US20130035325A1 - Kinase inhibitors - Google Patents

Kinase inhibitors Download PDF

Info

Publication number
US20130035325A1
US20130035325A1 US13/510,272 US201013510272A US2013035325A1 US 20130035325 A1 US20130035325 A1 US 20130035325A1 US 201013510272 A US201013510272 A US 201013510272A US 2013035325 A1 US2013035325 A1 US 2013035325A1
Authority
US
United States
Prior art keywords
substituted
unsubstituted
heteroaryl
heterocycloalkyl
alkyl
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
US13/510,272
Other languages
English (en)
Inventor
John William Taunton, Jr.
Rebecca Maglathlin
Iana Serafimova
Michael S. Cohen
Rand Miller
Ville Paavilainen
Jesse McFarland
Shyam KRISHNAN
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.)
University of California
Original Assignee
University of California
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 University of California filed Critical University of California
Priority to US13/510,272 priority Critical patent/US20130035325A1/en
Publication of US20130035325A1 publication Critical patent/US20130035325A1/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: UNIVERSITY OF CALIFORNIA, SAN FRANCISCO
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • 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
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/02Muscle relaxants, e.g. for tetanus or cramps
    • 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
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • 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
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/30Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members
    • C07D207/34Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D207/36Oxygen or sulfur atoms
    • C07D207/402,5-Pyrrolidine-diones
    • C07D207/4162,5-Pyrrolidine-diones with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to other ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/54Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings condensed with carbocyclic rings or ring systems
    • C07D231/56Benzopyrazoles; Hydrogenated benzopyrazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/26Heterocyclic compounds containing purine ring systems with an oxygen, sulphur, or nitrogen atom directly attached in position 2 or 6, but not in both
    • C07D473/32Nitrogen atom
    • C07D473/34Nitrogen atom attached in position 6, e.g. adenine

Definitions

  • the human genome contains at least 500 genes encoding protein kinases.
  • protein kinase genes constitute about 2% of all human genes. Protein kinases modify up to 30% of all human proteins and regulate the majority of cellular pathways, particularly those pathways involved in signal transduction.
  • kinase inhibitors are provided.
  • the kinase inhibitor has the structure of Formula I:
  • R 1 is substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.
  • L 1 is bond, —C(O)—, —C(O)N(L 3 R 2 )—, —C(O)O—, —S(O) n —, —O—, —N(L 3 R 2 )—, —P(O)(OL 3 R 2 )O—, —SO 2 N(L 3 R 2 )—, —P(O)(NL 3 R 2 )N—, substituted or unsubstituted alkylene, substituted or unsubstituted heteroalkylene, substituted or unsubstituted cycloalkylene, substituted or unsubstituted heterocycloalkylene, substituted or unsubstituted arylene, or substituted or unsubstituted heteroarylene.
  • n is 0, 1 or 2.
  • L 2 is a bond, —C(O)—, —C(O)N(L 3A R 2A ) t —, —C(O)O—, —S(O) t —, —O—, —N(L 3A R 2A ) t —, —P(O)(OL 3A R 2A )O—, —SO 2 N(L 3A R 2A )—, —P(O)(NL 3A R 2A )N—, substituted or unsubstituted alkylene, substituted or unsubstituted heteroalkylene, substituted or unsubstituted cycloalkylene, substituted or unsubstituted heterocycloalkylene, substituted or unsubstituted arylene, or substituted or unsubstituted heteroarylene.
  • the symbol t is 0, 1 or 2.
  • L 3 and L 3A are independently a bond, substituted or unsubstituted alkylene, substituted or unsubstituted heteroalkylene, substituted or unsubstituted cycloalkylene, substituted or unsubstituted heterocycloalkylene, substituted or unsubstituted arylene, or substituted or unsubstituted heteroarylene.
  • the symbol w is 0, 1 or 2.
  • R 2 and R 2A are independently hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.
  • E is an electron withdrawing group or together with L 2 forms an electron withdrawing group.
  • methods of inhibiting protein kinases include contacting a protein kinase with an effective amount of a kinase inhibitor provided herein.
  • the kinase inhibitor may have the structure of Formula I or Formula II (or any of the embodiments thereof described herein).
  • a method of treating a disease associated with kinase activity in a subject in need of such treatment includes administering to the subject an effective amount of a kinase inhibitor provided herein.
  • the kinase inhibitor may have the structure of Formula I or Formula II (or any of the embodiments thereof described herein).
  • FIG. 1A and FIG. 1B provide mass spectrometric results following incubation of DMSO or Cmpds 1-4 ( FIG. 1A ), and Cmpds 5-8 ( FIG. 1B ) with human RSK2 CTD for 1 hr at room temperature.
  • FIG. 2 depicts recovery of kinase activity of RSK2 CTD after inhibition by a selection of compounds disclosed herein and subsequent dialysis. Legend: Cmpd 6: checked; Cmpd 7: open box; Cmpd 1: Black box; Cmpd 9: diagonal stripes.
  • FIG. 3 depicts cyanoacrylate or cyanoacrylamide (Cmpds 4-7) dissociation from intact folded RSK2 CTD, as measured by competitive labeling with fluoromethylketone Cmpd 9 (FMK).
  • Upper left panel time course of FMK labeling of empty RSK2 CTD.
  • Upper right panel time course of dissociation of reversible covalent inhibits, Cmpds 4-7.
  • Lower panel tabular presentation of dissociation half-time (min) for the indicated compounds.
  • FIG. 4 depicts the formation and reversal of covalent bond formation between RSK2 and Cmpd 7 by UV/Visible spectrophotometry.
  • Upper panel normalized absorbance (400 nm) attributed to Cmpd 7 after reaction with a) C436V RSK2; b) WT RSK2; c) WT RSK2 plus proteinase K; d) WT RSK2 plus SDS; and e) WT RSK2 plus guanidine HCl.
  • Middle panel UV/Visible spectra for Cmpd 7, alone in buffer or in the presence of RSK2 or RSK2 plus Proteinase K (3 hr incubation).
  • Lower panel UV/Visible spectra for Cmpd 7, alone in buffer or in the presence of RSK2 or RSK2 plus SDS (1 min incubation).
  • FIG. 5A depicts the mass spectrometric analysis of the incubation of Cmpd 7 with 3 M guanidine HCl.
  • FIG. 5B depicts the mass spectrometric analysis of the incubation of Cmpd 7 incubated with RSK2 CTD prior to addition of 3 M guanidine HCl.
  • FIG. 6 depicts the inhibition of autophosphorylation of Ser386 of RSK2 by Cmpds 5-7 and Cmpd 9 in HEK-293 cells.
  • FMK 9 Cmpd 9
  • Cmpd 7 black box
  • Cmpd 6 grayed box
  • Cmpd 5 diagonal stripes.
  • Lower panel Western blot analysis with phospho-Ser386 RSK2 and anti-HA antibodies, as described herein.
  • FIG. 7 depicts modes of binding of Cmpds 6, 12 or 15 (top, middle, and lower panels, respectively) to Cys-436 of RSK2, based on X-ray crystallographic structures obtained as described herein.
  • FIG. 8 depicts modes of binding of Cmpd 40 (top panel) to Cys-436 of RSK2, and Cmpd 55 to Cys-345 of cSrc.
  • FIG. 9 depicts 1 H NMR spectrum before and after dilution of reaction mixture.
  • FIG. 10 depicts cyanoacrylamide absorbance spectrum and graphical representation of the absorbance values before and after dilution.
  • substituent groups are specified by their conventional chemical formulae, written from left to right, they equally encompass the chemically identical substituents that would result from writing the structure from right to left, e.g., —CH 2 O— is equivalent to —OCH 2 —.
  • alkyl by itself or as part of another substituent, means, unless otherwise stated, a straight (i.e. unbranched) or branched chain, or combination thereof, which may be fully saturated, mono- or polyunsaturated and can include di- and multivalent radicals, having the number of carbon atoms designated (i.e. C 1 -C 10 means one to ten carbons).
  • saturated hydrocarbon radicals include, but are not limited to, groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, (cyclohexyl)methyl, homologs and isomers of, for example, n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like.
  • An unsaturated alkyl group is one having one or more double bonds or triple bonds.
  • unsaturated alkyl groups include, but are not limited to, vinyl, 2-propenyl, crotyl, 2-isopentenyl, 2-(butadienyl), 2,4-pentadienyl, 3-(1,4-pentadienyl), ethynyl, 1- and 3-propynyl, 3-butynyl, and the higher homologs and isomers.
  • An alkoxy is an alkyl attached to the remainder of the molecule via an oxygen linker (—O—).
  • alkylene by itself or as part of another substituent means a divalent radical derived from an alkyl, as exemplified, but not limited, by —CH 2 CH 2 CH 2 CH 2 —, and further includes those groups described below as “heteroalkylene.”
  • an alkyl (or alkylene) group will have from 1 to 24 carbon atoms, with those groups having 10 or fewer carbon atoms being preferred in the present invention.
  • a “lower alkyl” or “lower alkylene” is a shorter chain alkyl or alkylene group, generally having eight or fewer carbon atoms.
  • heteroalkyl by itself or in combination with another term, means, unless otherwise stated, a stable straight or branched chain, or cyclic hydrocarbon radical, or combinations thereof, consisting of at least one carbon atoms and at least one heteroatom selected from the group consisting of O, N, P, Si and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized.
  • the heteroatom(s) O, N, P and S and Si may be placed at any interior position of the heteroalkyl group or at the position at which the alkyl group is attached to the remainder of the molecule.
  • Examples include, but are not limited to, —CH 2 —CH 2 —O—CH 3 , —CH 2 —CH 2 —NH—CH 3 , —CH 2 —CH 2 —N(CH 3 )—CH 3 , —CH 2 —S—CH 2 —CH 3 , —CH 2 —CH 2 , —S(O)—C 1-3 , —CH 2 —CH 2 —S(O) 2 —CH 3 , —CH ⁇ CH—O—CH 3 , —Si(CH 3 ) 3 , —CH 2 —CH ⁇ N—OCH 3 , —CH ⁇ CH—N(CH 3 )—CH 3 , O—CH 3 , —O—CH 2 —CH 3 , and —CN.
  • heteroalkylene by itself or as part of another substituent means a divalent radical derived from heteroalkyl, as exemplified, but not limited by, —CH 2 —CH 2 —S—CH 2 —CH 2 — and —CH 2 —S—CH 2 —CH 2 —NH—CH 2 —.
  • heteroatoms can also occupy either or both of the chain termini (e.g., alkyleneoxy, alkylenedioxy, alkyleneamino, alkylenediamino, and the like).
  • heteroalkyl groups include those groups that are attached to the remainder of the molecule through a heteroatom, such as —C(O)R′, —C(O)NR′, —NR′R′′, —OR′, —SR′, and/or —SO 2 R′.
  • heteroalkyl is recited, followed by recitations of specific heteroalkyl groups, such as —NR′R′′ or the like, it will be understood that the terms heteroalkyl and —NR′R′′ are not redundant or mutually exclusive. Rather, the specific heteroalkyl groups are recited to add clarity. Thus, the term “heteroalkyl” should not be interpreted herein as excluding specific heteroalkyl groups, such as —NR′R′′ or the like.
  • cycloalkyl and “heterocycloalkyl,” by themselves or in combination with other terms, represent, unless otherwise stated, cyclic versions of “alkyl” and “heteroalkyl”, respectively. Additionally, for heterocycloalkyl, a heteroatom can occupy the position at which the heterocycle is attached to the remainder of the molecule. Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, 1-cyclohexenyl, 3-cyclohexenyl, cycloheptyl, and the like.
  • heterocycloalkyl examples include, but are not limited to, 1-(1,2,5,6-tetrahydropyridyl), 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-morpholinyl, 3-morpholinyl, tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, tetrahydrothien-2-yl, tetrahydrothien-3-yl, 1-piperazinyl, 2-piperazinyl, and the like.
  • a “cycloalkylene” and a “heterocycloalkylene,” alone or as part of another substituent means a divalent radical derived from a cycloalkyl and heterocycloalkyl, respectively.
  • halo or “halogen,” by themselves or as part of another substituent, mean, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom. Additionally, terms such as “haloalkyl,” are meant to include monohaloalkyl and polyhaloalkyl.
  • halo(C 1 -C 4 )alkyl is meant to include, but not be limited to, fluoromethyl, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, 4-chlorobutyl, 3-bromopropyl, and the like.
  • acyl means —C(O)R where R is a substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl.
  • aryl means, unless otherwise stated, a polyunsaturated, aromatic, hydrocarbon substituent which can be a single ring or multiple rings (preferably from 1 to 3 rings) which are fused together (i.e. a fused ring aryl) or linked covalently.
  • a fused ring aryl refers to multiple rings fused together wherein at least one of the fused rings is an aryl ring.
  • heteroaryl refers to aryl groups (or rings) that contain from one to four heteroatoms selected from N, O, and S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quaternized.
  • heteroaryl includes fused ring heteroaryl groups (i.e. multiple rings fused together wherein at least one of the fused rings is a heteroaromatic ring).
  • a 5,6-fused ring heteroarylene refers to two rings fused together, wherein one ring has 5 members and the other ring has 6 members, and wherein at least one ring is a heteroaryl ring.
  • a 6,6-fused ring heteroarylene refers to two rings fused together, wherein one ring has 6 members and the other ring has 6 members, and wherein at least one ring is a heteroaryl ring.
  • a 6,5-fused ring heteroarylene refers to two rings fused together, wherein one ring has 6 members and the other ring has 5 members, and wherein at least one ring is a heteroaryl ring.
  • a heteroaryl group can be attached to the remainder of the molecule through a carbon or heteroatom.
  • Non-limiting examples of aryl and heteroaryl groups include phenyl, 1-naphthyl, 2-naphthyl, 4-biphenyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 3-pyrazolyl, 2-imidazolyl, 4-imidazolyl, pyrazinyl, 2-oxazolyl, 4-oxazolyl, 2-phenyl-4-oxazolyl, 5-oxazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2-furyl, 3-furyl, 2-thienyl, 3-thienyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidyl, 4-pyrimidyl, 5-benzothiazolyl, purinyl, 2-benzimidazolyl, 5-indolyl, 1-isoquinoly
  • arylene and heteroarylene alone or as part of another substituent means a divalent radical derived from an aryl and heteroaryl, respectively.
  • aryl when used in combination with other terms (e.g., aryloxy, arylthioxy, arylalkyl) includes both aryl and heteroaryl rings as defined above.
  • arylalkyl is meant to include those radicals in which an aryl group is attached to an alkyl group (e.g., benzyl, phenethyl, pyridylmethyl and the like) including those alkyl groups in which a carbon atom (e.g., a methylene group) has been replaced by, for example, an oxygen atom (e.g., phenoxymethyl, 2-pyridyloxymethyl, 3-(1-naphthyloxy)propyl, and the like).
  • alkyl group e.g., benzyl, phenethyl, pyridylmethyl and the like
  • an oxygen atom e.g., phenoxymethyl, 2-pyridyloxymethyl, 3-(1-naph
  • oxo as used herein means an oxygen that is double bonded to a carbon atom.
  • alkylsulfonyl as used herein means a moiety having the formula —S(O 2 )—R′, where R′ is an alkyl group as defined above. R′ may have a specified number of carbons (e.g. “C 1 -C 4 alkylsulfonyl”).
  • alkyl e.g., “alkyl,” “heteroalkyl,” “aryl” and “heteroaryl” are meant to include both substituted and unsubstituted forms of the indicated radical.
  • Preferred substituents for each type of radical are provided below.
  • Substituents for the alkyl and heteroalkyl radicals can be one or more of a variety of groups selected from, but not limited to: —OR′, ⁇ O, ⁇ NR′, ⁇ N—OR′, —NR′R′′, —SR′, -halogen, —SiR′R′′R′′′, —OC(O)R′, —C(O)R′, —CO 2 R′, —CONR′R′′, —OC(O)NR′R′′, —NR′′C(O)R′, —NR′—C(O)NR′′R′′′, —NR′′C(O) 2 R′, —NR—C(NR′R′′R′′′) ⁇ NR′′, —NR—C(NR′R′′R′′′) ⁇ NR′′, —OR′, ⁇ O, ⁇ NR′, ⁇ N—OR′, —NR′R′′, —SR′, -halogen, —SiR′R′′R′′′
  • R′, R′′, R′′′ and R′′′′ each preferably independently refer to hydrogen, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl (e.g., aryl substituted with 1-3 halogens), substituted or unsubstituted alkyl, alkoxy or thioalkoxy groups, or arylalkyl groups.
  • each of the R groups is independently selected as are each R′, R′′, R′′′ and R′′′′ groups when more than one of these groups is present.
  • R′ and R′′ are attached to the same nitrogen atom, they can be combined with the nitrogen atom to form a 4-, 5-, 6-, or 7-membered ring.
  • —NR′R′′ is meant to include, but not be limited to, 1-pyrrolidinyl and 4-morpholinyl.
  • alkyl is meant to include groups including carbon atoms bound to groups other than hydrogen groups, such as haloalkyl (e.g., —CF 3 and —CH 2 CF 3 ) and acyl (e.g., —C(O)CH 3 , —C(O)CF 3 , —C(O)CH 2 OCH 3 , and the like).
  • haloalkyl e.g., —CF 3 and —CH 2 CF 3
  • acyl e.g., —C(O)CH 3 , —C(O)CF 3 , —C(O)CH 2 OCH 3 , and the like.
  • substituents for the aryl and heteroaryl groups are varied and are selected from, for example: halogen, —OR′, —NR′R′′, —SR′, -halogen, —SiR′R′′R′′′, —OC(O)R′, —C(O)R′, —CO 2 R′, —CONR′R′′, —OC(O)NR′R′′, —NR′′C(O)R′, —NR′—C(O)NR′′R′′′, —NR′′C(O) 2 R′, —NR—C(NR′R′′R′′′) ⁇ NR′′′′, —NR—C(NR′R′′) ⁇ NR′′′, —S(O)R′, —S(O) 2 R′, —S(O) 2 NR′R′′, —NRSO 2 R′, —CN and —NO 2 , —R′, —N 3 , —
  • Two of the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally form a ring of the formula -T—C(O)—(CRR′) q —U—, wherein T and U are independently —NR—, —O—, —CRR′— or a single bond, and q is an integer of from 0 to 3.
  • two of the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with a substituent of the formula -A-(CH 2 ) r —B—, wherein A and B are independently —CRR′—, —O—, —NR—, —S—, —S(O)—, —S(O) 2 —, —S(O) 2 NR′— or a single bond, and r is an integer of from 1 to 4.
  • One of the single bonds of the new ring so formed may optionally be replaced with a double bond.
  • two of the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with a substituent of the formula —(CRR′) n —X′—(C′′R′′′) d —, where s and d are independently integers of from 0 to 3, and X′ is —O—, —NR′—, —S—, —S(O)—, —S(O) 2 —, or —S(O) 2 NR′—.
  • the substituents R, R′, R′′ and R′′′ are preferably independently selected from hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl.
  • heteroatom or “ring heteroatom” is meant to include oxygen (O), nitrogen (N), sulfur (S), phosphorus (P), and silicon (Si).
  • a “substituent group,” as used herein, means a group selected from the following moieties:
  • a “size-limited substituent” or “size-limited substituent group,” as used herein means a group selected from all of the substituents described above for a “substituent group,” wherein each substituted or unsubstituted alkyl is a substituted or unsubstituted C 1 -C 20 alkyl, each substituted or unsubstituted heteroalkyl is a substituted or unsubstituted 2 to 20 membered heteroalkyl, each substituted or unsubstituted cycloalkyl is a substituted or unsubstituted C 4 -C 8 cycloalkyl, and each substituted or unsubstituted heterocycloalkyl is a substituted or unsubstituted 4 to 8 membered heterocycloalkyl.
  • a “lower substituent” or “lower substituent group,” as used herein means a group selected from all of the substituents described above for a “substituent group,” wherein each substituted or unsubstituted alkyl is a substituted or unsubstituted C 1 -C 8 alkyl, each substituted or unsubstituted heteroalkyl is a substituted or unsubstituted 2 to 8 membered heteroalkyl, each substituted or unsubstituted cycloalkyl is a substituted or unsubstituted C 5 -C 7 cycloalkyl, and each substituted or unsubstituted heterocycloalkyl is a substituted or unsubstituted 5 to 7 membered heterocycloalkyl.
  • salts are meant to include salts of the active compounds which are prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein.
  • base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent.
  • pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amino, or magnesium salt, or a similar salt.
  • acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent.
  • Examples of pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, propionic, isobutyric, maleic, malonic, benzoic, succinic, suberic, fumaric, lactic, mandelic, phthalic, benzenesulfonic, p-tolylsulfonic, citric, tartaric, oxalic, methanesulfonic, and the like.
  • inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic,
  • salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galactunoric acids and the like (see, for example, Berge et al., “Pharmaceutical Salts”, Journal of Pharmaceutical Science, 1977, 66, 1-19).
  • Certain specific compounds of the present invention contain both basic and acidic functionalities that allow the compounds to be converted into either base or acid addition salts.
  • the compounds of the present invention may exist as salts, such as with pharmaceutically acceptable acids.
  • the present invention includes such salts.
  • examples of such salts include hydrochlorides, hydrobromides, sulfates, methanesulfonates, nitrates, maleates, acetates, citrates, fumarates, tartrates (e.g., (+)-tartrates, ( ⁇ )-tartrates or mixtures thereof including racemic mixtures), succinates, benzoates and salts with amino acids such as glutamic acid.
  • These salts may be prepared by methods known to those skilled in the art.
  • the neutral forms of the compounds are preferably regenerated by contacting the salt with a base or acid and isolating the parent compound in the conventional manner.
  • the parent form of the compound differs from the various salt forms in certain physical properties, such as solubility in polar solvents.
  • the present invention provides compounds, which are in a prodrug form.
  • Prodrugs of the compounds described herein are those compounds that readily undergo chemical changes under physiological conditions to provide the compounds of the present invention.
  • prodrugs can be converted to the compounds of the present invention by chemical or biochemical methods in an ex vivo environment. For example, prodrugs can be slowly converted to the compounds of the present invention when placed in a transdermal patch reservoir with a suitable enzyme or chemical reagent.
  • Certain compounds of the present invention can exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, the solvated forms are equivalent to unsolvated forms and are encompassed within the scope of the present invention. Certain compounds of the present invention may exist in multiple crystalline or amorphous forms. In general, all physical forms are equivalent for the uses contemplated by the present invention and are intended to be within the scope of the present invention.
  • Certain compounds of the present invention possess asymmetric carbon atoms (optical centers) or double bonds; the racemates, diastereomers, tautomers, geometric isomers and individual isomers are encompassed within the scope of the present invention.
  • the compounds of the present invention do not include those which are known in the art to be too unstable to synthesize and/or isolate.
  • the compounds of the present invention may also contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds.
  • the compounds may be radiolabeled with radioactive isotopes, such as for example tritium ( 3 H), iodine-125 ( 125 I) or carbon-14 ( 14 C). All isotopic variations of the compounds of the present invention, whether radioactive or not, are encompassed within the scope of the present invention.
  • R-substituted e.g. R 7 -substituted
  • R 7 substituent of a compound provided herein
  • the substituent is substituted with one or more of the named R groups (e.g. R 7 ) as appropriate.
  • the substituent is substituted with only one of the named R groups.
  • treating refers to any indicia of success in the treatment or amelioration of an injury, pathology or condition, including any objective or subjective parameter such as abatement; remission; diminishing of symptoms or making the injury, pathology or condition more tolerable to the patient; slowing in the rate of degeneration or decline; making the final point of degeneration less debilitating; improving a patient's physical or mental well-being.
  • the treatment or amelioration of symptoms can be based on objective or subjective parameters; including the results of a physical examination, neuropsychiatric exams, and/or a psychiatric evaluation.
  • the certain methods presented herein successfully treat cancer by decreasing the incidence of cancer, in inhibiting its growth and or causing remission of cancer.
  • an “effective amount” is an amount of a kinase inhibitor sufficient to contribute to the treatment, prevention, or reduction of a symptom or symptoms of a disease, or to inhibit the activity or a protein kinase relative to the absence of the kinase inhibitor. Where recited in reference to a disease treatment, an “effective amount” may also be referred to as a “therapeutically effective amount.”
  • a “reduction” of a symptom or symptoms means decreasing of the severity or frequency of the symptom(s), or elimination of the symptom(s).
  • a “prophylactically effective amount” of a drug is an amount of a drug that, when administered to a subject, will have the intended prophylactic effect, e.g., preventing or delaying the onset (or reoccurrence) a disease, or reducing the likelihood of the onset (or reoccurrence) of a disease or its symptoms.
  • the full prophylactic effect does not necessarily occur by administration of one dose, and may occur only after administration of a series of doses. Thus, a prophylactically effective amount may be administered in one or more administrations.
  • kinase refers to an enzyme that transfers a phosphate group from a donor molecule (e.g. ATP) to a substrate.
  • a donor molecule e.g. ATP
  • phosphorylation The process of transferring a phosphate group from a donor to a substrate is conventionally known as phosphorylation.
  • substrate in the context of protein phosphorylation refers to a compound (e.g. protein) which accepts a phosphate group and is thus phosphorylated.
  • kinase inhibitors are provided.
  • the kinase inhibitors are typically reversible kinase inhibitors.
  • the kinase inhibitor has the structure of Formula I:
  • R 1 is substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, or -L 1A -R 1A .
  • R 1A is substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl.
  • L 1 -R 1 and/or R 1 is/are generally designed to fit within a kinase ATP binding site and/or bind to amino acids within the kinase ATP binding site (e.g. a kinase ATP binding site moiety).
  • L 1 is a bond, —C(O)—, —C(O)N(L 3 R 2 )—, —C(O)O—, —S(O) n —, —O—, —N(L 3 R 2 )—, —P(O)(OL 3 R 2 )O—, —SO 2 N(L 3 R 2 )—, —P(O)(NL 3 R 2 )N—, substituted or unsubstituted alkylene, substituted or unsubstituted heteroalkylene, substituted or unsubstituted cycloalkylene, substituted or unsubstituted heterocycloalkylene, substituted or unsubstituted arylene, or substituted or unsubstituted heteroarylene.
  • L′ is a bond.
  • L 1A is a bond, —C(O)—, —C(O)N(L 3 R 2′ )—, —C(O)O—, —S(O) n′ —, —O—, —N(L 3 R 2′ )—, —P(O)(OL 3 R 2′ )O—, —SO 2 N(L 3 ′R 2′ )—, —P(O)(N L 3 ′R 2′ )N—, substituted or unsubstituted alkylene, substituted or unsubstituted heteroalkylene, substituted or unsubstituted cycloalkylene, substituted or unsubstituted heterocycloalkylene, substituted or unsubstituted arylene, or substituted or unsubstituted heteroarylene.
  • n′ is 0, 1 or 2.
  • L 2 is a bond, —C(O)—, —C(O)N(L 3A R 2A )—, —C(O)O—, —S(O) t —, —O—, —N(L 3A R 2A )—, —P(O)(OL 3A R 2A )O—, —SO 2 N(L 3A R 2A )—, —P(O)(NL 3A R 2A )N—, substituted or unsubstituted alkylene, substituted or unsubstituted heteroalkylene, substituted or unsubstituted cycloalkylene, substituted or unsubstituted heterocycloalkylene, substituted or unsubstituted arylene, or substituted or unsubstituted heteroarylene.
  • the symbol t is 0, 1 or 2.
  • L 3 , L 3′ and L 3A are independently a bond, substituted or unsubstituted alkylene, substituted or unsubstituted heteroalkylene, substituted or unsubstituted cycloalkylene, substituted or unsubstituted heterocycloalkylene, substituted or unsubstituted arylene, or substituted or unsubstituted heteroarylene.
  • the symbol w is 0, 1 or 2.
  • R 2 , R 2′ and R 2A are independently hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.
  • E is an electron withdrawing group or together with L 2 forms an electron withdrawing group (e.g. -L 2 -E may form an electron withdrawing group).
  • E is ring A or R 4 , wherein R 4 and ring A are is as defined below.
  • E may be hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl or -L 5A -R 4A .
  • E may also be hydrogen, R 23A -substituted or unsubstituted alkyl, R 23A -substituted or unsubstituted heteroalkyl, R 23A -substituted or unsubstituted cycloalkyl, R 23A -substituted or unsubstituted heterocycloalkyl, R 23A -substituted or unsubstituted aryl, or R 23A -substituted or unsubstituted heteroaryl.
  • E is a substituted or unsubstituted heteroaryl (e.g.
  • R 23A substituted or unsubstituted heteroaryl or substituted or unsubstituted heterocycloalkyl (e.g. R 23A -substituted or unsubstituted heterocycloalkyl).
  • E may simply be hydrogen.
  • electron withdrawing group refers to a chemical substituent that modifies the electrostatic forces acting on a nearby chemical reaction center by withdrawing negative charge from that chemical reaction center.
  • electron withdrawing groups draw electrons away from a reaction center.
  • the reaction center is fractionally more positive than it would be in the absence of the electron-withdrawing group.
  • the chemical reaction center is one of the two carbons forming the carbon-carbon double bond (olefin).
  • the chemical reaction center is the olefin carbon attached to -L 1 -R 1 .
  • the electron withdrawing group functions to draw charge or electrons away from this olefin carbon thereby making the olefin carbon electron deficient (relative to the absence of the electron withdrawing group).
  • the electron deficient olefin carbon is thereby rendered more reactive toward electron rich chemical groups, such as the sulfhydryl of a kinase active site cysteine.
  • E and -L 2 -E are typically substituents that sufficiently withdraw electrons from the reaction center olefin carbon to reversibly bind to the sulfhydryl of a kinase active cite cysteine (e.g. measurably reversibly binding when the kinase is fully denatured or partly denatured).
  • cysteine e.g. measurably reversibly binding when the kinase is fully denatured or partly denatured.
  • -L 2 -E is as set forth in one of the Formulae set forth below
  • Formula II -L 2 -E is —C(O)X(L 1 ′—R 3 ) z (L 5 -R 4 )
  • Formula IIIc -L 2 -NR 3 R 4 in Formula IIIc -L 2 -E is —WNR 3 R 4 , etc.
  • -L 2 -E may be as set forth in the Formulae provided herein and combined with the definitions and embodiments of -L 1 -R 1 provided herein.
  • -L 1 -R 1 may be as set forth in the Formulae provided herein (e.g. Formula IIIa to IIIe wherein -L 1 -R 1 includes a pyrrolopyrimidinyl) and combined with the definitions of -L 2 -E as provided herein.
  • groups capable of withdrawing electrons from a reaction center include, but are not limited to, —NO 2 , —N(R 2 ), —N(R 3 ) + , —N(H 3 ) + , —SO 3 H, —SO 3 R′, —S(O 2 )R′ (sulfone), —S(O)R′ (sulfoxide), —S(O 2 )NH 2 (sulfonamide), —SO 2 NHR′, —SO 2 NR′ 2 , —PO(OR′) 2 , —PO 3 H 2 , —PO(NR′ 2 ) 2 , pyridinyl (2-, 3-, 4-), pyrazolyl, indazolyl, imidazolyl, thiazolyl, benzothiazolyl, oxazolyl, benzimidazolyl, benzoxazolyl, isoxazolyl, benzisoxazolyl, triazoly
  • R, R′ and R′′ are independently hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, or similar Substituents (e.g. a substituent group, a size limited substituent group or a lower substituent group).
  • the term “electron withdrawing group” is distinguished from an “electron donating group” as known in the art.
  • the kinase inhibitors provided herein are reversible kinase inhibitors, and may measurably dissociate from the protein kinase when the protein kinase is not denatured, partially denatured, or fully denatured.
  • the covalent reversible kinase inhibitor measurably dissociates from the protein kinase only when the protein kinase is fully denatured or partially denatured, but does not measurably dissociate from the protein kinase when the protein kinase is intact, or dissociates at least 10, 1 ⁇ 10 2 , 1 ⁇ 10 3 , 1 ⁇ 10 4 , 1 ⁇ 10 5 , 1 ⁇ 10 6 , 1 ⁇ 10 7 , 1 ⁇ 10 8 , 1 ⁇ 10 9 , 1 ⁇ 10 10 fold slower when the protein kinase is intact relative to the dissociation when the protein kinase is fully or partially denatured (referred to herein as a “covalent reversible denatured kinase inhibitor”).
  • the protein kinase is denatured or fully denatured (i.e. not intact) when placed in denaturing solution, such as 6 N guanidine, 1% SDS, 50% MeCN, or similar protein denaturant, for seconds or minutes (e.g. 30 to 120 seconds, such as 60 seconds).
  • denaturing solution such as 6 N guanidine, 1% SDS, 50% MeCN, or similar protein denaturant
  • the reversible kinase inhibitors described herein, after covalently binding to the kinase active site cysteine residue are capable of dissociating from the kinase within seconds or minutes after denaturing/unfolding the kinase with 6 N guanidine, 1% SDS, 50% MeCN, or similar protein denaturant.
  • -L 2 -E is not —C(O)NH 2 .
  • L 1 is a bond and R 1 is a substituted 4-amino pyrrolopyrimidinyl
  • -L 2 -E is not —C(O)NH 2 .
  • L′ is a bond and R 1 is a substituted pyrrolopyrimidinyl
  • -L 2 -E is not —C(O)NH 2
  • L 1 is a bond and R 1 is a substituted or unsubstituted pyrrolopyrimidinyl
  • -L 2 -E is not —C(O)NH 2
  • R 1 is a substituted or unsubstituted pyrrolopyrimidinyl
  • -L 2 -E is not —C(O)NH 2 .
  • -L 2 -E is not —C(O)NH 2 .
  • -L 2 -E is not —C(O)OH, or —C(O)OR′′, wherein R′′ is an unsubstituted C 1 -C 10 alkyl (e.g. unsubstituted C 1 —O 5 alkyl such as methyl).
  • -L 2 -E is not —C(O)N(CH 3 ) 2 or —C(O)NH(CH 3 )
  • R′ and R 1A are independently R 7 -substituted or unsubstituted alkyl, R 7 -substituted or unsubstituted heteroalkyl, R 7 -substituted or unsubstituted cycloalkyl, R 7 -substituted or unsubstituted heterocycloalkyl, R 7 -substituted or unsubstituted aryl, or R 7 -substituted or unsubstituted heteroaryl.
  • R 7 is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 8 -substituted or unsubstituted alkyl, R 8 -substituted or unsubstituted heteroalkyl, R 8 -substituted or unsubstituted cycloalkyl, R 8 -substituted or unsubstituted heterocycloalkyl, R 8 -substituted or unsubstituted aryl, R 8 -substituted or unsubstituted heteroaryl, or -L 6 -R 7A .
  • L 6 is —O—, —NH—, —C(O)—, —C(O)NH—, —S(O) m —, or —S(O) m NH—, where m is 0, 1, or 2.
  • R 7A is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 8 -substituted or unsubstituted alkyl, R 8 -substituted or unsubstituted heteroalkyl, R 8 -substituted or unsubstituted cycloalkyl, R 8 -substituted or unsubstituted heterocycloalkyl, R 8 -substituted or unsubstituted aryl, or R 8 -substituted or unsubstituted heteroaryl.
  • R 8 is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 9 -substituted or unsubstituted alkyl, R 9 -substituted or unsubstituted heteroalkyl, R 9 -substituted or unsubstituted cycloalkyl, R 9 -substituted or unsubstituted heterocycloalkyl, R 9 -substituted or unsubstituted aryl, R 9 -substituted or unsubstituted heteroaryl or -L 9 -R 9A .
  • R 8 is independently —OH or unsubstituted alkyl.
  • L 9 is —O—, —NH—, —C(O)—, —C(O)NH—, —S(O) m ′—, or —S(O) m′ NH—, where m′ is 0, 1, or 2.
  • R 9A is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 10 -substituted or unsubstituted alkyl, R 10 -substituted or unsubstituted heteroalkyl, R 10 -substituted or unsubstituted cycloalkyl, R 10 -substituted or unsubstituted heterocycloalkyl, R 10 -substituted or unsubstituted aryl, or R 10 -substituted or unsubstituted heteroaryl.
  • R 9 is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 10 -substituted or unsubstituted alkyl, R 10 -substituted or unsubstituted heteroalkyl, R 10 -substituted or unsubstituted cycloalkyl, R 10 -substituted or unsubstituted heterocycloalkyl, R 10 -substituted or unsubstituted aryl, or R 10 -substituted or unsubstituted heteroaryl.
  • R 10 is independently halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , unsubstituted alkyl, unsubstituted heteroalkyl, unsubstituted cycloalkyl, unsubstituted heterocycloalkyl, unsubstituted aryl, or unsubstituted heteroaryl.
  • R 1 and R 1A are independently R 7 -substituted or unsubstituted 6,5 fused ring heteroaryl, R 7 -substituted or unsubstituted 5,6 fused ring heteroaryl, R 7 -substituted or unsubstituted 5,5 fused ring heteroaryl, R 7 -substituted or unsubstituted 6,6 fused ring heteroaryl, or R 7 -substituted or unsubstituted 5 or 6 membered heteroaryl having at least 2 (e.g. 2 to 4) ring nitrogens.
  • R 1 and R 1A are independently R 7 -substituted phenyl, R 7 -substituted piperidinyl, R 7 -substituted 6-membered heterocycloalkyl, R 7 -substituted or unsubstituted 6,5 fused ring heteroaryl, R 7 -substituted or unsubstituted 5,6 fused ring heteroaryl.
  • R 7 may be halogen, —CN, —OH, —COOH, R 8 -substituted or unsubstituted alkyl, R 8 -substituted or unsubstituted heteroalkyl, R 8 -substituted or unsubstituted cycloalkyl, R 8 -substituted or unsubstituted heterocycloalkyl, R 8 -substituted or unsubstituted aryl, or R 8 -substituted or unsubstituted heteroaryl or -L 6 -R 7A .
  • R 7A may be R 8 -substituted or unsubstituted cycloalkyl, R 8 -substituted or unsubstituted heterocycloalkyl, R 8 -substituted or unsubstituted aryl, or R 8 -substituted or unsubstituted heteroaryl.
  • L 6 may be —O—, —NH—, —C(O)—, —C(O)NH—, —S(O) m —, or —S(O) m NH—.
  • R 8 may be —OH, or R 9 -substituted or unsubstituted alkyl.
  • R 7 is independently R 8 -substituted or unsubstituted cycloalkyl, R 8 -substituted or unsubstituted heterocycloalkyl, R 8 -substituted or unsubstituted aryl, R 8 -substituted or unsubstituted heteroaryl, or -L 6 -R 7A .
  • R 7 is R 8 -substituted or unsubstituted heteroaryl, or -L 6 -R 7A .
  • L 6 may be —C(O)—.
  • R 7A may be R 8 -substituted or unsubstituted heteroaryl.
  • R 1 or R 1A is a substituted or unsubstituted heteroaryl, such as an R 7 -substituted or unsubstituted heteroaryl.
  • the heteroaryl may be a substituted or unsubstituted pyrrolopyrimidinyl, substituted or unsubstituted indolyl, substituted or unsubstituted pyrazolyl, substituted or unsubstituted indazolyl, substituted or unsubstituted imidazolyl, substituted or unsubstituted thiazolyl, substituted or unsubstituted benzothiazolyl, substituted or unsubstituted oxazolyl, substituted or unsubstituted benzimidazolyl, substituted or unsubstituted benzoxazolyl, substituted or unsubstituted isoxazolyl, substituted or unsubstituted benzisoxazolyl, substituted or un
  • R 1 or R 1A is a substituted or unsubstituted 6,5 fused ring heteroaryl, a substituted or unsubstituted 5,6 fused ring heteroaryl, a substituted or unsubstituted 5,5 fused ring heteroaryl, or a substituted or unsubstituted 6,6 fused ring heteroaryl.
  • R 1 or R 1A is a substituted or unsubstituted 5 or 6 membered heteroaryl having at least 2 (e.g. 2 to 4) ring nitrogens.
  • any R 1 substituent may be R 7 -substituted, including the substituents recited in this paragraph.
  • R 1 and/or -L 1 -R 1 is/are generally designed to be a kinase ATP binding site moiety. It has also been found herein that compounds of Formula I in which R 1 and/or -L 1 -R 1 is/are attached to the remainder of the compound via an sp2 carbon, stability of the compound is improved.
  • a “kinase ATP binding site moiety,” as used herein, is a moiety capable of fitting within a kinase ATP binding site and/or binding to amino acids within the kinase ATP binding site. Kinase ATP binding sites are well known for wide variety of kinases, and may be easily determined from the primary amino acid structure of a kinase using computer modeling techniques commonly employed in the art.
  • -L 1 -R 1 is a kinase ATP binding site moiety and the electron deficient olefin carbon binds to a sulfhydryl of a kinase active site cysteine.
  • the kinase inhibitors provided herein bind to at least two points of the protein kinase: at least one residue within the ATP binding site moiety and a sulfhydryl of a kinase active site cysteine.
  • -L 1 -R 1 does not have the formula:
  • -L 1 -R 1 is not a phenyl substituted with hydroxyl. In some embodiments, -L 1 -R 1 includes a substituted or unsubstituted heteroaryl or substituted or unsubstituted heteroarylene group.
  • L 1 -R 1 and/or R 1 is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl.
  • L 1 is a bond and R 1 is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl.
  • L 1 is a substituted or unsubstituted arylene or substituted or unsubstituted heteroarylene and R 1 or R 1A is a substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl.
  • L 1 may be substituted or unsubstituted arylene and R 1 or R 1A may be substituted or unsubstituted heteroaryl.
  • L 1A is —C(O)—, —C(O)NH—, —C(O)O—, —S(O)—, —SO 2 —. —S—, —O—, —NH— or —SO 2 NH—.
  • L 1 is a bond and R 1 is an R 7 -substituted phenyl.
  • R 7 is -L 6 -R 7A or R 8 -substituted or unsubstituted heteroaryl, wherein R 7A is R 8 -substituted or unsubstituted heteroaryl.
  • L 6 is a bond or —C(O)—.
  • the R 8 -substituted or unsubstituted heteroaryl is an R 8 -substituted or unsubstituted 6,5 fused ring heteroaryl or R 8 -substituted or unsubstituted 5,6 fused ring.
  • L 1 is a bond and R 1 is an R 7 -substituted phenyl, R 7 -substituted piperidinyl, R 7 -substituted piperizinyl, R 7 -substituted pyrrolidinyl, R 7 -substituted piperidinyl, R 7 -substituted azepanyl, or R 7 -substituted azetidinyl.
  • R 7 is -L 6 -R 7A or R 8 -substituted or unsubstituted heteroaryl, wherein R 7A is R 8 -substituted or unsubstituted heteroaryl.
  • L 6 is a bond or —C(O)—.
  • the R 8 -substituted or unsubstituted heteroaryl is an R 8 -substituted or unsubstituted 6,5 fused ring heteroaryl or R 8 -substituted or unsubstituted 5,6 fused ring.
  • L′ is a substituted or unsubstituted arylene (e.g. phenylene) and R 1 or R 1A is a substituted or unsubstituted heteroaryl (e.g. R 7 -substituted). In other embodiments, L′ is a substituted or unsubstituted heteroarylene.
  • L′ is a bond, —C(O)—, —C(O)N(L 3 R 2 )—, —C(O)O—, —S(O) n —, —O—, —S—, —N(L 3 R 2 )—, —P(O)(OL 3 R 2 )O—, —SO 2 N(L 3 R 2 )—, —P(O)(NL 3 R 2 )N—, R 11 -substituted or unsubstituted alkylene, R 11 -substituted or unsubstituted heteroalkylene, R 11 -substituted or unsubstituted cycloalkylene, R 11 -substituted or unsubstituted heterocycloalkylene, R 11 -substituted or unsubstituted arylene, or R 11 -substituted or unsubstituted or unsubstituted or unsubsti
  • L 1A may be a bond, —C(O)—, —C(O)N(L 3 R 2′ )-, —C(O)O—, —S(O) n′ —, —O—, —N(L 3 R 2′ )-, —P(O)(OL 3 R 2′ )O—, —SO 2 N(L 3 R 2′ )-, —P(O)(N L 3 R 2′ )N—, R 11 -substituted or unsubstituted alkylene, R 11 -substituted or unsubstituted heteroalkylene, R 11 -substituted or unsubstituted cycloalkylene, R 11 -substituted or unsubstituted heterocycloalkylene, R 11 -substituted or unsubstituted arylene, or R 11 -substituted or unsubstituted heteroarylene.
  • L 1 and L 1A may also independently be a bond, R 11 -substituted or unsubstituted alkylene, R 11 -substituted or unsubstituted heteroalkylene, R 11 -substituted or unsubstituted cycloalkylene, R 11 -substituted or unsubstituted heterocycloalkylene, R 11 -substituted or unsubstituted arylene, or R 11 -substituted or unsubstituted heteroarylene.
  • R 11 is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 12 -substituted or unsubstituted alkyl, R 12 -substituted or unsubstituted heteroalkyl, R 12 -substituted or unsubstituted cycloalkyl, R 12 -substituted or unsubstituted heterocycloalkyl, R 12 -substituted or unsubstituted aryl, or R 12 -substituted or unsubstituted heteroaryl.
  • R 12 is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 13 -substituted or unsubstituted alkyl, R 13 -substituted or unsubstituted heteroalkyl, R 13 -substituted or unsubstituted cycloalkyl, R 13 -substituted or unsubstituted heterocycloalkyl, R 13 -substituted or unsubstituted aryl, or R 13 -substituted or unsubstituted heteroaryl.
  • R 13 is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 14 -substituted or unsubstituted alkyl, R 14 -substituted or unsubstituted heteroalkyl, R 14 -substituted or unsubstituted cycloalkyl, R 14 -substituted or unsubstituted heterocycloalkyl, R 14 -substituted or unsubstituted aryl, or R 14 -substituted or unsubstituted heteroaryl.
  • R 14 is independently halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , unsubstituted alkyl, unsubstituted heteroalkyl, unsubstituted cycloalkyl, unsubstituted heterocycloalkyl, unsubstituted aryl, or unsubstituted heteroaryl.
  • L 2 may be a bond, —C(O)N(L 3A R 2A )—, —C(O)O—, —S(O) t —, —O—, —S—, —N(L 3A R 2A )—, —C(O)—P(O)(OL 3 R 2 ))-, —SO 2 N(L 3 R 2 )—, —P(O)(NL 3 R 2 )N—, R 19 -substituted or unsubstituted alkylene, R 19 -substituted or unsubstituted heteroalkylene, R 19 -substituted or unsubstituted cycloalkylene, R 19 -substituted or unsubstituted heterocycloalkylene, R 19 -substituted or unsubstituted arylene, or R 19 -substituted or unsubstituted heteroarylene.
  • R 19 is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R N -substituted or unsubstituted alkyl, R N -substituted or unsubstituted heteroalkyl, R 20 -substituted or unsubstituted cycloalkyl, R 20 -substituted or unsubstituted heterocycloalkyl, R 20 -substituted or unsubstituted aryl, or R 20 -substituted or unsubstituted heteroaryl.
  • R 20 is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 21 -substituted or unsubstituted alkyl, R 2′ -substituted or unsubstituted heteroalkyl, R 21 -substituted or unsubstituted cycloalkyl, R 21 -substituted or unsubstituted heterocycloalkyl, R 21 -substituted or unsubstituted aryl, or R 21 -substituted or unsubstituted heteroaryl.
  • R 21 is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 22 -substituted or unsubstituted alkyl, R 22 -substituted or unsubstituted heteroalkyl, R 22 -substituted or unsubstituted cycloalkyl, R 22 -substituted or unsubstituted heterocycloalkyl, R 22 -substituted or unsubstituted aryl, or R 22 -substituted or unsubstituted heteroaryl.
  • R 22 is independently halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , unsubstituted alkyl, unsubstituted heteroalkyl, unsubstituted cycloalkyl, unsubstituted heterocycloalkyl, unsubstituted aryl, or unsubstituted heteroaryl.
  • L 3 , L 3′ , and L 3A are independently a bond, R 27 -substituted or unsubstituted alkylene, R 27 -substituted or unsubstituted heteroalkylene, R 27 -substituted or unsubstituted cycloalkylene, R 27 -substituted or unsubstituted heterocycloalkylene, R 27 -substituted or unsubstituted arylene, or R 27 -substituted or unsubstituted heteroarylene.
  • R 27 is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 28 -substituted or unsubstituted alkyl, R 28 -substituted or unsubstituted heteroalkyl, R 28 -substituted or unsubstituted cycloalkyl, R 28 -substituted or unsubstituted heterocycloalkyl, R 28 -substituted or unsubstituted aryl, or R 28 -substituted or unsubstituted heteroaryl.
  • R 28 is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 29 -substituted or unsubstituted alkyl, R 29 -substituted or unsubstituted heteroalkyl, R 29 -substituted or unsubstituted cycloalkyl, R 29 -substituted or unsubstituted heterocycloalkyl, R 29 -substituted or unsubstituted aryl, or R 29 -substituted or unsubstituted heteroaryl.
  • R 29 is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R N -substituted or unsubstituted alkyl, R N -substituted or unsubstituted heteroalkyl, R 30 -substituted or unsubstituted cycloalkyl, R 30 -substituted or unsubstituted heterocycloalkyl, R 30 -substituted or unsubstituted aryl, or R 30 -substituted or unsubstituted heteroaryl.
  • R 30 is independently halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , unsubstituted alkyl, unsubstituted heteroalkyl, unsubstituted cycloalkyl, unsubstituted heterocycloalkyl, unsubstituted aryl, or unsubstituted heteroaryl.
  • R 2 , R 2′ , and R 2A are independently hydrogen, R 15 -substituted or unsubstituted alkyl, R 15 -substituted or unsubstituted heteroalkyl, R 15 -substituted or unsubstituted cycloalkyl, R 15 -substituted or unsubstituted heterocycloalkyl, R 15 -substituted or unsubstituted aryl, or R 15 -substituted or unsubstituted heteroaryl.
  • R 15 is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 16 -substituted or unsubstituted alkyl, R 16 -substituted or unsubstituted heteroalkyl, R 16 -substituted or unsubstituted cycloalkyl, R 16 -substituted or unsubstituted heterocycloalkyl, R 16 -substituted or unsubstituted aryl, R 16 -substituted or unsubstituted heteroaryl, or -L 7 -R 15A .
  • L 7 is independently —O—, —C(O)—, —C(O)NH—, —S(O) Y —, or —S(O) y NH—, where y is 0, 1, or 2.
  • R 15A is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 16 -substituted or unsubstituted alkyl, R 16 -substituted or unsubstituted heteroalkyl, R 16 -substituted or unsubstituted cycloalkyl, R 16 -substituted or unsubstituted heterocycloalkyl, R 16 -substituted or unsubstituted aryl, R 16 -substituted or unsubstituted heteroaryl.
  • R 16 is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 17 -substituted or unsubstituted alkyl, R 17 -substituted or unsubstituted heteroalkyl, R 17 -substituted or unsubstituted cycloalkyl, R 17 -substituted or unsubstituted heterocycloalkyl, R 17 -substituted or unsubstituted aryl, or R 17 -substituted or unsubstituted heteroaryl.
  • R 17 is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 18 -substituted or unsubstituted alkyl, R 18 -substituted or unsubstituted heteroalkyl, R 18 -substituted or unsubstituted cycloalkyl, R 18 -substituted or unsubstituted heterocycloalkyl, R 18 -substituted or unsubstituted aryl, or R 18 -substituted or unsubstituted heteroaryl.
  • R 18 is independently halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , unsubstituted alkyl, unsubstituted heteroalkyl, unsubstituted cycloalkyl, unsubstituted heterocycloalkyl, unsubstituted aryl, or unsubstituted heteroaryl.
  • the kinase inhibitor has the structure of Formula II:
  • W is —C(O)— or —S(O) 2 —
  • X is O or N
  • z is 0 or 1, provided, however, that if X is O, then z is 0.
  • L 1 and R 1 are defined as disclosed above for Formula I.
  • X is N. In other embodiments, X is O.
  • R 3 is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.
  • R 4 is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl or -L 5A -R 4A .
  • R 4A is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl.
  • R 3 and R 4 may be joined together with X to form a substituted or unsubstituted heterocycloalkyl or substituted or unsubstituted heteroaryl.
  • R 3 and R 4A may be joined together with X to form a substituted or unsubstituted heterocycloalkyl or substituted or unsubstituted heteroaryl.
  • L 4 and L 5 are independently a bond, —C(O)—, —C(O)N(L 3A R 2A )—, —C(O)O—, —S(O) t , —O—, —N(L 3A R 2A )—, —P(O)(OL 3A R 2A )O—, —SO 2 N(L 3A R 2A )—, —P(O)(NL 3A R 2A )N—, substituted or unsubstituted alkylene, substituted or unsubstituted heteroalkylene, substituted or unsubstituted cycloalkylene, substituted or unsubstituted heterocycloalkylene, substituted or unsubstituted arylene, or substituted or unsubstituted heteroarylene.
  • L 3A , R 2A , t are as defined above.
  • L 5A is a bond, —C(O)—, —C(O)N(L 3A′ R 2A′ )—, —C(O)O—, —S(O) t′ —, —O—, —N(L 3A′ R 2A′ )—, —P(O)(O L 3A′ R 2A′ )O—, —SO 2 N(L 3A′ R 2A′ )—, —P(O)(NL 3A′ R 2A′ )N—, substituted or unsubstituted alkylene, substituted or unsubstituted heteroalkylene, substituted or unsubstituted cycloalkylene, substituted or unsubstituted heterocycloalkylene, substituted or unsubstituted arylene, or substituted or unsubstituted heteroarylene.
  • the symbol t′ is 0, 1 or 2.
  • R 2A′ is independently hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.
  • R 2′ is independently hydrogen, R 15 -substituted or unsubstituted alkyl, R 15 -substituted or unsubstituted heteroalkyl, R 15 -substituted or unsubstituted cycloalkyl, R 15 -substituted or unsubstituted heterocycloalkyl, R 15 -substituted or unsubstituted aryl, or R 15 -substituted or unsubstituted heteroaryl.
  • R 15 is as defined above.
  • L 3A′ is independently a bond, substituted or unsubstituted alkylene, substituted or unsubstituted heteroalkylene, substituted or unsubstituted cycloalkylene, substituted or unsubstituted heterocycloalkylene, substituted or unsubstituted arylene, or substituted or unsubstituted heteroarylene.
  • L 3A′ is a bond, R 27 -substituted or unsubstituted alkylene, R 27 -substituted or unsubstituted heteroalkylene, R 27 -substituted or unsubstituted cycloalkylene, R 27 -substituted or unsubstituted heterocycloalkylene, R 27 -substituted or unsubstituted arylene, or R 27 -substituted or unsubstituted heteroarylene.
  • R 27 is as defined above.
  • L 4 and L 5 are independently a bond, —C(O)—, —C(O)N(L 3A R 2A )—, —C(O)O—, —S(O) t —, —O—, —N(L 3A R 2A )—, —P(O)(OL 3A R 2A )O—, —SO 2 N(L 3A R 2A )—, —P(O)(NL 3A R 2A )N—, R 19 -substituted or unsubstituted alkylene, R 19 -substituted or unsubstituted heteroalkylene, R 19 -substituted or unsubstituted cycloalkylene, R 19 -substituted or unsubstituted heterocycloalkylene, R 19 -substituted or unsubstituted arylene, or R 19 -substituted or unsubstituted or un
  • L 5A is a bond, —C(O)—, —C(O)N(L 3A′ R 2A′ )C(O)O—, —S(O) t′ —, —O—, —N(L 3A′ R 2′ )P(O)(O L 3A′ R 2A′ )O—, —SO 2 N(L 3A′ R 2A′ )—, —P(O)(NL 3A′ R 2A′ )N—, R 19 -substituted or unsubstituted alkylene, R 19 -substituted or unsubstituted heteroalkylene, R 19 -substituted or unsubstituted cycloalkylene, R 19 -substituted or unsubstituted heterocycloalkylene, R 19 -substituted or unsubstituted arylene, or R 19 -substituted or unsubstituted or un
  • At least one of L 5 -R 4 or L 4 -R 3 includes a substituted or unsubstituted heteroaryl or substituted or unsubstituted heteroarylene. In some embodiments, one of L 5 -R 4 or L 4 -R 3 includes a substituted or unsubstituted heteroaryl or substituted or unsubstituted heteroarylene. In some embodiments, one of L 5 -R 4 or L 4 -R 3 is a hydrogen.
  • R 1 is (3-(4-amino-5-p-tolyl-7H-pyrrolo[2,3-d]pyrimidin-7-yl)propan-1-ol)-6-yl, then at least one of R 3 and R 4 are not hydrogen.
  • R 1 or R 1A is R 7 -substituted or unsubstituted heterocycloalkyl, R 7 -substituted or unsubstituted aryl, or R 7 -substituted or unsubstituted heteroaryl, wherein R 7 is as described above.
  • R 3 is hydrogen, R 23 -substituted or unsubstituted alkyl, R 23 -substituted or unsubstituted heteroalkyl, R 23 -substituted or unsubstituted cycloalkyl, R 23 -substituted or unsubstituted heterocycloalkyl, R 23 -substituted or unsubstituted aryl, or R 23 -substituted or unsubstituted heteroaryl.
  • R 23 is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 24 -substituted or unsubstituted alkyl, R 24 -substituted or unsubstituted heteroalkyl, R 24 -substituted or unsubstituted cycloalkyl, R 24 -substituted or unsubstituted heterocycloalkyl, R 24 -substituted or unsubstituted aryl, R 24 -substituted or unsubstituted heteroaryl, or -L 8 -R 23A′ .
  • R 23A′ is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 24 -substituted or unsubstituted alkyl, R 24 -substituted or unsubstituted heteroalkyl, R 24 -substituted or unsubstituted cycloalkyl, R 24 -substituted or unsubstituted heterocycloalkyl, R 24 -substituted or unsubstituted aryl, R 24 -substituted or unsubstituted heteroaryl.
  • R 24 is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 25 -substituted or unsubstituted alkyl, R 25 -substituted or unsubstituted heteroalkyl, R 25 -substituted or unsubstituted cycloalkyl, R 25 -substituted or unsubstituted heterocycloalkyl, R 25 -substituted or unsubstituted aryl, or R 25 -substituted or unsubstituted heteroaryl.
  • R 25 is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 26 -substituted or unsubstituted alkyl, R 26 -substituted or unsubstituted heteroalkyl, R 26 -substituted or unsubstituted cycloalkyl, R 26 -substituted or unsubstituted heterocycloalkyl, R 26 -substituted or unsubstituted aryl, or R 26 -substituted or unsubstituted heteroaryl.
  • R 26 is independently halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , unsubstituted alkyl, unsubstituted heteroalkyl, unsubstituted cycloalkyl, unsubstituted heterocycloalkyl, unsubstituted aryl, or unsubstituted heteroaryl.
  • R 3 and R 4 are joined together with X to form a substituted or unsubstituted heteroaryl or substituted or unsubstituted heterocycloalkyl (e.g. R 23 -substituted or unsubstituted heteroaryl or R 23 -substituted or unsubstituted heterocycloalkyl).
  • R 3 and R 4 are joined together with X to form a 4 to 8 membered substituted or unsubstituted heterocycloalkyl or a 5 to 6 membered substituted or unsubstituted heteroaryl (e.g. R 23 -substituted species thereof).
  • R 3 and R 4 are joined together with X to form a 4 to 7 membered substituted or unsubstituted heterocycloalkyl or a 5 to 6 membered substituted or unsubstituted heteroaryl (e.g. R 23 -substituted species thereof). In some embodiments, R 3 and R 4 are joined together with X to form a 5 to 7 membered substituted or unsubstituted heterocycloalkyl or a 5 to 6 membered substituted or unsubstituted heteroaryl (e.g. R 23 -substituted species thereof).
  • R 3 and R 4 are joined together with X to form a substituted or unsubstituted morpholino, substituted or unsubstituted thiomorpholino (or oxidated ring thereof), substituted or unsubstituted pyridinyl, substituted or unsubstituted pyrazyinyl, substituted or unsubstituted pyrrolidinyl, substituted or unsubstituted piperidinyl, substituted or unsubstituted piperizinyl, substituted or unsubstituted pyrrolidinyl, substituted or unsubstituted azepanyl, or substituted or unsubstituted azetidinyl (e.g. R 23 -substituted substituents thereof).
  • R 4 and R 4A are hydrogen, R 23A -substituted or unsubstituted alkyl, R 23A -substituted or unsubstituted heteroalkyl, R 23A -substituted or unsubstituted cycloalkyl, R 23A -substituted or unsubstituted heterocycloalkyl, R 23A -substituted or unsubstituted aryl, or R 23A -substituted or unsubstituted heteroaryl. In some embodiments, R 4 and R 4A are not hydrogen.
  • R 23A is hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 24A -substituted or unsubstituted alkyl, R 24A -substituted or unsubstituted heteroalkyl, R 24A -substituted or unsubstituted cycloalkyl, R 24A -substituted or unsubstituted heterocycloalkyl, R 24A -substituted or unsubstituted aryl, R 24A -substituted or unsubstituted heteroaryl, or -L 7A -R 24B .
  • L 7A is independently —O—, —C(O)—, —C(O)NH—, —S(O) y′ —, or —S(O) y NH—, wherein y′ is 0, 1, or 2.
  • R 24B is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 24A -substituted or unsubstituted alkyl, R 24A -substituted or unsubstituted heteroalkyl, R 24A -substituted or unsubstituted cycloalkyl, R 24A -substituted or unsubstituted heterocycloalkyl, R 24A -substituted or unsubstituted aryl, R 24A -substituted or unsubstituted heteroaryl.
  • R 24A is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 25A -substituted or unsubstituted alkyl, R 25A -substituted or unsubstituted heteroalkyl, R 25A -substituted or unsubstituted cycloalkyl, R 25A -substituted or unsubstituted heterocycloalkyl, R 25A -substituted or unsubstituted aryl, or R 25A -substituted or unsubstituted heteroaryl.
  • R 25A is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 26A -substituted or unsubstituted alkyl, R 26A -substituted or unsubstituted heteroalkyl, R 26A -substituted or unsubstituted cycloalkyl, R 26A -substituted or unsubstituted heterocycloalkyl, R 26A -substituted or unsubstituted aryl, or R 26A -substituted or unsubstituted heteroaryl.
  • R 26A is independently halogen, —CN, —OH, —NH 2 , —COON, —CF 3 , unsubstituted alkyl, unsubstituted heteroalkyl, unsubstituted cycloalkyl, unsubstituted heterocycloalkyl, unsubstituted aryl, or unsubstituted heteroaryl.
  • R 4 or R 4A is R 23A -substituted or unsubstituted alkyl.
  • R 3 is substituted or unsubstituted pyridinyl (2-, 3-, 4-), substituted or unsubstituted pyrazolyl, substituted or unsubstituted indazolyl, substituted or unsubstituted imidazolyl, substituted or unsubstituted thiazolyl, substituted or unsubstituted benzothiazolyl, substituted or unsubstituted oxazolyl, substituted or unsubstituted benzimidazolyl, substituted or unsubstituted benzoxazolyl, substituted or unsubstituted isoxazolyl, substituted or unsubstituted benzisoxazolyl, substituted or unsubstituted triazolyl, substituted or unsubstituted benzotriazolyl, substituted or
  • L 1 , L 4 and L 5 are independently a bond
  • R 1 is R 7 -substituted or unsubstituted heterocycloalkyl, R 7 -substituted or unsubstituted aryl, or R 7 -substituted or unsubstituted heteroaryl.
  • R 7 is independently —NH 2 , R 8 -substituted or unsubstituted alkyl, R 8 -substituted or unsubstituted aryl, R 8 -substituted or unsubstituted heteroaryl, or -L 6 -R 7A .
  • L 6 may be —C(O)—.
  • R 7A may be R 8 -substituted or unsubstituted alkyl, R 8 -substituted or unsubstituted aryl, R 8 -substituted or unsubstituted heteroaryl.
  • R 8 may be —OH or R 9 -substituted or unsubstituted alkyl.
  • R 4 may be hydrogen or R 15 -substituted or unsubstituted alkyl, and R 3 may be hydrogen or R 23 -substituted or unsubstituted alkyl.
  • R 3 and R 4 may optionally be joined together with X to form a 4-7 membered (e.g. 5-7) membered heterocycloalkyl (e.g. an R 23 substituted species thereof).
  • R 7 is R 8 -substituted or unsubstituted heteroaryl, or -L 6 -R 7A .
  • L 4 may be —C(O)—
  • R 7A is R 8 -substituted or unsubstituted heteroaryl.
  • R 7 , R 8 , L 6 , R 7A are as defined above.
  • R 1 is R 7 -substituted phenyl.
  • R 7 is R 8 -substituted or unsubstituted heteroaryl or -L 6 -R 7A .
  • L 6 may be —C(O)—, and R 7A may be R 8 -substituted or unsubstituted heteroaryl.
  • R 3 and R 4 are hydrogen.
  • R 7 is R 8 -substituted or unsubstituted purinyl, R 8 -substituted or unsubstituted pyrimidinyl, R 8 -substituted or unsubstituted imidazolyl, R 8 -substituted or unsubstituted 1H-pyrrolo[2,3-b]pyridinyl, R 8 -substituted or unsubstituted pyrimidinyl, R 8 -substituted or unsubstituted 1H-indazolyl, or R 8 -substituted or unsubstituted 7H-pyrrolo[2,3-d]pyrimidinyl.
  • R 7 may also be R 8 -substituted or unsubstituted pyrrolopyrimidinyl, R 8 -substituted or unsubstituted indolyl, R 8 -substituted or unsubstituted pyrazolyl, R 8 -substituted or unsubstituted indazolyl, R 8 -substituted or unsubstituted imidazolyl, R 8 -substituted or unsubstituted thiazolyl, R 8 -substituted or unsubstituted benzothiazolyl, R 8 -substituted or unsubstituted oxazolyl, R 8 -substituted or unsubstituted benzimidazolyl, R 8 -substituted or unsubstituted benzoxazolyl, R 8 -substituted or unsubstituted isoxazo
  • R 1 is R 8 -substituted or unsubstituted 6,5 fused ring heteroaryl, R 8 -substituted or unsubstituted 5,6 fused ring heteroaryl, R 8 -substituted or unsubstituted 5,5 fused ring heteroaryl, or R 8 -substituted or unsubstituted 6,6 fused ring heteroaryl.
  • R 1 is a R 8 -substituted or unsubstituted 5 or 6 membered heteroaryl having at least 2 (e.g. 2 to 4) ring nitrogens.
  • R 7 is -L 4 -R 7A and R 7A is R 8 -substituted or unsubstituted 1H-pyrrolo[2,3-b]pyridinyl.
  • X is N
  • R 1 is R 7 -substituted 6-membered heterocycloalkyl.
  • R 7 may be R 8 -substituted or unsubstituted heteroaryl.
  • R 1 may be R 7 -substituted piperidinyl.
  • R 7 may be R 8 -substituted or unsubstituted purinyl, R 8 -substituted or unsubstituted pyrimidinyl, R 8 -substituted or unsubstituted imidazolyl, R 8 -substituted or unsubstituted 1H-pyrrolo[2,3-b]pyridinyl, R 8 -substituted or unsubstituted pyrimidinyl, R 8 -substituted or unsubstituted 1H-indazolyl, or R 8 -substituted or unsubstituted 7H-pyrrolo[2,3-d]pyrimidinyl.
  • R 3 and R 4 are hydrogen.
  • R 1 is R 7 -substituted or unsubstituted 6,5 fused ring heteroaryl, or R 7 -substituted or unsubstituted 5,6 fused ring heteroaryl.
  • R 7 may be —NH 2
  • R 8 -substituted or unsubstituted alkyl R 8 -substituted or unsubstituted aryl
  • R 8 is independently —OH or unsubstituted alkyl.
  • R 1 is R 7 -substituted or unsubstituted indazolyl, or R 7 -substituted or unsubstituted R 1 -pyrrolo[2,3-d]pyrimidinyl.
  • R 3 and R 4 are hydrogen.
  • R 1 is R 7 -substituted or unsubstituted indazole, or R 7 -substituted or unsubstituted 7H-pyrrolo[2,3-d]pyrimidinyl
  • R 3 is unsubstituted alkyl
  • R 4 is hydrogen
  • R 3 is phenylmethyl
  • R 4 is hydrogen.
  • R 3 and R 4 join with N to form a R 23 -substituted or unsubstituted pyrrolidinyl.
  • X is O
  • R 1 is R 7 -substituted or unsubstituted 6,5 fused ring heteroaryl, or R 7 -substituted or unsubstituted 5,6 fused ring heteroaryl.
  • R 7 may be —NH 2
  • R 8 may be —OH or unsubstituted alkyl, and R 3 may be unsubstituted alkyl.
  • R 1 is R 7 -substituted 7H-pyrrolo[2,3-d]pyrimidine, and R 7 is —NH 2 , R 8 -substituted or unsubstituted alkyl, or R 8 -substituted or unsubstituted phenyl.
  • the —C(O)X(L 4 -R 3 ) z (L 5 -R 4 ) substituent of Formula II is an electron withdrawing group. Therefore, the —C(O)X(L 4 -R 3 ) z (L 5 -R 4 ) is capable of withdrawing negative charge from the olefin moiety to which it is attached.
  • the —C(O)X(L 4 -R 3 ) z (L 5 -R 4 ) is capable of sufficiently withdrawing negative charge from the olefin moiety to which it is attached to allow a thiol adduct to form between an olefin carbon and the sulfhydryl of a kinase active site cysteine as discussed herein.
  • R 1 is substituted or unsubstituted heteroaryl.
  • R 1 is substituted or unsubstituted pyrazolopyrimidinyl (e.g. R 7 -substituted or unsubstituted pyrazolopyrimidinyl).
  • R 1 is substituted or unsubstituted pyrrolopyrimidinyl (e.g. R 7 -substituted or unsubstituted pyrrolopyrimidinyl).
  • the kinase inhibitor has the structure:
  • z is an integer from 1 to 4.
  • L 1 , L 2 and R 7 are as defined above.
  • L 1 is a bond.
  • the kinase inhibitor has the structure:
  • z is an integer from 1 to 4.
  • L 1 , L 2 , R 3 , R 4 and R 7 are as defined above.
  • L 1 is a bond.
  • the kinase inhibitor has the structure:
  • W is —C(O)— or —S(O) 2 —, z is an integer from 1 to 4.
  • L 1 , R 3 , R 4 and R 7 are as defined above. In some embodiments, L 1 is a bond.
  • the kinase inhibitor has the structure:
  • W is —C(O)— or —S(O) 2 —, z is an integer from 1 to 4.
  • L′, L 2 , R 3 , R 4 and R 7 are as defined above.
  • L′ is a bond.
  • the kinase inhibitor is compound 43, 44, 45, 46, 47, 48, 49 and 50.
  • the kinase inhibitor has the structure:
  • W is —C(O)— or —S(O) 2 —
  • w is an integer from 1 to 5 (e.g. 1).
  • L 1 , L 2 , R 3 , R 4 , R 7 and R 8 are as defined above.
  • L′ is substituted or unsubstituted arylene. In some embodiments, L′ is substituted or unsubstituted phenylene.
  • the kinase inhibitor has the structure:
  • R 1 , L 2 , E and R 11 are as defined above.
  • the symbol q is an integer from 1 to 4.
  • R 11 is hydrogen.
  • R 1 is substituted or unsubstituted heteroaryl.
  • R 1 is substituted or unsubstituted pyrrolopyrimidine.
  • R 1 is substituted or unsubstituted heteroaryl, q is 0, and L 1 is substituted or unsubstituted phenylene.
  • the kinase inhibitor has the structure:
  • R 7 , L 2 , E and R 11 are as defined above.
  • the symbol q is an integer from 0 to 4.
  • R 11 is hydrogen.
  • the symbol z is an integer from 1 to 4.
  • the kinase inhibitor has the structure:
  • R 7 , L 2 , and E are as defined above.
  • the symbol z is an integer from 1 to 4.
  • E is —NR 3 R 4 (as defined above).
  • the kinase inhibitor has the structure:
  • R 7 , L 2 , R 3 and R 4 are as defined above.
  • the symbol z is an integer from 1 to 4.
  • L 2 is —C(O)—.
  • the kinase inhibitor has the structure:
  • W is —C(O)— or —S(O) 2 —
  • R 7 , R 3 and R 4 are as defined above.
  • the symbol z is an integer from 1 to 4.
  • R 7 is independently —NH 2 , or substituted or unsubstituted aryl.
  • one of R 7 is —NH 2 , and another R 7 is substituted aryl.
  • the kinase inhibitor has the structure:
  • W is —C(O)— or —S(O) 2 —
  • R 7 , R 3 and R 4 are as defined above.
  • R 3 and R 4 are independently hydrogen, unsubstituted or substituted alkyl, or joined together to form a substituted or unsubstituted heteroalkyl.
  • R 7 may be substituted or unsubstituted phenyl.
  • the kinase inhibitor is compound 51, 52, 53, 54, 55, 56, 57, 58, 59, 60 or 61.
  • the kinase inhibitor has the structure:
  • R 1 , L 2 and E are as defined above.
  • E is —NR 3 R 4 as defined above).
  • L 2 may be —C(O)—.
  • the kinase inhibitor has the structure:
  • W is —C(O)— or —S(O) 2 —, R 1 , R 3 and R 4 are as defined above. In certain embodiments, R 1 is substituted alkyl.
  • the kinase inhibitor has the structure:
  • W is —C(O)— or —S(O) 2 —, R 3 and R 4 are as defined above.
  • the kinase inhibitor is compound 38 or 39.
  • R 1 is substituted or unsubstituted indazolyl
  • the kinase inhibitor has the structure of Formula VIa following, wherein R 7 is as defined herein.
  • the kinase inhibitor has the formula:
  • L 2 , E and R 7 are as defined above.
  • the symbol z is an integer from 1 to 4.
  • the kinase inhibitor has the formula:
  • W is —C(O)— or —S(O) 2 —, R 3 , R 4 and R 7 are as defined above.
  • the symbol z is an integer from 1 to 4.
  • the kinase inhibitor has the formula:
  • W is —C(O)— or —S(O) 2 —, R 3 , R 4 and R 7 are as defined above.
  • the kinase inhibitor is compound 37, 40, 41 or 42.
  • L 1 is substituted or unsubstituted heterocycloalkylene, L 2 is —C(O)—.
  • R 1 is substituted or unsubstituted heteroaryl, E is —NR 3 R 4 .
  • L 1 is piperidinyl.
  • R 1 is purinyl.
  • the kinase inhibitor has the formula:
  • W is —C(O)— or —S(O) 2 —, R 3 and R 4 are as defined above.
  • the kinase inhibitor is compound 36.
  • the kinase inhibitor may be a reversible kinase inhibitor (as discussed herein).
  • the kinase inhibitor is a reversible denatured kinase inhibitor (as discussed herein).
  • the kinase inhibitor is a covalent reversible kinase inhibitor (as discussed herein).
  • the kinase inhibitor is a covalent reversible denatured kinase inhibitor (as discussed herein).
  • the kinase inhibitor is a thiol covalent reversible denatured kinase inhibitor (as discussed herein).
  • the compounds of the Formulae provided herein, and embodiments thereof are stable at pH 7.5 (e.g. in phosphate-buffered saline at 37° C.). In some embodiments, where the compound of Formula I or II, and embodiments thereof, are stable at pH 7.5, the compound has a half life of greater than 6 hours, 12 hours, 24 hours, or 48 hours. In some embodiments, where the compounds of the Formulae provided herein, and embodiments thereof, are stable at pH 7.5, the compound has a half life of greater than 12 hours. In some embodiments, where the compounds of the Formulae provided herein, and embodiments thereof, are stable at pH 7.5, the compound has a half life of greater than 24 hours.
  • pH 7.5 e.g. in phosphate-buffered saline at 37° C.
  • the compound has a half life of greater than 48 hours.
  • the compounds of the Formulae provided herein, and embodiments thereof exhibit kinase inhibition within a cell.
  • the cell is a prokaryote or eukaryote.
  • the cell may be a eukaryote (e.g. protozoan cell, fungal cell, plant cell or an animal cell).
  • the cell is a mammalian cell such as a human cell, cow cell, pig cell, horse cell, dog cell and cat cell, mouse cell, or rat cell.
  • the cell is a human cell.
  • the cell may form part of an organ or an organism. In certain embodiments, the cell does not form part of an organ or an organism.
  • each substituted or unsubstituted alkyl is a substituted or unsubstituted C 1 -C 20 alkyl
  • each substituted or unsubstituted heteroalkyl is a substituted or unsubstituted 2 to 20 membered heteroalkyl
  • each substituted or unsubstituted cycloalkyl is a substituted or unsubstituted C 4 -C 8 cycloalkyl
  • each substituted or unsubstituted heterocycloalkyl is a substituted or unsubstituted 4 to 8 membered heterocycloalkyl
  • each substituted or unsubstituted alkylene is a substituted or unsubstituted C 1 -C 20 alkylene
  • each substituted or unsubstituted heteroalkylene is a substituted or unsubstituted 2 to 20 membered heteroalkylene
  • each substituted or unsubstituted cycloalkyl is a substituted or
  • each substituted or unsubstituted alkyl is a substituted or unsubstituted C 1 -C 8 alkyl
  • each substituted or unsubstituted heteroalkyl is a substituted or unsubstituted 2 to 8 membered heteroalkyl
  • each substituted or unsubstituted cycloalkyl is a substituted or unsubstituted C 5 -C 7 cycloalkyl
  • each substituted or unsubstituted heterocycloalkyl is a substituted or unsubstituted 5 to 7 membered heterocycloalkyl
  • each substituted or unsubstituted alkylene is a substituted or unsubstituted C 1 -C 8 alkylene
  • each substituted or unsubstituted heteroalkylene is a substituted or unsubstituted 2 to 8 membered heteroalkylene
  • the compounds of the Formulae provided herein is one or more of the compounds set forth in Table 1 and or Tables 2a-2e below. In other embodiments, the compound is one or more of the following:
  • the kinase inhibitor has the structure of Formula VIII:
  • ring A is a substituted or unsubstituted heteroaryl, such as an R 31 -substituted or unsubstituted heteroaryl.
  • R 1 and L 1 are as defined above.
  • R 31 is R 23A as defined above, or is hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 33 -substituted or unsubstituted alkyl, R 33 -substituted or unsubstituted heteroalkyl, R 33 -substituted or unsubstituted cycloalkyl, R 33 -substituted or unsubstituted heterocycloalkyl, R 33 -substituted or unsubstituted aryl, or R 33 -substituted or unsubstituted heteroaryl.
  • R 33 is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 34 -substituted or unsubstituted alkyl, R 34 -substituted or unsubstituted heteroalkyl, R 34 -substituted or unsubstituted cycloalkyl, R 34 -substituted or unsubstituted heterocycloalkyl, R 34 -substituted or unsubstituted aryl, or R 34 -substituted or unsubstituted heteroaryl.
  • R 34 is independently hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 35 -substituted or unsubstituted alkyl, R 35 -substituted or unsubstituted heteroalkyl, R 35 -substituted or unsubstituted cycloalkyl, R 35 -substituted or unsubstituted heterocycloalkyl, R 35 -substituted or unsubstituted aryl, or R 35 -substituted or unsubstituted heteroaryl.
  • R 35 is independently halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , unsubstituted alkyl, unsubstituted heteroalkyl, unsubstituted cycloalkyl, unsubstituted heterocycloalkyl, unsubstituted aryl, or unsubstituted heteroaryl.
  • the kinase inhibitor has the structure of Formula IX:
  • ring A is a substituted or unsubstituted heteroaryl, such as an R 31 -substituted or unsubstituted heteroaryl as set fort above.
  • R 1 and L 1 are as defined above.
  • ring A is R 31 -substituted or unsubstituted heteroaryl. In some embodiments of Formulae VIII and IX above, ring A is five-membered R 31 -substituted or unsubstituted heteroaryl or six-membered R 31 -substituted or unsubstituted heteroaryl.
  • the kinase inhibitor has the structure of Formula Xa:
  • X 1 is —C(R 31 R 32 )—, —N(R 31 )—, —S— or —O— when X 1 is not the point of attachment.
  • X 1 is C(R 31 ) or N.
  • X 2 , X 3 , X 4 and X 5 are independently —C(R 31 ) ⁇ or —N ⁇ when not the point of attachment.
  • X 2 , X 3 , X 4 or X 5 is the point of attachment, then the X 2 , X 3 , X 4 or X 5 that is the point of attachment is C.
  • At least one of X 1 , X 2 , X 3 , X 4 and X 5 is not carbon (i.e. not —C(R 31 R 32 )—, C(R 31 ) or —C(R 31 ) ⁇ as appropriate).
  • at least one of X 1 , X 2 , X 3 , X 4 and X 5 is nitrogen (i.e. —N(R 31 )— or —N ⁇ as appropriate).
  • at least 2 of X 1 , X 2 , X 3 , X 4 and X 5 is a carbon (e.g. —C(R 31 R 32 )—, C(R 31 ) or —C(R 31 ) ⁇ ).
  • R 31 is as defined above.
  • R 32 is hydrogen, halogen, —CN, —OH, —NH 2 , —COOH, —CF 3 , R 33 -substituted or unsubstituted alkyl, R 33 -substituted or unsubstituted heteroalkyl, R 33 -substituted or unsubstituted cycloalkyl, R 33 -substituted or unsubstituted heterocycloalkyl, R 33 -substituted or unsubstituted aryl, or R 33 -substituted or unsubstituted heteroaryl.
  • R 33 is defined as disclosed above.
  • the kinase inhibitor has the structure of Formula Xb:
  • X 1 is C(R 31 ) or N.
  • X 2 , X 3 , X 4 and X 5 are independently —C(R 31 ) ⁇ or —N ⁇ , provided, however, that at least one of X 1 , X 2 , X 3 , X 4 and X 5 is N.
  • at least 2 of X 1 , X 2 , X 3 , X 4 and X 5 is a carbon.
  • X 2 is C.
  • X 1 is —C(R 31 R 32 )—, —N(R 31 )—, —S— or —O—.
  • X 3 , X 4 and X 5 are independently —C(R 31 ) ⁇ or —N ⁇ , provided, however, that at least one of X 1 , X 3 , X 4 and X 5 is not carbon.
  • L 1 , R 1 and R 31 are defined as disclosed above.
  • at least one of X 1 , X 3 , X 4 and X 5 is carbon.
  • the kinase inhibitor has the structure of Formula Xc:
  • X 3 and X 5 are independently —C(R 31 ) ⁇ .
  • L 1 , R 1 and R 31 are as defined above.
  • the kinase inhibitor has the structure of Formula Xd:
  • X 3 , X 4 and X 5 are independently —C(R 31 ) ⁇ .
  • L 1 , R 1 and R 31 are defined as disclosed above.
  • the kinase inhibitor has the structure of Formula Xe:
  • X 4 and X 5 are independently —C(R 31 ) ⁇ .
  • X 2 is C.
  • L 1 , R 1 and R 31 are as defined as disclosed above.
  • the kinase inhibitor has the structure of Formula Xf:
  • X 4 and X 5 are independently —C(R 31 ) ⁇ .
  • L 1 , R 1 and R 31 are defined as disclosed above.
  • the kinase inhibitor has the structure of Formula XIa:
  • X 6 , X 7 , X 8 , X 9 and X 10 are independently —C(R 31 ) ⁇ , —N ⁇ , or +N—O—, provided, however, that at least one of X 6 , X 7 , X 8 , X 9 and X 10 is N or +N—O—.
  • L 1 , R 1 and R 31 are defined as disclosed above.
  • the kinase inhibitor has the structure of Formula XIb:
  • X 6 , X 7 , X 9 and X 10 are independently —C(R 31 ) ⁇ .
  • X 8 is N or +N—O—.
  • L 1 , R 1 and R 31 are defined as disclosed above.
  • the kinase inhibitor has the structure of Formula XIc:
  • X 6 , X 8 , X 9 and X 10 are independently —C(R 31 ) ⁇ .
  • X 7 is N or +N—O—.
  • L 1 , R 1 and R 31 are defined as disclosed above.
  • ring A is R 31 -substituted or unsubstituted furyl, R 31 -substituted or unsubstituted thienyl, R 31 -substituted or unsubstituted pyrrolyl, R 31 -substituted or unsubstituted imidazolyl, R 31 -substituted or unsubstituted pyrazolyl, R 31 -substituted or unsubstituted oxazolyl, R 31 -substituted or unsubstituted isoxazolyl, R 31 -substituted or unsubstituted thiazolyl, R 31 -substituted or unsubstituted isothiazolyl, R 31 -substituted or unsubstituted triazolyl, R 31 -substituted or unsubstituted ox
  • R 1 is R 7 -substituted or unsubstituted heterocycloalkyl, R 7 -substituted or unsubstituted aryl, or R 7 -substituted or unsubstituted heteroaryl.
  • L 1 is a bond.
  • a compound of Formulae I is one or more compounds set forth in Table 1, Tables 2a-2e below, or the following compounds:
  • a method of making a reversible kinase inhibitor includes the step of modifying a known non-reversible or irreversible kinase inhibitor to include a substituent having the formula:
  • W, L 2 , L 4 , L 5 , E, R 3 , R 4 , ring A, X 1 , X 2 , X 3 , X 4 , X 5 , X 6 , X 7 , X 8 , X 9 , X 10 , and z are as defined above.
  • the symbol represents the point of attachment of the substituent to the remained of the compound or solid support.
  • a protein adduct comprising a protein bound to a kinase inhibitor provided herein.
  • the adduct has the formula:
  • the symbols in the protein adduct formulae are as defined above.
  • the symbol Q represents a protein (e.g. peptide).
  • the sulfur attached to the Q typically forms part of a cysteine amino acid.
  • the cysteine linked to the inhibitor compounds is Cys-481 of BTK, Cys-909 of JAK3, or Cys-436 of RSK2.
  • methods of inhibiting protein kinases include contacting a protein kinase with an effective amount of a kinase inhibitor provided herein.
  • the kinase inhibitor may have the structure of the Formulae provided herein (or any of the embodiments thereof described above).
  • the methods of inhibiting a protein kinase are conducted within a cell.
  • methods of inhibiting a protein kinase within a cell are provided.
  • the method includes contacting a cell with an effective amount of a kinase inhibitor provided herein.
  • the kinase inhibitor may have the structure of the Formulae provided herein (or any of the embodiments thereof described above).
  • the cell is a prokaryote or eukaryote.
  • the cell may be a eukaryote (e.g. protozoan cell, fungal cell, plant cell or an animal cell).
  • the cell is a mammalian cell such as a human cell, cow cell, pig cell, horse cell, dog cell and cat cell, mouse cell, or rat cell.
  • the cell is a human cell.
  • the cell may form part of an organ or an organism. In certain embodiments, the cell does not form part of an organ or an organism.
  • the kinase inhibitor may be a reversible kinase inhibitor.
  • a reversible kinase inhibitor is a kinase inhibitor, as disclosed herein (e.g. the compounds of the Formulae provided herein and embodiments thereof), is capable of measurably dissociating from the protein kinase when the protein kinase is intact (i.e. not denatured) or denatured (e.g. partially denatured or fully denatured).
  • a “denatured” kinase is a kinase without sufficient tertiary or secondary structure sufficient to retain kinase activity.
  • an “intact” kinase is a kinase with sufficient tertiary or secondary structure sufficient to retain kinase activity. Therefore, in some embodiments, the method of inhibiting a protein kinase includes contacting a protein kinase with a reversible kinase inhibitor and allowing the reversible kinase inhibitor to reversibly bind to an active site cysteine residue, thereby inhibiting the protein kinase.
  • the reversible kinase inhibitor measurably dissociates from the protein kinase only when the protein kinase is denatured, but does not measurably dissociate from the protein kinase when the protein kinase is intact (or dissociates at least 1, 1 ⁇ 10 2 , 1 ⁇ 10 3 , 1 ⁇ 10 4 , 1 ⁇ 10 5 , 1 ⁇ 10 6 , 1 ⁇ 10 7 , 1 ⁇ 10 8 , 1 ⁇ 10 9 , 1 ⁇ 10 10 fold slower relative to the dissociation when the protein kinase is denatured).
  • a reversible kinase inhibitor that measurably dissociates (or dissociates at least 1, 1 ⁇ 10 2 , 1 ⁇ 10 3 , 1 ⁇ 10 4 , 1 ⁇ 10 5 , 1 ⁇ 10 6 , 1 ⁇ 10, 1 ⁇ 10 8 , 1 ⁇ 10 9 , 1 ⁇ 10 10 fold slower relative to the dissociation when the protein kinase is denatured) from the protein kinase only when the protein kinase is denatured, but does not measurably dissociate from the protein kinase when the protein kinase is intact is referred to herein as a “reversible denatured kinase inhibitor.” After dissociating from the kinase, the reversible denatured kinase inhibitor can bind to the same or another kinase.
  • the method of inhibiting the protein kinase includes contacting the protein kinase with a kinase inhibitor wherein the kinase inhibitor inhibits the protein kinase with an inhibition constant of less than 100 nM.
  • the method of inhibiting the protein kinase includes contacting the protein kinase with a reversible kinase inhibitor wherein the reversible kinase inhibitor inhibits the protein kinase with an inhibition constant of less than 100 nM.
  • kinase activity i.e. phosphorylation of a substrate molecule (e.g. a protein substrate)
  • the kinase inhibitor decreases the kinase activity 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 25, 50, 100, 250, 500, 1000, 5000, 10,000, 100,000, 500,000, 1,000,000, or more fold.
  • the kinase inhibitor inhibits the activity of the kinase with an inhibition constant (KO of less than 100 ⁇ M, 5 ⁇ M, 1 ⁇ M, 500 nM, 250 nM, 100 nM, 75 nM, 50 nM, 25 nM, 10 nM, 1 nM, 500 pM, 250 pM, 100 pM, 75 pM, 50 pM, 25 pM, 10 pM, or 1 pM.
  • an inhibition constant K of less than 100 ⁇ M, 5 ⁇ M, 1 ⁇ M, 500 nM, 250 nM, 100 nM, 75 nM, 50 nM, 25 nM, 10 nM, 1 nM.
  • the kinase inhibitor inhibits the activity of the kinase with an IC 50 of less than 100 ⁇ M, 10 ⁇ M, 5 ⁇ M, 1 ⁇ M, 500 nM, 250 nM, 100 nM, 75 nM, 50 nM, 25 nM, 10 nM, 1 nM, 500 pM, 250 pM, 100 pM, 75 pM, 50 pM, 25 pM, 10 pM, or 1 pM, when measured under the conditions set forth in the examples section.
  • a reversible kinase inhibitor reversibly binds to an active site cysteine residue
  • a reversible bond is formed between the active site cysteine residue and the reversible kinase inhibitor.
  • the reversible bond is typically a covalent bond.
  • a “covalent reversible kinase inhibitor,” as used herein, refers to a reversible kinase inhibitor that forms a covalent bond with the kinase.
  • the covalent reversible kinase inhibitor forms a reversible bond with an active site cysteine residue
  • the covalent reversible kinase inhibitor is referred to herein as a “thiol covalent reversible kinase inhibitor.”
  • the covalent reversible kinase inhibitor measurably dissociates from the protein kinase only when the protein kinase is denatured, but does not measurably dissociate (or dissociates at least 1, 1 ⁇ 10 2 , 1 ⁇ 10 3 , 1 ⁇ 10 4 , 1 ⁇ 10 5 , 1 ⁇ 10 6 , 1 ⁇ 10 7 , 1 ⁇ 10 8 , 1 ⁇ 10 9 , 1 ⁇ 10 10 fold slower relative to the dissociation when the protein kinas is fully or partially denatured) from the protein kinase when the protein kinase is intact (referred to herein as a “covalent reversible denatured kinase inhibitor”).
  • the protein kinase is denatured (i.e. not intact) when placed in denaturing solution, such as 6 N guanidine, 1% SDS, 50% MeCN, or similar protein denaturant, for second or minutes (e.g. 30 to 120 seconds, such as 60 seconds).
  • denaturing solution such as 6 N guanidine, 1% SDS, 50% MeCN, or similar protein denaturant
  • second or minutes e.g. 30 to 120 seconds, such as 60 seconds.
  • a covalent reversible denatured kinase inhibitor that forms a reversible bond with an active site cysteine residue is termed a “thiol covalent reversible denatured kinase inhibitor.”
  • the thiol covalent reversible kinase inhibitor forms a bond between the cysteine sulfhydryl groups and a carbon atom forming part of the carbon-carbon double bond (i.e. olefin) of the compound of the Formulae provided herein.
  • olefin carbon-carbon double bond
  • electrons of the sulfur atom of the active site cysteine sulfhydryl group attacks an electron deficient carbon atom of the carbon-carbon double bond (olefin).
  • the electron deficient carbon atom of the carbon-carbon double bond is distal to the electron withdrawing cyano group and the electron withdrawing -L 2 -E substituent of the kinase inhibitor of the Formulae provided herein and embodiments thereof (i.e. the carbon attached to -L 1 -R 1 ).
  • a thiol adduct is formed (e.g., Michael reaction with cysteine). Therefore, in some embodiments, the combination of cyano and -L 2 -E electron withdrawing groups bound to the olefinic moiety increases the reactivity of the olefin to form a thiol adduct with the active site cysteine residue.
  • the resulting thiol adduct is stable at about pH 2 to about pH 7 (e.g. about pH 3).
  • the reversible kinase inhibitors described herein, after covalently binding to the kinase active site cysteine residue as described herein is capable of dissociating from the kinase within seconds or minutes after denaturing/unfolding the kinase with 6 N guanidine, 1% SDS, 50% MeCN, or similar protein denaturant.
  • the cyano and -L 2 -E electron withdrawing groups function to increase the reversibility of thiol adduct that is formed.
  • the reversible kinase inhibitors set forth herein is completely reversible.
  • the term “completely reversible” means the reversible kinase inhibitor exhibits a measurable dissociation rate under conditions in which the kinase is not denatured.
  • the kinase inhibitors provided herein are not completely reversible (i.e. do not exhibit a measurable dissociation rate under conditions in which the kinase is intact). Dissociation may be measured using any appropriate means, including dialysis and mass spectrometry. Specific methods of measuring dissociation are set forth in the Examples section below.
  • the reversible denatured kinase inhibitor binds reversibly to cellular components other than the protein kinase that the reversible denatured kinase inhibitor inhibits (or specifically inhibits).
  • the cellular components may be GSH, proteins or protein fragments that are not targeted kinases (e.g. a kinase that does not include an active site cysteine or does not include an active site cysteine within sufficient proximity to an ATP binding site), protein fragments of targeted kinases (e.g.
  • the reversible denatured kinase inhibitor measurably dissociates from the kinase where the kinase is partly or fully digested.
  • a reversible denatured kinase inhibitor to measurably dissociate from cellular components other than the intact of full length protein kinase that the reversible denatured kinase inhibitor inhibits may provide decreased toxicity, including decreased immunogenic toxicity.
  • the -L 1 -R 1 group of the reversible denatured kinase inhibitor is a kinase ATP binding site moiety and the electron deficient olefin carbon binds to a sulfhydryl of a kinase active site cysteine.
  • the kinase inhibitors provided herein bind to at least two points of the protein kinase: at least one residue within the ATP binding site moiety and a sulfhydryl of a kinase active site cysteine.
  • physiological concentrations of glutathione e.g. 5 or 10 mM GSH
  • glutathione e.g. 5 or 10 mM GSH
  • the reversible kinase inhibitors e.g. provided herein to inhibit a protein kinase (e.g. eversible denatured kinase inhibitor only reversibly binds to GSH thereby enabling increased binding to the target kinase).
  • the IC50 or K i of the reversible kinase inhibitor is increased no more than 20%, 15%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.1%, 0.01% or 0.001% in the presence of physiological concentrations of glutathione(e.g. 5 or 10 mM GSH).
  • the IC50 or K i of the reversible kinase inhibitor is not measurably increased by the presence of physiological concentrations of glutathione (e.g. 5 or 10 mM GSH).
  • the physiological concentrations of glutathione e.g.
  • 5 or 10 mM GSH have little or no measurable effect on the ability of the reversible kinase inhibitors provided herein to inhibit a protein kinase wherein the reversible kinase inhibitor is present at low concentrations (e.g. less than 100 nM, 75 nM, 50 nM, 25 nM, 10 nM, 5 nM, 3 nM, 1 nM, 500 pM, 250 pM, 100 pM, 75 pM, 50 pM, 25 pM, 10 pM, or 1 pM).
  • the physiological concentrations of glutathione e.g.
  • 5 or 10 mM GSH have little or no measurable effect on the ability of the reversible kinase inhibitors provided herein to inhibit a protein kinase wherein the reversible kinase inhibitor is present at a concentration of less than 10 nM, 5 nM, 4 nM 3 nM, 2 nM or 1 nM.
  • the physiological concentrations of glutathione e.g. 5 or 10 mM GSH
  • physiological concentrations of adenosine triphosphate have little or no measurable effect on the ability of the reversible kinase inhibitors provided herein to inhibit a protein kinase.
  • the IC50 or K i of the reversible kinase inhibitor is increased no more than 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.1%, 0.01% or 0.001% in the presence of physiological concentrations of adenosine triphosphate (e.g. 1 mM ATP).
  • the IC50 or K 1 of the reversible kinase inhibitor is not measurably increased by the presence of physiological concentrations of adenosine triphosphate (e.g. 1 mM ATP).
  • the reversible kinase inhibitors provided herein reacts reversibly with GSH. In certain embodiments, the reversible kinase inhibitors provided herein react rapidly and reversibly with GSH. Thus, in certain embodiments, the reversible kinase inhibitors provided herein react reversibly with GSH (e.g. rapidly and reversibly) while also reversibly binding to an active site cysteine residue (e.g. at a concentration of less than 10 nM, 5 nM, 4 nM 3 nM, 2 nM or 1 nM).
  • the GSH may be at physiological concentration (e.g. 5-10 mM).
  • the protein kinase may be any appropriate kinase.
  • the protein kinase includes a cysteine residue in the active site.
  • a protein kinase active site is a portion of the protein kinase in which the protein kinase substrate is phosphorylated.
  • the kinase active site is typically a pocket or cleft containing amino acid residues that bind to a substrate (also referred to herein as kinase active site binding residues) and amino acid residues that participate in the catalytic phosphorylation reaction (also referred to herein as kinase active site catalytic residues).
  • the reversible kinase inhibitors provided herein are capable of inhibiting the kinase catalytic action by fitting into the kinase active site and disrupting the ability of the kinase to phosphorylate the substrate.
  • the active sites of many protein kinases are known in the art through structure determinations (e.g. X-ray crystallography or three dimensional NMR techniques). Where the three dimensional structure has not been determined, the structure of an active site of a protein kinase may be determined by the primary amino acid sequence using computer software modeling programs generally known in the art.
  • Protein kinases inhibited using the kinase inhibitors provided herein include, but are not limited to, serine/threonine-specific protein kinases, tyrosine-specific protein kinases, receptor tyrosine kinases, receptor-associated tyrosine kinases, histidine-specific protein kinases, and aspartic acid/glutamic acid-specific protein kinases, as known in the art.
  • the kinase is a tyrosine protein kinase or serine/threonine protein kinases.
  • kinases examples include SRC, YES, FGR, CHK2, FGFR1—4, BTK, EGFR, HER2, HER4, HER3, JAK3, PLK1-3, MPSI, RON, MEK1/2, ERK1/2, VEGFR, KIT, KDR, PDGFR, FLT3, CDK8, MEK7, ROR1, RSK1-4, MSK1/2, MEKK1, NEK2, MEK5, MNK1/2, MEK4, TGFbR2, ZAP70, WNK1-4, BMX, TEC, TXK, ITK, BLK, MK2/3, LIMK1, TNK1, CDK11, p70S6 Kb, EphB3, ZAK, and NOK.
  • a method of treating a disease associated with kinase activity in a subject in need of such treatment includes administering to the subject an effective amount (e.g. a therapeutically effective amount) of a compound having the structure of the Formulae provided herein (or an embodiment thereof as described above).
  • an effective amount e.g. a therapeutically effective amount
  • the disease associated with kinase activity is chronic disease.
  • the disease may be cancer, epilepsy, HIV infection, autoimmune disease (e.g. arthritis), ischemic disease (e.g. heart attack or stroke), stroke, neurodegenerative diseases, metabolic or inflammation.
  • the disease is cancer , including, for example, leukemia, carcinomas and sarcomas, such as cancer of the brain, breast, cervix, colon, pancreas, head & neck, liver, kidney, lung, non-small cell lung, prostate, melanoma, mesothelioma, ovary, sarcoma, stomach, uterus and medulloblastoma.
  • Additional examples include, Hodgkin's Disease, Non-Hodgkin's Lymphoma, multiple myeloma, neuroblastoma, ovarian cancer, rhabdomyosarcoma, primary thrombocytosis, primary macroglobulinemia, primary brain tumors, malignant pancreatic insulanoma, malignant carcinoid, urinary bladder cancer, premalignant skin lesions, testicular cancer, lymphomas, thyroid cancer, neuroblastoma, esophageal cancer, genitourinary tract cancer, malignant hypercalcemia, endometrial cancer, adrenal cortical cancer, neoplasms of the endocrine and exocrine pancreas.
  • the disease is liver cancer, colon cancer, breast cancer, melanoma, acute myelogenous leukemia, chronic myelogenous leukemia, or nonsmall-cell lung cancer.
  • the disease is cancers which have metastasized.
  • the disease is rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, scleroderma or polymyositis.
  • the disease is diabetes, obesity, or lipid disorders.
  • the disease may be caused by an infectious agent such as caused by bacteria, parasite or virus.
  • the disease is acute such as myocardial infarction, stroke or asthma.
  • the disease is Parkinson's disease or amyotrophic lateral sclerosis.
  • candidate kinase inhibitors may be easily assayed for their ability to inhibit any known protein kinase.
  • candidate kinase inhibitors having the structure of the Formulae provided herein or embodiments thereof may be first assayed using computer modeling techniques in order to assess potential binding contacts between kinase active site binding residues and/or kinase active site catalytic residues.
  • Such computer modeling techniques may also be referred to as in silico techniques.
  • the kinase active site binding residues and/or kinase active site catalytic residues are known or easily determined for any kinase in which the primary amino acid structure is known.
  • kinase inhibitors may react with a kinase active site cysteine residue with the electron deficient olefin carbon to form a thiol adduct.
  • the kinase inhibitor electron deficient olefin carbon is within 10 ⁇ of the kinase active site cysteine sulfhydryl, the potency and/or selectivity of kinase inhibitor may be improved (e.g. by 1000-10,000-fold).
  • computer modeling techniques may be used to assess the ability of candidate kinase inhibitors to fit into the kinase active site without creating stearic clashes.
  • —R 1 or -L-R 1 to fit within the kinase ATP binding site and/or make contacts with amino acid residues within the kinase ATP binding site. Therefore, computer modeling techniques may be used to assess the ability of —R 1 or -L-R 1 to fit within the kinase ATP binding site and/or make contacts with amino acid residues within the kinase ATP binding site.
  • the computer modeling assays described above may be used to assess the kinase inhibition ability of candidate kinase inhibitors having different general chemical scaffolds within the structure of the Formulae provided herein or embodiments thereof. In this way, new classes of chemical scaffolds may be assessed using computer modeling prior to performing in vitro activity assays.
  • In vitro assays may also be used to assess the kinase inhibiting properties of candidate kinase inhibitors having the structure of the Formulae provided herein or embodiments thereof.
  • In vitro kinase assays are well known in the art. High throughput techniques are known and useful for quickly assessing large numbers of kinase inhibitor candidates using binding assays for a large number of kinase panels. See, for example, Karaman et al., Nat. Biotechnol. 2008 January; 26(1):127-32.
  • Protein kinases can be found in native cells, isolated in vitro, or co-expressed or expressed in a cell. Certain protein kinases specifically phosphorylate particular substrates. Where specific substrates are known, the ability of a candidate kinase inhibitor to reduce phosphorylation of the specific substrate may be assayed. General, or non-specific, kinase substrates may also be employed.
  • the kinase inhibitors provided herein may also be tested in vitro for their ability to inhibit a mutant of a kinase that does not contain an active site cysteine.
  • the ability of a kinase inhibitor to decrease the catalytic activity of a kinase having an active site cysteine while not having the ability (or having measurably decreased ability) to decrease the catalytic activity of a mutant of the kinase that does not contain an active site cysteine is indicative of a kinase inhibitor that inhibits the kinase by binding to the active cite cysteine.
  • the C436V mutant of RSK2 may be resistant to certain kinase inhibitors (IC50>10 uM) that show strong inhibitory activity against the wild type RSK2. This result supports the conclusion that RSK2 inhibition requires the formation of a covalent bond between Cys436 and the inhibitor.
  • E and -L 2 -E are typically substituents that sufficiently withdraw electrons from the reaction center olefin carbon to reversibly bind to the sulfhydryl of a kinase active cite cysteine (e.g. when the kinase is partly or fully denatured).
  • the kinase inhibitors provided herein may also be tested in vitro for their ability to reversibly bind to the active site cysteine of a protein kinase by measuring association and dissociation of the kinase inhibitor from the protein kinase (e.g. partially or fully denatures) or from a thiol compound (e.g. 2-mercaptoethanol (BME)).
  • the ability of the reaction center carbon of a kinase inhibitor provided herein to reversibly bind to the sulfhydryl of a kinase active cite cysteine may be measured using any appropriate means, including dialysis, mass spectrometry, NMR and UV detection (see Examples section for more details).
  • the kinase inhibitors may be assayed by detecting the binding of a thiol compound such as BME.
  • the binding may be assessed using UV detection of compounds that typically become less UV active upon binding to a thiol compound or by detecting the binding using proton NMR.
  • the assays are conducted by titering in the thiol compound and examining a change in the endpoint binding detection parameter (e.g. UV activity or proton NMR). Reversibility is assessed by dilution. Specific examples are provided below in the Examples section (see Example 82).
  • the kinase inhibitors provided herein may also be tested in vitro for their stability at pH 7.5. Any appropriate method may be used to determine the stability of a kinase inhibitor set forth herein at pH 7.5. Appropriate methods include, for example, LC-MS (e.g. HPLC-MS) as well as measuring changes in UV absorption where the kinase inhibitor includes a chromophore group. UV absorption may be measured using high-throughput techniques (e.g. multiwell plated for scanning large numbers of kinase inhibitors simultaneously). Stability may be assessed using phosphate-buffered saline at pH 7.5 at 37° C. Compounds having half-lives greater than 6 hours, 12 hours, 24 hours, or 48 hours may be may be selected.
  • LC-MS e.g. HPLC-MS
  • UV absorption may be measured using high-throughput techniques (e.g. multiwell plated for scanning large numbers of kinase inhibitors simultaneously). Stability may be assessed using
  • Cellular assays may also be used to assess the kinase inhibiting properties of candidate kinase inhibitors having the structure of the Formulae provided herein or embodiments thereof.
  • Cellular assays include cells from any appropriate source, including plant and animal cells (such as mammalian cells). The cellular assays may also be conducted in human cells.
  • Cellular assays of kinase inhibition are well known in the art, and include methods in which a kinase inhibitor is delivered into the cell (e.g.
  • a kinase activity endpoint is measured, such as the amount of phosphorylation of a cellular substrate, the amount of expression of a cellular protein, or some other change in the cellular phenotype known to be affected by the catalytic activity of the particular kinase being measured.
  • phosphorylation of a particular cellular substrate may be assessed using a detection antibody specific or the phosphorylated cellular substrate followed by western blotting techniques and visualization using any appropriate means (e.g. fluorescent detection of a fluorescently labeled antibody).
  • Measuring the reduction in the protein kinase catalytic activity in the presence of a kinase inhibitor disclosed herein relative to the activity in the absence of the inhibitor may be performed using a variety of methods known in the art, such as the assays described in the Examples section below. Other methods for assaying the activity of kinase activity are known in the art. The selection of appropriate assay methods is well within the capabilities of those having ordinary skill in the art.
  • kinase inhibitors are identified that are capable of reducing kinase catalytic activity in vitro and/or in a cell
  • the compounds may be further tested for their ability to selectively inhibit kinase activity in animal models (e.g. whole animals or animal organs).
  • animal models e.g. whole animals or animal organs.
  • kinase inhibitors may be further tested in cell models or animal models for their ability to cause detectable changes in phenotype related to a particular kinase activity.
  • animal models may be used to test inhibitors of kinases for their ability to treat, for example, cancer in an animal model.
  • the present invention provides pharmaceutical compositions comprising a kinase inhibitor compound of the invention or a kinase inhibitor compound in combination with a pharmaceutically acceptable excipient (e.g. carrier).
  • a pharmaceutically acceptable excipient e.g. carrier
  • the pharmaceutical compositions include optical isomers, diastereomers, or pharmaceutically acceptable salts of the inhibitors disclosed herein.
  • the pharmaceutical compositions include a compound of the present invention and citrate as a pharmaceutically acceptable salt.
  • the kinase inhibitor included in the pharmaceutical composition may be covalently attached to a carrier moiety, as described above. Alternatively, the kinase inhibitor included in the pharmaceutical composition is not covalently linked to a carrier moiety.
  • a “pharmaceutically suitable carrier,” as used herein refers to pharmaceutical excipients, for example, pharmaceutically, physiologically, acceptable organic, or inorganic carrier substances suitable for enteral or parenteral application which do not deleteriously react with the extract.
  • suitable pharmaceutically acceptable carriers include water, salt solutions (such as Ringer's solution), alcohols, oils, gelatins and carbohydrates such as lactose, amylose or starch, fatty acid esters, hydroxymethycellulose, and polyvinyl pyrrolidine.
  • Such preparations can be sterilized and, if desired, mixed with auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like which do not deleteriously react with the compounds of the invention.
  • auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like which do not deleteriously react with the compounds of the invention.
  • the compounds of the invention can be administered alone or can be coadministered to the patient. Coadministration is meant to include simultaneous or sequential administration of the compounds individually or in combination (more than one compound).
  • the preparations can also be combined, when desired, with other active substances (e.g. to reduce metabolic degradation).
  • the kinase inhibitors of the present invention can be prepared and administered in a wide variety of oral, parenteral and topical dosage forms.
  • the compounds of the present invention can be administered by injection (e.g. intravenously, intramuscularly, intracutaneously, subcutaneously, intraduodenally, or intraperitoneally).
  • the compounds described herein can be administered by inhalation, for example, intranasally.
  • the compounds of the present invention can be administered transdermally. It is also envisioned that multiple routes of administration (e.g., intramuscular, oral, transdermal) can be used to administer the compounds of the invention.
  • the present invention also provides pharmaceutical compositions comprising a pharmaceutically acceptable carrier or excipient and one or more compounds of the invention.
  • pharmaceutically acceptable carriers can be either solid or liquid.
  • Solid form preparations include powders, tablets, pills, capsules, cachets, suppositories, and dispersible granules.
  • a solid carrier can be one or more substance, which may also act as diluents, flavoring agents, binders, preservatives, tablet disintegrating agents, or an encapsulating material.
  • the carrier is a finely divided solid, which is in a mixture with the finely divided active component.
  • the active component is mixed with the carrier having the necessary binding properties in suitable proportions and compacted in the shape and size desired.
  • the powders and tablets preferably contain from 5% to 70% of the active compound.
  • Suitable carriers are magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, a low melting wax, cocoa butter, and the like.
  • the term “preparation” is intended to include the formulation of the active compound with encapsulating material as a carrier providing a capsule in which the active component with or without other carriers, is surrounded by a carrier, which is thus in association with it.
  • cachets and lozenges are included. Tablets, powders, capsules, pills, cachets, and lozenges can be used as solid dosage forms suitable for oral administration.
  • a low melting wax such as a mixture of fatty acid glycerides or cocoa butter
  • the active component is dispersed homogeneously therein, as by stirring.
  • the molten homogeneous mixture is then poured into convenient sized molds, allowed to cool, and thereby to solidify.
  • Liquid form preparations include solutions, suspensions, and emulsions, for example, water or water/propylene glycol solutions.
  • liquid preparations can be formulated in solution in aqueous polyethylene glycol solution.
  • admixtures for the compounds of the invention are injectable, sterile solutions, preferably oily or aqueous solutions, as well as suspensions, emulsions, or implants, including suppositories.
  • carriers for parenteral administration include aqueous solutions of dextrose, saline, pure water, ethanol, glycerol, propylene glycol, peanut oil, sesame oil, polyoxyethylene-block polymers, and the like.
  • Ampules are convenient unit dosages.
  • the compounds of the invention can also be incorporated into liposomes or administered via transdermal pumps or patches.
  • compositions suitable for use in the present invention include those described, for example, in Pharmaceutical Sciences (17th Ed., Mack Pub. Co., Easton, Pa.) and WO 96/05309, the teachings of both of which are hereby incorporated by reference.
  • Aqueous solutions suitable for oral use can be prepared by dissolving the active component in water and adding suitable colorants, flavors, stabilizers, and thickening agents as desired.
  • Aqueous suspensions suitable for oral use can be made by dispersing the finely divided active component in water with viscous material, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, and other well-known suspending agents.
  • solid form preparations which are intended to be converted, shortly before use, to liquid form preparations for oral administration.
  • liquid forms include solutions, suspensions, and emulsions.
  • These preparations may contain, in addition to the active component, colorants, flavors, stabilizers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilizing agents, and the like.
  • the pharmaceutical preparation is preferably in unit dosage form.
  • the preparation is subdivided into unit doses containing appropriate quantities of the active component.
  • the unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, such as packeted tablets, capsules, and powders in vials or ampoules.
  • the unit dosage form can be a capsule, tablet, cachet, or lozenge itself, or it can be the appropriate number of any of these in packaged form.
  • the quantity of active component in a unit dose preparation may be varied or adjusted from 0.1 mg to 10000 mg, more typically 1.0 mg to 1000 mg, most typically 10 mg to 500 mg, according to the particular application and the potency of the active component.
  • the composition can, if desired, also contain other compatible therapeutic agents.
  • co-solvents include: Polysorbate 20, 60 and 80; Pluronic F-68, F-84 and P-103; cyclodextrin; and polyoxyl 35 castor oil. Such co-solvents are typically employed at a level between about 0.01% and about 2% by weight.
  • Viscosity greater than that of simple aqueous solutions may be desirable to decrease variability in dispensing the formulations, to decrease physical separation of components of a suspension or emulsion of formulation and/or otherwise to improve the formulation.
  • Such viscosity building agents include, for example, polyvinyl alcohol, polyvinyl pyrrolidone, methyl cellulose, hydroxy propyl methylcellulose, hydroxyethyl cellulose, carboxymethyl cellulose, hydroxy propyl cellulose, chondroitin sulfate and salts thereof, hyaluronic acid and salts thereof, combinations of the foregoing.
  • Such agents are typically employed at a level between about 0.01% and about 2% by weight.
  • compositions of the present invention may additionally include components to provide sustained release and/or comfort.
  • Such components include high molecular weight, anionic mucomimetic polymers, gelling polysaccharides and finely-divided drug carrier substrates. These components are discussed in greater detail in U.S. Pat. Nos. 4,911,920; 5,403,841; 5,212,162; and 4,861,760. The entire contents of these patents are incorporated herein by reference in their entirety for all purposes.
  • compositions provided by the present invention include compositions wherein the active ingredient is contained in a therapeutically effective amount, i.e., in an amount effective to achieve its intended purpose.
  • a therapeutically effective amount i.e., in an amount effective to achieve its intended purpose.
  • the actual amount effective for a particular application will depend, inter alia, on the condition being treated.
  • such compositions when administered in methods to treat cancer, such compositions will contain an amount of active ingredient effective to achieve the desired result (e.g. decreasing the number of cancer cells in a subject).
  • the dosage and frequency (single or multiple doses) of administered to a mammal can vary depending upon a variety of factors, including a disease that results in increased activity of kinase, whether the mammal suffers from another disease, and the route of administration; size, age, sex, health, body weight, body mass index, and diet of the recipient; nature and extent of symptoms of the disease being treated (e.g., cancer), type of concurrent treatment, complications from the disease being treated or other health-related problems.
  • Other therapeutic regimens or agents can be used in conjunction with the methods and compounds of the invention.
  • the therapeutically effective amount can be initially determined from cell culture assays.
  • Target concentrations will be those concentrations of active compound(s) that are capable of reducing the level of kinase catalytic activity, as measured, for example, using the methods described.
  • Therapeutically effective amounts for use in humans may be determined from animal models. For example, a dose for humans can be formulated to achieve a concentration that has been found to be effective in animals. The dosage in humans can be adjusted by monitoring kinase inhibition and adjusting the dosage upwards or downwards, as described above.
  • Dosages may be varied depending upon the requirements of the patient and the compound being employed.
  • the dose administered to a patient should be sufficient to affect a beneficial therapeutic response in the patient over time.
  • the size of the dose also will be determined by the existence, nature, and extent of any adverse side-effects.
  • treatment is initiated with smaller dosages which are less than the optimum dose of the compound. Thereafter, the dosage is increased by small increments until the optimum effect under circumstances is reached.
  • the dosage range is 0.001% to 10% w/v. In another embodiment, the dosage range is 0.1% to 5% w/v.
  • Dosage amounts and intervals can be adjusted individually to provide levels of the administered compound effective for the particular clinical indication being treated. This will provide a therapeutic regimen that is commensurate with the severity of the individual's disease state.
  • an effective prophylactic or therapeutic treatment regimen can be planned which does not cause substantial toxicity and yet is entirely effective to treat the clinical symptoms demonstrated by the particular patient.
  • This planning should involve the careful choice of active compound by considering factors such as compound potency, relative bioavailability, patient body weight, presence and severity of adverse side effects, preferred mode of administration and the toxicity profile of the selected agent.
  • the kinase inhibitors provided herein may be used in combination with other therapeutic modalities such as radiation therapy and surgery.
  • Dichloromethane, dimethylformamide, methanol, tetrahydrofuran, toluene, and diisopropylamine were dried using the solvent purification system manufactured by Glass Contour, Inc. (Laguna Beach, Calif.). All other solvents were of ACS chemical grade (Fisher) and used without further purification unless otherwise indicated. Analytical and preparative thin layer chromatography were performed with silica gel 60 F 254 glass plates (EM Science). Flash chromatography was conducted with 230-400 mesh silica gel (Selecto Scientific).
  • HPLC high performance liquid chromatography
  • Active JAK2 Human, residues 808-end
  • JAK3 Human, residues 781-end
  • Active kinase (3 nM) in 8 mM HEPES, pH 7.0, 200 uM EDTA, 10 mM MgCl 2 , 0.2 mg/mL BSA, 100 uM ATP and 10 mM GSH were pre-incubated with inhibitors (eight or ten concentrations, in duplicate) for 30 minutes at room temperature.
  • Kinase reactions were initiated by the addition of 0.3 ⁇ Ci/ ⁇ L of ⁇ - 32 P-ATP (6000 Ci/mmol, NEN) and 100 uM peptide substrate (JAK3-tide for JAK3 or PDK-tide for JAK2, Millipore) and incubated for 30 minutes at room temperature. Kinase activity was determined by spotting 5 ⁇ L of each reaction onto sheets of phosphocellulose. Each blot was washed once with 1% AcOH solution, twice with 0.1% H 3 PO 4 solution, and once with MeOH (5-10 minutes per wash). Dried blots were exposed for 30 minutes to a storage phosphor screen and scanned by a Typhoon imager (GE Life Sciences). The data were quantified using the SPOT program (Knight, Z. et al. Nature Protocols, 2 (10), 2459-66) and plotted using GraphPad Prism 4.0 software.
  • Btk human, full length was purchased (Invitrogen, catalog number PV3363) and used as specified in the product literature. Btk (2 nM final concentration) was pre-incubated with inhibitors (six or ten concentrations, in duplicate) for 30 minutes at room temperature. Kinase reactions were initiated by the addition of 0.16 ⁇ Ci/ ⁇ L of ⁇ - 32 P-ATP (6000 Ci/mmol, NEN) and 0.2 mg/mL substrate (poly[Glu:Tyr], 4:1 Glu:Tyr) and incubated for 30 minutes at room temperature. Kinase activity was determined by spotting 6 ⁇ L of each reaction onto sheets of phosphocellulose.
  • 6-Chloropurine (509 mg, 3.2 mmol), 4-piperidine methanol (737 mg, 6.4 mmol) and Et 3 N (2.25 mL, 25.6 mmol) was dissolved in n-BuOH (30 mL) and heated to 100° C. in a sealed vessel. The light orange solution was heated overnight, cooled to room temperature, and concentrated to afford a tan solid. The residue was triturated with 3:1 hexanes:MeOH (10 mL) and dried in vacuo to afford 401 mg (57%) of (1-(9H-purin-6-yl)piperidin-4-yl)methanol as a bright white solid.
  • 1-(9H-purin-6-yl)piperidine-4-carbaldehyde (11.2 mg, 0.048 mmol), 2-cyanoacetamide (6 mg, 0.071 mmol) and PPh 3 (6 mg, 0.023 mmol) were combined in THF (1 mL) and heated to 80° C. in a sealed vial. After 18 hours of heating, a white precipitate had formed but TLC indicated the presence of significant quantities of starting material. A spatula tip of 2-cyanoacetamide was added to the reaction and it was stirred for an additional 18 hours at 80° C.
  • N′-(3-cyano-4-p-tolyl-1H-pyrrol-2-yl)formimidamide (2.27 g, 10.2 mmol) was dissolved in MeOH (30 ml) and heated to reflux. Sodium methoxide (1.5 m, 25% wt in MeOH, 5.1 mmol) was added and the reaction was stirred at reflux for 3 hours after which the product precipitated out of solution. After cooling to room temperature, the solid was isolated by filtration and dried under high vacuum to give 2.3 g (100%) of pure 5-p-tolyl-7H-pyrrolo[2,3-d]pyrimidin-4-amine as a tan solid.
  • reaction was concentrated and the residue was purified by flash column chromatography in 50% EtOAc/hexanes to give a yellow powder that was dissolved in THF (1 ml) and 100 ⁇ l 1 N HCl was added.
  • the reaction was stirred at room temperature for 1 hour, diluted with ethyl acetate, and the layers were separated. The organic layer was washed with saturated aqueous sodium bicarbonate followed by brine and then dried over sodium sulfate, filtered, and concentrated.
  • 1-(9H-purin-6-yl)piperidine-4-carbaldehyde (17 mg, 0.086 mmol), 2-cyano-methylacetamide (11 mg, 0.112 mmol) and PPh 3 (28 mg, 0.106 mmol) were combined in THF (1 mL) and heated to 110 ⁇ C in a sealed vial. After 18 hours of heating, the reaction mixture was diluted with EtOAc (2 mL) and washed with water (1 mL).
  • This oil was dissolved in CH 2 Cl 2 (3 mL) and TFA (1.5 mL) was added. After 16 hours at 20-25° C., the reaction mixture was concentrated and the residue was redissolved in THF (6 mL) and 1M aq. HCl (2 mL) was added. The reaction mixture was maintained at ambient temperature for 8 hours, then quenched with satd. aq. NaHCO3 (20 mL) and brine (30 mL) and extracted with EtOAc (3 ⁇ 50 mL).
  • the solution was concentrated under reduced pressure and redissolved in methanol (10 mL).
  • the solution was heated to 80° C. and sodium methoxide (25% w/v in methanol, 1 mL) was added dropwise.
  • the mixture was refluxed for 2 h, cooled to room temperature and quenched by the addition of water.
  • the solution was extracted with ethyl acetate (3 ⁇ 50 mL).
  • the organic layer was dried over sodium sulfate, filtered, and concentrated under reduced pressure.
  • the resulting oil was purified by Si-gel chromatography (elute 1:1 EtOAc/Hex to 100% EtOAc). Intermediate product containing fractions were combined and concentrated under reduced pressure.
  • the organic layer was washed with 5% NaHCO 3 and then dried over sodium sulfate, filtered, and concentrated under reduced pressure.
  • the resulting residue was purified by Si-gel chromatography (elute 20% to 40% EtOAc/Hex).
  • the resulting oil was concentrated under reduced pressure and redissolved in ammonia/methanol solution (7 M, 10 mL) and the flask was tightly capped and stirred at room temperature over night.
  • the reaction mixture was concentrated under reduced pressure and redissolved ammonia/methanol solution (7 M, 10 mL), the flask tightly capped and stirred at room temperature for an additional 2 h.
  • RSK2 CTD and His6-ERK2 were expressed and purified as described (Cohen et al., Science, 308: 1318).
  • the C436V mutant of RSK2 CTD was generated by Quikchange mutagenesis (Stratagene) and was indistinguishable from WT RSK2 CTD in kinase activity assays, as described previously.
  • WT and C436V RSK2 CTD (10 ⁇ M) were activated by His6-ERK2 (10 ⁇ M) in 20 mM HEPES [pH 8.0], 10 mM MgCl 2 , 2.5 mM tris(2-carboxyethyl)phosphine (TCEP), 0.2 mg/mL BSA and 200 ⁇ M ATP for 30 min at 22° C.
  • RSK2 CTD 5 nM
  • 20 mM HEPES [pH 8.0] 10 mM MgCl 2
  • 2.5 mM tris(2-carboxyethyl)phosphine (TCEP) 0.25 mg/mL BSA
  • 100 ⁇ M ATP 100 ⁇ M ATP were pre-incubated with inhibitors (ten concentrations, in duplicate) for 30 min.
  • Kinase reactions were initiated by the addition of 5 ⁇ Ci of [ ⁇ - 32 P]ATP (6000 Ci/mmol, NEN) and 167 ⁇ M peptide substrate (RRQLFRGFSFVAK) (SEQ ID NO:1) and performed for 30 min at room temperature.
  • Kinase activity was determined by spotting 5 ⁇ L of each reaction onto dried sheets of nitrocellulose that had been pre-washed with 1 M NaCl in 0.1% H 3 PO 4 .
  • the nitrocellulose sheets were washed once with 1% AcOH solution and twice with a solution of 1 M NaCl in 0.1% H 3 PO 4 (5-10 min per wash).
  • Dried blots were exposed for 30 min to a storage phosphor screen and scanned by a Typhoon imager (GE Life Sciences). Data were quantified using the SPOT program (Knight, Z. et al. Nature Protocols, 2: 2459-66), and IC 50 values were determined using GraphPad Prism 4.0 software.
  • Table 1 provides half-maximal inhibitory concentrations (IC 50 in ⁇ M) for electrophilic pyrrolo[2,3-d]pyrimidines 1-8 toward WT RSK2 and C436V RSK2 C-terminal kinase domain (CTD).
  • Compounds 4-6 were additionally tested for RSK2 CTD inhibition in the presence of 10 mM reduced glutathione (GSH).
  • GSH reaction with compounds 4-6 was monitored by UV/visible spectroscopy at 350-400 nm) and being present at one million times the concentration of RSK2 CTD, glutathione had no effect on the inhibitory potency of 4-6.
  • IC 50 value for selected compounds and RSK2 species IC 50 ( ⁇ M) WT RSK2 + WT RSK2 C436V RSK2 10 mM GSH Me enone (1) 0.087 1.7 n/d Me acrylate (2) 0.25 0.19 n/d acrylonitrile (3) 0.75 1.5 n/d CN—OMe (4) 0.013 >10 0.015 CN—OtBu (5) 0.007 >10 0.006 CN—NH2 (6) 0.003 5 0.004 CN—NHiPr (7) 0.005 5.5 n/d CN—NHBn (8) 0.040 n/d n/d
  • RSK2 CTD human RSK2, 399-740
  • E. coli E. coli as a His 6 -tagged fusion protein and purified by Ni/NTA affinity chromatography, followed by cleavage of the His 6 -tag and further purification by size exclusion chromatography.
  • RSK2 CTD (5 ⁇ M) was incubated with the indicated compounds (25 ⁇ M, equiv) for 1 h at room temperature in buffer (20 mM HEPES [pH 8.0], 100 mM NaCl, 10 mM MgCl 2 ).
  • cyanoacrylates and cyanoacrylamides 4-8 do NOT irreversibly modify the RSK2 C-terminal kinase domain (CTD), as revealed by high-resolution mass spectrometry analysis.
  • CTD C-terminal kinase domain
  • Pyrrolo[2,3-d]pyrimidines 1-3 contain conventional electrophilic “warheads” and, as expected, formed irreversible 1:1 adducts with RSK2. This conclusion is supported by the formation of a new peak in the mass spectrum corresponding to the molecular mass of RSK2 CTD plus the molecular mass of the electrophilic compound ( FIG. 1A ). Note that modification of RSK2 CTD by acrylate 2 and acrylonitrile 3 was somewhat slower relative to enone 1, due to the lower intrinsic electrophilicity of the acrylate/acrylonitrile warheads.
  • pyrrolo[2,3-d]pyrimidines (1 ⁇ M) were added to a solution of RSK2 CTD (50 nM, pre-activated with 1 equiv of ERK2) in a buffer containing 20 mM HEPES [pH 8.0], 10 mM MgCl 2 , 2.5 mM tris(2-carboxyethyl)phosphine (TCEP), 0.25 mg/mL BSA, and 100 ⁇ M ATP.
  • the reactions were transferred to a dialysis cassette (0.1-0.5 mL Slide-A-Lyzer, MWCO 10 kDa, Pierce) and dialyzed against 2 L of buffer (20 mM Hepes [pH 8.0], 10 mM MgCl 2 , 1 mM DTT) at 4° C.
  • the dialysis buffer was exchanged after 2 h, and then was exchanged every 24 h until the end of the experiment. Aliquots were removed from the dialysis cassettes every 24 h, flash frozen in liquid nitrogen, and subsequently analyzed for RSK2 kinase activity in triplicate. Kinase activity for each sample was normalized to the DMSO control for that time point and expressed as the mean ⁇ SD.
  • RSK2 CTD kinase activity recovers from inhibition by pyrrolo[2,3-d]pyrimidines 6 and 7 upon dialysis, indicating that 6 and 7 are reversible inhibitors.
  • the data in FIG. 2 show that, upon extensive dialysis at 4° C., RSK2 kinase activity recovers in a time-dependent manner from inhibition by an excess (20 equiv, 1.0 ⁇ M) of cyanoacrylamides 6 ( ⁇ 60% recovery) and 7 ( ⁇ 25% recovery).
  • cyanoacrylamides 6 and 7 are slowly dissociating, reversible inhibitors of RSK2 with dissociation half-times of ⁇ 3 days and >4 days, respectively, under these conditions (dialysis at 4° C.). Note that dissociation is more rapid at room temperature and proceeds to completion in the presence of an irreversible competitor (see FIG. 3 ). In contrast to the partial recovery of kinase activity observed with cyanoacrylamides 6 and 7, RSK2 CTD remained completely inhibited by enone 1 and fluoromethylketone 9 during 4 days of dialysis, further demonstrating that these compounds are irreversible inhibitors. These results are consistent with the LCMS data, which show that enone 1 ( FIG.
  • FIG. 3 Further evidence that cyanoacrylate- and cyanoacrylamide-substituted pyrrolo[2,3-d]pyrimidines form slowly dissociating, fully reversible complexes with RSK2 is provided in FIG. 3 .
  • RSK2 CTD (5 ⁇ M) in 20 mM HEPES [pH 8.0], 10 mM MgCl 2 , 100 mM NaCl, 2.5 mM tris(2-carboxyethyl)phosphine (TCEP) and 0.2 mg/mL BSA was preincubated with 10 ⁇ M inhibitor 1, 4-7 (or DMSO control) for 60 min at room temperature. FMK 9 (100 ⁇ M) was then added, and aliquots were removed at different time points (0.5-1500 min) and immediately quenched by mixing with an equal volume of 0.4% formic acid. Samples were analyzed by LCMS with an LCT Premier mass spectrometer and MassLynx deconvolution software, as described in FIG. 1 .
  • the graph on the right side of FIG. 3 shows FMK labeling kinetics in the absence of any competitor.
  • Kinetic data (% FMK adduct vs. time) were fit to a single exponential (PRISM 4.0) to obtain dissociation half-times depicted in the table. Control experiments showed that C436V RSK2 was not modified by FMK 9 under these conditions.
  • cyanoacrylates/cyanoacrylamides 4-7 dissociate with a half-time of hours from intact, folded RSK2 CTD, as measured by competitive labeling with fluoromethylketone 9.
  • a large molar excess of fluoromethylketone 9 (100 ⁇ M, 20 equiv; “FMK” in FIG. 3 ) rapidly and irreversibly modified RSK2 CTD (t 1/2 ⁇ 2 min), as revealed by LCMS analysis of the RSK2 CTD ( FIG. 3 , upper left graph).
  • Absorbance spectra are shown in the middle and lower panels for experiments with proteinase K and SDS, respectively (absolute absorbance values are different in the two experiments because different concentrations of cyanoacrylamide 7 were used). Absorbance values for all three conditions were normalized to the absorbance of cyanoacrylamide 7 in buffer alone and are shown in the bar graph.
  • FIG. 5 shows LCMS chromatograms derived from similar experiments, proving that the addition of protein denaturants to the covalent complex formed between cyanoacrylamide 7 and RSK2 CTD results in the quantitative liberation of cyanoacrylamide 7.
  • glutathione an abundant cysteine-containing peptide, formed covalent adducts with compounds 4-8 (shown by UV/Visible spectroscopy) that rapidly dissociated, as shown by (1) rapid recovery of the cyanoacrylate/cyanoacrylamide chromophore upon dilution, and (2) unperturbed RSK2 inhibitory potency in the presence of 10 mM glutathione (Table 1).
  • the dependence of the rate of covalent bond dissociation of a protein thiol/electrophile adduct on the folded state of the protein has not, to our knowledge, been described.
  • Cyanoacrylamide 7 (250 ⁇ M) was incubated in the absence or presence of RSK2 CTD (300 ⁇ M) for 10 min in a total volume of 50 ⁇ L. Guanidine hydrochloride (50 ⁇ L, 6 M, pH 8) was added and the contents were mixed gently for 1 min, after which acetonitrile was added to a final concentration of 50%. The solution was filtered (0.2 ⁇ m pore size) and analyzed by LCMS (20 ⁇ L injection, Waters XTerra MS C18 column, 5-70% MeCN/water+0.1% formic acid over 20 min; Waters 2695 Alliance Separations Module, Waters Micromass ZQ mass spectrometer).
  • LCMS analysis revealed quantitative recovery of cyanoacrylamide 7 after denaturation of the RSK2 CTD/cyanoacrylamide 7 complex with 3 M guanidine.
  • HEK-293 cells in a 75 cm 2 flask were transfected with the pMT2 expression vector encoding HA-tagged RSK2 using Lipofectamine 2000 (Invitrogen) according to the manufacturer's protocol. After 12 h, the cells were trypsinized and seeded into 6-well plates at 600,000 cells per well in DMEM with 10% serum. After an additional 16 h, the cells were deprived of serum for 4 h and then treated with the indicated concentrations of inhibitors for 2 h in serum-free DMEM.
  • the cells were stimulated for 30 min with phorbol myristate acetate (PMA) (100 ng/ml), then washed with 2 mL cold PBS and frozen onto the plate at ⁇ 80° C.
  • PMA phorbol myristate acetate
  • the cells were thawed and scraped into 80 ⁇ L of lysis buffer (20 mM Hepes pH 7.9, 450 mM NaCl, 25% glycerol, 3 mM MgCl 2 , 0.5 mM EDTA) with protease (Complete, Roche) and phosphatase (Cocktails 1 and 2, Sigma-Aldrich) inhibitors.
  • the lysates were cleared by centrifugation and normalized via Bradford assay quantification.
  • cyanoacrylate 5 and cyanoacrylamides 6 and 7 inhibit RSK2 autophosphorylation of Ser386 in mammalian cells.
  • N-isopropyl cyanoacrylamide 7 was the most potent (IC 50 ⁇ 30 nM) among all the inhibitors tested, whereas cyanoacrylamide 6 had weak activity (IC 50 ⁇ 1000 nM).
  • the data show that the cyanoacrylate and cyanoacrylamide inhibitors are cell permeable and sufficiently potent to compete with high intracellular concentrations of ATP and glutathione.
  • NEK2 Full-length human T175A NEK2 (referred to as “NEK2”) was expressed and purified as previously described (Knapp S. et al. J. Biol. Chem., 2007, 282: 6833-6842).
  • the C22V mutant of NEK2 was generated by Quikchange mutagenesis (Stratagene) and was indistinguishable from NEK2 in kinase activity assays.
  • NEK2 kinases 60 nM in 20 mM Hepes, pH 7.6, 10 mM MgCl 2 , 1 mM EDTA, 0.2 mg/mL BSA, and 100 ⁇ M ATP were pre-incubated with inhibitors (8-10 concentrations, in duplicate) for 30 min at room temperature.
  • Kinase reactions were initiated by the addition of 0.4 ⁇ Ci/ ⁇ L of ⁇ - 32 P-ATP (6000 Ci/mmol, NEN) and 2.37 mg/mL ⁇ -casein (Sigma) and incubated for 30 minutes at room temperature.
  • Human PLK1 (Millipore, catalog number 14-777M) (7.2 nM) in 20 mM Hepes, pH 7.6, 10 mM MgCl 2 , 1 mM EDTA, 0.2 mg/mL BSA, and 100 ⁇ M ATP was pre-incubated with inhibitors (8-10 concentrations, in duplicate) for 30 min at room temperature.
  • Kinase reactions were initiated by the addition of 0.4 ⁇ Ci/ ⁇ L of ⁇ - 32 P-ATP (6000 Ci/mmol, NEN) and 0.5 mg/mL dephosphorylated ⁇ -casein (Sigma) and incubated for 30 min at room temperature.
  • Kinase activity was determined by spotting 5 ⁇ L of each reaction onto dried sheets of nitrocellulose pre-washed with 1M NaCl in 0.1% H 3 PO 4 . After blotting each kinase reaction, the nitrocellulose sheets were washed once with 1% AcOH solution. Kinase activity was determined by spotting 5 ⁇ L of each reaction onto dried sheets of nitrocellulose that had been pre-washed with 1 M NaCl in 0.1% H 3 PO 4 . The nitrocellulose sheets were washed once with 1% AcOH solution and twice with a solution of 1 M NaCl in 0.1% H 3 PO 4 (5-10 minutes per wash).
  • Dried blots were exposed for 30 minutes to a storage phosphor screen and scanned by a Typhoon imager (GE Life Sciences). Data were quantified using the SPOT program (Knight, Z. et al. Nature Protocols, 2: 2459-66), and IC 50 values were determined using GraphPad Prism 4.0 software.
  • cyanoacrylamides As cysteine-targeting moieties for the inhibition of therapeutically relevant proteins, we synthesized a panel of cyanoacrylamides that were substituted with heterocycles commonly found in kinase inhibitor drugs (e.g., azaindoles, indazoles, pyridines, pyrazoles, biaryls).
  • kinase inhibitor drugs e.g., azaindoles, indazoles, pyridines, pyrazoles, biaryls.
  • RSK2 CTD was expressed and purified as described above (see description for FIG. 1A AND FIG. 1B ) and concentrated to 20 mg/ml in 20 mM HEPES pH 8.0, 50 mM NaCl. Cyanoacrylamide 6, 12, or 15 (1 ⁇ l of a 10 mM stock solution in 100% DMSO) was then added to 19 ⁇ l of RSK2 CTD, to give 19 mg/ml protein and 0.5 mM inhibitor in 5% DMSO.
  • Cyanoacrylamides when appended to three different kinase inhibitor scaffolds, bind to the ATP binding pocket of RSK2 CTD and form a covalent bond with Cys-436.
  • pyrrolo-pyrimidine 6 (Table 1)
  • azaindole 12 (Table 2)
  • indazole 15 (Table 2).
  • the co-crystal structures of 6, 12, and 15 reveal non-covalent interactions that are typically observed in kinase/inhibitor co-crystal structures; e.g., hydrogen bonding between a heteroatom in the inhibitor and the backbone NH of RSK2 Met-496.
  • a covalent bond between the thiol of Cys-436 and the beta-carbon of the cyanoacrylamide moiety is clearly visible ( FIG. 7 , created from the structure coordinate files with PyMol).
  • FIG. 7 created from the structure coordinate files with PyMol
  • the kinase domain of chicken cSrc (S345C mutant) was expressed and purified as described (Blair et al., Nat. Chem. Biol., 2007) and concentrated to 3 mg/ml in 20 mM Tris pH 7.5, 100 mM NaCl, 1 mM DTT, 5% Glycerol. Protein (3 mg/mL) was incubated for 10 minutes on ice with inhibitor (200 ⁇ M) with 2% DMSO. Crystals of the complex were then grown in hanging drops by mixing 1 ⁇ l of protein/inhibitor—complex with 1 ⁇ l of precipitant solution composed of 100 mM MES, 50 mM NaOAc, 2% PEG(4000), pH 6.5 at 20° C. Typically crystals grew as thin plates to maximal dimensions in 1-2 days. Crystals were then transferred to a cryoprotectant solution consisting of mother liquor supplemented with 25% glycerol and frozen in a stream of liquid nitrogen at 100 K.
  • Wild-type and S345C mutant cSrc kinase domains were expressed and purified as described (Blair et al., Nat. Chem. Biol., 2007).
  • the purified cSrc kinase (2 nM final concentration) was pre-incubated with inhibitors (six or ten concentrations, in duplicate) for 30 minutes at room temperature in kinase reaction buffer (20 mM HEPES pH 7.4, 10 mM MgCl 2 0.2 mM EDTA) with 200 ⁇ M ATP, and 1 mg/mL BSA.
  • Kinase reactions were initiated by the addition of 0.05 ⁇ Ci/ ⁇ L of ⁇ - 32 P-ATP (6000 Ci/mmol, NEN) and 0.1 mM substrate peptide (LEIYGEFKKK) (SEQ ID NO:2) and incubated for 30 minutes at room temperature. Kinase activity was determined by spotting 6 ⁇ L of each reaction onto sheets of phosphocellulose. Each blot was washed once with 1% AcOH solution, twice with 0.1% H 3 PO 4 solution, and once with MeOH (5-10 minutes per wash). Dried blots were exposed for 30 minutes to a storage phosphor screen and scanned by a Typhoon imager (GE Life Sciences). The data were quantified using ImageQuant (v. 5.2, Molecular Dynamics) and plotted using GraphPad Prism 4.0 software.
  • the combined filtrate and wash were concentrated to afford the intermediate tert-butyl-2-chloroacetamide as white solid, which was carried on to the next step without any further purification.
  • the tert-butyl-2-chloroacetamide was dissolved in DMF (12 mL), and 2.31 g (35.4 mmol, 2.0 equiv) of finely crushed KCN was added. The reaction mixture was heated to 70° C. for 2 hours and then filtered. The filter cake was washed with EtOAc (2 ⁇ 30 mL). The combined washes and filtrate were concentrated to afford an amber oil.
  • the combined filtrate and wash were concentrated to afford the intermediate 1-adamantyl-2-chloroacetamide as white solid, which was carried on to the next step without any further purification.
  • the 1-adamantyl-2-chloroacetamide was dissolved in DMF (6 mL), and 1.61 g (24.7 mmol, 2.0 equiv) of finely crushed KCN was added. The reaction mixture was heated to 70° C. for 14 hours and then cooled to ambient temperature. The reaction mixture was diluted with EtOAc (100 mL) and washed with water (3 ⁇ 60 mL). The combined aqueous washes were extracted with EtOAc (70 mL).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Neurology (AREA)
  • Diabetes (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Communicable Diseases (AREA)
  • Neurosurgery (AREA)
  • Virology (AREA)
  • Obesity (AREA)
  • Biomedical Technology (AREA)
  • Pain & Pain Management (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Pulmonology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Endocrinology (AREA)
  • Cardiology (AREA)
  • AIDS & HIV (AREA)
  • Child & Adolescent Psychology (AREA)
  • Immunology (AREA)
  • Emergency Medicine (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Rheumatology (AREA)
  • Psychology (AREA)
  • Molecular Biology (AREA)
  • Vascular Medicine (AREA)
US13/510,272 2009-11-16 2010-11-16 Kinase inhibitors Abandoned US20130035325A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/510,272 US20130035325A1 (en) 2009-11-16 2010-11-16 Kinase inhibitors

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US26169609P 2009-11-16 2009-11-16
US33027110P 2010-04-30 2010-04-30
US13/510,272 US20130035325A1 (en) 2009-11-16 2010-11-16 Kinase inhibitors
PCT/US2010/056890 WO2011060440A2 (en) 2009-11-16 2010-11-16 Kinase inhibitors

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/056890 A-371-Of-International WO2011060440A2 (en) 2009-11-16 2010-11-16 Kinase inhibitors

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/462,158 Continuation US9505766B2 (en) 2009-11-16 2014-08-18 Kinase inhibitors

Publications (1)

Publication Number Publication Date
US20130035325A1 true US20130035325A1 (en) 2013-02-07

Family

ID=43992477

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/510,272 Abandoned US20130035325A1 (en) 2009-11-16 2010-11-16 Kinase inhibitors
US14/462,158 Active US9505766B2 (en) 2009-11-16 2014-08-18 Kinase inhibitors

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/462,158 Active US9505766B2 (en) 2009-11-16 2014-08-18 Kinase inhibitors

Country Status (9)

Country Link
US (2) US20130035325A1 (de)
EP (1) EP2558099A4 (de)
JP (1) JP2013510886A (de)
CN (1) CN102711765A (de)
AU (1) AU2010319964A1 (de)
BR (1) BR112012011528A2 (de)
CA (1) CA2781056A1 (de)
MX (1) MX2012005678A (de)
WO (1) WO2011060440A2 (de)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016007185A1 (en) * 2014-07-07 2016-01-14 Eternity Bioscience Inc. Aminopyridazinone compounds as protein kinase inhibitors

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5974084B2 (ja) 2011-05-17 2016-08-23 プリンシピア バイオファーマ インコーポレイテッド チロシンキナーゼ阻害剤
US9376438B2 (en) 2011-05-17 2016-06-28 Principia Biopharma, Inc. Pyrazolopyrimidine derivatives as tyrosine kinase inhibitors
DK2710007T3 (da) 2011-05-17 2020-01-27 Principia Biopharma Inc Kinasehæmmere
AU2013277582B2 (en) 2012-06-18 2017-11-30 Principia Biopharma Inc. Reversible covalent pyrrolo- or pyrazolopyrimidines useful for the treatment cancer and autoimmune diseases
WO2014036016A1 (en) 2012-08-31 2014-03-06 Principia Biopharma Inc. Benzimidazole derivatives as itk inhibitors
AU2013312296B2 (en) * 2012-09-10 2017-06-22 Principia Biopharma Inc. Pyrazolopyrimidine compounds as kinase inhibitors
WO2014093230A2 (en) 2012-12-10 2014-06-19 Merck Patent Gmbh Compositions and methods for the production of pyrimidine and pyridine compounds with btk inhibitory activity
US8957080B2 (en) 2013-04-09 2015-02-17 Principia Biopharma Inc. Tyrosine kinase inhibitors
EA033900B1 (ru) 2014-02-21 2019-12-06 Принсипиа Биофарма Инк. СОЛЕВЫЕ И АМОРФНЫЕ ФОРМЫ 2-[(3R)-3-[4-АМИНО-3-(2-ФТОР-4-ФЕНОКСИФЕНИЛ)ПИРАЗОЛО[3,4-d]ПИРИМИДИН-1-ИЛ]ПИПЕРИДИН-1-КАРБОНИЛ]-4-МЕТИЛ-4-[4-(ОКСЕТАН-3-ИЛ)ПИПЕРАЗИН-1-ИЛ]ПЕНТ-2-ЕННИТРИЛА, ФАРМАЦЕВТИЧЕСКАЯ КОМПОЗИЦИЯ НА ИХ ОСНОВЕ И СПОСОБ ЛЕЧЕНИЯ ПУЗЫРЧАТКИ ОБЫКНОВЕННОЙ И ПУЗЫРЧАТКИ ЛИСТОВИДНОЙ С ИХ ПРИМЕНЕНИЕМ
EA036269B1 (ru) 2014-12-18 2020-10-21 Принсипиа Биофарма Инк. Лечение пузырчатки
US20180305350A1 (en) 2015-06-24 2018-10-25 Principia Biopharma Inc. Tyrosine kinase inhibitors
CN106939002B (zh) * 2016-01-05 2019-09-24 江苏恒瑞医药股份有限公司 一种btk激酶抑制剂的结晶形式及其制备方法
JP6913274B2 (ja) * 2016-01-05 2021-08-04 ジエンス ヘンルイ メデイシンカンパニー リミテッドJiangsu Hengrui Medicine Co.,Ltd. Btkキナーゼ阻害剤の結晶形およびその製造方法
US20190231784A1 (en) 2016-06-29 2019-08-01 Principia Biopharma Inc. Modified release formulations of 2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile
JOP20180094A1 (ar) 2017-10-18 2019-04-18 Hk Inno N Corp مركب حلقي غير متجانس كمثبط بروتين كيناز
CN111205232B (zh) * 2020-02-26 2022-07-01 浙江天宇药业股份有限公司 一种替格瑞洛中间体的合成方法
WO2022251597A1 (en) 2021-05-28 2022-12-01 Verge Analytics, Inc. Methods of treating neurological disorders with modulators of ribosomal protein s6 kinase alpha-1 (rsk1) and ribosomal protein s6 kinase alpha-3 (rsk2)
CN116332996A (zh) * 2023-05-04 2023-06-27 南京颐媛生物医学研究院有限公司 抗冠状病毒化合物及其制备方法和应用
CN116284135A (zh) * 2023-05-04 2023-06-23 南京颐媛生物医学研究院有限公司 抗冠状病毒核苷类化合物的制备方法及其应用

Family Cites Families (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4444770A (en) * 1980-05-29 1984-04-24 Bayer Aktiengesellschaft New imidazoazole-alkenoic acid amide compounds, intermediate products for their production, their production, and their medicinal use
DE3216416A1 (de) * 1982-05-03 1983-11-03 Bayer Ag, 5090 Leverkusen Heteroarylthiovinyl-verbindungen, ein verfahren zu ihrer herstellung und ihre verwendung als schaedlingsbekaempfungsmittel
US4911920A (en) 1986-07-30 1990-03-27 Alcon Laboratories, Inc. Sustained release, comfort formulation for glaucoma therapy
DE3502264A1 (de) * 1985-01-24 1986-07-24 Bayer Ag, 5090 Leverkusen Imidazoalkensaeurederivate, ein verfahren zu ihrer herstellung und ihre verwendung fuer die herstellung pharmazeutischer wirkstoffe
FR2588189B1 (fr) 1985-10-03 1988-12-02 Merck Sharp & Dohme Composition pharmaceutique de type a transition de phase liquide-gel
WO1990007508A1 (en) * 1988-12-26 1990-07-12 Zenyaku Kogyo Kabushiki Kaisha 1-azaindolizine derivative, intermediate for synthesis thereof, and antiallergic containing 1-azaindolizine derivative
GB9004483D0 (en) * 1990-02-28 1990-04-25 Erba Carlo Spa New aryl-and heteroarylethenylene derivatives and process for their preparation
DK0495421T3 (da) 1991-01-15 1996-12-09 Alcon Lab Inc Anvendelse af carragenaner i topiske ophthalmiske sammensætninger
US5212162A (en) 1991-03-27 1993-05-18 Alcon Laboratories, Inc. Use of combinations gelling polysaccharides and finely divided drug carrier substrates in topical ophthalmic compositions
JPH05301838A (ja) * 1991-10-15 1993-11-16 Mitsubishi Kasei Corp スチレン誘導体
US5792771A (en) * 1992-11-13 1998-08-11 Sugen, Inc. Quinazoline compounds and compositions thereof for the treatment of disease
GB9226855D0 (en) * 1992-12-23 1993-02-17 Erba Carlo Spa Vinylene-azaindole derivatives and process for their preparation
AU2096895A (en) * 1994-03-07 1995-09-25 Sugen, Incorporated Receptor tyrosine kinase inhibitors for inhibiting cell proliferative disorders and compositions thereof
GB9406137D0 (en) * 1994-03-28 1994-05-18 Erba Carlo Spa N-substituted beta-aryl- and betaheteroaryl-alpha-cyanoacrylamide derivatives and process for their preparation
GB9412719D0 (en) * 1994-06-24 1994-08-17 Erba Carlo Spa Substituted azaindolylidene compounds and process for their preparation
US6331555B1 (en) * 1995-06-01 2001-12-18 University Of California Treatment of platelet derived growth factor related disorders such as cancers
WO1999043673A1 (fr) * 1998-02-26 1999-09-02 Zenyaku Kogyo Kabushiki Kaisha Derives de 1-aza-indolizine
DE602004009582T2 (de) * 2003-08-01 2009-01-02 Chugai Seiyaku K.K. Cyanoamid-verbindungen als nützliche malonyl-coa decarboxylase-hemmer
US7807719B2 (en) * 2004-09-14 2010-10-05 Chaim Roifman Compounds useful for modulating abnormal cell proliferation
KR20080066938A (ko) * 2005-10-07 2008-07-17 깃세이 야쿠힌 고교 가부시키가이샤 질소 함유 복소환 화합물 및 그것을 함유하는 의약 조성물
RU2456265C2 (ru) * 2006-03-31 2012-07-20 Дзе Борд Оф Риджентс Оф Дзе Юниверсити Оф Техас Систем Биологически доступная для перорального применения кофейная кислота, относящаяся к противоопухолевым лекарственным средствам
US7645748B2 (en) 2006-04-03 2010-01-12 Forbes Medi-Tech Inc. Sterol/stanol phosphorylnitroderivatives and use thereof
WO2008005954A2 (en) * 2006-06-30 2008-01-10 The Board Of Regents Of The University Of Texas System Tryphostin-analogs for the treatment of cell proliferative diseases
EP2471529A3 (de) * 2006-09-05 2012-10-10 Emory University Tyrosinkinasehemmer zur Vorbeugung oder Behandlung von Infektionen
US20120028981A1 (en) 2008-11-05 2012-02-02 Principia Biopharma Inc. Kinase Knockdown Via Electrophilically Enhanced Inhibitors
US20100113520A1 (en) 2008-11-05 2010-05-06 Principia Biopharma, Inc. Kinase knockdown via electrophilically enhanced inhibitors
US8426428B2 (en) 2008-12-05 2013-04-23 Principia Biopharma, Inc. EGFR kinase knockdown via electrophilically enhanced inhibitors

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
CAS RN 26272-41-3 (entered into STN 11/16/1984) *
CAS RN 63431-27-6 (entered into STN 11/16/1984) *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016007185A1 (en) * 2014-07-07 2016-01-14 Eternity Bioscience Inc. Aminopyridazinone compounds as protein kinase inhibitors
US9951077B2 (en) 2014-07-07 2018-04-24 Jiangsu Hengrui Medicine Co., Ltd. Aminopyridazinone compounds as protein kinase inhibitors
US10323037B2 (en) 2014-07-07 2019-06-18 Jiangsu Hengrui Medicine Co., Ltd. Aminopyridazinone compounds as protein kinase inhibitors

Also Published As

Publication number Publication date
EP2558099A4 (de) 2013-07-17
AU2010319964A2 (en) 2012-08-02
US20150045343A1 (en) 2015-02-12
JP2013510886A (ja) 2013-03-28
US9505766B2 (en) 2016-11-29
WO2011060440A3 (en) 2011-11-03
BR112012011528A2 (pt) 2019-09-24
WO2011060440A2 (en) 2011-05-19
MX2012005678A (es) 2012-09-07
EP2558099A2 (de) 2013-02-20
CA2781056A1 (en) 2011-05-19
CN102711765A (zh) 2012-10-03
AU2010319964A1 (en) 2012-06-07

Similar Documents

Publication Publication Date Title
US9505766B2 (en) Kinase inhibitors
CA2836449C (en) Kinase inhibitors
JP5465318B2 (ja) キナーゼ阻害剤としてのピリミジン置換プリン化合物
KR101546493B1 (ko) 이미다조[1,2-b]피리다진 및 피라졸로[1,5-a]피리미딘 유도체 및 단백질 키나제 억제제로서의 이의 용도
EP2551270B1 (de) Pyrazolopyrimidin-derivate zur verwendung as pi3 kinase antagonisten
US8637543B2 (en) Quinoline derivatives as kinase inhibitors
ES2856125T3 (es) Derivado de sulfonamida y composición farmacéutica que contiene la misma
CA3015484A1 (en) Novel condensed pyrimidine compound or salt thereof
TW200804387A (en) Novel imidazopyrazines as cyclin dependent kinase inhibitors
TW200400167A (en) Benzamide derivatives
US9475817B2 (en) Pyrazole substituted imidazopyrazines as casein kinase 1 d/e inhibitors
US20230142274A1 (en) Substituted aryl compound
TW201817724A (zh) 化合物
WO2018121400A1 (zh) 酰胺及硫代酰胺类衍生物及其制备方法和应用
AU2021270672A1 (en) Fused aza-heterocyclic amide compound and use thereof
CA3172186A1 (en) Pyrazolo[3,4-d]pyrimidin-6-yl-sulfonamide derivatives for the inhibition of sgk-1
KR102516260B1 (ko) Trap1 선택적 억제제로서의 화합물 및 이를 포함하는 암 예방 또는 치료용 조성물
US20230406853A1 (en) Covalent cdk2-binding compounds for therapeutic purposes
CN117062817A (zh) 用于抑制sgk-1的吡唑并[3,4-d]嘧啶-6-基-磺酰胺衍生物
JP5746777B2 (ja) キナーゼ阻害剤としてのピリミジン置換プリン化合物

Legal Events

Date Code Title Description
AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:UNIVERSITY OF CALIFORNIA, SAN FRANCISCO;REEL/FRAME:033193/0421

Effective date: 20140516

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION