Novartis Ag Compounds and compositions for inhibiting the activity of SHP2
US11186589B2 (en) 2016-06-30 2021-11-30 Janssen Pharmaceutica Nv Cyanoindoline derivatives as NIK inhibitors
US11136311B2 (en) 2016-06-30 2021-10-05 Janssen Pharmaceutica Nv Heteroaromatic derivatives as NIK inhibitors
US11534431B2 (en) 2016-07-05 2022-12-27 Beigene Switzerland Gmbh Combination of a PD-1 antagonist and a RAF inhibitor for treating cancer
WO2018019252A1 (en) * 2016-07-26 2018-02-01 Jacobio Pharmaceuticals Co., Ltd. Novel fused pyridine derivatives useful as fak/aurora kinase inhibitors
WO2018019204A1 (en) * 2016-07-26 2018-02-01 深圳市塔吉瑞生物医药有限公司 Amino pyrimidine compound for inhibiting protein tyrosine kinase activity
US11111233B2 (en) 2016-07-26 2021-09-07 Shenzhen Targetrx, Inc. Amino pyrimidine compound for inhibiting protein tyrosine kinase activity
US11591340B2 (en) 2016-08-16 2023-02-28 Beigene Switzerland Gmbh Crystalline form of (S)-7-(1-acryloylpiperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetra- hydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
US11814389B2 (en) 2016-08-16 2023-11-14 Beigene Switzerland Gmbh Crystalline form of (S)-7-(1-acryloylpiperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetra-hydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
US11884674B2 (en) 2016-08-16 2024-01-30 Beigene Switzerland Gmbh Crystalline form of (S)-7-(1-acryloylpiperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetra- hydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
US11851437B2 (en) 2016-08-16 2023-12-26 Beigene Switzerland Gmbh Crystalline form of (S)-7-(1-acryloylpiperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetra-hydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
US10927117B2 (en) 2016-08-16 2021-02-23 Beigene Switzerland Gmbh Crystalline form of (S)-7-(1-acryloylpiperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetra-hydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
US11701357B2 (en) 2016-08-19 2023-07-18 Beigene Switzerland Gmbh Treatment of B cell cancers using a combination comprising Btk inhibitors
WO2018053437A1 (en) 2016-09-19 2018-03-22 Mei Pharma, Inc. Combination therapy
WO2018085731A2 (en) 2016-11-03 2018-05-11 Juno Therapeutics, Inc. Combination therapy of a t cell therapy and a btk inhibitor
US11555038B2 (en) 2017-01-25 2023-01-17 Beigene, Ltd. Crystalline forms of (S)-7-(1-(but-2-ynoyl)piperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
US11498922B2 (en) 2017-04-07 2022-11-15 ACEA Therapeutics, Inc. Pharmaceutical composition comprising N-(3-((2-((3-fluoro-4-(4-methylpiperazin-1-yl phenyl)amino)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)oxy)phenylacrylamide
US11427577B2 (en) 2017-04-14 2022-08-30 Biogen Ma Inc. Inhibiting agents for Bruton's tyrosine kinase
US10189829B2 (en) 2017-04-14 2019-01-29 Biogen Ma Inc. Inhibiting agents for Bruton's tyrosine kinase
US11858926B2 (en) 2017-04-14 2024-01-02 Biogen Ma Inc. Inhibiting agents for bruton's tyrosine kinase
US10961237B2 (en) 2017-04-14 2021-03-30 Biogen Ma Inc. Inhibiting agents for Bruton's tyrosine kinase
US10227341B2 (en) 2017-04-14 2019-03-12 Biogen Ma Inc. Inhibiting agents for bruton's tyrosine kinase
US11142518B2 (en) 2017-04-20 2021-10-12 Otsuka Pharmaceutical Co., Ltd. 6-pyrimidin-isoindole derivative as ERK1/2 inhibitor
EP3640248A4 (en) * 2017-06-13 2021-03-31 Beijing Adamadle Biotechnology Limited Liability Company Aminopyrimidine compound, preparation method therefor and use thereof
US11597768B2 (en) 2017-06-26 2023-03-07 Beigene, Ltd. Immunotherapy for hepatocellular carcinoma
US11377449B2 (en) 2017-08-12 2022-07-05 Beigene, Ltd. BTK inhibitors with improved dual selectivity
US11247987B2 (en) 2017-10-06 2022-02-15 Forma Therapeutics, Inc. Inhibiting ubiquitin specific peptidase 30
US11786529B2 (en) 2017-11-29 2023-10-17 Beigene Switzerland Gmbh Treatment of indolent or aggressive B-cell lymphomas using a combination comprising BTK inhibitors
US11248003B2 (en) 2017-12-07 2022-02-15 Oncobix Co., Ltd. Pyrimidine derivative having effect of inhibiting cancer cell growth and pharmaceutical composition containing same
CN111747934A (en) * 2018-01-31 2020-10-09 迪哲(江苏)医药有限公司 ERBB/BTK inhibitors
US11504375B2 (en) 2018-01-31 2022-11-22 Dizal (Jiangsu) Pharmaceutical Co., Ltd. ErbB/BTK inhibitors
US11007198B2 (en) 2018-01-31 2021-05-18 Dizal (Jiangsu) Pharmaceutical Co., Ltd. ErbB/BTK inhibitors
US11501284B2 (en) 2018-02-19 2022-11-15 Newave Pharmaceutical Inc. Substituted pyrazines as inhibitors of BTK, and mutants thereof
WO2019177374A1 (en) * 2018-03-13 2019-09-19 포로노이바이오 주식회사 2, 4, 5-substituted pyrimidine derivative, preparation method therefor, and pharmaceutical composition comprising same as effective ingredient for prevention or treatment of cancer or inflammatory disease
WO2019200254A1 (en) 2018-04-13 2019-10-17 Tolero Pharmaceuticals, Inc. Pim kinase inhibitors for treatment of myeloproliferative neoplasms and fibrosis associated with cancer
US11220494B2 (en) 2018-04-26 2022-01-11 Pfizer Inc. Cyclin dependent kinase inhibitors
US10766884B2 (en) 2018-04-26 2020-09-08 Pfizer Inc. Cyclin dependent kinase inhibitors
WO2019208805A1 (en) 2018-04-27 2019-10-31 小野薬品工業株式会社 PREVENTIVE AND/OR THERAPEUTIC AGENT FOR AUTOIMMUNE DISEASE COMPRISING COMPOUND HAVING Btk INHIBITORY ACTIVITY AS ACTIVE INGREDIENT
WO2019213184A1 (en) 2018-05-03 2019-11-07 Juno Therapeutics, Inc. Combination therapy of a chimeric antigen receptor (car) t cell therapy and a kinase inhibitor
US11040038B2 (en) 2018-07-26 2021-06-22 Sumitomo Dainippon Pharma Oncology, Inc. Methods for treating diseases associated with abnormal ACVR1 expression and ACVR1 inhibitors for use in the same
US11814386B2 (en) 2018-10-05 2023-11-14 Forma Therapeutics, Inc. Fused pyrrolines which act as ubiquitin-specific protease 30 (USP30) inhibitors
US11535618B2 (en) 2018-10-05 2022-12-27 Forma Therapeutics, Inc. Fused pyrrolines which act as ubiquitin-specific protease 30 (USP30) inhibitors
US11066404B2 (en) 2018-10-11 2021-07-20 Incyte Corporation Dihydropyrido[2,3-d]pyrimidinone compounds as CDK2 inhibitors
US11866432B2 (en) 2018-10-11 2024-01-09 Incyte Corporation Dihydropyrido[2,3-d]pyrimidinone compounds as CDK2 inhibitors
US11471456B2 (en) 2019-02-12 2022-10-18 Sumitomo Pharma Oncology, Inc. Formulations comprising heterocyclic protein kinase inhibitors
US11384083B2 (en) 2019-02-15 2022-07-12 Incyte Corporation Substituted spiro[cyclopropane-1,5′-pyrrolo[2,3-d]pyrimidin]-6′(7′h)-ones as CDK2 inhibitors
US11472791B2 (en) 2019-03-05 2022-10-18 Incyte Corporation Pyrazolyl pyrimidinylamine compounds as CDK2 inhibitors
US11919904B2 (en) 2019-03-29 2024-03-05 Incyte Corporation Sulfonylamide compounds as CDK2 inhibitors
US11447494B2 (en) 2019-05-01 2022-09-20 Incyte Corporation Tricyclic amine compounds as CDK2 inhibitors
US11440914B2 (en) 2019-05-01 2022-09-13 Incyte Corporation Tricyclic amine compounds as CDK2 inhibitors
US11427558B1 (en) 2019-07-11 2022-08-30 ESCAPE Bio, Inc. Indazoles and azaindazoles as LRRK2 inhibitors
US11427567B2 (en) 2019-08-14 2022-08-30 Incyte Corporation Imidazolyl pyrimidinylamine compounds as CDK2 inhibitors
US11851426B2 (en) 2019-10-11 2023-12-26 Incyte Corporation Bicyclic amines as CDK2 inhibitors
WO2022094172A3 (en) * 2020-10-30 2022-07-07 Newave Pharmaceutical Inc. Inhibitors of btk
WO2022094354A1 (en) * 2020-10-30 2022-05-05 Lengo Therapeutics, Inc. Pyrimidine compounds, compositions, and medicinal applications thereof
WO2023076167A1 (en) * 2021-10-25 2023-05-04 Newave Pharmaceutical Inc. Inhibitor of btk and mutants thereof
WO2023220655A1 (en) 2022-05-11 2023-11-16 Celgene Corporation Methods to overcome drug resistance by re-sensitizing cancer cells to treatment with a prior therapy via treatment with a t cell therapy
US11786531B1 (en) 2022-06-08 2023-10-17 Beigene Switzerland Gmbh Methods of treating B-cell proliferative disorder
US11896596B2 (en) 2022-06-08 2024-02-13 Beigene Switzerland Gmbh Methods of treating B-cell proliferative disorder
US11911386B2 (en) 2022-06-08 2024-02-27 Beigene Switzerland Gmbh Methods of treating B-cell proliferative disorder

Also Published As

Publication number Publication date
US8609679B2 (en) 2013-12-17
JP2011526299A (en) 2011-10-06
CA2727455A1 (en) 2009-12-30
JP2014208674A (en) 2014-11-06
KR101955914B1 (en) 2019-03-11
RU2536584C2 (en) 2014-12-27
EP3549934A1 (en) 2019-10-09
JP2019194235A (en) 2019-11-07
TWI458721B (en) 2014-11-01
US20130165462A1 (en) 2013-06-27
EP2361248A1 (en) 2011-08-31
CN108047142A (en) 2018-05-18
IL209969A (en) 2017-02-28
TWI613196B (en) 2018-02-01
JP6554507B2 (en) 2019-07-31
TWI546290B (en) 2016-08-21
US10828300B2 (en) 2020-11-10
CA3031835A1 (en) 2009-12-30
NZ603525A (en) 2015-02-27
NZ624345A (en) 2016-07-29
KR101892989B1 (en) 2018-08-30
NZ589843A (en) 2012-12-21
RU2010151355A (en) 2012-08-10
US9212181B2 (en) 2015-12-15
JP2017137352A (en) 2017-08-10
TW201716387A (en) 2017-05-16
BRPI0914682B8 (en) 2021-05-25
KR20160145837A (en) 2016-12-20
IL230290A (en) 2016-06-30
JP6141800B2 (en) 2017-06-07
US20130072469A1 (en) 2013-03-21
IL250108A0 (en) 2017-03-30
PH12015501484A1 (en) 2017-06-05
CN108047142B (en) 2021-08-03
IL209969A0 (en) 2011-02-28
TW201437202A (en) 2014-10-01
KR20180045065A (en) 2018-05-03
CA2986640A1 (en) 2009-12-30
KR20110025224A (en) 2011-03-09
RU2734822C2 (en) 2020-10-23
ZA201009216B (en) 2018-11-28
WO2009158571A8 (en) 2010-02-25
MX357627B (en) 2018-07-17
MX360970B (en) 2018-11-23
AU2009262068A1 (en) 2009-12-30
EP2361248B1 (en) 2018-09-19
CA2986640C (en) 2019-03-26
US20130065879A1 (en) 2013-03-14
BRPI0914682B1 (en) 2020-09-24
AU2009262068C1 (en) 2015-07-02
RU2014117866A (en) 2015-11-10
BRPI0914682A2 (en) 2015-10-20
US20100029610A1 (en) 2010-02-04
TW201004940A (en) 2010-02-01
US8450335B2 (en) 2013-05-28
US20190117650A1 (en) 2019-04-25
CA3031835C (en) 2021-09-07
DK2361248T3 (en) 2019-01-14
AU2009262068B2 (en) 2014-12-11
MX2010014029A (en) 2011-01-21
ES2711249T3 (en) 2019-04-30
US9987276B2 (en) 2018-06-05
JP6853307B2 (en) 2021-03-31
US20160303121A1 (en) 2016-10-20
US9296737B2 (en) 2016-03-29
CN102083800A (en) 2011-06-01
CA2727455C (en) 2019-02-12
SG10201510696RA (en) 2016-01-28

Similar Documents

Publication Publication Date Title
AU2010343055B2 (en) Heteroaryl compounds and uses thereof
AU2009262068B2 (en) Heteroaryl compounds and uses thereof
AU2015202675B2 (en) Heteroaryl compounds and uses thereof
WO2010123870A1 (en) Heteroaryl compounds and uses thereof
AU2016253570B2 (en) Heteroaryl compounds and uses thereof
AU2013202496B2 (en) Heteroaryl compounds and uses thereof
BR112012018345B1 (en) HETEROARIL COMPOUNDS AND COMPOSITIONS INCLUDING THEM

Legal Events

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

Ref document number: 200980124410.4

Country of ref document: CN

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

Ref document number: 09771102

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2009262068

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2727455

Country of ref document: CA

Ref document number: 589843

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 209969

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: MX/A/2010/014029

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 12010502916

Country of ref document: PH

ENP Entry into the national phase

Ref document number: 2011516699

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2009262068

Country of ref document: AU

Date of ref document: 20090626

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2009771102

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20117002052

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 628/DELNP/2011

Country of ref document: IN

Ref document number: 2010151355

Country of ref document: RU

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 12015501484

Country of ref document: PH

ENP Entry into the national phase

Ref document number: PI0914682

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20101223

WWE Wipo information: entry into national phase

Ref document number: 250108

Country of ref document: IL