CA2920996A1 - Heteroaryl compounds and uses thereof - Google Patents

Heteroaryl compounds and uses thereof Download PDF

Info

Publication number
CA2920996A1
CA2920996A1 CA2920996A CA2920996A CA2920996A1 CA 2920996 A1 CA2920996 A1 CA 2920996A1 CA 2920996 A CA2920996 A CA 2920996A CA 2920996 A CA2920996 A CA 2920996A CA 2920996 A1 CA2920996 A1 CA 2920996A1
Authority
CA
Canada
Prior art keywords
ring
halogen
compound according
oxo
substituted
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA2920996A
Other languages
French (fr)
Inventor
Richland W. Tester
Juswinder Singh
Shomir Ghosh
Arthur F. Kluge
Russell C. Petter
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 Avilomics Research Inc
Original Assignee
Celgene Avilomics Research 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 Celgene Avilomics Research Inc filed Critical Celgene Avilomics Research Inc
Publication of CA2920996A1 publication Critical patent/CA2920996A1/en
Abandoned legal-status Critical Current

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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • C07B59/002Heterocyclic compounds
    • 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
    • C07D401/12Heterocyclic 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
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • 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
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/10Protein-tyrosine kinases (2.7.10)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/05Isotopically modified compounds, e.g. labelled

Abstract

The present invention provides compounds, pharmaceutically acceptable compositions thereof, and methods of using the same.

Description

HETEROARYL COKPOUNDS AND USES THEREOF
[0001]
TECHNICAL FIELD OF THE INVENTION
100021 The present invention relates to compounds useful as inhibitors of protein kinases.
The invention also provides pharmaceutically acceptable campositions comprising compounds of the present invention and methods of using said compositions in the treatment of various disorders.
BACKGROUND OF THE INVENTION
100031 The search for new therapeutic agents has been greatly aided in recent years by a better understanding of the structure of enzymes and other biornolecules associated with diseases. One important class of enzymes that has been the subject of extensive study is protein kinases.
100041 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.).
j00051 = 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., interleulcin-1 (IL-I) and tumor necrosis factor a (TNF-a)), and growth factors (e.g., granulocyte macrophage-colony-stimulating factor
2 (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.
[0006] 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
[0007] 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 the general formula I:

A13) (IR%
or a pharmaceutically acceptable salt thereof, wherein Ring A, Ring B, m, Rx, RY, W, and RI are as defined herein.
[0008] 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.
[0009] 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 lcinase inhibitors.
3 BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 depicts the EGF inhibiting activity of compound I-1.
Figure 2 depicts the results of compound I-1 in a "washout" experiment as compared with compound 1-93.
Figure 3 depicts dose response inhibition of EGFR phosphorylation and p42/p44 Erk phosphorylation with compounds 1-16 and 1-17 in A431 cells.
Figure 4 depicts dose response inhibition of EGFR phosphorylation and p42/p44 Erk phosphorylation with compound 1-19 in A431 cells.
Figure 5 depicts dose response inhibition of EGFR phosphorylation with compound 1-1 in A431 cells as compared with its "reversible control" compound (IR-3).
Figure 6 depicts the results of compound I-1 in a "washout" experiment as compared with its "reversible control" compound (IR-3).
Figure 7 depicts MS analysis confirming covalent modification of ErbB4 by compound I-1.
Figure 8 depicts MS analysis confirming covalent modification of ErbB1 at Cys797 by compound I-1.
Figure 9 depicts the inhibition of BTK signaling in Ramos cells by compound 1-13.
Figure 10 depicts the results of compound 1-13 in a "washout" experiment with BTK in Ramos cells.
Figure 11 depicts MS analysis of the tryptic digests confirming covalent modification of TEC
kinase by compound 1-13.
Figure 12 depicts MS analysis confirming covalent modification of BTK by compound 1-63.
Figure 13 depicts MS analysis confirming covalent modification of BTK by compound 1-66.
Figure 14 depicts an amino acid sequence for BTK (SEQ ID 1).
Figure 15 depicts an amino acid sequence for TEC (SEQ ID 2).
Figure 16 depicts an amino acid sequence for ITK (SEQ ID 3).
Figure 17 depicts an amino acid sequence for BMX (SEQ ID 4).
Figure 18 depicts an amino acid sequence for JAK3 (SEQ ID 5).
Figure 19 depicts an amino acid sequence for TXK (SEQ ID 6).
4 DETAILED DESCRIPTION OF CERTAIN EMBODIMENTS
1. General Description of Compounds of the Invention 100101 In certain embodiments, the present invention provides a compound of formula I:
R

NN
(Rx)m or a pharmaceutically acceptable salt thereof, wherein:
Ring A is an optionally substituted group selected from phenyl, an 8-10 membered bicyclic partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 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 phenyl, a 5-6 membered heteroaryl ring having 1-3 heteratoms independently selected from N, 0 or S, a 5-6 membered saturated heterocyclic ring having 1-2 heteroatoms independently selected from N, 0 or S, or an 8-10 membered bicyclic partially unsaturated or aryl ring having 1-3 heteroatoms independently selected from N, 0 or S;
RI is a warhead group;
RY is hydrogen, halogen, CN, lower alkyl, or lower haloallcyl;
W is a bivalent Ci_3 alkylene chain wherein one methylene unit of W is optionally replaced by ¨
NR2-, -N(R2)C(0)-, -C(0)N(R2)-, -N(R2)S02-, -SO2N(R2)-, -0-, -C(0)-, -0C(0)-, -C(0)0-, -S-, -SO- or -SO2-;
R2 is hydrogen or optionally substituted C1_6 aliphatic, or:
R2 and a substituent on Ring A are taken together with their intervening atoms to form a 4-6 membered saturated ring, or:
R2 and RY are taken together with their intervening atoms to form a 4-7 membered carbocyclic ring;
m is 0-4;

each le is independently selected from -R, halogen, ¨OR, ¨CN, ¨NO2, -SO2R, -SOR, -C(0)R, -CO2R, ¨C(0)N(R)2, -NRC(0)R, -N1.C(0)NR2, -NRSO2R, or ¨N(R)2; or:
Rx and RI 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; and 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.
[0011] In certain embodiments, the present invention provides a compound of formula N ).µA RY
(Rx)m or a pharmaceutically acceptable salt thereof, wherein:
Ring A is an optionally substituted group selected from phenyl, an 8-10 membered bicyclic partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 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;
RI is a warhead group;
RY is hydrogen, halogen, CN, lower alkyl, or lower haloallcyl;
G is CH, or N;
W is -NR2-, -S-, or -0-;
R2 is hydrogen or optionally substituted C1.6 aliphatic, or:
R2 and a substituent on Ring A are taken together with their intervening atoms to form a 4-6 membered saturated ring;

m is 0-4;
each le is independently selected from -R, halogen, -OR, -CN, -NO2, -SO2R, -SOR, -C(0)R, -CO2R, -C(0)N(R)2, -NRC(0)R, -NRC(0)NR2, -NRSO2R, or -N(R)2; or:
le and R1 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; and each R group is independently hydrogen or an optionally substituted group selected froth 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.
2. Compounds and Definitions 100121 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, 75e 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.
100131 The term "aliphatic" or "aliphatic group", as used herein, means a straight-chain (i.e., unbrancbed) or branched, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of tmsaturation, or a monocyclie hydrocarbon or bicyclic hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic (also referred to herein as "carbocycle" "cycloaliphatic"
or "cycloalkyl"), that has a single point of attachment to the rest of the molecule. Unless otherwise specified, aliphatic groups contain 1-6 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1-5 aliphatic carbon atoms. In other embodiments, aliphatic groups contain 1-4 aliphatic carbon atoms. In still other embodiments, aliphatic groups contain 1-3 aliphatic carbon atoms, and in yet other embodiments, aliphatic groups contain 1-2 aliphatic carbon atoms. In some embodiments, "cycloaliphatic" (or "carbocycle" or "cycloalkyl") refers to a monocyclic C3-C6 hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule. Suitable aliphatic groups include, but are not limited to, linear or branched, substituted or unsubstituted alkyl, alkenyl, allcynyl groups and hybrids thereof such as (cycloalkypalkyl, (cycloalkenypalkyl or (cycloalkyl)alkenyl.
[0014] The term "lower alkyl" refers to a C1_4 straight or branched alkyl group. Exemplary lower alkyl groups are methyl, ethyl, prOpyl, isopropyl, butyl, isobutyl, and tert-butyl.
[0015] The term "lower haloallcyl" refers to a Ci_4 straight or branched alkyl group that is substituted with one or more halogen atoms.
100161 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 quatemized form of any basic nitrogen or; a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4-dihydro-2H-pyn-oly1), NH (as in pyrrolidinyl) or NR+ (as in N-substituted pyrrolidinyl)).
[0017] The term "unsaturated", as used herein, means that a moiety has one or more units of unsaturation.
[0018] As used herein, the term "bivalent C1_8 (or C14 saturated or unsaturated, straight or branched, hydrocarbon chain", refers to bivalent alkylene, alkenylene, and alkynylene chains that are straight or branched as defined herein.
[0019] The term "alkylene" refers to a bivalent alkyl group. An "alkylene chain" is a polymethylene group, i.e., ¨(CH,)¨, 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.
[0020] 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.

100211 As used herein, the term "cyclopropylenyl" refers to a bivalent cyclopropyl group of the following structure: 7 .
[0022] The term "halogen" means F, Cl, Br, or I.
[0023] The term "aryl" used alone or as part of a larger moiety as in "arallcyl", "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.
100241 The terms "heteroaryl" and "beteroar¨", 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 it 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 quatemized 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, benzotlaienyl, benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzthiazolyl, quinolyl, isoquinolyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4H¨quinolizinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, and pyrido[2,3¨b]-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 "heteroarallcyl" refers to an alkyl group substituted by a heteroaryl, wherein the alkyl and beteroaryl portions independently are optionally substituted.
[0025] 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¨pyrroly1), NH (as in pyrrolidinyl), or +NR (as in N¨substituted pyrrolidinyl).
[0026] 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 "heterocyclylallcyl" refers to an alkyl group substituted by a heterocyclyl, wherein the alkyl and heterocyclyl portions independently are optionally substituted.
[0027] As used herein, the term "partially unsaturated" refers to a ring moiety that includes at least one double or triple bond. The term "partially unsaturated" is intended to encompass rings having multiple sites of unsaturation, but is not intended to include aryl or heteroaryl moieties, as herein defined.
[0028] 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.
[0029] Suitable monovalent substituents on a substitutable carbon atom of an "optionally substituted" group are independently halogen; -(CH2)0_41t ; -(CH2)0_40R ; -0(CH2)04R , -0-(CH2)0_4C(0)0R ; -(CH2)o-4CH(OR )2; -(CH2)o-4SIV; -(CH2)o-4Ph, which may be substituted with R ; -(CH2)o--40(CH2)0_1Ph which may be substituted with R ; -CH=CHPh, which may be substituted with R ; -(CH2)0_40(CH2)o-i-pyridyl which may be substituted with R ; -NO2; -CN;
-N3; -(CH2)o--4N(R )2; -(CH2)o-4N(R )C(0)R ; -N(R )C(S)R ; -(CH2)0_41\1(R
)C(0)NR 2;
-N(R )C(S)NR 2; -(CH2)0_4N(R )C(0)0R ; -N(R )N(R )C(0)R ; -N(R )N(R )C(0)NR 2;
-N(R )N(R )C(0)0R ; -(CH2)o--4C(0)R ; -C(S)R ; -(CH2)o-4C(0)0R ; -(CH2)0_4C(0)SR ;
-(CH2)0_4(0)0SiR 3; -(CH2)o--40C(0)R ; -0C(0)(CH2)0_4SR-, SC(S)SR ; -(CH2)0_4SC(0)R ;
-(CH2)0-4C(0)NR 2; -C(S)NR 2; -C(S)SR ; -SC(S)SR , -(CH2)0_40C(0)NR 2;
-C(0)N(OR )R ; -C(0)C(0)R ; -C(0)CH2C(0)R ; -C(NOR )R ; -(CH2)0-4SSW; -(CH2)0-4S(0)21V; -(CH2)0_4(0)20R ; -(CH2)0_40S(0)2R ; -S(0)2NR 2; -(CH2)0_4S(0)R ;
-N(R )S(0)2NR 2; -N(R )S(0)2R ; -N(OR )R ; -C(NH)NR 2; -P(0)2R ; -P(0)R 2; -0P(0)R 2;
-0P(0)(OR )2; SiR 3; -(C1-4 straight or branched allcylene)O-N(R )2; or -(C1.4 straight or branched alkylene)C(0)0-N(R )2, wherein each R may be substituted as defined below and is independently hydrogen, C1_6 aliphatic, -CH2Ph, -0(CH2)0_1Ph, -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.
[0030] 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., -(halon, -(CH2)0_20H, --(CH2)o-20R., -(CH2)0_2CH(0R.)2;
-0(haloR'), -CN, -N3, -(CH2)0-2C(0)1e, -(CH2)0-2C(0)0H, -(CH2)0_2C(0)01e, -(CH2)0-2SR", -(C1-12)0_2SH, -(CH2)0_2NI-12, -(CH2)0_2NHR", -(CH2)0_2NR"2, -NO2, -SiR."3, -0Si12..3, -C(0)SR', -(C1_4 straight or branched allcylene)C(0)01r, or -SSR" wherein each 11" is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently selected from C1_4 aliphatic, -CH2Ph, -0(CH2)0_1113, 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.
[0031] Suitable divalent substituents on a saturated carbon atom of an "optionally substituted" group include the following: =0, =S, =NNR*2, =NNHC(0)R*, =NNHC(0)01e, =NNHS(0)2R*, =NR*, =NOR*, -0(C(R.2))2-30-, or -S(C(R*2))2-3S-, wherein each independent occurrence of R* is selected from hydrogen, C1_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: -0(CR*2)2_30-, wherein each independent occurrence of R* is selected from hydrogen, C1 _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.
[0032] Suitable substituents on the aliphatic group of R.* include halogen, -R', -(haloR'), -OH, -OR', -0(halon, -CN, -C(0)0H, -C(0)0R', -NH2, -NHR", -N11.2, or -NO2, wherein each le is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently C1_4 aliphatic, -CH2Ph, -0(CH2)0_11Th, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.

[0033] Suitable substituents on a substitutable nitrogen of an "optionally substituted" group include -12.1., -C(0)Rt, -C(0)0Rt, -C(0)C(0)Rt, -C(0)CH2C(0)R1., -S(0)21e, -S(0)2NRI2, -C(S)NRt2, -C(NH)NRt2, or -N(11)S(0)2Rt; wherein each Rt is independently hydrogen, C1_6 aliphatic which may be substituted as defined below, unsubstituted -0Ph, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 beteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of RI, 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 beteroatoms independently selected from nitrogen, oxygen, or sulfur.
[0034] _Suitable substituents on the aliphatic group of le are independently halogen, -V', -(halon, -0H, -OR', -0(halon, -CN, --C(0)0H, -C(0)0R., -NH2, -NHR', -N11,2, or -NO2, wherein each R. is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently C1-4 aliphatic, -CH2Pb, -0(CH2)0-113h, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[0035] 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.
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, hydrobrornic 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, glucobeptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2¨hydroxy¨ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2¨naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, pahnitate, pamoate, pectinate, persulfate, 3¨phenylpropionate, phosphate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p¨toluenesulfonate, undecanoate, valerate salts, and the like.
[0036] Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N (C1_aalky1)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.
[0037] Unless otherwise stated, structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention.
Additionally, unless otherwise stated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures including the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope of this invention.
Such compounds are useful, for example, as analytical tools, as probes in biological assays, or as therapeutic agents in accordance with the present invention. In some embodiments, the R1 group of formula I comprises one or more deuterium atoms. Such compounds include compound 1-61 as depicted in Table 5.
[0038] 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 lcinase 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 lcinase 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.
[0039] 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.
[0040] 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 less than about 1 M, less than about 500 nM, less than about 100 nM, or less than about 10 nM.
[0041] A
compound of the present invention may be tethered to a detectable moiety. One of ordinary skill in the art will recognize that a detectable moiety may be attached to a provided compound via a suitable substituent on Ring A or a suitable It.' 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 (i.e. directly attached to Ring A or the aniline ring) or via a tethering group, such as a bivalent saturated or unsaturated hydrocarbon chain.

100421 As used herein, the term "detectable moiety" is used interchangeably with the term "label" and relates to any moiety capable of being detected, e.g., primary labels and secondary labels. Primary labels, such as radioisotopes (e.g., tritium, 32P, 33P, 35S, or "C), mass-tags, and fluorescent labels are signal generating reporter groups which can be detected without further modifications.
00431 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.
[00441 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:
AlexTFluor dyes (AlexirFluor 350, AlexTamFluor 488, AlexTamFluor 532, AlexTFluor 546, Alexr Fluor 568, AlexTamFluor 594, AlexFluor 633, AleZa-mFluor 660 and AlePaAFluor 680), AMCA, AMCA-S, BODIPY dyes (BODIPY FL, BODIPY R6G, BODIPY TMR, BODIPY TR, 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, Dialkylarninocournarin, 4',5'-Dichloro-2',7'-dimethoxy-fluorescein, DM-NERF, Eosin, Erythrosin, Fluorescein, FAM, Hydroxyeoumarin, IRDyes (IRD40, 1RD 700, 1RD
800), JOE, Lissamine rhodamine B, Marina Blue, Methoxycoumarin, Naphthofluoreseein, 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, TetrametItyl-rhodaminc (TMR), Carboxytetrarnethylrhiodamine (TAMRA), Texas Red, Texas Red-X.
100451 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-[344'-[(p-Methoxytetrafluorobenzypoxy]phenyl] -3 -methylglyc eronyl] i sonipecotic Acid, 4' Tetafluoro-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.
[0046] The terms "measurable affinity" and "measurably inhibit," as used herein, means a measurable change in at least one of ErbB I, 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 ErbBI, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3, in the absence of said compound, or composition thereof 3. Description of Exemplary Compounds [0047] According to one aspect, the present invention provides a compound of formula I, N

RY
(IV), or a pharmaceutically acceptable salt thereof, wherein:
Ring A is an optionally substituted group selected from phenyl, an 8-10 membered bicyclic partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 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 phenyl, a 5-6 membered heteroaryl ring having 1-3 heteratoms independently selected from N, 0 or S, a 5-6 membered saturated heterocyclic ring having 1-2 heteroatoms independently selected from N, 0 or S, or an 8-10 membered bicyclic partially unsaturated or aryl ring having 1-3 heteroatoms independently selected from N, 0 or S;
RI 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(0)-, -C(0)N(R)-, -N(R)S02-, -SO2N(R)-, -0-, -C(0)-, -0C(0)-, -C(0)0-, -S-, -SO-, -SO2-, -C(=S)-, -C(=NR)-, -N=N-, or 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 1-4 Re groups; and each Re is independently selected from ¨Q-Z, oxo, NO2, halogen, CN, a suitable leaving group, or a 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)-, -0C(0)-, -C(0)0-, -SO-, or -SO2-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S02-, or ¨SO2N(R)-; and Z is hydrogen or C1-6 aliphatic optionally substituted with oxo, halogen, NO2, or CN;
RY is hydrogen, halogen, CN, lower alkyl, or lower haloalkyl;
W is a bivalent C1_3 allcylene chain wherein one methylene .unit of W is optionally replaced by ¨
NR2-, -N(R2)C(0)-, -C(0)N(R2)-, -N(R2)S02-, -SO2N(R2)-, -0-, -C(0)-, -0C(0)-, -C(0)0-, -S-, -SO- or -SO2-;
R2 is hydrogen or optionally substituted Ci_6 aliphatic, or:
R2 and a substituent on Ring A are taken together with their intervening atoms to form a 4-6 membered saturated ring, or:
R2 and RY are taken together with their intervening atoms to form a 4-7 membered carbocyclic ring;
m is 0-4;

each Rx is independently selected from -R, halogen, ¨OR, ¨CN, ¨NO2, -SO2R, -SOR, -C(0)R, -CO2R, ¨C(0)N(R)2, -NRC(0)R, -NRC(0)NR2, -NRSO2R, or ¨N(R)2; or:
R" and RI 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; and 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.
[0048] In certain embodiments, the present invention provides a compound of formula H:

W

i , )) N N
H (Rx), II =
or a pharmaceutically acceptable salt thereof, wherein:
Ring A is an optionally substituted group selected from phenyl, an 8-10 membered bicyclic partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 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;
RI 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(0)-, -C(0)N(R)-, -N(R)S02-, -SO2N(R)-, -0-, -C(0)-, -0C(0)-, -C(0)0-, -S-, -SO-, -SO2-, -C(=S)-, -C(=NR)-, -N=N-, or -C(=N2)-;

Y is hydrogen, C .,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 C1 _6 aliphatic optionally substituted with oxo, halogen, NO2, or CN, wherein:
Q is a covalent bond or a bivalent C .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)-, -0C(0)-, -C(0)0-, -SO-, or -SO2-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S02-, or ¨SO2N(R)-; and Z is hydrogen or C1_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN;
RY is hydrogen, halogen, CN, lower alkyl, or lower haloaLkyl;
G is CH, or N;
W is -NR2-, -S-, or -0-;
R2 is hydrogen or optionally substituted C.6 aliphatic, or:
R2 and a substituent on Ring A are taken together with their intervening atoms to form a 4-6 membered saturated ring;
m is 0-4;
each Rx is independently selected from -R, halogen, ¨OR, ¨CN, ¨NO2, -SO2R, -SOR, -C(0)R, -CO2R, ¨C(0)N(R)2, -NRC(0)R, -NRC(0)NR2, -NRSO2R, or ¨N(R)2; or:
fe and RI are taken together with their intervening atoms to form a 5-7 membered saturated, partially unsaturated, or aryl ring haying 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 aliphatic; and 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.
100491 According to one aspect, the present invention provides a compound of formula II-a or 11-1):

5): R1 Ri NRY
N N N N
(Rx)m (Rx), II-a or a pharmaceutically acceptable salt thereof wherein each of Ring A, W, RI, G, RY, le and m are as defined above for formula II and as described herein.
[0050] In certain embodiments, the present invention provides a compound of formula II-b where in said compound is other than N6-m-tolyl-N4-p-tolylpyrimidine-4,6-diamine.
[0051] In certain embodiments, the present invention provides a compound of formula II-a wherein said compound is other than N4-(3-aminopheny1)-N6-(3-bromophenyppyrimidine-4,6-diamine, N-(3-(6-(3-(trifluoromethyl)phenylamino)pyrimidin-4-ylamino)phenyl)cyclopropane-carboxamide, N-(3-(6-(3-bromophenylamino)pyrimidin-4-ylamino)phenyl)propionamide, N4-(3-aminopheny1)-N6-m-tolylpyrimidine-4,6-diamine, or N4-(3-aminopheny1)-N6-methyl-N6-phenyl-pyrimidine-4,6-diamine.
[0052] As defined generally above, the Ring A group of formulae I and II is an optionally substituted group selected from phenyl, an 8-10 membered bicyclic partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 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 heteraryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some embodiments, Ring A is an optionally substituted diphenyl ether. In some embodiments, Ring A is an optionally substituted phenyl benzyl ether. In other embodiments, Ring A is an optionally substituted pyridine methoxy phenyl group.
[0053] In certain embodiments, the Ring A group of formulae I and II 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 -0(CH2)04R , wherein each R is as defined herein. Exemplary substituents on Ring A include Br, I, Cl, methyl, -CF3, -CE---CH, -OCH2phenyl, -OCH2(fluorophenyl), or -OCH2pyridyl.
100541 Exemplary Ring A groups of formulae I and ll are set forth in Table 1.
Table 1. Exemplary Ring A Groups /
Br CI-43 OCH3 Oil rS_ ro ' 411 µS
i ii iii iv =%.'"...) Oil (N' F CI CF3 0, 0lel F
,-(S-r-, ' 4111eSS' V Vi Vii Viii i N 1,4 µ =
y * 0 I

Y.: CH3 = gib = = 0 "1111,- 14I ,-5:
, 0111 ,r_ ix x F xi xii I.
0 0 0 0 Br IA F
/
N
N E. --SS' CI eSS:' 1414Pc-53:
Xiii XiV XV XVi XVii 0 0 . F, CI, CN op S . F, a CN
Y PPrs Y J-Prr xviii xix S F, CI, CN 0 * 40 C) . *

Y Sr XX XXi XXii C) () S' -XXiii XXiV XXV

F __ , a-o..s.,,,.,,,, "I_s xxvi xxvii Aa-vai Me0 0 NC 0 s K% si xxix xxx o I
N I R--, I
õisss C.,,,,,/ N
XXXi XXXii =CM

PAN * 0 I I
0. ss55 N sssS ' \ N-i7 Si XXXiv rx-xv xxxvi 0 0 .-,=.,,,C) * _.,---,,,.(3 0 I I I
SI si N , Si xxxvii zaa-viii xxxix ,7,N,õ,õ0 0 o ....----ry (7---.''' 1 1 N'..--') NN S 0-,4 Or,i3 S-' 0,,,, xliii xliv o 0 R----Cro . R-----Cr * R----Cr 0 HN--N
S

5535 .55IS SCSS
X/V X/Vi X/Vii R-------,,, 0 = 0 \ /
R --Cr R'N'N 51 = s/
0 Na , xlviii xlix 1 --,i-,, o I
rt\I
r------,N

=
0.,..,_ = 0 CI V
/i hi 0liii liv H H
(0 F3C O N y N 10 L. 0 CI
iv ivi [0055] As defined generally above, the Ring B group of formula I is phenyl, a 5-6 membered heteroaryl ring having 1-3 heteratoms independently selected from N, 0 or S, a 5-6 membered saturated heterocyclic ring having 1-2 heteroatoms independently selected from N, 0 or S, or an 8-10 membered bicyclic partially unsaturated or aryl ring having 1-3 heteroatoms independently selected from N, 0 or S.
[0056] In some embodiments, the Ring B group of formula I is phenyl. In some embodiments, Ring B is a 6-membered heteroaryl ring having 1-3 nitrogens. In some embodiments, Ring B is a 5-membered heteroaryl ring having 1 or 2 or 3 heteroatoms independently selected from N, 0 or S.
[0057] In some embodiments, the Ring B group of formula I 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.
[0058] Exemplary Ring B groups are set forth in Table 2.
Table 2. Ring B Groups al al al al al RI
N).-.= NN\ op \N 0 6 V------ N\
Rx it' Rx Rx Rx Rx i ii iii iv v vi RI
\ri / RI RI
RI RI .
I /
el III -)...'-' N .-----.1 _....-N
I I
.2Z2N
Rx Rx R' vii viii ix x xi xii , Ri \
Rx Rx xiii xiv [0059] In some embodiments, the m moiety of formula I, 11, Ha, or II13 is 1, 2, 3 or 4. In some embodiments, m is 1. In other embodiments, m is 0.
[0060] As defined generally above, each Rx group of formula I or II is independently selected from -R, halogen, ¨OR, ¨CN, ¨NO2, -SO2R, -SOR, -C(0)R, -CO2R, -C(0)N(R)2, -NRC(0)R, -NRC(0)NR2, -NRSO2R, or ¨N(R)2, or le and RI 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, wherein the warhead group is -Q-Z, and said ring is further substituted with 0-3 groups independently selected from oxo, halogen, CN, or C1_6 aliphatic.
[0061] In some embodiments, each instance of It.' is independently selected from ¨R, -OR or halogen. In certain embodiments, Rx is lower alkyl, lower alkoxy, or halogen.
Examplary Rx groups include methyl, methoxy, and chloro. In some embodiments, Rx is hydrogen.
[0062] In some embodiments, the G group of any of formula H, II-a, or 11-b is CH. In other embodiments, the G group of any of formula II, H-a, or H-b is N.
[0063] As defined generally above, the W group of formula I is a bivalent C1.3 alkylene chain wherein one methylene unit of W is optionally replaced by ¨NR2-, -N(R2)C(0)-, -C(0)N(R2)-, -N(R2)S02-, -SO2N(R2)-, -0-, -C(0)-, -0C(0)-, -C(0)0-, -S-, -SO-or -SO2-.
[0064] In certain embodiments, the W group of formula I is ¨NH-, -S-, or ¨0¨. In some embodiments, the W group of formula I is -CH20¨, ¨CH2S¨, or ¨CH2NH¨. In some aspects, W
is ¨OCH2¨, ¨SCH2¨, ¨NHC12¨, or -CH2CH2¨=
[0065] In some embodiments, the W group of formula I is ¨0- thus forming a compound of formula I-i:

N

N.011 = (Rx),õ
1-i or a pharmaceutically acceptable salt thereof, wherein each of Ring A, RI, le, RY, and m are as defined above and described in classes and subclasses above and herein.
100661 In some embodiments, W is ¨NR2- thus forming a compound of formula I-i:
N ¨ R2 RY R
N
= (Rx), or a pharmaceutically acceptable salt thereof, wherein each of Ring A, RI, R2, le, RY, and m are as defined above and described in classes and subclasses above and herein.
[00671 In some embodiments, W is ¨S- thus forming a compound of formula I-iii:
CO

N
N N
= (Rx),õ
or a pharmaceutically acceptable salt thereof, wherein each of Ring A, RI, le, RY, and m are as defined above and described in classes and subclasses above and herein.
[0068] In some embodiments, the W group of formula II is ¨0- thus forming a compound of formula II-i:

N RY G/
k )j N N
H (Rx)m or a pharmaceutically acceptable salt thereof, wherein each of Ring A, RI, Rx, RY, and m are as defined above and described in classes and subclasses above and herein.
100691 In some embodiments, the W group of formula II is ¨NR2- thus forming a compound of formula II-ii:

N¨R2 N ( j RI' G/R1 k, ) N N
II-ii or a pharmaceutically acceptable salt thereof, wherein each of Ring A, RI, R2, re, RY, and m are as defined above and described in classes and subclasses above and herein.
100701 In certain embodiments, R2 is hydrogen. In some embodiments, R2 is methyl. In still other embodiments, R2 is lower alkyl.
100711 In some embodiments, the W group of formula II is ¨S- thus forming a compound of formula II-iii:

S

N N
H-iii or a pharmaceutically acceptable salt thereof, wherein each of Ring A, RI, Rx, RY, and m are as defined above and described in classes and subclasses above and herein.

100721 In certain embodiments, the G group of formula II is CH, thus forming a compound of formula III:

N)I3Y
N N
(Rx), HI
or a pharmaceutically acceptable salt thereof, wherein each of Ring A, W, R, Rx, RY, and m are as defined above and described in classes and subclasses above and herein.
[0073] In certain embodiments, the compound of formula HI is of formula HI-a-i, HI-b-i, I11-a-ii, IH-b-ii, HI-a-iii, or III-b-iii:
A A

N RY N): R 1Y
EL.
N N(Rx)m N N \-"(Rix)m HI-a-i HI-b-i N )(RYfo N RY
N N
(Rx) N (Rx), HI-a-ii = A

N )):S RY
N )S RY R1 , N N N
(Rx)õ
or a pharmaceutically acceptable salt thereof, wherein each of Ring A, RI, R2, Rx, RY, and m are as defined above and described in classes and subclasses above and herein.
[0074] In certain embodiments, the G group of formula II is N, thus forming a compound of formula IV:

N) RY N
õ
N N
(Rx)n, IV
or a pharmaceutically acceptable salt thereof, wherein each of Ring A, W, RI, Rx, RY, and m are as defined above and described in classes and subclasses above and herein.
[0075] In certain embodiments, the compound of formula IV is of formula FV-a-i, IV-b-i, IV-a-ii, IV-b-ii, IV -a-iii, or IV -b-iii:

RY

N N N N
IV-a-i IV-b-i N¨R2 R1 N¨R2 N.J.-TRY
LNNN N
(Rx)õ, (Rx)õ, IV-a-ii IV-b-ii R1 .
Y ,R1 N R
'P.1) N N"
it, (Rx),, (Rx),, IV -a-iii IV -b-iii or a pharmaceutically acceptable salt thereof, wherein each of Ring A, RI, R2, le, RY, and m are as defined above and described in classes and subclasses above and herein.
100761 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 II-a-iv or 11-b-iv:
0 )1-3 ) Y

LNN)\X
11, µ0Rx)m N N (Rx)m H-a-iv H-b-iv or a pharmaceutically acceptable salt thereof, wherein each of Ring A, RI, le, and m are as defined above and described in classes and subclasses above and herein.
[0077] In certain embodiments, the present invention provides a compound of formula 11-a-v or H-b-v wherein Ring A is phenyl and said compound is of formula H-a-v or N):RY
N N N N
(Rx)m (Rx)m H-a-v H-b-v or a pharmaceutically acceptable salt thereof, wherein the Ring A phenyl moiety is optionally substituted and each of RI, Rx, RY, and m are as defined above and described in classes and subclasses above and herein. .
100781 In some embodiments, R2 is hydrogen. In other embodiments, R2 and RY
are taken together thereby forming a compound of formula II-a-vi or H-b-vi:
N N
)1-3 )1-3 R1 N N N N
(Rx) (Rx)m II-a-vi H-b-vi [0079] As defined generally above, the RI group of formulae I and H 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(0)-, -C(0)N(R)-, -N(R)S02-, -SO2N(R)-, -0-, -C(0)-, -0C(0)-, -C(0)0-, -S-, -SO-, -SO2-, -C(=S)-, -C(=NR)-, -N=N-, or 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 1-4 Re groups; and each Re is independently selected from ¨Q-Z, oxo, NO2, halogen, CN, a suitable leaving group, or a 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)-, -0C(0)-, -C(0)0-, -SO-, or -SO2-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S02-, or -SO2N(R)-; and Z is hydrogen or C1_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN.
[0080] In certain embodiments, L is a covalent bond.
[0081] In certain embodiments, L is a bivalent C1_8 saturated or unsaturated, straight or branched, hydrocarbon chain. In certain embodiments, L is -CH2-.
100821 In certain embodiments, L is a covalent bond, -CH2-, -NH-, -CH2NH-, -NHCH2-, -NHC(0)CH20C(0)-, -CH2NHC(0)-, -NI-IS02-, -NHSO2CH2-, -NHC(0)CH20C(0)-, or -SONH-.
[0083] In some embodiments, L is a bivalent C2-8 straight or branched, hydrocarbon chain wherein L has at least one double bond and one or two additional methylene units of L are optionally and independently replaced by -NRC(0)-, -C(0)NR-, -N(R)S02-, -SO2N(R)-, -S-, -S(0)-, -SO2-, -0C(0)-, -C(0)0-, cyclopropylene, -0-, -N(R)-, or -C(0)-.
[0084] 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(0)-, -C(0)NR-, -N(R)S02-, -SO2N(R)-, -S-, -S(0)-, -SO2-, -0C(0)-, 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)-.
[0085] In some embodiments, L is a bivalent C2-8 straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by -C(0)-, and one or two additional methylene units of L are optionally and independently replaced by cyclopropylene, -0-, -N(R)-, or -C(0)-.
(0086] As described above, in certain embodiments, L is a bivalent C2_8 straight or branched, hydrocarbon chain wherein L has at least one double bond. One of ordinary skill in the art will recognize that such a double bond may exist within the hydrocarbon chain backbone or may be "exo" to the backbone chain and thus forming an allcylidene group. By way of example, such an L group having an allcylidene 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(0)C(=CH2)CH2-.

100871 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(0)CH=CH(CH3)-, -C(0)CH=CHCH2NH(CH3)-, -C(0)CH=CH(CH3)-, -C(0)CH=CH-, -CH2C(0)CH=CH-, -CH2C(0)CH=CH(CH3)-, -CH2CH2C(0)CH=CH-, -CH2CH2C(0)CH=CHCH2-, -CH2CH2C(0)CH=CHCH2NH(CH3)-, or -CH2CH2C(0)CH=CH(CH3)-, or -CH(CH3)0C(0)CH=CH-.
[0088] 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 -0C(0)-.
[0089] 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(0)-, -C(0)NR-, -N(R)S02-, -SO2N(R)-, -S-, -S(0)-, -SO2-, -0C(0)-, 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)-. In some embodiments, L is -CH20C(0)CH=CHCH2-, -CH2-0C(0)CH=CH-, or -CH(CH=CH2)0C(0)CH=CH-.
[0090] In certain embodiments, L is -NRC(0)CH=CH-, -NRC(0)CH=CHCH2N(CH3)-, -NRC(0)CH=CHCH20-, -CH2NRC(0)CH=CH-, -NRSO2CH=CH-, -NRSO2CH=CHCH2-, -NRC(0)(C=N2)C(0)-, -NRC(0)CH=CHCH2N(CH3)-, -NRSO2CH=CH-, -NRSO2CH=CHCH2-, -NRC(0)CH=CHCH20-, -N1?.C(0)C(=CH2)CH2-, -CH2NRC(0)-, -CH2NRC(0)CH=CH-, -CH2CH2NRC(0)-, or -CH2NRC(0)cyclopropylene-, wherein each R is independently hydrogen or optionally substituted C1_6 aliphatic.
[0091] In certain embodiments, L is -NHC(0)CH=CH-, -NHC(0)CH=CHCH2N(CH3)-, -NHC(0)CH=CHCH20-, -CH2NHC(0)CH=CH-, -NHSO2CH=CH-, -NI-ISO2CH=CHCH2-, -NHC(0)(C=N2)C(0)-, -NHC(0)CH-CHCH2N(CH3)-, -NHSO2CH=CH-, -NHSO2CH=CHCH2-, -NHC(0)CH=CHCH20-, -NIC(0)C(=CH2)CH2-, -CH2NHC(0)-, -CH2NHC(0)CH=CH-, -CH2CH2NHC(0)-, or -CH2NHC(0)cyclopropylene-.
[0092] 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(0)-, -C(0)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(0)-, -C(0)-, -C(0)0-, or -0C(0)-, or -0-.
[0093] Exemplary L groups include -CEC-, -CF--CCH2N(isopropyI)-, -NliC(0)CCCH2CH2-, -CH2-CEC-CH2-, -CH2C(0)CEC-, -C(0)CEC-, or -CH20C(=0)CE-C-.
[0094] 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(0)-, -C(0)NR-, -N(R)S02-, or -SO2N(R)-. Exemplary L groups include -NHC(0)-cyclopropylene-S02- and -NHC(0)-cyclopropylene-.
[0095] 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, NO2, halogen, CN, a suitable leaving group, or C1.6 aliphatic, wherein Q is a covalent bond or a bivalent Ci_6 saturated or unsaturated, straight or branched, hydrocarbon chain, wherein one or two methylene units of Q are optionally and independently replaced by -N(R)-, -S-, -0-, -C(0)-, -0C(0)-, -C(0)0-, -SO-, or -SO2-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S02-, or -SO2N(R)-; and, Z is hydrogen or C1.6 aliphatic optionally substituted with oxo, halogen, NO2, or CN.
[0096] In certain embodiments, Y is hydrogen.
[0097] 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 allcynyl optionally substituted with oxo, halogen, NO2, or CN. In some embodiments, Y is C2_6 alkenyl. In other embodiments, Y is C2-4 allcynyl.
[00981 In other embodiments, Y is C1_6 alkyl substituted with oxo, halogen, NO2, or CN.
Such Y groups include -CH2F, -CH2C1, -CH2CN, and -CH2NO2.
[0099] 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.
[00100] In some embodiments, Y is a saturated 3-4 membered heterocyclic ring having 1 heteroatom selected from oxygen or nitrogen wherein said ring is substituted with 1-2 Re groups, wherein each Re is as defined above and described herein. Exemplary such rings are epoxide and oxetane rings, wherein each ring is substituted with 1-2 Re groups, wherein each Re is as defined above and described herein.
[00101] 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 defmed ,rs NR
( 6 ( 6 above and described herein. In certain embodiments, Y is 1-2 or Re) 1 -2 ( __ 151-2 , wherein each R, Q, Z, and Re is as defined above and described herein.
[00102] In some embodiments, Y is a saturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein.
In certain embodiments, Y is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl, wherein each ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein..
In certain embodiments, Y is , wherein Re is as defined above and described herein.
In certain embodiments, Y is cyclopropyl optionally substituted with halogen, CN or NO2.
1001031 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.
[00104] 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 (Ros)-3 above and described herein. In certain embodiments, Y is , wherein each Re is as defined above and described herein.

[00105] 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:

AO NR

Re)1 -2 (Re)1.2 (Re)i-2 wherein each R and Re is as defined above and described herein.
[00106] 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.
[00107] In some embodiments, Y is selected from:
srN , r; N rõ, r VI )1-4 (rµ e )1-4 lrµe)1 -3 '-)1-3 (Re)1-3 N
wherein each Re is as defined above and described herein.
[00108] 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:
N N, N, (N N ( N


Jr- srifx, NI
( 1\1 ce w Re (R )1-3 ________________ N N ¨
0 (31 ( N
("µ C\ N e __________________________________________ -(R)1.2 Re c, ( N çcN
Re wherein each R and Re is as defmed above and described herein.
[00109] 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.
[00110] 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 C16 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)-, -0C(0)-, -C(0)0-, -SO-, or -SO2-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S02-, or ¨SO2N(R)-; and Z is hydrogen or C _6 aliphatic optionally substituted with oxo, halogen, NO2, or CN.
[00111] 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.
[00112] 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 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-, -NRC(0)-, -C(0)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(0)-, -C(0)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(0)CH=CH2 or -C(0)CH=CH2.
[00113] 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(0)0CH2C1, -C(0)0CH2F, -C(0)0CH2CN, -C(0)CH2C1, -C(0)CH2F, -C(0)CH2CN, or -CH2C(0)C113.
1001141 In certain embodiments, Re is a suitable leaving group, ie a group that is subject to nucleophilic displacement. A "suitable leaving" is a chemical group that is readily displaced by a desired incoming chemical moiety such as the thiol moiety of a cysteine of interest. Suitable leaving groups are well known in the art, e.g., see, "Advanced Organic Chemistry," Jerry March, 5th Ed., pp. 351-357, John Wiley and Sons, N.Y. Such leaving groups include, but are not limited to, halogen, allcoxy, sulphonyloxy, optionally substituted allcylsulphonyloxy, optionally substituted alkenylsulfonyloxy, optionally substituted arylsulfonyloxy, acyl, and diazonium moieties. Examples of suitable leaving groups include chloro, iodo, bromo, fluor , acetoxy, methanesulfonyloxy (mesyloxy), tosyloxy, triflyloxy, nitro-phenylsulfonyloxy (nosyloxy), and bromo-phenylsulfonyloxy (brosyloxy).
1001151 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(0)-, -C(0)NR-, -N(R)S02-, -SO2N(R)-, -S-, -5(0)-, -SO2-, -0C(0)-, -C(0)0-, cyclopropylene, -0-, -N(R)-, or -C(0)- ; and Y is hydrogen or C1.6 aliphatic optionally substituted with oxo, halogen, NO2, or CN; or (b) L is a bivalent C2_8 straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by -C(0)-, -NRC(0)-, -C(0)NR-, -N(R)S02-, -SO2N(R)-, -S-, -S(0)-, -SO2-, -0C(0)-, 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 C1.6 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 C1_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 -0C(0)-; and Y
is hydrogen or C1_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN; or (f) L is -NRC(0)CH=CH-, -NRC(0)CHHCH2N(CH3)-, -NRC(0)CH=CHCH20-, -CH2NRC(0)CH=CH-, -NRSO2CH=CH-, -NRSO2CH=CHCH2-, -NRC(0)(C=N2)-, -NRC(0)(C=N2)C(0)-, -NRC(0)CH=CHCH2N(CH3)-, -NRSO2CH=CH-, -NRSO2CH=CHCH2-, -NRC(0)CH=CHCH20-, -NRC(0)C(=CH2)CH2-, -CH2NRC(0)-, -CH2NRC(0)CH=CH-, -CH2CH2NRC(0)-, or -CH2NRC(0)cyclopropylene-; wherein R
is H or optionally substituted C1_6 aliphatic; and Y is hydrogen or Ci_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN; or (g) L is -NHC(0)CH=CH-, -NHC(0)CH=CHCH2N(CH3)-, -NHC(0)CH=CHCH20-, -CH2NHC(0)CH=CH-, -NHSO2CH=CH-, -NHSO2CH=CHCH2-, -NHC(0)(C=N2)-, -NHC(0)(C=N2)C(0)-, -NHC(0)CH=CHCH2N(CH3)-, -NHSO2CH=CH-, -NHSO2CH=CHCH2-, -NHC(0)CH=CHCH20-, -NHC(0)C(=CH2)CH2-, -CH2NHC(0)-, -CH2NHC(0)CH=CH-, -CH2CH2NHC(0)-, or -CH2NHC(0)cyclopropylene-; and Y is hydrogen or C1-6 aliphatic optionally substituted with oxo, halogen, NO2, or CN; or (h) L is a bivalent Cm 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(0)-, -C(0)NR-, -N(R)S02-, -SO2N(R)-, -S-, -S(0)-, -SO2-, -0C(0)-, 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 C1_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(0)-, -C(0)NR-, -N(R)S02-, -SO2N(R)-, ¨S-, -S(0)-, -SO2-, -0C(0)-, or ¨C(0)0-, and Y is hydrogen or C1_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN; or (1) L is -C----CCH2N(isopropyl)-, -NHC(0)C--7--CCH2CH2-, -CH2-C7=-C-CH2-, -CE-CCH20-, -CH2C(0)Cm-C-, -C(0)CC-, or -CH20C(=0)CEC-; 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(0)-, -C(0)NR-, -N(R)S02-, -SO2N(R)-, ¨S-, -S(0)-, -SO2-, -0C(0)-, or ¨C(0)0-; and Y is hydrogen or C1_6 aliphatic optionally substituted with oxo, halogen, NO2, or CN; or (1) L is a covalent bond 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 allcynyl 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)i-2 Fe)i-2 NR

\ ________________________________ (vi) 1-2 5 __ ( 61-2 , Or 1-2 , wherein each R, Q, Z, and Re is as defined above and described herein; or (vii) a saturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or (viii) a partially unsaturated 3-6 membered monocyclic ring having 0-3 beteroatoms 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 L(Re)i -2 (X) , 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 N'jL NR
____________________ (R1-2 \\jfi) 1-2 (r\\1+1)1 -2 (Xii) 0 (Re)i -2 (Re)i -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 r( c_ -7¨(R e)1_4 1¨(Re)1_4 (Re )1-3 "7-(Re) (xiv) 1-3 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
(xvi) (Re)i -3 (Re) 1 -2 <' \\ 4 (13e )1-2 isf-Re r1 .3 (Re)1_3 (peN
____________________ N 11-2 -77-0Re)1.2 N __ I/ Re ,O, c, C, N N
--(Re)1 .3 _-(R)12 \1 (Re)i-2 ,S, zoS, C% c, N
¨(Re)12 _D%
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;
(m) L is --C(0)- 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 NN--1:1-ZCJNR "NrA-(vi) _______________________________ (\ (61-2 1-2 5 ( 61-2 7 or , wherein each R, Q, Z, and Re is as defined above and described herein; or (vii) a saturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or (viii) a partially unsaturated 3-6 membered monocyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or (ix) a partially unsaturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or (449.)-3 (x) , 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 /f1)1-2 c1-1+1)1-2 ef\J-(27 (Xii) 0 (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 , rii (xiv) N
fi r N
1¨(Re)1-4 -T(Re)14 -7-(Re)1-3 -T(Re)1-3 Re)1-3 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 _______ (Nõõ.. N, N, XVO ___ sil.rt, N rV, N
N .......-N
cN
N µ 1¨(Re)i-2 ( N-2 Re (Re)i -2 ¨7\_-_-./1"---(Re) i -2 ¨
2/--Re zS,...._ ,..,,S,õ.._ ,S, ,S, \\ --ifs(Re).1.3----i_..:7\r(Re)1-2 ---\\__T-2Li-(Re)i-2 --Ti_-_,;--- 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(0)- 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) C24 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 c (/NN¨Q-Z L 'N't\IR
(vi) 5¨C 01-2( 15 ( N-5 \ _______________________________ ( 151-2 ) 1-2 , or , wherein each R, Q, Z, and Re is as defined above and described herein; or (vii) a saturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or (viii) a partially unsaturated 3-6 membered monocyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or (Li) 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 (R)-3 .2-7--(Re) (X) 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 j(NR
"ii(Re)i-2 (;11ii) 1-2 1 -2 (Xii) 0 (Re)i -2 (Re)i -2 Or (Re)1 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 fr N rrNIN c, (Re)1-4 1¨(Re)1-4 ¨NI (Rni 3 7-(Re)1-3 (XiV) 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 < c N ( N
(XVi) (Re) -3 N
N N
Re)1 -2 ,O, tµ /7 5 Ci N .
\Ljifs (Re )1 _3 \L__N (R0)1-2 ¨(R)1.2 7 Re S S ,S
si% (Re) 1 _3 NL()1-2¨j(Re)12 -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 C1-8 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 allcynyl optionally substituted with oxo, halogen, NO2, or CN; or (iv) a saturated 3-4 membered heterocyclic ring having 1 beteroatom 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)i-2Re KN--CI-Z
( NR(- N-5 (.3 _ ( 6 \ (6 (vi) \ (61-2 1-2 , or 1-2 , wherein each R, Q, Z, and Re is as defined above and described herein; or (vii) a saturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or (viii) a partially unsaturated 3-6 membered monocyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or (ix) a partially unsaturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or '1 (R)12 (x) , 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 ANR
71¨(Re)1-2 1;11i1)1-2 (?)-1/-01-2 (xii) 0 (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 (rN1- r r ¨1-(Re )i-4 7-(R91-4 = (Re)i-3 7-(Re),_3 7(Re)1-3 (xiv) 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
trN )1 -3 (IVO -7/-- (Re)i -2 jx:A, N N
, N , N
x %
c -71(Re)1-3 c "z--ipt.el _____________________ N y = /1 -2 %--(Re)1-2 17rRe .,0, ,0 (:) , 0, ..
( N N
T7_(Re)1.3 t___Nj(Re)1 -2 v // (eR )1-2 ¨..sl Re zS z S S S, 11s c\ ........Q
\\ ir-(R)13 -(R11-2 (Re)1-2 _Ill _ - 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;
(3) L is a covalent bond, -CH2-, -NH-, -C(0)-, -CH2NH-, -NHCH2-, -NHC(0)->
-NHC(0)CH20C(0)-, -CH2NHC(0)-, -NHS02-, -NHSO2CH2-, -NHC(0)CH20C(0)-, or -SO2NH-; 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 allcynyl 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) i -2 NN'-"C)-Z \,-....õ.
( NR
\ (6 (vi) 1_2 __ , 1-2 , Or 1-2 , wherein each R, Q, Z, and Re is as defined above and described herein; or (vii) a saturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or (viii) a partially unsaturated 3-6 membered monocyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or (ix) a partially unsaturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 Re groups, wherein each Re is as defined above and described herein; or (c-49)-3 (x) , 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 defuied above and described herein; or ps--(Re)i-2 /f1)1-2 (xii) 0 (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
c, fr c, '1\1 Vrt J¨(Re)14 1¨(Re)14 -T-(R91-3 1 ,o'91-3 (XlV) 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 (xvi) v___N Vs /1-2 -77-(Re)1-2 azil., ..rift, N
N N
/ N
( ._._. N
c ________ I (Re)1 .3 -(Re)1-2 µ -77--N (Re)i-2 0 0 ,O, s(r s ( N
--L.:7%-(Re)i -3 -----NI--.-N---.(Re)1-2 ¨\)___/-7---(Re)i-2 . = 4 R
N
,S ,S, N i 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.
[00116] In certain embodiments, the Y group of formula I 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:

-N
N -N -Ni N CI , -..--- --N----CH3 i a b c d e I

CH3 ..., ,,,_,3 _, =s %.,, =-5{6 55r . , N NCI

g h i j k 1 CH3 CN N.___õCN 0 0.....(CN 0 --r -(2)4N .- S "(K.IN -(2)4N - N <N0 il sCN
m n 0 P 9 7 F
F CN

F
'2? L"'- F 1101 0 F 10 (.??0 icCN
F t27 "I? NO2 'a? CN '27 S t u v w x Y
ssr`i I I I I
õ.-N.,...,-;;;- N N N N ,-. N IV
CI N
Z aa bb cc dd ee 7 's's 'ssi 'sssy -sss' N../,- ==,.-IN N .,-N N.,,INI N.--,,'' N
-,---If a hh ii jj kk Re -5'.-f I
IReN Ny.,2' ,r,N NJ,,,N
I N.N N ,,--Re Re Re Re ii mm nn oo PP 49 H
Me I \N 0 I )¨ Re HNA_ ...--N
Re I 1\N
...---N
"4------H i'l-ri Re Re rr ss rt uu vv Me ¨N 0 N

Mey _.._ ,--0 \
N
fN---Re 1 /` Re I I
J[ )¨Re f )¨Re ';14---.7 N
Re ww xx YY zz aaa 0¨N\ õ..-SN
S S--N
N f )¨Re f)¨Re ,L)---\ R
'21 -----Re bbb ccc ddd eee Iff H H Me ....- N ....- N .... N
Hy µ ,-- N N //
I / N I ) __ \\ I / N I ) //
\ L)-----1--- µ t)/1 L,14CN
Me /
ggg hhh iii .11:1 kkk Me N
IN _____ //mer\ ___________________________________ , k/c.c. ,tr-0)___\ t >
-Z-?' N N \ `,17 0 µ=
III mmm mm 000 . ppp ,..--N
/\ N
f s. , ___ I
It ) , s,¨ N N
s`,/1)----''---/ \127 --'1- µ
qqq 177 SSS 111 UUU
H Me _...õ N (/'=õ. N N--N
Hrµ ____ I \ N iL ) __ = I /\ N
;1-) Vi ;I) `;-4-?"--'-/
t \\
vvv qqq WWW XXX YYY
N - N
( Me M e NI % = 9.- \ _ 1 0\N
N NI( / ____ ;11 N 121)----\\
zzz aaaa bbbb cccc dddd N
f ) ___ = 1 ) `, N =,:ziNg 0s....--N , = t) _____________ =
1-7 `,1-1.--- 0 2'-7 S

eeee ffff gggg hhhh iiii .LiN ,z,7 0 N __ \
%
iiii kkkk 1111 mmmm nnnn S 's-r. N N
i ____________________________________________________ 'I ) = NC_>__Re ,, el L ) N%
---, , \

0000 PPPP qqqg l717' SSSS

e' ;1 I. 0 -z7 ,,r,õ
-fill uuuu vvvv wwww xxxx 0 0 0 Te (22,1...,___ Re .'- 1-.s,õN, me =
YYYY zzzz aaaaa bbbbb ccccc wherein each Re is independently a suitable leaving group, NO2, CN, or oxo.
[00117] In certain embodiments, RI is -CsCH, -C¨=CCH2NH(isopropyl), -NHC(0)CEECCH2CH3, -CH2-C----C-CH3, -C----CCH2OH, -CH2C(0)C1=-CH, -C(0)C¨=CH, or -CH20C(=0)C-i-CH. In some embodiments, RI is selected from -NHC(0)CH=CH2, -NHC(0)CH=CHCH2N(CH3)2, or -CH2NHC(0)CH=CH2-1001181 In certain embodiments, RI 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 RI Groups 0 Me a b c d H H H
e f g h i O 0 Me 0 0 0 ),L,CI .---,NAõC1 ;1.,(N...ATC1 H 41, 0 H H ' : H
=
j k 1 m n o Me H CF3 )1,=-\,,k.-7.'-' :31,---,-k-%\, P q r s 1 0 ye 0 0 0 0 u v w x Y
Etr"---, o O 0 ,\,---riy=- ;:cõ,,N .,, ).L.,.,7,.., _.\--,,NK.
00 >1.---N
Et Z aa bb cc dd ee 0, 's' I II I I I I I I
N N,õ...;,-,- N NThrµl N .= N 14,,,, I
if a hh ii jj kk 7 'sssy 'sc 'ss -I) 1 I I
N N
',..,...I I I II
/ N--;-= ,,..,7 N N N .- N N,,,.,,,-2 /
I I I I I I I I
//mm nn oo PP qq -, . , , --...f, ITV-N N ,-- N N, N N N
-- N ,--I ,e-Re Re Re Re TT SS 11 Ull VV 'VW

H
..-N

NO
H X
R
xx YY zz aaa Me Me -N
N _--N N
1. Mey _Re 4N I ¨R" { ¨Fte N. X" N
M
R6 e bbb ccc ddd eee I N t ----Re t itz_i itt N N. 0 X
Re fff ggg hhh I N I --Re ,1 )¨Re jzzz)--Re t. S 'AL
Re irl kkk III mmm H H Me ..,-N N ..-N N
I \N 1 FIN % ___ Merry I \N
(___ /
nnn 000 PPP 999 Me ,-0\ õ..-0 N
I N __ //N .µ f ) t;17---t rrr sss fit uuu vvv s--N N /-===
I i\N \ , www XXX YYY zzz aaaa MIT

zHO=HOzHO 1-10zH3 cHO
9-10, N N
,-, ,,,,,,,,,,r..A-9-10, N.--..,;,---,r1V.,.õ.-42( cHO,N.---,,,,:,,,õThr,N,X
Ã1-16 0 cH0 0 cH6 0 um WPM 22222 f f aaaaa PPPPP .70333 N
, ,7.(r:!
A
0 ,õõro õ,.õ ,,, NH, ,,---- ,,, -,µ:

VIM VVV1111 2222 raid A.AA.-1 illniAL4-1 AMA
0 H a H H 0 o INJ''':. al\
N.J
A -',T,/%"Nri- cr.-NA N

d 0 d 0 d 0 nnnn nu SSSS .111.1 bbbb -sis:r ,Nrs.
N µ. /=-----10 'd--C1N = (N) \\( ) 11\1õ
N
dddd 0000 uuuu :murmur 1M
0 e<
N/ I N I
µN--j N S

YINY tiff !!!! Mil 2222 _ c,,,,,= ej,,, N/ I
N --S \ 0 N-- N-- 0 \ 1 ffJf aaaa PPPP 3333 qqq11 \, \\, N H
Lz.', N Y, NI / ______________ (1 _____________ , -- = ______________ ( j NI I
N--Nan N N N
N an H
an 9; *

0 0 0 \ z 0 0 mmmmm nnnnn 00000 PPPPP 99999 , N , H3C C n , , TIM SSsSS WU uuuuu 0µ ( 0 0 \z..---0 0 ,, '1',_) 1 0 v-L-CI = :ss.,,= CN (-0F
,,1,1 vvvvi, wwwww xxxxx yyyyy zzzzz aaaaaa bbbbbb 0,\ (0 0 CH3 0 S-----7:- CH ',t1,.."-Ii 1_,-, ;111.Thr-r CH3 '-`7-t-jC Ac sISS'141-1 3 0 0 0 CH3 CCCCCC dddddd eeeeee ffffff gggggg hhhhhh `,11c-y- C H3 ',Ell.
Thr'OAc --i-Th.-\
um: 111111 kkkkkk 111111 mmmmmm nnnnnn 0 OHO 0 OH ,,N*.,.F
)(0Et 74e,))-(0 Et 40µYLOEt /1/4),,,.õ ON \ I A
000000 PPPPPP 999999 rrrrrr ssssss -,'N jc.....- N 1-'4- -)LCDM e 'I'ltj H
-- st MU uuuuuu vvvvvv wwwwww or XYrrrX
wherein each Re is independently a suitable leaving group, NO2, CN, or oxo.
[001191 As defined generally above, RI is a warhead group, or, when RI 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 RI groups, i.e. warhead groups, are particularly suitable for covalently binding to a key cysteine residue in the binding domain of certain protein lcinases.
Protein lcinases having a cysteine residue in the binding domain are known to one of ordinary skill in the art and include ErbBl, ErbB2, and ErbB4, or a mutant thereof In certain embodiments, compounds of the present invention have a warhead group characterized in that inventive compounds target one or more of the following cysteine residues:
ERBB 1 SEQ ID 7: ITQLMPFGCLLDYVREH
ERBB2 SEQ ID 8: VTQLMPYGCLLDHVREN
ERBB4 SEQ ID 9: VTQLMPHGCLLEYVHEH
[00120] Thus, in some embodiments, RI is characterized in that the -L-Y moiety is capable of covalently binding to a cysteine residue thereby irreversibly inhibiting the enzyme. In certain embodiments, the cysteine residue is Cys797 of ErbBl, Cys805 of ErbB2 and Cys803 of ErbB4, or a mutant thereof where the provided residue numbering is in accordance with Uniprot (code P00533 for ErbBl; code P04626 for ErbB2, and Q15303 for ErbB4). It will be understood that the Cys of ErbBl (EGFR) is variably called 773 or 797 depending on whether the parent sequence contains the signal peptide or not. Thus, in accordance with the present invention, the relevant cysteine residue of ErbB1 may be described as Cys 773 or Cys 797 and these terms are used interchangeably.
[00121] 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 RI groups include, but are not limited to, those described herein and depicted in Table 4, infra. One of ordinary skill in the art will recognize that ErbB3 has no corresponding residue and, as recognized in the relevant art, is not catalytically active.
[00122] As depicted in Formula I supra, the RI warhead group can be in an ortho-, meta-, or para-position. In certain embodiments, the RI warhead group is in a meta-position of the phenyl ring relative to the rest of the molecule. Without wishing to be bound by any particular theory, it is believed that when RI is in such a meta-position, the warhead group is better positioned for covalent modification of the cysteine residue thus effecting irreversible inhibition of the enzyme.
Indeed, it has been surprisingly found that a compound having a warhead group at a meta-position (compound I-1) irreversibly binds to ErbBl whereas a compound having a warhead group at a para-position (compound 1-93) reversibly binds to ErbBl. These compounds have the following structures:
NH
BrBr IS
NH NH

Oi N N N N

[00123] This phenomenon was determined by performing a washout experiment using the protocol described in detail in Example 42, infra. The results of this experiment are depicted in Figure 2 where it is shown that compound 1-1 maintains enzyme inhibition after "washout"
whereas compound 1-93 was washed away in the experiment thereby resulting in reactivated enzyme activity.
[00124] In certain embodiments, RI 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.
[00125] In certain embodiments, RI 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.
[00126] In certain embodiments, RI 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.
[00127] In certain embodiments, RI 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.
[00128] In certain embodiments, RI 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.
[00129] In certain embodiments, RI 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.

100130] 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 RI groups include, but are not limited to, those described herein and depicted in Table 3, infra.
1001311 Exemplary compounds of formula I are set forth in Table 5 below.
Table 5. Exemplary Compounds of Formula I
a a 010/ o F
,k,%. F

HN.K%

NH
Br NH HN NH
N-1.'' L 1:
N N N''''' N41 Is1 k "- N
LJ ill H H H

F
CH3 CH3 a i 0 HN,..,"" 411 ,s,,..., 40 0 ) "I0 0 .K....., NH NH HN NH HN
NICII 011] rid 00 (11 0 N N N N N N
H H H

F
c cF, a 0 .
OP 0 .
..,õ
OP 1/4., Br ....s,.. NH HN)i.-= 411 0 HN.).
NH HN
N kk"
lel N(1), 410 N N N N
H H H

, NH
II Br 0 I I
NH

S HN
N ..., 0 ( .
N 0 NiCi''', .
N N N N N N
H H H

0 = 0 0 o o *

) )s*, 111 0 -5-c NL 1 0 (\
( V
N N N N N N
H H H

ra N
Me a =
* o HNA,.....% . 0 I F
H. NKip.,..., Nõ 0 o I
H_ N.J....,..,. N.,.
a NH kri C:r4H r\i) .
11,N N N 0 N N N N
H H H

N
Y CI N
F
N,r F 0 = 0 NH HN)...,1\1 0 II
CI a NH NH N--H
hi--L 010 NL 0 Nk)N),N 0 11,N,----,N
kN---'rµl H H H

õõ-=-=,,,...
'N--.

N
CI CI
CI

01.5) F 0 F 0 0 , NH
H,N * NH ,N NH HN,k( H
NC`* N(5. 40 1µ1)) 0 N
k.NN N N 1N N 0 H H H

\
IN--r----Ata N z "
\---I\I
CI CI
F F

NH
HN
, ) 1.1 ( 0 0 1 N)),.. N N".-L 0 NN*
N"-L

Q . 0 IN N k-N
H H H

N-..--\
Nj ,N
y N --'1 I
Me .

0 At )C r'l , a I S,N 0 le 0 NH HN NH NH HNI)CV
N''' 010 Nd le NIL 010 kN N
Y.N1 N N
H H H

F
CI CI N=< CI

0 N"7--.`

"
NH HNIA'40 NH H
-'1) NH HN 0 NS , N d, , .' I
0 tµI
N N N N 0 N '-- N
11.. jiNat , = i: N
H H H

F *F
NI \ N/ \
F F
CI CI

0sr" 0 NH II 0 0 F
µµ .......
NH NW- ) NH HN 0 N'L- 0 N),...õ 0 NICI), 0 E:N-,=-.NCN

H H

CI CI CI
F F

Oa ,NH IN 0 NH HNA6 0 H HN N

N-k.., N N

(5, N N N N
H H H

CI CI CI

NH HN)LxCN
NH HN 110 NH HN'k-----Li j 0 Kr-L 0 N).''''''- 0 Nkta- 0 t 1..N N kNN N N
H H H

----1\1 CI CI
CI
F * 0 0 vi AxCN NH HNAA NH HNAXS
= NNH H
N 0 N(5-, 0 Nik--1:j. , 0 k)).-N N N N N N
H H H

F
F = NC:\ F

= 0 0 CI 4111IPP NH HN-K-N, CI NH HN 0 CI NH MeN,K -,Nõ
N *
N it d N 0 d H H H

, F 0 F 0 F CI
HNCI CI NH HNI
CI SI NH HN--"'0 C NH
INJ). IS) N)i., 411:ttF
N 'L . F
Q.NN N N 11.N N
H H H

0 F .

F WI&
CI NH HN,/,--.,., N ,, CI NH CI N)C' NH HN
NJ) 0 ( )) 0 Q.NN
N N N N
H H H

Br A F F
0 0 0 (NH
NH HNL H,NNõ) H,N) WI NH CI --I'h = NH
N")-teN 0 1\N = NL 0 tt..N!,--.N
Q' (r H H H

F s 0 NH H. 0 1 ,A.7"--..,N CI NH HN)-'' 0 r NA' NH H-N o NC:-L1 0 N'L- 0 N N N NH N H ,,c) H H

0 *
o CI
NH HN)t o õ..,N ,CD3 0 NH HN o I
)1 0 N,µ, NH
NN---1'..' Nd NtC
N N N N N N
H H H

*
o 0 o HN)(õ.."

NH HN

N') .--'1 N'-'''Ll õ N,N N I
H H

a o 0 o )t, NH
)cL", *
HN,J1,,,.% õ.- _,-0 HN NH HN -:.'"
N'k N N").ks''- 0 N - k N.)--,,,j .NN is 0 N N
H H H

I H I H kiNiniiN
N...,-.7.-...irN,,i,,N.NN) _52,-....T,N,,fNN .,,N ,r--õõ*_Thr- ,T ,,.., ,.., ) 1 0 0.,_õ..,-0 ,.,,.,..-- -zyN
= 0 NH NH 46 0 . 0 0 la IIIPI
A

H H H H H H
N N ,-, ,--õ, ,N.NN.,,..N
'''INJ - Tr 1. )1 I
0 I .. =.,rN 0 IIIP N 0 =0 * NH =0 A 0, EL-I ZL-I IL-I
H H H H H
NN N N ===., õN õN N"Pi) .,,,..,,N,_,N
*.INJ----'"--%. ''''' ---' ''-== ) ir --1 --- ) I I
0N 0 :.,.- ',.=,N -..N,- N
NH
*
110 NH -,.,,..k.0 HN
0 d H H
N N N N
* 'cfiN
rµQ 10 L. NH 0 10 HN
0 A o 0 50. d N N
H H J
d I
s 0 *
0 j HN.,-...0 H
Nj'' =

N

.11 N';:L.` NA`="-Q'N"---14 N N
H H

= 0 . 0 NI-L-"''''-i 0 1\r'L-- 0 It.N--. N
H H H

. N
* 0 =
NH

$ 0 HO CH2 0¶
NH
N r-l'''= X:21>i . I 0 N
kN N
N N
H H

0y, NH
1\1L N NL 0 It. N--------,N ..-----...õ/ Qs.N N
H H

0 ..,_,.\ 10 0 0 I
LI: ,NH ill\l, N = N 0 N N N N
H H

HN.J-L--0 0 RN = 0 N-L'-` N'")'-'i N--I),, N"-j'' N N
H H

F =

I
CI 0 HNI\I Br = NH
H
N1r-, N-k`. 0 N -'--.NN0N N 0 H H

HN)L,.-:-." (0 el 0 HNo )0t,,,, 0 NLN--j'; fµlL Nj' kN--,..N.,,,,L ..N N
N
H H H

[00132] In certain embodiments, the present invention provides any compound depicted in Table 5, above, or a pharmaceutically acceptable salt thereof.
[00133] In certain embodiments, the present invention provides a compound selected from:
rn N
Me CI
=

HN.K 0 , 0 1 NH

a 0 NH H. NAN
NH
N.--1, 0 N-1, 40 N--L 0 kNN kN-'-' N
H H H

N
a o =

CI NH HNN NH HN
hrL N
NN N N

0 lip0 HN)L,s, N-j), %-''= 0 N N
NNNN

NH
NL
NN

or a pharmaceutically acceptable salt thereof.
[00134] 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-Icinase (e.g. BTK) and JAK3. One of ordinary skill in the art will recognize that certain compounds of the present invention are reversible inhibitors. In certain embodiments, such compounds are useful as assay comparator compounds. In other embodiments, such reversible compounds are useful as inhibitors of ErbBl, ErbB2, ErbB3, ErbB4, a TEC-Icinase, and/or JAK3, or a mutant thereof, and therefore useful for treating one or disorders as described herein. Exemplary reversible compounds of the present invention are set forth in Table 6, below.
=

Table 6. Reversible Inhibitors OLNH . 0 o Br NH2 F3 NH NH, Br NH HN

N'''L`

NN*N N N N
H H H
ift_i 1R-2 1R-3 F
a F 0 = 0 a NH NH2 H3 NH NH2 NH NH2 /4) L'''=
* el 10/ N
IL
L
N N N N
H H H
/1(4 1R-5 1R..6 0 'N a =
0111 0 ,õ..,,,..,,,=
k),H

NCJI *
C

N N
H H H
/12_7 /11_8 itt_9 o 0 NH _JoL_____ 0 0 NH NH2 * 0 NH2 HN
N--k''-0 NLN--1' N-L- N-)1 It.N N (N.---N---1--,-, NN
H H H

0 40 le 0 N N N
NH N

02N I.Br F 0 N
N N

or a pharmaceutically acceptable salt thereof.
4. Uses. Formulation and Administration Pharmaceutically Acceptable Compositions 1001351 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 ErbB 1, 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 ErbBI, 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.
[00136] The term "patient", as used herein, means an animal, preferably a mammal, and most preferably a human.
1001371 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 vehicle's 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.
[00138] 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.
[00139] 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-Icinase, and/or JAK3, or a mutant thereof.
[00140] 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 I,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.
[00141] 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 TweenZmSpans 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.
1001421 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.
1001431 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.
[00144] 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.
100145] 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.
100146] 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 can-iers. 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.
[00147] 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.
[00148] 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.
[00149] Most preferably, pharmaceutically acceptable compositions of this invention are formulated for oral administration.
[00150] 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.
[00151] 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 [00152] Compounds and compositions described herein are generally useful for the inhibition of protein kinase activity of one or more enzymes.
[00153] 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.
[00154] 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.
[00155] 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 change, decrease or abolish the inhibitory effect of the inhibitor on the target protein.
1001561 Examples of kinases that are inhibited by the compounds and compositions described herein and against which the methods described herein are useful include ErbB
I, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3, or a mutant thereof [00157] The activity of a compound utilized in this invention as an inhibitor of ErbB I, 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 ErbBI, 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 I, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3. Inhibitor binding may be measured by radiolabelling the inhibitor prior to binding, isolating the inhibitor/ErbBI, inhibitor/ErbB2, inhibitor/ErbB3, inhibitor/ErbB4, inhibitor/TEC-kinase, 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 ErbB1, 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.
[00158] 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.
[00159] 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 ErbB1 (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-1, 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 (PI3K) 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).
[00160] Interaction between family members is necessitated by deficiencies in ErbB2, which has no known ligand, and ErbB3, which is kinase dead. EGER. ErbB3, and ErbB4 bind ligand to induce ErbB receptor homodimerization or heterodimerizat ion, 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 Shc, Grb2, SOSI, Ras, Rafl, Mek, ERK1, ERK2, Elba, Alct, mTOR, FKHR, p27, Cyclin D1, FasL, GSK-3, Bad, and STAT3.
[00161] 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, ttunorigenic EGFR
vIll, 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). ErbB1 and/or ErbB2 amplification has also been implicated in squamous cell carcinomas, salivary gland carcinomas, ovarian carcinomas, and pancreatic cancers (Cooper, G.C. Oncogenes. rd ed. Sudbury: Jones and Barlett, 1995;
Zhang, Y., at 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 11: 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.
[00162] 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. (Ancirulis, LL., et al., J
Clin Oncol 16: 1340-9, 1998). =
[00163] Targeted blockade of ErbB signaling with trastuzumab (Hereeptin), 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)TM and paniturnurnab (Vectibi41.
[00164] Several small molecule tyrosine kinase inhibitors (TKIs) have been found to act selectively upon ErbB family members. Notable examples include gefitinib (Iressa) and erlotinib (TarcevT4, both of which target the EGFR. These small molecules compete with ATP
for binding to the kinase domain of the receptor. Compared to monoclonal antibodies, TKIs have several advantages in that they are orally bioavailable, well-tolerated, and appear to be active against truncated forms of ErbB2 and EGFR receptors (e.g., EGFR viii) in vitro. In addition, the small size of small molecule TKIs may allow them to penetrate sanctuary sites such as the central nervous system. Finally, the homology between kinase domains of ErbB receptors allows for development of TKIs that target more than one member of the ErbB
family simultaneously, the advantages of which are described herein.
[00165] 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).
[00166] 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.
Pan-ErbB regulators may be more successful in treating tumors than compounds that only target one ErbB receptor. Erbitux and Herceptin show success in a limited patient base (tumors having increased expression of EGFR or ErbB2), which could be partly due to lack of pan-ErbB
activity.
[00167] 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 the 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 ErbB I, ErbB2, ErbB3, and ErbB4. In some embodiments, provided compounds inhibit two or more of ErbB1 , ErbB2, ErbB3, and ErbB4, or a mutant thereof; and are therefore pan-ErbB inhibitors.
[00168] 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).

[00169] 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 Icinase domain.
Another irreversible pan-ErbB receptor tyrosine lcinase 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.
[00170] Neurofibromatosis type I (NF1) 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 NF1 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, with Schwann cells being the major (60-80%) cell type.
[00171] Abberant expression of the EGFR is associated with tumor development in NF1 and in animal models of NF1, 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 NF1 tumorigenesis and Schwann cell transformation (DeClue, J. E., et al., J Chin Invest 105: 1233-41, 2000).
[00172] Patients with NE I develop aggressive Schwann cell neoplasms 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 Icinases mediating NRG-1 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).
[00173] Deregulation of Schwann cell growth is a primary defect driving the development of both benign neurofibromas and MPNST in neurofibromatosis type I (NF1) 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).
[00174] 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.
[00175] 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).
[00176] 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).
[00177] 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.

[00178] Provided compounds are inhibitors of one of more of ErbB 1, ErbB2, ErbB3, and ErbB4 and are therefore useful for treating one or more disorders associated with activity of one of more of ErbBl, ErbB2, ErbB3, and ErbB4. Thus, in certain embodiments, the present invention provides a method for treating an ErbBl-mediated, an ErbB2-mediated, an ErbB3-mediated, and/or ErbB4-mediated disorder comprising the step of administering to a patient in need thereof a compound of the present invention, or pharmaceutically acceptable composition thereof.
[00179] As used herein, the terms "ErbBl-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 ErbBl, ErbB2, ErbB3, and/or ErbB4, or a mutant thereof, are known to play a role. Accordingly, another embodiment of the present invention relates to treating or lessening the severity of one or more diseases in which one or more of ErbB1, 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.
[00180] 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.
[00181] In certain embodiments, the present invention provides a method for treating or lessening the severity of neurofibromatosis type I (NF1), neurofibromatosis type II (NF2) Schwann cell neoplasms (e.g. MPNST's), or Schwannomas.
[00182] The TEC family of non-receptor tyrosine lcinases, referred to herein as "TEC-kinases," plays a central role in signaling through antigen-receptors such as the TCR, BCR and Fee receptors (reviewed in Miller A, et al. Current Opinion in Immunology 14;
331-340 (2002).
TEC-Icinases 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-g. Combined deletion of Itk and Rik in mice leads to a profound inhibition of TCR responses including proliferation, cytolcine 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-lcinases are also essential for B cell development and activation.
[00183] 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).
[00184] 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.
[00185] 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 autoimrnune, inflammatory, proliferative, and hyperproliferative diseases and immunologically-mediated diseases including rejection of transplanted organs or tissues and Acquired Immunodeficiency Syndrome (AIDS), wherein said method comprises administering to a patient in need thereof a composition of the present invention.
[00186] 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 thinitis 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.
[00187] 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, (pannus formation in) rheumatoid arthritis, seronegative spondyloarthropathis (including ankylosing spondylitis, psoriatic arthritis and Reiter's disease), Behcet's disease, Sjogren's syndrome, and systemic sclerosis.
1001881 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.
[00189] In some embodiments, the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with TEC-kinases including diseases and disorders of the gastrointestinal tract, including, without limitation, celiac disease, proctitis, eosinophilic gastro-enteritis, mastocytosis, pancreatitis, Crohn's disease, ulcerative colitis, food-related allergies which have effects remote from the gut, e.g., migraine, rhinitis and eczema.
[00190] In some embodiments, the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with TEC-kinases including those diseases and disorders of other tissues and systemic disease, including, without limiation, multiple sclerosis, artherosclerosis, lupus erythematosus, systemic lupus, erythematosus, Hashimoto's thyroiditis, myasthenia gravis, type I diabetes, nephrotic syndrome, eosinophilia fascitis, hyper IgE syndrome, lepromatous leprosy, sezary syndrome and idiopathic thrombocytopenia purpura, restenosis following angioplasty, tumours (for example leukemia, lymphomas including prostate cancers), artherosclerosis, and systemic lupus erythematosus.
[00191] In some embodiments, the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with 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.
[00192] 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.
[00193] Bruton's tyrosine kinase ("BTK"), a member of TEC-kinases, is a key signaling enzyme expressed in all hematopoietic cells types except T lymphocytes and natural killer cells.
BTK plays an essential role in the B-cell signaling spathway linking cell surface B-cell receptor (BCR) stimulation to downstream intracellular responses.
1001941 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 1NF-a production in macrophages, IgE receptor (FcepsilonRI) 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 ei a/. (1998), Current Biology 8(20): 1137-1140.

1001951 Patients with mutations in BTK have a profound block in B cell development, resulting in the almost complete absence of B lymphocytes and plasma cells, severely reduced Ig levels and a profound inhibition of humoral response to recall antigens (reviewed in Vihinen et al Frontiers in Bioscience 5 : d917-928). Mice deficient in BTK also have a reduced number of peripheral B cells and greatly decreased 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 (FceRI).
BTK deficient murine mast cells have reduced degranulation and decreased production of proinfllatnmatory cytokines following FceRI cross-linking (Kawakami et al.
Journal of leukocyte biology 65: 286-290).
1001961 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 an 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.
1001971 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, wherein said method comprises administering to a patient in need thereof a compound or composition according to the present invention.
1001981 In some embodiments, the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with BTK. In some embodiments, the disease or condition is an autoimmune disease, e.g., inflammatory bowel disease, arthritis, lupus, rheumatoid arthritis, psoriatic arthritis, osteoarthritis, Still's disease, juvenile arthritis, diabetes, myasthenia gravis, Hashimoto's thyroiditis, Ord's thyroiditis, Graves' disease, Sjogren's syndrome, multiple sclerosis, Guillain-Barre syndrome, acute disseminated encephalomyelitis, Addison's disease, opsoclonus-myoclonus syndrome, ankylosing spondylosis, antiphospholipid antibody syndrome, aplastic anemia, autoimmune hepatitis, coeliac 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, dysautonornia, endometriosis, interstitial cystitis, neuromyotonia, scleroderma, or vulvodynia.
1001991 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.
1002001 In some embodiments, the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with BTK, wherein the disease or condition is selected from an inflammatory disease, e.g., asthma, appendicitis, blepharitis, bronchiolitis, bronchitis, bursitis, cervicitis, cholangitis, cholecystitis, colitis, conjunctivitis, cystitis, dacryoadenitis, dermatitis, dennatomyositis, encephalitis, endocarditis, endometritis, enteritis, enterocolitis, epicondylitis, epididymitis, fasciitis, fibrositis, gastritis, gastroenteritis, hepatitis, hidradenitis suppurativa, laryngitis, mastitis, meningitis, myelitis myocarditis, myositis, nephritis, oophoritis, orchitis, osteitis, otitis, pancreatitis, parotitis, pericarditis, peritonitis, pharyngitis, pleuritis, phlebitis, pneumorntis, pneumonia, proctitis, prostatitis, pyelonephritis, rhinitis, salpingitis, sinusitis, stomatitis, synovitis, tendonitis, tonsillitis, uveitis, vaginitis, vasculitis, or vulvitis.
[00201] 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, B-cell prolymphocytic leukemia, lymphoplasmacytic lymphoma/Waldenstrom macroglobulinemia, splenic marginal zone lymphoma, plasma cell myeloma, plasmacytomaõ 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 or prostate cancer.
[002021 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.
1002031 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, appendictitis, pancreatitis, cholocystitus, agammaglobulinemia, psoriasis, allergy, Crohn's disease, irrtiable 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, athersclerosis, Addison's disease, Parkinson's disease, Alzheimer's disease, Type I diabetes, septic shock, systemic lupus erythematosus (SLE), rheumatoid arthritis, psoriatic arthritis, juvenile arthritis, osteoarthritis, chronic idiopathic thrombocytopenic purpura, Waldenstrom macroglobulinemia, myasthenia gravis, Hashimoto's thyroiditis, atopic dermatitis, degenerative joint disease, vitiligo, autoimmune hypopituatarism, Guillain-Barre syndrome, Behcet's disease, scleracierma, mycosis fungoides, acute inflammatory responses (such as acute respiratory distress syndrome and iscbemia/reperfusion injury), and Graves' disease.

[00204] 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 chromic lymphocytic leukemia, hairy cell leukemia, non-Hodgkin's lymphoma, irritable bowel syndrome, Crohn's Disease, lupus and renal transplant.
[00205] Interleulcin-2 tyrosine 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 Ilk (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).
[00206] 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).
[00207] 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.
[00208] It has been well established that T cells play an important role in regulating the immune response (Powrie and Coffman, 1993, Immunology Today, 14, 270-274).
Indeed, activation of T cells is often the initiating event in immunological disorders. Following activation of the TCR, there is an influx of calcium that is required for T cell activation. Upon activation, T
cells produce cytokines, including IL-2, 4, 5, 9, 10, and 13 leading to T cell proliferation, differentiation, and effector function. Clinical studies with inhibitors of IL-2 have shown that interference with T cell activation and proliferation effectively suppresses immune response in vivo (Waldmann, 1993, Immunology Today, 14, 264-270). Accordingly, agents that inhibit T
lymphocyte activation and subsequent cytokine production, are therapeutically useful for selectively suppressing the immune response in a patient in need of such immunosuppression.
[00209] Mast cells play a critical roll in asthma and allergic disorders by releasing pro-inflammatory mediators and cytokines. Antigen-mediated aggregation of Fc.epsilon.RI, the high-affinity receptor for IgE results in activation of mast cells (D. B. Corry et al., 1999, Nature, 402, B18-23). 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.
[00210] 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, Immu7lity 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 mucous production are drastically reduced in ITK-/-mice in response to challenge with the allergen OVA
(Mueller et al, Journal of Immunology 170: 5056-5063 (2003)). ITK has also been implicated in atopic dermatitis. This gene has been reported to be more highly expressed in peripheral blood T
cells from patients with moderate and/or severe atopic dermatitis than in controls or patients with mild atopic dermatitis (Matsumoto et al, International arcllives of Allergy and Immunology 129:
327-340 (2002)).

1002111 Splenocytes from RLK-/-mice secrete half the IL-2 produced by wild type animals in response to TCR engagement (Schaeffer et al, Science 284: 638-641 (1999) ), while combined deletion of ITK and RLK in mice leads to a profound inhibition of TCR-induced responses including proliferation and production of the cytokines IL-2, IL-4, IL-5 and IFN-y (Schaeffer et al, Nature Immunology 2 (12): 1183-1188 (2001) ), Schaeffer et al, Science 284: 638-641 (1999)). Intracellular signalling following TCR engagement is effected in ITK/RLK deficient T
cells; inositol triphosphate production, calcium mobilization, MAP lcinase activation, and activation of the transcription factors NFAT and AP-1 are all reduced (Schaeffer et al, Science 284: 638-641 (1999), Schaeffer et al, Nature Immunology 2 (12): 1183-1188 (2001)).
[00212] 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.
1002131' 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.
1002141 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.
1002151 In certain embodiments, the present invention provides a method for treating or lessening the severity of one or more diseases and conditions associated with ITK, wherein the disease or condition is acute or chronic inflammation, an allergy, contact dermatitis, psoriasis, rheumatoid arthritis, multiple sclerosis, type 1 diabetes, inflammatory bowel disease, Guillain-Barre syndrome, Crohn's disease, ulcerative colitis, cancer, graft versus host disease (and other forms of organ or bone marrow transplant rejection) or lupus erythematosus.
[00216] 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. Such conditions include those described in Readinger, J.A. et al., "Selective Targeting of ITK Blocks Multiple Steps of HIV
Replication", PNAS 2008, Vol. 105, No. 18 (May 6, 2008).
[00217] The Janus kinases (JAK) are a family of tyrosine kinases consisting of JAK1, JAK2, JAK3 and TYK2. The JAKs play a critical role in cytolcine 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 Mol. Med. 5 :
432-456 (1999) si Seidel, et al, Oncogene 19 : 2645-2656 (2000)].
[00218] 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.
[00219] 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) ].
[00220] 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.
[00221] 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) ].
[00222] 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 1L-4-associated JAK kinases are expressed in the RA synovium [Muller-Lather, et al, J. Immunol. 164: 3894-3901 (2000)].
[00223] 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)].
[00224] 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.
[00225] 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.

[00226] 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, autoirnmune diseases such as transplant rejection, graft versus host disease, rheumatoid arthritis, amyotrophic lateral sclerosis, and multiple sclerosis, neurodegenerative disorders such as familial amyotrophic lateral sclerosis (FALS), as well as in solid and hematologic malignancies such as leukemias and lymphomas, wherein said method comprises administering to a patient in need thereof a composition according to the present invention.
[00227] 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 nettrodegenerative 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.

[00228] Pharmaceutically acceptable compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracistemally, 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.
[00229] Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, inicroemulsions, 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.
[00230] 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 diglycericles. In addition, fatty acids such as oleic acid are used in the preparation of injectables.
[00231]
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.

[00232] 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.
[00233] 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.
[00234] 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, alginie 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.

[00235) 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 pacifying 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.
[00236] 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.
[00237) 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.
[00238] 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.
[002391 According to another embodiment, the invention relates to a method of inhibiting ErbBI, 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 JAK.3õ 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.
[00240] 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.
[00241] Inhibition of protein kinase, or a protein kinase selected from ErbB1, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3õ 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.
[00242] 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.
1002431 According to another embodiment, the invention relates to a method of inhibiting one or more of ErbB 1 , ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3õ 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 I , ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK3õ 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 ErbBI, ErbB2, ErbB3, ErbB4, a TEC-kinase, and/or JAK.3, 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.
1002441 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."
100245] 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, Adrianaycin, 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, actinonaycin D, dolastatin 10, cokhicine, ernetine, trimetrexate, metoprine, cyclosporine, daunorubicin, teniposide, arnphotericin, alkylating agents (e.g., chlorambucil), 5-fluorouracil, campthothecin, cisplatin, menonidazole, and GleevecTm, among others. In other embodiments, a compound of the present invention is TM TM
administered in combination with a biologic agent, such as Avastin or VECTII3DC.
1002461 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, aldesleuldia, Aldesleulcin, Alemtuzumab, Alitretinoin, Allopttrinol, Altretamine, Annfostine, Anastrozole, Arsenic trioxide, Asparaginase, Azacitidine, ECU Live, Bevacuzimab, Fluorouracil, Bexarotene, Bleomycin, Bortezomib, Busulfan, Calusterone, Capecitabine, Camptothecin, Carboplatin, Carmustine, Celecoxib, Cetuximab, Chlorambucil, Cladribine, Clofarabine, Cyclophosphamide, Cytarabine, Dactinomycin, Darbepoetin alfa, Daunorubicin, Denileulcin, 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, Oprelvelcin, 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, Temozolornide, Teniposide, VM-26, Testolactone, Thioguanine, 6-TG, Thiotepa, Topotecan, Toremifene, Tositumomab, Trastuzumab, Tretinoin, ATRA, Uracil Mustard, Valrubicin, Vinblastine, Vincristine, Vinorelbine, Zoledronate, or Zoledronic acid.
1002471 Other examples of agents the inhibitors of this invention may also be combined with include, without limitation: treatments for Alzheimer's Disease such as Aricept and Excelon ;
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 Rebie), Copaxone , and mitoxantrone; treatments for asthma such as albuterol and 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.

[00248] 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.
[00249] 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.
[00250] 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 compound of formula I, an additional therapeutic agent, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
[00251] 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.
[00252] 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 tig/kg body weight/day of the additional therapeutic agent can be administered.
[00253] 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.
[00254] 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.
EXEMPLIFICATION
[00255] 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.

SCHEME la Synthesis of Compounds of Formula H-a T(Rx)rn Ring A-NH2 , DIEA
1µ11 _____________________ N N
L, jt-(Rx)rn N CI Et0H or n-BuOH, 60-80 C CI n-BuOH, 120-150 C N N
[00256] Synthesis of (6-ehloro-pyrimidin-4-y1)-(3-bromo-phenyl)-amine: A
solution of 4,6-dichloropyrimidine (5 g, 33.6 mmol), 3-bromoaniline (5.8 g, 33.7 mmol) and N,N-diisopropylethylamine (DIEA) (5.2 g, 40.2 mmol) in ethanol (40 mL) was heated at 80 C for 16 hr. The reaction mixture was cooled to ambient temperature, diethylether (35 mL) was added while the mixture was being stirred. The product was precipitated, filtered, washed with water and dried to afford 5.9 g (62% yield) of a light colored solid. MS (m/z): MH+
= 284, 286, 288.
1002571 Synthesis of 3-16-(3-bromophenylamino)-pyrimidin-4-ylaminol-phenylamine: A
mixture of (6-chloro-pyrimidin-4-y1)-(3-bromo-phenyl)-amine (300 mg, 1.1 nunol) and benzene-1,3-diarnine (300mg, 2.75 mmol) in 3 mL of n-BuOH was heated in a sealed tube to 150 C for 16 hr. Solvent was removed by vacuum evaporation and the crude product was purified by flash chromatography on silica gel with Et0Ac/DCM solvent system to afford 255 mg (65% yield) of the title compound as a yellow solid. MS (m/z): mtv- = 356, 358.
1002581 The following compounds were prepared in a manner substantially similar to that described in Scheme la and the Examples above:
(a) 3-Bromoaniline and benzene-1,4-diamine gave 446-(3-bromophenylamino)-pyritnidin-4-ylarnino]phenylamine: MS (m/z): MH+ = 356, 358.
(b) 3-Chloro-4-fluoroaniline and benzene-1,3-diamine gave 34643 -chloro-4-fluorophenylamino)-pyrirnidin-4- ylamino]phenylamine (IR-4). MS (m/z):ME1+ =
330, 332.
(c) 3-Methylaniline and benzene-1,3-diamine gave 346-(3-methylphenylamino)-pyrimidin-4-ylarnino]phenylamine (IR-5). MS (m/z): MIT+ = 292.
(d) 3-Chloro-4-(3-fluorophenyl)methoxyaniline and benzene-1,3-diamine gave 3-{643-chloro-4-(3-fluorophenyl)methoxyphenylamino]-pyrimidin-4-ylaminolphenylamine (I'-
6). MS MS (m/z): MH+ = 436, 438.
(e) 3-Bromoaniline and 3-ethynylaniline gave /V'-(3-Bromopheny1)-N-(3-ethynylphenyl)pyrimidine-4,6-diamine (I-12). MS (M + H+) 365, 367; 'H NMR (400 MHz, d6-DMS0) 69.73 (s, 1H), 9.60 (s, 1H), 8.67 (s, 1H), 7.95 (t, 114), 7.76 (s, 114), 7.50 (d, 1H), 7.46 (d, 1H), 7.55 (t, 1H), 7.40 (t, 11-1), 7.20 (d, 11-1), 7.10 (d, 1H), 6.30 (s, 1H), 4.25 (s, 1H) ppm.
(0 3-Ethynylaniline gave /V4,N6-bis(3-ethynylphenyl)pyrimidine-4,6-diamine (I-11). MS (M
+ H+) 311, 312; 'H NMR (400 MHz, d6-DMS0) 69.24 (s, 2H), 8.32 (s, 1H), 7.78 (s, 2FI),
7.52 (d, 2H), 7.25 (t, 2H), 7.02 (d, 2H), 6.13 (s, 111), 4.12 (s, 2H) ppm.

(g) 4-Phenoxyaniline and 2,6-diaminopyridine gave 246-(4-phenoxypheny1)-aminopyrimidin-4-yl]arnino-6-aminopyridine (1R-11) MS (m/z): MH+ = 372, 371.
(h) 4-Phenoxyaniline and 1,4-diaminobenzene gave 446-(4-phenoxyphenyl)amino-pyrimidin-4-yl]amino-aminobenzene (IR-14) MS (m/z): MH+ ----- 370 SCHEME 2a Sequence A for Synthesis of 3-(6-mono- or disubstituted-amino)pyrinaidin-4-ylamino)-nhenylamines HN. Boc . Boc CI RN CO NR2 HN. Boc N= + E13N __ N)'`-' 40 Ring A-NH(R2) N.A1 kNr N 0.1r a Fi2 Et0H, 80 C Nr'N 120 C
Nrc_.2 NH2 50% TFA
__________________ NTh N N
[00259] Although a BOC protecting group is depicted in Scheme 2a above and the ensuing schemes below, one of ordinary skill in the art will recognize that other amine protecting groups are amenable for use in preparing compounds of the present invention.
Accordingly, a variety of amine protecting groups is contemplated.
1002601 Synthesis of tert-butyl 3-(6-chloropyrimidin-4-ylamino)phenylearbamate 4,6-Dichloropyrimidine (1.5 g, 10 mmol), tert-butyl 3-aminophenylcarbamate (2.1 g, 10 mmol) and triethylarnine (2.2 g, 20 mmol) were mixed in ethanol (20 mL), heated at 80 C
for 16 hr. The reaction mixture was cooled to RT and solvent was removed in vacuo. The gummy crude.
product was mixed with 20 mL of DCM and was stirred at RT to give an off-white solid, which was filtered and dried under vacuum (1.6 g, 5 mmol). MS (m/z): MH+ = 321, 323.
[00261] Synthesis of tert-butyl 3-(6-1N-methyl-N-phenylaminolpyrimidin-4-ylamino)pheny)-carbamate A mixture of tert-butyl 3-(6-chloropyrimidin-4-ylamino)phenylcarbamate (1.6g, 5 mmol) and N-methylaniline (1.07 g, 10 mmol) was heated at 120 C in a sealed tube for 2 hr. The reaction mixture was cooled to RT, mixed with 1 mL of IN

NaOH and 5 mL of DCM, stirred for 30 min, the product was filtered and dried under vacuum to give a solid product. MS (m/z): Mff+ = 392.
[00262] Synthesis of 3-f 6-(N-tnethyl-N-phenylamino)-pyrimidin-4-ylaminolphenylamine (IR-8) A mixture of tert-butyl 3-(64N-methyl-N-phenylarnino]pyrimidin-4-ylamino)phenylcarbamate (1.17 g, 3 mmol) and TFA (50% in DCM, 10 mL) was stirred at RT
for 4 hr. The reaction solvent was removed under vacuum. The crude product was mixed with 1 mL of 2N NaOH and 5 mL of Et0Ac, was stirred for 30 min, was filtered, and was dried under vacuum to give 0.65g (75%) of the title compound, an off-white solid. (IR-8) MS (m/z): ivar =
292.
[00263] The following compounds were prepared in a manner substiantially similar to that described in Scheme 2a:
(a) 4-Phenoxyphenyl amine gave 346-(4-phenoxyphenylamino)-pyrimidin-4-ylamino]phenylamine. (IR-7) MS (m/z): MH+ = 370.
(b) 3-Chloro-4-(2-pyridyl)methoxyaniline gave 3-116-(3-chloro-4-(2-pyridypmethoxyphenylamino)-pyrimidin-4-ylaminolphenylamine. (IR-9) MS (m/z):
MITF =
419, 421.
(c) 3 -Chl oro-4-(3-fluorob enzylo xy)aniline gave 3 -[6-(3 -chloro-4- {3-fluorobenzyloxy} phenylamino)-pyrimidin-4-ylaminol-phenyl amine. (IR-6) MS
(m/z): M+ =
436, 438.
(d) Aniline gave 3[6-(phenylarnino)-pyrimidin-4-yliaminophenylamine. (IR-15) MS (m/z):
MI-1+ = 278.

SCHEME 2b Sequence B for Synthesis of 3-(6-mono- or disubstituted-aminonyrimidin-4-yl)amino-phenvlamines NH- Boc 151 N, R2 14111 410 ,R2 HN-Bcc Rin A-NH R DIEA
( 2) , I el N CI Et0H or n-BuOH, 60-80 C N CI n-BuOH, 120-150 C N N

acidic deavage N N
[00264] Synthesis of 3-16-(4-bromo-2-fluorophenylamino)-pyrimidin-4-yl-aminol-phenylamine (IR-17). A solution of 4-bromo-2-fluoroaniline (701 mg, 3.69 mmol), diisopropylethylamine (0.70 mL, 4.03 mmol), and 4,6-dichloropyrimidine (500 mg, 3.36 mmol) in Et0H (10 mL) was heated in an 85 C oil bath for 5 d. Flash chromatography (2% Me0H /
CHC13) of the residue gave 380 mg (37%) of N-(4-bromo-2-fluoropheny1)-6-chloropyrimidin-4-amine as a pale yellow solid. MS (m/z): 304, 302.. A suspension of N-(4-bromo-fluoropheny1)-6-chloropyrimidin-4-amine (0.37 g, 1.22 mmol) and tert-butyl 3-aminophenylcarbamate (0.28 g, 1.35 mmol) in n-BuOH (4 mL) was heated in an oil bath at 120-130 C for 6 h. The reaction mixture was cooled and concentrated to give 0.68 g of a brown foam. Flash chromatography gave 0.16 g (28%) tert-butyl 3-(6-[4-bromo-2-fluorophenyliaminopyrimidin-4-yeaminophenylcarbamate as a white solid. MS
(m/z): (M + H) 476, 474. tert-Butyl 3-(6-[4-bromo-2-fluorophenyl]aminopyrimidin-4-yDaminophenylcarbamate (0.15 g, 0.32 mmol) was taken up in 4M HC1 in dioxane (5 mL). After several minutes a white solid began to precipitate. The mixture was allowed to stand for 3 h and was concentrated via rotary evaporation. Saturated aqueous sodium bicarbonate (5 mL) was added and the mixture was sonicated for several minutes. The solid was collected by filtration, was washed with water (5 mL), and was dried overnight under vacuum to give 0.12 g (100%) of 346-(4-bromo-2-fluorophenylamino)-pyrimidin-4-yl-amino]-phenylamine (I'-17) as a white powder. MS (m/z):
(M + H) 376, 374.
SCHEME 2c Synthesis of 3-(6-mono or disubstituted-aminopyrimidin-4-vflamino-N-substituted phenylamines CINHN-Bcic , R2 1) Li+ (TMS)2N", (CH3)3COCH3 ONR2 R2..N,Boc N, R2 R2.NH
2)R2I TFA, DCM
NrL N5 rµh 40 N N N N N N
[00265] Synthesis of tert-butyl N-3-(6-chloropyrimidin-4-ylamino)phenyl-N-methylcarbamate To a stirring solution of tert-butyl 3-(6-chloropyrimidin-4-ylamino)phenyl carbamate (2.000 g, 6.235 mmol) in 10 mL of THF at 0 C under N2 was added drop-wise a solution of 1.0 M lithium bis(trimethylsilyDamide in tert-butyl methyl ether (6.23 mL, 6.23 mmol). The light yellow solution was stirred at 0 C for 30 min then iodomethane (0.43 mL, 6.892 mmol, 1.1 eq) was added. The solution was allowed to slowly warm to room temperature overnight, was concentrated, and was then partitioned between Et0Ac and saturated ICH2PO4 solution. The organic extract was washed with brine solution, dried (MgSO4), was filtered, was concentrated in vacuo and was chromatographed (silica gel, 2% Me0H in CH2C12) to give 0.904 g of tert-butyl N-3-(6-chloropyrimidin-4-ylamino)phenyl-N-methylcarbamate as an off-white solid. MS (m/z) M+1 = 335/337 (100/44%), 1H NMR (CDC13) 8 8.48 (s, 1H), 7.49 (bs, 1H), 7.38 (m, 1H), 7.24 (m, 1H), 6.91 (m, 1H), 6.71 (bs, 1H), 6.30 (s, 1H), 3.48 (s, 3H), 1.53 (s, 9H).
[00266] Synthesis of N-346-(3-Chloro-4-fluorophenylamino)-pyrimidin-4-= ylamino]phenyl-N-methylamine (Int-C) A solution of tert-butyl N-3-(6-chloropyrimidin-4-ylamino)phenyl-N-methylcarbamate (0.486 g, 1.095 mmol) in 25 mL of CH2C12 was treated with trifluoroacetic acid (5 mL). The solution was stirred at room temperature under N2 for 2 h, was concentrated and was partitioned between CHC13 and 10% aq. NH4OH solution. The organic extract was washed with brine solution, was dried (MgSO4), was filtered and was concentrated to give 0.630 g of N-3- [6-(3 -chloro-4-fluorophenylamino)-pyrimidin-4-ylaminO]pheflYl-N-methylamine (Int-C) as yellow oil. MS (m/z): 344/346 (M+1, 100/68%). TLC
(Si02, 10%
Me0H in CHCI3): Rf 0.44.

SCHEME 3a Sequence B for Synthesis of 3-(6-mono- or disubstituted-aminopyrimidin-4-yl)amino-ohenylamines CI R2 0 R2 =

HN-jLL-Y
NC Y- -C(0)a L _______________________________________ N
kN N N N.PPP
wherein L' is a subset of L, as defined herein, such that Y-LIC(0)C1 results in formation of -provided compounds wherein RI is ¨L-Y wherein a terminal methylene unit of L
is replaced with ¨NHC(0)-.
[00267] Synthesis of N-1346-(3-bromophenylamino)-pyrimidin-4-ylaminol-pheny1}-propenamide (I-1) A solution of 346-(3-bromophenylamino)-pyrimidin-4-ylarnino]phenylamine (250 mg, 0.7 mmol) and triethylarnine (180 mg, 1.75 mmol) in 5 mL of THF was stirred at RT. Acryloyl chloride (80 mg, 0.9 mmol) was added into the reaction mixture and it was stirred at RI for lh. The solvent was removed by vacuum evaporation and the crude product was purified by flash chrornatography on silica gel with Et0Ac/DCM
solvent system to afford 115mg (40% yield) of the title compound as a light colored solid. MS
(rn/z): MH+ = 410, 412. JjjNMR (DMSO) : 10.15 (s, 111), 9.36 (s, 1H), 9.28 (s, 1H), 8.34 (s, 1H),
8.02 (s, 1H), 8.00 (s, 1H), 7.51-7.11 (m, 5H), 6.47 (dd, 1H, J1 = 10.1 Hz, J2 = 17.0 Hz), 6.27 (dd, 1H, .11 = 1.9 Hz, = 17.0 Hz), 6.20 (s, 1H), 5.76 (dd, 1H, J1= 10.1 Hz, J2 = 1.9 Hz) ppm.
[00268] The following compounds were prepared in a manner substantially similar to those described in Schemes 3a:
(a) 446-(3-Bromophenylamino)-pyrimidin-4-ylamino]phenylamine and acryoyl chloride gave N- {4-[6-(3 -bromoph enylamino)-pyrimid in-4-ylamino]ph eny1}-2-propenam i de (I-93). MS (m/z): MI-I' = 410, 412. 1H NMR (DMS0): 10.10 (s, 1H), 9.33 (s, 1H),
9.17 (s, 1H), 8.30 (s, 1H), 8.02 (s, 1H), 7.62 (m, 2H), 7.46 (m, 3H), 7.22 (t, 1H, J =
8.0 Hz), 7.10 (d, 1H, J = 8.0 Hz), 6.43 (dd, 1H, J1 = 10.0 Hz, J2 = 17.0 Hz), 6.24 (d, 1H, J
= 17.0 Hz), 6.13 (s, 1H), 5.73 (d, I H, J = 10.0 Hz) ppm.

(b) 316-(3-Bromophenylamino)-pyrimidin-4-ylaminolphenylamine and propionyl chloride gave N- {3-[6-(3-bromophenylarnino)-pyrirnidin-4-ylamino]phenyll-propionamide (IR-3).
MS (m/z): MH+ = 412, 414. '11 NMR (DMS0): 9.78 (s, 1H), 9.28 (s, 1H), 9.16 (s, 1H), 8.26 (s, 1H), 7.96 (s, 1H), 7.76 (s, 1H), 7.43 (d, 1H, J = 8.2 Hz), 7.20 (m, 2H), 7.05 (m, 3H), 6.12 (s, 1H), 2.27 (q, 2H, J = 7.6 Hz), 1.03 (t, 3H, J = 7.6 Hz) ppm.
(c) 346-(3-Bromophenylamino)-pyrimidin-4-ylamino]phenylamine and (E)-butenoylchloride gave (E)-N- (346-(3-bromophenylamino)-pyrimidin-4-ylamino]-pheny1}- 2-butenamide (1-8). MS (m/z): M+H+ = 424, 426. 'H NMR (DMS0): 9.87 (s, 1H), 9.29 (s, 1H), 9.18 (s, 1H ), 8.27 (s, 1H), 7.96 (s, 1H), 7.82 (s, 1H), 7.44 (d, 1H, J =
8.2 Hz) 7.20 (m, 4H), 7.05 (dd, 1H, J1 = 1.0 Hz, J2 = 7.8 Hz), 6.75 (m, 1H), 6.15 (m, 2H), 1.81 (d, 3H, J = 7.8 Hz) ppm.
(d) 316-(3-Chloro-4-fluorophenylamino)-pyrimidin-4-ylamino]phenylamine (IR-4) and acryoyl chloride gave N- {3-[6-(3-chloro-4-fluorophenylamino)-pyrimidin-4-ylamino]pheny1}- 2-propenamide (1-2). MS (m/z): MH+ = 384, 386. 'H NMR (DMS0):

9.29 (s, 1H), 9.20 (s, 1H), 8.27 (s, 1H), 7.93 (m, 1H), 7.86 (s, 1H), 7.41 (m, 1H), 7.25 (m, 5H), 6.42 (dd, 1H, Ji = 10.1 Hz, J2 = 17.0 Hz), 6.22 (dd, 1H, Ji = 1.9 Hz, J2 = 17.0 Hz), 6.09 (d, 1H, J1= 0.7 Hz), 5.69 (dd, 1H, J1= 10.1 Hz, J2 = 1.9 Hz) ppm.
(e)346-(3-Methylphenylamino)-pyrimidin-4-ylamino]phenylamine and acryolyl chloride gave N- {3- [6-(3-methylphenylarnino)-pyrirnidin-4-ylamino]pheny1}-2-propenamide (1-4). MS (rn/z): M+H+ = 346. 'H NMR (DMS0): 9.11 (s, 1H), 9.00 (s, 1H), 8.21 (s, 1H), 7.86 (d, 1H, J = 1.7 Hz), 7.31-7.05 (m, 7H), 6.74 (d, 1H, J = 7.6 Hz), 6.41 (dd, 1H, J1 =
10.0 Hz, J2 = 17.0 Hz), 6.20 (dd, 1H, Ji = 2.0 Hz, J2 = 17.0 Hz), 6.14 (s, 1H), 5.69 (dd, 1H, J1= 10.1 Hz, J2 = 2.0 Hz), 2.23 (s, 3H) PPm=
(f) 3- {6- [3 -Chloro-4-(3-fluorophenyl)methoxyphenylamino]-pyrmiidin-4-ylamino}phenylamine (IR-6) and acryolyl chloride gave N-{346-(3-chloro-4-(3-fluorophenypmetboxyphenylamino)-pyrimidin-4-ylarnino]phenyll-2-propenamide (II-6).
MS (m/z): M+H+ = 490, 492. 'H NMR (DMS0): 9.13 (s, 1H), 9.07 (s, 1H), 8.22 (s, 1H), 7.85 (d, 1H, J = 1.5 Hz), 7.40-7.10 (m, 10H), 6.41 (dd, I H, .11 = 10.0 Hz, J2 = 17.0 Hz), 6.20 (dd, 1H, JI = 2.0 Hz, J2 = 17.0 Hz), 6.05 (s, 1H), 5.70 (dd, 1H, .11=
10.1 Hz, 72 = 2.0 Hz), 5.14 (s, 2H) ppm.
(g) 4-Phenoxyphenyl amine gave 346-(4-phenoxyphenylamino)-pyrimidin-4-ylamino]phenylamine. (1R-7) and acryolyl chloride gave N-{34644-phenoxyphenylamino)-pyrimidin-4-ylamino]phenyl} -2 propenamide (1-13). MS
(tn/z):
MH+ = 424. 'H NMR (DMS0): 9.14 (s, 1H), 9.10 (s, 1H), 8.22 (s, 1H ), 7.89 (s, 1H), 7.52 (d, 2H, J = 9.0 Hz), 7.35 ¨ 6.92 (m, 11H), 6.42 (dd, 1H, J1 = 10.1 Hz, J2 = 16.9 Hz), 6.22 (dd, 1H, Ji = 1.9 Hz, J2 = 16.9 Hz), 6.12 (s, 1H), 5.70 (dd, 1H, J1 = 1.9 Hz, J2 =
10.1 Hz) ppm.
(h) 346-(N-methyl-N-phenylamino)-pyrimidin-4-ylamino]phenylamine (1R-8) and acryoyl chloride gave N- { 346-(N-methyl-N-phenylamino)-pyrimidin-4-ylamino]pheny11-2 propenamide (1-15): an off-white solid; 70mg; 20%; MS (m/z): MH+ = 346. 'H NMR

(DMS0): 10.02 (s, 1H), 9.00 (s, 1H), 8.25 (s, 1H ), 7.85 (s, 1H), 7.45 ¨ 7.12 (m, 6H), 6.45 (dd, 1H), 6.20 (d, 1H), 5.70 (m, 1H), 3.35 (s, 311) ppm.
(i) 3- ([6-(3-Chloro-4-(2-pyridyl)methoxyphenylamino)-pyrimidin-4-ylamino]phenylamine (IR-9) and acryloyl chloride gave N-(3-(6-( 3-chloro-4-(pyridin-2-ylmethoxy)phenylamino)pyrimidin-4-ylamino)pheny1)-2-propenamide (1-16). MS
(m/z):
MH+ = 473, 475(3:1). 'H NMR (DMS0): 10.10 (s, 1H), 9.17 (s, 1H), 9.08 (s, 1H
), 8.55 (m, 1H), 8.24 (s, 1H), 7.92 ¨ 7.73 (m, 4H), 7.57 (d, 1H), 7.38 ¨ 7.06 (m, 5H), 6.45 (dd, 1H), 6.23 (dd, 1H), 6.08 (s, 1H), 5.72 (dd, 1H), 5.20 (s, 2H) ppm.
(j) 346-(3-Chloro-4-fluorophenylamino)-pyrimidin-4-ylaminolphenylamine (IR-4) and 1-cyanocyclopropanecarbonyl chloride gave {3-chloro-4-fluorophenylamino }pyrimidin-4-Aamino]pheny1-1-cyanocyclopropanecarboxamide (I-47). MS (m/z): M+1 = 423/425, H NMR (DMSO-d6) 8 10.04 (s, 1H), 9.18 (s, 1H), 9.12 (s, 1H), 8.27 (d, 1H), 7.88 (s, 1H), 7.8 (s, 1H), 7.47-7.16 (m, 9H), 6.74 (b, 1H), 6.28 (d, 111), 6.1 (s, 1H), 5.19 (s, 2H), 3.05 (s, 211), 2.18 (s, 6H).
(k) 346-(3-Chloro-4-fluorophenylamino)-pyrirnidin-4-ylamino]phenylamine (IR-4) and chloroacetyl chloride gave 2-chloro-N-{346-(3-chloro-4-fluorophenylamino)-pyrimidin-4-ylaminol-phenyll-acetamide (1-49). MS (ES+): (M + 1)+ = 406 (100%), (M + 3)4-=

408 (75%). 11-1-NMR (DMSO-d6, 5 10.31 (s, 1H), 9.37 (s, 1H), 9.30 (s, 1H), 8.33 (s, 1H), 8.00 (m, 1H), 7.87 (s, 1H), 7.47-7.24 (m, 511), 6.15 (s, 1H), 4.26 (s, 2H).
(1) 3-[6-(3-Chloro-4-fluorophenypaminopyrimidin-4-yl]aminophenylamine (IR-4) and 2-chloropropionyl chloride gave 2-chloro-N-[3-(6- {3-chloro-4-fluorophenyl } aminopyrimidin-4-yDaminophenyl]propionamide (I-50). MS (ES): (M
+
1)+ = 420 (100%), (M + 3)4- = 422 (75%). 111-NMR (DMSO-d6, 5 10.32 (s, 1H), 9.37 (s, 1H), 9.28 (s, 1H), 8.33 (s, 1H), 8.00-7.98 (m, 1H), 7.87 (s, 1H), 7.47-7.26 (m, 5H), 6.14 (s, 1H), 4.69 (quartet, 1H), 1.61 (d, 311).
(m)346-(3-Chloro-4-fluorophenylamino)pyrimidin-4-ylamino]phenylarnine (IR-4) and 1-trifluoromethylcyclopropanecarbonyl chloride gave N-P-(6-{3-chloro-4-fluorophenyl}aminopyrimidin-4-yl)aminophenyli- 1-trifluoromethylcyclopropanecarboxamide (I-51). MS (ES) (m/z): 466/468 [M+1, 100/45%]. 11-1 NMR (DMSO-d6) 8 9.61 (m, 1H), 9.07-9.18 (m, 2H), 8.14 (m, 1H), 7.63-7.8 (m, 2H), 7.03-7.21 (m, 5H), 5.94 (bs, 1H), 1.12-1.27 (m, 4H).
(n) N-346-(3-Chloro-4-fluorophenylamino)-pyrimidin-4-ylamino]phenyl-N-methylamine (Int-D) and acryolyl chloride gave N-346-(3-chloro-4-fluorophenylamino)pyrimidin-4-yl]aminophenyl-N-methy1-2-propenamide (1-53). MS (m/z): 398/400 (M+1, 100/63%).
111 NMR (DMSO-d6) 5 10.32 (s, 1H), 9.21 (s, 1H), 8.33 (s, 1H), 7.99 (bs, 1H), 7.25-7.68 (m, 5H), 7.06 (bs, 1H), 6.41-6.47 (m, 1H), 6.26-6.29 (m, 1H), 5.71-5.80 (m, 2H).
(o) 3-[6-(4-Bromo-2-fluorophenypaminopyrimidin-4-yl]aminophenyl amine (1R.4 7)and acryloyl chloride gave N-346-(4-bromo-2-fluorophenylamino)pyrimidin-4-yl]aminopheny1]-2-propenamide (1-55). MS (M + H4) 430, 428. 'H NMR (d6-DMS0) 5 10.13 (s, 1H), 9.25 (s, 1H), 9.02 (s, 1H), 8.26 (s, 1H), 7.91 (m, 2H), 7.57 (d, J = 11 Hz, 111), 7.45-7.15 (m, 4H), 6.46 (dd, J = 12 and 10 Hz, 1H), 6.26 (d, J = 12 Hz, 1H), 6.23 (s, 111), 5.75 (d, J = 10 Hz, 1H).
(p) 246-(4-phenoxyphenyl)amino-pyrimidin-4-yl] amino-6-aminopyridine (IR41) and acryloyl chloride gave N-6-[6-(4-phenoxyphenypamino-pyrimidin-4-y1)]aminopyridin-2-ylpropenamide (1-63) 111 NMR (DMSO-d6) 8 ppm: 5.75 (d, J = 10.20 Hz, 1H), 6.29 (d, J
= 17.04 Hz, 111), 6.62 (dd, J = 10.08 & 16.92 Hz, 1H), 6.96-7.00 (m, 4H), 7.07-7.11 (m, 1H), 7.18 (bd, J= 3.28 Hz, 1H), 7.33-7.38 (m, 3H), 7.62-7.69 (m, 4H), 8.29 (s, I H), 9.13 (s, 1H), 9.66 (s, 1H), 10.15 (s, 1H); MS: m/z 425.3 (M+1).

(q) 2-[6-(3-methylphenoxy)-pyrimidin-4-yl]amino-6-aminopyridine (1R-12) and acryloyl chloride gave N-646-(3-methylphenoxy)-pyrimidin-4-y1)]aminopyridin-2-ylpropenamide (1-65) 11-1 NMR (CDC13) 8 ppm: 2.40 (s, 3H), 5.86 (d, J = 9.44 Hz, 1H), 6.49-6.61 (m, 2H), 6.83 (bs, 1H), 6.95-7.05 (m, 3H), 7.15 (d, J= 7.48 Hz, 1H), 7.34 (t, J =
15.08 Hz, 2H), 7.54 (bd, J= 5.56 Hz, 1H), 8.78 (s, 1H), 10.78 (s, 1H); LCMS: m/z 348.8 (M+1).
(r) 246-(4-phenoxyphenoxy)-pyrimidin-4-ylJamino-6-aminopyridine (I'-l3) and acryloyl chloride gave N-646-(4-phenoxyphenoxy)-pyrimidin-4-y1)]aminopyridin-2-yl}propenamide (1-66) 11-1 NMR (Me0D) 8 ppm: 5.92 (dd, J= 11.60, Hz, 1H), 6.50-6.54 (m, 2H), 6.75-7.28 (m, 10H), 7.37-7.41 (m, 2H), 7.91 (t, J= 16.08 Hz, 1H), 8.63 (s, 1H);
LCMS: m/z 426 (M+1).
(s) 3{6-(phenylamino)-pyrimidin-4-ylamino]phenylamine. (I'-l5) acryloyl chloride gave N-346-(phenylamino)-pyrimidin-4-yliaminophenylpropenamide (1-67) NMR
(DMSO-d6) 8 ppm: 5.72 (dd, J= 2.04 & 10.04 Hz, 1H), 6.19 (s, 1H), 6.25 (dd, J = 2 &
16.92 Hz, 1H), 6.46 (dd, J= 10.04 & 16.88 Hz, 1H), 6.96 (t, J= 7.36 Hz, 1H), 7.19-7.31 (m, 4H), 7.54 (d, J= 7.68 Hz, 2H), 7.91 (s, 1H), 8.26 (s, 1H), 9.13 (s, 1H), 9.18 (s, 1H), 10.11 (s, 1H); MS: m/z 332.8 (M+1).

SCHEME 4a Sequence A for Synthesis of N-3-(6-mono or disubstituted-aminouvrimidin-4-ynamino-N-mono-or unsubst ituted-pheny1-4-amino-subst ituted-2-butenami des 0. IL, .R2 R2.NH N.R2 R2.NY1N(R2)2 N*1 CICOCH=CHCH2N(R2)2 NjNop __________ NL:L1 N N

SCHEME 4b Sequence B for Synthesis of N-3-(6-mono or disubstituted-aminopyrimidin-4-ynamino-N-mono-or unsubstituted-phenyl-4-ammo-substituted-2-butenamides N-R2 R2, NH 0-N-R2 R2-N Br FiN(R2)2 0,N.R2 R2,Ni R-C(0)000CHHCH2Br id N 40 j, N N N N N N
[00269] Synthesis of (E)-N-(3-(6-(3-chloro-4-(pyridin-2-ylmethoxy)phenylamino)-pyrimidin-4-ylamino)pheny1)-4-(dimethylamino)but-2-enamide (1-19). 3- {[6-(3-chloro-4-(2-pyridyl)methoxyphenylamino)-pyrimidin-4-ylaminolphenylamine (IR-9) was dissolved in N-methylpyrrolidinone (1.2 mL) and added dropwise over 10 minutes to the ice-cold solution of (E)-4-(dimethylamino)but-2-enoyl chloride hydrochloride in acetonitrile. The reaction was stirred in an ice bath for 2 hr. To the mixture was added sodium bicarbonate to pH greater than 9. The oil that formed was extracted with Et0Ac (3 x 25 mL). A portion of the material was insoluble and set aside. The organic layer was dried (MgSO4) filtered and evaporated to a dark red oil. Both oils contained substantial amount of product as shown by TLC:
Si02 CHC13:Me0H/NH4OH (8:1) 19:1. The oils were combined and purified by flash column chromatography silica gel (25X300 mm) eluted first with 5% methanol/ammonium hydroxide 8/1 to remove non-polar impurities followed by 10% methanol/ammonium hydroxide 8/1 to elute 59 mg of product. The sample was further purified by a second flash column chromatography silica gel (25x250 mm) eluted with 10% methanol/ammonium hydroxide 8/1 to give the title compound (16.3 mg, 0.03 mmol, 6.4% yield). MS (ES+) 530(M+): 552, (M+Na); 11-1 NMR
(DMSO-d6, 500 MHz) 5 (ppm): 10.04 (s, 1H), 9.19 (s, 1H), 9.14(s, 1H), 8.60 (s, 1H), 8.27 (s, 1H), 7.88 (s, 2H), 7.57 (s, 1H), 7.36 (d, 1H, J = 7.4 Hz), 7.29 (d, 2H, J =
7.8 Hz), 7.22 (d, 2H, J
= 7.9 Hz), 7.18 (m, 2H), 6.74 (m, 1H), 6.30 (d, 1H, J = 15 Hz) 6.10 (s, 1H), 5.24 (s, 214), 3.06 (s, 2H), 2.18 (s, 6H) ppm; HPLC: tR = 5.15 min, 97.5% (YMC-Pack ODS-A 4.6x100 mm, 80%
water/20% acetonitrile to 5% water/95% acetonitrile over 5.5 min, hold to 9 min.).
[00270] Synthesis of (E)-N-3-(643-methylphenyllaminopyritnidin-4-Aarninophenyl-bromo-2-butenamide (Int-D) To a stirring solution of 4-bromo-but-2-enoic acid (0.72 g) at 0 C under nitrogen atmosphere and triethylamine (0.61 mL, 4.38 mmol) in 5 nil.
of THF was added iso-butyl chloroformate (0.56 mL, 4.32 mmol). The mixture was stirred for 15 min followed by dropwise addition of a solution of N-(3-amino-pheny1)-N'-3-methylphenylarnino-pyrimidine-4,6-diamine (1.015 g, 3.483 mmol) in 50 mL of THF. The reaction was allowed to warm to room temperature overnight. The sample was concentrated then partitioned between Et0Ac and sat. NaHCO3 solution. The organic extract was washed with brine solution, dried (MgSO4), filtered and was concentrated. The resultant brown foamy solid was washed with diethyl ether and vacuum dried to give 0.960 g of crude (E)-N-3-(643-methylphenyl]aminopyrimidin-4-yDaminopheny1-4-bromo-2-butenamide (Int-D) as brick-brown solid. MS (m/z): (M+1) 438/440 (71/75%).
[002711 Synthesis of (E)-N-3-(643-methylphenyllaminopyrimidin-4-yl)aminophenyl-(methyl-prop-2-ynyl)amino-2-butenatnide (1-58) To a stirring solution at 0 C
under N2 of N-[3-(6- {3-methylphenyl}amino-pyrimidin-4-ylamino)-4-bromo-2-butenamide (Int-D) (0.492 g, 1.122 mmol) and triethylamine (0.20 mL, 1.44 mmol) in 10 mL of THF was added (via syringe) N-methyl-propargylamine (0.11 mL, 1.173 mmol). The solution was allowed to warm to room temperature overnight, was concentrated and was then partitioned between Et0Ac and sat.
NaHCO3 solution. The organic extract was washed with brine solution, was dried (MgSO4), was filtered and was concentrated. The residue was chromatographed (silica gel, 10% Me0H in CHCI3) to give 0.110 g of (E)-N-3-(6-[3-methylphenyl]aminopyrimidin-4-yDaminophenyl-4-(methyl-prop-2-ynyl)amino-2-butenamide (1-58). MS (APCI) m/z 427 (M+1, 100%).

(DMSO-d6) 8 10.06 (s, 1H), 9.07 (s, IH), 9.17 (s, 1H), 8.27 (s, 1H), 7.90 (s, IH), 7.16-7.36 (m, 6H), 6.70-6.73 (m, 1H), 6.79 (d, 1H), 6.20 (s, IH), 6.32 (d, IH), 3.30-3.49 (m), 3.14-3.27 (m, 3H), 2.18-2.39 (m, 7H which contain singlets at 62.25 [3H] and 62.29 [3H]).
[00272) Synthesis of (E)-N-3-(6-13-methylphenyl]aminopyrimidin-4-yl)aminophenyl-4-piperaziny1-2-butenamide (1-57) To a stirring solution at 0 C under N2 of N-P-(6-{3-methylphenyl}amino-pyrimidin-4-ylamino)-4-bromo-2-butenamide (Int-D) (2.15 g, 4.91 mmol) and triethylamine (0.86 mL, 6.17 mmol) in 10 ad, of TI-IF was added dropwise a solution of 1-Boc-piperazine (0.92 g, 4.91 mmol) in 10 mL of THF. The solution was allowed to warm to room temperature overnight, was concentrated and was then partitioned between Et0Ae and sat.
NaHCO3 solution. The organic extract was washed with brine solution, was dried (MgSO4), was filtered and was concentrated to give 2.76 g of 4- [343-(6-{3-methylphenyl}amino-pyrimidin-4-ylamino)-phenylcarbamoylkally1}-piperazine-l-carboxylic acid ter/-butyl ester as gummy brown solid. This material was dissolved in 100 mL of CH2C12. 20 mL of trifluoroacetic acid was added and the mixture was stirred at room temperature under N2 for 2 h. The mixture was concentrated in vacuo, was basified with sat. NaHCO3 solution and was extracted with Et0Ac (2x 100 ml). The combined organic extract was dried (MgSO4), was filtered and was concentrated in vacuo. The residue was chromatographed (silica gel, 5% Me0H in CHC13 [500 la] then 1% NH4OH-10% Me0H in CHC13) to give 0.404 g of (E)-N-3-(643-methylphenyl]aminopyrimidin-4-yl)aminopheny1-4-piperaziny1-2-butenamide. (1-57) MS (m/z):
444 (M+1, 100%). IHNMR (DMSO-d6) 5 10.03 (s, 1H), 9.17 (s, 1H), 9.07 (s, 1H), 8.27 (s, 1H), 7.90 (s, 111), 7.17-7.36 (m, 6H), 6.79-6.80 (d, 1H), 6.72-6.75 (m, 1H), 6.29 (d, 1H), 6.20 (s, 1H), 3.08-3.39 (m, containing water and ¨3H), 2.70-2.72 (m, 4H), 2.30-2.42 (m, 4H), 2.29 (s, 3H.
[00273] The following compounds were prepared in a manner substantially similar to those described in Scheme 4a, above:
(a) 3-(6-[3-chloro-4-{3-fluorobenzyloxy}iphenylarnino-pyrimidin-4-yDaminophenylamine (IR-6) and 4-dimethylarnino-2-butenoyl chloride gave (E)-N-3-([6-(3-chloro-4-{3-fluorobenzyloxy}phenylarnino)-pyrimidin-4-yDaminopheny1-4-(dimethylamino)but-2-enamide (1-46). MS (m/z): IN/r = 547, 549(3:1), H-NMR (DMSO-d6) 5 10.04 (s, 1H), 9.18 (s, 1H), 9.12 (s, 1H), 8.27 (d, 1H), 7.88 (s, 1H), 7.8 (s, 1H), 7.47-7.16 (m, 9H), 6.74 (b, 1H), 6.28 (d, 1H), 6.1 (s, 1H), 5.19 (s, 2H), 3.05 (s, 2H), 2.18 (s, 6H).
(b) 346-(3-methylphenylamino)-pyrimidin-4-ylamino]phenylamine (1R-5) and (dimethylamino)-2-butenoyl chloride gave (E)-N-{346-(3-methylphenylamino)-pyrimidin-4-ylamino]pheny1}-4-dimethylamino-2-butenamide (1-17). MS (m/z): MH+
=
403.
(c) 346-(3-chloro-4-fluorophenylamino)-pyrimidin-4-ylannino]phenylamine (IR-4) and 4-(dimethylamino)-2-buteno yl chloride gave (E)-N-{346-(3-chloro-4-fluorophenylarnino)-pyrimidin-4-ylamino]pheny1}-4-dimethylamino-2-butenamide (1-18). MS (M + H+) 441, 443; IHNMR (400 MHz, d6-DMS0) 5 10.01 (s, 1H), 9.30 (s, 1H), 9.20 (s, 1H), 8.28 (s, 1H), 7.95 (dd, J = 7 and 3 Hz, 1H), 7.86 (s, 1H), 7.41 (m, 1H), 7.35-7.10 (m, 3H), 6.69 (dt, J = 15 and 5 Hz, 1 H), 6.26 (d, J = 15 Hz, 1H), 6.11 (s, 1H), 3.02 (d, J
= 5 Hz, 2H), 2.14 (s, 6H) ppm.
(d) N-346-(3-chloro-4-fluorophenylamino)-pyrimidin-4-ylaminolphenyl-N-methylamine (Int-C) and 4-dimethylamino-2-butenoyl chloride gave (E)-N-(346-(3-chloro-4-fluorophenylamino)-pyrimidin-4-ylamino]phenyl-N-methy1-4-(dimethylamino)but-2-enamide (1-48) MS (ES) (m/z): 455/457 (M+1, 37/13%) and 228 (100%). NMR
(DMSO-d6) 8 10.22 (s, IH), 9.20 (s, 1H), 8.33 (s, 1H), 7.99 (bs, 1H), 7.25-7.68 (m, 5H), 7.05 (bs, 1H), 6.73-6.76 (m, 1H), 6.26-6.29 (m, 1H), 5.71 (s, IH), 3.06 (bs, 2H), 1.99 (s, 6H).
(e) 346-(4-phenoxyphenylamino)-pyrimidin-4-yllaminophenylamine (IR-7) and 4-dimethylamino-2-butenoyl chloride gave (E)-N-346-(4-phenoxyphenyparnino-pyrimidin-4-yl]aminopheny1-4-(dimethylamino)but-2-enamide (1-62) NMR
(DMSO-d6) 8 ppm: 2.19 (s, 6H), 3.07 (d, J= 5.36 Hz, 2H), 6.16 (s, 1H), 6.29 (d, J =
15.4 Hz, 1H), 6.68-6.75 (m, 1H), 6.95-6.98 (m, 4H), 7.06-7.10 (m, 1H), 7.20 (dd, 1=7.88 &
8.12 Hz, 1H), 7.24 (d, J= 8.32 Hz, 2H), 7.36 (dd, J = 7.52 & 7.84 Hz, 2H), 7.55 (d, J=
8.76 Hz, 2H), 7.90 (s, 1H), 8.24 (s, 1H), 9.15 (d, 1= 8.84 Hz, 2H), 10.04 (s, 1H);
LCMS: m/z 481 (M+1).

SCHEME 5a Synthesis of N- {3[6-(arylamino)-pvrimidin-4-ylaminol-phenyll-ethensulfonamides N' R2 NH2R 0"e N:11 LY
k N N
1002741 wherein L' is a subset of L, as defined herein, such that Y-L'SO2C1 results in formation of provided compounds wherein RI is ¨L-Y wherein a terminal methylene unit of L is replaced with ¨NHS02-=
[00275] Synthesis of N-13-16-(3-ehloro-4-fluorophenylamino)-pyrimidin-4-ylaminolpheny1)- ethenesulfonamide (1-3) A solution of 346-(3-chloro-4-fluorophenylamino)-pyrimidin-4-ylamino]phenylamine (1R-4) (300 mg, 0.9 mmol) and triethylamine (500 mg, 5 mmol) in 10 mL of THF was stirred at RT. 2-Chloroethanesulfonyl chloride (360 mg, 2.25 mmol) was added into the reaction mixture and stirring was continued at RT for 1 hr. The crude product was purified by flash chromatography on silica gel with Et0Ac/heptane solvent system to afford 35 mg (9%) of the title compound, a brown colored solid. MS (m/z):
MB+ = 420, 422.

NMR (DMS0): 9.92 (s, 1H), 9.29 (s, 111), 9.21 (s, 1H), 8.26 (s, 1H), 7.92 (m, 111), 7.40-7.22 (m, 4H), 7.14 (m, 1H), 6.20 (m, 211), 6.05 (m, 3H) ppm..
[00276] The following compounds were prepared in a manner substantially similar to Schemes 5a:
(a) 3- { 6- [3-chloro-4-(3-fluorophenyl)methoxyphenylamino]-pyrimidin-4-ylamino}phenylamine (IR-6) gave N-{3-[6-(3-chloro-4-(3-fluorophenyl)methoxy-phenylamino)-pyrimidin-4-ylamino]pheny1}- ethenesulfonamide (I-7). MS (m/z):
M+H+
= 526, 528 (2:1). MS (m/z): M+H+ = 490, 492 (2:1). NMR
(DMS0): 9.91 (s, 1H), 9.16 (s, 1H), 9.07 (s, 1H), 8.22 (s, 1H), 7.73 (s, 1H), 7.40-7.10 (m, 9H), 6.70 (m, 2H), 6.12 (d, 1H, J = 17.0 Hz), 6.02 (m, 2H), 5.14 (s, 2H) ppm.
(b) 346-(3-methylphenylamino)-pyrimidin-4-ylamino]phenylamine (IR-5) gave N-{34643-methylphenylamino)-pyrimidin-4-ylamino]pheny1}- ethenesulfonamide (II-5). MS
(m/z):
M+H+ = 382. 11-1 NMR (DMS0): 9.90 (s, 111), 9.12 (s, 111), 9.00 (s, 1H), 8.21 (s, 1H), 7.37 (d, 1H, J = 1.7 Hz), 7.26 (m, 3H), 7.13 (m, 2H), 6.70 (m, 3H), 6.13 (m, 2H), 6.00 (d, 111, J = 10.0 Hz), 2.24 (s, 3H) ppm.

SCHEME 6a Synthesis of N2-acylated 6-(6-mono or disubstituted-aminopyrimidin-4-ynamino-2-aminopyridines NH2 4:11 2 Et3N _Ring A-NH(R2) rea.
N CI H2N Et0H, 80 C N N 120 C N N

NR2 HN 1_=-Y
Y-C-C(0 )CI N = ki NL
N N
[00277] Synthesis of N-(6-chloro-pyrimidin-4-y1)-pyridine-2,6-diamine A
mixture of 2,6-diaminopyridine (1.530 g, 14.020 mmol) and 4,6-dichloropyrimidine (2.610 g, 17.519 mmol) in 15 mL of n-butanol in a sealed vial was heated at 100 C for 72 h. The dark brown sample was cooled, was concentrated to remove most of the n-butanol, and was then partitioned between Et0Ac and sat. NaHCO3 solution. An emulsion formed, the sample was filtered through a pad of Celite and the layers were separated. The organic extract was washed with sat. KH2PO4 and brine solutions, was dried (MgSO4), was filtered and was concentrated to brown oily-solid. The sample was suspended into 50 nal, of CH2C12, was cooled and was filtered to give 1.017 g of N-(6-chloro-pyrimidin-4-y1)-pyridine-2,6-diamine as yellow-orange solid. MS (ES) (m/z) 222/224 (M+1, 100/63%). TLC (Si02, 50% EiOAc in hexanes): Rf 0.33.
[00278] Synthesis of N-(6-amino-pyridin-2-yI)-N'-(3-ehloro-4-fluoropheny1)-pyrimidine-4,6-diamine (Int-E) A mixture of N-(6-chloro-pyrimidin-4-y1)-pyridine-2,6-diamine (1.000 g, 4.512 mmol) and 3-chloro-4-fluoroaniline (1.380 mmol) in 10 inL of n-butanol in a sealed vial was heated at 120 C for 24 h. The sample was cooled, was concentrated to remove most of the n-butanol, and was then diluted with Et0Ac and sat. NaHCO3 solution. The sample was stirred at room temperature for 30 mm and was filtered. The solid was washed with fresh Et0Ac and water and vacuum dried to give 1.063 g of N-(6-amino-pyridin-2-y1)-N'-(3-chloro-4-fluoropheny1)-pyrimidine-4,6-diamine (Int-E) as tan solid. MS (ES) (m/z) 331/333 (M+1, 100/65%). TLC (Si02, 10% Me0H in CHC13): Rf 0.25.
[00279] Synthesis of (E)-N-[6-(3-chloro-4-fluorophenylamino)pyrimidin-4-ylaminopyridin-2-y11-4-(dimethylamino)but-2-enamide (1-52).
To a stirring suspension at 0 C under N2 of 4-dimethylamino-but-2-enoic acid, hydrochloride (0.500 g, 3.019 mmol) in 15 mL of THF containing 5 drops of DMF was added drop-wise (via syringe) oxalyl chloride (0.28 mL, 3.210 mmol). Gas formation started immediately. The sample was stirred at 0 C for ¨30 mm, room temperature for ¨2 h, re-cooled to 0 C, then treated with drop-wise addition of a solution of N-(6-amino-pyridin-2-y1)-AP-(3-chloro-4-fluoro-pheny1)-pyrimidine-4,6-diamine (Int-E) (0.500 g, 1.512 mmol) in 15 mL of THF
and 3 mL of NMP. The ice-bath was removed, the sample was stirred at room temperature for 2 h then was partitioned between Et0Ac and sat. NaHCO3 solution. The organic extract was washed with brine solution, dried (MgSO4), filtered and concentrated to a yellow solid.
The solid was suspended into Et0Ac (-25 mL), stirred at room temperature for ¨ 12 h, filtered and vacuum dried to give 0.459 g (69%) of (E)-N-[6-(3-chloro-4-fluorophenylamino)pyrimidin-4-ylaminopyridin-2-y1]-4-(dimethylamino)but-2-enamide (1-52) as light yellow solid. MS (m/z):
442/444 (M+1, 100/37%). 11-1 NMR (DMSO-d6) 8 10.19 (s, 1H), 9.78 (s, 1H), 9.43 (s, 1H), 8.36 (s, 1H), 8.05-8.07 (m, 1H), 7.54-7.72 (m, 4H), 7.32-7.35 (m, 1H), 7.10 (bs, 1H), 6.79-6.82 (m, 1H), 6.54-6.57 (m, 1H), 3.14 (bs, 2H), 2.23 (bs, 6H).

SCHEME 7a Synthesis o f N-acvlated 3-(6-mono or disubstituted-aminonvrimidin-4-yDamino-mono-substituted-phenylamines OR2 0, R2 NO2 I R x Ring A-NI DIEA
Nj.= H2N
I Nei R.
N CI Et0H or n-BuOH, 60-80 C N CI n-BuOH, 120-150 C N N

Y-1:-C(0)C1 reduction N N N N
[00280] Synthesis of N-(5-Amino-2-methylpheny1)-N'-(3-chloro-4-fluoropheny1)-pyrimidine-4,6-diamine (Int-F) A mixture of 4-(3-chloro-4-fluoropheny1)-6-chloropyrimidin-4-ylamine (IR-4) (1.2 g, 4.6 mmol), 2-methyl-5-nitroaniline (0.85 g, 5.5 mmol) and 1 mL of concentrated HC1 in 10 nit_ of n-butanol was heated at 120 C for 16 h. With stirring, 5 mL of Et0Ac was added to the reaction mixture. The light yellow colored product that precipitated was filtered and dried under vacuum to give N-(3-chloro-4-fluoropheny1)-N-(2-methy1-5-aitrophenyl)pyrimidine-4,6-diamine. MS (APCI) (m/z): 374/376 (M+1). A mixture of N-(3-chloro-4-fluoropheny1)-N-(2-methy1-5-nitrophenyppyrimidine-4,6-diamine (0.55 g, 1.5 mmol) and iron powder (35 mesh, 0.5 g, 5 eq) in 5 mL of HOAc and 2 mL of Me0H was heated at reflux for 2 h. The solvent was removed in vacuo and the dark colored residue was mixed with 150 mL of CH2C12 and 15 mi. of sat. K2CO3 solution and was stirred at room temperature for 30 min. The organic layer was dried (MgSO4), was concentrated, and was then purified by flash chromatography (silica gel, Me0H/NH4OH/CH2C12) to afford 0.115 g of N-(5-amino-inethylpheny1)-N-(3-chloro-4-fluorophenyppyrirnidine-4,6-diamine as a light colored solid.
(Int-F) MS (m/z): 344/346 (M+1).

=
[002811 In a manner substantially similar to that described above 4-(3-chloro-fluoropheny1amino)-6-ch1oropyrimidine and 2-methoxy-5-nitroaniline gave N-(5-amino-2-methoxypheny1)-/V-(3-chloro-4-fluorophenyl)pyrimidine-4,6-diamine. (In t-G) MS
(m/z):
360/362 (M+1).
[002821 Synthesis of (E)-N-13-(6-13-Chloro-4-fluorophenyl)amino-pyrimidin-4-ylamino)-4-methylpheny11-4-(dimethylamino)but-2-enamide (I-54) N-(5-amino-2-methylpheny1)-N-(3-chloro-4-fluorophenyl)pyrimidine-4,6-diarnine (Int-F) and 4-dirnethylamino-2-butenoyl chloride were combined in a manner similar to that described in Scheme 4a to give (E)-N-(3-(6-{3-chloro-4-fluorophenyl}amino-pyrirnidin-4-ylamino)-4-methylpheny1]-4-(dimethylarnino)but-2-enamide (1-54). MS (m/z): 455/457 (M4-1, 100/39%). NMR (DMSO-d6) 8 11.02 (bs, 1H), 10.66 (bs, 1H), 9.92 (bs, IH), 9.39 (bs, 1H), 7.92-7.94 (m, 1H), 7.75 (bs, 1H), 7.27-7.56 (m, 4H), .6.80 (in, 1H), 6.54 (d, 1H), 5.89 (bs, 1H), 3.92 (bs, 2H), 2.75 (bs, 6H), 2.17 (bs, 3H).
1002831 In a manner substantially similar to that described for the synthesis of (I-54) N-(5-Arnino-2-methoxypheny1)-N'-(3-chloro-4-fluorophenyl)pyrimidine-4,6-diamine (mnt-G) and 4-dimethylamino-2-butenoyl chloride gave (E)-N-13-(643-chloro-4-fluoropheny1)-aminopyrimidin-4-ylamino-4-methoxyphenyl]-4-(dimethylarnino)but-2-enamide (1-59). MS
(rn/z): 471/473 (M+1, 100/41%). 11-1 NMIZ (DMSO-d6) 8 9.97 (s, 1H), 9.28 (s, 1H), 8.46 (s, 1H), 8.26 (s, 1H), 7.93-7.98 (m, 2H), 7.44-7.51 (m, 2H), 7.30-7.33 (m, 1H), 7.02 (d, 1H), 6.69-6.72 (m, 1H), 6.26 (d, 1H), 6.03 (s, 1H), 3.80 (s, 3H), 3.05 (m, 214), 2.17 (s, 6H).

[00284] Synthesis of 2-[6-(3-methylphenoxy)-pyrimidin-4-yllamino-6-aminopyridine (141-12) A mixture of 2,6-diaminopyridine (1.530 g, 14.020 mmol) and 4,6-dichloropyrimidine (2.610 g, 17.519 mmol) in 15 mL of n-butanol in a sealed vial was heated at 100 oC for 72 h. The dark brown sample was cooled and was concentrated at reduced pressure to remove most of the n-butanol. The residue was then partitioned between Et0Ac and saturated. NaHCO3 solution. An emulsion formed, the sample was filtered through a pad of CelitTemand the layers were separated.
The organic extract was washed with sat .K.H2PO4 and brine solutions, dried (Mg SO4), filtered and concentrated to brown oily-solid. The sample was suspended into -50 rni, of CH2C12, cooled and filtered to give 1.017 g (36%) of N-(6-ebloro-pyrirnidin-4-yI)-pyridine-2,6-diamine as yellow-orange solid. MS: m/z 222/224 (M+1, 100/63%). To a solution of N-(6-chloro-pyrimidin-4-y1)-pyridine-2,6-diamine (100 mg, 0.45 mmol) in DMF (1 mL) was added 3-methylphenol (88 mg, 0.8 mmol) and anhydrous K2CO3 (93 mg, 0.6 mmol). The reaction mixture was heated at 145 C for 16 h. It was then cooled and DMF was removed under reduced pressure to get a yellow gummy residue. The residue was taken in Et0Ac (20 mL), was washed sequentially with water (5 mL) and brine (5 mL) and was dried over Na2SO4.
Filtration followed by concentration under reduced pressure afforded a yellow gum which was further purified by column chromatography (Si02, 60-120, Et0Ac/hexane, 5/5) to give 100 mg of methylphenoxy)-pyrimidin-4-yl]amino-6-aminopyridine (IR-12) as a pale yellow solid. MS: m/z 294 (M+H) [00285] The following compounds were prepared in a manner substantially similar to the procedure described above:
(a) 4-Phenoxyphenol and 2,6-diaminopyridime gave 246-(4-phenoxyphenoxy)-pyrimidin-4-yl]amino-6-aminopyridine (I1t-13) MS (m/z): MH+ = 372.
(b) 4-nitrophenol and 1,3-diaminobenzene gave 3-[6-(4-nitrophenoxy)-pyrimidin-yl]amino-aminobenzene (IR-16) MS (m/z): MH+ = 324 [00286] Synthesis of N-3-[6-(3-ethynylphenylamino)-pyrimidin-4-yl]aminopheny1-propenamide (1-68) To a stirred solution of IR-1 (150 mg, 0.42 mmol) in .dry DMF (4.0 mL) under N2 was added Pd(PPh3)2C12 (14.7 mg, 0.021 mmol), CuI (3.9 mg, 0.021 mmol), PPh3 (22.07 mg, 0.08 =top, and diethylamine (94.8 mg, 6.3 mmol). The reaction mixture was purged with N2 for additional 10 min, trimethylsilylacetylene (45.5 mg, 0.46 mmol) was added, and it was then subjected to microwave irradiation at 120 C for 30 min. The mixture was cooled, was diluted with 5 mL water, was filtered through celite and was extracted with Et0Ac (2x10 mL). The combined Et0Ac extract was washed with water, then with brine and was dried over Na2SO4. Concentration under reduced pressure gave a crude product that was purified by column chromatography (Si02, Me0H/CHC13, 1/99) to give 100 mg of a brown solid. A solution of this material in 2 mi.. of dry methanol under nitrogen atmosphere containing anhydrous K2CO3 (73.9 mg, 0.53 mmol) was stirred at rt for 16 h. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure to give a residue, which was further purified by column chromatography (Si02, 230-400, MeOHJCHC13, 1/99) to give 70 mg of a light brown solid. To a stirred solution of this material in NMP (1.0 mL) at 0 C was added acryloyl chloride (105 mg, 1.16 mmol), and the reaction mixture was stirred at 0 C for 1 h. The reaction mixture was then quenched with water, was basified with 10% NaHCO3 sol and was extracted with Et0Ac. The combined Et0Ac extract was washed sequentially with water and brine, was dried over Na2SO4 and was concentrated under reduced pressure. The residue obtained was further purified by preparative HPLC to give 8 mg of N-346-(3-ethynylphenypaminopyrimidin-4-yl]amino-pheny1-2-propenamide (1-68) as an off white solid. 111 NMR (DMSO-d6) 8 ppm: 4.15 (s, 1H), 5.73-5.76 (m, 111), 6.19 (s, 1H), 6.25 (dd, J= 1.96 & 17.00 Hz, 2H), 6.46 (dd, J= 10.2 &
16.96 Hz, 1H), 7.05 (d, J = 7.64 Hz, 1H), 7.21-7.33 (m, 3H), 7.54 (d, J = 7.96 Hz, 1H), 7.81 (s, 1H), 7.91 (s, 1H), 8.32 (s, 1H), 9.28 (d, J= 11.08 Hz, 2H), 10.13 (s, 1H); MS: m/z 356.8 (M+1).

[00287] Synthesis of N-3-16-(4-14-114-chloro-3-(trifluoromethyl)phenyllaminol-earbonyl]amino)phenoxy pyrimidin-4-yllaminophenylchloroacetamide (1-69) [00288] Stepl: To a solution of 346-(4-nitrophenoxy)-pyrimidin-4-yliamino-aminobenzene (IIR-16) (650 mg, 2.01 mmol) in THF (10 mL) was added Et3N (305 mg, 3.01 mmol) and (Boc)20 (525 mg, 2.4 mmol) under N2 atmosphere. The reaction mixture was further heated at 60 C for 16 h. A residue was obtained after cooling to room temperature and removal of solvent under vacuum. The residue was dissolved in Et0Ac (10 mL). The Et0Ac extract was washed sequentially with water (5 mL) and brine (2 mL), was dried over Na2SO4.
Concentration under reduced pressure followed by purification by column chromatography (Si02, 60-120, CHC13/Me0H, 9/1) gave 400 mg of the Boc derivative as a yellow solid.
1002891 Step 2: The material from Step 1 was dissolved in Me0H (8 mL) and 10%
Pd/C (40 mg) was added under N2 atmosphere. The reaction mixture was hydrogenated in a Parr apparatus (1-Y,2, 3 Kg, rt, 16 h). The reaction mixture was filtered through celite and the solvent was removed under reduced pressure to give 250 mg, of the amine as a yellow solid.
1002901 Step 3: To the material from Step 2 was added a toluene solution of 4-chloro-3-trifluoromethylphenylisocyanate prepared by reacting under nitrogen atmosphere at 0 C 24 mg of 4-chloro-3-trifluoromethylaniline in 10 mL toluene with 0.08 mL of a 20%-solution of phosgene in toluene followed by addition of Et3N (0.07 mL) and at 110 C for 16 h. The reaction mixture of the amine and the isocyanate was further heated at 110 C for 4 h and then was quenched with water (1 mL) and was extracted with Et0Ac (2x20 mL). The Et0Ac extract was washed with water (5 mL), brine (2 mL) and dried over Na2SO4. Filtration followed by concentration under vacuum offered a residue which was purified by a column chromatography (Si02, 230-400, hexane/Et0Ac, 9/1) to 20 mg of the Boc/urea intermediate as a yellow solid.
[00291] Step 4: To a solution of 45 mg of this intermediate in CH2C12 (2 mL) at 0 C was added TFA (0.01 mL) under N2 atmosphere. The reaction mixture stirred at room temperature for 4 h, was washed sequentially with 10% NaHCO3 solution (1 mL) and brine (1 mL) and was dried over Na2SO4. Concentration under reduced pressure gave 25 mg of the amine/urea intermediate as a brownish solid.
[00292] Step 5: To a solution of the amine/urea intermediate from Step 4 (45 mg, 0.05 mmol), in THF (2 mL) and Et3N (10 mg, 0.1 mmol) at 0 C under N2 atmosphere was added chloroacetyl chloride (55 mg, 0.1 mmol). The reaction mixture was allowed to come to rt and with stirring for 2 h. The reaction mixture was concentrated under reduced pressure and the residue was partitioned between Et0Ac (4 mL) and water (1 mL). The Et0Ac layer was separated, was washed with brine (2 mL) and was dried over Na2SO4. Filtration followed by concentration under reduced pressure gave a residue which was further purified by column chromatography (Si02, 230-400, CHC13/Me0H, 9/1) to give N-346-(444-[[[[4-chloro-3-(trifluoromethyl)phenyliamino]carbonyl]aminolphenoxy]phenyljamino-pyrirnidin-4-yl]aminophenylchloroacetamide (1-69) as a pale yellow solid. 11-1 NMR (Me0D) 8 ppm: 4:19 (s, 2H), 6.61 (s, 1H), 7.13 (d, J= 6.92 Hz, 211), 7.14-7.23 (m, 3H), 7.50-7.55 (m, 3H), 7.63-7.66 (m, 111), 7.95 (s, 1H), 8.00 (s, 1H), 8.30 (s, 1H); MS: m/z 593 (M+1).

1002931 Synthesis of (E)-N-3-(6-13-methylphenylaminol-pyrimidin-4-yDaminophenyl-4-(4-acetylpiperazin-ly1)-2-butenamide (1-60) To a stirring solution at 0 C under N2 of (E)-N-3-(643-methylphenyl]aminopyrimidin-4-ypaminopheny1-4-piperaziny1-2-butenamide (1-57) (0.291 g, 0.655 mmol) and triethylamine (0.14 mL, 1.004 mmol) in 10 mL of THF was added (via syringe) acetyl chloride (0.05 mL, 0.70 mrnol). The sample was allowed to warm to room temperature overnight, was concentrated and was then partitioned between Et0Ac and sat. NaHCO3 solution. The organic extract was washed with brine solution, was dried (MgSO4), was filtered, was concentrated and was ehromatographed (silica gel, 1% NH4OH-10% Me0H in CHC13) to give 0.0705 g of (E)-N-3-(6-[3-methylphenyl]aminopyrimidin-4-yDaminopheny1-4-(4-acetylpiperazin-ly1)-2-butenamide (I-60), a white solid. 1H NMR (DMSO-45 8 2.00 (s, 3H), 2.29 (s, 3H), 2.35-2.41 (m, 4H), 3.15-3.16 (m, 2H), 3.31-3.46 (m, containing water and ¨ 4H), 6.19 (s, 1H), 6.32 (d, 111), 6.73-6.80 (m, 21-1), 7.16-7.35 (m, 6H), 7.90 (s, IH), 8.27 (s, 111), 9.07 (s, 1H), 9.17 (s, 1H) and 10.05 (s, 1H) ;
MS: m/z 486 (M+1, 100%).

[00294] Synthesis of (E)-N-(3-16-(3-chloro-4-fluorophenylamino)-pyrimidin-4-ylaminolphenyl-N-methy1-4-(dimethyl-d6-amino)but-2-enamide (I-61) To a stirring solution of 4-bromo-but-2-enoic acid (0.28 g) at 0 C under nitrogen atmosphere and triethylamine (0.25 mL) in 3 mL of THF was added iso-butyl chlorofonnate (0.22 mL). The mixture was stirred for 15 mm followed by dropwise addition of a solution of 346-(3-chloro-4-fluorophenypaminopyrimidine-4-yljaminophenylamine (IR-4) (0.46 g) in 50 ml. of THF. The reaction was allowed to warm to room temperature overnight. The sample was concentrated then partitioned between Et0Ac and sat. NaHCO3 solution. The organic extract was washed with brine solution, dried (MgSO4), filtered and was concentrated. The resultant brown foamy solid was washed with diethyl ether and vacuum dried to give 0.378 g of (E)-N-3-(613-chloro-4-fluorophenylamino]pyrimidin-4-ylaminopheny1-4-bromo-2-butenamide (Int-E). MS
(m/z): 480, 478, 476 (25/100/80 %). To a stirring solution at 0 C under N2 of Int-E
(0.378 g, 0.794 mmol) and triethylamine (0.28 mL, 2.01 mmol) in 10 mL of THF was added in one portion dimethy1-d6-amine hydrochloride (0.070 g, 0.799 mmol). The sample was allowed to warm to room temperature overnight and then was partitioned between Et0Ae and sat. NaHCO3 solution. The organic extract was washed with brine solution, was dried (MgSO4), was filtered and was concentrated. The residue was chromatographed (silica gel, 1% NH.40H-10% Me0H
in CHC13) to give 0.0582 g (16%) of (E)-N-(346-(3-chloro-4-fluorophenylamino)-pyrimidin-Alaminopheny1-4-(dimethyl-d6-amino)but-2-enamide (I-61), a tan solid. 1H NMR
(DMSO-d6) 3.05-3.07 (m, 2H), 6.16 (s, 1H), 6.29-6.32 (in, 1H), 6.72-6.75 (m, 1H), 7.21-7.47 (in, 5H), 7.90 (s, I H), 7.92-8.01 (m, 1H), 8.32 (s, 1H), 9.26 (s, 1H), 9.36 (s, 1H) and 10.06 (s, 1H); MS: m/z 447/449 (M+1, 100/49%).

[00295] 1-80 may be prepared in a manner substantially similar to that described in Scheme 4a, above:

HN =
Nr-L N-ir-k 0 N N

446-(4-phenoxyphenyl)amino-pyrimidin-4-ylJamino-aminobenzene (I'-14) and acryloyl chloride gave N-416-(4-phenoxyphenyl)amino-pyrimidin-4-yl]aminophenylpropenamide (I-80), an off white solid. 11-1 NMR (DMSO-d6) 8 ppm : 5.73 (dd, J = 1.6 & 10.0 Hz, 1H), 6.08 (d, J = 5.6 Hz, 1H), 6.24 (dd, J = 2 & 16.8 Hz, 1H), 6.43 (dd, J = 10 & 16.8 Hz, 1H), 6.95-6.70 (m, 4H), 7.09 (t, J = 7.6 Hz, 1H), 7.35-7.39 (m, 211), 7.45-7.49 (m, 2H), 7.55-7.61 (m, 4H), 8.23 (s, 1H), 9.08 (s, 1H), 9.1 (s, 1H), 10.1 (s, 1H); LCMS : m/e 423.8 (M-F).

SCHEME 14a CI HN

NL

2 0 õ I

_____________________________________________________ =N
N N
step-1 step-2 [00296] Synthesis of N-6!-(6-(phenylamino)pyrimidin-4-ylamino)pyridine-2-yl)acrylamide (1-72) Step-1 A solution of N2-(6-chloropyrimidin-4-yl)pyridine-2,6-diamine (1) (0.25 g, 1.1 nnmol) and aniline (2) (0.16 g, 1.7 mmol) in n-BuOH (25 mL) was heated at 120 C for 12 h in a pressure tube. The reaction mixture was cooled, was dissolved in methanol (10 rnL) and was concentrated under reduced pressure. The residue was dissolved in ethyl acetate (35 mL) and was washed successively with with 10% sodium bicarbonate solution (20 mL), water (20 mL), and saturated brine (20 mL). The ethyl acetate extract was dried over Na2SO4 and was concentrated under reduced pressure to give a residue, which was triturated with diethyl ether to give (260 mg, 83%) N4-(6-aminopyridin-2-y1)-N6-phenylpyrirnidine-4,6-diamine (3) as an off-white solid.
Step-2 To a stirred solution of 3 (0.2 g, 0.7 mmol) in NMP (10 mL) was added acryloyl chloride (0.097 g, 1 mmol), drop wise at 0 C. The reaction mixture was allowed to stir at the same temperature for 20 min and then warmed to rt for 1.5 h. It was quenched with 10% sodium bicarbonate solution (4 mL) and was extracted with ethyl acetate (2x35 mL).
The combined ethyl acetate layer was washed with water (20 mL), saturated brine (20 mL), was dried over Na2SO4 and was concentrated under reduced pressure. The residue was further purified by column chromatography (Si02, 60-120, Petroleum ether/Et0Ac : 90/10) to give N-6-(6-(phenylamino)pyrimidin-4-ylamino)pyridine-2-ypacrylamide (1-72) as a brown solid. 11-1 NMR
(DMSO-d6) 5 ppm : 5.80 (dd, J = 1.84 & 10.12 Hz, 1H), 6.31 (dd, J = 1.8 &
16.96 Hz, 1H), 6.64 (dd, J = 10.08 & 16.88 Hz, 111), 6.96 (t, J = 7.32 Hz, 1H), 7.15-7.20 (m, 1H), 7.28 (t, J = 7.52 Hz, 2H), 7.34 (s, 1H), 7.60-7.70 (m, 4H), 8.30 (s, IH), 9.08 (s, 1H), 9.66 (s, 1H), 10.06 (s, 1H);
LCMS : m/e 332.6 (M+).

SCHEME 15a HN Cl Hy 0 p 0 NN"--.N NH2 ' [00297] Synthesis of (E)-4-(dimethylamino)-N-(6-(6-(phenylamino)pyrimidin-4-ylamino)pyridine-2-yl)but-2-enamide (1-73) To a stirred solution of acetonitrile (20 mL) and DMF (0.05 mL) under N2 was added IV,N-dimethylamino crotonic acid hydrochloride (0.47 g, 2.8 mmol). After 10 min this solution was cooled to 0-5 C. Oxalyl chloride (0.44 g, 3.5 mmol) was added and the reaction mixture was maintained at 0-5 C for 30 min. It was allowed to warm to rt and stirring was continued for 2 h.
It was then heated to 40 C for 5 min and again brought to rt and stirred for 10 min to get a light greenish colored solution of dimethylaminocrotonyl chloride that was used as such for next step.
To a stirred solution of .N4-(6-aminopyridin-2-yI)-/V6-phenylpyrimidine-4,6-diamine (0.2 g, 0.7 mmol) in NMP (10 mL) was added dropwise under N2 atmosphere at 0 C the solution of dimethylaminocrotonyl chloride. The reaction mixture was maintained at this temperature for 30 min and was warmed to A and stirred for 2 h. The mixture was quenched with sodium bicarbonate solution (1 mL) and was extracted with Et0Ac (2x35 mL). The combined ethyl acetate extract was washed with water (20 mL) and brine (20 mL), was dried over Na2SO4 and was concentrated under reduced pressure. The residue was purified by column chromatography (Si02, 60-120, product was eluted at 4-6% methanol in chloroform) to give (E)-(dimethylamino)-N-(6-(6-(phenylamino)pyritnidin-4-ylamino)pyridine-2-yl)but-2-enamide (I-73) as a yellowish solid. 1H NMR (DMSO-d6) 8 ppm : 2.24 (s, 6H), 3.13 (d, J =
5.76 Hz, 2H), 6.56 (d, J = 15.52 Hz, 1H), 6.80 (d, J = 15.36 Hz, 1H), 6.95 (t, J = 7.36 Hz, 1H), 7.11 (t, J =
1.16 Hz, 1H), 7.29 (t, J = 7.56 Hz, 2H), 7.58 (s, 1H), 7.65-7.7 (m, 4H), 8.31 (d, J = 2.44 Hz, III), 9.23 (s, 1H), 9.68 (s, 111), 10.16 (s, 1H); LCMS : m/e 390.3 (M+1).

SCHEME 16a el 0 ip HN
CI
'n 0 I
NNNN
1R_11 1-64 [00298] Synthesis of (E)-4-(dimethylamino)-N-(6-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)pyridine-2-yl)but-2-enamide (1-64) A solution of dimethylaminocrotonyl chloride was prepared by reaction of dimethylaminocrotonic acid hydrochloride (0.36 g, 2.16 mmol) in CH3CN (4 mL) containing DMF (I drop) with oxalyl chloride (0.34 g, 2.70 mmol) according to the procedure in Example 15a. This acid chloride was added drop wise at 0 C in to a stirred solution of N4-(6-aminopyridin-2-y1)-1V6-4-phenoxyphenylpyrimidine-4,6-diamine (1R1 1)(0.2 g, 0.54 mmol) in NMP (8 mL). The reaction was stirred at 0 C for 1 h, was diluted with Et0Ac (5 mL), and was washed with 10% NaHCO3 (2 mL), water (2 mL) and brine (2 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) to give (E)-(climethylamino)-N-(6-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)pyridine-2-yl)but-2-enamide (1-64) as a yellow solid. Ili NMR (DMSO-d6) 8 ppm : 2.18 (s, 6H), 3.05 (d, J = 5.2 Hz, 2H), 6.48 (d, J= 15.2 Hz, 1H), 6.79 (d, J= 6 & 15.6 Hz, 1H), 6.96-6.99 (m, 4H), 7.07-7.15 (m, 2H), 7.36 (t, J= 7.6 Hz, 2H), 7.42 (s, 1H), 7.64-7.7 (m, 4H), 8.30 (s, 1H), 9.12 (s, 1H), 9.69 (s, 1H), 10.07 (s, 1H); LCMS : m/e 482.4 (M+1).

SCHEME 17a n N NH2 step-1 L5 NNNN H2 step-2 N N

[00299] Synthesis of (E)-4-(dimethylamino)-N-(6-(6-(3-methylphenoxy)pyrimidin-ylamino)pyridine-2-yl)but-2-enamide (1-74) To a solution of N2-(6-chloropyrimidin-4-yl)pyridine-2,6-diamine (100 mg, 0.45 mmol) in DMI
(1 mL) was added 3-methylphenol (88 mg, 0.8 mmol) and anhydrous K2CO3 (93 mg, 0.6 mmol).
The reaction mixture was heated at 145 C for 16 h. It was then cooled and DMF
was removed under reduced pressure to get a yellow gummy residue. The residue was taken in Et0Ac (20 mL) and washed with water (5 mL), brine (5 mL) and dried over Na2SO4. Filtration followed by concentration under reduced pressure afforded a yellow gum which was further purified by column chromatography (Si02, 60-120, Et0Ac/hexane, 5/5) to give N2-(6-(3-methylphenoxy)pyrimidin-4-yOpyridine-2,6-diamine (I'-l2) (100 mg, 75%) as a pale yellow solid.
Step 2.

To a solution of I'-l2 (75 mg, 0.25 mmol) in NMP (2 mL) was added dimethylarninocrotonyl chloride (168 mg, 1.02 mmol) at 0 C. The reaction mixture was stirred at room temperature for 1 h and was then quenched with NaHCO3 solution (2 mL). The mixture was extracted with Et0Ac (3x5 mL) and the combined Et0Ac extract was washed with water (5 mL), brine (5 mL) and was dried over Na2SO4. Concentration under reduced pressure afforded a yellow oil, which was further purified by column chromatography (Si02, 60-120, Chloroform/Methanol, 9/1) to give (E)-4-(dimethylamino)-N-(6-(6-(3-methylphenoxy)pyrimidin-4-ylamino)pyridine-2-yl)but-2-enamide (1-74) as a light brown solid. 11-1 NMR (CD30D) 8 ppm: 2.35 (s, 6H), 2.38 (s, 3H), 3.25 (dd, J = 1.2 & 6.6 Hz, 2H), 6.40 (d, J= 13.16 Hz, 1H), 6.92-7.01 (rn, 3H), 7.12 (t, J = 8.12 Hz, 211), 7.34 (t, J = 7.84 Hz, 1H), 7.54 (s, 1H), 7.69 (t, J= 6.24 Hz, 1H), 7.79 (d, J = 8 Hz, 1H), 8.30 (s, 1H); LCMS: m/e 404.8 (M+).

SCHEME 18a o o 1110 o =
H =CI
HN
H2N HN H2N NH, N 0 'sk 1,1)1 ___________________________________ N-1 N'L 110 0 ¨ N
NI- a step-1 N CI step-2 L.

N N NH 2 step-3 N N

1003001 Synthesis of N-(3-(6-(3-phenoxyphenylamino)pyrimidin-4-ylamino)phenypacrylamide (1-75) Step 1 A solution of 4,6-dichloropyrimidine (0.5 g, 3.3 mmol), 3-phenoxyaniline (0.75 g, 4 mmol) and DIPEA (0.65 g, 5 mmol) in n-butanol (5 mL) was subjected to microwave irradiation (110 C, 30 mm). The reaction mixture was cooled, concentrated under reduced pressure and the residue was dissolved in Et0Ac (10 mL). This solution was washed with water (5 mL) and brine (5 mL), and was dried over Na2SO4. Concentrated under reduced pressure gave a residue that was purified by column chromatography (Si02, 60-120, hexane/ethylacetate, 8/2) to give 6-chloro-N-(3-phenoxyphenyl)pyrimidine-4-amine (0.56 g, 56%) as an off¨white solid.

Sta./
A solution of 6-chloro-N-(3-phenoxyphenyl)pyrimidine-4-amine (0.25 g, 0.8 mmol), 1,3-diaminobenzene (0.36 g, 3.3 mmol), n-butanol (10 inL) and conc. HC1 (61 mg, 1.6 mmol) was subjected to microwave irradiation (160 C, 15 mm). The reaction mixture was cooled, was concentrated under reduced pressure and the residue was taken in Et0Ac (10 mL). The Et0Ac solution was washed with water (5 mL) and brine (5 mL), and was dried over Na2SO4.
Concentration under reduced pressure gave a residue that was purified by column chromatography (Si02, 60-120, hexane/ethyl acetate, 6/4) to give N4-(3-aminopheny1)-N6-(3-phenoxyphenyppyrirnidine-4,6-diamine (0.1 g, 32%) as a brown solid.
Ste_pl To a stirred solution of N4-(3-aminopheny1)-N6-(3-phenoxyphenyl)pyrimidine-4,6-diamine (40 mg, 0.1 mmol), Et3N (0.03 mL, 0.2 mmol) and NMP (0.4 mL) in CH2C12 (2 mL), at 0 C, was added acryloyl chloride (6) (29 mg, 0.3 mmol). The reaction mixture was allowed to come to rt and stirred at this temperature for 3 h. It was washed with 10% NaHCO3 solution (2 mL), water (2 mL), brine (2 mL) and dried over Na2SO4. Filtration followed by concentration under reduced pressure offered a residue which was purified by column chromatography (Si02, 230-400, chloroform/methanol, 9/1) to gave N-(3-(6-(3-phenoxyphenylamino)pyrimidin-4-ylamino)phenyl)acrylamide (1-75) as a light brown solid. 1H NMR (DMSO-d6) 8 ppm : 5.73 (dd, J = 1.72 & 10 Hz, 1H), 6.18 (s, 1H), 6.24 (dd, J = 1.92 & 17 Hz, I H), 6.45 (dd, J = 10.04 &
= 16.88 Hz, 1H), 6.54-6.57 (m, 1H), 7.01-7.05 (m, 2H), 7.11-7.15 (dd, J =
0.88 & 7.48 Hz, 1H), 7.19-7.32 (s, 4H), 7.35-7.41 (m, 4H), 7.89 (s, 1H), 8.26 (s, 1H), 9.2 (s, 1H), 9.25 (s, 1H), 10.26 (s, 1H); LCMS : m/e 423.8 (M+1).

SCHEME 19a = =
Br Br lo 0 ,Thrc, 0 40 nN N N NH2 OH N N OH N NH2 N N Nn NH2 NNNN
H H
11-76 [00301] Compound 1-76 can be prepared according to Scheme 19a by coupling intermediate 1 with 3-bromophenol, followed by boronic acid coupling of intermediates 3 and 4 to give intermediate 5. Intermediate 5 can then be treated with acryloyl chloride using a protocol similar to that in Example 6 to give compound 1-76.

SCHEME 20a V V

0S ci)-N 0 Si N--L n ______________________________________ y. N').'''s 1 0 k-,---, ,a. ,--õ
k 1 = 1N N NH2 NNNNK,.N -.
H H H

[00302] Compound 1-77 can be prepared according to Scheme 20a by treatment of intermediate 5 with (E)-4-(dimethylamino)but-2-enoyl chloride using a protocol similar to that in Example 6.

SCHEME 21a y, H2N N NH2 I.

--%-;''=-, I
-..- _______________________________ ,.. .

step-1 step-2 H I step-3 "
. 40 0 so .,,y0 40 0 0 CI
HN HN
N,--J---step-4 H I H I

[00303] Synthesis of N-methyl-N-(6-(6-(4-phenoxyphenylamino)pyrimidin-4-ylarnino)pyridin-2-yl)acrylamide (1-78) Rai To a solution of 2,6-diaminopyridine (10 g, 91.63 mmol) in dry THF (100 mL) was added K2CO3 (18,8 g, 136.23 mmol) and CH3I (13 g, 91.63 mmol) and the reaction mixture was stirred at room temperature for 16 h. Water was added (10 mL) and the mixture was extracted with Et0Ac (100 mL). The Et0Ac layer was dried and was concentrated under reduced pressure. The residue was further purified by column chromatography (Si02, 60-120, chloroform) to give 2-methylamino-6-aminopyridine (1.1 g, 10%) as a brown solid.
Stu/
A mixture of 2-methylamino-6-aminopyridine (0.5 g, 4.04 mmol), 4,6-dichloropyrimidine (1.51 g, 10.13 mmol), DIPEA (1.5 g, 12.17 mmol) in n-butanol (5 mL) was heated at 120 C for 16 h.
The reaction mixture was cooled, was concentrated under reduced pressure and the residue was taken in dichloromethane (25 mL). The dichloromethane solution was washed with NaHCO3 solution (2 mL), water (2 mL) and brine (2 mL), was dried over Na2SO4 and was concentrated under reduced pressure. The residue was purified by column chromatography (Si02, 60-120, petroleum ether/ethyl acetate, 6/4) to give N2-(6-chloropyrimidin-4-y1)-N6-methylpyridine-2,6,diamine (0.3 g, 33%) as a yellow solid.
Ste_p_a A solution of N2-(6-chloropyrimidin-4-y1)-1V6-methylpyridine-2,6,diamine (0.3 g 1.27 mmol), 4-phenoxy aniline (0.28 g, 1.52 mmol) and conc. HC1 (2 drops) in n-butanol (2 mL) was subjected to microwave irradiation (120 C, 1 h). The reaction mixture was cooled, was concentrated under reduced pressure, and the residue was diluted with CH2C12 (5 mL). The dichloromethane solution was washed with NaHCO3 (2 mL), water (2 mL), and brine (2 mL), and was dried over Na2SO4.
Filtration followed by concentration under reduced pressure gave a residue which was purified by column chromatography (Si02, 60-120, chloroform/methanol, 9/1) to give N4-(6-(methylamino)pyridine-2-y1)-N6-(4-phenoxyphenyl)pyrimidine-4,6-diarnine (0.2 g, 41%) as a light brown solid.
S te.p To a solution of N4-(6-(methylamino)pyridine-2-y1)-N6-(4-phenoxyphenyl)pyrimidine-4,6-diamine (0.07 g, 0.18 mmol) in NIVIP (1 mL) was added acryloyl chloride (0.032 g, 0.36 mmol) at 0 C and the reaction mixture was stirred at rt for 1 h. The reaction mixture was diluted with dichloromethane (2 mL), was washed with NaHCO3 (1 mL), water (1 mL), and brine (1 mL).
The dichloromethane solution was dried over Na2SO4 and was concentrated under reduced pressure. The residue was purified by column chromatography (Si02, 60-120, chloroform/methanol, 9/1) to give N-methyl-N-(6-(6-(4--phenoxyphenylamino)pyrimidin-4-ylamino)pyriclin-2-yOacrylamide (1-78) as a yellow solid. 11-1 NMR (DMSO-d6) 5 ppm: 3.22 (s, 3E1), 5.60 (dd, J = 2.56 & 9.76 Hz, 1H), 6.12-6.16 (m, 2H), 6.79 (d, J = 7.56 Hz, 1H), 6.96 (d, J
= 8.64 Hz, 5H), 7.09 (t, .1 = 7.32 Hz, 1H), 7.23 (s, 1H), 7.33-7.37 (m, 4H), 7.45 (d, J = 8.2 Hz, 2E1), 7.32 (t, J = 7.8 Hz, 1H), 8.24 (s, 1H); LCMS: m/e 439.3 (M+1).

SCHEME 22a =0 ill HN HN =
_____________________________________ N 0 N
N N N NH NN N

1003041 Synthesis of (E)-4-(dimethylamino)-N-methyl-N-(6-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)pyridine-2-yl)but-2-enamide (1-79) To a solution of dimethylaminocrotonic acid hydrochloride (0.120 g, 0.72 mmol) in CH3CN (1.4 mL) was added DMF (1 drop) followed by oxalyl chloride (0.07 mL, 0.91 mmol) at 0 C under nitrogen atmosphere. The reaction was allowed to stir at this temperature for 30 min and then at rt for 2 h. This acid chloride was added, drop wise, at 0 C in to a stirred solution of N4-(6-(rnethylamino)pyridine-2-y1)-N6-(4-phenoxyphenyl)pyrimidine-4,6-diamine (0.07g, 0.18 mmol) in NM? (2.8 mL). The reaction was allowed to stir at 0 C for 1 h, diluted with Et0Ac (5 mL), and washed with 10% NaHCO3 (2 mL), water (2 inL) and brine (2 mL). drying over Na2SO4 followed by concentration under reduced pressure gave a residue which was further purified by column chromatography (Si02, 60-120, chloroform/methanol, 9/1) to give (E)-4-(dimethylamino)-N-methyl-N-(6-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)pyridine-2-yl)but-2-enamide (1-79) as a yellow solid. 'H NIAR. (DMSO-d6) 5 ppm: 2.05 (s, 6H), 2.91 (d, J
= 6 Hz, 2H), 3.27 (s, 3H), 6.08 (d, J = 15.2 Hz, 1H), 6.65 (dd, J = 5.6 & 14.8 Hz, 1H), 6.82 (d, J
= 7.6 Hz, 1H), 6.97-7.00 (m, 4H), 7.10 (t, J= 7.2 Hz, 1H), 7.20 (s, 1H), 7.35-7.39 (m, 2H), 7.46 (d, J= 8 Hz, 1H), 7.53 (d, J= 8.8 Hz, 2H), 7.75 (t, J= 8 Hz, 1H), 8.30 (s, 1H), 9.30 (s, 1H), 9.95 (s, 1H); LCMS: m/e 496 (M+1).

Scheme 23a fa el So, [00305] Synthesis of (E)-4-dimethylamino)-N-(6-(6-(4-phenoxyphenoxy)pyrimidin-ylamino)pyridin-2-yl)but-2-enamide (1-82) To a stirred solution of N2-(6-(4-phenoxyphenoxy)pyrimidin-4-yl)pyridine-2,6-diamine (0.65 g, 1.75 mmol) in NMP (10 mL) was added dimethylaminocrotonyl chloride (1.026 g, 7 mmol) at 0 C. The reaction mixture was allowed to come to room temperature and kept at it for 1 h. It was diluted with dichloromethane (10 mL), was washed with NaHCO3 solution (2 mL) and water (2 mL), and dried over Na2SO4. The dichloromethane solution was filtered and was concentrated under reduced pressure to give a residue that was purified by column chromatography (Si02, 60-120, chloroform/methanol, 9/1) to give (E)-4-dimethylamino)-N-(6-(6-(4-phenoxyphenoxy)pyrimidin-4-ylamino)pyridin-2-yObut-2-enamide (1-82) as an off white solid.
NMR (DMSO-d6) 8 ppm : 2.19 (s, 6H), 3.08 (d, J = 5.52 Hz, 2H), 6.50 (d, J =
15.4 Hz, 1H), 6.77 (td, J = 5.92 & 15.4 Hz, 1H), 7.00-7.07 (m, 5H), 7.15 (t, J = 7.36 Hz, 1H), 7.21 (dd, J = 2.2 & 8.92 Hz, 2H), 7.41 (t, J = 7.52 Hz, 2H), 7.68 (t, J = 7.96 Hz, 1H), 7.77 (d, J = 7.96 Hz, 1H), 7.95 (s, 1H), 8.35 (s, 1H), 10.20 (s, 1H), 10.40 (s, 1H); LCMS : m/e 483 (M+1).

Scheme 24a o N., 9O4 NH NH HO NH HO
=N
N N Step 1 N N Step 2 [l N N

[00306] Synthesis of 2-(hydroxy(3-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)phenyl)methyl)acrylamide (1-84) Step 1 To a stirred solution of N-methoxy-N-methy1-3-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)benzamide (0.75 g, 1.7 mmol) in THF (10 mL) was added LAH (3.4 mL, 3.4 mmol, 1 M solution in TliF) at -60 C. The reaction mixture was stirred at -60 C for 1 h, was quenched with Na2SO4 solution (2 mL) and was extracted with ethyl acetate (10 mL). The organic layer was separated and washed with water (2 mL) and with brine solution (2 mL) and was dried over anhydrous Na2SO4. Filtration followed by concentration under reduced pressure gave a residue that was purified by column chromatography (Si02, 60-120, chloroform/methanol, 9/1) to give 3-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)benzaldehyde (0.6 g, 92%) as an off yellow solid.
Step 2 To a stirred solution of 3-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)benzaldehyde (100 nig, 0.26 mmol) and acrylonitrile (36 mg, 0.52 mmol) in 1,4- dioxane/H20 (0.5 mL / 0.5 mL) was added DABCO (29 mg, 0.26 mmol) at rt. Stirring was continued at room temperature for 48 h after which time the reaction mixture was concentrated under reduced pressure. The residue obtained was further purified by column chromatography (Si02, 60-120, pet ether/ethyl acetate, 6/4) to give 2-(hydroxy(3-(6-(4-phenoxyphenylamino)pyrimidin-ylamino)phenyl)methyl)acrylamide (1-84) as a white solid. 11-1 NMR (DMSO-d6) 5 ppm : 5.33 (s, 1H), 6.06 (s, 1H), 6.20 (s, 1H), 6.25 (s, 1H), 6.80 (s, 1H), 6.88 (s, I
H), 6.95-7.05 (m, 4H), 7.09-7.13 (m, 2H), 7.16 (bd, J = 7.92 Hz, 1H), 7.32-7.39 (m, 5H), 7.47 (s, 1H), 8.31 (s, 1H);
LCMS : in/e436 (M+1).

Scheme 25a ,o 0 BOC ? HN HN

N
H
N CI step-1 N NH step-2 NNH step-3 N NH step-4 N, NH
BOC BOC
So NH

1003071 Synthesis of 1-(3-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)pyrrolidin-1-yl)prop-2-en-1-one (1-85) Step 1 A solution of 4,5-dichloropyrimidine (0.6 g, 4.02 mmol), 3-amino-Boc-pyrrolidine (0.5 g, 2.6 ramol) and D1PEA (1.73 g, 13.3 mmol) in n-butanol (5.0 mL) was heated in a pressure tube (120 C, 12 h). It was cooled, quenched with water (10 mL) and was extracted with Et0Ac (2x25 rnL). The combined Et0Ac extract was washed with water (5 mL), brine (5 mL), dried over Na2SO4 and concentrated under reduced pressure to afford tert-butyl 3-(6-chloropyrimidin-4-ylamino)pyrrolidine-l-carboxylate (0.4 g, 50%) as a yellow solid.

=

Step 2 To a stirring solution of tert-butyl 3-(6-chloropyrimidin-4-ylamino)pyrrolidin-l-carboxylate (0.5 g, 1.6 mmol) and 4-phenoxy aniline (0.309 g, 1.6 mmol) in ethanol (4 mL) was added acetic acid (0.1 mL) and the reaction mixture was heated at 100 C for 36 h. The reaction mixture was cooled, ethanol was removed under reduced pressure and the residue was taken in ethyl acetate (10 mL). It was washed with NaHCO3 solution (2 mL), brine (2 mL), dried over Na2SO4 and concentrated under reduced pressure. The residue was further purified by column chromatography (Si02, 60-120, chloroform/methanol, 9/1) to yield tert-butyl phenoxyphenylamino)pyrimidin-4-ylamino)pyrrolidine-1 -carboxylate (0.3 g, 42.8%) as a white solid.
Step 3 To a stirred solution of tert-butyl 3-(6-(4-phenoxyphenylamino)pyrimidin-4-ylarnino)pyrrolidin-l-carboxylate (0.1 g, 0.2 mmol) in dry CH2C12 (2.0 mL) at 0 C was added CF3COOH (2 mL, 20 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 3 h. The reaction mixture was concentrated under reduced pressure and the residue was quenched with water (2 mL), basified with NaHCO3 solution, and was extracted with ethyl acetate (2x8 mL). The combined ethyl acetate extract was washed with water (2 mL) and brine (2 mL), was dried over Na2SO4 and was concentrated under reduced pressure to give N4-(4-phenoxypheny1)-N6-(pyrrolidin-3-yl)pyrimidine-4,6-diamine (0.025 g, 32.4%) as a light a brown solid.
Step 4 To a stirred solution of N4-(4-phenoxypheny1)-/V6-(pyrrolidin-3-yppyrimidine-4,6-diamine (0.13 g, 0.3 mmol) in THF (1.5 mL) at -60 C were added DIPEA (0.07 g, 0.5 mmol) and acryloyl chloride (1 M solution in THF, 0.3 mL, 0.3 mmol), and the reaction mixture was stirred at -60 C
for 5 min. The reaction mixture was quenched by adding water and it was extracted with Et0Ac (2x5 mL). The combined Et0Ac extract was washed with brine (3 mL), was dried over Na2SO4 and was concentrated under reduced pressure. The residue obtained was purified by column chromatography (Si02, 230-400, chloroform/methanol: 98/2) to give 1434644-phenoxyphenylamino)pyrimidin-4-ylamino)pyrrolidin-1 -yl)prop-2-en- 1 -one (1-85) as a light green solid. NMR
(DMSO-d6) .5 ppm: 1.83-1.86 & 1.90-1.94 (m, 1H), 2.07-3.0 & 2.16-2.20 (m, 1H), 3.38-3.86 (m, 4H), 4.2-4.75 & 4.35-4.5 (bs, 111), 5.65 (dt, = 2 & 10 Hz, 1H), 5.78 (d, = 3.6 Hz, 1H), 6.11 & 6.15 (dd, J= 2.4 & 7.0 Hz & dd, J= 2.4 & 7.2 Hz, 1I-1), 6.51-6.64 (m, together 1H), 6.93-7.00 (m, 4H), 7.07-7.18 (m, 2H), 7.36 (t, J= 8 Hz, 2H), 7.51 (d, J = 8 Hz, 21-I), 8.12 (s, 1H), 8.93 (s, 1H); LCMS : m/e 401.8 (M+1).

Scheme 26a pi-Boc 401 ark 0 yi HN
steP-1 step-2 N step-3 [L.
N CI N NH N NH

HN HN
_step-4 11:L
N NH N NH
HN-100308] Synthesis of 1-(3-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)piperidin-1-yl)prop-2-en-1one (1-86) Step 1 A solution of 4,6-dichloropyrimidine (0.1 g, 0.671 mmol), 3-amino-Boc-piperidine (0.16 g, 0.80 mmol) and DIPEA (0.086 g, 6.71 mmol) in n-butanol (5.0 mL) was heated in a pressure tube (120 C, 12 h). The solution was cooled, was quenched with water (2 mL) and was extracted with Et0Ac (2x15 mL). The combined Et0Ac extract was washed with water (5 mL), brine (5 naL), was dried over Na2SO4 and was concentrated under reduced pressure to give tert-butyl 3-(6-chloropyrimidin-4-ylamino)piperidine- 1 -carboxylate, which was dried under high vacuum and was used as such for next step without further purification.
Step 2 A solution of tert-butyl 3-(6-chloropyrimidin-4-ylamino)piperidine- 1 -carboxylate (0.15 g, 0.48 mmol), 4-phenoxyaniline (0.089 g, 0.48 mmol), Pd(OAc)2 ( 0.010 g, 0.048 mmol), BINAP
(0.014 g, 0.024 mmol) and Cs2CO3 (0.39 g, 1.2 mmol) in degassed toluene (toluene was purged with N2 for 15 min) was heated for 12 h at 100 C under N2 atmosphere. The reaction mixture was cooled, was diluted with Et0Ac (20 mL) and was washed with water (4 mL) and brine brine (2 mL) and was dried over Na2SO4. The crude product obtained was purified by column chromatography (Si02, 230-400, chloroform/methanol : 99/1) to give tert-butyl 3-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)piperidine-l-carboxylate (90 mg, 40.9%) as a light yellow solid.
Step 3 To a stirred solution of tert-butyl 3-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)piperidine-l-carboxylate (110 mg, 0.238 mmol) in dry CH2C12 (1.0 mL) at 0 C was added CF3COOH (0.5 naL, 5 vol) and the reaction mixture was kept at this temperature for 30 mm.
It was allowed to come to rt and to stir at this temperature for 3 h. The reaction mixture was concentrated under reduced pressure and the residue was quenched with water (2 mL), was basified with NaHCO3 solution, and was extracted with ethyl acetate (2x8 mL). The combined ethyl acetate extract was washed with water (2 mL), brine (2 niL), dried over Na2SO4 and concentrated under reduced pressure to give N4-(4-phenoxypheny1)-.N6-(piperidin-3-yppyrimidine-4,6-diamine (0.07 g, 81%) as a light yellow solid.
Step 4 To a stirred solution of /V4-(4-phenoxypheny1)-A/6-(piperidin-3-yppyrimidine-4,6-diamine (0.025 g, 0.069 mmol) in NMI' (0.5 mL) at 0 C was added acryloyl chloride (0.007 g, 0.083 mmol), and the reaction mixture was stirred at 0 C for 5 min. The reaction mixture was quenched by adding 10% NaHCO3 solution and it was extracted with Et0Ac (2x5 mL). The combined Et0Ac extract was washed with water (3 mL) and brine (3 inL), was dried over Na2SO4 and was concentrated under reduced pressure. The residue obtained was purified by column chromatography (Si02, 230-400, chloroform/methanol: 98/2) to give 1434644-phenoxyphenylamino)pyrimidin-4-ylamino)piperidin-1-yl)prop-2-en-lone (1-86) as an off white solid. IHNMR (Me0D) 8 ppm: 1.5-1.75 (m, 2H), 1.80-2.00 (m, 1H), 2.0-2.20 (m, 1H), 2.70-2.90 (m, 1H), 2.90-3.05 (m, 1H), 3.80-4.00 (m, 2H), 4.30-4.45 (m, 1H), 5.65 & 5.75 (d, J=
10.8 Hz & d, J= 10.8 Hz respectively, together 1H), 5.81 (d, J = 10.8 Hz, I
H), 6.14 & 6.18 (d, J
= 17.2 Hz & d, J = 19.2 Hz respectively, together 1H), 6.60-6.70 & 6.70-6.85 (m, together 1H), 6.97-7.00 (m, 4H), 7.04 (t, J= 7.6 Hz, 1H), 7.32-7.38 (m, 4H), 8.04 & 8.07 (s, together 1H);
LCMS: m/e 416.1 (M+1).

SCHEME 27a COOMe CI CI NH 00Me NH 00H
step-1 N step-2 step-3 step-4 410:1 COOMe 0 NI-OMe 0 NH step-5 41" NH
N 40)NL 0, NN N N

[00309] Synthesis of 3-methy1-1-(34644-phenoxyphenylamino)pyrimidin-4-ylamino)phenyl)but-2-en-1-one (1-87) Step 1._ To a stirred solution of 4,6-dichloropyrimidine (0.5 g, 3.7 mmol) in n-butanol (10 mL) was added methyl 3-aminobenzoate (0.498 g, 3.7 mmol) and DIPEA (0.65 g, 5.0 mmol) and the reaction mixture was heated at 110 C for 12 h. It was cooled and excess n-butanol was removed under reduced pressure. The residue was extracted with Et0Ac (2x30 mL) and the combined Et0Ac extract was washed with water (5 mL), brine (2.5 mL), dried over Na2SO4 and concentrated under reduced pressure. The residue was stirred with pet ether (30 mL) for 30 min, the pet ether removed by decantation and the solid obtained was dried under high vacuum to give methyl 3-(6-chloropyrimidin-4-ylamino)benzoate (0.3 g, 34%) as light brown solid.
Step 2, To a stirring solution of methyl 3-(6-chloropyrimidin-4-ylamino)benzoate (1.0 g, 3.8 mmol) and 4-phenoxy aniline (0.703 g, 3.8 mmol) in ethanol (5 mL) was added acetic acid (0.22 mL) and the reaction mixture was heated at 100 C for 48 h. The reaction mixture was cooled, ethanol was removed under reduced pressure and the residue was dissolved in ethyl acetate (50 mL). It was washed with NaHCO3 solution (5 mL) and brine (5 mL), was dried over Na2SO4 and was concentrated under reduced pressure. The residue was purified by column chromatography (Si02, 60-120, chloroform/methanol, 9/1) to yield methyl 3-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)benzoate (1 g, 66%) as an off-white solid.
Step 3 To a stirred solution of methyl 3-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)benzoate (0.3 g, 0.72 mmol) in methanol/THE (2/2, 4 mL) was added LiOH (0.122 g, 2.9 mmol) in H20 (4 mL) and the reaction mixture was stirred at rt for 2 h. It was concentrated under reduced pressure. the residue was diluted with water (2 mL) and was extracted with dichloromethane (5 mL). The aqueous layer was separated and was acidified with 1.5 N HC1 (pH ¨5-6) to get a white precipitate, which was collected by filtration and dried under vacuum to give phenoxyphenylamino)pyrimidin-4-ylamino)benzoic acid (0.2 g 69%) as a white solid.
Step 4, To a stirred solution of 3-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)benzoic acid (0.05 g, 0.12 mmol) in DMF (2 mL) were added MeNH-OMe.HCI (0.0084 g, 0.12 mmol), (0.0361 g, 0.18 mmol), HOBT (0.0084 g, 0.062 mmol) and DIPEA (0.023 g, 0.18 mmol). The reaction mixture was stirred at room temperature for 1 h and was quenched with water. A white solid was isolated by filtration and dried under vacuum to give N-methoxy-N-methy1-3-(6-(4-phenoxyphenylamino)pyrirnidin-4-ylamino)benzamide (0.025 g, 44.5%) as a white solid.

Step 5 To a stirred solution of N-methoxy-N-methy1-3-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)benzamide (50 mg, 0.11 frump in THF (0.5 mL) at 0 C was added 2-rnethylpropenylmagnesium bromide (1.1 mL, 0.55 mmol, 0.5 M in THF). The reaction mixture . was allowed to stir at room temperature for 30 min. It was quenched with sat. NRICI solution (0.5 mL) and was extracted with Et0Ac (3x2 mL). The combined organic layer was washed with brine, was dried over anhydrous Na2SO4, was filtered and was concentrated under reduced pressure to give a white solid, which was further purified by column chromatography (Si02, 60-120, chloroform/methanol, 9/1) to give 3-methy1-1-(3-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)phenyl)but-2-en- 1-one (1-87) as an off white solid. 1H NMR (CDC13) 8 ppm: 2.01 (s, 3H), 2.20 (s, 3H), 6.14 (s, 1H), 6.71 (s, 1H), 6.95 (s, 1H), 6.99-7.02 (m, 5H), 7.11 (t, J= 7.36 Hz, 1H), 7.26-7.28 (m, 2H), 7.34 (t, J = 7.56 Hz, 2H), 7.40-7.53 (m, 2H), 7.65 (d, J = 7 Hz, 1H), 7.86 (s, 1H), 8.32 (s, 1H); LCMS : m/e 437.2 (M+1).

Scheme 28a O (110 0 N-Ome 0 NH NH
40) __________________________________ Nei 411 N N N N

1003101 Synthesis of 1-(3-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)phenyl)but-2-yn-1-one (1-88) To a stirred solution of N-methoxy-N-methy1-3-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)benzamide (50 mg, 0.11 mmol) in THF (0.5 mL) at 0 C was added a THF
solution of 1-butynylmagnesium bromide (1.1 mL, 1.1 mmol). The reaction mixture was allowed to come to rt and was stirred at 11 for 30 min. The reaction mixture was quenched with saturated NH4C1 solution (0.5 mL) arid was extracted with Et0Ac (2x3 mL). The combined Et0Ac layer was 1-i2 washed with brine, was dried over anhydrous Na2SO4, was filtered and was concentrated under reduced pressure to give a white solid, which h was further purified by column chromatography (Si02, 60-120, chloroform/methanol, 9/1) to give 1-(3-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)phenyl)but-2-yn- 1-one (1-88) as an off-yellow solid. IHNMR (DMSO-d6) 5 ppm: 2.22 (s, 3H), 6.16(s, IH), 6.97 (d, J= 8.6 Hz, 2H), 7.01 (d, J= 8.8 Hz, 2H), 7.1 (t, J= 7.2 Hz, 1H), 7.35-7.39 (m, 2H), 7.48 (t, J= 8 Hz, 1H), 7.56 (d, J= 8.8 Hz, 2H), 7.66 (d, J=
7.2 Hz, 1H), 7.95 (d, J= 7.6 Hz, 1H), 8.32 (s, 1H), 8.39 (s, 1H), 9.22 (s, 1H), 9.47 (s, 1H);
LCMS : m/e 421.1 (M+1).

Scheme 29a 0 N-0Me 0 NH NH
NC
LN I N 411:1 N N

[90311] Synthesis of (E,Z)-1-(3-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)phenyl)but-2-en-1-one (1-89) To a stirred solution of N-methoxy-N-methy1-3-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)benzamide (50 mg, 0.11 mmol) in THF (0.5 mL) at 0 C was added a THF
solution of propenylmagnesium bromide (2.2 mL, 1.1 mL, 0.5 M soln. in THF). The reaction mixture was allowed to stir at room temperature for 30 min. It was quenched with saturated NH4C1 solution (0.5 mL) and was extracted with Et0Ac (2x3 mL). The combined organic layer was washed with brine, was dried over anhydrous Na2SO4, was filtered and was concentrated under reduced pressure to get a white solid, which was further purified by column chromatography (Si02, 60-120, chloroform/methanol, 9/1) to give (E,Z)-1-(3-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)phenyl)but-2-en-l-one (1-89) as a pale yellow solid. 111 NMR (DMSO-d6) 8 ppm: 1.98 (dd, J = 1.6 & 6.8 Hz, 3H) & 2.13 (dd, J = 1.6 & 7.2 Hz, 3H), 6.10-6.13 (m, 1H), 6.75-6.90 (m, 1H), 6.90-7.13 (m, 7H), 7.25-7.27 (m, 1H), 7.32-7.34 (m, 2H), 7.34-7.50 (m, 2H), 7.63-7.65 (m, )H), 7.86-7.88 (m, 1H), 8.32 (s, 1H) ; LCMS : m/e 423 (M+1).

Scheme 30a NH
0 N'OMe 0 401 NH

NkN
I.
N N

1003121 Synthesis of 2-methy1-1-(3-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)phenyl)prop-2-en-1-one (1-83) To a N-methoxy-N-methy1-3-(6-(4-phenoxyphenylarnino)pyrimidin-4-ylamino)benzamide (0.150 g, 0.340 mmol) at 0 C was added 2-methylpropenylmagnesium bromide (6.8 mL, 3.4 rnmol, 0.5 M solution in THF). The reaction mixture was allowed to stir at room temperature for 30 min. It was quenched with saturated NH4C1 solution (0.5 mL) and was extracted with Et0Ac (2x3 mL). The combined organic layer was washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure to get a white solid, which was further purified by column chromatography (Si02, 60-120, product getting eluted in methanoUchloroform:
2/98) to give 2-methy1-1-(3-(6-(4-phenoxyphenylam ino)pyrimi din-4-ylamino)phenyl)prop-2-en-1 -one (1-83) as a white solid. NMR (DMSO-d6)45 ppm: 1.99 (s, 3H), 5.64 (s, 111), 6.03 (s, 1H), 6.14 (s, 1H), 6.96-7.02 (m, 4H), 7.10 (t, J= 7.6 Hz, 111), 7.25 (d, J= 7.6 Hz, 111), 7.35-7.43 (m, 311), 7.56 (d, I = 8.8 Hz, 211), 7.88 (d, J = 8 Hz, 111), 7.92 (s, 111), 8.29 (s, 111), 9.21 (s, 111), 9.38 (s, 1H);
LCMS : 423 m/e (M+1).

Scheme 31a OMe ito omi NH2 nor. OMpy thi 7-1 __________________ 0,.ryNH _______________ ONH
N N N NH2 Step-1 N N Step-2 N N

[00313] Synthesis of N-(6-(6-(3-methoxyphenoxy)pyrimidin-4-ylamino)pyridine-2-yl)acrylamide (1-90) Step 1, To a solution of N2-(6-chloropyrimidin-4-yl)pyridine-2,6-diamine (200 mg, 0.90 mmol) in dry DMF (2 mL) was added 3-methoxyphenol (112 mg, 0.90 mmol) and anhydrous K2CO3 (186 mg, 1.353 mmol). The reaction mixture was heated at 100 C for 16 h under N2 atmosphere. It was then cooled and DMF removed under reduced pressure to give a yellowish gummy residue that was taken in Et0Ac (10 mL). It was washed with water (5 mL), brine (5 mL), dried over Na2Sa4 and then concentrated under reduced pressure to give a crude product. This was further purified by column chromatography (Si02, 60-120, hexane/Et0Ac, 5/5) to give N2-(6-(3-methoxylphenoxy)pyrimidin-4-yl)pyridine-2,6-diamine (110 mg, 40.7%) as a pale yellow solid.
Step 2_ To a stirred solution of N2-(6-(3-methoxylphenoxy)pyrimidin-4-yl)pyridine-2,6-diamine (100 mg, 0.323 mmol) in THF/NMP (1 mL / 0.5 mL) was added acryloyl chloride (0.029 g, 0.3 nunol) under N2 atmosphere at -10 C. The stirring was continued at the same temperature for 2 h and the reaction mixture was concentrated under reduced pressure to give a residue that was further purified by column chromatography (Si02, 60-120, chloroform/methanol) to give N-(6-(6-(3-methoxyphenoxy)pyrimidin-4-ylamino)pyridine-2-yl)acrylamide (1-90) as a pale yellow solid. 'H NMR (DMSO-d6) 8 ppm: 3.75 (s, 3H), 5.79 (dd, J= 1.8 & 10.08 Hz, 1H), 6.30 (dd, J=
1.8 & 16.96 Hz, 1H), 6.65 (dd, J= 10.12 & 16.96 Hz, 1H), 6.74-6.76 (m, 2H), 6.82 (td, J= 1.52 & 9.24 Hz, 1H), 7.04 (d, J= 7.92 Hz, 1H), 7.33 (t, J= 8.28 Hz, 1H), 7.70 (t, J= 7.96 Hz, 1H), 7.76 (d, J= 8 Hz, IH), 7.90 (s, 1H), 8.34 (s, 1H), 10.20 (s, IH), 10.48 (s, 1H); LCMS : m/e 364 (M+1).

Scheme 32a SMe *Me icy NH2 n _______________________ 0 N
)nrH _______________________________________________ OyNH
N N N NH2 Step-1 N N Step-2 N N

[00314] Synthesis of N-(6-(6-(4-methoxyphenoxy)pyrimidin-4-ylamino)pyridine-2-yl)acrylamide (1-91) Step 1 To a solution of N2-(6-chloropyrimidin-4-y1)pyridine-2,6-diarnine (200 mg, 0.90 mmol) in dry DMF (2 mL) was added 4-methoxyphenol (168 mg, 1.3 mmol) and anhydrous K2CO3 (179 mg, 1.3 mmol). The reaction mixture was heated at 100 C for 16 h under N2 atmosphere. It was then cooled and DMF removed under reduced pressure to give a yellowish gummy residue that was taken in Et0Ac (10 mL). The solution was washed with water (5 mL) and brine (5 mL), was dried over Na2SO4 and was then concentrated under reduced pressure to get crude product. This was further purified by column chromatography (Si02, 60-120, hexane/Et0Ac, 5/5) to give N2-(6-(4-methoxylphenoxy)pyrimidin-4-yppyridine-2,6-diamine (160 mg, 59.2 %) as a pale yellow solid.
Step 2 To a stirred solution of N2-(6-(4-methoxylphenoxy)pyrimidin-4-yl)pyridine-2,6-diamine (150 mg, 0.474 mmol) in THF/NMP (1 mL / 0.5 mL) was added acryloyl chloride (64 mg, 0.712 ramol) at -10 C under N2 atmosphere. After stirring at this temperature for 30 min, the reaction was stopped and the reaction mixture was slowly added to NaHCO3 solution (10 mL). A white solid was precipitated which was isolated by filtration and was dissolved in a mixture of ethyl acetate (5 mL) and Et3N (0.5 mL). The solution was washed with water (2 mL) and brine (2 mL).
Drying over Na2SO4 followed by filtration and concentration under reduced pressure afforded a yellow solid. It was further purified by column chromatography (Si02, 60-120, chloroform/methanol, 9/1) to give N-(6-(6-(4-methoxyphenoxy)pyrimidin-4-ylamino)pyridine-2-ypacrylatnide (1-91) as an off white solid. 1HNMR (DMSO-d6) 8 ppm: 3.76 (s, 3H), 5.79 (dd, J
= l.84& 10.16 Hz, 1H), 6.30 (dd, J = l.84& 17 Hz, 1H), 6.65 (dd, J= 10.12&
16.92 Hz, 1H), 6.96-6.99 (m, 211), 7.02 (d, J= 7.88 Hz, 1H), 7.09-7.13 (m, 211), 7.69 (t, J=
7.96 Hz, 114), 7.76 (d, J= 7.44 Hz, 1H), 7.86 (s, 1H), 8.30 (d, J= 0.88 Hz, 1H), 10.17 (s, 1H), 10.17 (s, 111); LCMS
: m/e 364 (M+1).

Scheme 33a HN

N NH step-1 NH 2 step-2 N N
BOC

[00315] Synthesis of N-(3-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)phenyl)propionamide (1R-10) Step 1 A solution of tert-butyl 3-(6-chloropyrimidin-4-ylamino)phenylcarbamate (200 mg, 0.6 mmol), 4-phenoxyaniline (346 mg, 1.8 mmol) and concentrated HC1 (45 mg, 1.2 mmol) in n-butanol (8 mL) was subjected to microwave irradiation (160 C, 20 min). The reaction mixture was quenched with NaHCO3 solution (2 mL) and extracted with Et0Ac (2x10 mL). The combined Et0Ac extract was washed with water (5 mL), brine (5 mL), was dried over Na2SO4 and was concentrated under reduced pressure. The residue was further purified by column chromatography (Si02, 60-120, Chloroform/methanol, 9/1) to give 1V4-(3-aminopheny1)-N6-(4-plienoxyphenyl)pyrimidine-4,6-diamine (87 mg, 37.8%) as a brown solid.
Step 2 To a solution of propionic acid (12 mg, 0.1 mmol) in DMF (0.6 mL) was added HATU (92 mg, 0.2 mmol) and the reaction mixture was stirred at room temperature for 30 min.
To it was added N4-(3-aminopheny1)-N6-(4-phenoxyphenyOpyrimidine-4,6-diamine (60 mg, 0.1 mmol) followed by DIPEA (41 mg, 0.3 mmol) and the reaction mixture was stirred at this temperature for 16 h. It was concentrated under reduced pressure. The residue was diluted with CH2C12 (5 mL) and was washed with NaHCO3 solution (2 mL), water (2 mL), and brine (2 mL). Drying over Na2SO4 followed by concentration under reduced pressure offered a residue that was purified by column chromatography (Si02, 230-400, chloroform/methanol: 9/1) to give N-(3-(6-(4-phenoxyphenylamino)pyrimidin-4-ylamino)phenyl)propionamide (IR-10) as a brown solid. 111 NMR (DMSO-d6) 8 ppm: 1.06 (t, J= 7.56 Hz, 3H), 2.30 (q, J= 7.48 Hz, 2H), 6.13 (s, IH), 6.94-6.99 (m, 4H), 7.08 (t, J= 7.36 Hz, 1H), 7.16-7.24 (m, 3H), 7.36 (t, J = 7.44 Hz, 2H), 7.54 (d, J =
8.88 Hz, 2H), 7.81 (s, 1H), 8.23 (s, 1H), 9.12 (s, 2H), 9.81 (s, 1H); LCMS:
m/e 426.3 (M+1).

Scheme 34a a H

1 )<
I HN 0 --.- 11 - CI 5. N2 ----"- NI
t4 40 step 1 N -- 0 step 2 II
N .51 kN- N kN--1 0 N N
H H H
[00316] Synthesis of (E)-N-(3-(6-(3-chloro-4-fluorophenoxy)pyrimidin-4-ylamino)pheny1)-4-(dimethylamino)but-2-enamide (1-92) Step 1 A solution of terr-butyl 3-(6-chloropyrimidin-4-ylamino)phenylcarbamate (1.6g, 5 mmol), 3-chloro-4-fluorophenol (1.4g, 10 mmol) and potassium carbonate (1.4g, 10 mmol) in 15 mL of DMF was heated to 120 C for 16h. The reaction mixture was mixed in 15 mL of water, the crude product was precipitated, filtered, purified by flash chromatography on silica gel with Me0H/DCM solvent system to afford 750 mg (45% yield) of NI-(6-(3-chloro-4-fluorophenoxy)pyrimidin-4-yl)benzene-1,3-diamine as an off-white solid. MS
(m/z): MH+ = 331.
Step 2 Oxaly1 chloride (155 mg, 1.2 mmol) was added dropwise to a mixture of 4-/V,N-dimethyl aminocrotonic acid HC1 salt (200mg, 1.2 mmol) in 5 mL of THF at 0 C. To this mixture was added 3 drops of DMF/THF solution (made from 5 drops of DMF in 1 mi., of THF).
The reaction mixture was stirred at RT for 2 h, then was cooled to 0 C ice bath. A
solution of NI-(6-(3-chloro-4-fluorophenoxy)pyrimidin-4-yl)benzene-1,3-diamine (200mg, 0.6 mmol) 2 mL of NMP) was added to the dimethylaminocrotonyl chloride solution and the resulting mixture was stirred for 3 h at 0 C. The reaction was quenched with 3 mL of IN NaOH, and was extracted with Et0Ac (2 x 25 mL). The crude product mixture was purified by flash chromatography on silica gel with Me0H/NH4OH/DCM solvent system to afford (E)-N-(3-(6-(3-chloro-fluorophenoxy)pyrirnidin-4-ylamino)pheny1)-4-(dimethylamino)but-2-enamide (1-92) as a light colored solid. MS (m/z): M-1-1+ =- 442, 444(3:1), H-NMR (DMSO) 8 10.08 (s, I
H), 9.67 (s, 1H), 8.36 (s, 1H), 7.98 (s, 1H), 7.59 (d, 1H), 7.57 (t, 1H), 7.53-7.24 (m, 4H), 6.29 (b, 1H), 6.23 (s, I H), 3.51 (d, 2H), 2.21 (s, 6H).

Scheme 35a N Bo c F F
0 NH CI NH Oy , NL NH _____ LN) N N step 1 N N step 2 N N
[00317] Synthesis of 1-(3-(6-(3-chloro-4-fluorophenyamino)pyrimidin-4-ylamino)pyrrolidin-1-yl)prop-2-en-1-one (1-70) Step 1 A neat mixture of tert-butyl 3-(6-chloropyrimidin-4-ylamino)pyrrolidin-1-carboxylate (354 mg, 1.18 mmol) and 3-chloro-4-fluoroaniline (3.03 g, 20.8 mmol) was heated at 140 C for 21 hours.
Upon cooling to ambient temperature, the melt was diluted with Et0Ac and the mixture was stirred for 1 hour. A beige, amorphous precipitate was collected, washed with water and dried in vacuo at 50-60 C giving 292 mg (80%) of N4-(3-chloro-4-fluorophenyI)-N6-(pyrrolidin-3-yl)pyrimidine-4,6-diamine. MS (APCI): (M + 1) = 308, (M ¨ 1) = 306.
Step 2, To a solution of N4-(3-chloro-4-fluoropheny1)-N6-(pyrrolidin-3-yppyrimidine-4,6-diamine (287 mg, 0.93 mmol) and triethylamine (0.32 ml, 2.33 mmol) in anhydrous THF (5 ml) under nitrogen was added acryloyl chloride (91 j.il, 1.12 mmol). The reaction mixture was stirred at room temperature for 1 hour and was concentrated under reduced pressure. The residue was eluted through a flash column (silica gel 60, 230-400 mesh, 5% Me0H in Et0Ac to 10%
Me0H in Et0Ac) to give two products. The less polar product (Rf = 0.24 in 1 : 9 Me0H :
Et0Ac) was found to be a diacrylated analog. The more polar product (Rf = 0.14 in 1 : 9 Me0H : Et0Ac) was 1-(3-(6-(3-chloro-4-fluorophenyamino)pyrimidin-4-y1 amino)pyrrol idin-1- yl)prop-2-en-one (I-70). MS (APCI) (M + 1)1- = 362, (M ¨ 1)1- = 360: ill-NMR DMSO-d6) 8 9.11 (s, 1H), 8.14 (s, 1H), 7.92 (d, 1H), 7.38-7.23 (m, 3H), 6.58-6.48 (m, 1H), 6.13-6.08 (m, IH), 5.75 (d, 1H) 5.63-5.59 (m, 1H), 3.64-3.59 (m, 2H), 2.17-1.81 (m, 6H).

Scheme 36a F
I
CFI lel NH
CI NH
NNk, N N
step 1 step 2 1003181 Synthesis of 1-(3-(6-(3-ehloro-4-fluorophenyamino)pyrimidin-4-ylamino)piperidin-1-ypprop-2-en-1-one (1-71) Step 1 N-(3-Chloro-4-fluoropheny1)-N'-piperidin-3-ylpyrimidine-4,6-diamine.
CI CI
Boc CI NH

N N) A mixture of tert-butyl 3-(6-chloropyrimidin-4-ylamino)piperidine-1-carboxylate (295 mg, 0.94 ramol) and 3-chloro-4-fluoroaniline (2.83 g, 19.4 mmol) was heated neat at 140 C for 19 hours.
Upon cooling, the melt was stirred in Et0Ac and allowed to stand at room temperature for 1 hour. The precipitate was collected, washed with Et0Ac and dried to give 256 mg (85%) of N4 -(3-chloro-4-fluoropheny1)-/V6-(piperidin-3-yl)pyrimidine-4,6-diamine, a grayish-violet amorphous solid. MS (APCI): (M + 1)+ = 322.
Step 2 To a solution of N4-(3-chloro-4-fluoropheny1)-1V6-(piperidin-3-y1 )pyrimidine-4,6-diamine (251 mg, 0.78 mmol) and triethylamine (0.27 ml, 1.95 mmol) in anhydrous THF (7 ml) under nitrogen was added acryloyl chloride (76 jil, 0.94 mmol). The reaction mixture was stirred at room temperature for 1 hour and was concentrated under reduced pressure. The residue was eluted through a flash column (silica gel 60, 230-400 mesh with 5% Me0H in Et0Ac to give two products. The less polar product (Rf = 0.33 in 1 : 9 Me0H : Et0Ac) was found to be a diacrylated analog. The more polar product (Rf = 0.24 in 1 : 9 Me0H : Et0Ac) was 1434643-chloro-4-fluoroph enyamino)pyrimidin-4-ylamino)piperidin- 1-yl)prop-2-en-1-one (1-71). MS
(APO) (M + 1)4 = 376, (M ¨ 1)4 = 374: 1H-NMR DMSO-d6, .5 9.15 (br s, 1H), 8.18 (d, 1H), 7.95 (br s, 1H), 7.43-7.20 (m, 2H), 7.03-6.48 (m, 3H), 6.26-5.97 (m, 2H), 5.86-5.51 (m, 3H), 3.90-3.66 (m, 2H), 1.98-1.66 (m, 2H), 1.59-1.12 (m, 2H).

Scheme 37a Co el Co ?
r-o H tip -0 step 1 0 OH step 2 tr. N rsi NH2 step 3 0 N N

1003191 Synthesis of N-(6-(6-(2,3-dihydrobenzo [b][1,4]dioxin-6-yloxy)pyrimidin-4-ylamino)pyridin-2-yi)acrylamide (I-95) Step 1 To a stirred solution of 2,3-dihydrobenzo[b][1,4]dioxine-6-carbaldehyde (1 g, 6.09 mmol) in CH2C12 (16 mL) was added m-CPBA (4.204 g, 24.36 mmol). The suspension was heated at 50 C
for 2 days, was cooled to rt, was quenched with saturated NaHCO3 soln. and was extracted with CH2C12 (3x10 mL). The combined extract was concentrated under reduced pressure, and then was dissolved in Me0H containing NaOH. The solution was stirred at rt for 2 h, was acidified with HC1 and was extracted with ethyl acetate (3x10 mL). The combined extract was washed with saturated NaHCO3 solution and brine, was dried over anhydrous Na2SO4, was filtered and was concentrated under reduced pressure. The residue was dissolved in CH2C12 and was filtered.
The DCM solution was dried over anhydrous Na2SO4, was filtered and was concentrated under reduced pressure to give 2,3-dihydrobenzo[b][1,4]dioxin-6-ol (0.591 g, 63%) as a reddish brown oily liquid.

Step 2 A stirred mixture of 2,3-dihydrobenzo[b][1,4]dioxin-6-ol (0.137 g, 0.90 mmol), Cs2CO3 (0.734 g, 2.25 mmol), Cul (0.02 g, 10% w/w), and N2-(6-chloropyrimidin-4-yl)pyridine-2,6-diamine (0.29 g, 0.90 mmol) in NMP (1 mL) was heated at 100 C for 16 h. The reaction mixture was cooled to rt and slowly added to demineralised water. The solid that precipitated was collected by filtration and was further purified by column chromatography (Si02, 60-120, pet ether/ethyl acetate, 7/3) to give N2-(6-(2,3-dihydrobenzo[b] [1,4]dioxin-6-yloxy)pyrimidin-4-yl)pyridine-2,6-diarnine (0.088 g, 29%) as yellow solid.
Step 3 To a stirred solution of N2-(6-(2,3-dihydrobenzo [b][1,4]dioxin-6-yloxy)pyrimidin-4-yl)pyridine-2,6-diamine (0.080 g, 0.23 mmol) and potassium carbonate (0.065 g, 0.47 mmol) in NMP (0.8 mL) at 0 C was added acryloyl chloride (0.026 g, 0.29 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 was stirred at 0 C for 30 min. A white solid was isolated by filtration. The solid was washed with cold water and hexane and dissolved in a 2 mL of methanol/dichloromethane (1/1). This solution was 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 (2 x 5 mL). The combined ethyl acetate extract was washed with water (2 mL) and brine (2 mL), was dried over Na2SO4 and was concentrated under reduced pressure to obtain N-(6-(6-(2,3-dihydrobenzo[b][1,41dioxin-6-yloxy)pyrimidin-4-ylamino)pyridin-2-ypacrylamide (I-95) as a light yellow solid. 1H NMR (DMSO-d6) 5 ppm: 4.25 (s, 4H), 5.79 (dd, J = 1.84 &
10.08 Hz, 1H), 6.30 (dd, J = 1.84 & 16.96 Hz, 1H), 6.62-6.72 (m, 3H), 6.88 (d, J = 8.72 Hz, 1H), 7.03 (d, J= 7.84 Hz, 1H), 7.69 (t, J= 7.96 Hz, 1H), 7.70 (d, J= 7.84 Hz, 1H), 7.85 (s, 1H), 8.32 (d, J= 0.4 Hz, 1H), 10.16 (s, 1H), 10.47 (s, 1H); LCMS: m/e 392.
1003201 Described below are assays used to measure the biological activity of provided compounds as inhibitors of ErbB1 (EGFR), ErbB2, ErbB4, TEC, BTK, ITK, BMX, and JAK3.

Cloning, Expression and Purification of EGFR-WT and EGFR C797S Mutant Using Baculovirus and Insect Cells -I,Yubclonine of EGFR-WT and mutant kinase domains 1803211 Amino acids 696 to 1022 of the EGFR-WT kinase domain (NM_005228, NP_005219.2) was subcloned into the Ncol and HindlII sites of the pFastHTa vector (Invitrogen.,m 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.
(it) Expression 1003221 P1 baculovims stocks were generated in SF9 cells via Blue Sky Biotech's suspension transfection protocol (Worcester, MA). Expression analysis was conducted in 125 ml culture of SF21 insect cells ((grown in SF900I SFM (Invitrogen cat # 10902-088), supplemented with 10mg/L gentamicin (Invitrogen, Carlsbad, CA, cat# 15710-064)) using a viral load of 0.1m1 of virus per 100 ml of cell suspension. Expression was optimized using Blue Sky Biotech's Infection Kinetics Monitoring system (Worcester, MA)_ (iii) Purification (00323] 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 ul 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 2m1 column. The column was washed with 2 ml of wash buffer (Blue Sky Biotech, Worcester, MA, 1X WX, 25mM irnidazole ).The protein was eluted with 1X WX + imidazole at varying concentrations:
Elution 1: 75 mM imidazole ( 2 fractions, 1 column volume); Elution 2 : 150 m1V1 imidazole (2 fractions, 1 column volume); Elution 3: 300 niM imidazole (2 fractions, 1 column volume). All the elution fractions were analyzed by SOS page followed by Coomassie staining and Western Blotting using anti-petite-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 SoeqrometrY for EGFR
100324J EGFR wild type and EGFR mutant (C797S) was incubated with 10-fold excess of compound I-1 for 1 hr and 3 hrs. 1 ul aliquots of the samples (total volume 5-8 ul) were 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 was only observed for the wild type EGFR
with a new peak appearing at a mass consistent with a single site covalent modification with compound 1-1 which has a mass of 410 Da. (See Figure 8). The mutant EGFR (C797S) showed no significant reactivity even after 3 hrs, confirming modification of the eysteine of interest, Cys797.

Omnia Assay Protocol for Potency Assessment Against EGFR (WT) and EGFR
(T790M/L858R) Active Enzymes 1003251 The protocol below describes continuous-read Icinase assays to measure inherent potency of compounds against active forms of EGFR (WT) and EGFR (T790M/L858R) enzymes. Description of the mechanics of the assay platform may be obtained from the vendor (Invitrogen, Carlsbad, CA).
(003261 Briefly, 10X stocks of EGFR-WT (PV3872) from Invitrogen and EGFR-T790MTL858R (40350) from BPS Bioscience, San Diego, CA, 1.13X ATP (AS001A) and appropriate Tyr-Sox conjugated peptide substrates (KCZ1001) were prepared in 1X lcinase reaction buffer consisting of 20 mM Tris, pH 7.5, 5 mM MgC12, 1 mM EGTA, 5 mM

glycerophosphate, 5% glycerol (10X stock, 10002A) and 0.2 mM DTT (DS001A). 5 !IL of TM
each enzyme were pre-incubated in a Corning (#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% DIVISO and serially diluted compounds prepared in 50% .DMSO. Kinase reactions were started with the addition of 45 pL of the ATP/Tyr-Sox peptide substrate mix and monitored every 30-90 seconds for 60 minutes at X,,,360/),485 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 1050 from log[Inhibitor] vs Response, Variable Slope model in GraphPad Prism from GraphPad Software (San Diego, CA).
[00327] The EGFR-WT- and EGFR T790M/L858R-modified optimized reagent conditions are:
[EGFR-WT] =5 nIVI, [ATP] --- 15 ralvI, [Y12-Sox] =5 mM (ATP KMapp ¨ 12 mM);
and [EGFR-T790M/L858R] = 3 nM, [ATP] = 50 mM, [Y12-Sox] = 5 mIVI (ATP KMapp ¨45 mM).

[00328] Table 7 shows the activity of selected compounds of this invention in the EGFR
inhibition assay. The compound numbers correspond to the compound numbers in Table 5.
Compounds having an activity designated as "A" provided an 1050 <10 nM;
compounds having an activity designated as "B" provided an 1050 10-100 nM; compounds having an activity designated as "C" provided an IC50 of 100-1000 n/vl; compounds having an activity designated as "D" provided an ICso of 1000-10,000 nM; and compounds having an activity designated as "E" provided an IC59 >10,000 nM.
Table 7. EGFR Wild Type and EGFR (mutant C797S) Inhibition Data EGFR
EGFR
Compound #(T790M/L858R) inhibtion inhibiton EGFR
Compound # EGFR (T790M/1,858R) inhibtion inhibiton EGFR
Compound # EGFR (T790M/L858R) inhibtion inhibiton Cellular Assays for EGFR Activity [00329] 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 liAM 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. Figure 1 depicts the EGFR inhibiting activity of compound I-1.
[00330] 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 0.0014M test compound for 1 hr. Cells were then stimulated with 100 ng/ml EGF for 5 mm, and extracts were made as described in Fry, et al. 2Oug total protein from lysates were loaded on gel and blots were probed for either EGFR phosphorylation or p42/p44 Erk phosphorylation.
[00331] Dose response inhibition of EGFR phosphorylation and p42/p44 Erk phosphorylation with compound 1-16 and 1-17 in A431 cells is depicted in Figure 3. Dose response inhibition of EGFR phosphorylation and p42/p44 Erk phosphorylation with compound 1-19 in A431 cells is depicted in Figure 4. Dose response inhibition of EGFR phosphorylation with compound I-1 in A431 cells, as compared with its "reversible control" compound (IR-3), is depicted in Figure 5.

Washout Experiment for EGFR Activity [00332] 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 p.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 the compound with warmed compound-free medium, incubated for 2 hr, washed again, incubated another 2 hr, washed again, and then incubated another 2hr washed again and incubated for additional 2 hr and then stimulated with EGF. The results of this experiment are depicted in Figure 6 where it is shown that compound I-1 maintains enzyme inhibition after "washout" whereas its "reversible control" compound (IR-3) was washed away in the experiment thereby resulting in reactivated enzyme activity.

Mass Spectrometry for ErbB4 [00333] ErbB4 lcinase domain (Upstate) was incubated with compound for 90 minutes at 10-fold excess of compound I-1 to protein. 1 ul aliquots of the samples (total volume of 4.24u1) were 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).
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).

[00334] Intact ErbB4 protein occurs at MH+ of 35850 with corresponding sinapinic (matrix) adducts occurring about 200 Da higher. A stochiometric incorporation of the compound I-I (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 compound I-I as depicted in Figure 7.

ErbBl, ErbB2 and/or ErbB4 Kinase Inhibition [00335] 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 hivitrogen Corp (1nvitrogen Corporation, 1600 Faraday Avenue, Carlsbad, California, CA;
http://www.invitrogen.com/downloads/Z-LYTE_Brochure_1205.pdf) using the Z"-LYTErm 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.

[00336] Table 8 shows the activity of selected compounds of this invention in the ErbB
inhibition assay. The compound numbers correspond to the compound numbers in Table 5.
Table 8. ErbB1, ErbB2, and/or ErbB4 Inhibition Data ErbB1 ErbB2 ErbB4 Compound #
A Inhibition % Inhibition % Inhibition 11 99 76% @ 1 M 86% @ 1 M
-"Yo@ nM
61%@ 100 nM 64% @ 100 nM
98% @ 100 nM 96% @ 1 M
1-2 75% @ 1 M
75% @ 10 nM 39% @ 100 nM
96% @ nM
1-3 100 89% @ 1 M 95% M
56% @ 10 nM
1-4 100% @ 10 nM 86% @ 1 M 78% @ 1 M
1-5 lam@ 10 nM 86% @ 1 M 95% @ 1 M
100%! 100 nM
1-6 84% @ 1 tiM 97% 1 M
49% @ 10 nM
100% @ 100 nM
1-7 89% @ 1 !AM 100% @ 1 M
53%@lOnM
1-8 83% @ 10 nM 57% @ 1 M
1-9 100% @ 75% @ 1 p.M
1-10 96% @ 1 M

ErbB1 ErbB2 ErbB4 Compound #
A Inhibition A Inhibition % Inhibition I-11 79%@ 1 M
1-12 .82% @ 1 M
1-13 92% @ 1 M 98% @ 1 1-14 96% @ 1 !AM
1-15 98% @ 1 M
87% @ 100 nM 93% @ 100 nM
I-16 98Vo @ 1 1.11µ,4 24% @ 10 nM 26% @ 10 TIM
1-17 95% @ 1 M 89% @ 1 M 94% @ 1 p.M
1-18 91%@ 1 p.N1 94% @ 1 M
1-19 96% @ 1 M 98% @ 1 M 97% @ 1 M

Mass Spectrometry for TEC Kinase [00337] TEC kinase (45 pmols; Invitrogen) was incubated with (I-13) (450 pmols) for 3hrs at 10X access prior to tryptic digestion. Iodoacetamide was used as the alkylating agent after compound incubation. A control sample (45 pmols) was also prepared which did not have the addition of (I-13). For tryptic digests a Sul 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/m1 in 0.1%TFA:Acetonitrile 50:50).
[00338] As depicted in Figure 11, the expected peptide (GCLLNFLR) to be modified was immediately evident at MH+ of 1358.65. This is the mass to be expected when compound 1-13, with an adduct mass of 423.17, 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 1-13 indicating that the reaction was complete. There was no evidence of any other modified peptides.
[00339] Evidence of compound 1-13 was observed at MH+ of 424.20 in the low mass range of the spectra. The fragmentation spectra of the 424.20 peak did show many diagnostic fragments that were apparent in the PSD spectra of the modified peptide at 1358.65 (see Figure 11).
[00340] To further verify the presence of the modified peptide with compound 1-13, the peptide at MH+ of 1358.65 was subjected to PSD (MS/MS) analsysis. A
correlational analysis with the homosapien database identified the correct peptide modified by 1-13.

Instrumental:
[00341] 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 BTK
[00342] The Omnia Assay Protocol for potency assessment against BTK is performed in a substantially similar manner as that described in Example 25 above except that the modified BTK-optimized reagent conditions are:
[BTK] =5 nM, [ATP] =40 mM, [Y5-Sox] = 10 mM (ATP KMapp ¨ 36 mM).

[00343] Table 9 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 ICso of 100-1000 nM; compounds having an activity designated as "D" provided an ICso of 1000-10,000 nM; and compounds having an activity designated as "E" provided an ICso >10,000 nM.
Table 9. BTK Inhibition Data Compound # BTK Inhibition Compound # BTK Inhibition Compound # BTK Inhibition BTK Ramos Cellular Assay 1003441 Compounds 1-13 and (1-52) were assayed in Ramos human Burlcitt lymphoma cells.
Ramos cells were grown in suspension in T225 flasks, spun down, resuspended in 50mls 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 0.001uM. 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. 16ug total protein from lysates was loaded on gels and blots were probed for phosphorylation of the BTK substrate PLCy2. At 1 JIM 1-13 showed 85 % inhibition and (1-52) showed 50% inhibition of BTK
signaling in Ramos cells. Additional dose response inhibition of BTK signaling with 1-13 is depicted in Figure 9.
1003451 Table 10 provides inhibition data for selected compounds in Ramos cells. 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 of 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 ICso of >1000 nM.

Table 10. BTK Ramos Cellular Inhibition Data Compound # BTK Inhibition (nM) Washout Experiment with Ramos cells for BTK Activity [00346] Ramos cells were serum starved for one hour in RPM! media +1%
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 FNN0011) supplemented with Roche complete protease inhibitor tablets (Roche 11697498001) and phosphatase inhibitors (Roche 04 906 837 001) and 18 ug total protein lysate was loaded in each lane. Inhibition of Btk Icinase activity was assayed by measuring its substrate (PLCy2) phosphorylation by western blot with phospho-specific antibodies from Cell Signaling Technologies cat# 3871. Figure 10 depicts the results of compound 1-13 in the washout experiment at 0 hour, 4 hour, 6 hour, and 8 hour time points. As shown in Figure 10, compound 1-13 maintains inhibition of BTK for 8 hours.
[00347] Table 11 provides data for selected compounds in the Ramos washout assay.
Table 11. BTK Washout Data Compound # BTK Inhibition Type 1-13 irreversible 1-63 irreversible 1-64 irreversible Compound # BTK Inhibition Type 1-65 reversible 1-66 irreversible 1-82 irreversible Mass Spectrometry for BTK
[003481 Intact BTK was incubated for 1 hr at a 10x fold excess of compound 1-63 or 1-66 to protein. Aliquots (2 ul) of the samples were diluted with 10u1 of 0.1% TFA
prior to micro C4 ZipTipping directly onto the MALDI target using Sinapinic acid as the desorption matrix (10mg/m1 in 0.1%TFA:Acetonitrile 20:80). Mass spectrometry traces are shown in Figure 12 and Figure 13. The top panels of Figures 12 and 13 show the mass spec trace of the intact BTK
protein (m/z 81,169 Da). The bottom panels show the mass spec trace when BTK
was incubated with compound 1-63 (mw 424.5) or compound 1-66 (mw 425.5) in Figures 12 and 13, respectively. The centroid mass (m/z 81,593 kDa) in the bottom panel of Figure
12 shows a positive shift of about 424 Da, indicating complete modification of BTK by compound 1-63.
The centroid mass (m/z 81,593 kDa) in the bottom panel of Figure 13 shows a positive shift of about 407 Da, indicating complete modification of BTK by compound 1-66.

Omnia Assay Protocol for Potency Assessment Against Active Forms of ITK Kinase 100349) This example describes continuous-read kinase assays to measure inherent potency of compound against active forms of ITK enzymes as described in Example 10 above except that the modified ITK-optimized reagent conditions are:
[ITK] = 10 nM, [ATP] =25 p.M, [Y6-Sox] = 10 p.M (ATP Kmopp = 33 pM).

1003501 Table 12 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 1050 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 1050 >10,000 nM.
Table 12. ITK Inhibition Data Compound # ITK inhibition Omnia Assay Protocol for Potency Assessment Against Active Forms of BMX Kinase 100351] This example describes continuous-read lcinase assays to measure inherent potency of compound against active forms of BMX enzymes as described in Example 10 above except that the modified BMX-optimized reagent conditions are:
[BMX] = 2.5 nM, [ATP] = 100 M, [Y5-Sox] = 7.5 1.i.M (ATP Kmapp = 107 M).

1003521 Table 13 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 1050 <10 nM;
compounds having an activity designated as "B" provided an IC50 10-100 nM; compounds having an activity designated as "C" provided an 1050 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 1050 >10,000 nM.

Table 13. BMX Inhibition Data Compound # BMX inhibition Omnia Assay Protocol for Potency Assessment A2ainst the Active Form of Janus-3 Kinase (JAK3):
[00353] The Omnia Assay Protocol for potency assessment against JAK3 is performed in a substantially similar manner as that described in Example 25 above except that the modified JAK3-optimized reagent conditions are:
[JAK3] = 5 nM, [ATP] = 5 tM, [Y12-Sox] = 5 1.1M (ATP KMapp ¨5 1.1M).

[00354] Table 14 shows the activity of selected compounds of this invention in the JAK3 inhibition assay. The compound numbers correspond to the compound numbers in Table 5.
Compounds having an activity designated as "A" provided an IC50 <10 nM;
compounds having an activity designated as "B" provided an IC50 10-100 nM; compounds having an activity designated as "C" provided an IC50 of 100-1000 nM; compounds having an activity designated as "D" provided an IC50 of 1000-10,000 nM; and compounds having an activity designated as "E" provided an IC50 >10,000 nM.
Table 14. JAK3 Inhibition Data Compound #
Inhibition Compound #
Inhibition Compound #
Inhibition 1-92 B .

[00355] While we have described a number of embodiments of this invention, it is apparent that our 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 (60)

169We claim:
1. A compound of formula I:
or a pharmaceutically acceptable salt thereof, wherein:
Ring A is an optionally substituted group selected from phenyl, an 8-10 membered bicyclic partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 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 phenyl, a 5-6 membered heteroaryl ring having 1-3 heteratoms independently selected from N, O or S, a 5-6 membered saturated heterocyclic ring having 1-2 heteroatoms independently selected from N, O or S, or an 8-10 membered bicyclic partially unsaturated or aryl ring having 1-3 heteroatoms independently selected from N, O or S;
R1 is a warhead group;
R y is hydrogen, halogen, CN, lower alkyl, or lower haloalkyl;
G is CH, or N;
W is a bivalent C1-3 alkylene chain wherein one methylene unit of W is optionally replaced by -NR2-, -N(R2)C(O)-, -C(O)N(R2)-, -N(R2)SO2-, -SO2N(R2)-, -O-, -C(O)-, -OC(O)-, -C(O)O-, -S-, -SO- or -SO2-;
R2 is hydrogen or optionally substituted C1 -6 aliphatic, or:
R2 and a substituent on Ring A are taken together with their intervening atoms to form a 4-6 membered saturated ring, or:
R2 and R y are taken together with their intervening atoms to form a 4-7 membered carbocyclic ring;

m is 0-4;
each R x is independently selected from -R, halogen, -OR, -CN, -NO2, -SO2R, -SOR, -C(O)R, -CO2R, -C(O)N(R)2, -NRC(O)R, -NRC(O)NR2, -NRSO2R, or -N(R)2; or:
R x and R1 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 aliphatic; and 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.
2. The compound according to claim 1, wherein the compound is of formula IL
wherein:
W is -NR2-, -S-, or -O-;
R2 is hydrogen or optionally substituted C1-6 aliphatic, or:
R2 and a substituent on Ring A are taken together with their intervening atoms to form a 4-6 membered saturated ring; and G is CH or N.
3. The compound according to claim 2, wherein Ring A is selected from:

4. The compound according to claim 1, wherein Ring B is selected from:

5. The compound according to claim 2, wherein m is 1, 2, 3, or 4.
6. The compound according to claim 1, wherein said compound is of formula III or IV:
or a pharmaceutically acceptable salt thereof.
7. The compound according to claim 1, wherein said compound is of formula I-i, I-ii, or I- or a pharmaceutically acceptable salt thereof.
8. The compound according to claim 1, wherein said compound is of formula II-ii, II-ii, or II-iii:
or a pharmaceutically acceptable salt thereof.
9. The compound according to claim 1, wherein said compound is of formula i, III-a-ii, or III-b-ii, III-a-iii, or III-b-iii:
or a pharmaceutically acceptable salt thereof.
10. The compound according to claim 1, wherein said compound is of formula IV-a-i, IV-b-i, IV-a-ii, IV-b-ii, IV-a-iii, or IV-b-iii:
or a pharmaceutically acceptable salt thereof
11. The compound according to claim 1, wherein said compound is of formula II-a-iv, II-b-iv, II-a-v, II-b-v, II-a-vi, or ll-b-v:
or a pharmaceutically acceptable salt thereof.
12. The compound according to claim 1, wherein R1 is -L-Y, wherein:
L is a bivalent C2-8 straight or branched, hydrocarbon chain wherein L has at least one double bond and one or two additional methylene units of L are optionally and independently replaced by -NRC(O)-, -C(O)NR-, -N(R)SO2-, -SO2N(R)-, -S-, -S(O)-, -SO2-, -OC(O)-, -C(O)O-, cyclopropylene, -O-, -N(R)-, or -C(O)-;
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 1-4 R e groups; and each R e is independently selected from -Q-Z, oxo, NO2, halogen, CN, a suitable leaving group, or C3-6 aliphatic optionally substituted with oxo, halogen, NO2, or CN, wherein:
Q is a covalent bond or a bivalent C1 saturated or unsaturated, straight or branched, hydrocarbon chain, wherein one or two methylene units of Q are optionally and independently replaced by -N(R)-, -S-, -O-, -C(O)-, -OC(O)-, -C(O)O-, -SO-, or -SO2-, -N(R)C(O)-, -C(O)N(R)-, -N(R)SO2-, or -SO2N(R)-; and Z is hydrogen or C1-6 aliphatic optionally substituted with oxo, halogen, NO2, or CN.
13. The compound according to claim 12, wherein:
L is a bivalent C2-8 straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by -C(O)-, -NRC(O)-, -C(O)NR-, -N(R)SO2-, -SO2N(R)-, -S-, -S(O)-, -SO2-, -OC(O)-, or -C(O)O-, and one or two additional methylene units of L are optionally and independently replaced by cyclopropylene, -O-, -N(R)-, or -C(O)-; and Y is hydrogen or C1-6 aliphatic optionally substituted with oxo, halogen, NO2, or CN.
14. The compound according to claim 13, wherein L is a bivalent C2-8 straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by -C(O)-, and one or two additional methylene units of L are optionally and independently replaced by cyclopropylene, -O-, -N(R)-, or -C(O)-.
15. The compound according to claims 13, wherein L is a bivalent C2-8 straight or branched, hydrocarbon chain wherein L has at least one double bond and at least one methylene unit of L is replaced by -OC(O)-.
16. The compound according to claim 12, wherein L is -NRC(O)CH=CH-, -NRC(O)CH=CHCH2N(CH3)-, -NRC(O)CH=CHCH2O-, -CH2NRC(O)CH=CH-, -NRSO2CH=CH-, -NRSO2CH=CHCH2-, -NRC(O)(C=N2)-, -NRC(O)(C=N2)C(O)-, -NRC(O)CH=CHCH2N(CH3)-, -NRSO2CH=CH-, -NRSO2CH=CHCH2-, -NRC(O)CH=CHCH20-, -NRC(O)C(=CH2)CH2-, -CH2NRC(O)-, -CH2NRC(O)CH=CH-, -CH2CH2NRC(O)-, or -CH2NRC(O)cyclopropylene-; wherein R is H or optionally substituted C1-6 aliphatic; and Y is hydrogen or C1-6 aliphatic optionally substituted with oxo, halogen, NO2, or CN.
17. The compound according to claim 16, wherein L is -NHC(O)CH=CH-, -NHC(O)CH=CHCH2N(CH3)-, -N1-iC(O)CH=CHCH2O-, -CH2NHC(O)CH=CH-, -NHSO2CH=CH-, -NHSO2CH=CHCH2-, -NHC(O)(C=N2)-, -NHC(O)(C=N2)C(O)-, -NHC(O)CH=CHCH2N(CH3)-, -NHSO2CH=CH-, -NHSO2CH=CHCH2-, -NHC(O)CH=CHCH2O-, -NHC(O)C(=CH2)CH2-, -CH2NHC(O)-, -CH2NHC(O)CH=CH-, -CH2CH2NHC(O)-, or -CH2NHC(O)cyclopropylene-.
18. The compound according to claim 12, wherein L is a bivalent C2-8 straight or branched, hydrocarbon chain wherein L has at least one alkylidenyl double bond and at least one methylene unit of L is replaced by -C(O)-, -NRC(O)-, -C(O)NR-, -N(R)SO2-, -SO2N(R)-, -S-, -SO2-, -OC(O)-, or -C(O)O-, and one or two additional methylene units of L are optionally and independently replaced by cyclopropylene, -O-, -N(R)-, or -C(O)-.
19. The compound according to claim 1, wherein R1 is -L-Y, wherein:
L is a bivalent C2-8 straight or branched, hydrocarbon chain wherein L has at least one triple bond and one or two additional methylene units of L are optionally and independently replaced by -NRC(O)-, -C(O)NR-, -N(R)SO2-, -SO2N(R)-, -S-, -S(O)-, -SO2-, -OC(O)-, or -C(O)O-, Y is hydrogen, 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 1-4 R e groups; and each R e 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-, -O-, -C(O)-, -OC(O)-, -C(O)O-, -SO-, or -SO2-, -N(R)C(O)-, -C(O)N(R)-, -N(R)SO2-, or -SO2N(R)-; and Z is hydrogen or C1-6 aliphatic optionally substituted with oxo, halogen, NO2, or CN.
20. The compound according to claim 19, wherein Y is hydrogen or C1-6 aliphatic optionally substituted with oxo, halogen, NO2, or CN.
21. The compound according to claim 20, wherein L is -C.ident.C-, -C.ident.CCH2N(isopropyl)-, -NHC(O)C.ident.CCH2CH2-, -CH2-C.ident.C-CH2-, -C.ident.CCH2O-, -CH2C(O)C.ident.C-, -C(O)C.ident.C-, or -CH2OC(=O)C.ident.C-.
22. The compound according to claim 1, wherein R1 is -L-Y, wherein:
L is a bivalent C2-8 straight or branched, hydrocarbon chain wherein one methylene unit of L is replaced by cyclopropylene and one or two additional methylene units of L are independently replaced by -NRC(O)-, -C(O)NR-,' -N(R)SO2-, -SO2N(R)-, -S-, -S(O)-, -SO2-, -OC(O)-, or 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 1-4 R e groups; and each R e 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-, -O-, -C(O)-, -OC(O)-, -C(O)O-, -SO-, or -SO2-, -N(R)C(O)-, -C(O)N(R)-, -N(R)SO2-, or ¨SO2N(R)-; and Z is hydrogen or C1-6 aliphatic optionally substituted with oxo, halogen, NO2, or CN.
23. The compound according to claim 22, wherein Y is hydrogen or C1-6 aliphatic optionally substituted with oxo, halogen, NO2, or CN.
24. The compound according to claim 1, wherein R1 is ¨L-Y, wherein:
L is a covalent bond, -C(O)-, -N(R)C(O)-, or a bivalent C1-8 saturated or unsaturated, straight or branched, hydrocarbon chain; and Y is selected from the following (i) through (xvii):
(i) C1-6 alkyl substituted with oxo, halogen, NO2, or CN;
(ii) C1-4 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 R e groups; or (v) a saturated 5-6 membered heterocyclic ring having 1-2 heteroatom selected from oxygen or nitrogen wherein said ring is substituted with 1-4 R e groups; or (vi) , wherein each R, Q, Z; or (vii) a saturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 R e groups; or (viii) a partially unsaturated 3-6 membered monocyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 R e groups; or (ix) a partially unsaturated 3-6 membered carbocyclic ring, wherein said ring is substituted with 1-4 R e groups;

(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 R e groups; or (xiii) a 6-membered aromatic ring having 0-2 nitrogens wherein said ring is substituted with 1-4 R e groups; or wherein each R e is as defined above and described herein; or (xv) a 5-membered heteroaryl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-3 R e groups; or (xvii) an 8-10 membered bicyclic, saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, wherein said ring is substituted with 1-4 R e groups;
wherein:

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;
each R e 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-, -O-, -C(O)-, -OC(O)-, -C(O)O-, -SO-, or -SO2-, -N(R)C(O)-, -C(O)N(R)-, -N(R)SO2-, or ¨SO2N(R)-; and Z is hydrogen or C1-6 aliphatic optionally substituted with oxo, halogen, NO2, or CN.
25. The compound according to claim 24, wherein L is a covalent bond, ¨CH2-, -NH-, -C(O)-, -CH2NH-, -NHCH2-, ¨NHC(O)-, -NHC(O)CH2OC(O)-, -CH2NHC(O)-, -NHSO2-, -NHSO2CH2-, -NHC(O)CH2OC(O)-, or -SO2NH-.
26. The compound according to claim 25, wherein L is a covalent bond.
27. The compound according to any of claims 24, 25, and 26, wherein Y is selected from:
wherein each R e is independently selected from halogen.
28. The compound according to claim 1, wherein R1 is selected from:

wherein each R e is independently a suitable leaving group, NO2, CN, or oxo.
29. A compound selected from the group consisting of:
or a pharmaceutically acceptable salt thereof
30. A compound selected from the group consisting of:

or a pharmaceutically acceptable salt thereof.
31. A
composition comprising a compound according to any one of claims 1 through 30, and a pharmaceutically acceptable adjuvant, carrier, or vehicle.
32. The composition according to claim 31, in combination with an additional therapeutic agent.
33. The composition according to claim 32, wherein the additional therapeutic agent is a chemotherapeutic agent.
34. A method for inhibiting one or more of ErbB1, ErbB2, ErbB3, or ErbB4, or a mutant thereof, activity in a biological sample comprising the step of contacting said biological sample with a compound according to any of claims 1 through 30 or a composition according to claim 31.
35. A method for inhibiting one or more of ErbB1, ErbB2, ErbB3, or ErbB4, or a mutant thereof, activity in a patient comprising the step of administering to said patient a compound according to any of claims 1 through 30 or a composition according to claim 31.
36. The method according to either of claims 34 or 35, wherein the one or more of ErbB1, ErbB2, or ErbB4, or a mutant thereof, activity is inhibited irreversibly.
37. The method according to any one of claims 34, 35, or 36, wherein the one or more of ErbB1, ErbB2, or ErbB4, or a mutant thereof, activity is inhibited irreversibly by covalently modifying Cys797 of ErbB1, Cys805 of ErbB2 or Cys803 of ErbB4.
38. A method for treating an ErbB1-, ErbB2-, ErbB3-, or ErbB4-mediated disorder in a patient in need thereof, comprising the step of administering to said patient a compound according to any of claims 1 through 30 or a composition according to claim 31.
39. The method according to claim 38, wherein the disorder is a carcinoma selected from breast cancer, glioblastoma, lung cancer, cancer of the head and neck, colorectal cancer, bladder cancer, non-small cell lung cancer, squamous cell carcinoma, salivary gland carcinoma, ovarian carcinoma, or pancreatic cancer.
40. The method according to claim 38, wherein the disorder is selected from neurofibromatosis type I (NF1), neurofibromatosis type II (NF2) Schwann cell neoplasms (e.g.
MPNST's), or a Schwannoma.
41. A method for inhibiting one or more TEC-kinases, or a mutant thereof, activity in a biological sample comprising the step of contacting said biological sample with a compound according to any of claims 1 through 30 or a composition according to claim 3 L
42. A method for inhibiting one or more TEC-kinases, or a mutant thereof, activity in a patient comprising the step of administering to said patient a compound according to any of claims 1 through 30 or a composition according to claim 31.
43. The method according to either of claims 41 or 42, wherein the TEC-kinase, or a mutant thereof, activity is inhibited irreversibly.
44. The method according to claim 43 wherein the TEC-kinase is selected from one or more of TEC, ITK or BMX.
45. The method according to claim 44, where the one or more of TEC, ITK or BMX activity is inhibited irreversibly by colvalently modifying Cys 449 of TEC, Cys 442 of ITK or Cys 496 of BMX.
46. A method for treating a TEC-kinase-mediated disorder in a patient in need thereof, comprising the step of administering to said patient a compound according to any of claims 1 through 30 or a composition according to claim 31.
47. The method according to claim 46, wherein the disorder is an autoimrnune disorder, an inflammatory disorder, a proliferative disorder, a hyperproliferative disease, an immunological-mediated diseases, a disease of the respiratory tract, a disease of the bone and joints, a skin disorder, a gastrointestinal disorder, a systemic disease, or allograft rejection.
48. A method for inhibiting BTK, or a mutant thereof, activity in a biological sample comprising the step of contacting said biological sample with a compound according to any of claims 1 through 30 or a composition according to claim 31.
49. A method for inhibiting BTK, or a mutant thereof, activity in a patient comprising the step of administering to said patient a compound according to any of claims 1 through 30 or a composition according to claim 31.
50. The method according to either of claims 48 or 49, wherein the BTK, or a mutant thereof, activity is inhibited irreversibly.
51. The method according to claim 50, wherein the BTK, or a mutant thereof, activity is inhibited irreversibly by covalently modifying Cys 481 of BTK.
52. A method for treating a BTK-mediated disorder in a patient in need thereof, comprising the step of administering to said patient a compound according to any of claims 1 through 30 or a composition according to claim 31.
53. The method according to claim 52, wherein the disorder is an autoimmune disease, a heteroimmune disease, an inflammatory disease, a cancer, or a thromboembolic disorder.
54. The method according to claim 53, wherein the disorder is selected from rheumatoid arthritis, multiple sclerosis, B-cell chronic lymphocytic leukemia, hairy cell leukemia, non-Hodgkin's lymphoma, irritable bowel syndrome, Crohn's disease, lupus or disorders associated with renal transplant.
55. A method for inhibiting JAK3, or a mutant thereof, activity in a biological sample comprising the step of contacting said biological sample with a compound according to any of claims 1 through 30 or a composition according to claim 31.
56. A method for inhibiting JAK3, or a mutant thereof, activity in a patient comprising the step of administering to said patient a compound according to any of claims 1 through 30 or a composition according to claim 31.
57. The method according to either of claims 55 or 56, wherein the JAK3, or a mutant thereof, activity is inhibited irreversibly.
58. The method according to claim 57, wherein the JAK3, or a mutant thereof, activity is inhibited irreversibly by covalently modifying Cys 909 of JAK3.
59. A method for treating a JAK3-mediated disorder in a patient in need thereof, comprising the step of administering to said patient a compound according to any of claims 1 through 30 or a composition according to claim 31.
60. The method according to claim 59, wherein the disorder is selected from an autoimmune disorder, an inflammatory disorder, a neurodegenerative disorder, or a solid or hematologic malignancy.
CA2920996A 2007-10-19 2008-10-17 Heteroaryl compounds and uses thereof Abandoned CA2920996A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US98143207P 2007-10-19 2007-10-19
US60/981,432 2007-10-19
US5200208P 2008-05-09 2008-05-09
US61/052,002 2008-05-09
CA2702674A CA2702674C (en) 2007-10-19 2008-10-17 Heteroaryl compounds and uses thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CA2702674A Division CA2702674C (en) 2007-10-19 2008-10-17 Heteroaryl compounds and uses thereof

Publications (1)

Publication Number Publication Date
CA2920996A1 true CA2920996A1 (en) 2009-04-23

Family

ID=40567710

Family Applications (2)

Application Number Title Priority Date Filing Date
CA2920996A Abandoned CA2920996A1 (en) 2007-10-19 2008-10-17 Heteroaryl compounds and uses thereof
CA2702674A Expired - Fee Related CA2702674C (en) 2007-10-19 2008-10-17 Heteroaryl compounds and uses thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
CA2702674A Expired - Fee Related CA2702674C (en) 2007-10-19 2008-10-17 Heteroaryl compounds and uses thereof

Country Status (8)

Country Link
US (5) US7982036B2 (en)
EP (1) EP2214486A4 (en)
JP (2) JP5600063B2 (en)
CN (2) CN105367503A (en)
AU (3) AU2008314632B2 (en)
CA (2) CA2920996A1 (en)
TW (2) TWI552752B (en)
WO (1) WO2009051822A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113121575A (en) * 2014-08-25 2021-07-16 四川海思科制药有限公司 (substituted phenyl) (substituted pyrimidine) amino derivative and preparation method and pharmaceutical application thereof

Families Citing this family (96)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
HUE028086T2 (en) 2006-09-22 2016-11-28 Pharmacyclics Llc Inhibitors of bruton's tyrosine kinase
US20120101114A1 (en) 2007-03-28 2012-04-26 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase
US7989465B2 (en) * 2007-10-19 2011-08-02 Avila Therapeutics, Inc. 4,6-disubstituted pyrimidines useful as kinase inhibitors
TWI552752B (en) 2007-10-19 2016-10-11 賽基艾維洛米斯研究股份有限公司 Heteroaryl compounds and uses thereof
AU2009248923B2 (en) 2008-05-21 2015-01-29 Takeda Pharmaceutical Company Limited Phosphorous derivatives as kinase inhibitors
US9273077B2 (en) 2008-05-21 2016-03-01 Ariad Pharmaceuticals, Inc. Phosphorus derivatives as kinase inhibitors
US11351168B1 (en) 2008-06-27 2022-06-07 Celgene Car Llc 2,4-disubstituted pyrimidines useful as kinase inhibitors
US8338439B2 (en) 2008-06-27 2012-12-25 Celgene Avilomics Research, Inc. 2,4-disubstituted pyrimidines useful as kinase inhibitors
RU2536584C2 (en) 2008-06-27 2014-12-27 Авила Терапьютикс, Инк. Heteroaryl compounds and using them
WO2010009342A2 (en) 2008-07-16 2010-01-21 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase for the treatment of solid tumors
US20100185419A1 (en) * 2008-09-05 2010-07-22 Avila Therapeutics, Inc. Algorithm for designing irreversible inhibitors
WO2010129053A2 (en) 2009-05-05 2010-11-11 Dana Farber Cancer Institute Egfr inhibitors and methods of treating disorders
TW201103904A (en) 2009-06-11 2011-02-01 Hoffmann La Roche Janus kinase inhibitor compounds and methods
EP2440204B1 (en) 2009-06-12 2013-12-18 Bristol-Myers Squibb Company Nicotinamide compounds useful as kinase modulators
AR077695A1 (en) * 2009-08-04 2011-09-14 Schering Corp PIRIMIDINE DERIVATIVES AS INHIBITORS OF FACTOR IXA
US9556426B2 (en) * 2009-09-16 2017-01-31 Celgene Avilomics Research, Inc. Protein kinase conjugates and inhibitors
ES2467166T3 (en) * 2009-10-14 2014-06-12 Xenon Pharmaceuticals Inc. Methods of synthesis of spiro-oxindole compounds
WO2011082285A1 (en) * 2009-12-30 2011-07-07 Avila Therapeutics, Inc. Ligand-directed covalent modification of protein
US20120329784A1 (en) * 2010-01-13 2012-12-27 Kallander Lara S Compounds and methods
CN103003264B (en) 2010-05-21 2014-08-06 切米利亚股份公司 Novel pyrimidine derivatives
CA2800913C (en) 2010-06-03 2019-07-23 Pharmacyclics, Inc. The use of inhibitors of bruton's tyrosine kinase (btk)
SG187796A1 (en) 2010-08-10 2013-03-28 Celgene Avilomics Res Inc Besylate salt of a btk inhibitor
JP5956999B2 (en) 2010-11-01 2016-07-27 セルジーン アヴィロミクス リサーチ, インコーポレイテッド Heteroaryl compounds and uses thereof
TWI632134B (en) 2010-11-01 2018-08-11 阿維拉製藥公司 Heterocyclic compounds and uses thereof
WO2012064706A1 (en) 2010-11-10 2012-05-18 Avila Therapeutics, Inc. Mutant-selective egfr inhibitors and uses thereof
ES2587864T3 (en) 2011-03-24 2016-10-27 Noviga Research Ab Pyrimidine derivatives
KR101884010B1 (en) 2011-05-04 2018-07-31 어리어드 파마슈티칼스, 인코포레이티드 Compounds for inhibiting cell proliferation in egfr-driven cancers
RS63418B1 (en) * 2011-06-10 2022-08-31 Merck Patent Gmbh Compositions and methods for the production of pyrimidine and pyridine compounds with btk inhibitory activity
BR122014026094B1 (en) 2011-07-27 2020-10-13 Astrazeneca Ab compounds of formula ii, iii, iv and vi
TW201325593A (en) 2011-10-28 2013-07-01 Celgene Avilomics Res Inc Methods of treating a BRUTON'S tyrosine kinase disease or disorder
US9586965B2 (en) 2012-01-13 2017-03-07 Acea Biosciences Inc. Pyrrolo[2,3-d]pyrimidine compounds as inhibitors of protein kinases
US9034885B2 (en) 2012-01-13 2015-05-19 Acea Biosciences Inc. EGFR modulators and uses thereof
JP6353788B2 (en) 2012-01-13 2018-07-04 エイシア バイオサイエンシーズ インコーポレイテッド Heterocyclic compounds as anticancer agents and their use
US9464089B2 (en) 2012-01-13 2016-10-11 Acea Biosciences Inc. Heterocyclic compounds and uses thereof
SI2805940T1 (en) 2012-01-17 2017-04-26 Astellas Pharma Inc Pyrazine carboxamide compound
PT2825042T (en) 2012-03-15 2018-11-16 Celgene Car Llc Salts of an epidermal growth factor receptor kinase inhibitor
SG11201405691WA (en) 2012-03-15 2014-10-30 Celgene Avilomics Res Inc Solid forms of an epidermal growth factor receptor kinase inhibitor
EP2833886B1 (en) 2012-04-04 2020-08-12 HangzhouDeRenYuCheng Biotechnology Ltd. Substituted quinolines as bruton's tyrosine kinase inhibitors
WO2013161848A1 (en) * 2012-04-27 2013-10-31 カルナバイオサイエンス株式会社 Novel 1,2,4-triazine derivative
AU2013204563B2 (en) 2012-05-05 2016-05-19 Takeda Pharmaceutical Company Limited 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
CN110354132A (en) 2012-06-04 2019-10-22 药品循环有限责任公司 The crystalline form of bruton's tyrosine kinase inhibitor
EP2877598A1 (en) 2012-07-24 2015-06-03 Pharmacyclics, Inc. Mutations associated with resistance to inhibitors of bruton's tyrosine kinase (btk)
US9714450B2 (en) 2012-08-31 2017-07-25 New York University Methods for diagnosing and treating schizophrenia
WO2014040555A1 (en) * 2012-09-12 2014-03-20 山东亨利医药科技有限责任公司 Nitrogen-containing heteroaromatic ring derivative as tyrosine kinase inhibitor
BR112015011171A2 (en) 2012-11-15 2017-07-11 Pharmacyclics Inc pyrrolopyrimidine compounds as kinase inhibitors
WO2014100748A1 (en) 2012-12-21 2014-06-26 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
ES2805359T3 (en) 2013-02-08 2021-02-11 Celgene Car Llc ERK inhibitors and their uses
US9611283B1 (en) 2013-04-10 2017-04-04 Ariad Pharmaceuticals, Inc. Methods for inhibiting cell proliferation in ALK-driven cancers
NZ711540A (en) 2013-04-25 2018-08-31 Beigene Ltd Fused heterocyclic compounds as protein kinase inhibitors
EP2999702A4 (en) 2013-05-21 2017-01-11 Jiangsu Medolution Ltd. Substituted pyrazolopyrimidines as kinases inhibitors
BR112016000195A8 (en) 2013-07-11 2019-12-31 Acea Biosciences Inc heterocyclic compounds, pharmaceutical composition, combination, their use, and method for inhibiting a btk or jak tyrosine kinase activity, egfr, alk, pdgfr, blk, bmx / etk, flt3 (d835y), itk, tec, txk, and their respective ways
CA2919996A1 (en) 2013-08-02 2015-02-05 Pharmacyclics Llc Methods for the treatment of solid tumors
IN2013MU02611A (en) 2013-08-07 2015-06-12 Cadila Healthcare Ltd
ES2709509T3 (en) 2013-08-12 2019-04-16 Pharmacyclics Llc Procedures for the treatment of cancer amplified by HER2
WO2015025197A1 (en) 2013-08-22 2015-02-26 Jubilant Biosys Limited Substituted pyrimidine compounds, compositions and medicinal applications 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
EP4130044A1 (en) 2013-09-13 2023-02-08 BeiGene Switzerland GmbH Anti-pd1 antibodies and their use as therapeutics and diagnostics
CN105939998B (en) * 2013-10-21 2019-04-16 默克专利有限公司 Heteroaryl compound and application thereof as BTK inhibitor
CN111265531B (en) 2013-10-25 2023-11-10 药品循环有限责任公司 Methods for treating and preventing graft versus host disease
JP6879740B2 (en) * 2013-12-13 2021-06-02 ダナ−ファーバー キャンサー インスティテュート, インコーポレイテッド How to Treat Lymphatic Plasma Cell Lymphoma
US9415049B2 (en) 2013-12-20 2016-08-16 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
WO2015143400A1 (en) 2014-03-20 2015-09-24 Pharmacyclics, Inc. Phospholipase c gamma 2 and resistance associated mutations
TWI726608B (en) 2014-07-03 2021-05-01 英屬開曼群島商百濟神州有限公司 Anti-pd-l1 antibodies and their use as therapeutics and diagnostics
US9533991B2 (en) 2014-08-01 2017-01-03 Pharmacyclics Llc Inhibitors of Bruton's tyrosine kinase
SG11201700849XA (en) 2014-08-07 2017-03-30 Pharmacyclics Llc Novel formulations of a bruton's tyrosine kinase inhibitor
WO2016025561A1 (en) 2014-08-13 2016-02-18 Celgene Avilomics Research, Inc. Forms and compositions of an erk inhibitor
CN111170998B (en) 2014-11-05 2023-04-11 益方生物科技(上海)股份有限公司 Pyrimidine or pyridine compound, preparation method and medical application thereof
AU2016226279B2 (en) 2015-03-03 2021-11-11 Pharmacyclics Llc Pharmaceutical formulations of Bruton's tyrosine kinase inhibtor
CN104860890B (en) * 2015-04-29 2018-03-13 上海泓博智源医药股份有限公司 T790M mutant egfs R inhibitor and its application in antineoplastic is prepared
US20210323976A1 (en) * 2015-05-13 2021-10-21 Ariad Pharmaceuticals, Inc. Heteroaryl compounds for kinase inhibition
US20170209462A1 (en) 2015-08-31 2017-07-27 Pharmacyclics Llc Btk inhibitor combinations for treating multiple myeloma
CA3001744A1 (en) 2015-10-09 2017-04-13 ACEA Therapeutics, Inc. Pharmaceutical salts, physical forms, and compositions of pyrrolopyrimidine kinase inhibitors, and methods of making same
MY196809A (en) 2015-12-24 2023-05-03 Kyowa Kirin Co Ltd (),()-unsaturated amide compound
CN106928150B (en) * 2015-12-31 2020-07-31 恩瑞生物医药科技(上海)有限公司 Acrylamide aniline derivative and pharmaceutical application thereof
AU2017293423B2 (en) 2016-07-05 2023-05-25 Beigene, Ltd. Combination of a PD-1 antagonist and a RAF inhibitor for treating cancer
CN109563099B (en) 2016-08-16 2023-02-03 百济神州有限公司 Crystal form of compound, preparation and application thereof
WO2018033135A1 (en) 2016-08-19 2018-02-22 Beigene, Ltd. Use of a combination comprising a btk inhibitor for treating cancers
EP3515414B1 (en) 2016-09-19 2022-11-30 MEI Pharma, Inc. Combination therapy
MA46783A (en) 2016-11-03 2019-09-11 Juno Therapeutics Inc T-CELL THERAPY AND BTK INHIBITOR POLYTHERAPY
CN106749042B (en) * 2016-11-16 2019-01-22 大连医科大学 Sulfoamido pyrimidines, composition and purposes
CN110545826A (en) 2016-12-03 2019-12-06 朱诺治疗学股份有限公司 Methods and compositions for using therapeutic T cells in combination with kinase inhibitors
US11555038B2 (en) 2017-01-25 2023-01-17 Beigene, Ltd. Crystalline forms of (S)-7-(1-(but-2-ynoyl)piperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
US11498922B2 (en) 2017-04-07 2022-11-15 ACEA Therapeutics, Inc. Pharmaceutical composition comprising N-(3-((2-((3-fluoro-4-(4-methylpiperazin-1-yl phenyl)amino)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)oxy)phenylacrylamide
EP3712129A4 (en) * 2017-06-23 2020-12-09 Kyowa Kirin Co., Ltd. Alpha, beta-unsaturated amide compound
AU2018290532A1 (en) 2017-06-26 2019-11-21 Beigene, Ltd. Immunotherapy for hepatocellular carcinoma
CN110997677A (en) 2017-08-12 2020-04-10 百济神州有限公司 Btk inhibitors with improved dual selectivity
JP2019062834A (en) * 2017-10-03 2019-04-25 アークレイ株式会社 Probes, kits and methods for detecting c797s(g2390c) mutations in human egfr
WO2019108795A1 (en) 2017-11-29 2019-06-06 Beigene Switzerland Gmbh Treatment of indolent or aggressive b-cell lymphomas using a combination comprising btk inhibitors
SG11202010642TA (en) 2018-05-03 2020-11-27 Juno Therapeutics Inc Combination therapy of a chimeric antigen receptor (car) t cell therapy and a kinase inhibitor
CN108715834B (en) * 2018-06-01 2021-09-14 天晴干细胞股份有限公司 Preparation method of platelet lysate rich in CD41+ and CD81+ microcapsules
KR20240007289A (en) * 2021-08-02 2024-01-16 아비스코 테라퓨틱스 컴퍼니 리미티드 Pyrimidine-4,6-diamine derivatives and their preparation methods and pharmaceutical applications
CN114621922A (en) * 2022-02-15 2022-06-14 中山大学附属第五医院 Extraction, purification and culture method of rat cavernous nerve derived Schwann cells
WO2023220655A1 (en) 2022-05-11 2023-11-16 Celgene Corporation Methods to overcome drug resistance by re-sensitizing cancer cells to treatment with a prior therapy via treatment with a t cell therapy
US11786531B1 (en) 2022-06-08 2023-10-17 Beigene Switzerland Gmbh Methods of treating B-cell proliferative disorder
WO2024056077A1 (en) * 2022-09-15 2024-03-21 Cullgen (Shanghai) , Inc. Modified proteins and protein binders and degraders

Family Cites Families (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH0741461A (en) 1993-05-27 1995-02-10 Eisai Co Ltd Sulfonic acid ester derivative
GB9325217D0 (en) * 1993-12-09 1994-02-09 Zeneca Ltd Pyrimidine derivatives
US5977117A (en) 1996-01-05 1999-11-02 Texas Biotechnology Corporation Substituted phenyl compounds and derivatives thereof that modulate the activity of endothelin
US5994375A (en) 1996-02-12 1999-11-30 Berlex Laboratories, Inc. Benzamidine derivatives substituted by amino acid and hydroxy acid derivatives and their use as anti-coagulants
DK0945443T3 (en) 1998-03-27 2003-06-02 Janssen Pharmaceutica Nv HIV inhibitory pyrimidine derivatives
DE69943247D1 (en) 1998-03-27 2011-04-14 Janssen Pharmaceutica Nv HIV-inhibiting pyrimidine derivatives
US6303652B1 (en) * 1998-08-21 2001-10-16 Hughes Institute BTK inhibitors and methods for their identification and use
EP1107958B1 (en) * 1998-08-29 2006-08-16 AstraZeneca AB Pyrimidine compounds
US6262088B1 (en) 1998-11-19 2001-07-17 Berlex Laboratories, Inc. Polyhydroxylated monocyclic N-heterocyclic derivatives as anti-coagulants
US6495558B1 (en) 1999-01-22 2002-12-17 Amgen Inc. Kinase inhibitors
GB9905075D0 (en) * 1999-03-06 1999-04-28 Zeneca Ltd Chemical compounds
GB9907658D0 (en) 1999-04-06 1999-05-26 Zeneca Ltd Chemical compounds
AU5020400A (en) 1999-05-20 2000-12-12 E.I. Du Pont De Nemours And Company Heteroaryloxypyrimidine insecticides and acaricides
KR100820605B1 (en) 1999-09-24 2008-04-08 얀센 파마슈티카 엔.브이. Antiviral compositions
GB9924092D0 (en) * 1999-10-13 1999-12-15 Zeneca Ltd Pyrimidine derivatives
EA005028B1 (en) 2000-01-31 2004-10-28 Пфайзер Продактс Инк. Pyrimidine carboxamides useful as inhibitors of pde4 isozymes
US7427689B2 (en) 2000-07-28 2008-09-23 Georgetown University ErbB-2 selective small molecule kinase inhibitors
US20020137755A1 (en) * 2000-12-04 2002-09-26 Bilodeau Mark T. Tyrosine kinase inhibitors
US6881737B2 (en) 2001-04-11 2005-04-19 Amgen Inc. Substituted triazinyl acrylamide derivatives and methods of use
MXPA04004178A (en) 2001-11-01 2004-09-06 Janssen Pharmaceutica Nv Heteroaryl amines as glycogen synthase kinase 3beta inhibitors (gsk3 inhibitors).
CN100436427C (en) 2001-11-01 2008-11-26 詹森药业有限公司 Aminobenzamide derivatives as glycogen synthase kinase 3 beta inhibitors
IL162210A0 (en) * 2001-11-28 2005-11-20 Btg Int Ltd Preventives or remedies for alzheimer's disease oramyloid protein fibrosis inhibitors containing ni trogen-containing heteroaryl
AR039540A1 (en) 2002-05-13 2005-02-23 Tibotec Pharm Ltd MICROBICIDE COMPOUNDS WITH PIRIMIDINE OR TRIAZINE CONTENT
US6841664B2 (en) 2002-07-25 2005-01-11 Enanra Pharmaceuticals, Inc. 6,11-4-carbon bridged ketolides
AR037647A1 (en) * 2002-05-29 2004-12-01 Novartis Ag USED DIARILUREA DERIVATIVES FOR THE TREATMENT OF DEPENDENT DISEASES OF THE PROTEIN KINase
DE10226943A1 (en) * 2002-06-17 2004-01-08 Bayer Ag Phenylaminopyrimidines and their use
US20040002395A1 (en) * 2002-06-27 2004-01-01 Poynor Raymond L. Bridge weight for metal wood golf club
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
US20050014753A1 (en) 2003-04-04 2005-01-20 Irm Llc Novel compounds and compositions as protein kinase inhibitors
EP1620731A2 (en) 2003-04-07 2006-02-01 Praecis Pharmaceuticals Inc. Methods of measuring the ability of a test compound to inactivate a biological target in cells of a subject
WO2005007646A1 (en) 2003-07-10 2005-01-27 Neurogen Corporation Substituted heterocyclic diarylamine analogues
WO2005040135A1 (en) 2003-10-24 2005-05-06 Ono Pharmaceutical Co., Ltd. Antistress drug and medical use thereof
US20080312192A1 (en) * 2003-11-28 2008-12-18 Guido Bold Diaryl Urea Derivatives in the Treatment of Protein Kinase Dependent Diseases
GB2432834A (en) 2004-01-12 2007-06-06 Cytopia Res Pty Ltd Selective Kinase Inhibitors
US7558717B2 (en) * 2004-04-28 2009-07-07 Vertex Pharmaceuticals Incorporated Crystal structure of human JAK3 kinase domain complex and binding pockets thereof
US7879853B2 (en) * 2004-06-28 2011-02-01 Bayer Schering Pharma Ag 4,6-disubstituted pyrimidines and their use as protein kinase inhibitors
WO2006045066A2 (en) 2004-10-20 2006-04-27 Proteolix, Inc. Labeled compounds for proteasome inhibition
MY169441A (en) 2004-12-08 2019-04-11 Janssen Pharmaceutica Nv 2,4, (4,6) pyrimidine derivatives
DE102005016634A1 (en) 2005-04-12 2006-10-19 Merck Patent Gmbh Novel aza heterocycles as kinase inhibitors
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
DE602006017574D1 (en) * 2005-07-26 2010-11-25 Vertex Pharma USE AS PROTEIN KINASE INHIBITORS BENZIMIDAZOLE
CA2626479A1 (en) * 2005-11-03 2007-05-18 Irm Llc Protein kinase inhibitors
WO2007081630A2 (en) 2005-12-21 2007-07-19 Janssen Pharmaceutica, N.V. Substituted pyrimidinyl kinase inhibitors
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
US7560588B2 (en) 2006-04-27 2009-07-14 Intezyne Technologies, Inc. Poly(ethylene glycol) containing chemically disparate endgroups
HUE028086T2 (en) 2006-09-22 2016-11-28 Pharmacyclics Llc Inhibitors of bruton's tyrosine kinase
US8987233B2 (en) 2006-11-03 2015-03-24 Pharmacyclics, Inc. Bruton's tyrosine kinase activity probe and method of using
EP2114900B1 (en) 2007-01-31 2018-10-10 YM BioSciences Australia Pty Ltd Thiopyrimidine-based compounds and uses thereof
EP2014657A1 (en) * 2007-06-21 2009-01-14 Bayer Schering Pharma Aktiengesellschaft Diaminopyrimidines as modulators for an EP2 receptor
TWI552752B (en) 2007-10-19 2016-10-11 賽基艾維洛米斯研究股份有限公司 Heteroaryl compounds and uses thereof
US7989465B2 (en) 2007-10-19 2011-08-02 Avila Therapeutics, Inc. 4,6-disubstituted pyrimidines useful as kinase inhibitors
US8338439B2 (en) 2008-06-27 2012-12-25 Celgene Avilomics Research, Inc. 2,4-disubstituted pyrimidines useful as kinase inhibitors
RU2536584C2 (en) 2008-06-27 2014-12-27 Авила Терапьютикс, Инк. Heteroaryl compounds and using them

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113121575A (en) * 2014-08-25 2021-07-16 四川海思科制药有限公司 (substituted phenyl) (substituted pyrimidine) amino derivative and preparation method and pharmaceutical application thereof

Also Published As

Publication number Publication date
US20150328223A1 (en) 2015-11-19
TWI475996B (en) 2015-03-11
AU2015202675B2 (en) 2016-12-01
US7982036B2 (en) 2011-07-19
US20090137588A1 (en) 2009-05-28
CN105367503A (en) 2016-03-02
JP2014156489A (en) 2014-08-28
TWI552752B (en) 2016-10-11
CA2702674C (en) 2016-05-03
EP2214486A1 (en) 2010-08-11
JP5600063B2 (en) 2014-10-01
JP2011500685A (en) 2011-01-06
EP2214486A4 (en) 2011-03-09
US9393246B2 (en) 2016-07-19
CA2702674A1 (en) 2009-04-23
AU2008314632B2 (en) 2015-05-28
TW200924776A (en) 2009-06-16
CN101902911A (en) 2010-12-01
CN101902911B (en) 2015-12-16
US8445498B2 (en) 2013-05-21
US20110230494A1 (en) 2011-09-22
AU2016256754A1 (en) 2016-12-01
WO2009051822A1 (en) 2009-04-23
US9040541B2 (en) 2015-05-26
US20160311780A1 (en) 2016-10-27
TW201529072A (en) 2015-08-01
US20130217688A1 (en) 2013-08-22
AU2008314632A1 (en) 2009-04-23
AU2015202675A1 (en) 2015-06-11

Similar Documents

Publication Publication Date Title
CA2702674C (en) Heteroaryl compounds and uses thereof
CA2783624C (en) Heteroaryl compounds and uses thereof
CA2727455C (en) Heteroaryl compounds and uses thereof
US9296704B2 (en) Substituted pyrimidines as protein kinase inhibitors
US9549927B2 (en) Heteroaryl compounds and uses thereof

Legal Events

Date Code Title Description
EEER Examination request

Effective date: 20160216

FZDE Dead

Effective date: 20180427