WO2003051847A1 - (1-h-indazol-3-yl) -amide derivatives as gsk-3 inhibitors - Google Patents

(1-h-indazol-3-yl) -amide derivatives as gsk-3 inhibitors Download PDF

Info

Publication number
WO2003051847A1
WO2003051847A1 PCT/GB2002/005826 GB0205826W WO03051847A1 WO 2003051847 A1 WO2003051847 A1 WO 2003051847A1 GB 0205826 W GB0205826 W GB 0205826W WO 03051847 A1 WO03051847 A1 WO 03051847A1
Authority
WO
WIPO (PCT)
Prior art keywords
formula
compound
substituted
unsubstituted
aryl
Prior art date
Application number
PCT/GB2002/005826
Other languages
French (fr)
Inventor
Vincent Julien Eugene Bordas
Robert William Ward
Jason Witherington
Original Assignee
Smithkline Beecham P.L.C.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GB0130352A external-priority patent/GB0130352D0/en
Priority claimed from GB0206612A external-priority patent/GB0206612D0/en
Application filed by Smithkline Beecham P.L.C. filed Critical Smithkline Beecham P.L.C.
Priority to AU2002353186A priority Critical patent/AU2002353186A1/en
Publication of WO2003051847A1 publication Critical patent/WO2003051847A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/54Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings condensed with carbocyclic rings or ring systems
    • C07D231/56Benzopyrazoles; Hydrogenated benzopyrazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/04Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/04Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond

Definitions

  • GSK-3 is a serine/threonine protein kinase composed of two isoforms ( ⁇ and ⁇ ) which are encoded by distinct genes.
  • GSK-3 is one of several protein kinases which phosphorylates glycogen synthase (GS) (Embi et al, Eur. J. Biochem., (107), 519-527, (1980)).
  • GS glycogen synthase
  • Type II diabetes or Non-Insulin Dependent Diabetes Mellitus, NIDDM
  • Hyperglycaemia is due to insulin resistance in the liver, muscle and other tissues coupled with inadequate or defective secretion of insulin from pancreatic islets.
  • Skeletal muscle is the major site for insulin-stimulated glucose uptake and in this tissue, glucose removed from the circulation is either metabolised through glycolysis and the TCA cycle, or stored as glycogen.
  • Muscle glycogen deposition plays the more important role in glucose homeostasis and Type II diabetic subjects have defective muscle glycogen storage.
  • glycogen synthase The stimulation of glycogen synthesis by insulin in skeletal muscle results from the dephosphorylation and activation of glycogen synthase (Villar-Palasi C. and Lamer J., Biochim. Biophys. Ada., (39), 171-173, (1960), Parker PJ. et al, Eur. J. Biochem., (130), 227-234, (1983) and Cohen P., Biochem. Soc. Trans., (21), 555-567, (1993)).
  • the phosphorylation and dephosphorylation of GS are mediated by specific kinases and phosphatases.
  • GSK-3 is responsible for phosphorylation and deactivation of GS, while glycogen bound protein phosphatase 1 (PP1G) dephosphorylates and activates GS. Insulin both inactivates GSK-3 and activates PP1G (Srivastava A.K. and Pandey S.K., Mol and Cellular Biochem., (182), 135-141, (1998)). Chen et al. (Diabetes, (43), 1234-1241, (1994)) found that there was no difference in the mRNA abundance of PP1G between patients with Type II diabetes and control patients, suggesting that an increase in GSK-3 activity might be important in Type II diabetes.
  • GSK-3 is overexpressed in Type II diabetic muscle and that an inverse correlation exists between skeletal muscle GSK-3 activity and insulin action (Nikoulina et al, Diabetes, (49), 263-271, (2000)).
  • Overexpression of GSK-3 ⁇ and constirutively active GSK-3 ⁇ (S9A, S9E) mutants in 5 HEK-293 cells resulted in suppression of glycogen synthase activity (Eldar-Finkelman et al, PNAS, (93), 10228-10233, (1996)) and overexpression of GSK-3 ⁇ in CHO cells, expressing both insulin receptor and insulin receptor substrate 1 (IRS-1), resulted in an impairment of insulin action (Eldar-Finkelman and Krebs, PNAS, (94), 9660-9664, (1997)).
  • GSK-3 has been shown to phospliorylate other proteins in vitro including the eukaryotic initiation factor eIF-2B at Serine 540 (Welsh et al, FEBS Letts., (421), 125-
  • GSK-3 activity is subject to inhibitory phosphorylation by PI 3 kinase-mediated or Wnt-1 class-mediated signals that can be mimicked by treatment with Uthium, a low mM inhibitor of GSK-3 (Stambolic V., Ruel L. and Woodgett J.R., Curr. Biol, (6), 1664-8, (1996)).
  • 5 GSK-3 inhibitors may be of value as neuroprotectants in treatment of acute stroke and other neurotraumatic injuries.
  • GSK-3 is one of a number of PKB/akt substrates to be identified that can contribute to the inhibition of apoptosis via this pathway (Pap and Cooper, J. Biol. Chem., (273), 19929-19932, ((1998)).
  • Evidence 0 suggests that astrocytic glycogen can provide an alternative energy source to facilitate neuronal survival under conditions of glucose deprivation (for example, see Ransom B.R. and Fern R., Glia, (21), 134-141, (1997) and references therein).
  • Lithium is known to protect cerebellar granule neurons from death (D'Mello et al, Exp.
  • Tau and ⁇ -catenin, two known in vivo substrates of GSK-3, are of direct relevance in consideration of further aspects of the value of GSK-3 inhibitors in relation to treatment of chronic neurodegenerative conditions.
  • Tau hyperphosphorylation is an early event in neurodegenerative conditions such as Alzheimer's disease (AD), and is postulated to promote microtubule disassembly.
  • AD Alzheimer's disease
  • Lithium has been reported to reduce the phosphorylation of tau, enhance the binding of tau to microtubules, and promote microtubule assembly through direct and reversible inhibition of glycogen synthase kinase-3 (Hong M., Chen D.C., Klein P.S. and Lee V.M., J. Biol.
  • ⁇ -catenin is phosphorylated by GSK-3 as part of a tripartite complex with axin, resulting in ⁇ -catenin being targetted for degradation (D eda et al, J. EMBO., (17), 1371-1384, (1998)).
  • Inhibition of GSK-3 activity is a key mechanism by which cytosolic levels of catenin are stabilised and hence promote ⁇ -catenin-LEF-1/TCF transcnptional activity (Eastman, Grosschedl, Curr. Opin. Cell. Biol, (11), 233, (1999)).
  • the compounds of the present invention are indazole derivatives.
  • Other indazole derivatives have been described previously for use in alternative medicinal applications.
  • International Application, Publication Number WO 93/23404 describes a series of bicyclic compounds, including substituted indazoles, which are stated to modulate endothelin activity and may accordingly be of use in the treatment of conditions such as asthma, hypertension, renal failure and endotoxin shock.
  • International Application, Publication Number WO 94/14780 describes a series of heterocyclic compounds, including indazoles, which are stated to have biological activity as neuronal nitric oxide synthase inhibitors, and as such may be useful in the treatment of, for example, cerebral ischaemia.
  • indazoles are potent and selective inhibitors of GSK-3. These compounds are indicated to be useful for the treatment and/or prophylaxis of conditions associated with a need for inhibition of GSK-3, such as diabetes, conditions associated with diabetes, chronic neurodegenerative conditions including dementias such as Alzheimer's disease, Parkinson's disease, progressive supranuclear palsy, subacute sclerosing panencephalitic parkinsonism, postencephalitic parkinsonism, pugilistic encephalitis, guam parkinsonism-dementia complex, Pick's disease, corticobasal degeneration, frontotemporal dementia, Huntingdon's disease, ADDS associated dementia, amyotrophic lateral sclerosis, multiple sclerosis and neurotraumatic diseases such as acute stroke, mood disorders such as schizophrenia and bipolar disorders, promotion of functional recovery post stroke, cerebral bleeding (for example, due to solitary cerebral amyloid angiopathy), hair loss, obesity, atherosclerotic cardiovascular disease, hypertension
  • dementias such as Alzheimer'
  • the present invention provides a compound of formula ®,
  • Rl is unsubstituted or substituted alkyl, unsubstituted or substituted cycloalkyl, unsubstituted or substituted alkenyl, unsubstituted or substituted cycloalkenyl, unsubstituted or substituted aryl, aralkyl wherein the aryl and the alkyl moieties may each independently be unsubstituted or substituted, aralkenyl wherein the aryl and alkenyl moieties may each independently be unsubstituted or substituted, unsubstituted or substituted heterocyclyl, or heterocyclylalkyl wherein the heterocyclyl and the alkyl moieties may each independently be unsubstituted or substituted, unsubstituted or substituted heteroaryl, heteroarylalkyl wherem the heteroaryl and the alkyl moieties may each independently be unsubstituted or substituted;
  • R 2 is H
  • R3 is H, halo, alkyl, unsubstituted or substituted aryl or unsubstituted or substituted heteroaryl;
  • R4 is H, unsubstituted or substituted aryl; unsubstituted or substituted heteroaryl, unsubstituted or substituted heterocyclyl; or -X-aryl;
  • R 5 is H
  • X is O, NH, S or S(O) n and n is 1 or 2; with the proviso that when R 1 , R 2 and R ⁇ are as defined in relation to formula (I), and R ⁇ is H or halo, then R 4 is unsubstituted or substituted aryl; unsubstituted or substituted heteroaryl, unsubstituted or substituted heterocyclyl; or -X-aryl.
  • R* is alkyl, alkenyl, cycloC3_g alkyl, cycloC3_g alkenyl, di- alkylaminoalkyl, arylalkyl, arylalkenyl, heterocyclyl wherein the heterocyclyl group may be optionally substituted by one or more groups selected from alkyl, arylalkyl and alkoxyalkyl; heterocyclylalkyl wherein the heterocyclyl group may be optionally substituted by one or more groups selected from alkoxyalkyl, aryloxyalkyl, arylalkyl and alkyl; heteroarylalkyl wherein the heteroaryl may be optionally substituted by one or more groups selected from alkyl; heteroaryl wherein the heteroaryl may be optionally substituted by one or more groups selected from aryl and heteroaryl; aryl wherein the aryl group may be optionally substituted by heterocyclylalkyl and di-alkylaminoalky
  • R is alkyl, cycloC3_g alkyl or heterocyclyl wherein the heterocyclyl group is substituted by arylalkyl.
  • Rl is n-propyl, cyclopropyl, cyclopentyl and N- benzylpyrrolidin-3-yl.
  • R ⁇ is H, halo or aryl.
  • R ⁇ is H, bromo or phenyl.
  • R 4 is H, aryl wherein the aryl group may be optionally substituted by one or more substituents selected from halo, -SO2NH2, -OH, -NHSO2Me and -S ⁇ 2Me; heteroaryl wherein the heteroaryl group may be optionally substituted by one or more substituents selected from halo, alkyl and oxo; heterocyclyl wherein the heterocyclyl group may be optionally substituted by benzyloxycarbonyl; or -X-aryl.
  • R 4 is H, phenyl, 2-fluorophenyl, 3-fluorophenyl, 4-fluorophenyl, 2,3-difluorophenyl, 2,5- difluorophenyl, 3,5-difluoro ⁇ henyl, 3-H2NSO 2 -Ph, 4-H2NSO2-PI1, 3-MeSO2NH-Ph, 4-
  • MeSO2NH-Ph 4-methanesulfonylphenyl, 4-hydroxyphenyl, 2-furyl, 3-furyl, 2-pyrrolyl,
  • Rl is alkyl, cyck>C3_g alkyl or heterocyclyl wherein the heterocyclyl group is substituted by arylalkyl;
  • R 2 is H;
  • R3 is H, halo or aryl;
  • R4 is H, aryl wherein the aryl group may be optionally substituted by one or more substituents selected from halo, -SO2NH2, -OH, -NHSO2Me and -SO2Me; heteroaryl wherein the heteroaryl group may be optionally substituted by one or more substituents selected from halo, alkyl and oxo; heterocyclyl wherein the heterocyclyl group may be optionally substituted by aryloxycarbonyl; or -X-aryl; with the proviso that when R , R 2 and R ⁇ are as defined in relation to formula (IA), and
  • R3 is H or halo
  • R 4 is aryl wherein the aryl group may be optionally substituted by one or more substituents selected from halo, -SO2NH2, -OH, -NHSO2Me and -SO2Me; heteroaryl wherein the heteroaryl group may be optionally substituted by one or more substituents selected from halo, alkyl and oxo; heterocyclyl wherein the heterocyclyl group may be optionally substituted by aryloxycarbonyl; or -X-aryl.
  • Rl is n-propyl, cyclopropyl, cyclopentyl or N-benzyl ⁇ yrrolidin-3-yl;
  • R 2 is H;
  • R3 is H, bromo or phenyl;
  • R4 is H, phenyl, 2-fluorophenyl, 3 -fluorophenyl, 4-fluorophenyl, 2,3-difluorophenyl, 2,5- difluorophenyl, 3,5-difluorophenyl, 3-H 2 NSO 2 -Ph, 4-H 2 NSO 2 -Ph, 3-MeSO 2 NH-Ph, 4-
  • MeSO2 H-Ph 4-methanesulfonylphenyl, 4-hydroxyphenyl, 2-furyl, 3-furyl, 2-pyrrolyl,
  • R 5 is H; with the proviso that when R , R2 and R ⁇ are as defined in relation to formula (IB), and
  • R3 is H or bromo
  • R 4 is phenyl, 2-fluorophenyl, 3-fluorophenyl, 4-fluorophenyl, 2,3-difluorophenyl, 2,5-difluorophenyl, 3,5-difluorophenyl, 3-H 2 NSO 2 -Ph, 4-H2NSO2- Ph, 3-MeSO2NH-Ph, 4-MeS ⁇ 2NH-Ph, 4-methanesulfonylphenyl, 4-hydroxyphenyl, 2- furyl, 3-furyl, 2-pyrrolyl, 2-thienyl, 3-thienyl, 4-methylthien-2-yl, 5-bromothien-2-yl, indol-5-yl, pyrid-2-on-5-yl, -NHPh, -OPh, -SPh, -SO2PI1, piperazin-1-yl or 4- benzyloxycarbonylpiperazin-1-yl.
  • Certain compounds of formula (I) may contain chiral atoms and/or multiple bonds, and hence may exist in one or more stereoisomeric forms.
  • the present invention encompasses all of the isomeric forms of the compounds of formula (I) whether as individual isomers or as mixtures of isomers, including geometric isomers and racemic modifications.
  • alkyl refers to a straight or branched chain saturated aliphatic hydrocarbon radical containing 1 to 12 carbon atoms, suitably 1 to 6 carbon atoms.
  • alkyl groups in particular include methyl ("Me”), ethyl (“Et”), n-propyl (“Pr n “), ⁇ o-propyl ("Pr 1 "), n-butyl ("Bu n “), sec-butyl (“Bu s “), tert- butyl (“Bu*”), pentyl and hexyl.
  • alkyl groups may be substituted by one or more groups selected from halo (such as fluoro, chloro, bromo), -CN, -CF3, -
  • alkenyl as a group or part of a group refers to a straight or branched chain mono- or poly-unsaturated aliphatic hydrocarbon radical containing 2 to 12 carbon atoms, suitably 2 to 6 carbon atoms.
  • References to “alkenyl” groups include groups which may be in the E- or Z-form or mixtures thereof. Such alkenyl groups in particular include ethenyl, propenyl, butenyl, pentenyl and hexenyl.
  • alkenyl groups may be substituted by one or more groups selected from halo (such as fluoro, chloro, bromo), -CN, -CF3, -OH, -OCF3, C ⁇ _6 alkyl, C 3 _6 alkynyl, C . alkoxy, aryl and di-C ⁇ _6 alkylamino.
  • halo such as fluoro, chloro, bromo
  • alkynyl refers to hydrocarbon groups of either straight or branched configuration with one or more carbon-carbon triple bonds which may occur at any stable point in the chain, containing 3 to 12 carbon atoms, suitably 3 to 6 carbon atoms.
  • alkynyl groups in particular include propynyl, butynyl and pentynyl.
  • alkynyl groups may be substituted by one or more groups selected from halo (such as fluoro, chloro, bromo), -CN, -CF3, -OH, -OCF3, C ⁇ alkyl, C2-6 alkenyl,
  • alkoxy refers to an alkyl ether radical, wherein the term “alkyl” is defined above.
  • alkoxy groups in particular include methoxy, ethoxy, n-propoxy, tso-propoxy, n-butoxy, zso-butoxy, sec-butoxy and tert-butoxy.
  • alkoxy groups may be substituted by one or more groups selected from halo (such as fluoro, chloro, bromo), -CN, -CF3, -OH, -OCF3, Cj.g alkyl, C2_6 alkenyl, C2- alkynyl, aryl and di-Ci ⁇ g alkylamino.
  • halo such as fluoro, chloro, bromo
  • aryl as a group or part of a group refers to a carbocyclic aromatic radical.
  • aryl groups are 5-6 membered monocyclic groups or 8-10 membered fused bicyclic groups, especially phenyl ("Ph”), biphenyl and naphthyl, particularly phenyl.
  • Such aryl groups may be optionally substituted with one or more substituents, which maybe the same or different, selected from halo (such as fluoro, chloro, bromo), -CN, -CF3, -OH, -OCF3, -NO 2 , C ⁇ - alkyl, C2-6 alkenyl, 0, ⁇ -6 alkynyl, C j _g alkoxy and di-C ⁇ _g alkylamino.
  • substituents such as fluoro, chloro, bromo
  • heteroaryl as a group or part of a group refers to stable heterocyclic aromatic single and fused rings containing one or more hetero atoms independently selected from nitrogen, oxygen and sulfur.
  • a fused heteroaryl ring system may include carbocyclic rings and need include only one heteroaryl ring.
  • heteroaryl groups include furyl, thienyl, pyridazinyl, pyridyl, quinolinyl, indolyl, thiazolyl, benzoxazolyl, and benzothiazolyl.
  • Each ring may be optionally substituted with one or more substituents, which maybe the same or different, selected from halo (such as fluoro, chloro, bromo), -CN, -CF3, -OH, -NO2, -OCF3, C ⁇ _6 alkyl, C2-6 alkenyl, C3_6 alkynyl, C ⁇ _6 alkoxy, aryl, heteroaryl, and di-C 1 -6 alkylamino.
  • substituents such as fluoro, chloro, bromo
  • a fused heterocyclyl ring system may include carbocyclic rings and need include only one heterocyclic ring.
  • Such heterocyclyl groups include piperazinyl, piperidinyl and n orpholinyl.
  • Each ring may be optionally substituted with one or more substituents, which may be the same or different, selected from halo (such as fluoro, chloro, bromo), - CN, -CF3, -OH, -NO 2 , -OCF3, C ⁇ _6 alkyl, C 2 _6 alkenyl, C3_6 alkynyl, C _6 alkoxy, aryl, heteroaryl, and di-C ⁇ _6 alkylamino.
  • halo such as fluoro, chloro, bromo
  • -CN -CF3, -OH, -NO 2 , -OCF3, C ⁇ _6 alkyl, C 2 _6 alkenyl, C3_6 alkyny
  • halo include iodo, bromo, chloro or fluoro, suitably bromo, chloro and fluoro, especially bromo and chloro.
  • Composite terms such as “alkoxyalkyl” and “arylalkyl” refer to substituents comprising two interlinked groups, with the group named latterly in the term being the linking group, so that “alkoxyalkyl” means -(alkyl)-(alkoxy) whilst “arylalkyl” means - (alkyl)-(aryl).
  • the compounds of formula (I) or their salts or solvates are preferably in pharmaceutically acceptable or substantially pure form.
  • pharmaceutically acceptable form is meant, inter alia, having a pharmaceutically acceptable level of purity excluding normal pharmaceutical additives such as diluents and carriers, and including no material considered toxic at normal dosage levels.
  • a substantially pure form will generally contain at least 50% (excluding normal pharmaceutical additives), preferably 75%, more preferably 90% and still more preferably 95% of the compound of formula (I) or its salt or solvate.
  • One preferred pharmaceutically acceptable form is the crystalline form, including such form in pharmaceutical composition.
  • the additional ionic and solvent moieties must also be non-toxic.
  • Suitable salts are pharmaceutically acceptable salts.
  • Suitable pharmaceutically acceptable salts include the acid addition salts with the conventional pharmaceutical acids, for example maleic, hydrochloric, hydrobromic, phosphoric, acetic, fumaric, salicylic, citric, lactic, mandelic, tartaric, succinic, benzoic, ascorbic and methanesulfonic.
  • Suitable pharmaceutically acceptable salts include salts of acidic moieties of the compounds of formula (I) when they are present, for example salts of carboxy groups or phenolic hydroxy groups.
  • Suitable salts of acidic moieties include metal salts, such as for example aluminium, alkali metal salts such as lithium, sodium or potassium, alkaline earth metal salts such as calcium or magnesium and ammonium or substituted ammonium salts, for example those with lower alkylamines such as triethylamine, hydroxyalkylamines such as 2-hydroxyethylamine, bis-(2-hy ⁇ koxyethyl)-amine or tri-(2-hydroxyethyl)-amine, cycloalkylamines such as bicyclohexylamine, or with procaine, dibenzylpiperidine, N-benzyl- ⁇ -phenethylamine, dehydroabietylamine, N,N'-bisdehydroabietylamine, glucamine, N-methylglucamine or bases of the pyridine type such as pyridine, collidine, quinine or quinoline.
  • Suitable solvates are pharmaceutically acceptable solvates.
  • Suitable pharmaceutically acceptable solvates include hydrates.
  • diabetes mellitus especially Type 2 diabetes, and conditions associated with diabetes mellitus.
  • condition associated with diabetes includes those conditions associated with the pre-diabetic state, conditions associated with diabetes mellitus itself and complications associated with diabetes mellitus.
  • condition associated with the pre-diabetic state includes conditions such as insulin resistance, impaired glucose tolerance and hyperinsulinaemia.
  • condition associated with diabetes mellitus itself includes hyperglycaemia, insulin resistance and obesity. Further conditions associated with diabetes mellitus itself include hypertension and cardiovascular disease, especially atherosclerosis and conditions associated with insulin resistance. Conditions associated with insulin resistance include polycystic ovarian syndrome and steroid induced insulin resistance.
  • components associated with diabetes mellitus includes renal disease, especially renal disease associated with Type II diabetes, neuropathy and retinopathy.
  • Renal diseases associated with Type II diabetes include nephropathy, glomerulonephritis, glomerular sclerosis, nephrotic syndrome, hypertensive nephrosclerosis and end stage renal disease.
  • neurotraumatic diseases includes both open or penetrating head trauma, such as caused by surgery, or a closed head trauma injury, such as caused by an injury to the head region, ischaemic stroke including acute stroke, particularly to the brain area, transient ischaemic attacks following coronary by-pass and cognitive decline following other transient ischaemic conditions.
  • a process for the preparation of a compound of formula (I), or a salt and/or solvate thereof comprises reacting a compound of formula (II), wherein R 2 , R3, R4 nd R ⁇ are as defined in relation to formula (I) with a compound of formula (HI),
  • R is as defined in relation to formula (I) and X is a suitable leaving group and thereafter, if required, carrying out one or more of the following optional steps: (i) converting a compound of formula (I) to a further compound of formula (I); (ii) removing any necessary protecting group;
  • X is chloro. It will be appreciated that compounds of formula (III) may also include related carboxylic acid anhydrides.
  • the reaction between the compounds of formulae (II) and (HI) is carried out in a suitable solvent, under conventional conditions, at a suitable temperature, providing a suitable rate of formation of the required product, over a suitable reaction time.
  • a suitable solvent is pyridine.
  • Suitable reaction temperatures include those in the range of 20°C to 220°C and, as appropriate, the reflux temperature of the solvent. Suitable reaction times are those in the range 12 to 72 hours.
  • the reaction products are isolated using conventional methods. Conventional methods of heating and cooling may be employed, for example thermostatically controlled oil baths and ice/salt baths respectively.
  • the reaction products are typically purified by conventional methods, such as crystallisation, chromatography and trituration. Crystalline product may be obtained by standard methods.
  • a compound of formula (III) such as n-butyric anhydride
  • a compound of formula (II) is added to a solution of a compound of formula (II) in pyridine.
  • the reaction mixture is heated under reflux for 22 hours and allowed to cool to ambient temperature.
  • the resulting solution is concentrated in vacuo and purified by column chromatography using one or more suitable solvents, such as 10% v/v methanol in chloroform, to afford the desired compound of formula (I).
  • a compound of formula (IH) such as cyclopropanecarbonyl chloride
  • R 2 , R ⁇ , R4 and R ⁇ are as defined in relation to formula (I).
  • a process for the preparation of a compound of formula (I), or a salt and/or solvate thereof which process comprises reacting a compound of formula (IV) with a nucleophile and thereafter, if required, carrying out one or more of the following optional steps: (i) converting a compound of formula (I) to a further compound of formula (I); (ii) removing any necessary protecting group;
  • reaction between the compound of formula (TV) and a nucleophile is carried out optionally in a suitable solvent, under conventional conditions, at a suitable temperature, providing a suitable rate of formation of the required product, over a suitable reaction time.
  • a suitable nucleophile is an amine, such as a primary or secondary amine.
  • Suitable reaction temperatures include those in the range of 20°C to 100°C and, as appropriate, the reflux temperature of the solvent. Suitable reaction times are those in the range 1 to 48 hours.
  • the reaction products are isolated using conventional methods.
  • reaction products are typically purified by conventional methods, such as crystallisation, chromatography and trituration. Crystalline product may be obtained by standard methods.
  • a suitable nucleophile such as piperidine
  • water are added to a compound of formula (IV) under argon.
  • the reaction mixture is stirred for 3 hours at ambient temperature.
  • the resulting mixture is concentrated in vacuo and the crude product purified by preparative HPLC using one or more suitable solvents, such as 10-90% acetonitrile/0.1 % trifluoroacetic acid in water/0.1 % trifluoroacetic acid.
  • suitable solvents such as 10-90% acetonitrile/0.1 % trifluoroacetic acid in water/0.1 % trifluoroacetic acid.
  • the appropriate fractions are concentrated to dryness to afford the desired compound of formula (I).
  • the above-mentioned conversions of a compound of formula (I) into another compound of formula (I) include any conversion, which maybe effected using conventional procedures, but in particular the said conversions include any combination of:
  • Suitable conversions of one group R ⁇ into another group R ⁇ include:
  • R ⁇ is halo, such as bromo, with an arylboronic acid or an arylboronate, such as phenylboronic acid.
  • Suitable conversions of one group R 4 into another group R 4 include:
  • Rl, R 2 , R ⁇ and R ⁇ are as defined in relation to formula (I), may be converted to a compound of formula (I) where R 4 is - ⁇ HAr by reaction with a suitable acid, such as hydrochloric acid in 1,4-dioxane.
  • a suitable acid such as hydrochloric acid in 1,4-dioxane.
  • Compounds of formula (II) may be prepared by reaction of a compound of formula (VI), wherein,
  • R 2 , R3, R4 and R ⁇ are as defined in relation to formula (I), with hydrazine or a hydrate thereof.
  • the reaction between the compound of formula (VI) and hydrazine or a hydrate thereof, is carried out in a suitable solvent at a suitable temperature, generally an elevated temperature, providing a suitable rate of formation of the required product, over a suitable reaction time.
  • suitable solvents include pyridine and ethanol.
  • Suitable reaction temperatures include those in the range of 60 °C to 220 °C and, as appropriate, the reflux temperature of the solvent. Suitable reaction times are those in the range 1-72 hours.
  • the reaction products are isolated using conventional methods. Typically, the reaction mixture is cooled, the product isolated by filtration, and dried. Conventional methods of heating and cooling may be employed, for example thermostatically controlled oil baths and ice/salt baths respectively.
  • the reaction products may, if desired, be purified by conventional methods, such as crystallisation, chromatography and trituration.
  • hydrazine hydrate is added to a stirred solution of the compound of formula (VI) in pyridine.
  • the reaction mixture is stirred under reflux for 72 hours and cooled.
  • the resulting mixture is concentrated in vacuo, and the residue dissolved in ethyl acetate and washed with aqueous sodium bicarbonate solution.
  • the organic extract is dried over magnesium sulfate and concentrated in vacuo.
  • the crude product is triturated with a suitable solvent, such as dichloromethane to afford the desired compound of formula (II).
  • compounds of formula (fl) are believed to be novel and accordingly form a further aspect of the present invention.
  • compounds of formula (IV) may be prepared by reaction of a compound of formula (II) with a compound of formula (in), in the presence of a suitable solvent such as pyridine.
  • compounds of formula (V) may be prepared by reaction of a compound of formula (VII),
  • R 2 , R3, R4 and R ⁇ are as defined in relation to formula (I), with a compound of formula (HI).
  • the synthesis of compounds of formula (V) by reaction of a compound of formula (NH) with a compound of formula (Hi) may be undertaken using analogous acylation conditions to those used for the preparation of compounds of formula (I) as described hereinbefore.
  • a suitable base is KOAc.
  • Suitable reaction temperatures include those in the range of 60 °C to 220 °C and, as appropriate, the reflux temperature of the solvent. Suitable reaction times are those in the range 1-72 hours.
  • the reaction products are isolated using conventional methods. Typically, the reaction mixture is cooled, the product isolated by filtration, and dried. Conventional methods of heating and cooling may be employed, for example thermostatically controlled oil baths and ice/salt baths respectively.
  • the reaction products may, if desired, be purified by conventional methods, such as crystallisation, chromatography and trituration.
  • R 2 , R ⁇ and R $ are as defined in relation to formula (I), with a compound of formula (XI), R— Y (XI)
  • R 4 is as defined in relation to formula (I) and Y is halo, such as bromo, in the presence of a suitable catalyst.
  • the reaction between the compound of formula (X) and a compound of formula (XI) is carried out in a suitable solvent in the presence of a suitable catalyst and a suitable base, at a suitable temperature, generally an elevated temperature, providing a suitable rate of formation of the required product, over a suitable reaction time.
  • suitable solvents include a mixture of dimethylformamide, ethanol and water.
  • a suitable catalyst is PdCl2(dppf).
  • a suitable base is KOAc.
  • Suitable reaction temperatures include those in the range of 60 °C to 220 °C and, as appropriate, the reflux temperature of the solvent. Suitable reaction times are those in the range 1-72 hours.
  • the reaction products are isolated using conventional methods. Typically, the reaction mixture is cooled, the product isolated by filtration, and dried. Conventional methods of heating and cooling may be employed, for example thermostatically controlled oil baths and ice/salt baths respectively.
  • the reaction products may, if desired, be purified by conventional methods, such as crystallisation, chromatography and tri
  • the reaction between the compound of formula (XII) and a compound of formula (X ⁇ i) is carried out in a suitable solvent in the presence of a suitable base, at a suitable temperature, providing a suitable rate of formation of the required product, over a suitable reaction time.
  • a suitable solvent is dimethylformamide.
  • a suitable base is sodium hydride.
  • Suitable reaction temperatures include those in the range of 20 °C to 100 °C. Suitable reaction times are those in the range 1-48 hours.
  • the reaction products are isolated using conventional methods. Typically, the reaction mixture is cooled, the product isolated by filtration, and dried. Conventional methods of heating and cooling may be employed, for example thermostatically controlled oil baths and ice/salt baths respectively.
  • the reaction products may, if desired, be purified by conventional methods, such as crystallisation, chromatography and trituration.
  • a compound of formula (XIH) is added to a compound of formula (XII) in the presence of sodium hydride in dimethylformamide.
  • the reaction mixture is stirred at ambient temperature, under argon for 16 hours.
  • the resulting solution is concentrated to dryness in vacuo and purified by preparative HPLC using one or more suitable solvents, such as 10-90% acetonitrile (containing 0.1% trifluoroacetic acid) in water (containing 0.1% trifluoroacetic acid).
  • suitable solvents such as 10-90% acetonitrile (containing 0.1% trifluoroacetic acid) in water (containing 0.1% trifluoroacetic acid).
  • the appropriate fractions are concentrated to dryness to afford the desired compound of formula (VI).
  • Compounds of formula (X) may be prepared by reaction of a compound of formula (V ⁇ i) with bis(pinacolato)diboron in the presence of a suitable catalyst, such as l,r-Bis(diphenylphosphino)ferrocenedichloropalladium(II), i.e. PdCl2(dppf).
  • a suitable catalyst such as l,r-Bis(diphenylphosphino)ferrocenedichloropalladium(II), i.e. PdCl2(dppf).
  • the compounds of formula (I), or pharmaceutically acceptable salts or solvates thereof are indicated to be useful as inhibitors of glycogen synthase kinase-3.
  • the invention therefore provides a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, for use as an inhibitor of GSK-3.
  • the present invention also provides a method for the treatment of conditions associated with a need for inhibition of GSK-3 such as diabetes, conditions associated with diabetes, chronic neurodegenerative conditions including dementias such as Alzheimer's disease, Parkinson's disease, progressive supranuclear palsy, subacute sclerosing panencephalitic parkinsonism, postencephalitic parkinsonism, pugilistic encephalitis, guam parkinsonism-dementia complex, Pick's disease, corticobasal degeneration, frontotemporal dementia, Huntingdon's disease, AIDS associated dementia, amyotrophic lateral sclerosis, multiple sclerosis and neurotraurnatic diseases such as acute stroke, mood disorders such as schizophrenia and bipolar disorders, promotion of functional recovery post stroke, cerebral bleeding (for example, due to solitary cerebral amyloid angiopathy), hair loss, obesity, atheros
  • the present invention further provides a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, for use as an inhibitor of glycogen synthase kinase-3, and especially for use in the treatment of conditions associated with a need for the inhibition of GSK-3, such as diabetes, conditions associated with diabetes, chronic neurodegenerative conditions including dementias such as Alzheimer's disease, Parkinson's disease, progressive supranuclear palsy, subacute sclerosing panencephalitic parkinsonism, postencephalitic parkinsonism, pugilistic encephalitis, guam parkinsonism- dementia complex, Pick's disease, corticobasal degeneration, frontotemporal dementia, Huntingdon's disease, ADDS associated dementia, amyotrophic lateral sclerosis, multiple sclerosis and neurotraurnatic diseases such as acute stroke, mood disorders such as schizophrenia and bipolar disorders, promotion of functional recovery post stroke, cerebral bleeding (for example, due to solitary cerebral amyloid angiopathy), hair loss, obesity,
  • the present invention also provides the use of a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, for the manufacture of a medicament for the treatment of conditions associated with a need for the inhibition of GSK-3, such as diabetes, conditions associated with diabetes, chronic neurodegenerative conditions including dementias such as Alzheimer's disease, Parkinson's disease, progressive supranuclear palsy, subacute sclerosing panencephalitic parkinsonism, postencephalitic parkinsonism, pugilistic encephalitis, guam parkinsonism-dementia complex, Pick's disease, corticobasal degeneration, frontotemporal dementia, Huntingdon's disease, ADDS associated dementia, amyotrophic lateral sclerosis, multiple sclerosis and neurotraurnatic diseases such as acute stroke, mood disorders such as schizophrenia and bipolar disorders, promotion of functional recovery post stroke, cerebral bleeding (for example, due to solitary cerebral amyloid angiopathy), hair loss, obesity, atherosclerotic cardiovascular disease, hyper
  • the compounds of formula (I), or pharmaceutically acceptable salts or solvates thereof are administered as pharmaceutically acceptable compositions.
  • the invention also provides a pharmaceutical composition which comprises a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable carrier.
  • the active compounds are usually administered as the sole medicament agent but they may be administered in combination with other medicament agents as dictated by the severity and type of disease being treated.
  • the said combination comprises co-administration of a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, and an additional medicament agent or the sequential administration of a compound of formula (I), or a pharmaceutically acceptable derivative thereof, and the additional medicament agent.
  • Co-administration includes administration of a pharmaceutical composition which contains both a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, and the additional medicament agent or the essentially simultaneous administration of separate pharmaceutical compositions of a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, and the additional medicament agent.
  • compositions of the invention are preferably adapted for oral administration. However, they may be adapted for other modes of administration.
  • the compositions may be in the form of tablets, capsules, powders, granules, lozenges, suppositories, reconstitutable powders, or liquid preparations, such as oral or sterile parenteral solutions or suspensions.
  • a composition of the invention is in the form of a unit dose.
  • the composition are in unit dosage form.
  • a unit dose will generally contain from 0.1 to 1000 mg of the active compound.
  • an effective administered amount of a compound of the invention will depend on the relative efficacy of the compound chosen, the severity of the disorder being treated and the weight of the sufferer. However, active compounds will typically be administered once or more times a day for example 2, 3 or 4 times daily, with typical total daily doses in the range of from 0.1 to 800 mg/kg/day.
  • Suitable dose forms for oral administration may be tablets and capsules and may contain conventional excipients such as binding agents, for example syrup, acacia, gelatin, sorbitol, tragacanth, or polyvinylpyrrolidone; fillers, for example lactose, sugar, maize starch, calcium phosphate, sorbitol or glycine; tabletting lubricants, for example magnesium stearate; disintegrants, for example starch, polyvinylpyrrolidone, sodium starch glycollate or microcrystalline cellulose; or pharmaceutically acceptable wetting agents such as sodium lauryl sulfate.
  • binding agents for example syrup, acacia, gelatin, sorbitol, tragacanth, or polyvinylpyrrolidone
  • fillers for example lactose, sugar, maize starch, calcium phosphate, sorbitol or glycine
  • tabletting lubricants for example magnesium stearate
  • disintegrants for example starch
  • the solid oral compositions may be prepared by conventional methods of blending, filling or tabletting. Repeated blending operations may be used to distribute the active agent throughout those compositions employing large quantities of fillers. Such operations are of course conventional in the art.
  • the tablets may be coated according to methods well known in normal pharmaceutical practice, in particular with an enteric coating.
  • Oral liquid preparations may be in the form of, for example, emulsions, syrups, or elixirs, or may be presented as a dry product for reconstitution with water or other suitable vehicle before use.
  • Such liquid preparations may contain conventional additives such as suspending agents, for example sorbitol, syrup, methyl cellulose, gelatin, hydroxyethylcellulose, carboxymethylcellulose, aluminium stearate gel, hydrogenated edible fats; emulsifying agents, for example lecithin, sorbitan monooleate, or acacia; non-aqueous vehicles (which may include edible oils), for example almond oil, fractionated coconut oil, oily esters such as esters of glycerine, propylene glycol, or ethyl alcohol; preservatives, for example methyl or propyl p-hydroxybenzoate or sorbic acid; and if desired conventional flavouring or colouring agents.
  • suspending agents for example sorbitol, syrup, methyl cellulose,
  • fluid unit dosage forms are prepared utilizing the compound and a sterile vehicle, and, depending on the concentration used, can be either suspended or dissolved in the vehicle.
  • the compound can be dissolved in water for injection and filter sterilized before filling into a suitable vial or ampoule and sealing.
  • adjuvants such as a local anaesthetic, a preservative and buffering agents can be dissolved in the vehicle.
  • the composition can be frozen after filling into the vial and the water removed under vacuum.
  • Parenteral suspensions are prepared in substantially the same manner, except that the compound is suspended in the vehicle instead of being dissolved, and sterilization cannot be accomplished by filtration.
  • the compound can be sterilized by exposure to ethylene oxide before suspending in the sterile vehicle.
  • a surfactant or wetting agent is included in the composition to facilitate uniform distribution of the compound.
  • the formulations mentioned herein are carried out using standard methods such as those described or referred to in reference texts such as the British and US Pharmacopoeias, Remington's Pharmaceutical Sciences (Mack Publishing Co.), Martindale The Extra Pharmacopoeia (London, The Pharmaceutical Press) or the above- mentioned publications.
  • Suitable methods for preparing and suitable unit dosages for the additional medicament agent, such as the antidiabetic agent mentioned herein include those methods and dosages described or referred to in the above-mentioned reference texts.
  • GSK-3 assays used to test the compounds of the invention include the following protocol which is based on the ability of the kinase to phosphorylate a biotinylated 26 mer peptide, Biot- KYRRAAVPPSPSLSRHSSPHQ(S)EDEEE, the sequence of which is derived from the phosphorylation site of glycogen synthase, where (S) is a pre- phosphorylated serine as in glycogen synthase in vivo and the three consensus sites for GSK-3 specific phosphorylation are underlined.
  • the phosphorylated biotinylated peptide is then captured onto Streptavidin coated SPA beads (Amersham Technology), where the signal from the 3p [ s amplified via the scintillant contained in the beads.
  • GSK-3 was assayed in 50 mM MOPS buffer, pH 7.0, containing 5% glycerol, 0.01% Tween-20, 7.5 mM 2-mercaptoethanol, 10 mM magnesium acetate, 8 uM of the above peptide, and 10 uM [33p]_ATP. After incubation at room temperature, the reaction was stopped by addition of 50 mM EDTA solution containing the Streptavidin coated SPA beads to give a final 0.2 mgs. Following centrifugation, the microtitre plates are counted in a Trilux 1450 microbeta liquid scintillation counter (Wallac). IC50 values are generated for each compound by fitting to a four parameter model.
  • the most potent compounds of the present invention show IC50 values in the range of 1 to 500 nM.
  • Tefrakis(triphenylphosphine)palladium(0) (20 mg, 0.02 mmol) was added to a stirred solution of phenylboronic acid (56 mg, 0.46 mmol), N-(5-bromo-lH-indazol-3- yl)butyramide (100 mg, 0.35 mmol) and sodium carbonate (0.5 mL of 2M aqueous solution) in 1,2-dimethoxyethane (1 mL) and ethanol (0.5 mL). The resulting suspension was stirred at reflux for 20 hours, concentrated in vacuo and water (25 mL) added.
  • Cyclopropanecarbox lie acid (6-thiophen-2-yl-lH-indazol-3-yl)-amide
  • the filtrate was taken up in ethyl acetate (100 mL) and was washed with brine (3 x 100 mL).
  • the organic extract was dried (magnesium sulfate) and concentrated.
  • the crude solid was purified by chromatography (10% diethyl ether/60-80 petrol) to afford the title compound as a solid.
  • Examples containing a phenol substituent may be prepared by cleavage of an appropriate ether by the general procedure of Description 6. The product can then be acylated by a general method such as Synthetic Method D.
  • a hydrazone may be formed in the the cyclization step. Subsequent cleavage back to a carbonyl compound is exemplified in Description 7.
  • 6-[4-(l-Hydrazonoethyl)phenyl]-lH-indazol-3-ylamine (100 mg, 0.38 mmol) was stirred at reflux for 18 hours in a 4N hydrochloric solution in dioxane (10 mL) and water (1 mL). The reaction mixture was allowed to cool and was concentrated. The crude solid was taken up in ethyl acetate (50 mL) and washed with saturated sodium bicarbonate solution in water (50 mL). The organic extract was dried (magnesium sulfate) and concentrated. The resulting solid was purified by chromatography (ethyl acetate) to afford the title compound as a solid.
  • 6-Phenylsulfanyl-lH-indazol-3-ylamine (303 mg, 1.26 mmol) and cyclopropylcarbonyl chloride (114 ⁇ L, 1.26 mmol) were dissolved in anhydrous pyridine (5 mL). The solution was stirred for 16 hours under argon at ambient temperature. A further amount of cyclopropyl carbonyl chloride (114 ⁇ L, 1.26 mmol) was added and the reaction stirred for a further 2 hours at the same temperature. Water (2 mL) was added and the solution concentrated to dryness. Piperidine (20 mL) and water (3 mL) were added and the solution stirred at ambient temperature under argon for 3 hours.
  • Cyclopentanecarboxylic acid (6-benzenesulfonyl-lH-indazol-3-yl)-amide
  • Cyclopentanecarboxylic acid (6-benzenesulfanyl-lH-indazol-3-yl)-amide (100 mg, 0.324 mmol) and weto-chloroperbenzoic acid (168 mg, 0.972 mmol) were dissolved in dichloromethane (30 mL). The solution was stirred for 16 hours under argon at ambient temperature. A further 150 mL dichloromethane was added to dissolve the precipitate and the organic layer was washed with saturated sodium bicarbonate solution (3 x 150 mL).
  • Cyclopropanecarboxy lie acid (6-piperazin-l-yl-lH-indazol-3-yl)-amide

Abstract

Compounds of formula (I), or a salt thereof, or a solvate thereof, wherein, R1, R2, R3, R4 and R5 are as defined in the specification, a process for preparing such compounds, a pharmaceutical composition containing such compounds and the use of such compounds as GSK-3 in inhibitors.

Description

(1-H-INDAZO -3-Y ) -AMIDE DERIVATIVES AS GSK-3 INHIBITORS
This invention relates to novel compounds, in particular to novel indazole derivatives, to processes for the preparation of such compounds, to pharmaceutical compositions containing such compounds and to the use of such compounds in medicine. GSK-3 is a serine/threonine protein kinase composed of two isoforms (α and β) which are encoded by distinct genes. GSK-3 is one of several protein kinases which phosphorylates glycogen synthase (GS) (Embi et al, Eur. J. Biochem., (107), 519-527, (1980)). The α and β isoforms have a monomeric structure and are both found in mammalian cells. Both isoforms phospliorylate muscle glycogen synthase (Cross et al, BiochemicalJournal, (303), 21-26, (1994)) and these two isoforms show good homology between species (e.g. human and rabbit GSK-3α are 96% identical).
Type II diabetes (or Non-Insulin Dependent Diabetes Mellitus, NIDDM) is a multifactorial disease. Hyperglycaemia is due to insulin resistance in the liver, muscle and other tissues coupled with inadequate or defective secretion of insulin from pancreatic islets. Skeletal muscle is the major site for insulin-stimulated glucose uptake and in this tissue, glucose removed from the circulation is either metabolised through glycolysis and the TCA cycle, or stored as glycogen. Muscle glycogen deposition plays the more important role in glucose homeostasis and Type II diabetic subjects have defective muscle glycogen storage.
The stimulation of glycogen synthesis by insulin in skeletal muscle results from the dephosphorylation and activation of glycogen synthase (Villar-Palasi C. and Lamer J., Biochim. Biophys. Ada., (39), 171-173, (1960), Parker PJ. et al, Eur. J. Biochem., (130), 227-234, (1983) and Cohen P., Biochem. Soc. Trans., (21), 555-567, (1993)). The phosphorylation and dephosphorylation of GS are mediated by specific kinases and phosphatases. GSK-3 is responsible for phosphorylation and deactivation of GS, while glycogen bound protein phosphatase 1 (PP1G) dephosphorylates and activates GS. Insulin both inactivates GSK-3 and activates PP1G (Srivastava A.K. and Pandey S.K., Mol and Cellular Biochem., (182), 135-141, (1998)). Chen et al. (Diabetes, (43), 1234-1241, (1994)) found that there was no difference in the mRNA abundance of PP1G between patients with Type II diabetes and control patients, suggesting that an increase in GSK-3 activity might be important in Type II diabetes. It has also recently been demonstrated that GSK-3 is overexpressed in Type II diabetic muscle and that an inverse correlation exists between skeletal muscle GSK-3 activity and insulin action (Nikoulina et al, Diabetes, (49), 263-271, (2000)). Overexpression of GSK-3β and constirutively active GSK-3β(S9A, S9E) mutants in 5 HEK-293 cells resulted in suppression of glycogen synthase activity (Eldar-Finkelman et al, PNAS, (93), 10228-10233, (1996)) and overexpression of GSK-3β in CHO cells, expressing both insulin receptor and insulin receptor substrate 1 (IRS-1), resulted in an impairment of insulin action (Eldar-Finkelman and Krebs, PNAS, (94), 9660-9664, (1997)). Recent evidence for the involvement of elevated GSK-3 activity and the 10 development of insulin resistance and type II diabetes in adipose tissue has emerged from studies undertaken in diabetes and obesity prone C57BL/6J mice (Eldar-Finkelman et al, Diabetes, (48), 1662-1666, (1999)).
GSK-3 has been shown to phospliorylate other proteins in vitro including the eukaryotic initiation factor eIF-2B at Serine540 (Welsh et al, FEBS Letts., (421), 125-
15 130, (1998)). This phosphorylation results in an inhibition of eIF-2B activity and leads to a reduction in this key regulatory step of translation. In disease states, such as diabetes, where there is elevated GSK-3 activity this could result in a reduction of translation and potentially contribute to the pathology of the disease.
Several aspects of GSK-3 functions and regulation in addition to modulation of 0 glycogen synthase activity indicate that inhibitors of this enzyme may be effective in treatment of disorders of the central nervous system. GSK-3 activity is subject to inhibitory phosphorylation by PI 3 kinase-mediated or Wnt-1 class-mediated signals that can be mimicked by treatment with Uthium, a low mM inhibitor of GSK-3 (Stambolic V., Ruel L. and Woodgett J.R., Curr. Biol, (6), 1664-8, (1996)). 5 GSK-3 inhibitors may be of value as neuroprotectants in treatment of acute stroke and other neurotraumatic injuries. Roles for PI 3-kinase signalling through PKB/akt to promote neuronal cell survival are well established, and GSK-3 is one of a number of PKB/akt substrates to be identified that can contribute to the inhibition of apoptosis via this pathway (Pap and Cooper, J. Biol. Chem., (273), 19929-19932, ((1998)). Evidence 0 suggests that astrocytic glycogen can provide an alternative energy source to facilitate neuronal survival under conditions of glucose deprivation (for example, see Ransom B.R. and Fern R., Glia, (21), 134-141, (1997) and references therein). Lithium is known to protect cerebellar granule neurons from death (D'Mello et al, Exp. Cell Res., (211), 332- 338, (1994) and Nolonte et al, Neurosci. Letts., (172), 6-10, (1994)) and chronic lithium treatment has demonstrable efficacy in the middle cerebral artery occlusion model of stroke in rodents (Νonaka and Chuang, Neuroreport, (9), 2081-2084, (1998)). Wnt- induced axonal spreading and branching in neuronal culture models has been shown to correlate with GSK-3 inhibition (Lucas and Salinas, Dev. Biol, (192), 31-44, (1997)) suggesting additional value of GSK-3 inhibitors in promoting neuronal regeneration following neurotraumatic insult.
Tau and β-catenin, two known in vivo substrates of GSK-3, are of direct relevance in consideration of further aspects of the value of GSK-3 inhibitors in relation to treatment of chronic neurodegenerative conditions. Tau hyperphosphorylation is an early event in neurodegenerative conditions such as Alzheimer's disease (AD), and is postulated to promote microtubule disassembly. Lithium has been reported to reduce the phosphorylation of tau, enhance the binding of tau to microtubules, and promote microtubule assembly through direct and reversible inhibition of glycogen synthase kinase-3 (Hong M., Chen D.C., Klein P.S. and Lee V.M., J. Biol. Chem., (272), 25326- 32, (1997). β-catenin is phosphorylated by GSK-3 as part of a tripartite complex with axin, resulting in β-catenin being targetted for degradation (D eda et al, J. EMBO., (17), 1371-1384, (1998)). Inhibition of GSK-3 activity is a key mechanism by which cytosolic levels of catenin are stabilised and hence promote β-catenin-LEF-1/TCF transcnptional activity (Eastman, Grosschedl, Curr. Opin. Cell. Biol, (11), 233, (1999)). Rapid onset AD mutations in presenilin-1 (PS-1) have been shown to decrease the cytosolic β-catenin pool in transgenic mice. Further evidence suggests that such a reduction in available β- catenin may increase neuronal sensitivity to amyloid mediated death through inhibition of β-catenin-LEF-1/TCF transcriptional regulation of neuroprotective genes (Zhang et al, Nature, (395), 698-702, (1998)). A likely mechanism is suggested by the finding that mutant PS-1 protein confers decreased inactivation of GSK-3 compared with normal PS- 1 (Weihl C.C., Ghadge G.D., Kennedy S.G., Hay Ν, Miller RJ. and Roos R.P., J. Neurosci., (19), 5360-5369, (1999)). International Patent Application Publication Number WO 97/41854 (University of
Pennsylvania) discloses that an effective drug for the treatment of manic depression is lithium, but that there are serious drawbacks associated with this treatment. Whilst the precise mechanism of action of this drug for treatment of manic depression remains to be fully defined, current models suggest that inhibition of GSK-3 is a relevant target that contributes to the modulation of AP-1 DNA binding activity observed with this compound (see Manji et al, J. Clin. Psychiatry, (60) (suppl 2), 27-39, (1999) for review). GSK-3 inhibitors may also be of value in treatment of schizophrenia. Reduced levels of β-catenin have been reported in schizophremc patients (Cotter D., Kerwin R., al- Sarraji S., Brion J.P., Chadwich A., Lovestone S., Anderton B., and Everall I., Neuroreport, (9), 1379-1383, (1998)) and defects inpre-pulse inhibition to startle response have been observed in schizophremc patients (Swerdlow et al, Arch. Gen. Psychiat., (51), 139-154, (1994)). Mice lacking the adaptor protein dishevelled- 1, an essential mediator of Wnt-induced inhibition of GSK-3, exhibit both a behavioural disorder and defects in pre-pulse inhibition to startle response (Lijam N., Paylor R., McDonald M.P., Crawley J.N., Deng C.X., Herrup K, Stevens K.E., Maccaferri G., McBain C.J., SussmanDJ., and Wynshaw-Boris A., Cell, (90), 895-905, (1997)). Together, these findings implicate deregulation of GSK-3 activity as contributing to schizophrenia. Hence, small molecule inhibitors of GSK-3 catalytic activity maybe effective in treatment of this mood disorder.
The finding that transient β-catenin stabilisation may play a role in hair development (Gat et al, Cell, (95), 605-614, (1998)) suggests that GSK-3 inhibitors could be used in the treatment of baldness.
Studies on fibroblasts from the GSK-3β knockout mouse (Hoeflich K.P. et al, Nature, (406), 86-90, (2000)) support a role for this kinase in positively regulating the activity of NFkB. This transcription factor mediates cellular responses to a number of inflammatory stimuli. Therefore, pharmacologic inhibition of GSK-3 maybe of use in treating inflammatory disorders through the negative regulation of NFkB activity.
The compounds of the present invention are indazole derivatives. Other indazole derivatives have been described previously for use in alternative medicinal applications. For example, International Application, Publication Number WO 93/23404 describes a series of bicyclic compounds, including substituted indazoles, which are stated to modulate endothelin activity and may accordingly be of use in the treatment of conditions such as asthma, hypertension, renal failure and endotoxin shock. International Application, Publication Number WO 94/14780 describes a series of heterocyclic compounds, including indazoles, which are stated to have biological activity as neuronal nitric oxide synthase inhibitors, and as such may be useful in the treatment of, for example, cerebral ischaemia.
We have now discovered that a series of indazoles are potent and selective inhibitors of GSK-3. These compounds are indicated to be useful for the treatment and/or prophylaxis of conditions associated with a need for inhibition of GSK-3, such as diabetes, conditions associated with diabetes, chronic neurodegenerative conditions including dementias such as Alzheimer's disease, Parkinson's disease, progressive supranuclear palsy, subacute sclerosing panencephalitic parkinsonism, postencephalitic parkinsonism, pugilistic encephalitis, guam parkinsonism-dementia complex, Pick's disease, corticobasal degeneration, frontotemporal dementia, Huntingdon's disease, ADDS associated dementia, amyotrophic lateral sclerosis, multiple sclerosis and neurotraumatic diseases such as acute stroke, mood disorders such as schizophrenia and bipolar disorders, promotion of functional recovery post stroke, cerebral bleeding (for example, due to solitary cerebral amyloid angiopathy), hair loss, obesity, atherosclerotic cardiovascular disease, hypertension, polycystic ovary syndrome, syndrome X, ischaemia, traumatic brain injury, cancer, leukopenia, Down's syndrome, Lewy body disease, inflammation, and immunodeficiency.
Accordingly, in a first aspect, the present invention provides a compound of formula ®,
Figure imgf000006_0001
or a salt thereof, or a solvate thereof, wherein,
Rl is unsubstituted or substituted alkyl, unsubstituted or substituted cycloalkyl, unsubstituted or substituted alkenyl, unsubstituted or substituted cycloalkenyl, unsubstituted or substituted aryl, aralkyl wherein the aryl and the alkyl moieties may each independently be unsubstituted or substituted, aralkenyl wherein the aryl and alkenyl moieties may each independently be unsubstituted or substituted, unsubstituted or substituted heterocyclyl, or heterocyclylalkyl wherein the heterocyclyl and the alkyl moieties may each independently be unsubstituted or substituted, unsubstituted or substituted heteroaryl, heteroarylalkyl wherem the heteroaryl and the alkyl moieties may each independently be unsubstituted or substituted;
R2 is H;
R3 is H, halo, alkyl, unsubstituted or substituted aryl or unsubstituted or substituted heteroaryl;
R4 is H, unsubstituted or substituted aryl; unsubstituted or substituted heteroaryl, unsubstituted or substituted heterocyclyl; or -X-aryl;
R5 is H;
X is O, NH, S or S(O)n and n is 1 or 2; with the proviso that when R1, R2 and R^ are as defined in relation to formula (I), and R^ is H or halo, then R4 is unsubstituted or substituted aryl; unsubstituted or substituted heteroaryl, unsubstituted or substituted heterocyclyl; or -X-aryl.
Suitably, R* is alkyl, alkenyl, cycloC3_g alkyl, cycloC3_g alkenyl, di- alkylaminoalkyl, arylalkyl, arylalkenyl, heterocyclyl wherein the heterocyclyl group may be optionally substituted by one or more groups selected from alkyl, arylalkyl and alkoxyalkyl; heterocyclylalkyl wherein the heterocyclyl group may be optionally substituted by one or more groups selected from alkoxyalkyl, aryloxyalkyl, arylalkyl and alkyl; heteroarylalkyl wherein the heteroaryl may be optionally substituted by one or more groups selected from alkyl; heteroaryl wherein the heteroaryl may be optionally substituted by one or more groups selected from aryl and heteroaryl; aryl wherein the aryl group may be optionally substituted by heterocyclylalkyl and di-alkylaminoalkyl; or alkoxyalkyl wherein the alkoxy group may be optionally substituted by alkoxy. More suitably, R is alkyl, cycloC3_g alkyl or heterocyclyl wherein the heterocyclyl group is substituted by arylalkyl. Preferably, Rl is n-propyl, cyclopropyl, cyclopentyl and N- benzylpyrrolidin-3-yl. Suitably, R^ is H, halo or aryl. Preferably R^ is H, bromo or phenyl. Suitably, R4 is H, aryl wherein the aryl group may be optionally substituted by one or more substituents selected from halo, -SO2NH2, -OH, -NHSO2Me and -Sθ2Me; heteroaryl wherein the heteroaryl group may be optionally substituted by one or more substituents selected from halo, alkyl and oxo; heterocyclyl wherein the heterocyclyl group may be optionally substituted by benzyloxycarbonyl; or -X-aryl. Preferably, R4 is H, phenyl, 2-fluorophenyl, 3-fluorophenyl, 4-fluorophenyl, 2,3-difluorophenyl, 2,5- difluorophenyl, 3,5-difluoroρhenyl, 3-H2NSO2-Ph, 4-H2NSO2-PI1, 3-MeSO2NH-Ph, 4-
MeSO2NH-Ph, 4-methanesulfonylphenyl, 4-hydroxyphenyl, 2-furyl, 3-furyl, 2-pyrrolyl,
2-thienyl, 3-thienyl, 4-methylthien-2-yl, 5-bromotbien-2-yl, indol-5-yl, pyrid-2-on-5-yl, - NHPh, -OPh, -SPh, -SO2Ph, piperazin-1-yl or 4-benzyloxycarbonylpiperazin-l-yl. In a preferred aspect of the present invention there is provided a subset of compounds of formula (I), of formula (IA),
Figure imgf000008_0001
(IA)
or a salt thereof, or a solvate thereof, wherein,
Rl is alkyl, cyck>C3_g alkyl or heterocyclyl wherein the heterocyclyl group is substituted by arylalkyl;
R2 is H; R3 is H, halo or aryl; and
R4 is H, aryl wherein the aryl group may be optionally substituted by one or more substituents selected from halo, -SO2NH2, -OH, -NHSO2Me and -SO2Me; heteroaryl wherein the heteroaryl group may be optionally substituted by one or more substituents selected from halo, alkyl and oxo; heterocyclyl wherein the heterocyclyl group may be optionally substituted by aryloxycarbonyl; or -X-aryl; with the proviso that when R , R2 and R^ are as defined in relation to formula (IA), and
R3 is H or halo, then R4 is aryl wherein the aryl group may be optionally substituted by one or more substituents selected from halo, -SO2NH2, -OH, -NHSO2Me and -SO2Me; heteroaryl wherein the heteroaryl group may be optionally substituted by one or more substituents selected from halo, alkyl and oxo; heterocyclyl wherein the heterocyclyl group may be optionally substituted by aryloxycarbonyl; or -X-aryl.
In a further preferred aspect of the present invention there is provided a subset of compounds of formula (I), of formula (IB),
Figure imgf000009_0001
(IB)
or a salt thereof, or a solvate thereof, wherein,
Rl is n-propyl, cyclopropyl, cyclopentyl or N-benzylρyrrolidin-3-yl;
R2 is H; R3 is H, bromo or phenyl;
R4 is H, phenyl, 2-fluorophenyl, 3 -fluorophenyl, 4-fluorophenyl, 2,3-difluorophenyl, 2,5- difluorophenyl, 3,5-difluorophenyl, 3-H2NSO2-Ph, 4-H2NSO2-Ph, 3-MeSO2NH-Ph, 4-
MeSO2 H-Ph, 4-methanesulfonylphenyl, 4-hydroxyphenyl, 2-furyl, 3-furyl, 2-pyrrolyl,
2-thienyl, 3-thienyl, 4-methylthien-2-yl, 5-bromothien-2-yl, indol-5-yl, pyrid-2-on-5-yl, - NHPh, -OPh, -SPh, -SO2PI1, piperazin-1-yl or 4-benzyloxycarbonylpiperazin-l-yl; and
R5 is H; with the proviso that when R , R2 and R^ are as defined in relation to formula (IB), and
R3 is H or bromo, then R4 is phenyl, 2-fluorophenyl, 3-fluorophenyl, 4-fluorophenyl, 2,3-difluorophenyl, 2,5-difluorophenyl, 3,5-difluorophenyl, 3-H2NSO2-Ph, 4-H2NSO2- Ph, 3-MeSO2NH-Ph, 4-MeSθ2NH-Ph, 4-methanesulfonylphenyl, 4-hydroxyphenyl, 2- furyl, 3-furyl, 2-pyrrolyl, 2-thienyl, 3-thienyl, 4-methylthien-2-yl, 5-bromothien-2-yl, indol-5-yl, pyrid-2-on-5-yl, -NHPh, -OPh, -SPh, -SO2PI1, piperazin-1-yl or 4- benzyloxycarbonylpiperazin-1-yl.
Particularly preferred compounds of formula (I) which are of special interest as agents useful in the treatment and/or prophylaxis of conditions associated with a need for inhibition of GSK-3 are provided in Table 1 below.
Certain compounds of formula (I) may contain chiral atoms and/or multiple bonds, and hence may exist in one or more stereoisomeric forms. The present invention encompasses all of the isomeric forms of the compounds of formula (I) whether as individual isomers or as mixtures of isomers, including geometric isomers and racemic modifications.
As used herein the term "alkyl" as a group or part of a group refers to a straight or branched chain saturated aliphatic hydrocarbon radical containing 1 to 12 carbon atoms, suitably 1 to 6 carbon atoms. Such alkyl groups in particular include methyl ("Me"), ethyl ("Et"), n-propyl ("Prn"), ώo-propyl ("Pr1"), n-butyl ("Bun"), sec-butyl ("Bus"), tert- butyl ("Bu*"), pentyl and hexyl. Where appropriate, such alkyl groups may be substituted by one or more groups selected from halo (such as fluoro, chloro, bromo), -CN, -CF3, -
OH, -OCF3, C2-6 alkenyl, C^s alkynyl, C\. alkoxy, aryl and di-Cι_6 alkylamino.
As used herein the term "alkenyl" as a group or part of a group refers to a straight or branched chain mono- or poly-unsaturated aliphatic hydrocarbon radical containing 2 to 12 carbon atoms, suitably 2 to 6 carbon atoms. References to "alkenyl" groups include groups which may be in the E- or Z-form or mixtures thereof. Such alkenyl groups in particular include ethenyl, propenyl, butenyl, pentenyl and hexenyl. Where appropriate, such alkenyl groups may be substituted by one or more groups selected from halo (such as fluoro, chloro, bromo), -CN, -CF3, -OH, -OCF3, Cι _6 alkyl, C3_6 alkynyl, C . alkoxy, aryl and di-Cι_6 alkylamino.
As used herein the term "alkynyl" refers to hydrocarbon groups of either straight or branched configuration with one or more carbon-carbon triple bonds which may occur at any stable point in the chain, containing 3 to 12 carbon atoms, suitably 3 to 6 carbon atoms. Such alkynyl groups in particular include propynyl, butynyl and pentynyl. Where appropriate, such alkynyl groups may be substituted by one or more groups selected from halo (such as fluoro, chloro, bromo), -CN, -CF3, -OH, -OCF3, C\^ alkyl, C2-6 alkenyl,
C\. alkoxy, aryl and di-Cι_6 alkylamino.
As used herein, the term "alkoxy" as a group or part of a group refers to an alkyl ether radical, wherein the term "alkyl" is defined above. Such alkoxy groups in particular include methoxy, ethoxy, n-propoxy, tso-propoxy, n-butoxy, zso-butoxy, sec-butoxy and tert-butoxy. Where appropriate, such alkoxy groups may be substituted by one or more groups selected from halo (such as fluoro, chloro, bromo), -CN, -CF3, -OH, -OCF3, Cj.g alkyl, C2_6 alkenyl, C2- alkynyl, aryl and di-Ci^g alkylamino.
As used herein, the term "aryl" as a group or part of a group refers to a carbocyclic aromatic radical. Suitably such aryl groups are 5-6 membered monocyclic groups or 8-10 membered fused bicyclic groups, especially phenyl ("Ph"), biphenyl and naphthyl, particularly phenyl. Such aryl groups may be optionally substituted with one or more substituents, which maybe the same or different, selected from halo (such as fluoro, chloro, bromo), -CN, -CF3, -OH, -OCF3, -NO2, C\- alkyl, C2-6 alkenyl, 0,^-6 alkynyl, C j _g alkoxy and di-C \ _g alkylamino.
As used herein, the term "heteroaryl" as a group or part of a group refers to stable heterocyclic aromatic single and fused rings containing one or more hetero atoms independently selected from nitrogen, oxygen and sulfur. A fused heteroaryl ring system may include carbocyclic rings and need include only one heteroaryl ring. Such heteroaryl groups include furyl, thienyl, pyridazinyl, pyridyl, quinolinyl, indolyl, thiazolyl, benzoxazolyl, and benzothiazolyl. Each ring may be optionally substituted with one or more substituents, which maybe the same or different, selected from halo (such as fluoro, chloro, bromo), -CN, -CF3, -OH, -NO2, -OCF3, Cχ_6 alkyl, C2-6 alkenyl, C3_6 alkynyl, Cι_6 alkoxy, aryl, heteroaryl, and di-C 1 -6 alkylamino. As used herein, the terms "heterocyclyl" and "heterocyclic" as a group or part of a group refer to stable heterocyclic non-aromatic single and fused rings containing one or more hetero atoms independently selected from nitrogen, oxygen and sulfur. A fused heterocyclyl ring system may include carbocyclic rings and need include only one heterocyclic ring. Such heterocyclyl groups include piperazinyl, piperidinyl and n orpholinyl. Each ring may be optionally substituted with one or more substituents, which may be the same or different, selected from halo (such as fluoro, chloro, bromo), - CN, -CF3, -OH, -NO2, -OCF3, Cι _6 alkyl, C2_6 alkenyl, C3_6 alkynyl, C _6 alkoxy, aryl, heteroaryl, and di-Cι_6 alkylamino.
As used herein the terms "halo" include iodo, bromo, chloro or fluoro, suitably bromo, chloro and fluoro, especially bromo and chloro. Composite terms such as "alkoxyalkyl" and "arylalkyl" refer to substituents comprising two interlinked groups, with the group named latterly in the term being the linking group, so that "alkoxyalkyl" means -(alkyl)-(alkoxy) whilst "arylalkyl" means - (alkyl)-(aryl).
The compounds of formula (I) or their salts or solvates are preferably in pharmaceutically acceptable or substantially pure form. By pharmaceutically acceptable form is meant, inter alia, having a pharmaceutically acceptable level of purity excluding normal pharmaceutical additives such as diluents and carriers, and including no material considered toxic at normal dosage levels.
A substantially pure form will generally contain at least 50% (excluding normal pharmaceutical additives), preferably 75%, more preferably 90% and still more preferably 95% of the compound of formula (I) or its salt or solvate.
One preferred pharmaceutically acceptable form is the crystalline form, including such form in pharmaceutical composition. In the case of salts and solvates the additional ionic and solvent moieties must also be non-toxic. Suitable salts are pharmaceutically acceptable salts.
Suitable pharmaceutically acceptable salts include the acid addition salts with the conventional pharmaceutical acids, for example maleic, hydrochloric, hydrobromic, phosphoric, acetic, fumaric, salicylic, citric, lactic, mandelic, tartaric, succinic, benzoic, ascorbic and methanesulfonic. Suitable pharmaceutically acceptable salts include salts of acidic moieties of the compounds of formula (I) when they are present, for example salts of carboxy groups or phenolic hydroxy groups.
Suitable salts of acidic moieties include metal salts, such as for example aluminium, alkali metal salts such as lithium, sodium or potassium, alkaline earth metal salts such as calcium or magnesium and ammonium or substituted ammonium salts, for example those with lower alkylamines such as triethylamine, hydroxyalkylamines such as 2-hydroxyethylamine, bis-(2-hy<koxyethyl)-amine or tri-(2-hydroxyethyl)-amine, cycloalkylamines such as bicyclohexylamine, or with procaine, dibenzylpiperidine, N-benzyl-β-phenethylamine, dehydroabietylamine, N,N'-bisdehydroabietylamine, glucamine, N-methylglucamine or bases of the pyridine type such as pyridine, collidine, quinine or quinoline. Suitable solvates are pharmaceutically acceptable solvates.
Suitable pharmaceutically acceptable solvates include hydrates. For the avoidance of doubt when used herein the term "diabetes" includes diabetes mellitus, especially Type 2 diabetes, and conditions associated with diabetes mellitus. The term "conditions associated with diabetes" includes those conditions associated with the pre-diabetic state, conditions associated with diabetes mellitus itself and complications associated with diabetes mellitus.
The term "conditions associated with the pre-diabetic state" includes conditions such as insulin resistance, impaired glucose tolerance and hyperinsulinaemia. The term "conditions associated with diabetes mellitus itself includes hyperglycaemia, insulin resistance and obesity. Further conditions associated with diabetes mellitus itself include hypertension and cardiovascular disease, especially atherosclerosis and conditions associated with insulin resistance. Conditions associated with insulin resistance include polycystic ovarian syndrome and steroid induced insulin resistance. The term "complications associated with diabetes mellitus" includes renal disease, especially renal disease associated with Type II diabetes, neuropathy and retinopathy. Renal diseases associated with Type II diabetes include nephropathy, glomerulonephritis, glomerular sclerosis, nephrotic syndrome, hypertensive nephrosclerosis and end stage renal disease. The term "neurotraumatic diseases" includes both open or penetrating head trauma, such as caused by surgery, or a closed head trauma injury, such as caused by an injury to the head region, ischaemic stroke including acute stroke, particularly to the brain area, transient ischaemic attacks following coronary by-pass and cognitive decline following other transient ischaemic conditions. According to a further aspect of the present invention there is provided a process for the preparation of a compound of formula (I), or a salt and/or solvate thereof, which process comprises reacting a compound of formula (II),
Figure imgf000014_0001
wherein R2, R3, R4 nd R^ are as defined in relation to formula (I) with a compound of formula (HI),
Figure imgf000014_0002
wherein R is as defined in relation to formula (I) and X is a suitable leaving group and thereafter, if required, carrying out one or more of the following optional steps: (i) converting a compound of formula (I) to a further compound of formula (I); (ii) removing any necessary protecting group;
(iii) preparing an appropriate derivative of the compound so formed.
Suitably X is chloro. It will be appreciated that compounds of formula (III) may also include related carboxylic acid anhydrides.
The reaction between the compounds of formulae (II) and (HI) is carried out in a suitable solvent, under conventional conditions, at a suitable temperature, providing a suitable rate of formation of the required product, over a suitable reaction time. A suitable solvent is pyridine. Suitable reaction temperatures include those in the range of 20°C to 220°C and, as appropriate, the reflux temperature of the solvent. Suitable reaction times are those in the range 12 to 72 hours. The reaction products are isolated using conventional methods. Conventional methods of heating and cooling may be employed, for example thermostatically controlled oil baths and ice/salt baths respectively. The reaction products are typically purified by conventional methods, such as crystallisation, chromatography and trituration. Crystalline product may be obtained by standard methods. In a preferred aspect, a compound of formula (III), such as n-butyric anhydride, is added to a solution of a compound of formula (II) in pyridine. The reaction mixture is heated under reflux for 22 hours and allowed to cool to ambient temperature. The resulting solution is concentrated in vacuo and purified by column chromatography using one or more suitable solvents, such as 10% v/v methanol in chloroform, to afford the desired compound of formula (I). In a further preferred aspect, to a stirred solution of a compound of formula (II) in dry pyridine under argon is added a compound of formula (IH), such as cyclopropanecarbonyl chloride. The reaction mixture is stirred for 12 hours at room temperature. The resulting solution is concentrated in vacuo, and the crude oil is dissolved in ethyl acetate and washed with saturated sodium bicarbonate solution. The organic extract is dried over magnesium sulfate and further concentrated in vacuo. The resulting solid is triturated with a suitable solvent, such as dichloromethane, to afford the desired compound of formula (I).
It will be appreciated that treatment of a compound of formula (If) with a compound of formula (in), according to the above-mentioned process, may lead to the formation of a bw-acylated intermediate species of formula (IN),
Figure imgf000015_0001
wherein R2, R^, R4 and R^ are as defined in relation to formula (I). In a further aspect of the present invention, there is provided a process for the preparation of a compound of formula (I), or a salt and/or solvate thereof, which process comprises reacting a compound of formula (IV) with a nucleophile and thereafter, if required, carrying out one or more of the following optional steps: (i) converting a compound of formula (I) to a further compound of formula (I); (ii) removing any necessary protecting group;
(iii) preparing an appropriate derivative of the compound so formed. The reaction between the compound of formula (TV) and a nucleophile is carried out optionally in a suitable solvent, under conventional conditions, at a suitable temperature, providing a suitable rate of formation of the required product, over a suitable reaction time. Suitably the reaction is performed using the nucleophile as a solvent. A suitable nucleophile is an amine, such as a primary or secondary amine. Suitable reaction temperatures include those in the range of 20°C to 100°C and, as appropriate, the reflux temperature of the solvent. Suitable reaction times are those in the range 1 to 48 hours. The reaction products are isolated using conventional methods. Conventional methods of heating and cooling may be employed, for example thermostatically controlled oil baths and ice/salt baths respectively. The reaction products are typically purified by conventional methods, such as crystallisation, chromatography and trituration. Crystalline product may be obtained by standard methods.
In a preferred aspect, a suitable nucleophile, such as piperidine, and water are added to a compound of formula (IV) under argon. The reaction mixture is stirred for 3 hours at ambient temperature. The resulting mixture is concentrated in vacuo and the crude product purified by preparative HPLC using one or more suitable solvents, such as 10-90% acetonitrile/0.1 % trifluoroacetic acid in water/0.1 % trifluoroacetic acid. The appropriate fractions are concentrated to dryness to afford the desired compound of formula (I). The above-mentioned conversions of a compound of formula (I) into another compound of formula (I) include any conversion, which maybe effected using conventional procedures, but in particular the said conversions include any combination of:
(i) converting one group R into another group R ; (ii) converting one group R^ into another group R^; and
(iii) converting one group R4 into another group R4.
The above-mentioned conversions (i), (ii) and (iii) may be performed using any appropriate method under conditions determined by the particular groups chosen.
Suitable conversions of one group R^ into another group R^, as in conversion (iii) above, include:
(a) converting a group R3 which represents halo, such as bromo, into another group R3 which represents aryl, such as phenyl. Such a conversion may be performed using an appropriate arylation procedure, for example, by treating a compound of formula
(I) wherein R^ is halo, such as bromo, with an arylboronic acid or an arylboronate, such as phenylboronic acid.
Suitable conversions of one group R4 into another group R4, as in conversion (iv) above, include:
(b) converting a group R4 which represents -X-aryl, such as -SPh, into another group R4 which represents -X-aryl, such as -SO2PI1. Such a conversion may be performed using an appropriate oxidation procedure, for example, by treating a compound of formula (I) wherein R^ is -X-aryl, such as -SPh, with an appropriate oxidising agent such as wetβ-chloroperbenzoic acid.
It will be appreciated that the synthesis of compounds of formula (I) may involve the use of conventional protecting groups such as tert-butoxycarbonyl ("Boc"). Such protection/de-protection procedures may be performed using any appropriate method under conditions determined by the particular groups chosen. For example, a compound of formula (N),
Figure imgf000017_0001
wherein Rl, R2, R^ and R^ are as defined in relation to formula (I), may be converted to a compound of formula (I) where R4 is -ΝHAr by reaction with a suitable acid, such as hydrochloric acid in 1,4-dioxane. Compounds of formula (II) may be prepared by reaction of a compound of formula (VI),
Figure imgf000018_0001
wherein,
R2, R3, R4 and R^ are as defined in relation to formula (I), with hydrazine or a hydrate thereof. The reaction between the compound of formula (VI) and hydrazine or a hydrate thereof, is carried out in a suitable solvent at a suitable temperature, generally an elevated temperature, providing a suitable rate of formation of the required product, over a suitable reaction time. Suitable solvents include pyridine and ethanol. Suitable reaction temperatures include those in the range of 60 °C to 220 °C and, as appropriate, the reflux temperature of the solvent. Suitable reaction times are those in the range 1-72 hours. The reaction products are isolated using conventional methods. Typically, the reaction mixture is cooled, the product isolated by filtration, and dried. Conventional methods of heating and cooling may be employed, for example thermostatically controlled oil baths and ice/salt baths respectively. The reaction products may, if desired, be purified by conventional methods, such as crystallisation, chromatography and trituration.
In a preferred aspect, hydrazine hydrate is added to a stirred solution of the compound of formula (VI) in pyridine. The reaction mixture is stirred under reflux for 72 hours and cooled. The resulting mixture is concentrated in vacuo, and the residue dissolved in ethyl acetate and washed with aqueous sodium bicarbonate solution. The organic extract is dried over magnesium sulfate and concentrated in vacuo. The crude product is triturated with a suitable solvent, such as dichloromethane to afford the desired compound of formula (II).
Certain compounds of formula (fl) are believed to be novel and accordingly form a further aspect of the present invention. As mentioned hereinbefore, compounds of formula (IV) may be prepared by reaction of a compound of formula (II) with a compound of formula (in), in the presence of a suitable solvent such as pyridine. Compounds of formula (V) may be prepared by reaction of a compound of formula (VII),
Figure imgf000019_0001
wherein, R2, R3, R4 and R^ are as defined in relation to formula (I), with a compound of formula (HI). The synthesis of compounds of formula (V) by reaction of a compound of formula (NH) with a compound of formula (Hi) may be undertaken using analogous acylation conditions to those used for the preparation of compounds of formula (I) as described hereinbefore.
Compounds of formula (VI) may be prepared by reaction of a compound of formula (NTH),
Figure imgf000019_0002
wherein R2, R3 and R^ are as defined in relation to formula (I), with a compound of formula (LX),
Figure imgf000019_0003
wherein, 4 is as defined in relation to formula (I) and R^ is H or alkyl, in the presence of a suitable catalyst and a suitable base. The reaction between the compound of formula (VTH) and a compound of formula (IX) is carried out in a suitable solvent under an inert atmosphere in the presence of a suitable catalyst and a suitable base, at a suitable temperature, generally an elevated temperature, providing a suitable rate of formation of the required product, over a suitable reaction time. Suitable solvents include a mixture of dimethylformamide, ethanol and water. Suitable catalysts are tetrakis(triphenylphosρhine)palladium(0) and PdCl2(dρpf).
A suitable base is KOAc. Suitable reaction temperatures include those in the range of 60 °C to 220 °C and, as appropriate, the reflux temperature of the solvent. Suitable reaction times are those in the range 1-72 hours. The reaction products are isolated using conventional methods. Typically, the reaction mixture is cooled, the product isolated by filtration, and dried. Conventional methods of heating and cooling may be employed, for example thermostatically controlled oil baths and ice/salt baths respectively. The reaction products may, if desired, be purified by conventional methods, such as crystallisation, chromatography and trituration. In a preferred aspect, to a stirred solution of a compound of formula (VIH), a compound of formula (IX), and potassium acetate in dimethylformamide, ethanol and water is added PdCl2(dppf). The resulting mixture is stirred at 100 °C for 4 hours, allowed to cool and is filtered through celite. The filtrate is added to ethyl acetate and washed with brine. The organic extract is separated, dried with magnesium sulfate and concentrated in vacuo. The crude solid is purified by chromatography using one or more suitable solvents, such as 10% diethyl ether/60-80 petrol, to afford the desired compound of formula (I).
Compounds of formula (NT) may also be prepared by reaction of a compound of formula (X),
Figure imgf000020_0001
wherein, R2, R^ and R$ are as defined in relation to formula (I), with a compound of formula (XI), R— Y (XI)
wherein, R4 is as defined in relation to formula (I) and Y is halo, such as bromo, in the presence of a suitable catalyst.
The reaction between the compound of formula (X) and a compound of formula (XI) is carried out in a suitable solvent in the presence of a suitable catalyst and a suitable base, at a suitable temperature, generally an elevated temperature, providing a suitable rate of formation of the required product, over a suitable reaction time. Suitable solvents include a mixture of dimethylformamide, ethanol and water. A suitable catalyst is PdCl2(dppf). A suitable base is KOAc. Suitable reaction temperatures include those in the range of 60 °C to 220 °C and, as appropriate, the reflux temperature of the solvent. Suitable reaction times are those in the range 1-72 hours. The reaction products are isolated using conventional methods. Typically, the reaction mixture is cooled, the product isolated by filtration, and dried. Conventional methods of heating and cooling may be employed, for example thermostatically controlled oil baths and ice/salt baths respectively. The reaction products may, if desired, be purified by conventional methods, such as crystallisation, chromatography and trituration.
Compounds of formula (VI) wherein R4 is -X-aryl, where X is S, may be prepared by reaction of a compound of formula (XII),
Figure imgf000021_0001
wherein, R2, R3 and R^ are as defined in relation to formula (I), with a compound of formula (XTfl),
Ar— SH (XIH) wherein Ar is aryl as defined in relation to formula (I), in the presence of a suitable base. The reaction between the compound of formula (XII) and a compound of formula (Xπi) is carried out in a suitable solvent in the presence of a suitable base, at a suitable temperature, providing a suitable rate of formation of the required product, over a suitable reaction time. A suitable solvent is dimethylformamide. A suitable base is sodium hydride. Suitable reaction temperatures include those in the range of 20 °C to 100 °C. Suitable reaction times are those in the range 1-48 hours. The reaction products are isolated using conventional methods. Typically, the reaction mixture is cooled, the product isolated by filtration, and dried. Conventional methods of heating and cooling may be employed, for example thermostatically controlled oil baths and ice/salt baths respectively. The reaction products may, if desired, be purified by conventional methods, such as crystallisation, chromatography and trituration.
In a preferred aspect, a compound of formula (XIH) is added to a compound of formula (XII) in the presence of sodium hydride in dimethylformamide. The reaction mixture is stirred at ambient temperature, under argon for 16 hours. The resulting solution is concentrated to dryness in vacuo and purified by preparative HPLC using one or more suitable solvents, such as 10-90% acetonitrile (containing 0.1% trifluoroacetic acid) in water (containing 0.1% trifluoroacetic acid). The appropriate fractions are concentrated to dryness to afford the desired compound of formula (VI).
Compounds of formula (VH) may be prepared by analogous methods to those mentioned hereinbefore for the preparation of compounds of formula (II).
Compounds of formula (X) may be prepared by reaction of a compound of formula (Vπi) with bis(pinacolato)diboron in the presence of a suitable catalyst, such as l,r-Bis(diphenylphosphino)ferrocenedichloropalladium(II), i.e. PdCl2(dppf).
Compounds of formula (HI), (VIH), (LX), (XI), (XH) and (XIH) are either commercially available or are prepared by analogy with known conventional procedures such as those in standard reference texts of synthetic methodology, for example, J. March, Advanced Organic Chemistry, 4th Edition, 1992, Wiley Interscience. Compounds of formula (IV), (V), (VII) and (X) are believed to be novel and accordingly form a further aspect of the present invention. Compounds of formulae (I), (II), (V) and (VII) may exist as tautomers. The present invention encompasses all tautomeric forms of the compounds of (I), (II), (V) and (VII).
As stated above, the compounds of formula (I), or pharmaceutically acceptable salts or solvates thereof, are indicated to be useful as inhibitors of glycogen synthase kinase-3.
The invention therefore provides a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, for use as an inhibitor of GSK-3. Accordingly, the present invention also provides a method for the treatment of conditions associated with a need for inhibition of GSK-3 such as diabetes, conditions associated with diabetes, chronic neurodegenerative conditions including dementias such as Alzheimer's disease, Parkinson's disease, progressive supranuclear palsy, subacute sclerosing panencephalitic parkinsonism, postencephalitic parkinsonism, pugilistic encephalitis, guam parkinsonism-dementia complex, Pick's disease, corticobasal degeneration, frontotemporal dementia, Huntingdon's disease, AIDS associated dementia, amyotrophic lateral sclerosis, multiple sclerosis and neurotraurnatic diseases such as acute stroke, mood disorders such as schizophrenia and bipolar disorders, promotion of functional recovery post stroke, cerebral bleeding (for example, due to solitary cerebral amyloid angiopathy), hair loss, obesity, atherosclerotic cardiovascular disease, hypertension, polycystic ovary syndrome, syndrome X, ischaemia, traumatic brain injury, cancer, leukopenia, Down's syndrome, Lewy body disease, inflammation, and immunodeficiency, which method comprises the administration of a pharmaceutically effective, non-toxic amount of a compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof. The present invention further provides a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, for use as an inhibitor of glycogen synthase kinase-3, and especially for use in the treatment of conditions associated with a need for the inhibition of GSK-3, such as diabetes, conditions associated with diabetes, chronic neurodegenerative conditions including dementias such as Alzheimer's disease, Parkinson's disease, progressive supranuclear palsy, subacute sclerosing panencephalitic parkinsonism, postencephalitic parkinsonism, pugilistic encephalitis, guam parkinsonism- dementia complex, Pick's disease, corticobasal degeneration, frontotemporal dementia, Huntingdon's disease, ADDS associated dementia, amyotrophic lateral sclerosis, multiple sclerosis and neurotraurnatic diseases such as acute stroke, mood disorders such as schizophrenia and bipolar disorders, promotion of functional recovery post stroke, cerebral bleeding (for example, due to solitary cerebral amyloid angiopathy), hair loss, obesity, atherosclerotic cardiovascular disease, hypertension, polycystic ovary syndrome, syndrome X, ischaemia, traumatic brain injury, cancer, leukopenia, Down's syndrome, Lewy body disease, inflammation, and immunodeficiency.
The present invention also provides the use of a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, for the manufacture of a medicament for the treatment of conditions associated with a need for the inhibition of GSK-3, such as diabetes, conditions associated with diabetes, chronic neurodegenerative conditions including dementias such as Alzheimer's disease, Parkinson's disease, progressive supranuclear palsy, subacute sclerosing panencephalitic parkinsonism, postencephalitic parkinsonism, pugilistic encephalitis, guam parkinsonism-dementia complex, Pick's disease, corticobasal degeneration, frontotemporal dementia, Huntingdon's disease, ADDS associated dementia, amyotrophic lateral sclerosis, multiple sclerosis and neurotraurnatic diseases such as acute stroke, mood disorders such as schizophrenia and bipolar disorders, promotion of functional recovery post stroke, cerebral bleeding (for example, due to solitary cerebral amyloid angiopathy), hair loss, obesity, atherosclerotic cardiovascular disease, hypertension, polycystic ovary syndrome, syndrome X, ischaemia, traumatic brain injury, cancer, leukopenia, Down's syndrome, Lewy body disease, inflammation, and immunodeficiency.
In a further aspect of this invention, there is provided a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, for use as an active therapeutic substance.
Preferably, the compounds of formula (I), or pharmaceutically acceptable salts or solvates thereof, are administered as pharmaceutically acceptable compositions.
Accordingly, the invention also provides a pharmaceutical composition which comprises a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable carrier. The active compounds are usually administered as the sole medicament agent but they may be administered in combination with other medicament agents as dictated by the severity and type of disease being treated.
The said combination comprises co-administration of a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, and an additional medicament agent or the sequential administration of a compound of formula (I), or a pharmaceutically acceptable derivative thereof, and the additional medicament agent.
Co-administration includes administration of a pharmaceutical composition which contains both a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, and the additional medicament agent or the essentially simultaneous administration of separate pharmaceutical compositions of a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, and the additional medicament agent.
The compositions of the invention are preferably adapted for oral administration. However, they may be adapted for other modes of administration. The compositions may be in the form of tablets, capsules, powders, granules, lozenges, suppositories, reconstitutable powders, or liquid preparations, such as oral or sterile parenteral solutions or suspensions. In order to obtain consistency of administration it is preferred that a composition of the invention is in the form of a unit dose. Preferably the composition are in unit dosage form. A unit dose will generally contain from 0.1 to 1000 mg of the active compound.
Generally an effective administered amount of a compound of the invention will depend on the relative efficacy of the compound chosen, the severity of the disorder being treated and the weight of the sufferer. However, active compounds will typically be administered once or more times a day for example 2, 3 or 4 times daily, with typical total daily doses in the range of from 0.1 to 800 mg/kg/day.
Suitable dose forms for oral administration may be tablets and capsules and may contain conventional excipients such as binding agents, for example syrup, acacia, gelatin, sorbitol, tragacanth, or polyvinylpyrrolidone; fillers, for example lactose, sugar, maize starch, calcium phosphate, sorbitol or glycine; tabletting lubricants, for example magnesium stearate; disintegrants, for example starch, polyvinylpyrrolidone, sodium starch glycollate or microcrystalline cellulose; or pharmaceutically acceptable wetting agents such as sodium lauryl sulfate.
The solid oral compositions may be prepared by conventional methods of blending, filling or tabletting. Repeated blending operations may be used to distribute the active agent throughout those compositions employing large quantities of fillers. Such operations are of course conventional in the art. The tablets may be coated according to methods well known in normal pharmaceutical practice, in particular with an enteric coating.
Oral liquid preparations may be in the form of, for example, emulsions, syrups, or elixirs, or may be presented as a dry product for reconstitution with water or other suitable vehicle before use. Such liquid preparations may contain conventional additives such as suspending agents, for example sorbitol, syrup, methyl cellulose, gelatin, hydroxyethylcellulose, carboxymethylcellulose, aluminium stearate gel, hydrogenated edible fats; emulsifying agents, for example lecithin, sorbitan monooleate, or acacia; non-aqueous vehicles (which may include edible oils), for example almond oil, fractionated coconut oil, oily esters such as esters of glycerine, propylene glycol, or ethyl alcohol; preservatives, for example methyl or propyl p-hydroxybenzoate or sorbic acid; and if desired conventional flavouring or colouring agents.
For parenteral administration, fluid unit dosage forms are prepared utilizing the compound and a sterile vehicle, and, depending on the concentration used, can be either suspended or dissolved in the vehicle. In preparing solutions the compound can be dissolved in water for injection and filter sterilized before filling into a suitable vial or ampoule and sealing. Advantageously, adjuvants such as a local anaesthetic, a preservative and buffering agents can be dissolved in the vehicle. To enhance the stability, the composition can be frozen after filling into the vial and the water removed under vacuum. Parenteral suspensions are prepared in substantially the same manner, except that the compound is suspended in the vehicle instead of being dissolved, and sterilization cannot be accomplished by filtration. The compound can be sterilized by exposure to ethylene oxide before suspending in the sterile vehicle. Advantageously, a surfactant or wetting agent is included in the composition to facilitate uniform distribution of the compound. The formulations mentioned herein are carried out using standard methods such as those described or referred to in reference texts such as the British and US Pharmacopoeias, Remington's Pharmaceutical Sciences (Mack Publishing Co.), Martindale The Extra Pharmacopoeia (London, The Pharmaceutical Press) or the above- mentioned publications.
Suitable methods for preparing and suitable unit dosages for the additional medicament agent, such as the antidiabetic agent mentioned herein include those methods and dosages described or referred to in the above-mentioned reference texts.
GSK-3 Assay
GSK-3 assays used to test the compounds of the invention include the following protocol which is based on the ability of the kinase to phosphorylate a biotinylated 26 mer peptide, Biot- KYRRAAVPPSPSLSRHSSPHQ(S)EDEEE, the sequence of which is derived from the phosphorylation site of glycogen synthase, where (S) is a pre- phosphorylated serine as in glycogen synthase in vivo and the three consensus sites for GSK-3 specific phosphorylation are underlined. The phosphorylated biotinylated peptide is then captured onto Streptavidin coated SPA beads (Amersham Technology), where the signal from the 3p [s amplified via the scintillant contained in the beads.
Using microtitre plates, GSK-3 was assayed in 50 mM MOPS buffer, pH 7.0, containing 5% glycerol, 0.01% Tween-20, 7.5 mM 2-mercaptoethanol, 10 mM magnesium acetate, 8 uM of the above peptide, and 10 uM [33p]_ATP. After incubation at room temperature, the reaction was stopped by addition of 50 mM EDTA solution containing the Streptavidin coated SPA beads to give a final 0.2 mgs. Following centrifugation, the microtitre plates are counted in a Trilux 1450 microbeta liquid scintillation counter (Wallac). IC50 values are generated for each compound by fitting to a four parameter model.
The most potent compounds of the present invention show IC50 values in the range of 1 to 500 nM.
No adverse toxicological effects are expected for the compounds of the invention, when administered in accordance with the invention.
The following Descriptions and Examples illustrate the invention, but do not limit it in any way. Synthetic Method A
Example 1
N-(5-Bromo-lH-indazol-3-yl)butyramide n-Butyric anhydride (0.80 mL, 4.9 mmol) was added to a solution of 5-bromo-lH- indazol-3-ylamine (1.06 g, 5.0 mmol) in pyridine (5 mL). The reaction mixture was stirred at reflux for 22 hours, then allowed to cool. The solution was concentrated and purified by column chromatography (10% v/v methanol in chloroform) affording the title compound as a solid. MS (APCI+ve): [M+H]+ at m/z 282/284 (CπH12BrN3O requires [M+H]+ at m/z 282/284).
1HNMR 6 (DMSO-d6): 0.9 (3H, t), 1.6 (2H, appq), 2.4 (2H, t), 7.4 (2H, apps), 8.0 (1H, s), 10.4 (1H, s), 12.8 (1H, s).
Synthetic Method B Example 2 N-(5-Phenyl-lH-indazol-3-yl)-butyramide
Tefrakis(triphenylphosphine)palladium(0) (20 mg, 0.02 mmol) was added to a stirred solution of phenylboronic acid (56 mg, 0.46 mmol), N-(5-bromo-lH-indazol-3- yl)butyramide (100 mg, 0.35 mmol) and sodium carbonate (0.5 mL of 2M aqueous solution) in 1,2-dimethoxyethane (1 mL) and ethanol (0.5 mL). The resulting suspension was stirred at reflux for 20 hours, concentrated in vacuo and water (25 mL) added. The aqueous layer was extracted with ethyl acetate (x3) and the combined organic extracts were washed with brine, dried and concentrated. Purification by column chromatography (2% v/v methanol in dichloromethane) afforded the title compound as a solid.
MS (APCI+ve): [M+H]+ at m/z 280 (C17H17N3O requires [M+H]+ at m/z 280).
1H NMR δ (DMSO-d6): 0.9 (3H, t), 1.6 (2H, appq), 2.4 (2H, t), 7.7-7.3 (7H, m), 8.0 (1H, s), 10.3 (1H, s), 12.6 (1H, s). Synthetic Method C
Example 3
Cyclopentanecarboxylic acid (6-phenylamino-lH-indazoI-3-yl)-amide
[3 -[( 1 -Cycloρentyl-methanoyl)-amino] - 1 H-indazol-6-yl] -phenyl-carbamic acid 1,1- dimethylethyl ester (100 mg, 0.24 mmol) was dissolved in 4N hydrochloric acid in dioxane (5 mL) and the reaction mixture was stirred at room temperature for 2 hours. The reaction mixture was concentrated and the resulting crude oil purified by chromatography (2% methanol/dichloromethane) to afford a beige solid. The solid was washed with water and diethyl ether to afford the title compound as a solid. MS (ACPI+ve): [M+H]+ at m/z 321 (C19H20N4O requires [M+H]+ at m/z 321).
1H NMR δ (DMSO-d6): 1.58-1.90 (8H, m), 2.86-2.92 (1H, m), 7.75-7.79 (1H, dd), 7.84- 7.90 (1H, t), 7.97-7.98 (1H, d), 7.13-7.16 (2H, m), 7.24-7.30 (2H, m), 7.59-7.63 (1H, d), 8.29 (1H, s), 10.17 (1H, s), 12.09 (1H, s).
Description 1
2-Chloro-4-phenylamino-benzonitrile
To a stirred and degassed solution of 4-bromo-2-chlorobenzonitrile (2 g, 9.2 mmol), aniline (1 ml, 11.1 mmol), sodium tert-butoxide (1.3 g, 12.9 mmol) and BINAP (0.9 g,
1.4 mmol) in dry toluene was added Pd2(dba)3 (0.4 g, 0.5 mmol). The reaction mixture was stirred at 80°C for 18 hours, allowed to cool and filtered through celite. The filtrate was concentrated and the crude residue was purified by chromatography (20% ethyl acetate/60-80 petrol) to afford the title compound as a solid.
MS (ACPI+ve): [M+H]+ at m/z 229/231 (C13H9C1N2 requires [M+H]+ at m/z 229/231).
1H NMR (DMSO-d6): 6.97-7.01 (1H, dd), 7.07-7.10 (2H, m), 7.19-7.23 (2H, m), 7.35- 7.39 (2H, m), 7.65-7.68 (1H, d), 9.15 (1H, s).
Description 2
(3-Chloro-4-cyanophenyl)-phenyI-carbamic acid 1,1 -dimethylethyl ester
To a stirred solution of 2-chloro-4-phenylaminobenzonitrile (1.8 g, 7.9 mmol) in dry dimethylformamide (20 mL) was added at 0°C sodium hydride (0.5 g, 11.8 mmol) and the reaction mixture was stirred under argon for 1 hour. Di-tert-butyl dicarbonate (2.6 g, 11.8 mmol) was added and the reaction mixture was stirred at room temperature for 18 hours. The reaction mixture was quenched with water and concentrated. The crude oil was taken up in ethyl acetate (100 mL) and washed with brine (100 mL). The organic extract was dried (magnesium sulfate) and concentrated. The crude solid was purified by chromatography (20% ethyl acetate/60-80 petrol) to afford the title compound as a solid. MS (ACPI+ve): [M+H]+ at m z 329/331 (C18H17ClN2O2 requires [M+H]+ at m z 329/331).
1H NMR (DMSO-d6): 1.38 (9H, s), 7.20-7.22 (IH, dd), 7.27-7.29 (2H, m), 7.35-7.37 (IH, m), 7.43-7.45 (2H, m), 7.60-7.61 (IH, d), 7.88-7.90 (IH, d).
The general method of Description 1 was also employed to introduce the 4- benzyloxycarbonylpiperazin-1-yl substituent of Example 33 using 1- benzyloxycarbonylpiperazine and 4-bromo-2-chlorobenzonitrile
Synthetic Method D Example 4
Cyclopropanecarbox lie acid (6-thiophen-2-yl-lH-indazol-3-yl)-amide
To a stirred solution of 6-thiophen-2-yl-lH-indazol-3-ylamine (320 mg, 1.5 mmol) in dry pyridine (5 mL) under argon was added cyclopropanecarbonyl chloride (0.135 mL, 1.5 mmol) and the reaction was stirred at room temperature for 12 hours. The solution was concentrated. The crude oil was taken up in ethyl acetate (100 mL) and washed with saturated sodium bicarbonate solution in water (100 mL). The organic extract was dried (magnesium sulfate) and concentrated. The crude solid was triturated with dichloromethane to afford the title compound as a solid. MS (ACPI+ve): [M+H]+ at m/z 284 (C15H13N3OS requires [M+H]+ at m/z 284). 1H NMR (DMSO-d6): 0.82-0.90 (4H, m), 1.89-1.96 (IH, m), 7.14-7.18 (IH, m), 7.33- 7.40 (IH, dd), 7.55-7.61 (3H, m), 7.80-7.84 (IH, d), 10.7 (IH, s), 12.65 (IH, s).
Description 3 2-Chloro-4-thiophen-2-yl-benzonitrile To a stirred and degassed solution of 4-bromo-2-chlorobenzonitrile (1 g, 4.6 mmol), 2- thiophene boronic acid (0.7 g, 5.5 mmol) and potassium acetate (1.4 g, 13.9 mmol) in dimethylformamide (10 mL), ethanol (5 mL) and water (5 mL) was added PdCl2(dppf) (113 mg, 0.1 mmol). The reaction mixture was stirred at 100°C for 4 hours, allowed to cool and filtered through celite. The filtrate was taken up in ethyl acetate (100 mL) and was washed with brine (3 x 100 mL). The organic extract was dried (magnesium sulfate) and concentrated. The crude solid was purified by chromatography (10% diethyl ether/60-80 petrol) to afford the title compound as a solid.
MS (ACPI+ve): [M+H]+ at m/z 220/222 (CnH6ClNS requires [M+H]+ at m/z 220/222). 1H NMR (DMSO-d6): 7.21-7.24 (IH, m), 7.75-7.84 (3H, m), 7.97-8.00 (IH, d), 8.06-8.07 (lH, d).
Description 4
6-Thiophen-2-yl-lH-indazol-3-ylamine
To a stirred solution of 2-chloro-4-thiophen-2-yl-benzonitrile (900 mg, 4.1 mmol) in pyridine (5 mL) was added hydrazine hydrate (2 mL, 41 mmol). The reaction mixture was stirred at reflux for 72 hours, allowed to cool and concentrated. The crude oil was taken up in ethyl acetate (100 mL) and washed with saturated sodium bicarbonate solution in water (100 mL). The organic extract was dried (magnesium sulfate) and concentrated. The crude solid was triturated with dichloromethane to afford the title compound as a solid. MS (ACPI+ve): [M+H]+ at m/z 216 (CnH9N3S requires [M+H]+ at m/z 216). 1H NMR (DMSO-d6): 5.37 (2H, s), 7.12-7.16 (IH, t), 7.21-7.25 (IH, dd), 7.43 (IH, s), 7.52-7.54 (2H, m), 7.69-7.72 (IH, d), 11.42 (IH, s).
An alternative method for the introduction of a substituent R^ is provided the method of Description 5 with a subsequent coupling step. In the latter process an appropriate aryl or heteroaryl halide is reacted with the compound of Description 5 by the general procedure of Description 3 employing a suitable palladium catalyst.
Description 5 2-Chloro-4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-benzonitrile l,r-Bis(diphenylphosphino)ferrocenedichloropalladium(π) (1.7 g, 2.1 mmol) was added to a stirred and degassed solution of 4-bromo-2-chlorobenzonitrile (15 g, 69.3 mmol), bis(pinacolato)diboron (21.1 g, 83.2 mmol) and potassium acetate (20.4 g, 207.9 mmol) in dimethyl sulfoxide (200 mL). The reaction mixture was stirred at 100°C under argon for 18 hours, then allowed to cool. The solution was filtered through celite, then ethyl acetate (500 mL) was added and the organic phase was washed three times with brine (500 mL). The organic layer was dried (magnesium sulfate), concentrated and purified by chromatography (30% v/v ethyl acetate in petroleum ether) to afford a brown solid. Trituration with diethyl ether afforded the title compound as a solid. 1H NMR δ (DMSO-d6): 1.31 (12H, s), 7.73-7.76 (IH, dd), 7.82 (IH, app s), 7.96-8.00 (lH, d).
Examples containing a phenol substituent may be prepared by cleavage of an appropriate ether by the general procedure of Description 6. The product can then be acylated by a general method such as Synthetic Method D.
Description 6 4-(3-Amino-lH-mdazol-6-yl)-phenol
6-(4-Methoxy-phenyl)-lH-indazol-3-ylamine (150 mg, 0.63 mmol) was stirred at reflux for 4 hour in a 48% hydrobromic acid aqueous solution (5 mL). The reaction mixture was allowed to cool and was concentrated. The crude solid was triturated with water to afford the title compound as a solid. 1H NMR (DMSO-d6): 5.38 (2H, s), 6.83-6.86 (2H, d), 7.11-7.15 (IH, d), 7.31 (IH, s), 7.48-7.52 (2H, d), 7.71-7.77 (IH, d), 9.51 (IH, s), 11.35 (IH, s).
Where a ketone is present as an aryl substituent a hydrazone may be formed in the the cyclization step. Subsequent cleavage back to a carbonyl compound is exemplified in Description 7.
Description 7 l-[4-(3-Amino-lH-indazol-6-yl)-phenyl]-ethanone
6-[4-(l-Hydrazonoethyl)phenyl]-lH-indazol-3-ylamine (100 mg, 0.38 mmol) was stirred at reflux for 18 hours in a 4N hydrochloric solution in dioxane (10 mL) and water (1 mL). The reaction mixture was allowed to cool and was concentrated. The crude solid was taken up in ethyl acetate (50 mL) and washed with saturated sodium bicarbonate solution in water (50 mL). The organic extract was dried (magnesium sulfate) and concentrated. The resulting solid was purified by chromatography (ethyl acetate) to afford the title compound as a solid.
MS (ACPI+ve): [M+H]+ at m/z 252 (C15H13N3O requires [M+H]+ at m/z 252). 1H NMR (DMSO-d6): 2.62 (3H, s), 5.41 (2H, s), 7.25-7.29 (IH, dd), 7.52 (IH, s), 7.78- 7.87 (3H, m), 8.03-8.06 (2H, d), 11.53 (IH, s).
Synthetic Method E Example 11 Cyclopropanecarboxylic acid (6-benzenesulfanyl-lH-indazol-3-yl)-amide
6-Phenylsulfanyl-lH-indazol-3-ylamine (303 mg, 1.26 mmol) and cyclopropylcarbonyl chloride (114 μL, 1.26 mmol) were dissolved in anhydrous pyridine (5 mL). The solution was stirred for 16 hours under argon at ambient temperature. A further amount of cyclopropyl carbonyl chloride (114 μL, 1.26 mmol) was added and the reaction stirred for a further 2 hours at the same temperature. Water (2 mL) was added and the solution concentrated to dryness. Piperidine (20 mL) and water (3 mL) were added and the solution stirred at ambient temperature under argon for 3 hours. The solvents were removed in vacuo and the product purified by preparative HPLC (10-90% acetonitrile (0.1% trifluoroacetic acid) in water (0.1% trifluoroacetic acid) over 10 minutes, C18 column). The appropriate fractions were concentrated to dryness to yield the title compound, as a solid.
MS (APCI+ve): [M+H]+ at m/z 310 (C17H15N3OS requires [M+H]+ at m/z 310.)
IH NMR δ (DMSO-d6) : 0.78-0.84 (4H, m), 1.91 (IH, br m), 6.97 (IH, dd), 7.26 (IH, s), 7.37 (5H, m), 7.77 (IH, d), 10.67 (IH, s), 12.58 (IH, s).
The starting material for Synthetic Method E is prepared as shown below in Descriptions 8 and 9.
Description 8 2-Chloro-4-phenylsulfanyl-benzonitrile
Thiophenol (3.63 mL, 35.4 mmol) was added to 2-chloro-4-fluoroberιzonitrile (5 g, 32.1 mmol), sodium hydride (2.56 g, 64.2 mmol) in dimethylformamide (20 mL). The resulting solution was stirred at ambient temperature under argon for 16 hours. The solution was concentrated to dryness and purified by preparative HPLC (10-90% acetonitrile (0.1% trifluoroacetic acid) in water (0.1% trifluoroacetic acid) over 10 minutes, C18 column). The appropriate fractions were concentrated to dryness to yield the title compound as a solid.
MS (APCI-ve): [M-H]" at m z 168/170 (C13H8NC1S requires [M-H]" at m/z 244/246. Observed ion fits for loss of phenyl group).
IH NMR δ (DMSO-d6): 7.14 (IH, dd), 7.35 (IH, d), 7.56 (5H, m), 7.84 (IH, d).
Description 9
6-Phenylsulfanyl-lH-indazol-3-ylamine
Hydrazine hydrate (4 mL, 128 mmol) was added to 2-chloro-4-phenylsulfanyl- benzonitrile (1 g, 4.07 mmol) in pyridine (10 mL). The solution was heated at reflux for 96 hours, allowed to cool and concentrated to dryness. Purification was achieved by preparative HPLC (10-90% acetonitrile (0.1% trifluoroacetic acid) in water (0.1% trifluoroacetic acid) over 10 minutes, C18 column). The appropriate fractions were concentrated to dryness to yield the title compound as a solid.
MS (APCI+ve): [M+H]+ at m/z 242 (C13HπN3S requires [M+HJ+ at m/z 242.) IH NMR δ (DMSO-d6): 4.00-6.00 (2H, v. br s), 6.98 (IH, dd), 7.11 (IH, s), 7.42 (5H, m), 7.84 (IH, d). The pyrazole NH was not observed.
Synthetic Method F
Example 16
Cyclopentanecarboxylic acid (6-benzenesulfonyl-lH-indazol-3-yl)-amide Cyclopentanecarboxylic acid (6-benzenesulfanyl-lH-indazol-3-yl)-amide (100 mg, 0.324 mmol) and weto-chloroperbenzoic acid (168 mg, 0.972 mmol) were dissolved in dichloromethane (30 mL). The solution was stirred for 16 hours under argon at ambient temperature. A further 150 mL dichloromethane was added to dissolve the precipitate and the organic layer was washed with saturated sodium bicarbonate solution (3 x 150 mL). The organic layer was concentrated and purified by preparative HPLC (10-90% acetonitrile (0.1% trifluoroacetic acid) in water (0.1% trifluoroacetic acid) over 10 minutes, C18 column). The appropriate fractions were concentrated to dryness to yield the title compound as a solid.
MS (APCI+ve): [M+H]+ at m/z 370 (C19H19N3O3S requires [M+H]+ at m/z 370.) H NMR δ (DMSO-d6) : 1.54-1.89 (8H, m), 2.90 (IH, quint), 7.50 (IH, dd), 7.62 (2H, 5 t), 7.69 (IH, t), 7.95-7.99 (3H, m), 8.08 (IH, s), 10.48 (IH, s), 13.22 (IH, br s).
Synthetic Method G
Example 31
Cyclopropanecarboxy lie acid (6-piperazin-l-yl-lH-indazol-3-yl)-amide
10 10% Palladium on charcoal (16 mg) was added to a solution of 4-(3-[(l-cyclopropyl- methanoyl)-amino]-lH-indazol-6-yl)-piperazine-l-carboxylic acid benzyl ester (160 mg, 0.38 mmol) in ethanol (10 mL) and the reaction mixture was stirred at room temperature under 50 psi of hydrogen for 48 hours. The solution was filtered through celite and concentrated. The crude solid was purified by chromatography on silica gel (10%
15 methanol/dichloromethane then 10% (2N ammonia in methanol)/dichloromethane) to afford the title compound as a solid.
MS (ACPI+ve): [M+H]+ at m/z 286 (C15H19N5O requires [M+H]+ at m/z 286). 1H NMR δ (DMSO-d6): 0.78-0.83 (4H, m), 1.86-1.93 (IH, m), 2.77-2.78 (4H, m), 2.88- 2.92 (4H, m), 6.62 (IH, apps), 6.79-6.84 (IH, dd), 7.60-7.63 (IH, d), 10.50 (IH, s), 12.14 0 (lH, s).
The further Examples described herein were prepared by analogy with Synthetic Methods A-G described above.
5
0 Table 1
Figure imgf000036_0001
Figure imgf000036_0002
Figure imgf000037_0001

Claims

Claims
1. A compound of formula (I),
Figure imgf000038_0001
or a salt thereof, or a solvate thereof, wherein,
Rl is unsubstituted or substituted alkyl, unsubstituted or substituted cycloalkyl, unsubstituted or substituted alkenyl, unsubstituted or substituted cycloalkenyl, unsubstituted or substituted aryl, aralkyl wherein the aryl and the alkyl moieties may each independently be unsubstituted or substituted, aralkenyl wherein the aryl and alkenyl moieties may each independently be unsubstituted or substituted, unsubstituted or substituted heterocyclyl, or heterocyclylalkyl wherein the heterocyclyl and the alkyl moieties may each independently be unsubstituted or substituted, unsubstituted or substituted heteroaryl, heteroarylalkyl wherein the heteroaryl and the alkyl moieties may each independently be unsubstituted or substituted;
R2 is H;
R3 is H, halo, alkyl, unsubstituted or substituted aryl or unsubstituted or substituted heteroaryl;
R4 is H, unsubstituted or substituted aryl; unsubstituted or substituted heteroaryl, unsubstituted or substituted heterocyclyl; or -X-aryl;
R^ is H;
X is O, NH, S or S(O)n and n is 1 or 2; with the proviso that when Rl, R2 and R^ are as defined in relation to formula (I), and R3 is H or halo, then R^ is unsubstituted or substituted aryl; unsubstituted or substituted heteroaryl, unsubstituted or substituted heterocyclyl; or -X-aryl.
2. A compound of formula (I), as claimed in claim 1, of formula (IA),
Figure imgf000039_0001
(IA)
or a salt thereof, or a solvate thereof, wherein,
Rl is alkyl, cycloC3_g alkyl or heterocyclyl wherein the heterocyclyl group is substituted by arylalkyl;
R2 is H;
R is H, halo or aryl; and R4 is H, aryl wherein the aryl group may be optionally substituted by one or more substituents selected from halo, -SO2NH2, -OH, -NHSO2Me and -SO2Me; heteroaryl wherein the heteroaryl group may be optionally substituted by one or more substituents selected from halo, alkyl and oxo; heterocyclyl wherein the heterocyclyl group may be optionally substituted by aryloxycarbonyl; or -X-aryl; with the proviso that when Rl, R2 and R^ are as defined in relation to formula (LA), and
R is H or halo, then R4 is aryl wherein the aryl group may be optionally substituted by one or more substituents selected from halo, -SO2NH2, -OH, -NHSO2Me and -SO2Me; heteroaryl wherein the heteroaryl group may be optionally substituted by one or more substituents selected from halo, alkyl and oxo; heterocyclyl wherein the heterocyclyl group may be optionally substituted by aryloxycarbonyl; or -X-aryl.
3. A compound of formula (I), as claimed in claim 1 , of formula (IB),
Figure imgf000040_0001
(IB)
or a salt thereof, or a solvate thereof, wherein,
R is n-propyl, cyclopropyl, cyclopentyl or N-benzylpyrrolidin-3-yl;
R2 is H;
R3 is H, bromo or phenyl;
R4 is H, phenyl, 2-fluorophenyl, 3 -fluorophenyl, 4-fluorophenyl, 2,3-difluorophenyl, 2,5- difluorophenyl, 3,5-difluorophenyl, 3-H2NSO2-Ph, 4-H2NSO2-Ph, 3-MeSO2NH-Ph, 4-
MeSO2NH-Ph, 4-methanesulfonylphenyl, 4-hydroxyphenyl, 2-furyl, 3-furyl, 2-pyrrolyl,
2-thienyl, 3-thienyl, 4-methylthien-2-yl, 5-bromothien-2-yl, indol-5-yl, pyrid-2-on-5-yl, - NHPh, -OPh, -SPh, -SO2PI1, ρiperazin-1-yl or 4-benzyloxycarbonylpiperazin-l-yl; and
R5 is H; with the proviso that when R , R2 and R^ are as defined in relation to formula (IB), and
R3 is H or bromo, then R^ is phenyl, 2-fluorophenyl, 3 -fluorophenyl, 4-fluorophenyl, 2,3-difluorophenyl, 2,5-difluorophenyl, 3,5-difluorophenyl, 3-H2NSO2-PI1, 4-H2NSO2-
Ph, 3-MeSO2NH-Ph, 4-MeSO2NH-Ph, 4-methanesulfonylphenyl, 4-hydroxyphenyl, 2- furyl, 3-furyl, 2-pyrrolyl, 2-thienyl, 3-thienyl, 4-methylthien-2-yl, 5-bromothien-2-yl, indol-5-yl, pyrid-2-on-5-yl, -NHPh, -OPh, -SPh, -SO2Ph, piperazin-1-yl or 4- benzyloxycarbonylpiperazin- 1 -yl.
4. A process for the preparation of a compound of formula (I), or a salt and/or solvate thereof, as claimed in claim 1, which process comprises reacting a compound of formula (II),
Figure imgf000041_0001
wherein R2, R , R4 and R^ are as defined in relation to formula (I) with a compound of formula (IH),
Figure imgf000041_0002
wherein Rl is as defined in relation to formula (I) and X is a suitable leaving group and thereafter, if required, carrying out one or more of the following optional steps: (i) converting a compound of formula (I) to a further compound of formula (I); (ii) removing any necessary protecting group; (iii) preparing an appropriate derivative of the compound so formed.
5. A process for the preparation of a compound of formula (I), or a salt and/or solvate thereof, as claimed in claim 1, which process comprises reacting a compound of formula (IV),
Figure imgf000041_0003
wherein R2, R , R4 and R^ are as defined in relation to formula (I), with a nucleophile and thereafter, if required, carrying out one or more of the following optional steps: (i) converting a compound of formula (I) to a further compound of formula (I); (ii) removing any necessary protecting group; (iii) preparing an appropriate derivative of the compound so formed.
6. A compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, as claimed in claim 1, for use as an inhibitor of GSK-3.
7. A method for the treatment of conditions associated with a need for inhibition of GSK-3 such as diabetes, conditions associated with diabetes, chronic neurodegenerative conditions including dementias such as Alzheimer's disease, Parkinson's disease, progressive supranuclear palsy, subacute sclerosing panencephalitic parkinsonism, postencephalitic parkinsonism, pugilistic encephalitis, guam parkinsonism-dementia complex, Pick's disease, corticobasal degeneration, frontotemporal dementia, Huntingdon's disease, ADDS associated dementia, amyotrophic lateral sclerosis, multiple sclerosis and neurotraurnatic diseases such as acute stroke, mood disorders such as schizophrenia and bipolar disorders, promotion of functional recovery post stroke, cerebral bleeding (for example, due to solitary cerebral amyloid angiopathy), hair loss, obesity, atherosclerotic cardiovascular disease, hypertension, polycystic ovary syndrome, syndrome X, ischaemia, traumatic brain injury, cancer, leukopenia, Down's syndrome, Lewy body disease, inflammation, and immunodeficiency, which method comprises the administration of a pharmaceutically effective, non-toxic amount of a compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof, as claimed in claim 1.
8. Use of a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, as claimed in claim 1, for the manufacture of a medicament for the treatment of conditions associated with a need for the inhibition of GSK-3, such as diabetes, conditions associated with diabetes, chronic neurodegenerative conditions including dementias such as Alzheimer's disease, Parkinson's disease, progressive supranuclear palsy, subacute sclerosing panencephalitic parkinsonism, postencephalitic parkinsonism, pugilistic encephalitis, guam parkinsonism-dementia complex, Pick's disease, corticobasal degeneration, frontotemporal dementia, Huntingdon's disease, AIDS associated dementia, amyotrophic lateral sclerosis, multiple sclerosis and neurotraurnatic diseases such as acute stroke, mood disorders such as schizophrenia and bipolar disorders, promotion of functional recovery post stroke, cerebral bleeding (for example, due to solitary cerebral amyloid angiopathy), hair loss, obesity, atherosclerotic cardiovascular disease, hypertension, polycystic ovary syndrome, syndrome X, ischaemia, traumatic brain injury, cancer, leukopenia, Down's syndrome, Lewy body disease, inflammation, and immunodeficiency.
9. A pharmaceutical composition which comprises a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, as claimed in claim 1, and a pharmaceutically acceptable carrier.
PCT/GB2002/005826 2001-12-19 2002-12-19 (1-h-indazol-3-yl) -amide derivatives as gsk-3 inhibitors WO2003051847A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2002353186A AU2002353186A1 (en) 2001-12-19 2002-12-19 (1-h-indazol-3-yl) -amide derivatives as gsk-3 inhibitors

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
GB0130352A GB0130352D0 (en) 2001-12-19 2001-12-19 Novel compounds
GB0130352.8 2001-12-19
GB0206612A GB0206612D0 (en) 2002-03-20 2002-03-20 Novel compounds
GB0206612.4 2002-03-20

Publications (1)

Publication Number Publication Date
WO2003051847A1 true WO2003051847A1 (en) 2003-06-26

Family

ID=26246885

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2002/005826 WO2003051847A1 (en) 2001-12-19 2002-12-19 (1-h-indazol-3-yl) -amide derivatives as gsk-3 inhibitors

Country Status (2)

Country Link
AU (1) AU2002353186A1 (en)
WO (1) WO2003051847A1 (en)

Cited By (64)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003097610A1 (en) * 2002-05-17 2003-11-27 Pharmacia Italia S.P.A. Aminoindazole derivatives active as kinase inhibitors, process for their preparation and pharmaceutical compositions comprising them
WO2004007504A1 (en) * 2002-07-17 2004-01-22 Pharmacia Italia S.P.A. Heterobicyclic pyrazole derivatives as kinase inhibitors
WO2004113304A1 (en) * 2003-05-22 2004-12-29 Abbott Laboratories Indazole, benzisoxazole, and benzisothiazole kinase inhibitors
WO2005085206A1 (en) * 2004-02-27 2005-09-15 F. Hoffmann-La Roche Ag Indazole derivatives and pharmaceutical compositions containing them
FR2871158A1 (en) * 2004-06-04 2005-12-09 Aventis Pharma Sa SUBSTITUTED INDAZOLES, COMPOSITIONS CONTAINING SAME, METHOD OF MANUFACTURE AND USE
EP1606283A2 (en) * 2003-03-03 2005-12-21 Array Biopharma, Inc. P38 inhibitors and methods of use thereof
US7135575B2 (en) 2003-03-03 2006-11-14 Array Biopharma, Inc. P38 inhibitors and methods of use thereof
JP2006528212A (en) * 2003-05-15 2006-12-14 アジェンデ・キミケ・リウニテ・アンジェリニ・フランチェスコ・ア・チ・エレ・ア・エフェ・ソシエタ・ペル・アチオニ Indazole with analgesic activity
WO2007090493A1 (en) * 2006-02-06 2007-08-16 Merck Patent Gmbh Indazole heteroaryl derivatives
US7297709B2 (en) 2003-05-22 2007-11-20 Abbott Laboratories Indazole, benzisoxazole, and benzisothiazole kinase inhibitors
WO2008009335A2 (en) * 2006-07-18 2008-01-24 Merck Patent Gmbh Aminoindazole urea derivatives
JP2008502610A (en) * 2004-06-15 2008-01-31 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフトング 3-aminoindazole
WO2008086854A1 (en) 2007-01-18 2008-07-24 Merck Patent Gmbh 5-([1,3,4] oxadiazol-2-yl)-1h-indazol and 5-([1,3,4] thiadiazol-2-yl)-1h-indazol derivatives as sgk inhibitors for the treatment of diabetes
DE102007022565A1 (en) 2007-05-14 2008-11-20 Merck Patent Gmbh Heterocyclic indazole derivatives
WO2009013126A1 (en) * 2007-07-20 2009-01-29 Nerviano Medical Sciences S.R.L. Substituted indazole derivatives active as kinase inhibitors
JP2009525996A (en) * 2006-02-06 2009-07-16 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツング Aminoindazole derivatives
WO2009147189A1 (en) * 2008-06-05 2009-12-10 Glaxo Group Limited Novel compounds
DE102008038220A1 (en) 2008-08-18 2010-02-25 Merck Patent Gmbh oxadiazole
DE102008038221A1 (en) 2008-08-18 2010-02-25 Merck Patent Gmbh 7-azaindole derivatives
DE102008038222A1 (en) 2008-08-18 2010-02-25 Merck Patent Gmbh Indazol-5-carboxylic acid derivatives
WO2010064875A2 (en) * 2008-12-05 2010-06-10 Korea Institute Of Science And Technology Novel indazole derivatives or pharmaceutically acceptable salts thereof as protein kinase inhibitors for proliferative diseases treatment, and a pharmaceutical composition containing the same as an active ingredient
WO2010069966A1 (en) * 2008-12-18 2010-06-24 Nerviano Medical Sciences S.R.L. Substituted indazole derivatives active as kinase inhibitors
WO2010099217A1 (en) 2009-02-25 2010-09-02 Braincells, Inc. Modulation of neurogenesis using d-cycloserine combinations
EP2258359A2 (en) 2005-08-26 2010-12-08 Braincells, Inc. Neurogenesis by muscarinic receptor modulation with sabcomelin
EP2275095A2 (en) 2005-08-26 2011-01-19 Braincells, Inc. Neurogenesis by muscarinic receptor modulation
EP2314289A1 (en) 2005-10-31 2011-04-27 Braincells, Inc. Gaba receptor mediated modulation of neurogenesis
WO2011063115A1 (en) 2009-11-19 2011-05-26 Braincells Inc. Combination of nootropic agent with one or more neurogenic or neurogenic sensitizing agents for stimulating or increasing neurogenesis
JP2011522004A (en) * 2008-06-05 2011-07-28 グラクソ グループ リミテッド New compounds
WO2011091033A1 (en) 2010-01-20 2011-07-28 Braincells, Inc. Modulation of neurogenesis by ppar agents
JP2011522002A (en) * 2008-06-05 2011-07-28 グラクソ グループ リミテッド Benzpyrazole derivatives as inhibitors of PI3 kinase
JP2011522001A (en) * 2008-06-05 2011-07-28 グラクソ グループ リミテッド 4-Carboxamide indazole derivatives useful as inhibitors of PI3 kinase
US8039639B2 (en) 2006-01-31 2011-10-18 Array Biopharma Inc. Kinase inhibitors and methods of use thereof
EP2377530A2 (en) 2005-10-21 2011-10-19 Braincells, Inc. Modulation of neurogenesis by PDE inhibition
EP2377531A2 (en) 2006-05-09 2011-10-19 Braincells, Inc. Neurogenesis by modulating angiotensin
JP2013512879A (en) * 2009-12-03 2013-04-18 グラクソ グループ リミテッド Benzpyrazole derivatives as inhibitors of PI3 kinase
JP2013522292A (en) * 2010-03-16 2013-06-13 デイナ ファーバー キャンサー インスティチュート,インコーポレイテッド Indazole compounds and their use
WO2013100672A1 (en) * 2011-12-29 2013-07-04 제이더블유중외제약㈜ 3,6-disubstituted indazole derivative having protein kinase inhibiting activity
US8524751B2 (en) 2009-03-09 2013-09-03 GlaxoSmithKline Intellecutual Property Development 4-oxadiazol-2-YL-indazoles as inhibitors of P13 kinases
US8575162B2 (en) 2009-04-30 2013-11-05 Glaxosmithkline Intellectual Property Development Limited Compounds
US8993576B2 (en) 2010-10-27 2015-03-31 Glaxo Group Limited 6-(1H-indol-4-yl)-4-(5-{[4-1-methylethyl)-1-piperazinyl]methyl}-1,3-oxazol-2-yl)-1H-indazole hemi succinate salt, polymorphs and pharmaceutical compositions thereof
WO2015112445A1 (en) * 2014-01-24 2015-07-30 Abbvie Inc. 6-phenyl- or 6-(pyridin-3-yl)indazole derivatives and methods of use
WO2016096709A1 (en) 2014-12-16 2016-06-23 Eudendron S.R.L. Heterocyclic derivatives modulating activity of certain protein kinases
WO2016188214A1 (en) * 2015-05-27 2016-12-01 上海海和药物研究开发有限公司 Preparation and use of novel kinase inhibitor
WO2017045751A1 (en) * 2015-09-15 2017-03-23 Merck Patent Gmbh Compounds as asic inhibitors and uses thereof
US9758522B2 (en) 2012-10-19 2017-09-12 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged small molecules as inducers of protein degradation
US9862688B2 (en) 2014-04-23 2018-01-09 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged janus kinase inhibitors and uses thereof
US10000483B2 (en) 2012-10-19 2018-06-19 Dana-Farber Cancer Institute, Inc. Bone marrow on X chromosome kinase (BMX) inhibitors and uses thereof
US10017477B2 (en) 2014-04-23 2018-07-10 Dana-Farber Cancer Institute, Inc. Janus kinase inhibitors and uses thereof
US10085979B2 (en) 2014-12-02 2018-10-02 Ignyta, Inc. Combinations for the treatment of neuroblastoma
JP2018531266A (en) * 2015-10-22 2018-10-25 セルビタ エス.エー. Pyridone derivatives and their use as kinase inhibitors
US10112927B2 (en) 2012-10-18 2018-10-30 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US10144730B2 (en) 2011-11-17 2018-12-04 Dana-Farber Cancer Institute, Inc. Inhibitors of c-Jun-N-terminal kinase (JNK)
US10231965B2 (en) 2014-02-20 2019-03-19 Ignyta, Inc. Molecules for administration to ROS1 mutant cancer cells
US10398693B2 (en) 2017-07-19 2019-09-03 Ignyta, Inc. Pharmaceutical compositions and dosage forms
US10550121B2 (en) 2015-03-27 2020-02-04 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
US10702527B2 (en) 2015-06-12 2020-07-07 Dana-Farber Cancer Institute, Inc. Combination therapy of transcription inhibitors and kinase inhibitors
WO2020163812A1 (en) 2019-02-08 2020-08-13 Frequency Therapeutics, Inc. Valproic acid compounds and wnt agonists for treating ear disorders
US10869864B2 (en) 2015-12-18 2020-12-22 Ignyta, Inc. Combinations for the treatment of cancer
US10870651B2 (en) 2014-12-23 2020-12-22 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US10906889B2 (en) 2013-10-18 2021-02-02 Dana-Farber Cancer Institute, Inc. Polycyclic inhibitors of cyclin-dependent kinase 7 (CDK7)
US11007191B2 (en) 2017-10-17 2021-05-18 Ignyta, Inc. Pharmaceutical compositions and dosage forms
US11040957B2 (en) 2013-10-18 2021-06-22 Dana-Farber Cancer Institute, Inc. Heteroaromatic compounds useful for the treatment of proliferative diseases
US11142507B2 (en) 2015-09-09 2021-10-12 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
US11826365B2 (en) 2009-12-29 2023-11-28 Dana-Farber Cancer Institute, Inc. Type II raf kinase inhibitors

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE1280878B (en) * 1961-11-30 1968-10-24 Smith Kline French Lab 3-aminoindazoles
US4120811A (en) * 1976-09-02 1978-10-17 Kao Soap Co., Ltd. Safe bleaching compositions for colored and patterned fabrics
WO2001085726A1 (en) * 2000-05-10 2001-11-15 Lg Life Sciences Ltd. Indazoles substituted with 1,1-dioxoisothiazolidine useful as inhibitors of cell proliferation

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE1280878B (en) * 1961-11-30 1968-10-24 Smith Kline French Lab 3-aminoindazoles
US4120811A (en) * 1976-09-02 1978-10-17 Kao Soap Co., Ltd. Safe bleaching compositions for colored and patterned fabrics
WO2001085726A1 (en) * 2000-05-10 2001-11-15 Lg Life Sciences Ltd. Indazoles substituted with 1,1-dioxoisothiazolidine useful as inhibitors of cell proliferation

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
DAIDONE, GIUSEPPE ET AL: "Synthesis, crystallographic studies and biological evaluation of some 2-substituted 3-indazolyl-4(3H)-quinazolinones and 3-indazolyl-4(3H)- benzotriazinones", HETEROCYCLES (1996), 43(11), 2385-2396, XP002221089 *
RAFFA, DEMETRIO ET AL: "Synthesis and antiproliferative activity of novel 3-(indazol-3-yl)- quinazolin-4(3H)-one and 3-(indazol-3-yl)benzotriazin-4(3H)-one derivatives", ARCHIV DER PHARMAZIE (WEINHEIM, GERMANY) (1999), 332(9), 317-320, XP002221090 *

Cited By (138)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003097610A1 (en) * 2002-05-17 2003-11-27 Pharmacia Italia S.P.A. Aminoindazole derivatives active as kinase inhibitors, process for their preparation and pharmaceutical compositions comprising them
WO2004007504A1 (en) * 2002-07-17 2004-01-22 Pharmacia Italia S.P.A. Heterobicyclic pyrazole derivatives as kinase inhibitors
US7521447B2 (en) 2003-03-03 2009-04-21 Array Biopharma Inc. P38 inhibitors and methods of use thereof
US7799782B2 (en) 2003-03-03 2010-09-21 Array Biopharma Inc. P38 inhibitors and methods of use thereof
US8017641B2 (en) 2003-03-03 2011-09-13 Array Biopharma Inc. P38 inhibitors and methods of use thereof
EP1606283A2 (en) * 2003-03-03 2005-12-21 Array Biopharma, Inc. P38 inhibitors and methods of use thereof
US8518983B2 (en) 2003-03-03 2013-08-27 Array Biopharma Inc. P38 inhibitors and methods of use thereof
US7135575B2 (en) 2003-03-03 2006-11-14 Array Biopharma, Inc. P38 inhibitors and methods of use thereof
EP1606283A4 (en) * 2003-03-03 2007-05-23 Array Biopharma Inc P38 inhibitors and methods of use thereof
JP2006528212A (en) * 2003-05-15 2006-12-14 アジェンデ・キミケ・リウニテ・アンジェリニ・フランチェスコ・ア・チ・エレ・ア・エフェ・ソシエタ・ペル・アチオニ Indazole with analgesic activity
JP4865559B2 (en) * 2003-05-15 2012-02-01 アジェンデ・キミケ・リウニテ・アンジェリニ・フランチェスコ・ア・チ・エレ・ア・エフェ・ソシエタ・ペル・アチオニ Indazole with analgesic activity
US7598283B2 (en) 2003-05-22 2009-10-06 Abbott Laboratories, Inc. Indazole, benzisoxazole, and benzisothiazole kinase inhibitors
WO2004113304A1 (en) * 2003-05-22 2004-12-29 Abbott Laboratories Indazole, benzisoxazole, and benzisothiazole kinase inhibitors
EP2246333A1 (en) * 2003-05-22 2010-11-03 Abbott Laboratories Indazole, benzisoxazole, and benzisothiazole kinase inhibitors
US8642776B2 (en) 2003-05-22 2014-02-04 Abbvie Inc. Indazole, benzisoxazole, and benzisothiazole kinase inhibitors
US8299243B2 (en) 2003-05-22 2012-10-30 Abbvie Inc. Indazole, benzisoxazole, and benzisothiazole kinase inhibitors
US8063091B2 (en) 2003-05-22 2011-11-22 Abbott Laboratories Indazole, benzisoxazole, and benzisothiazole kinase inhibitors
US8940778B2 (en) 2003-05-22 2015-01-27 Abbvie Inc. Indazole, benzisoxazole, and benzisothiazole kinase inhibitors
US7297709B2 (en) 2003-05-22 2007-11-20 Abbott Laboratories Indazole, benzisoxazole, and benzisothiazole kinase inhibitors
WO2005085206A1 (en) * 2004-02-27 2005-09-15 F. Hoffmann-La Roche Ag Indazole derivatives and pharmaceutical compositions containing them
US7495015B2 (en) 2004-02-27 2009-02-24 Roche Palo Alto Llc Indazole derivatives and methods for using the same
US7517902B2 (en) 2004-06-04 2009-04-14 Aventis Pharma S.A. Substituted indazoles, compositions containing the same, and the preparation and use thereof
WO2006003276A1 (en) * 2004-06-04 2006-01-12 Aventis Pharma S.A. Substituted indazoles, compositions containing same, preparation and use
FR2871158A1 (en) * 2004-06-04 2005-12-09 Aventis Pharma Sa SUBSTITUTED INDAZOLES, COMPOSITIONS CONTAINING SAME, METHOD OF MANUFACTURE AND USE
EA012702B1 (en) * 2004-06-04 2009-12-30 Авентис Фарма С.А. Substituted indazoles, compositions containing same, preparation and use
AU2005259139B2 (en) * 2004-06-04 2011-09-08 Aventis Pharma S.A. Substituted indazoles, compositions containing same, preparation and use
AU2005254617B2 (en) * 2004-06-15 2011-05-26 Merck Patent Gmbh 3-aminoindazoles
US7872039B2 (en) * 2004-06-15 2011-01-18 Merck Patent Gesellschaft 3-aminoindazoles
JP2008502610A (en) * 2004-06-15 2008-01-31 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフトング 3-aminoindazole
EP2258359A2 (en) 2005-08-26 2010-12-08 Braincells, Inc. Neurogenesis by muscarinic receptor modulation with sabcomelin
EP2275095A2 (en) 2005-08-26 2011-01-19 Braincells, Inc. Neurogenesis by muscarinic receptor modulation
EP2258357A2 (en) 2005-08-26 2010-12-08 Braincells, Inc. Neurogenesis with acetylcholinesterase inhibitor
EP2258358A2 (en) 2005-08-26 2010-12-08 Braincells, Inc. Neurogenesis with acetylcholinesterase inhibitor
EP2275096A2 (en) 2005-08-26 2011-01-19 Braincells, Inc. Neurogenesis via modulation of the muscarinic receptors
EP2377530A2 (en) 2005-10-21 2011-10-19 Braincells, Inc. Modulation of neurogenesis by PDE inhibition
EP2314289A1 (en) 2005-10-31 2011-04-27 Braincells, Inc. Gaba receptor mediated modulation of neurogenesis
US8039639B2 (en) 2006-01-31 2011-10-18 Array Biopharma Inc. Kinase inhibitors and methods of use thereof
JP2009525996A (en) * 2006-02-06 2009-07-16 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツング Aminoindazole derivatives
US7884126B2 (en) 2006-02-06 2011-02-08 Merck Patent Gmbh Indazole-heteroaryl derivatives
JP2009525969A (en) * 2006-02-06 2009-07-16 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツング Indazole-heteroaryl derivatives
WO2007090493A1 (en) * 2006-02-06 2007-08-16 Merck Patent Gmbh Indazole heteroaryl derivatives
EP2377531A2 (en) 2006-05-09 2011-10-19 Braincells, Inc. Neurogenesis by modulating angiotensin
EP2382975A2 (en) 2006-05-09 2011-11-02 Braincells, Inc. Neurogenesis by modulating angiotensin
WO2008009335A3 (en) * 2006-07-18 2008-02-28 Merck Patent Gmbh Aminoindazole urea derivatives
US8207210B2 (en) 2006-07-18 2012-06-26 Merck Patent Gmbh Aminoindazolylurea derivatives
WO2008009335A2 (en) * 2006-07-18 2008-01-24 Merck Patent Gmbh Aminoindazole urea derivatives
DE102007002717A1 (en) 2007-01-18 2008-07-24 Merck Patent Gmbh Heterocyclic indazole derivatives
WO2008086854A1 (en) 2007-01-18 2008-07-24 Merck Patent Gmbh 5-([1,3,4] oxadiazol-2-yl)-1h-indazol and 5-([1,3,4] thiadiazol-2-yl)-1h-indazol derivatives as sgk inhibitors for the treatment of diabetes
DE102007022565A1 (en) 2007-05-14 2008-11-20 Merck Patent Gmbh Heterocyclic indazole derivatives
JP2010526840A (en) * 2007-05-14 2010-08-05 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツング Heterocyclic indazole derivatives
US8673893B2 (en) 2007-07-20 2014-03-18 Nerviano Medical Sciences S.R.L. Substituted indazole derivatives active as kinase inhibitors
US9085558B2 (en) 2007-07-20 2015-07-21 Nerviano Medical Sciences S.R.L. Substituted indazole derivatives active as kinase inhibitors
US9102662B2 (en) 2007-07-20 2015-08-11 Nerviano Medical Sciences S.R.L. Substituted indazole derivatives active as kinase inhibitors
JP2011502959A (en) * 2007-07-20 2011-01-27 ネルビアーノ・メデイカル・サイエンシーズ・エツセ・エルレ・エルレ Substituted indazole derivatives active as kinase inhibitors
WO2009013126A1 (en) * 2007-07-20 2009-01-29 Nerviano Medical Sciences S.R.L. Substituted indazole derivatives active as kinase inhibitors
KR101567763B1 (en) * 2007-07-20 2015-11-12 네르비아노 메디칼 사이언시스 에스.알.엘. Substituted indazole derivatives active as kinase inhibitors
AU2008280283B2 (en) * 2007-07-20 2013-11-07 Nerviano Medical Sciences S.R.L. Substituted indazole derivatives active as kinase inhibitors
US9616059B2 (en) 2007-07-20 2017-04-11 Nerviano Medical Sciences S.R.L. Substituted indazole derivatives active as kinase inhibitors
US9255087B2 (en) 2007-07-20 2016-02-09 Nerviano Medical Sciences S.R.L. Substituted indazole derivatives active as kinase inhibitors
CN103923072B (en) * 2007-07-20 2017-05-31 内尔维阿诺医学科学有限公司 As the substituted indazole derivative with kinase inhibitory activity
CN101754956B (en) * 2007-07-20 2014-04-23 内尔维阿诺医学科学有限公司 Substituted indazole derivatives active as kinase inhibitors
EA018503B1 (en) * 2007-07-20 2013-08-30 НЕРВИАНО МЕДИКАЛ САЙЕНСИЗ С.р.л. Substituted indazole derivatives active as kinase inhibitors
US10081622B2 (en) 2007-07-20 2018-09-25 Nerviano Medical Sciences S.R.L. Substituted indazole derivatives active as kinase inhibitors
CN103923072A (en) * 2007-07-20 2014-07-16 内尔维阿诺医学科学有限公司 Substituted Indazole Derivatives Active As Kinase Inhibitors
US9029356B2 (en) 2007-07-20 2015-05-12 Nerviano Medical Sciences S.R.L. Substituted indazole derivatives active as kinase inhibitors
US8299057B2 (en) 2007-07-20 2012-10-30 Nerviano Medical Sciences S.R.L. Substituted indazole derivatives active as kinase inhibitors
WO2009147189A1 (en) * 2008-06-05 2009-12-10 Glaxo Group Limited Novel compounds
JP2011522002A (en) * 2008-06-05 2011-07-28 グラクソ グループ リミテッド Benzpyrazole derivatives as inhibitors of PI3 kinase
US8765743B2 (en) 2008-06-05 2014-07-01 Glaxosmithkline Intellectual Property Development Limited Compounds
JP2011522003A (en) * 2008-06-05 2011-07-28 グラクソ グループ リミテッド New compounds
US8658635B2 (en) 2008-06-05 2014-02-25 Glaxosmithkline Intellectual Property Development Limited Benzpyrazol derivatives as inhibitors of PI3 kinases
US8536169B2 (en) 2008-06-05 2013-09-17 Glaxo Group Limited Compounds
JP2011522004A (en) * 2008-06-05 2011-07-28 グラクソ グループ リミテッド New compounds
JP2011522001A (en) * 2008-06-05 2011-07-28 グラクソ グループ リミテッド 4-Carboxamide indazole derivatives useful as inhibitors of PI3 kinase
WO2010020305A1 (en) 2008-08-18 2010-02-25 Merck Patent Gmbh Oxadiazole derivatives for treating diabetes
DE102008038220A1 (en) 2008-08-18 2010-02-25 Merck Patent Gmbh oxadiazole
DE102008038221A1 (en) 2008-08-18 2010-02-25 Merck Patent Gmbh 7-azaindole derivatives
DE102008038222A1 (en) 2008-08-18 2010-02-25 Merck Patent Gmbh Indazol-5-carboxylic acid derivatives
WO2010064875A3 (en) * 2008-12-05 2010-09-10 Korea Institute Of Science And Technology Novel indazole derivatives or pharmaceutically acceptable salts thereof as protein kinase inhibitors for proliferative diseases treatment, and a pharmaceutical composition containing the same as an active ingredient
JP2012510989A (en) * 2008-12-05 2012-05-17 コリア・インスティテュート・オブ・サイエンス・アンド・テクノロジー Novel indazole derivatives that are inhibitors of protein kinases for the treatment of abnormal cell proliferative diseases, pharmaceutically acceptable salts thereof, and pharmaceutical compositions containing them as active ingredients
WO2010064875A2 (en) * 2008-12-05 2010-06-10 Korea Institute Of Science And Technology Novel indazole derivatives or pharmaceutically acceptable salts thereof as protein kinase inhibitors for proliferative diseases treatment, and a pharmaceutical composition containing the same as an active ingredient
US8754209B2 (en) 2008-12-05 2014-06-17 Korea Institute Of Science And Technology Indazole derivatives or pharmaceutically acceptable salts thereof as protein kinase inhibitors for proliferative diseases treatment, and a pharmaceutical composition containing the same as an active ingredient
WO2010069966A1 (en) * 2008-12-18 2010-06-24 Nerviano Medical Sciences S.R.L. Substituted indazole derivatives active as kinase inhibitors
JP2012512830A (en) * 2008-12-18 2012-06-07 ネルビアーノ・メデイカル・サイエンシーズ・エツセ・エルレ・エルレ Substituted indazole derivatives active as kinase inhibitors
WO2010099217A1 (en) 2009-02-25 2010-09-02 Braincells, Inc. Modulation of neurogenesis using d-cycloserine combinations
US8524751B2 (en) 2009-03-09 2013-09-03 GlaxoSmithKline Intellecutual Property Development 4-oxadiazol-2-YL-indazoles as inhibitors of P13 kinases
US8575162B2 (en) 2009-04-30 2013-11-05 Glaxosmithkline Intellectual Property Development Limited Compounds
US8586590B2 (en) 2009-04-30 2013-11-19 Glaxosmithkline Intellectual Property Development Limited Compounds
US10383879B2 (en) 2009-04-30 2019-08-20 Glaxo Group Limited Compounds
US10624898B2 (en) 2009-04-30 2020-04-21 Glaxo Group Limited Compounds
US8580797B2 (en) 2009-04-30 2013-11-12 Glaxo Smith Kline Intellectual Property Development Limited Compounds
US8586583B2 (en) 2009-04-30 2013-11-19 Glaxosmithkline Intellectual Property Development Limited Compounds
US8609657B2 (en) 2009-04-30 2013-12-17 Glaxosmithkline Intellectual Property Development Limited Compounds
US10946025B2 (en) 2009-04-30 2021-03-16 Glaxo Group Limited Compounds
WO2011063115A1 (en) 2009-11-19 2011-05-26 Braincells Inc. Combination of nootropic agent with one or more neurogenic or neurogenic sensitizing agents for stimulating or increasing neurogenesis
JP2013512879A (en) * 2009-12-03 2013-04-18 グラクソ グループ リミテッド Benzpyrazole derivatives as inhibitors of PI3 kinase
US11826365B2 (en) 2009-12-29 2023-11-28 Dana-Farber Cancer Institute, Inc. Type II raf kinase inhibitors
WO2011091033A1 (en) 2010-01-20 2011-07-28 Braincells, Inc. Modulation of neurogenesis by ppar agents
US8987275B2 (en) * 2010-03-16 2015-03-24 Dana-Farber Cancer Institute, Inc. Indazole compounds and their uses
JP2013522292A (en) * 2010-03-16 2013-06-13 デイナ ファーバー キャンサー インスティチュート,インコーポレイテッド Indazole compounds and their use
US20130184287A1 (en) * 2010-03-16 2013-07-18 Dana-Farber Cancer Institute, Inc. Indazole compounds and their uses
US8993576B2 (en) 2010-10-27 2015-03-31 Glaxo Group Limited 6-(1H-indol-4-yl)-4-(5-{[4-1-methylethyl)-1-piperazinyl]methyl}-1,3-oxazol-2-yl)-1H-indazole hemi succinate salt, polymorphs and pharmaceutical compositions thereof
US10981903B2 (en) 2011-11-17 2021-04-20 Dana-Farber Cancer Institute, Inc. Inhibitors of c-Jun-N-terminal kinase (JNK)
US10144730B2 (en) 2011-11-17 2018-12-04 Dana-Farber Cancer Institute, Inc. Inhibitors of c-Jun-N-terminal kinase (JNK)
WO2013100672A1 (en) * 2011-12-29 2013-07-04 제이더블유중외제약㈜ 3,6-disubstituted indazole derivative having protein kinase inhibiting activity
US10787436B2 (en) 2012-10-18 2020-09-29 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US10112927B2 (en) 2012-10-18 2018-10-30 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US10000483B2 (en) 2012-10-19 2018-06-19 Dana-Farber Cancer Institute, Inc. Bone marrow on X chromosome kinase (BMX) inhibitors and uses thereof
USRE48175E1 (en) 2012-10-19 2020-08-25 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged small molecules as inducers of protein degradation
US9758522B2 (en) 2012-10-19 2017-09-12 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged small molecules as inducers of protein degradation
US11040957B2 (en) 2013-10-18 2021-06-22 Dana-Farber Cancer Institute, Inc. Heteroaromatic compounds useful for the treatment of proliferative diseases
US10906889B2 (en) 2013-10-18 2021-02-02 Dana-Farber Cancer Institute, Inc. Polycyclic inhibitors of cyclin-dependent kinase 7 (CDK7)
WO2015112445A1 (en) * 2014-01-24 2015-07-30 Abbvie Inc. 6-phenyl- or 6-(pyridin-3-yl)indazole derivatives and methods of use
JP2017503838A (en) * 2014-01-24 2017-02-02 アッヴィ・インコーポレイテッド 6-Phenyl- or 6- (pyridin-3-yl) indazole derivatives and methods of use
US10561651B2 (en) 2014-02-20 2020-02-18 Ignyta, Inc. Methods for treating neuroblastoma
US10231965B2 (en) 2014-02-20 2019-03-19 Ignyta, Inc. Molecules for administration to ROS1 mutant cancer cells
US10682348B2 (en) 2014-02-20 2020-06-16 Ignyta, Inc. Molecules for administration to ROS1 mutant cancer cells
EP3834827A1 (en) 2014-02-20 2021-06-16 Ignyta, Inc. N-[5-(3,5-difluorobenzyl)-1h-indazol-3-yl]-4-(4-methyl-piperazin-1-yl)-2-(tetrahydro-2h-pyran-4-ylamino) benzamide for treating patients with ros1,ntrk1, ntrk2 and ntrk3 mutant cancer cells
US10017477B2 (en) 2014-04-23 2018-07-10 Dana-Farber Cancer Institute, Inc. Janus kinase inhibitors and uses thereof
US9862688B2 (en) 2014-04-23 2018-01-09 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged janus kinase inhibitors and uses thereof
US10357490B2 (en) 2014-12-02 2019-07-23 Ignyta, Inc. Combinations for the treatment of neuroblastoma
US10085979B2 (en) 2014-12-02 2018-10-02 Ignyta, Inc. Combinations for the treatment of neuroblastoma
WO2016096709A1 (en) 2014-12-16 2016-06-23 Eudendron S.R.L. Heterocyclic derivatives modulating activity of certain protein kinases
US10870651B2 (en) 2014-12-23 2020-12-22 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US10550121B2 (en) 2015-03-27 2020-02-04 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
US11325910B2 (en) 2015-03-27 2022-05-10 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
WO2016188214A1 (en) * 2015-05-27 2016-12-01 上海海和药物研究开发有限公司 Preparation and use of novel kinase inhibitor
CN106279119A (en) * 2015-05-27 2017-01-04 上海海和药物研究开发有限公司 The preparation of a kind of Azaindole kinase inhibitors and application thereof
CN106279119B (en) * 2015-05-27 2020-06-16 上海海和药物研究开发有限公司 Preparation and application of novel kinase inhibitor
US10702527B2 (en) 2015-06-12 2020-07-07 Dana-Farber Cancer Institute, Inc. Combination therapy of transcription inhibitors and kinase inhibitors
US11142507B2 (en) 2015-09-09 2021-10-12 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
WO2017045751A1 (en) * 2015-09-15 2017-03-23 Merck Patent Gmbh Compounds as asic inhibitors and uses thereof
JP2018531266A (en) * 2015-10-22 2018-10-25 セルビタ エス.エー. Pyridone derivatives and their use as kinase inhibitors
US10869864B2 (en) 2015-12-18 2020-12-22 Ignyta, Inc. Combinations for the treatment of cancer
US10398693B2 (en) 2017-07-19 2019-09-03 Ignyta, Inc. Pharmaceutical compositions and dosage forms
US11253515B2 (en) 2017-07-19 2022-02-22 Ignyta, Inc. Pharmaceutical compositions and dosage forms
US11007191B2 (en) 2017-10-17 2021-05-18 Ignyta, Inc. Pharmaceutical compositions and dosage forms
WO2020163812A1 (en) 2019-02-08 2020-08-13 Frequency Therapeutics, Inc. Valproic acid compounds and wnt agonists for treating ear disorders

Also Published As

Publication number Publication date
AU2002353186A1 (en) 2003-06-30

Similar Documents

Publication Publication Date Title
WO2003051847A1 (en) (1-h-indazol-3-yl) -amide derivatives as gsk-3 inhibitors
US7105532B2 (en) Pyrazolo[3,4-c]pyridines as gsk-3 inhibitors
WO2003068773A1 (en) Pyrazolopyridine derivatives
WO2003045949A1 (en) Pyrazolopyridine derivatives
CA2673368C (en) 3-benzofuranyl-4-indolyl maleimides as potent gsk3 inhibitors for neurogenerative disorders
JP2002527419A (en) Pyrrole-2,5-diones as GSK-3 inhibitors
KR20160134865A (en) Amide derivatives and pharmaceutically acceptable salts thereof, preparation method therefor and medicinal application thereof
KR20090052884A (en) Pyrimidone compounds as gsk-3 inhibitors
WO2003080616A1 (en) Pyrazolopyridazine derivatives, process for preparation and use for the inhibition of gsk-3
KR20170018913A (en) Inhibitors of lysine specific demethylase-1
JP2016014061A (en) 1,2-disubstituted heterocyclic compound
US20040019052A1 (en) Pyrazolopyridines and pyrazolopyridazines as antidabetics
JP2009538309A (en) Thiophenecarboxamides useful as inhibitors of protein kinases
WO2014137723A1 (en) Compounds inhibiting leucine-rich repeat kinase enzyme activity
JP2009544625A (en) Benzothiophene inhibitors of RHO kinase
JP2006504711A (en) Indazole carboxamide derivatives, processes for their preparation and use as CDK1, CDK2 and CDK4 inhibitors
US20100256133A1 (en) Novel compounds having indazole frameworks, methods for preparing the same and pharmaceutical composition comprising the same
TW201331194A (en) Heteroaryls and uses thereof
AU2014234906B2 (en) Cycloalkyl nitrile pyrazolo pyridones as Janus kinase inhibitors
JP2009533452A (en) Thiophene-carboxamides useful as inhibitors of protein kinases
JP2009538305A (en) Thiophenecarboxamides useful as inhibitors of protein kinases
AU2017373758B2 (en) Compounds and pharmaceutical compositions for modulating SGK activity, and methods thereof
WO2003080609A1 (en) Pyrazolopyrimidines derivatives
CA2726666A1 (en) Derivatives of 2-oxo-alkyl-1-piperazin-2-one, preparation method thereof and therapeutic use of same
WO2003080617A1 (en) Pyrazolopyrimidine derivatives

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SK SL TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR IE IT LU MC NL PT SE SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP