WO2014026327A1 - 4-heteroaryl substituted benzoic acid compounds as rorgammat inhibitors and uses thereof - Google Patents

4-heteroaryl substituted benzoic acid compounds as rorgammat inhibitors and uses thereof Download PDF

Info

Publication number
WO2014026327A1
WO2014026327A1 PCT/CN2012/080131 CN2012080131W WO2014026327A1 WO 2014026327 A1 WO2014026327 A1 WO 2014026327A1 CN 2012080131 W CN2012080131 W CN 2012080131W WO 2014026327 A1 WO2014026327 A1 WO 2014026327A1
Authority
WO
WIPO (PCT)
Prior art keywords
chloro
trifluoromethyl
indazol
benzoyl
alkyl
Prior art date
Application number
PCT/CN2012/080131
Other languages
French (fr)
Inventor
Kenneth Jay BARR
Corey Beinstock
John Maclean
Hongjun Zhang
Richard Thomas Beresis
Dongshan Zhang
Original Assignee
Merck Sharp & Dohme Corp.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Merck Sharp & Dohme Corp. filed Critical Merck Sharp & Dohme Corp.
Priority to PCT/CN2012/080131 priority Critical patent/WO2014026327A1/en
Priority to AU2013302725A priority patent/AU2013302725A1/en
Priority to US14/421,048 priority patent/US9745265B2/en
Priority to CA2881688A priority patent/CA2881688A1/en
Priority to PCT/US2013/054887 priority patent/WO2014028589A2/en
Priority to CN201380053463.8A priority patent/CN104853757A/en
Priority to BR112015003188A priority patent/BR112015003188A2/en
Priority to EP13829282.6A priority patent/EP2884981B1/en
Priority to RU2015108894A priority patent/RU2015108894A/en
Priority to MX2015001941A priority patent/MX2015001941A/en
Priority to KR1020157006322A priority patent/KR20150042266A/en
Priority to JP2015527566A priority patent/JP6422438B2/en
Publication of WO2014026327A1 publication Critical patent/WO2014026327A1/en
Priority to IN1156DEN2015 priority patent/IN2015DN01156A/en
Priority to US15/647,437 priority patent/US10196354B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/10Indoles; Hydrogenated indoles with substituted hydrocarbon radicals attached to carbon atoms of the hetero ring
    • C07D209/18Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D209/26Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals with an acyl radical attached to the ring nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/4161,2-Diazoles condensed with carbocyclic ring systems, e.g. indazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/54Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings condensed with carbocyclic rings or ring systems
    • C07D231/56Benzopyrazoles; Hydrogenated benzopyrazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • 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
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • 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
    • C07D405/06Heterocyclic 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 linked by a carbon chain containing only aliphatic carbon atoms
    • 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
    • C07D405/12Heterocyclic 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 linked by a chain containing hetero atoms as chain links
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • naive T helper cells Upon activation by antigen-presenting cells naive T helper cells undergo clonal expansion and will ultimately differentiate in cytokine secreting effector T cells, such as Thl and Th2 subtypes.
  • cytokine secreting effector T cells such as Thl and Th2 subtypes.
  • Thl7 Upon activation by antigen-presenting cells naive T helper cells undergo clonal expansion and will ultimately differentiate in cytokine secreting effector T cells, such as Thl and Th2 subtypes.
  • This effector T helper cell subset can be distinguished based on its ability to produce large quantities of IL-17/F, IL-21 and IL-22, and is named Thl7 (Miossec et al, New Eng. J. Med. 2361 : 888-898, 2009).
  • Thl and Th2 effector cells express Tbet and GATA3, respectively.
  • a Thymocyte/T cell specific variant of Retinoic Acid Receptor-related Orphan Receptor (ROR), RORgammaT is highly expressed in Thl7 cells (He et al, Immunity 9: 797-806, 1998).
  • RORgammaT belongs to the nuclear hormone receptor superfamily (Hirose et al, Biochem. Biophys. Res. Comm. 205: 1976-1983, 1994). RORgammaT is a truncated form of
  • RORgamma lacking the first N-terminal 21 amino acids and is, in contrast to RORgamma which is expressed in multiple tissues (heart, brain, kidney, lung, liver and muscle), exclusively expressed in cells of the lymphoid lineage and embryonic lymphoid tissue inducers (Sun et al, Science 288: 2369-2372, 2000; Eberl et al, Nat Immunol. 5: 64-73, 2004).
  • RORgammaT was shown to play a crucial role in non-Thl7 lymphoid cells.
  • RORgammaT was critically important in innate lymphoid cells expressing Thyl, SCA-1 and IL-23R proteins. Genetic disruption of RORgamma in a mouse colitis model dependent on these innate lymphoid cells, prevented colitis development (Buonocore et al, Nature 464: 1371-1375, 2010).
  • RORgammaT was shown to play a crucial role in other non-Thl7 cells, such as mast cells (Hueber et al, J. Immunol. 184: 3336-3340, 2010).
  • RORgammaT has been identified as a key mediator in the pathogenesis of several diseases (Louten et al, J. Allergy Clin. Immunol. 123 : 1004-1011, 2009; Annuziato et al, Nat. Rev. Rheumatol. 5: 325-331, 2009). This was confirmed using several disease models
  • autoimmune diseases Genetic ablation of the RORgamma gene in mice prevented the development of experimental autoimmune diseases, such as experimental autoimmune encephalomyelitis (EAE) and colitis (Ivanov et al, Cell 126: 1121-33, 2006; Buonocore et al, Nature 464: 1371-1375, 2010).
  • EAE experimental autoimmune encephalomyelitis
  • colitis Ivanov et al, Cell 126: 1121-33, 2006; Buonocore et al, Nature 464: 1371-1375, 2010.
  • antagonism of the transcriptional activity of RORgammaT is expected to have a beneficial effect on
  • autoimmune diseases such as, but not limited to rheumatoid arthritis, psoriasis, multiple sclerosis, inflammatory bowel disease, Crohn's disease, and asthma (Annunziato et al, Nat. Rev. Immunol. 5: 325-331, 2009; Louten et al, J. Allergy Clin. Immunol. 123: 1004-1011, 2009).
  • Antagonism of RORgammaT may also be beneficial in other diseases, which are characterized by increased levels of Thl7 cells and/or elevated levels of Thl7 hallmark cytokines such as IL-17, IL-22 and IL-23. Examples of such diseases are Kawasaki Disease (Jia et al, Clin. Exp. Immunol.
  • agonists include T0901317 and SR1078 (Wang et al, ACS Chem. Biol. 5: 1029-1034, 2010).
  • antagonist have been reported such as 7-oxygenated sterols (Wang et al, J. Biol. Chem. 285: 5013-5025, 2009) and compounds described in EP2181710 Al .
  • psoriasis One exemplary immue disorder in need of better therapy is psoriasis.
  • Various therapeutics have been developed in an attempt to treat psoriasis.
  • the present invention provides compounds which alter the interaction of coregulator proteins with RORgammaT and thereby antagonize RORgammaT-mediated transcriptional ativity, their use for the treatment of RORgammaT-mediated diseases or conditions, in particular autoimmune diseases and inflammatory diseases, as well as pharmaceutical compositions comprising such compounds and pharmaceutical carriers.
  • the present invention provides a compound according to Formula I
  • Y is CH, CR a , or N;
  • n 0, 1, 2, 3 or 4;
  • a 4 is CR 4 or N
  • a 5 is CR 5 or N
  • a 6 is CR 6 or N
  • a 7 is CR 7 or N
  • R a is (Ci -4 )alkyl or (C 3 - 7 )cycloalkyl
  • R 2 is hydroxycarbonyl, hydroxycarbonyl(Ci-io)alkyl, (Ci-io)alkylsulfoxyaminocarbonyl, or carbamoyl;
  • R 3 is hydrogen, halogen, cyano, nitro, hydroxy, (Ci -4 )alkylcarbonyloxy, (Ci -4 ) alkylsulfonylamino, (Ci -4 ) alkylcarbonylamino, (Co -4 ) alkylamino, (Ci -4 )alkyl, or (Ci_ 4 )alkoxy, wherein (Ci -4 )alkyl and (Ci -4 )alkoxy are optionally substituted with one or more halogen;
  • R 4 -R 7 independently are hydrogen, halogen, amino, cyano, hydroxy, (Ci_3)alkoxy, (Ci_ 4 )alkyl, (Co-io)alkylaminocarbonyl, (Co-6)alkyoxycarbonylamino, (di)(Ci_
  • (iv) (C 3 -5)heterocycloalkylcarbonyl, optionally substituted with one or more groups selected from halogen, amino, amino(Ci -4 )alkyl, cyano, nitro, hydroxyl, oxo ( 0), H 2 NC(0), (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci -4 )alkyl or (Ci_ 3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci -4 )alkyl and (Ci-3)alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
  • (v) (C 3 -5)heterocycloalkylamino, optionally substituted with one or more groups selected from halogen, amino, amino(Ci-4)alkyl, cyano, nitro, hydroxyl, oxo ( 0), H 2 NC(0), (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci- 4 )alkyl or (Ci_ 3)alkoxy, wherein (Ci -3 )alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci -4 )alkyl and (Ci -3 )alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
  • (vi) (C 3- 5)cycloalkylaminocarbonyl, optionally substituted with one or more groups selected from halogen, amino, amino(Ci -4 )alkyl, cyano, nitro, hydroxyl, oxo ( 0), H 2 NC(0), (Ci -3 )alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci -4 )alkyl or (Ci_ 3)alkoxy, wherein (Ci -3 )alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci -4 )alkyl and (Ci -3 )alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
  • (vii) (C 3- 5)cycloalkylcarbonylamino, optionally substituted with one or more groups selected from halogen, amino, amino(Ci -4 )alkyl, cyano, nitro, hydroxyl, oxo ( 0), H 2 NC(0), (Ci -3 )alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci -4 )alkyl or (Ci_ 3)alkoxy, wherein (Ci -3 )alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci -4 )alkyl and (Ci -3 )alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
  • x is 0, 1, 2, 3, 4 or 5.
  • a first subset of the invention is a compound wherein Y is N.
  • first subset of the fourth embodiment is a compound wherein Y is N. In a first subset of the first embodiment is a compound having Formula Ie
  • a 4 , A 5 , A 6 , A 7 are selected from the group consisting of: (i) CR 4 , CR 5 , CR 6 , CR 7 ; (ii) N, CR 5 , CR 6 , CR 7 ; (iii) CR 4 , N, CR 6 , CR 7 ; (iv) CR 4 , CR 5 , N, CR 7 ; (v) CR 4 , CR 5 , CR 6 , N; (vi) N, N, CR 6 ,CR 7 ; (vii) CR 4 , N, N, CR 7 ; (viii) CR 4 , CR 5 , N, N; (ix) N, CR 5 , N, CR 7 ; (x) CR 4 , N, CR 6 , N; and (xi) N, CR 5 , CR 6 , N.
  • a 4 , A 5 , A 6 , A 7 are selected from the group consisting of: (i) CR 4 , CR 5 , CR 6 , CR 7 ; (ii) N, CR 5 , CR 6 , CR 7 ; and (iii) CR 4 , N, CR 6 , CR 7 .
  • a seventh subset is a compound wherein A 4 , A 5 , A 6 , A 7 is (i) CR 4 , CR 5 , CR 6 , CR 7 , or (ii) N, CR 5 , CR 6 , CR 7 ; and Y is N.
  • R 1 is (i) (C 2- 9)heteroaryl, or (ii) (C 6 -i 4 )aryl, optionally substituted with one, two, three, four or five R 8 .
  • R 1 is (C 6 -i 4 )aryl, optionally substituted with one or two R 8 .
  • R 1 is phenyl, optionally substituted with one or two R 8 .
  • a twelfth subset is a compound wherein R 2 is C(0)OH.
  • a thirteenth subset is a compoundwherein R 6 is
  • the invention also relates to those compounds wherein all specific definitions for A 1 through A 4 , R 1 through R 8 , R a , Y, m, n and x and all substituent groups in the various aspects of the inventions defined here above occur in any combination within the definition of the compound of Formula I.
  • Non-limiting examples of the compounds of the present invention include:
  • alkyl refers to an aliphatic hydrocarbon group having one of its hydrogen atoms replaced with a bond having the specified number of carbon atoms.
  • an alkyl group contains, for example, from 1 to 6 carbon atoms (Ci- C 6 alkyl) or from 1 to 3 carbon atoms (C1-C3 alkyl).
  • alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, neopentyl, isopentyl, n-hexyl, isohexyl and neohexyl.
  • an alkyl group is linear. In another embodiment, an alkyl group is branched.
  • alkyl includes both branched- and straight-chain saturated aliphatic hydrocarbon groups, including all isomers, having the specified number of carbon atoms; for example, “Ci-6 alkyl” (or “Ci-C 6 alkyl”) includes all of the hexyl alkyl and pentyl alkyl isomers as well as n-, iso-, sec- and t-butyl, n- and isopropyl, ethyl and methyl.
  • Alkylene refers to both branched- and straight-chain saturated aliphatic hydrocarbon groups, including all isomers, having the specified number of carbons, and having two terminal end chain attachments; for example, the term “A-C 4 alkylene-B” represents, for example, A-CH 2 - CH 2 -CH 2 -CH 2 -B, A-CH 2 -CH 2 -CH(CH 3 )-CH 2 -B, A-CH 2 -CH(CH 2 CH 3 )-B, A-CH 2 - C(CH 3 )(CH 3 )-B, and the like.
  • Alkoxy represents a linear or branched alkyl group of indicated number of carbon atoms attached through an oxygen bridge; for example “C1-C6 alkoxy” includes -OCH 3 , -OCH 2 CH 3 , -OCH(CH 3 ) 2 , -0(CH 2 ) 5 CH 3 , and the like.
  • alkyl groups are unsubstituted or substituted with 1 to 3 substituents on each carbon atom, with halo, Ci-C 20 alkyl, CF 3 , NH 2 , N(Ci-C 6 alkyl) 2 , N0 2 , oxo, CN, N 3 , -OH, -0(Ci-C 6 alkyl), C 3 - C10 cycloalkyl, (C 3- 7)cycloalkyl, (C 3- 5)heterocycloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, (Co-C 6 alkyl) S(0)o-2-, (C 0 -C 6 alkyl)S(0) 0 - 2 (Co-C 6 alkyl)-, (C 0 -C 6 alkyl)C(0)NH-, H 2 N-C(NH)-,
  • alkenyl means a straight or branched carbon chain having the specified number of carbon atoms with at least one carbon-carbon double bond.
  • alkenyl include, but are not limited to, vinyl, allyl, isopropenyl, pentenyl, hexenyl, heptenyl, 1- propenyl, 2-butenyl, 2-methyl-2-butenyl, 2,4-hexadienyl, and the like.
  • alkynyl means a straight or branched carbon chain having the specified number of carbon atoms with at least one carbon-carbon triple bond.
  • alkynyl include, but are not limited to ethynyl, propargyl, 1-propynyl, 2-butynyl, and the like.
  • carbocycle (and variations thereof such as “carbocyclic” or “carbocyclyl”) as used herein, unless otherwise indicated, refers to (i) a C 3 to C 8 monocyclic, saturated or unsaturated ring or (ii) a C 7 to C 12 bicyclic saturated or unsaturated ring system. Each ring in (ii) is either attached via a bond to, or fused (including spirofused) to, the other ring, and each ring is saturated or unsaturated.
  • the carbocycle may be attached to the rest of the molecule at any carbon atom which results in a stable compound.
  • Saturated carbocyclics form a subset of carbocycles in which the entire ring system (mono- or polycyclic) is saturated.
  • Saturated monocyclic carbocyclic rings are also referred to as cycloalkyl rings, e.g., cyclopropyl, cyclobutyl, etc.
  • the fused bicyclic carbocycles are a further subset of the carbocycles in which a C 7 to C 10 bicyclic ring system in which each ring is saturated or unsaturated and two adjacent carbon atoms (or in the case of spirofused, one carbon atom) are shared by each of the rings in the ring system.
  • a saturated bicyclic carbocycle is one in which both rings are saturated.
  • An unsaturated bicyclic carbocycle is one in which one ring is unsaturated and the other is unsaturated or saturated. Unless otherwise noted, carbocycle is unsubstituted or substituted with C 1-6 alkyl, C 1-6 alkenyl, C 1-6 alkynyl, aryl, halogen, H 2 or OH.
  • a subset of the fused bicyclic unsaturated carbocycles are those bicyclic carbocycles in which one ring is a benzene ring and the other ring is saturated or unsaturated, with attachment via any carbon atom that results in a stable compound.
  • Aromatic carbocycles form another subset of the carbocycles.
  • aryl refers to aromatic mono- and poly-carbocyclic ring systems in which the individual carbocyclic rings in the polyring systems are fused or attached to each other via a single bond. Suitable aryl groups include phenyl, naphthyl, and biphenyl.
  • cycloalkyl means a cyclic ring of an alkane having the specified total ring carbon atoms; for example cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl.
  • heterocycle (and variations thereof such as “heterocyclic” or
  • heterocyclyl broadly refers to (i) a stable 4- to 8-membered, saturated or unsaturated monocyclic ring, or (ii) a stable 7- to 12-membered bicyclic ring system, wherein each ring in (ii) is either attached via a bond to, or fused (including spirofused) to, the other ring, and each ring is saturated or unsaturated, and the monocyclic ring or bicyclic ring system contains one or more heteroatoms (e.g., from 1 to 6 heteroatoms, or from 1 to 4 heteroatoms) selected from N, O and S and a balance of carbon atoms (the monocyclic ring typically contains at least one carbon atom and the ring systems typically contain at least two carbon atoms); and wherein any one or more of the nitrogen and sulfur heteroatoms is optionally oxidized, and any one or more of the nitrogen heteroatoms is optionally quaternized.
  • heteroatoms e.g., from 1 to 6 heteroatoms, or
  • the heterocyclic ring may be attached at any heteroatom or carbon atom, provided that attachment results in the creation of a stable structure.
  • the heterocyclic ring has substituents, it is understood that the substituents may be attached to any atom in the ring, whether a heteroatom or a carbon atom, provided that a stable chemical structure results.
  • saturated heterocyclics form a subset of the heterocycles; i.e., the term “saturated heterocyclic” generally refers to a heterocycle as defined above in which the entire ring system (whether mono- or poly-cyclic) is saturated.
  • saturated heterocyclic ring refers to a 4- to 8-membered saturated monocyclic ring or a stable 7- to 12-membered bicyclic ring system which consists of carbon atoms and one or more heteroatoms selected from N, O and S.
  • Representative examples include piperidinyl, piperazinyl, azepanyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, oxazolidinyl, isoxazolidinyl, morpholinyl, thiomorpholinyl, thiazolidinyl, isothiazolidinyl, 1,4-dioxanyl, 1,4-thioxanyl,
  • Heteroaromatics form another subset of the heterocycles; i.e., the term “heteroaromatic” (alternatively “heteroaryl”) generally refers to a heterocycle as defined above in which the entire ring system (whether mono- or poly-cyclic) is an aromatic ring system.
  • heteroaryl generally refers to a heterocycle as defined above in which the entire ring system (whether mono- or poly-cyclic) is an aromatic ring system.
  • heteroaryl refers a 5- or 6-membered monocyclic aromatic ring or a 7- to 12- membered bicyclic aromatic ring, and which consists of carbon atoms and one or more heteroatoms selected from N, O and S.
  • substituted heteroaryl rings containing at least one nitrogen atom e.g., pyridine
  • substitutions can be those resulting in N-oxide formation.
  • monocyclic heteroaromatic rings include pyridyl, pyrrolyl, pyrazinyl, pyrimidinyl, pyridazinyl, thienyl (or thiophenyl), thiazolyl, furanyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, and thiadiazolyl.
  • bicyclic heteroaromatic rings include
  • heterocycles Another subset of heterocycles is unsaturated heterocycles in which one or both rings are unsaturated (provided the entire ring system is not aromatic).
  • unsaturated heterocycles include dihydrofuranyl, dihydrothienyl, dihydropyranyl,
  • dihydroimidazolyl indolinyl, isoindolinyl, chromanyl, isochromanyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, tetrahydronaphthyridinyl, 2,3-dihydrobenzofuranyl, 1,4- benzoxazinyl, 1,3-benzoxazolinyl, 2,3-dihydrobenzo-l,4-dioxinyl (i.e., eft O ), and benzo-
  • Cc°> 1,3-dioxolyl (i.e., Cc°> ).
  • C °> is alternatively referred to as phenyl having as a substituent methylenedioxy attached to two adjacent carbon atoms.
  • groups such as chromone and coumarin.
  • cycloalkyl Unless otherwise specifically noted as only unsubstituted or only substituted, cycloalkyl, cycloalkenyl, heterocycloalkyl, aryl (including phenyl) and heteroaryl groups are
  • substituents for substituted or optionally substituted cycloalkyl, heterocycloalkyl, cycloalkenyl, aryl (including phenyl, and as an isolated substituent or as part of a substituent such as in aryloxy and aralkyl), heteroaryl (as an isolated substituent or as part of a substituent such as in heteroaryloxy and heteroaralkyl) are one to three groups independently selected from halogen (or halo), C1-C5 alkyl optionally substituted with one to five fluorine, H 2 , N(Ci-C 6 alkyl) 2 , N0 2 , oxo, CN, N 3 , -OH, -0(Ci-C 6 alkyl) optionally substituted with one to five fluorine, C 3 -Cio cycloalkyl, (C 3- 7)
  • halogen refers to fluorine, chlorine, bromine and iodine
  • fluoro F
  • chloro CI
  • bromo Br
  • iodo I
  • haloalkyl means alkyl having the specified number of carbon atoms in which from one to all of the hydrogen atoms have been replaced by a halogen atom.
  • aralkyl and “heteroaralkyl” refer to an aryl/heteroaryl linked to the rest of the molecule via a Ci to C 4 alkylene.
  • Co as employed in expressions such as "Co-6 alkylene” means a direct covalent bond; or when employed in experessions such as "Co-6 alkyl” means hydrogen.
  • an integer defining the presence of a certain number of atoms in a group is equal to zero, it means that the atoms adjacent thereto are connected directly by a bond; for example, in the structure , wherein s is an integer equal to zero, 1 or 2, the structure is when s is zero; or it means that the indicated atom is absent; for example -S(0)o- means -S-.
  • an “unsaturated” ring is a partially or fully unsaturated ring.
  • an “unsaturated monocyclic C 6 carbocycle” refers to cyclohexene, cyclohexadiene, and benzene. Unless expressly stated to the contrary, all ranges cited herein are inclusive.
  • a heterocycle described as containing from “ 1 to 4 heteroatoms” means the heterocycle can contain 1, 2, 3 or 4 heteroatoms.
  • variable definition on each occurrence is independent of its definition at every other occurrence. Also, combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
  • variable definitions containing terms having repeated terms e.g., (CRiRj) r , where r is the integer 2, Ri is a defined variable, and Rj is a defined variable
  • the value of Ri may differ in each instance in which it occurs, and the value of Rj may differ in each instance in which it occurs.
  • Ri and Rj are independently selected from the group consisting of methyl, ethyl, propyl and butyl, then (CRiRj) 2 can be
  • (Ci-6)alkyl as used hereinabove means a branched or unbranched alkyl group having 1-6 carbon atoms, for example methyl, ethyl, propyl, isopropyl, butyl, tert-butyl, n- pentyl and n-hexyl. Preferred is (Ci-4)alkyl.
  • (Ci-5)alkyl means a branched or unbranched alkyl group having 1-5 carbon atoms, for example methyl, ethyl, propyl, isopropyl, butyl, tert-butyl and n-pentyl.
  • (Ci- 4 )alkyl as used herein means a branched or unbranched alkyl group having 1-4 carbon atoms, being methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl and tert-butyl.
  • (Ci-3)alkoxy means an alkoxy group having 1-3 carbon atoms, the alkyl moiety being branched or unbranched.
  • (Ci-3)alkoxycarbonyl means an alkoxycarbonyl group having 1-3 carbon atoms in the alkoxy moiety, the alkoxy moiety having the same meaning as previously defined.
  • (di)(Ci-6)alkylaminocarbonyl means an alkylaminocarbonyl group, the amino group of which is monosubstituted or disubstituted independently with an alkyl group which contains 1-6 carbon atoms and which has the same meaning as previously defined.
  • Preferred alkyl group is (Ci-4)alkyl.
  • (C 3 -7)cycloalkyl means a cycloalkyl group having 3-7 carbon atoms, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl. 5-6 Carbon atoms are preferred.
  • (C 3 -5)heterocycloalkyl means a heterocycloalkyl group having 3-5 carbon atoms, including 1-3 heteroatoms selected from N, O and/or S, which may be attached via a nitrogen if feasible, or a carbon atom.
  • Preferred number of heteroatoms is one or two. Most preferred number is one.
  • Preferred heteroatoms are N or O. Most preferred are piperazinyl, tetrahydropyranyl, morpholinyl and pyrrolidinyl.
  • a roup having the for ° means a heterocyclocarbonyl group such as , , and 5 each optionally substituted with one or more (Ci_ io)alkyl, halogen, amino, cyano, hydroxy, and (Ci_3)alkoxy.
  • (C 2 -9)heteroaryl means an aromatic group having 2-9 carbon atoms and 1-3 heteroatoms selected from N, O and S, like imidazolyl, thiadiazolyl, pyridinyl, pyrimidinyl, thiophenyl or furyl, pyrazolyl, isoxazolyl or quinolyl. Preferred number of heteroatoms is one or two. Preferred heteroaryl groups are pyrazolyl, thiophenyl, isoxazolyl, pyridyl and quinolyl.
  • the (C 2 -5)heteroaryl group may be attached via a carbon atom or a nitrogen, if feasible.
  • (C6-i 4 )aryl means an aromatic hydrocarbon group having 6-14 carbon atoms, such as phenyl, naphthyl, tetrahydronaphthyl, indenyl, anthracyl, More preferred are (C 6- io)aryl groups.
  • the most preferred aromatic hydrocarbon group is phenyl.
  • X a -Xb shall have the same meaning as the term "X a -b", wherein X is any atom and a and b are any integers.
  • X is any atom and a and b are any integers.
  • Ci-C 4 shall have the same meaning as “C 1-4 ".
  • a x shall have the same meaning, and be interchangeable with, “AX”, wherein “A” is any atom and “x” or “X” are any integer.
  • R 1 shall have the same meaning, and be
  • (Cl-4)alkylcarbonyloxy refers to, e.g. O
  • substituted means that one or more hydrogens on the designated atom/atoms is/are replaced with a selection from the indicated group, provided that the designated atom's normal valency under the existing circumstances is not exceeded, and that the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
  • Stable compound or “stable structure” is defined as a compound or structure that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
  • substituent and/or variable when referring to a substituent and/or variable means that one or more hydrogens on the designated atom/atoms is/are replaced with a selection from the indicated group, provided that the designated atom's normal valency under the existing circumstances is not exceeded, and that the substitution results in a stable compound.
  • salt is intended to equally apply to the salt, solvate, ester and prodrug of enantiomers, stereoisomers, rotamers, tautomers, positional isomers, racemates or prodrugs of the inventive compounds.
  • effective amount refers to an amount of the compound of Formula (I) and/or an additional therapeutic agent, or a composition thereof, that is effective in producing the desired therapeutic, ameliorative, inhibitory or preventative effect when administered to a subject suffering from an RORgammaT-mediated disease or disorder.
  • effective amount can refer to each individual agent or to the combination as a whole, wherein the amounts of all agents administered are together effective, but wherein the component agent of the combination may not be present individually in an effective amount.
  • a "subject” is a human or non-human mammal. In one embodiment, a subject is a human. In another embodiment, a subject is a chimpanzee.
  • the compounds of this invention include the prodrugs, hydrates or solvates of the compounds.
  • the compounds of Formula I may contain asymmetric or chiral centers, and, therefore, exist in different stereoisomeric forms. It is intended that all stereoisomeric forms of the compounds of Formula (I) as well as mixtures thereof, including racemic mixtures, form part of the present invention.
  • the present invention embraces all geometric and positional isomers. For example, if a compound of Formula (I) incorporates a double bond or a fused ring, both the cis- and trans-forms, as well as mixtures, are embraced within the scope of the invention.
  • Compounds described herein may contain an asymmetric center and may thus exist as enantiomers. Where the compounds according to the invention possess two or more asymmetric centers, they may additionally exist as diastereomers.
  • the present invention includes all such possible stereoisomers as substantially pure resolved enantiomers, racemic mixtures thereof, as well as mixtures of diastereomers.
  • the above Formula I is shown without a definitive stereochemistry at certain positions.
  • the present invention includes all stereoisomers of Formula I and pharmaceutically acceptable salts thereof.
  • Diastereoisomeric pairs of enantiomers may be separated by, for example, fractional crystallization from a suitable solvent, and the pair of enantiomers thus obtained may be separated into individual stereoisomers by conventional means, for example by the use of an optically active acid or base as a resolving agent or on a chiral UPLC column. Further, any enantiomer or diastereomer of a compound of the general Formula I may be obtained by stereospecific synthesis using optically pure starting materials or reagents of known configuration.
  • Diastereomeric mixtures can be separated into their individual diastereomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as, for example, by chromatography and/or fractional crystallization.
  • Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g. chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereomers and converting (e.g.
  • All stereoisomers for example, geometric isomers, optical isomers and the like
  • of the present compounds including those of the salts, solvates, esters and prodrugs of the compounds as well as the salts, solvates and esters of the prodrugs, such as those which may exist due to asymmetric carbons on various substituents, including enantiomeric forms (which may exist even in the absence of asymmetric carbons), rotameric forms, atropisomers, and diastereomeric forms, are contemplated within the scope of this invention, as are positional isomers.
  • Individual stereoisomers of the compounds of the invention may, for example, be substantially free of other isomers, or may be admixed, for example, as racemates or with all other, or other selected, stereoisomers.
  • the chiral centers of the present invention can have the S or R configuration as defined by the IUPAC 1974 Recommendations.
  • salts derived from such inorganic bases include aluminum, ammonium, calcium, copper (ic and ous), ferric, ferrous, lithium, magnesium, manganese (ic and ous), potassium, sodium, zinc and the like salts. Preferred are the ammonium, calcium, magnesium, potassium and sodium salts.
  • Salts prepared from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines derived from both naturally occurring and synthetic sources.
  • organic non-toxic bases from which salts can be formed include, for example, arginine, betaine, caffeine, choline, N,N'-dibenzyl- ethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol,
  • ethanolamine ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, dicyclohexylamine, lysine,
  • the compound of the present invention When the compound of the present invention is basic, its corresponding salt can be conveniently prepared from pharmaceutically acceptable non-toxic inorganic and organic acids.
  • Such acids include, for example, acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p-toluenesulfonic acid and the like.
  • Preferred are citric, hydrobromic, hydrochloric, maleic, phosphoric, sulfuric, and tartaric acids.
  • the compounds of Formula I can form salts which are also within the scope of this invention.
  • Reference to a compound of Formula I herein is understood to include reference to salts thereof, unless otherwise indicated.
  • pharmaceutically acceptable salt represents those salts which are, within the scope of medical judgement, suitable for use in contact for the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are
  • salts are well known in the art. They may be obtained during the final isolation and purification of the compounds of the invention, or separately by reacting the free base function with a suitable mineral acid such as hydrochloric acid, phosphoric acid, or sulfuric acid, or with an organic acid such as for example ascorbic acid, citric acid, tartaric acid, lactic acid, maleic acid, malonic acid, fumaric acid, glycolic acid, succinic acid, propionic acid, acetic acid, methanesulfonic acid, and the like.
  • the acid function can be reacted with an organic or a mineral base, like sodium hydroxide, potassium hydroxide, calcium hydroxide, calcium carbonate, ammonium (e.g. diethylamine) or lithium hydroxide.
  • solvates of compounds of Formula I.
  • the term "solvate” refers to a complex of variable stoichiometry formed by a solute (i.e., a compound of Formula I) or a pharmaceutically acceptable salt thereof and a solvent that does not interfere with the biological activity of the solute.
  • solvents include, but are not limited to water, ethanol, and acetic acid.
  • the solvent is water, the solvate is known as hydrate; hydrate includes, but is not limited to, hemi-, mono, sesqui-, di- and trihydrates.
  • the compounds of the invention may form hydrates or solvates. It is known to those of skill in the art that charged compounds form hydrated species when lyophilized with water, or form solvated species when concentrated in a solution with an appropriate organic solvent.
  • One or more compounds of the invention may exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like, and it is intended that the invention embrace both solvated and unsolvated forms.
  • “Solvate” may also mean a physical association of a compound of this invention with one or more solvent molecules. This physical association involves varying degrees of ionic and covalent bonding, including hydrogen bonding.
  • solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid.
  • solvent encompasses both solution-phase and isolatable solvates.
  • suitable solvates include ethanolates, methanolates, and the like.
  • the present invention includes within its scope the use prodrugs of the compounds of this invention.
  • prodrugs will be functional derivatives of the compounds of this invention which are readily convertible in vivo into the required compound.
  • the term “administering” shall encompass the treatment of the various conditions described with a compound of formula I or with a compound which may not be a compound of formula I, but which converts to a compound of formula I in vivo after administration to the patient.
  • Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in "Design of Prodrugs," ed. H. Bundgaard, Elsevier, 1985.
  • prodrugs means a compound (e.g, a drug precursor) that is transformed in vivo to yield a compound of Formula I or a pharmaceutically acceptable salt, hydrate or solvate of the compound. The transformation may occur by various mechanisms (e.g. by metabolic or chemical processes), such as, for example, through hydrolysis in blood.
  • prodrugs is provided by T. Higuchi and W. Stella, "Pro-drugs as Novel Delivery Systems," Vol. 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987.
  • the atoms may exhibit their natural isotopic abundances, or one or more of the atoms may be artificially enriched in a particular isotope having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number predominantly found in nature.
  • the present invention is meant to include all suitable isotopic variations of the compounds of generic Formula I.
  • different isotopic forms of hydrogen (H) include protium (1H) and deuterium ( 2 H).
  • Protium is the predominant hydrogen isotope found in nature. Enriching for deuterium may afford certain therapeutic advantages, such as increasing in vivo half-life or reducing dosage requirements, or may provide a compound useful as a standard for characterization of biological samples.
  • Isotopically-enriched compounds within generic Formula I can be prepared without undue experimentation by conventional techniques well known to those skilled in the art or by processes analogous to those described in the Schemes and Examples herein using appropriate isotopically-enriched reagents and/or intermediates.
  • Compounds of the present invention alter the interaction of coregulator proteins with Retinoic Acid Receptor-related Orphan Receptor gamma t (RORgammaT) and thereby antagonize RORgammaT-mediated transcriptional activity, and as such are useful in the treatment of diseases and conditions in which inhibition of RORgammaT is desirable, such as autoimmune and inflammatory diseases and disorders.
  • RORgammaT Retinoic Acid Receptor-related Orphan Receptor gamma t
  • another embodiment of the present invention provides a method for treating a disease or condition mediated by RORgammaT in a subject comprising
  • the compounds according to the invention can be used in therapy.
  • a further aspect of the invention resides in the use of compounds according to the invention or a pharmaceutically acceptable salt thereof for the treatment of RORgammaT- mediated diseases or RORgammaT mediated conditions.
  • Another aspect of the invention resides in the use of compounds or a pharmaceutically acceptable salt thereof having the general formula I for the treatment of autoimmune diseases, in particular those diseases in which Thl7 cells and non-Thl7 cells, which express Thl7 hallmark cytokines play a prominent role.
  • autoimmune diseases include, but are not limited to, the treatment of rheumatoid arthritis, psoriasis, inflammatory bowel disease, Crohn's disease, ankylosing spondylitis and multiple sclerosis.
  • compounds or a pharmaceutically acceptable salt thereof having the general formula I can be used for treatment of inflammatory diseases in which Thl7 cells and/or non-Thl7 cells, which express Thl7 hallmark cytokines play a prominent role such as, but not limited to respiratory diseases, osteoarthritis and asthma.
  • compounds or a pharmaceutically acceptable salt thereof having the general formula I can be used for treatment of infectious diseases in which Thl7 cells and/or non-Thl7 cells, which express Thl7 hallmark cytokines play a prominent role such as, but not limited to mucosal leishmaniasis.
  • Compounds or a pharmaceutically acceptable salt thereof having the general formula I can also be used for treatment of other diseases in which Thl7 cells and/or non-Thl7 cells, which express Thl7 hallmark cytokines play a prominent role such as, but not limited to Kawasaki disease and Hashimoto's thyroiditis.
  • the disease or condition is an autoimmune disease or inflammatory disease.
  • the disease or condition includes, but is not limited to, multiple sclerosis,
  • the compounds according to the invention can be used in therapies to treat or prevent multiple sclerosis, inflammatory bowel disease, Crohn's disease, psoriasis, rheumatoid arthritis, asthma, osteoarthritis, Kawasaki disease, Hashimoto's thyroiditis and mucosal leishmaniasis.
  • the compounds according to the invention can be used to treat or prevent psoriasis.
  • the compounds according to the invention can be used to treat inflammatory bowel disease.
  • This aspect of the present invention further includes the use of a compound of Formula I, la, lb, Ic, Id, Ie, If, Ig or Ih, or a pharmaceutically acceptable salt or solvate thereof, in the manufacture of a medicament for the treatment of a disease or condition mediated by
  • the compounds of this invention can be administered for the treatment or prevention of afflictions, diseases and illnesses according to the invention by any means that effects contact of the active ingredient compound with the site of action in the body of a warm-blooded animal.
  • administration can be oral, topical, including transdermal, ocular, buccal, intranasal, inhalation, intravaginal, rectal, intracisternal and parenteral.
  • parenteral refers to modes of administration which include subcutaneous, intravenous, intramuscular, intraarticular injection or infusion, intrasternal and intraperitoneal.
  • a warm-blooded animal is a member of the animal kingdom possessed of a homeostatic mechanism and includes mammals and birds.
  • the compounds can be administered by any conventional means available for use in conjunction with pharmaceuticals, either as individual therapeutic agents or in a combination of therapeutic agents. They can be administered alone, but are generally administered with a pharmaceutical carrier selected on the basis of the chosen route of administration and standard pharmaceutical practice.
  • the dosage administered will be dependent on the age, health and weight of the recipient, the extent of disease, kind of concurrent treatment, if any, frequency of treatment and the nature of the effect desired.
  • a daily dosage of active ingredient compound will be from about 1.0-2000 milligrams per day. Ordinarily, from 10 to 500 milligrams per day in one or more applications is effective to obtain desired results.
  • These dosages are the effective amounts for the treatment and prevention of afflictions, diseases and illnesses described above, e.g., autoimmune and inflammatory diseases and disorders.
  • compositions include e.g. those suitable for oral, sublingual, subcutaneous, intravenous, intramuscular, nasal, local, or rectal administration, and the like, all in unit dosage forms for administration.
  • the active ingredient may be presented as discrete units, such as tablets, capsules, powders, granulates, solutions, suspensions, and the like.
  • the pharmaceutical composition of the invention may be presented in unit-dose or multi-dose containers, e.g. injection liquids in predetermined amounts, for example in sealed vials and ampoules, and may also be stored in a freeze dried (lyophilized) condition requiring only the addition of sterile liquid carrier, e.g. water, prior to use.
  • sterile liquid carrier e.g. water
  • the active agent may be compressed into solid dosage units, such as pills, tablets, or be processed into capsules or suppositories.
  • the active agent can be applied as a fluid composition, e.g. as an injection preparation, in the form of a solution, suspension, emulsion, or as a spray, e.g. a nasal spray.
  • solid dosage units For making solid dosage units, the use of conventional additives such as fillers, colorants, polymeric binders and the like is contemplated. In general any pharmaceutically acceptable additive which does not interfere with the function of the active compounds can be used. Suitable carriers with which the active agent of the invention can be administered as solid compositions include lactose, starch, cellulose derivatives and the like, or mixtures thereof, used in suitable amounts. For parenteral administration, aqueous suspensions, isotonic saline solutions and sterile injectable solutions may be used, containing pharmaceutically acceptable dispersing agents and/or wetting agents, such as propylene glycol or butylene glycol.
  • compositions comprising a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof and one or more pharmaceutically acceptable excipients.
  • excipient and “carrier” may be used interchangeably.
  • composition as in pharmaceutical composition, is intended to encompass a product comprising the active ingredient(s), and the inert
  • ingredient(s) pharmaceutically acceptable excipients
  • compositions of the present invention encompass any composition made by admixing a compound of Formula I, additional active ingredient(s), and pharmaceutically acceptable excipients.
  • compositions of the present invention comprise a compound represented by Formula I (or pharmaceutically acceptable salts thereof) as an active ingredient, a pharmaceutically acceptable carrier and optionally other therapeutic ingredients or adjuvants.
  • the compositions include compositions suitable for oral, rectal, topical, and parenteral (including subcutaneous, intramuscular, and intravenous) administration, although the most suitable route in any given case will depend on the particular host, and nature and severity of the conditions for which the active ingredient is being administered.
  • the pharmaceutical compositions may be conveniently presented in unit dosage form and prepared by any of the methods well known in the art of pharmacy.
  • the active ingredient can be administered orally in solid dosage forms, such as capsules, tablets, troches, dragees, granules and powders, or in liquid dosage forms, such as elixirs, syrups, emulsions, dispersions, and suspensions.
  • solid dosage forms such as capsules, tablets, troches, dragees, granules and powders
  • liquid dosage forms such as elixirs, syrups, emulsions, dispersions, and suspensions.
  • the active ingredient can also be
  • sterile liquid dosage forms such as dispersions, suspensions or solutions.
  • Other dosages forms that can also be used to administer the active ingredient as an ointment, cream, drops, transdermal patch or powder for topical administration, as an ophthalmic solution or suspension formation, i.e., eye drops, for ocular administration, as an aerosol spray or powder composition for inhalation or intranasal administration, or as a cream, ointment, spray or suppository for rectal or vaginal administration.
  • Gelatin capsules contain the active ingredient and powdered carriers, such as lactose, starch, cellulose derivatives, magnesium stearate, stearic acid, and the like. Similar diluents can be used to make compressed tablets. Both tablets and capsules can be manufactured as sustained release products to provide for continuous release of medication over a period of hours. Compressed tablets can be sugar coated or film coated to mask any unpleasant taste and protect the tablet from the atmosphere, or enteric coated for selective disintegration in the gastrointestinal tract.
  • powdered carriers such as lactose, starch, cellulose derivatives, magnesium stearate, stearic acid, and the like. Similar diluents can be used to make compressed tablets. Both tablets and capsules can be manufactured as sustained release products to provide for continuous release of medication over a period of hours. Compressed tablets can be sugar coated or film coated to mask any unpleasant taste and protect the tablet from the atmosphere, or enteric coated for selective disintegration in the gastrointestinal tract.
  • Liquid dosage forms for oral administration can contain coloring and flavoring to increase patient acceptance.
  • water a suitable oil, saline, aqueous dextrose (glucose), and related sugar solutions and glycols such as propylene glycol or polyethylene gycols are suitable carriers for parenteral solutions.
  • Solutions for parenteral administration preferably contain a water soluble salt of the active ingredient, suitable stabilizing agents, and if necessary, buffer substances.
  • Antioxidizing agents such as sodium bisulfite, sodium sulfite, or ascorbic acid, either alone or combined, are suitable stabilizing agents.
  • citric acid and its salts and sodium EDTA are also used.
  • parenteral solutions can contain preservatives, such as benzalkonium chloride, methyl- or propylparaben, and chlorobutanol.
  • Suitable pharmaceutical carriers are described in Remington's Pharmaceutical Sciences, A. Osol, a standard reference text in this field.
  • the compounds of the present invention may be conveniently delivered in the form of an aerosol spray presentation from pressurized packs or nebulisers.
  • the compounds may also be delivered as powders which may be formulated and the powder composition may be inhaled with the aid of an insufflation powder inhaler device.
  • the preferred delivery system for inhalation is a metered dose inhalation (MDI) aerosol, which may be formulated as a suspension or solution of a compound of Formula I in suitable propellants, such as fluorocarbons or hydrocarbons.
  • MDI metered dose inhalation
  • an ophthalmic preparation may be formulated with an appropriate weight percent solution or suspension of the compounds of Formula I in an appropriate ophthalmic vehicle, such that the compound is maintained in contact with the ocular surface for a sufficient time period to allow the compound to penetrate the corneal and internal regions of the eye.
  • Useful pharmaceutical dosage-forms for administration of the compounds of this invention include, but are not limited to, hard and soft gelatin capsules, tablets, parenteral injectables, and oral suspensions.
  • a large number of unit capsules are prepared by filling standard two-piece hard gelatin capsules each with 100 milligrams of powdered active ingredient, 150 milligrams of lactose, 50 milligrams of cellulose, and 6 milligrams magnesium stearate.
  • a mixture of active ingredient in a digestible oil such as soybean oil, cottonseed oil or olive oil is prepared and injected by means of a positive displacement pump into gelatin to form soft gelatin capsules containing 100 milligrams of the active ingredient.
  • the capsules are washed and dried.
  • a large number of tablets are prepared by conventional procedures so that the dosage unit is 100 milligrams of active ingredient, 0.2 milligrams of colloidal silicon dioxide, 5 milligrams of magnesium stearate, 275 milligrams of microcrystalline cellulose, 11 milligrams of starch and 98.8 milligrams of lactose.
  • Appropriate coatings may be applied to increase palatability or delay absorption.
  • a parenteral composition suitable for administration by injection is prepared by stirring 1.5% by weight of active ingredient in 10% by volume propylene glycol. The solution is made to volume with water for injection and sterilized.
  • An aqueous suspension is prepared for oral administration so that each 5 milliliters contain 100 milligrams of finely divided active ingredient, 100 milligrams of sodium carboxymethyl cellulose, 5 milligrams of sodium benzoate, 1.0 grams of sorbitol solution, U.S.P., and 0.025 milliliters of vanillin.
  • the same dosage forms can generally be used when the compounds of this invention are administered stepwise or in conjunction with another therapeutic agent.
  • the dosage form and administration route should be selected depending on the compatibility of the combined drugs.
  • coadministration is understood to include the administration of the two agents concomitantly or sequentially, or alternatively as a fixed dose combination of the two active components.
  • the present invention also relates to a pharmaceutical composition
  • a pharmaceutical composition comprising compounds or pharmaceutically acceptable salts thereof having the general formula I in admixture with pharmaceutically acceptable auxiliaries and optionally other therapeutic agents.
  • the auxiliaries must be "acceptable” in the sense of being compatible with the other ingredients of the composition and not deleterious to the recipients thereof.
  • the invention further includes a pharmaceutical composition, as hereinbefore described, in combination with packaging material suitable for said composition, said packaging material including instructions for the use of the composition for the use as hereinbefore described.
  • composition thereof may vary with the particular compound, the route of administration, and the age and condition of the individual subject to whom the medicament is to be administered.
  • a dosage for humans preferably contains 0.0001-100 mg per kg body weight.
  • the desired dose may be presented as one dose or as multiple subdoses administered at appropriate intervals throughout the day.
  • the dosage as well as the regimen of administration may differ between a female and a male recipient.
  • Combination therapies according to the present invention thus comprise the administration of at least one compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof, or a physiologically functional derivative thereof, and the use of at least one other pharmaceutically active agent.
  • the compound(s) of formula (I) and the other pharmaceutically active agent(s) may be administered together or separately and, when administered separately this may occur simultaneously or sequentially in any order.
  • a compound of formula (I) may be combined with one or more other active agents such as: (1) T F-a inhibitors; (2) nonselective COX-I/COX-2 inhibitors; (3) COX-2 inhibitors; (4) other agents for treatment of inflammatory and autoimmune diseases including glucocorticoids, methotrexate, leflunomide, sulfasalazine, azathioprine, cyclosporin, tacrolimus, penicillamine, bucillamine, actarit, mizoribine, lobenzarit,
  • the other therapeutic ingredient(s) may be used in the form of salts, for example as alkali metal or amine salts or as acid addition salts, or prodrugs, or as esters, for example lower alkyl esters, or as solvates, for example hydrates, to optimise the activity and/or stability and/or physical characteristics, such as solubility, of the therapeutic ingredient. It will be clear also that, where appropriate, the therapeutic ingredients may be used in optically pure form.
  • compositions comprising a combination as defined above together with a pharmaceutically acceptable diluent or carrier represent a further aspect of the invention.
  • These combinations are of particular interest in respiratory diseases and are conveniently adapted for inhaled or intranasal delivery.
  • the individual compounds of such combinations may be administered either sequentially or simultaneously in separate or combined pharmaceutical compositions.
  • the individual compounds will be administered simultaneously in a combined pharmaceutical composition.
  • Appropriate doses of known therapeutic agents will be readily appreciated by those skilled in the art.
  • compositions of the present invention include those that also comprise at least one additional therapeutically active agent, in addition to the compound of Formula I, la, lb, Ic, Id, Ie, If, Ig or Ih.
  • the invention further includes a compound of Formula I in combination with one or more other drug(s).
  • EtOAc Ethyl acetate
  • PE Petroleum ether
  • EA Ethyl acetate
  • DCM Dichloro methane
  • DMF ⁇ , ⁇ -Dimethylformamide
  • Dppf 1, 1 '- Bis(diphenylphosphino)ferrocene
  • AcOH Acetic acid
  • DMAC N,N -Dimethylacetamide
  • DMAP N,N-dimethylpyridin-4-amine
  • TEA Triethylamine
  • PYAOP (7-Azabenzotriazol-l- yloxy)tripyrrolidinophosphonium hexafluorophosphat e;
  • Pd(PPh 3 )4 Tetrakis(Triphenylphosphine)Palladium(0); Pd(dppf)Cl 2 : [1, l'-Bis(diphe nylphosphino) ferrocene ]dichloropalladium (II); Pd 2 (dba) 3 : Tris(dibenzylideneace tone)dipalladium(O); BnBr: Benzyl bromide; Ac 2 0: Acetic an hydride; LiHMDS: Lithium bis(trimethylsilyl)amide; PhNTf 2 : N-Phenyl-bis(trifluoromethane sulfonimide); S-Phos: 2- Dicyclohexylphosphino-2',6'-dimethoxybiphenyl; X-Phos: 2-Dicyclohexylphosphino- 2',4',6'-triisopropylbipheny
  • Scheme 1 illustrates a genera method toward the preparation of compounds of formula I.
  • N-acylation with either carboxylic acids or corresponding acid chloride in the presence of base led to the formation of compound B.
  • Subsequent Suzuki coupling with pinacol boronic ester or acid followd by ester hydrolysis afforded the final compound.
  • ester hydrosis occurred under the Suzuki coupling condition and led to the formation of final product within one pot..
  • the final compound I could also be prepared by switching the order of reaction sequence between acylation and Suzuki coupling (see Scheme 2).
  • Suzuki coupling first by reacting halide A with pinacol boronic ester or acid gave intermediate B. Subsequent acylation followed by hydrolysis furnished final product.
  • the ester moiety could be hydrolyzed first followed by reacting with acid or acid chloride to give the final product.
  • Scheme 3 illustrates a general method for the prepartion of compounds of formula I , which contain an amide moiety at A position.
  • acylation followed by ester hydrolysis gave intermediate B.
  • Subsequent Suzuki coupling afforded acid C.
  • Standard amide coupling followed by hydrolysis led to the formation of the final product.
  • Scheme 4 illustrates a general method for compounds containing aryl or heteroaryl substituents at A 6 postion.
  • acylation followed by suzuki compound furnished common intermediate B.
  • Subsequent Suzuki coupling and ester hydrolysis gave the final product.
  • Compound B could be converted into pinacol boronic ester or acid first, followed by subsequent Suzukin coupling with appropriate aryl or heteraryl halide and hydrolysis delivered the final product.
  • Scheme 5 illustrates a general method for the preparation of compounds containing amine or lactam moiety at A 6 position. Starting from common intermedaite A (see Scheme 4 for its preparation), Pd-catalzyed reaction with primary or secondary amines or latams followed by ester hydrolysis furnished the final product.
  • Scheme 6 illustrates a general method for the preparation of compounds which contain alcohol or ketone moiety at A 6 position.
  • halide A acylation followed by reduction of the ester moitey with reducing agent (such as DIBAL-H) afforded compound B.
  • Suzuki coupling with boronic ester or acid gave compound C.
  • oxidation of the product from Grignard addition, followed by ester hydrolysis afford ketone derivative ⁇ .
  • Scheme 7 illustrates a general method for the preparation of compounds, which contain amide, sulfonamides or carbamate of primary or secondary amines at A 6 position.
  • acylation followed by reduction of N0 2 with reducing agent afforded compound B.
  • reducing agent such as SnCl 2
  • Suzuki coupling gave common intermedate C.
  • Stardard amide, sulfonamide, or carbamate formation reactions followed by ester hydrolysis gave the final compounds I.
  • Compound C could be alkylated first with halide in the presence of base to afford a new intermediate which contains a secondray amine.
  • Subsequent standard formation of amides, sulfonamides and carbamates and hydrolysis furnish the final compounds.
  • Scheme 8 illustrates a general strategy for the preparation of compounds, which contain heteroaryl substituetns at A 6 poistion, but could't access through Suzuki coupling as shown in Scheme 4.
  • carboxylic acid A see scheme 4 for its synthesis
  • amide coupling afforded intermediate B.
  • the same stratetgy was also used for the synthesis of a number of other analogs contain a different heterocyles at ths position. Construction of those heterocyles can follow those well-known routes in the literature.
  • Step 2 Preparation of methyl 2-chloro-6-cyclopropylbenzoate (i-lc).
  • Methyl 2-bromo-6- chlorobenzoate (i-lb) (0.8 g, 3.2 mmol), cyclopropylboronic acid (330 mg, 3.84 mmol), Pd(OAc) 2 (72 mg, 0.32 mmol), Cy 3 P (180 mg, 0.64 mmol) and K 3 P0 4 (2.0 g, 9.6 mmol) were mixed in toluene (12 ml) and H 2 0 (1.2 ml). The reaction mixture was stirred at 100 °C for overnight under N 2 atmosphere.
  • Step 3 Preparation of methyl 2-chloro-6-cyclopropoxybenzoic acid (i-2d).
  • 2-chloro-6-cyclopropoxybenzoate (i-2c) (226mg, 1.0 mmol) in CH 3 OH (10 ml) and H 2 0 (10 ml) was added KOH (200 mg).
  • LCMS (ESI) calc'd [M+H] + : 212.63, found: 212.9.
  • Step 4 Preparation of 2-chloro-6-cyclopropoxybenzoyl chloride (i-2).
  • methyl 2-chloro-6-cyclopropoxybenzoic acid (i-2d) 212 mg, 1 mmol
  • DMF 0.1 ml
  • oxalyl chloride 190 mg, 1.5 mmol
  • the mixture solution was protected by N 2 and stirred at r.t for 0.5 h. Then the solution was concentrated to afford 300 mg (crude) for the next step without further purification.
  • Step 1 Preparation of 4-bromo-3-hydroxy-2-nitrobenzoic acid (i-5b).
  • a solution of bromine (0.59 mL, 11.4 mmol) in acetic acid (3 mL) dropwise via a addition funnel over 0.5 h, then the reaction mixture was stirred in the dark at 60 °C. for 12 h. After cooled to room temperature, the reaction mixture was concentrated to give the desired product as a yellow solid (contained 30% of di-brominated products).
  • Step 3 Preparation of methyl 2-amino-4-bromo-3-hydroxybenzoate (i-5d).
  • LCMS (ESI) calc'd [M+H]+: 244.97, found: 246.
  • Step 4 Preparation of methyl 7-bromo-2-oxo-2,3-dihydrobenzo [d]oxazole -4- carboxylate (i-5e).
  • Methyl 2-amino-4-bromo-3-hydroxybenzoate (i-5d) (960mg, 3.9 mmmol) in THF (5ml) was added CDI (956mg, 5.9mmmol) .
  • the mixture was stirred at 80 °C for 3h.
  • LCMS (ESI) calc'd [M+H]+: 270.95, found: 272.
  • Step 5 Preparation of methyl 2-oxo-7-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan -2-yl)-2,3- dihydrobenzo[d]oxazole-4-carboxylate (i-5).
  • Methyl 7-bromo-2 -oxo-2,3- dihydrobenzo[d]oxazole-4-carboxylate (i-5e) (950mg, 3.5mmmol), Pin 2 B 2 (1.78g, 7 mmol), KOAc(1.37g, 14mmol) and (dppf)PdCl 2 (256mg, 0.35mmol) were mixed in dioxane(15ml) under N 2 protection.
  • Step 1 Preparation of 4-bromo-2,5-difluorobenzoic acid (i-6b). To the solution of 1,4- dibromo-2,5-difluorobenzene (i-6a) (27.0 g, 100 mmol) in THF(500 ml) was prepared.
  • Step 2 Preparation of methyl 4-bromo-2,5-difluorobenzoate (i-6c).
  • 4- bromo-2,5-difluorobenzoic acid (i-6b) (18.24 g, 77.3 mmol) in CH 3 OH (200 ml) was added Concentrated sulfuric acid (5 ml) drop wise.
  • the mixture solution was protected by N 2 and stirred at 85 °C for 30 h.
  • LCMS (ESI) calc'd [M+H] + : 251.01, found: 250.9.
  • Step 4 Preparation of 2 methyl 5-fluoro-2-methoxy-4-(4,4,5,5-tetramethyl -1,3,2- dioxaborolan-2-yl)benzoate (i-6).
  • Step 1 Preparation of methyl 2-amino-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborola- n-2- yl)benzoate (i-7).
  • i-7a methyl 2-amino -4-bromobenzoate
  • Pd(dppf)Cl 2 204 mg, 0.25 mmol
  • dppf(138 mg, 0.25 mmol) and KOAc (1.47g, 15 mmol) was added dioxane (20 ml), and the mixture was heated at 100° C under argon for 2 h.
  • CDCL3 57.84 (1H, d),7.12 (1H, s), 7.05(lH,d),5.68(2H, bs), 3.86( 3H, s), 1.33Q2H, s).
  • Step 1 Preparation of methyl lH-indazole-6-carboxylate (i-8b).
  • Methyl 3-amino-4- methylbenzoate (i-8a) (5.0 g, 30.2 mmol) was dissolved in AcOH (140 mL).
  • Sodium nitrite (2.1 g, 30.2 mmol) in water (3.5 mL) was added dropwise to the solution of starting material under ice-cooling at room temperature. The icebath was removed and the mixture was stirred overnight. Half of the solvents were then evaporated, the mixture was diluted with water (80 mL) and extracted with EtOAc (3 x 30 mL).
  • Step 2 Preparation of methyl 3-iodo-lH-indazole-6-carboxylate (i-8c).
  • Methyl 1H- indazole-6-carboxylate (i-8b) (5.0 g, 28.3 mmol) was dissolved in anhydrous DMAC (50 mL).
  • Iodine (14.4 g, 56.7 mmol) and potassium hydroxide (6.3 g, 113.5 mmol) were added in portions under ice-cooling at room temperature. The ice bath was removed and the mixture was stirred at room temperature for lh. The reaction was monitored by TLC (25% MeOH in chloroform) then it was slowly quenched with Sat.
  • Step 4 Preparation of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-iodo-lH-indazo- le-6- carboxylic acid (i-8).
  • Step 1 Preparation of 6-bromo-3-iodo-lH-indazole (i-9b). To a flask was added 6-bromo- lH-indazole (i-9a) (1.96 g, 10 mmol), KOH (1.68 g, 30 mmol) and DMF (60 mL), followed by the addition of I 2 (5.08 g, 20 mmol) in portions. The reaction mixture was stirred at RT for 1 h. The mixture was diluted with H 2 0, and the organic layer was separated. The aqueous layer was extracted with CH 2 C1 2 (3 * 50 mL). The combined organics were washed with H 2 0, brine, dried over Na 2 S0 4 , and concentrated.
  • Step 2 Preparation of (6-bromo-3-iodo-lH-indazol-l-yl)(2-chloro-6-(trifluorom- ethyl)phenyl)methanone (i-9c).
  • 6-bromo-3-iodo-lH-indazol e i-9b
  • DMAP 1.22 g, 10 mmol
  • TEA 2.77 mL, 20 mmol
  • DCM 50 mL
  • 2-chloro-6-(trifluoromethyl) benzoyl chloride (2.61 g, 10 mmol) slowly.
  • Step 3 Preparation of methyl 4-(6-bromo-l-(2-chloro-6-(trifluoromethyl) benzo- yl)-lH- indazol-3-yl)benzoate (i-9).
  • 6-bromo-3-iodo-lH-inda zol-l-yl)(2-chloro-6- (trifluoromethyl)phenyl)methanone (i-9c) 1.058 g, 2 mmol
  • Step 1 Preparation of methyl 3-fluoro-4-(lH-pyrazolo [4, 3-b]pyridin-3-yl) benzo- ate(A-2).
  • a mixture of 3-bromo-lH-pyrazolo[4,3-b]pyridine (A-1) (196.9 mg, 1 mol), 4- (methoxycarbonyl)phenylboronic acid (198 mg, 1 mol), Pd(PPh3)4 (115 mg, 0.1 mol) and K2C03 ( 420 mg, 3mol) were suspended in 1,4-dioxane (5 ml) and H20 (1 ml). The reation mixture was heated at 110°C in a microwave reactor for 2h.
  • Step 1 Preparation of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-iodo-lH -indazole-4- carbaldehyde (B-3).
  • B-3 The mixture of 2-chloro-6-(trifluoromethyl)benzoic acid (B-2) (0.46 g, 2.07 mol) and (COCl) 2 (0.32 mL, 3.76 mol) in DCM (10 mL) and DMF (5drops) was stirred at room temperature for lh. The solvent was removed and the residue was dissolved in DCM (10 mL).
  • Step 2 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-4- formyl-lH- indazol-3-yl)benzoate (B-5).
  • Step 3 Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-4-formyl- lH-indazol- 3-yl)benzoic acid (IB).
  • the mixture of methyl 4-(l-(2-chloro- 6-(trifluoromethyl)benzoyl)-4- formyl-lH-indazol-3-yl)benzoate (B-5) (40 mg, 0.08 mol) and LiOH (17 mg, 0.41 mol) in THF (4 mL) and H 2 0 (2 mL) was stirred at room temperature for 4h.
  • the reaction mixture was diluted with H 2 0 (20 mL).
  • EXAMPLE ID Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-lH- pyrazolo[4,3-b]pyridin-3-yl)-lH-indazole-7-carboxylic acid (ID) SCHEME D.
  • Step 1 Preparation of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-(4-(methoxy- carbonyl)phenyl)-lH-indazole-6-carboxylic acid (E-1).
  • E-1 A mixture of i-8 (300 mg, 0.61 mmol), 4-(methoxycarbonyl)phenylboronic acid (165 mg, 0.92 mmol), Pd(dppf) Cl 2 (50 mg, 0.061 mmol) and KOAc (181 mg, 1.83mmol) in 10 ml dioxane and 2 ml pure H 2 0 was heated to 95 °C for 2h with microwave.
  • Step 2 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) -6- (morpholine-4-carbonyl)-lH-indazol-3-yl)benzoate (E-2).
  • E-1 180 mg, 0.36mmol
  • CH 2 C1 2 15 mL
  • morpholine 37mg, 0.43 mmol
  • PYAOP 374 mg, 0.72mmol
  • TEA 0.16mL, 1.08mmol
  • Step 3 Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (morpholine-4- carbonyl)-lH-indazol-3-yl)benzoic acid (2A).
  • E-2 195mg, 0.34mmol
  • LiOH.H 2 0 72mg, 1.7mmol
  • 10 ml pure H 2 0 was stirred at RT for 2 hours.
  • the solvent was evaporated and the residue was dissolved in water.
  • the precipitated solid was filtered, washed with water and n-hexane, dried to afford an off white solid 2A (184mg, 97%).
  • EXAMPLE 3A Preparation of 2-acetamido-4-(l-(2-chloro-6-(trifluoromethyl) benzoyl)- lH-indazol -3-yl)benzoic acid (3A).
  • Step 2 Preparation of methyl 2-amino-4-(l-(2-chloro-6-(trifluoro methyl) benzoyl)-lH- indazol-3-yl)benzoate (F-3).
  • F-2 110 mg, 0.4 mmol
  • methyl 2-amino-4- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)benzoate 168 mg, 0.4 mmol
  • Pd(PPh 3 ) 4 46 mg, 0.04 mmol
  • K 2 C0 3 138 mg, 1 mmol
  • Step 2 Preparation of methyl 2-acetamido-4-(l-(2-chloro-6-(trifluoromethyl) benzoyl)- lH-indazol-3-yl)benzoate (F-4).
  • compound F-3 180mg, 0.38 mmol
  • acetyl chloride 36 mg, 0.46 mmol
  • DCM 30 mL
  • TEA 1.3 mL, 0.95 mmol
  • Step 1 Preparation of 3-bromo-6-nitro-lH-indazole (H-2).
  • Step 1 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(me- thylamino)-lH-indazol-3-yl)-3-fluorobenzoate (1-1).
  • Step 3 Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(N-methyl- acetamido)-lH-indazol-3-yl)-3-fluorobenzoic acid (5B).
  • Step 1 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(2- oxoazetidin-l-yl)-lH-indazol-3-yl)-3-fluorobenzoate (K-1).
  • Step 2 Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(2-oxoazeti- din-1- yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (6B).
  • methyl 4-(l-(2-chloro-6- (trifluoromethyl)benzoyl)-6-(2-oxoazetidin- 1 -yl)- 1 H-indazol-3 -yl)- 3 -fluorobenzoate (K-1) (53.1 mg, 0 . lmmol) in THF (30 ml) and water(10 ml) was added LiOH (240 mg, 10 mmol).
  • Step 1 Preparation of 4-(6-(2-carboxyethylamino)-l-(2-chloro-6-(trifluoromethy- l)benzoyl)-lH-indazol-3-yl)-3-fluorobenzoic acid (6C).
  • 6C 4-(l-(2- chloro-6-(trifluoromethyl)benzoyl)-6-(2-oxoazetidin- 1 -yl)- 1 H-indazol-3 -yl)-3 -fluorobenzoate (K-l) (53.1 mg, O. lmmol) in THF (30 ml) and waterQO ml) was added LiOH (240 mg, 10 mmol).
  • the reaction was then stirred at 60 °C for 48 hours.
  • the reaction mixture was poured slowly into a saturated solution of sodium bicarbonate (500 mL) at 0 °C.
  • the organic phase was retained and the aqueous phase extracted with dichloromethane (500 mL).
  • the combined organics were then concentrated to a brown oil which was dissolved in methanol (500 mL).
  • Aqueous sodium hydroxide (2 M, 500 mL) was added at 0 °C and the mixture stirred at ambient temperature for 1 hour before the methanol was removed in vacuo.
  • the aqueous mixture was then extracted with ethyl acetate (3 ⁇ 500 mL).
  • Step 6 Preparation of methyl l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-iodo- 1H- pyrazolo[4,3-b]pyridine-6-carboxylate (M-7).
  • M-7 methyl 3-iodo -1H- pyrazolo[4,3-b]pyridine-6-carboxylate
  • Et 3 N 290 mg
  • DMAP 32 mg, 0.26 mmol
  • 2-chloro-6- (trifluoromethyl)benzoyl chloride 630 mg, 2.64 mmol
  • Step 7 Preparation of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-iodo- lH-pyraz- olo[4,3-b]pyridine-6-carboxylic acid (M-8).
  • M-8 methyl l-(2-chloro- - (trifluoromethyl)benzoyl)-3-iodo-lH-pyrazolo[4,3-b]pyridine-6-carboxylate M-7) (700 mg, 1.37 mmol) in THF (15 mL ) and H 2 0 (5 mL) was added LiOH (242 mg, 10.9 mmol). The mixture solution was stirred at RT or 24 h.
  • Step 8 Preparation of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-(2-fluoro- 4- (methoxycarbonyl)phenyl)-lH-pyrazolo[4,3-b]pyridine-6-carboxylic acid (M-9). The mixture of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-iodo-lH- pyrazolo[4,3-b]
  • Step 9 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (morpholine-4-carbonyl)-lH-indazol-3-yl)benzoate (M-10).l-(2-Chloro-6-(trifluor- omethyl)benzoyl)-3 -(2-fluoro-4-(methoxycarbonyl)phenyl)- 1 H-pyrazolo [4,3 -b]pyrid- ine-6- carboxylic acid (M-9) (180 mg, 0.36 mmol) was dissolved in CH 2 C1 2 (15 mL).
  • Step 10 Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(morphol- ine-4- carbonyl)-lH-indazol-3-yl)benzoic acid (7A).
  • Step 1 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(met- hoxy(methyl)carbamoyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoate (N-2). l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(met- hoxy(methyl)carbamoyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoate (N-2). l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(met- hoxy(methyl)carbamoyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoate (N-2). l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(met
  • Step 2 Preparation of methyl 3-bromo-l-(2-chloro-6-(trifluoromethyl)benzoyl) -1H- pyrazolo[4,3-b]pyridine-6-carboxylate (0-3).
  • a solution of methyl 3-bromo -lH-pyrazolo[4,3-b]pyridine-6-carboxylate (0-2) (2.5 g, 9.8 mmol) in DCM (100 mL) was added TEA (2 mL, 14.7 mmol), DMAP (240 mg, 2 mmol), then 2-chloro-6- (trifluoromethyl)benzoyl chloride (3.1 g, 12.7 mmol) in DCM (10 mL) was added dropwise, then the reaction was stirred for 3 h at rt, then diluted with EtOAc (200 mL), washed with sat.
  • Step 3 Preparation of (3-bromo-6-(hydroxymethyl)-lH-pyrazolo[4,3-b] pyridin- l-yl)(2-chloro-6-(trifluoromethyl)phenyl)methanone (0-4). To a solution of methyl 3- bromo- 1 -(2-chloro-6-(trifluoromethyl)benzoyl)- 1 H-pyrazolo [4, 3 -b] pyridine
  • Step 4 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (hyd roxymethyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoate (0-5).
  • a 50 mL round-bottomed flask is desgassed with nitrogen, cooled to -78 °C, cyclopropylmagnesium bromide (4 mL, 2.0 mmol) was added, then methyl 4-(l-(2-chloro-6- (trifluoromethyl)benzoyl)-6-formyl- 1 H-pyrazolo [4,3 -b]pyridine-3 -yl)-3 -fluorobenzoate (0-6) (80 mg, 0.16 mmol) in THF (3 mL, anhydrous) was added slowly. The mixture was stirred from -78 °C to rt for 4 h. Water was added, and the solvent was evaporated.
  • Step 7 Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(cyclopropyl (hydroxy)methyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoic acid (8A).
  • Example 8B Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(cyclop ropane-carbonyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoic acid (8B) SCHEME P
  • Step 2 Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (cyclopropan ecarbonyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoic acid (8B).
  • Step 3 Preparation of methyl l-(2-chloro-6-(trifluoromethyl)benzoyl) -3-iodo-lH
  • Step 4 Preparation of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-iodo- 1H-Indaz ole-6-carboxylic acid (Q-6). A mixture of l-(2-chloro-6-(trifluoromethyl) benzoyl)-3
  • Step 5 Preparation of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-(4- (methoxycar bonyl)phenyl)-lH-indazole-6-carboxylic acid (Q-7).
  • Step 6 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) -6-(2- oxopropylcarbamoyl)-lH-indazol-3-yl)benzoate (Q-8). l-(2-Chloro-6- (trifluoro methyl)benzoyl)-3-(4-(methoxycarbonyl)phenyl)-lH-indazole-6-carboxylic acid (Q-7) (180 mg, 0.36 mmol) was dissolved in CH 2 C1 2 (15 mL).
  • Step 7 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) -6-(5- methyloxazol-2-yl)-lH-indazol-3-yl)benzoate (Q-9).
  • POCl 3 (3.5 mL) was added to a solution of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) -6-(2-oxopropylcarba moyl)-lH-indazol-3-yl)benzoate (Q-8) (191 mg, 0.34 mmol) in pyridine (7 mL) at 25 °C.
  • Step 8 Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) -6-(5-methylox azol-2-yl)-lH-indazol-3-yl)benzoic acid (Q-10).
  • Step 9 Preparation of sodium 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) -6-(5- methyloxazol-2-yl)-lH-indazol-3-yl)benzoate (9A). 4-(l-(2-Chloro-6- (trifluoro
  • Example 11 A Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(3-hy droxyprop-l-ynyl)-lH-indazol-3-yl)benzoic acid (11 A)
  • Step 1 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) (tetrahydro-2H-pyran-2-yloxy)prop-l-ynyl)-lH-indazol-3-yl)benzoate (S-2).
  • Step 2 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (3- hydroxyprop -1-ynyl)- lH-indazol-3-yl)benzoate (S-3).
  • S-3 To a solution of methyl 4-(l-(2- chloro-6-(trifluoromethyl)benzoyl)-6-(3-(tetrahydro-2H-pyran-2-yloxy)prop- 1-ynyl)- 1H- indazol-3-yl)benzoate (S-2) (200 mg, 0.34 mmol) in MeOH (10 mL) was added TsOH (12 mg, 0.07 mmol) at 0 °C .
  • Step 3 Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (3-hydroxy prop-l-ynyl)-lH-indazol-3-yl)benzoic acid (HA).
  • HA 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (3-hydroxy prop-l-ynyl)-lH-indazol-3-yl)benzoic acid (HA).
  • methyl 4-(l-(2- chloro-6-(trifluoromethyl)benzoyl)-6-(3 -hydroxyprop- 1 -ynyl)- 1 H-indazol-3 -yl)benzoate (S-3) 300 mg, 0.5 mmol
  • THF 8.0 mL
  • H 2 0 2.0 mL
  • LiOHH 2 0 108 mg, 2.5 mmol
  • Example 12A Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(3-hy droxybut-l-ynyl)-lH-indazol-3-yl)-3-fluorobenzoic acid (12A) SCHEME T
  • Step 1 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl) benzoyl)-6-(3- (tetrahydro-2H-pyran-2-yloxy)prop-l-ynyl)-lH-indazol-3-yl)-3-fluorobenzoate (T-2).
  • T-1 methyl 4-(6-bromo- l-(2-chloro-6-(trifluoromethyl)benzoyl)- lH-indazol-3-yl)-3-fluorobenzoate (T-1) (200 mg, 0.36 mmol) in DMF (10 mL) and TEA (20 mL) was added 2-(prop-2-ynyloxy)-tetrahydro-2H-pyran (60 mg, 0.43 mmol), copper(I) iodide (6.8 mg, 0.036 mmol) and PdCl 2 (PPh 3 ) 2 (25 mg, 0.036 mmol) under Argon . The mixture was stirred under Argon for 2 hours at 80 °C.
  • Step 2 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (3- hydroxyprop-l-ynyl)-lH-indazol-3-yl)-3-fluorobenzoate (T-3) To a solution of methyl 4- (l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(3-(tetrahydro-2H-pyran-2-ylox
  • T-2 y)prop-l-ynyl)-lH-indazol-3-yl)-3-fluorobenzoate (T-2) (111 mg, 0.18 mmol) in MeOH/H 2 0 (20/2 mL) was added TsOH (15 mg, 0.09 mmol) at 0 °C . The mixture was stirred for 12 hours at rt. The mixture was diluted with H 2 0, The aqueous layer was extracted with CH 2 C1 2 (3 x80 mL). The combined organics were washed with H 2 0, brine, dried over Na 2 S0 4 , and concentrated.
  • Step 3 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) -6-(3- oxoprop-l-ynyl)-lH- indazol-3-yl)-3-fluorobenzoate (T-4)
  • T-3 To a solution of methyl4-(l-(2- chloro-6-(trifluoromethyl)benzoyl)-6-(3 -hydroxyprop- 1 -ynyl)- 1 H-indazol-3 -yl)-3 - fluorobenzoate (T-3) (45 mg, 0.085 mmol) in DCM (20 mL) was added Dess-Martim reagent (108 mg, 0.25 mmol) at 0 °C.
  • Step 4 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) -6-(3- hydroxybut-l-ynyl)-lH -indazol-3-yl)-3-fluorobenzoate (T-5)
  • T-4 To a solution of methyl 4- ( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(3 -oxoprop- 1 -ynyl) - 1 H- indazol-3 -yl)-3 - fluorobenzoate (T-4) (40 mg, 0.076 mmol) in dry THF (5 mL) was added MeMgBr (0.18 mL, 0.53 mmol, 3 M in ether) at -60 °C.
  • Step 5 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) -6-(3- hydroxybut-l-ynyl)-lH -indazol-3-yl)-3-fluorobenzoate (12A).
  • T-5 methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl-6-(3-hydroxybut -l-ynyl)-lH -indazol-3- yl)-3 -fluorobenzoate (T-5) (41 mg, 0.075 mmol) was added THF (8.0 mL), H 2 0 (2.0 mL) and LiOHH 2 0 (32 mg, 0.75 mmol) and the solution was stirred at r.t. overnight. LCMS showed disappearance of staring material. The solution was adjusted to pH 4.0 using IN HC1 and poured into THF (30 mL), washed with brine (20 mL).
  • Example 13A Preparation of 4-(6-(3-aminoprop-l-ynyl)-l-(2-chloro-6-(trifluo romethyl)benzoyl)-lH-indazol-3-yl)-3-fluorobenzoic acid (13A)
  • Step 3 Preparation of 4-(6-(3-aminoprop-l-ynyl)-l-(2-chloro-6-(trifluo romethyl) benzoyl)-lH-indazol-3-yl)-3-fluorobenzoic acid (13A): To the solution of methyl 4-(6-(3- aminoprop- 1 -ynyl)- 1 -(2-chloro-6-(trifluoromethyl)benzoyl) - 1 H-indazol-3 -yl)
  • Example 14A Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) -6-ethy nyl-lH-indazol-3-yl)benzoic acid (14A)
  • V-2 methylsilyl)ethynyl)-lH-indazol-3-yl)benzoate (V-2).
  • V-1 methyl 4-(6-bromo- l-(2-chloro-6-(trifluoromethyl)benzoyl)-lH-indazol-3-yl)benzoate (V-1) (108 mg, 0.2 mmol) in DMF (10 mL) and TEA (20 mL) was added ethynyltrimethylsilane (24 mg, 0.24 mmol), copper(I) iodide (4 mg, 0.02 mmol) and PdCl 2 (PPh 3 ) 2 (14 mg, 0.02 mmol) under argon.
  • Step 2 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) -6-ethy nyl-lH-indazol-3-yl)benzoate (V-3).
  • V-3 methyl 4-(l-(2-chloro-6-(triflu oromethyl)benzoyl)-6-((trimethylsilyl)ethynyl)-lH-indazol-3-yl)benzoate (V-2) (50 mg, 0.09 mmol) in THF (10 mL) at 0 °C was added TBAF (23 mg, 0.09 mmol). The mixture was stirred for 12 hours at rt. The mixture was diluted with H 2 0.
  • Step 3 Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-ethynyl-lH- indazol-3-yl)benzoic acid (14A).
  • methyl 4-(l-(2-chloro-6- (trifluoromethyl)benzoyl)-6-ethynyl-lH-indazol-3-yl)benzoate (V-3) (170 mg, 0.35 mmol) was added THF (8.0 mL), H 2 0 (2.0 mL) and LiOHH 2 0 (74 mg, 1.76 mmol) and the solution was stirred at r.t. overnight.
  • Example 15A Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(3- methyl-l,2,4-oxadiazol-5-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (15A)
  • Step 2 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl) benzoyl) -6-(3- methyl-l,2,4-oxadiazol-5-yl)-lH-indazol-3-yl)-3-fluorobenzoate (W-4).
  • Step 3 Preparation of 4-(l-(2-chloro-6-(trifluoromethyl) benzoyl)-6-(3-methyl -1,2,4- oxadiazol-5-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (15A).
  • Example 16A Preparation 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(5-(hydr oxymethyl)oxazol-2-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (16A)
  • Step 1 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(prop
  • Step 2 Preparation of methyl 4-(6-(5-(acetoxymethyl)oxazol-2-yl)-l-(2-chloro-6- (trifluoromethyl)benzoyl)-lH-indazol-3-yl)-3-fluorobenzoate (X-3).
  • Step 3 Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(5-(hydroxyl methyl)oxazol-2-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (16A).
  • Example 17A Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(l-me thyl-lH-imidazol-4-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (17A)
  • Step 2 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(l-me thyl-lH-imidazol-4-yl)-lH-indazol-3-yl)-3-fluorobenzoate (Y-3). To the solution of methyl 4-(6-bromo- 1 -(2-chloro-6-(trifluoromethyl)benzoyl) - 1 H-indazol-3 -yl)-3 -fluor
  • Step 2 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(oxa zol-2-yl)-lH-indazol-3-yl)-3-fluorobenzoate (Z-3).
  • Step 4 Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) -6-(oxazol-2- yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (18A).
  • AA-2 thylcarbamoyl)-lH-indazol-3-yl)-3-fluorobenzoate
  • AA-1 The mixture of l-(2- chloro-6-(trifluoromethyl)benzoyl)-3-(2-fluoro-4-(methoxycarbonyl)phenyl)-lH-indazole-6- carboxylic acid (AA-1) (250 mg, 0.48 mmol), dimethylamine (2.0 M solution in THF, 0.36 mL, 0.72 mmol), HATU (220 mg, 0.58 mmol) and Et 3 N (0.13 mL, 0.96 mmol) in DCM (10 mL) was stirred at room temperature for 1 h.
  • Step 2 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(dime thylcarbamothioyl)-lH-indazol-3-yl)-3-fluorobenzoate (AA-3). The mixture of methyl4- (l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(dimethylcarbamoyl)-lH-indazol
  • AA-4 ylamino(ethylthio)methyl)-lH-indazol-3-yl)-3-fluorobenzoate (AA-4) (150 mg, 0.26 mmol), cyanamide (37 mg, 0.88 mmol) and Et 3 N ( 44 mg, 0.44 mmol) in MeOH (5 mL) was stirred at room temperature for 2 h. The result solution was diluted with water (30 mL) and the aqueous layer was extracted with EtOAc (20 mL> ⁇ 3).
  • Step 5 Preparation of (E)-4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(N'-cya no-N,N-dimethylcarbamimidoyl)-lH-indazol-3-yl)-3-fluorobenzoic acid (19A). The mixture of (E)-methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(N'-cyano-N,
  • N-dimethylcarbamimidoyl)-lH-indazol-3-yl)-3-fluorobenzoate (AA-5) (100 mg, 0.18 mmol) and LiOH (30 mg, 0.72 mmol) in THF (4 mL) and H 2 0 (2 mL) was stirred at room temperature for 3 h.
  • the reaction mixture was diluted with H 2 0 (20 mL).
  • the combined organic layers were washed with brine (20 mLx l), dried over anhydrous Na 2 S0 4 and concentrated.
  • Example 20A Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(5-oxo-4,5- dihydro-l,3,4-oxadiazol-2-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (20A)
  • Step 2 Preparation of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-iodo-lH-indaz ole-6-carbohydrazide (AB-3).
  • AB-3 methyl l-(2-chloro-6-(trifluoro methyl)benzoyl)-3-iodo-lH-indazole-6-carboxylate (AB-2) (1000 mg, 1.97 mmol) in ethanol (40 mL) was added H 2 H 2 H 2 0 (0. 31 ml, 9.83 mmol). The solution was stirred at reflux temperature overnight. LCMS showed complete transformation to the product.
  • Step 3 Preparation of 5-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-iodo-lH-inda zol-6-yl)-l,3,4-oxadiazol-2(3H)-one (AB-4).
  • AB-4 To a stirred solution of l-(2-chloro-6-(tr ifluoromethyl)benzoyl)-3-iodo-lH-indazole-6-carbohydrazide (AB-3) (100 mg, 0.196 mmol) in THF (2 mL) was added CDI (48 mg, 0.294 mmol) and Et 3 N (30 mg, 0.294 mmol). The solution was stirred at rt overnight.
  • Step 4 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(5-oxo -4,5-dihydro-l,3,4-oxadiazol-2-yl)-lH-indazol-3-yl)-3-fluorobenzoate (AB-5).
  • Step 5 Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(5-oxo-4,5-di hydro-l,3,4-oxadiazol-2-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (20A). To a stirred solution of methyl4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(5-oxo-4,5-di
  • Example 21A Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(4H- l,2,4-triazol-3-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (21A)
  • Step 4 Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(4H-l,2,4-tr iazol-3-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (21A).
  • Example 22A Preparation of 4-(l-(2-chloro-6-trifluorobenzoyl)-6-(thiazol-2-yl)-lH -indazol-3-yl)benzoic acid (22A)
  • AD-1 (20 mg, 0.034 mmol
  • 2-bromothiazole (AD-2) (0.051 mmol)
  • potassium carbonate 0.093 mL, 2M
  • dichloro l-l'-bis(diphenylphosphino)ferrocine palladium (II) dichloromethane adduct
  • 1,4-dioxane (1 mL).
  • the vessel was flushed with Argon and stirred overnight at 90 °C.
  • the reactions were concentrated under reduced pressure.
  • the remaining residue was dissolved in a 1 : 1 DCM/Methanol solution (1 mL) and SiliaBond DMT Resin (78 mg, 0.045 mmol) was then added.
  • Step 1 Preparation of 4-(l-(2-chloro-6-trifluorobenzoyl)-6-(pyridin-3-yl)-lH- indazol-3- yl)benzoic acid (23 A). To a 1 dram vial was added methyl 4-(6-bromo-l-(2-chloro-6- trifluorobenzoyl)-lH-indazol-3-yl)benzoate (i-9) (20 mg, 0.037 mmol), pyridin-3-ylboronic acid (6.9 mg, 0.056 mmol), potassium carbonate (2 M, 0.093 mL, 0.186 mmol), dichloro 1- ⁇ - bis(diphenylphosphino)ferrocine palladium(II) dichloromethane adduct (6.07 mg, 7.44 umol), and 1,4-dioxane (1 mL).
  • the vessel was flushed with Argon and stirred overnight at 90°C. The reactions were concentrated under reduced pressure. The remaining residue was dissolved in a 1 : 1 DCM/Methanol solution (1 mL) and SiliaBond DMT Resin (78 mg, 0.045 mmol) was then added. The mixture was stirred overnight at room temperature. The mixture was then filtered and concentrated under reduced pressure. Lithium hydroxide (1M, 0.186 mL), methanol (0.25 mL) and THF (0.5 mL) was then added and the reaction was stirred overnight at RT. The reactions were concentrated under reduced pressure.
  • Step 1 Preparation of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-(4-(methoxy- carbonyl)phenyl)-lH-indazole-6-carboxylic acid (AH-1).
  • AH-1 l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-(4-(methoxy- carbonyl)phenyl)-lH-indazole-6-carboxylic acid
  • i-8 300 mg, 0.61 mmol
  • 4-(methoxycarbonyl)phenylboronic acid 165 mg, 0.92 mmol
  • Pd(dppf)Cl 2 50 mg, 0.061 mmol
  • KOAc 181 mg, 1.83 mmol
  • Step 2 Preparation of methyl 4-(6-carbamoyl-l-(2-chloro-6-(trifluoromethyl)- benzoyl)- lH-indazol-3-yl)benzoate (AH-2).
  • AH-1 100 mg, 0.2 mmol
  • H 4 C1 13 mg, 0.24 mmol
  • PYAOP 208 mg, 0.4 mmol
  • TEA 0.16 mL, 1.08 mmol
  • Step 4 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (methylsulfonamidomethyl)-lH-indazol-3-yl)benzoate (AH-4).
  • compound AH-3 100 mg, 0.2 mmol
  • methanesulfonyl chloride 23 mg, 0.2 mmol
  • CH 2 C1 2 10 mL
  • TEA 0.1 mL, 0.6 mmol
  • Step 5 Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(methyl- sulfonamidomethyl)-lH-indazol-3-yl)benzoic acid (26A).
  • Step 2 Preparation of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-iodo-lH- indazole-6- carbaldehyde (AI-2).
  • AI- 2 To a stirred solution of (2-chloro-6- (trifluoromethyl)phenyl)(6- (hydroxymethyl)-3-iodo-lH-indazol-l-yl)methanone (AI- 1) (2.5 g, 0.52 mmol) in DCM (160 mL) was added Des Martin Periodinane (3.7 g, 0.78 mmol). The solution was stirred for lh.
  • Step 3 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)- 6-formyl-lH- indazol-3-yl)-3-fluorobenzoate (AI-4).
  • AI-4 l-(2-chloro-6- (trifluoromethyl)benzoyl)-3-iodo-lH-indazole-6-carbaldehyde
  • AI-3 2-fluoro-4-(methoxycarbonyl)phenylboronic acid
  • Pd(OAc) 2 trimer 28 mg, 0.125 mmol
  • K 3 PO 4 600 mg, 2.5 mmol
  • s-Phos 100 mg, 0.25 mmol
  • THF 8 mL
  • H 2 0 2 mL
  • Step 4 Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (2,5- dioxoimidazolidin-4-yl)-lH-indazol-3-yl)-3-fluorobenzoate (AI-5).
  • Step 5 Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(2,5- dioxoimidazolidin-4-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (27A).
  • methyl 4-(l-(2- chloro-6-(trifluoromethyl)benzoyl)-6-(2,5- dioxoimidazolidin-4- yl)-lH-indazol-3-yl)-3-fluorobenzoate (AI-5) was added LiOHH 2 0 (60 mg, 1.5 mmol) and H 2 0 (0.1 mL). The solution was stirred overnight.
  • Example 28A Preparation of 4-(5-(tert-butoxycarbonylamino)-l-(2-chloro-6- (trifluoromethyl)benzoyl)-lH-pyrrolo[2,3-c]pyridin-3-yl)-3-fluorobenzoic acid (28A).
  • Step 2 (3-bromo-5-chloro-lH-pyrrolo[2,3-c]pyridin-l-yl)(2-chloro-6-(trifluoro- methyl)phenyl)methanone (AJ-3).
  • 3-bromo-5-chloro- 1H- pyrrolo[2,3-c]pyridine (AJ-2) (2.9 g, 12.6 mmol) in anhydrous DMF (100 mL) was added 2- chloro-6-(trifluoromethyl)benzoyl chloride (4.6 g, 18.9 mmol) and NaH (60%) (1 g, 25.2 mmol). The solution was stirred at r.t for 2 hours.
  • Step 3 methyl 4-(5-chloro-l-(2-chloro-6-(trifluoromethyl)benzoyl)-lH-pyrrolo [2,3- c]pyridin-3-yl)-3-fluorobenzoate (AJ-4).
  • Step 4 4-(5-(tert-butoxycarbonylamino)-l-(2-chloro-6-(trifluoromethyl)benzoyl)- 1H- pyrrolo[2,3-c]pyridin-3-yl)-3-fluorobenzoic acid (28A). To a microwave tube was added 4-(5 -chloro- 1 -(2-chloro-6-(trifluoromethyl)benzoyl)- 1 H-pyrrolo [2, 3 -c] pyridin-3 -yl)-3 - fluorobenzoic acid (AJ-4) (50 mg, O.
  • Example 29A Preparation of 4-(6-(tert-butoxycarbonylamino)-l-(2-chloro-6- (trifluoromethyl)benzoyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoic acid (29A) and 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(methylamino)-lH- pyrazolo[4,3- b]pyridin-3-yl)-3-fluorobenzoic acid (29B)
  • Step 1 Preparation of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-(2-fluoro-4- (methoxycarbonyl)phenyl) -lH-pyrazolo[4,3-b]pyridine-6-carboxylic acid (AK-2).
  • Step 2 Preparation of methyl 4-(6-(tert-butoxycarbonylamino)-l-(2-chloro-6- (trifluoromethyl)benzoyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoate (AK-3). To a mixture of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-(2-fluoro-4-
  • Step 3 Preparation of 4-(6-(tert-butoxycarbonylamino)-l-(2-chloro-6- (trifluoromethyl)benzoyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoic acid (29A).
  • Step 4 Preparation of methyl 4-(6-(tert-butoxycarbonyl(methyl)amino)-l-(2- chloro-6- (trifluoromethyl)benzoyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoate (AK-4).
  • Example 30A Preparation of 4-(5-acetamido-l-(2-chloro-6-(trifluoromethyl)- benzoyl)- lH-pyrrolo[2,3-c]pyridin-3-yl)-3-fluorobenzoic acid (30A) and 4-(l-(2- chloro-6- (trifluoromethyl)benzoyl)-5-(methylamino)-lH-pyrrolo[2,3-c]pyridin-3-yl)-3- fluorobenzoic acid (30B)
  • Step 1 4-(5-(tert-butoxycarbonylamino)-l-(2-chloro-6-(trifluoromethyl)benzoyl) -1H- pyrrolo[2,3-c]pyridin-3-yl)-3-fluorobenzoic acid (AL-2).
  • Step 2 4-(5-amino-l-(2-chloro-6-(trifluoromethyl)benzoyl)-lH-pyrrolo[2,3-c] pyridin-3- yl)-3-fluorobenzoic acid (AL-3).
  • A-3 4-(5-(tert-butoxy carbonylamino)- 1 -(2-chloro-6-(trifluoromethyl)benzoyl)- 1 H-pyrrolo [2, 3 -c]pyridin-3 -yl)-3 -fluorobenzoic acid (AL-2) (40 mg, 0.07 mmol) in DCM (0.5 mL) and TFA (0.5 mL) was stirred under Ar at rt for 5 h. The mixture was evaporated, the crude product was used to the next step directly.
  • LCMS (ESI) calc'd for C 22 Hi 2 ClF 4 N303 [M+H] + : 478, found: 478.
  • Step 3 4-(5-acetamido-l-(2-chloro-6-(trifluoromethyl)benzoyl)-lH-pyrrolo[2,3-c] pyridin-3-yl)-3-fluorobenzoic acid (30A). To a stirred solution of 4-(5-amino-l-(2- chloro- 6-(trifluoromethyl)benzoyl)-lH-pyrrolo[2,3-c]pyridin-3-yl)-3-fluorobenzoic acid (AL-3) (30 mg, 0.06 mmol) in Ac 2 0 (1 mL) was added AcOH (0.2 mL) was stirred under Ar at 100 °C for 1 h.
  • A-3 4-(5-amino-l-(2- chloro- 6-(trifluoromethyl)benzoyl)-lH-pyrrolo[2,3-c]pyridin-3-yl)-3-fluorobenzoic acid
  • Step 4 Methyl 4-(5-(tert-butoxycarbonyl(methyl)amino)-l-(2-chloro-6-(trifluoro- methyl)benzoyl)-lH-pyrrolo[2,3-c]pyridin-3-yl)-3-fluorobenzoate (AL-4). To a stirred solution of 4-(5-acetamido-l-(2-chloro-6-(trifluoromethyl)benzoyl)-lH- pyrrolo[2,3- c]pyridin-3-yl)-3-fluorobenzoic acid (AL-2) (100 mg, 0.17 mmol) in anhydrous DMF (5 mL) at r.t.
  • Step 5 Methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-5-(methylamino)-lH pyrrolo[2,3-c]pyridin-3-yl)-3-fluorobenzoate (AL-5).
  • A-5 Methyl 4-(5- (tert-butoxycarbonyl(methyl)amino)- 1 -(2-chloro-6-(trifluoromethyl)- benzoyl)- 1 H- pyrrolo[2,3-c]pyridin-3-yl)-3-fluorobenzoate (AL-4) (120 mg, as generated from step 4) in DCM (4 mL) was added TFA (2 mL) under N 2 .
  • Step 6 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-5-(methylamino)-lH-pyrrolo [2,3- c]pyridin-3-yl)-3-fluorobenzoic acid (30B).
  • the compounds of the invention inhibit RORgammaT activity.
  • Activation of RORgammaT activity can be measured using e.g. biochemical TR-FRET assay.
  • TR-FRET assay
  • interaction of cofactor-derived peptides with human RORgammaT -Ligand Binding Domain (LBD) can be measured.
  • LBD RORgammaT -Ligand Binding Domain
  • the TR-FRET technique is a sensitive biochemical proximity assay that will give information concerning the interaction of a ligand with the LBD, in the presence of cofactor derived peptides (Zhou et al, Methods 25:54-61, 2001).
  • RORgammaT To identify novel antagonists of RORgammaT, an assay was developed which employs the interaction of RORgammaT with its co-activator peptide SRC 1 2. This peptide mimics the recruitment of co-activators to RORgammaT through its interaction with the LXXLL (eg R box) motifs (Xie et al, J. Immunol. 175: 3800-09, 2005; Kurebayashi et al, Biochem. Biophys. Res. Commun. 315: 919-27, 2004; Jin et al, Mol. Endocrinology 24:923-29, 2010).
  • LXXLL eg R box
  • HIS-tagged RORy-LBD protein was expressed in SF9 cells using a baculovirus expression system.
  • the RORy-LBD protein was purified by glutathione sepharose chromatography. Separately, SF9 cells not expressing any recombinant protein were lysed and the lysate was added to the purified RORy-LBD at 0.25 ⁇ lysate (from 10,000 SF9 cells)/nM purified protein. The mixture was then diluted in assay buffer (50 mM Tris pH 7.0, 50 mM KC1, 1 mM EDTA, 0.1 mM DTT) to obtain RORy-LBD final concentration of 3 nM in 384-well assay plate.
  • assay buffer 50 mM Tris pH 7.0, 50 mM KC1, 1 mM EDTA, 0.1 mM DTT
  • a stock of biotinylated-LXXLL peptide from coactivator SRC1 (Biotin- CPSSHSSLTERHKILHRLLQEGSPS) was prepared in assay buffer and added to each well (100 nM final concentration).

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Immunology (AREA)
  • Rheumatology (AREA)
  • Pulmonology (AREA)
  • Diabetes (AREA)
  • Endocrinology (AREA)
  • Dermatology (AREA)
  • Transplantation (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Pain & Pain Management (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Indole Compounds (AREA)
  • Peptides Or Proteins (AREA)

Abstract

Provided are compounds according to Formula I or a pharmaceutically acceptable salt or solvate thereof. Such compounds can be used in the treatment of RORgammaT-mediated diseases or conditions.

Description

4-HETEROARYL SUBSTITUTED BENZOIC ACID COMPOUNDS AS RORgammaT INHIBITORS AND USES THEREOF
BACKGROUND OF THE INVENTION
Upon activation by antigen-presenting cells naive T helper cells undergo clonal expansion and will ultimately differentiate in cytokine secreting effector T cells, such as Thl and Th2 subtypes. A third and distinct effector subset has been identified, which plays a key role in providing immunity to bacteria and fungi at mucosal surfaces (Kastelein et al, Annu. Rev. Immunol. 25: 221-242, 2007). This effector T helper cell subset can be distinguished based on its ability to produce large quantities of IL-17/F, IL-21 and IL-22, and is named Thl7 (Miossec et al, New Eng. J. Med. 2361 : 888-898, 2009).
Different T helper subsets are characterized by the expression of lineage specific master transcription factors. Thl and Th2 effector cells express Tbet and GATA3, respectively. A Thymocyte/T cell specific variant of Retinoic Acid Receptor-related Orphan Receptor (ROR), RORgammaT, is highly expressed in Thl7 cells (He et al, Immunity 9: 797-806, 1998).
RORgammaT belongs to the nuclear hormone receptor superfamily (Hirose et al, Biochem. Biophys. Res. Comm. 205: 1976-1983, 1994). RORgammaT is a truncated form of
RORgamma, lacking the first N-terminal 21 amino acids and is, in contrast to RORgamma which is expressed in multiple tissues (heart, brain, kidney, lung, liver and muscle), exclusively expressed in cells of the lymphoid lineage and embryonic lymphoid tissue inducers (Sun et al, Science 288: 2369-2372, 2000; Eberl et al, Nat Immunol. 5: 64-73, 2004).
Studies using heterozygous knock-in mice replacing the RORgammaT open reading frame with GFP, revealed a constitutive expression of GFP in approximately 10% of the CD4+ T cells in the small intestinal lamina propria (LP), co-expressing the Thl 7 cytokines IL-17/F and IL-22 (Ivanov et al, Cell 126: 1121-1133, 2006). In mice deficient for
RORgammaT, the number of Thl 7 cells was markedly decreased in the LP and in vitro stimulation of CD4+ T cells, under Thl 7 polarizing conditions resulted in a drastic decrease of IL-17 expression. These results were further substantiated via forced expression of
RORgammaT in naive CD4+ T cells, which resulted in an induction of IL-17/F and IL-22 (Ivanov et al, Cell 126: 1121-1133, 2006). Taken together demonstrating the importance of RORgammaT in differentiation and stabilization of the Thl7 lineage. In addition, a ROR family member, RORalpha has been demonstrated to be involved in Thl7 differentiation and stabilization (Yang et al, Immunity 28: 29-39, 2008).
Recently, RORgammaT was shown to play a crucial role in non-Thl7 lymphoid cells. In these studies, RORgammaT was critically important in innate lymphoid cells expressing Thyl, SCA-1 and IL-23R proteins. Genetic disruption of RORgamma in a mouse colitis model dependent on these innate lymphoid cells, prevented colitis development (Buonocore et al, Nature 464: 1371-1375, 2010). In addition, RORgammaT was shown to play a crucial role in other non-Thl7 cells, such as mast cells (Hueber et al, J. Immunol. 184: 3336-3340, 2010). Finally, RORgammaT expression and secretion of Thl7-type of cytokines was reported for Lymphoid Tissue Inducer cells, NK T-cells, NK cells (Eberl et al, Nat. Immunol. 5: 64-73, 2004) and gamma-delta T-cells (Sutton et al, Nat. Immunol. 31 : 331-341, 2009; Louten et al, J. Allergy Clin. Immunol. 123 : 1004-1011, 2009), suggesting an important function for RORgammaT in these subtypes of cells.
Based on the role of IL-17 producing cells (either Thl7 or non-Thl7 cells)
RORgammaT has been identified as a key mediator in the pathogenesis of several diseases (Louten et al, J. Allergy Clin. Immunol. 123 : 1004-1011, 2009; Annuziato et al, Nat. Rev. Rheumatol. 5: 325-331, 2009). This was confirmed using several disease models
representative of autoimmune diseases. Genetic ablation of the RORgamma gene in mice prevented the development of experimental autoimmune diseases, such as experimental autoimmune encephalomyelitis (EAE) and colitis (Ivanov et al, Cell 126: 1121-33, 2006; Buonocore et al, Nature 464: 1371-1375, 2010).
Being a critical mediator in Thl7-cells and other non-Thl7 cells, antagonism of the transcriptional activity of RORgammaT is expected to have a beneficial effect on
autoimmune diseases, such as, but not limited to rheumatoid arthritis, psoriasis, multiple sclerosis, inflammatory bowel disease, Crohn's disease, and asthma (Annunziato et al, Nat. Rev. Immunol. 5: 325-331, 2009; Louten et al, J. Allergy Clin. Immunol. 123: 1004-1011, 2009). Antagonism of RORgammaT may also be beneficial in other diseases, which are characterized by increased levels of Thl7 cells and/or elevated levels of Thl7 hallmark cytokines such as IL-17, IL-22 and IL-23. Examples of such diseases are Kawasaki Disease (Jia et al, Clin. Exp. Immunol. 162: 131-137, 2010) and Hashimoto's thyroiditis (Figueroa- Vega et al, J. Clin. Endocrinol. Metab. 95: 953-62, 2010). Another example includes infectious diseases, such as, but not limited to, mucosal leishmaniasis (Boaventura et al, Eur. J. Immunol. 40: 2830-2836, 2010). In each of the above examples the inhibition may be enhanced by simultaneous inhibition of RORalpha.
Compounds modulating RORgammaT have been reported. Examples of agonists include T0901317 and SR1078 (Wang et al, ACS Chem. Biol. 5: 1029-1034, 2010). In addition, antagonist have been reported such as 7-oxygenated sterols (Wang et al, J. Biol. Chem. 285: 5013-5025, 2009) and compounds described in EP2181710 Al .
Numerous immune and inflammatory disorders continue to afflict millions of patients worldwide. Although significant advances have been made in treating these disorders, current therapies do not provide satisfactory results for all patients due to, for example, detrimental side effects or insufficient efficacy. One exemplary immue disorder in need of better therapy is psoriasis. Various therapeutics have been developed in an attempt to treat psoriasis.
However, the traditional therapies for psoriasis often have toxic adverse effects. An exemplary inflammatory disorder in need of better treatment is rheumatoid arthritis.
Numerous therapeutics have been developed in an attempt to treat this disorder. However, some patients develop resistance to current therapies.
Accordingly, a need exists for improved treatments for immune disorders and inflammatory disorders. The present invention addresses this need and provides other related advantages.
SUMMARY OF THE INVENTION
The present invention provides compounds which alter the interaction of coregulator proteins with RORgammaT and thereby antagonize RORgammaT-mediated transcriptional ativity, their use for the treatment of RORgammaT-mediated diseases or conditions, in particular autoimmune diseases and inflammatory diseases, as well as pharmaceutical compositions comprising such compounds and pharmaceutical carriers. DETAILED DESCRIPTION OF THE INVENTION
The present invention provides a compound according to Formula I
Figure imgf000006_0001
I or a pharmaceutically acceptable salt or solvate thereof wherein,
Y is CH, CRa, or N;
n = 0, 1, 2, 3 or 4;
A4 is CR4 or N,
A5 is CR5 or N,
A6 is CR6 or N,
A7 is CR7 or N,
with the proviso that no more than one or two of A4- A7 can be N;
Ra is (Ci-4)alkyl or (C3-7)cycloalkyl;
R1 is
(i) (C3-i2)carbocyclyl(Co-4)alkyl;
(ii) a 4- to 12-membered heterocyclyl(Co-4)alkyl, or
(iii) (Ci-4)alkoxy,
each optionally substituted with one, two, three, four or five R8 ;
R2 is hydroxycarbonyl, hydroxycarbonyl(Ci-io)alkyl, (Ci-io)alkylsulfoxyaminocarbonyl, or carbamoyl;
R3 is hydrogen, halogen, cyano, nitro, hydroxy, (Ci-4)alkylcarbonyloxy, (Ci-4) alkylsulfonylamino, (Ci-4) alkylcarbonylamino, (Co-4) alkylamino, (Ci-4)alkyl, or (Ci_ 4)alkoxy, wherein (Ci-4)alkyl and (Ci-4)alkoxy are optionally substituted with one or more halogen;
R4-R7 independently are hydrogen, halogen, amino, cyano, hydroxy, (Ci_3)alkoxy, (Ci_ 4)alkyl, (Co-io)alkylaminocarbonyl, (Co-6)alkyoxycarbonylamino, (di)(Ci_
6)alkylaminocarbonyl, (Ci-6)alkylcarbonylamino, (Ci-4)alkylamino, amino(Ci-4)alkyl or formaldehyde, wherein (Ci-3)alkoxy, (Ci-4)alkyl, (Co-io)alkyl)aminocarbonyl, (di)(Ci_ 6)alkylaminocarbonyl, (Ci-4)alkylamino and amino(Ci-4)alkyl are optionally substituted with one or more halogen, hydroxyl or (Ci-3)alkoxy; or a group having the
Figure imgf000007_0001
, optionally substituted with one or more of the following: (Ci_ io)alkyl, halogen, amino, cyano, hydroxy, (Ci_3)alkoxy, and wherein m is 1, 2, 3, or 4; R6 is, additionally,
(i) (C3-7)cycloalkyl or (C3-5)heterocycloalkyl, both optionally substituted with one or more groups selected from halogen, amino, amino(Ci-4)alkyl, cyano, nitro, hydroxyl, oxo (=0), H2NC(0), (Ci-3)alkoxycarbonyl, (di)(Ci_
6)alkylaminocarbonyl, (Ci-4)alkyl or (Ci-3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci_3)alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
(ii) (C2-9)heteroaryl, optionally substituted with one or more groups selected from halogen, amino, amino(Ci-4)alkyl, cyano, nitro, hydroxyl, oxo (=0), H2NC(0), (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl or (Ci-3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci_ 3)alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
(iii) (C6-i4)aryl, optionally substituted with one or more groups selected from halogen, amino, amino(Ci-4)alkyl, cyano, nitro, hydroxyl, oxo (=0), H2NC(0), (Ci_ 3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl or (Ci-3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci_ 3)alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
(iv) (C3-5)heterocycloalkylcarbonyl, optionally substituted with one or more groups selected from halogen, amino, amino(Ci-4)alkyl, cyano, nitro, hydroxyl, oxo (=0), H2NC(0), (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl or (Ci_ 3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci-3)alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
(v) (C3-5)heterocycloalkylamino, optionally substituted with one or more groups selected from halogen, amino, amino(Ci-4)alkyl, cyano, nitro, hydroxyl, oxo (=0), H2NC(0), (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl or (Ci_ 3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci-3)alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
(vi) (C3-5)cycloalkylaminocarbonyl, optionally substituted with one or more groups selected from halogen, amino, amino(Ci-4)alkyl, cyano, nitro, hydroxyl, oxo (=0), H2NC(0), (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl or (Ci_ 3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci-3)alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
(vii) (C3-5)cycloalkylcarbonylamino, optionally substituted with one or more groups selected from halogen, amino, amino(Ci-4)alkyl, cyano, nitro, hydroxyl, oxo (=0), H2NC(0), (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl or (Ci_ 3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci-3)alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
(viii) (C3-5)cycloalkyl(Ci-4)alkyl, optionally substituted with one or more groups
selected from halogen, amino, amino(Ci-4)alkyl, cyano, nitro, hydroxyl, oxo (=0), H2NC(0), (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl or (Ci_ 3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci-3)alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
(ix) (C3-5)cycloalkylamino, optionally substituted with one or more groups selected from halogen, amino, amino(Ci-4)alkyl, cyano, nitro, hydroxyl, oxo (=0), H2NC(0), (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl or (Ci_ 3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci-3)alkoxy are optionally substituted with one or more halogen, amino or hydroxyl; (x) (C3-5)cycloalkylcarbonyl, optionally substituted with one or more groups selected from halogen, amino, amino(Ci-4)alkyl, cyano, nitro, hydroxyl, oxo (=0), H2NC(0), (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl or (Ci_ 3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci-3)alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
(xi) (C2-9)heteroaryl(Ci-4)alkyl, optionally substituted with one or more groups
selected from halogen, amino, amino(Ci-4)alkyl, cyano, nitro, hydroxyl, oxo (=0), H2NC(0), (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl or (Ci_ 3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci-3)alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
(xii) (C2-9)heteroarylcarbonyl, optionally substituted with one or more groups selected from halogen, amino, amino(Ci-4)alkyl, cyano, nitro, hydroxyl, oxo (=0), H2NC(0), (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl or (Ci_ 3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci-3)alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
(xiii)
Figure imgf000009_0001
(xiv) (C2-4)alkynyl, optionally substituted with one or more (Ci-4)alkyl, which (Ci_ 4)alkyl may be substituted with hydroxyl or amino; or
(xv) (Ci-6)alkoxycarbonylamino,
(Ci-6)alkylcarbonylamino,
(Ci-6)alkylsulfonylamino(Co-4)alkyl,
(Ci-6)alkylaminocarbonylamino,
(Ci-6)alkyoxycarbonylamino(Co-4)alkyl,
Hydroxycarbonyl(Ci-4)alkylamino,
Hydroxycarbonyl, or
(Ci-6)alkylamino, each optionally substituted with one or more (Ci-4)alkyl, hydroxyl or amino;
R8 is halogen, cyano, amino, nitro, hydroxy, oxo(=0), H2NC(0)-, (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl, (C3-7)cycloalkyl, (C3-5)heterocycloalkyl, (Ci_ 4)alkenyl, (C3-6)cycloalkoxy or (Ci-3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci_ 6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci_3)alkoxy are optionally substituted with one, two or three halogens; and
x is 0, 1, 2, 3, 4 or 5.
In a first embodiment of the compound having Formula I is a compound of having
Formula la
Figure imgf000010_0001
la
and a pharmaceutically acceptable salt or solvate thereof.
In a second embodiment of the compound having Formula I is a compound of claim 1 having Formula lb
Figure imgf000011_0001
and a pharmaceutically acceptable salt or solvate thereof.
In a first subset of the invention is a compound wherein Y is N.
In a third embodiment of the compound having Formula I is a compound having Formula Ic
Figure imgf000011_0002
and a pharmaceutically acceptable salt or solvate thereof.
In a fourth embodiment of the compound having Formula I is a compound having Formula Id
Figure imgf000012_0001
and a pharmaceutically acceptable salt or solvate thereof.
In a first subset of the fourth embodiment is a compound wherein Y is N. In a first subset of the first embodiment is a compound having Formula Ie
Figure imgf000012_0002
Ie
and a pharmaceutically acceptable salt or solvate thereof.
In a second subset is a compound having Formula If
Figure imgf000013_0001
If
and a pharmaceutically acceptable salt or solvate thereof.
In a third subset is a compound having Formula Ig
Figure imgf000013_0002
Ig
and a pharmaceutically acceptable salt or solvate thereof.
In a fourth subset is a compound having Formula Ih
Figure imgf000014_0001
and a pharmaceutically acceptable salt or solvate thereof.
In a fifth subset of the first embodiment is a compound wherein A4, A5, A6, A7 are selected from the group consisting of: (i) CR4, CR5, CR6, CR7; (ii) N, CR5, CR6, CR7; (iii) CR4, N, CR6, CR7; (iv) CR4, CR5, N, CR7; (v) CR4, CR5, CR6, N; (vi) N, N, CR6,CR7; (vii) CR4, N, N, CR7; (viii) CR4, CR5, N, N; (ix) N, CR5, N, CR7; (x) CR4, N, CR6, N; and (xi) N, CR5, CR6, N.
In a sixth subset is a compound wherein A4, A5, A6, A7 are selected from the group consisting of: (i) CR4, CR5, CR6, CR7; (ii) N, CR5, CR6, CR7; and (iii) CR4, N, CR6, CR7.
In a seventh subset is a compound wherein A4, A5, A6, A7 is (i) CR4, CR5, CR6, CR7, or (ii) N, CR5, CR6, CR7; and Y is N.
In an eighth subset is a compound wherein R1 is
(i) (C3-7)cycloalkyl or (C3-5)heterocycloalkyl, both optionally substituted with one or more R ;
(ii) (C2-9)heteroaryl(Co-4)alkyl, optionally substituted with one or more R8; or
(iii) (C6-i4)aryl(Co-4)alkyl, optionally substituted with one or more R8. In a ninth subset is a compound wherein R1 is (i) (C2-9)heteroaryl, or (ii) (C6-i4)aryl, optionally substituted with one, two, three, four or five R8.
In a tenth subset is a compound wherein R1 is (C6-i4)aryl, optionally substituted with one or two R8.
In an eleventh subset is a compound wherein R1 is phenyl, optionally substituted with one or two R8.
In a twelfth subset is a compound wherein R2 is C(0)OH. In a thirteenth subset is a compoundwherein R6 is
Figure imgf000015_0001
Figure imgf000016_0001
A still further embodiment of the compounds of Formula I, la, lb, Ic, Id, Ie, If, Ig and Ih, are compounds wherein one of R4, R5, R6, and R7 is other than hydrogen.
The invention also relates to those compounds wherein all specific definitions for A1 through A4, R1 through R8, Ra, Y, m, n and x and all substituent groups in the various aspects of the inventions defined here above occur in any combination within the definition of the compound of Formula I.
Non-limiting examples of the compounds of the present invention include:
(E)-4-( 1 -(2-chloro-6-(prop- 1 -enyl)benzoyl) - 1 H-pyrazolo [4,3 -b]pyridin-3 -yl)-3 - fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-4- formyl- 1 H-indazol-3 -yl)benzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)- 1 H- pyrazolo [4,3 -b]pyridin-3 -yl)- 1 H-indazole- 7-carboxylic acid;
4-(l-(2-chloro-6-cyclopropoxybenzoyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)benzoic acid; 3 -fluoro-4-( 1 -(2-phenylpropanoyl)- 1 H-indazol-3 -yl)benzoic acid;
3-fluoro-4-[l-(methoxyacetyl)-lH-indazol-3-yl]benzoic acid;
3 -fluoro-4- [ 1 -(pyridin-3 -ylcarbonyl)- 1 Hindazol-3 -yljbenzoic acid;
3 - fluoro-4- { 1 -[(2-oxopyrrolidin- 1 -yl)acetyl]- lHindazol-3 -yl }benzoic acid;
3 -fluoro-4- [ 1 -(naphthalen- 1 -ylcarbonyl)- 1 Hindazol-3 -yljbenzoic acid;
3 -fluoro-4- { 1 -[( 1 -methyl- 1 H-indol-2-yl)carbonyl] - 1 Hindazol-3 -yl } benzoic acid;
4- { 1 - [(2-bromo-3 -methylphenyl)carbonyl] - 1 H-indazol-3 -yl } -3 -fluorobenzoic acid;
4- [ 1 -(2, 3 -dihydro- 1 H-inden-4-ylcarbonyl)- 1 Hindazol-3 -yl] -3 -fluorobenzoic acid;
4-( 1 - { [3 -(tertbutoxycarbonyl)-3 -azabicyclo [3.1.0]hex-6-yl] carbonyl } - 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4- [ 1 -(2, 3 -dihydro- 1 -benzofuran-7-ylcarbonyl)- 1 Hindazol-3 -yl] -3 -fluorobenzoic acid;
4- [ 1 -( 1 -benzofuran-7-ylcarbonyl)- 1 Hindazol-3 -yl] -3 -fluorobenzoic acid;
4- { 1 - [(2-bromo-3 -chlorophenyl)carbonyl] - 1 H-indazol-3 -yl } -3 -fluorobenzoic acid;
3 - fluoro-4-( 1 -(tetrahydrofuran-2-carbonyl)- 1 H-indazol-3 -yl)benzoic acid;
4- (l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (morpholine-4-carbonyl)-l H-indazol-3 - yl)benzoic acid;
4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-((2S,6R)-2,6-dimethylmorpholine-4- carbonyl)-l H-indazol-3 -yl)-3 -fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(4-oxopiperidine- 1 -carbonyl)- 1 H-indazol-3 - yl)-3 -fluorobenzoic acid;
2-acetamido-4-(l-(2-chloro-6-(trifluoromethyl) benzoyl)- IH-indazol -3-yl)benzoic acid; 4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl)-2-
(methylsulfonamido)benzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl) benzoyl)-6- (3 -hydroxyazetidin- 1 -yl)- 1 H-indazol-3 - yl)benzoic acid;
4-(6-(azetidin- 1 -yl)- 1 -(2-chloro-6-(trifluoromethyl)benzoyl)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(cyclopropylamino)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(oxetan-3 -ylamino)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(3 -hydroxypyrrolidin- 1 -yl)- 1 H-indazol-3 - yl)-3 -fluorobenzoic acid;
4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-morpholino-l H-indazol-3 -yl)benzoic acid; 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (methoxycarbonylamino)-l H-indazol-3 -yl)-
3 -fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(N- methylacetamido)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(cyclopropanecarboxamido)-lH-indazol-3- yl)-3 -fluorobenzoic acid; 4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(methylsulfonamido)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(3 -methylureido)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4-(6-acetamido- 1 -(2-chloro-6-(trifluoromethyl)benzoyl)- 1 H-indazol-3 -yl)-3 -fluorobenzoic acid;
4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(N-methylmethylsulfonamido)-lH-indazol-
3 -yl)-3 -fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-( 1 , 3 -dimethylureido)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(2-oxo- imidazolidin- 1 -yl)- 1 H-indazol-3 -yl)- 3 -fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6- (2-oxoazetidin- 1 -yl)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4-(6-(2-carboxyethylamino)- 1 -(2-chloro-6- (trifluoromethyl)benzoyl)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(3 -methyl-2-oxoimidazolidin- 1 -yl)- 1 H- indazol-3 -yl)-3 -fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(2-oxopyrrolidin- 1 -yl)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(mor pholine-4-carbonyl)-l H-indazol-3 - yl)benzoic acid;
3- (4-carboxyphenyl)-l-(2-chloro-6-(trifluoromethyl) benzoyl)- lH-indazole-6-carboxylic acid;
4- ( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(cyclopropylcarbamoyl)- 1 H-pyrazolo [4,3- b]pyridin-3 -yl)-3 -fluorobenzoic acid;
4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(cyclopropyl(methyl)carbamoyl)-lH- pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6- (cyclo
Propyl (hydroxy)methyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl)-3 -fluorobenzoic acid;
4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(cyclop
ropane-carbonyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoic acid; 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(hydroxy(oxazol-2-yl)methyl)-lH- pyrazolo[4,3-b]pyridine-3-yl)-3-fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(oxazole-2-carbonyl)- 1 H-pyrazolo [4,3- b]pyridin-3 -yl)-3 -fluorobenzoic acid;
sodium 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(5-methyloxazol-2-yl)-lH-indazol-3- yl)benzoate;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(5 -methyloxazol-2-yl)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(3 -hydroxyprop- 1 -ynyl)- 1 H-indazol-3 - yl)benzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(3 -hydroxybut- 1 -ynyl)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4-(6-(3 -aminoprop- 1 -ynyl)- 1 -(2-chloro-6-(trifluo romethyl)benzoyl)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl) -6-ethynyl- 1 H-indazol-3 -yl)benzoic acid;
4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(3-methyl-l,2,4-oxadiazol-5-yl)-lH-indazol-
3 -yl)-3 -fluorobenzoic acid;
4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(5-(hydroxymethyl)oxazol-2-yl)-lH-indazol-
3 -yl)-3 -fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-( 1 -methyl- 1 H-imidazol-4-yl)- 1 H-indazol-3 - yl)-3 -fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(oxazol-2-yl)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4-(6-(5-bromooxazol-2-yl)- 1 -(2-chloro-6-(trifluoromethyl)benzoyl)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
(E)-4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(N'-cyano-N,N- dimethylcarbamimidoyl)-l H-indazol-3 -yl)-3 -fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(5 -oxo-4, 5 -dihydro- 1 , 3 ,4-oxadiazol-2-yl)-
1 H-indazol-3 -yl)-3 -fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(4H- 1 ,2,4-triazol-3 -yl)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid; 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(5 -methyl- 1 , 3 ,4-oxadiazol-2-yl)- 1 H-indazol- 3-yl)-3-fluorobenzoic acid;
1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-( 1 -methyl- 1 H-imidazol-2-yl)- 1 H-indazol-3 - yl)-3-fluorobenzoic acid;
^-il-(2-chloro-6-trifluorobenzoyl)-6-(thiazol-2-yl)-lH-indazol-3-yl)benzoic acid;
^-r i-{[2-chloro-6-(trifluoromethyl)phenyl]carbonyl}-6-(l -methyl- lH-imidazol-5-yl)- 1H- indazol-3-yl]benzoic acid;
l-{[2-chloro-6-(trifluoromethyl)phenyl]carbonyl}-6-(5-methylthiophen- 3 -yl)- 1 H-indazol-3 -yljbenzoic acid;
1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-pyrimidin-2-yl- 1 H-indazol-3 - yl)benzoic acid;
1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-pyrimidin-4-yl- 1 H-indazol-3 - yl)benzoic acid;
1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-( 1 -methyl- 1 H-imidazol-4-yl)- 1 H- indazol-3 -yljbenzoic acid;
1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-( 1 , 3 -thiazol-4-yl)- 1 H-indazol-3 - yljbenzoic acid;
6- [4-(aminomethyl)pyridin-2-yl] - 1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } - 1 H- indazol-3-yl)benzoic acid;
1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-pyridin-2-yl- 1 H-indazol-3 - yl)benzoic acid;
1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-( 1 , 3 -thiazol-5 -yl)- 1 H-indazol-3 - yljbenzoic acid;
^-(l-(2-chloro-6-trifluorobenzoyl)-6-(thiazol-2-yl)-lH-indazol-3-yl)benzoic acid;
\-( 1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-pyridin-4-yl- 1 H-indazol-3 - yl)benzoic acid;
1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-(4-cyanophenyl)- 1 H-indazol-3 - yljbenzoic acid;
1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-(3 -cyanophenyl)- 1 H-indazol-3 - yljbenzoic acid;
1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-(2-cyanophenyl)- 1 H-indazol-3 - yljbenzoic acid; 4- [ 1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-(2-fluorophenyl)- 1 H-indazol-3 - yljbenzoic acid;
4- [ 1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-(3 -fluorophenyl)- 1 H-indazol-3 - yljbenzoic acid;
4- [ 1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-(4-fluorophenyl)- 1 H-indazol-3 - yljbenzoic acid;
4- [ 1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-( 1 -methyl- 1 H-pyrazol-5 -yl)- 1 H- indazol-3 -yljbenzoic acid;
methyl 4-(l-(2-chloro-6-trifluorobenzoyl)-6-(4-hydroxyphenyl)-l H-indazol-3 -yl)benzoate; 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (methylsulfonamidomethyl)-lH-indazol-3- yl)benzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(2, 5 - dioxoimidazolidin-4-yl)- 1 H-indazol-3■ yl)-3-fluorobenzoic acid;
4-(5 -(tert-butoxycarbonylamino)- 1 -(2-chloro-6- (trifluoromethyl)benzoyl)- 1 H-pyrrolo [2,3- c]pyridin-3 -yl)-3 -fluorobenzoic acid;
4-(6-(tert-butoxycarbonylamino)- 1 -(2-chloro-6- (trifluoromethyl)benzoyl)- 1H- pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(methylamino)- 1 H- pyrazolo [4,3 -b]pyridin-
3 -yl)-3 -fluorobenzoic acid;
4-(5 -acetamido- 1 -(2-chloro-6-(trifluoromethyl)- benzoyl)- 1 H-pyrrolo [2,3 -c]pyridin-3 -yl)-
3 -fluorobenzoic acid; and
4-(l-(2- chloro-6-(trifluoromethyl)benzoyl)-5-(methylamino)-lH-pyrrolo[2,3-c]pyridin-3- yl)-3 -fluorobenzoic acid.
The terms used herein have their ordinary meaning and the meaning of such terms is independent at each occurrence thereof. That notwithstanding, and except where stated otherwise, the following definitions apply throughout the specification and claims. Chemical names, common names, and chemical structures may be used interchangeably to describe the same structure. If a chemical compound is referred to using both a chemical structure and a chemical name, and an ambiguity exists between the structure and the name, the structure predominates. These definitions apply regardless of whether a term is used by itself or in combination with other terms, unless otherwise indicated. Hence, the definition of "alkyl" applies to "alkyl" as well as the "alkyl" portions of "hydroxyalkyl," "fluoroalkyl," "alkoxy", etc.
As used herein, and throughout this disclosure, the following terms, unless otherwise indicated, shall be understood to have the following meanings:
The term "alkyl," as used herein, refers to an aliphatic hydrocarbon group having one of its hydrogen atoms replaced with a bond having the specified number of carbon atoms. In different embodiments, an alkyl group contains, for example, from 1 to 6 carbon atoms (Ci- C6 alkyl) or from 1 to 3 carbon atoms (C1-C3 alkyl). Non-limiting examples of alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, neopentyl, isopentyl, n-hexyl, isohexyl and neohexyl. In one embodiment, an alkyl group is linear. In another embodiment, an alkyl group is branched.
Unless specified otherwise, "alkyl" includes both branched- and straight-chain saturated aliphatic hydrocarbon groups, including all isomers, having the specified number of carbon atoms; for example, "Ci-6 alkyl" (or "Ci-C6 alkyl") includes all of the hexyl alkyl and pentyl alkyl isomers as well as n-, iso-, sec- and t-butyl, n- and isopropyl, ethyl and methyl.
"Alkylene" refers to both branched- and straight-chain saturated aliphatic hydrocarbon groups, including all isomers, having the specified number of carbons, and having two terminal end chain attachments; for example, the term "A-C4alkylene-B" represents, for example, A-CH2- CH2-CH2-CH2-B, A-CH2-CH2-CH(CH3)-CH2-B, A-CH2-CH(CH2CH3)-B, A-CH2- C(CH3)(CH3)-B, and the like. "Alkoxy" represents a linear or branched alkyl group of indicated number of carbon atoms attached through an oxygen bridge; for example "C1-C6 alkoxy" includes -OCH3, -OCH2CH3, -OCH(CH3)2, -0(CH2)5CH3, and the like.
Unless otherwise specifically noted as only "unsubstituted" or only "substituted", alkyl groups are unsubstituted or substituted with 1 to 3 substituents on each carbon atom, with halo, Ci-C20 alkyl, CF3, NH2, N(Ci-C6 alkyl)2, N02, oxo, CN, N3, -OH, -0(Ci-C6 alkyl), C3- C10 cycloalkyl, (C3-7)cycloalkyl, (C3-5)heterocycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, (Co-C6 alkyl) S(0)o-2-, (C0-C6 alkyl)S(0)0-2(Co-C6 alkyl)-, (C0-C6 alkyl)C(0)NH-, H2N-C(NH)-, H2N-C(0)(NH)-, -0(Ci-C6 alkyl)CF3, (C0-C6 alkyl)C(O)-, (C0-C6 alkyl)OC(O)-, (C0-C6 alkyl)0(Ci-C6 alkyl)-, (C0-C6 alkyl)C(0)i-2(Co-C6 alkyl)-, (C0-C6 alkyl)OC(0)NH-, -NH(Ci- C6 alkyl)NHC(0)NH(Ci-C6 alkyl), NHC(0)OCi-C6 alkyl, -NH(Ci-C6 alkyl)NHS02(Ci-C6 alkyl), -(Co-C6 alkyl)NHS02(Ci-C6 alkyl), aryl, aralkyl, heterocycle, heterocyclylalkyl, halo- aryl, halo-aralkyl, halo-heterocycle, halo-heterocyclylalkyl, cyano-aryl, cyano-aralkyl, cyano- heterocycle and cyano-heterocyclylalkyl.
The term "alkenyl" means a straight or branched carbon chain having the specified number of carbon atoms with at least one carbon-carbon double bond. Examples of alkenyl include, but are not limited to, vinyl, allyl, isopropenyl, pentenyl, hexenyl, heptenyl, 1- propenyl, 2-butenyl, 2-methyl-2-butenyl, 2,4-hexadienyl, and the like.
The term "alkynyl" means a straight or branched carbon chain having the specified number of carbon atoms with at least one carbon-carbon triple bond. Examples of alkynyl include, but are not limited to ethynyl, propargyl, 1-propynyl, 2-butynyl, and the like.
The term "carbocycle" (and variations thereof such as "carbocyclic" or "carbocyclyl") as used herein, unless otherwise indicated, refers to (i) a C3 to C8 monocyclic, saturated or unsaturated ring or (ii) a C7 to C12 bicyclic saturated or unsaturated ring system. Each ring in (ii) is either attached via a bond to, or fused (including spirofused) to, the other ring, and each ring is saturated or unsaturated. The carbocycle may be attached to the rest of the molecule at any carbon atom which results in a stable compound.
Saturated carbocyclics form a subset of carbocycles in which the entire ring system (mono- or polycyclic) is saturated. Saturated monocyclic carbocyclic rings are also referred to as cycloalkyl rings, e.g., cyclopropyl, cyclobutyl, etc. The fused bicyclic carbocycles are a further subset of the carbocycles in which a C7 to C10 bicyclic ring system in which each ring is saturated or unsaturated and two adjacent carbon atoms (or in the case of spirofused, one carbon atom) are shared by each of the rings in the ring system. A saturated bicyclic carbocycle is one in which both rings are saturated. An unsaturated bicyclic carbocycle is one in which one ring is unsaturated and the other is unsaturated or saturated. Unless otherwise noted, carbocycle is unsubstituted or substituted with C1-6 alkyl, C1-6 alkenyl, C1-6 alkynyl, aryl, halogen, H2 or OH. A subset of the fused bicyclic unsaturated carbocycles are those bicyclic carbocycles in which one ring is a benzene ring and the other ring is saturated or unsaturated, with attachment via any carbon atom that results in a stable compound.
Representative examples of this subset include the following:
Figure imgf000023_0001
Figure imgf000024_0001
Aromatic carbocycles form another subset of the carbocycles. The term "aryl" refers to aromatic mono- and poly-carbocyclic ring systems in which the individual carbocyclic rings in the polyring systems are fused or attached to each other via a single bond. Suitable aryl groups include phenyl, naphthyl, and biphenyl.
The term "cycloalkyl" means a cyclic ring of an alkane having the specified total ring carbon atoms; for example cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl.
The term "heterocycle" (and variations thereof such as "heterocyclic" or
"heterocyclyl") broadly refers to (i) a stable 4- to 8-membered, saturated or unsaturated monocyclic ring, or (ii) a stable 7- to 12-membered bicyclic ring system, wherein each ring in (ii) is either attached via a bond to, or fused (including spirofused) to, the other ring, and each ring is saturated or unsaturated, and the monocyclic ring or bicyclic ring system contains one or more heteroatoms (e.g., from 1 to 6 heteroatoms, or from 1 to 4 heteroatoms) selected from N, O and S and a balance of carbon atoms (the monocyclic ring typically contains at least one carbon atom and the ring systems typically contain at least two carbon atoms); and wherein any one or more of the nitrogen and sulfur heteroatoms is optionally oxidized, and any one or more of the nitrogen heteroatoms is optionally quaternized. Unless otherwise specified, the heterocyclic ring may be attached at any heteroatom or carbon atom, provided that attachment results in the creation of a stable structure. Unless otherwise specified, when the heterocyclic ring has substituents, it is understood that the substituents may be attached to any atom in the ring, whether a heteroatom or a carbon atom, provided that a stable chemical structure results.
Saturated heterocyclics form a subset of the heterocycles; i.e., the term "saturated heterocyclic" generally refers to a heterocycle as defined above in which the entire ring system (whether mono- or poly-cyclic) is saturated. The term "saturated heterocyclic ring" refers to a 4- to 8-membered saturated monocyclic ring or a stable 7- to 12-membered bicyclic ring system which consists of carbon atoms and one or more heteroatoms selected from N, O and S. Representative examples include piperidinyl, piperazinyl, azepanyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, oxazolidinyl, isoxazolidinyl, morpholinyl, thiomorpholinyl, thiazolidinyl, isothiazolidinyl, 1,4-dioxanyl, 1,4-thioxanyl,
tetrahydropyranyl, tetrahydrofuryl (or tetrahydrofuranyl), tetrahydrothienyl, and
tetrahydrothiopyrany 1. Heteroaromatics form another subset of the heterocycles; i.e., the term "heteroaromatic" (alternatively "heteroaryl") generally refers to a heterocycle as defined above in which the entire ring system (whether mono- or poly-cyclic) is an aromatic ring system. The term "heteroaromatic ring" refers a 5- or 6-membered monocyclic aromatic ring or a 7- to 12- membered bicyclic aromatic ring, and which consists of carbon atoms and one or more heteroatoms selected from N, O and S. In the case of substituted heteroaryl rings containing at least one nitrogen atom (e.g., pyridine), such substitutions can be those resulting in N-oxide formation. Representative examples of monocyclic heteroaromatic rings include pyridyl, pyrrolyl, pyrazinyl, pyrimidinyl, pyridazinyl, thienyl (or thiophenyl), thiazolyl, furanyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, and thiadiazolyl. Examples of bicyclic heteroaromatic rings include
benzotriazolyl, indolyl, benzoxazolyl, benzofuranyl, benzothienyl, benzothiazolyl, benzimidazolyl, isoindolyl, indazolyl, quinoxalinyl, quinazolinyl, cinnolinyl, quinolinyl, isoquinolinyl, naphthyridinyl, pyrazolo[3,4-b]pyridine, imidazo[2, l-b](l,3)thiazole, (i.e.,
Figure imgf000025_0001
), 6-(l-pyrrolyl)-3 -pyridyl, 4-(l-pyrrolyl)phenyl, 4-(pyrid-3-yl)phenyl, 4-(pyrid-
4-yl)phenyl, and benzothiophenyl (i.e.
Figure imgf000025_0002
Another subset of heterocycles is unsaturated heterocycles in which one or both rings are unsaturated (provided the entire ring system is not aromatic). Representative examples of unsaturated heterocycles include dihydrofuranyl, dihydrothienyl, dihydropyranyl,
dihydroimidazolyl, indolinyl, isoindolinyl, chromanyl, isochromanyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, tetrahydronaphthyridinyl, 2,3-dihydrobenzofuranyl, 1,4- benzoxazinyl, 1,3-benzoxazolinyl, 2,3-dihydrobenzo-l,4-dioxinyl (i.e., eft O ), and benzo-
1,3-dioxolyl (i.e., Cc°> ). In certain contexts herein, C °> is alternatively referred to as phenyl having as a substituent methylenedioxy attached to two adjacent carbon atoms. Also included are groups such as chromone and coumarin.
Unless otherwise specifically noted as only unsubstituted or only substituted, cycloalkyl, cycloalkenyl, heterocycloalkyl, aryl (including phenyl) and heteroaryl groups are
unsubstituted or substituted (also referred to as "optionally substituted"). Unless the substituents are specifically provided, substituents for substituted or optionally substituted cycloalkyl, heterocycloalkyl, cycloalkenyl, aryl (including phenyl, and as an isolated substituent or as part of a substituent such as in aryloxy and aralkyl), heteroaryl (as an isolated substituent or as part of a substituent such as in heteroaryloxy and heteroaralkyl) are one to three groups independently selected from halogen (or halo), C1-C5 alkyl optionally substituted with one to five fluorine, H2, N(Ci-C6 alkyl)2, N02, oxo, CN, N3, -OH, -0(Ci-C6 alkyl) optionally substituted with one to five fluorine, C3-Cio cycloalkyl, (C3-7)cycloalkyl, (C3-5)heterocycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, (C0-C6 alkyl) S(O)0-2-, aryl-S(O)0-2-, (C0- C6 alkyl)S(0)0-2(Co-C6 alkylene)-, (C0-C6 alkyl)C(0) H-, H2N-C( H)-, (C0-C6 alkyl)C(O)-, (Co-C6 alkyl)OC(O)-, (Co-C6alkyl)0(Ci-C6 alkylene)-, (C0-C6 alkyl)C(0)i-2(Co-C6 alkylene)-, (Co-C6 alkyl)2NC(0)-, (C0-C6 alkyl)OC(0) H-, aryl, aralkyl, heteroaryl, heteroaralkyl, halo- aryl, halo-aralkyl, halo-heteroaryl, halo-heteroaralkyl, cyano-aryl, cyano-aralkyl, cyano- heteroaryl and cyano-heteroaralkyl.
The term "halogen" (or "halo") refers to fluorine, chlorine, bromine and iodine
(alternatively referred to as fluoro (F), chloro (CI), bromo (Br), and iodo (I)).
The term "haloalkyl" means alkyl having the specified number of carbon atoms in which from one to all of the hydrogen atoms have been replaced by a halogen atom.
The terms "aralkyl" and "heteroaralkyl" refer to an aryl/heteroaryl linked to the rest of the molecule via a Ci to C4 alkylene.
The term "Co" as employed in expressions such as "Co-6 alkylene" means a direct covalent bond; or when employed in experessions such as "Co-6 alkyl" means hydrogen.
Similarly, when an integer defining the presence of a certain number of atoms in a group is equal to zero, it means that the atoms adjacent thereto are connected directly by a bond; for example, in the structure
Figure imgf000026_0001
, wherein s is an integer equal to zero, 1 or 2, the structure is
Figure imgf000026_0002
when s is zero; or it means that the indicated atom is absent; for example -S(0)o- means -S-.
Unless expressly stated to the contrary, an "unsaturated" ring is a partially or fully unsaturated ring. For example, an "unsaturated monocyclic C6 carbocycle" refers to cyclohexene, cyclohexadiene, and benzene. Unless expressly stated to the contrary, all ranges cited herein are inclusive. For example, a heterocycle described as containing from " 1 to 4 heteroatoms" means the heterocycle can contain 1, 2, 3 or 4 heteroatoms.
When any variable occurs more than one time in any constituent or in any formula depicting and describing compounds of the invention, its definition on each occurrence is independent of its definition at every other occurrence. Also, combinations of substituents and/or variables are permissible only if such combinations result in stable compounds. For variable definitions containing terms having repeated terms, e.g., (CRiRj)r, where r is the integer 2, Ri is a defined variable, and Rj is a defined variable, the value of Ri may differ in each instance in which it occurs, and the value of Rj may differ in each instance in which it occurs. For example, if Ri and Rj are independently selected from the group consisting of methyl, ethyl, propyl and butyl, then (CRiRj)2 can be
Figure imgf000027_0001
The term (Ci-6)alkyl as used hereinabove means a branched or unbranched alkyl group having 1-6 carbon atoms, for example methyl, ethyl, propyl, isopropyl, butyl, tert-butyl, n- pentyl and n-hexyl. Preferred is (Ci-4)alkyl.
The term (Ci-5)alkyl means a branched or unbranched alkyl group having 1-5 carbon atoms, for example methyl, ethyl, propyl, isopropyl, butyl, tert-butyl and n-pentyl.
The term (Ci-4)alkyl as used herein means a branched or unbranched alkyl group having 1-4 carbon atoms, being methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl and tert-butyl.
The term (Ci-3)alkoxy means an alkoxy group having 1-3 carbon atoms, the alkyl moiety being branched or unbranched.
The term (Ci-3)alkoxycarbonyl means an alkoxycarbonyl group having 1-3 carbon atoms in the alkoxy moiety, the alkoxy moiety having the same meaning as previously defined.
The term (di)(Ci-6)alkylaminocarbonyl means an alkylaminocarbonyl group, the amino group of which is monosubstituted or disubstituted independently with an alkyl group which contains 1-6 carbon atoms and which has the same meaning as previously defined. Preferred alkyl group is (Ci-4)alkyl.
The term (C3-7)cycloalkyl means a cycloalkyl group having 3-7 carbon atoms, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl. 5-6 Carbon atoms are preferred.
The term (C3-5)heterocycloalkyl means a heterocycloalkyl group having 3-5 carbon atoms, including 1-3 heteroatoms selected from N, O and/or S, which may be attached via a nitrogen if feasible, or a carbon atom. Preferred number of heteroatoms is one or two. Most preferred number is one. Preferred heteroatoms are N or O. Most preferred are piperazinyl, tetrahydropyranyl, morpholinyl and pyrrolidinyl.
A roup having the for ° , means a heterocyclocarbonyl group such as
Figure imgf000028_0001
, , and 5 each optionally substituted with one or more (Ci_ io)alkyl, halogen, amino, cyano, hydroxy, and (Ci_3)alkoxy.
The term (C2-9)heteroaryl means an aromatic group having 2-9 carbon atoms and 1-3 heteroatoms selected from N, O and S, like imidazolyl, thiadiazolyl, pyridinyl, pyrimidinyl, thiophenyl or furyl, pyrazolyl, isoxazolyl or quinolyl. Preferred number of heteroatoms is one or two. Preferred heteroaryl groups are pyrazolyl, thiophenyl, isoxazolyl, pyridyl and quinolyl. The (C2-5)heteroaryl group may be attached via a carbon atom or a nitrogen, if feasible.
The term (C6-i4)aryl means an aromatic hydrocarbon group having 6-14 carbon atoms, such as phenyl, naphthyl, tetrahydronaphthyl, indenyl, anthracyl, More preferred are (C6- io)aryl groups. The most preferred aromatic hydrocarbon group is phenyl.
As used herein, the term "Xa-Xb", shall have the same meaning as the term "Xa-b", wherein X is any atom and a and b are any integers. For example, "Ci-C4" shall have the same meaning as "C1-4". Additionally, when referring to a functional group generically, "Ax" shall have the same meaning, and be interchangeable with, "AX", wherein "A" is any atom and "x" or "X" are any integer. For example, "R1" shall have the same meaning, and be
interchangeable with, "Rl ". In the above definitions with multifunctional groups, the attachment point is at the last group. For example, the term (Ci-3)alkoxycarbonyl refers to, e.g.
Figure imgf000029_0001
, and the term
o
H3C A
(Cl-4)alkylcarbonyloxy refers to, e.g. O
The term "substituted" means that one or more hydrogens on the designated atom/atoms is/are replaced with a selection from the indicated group, provided that the designated atom's normal valency under the existing circumstances is not exceeded, and that the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds. "Stable compound" or "stable structure" is defined as a compound or structure that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent. Accordingly, the term "one or more" when referring to a substituent and/or variable means that one or more hydrogens on the designated atom/atoms is/are replaced with a selection from the indicated group, provided that the designated atom's normal valency under the existing circumstances is not exceeded, and that the substitution results in a stable compound.
The term "optionally substituted" means that a substitution with the specified groups, radicals or moieties, may or may not be made on the specified group.
When, in the definition of a substituent, is indicated that "all of the alkyl groups" of said substituent are optionally substituted, this also includes the alkyl moiety of an alkoxy group.
The use of the terms "salt", "solvate", "ester", "prodrug" and the like, is intended to equally apply to the salt, solvate, ester and prodrug of enantiomers, stereoisomers, rotamers, tautomers, positional isomers, racemates or prodrugs of the inventive compounds.
The term "effective amount" as used herein, refers to an amount of the compound of Formula (I) and/or an additional therapeutic agent, or a composition thereof, that is effective in producing the desired therapeutic, ameliorative, inhibitory or preventative effect when administered to a subject suffering from an RORgammaT-mediated disease or disorder. In the combination therapies of the present invention, as effective amount can refer to each individual agent or to the combination as a whole, wherein the amounts of all agents administered are together effective, but wherein the component agent of the combination may not be present individually in an effective amount. A "subject" is a human or non-human mammal. In one embodiment, a subject is a human. In another embodiment, a subject is a chimpanzee.
It should be noted that any carbon as well as heteroatom with unsatisfied valences in the text, schemes, examples and tables herein is assumed to have the sufficient number of hydrogen atom(s) to satisfy the valences.
The compounds of this invention include the prodrugs, hydrates or solvates of the compounds.
Optical Isomers - Diastereomers - Geometric Isomers - Tautomers
The compounds of Formula I may contain asymmetric or chiral centers, and, therefore, exist in different stereoisomeric forms. It is intended that all stereoisomeric forms of the compounds of Formula (I) as well as mixtures thereof, including racemic mixtures, form part of the present invention. In addition, the present invention embraces all geometric and positional isomers. For example, if a compound of Formula (I) incorporates a double bond or a fused ring, both the cis- and trans-forms, as well as mixtures, are embraced within the scope of the invention.
Compounds described herein may contain an asymmetric center and may thus exist as enantiomers. Where the compounds according to the invention possess two or more asymmetric centers, they may additionally exist as diastereomers. The present invention includes all such possible stereoisomers as substantially pure resolved enantiomers, racemic mixtures thereof, as well as mixtures of diastereomers. The above Formula I is shown without a definitive stereochemistry at certain positions. The present invention includes all stereoisomers of Formula I and pharmaceutically acceptable salts thereof. Diastereoisomeric pairs of enantiomers may be separated by, for example, fractional crystallization from a suitable solvent, and the pair of enantiomers thus obtained may be separated into individual stereoisomers by conventional means, for example by the use of an optically active acid or base as a resolving agent or on a chiral UPLC column. Further, any enantiomer or diastereomer of a compound of the general Formula I may be obtained by stereospecific synthesis using optically pure starting materials or reagents of known configuration.
When compounds described herein contain olefinic double bonds, unless specified otherwise, such double bonds are meant to include both E and Z geometric isomers. Some of the compounds described herein may exist with different points of attachment of hydrogen, referred to as tautomers. For example, compounds including
carbonyl -CH2C(0)- groups (keto forms) may undergo tautomerism to form hydroxyl - CH=C(OH)- groups (enol forms). Both keto and enol forms, individually as well as mixtures thereof, are included within the scope of the present invention.
Diastereomeric mixtures can be separated into their individual diastereomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as, for example, by chromatography and/or fractional crystallization. Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g. chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereomers and converting (e.g.
hydrolyzing) the individual diastereomers to the corresponding pure enantiomers. Also, some of the compounds of Formula (I) may be atropisomers (e.g. substituted biaryls) and are considered as part of this invention. Enantiomers can also be separated by use of chiral HPLC column.
It is also possible that the compounds of Formula I may exist in different tautomeric forms, and all such forms are embraced within the scope of the invention. Also, for example, all keto-enol and imine-enamine forms of the compounds are included in the invention.
All stereoisomers (for example, geometric isomers, optical isomers and the like) of the present compounds (including those of the salts, solvates, esters and prodrugs of the compounds as well as the salts, solvates and esters of the prodrugs), such as those which may exist due to asymmetric carbons on various substituents, including enantiomeric forms (which may exist even in the absence of asymmetric carbons), rotameric forms, atropisomers, and diastereomeric forms, are contemplated within the scope of this invention, as are positional isomers. Individual stereoisomers of the compounds of the invention may, for example, be substantially free of other isomers, or may be admixed, for example, as racemates or with all other, or other selected, stereoisomers. The chiral centers of the present invention can have the S or R configuration as defined by the IUPAC 1974 Recommendations.
Salts
The term "pharmaceutically acceptable salts" refers to salts prepared from
pharmaceutically acceptable non-toxic bases or acids. When the compound of the present invention is acidic, its corresponding salt can be conveniently prepared from
pharmaceutically acceptable non-toxic bases, including inorganic bases and organic bases. Salts derived from such inorganic bases include aluminum, ammonium, calcium, copper (ic and ous), ferric, ferrous, lithium, magnesium, manganese (ic and ous), potassium, sodium, zinc and the like salts. Preferred are the ammonium, calcium, magnesium, potassium and sodium salts. Salts prepared from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines derived from both naturally occurring and synthetic sources. Pharmaceutically acceptable organic non-toxic bases from which salts can be formed include, for example, arginine, betaine, caffeine, choline, N,N'-dibenzyl- ethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol,
ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, dicyclohexylamine, lysine,
methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine and the like.
When the compound of the present invention is basic, its corresponding salt can be conveniently prepared from pharmaceutically acceptable non-toxic inorganic and organic acids. Such acids include, for example, acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p-toluenesulfonic acid and the like. Preferred are citric, hydrobromic, hydrochloric, maleic, phosphoric, sulfuric, and tartaric acids.
The compounds of Formula I can form salts which are also within the scope of this invention. Reference to a compound of Formula I herein is understood to include reference to salts thereof, unless otherwise indicated.
The term pharmaceutically acceptable salt represents those salts which are, within the scope of medical judgement, suitable for use in contact for the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are
commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. They may be obtained during the final isolation and purification of the compounds of the invention, or separately by reacting the free base function with a suitable mineral acid such as hydrochloric acid, phosphoric acid, or sulfuric acid, or with an organic acid such as for example ascorbic acid, citric acid, tartaric acid, lactic acid, maleic acid, malonic acid, fumaric acid, glycolic acid, succinic acid, propionic acid, acetic acid, methanesulfonic acid, and the like. The acid function can be reacted with an organic or a mineral base, like sodium hydroxide, potassium hydroxide, calcium hydroxide, calcium carbonate, ammonium (e.g. diethylamine) or lithium hydroxide.
Solvates
The present invention includes within its scope solvates of compounds of Formula I. As used herein, the term "solvate" refers to a complex of variable stoichiometry formed by a solute (i.e., a compound of Formula I) or a pharmaceutically acceptable salt thereof and a solvent that does not interfere with the biological activity of the solute. Examples of solvents include, but are not limited to water, ethanol, and acetic acid. When the solvent is water, the solvate is known as hydrate; hydrate includes, but is not limited to, hemi-, mono, sesqui-, di- and trihydrates.
The compounds of the invention may form hydrates or solvates. It is known to those of skill in the art that charged compounds form hydrated species when lyophilized with water, or form solvated species when concentrated in a solution with an appropriate organic solvent. One or more compounds of the invention may exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like, and it is intended that the invention embrace both solvated and unsolvated forms. "Solvate" may also mean a physical association of a compound of this invention with one or more solvent molecules. This physical association involves varying degrees of ionic and covalent bonding, including hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. "Solvate" encompasses both solution-phase and isolatable solvates. Non- limiting examples of suitable solvates include ethanolates, methanolates, and the like.
"Hydrate" is a solvate wherein the solvent molecule is H20.
Prodrugs
The present invention includes within its scope the use prodrugs of the compounds of this invention. In general, such prodrugs will be functional derivatives of the compounds of this invention which are readily convertible in vivo into the required compound. Thus, in the methods of treatment of the present invention, the term "administering" shall encompass the treatment of the various conditions described with a compound of formula I or with a compound which may not be a compound of formula I, but which converts to a compound of formula I in vivo after administration to the patient. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in "Design of Prodrugs," ed. H. Bundgaard, Elsevier, 1985.
A discussion of prodrugs is provided in T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems (1987) 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, (1987) Edward B. Roche, ed., American Pharmaceutical Association and Pergamon Press. The term "prodrug" means a compound (e.g, a drug precursor) that is transformed in vivo to yield a compound of Formula I or a pharmaceutically acceptable salt, hydrate or solvate of the compound. The transformation may occur by various mechanisms (e.g. by metabolic or chemical processes), such as, for example, through hydrolysis in blood. A discussion of the use of prodrugs is provided by T. Higuchi and W. Stella, "Pro-drugs as Novel Delivery Systems," Vol. 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987.
Isotopes
In the compounds of generic Formula I, the atoms may exhibit their natural isotopic abundances, or one or more of the atoms may be artificially enriched in a particular isotope having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number predominantly found in nature. The present invention is meant to include all suitable isotopic variations of the compounds of generic Formula I. For example, different isotopic forms of hydrogen (H) include protium (1H) and deuterium (2H). Protium is the predominant hydrogen isotope found in nature. Enriching for deuterium may afford certain therapeutic advantages, such as increasing in vivo half-life or reducing dosage requirements, or may provide a compound useful as a standard for characterization of biological samples. Isotopically-enriched compounds within generic Formula I can be prepared without undue experimentation by conventional techniques well known to those skilled in the art or by processes analogous to those described in the Schemes and Examples herein using appropriate isotopically-enriched reagents and/or intermediates. Utilities
Compounds of the present invention alter the interaction of coregulator proteins with Retinoic Acid Receptor-related Orphan Receptor gamma t (RORgammaT) and thereby antagonize RORgammaT-mediated transcriptional activity, and as such are useful in the treatment of diseases and conditions in which inhibition of RORgammaT is desirable, such as autoimmune and inflammatory diseases and disorders.
Accordingly, another embodiment of the present invention provides a method for treating a disease or condition mediated by RORgammaT in a subject comprising
administering to the subject an amount of a compound having Formula I, la, lb, Ic, Id, Ie, If, Ig or Ih, or a pharmaceutically acceptable salt or solvate thereof, that is effective for treating the disease or condition mediated by RORgammaT in the subject.
The compounds according to the invention can be used in therapy.
A further aspect of the invention resides in the use of compounds according to the invention or a pharmaceutically acceptable salt thereof for the treatment of RORgammaT- mediated diseases or RORgammaT mediated conditions.
Another aspect of the invention resides in the use of compounds or a pharmaceutically acceptable salt thereof having the general formula I for the treatment of autoimmune diseases, in particular those diseases in which Thl7 cells and non-Thl7 cells, which express Thl7 hallmark cytokines play a prominent role. These include, but are not limited to, the treatment of rheumatoid arthritis, psoriasis, inflammatory bowel disease, Crohn's disease, ankylosing spondylitis and multiple sclerosis.
In another aspect, compounds or a pharmaceutically acceptable salt thereof having the general formula I can be used for treatment of inflammatory diseases in which Thl7 cells and/or non-Thl7 cells, which express Thl7 hallmark cytokines play a prominent role such as, but not limited to respiratory diseases, osteoarthritis and asthma. Also, compounds or a pharmaceutically acceptable salt thereof having the general formula I can be used for treatment of infectious diseases in which Thl7 cells and/or non-Thl7 cells, which express Thl7 hallmark cytokines play a prominent role such as, but not limited to mucosal leishmaniasis.
Compounds or a pharmaceutically acceptable salt thereof having the general formula I can also be used for treatment of other diseases in which Thl7 cells and/or non-Thl7 cells, which express Thl7 hallmark cytokines play a prominent role such as, but not limited to Kawasaki disease and Hashimoto's thyroiditis.
In one aspect is the disease or condition is an autoimmune disease or inflammatory disease. The disease or condition includes, but is not limited to, multiple sclerosis,
inflammatory bowel disease, Crohn's disease, ankylosing spondylitis, psoriasis, rheumatoid arthritis, asthma, osteoarthritis, Kawasaki disease, Hashimoto's thyroiditis or mucosal leishmaniasis.
In another aspect, the compounds according to the invention can be used in therapies to treat or prevent multiple sclerosis, inflammatory bowel disease, Crohn's disease, psoriasis, rheumatoid arthritis, asthma, osteoarthritis, Kawasaki disease, Hashimoto's thyroiditis and mucosal leishmaniasis.
In another aspect the compounds according to the invention can be used to treat or prevent psoriasis.
In yet another aspect the compounds according to the invention can be used to treat inflammatory bowel disease.
This aspect of the present invention further includes the use of a compound of Formula I, la, lb, Ic, Id, Ie, If, Ig or Ih, or a pharmaceutically acceptable salt or solvate thereof, in the manufacture of a medicament for the treatment of a disease or condition mediated by
RORgammaT.
Route of Administration/Dosage
The compounds of this invention can be administered for the treatment or prevention of afflictions, diseases and illnesses according to the invention by any means that effects contact of the active ingredient compound with the site of action in the body of a warm-blooded animal. For example, administration can be oral, topical, including transdermal, ocular, buccal, intranasal, inhalation, intravaginal, rectal, intracisternal and parenteral. The term "parenteral" as used herein refers to modes of administration which include subcutaneous, intravenous, intramuscular, intraarticular injection or infusion, intrasternal and intraperitoneal. For the purpose of this disclosure, a warm-blooded animal is a member of the animal kingdom possessed of a homeostatic mechanism and includes mammals and birds.
The compounds can be administered by any conventional means available for use in conjunction with pharmaceuticals, either as individual therapeutic agents or in a combination of therapeutic agents. They can be administered alone, but are generally administered with a pharmaceutical carrier selected on the basis of the chosen route of administration and standard pharmaceutical practice.
The dosage administered will be dependent on the age, health and weight of the recipient, the extent of disease, kind of concurrent treatment, if any, frequency of treatment and the nature of the effect desired. Usually, a daily dosage of active ingredient compound will be from about 1.0-2000 milligrams per day. Ordinarily, from 10 to 500 milligrams per day in one or more applications is effective to obtain desired results. These dosages are the effective amounts for the treatment and prevention of afflictions, diseases and illnesses described above, e.g., autoimmune and inflammatory diseases and disorders.
Compositions include e.g. those suitable for oral, sublingual, subcutaneous, intravenous, intramuscular, nasal, local, or rectal administration, and the like, all in unit dosage forms for administration.
For oral administration, the active ingredient may be presented as discrete units, such as tablets, capsules, powders, granulates, solutions, suspensions, and the like.
For parenteral administration, the pharmaceutical composition of the invention may be presented in unit-dose or multi-dose containers, e.g. injection liquids in predetermined amounts, for example in sealed vials and ampoules, and may also be stored in a freeze dried (lyophilized) condition requiring only the addition of sterile liquid carrier, e.g. water, prior to use.
Mixed with such pharmaceutically acceptable auxiliaries, e.g. as described in the standard reference, Gennaro, A.R. et al, Remington: The Science and Practice of Pharmacy (20th Edition., Lippincott Williams & Wilkins, 2000, see especially Part 5: Pharmaceutical Manufacturing), the active agent may be compressed into solid dosage units, such as pills, tablets, or be processed into capsules or suppositories. By means of pharmaceutically acceptable liquids the active agent can be applied as a fluid composition, e.g. as an injection preparation, in the form of a solution, suspension, emulsion, or as a spray, e.g. a nasal spray.
For making solid dosage units, the use of conventional additives such as fillers, colorants, polymeric binders and the like is contemplated. In general any pharmaceutically acceptable additive which does not interfere with the function of the active compounds can be used. Suitable carriers with which the active agent of the invention can be administered as solid compositions include lactose, starch, cellulose derivatives and the like, or mixtures thereof, used in suitable amounts. For parenteral administration, aqueous suspensions, isotonic saline solutions and sterile injectable solutions may be used, containing pharmaceutically acceptable dispersing agents and/or wetting agents, such as propylene glycol or butylene glycol.
Pharmaceutical Compositions
Another aspect of the present invention provides pharmaceutical compositions comprising a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof and one or more pharmaceutically acceptable excipients. The term "excipient" and "carrier" may be used interchangeably. The term "composition", as in pharmaceutical composition, is intended to encompass a product comprising the active ingredient(s), and the inert
ingredient(s) (pharmaceutically acceptable excipients) that make up the carrier, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients.
Accordingly, the pharmaceutical compositions of the present invention encompass any composition made by admixing a compound of Formula I, additional active ingredient(s), and pharmaceutically acceptable excipients.
The pharmaceutical compositions of the present invention comprise a compound represented by Formula I (or pharmaceutically acceptable salts thereof) as an active ingredient, a pharmaceutically acceptable carrier and optionally other therapeutic ingredients or adjuvants. The compositions include compositions suitable for oral, rectal, topical, and parenteral (including subcutaneous, intramuscular, and intravenous) administration, although the most suitable route in any given case will depend on the particular host, and nature and severity of the conditions for which the active ingredient is being administered. The pharmaceutical compositions may be conveniently presented in unit dosage form and prepared by any of the methods well known in the art of pharmacy.
The active ingredient can be administered orally in solid dosage forms, such as capsules, tablets, troches, dragees, granules and powders, or in liquid dosage forms, such as elixirs, syrups, emulsions, dispersions, and suspensions. The active ingredient can also be
administered parenterally, in sterile liquid dosage forms, such as dispersions, suspensions or solutions. Other dosages forms that can also be used to administer the active ingredient as an ointment, cream, drops, transdermal patch or powder for topical administration, as an ophthalmic solution or suspension formation, i.e., eye drops, for ocular administration, as an aerosol spray or powder composition for inhalation or intranasal administration, or as a cream, ointment, spray or suppository for rectal or vaginal administration.
Gelatin capsules contain the active ingredient and powdered carriers, such as lactose, starch, cellulose derivatives, magnesium stearate, stearic acid, and the like. Similar diluents can be used to make compressed tablets. Both tablets and capsules can be manufactured as sustained release products to provide for continuous release of medication over a period of hours. Compressed tablets can be sugar coated or film coated to mask any unpleasant taste and protect the tablet from the atmosphere, or enteric coated for selective disintegration in the gastrointestinal tract.
Liquid dosage forms for oral administration can contain coloring and flavoring to increase patient acceptance.
In general, water, a suitable oil, saline, aqueous dextrose (glucose), and related sugar solutions and glycols such as propylene glycol or polyethylene gycols are suitable carriers for parenteral solutions. Solutions for parenteral administration preferably contain a water soluble salt of the active ingredient, suitable stabilizing agents, and if necessary, buffer substances. Antioxidizing agents such as sodium bisulfite, sodium sulfite, or ascorbic acid, either alone or combined, are suitable stabilizing agents. Also used are citric acid and its salts and sodium EDTA. In addition, parenteral solutions can contain preservatives, such as benzalkonium chloride, methyl- or propylparaben, and chlorobutanol.
Suitable pharmaceutical carriers are described in Remington's Pharmaceutical Sciences, A. Osol, a standard reference text in this field.
For administration by inhalation, the compounds of the present invention may be conveniently delivered in the form of an aerosol spray presentation from pressurized packs or nebulisers. The compounds may also be delivered as powders which may be formulated and the powder composition may be inhaled with the aid of an insufflation powder inhaler device. The preferred delivery system for inhalation is a metered dose inhalation (MDI) aerosol, which may be formulated as a suspension or solution of a compound of Formula I in suitable propellants, such as fluorocarbons or hydrocarbons.
For ocular administration, an ophthalmic preparation may be formulated with an appropriate weight percent solution or suspension of the compounds of Formula I in an appropriate ophthalmic vehicle, such that the compound is maintained in contact with the ocular surface for a sufficient time period to allow the compound to penetrate the corneal and internal regions of the eye.
Useful pharmaceutical dosage-forms for administration of the compounds of this invention include, but are not limited to, hard and soft gelatin capsules, tablets, parenteral injectables, and oral suspensions.
A large number of unit capsules are prepared by filling standard two-piece hard gelatin capsules each with 100 milligrams of powdered active ingredient, 150 milligrams of lactose, 50 milligrams of cellulose, and 6 milligrams magnesium stearate.
A mixture of active ingredient in a digestible oil such as soybean oil, cottonseed oil or olive oil is prepared and injected by means of a positive displacement pump into gelatin to form soft gelatin capsules containing 100 milligrams of the active ingredient. The capsules are washed and dried.
A large number of tablets are prepared by conventional procedures so that the dosage unit is 100 milligrams of active ingredient, 0.2 milligrams of colloidal silicon dioxide, 5 milligrams of magnesium stearate, 275 milligrams of microcrystalline cellulose, 11 milligrams of starch and 98.8 milligrams of lactose. Appropriate coatings may be applied to increase palatability or delay absorption.
A parenteral composition suitable for administration by injection is prepared by stirring 1.5% by weight of active ingredient in 10% by volume propylene glycol. The solution is made to volume with water for injection and sterilized.
An aqueous suspension is prepared for oral administration so that each 5 milliliters contain 100 milligrams of finely divided active ingredient, 100 milligrams of sodium carboxymethyl cellulose, 5 milligrams of sodium benzoate, 1.0 grams of sorbitol solution, U.S.P., and 0.025 milliliters of vanillin.
The same dosage forms can generally be used when the compounds of this invention are administered stepwise or in conjunction with another therapeutic agent. When drugs are administered in physical combination, the dosage form and administration route should be selected depending on the compatibility of the combined drugs. Thus the term
coadministration is understood to include the administration of the two agents concomitantly or sequentially, or alternatively as a fixed dose combination of the two active components.
The present invention also relates to a pharmaceutical composition comprising compounds or pharmaceutically acceptable salts thereof having the general formula I in admixture with pharmaceutically acceptable auxiliaries and optionally other therapeutic agents. The auxiliaries must be "acceptable" in the sense of being compatible with the other ingredients of the composition and not deleterious to the recipients thereof.
The invention further includes a pharmaceutical composition, as hereinbefore described, in combination with packaging material suitable for said composition, said packaging material including instructions for the use of the composition for the use as hereinbefore described.
The exact dose and regimen of administration of the active ingredient, or a
pharmaceutical composition thereof, may vary with the particular compound, the route of administration, and the age and condition of the individual subject to whom the medicament is to be administered.
In general parenteral administration requires lower dosages than other methods of administration which are more dependent upon absorption. However, a dosage for humans preferably contains 0.0001-100 mg per kg body weight. The desired dose may be presented as one dose or as multiple subdoses administered at appropriate intervals throughout the day. The dosage as well as the regimen of administration may differ between a female and a male recipient.
Combination Therapy
Compounds of the present invention, and their salts and solvates, and physiologically functional derivatives thereof, may be employed alone or in combination with other therapeutic agents for the treatment of diseases and conditions associated with inappropriate IL-17 pathway activity. Combination therapies according to the present invention thus comprise the administration of at least one compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof, or a physiologically functional derivative thereof, and the use of at least one other pharmaceutically active agent. The compound(s) of formula (I) and the other pharmaceutically active agent(s) may be administered together or separately and, when administered separately this may occur simultaneously or sequentially in any order. The amounts of the compound(s) of formula (I) and the other pharmaceutically active agent(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect. For the treatment of the inflammatory and autoimmune diseases, rheumatoid arthritis, psoriasis, inflammatory bowel disease, ankylosing spondylitis, SLE, uveitis, atopic dermatitis, COPD, asthma and allergic rhinitis a compound of formula (I) may be combined with one or more other active agents such as: (1) T F-a inhibitors; (2) nonselective COX-I/COX-2 inhibitors; (3) COX-2 inhibitors; (4) other agents for treatment of inflammatory and autoimmune diseases including glucocorticoids, methotrexate, leflunomide, sulfasalazine, azathioprine, cyclosporin, tacrolimus, penicillamine, bucillamine, actarit, mizoribine, lobenzarit, ciclesonide, hydroxychloroquine, d-penicillamine, aurothiomalate, auranofin or parenteral or oral gold, cyclophosphamide, Lymphostat-B, BAFF/ APRIL inhibitors and CTLA-4-Ig or mimetics thereof; (5) leukotriene biosynthesis inhibitor, 5- lipoxygenase (5-LO) inhibitor or 5 -lipoxygenase activating protein (FLAP) antagonist; (6) LTD4 receptor antagonist; (7) PDE4 inhibitor; (8) antihistamine HI receptor antagonists; (9) al- and a2-adrenoceptor agonist; (10) anticholinergic agents; (1 1) β-adrenoceptor agonists; (12) insulin-like growth factor type I (IGF-1) mimetic; (13) glucocorticosteroids; (14) kinase inhibitors such as inhibitors of the Janus Kinases (JAK 1 and/or JAK2 and/or JAK 3 and/or TYK2), p38 MAPK and IKK2; ( 15) B-cell targeting biologies such as rituximab; (16) selective costimulation modulators such as abatacept; (17) interleukin inhibitors, such as IL-1 inhibitor anakinra, IL-6 inhibitor tocilizumab, and IL12/IL-23 inhibitor ustekinumab. It could also be combined with anti-IL17 antibodies to obtain additive/ synergistic responses for the treatment of inflammatory and autoimmune diseases.
It will be clear to a person skilled in the art that, where appropriate, the other therapeutic ingredient(s) may be used in the form of salts, for example as alkali metal or amine salts or as acid addition salts, or prodrugs, or as esters, for example lower alkyl esters, or as solvates, for example hydrates, to optimise the activity and/or stability and/or physical characteristics, such as solubility, of the therapeutic ingredient. It will be clear also that, where appropriate, the therapeutic ingredients may be used in optically pure form.
The combinations referred to above may conveniently be presented for use in the form of a pharmaceutical composition and thus pharmaceutical compositions comprising a combination as defined above together with a pharmaceutically acceptable diluent or carrier represent a further aspect of the invention. These combinations are of particular interest in respiratory diseases and are conveniently adapted for inhaled or intranasal delivery.
The individual compounds of such combinations may be administered either sequentially or simultaneously in separate or combined pharmaceutical compositions.
Preferably, the individual compounds will be administered simultaneously in a combined pharmaceutical composition. Appropriate doses of known therapeutic agents will be readily appreciated by those skilled in the art.
Accordingly, the pharmaceutical compositions of the present invention include those that also comprise at least one additional therapeutically active agent, in addition to the compound of Formula I, la, lb, Ic, Id, Ie, If, Ig or Ih.
The invention further includes a compound of Formula I in combination with one or more other drug(s).
METHODS OF SYNTHESIS
Methods for preparing the compounds of this invention are illustrated in the following schemes and examples. Other synthetic protocols will be readily apparent to those skilled in the art. The examples illustrate the preparation of the compounds of Formula I and as such are not to be considered as limiting the invention set forth in the claims appended hereto. Unless otherwise indicated, all variables are as previously defined.
All the end products of the formula I were analyzed by NMR, LCMS. Intermediates were analyzed by NMR and/or TLC and/or LCMS. Most compounds were purified by reverse phase HPLC, MPLC on silica gel, recrystallization and/or swish (suspension in a solvent followed by filtration of the solid). The course of the reactions was followed by thin layer chromatography (TLC) and/or LCMS and/or NMR and reaction times are given for illustration only.
Abbreviations used herein are as follows: EtOAc: Ethyl acetate; PE: Petroleum ether; EA: Ethyl acetate; DCM: Dichloro methane; DMF: Ν,Ν-Dimethylformamide; Dppf 1, 1 '- Bis(diphenylphosphino)ferrocene; AcOH: Acetic acid; DMAC: N,N -Dimethylacetamide; DMAP: N,N-dimethylpyridin-4-amine; TEA: Triethylamine; PYAOP: (7-Azabenzotriazol-l- yloxy)tripyrrolidinophosphonium hexafluorophosphat e;
Pd(PPh3)4:Tetrakis(Triphenylphosphine)Palladium(0); Pd(dppf)Cl2: [1, l'-Bis(diphe nylphosphino) ferrocene ]dichloropalladium (II); Pd2(dba)3: Tris(dibenzylideneace tone)dipalladium(O); BnBr: Benzyl bromide; Ac20: Acetic an hydride; LiHMDS: Lithium bis(trimethylsilyl)amide; PhNTf2: N-Phenyl-bis(trifluoromethane sulfonimide); S-Phos: 2- Dicyclohexylphosphino-2',6'-dimethoxybiphenyl; X-Phos: 2-Dicyclohexylphosphino- 2',4',6'-triisopropylbiphenyl; CPME: Cyclopentyl methyl ether.
Scheme 1 illustrates a genera method toward the preparation of compounds of formula I. Starting from halide A, N-acylation with either carboxylic acids or corresponding acid chloride in the presence of base led to the formation of compound B. Subsequent Suzuki coupling with pinacol boronic ester or acid followd by ester hydrolysis afforded the final compound. In certain cases, ester hydrosis occurred under the Suzuki coupling condition and led to the formation of final product within one pot..
SCHEME 1
C02alkyl
Figure imgf000044_0001
B
Alternatively the final compound I could also be prepared by switching the order of reaction sequence between acylation and Suzuki coupling (see Scheme 2). Suzuki coupling first by reacting halide A with pinacol boronic ester or acid gave intermediate B. Subsequent acylation followed by hydrolysis furnished final product. In some cases where the amide was unstable under hydrolysis conditions, the ester moiety could be hydrolyzed first followed by reacting with acid or acid chloride to give the final product.
SCHEME 2
Figure imgf000044_0002
Scheme 3 illustrates a general method for the prepartion of compounds of formula I , which contain an amide moiety at A position. Starting from halide A, acylation followed by ester hydrolysis gave intermediate B. Subsequent Suzuki coupling afforded acid C. Standard amide coupling followed by hydrolysis led to the formation of the final product.
Figure imgf000045_0001
Scheme 4 illustrates a general method for compounds containing aryl or heteroaryl substituents at A6 postion. Starting from halide A, acylation followed by suzuki compound furnished common intermediate B. Subsequent Suzuki coupling and ester hydrolysis gave the final product. Alternatively, Compound B could be converted into pinacol boronic ester or acid first, followed by subsequent Suzukin coupling with appropriate aryl or heteraryl halide and hydrolysis delivered the final product.
Figure imgf000046_0001
Scheme 5 illustrates a general method for the preparation of compounds containing amine or lactam moiety at A6 position. Starting from common intermedaite A (see Scheme 4 for its preparation), Pd-catalzyed reaction with primary or secondary amines or latams followed by ester hydrolysis furnished the final product.
SCHEME 5
Figure imgf000046_0002
I
Scheme 6 illustrates a general method for the preparation of compounds which contain alcohol or ketone moiety at A6 position. Starting with halide A, acylation followed by reduction of the ester moitey with reducing agent (such as DIBAL-H) afforded compound B. Suzuki coupling with boronic ester or acid gave compound C. Oxidation of the primary alcohol, followed by reacting with Grignard reagent and subsequent ester hydrolysis to give the final compound I. Alternatively, oxidation of the product from Grignard addition, followed by ester hydrolysis afford ketone derivative Γ. SCHEME 6
C02alkyl
Figure imgf000047_0001
C I Γ
Scheme 7 illustrates a general method for the preparation of compounds, which contain amide, sulfonamides or carbamate of primary or secondary amines at A6 position. Starting from compound A, acylation followed by reduction of N02 with reducing agent (such as SnCl2) afforded compound B. Subsequent Suzuki coupling gave common intermedate C. Stardard amide, sulfonamide, or carbamate formation reactions followed by ester hydrolysis gave the final compounds I. Alternatively, Compound C could be alkylated first with halide in the presence of base to afford a new intermediate which contains a secondray amine. Subsequent standard formation of amides, sulfonamides and carbamates and hydrolysis furnish the final compounds.
Figure imgf000048_0001
or Sulfonyl chloride, base
Scheme 8 illustrates a general strategy for the preparation of compounds, which contain heteroaryl substituetns at A6 poistion, but couldn't access through Suzuki coupling as shown in Scheme 4. Starting from carboxylic acid A (see scheme 4 for its synthesis), amide coupling afforded intermediate B. Subsequent cyclization by reacting with POCl3 in the presence of pyridine, followed by ester hydrolysis led to the formation of final product I, which contains an oxazole substituent at A6 position. The same stratetgy was also used for the synthesis of a number of other analogs contain a different heterocyles at ths position. Construction of those heterocyles can follow those well-known routes in the literature.
SCHEME 8
Figure imgf000049_0001
COMMERCIALLY AVAILABLE / PREVIOUSLY DESCRIBED MATERIALS The following table lists commercial sources, and previously disclosed synthetic routes for chemical materials employed in the synthesis of intermediates, and examples of the instant invention. The list is not intended to be exhaustive, exclusive, or limiting in any way.
Figure imgf000049_0002
Br Bellen H
Aldrich
■f Br Labpartner
H
1 Labpartner
H
Labpartner
Figure imgf000050_0001
Br Labpartner
H
Br
Λ / Sinova
H
Labpartner
H
1 Labpartner
H
°YCI Alfa
CIY CF3
Figure imgf000051_0001
Combi-blocks
Anisyn
Aldrich
Figure imgf000052_0001
Acros
Alfa Aesar
ADAMAS
Acros
Figure imgf000052_0002
Aldrich
Combi-blocks
H
°YCI Alfa
CIY CF3
Figure imgf000053_0001
INTERMEDIATES
Preparation of (E)-2-chloro-6-(prop-l-enyl)benzoic acid
SCHEME i-1
Figure imgf000054_0001
i-1
Step 1. Preparation of methyl 2-bromo-6-chlorobenzoate (i-lb). LiOH.H20 (180 mg, 4.24 mmol) was added to a solution of 2-bromo-6-chlorobenzoic acid (i-la) (1.0 g, 4.24 mmol) in THF (30 ml). The mixture was stirred at 25 °C for lh. Then the Me2S04 (1.1 g, 8.48 mmol) was added to the reaction mixture. The mixture was warmed to 85 °C and stirred at 85 °C for 21 h. After cooled, H3.H20 was added dropwise to the mixture until pH=7-8. The solution was poured into water and THF was evaporated. The water layer was extracted with EA (60 ml). The organic layer was dried over Na2S04 and concentrated to obtain 800 mg (75%) of the title compound. LCMS (ESI): calc'd for C8H6BrC102 [M+H]+: 251, found: 251.
Step 2. Preparation of methyl 2-chloro-6-cyclopropylbenzoate (i-lc). Methyl 2-bromo-6- chlorobenzoate (i-lb) (0.8 g, 3.2 mmol), cyclopropylboronic acid (330 mg, 3.84 mmol), Pd(OAc)2 (72 mg, 0.32 mmol), Cy3P (180 mg, 0.64 mmol) and K3P04 (2.0 g, 9.6 mmol) were mixed in toluene (12 ml) and H20 (1.2 ml). The reaction mixture was stirred at 100 °C for overnight under N2 atmosphere. After cooled, the mixture was poured into water (30 ml) and extracted with EA (50 ml). The organic layer was dried over Na2S04 and concentrated to obtain a residue. The residue was purified by chromatography on silica gel (PE/EA=10: 1) to obtain 350 mg (52%) of the title compound. LCMS (ESI): calc'd for CnHnC102 [M+H] : 211, found: 211.
Step 3. Preparation of (E)-2-chloro-6-(prop-l-enyl)benzoic acid (i-1). To a suspension of anhydrous Mgl2 (880 mg, 3.18 mmol) in toluene (15 ml) was added a solution of methyl 2- chloro-6-cyclopropylbenzoate (i-lc) (400 mg, 1.9 mmol) in toluene (5 ml). The mixture was refluxed under exclusion of moisture, cooled and poured into 10% aqueous NaHC03 (20 ml). The acid was isolated by acidification of the aqueous phase with 15% HCl followed by extraction with EA. The combined organic layers were washed with water, dried over Na2S04 and concentrated to obtain the crude product. The crude product was purified by chromatography on silica gel (PE/EA=3 : 1) to obtain 200 mg (54%) of the title compound. LCMS (ESI): calc'd for Ci0H9ClO2 [M+H]+: 197, found: 197.
SCHEME i-2
Figure imgf000055_0001
i-2d i-2
Preparation of methyl 2-chloro-6-hydroxybenzoate (i-2b). To the solution of 2-chloro-6- hydroxybenzoic acid (i-2a) (1.71 g, 10 mmol) in CH3OH (100 ml) was added concentrated sulfuric acid (10 ml) drop wise. The mixture solution was protected by N2 and stirred at 85 °C for 30 h. the solution was concentrated to be purified by chromatography column (EA:PE=1 :4) to afford 1.5 g product (81%). LCMS (ESI) calc'd [M+H]+: 186.59, found: 187.0. Step 2. Preparation of methyl 2-chloro-6-cyclopropoxybenzoate (i-2c). The mixture of i- 2b (186 mg, lmmol), bromocyclopropane (1.2 g, 10.0 mmol), Cs2C03 (414 mg, 3.0 mmol), DM AC (15 ml) was protected by N2 and stirred at 150 °C for 24 h. Then the solution was filtered and concentrated to be purified by chromatography column (EA: PE =1 :4) to afford 198 mg product (87%). LCMS (ESI) calc'd [M+H]+: 226.66, found: 227.1.
Step 3. Preparation of methyl 2-chloro-6-cyclopropoxybenzoic acid (i-2d). To the solution of 2-chloro-6-cyclopropoxybenzoate (i-2c) (226mg, 1.0 mmol) in CH3OH (10 ml) and H20 (10 ml) was added KOH (200 mg). The mixture solution was stirred at 85 °C for 12 h, the solution was acidified by aqueous HCl (1M) 50 ml, exacted with EA (30ml χ 3), concentrated and purified by chromatography column (EA:PE=1 : 1) to afford 186 mg product (87%). LCMS (ESI) calc'd [M+H]+: 212.63, found: 212.9.
Step 4. Preparation of 2-chloro-6-cyclopropoxybenzoyl chloride (i-2). To the solution of methyl 2-chloro-6-cyclopropoxybenzoic acid (i-2d) (212 mg, 1 mmol) and DMF (0.1 ml) dissolved in anhydrous DCM (10 ml) was added oxalyl chloride (190 mg, 1.5 mmol) drop wise. The mixture solution was protected by N2 and stirred at r.t for 0.5 h. Then the solution was concentrated to afford 300 mg (crude) for the next step without further purification.
Figure imgf000057_0001
Figure imgf000057_0002
1A
Step 1. Preparation of 4-bromo-3-hydroxy-2-nitrobenzoic acid (i-5b). To a suspension of 3-hydroxy-2-nitrobenzoic acid (i-5a) (2.0 g, 10.9 mmol) in acetic acid (4 mL) was added a solution of bromine (0.59 mL, 11.4 mmol) in acetic acid (3 mL) dropwise via a addition funnel over 0.5 h, then the reaction mixture was stirred in the dark at 60 °C. for 12 h. After cooled to room temperature, the reaction mixture was concentrated to give the desired product as a yellow solid (contained 30% of di-brominated products). The crude product was dissolved in THF (90 mL) and a solution of sodium hydrosulfite (11.2 g, 54.5 mmol) in water (50 mL) was added. The reaction mixture was stirred at 60 °C for 40 min. After the reaction mixture was cooled to room temperature, the aqueous layer was separated and extracted with ethyl acetate. The combined organics were dried with Na2S04, filtered and concentrated. The crude material was purified by column chromatography (PE:EA =3 : 1) to afford the desired product (1.17 g, 46%) as a pale-brown solid. LCMS (ESI) calc'd [M+H]+: 261.9, found: 262.
Step 2. Preparation of methyl 4-bromo-3-hydroxy-2-nitrobenzoate (i-5c). To the solution of methyl 4-bromo-3-hydroxy-2-nitrobenzoic acid (i-5b) (l . lg, 4.2mmol) in MeOH (30 ml), 2ml of 98%) sulfuric acid was added dropwise. The mixture solution was stirred at 90 °C for 16h. Cooled to RT, 200ml EA was added, washed with H2O(20ml x 3) and brine (50ml), dried with Na2S04, concentrated to afford 1.1 g product (yield: 95%). LCMS (ESI) calc'd [M+H]+: 274.9, found: 276.
Step 3. Preparation of methyl 2-amino-4-bromo-3-hydroxybenzoate (i-5d). To the solution of methyl 4-bromo-3-hydroxy-2-nitrobenzoate (i-5c) (l . lmg, 4 mmol) in HOAc(15ml) was added Fe( 670mg, 12mmol) , the mixture was stirring for 1 h at 60 °C. Cooled to RT, 100ml EA was added, filtered and concentrated. Purified with chromatogram (PE:EA=5: 1) to afford 960mg product (97%). LCMS (ESI) calc'd [M+H]+: 244.97, found: 246.
Step 4. Preparation of methyl 7-bromo-2-oxo-2,3-dihydrobenzo [d]oxazole -4- carboxylate (i-5e). Methyl 2-amino-4-bromo-3-hydroxybenzoate (i-5d) (960mg, 3.9 mmmol) in THF (5ml) was added CDI (956mg, 5.9mmmol) . The mixture was stirred at 80 °C for 3h. The solution was concentrated and purified with chromatogram (PE:EA=5: 1) to afford 950 mg product (95%). LCMS (ESI) calc'd [M+H]+: 270.95, found: 272.
Step 5. Preparation of methyl 2-oxo-7-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan -2-yl)-2,3- dihydrobenzo[d]oxazole-4-carboxylate (i-5). Methyl 7-bromo-2 -oxo-2,3- dihydrobenzo[d]oxazole-4-carboxylate (i-5e) (950mg, 3.5mmmol), Pin2B2(1.78g, 7 mmol), KOAc(1.37g, 14mmol) and (dppf)PdCl2 (256mg, 0.35mmol) were mixed in dioxane(15ml) under N2 protection. The mixture was stirred at 80 °C for 12h. The solution was concentrated and concentrated, purified with chromatogram (PE:EA=5: 1) to afford 670 mg product (95%). LCMS (ESI) calc'd [M+H]+: 270.9, found: 272.
SCHEME i-6
Figure imgf000059_0001
Step 1. Preparation of 4-bromo-2,5-difluorobenzoic acid (i-6b). To the solution of 1,4- dibromo-2,5-difluorobenzene (i-6a) (27.0 g, 100 mmol) in THF(500 ml) was
added n-BuLi (60 ml, 2M) dropwise at -78°C and stirred for 3 h which was protected by N2 .Then the solution was poured to excess dry ice over 0.5 h. Added 300 ml water to the solution, washed with EA (100 mix 3). The aqueous solution was acidified with HC1 (2M), extracted with EA (150 ml><3) and the organic layer was dried and concentrated. The crude material was purified by chromatography column (EA:PE = 1 : 10) to afford 18.24 g product (76%). LCMS (ESI) calc'd [M+H]+: 237.00, found: 237.1.
Step 2. Preparation of methyl 4-bromo-2,5-difluorobenzoate (i-6c). To the solution of 4- bromo-2,5-difluorobenzoic acid (i-6b) (18.24 g, 77.3 mmol) in CH3OH (200 ml) was added Concentrated sulfuric acid (5 ml) drop wise. The mixture solution was protected by N2 and stirred at 85 °C for 30 h. The solution was concentrated and purified by chromatography column (EA:PE=1 :20) to afford 17.6 g product (91%). LCMS (ESI) calc'd [M+H]+: 251.01, found: 250.9.
Step 3. Preparation of methyl 4-bromo-5-fluoro-2-methoxybenzoate (i-6d). To a solution of methyl 4-bromo-2,5-difluorobenzoate (i-6c) (17.6 g, 70.4 mmol) in anhydrous DMF (200 ml) was added CH3ONa (4.56g, 84.48 mmol). The mixture solution was protected by N2 and stirred at rt for 16 h.. The reaction was diluted with 500 ml EA and washed with water (100mlx3). The organic layer was dried and cone- entrated to be purified by chromatography column (EA:PE=1 :20) to afford 14.2 g product (77%). LCMS (ESI) calc'd [M+H]+: 263.06, found: 263.01.
Step 4. Preparation of 2 methyl 5-fluoro-2-methoxy-4-(4,4,5,5-tetramethyl -1,3,2- dioxaborolan-2-yl)benzoate (i-6). The mixture of methyl 4-bromo-5-fluoro-2-metho- xybenzoate (i-6d) (14.2 mg, 54.2mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(l,3,2- dioxaborolane) (13.8 g, 54.2 mmol), Pd(dppf)2Cl2 ( 224mg, 0.2 mmol), Dioxane(150 ml) was degassed and protected by N2 and stirred at 80 °C for 4 h. Then the solution was filtered and concentrated to be purified by chromatography column (EA:PE=1 :4) to afford 10.6 g product (63%). LCMS (ESI) calc'd [M+H]+: 310.13, found: 310.2.
SC H EME i-7
Figure imgf000060_0001
i-7
Step 1. Preparation of methyl 2-amino-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborola- n-2- yl)benzoate (i-7). To a mixture of methyl 2-amino -4-bromobenzoate (i-7a) (1.15 g, 5 mmol), 4,4,4',4',5,5,5',5'-octamethyl -2,2'-bi(l,3,2-dioxaborolane) (1.27 g, 5 mmol), Pd(dppf)Cl2 (204 mg, 0.25 mmol), dppf(138 mg, 0.25 mmol) and KOAc (1.47g, 15 mmol) was added dioxane (20 ml), and the mixture was heated at 100° C under argon for 2 h. The mixture was cooled down, diluted with CH2C12 (50 mL). The organic layer was separated, washed with brine, dried over Na2S04, concentrated. The residue was purified by flash chromatography (Pentane/EtOAc 5/1) to give 1.5 g (89 %) of the title compound. 1HNMR (500 MHz,
CDCL3) 57.84 (1H, d),7.12 (1H, s), 7.05(lH,d),5.68(2H, bs), 3.86( 3H, s), 1.33Q2H, s).
Figure imgf000061_0001
Figure imgf000061_0002
Step 1. Preparation of methyl lH-indazole-6-carboxylate (i-8b). Methyl 3-amino-4- methylbenzoate (i-8a) (5.0 g, 30.2 mmol) was dissolved in AcOH (140 mL). Sodium nitrite (2.1 g, 30.2 mmol) in water (3.5 mL) was added dropwise to the solution of starting material under ice-cooling at room temperature. The icebath was removed and the mixture was stirred overnight. Half of the solvents were then evaporated, the mixture was diluted with water (80 mL) and extracted with EtOAc (3 x 30 mL). The collected organic phase was washed with water and brine ( 2 x 200 mL ), dried and evaporated to afford i-8b (4.4 g, 83%). LCMS (ESI): calc'd for C9H8N202, [M+H]+: 177, found: 177.
Step 2. Preparation of methyl 3-iodo-lH-indazole-6-carboxylate (i-8c). Methyl 1H- indazole-6-carboxylate (i-8b) (5.0 g, 28.3 mmol) was dissolved in anhydrous DMAC (50 mL). Iodine (14.4 g, 56.7 mmol) and potassium hydroxide (6.3 g, 113.5 mmol) were added in portions under ice-cooling at room temperature. The ice bath was removed and the mixture was stirred at room temperature for lh. The reaction was monitored by TLC (25% MeOH in chloroform) then it was slowly quenched with Sat. Na2S203 aqueous (100 mL), diluted with water (50 mL) and extracted with EtOAc (3 x 100 mL). The organic phase was evaporated and triturated with n-hexane. The precipitated material was filtered and dried to afford a brown solid i-8c (5.3 g, 62%). LCMS (ESI): calc'd for C9H7IN202, [M+H]+: 303, found: 303. Step 3. Preparation of methyl l-(2-chloro-6-(trifluoromethyl)benzoyl) -3-iodo- 1H- indazole-6-carboxylate (i-8d). To a 250 mL round-bottomed flask, was added Methyl 3- iodo-lH-indazole-6-carboxylate (i-8c) (11.7 g, 38.7 mmol), 2-chloro-6-(trif- luoromethyl)benzoyl chloride (9.1 g, 38.7 mmol), DMAP (4.72 g, 38.7 mmol) and CH2C12 (30 mL). After stirring at r.t. for 3 minutes, TEA (11.2 mL, 77 mmol) was added slowly. The reaction mixture was stirred at r.t. for overnight. LC-MS showed no starting materials remained. Then the mixture was poured to 30 mL water, the lower (organic) and upper (aqueous) phases are separated. The aqueous phase is extracted twice with 20 mL CH2C12. The combined organic phases are washed successively with two 20 mL portions of water and 10 mL of brine. The reaction resulting organic phase is dried over anhydrous sodium sulfate, filtered and concentrated at reduced pressure to give a yellow solid. The residue was purified by column chromatography on 60 g of silica gel eluting with Petroleum ether /EtOAc from 50/1 to 10/1, to give a fawn solid i-8d (16.5 g, 84%). LCMS (ESI): calc'd for Ci7H9ClF3IN203, [M+H]+: 509, found: 509.
Step 4. Preparation of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-iodo-lH-indazo- le-6- carboxylic acid (i-8). A mixture of methyl l-(2-chloro-6-(trifluoromethyl) benz- oyl)-3-iodo- lH-indazole-6-carboxylate (i-8d) (16.5g, 32.5 mmol) and LiOH (3.40g, 162.40 mmol) in 10 ml THF and 50 ml H20 was stirred at RT overnight. The solvent was evaporated and the residue was dissolved in water. 5% HCl aqueous was added until pH~4-5. The precipitated solid was filtered, washed with water and n-hexane, dried to afford an off white solid i-8 (16.0g, 83%). LCMS (ESI): calc'd for Ci6H7ClF3IN203, , [M+H]+: 495, found: 495.
Figure imgf000063_0001
Figure imgf000063_0002
Step 1. Preparation of 6-bromo-3-iodo-lH-indazole (i-9b). To a flask was added 6-bromo- lH-indazole (i-9a) (1.96 g, 10 mmol), KOH (1.68 g, 30 mmol) and DMF (60 mL), followed by the addition of I2 (5.08 g, 20 mmol) in portions. The reaction mixture was stirred at RT for 1 h. The mixture was diluted with H20, and the organic layer was separated. The aqueous layer was extracted with CH2C12 (3 * 50 mL). The combined organics were washed with H20, brine, dried over Na2S04, and concentrated. The residue was purified by column chromatograph (PE/EA=10/1) to afford 2.84 g (88%) of the title compound. LCMS (ESI) calc'd for C7H4BrIN2 [M+H]+: 322.86, found: 323.
Step 2. Preparation of (6-bromo-3-iodo-lH-indazol-l-yl)(2-chloro-6-(trifluorom- ethyl)phenyl)methanone (i-9c). To a flask was added 6-bromo-3-iodo-lH-indazol e (i-9b) (3.22 g, 10 mmol), DMAP (1.22 g, 10 mmol), TEA (2.77 mL, 20 mmol) and DCM (50 mL), followed by the addition of 2-chloro-6-(trifluoromethyl) benzoyl chloride (2.61 g, 10 mmol) slowly. The reaction mixture was stirred at RT for 5 h. The mixture was diluted with H20, and the organic layer was separated. The aqueous layer was extracted with CH2C12 (3 x 50 mL), The combined organics were washed with H20, brine, dried over Na2S04, and concentrated. The residue was purified by column chromatograph (PE/EA=10/1) to afford 4.9 g (82 %) of the title compound. LCMS (ESI) calc'd for Ci5H6BrClF3IN20 [M+H] : 528.83, found: 529.
Step 3. Preparation of methyl 4-(6-bromo-l-(2-chloro-6-(trifluoromethyl) benzo- yl)-lH- indazol-3-yl)benzoate (i-9). To a mixture of (6-bromo-3-iodo-lH-inda zol-l-yl)(2-chloro-6- (trifluoromethyl)phenyl)methanone (i-9c) (1.058 g, 2 mmol), 4-
(methoxycarbonyl)phenylboronic acid (360 mg, 2 mmol), PdCl2(dppf)2 (82 mg, 0.1 mmol) and KF (290 mg, 5 mmol) was added dioxane (25 ml) and H20 (0.5 ml), and the mixture was heated at 90 °C under argon for 6 h. The mixture was cooled down, diluted with CH2C12 (180 ml). The organic layer was separated, washed with brine, dried over Na2S04, concentrated. The residue was purified by Si02 (PE/EA fromlO/1 to 20/1) to give 850 mg (81%) of the title compound. LCMS (ESI) calc'd for C^HoBrClFsN^ [M+H] +: 536.98, found: 537.
EXAMPLES
Example 1A: Preparation of (E)-4-(l-(2-chloro-6-(prop-l-enyl)benzoyl) -1H- pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoic acid (1A)
SCHEME A.
Figure imgf000065_0001
Figure imgf000065_0002
Step 1. Preparation of methyl 3-fluoro-4-(lH-pyrazolo [4, 3-b]pyridin-3-yl) benzo- ate(A-2). A mixture of 3-bromo-lH-pyrazolo[4,3-b]pyridine (A-1) (196.9 mg, 1 mol), 4- (methoxycarbonyl)phenylboronic acid (198 mg, 1 mol), Pd(PPh3)4 (115 mg, 0.1 mol) and K2C03 ( 420 mg, 3mol) were suspended in 1,4-dioxane (5 ml) and H20 (1 ml). The reation mixture was heated at 110°C in a microwave reactor for 2h. The result mixture was diluted with H20 (30 ml) and the aqueous layer was extracted with ethyl acetate (30 ml χ 2). The combined organic layers were washed with brine (30 ml x 1), dried over anhydrous Na2S04 and concentrated to get the crude product A-2 as brown oil. LCMS (ESI) calc'd for
C14H10FN3O2 [M+H] +: 272.08, found: 272.
Step 2. Preparation of (E)-methyl 4-(l-(2-chloro-6-(prop-l-enyl)benzoyl)-lH- pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoate (A-3). (COCl)2 (907 mg, 7.14 mmol) was added dropwise to a solution of (E)-2-chloro-6-(prop-l-enyl)benzoic acid (i-1) (700 mg, 3.57 mmol) and 3 drops of DMF in DCM (20 ml). The resulted solution was stirred at room temperature for 30 min. Then the solution was added to a solution of methyl 3-fluoro-4-(lH- pyrazolo[4,3-b]pyridin-3-yl)benzoate (A-2) (968 mg, 3.57 mmol), Et3N (720 mg, 7.14 mmol) and DMAP (436 mg, 3.57 mmol) in DCM (20 ml). The solution was stirred at room temperature for 3h. Then the reaction mixture was poured into water and separated. The organic layer was dried over Na2S04 and concentrated to obtain a crude product. The crude product was purified by on silica gel (PE/EA=6: 1) to afford 133 mg of the title compound. LCMS (ESI) calc'd for C24Hi7ClFN303 [M+H]+: 450, found: 450.
Step 3. Preparation of (E)-4-(l-(2-chloro-6-(prop-l-enyl)benzoyl)-lH -pyrazolo [4,3- b]pyridin-3-yl)-3-fluorobenzoic acid (1A). To a solution of (E)-methyl 4-(l-(2- chloro-6- (prop-l-enyl)benzoyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoate (A-3) (120 mg, 2.67 mmol) in THF (15mL) and H20 (15mL) was added LiOH.H20 (112 mg, 2.67 mmol), and the mixture was stirred at 30 °C for 2h. The mixture was neutralized with 2N HC1 to pH = 3-4. The mixture was extracted with EA (50 ml). The organic layer was dried over Na2S04 and concentrated to obtain a crude product. The crude product was purified by pre-HPLC to afford 17 mg (15%) of the title compound as white solid. LCMS (ESI): calc'd for C23Hi5ClFN303 [M+H]+: 436, found: 436; 1HNMR (400 MHz, DMSO) δ 8.94 (2H, m), 8.26- 8.23 (1H, t), 7.91-7.72 (4H, m), 7.57-7.48 (2H, m), 6.45-6.43 (1H, m), 6.34-6.30 (1H, d), 1.75-1.74 (3H, d).
EXAMPLE IB: Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-4- formyl-lH- indazol-3-yl)benzoic acid (IB)
HEME B.
Figure imgf000066_0001
Step 1. Preparation of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-iodo-lH -indazole-4- carbaldehyde (B-3). The mixture of 2-chloro-6-(trifluoromethyl)benzoic acid (B-2) (0.46 g, 2.07 mol) and (COCl)2 (0.32 mL, 3.76 mol) in DCM (10 mL) and DMF (5drops) was stirred at room temperature for lh. The solvent was removed and the residue was dissolved in DCM (10 mL). To the mixture of 3-iodo-lH-indazole-4-carbaldehyde (B-l) (0.51 g, 1.88 mol), DMAP (23 mg,0.19 mol) and Et3N (0.52 mL, 3.76 mol) in DCM (10 mL) was added the aboved DCM solution dropwise and the reaction mixture was stirred at room temperature overnight. The result solution was diluted with H20 (50 mL) and the aqueous layer was extracted with DCM (30 mLx3). The combined organic layers were washed with brine (30 mLx l),dried over anhydrous Na2S04 and concentrated. The residue was chromatographed on silica gel(PE:EA 8: 1 to 4: 1) to get the desired product B-3 as a white solid. LCMS (ESI) calc'd for Ci6H7ClF3lN202 [M+H]+: 479, found: 479.
Step 2. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-4- formyl-lH- indazol-3-yl)benzoate (B-5). A mixture of l-(2-chloro-6- (trifluorom- ethyl)benzoyl)-3-iodo- lH-indazole-4-carbaldehyde (B-3) (0.5 g, 1.05 mol), 4-(meth- oxycarbonyl)phenylboronic acid (B-4) (0.28 g, 1.57 mol), Pd(PPh3)4 (0.13 g, 0.11 mol) and K2C03 (0.43 g, 3.15 mol) were suspended in 1,4-dioxane (10 mL) and H20 (2 mL). The reation mixture was heated at 100 °C in a microwave reactor for 1.5h. The result mixture was diluted with H20 (50 mL) and the aqueous layer was extracted with ethyl acetate (50 mL χ 3). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2S04 and concentrated. The residue was chromatographed on silica gel (PE:EA = 4: 1) to get the desired product B-5 as a pale yellow solid. LCMS (ESI) calc'd for C24Hi4ClF3N204 [M+H]+:487, found: 487.
Step 3. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-4-formyl- lH-indazol- 3-yl)benzoic acid (IB). The mixture of methyl 4-(l-(2-chloro- 6-(trifluoromethyl)benzoyl)-4- formyl-lH-indazol-3-yl)benzoate (B-5) (40 mg, 0.08 mol) and LiOH (17 mg, 0.41 mol) in THF (4 mL) and H20 (2 mL) was stirred at room temperature for 4h. The reaction mixture was diluted with H20 (20 mL). 2M HC1 solution was added to adjust the pH=3 and the aqueous layer was extracted with ethyl acetate (20 mL χ 3). The combined organic layers were washed with brine (20 mL x 1), dried over anhydrous Na2S04 and concentrated. The residue was purified with Prep-HPLC to get the desired product IB as a white solid. LCMS (ESI): calc'd for C23Hi2ClF3N204 [M+H]+: 473, found: 473; 1HNMR (400 MHz, DMSO) δ 10.07 (1H, s), 8.89 (1H, d, J=8.4Hz), 8.13 (1H, d, J=7.2Hz), 8.03-8.07 (4H, m), 7.99 (1H, d, J=8.0Hz), 7.83-7.87 (1H, m), 7.70 (2H, d, J=8.4Hz).
EXAMPLE ID: Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-lH- pyrazolo[4,3-b]pyridin-3-yl)-lH-indazole-7-carboxylic acid (ID) SCHEME D.
Figure imgf000068_0001
1D
Step 1. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-lH-pyrazolo [4,3- b]pyridin-3-yl)-lH-indazole-7-carboxylic acid (ID). To a 1 dram vial was added methyl 3-fluoro-4-(lH-indazol-3-yl)benzoate (D-1) (30 mg, O. l l lmmol), LiOH (1M, 0.333ml), methanol (0.25ml) and THF (0.5ml), The reaction mixtures were stirred at RT for 2 hours. The mixtures were then evaporated under reduced pressure. The remaining residues were then dissolved in DCM (0.5ml) and added to other 1 dram vials that contained picolinic acid (27 mg, 0.222mmol), 2-chloro-l,3-dimethylimidazolinium chloride (37.5 mg, 0.222 mmol), and DCM (1 ml) which was stirring at RT for 4 h. The combined mixtures were stirred overnight at RT and then evaporated under reduced pressure. The reactions were then diluted with 1.0 mL DMSO, filtered, and purified by mass-triggered reverse phase HPLC, eluting with a 1% ammonium hydroxide buffered water/acetonitrile gradient over a Waters X-Bridge C-18 column, to afford desired products. LCMS (ESI) calc'd for C2oHi2FN303 [M+H]+: 362.1, found: 362.1.
The following examples shown in TABLE 1 were prepared following similar procedures described for Examples A, B, C, D in Schemes A-D, which can be achieved by those of ordinary skill in the art of organic synthesis.
TABLE 1
Figure imgf000069_0001
Figure imgf000069_0002
Figure imgf000070_0001
Figure imgf000071_0001
EXAMPLE 2A: Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (morpholine-4-carbonyl)-lH-indazol-3-yl)benzoic acid (2A)
SCHEME E
PYAOP
Figure imgf000072_0001
E-2 2A
Step 1. Preparation of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-(4-(methoxy- carbonyl)phenyl)-lH-indazole-6-carboxylic acid (E-1). A mixture of i-8 (300 mg, 0.61 mmol), 4-(methoxycarbonyl)phenylboronic acid (165 mg, 0.92 mmol), Pd(dppf) Cl2 (50 mg, 0.061 mmol) and KOAc (181 mg, 1.83mmol) in 10 ml dioxane and 2 ml pure H20 was heated to 95 °C for 2h with microwave. Then it was diluted with EtOAC (50 ml), washed with brine (50 ml x 2), dried over anhydrous Na2S04, concentrated. Then it was purified by silica gel column (Petroleum ether/EtOAc = 20/1) to get white solid E-1 (180 mg, 59%). LCMS (ESI): calc'd for C24Hi4ClF3N205, [M+H]+: 503.1, found: 503.1.
Step 2. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) -6- (morpholine-4-carbonyl)-lH-indazol-3-yl)benzoate (E-2). The compound E-1 (180 mg, 0.36mmol) was dissolved in CH2C12 (15 mL). morpholine (37mg, 0.43 mmol), PYAOP (374 mg, 0.72mmol) was added and the mixture was stirred at room temperature for 2 mins. TEA (0.16mL, 1.08mmol) was added and the mixture was stirred at room temperature for 2h. Then it was diluted with EtOAC (20 ml), washed with brine (2 x 20 ml), dried over anhydrous Na2S04, concentrated get white solid E-2 (195 mg, 95%). LCMS (ESI): calc'd for C28H2iClF3N305, [M+H]+: 572, found: 572.
Step 3. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (morpholine-4- carbonyl)-lH-indazol-3-yl)benzoic acid (2A). A mixture of E-2 (195mg, 0.34mmol) and LiOH.H20 (72mg, 1.7mmol) in 10 ml THF and 10 ml pure H20 was stirred at RT for 2 hours. The solvent was evaporated and the residue was dissolved in water. HC1 (5% sol in water) was added until pH=4-5. The precipitated solid was filtered, washed with water and n-hexane, dried to afford an off white solid 2A (184mg, 97%). LCMS (ESI): calc'd for C27Hi9ClF3N305, [M+H]+: 558.1 found: 558.1. 1HNMR (400MHz, DMSO) δ 8.55QH, s), 8.32-8.34QH, d, J = 8 Hz), 8.05-8.11(3H, m), 8.01-8.03 (1H, d, J = 8 Hz), 7.95-7.97 (2H, d, J = 8Hz), 7.87-7.91 (1H, m), 7.69-7.71 (1H, d, J = 8 Hz), 3.52-3.72 (8H, m).
The following examples shown in TABLE 2 were prepared following similar procedures described for Examples E in Schemes E, which can be achieved by those of ordinary skill in the art of organic synthesis.
TABLE 2
Figure imgf000073_0001
R4 LCMS [M+H]+
P Q
Ex. Chemical Name R* Found
Figure imgf000074_0001
acid
EXAMPLE 3A: Preparation of 2-acetamido-4-(l-(2-chloro-6-(trifluoromethyl) benzoyl)- lH-indazol -3-yl)benzoic acid (3A).
SCHEME F
Figure imgf000075_0001
F4 3A
Step 1. Preparation of (3-bromo-lH-indazol-l-yl)(2-chloro-6-
(trifluoromethyl)phenyl)methanone. (F-2). To a solution of 3-bromo-lH-indazole (F-1) (200 mg, 1.02 mmol) in DCM (20 mL) was added DMAP (12.5 mg, 0.1 mmol), TEA (0.3 mL, 2 mmol), 2-chloro-6-(trifluoromethyl)benzoyl chloride (370 mg, 1.53 mmol) in DCM (5 mL) was added slowly, the reaction mixture was stirred at RT for 3 h, then diluted with EA (100 mL), washed with Sat. NaHC03 aqueous, water and brine, concentrated and purified with flash chromatograph (PE:EA=10: 1) to give 400 mg (99%) of title compound as a yellow solid. LCMS (ESI) calc'd for Ci5H7BrClF3N20 [M+H]+, 402.9, found: 403, 405.
Step 2. Preparation of methyl 2-amino-4-(l-(2-chloro-6-(trifluoro methyl) benzoyl)-lH- indazol-3-yl)benzoate (F-3). To a mixture of F-2 (110 mg, 0.4 mmol), methyl 2-amino-4- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)benzoate(168 mg, 0.4 mmol), Pd(PPh3)4(46 mg, 0.04 mmol) and K2C03 (138 mg, 1 mmol) was added dioxane (15 ml) and H20 (1 ml), and the mixture was heated at 90° C under argon for 6 h. The mixture was cooled down, diluted with CH2C12 (50 mL). The organic layer was separated, washed with brine, dried over Na2S04, concentrated. The residue was purified by Prep-TLC (Pentane/EtOAc 10/1) to give 155 mg (71%) of the title compound. LCMS (ESI) calc'd for C23Hi5ClF3N303 [M+H]+, 474, found: 474.
Step 2. Preparation of methyl 2-acetamido-4-(l-(2-chloro-6-(trifluoromethyl) benzoyl)- lH-indazol-3-yl)benzoate (F-4). To a flask was added compound F-3 (180mg, 0.38 mmol), acetyl chloride (36 mg, 0.46 mmol), and DCM (30 mL), followed by the addition of TEA (1.3 mL, 0.95 mmol) slowly. The reaction mixture was stirred at rt for 2 h. The mixture was diluted with H20, and the organic layer was separated. The aqueous layer was extracted with CH2C12 (. The combined organics were washed with H20, brine, dried over Na2S04, and concentrated. The residue was purified by Prep-TLC (Pentane/EtOAc, 10/1) to afford 210 g (97 %) of the title compound. LCMS (ESI) calc'd for C25Hi7ClF3N304 [M+H]+: 516, found: 516.
Step 3. Preparation of 2-acetamido-4-(l-(2-chloro-6-(trifluoromethyl) benzoyl) -1H- indazol -3-yl)benzoic acid (3 A). To a stirred solution of compound F-4 (210mg, 0.41 mmol) was added THF (8.0 mL), H20 (2.0 mL) and LiOHH20 (172 mg, 4.1 mmol) and the solution was stirred at r.t. overnight. LCMS showed disappearance of staring material. The solution was adjusted to pH 4.0 using IN HC1 and poured into THF (30 mL), washed with Brine (20mL). The organic layer was dried over Na2S04, the organic layer was evaporated and submitted for Prep-HPLC. 45 mg product was collected (23 %). LCMS (ESI) calc'd for
C24Hi5ClF3N304 [M+H]+: 502, found: 502. 1HNMR (500 MHz, DMSO) 511.72 (lH,bs),9.09 (1H, s), 8.59(lH,d),8.23(lH, d), 8.06(3H,m), 7.88(2H, m),7.07(lH, s),7.49QH, d),2.15(3H,s).
TABLE 3
Figure imgf000076_0001
LCMS
Ex. Chemical Name A ring P Q [M+H]+
Found
3B 4-(l-(2-chloro-6- (trifluoromethyl)be
nzoyl)-lH- CI 0, -oH
pyrazolo[4,3- 539 b]pyridin-3-yl)-2- <¾ F3C
(methylsulfonamid
o)benzoic acid
Example 4A. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl) benzoyl)-6- (3- hydroxyazetidin-l-yl)-lH-indazol-3-yl)benzoic acid (4A)
SCH EME G
Figure imgf000077_0001
Step 1. Preparation of Methyl4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(3-hy- droxyazetidin-l-yl)-lH-indazol-3-yl)benzoate (G-1). To a mixture of methyl-4-(6- bromo- l-(2-chloro-6-(trifluoromethyl)benzoyl)-lH-indazol-3-yl)benzoate (i-9) (108 mg, 0.2 mmol), azetidin-3-ol (26mg, 0.24 mmol), Pd2(dba)3 (16 mg, 0.02 mmol), Xphos (20 mg, 0.04 mmol) and Cs2C03 (196 mg, 0.6 mmol) was added dioxane (10 ml), and the mixture was heated at 90°C under argon for 6 h. The mixture was cooled down, diluted with CH2C12 (100 ml). The organic layer was separated, washed with brine, dried over Na2S04, concentrated. The residue was purified by Prep-TLC (Pentane/EtOAc=10/l) to give 120 mg of the title compound (98%). LCMS (ESI) calc'd for C26H19CIF3N3O4 [M+H] +: 530.1 found: 530.
Step 2. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (3-hydroxyazetidin- l-yl)-lH-indazol-3-yl)benzoic acid (4A). To a stirred solution of Methyl 4-(l-(2-chloro-6- (trifluoromethyl)benzoyl)-6-(3 -hydroxyazetidin- 1 -yl)- 1 H- indazol-3 -yl)benzoate (G- 1) (159mg, 0.3 mmol) was added THF (8.0 ml), H20 (2.0 ml) and LiOHH20 (126 mg, 3 mmol) and the solution was stirred at r.t. overnight. LCMS showed disappearance of staring material. The solution was adjusted to pH 4.0 using IN HC1 and poured into THF (30 ml), washed with Brine (20mL). The organic layer was dried over Na2S04, the organic layer was evaporated and submitted for Prep-HPLC (ACN/H20). 55 mg product was collected (68 %). LCMS (ESI) calc'd for C25Hi7ClF3N304 [M+H] +: 516.09, found: 516; 1HNMR (500 MHz, DMSO) 513.24 (lH,s),8.05 (3H, d), 8.03 (2H, d), 7.87( 3H, t), 7.36QH, s),6.73 (lH,d), 5.72 (1H, bs), 4.67 (1H, m), 4.30 (2H, d), 3.76 (2H, d).
The following examples shown in TABLE 4 were prepared following similar procedures described for Examples 4A in Scheme G, which can be achieved by those of ordinary skill in the art of organic synthesis.
TABLE 4
Figure imgf000078_0001
LCMS
Example Chemical Name R [M+H]+
Found 4B 4-(6-(azetidin-l-yl)-l-(2- chloro-6-
(trifluoromethyl)benzoyl)- 518 1 H-indazol-3 -yl)-3 - fluorobenzoic acid
4D 4-(l-(2-chloro-6- (trifluoromethyl)benzoyl)- 6-(cyclopropylamino)- 518
Figure imgf000079_0001
1 H-indazol-3 -yl)-3 - fluorobenzoic acid
4F 4-(l-(2-chloro-6- (trifluoromethyl)benzoyl)- 6-(oxetan-3 -ylamino)- 1 H- 534 indazol-3-yl)-3- fluorobenzoic acid
4G 4-(l-(2-chloro-6- (trifluoromethyl)benzoyl)- 6-(3 -hydroxypyrrolidin- 1 - 548 yl)-lH-indazol-3-yl)-3- H 9O
fluorobenzoic acid
4H 4-(l-(2-chloro-6- (trifluoromethyl)benzoyl)-
530
6-morpholino- 1 H- indazol-3-yl)benzoic acid
Example 5A. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)bi (methoxycarbonylamino)-lH-indazol-3-yl)-3-fluorobenzoic acid (5A) Scheme H.
Figure imgf000080_0001
5A
Step 1. Preparation of 3-bromo-6-nitro-lH-indazole (H-2). A mixture of 6-nitro-lH- indazole (H-l) (5 g, 30 mmol) and NaOH (2 M, 20 ml) in 20 ml THF, Br2 (9.5 g, 60 mmol) dissolved in NaOH (2 M, 100 ml) was added, the mixture was stirred at RT for 1 night. The solvent was evaporated, The precipitated solid was filtered, washed with water (30 ml) and n- hexane (50 ml), dried to afford an off white solid H-2. LCMS (ESI): calc'd for C7H4BrN302 [M+H] +: 242 found: 242.
Step 2. Preparation of (3-bromo-6-nitro-lH-indazol-l-yl)(2-chloro-6-(trifluoro methyl)phenyl)methanone (H-3). To a 250 mL round-bottomed flask, was added 3-bromo- 6-nitro-lH-indazole (H-2) (9.4 g, 38.7 mmol), (2-chloro-6-(trifluoromethyl) benzoyl chloride) (10.3g, 42.6mmol), DMAP (472 mg, 3.87 mmol) and CH2C12 (lOOmL), after stirring at RT for 3 minutes, TEA (11.2 mL, 77 mmol) was added slowly. The reaction mixture is stirred at RT for overnight. LC-MS showed no starting materials remained. Then the mixture was poured to 30 mL water, the lower (organic) and upper (aqueous) phases are separated. The aqueous phase is extracted twice with 50 ml CH2C12. The combined organic phases are washed successively with two 20 ml portions of water and 10 ml of brine. The reaction resulting organic phase is dried over anhydrous sodium sulfate, filtered and concentrated at reduced pressure to give a yellow solid. The residue was purified by column chromatography (PE IE A from 50/1 to 10/1), to give a solid H-3. LCMS (ESI): calc'd for Ci5H6BrClF3N303 [M+H] +: 448, found: 448.
Step 3. Preparation of (6-amino-3-bromo-lH-indazol-l-yl)(2-chloro-6-(trifluoro methyl)phenyl)methanone (H-4). A mixture of (3-bromo-6-nitro-lH- indazol-l-yl) (2- chloro-6-(trifluoromethyl)phenyl)methanone (H-3) (lOg, 20 mmol) and SnCl2 (21 g, 10 mmol) in 100 ml EtOH , the mixture was stirred at 80 °C for 4 hours The solvent was evaporated with EtOAc(100 mix 3) and water (200 ml), The organic phase was collected and evaporated dried to afford an off white solid H-4. LCMS (ESI): calc'd for Ci5H8BrClF3N30, [M+H]+: 418 found:418.
Step 4. Preparation of methyl 4-(6-amino-l-(2-chloro-6-(trifluoromethyl)ben- zoyl)-lH- indazol-3-yl)-3-fluorobenzoate (H-5). A 30ml Microwave vial were charged with (6-amino- 3-bromo-lH-indazol-l-yl)(2-chloro-6-(trifluoromethyl) phen- yl)methanone (H-4) (2 g, 4.8 mmol), 2-fluoro-4-(methoxycarbonyl) phenylboronic acid (lg, 5.2 mmol), Pd(OAc)2 (54 mg, 0.24 mmol), Catacxium A (86 mg, 0.24 mmol) and KF (835 mg, 14.4 mmol) dissolved in unhydrous TUF (5 ml), A stir bar was added, the vial was sealed , the reaction was heated for 2 hours at a constant temperature of 80 °C, The mixture was filtered and the filtrate was collected , purified by column chromatography (DCM) to get the desired product H-5. LCMS (ESI): calc'd for C23Hi4ClF4N303 [M+H] +: 492 found: 492.
Step 5. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6 (methoxycarbonylamino)-lH-indazol-3-yl)-3-fluorobenzoate (H-6). Methyl 4-(6- amino- 1 -(2-chloro-6-(trifluoromethyl)benzoyl)- 1 H-indazol-3 -yl)-3 -fluorobenzoate (H-5)( 186 mg, 0.38mmol), methyl carbonochloridate (40 mg, 0.43mmol), DMAP (5 mg, 0.04 mmol) and CH2C12 (100 mL), after stirring at RT for 3 minutes, TEA (0.1 mL, 0.77 mmol) was added slowly. The reaction mixture is stirred at RT overnight. The solvent was concentrated at reduced pressure to give a yellow solid H-6. LCMS (ESI): calc'd for C25Hi6ClF4N305 [M+H] +: 550, found: 550. Step 6. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(methoxy- carbonylamino)-lH-indazol-3-yl)-3-fluorobenzoic acid (5A). A mixture of methyl 4-(l-(2- chloro-6-(trifluoromethyl)benzoyl)-6-(methoxycarbonylamino)-lH-indazol-3 -yl)-3- fluorobenzoate (H-6) (38 mg, 0.07 mmmol) and LiOH.H20 (16 mg, 0.37 mmmol) in 10 ml THF and 10 ml pure H20 was stirred at RT for 2 hours. The solvent was evaporated and the residue was dissolved in water. HC1 (5% sol in water) was added until pH =4-5. The precipitated solid was filtered, washed with water (10 ml) and n-hexane (10 ml), dried to afford an off white solid 5A. LCMS (ESI): calc'd for C^H^Cu^NsOs [M+H] +: 536 found: 536; IHNMR (400 MHz, DMSO) δ 13.54 (IH, s), 10.32 (IH, s), 8.93 (IH, s), 7.97-8.03 (2H, m), 7.83-7.91 (4H, m), 7.70-7.75 (IH, m), 7.61-7.63 (IH, d, J= 8 Hz), 3.76 (3H, s).
Example 5B. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(N- methylacetamido)-lH-indazol-3-yl)-3-fluorobenzoic acid (5B)
Scheme I
Figure imgf000082_0001
Figure imgf000082_0002
Step 1. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(me- thylamino)-lH-indazol-3-yl)-3-fluorobenzoate (1-1). A mixture of Methyl 4-(6-ami- no-1- (2-chloro-6-(trifluoromethyl)benzoyl)-lH-indazol-3-yl)-3-fluorobenzoate (H-5) (344 mg, 0.7 mmmol) in 10 ml DMF under ice bath, CH3I (0.07 ml, 0.14mmol) was added, and then the icebath was removed and stirred at RT for 5 hours. The solvent was extracted with EtOAc (30 ml) and water (3 χ 20 ml), the organic phase was collected and the residue purified by column chromatography of silic gel eluting with (PE/DCM=2: 1) to get the desired product 1-1. LCMS (ESI) calc'd for C24Hi6ClF4N303 [M+H] +: 506, found: 506.
Step 2. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(N-methyl- acetamido)-lH-indazol-3-yl)-3-fluorobenzoate (1-2). Methyl 4-(l-(2-chloro-6-(tri- fluoromethyl)benzoyl)-6-(methylamino)-lH-indazol-3-yl)-3-fluorobenzoate (1-1) (19 2 mg, 0.38mmol), acetyl chloride (33 mg, 0.43 mmol), DMAP (5 mg, 0.04 mmol) and CH2C12 (lOOmL), after stirring at RT for 3 minutes, TEA (0.1 ml, 0.77 mmol) was added slowly. The reaction mixture is stirred at RT for overnight. The solvent was concentrated at reduced pressure to give a yellow solid 1-3. LCMS (ESI): calc'd for C26Hi8ClF4N304 [M+H] +: 548, found: 548.
Step 3. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(N-methyl- acetamido)-lH-indazol-3-yl)-3-fluorobenzoic acid (5B). A mixture of 4-(l-(2- chloro-6- (trifluoromethyl)benzoyl)-6-(N-methylacetamido)-lH-indazol-3-yl)-3-fluorobenzoate (1-2) (38 mg, 0.07mmol) and LiOH.H20 (16mg, 0.37mmol) in 10 ml THF and 10 ml pure H20 was stirred at RT for 2 hours. The solvent was evaporated and the residue was dissolved in water. HC1 (5% in water) was added until pH = 4-5. The precipitated solid was filtered, washed with water (10 ml) and n-hexane (10 ml), dried to afford an off white solid 5B. LCMS (ESI): calc'd for C25Hi6ClF4N304[M+H] +: 534 found: 534; 1HNMR (400 MHz, DMSO) δ 8.46QH, s), 7.98-8.05(3H, m), 7.87-7.93 (3H, s), 7.76 (1H, s), 7.61-7.63QH, d, J= 8 Hz), 3.36 (3H, s), 2.01 (3H, s). The following examples shown in TABLE 5 were prepared following similar procedures described for Examples # 5A, 5B in Schemes H, I, which can be achieved by those of ordinary skill in the art of organic synthesis.
TABLE 5
Figure imgf000084_0001
LCMS
Example Chemical Name R [M+H]+
Found
5C 4-(l-(2-chloro-6- (trifluoromethyl)benzoyl)- 6-
546
(cyclopropanecarboxamid
o)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid
5D 4-(l-(2-chloro-6- (trifluoromethyl)benzoyl)- 0
6-(methylsulfonamido)- 556
0 H
1 H-indazol-3 -yl)-3 - fluorobenzoic acid
5E 4-(l-(2-chloro-6- 0
(trifluoromethyl)benzoyl)- HN X A 535
1 H
6-(3 -methylureido)- 1 H- indazol-3-yl)-3- fluorobenzoic acid
5F 4-(6-acetamido- 1 -(2- chloro-6-
(trifluoromethyl)benzoyl)- 520
H
1 H-indazol-3 -yl)-3 - fluorobenzoic acid
5G 4-(l-(2-chloro-6-
(trifluoromethyl)benzoyl)-
0
6-(N- I I
570
methylmethylsulfonamido 0 1
)-lH-indazol-3-yl)-3- fluorobenzoic acid
5H 4-(l-(2-chloro-6- (trifluoromethyl)benzoyl)-
6-( 1 , 3 -dimethylureido)- 549
1 H-indazol-3 -yl)-3 -
Figure imgf000085_0001
fluorobenzoic acid
Example 6A. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(2-oxo- imidazolidin-l-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (6A)
Scheme J
Figure imgf000085_0002
J-1 6A
i-9 Step 1. Preparation of methyl-4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(2- oxoimidazolidin-l-yl)-lH-indazol-3-yl)-3-fluorobenzoate (J-1). To a microwave tube was added methyl-4-(6-bromo-l-(2-chloro-6-(trifluoromethyl) benzoyl)- ΙΗ-ind- azol-3-yl)-3- fluorobenzoate (i-9) (70mg, 0.14 mmol), dioxane (1.5ml), imidazolidin -2-one (17mg,0.21mmol), Pd2(dba)3 (6.3mg, 0.007mmol), xant-phos (11.7 mg, 0.021 mmol), Cs2C03(86 mg,0.28 mmol). The solution was microwaved at 100°C for 2 hours and the organic layer was evaporated to use in next step without purificatiom. LCMS (ESI) calc'd for C26Hi7ClF4N404 [M+H] +: 561, found: 561.
Step 2. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(2-oxoimi- dazolidin-l-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (6A). To a stirred solution of methyl 4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(2-oxoimidazolidin- 1 -yl)- 1 H- indazol-3 -yl)-3 - fluorobenzoate (J-1) in dioxane(1.5ml) from last step was added LiOHH20 (59mg, 1.4mmol) and H20 (0.5 ml). The solution was stirred overnight and LCMS showed major product peak. The solution was adjusted to pH=3 using 1 N HCl. The upper organic layer was collected and the aqueous layer was extracted with THF (2xlmL). The combined organic layer was added 0.5mL MeOH and submitted for Prep-HPLC (H20/ACN) gave lOmg product, yield for two steps 26%. LCMS (ESI) calc'd for C25H15C1F4N404 [M+H]+: 547, found: 547; 1HNMR (400 MHz, DMSO) δ 13.44 (1H, s), 8.75 (1H, s), 8.01 (2H,m), 7.98-7.89 (5H, m), 7.71 (lH,d), 4.08-4.05(2H, d), 3.52-3.33 (2H, d).
Example 6B. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (2- oxoazetidin-l-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (6B)
Scheme K
Figure imgf000087_0001
Step 1. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(2- oxoazetidin-l-yl)-lH-indazol-3-yl)-3-fluorobenzoate (K-1). The mixture of methyl 4-(6- bromo- 1 -(2-chloro-6-(trifluoromethyl)benzoyl)- 1 H-indazol-3 -yl)-3 -fluorobenzo- ate (i-9) (554 mg, 1.0 mmol), Pd(dppf)2Cl2 (79 mg,0.1 mmol), azetidin-2-one (114 mg, 2.0 mmol), TEA (20 ml) and DMF (20 ml) was purged with N2 and stirred at 90°C overnight. LCMS showed the SM was consumed totally and the expected product appeared. The resulting solution was filtered and concentrated to purify by column chromatograph (PE/EA) to give 248 mg product as a white solid (45%). LCMS (ESI) calc'd for C26Hi6ClF4N304 [M+H] +: 545.87, found: 546.2.
Step 2. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(2-oxoazeti- din-1- yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (6B). To the solution of methyl 4-(l-(2-chloro-6- (trifluoromethyl)benzoyl)-6-(2-oxoazetidin- 1 -yl)- 1 H-indazol-3 -yl)- 3 -fluorobenzoate (K-1) (53.1 mg, 0 . lmmol) in THF (30 ml) and water(10 ml) was added LiOH (240 mg, 10 mmol). The mixture solution was stirred at 0°C for 2 h. Added 100 ml HCl aqueous, exacted with EA (30mlx3), the organic layer was concentrated to be purified by chromatography column (EA:PE=1 : 1) to afford 27 mg product (51%). LCMS (ESI) calc'd for C25Hi4ClF4N304 [M+H]+: 531.84, found: 532.1; 1HNMR (400 MHz, CDC13) 5: 13.5(lH,w), 8.46(lH,s),7.98(6H,m), 7.72(lH,t), 7.61(lH,d), 3.85(2H, m), 3.21(2H, m).
Example 6C. Preparation of 4-(6-(2-carboxyethylamino)-l-(2-chloro-6- (trifluoromethyl)benzoyl)-lH-indazol-3-yl)-3-fluorobenzoic acid (6C) Scheme L
Figure imgf000088_0001
Step 1. Preparation of 4-(6-(2-carboxyethylamino)-l-(2-chloro-6-(trifluoromethy- l)benzoyl)-lH-indazol-3-yl)-3-fluorobenzoic acid (6C). To the solution of methyl 4-(l-(2- chloro-6-(trifluoromethyl)benzoyl)-6-(2-oxoazetidin- 1 -yl)- 1 H-indazol-3 -yl)-3 -fluorobenzoate (K-l) (53.1 mg, O. lmmol) in THF (30 ml) and waterQO ml) was added LiOH (240 mg, 10 mmol). The mixture solution was stirred at rt for 12 h. Added 100 ml HCl aqueous, exacted with EA (30ml x 3), the organic layer was concentrated to be purified by chromatography column (EA:PE=1 : 1) to affor 39 mg product (yield:72%). LCMS (ESI) calc'd for C24H14CIF4N3O5 [M+H]+: 549.86, found: 550.1. 1HNMR (400 MHz, CDC13) 5: 13.1(2H,w), 7.9(5H,m), 7.66(lH,t), 7.541(2H,m),6.91(lH,m), 3.41(2H, m), 2.62(2H, m).
The following examples shown in TABLE 6 were prepared following similar procedures described for Examples #6A, 6B, 6C in Schemes J, K, L, which can be achieved by those of ordinary skill in the art of organic synthesis.
TABLE 6
Figure imgf000089_0001
Figure imgf000089_0002
Example 7A. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(mor pholine- 4-carbonyl)-lH-indazol-3-yl)benzoic acid (7 A)
Scheme M
Figure imgf000090_0001
7A
Step 1. Preparation of 5-bromo-2-methylpyridin-3-amine (M-2). To a solution of 5- bromo-2-methyl-3-nitropyridine (1) (15 g, 69.4 mmol) in EtOH (300 mL) and water (70 ml), was added iron powder (46.7 g, 833 mmol) and ammonium chloride (4.5 g, 83.4 mmol) successively at rt. The reaction mixture was heated to 90 °C for 40 min. The reaction was filtered hot and rinsed with EtOAc. The filtrate was washed with a saturated aqueous solution of sodium bicarbonate (200 mL), brine, dried over magnesium sulfate and solvent removed in vacuo to give the title compound as an orange solid, (11.7 g, 62.9 mmol, 90%). LCMS (ESI) calc'd for C6H7BrN2 [M+H]+: 187, found: 187, 188. Step 2. Preparation of N-(5-bromo-2-methylpyridin-3-yl)acetamide (M-3). To a solution of 5-bromo-2-methylpyridin-3-amine (M-2) (10.7 g, 57.5 mmol) in dichloromethane (575 mL) was added acetic anhydride (12 mL, 126.5 mmol) at 0 °C, followed by triethylamine (22 mL, 158 mmol). The mixture was allowed to warm to ambient temperature and stirred for 18 hours at which point a further equivalent of acetic anhydride (6 mL, 63 mmol) was added. The mixture was stirred at ambient temperature for a further 18 hours. The reaction mixture was quenched with a saturated aqueous solution of sodium bicarbonate (500 mL) and the organic phase washed with saturated aqueous sodium chloride (500 mL), dried over magnesium sulfate and concentrated in vacuo to give a brown solid. This solid was triturated with 30% ethyl acetate in hexanes to yield the title compound as an off-white solid, (8.28 g, 36 mmol, 63%). 1HNMR (400 MHz, CD3OD): δ ppm 8.31 (s, 1H), 8.18 (s, 1H), 2.43 (s, 3H), 2.18 (s, 3H). LCMS (ESI) calc'd for C8H9BrN20 [M+H]+: 228.99, found: 229, 230.
Step 3. Preparation of 6-bromo-lH-pyrazolo[4,3-b]pyridine (M-4). To a solution of N-(5- bromo-2-methylpyridin-3-yl)acetamide (M-3) (8.3 g, 36 mmol) in chloroform (550 mL) at ambient temperature was added potassium acetate (4.3 g, 43.6 mmol), acetic acid (2.5 mL, 43.6 mmol) and followed by acetic anhydride (6.9 mL, 72.6 mmol). The mixture was stirred at ambient temperature for 15 minutes before being heated to 40 °C. Jert-butyl nitrite (6.5 mL, 54 mmol) was then added dropwise. The reaction was then stirred at 60 °C for 48 hours. The reaction mixture was poured slowly into a saturated solution of sodium bicarbonate (500 mL) at 0 °C. The organic phase was retained and the aqueous phase extracted with dichloromethane (500 mL). The combined organics were then concentrated to a brown oil which was dissolved in methanol (500 mL). Aqueous sodium hydroxide (2 M, 500 mL) was added at 0 °C and the mixture stirred at ambient temperature for 1 hour before the methanol was removed in vacuo. The aqueous mixture was then extracted with ethyl acetate (3 χ 500 mL). The combined organics dried over magnesium sulfate, and the solvent removed in vacuo to give the title compound as a light brown solid (5.5 g, 27.9 mmol, 77%). 1HNMR (400, CD3OD): δ ppm 8.55 (s, 1H), 8.24 (s, 1H), 8.21 (s, 1H). LCMS (ESI) calc'd for C6H4BrN3 [M+H]+: 197.96, found: 198, 199.
Step 4. Preparation of methyl lH-pyrazolo[4,3-b]pyridine-6-carboxylate (M-5). To a solution of 6-bromo-lH-pyrazolo[4,3-b]pyridine (M-4) ( 0.5 g, 2.5 mmol) in methanol ( 15 ml) and acetonitrile ( 7 ml) was added Et3N ( 2.2 ml, 5.6 mmol ), Binap ( 0.17 g, 0.63 mmol) and palladium dichloride ( 0.17 g, 0.27 mmol). The mixture was placed under 20 bar of carbon monoxide, stirred at 100 °C for 18 h. The mixture was cooled, filtered and purified by Pre-TLC to give 310 mg white solid. ( 69 % ). LCMS (ESI) calc'd for [M+H]+: 178.1, found: 178.1.
Step 5. Preparation of methyl 3-iodo-lH-pyrazolo[4,3-b]pyridine-6-carboxyl- ate (M-6).
To a solution of methyl lH-pyrazolo[4,3-b]pyridine- 6-carboxylate (M-5) ( 316 mg, 1.8 mmol ) in DMAC ( 30 ml ) was added KOH ( 40 mg, 7.18 mmol ). The vigorously stirred mixture was treated with iodine (550 mg, 2.15 mmol) and added portionwise over 5 minutes then stirred for 60 minutes. The reaction was quenched with 20 ml of 20% citric acid solution, followed by 16 ml of saturated NaHS03 solution, then adjusted to pH = 8 with solid NaHC03 and partitioned between ethyl acetate and water. The organic extract was dried and concentrated to a dark-red oil contain DMAC for next step directly. LCMS (ESI) calc'd for [M+H]+: 304.1, found: 304.1.
Step 6. Preparation of methyl l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-iodo- 1H- pyrazolo[4,3-b]pyridine-6-carboxylate (M-7). To the solution of methyl 3-iodo -1H- pyrazolo[4,3-b]pyridine-6-carboxylate (M-6) (400 mg, 1.32 mmol), Et3N (290 mg) and DMAP (32 mg, 0.26 mmol) dissolved in anhydrous DCM (10 ml) was added 2-chloro-6- (trifluoromethyl)benzoyl chloride (630 mg, 2.64 mmol) in anhydrous DCM (10 ml) drop wise. The mixture solution was protected by N2 and stirred at rt for 20 h. Then the solution was concentrated to to afford 500 mg product ( 78 % ). LCMS (ESI) calc'd [M+H]+: 510.1, found: 510.1.
Step 7. Preparation of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-iodo- lH-pyraz- olo[4,3-b]pyridine-6-carboxylic acid (M-8). To the solution of methyl l-(2-chloro- - (trifluoromethyl)benzoyl)-3-iodo-lH-pyrazolo[4,3-b]pyridine-6-carboxylate M-7) (700 mg, 1.37 mmol) in THF (15 mL ) and H20 (5 mL) was added LiOH (242 mg, 10.9 mmol). The mixture solution was stirred at RT or 24 h. Added water (10 ml), acidified by HC1 ( 2 M ), extracted with EtOAc (20 ml χ 3), combined the organic layer, dried, filtered and concentrated, purified by Pre-HPLC to afford 670 mg (99%). LCMS (ESI) calc'd [M+H] : 496.1, found: 496.1.
Step 8. Preparation of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-(2-fluoro- 4- (methoxycarbonyl)phenyl)-lH-pyrazolo[4,3-b]pyridine-6-carboxylic acid (M-9). The mixture of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-iodo-lH- pyrazolo[4,3-b]
pyridine-6-carboxylic acid (M-8) (100 mg, 0.20 mmol), 2-fluoro-4-(methoxycarbonyl phenylboronic acid (100 mg, 0.5 mmol), Pd(dppf)Cl2 (44 mg, 0.06 mmol ), Na2C03 (50 mg, 0.6 mmol ) in dioxane (4 ml) and H20 (0.5 mL) was stirred at 100°C in MW for 1 h. Then the solution was filtered and concentrated to be purified by Pre-HPLC to afford 60 mg product (58 %). LCMS (ESI) calc'd [M+H]+: 522.0, found: 522.0.
Step 9. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (morpholine-4-carbonyl)-lH-indazol-3-yl)benzoate (M-10).l-(2-Chloro-6-(trifluor- omethyl)benzoyl)-3 -(2-fluoro-4-(methoxycarbonyl)phenyl)- 1 H-pyrazolo [4,3 -b]pyrid- ine-6- carboxylic acid (M-9) (180 mg, 0.36 mmol) was dissolved in CH2C12 (15 mL). morpholine (37 mg,0.43 mmol), PYAOP (374 mg, 0.72 mmol) was added and the mixture was stirred at room temperature for 2 min. TEA (0.16 mL, 1.08 mmol) was added and the mixture was stirred at room temperature for 2 h. Then it was diluted with EtOAc (20 ml), washed with brine (20 ml x 2), dried with anhydrous Na2S04, concentrated to get white solid 195 mg (95%). LCMS (ESI): calc'd for C28H2i C1F3N305, [M+H]+: 572, found: 572.
Step 10. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(morphol- ine-4- carbonyl)-lH-indazol-3-yl)benzoic acid (7A). A mixture of methyl 4-(l-(2-ch- loro-6- (trifluoromethyl)benzoyl)-6-(morpholine-4-carbonyl)- 1 H-indazol-3 -yl)benzoa- te (M- 10) (195 mg, 0.34 mmol) and LiOH (72 mg, 1.7 mmol) in 10 ml THF and 10 ml H20 was stirred at rt for 2 hour. The solvent was evaporated and the residue was dissolved in water. HC1 (5% sol. in water) was added until pH 4-5. The precipitated solid was filtered, washed with water and n-hexane, dried to afford an off-white solid 184 mg (97%). LCMS (ESI): calc'd for C27Hi9ClF3N305, [M+H]+: 558.1 found: 558.1. 1HNMR (400MHz, DMSO) δ 8.55 (1H, s), 8.32-8.34 (1H, d, J = 8 Hz), 8.05-8.11 (3H, m), 8.01-8.03 (1H, d, J = 8 Hz), 7.95-7.97 (2H, d, J = 8Hz), 7.87-7.91 (1H, m), 7.69-7.71 (1H, d, J = 8 Hz), 3.52-3.72 (8H, m). Example 7B: Preparation of 3-(4-carboxyphenyl)-l-(2-chloro-6-(trifluoromethyl) benzoyl)-lH-indazole-6-carboxylic acid (7B)
Scheme N
Figure imgf000094_0001
Step 1. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(met- hoxy(methyl)carbamoyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoate (N-2). l-(2-
Chloro-6-(trifluoromethyl)benzoyl)-3-(2-fluoro-4-(methoxycarbonyl)phenyl) -1H- pyrazolo[4,3-b]pyridine-6-carboxylic acid (M-9) (100 mg, 0.19 mmol) was dissolved in CH2C12 (10 mL), Ο,Ν-dimethylhydroxylamine (22 mg, 0.23 mmol), HATU (94 mg, 0.25 mmol) was added and the mixture was stirred at room temperature for 2 min. TEA (23 mg, 0.23 mmol) was added and the mixture was stirred at room temperature overnight. Then it was diluted with EtOAc (15 ml), washed with brine (15 ml><2), dried over anhydrous Na2S04, concentrated to got crude solid N-2 (80 mg). LCMS (ESI): calc'd for C25Hi7ClF4N405, [M+H]+: 564.7, found: 564.7.
Step 2. Preparation of 3-(4-carboxy-2-fluorophenyl)-l-(2-chloro-6-(trifluoromet hyl)benzoyl)-lH-pyrazolo[4,3-b]pyridine-6-carboxylic acid (7B). A mixture of methyl 4- (l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(methoxy(methyl)carbamoyl)-l H-pyrazolo[4,3- b]pyridin-3-yl)-3-fluorobenzoate (N-1) (80 mg, 0.14 mmol) and LiOH»H20 (30 mg, 0.7 mmol) in 5 ml THF and 5 ml H20 was stirred at room temperature for 2 hours. The solvent was evaporated and the residue was dissolved in water. HC1 (5% sol. in water) was added until pH 4-5. The product was extracted by EtOAc, concentrated to obtain crude solid. The product was purified by pre-HPLC to get 7B (10 mg), yield 14.1%. Physical characterization data for 7B is as follows: LCMS (ESI): calc'd for C22HioClF4N305, [M+H]+: 508, found: 508. 1HNMR (400 MHz, MeOD) δ 9.43-9.47 (2H, d, J=13.6Hz), 8.35-8.38 (1H, m), 7.99-8.02 (1H, d, J = 8 Hz), 7.89-7.93 (2H, m), 7.80-7.88 (2H, m).
TABLE 7
Figure imgf000095_0001
Example 8A: Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (cyclo
Propyl (hydroxy)methyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoic acid (8A)
SCHEME O
Figure imgf000096_0001
p 1. Preparation of methyl 3-bromo-lH-pyrazolo[4,3-b]pyridine -6-carboxylat e (0-2). To a solution of methyl lH-pyrazolo[4,3-b]pyridine -6-carboxylate (O-l) (2 g, 11.3 mmol) in THF (50 mL) was added BS (3 g, 16.9 mmoL), the reaction mixture was stirred overnight at rt, then methanol was added to quench the reaction, concentrated to give a crude product, triturated with EtOAc, filtered and collected the solid to give 2.5 g (87%) of title compound as a white solid. LCMS (ESI): calc'd for C8H6BrN302 [M+H]+: 256, found: 256.
Step 2. Preparation of methyl 3-bromo-l-(2-chloro-6-(trifluoromethyl)benzoyl) -1H- pyrazolo[4,3-b]pyridine-6-carboxylate (0-3). To a solution of methyl 3-bromo -lH-pyrazolo[4,3-b]pyridine-6-carboxylate (0-2) (2.5 g, 9.8 mmol) in DCM (100 mL) was added TEA (2 mL, 14.7 mmol), DMAP (240 mg, 2 mmol), then 2-chloro-6- (trifluoromethyl)benzoyl chloride (3.1 g, 12.7 mmol) in DCM (10 mL) was added dropwise, then the reaction was stirred for 3 h at rt, then diluted with EtOAc (200 mL), washed with sat. NaHC03 aqueous, water and brine, dried over Na2S04, concentrated, purified by flash chromatograph (PE:EA=10: 1) to give 4.3 g (95%) of title compound as a light yellow solid. LCMS (ESI): calc'd for Ci6H8BrClF3N303 [M+H]+: 462, found: 462.
Step 3. Preparation of (3-bromo-6-(hydroxymethyl)-lH-pyrazolo[4,3-b] pyridin- l-yl)(2-chloro-6-(trifluoromethyl)phenyl)methanone (0-4). To a solution of methyl 3- bromo- 1 -(2-chloro-6-(trifluoromethyl)benzoyl)- 1 H-pyrazolo [4, 3 -b] pyridine
-6-carboxylate (0-3) (2 g, 4.3 mmol) in dried THF (50 mL) was added DIBAL (1M in THF, 13 mL) slowly at -40 °C, then the reaction mixture was warmed to rt and stirred for 3 h, the reaction mixture was cooled to 0 °C, 15% NaOH aqueous (5 ml), water (5 mL) was added slowly successively, and stirred for another 30 min, filtered and concentrated, purified with flash chromatograph (PE:EA=3 : 1) to give 1.4 g (75%) of title compound as a light yellow solid. LCMS (ESI): calc'd for Ci6H8BrClF3N303 [M+H]+: 434, found: 434.
Step 4. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (hyd roxymethyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoate (0-5). A mixture of (3- bromo-6-(hydroxymethyl)-l H-pyrazolo [4,3-b]pyridin-l-yl) (2-chloro-6- (trifluo
romethyl)phenyl)methanone (0-4) (1.3 g, 3.0 mmol), 2-fluoro-4- (methoxycarbonyl) phenylboronic acid (1.2 g, 6.0 mmol), Pd(dppf)Cl2 (367 mg, 0.45 mmol), and K3P04 (1.9 g, 9.0 mmol) in 8 mL dioxane and water (7: 1) was heated at 10 °C for 1 h with oil-bath. Then it was filtrated and washed with EtOAc, the organic phase was concentrated and the product was purified with column chromatography (PE:EA=3 : 1) to get yellow solid 0-5 (800 mg), yield 52.6%. Physical characterization data for 0-5 is as follows: LCMS (ESI): calc. C23Hi4ClF4N304, 507.7; found: M+H=508.7.
Step 5. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- formyl-lH- pyrazolo [4,3-b] pyridin-3-yl)-3-fluorobenzoate (0-6). 4-( 1 -(2-Chloro-6-
(trifluoromethyl)benzoyl)-6-(hydroxymethyl)- 1 H-pyrazolo [4,3 -b]pyridine-3 -yl)- 3 - fluorobenzoate (0-5) (670 mg, 1.32 mmol) was dissolved in DCM (15 mL), Dess-Martin reagent (840 mg, 1.98 mmol) was added, then the mixture was stirred at room temperature for 2 h. The solvent was evaporated and the product was purified with column chromatography (PE:EA=3 : 1) to get solid 0-6 (580 mg), yield 87%. Physical characterization data for 0-6 is as follows: LCMS (ESI): calc. C23Hi2ClF4N304, 505.7; found: M+H=506.7.
Step 6. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (cyclopropyl(hydroxy)methyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoate (0-7).
A 50 mL round-bottomed flask, is desgassed with nitrogen, cooled to -78 °C, cyclopropylmagnesium bromide (4 mL, 2.0 mmol) was added, then methyl 4-(l-(2-chloro-6- (trifluoromethyl)benzoyl)-6-formyl- 1 H-pyrazolo [4,3 -b]pyridine-3 -yl)-3 -fluorobenzoate (0-6) (80 mg, 0.16 mmol) in THF (3 mL, anhydrous) was added slowly. The mixture was stirred from -78 °C to rt for 4 h. Water was added, and the solvent was evaporated. The product was purified with Pre-HPLC to get white solid 0-7 (26 mg), yield 29.7%. Physical characterization data for 0-7 is as follows: LCMS (ESI): calc. C26Hi8ClF N304, 547.7; found: M+H=548.7.
Step 7. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(cyclopropyl (hydroxy)methyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoic acid (8A). A mixture of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(cyclopropyl (hydroxy)methyl)-lH- pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoate (0-7) (26 mg, 0.05 mmol) and LiOH»H20 (10 mg, 0.24 mmol) in 2 ml THF and 2 ml H20 was stirred at room temperature for 2 hours. The solvent was evaporated and the residue was dissolved in water. HC1 (5% sol in water) was added until pH 4-5. The product was extracted by EtOAc, concentrated to obtain crude solid. The product was purified by pre-HPLC to get 0-8 (4 mg), yield 15.79%). Physical characterization data for 8A is as follows: LCMS (ESI): calc. C25Hi6ClF N304, 533.7; found: M+H=533.7. 1HNMR (400 MHz, MeOD) δ 8.97-9.00 (2H, d, J=12 Hz), 8.26-8.30 (1H, m), 7.98-8.00 (1H, d, J = 8Hz), 7.84-7.90 (3H, m), 7.77-7.81 (1H, m), 4.37-4.39 (1H, d, J = 8.4Hz), 1.29-1.33 (1H, m), 0.74-0.78 (1H, m), 0.67-0.72 (2H, m), 0.57-0.63 (1H, m).
Example 8B: Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(cyclop ropane-carbonyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoic acid (8B) SCHEME P
Figure imgf000099_0001
Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-
(cyclopropanecarbonyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoate (P-2). Methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(cyclopropyl(hydroxy)methyl) -1H- pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoate (P-1) (25 mg, 0.05 mmol) was dissolved in DCM (5 mL), Dess-Martin reagent (29 mg, 0.069 mmol) was added, then the mixture was stirred at room temperature for 3 h. The solvent was evaporated and the product was purified with column chromatography (PE:EA=4: 1) to get crude solid P-2 (10 mg). Physical characterization data for P-2 is as follows: LCMS (ESI): calc. C26H16CIF4N3O4, 545.7; found: M+H=546.7.
Step 2. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (cyclopropan ecarbonyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoic acid (8B). A mixture of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(cyclopro panecarbonyl)-lH- pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoate (P-2) (10 mg, 0.02 mmol) and LiOH»H20 (8 mg, 0.18 mmol) in 2 mL THF and 2 mL H20 was stirred at room temperature for 2 hours. The solvent was evaporated and the residue was dissolved in water. HC1 (5% sol. in water) was added until pH 4-5. The product was extracted by EtOAc, concentrated to obtain crude solid. The product was purified by Pre-HPLC to get 3 (2 mg), yield 2.35%. Physical characterization data for 8B is as follows: LCMS (ESI): calc. C25Hi4ClF4N304, 531.7; found: M+H=531.7. 1HNMR (400 MHz, DMSO) δ 9.553 (1H, s), 9.312 (1H, s), 8.377 (1H, s), 8.04- 8.08 (1H, m), 7.96-8.01 (1H, m), 7.88-7.92 (1H, m), 7.01-7.84 (1H, d, J=10.4Hz), 3.16-3.18 (1H, m), 1.15-1.22 (4H, m).
TABLE 8:
Figure imgf000100_0001
acid
Example 9A: Preparation of sodium 4-(l-(2-chloro-6-(trifluoromethyl)bi -6- 5-methyloxazol-2-yl)-lH-indazol-3-yl)benzoate (9 A)
Figure imgf000101_0001
Qio 9A Step 1:
Preparation of methyl lH-indazole-6-carboxylate (Q-2). Methyl 3-amino-4 -methylbenzoate (Q-l) (5.0 g, 30.2 mmol) was dissolved in AcOH (140 mL). Sodium nitrite (2.1 g, 30.2 mmol) in water (3.5 mL) was added drop-wise to the solution of starting material under ice-cooling at room temperature. The ice-bath was removed and the mixture was stirred overnight. Half of the solvents were evaporated, the mixture was diluted with water (80 mL) and extracted with EtOAc (30 mLx3). The collected organic phase was washed with water and brine (200 mL><2), dried and evaporated to afford the crude product Q-2 (4.4 g). LCMS (ESI): calc'd for C9H8N202, [M+H]+: 177.1, found: 177.1. Step 2: Preparation of methyl 3-iodo-lH-indazole-6-carboxylate (Q-3). Methyl 1H- indazole-6-carboxylate (Q-2) (5.0 g, 28.3 mmol) was dissolved in anhydrous DMAC (50 mL). Iodine (14.4 g, 56.7 mmol) and potassium hydroxide (6.3 g, 113.5 mmol) were added in portions under ice-cooling at room temperature. The ice-bath was removed and the mixture was stirred at room temperature for 1 h. The reaction was monitored by TLC (25% MeOH in chloroform) then it was slowly quenched with Na2S203 (sat. sol. in water, 100 mL), diluted with water (50 mL) and extracted with EtOAC (100 mLx3). The organic phase was evaporated and triturated with n-hexane. The precipitated material was filtered and dried to afford a brown solid Q-3 5.3 g (62%). LCMS (ESI): calc'd for C9H7IN202, [M+H]+: 302.9, found: 302.9.
Step 3: Preparation of methyl l-(2-chloro-6-(trifluoromethyl)benzoyl) -3-iodo-lH
-indazole-6-carboxylate (Q-5). To a 250 mL round-bottomed flask, was added compound methyl 3-iodo-lH-indazole-6-carboxylate (Q-3) (11.7 g, 38.7 mmol), 2-chloro-6- (trifluoromethyl)benzoyl chloride (Q-4) (10.3 g, 42.6 mmol), DMAP (4.72 g, 38.7 mmol) and CH2C12 (100 mL), after stirring at rt for 3 minutes, TEA (11.2 mL, 77 mmol) was added slowly. The reaction mixture is stirred at rt for overnight. LCMS showed no starting materials remained. Then the mixture was poured to 30 mL water, the lower (organic) and upper (aqueous) phases were separated. The aqueous phase is extracted twice with 20 mL CH2C12. The combined organic phases are washed successively with two 20 mL portions of water and 10 mL of brine. The resulting organic phase was dried over anhydrous sodium sulfate, filtered and concentrated at reduced pressure to give a yellow solid. The residue was purified by column chromatography on 60 g of silica gel eluting with PE /EA from 50/1 to 10/1, to give a fawn solid Q-5 16.5 g (84%). LCMS (ESI): calc'd for Ci7H9ClF3IN203, [M+H]+: 508.9, found: 508.9.
Step 4: Preparation of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-iodo- 1H-Indaz ole-6-carboxylic acid (Q-6). A mixture of l-(2-chloro-6-(trifluoromethyl) benzoyl)-3
-iodo-lH-indazole-6-carboxylate (Q-5) (16.5 g, 32.48 mmol) and LiOH (3.40 g, 162.40 mmol) in 10 mL THF and 50 mL pure H20 was stirred at rt overnight. The solvent was evaporated and the residue was dissolved in water. HC1 (5% sol. in water) was added until pH 4-5. The precipitated solid was filtered, washed with water and n-hexane, dried to afford an off white solid Q-6 16.0 g (83%). LCMS (ESI): calc'd for C16H7CIF3IN2O3, [M+H]+: 494.9, found: 494.9.
Step 5: Preparation of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-(4- (methoxycar bonyl)phenyl)-lH-indazole-6-carboxylic acid (Q-7). A mixture of l-(2-chloro-6- (trifluoromethyl)benzoyl)-3-iodo-lH-indazole-6-carboxylic acid (Q-6) (300 mg, 0.61 mmol), 4-(methoxycarbonyl) phenylboronic acid (165 mg, 0.92 mmol), Pd(dppf)Cl2 (50 mg, 0.061 mmol) and KOAc (181 mg, 1.83 mmol) in 10 mL dioxane and 2 mL pure H20 was heated to 95 C for 2 h with microwave. Then it was diluted with EA (50 mL), washed with brine (50 mLx2), dried over anhydrous Na2S04, concentrated. Then it was purified by silica gel column (Petroleum ether /EtOAc =20/1) to get white solid Q-7 180 mg (59%). LCMS (ESI): calc'd for C24Hi4ClF3N205, [M+H]+: 503.1, found: 503.1.
Step 6: Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) -6-(2- oxopropylcarbamoyl)-lH-indazol-3-yl)benzoate (Q-8). l-(2-Chloro-6- (trifluoro methyl)benzoyl)-3-(4-(methoxycarbonyl)phenyl)-lH-indazole-6-carboxylic acid (Q-7) (180 mg, 0.36 mmol) was dissolved in CH2C12 (15 mL). l-aminopropan-2-one hydrochloride (47 mg, 0.43 mmol), PYAOP (374 mg, 0.72 mmol) was added and the mixture was stirred at room temperature for 2 mins. TEA (0.16 mL, 1.08 mmol) was added and the mixture was stirred at room temperature for 2 h. Then it was diluted with EtOAC (20 ml), washed with brine (20 ml><2), dried over anhydrous Na2S04, concentrated to get crude product Q-8 (191 mg), which was used to the next step without further purification. LCMS (ESI): calc'd for C27Hi9ClF3N305, [M+H]+: 558.1, found: 558.1.
Step 7: Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) -6-(5- methyloxazol-2-yl)-lH-indazol-3-yl)benzoate (Q-9). POCl3 (3.5 mL) was added to a solution of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) -6-(2-oxopropylcarba moyl)-lH-indazol-3-yl)benzoate (Q-8) (191 mg, 0.34 mmol) in pyridine (7 mL) at 25 °C. The resulted solution was then warmed to 70 °C and stirred for 6 hours. Upon completion, the reaction mixture was cooled to 25 °C, diluted with EtOAc (10 mL), poured into a cold (0 °C) solution of saturated aqueous NaHC03 (50 mL), and extracted with EtOAc ( 25 mLx3). The combined organic layers were then washed with water (50 mL) and brine (50 mL), dried (MgS04), and concentrated to get a crude product Q-9 (40 mg). LCMS (ESI): calc'd for C27Hi7ClF3N304, [M+H]+: 540.1, found: 540.1.
Step 8: Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) -6-(5-methylox azol-2-yl)-lH-indazol-3-yl)benzoic acid (Q-10). A mixture of methyl 4-(l-(2-chloro-6- (trifluoromethyl)benzoyl)-6-(5-methyloxazol-2-yl)-lH-indazol-3-yl)benzoate (Q-9) (40 mg, 0.07 mmol) and LiOH (16 mg, 0.37 mmol) in 10 mL THF and 10 mL pure H20 was stirred at RT for 2 hours. The solvent was evaporated and the residue was dissolved in water. HC1 (5% sol in water) was added until pH 4-5. The precipitated solid was filtered, washed with water and n-hexane, dried to afford an off white solid Q-10 36 mg (98%). LCMS (ESI): calc'd for C26Hi5ClF3N304, [M+H]+: 526 found: 526.
Step 9: Preparation of sodium 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) -6-(5- methyloxazol-2-yl)-lH-indazol-3-yl)benzoate (9A). 4-(l-(2-Chloro-6- (trifluoro
methyl)benzoyl)-6-(5-methyloxazol-2-yl)-lH-indazol-3-yl) benzoic acid (Q-10) (36 mg, 0.069 mmol) was added to pure H20 (10 mL) to disperse equably with ultrasonic. Then 0.1 mol/L NaOH (0.7 mL, 0.07 mmol) was added to the solution at ice-bath. Then it was stirred at 0 °C for 30 mins. Then it was dried by Freeze dryer to get compound 9A 38 mg (100%). LCMS (ESI): calc'd for C26Hi4ClF3N3Na04, [M+H]+: 548, found: 548. 1HNMR (400 MHz, DMSO) δ 9.04 (1H, s), 8.37-8.39 (1H, d, J = 8 Hz), 8.18-8.20 (1H, d, J = 8 Hz), 8.06-8.08 (1H, d, J = 8 Hz), 8.00-8.02 (3H, d, J = 8.8 Hz), 7.87-7.91 (1H, m), 7.78-7.80 (2H, d, J = 8 Hz), 7.16 (lH,s),2.48 (3H, s).
TABLE 9:
Figure imgf000104_0001
Chemical LCMS
A ring P Q
Ex. Name [M+H]+
Figure imgf000105_0001
acid
Example 11 A: Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(3-hy droxyprop-l-ynyl)-lH-indazol-3-yl)benzoic acid (11 A)
SCHEME S
Figure imgf000105_0002
Step 1. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) (tetrahydro-2H-pyran-2-yloxy)prop-l-ynyl)-lH-indazol-3-yl)benzoate (S-2). solution of methyl 4-(6-bromo-l-(2-chloro-6-(trifluoromethyl) benzoyl)- IH-ind azol-3-yl)benzoate (S-l) (53.5 mg, 0.1 mmol) in DMF (5 mL) and TEA (10 mL) was added 2-(prop-2-ynyloxy)-tetrahydro-2H-pyran (16.8 mg, 0.12 mmol), copper(I) iodide (10 mg, 0.01 mmol) and PdCl2(PPh3)2 (10 mg, 0.01 mmol) under Argon . The mixture was stirred under Argon for 2 hours at 80 °C. The mixture was diluted with H20. The aqueous layer was extracted with CH2C12 (3 x50 mL). The combined organics were washed with H20, brine, dried over Na2S04, and concentrated. The residue was purified by Prep-TLC (Pentane/EtOAc, 5/1) to afford 48 mg (58%) of the title compound. LCMS (ESI) calc'd for C3iH24ClF3N205 [M+H]+: 597.1, found: 597.1.
Step 2. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (3- hydroxyprop -1-ynyl)- lH-indazol-3-yl)benzoate (S-3). To a solution of methyl 4-(l-(2- chloro-6-(trifluoromethyl)benzoyl)-6-(3-(tetrahydro-2H-pyran-2-yloxy)prop- 1-ynyl)- 1H- indazol-3-yl)benzoate (S-2) (200 mg, 0.34 mmol) in MeOH (10 mL) was added TsOH (12 mg, 0.07 mmol) at 0 °C . The mixture was stirred for 12 hours at rt. The mixture was diluted with H20, The aqueous layer was extracted with CH2Cl2 (3 x80 mL). The combined organics were washed with H20, brine, dried over Na2S04, and concentrated. The residue was purified by Prep-TLC (Pentane/EtOAc, 3/1) to afford 110 mg (64%) of the title compound. LCMS (ESI) calc'd for C26Hi6ClF3N204 [M+H]+: 513.1, found: 513.1.
Step 3. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (3-hydroxy prop-l-ynyl)-lH-indazol-3-yl)benzoic acid (HA). To a stirred solution of methyl 4-(l-(2- chloro-6-(trifluoromethyl)benzoyl)-6-(3 -hydroxyprop- 1 -ynyl)- 1 H-indazol-3 -yl)benzoate (S-3) (300 mg, 0.5 mmol) was added THF (8.0 mL), H20 (2.0 mL) and LiOHH20 (108 mg, 2.5 mmol) and the solution was stirred at r.t. overnight. LCMS showed disappearance of staring material. The solution was adjusted to pH 4.0 using IN HC1 and poured into THF (30 mL), washed with brine (20 mL). The organic layer was dried over Na2S04, the organic layer was evaporated and submitted for Prep-HPLC. 100 mg product was collected. Yield: 32%. LCMS (ESI) calc'd for C25Hi4ClF3N204 [M+H]+: 499.1, found: 499.1. 1HNMR (400 MHz, DMSO) δ 8.544 (1H, s), 8.28 (1H, d), 8.10 ( 8H, m), 7.70 (1H, d), 4.41 (1H, s).
Example 12A: Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(3-hy droxybut-l-ynyl)-lH-indazol-3-yl)-3-fluorobenzoic acid (12A) SCHEME T
Figure imgf000107_0001
Step 1. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl) benzoyl)-6-(3- (tetrahydro-2H-pyran-2-yloxy)prop-l-ynyl)-lH-indazol-3-yl)-3-fluorobenzoate (T-2). To a solution of methyl 4-(6-bromo- l-(2-chloro-6-(trifluoromethyl)benzoyl)- lH-indazol-3-yl)-3-fluorobenzoate (T-1) (200 mg, 0.36 mmol) in DMF (10 mL) and TEA (20 mL) was added 2-(prop-2-ynyloxy)-tetrahydro-2H-pyran (60 mg, 0.43 mmol), copper(I) iodide (6.8 mg, 0.036 mmol) and PdCl2(PPh3)2 (25 mg, 0.036 mmol) under Argon . The mixture was stirred under Argon for 2 hours at 80 °C. The mixture was diluted with H20, The aqueous layer was extracted with CH2C12 (3 ><50 mL). The combined organics were washed with H20, brine, dried over Na2S04, and concentrated. The residue was purified by Prep-TLC (Pentane/EtOAc, 5/1) to afford 111 mg (51%) of the title compound. LCMS (ESI) calc'd for C3iH23ClF4N205 [M+H]+: 615, found: 615.
Step 2. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (3- hydroxyprop-l-ynyl)-lH-indazol-3-yl)-3-fluorobenzoate (T-3) To a solution of methyl 4- (l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(3-(tetrahydro-2H-pyran-2-ylox
y)prop-l-ynyl)-lH-indazol-3-yl)-3-fluorobenzoate (T-2) (111 mg, 0.18 mmol) in MeOH/H20 (20/2 mL) was added TsOH (15 mg, 0.09 mmol) at 0 °C . The mixture was stirred for 12 hours at rt. The mixture was diluted with H20, The aqueous layer was extracted with CH2C12 (3 x80 mL). The combined organics were washed with H20, brine, dried over Na2S04, and concentrated. The residue was purified by Prep-TLC (Pentane/EtOAc, 3/1) to afford filtered and concentrated to give 45 mg (47%) of the title compound. LCMS (ESI) calc'd for C26Hi5ClF4N204 [M+H]+: 531, found: 531.
Step 3. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) -6-(3- oxoprop-l-ynyl)-lH- indazol-3-yl)-3-fluorobenzoate (T-4) To a solution of methyl4-(l-(2- chloro-6-(trifluoromethyl)benzoyl)-6-(3 -hydroxyprop- 1 -ynyl)- 1 H-indazol-3 -yl)-3 - fluorobenzoate (T-3) (45 mg, 0.085 mmol) in DCM (20 mL) was added Dess-Martim reagent (108 mg, 0.25 mmol) at 0 °C. The mixture was stirred for 12 hours at rt. The mixture was diluted with H20 and extracted with CH2C12 (3 x80 mL). The combined organics were washed with H20, brine, dried over Na2S04, and concentrated. The residue was purified by Prep-TLC (Pentane/EtOAc, 3/1) to afford 40 mg (88%) of the title compound. LCMS (ESI) calc'd for C26Hi3ClF4N204 [M+H]+: 529.1, found: 529.1.
Step 4. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) -6-(3- hydroxybut-l-ynyl)-lH -indazol-3-yl)-3-fluorobenzoate (T-5) To a solution of methyl 4- ( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(3 -oxoprop- 1 -ynyl) - 1 H- indazol-3 -yl)-3 - fluorobenzoate (T-4) (40 mg, 0.076 mmol) in dry THF (5 mL) was added MeMgBr (0.18 mL, 0.53 mmol, 3 M in ether) at -60 °C. The mixture was stirred for 2 hours at rt, the mixture was quenched with saturated H4C1, The aqueous layer was extracted with EtOAc (3 x80 mL); the combined organics were washed with H20, brine, dried over Na2S04, and concentrated. The residue was purified by Prep-TLC (Pentane/EtOAc, 3/1) to afford 41 mg (85%) of the title compound. LCMS (ESI) calc'd for C27Hi7ClF4N204 [M+H]+: 545.1, found: 545.1.
Step 5. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) -6-(3- hydroxybut-l-ynyl)-lH -indazol-3-yl)-3-fluorobenzoate (12A). To a stirred solution of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl-6-(3-hydroxybut -l-ynyl)-lH -indazol-3- yl)-3 -fluorobenzoate (T-5) (41 mg, 0.075 mmol) was added THF (8.0 mL), H20 (2.0 mL) and LiOHH20 (32 mg, 0.75 mmol) and the solution was stirred at r.t. overnight. LCMS showed disappearance of staring material. The solution was adjusted to pH 4.0 using IN HC1 and poured into THF (30 mL), washed with brine (20 mL). The organic layer was dried over Na2S04, the organic layer was evaporated and submitted for Prep-HPLC to give 5 mg of product (yield 12%). LCMS (ESI) calc'd for C26Hi5ClF4N204 [M+H]+: 531, found: 531. 1HNMR (400 MHz, DMSO) δ 13.5 (1H, bs), 8.526 (1H, s), 7.95 (6H, m), 7.76 (1H, t), 7.63 (1H, t),5.61 (lH,d) , 4.57 (1H, t), 1.449- 1.464 (3H, d).
Example 13A: Preparation of 4-(6-(3-aminoprop-l-ynyl)-l-(2-chloro-6-(trifluo romethyl)benzoyl)-lH-indazol-3-yl)-3-fluorobenzoic acid (13A)
SCHEME U
Figure imgf000109_0001
13A
Step 1. Preparation of methyl 4-(6-(3-(tert-butoxycarbonylamino)prop-l- ynyl)-l -(2-chloro-6-(trifluoromethyl)benzoyl)-lH-indazol-3-yl)-3-fluorobenzoate (U-2). The mixture of methyl 4-(6-bromo-l-(2-chloro-6-(trifluoromethyl)benzoyl) -lH-indazol-3-yl)-3- fluorobenzoate (U-l) (554 mg, 1.0 mmol), Pd(PPh3)2Cl2 (105 mg,0.1 mmol), Cul (60 mg, 0.15 mmol), Et3N (20 mL) and DMF (20 mL) was purged with N2 and stirred at 80 °C overnight. LCMS showed the starting material was consumed totally and formation of the desired product. The resulting solution was filtered and concentrated to purify by column chromatography to give 432 mg of product as a white solid (68.7%). LCMS (ESI) calc'd for [M+H]+: 629.99, found: 630.1.
Step 2. Preparation of methyl 4-(6-(3-aminoprop-l-ynyl)-l-(2-chloro-6- (trifluoro methyl)benzoyl)-lH-indazol-3-yl)-3-fluorobenzoate (U-3): To the solution of methyl 4-(6- (3 -(tert-butoxycarbonylamino)prop- 1 -ynyl)- 1 -(2-chloro-6- (trifluorometh
yl)benzoyl)-lH-indazol-3-yl)-3-fluorobenzoate (U-2) (629 mg, 1.0 mmol) in THF (30 mL) and water (10 mL) was added TFA (970 mg, 10 mmol). The mixture solution was stirred at rt for 10 h. The mixture was diluted with H20 and exacted with EtOAc (30 mL><3). The combined organic layer was concentrated and purified by chromatography column (EA:PE=1 : 1) to afford 461 mg of product (yield: 87.5%). LCMS (ESI) calc'd [M+H]+: 529.87, found: 530.1.
Step 3. Preparation of 4-(6-(3-aminoprop-l-ynyl)-l-(2-chloro-6-(trifluo romethyl) benzoyl)-lH-indazol-3-yl)-3-fluorobenzoic acid (13A): To the solution of methyl 4-(6-(3- aminoprop- 1 -ynyl)- 1 -(2-chloro-6-(trifluoromethyl)benzoyl) - 1 H-indazol-3 -yl)
-3-fluorobenzoate (U-3) (529 mg, 1.0 mmol) in THF (30 mL) and water (10 mL) was added LiOH (240 mg, 10 mmol). The mixture solution was stirred at rt for 10 h. The mixture was acidified with 2N HC1 and exacted with EtOAc (30 mL><3). The combiend organic layer was purified by flash chromatography (EA:PE=1 : 1) to afford 431 mg of final product (yield: 84%). LCMS (ESI) calc'd [M+H]+: 515.84, found: 516.1. 1HNMR (400 MHz, CDC13) δ: 8.63 (1H, s), 8.41(2H, w), 8.02 (3H, m), 7.89 (3H, m), 7.76 (1H, t), 7.68 (1H, s), 4.12 (1H, s).
Example 14A: Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) -6-ethy nyl-lH-indazol-3-yl)benzoic acid (14A)
SCHEME V
Figure imgf000111_0001
4A Step 1.
Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl) benzoyl)-6-((tri
methylsilyl)ethynyl)-lH-indazol-3-yl)benzoate (V-2). To a solution of methyl 4-(6-bromo- l-(2-chloro-6-(trifluoromethyl)benzoyl)-lH-indazol-3-yl)benzoate (V-1) (108 mg, 0.2 mmol) in DMF (10 mL) and TEA (20 mL) was added ethynyltrimethylsilane (24 mg, 0.24 mmol), copper(I) iodide (4 mg, 0.02 mmol) and PdCl2(PPh3)2 (14 mg, 0.02 mmol) under argon. The mixture was stirred under argon for 2 hours at 80 °C. The mixture was diluted with H20. The aqueous layer was extracted with CH2C12 (3 ><50 mL). The combined organics were washed with H20, brine, dried over Na2S04, and concentrated. The residue was purified by Prep-TLC (Pentane/EtOAc, 5/1) to afford 90 mg (87%) of the title compound. LCMS (ESI) calc'd for C28H22ClF3N203Si [M+H]+: 555.1, found: 555.1.
Step 2. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) -6-ethy nyl-lH-indazol-3-yl)benzoate (V-3). To a solution of methyl 4-(l-(2-chloro-6-(triflu oromethyl)benzoyl)-6-((trimethylsilyl)ethynyl)-lH-indazol-3-yl)benzoate (V-2) (50 mg, 0.09 mmol) in THF (10 mL) at 0 °C was added TBAF (23 mg, 0.09 mmol). The mixture was stirred for 12 hours at rt. The mixture was diluted with H20. The aqueous layer was extracted with CH2C12 (3 x80 mL). The combined organics were washed with H20, brine, dried over Na2S04, and concentrated. The residue was purified by Prep-TLC (Pentane/EtOAc, 5/1) to afford 35 mg (92%) of the title compound. LCMS (ESI) calc'd for C25H14CIF3N2O3 [M+H]+: 483, found: 483.
Step 3. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-ethynyl-lH- indazol-3-yl)benzoic acid (14A). To a stirred solution of methyl 4-(l-(2-chloro-6- (trifluoromethyl)benzoyl)-6-ethynyl-lH-indazol-3-yl)benzoate (V-3) (170 mg, 0.35 mmol) was added THF (8.0 mL), H20 (2.0 mL) and LiOHH20 (74 mg, 1.76 mmol) and the solution was stirred at r.t. overnight. The solution was acidified with IN HC1 to pH = ~ 4.0, and diluted with THF (30 mL). The organic layer was separated and washed with brine (20 mL). The organic layer was dried over Na2S04, concentrated, and purified by Prep-HPLC to afford 43 mg of final product (yield: 26%). LCMS (ESI) calc'd for C25Hi4ClF3N203 [M+H]+: 483.1, found: 483.1. 1HNMR (500 MHz, DMSO) δ 13.36 (1H, bs), 8.56 (1H, s), 8.30 (1H, d),8.10 (4H, d), 8.05 (3H, m), 7.76 (1H, d), 4.59 (1H, s).
Example 15A: Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(3- methyl-l,2,4-oxadiazol-5-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (15A)
SCHEME W
Figure imgf000112_0001
15A St ep 1. Preparation of (2-chloro-6-(trifluoromethyl)phenyl)(3-iodo-6- (3-methyl- l,2,4-oxadiazol-5-yl)-lH-indazol-l-yl)methanone (W-2). The mixture of l-(2-chloro-6- (trifluoromethyl)benzoyl)-3-iodo-lH-indazole-6-carboxylic acid (W-1) (0.2 g, 0.4 mmol), N'- hydroxyacetimidamide (59 mg, 0.8 mmol), HATU (0.3 g, 0.8 mmol) and DIPEA (0.13 mL, 0.80 mmol) in DCM (20 mL) was stirred at rt for overnight. The reaction mixture was washed with 1M HC1 solution, saturated NaHC03 solution, and brine respectively, dried over anhydrous Na2S04 and concentrated. The residue was dissolved in 1,4-dioxane (20 mL ) and heated at 100 °C for overnight. The solvent was removed under reduced pressure and the residue was diluted with H20 (50 mL) and the aqueous layer was extracted with ethyl acetate (50 mLx3). The combined organic layers were washed with brine (50 mLx l), dried over anhydrous Na2S04 and concentrated. The residue was purified with Prep-TLC (PE:EA=5:2) to get the desired product 2 as a white solid (85 mg, 40%). LCMS (ESI) calc'd for Ci8H9ClF3IN402 [M+H]+: 533, found: 533.
Step 2. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl) benzoyl) -6-(3- methyl-l,2,4-oxadiazol-5-yl)-lH-indazol-3-yl)-3-fluorobenzoate (W-4). A mixture of (2- chloro-6-(trifluoromethyl)phenyl)(3-iodo-6-(3-methyl-l,2,4-oxa diazol-5-yl)-lH
-indazol-l-yl)methanone (W-2) (100 mg, 0.19 mol), W-3 (56 mg, 0.29 mol), Pd(OAc)2 (4 mg, 0.019 mol), s-phos (8 mg, 0.019 mol) and K3P04 (121 mg, 0.57 mol) were suspended in 1,4- dioxane (5 mL) and H20 (1 mL). The reation mixture was heated at 100 °C in a microwave reactor for 2 h. The result mixture was diluted with H20 (50 mL) and the aqueous layer was extracted with ethyl acetate (50 mLx3). The combined organic layers were washed with brine (50 mLx l), dried over anhydrous Na2S04 and concentrated to get the desired product W-4 as a yellow solid (65 mg, 45%). LCMS (ESI) calc'd for C26Hi5ClF4N404 [M+H]+: 559, found: 559.
Step 3. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl) benzoyl)-6-(3-methyl -1,2,4- oxadiazol-5-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (15A). A mixture of methyl 4-(l-(2- chloro-6-(trifluoromethyl)benzoyl)-6-(3-methyl -l,2,4-oxadiazol-5-yl) -lH-indazol-3-yl)-3- fluorobenzoate (W-4) (150 mg, 0.27 mmol) and LiOH (57 mg, 1.35 mmol) in THF (4 mL) and H20 (2 mL) was stirred at room temperature for 4 h.The reaction mixture was diluted with H20 (20 mL), and acidified with 2N HC1 solution to pH=~3. The mixture was extracted with ethyl acetate (20 mLx3). The combined organic layers were washed with brine (20 mLx l), dried over anhydrous Na2S04 and concentrated. The residue was purified with Prep- HPLC to get the desired product 15A as a white solid (130 mg, 90%). LCMS (ESI): calc'd for C25Hi3ClF4N404 [M+H]+: 545, found: 545; 1HNMR (400 MHz, MeOD) δ 8.36 (1H, s), 8.34 (1H, d, J=8.4 Hz), 8.17 (1H, d, J=8.4Hz), 7.97-7.99 (2H, m), 7.89-7.94 (2H, m), 7.81 J = 8.4 Hz), 7.73-7.78 (1H, m), 2.54 (3H, s).
Example 16A: Preparation 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(5-(hydr oxymethyl)oxazol-2-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (16A)
SCHEME X
Figure imgf000114_0001
16A Step 1: Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(prop
-2-ynylcarbamoyl)-lH-indazol-3-yl)-3-fluorobenzoate (X-2). l-(2-Chloro -6-triflu oromethyl)benzoyl)-3-(2-fluoro-4-(methoxycarbonyl)phenyl )-lH-indazole-6-carboxylic acid (X-1) (100 mg, 0.2 mmol) was dissolved in CH2C12 (15 mL), followed by the additon of prop- 2-yn-l-amine (13 mg, 0.24 mmol) and PYAOP (208 mg, 0.4 mmol). The mixture was stirred at room temperature for 2 mins, followed by the addition of TEA (0.16 mL, 1.08 mmol). The mixture was stirred at room temperature for 2 h, diluted with EtOAC (20 mL), washed with brine (20 mL><2), dried over anhydrous Na2S04, concentrated to give 85 mg of crude product, which was used to the next step without further purification. LCMS (ESI): calc'd for C27Hi6ClF4N304, [M+H]+: 557.1, found: 557.1.
Step 2: Preparation of methyl 4-(6-(5-(acetoxymethyl)oxazol-2-yl)-l-(2-chloro-6- (trifluoromethyl)benzoyl)-lH-indazol-3-yl)-3-fluorobenzoate (X-3). Methyl-(l-(2- chloro-6-(trifluoromethyl)benzoyl)-6-(prop-2-ynylcarbamoyl)-lH-indazol-3-yl)-3- fluorobenzoate (X-2) (85 mg, 0.15 mmol) was dissolved in AcOH (15 mL), followed by the addition of (diacetoxyiodo) benzene (73 mg, 0.23 mmol). The mixture was stirred at 90 °C for 12 hours. Then it was diluted with EtOAC (20 mL), washed with brine (20 mL><2), dried over anhydrous Na2S04, concentrated to get the crude product X-3 (60 mg), which was used to the next step without further purification. LCMS (ESI): calc'd for C3oHi9ClF4N206, [M+H]+: 614.1, found: 614.1.
Step 3: Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(5-(hydroxyl methyl)oxazol-2-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (16A). A mixture of methyl 4- (l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(prop-2-ynyl carbamoyl)- lH-ind
azol-3-yl)-3 -fluorobenzoate (X-3) (25 mg, 0.04 mmol) and LiOHH20 (10 mg, 0.25 mmol) in 10 mL THF and 10 mL pure H20 was stirred at RT for 2 hours. The solvent was evaporated and the residue was dissolved in water. HC1 (5% sol. in water) was added until pH 4-5. The precipitated solid was filtered, washed with water and n-hexane, dried to afford an off white solid 16A (15 mg, 80%). LCMS (ESI): calc'd for C27Hi5ClF4N205, [M+H]+: 558.1 found: 558.1. 1HNMR (400 MHz, DMSO) δ 9.09 (1H, s), 8.22-8.24 (1H, d, J = 8 Hz), 8. 11-8.13 (1H, d, J = 8 Hz), 8.05-8.07 (1H, d, J = 8 Hz), 8.00-8.02QH, d, J = 8 Hz), 7. 88-7.95 (3H, m), 7.77-7.81 (1H, m), 7.35 (1H, s), 4.63 (2H, s).
Example 17A: Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(l-me thyl-lH-imidazol-4-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (17A)
Figure imgf000115_0001
Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(4,4,5,
5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-indazol-3-yl)-3-fluorobenzoate (Y-2). To the solution of methyl 4-(6-bromo-l-(2-chloro-6-(trifluoromethyl)benzoyl)-lH-ind
azol-3-yl)-3 -fluorobenzoate (Y-l) (161 mg, 0.29 mmol) in dioxane (5 mL) was added Pin2B2
(151 mg, 0.58 mmol), KOAc (115 mg, 1.16 mmol) and (dppf)PdCl2 (21 mg, 0.029 mmol) under N2 protection. The mixture was heated at 90 °C for 6 h. Then the mixture was cooled down and diluted with EtOAc (100 mL), washed with H20 (20 mL 3), and brine (20 mL), dried and concentrated. The residue was purified with chromatography (PE:EA=6: 1) to afford 125 mg of final product (yield: 71%). LCMS (ESI) calc'd [M+H]+: 602.0, found:602.1.
Step 2. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(l-me thyl-lH-imidazol-4-yl)-lH-indazol-3-yl)-3-fluorobenzoate (Y-3). To the solution of methyl 4-(6-bromo- 1 -(2-chloro-6-(trifluoromethyl)benzoyl) - 1 H-indazol-3 -yl)-3 -fluor
obenzoate (Y-2) (50 mg, 0.083 mmol) in dioxane (5 mL) was added 4-bromo-l-meth yl-lH-imidazole (20 mg, 0.1 mmol) , K2C03 (30 mg, 0.2 mmol) and (PPh3)4Pd (10 mg, 0.0083 mmol) under N2 protection. The mixture was protected by N2 and stirred at 85 °C for 16 h. The solution was cooled down, concentrated, and purified by Prep-HPLC (ACN:H20) to afford 5 mg of product (yield: 7.2 %). LCMS (ESI) calc'd [M+H]+: 557, found: 557.
Step 3. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(l-methyl-lH
-imidazol-4-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (17 A). To the solution of methyl 4- ( 1 -(2-chloro-6-(trifluoromethyl)benzoyl) -6-( 1 -methyl- 1 H-imidazol-4-yl)- 1
H-indazol-3 -yl)-3-fluorobenzoate (Y-3) (5 mg, 0.009 mmol) in THF (2 mL) and H20 (0.5 mL) was added LiOH (1 mg, 0.045 mmol). The mixture solution was stirred at rt for 16 h, acidified by HC1 (2N), extracted with EtOAc (10 mL><3). The organic layer was dried and concentrated to be purified by Prep-HPLC (ACN:H20) to afford 5 mg of final product (yield: 100%). LCMS (ESI) calc'd [M+H]+: 543, found: 543.
Example 18A: Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(oxaz ol-2-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (18A)
Figure imgf000117_0001
p 1. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(4,5- dihydrooxazol-2-yl)-lH-indazol-3-yl)-3-fluorobenzoate (Z-2). A mixture of l-(2-chloro-6- (trifluoromethyl)benzoyl)-3-(2-fluoro-4-(methoxycarbonyl)phenyl)-lH- indazole-6-carboxylic acid (Z-l) (286 mg, 0.55 mmol) and (COCl)2 (0.14 mL, 1.65 mmol) in DCM (5 mL) and DMF (2 drops) was stirred at room temperature for 1 h. The solvent was removed and the residue was dissolved in anhydrous toluene (5 mL). The resulting solution was added to a mixture of 2-bromoethanamine (86 mg, 0.7 mmol) and Et3N (167 mg, 1.65 mmol) in anhydrous toluene (5 mL). The reaction mixture was stirred at 85 °C for 3 hours. The solvent was evaporated and the residue was purified by flash chromatography (PE:DCM=1 : 10) to give the desired product Z-2. LCMS (ESI) calc'd for C26Hi6ClF4N304 [M+H]+: 546, found: 546.
Step 2. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(oxa zol-2-yl)-lH-indazol-3-yl)-3-fluorobenzoate (Z-3). The mixture of methyl 4-(l-(2-chloro-6- (trifluoromethyl)benzoyl)-6-(4, 5 -dihydrooxazol-2-yl)- 1 H-indazol-3 -yl)-3 -fluorobenzoate (Z- 2) (300 mg, 0.55 mmol), AIBN (9 mg, 0.055 mmol) and NBS (587 mg, 3.3 mmol) in CC14 (15 mL) under argon, the reaction mixture was refluxed for 12 hours. The solvent was evaporated and the residue was purified by column chromatography on silica gel eluting with (PE:DCM=1 :4) to get the desired product Z-3 (200 mg, 66%). LCMS (ESI) calc'd for C26Hi4ClF4N304 [M+H]+: 544, found: 544. Step 3. Preparation of methyl 4-(6-(5-bromooxazol-2-yl)-l-(2-chloro-6-(trifluor omethyl)benzoyl)-lH-indazol-3-yl)-3-fluorobenzoate (Z-4). The same to the preparation of compound Z-3 to get the desired product Z-4. LCMS (ESI) calc'd for C26Hi3BrClF4N30 [M+H]+: 622, found: 622.
Step 4. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl) -6-(oxazol-2- yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (18A). A mixture of methyl 4-(l-(2-chloro-6- (trifluoromethyl)benzoyl)-6-(oxazol-2-yl)-lH-indazol-3-yl)-3-fluorobenzoate (Z-3) (38 mg, 0.07 mmmol) and LiOHH20 (16 mg, 0.37 mmol) in 10 mL THF and 10 mL pure H20 was stirred at RT for 2 hours. The solvent was evaporated and the residue was dissolved in water. HC1 (5% sol. in water) was added until pH = 4-5. The precipitated solid was filtered, washed with water (10 mL) and n-hexane (10 mL), dried to afford an off white solid 18A (32 mg, 93%). LCMS (ESI): calc'd for C25Hi2ClF4N304, [M+H]+: 530 found: 530; 1HNMR (400 MHz, DMSO) δ 13.57 (IH, s), 9.12 (IH, s), 8.40 (IH, s), 8.24-8.26 (IH, d, J= 8 Hz), 8.12-8.14 (IH, d, J= 8 Hz), 8.00-8.07 (2H, m), 7.88-7.95 (3H,m), 7.78-7.81 (IH, m) , 7.55 (IH, s).
Step 5. Preparation of 4-(6-(5-bromooxazol-2-yl)-l-(2-chloro-6-(trifluorometh
yl)benzoyl)-lH-indazol-3-yl)-3-fluorobenzoic acid (18B). The same to the preparation of compound 18A to get the desired product 18B. LCMS (ESI): calc'd for C25HnBrClF4N304, [M+H]+: 608 found: 608; 1HNMR (400 MHz, DMSO) δ 13.57 (IH, s), 9.05 (IH, s), 8.19- 8.21 (IH, d, J= 8 Hz), 8.12-8.14 (IH, d, J= 8 Hz), 8.05-8.06 (IH, d, J= 4 Hz), 8.00-8.02 (IH, d, J= 8 Hz), 7.93-7.95 (IH, d, J= 8 Hz), 7.86-7.88 (2H, m), 7.77-7.80 (IH, m), 7.61 (IH, s).
Example 19A: Preparation of (E)-4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (N'-cyano-N,N-dimethylcarbamimidoyl)-lH-indazol-3-yl)-3-fluorobenzoic acid (19A)
Figure imgf000119_0001
Figure imgf000119_0002
Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(dime
thylcarbamoyl)-lH-indazol-3-yl)-3-fluorobenzoate (AA-2). The mixture of l-(2- chloro-6-(trifluoromethyl)benzoyl)-3-(2-fluoro-4-(methoxycarbonyl)phenyl)-lH-indazole-6- carboxylic acid (AA-1) (250 mg, 0.48 mmol), dimethylamine (2.0 M solution in THF, 0.36 mL, 0.72 mmol), HATU (220 mg, 0.58 mmol) and Et3N (0.13 mL, 0.96 mmol) in DCM (10 mL) was stirred at room temperature for 1 h. The solvent was removed under reduced pressure and the residue was chromatographed on silica gel (PE:EA 1 : 1) to get the desired product as a white solid (200 mg, 86%). LCMS (ESI) calc'd for C26Hi8ClF4N304 [M+H]+: 548, found: 548.
Step 2. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(dime thylcarbamothioyl)-lH-indazol-3-yl)-3-fluorobenzoate (AA-3). The mixture of methyl4- (l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(dimethylcarbamoyl)-lH-indazol
-3-yl)-3-fluorobenzoate (AA-2) (240 mg, 0.44 mmol) and Lawesson Reagent (360 mg, 0.88 mmol) in toluene (10 mL) was stirred at 100 °C for 4 h. The solvent was removed under reduced pressure and the residue was chromatographed on silica gel (PE:EA 5: 1) to get the desired product as a yellow solid (180 mg, 70%). LCMS (ESI) calc'd for C26Hi8ClF4N303S [M+H]+: 564, found: 564.
Step 3. Preparation of tetrafluoroborate of methyl 4-(l-(2-chloro-6-(trifluoro methyl) benzoyl) -6-((dimethylamino)(ethylthio)methyl)-lH-indazol-3-yl)-3-fl uorobenzoate (AA-
4). The mixture of methyl 4-(l-(2-chloro-6-(trifluor omethyl)benzoyl)-6- (dimethylcarbamothioyl)-lH-indazol-3-yl)-3-fluorobenzoate (AA-3) (145 mg, 0.26 mmol) and triethyloxonium tetrafluoroborate (1.0 M solution in DCM, 0.31 mL, 0.31 mmol) in DCE (6 mL) was stirred at 85 °C for overnight. The solvent was removed under reduced pressure to get the crude product as a yellow solid. The crude product was used in the next step without further purification. LCMS (ESI) calc'd for C28H23BC1F8N303S [M+H]+: 592, found: 592.
Step 4. Preparation of (E)-methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6
-(N'-cyano-N,N-dimethylcarbamimidoyl)-lH-indazol-3-yl)-3-fluorobenzoate (AA-5). The mixture of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-((dimeth
ylamino)(ethylthio)methyl)-lH-indazol-3-yl)-3-fluorobenzoate (AA-4) (150 mg, 0.26 mmol), cyanamide (37 mg, 0.88 mmol) and Et3N ( 44 mg, 0.44 mmol) in MeOH (5 mL) was stirred at room temperature for 2 h. The result solution was diluted with water (30 mL) and the aqueous layer was extracted with EtOAc (20 mL><3). The combined organic layers were washed with IN HCl solution (20 mLx l) then brine (20 mLx l), dried over anhydrous Na2S04 and concentrated to get the desired crude product AA-5 (160 mg) as a yellow solid. LCMS (ESI) calc'd for C27Hi8ClF4N503 [M+H]+: 572, found: 572.
Step 5. Preparation of (E)-4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(N'-cya no-N,N-dimethylcarbamimidoyl)-lH-indazol-3-yl)-3-fluorobenzoic acid (19A). The mixture of (E)-methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(N'-cyano-N,
N-dimethylcarbamimidoyl)-lH-indazol-3-yl)-3-fluorobenzoate (AA-5) (100 mg, 0.18 mmol) and LiOH (30 mg, 0.72 mmol) in THF (4 mL) and H20 (2 mL) was stirred at room temperature for 3 h. The reaction mixture was diluted with H20 (20 mL). 2 M HCl solution was added to adjust the pH=3 and the aqueous layer was extracted with ethyl acetate (20 mLx3). The combined organic layers were washed with brine (20 mLx l), dried over anhydrous Na2S04 and concentrated. The residue was purified with Prep-HPLC (acetonitrile- water system) to get the desired product 6 as a white solid (50 mg, 50%). LCMS (ESI) calc'd for C26Hi6ClF4N503 [M+H]+: 558, found: 558; 1HNMR (400 MHz, MeOD) δ 8.68 (1H, s), 8.15-8.18 (1H, m), 7.93-7.98 (2H, m), 7.87-7.91 (2H, m), 7.77-7.81 (1H, m), 7.72-7.76 (1H, m), 7.61 (1H, d, J=8.4Hz), 3.37 (3H, s), 3.09 (3H, s).
Example 20A: Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(5-oxo-4,5- dihydro-l,3,4-oxadiazol-2-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (20A)
SCHEME AB
Figure imgf000121_0001
AB-4 AB-5 20A Sfe p 1. Preparation of methyl l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-iodo-lH
-indazole-6-carboxylate (AB-2). To a stirred solution of methyl 3-iodo-lH-indazole
-6-carboxylate (AB-1) (2 g, 6.62 mmol) in anhydrous DCM (60 mL) at rt was added 2- chloro-6-(trifluoromethyl)benzoyl chloride (2.4 g, 9.93 mmol), DMAP (161 mg, 1.32 mmol), Et3N (1.47 mg, 14.57 mmol). The solution was stirred at rt overnight. The solution was diluted with EtOAc (50 mL), filtered through celite and washed with DCM (40 mL). The combined organic layer was washed with H20 (20 mL), brine (20 mL) and dried over anhydrous Na2S04. The solution was evaporated with silica gel and loaded on a column. SGC (DCM) afforded 3.25 g product. Yield 98.5%. LCMS (ESI) calc'd for Ci7H9ClF3IN203 [M+H]+: 509, found: 509.
Step 2. Preparation of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-iodo-lH-indaz ole-6-carbohydrazide (AB-3). To a stirred solution of methyl l-(2-chloro-6-(trifluoro methyl)benzoyl)-3-iodo-lH-indazole-6-carboxylate (AB-2) (1000 mg, 1.97 mmol) in ethanol (40 mL) was added H2 H2 H20 (0. 31 ml, 9.83 mmol). The solution was stirred at reflux temperature overnight. LCMS showed complete transformation to the product. The solution was diluted with H20 (100 mL), extracted with EtOAc (3 x60 mL). The combined organic layer was washed with H20 (2x20 mL), brine (30 mL) and dried over anhydrous Na2S04. The solution was evaporated with silica gel and loaded on a column. Prep-TLC (DCM) afforded 176 mg product. Yield 18%. LCMS (ESI) calc'd for Ci6H9ClF3IN402 [M+H]+: 509, found: 509.
Step 3. Preparation of 5-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-iodo-lH-inda zol-6-yl)-l,3,4-oxadiazol-2(3H)-one (AB-4). To a stirred solution of l-(2-chloro-6-(tr ifluoromethyl)benzoyl)-3-iodo-lH-indazole-6-carbohydrazide (AB-3) (100 mg, 0.196 mmol) in THF (2 mL) was added CDI (48 mg, 0.294 mmol) and Et3N (30 mg, 0.294 mmol). The solution was stirred at rt overnight. The solution was diluted with H20 (30 mL), extracted with EtOAc (2x30 mL) and organic layer was washed with H20 (30 mL) and brine (30 mL) and dried over anhydrous Na2S04. The organic layer was evaporated with silica gel and loaded on a silica gel column. SGC (PE/EA: 2/1) gave 60 mg product, yield 57%. LCMS (ESI) calc'd for Ci7H7ClF3IN403 [M+H]+: 535, found: 535.
Step 4. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(5-oxo -4,5-dihydro-l,3,4-oxadiazol-2-yl)-lH-indazol-3-yl)-3-fluorobenzoate (AB-5). To a microwave tube was added 5-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-iodo-lH- indazol-6-yl)-l,3,4-oxadiazol-2(3H)-one (AB-4) (60 mg, 0.11 mmol), methyl 3-fluoro -4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)benzoate (44 mg, 0.22 mmol), Pd(OAc)2 (trimer) (1.23 mg, 0.01 mmol), s-Phos (4.4 mg, O.Olmmol), K3P04 (80 mg, 0.33 mmol), THF(1.5 mL), H20 (0.3 mL). The solution was microwaved under Argon at 110 °C for 2 hours. LCMS showed major product peak. The upper solution was filtered and submitted for next step without further purification. LCMS (ESI) calc'd for C25Hi3ClF4N405[M+H]+: 560, found: 560.
Step 5. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(5-oxo-4,5-di hydro-l,3,4-oxadiazol-2-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (20A). To a stirred solution of methyl4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(5-oxo-4,5-di
hydro-l,3,4-oxadiazol-2-yl)-lH-indazol-3-yl)-3-fluorobenzoate (AB-5) (as generated in step 4) was added LiOHH20 (10 mg, 0.4 mmol) and H20 (0.1 mL). The solution was stirred overnight and LCMS showed major product peak. The solution was adjusted to PH 3.0 using 1 N HCl. The upper organic layer was collected and the aqueous layer was extracted with THF (3 x 1 mL). The combined organic layer was added 0.5 mL MeOH and submitted for Prep-HPLC. Prep-HPLC (H20/ACN, 0.05% TFA) gave 15 mg product, yield for two steps 25%. LCMS (ESI) calc'd for C^HnCu^^Os [M+H]+: 547, found: 547. 1HNMR (400 MHz, DMSO) δ 13.60 (1H, s), 13.00 (1H, s), 8.87 (1H, s), 8.15-8.07 (1H, m), 8.06-8.03(3H, m), 8.02-8.00(3H, m), 7.79-7.76 (1H, m).
Example 21A: Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(4H- l,2,4-triazol-3-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (21A)
SCHEME AC
Figure imgf000123_0001
Step 1. Preparation of methyl 4-(6-carbamoyl-l-(2-chloro-6-(trifluoromethyl)ben zoyl)-lH-indazol-3-yl)-3-fluorobenzoate (AC-2). The mixture of l-(2-chloro-6- (trifluoromethyl)benzoyl)-3-(2-fluoro-4-(methoxycarbonyl)phenyl)-lH-indazole-6-carboxylic acid (AC-1) (95 mg, 0.18 mmol), ammonium chloride (14.3 mg, 0.27 mmol), HATU (82 mg, 0.22 mmol) and Et3N (76 uL, 0.54 mmol) in DCM (5 mL) was stirred at room temperature for 1 h. The solvent was removed under reduced pressure and the residue was chromatographed on silica gel (PE:EA 1 : 1) to get the desired product as a white solid (60 mg, 68%). LCMS (ESI) calc'd for C24Hi4ClF4N304 [M+H]+: 520, found: 520.
Step 2. Preparation of (E)-methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(( dimethylamino)methylenecarbamoyl)-lH-indazol-3-yl)-3-fluorobenzoate (AC-3). The mixture of methyl 4-(6-carbamoyl-l-(2-chloro-6-(trifluoromethyl)benzoyl)-lH- indazol-3-yl)-3-fluorobenzoate (AC-2) (80 mg, 0.15 mmol) and dimethoxy-N,N- dimethylmethanamine (92 mg, 0.75 mmol) in EtOAc (5 mL) was stirred at 60 °C for overnight. The solvent was removed under reduced pressure to get the crude product as a white solid. The crude product was used in the next step without further purification. LCMS (ESI) calc'd for C27Hi9ClF4N404 [M+H]+: 575, found: 575.
Step 3. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(4H- l,2,4-triazol-3-yl)-lH-indazol-3-yl)-3-fluorobenzoate (AC-4). The mixture of (E)-methyl 4- (l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-((dimethylamino)methylene
carbamoyl)- lH-indazol-3-yl)-3-fluorobenzoate (AC-3) (100 mg, 0.17 mmol) and hydrazine (27 mg, 0.85 mmol) in AcOH (5 mL) was stirred at room temperature for 2 h. The result solution was diluted with water (30 mL). NaHC03 solid was added to adjust the pH=8 and the aqueous layer was extracted with EtOAc (20 mL χ3). The combined organic layers were washed with brine (20 mL x l), dried over anhydrous Na2S04 and concentrated to get the desired product as a white solid (50 mg, 50%). LCMS (ESI) calc'd for C25Hi4ClF4N503 [M+H]+: 544, found: 544.
Step 4. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(4H-l,2,4-tr iazol-3-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (21A). The mixture of methyl 4-(l-(2- chloro-6-(trifluoromethyl)benzoyl)-6-(4H-l,2,4-triazol-3-yl)-lH-indazol-3-yl)-3- fluorobenzoate (AC-4) (100 mg, 0.18 mmol) and LiOH (38 mg, 0.90 mmol) in THF (4 mL) and H20 (2 mL) was stirred at 30 °C for lh. The reaction mixture was diluted with H20 (20 mL). 2M HC1 solution was added to adjust the pH=3 and the aqueous layer was extracted with ethyl acetate (20 mL x3). The combined organic layers were washed with brine (20 mLx l), dried over anhydrous Na2S04 and concentrated. The residue was purified with Prep- HPLC (acetonitrile-water system) to get the desired product 21A as a white solid (95 mg, 96%). LCMS (ESI) calc'd for C24Hi2ClF4N503 [M+H]+: 530, found: 530. 1HNMR (400 MHz, MeOD) δ 9.32 (IH, s), 8.56 (IH, s), 8.30 (IH, d, J=8.4Hz), 8.02-8.05 (IH, m), 7.92-7.97 (2H, m), 7.86-7.90 (2H, m), 7.72-7.80 (2H, m).
TABLE 10:
Figure imgf000125_0001
Figure imgf000125_0002
imidazol-2-yl)- lH-indazol-3-yl)- 3-fluorobenzoic
acid
Example 22A: Preparation of 4-(l-(2-chloro-6-trifluorobenzoyl)-6-(thiazol-2-yl)-lH -indazol-3-yl)benzoic acid (22A)
SCHEME AD
Figure imgf000126_0001
Step: Preparation of 4-(l-(2-chloro-6-trifluorobenzoyl)-6-(thiazol-2-yl)-lH-indazol -3-yl)benzoic acid (22 A). To a 1 dram vial was added methyl 4-(l-(2-chloro-6- trifluorobenzoyl)-6-(4,4, 5 , 5 -tetramethyl- 1 , 3 ,2-dioxaborolan-2-yl)- 1 H-indazol-3 -yl)benzoate
(AD-1) (20 mg, 0.034 mmol), 2-bromothiazole (AD-2) (0.051 mmol), potassium carbonate (0.093 mL, 2M), dichloro l-l'-bis(diphenylphosphino)ferrocine palladium (II) dichloromethane adduct (5.59 mg, 6.84 umol), and 1,4-dioxane (1 mL). The vessel was flushed with Argon and stirred overnight at 90 °C. The reactions were concentrated under reduced pressure. The remaining residue was dissolved in a 1 : 1 DCM/Methanol solution (1 mL) and SiliaBond DMT Resin (78 mg, 0.045 mmol) was then added. The mixture was stirred overnight at room temperature. The mixture was then filtered and concentrated under reduced pressure. Lithium hydroxide (1M, 0.186 mL), methanol (0.25 mL), and THF (0.5 mL) was then added and the reaction was stirred overnight at RT. The reactions were concentrated under reduced pressure. The reaction was then diluted with 1.0 mL DMSO, filtered, and purified by mass-triggered reverse phase HPLC, eluting with a 1% trifluoroacetic acid buffered water/acetonitrile gradient over a Waters X-B ridge C-18 column, to afford desired product (5 mg, 18% over two steps). LCMS (ESI) calc'd for C24H13CIFN3O3S [M+H]+: 478, found: 478. TABLE 11:
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
yl]benzoic acid
Example 23A: Preparation of 4-(l-(2-chloro-6-trifiuorobenzoyl)-6-(thiazol-2-yi)-lH- indazol-3-yl)benzoic acid (23A) Scheme AE
Figure imgf000130_0001
Step 1: Preparation of 4-(l-(2-chloro-6-trifluorobenzoyl)-6-(pyridin-3-yl)-lH- indazol-3- yl)benzoic acid (23 A). To a 1 dram vial was added methyl 4-(6-bromo-l-(2-chloro-6- trifluorobenzoyl)-lH-indazol-3-yl)benzoate (i-9) (20 mg, 0.037 mmol), pyridin-3-ylboronic acid (6.9 mg, 0.056 mmol), potassium carbonate (2 M, 0.093 mL, 0.186 mmol), dichloro 1-Γ- bis(diphenylphosphino)ferrocine palladium(II) dichloromethane adduct (6.07 mg, 7.44 umol), and 1,4-dioxane (1 mL). The vessel was flushed with Argon and stirred overnight at 90°C. The reactions were concentrated under reduced pressure. The remaining residue was dissolved in a 1 : 1 DCM/Methanol solution (1 mL) and SiliaBond DMT Resin (78 mg, 0.045 mmol) was then added. The mixture was stirred overnight at room temperature. The mixture was then filtered and concentrated under reduced pressure. Lithium hydroxide (1M, 0.186 mL), methanol (0.25 mL) and THF (0.5 mL) was then added and the reaction was stirred overnight at RT. The reactions were concentrated under reduced pressure. The reaction was then diluted with 1.0 mL DMSO, filtered, and purified by mass-triggered reverse phase HPLC, eluting with a 1% trifluoroacetic acid buffered water/acetonitrile gradient over a Waters X-B ridge C-18 column, to afford desired product. LCMS (ESI) calc'd for C26H15CIFN3O3 [M+H]+: 472, found: 472. 1H 1H MR δ (ppm)(DMSO-d6): 7.66 (1 H, t, J = 6.33 Hz), 7.85 (1 H, t, J = 8.15 Hz), 7.99-7.98 (6 H, m), 8.08 (2 H, d, J = 8.17 Hz), 8.37 (2 H, d, J = 8.35 Hz), 8.71 (1 H, d, J = 4.84 Hz), 8.78 (1 H, s), 9.09 (1 H, s).
TABLE 5:
Figure imgf000131_0001
LCMS [M+H]+
Ex. Chemical Name Structure
Calc'd/Found 3B 4-(l-{[2-chloro-6- 472/472
(trifluoromethyl)ph
enyl]carbonyl}-6- pyridin-4-yl- 1 H-indazol- 3-yl)benzoic acid
Figure imgf000131_0002
3C 4-[l-{[2-chloro-6- 0. 496/496
(trifluoromethyl)ph
enyl]carbonyl}-6- (4-cyanophenyl)- lH-indazol-3-yl]benzoic
acid F3C
Figure imgf000132_0001
Figure imgf000133_0001
Example 26A: Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)bi
(methylsulfonamidomethyl)-lH-indazol-3-yl)benzoic acid (26A)
Scheme AH
Figure imgf000134_0001
Step 1. Preparation of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-(4-(methoxy- carbonyl)phenyl)-lH-indazole-6-carboxylic acid (AH-1). A mixture of i-8 (300 mg, 0.61 mmol), 4-(methoxycarbonyl)phenylboronic acid (165 mg, 0.92 mmol), Pd(dppf)Cl2 (50 mg, 0.061 mmol) and KOAc (181 mg, 1.83 mmol) in 10 mL dioxane and 2 mL pure H20 was heated to 95 °C for 2h with microwave. Then it was diluted with EtOAC (50 mL), washed with brine (50 mL x 2), dried over anhydrous Na2S04, concentrated. Then it was purified by silica gel column (PE/EtOAc=20/l) to get white solid 180 mg (59%). LCMS (ESI): calc'd for C24Hi4ClF3N205, [M+H]+: 503.1, found: 503.1
Step 2. Preparation of methyl 4-(6-carbamoyl-l-(2-chloro-6-(trifluoromethyl)- benzoyl)- lH-indazol-3-yl)benzoate (AH-2). The compound AH-1 (100 mg, 0.2 mmol) was dissolved in CH2C12 (15 mL). H4C1 (13 mg, 0.24 mmol), PYAOP (208 mg, 0.4 mmol) was added and the mixture was stirred at room temperature for 2 min. TEA (0.16 mL, 1.08 mmol) was added and the mixture was stirred at room temperature for 2h. Then it was diluted with EtOAC (20 mL), washed with brine (20 mL x 2), dried over anhydrous Na2S04, concentrated get white solid AH-2 90 mg (90%). LCMS (ESI): calc'd for C24Hi5ClF3N304, [M+H]+: 502, found: 502. Step 3. Preparation of methyl 4-(6-(aminomethyl)-l-(2-chloro-6-(trifluoromethyl) benzoyl)-lH-indazol-3-yl)benzoate (AH-3). The compound AH-2 (90 mg, 0.18 mmol) was dissolved in anhydrous THF (20 mL) under argon, BH3.THF (0.9 mL, 0.9 mmol) was added and the mixture was refluxed for 12 h. MeOH was added to quench the excess BH3. The mixture was evaporated and then got white solid 75 mg. LCMS (ESI): calc'd for C24Hi7ClF3N303, [M+H]+: 488.1, found: 488.1.
Step 4. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (methylsulfonamidomethyl)-lH-indazol-3-yl)benzoate (AH-4). To a 50 mL round- bottomed flask was added compound AH-3 (100 mg, 0.2 mmol), methanesulfonyl chloride (23 mg, 0.2 mmol) and CH2C12 (10 mL), after stirring at rt for 3 min, TEA (0.1 mL, 0.6 mmol) was added slowly. The reaction mixture was stirred at rt overnight. Then the mixture was poured to 30 mL water, the lower (organic) and upper (aqueous) phases were separated. The aqueous phase was extracted CH2C12 (20 mL x 2). The combined organic phases were washed successively with water (20 mL x 2) and 10 mL of brine. The organic phase was dried over anhydrous sodium sulfate, filtered and concentrated at reduced pressure to give solid 22 mg. LCMS (ESI): calc'd for C25Hi9ClF3N305S [M+H]+: 566.1, found: 566.1.
Step 5. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(methyl- sulfonamidomethyl)-lH-indazol-3-yl)benzoic acid (26A). A mixture of methyl 4-(l-(2- chloro-6-(trifluoromethyl)benzoyl)-6-(methylsulfonamidomethyl)-lH-indazol-3-yl)benzoate (AH-4) (22 mg, 0.04 mmol) and LiOH.H20 (8 mg, 0.19mmol) in 10 mL THF and 10 mL pure H20 was stirred at rt for 2h. The solvent was evaporated and the residue was dissolved in water. HC1 (5% sol in water) was added until pH 4-5. The precipitated solid was filtered, washed with water and n-hexane, dried to afford an off-white solid 15 mg (80%). LCMS (ESI): calc'd for C24Hi7ClF3N305S, [M+H]+: 552.1 found: 552.1; 1HNMR (400MHz, DMSO- d6) δ 8.59 (s, 1H), 8.23-8.25 (d, lH, J =8Hz), 8.09-8.11 (d, 2H, J=8Hz), 8.04-8.06 (d, 1H, J=8Hz), 7.99-8.01 (d, 1H, J=8Hz), 7.94-7.96 (d, 2H, J=8Hz), 7.86-7.90 (m, 2H), 7.64-7.66 (d, 1H, J=8Hz), 4.46-4.48 (d, 2H, J=8Hz), 2.99 (s, 3H). Example 27A: Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(2,5- dioxoimidazolidin-4-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (27A)
Scheme Al
Figure imgf000136_0001
27A
Step 1. Preparation of (2-chloro-6-(trifluoromethyl)phenyl)(6-(hydroxymethyl)- 3-iodo- lH-indazol-l-yl)methanone (AI-1). To a stirred solution of l-(2-chloro-6- (trifluoromethyl)benzoyl)-3-iodo-lH-indazole-6-carboxylic acid (i-8) (2.00 g, 4.0 mmol) in THF (60 mL) was added BH3 THF (20 mL, 20 mmol). The solution was refluxed overnight. The solution was evaporated and the solid weighted 2.5 g. It proceeded to next step without further purification. LCMS (ESI) calc'd for Ci6H9ClF3IN202 [M+H]+: 480.7, found: 480.9.
Step 2. Preparation of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-iodo-lH- indazole-6- carbaldehyde (AI-2). To a stirred solution of (2-chloro-6- (trifluoromethyl)phenyl)(6- (hydroxymethyl)-3-iodo-lH-indazol-l-yl)methanone (AI- 1) (2.5 g, 0.52 mmol) in DCM (160 mL) was added Des Martin Periodinane (3.7 g, 0.78 mmol). The solution was stirred for lh. The mixture was filtered, the filtrate was washed with H20 (100 mL) and brine (100 mL) and dried over Na2S04. The solvent was then evaporated and purified with column chromatography (EtOAc/Hexanes=l/10) to give 1.2 g product. Yield for two steps 60%. LCMS (ESI) calc'd for Ci6H7ClF3IN202 [M+H]+: 478.7, found: 479.0.
Step 3. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)- 6-formyl-lH- indazol-3-yl)-3-fluorobenzoate (AI-4). To a microwave tube was added l-(2-chloro-6- (trifluoromethyl)benzoyl)-3-iodo-lH-indazole-6-carbaldehyde (AI-2) (400 mg, 0.83 mmol), 2-fluoro-4-(methoxycarbonyl)phenylboronic acid (AI-3) (320 mg, 1.66 mmol), Pd(OAc)2 trimer (28 mg, 0.125 mmol), K3PO4(600 mg, 2.5 mmol), s-Phos (100 mg, 0.25 mmol), THF (8 mL), H20 (2 mL). The mixture was micro waved at 110°C for 2 hours and the compound was submitted for Prep-HPLC. 178 mg product was obtained with a yield of 40%. LCMS (ESI) calc'd for C24Hi3ClF4N204 [M+H]+: 505, found: 505.
Step 4. Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (2,5- dioxoimidazolidin-4-yl)-lH-indazol-3-yl)-3-fluorobenzoate (AI-5). To a microwave tube was added methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- formyl-lH-indazol-3-yl)-3- fluorobenzoate (AI-4) (100 mg, 0.20 mmol), NaCN (19.6 mg, 0.40 mmol), ( H4)2C03 (76.8 mg, 0.8 mmol), ethanol (0.8 mL) and H20 (0.8 mL). The mixture was micro waved under Argon at 110°C for 2 hours. The mixture was filtered and submitted for next step without further purification. LCMS (ESI) calc'd for C26Hi5ClF4N405 [M+H]+: 575, found: 575.
Step 5. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(2,5- dioxoimidazolidin-4-yl)-lH-indazol-3-yl)-3-fluorobenzoic acid (27A). To a stirred above solution of methyl 4-(l-(2- chloro-6-(trifluoromethyl)benzoyl)-6-(2,5- dioxoimidazolidin-4- yl)-lH-indazol-3-yl)-3-fluorobenzoate (AI-5) was added LiOHH20 (60 mg, 1.5 mmol) and H20 (0.1 mL). The solution was stirred overnight. The solution was adjusted to PH 3.0 using 1 N HC1. The reaction mixture was extracted with THF (1 mL x 3). The combined organic layer was added 0.5 mL MeOH and submitted for Prep-HPLC. Prep-HPLC (H20/ACN, 0.05% TFA) gave 5 mg product, yield for two steps 4.5%. LCMS (ESI) calc'd for
C25H13CIF4N4O5 [M+H]+: 561, found: 561 ; 1HNMR (400MHz, DMSO-d6) δ 13.60 (s, 1H), 10.99 (s, 1H), 8.68 (s, 1H), 8.66 (s, 1H), 8.56-7.98 (m, 3H), 7.93-7.88 (m, 3H), 7.84-7.75 (d, lH),7.73-7.60 (d, 1H).
Example 28A: Preparation of 4-(5-(tert-butoxycarbonylamino)-l-(2-chloro-6- (trifluoromethyl)benzoyl)-lH-pyrrolo[2,3-c]pyridin-3-yl)-3-fluorobenzoic acid (28A).
Scheme AJ
Figure imgf000138_0001
28A
Step 1. 3-bromo-5-chloro-lH-pyrrolo[2,3-c]pyridine (AJ-2). To a stirred solution of 5- chloro-lH-pyrrolo[2,3-c]pyridine (AJ-1) (2.0 g, 13.3 mmol) in anhydrous DMF (80 mL) at r.t.was added Br2 (0.68 mL, 13.3 mmol) dropwise. The solution was stirred at r.t for one hour. The solution was quenched with 10% Na2S203 solution (100 mL) and diluted with H20 (400 mL). The aqueous layer was extracted with EtOAc (100 mL x 4) and combined organic layer was washed with H20 (50 mL x 3) and brine (50 mL x 3) and dried over anhydrous Na2S04. The solution was evaporated and dried over vacuo and 2.9 g product (95%) was collected. LCMS (ESI) calc'd for C7H4BrClN2 [M+H]+: 231, found: 231.
Step 2. (3-bromo-5-chloro-lH-pyrrolo[2,3-c]pyridin-l-yl)(2-chloro-6-(trifluoro- methyl)phenyl)methanone (AJ-3). To a stirred solution of 3-bromo-5-chloro- 1H- pyrrolo[2,3-c]pyridine (AJ-2) (2.9 g, 12.6 mmol) in anhydrous DMF (100 mL) was added 2- chloro-6-(trifluoromethyl)benzoyl chloride (4.6 g, 18.9 mmol) and NaH (60%) (1 g, 25.2 mmol). The solution was stirred at r.t for 2 hours. The solution was quenched with H20 (400 mL). The suspension was extracted with EtOAc (150 mL x 3). The combined organic layer was washed with H20 (100 mL x 2) and brine (100 mL x 2) and dried over anhydrous Na2S04. The solution was evaporated and dried over vacuo and 5.7 g product was obtained. LCMS (ESI) calc'd for Ci5H6BrCl2F3N20 [M+H]+: 437, found: 437.
Step 3. methyl 4-(5-chloro-l-(2-chloro-6-(trifluoromethyl)benzoyl)-lH-pyrrolo [2,3- c]pyridin-3-yl)-3-fluorobenzoate (AJ-4). To a microwave tube was added (3- bromo-5- chloro- 1 H-pyrrolo [2,3 -c]pyridin- 1 -yl)(2-chloro-6-(trifluoromethyl)phenyl)-methanone (A J-3) (650 mg, 1.5 mmol), 2-fluoro-4-(methoxycarbonyl)phenylboronic acid (450 mg, 2.25 mmol), Pd(dppf)Cl2 (73 mg, 0.10 mmol), KOAc (300 mg, 3.0 mmol) and dioxane (12 mL). The mixture was microwaved at 110 °C for three hours and filtered through celite. The solvent was evaporated, the cude product was purified with columm chromatography (DCM/Hexanes: 1/1) to give 450 mg product (yield 60%). LCMS (ESI) calc'd for C23Hi2Cl2F4N203 [M+H]+: 511, found: 511.
Step 4. 4-(5-(tert-butoxycarbonylamino)-l-(2-chloro-6-(trifluoromethyl)benzoyl)- 1H- pyrrolo[2,3-c]pyridin-3-yl)-3-fluorobenzoic acid (28A). To a microwave tube was added 4-(5 -chloro- 1 -(2-chloro-6-(trifluoromethyl)benzoyl)- 1 H-pyrrolo [2, 3 -c] pyridin-3 -yl)-3 - fluorobenzoic acid (AJ-4) (50 mg, O. lmmol), Boc H2 (35 mg, 0.3mmol), NaOH (20 mg, 0.5 mmol), Pd(OAc)2 (4 mg, 0.02 mmol), xant-Phos( 20 mg, 0.04 mmol), dioxane (1 mL) and H20 (0.05 mL). The mixture was microwaved at 90 °C for 1 hr. After filtration and evaporation, the crude product was submitted for prep-HPLC purification, which gave 5 mg title product (yield 17%). LCMS (ESI) calc'd for C27H2oClF4N305 [M+H]+: 578, found: 578; 1HNMR (400MHz, DMSO-d6) δ 13.40 (bs, 1H), 10.00 (s, 1H), 9.37(s, 1H), 8.02-8.09 (m, 3H), 7.88-7.93 (m, 3H), 7.81-7.84 (d, 1H), 7.70-7.74 (m, 1H), 1.48 (s, 9H).
Example 29A: Preparation of 4-(6-(tert-butoxycarbonylamino)-l-(2-chloro-6- (trifluoromethyl)benzoyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoic acid (29A) and 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(methylamino)-lH- pyrazolo[4,3- b]pyridin-3-yl)-3-fluorobenzoic acid (29B)
Scheme AK
Figure imgf000140_0001
Step 1. Preparation of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-(2-fluoro-4- (methoxycarbonyl)phenyl) -lH-pyrazolo[4,3-b]pyridine-6-carboxylic acid (AK-2). To a mixture of 3-bromo-l-(2-chloro-6-(trifluoromethyl)benzoyl)-lH-pyrazolo[4,3-b] pyridine-6- carboxylic acid (AK-1) (224 mg, 0.5 mmol), boric acid (128 mg, 0.60 mmol), PdCl2(dppf)2 (48 mg, 0.05 mmol) and KF (90 mg, 1.5 mmol) was added dioxane (25 mL) and H20 (0.5 mL), and the mixture was heated at 90 °C under argon for 16 h. The mixture was cooled down, diluted with CH2C12 (80 mL). The organic layer was separated, washed with brine, dried over Na2S04, and concentrated. The residue was purified by Prep-TLC (EtOAc) to afford 220 mg (85 %) of the title compound as a white solid. LCMS (ESI) calc'd for C23Hi2ClF4N305 [M+H]+: 522.1, found: 522.1.
Step 2. Preparation of methyl 4-(6-(tert-butoxycarbonylamino)-l-(2-chloro-6- (trifluoromethyl)benzoyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoate (AK-3). To a mixture of l-(2-chloro-6-(trifluoromethyl)benzoyl)-3-(2-fluoro-4-
(methoxycarbonyl)phenyl)-lH-pyrazolo[4,3-b]pyridine-6-carboxylic acid (AK-2) (50 mg, 0.1 mmol), DPP A (41 mg, 0.15 mmol), DIPEA (38 mg, 0.3 mmol) and t-BuOH (10 mL) was heated at 90 °C under argon for 16h. The mixture was cooled down, diluted with CH2C12 (80 mL). The organic layer was separated, washed with brine, dried over Na2S04, concentrated. The residue was purified by Prep-TLC (EtOAc) to afford 38 mg (64 %) of the title compound as a white solid. LCMS (ESI) calc'd for C27H21C1F4N405 [M+H]+: 593.1 , found: 593.1.
Step 3. Preparation of 4-(6-(tert-butoxycarbonylamino)-l-(2-chloro-6- (trifluoromethyl)benzoyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoic acid (29A).
To a stirred solution of methyl 4-(6-(tert-butoxycarbonylamino)-l-(2-chloro- 6- (trifluoromethyl)benzoyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoate (AK-3) (80 mg, 0.14 mmol) was added THF (5.0 mL), H20 (1.0 mL) and LiOHH20 (57 mg, 1.4 mmol) and the solution was stirred at r.t. overnight. The solution was adjusted to PH 4.0 using IN HC1 and poured into THF (30 mL) and washed with brine (20 mL). The organic layer was dried over Na2S04, the solvent was evaporated and submitted for Prep-HPLC to get 60 mg title product with a yield of 73%. LCMS (ESI) calc'd for C26Hi9ClF4N405 [M+H]+: 579.1, found: 579.1; 1HNMR (500MHz, DMSO-d6) δ 13.75 (bs, 1H), 10.35 (s, 1H), 9.25 (s, 1H), 8.81-8.81 (d, 1H), 8.34 (t, 1H), 7.98 (m, 5H), 1.55 (s, 9H).
Step 4. Preparation of methyl 4-(6-(tert-butoxycarbonyl(methyl)amino)-l-(2- chloro-6- (trifluoromethyl)benzoyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoate (AK-4). To a solution of methyl 4-(6-(tert-butoxycarbonylamino)-l-(2-chloro-6- (trifluoromethyl)benzoyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoate (AK-3) (300 mg, 0.5 mmol) in DMF (10 mL) was added NaH (81 mg, 2 mmol, 60 %) portions over 5 min then stirred at 0 °C for 1 h. And then CH3I (108 mg, 0.76 mmol) in THF (1 mL) was dropwise by syring, the reaction mixture was stirred at 0°C for 12 h, quenched with ice-water, and the aqueous layer was extracted with CH2C12 (50 mL x 3). The combined organics were washed with H20, brine, dried over Na2S04, and concentrated. The residue was purified by silica gel column chromatography (Pentane/EtOAc=10/l) to afford 280 mg (91 %) of the title compound. LCMS (ESI) calc'd for C28H23C1F4N405 [M+H]+: 607, found: 607.
Step 5. Preparation of 4-(6-(tert-butoxycarbonyl(methyl)amino)-l-(2-chloro-6- (trifluoromethyl)benzoyl)- lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoic acid (AK-5).
To a stirred solution of methyl 4-(6-(tert-butoxycarbonyl(methyl)amino)-l- (2-chloro-6- (trifluoromethyl)benzoyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoate (AK-4) (85 mg, 0.14 mmol) was added THF (5.0 mL), H20 (1.0 mL) and LiOHH20 (57 mg, 1.4 mmol) and the solution was stirred at r.t. overnight. The solution was adjusted to pH 4.0 using IN HCl and poured into THF (30 mL), washed with brine (20 mL). The organic layer was dried over Na2S04, the organic solvent was evaporated and submitted for prep-HPLC. 61 mg product was collected. Yield: 71%. LCMS (ESI) calc'd for C27H2iClF4N405 [M+H]+: 593.1, found: 593.1.
Step 6. Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(methyl- amino)-lH- pyrazolo [4,3-b]pyridin-3-yl)-3-fluorobenzoic acid (28B). To a stirred solution of 4-(6- (tert-butoxycarbonyl(methyl)amino)- 1 -(2-chloro-6-(trifluoromethyl) benzoyl)- 1 H- pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoic acid (AK-5) (100 mg, 0.17 mmol) was added THF (5.0 mL), and 6 N HCl (4 mL) and the solution was stirred at room temperature for 4 h. The solution was adjusted to pH 4.0 using 2N NaOH and poured into THF (30 mL), washed with brine (20 mL). The organic layer was dried over Na2S04, the organic solvent was evaporated and submitted for prep-HPLC. 20 mg product was collected. Yield: 25%. LCMS (ESI) calc'd for C22Hi3ClF4N403 [M+H]+: 493.1, found: 493.1; 1HNMR (500MHz, DMSO- d6) δ 13.74 (bs, 1H), 8.328 (s, 1H), 8.268 (t, 1H), 8.01 (d, 1H), 7.94 (d, 1H), 7.89 (m, 1H), 7.84 (d, 1H), 7.74 (m, 1H), 7.19 (d, 1H), 2.87 (d, 1H).
Example 30A: Preparation of 4-(5-acetamido-l-(2-chloro-6-(trifluoromethyl)- benzoyl)- lH-pyrrolo[2,3-c]pyridin-3-yl)-3-fluorobenzoic acid (30A) and 4-(l-(2- chloro-6- (trifluoromethyl)benzoyl)-5-(methylamino)-lH-pyrrolo[2,3-c]pyridin-3-yl)-3- fluorobenzoic acid (30B)
Scheme AL
Figure imgf000143_0001
Step 1. 4-(5-(tert-butoxycarbonylamino)-l-(2-chloro-6-(trifluoromethyl)benzoyl) -1H- pyrrolo[2,3-c]pyridin-3-yl)-3-fluorobenzoic acid (AL-2). To a stirred solution of methyl 4- (5 -chloro- 1 -(2-chloro-6-(trifluoromethyl)benzoyl)- 1 H-pyrrolo [2, 3 -c] pyridin-3 -yl)-3 - fluorobenzoate (AL-1) (550 mg, 1.1 mmol) in dioxane (14 mL) and water (0.5 mL) was added Boc H2 (389 mg, 3.3 mmol), Pd(OAc)2 (50 mg, 0.22 mmol), xant-phos (256 mg, 0.44 mmol) and NaOH (222 mg, 5.5 mmol). The mixture was stirred under Ar at 90 °C for 1 h in the microwave reactor. The mixture was filtered and evaporated, MeOH (5 mL) was added. The resultant solution was submitted for Prep-HPLC under 0.01% TFA, 300 mg solid was obtained, yield: 47%. LCMS (ESI) calc'd for C^H^Ch^NsOs [M+H]+: 578, found: 578.
Step 2. 4-(5-amino-l-(2-chloro-6-(trifluoromethyl)benzoyl)-lH-pyrrolo[2,3-c] pyridin-3- yl)-3-fluorobenzoic acid (AL-3). To a stirred solution of 4-(5-(tert-butoxy carbonylamino)- 1 -(2-chloro-6-(trifluoromethyl)benzoyl)- 1 H-pyrrolo [2, 3 -c]pyridin-3 -yl)-3 -fluorobenzoic acid (AL-2) (40 mg, 0.07 mmol) in DCM (0.5 mL) and TFA (0.5 mL) was stirred under Ar at rt for 5 h. The mixture was evaporated, the crude product was used to the next step directly. LCMS (ESI) calc'd for C22Hi2ClF4N303 [M+H]+: 478, found: 478.
Step 3. 4-(5-acetamido-l-(2-chloro-6-(trifluoromethyl)benzoyl)-lH-pyrrolo[2,3-c] pyridin-3-yl)-3-fluorobenzoic acid (30A). To a stirred solution of 4-(5-amino-l-(2- chloro- 6-(trifluoromethyl)benzoyl)-lH-pyrrolo[2,3-c]pyridin-3-yl)-3-fluorobenzoic acid (AL-3) (30 mg, 0.06 mmol) in Ac20 (1 mL) was added AcOH (0.2 mL) was stirred under Ar at 100 °C for 1 h. Evaporated the solvent, then added H20 (1 mL), mixture was stirred at rt for 2 h. The mixture was purified by prep-HPLC under 0.01% TFA twice to give 10 mg product (yield: 24.5%). LCMS (ESI) calc'd for C24Hi4ClF4N304 [M+H]+: 520, found: 520; 1HNMR (400MHz, DMSO-de) δ 13.37 (bs, 1H), 10.78 (s, 1H), 9.43 (s, 1H), 8.43 (s, 1H), 8.04 (dd, 2H), 7.93-7.87 (m, 3H), 7.81 (d, 1H), 7.71 (t, 1H), 2.12 (s, 3H).
Step 4. Methyl 4-(5-(tert-butoxycarbonyl(methyl)amino)-l-(2-chloro-6-(trifluoro- methyl)benzoyl)-lH-pyrrolo[2,3-c]pyridin-3-yl)-3-fluorobenzoate (AL-4). To a stirred solution of 4-(5-acetamido-l-(2-chloro-6-(trifluoromethyl)benzoyl)-lH- pyrrolo[2,3- c]pyridin-3-yl)-3-fluorobenzoic acid (AL-2) (100 mg, 0.17 mmol) in anhydrous DMF (5 mL) at r.t. was added NaH (42 mg, 60%, 1 mmol) and then Mel (98 mg, 0.69 mmol). The solution was stirred at r.t for 3 hours. Ethyl Acetate (200 mL) was added and the solution was washed with brine (70 mL x 2). The solution was evaporated and 120 mg crude product was got and it was used in the next step without further purification. LCMS (ESI) calc'd for C29H24C1F4N305 [M+H]+: 606, found: 606.
Step 5. Methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-5-(methylamino)-lH pyrrolo[2,3-c]pyridin-3-yl)-3-fluorobenzoate (AL-5). To a stirred solution of methyl 4-(5- (tert-butoxycarbonyl(methyl)amino)- 1 -(2-chloro-6-(trifluoromethyl)- benzoyl)- 1 H- pyrrolo[2,3-c]pyridin-3-yl)-3-fluorobenzoate (AL-4) (120 mg, as generated from step 4) in DCM (4 mL) was added TFA (2 mL) under N2. The solution was stirred at r.t. for 3 hours and evaporated. 70 mg crude product was collected. It was used for next step without further purification. LCMS (ESI) calc'd for C24Hi6ClF4N303 [M+H]+: 506, found: 506.
Step 6. 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-5-(methylamino)-lH-pyrrolo [2,3- c]pyridin-3-yl)-3-fluorobenzoic acid (30B). To a stirred solution of methyl 4- (l-(2-chloro- 6-(trifluoromethyl)benzoyl)-5-(methylamino)-lH-pyrrolo[2,3-c]pyridin-3-yl)-3- fluorobenzoate (AL-5) (71mg, as generated from step 5) in THF (2 mL) was added H20 (2 mL) and LiOHH20 (58 mg, 1.4 mmol). The solution was stirred at r.t for overnight and adjusted to PH~2 using IN HC1. The upper layer was evaporated and submitted for Prep- HPLC (CH3CN/H20), which gave 52 mg product. Yield for three steps 60%. LCMS (ESI) calc'd for C23Hi4ClF4N303 [M+H]+: 492, found: 492.
Biological Assays
The compounds of the invention inhibit RORgammaT activity. Activation of RORgammaT activity can be measured using e.g. biochemical TR-FRET assay. In such an assay, interaction of cofactor-derived peptides with human RORgammaT -Ligand Binding Domain (LBD) can be measured. The TR-FRET technique is a sensitive biochemical proximity assay that will give information concerning the interaction of a ligand with the LBD, in the presence of cofactor derived peptides (Zhou et al, Methods 25:54-61, 2001).
To identify novel antagonists of RORgammaT, an assay was developed which employs the interaction of RORgammaT with its co-activator peptide SRC 1 2. This peptide mimics the recruitment of co-activators to RORgammaT through its interaction with the LXXLL (eg R box) motifs (Xie et al, J. Immunol. 175: 3800-09, 2005; Kurebayashi et al, Biochem. Biophys. Res. Commun. 315: 919-27, 2004; Jin et al, Mol. Endocrinology 24:923-29, 2010). The RORy-Ligand Binding Domain TR-FRET Assay was run according to the following protocol.
HIS-tagged RORy-LBD protein was expressed in SF9 cells using a baculovirus expression system. The RORy-LBD protein was purified by glutathione sepharose chromatography. Separately, SF9 cells not expressing any recombinant protein were lysed and the lysate was added to the purified RORy-LBD at 0.25 μΐ lysate (from 10,000 SF9 cells)/nM purified protein. The mixture was then diluted in assay buffer (50 mM Tris pH 7.0, 50 mM KC1, 1 mM EDTA, 0.1 mM DTT) to obtain RORy-LBD final concentration of 3 nM in 384-well assay plate.
Compounds to be tested were injected to the assay plate using Acoustic Droplet Ejection technology by Echo 550 liquid handler (Labcyte, CA).
A stock of biotinylated-LXXLL peptide from coactivator SRC1 (Biotin- CPSSHSSLTERHKILHRLLQEGSPS) was prepared in assay buffer and added to each well (100 nM final concentration). A solution of Europium tagged anti-HIS antibody (1.25 nM final concentration) and APC conjugated streptavidin (8 nM final concentration) were also added to each well. The final assay mixture was incubated for overnight at 4°C, and the fluorescence signal was measured on an Envision plate reader: (Excitation filter = 340 nm; APC emission = 665 nm; Europium emission = 615 nm; dichroic mirror = D400/D630; delay time = 100 μβ, integration time = 200 μ8).ΙΟ50 values for test compounds were calculated from the quotient of the fluorescence signal at 665 nm divided by the fluorescence signal at 615 nm.
BIOLOGICAL DATA
The following table tabulates the biological data disclosed for the instant invention.
Figure imgf000146_0001
4D 130
4F 23
4G 1060
4H 4321
5A 255
5B 153
5C 314
5D 493
5E 64
5F 48
5G 1502
5H 491
6A 85
6B 25
6C 22
6D 1511
6E 224
7A 69
7B 317
7C 87
7D 14
8A 143
8B 283
8C 218
8D 57
9A 63
9B 14
11A 48
12A 105
13A 20
14A 137
15A 241
16A 21
17A 12
18A 24
18B 26 60Z ao ε
9 νοε
£1 mz
161 Y6Z
281 V8Z
£ζ YLZ
9L Y9Z
OOOOK ¾εζ
OOOOK i£Z
OOOOK l£Z
OOOOK H£Z
OOOOK Ό£Ζ
OOOOK d£Z
OOOOK £Ζ
OOOOK Q£Z
OOOOK 3£Z
OOOOK Q£Z
OOOOK Y£Z
16C9 iZZ
IZ IZZ
L£ HZZ
9ζ£ ΌΖΖ
6£ dZZ
IfrH ΉΖΖ
ζζ \ QZZ
OOOOK JZZ
909S QZZ
P9L YZZ
S t JIZ
LI mz
ς YIZ
OZl YOZ
888 V61
9tl
I£l080/ZT0ZN3/I3d /.Z£9Z0/H0Z OAV

Claims

A compound according to Formula I
Figure imgf000149_0001
I or a pharmaceutically acceptable salt or solvate thereof wherein,
Y is CH, CRa, or N;
n = 0, 1, 2, 3 or 4;
A4 is CR4 or N,
A5 is CR5 or N,
A6 is CR6 or N,
A7 is CR7 or N,
with the proviso that no more than one or two of A4- A7 can be N;
Ra is (Ci-4)alkyl or (C3-7)cycloalkyl;
R1 is
(i) (C3-i2)carbocyclyl(Co-4)alkyl;
(ii) a 4- to 12-membered heterocyclyl(Co-4)alkyl, or
(iii) (Ci-4)alkoxy,
each optionally substituted with one, two, three, four or five R8 ;
R2 is hydroxycarbonyl, hydroxycarbonyl(Ci-io)alkyl, (Ci-io)alkylsulfoxyaminocarbonyl, or carbamoyl;
R3 is hydrogen, halogen, cyano, nitro, hydroxy, (Ci-4)alkylcarbonyloxy, (Ci-4) alkylsulfonylamino, (C1-4) alkylcarbonylamino, (C0-4) alkylamino, (Ci-4)alkyl, or (Ci_ 4)alkoxy, wherein (Ci-4)alkyl and (Ci-4)alkoxy are optionally substituted with one or more halogen;
R4-R7 independently are hydrogen, halogen, amino, cyano, hydroxy, (Ci_3)alkoxy, (Ci_ 4)alkyl, (Co-io)alkylaminocarbonyl, (Co-6)alkyoxycarbonylamino, (di)(Ci_
6)alkylaminocarbonyl, (Ci-6)alkylcarbonylamino, (Ci-4)alkylamino, amino(Ci-4)alkyl or formaldehyde, wherein (Ci-3)alkoxy, (Ci-4)alkyl, (Co-io)alkyl)aminocarbonyl, (di)(Ci_ 6)alkylaminocarbonyl, (Ci-4)alkylamino and amino(Ci-4)alkyl are optionally substituted with one or more halogen, hydroxyl or (Ci-3)alkoxy; or a group having the formula ° , optionally substituted with one or more of the following: (Ci_ io)alkyl, halogen, amino, cyano, hydroxy, (Ci_3)alkoxy, and wherein m is 1, 2, 3, or 4; R6 is, additionally,
(i) (C3-7)cycloalkyl or (C3-5)heterocycloalkyl, both optionally substituted with one or more groups selected from halogen, amino, amino(Ci-4)alkyl, cyano, nitro, hydroxyl, oxo (=0), H2NC(0), (C1.3)alkoxycarbonyl, (di)(Ci_
6)alkylaminocarbonyl, (Ci-4)alkyl or (Ci-3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci_3)alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
(ii) (C2-9)heteroaryl, optionally substituted with one or more groups selected from halogen, amino, amino(Ci-4)alkyl, cyano, nitro, hydroxyl, oxo (=0), H2NC(0), (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl or (Ci-3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci_ 3)alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
(iii) (C6-i4)aryl, optionally substituted with one or more groups selected from halogen, amino, amino(Ci-4)alkyl, cyano, nitro, hydroxyl, oxo (=0), H2NC(0), (Ci_ 3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl or (Ci-3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci_ 3)alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
(iv) (C3-5)heterocycloalkylcarbonyl, optionally substituted with one or more groups selected from halogen, amino, amino(Ci-4)alkyl, cyano, nitro, hydroxyl, oxo (=0), H2NC(0), (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl or (Ci_ 3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci-3)alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
(v) (C3-5)heterocycloalkylamino, optionally substituted with one or more groups selected from halogen, amino, amino(Ci-4)alkyl, cyano, nitro, hydroxyl, oxo (=0), H2NC(0), (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl or (Ci_ 3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci-3)alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
(vi) (C3-5)cycloalkylaminocarbonyl, optionally substituted with one or more groups selected from halogen, amino, amino(Ci-4)alkyl, cyano, nitro, hydroxyl, oxo (=0), H2NC(0), (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl or (Ci_ 3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci-3)alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
(vii) (C3-5)cycloalkylcarbonylamino, optionally substituted with one or more groups selected from halogen, amino, amino(Ci-4)alkyl, cyano, nitro, hydroxyl, oxo (=0), H2NC(0), (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl or (Ci_ 3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci-3)alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
(viii) (C3-5)cycloalkyl(Ci-4)alkyl, optionally substituted with one or more groups
selected from halogen, amino, amino(Ci-4)alkyl, cyano, nitro, hydroxyl, oxo (=0), H2NC(0), (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl or (Ci_ 3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci-3)alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
(ix) (C3-5)cycloalkylamino, optionally substituted with one or more groups selected from halogen, amino, amino(Ci-4)alkyl, cyano, nitro, hydroxyl, oxo (=0), H2NC(0), (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl or (Ci_ 3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci-3)alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
(x) (C3-5)cycloalkylcarbonyl, optionally substituted with one or more groups selected from halogen, amino, amino(Ci-4)alkyl, cyano, nitro, hydroxyl, oxo (=0), H2NC(0), (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl or (Ci_ 3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci-3)alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
(xi) (C2-9)heteroaryl(Ci-4)alkyl, optionally substituted with one or more groups
selected from halogen, amino, amino(Ci-4)alkyl, cyano, nitro, hydroxyl, oxo (=0), H2NC(0), (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl or (Ci_ 3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci-3)alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
(xii) (C2-9)heteroarylcarbonyl, optionally substituted with one or more groups selected from halogen, amino, amino(Ci-4)alkyl, cyano, nitro, hydroxyl, oxo (=0), H2NC(0), (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl or (Ci_ 3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci-3)alkoxy are optionally substituted with one or more halogen, amino or hydroxyl;
(xiii)
Figure imgf000152_0001
(xiv) (C2-4)alkynyl, optionally substituted with one or more (Ci-4)alkyl, which (Ci_ 4)alkyl may be substituted with hydroxyl or amino; or
(xv) (Ci-6)alkoxycarbonylamino,
(Ci-6)alkylcarbonylamino,
(Ci-6)alkylsulfonylamino(Co-4)alkyl,
(Ci-6)alkylaminocarbonylamino,
(Ci-6)alkyoxycarbonylamino(Co-4)alkyl,
Hydroxycarbonyl(Ci-4)alkylamino, Hydroxycarbonyl, or
(Ci-6)alkylamino,
each optionally substituted with one or more (Ci-4)alkyl, hydroxyl or amino; R8 is halogen, cyano, amino, nitro, hydroxy, oxo(=0), H2NC(0)-, (Ci-3)alkoxycarbonyl, (di)(Ci-6)alkylaminocarbonyl, (Ci-4)alkyl, (C3-7)cycloalkyl, (C3-5)heterocycloalkyl, (Ci_ 4)alkenyl, (C3-6)cycloalkoxy or (Ci-3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (di)(Ci_ 6)alkylaminocarbonyl, (Ci-4)alkyl and (Ci_3)alkoxy are optionally substituted with one, two or three halogens; and
x is 0, 1, 2, 3, 4 or 5.
The compound of claim 1 having Formula la
Figure imgf000153_0001
la and a pharmaceutically acceptable salt or solvate thereof.
The compound of claim 1 having Formula lb
Figure imgf000154_0001
and a pharmaceutically acceptable salt or solvate thereof. The compound of claim 3, wherein Y is N.
The compound of claim 3 having Formula Ic
Figure imgf000154_0002
and a pharmaceutically acceptable salt or solvate thereof.
The compound of claim 5 having Formula Id
Figure imgf000155_0001
and a pharmaceutically acceptable salt or solvate thereof. 7. The compound of claim 6, wherein Y is N.
8. The compound of claim 2 having Formula Ie
Figure imgf000155_0002
Ie
and a pharmaceutically acceptable salt or solvate thereof.
9. The compound of claim 8 having Formula If
Figure imgf000156_0001
and a pharmaceutically acceptable salt or solvate thereof.
10. The compound of claim 9 having Formula Ig
Figure imgf000156_0002
Ig
and a pharmaceutically acceptable salt or solvate thereof.
The compound of claim 10 having Formula Ih
Figure imgf000157_0001
and a pharmaceutically acceptable salt or solvate thereof.
12. The compound of claim 1, wherein A4, A5, A6, A7 are selected from the group
consisting of: (i) CR4, CR5, CR6, CR7; (ii) N, CR5, CR6, CR7; and (iii) CR4, N, CR6, CR7.
13. The compound of claim 12, wherein A4, A5, A6, A7 is (i) CR4, CR5, CR6, CR7, or (ii) N, CR5, CR6, CR7; and Y is N.
14. The compound of claim 1, wherein R1 is
(i) (C3-7)cycloalkyl or (C3-5)heterocycloalkyl, both optionally substituted with one or more R ;
(ii) (C2-9)heteroaryl(Co-4)alkyl, optionally substituted with one or more R8; or
(iii) (C6-i4)aryl(Co-4)alkyl, optionally substituted with one or more R8.
15. The compound of claim 14, wherein R1 is (i) (C2-9)heteroaryl, or (ii) (C6-i4)aryl,
optionally substituted with one, two, three, four or five R8.
16. The compound of claim 15, wherein R1 is (C6-i4)aryl, optionally substituted with one or two R8.
17. The compound of claim 16, wherein R1 is phenyl, optionally substituted with one or two
18. The compound of claim 17, wherein R2 is C(0)OH.
19. The compound of claim 1, wherein R6 is
Figure imgf000158_0001
20. A compound according to claim 1 selected from:
(E)-4-( 1 -(2-chloro-6-(prop- 1 -enyl)benzoyl) - 1 H-pyrazolo [4,3 -b]pyridin-3 -yl)-3 - fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-4- formyl- 1 H-indazol-3 -yl)benzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)- 1 H- pyrazolo [4,3 -b]pyridin-3 -yl)- 1 H-indazole- 7-carboxylic acid;
4-(l-(2-chloro-6-cyclopropoxybenzoyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)benzoic acid; 3 -fluoro-4-( 1 -(2-phenylpropanoyl)- 1 H-indazol-3 -yl)benzoic acid;
3-fluoro-4-[l-(methoxyacetyl)-lH-indazol-3-yl]benzoic acid;
3 -fluoro-4- [ 1 -(pyridin-3 -ylcarbonyl)- 1 Hindazol-3 -yljbenzoic acid;
3 - fluoro-4- { 1 -[(2-oxopyrrolidin- 1 -yl)acetyl]- lHindazol-3 -yl }benzoic acid;
3 -fluoro-4- [ 1 -(naphthalen- 1 -ylcarbonyl)- 1 Hindazol-3 -yljbenzoic acid;
3 -fluoro-4- { 1 -[( 1 -methyl- 1 H-indol-2-yl)carbonyl] - 1 Hindazol-3 -yl } benzoic acid;
4- { 1 - [(2-bromo-3 -methylphenyl)carbonyl] - 1 H-indazol-3 -yl } -3 -fluorobenzoic acid;
4- [ 1 -(2, 3 -dihydro- 1 H-inden-4-ylcarbonyl)- 1 Hindazol-3 -yl] -3 -fluorobenzoic acid;
4-( 1 - { [3 -(tertbutoxycarbonyl)-3 -azabicyclo [3.1.0]hex-6-yl] carbonyl } - 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4- [ 1 -(2, 3 -dihydro- 1 -benzofuran-7-ylcarbonyl)- 1 Hindazol-3 -yl] -3 -fluorobenzoic acid;
4- [ 1 -( 1 -benzofuran-7-ylcarbonyl)- 1 Hindazol-3 -yl] -3 -fluorobenzoic acid;
4- { 1 - [(2-bromo-3 -chlorophenyl)carbonyl] - 1 H-indazol-3 -yl } -3 -fluorobenzoic acid;
3 - fluoro-4-( 1 -(tetrahydrofuran-2-carbonyl)- 1 H-indazol-3 -yl)benzoic acid;
4- (l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (morpholine-4-carbonyl)-l H-indazol-3 - yl)benzoic acid;
4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-((2S,6R)-2,6-dimethylmorpholine-4- carbonyl)-l H-indazol-3 -yl)-3 -fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(4-oxopiperidine- 1 -carbonyl)- 1 H-indazol-3 - yl)-3 -fluorobenzoic acid;
2-acetamido-4-(l-(2-chloro-6-(trifluoromethyl) benzoyl)- IH-indazol -3-yl)benzoic acid; 4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl)-2-
(methylsulfonamido)benzoic acid; -( 1 -(2-chloro-6-(trifluoromethyl) benzoyl)-6- (3 -hydroxy azetidin- 1 -yl)- 1 H-indazol-3 - yl)benzoic acid;
-(6-(azetidin- 1 -yl)- 1 -(2-chloro-6-(trifluoromethyl)benzoyl)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(cyclopropylamino)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(oxetan-3 -ylamino)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(3 -hydroxypyrrolidin- 1 -yl)- 1 H-indazol-3 - yl)-3 -fluorobenzoic acid;
-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-morpholino- 1 H-indazol-3 -yl)benzoic acid;-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (methoxycarbonylamino)-lH-indazol-3-yl)-
3 -fluorobenzoic acid;
-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(N- methylacetamido)-lH-indazol-3-yl)-3- fluorobenzoic acid;
-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(cyclopropanecarboxamido)-lH-indazol-3- yl)-3 -fluorobenzoic acid;
-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(methylsulfonamido)- lH-indazol-3 -yl)-3 - fluorobenzoic acid;
-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(3 -methylureido)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
-(6-acetamido- 1 -(2-chloro-6-(trifluoromethyl)benzoyl)- 1 H-indazol-3 -yl)-3 -fluorobenzoic acid;
-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(N-methylmethylsulfonamido)-lH-indazol-
3 -yl)-3 -fluorobenzoic acid;
-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-( 1 , 3 -dimethy lureido)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(2-oxo- imidazolidin- 1 -yl)- 1 H-indazol-3 -yl)- 3 -fluorobenzoic acid;
-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6- (2-oxoazetidin- 1 -yl)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid; 4-(6-(2-carboxyethylamino)- 1 -(2-chloro-6- (trifluoromethyl)benzoyl)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(3 -methyl-2-oxoimidazolidin- 1 -yl)- 1 H- indazol-3 -yl)-3 -fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(2-oxopyrrolidin- 1 -yl)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(mor pholine-4-carbonyl)-l H-indazol-3 - yl)benzoic acid;
3- (4-carboxyphenyl)-l-(2-chloro-6-(trifluoromethyl) benzoyl)- lH-indazole-6-carboxylic acid;
4- ( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(cyclopropylcarbamoyl)- 1 H-pyrazolo [4,3- b]pyridin-3 -yl)-3 -fluorobenzoic acid;
4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(cyclopropyl(methyl)carbamoyl)-lH- pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6- (cyclo
Propyl (hydroxy)methyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl)-3 -fluorobenzoic acid;
4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(cyclop
ropane-carbonyl)-lH-pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoic acid;
4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(hydroxy(oxazol-2-yl)methyl)-lH- pyrazolo[4,3-b]pyridine-3-yl)-3-fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(oxazole-2-carbonyl)- 1 H-pyrazolo [4,3- b]pyridin-3 -yl)-3 -fluorobenzoic acid;
sodium 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(5-methyloxazol-2-yl)-lH-indazol-3 yl)benzoate;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(5 -methyloxazol-2-yl)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(3 -hydroxyprop- 1 -ynyl)- 1 H-indazol-3 - yl)benzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(3 -hydroxybut- 1 -ynyl)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4-(6-(3 -aminoprop- 1 -ynyl)- 1 -(2-chloro-6-(trifluo romethyl)benzoyl)- 1 H-indazol-3 -yl)-3 fluorobenzoic acid; 4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl) -6-ethynyl- 1 H-indazol-3 -yl)benzoic acid; 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(3-methyl-l,2,4-oxadiazol-5-yl)-lH-indazol-
3-yl)-3-fluorobenzoic acid;
4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(5-(hydroxymethyl)oxazol-2-yl)-lH-indazol-
3-yl)-3-fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-( 1 -methyl- 1 H-imidazol-4-yl)- 1 H-indazol-3 - yl)-3-fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(oxazol-2-yl)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4-(6-(5-bromooxazol-2-yl)- 1 -(2-chloro-6-(trifluoromethyl)benzoyl)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
(E)-4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6-(N'-cyano-N,N- dimethylcarbamimidoyl)-l H-indazol-3 -yl)-3 -fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(5 -oxo-4, 5 -dihydro- 1 , 3 ,4-oxadiazol-2-yl)-
1 H-indazol-3 -yl)-3 -fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(4H- 1 ,2,4-triazol-3 -yl)- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(5 -methyl- 1 , 3 ,4-oxadiazol-2-yl)- 1 H-indazol-
3 -yl)-3 -fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-( 1 -methyl- 1 H-imidazol-2-yl)- 1 H-indazol-3 - yl)-3 -fluorobenzoic acid;
4-(l-(2-chloro-6-trifluorobenzoyl)-6-(thiazol-2-yl)-lH-indazol-3-yl)benzoic acid;
4-[l-{[2-chloro-6-(trifluoromethyl)phenyl]carbonyl}-6-(l -methyl- lH-imidazol-5-yl)- 1H- indazol-3-yl]benzoic acid;
4-[l-{[2-chloro-6-(trifluoromethyl)phenyl]carbonyl}-6-(5-methylthiophen-
3 - yl)- 1 H-indazol-3 -yljbenzoic acid;
4- ( 1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-pyrimidin-2-yl- 1 H-indazol-3 - yl)benzoic acid;
4-( 1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-pyrimidin-4-yl- 1 H-indazol-3 - yl)benzoic acid;
4-[l-{[2-chloro-6-(trifluoromethyl)phenyl]carbonyl}-6-(l -methyl- lH-imidazol-4-yl)- 1H- indazol-3 -yljbenzoic acid; 4- [ 1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-( 1 , 3 -thiazol-4-yl)- 1 H-indazol-3 - yljbenzoic acid;
4-(6- [4-(aminomethyl)pyridin-2-yl] - 1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } - 1 H- indazol-3-yl)benzoic acid;
4-( 1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-pyridin-2-yl- 1 H-indazol-3 - yl)benzoic acid;
4- [ 1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-( 1 , 3 -thiazol-5 -yl)- 1 H-indazol-3 - yljbenzoic acid;
4-(l-(2-chloro-6-trifluorobenzoyl)-6-(thiazol-2-yl)-lH-indazol-3-yl)benzoic acid;
4-( 1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-pyridin-4-yl- 1 H-indazol-3 - yl)benzoic acid;
4- [ 1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-(4-cyanophenyl)- 1 H-indazol-3 - yljbenzoic acid;
4- [ 1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-(3 -cyanophenyl)- 1 H-indazol-3 - yljbenzoic acid;
4- [ 1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-(2-cyanophenyl)- 1 H-indazol-3 - yljbenzoic acid;
4- [ 1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-(2-fluorophenyl)- 1 H-indazol-3 - yljbenzoic acid;
4- [ 1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-(3 -fluorophenyl)- 1 H-indazol-3 - yljbenzoic acid;
4- [ 1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-(4-fluorophenyl)- 1 H-indazol-3 - yljbenzoic acid;
4- [ 1 - { [2-chloro-6-(trifluoromethyl)phenyl] carbonyl } -6-( 1 -methyl- 1 H-pyrazol-5 -yl)- 1 H- indazol-3 -yljbenzoic acid;
methyl 4-(l-(2-chloro-6-trifluorobenzoyl)-6-(4-hydroxyphenyl)-l H-indazol-3 -yl)benzoate; 4-(l-(2-chloro-6-(trifluoromethyl)benzoyl)-6- (methylsulfonamidomethyl)-lH-indazol-3- yl)benzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(2, 5 - dioxoimidazolidin-4-yl)- 1 H-indazol-3 - yl)-3-fluorobenzoic acid;
4-(5 -(tert-butoxycarbonylamino)- 1 -(2-chloro-6- (trifluoromethyl)benzoyl)- 1 H-pyrrolo [2,3- c]pyridin-3 -yl)-3 -fluorobenzoic acid; 4-(6-(tert-butoxycarbonylamino)- 1 -(2-chloro-6- (trifluoromethyl)benzoyl)- 1H- pyrazolo[4,3-b]pyridin-3-yl)-3-fluorobenzoic acid;
4-( 1 -(2-chloro-6-(trifluoromethyl)benzoyl)-6-(methylamino)- 1 H- pyrazolo [4,3 -bjpyridin-
3-yl)-3-fluorobenzoic acid;
4-(5 -acetamido- 1 -(2-chloro-6-(trifluoromethyl)- benzoyl)- 1 H-pyrrolo [2,3 -c]pyridin-3 -yl)-
3-fluorobenzoic acid; and
4-(l-(2- chloro-6-(trifluoromethyl)benzoyl)-5-(methylamino)-lH-pyrrolo[2,3-c]pyridin-3- yl)-3-fluorobenzoic acid.
21. A pharmaceutical composition comprising a compound of claim 1 or a
pharmaceutically acceptable salt or solvate thereof and one or more pharmaceutically acceptable excipients.
22. The pharmaceutical composition of claim 21, which further comprises at least one
additional therapeutically active agent.
23. Use of a compound of claim 1, or a pharmaceutically acceptable salt or solvate thereof, in the manufacture of a medicament for the treatment of a disease or condition mediated by Retinoic acid receptor-related Orphan Receptor gamma t (RORgammaT).
24. A method for treating a disease or condition mediated by RORgammaT in a subject comprising administering to the subject an amount of a compound of claim 1, or a pharmaceutically acceptable salt or solvate thereof, that is effective for treating the disease or condition mediated by RORgammaT in the subject.
25. The method of claim 24, wherein the disease or condition is an autoimmune disease or inflammatory disease.
26. The method of claim 25, wherein the disease or condition is multiple sclerosis,
inflammatory bowel disease, Crohn's disease, ankylosing spondylitis, psoriasis, rheumatoid arthritis, asthma, osteoarthritis, Kawasaki disease, Hashimoto's thyroiditis or mucosal leishmaniasis.
PCT/CN2012/080131 2012-08-15 2012-08-15 4-heteroaryl substituted benzoic acid compounds as rorgammat inhibitors and uses thereof WO2014026327A1 (en)

Priority Applications (14)

Application Number Priority Date Filing Date Title
PCT/CN2012/080131 WO2014026327A1 (en) 2012-08-15 2012-08-15 4-heteroaryl substituted benzoic acid compounds as rorgammat inhibitors and uses thereof
EP13829282.6A EP2884981B1 (en) 2012-08-15 2013-08-14 4-HETEROARYL SUBSTITUTED BENZOIC ACID COMPOUNDS AS RORgammaT INHIBITORS AND USES THEREOF
RU2015108894A RU2015108894A (en) 2012-08-15 2013-08-14 COMPOUNDS OF 4-HETEROARYL SUBSTITUTED BENZOIC ACID AS RORGAMMAT INHIBITORS AND THEIR APPLICATIONS
CA2881688A CA2881688A1 (en) 2012-08-15 2013-08-14 4-heteroaryl substituted benzoic acid compounds as rorgammat inhibitors and uses thereof
PCT/US2013/054887 WO2014028589A2 (en) 2012-08-15 2013-08-14 4-HETEROARYL SUBSTITUTED BENZOIC ACID COMPOUNDS AS RORgammaT INHIBITORS AND USES THEREOF
CN201380053463.8A CN104853757A (en) 2012-08-15 2013-08-14 4-heteroaryl substituted benzoic acid compounds as rorgammat inhibitors and uses thereof
BR112015003188A BR112015003188A2 (en) 2012-08-15 2013-08-14 COMPOUND, PHARMACEUTICAL COMPOSITION, COMPOUND USE, AND METHOD FOR TREATMENT OF A RORGAMMAT-MEDIATED DISEASE OR CONDITION IN A PATIENT
AU2013302725A AU2013302725A1 (en) 2012-08-15 2013-08-14 4-heteroaryl substituted benzoic acid compounds as RORgammaT inhibitors and uses thereof
US14/421,048 US9745265B2 (en) 2012-08-15 2013-08-14 4-heteroaryl substituted benzoic acid compounds as RORgammaT inhibitors and uses thereof
MX2015001941A MX2015001941A (en) 2012-08-15 2013-08-14 4-HETEROARYL SUBSTITUTED BENZOIC ACID COMPOUNDS AS RORgammaT INHIBITORS AND USES THEREOF.
KR1020157006322A KR20150042266A (en) 2012-08-15 2013-08-14 4-HETEROARYL SUBSTITUTED BENZOIC ACID COMPOUNDS AS RORgammaT INHIBITORS AND USES THEREOF
JP2015527566A JP6422438B2 (en) 2012-08-15 2013-08-14 4-Heteroaryl-substituted benzoic acid compounds and their use as RORγT inhibitors
IN1156DEN2015 IN2015DN01156A (en) 2012-08-15 2015-02-12
US15/647,437 US10196354B2 (en) 2012-08-15 2017-07-12 4-heteroaryl substituted benzoic acid compounds as RORgammaT inhibitors and uses thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2012/080131 WO2014026327A1 (en) 2012-08-15 2012-08-15 4-heteroaryl substituted benzoic acid compounds as rorgammat inhibitors and uses thereof

Publications (1)

Publication Number Publication Date
WO2014026327A1 true WO2014026327A1 (en) 2014-02-20

Family

ID=50101185

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/CN2012/080131 WO2014026327A1 (en) 2012-08-15 2012-08-15 4-heteroaryl substituted benzoic acid compounds as rorgammat inhibitors and uses thereof
PCT/US2013/054887 WO2014028589A2 (en) 2012-08-15 2013-08-14 4-HETEROARYL SUBSTITUTED BENZOIC ACID COMPOUNDS AS RORgammaT INHIBITORS AND USES THEREOF

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/US2013/054887 WO2014028589A2 (en) 2012-08-15 2013-08-14 4-HETEROARYL SUBSTITUTED BENZOIC ACID COMPOUNDS AS RORgammaT INHIBITORS AND USES THEREOF

Country Status (11)

Country Link
US (2) US9745265B2 (en)
EP (1) EP2884981B1 (en)
JP (1) JP6422438B2 (en)
KR (1) KR20150042266A (en)
AU (1) AU2013302725A1 (en)
BR (1) BR112015003188A2 (en)
CA (1) CA2881688A1 (en)
IN (1) IN2015DN01156A (en)
MX (1) MX2015001941A (en)
RU (1) RU2015108894A (en)
WO (2) WO2014026327A1 (en)

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015139621A1 (en) * 2014-03-18 2015-09-24 北京韩美药品有限公司 4-heteroaryl substituted benzoic acid or benzamides compound
WO2016039358A1 (en) * 2014-09-10 2016-03-17 第一三共株式会社 Carboxylic acid derivative
WO2016061160A1 (en) * 2014-10-14 2016-04-21 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ror-gamma
WO2016130818A1 (en) * 2015-02-11 2016-08-18 Merck Sharp & Dohme Corp. SUBSTITUTED PYRAZOLE COMPOUNDS AS RORgammaT INHIBITORS AND USES THEREOF
US9481674B1 (en) 2016-06-10 2016-11-01 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US9522134B2 (en) 2014-10-24 2016-12-20 Orca Pharmaceuticals Limited Compounds
US9624217B2 (en) 2014-02-03 2017-04-18 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
WO2017075182A1 (en) * 2015-10-27 2017-05-04 Merck Sharp & Dohme Corp. Substituted indazole compounds as rorgammat inhibitors and uses thereof
US9845308B2 (en) 2014-11-05 2017-12-19 Vitae Pharmaceuticals, Inc. Isoindoline inhibitors of ROR-gamma
WO2018130537A1 (en) * 2017-01-10 2018-07-19 ETH Zürich Cell-protective compounds and their use
US20190062328A1 (en) * 2016-01-21 2019-02-28 Zibo Biopolar Changsheng Pharmaceutical Co., Ltd. Bruton's tyrosine kinase inhibitors
US10301261B2 (en) 2015-08-05 2019-05-28 Vitae Pharmaceuticals, Llc Substituted indoles as modulators of ROR-gamma
US10344000B2 (en) 2015-10-27 2019-07-09 Merck Sharp & Dohme Corp. Substituted bicyclic pyrazole compounds as RORgammaT inhibitors and uses thereof
US10508086B2 (en) 2014-12-19 2019-12-17 Cancer Research Technology Limited PARG inhibitory compounds
US10508113B2 (en) 2018-03-12 2019-12-17 Abbvie Inc. Inhibitors of tyrosine kinase 2 mediated signaling
US10584121B2 (en) 2015-10-27 2020-03-10 Merck Sharp & Dohme Corp. Heteroaryl substituted benzoic acids as RORgammaT inhibitors and uses thereof
US10829481B2 (en) 2016-01-29 2020-11-10 Vitae Pharmaceuticals, Llc Benzimidazole derivatives as modulators of ROR-gamma
US10913739B2 (en) 2017-07-24 2021-02-09 Vitae Pharmaceuticals, LLC (121374) Inhibitors of RORγ
US11001583B2 (en) 2014-11-05 2021-05-11 Vitae Pharmaceuticals, Llc Dihydropyrrolopyridine inhibitors of ROR-gamma
US11008340B2 (en) 2015-11-20 2021-05-18 Vitae Pharmaceuticals, Llc Modulators of ROR-gamma
US11186573B2 (en) 2017-07-24 2021-11-30 Vitae Pharmaceuticals, Llc Inhibitors of ROR gamma
WO2022242697A1 (en) * 2021-05-19 2022-11-24 南京药石科技股份有限公司 Tyk2 selective inhibitor and use thereof

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2487159A1 (en) 2011-02-11 2012-08-15 MSD Oss B.V. RorgammaT inhibitors
WO2014026327A1 (en) 2012-08-15 2014-02-20 Merck Sharp & Dohme Corp. 4-heteroaryl substituted benzoic acid compounds as rorgammat inhibitors and uses thereof
WO2014026330A1 (en) 2012-08-15 2014-02-20 Merck Sharp & Dohme Corp. 3-AMINOCYCLOALKYL COMPOUNDS AS RORgammaT INHIBITORS AND USES THEREOF
WO2014026329A1 (en) 2012-08-15 2014-02-20 Merck Sharp & Dohme Corp. N-alkylated indole and indazole compounds as rorgammat inhibitors and uses thereof
WO2014026328A1 (en) * 2012-08-15 2014-02-20 Merck Sharp & Dohme Corp. 3-cyclohexenyl substituted indole and indazole compounds as rorgammat inhibitors and uses thereof
WO2015095795A1 (en) 2013-12-20 2015-06-25 Merck Sharp & Dohme Corp. TETRAHYDRONAPHTHYRIDINE, BENZOXAZINE, AZA-BENZOXAZINE, AND RELATED BICYCLIC COMPOUNDS FOR INHIBITION OF RORgamma ACTIVITY AND THE TREATMENT OF DISEASE
JP2017507950A (en) 2014-02-27 2017-03-23 リセラ・コーポレイションLycera Corporation Adoptive cell therapy using retinoic acid receptor-related orphan receptor gamma agonists and related therapeutic methods
US10189777B2 (en) 2014-05-05 2019-01-29 Lycera Corporation Benzenesulfonamido and related compounds for use as agonists of RORγ and the treatment of disease
AU2015256190B2 (en) 2014-05-05 2019-08-15 Lycera Corporation Tetrahydroquinoline sulfonamide and related compounds for use as agonists of rory and the treatment of disease
EP3194392B1 (en) * 2014-09-15 2020-01-01 Plexxikon, Inc. Heterocyclic compounds and uses thereof
US10435402B2 (en) * 2015-01-08 2019-10-08 Advinus Therapeutics Limited Bicyclic compounds, compositions and medicinal applications thereof
EP3268087A4 (en) 2015-03-12 2018-08-29 The Regents of the University of California METHODS FOR TREATING CANCER WITH RORgamma INHIBITORS
US10421751B2 (en) 2015-05-05 2019-09-24 Lycera Corporation Dihydro-2H-benzo[b][1,4]oxazine sulfonamide and related compounds for use as agonists of RORγ and the treatment of disease
CN107980042B (en) 2015-06-11 2021-10-08 莱斯拉公司 Aryldihydro-2H-benzo [ b ] [1,4] oxazine sulfonamides and related compounds as ROR gamma agonists and for the treatment of disease
EP4071149A3 (en) * 2015-09-21 2023-01-25 Opna Immuno Oncology, SA Heterocyclic compounds and uses thereof
TWI817929B (en) * 2016-07-29 2023-10-11 美商瑞佩特治療公司 Chemokine receptor modulators and uses thereof
CA3056501A1 (en) 2017-04-11 2018-10-18 Sunshine Lake Pharma Co., Ltd. Fluorine-substituted indazole compounds and uses thereof
WO2018219797A1 (en) 2017-06-02 2018-12-06 Basf Se Substituted oxadiazoles for combating phytopathogenic fungi
GB201811825D0 (en) * 2018-07-19 2018-09-05 Benevolentai Bio Ltd Organic compounds

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0429257A2 (en) * 1989-11-17 1991-05-29 Glaxo Group Limited Indole derivatives
WO2006063167A1 (en) * 2004-12-08 2006-06-15 Smithkline Beecham Corporation 1h-pyrrolo[2,3-b]pyridines
EP2181710A1 (en) * 2008-10-28 2010-05-05 Phenex Pharmaceuticals AG Ligands for modulation of orphan receptor-gamma (NR1F3) activity
WO2012106995A1 (en) * 2011-02-11 2012-08-16 Merck Sharp & Dohme Corp. Rorgammat inhibitors

Family Cites Families (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4670447A (en) 1983-08-22 1987-06-02 Hoechst-Roussel Pharmaceuticals Inc. Antipsychotic 3-(piperidinyl)- and 3-(pyrrolidinyl)-1H-indazoles
US5639780A (en) 1995-05-22 1997-06-17 Merck Frosst Canada, Inc. N-benzyl indol-3-yl butanoic acid derivatives as cyclooxygenase inhibitors
US6133290A (en) 1998-07-31 2000-10-17 Eli Lilly And Company 5-HT1F agonists
GB0117577D0 (en) 2001-02-16 2001-09-12 Aventis Pharm Prod Inc Novel heterocyclic substituted carbonyl derivatives and their use as dopamine D receptor ligands
US7355042B2 (en) 2001-10-16 2008-04-08 Hypnion, Inc. Treatment of CNS disorders using CNS target modulators
US7592363B2 (en) 2004-08-03 2009-09-22 Wyeth Indazoles
CA2583428A1 (en) 2004-09-03 2006-03-09 Plexxikon, Inc. Bicyclic heteroaryl pde4b inhibitors
SE0402762D0 (en) 2004-11-11 2004-11-11 Astrazeneca Ab Indazole sulphonamide derivatives
CN100516049C (en) 2004-11-16 2009-07-22 永信药品工业股份有限公司 Synthesis of N2 - (substituted arylmethyl) -3- (substituted phenyl) indazoles as anti-angiogenic agents
AU2007217040A1 (en) 2006-02-17 2007-08-30 Memory Pharmaceuticals Corporation Compounds having 5-HT6 receptor affinity
JP4675801B2 (en) 2006-03-06 2011-04-27 日本メナード化粧品株式会社 ROR activator
JP2009529047A (en) 2006-03-07 2009-08-13 アレイ バイオファーマ、インコーポレイテッド Heterobicyclic pyrazole compounds and uses thereof
GB0611587D0 (en) 2006-06-12 2006-07-19 Glaxo Group Ltd Novel compounds
EP2134685B1 (en) 2007-04-16 2015-09-02 AbbVie Inc. 7-nonsubstituted indole derivatives as mcl-1 inhibitors
GB0708141D0 (en) 2007-04-26 2007-06-06 Syngenta Participations Ag Improvements in or relating to organic compounds
AR067326A1 (en) 2007-05-11 2009-10-07 Novartis Ag IMIDAZOPIRIDINES AND PIRROLO -PIRIMIDINES REPLACED AS INHIBITORS OF LIPIDO KINASE
NZ581698A (en) 2007-06-05 2012-09-28 Merck Sharp & Dohme Polycyclic indazole derivatives and their use as erk inhibitors for the treatment of cancer
JP2010534665A (en) 2007-07-25 2010-11-11 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Glucocorticoid mimetics, methods for their production, pharmaceutical compositions, and uses thereof
WO2010050837A1 (en) 2008-10-28 2010-05-06 Haskin Lev Yakovlevich Wind power plant
EP2379561B1 (en) 2008-11-25 2015-11-04 University Of Rochester Mlk inhibitors and methods of use
WO2010150837A1 (en) 2009-06-25 2010-12-29 第一三共株式会社 Indoline derivative
EP2459184A1 (en) 2009-07-31 2012-06-06 The Brigham and Women's Hospital, Inc. Modulation of sgk1 expression in th17 cells to modulate th17-mediated immune responses
CA2784753A1 (en) 2009-12-18 2011-06-23 Janssen Pharmaceutica Nv Substituted aminothiazolone indazoles as estrogen related receptor-.alpha. modulators
EP2536275A1 (en) 2010-02-16 2012-12-26 UWM Research Foundation, Inc. Methods of reducing virulence in bacteria
EP2571361A4 (en) 2010-05-19 2013-11-13 Univ North Carolina Pyrazolopyrimidine compounds for the treatment of cancer
EP2638014B1 (en) 2010-11-08 2017-01-04 Lycera Corporation N-sulfonylated tetrahydroquinolines and related bicyclic compounds for inhibition of ror-gamma activity and the treatment of diseases
KR20120063283A (en) 2010-12-07 2012-06-15 제일약품주식회사 Novel pyrazolopyridine derivatives or pharmaceutically acceptable salts thereof, process for the preparation thereof and pharmaceutical composition comprising the same
CN108586500B (en) 2010-12-22 2021-06-25 慕尼黑路德维希马克西米利安斯大学 Zinc coordination complex and method for preparing organic zinc reagent
JP2014166961A (en) 2011-06-20 2014-09-11 Dainippon Sumitomo Pharma Co Ltd Novel indazole derivative
WO2014026329A1 (en) 2012-08-15 2014-02-20 Merck Sharp & Dohme Corp. N-alkylated indole and indazole compounds as rorgammat inhibitors and uses thereof
WO2014026328A1 (en) 2012-08-15 2014-02-20 Merck Sharp & Dohme Corp. 3-cyclohexenyl substituted indole and indazole compounds as rorgammat inhibitors and uses thereof
WO2014026327A1 (en) 2012-08-15 2014-02-20 Merck Sharp & Dohme Corp. 4-heteroaryl substituted benzoic acid compounds as rorgammat inhibitors and uses thereof
WO2014026330A1 (en) 2012-08-15 2014-02-20 Merck Sharp & Dohme Corp. 3-AMINOCYCLOALKYL COMPOUNDS AS RORgammaT INHIBITORS AND USES THEREOF

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0429257A2 (en) * 1989-11-17 1991-05-29 Glaxo Group Limited Indole derivatives
WO2006063167A1 (en) * 2004-12-08 2006-06-15 Smithkline Beecham Corporation 1h-pyrrolo[2,3-b]pyridines
EP2181710A1 (en) * 2008-10-28 2010-05-05 Phenex Pharmaceuticals AG Ligands for modulation of orphan receptor-gamma (NR1F3) activity
WO2012106995A1 (en) * 2011-02-11 2012-08-16 Merck Sharp & Dohme Corp. Rorgammat inhibitors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
WANG YONGJUN ET AL.: "Identification of SRI 078, a synthetic agonist for the orphan nuclear receptors ROR a and RORy", ACS CHEMICAL BIOLOGY, vol. 5, no. 11, 2010, pages 1029 - 1034 *

Cited By (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10399976B2 (en) 2014-02-03 2019-09-03 Vitae Pharmaceuticals, Llc Dihydropyrrolopyridine inhibitors of ROR-gamma
US10807980B2 (en) 2014-02-03 2020-10-20 Vitae Pharmaceuticals, Llc Dihydropyrrolopyridine inhibitors of ROR-gamma
US10047085B2 (en) 2014-02-03 2018-08-14 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US11535614B2 (en) 2014-02-03 2022-12-27 Vitae Pharmaceuticals, Llc Dihydropyrrolopyridine inhibitors of ROR-gamma
US9624217B2 (en) 2014-02-03 2017-04-18 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
WO2015139621A1 (en) * 2014-03-18 2015-09-24 北京韩美药品有限公司 4-heteroaryl substituted benzoic acid or benzamides compound
WO2016039358A1 (en) * 2014-09-10 2016-03-17 第一三共株式会社 Carboxylic acid derivative
WO2016061160A1 (en) * 2014-10-14 2016-04-21 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ror-gamma
CN107074852B (en) * 2014-10-14 2019-08-16 生命医药有限责任公司 The pyrrolin and pyridine inhibitors of ROR- γ
EA031967B1 (en) * 2014-10-14 2019-03-29 Вайтаи Фармасьютиклз, Инк. Dihydropyrrolopyridine inhibitors of ror-gamma
AU2015333610B2 (en) * 2014-10-14 2019-11-07 Vitae Pharmaceuticals, Llc Dihydropyrrolopyridine inhibitors of ROR-gamma
US10087184B2 (en) 2014-10-14 2018-10-02 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of RORγ
US9796710B2 (en) 2014-10-14 2017-10-24 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
CN107074852A (en) * 2014-10-14 2017-08-18 生命医药公司 ROR γ pyrrolin and pyridine inhibitors
US9522134B2 (en) 2014-10-24 2016-12-20 Orca Pharmaceuticals Limited Compounds
US9861612B2 (en) 2014-10-24 2018-01-09 Orca Pharmaceuticals Limited Indolyl-containing RORγt inhibitors
US11001583B2 (en) 2014-11-05 2021-05-11 Vitae Pharmaceuticals, Llc Dihydropyrrolopyridine inhibitors of ROR-gamma
US9845308B2 (en) 2014-11-05 2017-12-19 Vitae Pharmaceuticals, Inc. Isoindoline inhibitors of ROR-gamma
US10995073B2 (en) 2014-12-19 2021-05-04 Cancer Research Technology Limited PARG inhibitory compounds
US10508086B2 (en) 2014-12-19 2019-12-17 Cancer Research Technology Limited PARG inhibitory compounds
US10221142B2 (en) 2015-02-11 2019-03-05 Merck Sharp & Dohme Corp. Substituted pyrazole compounds as RORgammaT inhibitors and uses thereof
WO2016130818A1 (en) * 2015-02-11 2016-08-18 Merck Sharp & Dohme Corp. SUBSTITUTED PYRAZOLE COMPOUNDS AS RORgammaT INHIBITORS AND USES THEREOF
US10301261B2 (en) 2015-08-05 2019-05-28 Vitae Pharmaceuticals, Llc Substituted indoles as modulators of ROR-gamma
US10829448B2 (en) 2015-08-05 2020-11-10 Vitae Pharmaceuticals, Llc Substituted benzoimidazoles as modulators of ROR-γ
US10287272B2 (en) 2015-10-27 2019-05-14 Merck Sharp & Dohme Corp. Substituted indazole compounds as RORgammaT inhibitors and uses thereof
WO2017075182A1 (en) * 2015-10-27 2017-05-04 Merck Sharp & Dohme Corp. Substituted indazole compounds as rorgammat inhibitors and uses thereof
US10344000B2 (en) 2015-10-27 2019-07-09 Merck Sharp & Dohme Corp. Substituted bicyclic pyrazole compounds as RORgammaT inhibitors and uses thereof
CN108431009A (en) * 2015-10-27 2018-08-21 默沙东公司 Substituted indazole compound and application thereof as ROR γ T inhibitor
JP2018535958A (en) * 2015-10-27 2018-12-06 メルク・シャープ・アンド・ドーム・コーポレーションMerck Sharp & Dohme Corp. Substituted indazole compounds as RORγT inhibitors and uses thereof
US10584121B2 (en) 2015-10-27 2020-03-10 Merck Sharp & Dohme Corp. Heteroaryl substituted benzoic acids as RORgammaT inhibitors and uses thereof
US10689369B2 (en) 2015-10-27 2020-06-23 Merck Sharp & Dohme Corp. Substituted indazole compounds as RORgammaT inhibitors and uses thereof
US11008340B2 (en) 2015-11-20 2021-05-18 Vitae Pharmaceuticals, Llc Modulators of ROR-gamma
US10662187B2 (en) * 2016-01-21 2020-05-26 Zibo Biopolar Changsheng Pharmaceutical Co. Ltd. Bruton's tyrosine kinase inhibitors
US20190062328A1 (en) * 2016-01-21 2019-02-28 Zibo Biopolar Changsheng Pharmaceutical Co., Ltd. Bruton's tyrosine kinase inhibitors
US10829481B2 (en) 2016-01-29 2020-11-10 Vitae Pharmaceuticals, Llc Benzimidazole derivatives as modulators of ROR-gamma
US9481674B1 (en) 2016-06-10 2016-11-01 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
WO2018130537A1 (en) * 2017-01-10 2018-07-19 ETH Zürich Cell-protective compounds and their use
CN110494136A (en) * 2017-01-10 2019-11-22 瑞士苏黎世联邦理工学院 Cytoprotective compounds and application thereof
US11628159B2 (en) 2017-01-10 2023-04-18 Eth Zurich Cell-protective compounds and their use
US10913739B2 (en) 2017-07-24 2021-02-09 Vitae Pharmaceuticals, LLC (121374) Inhibitors of RORγ
US11186573B2 (en) 2017-07-24 2021-11-30 Vitae Pharmaceuticals, Llc Inhibitors of ROR gamma
US10508113B2 (en) 2018-03-12 2019-12-17 Abbvie Inc. Inhibitors of tyrosine kinase 2 mediated signaling
WO2022242697A1 (en) * 2021-05-19 2022-11-24 南京药石科技股份有限公司 Tyk2 selective inhibitor and use thereof

Also Published As

Publication number Publication date
US9745265B2 (en) 2017-08-29
AU2013302725A1 (en) 2015-03-05
EP2884981A2 (en) 2015-06-24
US20180022701A1 (en) 2018-01-25
US10196354B2 (en) 2019-02-05
RU2015108894A (en) 2016-10-10
JP2015526441A (en) 2015-09-10
JP6422438B2 (en) 2018-11-14
CA2881688A1 (en) 2014-02-20
EP2884981B1 (en) 2018-06-20
WO2014028589A2 (en) 2014-02-20
KR20150042266A (en) 2015-04-20
BR112015003188A2 (en) 2017-08-22
MX2015001941A (en) 2015-08-14
EP2884981A4 (en) 2016-03-23
IN2015DN01156A (en) 2015-06-26
WO2014028589A3 (en) 2014-05-08
US20150218096A1 (en) 2015-08-06

Similar Documents

Publication Publication Date Title
US10196354B2 (en) 4-heteroaryl substituted benzoic acid compounds as RORgammaT inhibitors and uses thereof
US10577367B2 (en) IRAK4 inhibiting agents
EP2683693B1 (en) Rorgammat inhibitors
EP2884977B1 (en) N-ALKYLATED INDOLE AND INDAZOLE COMPOUNDS AS RORgammaT INHIBITORS AND USES THEREOF
EP2920149B1 (en) 3-aminocycloalkyl compounds as ror-gamma-t inhibitors and uses thereof
JP6242390B2 (en) 2-Aminopyrazine derivatives as CSF-1R kinase inhibitors
EP2884976A2 (en) 3-CYCLOHEXENYL AND CYCLOHEXYL SUBSTITUTED INDOLE AND INDAZOLE COMPOUNDS AS RORgammaT INHIBITORS AND USES THEREOF
JP2017531679A (en) Indole carboxamides useful as kinase inhibitors
EP3209664A1 (en) Bicyclic heteroaryl amine compounds as pi3k inhibitors
WO2014151630A2 (en) Compounds and compositions for the treatment of parasitic diseases
JP2022540200A (en) Indazole and Azaindazole as LRRK2 Inhibitors
WO2024029489A1 (en) Kit inhibitors, compounds, pharmaceutical compositions, and methods of use thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 12883012

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12883012

Country of ref document: EP

Kind code of ref document: A1