AU2014250696A1 - Intracellular kinase inhibitors - Google Patents

Intracellular kinase inhibitors Download PDF

Info

Publication number
AU2014250696A1
AU2014250696A1 AU2014250696A AU2014250696A AU2014250696A1 AU 2014250696 A1 AU2014250696 A1 AU 2014250696A1 AU 2014250696 A AU2014250696 A AU 2014250696A AU 2014250696 A AU2014250696 A AU 2014250696A AU 2014250696 A1 AU2014250696 A1 AU 2014250696A1
Authority
AU
Australia
Prior art keywords
kinase
active site
inhibitor
compounds
hydrogen
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
AU2014250696A
Inventor
David William Brandt
Subrata Chakravarty
Mary Faris
Gary A. Flynn
Sandra Aeyoung Lee
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.)
Pharmacyclics LLC
Original Assignee
Mannkind Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2013200229A external-priority patent/AU2013200229B2/en
Application filed by Mannkind Corp filed Critical Mannkind Corp
Priority to AU2014250696A priority Critical patent/AU2014250696A1/en
Publication of AU2014250696A1 publication Critical patent/AU2014250696A1/en
Assigned to PHARMACYCLICS, INC. reassignment PHARMACYCLICS, INC. Request for Assignment Assignors: MANNKIND CORPORATION
Assigned to PHARMACYCLICS LLC reassignment PHARMACYCLICS LLC Amend patent request/document other than specification (104) Assignors: PHARMACYCLICS, INC.
Abandoned legal-status Critical Current

Links

Abstract

INTRACELLULAR KINASE INHIBITORS Intracellular kinase inhibitors and their therapeutic uses for patients with T cell malignancies, B cell malignancies, autoimmune disorders and transplanted organs.

Description

INTRACELLULAR KINASE INHIBITORS [01] This application claims the benefit of and incorporates by reference co-pending provisional application Serial No. 60/801,074 filed May 18, 2006 and Serial No. 60/869,664 filed December 12, 2006. FIELD OF THE INVENTION 1021 The invention relates to intracellular kinase inhibitors and their therapeutic uses. BACKGROUND OF THE INVENTION [03] Intracellular kinases play important functions in cells of the immune system. For example, interleukin-2 inducible tyrosine kinase (ITK) plays a key role in T cell development and differentiation; it regulates IL-2 production via phospholipase C y1 (PLCyl) and nuclear factor of activated T cells (NFAT); it mediates Th2 cell differentiation; and it regulates T cell migration and recruitment to lymphatic organs. Bruton's tyrosine kinase (BTK) is involved in signal transduction pathways which regulate growth and. differentiation of B lymphoid cells. BTK also is involved in platelet physiology by regulating the glycoprotein VI/Fe receptor y chain (GPVI-FcRy)-coupled collagen receptor signaling pathway. For these reasons, inhibitors of intracellular kinases are useful for treating blood cell malignancies, solid tumors and for suppressing the immune system, for example in patients with autoimmune disorders or organ transplants. Intracellular kinase inhibitors also are useful for preventing or reducing the risk of thromboembolism. BRIEF DESCRIPTION OF THE DRAWING [04] FIG. 1. Results of a BIACORE* experiment in which the ITK kinase domain was immobilized on a biosensor and evaluated for its ability to bind and dissociate from a small molecule. [051 FIG. 2. Alignment of human ITK (SEQ ID NO:1) and BTK (SEQ ID NO:2). 1 [06] FIG. 3. Alignment of kinase domains. Bolded amino acids, hinge; bolded and underlined amino acids, gatekeeper; italicized and bolded amino acids, Cys442 equivalents. DETAILED DESCRIPTION OF THE INVENTION [071 The invention provides compounds which inhibit intracellular kinases, particularly ITK and BTK, with an IC 50 of 1 pM or below in an in vitro kinase assay as disclosed herein. The invention also provides pharmaceutical compositions and methods of using the compounds therapeutically. Patients who can be treated include those with blood cell malignancies, solid tumors, autoimmune disorders, and transplanted organs. [08] A review of the literature and patent database revealed the existence of compounds that inhibit ITK or BTK kinases. However, these compounds differ significantly from the compounds disclosed herein. In several instances, the compounds are pyrrolopyridines (e.g., US 2005/0215582). In other instances, the compounds are methyl dimethylbenzoates that belong thiazolyl family of compounds (e.g., US 2004/0077695). In all cases, these published compounds differ from the compounds disclosed herein based on the following parameters: the compounds do not correspond to the general structure shown in this application, do not require the amino acid triad DKC found in the kinase binding site and necessary for optimal compound inhibitory capability described herein, do not undergo elimination, and do not bind covalently to the kinase binding pocket. [09] Compounds of the invention which inhibit ITK can be used, e.g., to treat T cell malignancies. Preferred compounds of the invention inhibit both ITK and BTK with an
IC
5 o of 1 pM or below for each enzyme. Such compounds can be used, e.g., to treat both T and B cell malignancies, as well as EGFR or HER positive tumors. [10] The Tec family of kinases share a common subunit structure composed of a Src homology domain 2 (SH 2 ), .an SH 3 and a catalytic kinase domain. Further, they are uniquely identified by the presence of a Tec -homology region (TH) and a pleckstrin homology (PH) domain. There are-four known crystallographic structures described for 2 the Tec family of kinases. These.- include (a) two structures representing the phosphorylated and unphosphorylated staurosporine-bound ITK (PDB codes 1SM2, 1SNU); (b) one structure of the unphosphorylated apo-form of ITK (PDB code 1SNX), and (c) one structure for the unphosphorylated apo-form of BTK (Mao et al. J. Biol Chem. 2001, 276, 41435-41443). For the purpose of clarity of explanation, this disclosure will represent these kinase structures with those of the nearly identical ITK structures in (a) and (b) incorporated herein by reference (Brown et al. J. Bio. Chem. 2004, 279, 18727-18732) focusing attention on the ATP binding site. For the sake of uniformity, the residue numbering in these kinase structures as represented in the Protein Data Bank have been incorporated throughout this document to describe the kinase domain. The amino acid sequence of human ITK is shown in SEQ ID NO:1. The amino acid sequence of human BTK is shown in SEQ ID NO:2. Homologous residues in the other kinases and sequences from other sources may be numbered differently. [111 Referring to Figure 2, The ITK kinase domain (residues 357-620) can be broken down into two components: the N-terminal lobe (residues 357-437) and the C-terminal lobe (residues 438-620). Like most kinases, the connecting region between the two lobes is a flexible hinge region described below, that forms part of the catalytic active site. The ordered nature of the C-helix places the catalytically important residues of Glu406, Lys391 and Asp500 in an orientation typical of the active form of a protein kinase. The Gly-rich loop (residues 362-378), commonly observed in kinases, assumes an extended and open conformation typical of an active kinase. [12] The boundaries of the ATP binding site are demarcated by the following residues: (a) the glycine-rich loop (Gly370, Ser371, Gly372, Gln373, Phe374 and Gly375); (b). the hinge region (Phe435, Glu436, Phe437, Met438, Glu439, His440, Gly441 and Cys442); and (c) the catalytic residues Lys391 and Asp500. Additionally, the active site also comprises several other hydrophobic residues including Ala389, Ile369, Val377, Val419, and Leu 489 as well as the hydrophilic residue Ser499. [13] Similar to other kinases, the hinge region of ITK contains two backbone carbonyls and one backbone amino group as potential hydrogen bond acceptor and donor sites 3 respectively. Similar backbone interactions have been observed in the interaction of kinases with the adenine base of ATP and several competitive inhibitors have been designed pursuing this concept. At the N-terminal end of the hinge region lies the "gatekeeper" residue, Phe435. This residue blocks access to an internal hydrophobic pocket, and, at the same time, provides a potential site of interaction for aromatic or hydrophobic groups. This "gatekeeper" residue is a significant difference between ITK and BTK. Despite the strong overall sequence identity between BTK and ITK, the presence of the smaller threonine residue as a gatekeeper in the active site of BTK justifies a key similarity of the latter to the active site of several kinases such as Src/Abl/EGFR. The absence of the bulkier Phe gatekeeper allows access to an internal hydrophobic pocket for these kinases, a fact that has been exploited for the design of allosteric inhibitors, and to improve the affinity of ATP-competitive inhibitors through the addition of a hydrophobic pharmacophore. Definitions [14] "Alkyl" is a monovalent linear or branched saturated hydrocarbon radical and can be substituted or unsubstituted. Linear or branched alkyls typically have between 1 and 12 carbon atoms (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12). Lower alkyls, or "C-C 6 alkyls," have between 1 and 6 carbon atoms (e.g., 1, 2, 3, 4, 5, or 6). Optional substitutents include halogen, hydroxyl, alkoxy, aryloxy, amino, N-alkylamino, N,N-dialkylamino, alkylcarbamoyl, arylcarbamoyl, aminocarbamoyl, N-alkylaminocarbamoyl,
N,N
dialkylaminocarbamoyl, alkylsulfonylamino, arylsulfonylamino, carboxy, carboxyalkyl, N-alkylcarboxamido, N,N-dialkylcarboxamido, alkylthio, alkylsulfinyl, alkylsulfonyl, trihaloalkylsulfonylamino (e.g., trifluoromethylsulfonylamino), arylthio, arylsulfinyl, arylsulfonyl, and heterocyclyl. Examples of linear or branched C-C 6 alkyl are methyl, ethyl, propyl, isopropyl, sec-butyl, tert-butyl, n-butyl, n-pentyl, sec-pentyl, tert-pentyl, n hexyl, isopentyl, fluoromethyl, trifluoromethyl, hydroxybutyl, dimethylcarboxyalkyl, aminoalkyl, and benzylpropyl. 4 [15] "Acyl" (or "alkylcarbonyl") is the radical -C(O)R, wherein R 8 is an optionally substituted lower alkyl. Examples. of acyl include, but are not limited to, acetyl, propionyl, n-butyryl, sec-butyryl, t-butyryl, iodoacetyl, and benzylacetyl. [16] "Acyloxy" is the radical -OC(O)R 8 , wherein R 8 is an optionally substituted lower alkyl. Examples of acyloxy include, but are not limited to, acetoxy, propionyloxy, butyryloxy, trifluoroacetoxy, and diiodobutyryloxy. [17] "Alkoxy" is the radical -OR 8 , wherein R 8 is an optionally substituted lower alkyl. Examples of alkoxy include methoxy, ethoxy, propoxy, 2-propoxy, butoxy, sec-butoxy, tert-butoxy, pentyloxy, hexyloxy, fluoromethoxy, and iodoethoxy. [181 "Alkylamino" is the radical -NR 7 R , wherein R 7 is hydrogen or an optionally substituted lower alkyl and R 8 is an optionally substituted lower alkyl. Examples of alkylamino groups are methylamino, ethylamino, isopropylamino, dimethylamino, diethylamino, and trifluoromethylamino. [19] "Alkylaminocarbonyl" (or "alkylcarbamoyl") is the radical -C(O)NR 7
R
8 , wherein R 7 is hydrogen or an optionally substituted lower alkyl and R 8 is an optionally substituted lower alkyl. Examples of alkylaminocarbonyl include, but are not limited to, methylaminocarbonyl, dimethylaminocarbonyl, t-butylaminocarbonyl, n butylaminocarbonyl, iso-propylaminocarbonyl, and trifluoromethylaminocarbonyl. [201 "Alkylaminosulfonyl" is the radical -S(O) 2 NR 7R, wherein R 7 is hydrogen or. an optionally substituted lower alkyl and R 8 is an optionally substituted lower alkyl. Examples of alkylaminosulfonyl include, but are not limited to, methylaminosulfonyl, dimethylaminosulfonyl, and triiodomethylaminosulfonyl. [21] "Alkoxycarbonyl" or "alkyl ester" is the radical -C(O)OR , wherein R8 is an optionally substituted lower alkyl. Examples of alkoxycarbonyl radicals include, but are not limited to, methoxycarbonyl, ethoxycarbonyl, sec-butoxycarbonyl, isopropyloxycarbonyl, and difluoromethoxycarbonyl. 5 [22] "Alkylcarbonylamino" is the radical -NR 7 C(O)Rs, wherein R 7 is hydrogen or an optionally substituted lower alkyl and R 8 is an optionally substituted lower alkyl. Examples of alkylcarbonylamino include, but are not limited to, methylcarbonylamino, iso-propylcarbonylamino, and t-butylcarbonylamino. [231 "Alkylcarboxamido" is the radical -C(O)NR 7
R
8 , wherein R 7 is hydrogen or an optionally substituted lower alkyl and R 8 is an optionally substituted lower alkyl. Examples of alkylcarboxamidos are methylcarboxamido, ethylcarboxamido, isopropylcarboxamido, and n-propylcarboxamido. [24] "Alkylsulfonyl" is the radical -S(O) 2 R , wherein R 8 is an optionally substituted lower alkyl. Examples of alkylsulfonyl include, but are not limited to, methylsulfonyl, trifluoromethylsulfonyl, and propylsulfonyl. [25] "Alkylsulfonylamino" is the radical -NR 7
S(O)
2
R
8 , wherein R 7 is hydrogen or an optionally substituted lower alkyl and R 8 is an optionally substituted lower alkyl. Examples of alkylsulfonylamino include, but are not limited to, methylsulfonylamino, propylsulfonylamino, and trifluoromethylsulfonylamino. [26] "Aryl" is the monovalent aromatic carbocyclic radical of one individual aromatic ring or two or three fused rings in which at least one of the fused rings is aromatic. Aryls can be optionally substituted on one or more rings with one or more of halogen, hydroxyl, alkoxy, aryloxy, amino, N-alkylamino, N,N-dialkylamino, alkylcarbamoyl, arylcarbamoyl, aminocarbamoyl, N-alkylaminocarbamoyl, N,N-dialkylaminocarbamoyl, alkylsulfonylamino, arylsulfonylamino, carboxy, carboxyalkyl, N-alkylcarboxamido, N,N-dialkylcarboxamido, alkylthio, alkylsulfinyl, alkylsulfonyl, trifluoromethylsulfonylamino, arylthio, arylsulfinyl, arylsulfonyl, hydroxyalkyl, alkoxyalkyl, aryloxalkyl, aminoalkyl, N-alkylaminoalkyl, N,N-dialkylaminoalkyl, alkylcarbamoylalkyl, arylcarbamoylalkyl, aminocarbanoylalkyl,
N
alkylaminocarbamoylalkyl N,N-dialkylaminocarbamoylalkyl, alkylsulfonylaminoalkyl, arylsulfonylaminoalkyl, alkylcarboxy, alkyicarboxyalkyl, N-alkylcarboxamindoalkyl, N,N-dialkylcarboxamindoalkyl, alkylthioalkyl, alkylsulfinylalkyl, alkylsulfonylalkyl, 6 trifluoromethylsulfonylaminoalkyl, arylthioalkyl, arylsulfinylalkyl, and arylsulfonylalkyl. Examples of aryls are phenyl, naphthyl, tetrahydronaphthyl, indanyl, indanonyl, tetralinyl, tetralonyl, fluorenonyl, phenanthryl, anthryl, and acenaphthyl. [271 "Arylalkoxycarbonyl" or "arylalkyl ester" is the radical -C(O)OR 8 X, wherein R 8 is an optionally substituted lower alkyl and X is an optionally substituted aryl. Examples of .aryloxycarbonyl radicals include, but are not limited to, benzyl ester, phenyl ethyl ester, and dimethylphenyl ester. [28] "Arylalkylcarbamoyl" is the radical -C(O)NHR8X, wherein R8 is an optionally substituted lower alkyl and X is an optionally substituted 'aryl. Examples of arylalkylcarbamoyl include, but are not limited to, benzylcarbamoyl, phenylethylcarbamoyl, and cyanophenylcarbamoyl. [29] "Arylalkylcarbonyl" (or "aralkylcarbonyl") is the radical -C(O)RgX, wherein R 8 is an optionally substituted lower alkyl and X is an optionally substituted aryl. Examples of arylalkylcarbonyl radicals include, but are not limited to, phenylacetyl and fluorophenylacetyl. [30] "Arylaminocarbonyl" (or "arylcarbamoyl") is the radical -C(O)NXX', wherein X is an optionally substituted aryl and X' is hydrogen or an optionally substituted aryl. Examples of arylaminocarbonyl include, but are not limited to, phenylaminocarbonyl, methoxyphenylaminocarbonyl, diphenylaminocarbonyl, and dimethoxyphenyl aminocarbonyl. [31] "Arylaminosulfonyl" is the radical -S(O) 2 NXX', wherein X is an optionally substituted aryl and X' is hydrogen or an optionally substituted aryl. Examples of arylaminosulfonyl include, but are not limited to, phenylaminosulfonyl, methoxyphenylaminosulfonyl, and triiodomethylaminosulfonyl. [32] "Arylcarbonyl" is the radical -C(O)X, wherein X is anoptionally substituted aryl. Examples of arylcarbonyl radicals include, but are not limited to, benzoyl, naphthoyl, and difluorophenylcarbonyl. 7 [33] "Arylcarbonylamino" is the radical -NHC(O)X, wherein X is an optionally substituted aryl. Examples of arylcarbonylamino include, but are not limited to, phenylcarbonylamino, tosylcarbonylamino, and cyanophenylcarbonylamino. [34] "Aryloxy" is -OX, wherein X is an optionally substituted aryl. Examples of aryloxys include phenyloxy, naphthyloxy, tetrahydronaphthyloxy, indanyloxy, indanonyloxy, biphenyloxy, tetralinyloxy, tetralonyloxy, fluorenonyloxy, phenanthryloxy, anthryloxy, and acenaphthyloxy. [35] "Aryloxycarbonyl" or "aryl ester" is the radical -C(O)OX, wherein X is an optionally substituted aryl. Examples of aryloxycarbonyl radicals include, but are not limited to, phenyl ester, naphthyl ester, dimethylphenyl ester, and trifluorophenyl ester. [36] "Arylsulfonyl" is the radical -S(O) 2 X, wherein X is an optionally substituted aryl. Examples of arylsulfonyl include, but. are not limited to, phenylsulfonyl, nitrophenylsulfonyl, methoxyphenylsulfonyl, and 3,4,5-trimethoxyphenylsulfonyl. [37] "Arylsulfonylamino" is the radical --NS(O) 2 X, wherein X is an optionally substituted aryl. Examples of arylsulfonylamino include, but are not limited to, phenylsulfonylamino, naphthylsulfonylamino, 2-butoxyphenylsulfonylanino, 4-chlorophenylsulfonylamino, 2,5-diethoxysulfonylamino, 4-hexyloxyphenylsulfonylainino, 4-methylphenylsulfonyl amino, naphtylsulfonylamino, 4-methoxyphenylsulfonylamino, N-methylphenylsulfonyl amino, and 4-cyanophenylsulfonylamino, phenylsulfonylamino,-4-methylphenylsulfonyl amino, naphtylsulfonylamino. phenylsulfonylamino, and 4-metylphenylsulfonylamino. [38] "Arylsulfonyloxy" is the radical -OS(O) 2 X, wherein X is an optionally substituted aryl. Examples of arylsulfonyloxy include, but are not limited to, benzenesulfonyloxy and 4 chloro-benzenesulfonyloxy. [39] "Cycloalkyl" is a monovalent saturated carbocyclic radical consisting of one or more rings, preferably one, of three to seven carbons per ring and can be optionally substituted with one or more of hydroxyl, alkoxy, aryloxy, amino, N-alkylamino, N,N-dialkylamino, alkylcarbamoyl, arylcarbamoyl, aminocarbamoyl, N-alkylaminocarbamoyl, N,N 8 dialkylaminocarbamoyl, alkylsulfonylamino, arylsulfonylamino, carboxy, carboxyalkyl, N-alkylcarboxamido, NN-dialkylcarboxamido, alkylthio, alkylsulfinyl, alkylsulfonyl, trifluoromethylsulfonylamino, arylthio, arylsulfinyl, arylsulfonyl, hydroxyalkyl, alkoxyalkyl, aryloxalkyl, aminoalkyl, N-alkylaminoalkyl, N,N-dialkylaminoalkyl, alkylcarbamoylalkyl, arylcarbamoylalkyl, aminocarbamoylalkyl, N-' alkylaminocarbamoylalkyl N,N-dialkylaminocarbamoylalkyl, alkylsulfonylaminoalkyl, arylsulfonylaminoalkyl, alkylcarboxy, alkylcarboxyalkyl, N-alkylcarboxamindoalkyl, N,N-dialkylcarboxamindoalkyl, alkylthioalkyl, alkylsulfinylalkyl, alkylsulfonylalkyl, trifluoromethylsulfonylaminoalkyl, arylthioalkyl, arylsulfinylalkyl, and arylsulfonylalkyl. Examples of cycloalkyls are cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, adamantyl, cyclooctyl, cycloheptyl, tetrahydro-naphthalene, methylenecylohexyl, indanyl, indenyl, and fluorenyl. [401 "Cycloalkylcarbonyl" is the radical -C(O)R, wherein R is an optionally substituted cycloalkyl radical. Examples of cycloalkylcarbonyl radicals include, but are not. limited to, cyclobutanoyl, cyclopentanoyl, cyclohexanoyl, and trifluorocyclopentanoyl. [41] "Halogen" includes fluorine, chlorine, bromine, and iodine. [421 "Heteroaryl" is a monovalent aromatic cyclic radical having one or more rings, preferably one to three rings, of four to eight atoms per ring, incorporating one or more heteroatoms selected independently from nitrogen, oxygen, silicon, and sulfur. Heteroaryls can be optionally substituted on one or more rings with one or more of halogen, hydroxyl, alkoxy, aryloxy, amino, N-alkylamino, N,N-dialkylamino, alkylcarbamoyl, arylcarbamoyl, aminocarbamoyl, N-alkylaminocarbamoyl, N,N dialkylaminocarbamoyl, alkylsulfonylamino, arylsulfonylamino, carboxy, carboxyalkyl, N-alkylcarboxamido, NN-dialkylcarboxamido, alkylthio, alkylsulfinyl, alkylsulfonyl, trifluoromethylsulfonylamino, arylthio, arylsulfinyl, arylsulfonyl, hydroxyalkyl, alkoxyalkyl, aryloxalkyl, aminoalkyl, N-alkylaminoalkyl, N,N-dialkylaminoalkyl, alkylcarbamoylalkyl, arylcarbamoylalkyl, aminocarbamoylalkyl, N alkylaminocarbamoylalkyl N,N-dialkylaminocarbamoylalkyl, alkylsulfonylaminoalkyl, arylsulfonylaminoalkyl, alkylcarboxy, alkylcarboxyalkyl, N-alkylcarboxamindoalkyl, 9 N,N-dialkylcarboxamindoalkyl, alkylthioalkyl, alkylsulfinylalkyl, alkylsulfonylalkyl, trifluoromethylsulfonylaminoalkyl, arylthioalkyl, arylsulfinylalkyl, and arylsulfonylalkyl. [43] Representative examples of monocyclic ring system heteroaryls include, but are not limited to, azetidinyl, azepinyl, aziridinyl, diazepinyl, 1,3-dioxolanyl, dioxanyl, dithianyl, furyl, imidazolyl, imidazolinyl, imidazolidinyl, isothiazolyl, isothiazolinyl, isothiazolidinyl, isoxazolyl, isoxazolinyl, isoxazolidinyl, morpholinyl, oxadiazolyl, oxadiazolinyl, oxadiazolidinyl, oxazolyl, oxazolinyl, oxazolidinyl, piperazinyl, piperidinyl, pyranyl, pyrazinyl, pyrazolyl, pyrazolinyl, pyrazolidinyl, pyridyl, pyrimidinyl, pyridazinyl, pyrrolyl, pyrrolinyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, tetrazinyl, tetrazolyl, thiadiazolyl, thiadiazolinyl, thiadiazolidinyl, thiazolyl, thiazolinyl, thiazolidinyl, thiophenyl, thiomorpholinyl, 1,1 dioxidothiomorpholinyl, thiopyranyl, triazinyl, triazolyl, and trithianyl. [44] Bicyclic ring systems include any of the above monocyclic ring systems fused to an aryl group, a cycloalkyl group, or another heteroaryl monocyclic ring system. Representative examples of bicyclic ring systems include but are not limited to, benzimidazolyl, benzothiazolyl, benzothiophenyl, benzoxazolyl, benzofuranyl, benzopyranyl, benzothiopyranyl, benzodioxinyl, 1,3-benzodioxolyl, cinnolinyl, indazolyl, indolyl, indolinyl, indolizinyl, naphthyridinyl, isobenzofuranyl, isobenzothiophenyl, isoindolyl, isoindolinyl, isoquinolyl, phthalazinyl, pyranopyridyl, quinolyl, quinolizinyl, quinoxalinyl, quinazolinyl, tetrahydroisoquinolyl, tetrahydroquinolyl, and thiopyranopyridyl. [45] Tricyclic rings systems include any of the above bicyclic ring systems fused to an aryl group, a cycloalkyl group, or a heteroaryl monocyclic ring system. Representative examples of tricyclic ring systems include,'but are not limited to, acridinyl, carbazolyl, carbolinyl, dibenzofuranyl, dibenzothiophenyl, naphthofuranyl, naphthothiophenyl, oxanthrenyl, phenazinyl, phenoxathiinyl, phenoxazinyl, phenothiazinyl, thianthrenyl, thioxanthenyl, and xanthenyl. 10 [46] "Heteroarylaminocarbonyl" is the radical -C(O)NZZ', wherein Z is an optionally substituted heteroaryl and Z' is hydrogen or an optionally substituted heteroaryl. Examples of heteroarylaminocarbonyl -include, but are not limited to, pyridinylaminocarbonyl, and thienylaminocarbonyl, furanylaminocarbonyl. [47] "Heteroarylaminosulfonyl" is the radical -S(O) 2 N ZZ', wherein Z is an optionally substituted heteroaryl and Z' is hydrogen or an optionally substituted heteroaryl. Examples of heteroarylaminosulfonyl include, but are not limited to, thienylaminosulfonyl, piperidinylaminosulfonyl, furanylaminosulfonyl, and imidazolylaminosulfonyl. [481 "Heteroarylcarbonyl" is the radical -C(O)Z, wherein Z is an optionally substituted heteroaryl. Examples of heteroarylcarbonyl radicals include, but are not limited to, pyridinoyl, 3-methylisoxazoloyl, isoxazoloyl, thienoyl, and furoyl. [491 "Heteroarylsulfonyl" is the radical -S(O) 2 Z, wherein Z is an optionally substituted heteroaryl. Examples of heteroarylsulfonyl include, but are not limited to, thienylsulfonyl, furanylsulfonyl, imidazolylsulfonyl, and N-methylimidazolylsulfonyl. [501 "Heteroarylsulfonyloxy" is the radical - OS(O) 2 Z, wherein Z is an optionally substituted heteroaryl. An examples of hetroarylsulfonyloxy is thienylsulfonyloxy. [51] "Heterocycle" is a saturated or partially unsaturated carbocyclic radical having one, two, or three rings each containing one or more heteroatoms selected independently from nitrogen, oxygen, silicon, and sulfur. A heterocycle can be unsubstituted or substituted on any or all of the rings with one or more of halogen, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, amino, N-alkylamino, N,N-dialkylamino, alkylsulfonylamino, arylsulfonyl amino, alkylcarbamoyl, arylcarbamoyl, aminocarbamoyl,
N
alkylaminocarbamoyl, N,N-dialkylaminocarbamoyl, carboxy, alkylcarboxy,
N
alkylcarboxamido, N,N-dialkylcarboxamido, alkylthio, alkylsulfinyl, alkylsulfonyl, trifluoromethylsulfonylamino, arylthio, arylsulfinyl, arylsulfonyl, carboxyalkyl, hydroxyalkyl, alkoxyalkyl, aryloxalkyl, aminoalkyl, N-alkylaminoalkyl,
N,N
dialkylaminoalkyl, alkylcarbamoylalkyl, arylcarbamoylalkyl, aminocarbamoylalkyl,
N
11 alkylaminocarbamoylalkyl N,N-dialkylaminocarbamoylalkyl, alkylsulfonylaminoalkyl, arylsulfonylaminoalkyl, alkylcarboxyalkyl, N-alkylcarboxamindoalkyl,
N,N
dialkylcarboxamindoalkyl, alkylthioalkyl, alkylsulfinylalkyl, alkylsulfonylalkyl, trihaloalkylsulfonylaninoalkyl, arylthioalkyl, arylsulfinylalkyl, and arylsulfonylalkyl. Examples of heterocycles include piperazinyl, piperidinyl, pyrrolidinyl, morpholinyl, thiamorpholinyl, pyrrolyl, phthalamide, succinamide, and maleimide. [52] "Heterocyclylcarbonyl" (or "heterocyclocarbonyl") is the radical -C(O)M', wherein M' is an optionally substituted heterocycle.. Examples of heterocyclylcarbonyl include, but are not limited to, piperazinoyl, morpholinoyl, and pyrrolindinoyl. [53] "Heterocyclylsulfonyl" is the radical -S(O) 2 Z', wherein M' is an optionally substituted heterocycle. Examples of heterocyclylsulfonyl include, but are not limited 'to, piperidinylsulfonyl and piperazinylsulfonyl. [54] "Heterocyclylsulfonyloxy" is the radical - OS(O) 2 M', wherein M' is an optionally substituted heterocycle. Examples of heterocyclylsulfohyloxy include, but are not limited to, 3 ,5,dimethyl-isoxazolesulfonyloxy and pyrrolidinylsulfonyloxy. Compounds [55] This invention provides compounds which inhibit tyrosine kinases, particularly Tec (e.g., ITK, BTK), Src (Src, Lck, etc.) and EGFR kinases (e.g., EGFR1, Her 2, Her 4), and Jak kinase (e.g., Jak3), having structures that exploit a discrete mechanistic rationale described herein. This mechanism provides for the utilization of the kinase catalytic machinery, described in the ITK crystallographic structures as the acid-base pair residues Lys391 and Asp500 (herein referred to as the "catalytic dyad"), to trigger a transformation that activates the proposed inhibitory compounds within the enzyme active site. This transformation involves the elimination of a leaving group, resulting in the in situ formation of an electrophilic intermediate capable of forming a covalent adduct with an'active site cysteine residue thereby irreversibly inhibiting the function of the target enzyme. This cysteine residue is identifiable as Cys442 in the ITK crystallographic structure. The group of kinases with the above described triad, including 12 ITK, BTK,.BMX, Tec, TXK, BLK, EGFr, Her 2, Her 4 and JAK3, will be referred to as the DKC triad kinases. Various embodiments of the invention relate to this group, its possible sub-groupings, and to its individual members. [56] It is known that several compounds, typically containing electrophilic Michael acceptors, form covalent adducts with enzymatic nucleophiles present in the active site to irreversibly inhibit the target enzyme (Slichenmeyer, W.J.; Elliott, W.C.; Fry, D.W. Semin. Oncol. 2001, 28, 80-85; Shimamura, T.; Ji, H.; Minami, Y.; Thomas, R. K.; Lowell, A.M.; Sha, K.; Greulich, H.; Glatt, K.A.; Meyerson, M.; Shapiro, I.; Wong, K.
K. Cancer Res. 2006, 66, 6487-6491). However, the compounds described in this invention are unique in that the transformation that forms the electrophilic intermediate takes place preferentially in situ, i.e. within the enzyme active site. Outside of an appropriate active site, these compounds are much less likely to undergo beta-elimination and form adducts with other proteins. The compounds described within must first bind in the active site of the target kinase and achieve a specific conformational geometry with respect to the relevant catalytic residues in order to effectively trigger elimination of the leaving group, thereby unmasking the adduct-forming intermediate. This intermediate forms a covalent, irreversible adduct with the proximal active site cysteine residue. In some embodiments the reaction proceeds stepwise; in other embodiments it is concerted. In preferred embodiments additional portions of the inhibitor molecule interact with other portions of the kinase, particularly in the active site, to promote favorable binding affinity and positioning. Such interactions contribute to the specificity of various inhibitors so that some inhibitors are inhibit a single kinase whereas others inhibit multiple kinases with similar or different IC 5 0 s. To our.knowledge, this is the first example of an in situ formation of an active inhibitor in a kinase active site. Compound interaction with the kinase domain [57] Without specifying the kinetics of the reaction, the inhibition of the target kinase goes through the following sequence of steps to form the adduct with the inhibitory compounds: 13 (1) The catalytic lysine N-H is positioned within hydrogen bonding distance (approximately 1.8 - 4.0 Angstroms) of a hydrogen bond acceptor Y in the compound that exists in the form of a C=Y (Y=O, S, NOR) functionality. Polarization of the C=Y bond results in increasing the acidity of the proton (HA) at a carbon atom alpha to the C=Y group. (2) Acting as a base, the aspartate of the catalytic dyad extracts the acidic proton HA, leaving behind a conjugated carbanion that forms for Y 0, an enol, H-bonded enolate through standard electronic rearrangement. For Y = S, it would form a thioenol or H-bonded thioenolate, and for Y = NOR, it would form an alkoxy (R = alkyl), aryloxy (R=aryl) or hydroxy (R=H) enamine. (3) The formation of the enol/thioenol/enamine facilitates the elimination of the leaving group attached at a carbon beta to C=Y, through a process known as "P-elimination." The leaving group, attached to the compound through protonatable heteroatom Z, may optionally be additionally tethered to the rest of the compound. (4) Being a strong nucleophilic species, the sulfhydryl group of the neighboring cysteine residue reacts with the newly formed electrophilic elimination product. This addition reaction (thioalkylation) forms the covalent adduct to the kinase resulting in its irreversible inhibition and abrogation of activity. [58] The inhibitory activity of this class of compounds toward select kinases is dependent on their ability to bind effectively in proximity to the appropriate calalytic environment, the existence of a polarizable C=Y group (C=O in formula (I), below) with appropriate reactivity and an adjacent alpha proton to allow elimination of the beta leaving group. [59] In turn, the elimination process that generates a reactive electrophilic species requires removal of the abstractable alpha proton that is facilitiated by adequate positioning of the C=Y group in the catalytic environment. The generated electrophilic Michael acceptor, in turn is required to be positioned within reactive distance of the key cysteine residue. 14 The appropriate positioning of the abstractable proton in the kinase binding site is achieved through pharmacophoric elements that include: (i) a C=Y moiety that serves the dual purpose of polarizing the proximal C-H bond of the abstractable proton, and hydrogen bonding to the lysine residue of the catalytic pair; (ii) a hydrophobic aryl or heteroaryl group that interacts with specific hydrophobic residues in the binding site at an approximate distance of 3-5 A from Y, (iii) several (one to 3) hydrophilic pharmacophores that interact with the backbone in the hinge region, (vi) a carbon atom in the beta position from the C=Y carbon atom, that is positioned within reactive distance of the sulfhydryl group of the relevant cysteine as explained below. [60] The effective "reactive distance" to the cysteine sulfhydryl group as stated above is observed in the range of about 3-10 A using computational design methods that test the binding of inhibitors to the ITK ATP binding site, wherein the enzyme is maintained in a fixed conformation. While a distance of 10 A in a rigid system would be too far to effect a chemical reaction, the enzymatic nucleophilic moiety and the inhibitor's electrophilic moiety can readily be brought together through a series of low energy barrier rotations around the flexible inhibitor bonds as well as the cysteinyl side chain. Overall global conformational changes, common to kinase systems, cannot be ruled out either but are not readily measurable. Such conformational changes, which can be envisioned by computational predictions, are adequate in bringing the two reactive pieces in close enough proximity to effect covalent bond formation. 15 [611 Compounds according to the invention have the structural formula: 0 R4 R3 Ar N R R 5 (I) wherein: Ar is optionally substituted aryl or optionally substituted heteroaryl;
R
3 , R 4 , R', and R 6 are independently hydrogen or optionally substituted C-C 6 alkyl; and R' and R 2 (a) are independently hydrogen, optionally substituted CI-C 6 alkyl, piperidine, or furanyl; or (b) are taken together with the nitrogen atom to which they are attached to form (i) a 5- to 7-membered optionally substituted aryl, (ii) a 5:- to 7-membered optionally substituted heteroaryl, or (iii) a 5- to 7-membered optionally substituted heterocycle which may be unfused or fused to an optionally substituted aryl. [621 In some embodiments Ar is selected from the group consisting of: D Cand D ,wherein A, B, E, and Q are independently CH, 0, or N; and D and D' are independently CH 2 , NH, 0, or S. 16 [631 In other embodiments Ar is selected from the group consisting-of: ; ; CN - N H N ;and H 1641 Examples of 5- to 7-membered heterocycles include: NN (H2) RN R 0 0 N - R' L kN 12 K & .1 W-R R R 4HH 2 )- KCHNR1 and wherein: G is N, CH, or S; Gis NH, CH, or S; n = 0-2; R' and R 2 are as defined above; and 17 R7 is hydrogen, optionally substituted C 1
-C
6 alkyl, optionally substituted aryl, or optionally substituted heteroaryl. [651 Preferred 5- to 7-membered heterocycles are piperazinyl, piperidinyl, pyrrolidinyl, and morpholinyl. Preferred substituents for piperazinyl are C 1
-C
6 alkyl, dialkyl Ci-C 6 aminoalkyl, aryl, aralkyl, cycloalkyl, and cycloalkyl-alkyl. Preferred substituents for piperidinyl are C 1
-C
6 alkyl and aralkyl. In some embodiments piperidinyl is benzofused to form isoquinolinyl. Preferred substituents for pyrrolidinyl are CI-C 6 alkyl, aryl, and aralkyl. In some embodiments pyrrolidinyl is benzofused to form isoindolyl. Preferred substituents for morpholinyl are CI-C 6 alkyl and arylalkyl. [661 Some compounds have the structural formula: 0 Rti3
R
9 N (II) wherein:
R
3 , R 4 , R 5 , and R 6 are as defined above;
R
10 H R
R
9 is selected from, O 0 NN Nc 18 - ~ NN 0 0 18 HN N N H N H
H
2 N H N HHNCO N O H N0 N
OCH
3 H OCH 3 O NN 'o 1? N 00 HH H3CO1 N N HN 7 HN
H
2 N I N NS N I 0 S
NH
2 HN HN N NN N KNH H SH-/O H 2 N 'O NH HIN ,and ;and R'0 is hydrogen, -OH-, -COOH, -CONH2, or -NCO, wherein if R 9 is naphthyl; then R 5 and R 6 are not both methyl. 20 [67] Examples of these compounds include: N No 00 H N ~NN 0 ~ 6 0 o 0 0 0 N NN NH 0 00 21 0 N HO HO HNN N H2N O 6 NO HN N ~IN 00 S 0 N HNN S0n
NHN
00 0N 0 0NC H OCH 22 NN 0 'N H OCH 3 H0 0 0 0 < I N' </ 0 0 0 H N N 0 14" N 0o S \ I ~ c20 0
H
2 N - 12 HN' IN~ S c /-S S> 23 0 NAN HN+:) NH0 0 00 N N 0& IHN 0 0 NH 00 H H HN \ NF0 H2N--t-'O 24 [68] Other compounds have the structural formula: 0 Rh3 R12 N R13 wherein:
R
3 , R 4 , R 5 , and R 6 are as defined above; R" and R' 2 are independently selected from hydrogen, -OCH 3 , halogen, -NO 2 , -CN, -CF 3 , -NCOR' (wherein R' is hydrogen or CI-C 4 alkyl), phenyloxy, -OCF 3 , -NR'R" (wherein R' and R" are independently hydrogen or CI-C 4 alkyl), CI-C 4 alkyl, Ci
C
4 alkoxy, and -SO 2 R'(wherein R' is hydrogen or CI-C 4 alkyl); and NN R1 3 is hydrogen, Ci-C 4 alkyl, NH 2 , NH 2 , NH 2 HN _ O N.'N NH2 , and HN with the proviso that formula (III) does not include the following compounds: 25 0 F Oiii 0 H0 N 0 ~C~0 c' N F 0 0 FWC O a ON o oF F II 0 0 03,* 0 0 0 0' 0-~ o 0 0 0 1 Q.O F2 CF 0 0 0 aCF 0 26 0 0 0 Nc I N 0 Q FF o F 0 N and 00 F One example of a compound of formula (III) is F [69] Some compounds of the invention have the structural formula: 0 NN
R
2 0 wherein R' and R 2 are as defined above, with the exception of 27 [70] Examples of such compounds include those of with the following structural formulae: o 0 NNN N K , N K,an 00 in which: GG is hydrogen, dimethylamninoalkyl, aryl, C 1
-C
6 alkyl, cyclohexylalkyl, 0 NN pyridine, -COCF 3 ; -CONR'R", or.; J is hydrogen, aralkyl, C 1
-C
6 alkyl, -CNHCOOR', or NR'R"; K is hydrogen, pyridine, aryl, --COOH, -CONR'R", -COH, or -CNR'R"; L is hydrogen or alkyloxy; and
R
2 is as defined above. 28.
171] Other compounds of formula (IV) include: 0 0 N NN N CFHO NN 0 0 00 NNH. * OH 0 0 OH O 0 K-K 1 N * /H 02 0 NHMO NrO N OS O N N H N0 00 N I ,and 0 OQNNh Nb [721 Other compounds have the structural formula: 0 R4F3 R. R- -(V) 30 wherein R 3 , R 4 , R 5 , R 6 , and R' 0 are as defined above. [731 Other compounds have the structural formula: 0 Rj 3 R14 R 04 R ( V I ) wherein: R', R R, and R6 are as defined above; and R1 4 is hydrogen or =0; and D is CH or NH, with the exception of: 0 0 0y NQ 0 N~ 0 and [741 Other compounds have the structural formula: O Ra 2 O (VII) wherein:
R
3 , R 4 , R', and R6 are as defined above; and 31 R' and R 2 are independently hydrogen, CI-C 4 alkyl, (wherein R1 5 is halogen or Ci-C 4 alkyl and Ri 6 is CI-C 4 alkyl), or R' and R 2 together with the SN A nitrogen to which they are attached form an aryl group selected from N R 1 7 I2O R 18 7 1 (wherein R' 7 and R" are independently hydrogen or -OCH 3 ), N
R
1 N R 2 (wherein R' and R 2 are independently hydrogen or CI-C 4 alkyl), N R A N R 2 (n = 1-4), phenyl-CI-C 4 alkyl (optionally substituted with halogen), 0 O : N N with the exception of [751 Other compounds have the structural formula: 0 R 3 O 1 , O N R 5 - 0( V I I I ) wherein R', R 4 , R 5 , and R 6 are as defined above. 32 [761 Other compounds have the structural formula: O R4Ra 12 R- R5 R 2 (IX) wherein R 3 , R 4 , R 5 , and R 6 are as defined above and wherein R 1 is hydrogen and R 2 is n0 RO NH N , wherein R1 9 is selected from hydrogen and ; or N ' R 20 R' and R 2 together with the nitrogen to which they are attached are H0NRo 'NR / R ~OH K .AK I 9 18 K N N NNN and A is N or 0;
R
20 is phenyl-C 1
-C
4 alkyl optionally substituted with one or more halogens, hydrogen, R2 N C 1
-C
4 alkyl, amino- C 1
-C
4 alkyl, , " ; 33 R1 7 and R 8 are independently hydrogen or -OCH 3 ; H N H NO
R
2 ' is -CONR'R", -COR', , N N CF 3 , and ; and R' and R" are independently selected from hydrogen and CI-C 4 alkyl. [771 In other embodiments compounds have the structural formula: o R' 3 R22(X) wherein R 3 , R 4 , R 5 , and R 6 are as defined above and wherein R 2 2 is selected from hydrogen, CI-C 4 alkyl, -NR'R", -COH, -COOH, -CNR'R", and -CONHR', wherein R' and R" are as defined above. [78] In other embodiments compounds have the structural formula: 0 R4s3 R 2 3 (X) 34 wherein R 3 , R 4 , Rs, R6, G, and G' are as defined above; and R 3 is hydrogen, -NR'R" C 1 C 4 linear alkyl, Ci-C 4 alkyl, phenyl-Cl-C 4 alkyl, -CONH 2 , and -CO R'R", wherein R' and R" are as defined above. . [791 In other embodiments compounds have the structural formula: 0 Rj3 (XII) R3, R, R5, and R 6 are as defined above and wherein R 24 is H [801 In other embodiments compounds have the structural formula: -L (XIII) O Rh3 R5 N wherein L is ,and wherein R 3 , R 4 , R5, and R 6 are as defined above. 35 [811 Some compounds have the structural formula: U W
-
0 V R25 N (XIV) wherein T, U, V, and W independently are selected from hydrogen; halogen; -0; C-C 3 alkyl; and C-C 3 alkyloxy; and wherein R 2 5 is hydrogen or C-C 3 alkyl. Representative compounds include: -- -. 0 0 0 -0 0 ~~N and N [82] Other compounds have the structural formula: O N W NH T //I- N U\ / u v (XV), 36 wherein T, U, V, and W independently are selected from hydrogen; halogen; -0; CI-C 3 alkyl; and CI-C 3 alkyloxy; and wherein R 8 is hydrogen or CI-C 3 alkyl. [83] Still other compounds have the structural formula: T W NH O N C NU V O- (XVI), wherein T, U, V, and W independently are selected from hydrogen; halogen; -0; Ci-C 3 alkyl; and C-C 3 alkyloxy; and wherein R 8 is hydrogen or CI-C 3 alkyl. [84] Other compounds have the structural fonnula: 0 O (XVII) 0 wherein D is S, 0, or NH; i.e., 0 0 and H 37 [85] Other compounds of the invention include those with the structural formula: 0 cD -(XVIII), 0 cch-o wherein D is defined above; i.e., S 0 0 Q* NN)I l~ ,and H [86] Other compounds of the invention include those with the structural formula: .0
.
N I G 0 (XIX), 0 0 NH N N wherein G' is NH or CH; i.e., 0 or 0 38 [87] The invention also includes the compounds identified in Examples 15 and 16. [88] The compounds of the present invention may have asymmetric centers and may occur as racemates, stereoisomers, and tautomers. The invention includes all possible racemates, tautomers, stereoisomers, and mixtures thereof [89] Suitable methods of preparing compounds of the invention are illustrated by the representative examples provided below. Starting materials are known compounds and can be obtained by standard procedures of organic chemistry. Provisos for compound claims [90] Compounds of the invention preferably do not have one or more of the following activities: vasodilator, hypotensive, bradycardiac, anti-depressant, anti-arrhythmic, anti arteriosclerotic, serum cholesterol lowering, triglyceride level lowering, neuroleptic, anti inflammatory, tranquilizing, anti-convulsant, anesthetic, muscle relaxing, anti-fungal, anti-bacterial, insecticidal, fumigant, anti-parasitic, central nervous system depressant, antagonization of sedation, antipollakiurea, antihistamine, anti-allergy, bronchodilating, analgesic, spasmolytic, muscarinic antagonist, preventing or decreasing production of abnormally phosphorylated paired helical filament epitopes associated with Alzheimer's Disease, hypolipidemic, male anti-fertility, anti-sporicidal, inhibition of nitric oxide production, or central nervous system stimulant activities. [91] To the extent any of the following compounds are not novel, Applicants reserve the right to present compound and/or composition claims which include a proviso excluding the compounds and/or their pharmaceutically acceptable salts from the scope of the claims: a. compounds having the structural formula: 0 Y, N I-I
R
2 39 wherein n is 0, 1, 2, or 3 and R' and R 2 together with the nitrogen atom to N N N which they are attached are , , or and Y is alkyl, halogen, halogenoalkyl, alkyoxy, akylthio, halogenoalkyloxy, halogenoalkylthio, cycloalkyl, or a cyane radical; b. compounds of formula (1) in which Ar is phenyl, if R 3 , R 4 , R5, and R 6 are each hydrogen, and R' and R 2 together form a ring with the nitrogen atom to which they are attached c. compounds having the structural fohnula formula: Il Ph -C-CH-CH 2 -Am Alk in which Ph is an optionally substituted monocyclic carbocyclic aryl radical, Alk is CI-C 3 lower alkyl, and Am is a tertiary amino group, salts, N-oxides, or quaternary ammonium derivatives thereof; d. compounds having the structural formula: 0 Ph 1 -- C-CH-CH 2 -N Ph 2 in which Ph, and Ph 2 are monocyclic carboxylic aryl radicals and the acid addition salts thereof; 40 e. compounds having the structural formula:
H
RR-CO-C-C--N(XX) I H2 RR, in which RR is selected from the group consisting of aliphatic, aromatic, and araliphatic radicals; RR' is selected from the group consisting of hydrogen, aliphatic, aromatic, and araliphatic radicals; -N(XX) is the residue of a secondary amine selected from the group consisting of dialkylamine and dialkylamines; f. compounds having the structural formula: 0 N R 12 R wherein R' and R 2 are as defined in formula (I), including the compound 0 N 1 i g. compounds having the structural formula: 0 No R3o wherein R 30 is an ethyl-, propyl-, isopropyl-, butyl-, or isobutyl group or a cycloalkyl group having 5-7 carbon atoms; 41 h. compounds having the structural formula: 0 I 14 M M in which M 2 is hydrogen, halogen, or C 1
-C
2 alkoxy, M1 is hydrogen or halogen, and M' and M 4 are lower alkyl or, taken together with the nitrogen atom to which they are attached, (a) are a heterocyclic amino group or an N-lower alkyl quaternary heterocyclic ammonium group or (b) a tri-lower alkyl-ammonium; i. compounds having the structural formula: 0 M 5-Me or a picrate salt thereof, wherein M 5 is a simple or substituted aryl group and M6 is a simple or substituted amino group; j. compounds having the structural formula: 0 M7)< N MI N in which M 7 is thienyl, phenyl or substituted phenyl; 42 k. compounds having the structural formula: OX2 0 x4 x'O - ox 3 in which each of X1, X 2 , and X 3 are independently hydrogen or an alkyl group, and each of X 5 and X 4 are independently hydrogen or an alkyl group or, together with the nitrogen atom to which they are attached, form a heterocyclic group with 5, 6, or 7 ring atoms, optionally containing, in addition to N, a further heteroatom selected from N, S, and 0; 1. compounds of formula (II) in which R 9 is phenyl and R 3 , R 4 , R 5 , and R 6 are each hydrogen; m. compounds having the structural formula: 0 S NX 6 in which X 6 forms with the nitrogen atom pyrrolidine, piperidine, morpholine, hexamethyleneimine, or 3-azabicyclo-3,2,2 nonane, including 0 S Q the compound 43 n. compounds having the structural formula: 0 N in which X 7 is hydrogen or fluorine; X 8 is N(X 9 )phenyl (wherein the phenyl 'is optionally monosubstituted with C 1
-C
8 alkoxy, C-Cs alkyl, trifluoromethyl, or halogen), -C(OH)(X 9 ) phenyl (wherein the phenyl is optionally monosubstituted with C-C 8 alkoxy, C-C 8 alkyl, trifluoromethyl, or halogen), or phenyl (wherein the phenyl is optionally monosubstituted with C-C 8 alkoxy, C 1
-C
8 alkyl, trifluoromethyl, or halogen); and X 9 is hydrogen, C-C 8 alkyl, or lower alkanoyl; o. compounds having the structural formula: 0 S N - I kIo wherein X 9 and X 10 each designate a saturated or unsaturated aliphatic hydrocarbon having I to 4 carbon atoms or, together with the nitrogen to which they are attached, form a heterocyclic radical selected from pyrrolidino, piperidine, perhydroazepino, and morpholino; p. compounds having the structural formula: 44 0 NQ in which X" is C 2
-C
3 alkyl; q. compounds having the structural formula: O N NI~I x 11 in which X" is hydrogen, halogen, CI-C 4 alkoxy,. nitro, or Ci-C4 secondary amine; X1 2 is (CH2)OX1 3 ; n is 2 or 3; and X1 3 is Ci-C 4 alkoxyphenyl, nitrophenyl, trifluoromethylphenyl, or phenyl disubstituted with two halogens, two Ci-C 4 alkyls, halogen and nitro, halogen and C 1 -C4 alkyl, halogen and CI-C 4 alkoxy, or C,-C 4 alkoxy and Ci-C 4 alkoyl; r. compounds having the structural formula: 0 x 14 X15 x17 XX8 x 16 in which X1 4 , X 15 , and X 16 are independently hydrogen, halogen, CI-C 4 alkyl, halogeno- Ci-C 4 alkyl, CI-C 4 alkoxy, or a cycloalkyl group having 3-8 carbon atoms and two of X1 4 , X' 5 , and X1 6 may combine to form methylenedioxy or ethyleneoxy; X 8 is hydrogen or C 1
-C
4 alkyl; and X1 7 is pyrrolidinyl-, piperidinyl, morpholinyl-, or azepinyl; 45 s. compounds having the structural formula: 0 Ar ~N-X- X 21 S0 3 H 120 Ar denotes an aryl radical; and X19 and X20 (a) are both Ci-C6 alkyl or (b) together with the N atom form the remaining members of a saturated heterocyclic radical and X21 is -OH, C I-C6 alkyl, or aryl; t. compounds hav ing the structural formula: O N 0 N'R x23_ 3 12 R R wherein R and R2 ica;endently represent an alkyl radical; or R and R2, together with the nitrogen atom to which they are bonded complete an optionally substituted heterocyclic radical of the formula /- No or /-0 ; R 3 is hydrogen or CI-C4 alkyl; and X2, X3, and X24 are independently CI-C4 alkyl, halogen, or a halogeno- CI-C4 alkyl, CI-C4 alkoxy, alkylthio, halogeno- CI-C4 alkoxy, halogeno- C1-C4 alkylthio, cycloalkyl 3 to 7 carbon atoms, or cyano; u. compounds having the structural formula: 0 Ar x25 -R46 244 wherein Ar is non-substituted aryl or aryl substituted with a hydroxyl group, lower alkoxy group or halogen, or non-substituted benzo[b]thienyl group or benzo[b]thienyl group substituted by hydroxyl group, lower alkyl group, lower alkoxy group, aryl group or halogen; R 5 is hydrogen or CI-C4 alkyl; and X 5 is a group other than piperidine; v. compounds having the structural formula: 0 L 6 2L LD; L 3 L2
L
4 L wherein L' and L2 are independently halogen or alky]; L 6 and LJ are independently hydrogen or alkyl; and L 3 and L 4 are independently hydrogen or an aliphatic group or combine together with the nitrogen to which they are attached to form a ring; w. compounds of formula (I), (IV), (VI), (VII), (IX), and (XI) in which if R 3 and R 4 N2 N Ar are hydrogen, then R is not La- or NL A wherein L 8 is a carbonyl, sulfonyl, methylene, or methylene substituted with optionally substituted phenyl; and Ar is an aryl group; x. compounds having the structural formula: 0 T3 Ar N 4 4 47 in whicli T' is 0, S, or NT 7 ; T 7 is hydrogen, CI-C 4 alkyl, and
CH
2
CH
2 COAri; T is hydrogen, CI-C 6 alkyl, or T 6 and a substituent on the aryl group together represent CH 2 , CH 2
CH
2 , CH 2 0, or CH 2 S to form a five or six membered ring where the ring is optionally substituted with
CI-C
6 alkyl or phenyl; Ts is hydrogen, CI-C 6 alkyl, or optionally substituted phenyl; T 2 , T 3 , and T4 are independently hydrogen or CI-C 6 alkyl; and Ar and Arl are aryl or optionally substituted phenyl; and y. the following compounds: 0 0
H
3 CO N F N 0
CH
3 N F Nand 0 No
H
3 CO Pharmaceutical Preparations [921 Compounds of the invention can be formulated as pharmaceuticals using methods well known in the art. Pharmaceutical formulations of the invention typically comprise at least one compound of the invention mixed with a carrier, diluted with a diluent, and/or enclosed or encapsulated by an ingestible carrier in the form of a capsule, sachet, cachet, paper or other container or by a disposable container such as an ampoule. 48 [931 A carrier or diluent can be a solid, semi-solid or liquid material. Some examples of diluents or carriers which may be employed in the pharmaceutical compositions of the present invention are lactose, dextrose, sucrose, sorbitol, mannitol, propylene glycol, liquid paraffin, white soft paraffin, kaolin, microcrystalline cellulose, calcium silicate, silica polyvinylpyrrolidone, cetostearyl alcohol, starch, gum acacia, calcium phosphate, cocoa butter, oil of theobroma, arachis oil, alginates, tragacanth, gelatin, methyl cellulose, polyoxyethylene sorbitan monolaurate, ethyl lactate, propylhydroxybenzoate, sorbitan trioleate, sorbitan sesquioleate and oleyl alcohol. [94] Pharmaceutical compositions of the invention can be manufactured by methods well known in the art, including conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping, or lyophilizing processes. [95] For injection, the agents of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as acetate, Hanks's solution, Ringer's solution, or physiological saline buffer. Preferably the solutions are sterile and non-pyrogenic. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art. [96] For oral administration, the active compound(s) can be combined with pharmaceutically acceptable carriers which enable the compound(s) to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like. Fillers can be used, such as gelatin, sugars (e.g., lactose, sucrose, mannitol, or sorbitol); cellulose preparations .(e.g., maize starch, wheat starch, rice starch, potato starch, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose); and/or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate. [971 Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, 49 and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses. [98] Pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compound(s) may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration preferably are in dosages suitable for such administration. [99] For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner. [100] For administration by inhalation, pharmaceutical preparations of the invention can be delivered in the form of an aerosol sprays from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide, or other suitable gas. If desired, a valve can be used to deliver a metered amount. Capsules and cartridges of e.g., gelatin for use in an inhaler or insufflator, may be formulated containing a powder mix of a compound and a suitable powder base such as lactose or starch. [101] Compounds of the invention can be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection can be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions can take such forms as suspensions, solutions or - emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. [1021 Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds. Additionally, suspensions of the active compounds may be .50 prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. [1031 Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use. [104] The compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides. [1051 Compounds of the invention typically are soluble and stable in 50 mM acetate at a concentration of 10 mg/ml or above, and can be delivered intraperitoneally and orally in this buffer. Some compounds are soluble in hydroxypropyl-b-cyclodextrin (HBPCD, 3 5%), and can be delivered intraperitoneally and orally in this solvent. For intravenous delivery, compounds can be suspended or dissolved in 5% mannitol. [106] In addition to the formulations described previously, the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as. sparingly soluble derivatives, for example, as a sparingly soluble salt. [107] The pharmaceutical compositions also may comprise suitable solid or gel phase carriers or excipients. Examples of such carriers or excipients include but are not limited to, calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols. 51 [1081 In addition to the common dosage forms set out above, the compounds of the present invention may also be administered by controlled release means and/or delivery devices including ALZET@ osmotic pumps which are available from Alza Corporation. Suitable delivery devices are described in U.S. Pat. Nos. 3,845,770; 3,916,899; 3,536,809; 3,598,123; 3,944,064 and 4,008,719. Therapeutic Methods [1091 The identified compounds can be administered to a human patient, either alone or in pharmaceutical compositions where they. are mixed with suitable carriers or excipient(s) at doses to treat or ameliorate blood-related cancers (e.g., lymphomas and leukemias) and. autoimmune disorders. Reduction of intracellular kinase activity also is useful to suppress the immune system of transplant patients prior to, during, and/or after transplant. [1101 Lymphomas are malignant growths of B. or T cells in the lymphatic system, including Hodgkin's lymphoma and non-Hodgkin's lymphoma. Non-Hodgkin's lymphomas include cutaneous T cell lymphomas (e.g., Sezary syndrome and Mycosis fungoides), diffuse large cell lymphoma, HTLV-1 associated T cell. lymphoma, nodal peripheral T cell lymphoma, extranodal peripheral T cell lymphoma, central nervous system lymphoma, and AIDS-related lymphoma. [1111 Leukemias include acute and chronic types of both lymphocytic and myelogenous leukemia (e.g, acute lymphocytic or lymphoblastic leukemia, acute myelogenous leukemia, acute myeloid leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, T cell prolymphocytic leukemia, adult T cell leukemia, and hairy cell leukemia). [1121 Autoimmune disorders include systemic lupus erythematosus, anti-phospholipid antibody syndrome, multiple sclerosis, ulcerative colitis, Crohn's disease, rheumatoid arthritis, asthma, Hashimoto's thyroiditis, Reiter's syndrome, Sjogren's syndrome, Guillain-Barr6 syndrome, myasthenia gravis, large vessel vasculitis, medium vessel vasculitis, 52 polyarteritis nodosa, pemphigus vulgaris, scleroderma, Goodpasture's syndrome, glomerulonephritis, primary biliary cirrhosis, Grave's disease, membranous nephropathy, autoimmune hepatitis, celiac sprue, Addison's disease, polymyositis, dermatomyositis, monoclonal gammopathy, Factor VIII deficiency, cryoglobulinemia, peripheral neuropathy, IgM polyneuropathy, chronic neuropathy, autoimmune hemolytic anemia, autoimmune thrombocytopenic purpura, pernicious anemia, ankylosing spondylitis, vasculitis, inflammatory bowel disease, and type I diabetes mellitus. The autoimmune disease may involve a secretory cell, such as a T lymphocyte, B lymphocyte, Mast cell, or dendritic cell. Compounds of the invention also can be used to treat patients who undergo protein replacement therapies and who develop antibodies to the replacement. Routes of administration [113] Pharmaceutical preparations of the invention can be administered locally or systemically. Suitable routes of administration include oral, pulmonary, rectal, transmucosal, intestinal, parenteral (including intramuscular, subcutaneous, intramedullary routes), intranodal, intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, intraocular, transdermal, topical, and vaginal routes. As described in more detail above, dosage forms include, but are not limited to, tablets, troches, dispersions, suspensions, suppositories, solutions, capsules, creams, patches, minipumps and the like. Targeted delivery systems also can be used (for example, a liposome coated with target-specific antibody). Dosage [1141 A pharmaceutical composition of the invention comprises at least one active ingredient in a therapeutically effective amount. A "therapeutically effective dose" is the amount of an active agent which, when administered to a patient, results in a measurable improvement in a characteristic of the disease being treated (e.g., improved laboratory values, retarded development of a symptom, reduced severity of a symptom, improved levels of a biological marker such as CD25a or IL2). The improvement can be evident after a single administration of the therapeutically effective dose. More usually - multiple administrations are utilized in order to achieve or maintain optimal effect. In preferred 53 embodiments frequency of administration can range from twice a month to once a week to several times a day, for example 1-4 times a day. In alternative embodiments administration can be by time-release formulations, or extended or continuous infusions. The frequency of administration can be selected to achieve a systemic or local concentration at or above some predetermined level for a period of time. The period of time can be all or a substantial portion of the interval between administrations or comprise the period of time-release or infusion. In some embodiments, the treatment schedule can require that a concentration of the compound be maintained for a period of time (e.g., several days or a week) and then allowed to decay by ceasing administration for a period of time (e.g., 1, 2, 3, or 4 weeks). 11151 Determination of therapeutically effective amounts is well within the capability of those skilled in the art. A therapeutically effective dose initially can be estimated from in vitro enzyme assays, cell culture assays and/or animal models. For example, a dose can be formulated in an animal model to achieve a circulating concentration range that includes the IC 50 as determined in an in vitro enzyme assay or in a cell culture (i.e., the concentration of the test compound which achieves a half-maximal inhibition of ITK or BTK activity). Such information can be used to more accurately determine useful doses in humans. [116] Appropriate animal models for the relevant diseases are known in the art. See, e.g., Exp Hematol. 34, 284-88, 2006 (aggressive systemic mastocytosis and mast cell leukemia); Leuk. Lymphoma. 47, 521-29, 2006 (acute myeloid leukemia); Leuk. Lymphoma. 7, 79 86, 1992 (disseminated human B-lineage acute lymphoblastic leukemia and non Hodgkins lymphoma); J. Virol. 79, 9449-57, 2006 (adult T-cell leukemia); Neoplasia 7, 984-91, 2005 (lymphoma); Oligonucleotides 15, 85-93, 005 (lymphoma); Transfus. Apher. Sci. 32, 197-203, 2005 (cutaneous T cell lymphoma); Nature 17, 254-56, 1991 (follicular lymphoma and diffuse large cell lymphoma); Cell. Mol. Immunol. 2, 461-65, 2005 (myasthenia gravis); Proc. Natl. Acad. Sci. USA 102, 11823-28, 2005 (type I diabetes); Arthritis Rheum. 50, 3250-59, 2004 (lupus erythymatosus); Clin. Exp. Immunol. 99, 294-302, 1995 (Grave's disease); J. Clin. Invest. 116, 905-15, 2006 54 (multiple sclerosis); Pharmacol Res. e-published Feb. 1, 2006 (ulcerative colitis); J. Pathol. e-published March 21, 2006 (Crohn's disease); J. Clin. Invest. 116, 961-973, 2006 (rheumatoid arthritis); Endocrinol. 147, 754-61, 2006 (asthma); Exp Mol PathoL. 77, 161-67, 2004 (Hashimoto's thyroiditis); J. Rheumatol. Supply. 11, 114-17, 1983 (Reiter's syndrome); Rheumatol. 32, 1071-75, 2005 (Sj6gren's syndrome); Brain PathoL. 12, 420-29,'2002 (Guillain-Barr6 syndrome); J. Clin. Invest. 110, 955-63, 2002 (vessel vasculitis); Vet. PathoL. 32, 337-45, 1995 (polyarteritis nodosa); Immunol. Invest. 3,47 61, 2006 (pemphigus vulgaris); Arch. Dermatol. Res. 297, 333-44, 2006 (scleroderma); J. Exp. Med. 191, 899-906, 2000 (Goodpasture's syndrome); J. Vet. Med. Sci. 68, 65-68, 200.6 (glomerulonephritis); Liver Int. 25, 595-603, 2005 (primary biliary cirrhosis); Clin. Exp. Immunol. 99, 294-302, 1995 (Grave's disease); J. Clin. Invest. 91, 1507-15, 1993 (membranous nephropathy); J. Immunol. 169, 4889-96, 2002 (autoimmune hepatitis); Isr. J. Med. Sci. 15, 348-55, 1979 (celiac sprue); Surgery 128, 999-1006, 2000 (Addison's disease); J. Neuroimmunol. 98, 130-35, 1999 (polymyositis); Am. J. Pathol. 120, 323-25, 1985 (dermatomyositis); Bone 20, 515-20, 1997 (monoclonal gammopathy); Haemophilia 11, 227-32, 2005 (Factor VIII deficiency); Proc. NatL. Acad. Sci. USA 94, 233-36, 1997 (cryoglobulinemia); Pain 110, 56-63, 2004 (peripheral neuropathy); Ann. Neurol. 49, 712-20, 2001 (IgM polyneuropathy); J. Neurosci. Res: 44, 58-65, 1996 (chronic neuropathy); Eur. J. Immunol. 32, 1147-56, 2002 (autoimmune hemolytic anemia); Haematologica 88, 679-57, 2003 (autoimmune thrombocytopenic purpura); Curr. Top. Microbiol. Immunol. 293, 153-77, 2005 (pernicious anemia); J. Immunol. 175, 2475-83, 2005 (ankylosing spondylitis); Inflamm. Res. 53, 72-77, 2004 (vasculitis); Vet. Pathol. 43, 2-14, 2006 (inflammatory bowel disease); and J. Biol. Chem. 276, -13821, 2001 (anti-phospholipid antibody syndrome). 11171 LDso (the dose lethal to 50% of the population) and the ED 5 o (the dose therapeutically effective in 50% of the population) can be determined by standard pharmaceutical procedures in cell cultures and/or experimental animals. Data obtained from cell culture assays or animal studies can be used to determine initial human doses. As is known in the art, the dosage may vary depending upon the dosage form and route of administration used. 55 11181 As is well known, the FDA guidance document "Guidance for Industry and Reviewers Estimating the Safe Starting Dose in Clinical Trials for Therapeutics in Adult Healthy Volunteers" (HFA-305) provides an equation for use in calculating a human equivalent dose (HED) based on in vivo animal studies. Based on the studies described in Example 16, below, the human equivalent dose ranges between 1.5 mg/kg and 8 mg/kg, with some' compounds showing considerable efficacy at lower or higher doses than those estimated by the HED. Thus, human dosages for systemic administration can range from, e.g., 1.5 mg/kg to 3 mg/kg; 2 mg/kg to 4 mg/kg; 5 mg/kg to 7 mg/kg; and 4 mg/kg to 8 mg/kg. The amount of composition administered will, of course, be dependent on the subject being treated, on the subject's weight, the severity of the disorder, the manner of administration and the judgment of the prescribing physician. [119] All patents, patent applications, and references cited in this disclosure are expressly incorporated herein by reference. The above disclosure generally describes the present invention. A more complete understanding can be obtained by reference to the following specific examples, which are provided for purposes of illustration only and are not intended to limit the scope of the invention. EXAMPLE 1 Preparation of 1-naphthalen-2-yl-prop-2-en-1-ol H OH [1201 Naphthaldehyde (5.0 g, 32.0 mmole) was dissolved in anhydrous tetrohydrofuran and stirred at -78 0C under N 2 (g) atmosphere. To the mixture was added vinyl magnesium bromide (50 ml, 1 M solution in THF) and the reaction was warmed to room temperature and stirred overnight. The reaction was quenched with water and partitioned between EtOAc and water. The organic layer was washed with brine, dried over sodium sulfate, 56 filtered, and concentrated under vacuum to give the desired product as yellow oil (5.0 g, 85%). ESI-MS m/z 185 (M+H)+ EXAMPLE 2 Preparation of 1-naphthalen-2-yl-propenone OH 0 [1211 To a solution of 1-naphthalen-2-yl-prop-2-en-l-ol (1.3 g, 7.0 mmole) in 30 ml of dichloromethane was added pyridinium chlorochromate (1.5 g, 7.0 mmole). The mixture was stirred at room temperature until oxidation was complete. The solution was filtered through celite and the solvent was concentrated under vacuum. The residue was re dissolved in EtOAc and washed with water and brine, dried over sodium sulfate, filtered, and concentrated under vacuum. The residue was purified by HPLC using a 0-100% EtOAc-Hx gradient to give the desired product as yellow oil (280 mg, 22%). ESI-MS m/z 183 (M+H)*. EXAMPLE 3 Preparation of 1-naphthalen-2-yl-3-piperidin-1-yl-propan-1 -one 0 0 N [122] 1-Naphthalen-2-yl-propenone (10 mg, 0.05 mmole) was dissolved in 100 P1 of DMSO in one well of a 96 well polypropylene plate. To the mixture was added piperidine (12 pl, 0.10 mmole) and diisopropylethyl amine (17 1, 0.1mmole). After completion, the 57 product was .purified using HPLC to give the desired product (50 mm x 10 mm Phenomenex GEMINITM column using a 30-100% acetonitrile-water gradient). ESI-MS m/z 268 (M+H)*. EXAMPLE 4 Preparation of JH-Pyrrolo[2,3-bipyridine 7-oxide N N N N H H 0 [123] 7-Azaindole (10g, 84.7 mmol) was dissolved in ether (300 mL) at room temperature. M CPBA (29.1 g, 1.5 eq.) was added in portions and stirred by manual agitation. After all oxidant was added, the mixture was stirred at room temperature for a further 3 hours. LC/MS showed complete conversion. The mixture was filtered, and the solid was washed with ether (40 mL X 3) and air-dried. NMR analysis of this solid in d6-DMSO obtained showed the product as mostly the meta-Chloro benzoic acid salt of 1H-Pyrrolo[2,3 b]pyridine 7-oxide (off white, 17.9 g); MS: m/z 135.3 [MH*]. EXAMPLE 5 Preparation of4-Chloro-JH-pyrrolo[2,3-bipyridine Cl N' N H H 0 58 [124] The m-CBA salt of 1H-Pyrrolo[2,3-b]pyridine 7-oxide (9 g) was taken into POCl 3 (46 mL, 7.5 eq.). The mixture was heated at 90 *C for 15 hours and to 106 "C for another 4 hours. The mixture was cooled to room temperature, and most of the POCl 3 was distilled off under high vacuum. The residue was dissolved in CH 3 CN (10 mL). Water (20 mL) was added slowly to quench the reaction. The resulted mixture was adjusted to pH ~ 9 using 10 N NaOH. The solid was filtered. The crude solid was redissolved in several ml of THF and combiflashed using 0-10% MeOH in DCM to give 4-Chloro-iH pyrrolo[2,3-b]pyridine as a slightly yellowish solid. (4 g). MS: mlz 154.9 [MH*]. EXAMPLE 6 Preparation of J-[4-(JH-Pyrrolo[2,3-b]pyridin-4-yl)-phenyl]-ethanone 0 0 C N N o o N N [1251 4-Chloro-1H-pyrrolo[2,3-b]pyridine (500 mg, 3.27 mmol) was dissolved in dioxane (11 mL). 4-Acetyl phenylboronic acid (802 mg, 4.9 mmol, 1.5 eq), dppfPdCl 2 (41 mg, 0.03mmol, 0.01 eq) and Na 2
CO
3 (2 N aq., 8.6 mL) were charged. The mixture was vacuumed and flushed with N 2 and microwaved at 160 "C for 15 minutes. Six batches of this same reaction were carried out. The crude mixture was pooled and partitioned between DCM (40 mL) and water (20 mL). Combi-flash of the residue using hexane/EtOAc (0% to 100%) gave the free base azaindole derivative as a slightly yellowish solid. The solid was redissolved in DCM (20 mL) and stirred in an ice bath. A 2M HC solution in ether (10 mL) was added dropwise. The precipitate was filtered and dried to give 1-[ 4 -(1H-Pyrrolo[2,3-b]pyridin-4-yl)-phenyl]-ethanone. (2.5g, 48%). MS: m/z 237.3 [MH*]. 59 EXAMPLE 7 Preparation of l-[3-(2-Chloro-pyridin-4-yl)-phenyl]-ethanone 0 HO, ,OH O B Kj+ ------ It N CI Br N CI [126] 2-Chloropyridine-4-boronic acid (11.0 g, 69.9 mmol), 3-Bromoacetophenone (11.2 mL, 83.9 mmol, 1.2 eq.), Na 2
CO
3 (35 mL,'244.65 mmol, 3.5 eq.) and dppfPdCl 2 (572 mg, 0.07 mmol, 0.01 eq.) were mixed in THF (200 mL). The mixture was heated to reflux and continued at this temperature for 6 hours. It was then cooled and concentrated in vacuo. The residue was partitioned between DCM and water (100 mL/40 mL). The layers were separated and the aqueous layer was washed further with DCM (2 x 40 mL). The combined organic layer was dried (Na 2
SO
4 ) and filtered. The filtrate was concentrated, and the residue was chromatographed using 1/1 hexane/EtOAc to give 1-[3-(2-Chloro pyridin-4-yl)-phenyl]-ethanone as a white solid (9.5 g, 58%). MS: m/z 232.1 [MHW]. EXAMPLE 8 Preparation of N-[4-(3-Acetyl-phenyl)-pyridin-2-yl]-benzamide 60 N NN O C1\ / [1271 A degassed mixture of 1-[3-(2-Chloro-pyridin-4-yl)-phenyl]-ethanone (500mg, 2.16 mmol), benzamide (523mg, 4.32 mmol, 2 eq.), Xantphos (120 mg, 0.21 mmol, 0.1 eq. ), Pd(OAc) 2 (24 mg, 0.10 mmol, 0.05 eq.), K 2 C0 3 (448 mg, 3.24 mmol, 1.5 eq.) in dioxane (12 mL) was irradiated with microwaves at 150 "C for 1 hour. LC/MS. control. Conversion was mostly 100% based on disappearance of starting material. Dimer (M+: 392 ) being the major by-product. If any starting material is unreacted at this point, another portion of Xantphos and Pd(OAc) 2 may be added and the mixture microwaved for another 30 minutes. The mixture was then partitioned between DCM and water (20 mL/10 mL). The layers were separated and the aqueous layer was washed further with DCM (2 x 20 mL). The combined organic layer was dried (Na 2
SO
4 ) and filtered. The filtrate was concentrated and the residue was chromatographed using 1/1 Hexane/EtOAc to give N-[4-(3-Acetyl-phenyl)-pyridin-2-yl]-benzamide as a white solid (375 mg, 55%). MS: m/z 317.1 [MH*]. EXAMPLE 9 61 Preparation ofN- 4
-[
3
-(
3 -Morpholin-4-yl-propionyl)-phenyl]-pyridin-2-yl}-benzamide 0 N 0 0 0 N N N N [1281 N-[4-(3-Acetyl-phenyl)-pyridin-2-yl]-benzamide (200mg, 0.632 mmol), morpholine HCI salt (78 mg, 0.632 mmol, I eq.) and paraformaldehyde (19 mg, 0.632 mmol, 1 eq.) were mixed with dioxane (2 mL) in a microwave tube. It was irradiated at 180 *C for 15 minutes. The mixture was partitioned between DCM/water (10 mL/5 mL). The aqueous layer was washed further with DCM (2 x 10 mL). The combined organic layer was dried (Na 2
SO
4 ) and filtered. The filtrate was concentrated and the residue was chromatographed using 20/1 DCM/MeOH to give N-{4-[3-(3-Morpholin-4-yl propionyl)-phenyl]-pyridin-2-yl}-benzamide as a slightly yellow solid (100 mg, 38%). MS: m/z 416.3 [MH*]. EXAMPLE 10 Preparation of J-[3-( 2 -Amino-pyridin-4-yl)-phenyl]-3-morpholin-4-yl-propan-1-one 62 0 0 N O N N N N N [1291 N-{4-[3-(3-Morpholin-4-yl-propionyl)-phenyl]-pyridin-2-y}-benzamide (100 mg, 0.32 mmol) was dissolved in HC (2 mL, 6 N). The mixture was irradiated with microwaves at 140 *C for 30 minutes. The mixture was diluted with DCM (20 mL) and neutralized with NaOH to pH - 9. The layers were separated and the aqueous layer was washed further with DCM (2 X 15 mL). The combined organic layer was dried (Na 2
SO
4 ) and filtered. The filtrate was concentrated and the residue was purified to give 1-[3-(2-Amino-pyridin 4-yl)-phenyl]-3-morpholin-4-yl-propan-l-one (TFA salt) as a white solid (84 mg, 78%). MS: m/z 312.3 [MH*] 63 EXAMPLE 11 Preparation ofN-[4-(3-Acetyl-phenyl)-pyridin-2-yl]-4-tert-butyl-benzamide 0 0 N C1 N N [130] According the same procedure for the preparation of N-[4-(3-Acetyl-phenyl)-pyridin-2 yl]-benzamide, N-[4-(3-Acetyl-phenyl)-pyridin-2-yl]-4-tert-butyl-benzamide (130 mg, 81%, slight impurity) was obtained from 1-[3-(2-Chloro-pyridin-4-yl)-phenyl]-ethanone (100 mg, 0.43 mmol) and 4-tert-butylbenzamide (153 mg, 0.86 mmol). MS: m/z 373.1 [MH*]. 64 EXAMPLE 12 Preparation of 4-tert-Butyl-N-{4-3-(3-morpholin-4-yl-propionyl)-phenyl]-pyridin-2-yl} benzamide 0 N N N N N [131] According to the same procedure for the preparation of 1-[3-(2-Amino-pyridin-4-yl) phenyl]-3-morpholin-4-yl-propan-1-one, 4-tert-Butyl-N- {4-3-(3-morpholin-4-yl propionyl)-phenyl]-pyridin-2-yl}-benzamide (12 mg, 30%) was obtained from N-[4-(3 Acetyl-phenyl)-pyridin-2-yl]-4-tert-butyl-benzamide (36 mg, 0.1 mmol). MS: m/z 472.3 [MH*]. 65 EXAMPLE 13 Preparation ofN-[4-(3-Acetyl-phenyl)-pyridin-2-yl]-acetamide o 0 N CI N [132] According the same procedure for the preparation of N-[4-(3-Acetyl-phenyl)-pyridin-2 yl]-benzamide, N-[ 4 -(3-Acetyl-phenyl)-pyridin-2-yl]-acetamide (50 mg, 50%, slight impurity) was obtained from 1-[3-(2-Chloro-pyridin-4-yl)-phenyl]-ethanone (100 mg, 0.43 mmol) and acetamide (26 mg, 0.86 mmol). MS: m/z 255.1 [MH]. EXAMPLE 14 Preparation ofN-{ 4
-[
3
-(
3 -Morpholin-4-yl-propionyl)-phenyl]-pyridin-2-yl}-acetamide 0 0 N O N N N N 66 [1331 According to the same procedure for the preparation of 1-[ 3 -(2-Amino-pyridin-4-yl) phenyl]-3-morpholin-4-yl-propan-1 -one, N-{4-[3-(3-Morpholin-4-yl-propionyl)-phenyl] pyridin-2-yl}-acetamide (10 mg, 20%) was obtained from N-[4-(3-Acetyl-phenyl) pyridin-2-yl]-acetamide (50 mg, 0.2 mmol). MS, m/z 354.3 [MH*]. EXAMPLE 15 In vitro assays Measurement of IL-2 Production [1341 Human T cell lines were plated in 96 well plates pre-coated with anti-CD3 monoclonal antibodies. Wells were either left untreated or treated with anti-CD28 for 2 days. The supernatant was collected and tested for IL-2 production in the presence or absence of a test compound using a human IL-2 ELISA assay. T Cell Proliferation Assay [135] Human T cell lines were plated in 96 well plates pre-coated with anti-CD3 monoclonal antibodies. Wells were either left untreated or treated with anti-CD28 for 2 days. Cell proliferation was measured in the presence or absence of a test compound using a commercially available CELLTITER-GLOTM assay (Promega). In vitro Kinase Assays [136] Compounds were screened using the HITHUNTERTM enzyme fragment complementation method (Discoverx). Briefly, a recombinantly produced, N-terminally His-tagged ITK kinase domain (amino acids 352-617) was incubated with various concentrations of individual compounds. ATP and substrate were added, and the kinase reaction was allowed to proceed for 2-16 hours. Commercially available detection reagents were added and allowed to react for 2-4 hours. The reaction was evaluated by luminescence. Initial results were confirmed using full-length recombinant ITK protein. 67 [137] Similarly, commercially available reagents such as HITHUNTERTM were used to evaluate the effect of compounds on the activity of additional kinases. The kinase domains of BTK, LCK and ERK were expressed as recombinant purified proteins were used for these studies. [1381 The compounds in Table 1 were tested-and shown to inhibit IL-2 production, to inhibit T cell proliferation, and to inhibit ITK with an IC 50 of less than 1 pM. Table 1 Compound
IC
50 (pM) 0.01807 0 0.00954 0 NF F 0.01355 0> N 0 0.02851 68 0 0.00533 NLN 0.00426 0 0.05043 0 ~N 0.01 14 0 *N. N I~hhJ 0.01327 69 o 0.00686 N 0.02855 1N~ .0.01825 0.00085 0.019 0.01964 00 70 [139] The compounds in Tables 2-5 were tested in in vitro kinase assays: Table 2 Compound IC 5 o ITK ICso BTK IC 5 o LCK . (pM) (I'M) (p'M) 0 0.005 0.42482 12.55299 0 0 0 0.040 0.27584 2.89341 0 0.04022 0.0369 8.03843 N O F 0.013 1.41274 27.7419 o 0.013 0.10223 -34.05941 CI 0 0.014 1.83528 23.89837 XY71 71 o 0 0.020 O11 N o 0.025 0.36501 NO IC50 Br o 0.029 0.64341 413.06105 NNC o 0.035 0.94241 16.4214 0 N o 0.036 0.039 19.969 o 0.043 0.6561 27.11277 F3C o 0.056 0.86517 NO IC50 F _ o 0.065 1.24489 18.43928 0 72 O 0.067 0.12463 28.09552 CN N 0.072 0.7368 NO IC50 0 0.065 0.39763 23.16665 N 0 0.091 0.09415 18.46087 0 0.077 0.77538 47.61179 Cf o 0.096 1.53948 20.8277 o 0.104 0.23242 NO IC50 * N Br O 0 0.148 0.77352 28.01341 73 o 0.180 1.52018 163.63704 0 0.186 3.67569 20.64831 N o 0.199 0.4735 NO IC50 FCo_ 0 0.207 0.09415 18.46087 0 0.208 2.89272 33.1157 F CI 0 0.207 0.08071 NO IC50 0 0.219 1.30729 NO IC50 F3CNI0j aC O 0.223 1.47599 21.15799 Do 0 0.241 0.81405 NO IC50 c74 74 C 0.290 0.68214 25.86619 0 0.305 0.74064 NO 1C50 ~Br __ 0 0.345 3.1355 21.10834
H
3 CO OCH 3 o 0.381 3.03351 32.57859 F o 0.385 1.47531 25.34326 O 0.385 3.92321 23.25 o 0.385 0.75252 23.94596 0 0 0 0.468 1.21899 NO IC50 F CF 3 ____ 75 o 0.560 3.06627 24.36134 N O 0.569 1.01979 NO IC50 CF3 _ _ _ _ o 0.611 2.31114 NO IC50 O 0.797 3.62429 NO IC50 o 0.935 0.99267 29.52378 F o 0.874 2.57662 NO IC50 F o 1.279 0.55617 704.77096 Z _ _ O 1.406 4.1378 27.08267 N 76 -4 04 0 C0 0 0 0 0 1 //g 00 r- O 0 00 6 6 0 0 0 0 Cf) 0I Z 0 a:Z 0 0 6 o0/ 0) 77 00 C14 I'D 00 LL 5b -K 0 00 0 6 607 0 00 ~0 00 0 0 00 JZ 0 0I) 0>~~~ 0 C 78 Table 4 Compound IC5 0 ITK (pM) IC 5 o BTK (pM) 0.05666 0
CF
3 0.03034 0 0.09281 0 N 0.02285 0 N 0.07489 79 c 0.020 0.039 0 0.048 5.001 o 1.684 no IC 50 o 0.189 0.100 N N C 0 0.179 0.174 N HN--N O 0 0.049 0.296 N 0 0.134 0.611 S O 80 0 0.075 2.038 oN o 0.060 0.334 SN 0 56.392 no IC 50 0 0.010 0.017 O 0.030 0.006 a HO"' 0 3.339 2.364 OC I OH 81 0 0.005 0.001
NH
2 0 0.042 0.002 HN O Table 5 Compound
IC
50 ITK (ptM) IC50 BTK (ptM) 0 0.0205 0.0395 NN 0 0.1369 8.21849 0 0.0080 0.06706 N.N No82 82 o 0.0169 N N 0.0602 0.12799 0.0148 cc N o 0.5375 0.8516 0 0.0309 N83 83 0 0.0212 0 0.1609 0 0.0242 NN O 0.007 0.11532 N N 0.3096 N 0.0069 0.0492 84 0 0.0187 0 0.0095 CO aN~ o 0.0162 ~N.C'. 0 0.0359 N
N
0 0.0147 85 0 0.0092 coK 0 0.0062 0.16054 O j 0.0163 N NH N 0.023 0.153
NH
2 86 0.056 0.452 N 0.060 0.242 (N) 0.066 0.089 0.064 0.360 cN r 87 0.054 0.018 N 0.087 0.051 0 0.031 0.071 o 0.066 0.117 0 0 0.049 0.123 OH 88 0 0.086 -0.084
NH
2 o 0.284 0.486 N N N 0 .0 N o0.217 0.266 HN 0 CF3 NH0.163 0.100 H "' NH 0.036 0.004 N H 8 N 89 0 0.737 0.373 N H N 0 N EXAMPLE 16 In vivo studies [140] Several representative compounds were evaluated for efficacy in mouse in vivo tumor models. NOD/SCID mice were implanted intraperitoneally with T cell leukemia/lymphoma cells. One group was treated with vehicle alone (mock treatment) while the other groups were treated with several small molecule inhibitors via intraperitoneal route. Tumor growth was evaluated by peritoneal savage and FACS analysis. Table 6 summarizes percent inhibition of tumor growth relative to a mock group treated with vehicle alone. Doses of compounds evaluated in this study were below the maximal tolerated- dose, and showed minimal toxicity. L141] The compounds in Table 6 were tested and inhibited tumor growth by at least 50% at the concentrations shown. 90 Table 6 Compound mg/kg % inhibition tumor growth 0 100 70-90 N~Q
/N
o 100 85-98 a 00 0 0 80 92-99 o 80 50-80 O F H 0 80 90-99 N N N H o 80 99. NN 0 80 67-81 N 91 0 80 92-99 N O N o 100 99 H2 0 80 81-95 o, N N:: 0 00 0 80 9-99 N N 00 0 20 99 01 0 80 81-95 N.- Na 050 H0 20 40 vehicle 0 92 EXAMPLE 17 Compound activity mechanism [142] The compound class interacts selectively with kinase domains of such kinase families as Tec and EGFR, as well as a few additional kinases. There is evidence indicating that this class of compounds reacts irreversibly at the ATP binding site of the kinase binding domain, through a mechanism that involves the exposure of a reactive aminoethyl C=Y warhead through the in situ elimination of a leaving group. The compounds contain an abstractable proton adjacent to the C=Y group, which upon exposure to an appropriate catalytic environment in the active site of a kinase of interest will promote elimination of the beta-amino functionality. This elimination thus generates a reactive electrophilic species (commonly termed a Michael acceptor moiety) which, due to the existence of a proximal cysteine residue in the kinase active site, rapidly forms a covalent adduct between this cysteine residue and the in situ generated electrophylic species. The combination of a kinase with the catalytic environment in close proximity to a nucleophilic cysteine, is a vital and unique requirement that describes this mechanism of action. The data below support that in situ elimination promotes the inhibitory activity of compounds in depicted in this invention. When a compound is modified in a manner that prevents elimination, the compound fails to. exhibit inhibitory activity. 93 Table 7 compound
IC
50 ITK O 0.0446 N Q OH no IC 50 N O no IC 5 0 EXAMPLE 18 Covalent binding to Select Kinases [143] As a result of elimination in proximity of a relevant cysteine, a covalent adduct is formed between the compound and the kinase domain. The irreversible binding that ensues can be demonstrated by several methods, including surface plasmon resonance (SPR) and co precipitation of the compound with the kinase. [144] BIACORE* is a SPR-based protein interaction approach, whereby the kinase is immobilized on the sensor chip, and. a small molecule solution allowed to interact with the kinase. Detection of small molecule/kinase interaction occurs in real time, and is detected as a difference in SPR response. Figure 1 shows a BIACOREeexperiment in which the ITK kinase domain was immobilized on a biosensor, and evaluated for its 94 ability to bind and dissociate form a small molecule. The data indicates that compounds depicted in this application bind to the ITK kinase domain irreversibly. [145] In the co-precipitation assay, 1-10 mM labeled compound is incubated with cell lysates from either kinase expressing or kinase lacking cells. The label is then used to precipitate the compound and any bound proteins. The mixture is separated by SDS-PAGE and proteins are identified by western blotting and/or Mass spectrophotometry. EXAMPLE 19 Contribution of Cysteine 442 to adduct formation [146] In order to confirm the mechanism by which compounds depicted herein interact with the. kinase domain of Tec and EGFR kinases, we created a point mutant of the ITK kinase domain, whereby the key amino acid, namely C442 was mutated to alanine. The protein was expressed in a commercial baculovirus expression system using the manufacturer's general protocol (Invitrogen, pBlueBac). Protein was expressed and purified using standard techniques. Both wild type (WT) ITK kinase domain and C442A kinase domain exhibited kinase activity. While the activity of WT-ITK was inhibited by compounds depicted in this application, the same compounds had no activity towards the C442A mutant kinase domain. 95 Table 8 compound IC 50 (;M) wild-type ITK C442A-ITK control (BMS-488516) 0.0392 0.0532 0 0.011 >10 1&01
N
0 0.0496 >10 Fj: 0.0111 >10
H
2 N 0 96

Claims (8)

1. A protein kinase inhibitor that binds to a DKC triad kinase active site, comprising: (a) a proton acceptor placed in a DKC triad kinase active site within hydrogen bonding distance of an amino group of a lysine of a catalytic dyad in the kinase active site; (b) an abstractable proton in hydrogen-bonding proximity of an aspartate of the catalytic dyad in the kinase active site, wherein removal of the abstractable proton creates a conjugated system capable of electronic rearrangement to an enol/enolate or thiol/thiolate or enamine; (c) a leaving group, wherein further electronic rearrangement leads to -elimination of the leaving group, whereby a Michael acceptor in the inhibitor is created, and wherein in at least one conformation of the inhibitor and the kinase is such that the Michael acceptor moiety is located within a distance of 3-10 A from a cysteinyl nucleophile, causing a reaction to form a Michael adduct with the enzyme; and (d) a kinase binding moiety with affinity for a portion of an ATP binding site selected from the group consisting of a hinge region of the kinase, hydrophobic amino acid residues, hydrophilic amino acid residues, and combinations thereof, with the proviso that the protein kinase inhibitor is not or V
2. The protein kinase inhibitor of claim 1, wherein the protein kinase inhibitor forms an irreversible adduct with the DKC triad kinase active site.
3. A composition comprising: a pharmaceutically acceptable carrier; and a protein kinase inhibitor that binds to an aspartate-lysine-cysteine (DKC) triad kinase active site, and that comprises: (a) a proton acceptor placed in a DKC triad kinase active site within hydrogen bonding distance of an amino group of a lysine of a catalytic dyad in the kinase active site; (b) an abstractable proton in hydrogen-bonding proximity of an aspartate of the catalytic dyad in the kinase active site, wherein removal of the abstractable proton creates a 98 conjugated system capable of electronic rearrangement to an enol/enolate or thiol/thiolate or enamine; (c) a leaving group, wherein further electronic rearrangement leads to j-elimination of the leaving group, whereby a Michael acceptor in the inhibitor is created, and wherein in at least one conformation of the inhibitor and the kinase is such that the Michael acceptor moiety is located within a distance of 3-10 A from a cysteinyl nucleophile, causing a reaction to form a Michael adduct with the enzyme; and (d) a kinase binding moiety with affinity for a portion of an ATP binding site selected from the group consisting of a hinge region of the kinase, hydrophobic amino acid residues, hydrophilic amino acid residues, and combinations thereof, with the proviso that the protein kinase inhibitor is not or
4. The composition of claim 3, wherein the protein kinase inhibitor forms an irreversible adduct with the DKC triad kinase active site.
5. An adduct comprising: an aspartate-lysine-cysteine (DKC) triad kinase; and a DKC triad kinase inhibitor that binds to a DKC triad kinase active site, and that comprises: (a) a proton acceptor placed in a DKC triad kinase active site within hydrogen bonding distance of an amino group of a lysine of a catalytic dyad in the kinase active site; (b) an abstractable proton in hydrogen-bonding proximity of an aspartate of the catalytic dyad in the kinase active site, wherein removal of the abstractable proton creates a conjugated system capable of electronic rearrangement to an enol/enolate or thiol/thiolate or enamine; (c) a leaving group, wherein further electronic rearrangement leads to j-elimination of the leaving group, whereby a Michael acceptor in the inhibitor is created, and wherein in at least one conformation of the inhibitor and the kinase is such that the Michael acceptor moiety is located within a distance of 3-10 A from a cysteinyl nucleophile, causing a reaction to form a Michael adduct with the enzyme; and 99 (d) a kinase binding moiety with affinity for a portion of an ATP binding site selected from the group consisting of a hinge region of the kinase, hydrophobic amino acid residues, hydrophilic amino acid residues, and combinations thereof, with the proviso that the protein kinase inhibitor is not or
6. The adduct of claim 5, wherein the adduct is an irreversible adduct.
7. A complex comprising a protein kinase inhibitor that is bound to an aspartate-lysine cysteine (DKC) triad kinase, wherein the protein kinase inhibitor comprises: (a) a proton acceptor placed in a DKC triad kinase active site within hydrogen bonding distance of an amino group of a lysine of a catalytic dyad in the kinase active site; (b) an abstractable proton in hydrogen-bonding proximity of an aspartate of the catalytic dyad in the kinase active site, wherein removal of the abstractable proton creates a conjugated system capable of electronic rearrangement to an enol/enolate or thiol/thiolate or enamine; (c) a leaving group, wherein further electronic rearrangement leads to -elimination of the leaving group, whereby a Michael acceptor in the inhibitor is created, and wherein in at least one conformation of the inhibitor and the kinase is such that the Michael acceptor moiety is located within a distance of 3-10 A from a cysteinyl nucleophile, causing a reaction to form a Michael adduct with the enzyme; and (d) a kinase binding moiety with affinity for a portion of an ATP binding site selected from the group consisting of a hinge region of the kinase, hydrophobic amino acid residues, hydrophilic amino acid residues, and combinations thereof, with the proviso that the protein kinase inhibitor is not or F 100
8. The complex of claim 7, wherein the protein kinase inhibitor is irreversibly bound to the DKC triad kinase active site. MannKind Corporation Patent Attorneys for the Applicant/Nominated Person SPRUSON & FERGUSON
AU2014250696A 2006-05-18 2014-10-17 Intracellular kinase inhibitors Abandoned AU2014250696A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2014250696A AU2014250696A1 (en) 2006-05-18 2014-10-17 Intracellular kinase inhibitors

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US60/801,074 2006-05-18
US60/869,664 2006-12-12
AU2013200229A AU2013200229B2 (en) 2006-05-18 2013-01-16 Intracellular kinase inhibitors
AU2014250696A AU2014250696A1 (en) 2006-05-18 2014-10-17 Intracellular kinase inhibitors

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2013200229A Division AU2013200229B2 (en) 2006-05-18 2013-01-16 Intracellular kinase inhibitors

Publications (1)

Publication Number Publication Date
AU2014250696A1 true AU2014250696A1 (en) 2014-11-06

Family

ID=51845226

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2014250696A Abandoned AU2014250696A1 (en) 2006-05-18 2014-10-17 Intracellular kinase inhibitors

Country Status (1)

Country Link
AU (1) AU2014250696A1 (en)

Similar Documents

Publication Publication Date Title
AU2007254179B2 (en) Intracellular kinase inhibitors
US11337956B2 (en) IRE-1α inhibitors
TWI671298B (en) Substituted polycyclic pyridone derivatives and prodrug thereof
JP5941173B2 (en) IRE-1α inhibitor
JP4838128B2 (en) Piperazine derivatives and their use as therapeutic agents
JP2013053161A (en) Heterocyclic derivative and use thereof as therapeutic agent
JP2005508867A (en) HIV protease inhibitors, compositions containing them, their pharmaceutical use and materials for their synthesis
KR20090114439A (en) Acylguanidine derivative
ES2782357T3 (en) IRE 1 alpha inhibitors
Wu et al. Synthesis of Pyrazine‐1, 3‐thiazine Hybrid Analogues as Antiviral Agent Against HIV‐1, Influenza A (H1N1), Enterovirus 71 (EV 71), and Coxsackievirus B3 (CVB 3)
AU2013200229B2 (en) Intracellular kinase inhibitors
AU2014250696A1 (en) Intracellular kinase inhibitors
CN112209917B (en) Application of heterocyclic substituted phenylpiperazine or phenylpiperidine derivatives in antidepressant drugs
RU2671179C2 (en) 1-(3-aminopropyl)-substituted cyclic amine compounds, preparation method therefor and pharmaceutical compositions and uses thereof
CA3153006A1 (en) Modulators of circadian rhythms and uses thereof
NZ791086A (en) Substituted polycyclic pyridone derivative and pharmaceutical composition containing prodrug thereof
Awasthi Dual Targeting Ligands Of Sigma Receptors And Dopamine Transporters As Potential Pharmacotherapy For Cocaine Abuse

Legal Events

Date Code Title Description
PC1 Assignment before grant (sect. 113)

Owner name: PHARMACYCLICS, INC.

Free format text: FORMER APPLICANT(S): MANNKIND CORPORATION

MK5 Application lapsed section 142(2)(e) - patent request and compl. specification not accepted