WO2014026329A1 - N-alkylated indole and indazole compounds as rorgammat inhibitors and uses thereof - Google Patents

N-alkylated indole and indazole compounds as rorgammat inhibitors and uses thereof Download PDF

Info

Publication number
WO2014026329A1
WO2014026329A1 PCT/CN2012/080134 CN2012080134W WO2014026329A1 WO 2014026329 A1 WO2014026329 A1 WO 2014026329A1 CN 2012080134 W CN2012080134 W CN 2012080134W WO 2014026329 A1 WO2014026329 A1 WO 2014026329A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
pyridin
alkyl
pyrazolo
chloro
Prior art date
Application number
PCT/CN2012/080134
Other languages
French (fr)
Inventor
Kenneth Jay BARR
Corey Beinstock
John Maclean
Hongjun Zhang
Richard Thomas Beresis
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/CN2012/080134 priority Critical patent/WO2014026329A1/en
Priority to CA2881693A priority patent/CA2881693A1/en
Priority to US14/421,052 priority patent/US9556168B2/en
Priority to PCT/US2013/054893 priority patent/WO2014028591A2/en
Priority to RU2015108935A priority patent/RU2015108935A/en
Priority to CN201380053436.0A priority patent/CN104822377A/en
Priority to JP2015527567A priority patent/JP6411345B2/en
Priority to EP13829125.7A priority patent/EP2884977B1/en
Priority to BR112015003397A priority patent/BR112015003397A2/en
Priority to AU2013302727A priority patent/AU2013302727A1/en
Priority to KR20157006321A priority patent/KR20150041659A/en
Publication of WO2014026329A1 publication Critical patent/WO2014026329A1/en

Links

Classifications

    • 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
    • A61K31/41Heterocyclic 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
    • A61K31/41Heterocyclic 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
    • A61K31/41621,2-Diazoles condensed with heterocyclic ring 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
    • A61K31/41Heterocyclic 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
    • A61K31/42Oxazoles
    • A61K31/422Oxazoles not condensed and containing further heterocyclic rings
    • 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
    • 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
    • A61P17/16Emollients or protectives, e.g. against radiation
    • 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
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • 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
    • 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
    • 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
    • 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
    • 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.
  • 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 activity, 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, N or CR a ;
  • 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
  • R 2 is hydroxycarbonyl, hydroxycarbonyl(Ci-io)alkyl, (Ci-io)alkylsulfoxyaminocarbonyl, or carbamoyl;
  • R 4 -R 7 independently are H, halogen, amino, cyano, hydroxy, (Ci -3 )alkoxy, (Ci -4 )alkyl, (Co-io)alkyl)aminocarbonyl, (di)(Ci-6)alkylaminocarbonyl or amino(Ci -4 )alkyl, wherein (Ci_3)alkoxy, (Ci -4 )alkyl, (Co-io)alkyl)aminocarbonyl, (di)(Ci-6)alkylaminocarbonyl and amino(Ci -4 )alkyl are optionally substituted with one or more halogen, hydroxyl or (Ci_
  • R 8 is halogen, cyano, amino, nitro, hydroxy, H 2 NC(0)-, (Ci -3 )alkoxycarbonyl, (di)(Ci_
  • (Ci -3 )alkoxy are optionally substituted with one, two or three halogens;
  • x 0, 1, 2, 3, 4 or 5;
  • R 9 is hydrogen or (Ci -4 )alkyl.
  • first subset of the second embodiment is a compound wherein Y is N.
  • Y is N.
  • a first subset of the first embodiment is a compound having Formula le
  • 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 , CR 5 , N, CR 7 .
  • a fourth subset of the first embodiment 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 3 -7)cycloalkyl or (C 3 -5)heterocycloalkyl, both optionally substituted with one or more R 8 , wherein R 8 is selected from halogen, amino, cyano, nitro, hydroxy, 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 halogens; (ii) (C 2- 9)heteroaryl, optionally substituted with one or more R 8 , wherein R 8 is selected from halogen, amino, cyano, nitro, hydroxy, H
  • R 1 is (C 2- 9)heteroaryl, or (ii) (C 6 -i 4 )aryl, optionally substituted with one, two, three, four or five R 8 .
  • R 8 is selected from halogen, amino, cyano, nitro, hydroxy, (Ci-3)alkoxycarbonyl, (Ci -4 )alkyl, (Ci_3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (Ci -4 )alkyl and (Ci_3)alkoxy are optionally substituted with one or more halogens.
  • R 1 is phenyl, optionally substituted with one, two or three R 8 .
  • R 8 is selected from halogen, amino, cyano, nitro, hydroxy, (Ci-3)alkoxycarbonyl, (Ci -4 )alkyl or (Ci-3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (Ci -4 )alkyl and (Ci_3)alkoxy are optionally substituted with one or more halogens.
  • R 2 is C(0)OH.
  • a still further embodiment of the compounds of Formula I, la, lb, Ic, Id, Ie, If, Ig, and Ih are compounds wherein one of R 4 , R 5 , R 6 , and R 7 is other than hydrogen.
  • the invention also relates to those compounds wherein all specific definitions for A 1 through A 4 , R 1 through R 9 , 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.
  • 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, 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.
  • 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-
  • C °> is alternatively referred to as phenyl having as a substituent methylenedioxy attached to two adjacent carbon atoms.
  • 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
  • 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)
  • 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.
  • 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.
  • 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
  • the attachment point is at the last group.
  • (Ci-3)alkoxycarbonyl refers to, e.g. 5 and the term
  • (Cl-4)alkylcarbonyloxy refers to, e.g.
  • 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.
  • an 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.
  • 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 HPLC 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.
  • 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.
  • 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
  • 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.
  • 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 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 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, inflammatory bowel disease, Crohn's disease, ankylosing spondylitis, psoriasis, rheumatoid arthritis, asthma, osteoarthritis, Kawasaki disease, Hashimoto's thyroiditis or mucosal leishmaniasis.
  • 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 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.
  • aqueous suspensions, isotonic saline solutions and sterile injectable solutions may be used, containing
  • 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) 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.
  • 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.
  • 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.
  • 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.
  • 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).
  • tetrahydrofuran DMSO: Dimethyl sulfoxide; TBAI: Tetrabutylammonium iodide; TsCl: 4-toluene sulfonyl chloride; DMAP: N,N-dimethylpyridin-4-amine; Et3N: triethylamine; ACN: acetonitrile; MsCl: methanesulfonyl chloride; (COCl) 2 : oxalyl dichloride; LiBH 4 : lithium tetrahydroborate; t-BuOK: Potassium tert-butoxide.
  • DMSO Dimethyl sulfoxide
  • TBAI Tetrabutylammonium iodide
  • TsCl 4-toluene sulfonyl chloride
  • DMAP N,N-dimethylpyridin-4-amine
  • Et3N triethylamine
  • ACN acetonitrile
  • A-6 1A i) Preparation of 2-chloro-6-(trifluoromethyl)benzyl 4-methylbenzenesulfonate (A-2).
  • the compounds of the invention inhibit RORgammaT activity.
  • Activation of RORgammaT activity can be measured using e.g. biochemical TR-FRET assay.
  • TR-FRET interaction of cofactor-derived peptides with human RORgammaT-Ligand Binding Domain (LBD) can be measured.
  • LBD human 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).
  • 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).
  • LXXXLL 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

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Rheumatology (AREA)
  • Pulmonology (AREA)
  • Neurosurgery (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Dermatology (AREA)
  • Pain & Pain Management (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Transplantation (AREA)
  • Toxicology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Tropical Medicine & Parasitology (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)
  • Peptides Or Proteins (AREA)

Abstract

Disclosed are compounds of 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

N-ALKYLATED INDOLE AND INDAZOLE 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 activity, 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 imgf000005_0001
I or a pharmaceutically acceptable salt or solvate thereof wherein,
Y is CH, N or CRa;
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;
R1 is
(i) (C3-i2)carbocyclyl; or
(ii) a 4- to 12-membered heterocyclyl,
both (i) and (ii) 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, (Cl-3)alkylC(0)0-, (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 H, halogen, amino, cyano, hydroxy, (Ci-3)alkoxy, (Ci-4)alkyl, (Co-io)alkyl)aminocarbonyl, (di)(Ci-6)alkylaminocarbonyl or amino(Ci-4)alkyl, wherein (Ci_3)alkoxy, (Ci-4)alkyl, (Co-io)alkyl)aminocarbonyl, (di)(Ci-6)alkylaminocarbonyl 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;
R8 is halogen, cyano, amino, nitro, hydroxy, H2NC(0)-, (Ci-3)alkoxycarbonyl, (di)(Ci_
6)alkylaminocarbonyl, (Ci-4)alkyl, (C3-7)cycloalkyl, (C3-5)heterocycloalkyl, 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;
x is 0, 1, 2, 3, 4 or 5;
R9 is hydrogen or (Ci-4)alkyl.
In a first embodiment of the compound having Formula I is a compound having Formula la
Figure imgf000006_0001
la and a pharmaceutically acceptable salt or solvate thereof. In a second embodiment of the compound having Formula I is a compound having Formula lb
Figure imgf000007_0001
lb and a pharmaceutically acceptable salt or solvate thereof.
In a first subset of the second embodiment is a compound wherein Y is N.
In a second subset of the second embodiment is a compound having Formula Ic
Figure imgf000007_0002
Ic
and a pharmaceutically acceptable salt or solvate thereof. a subset of the compound having Formula Ic, is a compound having Formula Id
Figure imgf000008_0001
and a pharmaceutically acceptable salt or solvate thereof. In a further subset, Y is N. In a first subset of the first embodiment is a compound having Formula le
Figure imgf000008_0002
le
and a pharmaceutically acceptable salt or solvate thereof.
In a subset of the compound having Formula le is a compound having Formula If
Figure imgf000009_0001
If
and a pharmaceutically acceptable salt or solvate thereof.
In a subset of the compound having Formula If is a compound having Formula Ig
Figure imgf000009_0002
Ig
and a pharmaceutically acceptable salt or solvate thereof.
In a subset of the compound having Formula Ig is a compound having Formula Ih
Figure imgf000010_0001
Ih and a pharmaceutically acceptable salt or solvate thereof.
In a second 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 third 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; and (iii) CR4, CR5, N, CR7.
In a fourth subset of the first embodiment 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 a fifth subset of the first embodiment is compound wherein R1 is (i) (C3-7)cycloalkyl or (C3-5)heterocycloalkyl, both optionally substituted with one or more R8, wherein R8 is selected from halogen, amino, cyano, nitro, hydroxy, 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 halogens; (ii) (C2-9)heteroaryl, optionally substituted with one or more R8, wherein R8 is selected from halogen, amino, cyano, nitro, hydroxy, 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 halogens; or (iii) (C6-i4)aryl, optionally substituted with one or more R8, wherein R8 is selected from halogen, amino, cyano, nitro, hydroxy, 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 or (Ci-3)alkoxy are optionally substituted with one or more halogens.
In a sixth subset of the first embodiment is compound wherein R1 is (C2-9)heteroaryl, or (ii) (C6-i4)aryl, optionally substituted with one, two, three, four or five R8. In a further subset R8 is selected from halogen, amino, cyano, nitro, hydroxy, (Ci-3)alkoxycarbonyl, (Ci-4)alkyl, (Ci_3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (Ci-4)alkyl and (Ci_3)alkoxy are optionally substituted with one or more halogens.
In a seventh subset of the first embodiment, R1 is (C6-i4)aryl, optionally substituted with one, two, three, four or five R8. In a further subset R8 is selected from halogen, cyano, (C1-3)- alkoxycarbonyl, (Ci-4)alkyl or (Ci-3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (Ci-4)alkyl and (Ci_3)alkoxy are optionally substituted with one, two or three halogens.
In an eighth subset of the first embodiment, R1 is phenyl, naphthyl, pyridinyl, quinolinyl, benzooxadiazolyl, thiophenyl, isoxazolyl, or benzothiophenyl, each optionally substituted with one or more R8. In a further subset R8 is selected from halogen, amino, cyano, nitro, hydroxy, (Ci-3)alkoxycarbonyl, (Ci-4)alkyl or (Ci-3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (Ci-4)alkyl and (Ci-3)alkoxy are optionally substituted with one or more halogens.
In a ninth subset of the first embodiment, R1 is phenyl, optionally substituted with one, two or three R8. In a further subset R8 is selected from halogen, amino, cyano, nitro, hydroxy, (Ci-3)alkoxycarbonyl, (Ci-4)alkyl or (Ci-3)alkoxy, wherein (Ci-3)alkoxycarbonyl, (Ci-4)alkyl and (Ci_3)alkoxy are optionally substituted with one or more halogens.
In a tenth subset of the first embodiment, R2 is C(0)OH. 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 R9, 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 compound of the present invention include:
4- { 1 - [2-chloro-6-(trifluoromethyl)benzyl] - 1 H-pyrazolo [3 ,4-c]pyridin-3 -yl } -3 -fluorobenzoic acid;
4- [ 1 -(2-bromo-6-fluorobenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl] -3 -fluorobenzoic acid;
4- { 1 - [2-chloro-6-(trifluoromethyl)benzyl] -4-fluoro- 1 H-indazol-3 -yl } -3 -fluorobenzoic acid;
4- { 1 - [2-chloro-6-(trifluoromethyl)benzyl] -4-fluoro- 1 H-indazol-3 -yl } benzoic acid;
4- { 1 - [2-chloro-6-(trifluoromethyl)benzyl] -4-fluoro- 1 H-indazol-3 -yl } -2, 5 -difluorobenzoic acid;
4-( 1 - { 1 -[2-chloro-6-(trifluoromethyl)phenyl] ethyl } -4-fluoro- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4-( 1 - { ( 1 R or 1 S) - 1 - [2-chloro-6-(trifluoromethyl)phenyl] ethyl } -4-fluoro- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4-( 1 - { ( 1 S or 1 R) - 1 - [2-chloro-6-(trifluoromethyl)phenyl] ethyl } -4-fluoro- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4- [ 1 -(2-bromo-3 -fluorobenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl] -3 -fluorobenzoic acid;
4- [ 1 -(5-chloro-2-cyanobenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl] -3 -fluorobenzoic acid;
3 -fluoro-4-( 1 - { 1 - [2-(trifluoromethyl)phenyl] ethyl } - 1 H-pyrazolo [4,3 -b]pyridin-3 -yl)benzoic acid;
4- [ 1 -(6-chloro-2-fluoro-3 -methylbenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl] -3 -fluorobenzoic acid;
4- [ 1 -(2-chloro-3 , 6-difluorobenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl] -3 -fluorobenzoic acid; 3 -fluoro-4- [ 1 -(2, 3 , 6-trifluorobenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl]benzoic acid; 3 -fluoro-4- { 1 -[2-fluoro-6-(trifluoromethyl)benzyl] - 1 H-pyrazolo [4,3 -b]pyridin-3 -yl } benzoic acid;
4- [ 1 -(2, 6-difluorobenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl] -3 -fluorobenzoic acid;
4- [ 1 -(2-chloro-6-fluorobenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl] -3 -fluorobenzoic acid;
4- [ 1 -(6-chloro-2-fluoro-3 -methoxybenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl] -3 -fluorobenzoic acid;
4- [ 1 -(2-chloro-6-fluoro-3 -methoxybenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl] -3 -fluorobenzoic acid;
4- [ 1 -(2, 3 -dichloro-6-fluorobenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl] -3 -fluorobenzoic acid; 4- [ 1 -( 1 -benzothiophen-7-ylmethyl)- 1 H-pyrazolo [4, 3 -b]pyridin-3 -yl] -3 -fluorobenzoic acid; 4- { 1 - [2, 6-dichloro-3 -(trifluoromethyl)benzyl] - 1 H-pyrazolo [4,3 -b]pyridin-3 -yl } -3 - fluorobenzoic acid;
4- [ 1 -(3 , 6-dichloro-2-fluorobenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl] -3 -fluorobenzoic acid; 4- { 1 - [2-chloro-6-(methoxycarbonyl)benzyl] - 1 H-pyrazolo [4,3 -b]pyridin-3 -yl } -3 - fluorobenzoic acid;
4- [ 1 -(2-bromo-6-chlorobenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl] -3 -fluorobenzoic acid;
4- { 1 - [2-chloro-6-(trifluoromethyl)benzyl] - 1 H-pyrazolo [4,3 -b]pyridin-3 -yl } -3 -fluorobenzoic acid;
4- { 1 - [2-chloro-6-(trifluoromethyl)benzyl] - 1 H-pyrrolo [3 ,2-b]pyridin-3 -yl } -3 -fluorobenzoic acid; and
4- { 1 - [2-chloro-6-(trifluoromethyl)benzyl] - 1 H-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 imgf000015_0001
Figure imgf000016_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 imgf000017_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 imgf000017_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 , wherein s is an integer equal to zero, 1 or 2, the structure is
Figure imgf000018_0001
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 imgf000019_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 imgf000020_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, "C1-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 imgf000021_0001
5 and the term
(Cl-4)alkylcarbonyloxy refers to, e.g.
Figure imgf000021_0002
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 HPLC 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 actvity, 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 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; THF:
tetrahydrofuran; DMSO: Dimethyl sulfoxide; TBAI: Tetrabutylammonium iodide; TsCl: 4-toluene sulfonyl chloride; DMAP: N,N-dimethylpyridin-4-amine; Et3N: triethylamine; ACN: acetonitrile; MsCl: methanesulfonyl chloride; (COCl)2: oxalyl dichloride; LiBH4: lithium tetrahydroborate; t-BuOK: Potassium tert-butoxide.
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.
Figure imgf000036_0001
A B I
Alternatively the final compound I could also be prepared by switching the order of reaction sequence between N-alkylation and Suzuki coupling (see Scheme 2). Suzuki coupling first by reacting halide A with pinacol boronic ester or acid gave intermediate B. Subsequent N-alkylation followed by hydrolysis furnished final product.
Scheme 2.
Figure imgf000036_0002
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 imgf000037_0001
INTERMEDIATES Example i-1: Preparation of (2-chloro-6-(trifluoromethyl)phenyl)methanol (i-1)
Scheme i-1
Figure imgf000038_0001
-1a -1b i) . Preparation of methyl 2-chloro-6-(trifluoromethyl)benzoate (i-lb).
The mixture of 2-chloro-6-(trifluoromethyl)benzoic acid (i-la) (1.5 g, 6.70 mol) and (COCl)2 (1.1 ml, 13.4 mol) in DCM (20 ml) and DMF (5drops) was stirred at room temperature for 2h.Then MeOH (0.41 ml, 13.4 mol) was added dropwise and the reaction mixture was stirred at room temperature for another 30min.The result solution was diluted with H20 (50 ml) and the aqueous layer was extracted with DCM(50ml><2). The combined organic layers were washed with brine (50 ml x 1), dried over anhydrous Na2S04 and concentrated to get the desired product i-lb as a pale yellow oil. LCMS (ESI) calc'd for C9H6C1F302 [M+H] +: 239, found: 239. ii) Preparation of (2-chloro-6-(trifluoromethyl)phenyl)methanol (i-1).
The mixture of methyl 2-chloro-6-(trifluoromethyl)benzoate (i-lb) (1.0 g, 4.20 mol) and LiBH (0.18 g, 8.40 mol) in THF (10 ml) was stirred at room temperature for overnight. 2 M HC1 solutions (10 ml) were added to quench the reaction and the aqueous layer was extracted with EtOAc (20 ml x3). The combined organic layers were washed with brine (20 mix 1), dried over anhydrous Na2S04 and concentrated to get the desired product i-1 as a pale yellow oil. LCMS (ESI) calc'd for C8H6C1F30 [M+H] +: 211, found: 211.
Example i-2: Preparation of l-(2-chloro-6-(trifluoromethyl)phenyl)ethanol (i-2) Scheme i-2
Figure imgf000039_0001
i-1 i-2b i-2 i) Preparation of 2-chloro-6-(trifluoromethyl)benzaldehyde (i-2b).
The mixture of (2-chloro-6-(trifluoromethyl)phenyl)methanol (i-1) (0.7 g, 3.33 mol) and Dess-Martin periodinane (2.8g, 6.66 mol) in DCM (15 ml) was stirred at room temperature for overnight. The result solution was diluted with H20 (30mL) and the aqueous layer was extracted with DCM (30 ml><3). 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=10: 1) to get the desired product i-2b as a pale yellow solid. LCMS (ESI) calc'd for C8H4C1F30 [M+H] +: 209, found: 209. ii) Preparation of l-(2-chloro-6-(trifluoromethyl)phenyl)ethanol (i-2).
The mixture of 2-chloro-6-(trifluoromethyl)benzaldehyde (i-2b) (0.25 g, 1.20 mol) in anhydrous THF (10 ml) was cooled to 0°C in an ice-water bath and CH3MgBr (3.0M solution in ether, 2.0 ml, 6.0 mmol) was added dropwise. The reaction mixture was warmed to room temperature and stirred for 2h. Saturated H4C1 solution (20 ml) was added to quench the reaction and the aqueous layer was extracted with ethyl acetate (20 mix 3). The combined organic layers were washed with brine (20 mix 1), dried over anhydrous Na2S04 and concentrated to get the desired product i-2 as a pale yellow oil. LCMS (ESI): calc'd for C9H8C1F30 [M+H] +: 225, found: 225.
EXAMPLES
Example 1A: Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzyl)-lH-pyraz olo[3,4-c]pyridin-3-yl)-3-fluorobenzoic acid (1A). Scheme A
Figure imgf000040_0001
A-6 1A i) Preparation of 2-chloro-6-(trifluoromethyl)benzyl 4-methylbenzenesulfonate (A-2).
The mixture of (2-chloro-6-(trifluoromethyl)phenyl)methanol (i-1) (0.35 g, 1.67 mol), TsCl (0.64 g, 3.34 mol), DMAP (0.20 g, 1.67 mol) and Et3N (0.48 ml, 3.34 mol) in DCM (10 ml) was stirred at 25 °C for 23h. The result solution was diluted with H20 (30 ml) and the aqueous layer was extracted with DCM (20 ml χ3). The combined organic layers were washed with 1M HC1 solution (10 mix 2), then brine (20 ml x l), dried over anhydrous Na2S04 and concentrated to get the desired product A-2 as a yellow oil. LCMS (ESI) calc'd for CI5HI2C1F303S [M+ H4] +: 382, found: 382. ii) Preparation of 3-bromo-l-(2-chloro-6-(trifluoromethyl)benzyl)-lH-pyrazolo
[4,3-b] pyridine (A-4).
The mixture of 2-chloro-6-(trifluoromethyl)benzyl 4-methylbenzenesulfonate (A-2) (0.19 g, 0.51 mol), 3-bromo-lH-pyrazolo[3,4-c]pyridine (A-3) (0.1 g, 0.51 mol), t-BuOK (0.1 1 g, 1.02 mol) and TBAI (75 mg, 0.20 mol) in THF (5 ml) was heated to 60 °C and stirred at this temperature for overnight. Saturated H4C1 solution (20 ml) was added to quench the reaction and the aqueous layer was extracted with ethyl acetate (30 ml χ2). The combined organic layers were washed with brine (20 ml x 1), dried over anhydrous Na2S04 and concentrated to get the desired product A-4 as a brown oil. LCMS (ESI) calc'd for Ci4H8BrClF3N3 [M+H] +: 390, found: 390. iii) Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzyl)-lH-pyrazolo
[4,3-b] pyridin-3-yl)-3-fluorobenzoate (A-6).
A mixture of 3-bromo-l-(2-chloro-6-(trifluoromethyl)benzyl)-lH-pyrazolo[4,3-b]pyri dine (A-4) (120 mg, 0.31 mol), 4-(methoxycarbonyl)phenylboronic acid (A-5) ( 73 mg, 0.37 mol), Pd(PPh3)4 ( 36 mg, 0.031 mol) and K2C03 ( 128 mg, 0.93 mol) were suspended in 1,4- dioxane ( 5 ml) and H20 (1 ml). The reaction 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 χ2). The combined organic layers were washed with brine (30 ml x 1), dried over anhydrous Na2S04 and concentrated to get the crude product A-6 as a brown oil. LCMS (ESI) calc'd for C22Hi4ClF4N302 [M+H] +: 464, found: 464. iv) Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzyl)-lH-pyrazolo[3,4-c]p yridin-3-yl)-3-fluorobenzoic acid (1A).
The mixture of 4-(l-(2-chloro-6-(trifluoromethyl)benzyl)-lH-pyrazolo[4,3-b]pyridine -3-yl)-3-fluorobenzoate (A-6) (100 mg, 0.22 mol) and LiOH (28 mg, 0.66 mol) in THF (4 ml) and H20 (2 ml) was stirred at room temperature for overnight. The reaction mixture was diluted with H20 (30 ml). 2M HC1 solution was added to adjust the pH=3 and the aqueous layer was extracted with ethyl acetate (20 ml χ3). The combined organic layers were washed with brine (20 ml x l), dried over anhydrous Na2S04 and concentrated. The residue was purified with Prep-HPLC (ACN/H20) to get the desired product 1A as a white solid.. LCMS (ESI) calc'd for C2IHI2C1F4N302 [M+H]+: 450, found: 450; 1HNMR (400 MHz, MeOD) δ 8.63 (1H, d, J=4.4Hz), 8.20 (1H, d, J=8.8Hz), 8.05-8.09 (1H, m), 7.94 (1H, d, J=7.6Hz), 7.79- 7.87 (3H, m), 7.62-7.66 (1H, m), 7.52-7.55 (1H, m),5.97 (2H, s).
Example IB: Preparation of 4-(l-(2-bromo-6-fluorobenzyl)-lH-pyrazolo[4,3-b] pyridin-3-yl)-3-fluorobenzoic acid (IB) Scheme B
Figure imgf000042_0001
i) Preparation of methyl 3-fluoro-4-(lH-pyrazolo[4,3-b]pyridin-3-yl)benzoate (B-2)
A mixture of 3-bromo-lH-pyrazolo[4,3-b]pyridine (B-1) (196.9mg, 1 mol),4- (methoxycarbonyl)phenylboronic acid ( 198 mg, lmol), Pd(PPh3)4 (115mg, 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 χ2). The combined organic layers were washed with brine (30 ml x 1), dried over anhydrous Na2S04 and concentrated to get the crude product B-2 as a brown oil. LCMS (ESI) calc'd for Ci4HioFN302 [M+H] +: 272.08, found: 272. ii) Preparation of 4-(l-(2-bromo-6-fluorobenzyl)-lH-pyrazolo[4,3-b] pyridin-3 -yl)-3- fluorobenzoic acid (IB)
To a 1 dram vial was added methyl 3-fluoro-4-(lH-pyrazolo[4,3-b]pyridin-3-yl)benz oate (B-2) (30mg, O. l lmmol), l-bromo-2-(bromomethyl)-3-fluorobenzene (29.4mg, 0.11 lmmol), cesium carbonate(72 mg,0.22 mmol), and DMF(lml), The reaction mixtures were stirred at RT overnight. The mixtures were then evaporated under reduced pressure. THF (0.5ml), Methanol (0.25ml), and LiOH (1M, 0.332mmol) were then added and the reaction mixtures were stirred overnight at room temperature. The mixtures were then evaporated under reduced pressure. The reactions were then diluted with 2.0 ml DMSO, filtered, and purified by purified with Prep-HPLC (ACN/H20) to afford desired products. LCMS (ESI) calc'd for C2IHI2C1F4N302 [M+H]+: 444, found: 444. The following examples shown in TABLE 1 were prepared following similar procedures described for Examples #1A, IB in Schemes A, B. which can be achieved by those of ordinary skill in the art of organic synthesis.
TABLE 1
Figure imgf000043_0001
Figure imgf000043_0002
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Example 2A
Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzyl)-lH-pyrrolo[3,2-b]pyridi n-3-yl)-3-fluorobenzoic acid (2A).
Figure imgf000048_0002
C- 2A i) Preparation of 2-chloro-6-(trifluoromethyl)benzyl methanesulfonate (C-2) To the solution of (2-chloro-6-(trifluoromethyl)phenyl)methanol (i-1) (210 mg, 1 mmol) and Et3N (3 ml) dissolved in anhydrous THF (10 ml) was added MsCl (228 mg,2.0 mmol) drop wise. The mixture solution was protected by N2 and stirred at r.t for 0.5 h. Then the solution was filtered and concentrated to afford 267 mg product C-2 (yield: 93%). LCMS (ESI) calc'd [M+H] +: 288.67, found: 288.9. ii) Preparation of 3-bromo-l-(2-chloro-6-(trifluoromethyl)benzyl)-lH-pyrrolo[3
,2-b]pyridine (C-3)
The mixture of 2-chloro-6-(trifluoromethyl)benzyl methanesulfonate (C-2) (288 mg, lmmol), 3-bromo-lH-pyrrolo[3,2-b]pyridine (196 mg, 1.0 mmol), tBuOK (336 mg, 3.0 mmol), TBAI (106 mg, 0.4 mmol), THF(15 ml) and was protected by N2 and stirred at 60 °C for 3 h. Then the solution was filtered and concentrated to be purified by chromatography column (EA/PE=1 :4) to afford 289 mg product C-3 (yield: 74.5%). LCMS (ESI) calc'd
[M+H] +: 389.60, found: 389.8. iii) . Preparation of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzyl)-lH-pyrrolo
[3,2-b] pyridin-3-yl)-3-fluorobenzoate (C-4).
The mixture of 3-bromo-l-(2-chloro-6-(trifluoromethyl)benzyl)-lH-pyrrolo[3,2-b]py ridine (C-3) (216 mg, lmmol), 2-fluoro-4-(methoxycarbonyl)phenylboronic acid (298 mg, 1.5 mmol), Pd(PPh3)4 (5 mg), K2C03(414 mg, 3.0 mmol), Dioxane(15 ml) and H20(5 ml) was protected by N2 and stirred at 100°C for 16 h. Then the solution was filtered and concentrated to be purified by chromatography column (EA/PE=1 :4) to afford 364 mg product C-4 (yield: 78.8%). LCMS (ESI) calc'd [M+H] +: 462.86, found: 463.0. iv) . Preparation of 4-(l-(2-chloro-6-(trifluoromethyl)benzyl)-lH-pyrrolo[3,2-b]py ridin-3-yl)-3-fluorobenzoic acid (2A)
To the solution of methyl 4-(l-(2-chloro-6-(trifluoromethyl)benzyl)-lH-pyrrolo[3,2-b] pyridin-3-yl)-3-fluorobenzoate (C-4) (100 mg, 0.21 mmol) in THF (20 ml) and H20 (5 ml) was added LiOH (48 mg,3.0 mmol). The mixture solution was stirred at r.t for 16 h. Added water (30 ml), acidified by HC1 (2M), extracted with EA(20 ml X 3), combined the organic layer, dried and concentrated to be purified by pre-HPLC (ACN/H20) to afford 79.4 mg product 2A (yield: 82.3 %).¾Ν Κ (400 MHz, CDC13) δ: 8.61(lH,d), 8.52(lH,d), 8.32(lH,t), 7.91(3H,t), 7.82(1H,2), 7.70(2H,m), 7.53(lH,m),5.81(2H,s).
The following examples shown in TABLE 2 were prepared following similar procedures described for Example #2A in Scheme C. which can be achieved by those of ordinary skill in the art of organic synthesis.
TABLE 2
Figure imgf000050_0001
Figure imgf000050_0002
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. Examples Fret ICso (nM)
1A 142
IB 2293
1C 39
ID 65
IE 219
IF 1553
1G 725
1H 358
11 8228
1J >10000
IK >10000
1L 2122
1M 3102
IN >10000
10 1188
IP >10000
1Q 3792
1R 4306
I S 3708
IT 1916
1U 9120
IV 2170
1W 4199
IX 3264
1Y 599
1Z 380
2A 283
2B 33

Claims

A compound according to Formula I
Figure imgf000053_0001
I or a pharmaceutically acceptable salt or solvate thereof wherein,
Y is CH, N or CRa;
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;
R1 is
(i) (C3-i2)carbocyclyl; or
(ii) a 4- to 12-membered heterocyclyl,
both (i) and (ii) 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, (Cl-3)alkylC(0)0-, (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 H, halogen, amino, cyano, hydroxy, (Ci-3)alkoxy, (Ci-4)alkyl, (Co-io)alkyl)aminocarbonyl, (di)(Ci-6)alkylaminocarbonyl or amino(Ci-4)alkyl, wherein (Ci_3)alkoxy, (Ci-4)alkyl, (Co-io)alkyl)aminocarbonyl, (di)(Ci-6)alkylaminocarbonyl and amino(Ci-4)alkyl are optionally substituted with one or more halogen, hydroxyl or (Ci_
3) alkoxy; or a group having the
Figure imgf000054_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;
R8 is halogen, cyano, amino, nitro, hydroxy, H2NC(0)-, (Ci-3)alkoxycarbonyl, (di)(Ci_
6)alkylaminocarbonyl, (Ci-4)alkyl, (C3-7)cycloalkyl, (C3-5)heterocycloalkyl,
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; x is 0, 1, 2, 3, 4 or 5;
R9 is hydrogen or (Ci-4)alkyl.
The compound of claim 1 having Formula la
Figure imgf000054_0002
la and a pharmaceutically acceptable salt or solvate thereof. The compound of claim 1 having Formula lb
Figure imgf000055_0001
lb 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 imgf000055_0002
Ic
and a pharmaceutically acceptable salt or solvate thereof.
The compound of claim 5 having Formula Id and a pharmaceutically acceptable salt or solvate thereof.
The compound of claim 6, wherein Y is N.
The compound of claim 2 having Formula Ie
Figure imgf000056_0001
Ie
and a pharmaceutically acceptable salt or solvate thereof.
The compound of claim 8 having Formula If If
and a pharmaceutically acceptable salt or solvate thereof.
The compound of claim 9 having Formula Ig
Figure imgf000057_0001
11. The compound of claim 10 having Formula Ih
Figure imgf000058_0001
Ih 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, CR5, N, 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 12, wherein R1 is (C2-9)heteroaryl, or (ii) (C6-14)aryl, optionally substituted with one, two, three, four or five R8.
15. The compound of claim 14, wherein R1 is (C6-i4)aryl, optionally substituted with one, two, three, four or five R8.
16. The compound of claim 14, wherein R1 is phenyl, naphthyl, pyridinyl, quinolinyl,
benzooxadiazolyl, thiophenyl, isoxazolyl, or benzothiophenyl, each optionally substituted with one or more R8. 17. The compound of claim 16, wherein R1 is phenyl, optionally substituted with one, two or three R8.
18. The compound of claim 17, wherein R2 is C(0)OH.
19. A compound according to claim 1 selected from:
4- { 1 - [2-chloro-6-(trifluoromethyl)benzyl] - 1 H-pyrazolo [3 ,4-c]pyridin-3 -yl } -3 - fluorobenzoic acid;
4- [ 1 -(2-bromo-6-fluorobenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl] -3 -fluorobenzoic acid; 4- { 1 - [2-chloro-6-(trifluoromethyl)benzyl] -4-fluoro- 1 H-indazol-3 -yl } -3 -fluorobenzoic acid;
4- { 1 - [2-chloro-6-(trifluoromethyl)benzyl] -4-fluoro- 1 H-indazol-3 -yl } benzoic acid; 4- { 1 - [2-chloro-6-(trifluoromethyl)benzyl] -4-fluoro- 1 H-indazol-3 -yl } -2, 5 - difluorobenzoic acid;
4-( 1 - { 1 -[2-chloro-6-(trifluoromethyl)phenyl] ethyl } -4-fluoro- 1 H-indazol-3 -yl)-3 - fluorobenzoic acid;
4-( 1 - { ( 1 R or 1 S) - 1 - [2-chloro-6-(trifluoromethyl)phenyl] ethyl } -4-fluoro- 1 H-indazol-3 - yl)-3 -fluorobenzoic acid;
4-( 1 - { ( 1 S or 1 R) - 1 - [2-chloro-6-(trifluoromethyl)phenyl] ethyl } -4-fluoro- 1 H-indazol-3 - yl)-3 -fluorobenzoic acid;
4- [ 1 -(2-bromo-3 -fluorobenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl] -3 -fluorobenzoic acid; 4- [ 1 -(5-chloro-2-cyanobenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl] -3 -fluorobenzoic acid; 3 -fluoro-4-( 1 - { 1 - [2-(trifluoromethyl)phenyl] ethyl } - 1 H-pyrazolo [4,3 -b]pyridin-3 - yl)benzoic acid;
4- [ 1 -(6-chloro-2-fluoro-3 -methylbenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl] -3 - fluorobenzoic acid;
4- [ 1 -(2-chloro-3 , 6-difluorobenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl] -3 -fluorobenzoic acid;
3 - fluoro-4- [ 1 -(2, 3 , 6-trifluorobenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl]benzoic acid; 3 -fluoro-4- { 1 -[2-fluoro-6-(trifluoromethyl)benzyl] - 1 H-pyrazolo [4,3 -b]pyridin-3 - yl}benzoic acid;
4- [ 1 -(2, 6-difluorobenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl] -3 -fluorobenzoic acid; 4- [ 1 -(2-chloro-6-fluorobenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl] -3 -fluorobenzoic acid; 4- [ 1 -(6-chloro-2-fluoro-3 -methoxybenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl] -3 - fluorobenzoic acid;
4- [ 1 -(2-chloro-6-fluoro-3 -methoxybenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl] -3 - fluorobenzoic acid;
4- [ 1 -(2, 3 -dichloro-6-fluorobenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl] -3 -fluorobenzoic acid;
4- [ 1 -( 1 -benzothiophen-7-ylmethyl)- 1 H-pyrazolo [4, 3 -b]pyridin-3 -yl] -3 -fluorobenzoic acid;
4- { 1 - [2, 6-dichloro-3 -(trifluoromethyl)benzyl] - 1 H-pyrazolo [4,3 -b]pyridin-3 -yl } -3 - fluorobenzoic acid;
4- [ 1 -(3 , 6-dichloro-2-fluorobenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl] -3 -fluorobenzoic acid;
4- { 1 - [2-chloro-6-(methoxycarbonyl)benzyl] - 1 H-pyrazolo [4,3 -b]pyridin-3 -yl } -3 - fluorobenzoic acid;
4- [ 1 -(2-bromo-6-chlorobenzyl)- 1 H-pyrazolo [4,3 -b]pyridin-3 -yl] -3 -fluorobenzoic acid; 4- { 1 - [2-chloro-6-(trifluoromethyl)benzyl] - 1 H-pyrazolo [4,3 -b]pyridin-3 -yl } -3 - fluorobenzoic acid;
4- { 1 - [2-chloro-6-(trifluoromethyl)benzyl] - 1 H-pyrrolo [3 ,2-b]pyridin-3 -yl } -3 - fluorobenzoic acid; and
4- { 1 - [2-chloro-6-(trifluoromethyl)benzyl] - 1 H-pyrrolo [2,3 -c]pyridin-3 -yl } -3 - fluorobenzoic acid.
20. A pharmaceutical composition comprising a compound of claim 1 or a
pharmaceutically acceptable salt or solvate thereof and one or more pharmaceutically acceptable excipients.
21. The pharmaceutical composition of claim 20, which further comprises at least one additional therapeutically active agent. 22. 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).
23. 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.
24. The method of claim 23, wherein the disease or condition is an autoimmune disease or inflammatory disease.
25. The method of claim 24, 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/CN2012/080134 2012-08-15 2012-08-15 N-alkylated indole and indazole compounds as rorgammat inhibitors and uses thereof WO2014026329A1 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
PCT/CN2012/080134 WO2014026329A1 (en) 2012-08-15 2012-08-15 N-alkylated indole and indazole compounds as rorgammat inhibitors and uses thereof
CA2881693A CA2881693A1 (en) 2012-08-15 2013-08-14 N-alkylated indole and indazole compounds as rorgammat inhibitors and uses thereof
US14/421,052 US9556168B2 (en) 2012-08-15 2013-08-14 N-alkylated indole and indazole compounds as RORgammaT inhibitors and uses thereof
PCT/US2013/054893 WO2014028591A2 (en) 2012-08-15 2013-08-14 N-ALKYLATED INDOLE AND INDAZOLE COMPOUNDS AS RORgammaT INHIBITORS AND USES THEREOF
RU2015108935A RU2015108935A (en) 2012-08-15 2013-08-14 Compounds of N-Alkylated Indoles and Indazoles as RorgammaT Inhibitors and Methods of Their Use
CN201380053436.0A CN104822377A (en) 2012-08-15 2013-08-14 N-alkylated indole and indazole compounds as RORgammat inhibitors and uses thereof
JP2015527567A JP6411345B2 (en) 2012-08-15 2013-08-14 N-alkylated indole and indazole compounds as RORγT inhibitors and their use
EP13829125.7A EP2884977B1 (en) 2012-08-15 2013-08-14 N-ALKYLATED INDOLE AND INDAZOLE COMPOUNDS AS RORgammaT INHIBITORS AND USES THEREOF
BR112015003397A BR112015003397A2 (en) 2012-08-15 2013-08-14 compound, pharmaceutical composition, use of the compound and method for treating a rorgammat-mediated disease or condition in a patient.
AU2013302727A AU2013302727A1 (en) 2012-08-15 2013-08-14 N-alkylated indole and indazole compounds as RORgammaT inhibitors and uses thereof
KR20157006321A KR20150041659A (en) 2012-08-15 2013-08-14 N-ALKYLATED INDOLE AND INDAZOLE COMPOUNDS AS RORgammaT INHIBITORS AND USES THEREOF

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2012/080134 WO2014026329A1 (en) 2012-08-15 2012-08-15 N-alkylated indole and indazole compounds as rorgammat inhibitors and uses thereof

Publications (1)

Publication Number Publication Date
WO2014026329A1 true WO2014026329A1 (en) 2014-02-20

Family

ID=50101187

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/CN2012/080134 WO2014026329A1 (en) 2012-08-15 2012-08-15 N-alkylated indole and indazole compounds as rorgammat inhibitors and uses thereof
PCT/US2013/054893 WO2014028591A2 (en) 2012-08-15 2013-08-14 N-ALKYLATED INDOLE AND INDAZOLE COMPOUNDS AS RORgammaT INHIBITORS AND USES THEREOF

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/US2013/054893 WO2014028591A2 (en) 2012-08-15 2013-08-14 N-ALKYLATED INDOLE AND INDAZOLE COMPOUNDS AS RORgammaT INHIBITORS AND USES THEREOF

Country Status (9)

Country Link
US (1) US9556168B2 (en)
EP (1) EP2884977B1 (en)
JP (1) JP6411345B2 (en)
KR (1) KR20150041659A (en)
AU (1) AU2013302727A1 (en)
BR (1) BR112015003397A2 (en)
CA (1) CA2881693A1 (en)
RU (1) RU2015108935A (en)
WO (2) WO2014026329A1 (en)

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9266886B2 (en) 2014-02-03 2016-02-23 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
WO2016110821A1 (en) 2015-01-08 2016-07-14 Advinus Therapeutics Limited Bicyclic compounds, compositions and medicinal applications thereof
WO2016120850A1 (en) * 2015-01-30 2016-08-04 Pfizer Inc. Sulfonamide-subsituted indole modulators of rorc2 and methods of use thereof
WO2016128908A1 (en) 2015-02-12 2016-08-18 Advinus Therapeutics Limited Bicyclic compounds, compositions and medicinal applications thereof
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
US9663515B2 (en) 2014-11-05 2017-05-30 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US9796710B2 (en) 2014-10-14 2017-10-24 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US9845308B2 (en) 2014-11-05 2017-12-19 Vitae Pharmaceuticals, Inc. Isoindoline inhibitors of ROR-gamma
CN108586432A (en) * 2018-05-31 2018-09-28 温州医科大学 A kind of 3-(Indoles -5- bases)Indazole derivative and its application
US20190062328A1 (en) * 2016-01-21 2019-02-28 Zibo Biopolar Changsheng Pharmaceutical Co., Ltd. Bruton's tyrosine kinase inhibitors
US10287272B2 (en) 2015-10-27 2019-05-14 Merck Sharp & Dohme Corp. Substituted indazole 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
US10344000B2 (en) 2015-10-27 2019-07-09 Merck Sharp & Dohme Corp. Substituted bicyclic pyrazole compounds as RORgammaT 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
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
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

Families Citing this family (13)

* 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
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
WO2014026330A1 (en) 2012-08-15 2014-02-20 Merck Sharp & Dohme Corp. 3-AMINOCYCLOALKYL COMPOUNDS AS RORgammaT INHIBITORS AND USES THEREOF
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
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
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
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
KR20190078646A (en) 2016-11-18 2019-07-04 머크 샤프 앤드 돔 코포레이션 Indazole derivatives useful as inhibitors of diacylglyceride O-acyltransferase 2
CA3056501A1 (en) * 2017-04-11 2018-10-18 Sunshine Lake Pharma Co., Ltd. Fluorine-substituted indazole compounds and uses thereof

Citations (5)

* 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
US5958683A (en) * 1994-03-30 1999-09-28 Novartis Corporation Screening method using the RZR receptor family
CN101087784A (en) * 2004-09-21 2007-12-12 阿特西斯公司 Indole acetic acids exhibiting CRTH2 receptor antagonism and uses thereof
US7427600B2 (en) * 2000-08-22 2008-09-23 Shire Llc Active agent delivery systems and methods for protecting and administering active agents
WO2011055270A1 (en) * 2009-11-04 2011-05-12 Wyeth Llc Indole based receptor crth2 antagonists

Family Cites Families (35)

* 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
US6387942B2 (en) * 2000-06-19 2002-05-14 Yung Shin Pharmaceutical Ind. Co. Ltd Method of treating disorders related to protease-activated receptors-induced cell activation
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
KR20060043654A (en) * 2004-03-15 2006-05-15 융신 파마슈티칼 인더스트리얼 컴파니 리미티드 Preferential inhibition of release of pro-inflammatory cytokines
US7592363B2 (en) 2004-08-03 2009-09-22 Wyeth Indazoles
CA2583428A1 (en) 2004-09-03 2006-03-09 Plexxikon, Inc. Bicyclic heteroaryl pde4b inhibitors
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
US20090233955A1 (en) 2004-12-08 2009-09-17 Frazee James S 1H-Pyrrolo[2,3-B]Pyridnes
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
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
EP2346508B1 (en) * 2008-09-26 2016-08-24 Intellikine, LLC Heterocyclic kinase inhibitors
EP2181710A1 (en) 2008-10-28 2010-05-05 Phenex Pharmaceuticals AG Ligands for modulation of orphan receptor-gamma (NR1F3) activity
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
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
EP2487159A1 (en) 2011-02-11 2012-08-15 MSD Oss B.V. RorgammaT inhibitors
JP2014166961A (en) 2011-06-20 2014-09-11 Dainippon Sumitomo Pharma Co Ltd Novel indazole derivative
WO2014026330A1 (en) 2012-08-15 2014-02-20 Merck Sharp & Dohme Corp. 3-AMINOCYCLOALKYL 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

Patent Citations (5)

* 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
US5958683A (en) * 1994-03-30 1999-09-28 Novartis Corporation Screening method using the RZR receptor family
US7427600B2 (en) * 2000-08-22 2008-09-23 Shire Llc Active agent delivery systems and methods for protecting and administering active agents
CN101087784A (en) * 2004-09-21 2007-12-12 阿特西斯公司 Indole acetic acids exhibiting CRTH2 receptor antagonism and uses thereof
WO2011055270A1 (en) * 2009-11-04 2011-05-12 Wyeth Llc Indole based receptor crth2 antagonists

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
ARMELLE MELET ET AL.: "ANALYSIS OF HUMAN CYTOCHROME P450 2C8 SUBSTRATE SPECIFICITY USING A SUBSTRATE PHARMACOPHORE AND SITE-DIRECTED MUTANTS", BIOCHEMISTRY, vol. 43, 2004, pages 15379 - 15392 *
VOLKER FISCHER ET AL.: "THE 3-HYDROXY 3-METHYLGLUTARYL COENZYME A DUCTASE INHIBITOR FLUVASTATIN: EFFECT ON HUMAN CYTOCHROME P-450 AND IMPLICATIONS FOR METABOLIC DRUG INTERACTIONS", DRUG METABOLISM AND DISPOSITION, vol. 27, no. 3, 1998, pages 410 - 416, XP000997302 *

Cited By (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10047085B2 (en) 2014-02-03 2018-08-14 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US10807980B2 (en) 2014-02-03 2020-10-20 Vitae Pharmaceuticals, Llc Dihydropyrrolopyridine inhibitors of ROR-gamma
US9266886B2 (en) 2014-02-03 2016-02-23 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US10399976B2 (en) 2014-02-03 2019-09-03 Vitae Pharmaceuticals, Llc 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
US9796710B2 (en) 2014-10-14 2017-10-24 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US10087184B2 (en) 2014-10-14 2018-10-02 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of RORγ
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
US9663515B2 (en) 2014-11-05 2017-05-30 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
US9845308B2 (en) 2014-11-05 2017-12-19 Vitae Pharmaceuticals, Inc. Isoindoline inhibitors of ROR-gamma
US11001583B2 (en) 2014-11-05 2021-05-11 Vitae Pharmaceuticals, Llc Dihydropyrrolopyridine inhibitors of ROR-gamma
WO2016110821A1 (en) 2015-01-08 2016-07-14 Advinus Therapeutics Limited Bicyclic compounds, compositions and medicinal applications thereof
JP2018510131A (en) * 2015-01-30 2018-04-12 ファイザー・インク RORC2 sulfonamide substituted indole modulator and method of use thereof
US10385036B2 (en) 2015-01-30 2019-08-20 Pfizer Inc. Sulfonamide-substituted indole modulators of RORC2 and methods of use thereof
WO2016120850A1 (en) * 2015-01-30 2016-08-04 Pfizer Inc. Sulfonamide-subsituted indole modulators of rorc2 and methods of use thereof
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
WO2016128908A1 (en) 2015-02-12 2016-08-18 Advinus Therapeutics Limited Bicyclic compounds, compositions and medicinal applications thereof
US10829448B2 (en) 2015-08-05 2020-11-10 Vitae Pharmaceuticals, Llc Substituted benzoimidazoles as modulators of ROR-γ
US10301261B2 (en) 2015-08-05 2019-05-28 Vitae Pharmaceuticals, Llc Substituted indoles as modulators of ROR-gamma
US10287272B2 (en) 2015-10-27 2019-05-14 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
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
US20190062328A1 (en) * 2016-01-21 2019-02-28 Zibo Biopolar Changsheng Pharmaceutical Co., Ltd. Bruton's tyrosine kinase inhibitors
US10662187B2 (en) * 2016-01-21 2020-05-26 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
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
CN108586432A (en) * 2018-05-31 2018-09-28 温州医科大学 A kind of 3-(Indoles -5- bases)Indazole derivative and its application

Also Published As

Publication number Publication date
BR112015003397A2 (en) 2019-09-24
WO2014028591A3 (en) 2014-04-17
AU2013302727A1 (en) 2015-03-05
JP6411345B2 (en) 2018-10-24
CA2881693A1 (en) 2014-02-20
US9556168B2 (en) 2017-01-31
RU2015108935A (en) 2016-10-10
US20150210687A1 (en) 2015-07-30
KR20150041659A (en) 2015-04-16
EP2884977A4 (en) 2016-05-11
WO2014028591A2 (en) 2014-02-20
JP2015526442A (en) 2015-09-10
EP2884977A2 (en) 2015-06-24
EP2884977B1 (en) 2017-10-11

Similar Documents

Publication Publication Date Title
WO2014026329A1 (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
WO2014026328A1 (en) 3-cyclohexenyl substituted indole and indazole compounds as rorgammat inhibitors and uses thereof
EP2683693B1 (en) Rorgammat inhibitors
AU2016219183B2 (en) Substituted pyrazole compounds as RORgammaT inhibitors and uses 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: 12883070

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: 12883070

Country of ref document: EP

Kind code of ref document: A1