WO2007084391A2 - Thiazole compounds as protein kinase b ( pkb) inhibitors - Google Patents

Thiazole compounds as protein kinase b ( pkb) inhibitors Download PDF

Info

Publication number
WO2007084391A2
WO2007084391A2 PCT/US2007/000871 US2007000871W WO2007084391A2 WO 2007084391 A2 WO2007084391 A2 WO 2007084391A2 US 2007000871 W US2007000871 W US 2007000871W WO 2007084391 A2 WO2007084391 A2 WO 2007084391A2
Authority
WO
WIPO (PCT)
Prior art keywords
amino
alkyl
thiazol
compound
phenyl
Prior art date
Application number
PCT/US2007/000871
Other languages
English (en)
French (fr)
Other versions
WO2007084391A3 (en
Inventor
Qingping Zeng
John G. Allen
Matthew P. Bourbeau
Celia Dominguez
Christopher H. Fotsch
Nianhe Han
Fang-Tsao Hong
Xin Huang
Matthew R. Lee
Aiwen Li
Qingyian Liu
James T. Rider
Seifu Tadesse
Andrew S. Tasker
Vellarkad N. Viswanadhan
Xianghong Wang
Kurt E. Weiler
George E. Wohlhieter
Guomin Yao
Chester Chenguang Yuan
Original Assignee
Amgen Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Amgen Inc. filed Critical Amgen Inc.
Priority to CA2636077A priority Critical patent/CA2636077C/en
Priority to AU2007207743A priority patent/AU2007207743B2/en
Priority to EA200801716A priority patent/EA200801716A1/ru
Priority to ES07718125T priority patent/ES2389062T3/es
Priority to JP2008551297A priority patent/JP5199885B2/ja
Priority to BRPI0706621-0A priority patent/BRPI0706621A2/pt
Priority to EP07718125A priority patent/EP1981884B1/en
Publication of WO2007084391A2 publication Critical patent/WO2007084391A2/en
Publication of WO2007084391A3 publication Critical patent/WO2007084391A3/en
Priority to IL192751A priority patent/IL192751A0/en
Priority to NO20083572A priority patent/NO20083572L/no

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/04Ortho-condensed systems

Definitions

  • the invention relates to thiazole compounds useful for treating diseases mediated by protein kinase B (PKB).
  • PPKB protein kinase B
  • the invention also relates to the therapeutic use of such thiazole compounds and compositions thereof in treating disease states associated with abnormal cell growth, cancer, inflammation, and metabolic disorders.
  • Protein kinases represent a large family of proteins which play a central role in the regulation of a wide variety of cellular processes, maintaining control over cellular function.
  • a partial list of such kinases includes abl, bcr-abl, BIk, Brk, Btk, c-kit, c-met, c-src, c-fms, CDKl, CDK2, CDK3, CDK4, CDK5, CDK6, CDK7, CDK8, CDK.9, CDKlO, cRafl, CSFlR, CSK, EGFR, ErbB2, ErbB3, ErbB4, Erk, Fak, fes, FGFRl, FGFR2, FGFR3, FGFR4, FGFR5, Fgr, flt-1, Fps, Frk, Fyn, GSK3 ⁇ , GSK3 ⁇ , Hck, IGF- IR, INS-R, Jak, KDR, Lck, Lyn, MEK
  • AKT also known as protein kinase B (PKB) or RAC-PK
  • PKT protein kinase B
  • RAC-PK protein kinase B
  • AKT2/PKB ⁇ /RAC-PK ⁇ J AKT3/PKB ⁇ /RAC- PK ⁇ has been identified as a serine/threonine protein kinase.
  • PKB mediates many effects of IG F-I and other growth factors on tumor growth and inhibition of apoptosis.
  • PKB plays an important role in cell proliferation, apoptosis and response to insulin. For these reasons, modulation
  • PKBs are of interest in the treatment of tumorigenesis, abnormal cell proliferation, and diabetes.
  • the molecular structure of the PKBs comprises a regulatory site near the carboxy terminus of the polypeptide, a catalytic domain with an activation loop having a threonine, and an amino-terminal pleckstrin homology domain.
  • the pleckstrin homology domain permits anchorage of the enzyme to the cell membrane through interaction with phospholipids, which triggers the activation of the PKBs.
  • the role of the pleckstrin homology domain requires phosphorylation of phosphatidylinositol at the D-3 position via phosphatidylinositol 3-kinase PI3K, an SH2 domain protein that associates with activated receptor tyrosine kinases, particularly IGF-I R.
  • phosphoinositol-3- kinase when activated by receptor tyrosine kinase, catalyzes the synthesis of phosphoinositol-3,4-diphosphate and phosphatidylinositol 3,4,5-triphosphate.
  • the pleckstrin homology domain binds 3-phosphoinositides, which are synthesized by PI3K upon stimulation by growth factors such as platelet derived growth factor (PDGF), nerve growth factor (NGF) and insulin-like growth factor (IGF-I ).
  • PDGF platelet derived growth factor
  • NTF nerve growth factor
  • IGF-I insulin-like growth factor
  • Activation of PKB can also occur by inhibiting the D-3 phosphoinositide specific phosphatase, PTEN, which is a membrane-associated FYVE finger phosphatase commonly inactivated in many cancers due to genetic alteration, including prostate cancer.
  • PTEN D-3 phosphoinositide specific phosphatase
  • the catalytic domain of PKB is responsible for the phosphorylation of serine or threonine in the target protein.
  • PKB mediates several cellular functions including proliferation, cell growth, and promotion of survival. Intracoronary, adenovirus-mediated akt gene transfer in heart limits infarct size following ischemia-reperfusion injury in vivo. Miao et al, J. MoI. Cell. Cardiol., 2000, 32, 2397-2402.
  • the antiapoptotic function of PKB is reported to be mediated by its ability to phosphorylate apoptosis regulatory molecules including BAD, caspase 9, IKK-, and the forkhead transcriptional factor FKHRLl.
  • PKB kinase activity is constitutively activated in tumors with PTEN mutation, PI 3-kinase mutation and overexpression, and receptor tyrosine kinase overexpression.
  • PKB is also a mediator of normal cell functions in response to growth factor signaling. Expression of the PKB gene was found to be amplified in 15% of human ovarian carcinoma cases. Cheng, et ah, Proc. Natl. Acad. Sci. U.S.A., 1992, 89, 9267-9271. PKB has also been found to be over expressed in 12% of pancreatic cancers. Cheng, et al., Proc.
  • PKB ⁇ is especially abundant in highly insulin-responsive tissues, including brown fat; PKB ⁇ is widely expressed in most of the tissues; and PKB ⁇ is more abundant in brain and testes.
  • Modulation of PKB by small molecules can be achieved by identifying compounds that bind to and activate or inhibit one or more PKBs.
  • ROCK Rho-associated coiled-coil forming protein serine/threonine kinase
  • the AGC sub-family of kinases includes protein kinase A (PKA),
  • Triciribine has been reported to inhibit cell growth in PKB ⁇ overexpressing cells, transformed cells, and was effective at a concentration of 50 nM.
  • thiazole derivatives discloses certain thiazole derivatives, a method of obtaining them, and pharmaceutical compositions containing them.
  • the derivatives are described as adenosine antagonists useful in the prevention and/or treatment of cardiac and circulatory disorders, degenerative disorders of the central nervous system, respiratory disorders, and many diseases for which diuretic treatment is suitable.
  • This invention encompasses novel compounds useful for treating diseases or conditions mediated by PKB.
  • the invention also encompasses the therapeutic use of such compounds and compositions thereof in the treatment of disease states associated with abnormal cell growth, such as cancer, or metabolic disease states, such as diabetes, or inflammation.
  • the invention further provides pharmaceutical compositions that include the compounds of the invention and the use of the compounds in the preparation of medicaments for treating various conditions and disease states.
  • the invention comprises a compound of Formula I
  • Y is -N(R 5 )R 6 or -OR 6 ;
  • X is O, S, or -N(R 7 );
  • R 1 is R 8 , -CHR 1 '-N(H)-R 8 , -CHR 1 '-O-R 8 , C 2 -C 6 alkynyl, C 2 -C 6 hydroxyalkynyl, or
  • R 2 is aryl or heteroaryl
  • R 3 is — H, C)-C 6 alkyl which may be interrupted by one or more hetero atoms,
  • R 4 is C 1 -C 6 alkyl which may be interrupted by one or more hetero atoms, -(CR 9 R °) t (aryl), -(CR 9 R 1 °),(heteroaryl), -(CR 9 R 10 ) t (cycloalkyl), or -(CR 9 R I0 ),(heterocyclyl), or R 3 and R 4 , together with the carbon atom to which they are both attached, join to form a C 3 -Ci 0 heterocyclic or carbocyclic ring system, or R 4 and R 7 join to form a C 3 -C 10 heterocyclic ring;
  • R 5 is -H, C 1 -C 8 alkyl, -C(O)(CR 9 R I0 ),)N(R 7 ) 2 , -C(O)(CR 9 R 10 ),, -C(O) 2 (CR 9 R 10 ),, -(CR 9 R 10 ),(aryl), -(CR 9 R 10 ) t (heteroaryl), -(CR 9 R 10 ),(cycloalkyl), or -(CR 9 R 1 °),(heterocycly I), or R 4 and R 5 join to form a C 3 -C 10 heterocyclic ring;
  • R 6 and R 7 are independently selected from — H, Ci-C 8 alkyl, -(C]-C 6 alkyl)aryl, or -C(O)(Ci-C 6 alkyl), or R 6 and R 7 , together with the atoms to which they are linked, join to form a 5 to 6-membered heterocyclic ring, or
  • R s and R 6 together with the nitrogen atom to which they are linked, join to form a 5 to 6- membered heterocyclic or heteroaryl ring;
  • R 8 is -H, Ci-C 6 alkyl, -(C, -C 6 alkyl)aryl, aryl, or heteroaryl;
  • R 9 , R 10 , and R 1 ' are independently selected from -H, C 1 -C 6 alkyl, or aryl; wherein n is an integer from 1 to 6; m is an integer from 0 to 2; and each t is independently an integer from 0 to 3; wherein each of the above alkyl, aryl, heteroaryl, cycloalkyl, and heterocyclyl moieties and heterocyclic and carbocyclic rings are optionally and independently substituted by 1- 3 substituents selected from amino, aryl, heteroaryl, cycloalkyl, or heterocyclyl optionally substituted by 1-5 substituents selected from
  • Ci-C 6 alkyl optionally substituted by halo, aryl, halo, hydroxy!, heteroaryl,
  • Ci-C 6 hydroxyalkyl or
  • the invention comprises a compound of Formula I, wherein A is
  • the invention comprises a compound of Formula
  • the invention comprises a compound of Formula
  • X is -N(R 7 )
  • Y is -N(R 5 )(R 6 )
  • m, n, and t are 1.
  • the invention comprises a compound of Formula
  • A is and X is -N(R 7 ), Y is -N(R 5 )(R 5 ), R 2 is heteroaryl, R 3 is -H, R 4 is -(CR 9 R lo )t(aryl) or -(CR 9 R 1 °) t (heteroaryl), m, n, and t are 1, R 5 , R 6 , and R 7 are -H, and R 9 and R 10 are independently selected from H or Cj-C 3 alkyl.
  • the invention comprises a compound of Formula
  • X is -N(R 7 )
  • Y is -N(R S )(R 6 )
  • R 2 is bicyclic heteroaryl
  • R 3 is -H
  • R 4 is -(CR 9 R l0 ),(monocycIic aryl) or -(CR 9 R I0 ),(bicyclic heteroaryl)
  • m, n, and t are 1, and R 5 , R 6 , R 7 , R 9 and R 10 are -H.
  • the invention comprises a compound of Formula
  • R 2 is bicyclic heteroaryl
  • R 3 is -H
  • R 4 is -(CR 9 R 10 ),(monocyclic aryl) or -(CR 9 R l0 ),(bicyclic heteroaryl)
  • m, n, and t are 1, and R 5 , R 6 , R 7 , R 9 and R 10 are -H
  • the bicyclic heteroaryl group of R 2 is isoquinolinyl, lH-indazolyl, thiazolo[5,4-c]pyridinyl, benzo[d]thiazole-2(3H)-onyl, phthalazinyl, indolin-2-onyl, 3,4-dihydroquinolin-2(lH)-onyl, benzo[d ⁇ isoxazolyl, benzo[d]oxazol- 2(3H)-onyl, benzo[d ⁇ isoxazolyl, benzo[d]oxazol- 2
  • the bicyclic heteroaryl group of R 2 is isoquinolin-6-yl, 3-aminoisoquinolin-6-yI, lH-indazol-5-yl, 1 H- indazol-6-yl, 3-amino-l H-indazol-5-yl, 3-amino-lH-indazol-6-yl, 3 -amino- 1 -methyl- 1H- indazol-6-yl, 3-methylamino- l H-indazol-5-yl, 3-methyl-lH-indazol-5-yl, thiazolo[5,4- c]pyridin-2-yl, benzo[d]thiazole-2(3H)-on-6-yl, l-hydroxyphthalazin-6-yl, phthalazin-6- yl, indolin-2-on-5-yl, 3-methylindolin-2-on-5-yl, 3-(furan-2-ylmethylene)in
  • the monocyclic aryl group of R 4 is phenyl, 3-chlorophenyl, 4-chlorophenyI, 4-methoxyphenyI, 3-(trifluoromethyl)phenyl, or 4-(trifluoromethyl)phenyl, or the bicycHc heteroaryl group of R 4 is lH-indol-3-yl.
  • the invention comprises a compound of Formula
  • R 2 is bi cyclic heteroaryl
  • R 3 is -H
  • R 4 is -(CR 9 R 10 ),(monocyclic aryl), m, n, and t are 1
  • R 5 , R 6 , R 7 , R 9 and R 10 are -H
  • the bicyclic heteroaryl group of R 2 is isoquinolin-6-yl, 3-aminoisoquinolin-6-yl, 1H- indazol-5-yl, 3-methyl-lH-indazol-5-yl, thiazolo[5 s 4-c]pyridin-2-yl, benzo[d]oxazol- 2(3H)-on-6-yl, or l,6-naphthyridin-2-yl
  • the monocyclic aryl group of R 4 is 4- chlorophenyl, 3-(trifluoromethyl
  • the invention comprises a compound of Formula I having any of the features of any of the embodiments described above in which R 1 is — H, -CH 3 , -CH 2 CH 3 , -CH 2 OCH 3 , -CH 2 OCH 2 CH 3 , -CH 2 OH, -CH 2 OCH 2 CF 3 , -CH 2 N(H)CH 3 , -CH(CH 3 )OCH 3 , furanyl, phenyl, pyridyl, or -CsN.
  • R 1 is — H, -CH 3 , -CH 2 OCH 3 , -CH 2 OCH 2 CH 3 , -CH 2 OH, or furan-2-yl.
  • R 1 is -CH 3 , -CH 2 CH 3 , -CH 2 OCH 3 , -CH 2 OCH 2 CH 3 , -CH 2 OH, -CH 2 OCH 2 CF 3 , -CH 2 N(H)CH 3 , -CH(CH 3 )OCH 3 , furanyl, phenyl, pyridyl, or -C ⁇ N.
  • R 1 is -CH 3 , -CH 2 OCH 3 , -CH 2 OCH 2 CH 3 , -CH 2 OH, or furan-2-yl.
  • R 1 is -(CpC 6 alkyl), aryl, heteroaryl, -CHR 1 '-N(H)- R 8 , -CHR n -O-R 8 , or -CsN. In still other such embodiments, R 1 is -(C 1 -C 6 alkyl), heteroaryl, or -CHR 1 '-O-R 8 . . [0029] In another embodiment, the invention comprises a compound of Formula
  • A is aryl
  • X is -N(R 7 )
  • R 2 is heteroaryl
  • m is 1 .
  • the invention comprises a compound of Formula
  • the invention comprises a compound of Formula I, wherein A is aryl, X is -N(R 7 ), R 2 is a bicyclic heteroaryl, m is 1 , and R 7 is -H.
  • the invention comprises a compound of Formula
  • A is a monocyclic aryl
  • X is -N(R 7 )
  • R 1 is -H
  • R 2 is thiazolo[5,4-c]pyridin-2-yl
  • m is 1
  • R 7 is -H.
  • the invention provides a compound of Formula II
  • R 1 is -H, halo, -OR 8 , C 1 -C 6 alky], -(C 1 -C 6 alkyl)-O-R s , -(C 1 -C 6 haloalkyl)-O-R 8 , -(C 2 -C 6 alkenyl)-O-R 8 , -(C 1 -C 6 alkyl)N(R 7 ) 2 , -(C 1 -C 6 alkyl)aryl, -C(O)R 8 , -C(O)O-R 8 , -C(O)O-R 8 ,
  • R 2 is a carbocyclic ring system or is a heterocyclic ring system
  • R 5 is -H, C 1 -C 8 alkyl, -C(O)(CR 9 R 10 ),)N(R 7 ) 2i -C(O)(CR 9 R 10 ),, -C(O) 2 (CR 9 R 10 ),,
  • R 6 and R 7 are independently selected from — H, C 1 -C 8 alkyl, -(C ⁇ -C & alkyl)aryl, or -C(O)(C 1 -C 6 alkyl);
  • R 8 is selected from -H, C r C 6 alkyl, C 1 -C 6 haloalkyl, -(C r C 6 alkyl)aryl, aryl, heteroaryl,
  • R 9 and R 10 in each instance, and R 1 1 are independently selected from -H, Ci-C 6 alkyl, or aryl;
  • R 12 is -H, -OR 8 , -0-(Ci-C 6 alkyl)-O-R 8 , Ci-C 6 alkyl, C 1 -C 6 alkenyl, -(C 1 -C 6 alkyl)-O-R 8 , or -(Ci-C 6 alkyl)-O-C(O)-R 8 ;
  • R 13 is -H, or C,-C6 alkyl;
  • R 14 is -H, -OR 8 , -O-(C r C 6 alkyl)-O-R 8 , C 1 -C 6 alkyl, C 1 -C 6 alkenyl, -(C,-C 6 alkyl)-O-R 8 , or -(Ci-C 6 alkyl)-O-C(O)-R 8 ; each t is independently selected from 0, 1, 2, or 3; and Z is aryl or heteroaryl; wherein each of the above alkyl, aryl, heteroaryl, cycloalkyl, and heterocyclyl moieties and heterocyclic and carbocyclic rings are optionally and independently substituted by 1 - 3 substituents selected from amino, aryl, heteroaryl, cycloalkyl, or heterocyclyl optionally substituted by 1-5 substituents selected from Ci-C 6 alkoxy,
  • C]-C 6 alkyl optionally substituted by halo, aryl, halo, hydroxyl, heteroaryl,
  • the compound of Formula II has the Formula HB
  • the compound of Formula II has the Formula HC
  • the compound of Formula II has the Formula IID
  • the compound of Formula II has the Formula HE
  • R 1 is -H.
  • R 12 is — H or Q-C O alkyl. In some such embodiments, R 12 is — H or methyl.
  • R 13 is -H.
  • R 14 is — H.
  • R 14 is -OR 8 .
  • R 14 is selected from — H, methyl, ethyl, propyl, ethenyl, propenyl, hydroxymethyl, methoxymethyl,
  • Z is selected from optionally substituted phenyl, optionally substituted indolyl, optionally substituted naphthyl, optionally substituted pyridyl, or optionally substituted thiophenyl.
  • Z is selected from phenyl, indolyl, naphthyl, pyridyl, or thiophenyl, each of which is optionally substituted with 1 -3 substituents selected from -CI, -F, -CF 3 ,
  • Z is selected from phenyl, indolyl, naphthyl, pyridyl, thiophenyl, 4-chlorophenyl, 4-trifluormethylphenyI, 3- chlorophenyl, 3-trifluoromethylphenyl, 4-methoxyphenyl, 3-fluoro-4- trifluoromethylphenyl, 4-chloro-3 -fluorophenyl, 4-(3-chloropropoxy)phenyl > 4-(3- hydroxypropoxy)phenyl, 3,4-dichlorophenyl, 4-fluorophenyl, 2,4-dichlorophenyl, 4- methylphenyl, 3,4-difluorophenyl, 3-fiuoro-4-methoxyphenyl, 3,5-difluorophenyl, 6- trifluoromethylpyridin-3-yl, 5-methoxy-6-trifluoromethylpyridin-3-yl, 5-methoxy-6-trifluoromethylpyri
  • R 7 is H. In some such embodiments, R 5 and K 6 are both H.
  • R 5 and R 6 are both
  • R 12 is — H or Ci-C 6 alkyl
  • R 13 is -H
  • R 14 is -H
  • -OR 8 -0-(Ci-C 6 alkyl)-O-R 8 , C 1 -C 6 alkyl, C x -C 6 alkenyl, -(Ci-C 6 alkyl)-O-R 8 , or -(C 1 -C 6 alky I)-O-C(O)-R 8 .
  • R 14 is -OR 8 , -0-(Ci-C 6 alky])-O-R 8 , C 1 -C 6 alkyl, C r C 6 alkenyl, -(Ci-C 6 alkyl)-O-R 8 , or -(Ci-C 6 alky I)-O-C(O)-R 8 .
  • R 5 , R 6 , and R 7 are all H.
  • the carbocyclic ring system or the heterocyclic ring system of R 2 includes at least one aromatic ring.
  • R 2 is selected from optionally substituted phenyl, pyridyl, indazolyl, isoquinolinyl, thiazolopyridinyl, benzothiazolonyl, dihydroquinoli ⁇ onyl, benzoisoxazolyl, benzooxazolonyl, indolinonyl, benzoimidazolonyl, phthalazinyl, naphthyridinyl, thienopyridinyl, benzodioxolyl, isoindolinonyt, quinazolinyl, or cinnolinyl.
  • R 2 is selected from isoquinolinyl, 1 H-indazolyl, thiazolo[5,4-c]pyridinyl, benzo[d]thiazole-2(3H)-onyl, phthalazinyl, indolin-2-onyl, 3,4-dihydroquinolin-2(l H)-onyl, benzo[d]isoxazolyl, benzo[d]oxazol-2(3H)-onyl, benzo[d]imidazol-2(3 H)-ony 1, 1 ,6-naphthyridinyI, quinazolin-7-yl, or cinnolin-6-yl.
  • R 2 is isoquinolin-6-yl, 3- aminoisoquinolin-6-yl, lH-indazol-5-yl, lH-indazol-6-yl, 3-amino-l H-indazol-5-yl, 3- amino-l H-indazol-6-yl, 3-amino-l-methyl-l H-indazol-6-yl, 3-methylamino-l H-indazol- 5-yl, 3-methyl- lH-indazol-5-yl, thiazolo[5,4-c]pyridin-2-yl, benzo[d]thiazole-2(3H)-on- 6-yl, l -hydroxyphthalazin-6-yl, phthalazin-6-yl, indolin-2-on-5-yl, 3-methylindoIin-2-on- 5-yl, 3-(furan-2-ylmethylene)indolin-2-on-5-yl, 3-(l
  • R 2 is selected from one of the following groups, where the wavy line indicates the point of attachment to the thiazole:
  • R 1 is selected from
  • the invention comprises a pharmaceutically acceptable salt, hydrate, or solvate of a compound of Formula I or Formula II or any of the compounds listed above.
  • the pharmaceutically acceptable salts of Formula I compounds or Formula II compounds are selected from ammonium trifluoroacetate and ammonium chloride.
  • the invention comprises a pharmaceutical composition comprising a pharmaceutically-acceptable carrier and a compound of Formula I or Formula II, a compound of any of the embodiments described herein, and/or a salt of any of the compounds of any of the embodiments.
  • the invention also provides the use of a compound of any of the embodiments in the manufacture of a medicament for carrying out any of the methods of any of the embodiments of the invention.
  • Such compositions and medicaments may further include one or more additional therapeutic agent. Therefore, in some embodiments, the composition or medicament includes at least one additional therapeutic agent.
  • the invention comprises a method for treating a kinase- mediated disorder in a mammal comprising administering to the mammal a therapeutically effective amount of a compound of Formula I or Formula II or a pharmaceutical composition of the invention.
  • the invention provides the use of a compound of Formula I or Formula II or a pharmaceutical composition of the invention for treating a kinase-mediated disorder in a mammal.
  • the disorder can be one that is mediated by kinases including IGF-I R, Insulin Receptor, KDR, Tie2, EGFR, PKA, PKB, PKC, FKHR, TSC 1/2, SGK, LCK, BTK, Erk, MSK, MK2, MSK, p38, P7OS6K, PlM l, PIM2, ROCK2, GSK3, or a CDK complex.
  • the disorder is mediated by PKB, and in some embodiments is mediated by PKB ⁇ .
  • the method comprises selective inhibition of PKB. In some such embodiments, the method comprises selective inhibition of PKB ⁇ .
  • the invention encompasses Formula I compounds or Formula II compounds that have selective kinase activity ⁇ i.e., they possess significant activity against one specific kinase while possessing less or minimal activity against a different kinase.
  • the compounds have selective PKB inhibition activity.
  • the compounds have selective PKB ⁇ inhibition activity.
  • the invention provides the use of a compound of Formula I or Formula II or a pharmaceutical composition of the invention for selectively inhibiting a kinase activity.
  • PKB is selectively inhibited.
  • PKB ⁇ is selectively inhibited.
  • the invention provides a method of treating a proliferation-related disorder in a mammal in need thereof. Such methods include administering to the mammal a therapeutically effective amount of a compound of any of the embodiments described herein or a pharmaceutical composition comprising the compound. Another embodiment of the invention comprises treating abnormal cell growth by administering a therapeutically effective amount of a compound of the invention or a pharmaceutical composition of the invention to a subject in need thereof. In some embodiments, the invention provides the use of a compound of Formula I or Formula II or a pharmaceutical composition of the invention for treating abnormal cell growth. The abnormal cell growth can be a benign growth or a malignant growth.
  • the abnormal cell growth can be a carcinoma, sarcoma, lymphoma, or leukemia.
  • the abnormal cell growth is a cancer, including, but not limited to, lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, colon cancer, breast cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, chronic or acute leukemia, lymphocytic lymphomas,
  • the method of the invention also comprises treating a patient having cancer wherein the cancer is selected from the group consisting of small cell lung carcinoma, non-small cell lung carcinoma, esophageal cancer, kidney cancer, pancreatic cancer, melanoma, bladder cancer, breast cancer, colon cancer, liver cancer, lung cancer, sarcoma, stomach cancer, cholangiocarcinoma, mesothelioma, or prostate cancer.
  • said abnormal cell growth is a benign proliferative disease, including, but not limited to, psoriasis, benign prostatic hypertrophy or restenosis.
  • the invention comprises a method of administering a therapeutically effective amount of a Formula I or Formula II compound to a mammal for treating disease states or conditions selected from diabetes, inflammation, and metabolic disorders.
  • the invention provides the use of a compound of Formula I or Formula II or a pharmaceutical composition of the invention for treating a disease state or a condition selected from diabetes, inflammation, and metabolic disorders.
  • the invention encompasses a method for treating or preventing cancer in a patient in need thereof, comprising administering to the patient a therapeutically or prophylactically effective amount of a compound according to Formula I or Formula II and a pharmaceutically acceptable excipient, carrier, or vehicle.
  • the invention provides the use of a compound of Formula I or Formula II or a pharmaceutical composition of the invention for treating or preventing cancer in a patient such as in a human cancer patient.
  • the cancer is a tumor.
  • the invention encompasses a method for treating or preventing cancer in a patient in need thereof, comprising administering to the patient a therapeutically or prophylactically effective amount of a Formula I or Formula Il compound and at least one additional therapeutic agent.
  • alkyl means a saturated straight chain or branched non-cyclic hydrocarbon having from 1 to 20 carbon atoms, preferably 1-10 carbon atoms and most preferably 1-4 carbon atoms.
  • saturated straight chain alkyls include, but are not limited to, -methyl, - ethyl, -n-propyl, -n-butyl, -n-pentyl, -n-hexyl, -n-heptyl, -n-octyl, -n-nonyl and -n-decyl; while saturated branched alkyls include, but are not limited to, -isopropyl, -sec-butyl, - isobutyl, -tert-butyl, -isopentyl, 2-methylbutyl, 3-methylbutyl, 2-methylpentyl, 3- methylpentyl, 4-methylpentyl, 2-methyIhexyI, 3-methylhexyl, 4-methylhexyl, 5- methylhexyl, 2,3-dimethylbutyl, 2,3-dimethylpentyl, 2,4-dimethylpentyl,
  • An alkyl group can be unsubstituted or substituted.
  • An alkyl group may be designated as having a certain number of carbon atoms.
  • an alkyl group having from 1 to 8 carbon atoms may be designated as a Cj-Cg alkyl group whereas an alkyl group having from 1 to 6 carbon atoms may be designated as a C 1 -C 6 alkyl group.
  • the "-" symbol indicates the point of attachment to the rest of the molecule, and the term indicates that one of the hydrogens of the alkyl group is replaced by a bond to an aryl group.
  • -C 2 alkyl)aryl includes such groups as — CH 2 Ph, -CH 2 CH 2 Ph, and -CH(Ph)CH 3 .
  • an alkyl group can be interrupted by one or more heteroatoms such as N, O, S, or Si atoms. Insertion of a heteroatom in the alkyl group forms a heteroalkyl group.
  • the heteroatom is a N, O, or S atom.
  • heteroalkyl by itself or in combination with another term, means, unless otherwise stated, a stable straight or branched chain radical, or combination thereof, that includes carbon atoms and from one to three heteroatoms selected from the group consisting of O, N, and S.
  • the nitrogen and sulfur atoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized.
  • the heteroatom(s) O, N, and S may be placed at any position in the heteroalkyl group. Examples include -CH 2 -CH 2 -O- CH 3 , -CH 2 -CH 2 -NH-CH 3 , -CH 2 -CH 2 -N(CHs)-CH 3 , -CH 2 -S-CH 2 -CH 3 , -CH 2 -CH 2 -S(O)- CH 3 , and -CH 2 -CH 2 -S(O) 2 -CH 3 . Up to two heteroatoms may be consecutive or adjacent to one another, such as, for example, in -CH 2 -NH-OCH 3 .
  • a prefix such as (C 2 -C 8 ) is used to refer to a heteroalkyl group
  • the number of carbons (2 to 8, in this example) is meant to include the heteroatoms as well.
  • a C 2 -heteroalkyl group is meant to include, for example, -CH 2 OH (one carbon atom and one heteroatom replacing a carbon atom) and -CH 2 SH.
  • a heteroalkyl group is an oxyalkyl group.
  • (C 2- C 5 )oxyalkyl is meant to include, for example -CH 2 -O-CH 3 (a C 3 -oxyalkyl group with two carbon atoms and one oxygen replacing a carbon atom), -CH 2 CH 2 CH 2 CH 2 OH, and the like.
  • alkenyl means an unsaturated straight chain or branched non-cyclic hydrocarbon having from 2 to 20 carbon atoms and at least one carbon-carbon double bond.
  • an alkenyl has 2 to 10 carbon atoms and most preferably has 2 to 4 carbon atoms.
  • Exemplary straight chain alkenyls include, but are not limited to, -but-3-ene, -hex-4-ene, and — oct-1 -ene.
  • Exemplary branched chain alkenyls include, but are not limited to, -2-methyl-but-2-ene, - l-methyl-hex-4-ene, and -4-ethyl-oct-l-ene.
  • An alkenyl group can be substituted or unsubstituted.
  • An alkenyl group may be designated as having a certain number of carbon atoms. For example, an alkenyl group having from 2 to 8 carbon atoms may be designated as a C 2 -C 8 alkenyl group whereas an alkenyl group having from 2 to 6 carbon atoms may be designated as a C 2 -C 6 alkenyl group.
  • alkynyl means an alkyl group in which one or more carbon-carbon single bonds is replaced with an equivalent number of carbon-carbon triple bonds.
  • An alkynyl group must comprise at least two carbon atoms, and can be substituted or unsubstituted.
  • An alkynyl group may be designated as having a certain number of carbon atoms. For example, an alkynyl group having from 2 to 8 carbon atoms may be designated as a C 2 -C 8 alkynyl group whereas an alkynyl group having from 2 to 6 carbon atoms may be designated as a C 2 -Ce alkynyl group.
  • halo means a halogen atom such as a fluorine, chlorine, bromine, or iodine atom (-F, -Cl, -Br, or —1).
  • haloalkyl means an alkyl group in which one or more hydrogens has been replaced by a halogen atom.
  • a halogen atom is a fluorine, chlorine, bromine, or iodine atom.
  • the number of halogen atoms in a haloalkyl group may range from one to (2m' + 1), where m' is the total number of carbon atoms in the alkyl group.
  • haIo(Ci.C 4 )alkyr' is meant to include trifluoromethyl, 2,2,2-trifluoroethyl, 4-chlorobutyl, 3-bromopropyl, and the like.
  • haloalkyl includes monohaloalkyl (alkyl substituted with one halogen atom) and polyhaloalkyl (alkyl substituted with halogen atoms in a number ranging from two to (2m' + 1) halogen atoms).
  • perhaloalkyl means, unless otherwise stated, an alkyl substituted with (2m' + 1) halogen atoms, where m' is the total number of carbon atoms in the alkyl group.
  • perhaIo(Ci-C 4 )alkyI is meant to include trifluoromethyl, pentachloroethyl, l,l,l-trifluoro-2-bromo-2-chloroethyl, and the like.
  • cyano means a -C- ⁇ N group.
  • nitro means a -NO 2 group.
  • hydroxy and "hydroxyl” mean an -OH group.
  • hydroxyalkyl means an alkyl group in which one or more hydrogens has been replaced with a hydroxy] group.
  • hydroxyalkenyl means an alkenyl group in which one or more hydrogens has been replaced with a hydroxyl group.
  • hydroxyalkynyl means an alkynyl group in which one or more hydrogens has been replaced with a hydroxyl group.
  • alkoxy means a structure of the formula — O-alkyl where alkyl has the meaning set forth above.
  • haloalkoxy means an alkoxy group in which one or more hydrogen is replaced by a halogen atom.
  • hydroxyalkoxy means an alkoxy group in which one or more hydrogen is replaced by a hydroxy group.
  • alkylsulfonyl means a structure of the formula -S(O) 2 -alkyI.
  • amino means an — NH 2 group.
  • alkylamino and dialkylamino mean a structure of the formula -NH-alkyl and -N(alkyl)alkyl, respectively, wherein the alkyl is as defined above.
  • the alkyl groups in dialkylamino groups may be the same or different.
  • alkanoyl alone or in combination with another term, means a radical of the type "R-C(O) — " wherein “R” is an alkyl radical as defined above and "-
  • C(O) — " is a carbonyl radical.
  • alkanoyl radicals include, but are not limited to, acetyl, trifluoroacetyl, hydroxyacetyl, propionyl, butyryl, valeryl, 4- methylvaleryl, and the like.
  • alkanoylamino and “alkanoyloxy” mean -NH- alkanoyl and -O-alkanoyl, respectively.
  • alkoxy carbonyl amino means a structure of the formula
  • alkylsulfonyl amino means a structure of the general formula
  • Carbocyclic ring system and “carbocyclic” mean a ring system in which all the ring members are carbon atoms.
  • Carbocyclic ring systems typically include from 3 to 14 ring atoms.
  • Carbocyclic ring systems may be aromatic or may be non-aromatic.
  • Carbocyclic ring systems include cycloalkyl rings and may also include fused ring systems. Examples of fused ring carbocyclic ring systems include, but are not limited to, decalin, norbornane, tetrahydronaphthalene, naphthalene, indene, and adamantane.
  • the ring atoms in a carbocyclic ring system may be substituted or unsubstituted.
  • heterocyclic ring system As used herein, the terms "heterocyclic ring system”, “heterocyclic” and
  • heterocyclyl means a carbocyclic ring system in which at least one ring atom is a heteroatom such as a N, O, S, or Si. In some embodiments, the heterocyclic ring system includes from 1 to 4 heteroatoms. In some embodiments, the heteroatom is selected from N, O, or S.
  • Heterocyclic ring systems may include one ring or may include fused ring systems. By way of nonlimiting example, heterocycic ring systems may include two six membered rings that are fused to one another or may include one five membered ring and one six membered ring that are fused to one another. Heterocyclic ring systems may be aromatic or may be non-aromatic and may be unsaturated, partially unsaturated, or saturated. The ring atoms in a heterocyclic ring system may be substituted or unsubstituted.
  • aryl means a carbocyclic ring or ring system containing from 6 to 14 ring atoms wherein at least one ring is aromatic.
  • the ring atoms of a carbocyclic aryl group are all carbon atoms.
  • Aryl groups include mono-, bi-, and tricyclic groups as well as benzo-fused carbocyclic moieties such as, but not limited to, 5,6,7,8-tetrahydronaphthyl and the like.
  • the aryl group is a monocyclic ring or is a bicyclic ring.
  • aryl groups include, but are not limited to, phenyl, tolyl, anthracenyl, fluorenyl, indenyl, azulenyl, phenanthrenyl and naphthyl.
  • An aryl group can be unsubstituted or substituted.
  • heteroaryl means an aryl group in which one or more, but not all, of the ring carbon atoms in any ring, whether aromatic or not, is replaced by a hetero atom.
  • pyridine is a heteroaryl group as is a compound in which benzene is fused to a nonaromatic ring that includes at least one heteroatom.
  • Exemplary heteroatoms are N, O, S, and Si.
  • the heteroatoms are N, O, or S.
  • a heteroaryl group can be unsubstituted or substituted.
  • aryl and heteroaryl groups include phenyl, 1 -naphthyl, 2-naphthyl, 4-biphenyl, 1-pyrrolyl, 2-pyrrolyl, 3- pyrrolyl, 1-pyrazolyl, 3-pyrazolyl, 5-pyrazolyl, 2-imidazolyl, 4-imidazolyl, pyrazinyl, 2- oxazolyl, 4-oxazolyl, 2-phenyl-4-oxazolyl, 5-oxazolyl, 3-isoxazolyl, 4-isoxazolyI, 5- isoxazolyl, 2-thiazolyI, 4-thiazolyl, 5-thiazolyl, 2-furanyl, 3-furanyI, dibenzofuryl, 2- thienyl (2-thiophenyl), 3-thienyl (3-thiophenyl
  • Non-limiting examples of other heteroaryl groups include pyridyl, indazolyl, isoquinolinyl, thiazolopyridinyl, benzothiazolonyl, dihydroquinolinonyl, benzoisoxazolyl, benzooxazolonyl, indolinonyl, benzoimidazolonyl, phthalazinyl, naphthyridinyl, thienopyridinyl, benzodioxolyl, isoindolinonyl, quinazolinyl, or cinnolinyl.
  • cycloalkyl means an unsaturated or saturated hydrocarbon that forms at least one ring, having from 3 to 20 ring carbon atoms, and in some embodiments, from 3 to 10 ring, from 3 to 8, or from 3 to 6 carbon atoms.
  • the rings in a cycloalkyl group are not aromatic.
  • a cycloalkyl group can be unsubstituted or substituted.
  • compounds of the invention may optionally be substituted with one or more substituents, such as are illustrated generally above, or as exemplified by particular classes, subclasses, and species of the invention. It will be appreciated that the phrase “optionally substituted” is used interchangeably with the phrase “substituted or unsubstituted.” In general, the term “substituted”, whether preceded by the term “optionally” or not, refers to the replacement of hydrogen radicals in a given structure with the radical of a specified substituent.
  • an optionally substituted group may have a substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position. Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds.
  • PKT protein kinase B, also known as AKT.
  • preventing refers to the ability of a compound or composition of the invention to prevent a disease identified herein in mammals diagnosed as having the disease or who are at risk of developing such disease. The term also encompasses preventing further progression of the disease in mammals that are already suffering from or have symptoms of the disease.
  • mamal refers to non-human animals or humans.
  • the term "patient” or “subject” means an animal (e.g., cow, horse, sheep, pig, chicken, turkey, quail, cat, dog, mouse, rat, rabbit, guinea pig, etc.) or a mammal, including chimeric and transgenic animals and mammals.
  • the term "patient” or “subject” preferably means a monkey or a human, most preferably a human.
  • the patient or subject is afflicted by a cancer.
  • a "therapeutically effective amount” refers to an amount of a compound of the invention, or prodrug thereof, sufficient to provide a benefit in the treatment or prevention of a condition or disease such as cancer, to delay or minimize symptoms associated with the condition or disease, or to cure or ameliorate the disease or cause thereof.
  • a therapeutically effective amount means an amount sufficient to provide a therapeutic benefit in vivo.
  • the term preferably encompasses a non-toxic amount that improves overall therapy, reduces or avoids symptoms or causes of disease, or enhances the therapeutic efficacy of or synergies with another therapeutic agent.
  • a prophylactically effective amount refers to an amount of a compound of the invention or other active ingredient sufficient to result in the prevention of a condition or disease such as cancer, or recurrence or metastasis of cancer.
  • a prophylactically effective amount may refer to an amount sufficient to prevent initial disease or the recurrence or spread of the disease.
  • the term preferably encompasses a non-toxic amount that improves overall prophylaxis or enhances the prophylactic efficacy of or synergies with another prophylactic or therapeutic agent.
  • combination refers to the use of more than one prophylactic and/or therapeutic agents simultaneously or sequentially.
  • the agents may be selected and administered in such a manner that their respective effects are additive or synergistic.
  • the term "pharmaceutically acceptable salts” refers to salts prepared from pharmaceutically acceptable non-toxic acids or bases including inorganic and organic acids and bases.
  • the desired pharmaceutically acceptable salt may be prepared by any suitable method available in the art, for example, treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, or with an organic acid, such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, a pyranosidyl acid, such as glucuronic acid or galacturonic acid, an alpha- hydroxy acid, such as citric acid or tartaric acid, an amino acid, such as aspartic acid or glutamic acid, an aromatic acid, such as benzoic acid or c
  • the desired pharmaceutically acceptable salt may be prepared by any suitable method, for example, treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary or tertiary), an alkali metal hydroxide or alkaline earth metal hydroxide, or the like.
  • suitable salts include organic salts derived from amino acids, such as glycine and arginine, ammonia, primary, secondary, and tertiary amines, and cyclic amines, such as piperidine, morpholine and piperazine, and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.
  • the neutral forms of the compounds may be regenerated from the salt by contacting the salt with a base or acid and isolating the parent compound in the conventional manner.
  • the parent form of the compound differs from the various salt forms in certain physical properties, such as solubility in polar solvents, but otherwise the salts are equivalent to the parent form of the compound for the purposes of the invention.
  • the invention provides compounds which are in a. prodrug form.
  • prodrug is intended to mean any chemical entity that, after administration, is converted to a different therapeutically effective chemical entity.
  • Prodrugs of the compounds described herein are those compounds that readily undergo chemical changes under physiological conditions to provide the compounds of the invention.
  • prodrugs can be converted to the compounds of the invention by chemical or biochemical methods in an ex vivo environment.
  • prodrugs can be slowly converted to the compounds of the invention when placed in a transdermal patch reservoir with a suitable enzyme or chemical reagent.
  • Prodrugs are often useful because, in some situations, they may be easier to administer than the parent drug. They may, for instance, be bioavailable by oral administration whereas the parent drug is not.
  • the prodrug may also have improved solubility in pharmaceutical compositions over the parent drug.
  • a wide variety of prodrug derivatives are known in the art, such as those that rely on hydrolytic cleavage or oxidative activation of the prodrug.
  • prodrug a compound of the invention which is administered as an ester (the "prodrug"), but then is metabolically hydrolyzed to the carboxylic acid, the active entity.
  • prodrug a compound of the invention which is administered as an ester (the "prodrug"), but then is metabolically hydrolyzed to the carboxylic acid, the active entity.
  • Additional examples include peptidyl derivatives of a compound.
  • solvate refers to a compound of the present invention or a salt thereof, that further includes a stoichiometric or non-stoichiometric amount of solvent bound by non-covalent intermolecular forces. Where the solvent is water, the solvate is a hydrate.
  • the compounds of this invention may contain one or more asymmetric centers and thus occur as racemates and racemic mixtures, scalemic mixtures, single enantiomers, individual diastereomers, and diastereomeric mixtures. All such isomeric forms of these compounds are expressly included in the present invention.
  • the term "optically pure” or "stereomerically pure” means a composition that comprises one stereoisomer of a compound and is substantially free of other stereoisomers of that compound. For example, a stereomerically pure compound having one chiral center will be substantially free of the opposite enantiomer of the compound.
  • a typical stereomerically pure compound comprises greater than about 80% by weight of one stereoisomer of the compound and less than about 20% by weight of other stereoisomers of the compound, more preferably greater than about 90% by weight of one stereoisomer of the compound and less than about 10% by weight of the other stereoisomers of the compound, even more preferably greater than about 95% by weight of one stereoisomer of the compound and less than about 5% by weight of the other stereoisomers of the compound, and most preferably greater than about 97% by weight of one stereoisomer of the compound and less than about 3% by weight of the other stereoisomers of the compound.
  • This invention encompasses the use of stereomerically pure forms of such compounds, as well as the use of mixtures of those forms.
  • mixtures comprising equal or unequal amounts of the enantiomers of a particular compound of the invention may be used in methods and compositions of the invention.
  • These isomers may be asymmetrically synthesized or resolved using standard techniques such as chiral columns or chiral resolving agents. See, e.g., Jacques, J., et ai, Enantiomers, Racemates and Resolutions (Wiley-lnterscience, New York, 1981); Wilen, S. H., et al. (1997) Tetrahedron 33:2725; EHeI, E. L., Stereochemistry of Carbon Compounds (McGraw-Hill, NY, 1962); and Wilen, S. H., Tables of Resolving Agents and Optical Resolutions p. 268 (E.L. EHeI, Ed., Univ. of Notre Dame Press, Notre Dame, IN, 1972).
  • the compounds of the invention may exhibit the phenomenon of tautomerism. While the structural formulas set forth herein cannot expressly depict all possible tautomeric forms, it is to be understood that these structures are intended to represent all tautomeric forms of the depicted compound and are not to be limited merely to the specific compound form depicted by the formula drawings. [00109J Certain compounds of the invention may exist in multiple crystalline or amorphous forms. In general, all physical forms are equivalent for the uses contemplated by the invention and are intended to be within the scope of the invention. [00110] The compounds of the invention may also contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds.
  • the compounds may be radiolabeled with radioactive isotopes, such as for example tritium ( 3 H), iodine-125 ( 125 I) or carbon-14 ( 14 C).
  • Radiolabeled compounds are useful as therapeutic or prophylactic agents, research reagents, e.g., assay reagents, and diagnostic agents, e.g., in vivo imaging agents. All isotopic variations of the compounds of the invention, whether radioactive or not, are intended to be encompassed within the scope of the invention.
  • the compounds described herein are useful for treating diseases or conditions mediated by various kinases such as PKB.
  • the invention encompasses the therapeutic use of such compounds and compositions thereof in the treatment of disease states associated with abnormal cell growth, such as cancer, or metabolic disease states, such as diabetes, or inflammation.
  • the invention further provides pharmaceutical compositions that include the compounds of the invention and the use of the compounds in the preparation of medicaments or pharmaceutical formulations or compositions for treating various conditions and disease states.
  • the invention comprises a compound of Formula I
  • Y is -N(R 5 )R 6 or -OR 6 ;
  • X is O, S, or-N(R 7 );
  • R 1 is R 8 , -CHR"-N(H)-R 8 , -CHR 1 '-O-R 8 , C 2 -C 6 alkynyl, C 2 -C 6 hydroxyalkynyl, or
  • R 2 is aryl or heteroaryl
  • R 3 is — H, Ct-C 6 alkyl which may be interrupted by one or more hetero atoms,
  • R 4 is Ci-C 6 alkyl which may be interrupted by one or more hetero atoms, -(CR 9 R 10 ) t (aryl),
  • R 5 is -H, C 1 -C 8 alkyl, -C(O)(CR 9 R 10 ),)N(R 7 ) 2 , -C(O)(CR 9 R 10 ),, -C(O) 2 (CR 9 R 10 ),, -(CR 9 R l0 ),(aryl), -(CR 9 R 1 °),(heteroaryl), -(CR 9 R l0 ) t (cycloalkyl), or -(CR 9 R 10 ),(heterocycIyl), or R 4 and R 5 join to form a C 3 -C] 0 heterocyclic ring;
  • R 6 and R 7 are independently selected from -H, Ci-Cs alkyl, -(C]-C 6 alkyl)aryl, or -C(O)(C]-C 6 alkyl), or R ⁇ and R 7 , together with the atoms to which they are linked, join to form a 5 to 6-membered heterocyclic ring, or
  • R 5 and R 6 together with the nitrogen atom to which they are linked, join to form a 5 to 6- membered heterocyclic or heteroaryl ring;
  • R 8 is -H, CpC 6 alkyl, -(Cj-C 6 alkyl)aryl, aryl, or heteroaryl; and R 9 , R 10 , and R 11 are independently selected from — H, CpC 6 alkyl, or aryl; wherein n is an integer from 1 to 6; m is an integer from 0 to 2; and each t is independently an integer from 0 to 3; wherein each of the above alkyl, aryl, heteroaryl, cycloalkyl, and heterocyclyl moieties and heterocyclic and carbocyclic rings are optionally and independently substituted by 1 - 3 substituents selected from amino, aryl, heteroaryl, cycloalkyl, or heterocyclyl optionally substituted by 1-5 substituents selected from C i -Ce alkoxy,
  • C 1 -C 6 alkyl optionally substituted by halo, aryl, halo, hydroxyl, heteroaryl,
  • Ci-C 6 hydroxyalkyl or -NHS(O) 2 -(Ci-C 6 alkyl);
  • the invention comprises a compound of Formula I, wherein A is
  • the invention comprises a compound of Formula
  • the invention comprises a compound of Formula
  • X is -N(R 7 )
  • Y is -N(R 5 )(R 6 )
  • m, n, and t are 1.
  • the invention comprises a compound of Formula
  • X is -N(R 7 )
  • Y is -N(R 5 )(R 6 )
  • R 2 is heteroaryl
  • R 3 is -H
  • R 4 is -(CR 9 R 10 ),(aryl) or -(CR 9 R l0 ) t (heteroaryl)
  • m, n, and t are 1, R 5 , R 6 , and R 7 are -H
  • R 9 and R 10 are independently selected from H or Q-C3 alky I.
  • the invention comprises a compound of Formula
  • X is -N(R 7 )
  • Y is -N(R 5 )(R 6 )
  • R 2 is bicyclic heteroaryl
  • R 3 is -H
  • R 4 is -(CR 9 R 10 ),(monocyclic aryl) or -(CR 9 R 10 ) t (bicyclic heteroaryl)
  • m, n, and t are 1
  • R 5 , R 6 , R 7 , R 9 and R 10 are -H.
  • the invention comprises a compound of Formula
  • R 2 is bicyclic heteroaryl
  • R 3 is -H
  • R 4 is -(CR 9 R 10 ),(monocyclic aryl) or -(CR 9 R l0 ) t (bicyclic heteroaryl)
  • m, n, and t are 1, and R 5 , R 6 , R 7 , R 9 and R 10 are — H
  • the bicyclic heteroaryl group of R 2 is isoquinolinyl, lH-indazolyl, thiazolo[5,4-c]pyridinyl, benzo[d]thiazole-2(3H)-onyl, phthalazinyl, indolin-2-onyl, 3,4-dihydroquinolin-2(lH)-onyl, benzo[d]isoxazolyl, benzo[d]oxazol- 2(3H)-on
  • the bicyclic heteroaryl group of R 2 is isoquinoIin-6-yl, 3-aminoisoquinolin-6-yl, lH-indazol-5-yl, 1H- indazol-6-yl, 3-amino-l H-indazol-5-yl, 3-amino-lH-indazol-6-yl, 3-amino-l-methyl-lH- indazol-6-yl, 3-methylamino-lH-indazol-5-yI, 3-methyl-lH-indazol-5-yl, thiazolo[5,4- c]pyridin-2-yl, benzo[d]thiazo!e-2(3H)-on-6-yl, l-hydroxyphthalazin-6-yl, phthalazin-6- yl, indolin-2-on-5 ⁇ yl, 3-methylindolin-2-on-5-yl, 3-(furan-2-ylmethylene)indolin-2
  • the monocyclic aryl group of R 4 is phenyl, 3-chlorophenyl, 4-chIorophenyl, 4-methoxyphenyI, 3-(trifluoromethyl)phenyl, or 4-(trifluoromethyl)phenyl, or the bicyclic heteroaryl group of R 4 is l H-indol-3-yl.
  • the invention comprises a compound of Formula
  • R 2 is bicyclic heteroaryl
  • R 3 is -H
  • R 4 is -(CR 9 R 10 ),(monocyclic aryl), m, n, and t are 1, and R s , R 6 , R 7 , R 9 and R 10 are -H
  • the bicyclic heteroaryl group of R 2 is isoquinolin-6-yl, 3-aminoisoquinolin-6-yl., 1H- indazol-5-yl, 3-methyl-lH-indazol-5-yl, thiazolo[5,4-c]py ⁇ din-2-yl, benzo[d]oxazol- 2(3H)-on-6-yl, or l,6-naphthyridin-2-yl, and the monocyclic aryl group of R 4 is 4- chlorophenyl, 3-(trifIuoro
  • the invention comprises a compound of Formula 1 having any of the features of any of the embodiments described above in which R 1 is -H, -CH 3 , -CH 2 CH 3 , -CH 2 OCH 3 , -CH 2 OCH 2 CH 3 , -CH 2 OH, -CH 2 OCH 2 CF 3 , -CH 2 N(H)CH 3 , -CH(CH 3 )OCH 3 , furanyl, phenyl, pyridyl, or -C ⁇ N.
  • R 1 is - H, -CH 3 , -CH 2 OCH 3 , -CH 2 OCH 2 CH 3 , -CH 2 OH, or furan-2-yl.
  • R 1 is -CH 3 , -CH 2 CH 3 , -CH 2 OCH 3 , -CH 2 OCH 2 CH 3 , -CH 2 OH, -CH 2 OCH 2 CF 3 , -CH 2 N(H)CH 3 , -CH(CH 3 )OCH 3 , furanyl, phenyl, pyridyl, or -CsN.
  • R 1 is -CH 3 , -CH 2 OCH 3 , -CH 2 OCH 2 CH 3 , -CH 2 OH, or furan-2-yl.
  • R 1 is -(C 1 -C 6 alkyl), -(C 1 -C 6 alkyl)aryl, aryl, heteroaryl, -CHR ! '-N(H)-R 8 , -CHR 1 '-O-R 8 , or -C ⁇ N.
  • R 1 is -(C r C 6 alkyl), aryl, heteroaryl, -CHR 1 '-N(H)- R 8 , -CHR"-O-R 8 , or -C ⁇ N. In still other such embodiments, R 1 is -(Ci-C 6 alkyl), heteroaryl, or -CHR U -O-R 8 .
  • the invention comprises a compound of Formula
  • the invention comprises a compound of Formula
  • the invention comprises a compound of Formula I, wherein A is aryl, X is -N(R 7 ), R 2 is a bicyclic heteroaryl, m is 1 , and R 7 is -H.
  • the invention comprises a compound of Formula
  • A is a monocyclic aryl
  • X is -N(R 7 )
  • R 1 is -H
  • R 2 is thiazolo[5,4-c]pyridin-2-yl
  • m is 1, and R 7 is -H.
  • the invention comprises a compound of Formula
  • the invention provides a compound of Formula II
  • R 1 is -H, halo, -OR 8 , C 1 -C 6 alkyl, -(C 1 -C 6 aIkyl)-O-R 8 , -(C 1 -C 6 haloalkyl)-O-R 8 , -(C 2 -C 6 aIkenyl)-O-R s , -(C 1 -C 6 alkyl)N(R 7 ) 2 , -(C 1 -C 6 alkyl)aryl, -C(O)R 8 , -C(O)O-R 8 , -C(O)O-R 8 ,
  • -C N, -(C 2 -C 6 alkynyl)(C 3 -C 3 cycloalkyl), -(C 2 -C 6 alkynyl)(C 5 -C 8 cycloalkenyl), -(C 2 -C 6 alkynyI)-N(R 7 )S(O) 2 -R 8 , aryl, heteroaryl, cycloalkyl, or heterocyclyl;
  • R 2 is a carbocyclic ring system or is a heterocyclic ring; system; R 5 is -H, C 1 -C 8 alkyl, -C(O)(CR 9 R 1 °),)N(R 7 ) 2 , -C(O)(CR 9 R 10 ),, -C(O) 2 (CR 9 R 1 ⁇ -(CR 9 R 10 ),(aryl), -(CR 9 R t0 ),(heteroaryl), -(CR 9 R 10 ),(cycloalkyl), or -(CR 9 R'°),(heterocycIyl);
  • R 6 and R 7 are independently selected from -H, C r C 8 alkyl, -(CpC 6 alkyl)aryl, or -C(O)(C 1 -C 6 alkyl);
  • R 8 is selected from -H, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, -(C]-C 6 alkyl)aryl, aryl, heteroaryl, C-C 6 hydroxyalkyl, or -(C r C 6 alkyl)-O-(C,-C 6 alkyl), cycloalkyl, or heterocyclyl;
  • R 9 and R 10 in each instance, and R 11 are independently selected from -H, Ci-C 6 alkyl, or aryl;
  • R 12 is -H, -OR 8 , -0-(C 1 -C 6 alkyl)-O-R 8 , C 1 -C 6 alkyl, C 1 -C 6 alkenyl, -(C 1 -C 5 alkyl)-O-R s , Or -(C 1 -C 6 alkyl)-O-C(O)-R 8 ;
  • R' 3 is -H, or C 1 -C 6 alkyl;
  • R 14 is -H, -OR 8 , -0-(C 1 -C 6 C r C 6 alkyl, C 1 -C 6 alkenyl, -(C 1 -C 6 alkyl)-O-R s , or -(C 1 -C 6 alky I)-O-C(O)-R 8 ; each t is independently selected from O, 1, 2, or 3; and Z is aryl or heteroaryl; wherein each of the above alkyl, aryl, heteroaryl, cycloalkyl, and heterocyclyl moieties and heterocyclic and carbocyclic rings are optionally and independently substituted by 1- 3 substituents selected from amino, aryl, heteroaryl, cycloalkyl, or heterocyclyl optionally substituted by 1-5 substituents selected from Ci-C 6 alkoxy,
  • C)-C 6 alkyl optionally substituted by halo, aryl, halo, hydroxy 1, heteroaryl,
  • Ci-C 6 hydroxyalkyl or -NHS(O) 2 -(C 1 -C 6 alkyl);
  • R 1 is -H, halo, -OR 8 , C 1 -C 6 alkyl, -(C 1 -C 6 alkyl)-O-R ⁇ -(C 1 -C 6 haloalkyl)-O-R s , -(C 2 -C 6 alkenyl)-O-R s , -(C r C 6 alkyl)N(R 7 ) 2 , -(Ci-C 6 alkyl)aryl, -C(O)R 8 , -C(O)O-R 8 , -C(O)N(R 7 ) 2 , -CHR" -N (H)-R 8 , -CHR 1 '-O-R 8 , C 2 -C 6 alkynyl, (C 2 -C 6 alkynyl)-O-R s , -C ⁇ N, -(C 2 -C 6 alkynyl)(C 3 -C
  • R 6 and R 7 are independently selected from — H, Ci-C 8 alkyl, -(Ci-C 6 alkyl)aryl, Or -C(O)(C 1 -C 6 alkyl);
  • R 8 is selected from -H, C 1 -C 6 alkyl, C r C 6 haloalkyl, -(CpC 6 alkyl)aryl, aryl, heteroaryl, Ci-C 6 hydroxyalkyl, or -(C r C 6 alkyl)-O-(CrC 6 alkyl), cycloalkyl, or heterocyclyl;
  • R 9 and R 10 in each instance, and R 1 1 are independently selected from -H, Ci-C 6 alkyl, or aryl;
  • R 12 is -H, -OR 8 , -O-(C,-C 6 alkyl)-O-R 8 , C 1 -C 6 alkyl, C 1 -C 6 alkenyl, -(C 1 -C 6 alkyl)-O-R 8 , or -(C-C 6 alkyl)-O-C(O)-R s ;
  • R 13 is -H, or C 1 -C 6 alkyl;
  • R 14 is -H, -OR 8 , -0-(C 1 -C 6 alkyl)-O-R 8 , C 1 -C 6 alkyl, C 1 -C 6 alkenyl, -(C 1 -C 6 alkyl)-O-R 8 , or -(Ci-C 6 alkyl)-O-C(O)-R 8 ; each t is independently selected from 0, 1 , 2, or 3; and
  • Z is aryl or heteroaryl optionally substituted with from 1-3 substituents independently selected from -F, -Cl, -Br, -I, -CN, -NH 2 , -NO 2 , -CF 3 , -OH, -0-(C 1 -C 6 alkyl), -0-(C 1 -C 6 alkyl)-Cl, -0-(C 1 -C 6 alkyl)-OH, -C 1 -C 6 alkyl, -OCF 3 , -NH(C 1 -C 6 alkyl), -N(C 1 -C 6 alkyl) 2 ,
  • the compound of Formula II has the Formula ILA
  • the compound of Formula II has the Formula HB
  • the compound of Formula II has the Formula HC
  • the compound of Formula II has the Formula HD
  • the compound of Formula II has the Formula HE
  • R 1 is -H.
  • R 12 is — H or Cj-C 6 alkyl. In some such embodiments, R 12 is — H or methyl.
  • R 13 is -H.
  • R 14 is — H.
  • R 14 is -OR 8 .
  • R 14 is selected from -H, methyl, ethyl, propyl, ethenyl, propenyl, hydroxymethyl, methoxymethyl, -CH 2 -O-C(O)-(C 1 -C 6 alkyl), 1 -hydroxyethyl,or methoxymethoxy.
  • Z is selected from optionally substituted phenyl, optionally substituted indolyl, optionally substituted naphthyl, optionally substituted pyridyl, or optionally substituted thiophenyl.
  • Z is selected from phenyl, indolyl, naphthyl, pyridyl, or thiophenyl, each of which is optionally substituted with 1-3 substituents selected from -Cl, -F, -CF 3 , -OH, -0-(C 1 -C 6 alkyl), -0-(C 1 -C 6 alkyl)-CI, -0-(C 1 -C 6 alkyl)-OH, -C 1 -C 6 alkyl, -OCF 3 ,
  • Z is selected from phenyl, indolyl, naphthyl, pyridyl, thiophenyl, 4-chIorophenyl, 4-trifluormethylphenyl, 3- chlorophenyl, 3-trifluoromethylphenyl, 4-methoxyphenyl, 3-fluoro-4- trifluoromethylphenyl, 4-chloro-3-fluorophenyl, 4-(3-chloropropoxy)phenyl, 4-(3- hydroxypropoxy)phenyl, 3,4-dichlorophenyl, 4-fluorophenyl, 2,4-dichloro ⁇ henyl, 4- methylphenyl, 3,4-difluorophenyl, 3-fluoro-4-methoxyphenyl, 3,5-difluorophenyl, 6- trifluoromethylpyridin-3-yl, 5-methoxy-6-trifluoromethylpyridin-3-yl,
  • R 7 is H. In some such embodiments, R 5 and R 6 are both H.
  • R s and R 6 are both
  • R 12 is — H Or C 1 -C 6 alkyl
  • R 13 is -H
  • R 14 is -H
  • -OR 8 -0-(C 1 -C 6 alkyl)-O-R ⁇ C 1 -C 6 alkyl, C 1 -C 6 alkenyl
  • R 14 is '
  • R 5 , R 6 , and R 7 are all H.
  • C 1 -C 6 haloalkyl C 1 -C 6 hydroxyalkyl, Ci-C 6 alkoxy, C 1 -C 6 haloalkoxy, C,-C 6 hydroxyalkoxy,Ci-C 6 alkylamino, C 2 -C 6 alkenyl, -(C 2 -C 4 alkenyl)heterocyclyl, or -(C 2 -C 4 alkenyl)cycloalkyl.
  • the carbocyclic ring system or the heterocyclic ring system of R 2 includes at least one aromatic ring.
  • the aromatic ring may be carbocyclic, or the aromatic ring may include one or more heteroatoms such as, but not limitede to, in pyridine or pyrimidine rings.
  • the at least one aromatic ring in the carbocyclic ring system or the heterocyclic ring system of R 2 is a phenyl ring or is a pyridyl ring.
  • the carbocyclic ring system or the heterocyclic ring system of R 2 includes an aromatic ring, and the aromatic ring is bonded to the thiazole shown in the structure of Formula II.
  • the carbocyclic ring system or the heterocyclic ring system of R 2 is a fused ring system comprising at least two rings.
  • R 2 is a heterocyclic ring system comprising two six membered rings or one six membered ring and one 5 membered ring.
  • R 2 is selected from optionally substituted phenyl, pyridyl, indazolyl, isoquinolinyl, thiazolopyridinyl, benzothiazolonyl, dihydroquinolinonyl, benzoisoxazolyl, benzooxazolonyl, indolinonyl, benzoimidazolonyl, phthalazinyl, naphthyridinyl, thienopyridinyl, benzodioxolyl, isoindolinonyl, quinazolinyl, or cinnolinyl.
  • R 2 is selected from isoquinolinyl, lH-indazolyl, thiazolo[5,4-c]pyridinyl, benzo[d]thiazo!e-2(3H)-onyl, phthalazinyl, indolin-2-onyl, 3,4-dihydroquinolin-2(l H)-onyl, benzo[d]isoxazolyl, benzo[d]oxazol-2(3H)-onyl, benzo[d]imidazol-2(3H)-onyl, 1,6-naphthyridinyI, quinazolin-7-yl, or cinnolin-6-yl.
  • R 2 is isoquinolin-6-yl, 3- aminoisoquinolin-6-yL, l H-indazol-5-yl, 1 H-indazol-6-yl, 3-amino-lH-indazol-5-yl, 3- amino-lH-indazol-6-yI, 3-amino-l-methyl-lH-indazol-6-yI, 3-methylamino-lH-indazol- 5-yl, 3-methyl-lH-indazol-5-yl, thiazolo[5,4-c]pyridin-2-y!, benzo[d]thiazole-2(3H)-on- 6-yl, 1 -hydroxy ⁇ hthalazin-6-yl, phthalazin-6-yl, indolin-2-on-5-yl, 3-methylindolin-2-on- 5-yl, 3-(furan-2-ylmethylene)indolin-2-on-5-yl, 3-(l H-imi
  • R 2 is selected from one of the following groups which may optionally be substituted and where the wavy line indicates the point of attachment to the thiazole:
  • R 2 is selected from one of the following groups, where the wavy line indicates the point of attachment to the thiazole:
  • R 1 is selected from
  • the invention comprises one or more compound selected from any one or all of Examples 1 — 255 shown in Table 1, or a pharmaceutically acceptable salt, hydrate, or stereoisomer thereof.
  • Each of the different groups of Examples 1 — 255 that correspond to any of the variables in the compounds of Formula I and/or Formula II is preferred.
  • the invention comprises a pharmaceutically acceptable salt, hydrate, or solvate of a compound of Fo ⁇ nula I or Formula II or any of the compounds of any of the embodiments described herein.
  • the pharmaceutically acceptable salt is selected from a chloride or trifluoroacetate salt.
  • the salt is an ammonium trifluoroacetate, ammonium chloride, or hydrochloride salt.
  • compositions and individual dosage forms of the invention may be suitable for oral, mucosal (including sublingual, buccal, rectal, nasal, or vaginal), parenteral (including subcutaneous, intramuscular, bolus injection, intra-arterial, or intravenous), transdermal, or topical administration.
  • compositions and dosage forms of the invention typically also comprise one or more pharmaceutically acceptable carrier, excipient, or diluent. Sterile dosage forms are also contemplated.
  • composition as used herein is intended to encompass a product comprising the specified ingredients (and in the specified amounts, if indicated), as well as any product which results, directly or indirectly, from combination of the specified ingredients.
  • pharmaceutically acceptable carrier, excipient, or diluent means that the carrier, excipient, or diluent is compatible with the other ingredients of the formulation and is not deleterious to the recipient thereof.
  • Composition formulation may improve one or more pharmacokinetic properties (e.g., oral bioavailability, membrane permeability) of a compound of the invention (herein referred to as the active ingredient).
  • compositions of the invention may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art. All methods include the step of bringing the active ingredient such as a compound of any of the embodiments into association with the carrier which constitutes one or more accessory ingredients.
  • the pharmaceutical compositions are prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired formulation.
  • the active object compound is included in an amount sufficient to produce the desired effect in the subject.
  • compositions include a Formula I or Formula II compound of the invention, or a pharmaceutically acceptable salt, hydrate or stereoisomer thereof, and at least one additional therapeutic agent.
  • additional therapeutic agents include, but are not limited to, those listed above.
  • Such compositions may include one or more pharmaceutically acceptable carrier, excipient, or diluent.
  • composition, shape, and type of dosage forms of the invention will typically vary depending on their use.
  • a dosage form used in the acute treatment of a disease or a related disease may contain larger amounts of one or more of the active ingredients it comprises than a dosage form used in the chronic treatment of the same disease.
  • a parenteral dosage form may contain smaller amounts of one or more of the active ingredients it comprises than an oral dosage form used to treat the same disease or disorder.
  • dosage forms include, but are not limited to, tablets; caplets; capsules, such as soft elastic gelatin capsules; cachets; troches; lozenges; dispersions; suppositories; ointments; cataplasms (poultices); pastes; powders; dressings; creams; plasters; solutions; patches; aerosols (e.g., nasal sprays or inhalers); gels; liquid dosage forms suitable for oral or mucosal administration to a patient, including suspensions (e.g.
  • aqueous or non-aqueous liquid suspensions oil-in-water emulsions, or a water-in-oil liquid emulsions
  • solutions and elixirs
  • liquid dosage forms particularly suitable for parenteral administration to a patient and sterile solids (e.g., crystalline or amorphous solids) that can be reconstituted to provide liquid dosage forms suitable for parenteral administration to a patient.
  • sterile solids e.g., crystalline or amorphous solids
  • compositions containing the active ingredient may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs.
  • Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions. Such compositions may contain one or more agents selected from sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with other nontoxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • excipients may be, for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid, or talc.
  • the tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate may be employed. They may also be coated by the techniques described in U.S. Patent Nos. 4,256, 108, 4, 160,452, and 4,265,874 to form osmotic therapeutic tablets for control release.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate, or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate, or kaolin
  • water or an oil medium for example peanut oil, liquid paraffin, or olive oil.
  • Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydroxy-propylmethylcellulose, sodium alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxy-ethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbit
  • the aqueous suspensions may also contain one or more preservatives, for example ethyl, or n-propyl, p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose or saccharin.
  • preservatives for example ethyl, or n-propyl, p-hydroxybenzoate
  • coloring agents for example ethyl, or n-propyl, p-hydroxybenzoate
  • coloring agents for example ethyl, or n-propyl, p-hydroxybenzoate
  • flavoring agents for example ethyl, or n-propyl, p-hydroxybenzoate
  • sweetening agents such as sucrose or saccharin.
  • Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil, or coconut oil, or in a mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent, for example beeswax, hard paraffin, or cetyl alcohol. Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • a dispersing or wetting agent e.g., sodium EDTA
  • suspending agent e.g., sodium EDTA
  • preservatives e.g., sodium EDTA, sodium bicarbonate, sodium bicarbonate
  • the pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions.
  • the oily phase may be a vegetable oil, for example olive oil or arachis oil, or a mineral oil, for example liquid paraffin or mixtures of these.
  • Suitable emulsifying agents may be naturally-occurring gums, for example gum acacia or gum tragacanth, naturally-occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate.
  • the emulsions may also contain sweetening and flavoring agents.
  • Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, and flavoring and coloring agents.
  • the pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleagenous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1,3-butane diol.
  • Suitable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • compositions may also be administered in the form of suppositories for rectal administration of the drug.
  • These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritating excipient include, for example, cocoa butter and polyethylene glycols.
  • compositions of the invention for topical use, creams, ointments, jellies, solutions, or suspensions, etc., containing the compounds of the invention are employed. As used herein, topical application is also meant to include the use of mouthwashes and gargles.
  • topical application is also meant to include the use of mouthwashes and gargles.
  • typical dosage forms of the invention comprise a Formula I or Formula II compound of the invention, or a pharmaceutically acceptable salt, hydrate, or stereoisomer thereof in an amount of from 0.1 mg to 1500 mg per unit to provide doses of about 0.01 to 200 mg/kg per day.
  • the invention further provides the use of a compound of Formula 1 or
  • composition or medicament may be used to treat a disease mediated by a kinase such as PKB.
  • the disease is mediated by PKB ⁇ .
  • the disease is cancer and in some such embodiments, the cancer is a solid tumor.
  • the compounds of the invention may be used to treat or prevent various kinase-related disorders.
  • the present invention provides methods for treating or preventing such disorders.
  • the invention provides a method for treating a kinase-mediated disorder in a subject that includes administering a therapeutically effective amount of a compound of any of the embodiments of the invention or a pharmaceutical composition to the subject.
  • the subject is a mammal, and in some such embodiments is a human.
  • the disorder is mediated by IGF- I R, Insulin Receptor, KDR, Tie2, EGFR, PKA, PKB, PKC, FKHR, TSC 1/2, SGK, LCK, BTK, Erk, MSK, MK2, MSK, p38, P70S6K, PIMl, PIM2, ROCK2, GSK3, or a CDK complex.
  • the disorder is mediated by PKB.
  • the administration of the compound or pharmaceutical composition produces selective inhibition of PKB, and in some cases PKB ⁇ , in the subject after administration.
  • the disorder is cancer. The present invention thus provides methods for treating or preventing PKB-mediated disease states, such as cancer.
  • the cancer is a tumor such as a solid tumor.
  • the compounds of the invention may also be used to treat proliferation- related disorders.
  • the invention further provides methods for treating such proliferation-related disorders in a subject. Such methods include administering to a subject in need thereof a therapeutically effective amount of the compound or pharmaceutical composition of any of the embodiments.
  • the subject is a mammal.
  • the mammal is a human.
  • the proliferation-related disorder is abnormal cell growth.
  • the disorder is inflammation or an inflammation-related disorder.
  • the disorder is a metabolic disease such as diabetes.
  • the disorder is cancer.
  • the cancer is a solid tumor.
  • Formula II compound of the invention or a pharmaceutically acceptable salt, solvate, hydrate, or stereoisomer thereof in the acute or chronic treatment or prevention of a cancer or other disease or condition will vary with the nature and aggressiveness of the condition, and the route by which the active ingredient is administered.
  • the dose, and in some cases the dose frequency will also vary according to the condition to be treated, the age, body weight, and response of the individual patient. Suitable dosing regimens can be readily selected by those skilled in the art with due consideration of such factors.
  • the dose administered depends upon the specific compound to be used, and the weight and condition of the patient.
  • the dose per day is in the range of from about 0.001 to 100 mg/kg, preferably about 1 to 25 mg/kg, more preferably about 1 to about 5 mg/kg.
  • about 0.1 mg to about 15 g per day is administered in about one to four divisions a day, preferably 10 mg to 12 g per day, more preferably from 40 mg to 500 mg per day.
  • the compounds of the invention are administered from 40 mg to 500 mg per day in about one to four divisions a day.
  • the recommended daily dose can be administered in cycles as single agents or in combination with other therapeutic agents.
  • the daily dose is administered in a single dose or in equally divided doses.
  • the recommended daily dose can be administered one time per week, two times per week, three times per week, four times per week or five times per week.
  • the compounds of the invention can be administered to provide systemic distribution of the compound within the patient. Therefore, in some embodiments, the compounds of the invention are administered to produce a systemic effect in the body.
  • the compounds of the invention may also be administered directly to a site affected by a condition, as, for example, an in the treatment of an accessible area of skin or an esophageal cancer.
  • the compounds of the invention may be administered via oral, mucosal (including sublingual, buccal, rectal, nasal, or vaginal), parenteral (including subcutaneous, intramuscular, bolus injection, intra-arterial, or intravenous), transdermal, or topical administration.
  • the compounds of the invention are administered via mucosal (including sublingual, buccal, rectal, nasal, or vaginal), parenteral (including subcutaneous, intramuscular, bolus injection, intra-arterial, or intravenous), transdermal, or topical administration.
  • the compounds of the invention are administered via oral administration.
  • the compounds of the invention are not administered via oral administration.
  • Some methods of the invention comprise the administration of a compound of the invention and an additional therapeutic agent (i.e., a therapeutic agent other than a compound of the invention).
  • an additional therapeutic agent i.e., a therapeutic agent other than a compound of the invention.
  • the compounds of the invention can be used in combination with at least one other therapeutic agent.
  • additional therapeutic agents include, but are not limited to, antibiotics, anti-emetic agents, antidepressants, antifungal agents, anti-inflammatory agents, antineoplastic agents, antiviral agents, cytotoxic agents, and other anticancer agents, immunomodulatory agents, alpha-interferons, ⁇ -interferons, alkylating agents, hormones, and cytokines.
  • the invention encompasses administration of an additional therapeutic agent that demonstrates anti-cancer activity.
  • an additional therapeutic agent that demonstrates cytotoxic activity is administered to a subject such as a cancer patient.
  • the compounds of the invention and the other therapeutics agent can act additively or, preferably, synergistically.
  • a composition comprising a compound of the invention is administered concurrently with the administration of another therapeutic agent, which can be part of the same composition or can be in a different composition from the one that comprises the compound of the invention.
  • a compound of the invention is administered prior to, or subsequent to, administration of another therapeutic agent.
  • a compound of the invention is administered to a patient who has not previously undergone or is not currently undergoing treatment with another therapeutic agent.
  • a compound of the invention may be administered to a subject that has had, is currently undergoing, or is scheduled to receive radiation therapy. In some such embodiments, the subject is a cancer patient.
  • the therapeutic agents can be formulated as separate compositions that are administered at the same time or sequentially at different times, or the therapeutic agents can be given as a single composition.
  • co-therapy in defining use of a compound of the present invention and another pharmaceutical agent, is intended to embrace administration of each agent in a sequential manner in a regimen that will provide beneficial effects of the drug combination, and is intended as well to embrace coadministration of these agents in a substantially simultaneous manner, such as in a single capsule having a fixed ratio of these active agents or in multiple, separate capsules for each agent.
  • the administration of compounds of the present invention may be in conjunction with additional therapies known to those skilled in the art in the prevention or treatment of neoplasia, such as with radiation therapy or with cytostatic or cytotoxic agents.
  • antineoplastic agents available in commercial use, in clinical evaluation and in pre-clinical development, which may be selected for treatment of neoplasia by combination drug chemotherapy. Such antineoplastic agents fall into several major categories, namely, antibiotic-type agents, alkylating agents, antimetabolite agents, hormonal agents, immunological agents, interferon-type agents and a category of miscellaneous agents.
  • antibiotic-type agents antibiotic-type agents
  • alkylating agents antimetabolite agents
  • hormonal agents include antimetabolite agents, hormonal agents, immunological agents, interferon-type agents
  • interferon-type agents and a category of miscellaneous agents.
  • a first family of antineoplastic agents which may be used in combination with compounds of the present invention consists of antimetabolite-type/thymidilate synthase inhibitor antineoplastic agents.
  • Suitable antimetabolite antineoplastic agents may be selected from, but are not limited to, the group consisting of 5-FU-fibrinogen, acanthifolic acid, aminothiadiazole, brequinar sodium, carmofur, Ciba-Geigy CGP- 30694, cyclopentyl cytosine, cytarabine phosphate stearate, cytarabine conjugates, Lilly DATHF, Merrel Dow DDFC, dezaguanine, dideoxycytidine, dideoxyguanosine, didox, Yoshitomi DMDC, doxifluridine, Wellcome EHNA, Merck & Co.
  • EX-015 benzrabine, floxuridine, fludarabine phosphate, 5-fluorouracil, N-(2'-furanidyl)-5-fIuorouracil, Daiichi Seiyaku FO-152, isopropyl pyrrolizine, Lilly LY-18801 1 , Lilly LY-264618, methobenzaprim, methotrexate, Wellcome MZPES, norspermidine, NCI NSC-127716, NCI NSC-264880, NCI NSC-39661, NCI NSC-612567, Warner-Lambert PALA, pentostatin, piritrexim, plicamycin, Asahi Chemical PL-AC, Takeda TAC-788, thioguanine, tiazofurin, Erbamont TIF, trimetrexate, tyrosine kinase inhibitors, Taiho UFT, and uricytin.
  • a second family of antineoplastic agents which may be used in combination with compounds of the present invention consists of alkylating-type antineoplastic agents.
  • Suitable alkylating-type antineoplastic agents may be selected from, but are not limited to, the group consisting of Shionogi 254-S, aldo-phosphamide analogues, altretamine, anaxirone, Boehringer Mannheim BBR-2207, bestrabucil, budotitane, Wakunaga CA-102, carboplatin, carmustine, Chinoin-139, Chinoin-153, chlorambucil, cisplatin, cyclophosphamide, American Cyanamid CL-286558, Sanofi CY- 233, cyplatate, Degussa D- 19-384, Sumimoto DACHP(Myr)2, diphenylspiromustine, diplatinum cytostatic, Erba distamycin derivatives, Chugai DWA-21
  • a third family of antineoplastic agents which may be used in combination with compounds of the present invention consists of antibiotic-type antineoplastic agents.
  • Suitable antibiotic-type antineoplastic agents may be selected from, but are not limited to, the group consisting of Taiho 4181-A, aclarubicin, actinomycin D, actinoplanone, Erbamont ADR-456, aeroplysinin derivative, Ajinomoto AN-201- ⁇ 1, Ajinomoto AN-3, Nippon Soda anisomycins, anthracycline, azino-mycin-A, bisucaberin, Bristol-Myers BL-6859, Bristol-Myers BMY-25067, Bristol-Myers BMY- 25551 , Bristol-Myers BMY-26605, Bristol-Myers BMY-27557, Bristol-Myers BMY- 28438, bleomycin sulfate, bryostatin- 1 , Taiho C- 1027,
  • a fourth family of antineoplastic agents which may be used in combination with compounds of the present invention consists of a miscellaneous family of antineoplastic agents, including tubulin interacting agents, topoisomerase II inhibitors, topoisomerase I inhibitors and hormonal agents, selected from, but not limited to, the group consisting of ⁇ -carotene, ⁇ -difluoromethyl-arginine, acitretin, Biotec AD-5, Kyorin AHC-52, alstonine, amonafide, amphethinile, amsacrine, Angiostat, ankinomycin, anti-neoplaston AlO, antineoplaston A2, antineoplaston A3, antineoplaston A5, antineoplaston AS2-1, Henkel APD, aphidicolin glycinate, asparaginase, Avarol, baccharin, batracylin, benfluron, benzotript, Ipsen-Beaufour BIM-
  • the present compounds may also be used in co-therapies with other anti-neoplastic agents, such as acemannan, aclarubicin, aldesleukin, alemtuzumab, alitretinoin, altretamine, amifostine, aminolevulinic acid, amrubicin, amsacrine, anagrelide, anastrozole, ANCER, ancestim, ARGLABIN, arsenic trioxide, BAM 002 (Novelos), bexarote ⁇ e, bicalutamide, broxuridine, capecitabine, celmoleukin, cetrorelix, cladribine, clotrimazole, cytarabine ocfosfate, DA 3030 (Dong-A), daclizumab, denileukin diftitox, deslorelin, dexrazoxane, dilazep, docetaxel, docos
  • the compounds of the invention may further be used with VEGFR inhibitors.
  • Other compounds described in the following patents and patent applications can be used in combination therapy: US 6,258,812, US 2003/0105091, WO 01/37820, US 6,235,764, WO 01/32651, US 6,630,500, US 6,515,004, US 6,713,485, US 5,521,184, US 5,770,599, US 5,747,498, WO 02/68406, WO 02/66470, WO 02/55501, WO 04/05279, WO 04/07481, WO 04/07458, WO 04/09784, WO 02/591 10, WO 99/45009, WO 00/59509, WO 99/61422, US 5,990,141, WO 00/12089, and WO 00/02871.
  • the combination comprises a composition of the present invention in combination with at least one anti-angiogenic agent.
  • Agents are inclusive of, but not limited to, in vitro synthetically prepared chemical compositions, antibodies, antigen binding regions, radionuclides, and combinations and conjugates thereof.
  • An agent can be an agonist, antagonist, allosteric modulator, toxin or, more generally, may act to inhibit or stimulate its target (e.g., receptor or enzyme activation or inhibition), and thereby promote cell death or arrest cell growth.
  • anti-tumor agents include HERCEPTINTM (trastuzumab), which may be used to treat breast cancer and other forms of cancer, and RITUXANTM (rituximab), ZEVALINTM (ibritumomab tiuxetan), and LYMPHOC1DETM (epratuzumab), which may be used to treat non-Hodgkin's lymphoma and other forms of cancer, GLEEV ACTM which may be used to treat chronic myeloid leukemia and gastrointestinal stromal tumors, and BEXXARTM (iodine 131 tositumomab) which may be used for treatment of non-Hodgkins's lymphoma.
  • Exemplary anti-angiogenic agents include ERBITUXTM (IMC-C225),
  • KDR (kinase domain receptor) inhibitory agents e.g., antibodies and antigen binding regions that specifically bind to the kinase domain receptor
  • anti-VEGF agents e.g., antibodies or antigen binding regions that specifically bind VEGF, or soluble VEGF receptors or a Iigand binding region thereof
  • AVASTINTM or VEGF-TRAPTM e.g., antibodies or antigen binding regions that specifically bind thereto
  • EGFR inhibitory agents e.g., antibodies or antigen binding regions that specifically bind thereto
  • anti-Angl and anti-Ang2 agents e.g., antibodies or antigen binding regions specifically binding thereto or to their receptors, e.g., Tie2/Tek
  • anti-Tie2 kinase inhibitory agents e.g., antibodies or antigen binding regions that specifically bind to the kinase domain receptor
  • anti-VEGF agents e.g., antibodies or antigen binding regions that specifically bind VEGF, or
  • compositions of the present invention can also include one or more agents (e.g., antibodies, antigen binding regions, or soluble receptors) that specifically bind and inhibit the activity of growth factors, such as antagonists of hepatocyte growth factor (HGF, also known as Scatter Factor), and antibodies or antigen binding regions that specifically bind its receptor "c-met".
  • agents e.g., antibodies, antigen binding regions, or soluble receptors
  • HGF hepatocyte growth factor
  • c-met hepatocyte growth factor
  • Other anti-angiogenic agents include Campath, IL-8, B-FGF, Tek antagonists (Ceretti et al., U.S. Publication No. 2003/0162712; U.S. Patent No.
  • anti-TWEAK agents e.g., specifically binding antibodies or antigen binding regions, or soluble TWEAK receptor antagonists; see, Wiley, U.S. Patent No. 6,727,225
  • ADAM distintegrin domain to antagonize the binding of integrin to its ligands Fanslow et al., U.S. Publication No. 2002/0042368
  • specifically binding anti-eph receptor and/or anti-ephrin antibodies or antigen binding regions U.S. Patent Nos.
  • anti-PDGF-BB antagonists e.g., specifically binding antibodies or antigen binding regions
  • antibodies or antigen binding regions specifically binding to PDGF- BB ligands
  • PDGFR kinase inhibitory agents e.g., antibodies or antigen binding regions that specifically bind thereto
  • Additional anti-angiogenic/anti-tumor agents include: SD-7784 (Pfizer.,
  • the present compounds may also be used in co-therapies with other anti-neoplastic agents, such as VEGF antagonists, other kinase inhibitors including p38 inhibitors, KDR inhibitors, EGF inhibitors and CDK inhibitors, TNF inhibitors, matrix metal loproteinases (MMP) inhibitors, COX-2 inhibitors including celecoxib, NSAID's, or ⁇ v ⁇ 3 inhibitors.
  • VEGF antagonists such as VEGF antagonists, other kinase inhibitors including p38 inhibitors, KDR inhibitors, EGF inhibitors and CDK inhibitors, TNF inhibitors, matrix metal loproteinases (MMP) inhibitors, COX-2 inhibitors including celecoxib, NSAID's, or ⁇ v ⁇ 3 inhibitors.
  • MMP matrix metal loproteinases
  • Preparative HPLC was conducted on a Varian, Shimadzu, Beckman, or Waters HPLC system with 0.1 % TFA/H 2 O and 0.1 % TFA/CH 3 CN as mobile phase. The flow rate was at 20 mL/minute and the gradient method was used. 1 H NMR spectra were obtained with super conducting FT NMR spectrometers operating at 400 MHz or a Varian 300 MHz instrument. Chemical shifts are expressed in ppm downfield from the tetramethylsilane internal standard. AH compounds showed NMR spectra consistent with their assigned structures.
  • Mass spectra were obtained using a Perkin Elmer-SCIEX API 165 electrospray mass spectrometer (positive and/or negative) or an MP 1 100 MSD LC-MS with electrospray ionization and quadrupole detection. All parts are by weight and temperatures are in degrees centigrade unless otherwise indicated. [00194] The following abbreviations are used: AcOH (acetic acid), ATP
  • Example 1 was synthesized starting with commercially available 5-bromothiazol-2-amine and (S)-2-amino-3-phenylpropan-l-ol. HRMS Theoretical (M+H) 364.15904, found 364.15915. [00197] N-(5-bromothiazol-2-yl)acetamide:
  • N-(5-Bromo-thiazol-2-yl)-acetamide) 442.0 mg, 2.0 mmol
  • (S)-2-(l- hydroxy-3-phenylpropan-2-yl)isoindoline-l,3-dione 281.0 mg, 1.0 mmol
  • triphenylphosphine 303.0 mg, 1.5 mmol
  • THF 15.0 mL
  • tetrakis(triphenylphosphi ⁇ e)palladium(0) (10.0 mg,0.0083 mmol) was added to the tube, and the mixture was heated at 150°C in a Smith microwave synthesizer. After heating for 25 minutes, the reaction mixture was filtered through a pad of Celite and washed with MeOH. The filtrate was evaporated off, and the resulting residue was diluted with 1.5 mL MeOH, 1.5 mL water, and 1.5 mL hydrazine. The resulting mixture was transferred to a 5 mL microwave tube and heated at 150 0 C. After heating for 33 minutes, silica gel (10.0 g) was added to the reaction mixture, and the mixture was concentrated to dryness.
  • Examples 2-3 were synthesized in a manner similar to that shown in Scheme 1.
  • Example 2 N-((S)-2-amino-3-(lH-indol-3-yI)propyl)-5-(3-methyl-lH- indazoI-5-yI)thiazoI-2-amine: HRMS Theoretical (M+H) 403.16994, found 403.16939.
  • Example 3 N-((S)-2-amino-3-(4-chlorophenyl)propyl)-5-(3-methyl- lH-indazol-5-yI)thiazol-2-amine: HRMS Theoretical (M+H) 398.12007, found 398.11981.
  • This compound was prepared in a manner similar to that shown in
  • Examples 5-6 Examples 5-6 were synthesized in a manner similar to that shown in Scheme 1 using isoquinolin-6-ylboronic acid to couple with the corresponding amino thiazole bromide, ⁇ soquinoiin-6-ylboronic acid was prepared as shown in Scheme 2.
  • Examples 7-10 were synthesized using a procedure similar to that shown in Scheme 1 with a coupling reaction between the corresponding bromothiazole intermediate and the boronic acid or esters.
  • the 4-substituted 2- aminothiazoles were prepared by treating the commercially available 2-bromo-ketones with thiourea as shown in a similar manner in Scheme 3.
  • Examples 10-12 were synthesized using a procedure similar to that shown in Scheme 1 with a coupling reaction between the corresponding bromothiazole intermediate and the boronic acid or esters.
  • the 4-hydroxy methyl -2- aminothiazole intermediate was prepared as shown in Scheme 3.
  • Examples 13-15 were synthesized using a procedure similar to that shown in Scheme 1 using a coupling reaction between the corresponding bromothiazole intermediate and the boronic acid or esters.
  • the 4-methoxylmethyl-2- aminothiazole intermediate was prepared as shown in Scheme 4.
  • Butan-2-one (2O.g, 277 mmol) and 200 mL MeOH were charged into a 1
  • Example 17 was synthesized as shown in Scheme 6.
  • N-(4-(methoxymethyl)-5-(thiazolo[5,4-c]pyridin-2-yl)thiazol-2- yljacetamide [00258] Crude 4-(2-acetamido-4-(methoxymethyl)thiazole-5- carboxamido)pyridin-3-yI diethylcarbamodithioate (4.485 g, 9.89 mmol) and formic acid (0.379 mL, 9.89 mmol) were added to a 150 rtiL round-bottomed flask, and the solution was stirred at reflux for approximately 24 hours until LCMS indicated that the reaction was complete.
  • Examples 28-30 were made using a procedure similar to that shown in Scheme 6, but starting from readily available 2-(benzylamino)thiazole-5- carboxylic acid as shown in Scheme 7.
  • Scheme 7
  • Example 30 N-((S)-2-amino-3-(4-(trinuoromethyl)phenyl)propyI)-N- benzy]-5-(thiazolo[5,4-c]pyridin-2-yl)thiazol-2-amine: LCMS (M+H) 526.1 calc. for C 26 H 23 F 3 N 5 S 2 526.13.
  • Examples 31-32 were made using a procedure similar to that shown in Scheme 6, but starting from readily available methyl 2-aminothiazole-5- carboxylate to make the intermediate N-(5-(thiazolo[5,4-c]pyridin-2-yl)thiazol-2- yl)acetamide.
  • Examples 33-34 were made using the procedure shown in Scheme 8 starting from N-(4-(methoxymethyl)-5-(thiazolo[5,4-c]pyridin-2- yl)thiazol-2-yl)acetamide that was prepared as shown in Scheme 6.
  • Example 33 N-(4-(methoxymethyl)-5-(thiazolo[5,4-c]pyridin-2- yl)thiazol-2-yl)-N-((S)-2-(2-morpholinoethylamino)-3-(4-
  • Examples 37-40 were synthesized using a procedure similar to that shown in Scheme 1 using a coupling reaction between the corresponding bromothiazole intermediate as described in Example 35 or Example 36 and the corresponding boronic acids or esters.
  • the title compound was obtained by treating 5-(2-((S)-2-amino-3-(lH- indoI-3-yl)propylamino)thiazol-5-yl)-3,3-difluoro-l-(methoxymethyl)indolin-2-one with 4N HCl in dioxane in a glass microwave reaction vessel and heated in a Discover® model microwave reactor (CEM, Matthews, NC) at 100 0 C for 5 minutes to remove the MOM group. The reaction mixture was concentrated and directly purified by preparative HPLC to give the title compound. MS m/z: 469 (M+ 1).
  • 3,3-difluoro-l- (methoxymethyl)-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)indolin-2-on was prepared using a procedure similar to that shown in Scheme 1 using 5-bromo-3,3- difluoro-l -(methoxymethyl)indolin-2-one as the starting material.
  • 5-Bromo-3,3-difluoro- l-(methoxymethyI)indolin-2-one was prepared using a two step procedure as described below.
  • the organic layer was dried over sodium sulfate, evaporated onto a plug of silica gel, and purified by chromatography through a Redi-Sep® pre-packed silica gel column (120 g), eluting with a gradient of 0 % to 100 % EtOAc in hexane, to provide 5-bromo-3,3-difluoro-l-(rnethoxymethyl)indolin- 2- one (1.36 g, 84% yield) as a off-white crystalline.
  • Examples 42-44 were synthesized using a procedure similar to that used in Examples 35 and 36. [00308] Example 42, 5-(2-((S)-2-amino-3-(4-
  • Example 35 using tert-butyl 5-bromo-3-(tert-butoxycarbonyl)-l H-indazole-l -carboxyIate as one of the starting material that led to the product.
  • tert-Butyl 5-bromo-3-(tert- butoxycarbonyl)-lH-indazoIe-l-carboxylate was synthesized from commercially available 5-bromo-2-fluorobenzonitrile. MS m/z: 418 (M+l).
  • Example 52 using methyl hydrazine instead of hydrazine as the reagent in the last step.
  • LCMS (M+H) 447 calc. for C 2I H 22 F 3 N 6 S 447.15.
  • Example 54 5-(2-((S)-2-Amino-3-(4- chlorophenyl)propyIamino)thiazol-5 ⁇ yI)indolin-2-one:
  • Example 1 and Example 36 The coupling reaction used CtBu 2 PPh) 2 PdCl 2 as the catalyst with KOAc as the base instead of Pd(PPh 3 ) 4 with Na 2 COs as the base as shown in Scheme 1.
  • the reaction started with the commercially available 5-bromoindolin-2-one. LCMS (M+H) 399 calc.
  • Example 544 It started with the commercially available 6-chlorobenzo[d]oxazol-2(3H)- one.
  • Example 56 N-((S)-2-amino-3-(4-(trifluoromethyl)phenyl)propyl)-5-
  • 6-bromo-l -methyl-1 H-benzo[d]imidazol-2(3H)-one was prepared by treating tert-butyl 5-bromo-2-oxo-2,3-dihydrobenzo[d]imidazole-l- carboxylate with dimethyl sulfate in the presence of sodium carbonate followed by a crystallization step in the wet MeOH.
  • tert-Butyl 5-bromo-2-oxo-2,3- dihydrobenzo[d]imidazole-l-carboxylate was prepared as described by Puwen Zhang, et. ah, in Bioorganic & Medicinal Chemistry Letters 1 1 (2001) 2747-2750.
  • Example 57 using the same starting 5- bromothiazole intermediate used in Example 14.
  • Example 60 N-((S)-2-amino-3-(4-(tri ⁇ uoromethyl)phenyl)propyl)-4-
  • Examples 62-63 were synthesized using a procedure similar to that used for Example 61 .
  • Example 64 ⁇ -fi(-S)-2-amino-3-(4-(trifluoroinethyI)phenyl)propyI)-5-
  • Example 64 using tert-butyl 5-(l-hydroxyphthalazin-6-yl)thiazoI-2-y [carbamate as the starting material.
  • LCMS (M+H + ) 446.2 calc. for C 2 ]Hi 8 F 3 N 5 SO, 446.2; 1 H NMR (400 MHz, CDCl 3 ) ⁇ ppm 8.29 (s, 1H), 8.27 (d, 1H), 7.95 (d, 1H), 7.85 (s, 1H), 7.68 (s, 1H) 7.62 (d, 2H) 7.47 (d, 2H), 3.92 (dd, 2H), 3.55 (dd, 1H), 3.25 (tt, 1H) 2.98 (dd, 1 H), 2.89 (t, 2H), 2.72 (b, 1H).
  • Example 64 by coupling 2-iodo-l,6-naphthyridine with tert-butyl 5- (tributylstannyl)thiazol-2-ylcarbamate.
  • the resulting tert-butyl 5-(l,6-naphthyridin-2-yl) thiazol-2-ylcarbamate was treated with the cyclic sulfamidate as described in Scheme 10 for Example 36.
  • the intermediate 2-iodo-l,6-naphthyridine was prepared as shown in Scheme 17.
  • 6-Naphthyridin-2-amine (1.6 g, 5.5 mmol), I 2 (0.70 g, 2.8 mmol), copper(I) iodide (0.32 mg, 1.7 mmol), cesium iodide (1.43 g, 5.5 mmol), isoamy] nitrite (3.9 g, 33.1 mmol), and 60 mL of DME were added to a 250 mL round-bottom flask.
  • the reaction mixture was heated to 60 0 C for 16 hours and then 70 mL of EtOAc was added to the reaction mixture.
  • the mixture was washed with 70 mL of 20 % NH 3 solution and 70 mL of 1 M Na 2 S 2 O 3 solution.
  • Example 70 5-(2-((S)-2-amino-3-phenylpropylamino)thiazol-5-yl)-3- methyIindoIin-2-one:
  • ACN 400 mL was stirred at room temperature and treated with pyridine (13 mL, 158 mmol) and then di-tert-butyl dicarbonate (23 g, 105 mmol). The reaction mixture was stirred at room temperature overnight. The solvent was reduced in vacuo to approximately 20 mL, and the mixture was partitioned between EtOAc (200 mL) and 1 N HCl (150 mL). The aqueous layer was extracted again with EtOAc (150 mL), and the combined organic phases were washed with 1 N HCl (75 mL), saturated NaHCO 3 (75 mL), and saturated NaCl (50 mL).
  • Example 72 (E)- and (Z)-5-(2-((S)-2-amino-3-(lH-indol-3- yl)propylamino)thiazol-5-yl)-3-(furan-2-yImethylene)indolin-2-one ditrifluoroacetates:
  • Example 76 6-(2-((S)-2-Amino-3-(4- chlorophenyl)propylamino)thiazol-5-yl)isoq ⁇ iinolin-3 ⁇ amine:
  • Example 21 LCMS (M+H) 460.1 calcd for C 21 H 23 ClN 5 OS 2 460.09. [00427]
  • Example 78 6-(2-((S)-2-amino-3-(4- ⁇ iethoxyphe ⁇ yl)propylamiiio)thiazol-5-yJ)beiizo[d)oxazol-2(3H)-one:
  • Example 36 LCMS m/z: 397 (M+l).
  • Example 79 4-(2-((S)-2-Amino-3-(4-
PCT/US2007/000871 2006-01-18 2007-01-11 Thiazole compounds as protein kinase b ( pkb) inhibitors WO2007084391A2 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
CA2636077A CA2636077C (en) 2006-01-18 2007-01-11 Thiazole compounds as protein kinase b (pkb) inhibitors
AU2007207743A AU2007207743B2 (en) 2006-01-18 2007-01-11 Thiazole compounds as protein kinase B (PKB) inhibitors
EA200801716A EA200801716A1 (ru) 2006-01-18 2007-01-11 Тиазольные соединения и их применение
ES07718125T ES2389062T3 (es) 2006-01-18 2007-01-11 Compuestos de tiazol como inhibidores de proteína cinasa B (PKB)
JP2008551297A JP5199885B2 (ja) 2006-01-18 2007-01-11 プロテインキナーゼb(pkb)阻害剤としてのチアゾール化合物
BRPI0706621-0A BRPI0706621A2 (pt) 2006-01-18 2007-01-11 composto, composição farmacêutica, métodos para tratar um distúrbio mediado por quinase em um mamìfero e para tratar um distúrbio relacionado com a proliferação em um mamìfero, e , uso do composto
EP07718125A EP1981884B1 (en) 2006-01-18 2007-01-11 Thiazole compounds as protein kinase b (pkb) inhibitors
IL192751A IL192751A0 (en) 2006-01-18 2008-07-10 Thiazole compounds as protein kinase b (pkb) inhibitors
NO20083572A NO20083572L (no) 2006-01-18 2008-08-18 Tiazolforbindelser som proteinkinase-B (PKB)-inhibitorer

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US75954606P 2006-01-18 2006-01-18
US60/759,546 2006-01-18

Publications (2)

Publication Number Publication Date
WO2007084391A2 true WO2007084391A2 (en) 2007-07-26
WO2007084391A3 WO2007084391A3 (en) 2008-03-20

Family

ID=38180602

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/000871 WO2007084391A2 (en) 2006-01-18 2007-01-11 Thiazole compounds as protein kinase b ( pkb) inhibitors

Country Status (21)

Country Link
US (2) US7514566B2 (US08084479-20111227-C00086.png)
EP (1) EP1981884B1 (US08084479-20111227-C00086.png)
JP (1) JP5199885B2 (US08084479-20111227-C00086.png)
KR (1) KR20080091369A (US08084479-20111227-C00086.png)
CN (1) CN101421265A (US08084479-20111227-C00086.png)
AR (1) AR059064A1 (US08084479-20111227-C00086.png)
AU (1) AU2007207743B2 (US08084479-20111227-C00086.png)
BR (1) BRPI0706621A2 (US08084479-20111227-C00086.png)
CA (1) CA2636077C (US08084479-20111227-C00086.png)
CR (1) CR10211A (US08084479-20111227-C00086.png)
EA (1) EA200801716A1 (US08084479-20111227-C00086.png)
ES (1) ES2389062T3 (US08084479-20111227-C00086.png)
IL (1) IL192751A0 (US08084479-20111227-C00086.png)
MY (1) MY149143A (US08084479-20111227-C00086.png)
NO (1) NO20083572L (US08084479-20111227-C00086.png)
PE (1) PE20071114A1 (US08084479-20111227-C00086.png)
TW (1) TW200738657A (US08084479-20111227-C00086.png)
UA (1) UA91895C2 (US08084479-20111227-C00086.png)
UY (1) UY30098A1 (US08084479-20111227-C00086.png)
WO (1) WO2007084391A2 (US08084479-20111227-C00086.png)
ZA (1) ZA200806386B (US08084479-20111227-C00086.png)

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008100456A2 (en) * 2007-02-13 2008-08-21 Schering Corporation Functionally selective alpha2c adrenoreceptor agonists
WO2009011880A2 (en) * 2007-07-17 2009-01-22 Amgen Inc. Heterocyclic modulators of pkb
WO2009137391A3 (en) * 2008-05-06 2009-12-30 Smithkline Beecham Corporation Benzene sulfonamide thiazole and oxazole compounds
US7700636B2 (en) 2004-10-18 2010-04-20 Amgen Inc. Thiadiazole compounds and methods of use
WO2010083246A1 (en) * 2009-01-15 2010-07-22 Amgen Inc. Fluoroisoquinoline substituted thiazole compounds and methods of use
US7919504B2 (en) 2007-07-17 2011-04-05 Amgen Inc. Thiadiazole modulators of PKB
WO2011044415A1 (en) * 2009-10-08 2011-04-14 Glaxosmithkline Llc Combination
WO2011044414A1 (en) * 2009-10-08 2011-04-14 Glaxosmithkline Llc Combination
WO2011046894A1 (en) * 2009-10-12 2011-04-21 Glaxosmithkline Llc Combination
EP2326631A2 (en) * 2008-08-18 2011-06-01 Yale University Mif modulators
WO2011115071A1 (ja) * 2010-03-15 2011-09-22 国立大学法人広島大学 チエノピリジン誘導体及びその製造方法、並びにそれを用いた有機半導体デバイス
WO2011130921A1 (en) * 2010-04-23 2011-10-27 Merck Sharp & Dohme Corp. Inhibitors of akt activity
US8084479B2 (en) 2006-01-18 2011-12-27 Amgen Inc. Thiazole compounds and methods of use
JP2013525303A (ja) * 2010-04-16 2013-06-20 アッヴィ・インコーポレイテッド フタラジン−(2h)−オン系キナーゼ阻害薬
WO2013173382A1 (en) 2012-05-15 2013-11-21 Amgen Inc. Benzothiophene sulfonamides and other compounds that interact with glucokinase regulatory protein
EP2832731A4 (en) * 2012-03-27 2015-08-19 Shionogi & Co AROMATIC HETEROCYCLIC CHAIN 5-CORE DERIVED DERIVATIVE HAVING TRPV4 INHIBITORY ACTIVITY
CN106243100A (zh) * 2008-10-10 2016-12-21 Vm生物医药公司 治疗酒精使用障碍,疼痛和其他疾病的药物组合与方法
US9540322B2 (en) 2008-08-18 2017-01-10 Yale University MIF modulators
US9643922B2 (en) 2008-08-18 2017-05-09 Yale University MIF modulators
EP2445502B1 (en) * 2009-06-25 2017-06-21 Alkermes Pharma Ireland Limited Heterocyclic compounds for the treatment of neurological and psychological disorders
JP2018510857A (ja) * 2015-03-02 2018-04-19 ライジェル ファーマシューティカルズ, インコーポレイテッド TGF−β阻害剤
US10342786B2 (en) 2017-10-05 2019-07-09 Fulcrum Therapeutics, Inc. P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD
CN111603466A (zh) * 2020-06-29 2020-09-01 江南大学 一种乙酮类化合物在制备治疗肿瘤药物中的应用
CN114014787A (zh) * 2022-01-10 2022-02-08 苏州开元民生科技股份有限公司 一种制备(2s,3r)-对甲砜基苯丝氨酸乙酯的不对称合成方法
US11273158B2 (en) 2018-03-05 2022-03-15 Alkermes Pharma Ireland Limited Aripiprazole dosing strategy
US11291659B2 (en) 2017-10-05 2022-04-05 Fulcrum Therapeutics, Inc. P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD
US11566003B2 (en) 2017-03-30 2023-01-31 Genentech, Inc. Isoquinolines as inhibitors of HPK1
US11612606B2 (en) 2018-10-03 2023-03-28 Genentech, Inc. 8-aminoisoquinoline compounds and uses thereof

Families Citing this family (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2011525929A (ja) * 2008-06-26 2011-09-29 グラクソスミスクライン・リミテッド・ライアビリティ・カンパニー Akt活性の阻害剤
WO2010080503A1 (en) * 2008-12-19 2010-07-15 Genentech, Inc. Heterocyclic compounds and methods of use
EP2440556A1 (en) * 2009-06-10 2012-04-18 Vertex Pharmaceuticals Incorporated Inhibitors of phosphatidylinositol 3-kinase
WO2011049625A1 (en) 2009-10-20 2011-04-28 Mansour Samadpour Method for aflatoxin screening of products
SG10201500895XA (en) 2009-11-05 2015-04-29 Rhizen Pharmaceuticals Sa Chromen-4-one Derivatives As Kinase Modulators
JP5578705B2 (ja) * 2010-03-29 2014-08-27 公益財団法人相模中央化学研究所 (アリール)ジフルオロ酢酸エステル誘導体及びその製造方法
US20130102605A1 (en) * 2010-04-23 2013-04-25 Peng Liang Inhibitors of akt activity
JO2998B1 (ar) 2010-06-04 2016-09-05 Amgen Inc مشتقات بيبيريدينون كمثبطات mdm2 لعلاج السرطان
CA2833935C (en) 2011-05-04 2020-09-15 Dhanapalan Nagarathnam Novel compounds as modulators of protein kinases
MX352672B (es) 2011-09-27 2017-12-04 Amgen Inc Compuestos heterocíclicos como inhibidores de mdm2 para el tratamiento del cáncer.
CN103304468A (zh) * 2012-03-13 2013-09-18 华东师范大学 一种氧化吲哚的一锅法串联合成方法
PL2870157T3 (pl) 2012-07-04 2018-01-31 Rhizen Pharmaceuticals S A Selektywne inhibitory pi3k delta
US20160002185A1 (en) 2013-02-19 2016-01-07 Amgen Inc. Cis-morpholinone and other compounds as mdm2 inhibitors for the treatment of cancer
RU2015143526A (ru) * 2013-03-13 2017-04-19 Бостон Байомедикал, Инк. Производные 3-(арил или гетероарил)метилениндолин-2-она в качестве ингибиторов киназного пути раковых стволовых клеток для лечения рака
CA2906538C (en) 2013-03-14 2021-02-02 Ana Gonzalez Buenrostro Heteroaryl acid morpholinone compounds as mdm2 inhibitors for the treatment of cancer
JOP20200296A1 (ar) 2013-06-10 2017-06-16 Amgen Inc عمليات صنع وأشكال بلورية من mdm2 مثبط
MX2020013670A (es) 2013-11-11 2022-09-30 Amgen Inc Terapia de combinacion que incluye un inhibidor mdm2 y uno o mas agentes farmaceuticamente activos adicionales para el tratamiento de canceres.
WO2017151409A1 (en) 2016-02-29 2017-09-08 University Of Florida Research Foundation, Incorporated Chemotherapeutic methods
MX2018010894A (es) * 2016-03-10 2018-11-09 Astrazeneca Ab Nuevos inhibidores de fosfatidilinositol 3-quinasa gamma.
JOP20190272A1 (ar) 2017-05-22 2019-11-21 Amgen Inc مثبطات kras g12c وطرق لاستخدامها
AU2019262589B2 (en) 2018-05-04 2022-07-07 Amgen Inc. KRAS G12C inhibitors and methods of using the same
JP7377679B2 (ja) 2018-11-19 2023-11-10 アムジエン・インコーポレーテツド がん治療のためのkrasg12c阻害剤及び1種以上の薬学的に活性な追加の薬剤を含む併用療法
EP3738593A1 (en) 2019-05-14 2020-11-18 Amgen, Inc Dosing of kras inhibitor for treatment of cancers
CR20210665A (es) 2019-05-21 2022-01-25 Amgen Inc Formas en estado sólido
CN114391012A (zh) * 2019-08-02 2022-04-22 美国安进公司 作为kif18a抑制剂的吡啶衍生物
WO2021076655A1 (en) 2019-10-15 2021-04-22 Amgen Inc. Combination therapy of kras inhibitor and shp2 inhibitor for treatment of cancers
WO2021126816A1 (en) 2019-12-16 2021-06-24 Amgen Inc. Dosing regimen of a kras g12c inhibitor
CN116813608B (zh) * 2023-06-08 2024-03-22 英矽智能科技(上海)有限公司 噻唑类化合物及其应用

Family Cites Families (81)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3591600A (en) * 1969-07-07 1971-07-06 Stauffer Chemical Co 2-aminothiazole phosphates and phosphonates
GB1435139A (en) * 1972-08-17 1976-05-12 Sumitomo Chemical Co Thiazole derivatives
JPS5239827B2 (US08084479-20111227-C00086.png) 1973-11-08 1977-10-07
US4086239A (en) * 1977-07-01 1978-04-25 Stauffer Chemical Company Thiazole bis-phosphates and phosphonates, intermediates, and insecticidal compositions and methods
DK150068C (da) 1978-06-02 1987-06-29 Pfizer Analogifremgangsmaade til fremstilling af aminothiazoler
US4297365A (en) * 1978-08-04 1981-10-27 Ciba-Geigy Corporation Benzimidazoles and pharmaceutical preparations containing such compounds
US4451471A (en) * 1981-03-18 1984-05-29 Ciba-Geigy Corporation Certain 2,4,5-tri-substituted thiazoles, pharmaceutical compositions containing same and methods of using same
FR2511375A1 (fr) * 1981-08-17 1983-02-18 Rhone Poulenc Sante Nouveaux derives de la cephalosporine, leur preparation et les medicaments qui les contiennent
DE3630732A1 (de) * 1986-09-10 1988-03-17 Bayer Ag 2-cyano-2-alkoximino-acetamide
US5232921A (en) * 1987-03-12 1993-08-03 Sanofi Thiazole derivatives active on the cholinergic system, process for obtention and pharmaceutical compositions
JPH0753666B2 (ja) 1987-09-14 1995-06-07 久光製薬株式会社 置換ジフェニルチアゾール誘導体からなる抗炎症剤
FR2636628B1 (fr) * 1988-08-25 1990-12-28 Sanofi Sa Derives du thiadiazole-1,3,4, leur procede d'obtention et compositions pharmaceutiques en contenant
US5302608A (en) * 1988-11-18 1994-04-12 Takeda Chemical Industries, Ltd. Age formation inhibitors
US5145860A (en) * 1989-01-05 1992-09-08 Fujisawa Pharmaceutical Co., Ltd. Thiazole compounds and pharmaceutical composition comprising the same
US5550138A (en) * 1992-03-25 1996-08-27 Takeda Chemical Industries, Ltd. Condensed thiadiazole derivative, method of its production, and use thereof
WO1996016650A1 (fr) * 1994-11-29 1996-06-06 Hisamitsu Pharmaceutical Co., Inc. Antibacterien et bactericide comprenant un derive de 2-aminothiazole et sels de ce derive
FR2735777B1 (fr) * 1995-06-21 1997-09-12 Sanofi Sa Derives de 4-phenylaminothiazole, leur procede de preparation et les compositions pharmaceutiques les contenant
US6407124B1 (en) * 1998-06-18 2002-06-18 Bristol-Myers Squibb Company Carbon substituted aminothiazole inhibitors of cyclin dependent kinases
CA2332325A1 (en) 1998-06-18 1999-12-23 Bristol-Myers Squibb Company Carbon substituted aminothiazole inhibitors of cyclin dependent kinases
GB9823873D0 (en) * 1998-10-30 1998-12-30 Pharmacia & Upjohn Spa 2-ureido-thiazole derivatives,process for their preparation,and their use as antitumour agents
EP1150565A1 (en) 1999-02-08 2001-11-07 LION bioscience AG Thiazole derivatives and combinatorial libraries thereof
AUPP873799A0 (en) * 1999-02-17 1999-03-11 Fujisawa Pharmaceutical Co., Ltd. Pyridine compounds
UA71971C2 (en) * 1999-06-04 2005-01-17 Agoron Pharmaceuticals Inc Diaminothiazoles, composition based thereon, a method for modulation of protein kinases activity, a method for the treatment of diseases mediated by protein kinases
JP2001114690A (ja) * 1999-08-06 2001-04-24 Takeda Chem Ind Ltd p38MAPキナーゼ阻害剤
CA2381215A1 (en) * 1999-08-06 2001-02-15 Takeda Chemical Industries, Ltd. P38map kinase inhibitors
WO2001074811A2 (en) * 2000-03-30 2001-10-11 Takeda Chemical Industries, Ltd. Substituted 1,3-thiazole compounds, their production and use
US6586453B2 (en) * 2000-04-03 2003-07-01 3-Dimensional Pharmaceuticals, Inc. Substituted thiazoles and the use thereof as inhibitors of plasminogen activator inhibitor-1
US7074934B2 (en) * 2000-06-13 2006-07-11 Tularik Limited Serine protease inhibitors
GT200100147A (es) 2000-07-31 2002-06-25 Derivados de imidazol
JP4689801B2 (ja) 2000-08-09 2011-05-25 ケミプロ化成株式会社 アミノチアゾール誘導体の製造方法
JP2002053566A (ja) 2000-08-11 2002-02-19 Japan Tobacco Inc チアゾール化合物及びその医薬用途
GB0114185D0 (en) * 2001-06-12 2001-08-01 Protherics Molecular Design Lt Compounds
US20040097555A1 (en) * 2000-12-26 2004-05-20 Shinegori Ohkawa Concomitant drugs
AR032653A1 (es) * 2001-02-09 2003-11-19 Telik Inc Inhibidores heterociclicos del trasportador de glicina 2 composiciones farmaceuticas, uso y metodos.
DE60208630T2 (de) * 2001-05-11 2006-08-17 Pfizer Products Inc., Groton Thiazolderivate und ihre Verwendung als cdk-Inhibitoren
EP1402900A1 (en) * 2001-06-11 2004-03-31 Takeda Chemical Industries, Ltd. Medicinal compositions
AR039059A1 (es) * 2001-08-06 2005-02-09 Sanofi Aventis Compuesto derivado de acilaminotiazol, su utilizacion, procedimientos para prepararlo, composicion farmaceutica que lo comprende, y compuestos intermediarios
MXPA04002914A (es) * 2001-09-28 2005-06-20 Cyclacel Ltd N- (4- (4-metiltiazol-5-il)pirimidin-2-il) -n-fenilaminas como compuestos antiproliferativos.
GB0123589D0 (en) * 2001-10-01 2001-11-21 Syngenta Participations Ag Organic compounds
US20050004134A1 (en) * 2001-11-08 2005-01-06 Hideo Tsutsumi Thiazole derivative and pharmaceutical use thereof
CN1633293A (zh) 2002-02-13 2005-06-29 药物基因实验室有限公司 5-ht2b受体拮抗剂
TWI314928B (en) * 2002-02-28 2009-09-21 Novartis A 5-phenylthiazole derivatives and use as pi3 kinase inhibitors
DE60316411T2 (de) * 2002-02-28 2008-06-12 F. Hoffmann-La Roche Ag Thiazolderivate als npy-rezeptorantagonisten
WO2004026226A2 (en) * 2002-05-10 2004-04-01 Cytokinetics, Inc. Compounds, compositions and methods
FR2842523A1 (fr) * 2002-07-17 2004-01-23 Sanofi Synthelabo Derives d'acylaminothiazole, leur preparation et leur application en therapeutique
WO2004033439A1 (en) * 2002-10-09 2004-04-22 Pfizer Products Inc. Thiazole compounds for the treatment of neurodegenerative disorders
US20040122016A1 (en) 2002-10-30 2004-06-24 Jingrong Cao Compositions useful as inhibitors of rock and other protein kinases
GB0226583D0 (en) * 2002-11-14 2002-12-18 Cyclacel Ltd Compounds
TWI335913B (en) 2002-11-15 2011-01-11 Vertex Pharma Diaminotriazoles useful as inhibitors of protein kinases
US20050004186A1 (en) 2002-12-20 2005-01-06 Pfizer Inc MEK inhibiting compounds
FR2850380B1 (fr) * 2003-01-23 2006-07-07 Sanofi Synthelabo Derives d'acylaminothiazole, leur preparation et leur utilisation en therapeutique
JP2006182648A (ja) 2003-04-08 2006-07-13 Dai Ichi Seiyaku Co Ltd 7員複素環誘導体
AR044519A1 (es) * 2003-05-02 2005-09-14 Novartis Ag Derivados de piridin-tiazol amina y de pirimidin-tiazol amina
JP2007512230A (ja) * 2003-08-20 2007-05-17 バーテックス ファーマシューティカルズ インコーポレイテッド プロテインキナーゼ阻害剤として有用な(4−アミノ−1,2,5−オキサジアゾール−4−イル)−ヘテロ芳香族化合物
ZA200602755B (en) * 2003-09-06 2007-06-27 Vertex Pharma Modulators of ATP-binding cassette transporters
GB0327380D0 (en) 2003-11-25 2003-12-31 Cyclacel Ltd Method
GB0329275D0 (en) 2003-12-18 2004-01-21 Merck Sharp & Dohme Therapeutic treatment
AU2005205201B2 (en) 2004-01-12 2011-05-12 Merck Serono Sa Thiazole derivatives and use thereof
BRPI0506880A (pt) * 2004-01-16 2007-06-26 Sanofi Aventis derivados de acilaminotiazol, sua preparação e sua aplicação em terapêutica
EP1709018B1 (fr) * 2004-01-16 2011-07-20 Sanofi-Aventis Derives d'acylaminothiazole et leur application comme inhibiteurs de beta-amyloide
US20070185152A1 (en) * 2004-03-02 2007-08-09 Smithkline Beecham Corporation Inhibitors of akt activity
AU2005225635B2 (en) 2004-03-23 2010-06-24 Pfizer Products Inc. Imidazole compounds for the treatment of neurodegenerative disorders
CN1989131A (zh) * 2004-03-30 2007-06-27 希龙公司 取代的噻吩衍生物用作抗癌药
CA2563374A1 (en) * 2004-04-27 2005-12-08 Research Development Foundation Antagonism of tgf-.beta.superfamily receptor signaling
WO2005113579A1 (en) * 2004-05-21 2005-12-01 Mpex Pharmaceuticals, Inc. Bacterial efflux pump inhibitors and methods of treating bacterial infections
GB0411791D0 (en) 2004-05-26 2004-06-30 Cyclacel Ltd Compounds
RU2007109073A (ru) 2004-08-13 2008-09-20 Дженентек, Инк. (Us) Ингибиторы для атф-зависимого фермента на основе тиазола
WO2006038734A1 (en) 2004-10-08 2006-04-13 Astellas Pharma Inc. Pyridazinone derivatives cytokines inhibitors
CA2583217C (en) * 2004-10-18 2011-05-31 Amgen Inc. 1,3,4-thiadiazole compounds as protein kinase inhibitors
SE0402735D0 (sv) 2004-11-09 2004-11-09 Astrazeneca Ab Novel compounds
US20060178388A1 (en) * 2005-02-04 2006-08-10 Wrobleski Stephen T Phenyl-substituted pyrimidine compounds useful as kinase inhibitors
AU2006219231B2 (en) * 2005-02-28 2010-01-14 Japan Tobacco Inc. Novel aminopyridine compound with Syk inhibitory activity
RU2447060C2 (ru) * 2005-06-17 2012-04-10 Эпоуджи Биотекнолоджи Корпорейшн Ингибиторы сфингозинкиназы
WO2007008541A2 (en) 2005-07-08 2007-01-18 Kalypsys, Inc. Cellular cholesterol absorption modifiers
DE102005048072A1 (de) 2005-09-24 2007-04-05 Bayer Cropscience Ag Thiazole als Fungizide
EP1970377A4 (en) 2005-12-09 2013-02-27 Meiji Seika Kaisha LINCOMYCIN DERIVATIVE AND ANTIBACTERIAL AGENT CONTAINING THIS AS AN ACTIVE SUBSTANCE
CA2633569A1 (en) 2005-12-12 2007-06-21 Genelabs Technologies, Inc. N-(5-membered heteroaromatic ring)-amido anti-viral compounds
NZ569814A (en) 2006-01-18 2011-10-28 Schering Corp Cannibinoid receptor modulators
MY149143A (en) 2006-01-18 2013-07-15 Amgen Inc Thiazole compounds as protien kinase b (pkb) inhibitors
EP1979338A1 (en) 2006-01-18 2008-10-15 F.Hoffmann-La Roche Ag Cis-4, 5-biaryl-2-heterocyclic-imidazolines as mdm2 inhibitors
US20080242694A1 (en) 2006-09-18 2008-10-02 D Sidocky Neil R Amino-substituted heterocycles, compositions thereof, and methods of treatment therewith

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None

Cited By (67)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7919514B2 (en) 2004-10-18 2011-04-05 Amgen Inc. Thiadiazole compounds and methods of use
US7700636B2 (en) 2004-10-18 2010-04-20 Amgen Inc. Thiadiazole compounds and methods of use
US8084479B2 (en) 2006-01-18 2011-12-27 Amgen Inc. Thiazole compounds and methods of use
WO2008100456A3 (en) * 2007-02-13 2008-11-06 Schering Corp Functionally selective alpha2c adrenoreceptor agonists
WO2008100456A2 (en) * 2007-02-13 2008-08-21 Schering Corporation Functionally selective alpha2c adrenoreceptor agonists
JP2010518159A (ja) * 2007-02-13 2010-05-27 シェーリング コーポレイション 機能選択性α2Cアドレナリン受容体アゴニスト
US8501747B2 (en) 2007-02-13 2013-08-06 Merck Sharp & Dohme Corp. Functionally selective alpha2C adrenoreceptor agonists
US7919504B2 (en) 2007-07-17 2011-04-05 Amgen Inc. Thiadiazole modulators of PKB
WO2009011880A3 (en) * 2007-07-17 2009-04-02 Amgen Inc Heterocyclic modulators of pkb
US7897619B2 (en) 2007-07-17 2011-03-01 Amgen Inc. Heterocyclic modulators of PKB
WO2009011880A2 (en) * 2007-07-17 2009-01-22 Amgen Inc. Heterocyclic modulators of pkb
WO2009137391A3 (en) * 2008-05-06 2009-12-30 Smithkline Beecham Corporation Benzene sulfonamide thiazole and oxazole compounds
KR20110013456A (ko) * 2008-05-06 2011-02-09 글락소스미스클라인 엘엘씨 벤젠 설폰아미드 티아졸 및 옥사졸 화합물
US9233956B2 (en) 2008-05-06 2016-01-12 Novartis Ag Benzene sulfonamide thiazole and oxazole compounds
KR101628746B1 (ko) 2008-05-06 2016-06-13 노바르티스 아게 벤젠 설폰아미드 티아졸 및 옥사졸 화합물
EA019349B1 (ru) * 2008-05-06 2014-03-31 ГЛАКСОСМИТКЛАЙН ЭлЭлСи Соединения бензолсульфонамидтиазола и оксазола
CN102083312A (zh) * 2008-05-06 2011-06-01 葛兰素史密丝克莱恩有限责任公司 苯磺酰胺噻唑和噁唑化合物
US7994185B2 (en) 2008-05-06 2011-08-09 Glaxo Smith Kline LLC Benzene sulfonamide thiazole and oxazole compounds
AU2009244491B2 (en) * 2008-05-06 2014-08-07 Novartis Ag Benzene sulfonamide thiazole and oxazole compounds
US8415345B2 (en) 2008-05-06 2013-04-09 Glaxo SmithKline LLC Benzene sulfonamide thiazole and oxazole compounds
US8642759B2 (en) 2008-05-06 2014-02-04 Glaxosmithkline Llc Benzene sulfonamide thiazole and oxazole compounds
EP2326631A4 (en) * 2008-08-18 2012-03-21 Univ Yale MODULATORS OF MIF
US9540322B2 (en) 2008-08-18 2017-01-10 Yale University MIF modulators
US9643922B2 (en) 2008-08-18 2017-05-09 Yale University MIF modulators
EP2326631A2 (en) * 2008-08-18 2011-06-01 Yale University Mif modulators
US10202343B2 (en) 2008-08-18 2019-02-12 Yale University MIF modulators
US11584717B2 (en) 2008-08-18 2023-02-21 Yale University MIF modulators
CN106243100A (zh) * 2008-10-10 2016-12-21 Vm生物医药公司 治疗酒精使用障碍,疼痛和其他疾病的药物组合与方法
CN106243100B (zh) * 2008-10-10 2019-04-09 Vm生物医药公司 治疗酒精使用障碍,疼痛和其他疾病的药物组合与方法
WO2010083246A1 (en) * 2009-01-15 2010-07-22 Amgen Inc. Fluoroisoquinoline substituted thiazole compounds and methods of use
US10023537B2 (en) 2009-06-25 2018-07-17 Alkermes Pharma Ireland Limited Heterocyclic compounds for the treatment of neurological and psychological disorders
US11518745B2 (en) 2009-06-25 2022-12-06 Alkermes Pharma Ireland Limited Heterocyclic compounds for the treatment of neurological and psychological disorders
US10112903B2 (en) 2009-06-25 2018-10-30 Alkermes Pharma Ireland Limited Heterocyclic compounds for the treatment of neurological and psychological disorders
EP2445502B1 (en) * 2009-06-25 2017-06-21 Alkermes Pharma Ireland Limited Heterocyclic compounds for the treatment of neurological and psychological disorders
US10351529B2 (en) 2009-06-25 2019-07-16 Alkermes Pharma Ireland Limited Heterocyclic compounds for the treatment of neurological and psychological disorders
US10822306B2 (en) 2009-06-25 2020-11-03 Alkermes Pharma Ireland Limited Heterocyclic compounds for the treatment of neurological and psychological disorders
US8835450B2 (en) 2009-10-08 2014-09-16 Glaxosmithkline Llc Combination of inhibitor of B-Raf and an inhibitor of Akt in the treatment of cancer
AU2010303364B2 (en) * 2009-10-08 2013-09-19 Novartis Ag Combination
EA022982B1 (ru) * 2009-10-08 2016-04-29 ГЛЭКСОСМИТКЛАЙН ЭлЭлСи Комбинация для лечения рака или предраковых синдромов
EA023018B1 (ru) * 2009-10-08 2016-04-29 ГЛЭКСОСМИТКЛАЙН ЭлЭлСи Комбинация для лечения рака или предраковых синдромов
AU2010303363B2 (en) * 2009-10-08 2013-09-05 Novartis Ag Combination
US9402846B2 (en) 2009-10-08 2016-08-02 Novartis Ag Combination of inhibitor of B-Raf and inhibitor of AKT in the treatment of cancer
WO2011044414A1 (en) * 2009-10-08 2011-04-14 Glaxosmithkline Llc Combination
US8796298B2 (en) 2009-10-08 2014-08-05 Glaxosmithkline Llc Combination of a B-Raf inhibitor: N-{3-[5-(2-Amino-4-pyrimidinyl)-2-(1,1- dimethylethyl)-1,3-thiazol-4-yl]-2-fluorophenyl}-2,6-difluorobenzenesulfonamide and the Akt inhibitor: N-{ (1S)-2-amino-1-[(3- fluorophenyl)methyl]ethyl}-5-chIoro-4-(4-chIoro- 1 -methyl- 1 H-pyrazol-5-yl)-2-10 thiophenecarboxamide useful in the treatment of cancer
WO2011044415A1 (en) * 2009-10-08 2011-04-14 Glaxosmithkline Llc Combination
EA020965B1 (ru) * 2009-10-12 2015-03-31 ГЛЭКСОСМИТКЛАЙН ЭлЭлСи Способ и фармацевтическая комбинация для лечения рака
AU2010307043C1 (en) * 2009-10-12 2014-03-13 Glaxosmithkline Llc Combination
AU2010307043B2 (en) * 2009-10-12 2013-10-31 Glaxosmithkline Llc Combination
WO2011046894A1 (en) * 2009-10-12 2011-04-21 Glaxosmithkline Llc Combination
WO2011115071A1 (ja) * 2010-03-15 2011-09-22 国立大学法人広島大学 チエノピリジン誘導体及びその製造方法、並びにそれを用いた有機半導体デバイス
CN102884068A (zh) * 2010-03-15 2013-01-16 国立大学法人广岛大学 噻吩并吡啶衍生物及其制备方法、以及使用该衍生物的有机半导体器件
JP2013525303A (ja) * 2010-04-16 2013-06-20 アッヴィ・インコーポレイテッド フタラジン−(2h)−オン系キナーゼ阻害薬
WO2011130921A1 (en) * 2010-04-23 2011-10-27 Merck Sharp & Dohme Corp. Inhibitors of akt activity
EP2832731A4 (en) * 2012-03-27 2015-08-19 Shionogi & Co AROMATIC HETEROCYCLIC CHAIN 5-CORE DERIVED DERIVATIVE HAVING TRPV4 INHIBITORY ACTIVITY
US9499533B2 (en) 2012-03-27 2016-11-22 Shionogi & Co., Ltd. Aromatic 5-membered heterocyclic derivative having TRPV4-Inhibiting activity
WO2013173382A1 (en) 2012-05-15 2013-11-21 Amgen Inc. Benzothiophene sulfonamides and other compounds that interact with glucokinase regulatory protein
JP2018510857A (ja) * 2015-03-02 2018-04-19 ライジェル ファーマシューティカルズ, インコーポレイテッド TGF−β阻害剤
US11566003B2 (en) 2017-03-30 2023-01-31 Genentech, Inc. Isoquinolines as inhibitors of HPK1
US11479770B2 (en) 2017-10-05 2022-10-25 Fulcrum Therapeutics, Inc. Use of p38 inhibitors to reduce expression of DUX4
US11291659B2 (en) 2017-10-05 2022-04-05 Fulcrum Therapeutics, Inc. P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD
US10537560B2 (en) 2017-10-05 2020-01-21 Fulcrum Therapeutics. Inc. P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD
US10342786B2 (en) 2017-10-05 2019-07-09 Fulcrum Therapeutics, Inc. P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD
US11273158B2 (en) 2018-03-05 2022-03-15 Alkermes Pharma Ireland Limited Aripiprazole dosing strategy
US11612606B2 (en) 2018-10-03 2023-03-28 Genentech, Inc. 8-aminoisoquinoline compounds and uses thereof
CN111603466B (zh) * 2020-06-29 2022-07-22 江南大学 一种乙酮类化合物在制备治疗肿瘤药物中的应用
CN111603466A (zh) * 2020-06-29 2020-09-01 江南大学 一种乙酮类化合物在制备治疗肿瘤药物中的应用
CN114014787A (zh) * 2022-01-10 2022-02-08 苏州开元民生科技股份有限公司 一种制备(2s,3r)-对甲砜基苯丝氨酸乙酯的不对称合成方法

Also Published As

Publication number Publication date
US7514566B2 (en) 2009-04-07
KR20080091369A (ko) 2008-10-10
BRPI0706621A2 (pt) 2011-04-05
NO20083572L (no) 2008-10-17
ZA200806386B (en) 2009-11-25
IL192751A0 (en) 2009-02-11
US20070173506A1 (en) 2007-07-26
CN101421265A (zh) 2009-04-29
EA200801716A1 (ru) 2009-04-28
UA91895C2 (en) 2010-09-10
AU2007207743B2 (en) 2010-07-08
JP5199885B2 (ja) 2013-05-15
TW200738657A (en) 2007-10-16
AR059064A1 (es) 2008-03-12
US20090270445A1 (en) 2009-10-29
AU2007207743A1 (en) 2007-07-26
US8084479B2 (en) 2011-12-27
CA2636077A1 (en) 2007-07-26
UY30098A1 (es) 2007-10-31
MY149143A (en) 2013-07-15
JP2009525960A (ja) 2009-07-16
PE20071114A1 (es) 2008-01-10
ES2389062T3 (es) 2012-10-22
CR10211A (es) 2008-10-03
WO2007084391A3 (en) 2008-03-20
EP1981884A2 (en) 2008-10-22
CA2636077C (en) 2012-01-03
EP1981884B1 (en) 2012-06-13

Similar Documents

Publication Publication Date Title
EP1981884B1 (en) Thiazole compounds as protein kinase b (pkb) inhibitors
US7919504B2 (en) Thiadiazole modulators of PKB
US7897619B2 (en) Heterocyclic modulators of PKB
US20110263647A1 (en) Fluoroisoquinoline substituted thiazole compounds and methods of use
ES2538581T3 (es) Compuestos de pirimidina que inhiben la quinasa de linfoma anaplásico
US6822097B1 (en) Compounds and methods of uses
US9505749B2 (en) Quinazolinone compounds and derivatives thereof
EP2822944A1 (en) Oxazolidinone compounds and derivatives thereof
WO2003101985A1 (en) 2-oxo-1,3,4-trihydroquinazolinyl derivatives for the treatment of cell proliferation-related disorders
US20040147561A1 (en) Pyrid-2-one derivatives and methods of use
CA2486530A1 (en) 2-oxo-1,3,4-trihydroquinazolinyl derivatives for the treatment of cell proliferation-related disorders

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2636077

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2008071135

Country of ref document: EG

WWE Wipo information: entry into national phase

Ref document number: 12008501616

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 569706

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 192751

Country of ref document: IL

Ref document number: 2007207743

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: MX/a/2008/009048

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2007718125

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2008551297

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 3974/CHENP/2008

Country of ref document: IN

ENP Entry into the national phase

Ref document number: 2007207743

Country of ref document: AU

Date of ref document: 20070111

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1020087019941

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: CR2008-010211

Country of ref document: CR

Ref document number: 200801716

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 200780009708.1

Country of ref document: CN

ENP Entry into the national phase

Ref document number: PI0706621

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20080718