CA2783624A1 - Heteroaryl compounds and uses thereof - Google Patents

Heteroaryl compounds and uses thereof Download PDF

Info

Publication number
CA2783624A1
CA2783624A1 CA2783624A CA2783624A CA2783624A1 CA 2783624 A1 CA2783624 A1 CA 2783624A1 CA 2783624 A CA2783624 A CA 2783624A CA 2783624 A CA2783624 A CA 2783624A CA 2783624 A1 CA2783624 A1 CA 2783624A1
Authority
CA
Canada
Prior art keywords
ring
compound
moiety
substituted
group
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CA2783624A
Other languages
French (fr)
Other versions
CA2783624C (en
Inventor
Juswinder Singh
Russell Petter
Richland Wayne Tester
Arthur F. Kluge
Hormoz Mazdiyasni
William Frederick Westlin, Iii
Deqiang Niu
Lixin Qiao
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Celgene CAR LLC
Original Assignee
Avila Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Avila Therapeutics Inc filed Critical Avila Therapeutics Inc
Publication of CA2783624A1 publication Critical patent/CA2783624A1/en
Application granted granted Critical
Publication of CA2783624C publication Critical patent/CA2783624C/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • 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
    • 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
    • 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
    • A61K31/495Heterocyclic 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
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/541Non-condensed thiazines containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs 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
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • 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
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • 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
    • 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
    • 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
    • C07D453/00Heterocyclic compounds containing quinuclidine or iso-quinuclidine ring systems, e.g. quinine alkaloids
    • C07D453/02Heterocyclic compounds containing quinuclidine or iso-quinuclidine ring systems, e.g. quinine alkaloids containing not further condensed quinuclidine ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D487/02Heterocyclic 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
    • C07D487/08Bridged 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Abstract

The present invention provides heteroaryl compounds according to formula I-a or I-b, pharmaceutically acceptable compositions thereof, and methods of using the same.
(see above formula)

Description

HETEROARYL COMPOUNDS AND USES THEREOF

CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] The present application claims priority to United States serial number 12/648693, filed December 29, 2009, which is a continuation-in-part of United States serial number 12/492,180, filed June 26, 2009, which claims priority to United States provisional application serial number 61/076,450, filed June 27, 2008, United States provisional application serial number 61/148,388, filed January 29, 2009, and United States provisional application serial number 61/170,874, filed April 20, 2009, the entirety of each of which is hereby incorporated by reference.

TECHNICAL FIELD OF THE INVENTION
[0002] 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
[0003] 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.
[0004] 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.).
[0005] 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 H202), cytokines (e.g., interleukin-1 (IL-1) and tumor necrosis factor a (TNF-a)), 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.
[00061 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
[00071 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:

(RV R1 A (RV A

y R1 y N N
B (RX) `LJ (RX) I-a I-b or a pharmaceutically acceptable salt thereof, wherein Ring A, Ring B, m, p, W, Ry, R ,W', W2, and Rl are as defined herein.
[00081 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.
[00091 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 ofp-plc gamma2 with compound 1-35 in Ramos cells.
Figure 5 depicts dose response inhibition ofp-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 [00101 In certain embodiments, the present invention provides a compound of formula I-a or I-b:

( V~p R1 A (R"p A

y R1 Y

B (Rx)m 1 &(RX~m 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;
R' is a warhead group;
Ry is hydrogen, halogen, -CN, -CF3, C1_4 aliphatic, C1_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;
W' and W2 are each independently a covalent bond or a bivalent CI-3 alkylene chain wherein one methylene unit of W' or W2 is optionally replaced by -NR2-, -N(R2)C(O)-, -C(O)N(R2)-, -N(R2)SO2-, -SO2N(R2)-, -0-, -C(0)-, -OC(O)-, -C(0)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;
in and p are independently 0-4; and RX and R are independently selected from -R, halogen, -OR, -O(CH2)gOR, -CN, -N02, -S02R, -S02N(R)2, -SOR, -C(O)R, -C02R, -C(O)N(R)2, -NRC(O)R, -NRC(O)NR2, -NRS02R, or -N(R)2, wherein q is 1-4; or:
Rx and RI 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 and RI 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 [00111 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.
[00121 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"
6 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.
[00131 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.
[00141 The term "lower haloalkyl" refers to a Ci_4 straight or branched alkyl group that is substituted with one or more halogen atoms.
[00151 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)).
[00161 The term "unsaturated", as used herein, means that a moiety has one or more units of unsaturation.
[00171 As used herein, the term "bivalent Ci_s (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.
[00181 The term "alkylene" refers to a bivalent alkyl group. An "alkylene chain" is a polymethylene group, i.e., -(CH,)ri , 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.
7 [00191 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.
[00201 As used herein, the term "cyclopropylenyl" refers to a bivalent cyclopropyl group of the following structure:
[00211 The term "halogen" means F, Cl, Br, or I.
[00221 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.
[00231 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 ic 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,
8 quinazolinyl, quinoxalinyl, 4H-quinolizinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, and pyrido[2,3-b]-1,4-oxazin-3(4H)-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.
[0024] 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 N-substituted pyrrolidinyl).
[0025] 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.
[0026] 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
9 rings having multiple sites of unsaturation, but is not intended to include aryl or heteroaryl moieties, as herein defined.
[0027] 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.
[0028] Suitable monovalent substituents on a substitutable carbon atom of an "optionally substituted" group are independently halogen; -(CH2)0 4R ; -(CH2)0_40R ; -O(CH2)0_4R , -0-(CH2)0 4C(O)OR ; -(CH2)0 4CH(OR )2; -(CH2)0 4SR ; -(CH2)o4Ph, which may be substituted with R ; -(CH2)0_40(CH2)o-iPh which may be substituted with R ; -CH-CHPh, which may be substituted with R ; -(CH2)o40(CH2)o-i-pyridyl which may be substituted with R
; -NO2; -CN;
-N3; -(CH2)0 4N(R )2, -(CH2)0 4N(R )C(O)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(O)OR ; -N(R )N(R )C(O)R ; -N(R )N(R )C(O)NR 2;
-N(R )N(R )C(O)OR ; -(CH2)0 4C(O)R ; -C(S)R ; -(CH2)0 4C(O)OR ; -(CH2)0 4C(O)SR ;
-(CH2)0 4C(O)OSiR 3; -(CH2)0 4OC(O)R ; -OC(O)(CH2)0 4SR , SC(S)SR ; -(CH2)0 4SC(O)R ;
-(CH2)0 4C(O)NR 2, -C(S)NR 2, -C(S)SR ; -SC(S)SR , -(CH2)0-40C(O)NR 2, -C(O)N(OR )R ; -C(O)C(O)R ; -C(O)CH2C(O)R ; -C(NOR )R ; -(CH2)0 4SSR ; -(CH2)0-4S(0)2R ; -(CH2)0-4S(0)20R ; -(CH2)0 4OS(0)2R ; -S(0)2NR 2; -(CH2)0_4S(O)R ;
-N(R )S(0)2NR 2; -N(R )S(0)2R ; -N(OR )R ; -C(NH)NR 2; -P(0)2R ; -P(O)R 2; -OP(O)R 2;
-OP(O)(OR )2; SiR 3; -(C1 straight or branched alkylene)O-N(R )2, or -(C1_4 straight or branched alkylene)C(O)0 N(R )2, wherein each R may be substituted as defined below and is independently hydrogen, Ci_6 aliphatic, -CH2Ph, -O(CH2)o_iPh, -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 R , 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.
[00291 Suitable monovalent substituents on R (or the ring formed by taking two independent occurrences of R together with their intervening atoms), are independently halogen, -(CH2)0_2R', -(haloR'), -(CH2)0_20H, -(CH2)0_20R', -(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)0_2SH, -(CH2)o_2NH2, -(CH2)0_2NHR', -(CH2)0_2NR'2, -NO2, -SiR'3, -OSiR'3, -C(O)SR', -(C1 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_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. Suitable divalent substituents on a saturated carbon atom of R
include -0 and -S.
[00301 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(0)2R*, -NR*, -NOR*, -O(C(R*2))2_30-, 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_30-, 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.
[00311 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 -N02, wherein each R' is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently C1_4 aliphatic, -CH2Ph, -O(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.
[00321 Suitable substituents on a substitutable nitrogen of an "optionally substituted" group include -Rt, -NRt2, -C(O)W, -C(O)ORt, -C(O)C(O)Rt, -C(O)CH2C(O)Rt, -S(O)2Rt, -S(O)2NRt2, -C(S)NRt2, -C(NH)NRt2, or -N(R)S(O)2Rt; wherein each Rt 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 Rt, 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.
[00331 Suitable substituents on the aliphatic group of Rt are independently halogen, -R', -(haloR'), -0H, -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)o-iPh, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[00341 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.
[00351 Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N+(Cl 4alkyl)4 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.
[00361 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 Rl group of formula I-a and I-b comprises one or more deuterium atoms.
[00371 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 radiolabelled inhibitor, to show covalent modification of the enzyme, as well as other methods known to one of skill in the art.
[00381 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.
[00391 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 M, less than about 1 M, less than about 500 nM, less than about 100 nM, or less than about 10 nM, [00401 The terms "measurable affinity" and "measurably inhibit," as used herein, means a measurable change in at least one of ErbB 1, 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 ErbB 1, 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 [00411 According to one aspect, the present invention provides a compound of formula I-a or I-b, (R"gyp R1 (R"p A

y R1 Y
N N ~~
B (RX)m ~, / (RX)m 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;
R' 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)S02-, -SO2N(R)-, -0-, -C(0)-, -OC(O)-, -C(0)O-, -S-, -SO-, -SO2-, -C(-S)-, -C(-NR)-, -N-N-, or -C(-N2)-;
Y is hydrogen, C]_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 C1_6 saturated or unsaturated, straight or branched, hydrocarbon chain, wherein one or two methylene units of Q are optionally and independently replaced by -NR-, -5-, -0-, -C(0)-, -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;
W' and W2 are each independently a covalent bond or a bivalent C1_3 alkylene chain wherein one methylene unit of W' or W2 is optionally replaced by -NR2-, -N(R2)C(O)-, -C(O)N(R2)-, -N(R2)S02-, -SO2N(R2)-, -0-, -C(0)-, -OC(O)-, -C(0)O-, -5-, -SO- or -SO2-;
R2 is hydrogen, optionally substituted C1_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 R are independently selected from -R, halogen, -OR, -O(CH2)qOR, -CN, -NO2, -S02R, -S02N(R)2, -SOR, -C(O)R, -C02R, -C(O)N(R)2, -NRC(O)R, -NRC(O)NR2, -NRS02R, or -N(R)2, or:
Rx and R' 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.
[00421 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.
[00431 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, R , or -(CH2)0- 40R , or -O(CH2)0_4R , wherein each R is as defined herein.
Exemplary substituents on Ring A include Br, I, Cl, methyl, -CF3, -C=CH, -OCH2phenyl, -OCH2(fluorophenyl), or -OCH2pyridyl.
[00441 Exemplary Ring A groups are set forth in Table 1.
Table 1. Exemplary Ring A Groups Br CH3 OCH3 6o"

i ii iii iv \N \ F CI CF3 0\ C 0\ C F\ I \

v vi vii viii I
n N
N iN iN

o\ I o o\ I o\
ix x xi xii F

/ : 0 C~ )N CL, Br F
N cl): jkIP-xiii xiv xv xvi xvii \ 0 F, Cl, CN S F, Cl, CN

xviii xix ys F, CI,CN O / \ O /
N N\
xx xxi X -vii a N N

Qo7O1, S V
xxiii xxiv xxv F CI

xxvi xxvii xxviii Me0 I / \ NC cvoci]

xxrx xxx O o N
R
SSS
O N S O N\ ssss xxxi xxxii xxxiii O
flN)LN

O J/
xxxiv xxxv xxxvi N I N ,N "\N/

xxxvii xxxviii .x ix O ~ N O N/

NON p \ I 'S SS
S' Si xl xli xlii N ~N
S O~
s' xliii xliv O
R _ O R O R y SSSS
~ __~___~` OWN SAN HNC

xlv xlvi xlvii R / I
NiN \
Rt xlviii R O
\ N

Is -Na S, ,S~ , 0 S"
xlix l li / rs \ N ~ I \ F / \ I I \~ N / N J,QN

lii liii liv lv lvi lvii R1`N // (:~o / COX, R R
lviii lix lx lxi lxii / N :a R1 / CF3 R1~N
\ I N \ I N \ I N
R H R
lxiii lxiv lxv lx i lxvii lx iii H
qNO-, N N O R1_N
NC CI

ss`f R

Ixix lxx lxxi lxxii lxxiii lxxiv /
R1~
NO N \ `` Na NC. R1'Nal" 0~
R1' '11 R1 R1 ~: R1`' /"

lxxv lxxvi lxxvii lxxviii lxxix lxxx / O1 O~~O / O' R1 ~ ~ O ~ O
Ixxxi lxxvii lx=iii lx=iv or lxxxv, wherein each R , Rt, and R' is as defined above and described in classes and subclasses herein.
[0045] In certain embodiments, Ring A is selected from i, ii, iv, v, vi, vii, ix, xiv, xvi, Iii, lxiii, lxxi, lxxiv, lxxvi, lxxviii, and lxxxi.
[0046] 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.

[0047] In some embodiments, Ring B is phenyl. In some embodiments, Ring B is a 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.
[00481 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.
[00491 In some embodiments, Ring B is an optionally substituted group selected from phenyl, pyridyl, pyrazinyl, pyrimidinyl, imidazolyl, pyrrolidinyl, piperdinyl, indolinyl, indazolyl, and isoindolinyl.
[00501 Exemplary Ring B groups are set forth in Table 2.
Table 2. Ring B Groups N/ i N
--'~ \ ) \):: I
N
)--N
Rx Rx Rx Rx Rx Rx l ii iii iv v Vi R' R1 N N

N N .~.
\f-11) Rx Rx NZ
s'0 vii viii lx x xi xii xiii xiv 0 S'NHZ

F / \ \ / \ CI \ OMe \ I I N CI N

xv xvi xvii xviii xix xx xxi N
N) N
OH OMe "'Co xxii xxiii xxiv xxv O''Co I \\
p 0\ O o ~ al~

,,,a CF3 `~
"~.
H
xxvi xxvii xxviii xxlx CI

~p\/
ao-, N \ rO,r xxx xxxi xxxii xxxiii S;p O~~pi ON- OH NJ CI
N
OMe xxxiv xxxv xxxvi xxxvii ' \
\ /
N N N
xxxviii xxxix xl xli N" I \
0" O OS /

N I N N"- ~
OH
xlii xliii xliv \ CI

/ N \ \ 0 C \
0/ I/ ~OH N ~D , I/ F
xlv xlvi xlvii xlviii N ,,,-\ / I \\ I \ "r ,-,, iOH N
0 4`'z 0 xlix l li lii aN 0 \ \
. OMe '''a O 0 N
N / I N
IN liv lv lvi N ,_,,\OH
IS O N

lvii lviii lix lx aN 0 0 N ~ \ ~ \ 0 \ r0 ~ 1, ~/ -~NJ
R l 0 0 lxi lxii lxiii lxiv lxv \ O 0 \
N N I i x \
O 'SN, N N CF3 OH

lxvi lxvii lxvvii lxix N N, \ N, N 0 I ~N N
lxx lxxi lxxii lxxiii H \ N N N I /

H
lxxiv lxxv lxxvi lxxvii \ I \

\ I / ~/~ / 0~~ N . CN
/ ~`+ 0 N N

lxxviii lxxix lxxx lxxxi ,a N F H R
/ ~\ I 0 OH \ C

lxxxii lxxxiii lxxxiv lxxxv lxxxvi 4' 0 1CNH2+ O 1CNH2 X 1CNH2 lxxxvii lxxxviii lxxxix \ N~ I \ N~ ~'' I / N CO
xc xci xcii xciii 0",~,aNH2 N NH2 ao"~
/ /~I '\/ , '\/ 0 N H

xciv xcv xcvi H
'~)'N'2 xcvii xcviii xcix N 0"",a N OC \ 0.,'ao*Co off Co c ci cii ciii OH

\\ O~/S\ I \ CI ~ I CNH \ O\` ` H
F 0 =OH N F
civ cv cvi cvii N
Oi O O

N N
cviii cix cx cxi CI
0 \/ \ O~/~Oi\i N ~O \ O~/~Oi\i N3 cxii cxiii cxiv \ O OH I \ O~'OH I \ o' -('O C HZ I \ 0'-"^N3 " F ~ F ~ F

cxv cxvi cxvii cxviii F "'(\'O' 0,,,,0 C H2 I \ 0\ XO~ D I \ 0~~0~~0~~ N3 %
cxxix cxx cxii rN
NJ
N

01*11 or cxxii, wherein each R' and RX is as defined above and described in classes and subclasses herein.
[00511 In certain embodiments, Ring B is selected from i, ii, iii, iv, v, ix, x, xi, xiii, xvi, xvii, xix, xx, xxv, xl'vi, xxxii, xxxiv, xxxv, xxxviii, xlii, xlvi, xlviii,1, lviii, lxiv, 1xxviii, L xxiii, lxxxvi, xciv, c, ci, cii, ciii, civ, and cv.

[0052] In some embodiments, the in moiety of formula I is 1, 2, 3 or 4. In some embodiments, in is 1. In other embodiments, in is 0.
[0053] 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.
[0054] As defined generally above, each RX group of formula I is independently selected from -R, halogen, -OR, -O(CH2)qOR, -CN, -N02, -S02R, -S02N(R)2, -SOR, -C(O)R, -C02R, -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.
[0055] In some embodiments, each instance of R" 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.
[0056] As defined generally above, each R group of formula I is independently selected from -R, halogen, -OR, -O(CH2)qOR, -CN, -N02, -S02R, -SO2N(R)2, -SOR, -C(O)R, -C02R, -C(O)N(R)2, -NRC(O)R, -NRC(O)NR2, -NRSO2R, or -N(R)2, wherein q is 1-4, or R
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 C1_6 aliphatic.
[0057] In some embodiments, each instance of R is independently selected from -R, -OR, -O(CH2)qOR, or halogen. In certain embodiments, R is lower alkyl, lower alkoxy, lower alkoxyalkoxy, or halogen. Exemplary R groups include methyl, methoxy, trifluoromethyl, methoxyethoxy, and chloro. In some embodiments, R is hydrogen.
[0058] 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.
[0059] As defined generally above, Ry is hydrogen, halogen, -CN, -CF3, Ci4 aliphatic, C14 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.
[00601 As generally defined above, W' and W2 are each independently a covalent bond or a bivalent C1_3 alkylene chain wherein one methylene unit of W' or W2 is optionally replaced by -NR2-, -N(R2)C(O)-, -C(O)N(R2)-, -N(R')S02-, -SO2N(R2)-, -0-, -C(0)-, -OC(O)-, -C(0)O-, -5-, -SO- or -S02-. In certain embodiments, W1 and W2 are the same. In some embodiments, W' and W2 are different.
[00611 In some embodiments, W1 is a covalent bond. In certain embodiments, W' is a bivalent C1.3 alkylene chain wherein one methylene unit of W1 is optionally replaced by -NR2-, -N(R2)C(O)-, -C(O)N(R2)-, -N(R2)S02-, -S02N(R2)-, -0-, -C(0)-, -OC(O)-, -C(0)O-, -5-, -SO-or -SO2-, In certain embodiments, W' is -C(-0), -NR2-, -5-, or -0-. In some embodiments, W' is -NR2-. In other embodiments, W' is -0-. In certain embodiments, W1 is -NH-, -5-, or -0-.
In some embodiments, W' is -CH2O-, -CH2S-, or -CH2NH-. In some aspects, W1 is -OCH2-, -SCH2-, NHCHz-, or -CH2CH2-.
[00621 In certain embodiments, W2 is a covalent bond. In some embodiments, W2 is a bivalent C1_3 alkylene chain wherein one methylene unit of W2 is optionally replaced by -NR2-, -N(R2)C(O)-, -C(O)N(R2)-, -N(R2)S02-, -SO2N(R2)-, -0-, -C(0)-, -OC(O)-, -C(0)O-, -5-, -SO-or -SO2-. In certain embodiments, W2 is -C(-0), -NR2-, -5-, or -0-. In some embodiments, W2 is -NR2-. In other embodiments, W2 is -0-. In certain embodiments, W2 is -NH-, -5-, or -0-.
In some embodiments, W2 is -CH2O-, -CHzS-, or -CH2NH-. In some aspects, W2 is -OCH2-, -SCH2-, -NHCH2-, or -CH2CH2-.
[00631 In some embodiments, Ring B is phenyl, thus forming a compound of formula 11-a or II-b:

(RV)p R1 A W1 R1 (RV n A W1 Y

2 / (Rx)m ' Z I = (Rx)m N w N W
II-a II-b or a pharmaceutically acceptable salt thereof, wherein each of Ring A, in, p, Rx, Ry, R ,W', W2, and R' is as defined above and described in classes and subclasses above and herein.
[0064] In certain embodiments, Ring A is phenyl, thus forming a compound of formula 111-a or III-b:

(Rv)p R1 (Rv )pT/

Ry N II WzB (R )m I~RX)m /~ N Wz 111-a 111-b or a pharmaceutically acceptable salt thereof, wherein each of Ring B, in, p, W, Ry, R ,W', W2, and R1 is as defined above and described in classes and subclasses above and herein.
[0065] In certain embodiments, Ring A is phenyl and Ring B is phenyl, thus forming a compound of formula IV-a or fV-b:

(Rv)p R1 (R )p ~% \ 1 Y/ v N N / I X
J,Wz \ I ~R )m 2 (R"). N
N W
IV-a IV-b or a pharmaceutically acceptable salt thereof, wherein each of m, p, Rx, Rb', R ,W', W2, and R1 is as defined above and described in classes and subclasses above and herein.
[0066] As defined generally above, each R2 is independently hydrogen, optionally substituted C1_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 C1_6 aliphatic group.

[00671 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-i or I-b-i:

N )1-3 '~6N 43 R~

Ry I\ N B (RX) RY I\ N B (R") NW2 NWz I-a-i I-b-i or a pharmaceutically acceptable salt thereof, wherein each of Ring A, R1, RX, and in are as defined above and described in classes and subclasses above and herein.
[00681 Similar to the formation of compounds of formulae I-a-i and I-b-i above, it will be understood by one skilled in the art that compounds of formulae 11-a, 11-b, 111-a, 111-b, IV-a, and IV-b, will form corresponding compounds 11-a-i, II-b-i, 111-a-i, III-W, IV-a-i, and IV-b-i when 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.
[00691 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-ii or I-b-ii:

V)p ~ V)p A A

a2 N N N D R)m N N (Rx)m N B (~. D / 1 W2 N W2 I-a-ii I-b-ii or a pharmaceutically acceptable salt thereof, wherein each of Ring A, R1, RX, and in are as defined above and described in classes and subclasses above and herein.
[00701 Similar to the formation of compounds of formulae I-a-ii and I-b-ii above, it will be understood by one skilled in the art that compounds of formulae 11-a, 11-b, 111-a, 111-b, IV-a, and IV-b, will form corresponding compounds II-a-ii, II-b-ii, III-a-ii, III-b-ii, IV-a-ii, and IV-b-ii when R2 and RY are taken together with their intervening atoms to form a 4-7 membered partially unsaturated ring.

[00711 As defined generally above, the R1 group of formulae I and II is -L-Y, wherein:
L is a covalent bond or a bivalent C1_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)S02-, -SO2N(R)-, -0-, -C(0)-, -OC(O)-, -C(0)O-, -S-, -SO-, -SO2-, -C(-S)-, -C(-NR)-, -N-N-, or -C(-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 C1_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)-, -5-, -0-, -C(0)-, -OC(O)-, -C(0)O-, -SO-, or -SO2-, -N(R)C(O)-, -C(O)N(R)-, -N(R)S02-, or -SO2N(R)-; and Z is hydrogen or C1_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN.
[00721 In certain embodiments, L is a covalent bond.
[00731 In certain embodiments, L is a bivalent Ci_8 saturated or unsaturated, straight or branched, hydrocarbon chain. In certain embodiments, L is -CH2-.
[00741 In certain embodiments, L is a covalent bond, -CHz-, -NH-, -CH2NH-, -NHCH2-, -NHC(O)-, -NHC(O)CH2OC(O)-, -CH2NHC(O)-, -NHSO2-, -NHSO2CH2-, -NHC(O)CH2OC(O)-, or -SO2NH-.
[00751 In some embodiments, L is a bivalent Cz_g 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)S02-, -SO2N(R)-, -S-, -S(O)_'_ S02-, -OC(O)-, -C(0)O-, cyclopropylene, -0-, -N(R)-, or -C(0)-.
[00761 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(0)-, -NRC(O)-, -C(O)NR-, -N(R)SO2-, -SO2N(R)-, -S-, -S(0)-, -SO2-, -OC(O)-, or -C(0)0-, and one or two additional methylene units of L are optionally and independently replaced by cyclopropylene, -0-, -N(R)-, or -C(0)-.
[0077] In some embodiments, L is a bivalent Cz_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(0)-, and one or two additional methylene units of L are optionally and independently replaced by cyclopropylene, -0-, -N(R)-, or -C(0)-.
[0078] As described above, in certain embodiments, L is a bivalent C2 2 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-.
[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 -C(0)-. 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-.
[0080] 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)-.
[0081] 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 -NRC(O)-, -C(O)NR-, -N(R)S02-, -SO2N(R)-, -S-, -S(0)-, -SO2-, -OC(O)-, or -C(0)O-, and one or two additional methylene units of L are optionally and independently replaced by cyclopropylene, -0-, -N(R)-, or -C(0)-. In some embodiments, L is -CH2OC(O)CH-CHCH2-, -CH2-OC(O)CH-CH-, or -CH(CH-CH2)OC(O)CH-CH-.

[0082] In certain embodiments, L is -NRC(O)CH-CH-, -NRC(O)CH-CHCH2N(CH3)-, -NRC(O)CH-CHCH2O-, -CH2NRC(O)CH-CH-, -NRS02CH-CH-, -NRS02CH-CHCH2-, -NRC(O)(C-N2)C(O)-, -NRC(O)CH-CHCH2N(CH3)-, -NRS02CH-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.
[00831 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-.
[00841 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_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-, -S-, -S(0)-, -SO2-, -C(-S)-, -C(-NR)-, -0-, -N(R)-, or -C(0)-. 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(0)-, -C(0)0-, or -OC(O)-, or -0-.
[00851 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-.
[00861 In certain embodiments, 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 -C(0)-, -NRC(O)-, -C(O)NR-, -N(R)S02-, or -SO2N(R)-. Exemplary L groups include -NHC(O)-cyclopropylene-S02- and -NHC(O)-cyclopropylene-.
[00871 As defined generally above, Y is hydrogen, C1_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, N02, halogen, CN, a suitable leaving group, or Ci_6 aliphatic, wherein Q is a covalent bond or a bivalent C1_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(0)-, -OC(O)-, -C(0)O-, -SO-, or -SO2-, -N(R)C(0)-, -C(O)N(R)-, -N(R)S02-, or -SO2N(R)-; and, Z is hydrogen or C1_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN.
[0088] In certain embodiments, Y is hydrogen.
[0089] In certain embodiments, Y is C1_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, N02, or CN. In some embodiments, Y is C2.6 alkenyl. In other embodiments, Y is C2.4 alkynyl.
[0090] In other embodiments, Y is Ci_6 alkyl substituted with oxo, halogen, N02, or CN.
Such Y groups include -CH2F, -CH2C1, -CH2CN, and -CH2NO2.
[0091] 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.
[0092] 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 R' 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.
[0093] 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 (Re)1-2 ~~N-Q-Z NR
above and described herein. In certain embodiments, Y is 1-2 1-2, or (Re)1-2 QX`q ~

1-2 e wherein each R, Q, Z, and R' is as defined above and described herein.
[0094] 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 R' groups, wherein each R' is as defined above and described herein..

Re In certain embodiments, Y is , wherein Re is as defined above and described herein.
In certain embodiments, Y is cyclopropyl optionally substituted with halogen, CN or NO2.
[0095] 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.
[0096] 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 (RI)1-2 and described herein. In certain embodiments, Y is wherein each Re is as defined above and described herein.
[0097] 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:

S~N /]
(Re)1-2 12 -2 +//1-2 0 (RI)1-2 (Re )1-2 (Re )1-2 wherein each R and Re is as defined above and described herein.
[0098] 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.
[0099] In some embodiments, Y is selected from:

N N N N
!/ fL 1 (Re)1 4 f~l `l (Re)1-4 N ~Re~1 3 k /JN (Re)1-3 (Re)1-3 N

wherein each Re is as defined above and described herein.
[00100] 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:
R R R R
N % N N. ` N~

~ (Re)1 3 N (Re)1 z N(Re)1 2 NRe .llJ1. .l11~.
N NI N
l NON
` \ e N < N e \ , (Re)1-3 N (R )1-2 (Re)1-2 N- R

0 0 01% ON, ( N
`L_Js(Re)1-3 N (Re)1-2 `V\(Re)1-2 IVY Re /S ~S S S*%
C` \\ -'.I N e /\(Re)1 3 L~ --N (R )12 ~(Re)1-2 N R
wherein each R and Re is as defined above and described herein.
[00101] 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.

[001021 As defined generally above, each Re group is independently selected from -Q-Z, oxo, NO2, halogen, CN, a suitable leaving group, or C1_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN, wherein Q is a covalent bond or a bivalent C1_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(0)-, -OC(O)-, -C(0)0-, -SO-, or -SO2-, -N(R)C(0)-, -C(O)N(R)-, -N(R)S02-, or -SO2N(R)-; and Z is hydrogen or C1_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN.
[001031 In certain embodiments, Re is C1_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN. In other embodiments, Re is oxo, NO2, halogen, or CN.
[001041 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-, -0-, -C(0)-, -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(0)-, -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-CHz.
[001051 In certain embodiments, each Re is independently selected from from oxo, NO2, CN, fluoro, chloro, -NHC(0)CH-CH2, -C(0)CH-CH2, -CH2CH-CH2, -C=CH, -C(0)OCH2C1, -C(O)OCH2F, -C(0)OCH2CN, -C(0)CH2C1, -C(0)CH2F, -C(0)CH2CN, or -CH2C(O)CH3.
[001061 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, 5`h 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).

[001071 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)S02-, -SO2N(R)-, -S-, -S(0)-, -SO2-, -OC(O)-, -C(0)O-, cyclopropylene, -0-, -N(R)-, or -C(0)- ; and Y is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, N02, 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(0)-, -NRC(O)-, -C(O)NR-, -N(R)S02-, -SO2N(R)-, -5-, -S(0)-, -SO2-, -OC(O)-, or -C(0)O-, and one or two additional methylene units of L are optionally and independently replaced by cyclopropylene, -0-, -N(R)-, or -C(0)-; and Y is hydrogen or CJ-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(0)-, and one or two additional methylene units of L are optionally and independently replaced by cyclopropylene, -0-, -N(R)-, or -C(0)-; 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(0)-; and Y
is hydrogen or C1_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 C1_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-, -NRS02CH-CH-, -NRS02CH-CHCH2-, -NRC(O)(C-N2)-, -NRC(O)(C-N2)C(O)-, -NRC(O)CH-CHCH2N(CH3)-, -NRS02CH-CH-, -NRS02CH-CHCH2-, -NRC(O)CH-CHCH2O-, -NRC(O)C(-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, N02, or CN; or (g) L is -NHC(O)CH-CH-, -NHC(O)CH-CHCH2N(CH3)-, -NHC(O)CH-CHCH2O-1 -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-, -NHS02CH=CHCH2-, -NHC(O)CH=CHCH2O-, -NHC(O)C(=CH2)CH2-1 -CH2NHC(O)-, -CH2NHC(O)CH=CH-, -CH2CH2NHC(O)-, or -CH2NHC(O)cyclopropylene-; and Y is hydrogen or C1_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN; or (h) L is a bivalent Cz_g 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(0)-, -NRC(O)-, -C(O)NR-, -N(R)S02-, -SO2N(R)-, -S-, -S(0)-, -SO2-, -OC(O)-, or -C(0)0-, and one or two additional methylene units of L are optionally and independently replaced by cyclopropylene, -0-, -N(R)-, or -C(0)-; 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)S02-, -SO2N(R)-, -S-, -S(0)-, -SO2-, -OC(O)-, or -C(0)O-, and Y is hydrogen or CI-6 aliphatic optionally substituted with oxo, halogen, NO2, or CN; or (j) 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(-0)C=C-; and Y is hydrogen or C1_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)S02-, -SO2N(R)-, -S-, -S(0)-, -SO2-, -OC(O)-, or -C(0)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;
(ii) C2_6 alkenyl optionally substituted with oxo, halogen, NO2, or CN; or (iii) C2_6 alkynyl optionally substituted with oxo, halogen, NO2, or CN; or (iv) 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 (Re)1-2 (Re)1-2 \'N
~^-N-Q-Z `(\~NR

1-21-2 1-2 e (vi) , , or , wherein each R, Q, Z, and R and R 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) (Re)1-2, 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 A'O KNR N
PL- 1-2 I12 \I~/12 (xaa) (Re)1-2 (Re)1-2 or (Re)1-2 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 N SS lI N1 c ICI ~1 (R')1-4 c f~l ~~ (Re)1-4 ( (Re)1-3 ~ (Re)1-3 C \ % ~Re)1 3 (xiv) cS-\% 'O N Ild 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 R

N
N N R N
(Re)1 3-,-(R-)1-2 U~(Re)1 2 NRe (xvi) N N N`
N
\ \\, (Re)1_3 \ l \ N
-Re (Re)1-2 (Re)1N -2 N--D' N
e L / (Re)1-3 ~N (Re)1-2 J\(Re)1-2 N~ R

S/1 \\S// S\N S
`\N e /\(Re)1-3 N (Re)1-2 i_(Re)12 ND R

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;
(m) L is -C(O)- and Y is selected from:
(i) C1-6 alkyl substituted with oxo, halogen, N02, or CN; or (ii) C2-6 alkenyl optionally substituted with oxo, halogen, NO2, or CN; or (iii) C2-6 alkynyl optionally substituted with oxo, halogen, NO2, or CN; or (iv) 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 (Re)1-2 (Re)1-2 ~^N~Q-Z `(\~NR
\~N`7 VC4~1-2 1-2 1-2 e (vi) , , or , wherein each R, Q, Z, and R' 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) (Re), -2 wherein each Reis 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 A'O ANR N
~ (Re)1-2 1-2 1-2 1-2 (xaa) G (Re)1-2 (Re)1-2 or (Re)1-2 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 (Re)1 s (Re)1-4 f~l ~1 (Re)1-4 ~ (Re) -3 N -(Re)j-3 (xiv) N
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 R R R R
N N N\ NN N
(Re)1-3 (Re)1 z c!(Re)l2 -R
(xvi) N N N` N NON
('(Re)i3 \ N (Re)1-2 \ 3/~-(Re)1 2 N Re O /O\ /OW /OW
N N
% ) (Re)1-3 \`N (Re)1-2 (Re)1-2 `N Re (S` \\S// \\S\N S~
lJ>~(Re)1-3 LN (Re)1-2 i _(Re) 1-2 N~ Re 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;
(n) L is -N(R)C(O)- and Y is selected from:
(i) CI-6 alkyl substituted with oxo, halogen, NO2, or CN; or (ii) C2-6 alkenyl optionally substituted with oxo, halogen, NO2, or CN; or (iii) C2-6 alkynyl optionally substituted with oxo, halogen, NO2, or CN; or (iv) 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 (Re)1-2 (Re)1-2 ~(N-Q-Z ~(\~NR \'`N

1.21-2 Q1-2 e (vi) , or , wherein each R, Q, Z, and R' 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) (R
e)1-z wherein each Reis 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 N )~,' A'O A'NR N
(Re)1-2 1-2 `I \ 1-2 (xaa) (Re)1-2 (Re)1-2 or (Re)1-2 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 N\ N1 (Re)1-4 fly /JI (Re)1-4 fl` '-(Re)1-3 I N (Re)1-3 -(R')1-3 (xiv) v v v N v N
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 N
N N R N
~~~ (RI),-3 N (Re)1 2q (Re)1 2 -NRe (xvi) N N N N
IN
N \` e \ (Re)1-3 (..)(Re)12 N (Re)1-2 N R
N
4(Re)13 ~N (Re)1-2 ``J\(Re)1-2 Re S~ S\ S S
\N )1_3 \~ N (Re)1-2 i(Re)12 N-3-Re 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;
(o) L is a bivalent CI-8 saturated or unsaturated, straight or branched, hydrocarbon chain; and Y is selected from:
(i) C1-6 alkyl substituted with oxo, halogen, NO2, or CN;
(ii) C2-6 alkenyl optionally substituted with oxo, halogen, NO2, or CN; or (iii) C2-6 alkynyl optionally substituted with oxo, halogen, N02, or CN; or (iv) 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 (Re)1-2 (Re)1-2 ~(N-Q-Z ~(\~NR \'`N

1.21-2 Q1-2 e (vi) , or , wherein each R, Q, Z, and R' 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) (R
e)1-z wherein each Reis 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 N )~,' A'O A'NR N
(Re)1-2 1-2 `I \ 1-2 (xaa) (Re)1-2 (Re)1-2 or (Re)1-2 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 N\ N1 (Re)1-4 fly /JI (Re)1-4 fl` '-(Re)1-3 I N (Re)1-3 -(R')1-3 (xiv) v v v N v N
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 R

N
N N R N
)1 2 -NRe ~~~ (RI),-3 clR2q (Re (xvi) N N N N
IN
N \` e \ (Re)1-3 (..)(Re)12 N (Re)1-2 N R
N
4(Re)13 ~N (Re)1-2 ``J\(Re)1-2 Re S~ S\ S S
\N )1_3 \~ N (Re)1-2 i(Re)12 N-3-Re 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(0)-, -CH2NH-, -NHCH2-, -NHC(O)-, -NHC(O)CH2OC(O)-, -CH2NHC(O)-, -NHSO2-, -NHSO2CH2-, -NHC(O)CH2OC(O)-, or -SO2NH-; and Y is selected from:
(i) C1-6 alkyl substituted with oxo, halogen, NO2, or CN; or (ii) C2-6 alkenyl optionally substituted with oxo, halogen, NO2, or CN; or (iii) C2-6 alkynyl optionally substituted with oxo, halogen, NO2, or CN; or (iv) 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 (Re)1-2 (Re)1-2 ~(N-Q-Z ~(\~NR \'`N

1.21-2 Q1-2 e (vi) , or , wherein each R, Q, Z, and R' 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) (R
e)1-z wherein each Reis 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 N )~,' A'O A'NR N
(Re)1-2 1-2 `I \ 1-2 (xaa) (Re)1-2 (Re)1-2 or (Re)1-2 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 N\ N1 (Re)1-4 fly /JI (Re)1-4 fl` '-(Re)1-3 I N (Re)1-3 -(R')1-3 (xiv) v v v N v N
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 R

N
N N R N
~~~ (RI),-3 N (Re)1 2q (Re)1 2 N-NRe (xvi) J1lL
N N N N (N11 e \ \(Re)1-3 (..)(Re)12 N (Re)1-2 N R

/OW ( N .
N
(Re)1 3 ~N (Re)1-2 ``J\(Re)1-2 Re S~ \\ S\ S S
\N
~/~(Re)1_3 \~ N (Re)1-2 \j/\(Re)1-2 N-3-Re 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.
[00108] In certain embodiments, the Y group of formula la 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.
Table 3. Exemplary Y groups:

5-N I ~~ I ~-N I -N I ~-N CI 5-N I CH3 a b c d e f O CI
I\
I \ S' CH \

g h i j k l N
V ~:z'CH CN N CN 0 4, 0 S - C\( N N
CN
m n o p q r F

CN
F F I\ F I F F I\ F ol~N

F / / N02 CN / s t u v w x y sI % 3 I \ s' I \ s'~
N
~ % \ sY
N
N
N N N NI IN INI /
z as bb cc dd ee I N N N N ~N NON N

II
if sgg hh kks nC, Sn Re N / N INI IN INI IN INI
Re N ~/ \/
Re Re Re Re ll mm nn oo pp qq H N Me J:p~= / N~ 0 g ~Re HNRe 4N

Re Re rr ss It uu vv M
0\ N
N e MeN N O
/> - Re ~\ Re I / N /Re Re Re ww xx yy zz aaa OWN qN /Re I ~Re ~~' Re 'IN
Re bbb ccc ddd eee fff Me N N HN' N N\ I )_/
N I / \ /N
/ ~h N
N '? Me L /

ggg hhh iii /11 kkk Me IMeN' OW
N I / I
111 11111 nnn 000 ppp N S-~ o,N
h >-\

ggg rrr sss ttl 111111 N
H e N\N N N N HN' / ~N~ /

\\ \\
vvv qqq www xxx yyy Me /~ Me IN_' -- 0~
/ N
N
o zzz aaaa bbbb cccc dddd 0 Nq S.~ N N

N 0 ;2t S

eeee ffff gggg hhhh iiii N CN~ I S
/N CN-c kkkk 1111 mmmm SS nnnn N
N~ Re I I N
N ~ .Lj N" \\ L
0000 pppp qqqq rrrr ssss XO

tttt uuuu vvvv wwww xxxx 0 00 0~ Me 0 J Re N
Me yyyy zzzz aaaaa bbbbb ccccc ddddd wherein each Re is independently a suitable leaving group, NO2, CN, or oxo.
[001091 In certain embodiments, RI 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(-0)C=CH. In some embodiments, R1 is selected from -NHC(O)CH-CHz, -NHC(O)CH-CHCH2N(CH3)2, or -CH2NHC(O)CH-CH2.
[001101 In certain embodiments, R' 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.
Table 4: Exemplary R1 Groups 0 H Me I

~~z^H~N N N
V O O

a b c d 0 0 0 0 Me H H H
e f g h i 0 0 Me 0 0 0 ,\'~'N'ICI N N\ ^ NCI a~N~CI NCI
H 0\ H H H
j k l m n o ~J
N ~ X-- IN Me v H ICF3'~/~%

p q r s t IN, Me xlj,~ -1,1~

n v w x y Et 0 z as bb cc dd ee N
~
N IN N IN NON N /
if gg hh ii jj kk -I- rj~ Y, N IN IN IN ,N NON N

II
ll mm nn oo pp qq s' I \ ? I \ s' I \ s'- Re N
N N INI IN N N INI
Re N Y
Re Re Re Re rr ss It uu vv ww H N
NN 0 \>-Re HN N Re Re H

xx yy zz aaa Me Me N N
\ I % N {,)_Re \) Re MeN' Re N N
Re Me bbb ccc ddd eee 0 0 N\~ N
N />-Re / Re Re `s N .`~. 0 Re fff ggg hhh iii s S N S- N
%N {,)_Re \ -Re Re :+z N .~, S
Re jjj kkk 111 mmm H H Me N N HN~N McN~N / N
II ,~ ~~ \ \~ I N
N

nnn 000 ppp qqq 0 O N ,N [N,)/
c L

rrr sss ttt uuu vvv S` S fN SAN ao iN
N , www xxx yyy zzz aaaa H Ne jN MeN' N N
L ~ j />

bbbb cccc dddd eeee ffff ,[O)AN
0 ~Z?~O N L-=- N i>-N J0> ool \\

gggg hhhh iiii kkkk I sN 9IN NN
~N
o' S
\\ \\
liii mmmm nnnn 0000 pppp N
N I N'N
I_ I F, Cl, Br v~ \\ N

qqqq rrrr ssss tttt uuuu 0 O\ 0 O F O F O F
,`'~% I~,~N zzN I /N \ H \ H -~6 vvvv wwww xxxx yyyy zzzz aaaaa bbbbb 0~ VNR",,, c N
ccccc ddddd eeeee fffff ggggg hhhhh iiiii N.
CH3 +- N CH3 + N CH3 CH3 CH2CH3 CH2CH=CH2 jjjjj kkkkk lllll mmmmm nnnnn 00000 ppppp qqqqq ~~ 0 0 CH3 (.~ o CH3 - ANN A N)\% N)\%~N~

rrrrr sssss ttttt uuuuu o' 0 0 0 S~0 O

vvvvv wwwww xxxxx yyyyy zzzzz aaaaaa bbbbbb 0~0 0 CH3 0 NS;:::--NH' 'I' v CH3 ;4~Ac 101 CH3 cccccc dddddd eeeeee ffffff gggggg hhhhhh _N 0 0 0 CH3 0 OH N

kkkkkk 111111 mmmmmm nnnnnn 0 OH O 0 OH N\YF
OEt OEt O OEt CN I N

000000 pppppp qqqqqq rrrrrr ssssss 0 O~~ F
-IN N -0, 0 \ O 0 H S~F~
tttttt uuuuuu vvvvvv wwwwww xxxxxx or yyyyyy wherein each Re is independently a suitable leaving group, NO2, CN, or oxo.
[00111] As defined generally above, R' is a warhead group, or, when R' 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 R' 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 ErbBI, 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:

[00112] Thus, in some embodiments, R' 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 ErbB 1, Cys805 of ErbB2 and Cys803 of ErbB4, or a mutant thereof, where the provided residue numbering is in accordance with Uniprot (code P00533 for ErbBl; code P04626 for ErbB2, and Q15303 for ErbB4). It will be understood that the Cys of ErbB 1 (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 ErbB 1 may be described as Cys 773 or Cys 797 and these terms are used interchangeably.
[00113] 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 R' groups include, but are not limited to, those described herein and depicted in Table 4, infra.
[00114] As depicted in formulae I-a and I-b supra, the R' warhead group can be in an ortho-, meta-, or para-position. In certain embodiments, the R' warhead group is in a meta-position of the phenyl ring relative to the rest of the molecule.
[00115] In certain embodiments, R' 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.

[001161 In certain embodiments, R' 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.
[001171 In certain embodiments, R' 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.
[001181 In certain embodiments, R' 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.
[001191 In certain embodiments, R' 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.
[001201 In certain embodiments, R' 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.
[001211 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 R' groups include, but are not limited to, those described herein and depicted in Table 3, infra.
[001221 Exemplary compounds of the present invention are set forth in Table 5 below.
Table 5. Exemplary Compounds a NH HN NH HN NH H N
F
NN NN NN
H H H

HN I \

/ I0J I aNH 0J F N 0 /~ NH HN N
NH HN HN N NH
F

N!N \ I I N!N \ I b'Nk~~
H H H

NH Br / ,a NH 0 b,NH \ NH \ NH HN / HN"

IAN /I N I~
N H N H NIN H N

0 7 0S-MNH CI~N ;NH2 N N\I
,NH2 O,S NS
H OS
NIN\I
H H

\ 0 / 0 0 /

NH HN" v 7 N \ NH

I \ I NJ.N \

N H H CI

~I
HN H) HN H HN H
CI
CI N
~I XNO
N CI H H H

~1 0 \I ~I 0 HN N
HN N H N N H
`N , N
oMe ~~ \ IA n N N j/
N H N H N H

\ I 0 N \ I N
N H H HN H
N N\ N N-\ N N\
H H H

NH HN HN

N N N N
H H

HNN

N
HN \ I 0 I H N \ 1 N" v i N H H H \I

~I ~I
N
HN H S H
F

N N N N
H H

HN H I/ I
NH
N
F
I
N N N~ \ I\%
~N"~OH H H

\I 0 IoI /' H N
N \%
N\ NH H N H

I _ N \I S'NH2 ! \I
\I I N
N H H H Ol0 N H

O O

HN \ N / H N \ N / H N \ N
H N~
HO
I\~ /I ~ /I N /I
H\ 0^` N H\ N H\

H r ~J
N N
HN ~
\IH HN \I 0 HN

~~ \ I \ I N~`N \ I S.NH2 N N N N H

Ni O

0 Z'~ HN
HN HN Q--H

/ F3C N N F / N 0, N!N \ S,NH2 1 \ 1 1 / C
H ~ N N O N N
O O H H

HN HN N HN
H
N
F3c I J,, N 'N Cz ~ \ F
1 0~0 N N O N N 0 N O j,~
N N
H H H H

HN HN HN

NANH/I NINH/I NN N '/I
\

0\ 0 0\ 1 H

I~~N\ /I N N/I
\
N N\ N
H H H

HN I N
FN F

N~NH F F HN H HN H
/ I\ 0'' 0 F I ~ / I CI
b,, 0 F
.N
N NNN NN N N

HN" v NH \ I \
NH NH
~N /
NON \ I \ I F

H N NH 'o NH O F

NH

\ NH NH NH

\~ N I \I I \I ~F
N' N H N N H N N H 0 F

0 0 NH v - N~l / I I~ \ I ~NH\ O~
N NH O"' N
N- NH v _O---~N-, NH v p NH v v NN, b'o NH 6LO
N 5p, N N H O`\ Q N N H O"\
N NH

N~
H HN H N NH N p O F Z, N N N / F HN I ! *,"co I N
J. ' ' ' N H N H
N NH H H

N

HO

HN HN H Sr' ~HN H
F I ~ / I 0 F I B NJ O F I ~ / I off NNN N N N NNja N
H H H

\ I O / F HN H
O ri%
QN7 0 H I N _ H F N N! N
HN
H
F/ N I\ Cl N!H a 0 O
N H O N

' L
HN N
H
N! NH

,I ~ N' ~
N N
HN H HN H

N F \ / 0 F 1 f 1 N N

F ~
aN 0 F IHN H
HN H 0 NNH ~N
F H 0 N~N"
N N H
wN
N N H

HN I
N
F H

N \I N" N NH N-HN H O H
F -I i F I ~N i N IN I NlN I 0 H O H N--) "OH OH

HN
H H N N HN I
F H HN N N
H
F F N
N NH NNH N! NH
qoH

NI o /0 o\

O
a 0 ~~ HN H
HN / I N' ~%\ F N

F ~~
/ N H ON NH
NNH /

O N' HN H
O F
I , O N
N I I I H

O
HN I N'v 0 H
F HN H
N, NH
F I N HN \ N

NNH

f 0 HO o o ~0 HN" ~

~
HN\ IN NH HN\ IN
H H
F\ N ODD F/ N / OMe F N O OAS
N N"~ N N F N N~
H D H H

OH

\ NH HN \ NH HN" v v N
F I~ /~ N' F I\N /~
N'N \ N"N \
H H

0 N~ / 0 \ I NH HN N ~f\% N, NH HN/ ~NH
F I ~I \ F !\
N N N N
H H

NH HN H N N
F I ~ F - _ 0 NN'N/ \ NN \N \~0' H H

OH
O
~~ NO N
NH HN HN H
F - _ N F N
N INN/ O
I \ i~OH
H H

Cl p CI

/I H
HN \ H F I ~N
F N NH

Ni \ NH N I
F N \ N /0 p J( N H O

\/~ HN H
HN
N~ H F
/ O J
\ I N!N \ I O Nom/

F O HN aN)t' /
H II
N Jao F / 0 HN
NIH 0 \ ) F N /
\/\ NNH H J~ \
N N O

\ I I \ N I O\ I N~
H
HN I HN F

F O F I H

I \ N I ~~ \ I I NN \ I O

N'\% O / 0 NH
/~/ N \ I N
O HN H NH HN
F I ~~ \ N 0 F I , \ I OH F \
N H N H O N!H

,,,a NH \ I N _/~ l 0 H N \ N
NH HN O H

F INN/ N / ~ O~~OH
\ I N~
N N
N
H H

0 HN N \ IN-/
H NH H N ~ 0 N
N; N O I \
N N N
H H

O

H HN N H
H
o - y F N F ANZ

H H H

~ I ) / o OHN N J
N 0 HN/\ ~N

N~NH N O' O N~NH
N llaW~ H N F N I ~N

H N
H N 0 fo HN~ 0 HNI

FN N ~ O O~OMe F I \ I i N N \ I N N \ I N'I

H H H

0 / 0 ,ON
~'N I ' HN
~~N
HN H O' HN H F N 0 F
N
INNN \ I 0' , \ I / N H \
H O N H O O

N
\I I ja o F %,,CN HN N
N /I s H
/
J~ \ F 0 FN
N H O 0 I N ~ ~ ~
v S~ NlN \ l N H 0~\H
0 O' 0 HN N
~ O 0 F I I N 0 HN" `/ HNC NNH
~\Oj NH NH OAS, N / 0 F N N~ 0 OMe 0 J( N H N H~~ O

~ ~ ~ HN N

N
N 0' v / 0 HN \ H ' N N \ F
N H
HN 0 N ~

N N F OF N H 0-N/~N

N
I N
~N 0 \ HN
HN H

F \ N ' F I ~ \ I O F I iN 0 OH N N O N N \
N H H ~~ H CI

I
O HN\N
N!NH

N/ '~I I
O ~~/\
H 0= CI NH 0 N") F , , F 0 O
Ni \ N \ 0 I IlJI, 0~
N H
H O I

\I \I
F HN H N 0 HN N` 0 HN N

N N NH
~ I O %N
N N N N N N /N

\I ) \I \I

0 "~ I ~~ \ I NN I\ NN
N I N Z
H\ /N N H N N

\I \I
N H N H N H H aN 0 0 N H 0 I \N I ~ IN / I 0 I \ N
N NC N N' I 0 N N
H H H

OH
/ O ~N O

N
F HN \ H H N \ F O N H

'IN \ I N"H ON
N_ Na S~0 1 H 'NHZ N H

1-160 1-161 1-162 N 1 I \

j1 F
O
HN HN JaNlkv NINH
H
F F \ 0 N
F \ /
N N O F N N 0l-,,,)ND 0 H H

~I
HN I,N HN NH
F ,I
~N r 'O I N O' / O
NNH NNH II \

0 C\0 N N, 1 1 N ~
N!N N
O~ O~1 H

\1 \I \1 O HN N 0 HN N~ O N N

N~ I O~ N N
H
N N N CN N N
H H H

HN \ IN
F H

N ~ I N\ HN H HN H
/
/ O0 I \ I J
NINN N NI N N
H H H

0 0 N" v \ I k N N N N
H
HN N NNH H
N j p N) NH

N N N N- b,o H
0~0 i0 I

/ I p N

H Y~l F O
H2N F ~N O I N
NINH NNH NNH

\ I N
O \ IO
I I ~0 \ I

F IN H
HN \ I N' N
N i~l NH
F H
N! NH / 0 / HN N

0NH s I N_ \ IO AOk ~ H H

~I ~I

H H
F /

N N
)a Y -N N N N N\
H H H

IN OMe N j OMe N j OMe ! !
NIN N N N N
H H H

N N

fJ 0 ~
0 O HN \N
F H
N / I F

\ N I 1 N
H ~ N H N H

' 0 \I
p HN N p~
N F I' 0-;I N
O~,. N NH HNIO
F , N I F \N ' H O~ H

HN HN HN / N H
O-/
F F 0 F H 0,_,,~~I
\ N lI
NI / i IN N
\ I N I \ I 0 H H H

p p \ / \ U
HN 0 0~a H) HN
F
F
a a F H N
N \l ~

N No O F H NH

~0 OI pI HN" v N N /
HN
HN HN
F N / O F N O F I \N ' \ N N N N O N--\
N H N H H I N

0 , 0 N I \ I \
HN 0,, NH NH

\ 0 F \ I F i~ \ I N
N N N N N l H H H

\ I I 0 H2N N \ H
\ I H2N I N
HN N NH NNH
F I N

N H\ O^~N N b"O

it I\ 0 O

NH O HN F HN
F
N F IN F IN
tj \ I i N 'l H N H NIH O

i oo ~HN N

N
N NO"""-"-\NH
H

H
N H
\ IO 0 NH J,~

F H N I N F I N0 Oi H

H

N N O\ F N 0\ F
H N J\ N O
N N N N N NN
H H H

H
I o I 0 H IN H
N N N N N N
H H H

\ I ~% \ I HN

H N F- H \ I \ F XNNH2 N H

H ~0 0 CN ^ HN"

F HN F HN F HN
F F F
F I I F I \ i F I i~ \ I i N N O N IN O N N O
H H H

I0I ~JN 0 H H
F N F I \N / H2N N /
.~ / J. C
N N 'O N LNJ~N
H H H

/ I NH
HN

\ / 0 N NH F

H I \ I N
N H 1"N I-231 1-232 1-233 HN" 0 H
HN F FF FHN
F FHN

Z F N / F I N /
N N \ N N!N \ O~ N~N \ O~
H H H

HN N" -H HN~
N'N O"~\NH
H HN

HHN NH NIH N

\ NH HN" v v N--~~NH
F I I'b 0 N N H

H
H,HN 0 NH
S ~H

~0 0 \ 0 / HN"
HN H v H \ F I ~N \
NH F
NNH FHN

N H O N H

/ HN
II HN \
HN \ ~N F F FHN
F I \I / 0 N!N \ I O F N n/
N H F '/
N H FF N H

HN)t"~
Z~ANH
HN \ I 0 \\/ HN \ I I/ O
I I
~ 0 F F

N N N N N N
H H H

NH

\ )~ II o NH2 b,, HN H HN H NH
F N CI OHF I SIN / O NI Na F/F
NJIH N H F N a O F

HN H HN" v IO
HN \ I HN b,o N~
F % N \ N ~ % IN F I %N /
J~ ! \
N H Nl H N H 0 \\

0 \ 0 0 FFHN FFHN FFHN
F F N F N
I \ I i I \/ I \ I i H H H

HN' HNC -CI O
\ \ N
F HN F HN HN H
F FI INH N NO
F FI Nl IN H N O F Nl IN H " N
NJ~ i i \ N
H

O
1 N~, ,/ 0 /~ 0 H ja ~\ O ~
HN H O H O H
F NN 0 ao ~0 N / I 0~
\ N
N N N N NN
H H H

N Ql--H N N

N N
HzN I N \ I HzN I / I 0 H \
N N N N
H N H H

HN IIII n N O
O N~ N
O O H F HN O
Hy N \ N I F I \ N O F N IN O N N O \ H N H H H

O I
N / O
p p \ H I/ \I

f-~ F F F 0 ! IAN /I
N
N !
N H\Ip H O, .NH2 N H\ phi / N'~ o/I N~

O HN
F F N / F N
NH \ Sr JI N N ;
N N N /

0III '' HN HN /N
\%
F,_ I --N
/ I F IIIN \ p`~`O~
N N \ S'o I /
H ~~ 'NH N N O~~O~

N H IN JaN
H
HN H F
F O N /
\
N~N
NN \ O~
H " N

O
N /~
HN
' N
. ~
O HN \ H
F
NI, F N
/ O IN F N
_G N, 0 H N
N -Q
H CI N H

CN
HN" / I O

. HN \ N
H
N I N \ I 0 F / N
H ON \ I N
N N
H

N I

F O 0 aN
N
0 H O Hp~

O
NH \ / O F 'N O' F
pN N ' / NIN \ / \
H H O__ N aN 0 0~ N

N~H F ~ \ I N~ F I N / N
O~~N J N H H H

O
O

HN \ N NH2 HN H
F
F \N / N 1 0 -Ira N" H 0 N N N
H H
)"'~NH2 O O

HN H F
F N / I O N N N \ I O
N N H

N ~ N~ N Br ~-N H N H N H N H
F \N 0 F 1001\/~ F i NN \ I NN \ I N \ I ^p H CI H CI H O',I~/
N

N N

F IN i F ,I
N~N l I N!N
H N H O'er N

N

F ( ~~N/ I

N N" ~/\
H O~~) N H N~/

/ I ; 0 F
\N I HN
N H N F I

~,N N~N N I
"OH H N

l'u N

HN N
F \ \ N-HN N O
NIN I F 0~
H O I N
N N H N

OH aNH 0 NH
)I~%, iNN HN
NH HN \%\~ F
F I ~ / I N
N N
NIN \ H
H

ci 0 NH
HN HN N
F I N 0_ F N
N~N O
\~---~ NHZ N N
H V H

II II
HO HO
\ I )\, \ N IN
HN N HN
F N Ja O~~Oi F I \N / I O F , _ / Ov OH
N N ! \ N
NN H i N 'N

I N I N I N
HN H HN H HN H
F , _ F , , F , N N N N N N
H H H

~ N HN H N
HN H F 0,,, Co F HN H 0 N S~
N O I F O, O 0 F LI
N N
N N F H NIN F
H H

N' " HN H
HN H F N
F - _ q-OH H F
~/ ` N H
N H H

r1l a 0 N
HN H HN HN
Cl F N F N O

N N N N F NNN F
0 1 ' H H H

N

HN HN HN
F N / O~~Oi F N 0-,~ i F 0 N N\ I F N N\ F I N IN C I F
H H H

cro HN HN HN
F I N JX i F N O~/~Oi F I 4xo ~Nll N N F Nl N F NIN F
H H H

HN HN HN H

F I N I F N 0~~0~ F I / OOH
N N F Nl N F NIN F
H H H

O-N N OH sDLN'L"

NIJ
51, N, I F N 0 OH F N Ov _OH F N Ov OH -'a N N F N N F N N
H H H

O-N
HN N O~ ~aNH HN
H HN H \%
F CI F N CI F N
~OH \ ~OH
NNN O N N 0 N N \ I
H H H

HN \ H N~ N
F HN H HN H
N \ I F CI F N CI
~0 I \ I C0 NJ`H O OH
NY' N 0 N N 0~
OH H H

CN
o N

CI NH Orl~ N") HN
F I H N / I O I\ H FI N / O F N 0 ~. ,N \I I \~
N N N N N N F
H H H

HN\IN
0 F , , H 0 N~NH
N N
HN HN
O I
F I N / I O~~Oi F
N N NNN/ I O,, H N H

'k HN H
0 N\\
0 HN \ N
H NNH
~
N I
NINH I
HN F
prp O
FN
0 p~
N H / I ~0 I

o F F HN N
H
F N

H N)'-NH / 0 / N\\
U HN H
HN F F / OCI

Ik \ I OH
F f 11, H OH

1 \
OH
N N
HN H HN H
F CI F N
IN / -'co I F H
N IN N N
H H

v HN H
HN \ H ' H
F N / 0"-- O-\iN0,/ F N / CI
\ 0 I \ ~0H
N N F N N O
H H

O
N
HN H H N N H

CI F N N N NN F
F L N Z(O"-~OH
H H

HN \ ~ HN H

0 I I N I ~\NH
F
N /~/
N H H N H =0H

/ o / o HN \ I N~ H HN \ I N
H
F _j \ I N 0 F iõ N 0 N N F N N
H H

NH NH
HN H HNf _ H
0 N ro 0 Nr0 S NH S NH

H N

/ 0 4c 0 HN \ I0 F L / H
N
F / ON 0 I N N 0/\O~i N 0 H
NIN
H

NH NH
HIIN H HN H
0 Nr0 0 Nr0 S NH S NH

F HN \ I N H N\ HN H H
F / 0~-0~~ N 0 / ON 0 NIN \ I N~N \
F F
H H

NH NH
HN H HN H
N'rO O N,~O
O
S NH S NH

5~
H
F C N n~0 e 11N~ N-NH

NH

HN H
O NrO
S NH

HN H
HN N
F N / Cl F H ON" ~O,~NUO
N l O ~r N'N" v 0 N
HN N HN H
H
F N / ON3 F N / I O(OH
J, ZZ-1 N N N N F
H H

N~ NXl///

F N / 0 OH F I N / I O~O CH2 N N F N N F
H H

O
\ I N I~
HN H HN H N
CI

N N NN N
H H

O

N N F N N F
H H

' N "'o NH HN- v HN
H
F N / Na F I I N /
N N N N
H H

/ 0 0~
N' N I iN\
F I IN\ I F N
H H

~0 0 HN" `~ HN" v N N

b,'NH \ NH N Nom/
CI / N F /
H \ I N
NI N!N \ I
H i0 H /O

O

N~ INS

0 ll,NH NJ NH N
CI NCr CI N
NIN / \ I NIN / \

H i0 H i N

HN HN
F N I'aoF' N N F N N H H

N--O

HN HN
F I
/ I O~/~Oi F N
O

NNN F N N
H H

HN/'~
N
O

/ 1 / \ o HN HN
F I N
J O-/~O F N / O

N N N N
H H

CN
N
H
O

HN
F I NN Otro N IN

[00123] In certain embodiments, the present invention provides any compound depicted in Table 5, above, or a pharmaceutically acceptable salt thereof.

[00124] In certain embodiments, the present invention provides a compound selected from:

~IH HN~I
HN
/ F , \ I )\, \ N
N N N N N
J!a0 H H H

HN N
F H
O
HN" N,NH O
N
O
NH
F?N"' N ao\---\OMe O F
Of \
H F NIH O

O
/ I 11 Na N
HN H H' N O
F I C' OH F H O \
p I \ I I ~N H
N H NIH

or a pharmaceutically acceptable salt thereof.
[001251 As described herein, compounds of the present invention are irreversible inhibitors of at least one of ErbB 1, 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 ErbB 1, 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.

v `N

N
I eo or a pharmaceutically acceptable salt thereof.
4. Uses, Formulation and Administration Pharmaceutically Acceptable Compositions [001261 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.
[001271 The term "patient", as used herein, means an animal, preferably a mammal, and most preferably a human.
[001281 The term "pharmaceutically acceptable carrier, adjuvant, or vehicle"
refers to a non-toxic 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.
[001291 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.
[001301 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.
[001311 Compositions of the present invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
The term "parenteral" as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques. Preferably, the compositions are administered orally, intraperitoneally or intravenously. Sterile injectable forms of the compositions of this invention may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1,3-butanediol.
Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium.
[001321 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.
[001331 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.
[001341 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.
[001351 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.
[001361 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.
[001371 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.
[001381 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.
[001391 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.
[001401 Most preferably, pharmaceutically acceptable compositions of this invention are formulated for oral administration.
[001411 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.
[001421 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 [001431 Compounds and compositions described herein are generally useful for the inhibition of protein kinase activity of one or more enzymes.
[001441 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.
[001451 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.
[001461 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.
[001471 Examples of kinases that are inhibited by the compounds and compositions described herein and against which the methods described herein are useful include ErbB
1, ErbB2, ErbB3, ErbB4, a TEC-kinase (including BTK, ITK, TEC, BMX and RLK), and/or JAK3, or a mutant thereof.
[001481 The activity of 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, 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 ErbB 1, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3. Inhibitor binding may be measured by radiolabeling the inhibitor prior to binding, isolating the inhibitor/ErbB1, 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.

[001491 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 [001501 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 ErbBI
(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, TGFa, AR, BTC, EPR, HB-EGF, NRG-l, 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 (P13K) 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).
[001511 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, SOS I, Ras, Rafl, Mek, ERK1, ERK2, ERa, Akt, mTOR, FKHR, p27, Cyclin Dl, FasL, GSK-3, Bad, and STAT3.
[001521 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 All, 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). ErbB l 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 18: 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.
[001531 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, I.L., et al., J
Clin Oncol 16: 1340-9, 1998).
[001541 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).
[001551 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 vlll) 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.
[00156] 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., Mol Cancer Ther 3: 1335-42, 2004).
[00157] 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.
[00158] 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 ErbB 1, ErbB2, ErbB3, and ErbB4, or a mutant thereof, and are therefore pan-ErbB inhibitors.
[00159] 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).
[00160] 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, I.L., et al., J
Clin Oncol 16: 1340-9, 1998). Irreversible inhibitors have demonstrated superior antitumor activity in comparison with reversible inhibitors.
[001611 Neurofibromatosis type I (NFI) 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 NF I 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.
[001621 Abberant expression of the EGFR is associated with tumor development in NF 1 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 NF1 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).
[001631 Patients with NFI 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-l responses (ErbB2, ErbB3, ErbB4).
Neuregulin-1 (NRG-1) proteins promote the differentiation, survival, and/or proliferation of many cell types in the developing nervous system, and overexpression of NRG-1 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).
[001641 Deregulation of Schwann cell growth is a primary defect driving the development of both benign neurofibromas and MPNST in neurofibromatosis type I (NF 1) 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).
[001651 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.
[001661 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).
[001671 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. I., et al., Genes and Development 19: 2265-77, 2005; Curto, M. C., et al., J Cell Biol 177: 893-903, 2007).
[001681 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.
[001691 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 ErbB 1, ErbB2, ErbB3, and ErbB4. Thus, in certain embodiments, the present invention provides a method for treating an ErbB I -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.
[001701 As used herein, the terms "ErbB I -mediated", "ErbB2-mediated," "ErbB3-mediated,"
and/or "ErbB4-mediated" disorders or conditions as used herein means any disease or other deleterious condition in which one or more of ErbBI, 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 ErbB 1, 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.
[001711 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.
[001721 In certain embodiments, the present invention provides a method for treating or lessening the severity of neurofibromatosis type I (NF 1), neurofibromatosis type II (NF2) Schwann cell neoplasms (e.g. MPNST's), or Schwannomas.
(b) TECFamily [001731 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, Rlk and, are activated downstream of antigen receptor engagement in T cells and transmit signals to downstream effectors, including PLC-y. Combined deletion of Itk and Rlk 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.
[001741 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, zebrafish (Danio rerio), skate (Raja eglanteria), and sea urchin (Anthocidaris crassispina).
[001751 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.
[001761 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.
[001771 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.
[001781 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.
[001791 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.
[001801 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, eosinophiic 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.
[001811 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, 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. In certain embodiments, the diabetes is type I diabetes.
[001821 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.
[001831 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) Bruton's tyrosine kinase (BTK) [001841 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.
[001851 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-a production in macrophages, IgE receptor (Fc_epsilonRI) signaling in mast cells, inhibition of Fas/APO-1 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.

[001861 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 Immoral 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 Fe epsilon RI
cross-linking (Kawakami et al. Journal of Leukocyte Biology 65: 286-290).
[001871 BTK has been implicated in diabetes. BTK deficiency in non-obese diabetic mice dramatically protects against diabetes and improves B cell-related tolerance, as indicated by failure to generate autoantibodies to insulin (Kendall, et al. J. Immunol.
183: 6403-6412 (2009)).
Modulation of BTK and improvement of B cell-related tolerance can therefore be used in treatment of diabetes, particularly T cell-mediated autoimmune diabetes, e.g.
type I diabetes.
[001881 BTK is also implicated in various cancers. For example, BTK is upregulated in pancreatic cancer cells compared with normal pancreas cells, and BTK is also upregulated in chronic pancreatitis cells, which is sometimes a precursor to pancreatic cancer (Crnogorac-Jurcevic, et al. Gastroenterology 129: 1454-1463 (2005)). Due to the key role of BTK in regulation of B-cell development, activation, signaling, and survival, BTK is involved in many B
cell-related cancers.
[001891 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.
[001901 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.
[001911 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, systemic lupus erythematosus (SLE), vasculitis, idiopathic thrombocytopenic purpura (ITP), rheumatoid arthritis, psoriatic arthritis, osteoarthritis, Still's disease, juvenile arthritis, diabetes, myasthenia gravis, Hashimoto's thyroiditis, Ord's thyroiditis, Graves' disease, autoimmune thyroiditis, Sjogren's syndrome, multiple sclerosis, systemic sclerosis, Lyme neuroborreliosis, Guillain-Barre syndrome, acute disseminated encephalomyelitis, Addison's disease, opsoclonus-myoclonus syndrome, ankylosing spondylosis, antiphospholipid antibody syndrome, aplastic anemia, autoimmune hepatitis, autoimmune gastritis, pernicious anemia, 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, membranous glomerulonephropathy, endometriosis, interstitial cystitis, pemphigus vulgaris, bullous pemphigoid, 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). In certain embodiments, the diabetes is type I diabetes.
[001921 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.

[001931 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, atopic dermatitis, asthma, allergy, blepharitis, bronchiolitis, bronchitis, bursitis, cervicitis, cholangitis, cholecystitis, chronic graft rejection, colitis, conjunctivitis, Crohn's disease, cystitis, dacryoadenitis, dermatitis, dermatomyositis, encephalitis, endocarditis, endometritis, enteritis, enterocolitis, epicondylitis, epididymitis, fasciitis, fibrositis, gastritis, gastroenteritis, Henoch-Schonlein purpura, hepatitis, hidradenitis suppurativa, immunoglobulin A
nephropathy, interstitial lung disease, laryngitis, mastitis, meningitis, myelitis myocarditis, myositis, nephritis, oophoritis, orchitis, osteitis, otitis, pancreatitis, parotitis, pericarditis, peritonitis, pharyngitis, pleuritis, phlebitis, pneumonitis, pneumonia, polymyositis, proctitis, prostatitis, pyelonephritis, rhinitis, salpingitis, sinusitis, stomatitis, synovitis, tendonitis, tonsillitis, ulcerative colitis, uveitis, vaginitis, vasculitis, or vulvitis.
[001941 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. In certain embodiments, the cancer is colorectal cancer or pancreatic cancer.
[001951 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.
[001961 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.
[001971 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, 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. In certain embodiments, the diabetes is type I
diabetes.

[001981 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, diabetes (e.g. type I
diabetes), irritable bowel syndrome, Crohn's disease, lupus and renal transplant.
(d) ITK
[001991 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).
[002001 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).
[002011 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.

[002021 It has been well established that T cells play an important role in regulating the immune response (Powrie and Coffinan, 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.
[002031 Mast cells play a critical roll in asthma and allergic disorders by releasing pro-inflammatory mediators and cytokines. Antigen-mediated aggregation of Fc.epsilon.Rl, 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.
[002041 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 at, International Archives of Allergy and Immunology 129:
327-340 (2002)).
[00205] Splenocytes from RLK-/-mice secrete half the IL-2 produced by wild type animals in response to TCR engagement (Schaeffer et at, 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 at, Nature Immunology 2 (12): 1183-1188 (2001), Schaeffer et at, 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-1 are all reduced (Schaeffer et at, Science 284: 638-641 (1999), Schaeffer et at, Nature Immunology 2 (12): 1183-1188 (2001)).
[00206] 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.
[00207] 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.
[00208] 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.
[00209] 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, diabetes (e.g. 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.
[00210] 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 [002111 The Janus kinases (JAK) are a family of tyrosine kinases consisting of JAK1, 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, Mot. Med. 5 432-456 (1999) & Seidel, et al, Oncogene 19 : 2645-2656 (2000)].
[00212] JAK1, 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.
[00213] 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) ].
[00214] 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.
[00215] 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)].
[00216] 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)].
[00217] 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)].
[00218] 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, LC1 ; 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.

[002191 JAK3 has also been implicated in diabetes. A JAK3 inhibitor exhibited potent immunomodulatory activity and delayed the onset of diabetes in the NOD mouse model of autoimmune type 1 diabetes (Cetkovic-Cvrlje, et al. Clin. Immunol. 106: 213-225 (2003)).
[002201 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.
[002211 The term "JAK3-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, diabetes (e.g. type I
diabetes), 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.
[002221 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.
[002231 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.
[002241 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.
[002251 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.
[00226] 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.
[00227] 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.
[00228] 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.
[00229] 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, 0 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.
[002301 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.
[002311 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.
[002321 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.
[002331 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.
[002341 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 ErbB 1, 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.
[002351 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.
[002361 Inhibition of protein kinase, or a protein kinase selected from ErbB1, 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.
[002371 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.

[002381 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 ErbB 1, 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.
[002391 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."
[002401 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 GleevecTM, among others. In other embodiments, a compound of the present invention is administered in combination with a biologic agent, such as Avastin or VECTIBIX.
[002411 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, fulvestrant, 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.
[002421 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-1 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, anti-convulsants, 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, anti-leukemic agents, and growth factors; and agents for treating immunodeficiency disorders such as gamma globulin.
[002431 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.
[002441 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.
[002451 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.
[002461 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.
[002471 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 pg/kg body weight/day of the additional therapeutic agent can be administered.
[002481 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.
[002491 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 [002501 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.
[002511 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, Rr, 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. One of ordinary skill in the art will appreciate that when a tethering moiety is attached to R', R' is a bivalent warhead group denoted as Rif. 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:

( ")p R1, T-R' (R"p A

y R1, T-Rt y N /
2 B (RX)m ~NW2&(RX)m N
V-a V-b (R")p R1 T-Rt A (R"p A

y R1 Y T-Rt N INI
B (Rx)m B (RX)m VI-a VI-b ( ")p R1 T-Rt (R" A

Y R1 t Y
N T-R / N
N~W2 B (RX)m ~(RX)m VII-a VII-b, wherein each of Ring A, Ring B, R', in, p, Rx, RY, R ,W', and W2 is as defined above with respect to formulae I-a and I-b, and described in classes and subclasses herein, R', is a bivalent warhead group, T is a bivalent tethering moiety; and Rt is a detectable moiety.
[00252] In some embodiments, Rt is a detectable moiety selected from a primary label or a secondary label. In certain embodiments, Rt 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.
[00253] 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).

[002541 Primary labels, such as radioisotopes (e.g., tritium, 32P 33P 35S 140 123I 124I 125I or 131I), mass-tags including, but not limited to, stable isotopes (e.g., 13C,2 H170 180, 15N 19F> and 127I , positron emitting isotopes (e.g., 110 18F 13N 124I and 150) 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.
[002551 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.
[002561 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, YOYO-1,5-FAM, BCECF, dichlorofluorescein, rhodamine 110, rhodamine 123, YO-PRO-1, SYTOX Green, Sodium Green, SYBR Green I, Alexa Fluor 500, FITC, Fluo-3, Fluo-4, fluoro-emerald, YoYo-1 ssDNA, YoYo-1 dsDNA, YoYo-1, 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-1,3-Diazole, Bexl, Doxorubicin, Lumio Green, and SuperGlo GFP.
[00257] 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, 170, 180, and '5N) may also be used as mass-tags.
[002581 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-1,4-phthalazinedione) reacts with oxidants like hydrogen peroxide (H202) in the presence of a base and a metal catalyst to produce an excited state product (3-aminophthalate, 3-APA).
[002591 The term "chromophore," as used herein, refers to a molecule which absorbs light of visible wavelengths, UV wavelengths or IR wavelengths.
[002601 The term "dye," as used herein, refers to a soluble, coloring substance which contains a chromophore.
[002611 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.
[002621 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.
[002631 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.

[00264] 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.
[00265] 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.
[00266] The term "photoisomerizable moiety," as used herein, refers to a group wherein upon illumination with light changes from one isomeric form to another.
[00267] 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.
[00268] 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.
[00269] 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).
[00270] 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.
[00271] 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, 552-57.
In some embodiments, a click ready inhibitor moiety is provided and reacted with a click ready -T-R' 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-W 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).
[002721 In certain embodiments, the click ready inhibitor moiety is of one of the following formulae:

(R"p A (R"p A

v/ N 0 r^` N3 v/ N2 W(,!~ N R1 (R")p (R A 0+0/ N3 A 0\ ~O~ /N3 " v 1 /

v R1 F~_e F

@ (RX)m B (RX) N W2 e'N W2 ~0 1 HN" ~N N3 (")p (R"p A 0 W1 W1 HN" v v N4-_O~N 3 v v q (R X)m ~(Rx)m N WZ or N WZ

wherein Ring A, Ring B, W1, W2, Ry, R , p, R", and in are as defined above with respect to Formula I and described herein, and q is 1, 2, or 3.

[002731 Exemplary click ready inhibitors include:

\ I N N3 HN \H NH HN +0 F

N N O--~ *N3 NN
H 2 H and HN N
F I N / I O\~0~/N3 N~N \ F
H
[00274] In some embodiments, the click ready -T-W moiety is of formula:
MeO - (O \__\,A

Me0` N 0-,\-o S H
J NH

0 \\O
[00275] An exemplary reaction in which a click ready inhibitor moiety and a click ready -T-Rt moiety are joined through a [2+3]-cycloaddition is as follows:

HN \ H" v I0I /
IN HN \ I N"
NN \ I O O N
H aI \ I 0 N
+ N H 0\ /2 N' N

Nom/ OMe MeO o S //
\_~H O CHN OMe Me0N \00 H H Nk\ -^ JNH 0 0 yN H HN-~

[00276] In some embodiments, the detectable moiety, Rr, is selected from 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, or a combination thereof.
[002771 In some embodiments, Rt is biotin or an analog thereof. In certain embodiments, Rt is biotin. In certain other embodiments, Rt is biotin sulfoxide.
[002781 In another embodiment, Rt 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 5301550, 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, YOYO-1,5-FAM, BCECF, dichlorofluorescein, rhodamine 110, rhodamine 123, YO-PRO-1, SYTOX
Green, Sodium Green, SYBR Green I, Alexa Fluor 500, FITC, Fluo-3, Fluo-4, fluoro-emerald, YoYo-1 ssDNA, YoYo-l dsDNA, YoYo-1, 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, Benzylamino-4-Nitrobenz-2-Oxa-1,3-Diazole, Bexl, Doxorubicin, Lumio Green, or SuperGlo GFP.
[00279] 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.
[00280] 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(0)2. In other embodiments, the water soluble polymer is a PEG group.
[00281] 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.
[002821 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, Rr, 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.
[002831 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.

[002841 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.
[00285] One of ordinary skill in the art will appreciate that when -T-R 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 -R''-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 -R''-T- may be substituted with an -L-Y warhead group, wherein such groups are as described herein. Alternatively, the tethering moiety formed by -R''-T- has the appropriate features of a warhead group incorporated within the tethering moiety. For example, 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'-T- tethering moiety are depicted below.
[00286] In some embodiments, a methylene unit of an -R"-T- tethering moiety is replaced by a bivalent -L-Y'- moiety to provide a compound of formula V-a-iii or V-b-iii:

~RV~n L_Y _T_Rt A ~RVp A

Y ~ L-Y'-T-Rt y INI
N (Rx)m &(Rx),, N W2 \NW2 V-a-iii V-b-iii wherein each of Ring A, Ring B, in, p, Rx, Ry, R,W', W2, T, L, Y', and Rt 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.
[00287] In some embodiments, a methylene unit of an -R"-T- tethering moiety is replaced by an -L(Y)- moiety to provide a compound of formula V-a-iv or V-b-iv:

Y
( ")p L-T-Rt W1 Y (RV p A W1 Fly L-T-Rt y N
Il 2 g (RX)m NW2 B (RX)m N W
V-a-iv V-b-iv wherein each of Ring A, Ring B, m, p, RX, Ry, R,W', W2, T, L, Y, and Rt is as defined above and described in classes and subclasses herein.
[002881 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:

Rt ( V)p T-L-Y
Y (RVp A
1 i W1 y T-Rt y N

N -(RX)m B (RX)m \N W2 N~W2 V-a-v V-b-v wherein each of Ring A, Ring B, m, p, RX, Ry, R,W', W2, T, L, Y, and Rt is as defined above and described in classes and subclasses herein.
[002891 In certain embodiments, the tethering moiety, -T-, has one of the following structures:

)J 'J
H H H
[002901 In some embodiments, the tethering moiety, -T-, has the following structure:
IOJ _OJ

H H H
[002911 In other embodiments, the tethering moiety, -T-, has the following structure:
1O--'~~ N
H
[002921 In certain other embodiments, the tethering moiety, -T-, has the following structure:

HN' HOOC HH -YO IOI

[00293] In yet other embodiments, thes tethering moiety, -T-, has the following structure:
N
NNk H
[00294] In some embodiments, the tethering moiety, -T-, has the following structure:

N-N OMe ,_/-N"' H
N / OMe /~0~~0 N` 0,-)l "2 O ~0 N N
H

[00295] In some embodiments, -T-W is of the following structure:

0 0 0 HN -,// 0 NH
H H H H S

[00296] In other embodiments, -T-W is of the following structure:

O 0 0 i0 H~ HN f -NN-- ~O--~ON/~O-'-~H H NH
H S

[00297] In certain embodiments, -T-W is of the following structure:

N_N We /-_/\H
S
N r^ 0~ N t!: OMe /--/
1,2 0 NO H H
N H
HNNH

[00298] 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.
[00299] In certain embodiments, the probe compound is one of the following structures:
a ' \
HN N
H
N

N)N O" -"'~NH
H

H
0 NH J,~~ 0S

I-215 0v 0 V `0' V `N
H

\ NH HN NH HN \ H H
F I
N O
/ I O I N/

F
NNH\ H

0 HN. O O
N~/~0~~0-/~O" v l*' / O~~O~~NH
H r I 1-362 H,HNO HN H

S H S NH

HN N

NIN"
H
O
HN H
O Nr0 S NH

N` 0 4c 0 HN \ IO F /
N
F , I N N\ I 0/\O~i N O
H
NIN
H

NH NH
HIIN H HN H
0 Nr0 0 Nr0 S NH S NH

F HN \ I N H N\ HN H H
F / 0"/-0~~ N 0 / 0N 0 NIN \ I NN \
F F
H H

NH NH
HN H HN H
N'rO 0 N,~O
O
S NH S NH

HN H
F N
0 e 11N~ N-NH

NH

HN H
N,?:r,O
O
S NH
1-368.
[003001 It will be appreciated that many -T-W 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-PEG]2-Biotin.
[003011 In some embodiments, analogous probe structures to the ones exemplified above are prepared using click-ready inhibitor moieties and click-ready -T-W moieties, as described herein.
[003021 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.
[003031 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.
[003041 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.
[003051 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 ErbB 1, ErbB2, or ErbB4. In yet other embodiments, the protein kinase is TEC, ITK, or BMX.
[003061 In some embodiments, the probe compound comprises the irreversible inhibitor for which occupancy is being determined.
[003071 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.
[003081 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.
[003091 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.
[00310] 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.
[00311] 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.
[00312] 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.
[00313] In certain embodiments, the probe compound is detected by binding to avidin, streptavidin, neutravidin, or captavidin.
[00314] 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.
[00315] 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 multi-dimensional LC-MS-MS to identify protein kinases modified by the probe compound and the adduction sites of said kinases.
[00316] 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.

[003171 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
[003181 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.
[003191 Compound numbers utilized in the Examples below correspond to compound numbers set forth in Table 5, supra.

[003201 Preparation of N-(3-(5-methyl-2-(phenylamino)pyrimidin-4-ylamino)phenyl) acrylamide 1-7 N

[003211 The title compound was prepared according to the schemes, steps and intermediates described below.

CI NH H N \ NH clod / NH
NH2 z -~' 1, 2 4 J. 6 ~ \
N CI Step-1 N CI step-2 N N step-3 N N

A) DIPEA, n-BuOH, 120 C, 30 min, MW; B) NMP, 200 C, 10 min, MW; C) NMP, 0 C-min, rt-30 min.
[003221 Step-1 / NH
N
N CI

[003231 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 C 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 (Si02, 60-120 mesh, EtOAc/CHC13 :
15/85) gave 3 (1.3 g, 45 %) as a dark brown solid.
[003241 Step_2 / NH
N
NN \
H
[003251 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 C, 10 min). The reaction mixture was cooled, diluted with water (100 mL) and extracted with EtOAc (3x 100 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 (Si02, CHC13/MeOH : 98/2) gave 5 (0.5 g, 40.3%) as a light brown solid.
[00326] Step-3 vxNH

NH
N ~
NN \
H

[00327] To a stirred solution of 5 (200 mg, 0.68 mmol) in NMP (2.0 mL) at 0 C
was added acryloyl chloride (248 mg, 0.2.74 mmol) and the reaction mixture was stirred at 0 C for 60 min.. The reaction mixture was then stirred with hexane for 1/2 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 (Si02, 230-400, MeOH/ CHC13 : 10/90) gave 1-7 (110 mg, 46.4%) as a brown solid. 'HNMR (DMSO-d6) 6 ppm: 2.10 (s, 3H), 5.73 (dd, 1.88& 10.42 Hz, 1H), 6.24 (dd, J-1.88 & 17 Hz, 1H), 6.44 (dd, J - 10.08 & 16.92 Hz, 1H), 6.78 (t, J- 7.36 Hz, 1H), 7.06-7.11 (m, 2H), 7.26 (t, J- 8.08 Hz, 1H), 7.38-7.40 (bm, 2H), 7.65 (d, J- 8.52 Hz, 2H), 7.88 (s, 1H), 7.92 (s, 1H), 8.37 (s, 1H), 8.91 (s, 1H), 10.09 (s, 1H); LCMS: m/e 346.8 (M+l).

[00328] Preparation of N-(3-(4-(m-tolylamino)pyrimidin-2-ylamino)phenyl)acrylamide I-1 \ I N N' v [003291 The title compound was prepared according to the schemes, steps and intermediates described below.
N

\\ NN step-1 step-2 aNH2 step-3 NH

NIN NH
H

A) DIEA, n-BuOH, 110 C, 30 min, microwave; B) NMP, 200 C, 10 min, microwave;
C) acryloyl chloride, NMP, 0 C-30 min, rt-30 min.
[003301 Step-1 NH
NICI

[003311 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 C
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 (Si02, 60-120 mesh, CHC13/MeOH : 99/1) gave 3 (0.4 g, 54.2%) as a yellow solid.
[00332] Step_2 NH
NN \ NH2 [00333] 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 C, 10 min). Then the reaction mixture was cooled, diluted with water (10 mL) and extracted with CH2C12 (3x15 mL). The combined CH2CI2 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 (Si02, CHC13/MeOH : 98/2) and gave 5 (0.14 g, 53%) as a light yellow solid.
[00334] Step-3 NH

/
N"N \ NH
H
0 ~), [00335] To a stirred solution of 5 (0.075 g, 0.25 mmol) in NMP (1.0 mL) at 0 C was added acryloyl chloride (0.19 g, 2.0 mmoL) and the reaction mixture was stirred at 0 C for 30 min followed by stirring at rt for 30 min. The neat reaction mixture was subjected to purification by column chromatography (neutral A1203, CHC13/MeOH : 98/2) gave I-1 (0.04 g, 45%) as a white solid. 'H NMR (DMSO-d6) 6 ppm: 2.56 (s, 3H), 5.71 (dd, J= 2.0 & 10.08 Hz, lH), 6.20-6.25 (m, 2H), 6.45 (dd, J= 10.12 & 17.00 Hz, 1H), 6.78 (d, J= 7.52 Hz, 1H), 7.12-7.19 (m, 2H), 7.31 (d, J = 8.44 Hz, 1H), 7.46-7.53 (m, 3H), 7.87 (s, 1H), 7.99 (d, J = 5.76 Hz, 1H), 9.15 (s, 1H), 9.24 (s, 1H), 10.03 (s, 1H); LCMS : m/e 346.4 (M+l).

[00336] Preparation of N-(3-(5-methyl-4-(m-tolylamino)pyrimidin-2-ylamino)phenyl) acrylamide 1-2 N \0 N

[00337] The title compound was prepared according to the schemes, steps and intermediates described below.

I NH
CI NH2 0NH H N~NH

~ step-1 N step-2 N /
N CI A N CI B \N~H \ NH2 C step-3 'a NH

N I~N ~ I N" v H H

A) DIPEA, n-BuOH, 110 C, 30 min, MW; B) NMP, 200 C, 15 min, MW; C) acryloyl chloride, NMP, 0 C-30 min, rt-30 min.
[00338] Step-1 i \ NH
NICI

[003391 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 C for 90 min. The reaction mixture was cooled, concentrated under reduced pressure and the residue obtained was further purified by column chromatography (Si02, Methanol/chloroform mixtures) gave 3 (0.05 g, 34 %) as an off white solid.
[003401 Step_2 NH

[003411 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 C, 15 min). Then the reaction mixture was cooled, diluted with water (15 mL) and extracted with EtOAc (3x 15 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 (Si02, CHC13/MeOH: 98/2) gave 5 (0.03 g, 46%) as a grey solid.
[003421 Sten-3 NH

-~-- 1 0 N N N
H H
1.2 [003431 To a stirred solution of 5 (0.025 g, 0.082 mmol) in NMP (0.5 mL) at 0 C was added acryloyl chloride (0.073 g, 0.821 mmol) and the reaction mixture was stirred at 0 C for 30 min followed by stirring at rt for 30 min. The crude reaction mixture was passed through an alumina column (neutral A1203, chloroform/methanol mixtures) gave 1-2 (0.012 g, 41%) as a pale brown solid. 'H NMR (DMSO-d6) 6 ppm: 2.10 (s, 3H), 2.27 (s, 3H), 5.72 (dd, J- 2 &
10.04 Hz, 1H), 6.22 (dd, J- 1.96 & 16.92 Hz, 1H), 6.45 (dd, J- 10.08 & 16.92 Hz, 1H), 6.83 (d, J- 7.36 Hz, 1H), 7.09 (t, J- 8.06 Hz, 1H), 7.17 (t, J- 7.78 Hz, 1H), 7.26 (d, J- 7.80 Hz, 1H), 7.47 (d, J-1.08 Hz, 1H), 7.53 (s, 1H), 7.58 (d, J - 8.60 Hz, 1H), 7.78 (s, 1H), 7.88 (s, 1H), 8.15 (s, 1H), 9.01 (s, 1H), 9.99 (s, 1H); LCMS: m/e 360.1 (M+1).

[003441 Preparation of N-(3-(5-fluoro-4-(m-tolylamino)pyrimidin-2-ylamino)phenyl) acrylamide 1-3 \ N N' L
F
N

[003451 The title compound was prepared according to the schemes, steps and intermediates described below.

NH2 H2N \ \

F N step-1 F N step-2 F
N
N" 'CI A NCI B
NIH \

0 C step-3 CI

"a NH HN" v F

NIN
H

A) n-butanol, DIPEA, 110 C, 45 min, MW; B) NMP, 200 C, 10 min, MW; C) NMP, DMAP, 0 C, 30 min.

[003461 Step-1 NH
F ~
NICI

[003471 To a solution of 1 (0.5 g, 3 mmol) in n-butanol (5.0 mL) 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 C 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.
[003481 Step_2 NH
F ~N
N~N \

[003491 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 C 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 (Si02, 60-120, Chloroform/Ethyl acetate: 90/10) gave 5 (0.23 g, 41%) as a light yellow solid.
[003501 Step-3 "a NH HN" v F N

H

[003511 To a stirred solution of 5 ( 0.075 g, 0.24 mmol), in NMP(1.5 mL) at 0 C 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 (A1203, Chloroform/Methanol: 98/2) gave 1-3 (0.01 g, 11.3%) as an off white solid. 1H NMR (DMSO-d6) b ppm: 2.27 (s, 3H), 5.72 (d, J- 9.84 Hz, 1H), 6.22 (d, J- 16.92 Hz, 1 H), 6.44 (dd, J - 10.2 & 17.02 Hz, 1 H), 6.85 (d, J - 7.12 Hz, 1 H), 7.12-7.19 (m, 2H), 7.29 (d, J
- 7.68 Hz, 1H), 7.43 (d, J- 7.92 Hz, 1H), 7.61-7.63 (m, 2H), 7.82 (s, 1H), 8.08 (s, 1H), 9.23 (bs, 2H), 10.03 (s, 1H); LCMS: m/e 364.2 (M+l).

[003521 Preparation of (E)-4-(dimethylamino)-N-(3-(5-fluoro-4-(m-tolylamino)pyrimidin-2-ylamino)phenyl)but-2-enamide 1-4 / I
N o F I ~N /

[003531 The title compound was prepared according to the schemes, steps and intermediates described below.

F~, step-1 F ,I N 4 NHz F ,,I

~1 Ili, Nom" CI ~ NCI step-2 N N\ Q

CIOC HOOC
C
step-3 D step-2' HCI
iN /N,_ NH HN v vN, F

NN \
H

A) n-butanol, DIPEA, 110 C, 45 min., MW; B) NMP, 200 C, 10 min., MW; C) oxalyl chloride, CH3CN, 1/2 h at 0 C, 2 h at 25 C, 5 min at 45 C; D) NMP, 0 C to 10 C, 30 min.
[003541 Step-1 NH
F N
N" 'CI

[003551 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 C, 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.
[003561 Step_2 NH
F ~N
N~H

[003571 A solution of 3 (0.45 g, 1.8 mmol), 4 (0.41 g, 3.7 mmol) in NMP (4.5 mL) was subjected to microwave irradiation (200 C, 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 (Si02, Chloroform/Ethyl acetate:
90/10) gave 5 (0.23 g, 41%) as a light yellow solid.
[003581 Step-3a COCI
N, N-/-/

[003591 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 C. The reaction mixture was allowed to stir at 0 C for 1/2 h and then at RT for 2 h. Finally it was heated at 45 C for 5 min, cooled and the reaction mixture was taken for next step without further purification.
[003601 Step-3 i NH HN' N' N N L
H

[003611 To a stirred solution of 5 (0.05 g, 0.16 mmol) in NMP (1.0 mL) was added 7 at 0 C.
The reaction mixture was stirred at 0 C for 30 min and at 10 C for 30 min.
It was quenched with sat. Sodium bicarbonate sole. (5 mL) and extracted with CH2Clz (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 (Si02, 230-400, CHC13/MeOH, 95/5) gave 1-4 (0.02 g, 29.4%) as a white solid.
iH NMR
(DMSO-d6) 6 ppm: 2.21 (s, 6H), 2.28 (s, 3H), 3.08 (bd, J- 5.6 Hz, 2H), 6.29 (d, J- 15.60 Hz, 1H), 6.67-6.74 (m, 1H), 6.86 (d, J- 7.20 Hz, 1H), 7.12-7.20 (m, 2H), 7.27 (d, J- 8.00 Hz, 1H), 7.43 (d, J- 8.00 Hz, 1H), 7.62-7.64 (m, 2H), 7.82 (s, 1H), 8.08 (d, J- 3.6 Hz, 1H), 9.23 (s, 1H), 9.24 (s, 1H), 9.96 (s, 1H); LCMS: m/e 421.2 (M+1).

[00362] Preparation of N-(3-(5-methyl-4-(phenylamino)pyrimidin-2-ylamino)phenyl) acrylamide I-5 N
I~~I

[00363] The title compound was prepared according to the schemes, steps and intermediates described below.

CI NH, ~ I I ~ NH
2 NH H ZN 4 NH, IN step-1 IN IN
step-2 II
N CI A N CI B N H N NHZ

C step-3 NH

eN 00 I N '01 , N"
H H

A) DIPEA, n-BuOH, 110 C, 30 min, MW; B) NMP, 200 C, 15 min, MW; C) acryloyl chloride, NMP, 0 C-30 min, rt-30 min.
[00364] Step-1 NH
N"CI

[00365] 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 C for 90 min. The reaction mixture was cooled, concentrated under reduced pressure and the residue was further purified by column chromatography (Si02, 60-120, Methanol/chloroform: 1/9) gave 3 (0.08 g, 59 %) as a white solid [003661 Step_2 NH

[003671 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 C, 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 (Si02, 60-120, CHC13/MeOH: 98/2) gave 5(0.06 g, 60%) as a light grey solid.
'H NMR (DMSO-d6) 6 ppm: 2.09 (s, 3H), 4.74 (s, 2H), 6.09-6.11 (m, 1H), 6.77-6.85 (m, 2H), 6.91 (t, J - 1.72 Hz, I H), 7.02 (t, J - 7.36 Hz, I H), 7.31 (t, J - 7.52 Hz, 2H), 7.75 (d, J - 7.68 Hz, 2H), 7.84 (s, 1H), 8.18 (s, 1H), 8.65 (s, 1H); LCMS: m/e 293.2 (M+1).
[003681 Step-3 C~NH
e'N I N N
H H

[003691 To a stirred solution of 5 (60 mg, 0.205 mmol) in NMP (2.0 mL) at 0 C
was added acryloyl chloride (0.148 g, 1.64 mmol) and the reaction mixture was stirred at 0 C for 30 min..
The neat reaction mixture was passed through an alumina column (neutral Al203, chloroform/methanol, 99/1) gave 1-5 (0.013 g, 18.5%) as an off-white solid. 'H
NMR (DMSO-d6) 6 ppm: 2.11 (s, 3H), 5.72 (dd, J- 1.92 & 10.04 Hz, 1H), 6.22 (dd, J- 1.92 & 16.92 Hz, 111), 6.45 (dd, J - 9.32 & 16.92 Hz, I H), 7.00 (t, J - 7.28 Hz, 1H), 7.09 (t, J -8.04 Hz, 111), 7.23-7.30 (m, 3H), 7.43 (d, J- 8.04 Hz, 1H), 7.75-7.77 (m, 2H), 7.83 (s, 1H), 7.88 (s, 1H), 8.22 (s, 1H), 9.00 (s, 1H), 9.99 (s, 1H); LCMS: m/e 346 (M+l).

[003701 Preparation of N-(4-methyl-3-(5-methyl-4-(m-tolylamino)pyrimidin-2-ylamino) phenyl)acrylamide 1-8 /I

\ N LO
N
I~~I

[003711 The title compound was prepared according to the schemes, steps and intermediates described below.
N H, A

step-1' 1 A' NH(BOC) CI 2 NH2 NH NH, / NH NH(BOC) NH NH2 iN Step -1 N 4 ~'NIN step -3 %N
N CI A N CI step-2 C N N

B 1 step-4 NH :N'0 N
NN
H

A) DIPEA, n-BuOH, 120 C, 60 min., MW; A') (BOC)20, MeOH, -10 C4 4 h; B) Pd(OAc)2, BINAP, Cs2CO3, toluene, 110 C, 12 h; C) TFA, CH202, 0 C-30 min, rt-2 h; D) acryloyl chloride, NMP, 0 C-30 min, rt-30 min.

[003721 Step 1, NH(BOC) [003731 To a stirred solution of A (5 g, 0.04 mmol) in McOH (75 mL) was added (BOC)20 (11.59 g, 0.050 mmol), slowly at -10 C. 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.
[003741 Step 1 NH
NICI

[003751 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 C, 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 (Si02, 60-120, CHC13/MeOH : 9/1) gave 3 (0.35 g, 49%) as an off-white solid.
[003761 Step 2 6NH NH(BOC) e'N II N
H

[00377] 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 (Si02, 60-120, CHCl3/MeOH : 9/1) gave 5 (40 mg, 22%) as an off-white solid.
[00378] Sten-3 /I

INiN
H

[00379] To a stirred solution of 5 (0.04 g, 0.095 mmol) in dry CH2C12 (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.
[00380] Step-4 aNH HN 0 NIN
H

[00381] To a stirred solution of 6 (0.2 g, 0.63 mmol) in NMP (4 mL) at 0 C
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 (2x 10 mL). The combined EtOAc extract was washed with water (2 mL), brine (2 mL) and dried over Na7SO4. Filtration followed by concentration under reduced pressure offered a residue which was further purified by column chromatography (Si02, 230-400, CHC13/MeOH
: 9/1) gave 1-8 (10 mg, 4%) as a white solid. 1H NMR (DMSO-d6) 6 ppm: 2.07 (s, 3H), 2.13 (s, 6H), 5.70 (dd,J=1.92&10.08Hz,lH),6.20(dd,J=1.96&16.88Hz,1H),6.41(dd,J=10.16&16.96 Hz, 1H), 6.69 (d, J = 7.36 Hz, 1H), 6.98 (t, J = 7.76 Hz, 1H), 7.11 (d, J =
8.24 Hz, 1H), 7.41 (q, J
= 9.92 Hz, 1H), 7.49-7.51 (m, 2H), 7.73 (s, 1H), 7.80 (s, 1H), 7.97 (s, 1H), 8.16 (s, 1H), 10.00 (s, 1H); LCMS : m/e 374 (M+l).

[003821 Preparation of N-(3-(4-(3-bromophenylamino)-5-methylpyrimidin-2-ylamino)phenyl) acrylamide 1-9 Br OO
N N' v IN
N

[003831 The title compound was prepared according to the schemes, steps and intermediates described below.

Br Br CI Br \ NH NH NH2 \N 2 N 4 N /~
NCI step-1 NLCI step-2 NN \

C step-3 Br OO
NH HN" `:
/ II
N N \
H

A) DIPEA, n-butanol, 110 C, 1 h, MW; B) 1.5 N HCl, ethanol, 90 C, 30 min., MW; C) acryloyl chloride, NMP, 0 C, 30 min.
[003841 Step 1 Br NH
NICI

[003851 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 C, 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 (Si02, 60-120, chloroform/methanol, 9/1) gave 3 (0.125 g, 13%) as a brown solid.

[003861 Step 2 Br \N II N Jb H
[003871 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 C, 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 (Si02, 60-120, chloroform/methanol, 9/1) gave 5 (0.06 g, 32%) as a light brown solid.
[003881 Step 3 Br NH HN" v e-N N
H

[003891 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 C. 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 (Si02, 60-120, chloroform/methanol, 9/1) gave 1-9 (0.016 g, 23%) as a pale brown solid. 1H NMR (DMSO-d6) 6 ppm: 2.16 (s, 3H), 5.75 (dd, J - 1.72 & 10 Hz, 1H), 6.23 (dd, J- 1.76 & 16.88, Hz, 1H), 6.45 (dd, J- 10.08 & 16.92 Hz, 1H), 7.22-7.34 (m, 4H), 7.38 (d, J- 8.00 Hz, 1H), 7.63 (d, J- 8.08 Hz, 1H), 7.77 (s, 1H), 7.82 (s, 1H), 7.93 (s, 1H), 9.68 (s, I H), 10.26 (s, I H), 10.34 (s, I H); LCMS: m/e 426 (M+l ).

[00390] Preparation of 3-(4-(2-(cyclopropylsulfonyl)-1,2,3,4-tetrahydroisoquinolin-6-ylamino)-5-methylpyrimidin-2-ylamino)benzenesulfonamide 1-10 Y

MN (~=~N
N

[00391] The title compound was prepared according to the schemes, steps and intermediates described below.

~ N ,BOC Step-1 BOG
ZBCHN ~2 A' HO'C ^ rI^N, B' Sep-2 CI I NBOC HN N,BOC HN I ~---,NH

'N S N 6 N
N'-CI Step-3 NCI step-4 N-~ HQ
4 q 5 B 7 SO2NH2 C Step-5 OSO

HN
\N -NK
N Q
H

A') DPPA, Benzyl alcohol, Et3N, toluene, 110 C, 12 h.; B') Pd(OH)2, Ammonium formate, EtOH, reflux, 6 h; A) DIPEA, n-BuOH, 120 C, 1 h., MW; B) 1.5 N HCl, EtOH, reflux 12 h.; C) Cyclopropylsulphonyl chloride, DIPEA, THF, rt, 12 h.

[003921 Steps 1-4 The procedure for synthesizing scaffold 7 is described in the experimental for Compound I-11 herein.
[003931 Step 5 HN
/N
N' N Q

[003941 To a stirred solution of 7 (0.05 g, 0.0121 mmol) in THE (4 mL) at 0 C, 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 (Si02, 60-120, pet ether/ethyl acetate, 6/4) gave 1-10 (0.035 g, 56%) as a yellow solid. 'H NMR (DMSO-d6) 6 ppm: 0.97-0.1.00 (m, 4H), 2.12 (s, 3H), 2.60-2.66 (m, 1H), 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, 1H), 7.27 (s, 2H), 7.31-7.35 (m, 2H), 7.53 (s, 1H), 7.59 (d, J-8.4 Hz, 1H), 7.92 (s, 1H), 8.03-8.04 (m, 2H), 8.45 (s, 1H), 9.40 (s, 1H);
LCMS: nile 515 (M+1).

[003951 Preparation of 3-(4-(2-(2-chloroacetyl)-1,2,3,4-tetrahydroisoquinolin-6-ylamino)-5-methylpyrimidin-2-ylamino)benzenesulfonamide I-11 C1j MN 0\\S<-N
0%

[00396] The title compound was prepared according to the schemes, steps and intermediates described below.

N, BOO Step -1 N,BOC
ZBCHN A' H02C

B, Sep-2 HN N,BOC HN ~-'-NH
CI BOC

3 N 6 'N
CI Step-3 N'-CI step-4 N~
/
N -Q

C Step-4 HN Cl /NH \ /

A') DPPA, Benzyl alcohol, Et3N, toluene, 110 C, 12 h.; B') Pd(OH)2, Ammonium formate, EtOH, reflux, 6 h; A) DIPEA, n-BuOH, 120 C, 1 h., MW; B) 1.5 N HC1, EtOH, reflux 12 h.; C) C1-CH2-0001, Et3N, THF, rt, 12 h.
[00397] Step-1 N BOC
ZBCHN

[00398] 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 (Si02, 60-120, chloroform/methanol, 9/1) gave 2 (2.0 g, 97%) as a white solid.

[003991 Sten_2 jN BOC

[004001 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.
[004011 Step-3 N,BOC
HW'f~

N iCI
[004021 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 C 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 (Si02, 60-120, chloroform/methanol, 9/1) gave 5 (1.1 g, 52%) as a cream colored solid.
[004031 Step-4 ~ 'NH
HN'J!

N={
N \ Q

[004041 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.HC1 and the reaction mixture was refluxed for 12 h at 100 C. 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.

[004051 Step-5 N'~
HN"' CI
N-N \
H

[004061 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 C 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 (Si02, 60-120, chloroform/methanol, 9/1) gave I-11 (0.038 g, 16%) as a light yellow solid. 'H NMR (DMSO-d6) 6 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, 1H), 7.24 (s, 2H), 7.30-7.32 (m, 2H), 7.50-7.65 (m, 2H), 7.91 (s, 1H), 8.04-8.05 (m, 2H), 8.27 (s, 1H), 9.31 (s, 1H), LCMS: m/e 486.8 (MH).

[004071 Preparation of N-(3-(5-methyl-4-(4-phenoxyphenylamino)pyrimidin-2-ylamino) phenyl)acrylamide 1-23 01Ph HN

TXNONH
H

[004081 The title compound was prepared according to the schemes, steps and intermediates described below.

Ph / Ph Ph NH2 ii HZN ~~ k" \I \I 'Ph N step-1 N step-2 N step-3 NN
NCI `4 NCI B NH I NHz C NH NH
1 3 5 1-23 01), A) DIPEA, n-butanol, 100 C, 1 h, MW; B) conc.HCl, n-BuOH, 160 C, 20 min., MW; C) Acryloyl chloride 0 C, rt, 1 h.

[004091 Step-1 Ph O
HN

NICI

[004101 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 mL) was subjected to microwave irradiation (100 C 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 (Si02, 60-120, chloroform/methanol, 9/1) gave 3 (0.11 g, 28.9%) as a light brown solid.
[004111 Step_2 Ph O
HN

~
NN \ I NH2 H
[004121 To a solution 3 (0.11 g, 0.3 mmol), 4 (0.114 g, 1.05 mmol) in n-butanol (1 mL) was added conc. HCl (1 drop) and the mixture was subjected to microwave irradiation (165 C 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 mL). Drying over Na2SO4 followed by concentration under reduced pressure offered residue which was purified by column chromatography (Si02, 60-120, chloroform/methanol, 9/1) gave 5 (0.08 g, 65%) as a brown solid.
[00413] Step-3 0, Ph HN

1\N /
NXN \ NH
H

1-23 0101), [00414] 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 C. 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 (Si02, 60-120, chloroform/methanol, 9/1) gave 1-23 (0.004 g, 23%) as a brown solid. 400 MHz, MeOD: 6 2.14 (s, 3H), 5.71 (d, J= 11.20 Hz, 1H), 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, 1H), 7.34-7.36 (m, 3H), 7.63 (d, J = 8.8 Hz, 2H), 7.79 (s, 2H); LCMS: m/e 437 (M+l).

[00415] Preparation of N-(3-(5 -methyl-2-(3 -sulfamoylphenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-33 oo H N' H
H;

N as-*' NH, H/\\

[00416] The title compound was prepared according to the schemes, steps and intermediates described below.

CI
N
NCI
1.

step-1 A

~~ HN\I 0 HNNH2 \
HN" v `NH2 2 H2N~ HN
ON iL 0 H

N 2 I \ I %0 4 \ I
N N a NCI step-2 H H2N'0 step- 3 N H H2N.o A) DIPEA, n-BuOH, 120 C, 30 min., MW; B) 1.5 N HC1, Ethanol, 100 C, 12 h; C) NMP, 0 C
to rt, 1 h.
[004171 Step-1 HN \ NH2 xcI
[004181 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 mL) was subjected to microwave irradiation (120 C, 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 (Si02, 60-120, chloroform/ethyl acetate, 9/1) gave 1 (0.25 g, 34.77%) as a light brown solid.
[004191 Step_2 HN \ NH2 NI
J\ \ .,O

[004201 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 C, 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.
[00421] Step-3 HN N
H
SN ~
I '0 N! H H2N'1i0 [00422] To a stirred solution of 5 (0.1 g, 0.27 mmol) in NMP (2 mL) was added acryloyl chloride (0.037 g, 0.425 mmol) at 0 C, 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) 6 ppm: 2.17 (s, 3H), 5.78 (dd, J= 2.36 & 9.52 Hz, 1H), 6.34-6.48 (m, 2H), 7.26-7.43 (m, 5H), 7.87 (s, 1H), 7.96-8.03 (m, 3H); LCMS: m/e 425 (M+1).

[00423] Preparation of N-(3-(methyl(5-methyl-2-(phenylamino)pyrimidin-4-yl)amino) phenyl)acrylamide 1-34 o H
tj~N

H

[00424] The title compound was prepared according to the schemes, steps and intermediates described below.

CI

~~1111 N CI HN" NO2 N ND2 N \ I NOz Me 'N step-2 Me 'N step-3 MeN -H2N 1 I NO2 step-1 N 3 CI B I N CI ~H
N N

C 1 step-4 \N \ I N ^ /CI IN \ I NHz H 0 MeN
Me I E ~ _ INN \ I E "H

A) Pd(OAC)2, BINAP, Cs2CO3, Toluene, 100 C, 16 h; B) NaH, CH3I, THF, 0 C-30 min, rt-16 h.; C) Aniline, conc.HC1, Ethanol, 90 C, 60 min; D) H2, Pd/C, Ethanol, 16 h;
E) acryloyl chloride, NMP, 0 C, 1 h.
[004251 Step-1 HN \ NO2 Me N! C1 [004261 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, 03 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 C 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 (Si02, 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.

[004271 Sten_2 \N a N02 Me LNCI

[004281 To a stirred mixture of NaH (0.1 g, 2.5 mmol, 60% dispersion in paraffin oil) in dry THE (10.0 mL) was added 3 (0.5 g, 1.89 mmol) at 0 C, 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 (Si02, CHC13/MeOH: 99/1) gave 4 (0.12 g, 22.7%) as a light yellow solid.
[004291 Sten-3 i N. N \ NO2 Me [004301 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 C 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 (Si02, CHC13/MeOH: 99/1) gave 5 (0.11 g, 76%) as a light yellow solid.

[004311 Step-4 a N, N NH2 Me 6N Ni [004321 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 (Si02, 60-120, methanol/chloroform: 1/99) gave 6 (0.07 g, 69.9%) as a colorless viscous liquid.
[004331 Step-5 i ~N \ I N v Me H

N N
H

[004341 To a stirred solution of 6 (0.070 g, 0.23 mmol) in NMP (1.5 mL) at 0 C was added acryloyl chloride (0.083 g, 0.916 mmol) and the reaction mixture was stirred at 0 C 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. 'H NMR (DMSO-d6) 6 ppm: 6 1.47 (s, 3H), 3.45 (s, 3H), 5.74 (dd, J- Hz, 1H), 6.22 (dd, J- 2.0 & 16.98 Hz, 1H), 6.38 (dd, J -& 16.94 Hz, 1 H), 6.85-6.91 (m, 2H), 7.21-7.25 (m, 2H), 7.32 (t, J - 8.02 Hz, 1 H), 7.43-7.47 (m, 2H), 7.77-7.79 (m, 2H), 7.90 (s, 1H), 9.22 (s, 1H), 10.18 (s, 1H); LCMS:
m/e 360.8 (M+1).

[00435] Preparation of N-(3-(5-methyl-2-(3-(prop-2-ynyloxy)phenylamino)pyrimidin-4-ylamino)phenyl) acrylamide 1-35 oo H aN v H
N

H

[00436] The title compound was prepared according to the schemes, steps and intermediates described below.
\ I step-1 \

02N OH 02N O"
1a 2b \ I step-2 B \

N step-3 N step-4 N /

CI
E step-5 O

HN \ N" v N
NN_ O"'-~' H

A) K2CO3, CH3CN665 C, 8 h; B) Fe powder, NH4Cl, McOH, H20, 80 C, 4 h; C) 1,3-pheneylendiamine, DIPEA, n-BuOH, 120 C, 30 min, MW; D) Con.HC1, absolute ethanol, 110 C, 2 h; E) NMP, 0 C, 1 h.
[00437] Step-1 2b [004381 To a stirred solution of la (4 g, 0.0287 mot) and K2CO3 (5.6 g, 0.0574 mot) in CH3CN (15 mL) was added propargyl bromide (4.1 g, 0.0345 mot) 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.
[004391 Step_2 H2N \ 0~

[004401 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 mot) and iron powder (6.8 g, 0.121 mot) respectively. Resulting mixture was refluxed at 80 C 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 (Si02, 60-120, gravity column chromatography, the expected product was eluted with CHC13/MeOH : 96/4) gave 3 (3.2 g, 91%) as a brownish solid.
[004411 Step-3 i HN \ NH2 N! C1 [004421 A solution of 2, 4-dichloro-5-methyl pyrimidine 1 (0.3 g, 0.0018 mot), 1,3-phenylene diamine (0.24 g, 0022 mot), DIPEA (0.35 g, 0.0027 mot) in n-BuOH (3 mL) was subjected to microwave irradiation (120 C, 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 (Si02, 60-120) gave 2 (0.15 g, 35%) as a brownish solid.

[004431 Step-4 i HN \ NH2 /
N

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 C 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 (Si02, 60-120, gravity column chromatography, expected compound getting eluted in CHC13/MeOH : 94/6) gave 4 (125 mg, 56%) as a light brown solid.

[004441 Step-5 0 HN N
H

NIH \ 0' [004451 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 T. 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 (2x 15 mL). The combined EtOAc extract was washed with water (10 mL), brine (10 mL) dried over Na7SO4 and concentrated under reduced pressure. The residue obtained was purified by column chromatography (Si02, 60-120, gravity column chromatography, expected compound getting eluted in CHC13/MeOH : 90/10) gave 1-35 (20 mg, 18%) as an off-white solid. iH
NMR
(DMSO-d6) 6 ppm: 2.11 (s, 3H), 3.51 (s, lH), 4.61 (s, 2H), 5.74 (d, J = 9.08 Hz, lH), 6.25 (d, J =
15.84 Hz, lH), 6.45 (s, 2H), 7.02 (s, lH), 7.27-7.45 (m, 5H), 7.91 (d, J =
8.84 Hz, 2H), 8.36 (s, 1H), 8.93 (s, lH), 10.09 (s, lH), LCMS: m/e 400 (M+1).

[004461 Preparation of (E)-4-(dimethylamino)-N-(3-(5-methyl-2-(phenylamino)pyrimidin-4-ylamino) phenyl)but-2-enamide 1-38 HN / N,, 6'-~NH

I~~I

H

[004471 The title compound was prepared according to the schemes, steps and intermediates described below.

HOOCH` k I
6 ~- ^ I00 N
NH2 NH2 NHZ 'NH
step-2' /IL C

CI (/ NH2 NH H2N \ I NH N~ NH

e, il N CI step-1 N CI step-2 N JLN \ step-3 N N
H H

A) DIEA, n-BuOH, 120 C, 30 min, MW; B) NMP, 200 C, 10 min, MW; C)) oxalyl chloride, CH3CN, 30 min at 0 C, 2 h at 25 C, 5 min at 45 C, D) NMP, 0 C, 1 h.
[004481 Step-1 / NH
eUcl [004491 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 C 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 (Si02, 60-120 mesh, EtOAc/CHC13:15/85) gave 3 (1.3 g, 45 %) as a dark brown solid.
[004501 Step_2 & NH

N N
H
[004511 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 C, 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, CHC13/MeOH : 98/2) gave 5 (0.5 g, 40.3%) as a light brown solid.
[004521 Step-2' CIOC

[004531 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 C. The reaction mixture was allowed to stir at 0 C for i/2 h and then at rt for 2 h. Finally it was heated at 45 C for 5 min, cooled and the reaction mixture was taken for the next step without further purification.

[004541 Sten-3 /N'~'NH

NH
N N
H

[004551 To a stirred solution of 5 (75 mg, 0.12 mmol) in NMP (1 mL) was added 6 at 0 C.
The reaction mixture was stirred at 0 C for 1 h, quenched with cold water (5 mL), basified with Et3N and extracted with CH2CI2 (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. 'H NMR
(DMSO-d6) 6 ppm: 2.15 (s, 3H), 2.32 (s, 6H), 3.21 (d, J- 5.76 Hz, 2H), 6.27 (d, J- 15.36 Hz, 1H), 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, 1H), 7.53 (d, J- 7.72 Hz, 2H), 7.80 (s, 1H), 8.00 (s, 1H); LCMS: m/e 402.8 (M+1).

[004561 Preparation of N-(4-(5-methyl-2-(phenylamino)pyrimidin-4-ylamino)phenyl) acrylamide 1-39 H

H
M N

\
H

[004571 The title compound was prepared according to the schemes, steps and intermediates described below.
/ NHZ H
HZN \ I \ I NHZ 0,NH2 \ I NMZ \ I NO

IN step-] IN step-2 'N step-3 IN /
1 1), CNC1 N 0 B CN N\ I C I N LN \ I
H H

A) DIPEA, n-BuOH, 110 C, 45 min, MW; B) Conc. HC1, n-BuOH, 150 C, 10 min, MW;
C) Acryloyl chloride, NMP, 0 C-30 min, rt-2 h.
[004581 Step-1 HN

N
NY'CI

[004591 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 C, 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 (Si02, 60-120, CHC13/MeOH : 99/1) gave 3 (350 mg, 62%) as an off-white solid.
[004601 Step_2 HN

/
NN \
H
[004611 A solution of 3 (0.2 g, 0.8 mmol), 4 (0.63 g, 6.8 mmol) and con.HC1 (0.03 g, 0.8 mmol) in n-BuOH (10 mL) was subjected to microwave irradiation (150 C, 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, CHC13/MeOH
: 97/3) gave (110 mg, 47%) as a brown colored gummy solid.
[00462] Step-3 H
N`^

N"N
H

[00463] 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 C. 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 (Si02, 60-120) and finally by preparative HPLC gave 1-39 (10 mg, 16%) as an off-white solid. 'H NMR (DMSO-d6) 6 ppm:
2.10 (s, 3H), 5.71-5.76 (m, 1H), 6.25 (dd, J 2.04 & 16.96 Hz, 1H), 6.45 (dd, J
- 10.08 & 16.92 Hz, 1H), 6.84 (t, J - 7.30 Hz, 1H), 7.14-7.18 (m, 2H), 7.62-7.68 (m, 6H), 7.86 (s, 1H), 8.26 (s, 1H), 8.94 (s, 1H), 10.11 (s, 1H), LCMS: m/e 346 (M+1).

[00464] Preparation of N-(3-(5-methyl-2-(phenylamino)pyrimidin-4-ylamino)phenyl) propionamide IR-7 v `N

N

N

o [004651 The title compound was prepared according to the schemes, steps and intermediates described below.

NH2 NH2 NH2 v NH

IIINI :p, 0 CI N CI step-2 N N step-3 N N
H H

A) DIPEA, n-BuOH, 120 C, 30 min, MW; B) NMP, 200 C, 10 min, MW; C) 6, NMP, 0 C, 60 min.
[004661 Step-1 & NH

\N CI

[004671 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 C 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 (Si02, 60-120 mesh, EtOAc/CHC13 : 15/85) gave 3 (1.3 g, 45%) as a dark brown solid.
[004681 Step_2 & NH
e'N!NJO
H

[004691 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 C, 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 (Si02, CHC13/MeOH : 98/2) gave 5 (0.5 g, 40.3%) as a light brown solid.
[004701 Sten-3 --'~NH

NH
e'N IN
H

[004711 To a stirred solution of 5 (75 mg, 0.25 mmol) in NMP (1.0 mL) at 0 C
was added propanoyl chloride (6) (72 mg, 0.75 mmol) and the reaction mixture was stirred at 0 C 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 (Si02, 230-400, methanol/chloroform :
2/98) gave IR-7 (0.025 g, 28.73%) as an off white solid. 1H NMR (DMSO-d6) 6 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, 1H), 7.11 (t, J - 8 Hz, 2H), 7.21-7.25 (m, 1H), 7.31 (d, J - 8.40 Hz, 1H), 7.36 (d, J - 8.00 Hz, 1H), 7.66 (d, J - 8.40 Hz, 2H), 7.86 (s, 1H), 7.89 (s, 1H), 8.35 (s, 1H), 8.93 (s, 1H), 9.81 (s, 1H);
LCMS: m/e 348.3 (M+ 1).

[00472] Preparation of N-(4-methyl-3-(4-(pyridin-3-yl)pyrimidin-2-ylamino)phenyl) acrylamide 1-56 H
N\rN N`

/I
N

[00473] The title compound was prepared according to the schemes, steps and intermediates described below.
H
N N \ NHZ N\ N
\
N I/ step-1 N 0 A
N N

A) acryloyl chloride, Et3N, DMF, it, 12 h [00474] Step-1 H
N\/N N

I
N

[00475] 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 C 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) 6 ppm:
2.19 (s, 3H), 5.72 (dd, J- 2 & 10.08 Hz, 1H), 6.22 (dd, J- 2 & 16.92 Hz, 1H), 6.45 (dd, J -10 & 17 Hz, 1H), 7.16 (d, J- 8.36 Hz, t H), 7.32 (dd, J- 1.92 & 8.16 Hz, t H), 7.42 (d, J- 5.12 Hz, t 1l), 7.50-7.53 (m, 1H), 7.95 (d, J- 1.68 Hz, 1H), 8.45 (dd, J- 6.16 & 8.16 Hz, 1H), 8.49 (d, J- 5.16 Hz, 1H), 8.68 (dd, J- 1.56 & 4.76 Hz, 1H), 8.95 (s, 1H), 9.25 (d, J- 1.56 Hz, 1H), 10.08 (s, 1H);
LCMS: m/e 332.4 (M+1).

[004761 General method for preparing compounds having an enone-containing warhead, e.g., 3-methyl-l-(3-(5-methyl-2-(phenylamino)pyrimidin-4-ylamino)phenyl)but-2-en-l-one 1-47 NH
I \N N
H

[004771 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.

CI 0 01-1 / II IN + NH

\N CI / NH2 2 3 \N'CI

0 011~ 0 OH
O,NH2 NH KOH \
NH

N N
N

O N,i O
EDC/HOBT O ~--`MgBr NH

e'NH
HN'O~
N / \
~N N N
N H / I H

[004781 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.

[00479] Preparation of N-(3-(5-fluoro-2-(4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-182 HN N
F H
NNH

/O
OJr [00480] The title compound was prepared according to the schemes, steps and intermediates described below.

CI H2N I HxOk HN \ I N O H2N ~

N CI step-1 step-2 ~ZN

N \ NH2 F H
step-3 F I N / O~
N N~ 0~ C
H H

CI~~ HN \ N"

F
ste I ~ N /O ~
C N' `N \O
H

A) 2, DIPEA, THF, reflux; B) 4, t-amyl alcohol, HOAc, reflux; C) TFA, DCM; D) 7, DIPEA, THF, -10 C.

[004811 Step-1 HN N)L0 H
F
N! CI

[004821 1 (800mg, 4.8mmoL), 2 (996mg, 4.8mmoL) and Hunig's base (948uL, 5.75mmoL) were dissolved in THE (20mL). 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, lg.
LC/MS (RT -2.03/(M+1)) 339.1.
[004831 Step_2 F
N H
NINH
fo O

[004841 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 THE (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/(M+1)) 470.2. Additional 150 mg of product minus the (BOC) protecting group crystallized from the aqueous layer.

[004851 Sten-3 /I
HN \ NH2 F

NI NH

oJ( [004861 To a solution of 6 (550 mg, 1.17 mmol) in DCM (20 mL) was added TFA (2 mL).
Stirred for 30 min at rt for 4h; removed solvent via rotary evaporation and partitioned oil with cold (0 C) 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 combiflash system gave 309 mg of a light pink solid. LC/MS (RT -2.78/(M+l)) 370.2.
[004871 Sten-4 HN N
F H
N! NH

O
OJ[

[004881 A solution of 6 (309 mg, 0.84 mmol) in THE (10 mL) was cooled in a water/ice-MeOH bath (-10 C). To this was added 7 (71 L, 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 mL), 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.

[00489] Preparation of N-(3-(2-(3-chloro-4-(pyridin-2-ylmethoxy)phenylamino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-86 HN N
F H
XLNH

\ CI

N

[00490] 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.

[00491] Preparation of N-(3-(5-fluoro-2-(4-(2-(2-oxopyrrolidin-l-yl)ethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-92 HN N
F H
NNH

O
O N J[

[00492] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 1-(2-(4-aminophenoxy)ethyl)pyrrolidin-2-one in the place of 4 in Step 2. LC/MS (RT - 2.718/(M + H)) 477.1.

[00493] Preparation of N-(3-(5-fluoro-2-(4-(1-hydroxy-2-methylpropan-2-yloxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-93 HN N
F H
~N
N!NH

O
HO

[004941 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.

[004951 Preparation of N-(3-(5-fluoro-2-(6-isopropoxypyridin-3-ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-172 HN N
F H
NNH

N~
\/O

[004961 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.

[004971 Preparation of N-(3-(5-fluoro-2-(2-oxoindolin-5-ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-181 HN N
F H
X,LNH

i-NH
O

[004981 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.

[004991 Preparation of N-(2-chloro-5-(5-fluoro-2-(4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-108 CIO
HN N
F H
~N
N! NH

O
OJr [005001 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.

[00501] Preparation of N-(2-chloro-5-(5-fluoro-2-(6-isopropoxypyridin-3-ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-107 CIO
HN N
F H
N~NH
I
N

[00502] 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.

[00503] Preparation of N-(2-fluoro-5-(5-fluoro-2-(4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-87 F O
HN
F H
N~NH
O
OJ[

[005041 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)) 442Ø

[005051 Preparation of N-(3-(5-fluoro-2-(4-((l-methylpiperidin-4-yl)methoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-90 HN N
F H
~N
N~NH

O
N

[005061 The title compound was prepared according to the schemes, steps and intermediates described below.

CI H2N ON AO~ IN J~Oj\ H2N 4 F ~N F H
N
NLCI step-1 ! step-2 A N ~CI 3 B
HN N 0 ja F
N \ / O~N~ step-3 F ~ / O N
N N C N N
H H

CI HN \ N v F
step I N , O\~N
D N,N \
H

A) 2, DIPEA, THF, reflux; B) 4, Pd(OAc)2, X-Phos, CsC03, dioxane, reflux, 12 h; C) TFA, DCM; D) 7, DIPEA, THF, -10 C.
[005071 Step-1 \ N /\ k H
F
N
L/
N" 'CI

[005081 1 (800mg, 4.8mmoL), 2 (996mg, 4.8mmoL) and Hunig's base (948uL, 5.75mmoL) were dissolved in THF (20mL). 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+1)) 339.1.

[005091 Sten_2 ~~N 0,k HN
F H
IN
N~NH

N

[005101 3 (205 mg, 0.61 mmoL) and 4 (150 mg, 0.73 mmoL) was dissolved in dioxane (4 mL). 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+1)) 523.2.
[005111 Step-3 it HN \ NH2 F N

N 'NH
O

N

[005121 To a solution of 5 (144 mg, 0.27 mmol) in DCM (10 mL) was added TFA (1 mL).
Stirred for 30 min at rt for 12 h; removed solvent via rotary evaporation and partitioned oil with cold (0 C) 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.
[005131 Step-4 HN N
F H
~N
NNH

N

[005141 A solution of 6 (105 mg, 0.25 mmol) in THE (3 mL) was cooled in water/ice-MeOH
bath (-10 C). To this was added 7 (21 L, 0.26 mmoL), stirred for 10 min, then added Hunig's base (51 L, 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.

[00515] Preparation of N-(3-(5-fluoro-2-(6-((2-methoxyethyl)(methyl)amino)pyridin-3-ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-77 ~I
HN N
F H

NNH
N~
N
OJ( [00516] 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. LGMS (RT - 2,739/(M + H)) 438.1.

[00517] Preparation of 1-(6-(5-fluoro-2-(6-methoxypyridin-3-ylamino)pyrimidin-4-ylamino)-2H-benzo[b][l,4]oxazin-4(3H)-yl)prop-2-en-1-one 1-194 HN ,~N

F I ~ 0' 1 NI NH hIl N~

0.

[00518] The title compound was prepared according to the schemes, steps and intermediates described below.

'CO 0 N, O.
H2N N~ I
1 0 2 0 \ x H N

F ~N F
N O O
N CI step-I step-2 HN N HN
F H

1j 0 / step-3 0 N H/ 0' C N N
N H

O ~~
CI ~ ~ HN N

F , , step 0 - /

D N H \ / O
N

A) 2, DIPEA, THF, reflux; B) 4, HOAc, tert-amyl alcohol, reflux, 12 h; C) TFA, DCM; D) 7, DIPEA, DCM, NMP, -10 C.
[005191 Step-1 O
HN N
F N 0-~-Oj<
N 'CI

[005201 1 (186 mg, 1.1 mmoL), 2 (280mg, 1.lmmoL) and Hunig's base (220 L, 1.3 mmoL) were dissolved in THE (6 mL). 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.

[005211 Sten_2 O
HN \ N
F ;--j<
0I ~ 0 N NH

N
[005221 3 (215 mg, 0.56 mmoL) and 4 (83 mg, 0.66 mmoL) was suspended in tert-amyl alcohol (6 mL) 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 mL 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.
[005231 Sten-3 HN \ N
H
F

LN" _NH
0:11 1 N

[005241 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 C) 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 a pink solid. LC/MS (RT - 2.782/(M+l)) 369.1.
[005251 Step-4 HN N
F
--- N 0)11 LNLNH
0:11 1 N

[005261 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 C). To this was added 7 (34 L,, 0.43 mmoL), stirred for 10min, then added Hunig's base (70 L, 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.

[00527] Preparation of 1-(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 / o \~
HN N
F

N NH

[00528] 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.
LGMS (RT - 2.845!(M + H)) 466.2.

[00529] Preparation of 1-(6-(5-fluoro-2-(6-methoxypyridin-3-ylamino)pyrimidin-ylamino)indolin-l-yl)prop-2-en-l-one 1-166 HN / N
F

NNH

N

0N., [00530] The title compound was prepared according to the schemes, steps and intermediates described in Example 31, by using tert-butyl 6-aminoindoline-1-carboxylate in the place of 2 in Step 1. LC/MS (RT - 2.825/(M + H)) 407.1.

[00531] Preparation of 1-(5-(5-fluoro-2-(6-methoxypyridin-3-ylamino)pyrimidin-ylamino)isoindolin-2-yl)prop-2-en-l-one 1-165 HN

F N
LN~NH
NN1%

0IN.

[00532] 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.

[00533] Preparation of 1-(6-(4-(3-chlorophenylamino)-5-fluoropyrimidin-2-ylamino)-2H-benzo[b][ l,4]oxazin-4(3H)-yl)prop-2-en-l-one 1-149 HN CI
F -_ N / I O
N N N
H
O

[005341 The title compound was prepared according to the schemes, steps and intermediates described below.

~ N
/ H2Ni I
~
I ~ CI I 0 'Z 0 I H2N k N F ~
NCI step-1 N step-2 A N CI B

~I
HN CI HN/ CI
F / 0 F , 0 I step-3 I I / II
N H O N N N
0 01~ H 6 H
o ~I

7 F , / 0 step -4 Z~Nll D N N N
H

A) 2, DIPEA, THF, reflux; B) 4, HOAc, tert-amyl alcohol, reflux, 12 h; C) TFA, DCM; D) 7, DIPEA, THF, -10 C.
[005351 Step-1 HN CI
F
N
LNCI

[005361 1(484 mg, 2.9 mmoL), 2 (305 mg, 2.9 mmoL) and Hunig's base (526 L, 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.
[00537] Step_2 HN CI

N N N
H
O)--~ 0 ,1~
[00538] 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.
[00539] Step-3 HN CI
F / O
/
N N N
H H

[00540] 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 C) 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.

[00541] Sten-4 HN CI
F ~N / I 0 NN N
H

[00542] A solution of 6 (150 mg, 0.4 mmol) in THE (3 mL) was cooled in water/ice-MeOH
bath (-10 C). To this was added 7 (34 L, 0.42 mmoL), stirred for 10 min, then added Hunig's base (70 L, 0.42 mmoL), and stirred for 10min. 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.

[00543] Preparation of 5-(2-(4-acryloyl-3,4-dihydro-2H-benzo[b][1,4]oxazin-6-ylamino)-5-fluoropyrimidin-4-ylamino)indolin-2-one 1-130 O
HN
/I

~ NH O N
F I O
N

N" _N
H

[00544] 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.

[00545] Preparation of 4-(3-acrylamidophenylamino)-2-(phenylamino)pyrimidine-5-carboxamide 1-230 0 HN "'a N H

H2N yoo~
~ NN 'O

[00546] The title compound was prepared according to the schemes, steps and intermediates described below.

=0 NH2 0 CI H2N I NA0 H
CI `N 2 NN 4 N CI step-1 Me0 I / H I NCI step-2 J, 0 0 HN H O a6 NH2 ~I

~N H
\\~ I
MeO H N CI step-3 N N
C MeC " N N"0 0 HN \ NH2 CI 9 0 HN/H v step-4 y \ \ \
I j H~ Step -5 H \ /
MeO N N"\\~ / E Me0 N N'O

o 0 HN \ N" v H
step-6 H2N `N
F N" -N O
H

A) 2, NEt3, DCM, 0 C 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, THE, 0 C; F) TFA, TfOH, DCM, rt.

[005471 Step-1 H N
JOI""' O NCI

[005481 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 mL) and chilled in an ice/water bath (0 C). 2 (309 L, 2.4 mmoL) was added and the mixture stirred for 10 min. Triethylamine (365 L, 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. LGMS (RT - 2.789/(M+l)) 312.
[005491 Step_2 0 HN \ NAO
H
~N
I H NJL%jj O \ NCI
[005501 3 (170 mg, 0.55 mmoL), 4 (113 mg, 0.55 mmoL) and Hunig's base (108 L, 0.65 mmoL) were dissolved in THE (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+l)) 484.
[005511 Step-3 \ 0 H I/

LN NH

[005521 5 (230 mg, 0.48 mmol), 6 (126 L, 1.4 mmoL) and Hunig's base (94 L, 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+l)) 541.2.
[005531 Step-4 \ I H
O N,LNH

[005541 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
(1N, 25mL).
Agitated, precipitate formed, filtered and dry to give a white solid. LC/MS
(RT - 2.934/(M+l)) 441.1.
[005551 Step-5 \ 0 0 HN I / N" v H
\ I H I \N
O NNH

[005561 A suspension of 8 (130 mg, 0.29 mmol) in THE (6 mL) was cooled in water/ice (0 C). To this was added 9 (25 L (plus additional 5 L), 0.38 mmoL (total)), then added triethyl amine (43 L (plus additional 11 L), 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.

[005571 Sten-6 \ 0 0 HN I / N v H

NINH

[005581 To a suspension of 1-231 (30 mg, 0.061 mmol) in DCM (4 mL) was added TFA (200 L) and triflic acid (68 L, 0.6lmmoL). Stirred at rt 1 h. Removed solvent under reduce pressure via rotary evaporation and partitioned with cold (0 C) 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 - 2.7151(M + H)) 375.1.

[005591 Preparation of 4-(3-acrylamidophenylamino)-N-phenyl-2-(phenylamino)pyrimidine-5-carboxamide 1-222 O HN \ N" v H
N
H Y'O"~
N
N
H

[005601 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 - 2.99l/(M + H)) 451.2.

[00561] Preparation of 4-(3-acrylamidophenylamino)-N-cyclopropyl-2-(phenylamino)pyrimidine-5-carboxamide I-221 0 HN \ N"
H
&NAfvLN /
H N \
H

[00562] 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.

[00563] Preparation of 4-(3-acrylamidophenylamino)-2-(3-methoxyphenylamino)pyrimidine-5-carboxamide 1-210 / OO
0 HN \ N"
H

H2N Y'~ NNH

[00564] The title compound was prepared according to the schemes, steps and intermediates described in Example 37, by using 3-methoxyaniline in the place of 6 in Step 3. LC/MS (RT
2.743/(M + H)) 405.1.

[00565] Preparation of 4-(3-acrylamidophenylamino)-2-(6-methoxypyridin-3-ylamino)pyrimidine-5-carboxamide 1-209 0 HN N"
H
H2N I ~~
N N NH

N z ON.11-209 [00566] 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.

[00567] Preparation of 1-{6-[5-Acetyl-2-(6-methoxy-pyridin-3-ylamino)-pyrimidin-4-ylamino]-2,3-dihydro-benzo[1,4]oxazin-4-yl}-propenone 1-170 O HN N lkl~
H

N NH
N

[00568] The title compound was prepared according to the schemes, steps and intermediates described below.

o 0 0 ~I
CI H ZN I N HN I N" . 0 HN \ N"
Br N 2 Boc Br Boc Bu3Sn 4 OR
\ Boc NCI step-1 NCI step-2 N'CI step-3 / I 0 1 x0 / 0 H2N
J r CI I 0 HN aN
0 HN" v NJ 7 0 HN" v N N OMe \
H 9N o N
step -4 N 0 step-5 N NH
N CI D
6 ry 8 CI E 1-170 I N OMe A) 2, DIPEA, THF, 70 C, 16 h; B) (a) 4, PdC12(PPh3)2, DMF, 70 C; (b) 1N HCl, acetone, 60 C, 15 min; C) HCI/dioxane, DCM; D) 7, DIPEA, NMP, DCM, -20 C to rt; E) 9, pTsOH, dioxane, 100 C, 15 min.
[005691 Step-1 Jl HN N
Br Boc NNCI

[005701 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 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-).

[00571] Sten_2 Jl Boc AIN-CI
[00572] 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 C
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.
[00573] After filtration and concentration, the residue was dissolved in 20 mL
of acetone, followed by additon of 3 mL of 1.0 N aqueous HC1 solution. The mixture was heated at 60 C for 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-).
[00574] Sten-3 O HN N
H
N
NCI

[00575] To a mixture of 1.28 g of intermediate 1-2 in 10 mL of dichloromethane, was added 10 mL of 4.0 N HC1 in dioxane. After stirring at it overnight, the solvent was removed, and the residue was dried in vacuum. LC-MS: m/z 305.1 (ES+), 303.1 (ES-).

[005761 Step-4 O HN' N
N O

N CI

[005771 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 C, 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-).
[005781 Step-5 N
0 HN ~`~
~N O' N-NH
H

N OMe [005791 The mixture of 30 mg intermediate 8 (84 mol) and 13 mg of 9 (1.2 equiv) in 1 mL
of 0.08 M p-TsOH dioxane solution was heated at 100 C 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-).

[00580] Preparation of 1-{6- [5-Acetyl-2-(4-morpholin-4-yl-phenylamino)-pyrimidin-4-ylamino]-2,3-dihydro-benzo[1,4]oxazin-4-yl}-propenone 1-169 ~
O HN, N
NNH

N~

[00581] 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-).

[00582] Preparation of 1-{6-[5-Acetyl-2-(6-morpholin-4-yl-pyridin-3-ylamino)-pyrimidin-4-ylamino]-2,3-dihydro-benzo[1,4]oxazin-4-yl}-propenone 1-168 0\
J
O HN N

N O' NNH n N N

[00583] 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-).

[00584] Preparation of 1-{6-[5-Acetyl-2-(1-methyl-lH-indazol-6-ylamino)-pyrimidin-4-ylamino]-2,3-dihydro-benzo[1,4]oxazin-4-yl}-propenone 1-154 Jl N'%H
N
~N

[00585] The title compound was prepared according to the schemes, steps and intermediates described in Example 42, by using 1-methyl-1H-indazol-6-ylamine in the place of 9 in Step 5.
LC-MS: m/z 470.1 (ES+), 468.1 (ES-).

[00586] Preparation of 1-{6-[5-Acetyl-2-(lH-indazol-6-ylamino)-pyrimidin-4-ylamino]-2,3-dihydro-benzo[1,4]oxazin-4-yl}-propenone 1-153 Jl N 0' H
NNH ~~ N\
II N

[00587] The title compound was prepared according to the schemes, steps and intermediates described in Example 42, by using 1H-indazole-6-ylamine in the place of 9 in Step 5. LC-MS:
m/z 456.1 (ES+), 454.2 (ES-).

[00588] Preparation of 1-{4-[5-Acetyl-4-(4-acryloyl-3,4-dihydro-2H-benzo[1,4]oxazin-6-ylamino)-pyrimidin-2-ylamino]-phenyl}-pyrrolidin-2-one 1-152 0 HN,XN

N~-NH

[00589] The title compound was prepared according to the schemes, steps and intermediates described in Example 42, by using 1-(4- amino-phenyl) -pyrrolidin-2- one in the place of 9 in Step 5. LC-MS: m/z 456.1 (ES+), 454.2 (ES-).

[00590] Preparation of 1-(6-{5-Acetyl-2-[4-(2-methoxy-ethoxy)-phenylamino]-pyrimidin-4-ylamino}-2,3-dihydro-benzo[1,4]oxazin-4-yl)-propenone 1-150 O HN aN
N 0' v N ~NH

0--\\--0 [00591] 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-).

[00592] Preparation of 5-[5-Acetyl-4-(4-acryloyl-3,4-dihydro-2H-benzo[1,4]oxazin-6-ylamino)-pyrimidin-2-ylamino]-1,3-dihydro-indol-2-one 1-129 O\
J
O HN N

N 0' N ~NHIlll) N
H

[00593] 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-).

[00594] Preparation of 1-(6-{5-Acetyl-2-[6-(2-hydroxy-ethoxy)-pyridin-3-ylamino]-pyrimidin-4-ylamino}-2,3-dihydro-benzo[1,4]oxazin-4-yl)-propenone 1-128 O\
J
O HN N

N 0' N~NH ~
~ N
H

[00595] 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-).

[00596] Preparation of N-{3-[5-Acetyl-2-(6-methoxy-pyridin-3-ylamino)-pyrimidin-4-ylamino]-phenyl}-acrylamide 1-189 N
N~NH

N O

[00597] 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-).

[00598] Preparation of N-{3-[5-Acetyl-2-(6-methoxy-pyridin-3-ylamino)-pyrimidin-4-yloxy]-phenyl} -acrylamide 1-188 H
N
N~NH

[00599] 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-).

[00600] Preparation of 1-{3-[5-Acetyl-2-(6-methoxy-pyridin-3-ylamino)-pyrimidin-4-ylamino]-azetidin-l-yl}-propenone 1-187 N
N~NH

N O

[00601] 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-).

[00602] Preparation of N-(3-{5-Acetyl-2-[4-(2-methoxy-ethoxy)-phenylamino]-pyrimidin-4-ylamino}-phenyl)-acrylamide 1-124 O HN H
N
N~NH
0,-,,,~0\

[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 4-(2-methoxy-ethoxy)-phenylamine in the place of 9 in Step 5. LC-MS: m/z 448.2 (ES+), 446.3 (ES-).

[00604] Preparation of N-(3-{5-Acetyl-2-[6-(2-methoxy-ethoxy)-pyridin-3-ylamino]-pyrimidin-4-ylamino}-phenyl)-acrylamide 1-122 O HN H
N
N~NH

N 0\i0\

[00605] 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-).

[00606] Preparation of N-(3-{5-Acetyl-2-[6-(2-hydroxy-ethoxy)-pyridin-3-ylamino]-pyrimidin-4-ylamino}-phenyl)-acrylamide 1-121 O HN H
N
N~NH

N O,-~O H

[00607] 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-).

[006081 Preparation of 4-(3-acrylamidophenoxy)-2-(3-methoxyphenylamino)-pyrimidine-5-carboxylic acid phenylamide 1-200 \ H
N
H 'J.
N N We H

[006091 The title compound was prepared according to the schemes, steps and intermediates described below.

\ /
/ .Boc O 0 \ I N.Bac HO N H
Et-, U N 2 N H Et~O N
"
S step 1 NS step 2 Boc H2N 5 \ I Boc HO I step 3 H N step 4 N S C N S D

.Boc H2N OMe 0 O N"Boc I
H ~0 ~ H N / step 6 N S step 5 NN \ OMe F
ii E H

\ I yCI /

N I \ I step 7 \ H
N N N OMe G N N OMe A) 2, NaH, THF, 0 C; B) NaOH, THF, MeOH; C) 5, TBTU, DIPEA, CH3CN, 0 C; D) MCPBA, CH2C2, 0 C; E) 8,50T, 3 h; F) TFA, CH2C12; G) 11, DIPEA, CH2C12 [006101 Step 1 0 O \ I N.Boc ERO N H
N!

[006111 To a stirred solution of (3-hydroxyphenyl)carbamic acid tert-butyl ester 2 (1.79 g, 8.59 mmol) at 0 C was added a suspension of sodium hydride (60% dispersion in mineral oil) (0.34 g, 8.9 mmol) in anhdyrous THE (30 mL). The mixture was stirred at 0 C
for 20 minutes.

The phenoxide solution was then added dropwise at 0 C to a solution of 4-chloro-(2-methylsulfanyl)pyrimidine-5-carboxylic acid ethyl ester 1 (2 g, 8.59 mmol) in THE (20 mL).
The reaction mixture was stirred at 0 C 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 CH2C12:hexane (1:9) to afford the title compound 3 as a white solid (2.43 g, 70%).
[00612] Step_2 0 O \ I NBoc H
HO I ~N

NS

[00613] To a stirred solution of 4-(3-tent-butoxycarbonylaminophenoxy)-2-(methylsulfanyl-pyrimidine)-5-carboxylic acid ethyl ester 3 (2 g, 4.93 mmol) in THE (60 mL), was added methanol (60 mL) at -10 C, 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%).
[00614] Sten-3 0 O \ I NBoc \ H
H /N
N~S

[00615] To a stirred solution of 4-(3-tent-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 0 C was added DIPEA (1.36g, 10.6 mmol) followed by aniline (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%).

[006161 Sten-4 Boc H

H N
NS
11 [00617] To a stirred solution of [3-(2-methylsulfanyl-5-phenylcarbamoylpyrimidin-4-yloxy)-phenyl]-carbamic acid tent-butyl ester 6 (1.5 g, 3.31 mmol) in CH2,C12 at 0 C
was added a solution m-CPBA (70%, 1.62 g, 2 eq) in CH2C12 (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 CH2C12:hexane (1:9) to afford the title compound 7 as a white solid (1.16 g, 73%).
[006181 Step-5 0 O \ I N,Boc H
N ~N ~
H \
N N OMe H

[00619] Excess 3-methoxyaniline (8) (2 mL) was added to solid [3-(2-methanesulfonyl-5-phenylcarbamoyl-pyrimidin-4-yloxy)-phenyl]-carbamic acid tert-butyl ester 7 (0.5 g, 1.03 mmol) and the resulting mixture was heated to 50 C 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).
[00620] Step-6 II 0 O \ NH2 H \I
N N OMe H

[00621] To a solution of {3-[2-(3-methoxy-phenylamino)-5-phenylcarbamoyl-pyrimidin-4-yloxy]-phenyl}-carbamic acid tent-butyl ester 9 (0.3 g, 0.56 mmol) in CH2C12 (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 CH2C12, washed with 10% aqueous NaHCO3 solution, dried (Na2SO4), filtered, and evaporated under reduced pressure to provide the free amine 10 as white solid.
[00622] Step_7 H
N N
H
N N We H

[00623] To a stirred solution of amine 10 (0.24 g, 0.56 mmol) in dichloromethane (20mL) under argon atmosphere cooled to -70 C 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 C for 5 minutes, and the reaction mixture diluted with CH2C12 (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-CHC13 5:95) as eluent to provide the target compound 11 (0.094 g, 35%) as white solid: 'H NMR (200 MHz, DMF-d7) 6 8.9 (s, 1H), 8.10-7.70 (m, 6H), 7.60-7.10 (m, 6H),6.60 (m, 2H) 6.40 (dd, 1H, J- 8.0, 2.0 Hz ), 5.80 (m, 2H), 3.70 (s, 3H).

[00624] Preparation of 4-(3-acrylamidophenoxy)-2-(6-methoxypyridin-3-ylamino)-pyrimidine-5-carboxylic acid phenylamide 1-159 H
H I N \ N
NN
H

[006251 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. 'H NMR
(200 MHz, DMSO-d6 6 8.90 (s, 1H), 8.20 (brs, 1H), 7.90-7.60 (m, 4H), 7.45(m, 4H), 7.10 (m, 2H), 6.50 (m, 1H), 6.20 (m, 2H), 5.90 (dd, J- 8.0, 2.0 Hz, 1H), 3.90 (s, 3H).

[006261 Preparation of 4-(3-acrylamidophenoxy)-2-(3-methoxyphenylamino)-pyrimidine-5-carboxylic acid cyclopropylamide 1-177 O O H

N N OMe H

[006271 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. 'H
NMR (200 MHz, CD30D) 6 9.0 (s, 1H), 7.90 (brs, 1H), 7.50 (m, 3H), 7.0 (m, 4H), 6.50 (m, 1H), 6.40 (d, J- 8.0 Hz, 2H), 5.80 (dd, J- 8.2, 3.0 Hz, 1H), 3.60 (s, 3H), 0.90 (m, 2H), 0.62 (m, 2H).

[006281 Preparation of 4-(3-acrylamidophenoxy)-2-(6-methoxypyridin-3-ylamino)-pyrimidine-5-carboxylic acid cyclopropylamide 1-176 N I 0~
H N
N N
H

[006291 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. 'H NMR (200 MHz, CD30D) 6 8.90 (s, 1H), 7.95 (brs, 1H), 7.90-7.82 (m, 3H), 7.40 (m, 3H), 6.98 (d, J - 6.0 Hz, 1H), 6.42 (m, 2H), 5.90 (dd, J - 8.0, 2.0 Hz, 1H), 3.90 (s, 3H), 0.95 (m, 2H), 0.83 (m, 2H).

[006301 Preparation of 4-(3-acrylamidophenoxy)-2-(3-methoxyphenylamino)pyrimidine-5-carboxylic acid amide 1-178 ZIIN

H

N N \ We H

[006311 The title compound was prepared according to the schemes, steps and intermediates described below.

0 CI HO I/ NBoc 0 O\ N Boc Et,O 2 H Et.O H

NI SI step 1 N,S step 2 \ OMe Boc , Boc H ~ H
HO N step 3 HN step 4 NISI D
N~S C I \ 6 4 MeO
/ I \ I Boc \ Boc H2N OMe 0 0 N' step 6 \ NIS step 5 HN
NN \ I OMe \ 9 ii E / H F
MeO I 7 0 MeO

~C1 0 0 \ NH2 \ I ~%

HN XNOM s t HN step 8 e G\ N H OMe H
Me0 10 MeO 11 H
H2N ~ /
N N \ OMe H

A) 2, NaH, THF, 0 C; B) LiOH, THF, H20; C) 5, TBTU, DIPEA, CH3CN; D) MCPBA, CHC130 0 C; E) 8, DMA, 90 C, 24 h; F) 4N HCI, dioxane; G) 11, CH2C12; H) triflic acid, TFA, CH2C12 [006321 Step-1 0 O I NBoc Et,, H

[006331 Step 1 was carried out in a manner similar to Step 1 in Example 57.
[006341 Step_2 N,Boc H

NiS

[006351 Saponification of 3 (4.58 g, 11.3 mmol) by LiOH (500 mg, 20 mmol) in 80 mL
THE/H20 (1:1) and usual workup with 1 N HC1 gave free acid 4.
[006361 Step-3 N Boc .
H
HN I

NJ S
Me0 / 6 [006371 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).
[006381 Step-4 N,Boc H
HN N

Me0 [00639] Step 4 was run in a manner similar to Step 4 in Example 57 with CHC13 being substituted for CH2C12 as the solvent.
[00640] Step-S

N,Boc H
HN ~ /

N H \ OMe MeO g [00641] The 2-methylsulfone of 7 (1.0 g, 1.9 mmol) was mixed with 3-methoxyaniline (420 mg, 3.4 mmol) in DMA and the mixture was heated at 90 C 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).
[00642] Steps-6, 7, and 8 ~I

H

N N OMe H

[00643] 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 L, 0.52 mmol) in 15 mL DCM at -40 C. 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 L, 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.

[00644] Preparation of tert-butyl 3-(3-(4-(3-acrylamidophenylamino)-5-methylpyrimidin-2-ylamino)phenoxy)propylcarbamate 1-45 ~I
HN N

H 'I N N H 0 H N 'k 0 [00645] The title compound was prepared according to the schemes, steps and intermediates described below.

J~ k 3 HONO step-1 - MsO-I~NA0 H s A

O N \ I ON~O H2N \ I ON 0~
z H step-3 H

HN' v `NOz H

N CI step-4 step -5 p N CI E

/ / CI"

HN \ NOZ HN \ NHZ 11 ~ SteF 6 I % \ 0 stepC 7 H H H H

HN N
H

H H

A) 1, methanesulfonyl chloride, CH2Cl2, Et3N, rt, 1 h; B) 3, K2CO3, DMF, 60 C; C) H2, Pd/C, EtOH, rt, 16 hr; D) 6, 7, Pd(OAc)2, BINAP, Cs2CO3, toluene, 100 C, 16 hr; E) 5, AcOH, EtOH, 90 C, 16 hr; F) H2, Pd/C, EtOH, rt, 16 hr; G) 11, NMP, 0 C, 15 min [006461 Step-1 MsO~~N 0 H

[006471 To a stirring solution of 1 (1.0 g, 5.7 mmol) in dichloromethane (20.0 mL) 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 sole. (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.
[006481 Step_2 02N ~~0 H 0k [006491 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 C
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.
[006501 Sten-3 az~10 H A

[006511 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.

[00652] Step-4 HN \ NO2 N! C1 [00653] To a solution of 6 (1.69 g, 12.26 mmol), in toluene (50.0 mL) 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 C 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 (Si02, 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.
[00654] Step-5 HN \ NO2 N N ao-~~N O
H H

[00655] 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 C. The reaction mixture was cooled, concentrated under reduced pressure. The residue was quenched with 10% sodium bicarbonate sole. (10.0 mL) and extracted with ethyl acetate (3xl5 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 (Si02, EtOAc/Hexane: 50/50) to get 9 (0.57 g, 61%) as a light yellow solid.

[006561 Sten-6 I ~. H H
[006571 To a solution of 9 (0.56 g, 1.13 mmol) in ethanol (25 mL)) 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.
[006581 Step-7 ~I
HN N
H
N H 0 H N 'k 0 [006591 To a stirred solution of 10 (0.25 g, 0.5382 mmol) in NMP (2.5 mL) at 0 C was added acryloyl chloride (0.073 g, 0.807 mmol) and the reaction mixture was stirred at 0 C 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 C, 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. 'H NMR (DMSO-d6) 6 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, 1H), 6.24 (dd, J- 1.84 & 15.16 Hz, 1H), 6.35-6.47 (m, 2H), 6.80-6.90 (bs, 1H), 6.97 (t, J- 8.28 Hz, 1H), 7.23-7.27 (m, 2H), 7.31 (s, 1H), 7.37 (d, J- 8.2 Hz, 1H), 7.46 (d, J- 7.48 Hz, 1H), 7.90-7.90-7.91 (m, 2H), 8.36 (s, 1H), 8.87 (s, 1H), 10.07 (s, 1H);
LCMS: m/e 519 (M+l).

[006601 Preparation of tert-butyl 3-(3-(4-(3-acrylamidophenylamino)-5-fluoropyrimidin-2-ylamino)phenoxy)propylcarbamate 1-183 ~I
HN N
F H

N H O H O

[006611 The title compound was prepared according to the schemes, steps and intermediates described below.

2' H

A step-1' / 0 step-2' a V
~ I + MsO,~\N~Ox J
B ~\

C step-3 X
~~H
H2N 0~ 0 A) methanesulfonyl chloride, CH2C12, Et3N, rt, 1 h; B) K2CO3, DMF, 60 C, 16 h; C) Pd-C, H2, ethanol, rt, 16 h.
[006621 Step 1' 0 u MsO"~ NId, O" \

[00663] 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.
[00664] Step 2' ~~O
OpN "'~H ~OX

[00665] 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 C 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.
[00666] Step 3' ~~0~H ~0X

[00667] 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.

CI H2N I / N02 HN \ I N02 H2N O N~O \ I

F--~- step-1 F N step-2 NI / R

C l step-3 HN \ I N HN \ I NH2 H
step-4 F
N N\ I O~\N II O~ D N N\ I C"-O~

[006681 Step 1 F ~N
N! CI

[006691 A pressure tube was charged with 2 (10.0 g, 0.072 mot), 1 (24.1 g, 0.145 mot), n-BuOH (100 mL) and DIPEA (13.9 g, 0.108 mol), and the contents were stirred at 120 C 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.
[006701 Step 2 HN \ N02 F ,. a 0 N N O N A O X
H H

[006711 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 C for 48 h. The reaction mixture was cooled and concentrated under reduced pressure; the residue was quenched with 10% sodium bicarbonate solution (20.0 mL) 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 A1203, McOH/Chloroform:
0.5/99.5) to give 5 (1.4 g, 50.3%) as a brown solid.
[00672] Step 3 HN \ NH2 FI!~I

[00673] 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 A1203, 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.
[00674] Step 4 HN N
F H

H H

[00675] 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 C was added acryloyl chloride (0.060 g, 0.665 mmol), and the reaction mixture was stirred at 0 C for 30 min. The reaction mixture was added dropwise to a cold, stirring solution of 10% NaHCO3 and stirred at the same temperature (0 C) 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. 'H NMR (DMSO-d6) 6 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, I
H), 6.24 (d, J =
16.84 Hz, 1H), 6.41-6.48 (m, 2H), 6.88 (s, 1H), 7.03 (t, J= 8.24 Hz, 1H), 7.23-7.31 (m, 3H), 7.41 (d, J= 8.28 Hz, 1H), 7.56 (d, J= 7.96 Hz, 1H), 7.90 (s, 1H), 8.11 (d, J=
3.56 Hz, 1H), 9.11 (s, 1H), 9.43 (s, 1H), 10.10 (s, 1H); LCMS : m/e 523.1 (M+1).

[006761 Preparation of tert-butyl 3-(4-(4-(3-acrylamidophenylamino)-5-fluoropyrimidin-2-ylamino)phenoxy)propylcarbamate 1-198 F N / 0'/~N0 H
N N
H

[006771 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. iH NMR (DMSO-d6) 6 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, 1H), 6.24 (dd, J- 1.92 & 16.92 Hz, 1 H), 6.45 (dd, J = 10.08 & 16.92 Hz, 1 H), 6.72 (d, J = 9 Hz, 2H), 6.89 (t, J - 5.4 Hz, 1 H), 7.26 (t, J- 8.08 Hz, 1H), 7.40 (d, J- 8.12 Hz, 1H), 7.48-7.52 (m, 3H), 7.92 (s, 1H), 8.05 (d, J=
3.72 Hz,1H), 8.95 (s, 1H), 9.36 (s, 1H), 10.12 (s, 1H); LCMS : m/e 523.2 (M+1).
[006781 The intermediate tert-butyl 3-(4aminophenoxy)propylcarbamate was prepared by the scheme shown below.

~~
0 1 1 < step -1 J
0 N 0 step-2 H2N \

H

A) NaH, THF, rt, 16 h; B) H2, Pd/C, EtOH, rt, 16 hr [006791 Step-1 [006801 To a stirring solution of 1 (1.7 g, 9.7 mmol) in dry THE (40 mL) was added NaH
(0.72 g, 18.0 mmol, 60% dispersion in paraffin oil) at 0 C 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 (2.0g, 69.5%) as a yellow crystalline solid.
[006811 Step_2 [006821 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.

[006831 Preparation of 4-(3 acrylamidophenylamino)-5-fluoro-2-(3,4-dimethoxyphenylamino)-pyrimidine 1-134 a HN N
H
F , /I 0, H

[006841 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.
'H NMR (200 MHz, CD30D) 6 8.50 (s, 1H), 7.80 (d, J- 6.5 Hz, 1H), 7.70-7.66 (m, 2H), 7.20 (m, 1H), 7.0 (m, 2H), 6.41 (m, 2H), 5.92 (dd, J- 8.0, 2.0 Hz, 1H), 3.89 (s, 6H).

[006851 Preparation of 4-(3-acrylamidophenylamino)-5-fluoro-2-(3,4,5-trimethoxyphenylamino)-pyrimidine 1-133 HN

HN
"
F N O

NN \ O
H

[006861 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. 'H NMR (200 MHz, CD30D) 6 8.10 (s, 1H), 8.0 (d, J- 6.0 Hz, 1H), 7.50 (m, 2H), 7.30 (m, 1H), 7.0 (m, 2H), 6.45 (m, 2H), 5.90 (dd, J- 8.0, 2.0 Hz, 1H), 3.90 (s, 3H), 3.89 (s, 9H).

[00687] Preparation of 4-(3-acrylamidophenylamino)-5-fluoro-2-(3-(hydroxymethyl)phenylamino)-pyrimidine 1-145 F H
NIJ \
NJ N I OH
H

[00688] 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-d6) 6 ppm: 4.38 (d, J - 5.6 Hz, 2H), 5.07 (t, J - 5.68 Hz, 1H), 5.75 (d, J - 10.84 Hz, I H), 6.24 (dd, J- 16.96 Hz, 1H), 6.44 (dt, J- 10.04 & 17.0 Hz, 111), 6.83 (d, J- 7.4 Hz, 111), 7.10 (t, J- 7.72 Hz, 1H), 7.28 (t, J- 8.16 Hz, 1H), 7.40 (d, J- 8.08 Hz, 1H), 7.55-7.59 (m, 3H), 7.92 (s, 1H), 8.09 (d, J- 3.6 Hz, 1H), 9.11 (s, 1H), 9.40 (s, 1H), 10.1 (s, 1H); LCMS : m/e 378.0 (M+ 1).

[00689] Preparation of 4-(3-acrylamidophenylamino)-5-fluoro-2-(3-(3-(2-oxopyrrolidin-l-yl)propoxy)phenylamino)-pyrimidine 1-144 HN N
F H
N N N
H

[00690] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 3-(2-oxopyrrolidin-1-yl)propoxyaniline in place of 4 in Step-2. 1H NMR (DMSO-d6) 6 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, 1H), 6.24 (d, J- 15.64 Hz, 1H), 6.41-6.80 (m, 2H), 7.04 (t, J- 8.16 Hz, 1H), 7.22-7.29 (m, 2H), 7.33 (s, 1H), 7,42 (d, J- 8.08 Hz, 1H), 7.55 (d, J-7.52 Hz, t H), 7.91 (s, t 1l), 8.11 (d, J- 3.48 Hz, t H), 9.13 (s, t H), 9.43 (s, t H), 10.11 (s, t 1l);
LCMS : m/e 491 (M+1).

[006911 Preparation of 4-(3-acrylamidophenylamino)-5-fluoro-2-(3-(3-(methylsulfonyl)propoxy)phenylamino)-pyrimidine 1-138 HN N
F H
NIN /

LN NO'S"
H II

[006921 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.
iH NMR (DMSO-d6) 6 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, 1H), 6.25 (dd, J- 1.8 & 16.88 Hz, 1H), 6.44 (dd, J
= 10.16 & 16.84 Hz, 2H), 7.05 (t, J= 8.16 Hz, 1H), 7.24-7.30 (m, 2H), 7.35 (s, 1H), 7.42 (d, J=
8.2 Hz, 1H), 7.55 (d, J= 8 Hz, 1H), 7.90 (s, 1H), 8.11 (d, J= 3.6 Hz, 1H), 9.14 (s, 1H), 9.43 (s, 1H), 10.10 (s, 1H); LCMS : m/e 484 (M+1).
[006931 The intermediate 3-(3-(methylsulfonyl)propoxyaniline was prepared by the scheme shown below.

HOSV

\ step-1 step-2 Boc.N I / OH A Boc.N JaOS B

s tep-3Boc.H 0S, /
Ja A) DEAD, Ph3P, Et3N, THF, rt, 1 hr; B) MCPBA, CH2C12, rt, 30 min; C) TFA, CH2C12, rt, 1 hr [006941 Step-1 (BOC)HN I 0^~S-1' [006951 To a stirred solution of 2 (1.1 g, 10.3 mmol) in THE (20 mL) 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 C 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 (Si02, 60-120, pet ether/ethyl acetate, 8/2) to get 3 (2 g 60.6%) as a white solid.
[006961 Step_2 (COB)HN 4 O OS 0 [006971 To a stirred solution of 3 (2 g, 6.7 mmol) in CH2C12 (25 mL) was added m-CPBA
(4.13 g, 26.7 mmol) at -10 C. The reaction mixture was allowed to come to rt and stirred for 30 min. It was quenched with Na2CO3 solution (10 mL), extracted with CH2C12 (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was further purified by column chromatography (Si02, 60-120, chloroform/methanol 9/1) to get 4 (1.05 g, 68.8%) as a yellow oil.
[006981 Step-3 O
[006991 To a stirred solution of 4 (0.75 g, 2.2 mmol) in CH2C12 (7.5 mL) was added TFA (3 vol.) at 0 C. 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 CH2C12 (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.

[00700] Preparation of N-(3-(5-fluoro-2-(3-(2-hydroxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-105 HN N

F , NN O~~OH
H

[00701] 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-d6) 6 ppm: 3.67 (dd, J - 4.5 & 10 Hz, 2H), 3.85-3.87 (m, 2H), 4.83 (t, J
- 5.6 Hz, 1H), 5.75 (bd, J - 10 Hz, 1 H), 6.25 (d, J - 15.6 Hz, 1 H), 6.42-6.46 (m, 2H), 7.05 (t, J - 8.4 Hz, 1 H), 7.26 (d, J- 8 Hz, 1H), 7.30 (d, J- 8 Hz, 1H), 7.33 (s, 1H), 7.41 (d, J- 8 Hz, 1H), 7.57 (d, J- 8 Hz, 1 H), 7.92 (s, 1 H), 8.11 (d, J - 3.6 Hz, 1 H), 9.11 (s, 1 H), 9.42 (s, 1 H), 10.11 (s, 1 H); LCMS
m/e 409.9 (M+1).
[00702] The intermediate 3-(2-hydroxy)ethoxyaniline was prepared by the scheme shown below.
Br-CH2COOEt step-1 step-2 O N I OH 02N I 0 OEt step-3 \
OD O
H ~iOH
C zNI a zN 0 " H

A) K2CO3, DMF, 70 C, 12 h; B) Pd-C, H2, ethanol, rt, 10 h; C) 1M LAH
solution, THF, -15 C, 45 min.
[00703] Step-1 02N I O'*'-r OB

[007041 To a stirring solution of 1 (2.0 g, 14.37 mmol) and K2C03 (3.95 g, 28.6 mmol) in dry DMF (15 mL) was added 2 (2.88 g, 17.25 mmol) and the reaction was stirred at rt 70 C 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.
[007051 Step_2 H2N I O^ /OEt [007061 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.
[007071 Step-3 H 2N I a Oi~OH
[007081 To a stirring solution of 4 (1.2 g, 6.14 mmol) in dry THE (12 mL)) was added lithium aluminum hydride (9.2 mL, 9.20 mmol, 1.0 M soln. in THF) at -15 C, 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.

[00709] Preparation of N-(3-(5-fluoro-2-(3-(2-hydroxy-2-methylpropoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-118 HN \IN
F H
~N \
OH
H O

[00710] The title compound was prepared according to the schemes, steps and intermediates described below.

CI H2N N'Boc Boc H2N Al; OCOOEt F I N step-1 F I N step-2 N CI A NCI B

\ I ,Boc step-3 \ I - Boc HN H HN H step-4 F F I ~NI \
/ C D
N N \O^COOEt N H 0~ OH

i80 \ I
HN\ INH2 HN H
Step -5 F I 'I OH
E

N H 0 N H 0~

A) DIPEA, n-BuOH, 110 C, 16 h; B) Pd(OAc)2, BINAP, Cs2CO3, toluene, 100 C, 16 h; C) MeMgBr (3M solution in ether), THF, -78 C, 3 h; D) TFA, CH2CI2, rt, 3 h.
E) K2CO3, NMP, rt, 45 min.

[007111 Step-1 /
HN \ I N,Boc H
N CI

[007121 Compound 3 was prepared according to the schemes, steps and intermediates described in Example 20.
[007131 Step_2 HN \ NH(BOC) F SN

N I N O COOEt H
[007141 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 C 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 (Si02, 60-120, pet ether/ethyl acetate, 6/4) to get 5 (700 mg, 40%) as a white solid.
[007151 Step-3 HN \ NH(BOC) F N \ N ~ / OH
N H O~

[007161 To a stirred solution of 5 (0.4 g, 0.8 mmol) in THE (10 mL) was added Methyl magnesium bromide ((3 M solution in ether, 1.6 mL, 4.8 mmol) at -78 C. The reaction mixture was allowed to warm to -30 C over 3 h, cooled again to -78 C 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.
[00717] Step-4 F

CH
N H N O

[00718] To a stirred solution of 6 (0.2 g, 0.4 mmol) in CH2C12 (7.5 mL) was added TFA (3 vol.) at 0 C. 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 NaHCO3 solution (5 mL) and extracted with CH2C12 (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 (Si02, 60-120, pet ether/ethyl acetate, 6/4) to get 7 (130 mg, 86%) as a white solid.
[00719] Step-S

HN N
F H
N
~ 0 OH
N

[00720] 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 C was added 8 (0.023 g, 0.22 mmol) and the reaction mixture was stirred at 0 C 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 C) 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 mL), dried over Na2SO4 and concentrated under reduced pressure. The residue obtained was further purified by column chromatography (Si02, 60-120, pet ether/ethyl acetate, 5/5) to get 1-118 (35 mg, 38%) as a white solid. iH NMR
(CDOD) 6 ppm: 1.27 (s, 6H), 3.67 (s, 2H), 5.76 (dd, J = 2.4 & 9.6 Hz, I H), 6.34 (dd, J = 2 &
16,8 Hz, 1 H), 6.42 (dd, J = 9.6 & 16.8 Hz, 1 H), 6.54 (td, J = 2 & 7.2 Hz, 1H), 7.07-7.12 (m, 2H), 7.27-7.31 (m, 2H), 7.40 (d, J = 8 Hz, 1H), 7.45 (d, J - 8 Hz, 1 H), 7.92 (d, J
- 4 Hz, 1 H), 8.07 (d, J= 2 Hz, 1H); LCMS : m/e 436.2 (M-1).

[00721] Preparation of N-(3-(5-fluoro-2-(3-(2-morpholino-2-oxoethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-110 HN N
H
a4-1 O
NH O~N,_) O

[00722] 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. 'H NMR (DMSO-d6) 6 ppm: 3.4-3.5 (bm, 4H), 3.5-3.6 (bm, 4H), 4.69 (s, 2H), 5.75 (dd, J= 2 & 10 Hz, 1 H), 6.25 (dd, J = 2 & 17.2 Hz, 1 H), 6.42-6.49 (m, 2H), 7.05 (t, J
= 8 Hz, 1 H), 7.29 (t, J= 8 Hz, 3H), 7.41 (d, J= 8 Hz, 1H), 7.57 (d, J= 8.8 Hz, 1H), 7.91 (s, 1H), 8.12 (d, J= 3.6 Hz, 1H), 9.15 (s, 1H), 9.45 (s, 1H), 10.12 (s, 1H); LCMS : m/e 491.0 (M-2).
[00723] The intermediate 4-[(3-aminophenoxy)acetyl]-morpholine was prepared by the scheme shown below.

/ step-1 / step-2 O2N \ O~COOEt A OZN I 0^[ /OH B

al r' 0 step-3 ro 02N O"Nv J C H2N N,,) A) LiOH, THF, MeOH, H20, rt, 4 h; B) SOC12, 85 C, morpholine, 0 C, 30 min;
C) Pd-C, H2, ethyl acetate, rt, 2 h.
[007241 Step-1 [007251 To a stirred solution of 1 (1.0 g, 4.44 mmol) in methanol/THF/water :
5 mL/ 5 mL/5 mL 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 HC1 (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.
[007261 Step_2 i r o 02N 0~ ,/

[007271 Thionyl chloride (2.0 ml, 27.56 mmol) was added to 2 (0.2 g, 1.014 mmol) under nitrogen atmosphere. A drop of N,N Dimethylformamide was added to the mixture and the contents were stirred at 85 C for 2 h. After cooling to rt thionyl chloride was removed by concentration under reduced pressure. The residue was cooled to 0 C, morpholine (0.5 g, 5.74 mmol) was added to it in small portions and the reaction mixture was stirred at 0 C 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.

[007281 Sten-3 H2N O~ N,_) [007291 To a solution of 3 (0.180 g, 0.676 mmol) in ethyl acetate (10 mL)) 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.

[007301 Preparation of N-(3-(5-fluoro-2-(3-(1-hydroxy-2-methylpropan-2-yloxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-91 HN N

F _ , N N / 0y,,OH
H

[007311 The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 3-(1-hydroxy-2-methylpropan-2-yloxy)aniline in place of 4 in Step-2. 1H NMR (DMSO-d6) 6 ppm: 1.16 (s, 6H), 3.32-3.35 (m, 2H), 4.81 (t, J =
5.74 Hz, 1H), 5.74 (dd, J = 1.84 & 10.04 Hz, 1H), 6.24 (dd, J = 1.88 & 16.96 Hz, 1H), 6.44 (dd, J = 10.12 &
16.96 Hz, 1H), 6.50 (dd, J = 2.12 & 7.96 Hz, 1H), 7.02 (t, J = 8.12 Hz, 1H), 7.26-7.30 (m, 2H), 7.41 (d, J = 8.16 Hz, 1H), 7.48 (d, J = 8.24 Hz, 1H), 7.57 (d, J = 8.12 Hz, 1H), 7.92 (s, 1H), 8.09 (d, J= 3.6 Hz, 1H), 9.07 (s, 1H), 9.41 (s, 1H), 10.09 (s, 1H); LCMS : m/e 438.0 (M+1).
[007321 The intermediate 3-(1-hydroxy-2-methylpropan-2-yloxy)aniline was prepared by the scheme shown below.

Br COOEt step-1 \ I ~ / step-2 02N OH A 02N O' COOEt B

step-3 \ OH
2N 0 COOEt C H2N O~

A) K2CO3, DMF, 16 h, rt; B) Pd/C, ethanol, 5 h, rt; C) LAH (1M in THE
solution), 0 C to rt, 2 h.
[007331 Step-1 /I
02N \ O COOEt [007341 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%).
[007351 Step_2 H2N \ O COOEt [007361 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.
[007371 Step-3 H2N\ I0~OH

[00738] To a stirred solution of 4 (0.25 g, 1.15 mmol) in THE (5 mL) under N2 was added LAH (3.45 mL, 3.35 mmol, 1M solution in THF,) at 0 C. 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 (Si02, 60-120, pet ether/ethyl acetate, 6/4) to give 5 as a light brown liquid (0.15 g, 71%).

[00739] Preparation of N-(3-(5-fluoro-2-(3-(2-(2-oxopyrrolidin-l-yl)ethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-164 F ,I
~'NI I ~

H

[00740] 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. iH NMR (DMSO-d6) 8 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, 1 H), 6.25 (dd, J - 2 & 16.84 Hz, 1 H), 6.42-6.49 (m, 2H), 7.05 (t, J - 8.4 Hz, 1 H), 7.28 (t, J - 8 Hz, 2H), 7.33 (s, t H), 7.43 (d, J- 8 Hz, t H), 7.57 (d, J- 8 Hz, I H), 7.92 (s, t H), 8.12 (d, J- 3.6 Hz, 1H), 9.15 (s, lH), 9.45 (s, 1H), 10.13 (s, lH); LCMS : m/e 475 (M-2).

[00741] Preparation of N-(3-(5-fluoro-2-(6-(3-(methylsulfonyl)propoxy)pyridin-ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-80 HN N
H 0`

N NN
H

[007421 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. 'H NMR (DMSO-d6) 6 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, 1H), 6.25 (dd, J -1.8 & 16.96 Hz, 1H), 6,45 (dd, J - 10.04 & 16.92 Hz, 1H), 6.65 (d, J - 8.88 Hz, 1H), 7.27 (t, J - 8.08 Hz, I H), 7.39 (d, J - 8.08 Hz, 1H), 7.49 (d, J - 8 Hz, 1H), 7.92 (s, 1H), 7.99 (dd, J -2.6 & 8.76 Hz, 1H), 8.07 (d, J - 3.64 Hz, 1H), 8.31 (d, J - 2.28 Hz, 1H), 9.10 (s, 1H), 10.11 (s, 1H); LCMS : m/e 486.9 (M+l).

[007431 Preparation of N-(3-(2-(6-cyclobutoxypyridin-3-ylamino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-79 HN N
"0 F N O

N N N
H

[007441 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. 'H
NMR (DMSO-d6) 6 ppm: 1.57-1.66 (m, 1H), 1.71-1.78 (m, 1H), 1.94-2.04 (m, 2H), 2.32-2.38 (m, 2H), 4.95-5.05 (m, 1H), 5.73-5.76 (m, 1H), 6.25 (dd, J - 1.92 & 16.92 Hz, 1H), 6.45 (dd, J
10.08 & 16.92 Hz, 1H), 6.58 (d, J - 8.84 Hz, 1H), 7.25 (t, J - 8.04 Hz, 1H), 7.39 (d, J - 7.84 Hz, 1H), 7.45-7.55 (m, 1H), 7.90 (s, 1H), 7.94 (dd, J - 2.72 & 8.88 Hz, 1H), 8.06 (d, J - 3.68 Hz, 1 H), 8.27 (d, J - 2.6 Hz, 1 H), 9.04 (s, 1 H), 9.41 (s, 1 H), 10.1 (s, 1 H); LCMS : m/e 421.2 (M+1).

[00745] Preparation of N-(3-(5-fluoro-2-(6-((1-methylpiperidin-4-yl)methoxy)pyridin-3-ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-78 HN \ N ~JN' F H 0:/\/
N /

N/LN N
H

[00746] The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 3-amino-6-(1-methylpiperidin-4-yl)methoxypyridine in place of 4 in Step-2. iH NMR (DMSO-d6) 6 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, 1H), 6.24 (dd, J - 1.96 & 16.92 Hz, I H), 6.45 (dd, J - 10.08 &
16.92 Hz, I H), 6.62 (d, J - 8.88 Hz, 1H), 7.27 (t, J - 8.08 Hz, 1H), 7.40 (d, J - 8.88 Hz, 1H), 7.47 (d, J - 7.6 Hz, 1H), 7.92 (s, 1H), 7.97 (dd, J- 2.76 & 8.92 Hz, 1H), 8.07 (d, J- 3.72 Hz, 1H), 8.28 (d, J- 2.64 Hz, 1H), 9.07 (s, 1H), 9.41 (s, 1H), 10.11 (s, 1H); LCMS : m/e 478.0 (M+1).

[00747] Preparation of N-(3-(5-fluoro-2-(4-chloro-3-methoxyphenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-74 HN N
F- H j CI
N
N H OMe [00748] 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-d6) 6 ppm: 3.64 (s, 3H), 5.74 (dd, J - 2.12 & 9.96 Hz, 1H), 6.24 (dd, J -1.84 & 17 Hz, 1H), 6.44 (dd, J- 10 & 16.84 Hz, 1H), 7.13 (s, 1H), 7.28 (t, J- 8.08 Hz, 1H), 7.36 (dd, J- 2.12 & 8.68 Hz, 1H), 7.40 (d, J - 7.64 Hz, 1H), 7.45 (d, J - 2.04 Hz, 1H), 7.50 (d, J - 8.12 Hz, 1H), 7.91 (s, 1H), 8.13 (d, J - 3.52 Hz, 1H), 9.28 (s, 1H), 9.48 (s, 1H), 10.12 (s, 1H); LCMS : m/e 414.0 (M+1).

[007491 Preparation of N-(3-(5-fluoro-2-(4-(2-hydroxy-2-methylpropoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-73 HN H OH
F N O

N N
H

[007501 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.
'H NMR (MeOD) 6 ppm: 1.33 (s, 6H), 3.75 (s, 2H), 5.80 (dd, J - 3.28 & 10.64 Hz, 1H), 6.39 (dd, J - 2.24 & 16.96 Hz, 1H), 6.47 (dd, J - 9.6 & 16.96 Hz, 1H), 6.84 (td, J -3.48 & 9.0 Hz, 2H), 7.30 (t, J- 7.72 Hz, 1H), 7.41-7.50 (m, 4H), 7.89 (d, J- 3.88 Hz, 1H), 8.09 (s, 1H); LCMS
m/e 438 (M+1).

[007511 Preparation of N-(3-(5-fluoro-2-(6-(l,1-dioxidothiomorpholin-4-yl) pyridin-3-ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-72 Nl'-5~
7 b ~0 HN ~Sz- 0 F N NJ
NN N
H

[007521 The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 3-amino-6-(1,1-dioxidothiomorpholin-4-yl) pyridine in place of 4 in Step-2. iH NMR (DMSO-d6) 6 ppm: 3.00-3.15 (bm, 4H), 3.90-4.10 (bm, 4H), 5.76 (dd, J
1.64 & 10.04 Hz, 111), 6.26 (dd, J - 1.72 & 16.92 Hz, 111), 6.46 (dd, J -10.04 & 16.88 Hz, I H), 6.87 (d, J - 9.04 Hz, 1H), 7.20 (t, J - 8.04 Hz, 1H), 7.39 (d, J - 8.24 Hz, 1H), 7.50 (d, J -7.68 Hz, 1 H), 7.90-7.93 (m, 2H), 8.06 (d, J = 3.6 Hz, 1 H), 8.3 5 (d, J = 2.4 Hz, 1 H), 9.0 (s, 1 H), 9.40 (s, 1H), 10.12 (s, 1H); LCMS : m/e 484 (M+1).

[00753] Preparation of N-(3-(5-fluoro-2-(6-(2-(2-oxopyrrolidin-1-yl)ethoxy)pyridin-3-ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-70 HN

F N Zz,,, 0~\N
N
N N
H

[00754] 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) 6 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, 1H), 6.25 (d, J = 17.04 Hz, 1H), 6.45 (dd, J = 10.12 & 16.84 Hz, 1H), 6.63 (d, J =
8.96 Hz, 1H), 7.27 (t, J = 8.04 Hz, 1H), 7.39 (d, J = 7.56 Hz, 1H), 7.47 (d, J
= 7.32 Hz, 1H), 7.92 (s, 1H), 7.98 (dd, J = 2.36 & 8.84 Hz, 1H), 8.08 (d, J = 3.3 Hz, 1H), 8.31 (d, J = 2.24 Hz, 1H), 9.10 (s, 1H), 9.44 (s, 1H), 10.11 (s, 1H); LCMS : m/e 478.0 (M+1).

[00755] Preparation of (R)-N-(3-(5-fluoro-2-(6-(tetrahydrofuran-3-yloxy)pyridin-3-ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-69 HN N
F ~N H

~ N N
H

[00756] 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) 6 ppm: 1.91-1.99 (m, 1H), 2.14-2.23 (m, 1H), 3.70-3.77 (m, 2H), 3.81 (dd, J - 7.90 & 15.48 Hz, 1H), 3.88 (dd, J - 4.76 & 10.16 Hz, 1H), 5.38 (t, J - 4.68 Hz, 1H), 5.75 (dd, J - 1.72 & 10.08 Hz, 1H), 6.24 (d, J - 16.92 Hz, 1H), 6.45 (dd, J - 10.16 &
16.88 Hz, 1H), 6.63 (d, J - 8.84 Hz, 1H), 7.26 (d, J - 7.64 Hz, 1H), 7.39 (d, J-- 7.92 Hz, 1H), 7.46 (d, J - 7.64 Hz, 1H), 7.92 (s, 1H), 7.97 (dd, J - 2.6 & 8.83 Hz, 1H), 8.07 (d, J - 3.6 Hz, 1H), 8.32 (d, J - 2.48 Hz, 1H), 9.08 (s, 1H), 9.42 (s, 1H), 10.10 (s, 1H);
LCMS : m/e 437.2 (M+ 1).

[00757] Preparation of N-(3-(2-(4-chloro-3-(3-(methylsulfonyl)propoxy)phenylamino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-55 HN N
F H CI
iI

[00758] 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. iH NMR (DMSO-d6) 6 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, 1H), 6.24 (dd, J -1.84 & 16.92 Hz, 1H), 6.44 (dd, J = 10.12 & 16.96 Hz, 1H), 7.15 (d, J = 8.72 Hz, 1H), 7.30 (t, J - 8.08 Hz, 1H), 7.35 (dd, J = 2.2 & 8.8 Hz, 1H), 7.43 (d, J = 8 Hz, 1H), 7.45-7.55 (m, 2H), 7.91 (s, 1H), 8.14 (d, J - 3.56 Hz, 1H), 9.31 (s, 1H), 9.49 (s, 1H), 10.14 (s, 1H); LCMS : m/e 520.0 (M+1).

[007591 Preparation of N-(3-(2-(3-fluoro-4-(2-methoxyethoxy)phenylamino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-96 ~O
HN" v NH
F N O~~OMe N N IIF
H

[007601 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.
iH NMR (DMSO, 400 MHz) 6 10.13 (s, 1H), 9.43 (s, 1H), 9.18 (s, 1H), 8.09 (d, 1H, J = 3.68 Hz), 7.92 (s, 1H), 7.65 (dd, 1H, J = 2.3, 14.2 Hz), 7.47 (d, 1H, J = 8.24 Hz), 7.41 (d, 1H, J = 8.28 Hz), 7.27 (t, 2H, J = 8.0 Hz), 6.94 (t, 1H, J = 9.4 Hz), 6.44 (dd, 1H, J =
16.96, 10.1 Hz), 6.23 (dd, 1H, J =1.84, 16.96 Hz), 5.73 (dd, 1H, J=1.4, 10.1 Hz), 4.04 (m, 2H), 3.61 (m, 2H), 3.29 (s, 3H). MS m/z: 442.0 (M+H+).

[00761] Preparation of N-(3-(2-(4-tert-butoxycarbonyl-2,3-dihydrobenzo[1,4]oxazin-6-yl)amino-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-175 lL
HN

NH BOC
F N-~

H\

[00762] 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[1,4]oxaxine in place of 4 in Step-2. MS m/z: 507.1 (M+H ).

[00763] Preparation of N-(3-(2-(4-tert-butoxycarbonyl-2,3-dihydrobenzo[1,4]oxazin-6-yl)amino-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-174 HN

NH
F N H N

~N H \ / 0 [00764] 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 +).

[00765] Preparation of N-(3-(2-(4-trifluoroacetyl-2,3-dihydrobenzo[1,4]oxazin-6-yl)amino-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-143 HN

N N
I
N N O
H

[00766] 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 +).

[00767] Preparation of N-(3-(2-(4-methylsulfonyl-2,3-dihydrobenzo[1,4]oxazin-6-yl)amino-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-140 HN

O/
NH O.;S, F N
II INI
~N \ / O
H

[00768] The title compound was prepared by treating the product of Example 85 with mesyl chloride Et3N in CH2C12 at 0 C 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+).

[00769] Preparation of N-(3-(2-(4-methyl-2,3-dihydrobenzo[1,4]oxazin-6-yl)amino-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-126 HN

NH
F N
I
"N N \ 0 H

[00770] 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[1,4]oxazine in place of 4 in Step-2. MS m/z: 421.1 (M+H ).

[00771] Preparation of N-(3-(2-(4-acetyl-2,3-dihydrobenzo[1,4]oxazin-6-yl)amino-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-112 HN

LNH
F N
I
N O
H

[00772] The title compound was prepared by treating the product of Example 85 with acetic anhydride and pyridine in CH2C12 at rt for 1 hr, followed by washing with 1N
HCl, then with aqueous NaHCO3, drying over Na2SO4 and removal of solvents in vacuo. MS m/z:
449.1 (M+H

[007731 Preparation of N-(3-(2-(l-tert-butoxycarbonyl-lH-indazol-5-yl)amino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-151 lL
HN

F N N
\N" N N'BOC
H

[007741 The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 5-amino-N-(tert-butoxycarbonyl)-1H-indazole in place of 4 in Step-2. MS m/z: 490.2 (M+H+).

[007751 Preparation of N-(3-(2-(1H-indazol-5-yl)amino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-156 HN

NH
F / N ~N
~~ NH
N H

[007761 The title compound was prepared by treating the product of Example 90 with 4N HC1 in dioxane at rt for 1 hr followed by removal of solvents in vacuo. MS m/z:
390.1 (M+H+).

[007771 Preparation of N-(3-(2-(l-methyl-lH-indazol-5-yl)amino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-155 HN

NH
F N N
N
N
H

[007781 The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 5-amino-l-methyl-lH-indazole in place of 4 in Step-2. MS
m/z: 404.2 (M+H ).

[007791 Preparation of N-(3-(5-fluoro-2-(3-sulfamoylphenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-160 HN N
F - , /

[007801 The title compound was prepared according to the schemes, steps and intermediates described below.

\ SO2NH2 F step-1 FN I N step-2 NiCI A N ~'CI B

CI

HN \ I N02 HN ~aNH2 F NI / step-3 F IN / step-4 HN N
F H
N
N N

A) DIPEA, n-butanol, 120 C, 2 h, pressure tube; B) AcOH, ethanol, 90 C, 16 h; C) Pd-C, H2, ethanol, rt, 3 h; D) acryloyl chloride, K2CO3, NMP, 0 C, 60 min.
[007811 Step-1 HN \ NO

F ' _ N! C1 [007821 A pressure tube was charged with 2 (10.0 g, 0.072 mot), 1 (24.1 g, 0.145 mot), n-BuOH (100 mL) and DIPEA (13.9 g, 0.108 mol) and the contents were stirred at 120 C 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.

[007831 Sten_2 i HN \ N02 F N N H SOZNHZ

[007841 To a solution of 3 (0.25 g, 0.93 mmol) and 4 (0.16 g, 0.93 mmol) in ethanol (2.5 mL) was added glacial acetic acid (0.083 g, 1.39 mmol), and the reaction mixture was stirred in a pressure tube at 90 C 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.
[007851 Step-3 F N N H SOZNHZ

[007861 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 H., 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.

[007871 Step-4 0 HN N
F I H

[007881 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 C was added acryloyl chloride (0.021 g, 0.23 mmol) and the reaction mixture was stirred at 0 C 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 C) 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 A1203, MeOH/chloroform: 3/97) to get 1-160 (0.028 g, 35%) as light brown solid. 'H NMR (DMSO-d6) 6 ppm: 5.75 (dd, J- 1.68 & 10.24 Hz, 1H), 6.25 (dd, J-1.8 & 17 Hz, 1H), 6.43 (dd, J- 10 & 16.92 Hz, 1H), 7.27-7.35 (m, 5H), 7.40 (d, J- 8 Hz, 1H), 7.60 (d, J - 8.16 Hz, 1H), 7.92 (s, 1H), 7.95-8.05 (m, 1H), 8.07 (s, 1H), 8.14 (d, J - 3.52 Hz, 1H), 9.50 (s, 2H), 10.12 (s, 1H); LCMS : m/e 428.9 (M+l).

[007891 Preparation of N-(3-(5-cyano-2-(4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-109 HN" v HN \
NC I NN/ I
N
H

[007901 The title compound was prepared according to the steps and intermediates as described below.

HN'Boc HN-Boc HN'Boc Br CI 2I NHZ NH HZN q NH
/ N
Br N Br / INI0~~0 N CI step-1 /, step-2 \
1 A Ni ICI B N H

HN'Boc HN" v NH r0~ NH
NC / N / 0 -> NC / 0 step-3 \ step-4 C H D N H

A) DMA, K2CO3, rt, 10 h, pressure tube; B) PTSA, dioxane, 100 C, 2 h; C) Zn(CN)2, Ph3P, DMF, 120 C, 12 h; D) 4N HCl, dioxane, rt, 1 hr; then acryloyl chloride, Et3N, DCM, -10 C, min.
[007911 Step-1 . Boc a NH
Br N CI

[007921 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).

[007931 Step_2 Boc HN

NH
Br N N
H
[007941 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 C 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+).
[007951 Step-3 ,Boc HN

N C , , , N N 'a [007961 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 C 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+1).

[007971 Sten-4 a NH
NH
NC / 0-1-1,1_0 N N
H

[007981 Compound 6 (0.10 g, 0.21 mmol) was dissolved in 4 N HC1 (2 mL) 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+).
[007991 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 C.
The reaction was stirred for 10 minutes at -10 C 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+).

[00800] Preparation of N-(3-(5-cyano-2-(6-methoxypyridin-3-ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-173 HN

NH
N C / INII / INO
1, N N
H

[00801] 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:
3 88.2 (M+H +).

[00802] Preparation of N-(3-(5-cyclopropyl-2-(4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-139 HNv NH

/ N / II O---IOMe N N
H

[00803] The title compound was prepared according to the steps and intermediates as described below.

H N' Boc H N' Boc \ \ O
NH NH ~ I ~\0 Br / N HZN 3 step-1 N CI step-2 \N CI
A

HN'Boc 0 HN" v \
NH
~NH

\ I step-3 e'N N0~~0 A) Potassium cyclopropyltrifluoroborate, Pd(OAc)2, Xanphos, Cs2(CO3), toluene, 100 C, 12 h;
B) PTSA, dioxane, 100 C, 2 h; C) Zn(CN)2, Ph3P, DMF, 120 C, 12 h; D) 4N HCI, dioxane, rt, 1 hr; then acryloyl chloride, Et3N, DCM, -10 C, 10 min.
[008041 Step-1 ,Boc HN

\
NH

N
I
N CI

[008051 Potassium cyclopropyltrifluoroborate (0.4g, 3.0 mmol), compound 1 (1.0g, 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 C 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+).

[008061 Sten-2 ,Boc aNH
N N
H

[008071 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+).
[008081 Sten-3 ~O
HN" v NH
O
e'NN N
H

[008091 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+).

[008101 Preparation of N-(3-(5-cyclopropyl-2-(6-methoxypyridin-3-ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-167 ~0 HN" v / NH
N / I We \ N
N N
H

[008111 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 +).

[008121 Preparation of N-(3-(5-fluoro-2-(3-(3-(2-oxopyrrolidin-l-yl)propoxy)phenylamino)pyrimidin-4-yloxy)phenyl)acrylamide 1-162 ~I

F H
N N O N
H

[008131 The title compound was prepared according to the schemes, steps and intermediates described below.

HO I/ N,Boc \ I ,Boc H N O
I 2 H 0 H z 4 F N step-1 F step-2 0 NCI A N CI B

CI
/ I '01, \ N,Boc H step-3 0 N HZ 0 F ~N \ 0 F -~N '0'0' 0 ~~ C step-4 \N D
vv 6 0 \ I N
H
F
\ 0 N H N 0~

A) DIPEA, n-BuOH, 110 C, 16 h; B) Pd(OAc)2, BINAP, Cs2CO3, toluene, 100 C, 16 h; C) TFA, CH2Cl2, rt. 2 h; D) K2CO3, NMP, rt, 45 min.

[008141 Step-1 0 ja NH(BOC) F , , NCI

[008151 A pressure tube was charged with 2 (2.0 g, 9.61 mmol), 1 (3.21 g, 19.23 mmol), n-BuOH (30 mL) and DIPEA (1.86 g, 14.42 mmol) and the contents were stirred at 110 C 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.
[008161 Step_2 O NH(BOC) F , , I ~~ 0 H
[008171 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 C 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 (Si02, 60-120, product getting eluted in 3% methanoUchloroform: 3/97) to get 5 (0.3 g, 60%) as yellow solid.

[008181 Sten-3 a F N
I ~~ O
N N O N
H

[008191 To a stirred solution of 5 (0.25 g, 0.46 mmol) in CH2C12 (10 mL) was added TFA (1.0 mL) at 0 C 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 [008201 Sten-4 0 ~I
L

F H

N I a O' [008211 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 C was added acryloyl chloride (0.020 g, 0.22 mmol) and the reaction mixture was stirred at 0 C 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 C) 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, 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) 6 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, 1H), 6.25 (dd, J- 1.88 & 16.9 Hz, 1H), 6.38-6.45 (m, 2H), 6.93 (t, J- 8.12 Hz, 1H), 7.02-7.04 (m, 2H), 7.11 (s, M), 7.43 (t, J - 8.16 Hz, 1H), 7.55 (d, J - 8.24 Hz, 1H), 7.68 (d, J- 1.8 Hz, 1H), 8.56 (d, J- 2.88 Hz, 1H), 9.56 (s, 1H), 10.34 (s, 1H); LCMS :
m/e 490.0 (M-2).
[008221 The intermediate 3-(3-(2-oxopyrrolidin-1-yl)propoxyaniline 4 was prepared according to the scheme shown below.

I
zzt"
0 02N ~F

step-1 step-2 A 02N 0~\~ B

O
H2N 0~\N

A) NaH, DMF, rt, 16 h; B) SnClz, Cone. HCl, 50 C, 2 h.
[008231 Step-1 02N 0-"'~

[008241 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 C. 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.
[008251 Step_2 all:" H2N O~~\N

[008261 To a stirred solution of 3 (2 g, 7.57 mmol) in cone. HCl (20 mL) was added SnC12 (7.5 g, 34.06 mmol) in small portions. The reaction mixture was stirred at 50 C for 2 h, cooled and basified with NaHCO3. 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.

[008271 Preparation of N-(4-(5-fluoro-2-(3-(2-(2-oxopyrrolidin-l-yl)ethoxy)phenylamino)pyrimidin-4-yloxy)benzyl)-N-methylacrylamide 1-146 ~ N

F
N

N H N
ONE/

[008281 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) 6 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, 111), 6.43 (d, J - 16.72 Hz, 111), 6.50 (d, J
5.72 Hz, 1H), 6.60-6.75 (m, 1H), 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, 1H), 8.21 (d, J- 2.36 Hz, 1H); LCMS : m/e 506.2 (M+1).

[00829] Preparation of N-(4-(5-fluoro-2-(3-(3-(methylsulfonyl)propoxy)phenylamino)pyrimidin-4-yloxy)benzyl)-N-methylacrylamide 1-136 N
O \I
F ~
NN
N O

[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-(3-methylsulfonyl)propoxyaniline in place of 4 in Step-2. 1H NMR
(CDC13) 6 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, 1H), 4.72 (s, 1H), 5.7-5.82 (m, 1H), 6.43 (dd, J- 1.96 & 16.96 Hz, 1H), 6.49-6.53 (m, 1H), 6.6-6.75 (m, 1H), 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, 1H), 8.21 (bd, J- 2.52 Hz, 1H); LCMS : m/e 515.0 (M+1).

[00831] Preparation of N-(4-(5-fluoro-2-(6-methoxypyridin-3-ylamino)pyrimidin-yloxy)benzyl)-N-methylacrylamide 1-117 N
JO'~
F N COMe H

[00832] 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. iH
NMR (DMSO-d6) 6 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 t 1l), 6.20 (dd, J - 1.2 & 16.4 Hz, 1H), 6.56 (d, J - 8.8 Hz, t 1l), 6.82-6.90 (m, 1H), 7.28-7.35 (m, 4H), 7.71 (bd, J - 7.6 Hz, 1H), 8.14 (s, 1H), 8.44 (bd, J- 2.8 Hz, 1H), 9.48 (s, 1H); LCMS : m/e 410 (M+1).

[008331 Preparation of N-(4-(5-fluoro-2-(3-(3-(2-oxopyrrolidin-l-yl)propoxy)phenylamino)pyrimidin-4-yloxy)benzyl)-N-methylacrylamide I-111 O

NIN O
H

[008341 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-1-yl)propoxy)aniline in place of 4 in Step-2. 'H
NMR (DMSO-d6) 6 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, 1H), 6.20 (dd, J- 2.4 & 16.8 Hz, 1H), 6.24 (bd, J- 8 Hz, 1H), 6.86 (dd, J- 10.4 & 16.8 Hz, 1H), 6.93 (t, J- 8 Hz, 1H), 7.08 (t, J- 8 Hz, 2H), 7.28-7.36 (m, 4H), 8.48 (d, J- 2.8 Hz, 1H), 9.51 (s, 1H);
LCMS : m/e 520.2 (M+1).

[008351 Preparation of N-(3 -(5-fluoro-2-(3-(2-(2-oxopyrrolidin-l-yl)ethoxy)phenylamino)pyrimidin-4-yloxy)phenyl)acrylamide 1-184 0 N'\1 N N AO
F \N 0 ,~ND

H

[008361 The title compound was prepared according to the schemes, steps and intermediates described below.

HO NO \ I H2N \ I O,~N
CI 2 2 OIaNO2 4 FI L N step-1 F , I, step-2 ) N CI A N CI B

\I \I
O NO2 \\~ step-3 0 NH2 step-4 F I / ~ j 0 r \ = N0 N NO~ N N O D

F \N \ O
N N O
H

A)) K2C03, DMF, rt, 16 h; B) Pd(OAc)2, BINAP, Cs2CO3, toluene, 100 C, 8 h; C) Pd-C, H2, methanol, rt, 16 h. D)) acryloyl chloride, K2CO3, NMP, 0 C, 30 min.
[008371 Step-1 ~I
O \ NO2 Ft N
NCI

[008381 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 (10 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.

[008391 Sten_2 ~I
O \ NO2 NN , O,-,,~N
H

[008401 To a solution of 3 (0.9 g, 3.3 mmol) in toluene (30 mL) 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 C 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 (Si02, 60-120, product getting eluted in methanol/chloroform: 8/92) to get 5 (0.50 g, 33%) as a white solid.
[008411 Step-3 ~I
O \ NH2 N
N'~'N , 0~~ND
H

[008421 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.
[008431 Step-4 N NNI
N')~' N I o,- N
H

[008441 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 mL) at 0 C was added acryloyl chloride (0.053 g, 0.6 mmol) and the reaction mixture was stirred at 0 C 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 C) 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 methanolldichloromethane (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. tH
NMR (DMSO-d6) 6 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, 1H), 6.26 (dd, J- 1.6 & 17.2 Hz, 1H), 6.39-6.46 (m, 2H), 6.95 (t, J- 8.4 Hz, 1H), 7.03-7.12 (m, 3H), 7.44 (t, J- 8.4 Hz, 1H), 7.56 (d, J- 8.4 Hz, 1H), 7.70 (s, 1H), 8.51 (d, J- 2.8 Hz, 1H), 9.56 (s, 1H), 10.35 (s, 1H); LCMS : m/e 478 (M+1).
[008451 The intermediate 3-(2-(2-oxopyrrolidin-1-yl)ethoxyaniline 4 was prepared according to the scheme shown below.

n N-\-OH step-1 N step-2 / N
02NI0^~ B H2NICI
A

A) NaH, THF, rt,16 h; B) Pd-C, H2, methanol, rt, 16 h.
[008461 Step-1 02N I O,,~N

[008471 To a stirring solution of NaH (3.4 g, 141.6 mmol, 60% dispersion in paraffin oil) in dry THE (50 mL) was added 1 (6 g, 46.0 mmol) at 0 C 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 THE
(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 mL), brine (25 mL), dried over Na2SO4 and concentration under reduced pressure to get a residue which was purified by column chromatography (Si02, 60-120, product getting eluted in methanol/chloroform: 10/90) to get 3 (2.5 g, 30%) as a brownish liquid.
[008481 Step_2 H2N I / Oi~N

[008491 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.

[008501 Preparation of N-(3-(2-(6-methoxypyridin-3-ylamino)-5-methylpyrimidin-yloxy)phenyl)acrylamide 1-186 HN"
0 '6 N
N N
H

[008511 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-d6) 6 ppm:
2.16 (s, 3H), 3.73 (s, 3H), 5.76 (dd, J- 1,92 & 10.04 Hz, 1H), 6.24 (dd, J- 1.92 & 16.92 Hz, 1H), 6.39-6.49 (m, 2H), 6.92 (dd, J - 1.48 & 8 Hz, 1H), 7.40 (t, J - 8.08 Hz, t H), 7.49 (d, J - 8.16 Hz, 1H), 7.64 (d, J- 1.84 Hz, 1H), 7.77-7.79 (m, 1H), 8.17 (bs, 1H), 8.20 (s, 1H), 9.27 (s, 1H), 10.29 (s, 1H); LCMS : m/e 378 (M+l).

[00852] Preparation of N-(3-(5-methyl-2-(phenylamino)pyrimidin-4-yloxy)phenyl)acrylamide ~0 ~' NH

N N
H

[00853] The title compound was prepared according to the schemes, steps and intermediates described below.

,Boc .Boc HNBoc ao CI aOH O 2 N step-1 N step -2 step -3 N CI A N CI B N H C

O

NH
&0 0 I- I
\ I step -4 \ i \
N H D N H

A) K2CO3, DMF, rt, 24 h; A') (Boc)20, THF, 60 C, 2 h; B) aniline, cone. HC1, EtOH, 80 C, 1 h; C) TFA, CH2C12, 0 C to rt, 1/2 h; D) acryloyl chloride, NMP, 0 C, 10 min.

[00854] Step-1 HN'BOC

N
I
\N CI

[00855] 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.
[00856] Step_2 HN'BOC

NIN
H

[00857] A pressure tube was charged with 3 (75 mg, 0.224 mmol), conc. HC1 (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 C 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 soln. 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 (Si02, 60-120 mesh, EtoAc/Hexane: 50/50) to get 4 (0.04 g, 45.9%) as an off-white sold.

[008581 Sten-3 & 0 e'N!NJO
H
[008591 To a stirring solution of 4 (160 mg, 0.40 mmol) in dichloromethane (4.0 mL) was added at 0 C, trifluoroacetic acid (0.8 mL). 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.
[008601 Step-4 NH

N N
H

[008611 To a stirred solution of 5 (75 mg, 0.256 mmol) in NMP (0.8 mL) at 0 C
was added acryloyl chloride (34.8 mg, 0.38 mmol) and the reaction mixture was stirred at 0 C 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 (Si02, 60-120 mesh, CHC13/MeOH: 99/1) to get 1-248 (0.035 g, 39.6%) as a white colored solid. 'H NMR (DMSO-d6) 6 ppm: 2.15 (s, 3H), 5.75 (dd, J - 1.92 & 10.04 Hz, 1H), 6.24 (dd, J - 1.96 & 16.96 Hz, 1H), 6.41 (dd, J - 10.6 & 17 Hz, 1H), 6.78-6.8 (m, 1H), 6.94 (dd, J - 1.44 & 8.04 Hz, 1H), 7.00 (t, J -7.52 Hz, 2H), 7.40-7.44 (m, 3H), 7.53 (d, J - 8.24 Hz, 1 H), 7.6 (t, J - 2 Hz, 1 H), 8.23 (d, J -1.04 Hz, 1 H), 9.36 (s, 1H), 10.30 (s, 1H); LCMS: m/e 346.8 (M+l).

[008621 Preparation of 1-(4-(5-fluoro-2-(phenylamino)pyrimidin-4-ylamino)piperidin-l-yl)prop-2-en-l-one 1-229 D
HN
N N
H

[008631 The title compound was prepared according to the schemes, steps and intermediates described in Example 20 by using 1-tert-butyloxycarbonyl-4-aminopiperidine in place of 2 in Step-1 and aniline in place of 4 in Step-2. 'H NMR (DMSO-d6) 6 ppm:l.35-1.50 (m, 2H), 1.90-2.05 (m, 2H), 2.7-2.85 (m, 1H), 3.10-3.20 (m, 1H), 4.11-4.15 (m, 2H), 4.46 (bd, J- 13.72 Hz, I H), 5.67 (dd, J- 2.44 & 10.4 Hz, 1H), 6.10 (dd, J- 2.44 & 16.6 Hz, 1H), 6.82-6.88 (m, 2H), 7.22 (t, J- 7.44 Hz, 2H), 7.35 (d, J- 7.56 Hz, 1H), 7.70 (d, J- 7.72 Hz, 2H), 7.87 (d, i - 3.76 Hz, 1H), 9.07 (s, 1H); LCMS : m/e 341.383 (M+l).

[008641 Preparation of 2-((3-(5-fluoro-2-(4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)(hydroxy)methyl)acrylonitrile 1-71 CN
HO

HN
F I ~ N / O Me I\/
N N
H

[008651 The title compound was prepared according to the schemes, steps and intermediates described below.
0 OMe O OMe 0 OMe \ I / I \ I 0 OMe I OMe H N

F N sty F I N step-2 F I N N \ I O step-N CI A Ni CI B N, C
N

O OH 0 N'OMe OMe OMe HN step4 HN step-5 F
E
IH
JIJ D I N
N%~H J~ <To O H CN
HO
OMe HN
F N 0 9 ~CN HN
F O
8 F step-6 I N 'IN I OMe N N
H
H

A) 2, DIPEA, n-BuOH, 120 C, 12 h; B) conc. HCI, ethanol, 100 C, 5 h; C) LiOH, MeOH/THF/H20, rt, 6 h; D) Me-NH-OMe.HCI, EDCI.HCI, HOBT, DIPEA, DMF, rt, 8 h;
E) LAH (1.0 M soln. in THF), -78 C, 30 min; F) DABCO, 1,4-dioxan/water, rt, 48 h.

[008661 Step-1 0 OMe HN \

F 'N
N ICI

[008671 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 mL) was heated in a pressure tube (120 C, 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.
[008681 Step_2 0 OMe HN

NIN
H
[008691 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 C 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%).

[008701 Sten-3 HN
F / O
NIN r H

[008711 To a stirred solution of 5 (0.6 g, 1.4 mmol) in methanol/THF/water: 6 mL/6 mL/3 mL
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.
[008721 Step-4 0 N,OMe O
HN
O
F NN/ I

N
H

[008731 To a stirred solution of 6 (0.4 g, 1 mmol) in DMF (3 mL) were added McNH-OMe.HC1 (0.102 g, 0.1 mmol), EDCI.HCl (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 Na7SO4, filtered and concentrated under reduced pressure to get 7 (0.4 g, 90.9%) as a white solid.

[00874] Step-5 O H

HN

NIN
H

[00875] To a stirred solution of 7 (0.4 g, 0.9 mmol) in THE (10 mL) was added LAH (1.8 mL, 1.8 mmol) at -78 C. 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 (Si02, 60-120, pet ether/ethyl acetate 7/3) to get 8 (200 mg, 58%) as a yellow solid.
[00876] Step-6 CN
HO

HN
F NI OMe N
H

[00877] To a stirred solution of 8 (200 mg, 0.523 mmol) and 9 (69 mg, 1.3 mmol) in 1,4-dioxane/H20 (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 (Si02, pet ether/ethyl acetate, 6/4) to get 1-71 as greenish gummy material (0.05 g, 22.7%).'H NMR
(DMSO-d6) 6 ppm: 3.48 (s, 3H), 3.77 (t, J - 4.4 Hz, 2H), 4.11-4.16 (m, 2H), 5.11 (s, 1H), 5.99 (s, 1H), 6.06 (s, 1H), 6.85 (s, 1H), 6.91 (d, J - 8.84 Hz, 2H), 7.15 (d, J -7.44 Hz, 1H), 7.30-7.40 (m, ; LCMS : m/e (M+1).

[008781 Preparation of 2-((4-(5-fluoro-2-(phenylamino)pyrimidin-4-ylamino)phenyl)(hydroxy)methyl)acrylonitrile 1-161 OH
,N
HN

F N C
N N
H

[008791 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. 'H NMR
(CDCI3) 6 ppm: 5.32 (s, 1H), 6.07 (d, J- 0.8 Hz, 1H), 6.15 (d, J- 1.6 Hz, 1H), 6.84 (d, J- 2.8 Hz, 1H), 7.03-7.06 (m, 2H), 7.29 (t, J - 1.6 Hz, 2H), 7.3 8 (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, 1H); LCMS : m/e 361.8 (M+1).

[008801 Preparation of 2-((4-(5-fluoro-2-(3-trifluoromethoxyphenylamino)pyrimidin-4-ylamino)phenyl)(hydroxy)methyl)acrylonitrile 1-163 OH
HN

H

[008811 The title compound was prepared according to the schemes, steps and intermediates described in Example 107 by using 3-trifluoromethoxyaniline in place of 4 in Step-2. 'H NMR
(DMSO-d6) 6 ppm: 5.29 (d, J- 3.8 Hz, 1H), 6.13 (s, 1H), 6.19 (s, 1H), 6.31 (dd, J- 3.8 Hz, 1H), 6.83 (d, J - 7.76 Hz, 1 H), 7.11 (d, J - 7.8 Hz, 1 H), 7.30-7.37 (m, 2H), 7.61-7.63 (m, 2H), 7.81 (s, 1H), 7.90 (d, J- 7.4 Hz, 1H), 8.16 (dd, J- 1.44 & 3.56 Hz, 1H), 9.45 (s, 1H), 9.53 (s, 1H);
LCMS : m/e 446 (M+1).

[008821 Preparation of N-(3-(5-fluoro-2-(3-(3-(methylsulfonyl)propoxy)phenylamino)pyrimidin-4-yloxy)phenyl)acrylamide 1-116 I

F H
N

N H 0 'S"O
O

[008831 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.
iH NMR (DMSO-d6) 6 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, 1H), 6.25 (dd, J- 1.72 & 16.88 Hz, 1H), 6.43 (d, J- 9.96 & 16.76 Hz, 2H), 6.95 (t, J- 8.12 Hz, 1H), 7.06 (t, J- 7.48 Hz, 2H), 7.13 (s, I H), 7.44 (t, J- 8.12 Hz, 1H), 7.56 (d, J- 8.44 Hz, 1H), 7.68 (s, 1H), 8.50 (d, J- 2.84 Hz, 1H), 9.57 (s, 1 H), 10.34 (s, 1 H); LCMS : m/e 487.0 (M+2).

[008841 Preparation of N-(3-(5-cyclopropyl-2-(4-(2-methoxyethoxy)phenylamino)pyrimidin-4-yloxy)phenyl)acrylamide 1-131 HN
/I

0 \
/ 0 '/~0 N N
H

[008851 The title compound was prepared according to the steps and intermediates as described below.

HN"Boc Boc HNBoc HN
0-/~O

Br CI 2 / \OH 6,0 H2N\

N B e,,,, Br N O~\O
NCI step-1 step2 step 3 N N
N CI H

HN'Boc HN
O
0 step-4 N N H N H

A) K2CO3, DMA, rt, 5 h; B) PTSA, dioxane, 100 C, 2 h; C) potassium cyclopropyltrifluoroborate, Pd(OAc)2, Xanphos, Cs2CO3, toluene, 100 C, 12 h;
D) 4N HCL, dioxane, rt, 1 hr; then acryoyl chloride, Et3N, DCM, -10 C, 10 min [008861 Step-1 .Boc ao Br ~ I
N CI

[008871 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).

[008881 Sten_2 Boc HN
NNI
Br N 0-1/~0 N N
H
[008891 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 100 C 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 ).
[008901 Sten-3 Boc HN

N / 0~\0 N N
H

[008911 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 C 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).

[008921 Sten-4 HN

/ 0_~_\O
N N
H

[008931 The title compound was prepared from compound 6 following the procedure described in Example 96. MS m/z: 447.1 (M+H+).

[008941 Preparation of 1-(4-(5-fluoro-2-(3-(2-dimethylaminoethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-2-methylprop-2-en-l-one 1-207 NMe2 Jf F !b N N
H

[008951 The title compound was prepared according to the schemes steps and intermediates described below.

/NMe2 0 OMe ) CO2Me CO2Me / I \ I / I NMe2 H2N 2 HN \ H2N \ 0f F I N step-1 F I N step-2 F N / I step-3 I'll NCI A NCI B N~N \ C

CO2H 0 N,OMe 0 \
/ NMe2 NMe2 NMe2 HN 0 HN O 8 MgBr HN 0 F IAN /I step F IAN /I F I\N /I
NN \ p N~N \ step 5 NN

A) 2, DIPEA, n-BuOH, 90 C, 12 h; B) 4, Pd(OAc)2, BINAP, Cs2CO3, toluene, 110 C, 16 h; C) LiOH, MeOH/THF/H20, rt, 6 h; D) McNHOMe.HC1, EDCI.HC1, HOBT, DIPEA, DMF, rt, 3 h;
E) 8, THF, 0 C to rt, 2 h.
[008961 Step-1 0 OMe HN
F , NCI

[008971 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 C, 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.

[008981 Sten_2 ~O 0 NH
F
NN
N O
H
[008991 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 C 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 (Si02, 60-120, chloroform/methanol, 9/1) to get 4 (0.63 g, 84%) as yellow solid.
[009001 Step-3 NH
F JN
I
N N O
H

[009011 To a stirred solution of 5 (0.3 g, 0.70 mmol) in methanol/THF/water: 1 mL/I 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.

[009021 Sten-4 0'N 0 NH
F '/
tiN

H

[009031 To a stirred solution of 6 (0.29 g, 0.70 mmol) in DMF (3 mL) were added McNH-OMe.HCl (0.068 g, 0.70 mmol), EDCI.HC1 (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 (Si02, 60-120, methanol/chloroform : 20/80) to get 7 (0.061 g, 19%) as gummy yellow solid.
[009041 Sten-5 NH

N~

H

[009051 To a stirred solution of 7 (100 mg, 0.22 mmol) in THE (1 mL) at 0 C
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 NH4C1 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 (Si02, 60-120, product getting eluted in 20%
methanol/chloroform) to get 1-207 (9 mg. 9%) as gummy yellow solid. 'H NMR (DMSO-d6) 6 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, 1H), 6.82 (s, 1H), 7.04 (t, J - 8.16 Hz, I H), 7.18 (d, J- 8.16 Hz, 1H), 7.33 (s, 1H), 7.47 (t, J- 7.88 Hz, 1H),7.62 (d, J - 7.72 Hz, 1H), 8.13-8.16 (m, 3H), 9.24 (s, 1H), 9.56 (s, 1H); LCMS : m/e 450.1 (M+1).

[009061 Preparation of 1-(4-(5-fluoro-2-(phenylamino)pyrimidin-4-ylamino)phenyl)-3-methylbut-2-en- l -one I-206 HN

F \
NN
NN
H

[009071 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. 'H NMR (DMSO-d6) 6 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, 1H), 9.31 (s, 1H), 9.68 (s, 1H); LCMS : m/e 363.0 (M+1).

[009081 Preparation of 1-(3-(5-fluoro-2-(3-(prop-2-ynyloxy)phenylamino)pyrimidin-4-ylamino)phenyl)-3-methylbut-2-en- l -one 1-211 HN
F I %~ \

N H N 0/\

[009091 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) 6 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, 1H), 4.59 (d, J - 2.36 Hz, 2H), 6.79-6.81 (m, 2H), 7.03 (dd, J- 3.12 & 8.04 Hz, 1H), 7.14 (t, J- 2.2 Hz, I H), 7.23 (t, J- 8.12 Hz, I H), 7.54 (t, J - 7.92 Hz, I H), 7.83 (d, J-7.96 Hz, I H), 7.86 (dd, J- 2.08 & 8.08 Hz, 1H), 8.03 (d, J- 4.96 Hz, 1H), 8.21 (t, J- 1.88 Hz, 1H);
LCMS : m/e 417.0 (M+1).

[009101 Preparation of 1-(3-(5-fluoro-2-(3-(trifluoromethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-3-methylbut-2-en-l-one 1-223 O
HN
F ~NI /
'CF3 N N O
H

[009111 The title compound was prepared according to the schemes, steps and intermediates described in Example 112 by using 3-trifluoromethoxyaniline in place of 4 in step-2. 1H NMR
(CDC13) 6 ppm: 2.0 (d, J - 1.08 Hz, 3H), 2.24 (d, J - 1.04 Hz, 3H), 6.74 (t, J
- 1.24 Hz, 1H), 6.85 (dd, J - 1.08 & 7.0 Hz, 1H), 6.90 (s, 1H), 7.08 (s, 1H), 7.24-7.28 (m, 1H), 7.35 (td, J - 1.2 & 7.44 Hz, 1 H), 7.49 (t, J - 7.8 8 Hz, 1 H), 7.63 (s, 1 H), 7.72 (td, J -1.04 & 7.76 Hz, 1 H),7.90-7.92 (m, 1H), 8.00-8.05 (m, 2H); LCMS : m/e 447 (M+1).

[00912] Preparation of 1-(3-(5-fluoro-2-(3-(trifluoromethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-2-methylprop-2-en-l-one 1-199 HN

F , , N H OCF3 [00913] The title compound was prepared according to the schemes, steps and intermediates described in Example 112 by using 3-trifluoromethoxyaniline in place of 4 in step-2 and isopropenylmagnesium bromide in place of 8 in step-5. 'H NMR (DMSO-d6) 6 ppm:
1.97 (s, 3H), 5.6 (s, 1H), 6.0 (d, J- 0.96 Hz, 111), 6.82 (d, J- 8.08 Hz, 111), 7.27 (t, J- 8.2 Hz, 111), 7.41 (dd, J - 1.12 & 7.56 Hz, I H), 7.48 (t, J - 7.76 Hz, 111), 7.60 (dd, J -1.28 & 7.88 Hz, 111), 7.78 (s, I H), 7.90 (d, J - 1.64 Hz, 1H), 8.15 (d, J - 8 Hz, 1H), 8.19 (d, J -3.64 Hz, 1H), 9,55 (s, 1H), 9.65 (s, 1H); LCMS : m/e 433 (M+1).

[00914] Preparation of 1-(4-(5-fluoro-2-(phenylamino)pyrimidin-4-ylamino)phenyl)-2-methylprop-2-en-1-one 1-185 HN
F

N N
H

[00915] 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.
'H NMR

(DMSO-d6) 6 ppm: 1.99 (s, 3H), 5.54 (s, 1H), 1.01 (s, 1H), 6.93 (t, J- 7.36 Hz, 1H), 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, 1H), 9.32 (s, 1H), 9.72 (s, 1H); LCMS : m/e 348.8 (M+1).

[00916] Preparation of N-(3-(2-(3-(2-(dimethylamino)ethoxy)phenylamino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-233 ~0 ~' NH

b /I
NH
F N
\ ~N\

H

[00917] 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. iH
NMR (CD30D) 6 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, 1H), 6.38-6.42 (m, 2H), 6.52-6.55 (m, 1H), 7.1-7.11 (m 2H), 7.30 (t, J - 8.0 Hz, 1H), 7.36 (s, 1H), 7.42-7.48 (m, 2H), 7.94 (d, J- 3.6 Hz, 1H), 8.05 (s, 1H);
LCMS : m/e 437 (M+1).

[00918] Preparation of N-(3-(5-fluoro-4-(4-phenoxyphenoxy)pyrimidin-2-ylamino)phenyl)acrylamide 1-130 O \ I
F

N N N
H H

[00919] 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. iH
NMR (DMSO-d6) 6 ppm: 5.71 (dd, J- 1.6 & 10 Hz, 1H), 6.22 (dd, J- 1.6 & 16.8 Hz, 1H), 6.44 (dd, J- 10.4 & 17.2 Hz, 1H), 6.98-7.05 (m, 3H), 7.1-7.12 (m, 2H), 7.17 (t, J-7.2 Hz, 1H), 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, 1H), 8.5 (s, 1H), 9.6 (s, 1H), 10.05 (s, 1H); LCMS : m/e 443,0 (M+l).

[00920] Preparation of (S)-N-(3 -(5-fluoro-2-(4-(tetrahydrofuran-3-yloxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-43 / O
HN \ N' F N H O/, I~NI/ C0 N H

[00921] 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.
iH NMR (DMSO-d6, 500 MHz): 6 10.10 (s, 1H), 9.35 (s, 1H), 8.95 (s, 1H), 8.05 (d, J- 4.0 Hz, 1H), 7.92 (s, 1H), 7.52 (d, J - 9.0 Hz, 2H), 7.47 (d, J - 7.5 Hz, 1H), 7.41 (d, J - 8.5 Hz, 1H), 7.27 (t, J - 8.0 Hz, 1H), 6.72 (d, J - 9.0 Hz, 2H), 6.45 (dd, J - 1.5, 17.0 Hz, 1H), 6.25 (dd, J -1.1, 16.5 Hz, 1H), 5.75 (dd, J- 1.1, 10.0 Hz, 1H), 4.93 - 4.84 (m, 1H), 3.88 -3.72 (m, 4H), 2.20 - 2.10 (m, 1H), 1.97 - 1.90 (m, 1H). MS m/e - 436 [M++l]

[00922] Preparation of (R)-N-(3-(5-fluoro-2-(4-(tetrahydrofuran-3-yloxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-46 HN

HN \
FN
01'- 0 ""CO
INN
H

[00923] 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.
iH NMR (DMSO-d6, 500 MHz): 6 10.10 (s, 1H), 9.35 (s, 1H), 8.95 (s, 1H), 8.05 (d, J- 4.0 Hz, 1H), 7.92 (s, 1H), 7.52 (d, J- 9.0 Hz, 2H), 7.47 (d, J- 7.5 Hz, 1H), 7.41 (d, J- 8.5 Hz, 1H), 7.27 (t, J - 8.0 Hz, 1H), 6.72 (d, J - 9.0 Hz, 2H), 6.45 (dd, J- 1.5, 17.0 Hz, 1H), 6.25 (dd, J -1.1, 16.5 Hz, 1H), 5.75 (dd, J- 1.1, 10.0 Hz, 1H), 4.93 - 4.84 (m, 1H), 3.88 -3.72 (m, 4H), 2.22 - 2.14 (m, 1H), 1.97 - 1.90 (m, 1H). MS: m/e - 436 [M++l]

[00924] Preparation of N-(3-(5-fluoro-2-(3-((1-methylpiperidin-3-yl)methoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-acrylamide I- 76) HN
/I
HN
F N

N N a0`0 " N

[00925] The title compound was prepared according to the steps, schemes and intermediates described in Example 20 by using 3-(1-methylpiperidin-3-yl)methoxyaniline in place of 4 in step-2. ' H NMR (DMSO-d6, 500 MHz): 6 10.08 (s, 1 H), 9.41 (s, 1 H), 9.09 (s, 1 H), 8.11 (d, J -3.5 Hz, 1H), 7.90 (s, 1H), 7.57 (d, J- 8.0 Hz, 1H), 7.41 (d, J- 8.0 Hz, 1H), 7.32 (s, 1H), 7.30 -7.20 (m, 2H), 7.03 (t, J - 8.0 Hz, 1 H), 6.50 - 6.40 (m, 2H), 6.24 (dd, J -1.5, 17.0 Hz, 1 H), 5.74 (dd, J - 2.0, 10.5 Hz, 1 H), 3.75 - 3.65 (m, 2H), 2.73 (d, J - 10 Hz, 1 H), 2.60 (d, J - 10.5 Hz, 1H), 2,13 (s, 3H), 1.98 - 1.83 (m, 2H), 1.75 - 1.58 (m, 3H), 1.55 - 1.45 (m, 1H), 1.05 - 0.95 (m, 1H). MS: m/e - 477 (M++1).

[009261 Preparation of N-(3-(5-fluoro-2-(3-((1-methylpiperidin-4-yl)methoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-acrylamide 1-82 HN' HN
FN

N~

[009271 The title compound was prepared according to the steps, schemes and intermediates described in Example 20 by using 3-(1-methylpiperidin-4-yl)methoxyaniline in place of 4 in step-2. 'H-NMR (CDC13+DMSO-D6, 500 MHz): 6 9.04 (bs, 1H), 8.30 (s, 1H), 7.96 (s, 1H), 7.75 (s, 1H), 7.63 (s, 1H), 7.59 (d, J - 7.0 Hz, 1H), 7.38 - 7.33 (m, 2H), 7.27 (t, J - 8.5 Hz, 1H), 7.16 (t, J - 8 Hz, 1 H), 7.09 (d, J - 8.5 Hz, 1 H), 6.52 (d, J - 7.0 Hz, 1 H), 6.51 - 6.3 8 (m, 2H), 5.73 (dd, J - 2.0, 9.0 Hz, I H), 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, 1H), 1,85 - 1.74 (m, 2H), 1.45 - 1.32 (m, 2H), 1.28 - 1.25 (m, 2H).
MS: m/e - 477 (M++ 1).

[00928] Preparation of N-(3-(2-(3-(4-(2-hydroxyethyl)piperazin-1-yl)phenylamino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-83 HN
HN
FN
N N
H N
OH

[00929] The title compound was prepared according to the steps, schemes and intermediates described in Example 20 by using 3-(4-(2-t-butyldimethylsilyloxyethyl)piperazin-1-ylaniline in place of 4 in step-2 and deprotecting the TBS ether with TFA in DCM as a step-5. iH NMR
(DMSO-d6, 500 MHz): 6 8.99 (s, 1H), 8.85 (s, 1H), 8.04 (d, J- 4.0 Hz, 1H), 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, 1H), 6.35 - 6.31 (m, 1H), 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++1).

[00930] Preparation of N-(4-(5-fluoro-2-(3-methoxyphenylamino)pyrimidin-4-ylamino)benzyl)-N-methylacrylamide 1-113 HN \
F ,,I ~N a 5;1"

H

[00931] 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-methoxyaniline in place of 4 in step-2. 'H-NMR
(DMSO-d6, 200 MHz): 6 9.36 (bs, 1H), 9.16 (bs, 1H), 8.10 (d, J - 3.4 Hz, 1H), 7.83 - 7.70 (m, 2H), 7.34 (bs, 1H), 7.26 - 7.01 (m, 4H), 6.86 - 6.73 (m, 1H), 6.48 (d, J - 8.0 Hz, 1H), 6.21 (dd, J - 16.4, 2.2 Hz, 1H), 5.76- 5.64 (m, 1H), 4.64 - 4.53 (two s, 2H), 3.65 (s, 3H), 3.00 -2.88 (two s, 3H). MS:
m/e - 408.2 [M++1].

[00932] Preparation of N-(4-(5-fluoro-2-(6-methoxypyridin-3-ylamino)pyrimidin-ylamino)benzyl)-N-methylacrylamide 1-114 HN
FN / 0~
N N N
H

[00933] 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. 'H-NMR (DMSO-D6, 200 MHz): 6 9.36 (bs, 1H), 9.09 (bs, 1H), 8.32 (bs, 1H), 8.06 (dd, J - 3.8 Hz, 1H), 7.97-7.92 (m, I H), 7.76- 7.68 (m, 2H), 7.20 - 7.08 (m, 2H), 6.87 - 6.75 (m, I H), 6.72 (d, J - 8.4 Hz, 1H), 6,22 (dd, J - 19.4, 2.6 Hz, 1H), 5.74 - 5.65 (m, 1H), 4.64 - 4.53 (two s, 2H), 3.78 (s, 3H), 3.00 - 2.88 (two s, 3H). MS: m/e - 409 (M+l).

[00934] Preparation of N-(3-(5-fluoro-2-(3-methyoxyphenylamino)pyrimidin-4-ylamino)benzyl)-N-methylacrylamide I-115 O
HN \ I N

F ~N 410 N N H

[00935] 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-methoxyaniline in place of 4 in step-2. 'H-NMR
(DMSO-d6, 200 MHz): 6 9.50 - 9.30 (m, 1H), 9.15 - 8.96 (m, 1H), 8.15 (bs, 1H), 7.82 - 7.59 (m, 2H), 7.45 - 7.00 (m, 4H), 6.97 - 6.65 (m, 2H), 6.55 - 6.45 (m, 1H), 6.26 - 6.12 (m, I H), 5.78 -5.60 (m, I H), 4.68 (s, 1H), 4.55 & 3.75 (two s, 3H), 2.90 & 3.00 (two s, 3H). MS: m/e - 408.2 [M++l].

[00936] Preparation of N-(3-(5-fluoro-2-(3-(4-(2-hydroxyethyl)piperazin-1-yl)phenylamino)pyrimidin-4-yloxy) phenyl)acrylamide 1-84 HN
/I

F I 'z N

N H aN
N O H

[00937] The title compound was prepared according to the steps, schemes and intermediates described in Example 98 by using 3-(4-(2-t-butyldimethylsilyloxyethyl)piperazin-1-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): 6 8.19 (s, 1H), 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, I H), 6.93 (s, I H), 6.72 (d, J - 6.5 Hz, I
H), 6.57 - 6.54 (m, I H), 6.44 (d, J - 17.0 Hz, 1H), 6.29 - 6. 20 (m, 1H), 5.78 (d, J - 10.0 Hz, 1H), 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).

[009381 Preparation of N-(3-(5-fluoro-2-(3-((l-methylpiperidin-3-yl)methoxy)phenylamino)pyrimidin-4-yloxy)phenyl)acrylamide 1-81 O
HN' F 'N

N Na\O T 1 " `J
N

[009391 The title compound was prepared according to the steps, schemes and intermediates described in Example 98 by using 3-(1-methylpiperidin-3-yl)methoxyaniline in place of 4 in step-2. iH-NMR (CDC13, 500 MHz): 6 8.19 (d, J- 2.5 Hz, 1H), 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, 1H), 6.91 (s, 1H), 6.79 (d, J - 7.5 Hz, 1H), 6.48 - 6.44 (m, 1H), 6.42 (s, 1H), 6.29 - 6.23 (m, 1H), 5.77 (d, J- 10 Hz, 1H), 3.67 - 3.62 (m, 2H), 2.95 - 2.91 (m, 1H), 2.82 - 2.76 (m, 1H), 2.28 (s, 3H), 2.11 - 2.05 (m, 1H), 1.98 - 1.92 (m, 1H), 1.79 - 1.70 (m, 3H), 1.11 - 1.04 (m, 1H). MS: m/e - 478 (M++1).

[009401 Preparation of N-(3-(5-fluoro-2-(3-((1-methylpiperidin-4-yl)methoxy)phenylamino)pyrimidin-4-yloxy)phenyl)-acrylamide 1-75 HN' O \
F I N

N~

[009411 The title compound was prepared according to the steps, schemes and intermediates described in Example 98 by using 3-(1-methylpiperidin-4-yl)methoxyaniline in place of 4 in step-2. 'H-NMR (DMSO-D6, 500 MHz): 6 10.31(s, 1H), 9.50 (s, 1H), 8.50 (s, 1H), 7.67 (s, 1H), 7.55 (d, J - 8.0 Hz, I H), 7.42 (t, J - 8.0 Hz, I H), 7.10 (s, I H), 7.04 (d, J - 7.0 Hz, 2H ), 6.94 (t, J - 8.0 Hz, 1H), 6.45 - 6.39 (m, 2H), 6.27 (d, J - 15.0 Hz, I H ), 5.78 (dd, J
- 2.0, 10.5 Hz, I H), 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++1).

[009421 Preparation of N-(3-(5-cyano-2-(phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-157 ~I
HN N
N~~ H

\N /
N!N
H

[009431 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).

[009441 Preparation of N-(3-(5-fluoro-2-(3-(trifluoromethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-244 HN" v HN
F

N N \ 0 H )< F
F F

[00945] 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).

[00946] Preparation of N-(3-(5-fluoro-2-(pyridin-3-ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-234 HN- ~
HN

F SINN
N
NI
H

[00947] 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+1).

[00948] Preparation of N-(3-(5-fluoro-2-(4-fluorophenylamino)pyrimidin-4-ylamino)phenyl)acrylamide I- 247 HN"
HN
F
N N
H

[00949] 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+1) [00950] Preparation of N-(3-(5-fluoro-2-(3-(3-morpholinopropoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-208 ~0 HN"

HN
F

N H O

[00951] 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).

[00952] Preparation of N-(3-(2-(3-(3-(1H-imidazol-1-yl)propoxy)phenylamino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide I- 204 ~0 HN" v HN
F N
~~

[00953] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 3-(3-(1H-imidazol-l-yl)propoxy)aniline in the place of 4 in Step 2. MS 474.3 (M+Na).

[00954] Preparation of N-(3-(2-(l-acetylpiperidin-3-ylamino)-5-fluoropyrimidin-ylamino)phenyl)acrylamide 1-238 HN

HN
F

N!N N
H

[00955] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 1-(3-aminopiperidin-1-yl)ethanone in the place of 4 in Step 2.
MS 421.1 (M+Na).

[00956] Preparation of N-(3-(5-fluoro-2-(phenylamino)pyrimidin-4-yloxy)phenyl)acrylamide F N O
H

[00957] 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+1).

[00958] Preparation of N-(3-(5-fluoro-2-(6-methoxypyridin-3-ylamino)pyrimidin-ylamino)phenyl)acrylamide 1-243 HN N
H
F IN I O"
N
NIN
H

[00959] 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).

[00960] Preparation of N-(3-(5-methoxy-2-(3-methoxyphenylamino)pyrimidin-4-ylamino)phenyl)acrylamide. 1-158 I
HN \ N
O H

N N/ o H

[00961] 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-methoxyaniline in place of 4 in step 2. MS 392.3 (M+l).

[00962] Preparation of N-(3-(5-methoxy-2-(6-methoxypyridin-3-ylamino)pyrimidin-ylamino)phenyl)acrylamide 1-192 HN N
H
,0 NNN 0~
N
H

[00963] 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).

[00964] Preparation of N-(3-(5-fluoro-2-(6-methoxypyridin-3-ylamino)pyrimidin-yloxy)phenyl)acrylamide 1-222 0 H
F IN / I N ' NI N

H

[00965] 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).

[009661 Preparation of 4-(3-acrylamidophenylamino)-N-tert-butyl-2-(6-methoxypyridin-3-ylamino)pyrimidine-5-carboxamide 1-216 H I \ N
N N
H

[009671 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).

[009681 Preparation of (R)-1-(3-(5-fluoro-2-(6-methoxypyridin-3-ylamino)pyrimidin-4-ylamino)piperidin-l-yl)prop-2-en- l -one 1-202 N
HN
F ~NI / 0 N
N N
H

[009691 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).

[00970] Preparation of (R)-l-(3-(5-fluoro-2-(3-methoxyphenylamino)pyrimidin-4-ylamino)piperidin-1-yl)prop-2-en-l-one 1-195 O I
N
HN
F

N NN O
H

[00971] 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-methoxyaniline in place of 4 in step 2. MS 394.3 (M+Na).

[00972] Preparation of (S)-1-(3-(5-fluoro-2-(6-methoxypyridin-3-ylamino)pyrimidin-4-ylamino)piperidin-l-yl)prop-2-en-l-one 1-197 O I
N
,'U
HN
F O
NI N
N
H

[00973] 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).

[00974] Preparation of (S)-1-(3-(5-fluoro-2-(3-methoxyphenylamino)pyrimidin-4-ylamino)piperidin-1-yl)prop-2-en-l-one 1-196 N
' HN
F

N N O
H

[00975] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using (S)-tert-butyl 3-aminopiperidine-1-carboxylate in the place of 2 in Step 1 and 3-methoxyaniline in place of 4 in step 2. MS 372.3 (M+1).

[00976] Preparation of (R)-1-(3-(5-fluoro-2-(6-methoxypyridin-3-ylamino)pyrimidin-4-yloxy)piperidin-l-yl)prop-2-en-l-one 1-180 N
O

F 'N O
N N JzCJI N
H

[00977] 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+1).

[00978] Preparation of (R)-l-(3-(5-fluoro-2-(3-methoxyphenylamino)pyrimidin-4-yloxy)piperidin-l-yl)prop-2-en-l-one 1-190 N

F

N IN O
H

[00979] 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-methoxyaniline in place of 4 in step 2. MS 395.3 (M+Na).

[00980] Preparation of (S)-1-(3-(5-fluoro-2-(6-methoxypyridin-3-ylamino)pyrimidin-4-yloxy)piperidin-l-yl)prop-2-en-l-one 1-193 N
o N N N
H

[00981] 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).

[00982] Preparation of (S)-1-(3-(5-fluoro-2-(3-methoxyphenylamino)pyrimidin-4-yloxy)piperidin-l-yl)prop-2-en-l-one 1-179 N
s O
F

N N O
H

[00983] 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-methoxyaniline in place of 4 in step 2. MS 395.3 (M+Na).

[00984] Preparation of 1-(3-(5-fluoro-2-(6-methoxypyridin-3-ylamino)pyrimidin-ylamino)pyrrolidin-l-yl)prop-2-en-l-one 1-203 O I
N
HN Q I

N N N
H

[00985] 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).

[00986] Preparation of 1-(3-(5-fluoro-2-(3-methoxyphenylamino)pyrimidin-4-ylamino)pyrrolidin-1-yl)prop-2-en-1-one 1-201 O I
N
HN .0 F ~
NN
NN O
H

[00987] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using tert-butyl 3-aminopyrrolidine-1-carboxylate in the place of 2 in Step 1 and 3-methoxyaniline in place of 4 in step 2. MS 358.3 (M+1).

[00988] Preparation of (R)-l-(3-(5-fluoro-2-(3-methoxyphenylamino)pyrimidin-4-ylthio)piperidin-1-yl)prop-2-en-l-one 1-137 N
S~
F O
~
NN \
H O, [00989] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using (S)-tert-butyl 3 -mercaptopiperidine- 1 -carboxylate in the place of 2 in Step 1 and 3-methoxyaniline in place of 4 in step 2. MS 411.1 (M+Na).

[009901 Preparation of (R)-l-(3-(2-(3-chlorophenylamino)-5-fluoropyrimidin-4-ylamino)piperidin-1-yl)prop-2-en-l-one 1-147 HN"

N t N I N
H CI

[009911 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+1).

[009921 Preparation of (R)-1-(3-(5-fluoro-2-(3-(2-morpholinoethoxy)phenylamino)pyrimidin-4-ylamino)piperidin-l-yl)prop-2-en-l-one 1-135 N
HN
F O

A-N,N\,j [009931 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).

[00994] Preparation of (E)-4-(dimethylamino)-N-(3-(5-fluoro-2-(6-methoxypyridin-3-ylamino)pyrimidin-4-ylamino)phenyl)but-2-enamide 1-125 O
HN \ I N" .. " N~
H
F INN I O~
NI N
H

[00995] 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).

[00996] Preparation of 2-((1H-pyrazol-1-yl)methyl)-N-(3-(5-fluoro-4-(m-tolylamino)pyrimidin-2-ylamino)phenyl)acrylamide. 1-98 O
/ I

\ NH HN
F N / N.N
N"N \ ,~~J~
H

[00997] The title compound was prepared according to the schemes, steps and intermediates described in Example 4, by using 2-((1H-pyrazol-1-yl)methyl)acryloyl chloride in the place of 6 in Step 3. MS 466.1 (M+Na).

[00998] Preparation of (E)-4-(azetidin-1-yl)-N-(3-(5-fluoro-4-(m-tolylamino)pyrimidin-2-ylamino)phenyl)but-2-enamide 1-123 /I D
\ NH HN" v , N
Ft IN
N
H

[00999] 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).

[001000] Preparation of (E)-N-(3-(5-fluoro-4-(m-tolylamino)pyrimidin-2-ylamino)phenyl)-4-morpholinobut-2-enamide 1-102 7\ I 0 0 NH HN/J~~%/
F

NIN
H

[001001] 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).

[0010021 Preparation of (E)-4-((1S,4S)-2,5-diazabicyclo[2.2.1]heptan-2-yl)-N-(3-(5-fluoro-4-(m-tolylamino)pyrimidin-2-ylamino)phenyl)but-2-enamide 1-101 /I

\ NH HN ~N JNH
IN)N

H

[0010031 The title compound was prepared according to the schemes, steps and intermediates described in Example 4, by using (E)-4-((1S,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).

[0010041 Preparation of (E)-N-(3-(5-fluoro-4-(m-tolylamino)pyrimidin-2-ylamino)phenyl)-4-((2-methoxyethyl)(methyl)amino)but-2-enamide 1-120 N H HN .. " N
F
I
NJN\ I
H

[0010051 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).

[0010061 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 OH

NH HN" v vN
F ~
NIN \ I
H

[0010071 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) [0010081 Preparation of (R,E)-N-(3-(5-fluoro-4-(m-tolylamino)pyrimidin-2-ylamino)phenyl)-4-(3-hydroxypyrrolidin-l-yl)but-2-enamide 1-104 OH
O
\ INH N0 HN
F _ ~N'b N" -H

[0010091 The title compound was prepared according to the schemes, steps and intermediates described in Example 4, by using (R,E)-4-(3-hydroxypyrrolidin-1-yl)but-2-enoyl in the place of 6 in Step 3. MS 485.3 (M+Na).

[0010101 Preparation of (E)-N-(3-(5-fluoro-4-(m-tolylamino)pyrimidin-2-ylamino)phenyl)-4-(1H-pyrazol-1-yl)but-2-enamide 1-100 CNH H N" N
F , _ LN N
H

[0010111 The title compound was prepared according to the schemes, steps and intermediates described in Example 4, by using (E)-4-(1H-imidazol-l-yl)but-2-enoyl chloride in the place of 6 in Step 3. MS 466.1 (M+Na).

[0010121 Preparation of (R,E)-N-(3-(5-fluoro-2-(4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-4-(3-hydroxypyrrolidin-l-yl)but-2-enamide 1-89 / I ~O
HN ~
N
F H
N N

NNH
'O H
O

[0010131 The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using (R,E)-4-(3-hydroxypyrrolidin-1-yl)but-2-enoyl chloride in the place of 7 in Step 4. MS 545.3 (M+Na).

[001014] 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 O
HN I N
F H /
N N
I
\NjNH
qH

[001015] 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).

[001016] Preparation of 2-((lH-pyrazol-1-yl)methyl)-N-(3-(5-fluoro-2-(4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-85 O
HN I
F H
N -Ir N NH

O

[001017] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 2-((1H-pyrazol-l-yl)methyl)acryloyl chloride in the place of 7 in Step 4. MS 526.1 (M+Na).

[001018] Preparation of N-(3-(5-fluoro-2-(phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-28 HN N
F , _ /
N,N \
H

[001019] 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).

[001020] 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 O NH
\\~N~
NH HN
F

L IN\
N
H

[001021] 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-1-yl)but-2-enoyl chloride in the place of 6 in Step 3. MS 512.3 (M+Na).

[0010221 Preparation of 1-(3-(5-methyl-2-(3-aminosulfonylphenylamino)pyrimidin-ylamino)phenyl)-3-methylbut-2-en-l-one 1-224 HN \

/
N \ S02NH2 [0010231 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. 'H NMR (DMSO-d6) 6 ppm: 1.97 (s, 3H), 2.14 (s, 6H), 6.88 (s, 1H), 7.25-7.30 (m, 4H), 7.47 (t, J- 7.92 Hz, 1H), 7.62 (d, J- 7.72 Hz. 1H), 7.96 (s, 1H), 8.0-8.07 (m, 3H), 8.20 (t, J- 7.36 Hz, 1H), 8.55 (s, 1H), 9.37 (s, 1H);
LCMS : m/e 438 (M+l).

[0010241 Preparation of N-(3-acrylamidophenyl)-N-(5-cyano-2-(6-methoxypyridin-ylamino)pyrimidin-4-yl)acrylamide 1-171 HN

, N C N O
J~ \
N N N
H

[0010251 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 +).

[0010261 Preparation of N-3-(N-methyl-N-(5-fluoro-2-(4-methyl-3,4-dihydro-2H-benzo[b][1,4]oxazin-6-ylamino)pyrimidin-4-yl)aminophenylacrylamide 1-127 HN

H

[0010271 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+).

[0010281 Preparation of N-(3-(5-methyl-2-(phenylamino)pyrimidin-4-ylamino)benzyl)acrylamide 1-205 H

HN

NIN\
H

[0010291 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. 'H-NMR (CDC13, 500 MHz): 6 7.91 (s, 1H), 7.77 (s, 1H), 7.57 (d, J- 9.0 Hz, 2H), 7.41 (d, J- 8.0 Hz, 1H), 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, 1H), 5.78 (bs, 1H), 5.63 (d, J - 10.5 Hz, 1H), 4.51 (d, J - 6.0 Hz, 2H), 2.12 (s, 3H). MS: m/e - 360 (M++1).

[0010301 Preparation of (E)-3-(5-methyl-2-(phenylamino) pyrimidin-4-ylamino) benzyl but-2-enoate 1-246 \ i O
HN

~N \ O
N N
H

[0010311 The title compound was prepared according to the schemes, steps, and intermediates described below.
\

step-1 I N step-2 A
B
N iCI N CI

J::)""O H 6 OB H N \
HN ~

N ~/ step-3 C i I~ 0 N H N H

A) 2, Xanthophos, Pd2(dba)3, Cs2CO3, CH3CN, 90 C, 12 hr; B) 4, t-BuOH, 90 C, 4 hr; C) 6, TEA, DCM, -30 C, 5 min [0010321 Step-1 HN \ OlTBDMS
NCI

[0010331 To a stirred solution of 1 (0.34 g, 2.08 mmol) in acetonitrile (5 mL) 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 It and stirred at 90 C 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. 'H-NMR (CDC13, 500 MHz): 6 8.0 (s, 1H), 7.55 (s, 1H), 7.51 (d, J - 8.0 Hz, 1H), (t, J - 7.5 Hz, 1H), (d, J - 7.5 Hz, 1H), 6.48 (s, 1H), 4.76 (s, 2H), 2.18 (s, 3H), 0.95 (s, 9H), 0.10 (s, 6H). MS: m/e - 364 [M++1].
[0010341 Step-2 i HN OH
'J:

ININ ~/

H
[0010351 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 C 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. 'H-NMR (DMSO-d6, 500 MHz): 6 10.45 (s, 1H), 9.82 (s, 1H), 7.91 (s, 1H), 7.58-7.01 (m, 9H), 4.50 (s, 2H), 2.16 (s, 3H). MS: m/e 307 [M++l].
[0010361 Step-3 HN \ I O II

N N
H

[0010371 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 C under N2 atmosphere.
The reaction mixture was stirred for 5 minutes at -30 C 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. 'H-NMR (CDC13, 500 MHz): 6 7.87 (s, 1H), 7.65 (s, 1H), 7.58-7.51 (m, 3H), 7.34 (t, J- 7.5 Hz, 1H), 7.30-7.23 (m, 3H), 7.13 (d, J- 7.5 Hz, 1H), 7.08-6.96 (m, 2H), 6.40 (s, 1H), 5.87 (dd, J-1.5, 15.5 Hz, 1H), 5.16 (s, 2H), 2.13 (s, 3H), 1.87 (dd, J- 2.0, 7.0 Hz, 3H). 13C-NMR (CDC13,125 MHz): 6 166.3, 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].

[0010381 Preparation of (E)-4-(5-methyl-2-(phenylamino) pyrimidin-4-ylamino) benzyl but-2-enoate 1-60 HN

N i NJO
H

[0010391 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-l. 'H-NMR (CDC13, 500 MHz): 6 8.19 (bs, 1H), 7.81 (s, 1H), 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, 1H), 7.08-6.98 (m, 2H), 6.54 (s, 1H), 5.89 (dd, J - 12.5, 14.0 Hz, 1H), 5.17 (s, 2H), 2.15 (s, 3H), 1.89 (dd, J - 1.5, 7.0 Hz, 3H). MS: m/e - 375 [M++l].

[0010401 Preparation of N-methyl-N-(3-(5-methyl-2-(phenylamino)pyrimidin-4-ylamino)benzyl)acrylamide 1-220 HN N II
~N \ 0 N N
H

[0010411 The title compound was prepared according to the schemes, steps, and intermediates described below.

N /
H N,Boc /
CI 2 2 \ I N` H2N
H N Boc 4 step-1 I N step-2 / I / I 1 ^/ CI
p HN \ N, Boc vNH

N step-3 N I \ step-4 NH C / D
N H

/
HN \ I N II

A) 2, Xanthophos, Pd2(dba)3, Cs2CO3, CH3CN, 100 C, 12 hr; B) 4, t-BuOH, 90 C, 4 hr; C) 10 N HC1, DCM, rt, 30 min: D) 7, TEA, DCM, -10 C, 10 min.

[0010421 Step-1 HN Boc N CI

[0010431 To a stirred solution of 1 (2.6 g, 15.7 mmol) in acetonitrile (26.7 mL) 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 lh and further heated to 100 C 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
(CDC13, 500 MHz): ^ 8.01 (s, 1H), 7.56 (d, J - 7.5 Hz, 1H), 7.43 (s, 1H), 7.35 (t, J - 7.0 Hz, 1H), 7.05 (s, 1H), 6.80 (bs, 1H), 4.45 (s, 2H), 2.87 (s, 3H), 2.30 (s, 3H), 1.48 (s, 9H).
[0010441 Step_2 HN \ N~Boc -"N

N~N I / -H
[0010451 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. iH-NMR (DMSO-d6, 500 MHz): 6 10.32 (s, 1H), 9.78 (s, 1H), 7.91 (s, 1H), 7.50 (d, J -8.0 Hz, 1H), 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++1).

[001046] Len-3 HN NH
ININ
O
H

[001047] To a stirred solution of 5 (0.05 mg, 0.11 mmol) in DCM (5 mL) was added 37%
HC1 (1.0 mL) 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 C, basified 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. 'H-NMR (CDCl3, 500 MHz): 6 7.95 - 7.85 (m, 2H), 7.68 -7.60 (m, 3H), 7.42 (d, J = 8.0 Hz, 1H), 7.32 - 7.20 (m, 4H), 7.05 (d, J = 7.5 Hz, 1H), 7.00 - 6.95 (m, 1H), 6.40 (s, 1H), 3.84 (s, 2H), 2.48 (s, 3H), 2.06 (s, 3H). MS: m/e = 320 (M++l).
[001048] Step-4 HN \ I N II
NH

[001049] 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 C under inert atmosphere. The reaction mixture was then stirred at -10 C 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. 'H-NMR (DMSO-d6, 500 MHz, at 80 C): 6 8.48 (bs, 1H), 8.07 (s, 1H), 7.88 (s, 1H), 7.69 - 7.58 (m, 4H), 7.28 (t, J -8.0 Hz, 1H), 7.16 (t, J

- 8.0 Hz, 2H), 6.91 - 6.85 (m, 2H), 6.75 (dd, J- 2.5, 15.3 Hz, 1H), 6.13 (d, J-15.0 Hz, 1H), 5.66 (d, J - 7.5 Hz, 1H), 4.60 (s, 2H), 2.95 (s, 3H), 2.12 (s, 3H). MS: m/e -374 (M++1).

[001050] Preparation of N-methyl-N-(4-(5-methyl-2-(phenylamino)pyrimidin-4-ylamino)benzyl)acrylamide 1-219 \ I H
HN

NI/
NI

H

[001051] 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. 'H NMR (DMSO-d6, 500 MHz at 80 C): 6 8.55 (s, 1H), 8.03 (s, 1H), 7.86 (s, 1 H), 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, 1H), 5.67 (d, J- 15.0 Hz, 1H), 4.58 (s, 2H), 2.96 (s, 3H), 2.10 (s, 3H). MS: m/e - 374 (M++1).

[001052] Preparation of N-(5-(5-acetyl-4-(4-acryloyl-3,4-dihydro-2H-benzo[b][1,4]oxazin-6-ylamino)pyrimidin-2-ylamino)pyridin-2-yl)-2,2,2-trifluoro-N-methylacetamide N") \ N\/CF3 N N N Y
I ~N 0 H

[001053] 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-).

[001054] Preparation of 1-(6-(5-fluoro-2-(4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)indolin-l-yl)prop-2-en-l-one 1-94 05:r N
HN

F I IN Z OMe H

[001055] 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-).

[001056] Preparation of N-(3-(5-fluoro-2-(6-(2-methoxyethoxy)pyridine-3-ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-103 HN N
H
F
NNN
N
H

[001057] 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. 'H NMR (CDC13 + trace of DMSO-d6) 6 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, 1H), 6.45 (m, 1H), 6.80 (m, 3H), 7.17 (m, I H), 7.29 (m, t H), 7.43 (m, t H), 7.49 (m, 1H), 7.60 (m, t H), 7.80 (dd, J- 2.8 & 9.2 Hz, 1H), 7.94 (d, J - 3.1 Hz, 1H), 8.14 (s, 1H), 8.32 (d, J- 2.9 Hz, 1H); LCMS :
m/e 425.1 (M+1).

[0010581 Preparation of N-(3-(5-fluoro-2-(4-(3-methylsulfonylpropoxy)phenyl)aminopyrimidin-4-ylamino)phenyl)acrylamide 1-97 HN N
~1 /
F N H \ O~SO
N N
H

[0010591 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. 'H NMR (CDC13 + trace of DMSO-d6) 6 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, 1H), 6.07 (m, 2H), 6.48 (d, J - 8.2 Hz, 1 H), 6.77 (m, 4H), 7.09 (d, J - 7.4 Hz, 1 H), 7.51 (d, J - 4.1 Hz, 1 H), 7.70 (br, 1 HA; LCMS :
m/e 486.1 (M+1).

[0010601 Preparation of N-(3-(5-fluoro-2-(6-(trideutereomethoxy)pyridine-3-ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-95 HN N

N NN
H

[0010611 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. 'H NMR (CDC13 + trace of DMSO-d6) 6 ppm: 5.78 (dd, J -3.7 & 7.8 Hz, 1H), 6.40 (m, 2H), 6.71 (d, J= 8.7 Hz, 1H), 7.3 (m, 3H), 7.75 (dd, J= 2.7 &
8.7 Hz, 1H), 7.82 (d, J - 4.6 Hz, 1H), 7.95 (s, 1H), 8.33 (d, J - 2.3 Hz, 1H); LCMS : m/e 384.1 (M+1).
[001062] The intermediate 6-(trideutratedmethoxy)pyridin-3-amine was prepared by the scheme shown below.

02N step-1 H2N

A) NaH, CD30D, it; BH3=NMe3, Pd(OH)2 [001063] Step 1 [001064] To NaH (60%, 0.30 g) in 5 mL of CD30D at 0 C 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). 'H NMR (CDC13) 6 ppm: 3.30 (br, 2H), 6.60 (d, J = 8.7 Hz, 1H), 7.03 (dd, J = 3.2 & 8.7 Hz, 1H), 7.66 (d, J = 3.2 Hz, 1H).

[001065] Preparation of N-(3-(5-fluoro-2(3,4,5-trimethoxyphenylamino)pyrimidin-yloxy)phenyl)acrylamide 1-148 HN
/ zI-O Me F N \ OMe N N OMe H

[001066] 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+1 ].

[0010671 Preparation of 3-methyl-l-(3-(5-methyl-2-(phenylamino)pyrimidin-4-ylamino)phenyl)but-2-en-l-one 1-232 NH
N N
H

[0010681 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. 'H NMR (CDC13) 6 ppm: 1.97 (s, 3H), 2.15 (s, 3H), 2.22 (s, 3H), 6.47 (s, 1H), 6.71 (s, 1H), 6.97 (t, J- 9.8 Hz, 1H), 7.17 (s, 1H), 7.24 (t, J- 10.36 Hz, 1H), 7.27 (s, 1H), 7.44 (t, J- 10.64 Hz, 1H), 7.53 (d, J- 10.48 Hz, 2H), 7.68 (d, J- 10.28 Hz, 1H), 7.94 (d, J- 10 Hz, 1H), 7.98 (s, 1H); LCMS : m/e 359 (M+1).

[0010691 Preparation of 1-(3-(5-methyl-2-phenylamino)pyrimidin-4-ylamino(piperidin-l-yl)prop-2-en-one 1-27 HN

N NH

[0010701 The title compound was prepared according to the schemes, steps and intermediates described in Example 1 using 1-tert-butoxycarbonyl-3-aminopiperidine in place of 1 in step-1. 'H NMR (DMSO-d6) 6 ppm: 1.30-1.50 (m, 1H), 1.55-1.75 (m, 1H), 1.75-1.90 (m, 1H), 1.92 (s, 3H), 1.95-2.05 (m, 1H), 2.75-3.31 (m, 2H), 3.99-4.09 (m, 2H), 4.10-4.15 & 4.40-4.47 (m, 1H), 5.49 & 5.70 (d, J - 10.8 Hz & d, J - 9.2 Hz respectively, together 1H), 6.02 &
6.13 (d, J- 17.6 Hz & d, J- 16.8 Hz respectively, together 1H), 6.25-6.40 (m, 1H), 6.63 & 6.80-6.90 (dd, J- 10.8, 16.8 Hz & mrespectively, together 1H), 6.75-6.85 (m, 1H), 7.15 (t, J- 8 Hz, 2H), 7.69 (bs, 3H), 8.81 (s, 1H); LCMS: m/e 337.8 (M+1).

[0010711 Preparation of 3-(4-(2-acryloyl- 1,2,3,4-tetrahydroisoquinolin-6-ylamino)-5-methylpyrimidin-2-ylamino)benzenesulfonamide 1-40 \ I N
HN N

[0010721 The title compound was prepared according to the schemes, steps and intermediates described in Example 1 using 6-amino-2-tert-butoxycarbonyl-1,2,3,4-tetrahydroisoquinoline in place of 1 in step-1. 'H NMR (DMSO-d6) 6 ppm: 2.10 (s, 3H), 2.80-2.83 (m, 2H), 3.75-3.90 (m, 2H), 4.66 (s, 1H), 4.76 (s, 1H), 5.71-5.74 (m, 1H), 6.16 (dd, J- 2.32 & 16.76 Hz, 1H), 6.87-6.91 (m, 1H), 7.13-7.18 (m, 1H), 7.25-7.31 (m, 4H), 7.53-7.57 (m, 2H), 7.90 (s, 1H), 8.05 (s, 2H), 8.28 (s, 1H), 9.31 (s, 1H); LCMS: m/e 464.8 (M+1).

[0010731 Preparation of (S)-N-(3-(5-fluoro-2-(tetrahydrofuran-3-yloxy)pyridine-ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-54 HN

HN
C

F ZL~N 01"
N N
H

[0010741 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+l].

[0010751 Preparation of N-(3-(5-trifluoromethyl-2-(phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-245 HN N
H
F3C \

N N 'JO

[0010761 The title compound was prepared according to the schemes, steps, and intermediates described below.

1NH2 \
CI I H2N / N-Boc NNCI step-1 N N step-2 H
A

OO
- / NH
HN \ N Boc HN\ I CI

/
F3C I ~ N / I F3C I NI \ NH D
p Ni N \ step-3 ~ step 4 HN N
H
F3C 1 \
N N 'O

A) 2, ZnC12, DCE, t-BuOH (1:1), 0 C, 30 min; B) 4, DMF, DIEPA, 70 C, 16 hr;
C) TFA, DCM, rt, 1 hr; D) 7, TEA, DCM.
[0010771 Step-1 CI

N N
H

[0010781 To a cold (0 C) 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%).
[001079] Step_2 aZz~~'N-Boc HN HF3C \
N N 'O

[001080] 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 C
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%).
[001081] Step-3 HN \ NH2 N NJO
H

[001082] To a solution of 6 (0.25 g, 0.63 mmol) in CH2C12 (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 CH2C12, washed with 10% aqueous NaHCO3 solution, dried (Na2SO4), filtered, and evaporated under reduced pressure to provide the free amine as white solid.
[001083] Step-4 HN N
H
F3C I I \
N N
H

[0010841 To a stirred solution of 6 (0.2 g) in DCM (20 mL) under argon atmosphere cooled to -40 C was added triethylamine followed by dropwise addition of 7 (0.069 g, 0.686 mmol).
The resulting mixture was stirred at -40 C 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. 'H NMR (200 MHz, CD30D) 6 8.25 (s, 1H), 7.80 (s, 1H), 7.60-7.05 (m, 7H), 6.90 (m, 1H), 6.35 (m, 2H), 5.75 (dd, J- 8.0, 2.0 Hz, 1H).

[0010851 Preparation of N-(3-(5-trifluoromethyl-2-(3-methoxyphenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-242 HN N
H

N H N OMe [0010861 The title compound was prepared according to the schemes, steps and intermediates described in Example 189 using 3-methoxyaniline in place of 2 in step-l. 'H
NMR (200 MHz, CD30D) 6 8.31 (s, 1H), 7.84 (s, 1H), 7.59 (m, 1H), 7.37-7.09 (m, 5H) 6.53 (m, 1H), 6.41 (m, 2H), 5.79 (dd, J- 8.0, 2.0, Hz, 1H), 3.66 (s, 3H).

[0010871 Preparation of N-(4-(5-trifluoromethyl-2-(3-methoxyphenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-236 H
/ I N
HN \ O

N H OMe [001088] The title compound was prepared according to the schemes, steps and intermediates described in Example 189 using 3-methoxyaniline in place of 2 in step-1 and 4-amino-N-tert-butoxycarbonylaniline in place of 4 in step-2. 'H NMR (200 MHz, CD30D) 6 8.27 (s, I H), 7.70 (d, J - 6.0Hz) I H), 7.46 (d, J - 6.0Hz, I H), 7.09 (brs, I H), 7.07 (m, 2H) 6.51 (m, 1H), 6.44 (m, 2H), 5.80 (dd, J- 8.0, 2.O Hz, 1H), 3.56 (s, 3H).

[001089] Preparation of N-(4-(5-trifluoromethyl-2-(3-methoxyphenylamino)pyrimidin-4-ylamino)phenyl)methylacrylamide 1-235 HN

HN
C
N

NN OMe H

[001090] The title compound was prepared according to the schemes, steps and intermediates described in Example 189 using 3-methoxyaniline in place of 2 in step-1 and 4-aminophenylmethyl-N-tert-butoxycarbonylamine in place of 4 in step-2. 1H NMR
(200 MHz, CD30D) 6 8.30 (s, 1H), 7.49 (d, J- 8.0 Hz, 1H), 7.37 (d, J- 8.0 Hz, 1H), 7.10 (m, 3H), 6.60 (m, 1H) 6.34 (m, 2H), 5.75 (dd, J- 8.0, 2.0 Hz, 1H), 4.51(s, 2H), 3.68 (s, 3H).

[001091] Preparation of N-(4-chloro-3-(5-trifluoromethyl-2-(3-methoxyphenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-227 HN lk~
CI
HN

F3C N COMe N H

[001092] The title compound was prepared according to the schemes, steps and intermediates described in Example 189 using 3-methoxyaniline in place of 2 in step-1 and N-tert-butoxycarbony-3-amino- 6-chloroaniline in place of 4 in step-2. iH NMR
(200 MHz, CD30D) 6 8.33 (s, 1H), 6 8.08 (s, 1H), 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, 1H), 3.71 (s, 3H).

[001093] Preparation of N-(3-(5-trifluoromethyl-2-(3-methoxyphenylamino)pyrimidin-4-ylamino)phenyl)methylacrylamide 1-226 H
N

HN

F3C %N Na J~

N H We [001094] The title compound was prepared according to the schemes, steps and intermediates described in Example 189 using 3-methoxyaniline in place of 2 in step-1 and 3-aminophenylmethyl-N-tert-butoxycarbonylamine in place of 4 in step-2. 1H NMR
(200 MHz, CD30D) 6 8.31 (s, I H), 7.71-7.33 (m, 3H), 7.21-7.08 (m, 4H), 6.57 (m, I H), 6.26 (d, J - 4Hz, 2H), 5.69 (dd, J- 8.0, 2.0 Hz, 1H), 4.47 (s, 2H), 3.67 (s, 3H).

[001095] Preparation of N-(4-(5-trifluoromethyl-2-(6-methoxypyridin-3-ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-218 HN N
F3C N j 0"
I
N N N
H

[001096] 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, CD30D) 6 8.21 (s, 1H), 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, 1H), 3.81 (s, 3H).

[001097] Preparation of N-(4-(5-trifluoromethyl-2-(5-methoxypyridin-3-ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-214 H
N

HN

N N COMe H

[001098] 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].

[0010991 Preparation of 1-(3-(5-trifluoromethyl-2-(3-methoxyphenylamino)pyrimidin-4-ylamino)phenyl)-3-methyl-but-2-en-1-one 1-225 HN

N ~

N \ OMe H

[0011001 The title compound was prepared according to the schemes, steps and intermediates described in Example 189 using 3-methoxyaniline in place of 2 in step-1 and 1-(3-aminophenyl)-3-methylbut-2-en-l-one in place of 4 in step-2. iH NMR (200 MHz, CDC13) 6 8.33 (s, 1H), 6 8.38 (s, 1H), 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).
[0011011 1-(3-Aminophenyl)-3-methylbut-2-en-l-one was prepared according to the scheme, steps, and intermediates described below.

H02C 1 NH2 step-1 HO2C NHBoc step-2 O NHBoc BrMg",,~_ step-3 \ NHBoc step-4 NH2 A) Boc20, NEt3, DMAP, DCM; B) NHMe(OMe)-HC1, TBTU, DCM, 0 C to rt; C) 4, THE, to rt; D) TFA, DCM.
[0011021 Step-1 HO2C JaNHBoc [001103] Di-tert-butyldicarbonate (6.54 g, 30 mmol, 1.5 eq.) was added to a solution of 1 (2.70 g, 20 mmol) in CH2C12 (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 CaC12 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
HC1 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%).
[001104] Sten_2 O"N \ NHBoc [001105] 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 C was added NEt3 (2.7 mL, 19 mmol, 3 eq.). The mixture was stirred for 30 min at 0 C 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.
[001106] Step-3 NHBoc [001107] To a solution of 3 (546 mg, 1.95 mmol) in THE (3 mL) at 0 C under At 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 C 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.

[001108] Len-4 [001109] A sample of 400 mg of 5 was treated with 5 mL 1:3 CH2C12 :
trifluoroacetic acid and the resulting solution stirred at room temperature for 15 minutes. The solvents were removed in vacuo and the residue redissolved in CH2C12 and re-evaporated three times. The residue was again taken up in CH2C12 and the solution washed with saturated sodium bicarbonate solution. The CH2C12 layer was dried over sodium sulphate, filtered and evaporated to afford 6 as a white solid that was used directly in the next reaction.

[001110] Preparation of 1-(3-(2-(3-Methoxy-phenylamino)-5-trifluoromethyl-pyrimidin-4-ylamino)-cyclohexyl)-3-methyl-but-2-en-l-one 1-213 HN

N 5:;~' NN OMe H

[001111] The title compound was prepared according to the schemes, steps and intermediates described in Example 189 using 3-methoxyaniline in place of 2 in step-1 and (d,l)-cis-1-(3-Amino-cyclohexyl)-3-methyl-but-2-en-l-one in place of 4 in step-2. 'H
NMR (200 MHz, CDC13) 6 8.07 (s, 1H), 7.33 (s, 1H), 7.19-7.0 (m, 3H), 6.54 (d, J- 2.7 Hz, 1H), 6.02 (s, 1H), 4.04 (m, 1H), 3.75 (s, 3H), 2.50 (m, 1H), 2.10 (m, 1H), 1.89-1.15 (m, 14H).
[001112] (D,L)-cis-1-(3-Amino-cyclohexyl)-3-methyl-but-2-en-l-one was prepared according to the scheme, steps, and intermediates described below.

H2N OH BocHN OH BocHN N,O~

step-1 step-2 BrMg~ O O
4 BocHN H2N
step-3 step-4 C D

A) Boc2O, Na2CO3, acetone, H20; B) NHMe(OMe)-HC1, TBTU, DCM, 0 C to rt; C) 4, THF, 0 C to rt; D) TFA, DCM.
[001113] Step-1 BocH N
OH

[001114] To a stirred solution of ( )-1 (4.05 g, 28.2 mmol) in water (150 mL) 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 25 C 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.
[001115] Step_2 BocHN N,O1~1 [001116] To a stirred solution of 2 (2.45 g, 10.1 mmol), TBTU (3.44 g, 10.6 mmol, 1.05 eq) and N-methyl-N-methoxyamine hydrochloride (1.03 g, 10.6 mmol, 1.05 eq) in CH2C12 (40 mL) at 0 C was added triethylamine (4.25 mL, 30.3 mmol, 3 eq). The mixture was stirred at 0 C for 20 min and the bath was removed and stirring was continued for 3 h at 25 C.
After quenching with water, the CH2C12 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.
[001117] Step-3 BocH N -C, [001118] To a solution of 3 (1.23 g, 4.32 mmol) in THE (20 mL) at 0 C 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 C for 30 min, then at rt for 1 h. The reaction mixture was cooled to 0 C 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.
[001119] Step-4 H2N -.10 [001120] A sample of 500 mg of 5 was treated with 6 mL 1:3 CH2C12 :
trifluoroacetic acid and the resulting solution stirred at room temperature for 15 minutes. The solvents were removed in vacuo and the residue redissolved in CH2C12 and re-evaporated three times. The residue was again taken up in CH2C12 and the solution washed with saturated sodium bicarbonate solution. The CH2C12 layer was dried over sodium sulphate, filtered and evaporated to afford 6 as a white solid that was used directly without purification.

[001121] Preparation of 1-(5-(5-trifluoromethyl-2-(3-methoxyphenylamino)pyrimidin-4-yl)amino-1,3-dihydroisoindol-2-yl)2-propen-l-one 1-132 N
HN JOC

N ~
NN OMe H

[001122] The title compound was prepared according to the schemes, steps and intermediates described in Example 189 using 3-methoxyaniline 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].

[001123] Preparation of 3-(2-(2-acryloylisoindolin-5-ylamino)-5-fluoropyrimidin-4-ylamino)benzonitrile 1-106 /
P
NC \ NH N
F

N N
H

[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-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 [001125] Preparation of N-(3-(5-fluoro-4-((6-(trifluoromethyl)pyridin-3-yl)methylamino)pyrimidin-2-ylamino)phenyl)acrylamide 1-53 HN "' N
F N I F
F
NNH F

b,, 0 N
H

[001126] 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-l. LC/MS (RT =
2.805/(M + H)) 433.0 [001127] Preparation of N-(3-(4-((2,3-dihydrobenzofuran-5-yl)methylamino)-5-fluoropyrimidin-2-ylamino)phenyl)acrylamide 1-6 F HN -'~"NCO
'N 0 N" 'NH

6~NL

[001128] 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 [001129] Preparation of N-(3-(5-fluoro-2-(4-methoxybenzylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-241 ~I
HN N
F H

NNH

O

[001130] 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 [001131] Preparation of Ni-(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 HN N"
H
N

NON O""'~NH
H

HH

N
IC
HH

0 If ~0NI'-~0--O~N
H

[001132] The title compound was prepared according to the schemes steps and intermediates described below.

' e HN I N v HN N
H H
I )Ij< step- I N~a Ia"'~ 0 N 0 N N O_~NH2 N N
H H A H

step-2 B
HN N'J"~
H
`N
N-1N \ I O"-' NH
H

HH

O=( S
N

LI,10"_~_0__'~ )H
A) TFA, DCM; B) N-Biotinyl-NH-(PEG)2-COOH-DIPEA, HOBt, EDC, NMM, DMF.
[001133] Step-1 ~I
HN N
H
N

N)N I O--~NH2 H

[001134] 1-45 (97 mg, 0.19 mmol; synthesis of 1-45 provided in Example 62) was dissolved in DCM (10 mL). Trifluoroacetic acid (200 L) 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) [001135] Step-2 HN N"
H
,jam :;e N N O^~NH
H

HH

O=( JCS
N
HH
O I'f H

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 L, 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).

[0011361 Preparation of N-(3-(3-(4-(3-acrylamidophenylamino)-5-methylpyrimidin-ylamino)phenoxy)propyl)-5-((3aS,4S,6aR)-2-oxohexahydro-lH-thieno[3,4-d]imidazol-4-yl)pentanamide 1-237 o HN N
H
N "a 0"^"v `N H
H
S
O
.,m %H

NH
HN` /

O

[0011371 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 [0011381 Preparation of (R)-N-(3-(5-fluoro-2-(3-fluoro-4-(tetrahydrofuran-3-yloxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-316 HN N
F j 0 N F ~/
H

[0011391 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. 'H NMR (DMSO-d6) 6 ppm: 1.85-2.00 (m, 1H), 2.20 (m, 1H), 3.70-3.90 (m, 4H), 4.90 (s, 1H), 5.73 (dd, J - 1.56 & 10.04 Hz, 1H), 6.23 (dd, J - 1.76 & 17.00 Hz, 1H), 6.44 (dd, J - 10.08 & 16.88 Hz, 1H), 7.28 (t, J - 8.04 Hz, 1H), 7.40-7.47 (m, 2H), 7.67-7.71 (m, 2H), 7.68 (dd, J - 1.96 & 14.08 Hz, 1H), 7.92 (s, 1H), 8.1 (d, J - 3.64 Hz, 1H), 9.21 (s, 1H), 9.44 (s, 1H), 10.12 (s, 1H); LCMS : m/e 452.0 (M-1).

[0011401 Preparation of 1-(4-(5-fluoro-2-(3-fluoro-4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-3-methylbut-2-en-l-one 1-HN
F \ / II 0, N N F
H

[0011411 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-d6) 6 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, 1H), 7.08 (t, J - 9.44 Hz, 1H), 7,27 (d, J - 8.88 Hz, I H), 7.74 (dd, J - 2.44 & 14,24 Hz, 1H), 7.93 (d, J - 8.96 Hz, 2H), 7.98 (d, J -8.92 Hz, 2H), 8.20 (d, J- 3.64 Hz, 1H), 9.35 (s, 1H), 9.73 (s, 1H); LCMS : m/e 455 (M+1).

[001142] Preparation of 1-(3-(5-fluoro-2-(3-fluoro-4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-3-methylbut-2-en-1-one 1-HN
F IAN /I 0~`0 N N F
H

[001143] 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. 'H NMR (DMSO-d6) b 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, 1H), 6.97 (t, J - 9.52 Hz, 1H), 7.26 (bd, J
- 8.32 Hz, 1H), 7.48 (t, J - 7.92 Hz, 1H), 7.62-7.67 (m, 2H), 8.08 (bd, J -7.04 Hz, 1H), 8.15-8.16 (m, 2H), 9.29 (s, 1H), 9.58 (s, 1H); LCMS : m/e 455 (M+l).

[001144] Preparation of 1-(4-(5-fluoro-2-(3-fluoro-4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-2-methylprop-2-en- l -one 1-324 /I

HN

N N F
H

[001145] 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. 1H NMR (DMSO-d6) b 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, 1H), 5.94 (s, 1H), 7.07 (t, J - 9.32 Hz, 1H), 7.26 (t, J - 9.4 Hz, 1H), 7.72-7.76 (m, 3H), 7.99 (d, J - 8.44 Hz, 2H), 8.21 (d, J - 3.56 Hz, 1H), 9.38 (s, 1H), 9.75 (s, 1H); LCMS : m/e 441.2 (M+1).

[0011461 Preparation of 1-(4-(5-fluoro-2-(3-fluoro-4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-3-methylbut-3-en-2-one 1-HN

N N F
H

[0011471 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. iH NMR (DMSO-d6) 8 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, 1H), 6.27 (s, 1H), 7.01 (t, J - 9.44 Hz, 1H), 7.15 (d, J - 8.52 Hz, 2H), 7.28 (d, J - 8.88 Hz, 1H), 7.64-7.70 (m, 3H), 8.08 (d, J -3.72 Hz, 1H), 9.19 (s, 1H), 9.33 (s, 1H); LCMS : m/e 455.3 (M+l).

[0011481 Preparation of l-(4-(5-fluoro-2-(3-fluoro-4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-4-methylpent-3-en-2-one \
\ I 0 HN

N N F
H

[0011491 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. 'H NMR
(DMSO-d6) 8 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, 1H), 7.01 (t, J = 8.68 Hz, 1H), 7.16 (d, J - 8.48 Hz, 2H), 7.26 (d, J = 8.96 Hz, 1H), 7.65-7.72 (m, 3H), 8.08 (d, J = 3.72 Hz, 1H), 9.20 (s, 1H), 9.34 (s, 1H); LCMS : m/e 469.3 (M+l).

[001150] Preparation of 1-(3-(5-fluoro-2-(3-fluoro-4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-2-methylprop-2-en-l-one O
HN

F N O~/\O
I
N N / F
H

[001151] The title compound was prepared according to the schemes, steps and intermediates described in Example 112, by using 1,3-fluoro-4-(2-methoxyethoxy)aniline in place of 4 in step 2 and isopropenylmagnesium bromide in place of 8 in step 5.
'H NMR
(DMSO-d6) 8 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, 1H), 6.00 (s, 1H), 6.99 (t, J - 9.24 Hz, 1H), 7.26 (d, J - 8.92 Hz, 1H), 7.39 (d, J - 7.56 Hz, 1H), 7.46 (t, J - 7.72 Hz, 1H), 7.62 (bd, J - 14.4 Hz, 1H), 7.93 (s, 1H), 8.12-8.14 (m, 2H), 9.25 (s, 1H), 9.58 (s, 1H); LCMS : m/e 441.2 (M+1).

[001152] Preparation of l-(3-(5-fluoro-2-(3-fluoro-4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-3-methylbut-3-en-2-one 1-/
HN
F IAN 0"'-`0 N N F
H

[001153] 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) 6 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, 1H), 6.23 (s, 1H), 6.92 (d, J -7.52 Hz, 1H), 7.02 (t, J - 9.4 Hz, 1H), 7.27 (t, J - 7.8 Hz, 2H), 7.50 (s, 1H), 7.66-7.72 (m, 2H), 8.10 (d, J- 3.56 Hz, 1H), 9.21 (s, 1H), 9.36 (s, 1H); LCMS : m/e 455.1 (M+l).

[001154] Preparation of 2-((3-(5-fluoro-2-(3-fluoro-4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)(hydroxy)methyl)acrylonitrile 1-326 HO
CN
HN

F N I O~/\O
Z~N
N N / F
H

[001155] 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 (DMSO-d6) 8 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, 1H), 6.13 (s, 1H), 6.19 (s, 1H), 6.31 (d, J = 4.04 Hz, 1H), 7.03 (t, J =
9.24 Hz, 1H), 7.10 (d, J = 7.44 Hz, 1H), 7.28 (d, J = 8.72 Hz, 1H), 7.36 (t, J
= 7.8 Hz, 1H), 7.62 (s, 111), 7.66 (dd, J = 2.32 & 14.44 Hz, 1H), 7.89 (d, J = 8.2 Hz, I H), 8.10 (d, J = 3.68 Hz, 1H), 9.14 (s, 1H), 9.45 (s, 1H); LCMS : m/e 454 (M+1).

[001156] Preparation of 2-((4-(5-fluoro-2-(3-fluoro-4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)(hydroxy)methyl)acrylonitrile 1-327 OH

HN\ I CN
F N
N /F
H

[001157] 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-d6) 6 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, 1H), 6.11 (s, 1H), 6.21 (s, 1H), 6.24 (d, J= 4.12 Hz, 1H), 7.01 (t, J= 9.4 Hz, 1H), 7.29-7.34 (m, 3H), 7.68 (dd, J = 2.2 & 14.16 Hz, 1H), 7.77 (d, J = 8.52 Hz, 2H), 8.11 (d, J
= 3.68 Hz, 1H), 9.23 (s, 1H), 9.42 (s, 1H); LCMS : m/e 454.0 (M+1).

[001158] Preparation of N-(3-(2-(4-chloro-3-(2-hydroxy-2-methylpropoxy)phenylamino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-249 HN N
F N C I

N N \ I O OH
H

[001159] 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. 'H NMR (DMSO-d6) 6 ppm: 1.20 (s, 6H), 3.61 (s, 2H), 4.61 (s, 1H), 5.75 (d, J - 11.4 Hz, 1H), 6.24 (d, J - 18.36 Hz, 1H), 6.44 (dd, J - 10.32 &
17,08 Hz, 1H), 7.13 (d, J - 8.64 Hz, 1H), 7.28 (t, J - 8 Hz, 1H), 7.37-7.44 (m, 3H), 7.55 (d, J
7.08 Hz, 1 H), 7.93 (s, 1 H), 8.12 (d, J - 3.44 Hz, 1 H), 9.23 (s, 1 H), 9.47 (s, 1 H), 10.11 (s, 1 H);
LCMS : m/e 472.0 (M+l).

[001160] Preparation of N-(3-(5-fluoro-2-(3-fluoro-4-(2-hydroxy-2-methylpropoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-315 HN H
F,11 O'OH
N N F
H

[001161] 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. 'H NMR (DMSO-d6) 6 ppm: 1.19 (s, 6H), 3.67 (s, 2H), 4.62 (s, 1 H), 5.75 (d, J - 10.4 Hz, 1H), 6.25 (d, J - 17.2 Hz, 1H), 6.45 (dd, J - 10 & 16.8 Hz, I
H), 6.94 (t, J - 9.2 Hz, 1 H), 7.29 (t, J - 8 Hz, 2H), 7.43 (d, J - 8.4 Hz, 1 H), 7.49 (d, J - 7.6 Hz, 1 H), 7.67 (d, J
13.6 Hz, t H), 7.94 (s, t H), 8.11 (d, J - 3.6 Hz, 1 H), 9.19 (s, t H), 9.45 (s, 1 H), 10.14 (s, t 1l);
LCMS : m/e 456 (M-1).

[001162] Preparation of N-(3-(5-fluoro-2-(3-fluoro-4-(1-hydroxypropan-2-yloxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-333 I
HN \ N

F N O-rOH
N N IF
H

[001163] 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) 6 ppm: 1.16 (d, J - 6.12 Hz, 3H), 3.40-3.46 (m, 1H), 3.50-3.56 (m, 1H), 4.22 (sextet, J - 5.6 Hz, 1H), 4.84 (t, J - 5.68 Hz, 1H), 5.75 (dd, J
1.96 & 10.08 Hz, 1H), 6.25 (dd, J - 1.92 & 16.92 Hz, 1H), 6.46 (dd, J - 10.08 & 16.92 Hz, 1H), 6.98 (t, J - 9.32 Hz, 1H), 7.26-7.31 (m, 2H), 7.43 (d, J - 8.76 Hz, 1H), 7.49 (d, J - 8 Hz, 1H), 7.68 (dd, J - 2.44 & 14.28 Hz, 1H), 7.95 (s, 1H), 8.11 (d, J - 3.68 Hz, 1H), 9.23 (s, 1H), 9.46 (s, 1H), 10.17 (s, 1H); LCMS : m/e 442.2 (M+1).

[001164] Preparation of N-(3-(2-(4-(2,3-dihydroxypropoxy)-3-fluorophenylamino)-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-334 \ IN OH
HN
H
F I ANI O OH
N N F
H

[001165] 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. 'H NMR (DMSO-d6) 8 ppm: 3.42 (t, J = 5.6 Hz, 2H), 3.7-3.8 (m, 1H), 3.8-3.9 (m, 1H), 3.94 (dd, J = 4.36 & 9.92 Hz, 1H), 4.65 (t, J = 5.64 Hz, 1H), 4.93(d, J = 5.08 Hz, 1H), 5.7-5.8 (m, 1H), 6.24 (dd, J = 1.64 & 16.84 Hz, 1H), 6.44 (dd, J = 10 &
16.96 Hz, 1H), 6.94 (t, J = 9.32 Hz, 1H), 7.28 (t, J = 7.96 Hz, 2H), 7.40 (d, J = 8.28 Hz, 1H), 7.49 (d, J = 7.44 Hz, 1H), 7.66 (d, J = 14.24 Hz, 1H), 7.92 (s, 1H), 8.09 (d, J = 3.6 Hz, 1H), 9.17 (s, 1H), 9.45 (s, 1H), 10.15 (s, 1H); LCMS : m/e 456 (M-1).

[001166] Preparation of N-(3 -(2-(4- chloro-3 -(1-hydroxy-2-methylpropan-2-yloxy)phenylamino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-336 HN\ INL%
F H CI
IN
NJ~N OOH
Y
H

[001167] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 4-chloro-3-(1-hydroxy-2-methylpropan-2-yloxy)aniline in place of 4 in step 2. 'H NMR (DMSO-d6) 6 ppm: 1.22 (s, 6H), 3.47 (d, J = 5.88 Hz, 2H), 4.88 (t, J = 5.84 Hz, 1H), 5.75 (dd, J = 3.24 & 10 Hz, 1H), 6.25 (dd, J = 2 & 16.92 Hz, 1H), 6.46 (dd, J = 10.12 & 17.08 Hz, 1H), 7.15 (d, J = 8.8 Hz, 1H), 7.31 (t, J
= 8.2 Hz, 1H), 7.40-7.45 (m, 1H), 7.51-7.60 (m, 3H), 7.93 (s, 1H), 8.13 (d, J = 3.56 Hz, 1H), 9.24 (s, 1H), 9.47 (s, 1H), 10.12 (s, 1H); LCMS : m/e 472.2 (M+1).

[001168] Preparation of N-(3-(2-(4-chloro-3-(1-hydroxypropan-2-yloxy)phenylamino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-337 HN N
F,, j CI
N
N I N \ I OOH
H

[0011691 The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 4-chloro-3-(1-hydroxypropan-2-yloxy)aniline in place of 4 in step 2. 'H NMR (DMSO-d6) 6 ppm: 1.18 (d, J = 6.12 Hz, 3H), 3.40-3.47 (m, 1H), 3.50-3.56 (m, 1H), 4.20-4.30 (m, 1H), 4.82 (t, J = 5.6 Hz, 1H), 5.75 (dd, J = 1.88 & 10.08 Hz, 1H), 6.25 (dd, J = 1.92 & 16.92 Hz, 1H), 6.45 (dd, J = 10.08 & 16.92 Hz, 1H), 7.12 (d, J =
8.76 Hz, 1H), 7.29 (t, J = 8.08 Hz, 1H), 7.40-7.44 (m, 3H), 7.52 (d, J = 8.44 Hz, 1H), 7.91 (s, 1H), 8.12 (d, J = 3.64 Hz, 1H), 9.21 (s, 1H), 9.45 (s, 1H), 10.12 (s, 1H);
LCMS : m/e 458.0 (M+ 1).

[0011701 Preparation of N-(3-(2-(4-(2,3-dihydroxypropoxy)phenylamino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-335 HN I N OH
H

F "zN OH
N N
H

[0011711 The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 4-(1-hydroxypropan-2-yloxy)aniline in place of 4 in step 2. iH NMR (DMSO-d6) 6 ppm: 3.43 (dd, J = 0.64 & 6.84 Hz, 2H), 3.70-3.80 (m, 2H), 3.85-3.95 (m, 1H), 4.62 (t, J = 5.6 Hz, 1H), 4.88 (d, J = 4.76 Hz, 1H), 5.75 (dd, J = 1.76 &
10.08 Hz, 1H), 6.25 (dd, J = 1.72 & 16.92 Hz, 1H), 6.45 (dd, J = 10.08 & 16.88 Hz, 1H), 6.74 (d, J = 9 Hz, 2H), 7.27 (t, J = 8.08 Hz, 1H), 7.39 (d, J = 8.04 Hz, 1H), 7.38-7.53 (m, 3H), 7.93 (s, 1H), 8.05 (d, J = 3.68 Hz, 1H), 8.93 (s, 1H), 9.35 (s, 1H), 10.11 (s, 1H);
LCMS : m/e 440.3 (M+ 1).

[0011721 Preparation of (R)-N-(3-(2-(4-chloro-3-(tetrahydrofuran-3-yloxy)phenylamino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-341 F j CI
N
Ni N 01' CO

[0011731 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. 'H NMR (DMSO-d6) 6 ppm: 1.85-1.95 (m, 1H), 2.0-2.15 (m, 1H), 3.60-3.70 (m, 1H), 3.73-3.83 (m, 3H), 4.68 (s, 1H), 5.74 (dt, J -1.92 & 10.0 Hz, 1H), 6.23 (dd, J - 1.8 8 & 16.92 Hz, 1 H), 6.43 (dd, J - 10.12 & 16.96 Hz, 1 H), 7.14 (d, J - 8.72 Hz, 1H), 7.29 (t, J - 8.08 Hz, 1H), 7.33 (dd, J - 4,16 & 8.76 Hz, 1H), 7.41-7.47 (m, 3H), 7.90 (s, 1H), 8.13 (d, J - 3.56 Hz, 1H), 9.28 (s, 1H), 9.47 (s, 1H), 10.13 (s, 1H);
LCMS : rrt/e 469.8 (M+ 1).

[0011741 Preparation of N-(3-(5-fluoro-2-(3-fluoro-4-(1-hydroxy-2-methylpropan-yloxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-332 ~I
HN NL
F - _ Xo<OH

N N F
H

[0011751 The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 3-fluoro-4-(1-hydroxy-2-methylpropan-2-yloxy)aniline in place of 4 in step 2, 'H NMR (DMSO-d6) 6 ppm: 1.13 (s, 6H), 3.36 (d, J - 5.84 Hz, 2H), 4.86 (t, J - 5.84 Hz, 1H), 5.73 (dd, J - 1.96 & 10.04 Hz, 1H), 6.24 (dd, J - 1.96 &
16.96 Hz, 1H), 6.44 (dd, J - 10.08 & 16.92 Hz, 1H), 6.95 (t, J - 9.16 Hz, 1H), 7.23 (dd, J -1.64 & 8.96 Hz, 1H), 7.28 (t, J - 8.12 Hz, 1H), 7.43 (d, J - 8.8 Hz, 1H), 7.48 (d, J - 7.92 Hz, 1H), 7.70 (dd, J - 2.48 & 13.84 Hz, 1H), 7.92 (s, 1H), 8.11 (d, J - 3.68 Hz, 1H), 9.25 (s, 1H), 9.45 (s, 1H), 10.10 (s, 1H); LCMS: m/e 456.2 (M+1).

[0011761 Preparation of N-(3-(2-(3-(2,3-dihydroxypropoxy)phenylamino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-339 ~I
HN N
F H
/
N,N \ O
H
OH
OH

[0011771 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. iH NMR (MeOD) 6 ppm: 3.59-3.69 (m, 2H), 3.88-3.98 (m, 3H), 5.78 (dd, J - 2.16 &
9.6 Hz, 1H), 6.36 (dd, J- 2.24 & 17.04 Hz, 1H), 6.44 (dd, J- 9.56 & 16.96 Hz, 1H), 6.54-6.57 (m, 1H), 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, 1H), 8.09 (s, 1H); LCMS : m/e 440.1 (M+1).

[0011781 Preparation of N-(4-(5-fluoro-2-(3-fluoro-4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-351 H
al N

HN
F - , , / I O
~
N N F
H

[001179] 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-d6) 6 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, 1H), 6.25 (dd, J = 1.96 & 16.92 Hz, 1H), 6.44 (dd, J = 10.04 &
16.96 Hz, 1H), 7.02 (t, J = 9.48 Hz, 1H), 7.23 (bd, J = 7.44 Hz, 1H), 7.64 (d, J = 9 Hz, 2H), 7.70-7.74 (m, 3H), 8.07 (d, J= 3.72 Hz, 1H), 9.19 (s, 1H), 9.34 (s, 1H), 10.13 (s, 1H); LCMS :
m/e 442.0 (M+l).

[001180] Preparation of 2-((3-(5-fluoro-2-(6-(2-hydroxy-2-methylpropoxy)pyridin-3-ylamino)pyrimidin-4-ylamino)phenyl)(hydroxy)methyl)acrylonitrile 1-312 N
HO

HN

I
N N N
H

[001181] 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) b ppm: 1.18 (s, 6H), 3.98 (s, 2H), 4.61 (s, 1H), 5.31 (d, J= 3.88 Hz, 1H), 6.13 (s, 1H), 6.19 (s, 1H), 6.32 (d, J= 4 Hz, 1H), 6.75 (d, J= 8.88 Hz, 1H), 7.10 (d, J= 7.72 Hz, 1H), 7.33 (t, J= 7.84 Hz, 1H), 7.68 (s, 1H), 7.84 (d, J = 7.52 Hz, 1H), 7.96 (dd, J = 2.72 & 8.88 Hz, 1H), 8.08 (d, J = 3.64 Hz, 1H), 8.33 (d, J =
1.76 Hz, 1H), 9.06 (s, 1H), 9.44 (s, 1H); LCMS : m/e 451 (M+1).

[0011821 Preparation of 4-(4-(4-(3-acrylamidophenylamino)-5-fluoropyrimidin-2-ylamino)phenoxy)-N-methylpicolinamide 1-342 azzt~' 'C 1-1 NI O
NJ'N \ I &N
H

[0011831 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 [0011841 Preparation of (R)-1-(3-(3-fluoro-4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)piperidin-l-yl)prop-2-en-l-one 1-344 0'~
HN
F N O
N N / F
H

[0011851 The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using (R)-1-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.

[0011861 Preparation of (R)-l-(3-(4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)piperidin-1-yl)prop-2-en-l-one 1-345 N
HN
F N, N O,0 "/I I'll LN N
H

[0011871 The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using (R)-1-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 [0011881 Preparation of 4-(4-(4-(3-acrylamidophenylamino)-5-fluoropyrimidin-2-ylamino)phenoxy) pyridine 1-346 HN\I

F INNj 0 \
\ I I iN
NN
H

[0011891 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. LGMS (RT - 2.802/(M + H)) 500.2 [0011901 Preparation of 1-((R)-3-(5-fluoro-2-(4-((S)-tetrahydrofuran-3-yloxy)phenylamino)pyrimidin-4-ylamino)piperidin-l-yl)prop-2-en-1-one 1-347 0r HN
F ~ SO,.
I CO
N N
H

[0011911 The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using (R)-1-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. LGMS (M
+ H) 428.3.

[0011921 Preparation of 1-((R)-3-(5-fluoro-2-(4-((R)-tetrahydrofuran-3-yloxy)phenylamino)pyrimidin-4-ylamino)piperidin-l-yl)prop-2-en-1-one 1-348 HN

~N'I N
H

[0011931 The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using (R)-1-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. LGMS (M
+ H) 428.3.

[0011941 Preparation of N-(3-(2-(2,3-dihydrobenzo[b] 1,4]dioxin-6-ylamino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-349 \I

F,~ H
j N N O
H

[0011951 The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using 6-amino-2,3-dihydrobenzo[b]l,4]dioxane in place of 4 in step 2. LC/MS (M + H) 408.

[0011961 Preparation of 1-(6-(4-(3-chloro-4-(pyridine-2-ylmethoxy)phenylamino)-fluoropyrimidin-2-ylamino)-2H-benzo[b][l,4]oxazin-4(3H)-yl)prop-2-en-l-one 1-\ I 0 N IN
N
H
0 \%

[0011971 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.

[001198] Preparation of N-(3-(5-cyano-2-(3-fluoro-4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-350 HN

HN
NC I N 0~'-~ Oi N N F
H

[001199] 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 [001200] Preparation of N-(3-(5-trifluoromethyl-2-(3-fluoro-4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-352 HN' HN
F3C N 0"- 0 N N F
H

[001201] 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 [001202] Preparation of N-(3-(2-(4-chloro-3-(2-methoxyethoxy)phenylamino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-321 a HN N
F H CI
N

NN O
H

[001203] 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. 'H NMR (DMSO-d6) 6 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, 1H), 6.24 (dd, J- 1.8 & 16.88 Hz, 1H), 6.44 (dd, J
- 4.36 & 10.0 Hz, 1H), 7.13 (d, J - 8.72 Hz, 1H), 7.28 (t, J - 8.04 Hz, 1H), 7.33 (dd, J - 2.16 & 8.8 Hz, 1H), 7.41 (d, J - 7.96 Hz, 1H), 7.47-7.49 (m, 2H), 7.84 (s, 1H), 8.13 (d, J - 3.6 Hz, 1H), 9.27 (s, 1H), 9.47 (s, 1H), 10.12 (s, 1H); LCMS : m/e 458.0 (M+1).

[001204] Preparation of N-(3-(5-fluoro-2-(6-(2-hydroxy-2-methylpropoxy)pyridin-ylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-313 HN N lk~
H

N N N
H

[001205] 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. 'H NMR (DMSO-d6) 6 ppm: 1.16 (s, 6H), 3.93 (s, 2H), 4.57 (s, 1H), 5.74 (dd, J - 1.68 & 10.04 Hz, 1H), 6.24 (dd, J - 1.84 & 16.92 Hz, 1H), 6.45 (dd, J - 10.04 & 16.88 Hz, 1H), 6.65 (d, J - 8.88 Hz, 1H), 7.26 (t, J -8.04 Hz, 1H), 7.39 (d, J- 8 Hz, 1H), 7.48 (d, J- 7.8 Hz, 1H), 7.91 (s, 1H), 7.99 (dd, J- 2.72 &
8.92 Hz, 1H), 8.07 (d, J - 3.68 Hz, 1H), 8.27 (d, J - 2.52 Hz, 1H), 9.06 (s, 1H), 9.41 (s, 1H), 10.1 (s, 1H); LCMS
m/e 439.0 (M+1).

[0012061 Preparation of N-(3-(5-fluoro-2-(3-fluoro-4-(3-(methylsulfonyl)propoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-HN H O
F N / O~i~\
aI O
N N \ F
H

[0012071 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, 'H NMR (DMSO-d6) 6 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, 1H), 6.24 (dd, J - 1.72 &
16.96 Hz, 1H), 6.44 (dd, J - 10 & 16.84 Hz, 1H), 6.96 (t, J - 9.36 Hz, 1H), 7.28 (t, J - 8.04 Hz, 2H), 7.40 (d, J - 7.8 Hz, I H), 7.48 (d, J - 8.32 Hz, t H), 7.69 (dd, J - 2.2 & 14.4 Hz, I H), 7.91 (s, 111), 8.10 (d, J - 3.64 Hz, 1H), 9.20 (s, 1H), 9.44 (s, 1H), 10.12 (s, 111); LCMS : m/e 504.2 (M+ 1).

[001208] Preparation of 1-(3-(5-fluoro-2-(3-fluoro-4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)-4-methylpent-3-en-2-one HN
N / I O
N N "a H

[001209] 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. iH
NMR (DMSO-d6) 6 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, 1H), 6.92 (d, J = 7.44 Hz, 1H), 7.01 (t, J = 9.36 Hz, 1H), 7.27 (t, J = 7.84 Hz, 2H), 7.51 (s, 1H), 7.64-7.71 (m, 2H), 8.09 (d, J = 3.64 Hz, 1H), 9.19 (s, 1H), 9.34 (s, 1H);
LCMS : m/e 469.1 (M+1).

[001210] Preparation of N-(3-(2-(4-chloro-3-(2,3-dihydroxypropoxy)phenylamino)-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-353 HN N
F N N \

H
OH
OH

[001211] The title compound was prepared according to the schemes, steps and intermediates described below.

,/

CI x HN NHBOC ZH2 step -I NHBOC HN \ I NH2 \ -1 F t,~, / CI step-2 F t,~
CI
B IN
NI IN
1~
N CI N N OO N N O
1 3 H 0~ 4 H
IT---OH
C step-3 OH

HN \ N" v F H CI
N
NN \ I O
H

OH

A) Pd(OAc)2, BINAP, Cs2CO3, toluene, 110 C 16 h; B) TFA, CH2C12, rt, 2 h; C) acryloyl chloride, K2CO3, NMP, rt, 45 min.
[0012121 Step-1 HN \ NHBOC
F - , 1 N H 3 0[0012131 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 Na for 30 min) was heated for 16 h at 100 C
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.

[0012141 Sten_2 HN \ NH2 F / CI
N

N N O
H

OH
[0012151 To a stirred solution of 3 (300 mg, 0.5 mmol) in dry CH2C12 (6 mL) at 0 C 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), basified with NaC03 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.

[0012161 ten-3 0 ~I
HN N
F_ H j CI

H

OH
[0012171 To a stirred solution of 4 (240 mg, 0.5 mmol), in NMP (1.5 mL) at 0 C 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 C 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 C 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. 'H NMR (DMSO-d6) S ppm:
3.43-3.50 (m, 2H), 3.78-3.85 (m, 2H), 3.89-3.92 (m, 1H), 4.65 (t, J= 5.6 Hz, 1H), 4.93 (d, J= 4.8 Hz, 1H), 5.76 (dd, J- 1.92 & 10.04 Hz, 1H), 6.26 (dd, J- 1.92 & 16.92 Hz, I H), 6.46 (dd, J = 10.08 &
16.92 Hz, 1H), 7.14 (d, J= 8.72 Hz, 1H), 7.30 (t, J= 8.08 Hz, 1H), 7.39-7.43 (m, 2H), 7.46 (dd, J= 2.2 & 8.72 Hz, 1H), 7.56 (d, J= 8.04 Hz, 1H), 7.94 (s, 1H), 8.14 (d, J= 3.6 Hz, 1H), 9.24 (s, 1H), 9.48 (s, 1H), 10.13 (s, 1H); LCMS : m/e 473.8 (M+).

[0012181 Synthesis of intermediate 2 HO
O

CI CI step-1 I \\ CI B step-2 I \\ CI
02N OH A o 2N H 2N O

A) DIAD, PPh3, Et3N, dry THF, rt, 1 h; B) H2, Ra Ni, methanol, 2 h.
[0012191 Step-1 CI
J:::~O
02N 3' 0`/

Ox [0012201 To a stirred solution of 2' (0.640 g, 3.7 mmol) in THE (20 mL) 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 C 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 (3x 10 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 (Si02, 60-120, pet ether/ethyl acetate, 9/1) to give 3' (0.6 g, 60 %) as a white solid.

[0012211 Len-2 JZ~ CI

[0012221 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.

[0012231 Preparation of (S)-N-(3-(2-(4-chloro-3-(tetrahydrofuran-3-yloxy)phenylamino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-354 HN N

F, H j XtcO

[0012241 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).

[001225] 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 \ I ~ H
HN N O
H
/ 0""'. N
\I H
N N F
H

[001226] The title compound was prepared according to the schemes, steps and intermediates described below.

0..6 / I ~. N
/ H2N \ F Boc \ I 2 O-TBDMS
F HN N02 step-1 F HN \ NO2 O L) step-2 A B
I Na N, CI N'/ a F Boc CTBDMS \ I i OTBDMS
HN NH2 step -3 HN N O step 'C~ - F / 0~%`' N F_ H 04 NNN\ I F Boc I N N IF Boc H

OH OH
HN H 0 step 5 HN H N 0 F / 0~.. N 6 E F N 0' N
\ I Boc \ a H
N N F N N / F
H H

A) Pd(OAc)2, BINAP, Cs2CO3, toluene, 110 C, 6 h; B) TFA, CH2C12, rt, 1 h; C) (Boc)20, 30 min then acryloyl chloride, K2CO3, NMP, 0 C, 90 min; D) HF (49% aq.
Solution), CH3CN, rt, 2 h; E) TFA, DCM, rt, 2 h.

[0012271 Step-1 p-TBDMS
HN \ NO2 Boc N N F

[0012281 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 C 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.
[0012291 Step_2 / I OTBDMS
HN \ NH2 F / N
Boc N N

[0012301 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.
[0012311 Step-3 HN N O
H

Boc N N N F
H
[0012321 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 C 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 C 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 C, 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 (2x 10 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 (Si02, methanol/chloroform: 4/96) to give 5 (0.075 g, 71.4%) as yellow solid.
[0012331 Lt-en-4 OH
H H
ON
F N N N F
Boc H

[0012341 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 C. 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.
[0012351 Step-5 \ I ~ OH
HN N O
H
F ON
H
N N N / F
H

[0012361 To a stirred solution of 6 (0.008 g, 0.013 mmol) in CH2C12 (0.024 mL) was added TFA (0.016 mL) at 0 C. 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 mL) and the combined EtOAc extract was washed with brine (1 mL), was dried over Na2SO4 and was concentrated under reduced pressure. The crude residue was further purified by column chromatography (Si02, 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+).
[0012371 Compound 2 was prepared according to the schemes, steps and intermediates described below.

OH OH OH
step-1 ~ step-2 /~ step-3 HOOC`' N A Me000` N B Me000`' N C
H H Boc 3' / OH

0-TBDMS O-TBDMS 02 N 6' F
step-4 step-5 ' HO"w==d "d ---- McOOC N D N E

Boc Boc 4' 5' O-TBDMS O-TBDMS
step-6 ,=C~
N N
02N F Boc F H2N F Boc 7' 2 A) McOH, SOC12, reflux, 5 h; B) (Boc)20, Et3N, CH2C12, rt, 5 h; C) TBDMS-Cl, imidazole, DMF, rt, 16 h; D) LAH solution (1M in THF), -20 C, 20 min; E) DIAD, PPh3, Et3N, THF, 16 h;
F) H2, Pd/C, methanol, rt, 16 h.
[0012381 Step-1 OH
McOOC N
H

[0012391 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.
[001240] Step-2 OH
McOOC"' N
Boc 3' [001241] To a stirred solution of 2' (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 (Si02, 60-120, 100% ethyl acetate) to give 3' (3.2 g, 64%) as a white solid.
[001242] Step-3 MeOOC"' C~
N
Boc 4' [001243] 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 Na2S04. Filtration followed by concentration under reduced pressure offered a residue which was purified by column chromatography (Si02, 60-120, petroleum ether/
ethyl acetate 6/4) to give 4' (3.2 g, 80%) as a colorless liquid.
[001244] Step-4 O-TBDMS
HO"',C~
N
X
Boc 5' [001245] To a stirred solution of 4' (0.5 g, 1.39 mmol) in THE (5 mL) was added LAH
(1.39 mL, 1M solution, 1.39 mmol) at -20 C. 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.
[0012461 Step-5 O-TBDMS
O ,=N
~~` N
Boc [0012471 To a stirred solution of 5' (0.1 g, 0.3 mmol) in THE (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 C 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 (Si02, 60-120, pet ether/ethyl acetate, 9/1) to give 7' (0.120 g, 85%) as a yellow solid [0012481 Step-6 O-TBDMS
0"""'6 / I ~ N
Boc N \ F

[0012491 To a solution of 7' (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.

[001250] Preparation of tent-butyl 2-(2-(4-(4-(3-acrylamidophenylamino)-5-fluoropyrimidin-2-ylamino)-2-fluorophenoxy)ethoxy)ethylcarbamate 1-356 ~I
HN N
F L N j l 0~,O/NHBoc N N F
H

[001251] The title compound was prepared according to the schemes, steps and intermediates described below.

~C~N HBoc \ ~ H2N 2 F ~

HN NHBoc step-1 HN NHBoc NHBoc step-2 /
A \ I B
N N CI N H F

HN \INH2 HN\~NL
F 0\~ NH2 H 0,,,--., ^NHBoc F I N / I C
CIC C step-3 N H F C N \ F

A) Pd(OAc)2, BINAP, Cs2CO3, toluene, 110 C, 6 h; B) TFA, CH2Cla, RT 1 h; C) (Boc)20, 30 min then acryloyl chloride, K2CO3, NMP, 0 C, 90 min.
[001252] Step-1 i HN \ NHBoc F C\~ NHBoc N!N \ F

[001253] 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, 0.3975mmo1) in degassed toluene (toluene was purged with N2 for 30 min) was heated at 110 C 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.
[001254] Step_2 HN \ NH2 F

NN ~N /
~ F

[001255] To a stirred solution of 3 (0.047 g, 0.0762 mmol) in dry CH2C12 (3 mL) at 0 C
was added CF3COOH (1.0 mL) and the reaction mixture was stirred at 0 C 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 basified 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.
[001256] Sten-3 O
HN \ N"
H 0.'.'~O^_',N H Boc N' N \ F
H

[001257] To a stirred solution of 4 (0,028 g, 0.06731 mmol) in NMP (1.0 mL) at rt was added (Boc)20 (0.016 g, 0.07404 mmol) and the reaction mixture was stirred at rt for 30 min. It was cooled to 0 C 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 C 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 C, 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) 6 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, 1 H), 6.25 (d, J -16.76 Hz, 1 H), 6.46 (dd, J - 10.12 & 16.84 Hz, 1 H), 6.81 (s, 1 H), 6.96 (t, J - 9.12 Hz, 1 H), 7.24-7.31 (m, 2H), 7.43 (d, J- 7.96 Hz, 1H), 7.49 (d, J- 7.56 Hz, 1H), 7.68 (d, J- 14 Hz, 1H), 7.94 (s, 1H), 8.11 (d, J - 3.24 Hz, 1H), 9.21 (s, 1H), 9.46 (s, 1H), 10.15 (s, 1H); LCMS : m/e 571.1 (M+l).
[0012581 The intermediate 2 was prepared according to the schemes, steps and intermediates described below.

OH
Oz N 3' F

HO0~,~ 0- ~NH2 step-1, HO,,,-.0,- NHBoc sty A B
2' 0,,/~0 - ~N HBoc step-3 \ I 0 - ~NHBoc 4' 2 A) (Boc)20, aq. NaOH, it, 16 h; B) DIAD, PPh3, Et3N, dry THF, rt, 1 h; C) Hz, Pd/C, ethanol, rt, 16 h.

[001259] Step-1 HO,_,-,, O-,,_,NHBoc 2' [001260] To a solution of NaOH (0.76 g, 0.0 19 mmol) in water (9.6 mL) 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 THE (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 2' (3.2 g, 82%) as a viscous oil.
[001261] Step_2 O'-'--,O,-,_,NHBoc 4' [001262] To a stirred solution of 2' (0.38 g, 1.851 mmol) in THE (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 C 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 (Si02, 60-120, pet ether/ethyl acetate, 9/1) to give 4' (0.360 g, crude) as a yellow solid [001263] Step-3 O'-.'~--,O,-,~,NHBoc [001264] 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.

[001265] Preparation of N'-(2-(2-(4-(4-(3-acrylamidophenylamino)-5-fluoropyrimidin-2-ylamino)-2-fluorophenoxy)ethoxy)ethyl)-Ns-(15-oxo-18-((3 aR,4R,6aS)-2-oxohexahydro-1 H-thieno[3,4-d]imidazol-4-yl)-4,7,10-trioxa-14-azaoctadecyl)glutaramide 1-362 N I I HN NH
F HN OIN
H 0 N H H O~~O^/O~~ S H
N II
~tll N N" ' F 0 O 1-362 H

[001266] 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. 'H NMR (DMSO-d6) 6 ppm: 10.1 (s, 1H), 9.87 (s, 1H), 9.52 (s, 1H), 8.09 (d, J- 4.1 Hz, 1H), 7.86 (s, 1H), 7.78 (t, J- 5.5 Hz, 1H), 7.66 (m, 3H), 7.48 (dd, J- 2.3 & 13.8 Hz, 1H), 7.33 (m, 2H), 7.21 (t, J- 7.8 Hz, 1H), 7.11 (d, J
- 9.2 Hz, 1H), 6.90 (t, J- 9.2 Hz, 1H), 6.34 (m, 2H), 6.14 (dd, J- 2.3 & 17.0 Hz, 1H), 5.66 (dd, J- 2.3 & 17.0 Hz, 1H), 4.20 (dd, J- 5.0 & 7.3 Hz, 1H), 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+l).

[001267] Preparation of N-(3-(5-fluoro-2-(3-fluoro-4-(2-(2-methoxyethoxy)ethoxy)-phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-359 / I O
HN \ N
H
F I N 0 0.i O"

N,LN F

[001268] The title compound was prepared according to the schemes, steps and intermediates described below.

HO,_,-\0'-""0' OH step-1 I 0n0-"'0\ step-2 O--\Oi"O\

HN NH(BOC) step-3 HN NH(BOC) step-4 HN NH
F i~ C F\ N 0D F I O\/\O'~0\
N CI Ni,N F \N N F

v CI I step-5 g tl E

HN \ N"
H
\N'N C F
H

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 C, 6 h; D) TFA, CH2C12, rt, 1 h; E) (BOC)20, 30 min, then K2C03, NN P, 0 C, 15 min.
[001269] Step-1 [001270] To a stirred solution of 1 (0.5 g, 3.18 mmol) in THE (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 C 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 (Si02, 60-120, pet ether/ethyl acetate, 7/3) to give 3 (0.61 g, 65%) as a white solid.

[001271] Len-2 [001272] 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.
[001273] ten-3 HN \ NH(BOC) N !Nj~~F
H

[001274] 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 C 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 (Si02, 60-120, pet ether/ethyl acetate 5/5) to give 6 (0,350 g, 55%) as a yellow solid.
[001275] Step-4 i HN \ NH2 F / IN
N F
H

[001276] To a stirred solution of 6 (0.3 g, 0.56 mmol) in dry CH2C12 (3 mL) at 0 C was added CF3COOH (1.0 mL) and the reaction mixture was stirred at 0 C 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 mL).
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.

[0012771 Step-5 0 HN N
H

N F
H

[0012781 To a cooled solution of 7 (0.1 g, 0.23 mmol) in NMP (1.0 mL) at about 0 C 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 C 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 C, 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) 6 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, 1 H), 6.23 (d, J =
16.8 Hz, 1H), 6.45 (dd, J= 10.12 & 16.92 Hz, 1H), 6.95 (t, J= 9.4 Hz, 1H), 7.28-7.30 (m, 2H), 7.42 (d, J = 8.04 Hz, 1 H), 7.48 (d, J = 7.48 Hz, 1 H), 7.67 (d, J = 14.36 Hz, I H), 7.93 (s, l H), 8.10 (d, J= 3.44 Hz, lH), 9.20 (s, lH), 9.44 (s, lH), 10.14 (s, 1H); LCMS :
m/e 486.1 (M+l).

[001279] Preparation of (S)-N-(3-(2-(4-chloro-3-(1-hydroxypropan-2-yloxy)phenylamino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-357 a HN N
F I N H I CI
~OH

H

[001280] The title compound was prepared according to the schemes, steps and intermediates described below.
CI
I
02N \ OH

step-1 step-2 step-3 HO~,~OH A HO^,OTBDMS B 02N \ O1~1OTBDMS C HzN \ O-~' OTBDMS

CI
\
HzN OItIOTBDMS
\ 5 \ \
HN NH(BOC) step-4 HN NH(BOC) step-5 HN NHz F F CI F CI
N i D \ ~OTBDMS E i N
\ I ~OH

CI I step-6 9 /`

HN \ N
F~ IN H CI
NN OJOH
H

A) TBDMSC1, imidazole, CH2C12, 0 C, 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 C, 6 h; E) TFA, CH2C12, rt, 1 h; F) (BOC)20, 30 min, then K2CO3, NMP, 0 C, 15 min.

[001281] Step-1 HO^,OTBDMS

[001282] To stirred solution of 1 (1 g, 13.1 mmol) in DCM was added at 0 C, 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.
[001283] Step_2 CI
\ I ~OTBDMS
OZN O

[001284] To a stirred solution of 2 (1.5 g, 7.89 mmol) in THE (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 C 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 (Si02, 60-120, pet ether/ethyl acetate, 7/3) to give 4 (2.1 g, 76.9%) as a yellow oil.
[001285] Step-3 CI
\ I ~OTBDMS

[001286] 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.

[0012871 Len-4 HN \ NH(BOC) F / CI

NIN \ I Oj,,~OTBDMS
H

[0012881 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 C 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 (Si02, 60-120, pet ether/ethyl acetate 7/3) to give 7 (0.15 g, 38.4%) as a yellow solid.
[0012891 Step-5 i HN \ NH2 F L / CI
N
NN OOH

[0012901 To a stirred solution of 7 (0.15 g, 0.24 mmol) in dry CH2C12 (5 mL) at 0 C was added CF3COOH (1.5 mL) and the reaction mixture was stirred at 0 C 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 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.

[0012911 Len-6 0 \I '%
HN N
F H CI
IN
NLN \ I O1,OH
H

[0012921 A stirred solution of 8 (0.085 g, 0.21 mmol) in NMP (2.0 mL) was cooled to 0 C
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 C 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 C, 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. 'H NMR (DMSO-d6) 6 ppm: 1.18 (d, J - 6.12 Hz, 3H), 3.40-3.47 (m, 1H), 3,50-3.56 (m, 1H), 4.20-4.30 (m, 1H), 4.82 (t, J - 5.6 Hz, 1H), 5.75 (dd, J - 1.88 & 10.08 Hz, 1H), 6.25 (dd, J - 1.92 & 16.92 Hz, 1H), 6.45 (dd, J - 10.08 & 16,92 Hz, 1H), 7.12 (d, J
8.76 Hz, 1H), 7.29 (t, J - 8.08 Hz, 1H), 7.40-7.44 (m, 3H), 7.52 (d, J - 8.44 Hz, 1H), 7.91 (s, 1H), 8.12 (d, J - 3.64 Hz, 1H), 9.21 (s, 1H), 9.45 (s, 1H), 10.12 (s, 1H);
LCMS : m/e 458.0 (M+1).

[001293] Preparation of (R)-N-(3-(2-(4-chloro-3-(1-hydroxypropan-2-yloxy)phenylamino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-358 a HN N
F ,I SIN j I CI

J~i~,OH

H

[001294] The title compound was prepared according to the schemes, steps and intermediates described in Example 248 by using (R)-propane-1,2-diol in place of 1 in Step-1.
'H NMR (DMSO-d6) 6 ppm: 1.18 (d, J - 6.12 Hz, 3H), 3.40-3.47 (m, 1H), 3.50-3.56 (m, 1H), 4.20-4.30 (m, 1H), 4.82 (t, J - 5.6 Hz, 1H), 5.75 (dd, J - 1.88 & 10.08 Hz, 1H), 6.25 (dd, J -1.92 & 16.92 Hz, 1H), 6.45 (dd, J - 10.08 & 16.92 Hz, 1H), 7.12 (d, J - 8.76 Hz, 1H), 7.29 (t, J
- 8.08 Hz, 1H), 7.40-7.44 (m, 3H), 7.52 (d, J - 8.44 Hz, 1H), 7.91 (s, 1H), 8.12 (d, J - 3.64 Hz, 1H), 9.21 (s, 1H), 9.45 (s, 1H), 10.12 (s, 1H); LCMS : m/e 458.0 (M+1).

[001295] Preparation of (E)-4-(dimethylamino)-N-(3-(5-methyl-4-(m-tolylamino)pyrimidin-2-ylamino)phenyl)but-2-enamide 1-360 HN \

H H

[001296] 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. 'H NMR (DMSO-d6) 6 ppm: 7.91 (s, 1H), 7.85 (s, 1H), 7.52 (d, J- 6.4 Hz, 1H), 7.45 (d, J- 7.8 Hz, 1H), 7.34 (s, 1H), 7.31-7.26 (m, 1H), 7.21 (dd, J- 8.2, 8.0 Hz, 1 H), 7.01-6.92 (m, 4H); 6.27 (s, 1 H), 6.06 (d, J - 15.1 Hz, 1 H), 3.14 (d, J - 5.5 Hz, 2H), 2.37 (s, 3H), 2.31 (s, 6H), 2.13 (s, 3H); LCMS m/z 417 (M+l).

[001297] Preparation of N-(3-(2-(3,4-bis(2-methoxyethoxy)phenylamino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-277 ~I
HN N
F , _ N 0 N N O~~ ~
H

[001298] The title compound was prepared according to the schemes, steps and intermediates described below.

+ CI-~OMe St A OMe Step B OWN / O~~OMe Step~ HEN a':' Oi Me C(QH ~OMe HN \ N02 Step-4 F IIINI
D ~~
N CI

HN \ I H HN \ I NH2 HN I NOz F / IIN 0' We Step-6 F / IIN O'~OMe Step-5 F / IIN O~"OMe ~NN O"'OMe F NN I/ 0^~OMe E ~NN ^,OMe H H H
1.277 8 7 A) K2CO3, Cs2CO3, DMF, 90 C, 16 h; B) HN03, Acetic acid, r.t., lh;
C) Pd/C, methanol, r.t., 3 h; D) Acetic acid, ethanol, 80 C, 16 h;
E) Pd/C, methanol, r.t., 16 h; F) Acryloyl chloride, K2CO3, NMP, r.t., 30 min.
[001299] Step-1 C ~ O~~OMe / CO'-"-'OMe [001300] To a stirred solution of 1 (5 g, 45.4 mmol) in DMF (100 mL) were added K2CO3 (15.6 g, 112.9 mmol), Cs2CO3 (36.6 g, 112.3 mmol) followed by 2 (12.4 mL, 135.8 mmol) and the reaction mixture was heated to 90 C for 16 h. Then the reaction mixture was cooled, filtered and the filtrate was concentrated under reduced pressure. The residue was diluted with ethyl acetate, washed with water and brine, dried over anhydrous Na2SO4, filtered and concentrated to give 3 (9.9 g, 96%) as brown liquid.
[001301] Step_2 (( OMe 02N O,,N,~,OMe [001302] To a stirred solution of 3 (2.5 g, 11 mmol) in acetic acid (50 mL) was added fuming nitric acid (0.83 g, 13.2 mmol) at 0 C drop wise and stirred at room temperature for 1 h.
The reaction mixture was poured onto crushed ice and the solid obtained was collected by filtration and dried under vacuum to give 4 (2.23 g, 74%) as yellow solid.
[001303] Sim-3 0`~OMe H2N O,^,,~,,OMe [001304] To a solution of 4 (1 g, 3.68 mmol) in methanol (20 mL) was added Pd/C (0.2 g, 20%w/w) under N2 atmosphere. Then the reaction mixture was stirred under H2 pressure (bladder) for 3 h. It was filtered through celite bed and the filtrate was concentrated to give 5 (0.87 g, 98%) as brown oil.
[001305] Step-4 HN \ N02 F N O`,~OMe N N O i,,.,OMe H

[001306] To a solution of 6 (0.45 g, 1.675 mmol) in ethanol (4 mL) were added 5 (0.4 g, 1.658 mmol) and acetic acid (0.2 ml) and the reaction mixture was heated to 80 C for 16 h. Then it was concentrated under vacuum and the residue was purified by column chromatography (Si02, petroleum ether : ethyl acetate, 6:4) to get 7 (0.13 g, 17%) as yellow solid.

[001307] Step-5 i HN \ NH2 F / N O~*'OMe /
N N O /,,.,OMe H

[001308] To a solution of 7 (0.13 g, 0.275 mmol) in methanol (3 mL) was added Pd/C (26 mg, 20% w/w) under N2 atmosphere. Then the reaction mixture was stirred at room temperature under H2 pressure (bladder) for 16 h. It was filtered through celite bed and the filtrate was concentrated to give 8 (0.10 g, 82%) as green solid.

[001309] Step-6 0 HNaN~
H
F N O _ OMe N N Oi'OMe H

[001310] To a cooled solution of 8 (0.1 g, 0.225 mmol) in NMP (1.0 mL) was added K2CO3 (0.15 g, 1.1 mmol), acryloyl chloride (0.002 g, 0.25 mmol) and the reaction mixture was stirred at 0 C for 30 min. Then the reaction mixture added drop-wise to a cold, stirring solution of 10%
NaHCO3 (-5 ml) and stirred at 0 C for 30 min. A solid precipitated out was isolated by filtration through a Buchner funnel. The solid was dissolved in methanol: dichloromethane (1:1, 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. The combined ethyl acetate extract was washed with water and brine, dried over Na2SO4 and concentrated under reduced pressure to get 1-277 (0.049 g, 44%) as yellow solid.
iH NMR (DMSO-d6): 6 = 3.28 (s, 3H), 3.29 (s, 3H), 3.56-3.60 (m, 4H), 3.88 (t, J = 4.8 Hz, 2H), 3.98 (t, J - 4.9 Hz, 2H), 5.74 (d, J - 12.1 Hz, 1H), 6.24 (d, J - 14 Hz, 114), 6.45 (dd, J - 16, Hz, 1H), 6.75 (d, J= 8.7 Hz, 1H), 7.21-727 (m, 3H), 7.41 (d, J= 8 Hz, 1H), 7.52 (d, J= 8 Hz, 1H), 7.89 (s, 1H), 8.07 ( d, J= 3.7 Hz, 1H), 8.95 (s, 1H), 9.38 (s, 1H), 10.12 (s, 1H); LCMS:
m/e: 498.2 (M+1).

[0013111 Preparation of tent-butyl 2-(2-(4-(4-(3-acrylamidophenylamino)-5-fluoropyrimidin-2-ylamino)phenoxy)ethoxy)ethylcarbamate 1-370 HN /
H H
F - _ / N XO~
/ I
N~N 0 H

[0013121 The title compound was prepared according to the schemes, steps and intermediates described in Example 245, by using tert-butyl 2-(2-(4-aminophenoxy)ethoxy)ethylcarbamate in the place of 2 in Step 1. 'H NMR (DMSO-d6): 6 -1.37 (s, 9H), 3.09 (m, 2H), 3.43 (t, J- 6.0 Hz, 2H), 3.68 (d, J- 4.3 Hz, 2H), 3.98 (t, J- 4.3 Hz, 2H), 5.75 (d, J- 11.4 Hz, 1H), 6.25 (d, J- 15.6 Hz, 1H), 6.45 (dd, J- 16.9, 10 Hz, 111), 6.73-6.81 (m, 3H), 7.27 (t, J- 8.1 Hz, 1H), 7.40 (d, J- 7.9 Hz, 1H), 7.47-7.53 (m, 3H), 7.92 (s, 1H), 8.06 (d, J- 3.6 Hz, 1H), 8.96 (s, 1H), 9.36 (s, 1H), 10.12 (s, 1H); LCMS: m/e:
553.2 (M+1).

[0013131 Preparation of N1-(2-(2-(4-(4-(3-acrylamidophenylamino)-5-fluoropyrimidin-2-ylamino)phenoxy)ethoxy)ethyl)-N5-(15-oxo-19-((3aS,4S,6aR)-2-oxohexahydro-lH-thieno[3,4-d]imidazol-4-yl)-4,7, 10-trioxa-14-azanonadecyl)glutaramide 1-363 O
HN \ H
\N 0~\0^~NH 0 ON
~
N~V ~0 HN
H

NH

[0013141 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)phenoxy)ethoxy)ethylcarbamate (1-370, described in Example 252) in the place of 1-45 in Step 1. LC/MS (RT - 2.045/
(M + H)) 995.4.

[001315] Preparation of (R)-N-(3-(5-fluoro-2-(3-fluoro-4-(1-hydroxypropan-2-yloxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-372 HN H

F N I O-COH
N N F
H

[001316] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using (R)-2-(4-amino-2-fluorophenoxy)propan-l-ol in the place of 4 in Step 2. 'HNMR (DMSO-d6): 8 = 1.17 (d, J = 6.2 Hz, 3H), 3.40-3.46 (m, 1H), 3.50-3.56 (m, 1H), 4.22 (q, J= 5.9 Hz, lH), 4.83 (t, J= 5.8 Hz, lH), 5.75 (dd, J= 10.1, 1.9 Hz, 1H), 6.25 (dd, J=16.9, 1.9 Hz, 1H), 6.46 (dd, J= 16.9, 10.1 Hz, 1H), 6.98 (t, J= 9.4 Hz, 1H), 7.24-7.31 (m, 2H), 7.42 (d, J= 8.2 Hz, 1H), 7.49 (d, J= 7.9 Hz, 1H), 7.68 (dd, J= 14.3, 2.4 Hz, 1H), 7.93 (s, 1H), 8.11 (d, J= 3.7 Hz, 1H), 9.20 ( s, 1H), 9.45 (s, 1H), 10.14 (s, 1H); LCMS
m/e 442.2 (M+1).

[001317] Preparation of (S)-N-(3-(5-fluoro-2-(3-fluoro-4-(1-hydroxypropan-2-yloxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide 1-373 O
N
HN H

F /r ' OH
NNN F
H

[001318] The title compound was prepared according to the schemes, steps and intermediates described in Example 20, by using (S)-2-(4-amino-2-fluorophenoxy)propan-l-ol in the place of 4 in Step 2. LC/MS (RT - 3.316/(M + H)) 442Ø

[001319] Preparation of (S)-((R)-2-(4-(4-(3-acrylamidophenylamino)-5-fluoropyrimidin-2-ylamino)-2-fluorophenoxy)propyl) 2-amino-3-methylbutanoate 1-374 N

F 0~0, NHZ
N N N / I F

H

[001320] The title compound was prepared according to the schemes, steps and intermediates described below.
/ o /I 00 HN N" 'eO HN \ N" v~ 0 H
H H Step-1 H
F O N F O N Step-2 IN ~OH + HO boc I IN / I ~O boc N H F N H F

O
HN \ N" 0 F H O NH
\N / r0 2 NN \ F
H

A) EDC.HC1, HOBT, DMAP, NMM, DMF, r.t., 16 h;
B) HC1(1M in Diethylether), 0 C, 1 h.

[001321] Step-1 HN H N

N I 00 boc N N F
H

[001322] To a stirred solution of 1-372 (0.08 g, 0.18 mmol) in DMF (0.8 mL) were added 2 (0.057 g, 0.26 mmol), EDC.HC1 (0.086 g, 0.45 mmol), HOBT (0.06 g, 0.44 mmol), (0.008) and NMM (0.036 g, 0.36 mmol). And the reaction mixture was stirred at room temperature for 16 h. Then it was quenched with water and extracted with ethyl acetate. The ethyl acetate layer was washed with water and brine, dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by column chromatography (Si02, chloroform:methanol, 9:1) to obtain 3 (0.06 g, 51.7%) as off white solid.
[0013231 Step-2 5111, 0 aZz~~' F H OroNH 2 \ I

N IN F
H

[0013241 A cold solution of HC1 in diethyl ether (1M, 6 ml) was added to 3 (60 mg, 0.09 mmol) and the reaction mixture was stirred at 0 C for 45 min. Then the reaction mixture was concentrated and the solid obtained was washed with diethyl ether and dried to yield 1-374 (as its HC1 salt) as yellow solid (20 mg). 1H NMR (DMSO-d6) 6 = 0.77 (d, J = 6.8 Hz, 3H), 0.83 (d, J
= 6.8 Hz, 3H), 1.23 (d, J = 6.3 Hz, 3H), 1.75-1.83 (m, 1H), 3.10 (d, J = 5.2 Hz, 1H), 4.05-4.11 (m, I H), 4.22 (dd, J- 11.7, 3.3 Hz, 1H), 4.40-4.50 (m, 1H), 5.73 (d, J- 10.2 Hz, 1H), 6.23 (d, J
17.1 Hz, 111), 6.44 (dd, J- 17, 10.2 Hz, 1H), 6.98 (t, J- 9.2 Hz, I H), 7.24-7.29 (m, 2H), 7.41 (d, J= 7.8 Hz, 1H), 7.48 (d, J= 7.8 Hz, 1H), 7.68 (d, J= 14.3 Hz, 1H), 7.91 (s, 1H), 8.10 (d, J-3.5 Hz, 1H), 9.23 (s, 1H), 9.44 (s, 1H), 10.13 (s, 1H). LCMS : m/e 541.2 (M+l).

[0013251 Preparation of (S)-((S)-2-(4-(4-(3-acrylamidophenylamino)-5-fluoropyrimidin-2-ylamino)-2-fluorophenoxy)propyl) 2-amino-3-methylbutanoate 1-377 a HN H 0 F N / I 0~~0 NH2 N N F
H

[001326] The title compound was prepared according to the schemes, steps and intermediates described in Example 256, by using (S)-N-(3-(5-fluoro-2-(3-fluoro-4-(l-hydroxypropan-2-yloxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide (1-373 described in example 255) in the place of 1-372 in Step 1. LC/MS (RT = 3.227/(M + H)) 542.2.

[001327] Preparation of (S)-((S)-2-(5-(4-(3-acrylamidophenylamino)-5-fluoropyrimidin-2-ylamino)-2-chlorophenoxy)propyl) 2-amino-3-methylbutanoate 1-369 a HN H
F / CI

NNH 0 ~NH2 [001328] The title compound was prepared according to the schemes, steps and intermediates described in Example 256, by using (S)-N-(3-(2-(4-chloro-3-(1-hydroxypropan-2-yloxy)phenylamino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide (1-357 described in example 248) in the place of 1-372 in Step 1. LC/MS (RT = 3.54/(M + H)) 557.2.

[001329] Preparation of N-(3-(2-(4-(2-(2-azidoethoxy)ethoxy)phenylamino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-371 HN I N-F N a'~' 01 rN3 '~' '15 N N 0 J
H

[001330] The title compound was prepared according to the schemes, steps and intermediates described below.

N0 F + HOlO /OH St NOZ \ O` O /OH St B Z HZN \ I O1O fOH

1 2 3` J 4 N I / Nbo, Step-3 HN I / Hboc Step-4 HN / Hboc Step-5 4 + F F 0 CH F \ 0 OMs E

NC1 C NN `O~ D NN \O~
H H

~ ~ ~ IOxI
HN I / Nb0c Ste p-6 HN / NH2 Ste 7 HN / N
F N J 'õ \ 010; N3 N N N \ I 0 \ OJ /Na F I N i N \ 0 0 /N3 N I` . I`J
H H H

A) K2CO3, DMSO, r.t., 16 h; B) Pd/C, Ethanol, 16 h; C) Pd(OAc)2, BINAP, Cs2CO3, Toluene, 110 C, 16 h; D) Mesyl chloride, Et3N, CHzClz, r.t., 1 h; E) Sodium azide, DMF, 70 C, 4 h; F) TFA, CHzClz, 0 C-r.t., 1.5 h; G) Acryloyl chloride, NMP, K2CO3, 30 min.

[0013311 Step-1 [0013321 To a stirred solution of 1 (1 g, 7.09 mmol) in DMSO (20 mL) was added 0.96 g, 6.95 mmol) and stirred at room temperature for 15 min. Then 2 (3.7 g, 34.86 mmol) was added and stirring continued at room temperature for 16 h. The reaction mixture was poured into ice water and extracted with ethyl acetate. The organic layer was washed with water and brine, dried over anhydrous Na2SO4, filtered and concentrated to obtain 3 as yellow solid (1.0 g, 62%).
[0013331 Step_2 Jr [0013341 To a solution of 3 (1.0 g, 4.4 mmol) in ethanol was added Pd/C (0.1 g, 10% w/w) under N2 and then the reaction mixture was stirred under H2 pressure (bladder) for 16 h. Then the reaction mixture was filtered and the filtrate was concentrated to obtain 4 as brown viscous liquid (0.8 g, 92.2%).
[001335] Step-3 H N I N,boc N N f 0 f H

[001336] A mixture of 4 (0.3 g, 1.52 mmol), 5 (0.61 g, 1.8 mmol), Pd(OAc)2 (0.017 g, 0.076 mmol), BINAP (0.066 g, 0.106 mmol) and Cs2CO3 (1.23 g, 3.78 mmol) in degassed toluene (20 mL) (toluene was purged with N2 for 30 min) was heated at 110 C
for 16 h under N2 atmosphere. The reaction mixture was cooled, diluted with EtOAc, washed with water and brine and dried over Na2SO4. Filtration followed by concentration under reduced pressure offered a residue which was further purified by column chromatography (Si02, petroleum ether : ethyl acetate; 6:4) to obtain 6 (0.3 g, 39.5%) as a brown solid.
[001337] Step-4 HN Nboc F N H 0 OMs N N f O f H

[001338] To a stirred solution of 6 (0.35 g, 0.7 mmol) and triethylamine (0.2 mL, 1.4 mmol) in dichloromethane (10 mL) at 0 C was added mesyl chloride (0.12 g, 1.05 mmol) and then the reaction mixture stirred at room temperature for 1 h. It was quenched with water; the organic layer was separated, dried over anhydrous Na2SO4 and concentrated to obtain 7 as colorless oil (0.4 g, crude).

[0013391 Len-5 HN N.boc F N j 0 N3 Ni N N ~f O f H

[0013401 To a stirred solution of 7 (0.4 g, 0.69 mmol) in DMF (5 mL) was added sodium azide (0.054 g, 0.83 mmol) and the reaction mixture was heated to 70 C for 4 h. Then it was treated with ice water and extracted with ethyl acetate. The ethyl acetate layer was washed with water and with brine, dried over anhydrous Na2SO4, filtered and concentrated to obtain 8 as viscous oil 8 (0.3 g, 82.6%).
[0013411 Step-6 N N ; N 1 0 f H

[0013421 To a stirred solution of 3 (0.3 g, 0.57 mmol) in dry CH2C12 (10 mL) at 0 C was added CF3COOH (1.0 mL) and the reaction mixture was stirred at 0 C for 30 min and then at r.t.
for 1 h. It was concentrated under reduced pressure and the residue was treated with NaHCO3 solution and extracted with ethyl acetate. The combined EtOAc extract was washed with water.
Then it was extracted with 10% citric acid solution. The combined citric acid extract was basified with 10% NaOH solution and then extracted with EtOAc. The EtOAc extract was washed with water and brine, dried over Na2SO4 and concentrated to get 9 (0.17 g, 70%) as brown solid.

[0013431 Sten_7 HN I N"

F N ja 0 N3 N N O
H

[0013441 To a stirred solution of 9 (0.075 g, 0.177 mmol) in NMP (1.5 mL) at 0 C was added K2C03 (0.05 g, 0.36 mmol) followed by acryloyl chloride (0.0 17 g, 0.188 mmol) and the reaction mixture was stirred at 0 C for 30 min. Then 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 C and the solid separated was isolated by filtration through a Buchner funnel. The solid was washed with cold water, hexane and was dissolved in methanol:dichloromethane (1:1, 5 mL) and then concentrated under reduced pressure. The residue obtained was suspended in cold water (3 mL) and EtN (-1 ml), stirred for a while and extracted with ethyl acetate. The combined ethyl acetate extract was washed with water and brine, dried over Na2SO4 and concentrated under reduced pressure. The residue obtained was purified by column chromatography twice (Si02, petroleum ether:ethyl acetate, 1:1) to get 1-371 (0.04g,47.3%) as white solid.'H NMR (DMSO-d6): 6 = 3.42 (t, J= 4.9 Hz, 1H), 3.66 (t, J= 4.9 Hz, 1H), 3.72-3.77 (m, 4H), 4.00 (t, J= 4.6 Hz, 2H), 5.75 (dd, J= 10.1, 1.8 Hz, 1H), 6.25 (dd, J
17, 1.9 Hz, 1 H), 6.45 (dd, J = 16.9, 10.1 Hz, 1 H), 6.75 (d, J = 9 Hz, 2H), 7.27 (t, J = 8.1 Hz, 1H), 7.40 (d, J= 8 Hz, 1H), 7.47-7.53 in, 3H), 7.92 (s, 1H), 8.06 (d, J= 3.7 Hz, 1H), 8.97 (s, 1H), 9.37 (s, 1H), 10.12 (s, 1H); LCMS : m/e 479.2 (M+1).

[0013451 Preparation of N-(3-(2-(4-(2-azidoethoxy)phenylamino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-376 o HN i N
H
I C Ns '-I
NN
H

[001346] The title compound was prepared according to the schemes, steps and intermediates described in Example 259, by using 2-(4-aminophenoxy) ethanol in the place of 4 in Step 3. LC/MS (RT - 3.447/(M + H)) 435.2.

[001347] Preparation of N-(3-(2-(4-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)phenylamino)-5-fluoropyrimidin-4-ylamino)phenyl)acrylamide 1-379 HN C H

N N
H

[001348] The title compound was prepared according to the schemes, steps and intermediates described in Example 259, by using 2,2'-(ethane-1,2-diylbis(oxy))diethanol in the place of 2 in Step 1. LC/MS (RT - 4.4541(M + H))475.6.

[001349] Preparation of (E)-N-(3-(2-(4-chloro-3-(2-methoxyethoxy)phenylamino)-fluoropyrimidin-4-ylamino)phenyl)but-2-enamide 1-375 ~ I
HN H
F L N

NIN
H

[001350] The title compound was prepared according to the schemes, steps and intermediates described in Example 238, by using (E)-but-2-enoyl chloride in the place of 7 in Step 4. LC/MS (RT - 4.026/(M + H)) 473.2.

[001351] Preparation of N-(3-(5-fluoro-4-(4-(2-methoxyethoxy)phenylamino)pyrimidin-2-ylamino)phenyl)acrylamide 1-381 ~aNH HN
F
NIN \ I
H

[001352] The title compound was prepared according to the schemes, steps and intermediates described in Example 4, by using 4-(2-methoxyethoxy)aniline in the place of 2 in Step 1. LC/MS (RT - 2.275/(M + H)) 424.1.

[001353] Preparation of N-(3-(5-fluoro-4-(3-(2-morpholinoethoxy)phenylamino)pyrimidin-2-ylamino)phenyl)acrylamide 1-382 N' O \ NH HN" v F

N N
H

[001354] The title compound was prepared according to the schemes, steps and intermediates described in Example 4, by using 3-(2-morpholinoethoxy)aniline in the place of 2 in Step 1. LC/MS (RT - 2.108/(M + H)) 479.2.

[001355] Preparation of (S)-N-(3-(5-fluoro-4-(4-(tetrahydrofuran-3-yloxy)phenylamino)pyrimidin-2-ylamino)phenyl)acrylamide 1-338 O
~o / o NH HN
F ~N
N'N I
H

[001356] The title compound was prepared according to the schemes, steps and intermediates described in Example 4, by using (S)-4-(tetrahydrofuran-3-yloxy)aniline in the place of 2 in Step 1. LC/MS (RT - 1.71/(M + H)) 458.1.

Biological Examples [001357] Described below are assays used to measure the biological activity of provided compounds as inhibitors of BTK, TEC, ITK, BMX, ErbB 1 (EGFR), ErbB2, ErbB4, and JAK3.

Omnia Assay Protocol for Potency Assessment Against BTK
[001358] Below describes the protocol using EGFR-WT and EGFR-T790M/L858R and the protocol BTK-optimized reagent conditions then follow.
[001359] 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.cfin?pageid-l 1338 or www.invitrogen.com/site/uslen/home/Products-and-Services/Applications/Drug-Discovery/Target-and-Lead-Identification-and-Validation/KinaseBiology/KB-Misc/Biochemical-Assays/Omnia-Kinase-Assays.html.
[001360] Briefly, lOX stocks of EGFR-WT (PV3872) from Invitrogen and EGFR-T790M/L858R (40350) from BPS Bioscience, San Diego, CA, 1.13X ATP (ASOO1A) and appropriate Tyr-Sox conjugated peptide substrates (KCZ1001) were prepared in 1X kinase reaction buffer consisting of 20 mM Tris, pH 7.5, 5 mM MgC12, 1 mM EGTA, 5 mM
(3-glycerophosphate, 5% glycerol (lOX stock, KBO02A) and 0.2 mM DTT (DSOO1A). 5 L of each enzyme were pre-incubated in a Coming (#3574) 384-well, white, non-binding surface microtiter plate (Corning, NY) for 30 min. at 27 C with a 0.5 L volume of 50%
DMSO and serially diluted compounds prepared in 50% DMSO. Kinase reactions were started with the addition of 45 L of the ATP/Tyr-Sox peptide substrate mix and monitored every 30-90 seconds for 60 minutes at &ex360/&em485 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).
[0013611 The modified BTK-optimized reagent conditions for the above protocol are:
[BTK] - 5 nM, [ATP] - 40 mM, [Y5-Sox] - 10 mM (ATP KMapp - 36 mM).

[0013621 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 ICso <10 nM;
compounds having an activity designated as "B" provided an ICso 10-100 nM; compounds having an activity designated as "C" provided an ICs0 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.
Table 6. BTK Inhibition Data Compound # BTK Inhibition Compound # BTK Inhibition Compound # BTK Inhibition Compound # BTK Inhibition Compound # BTK Inhibition Compound # BTK Inhibition Compound # BTK Inhibition BTK Ramos Cellular Assay [0013631 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 M. Ramos cells were incubated with compound for 1 hour, washed again and resuspended in 100u1 serum-free media.
Cells were then stimulated with 1 g 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 g total protein from lysates were loaded on gel and blots were probed for phosphorylation of the BTK substrate PLC72. Dose response inhibition of BTK signaling in Ramos cells is depicted in Figures 1, 2, 3, 4 and 5.
[001364] 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 >l0,000 nM.
Table 7. BTK Ramos Cellular Inhibition Data Compound # BTK Inhibition Compound # BTK Inhibition Compound # BTK Inhibition Washout Experiment with Ramos cells [001365] Ramos cells were serum starved for one hour in RPMI media +l%
glutamine at 37 C. After starvation, Ramos cells were treated with 100nM 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 FNNOO11) supplemented with Roche complete protease inhibitor tablets (Roche 11697498001) and phosphatase inhibitors (Roche 04 906 837 001) and l8ug total protein lysate was loaded in each lane. Inhibition of BTK kinase activity was assayed by measuring its substrate (PLCy2) 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.
[001366] Table 8 provides data for selected compounds in the Ramos washout assay.
Table 8. BTK Washout Data Compound # BTK Inhibition T e 1-2 irreversible 1-4 irreversible 1-7 irreversible 1-28 irreversible 1-35 irreversible 1-38 reversible 1-228 irreversible I-230 irreversible 1-242 irreversible 1-243 irreversible 1-247 irreversible 1-248 irreversible Mass Spectrometry for BTK
[001367] Intact BTK was incubated for 1 hr at a l OX fold excess of 1-7 to protein. Aliquots (2 l) of the samples were diluted with 10 l 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.

Human primary B cell proliferation assay [0013681 Human naive B cells were purified from 100 mL whole blood using a MACS
purification kit designed to isolate CD19+, IgD+ cells by negative selection.
Purified naive B
cells were resuspended in RPMI complete and stimulated with 5 g/ml a-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 a-IgM stimulation. Importantly, a molecule with the same scaffold as 1-7 but biochemically inactive against BTK, IR-7, is not active in the naive B cell proliferation assay.

Table 9.

Compound # EC50 (nM) IR-7 >1000 B cell lymphoma proliferation assay [0013691 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 M;
compounds having an activity designated as "B" provided an EC50 0,1-1 M; compounds having an activity designated as "C" provided an EC50 of 1-10 M; and compounds having an activity designated as "D" provided an EC50 of >10 M.

Table 10.

Compound # EC50 (M) Compound # EC50 M

Compound # EC50 M

In vivo Thymus-Independent (TI-2) B cell Activation [0013701 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 g TNP-Ficoll on day 1, serum was collected on day 6 and analyzed for circulating a-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.
Table 11.

Compound # % Inhibition I M (1:1600) IgG3 (1:100) 1-96 43 37.2 Collagen Antibody Induced Arthritis Model [0013711 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 g LPS in 200 l 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.

Table 12.

Compound # Dose % inhibition foot pad swellin 1-7 30 mg/kg 83 PG-PS Arthritis Model [0013721 On day 0, female Lewis rats received an intraperitoneal (IP) bolus of peptidoglycan-polysaccharide (PG-PS) in an amount of 15 g/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).
Table 13.

Compound # n % inhibition ankle swelling 1-7 1 77.5 1-96 2 82.8 Mass Spectrometry for TEC Kinase (Compound 1-2) [0013731 TEC kinase (45 pmols; Invitrogen) was incubated with (1-2) (450 pmols) for 3 hrs at 10X excess prior to tryptic digestion. lodoacetamide 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 C18 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).
[0013741 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 lodacetamide 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.
[001375] 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).
[001376] 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:
[001377] 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 P 14R fragmentation calibration for the Curved field Reflectron.

Mass Spectrometry for TEC Kinase (Compound 1-4) [001378] TEC kinase (45 pmols; Invitrogen) was incubated with (1-4) (450 pmols) for 3hrs at lOX excess prior to tryptic digestion. lodoacetamide 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 C18 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).
[001379] 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 lodacetamide 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.
[001380] 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).
[001381] 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 I-4.
Instrumental: .
[001382] 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 P 14R fragmentation calibration for the Curved field Reflectron.

Mass Spectrometry for TEC Kinase (Compound 1-7) [001383] TEC kinase (45 pmols; Invitrogen) was incubated with (1-7) (450 pmols) for 3hrs at lOX excess prior to tryptic digestion. lodoacetamide 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 C18 Zip Tipping directly onto the MALDI target using alpha cyano-4-hydroxy cinnamic acid as the matrix (5mg/ml in 0.l%TFA:Acetonitrile 50:50).
[001384] 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 lodacetamide 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 (TIDELVECEETFGR).
[001385] 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).
[001386] 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:
[001387] 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 P 14R fragmentation calibration for the Curved field Reflectron.

Omnia Assay Protocol for Potency Assessment Against Active Forms of ITK Kinase [001388] 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 M, [Y6-Sox] - 10 M (ATP KMapp - 33 LM).

[001389] 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 ICso 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.
Table 14. ITK Inhibition Data Compound # ITK Inhibition Compound # ITK Inhibition Compound # ITK Inhibition Compound # ITK Inhibition Omnia Assay Protocol for Potency Assessment Against Active Forms of BMX Kinase [0013901 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 M, [Y5-Sox] - 7.5 M (ATP KMapp - 107 M).

[0013911 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.
Table 15. BMX Inhibition Data Compound # BMX Inhibition Compound # BMX Inhibition Cloning, Expression and Purification of EGFR-WT and EGFR C797S Mutant Using Baculovirus and Insect Cells (i) Subcloninj of EGFR- WT and mutant kinase domains [0013921 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.

(ii) Expression [0013931 P1 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 SF9001 SFM (Invitrogen cat # 10902-088), supplemented with lOmg/L gentamicin (Invitrogen, Carlsbad, CA, cat# 15710-064)) using a viral load of 0.lml of virus per 100 ml of cell suspension. Expression was optimized using Blue Sky Biotech's Infection Kinetics Monitoring system (Worcester, MA).

(iii) Purification [001394] Infected insect cells were pelleted. Cell pellets were resuspended in Blue Sky Biotech's lysis buffer (Worcester, MA, 1X 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 l 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, lX WX, 25mM imidazole ).The protein was eluted with lX 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.

Mass Spectrometry for EGFR
[001395] EGFR wild type and EGFR (mutant C797S) is incubated with 10-fold excess of test compound for 1 hr and 3 hrs. 1 l 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.

Omnia Assay Protocol for Potency Assessment Against EGFR (WT) and EGFR
(T790M/L858R) Active Enzymes [0013961 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-modified optimized reagent conditions are:
[0013971 [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).

[0013981 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 ICso 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 ICso >10,000 nM.
Table 16. EGFR Wild Type Inhibition Data Compound EGFR

Compound # EGFR

Compound # EGFR

Compound # EGFR

Compound # EGFR

Compound # EGFR

Compound # EGFR

Table 17. EGFR mutant (T790M/L858R and T790M) Inhibition Data Compound # EGFR (T790M/L858R) EGFR (T790M) Compound # EGFR (T790M/L858R) EGFR (T790M) Cellular Assays for EGFR Activity [0013991 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 Vol 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 M 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.
[0014001 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 0.001 M 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.

Washout Experiment for EGFR Activity [0014011 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 M 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.

Washout Experiment in HCC827 cells containing EGFR deletion mutantHCC827 [0014021 Cells (ATCC, Manassas,VA) were plated in Growth Media (RPMI 1640) supplemented with 10% FBS,10uM HEPES, 2mM 1-glutamine, 1mM NaPyruvate and pen/strep (Invitrogen, Carlsbad, CA) at a density of 2.5 x 10 5 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).
[0014031 The following morning the media was removed and 2 ml fresh Basal Media containing luM 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.
[0014041 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
(Tyr1068) (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.

Mass Spectrometry for ERBB4 [0014051 Erbb4 kinase domain (Upstate) was incubated with compound for 60 minutes at 10-fold excess of compound 1-4 and I-11 to protein. 1 l aliquots of the samples (total volume of 4.24u1) were diluted with 10 l 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).
[0014061 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 I-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 I-11.

ErbB1, ErbB2 and/or ErbB4 Kinase Inhibition [0014071 Compounds of the present invention were assayed as inhibitors of one or more of ErbB 1, 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/downloads/Z-LYTE_Brochure_1205.pdf) using the Z'-LYTETM
biochemical assay procedure or similar biochemical assay. The Z'-LYTETM
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 ERBB1 with an IC50 of 2,233 nM. Using this assay, Compound 1-56 was found to inhibit ERBB4 (HER4) with an ICso of 2,165 nM.

Mass Spectrometry for Janus-3 Kinase (JAK3) [0014081 JAK3 kinase (33 pmols; Invitrogen) was incubated with (1-7) (327 pmols) for 3hrs at lOX 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 C18 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).

[001409] 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.
[001410] 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).
[001411] 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 I-11 was also tested using the same procedure and showed measurable modification.
Instrumental:
[001412] 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 P 14R fragmentation calibration for the Curved field Reflectron.

Omnia Assay Protocol for Potency Assessment Against the Active Form of JAK3:
[001413] 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 JAK3-optimized reagent conditions were:

[JAK3] - 5 nM, [ATP] - 5 M, [Y12-Sox] - 5 M (ATP KMapp -5 M).

[001414] 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 ICso 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.
Table 18. JAK3 Inhibition Data Compound # JAK3 Inhibition Compound # JAK3 Inhibition JAK3 Cellular Assay Protocol in CTLL2 Cells [0014151 Compounds 1-2, 1-4 and 1-7 were tested in the following protocol.
CTLL2: murine lymphoma cell line ATCC: TIB-214. 5x106 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 I OOnM 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.

BTK occupancy in Ramos cells with 1-7 and 1-215 using streptavidin beads [0014161 Ramos cells were incubated with 0.1, 0.05, 0.01, or 0.001 pM 1-7 in serum free media for 1 hour at 37 T. 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 M 1-215 for 1 hour at room temperature, then incubated with streptavidin-coupled agarose beads (ThermoFisher) overnight at 4 T. The beads were washed three times with lysis buffer and the bound proteins were boiled off the beads at 95 C 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 I-215.

Washout Experiment with 1-7 and probe compound 1-215 [001417] Ramos cells were incubated with 0.1 M 1-7 or a reversible BTK
inhibitor control compound in serum free media for 1 hour at 37 C. 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 M 1-215 for 1 hour at room temperature, then overnight at 4 C 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.

Measuring BTK occupancy from in vitro samples by ELISA
[001418] In order to determine the amount of free B'TK 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).
[001419] 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 M 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.
[0014201 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.
Table 19.

Compound # >50% occupancy >90% occupancy Human primary B cell covalent probe occupancy in vitro [0014211 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 T. After incubation, the cells were pelleted, washed with 1X 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 I-215. 1-96 and 1-182 occupied at least 50% of BTK at concentrations greater than 10 nM.

Dog primary B cell covalent probe occupancy in vitro [0014221 Canine whole blood (30 mL) was diluted to 50 mL total with 1X 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 mL RBC lysis buffer (Boston Bioproducts) and the remaining PBMCs were washed 3 times in 1X PBS at 250 x g. PBMCs were treated with compound at a 1:1000 dilution for one hour at 37 C, 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.

Measuring BTK occupancy from in vivo samples by ELISA
[0014231 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 l. 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.
Table 20.

Treatment % BTK Occupancy 2 h % BTK Occupancy 24 h Vehicle 0 0 Proteomics analysis [0014241 Proteins that are covalently bound to 1-215 in a cell lysate are identified using mass spectrometry. Cell lysate is incubated with 1 M 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.

[0014251 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

1. A compound selected from the group consisting of:

or a pharmaceutically acceptable salt thereof.
CA2783624A 2009-12-29 2010-12-29 Heteroaryl compounds and uses thereof Active CA2783624C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US12/648,693 2009-12-29
US12/648,693 US8338439B2 (en) 2008-06-27 2009-12-29 2,4-disubstituted pyrimidines useful as kinase inhibitors
PCT/US2010/062432 WO2011090760A1 (en) 2009-12-29 2010-12-29 Heteroaryl compounds and uses thereof

Publications (2)

Publication Number Publication Date
CA2783624A1 true CA2783624A1 (en) 2011-07-28
CA2783624C CA2783624C (en) 2019-08-20

Family

ID=44307129

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2783624A Active CA2783624C (en) 2009-12-29 2010-12-29 Heteroaryl compounds and uses thereof

Country Status (14)

Country Link
US (5) US8338439B2 (en)
EP (1) EP2519235B1 (en)
JP (2) JP5879270B2 (en)
KR (2) KR101747159B1 (en)
CN (2) CN102740847B (en)
AU (2) AU2010343055B2 (en)
CA (1) CA2783624C (en)
IL (2) IL220155B (en)
MX (2) MX2012007507A (en)
NZ (2) NZ600631A (en)
RU (1) RU2636584C2 (en)
SG (1) SG181962A1 (en)
TW (1) TWI592405B (en)
WO (1) WO2011090760A1 (en)

Families Citing this family (280)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7982036B2 (en) 2007-10-19 2011-07-19 Avila Therapeutics, Inc. 4,6-disubstitued pyrimidines useful as kinase inhibitors
US7989465B2 (en) * 2007-10-19 2011-08-02 Avila Therapeutics, Inc. 4,6-disubstituted pyrimidines useful as kinase inhibitors
MX2010012703A (en) 2008-05-21 2010-12-21 Ariad Pharma Inc Phosphorous derivatives as kinase inhibitors.
US9273077B2 (en) 2008-05-21 2016-03-01 Ariad Pharmaceuticals, Inc. Phosphorus derivatives as kinase inhibitors
US8338439B2 (en) 2008-06-27 2012-12-25 Celgene Avilomics Research, Inc. 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
CA2727455C (en) 2008-06-27 2019-02-12 Avila Therapeutics, Inc. Heteroaryl compounds and uses thereof
EP2440559B1 (en) 2009-05-05 2018-01-10 Dana-Farber Cancer Institute, Inc. Egfr inhibitors and methods of treating disorders
AR076550A1 (en) * 2009-05-06 2011-06-22 Portola Pharm Inc INHIBITORS OF THE JANUS TIROSINA KINASA (JAK)
US8586751B2 (en) 2009-06-12 2013-11-19 Bristol-Myers Squibb Company Nicotinamide compounds useful as kinase modulators
RU2012114902A (en) 2009-09-16 2013-10-27 Авила Терапьютикс, Инк. CONJUGATES AND PROTEINKINASE INHIBITORS
US8466155B2 (en) * 2009-10-02 2013-06-18 Boehringer Ingelheim International Gmbh Pyrimidines
WO2011082285A1 (en) 2009-12-30 2011-07-07 Avila Therapeutics, Inc. Ligand-directed covalent modification of protein
EP2571361A4 (en) 2010-05-19 2013-11-13 Univ North Carolina Pyrazolopyrimidine compounds for the treatment of cancer
CA2798578C (en) 2010-05-21 2015-12-29 Chemilia Ab Novel pyrimidine derivatives
US8354420B2 (en) 2010-06-04 2013-01-15 Genentech, Inc. Aminopyrimidine derivatives as LRRK2 inhibitors
MX336875B (en) 2010-08-10 2016-02-04 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
ES2776002T3 (en) 2010-11-01 2020-07-28 Celgene Car Llc Heterocyclic compounds and their uses
CN103313978B (en) 2010-11-10 2015-04-15 霍夫曼-拉罗奇有限公司 Pyrazole aminopyrimidine derivatives as LRRK2 modulators
ES2665013T3 (en) 2010-11-10 2018-04-24 Celgene Car Llc EGFR selective mutant inhibitors and uses thereof
WO2012088266A2 (en) 2010-12-22 2012-06-28 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of fgfr3
CA2830129C (en) 2011-03-24 2016-07-19 Chemilia Ab Novel pyrimidine derivatives
JP6147727B2 (en) * 2011-04-01 2017-06-14 ユニヴァーシティー オブ ユタ リサーチ ファウンデーション Substituted N- (3- (pyrimidin-4-yl) phenyl) acrylamide analogs as tyrosine receptor kinase BTK inhibitors
PL2693881T3 (en) 2011-04-01 2020-03-31 University Of Utah Research Foundation Substituted n-phenylpyrimidin-2-amine analogs as inhibitors of the axl kinase
CN111471021B (en) 2011-04-22 2024-04-02 西格诺药品有限公司 Substituted diamino carboxamides and diamino carbonitrile pyrimidines, compositions thereof, and methods of treatment therewith
AU2012249801B2 (en) 2011-04-25 2016-05-19 Usher Iii Initiative, Inc. Pyrazolopyridazines and methods for treating retinal-degenerative diseases and hearing loss associated with Usher syndrome
KR101884010B1 (en) 2011-05-04 2018-07-31 어리어드 파마슈티칼스, 인코포레이티드 Compounds for inhibiting cell proliferation in egfr-driven cancers
ES2900230T3 (en) 2011-07-27 2022-03-16 Astrazeneca Ab 2-(2,4,5-anilino substituted)pyrimidine compounds
AU2012318896B2 (en) 2011-10-03 2017-03-02 The University Of North Carolina At Chapel Hill Pyrrolopyrimidine compounds for the treatment of cancer
EP2770830A4 (en) * 2011-10-28 2015-05-27 Celgene Avilomics Res Inc Methods of treating a bruton's tyrosine kinase disease or disorder
US9464089B2 (en) 2012-01-13 2016-10-11 Acea Biosciences Inc. Heterocyclic compounds and uses thereof
US9034885B2 (en) 2012-01-13 2015-05-19 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
CN110194748A (en) 2012-01-13 2019-09-03 艾森生物科学公司 Heterocyclic compound and its purposes as anticarcinogen
KR101985050B1 (en) 2012-01-17 2019-05-31 아스테라스 세이야쿠 가부시키가이샤 Pyrazine carboxamide compound
KR102081042B1 (en) * 2012-03-15 2020-02-26 셀젠 카르 엘엘씨 Solid forms of an epidermal growth factor receptor kinase inhibitor
WO2013138502A1 (en) 2012-03-15 2013-09-19 Celgene Avilomics Research, Inc. Salts of an epidermal growth factor receptor kinase inhibitor
WO2013169401A1 (en) * 2012-05-05 2013-11-14 Ariad Pharmaceuticals, Inc. Compounds for inhibiting cell proliferation in egfr-driven cancers
WO2013173518A1 (en) * 2012-05-16 2013-11-21 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase
AU2013266438B2 (en) 2012-05-22 2017-09-07 The University Of North Carolina At Chapel Hill Pyrimidine compounds for the treatment of cancer
NZ730134A (en) 2012-06-13 2018-07-27 Incyte Holdings Corp Substituted tricyclic compounds as fgfr inhibitors
WO2014026125A1 (en) 2012-08-10 2014-02-13 Incyte Corporation Pyrazine derivatives as fgfr inhibitors
EP2892538A4 (en) * 2012-09-04 2016-04-20 Celgene Avilomics Res Inc Methods of treating a bruton's tyrosine kinase disease or disorder
JP6313772B2 (en) 2012-10-04 2018-04-18 ユニヴァーシティー オブ ユタ リサーチ ファウンデーション Substituted N- (3- (pyrimidin-4-yl) phenyl) acrylamide analogs that are tyrosine receptor kinase BTK inhibitors
EP3680238A1 (en) * 2012-10-04 2020-07-15 University of Utah Research Foundation Substituted n-(3-(pyrimidin-4-yl)phenyl)acrylamide analogs as tyrosine receptor kinase btk inhibitors
EP2909211A4 (en) 2012-10-17 2016-06-22 Univ North Carolina Pyrazolopyrimidine compounds for the treatment of cancer
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
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
TW201427668A (en) * 2012-11-20 2014-07-16 Celgene Avilomics Res Inc Methods of treating a disease or disorder associated with Bruton's tyrosine kinase
TW201427666A (en) * 2012-11-20 2014-07-16 Celgene Avilomics Res Inc Methods of treating a disease or disorder associated with Bruton's tyrosine kinase
US20140142128A1 (en) * 2012-11-20 2014-05-22 Celgene Avilomics Research, Inc. Methods of treating a disease or disorder associated with bruton's tyrosine kinase
TW201427667A (en) * 2012-11-20 2014-07-16 Celgene Avilomics Res Inc Methods of treating a disease or disorder associated with Bruton's tyrosine kinase
EP2925752A4 (en) 2012-11-27 2016-06-01 Univ North Carolina Pyrimidine compounds for the treatment of cancer
US9266892B2 (en) 2012-12-19 2016-02-23 Incyte Holdings Corporation Fused pyrazoles as FGFR inhibitors
US9126950B2 (en) * 2012-12-21 2015-09-08 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
CA2899281C (en) 2013-01-25 2022-05-31 Rigel Pharmaceuticals, Inc. Pyrimidinediamine compounds for use in treating or preventing autoimmune alopecia
MX2015009952A (en) * 2013-02-08 2015-10-05 Celgene Avilomics Res Inc Erk inhibitors and uses thereof.
US20140357636A1 (en) * 2013-02-21 2014-12-04 Wayne Rothbaum Treatment of Skeletal-Related Disorders
TW201444798A (en) 2013-02-28 2014-12-01 必治妥美雅史谷比公司 Phenylpyrazole derivatives as potent ROCK1 and ROCK2 inhibitors
EP2961746B1 (en) 2013-02-28 2018-01-03 Bristol-Myers Squibb Company Phenylpyrazole derivatives as potent rock1 and rock2 inhibitors
US20160016934A1 (en) 2013-03-14 2016-01-21 Respivert Limited Kinase inhibitors
KR102334260B1 (en) 2013-03-14 2021-12-02 스미토모 다이니폰 파마 온콜로지, 인크. Jak2 and alk2 inhibitors and methods for their use
US9611283B1 (en) 2013-04-10 2017-04-04 Ariad Pharmaceuticals, Inc. Methods for inhibiting cell proliferation in ALK-driven cancers
CN109912594A (en) 2013-04-19 2019-06-21 因赛特控股公司 Bicyclic heterocycle as FGFR inhibitor
EA028756B1 (en) 2013-04-25 2017-12-29 Бэйджин, Лтд. Fused heterocyclic compounds as protein kinase inhibitors
WO2014182593A1 (en) * 2013-05-06 2014-11-13 Clovis Oncology, Inc. Salts of an epidermal growth factor receptor kinase inhibitor
GB201309085D0 (en) 2013-05-20 2013-07-03 Redx Pharma Ltd Compounds
PL3013337T3 (en) 2013-06-26 2019-06-28 Abbvie Inc. Primary carboxamides as btk inhibitors
CN117486865A (en) 2013-07-11 2024-02-02 艾森医药公司 Pyrimidine derivatives as kinase inhibitors
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
CA2921208C (en) 2013-08-12 2018-02-20 Taiho Pharmaceutical Co., Ltd. Fused pyrimidine compound or salt thereof
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
MX2016003292A (en) 2013-09-13 2016-06-24 Beigene Ltd Anti-pd1 antibodies and their use as therapeutics and diagnostics.
CN104262328B (en) * 2013-09-18 2016-09-07 北京韩美药品有限公司 The compound of suppression BTK and/or JAK3 kinase activity
SG10201901902WA (en) 2013-10-21 2019-04-29 Merck Patent Gmbh Heteroaryl compounds as btk inhibitors and uses thereof
EP4115886A1 (en) 2013-10-25 2023-01-11 Pharmacyclics LLC Methods of treating and preventing graft versus host disease
US9415049B2 (en) * 2013-12-20 2016-08-16 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
NZ715903A (en) 2014-01-30 2017-06-30 Signal Pharm 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
US11039621B2 (en) 2014-02-19 2021-06-22 Corning Incorporated Antimicrobial glass compositions, glasses and polymeric articles incorporating the same
US9622483B2 (en) 2014-02-19 2017-04-18 Corning Incorporated Antimicrobial glass compositions, glasses and polymeric articles incorporating the same
US11039620B2 (en) 2014-02-19 2021-06-22 Corning Incorporated Antimicrobial glass compositions, glasses and polymeric articles incorporating the same
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
CA2940666C (en) 2014-02-28 2022-08-23 Nimbus Lakshmi, Inc. Tyk2 inhibitors and uses thereof
EP3113779A4 (en) * 2014-03-07 2017-09-20 Celgene Avilomics Research, Inc. Methods of treating a bruton's tyrosine kinase disease or disorder
JP2017513942A (en) 2014-04-11 2017-06-01 ザ・ユニヴァーシティ・オヴ・ノース・キャロライナ・アト・チャペル・ヒル MERTK-specific pyrrolopyrimidine compounds
US9879008B2 (en) 2014-04-14 2018-01-30 Shanghai Haiyan Pharmaceutical Technology Co., Ltd. 2,3,4,6-tetra-substituted benzene-1,5-diamine derivatives, preparation method therefor and medicinal use thereof
WO2015160869A1 (en) * 2014-04-16 2015-10-22 Signal Pharmaceuticals, Llc Methods for treating cancer using tor kinase inhibitor combination therapy
US10300058B2 (en) 2014-04-18 2019-05-28 Xuanzhu Pharma Co., Ltd. Tyrosine kinase inhibitor and uses thereof
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
GB201410430D0 (en) 2014-06-11 2014-07-23 Redx Pharma Ltd Compounds
JP6526189B2 (en) 2014-07-03 2019-06-05 ベイジーン リミテッド Anti-PD-L1 antibodies and their use for therapy and diagnosis
CN105315285B (en) * 2014-07-25 2017-12-08 上海海雁医药科技有限公司 2,4 2 substitution 7H pyrrolo-es [2,3 d] pyrimidine derivatives, its preparation method and purposes pharmaceutically
PE20170684A1 (en) 2014-08-04 2017-06-15 Nuevolution As OPTIONALLY CONDENSED PYRIMIDINE HETEROCICLYL DERIVATIVES USEFUL FOR THE TREATMENT OF INFLAMMATORY, METABOLIC, ONCOLOGICAL AND AUTOIMMUNE DISEASES
AU2015303835B2 (en) 2014-08-12 2020-04-09 Monash University Lymph directing prodrugs
US10005760B2 (en) 2014-08-13 2018-06-26 Celgene Car Llc Forms and compositions of an ERK inhibitor
EP4070795A1 (en) 2014-08-25 2022-10-12 Salk Institute for Biological Studies Novel ulk1 inhibitors and methods using same
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
CN105399686B (en) * 2014-09-16 2018-05-22 深圳微芯生物科技有限责任公司 Pyrimidine derivatives, its preparation method and its application
CN105481778B (en) * 2014-09-16 2019-06-04 深圳微芯生物科技股份有限公司 Pyrimidine derivatives, preparation method and its application
US9458175B2 (en) 2014-09-17 2016-10-04 Celgene Avilomics Research, Inc. MK2 inhibitors and uses thereof
CA2959194A1 (en) * 2014-10-11 2016-04-14 Shanghai Hansoh Biomedical Co., Ltd. 4-substituted-2-(n-(5-substituted allyl amide)phenyl)amino)pyrimidine compound, its use as an egfr inhibitor, and preparation and application thereof
ES2699948T3 (en) 2014-10-14 2019-02-13 Sunshine Lake Pharma Co Ltd Substituted heteroaryl compounds and methods of use
US10851105B2 (en) 2014-10-22 2020-12-01 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
CN105601573B (en) * 2014-11-24 2021-07-02 中国科学院上海药物研究所 2-aminopyrimidine compound and pharmaceutical composition and application thereof
EP3230737A1 (en) 2014-12-11 2017-10-18 Merck Patent GmbH Assays for btk inhibitors
NZ732793A (en) 2014-12-16 2023-07-28 Signal Pharm Llc Formulations of 2-(tert-butylamino)-4-((1r,3r,4r)-3-hydroxy-4-methycyclohexylamino)-pyrimidine-5-carboxamide
WO2016100308A1 (en) 2014-12-16 2016-06-23 Signal Pharmaceuticals, Llc Methods for measurement of inhibition of c-jun n-terminal kinase in skin
CA2975260A1 (en) 2015-01-29 2016-08-04 Signal Pharmaceuticals Llc Isotopologues of 2-(tert-butylamino)-4-((1r,3r,4r)-3-hydroxy-4-methylcyclohexylamino)-pyrimidine-5-carboxamide
ES2667247T3 (en) 2015-01-30 2018-05-10 Taiho Pharmaceutical Co., Ltd. Novel salt of condensed pyrimidine compound and crystal thereof
PT3120852T (en) * 2015-01-30 2019-07-16 Taiho Pharmaceutical Co Ltd Preventive and/or therapeutic agent for immune diseases
US9580423B2 (en) 2015-02-20 2017-02-28 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
MA41551A (en) 2015-02-20 2017-12-26 Incyte Corp BICYCLIC HETEROCYCLES USED AS FGFR4 INHIBITORS
TWI712601B (en) 2015-02-20 2020-12-11 美商英塞特公司 Bicyclic heterocycles as fgfr inhibitors
ES2930585T3 (en) 2015-02-27 2022-12-19 Nimbus Lakshmi Inc TYK2 inhibitors and uses thereof
WO2017004134A1 (en) 2015-06-29 2017-01-05 Nimbus Iris, Inc. Irak inhibitors and uses thereof
JP6853234B2 (en) * 2015-07-09 2021-03-31 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 derivatives and their use as BTK inhibitors
EP3325432B1 (en) 2015-07-24 2020-09-02 Celgene Corporation Methods of synthesis of (1r,2r,5r)-5-amino-2-methylcyclohexanol hydrochloride and intermediates useful therein
WO2017040617A1 (en) 2015-08-31 2017-03-09 Pharmacyclics Llc Btk inhibitor combinations for treating multiple myeloma
EP4327809A2 (en) 2015-09-02 2024-02-28 Takeda Pharmaceutical Company Limited Tyk2 inhibitors and uses thereof
AU2016318229A1 (en) 2015-09-08 2018-03-29 Monash University Lymph directing prodrugs
US10683308B2 (en) 2015-09-11 2020-06-16 Navitor Pharmaceuticals, Inc. Rapamycin analogs and uses thereof
CA2998796A1 (en) 2015-09-16 2017-03-23 Loxo Oncology, Inc. Pyrazolopyrimidine derivatives as btk inhibitors for the treatment of cancer
CN106554347B (en) * 2015-09-25 2020-10-30 浙江博生医药有限公司 EGFR kinase inhibitor and preparation method and application thereof
RU2018115334A (en) 2015-10-09 2019-11-11 Ацея Терапьютикс, Инк. PHARMACEUTICAL SALTS, PHYSICAL FORMS AND COMPOSITIONS OF KYNASE, Pyrrolopyrimidine Inhibitors, and METHODS FOR PRODUCING THEM
TWI747846B (en) * 2015-10-21 2021-12-01 日商大塚製藥股份有限公司 Benzolactam compounds
AU2016342027B2 (en) 2015-10-23 2021-05-13 Navitor Pharmaceuticals, Inc. Modulators of Sestrin-GATOR2 interaction and uses thereof
EP3389664A4 (en) 2015-12-14 2020-01-08 Raze Therapeutics Inc. Caffeine inhibitors of mthfd2 and uses thereof
MX2021003478A (en) 2015-12-16 2022-07-25 Loxo Oncology Inc Compounds useful as kinase inhibitors.
CN107021963A (en) 2016-01-29 2017-08-08 北京诺诚健华医药科技有限公司 Pyrazole fused ring analog derivative, its preparation method and its application in treating cancer, inflammation and immunity disease
WO2017156179A1 (en) 2016-03-09 2017-09-14 Raze Therapeutics, Inc. 3-phosphoglycerate dehydrogenase inhibitors and uses thereof
SI3426243T1 (en) 2016-03-09 2021-11-30 Raze Therapeutics, Inc. 3-phosphoglycerate dehydrogenase inhibitors and uses thereof
US10709708B2 (en) 2016-03-17 2020-07-14 The University Of North Carolina At Chapel Hill Method of treating cancer with a combination of MER tyrosine kinase inhibitor and an epidermal growth factor receptor (EGFR) inhibitor
CN109153722A (en) 2016-04-08 2019-01-04 X4 制药有限公司 Method for treating cancer
CN105968056A (en) * 2016-05-28 2016-09-28 大连医科大学 Diarylpyrimidine compound, composition and application
WO2017223243A1 (en) 2016-06-21 2017-12-28 X4 Pharmaceuticals, Inc. Cxcr4 inhibitors and uses thereof
CA3027495A1 (en) 2016-06-21 2017-12-28 X4 Pharmaceuticals, Inc. Cxcr4 inhibitors and uses thereof
EP3471727B1 (en) 2016-06-21 2020-12-09 X4 Pharmaceuticals, Inc. Cxcr4 inhibitors and uses thereof
NZ749997A (en) 2016-07-05 2022-11-25 Beigene Ltd Combination of a pd-l antagonist and a raf inhibitor for treating cancer
BR112019003205A8 (en) 2016-08-16 2023-01-17 Beigene Ltd CRYSTALLINE FORM OF (S) -7- (1-ACRYLOYLPIPERIDIN-4-IL) -2- (4-PHENOXYPHENYL) -4,5,6,7-TETRAHYDROPYRAZOLE [1,5-A]PYRIMIDINE-3-CARBOXAMIDE , ITS PREPARATION AND USES
DK3500299T3 (en) 2016-08-19 2024-01-29 Beigene Switzerland Gmbh Combination of zanubrutinib with an anti-CD20 or an anti-PD-1 antibody for use in the treatment of cancer
CN106316886A (en) * 2016-08-24 2017-01-11 泰州精英化成医药科技有限公司 Cis form-3- (tert butoxycarbonylamino) naphthenic acid preparing method
EP3515414B1 (en) 2016-09-19 2022-11-30 MEI Pharma, Inc. Combination therapy
ES2930198T3 (en) 2016-10-14 2022-12-07 Nimbus Lakshmi Inc TYK2 inhibitors and uses thereof
US10647713B2 (en) 2016-10-21 2020-05-12 Nimbus Lakshmi, Inc. TYK2 inhibitors and uses thereof
CN107973754B (en) * 2016-10-25 2020-04-14 清华大学 Small molecule inhibitor, preparation method thereof and application thereof in treatment of multiple myeloma
JP2020023441A (en) * 2016-11-02 2020-02-13 国立大学法人九州大学 Novel compound useful for egfr inhibition and tumor treatment
MA46783A (en) 2016-11-03 2019-09-11 Juno Therapeutics Inc T-CELL THERAPY AND BTK INHIBITOR POLYTHERAPY
US10414727B2 (en) 2016-11-08 2019-09-17 Navitor Pharmaceuticals, Inc. Phenyl amino piperidine mTORC inhibitors and uses thereof
EP3548005A4 (en) 2016-11-29 2020-06-17 Puretech Health LLC Exosomes for delivery of therapeutic agents
US11091451B2 (en) 2016-12-05 2021-08-17 Raze Therapeutics, Inc. SHMT inhibitors and uses thereof
WO2018115203A1 (en) 2016-12-23 2018-06-28 Bicyclerd Limited Peptide derivatives having novel linkage structures
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
EP3565638B8 (en) 2017-01-06 2024-04-10 BicycleRD Limited Bicycle conjugate for treating cancer
CN108329274B (en) * 2017-01-13 2021-09-03 正大天晴药业集团股份有限公司 Bruton's tyrosine kinase inhibitors
EP3573989A4 (en) 2017-01-25 2020-11-18 Beigene, Ltd. Crystalline forms of (s) -7- (1- (but-2-ynoyl) piperidin-4-yl) -2- (4-phenoxyphenyl) -4, 5, 6, 7-tetrahy dropyrazolo [1, 5-a]pyrimidine-3-carboxamide, preparation, 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
EP3592746B1 (en) 2017-03-08 2024-01-24 Takeda Pharmaceutical Company Limited Tyk2 inhibitors, uses, and methods for production thereof
EP3375778A1 (en) 2017-03-14 2018-09-19 Artax Biopharma Inc. Aryl-piperidine derivatives
EP3375784A1 (en) 2017-03-14 2018-09-19 Artax Biopharma Inc. Aza-dihydro-acridone derivatives
CA3059072A1 (en) 2017-04-07 2018-10-11 ACEA Therapeutics, Inc. Pharmaceutical salts, physical forms, and compositions of pyrrolopyrimidine kinase inhibitors, and methods of making same
JOP20190233A1 (en) 2017-04-14 2019-10-02 Biogen Ma Inc Benzoazepine analogs as inhibiting agents for bruton's tyrosine kinase
GB201706327D0 (en) 2017-04-20 2017-06-07 Otsuka Pharma Co Ltd A pharmaceutical compound
US10857196B2 (en) 2017-04-27 2020-12-08 Bicycletx Limited Bicyclic peptide ligands and uses thereof
AR111960A1 (en) 2017-05-26 2019-09-04 Incyte Corp CRYSTALLINE FORMS OF A FGFR INHIBITOR AND PROCESSES FOR ITS PREPARATION
CN110996955A (en) 2017-06-22 2020-04-10 细胞基因公司 Treatment of hepatocellular carcinoma characterized by hepatitis B virus infection
CN111032678A (en) 2017-06-26 2020-04-17 拜西克尔德有限公司 Bicyclic peptide ligands with detectable moieties and uses thereof
EP3645569A4 (en) 2017-06-26 2021-03-24 BeiGene, Ltd. Immunotherapy for hepatocellular carcinoma
JP7216705B2 (en) 2017-07-28 2023-02-02 ニンバス ラクシュミ, インコーポレイテッド TYK2 inhibitors and methods of use thereof
CN110997677A (en) 2017-08-12 2020-04-10 百济神州有限公司 Btk inhibitors with improved dual selectivity
US20200283482A1 (en) 2017-08-14 2020-09-10 Bicyclerd Limited Bicyclic peptide ligand prr-a conjugates and uses thereof
EP3668887A1 (en) 2017-08-14 2020-06-24 Bicyclerd Limited Bicyclic peptide ligand sting conjugates and uses thereof
CN111032630B (en) * 2017-08-18 2022-12-16 北京韩美药品有限公司 Compound, pharmaceutical composition, application and application thereof
US11883497B2 (en) 2017-08-29 2024-01-30 Puretech Lyt, Inc. Lymphatic system-directing lipid prodrugs
CA3077739A1 (en) 2017-08-29 2019-03-07 Puretech Lyt, Inc. Lymphatic system-directing lipid prodrugs
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
WO2019060693A1 (en) 2017-09-22 2019-03-28 Kymera Therapeutics, Inc. Crbn ligands and uses thereof
IL273432B (en) 2017-09-22 2022-09-01 Kymera Therapeutics Inc Protein degraders and uses thereof
US11166959B2 (en) 2017-11-06 2021-11-09 Bristol-Myers Squibb Company Isofuranone compounds useful as HPK1 inhibitors
EP3710006A4 (en) 2017-11-19 2021-09-01 Sunshine Lake Pharma Co., Ltd. Substituted heteroaryl compounds and methods of use
CN111801334B (en) 2017-11-29 2023-06-09 百济神州瑞士有限责任公司 Treatment of indolent or invasive B-cell lymphomas using combinations comprising BTK inhibitors
US11304954B2 (en) 2017-12-19 2022-04-19 Puretech Lyt, Inc. Lipid prodrugs of mycophenolic acid and uses thereof
WO2019126378A1 (en) 2017-12-19 2019-06-27 Ariya Therapeutics, Inc. Lipid prodrugs of mycophenolic acid and uses thereof
TWI825046B (en) 2017-12-19 2023-12-11 英商拜西可泰克斯有限公司 Bicyclic peptide ligands specific for epha2
US11608345B1 (en) 2017-12-19 2023-03-21 Puretech Lyt, Inc. Lipid prodrugs of rapamycin and its analogs and uses thereof
GB201721265D0 (en) 2017-12-19 2018-01-31 Bicyclerd Ltd Bicyclic peptide ligands specific for EphA2
SG11202005912PA (en) 2017-12-26 2020-07-29 Kymera Therapeutics Inc Irak degraders and uses thereof
US11485743B2 (en) 2018-01-12 2022-11-01 Kymera Therapeutics, Inc. Protein degraders and uses thereof
EP3737675A4 (en) 2018-01-12 2022-01-05 Kymera Therapeutics, Inc. Crbn ligands and uses thereof
SG11202006832YA (en) 2018-01-29 2020-08-28 Merck Patent Gmbh Gcn2 inhibitors and uses thereof
MX2020007799A (en) 2018-01-29 2020-09-18 Merck Patent Gmbh Gcn2 inhibitors and uses thereof.
TWI798334B (en) 2018-01-31 2023-04-11 大陸商迪哲(江蘇)醫藥股份有限公司 Erbb/btk inhibitors
IL301120A (en) 2018-02-27 2023-05-01 Artax Biopharma Inc Chromene derivatives as inhibitors of tcr-nck interaction
WO2019195753A1 (en) 2018-04-05 2019-10-10 Tolero Pharmaceuticals, Inc. Axl kinase inhibitors and use of the same
EP3784669B1 (en) 2018-04-24 2023-10-25 Vertex Pharmaceuticals Incorporated Pteridinone compounds and uses thereof
US10815225B2 (en) 2018-04-24 2020-10-27 Merck Patent Gmbh Antiproliferation compounds and uses thereof
WO2019207463A1 (en) 2018-04-26 2019-10-31 Pfizer Inc. 2-amino-pyridine or 2-amino-pyrimidine derivatives as cyclin dependent kinase inhibitors
CN112584902A (en) 2018-05-03 2021-03-30 朱诺治疗学股份有限公司 Combination therapy of Chimeric Antigen Receptor (CAR) T cell therapy and kinase inhibitors
MA52494A (en) 2018-05-04 2021-03-10 Incyte Corp SOLID FORMS OF FGFR INHIBITOR AND THEIR PREPARATION PROCEDURES
CR20200591A (en) 2018-05-04 2021-03-31 Incyte Corp Salts of an fgfr inhibitor
BR112020025283A2 (en) 2018-06-15 2021-03-09 Navitor Pharmaceuticals, Inc. RAPAMICIN ANALOGS AND USES OF THE SAME
US11180531B2 (en) 2018-06-22 2021-11-23 Bicycletx Limited Bicyclic peptide ligands specific for Nectin-4
GB201810316D0 (en) 2018-06-22 2018-08-08 Bicyclerd Ltd Peptide ligands for binding to EphA2
WO2020010177A1 (en) 2018-07-06 2020-01-09 Kymera Therapeutics, Inc. Tricyclic crbn ligands and uses thereof
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
US10548889B1 (en) 2018-08-31 2020-02-04 X4 Pharmaceuticals, Inc. Compositions of CXCR4 inhibitors and methods of preparation and use
WO2020051424A1 (en) 2018-09-07 2020-03-12 Pic Therapeutics Eif4e inhibitors and uses thereof
US11066404B2 (en) 2018-10-11 2021-07-20 Incyte Corporation Dihydropyrido[2,3-d]pyrimidinone compounds as CDK2 inhibitors
AU2019360941A1 (en) 2018-10-15 2021-04-29 Takeda Pharmaceutical Company Limited TYK2 inhibitors and uses thereof
US10919937B2 (en) 2018-10-23 2021-02-16 Bicycletx Limited Bicyclic peptide ligands and uses thereof
CN109608444B (en) * 2018-11-27 2022-02-11 中国药科大学 Isoindolinone-containing ERK inhibitor and preparation method and application thereof
JP2022509260A (en) 2018-11-30 2022-01-20 ニンバス ラクシュミ, インコーポレイテッド TYK2 inhibitors and their use
MX2021006154A (en) 2018-11-30 2021-08-24 Kymera Therapeutics Inc Irak degraders and uses thereof.
EP3670659A1 (en) 2018-12-20 2020-06-24 Abivax Biomarkers, and uses in treatment of viral infections, inflammations, or cancer
JP2022518505A (en) 2019-01-23 2022-03-15 ニンバス ラクシュミ, インコーポレイテッド TYK2 inhibitors and their use
CN109776495A (en) * 2019-01-31 2019-05-21 李佳睿 Antitumoral compounds and preparation method thereof and purposes
WO2020165600A1 (en) 2019-02-14 2020-08-20 Bicycletx Limited Bicyclic peptide ligand sting conjugates and uses thereof
WO2020168197A1 (en) 2019-02-15 2020-08-20 Incyte Corporation Pyrrolo[2,3-d]pyrimidinone compounds as cdk2 inhibitors
US11472791B2 (en) 2019-03-05 2022-10-18 Incyte Corporation Pyrazolyl pyrimidinylamine compounds as CDK2 inhibitors
WO2020185532A1 (en) 2019-03-08 2020-09-17 Incyte Corporation Methods of treating cancer with an fgfr inhibitor
US11919904B2 (en) 2019-03-29 2024-03-05 Incyte Corporation Sulfonylamide compounds as CDK2 inhibitors
WO2020201753A1 (en) 2019-04-02 2020-10-08 Bicycletx Limited Bicycle toxin conjugates and uses thereof
CN113939300A (en) 2019-04-05 2022-01-14 凯麦拉医疗公司 STAT degradants and uses thereof
WO2020223558A1 (en) 2019-05-01 2020-11-05 Incyte Corporation Tricyclic amine compounds as cdk2 inhibitors
WO2020223469A1 (en) 2019-05-01 2020-11-05 Incyte Corporation N-(1-(methylsulfonyl)piperidin-4-yl)-4,5-di hydro-1h-imidazo[4,5-h]quinazolin-8-amine derivatives and related compounds as cyclin-dependent kinase 2 (cdk2) inhibitors for treating cancer
MX2021013662A (en) 2019-05-10 2022-03-11 Deciphera Pharmaceuticals Llc Heteroarylaminopyrimidine amide autophagy inhibitors and methods of use thereof.
BR112021022504A2 (en) 2019-05-10 2022-04-12 Deciphera Pharmaceuticals Llc Phenylaminopyrimidine amide autophagy inhibitors and methods of using them
KR20220034739A (en) 2019-05-31 2022-03-18 이케나 온콜로지, 인코포레이티드 TEAD inhibitors and uses thereof
CA3143489A1 (en) 2019-06-17 2020-12-24 Deciphera Pharmaceuticals, Llc Aminopyrimidine amide autophagy inhibitors and methods of use thereof
US20220388968A1 (en) * 2019-06-18 2022-12-08 Purdue Research Foundation Inhibitors of erythrocyte band 3 tyrosine phosphorylation and uses thereof
WO2021007269A1 (en) 2019-07-09 2021-01-14 Incyte Corporation Bicyclic heterocycles as fgfr inhibitors
CN110305140B (en) * 2019-07-30 2020-08-04 上海勋和医药科技有限公司 Dihydropyrrolopyrimidines selective JAK2 inhibitors
MX2022001940A (en) 2019-08-14 2022-05-10 Incyte Corp Imidazolyl pyrimidinylamine compounds as cdk2 inhibitors.
AU2020345962A1 (en) 2019-09-11 2022-03-31 Vincere Biosciences, Inc. USP30 inhibitors and uses thereof
BR112022004451A2 (en) 2019-09-13 2022-06-21 Nimbus Saturn Inc hpk1 antagonists and uses thereof
CN112574207B (en) * 2019-09-30 2023-04-11 南京药石科技股份有限公司 ERK1/2 protein kinase inhibitor and application thereof
PE20221905A1 (en) 2019-10-11 2022-12-23 Incyte Corp BICYCLIC AMINES AS INHIBITORS OF CDK2
BR112022007163A2 (en) 2019-10-14 2022-08-23 Incyte Corp BICYCLIC HETEROCYCLES AS FGFR INHIBITORS
US11566028B2 (en) 2019-10-16 2023-01-31 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
JP2023500352A (en) * 2019-11-08 2023-01-05 深▲セン▼微芯生物科技股▲フン▼有限公司 Compounds for preventing or treating graft-versus-host disease
US20230000863A1 (en) * 2019-11-26 2023-01-05 Shenzhen Chipscreen Biosciences Co., Ltd. Composition containing aromatic heterocyclic compound in amorphous form, and preparation method therefor and use thereof
CA3162010A1 (en) 2019-12-04 2021-06-10 Incyte Corporation Derivatives of an fgfr inhibitor
WO2021113479A1 (en) 2019-12-04 2021-06-10 Incyte Corporation Tricyclic heterocycles as fgfr inhibitors
MX2022006807A (en) 2019-12-05 2022-09-12 Anakuria Therapeutics Inc Rapamycin analogs and uses thereof.
MX2022007576A (en) 2019-12-17 2022-09-23 Kymera Therapeutics Inc Irak degraders and uses thereof.
WO2021127283A2 (en) 2019-12-17 2021-06-24 Kymera Therapeutics, Inc. Irak degraders and uses thereof
WO2021124277A1 (en) 2019-12-20 2021-06-24 Nuevolution A/S Compounds active towards nuclear receptors
CA3162502A1 (en) 2019-12-23 2021-07-01 Yi Zhang Smarca degraders and uses thereof
CN111620852B (en) * 2020-01-19 2023-05-09 镇江植生源农业科技有限公司 5-chloro-pyrimidine-2, 4-diamine compound, and preparation method and application thereof
IL296139A (en) 2020-03-03 2022-11-01 Pic Therapeutics Inc Eif4e inhibitors and uses thereof
TW202200543A (en) 2020-03-19 2022-01-01 美商凱麥拉醫療公司 Mdm2 degraders and uses thereof
EP4126875A1 (en) 2020-03-31 2023-02-08 Nuevolution A/S Compounds active towards nuclear receptors
CA3174252A1 (en) 2020-03-31 2021-10-07 Nuevolution A/S Compounds active towards nuclear receptors
TW202210483A (en) 2020-06-03 2022-03-16 美商凱麥拉醫療公司 Crystalline forms of irak degraders
CN111646978B (en) * 2020-06-11 2021-12-21 浙江大学 N2-substituted alkoxy aromatic ring-2-aminopyrimidine derivatives and application thereof
US20230257350A1 (en) * 2020-07-02 2023-08-17 The Cleveland Clinic Foundation Compounds and methods of promoting oligodendrocyte precursor differentiation
EP4196792A1 (en) 2020-08-17 2023-06-21 BicycleTX Limited Bicycle conjugates specific for nectin-4 and uses thereof
WO2022072632A1 (en) * 2020-09-30 2022-04-07 Scorpion Therapeutics, Inc. Bicyclic compounds for use in the treatment cancer
CA3200814A1 (en) 2020-12-02 2022-06-09 Alfredo C. Castro Tead inhibitors and uses thereof
WO2022120353A1 (en) 2020-12-02 2022-06-09 Ikena Oncology, Inc. Tead inhibitors and uses thereof
JP2024506858A (en) * 2021-02-02 2024-02-15 リミナル・バイオサイエンシーズ・リミテッド GPR84 antagonists and their uses
CA3206501A1 (en) 2021-02-02 2022-08-11 Shaun Abbott Gpr84 antagonists and uses thereof
AU2022220869A1 (en) 2021-02-15 2023-08-24 Kymera Therapeutics, Inc. Irak4 degraders and uses thereof
JP2024509192A (en) 2021-03-05 2024-02-29 ニンバス サターン, インコーポレイテッド HPK1 antagonists and their uses
WO2022212893A1 (en) 2021-04-02 2022-10-06 Biogen Ma Inc. Combination treatment methods of multiple sclerosis
EP4323066A1 (en) 2021-04-16 2024-02-21 Ikena Oncology, Inc. Mek inhibitors and uses thereof
TW202313611A (en) 2021-06-09 2023-04-01 美商英塞特公司 Tricyclic heterocycles as fgfr inhibitors
US20230150997A1 (en) 2021-08-25 2023-05-18 PIC Therapeutics, Inc. Eif4e inhibitors and uses thereof
US20230134932A1 (en) 2021-08-25 2023-05-04 PIC Therapeutics, Inc. Eif4e inhibitors and uses thereof
WO2023114984A1 (en) 2021-12-17 2023-06-22 Ikena Oncology, Inc. Tead inhibitors and uses thereof
WO2023173057A1 (en) 2022-03-10 2023-09-14 Ikena Oncology, Inc. Mek inhibitors and uses thereof
WO2023173053A1 (en) 2022-03-10 2023-09-14 Ikena Oncology, Inc. Mek inhibitors and uses thereof
WO2023211889A1 (en) 2022-04-25 2023-11-02 Ikena Oncology, Inc. Polymorphic compounds and uses 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
WO2023230205A1 (en) 2022-05-25 2023-11-30 Ikena Oncology, Inc. Mek inhibitors and uses thereof
US11786531B1 (en) 2022-06-08 2023-10-17 Beigene Switzerland Gmbh Methods of treating B-cell proliferative disorder
WO2024028364A1 (en) 2022-08-02 2024-02-08 Liminal Biosciences Limited Aryl-triazolyl and related gpr84 antagonists and uses thereof
WO2024028365A1 (en) 2022-08-02 2024-02-08 Liminal Biosciences Limited Substituted pyridone gpr84 antagonists and uses thereof
WO2024028363A1 (en) 2022-08-02 2024-02-08 Liminal Biosciences Limited Heteroaryl carboxamide and related gpr84 antagonists and uses thereof
CN115448906B (en) * 2022-09-26 2024-04-02 深圳大学 2-anilinopyrimidine derivative and preparation method and application thereof

Family Cites Families (174)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4032637A (en) 1972-09-26 1977-06-28 Sandoz Ltd. Method of promoting sleep
US5516931A (en) 1982-02-01 1996-05-14 Northeastern University Release tag compounds producing ketone signal groups
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
GB8608335D0 (en) 1986-04-04 1986-05-08 Pfizer Ltd Pharmaceutically acceptable salts
US5453510A (en) 1990-07-13 1995-09-26 Burroughs Wellcome Co. Neuromuscular blocking agents
JPH0741461A (en) 1993-05-27 1995-02-10 Eisai Co Ltd Sulfonic acid ester derivative
EP0813525B9 (en) 1995-03-10 2004-02-04 Berlex Laboratories, Inc. Benzamidine derivatives their preparation and their use as anti-coagulants
GB9523675D0 (en) * 1995-11-20 1996-01-24 Celltech Therapeutics Ltd Chemical compounds
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
US6432963B1 (en) 1997-12-15 2002-08-13 Yamanouchi Pharmaceutical Co., Ltd. Pyrimidine-5-carboxamide derivatives
US6303652B1 (en) * 1998-08-21 2001-10-16 Hughes Institute BTK inhibitors and methods for their identification and use
AU5438299A (en) * 1998-08-29 2000-03-21 Astrazeneca Ab Pyrimidine compounds
HU230394B1 (en) 1998-11-10 2016-04-28 Janssen Pharmaceutica N.V. Hiv replication inhibiting pyrimidines
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
CA2394727A1 (en) 1999-12-28 2001-07-05 Pharmacopeia, Inc. Pyrimidine and triazine kinase inhibitors
EP1257546A1 (en) * 2000-02-17 2002-11-20 Amgen Inc. Kinase inhibitors
GB0004887D0 (en) * 2000-03-01 2000-04-19 Astrazeneca Uk Ltd Chemical compounds
GB0004888D0 (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
US7034019B2 (en) 2000-05-08 2006-04-25 Janssen Pharmaceutica N.V. Prodrugs of HIV replication inhibiting pyrimidines
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
NZ531853A (en) * 2001-11-01 2006-02-24 Janssen Pharmaceutica Nv Heteroaryl amines as glycogen synthase kinase 3beta inhibitors (gsk3 inhibitors)
TWI329105B (en) * 2002-02-01 2010-08-21 Rigel Pharmaceuticals Inc 2,4-pyrimidinediamine compounds and their uses
JP2005522438A (en) * 2002-02-08 2005-07-28 スミスクライン ビーチャム コーポレーション Pyrimidine compounds
GB0206215D0 (en) * 2002-03-15 2002-05-01 Novartis Ag Organic compounds
CA2478981A1 (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
IL166241A (en) 2002-07-29 2011-12-29 Rigel Pharmaceuticals Inc 2,4-pyrimidinediamine compounds for use in the treatment of autoimmune diseases
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
NZ541902A (en) 2003-02-07 2008-12-24 Janssen Pharmaceutica Nv Pyrimidine derivatives for the prevention of HIV infection
US7514446B2 (en) * 2003-02-20 2009-04-07 Smithkline Beecham Corporation Pyrimidine compounds
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
JP4886511B2 (en) * 2003-07-30 2012-02-29 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 Method for treating or preventing autoimmune diseases with 2,4-pyrimidinediamine compounds
EP1663242B1 (en) * 2003-08-07 2011-04-27 Rigel Pharmaceuticals, Inc. 2,4-pyrimidinediamine compounds and uses as anti-proliferative agents
TWI378923B (en) * 2003-08-15 2012-12-11 Novartis Ag Pyrimidine derivatives
GB0321710D0 (en) 2003-09-16 2003-10-15 Novartis Ag Organic compounds
MXPA06003054A (en) * 2003-09-18 2006-05-31 Novartis Ag 2,4-di (phenylamino) pyrimidines useful in the treatment of proliferative disorders.
JP2007514775A (en) 2003-12-19 2007-06-07 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 Stereoisomers and stereoisomer mixtures of 1- (2,4-pyrimidinediamino) -2-cyclopentanecarboximide synthesis intermediates
JP5283336B2 (en) * 2004-01-12 2013-09-04 ワイエム・バイオサイエンシズ・オーストラリア・ピーティーワイ・リミテッド Selective kinase inhibitor
TW200526219A (en) 2004-01-16 2005-08-16 Wyeth Corp Quinoline intermediates of receptor tyrosine kinase inhibitors and the synthesis thereof
ATE545413T1 (en) * 2004-03-15 2012-03-15 Lilly Co Eli ANTAGONISTS OF THE OPIOID RECEPTOR
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
EP1766068A4 (en) * 2004-06-04 2010-03-03 Genentech Inc Egfr mutations
US7521457B2 (en) 2004-08-20 2009-04-21 Boehringer Ingelheim International Gmbh Pyrimidines as PLK inhibitors
GB0419161D0 (en) * 2004-08-27 2004-09-29 Novartis Ag Organic compounds
WO2006034474A2 (en) * 2004-09-23 2006-03-30 Reddy Us Therapeutics, Inc. Novel pyridine compounds, process for their preparation and compositions containing them
EP1797048B1 (en) * 2004-09-30 2011-08-17 Tibotec Pharmaceuticals Hiv inhibiting 5-carbo- or heterocyclic substituted pyrimidines
CN101039919A (en) * 2004-10-13 2007-09-19 惠氏公司 N-benzenesulfonyl substituted anilino-pyrimidine analogs
AU2005295183A1 (en) 2004-10-20 2006-04-27 CAOnyx Therapeutics, Inc. Labeled compounds for proteasome inhibition
TW200628463A (en) 2004-11-10 2006-08-16 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
ES2380550T3 (en) 2004-11-24 2012-05-16 Rigel Pharmaceuticals, Inc. Spiro-2,4-pyrimidinediamine compounds and their uses
MY169441A (en) 2004-12-08 2019-04-11 Janssen Pharmaceutica Nv 2,4, (4,6) pyrimidine derivatives
ATE519759T1 (en) 2004-12-30 2011-08-15 Exelixis Inc PYRIMIDINE DERIVATIVES AS KINASE MODULATORS AND METHODS OF APPLICATION
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
US7449458B2 (en) 2005-01-19 2008-11-11 Rigel Pharmaceuticals, Inc. Prodrugs of 2,4-pyrimidinediamine compounds and their uses
CA2597673C (en) * 2005-02-11 2014-07-08 Harold Varmus Methods and compositions for detecting a drug resistant egfr mutant
MX2007011500A (en) * 2005-03-16 2007-11-21 Targegen Inc Pyrimidine compounds and methods of use.
DE102005016634A1 (en) 2005-04-12 2006-10-19 Merck Patent Gmbh Novel aza heterocycles as kinase inhibitors
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
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
US20070203161A1 (en) * 2006-02-24 2007-08-30 Rigel Pharmaceuticals, Inc. Compositions and methods for inhibition of the jak pathway
ZA200710206B (en) * 2005-06-08 2009-12-30 Rigel Pharmaceuticals Inc Compositions and methods for inhibition of the JAK path-way
AU2006283592A1 (en) 2005-08-22 2007-03-01 Amgen Inc. Pyrazolopyridine and pyrazolopyrimidine compounds useful as kinase enzymes modulators
CA2626789A1 (en) 2005-10-21 2007-04-26 Exelixis, Inc. Pyrimidinones as casein kinase ii (ck2) modulators
RU2448959C2 (en) 2005-11-01 2012-04-27 Таргеджен, Инк. Bi-aryl-metha-pyrimidine kinase inhibitors
CN101300234A (en) * 2005-11-03 2008-11-05 Irm责任有限公司 Compounds and compositions for protein kinase inhbitors
US7713987B2 (en) 2005-12-06 2010-05-11 Rigel Pharmaceuticals, Inc. Pyrimidine-2,4-diamines and their uses
EP1968950A4 (en) 2005-12-19 2010-04-28 Genentech Inc Pyrimidine kinase inhibitors
TW200736232A (en) * 2006-01-26 2007-10-01 Astrazeneca Ab Pyrimidine derivatives
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
EP1984357B1 (en) 2006-02-17 2013-10-02 Rigel Pharmaceuticals, Inc. 2,4-pyrimidinediamine compounds for treating or preventing autoimmune diseases
ES2622493T3 (en) * 2006-02-24 2017-07-06 Rigel Pharmaceuticals, Inc. Compositions and methods for inhibiting the JAK route
DK2004632T3 (en) 2006-03-30 2014-06-16 Janssen R & D Ireland HIV INHIBITIVE 5-AMIDOSUBSTITUTED PYRIMIDINES
SI2004641T1 (en) 2006-03-30 2011-02-28 Tibotec Pharm Ltd Hiv inhibiting 5-(hydroxymethylene and aminomethylene) substituted pyrimidines
GB0608386D0 (en) 2006-04-27 2006-06-07 Senexis Ltd Compounds
EP2865381A1 (en) 2006-05-18 2015-04-29 Pharmacyclics, Inc. ITK inhibitors for treating blood cell malignancies
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
AU2007269540B2 (en) 2006-07-05 2013-06-27 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
CN101610676B (en) * 2006-09-22 2013-03-27 药品循环公司 Inhibitors of bruton's tyrosine kinase
US8193197B2 (en) * 2006-10-19 2012-06-05 Rigel Pharmaceuticals, Inc. Compositions and methods for inhibition of the JAK pathway
WO2008054827A2 (en) * 2006-11-03 2008-05-08 Pharmacyclics, Inc. Bruton's tyrosine kinase activity probe and method of using
EP2420505A1 (en) 2006-11-21 2012-02-22 Rigel Pharmaceuticals, Inc. Prodrug salts of 2, 4- pyrimidinediamine compounds and their uses
CN103641833A (en) * 2006-12-08 2014-03-19 Irm责任有限公司 Compounds and compositions as protein kinase inhibitors
US20100144732A1 (en) 2006-12-19 2010-06-10 Krueger Elaine B Pyrimidine kinase inhibitors
EP1939185A1 (en) 2006-12-20 2008-07-02 Bayer Schering Pharma Aktiengesellschaft New types of hetaryl-phenylendiamin-pyrimidines as protein kinase inhibitors for the treatment of cancer
MX2009007006A (en) 2006-12-29 2009-07-09 Tibotec Pharm Ltd Hiv inhibiting 5,6-substituted pyrimidines.
AU2007341227C1 (en) 2006-12-29 2013-08-29 Janssen Sciences Ireland Uc HIV inhibiting 6-substituted pyrimidines
PE20081636A1 (en) * 2007-01-26 2009-01-10 Smithkline Beecham Corp ANTHRANILAMIDE INHIBITORS FOR AURORA KINASE
AU2008210266B2 (en) 2007-01-31 2013-09-05 Ym Biosciences Australia Pty Ltd Thiopyrimidine-based compounds and uses thereof
DE102007010801A1 (en) 2007-03-02 2008-09-04 Bayer Cropscience Ag Use of new and known 2,4-diaminopyrimidine derivatives as fungicides, especially for controlling phytopathogenic fungi
BRPI0809188A2 (en) 2007-03-20 2014-09-16 Smithkline Beecham Corp COMPOUND OR A SALT THEREOF, PHARMACEUTICAL COMPOSITION, METHODS FOR TREATING DISTURBANCE IN A MAMMALIAN AND TREATING CANCER IN A MAMMALIAN, AND USING A COMPOUND
KR20090121399A (en) 2007-03-20 2009-11-25 스미스클라인 비참 코포레이션 Chemical compounds
WO2008118822A1 (en) 2007-03-23 2008-10-02 Rigel Pharmaceuticals, Inc. Compositions and methods for inhibition of the jak pathway
US7834024B2 (en) * 2007-03-26 2010-11-16 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
US20120101114A1 (en) 2007-03-28 2012-04-26 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase
EP2152079A4 (en) * 2007-06-04 2011-03-09 Avila Therapeutics Inc Heterocyclic compounds and uses thereof
BRPI0814432A2 (en) * 2007-07-17 2017-05-09 Rigel Pharmaceuticals Inc cyclic amine substituted pyrimidinadiamines as pkc inhibitors
WO2009017838A2 (en) * 2007-08-01 2009-02-05 Exelixis, Inc. Combinations of jak-2 inhibitors and other agents
AU2008296479A1 (en) 2007-08-28 2009-03-12 Dana Farber Cancer Institute Amino substituted pyrimidine, pyrollopyridine and pyrazolopyrimidine derivatives useful as kinase inhibitors and in treating proliferative disorders and diseases associated with angiogenesis
WO2009029682A1 (en) 2007-08-28 2009-03-05 Rigel Pharmaceuticals, Inc. Combination therapy with syk kinase inhibitor
BRPI0815979A2 (en) 2007-08-28 2017-06-13 Irm Llc kinase inhibitor compounds and compositions, as well as their use
US7982036B2 (en) * 2007-10-19 2011-07-19 Avila Therapeutics, Inc. 4,6-disubstitued pyrimidines useful as kinase inhibitors
US7989465B2 (en) * 2007-10-19 2011-08-02 Avila Therapeutics, Inc. 4,6-disubstituted pyrimidines useful as kinase inhibitors
WO2009080638A2 (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
CA2717529A1 (en) 2008-03-11 2009-09-17 Cellzome Limited Sulfonamides as zap-70 inhibitors
US8205348B2 (en) * 2008-03-19 2012-06-26 Zashiki-Warashi Manufacturing Inc. Tile spacer and holder therefor
CL2009000600A1 (en) 2008-03-20 2010-05-07 Bayer Cropscience Ag Use of diaminopyrimidine compounds as phytosanitary agents; diaminopyrimidine compounds; its preparation procedure; agent that contains them; procedure for the preparation of said agent; and method for combating pests of animals and / or harmful plant pathogenic fungi.
DE102008018910A1 (en) * 2008-04-14 2009-12-31 Basf Coatings Ag Process for the preparation of color pigments containing effect pigments
WO2009127642A2 (en) 2008-04-15 2009-10-22 Cellzome Limited Use of lrrk2 inhibitors for neurodegenerative diseases
ES2597441T3 (en) 2008-04-16 2017-01-18 Portola Pharmaceuticals, Inc. 2,6-diamino-pyrimidin-5-yl-carboxamides as inhibitors of syk or JAK kinases
VN29259A1 (en) * 2008-04-16 2012-04-25 Portola Pharm Inc 2, 6-diamino- pyrimidin- 5-yl-carboxamides as syk or JAK kinases inhibitors
US8138339B2 (en) * 2008-04-16 2012-03-20 Portola Pharmaceuticals, Inc. Inhibitors of protein kinases
US8871753B2 (en) 2008-04-24 2014-10-28 Incyte Corporation Macrocyclic compounds and their use as kinase inhibitors
MX2010012703A (en) 2008-05-21 2010-12-21 Ariad Pharma Inc Phosphorous derivatives as kinase inhibitors.
US8338439B2 (en) 2008-06-27 2012-12-25 Celgene Avilomics Research, Inc. 2,4-disubstituted pyrimidines useful as kinase inhibitors
CA2727455C (en) 2008-06-27 2019-02-12 Avila Therapeutics, Inc. Heteroaryl compounds and uses thereof
WO2010009342A2 (en) 2008-07-16 2010-01-21 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase for the treatment of solid tumors
US20110245284A1 (en) 2008-09-03 2011-10-06 Bayer Cropscience Ag Alkoxy- and Alkylthio-Substituted Anilinopyrimidines
WO2010077740A2 (en) 2008-12-09 2010-07-08 Cytokine Pharmasciences, Inc. Novel antiviral compounds, compositions, and methods of use
SG172927A1 (en) 2009-01-15 2011-08-29 Hoffmann La Roche Antibodies against human epo receptor
US20120270237A9 (en) 2009-04-03 2012-10-25 Cellzone AG Methods for the identification of kinase interacting molecules and for the purification of kinase proteins
EP2440559B1 (en) * 2009-05-05 2018-01-10 Dana-Farber Cancer Institute, Inc. Egfr inhibitors and methods of treating disorders
EP3009428B1 (en) 2009-05-08 2018-02-21 Astellas Pharma Inc. Diamino heterocyclic carboxamide compound
CA2763720A1 (en) 2009-06-18 2010-12-23 Cellzome Limited Sulfonamides and sulfamides as zap-70 inhibitors
US7741330B1 (en) 2009-10-12 2010-06-22 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
US20130137709A1 (en) 2010-05-05 2013-05-30 Nathanael S. Gray Compounds that modulate EGFR activity and methods for treating or preventing conditions therewith
US20120071497A1 (en) 2010-06-03 2012-03-22 Pharmacyclics, Inc. Methods of treating abc-dlbcl using inhibitors of bruton's tyrosine kinase
NZ702485A (en) 2010-06-03 2016-04-29 Pharmacyclics Llc The use of inhibitors of bruton’s tyrosine kinase (btk)
WO2011153553A2 (en) 2010-06-04 2011-12-08 The Regents Of The University Of California Methods and compositions for kinase inhibition
MX336875B (en) 2010-08-10 2016-02-04 Celgene Avilomics Res Inc Besylate salt of a btk inhibitor.
WO2012061415A1 (en) 2010-11-01 2012-05-10 Portola Pharmaceuticals, Inc. Oxypyrimidines as syk modulators
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
ES2776002T3 (en) 2010-11-01 2020-07-28 Celgene Car Llc Heterocyclic compounds and their uses
ES2665013T3 (en) 2010-11-10 2018-04-24 Celgene Car Llc EGFR selective mutant 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
KR101884010B1 (en) 2011-05-04 2018-07-31 어리어드 파마슈티칼스, 인코포레이티드 Compounds for inhibiting cell proliferation in egfr-driven cancers
WO2012158843A2 (en) 2011-05-17 2012-11-22 The Regents Of The University Of California Kinase inhibitors
US20140303191A1 (en) 2011-10-19 2014-10-09 Pharmacyclics, Inc. 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
KR102081042B1 (en) 2012-03-15 2020-02-26 셀젠 카르 엘엘씨 Solid forms of an epidermal growth factor receptor kinase inhibitor
EP2914263A4 (en) 2012-11-02 2016-04-27 Pharmacyclics Inc Tec family kinase inhibitor adjuvant therapy
EP3027192A4 (en) 2013-08-02 2017-03-22 Pharmacyclics, LLC Methods for the treatment of solid tumors

Also Published As

Publication number Publication date
WO2011090760A1 (en) 2011-07-28
CN106146407A (en) 2016-11-23
NZ600631A (en) 2014-10-31
KR20170086547A (en) 2017-07-26
US20140213574A1 (en) 2014-07-31
US8338439B2 (en) 2012-12-25
EP2519235A4 (en) 2013-07-31
WO2011090760A4 (en) 2011-09-29
TWI592405B (en) 2017-07-21
EP2519235A1 (en) 2012-11-07
TW201124399A (en) 2011-07-16
CA2783624C (en) 2019-08-20
MX341825B (en) 2016-09-05
US10596172B2 (en) 2020-03-24
US8710222B2 (en) 2014-04-29
CN102740847A (en) 2012-10-17
AU2010343055A1 (en) 2012-06-21
RU2636584C2 (en) 2017-11-24
BR112012018345A2 (en) 2018-07-10
IL235621A0 (en) 2014-12-31
US20100249092A1 (en) 2010-09-30
MX2012007507A (en) 2012-08-01
KR101747159B1 (en) 2017-06-15
NZ700236A (en) 2016-06-24
KR20120118020A (en) 2012-10-25
US10010548B2 (en) 2018-07-03
US20190192512A1 (en) 2019-06-27
AU2017200697A1 (en) 2017-02-23
CN102740847B (en) 2016-06-29
US20170100397A1 (en) 2017-04-13
IL220155A0 (en) 2012-07-31
JP2013515786A (en) 2013-05-09
US20130065899A1 (en) 2013-03-14
AU2010343055B2 (en) 2016-11-10
EP2519235B1 (en) 2018-11-21
RU2012123546A (en) 2014-02-10
JP2016117749A (en) 2016-06-30
JP5879270B2 (en) 2016-03-08
US9409921B2 (en) 2016-08-09
IL220155B (en) 2019-05-30
SG181962A1 (en) 2012-08-30

Similar Documents

Publication Publication Date Title
AU2009262068B2 (en) Heteroaryl compounds and uses thereof
AU2010343055B2 (en) Heteroaryl compounds and uses thereof
AU2015202675B2 (en) Heteroaryl compounds and uses thereof
CA2759655A1 (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
EEER Examination request

Effective date: 20151119