WO2014181287A1 - Heterocyclyl compounds and uses thereof - Google Patents

Heterocyclyl compounds and uses thereof Download PDF

Info

Publication number
WO2014181287A1
WO2014181287A1 PCT/IB2014/061299 IB2014061299W WO2014181287A1 WO 2014181287 A1 WO2014181287 A1 WO 2014181287A1 IB 2014061299 W IB2014061299 W IB 2014061299W WO 2014181287 A1 WO2014181287 A1 WO 2014181287A1
Authority
WO
WIPO (PCT)
Prior art keywords
phenyl
alkyl
cyano
urea
triazol
Prior art date
Application number
PCT/IB2014/061299
Other languages
French (fr)
Inventor
Rajiv Sharma
Arunkumar JAIN
Bichismita Sahu
Deepak Singh
Sunil MALI
Original Assignee
Piramal Enterprises Limited
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 Piramal Enterprises Limited filed Critical Piramal Enterprises Limited
Publication of WO2014181287A1 publication Critical patent/WO2014181287A1/en

Links

Classifications

    • 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
    • A61K31/41641,3-Diazoles
    • 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
    • A61K31/41921,2,3-Triazoles
    • 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
    • A61K31/41961,2,4-Triazoles
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/4261,3-Thiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/42One nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D249/00Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms
    • C07D249/02Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D249/041,2,3-Triazoles; Hydrogenated 1,2,3-triazoles
    • C07D249/061,2,3-Triazoles; Hydrogenated 1,2,3-triazoles with aryl radicals directly attached to ring atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D261/00Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings
    • C07D261/02Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings
    • C07D261/06Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings having two or more double bonds between ring members or between ring members and non-ring members
    • C07D261/08Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings having two or more double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D261/00Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings
    • C07D261/02Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings
    • C07D261/06Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings having two or more double bonds between ring members or between ring members and non-ring members
    • C07D261/10Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings having two or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D261/14Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • 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
    • C07D403/12Heterocyclic 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 chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/107Spiro-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring

Definitions

  • the present invention relates to heterocyclyl compounds (the compounds of formula I as described herein), processes for their preparation, pharmaceutical compositions containing the said compounds, their use as Interleukin 17 (IL-1 7) inhibitors or Tumor Necrosis Factor alpha (TNF-a) inhibitors, and methods for using said compounds in the treatment or prevention of diseases or disorders mediated by IL-17 and TNF-a, such as autoimmune or inflammatory diseases or disorders; and metabolic diseases or disorders.
  • IL-1 Interleukin 17
  • TNF-a Tumor Necrosis Factor alpha
  • Inflammation is part of the non-specific immune response that occurs in reaction to body injury (e.g. trauma, ischemia, and foreign particles) or infection (e.g. bacterial or viral infection). Inflammation is an inherent part of various autoimmune or inflammatory disorders, such as rheumatoid arthritis, atherosclerosis, inflammatory myopathies, septic shock syndrome, ulcerative colitis, inflammatory bowel disease, psoriasis, multiple sclerosis, and others. It has been reported that inflammation is commonly associated with the metabolic diseases and disorders such as insulin resistance and obesity (Brent E. Wisse, Journal of the American Society of Nephrology, 2004, 15, 2792-2800). Inflammation can be classified as either acute or chronic.
  • Th 7 cells are involved in mediating inflammatory diseases and immune defense against extracellular bacteria.
  • Th 17 cells are a subset of T cells that produce pro-inflammatory cytokines such as the interleukins (IL-6, IL-17A, IL-17F, IL-21 , IL-22), and TNF-a (Tumor necrosis factor-alpha).
  • Th 7 cells can be generated from naive precursors in the presence of TGF- ⁇ and IL-6 or IL-21 , which can be further augmented by TNF or I L-1 ⁇ which are the inflammatory cytokines that inhibit Th1 and Th2 cell differentiation.
  • TGF- ⁇ and IL-6 or IL-21 TGF- ⁇ and IL-6 or IL-21
  • TNF or I L-1 ⁇ TNF or I L-1 ⁇ which are the inflammatory cytokines that inhibit Th1 and Th2 cell differentiation.
  • T reg cells are specified by transcription factors such as Tbet, GATA3, and Foxp3, respectively, whereas that of Th-
  • the pro-inflammatory cytokine, Interleukin 17 (family) comprises six members namely IL-17A, IL-17B, IL-17C, IL-17D, IL-17E, and IL-1 7F, out of which IL-1 7A and IL-1 7F are clearly associated with inflammation as these are produced by Th 7 cells.
  • IL-17 deploys neutrophils to the site of inflammation and this ability imparts IL-17 with a key role in several types of diseases.
  • IL-1 7 is predominantly produced by activated memory T cells and acts on the cell by binding to a specific cell surface receptor IL-1 7R.
  • Human IL-1 7 is a 20-30 kDa glycoprotein configured with 155 amino acids, comprising a signal peptide at the N-end.
  • IL-17 has been found to be elevated in patients with rheumatoid arthritis, systemic lupus erythematosus (SLE), Behget's disease, inflammatory bowel disease, asthma, multiple sclerosis, and other related diseases or disorders. It has further been reported that IL-1 7 plays a pivotal role in the obesity induced inflammatory conditions (Mario Galgani et al., Journal of Leukocyte Biology, 201 0, 87, 17-1 8).
  • Th 7 cells which stimulate the secretion of a wide range of pro-inflammatory cytokines including IL-17A and TNF-oc among others, appear to depend on the retinoid-related orphan receptor RORgt. Accordingly, transcription factor 'Retinoid-related orphan receptor gamma t (RORgt)' plays a pivotal role in the production of IL-17.
  • RORgt is a member of steroid hormone nuclear receptor superfamily and it is encoded by ROR c gene. RORgt is exclusively expressed in the T cells of the immune system. It is known that RORgt controls differentiation of proinflammatory T helper 17 (Th 17 ) cells.
  • Th17-dependent autoimmune disease It is reported that RORgt is required for the induction of IL-1 7 transcription and for the manifestation of Th17-dependent autoimmune disease. Multiple other transcription factors have also been shown to be important for the development of Th1 7 cells: BATF, STAT3, IRF4, RUNX1 , and ⁇ . Unlike RORgt, these factors are not NHRs (Nuclear hormone receptors) and thus do not contain ligand binding domains, which renders them less attractive targets for drug development.
  • Tumor Necrosis Factor-alpha (TNF-a) / cachectin is a pleiotrophic cytokine that plays pivotal role in inflammatory disorders and conditions. TNF-a is a soluble homotrimer of 17 kD protein subunits.
  • TNF-a is produced by monocytes, macrophages, and T lymphocytes. It shows self-regulatory, growth stimulating and growth inhibitory properties. Following to infection or injury, TNF-a initiates inflammatory cascade of cytokines, thereby recruiting macrophage and neutrophils to a site of infection. It activates granulocytes, and increases Major Histocompatibility Complex class II (MHC Class II) expression. It also promotes secretion of matrix metalloproteinases (MMPs), leading to cartilage matrix degradation. Because TNF-a initiates an inflammatory cascade, and has been found to be increased in close proximity to inflamed or injured tissue, TNF-a inhibition is an appealing target for the development of new antiinflammatory drugs.
  • MHC Class II Major Histocompatibility Complex class II
  • Treatments for autoimmune and inflammatory disorders vary depending on the particular medical disorder, and often involve use of immunosuppressive drugs. In view of this, there is a continuing need to provide newer and effective treatment options to the patients suffering from autoimmune, inflammatory and related disorders.
  • the present invention addresses the need of newer and effective treatment options to the patients suffering from autoimmune, inflammatory and related disorders by providing compounds that are IL-17 and/or TNF-a inhibitors.
  • the present invention relates to a compound of formula I (as described herein), or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, S-oxide or N-oxide thereof.
  • processes for the preparation of the compounds of formula I, or pharmaceutically acceptable salts thereof are provided.
  • a pharmaceutical composition comprising one or more of the compounds of formula I or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof; and at least one pharmaceutically acceptable carrier or excipient.
  • a compound of formula I or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof; for use as TNF-a inhibitor for use as IL-17 inhibitor.
  • a compound of formula I or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof for use in the treatment of a disease or a disorder mediated by IL-1 7 or TNF-a.
  • a method for the treatment of a disease or disorder mediated by IL-1 7 or TNF-a comprising administering to a subject in need thereof; a therapeutically effective amount of the compound of formula I or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof.
  • a compound of formula I or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof for the manufacture of a medicament for the treatment of a disease or a disorder mediated by IL-1 7 or TNF-a.
  • the present invention relates to use of a compound of formula I or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt or a solvate thereof; in combination with at least one further therapeutically active agent for the treatment of a disease or a disorder mediated by IL-1 7 or TNF-a.
  • the present invention relates to a compound of formula I:
  • A is triazole, pyrazole, imidazole, oxazole, isoxazole, thiazole, pyrrole, a 5- or 6- membered saturated heterocyclic ring system or a heteroaryl ring system selected from:
  • Ri and R3 are independently selected from the group consisting of hydrogen, hydroxy, halogen, cyano, nitro, (CrC 6 )-alkyl, (CrC 6 )-alkoxy, (CrC 6 )-alkoxyalkyl, (CrC 6 )- alkylamino, (C 3 -Ci 2 )-cycloalkyl, 0-(C 3 -Ci 2 )-cycloalkyl, halo(CrC 6 )alkyl, halo(d- C 6 )alkoxy, (C 6 -C 14 )-aryl, heterocyclyl, 0-(C 6 -Ci 4 )aryl, (C 6 -Ci 4 )ar(C C 6 )alkyl, (C 6 - Ci 4 )ar(C C 6 )alkyloxy, heteroaryl, C(0)R a , C(0)OR a , S(0) n R
  • R 2 is (C r C 6 )-alkyl, (C 1 -C 6 )-alkoxy, (C Ce lkoxyid-C ⁇ - lkyl, (C 3 -C 12 )-cycloalkyl, O- (C 3 -C 12 )-cycloalkyl, (C 6 -Ci 4 )-aryl, 0-(C 6 -C 14 )aryl, (C 6 -Ci 4 )ar(CrC 6 )alkyl, (C 6 -Ci 4 )ar(C C 6 )alkyloxy, heterocyclyl or heteroaryl;
  • R 2 together with R can form a 5- or 6- membered saturated or unsaturated ring optionally containing 1 to 3 heteroatoms independently selected from the group consisting of O, N and S, which ring is unsubstituted or substituted with 1 to 5 groups independently selected from the group consisting of halogen, hydroxy, nitro, cyano, (d- C 6 )-alkyl, (C C 6 )-alkoxy, (C C 6 )-alkoxyalkyl, (CrC 6 )-alkylamino, (C 3 -Ci 2 )-cycloalkyl, O- (C 3 -C 12 )-cycloalkyl, halo(C C 6 )alkyl, halo(C C 6 )alkoxy, (C 6 -C 14 )-aryl, 0-(C 6 -Ci 4 )aryl, (C 6 -Ci 4 )ar(Ci-C 6 )alkyl, (C 6 -
  • L is * N(R 4 )-C(0)-N(R 5 ), * C(0)N(R 5 ), * N(R 4 )-C(0), * CH(R 6 )-C(0)-N(R 5 ), * CH(R 6 )N(R 5 ), * N(R 4 )-S(0) 2 or * NR 4 ; wherein * indicates the point of attachment to A;
  • R , R 5 and R 6 are independently selected from the group consisting of hydrogen, hydroxy, cyano, (Ci-C6)-alkyl, (Ci-C6)-alkoxy, (Ci-C6)-alkoxyalkyl, (Ci-C6)-alkylamino, (C 3 -Ci 2 )-cycloalkyl, 0-(C 3 -Ci 2 )-cycloalkyl, halo(C C 6 )alkyl, halo(C C 6 )alkoxy, hydroxy(Ci-C 6 )-alkyl, C(0)R a , C(0)OR a , S(0) n R a , P(0)n(0(Ci-C 6 )alkyl) 2 , O-P(O) (0(C C 6 )alkyl) 2 and C(0)NR a R b ;
  • Xi , X 2 , X 4 and X 5 are independently selected from N and C(R 7 ); wherein R 7 is hydrogen, halogen, hydroxy, nitro, cyano, (CrC 6 )-alkyl, (CrC 6 )-alkoxy, (d-C 6 )- alkoxy(CrC 6 )-alkyl, (Ci-C 6 )-alkylamino, (C 3 -Ci 2 )-cycloalkyl, halo(C C 6 )alkyl, halo(C C 6 )alkoxy, (C 2 -C 6 )-alkenyl, (C 2 -C 6 )-alkynyl, (C 6 -Ci 4 )-aryl, 0-(C 6 -Ci 4 )aryl, (C 6 -Ci 4 )ar(C C 6 )alkyl, (C 6 -Ci 4 )ar(Ci-C 6
  • X 3 is N or C(R 8 ), wherein R 8 is hydrogen, halogen, hydroxy, cyano, nitro, (CrC 6 )-alkyl, (Ci -C 6 )-alkoxy, (Ci -C 6 )-alkoxy(Ci -C 6 )alkyl, (C C 6 )-alkylamino, (C 3 -C 12 )-cycloalkyl, O- (C 3 -Ci 2 )-cycloalkyl, halo(C C 6 )alkyl, halo(C C 6 )alkoxy, (C 6 -Ci 4 )-aryl, 0-(C 6 -Ci 4 )aryl, (C6-Ci 4 )ar(CrC6)alkyl, (C6-Ci 4 )ar(CrC6)alkyloxy, heterocyclyl, O-heterocyclyl, heteroaryl, C(0)R a , C(0)
  • R 7 groups on the two adjacent carbon atoms can form a 3-7 membered unsaturated or saturated fused ring optionally containing 1 to 3 heteroatoms independently selected from the group consisting of O, N and S; wherein the ring can be unsubstituted or substituted with 1 -3 groups of R 0 ; or
  • R 7 group together with the R 8 group can form a 3-7 membered fused ring optionally containing 1 to 3 heteroatoms selected from O, N or S; wherein the ring can be unsubstituted or substituted with 1 -3 groups of R c ;
  • R a and R b are independently selected from the group consisting of hydrogen, (CrC 6 )- alkyl, (C 3 -C 12 )-cycloalkyl, halo(C C 6 )alkyl, (C 6 -C 14 )-aryl, (C 6 -Ci4)ar(C C6)alk l, heterocyclyl, heteroaryl, and S(0) n R a ; or
  • R a and R b can form a 3-7 membered saturated or unsaturated ring optionally containing one or more heteroatoms selected from N, O or S, which is unsubstituted or substituted with one or more groups independently selected from the group consisting of halogen, hydroxy, cyano, nitro, (C C 6 )-alkyl, (CrC 6 )-alkoxy, (CrC 6 )-alkylamino, (C 3 - Ci 2 )-cycloalkyl, 0-(C 3 -Ci 2 )-cycloalkyl, halo(C r C 6 )alkyl, halo(C r C 6 )alkoxy, (C 6 -C )-aryl, 0-(C 6 -C )aryl, (C 6 -Ci 4 )ar(Ci-C 6 )alkyl, heterocyclyl, heteroaryl, C(0)R a , C(0)OR a , S
  • -C 6 )-alkoxy can be unsubstituted or substituted with one or more groups independently selected from the group consisting of halogen, hydroxy, nitro, cyano, (Ci-C 6 )-alkoxy-(R c )o-3, (C 3 -Ci 2 )-cycloalkyl-(R c )o- 3!
  • R c is halogen, hydroxy, cyano, nitro, (Ci-C 6 )-alkyl-(R c )o-i , (Ci-C 6 )-alkoxy-(R c )o-i , (C 3 - Ci 2 )-cycloalkyl-(R c )o-i , O-(C 3 -Ci 2 )-cycloalkyl-(R c ) 0 -i , hydroxy(Ci-C 6 )-alkyl-(R c )o-i , halo(CrC 6 )alkyl, halo(C C 6 )alkoxy, (C 6 -Ci 4 )-aryl-(R c ) 0 -i, O-(C 6 -Ci 4 )aryl-(R c ) 0 -i, (C 6 - Ci 4 )ar(CrC 6 )alkyl-(R
  • substitution means that one or more hydrogens of the specified moiety are replaced with a suitable substituent and includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and results in a stable compound.
  • (C-i-C 6 )-alkyr or "alkyl” refers to the radical of saturated aliphatic groups, including straight or branched-chain alkyl groups.
  • An alkyl group can have a straight chain or branched chain containing carbon atoms.
  • a straight-chain or branched chain alkyl has six or fewer carbon atoms in its backbone, for instance, CrC 6 for straight chain and C 3 -C 6 for branched chain.
  • Suitable alkyl groups containing from one to six carbon atoms include methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, iso-butyl, sec-butyl, n-pentyl, isopentyl, 2-pentyl, 3-pentyl, neo-pentyl, n-hexyl, isohexyl, 2-hexyl and 3- hexyl.
  • alkyl groups may be unsubstituted or substituted with one or more substituents.
  • a substituted alkyl refers to a (CrC 6 )-alkyl substituted with one or more groups, preferably 1 -3 groups independently selected from the group consisting of halogen, hydroxy, nitro, cyano, (C-i-C 6 )-alkoxy-(R c )o-3, (C 3 -C 12 )- cycloalkyl-(R c )o-3, 0-(C 3 -Ci 2 )-cycloalkyl-(R c )o-3, halo(C C 6 )alkyl, halo(C C 6 )alkoxy, (C 6 - Ci4)-aryl-(R 0 )o-3, 0-(C 6 -Ci 4 )aryl-(R c )o-3, (C 6 -Ci 4 )ar(Ci-C 6 )
  • R a and R b are independently selected from the group consisting of hydrogen, (C C 6 )-alkyl, (C 3 -Ci 2 )-cycloalkyl, halo(C C 6 )alkyl, (C 6 -Ci 4 )-aryl, (C 6 - Ci 4 )ar(CrC 6 )alkyl, heterocyclyl, heteroaryl, and S(0) n R a ; or
  • R a and R b can form a 3-7 membered saturated or unsaturated ring optionally containing one or more heteroatoms selected from N, O or S, which is unsubstituted or substituted with one or more groups independently selected from the group consisting of halogen, hydroxy, cyano, nitro, (Ci-C6)-alkyl, (Ci-C6)-alkoxy, (Ci-C6)-alkylamino, (C 3 -Ci 2 )- cycloalkyl, 0-(C 3 -Ci 2 )-cycloalkyl, halo(C C 6 )alkyl, halo(C C 6 )alkoxy, (C 6 -C 14 )-aryl, O- (C 6 -C 14 )aryl, (C 6 -Ci 4 )ar(C C 6 )alkyl, heterocyclyl, heteroaryl, C(0)R a , C(0)OR a , S(0) n Ra,
  • R c is halogen, hydroxy, cyano, nitro, (C C 6 )-alkyl-(R c ) 0 -i, (CrC 6 )-alkoxy-(R c )o-i, (C 3 - Ci 2 )-cycloalkyl-(R c )o-i, O-(C 3 -Ci 2 )-cycloalkyl-(R c ) 0 -i, hydroxy(C r C 6 )-alkyl-(R c )o-i, halo(CrC 6 )alkyl, halo(C C 6 )alkoxy, (C 6 -Ci 4 )-aryl-(R c ) 0 -i, O-(C 6 -Ci 4 )aryl-(R c ) 0 -i, (C 6 - Ci4)ar(Ci-C 6 )alkyl-(R c )o-i
  • substituted alkyls include, but are not limited to, chloromethyl, hydroxymethyl, hydroxyethyl, aminoethyl and the like.
  • halogen refers to a fluorine, chlorine, bromine, or iodine atom.
  • halo(Ci-C 6 )alkyr When the alkyl group is substituted with one or more halogens, it is specifically referred to as "halo(Ci-C 6 )alkyr or "haloalkyl".
  • a monohalo(Ci-C 6 )alkyl radical for example, may have a chlorine, bromine, iodine or fluorine atom.
  • Dihalo and polyhalo(CrC6)alkyl radicals may have two or more of the same or different halogen atoms.
  • halo(CrC 6 )alkyl examples include, but are not limited to, chloromethyl, dichloromethyl, trichloromethyl, dichloroethyl, dichloropropyl, fluoromethyl, difluoromethyl, trifluoromethyl, pentafluoroethyl, heptafluoropropyl, difluorochloromethyl, dichlorofluoromethyl, difluoroethyl, difluoropropyl or the like groups.
  • halo(d-C6)alkoxy or "haloalkoxy” refers to radicals wherein one or more of the hydrogen atoms of the alkoxy group are substituted with one or more halogens.
  • haloalkoxy or “halo(CrC 6 )alkoxy” groups include, but not limited to, difluoromethoxy (OCHF 2 ), trifluoromethoxy (OCF 3 ) or trifluoroethoxy (OCH 2 CF 3 ).
  • (C 2 -C 6 )-alkenyl refers to an alkenyl group having 2 to 6 (both inclusive) carbon atoms.
  • the geometry of the double bond may be
  • E), or sixteen (Z), cis or trans examples of alkenyl include, but are not limited to, vinyl, allyl and 2-propenyl.
  • the alkenyl groups can be unsubstituted or substituted with one or more groups, preferably 1 -3 groups independently selected from the group consisting of halogen, hydroxy, nitro, cyano, (CrC6)alkyl-(R c )o-3, (Ci-C6)-alkoxy-(R c )o-3, (C 3 -C 12 )-cycloalkyl-(R c )o- 3 , 0-(C 3 -Ci 2 )-cycloalkyl-(R c )o- 3 , halo(C r C 6 )alkyl, halo(C C 6 )alkoxy, (C 6 -C 14 )-aryl-(R c ) 0 - 3 , O-(C 6 -C 14 )aryl-(R c ) 0 - 3 , (C 6 -C 14 )ar(C C 6 )alkyl-(R
  • alkynyl or "(C 2 -C6)alkynyl” whether used alone or as part of a substituent group, refers to a straight or branched chain hydrocarbon radical containing the indicated number of carbon atoms and at least one carbon-carbon triple bond (two adjacent sp carbon atoms).
  • (C 2 -C 6 )alkynyl refers to an alkynyl group having 2 to 6 (both inclusive) carbon atoms.
  • Examples of (C 2 -Ce)alkynyl include, but are not limited to, ethynyl, 1 -propynyl, 3-propynyl and 3-butynyl.
  • alkynyl can be unsubstituted or substituted with one or more groups, preferably 1 - 3 groups independently selected from the group consisting of halogen, hydroxy, nitro, cyano, (Ci-C 6 )alkyl-(R c ) 0 - 3 , (Ci-C 6 )-alkoxy-(R c )o-3, (C 3 -Ci 2 )-cycloalkyl-(R c ) 0 - 3 , 0-(C 3 -Ci 2 )- cycloalkyl-(R c )o-3, halo(C C 6 )alkyl, halo(C C 6 )alkoxy, (C 6 -Ci 4 )-aryl-(R c )o- 3 , 0-(C 6 - Ci 4 )aryl-(R c )o-3, (C 6 -C 14 )ar(C 1 -C
  • 2 )-cycloalky or "cycloalkyl” refers to a saturated or partially unsaturated cyclic hydrocarbon radical including 1 , 2 or 3 rings and including a total of 3 to 12 carbon atoms forming the rings.
  • the term cycloalkyl includes bridged, fused and spiro ring systems.
  • (C 3 -C 2 )-cycloalkyl refers to a cycloalkyl group having 3 to 8 (both inclusive) carbon atoms.
  • cycloalkyl examples include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, adamantyl, norbornyl, bicyclo[2.1 .0]pentane, bicyclo[2.2.1 ]heptyl, bicyclo[2.2.1 ]hept-2-ene, spiro[3.3]heptane, 1 ,2,3,3a-tetrahydropentalene and the like.
  • the "cycloalkyl” group can be unsubstituted or substituted with one or more groups, preferably 1 -3 groups independently selected from the group consisting of halogen, hydroxy, nitro, cyano, (Ci-C 6 )alkyl-(Rc)o-3, (Ci-C 6 )-alkoxy-(R c )o-3, (C 3 -C 12 )-cycloalkyl-(R c )o-3, 0-(C 3 -Ci 2 )-cycloalkyl-(R c )o- 3 , halo(C r C 6 )alkyl, halo(C C 6 )alkoxy, (C 6 -Ci 4 )-aryl-(R c )o- 3 , 0-(C 6 -Ci 4 )aryl-(R c )o- 3 , (C 6 -Ci 4 )ar(Ci-C 6 )alky
  • (CrC 6 )alkoxy refers to a (C C 6 )-alkyl having an oxygen radical attached thereto.
  • Representative alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy, isopropoxy, n-butoxy, isobutoxy, t-butoxy and the like.
  • (C 3 -Ci 2 )-cycloalkyloxy or cycloalkyloxy or 0-(C 3 -Ci 2 )-cycloalkyl refers to a (C3-Ci 2 )-cycloalkyl having an oxygen radical attached thereto.
  • Representative cycloalkyloxy groups include, but are not limited to, cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, cyclohexyloxy and the like.
  • the alkoxy and cycloalkyloxy can be unsubstituted or substituted with one or more groups selected from the group consisting of halogen, hydroxy, nitro, cyano, (C 1 -C 6 )alkyl-(R c ) 0 - 3 , (Ci-C 6 )-alkoxy-(R c ) ⁇ w, (C 3 -C 12 )-cycloalkyl- (R c )o-3, 0-(C 3 -Ci 2 )-cycloalkyl-(R c )o-3, halo(C C 6 )alkyl, halo(CrC 6 )alkoxy, (C 6 -Ci )-aryl- (Rc)o-3, 0-(C 6 -Ci4)aryl-(R c )o- 3!
  • substituted (CrC 6 )-alkoxy examples include, but are not limited to, chloromethoxy, 2-cyanoethoxy, trifluoromethoxy and benzyloxy group.
  • a benzyloxy group refers to a benzyl having an oxygen radical attached thereto.
  • (Ci-C 6 )-alkoxyalkyl or “alkoxyalkyl” as used herein refers to a (C-
  • (C 6 -C 4 )-aryr or “aryl” as used herein refers to monocyclic or bicyclic hydrocarbon groups having 6 to 14 ring carbon atoms, preferably 6 to 10 carbon atoms in which the carbocyclic ring(s) present have a conjugated pi electron system.
  • Examples of (C 6 -Ci 4 )-aryl residues are phenyl, naphthyl, fluorenyl or anthracenyl.
  • Preferred examples of (C6-Cu)-aryl residues are phenyl or naphthyl.
  • Aryl groups can be unsubstituted or substituted by one or more groups, for example 1 , 2, 3, 4 or 5 groups independently selected from the group consisting of halogen, hydroxy, nitro, cyano, (C C 6 )alkyl-(R c ) 0 -3, (Ci-C 6 )-alkoxy-(R c )o-3, (C 3 -Ci 2 )-cycloalkyl-(R c )o-3, 0-(C 3 -Ci 2 )-cycloalkyl- (R c )o-3, halo(CrC 6 )alkyl, halo(C r C 6 )alkoxy, (C 6 -Ci 4 )-aryl-(R c )o- 3 , 0-(C 6 -Ci 4 )aryl-(R c )o-3, (C 6 -C 14 )ar(Ci-C 6 )alkyl-(R c
  • the substituent can be located in the 2-position, the 3-position or the 4-position.
  • phenyl carries two substituents, they can be located in 2, 3-position, 2, 4-position, 2, 5- position, 2, 6-position, 3, 4-position or 3, 5-position.
  • monosubstituted phenyl groups include, but are not limited to, 3-trifluoromethylphenyl, 4-chlorophenyl and 4-cyanophenyl.
  • disubstituted phenyl groups include, but are not limited to, 3, 5-difluorophenyl, and 3, 4-dimethoxyphenyl.
  • (C 6 -Ci 4 )-aryloxy or "aryloxy” or "0-(C 6 -Ci 4 )aryl” refers to an "(C 6 -Ci 4 )-aryr group having an oxygen radical attached thereto.
  • the aryl of aryloxy group can be unsubstituted or substituted as explained in the definition of substituted (C 6 -Ci 4 )-aryl herein above. Examples of aryloxy groups include, but not limited to, phenoxy, 4-chlorophenoxy, and 3, 4-dimethoxyphenoxy.
  • aralkyl or "(C 6 -Ci 4 )ar(C-
  • Exemplary aralkyl groups include (CH 2 )p-phenyl, wherein p is an integer from 1 to 6, such as benzyl wherein p is 1 .
  • the aryl of the (C 6 -Ci 4 )-aralkyl group can be unsubstituted or substituted as explained in the definition of substituted aryl herein above.
  • aralkyloxy or "(C 6 -Ci 4 )ar(C-
  • the aryl of aralkyloxy group may be unsubstituted or substituted as explained in the definition of substituted aryl herein above.
  • heteroatom as used herein, includes nitrogen (N), oxygen (O) and sulfur (S). Any heteroatom with unsatisfied valency is assumed to have a hydrogen atom to satisfy the valency.
  • heterocyclyl or “heterocyclic” whether used alone or as part of a substituent group, unless indicated otherwise, refers to a saturated, partially unsaturated, monocyclic or polycyclic ring system containing 1 to 1 0 carbon atoms and 1 to 4 identical or different heteroatoms selected from the group consisting of oxygen, nitrogen and sulfur. Unless indicated otherwise, the term “heterocyclyl” primarily refers to a 3- to 12- membered ring system, preferably, 3- to 1 0- membered ring system which can be a monocyclic or a bicyclic ring.
  • heterocyclyls include, but are not limited to, pyrrolyl, pyrrolidinyl, pyrazolyl, imidazolyl, pyrazinyl, piperazinyl, oxazolyl, oxadiazolyl, isoxazolyl, triaziolyl, thiazolyl, tetrazolyl, furyl, thienyl, purinyl, pyridinyl, pyridazinyl, pyrimidinyl, piperidyl, benzoxazolyl, benzothiazolyl, benzofuranyl, purinyl, benzirmidazolyl, benzoxazolyl, indolyl, indazolyl, isoindolyl, isothiazolyl, isoquinolyl, isoquinolyl, morpholinyl, thiomorpholinyl, thiomorpholinyl-1 , 1 - dioxide, quinoxalinyl
  • heterocyclyl having an aromatic ring containing heteroatom/s are herein referred to by the customary term "heteroaryl". Unless indicated otherwise, within the context of the present invention and as used herein, the term “heteroaryl” refers to a 5- to 10- membered aromatic monocyclic or bicyclic ring system containing 1 to 4 identical or different hetero atoms selected from the group consisting of nitrogen, sulphur and oxygen.
  • heteroaryls include, but are not limited to, pyrrole, pyrazole, imidazole, pyrazine, furan, thiophene, oxazole, thiazole, benzimidazole, benzoxazole, benzothiazole, benzofuran, indole, indazole, isoindole, isoquinoline, isooxazole, triazine, purine, pyridine, quinoline, oxadiazole, thiene, pyridazine, pyrimidine, isothiazole, quinoxaline (benzopyrine) and tetrazole.
  • heterocyclyl or heteroaryl group can be unsubstituted or substituted.
  • a substituted heterocyclyl or heteroaryl refers to a heterocyclyl or heteroaryl substituted with 1 -5 groups independently selected from the group consisting of halogen, hydroxy, nitro, cyano, (Ci-C 6 )alkyl-(R c ) 0 -3, (CrC 6 )-alkoxy-(R c )o-3, (C 3 -Ci 2 )-cycloalkyl-(R c )o-3, O-(C 3 -Ci 2 )-cycloalkyl-(R c ) 0 - 3 , halo(C r C 6 )alkyl, halo(C C 6 )alkoxy, (C 6 -Ci 4 )-aryl-(R c ) 0 - 3 , O-(C 6 -Ci 4 )aryl-(R c ) 0 - 3 , (C 6 -Ci4)ar(Ci-C 6
  • isotopic forms or “isotopically labeled forms” is a general term used for isotopic forms of compounds of formula I, wherein one or more atoms of compounds of formula I are replaced by their respective isotopes. All isotopes of any particular atom or element as specified are contemplated within the scope of the compounds of the invention.
  • isotopes that can be incorporated into the compounds disclosed herein include, but are not limited to, isotopes of hydrogen such as 2 H (deuterium or D) and 3 H, carbon such as 1 1 C, 13 C and 14 C, nitrogen such as 13 N and 15 N, oxygen such as 15 0, 17 0 and 18 0, chlorine such as 36 CI, fluorine such as 18 F and sulphur such as 35 S.
  • isotopes of hydrogen such as 2 H (deuterium or D) and 3 H
  • carbon such as 1 1 C, 13 C and 14 C
  • nitrogen such as 13 N and 15 N
  • oxygen such as 15 0, 17 0 and 18 0, chlorine such as 36 CI
  • fluorine such as 18 F
  • sulphur such as 35 S.
  • Substitution with heavier isotopes, for example, replacing one or more key carbon- hydrogen bonds with carbon-deuterium bond may show certain therapeutic advantages, resulting from longer metabolism cycles, (e.g., increased in vivo half life or reduced dosage requirements), improved safety or
  • isotopic forms of the compounds of formula I can include, without limitation, deuterated compounds of formula I.
  • deuterated as used herein, by itself or used to modify a compound or group, refers to replacement of one or more hydrogen atom(s), which is attached to carbon(s), with a deuterium atom.
  • the compounds of formula I can include in the definitions of one or more of its various variables, wherever applicable, deuterium, deuterated-alkyl, deuterated-alkoxy, deuterated-cycloalkyl, deuterated-heterocyclyl, deuterated-aryl, deuterated-heteroaryl and the like.
  • deuterated-alkyl refers to an (d-C6)-alkyl group as defined herein, wherein at least one hydrogen atom bound to carbon is replaced by a deuterium. That is, in a deuterated alkyl group, at least one carbon atom is bound to a deuterium. In a deuterated alkyl group, it is possible for a carbon atom to be bound to more than one deuterium; it is also possible that more than one carbon atom in the alkyl group is bound to a deuterium.
  • deuterated and the terms deuterated-heterocyclyl, deuterated-heteroaryl, deuterated-cycloalkyl, deuterated-aryl, deuterated-alkoxy each refer to the corresponding chemical moiety wherein at least one carbon is bound to a deuterium.
  • solvate(s) refers to a compound formed by the interaction of a solute (in the present invention, a compound of formula I or a pharmaceutically acceptable salt thereof) and a solvent.
  • solvents for the purpose of the invention may not interfere with the biological activity of the solute.
  • the solvent used is a pharmaceutically acceptable solvent.
  • suitable pharmaceutically acceptable solvents include, without limitation, water, ethanol and acetic acid. Most preferably, the solvent used is water and the solvates obtained are referred to as hydrates. Examples for suitable solvates are the mono- or di-hydrates or alcoholates of the compounds according to the invention.
  • stereoisomer or “stereoisomeric form” is a general term used for all isomers of individual compounds (in the present invention, a compound of formula I) that differ only in the orientation of their atoms in space.
  • stereoisomer includes mirror image isomers (enantiomers), mixtures of mirror image isomers (racemates, racemic mixtures), geometric (cis/trans or E/Z) isomers, and isomers of compounds with more than one chiral center that are not mirror images of one another (diastereoisomers).
  • tautomer or “tautomeric form” refers to the coexistence of two (or more) compounds that differ from each other only in the position of one (or more) mobile atoms and in electron distribution, for example, keto-enol tautomers.
  • the term “pharmaceutically acceptable” means that the carrier, diluent, excipients, and/or salt must be compatible with the other ingredients of the formulation, and not deleterious to the recipient thereof.
  • pharmaceutically acceptable salt(s) as used herein includes salts of the active compound i.e. the compound of formula I, which retain the desired biological activity of the subject compound and exhibit minimal undesired toxicological effects; and are prepared with suitable acids or bases, depending on the particular substituents found on the compounds described herein.
  • N-oxide refers to the oxide of the nitrogen atom of a nitrogen-containing heteroaryl or heterocycle. N-oxide can be formed in the presence of an oxidizing agent such as m-chloro-perbenzoic acid or hydrogen peroxide. N-oxide refers to an amine oxide, also known as amine-N-oxide, and is a chemical compound that contains N->0 bond.
  • a prodrug or “prodrugs” refers to any compound, which are derivatives of parent compound (in the context of the present invention, a compound of formula I), which following administration, release(s) the parent compound in vivo via a chemical or physiological process, e.g., a prodrug on being brought to the physiological pH or through enzyme action is converted to the parent compound.
  • the term "compound(s) of formula I" or “compounds of the present invention” are used interchangeably and includes all the stereoisomeric and tautomeric forms and mixtures thereof in all ratios, and pharmaceutically acceptable salts, pharmaceutically acceptable solvates, pharmaceutically acceptable prodrugs, N-oxides and pharmaceutically acceptable polymorphs thereof.
  • the compound(s) of formula I can also be referred to herein as "the active compound” or "the active ingredient”.
  • therapeutically effective amount means an amount of a compound of formula I or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof; or a composition comprising the compound of formula I or a pharmaceutically acceptable salt thereof, effective in producing the desired therapeutic response in a particular patient suffering from a disease or disorder mediated by IL1 7 or TNF-a.
  • therapeutically effective amount includes the amount of the compound, when administered, that induces a positive modification in the disease or disorder to be treated or is sufficient to prevent development of, or alleviate to some extent, one or more of the symptoms of the disease or disorder being treated in a subject.
  • the amount of the compound used for the treatment of a subject is low enough to avoid undue or severe side effects, within the scope of sound medical judgment.
  • the therapeutically effective amount of the compound or composition will vary with the particular condition being treated, the age and physical condition of the end user, the severity of the condition being treated or prevented, the duration of the treatment, the nature of concurrent therapy, the specific compound or composition employed the particular pharmaceutically acceptable carrier utilized.
  • the term "pharmaceutically acceptable carrier” refers to a material that is non-toxic, inert, solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type which is suitable for a subject, preferably a mammal, more preferably a human, and is suitable for delivering an active agent to the target site without terminating the activity of the agent.
  • disease(s) or disorder(s) mediated by IL-17 or TNF-a encompasses all diseases, disorders and medical conditions in which pro-inflammatory cytokines IL-1 7 or TNF- ⁇ plays a role, whether directly or indirectly, including the causation, development, progression, persistence or pathology of the disease or disorder.
  • disease(s) or disorder(s) mediated by IL-17 encompasses all diseases, disorders and medical conditions in which pro-inflammatory cytokine IL-17 plays a role, whether directly or indirectly, including the causation, development, progression, persistence or pathology of the disease or disorder.
  • TNF-a disease(s) or disorder(s) mediated by TNF-a
  • the diseases or disorders mediated by IL-17 or TNF-a can be selected from the group consisting of an autoimmune or inflammatory disease or disorder; and a metabolic disease or disorder.
  • subject refers to an animal, preferably a mammal, and most preferably a human.
  • mammal refers to warm-blooded vertebrate animals of the class 'mammalia', including humans, characterized by a covering of hair on the skin and, in the female, milk-producing mammary glands for nourishing the young.
  • mammal includes animals such as cat, dog, rabbit, bear, fox, wolf, monkey, deer, mouse, pig and human.
  • subject may be used interchangeably with the term patient.
  • a subject in need thereof means a subject in need of the treatment for the disease or disorder that is mediated by IL1 7 or TNF-a.
  • a subject in need thereof means a subject (patient) diagnosed having a disease or disorder that is mediated by IL17 or TNF-a.
  • treatment refers to alleviate, slow the progression, attenuation or cure of existing diseases or condition (e.g. rheumatoid arthritis). Treatment also includes treating, preventing development of, or alleviating to some extent, one or more of the symptoms of the diseases or condition.
  • the present invention relates to a compound of formula I,
  • A is triazole, pyrazole, imidazole, oxazole, isoxazole, thiazole, pyrrole or a heteroaryl ring system selected from:
  • R 1 5 R 2 , R3 and n are as defined in the first aspect with the provisos that:
  • the present invention relates to a compound of formula I, wherein,
  • A is triazole, pyrazole, imidazole, oxazole, isoxazole, thiazole or pyrrole;
  • the present invention relates to a compound of formula I;
  • A is triazole, pyrazole, isoxazole or thiazole
  • L is * N(R 4 )-C(0)-N(R 5 ), * C(0)N(R 5 ), * N(R 4 )-C(0), * CH(R 6 )-C(0)-N(R 5 ), * CH(R 6 )N(R 5 ), * N(R 4 )-S(0) 2 or * NR 4 ; wherein * indicates the point of attachment to A;
  • R , R 5 and R 6 are as defined in the first aspect with the provisos that:
  • the present invention relates to a compound of formula I;
  • A is triazole
  • the present invention relates to a compound of formula I; wherein:
  • A is pyrazole
  • L is * N(R 4 )-C(0)-N(R 5 ); * CH(R 6 )-C(0)-N(R 5 ); * CH(R 6 )N(R 5 ); * N(R 4 )-S(0) 2 or * NR 4 ; wherein * indicates the point of attachment to A;
  • R 4 , R 5 and R 6 are as defined in the first aspect with the proviso that when L is * N(R 4 )- C(0)-N(R 5 ), then the said L group is not attached to 5-position of pyrazole;
  • the present invention relates to a compound of formula I;
  • A is isoxazole
  • the present invention relates to a compound of formula I;
  • A is thiazole
  • the present invention provides a compound of formula I ; wherein A is
  • R 1 ; R 2 , R3 and n are as defined in the first aspect
  • the present invention provides a compound of formula I ; wherein A is
  • the present invention relates to a compound of formula I; wherein A is
  • R 1 ; R 2 , R 3 and n are as defined in the first aspect
  • the present invention relates to a compound of formula I; wherein each of Xi , X 2 , X 4 and X 5 is C(R 7 ); and X 3 is C(R 8 ); and
  • R 7 and R 8 are as defined in the first aspect; with the provisos that:
  • L is a group other than * C(0)N(R 5 ) or * N(R 4 )-C(0);
  • the present invention relates to a compound of formula I; wherein A is selected from a triazole, pyrazole, isoxazole or thiazole;
  • each of Xi , X 2 , X 4 and X 5 is C(R 7 ); and X 3 is C(R 8 ); and
  • R 7 and R 8 are as defined in the first aspect; with the provisos that:
  • L is a group other than * C(0)N(R 5 ) or * N(R 4 )-C(0);
  • the present invention relates to a compound of formula I; wherein A is triazole or pyrazole;
  • L is *N(R 4 )-C(0)-N(R 5 ); wherein * indicates the point of attachment to A;
  • each of Xi , X 2 , X 4 and X 5 is C(R 7 ); and X 3 is C(R 8 ); and
  • R 4 , R 5 , R 7 and R 8 are as defined in the first aspect; with the proviso that when A is pyrazole the said L group is not attached to 5-position of pyrazole;
  • the present invention relates to a compound of formula I; wherein A is triazole;
  • L is * N(R 4 )-C(0)-N(R 5 ); * C(0)N(R 5 ), * CH(R 6 )-C(0)-N(R 5 ); * CH(R 6 )N(R 5 ) or * N(R 4 )- S(0) 2 ; wherein * indicates the point of attachment to A;
  • each of Xi , X 2 , X 4 and X5 is C(R 7 ); and X3 is C(R 8 ); and
  • R , R 5 , R 6 , R 7 and R 8 are as defined in the first aspect
  • the present invention relates to a compound of formula I; wherein A pyrazole;
  • L is *N(R 4 )-C(0)-N(R 5 ); N(R 4 )-S(0) 2 or *CH(R 6 )N(R 5 ); wherein * indicates the point of attachment to A;
  • each of Xi , X 2 , X and X 5 is C(R 7 ); and X 3 is C(R 8 ); and
  • R 4 , R5, R 7 and R 8 are as defined in the first aspect; with the proviso that when A is pyrazole and L is * N(R 4 )-C(0)-N(R 5 ), then the said L group is not attached to 5-position of pyrazole;
  • the present invention relates to a compound of formula I; wherein A is
  • L is *N(R 4 )-C(0)-N (R 5 ); *C(0)N(R 5 ); *N(R 4 )-S(0) 2 or *N R 4 ; wherein * indicates the point of attachment to A;
  • each of Xi , X 2 , X 4 and X 5 is C(R 7 ); and X 3 is C(R 8 ) ;
  • Ri , R2, R3, R4, R5, R7, Re and n are as defined in the first aspect
  • the present invention relates to a compound of formula I; wherein A is
  • L is *N(R 4 )-C(0)-N(R 5 ) ; *C(0)N(R 5 ); N(R 4 )-S(0) 2 or *NR 4 ; wherein * indicates the point of attachment to A;
  • each of Xi , X 2 , X 4 and X 5 is C(R 7 ); and X 3 is C(R 8 ) ;
  • Ri , R2, R3, R4, R5, R7, Re and n are as defined in the first aspect
  • the present invention relates to a compound of formula I; wherein one of Xi , X 2 , X 4 or X 5 is N, while the remaining are C(R 7 ); X3 is N or C(Rs); or any two of Xi , X 2 , X 4 or X 5 are N, while the remaining are C(R 7 ); X3 is N or C(Rs) ; and R 7 and R 8 are as defined in the first aspect; with the provisos that:
  • L is a group other than *C(0)N(R 5 ) or *N(R 4 )-C(0); and (b) when A is pyrazole and L is *N(R 4 )-C(0)-N(R 5 ), then said L group is not attached to 5-position of pyrazole; or an isotopic form, stereoisomer or a tautomer or a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
  • Representative compounds of formula I encompassed in accordance with the present invention include:
  • the compounds of the present invention include all isotopic forms, stereoisomeric and tautomeric forms and mixtures thereof in all ratios and their pharmaceutically acceptable salts, pharmaceutically acceptable solvates, prodrugs, N-oxides, S-oxides and polymorphs. According to another aspect of the present invention, there are provided processes for the preparation of the compounds of formula I.
  • the compound of formula I can be prepared by various methods including using methods well known to the person skilled in the art. Examples of processes for the preparation of the compounds of formula I are described below and illustrated in the following scheme, but are not limited thereto. It will be appreciated by the person skilled in the art that within certain of the processes described herein, the order of the synthetic steps employed can be varied and will depend inter alia on factors such as the nature of functional groups present in a particular substrate and the protecting group strategy (if any) to be adopted. Clearly, such factors will also influence the choice of reagent such as bases, solvents, coupling agents to be used in the reaction steps.
  • Scheme 1 depicts a process for the preparation of the compounds of formula I, wherein A, R-i , R 2 , F?3, X-i , X2, X3, X 4 , X5 and n are as defined in the first aspect of the present invention, and L is * N(R 4 )-C(0)-N(R 5 ); wherein * indicates the point of attachment to A; and R 4 and R5 are hydrogen.
  • A, R-i , R 2 , R3 and n are as defined in the first aspect; is hydrolysed by reacting it with NaOH in a solvent such as a mixture of tetrahydrofuran (THF) and water at a temperature ranging from 70 °C to 90 °C to obtain a compound 2 of the following formula;
  • a solvent such as a mixture of tetrahydrofuran (THF) and water at a temperature ranging from 70 °C to 90 °C to obtain a compound 2 of the following formula;
  • step (1 a) The compound of formula 2 as obtained in step (1 a) is reacted with reagents namely trichloroacetonitrile, triphenyl phosphine and sodium azide in a solvent such as acetone at a temperature ranging from 20 °C to 35 °C to obtain a compound 3 of the following formula;
  • step (1 a) The compound of formula 2 as obtained in step (1 a) is reacted with reagents namely sodium azide and triphosgene in the presence of a base such as triethylamine (TEA) in a solvent such as acetone or THF at a temperature ranging from 20 °C to 35 °C to obtain a compound 3 of the above formula.
  • a base such as triethylamine (TEA)
  • a solvent such as acetone or THF
  • step (1 b) The compound of formula 3 as obtained in step (1 b) is reacted with a compound 4 of the following formula;
  • X 1 ; X 2 , X 3 , X 4 and X 5 are as defined in the first aspect;
  • Scheme 2 depicts a process for the preparation of the compounds of formula I, wherein A, R-i , R 2 , R3, X-i , X 2 , X3, X 4 , X5 and n are as defined in the first aspect of the present invention, and L is * C(0)N(R 5 ); wherein * indicates the point of attachment to A; and R 5 is hydrogen.
  • X 1 ; X 2 , X3, X 4 and X 5 are as defined in the first aspect;
  • Scheme 3 depicts a process for the preparation of the compounds of formula I, wherein A, R-i , R 2 , R3, X-i , X 2 , X3, X 4 , X5 and n are as defined in the first aspect of the present invention, and L is *N(R 4 )-S(0) 2 ; wherein * indicates the point of attachment to A; and R is hydrogen.
  • X 1 ; X 2 , X 3 , X 4 and X 5 are as defined in the first aspect;
  • step 3(a) The compound of formula 5 as obtained in step 3(a) is reacted with a compound 6 of the following formula;
  • Scheme 4 depicts a process for the preparation of the compounds of formula I, wherein A, R-i , R 2 , R3, Xi , X 2 , X3, X 4 , X5 and n are as defined in the first aspect of the present invention, and L is *N(R 4 )-C(0); wherein * indicates the point of attachment to A; and R 4 is hydrogen.
  • step 4(a) The compound of formula 7 as obtained in step 4(a) is reacted with compound 8 of the following formula;
  • X 1 ; X 2 , X 3 , X 4 and X 5 are as defined in the first aspect;
  • Scheme 5 depicts a process for the preparation of the compounds of formula I wherein, A, Ri , R 2 , R3, X-i , X2, X3, X4, X5 and n are as defined in the first aspect of the present invention, and L is * NR 4 ; wherein * indicates the point of attachment to A; and R 4 is hydrogen.
  • Scheme 6 depicts a process for the preparation of the compounds of formula I wherein, A, R-i, R 2 , R3, X-i, X 2 , X3, X4, X5 and n are as defined in the first aspect of the present invention, and L is * CH(R 6 )N(R 5 ); wherein * indicates the point of attachment to A; R 5 is hydrogen and R 6 is hydrogen or cyano.
  • L is *CH(R 6 )N(R 5 ); R 5 is hydrogen; and R 6 is cyano.
  • A, Ri , R 2 , R3 and n are as defined in the first aspect; is reacted with lithium aluminium hydride (LAH) in a solvent such as tetrahydrofuran at a temperature ranging from -5 °C to 5 °C to obtain a compound 10 of the following formula;
  • LAH lithium aluminium hydride
  • A, R ; R 2 , R3 and n are as defined in the first aspect.
  • step 6(a) The compound of formula 10 as obtained in step 6(a) is reacted with manganese dioxide in a solvent such as chloroform at a temperature ranging from 65 °C to 75 °C to obtain a compound 1 1 of the following formula;
  • step 6(b) The compound of formula 1 1 as obtained in step 6(b) is reacted with compound 4 of the following formula; wherein, Xi , X 2 , X3, X 4 and X5 are as defined in the first aspect;
  • the compound of formula 1 2 as obtained in step 6(c) is reduced by using a reducing agent such as sodium borohydride or sodium cyanoborohydride in a solvent such as tetrahydrofuran at a temperature ranging from 0 °C to 25 °C to obtain a compound of formula I wherein, A, R-i , R 2 , R3, X-i , X2, X3, X 4 , X5 and n are as defined in the first aspect of the present invention, and L is *CH(R 6 )N(R 5 ); wherein * indicates the point of attachment to A; and R 5 and R 6 are hydrogen.
  • a reducing agent such as sodium borohydride or sodium cyanoborohydride in a solvent such as tetrahydrofuran
  • step 6(c) The compound of formula 1 2 as obtained in step 6(c) is reacted with trimethylsilanecarbonitrile and tetrabutylammonium difluorotriphenylsilicate in a solvent such as tetrahydrofuran at a temperature ranging from 0 °C to 25 °C to obtain a compound of formula I; wherein, A, Ri , R 2 , R3, Xi , X2, X3, X4, X5 and n are as defined in the first aspect of the present invention, and L is * CH(R 6 )N(R 5 ); wherein * indicates the point of attachment to A; R 5 is hydrogen and R 6 is cyano.
  • Scheme 7 depicts a process for the preparation of the compounds of formula I wherein, A, R-i , R 2 , R 3 , X-i , X 2 , X3, X 4 , X5 and n are as defined in the first aspect of the present invention, and L is * CH(R 6 )-C(0)-N(R 5 ); wherein * indicates the point of attachment to A; R 5 is hydrogen and R 6 is hydroxy.
  • the compound of formula I as obtained in Schemes 1 , 2, 3, 4, 5, 6 and 7 can be optionally converted into its corresponding pharmaceutically acceptable salt.
  • pharmaceutically acceptable salts refers to organic and inorganic salts of a compound of the invention, depending on the particular group (acidic or basic group) present in the compounds of formula I described herein.
  • base addition salts can be obtained by contacting the compounds of formula I with a sufficient amount of an appropriate base, either neat or in a suitable inert solvent.
  • pharmaceutically acceptable base addition salts include sodium, potassium, magnesium, calcium, ammonium, zinc or organic base salt.
  • pharmaceutically acceptable organic base addition salts include those derived from organic bases such as lysine, arginine, choline, guanidine, diethanolamine, tromethamine, metformin and the like.
  • acid addition salts can be obtained by contacting the compounds of formula I with a sufficient amount of an appropriate acid, either neat or in a suitable inert solvent.
  • suitable inert solvent examples include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, mono- hydrogensulfuric or hydriodic acids and the like, as well as the salts derived from organic acids such as acetic, ascorbic, isobutyric, benzoic, citric, oxalic, succinic, suberic, fumaric, propionic, mandelic, phthalic, benzenesulfonic, p-tolylsulfonic, lactic, maleic, malonic, mandelic, tartaric, methanesulfonic, ethanesulfonic, glucuronic or galacturonic acids
  • the pharmaceutically acceptable salts of the compounds of formula I can be synthesized from the compound of formula I, which contains a basic or an acidic group, by using conventional chemical methods. Generally, the salts are prepared by treating the compound of formula I which may be a free base or an acid with a suitable salt- forming inorganic or an organic acid or a base in a suitable solvent or dispersant or from another salt by cation or anion exchange.
  • suitable solvents that can be used for the preparation of pharmaceutically acceptable salts include, but are not limited to, ethyl acetate, diethyl ether, methanol, ethanol, acetone, tetrahydrofuran, dioxane or mixtures of these solvents.
  • the compounds of formula I can be regenerated from their corresponding salts by contacting the salt with an appropriate base or acid depending on the type of salt and isolating the parent compound in the conventional manner.
  • the compound differs from the various salt forms in certain physical properties.
  • An example of physical properties that may differ is solubility in polar solvents.
  • the present invention also encompasses within its scope the solvates of the compounds of formula I.
  • Certain compounds of the present invention can exist in unsolvated forms as well as solvated forms, including hydrated forms.
  • Certain compounds of the present invention may exist in multiple crystalline or amorphous forms. In general, all physical forms are suitable for the uses contemplated by the present invention and are intended to be within the scope of the present invention.
  • polymorphs of compounds of formula I can be prepared by crystallization of the compounds under different conditions.
  • the different conditions are, for example, using different solvents or their mixtures for crystallization; crystallization at different temperatures; various modes of cooling, ranging from very fast to very slow cooling during crystallizations.
  • Polymorphs can also be obtained by heating or melting the compound followed by gradual or fast cooling.
  • the presence of polymorphs can be determined by IR (infra-red) spectroscopy, solid probe NMR (nuclear magnetic resonance) spectroscopy, differential scanning calorimetry, powder x-ray diffraction or such other techniques.
  • the present invention includes all possible stereoisomers and geometric isomers of formula I and includes not only racemic compounds but also the optically active isomers as well.
  • a compound of formula I When a compound of formula I is desired as a single enantiomer, it can be obtained either by resolution of the final product or by stereospecific synthesis from either isomerically pure starting material or an appropriate intermediate. Resolution of the final product, an intermediate or a starting material may be effected by any suitable method known in the art, for example, Chiral reagents for asymmetric synthesis by Leo A. Paquette; John Wiley & Sons Ltd (2003).
  • the present invention is intended to include all tautomeric forms of the compounds.
  • prodrugs of the compound of formula I are those compounds that are converted to their parent compound intracellular ⁇ , where the cellular converting location is the site of therapeutic action.
  • the prodrugs of the compounds of the present invention are derivatives, particularly simple derivatives of the said compounds which upon administration to a subject in need thereof undergoes conversion by metabolic or chemical processes to release the parent drug (e.g. the compound of formula I) in wVo from which the prodrug is derived.
  • preferred produgs are pharmaceutically acceptable ester derivatives convertible by solvolysis under physiological conditions to the parent carboxylic acid, e.g., alkyl esters, cycloalkyl esters, alkenyl esters, benzyl esters, mono- or di-substituted alkyl esters such as the pivaloyloxymethyl ester and the like conventionally used in the art (An introduction to Medicinal Chemistry, Graham. L. Patrick, Second Edition, Oxford University Press, pg 239-248; Prodrugs: Challenges and Rewards, Part 1 and Part 2, AAPS Press, Edited by Valentino J. Stella, Renald T. Borchardt, Michael J. Hagemon, Reza Oliyai, Hans Maag, Jefferson W. Tilley).
  • alkyl esters e.g., alkyl esters, cycloalkyl esters, alkenyl esters, benzyl esters, mono- or di-substituted alkyl esters
  • the present invention relates to a method for the treatment of a disease or disorder mediated by IL-17 or TNF- ⁇ , comprising administering to a subject in need thereof; a therapeutically effective amount of the compound of formula I or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof.
  • the present invention provides a compound of formula I or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof; for use as IL-17 inhibitor.
  • the present invention provides a compound of formula I or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof; for use as TNF-a inhibitor.
  • a compound of formula I or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or solvate thereof for use in the treatment of a disease or a disorder mediated by IL-17 or TNF-a.
  • the present invention provides use of a compound of formula I or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof; for the manufacture of a medicament for the treatment of a disease or a disorder mediated by IL-1 7 or TNF-a.
  • the present invention encompasses within its scope all the diseases or disorders wherein IL-17 is implicated. In another embodiment of the present invention, the present invention encompasses within its scope all the diseases or disorders wherein TNF-a is implicated.
  • the disease or disorder mediated by the pro-inflammatory cytokines IL-17 or TNF-a is selected from the group consisting of an autoimmune or inflammatory disease or disorder; and a metabolic disease or disorder.
  • the autoimmune disorder, inflammatory disorder or metabolic disorder can be selected from the group consisting of: inflammatory bowel disease, rheumatoid arthritis, juvenile rheumatoid arthritis, psoriatic arthritis, osteoarthritis, refractory rheumatoid arthritis, chronic non-rheumatoid arthritis, ankylosing spondylitis, osteoporosis/bone resorption, chronic graft-versus-host disease, acute graft-versus-host disease, multiple sclerosis, systemic lupus erythematosus, Celiac Sprue, idiopathic thrombocytopenic thrombotic purpura, myasthenia gravis, Sjogren's syndrome, scleroderma, asthma, bronchitis, epidermal hyperplasia, Crohn's disease, atherosclerosis, septic shock syndrome, coronary heart disease, vasculitis, ulcerative colitis, psoria
  • the metabolic disease or disorder is selected from the group consisting of obesity, dyslipidemia, hyperlipidemia, hypertriglyceridemia, low adiponectin level, dyslipoproteinemia, impaired glucose tolerance, insulin resistance, increase in HbA1 c (glycosylated haemoglobin) level, reduced metabolic activity and Type II diabetes.
  • the present invention also encompasses within its scope the use of a compound of formula I or its pharmaceutically acceptable salt or a solvate in combination with other therapeutically active agents; wherein the compound of formula I and the further therapeutic agent are administered either simultaneously or sequentially.
  • the present invention relates to pharmaceutical composition(s) containing a therapeutically effective amount of at least one compound of formula I or its pharmaceutically acceptable salt or solvate thereof and a conventional pharmaceutically acceptable carrier.
  • the present invention also relates to a process for the production of a pharmaceutical composition, which includes bringing at least one compound of formula I, into a suitable administration form using a pharmaceutically acceptable excipient and, if appropriate, further suitable active compounds, additives or auxiliaries can be added.
  • composition(s) of the present invention can be administered orally, for example in the form of pills, tablets, coated tablets, capsules, granules or elixirs. Administration, however, can also be carried out rectally, for example in the form of suppositories, or parenterally, for example intravenously, intramuscularly or subcutaneously, in the form of injectable sterile solutions or suspensions, or topically, for example in the form of ointments or creams or transdermal ⁇ , in the form of patches, or in other ways, for example in the form of aerosols or nasal sprays.
  • composition(s) according to the invention is/are prepared in a manner known and familiar to one skilled in the art.
  • Pharmaceutically acceptable inert inorganic and/or organic carriers and/or additives can be used in addition to the compound(s) of formula I, and/or its (their) physiologically tolerable salt(s).
  • Carriers for soft gelatin capsules and suppositories are, for example, fats, waxes, natural or hardened oils, etc.
  • Suitable carriers for the production of solutions for example injection solutions, or of emulsions or syrups are, for example, water, physiological sodium chloride solution or alcohols, for example, ethanol, propanol or glycerol, sugar solutions, such as glucose solutions or mannitol solutions, or a mixture of the various solvents which have been mentioned.
  • the pharmaceutical composition(s) normally contain about 1 % to 99 %, for example, about 5 % to 70 %, or from about 1 0 % to about 30 % by weight of the compound of formula I or its pharmaceutically acceptable salt.
  • the amount of the compound of formula I or its pharmaceutically acceptable salt in the pharmaceutical composition(s) can range from about 1 mg to about 1000 mg or from about 2.5 mg to about 500 mg or from about 5 mg to about 250 mg or in any range falling within the broader range of 1 mg to 1000 mg or higher or lower that the specified range.
  • the dosage range which are suitable in a specific case depend on the type of inflammatory disorder or autoimmune disorder to be treated and on the state of the respective condition or disorder.
  • the selected dosage level can be readily determined by a skilled medical practitioner in the light of the relevant circumstances, including the condition (inflammatory disorder or autoimmune disorder) to be treated, the chosen route of administration including other factors such as age, weight and physical health and response of the individual patient, pharmacokinetics, severity of the disease and the like, factors known in the medical art.
  • Actual dosage levels of the active ingredients i.e.
  • the compounds of formula I in the pharmaceutical composition of this present invention can be varied so as to obtain an amount of the active ingredient, which is effective to achieve the desired therapeutic response for a particular patient (subject in need of the treatment), composition, and mode of administration without being toxic to the patient.
  • the dose of the compounds of formula I or pharmaceutically acceptable salts thereof, which is to be administered can cover a wide range.
  • the dose to be administered daily is to be selected to suit the desired effect.
  • a suitable dosage is about 0.01 mg/kg/day to about 200 mg/kg/day of the compound of formula I or its pharmaceutically acceptable salt, for example, about 0.1 mg/kg/day to 100 mg/kg/day of a compound of formula I or its pharmaceutically acceptable salt. If required, higher or lower daily doses can also be administered.
  • the pharmaceutical compositions can contain additives such as, for example, fillers, antioxidants, dispersants, emulsifiers, defoamers, flavors, preservatives, solubilizers or colorants.
  • Pharmaceutical compositions can also contain two or more compounds of formula I or their physiologically tolerable salts.
  • the pharmaceutical preparations can also contain one or more other therapeutically or prophylactically active ingredients.
  • the present invention also encompasses within its scope use of a compound of formula I or its pharmaceutically acceptable salt or a solvate in combination with other therapeutically active agents.
  • the compound of formula I or its pharmaceutically acceptable salt can be administered either simultaneously or sequentially in combination with the further therapeutically active agents.
  • a pharmaceutical composition including a compound of formula I or a pharmaceutically acceptable salt can be administered to a mammal, in particular a human, with any other pharmaceutically active compounds, in mixtures with one another or in the form of pharmaceutical compositions.
  • the therapeutically active agents that can be used in combination with one or more compounds of formula I or its pharmaceutically acceptable salt can be selected from: monoclonal antibodies, non-steroidal anti-inflammatory drugs (NSAIDs) such as COX inhibitors or PDE4 inhibitors.
  • NSAIDs non-steroidal anti-inflammatory drugs
  • monoclonal antibodies include, but not limited to, rituximab, etanercept, infliximab, adalimumab, natalizumab and vedolizumab.
  • NSAIDs include, but not limited to, diclofenac, indomethacin, sulindac, mefenamic acid, piroxicam, ibuprofen, naproxen, ketoprofen, phenylbutazone, aspirin, diflunisal, nimesulide, celecoxib, valdecoxib, etorcoxib and meloxicam.
  • PDE4 inhibitors include, but not limited to, rolipram, ibudilast, luteolin and roflumilast. It is understood that modifications that do not substantially affect the activity of the various aspects of this invention are included. Accordingly, the following examples are intended to illustrate but not to limit the present invention.
  • Step 2) Azido(1-(4-(trifluoromethyl)phenyl)-5-methyl-1 H-pyrazol-4-yl) methanone (intermediate 2)
  • step 1 To a stirred solution of the compound obtained in step 1 [intermediate 1 ] (3.90 mmol) in acetone (10 mL) in an ice bath, was added trichloroacetonitrile (7.81 mmol), triphenyl phosphine (7.81 mmol) and sodium azide (4.68 mmol). The reaction mixture was stirred for 2 h at room temperature. On completion of reaction, the reaction was quenched with water (10 mL). The reaction mixture was extracted with ethyl acetate (25 mL). The organic layer was separated, washed with brine (1 0 mL), dried with Na 2 SO 4 , concentrated and purified using column chromatography (silica gel, ethyl acetate and petroleum ether gradient) to afford the title compound.
  • step 1 To a stirred solution of the compound obtained in step 1 [intermediate 1 ] (2.23 mmol) in dry acetone (5 mL) was added sodium azide (4.47 mmol), triethylamine (4.47 mmol) at
  • step 2 [intermediate 2] (0.339 mmol) and 2-(trifluoromethyl)- 3H-benzo[d]imidazol-5-amine (0.339 mmol) in toluene (3 mL) was refluxed for 5 h. The reaction mixture was allowed to cool to the room temperature. The precipitated compound was filtered, washed with petroleum ether and cold dichloromethane, and dried to afford the title compound.
  • the compound of example 2 was prepared analogous to the procedure described in example 1 by reaction of 5-methyl-1 -(4-(trifluoromethyl)phenyl)-1 H-pyrazole-4-carbonyl azide with 5-amino-2-(4-(4-chlorophenyl)-4-hydroxypiperidin-1 -yl)benzonitrile in step 3.
  • the compound of example 3 was prepared analogous to the procedure described in example 1 by reaction of 5-methyl-1 -(4-(trifluoromethyl)phenyl)-1 H-pyrazole-4-carbonyl azide with 5-amino-2-(4-(2-hydroxyethyl)piperidin-1 -yl)benzonitrile in step 3.
  • the compound of example 4 was prepared analogous to the procedure described in example 1 by reaction of 5-methyl-1 -phenyl-1 H-pyrazole-4-carbonyl azide with naphthalen-1 -amine in step 3.
  • the compound of example 5 was prepared analogous to the procedure described in example 1 by reaction of 1 -(4-chlorophenyl)-5-methyl-1 H-pyrazole-4-carbonyl azide with 5-amino-2-(4-morpholinopiperidin-1 -yl)benzonitrile in step 3.
  • the compound of example 6 was prepared analogous to the procedure described in example 1 by reaction of 5-methyl-1 -(4-(trifluoromethyl)phenyl)-1 H-pyrazole-4-carbonyl azide with 5-amino-2-(4-morpholinopiperidin-1 -yl)benzonitrile in step 3.
  • the compound of example 7 was prepared analogous to the procedure described in example 1 by reaction of 5-methyl-1 -(4-(trifluoromethyl)phenyl)-1 H-pyrazole-4-carbonyl azide with 5-amino-2-(piperazin-1 -yl)benzonitrile in step 3.
  • the compound of example 9 was prepared analogous to the procedure described in example 1 by reaction of 5-methyl-1 -(4-(trifluoromethyl)phenyl)-1 H-pyrazole-4-carbonyl azide with 5-amino-2-(4-(4-fluoro-3-(trifluoromethyl)benzoyl)piperazin-1 -yl)benzonitrile in step 3.
  • the compound of example 10 was prepared analogous to the procedure described in example 1 by reaction of 5-methyl-1 -(4-(trifluoromethyl)phenyl)-1 H-pyrazole-4-carbonyl azide with 5-(methylamino)-2-(4-morpholinopiperidin-1 -yl)benzonitrile in step 3.
  • Step 1) 1-(4-chlorophenyl)-1 H-1 ,2,3-triazole-4-carboxylic acid (intermediate 1)
  • H 2 0 (1 :1 vol, 20 mL)
  • NaOH 25.50 mmol
  • Step 2) 1-(4-chlorophenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide (intermediate 2)
  • step 1 To a stirred solution of the compound obtained in step 1 [intermediate 1 ] (3.90 mmol) in acetone (10 mL) in an ice bath, was added trichloroacetonitrile (7.81 mmol), triphenyl phosphine (7.81 mmol) and sodium azide (4.68 mmol). The reaction mixture was stirred for 2 h at room temperature. On completion of reaction, the reaction was quenched with water (10 mL). The reaction mixture was extracted with ethyl acetate (25 mL). The organic layer was separated, washed with brine (1 0 mL), dried with Na 2 SO 4 , concentrated and purified using column chromatography (silica gel, ethyl acetate and petroleum ether gradient) to afford the title compound.
  • step 1 To a stirred solution of the compound obtained in step 1 [intermediate 1 ] (2.23 mmol) in dry acetone (5 mL) was added sodium azide (4.47 mmol), triethylamine (4.47 mmol) at
  • step 2 [intermediate 2] (0.339 mmol) and 2-(trifluoromethyl)- 3H-benzo[d]imidazol-5-amine (0.339 mmol) in toluene (3 mL) was refluxed for 5 h. The reaction mixture was allowed to cool to the room temperature. The precipitated compound was filtered, washed with petroleum ether and cold dichloromethane, and dried to afford the title compound.
  • the compound of example 12 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-chlorophenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 2- ([1 ,4'-bipiperidin]-1 '-yl)-5-aminobenzonitrile in step 3.
  • the compound of example 13 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 2-(1 -(4-amino-2-cyanophenyl)piperidin-4-yl)ethyl diethyl phosphate in step 3.
  • the compound of example 14 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5-amino-2-(4-(2-methoxyethyl)piperidin-1 -yl)benzonitrile in step 3.
  • the compound of example 15 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 4-(4-morpholinopiperidin-1 -yl)-2-(trifluoromethyl)aniline in step 3.
  • the compound of example 16 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-morpholinophenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5-amino-2-(4-morpholinopiperidin-1 -yl)benzonitrile in step 3.
  • the compound of example 17 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-isopropylphenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with
  • the compound of example 18 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-chlorophenyl)-5-methyl-1 H-1 ,2,3-triazole-4-carbonyl azide with 5-amino-2-(4-morpholinopiperidin-1 -yl)benzonitrile in step 3.
  • the compound of example 20 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 4-(1 -(4-amino-2-cyanophenyl)piperidin-4-yl)butanoic acid in step 3.
  • the compound of example 22 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-chlorophenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5- amino-2-(piperazin-1 -yl)benzonitrile in step 3.
  • the compound of example 23 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-chlorophenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5- (4-morpholinopiperidin-1 -yl)pyridin-2-amine in step 3.
  • the compound of example 24 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-chlorophenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 2- amino-5-(4-morpholinopiperidin-1 -yl)isonicotinonitrile in step 3.
  • the compound of example 25 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 6-(4-morpholinopiperidin-1 -yl)pyridin-3-amine in step 3.
  • the compound of example 26 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 1 -(5-aminopyridin-2-yl)piperidine-4-carboxylic acid in step 3.
  • the compound of example 27 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(3-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 6-(4-(pyrimidin-2-yl)piperazin-1 -yl)-5-(trifluoromethyl)pyridin-3-amine in step 3.
  • the compound of example 28 was prepared analogous to the procedure described in example 11 by reaction of 1-(4-chlorophenyl)-1H-1,2,3-triazole-4-carbonyl azide with 5- chloro-6-(4-morpholinopiperidin-1-yl)pyridin-3-amine in step 3..
  • the compound of example 29 was prepared analogous to the procedure described in example 11 by reaction of 1-(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5-methyl-6-(4-morpholinopiperidin-1-yl)pyridin-3-amine in step 3.
  • the compound of example 30 was prepared analogous to the procedure described in example 11 by reaction of 1-(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5-methyl-6-(4-(pyrimidin-2-yl)piperazin-1 -yl)pyridin-3-amine in step 3.
  • the compound of example 31 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5-amino-2-(4-(pyrimidin-2-yl)piperazin-1 -yl)nicotinonitrile in step 3.
  • the compound of example 32 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(3-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 1 -(5-aminopyridin-2-yl)-4-phenylpiperidine-4-carbonitrile in step 3.
  • the compound of example 33 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5-amino-2-(piperidin-1 -yl)benzonitrile in step 3.
  • the compound of example 34 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5-amino-2-(2-oxa-6-azaspiro[3.3]heptan-6-yl)benzonitrile in step 3.
  • the compound of example 35 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-chlorophenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5- amino-2-(4-(cyclopropylmethyl)piperazin-1 -yl)benzonitrile in step 3.
  • the compound of example 36 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-chlorophenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5- amino-2-(4-methoxypiperidin-1 -yl)benzonitrile in step 3.
  • the compound of example 38 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(3-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5-amino-2-(4-morpholinopiperidin-1 -yl)benzonitrile in step 3.
  • Step 1) Synthesis of 1-(1-(4-chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(4- hydroxypiperidin-1 -yl)phenyl)urea
  • step 1 The compound of step 1 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-chlorophenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5- amino-2-(4-hydroxypiperidin-1 -yl)benzonitrile in step 3.
  • Step 2) Synthesis of 1-(1-(4-chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(4- hydroxypiperidin-1 -yl)phenyl)-1 ,3-dimethylurea
  • the compound of example 40 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5-amino-2-(4-(2-hydroxypropan-2-yl)piperidin-1 -yl)benzonitrile in step 3.
  • the compound of example 41 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5-amino-2-(piperazin-1 -yl)benzonitrile in step 3.
  • the compound of example 41 was further treated with hydrochloric acid to obtain hydrochloric salt of compound of example 41 A.
  • Step 2) 3-(4-fluorophenyl)-5-methylisoxazole-4-carbonyl azide (intermediate 2)
  • step 1 To a stirred solution of the compound obtained in step 1 [intermediate 1 ] (3.90 mmol) in acetone (10 mL) in an ice bath, was added trichloroacetonitrile (7.81 mmol), triphenyl phosphine (7.81 mmol) and sodium azide (4.68 mmol). The reaction mixture was stirred for 2 h at room temperature. On completion of reaction, the reaction was quenched with water (10 mL). The reaction mixture was extracted with ethyl acetate (25 mL). The organic layer was separated, washed with brine (1 0 mL), dried with Na 2 SO 4 , concentrated and purified using column chromatography (silica gel, ethyl acetate and petroleum ether gradient) to afford the title compound.
  • step 1 To a stirred solution of the compound obtained in step 1 [intermediate 1 ] (2.23 mmol) in dry acetone (5 mL) was added sodium azide (4.47 mmol), triethylamine (4.47 mmol) at
  • Step 3) 1-(3-cyano-4-(4-morpholinopiperidin-1-yl)phenyl)-3-(3-(4-fluorophenyl)-5- methylisoxazol-4-yl)urea (final compound)
  • step 2 [intermediate 2] (0.339 mmol) and 5-amino-2-(4- morpholinopiperidin-1 -yl)benzonitrile (0.339 mmol) in toluene (3 mL) was refluxed for 5 h. The reaction mixture was allowed to cool to the room temperature. The precipitated compound was filtered, washed with petroleum ether and cold dichloromethane, and dried to afford the title compound.
  • the compound of example 44 was prepared analogous to the procedure described in example 43 by reaction of 1-(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazole-4-carboxylic acid with 5-amino-2-(4-hydroxypiperidin-1 -yl)benzonitrile.
  • the compound of example 45 was prepared analogous to the procedure described in example 43 by reaction of 1-(4-chlorophenyl)-1 H-1 ,2,3-triazole-4-carboxylic acid with 5- amino-2-(4-morpholinopiperidin-1-yl)benzonitrile.
  • the compound of example 46 was prepared analogous to the procedure described in example 43 by reaction of 1 -(4-chlorophenyl)-1 H-1 ,2,3-triazole-4-carboxylic acid with 5- amino-2-(4-(pyrimidin-2-yl)piperazin-1 -yl)benzonitrile.
  • the compound of example 47 was prepared analogous to the procedure described in example 43 by reaction of 5-pentyl-1 -(3-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazole-4- carboxylic acid with 5-amino-2-(4-morpholinopiperidin-1 -yl)benzonitrile.
  • the compound of example 48 was prepared analogous to the procedure described in example 43 by reaction of 1 -(4-chlorophenyl)-1 H-1 ,2,3-triazole-5-carboxylic acid with 5- amino-2-(4-morpholinopiperidin-1 -yl)benzonitrile.
  • the compound of example 48 was further treated with hydrochloric acid to obtain hydrochloric salt of compound of example 48A.
  • Step 2) Synthesis of_4-chloro-N-(1-(4-chlorophenyl)-5-methyl-1 H-pyrazol-4-yl)-3- cyanobenzenesulfonamide (Intermediate 2)
  • step 1 To the stirred solution of compound obtained in step 1 [intermediate 1 ] (0.193 g, 0.81 9 mmol) in dichloromethane (10 mL) was added 1 -(4-chlorophenyl)-5-methyl-1 H-pyrazol- 4-amine hydrochloride (0.200 g, 0.81 9 mmol) and triethylamine (0.166 g, 1 .639 mmol) at room temperature. After completion of the reaction, the reaction was quenched with water. The crude compound obtained was extracted with dichloromethane, dried over anhydrous Na 2 S0 4 and purified using column chromatography (silica gel, methanol and dichloromethane gradient) to afford the title compound.
  • Step 3) Synthesis of N-(1-(4-chlorophenyl)-5-methyl-1 H-pyrazol-4-yl)-3-cyano-4- (4-morpholinopiperidin-1 -yl)benzenesulfonamide (final compound)
  • step 2 To the stirred solution of compound obtained in step 2 [intermediate 2] (0.50 g, 1 .228 mmol) in acetonitrile (10 mL) was added morpholinopiperidine (0.209 g, 1 .228 mmol) and triethylamine (0.248 g, 2.455 mmol). The resulting reaction mixture was heated to 65-70 e C for about 1 2-16 h. The solvent was removed, and the residue was diluted with ethylacetate and filtered through celite. The organic layers obtained were combined and dried over anhydrous Na 2 S0 4 . The solvent was removed and the crude product obtained was further purified using column chromatography (silica gel, methanol and dichloromethane gradient) to afford the title compound.
  • the compound of example 53 was prepared analogous to the procedure described in example 52 by reaction of 4-phenylthiazol-2-amine with 2-chloro-4-(4- (trifluoromethoxy)phenyl)pyrimidine.
  • the compound of example 55 was prepared analogous to the procedure described in example 52 by reaction of 6-fluorobenzo[d]thiazol-2-amine with 5-chloro-2-(4- morpholinopiperidin-1 -yl)benzonitrile.
  • the compound of example 57 was prepared analogous to the procedure described in example 56 by reaction of 2-chloro-4-(3,5-dimethoxyphenyl)pyrimidine with 5-amino-2- (4-(hydroxymethyl)piperidin-1 -yl)benzonitrile.
  • the compound of example 58 was prepared analogous to the procedure described in example 56 by reaction of 2-chloro-4-(2-methoxyphenyl)pyrimidine with 3- (trifluoromethyl)-4-(4-(pyrimidin-2-yl)piperazin-1 -yl)benzenamine.
  • the compound of example 59 was prepared analogous to the procedure described in example 56 by reaction of 2-chloro-4-(3-methoxyphenyl)pyrimidine with 2-(4- aminophenyl)-1 ,1 ,1 ,3,3,3-hexafluoropropan-2-ol.
  • the compound of example 60 was prepared analogous to the procedure described in example 56 by reaction of 2-chloro-4-(3-methoxyphenyl)pyrimidine with 5-amino-2-(4- hydroxypiperidin-1 -yl)benzonitrile.
  • the compound of example 61 was prepared analogous to the procedure described in example 56 by reaction of 2-chloro-4-morpholinopyrimidine with 5-amino-2-(4-(pyridin- 2-yl)piperazin-1 -yl)benzonitrile.
  • the compound of example 62 was prepared analogous to the procedure described in example 56 by reaction of 2-chloro-4-morpholinopyrimidine with 5-amino-2- morpholinobenzonitrile.
  • the compound of example 64 was prepared analogous to the procedure described in example 56 by reaction of 2,4-dichloro-6-methoxyquinoline with 5-amino-2-(4- morpholinopiperidin-1-yl)benzonitrile.
  • the compound of example 65 was prepared analogous to the procedure described in example 56 by reaction of 2-chloroquinoline with 5-amino-2-(4-morpholinopiperidin-1- yl)benzonitrile.
  • Step 1) Synthesis of (1-(3-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)methanol (intermediate 1)
  • Step-2 Synthesis of 1-(3-(trifluoromethyl) phenyl)-1 H-1 , 2, 3-triazole-4- carbaldehyde (intermediate 2)
  • step 1 To a stirred solution of the compound obtained in step 1 [intermediate 1 ] (1 1 .10 mmol) in chloroform (30 mL) was added manganese dioxide (1 1 .10 mmol). The resulting reaction mixture was heated at 70 °C for 3 h. On completion of the reaction, the reaction mixture was filtered through celite bed and residue was washed 2-3 times with chloroform. The combined filtrate was dried over anhydrous Na 2 SO 4 to afford the title compound.
  • Step-3 Synthesis of 5-((cyano(1-(3-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazol-4- yl)methyl)amino)-2-(4-(2-hydroxypropan-2-yl)piperidin-1-yl)benzonitrile (Final compound)
  • step 2 To a stirred solution of the compound obtained in step 2 [intermediate 2] (0.829 mmol) in tetrahydrofuran (20 mL) was added 5-amino-2-(4-(2-hydroxypropan-2-yl)piperidin-1 - yl)benzonitrile (0.829 mmol) and trifluoroacetic acid (0.035 mmol). The resulting reaction mixture was stirred at room temperature for 3 h. After the formation of imine compound, trimethylsilanecarbonitrile (1 .244 mmol) and tetrabutylammonium difluorotriphenylsilicate (1 .244 mmol) was added to the reaction mixture. The resulting reaction mixture was stirred for about 1 2-16 h.
  • the compound of example 67 was prepared analogous to the procedure described in example 66 by reaction of 1 -(3-(trifluoromethyl) phenyl)-1 H-1 , 2, 3-triazole-4- carbaldehyde with 5-amino-2-(4-(pyrimidin-2-yl)piperazin-1 -yl)benzonitrile in step 3.
  • Step 1) Synthesis of (1-(4-chlorophenyl)-5-methyl-1 H-pyrazol-4-yl)methanol (intermediate 1)
  • Step-2 Synthesis of 1-(4-chlorophenyl)-5-methyl-1 H-pyrazole-4-carbaldehyde (intermediate 2)
  • step 2 To a stirred solution of the compound obtained in step 2 [intermediate 2] (0.829 mmol) in tetrahydrofuran (20 ml_) was added 5-amino-2-chlorobenzonitrile (0.829 mmol) and trifluoroacetic acid (0.035 mmol). The resulting reaction mixture was stirred at room temperature for 3 h. After the formation of imine compound, sodium borohydride was added to the reaction mixture. The resulting reaction mixture was stirred for about 12- 16 h. The solvent was removed and the crude residue was further purified using column chromatography (silica gel, methanol and dichloromethane gradient) to afford the title compound.
  • the compound of example 69 was prepared analogous to the procedure described in example 68 by reaction of 1 -(4-chlorophenyl)-5-methyl-1 H-pyrazole-4-carbaldehyde with 5-amino-2-(4-morpholinopiperidin-1 -yl)benzonitrile in step 3.
  • the compound of example 71 was prepared analogous to the procedure described example 70 by reaction of 2-hydroxy-2-(1 -(3-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazol yl)acetic acid with 5-amino-2-(4-methoxypiperidin-1 -yl)benzonitrile.
  • Biological Assays The biological activity of the compounds of the present invention as IL-17 and TNF-a inhibitor can be confirmed by a number of biological assays known in the art.
  • the exemplified biological assays, given below, have been carried out with certain representative compounds of the formula I (referred to as the test compounds) synthesized in the above examples.
  • Test System Human peripheral blood cells isolated from healthy volunteers.
  • PBMC peripheral blood mononuclear cells
  • Cytokine release assay The cells were uniformly plated in 96-well tissue culture plates (Nunc 167008) at a concentration of 1 x1 0 5 cells/well. After plating, different concentrations (0.003, 0.01 , 0.03, 0.1 , 0.3, 1 , 3 and 1 0 ⁇ g/mL) of the test compounds dissolved in dimethylsulfoxide (DMSO) were added to the cells and incubated at 37 °C for 30 min in 5 % CO 2 atmosphere. The final concentration of DMSO was maintained at 0.5 %. The vehicle (0.5 % DMSO) was used as control.
  • DMSO dimethylsulfoxide
  • the cells were then stimulated with 25 ng/mL of PMA (P1585, SIGMA) and 1 ⁇ ionomycin (SIGMA, I0634).
  • the plates were incubated in 5 % CO 2 air humidified atmosphere at 37 °C for 48 h. At the end of 48 h, the supernatant from the cells were collected and ELISA was performed to detect the level of IL-17 (R&D systems, DY31 7E) and TNF-a (BD Biosciences, 555212) in the supernatant.
  • Lymphocyte proliferation assay by 3 H thymidine uptake The cells were uniformly plated in 96-well tissue culture plates (Nunc 167008) at a concentration of 1 x1 0 5 cells/well. After plating, different concentrations (0.003, 0.01 , 0.03, 0.1 , 0.3, 1 , 3 and 1 0 ⁇ g/mL) of the test compounds dissolved in DMSO were added to the cells and incubated at 37 °C for 30 min in 5 % CO 2 atmosphere. The final concentration of DMSO was maintained at 0.5 %. The vehicle (0.5 % DMSO) was used as control.
  • the cells were then stimulated with 25 ng/mL of PMA (P1585, SIGMA) and 1 ⁇ ionomycin (SIGMA, I0634).
  • the plates were incubated in 5 % CO 2 air humidified atmosphere at 37 °C for 48 h.
  • cells are pulsed with 0.5 ⁇ / well of 3[H]-Thymidine (BRIT, LCT3).
  • BRIT, LCT3 3[H]-Thymidine
  • the cells were washed twice with PBS to remove excess radioactivity. Radioactivity in the incorporated cells was captured on lysis with Microscint-40, Perkin Elmer and measured by using a liquid scintillation counter (TopCount, Perkin Elmer).
  • the anti-proliferative effect of the test compounds was measured using the following formula.
  • Controls consisted of PBMC with PMA and ionomycin (Stimulated) and PBMC with RPMI (Un-stimulated)
  • the test compounds exhibited IL-17 and TNF-a inhibitory activity in the cytokine release assay in which the compounds were tested.
  • the IC 50 values for test compounds inhibiting IL-17 are given in Table 1
  • the IC50 values for test compounds inhibiting TNF-a is given in Table 2.
  • Table 1 IC 50 values of the test compounds (IL-17 inhibition)
  • the IC 50 values determined for the test compounds are indicative of their IL17 and TNF-a inhibitory activity.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Diabetes (AREA)
  • Emergency Medicine (AREA)
  • Endocrinology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Virology (AREA)
  • Pulmonology (AREA)
  • Dermatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention provides heterocyclyl compounds of formula I, isotopic forms, stereoisomers or tautomers thereof, or pharmaceutically acceptable salts, solvates, N- oxides, S-oxides and polymorphs thereof, and processes for their preparation. The invention further relates to pharmaceutical compositions containing the compounds and their use in the treatment of inflammatory diseases, autoimmune disorders and other related disorders. The heterocyclyl compounds find use as Interleukin 17 (IL-17) inhibitors and Tumor Necrosis Factor alpha (TNF-α) inhibitors.

Description

HETEROCYCLYL COMPOUNDS AND USES THEREOF
FIELD OF THE INVENTION The present invention relates to heterocyclyl compounds (the compounds of formula I as described herein), processes for their preparation, pharmaceutical compositions containing the said compounds, their use as Interleukin 17 (IL-1 7) inhibitors or Tumor Necrosis Factor alpha (TNF-a) inhibitors, and methods for using said compounds in the treatment or prevention of diseases or disorders mediated by IL-17 and TNF-a, such as autoimmune or inflammatory diseases or disorders; and metabolic diseases or disorders.
BACKGROUND OF THE INVENTION Inflammation is part of the non-specific immune response that occurs in reaction to body injury (e.g. trauma, ischemia, and foreign particles) or infection (e.g. bacterial or viral infection). Inflammation is an inherent part of various autoimmune or inflammatory disorders, such as rheumatoid arthritis, atherosclerosis, inflammatory myopathies, septic shock syndrome, ulcerative colitis, inflammatory bowel disease, psoriasis, multiple sclerosis, and others. It has been reported that inflammation is commonly associated with the metabolic diseases and disorders such as insulin resistance and obesity (Brent E. Wisse, Journal of the American Society of Nephrology, 2004, 15, 2792-2800). Inflammation can be classified as either acute or chronic. Infiltration of innate immune system cells, specifically neutrophils and macrophages, characterizes the acute inflammation, while infiltration of T lymphocytes and plasma cells are features of chronic inflammation. Recent studies have established that Th 7 cells are involved in mediating inflammatory diseases and immune defense against extracellular bacteria. Th17 cells are a subset of T cells that produce pro-inflammatory cytokines such as the interleukins (IL-6, IL-17A, IL-17F, IL-21 , IL-22), and TNF-a (Tumor necrosis factor-alpha). Th 7 cells can be generated from naive precursors in the presence of TGF-β and IL-6 or IL-21 , which can be further augmented by TNF or I L-1 β which are the inflammatory cytokines that inhibit Th1 and Th2 cell differentiation. Developments of Th1 , Th2, and regulatory T cells (T reg cells) are specified by transcription factors such as Tbet, GATA3, and Foxp3, respectively, whereas that of Th-|7 cells appears to depend on the retinoid-related orphan receptor RORgt, RORa, and signal transducer and activator of transcription (J Exp Med. 201 1 October 24; 208(1 1 ): 2321 -2333).
The pro-inflammatory cytokine, Interleukin 17 (family) comprises six members namely IL-17A, IL-17B, IL-17C, IL-17D, IL-17E, and IL-1 7F, out of which IL-1 7A and IL-1 7F are clearly associated with inflammation as these are produced by Th 7 cells. IL-17 deploys neutrophils to the site of inflammation and this ability imparts IL-17 with a key role in several types of diseases. IL-1 7 is predominantly produced by activated memory T cells and acts on the cell by binding to a specific cell surface receptor IL-1 7R. Human IL-1 7 is a 20-30 kDa glycoprotein configured with 155 amino acids, comprising a signal peptide at the N-end. The expression of IL-17 has been found to be elevated in patients with rheumatoid arthritis, systemic lupus erythematosus (SLE), Behget's disease, inflammatory bowel disease, asthma, multiple sclerosis, and other related diseases or disorders. It has further been reported that IL-1 7 plays a pivotal role in the obesity induced inflammatory conditions (Mario Galgani et al., Journal of Leukocyte Biology, 201 0, 87, 17-1 8).
It is indicated herein above that Th 7 cells, which stimulate the secretion of a wide range of pro-inflammatory cytokines including IL-17A and TNF-oc among others, appear to depend on the retinoid-related orphan receptor RORgt. Accordingly, transcription factor 'Retinoid-related orphan receptor gamma t (RORgt)' plays a pivotal role in the production of IL-17. RORgt is a member of steroid hormone nuclear receptor superfamily and it is encoded by RORc gene. RORgt is exclusively expressed in the T cells of the immune system. It is known that RORgt controls differentiation of proinflammatory T helper 17 (Th17) cells. It is reported that RORgt is required for the induction of IL-1 7 transcription and for the manifestation of Th17-dependent autoimmune disease. Multiple other transcription factors have also been shown to be important for the development of Th1 7 cells: BATF, STAT3, IRF4, RUNX1 , and ΙκΒζ. Unlike RORgt, these factors are not NHRs (Nuclear hormone receptors) and thus do not contain ligand binding domains, which renders them less attractive targets for drug development. Tumor Necrosis Factor-alpha (TNF-a) / cachectin is a pleiotrophic cytokine that plays pivotal role in inflammatory disorders and conditions. TNF-a is a soluble homotrimer of 17 kD protein subunits. TNF-a is produced by monocytes, macrophages, and T lymphocytes. It shows self-regulatory, growth stimulating and growth inhibitory properties. Following to infection or injury, TNF-a initiates inflammatory cascade of cytokines, thereby recruiting macrophage and neutrophils to a site of infection. It activates granulocytes, and increases Major Histocompatibility Complex class II (MHC Class II) expression. It also promotes secretion of matrix metalloproteinases (MMPs), leading to cartilage matrix degradation. Because TNF-a initiates an inflammatory cascade, and has been found to be increased in close proximity to inflamed or injured tissue, TNF-a inhibition is an appealing target for the development of new antiinflammatory drugs. Although, significant advances have been made in providing treatment options for inflammatory and autoimmune disorders, the treatment options do not provide satisfactory results for all the patients due to, for example, detrimental side effects or efficacy, or inadequacy as to patient compliance. Treatments for autoimmune and inflammatory disorders vary depending on the particular medical disorder, and often involve use of immunosuppressive drugs. In view of this, there is a continuing need to provide newer and effective treatment options to the patients suffering from autoimmune, inflammatory and related disorders.
It is evident from the above discussion that the pro-inflammatory cytokines such as IL- 17 and TNF-a constitute attractive targets for new drug developments for inflammatory disorders, autoimmune disorders and other related disorders. The present invention addresses the need of newer and effective treatment options to the patients suffering from autoimmune, inflammatory and related disorders by providing compounds that are IL-17 and/or TNF-a inhibitors.
SUMMARY OF THE INVENTION
In one aspect, the present invention relates to a compound of formula I (as described herein), or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, S-oxide or N-oxide thereof. According to another aspect of the present invention, there are provided processes for the preparation of the compounds of formula I, or pharmaceutically acceptable salts thereof.
According to a further aspect of the present invention, there is provided a pharmaceutical composition comprising one or more of the compounds of formula I or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof; and at least one pharmaceutically acceptable carrier or excipient.
According to a further aspect of the present invention, there is provided a compound of formula I or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof; for use as IL-17 inhibitor. According to another aspect of the present invention, there is provided a compound of formula I or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof; for use as TNF-a inhibitor.
According to yet another aspect of the present invention, there is provided a compound of formula I or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof; for use in the treatment of a disease or a disorder mediated by IL-1 7 or TNF-a.
According to a further aspect of the present invention, there is provided a method for the treatment of a disease or disorder mediated by IL-1 7 or TNF-a, comprising administering to a subject in need thereof; a therapeutically effective amount of the compound of formula I or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof. According to yet another aspect of the present invention, there is provided use of a compound of formula I or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof; for the manufacture of a medicament for the treatment of a disease or a disorder mediated by IL-1 7 or TNF-a. In a further aspect, the present invention relates to use of a compound of formula I or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt or a solvate thereof; in combination with at least one further therapeutically active agent for the treatment of a disease or a disorder mediated by IL-1 7 or TNF-a.
One or more further aspects of the present inventions are discussed in detail herein below. These and other objectives and advantages of the present invention will be apparent to those skilled in the art from the following description.
DETAILED DESCRIPTION OF THE INVENTION According to the first aspect, the present invention relates to a compound of formula I:
Figure imgf000006_0001
Formula I or an isotopic form, a stereoisomer or a tautomer, a pharmaceutically acceptable salt, solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof; wherein,
A is triazole, pyrazole, imidazole, oxazole, isoxazole, thiazole, pyrrole, a 5- or 6- membered saturated heterocyclic ring system or a heteroaryl ring system selected from:
Figure imgf000006_0002
wherein * indicates the point of attachment to L; Ri and R3 are independently selected from the group consisting of hydrogen, hydroxy, halogen, cyano, nitro, (CrC6)-alkyl, (CrC6)-alkoxy, (CrC6)-alkoxyalkyl, (CrC6)- alkylamino, (C3-Ci2)-cycloalkyl, 0-(C3-Ci2)-cycloalkyl, halo(CrC6)alkyl, halo(d- C6)alkoxy, (C6-C14)-aryl, heterocyclyl, 0-(C6-Ci4)aryl, (C6-Ci4)ar(C C6)alkyl, (C6- Ci4)ar(C C6)alkyloxy, heteroaryl, C(0)Ra, C(0)ORa, S(0)nRa, NRaRb and C(0)NRaRb; n is an integer from 0 to 2;
R2 is (CrC6)-alkyl, (C1-C6)-alkoxy, (C Ce lkoxyid-C^- lkyl, (C3-C12)-cycloalkyl, O- (C3-C12)-cycloalkyl, (C6-Ci4)-aryl, 0-(C6-C14)aryl, (C6-Ci4)ar(CrC6)alkyl, (C6-Ci4)ar(C C6)alkyloxy, heterocyclyl or heteroaryl;
R2 together with R can form a 5- or 6- membered saturated or unsaturated ring optionally containing 1 to 3 heteroatoms independently selected from the group consisting of O, N and S, which ring is unsubstituted or substituted with 1 to 5 groups independently selected from the group consisting of halogen, hydroxy, nitro, cyano, (d- C6)-alkyl, (C C6)-alkoxy, (C C6)-alkoxyalkyl, (CrC6)-alkylamino, (C3-Ci2)-cycloalkyl, O- (C3-C12)-cycloalkyl, halo(C C6)alkyl, halo(C C6)alkoxy, (C6-C14)-aryl, 0-(C6-Ci4)aryl, (C6-Ci4)ar(Ci-C6)alkyl, (C6-Ci4)ar(Ci-C6)alkyloxy, heterocyclyl, heteroaryl, C(0)Ra, C(0)ORa, S(0)nRa, NRaRb and C(0)NRaRb;
L is *N(R4)-C(0)-N(R5), *C(0)N(R5), *N(R4)-C(0), *CH(R6)-C(0)-N(R5), *CH(R6)N(R5), *N(R4)-S(0)2 or *NR4; wherein * indicates the point of attachment to A;
R , R5 and R6 are independently selected from the group consisting of hydrogen, hydroxy, cyano, (Ci-C6)-alkyl, (Ci-C6)-alkoxy, (Ci-C6)-alkoxyalkyl, (Ci-C6)-alkylamino, (C3-Ci2)-cycloalkyl, 0-(C3-Ci2)-cycloalkyl, halo(C C6)alkyl, halo(C C6)alkoxy, hydroxy(Ci-C6)-alkyl, C(0)Ra, C(0)ORa, S(0)nRa, P(0)n(0(Ci-C6)alkyl)2, O-P(O) (0(C C6)alkyl)2 and C(0)NRaRb;
Xi , X2, X4 and X5 are independently selected from N and C(R7); wherein R7 is hydrogen, halogen, hydroxy, nitro, cyano, (CrC6)-alkyl, (CrC6)-alkoxy, (d-C6)- alkoxy(CrC6)-alkyl, (Ci-C6)-alkylamino, (C3-Ci2)-cycloalkyl, halo(C C6)alkyl, halo(C C6)alkoxy, (C2-C6)-alkenyl, (C2-C6)-alkynyl, (C6-Ci4)-aryl, 0-(C6-Ci4)aryl, (C6-Ci4)ar(C C6)alkyl, (C6-Ci4)ar(Ci-C6)alkyloxy, heterocyclyl, O-heterocyclyl, heteroaryl, C(0)Ra, C(0)ORa, S(0)nRa, P(0)n(0(Ci -Ce)alkyl)2l 0-P(0)(0(C C6)alkyl)2, NRaRb or C(0)NRaRb;
X3 is N or C(R8), wherein R8 is hydrogen, halogen, hydroxy, cyano, nitro, (CrC6)-alkyl, (Ci -C6)-alkoxy, (Ci -C6)-alkoxy(Ci -C6)alkyl, (C C6)-alkylamino, (C3-C12)-cycloalkyl, O- (C3-Ci2)-cycloalkyl, halo(C C6)alkyl, halo(C C6)alkoxy, (C6-Ci4)-aryl, 0-(C6-Ci4)aryl, (C6-Ci4)ar(CrC6)alkyl, (C6-Ci4)ar(CrC6)alkyloxy, heterocyclyl, O-heterocyclyl, heteroaryl, C(0)Ra, C(0)ORa, S(0)nRa, NRaRb or C(0)NRaRb;
provided that no more than three of the X to X5 positions can be simultaneously N; when two or more of X-i , X2, X4 and X5 represent C(R7) then R7 groups on the two adjacent carbon atoms can form a 3-7 membered unsaturated or saturated fused ring optionally containing 1 to 3 heteroatoms independently selected from the group consisting of O, N and S; wherein the ring can be unsubstituted or substituted with 1 -3 groups of R0; or
when any one of X2 and X4 represent C(R7) and X3 represents C(R8) then the R7 group together with the R8 group can form a 3-7 membered fused ring optionally containing 1 to 3 heteroatoms selected from O, N or S; wherein the ring can be unsubstituted or substituted with 1 -3 groups of Rc;
Ra and Rb are independently selected from the group consisting of hydrogen, (CrC6)- alkyl, (C3-C12)-cycloalkyl, halo(C C6)alkyl, (C6-C14)-aryl, (C6-Ci4)ar(C C6)alk l, heterocyclyl, heteroaryl, and S(0)nRa; or
Ra and Rb, can form a 3-7 membered saturated or unsaturated ring optionally containing one or more heteroatoms selected from N, O or S, which is unsubstituted or substituted with one or more groups independently selected from the group consisting of halogen, hydroxy, cyano, nitro, (C C6)-alkyl, (CrC6)-alkoxy, (CrC6)-alkylamino, (C3- Ci2)-cycloalkyl, 0-(C3-Ci2)-cycloalkyl, halo(CrC6)alkyl, halo(CrC6)alkoxy, (C6-C )-aryl, 0-(C6-C )aryl, (C6-Ci4)ar(Ci-C6)alkyl, heterocyclyl, heteroaryl, C(0)Ra, C(0)ORa, S(0)nRa, P(0)n(0(Ci -C6)alkyl)2j 0-P(0)(0(C C6)alkyl)2, NRaRb and C(0)NRaRb; wherein:
each of the (CrC6)alkyl and (C-|-C6)-alkoxy can be unsubstituted or substituted with one or more groups independently selected from the group consisting of halogen, hydroxy, nitro, cyano, (Ci-C6)-alkoxy-(Rc)o-3, (C3-Ci2)-cycloalkyl-(Rc)o-3! 0-(C3-Ci2)-cycloalkyl- (Rc)o-3, halo(CrC6)alkyl, halo(CrC6)alkoxy, (C6-Ci4)-aryl-(Rc)0-3, 0-(C6-Ci4)aryl-(Rc)o-3, (C6-C14)ar(CrC6)alkyl-(Rc)o-3, heterocyclyl-(Rc)0-3, heteroaryl-(Rc)0-3, C(0)Ra, C(0)ORa, S(0)nRa, P(0)n(0(Ci-C6)alkyl)2, 0-P(0)(0(Ci-C6)alkyl)2, NRaRb and C(0)NRaRb; heterocyclyl refers to a 3-1 2 membered saturated or partially unsaturated monocyclic or bicyclic ring system containing one to four identical or different heteroatoms independently selected from the group consisting of N, S and O; heteroaryl refers to a 5-10 membered aromatic monocyclic or bicyclic ring system containing one to four identical or different heteroatoms independently selected from the group consisting of N, S and O; each of the (C2-C6)-alkenyl, (C2-C6)-alkynyl, (C3-Ci2)-cycloalkyl, (C6-Ci4)-aryl, (C6- Ci4)ar(CrC6)alkyl, heterocyclyl and heteroaryl can be unsubstituted or substituted with one or more groups independently selected from the group consisting of halogen, hydroxy, nitro, cyano, (Ci-C6)alkyl-(Rc)o-3, (CrC6)-alkoxy-(Rc)o-3, (C3-C12)-cycloalkyl- (Ftc)o-3, 0-(C3-Ci2)-cycloalkyl-(Rc)o-3, halo(CrC6)alkyl, halo(C C6)alkoxy, (C6-C )-aryl- (Rc)o-3, 0-(C6-Ci4)aryl-(Rc)o-3, (C6-Ci4)ar(Ci-C6)alkyl-(Rc)o-3, heterocyclyl-(Rc)0-3, heteroaryl-(Rc)o-3, C(0)Ra, C(0)ORa, S(0)nRa, P(0)n(0(Ci-C6)alkyl)2, O-P(O) (0(C C6)alkyl)2, NRaRb and C(0)NRaRb; and
Rc is halogen, hydroxy, cyano, nitro, (Ci-C6)-alkyl-(Rc)o-i , (Ci-C6)-alkoxy-(Rc)o-i , (C3- Ci2)-cycloalkyl-(Rc)o-i , O-(C3-Ci2)-cycloalkyl-(Rc)0-i , hydroxy(Ci-C6)-alkyl-(Rc)o-i , halo(CrC6)alkyl, halo(C C6)alkoxy, (C6-Ci4)-aryl-(Rc)0-i, O-(C6-Ci4)aryl-(Rc)0-i, (C6- Ci4)ar(CrC6)alkyl-(Rc)0-i, heterocyclyl-(Rc)0-i , heteroaryl-(Rc)0-i , C(0)Ra, C(0)ORa, SRa, S(0)nRa, P(0)n(0(Ci-C6)alkyl)2, 0-P(0)(0(C C6)alkyl)2, NRaRb or C(0)NRaRb; and with the provisos that:
(a) when A is pyrazole then L is a group other than *C(0)N(R5) and *N(R4)-C(0); and
(b) when A is pyrazole and L is *N(R4)-C(0)-N(R5), then said L group is not attached to 5-position of pyrazole.
Definitions
Unless otherwise indicated, the following definitions are set forth to illustrate and define the meaning and scope of the various terms used to describe the invention herein and the appended claims. These definitions should not be interpreted in the literal sense as they are not general definitions and are relevant only for this application.
It will be understood that "substitution", "substituted" or "substituted with" means that one or more hydrogens of the specified moiety are replaced with a suitable substituent and includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and results in a stable compound.
The terms "a", "an" and "the" refers to "one or more" when used in the subject specification, including the claims. Thus, for example, reference to "a compound" may include a plurality of such compounds, or reference to "a disease" or "a disorder" includes a plurality of diseases or disorders.
It should also be noted that the term "or" is generally employed in its sense including "and/or" unless the content clearly dictates otherwise.
Also, use of "(s)" as part of a term, includes reference to the term singly or in plurality, e.g. the term compound(s) may indicate a single compound or more compounds.
The term "independently" when used in the context of selection of substituents for a variable, it means that where more than one substituent is selected from a number of possible substituents, those substituents may be the same or different.
As used herein, the term "(C-i-C6)-alkyr or "alkyl" whether used alone or as part of a substituent group, refers to the radical of saturated aliphatic groups, including straight or branched-chain alkyl groups. An alkyl group can have a straight chain or branched chain containing carbon atoms. A straight-chain or branched chain alkyl has six or fewer carbon atoms in its backbone, for instance, CrC6 for straight chain and C3-C6 for branched chain. Suitable alkyl groups containing from one to six carbon atoms, for example, include methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, iso-butyl, sec-butyl, n-pentyl, isopentyl, 2-pentyl, 3-pentyl, neo-pentyl, n-hexyl, isohexyl, 2-hexyl and 3- hexyl.
Furthermore, unless stated otherwise, the alkyl groups may be unsubstituted or substituted with one or more substituents. A substituted alkyl refers to a (CrC6)-alkyl substituted with one or more groups, preferably 1 -3 groups independently selected from the group consisting of halogen, hydroxy, nitro, cyano, (C-i-C6)-alkoxy-(Rc)o-3, (C3-C12)- cycloalkyl-(Rc)o-3, 0-(C3-Ci2)-cycloalkyl-(Rc)o-3, halo(C C6)alkyl, halo(C C6)alkoxy, (C6- Ci4)-aryl-(R0)o-3, 0-(C6-Ci4)aryl-(Rc)o-3, (C6-Ci4)ar(Ci-C6)alkyl-(Rc)o-3, heterocyclyl-(Rc)0- 3, heteroaryl-(Rc)o-3, C(0)Ra, C(0)ORa, S(0)nRa, P(0)n(0(Ci-C6)alkyl)2, 0-P(0)(0(C C6)alkyl)2, NRaRb and C(0)NRaRb;
wherein n is 0-2; Ra and Rb are independently selected from the group consisting of hydrogen, (C C6)-alkyl, (C3-Ci2)-cycloalkyl, halo(C C6)alkyl, (C6-Ci4)-aryl, (C6- Ci4)ar(CrC6)alkyl, heterocyclyl, heteroaryl, and S(0)nRa; or
Ra and Rb can form a 3-7 membered saturated or unsaturated ring optionally containing one or more heteroatoms selected from N, O or S, which is unsubstituted or substituted with one or more groups independently selected from the group consisting of halogen, hydroxy, cyano, nitro, (Ci-C6)-alkyl, (Ci-C6)-alkoxy, (Ci-C6)-alkylamino, (C3-Ci2)- cycloalkyl, 0-(C3-Ci2)-cycloalkyl, halo(C C6)alkyl, halo(C C6)alkoxy, (C6-C14)-aryl, O- (C6-C14)aryl, (C6-Ci4)ar(C C6)alkyl, heterocyclyl, heteroaryl, C(0)Ra, C(0)ORa, S(0)nRa, P(0)n(0(C C6)alkyl)2, 0-P(0)(0(CrC6)alkyl)2, NRaRb and C(0)NRaRb; and
Rc is halogen, hydroxy, cyano, nitro, (C C6)-alkyl-(Rc)0-i, (CrC6)-alkoxy-(Rc)o-i, (C3- Ci2)-cycloalkyl-(Rc)o-i, O-(C3-Ci2)-cycloalkyl-(Rc)0-i, hydroxy(CrC6)-alkyl-(Rc)o-i, halo(CrC6)alkyl, halo(C C6)alkoxy, (C6-Ci4)-aryl-(Rc)0-i, O-(C6-Ci4)aryl-(Rc)0-i, (C6- Ci4)ar(Ci-C6)alkyl-(Rc)o-i, heterocyclyl-(Rc)0-i, heteroaryl-(Rc)0-i, C(0)Ra, C(0)ORa, SRa, S(0)nRa, P(0)n(0(Ci-C6)alkyl)2, 0-P(0)(0(C C6)alkyl)2, NRaRb or C(0)NRaRb.
Examples of substituted alkyls include, but are not limited to, chloromethyl, hydroxymethyl, hydroxyethyl, aminoethyl and the like.
The term "halogen" refers to a fluorine, chlorine, bromine, or iodine atom.
When the alkyl group is substituted with one or more halogens, it is specifically referred to as "halo(Ci-C6)alkyr or "haloalkyl". A monohalo(Ci-C6)alkyl radical, for example, may have a chlorine, bromine, iodine or fluorine atom. Dihalo and polyhalo(CrC6)alkyl radicals may have two or more of the same or different halogen atoms. Examples of halo(CrC6)alkyl include, but are not limited to, chloromethyl, dichloromethyl, trichloromethyl, dichloroethyl, dichloropropyl, fluoromethyl, difluoromethyl, trifluoromethyl, pentafluoroethyl, heptafluoropropyl, difluorochloromethyl, dichlorofluoromethyl, difluoroethyl, difluoropropyl or the like groups. As used herein, the term "halo(d-C6)alkoxy" or "haloalkoxy" refers to radicals wherein one or more of the hydrogen atoms of the alkoxy group are substituted with one or more halogens. Representative examples of "haloalkoxy" or "halo(CrC6)alkoxy" groups include, but not limited to, difluoromethoxy (OCHF2), trifluoromethoxy (OCF3) or trifluoroethoxy (OCH2CF3).
As used herein, the term "(C2-C6)-alkenyr or "alkenyl", as used herein; alone or as part of a substituent group, refers to a straight or branched chain hydrocarbon radical containing the indicated number of carbon atoms and at least one carbon-carbon double bond (two adjacent sp2 carbon atoms). For example, (C2-C6)-alkenyl refers to an alkenyl group having 2 to 6 (both inclusive) carbon atoms. Depending on the placement of double bond and substituents if any, the geometry of the double bond may be entgegen (E), or zusammen (Z), cis or trans. Examples of alkenyl include, but are not limited to, vinyl, allyl and 2-propenyl.
Unless stated otherwise, the alkenyl groups can be unsubstituted or substituted with one or more groups, preferably 1 -3 groups independently selected from the group consisting of halogen, hydroxy, nitro, cyano, (CrC6)alkyl-(Rc)o-3, (Ci-C6)-alkoxy-(Rc)o-3, (C3-C12)-cycloalkyl-(Rc)o-3, 0-(C3-Ci2)-cycloalkyl-(Rc)o-3, halo(CrC6)alkyl, halo(C C6)alkoxy, (C6-C14)-aryl-(Rc)0-3, O-(C6-C14)aryl-(Rc)0-3, (C6-C14)ar(C C6)alkyl-(Rc)o-3, heterocyclyl-(Rc)o-3, heteroaryl-(Rc)0-3, C(0)Ra, C(0)ORa, S(0)nRa, P(0)n(0(C C6)alkyl)2, O-P(O) (0(C C6)alkyl)2, NRaRb and C(0)NRaRb; wherein Ra, Rb, Rc and n are as defined herein above.
As used herein, the term "alkynyl" or "(C2-C6)alkynyl" whether used alone or as part of a substituent group, refers to a straight or branched chain hydrocarbon radical containing the indicated number of carbon atoms and at least one carbon-carbon triple bond (two adjacent sp carbon atoms). For example, (C2-C6)alkynyl refers to an alkynyl group having 2 to 6 (both inclusive) carbon atoms. Examples of (C2-Ce)alkynyl include, but are not limited to, ethynyl, 1 -propynyl, 3-propynyl and 3-butynyl. Unless stated otherwise, the "alkynyl" can be unsubstituted or substituted with one or more groups, preferably 1 - 3 groups independently selected from the group consisting of halogen, hydroxy, nitro, cyano, (Ci-C6)alkyl-(Rc)0-3, (Ci-C6)-alkoxy-(Rc)o-3, (C3-Ci2)-cycloalkyl-(Rc)0-3, 0-(C3-Ci2)- cycloalkyl-(Rc)o-3, halo(C C6)alkyl, halo(C C6)alkoxy, (C6-Ci4)-aryl-(Rc)o-3, 0-(C6- Ci4)aryl-(Rc)o-3, (C6-C14)ar(C1-C6)alkyl-(Rc)o-3, heterocyclyl-(Rc)o-3, heteroaryl-(Rc)0-3, C(0)Ra, C(0)ORa, S(0)nRa, P(0)n(0(Ci-C6)alkyl)2, O-P(O) (0(Ci-C6)alkyl)2l NRaRb and C(0)NRaRb; wherein Ra, Rb, Rc and n are as defined herein above. As used herein, the term "(C3-C-|2)-cycloalky or "cycloalkyl" whether used alone or as part of a substituent group, refers to a saturated or partially unsaturated cyclic hydrocarbon radical including 1 , 2 or 3 rings and including a total of 3 to 12 carbon atoms forming the rings. The term cycloalkyl includes bridged, fused and spiro ring systems. For example, (C3-C 2)-cycloalkyl refers to a cycloalkyl group having 3 to 8 (both inclusive) carbon atoms. Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, adamantyl, norbornyl, bicyclo[2.1 .0]pentane, bicyclo[2.2.1 ]heptyl, bicyclo[2.2.1 ]hept-2-ene, spiro[3.3]heptane, 1 ,2,3,3a-tetrahydropentalene and the like. Unless stated otherwise, the "cycloalkyl" group can be unsubstituted or substituted with one or more groups, preferably 1 -3 groups independently selected from the group consisting of halogen, hydroxy, nitro, cyano, (Ci-C6)alkyl-(Rc)o-3, (Ci-C6)-alkoxy-(Rc)o-3, (C3-C12)-cycloalkyl-(Rc)o-3, 0-(C3-Ci2)-cycloalkyl-(Rc)o-3, halo(CrC6)alkyl, halo(C C6)alkoxy, (C6-Ci4)-aryl-(Rc)o-3, 0-(C6-Ci4)aryl-(Rc)o-3, (C6-Ci4)ar(Ci-C6)alkyl-(Rc)o-3, heterocyclyl-(Rc)o-3, heteroaryl-(Rc)0-3, C(0)Ra, C(0)ORa, S(0)nRa, P(0)n(0(C C6)alkyl)2, O-P(O) (0(CrC6)alkyl)2, NRaRb and C(0)NRaRb; wherein Ra, Rb, Rc and n are as defined herein above.
As used herein, the term "(CrC6)alkoxy" or "alkoxy" refers to a (C C6)-alkyl having an oxygen radical attached thereto. Representative alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy, isopropoxy, n-butoxy, isobutoxy, t-butoxy and the like.
The term, (C3-Ci2)-cycloalkyloxy or cycloalkyloxy or 0-(C3-Ci2)-cycloalkyl refers to a (C3-Ci2)-cycloalkyl having an oxygen radical attached thereto. Representative cycloalkyloxy groups include, but are not limited to, cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, cyclohexyloxy and the like.
Unless stated otherwise, the alkoxy and cycloalkyloxy can be unsubstituted or substituted with one or more groups selected from the group consisting of halogen, hydroxy, nitro, cyano, (C1-C6)alkyl-(Rc)0-3, (Ci-C6)-alkoxy-(Rc)<w, (C3-C12)-cycloalkyl- (Rc)o-3, 0-(C3-Ci2)-cycloalkyl-(Rc)o-3, halo(C C6)alkyl, halo(CrC6)alkoxy, (C6-Ci )-aryl- (Rc)o-3, 0-(C6-Ci4)aryl-(Rc)o-3! (C6-Ci4)ar(Ci-C6)alkyl-(Rc)o-3, heterocyclyl-(Rc)o-3, heteroaryl-(Rc)o-3, C(0)Ra, C(0)ORa, S(0)nRa, P(0)n(0(Ci-C6)alkyl)2, O-P(O) (0(C C6)alkyl)2, NRaRb and C(0)NRaRb; wherein Ra, Rb, Rc and n are as defined herein above.
Examples of substituted (CrC6)-alkoxy include, but are not limited to, chloromethoxy, 2-cyanoethoxy, trifluoromethoxy and benzyloxy group. A benzyloxy group refers to a benzyl having an oxygen radical attached thereto.
The term "(Ci-C6)-alkoxyalkyl" or "alkoxyalkyl" as used herein refers to a (C-|-C6)-alkyl group, which is substituted by a (CrC6)-alkoxy group. The term "(C6-C 4)-aryr or "aryl" as used herein refers to monocyclic or bicyclic hydrocarbon groups having 6 to 14 ring carbon atoms, preferably 6 to 10 carbon atoms in which the carbocyclic ring(s) present have a conjugated pi electron system. Examples of (C6-Ci4)-aryl residues are phenyl, naphthyl, fluorenyl or anthracenyl. Preferred examples of (C6-Cu)-aryl residues are phenyl or naphthyl. Aryl groups can be unsubstituted or substituted by one or more groups, for example 1 , 2, 3, 4 or 5 groups independently selected from the group consisting of halogen, hydroxy, nitro, cyano, (C C6)alkyl-(Rc)0-3, (Ci-C6)-alkoxy-(Rc)o-3, (C3-Ci2)-cycloalkyl-(Rc)o-3, 0-(C3-Ci2)-cycloalkyl- (Rc)o-3, halo(CrC6)alkyl, halo(CrC6)alkoxy, (C6-Ci4)-aryl-(Rc)o-3, 0-(C6-Ci4)aryl-(Rc)o-3, (C6-C14)ar(Ci-C6)alkyl-(Rc)0-3! heterocyclyl-(Rc)o-3, heteroaryl-(Rc)0-3, C(0)Ra, C(0)ORa, S(0)nRa, P(0)n(0(CrC6)alkyl)2, O-P(O) (0(CrC6)alkyl)2, NRaRb and C(0)NRaRb; wherein Ra, Rb, Rc and n are as defined herein above. In monosubstituted phenyl, the substituent can be located in the 2-position, the 3-position or the 4-position. If the phenyl carries two substituents, they can be located in 2, 3-position, 2, 4-position, 2, 5- position, 2, 6-position, 3, 4-position or 3, 5-position. Examples of monosubstituted phenyl groups include, but are not limited to, 3-trifluoromethylphenyl, 4-chlorophenyl and 4-cyanophenyl. Examples of disubstituted phenyl groups include, but are not limited to, 3, 5-difluorophenyl, and 3, 4-dimethoxyphenyl.
As used herein, the term "(C6-Ci4)-aryloxy" or "aryloxy" or "0-(C6-Ci4)aryl" refers to an "(C6-Ci4)-aryr group having an oxygen radical attached thereto. The aryl of aryloxy group can be unsubstituted or substituted as explained in the definition of substituted (C6-Ci4)-aryl herein above. Examples of aryloxy groups include, but not limited to, phenoxy, 4-chlorophenoxy, and 3, 4-dimethoxyphenoxy. As used herein, the term "aralkyl" or "(C6-Ci4)ar(C-|-C6)alkyr refers to (CrC6)alkyl group substituted with an (C6-Ci4)-aryl group, wherein the terms alkyl and aryl are as defined above. Exemplary aralkyl groups include (CH2)p-phenyl, wherein p is an integer from 1 to 6, such as benzyl wherein p is 1 . The aryl of the (C6-Ci4)-aralkyl group can be unsubstituted or substituted as explained in the definition of substituted aryl herein above.
As used herein, the term "aralkyloxy" or "(C6-Ci4)ar(C-|-C6)alkyloxy" refers to an aralkyl group having an oxygen radical attached thereto. The aryl of aralkyloxy group may be unsubstituted or substituted as explained in the definition of substituted aryl herein above.
The term "heteroatom" as used herein, includes nitrogen (N), oxygen (O) and sulfur (S). Any heteroatom with unsatisfied valency is assumed to have a hydrogen atom to satisfy the valency.
As used herein, the terms "heterocyclyl" or "heterocyclic" whether used alone or as part of a substituent group, unless indicated otherwise, refers to a saturated, partially unsaturated, monocyclic or polycyclic ring system containing 1 to 1 0 carbon atoms and 1 to 4 identical or different heteroatoms selected from the group consisting of oxygen, nitrogen and sulfur. Unless indicated otherwise, the term "heterocyclyl" primarily refers to a 3- to 12- membered ring system, preferably, 3- to 1 0- membered ring system which can be a monocyclic or a bicyclic ring. Representative examples of heterocyclyls include, but are not limited to, pyrrolyl, pyrrolidinyl, pyrazolyl, imidazolyl, pyrazinyl, piperazinyl, oxazolyl, oxadiazolyl, isoxazolyl, triaziolyl, thiazolyl, tetrazolyl, furyl, thienyl, purinyl, pyridinyl, pyridazinyl, pyrimidinyl, piperidyl, benzoxazolyl, benzothiazolyl, benzofuranyl, purinyl, benzirmidazolyl, benzoxazolyl, indolyl, indazolyl, isoindolyl, isothiazolyl, isoquinolyl, isoquinolyl, morpholinyl, thiomorpholinyl, thiomorpholinyl-1 , 1 - dioxide, quinoxalinyl, quinolinyl and thiophenyl. The oxidized form of the ring nitrogen and sulfur atom of the heterocyclyl to provide N-oxide, S-oxide or S, S-dioxide is also encompassed. Heterocyclyl having an aromatic ring containing heteroatom/s are herein referred to by the customary term "heteroaryl". Unless indicated otherwise, within the context of the present invention and as used herein, the term "heteroaryl" refers to a 5- to 10- membered aromatic monocyclic or bicyclic ring system containing 1 to 4 identical or different hetero atoms selected from the group consisting of nitrogen, sulphur and oxygen. Examples of heteroaryls include, but are not limited to, pyrrole, pyrazole, imidazole, pyrazine, furan, thiophene, oxazole, thiazole, benzimidazole, benzoxazole, benzothiazole, benzofuran, indole, indazole, isoindole, isoquinoline, isooxazole, triazine, purine, pyridine, quinoline, oxadiazole, thiene, pyridazine, pyrimidine, isothiazole, quinoxaline (benzopyrine) and tetrazole. The oxidized form of the ring nitrogen and sulfur atom of the heteroaryl to provide N-oxide, S-oxide or S,S-dioxide is also encompassed. Unless stated otherwise, heterocyclyl or heteroaryl group can be unsubstituted or substituted. A substituted heterocyclyl or heteroaryl refers to a heterocyclyl or heteroaryl substituted with 1 -5 groups independently selected from the group consisting of halogen, hydroxy, nitro, cyano, (Ci-C6)alkyl-(Rc)0-3, (CrC6)-alkoxy-(Rc)o-3, (C3-Ci2)-cycloalkyl-(Rc)o-3, O-(C3-Ci2)-cycloalkyl-(Rc)0-3, halo(CrC6)alkyl, halo(C C6)alkoxy, (C6-Ci4)-aryl-(Rc)0-3, O-(C6-Ci4)aryl-(Rc)0-3, (C6-Ci4)ar(Ci-C6)alkyl-(Rc)o^, heterocyclyl-(Rc)o-3, heteroaryl-(Rc)0-3, C(0)Ra, C(0)ORa, S(0)nRa, P(0)n(0(C C6)alkyl)2, O-P(O) (0(CrC6)alkyl)2, NRaRb and C(0)NRaRb; wherein Ra, Rb, Rc and n are as defined herein above. The substituents can be present on either the ring carbon or the ring nitrogen atom(s). The substituents can be present at one or more positions provided that a stable molecule results.
Within the context of this present application and as used herein the term "isotopic forms" or "isotopically labeled forms" is a general term used for isotopic forms of compounds of formula I, wherein one or more atoms of compounds of formula I are replaced by their respective isotopes. All isotopes of any particular atom or element as specified are contemplated within the scope of the compounds of the invention. Examples of isotopes that can be incorporated into the compounds disclosed herein include, but are not limited to, isotopes of hydrogen such as 2H (deuterium or D) and 3H, carbon such as 1 1C, 13C and 14C, nitrogen such as 13N and 15N, oxygen such as 150, 170 and 180, chlorine such as 36CI, fluorine such as 18F and sulphur such as 35S. Substitution with heavier isotopes, for example, replacing one or more key carbon- hydrogen bonds with carbon-deuterium bond may show certain therapeutic advantages, resulting from longer metabolism cycles, (e.g., increased in vivo half life or reduced dosage requirements), improved safety or greater effectiveness and hence may be preferred in certain circumstances.
Representative examples of isotopic forms of the compounds of formula I can include, without limitation, deuterated compounds of formula I. The term "deuterated" as used herein, by itself or used to modify a compound or group, refers to replacement of one or more hydrogen atom(s), which is attached to carbon(s), with a deuterium atom. For example, the compounds of formula I can include in the definitions of one or more of its various variables, wherever applicable, deuterium, deuterated-alkyl, deuterated-alkoxy, deuterated-cycloalkyl, deuterated-heterocyclyl, deuterated-aryl, deuterated-heteroaryl and the like. The term "deuterated-alkyl" refers to an (d-C6)-alkyl group as defined herein, wherein at least one hydrogen atom bound to carbon is replaced by a deuterium. That is, in a deuterated alkyl group, at least one carbon atom is bound to a deuterium. In a deuterated alkyl group, it is possible for a carbon atom to be bound to more than one deuterium; it is also possible that more than one carbon atom in the alkyl group is bound to a deuterium. Analogously, the term "deuterated" and the terms deuterated-heterocyclyl, deuterated-heteroaryl, deuterated-cycloalkyl, deuterated-aryl, deuterated-alkoxy each refer to the corresponding chemical moiety wherein at least one carbon is bound to a deuterium.
The term "pharmaceutically acceptable solvate(s)" or "solvate(s)" as used herein refers to a compound formed by the interaction of a solute (in the present invention, a compound of formula I or a pharmaceutically acceptable salt thereof) and a solvent. Such solvents for the purpose of the invention may not interfere with the biological activity of the solute. Preferably, the solvent used is a pharmaceutically acceptable solvent. Examples of suitable pharmaceutically acceptable solvents include, without limitation, water, ethanol and acetic acid. Most preferably, the solvent used is water and the solvates obtained are referred to as hydrates. Examples for suitable solvates are the mono- or di-hydrates or alcoholates of the compounds according to the invention. Within the context of the present invention and as used herein, the term "stereoisomer" or "stereoisomeric form" is a general term used for all isomers of individual compounds (in the present invention, a compound of formula I) that differ only in the orientation of their atoms in space. The term stereoisomer includes mirror image isomers (enantiomers), mixtures of mirror image isomers (racemates, racemic mixtures), geometric (cis/trans or E/Z) isomers, and isomers of compounds with more than one chiral center that are not mirror images of one another (diastereoisomers).
The term "tautomer" or "tautomeric form" refers to the coexistence of two (or more) compounds that differ from each other only in the position of one (or more) mobile atoms and in electron distribution, for example, keto-enol tautomers.
As used herein, the term "pharmaceutically acceptable" means that the carrier, diluent, excipients, and/or salt must be compatible with the other ingredients of the formulation, and not deleterious to the recipient thereof. The term "pharmaceutically acceptable salt(s)" as used herein includes salts of the active compound i.e. the compound of formula I, which retain the desired biological activity of the subject compound and exhibit minimal undesired toxicological effects; and are prepared with suitable acids or bases, depending on the particular substituents found on the compounds described herein.
Within the context of the present invention and as used herein the term "polymorph(s)" or "polymorphic form" refers to crystals of the same compound that differs only in the arrangement and/or conformation of the molecule in the crystal lattice. Within the context of the present invention and as used herein, "N-oxide" refers to the oxide of the nitrogen atom of a nitrogen-containing heteroaryl or heterocycle. N-oxide can be formed in the presence of an oxidizing agent such as m-chloro-perbenzoic acid or hydrogen peroxide. N-oxide refers to an amine oxide, also known as amine-N-oxide, and is a chemical compound that contains N->0 bond.
Within the context of the present invention and as used herein, "a prodrug" or "prodrugs" refers to any compound, which are derivatives of parent compound (in the context of the present invention, a compound of formula I), which following administration, release(s) the parent compound in vivo via a chemical or physiological process, e.g., a prodrug on being brought to the physiological pH or through enzyme action is converted to the parent compound. In the context of the present invention, the term "compound(s) of formula I" or "compounds of the present invention" are used interchangeably and includes all the stereoisomeric and tautomeric forms and mixtures thereof in all ratios, and pharmaceutically acceptable salts, pharmaceutically acceptable solvates, pharmaceutically acceptable prodrugs, N-oxides and pharmaceutically acceptable polymorphs thereof. The compound(s) of formula I can also be referred to herein as "the active compound" or "the active ingredient". The term, "therapeutically effective amount" as used herein means an amount of a compound of formula I or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof; or a composition comprising the compound of formula I or a pharmaceutically acceptable salt thereof, effective in producing the desired therapeutic response in a particular patient suffering from a disease or disorder mediated by IL1 7 or TNF-a. Particularly, the term "therapeutically effective amount" includes the amount of the compound, when administered, that induces a positive modification in the disease or disorder to be treated or is sufficient to prevent development of, or alleviate to some extent, one or more of the symptoms of the disease or disorder being treated in a subject. In respect of the therapeutic amount of the compound, consideration is also given that the amount of the compound used for the treatment of a subject is low enough to avoid undue or severe side effects, within the scope of sound medical judgment. The therapeutically effective amount of the compound or composition will vary with the particular condition being treated, the age and physical condition of the end user, the severity of the condition being treated or prevented, the duration of the treatment, the nature of concurrent therapy, the specific compound or composition employed the particular pharmaceutically acceptable carrier utilized.
As used herein, the term "pharmaceutically acceptable carrier" refers to a material that is non-toxic, inert, solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type which is suitable for a subject, preferably a mammal, more preferably a human, and is suitable for delivering an active agent to the target site without terminating the activity of the agent.
The term "disease(s) or disorder(s) mediated by IL-17 or TNF-a" encompasses all diseases, disorders and medical conditions in which pro-inflammatory cytokines IL-1 7 or TNF-α plays a role, whether directly or indirectly, including the causation, development, progression, persistence or pathology of the disease or disorder.
The term "disease(s) or disorder(s) mediated by IL-17" encompasses all diseases, disorders and medical conditions in which pro-inflammatory cytokine IL-17 plays a role, whether directly or indirectly, including the causation, development, progression, persistence or pathology of the disease or disorder.
The term "disease(s) or disorder(s) mediated by TNF-a" encompasses all diseases, disorders and medical conditions in which pro-inflammatory cytokine TNF-a plays a role, whether directly or indirectly, including the causation, development, progression, persistence or pathology of the disease or disorder.
The diseases or disorders mediated by IL-17 or TNF-a can be selected from the group consisting of an autoimmune or inflammatory disease or disorder; and a metabolic disease or disorder.
The term "subject" as used herein refers to an animal, preferably a mammal, and most preferably a human. The term "mammal" used herein refers to warm-blooded vertebrate animals of the class 'mammalia', including humans, characterized by a covering of hair on the skin and, in the female, milk-producing mammary glands for nourishing the young. The term mammal includes animals such as cat, dog, rabbit, bear, fox, wolf, monkey, deer, mouse, pig and human. The term "subject" may be used interchangeably with the term patient. In the context of the present invention the phrase "a subject in need thereof" means a subject in need of the treatment for the disease or disorder that is mediated by IL1 7 or TNF-a. Alternatively, the phrase "a subject in need thereof means a subject (patient) diagnosed having a disease or disorder that is mediated by IL17 or TNF-a. As used herein, the terms "treatment" "treat" and "therapy" and the like refer to alleviate, slow the progression, attenuation or cure of existing diseases or condition (e.g. rheumatoid arthritis). Treatment also includes treating, preventing development of, or alleviating to some extent, one or more of the symptoms of the diseases or condition. Embodiments
The invention encompasses all the compounds described by the formula I without limitation, however, for the purpose of further illustrations, preferred aspects and elements of the invention are discussed herein in the form of the following embodiments.
In an embodiment, the present invention relates to a compound of formula I,
wherein,
A is triazole, pyrazole, imidazole, oxazole, isoxazole, thiazole, pyrrole or a heteroaryl ring system selected from:
Figure imgf000021_0001
wherein * indicates the point of attachment to L; and
L, R1 5 R2, R3 and n are as defined in the first aspect with the provisos that:
(a) when A is pyrazole then L is a group other than *C(0)N(R5) and *N(R4)-C(0); and
(b) when A is pyrazole and L is *N(R4)-C(0)-N(R5), then said L group is not attached to 5-position of pyrazole;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
In another embodiment, the present invention relates to a compound of formula I, wherein,
A is triazole, pyrazole, imidazole, oxazole, isoxazole, thiazole or pyrrole;
with the provisos that: (a) when A is pyrazole then L is a group other than *C(0)N(R5) and *N(R4)-C(0); and
(b) when A is pyrazole and L is *N(R4)-C(0)-N(R5), then said L group is not attached to 5-position of pyrazole;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
In another embodiment, the present invention relates to a compound of formula I;
wherein,
A is triazole, pyrazole, isoxazole or thiazole;
L is *N(R4)-C(0)-N(R5), *C(0)N(R5), *N(R4)-C(0), *CH(R6)-C(0)-N(R5), *CH(R6)N(R5), *N(R4)-S(0)2 or *NR4; wherein * indicates the point of attachment to A;
R , R5 and R6, are as defined in the first aspect with the provisos that:
(c) when A is pyrazole then L is a group other than *C(0)N(R5) and *N(R4)-C(0); and
(d) when A is pyrazole and L is *N(R4)-C(0)-N(R5), then said L group is not attached to 5-position of pyrazole;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
In another embodiment, the present invention relates to a compound of formula I;
wherein,
A is triazole;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
In another embodiment, the present invention relates to a compound of formula I; wherein:
A is pyrazole;
L is *N(R4)-C(0)-N(R5); *CH(R6)-C(0)-N(R5); *CH(R6)N(R5); *N(R4)-S(0)2 or *NR4; wherein * indicates the point of attachment to A;
R4, R5 and R6, are as defined in the first aspect with the proviso that when L is *N(R4)- C(0)-N(R5), then the said L group is not attached to 5-position of pyrazole;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof. In another embodiment, the present invention relates to a compound of formula I;
wherein A is isoxazole;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
In another embodiment, the present invention relates to a compound of formula I;
wherein A is thiazole;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
In another embodiment, the present invention provides a compound of formula I ; wherein A is
Figure imgf000023_0001
wherein * indicates the point of attachment to L;
and L, R1 ; R2, R3 and n are as defined in the first aspect;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof. In another embodiment, the present invention provides a compound of formula I ; wherein A is
Figure imgf000023_0002
wherein * indicates the point of attachment to L; and L, R1 ; R2, R3 and n are as defined in the first aspect;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof. In another embodiment, the present invention relates to a compound of formula I; wherein A is
Figure imgf000024_0001
wherein * indicates the point of attachment to L;
and L, R1 ; R2, R3 and n are as defined in the first aspect;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
In another embodiment, the present invention relates to a compound of formula I; wherein each of Xi , X2, X4 and X5 is C(R7); and X3 is C(R8); and
R7 and R8 are as defined in the first aspect; with the provisos that:
(a) when A is pyrazole then L is a group other than *C(0)N(R5) or *N(R4)-C(0); and
(b) when A is pyrazole and L is *N(R4)-C(0)-N(R5), then said L group is not attached to 5-position of pyrazole;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
In another embodiment, the present invention relates to a compound of formula I; wherein A is selected from a triazole, pyrazole, isoxazole or thiazole;
each of Xi , X2, X4 and X5 is C(R7); and X3 is C(R8); and
R7 and R8 are as defined in the first aspect; with the provisos that:
(a) when A is pyrazole then L is a group other than *C(0)N(R5) or *N(R4)-C(0); and
(b) when A is pyrazole and L is *N(R4)-C(0)-N(R5), then said L group is not attached to 5-position of pyrazole;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof. In another embodiment, the present invention relates to a compound of formula I; wherein A is triazole or pyrazole;
L is *N(R4)-C(0)-N(R5); wherein * indicates the point of attachment to A;
each of Xi , X2, X4 and X5 is C(R7); and X3 is C(R8); and
R4, R5, R7 and R8 are as defined in the first aspect; with the proviso that when A is pyrazole the said L group is not attached to 5-position of pyrazole;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof. In another embodiment, the present invention relates to a compound of formula I; wherein A is triazole;
L is *N(R4)-C(0)-N(R5); *C(0)N(R5), *CH(R6)-C(0)-N(R5); *CH(R6)N(R5) or *N(R4)- S(0)2 ; wherein * indicates the point of attachment to A;
each of Xi , X2, X4 and X5 is C(R7); and X3 is C(R8); and
R , R5, R6, R7 and R8 are as defined in the first aspect;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
In another embodiment, the present invention relates to a compound of formula I; wherein A pyrazole;
L is *N(R4)-C(0)-N(R5); N(R4)-S(0)2 or *CH(R6)N(R5); wherein * indicates the point of attachment to A;
each of Xi , X2, X and X5 is C(R7); and X3 is C(R8); and
R4, R5, R7 and R8 are as defined in the first aspect; with the proviso that when A is pyrazole and L is *N(R4)-C(0)-N(R5), then the said L group is not attached to 5-position of pyrazole;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof. In another embodiment, the present invention relates to a compound of formula I; wherein A is
Figure imgf000026_0001
wherein * indicates the point of attachment to L;
L is *N(R4)-C(0)-N (R5); *C(0)N(R5); *N(R4)-S(0)2 or *N R4; wherein * indicates the point of attachment to A;
each of Xi , X2, X4 and X5 is C(R7); and X3 is C(R8) ; and
Ri , R2, R3, R4, R5, R7, Re and n are as defined in the first aspect;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof. In another embodiment, the present invention relates to a compound of formula I; wherein A is
Figure imgf000026_0002
wherein * indicates the point of attachment to L;
L is *N(R4)-C(0)-N(R5) ; *C(0)N(R5); N(R4)-S(0)2 or *NR4; wherein * indicates the point of attachment to A;
each of Xi , X2, X4 and X5 is C(R7); and X3 is C(R8) ; and
Ri , R2, R3, R4, R5, R7, Re and n are as defined in the first aspect;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
In another embodiment, the present invention relates to a compound of formula I; wherein one of Xi , X2, X4 or X5 is N, while the remaining are C(R7); X3 is N or C(Rs); or any two of Xi , X2, X4 or X5 are N, while the remaining are C(R7); X3 is N or C(Rs) ; and R7 and R8 are as defined in the first aspect; with the provisos that:
(a) when A is pyrazole then L is a group other than *C(0)N(R5) or *N(R4)-C(0); and (b) when A is pyrazole and L is *N(R4)-C(0)-N(R5), then said L group is not attached to 5-position of pyrazole; or an isotopic form, stereoisomer or a tautomer or a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
Representative compounds of formula I encompassed in accordance with the present invention include:
1 -(5-Methyl-1 -(4-(trifluoromethyl)phenyl)-1 H-pyrazol-4-yl)-3-(2-(trifluoromethyl)-1 H- benzo[d]imidazol-6-yl)urea;
1 -(4-(4-(4-Chlorophenyl)-4-hydroxypiperidin-1 -yl)-3-cyanophenyl)-3-(5-methyl-1 -(4- (trifluoromethyl)phenyl)-1 H-pyrazol-4-yl)urea;
1 -(3-Cyano-4-(4-(2-hydroxyethyl)piperidin-1 -yl)phenyl)-3-(5-methyl-1 -(4-
(trifluoromethyl)phenyl)-1 H-pyrazol-4-yl)urea;
1 -(5-Methyl-1 -phenyl-1 H-pyrazol-4-yl)-3-(naphthalen-1 -yl)urea;
1 -(1 -(4-Chlorophenyl)-5-methyl-1 H-pyrazol-4-yl)-3-(3-cyano-4-(4-morpholino piperidin-
1 -yl) phenyl) urea;
1 -(3-Cyano-4-(4-morpholinopiperidin-1 -yl) phenyl)-3-(5-methyl-1 -(4-(trifluoromethyl) phenyl)-1 H-pyrazol-4-yl) urea;
1 -(3-Cyano-4-(piperazin-1 -yl) phenyl)-3-(5-methyl-1 -(4-(trifluoromethyl) phenyl)-1 H- pyrazol-4-yl) urea;
1 -(5-Chloro-6-morpholinopyridin-3-yl)-3-(5-methyl-1 -(4-(trifluoromethyl) phenyl)-1 H- pyrazol-4-yl) urea;
1 - (3-Cyano-4-(4-(4-fluoro-3-(trifluoromethyl) benzoyl) piperazin-1 -yl) phenyl)-3-(5- methyl-1 -(4-(trifluoromethyl)phenyl)-1 H-pyrazol-4-yl)urea;
1 -(3-Cyano-4-(4-morpholinopiperidin-1 -yl) phenyl)-1 -methyl-3-(5-methyl-1 -(4- (trifluoromethyl) phenyl)-1 H-pyrazol-4-yl) urea;
1 -(1 -(4-Chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(4-morpholinopiperidin -1 - yl)phenyl)urea;
1 -(4-([1 ,4'-Bipiperidin]-1 '-yl)-3-cyanophenyl)-3-(1 -(4-chlorophenyl)-1 H-1 ,2,3-triazol-4- yl)urea;
2- (1 -(4-(3-(1 -(4-(Tert-butyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)ureido)-2-cyanophenyl) piperidin-4-yl)ethyl diethyl phosphate;
1 -(1 -(4-(Tert-butyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(4-(2-methoxyethyl) piperidin-1 -yl)phenyl)urea;
1 -(1 -(4-(Tert-butyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(4-(4-morpholinopiperidin-1 -yl)-2- (trifluoromethyl)phenyl)urea; 1 -(3-Cyano-4-(4-morpholinopiperidin-1 -yl)phenyl)-3-(1 -(4-morpholinophenyl)-1 H-1 ,2,3- triazol-4-yl)urea;
1 -(4-(1 ,1 ,1 ,3,3,3-Hexafluoro-2-hydroxypropan-2-yl)phenyl)-3-(1 -(4-isopropylphenyl) - 1 H-1 ,2,3-triazol-4-yl)urea;
1 -(1 -(4-Chlorophenyl)-5-methyl-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(4-morpholino piperidin-1 -yl)phenyl)urea;
3- (3-(1 -(4-Chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)ureido)-N-(4-hydroxy-3-methoxy benzyl)benzamide;
4- (1 -(2-Cyano-4-(3-(1 -(4-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)ureido) phenyl)piperidin-4-yl)butanoic acid;
1 -(3-Cyano-4-(4-(pyrimidin-2-yl)piperazin-1 -yl)phenyl)-3-(1 -(4-(trifluoromethyl) phenyl)- 1 H-1 ,2,3-triazol-4-yl)urea;
1 -(1 -(4-Chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(piperazin-1 -yl)phenyl) urea; 1 -(1 -(4-Chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(5-(4-morpholinopiperidin-1 -yl) pyridin-2- yl)urea;
1 -(1 -(4-Chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(4-cyano-5-(4-morpholinopiperidin -1 - yl)pyridin-2-yl)urea;
1 -(6-(4-Morpholinopiperidin-1 -yl)pyridin-3-yl)-3-(1 -(3-(trifluoromethyl)phenyl)-1 H-1 ,2,3- triazol-4-yl)urea;
1 -(5-(3-(1 -(4-(Tert-butyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)ureido)pyridin-2-yl)piperidine-4- carboxylic acid;
1 -(6-(4-(Pyrimidin-2-yl)piperazin-1 -yl)-5-(trifluoromethyl)pyridin-3-yl)-3-(1 -(3- (trifluoromethyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)urea;
1 -(5-Chloro-6-(4-morpholinopiperidin-1 -yl)pyridin-3-yl)-3-(1 -(4-chlorophenyl)-1 H-1 ,2,3- triazol-4-yl)urea;
1 -(1 -(4-(Tert-butyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(5-methyl-6-(4-morpriolino piperidin- 1 -yl)pyridin-3-yl)urea;
1 -(1 -(4-(Tert-butyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(5-methyl-6-(4-(pyrimidin-2- yl)piperazin-1 -yl)pyridin-3-yl)urea;
1 -(1 -(4-(Tert-butyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(5-cyano-6-(4-(pyrimidin-2- yl)piperazin-1 -yl)pyridin-3-yl)urea;
1 -(6-(4-Cyano-4-phenylpiperidin-1 -yl)pyridin-3-yl)-3-(1 -(3-(trifluoromethyl) phenyl)-1 H- 1 ,2,3-triazol-4-yl)urea; 1 -(3-Cyano-4-(piperidin-1 -yl)phenyl)-3-(1 -(4-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazol-4- yl)urea;
1 -(3-Cyano-4-(2-oxa-6-azaspiro[3.3]heptan-6-yl)phenyl)-3-(1 -(4-(trifluoro
methyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)urea;
1 -(1 -(4-Chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(4-(cyclopropylmethyl) piperazin-1 -yl)phenyl)urea;
1 -(1 -(4-Chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(4-met oxypiperidin-1 - yl)phenyl)urea;
1 -(3-Cyano-4-(4-(3-hydroxyphenyl)piperazin-1 -yl)phenyl)-3-(1 -(p-tolyl)-l H-1 ,2,3-triazol- 4-yl)urea;
1 -(3-Cyano-4-(4-morpholinopiperidin-1 -yl)phenyl)-3-(1 -(3-(trifluoromethyl) phenyl)-1 H- 1 ,2,3-triazol-4-yl)urea;
1 -(1 -(4-Chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(4-hydroxypiperidin-1 - yl)phenyl)-1 ,3-dimethylurea;
1 -(4-(3-(1 -(4-(Tert-butyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)ureido)-2-cyanophenyl)-4-(2- hydroxypropan-2-yl)piperidin-1 -ium met anesulfonate;
1 -(3-Cyano-4-(piperazin-1 -yl)phenyl)-3-(1 -(4-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazol-4- yl)urea hydrochloride;
1 -(3-Cyano-4-(4-morpholinopiperidin-1 -yl)phenyl)-3-(3-(4-fluorophenyl)-5- methylisoxazol-4-yl)urea;
1 -(4-(Tert-butyl) phenyl)-N-(3-cyano-4-(4-(2-hydroxyethyl) piperidin-1 -yl) phenyl)-1 H-1 , 2, 3-triazole-4-carboxamide;
1 -(4-(Tert-butyl) phenyl)-N-(3-cyano-4-(4-hydroxypiperidin-1 -yl) phenyl)-1 H-1 , 2, 3- triazole-4-carboxamide;
1 -(4-Chlorophenyl)-N-(3-cyano-4-(4-morpholinopiperidin-1 -yl)phenyl)-1 H-1 ,2,3-triazole- 4-carboxamide;
1 -(4-Chlorophenyl)-N-(3-cyano-4-(4-morpholinopiperidin-1 -yl)phenyl)-1 H-1 ,2,3-triazole-
4- carboxamide hydrochloride;
1 -(4-Chlorophenyl)-N-(3-cyano-4-(4-(pyrimidin-2-yl)piperazin-1 -yl)phenyl)-1 H-1 ,2,3- triazole-4-carboxamide;
N-(3-Cyano-4-(4-morpholinopiperidin-1 -yl) phenyl)-5-pentyl-1 -(3-(trifluoromethyl) phenyl)-1 H-1 ,2,3-triazole-4-carboxamide;
N-(3-Cyano-4-(4-morpholinopiperidin-1 -yl)phenyl)-3-(4-fluorophenyl)-4-rnethylisoxazole-
5- carboxamide; 3- (4-Chlorophenyl)-N-(3-cyano-4-(4-morpholinopiperidin-1 -yl)phenyl)isoxazole-5- carboxamide;
N-(1 -(4-Chlorophenyl)-5-methyl-1 H-pyrazol-4-yl)-3-cyano-4-(4-morpholino piperidin-1 - yl)benzenesulfonamide;
N-(4-(3-Methoxyphenyl)pyrimidin-2-yl)-5^henylthiazol-2-arnine;
5-Phenyl-N-(4-(4-(trifluoromethoxy)phenyl)pyrimidin-2-yl)thiazol-2-arnine;
N-(4-(2-Methoxyphenyl)pyrimidin-2-yl)-5-phenylthiazol-2-arnine;
5-((6-Fluorobenzo[d]thiazol-2-yl) amino)-2-(4-morpholinopiperidin-1 -yl) benzonitrile; 5-((4-(3, 5-Dimethoxyphenyl) pyrimidin-2-yl) amino)-2-(4-morpholinopiperidin-1 -yl) benzonitrile;
5-((4-(3, 5-Dimethoxyphenyl) pyrimidin-2-yl) amino)-2-(4-(hydroxymethyl) piperidin-1 -yl) benzonitrile;
4- (2-Methoxyphenyl)-N-(4-(4-(pyrimidin-2-yl)piperazin-1 -yl)-3-(trifluoromethyl) phenyl) pyrimidin-2-amine;
1 ,1 ,1 ,3,3,3-Hexafluoro-2-(4-((4-(3-methoxyphenyl)pyrimidin-2-yl)amino)phenyl) propan- 2-ol;
2-(4-Hydroxypiperidin-1 -yl)-5-((4-(3-methoxyphenyl)pyrimidin-2-yl)amino) benzonitrile;
5- ((4-Morpholinopyrimidin-2-yl)amino)-2-(4-(pyridin-2-yl)piperazin-1 -yl)benzonitrile 2-morpholino-5-((4-morpholinopyrimidin-2-yl)amino)benzonitrile;
5-((4-((2-Morpholinoethyl)amino)pyrimidin-2-yl)amino)-2-(4-morpholino piperidin-1 - yl)benzonitrile;
5-((2-Chloro-6-methoxyquinolin-4-yl) amino)-2-(4-morpholinopiperidin-1 -yl) benzonitrile;
2-(4-Morpholinopiperidin-1 -yl)-5-(quinolin-2-ylamino) benzonitrile;
5-((Cyano(1 -(3-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)methyl)amino)-2-(4-(2- hydroxypropan-2-yl)piperidin-1 -yl)benzonitrile;
5-((Cyano(1 -(3-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)methyl)amino)-2-(4- (pyrimidin-2-yl)piperazin-1 -yl)benzonitrile;
2-Chloro-5-(((1 -(4-chlorophenyl)-5-methyl-1 H-pyrazol-4-yl)methyl)amino) benzonitrile; 5-(((1 -(4-Chlorophenyl)-5-methyl-1 H-pyrazol-4-yl)methyl)amino)-2-(4-morpholino piperidin-1 -yl)benzonitrile;
Diethyl (((4-chloro-3-cyanophenyl)amino)(1 -(4-chlorophenyl)-5-methyl-1 H-pyrazol-4- yl)methyl)phosphonate;
N-(4-(1 ,1 ,1 ,3,3,3-Hexafluoro-2-hydroxypropan-2-yl)phenyl)-2-hydroxy-2-(1 -(3- (trifluoromethyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)acetamide; or N-(3-Cyano-4-(4-methoxypiperidin-1 -yl)phenyl)-2-hydroxy-2-(1 -(3 (trifluoromethyl) phenyl)-1 H-1 ,2,3-triazol-4-yl)acetamide; or
a stereoisomer, a tautomer or a pharmaceutically acceptable salt thereof. The compounds of the present invention include all isotopic forms, stereoisomeric and tautomeric forms and mixtures thereof in all ratios and their pharmaceutically acceptable salts, pharmaceutically acceptable solvates, prodrugs, N-oxides, S-oxides and polymorphs. According to another aspect of the present invention, there are provided processes for the preparation of the compounds of formula I.
Thus, the compound of formula I can be prepared by various methods including using methods well known to the person skilled in the art. Examples of processes for the preparation of the compounds of formula I are described below and illustrated in the following scheme, but are not limited thereto. It will be appreciated by the person skilled in the art that within certain of the processes described herein, the order of the synthetic steps employed can be varied and will depend inter alia on factors such as the nature of functional groups present in a particular substrate and the protecting group strategy (if any) to be adopted. Clearly, such factors will also influence the choice of reagent such as bases, solvents, coupling agents to be used in the reaction steps.
The reagents, reactants and intermediates used in the following process are either commercially available or can be prepared according to standard procedures known in the art, for instance those reported in the literature references. In the following schemes and the description of the processes for the synthesis of the compounds of formula I, the starting compounds and the intermediates used for the synthesis of compounds of the present invention, are designated as intermediates 1 -13 respectively, for ease of reference. Throughout the process description, the corresponding substituent groups in the various formulae representing starting compounds and intermediates have the same meanings as that for the compound(s) of formula I as indicated in one or more embodiments described above, unless stated otherwise.
The processes for the preparation of the compounds of formula I of the present invention are depicted in schemes 1 to 7 presented below. For ease of reference, the reaction steps shown in schemes 1 to 7, are referred to by using general symbols namely (1 a), (1 b) and (1 c) in the scheme 1 ; (2a) in the scheme 2, and the like.
Scheme 1 depicts a process for the preparation of the compounds of formula I, wherein A, R-i , R2, F?3, X-i , X2, X3, X4, X5 and n are as defined in the first aspect of the present invention, and L is *N(R4)-C(0)-N(R5); wherein * indicates the point of attachment to A; and R4 and R5 are hydrogen.
Scheme 1
Figure imgf000032_0001
1
wherein, A, R-i , R2, R3 and n are as defined in the first aspect; is hydrolysed by reacting it with NaOH in a solvent such as a mixture of tetrahydrofuran (THF) and water at a temperature ranging from 70 °C to 90 °C to obtain a compound 2 of the following formula;
Figure imgf000033_0001
2
wherein, A, R-i , R2, R3 and n are as defined in the first aspect.
Step (1 b) Method A:
The compound of formula 2 as obtained in step (1 a) is reacted with reagents namely trichloroacetonitrile, triphenyl phosphine and sodium azide in a solvent such as acetone at a temperature ranging from 20 °C to 35 °C to obtain a compound 3 of the following formula;
Figure imgf000033_0002
3
wherein, A, R ; R2, R3 and n are as defined in the first aspect. Method B:
The compound of formula 2 as obtained in step (1 a) is reacted with reagents namely sodium azide and triphosgene in the presence of a base such as triethylamine (TEA) in a solvent such as acetone or THF at a temperature ranging from 20 °C to 35 °C to obtain a compound 3 of the above formula.
Step (1 c)
The compound of formula 3 as obtained in step (1 b) is reacted with a compound 4 of the following formula;
Figure imgf000034_0001
4
wherein, X1 ; X2, X3, X4 and X5 are as defined in the first aspect;
in a solvent such as DMF (N, N-Dimethylformamide) or toluene at a temperature ranging from 100 °C to 120 °C to obtain a compound of formula I; wherein R-i , R2, R3, Xi , X2, X3, X4, X5 and n are as defined in the first aspect, L is *N(R4)-C(0)-N(R5); wherein * indicates the point of attachment to A; and R4 and R5 are hydrogen.
Scheme 2 depicts a process for the preparation of the compounds of formula I, wherein A, R-i , R2, R3, X-i , X2, X3, X4, X5 and n are as defined in the first aspect of the present invention, and L is *C(0)N(R5); wherein * indicates the point of attachment to A; and R5 is hydrogen.
Scheme 2
Figure imgf000034_0002
Step 2(a)
The compound 2 of the following formula;
Figure imgf000034_0003
2
wherein, A, R-i , R2, R3 and n are as defined in the first aspect;
is reacted with the compound 4 of the following formula;
Figure imgf000035_0001
4
wherein, X1 ; X2, X3, X4 and X5 are as defined in the first aspect;
in the presence of 1 -ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC) and hydroxybenzotriazole (HOBt) as a catalysts, in the presence of a base such as tnethylamine, in a solvent such as DMF (N, N-Dimethylformamide) at a temperature ranging from 20 °C to 35 °C to obtain a compound of formula I, wherein A, Ri , R2, R3, Xi , X2, X3, X4, X5 and n are as defined in the first aspect of the present invention, and L is *C(0)N(R5); wherein * indicates the point of attachment to A; and R5 is hydrogen.
Scheme 3 depicts a process for the preparation of the compounds of formula I, wherein A, R-i , R2, R3, X-i , X2, X3, X4, X5 and n are as defined in the first aspect of the present invention, and L is *N(R4)-S(0)2; wherein * indicates the point of attachment to A; and R is hydrogen.
Scheme 3
Figure imgf000035_0002
Step (3a)
Compound 4 of the following formula;
Figure imgf000035_0003
4
wherein, X1 ; X2, X3, X4 and X5 are as defined in the first aspect;
is reacted with cuprous chloride and sulphur dioxide in the presence of a base such as sodium nitrite in a solvent such as mixture of concentrated HCI and glacial acetic acid or acetic acid at a temperature ranging from -5 °C to 5 °C to obtain a compound 5 of the following formula;
Figure imgf000036_0001
5
wherein, X1 ; X2, X3, X4 and X5 are as defined in the first aspect; Step 3(b)
The compound of formula 5 as obtained in step 3(a) is reacted with a compound 6 of the following formula;
Figure imgf000036_0002
6
wherein, A, R ; R2, R3 and n are as defined in the first aspect;
in the presence of a base such as triethylamine (TEA) in a solvent such dichloromethane (MDC) at a temperature ranging from 20 °C to 35 °C to obtain a compound of formula I, wherein A, R ; R2, R3, Xi , X2, X3, X4, X5 and n are as defined in the first aspect of the present invention, and L is *N(R4)-S(0)2; wherein * indicates the point of attachment to A; and R4 is hydrogen.
Scheme 4 depicts a process for the preparation of the compounds of formula I, wherein A, R-i , R2, R3, Xi , X2, X3, X4, X5 and n are as defined in the first aspect of the present invention, and L is *N(R4)-C(0); wherein * indicates the point of attachment to A; and R4 is hydrogen.
Scheme 4
Figure imgf000036_0003
8
Step 4(a)
Compound 3 of following formula;
Figure imgf000037_0001
wherein, A, R-i , R2, R3 and n are as defined in the first aspect;
is refluxed in a solvent such as terf-butanol and reacted with saturated HCI in ethyl acetate at a temperature ranging from 20 °C to 35 °C to obtain a compound 7 of the following formula;
Figure imgf000037_0002
wherein, A, R-i , R2, R3 and n are as defined in the first aspect.
Step 4(b)
The compound of formula 7 as obtained in step 4(a) is reacted with compound 8 of the following formula;
Figure imgf000037_0003
8
wherein, X1 ; X2, X3, X4 and X5 are as defined in the first aspect;
in the presence of 1 -ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC) and hydroxybenzotriazole (HOBt) as a catalysts in a solvent such as dichloromethane at a temperature ranging from 20 °C to 35 °C to obtain a compound of formula I, wherein A, RT , R2, R3, X-i , X2, X3, X4, X5 and n are as defined in the first aspect of the present invention, and L is *N(R4)-C(O); wherein * indicates the point of attachment to A; and R4 is hydrogen.
Scheme 5 depicts a process for the preparation of the compounds of formula I wherein, A, Ri , R2, R3, X-i , X2, X3, X4, X5 and n are as defined in the first aspect of the present invention, and L is *NR4; wherein * indicates the point of attachment to A; and R4 is hydrogen.
Scheme 5
Figure imgf000038_0001
Formula I
Step 5(a)
Compound 7 of the following formula;
Figure imgf000038_0002
7
wherein, A, R-i, R2, R3 and n are as defined in the first aspect;
is reacted with a compound 9 of the following formula;
Figure imgf000038_0003
9
wherein, Xi, X2, X3, X4 and X5 are as defined in the first aspect;
in presence of a base such as xantphos and cesium carbonate and a catalyst namely tris(dibenzylidineacetone)dipd(0) in a solvent such as dioxane at a temperature ranging from 80 °C to 120 °C to obtain a compound of formula I, wherein A, Ri , R2, R3, X-i, X2, X3, X4, X5 and n are as defined in the first aspect of the present invention, and L is *NR4; wherein * indicates the point of attachment to A; and R4 is hydrogen.
Scheme 6 depicts a process for the preparation of the compounds of formula I wherein, A, R-i, R2, R3, X-i, X2, X3, X4, X5 and n are as defined in the first aspect of the present invention, and L is *CH(R6)N(R5); wherein * indicates the point of attachment to A; R5 is hydrogen and R6 is hydrogen or cyano.
Scheme 6
Figure imgf000039_0001
(wherein,
L is *CH(R6)N(R5); R5 is hydrogen; and R6 is cyano.)
Step 6(a)
Compound 1 of the following form
Figure imgf000039_0002
1
wherein, A, Ri , R2, R3 and n are as defined in the first aspect; is reacted with lithium aluminium hydride (LAH) in a solvent such as tetrahydrofuran at a temperature ranging from -5 °C to 5 °C to obtain a compound 10 of the following formula;
Figure imgf000040_0001
10
wherein, A, R ; R2, R3 and n are as defined in the first aspect.
Step 6(b)
The compound of formula 10 as obtained in step 6(a) is reacted with manganese dioxide in a solvent such as chloroform at a temperature ranging from 65 °C to 75 °C to obtain a compound 1 1 of the following formula;
Figure imgf000040_0002
wherein, A, R-i , R2, R3 and n are as defined in the first aspect. Step 6(c)
The compound of formula 1 1 as obtained in step 6(b) is reacted with compound 4 of the following formula;
Figure imgf000040_0003
wherein, Xi , X2, X3, X4 and X5 are as defined in the first aspect;
and trifluoroacetic acid in a solvent such as tetrahydrofuran at a temperature ranging from 25 °C to 30 °C to obtain a compound 1 2 of the following formula;
Figure imgf000041_0001
wherein, A, R-i , R2, R3,n, X-i , X2, X3, X4 and X5 are as defined in the first aspect;
which is telescoped to next step without isolation. Step 6(d)
The compound of formula 1 2 as obtained in step 6(c) is reduced by using a reducing agent such as sodium borohydride or sodium cyanoborohydride in a solvent such as tetrahydrofuran at a temperature ranging from 0 °C to 25 °C to obtain a compound of formula I wherein, A, R-i , R2, R3, X-i , X2, X3, X4, X5 and n are as defined in the first aspect of the present invention, and L is *CH(R6)N(R5); wherein * indicates the point of attachment to A; and R5 and R6 are hydrogen.
Step 6(e)
The compound of formula 1 2 as obtained in step 6(c) is reacted with trimethylsilanecarbonitrile and tetrabutylammonium difluorotriphenylsilicate in a solvent such as tetrahydrofuran at a temperature ranging from 0 °C to 25 °C to obtain a compound of formula I; wherein, A, Ri , R2, R3, Xi , X2, X3, X4, X5 and n are as defined in the first aspect of the present invention, and L is *CH(R6)N(R5); wherein * indicates the point of attachment to A; R5 is hydrogen and R6 is cyano.
Scheme 7 depicts a process for the preparation of the compounds of formula I wherein, A, R-i , R2, R3, X-i , X2, X3, X4, X5 and n are as defined in the first aspect of the present invention, and L is *CH(R6)-C(0)-N(R5); wherein * indicates the point of attachment to A; R5 is hydrogen and R6 is hydroxy.
Scheme 7
Figure imgf000041_0002
13 4 Formula I Step 7(a)
Compound 13 of the following formula;
Figure imgf000042_0001
13
wherein, A, R-i , R2, R3 and n are as defined in the first aspect;
is reacted with the compound 4 of the following formula;
Figure imgf000042_0002
4
wherein, Xi , X2, X3, X4 and X5 are as defined in the first aspect;
in the presence of 1 -ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC) and hydroxybenzotriazole (HOBt) as a catalysts, in presence of a base such as triethylamine, in a solvent such as DMF (N, N-Dimethylformamide) at a temperature ranging from 20 °C to 35 °C to obtain a compound of formula I, wherein A, R-i , R2, R3, Xi , X2, X3, X4, X5 and n are as defined in the first aspect of the present invention, and L is *CH(R6)-C(0)-N(R5); wherein * indicates the point of attachment to A; R5 is hydrogen and R6 is hydroxy.
The compound of formula I, as obtained in Schemes 1 , 2, 3, 4, 5, 6 and 7 can be optionally converted into its corresponding pharmaceutically acceptable salt. The term "pharmaceutically acceptable salts" as used herein refers to organic and inorganic salts of a compound of the invention, depending on the particular group (acidic or basic group) present in the compounds of formula I described herein. When compounds of the present invention contain relatively acidic groups, base addition salts can be obtained by contacting the compounds of formula I with a sufficient amount of an appropriate base, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable base addition salts include sodium, potassium, magnesium, calcium, ammonium, zinc or organic base salt. Examples of pharmaceutically acceptable organic base addition salts include those derived from organic bases such as lysine, arginine, choline, guanidine, diethanolamine, tromethamine, metformin and the like.
When compounds of the present invention contain relatively basic groups, acid addition salts can be obtained by contacting the compounds of formula I with a sufficient amount of an appropriate acid, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, mono- hydrogensulfuric or hydriodic acids and the like, as well as the salts derived from organic acids such as acetic, ascorbic, isobutyric, benzoic, citric, oxalic, succinic, suberic, fumaric, propionic, mandelic, phthalic, benzenesulfonic, p-tolylsulfonic, lactic, maleic, malonic, mandelic, tartaric, methanesulfonic, ethanesulfonic, glucuronic or galacturonic acids and the like.
The pharmaceutically acceptable salts of the compounds of formula I can be synthesized from the compound of formula I, which contains a basic or an acidic group, by using conventional chemical methods. Generally, the salts are prepared by treating the compound of formula I which may be a free base or an acid with a suitable salt- forming inorganic or an organic acid or a base in a suitable solvent or dispersant or from another salt by cation or anion exchange. Suitable solvents that can be used for the preparation of pharmaceutically acceptable salts include, but are not limited to, ethyl acetate, diethyl ether, methanol, ethanol, acetone, tetrahydrofuran, dioxane or mixtures of these solvents.
The compounds of formula I can be regenerated from their corresponding salts by contacting the salt with an appropriate base or acid depending on the type of salt and isolating the parent compound in the conventional manner. The compound differs from the various salt forms in certain physical properties. An example of physical properties that may differ is solubility in polar solvents.
The present invention also encompasses within its scope the solvates of the compounds of formula I. Certain compounds of the present invention can exist in unsolvated forms as well as solvated forms, including hydrated forms. Certain compounds of the present invention may exist in multiple crystalline or amorphous forms. In general, all physical forms are suitable for the uses contemplated by the present invention and are intended to be within the scope of the present invention.
Various polymorphs of compounds of formula I can be prepared by crystallization of the compounds under different conditions. The different conditions are, for example, using different solvents or their mixtures for crystallization; crystallization at different temperatures; various modes of cooling, ranging from very fast to very slow cooling during crystallizations. Polymorphs can also be obtained by heating or melting the compound followed by gradual or fast cooling. The presence of polymorphs can be determined by IR (infra-red) spectroscopy, solid probe NMR (nuclear magnetic resonance) spectroscopy, differential scanning calorimetry, powder x-ray diffraction or such other techniques.
Those skilled in the art will recognize that stereocentres exist in compounds of formula I. Accordingly, the present invention includes all possible stereoisomers and geometric isomers of formula I and includes not only racemic compounds but also the optically active isomers as well. When a compound of formula I is desired as a single enantiomer, it can be obtained either by resolution of the final product or by stereospecific synthesis from either isomerically pure starting material or an appropriate intermediate. Resolution of the final product, an intermediate or a starting material may be effected by any suitable method known in the art, for example, Chiral reagents for asymmetric synthesis by Leo A. Paquette; John Wiley & Sons Ltd (2003).
Additionally, in situations wherein tautomers of the compounds of formula I are possible, the present invention is intended to include all tautomeric forms of the compounds.
The present invention also encompasses within its scope prodrugs of the compound of formula I. Preferably, prodrugs are those compounds that are converted to their parent compound intracellular^, where the cellular converting location is the site of therapeutic action. The prodrugs of the compounds of the present invention are derivatives, particularly simple derivatives of the said compounds which upon administration to a subject in need thereof undergoes conversion by metabolic or chemical processes to release the parent drug (e.g. the compound of formula I) in wVo from which the prodrug is derived. For instance, preferred produgs are pharmaceutically acceptable ester derivatives convertible by solvolysis under physiological conditions to the parent carboxylic acid, e.g., alkyl esters, cycloalkyl esters, alkenyl esters, benzyl esters, mono- or di-substituted alkyl esters such as the pivaloyloxymethyl ester and the like conventionally used in the art (An introduction to Medicinal Chemistry, Graham. L. Patrick, Second Edition, Oxford University Press, pg 239-248; Prodrugs: Challenges and Rewards, Part 1 and Part 2, AAPS Press, Edited by Valentino J. Stella, Renald T. Borchardt, Michael J. Hagemon, Reza Oliyai, Hans Maag, Jefferson W. Tilley).
In one aspect, the present invention relates to a method for the treatment of a disease or disorder mediated by IL-17 or TNF-α, comprising administering to a subject in need thereof; a therapeutically effective amount of the compound of formula I or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof.
In another aspect, the present invention provides a compound of formula I or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof; for use as IL-17 inhibitor.
In another aspect, the present invention provides a compound of formula I or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof; for use as TNF-a inhibitor. In yet another aspect of the present invention, there is provided a compound of formula I or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or solvate thereof; for use in the treatment of a disease or a disorder mediated by IL-17 or TNF-a.
In yet another aspect, the present invention provides use of a compound of formula I or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof; for the manufacture of a medicament for the treatment of a disease or a disorder mediated by IL-1 7 or TNF-a.
In an embodiment of the present invention, the present invention encompasses within its scope all the diseases or disorders wherein IL-17 is implicated. In another embodiment of the present invention, the present invention encompasses within its scope all the diseases or disorders wherein TNF-a is implicated.
In an embodiment of the present invention, the disease or disorder mediated by the pro-inflammatory cytokines IL-17 or TNF-a is selected from the group consisting of an autoimmune or inflammatory disease or disorder; and a metabolic disease or disorder.
In an embodiment of the present invention, the autoimmune disorder, inflammatory disorder or metabolic disorder can be selected from the group consisting of: inflammatory bowel disease, rheumatoid arthritis, juvenile rheumatoid arthritis, psoriatic arthritis, osteoarthritis, refractory rheumatoid arthritis, chronic non-rheumatoid arthritis, ankylosing spondylitis, osteoporosis/bone resorption, chronic graft-versus-host disease, acute graft-versus-host disease, multiple sclerosis, systemic lupus erythematosus, Celiac Sprue, idiopathic thrombocytopenic thrombotic purpura, myasthenia gravis, Sjogren's syndrome, scleroderma, asthma, bronchitis, epidermal hyperplasia, Crohn's disease, atherosclerosis, septic shock syndrome, coronary heart disease, vasculitis, ulcerative colitis, psoriasis, adult respiratory distress syndrome, myolitis, polymyolitis, dermatomyolitis, polyarteritis nodossa, Wegener's granulomatosis, arteritis, polymyalgia rheumatica, sarcoidosis, sclerosis, primary biliary sclerosis, sclerosing cholangitis, dermatitis, atopic dermatitis, Still's disease, chronic obstructive pulmonary disease, Guillain-Barre disease, Type I diabetes, Graves' disease, Addison's disease, Raynaud's phenomenon, autoimmune hepatitis, psoriatic epidermal hyperplasia and delayed type hypersensitivity in skin disorders. In an embodiment of the present invention, the metabolic disease or disorder is selected from the group consisting of obesity, dyslipidemia, hyperlipidemia, hypertriglyceridemia, low adiponectin level, dyslipoproteinemia, impaired glucose tolerance, insulin resistance, increase in HbA1 c (glycosylated haemoglobin) level, reduced metabolic activity and Type II diabetes.
The present invention also encompasses within its scope the use of a compound of formula I or its pharmaceutically acceptable salt or a solvate in combination with other therapeutically active agents; wherein the compound of formula I and the further therapeutic agent are administered either simultaneously or sequentially. In another further aspect, the present invention relates to pharmaceutical composition(s) containing a therapeutically effective amount of at least one compound of formula I or its pharmaceutically acceptable salt or solvate thereof and a conventional pharmaceutically acceptable carrier. The present invention also relates to a process for the production of a pharmaceutical composition, which includes bringing at least one compound of formula I, into a suitable administration form using a pharmaceutically acceptable excipient and, if appropriate, further suitable active compounds, additives or auxiliaries can be added. The pharmaceutical composition(s) of the present invention can be administered orally, for example in the form of pills, tablets, coated tablets, capsules, granules or elixirs. Administration, however, can also be carried out rectally, for example in the form of suppositories, or parenterally, for example intravenously, intramuscularly or subcutaneously, in the form of injectable sterile solutions or suspensions, or topically, for example in the form of ointments or creams or transdermal^, in the form of patches, or in other ways, for example in the form of aerosols or nasal sprays.
The pharmaceutical composition(s) according to the invention is/are prepared in a manner known and familiar to one skilled in the art. Pharmaceutically acceptable inert inorganic and/or organic carriers and/or additives can be used in addition to the compound(s) of formula I, and/or its (their) physiologically tolerable salt(s). For the production of pills, tablets, coated tablets and hard gelatin capsules it is possible to use, for example, lactose, corn starch or derivatives thereof, gum arabica, magnesia or glucose, etc. Carriers for soft gelatin capsules and suppositories are, for example, fats, waxes, natural or hardened oils, etc. Suitable carriers for the production of solutions, for example injection solutions, or of emulsions or syrups are, for example, water, physiological sodium chloride solution or alcohols, for example, ethanol, propanol or glycerol, sugar solutions, such as glucose solutions or mannitol solutions, or a mixture of the various solvents which have been mentioned.
The pharmaceutical composition(s) normally contain about 1 % to 99 %, for example, about 5 % to 70 %, or from about 1 0 % to about 30 % by weight of the compound of formula I or its pharmaceutically acceptable salt. The amount of the compound of formula I or its pharmaceutically acceptable salt in the pharmaceutical composition(s) can range from about 1 mg to about 1000 mg or from about 2.5 mg to about 500 mg or from about 5 mg to about 250 mg or in any range falling within the broader range of 1 mg to 1000 mg or higher or lower that the specified range.
As is customary, the dosage range which are suitable in a specific case depend on the type of inflammatory disorder or autoimmune disorder to be treated and on the state of the respective condition or disorder. The selected dosage level can be readily determined by a skilled medical practitioner in the light of the relevant circumstances, including the condition (inflammatory disorder or autoimmune disorder) to be treated, the chosen route of administration including other factors such as age, weight and physical health and response of the individual patient, pharmacokinetics, severity of the disease and the like, factors known in the medical art. Actual dosage levels of the active ingredients i.e. the compounds of formula I in the pharmaceutical composition of this present invention can be varied so as to obtain an amount of the active ingredient, which is effective to achieve the desired therapeutic response for a particular patient (subject in need of the treatment), composition, and mode of administration without being toxic to the patient. Typically, the dose of the compounds of formula I or pharmaceutically acceptable salts thereof, which is to be administered, can cover a wide range. The dose to be administered daily is to be selected to suit the desired effect. A suitable dosage is about 0.01 mg/kg/day to about 200 mg/kg/day of the compound of formula I or its pharmaceutically acceptable salt, for example, about 0.1 mg/kg/day to 100 mg/kg/day of a compound of formula I or its pharmaceutically acceptable salt. If required, higher or lower daily doses can also be administered.
In addition to the compound of the formula I or its pharmaceutically acceptable salt and carrier substances, the pharmaceutical compositions can contain additives such as, for example, fillers, antioxidants, dispersants, emulsifiers, defoamers, flavors, preservatives, solubilizers or colorants. Pharmaceutical compositions can also contain two or more compounds of formula I or their physiologically tolerable salts. Furthermore, in addition to at least one compound of formula I or its physiologically tolerable salt, the pharmaceutical preparations can also contain one or more other therapeutically or prophylactically active ingredients.
The present invention also encompasses within its scope use of a compound of formula I or its pharmaceutically acceptable salt or a solvate in combination with other therapeutically active agents. The compound of formula I or its pharmaceutically acceptable salt, can be administered either simultaneously or sequentially in combination with the further therapeutically active agents. For instance, a pharmaceutical composition including a compound of formula I or a pharmaceutically acceptable salt can be administered to a mammal, in particular a human, with any other pharmaceutically active compounds, in mixtures with one another or in the form of pharmaceutical compositions.
The therapeutically active agents that can be used in combination with one or more compounds of formula I or its pharmaceutically acceptable salt can be selected from: monoclonal antibodies, non-steroidal anti-inflammatory drugs (NSAIDs) such as COX inhibitors or PDE4 inhibitors. Examples of monoclonal antibodies include, but not limited to, rituximab, etanercept, infliximab, adalimumab, natalizumab and vedolizumab. Examples of NSAIDs include, but not limited to, diclofenac, indomethacin, sulindac, mefenamic acid, piroxicam, ibuprofen, naproxen, ketoprofen, phenylbutazone, aspirin, diflunisal, nimesulide, celecoxib, valdecoxib, etorcoxib and meloxicam. Examples of PDE4 inhibitors include, but not limited to, rolipram, ibudilast, luteolin and roflumilast. It is understood that modifications that do not substantially affect the activity of the various aspects of this invention are included. Accordingly, the following examples are intended to illustrate but not to limit the present invention.
Abbreviations, which may be used in the description of the schemes and in the examples that follow, include:
CH3CN Acetonitrile
DCM Dichloromethane
DMF N, N-Dimethylformamide
DMSO Dimethyl sulfoxide
EDC 1 -Ethyl-3-(3-dimethylaminopropyl)carbodiimide
FBS Fetal bovine serum
g / gm Gram
h Hour(s)
3H Tritium
H2O Water
HCI Hydrochloric acid
HOBt Hydroxybenzotriazole hPBMC Human peripheral blood mononuclear cells
IL-17 lnterleukin-1 7
LAH Lithium aluminium hydride
MEM EBS Minimum essential medium with Earle's balance salt solution
mmol Millimolar
mL Millilitre
NaOH Sodium hydroxide
Na2S04 Sodium sulphate
NaHC03 Sodium bicarbonate
PBS Phosphate buffered saline
PMA Phorpbol myristate acetate
PTSA p-Toluenesulfonic acid
RT Room temperature (25-30 °C)
REG3A Regenerating islet-derived protein 3-alpha
RPMI Roswell Park Memorial Institute Medium
THF Tetrahydrofuran
TEA Triethylamine
Th17 T-helper cell - 17
TNF-oc Tumor necrosis factor - alpha
t-BuOH fert-Butyl alcohol
Vol Volume
Xantphos 4,5-Bis(diphenylphosphino)-9,9-dimethylxanthene
Examples
Example 1 :
1 -(5-Methyl-1 -(4-(trif luoromethyl)phenyl)-1 H-pyrazol-4-yl)-3-(2-(trif luoromethyl)- 1 H-benzo[d]imidazol-6-yl)urea Step 1) 1-(4-(trifluoromethyl)phenyl)-5-methyl-1 H-pyrazole-4-carboxylic acid (intermediate 1 )
To the stirred solution of ethyl 1 -(4-(trifluoromethyl)phenyl)-5-methyl-1 H-pyrazole-4- carboxylate (1 2.75 mmol) in THF: H20 (1 :1 vol, 20 mL) was added NaOH (25.50 mmol) and the resulting mixture was heated to reflux for 4 h. The reaction mixture was cooled to a room temperature and THF was removed. The aqueous layer was separated, washed with ethyl acetate and acidified with dilute HCI to afford the title compound.
Step 2): Azido(1-(4-(trifluoromethyl)phenyl)-5-methyl-1 H-pyrazol-4-yl) methanone (intermediate 2)
Method A:
To a stirred solution of the compound obtained in step 1 [intermediate 1 ] (3.90 mmol) in acetone (10 mL) in an ice bath, was added trichloroacetonitrile (7.81 mmol), triphenyl phosphine (7.81 mmol) and sodium azide (4.68 mmol). The reaction mixture was stirred for 2 h at room temperature. On completion of reaction, the reaction was quenched with water (10 mL). The reaction mixture was extracted with ethyl acetate (25 mL). The organic layer was separated, washed with brine (1 0 mL), dried with Na2SO4, concentrated and purified using column chromatography (silica gel, ethyl acetate and petroleum ether gradient) to afford the title compound.
Method B:
To a stirred solution of the compound obtained in step 1 [intermediate 1 ] (2.23 mmol) in dry acetone (5 mL) was added sodium azide (4.47 mmol), triethylamine (4.47 mmol) at
0
0 C. Triphosgene (1 .1 1 mmol) dissolved in dry acetone (5 mL) was added to the resulting reaction mixture drop wise over a period of 15 minutes. The resulting reaction mixture was stirred at room temperature for 12 h. After completion of the reaction, the white precipitate was filtered out, and washed with acetone. The resulting organic layer was evaporated to afford a crude product, which was further purified using column chromatography (silica gel, ethyl acetate and petroleum ether gradient) to afford the title compound.
Step 3): 1-(5-Methyl-1-(4-(trifluoromethyl)phenyl)-1 H-pyrazol-4-yl)-3-(2-(trifluoro methyl)-1 H-benzo[d]imidazol-6-yl)urea (final compound)
The compound obtained in step 2 [intermediate 2] (0.339 mmol) and 2-(trifluoromethyl)- 3H-benzo[d]imidazol-5-amine (0.339 mmol) in toluene (3 mL) was refluxed for 5 h. The reaction mixture was allowed to cool to the room temperature. The precipitated compound was filtered, washed with petroleum ether and cold dichloromethane, and dried to afford the title compound. 1 H NMR (DMSO-d6, 300 MHz): δ 2.35 (s, 3H), 7.21 (br s, 1 H), 7.62 (m, 1 H), 7.79 (d, J= 8.1 Hz, 2H), 7.89 (s, 3H), 8.05 (d, J= 1 1 .7 Hz, 2H), 8.86 (s, 1 H), 1 3.68 (br s, 1 H); MS: m/z 469.0 (M+H)+. Example 2:
1-(4-(4-(4-Chlorophenyl)-4-hydroxypiperidin-1-yl)-3-cyanophenyl)-3-(5-methyl-1- (4-(trifluoromethyl)phenyl)-1 H-pyrazol-4-yl)urea
The compound of example 2 was prepared analogous to the procedure described in example 1 by reaction of 5-methyl-1 -(4-(trifluoromethyl)phenyl)-1 H-pyrazole-4-carbonyl azide with 5-amino-2-(4-(4-chlorophenyl)-4-hydroxypiperidin-1 -yl)benzonitrile in step 3. 1 H NMR (DMSO-de, 300 MHz): δ 1 .68-1 .72 (m, 2H), 1 .98-2.00 (m, 2H), 2.29 (s, 3H), 3.22-3.40 (m, 2H), 3.89-3.93 (m, 2H), 7.36 (d, J= 7.2 Hz, 2H), 7.50 (d, J= 8.1 Hz, 2H), 7.79-7.91 (m, 5H), 8.20(s, 1 H), 8.27 (s, 1 H), 8.44 (s, 1 H), 8.74 (s, 1 H); MS: m/z 595.8 (M+H)+.
Example 3:
1-(3-Cyano-4-(4-(2-hydroxyethyl)piperidin-1-yl)phenyl)-3-(5-methyl-1-(4-(trifluoro methyl)phenyl)-1 H-pyrazol-4-yl)urea
The compound of example 3 was prepared analogous to the procedure described in example 1 by reaction of 5-methyl-1 -(4-(trifluoromethyl)phenyl)-1 H-pyrazole-4-carbonyl azide with 5-amino-2-(4-(2-hydroxyethyl)piperidin-1 -yl)benzonitrile in step 3.
1 H NMR (DMSO-de, 300 MHz): δ 1 .29-1 .32 (m, 2H), 1 .41 -1 .43 (m, 2H), 1 .75-1 .79 (m, 2H), 2.32 (s, 3H), 2.68 (t, J= 1 1 .1 Hz, 2H), 3.46-3.48 (m, 3H), 4.38 (t, J= 4.8 Hz, 1 H), 7.13 (d, J= 8.7 Hz, 1 H), 7.54-7.57 (m, 1 H), 7.78-7.90 (m, 6H), 8.14 (s, 1 H), 8.74 (s, 1 H); MS: m/z 51 3.1 (M+H).
Example 4:
1 -(5-Methyl-1 -phenyl-1 H-pyrazol-4-yl)-3-(naphthalen-1 -yl)urea
The compound of example 4 was prepared analogous to the procedure described in example 1 by reaction of 5-methyl-1 -phenyl-1 H-pyrazole-4-carbonyl azide with naphthalen-1 -amine in step 3.
1 H NMR (DMSO-de, 300 MHz): δ 2.32 (s, 3H), 7.42-7.63 (m, 1 0H), 7.85 (s, 1 H), 7.93 (d, J= 7.8 Hz, 1 H), 8.04 (d, J= 7.5 Hz, 1 H), 8.13 (d, J= 7.8 Hz, 1 H), 8.40 (s, 1 H), 8.78 (s, 1 H); MS: m/z 343.0 (M+H). Example 5:
1-(1-(4-chlorophenyl)-5-methyl-1 H-pyrazol-4-yl)-3-(3-cyano-4-(4-morpholino piperidin-1-yl) phenyl) urea
The compound of example 5 was prepared analogous to the procedure described in example 1 by reaction of 1 -(4-chlorophenyl)-5-methyl-1 H-pyrazole-4-carbonyl azide with 5-amino-2-(4-morpholinopiperidin-1 -yl)benzonitrile in step 3.
1 H NMR (DMSO-de, 300 MHz): δ 1 .54-1 .58 (m, 2H), 1 .88-1 .92 (m, 2H), 2.08 (br s, 1 H), 2.20-2.30 (m, 6H), 2.73 (t, J= 1 1 .0 Hz, 2H), 3.39-3.42 (m, 4H), 3.58 (br s, 4H), 7.12 (d, J= 8.7 Hz, 1 H), 7.58 (br s, 4H), 7.78 (br s, 1 H), 7.84 (br s, 1 H), 8.1 0 (br s, 1 H), 8.74 (s, 1 H) ; MS: m/z 520.3 (M)+.
Example 6:
1-(3-cyano-4-(4-morpholinopiperidin-1-yl) phenyl)-3-(5-methyl-1-(4-(trifluoro methyl) phenyl)-1 H-pyrazol-4-yl) urea
The compound of example 6 was prepared analogous to the procedure described in example 1 by reaction of 5-methyl-1 -(4-(trifluoromethyl)phenyl)-1 H-pyrazole-4-carbonyl azide with 5-amino-2-(4-morpholinopiperidin-1 -yl)benzonitrile in step 3.
1 H NMR (DMSO-de, 300 MHz): δ 1 .51 -1 .62 (m, 2H), 1 .88-1 .92 (m, 2H), 2.25-2.28 (m, 3H), 2.33 (s, 3H), 2.73 (t, 2H, J = 1 1 .4 Hz), 3.33-3.42 (m, 4H), 3.58 (br s, 4H), 7.13 (d, 1 H, J = 9.0 Hz), 7.57 (d, 1 H, J = 7.5 Hz), 7.79-7.91 (m, 6H), 8.1 5 (s, 1 H), 8.76 (s, 1 H); MS: m/z 554.2 (M+H)+.
Example 7:
1-(3-cyano-4-(piperazin-1-yl) phenyl)-3-(5-methyl-1-(4-(trifluoromethyl) phenyl)-"! H- pyrazol-4-yl) urea
The compound of example 7 was prepared analogous to the procedure described in example 1 by reaction of 5-methyl-1 -(4-(trifluoromethyl)phenyl)-1 H-pyrazole-4-carbonyl azide with 5-amino-2-(piperazin-1 -yl)benzonitrile in step 3.
1 H NMR (DMSO-d6! 300 MHz): δ 2.35 (s, 3H), 2.49 (br s, 4H), 3.84 (br s, 4H), 7.22 (d, 1 H, J = 8.7 Hz), 7.61 (d, 1 H, J = 8.7 Hz), 7.80 (d, 2H, J = 8.1 Hz), 7.89 (d, 4H, J = 8.1 Hz), 8.63 (s, 1 H), 9.08 (s, 1 H), 9.48 (s, 1 H); MS: m/z 470.1 (M+H)+.
Example 8:
1-(5-chloro-6-morpholinopyridin-3-yl)-3-(5-methyl-1-(4-(trifluoromethyl) phenyl)- 1 H-pyrazol-4-yl) urea The compound of example 8 was prepared analogous to the procedure described in example 1 by reaction of 5-methyl-1 -(4-(trifluoromethyl)phenyl)-1 H-pyrazole-4-carbonyl azide with 5-chloro-6-morpholinopyridin-3-amine in step 3.
1 H NMR (DMSO-de, 300 MHz): δ 2.33 (s, 3H), 3.14 (br s, 4H), 3.73 (br s, 4H), 7.80 (d, 2H, J = 8.4 Hz), 7.85 (br s, 1 H), 7.89 (d, 2H, J = 8.1 Hz), 8.1 1 (s, 1 H), 8.25 (br s, 2H), 8.83 (s, 1 H); MS: m/z 481 .2 (M+H)+.
Example 9:
1-(3-cyano-4-(4-(4-fluoro-3-(trifluoromethyl) benzoyl) piperazin-1-yl) phenyl)-3-(5- methyl-1 -(4-(trif luoromethyl)phenyl)-1 H-pyrazol-4-yl)urea
The compound of example 9 was prepared analogous to the procedure described in example 1 by reaction of 5-methyl-1 -(4-(trifluoromethyl)phenyl)-1 H-pyrazole-4-carbonyl azide with 5-amino-2-(4-(4-fluoro-3-(trifluoromethyl)benzoyl)piperazin-1 -yl)benzonitrile in step 3.
1 H NMR (DMSO-d6, 300 MHz): δ 2.29 (s, 3H), 3.07 (br s, 4H), 3.60 (br s, 4H), 7.16 (d, 1 H, J = 9.0 Hz), 7.60-7.65 (m, 2H), 7.78-7.88 (m, 8H), 8.20 (s, 1 H), 8.84 (s, 1 H); MS: m/z 659.8 (M)+.
Example 10:
1-(3-Cyano-4-(4-morpholinopiperidin-1-yl) phenyl)-1-methyl-3-(5-methyl-1-(4- (trifluoromethyl) phenyl)-"! H-pyrazol-4-yl) urea
The compound of example 10 was prepared analogous to the procedure described in example 1 by reaction of 5-methyl-1 -(4-(trifluoromethyl)phenyl)-1 H-pyrazole-4-carbonyl azide with 5-(methylamino)-2-(4-morpholinopiperidin-1 -yl)benzonitrile in step 3.
Example 11 :
1-(1-(4-chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(4-morpholinopiperidin -1- yl)phenyl)urea
Step 1): 1-(4-chlorophenyl)-1 H-1 ,2,3-triazole-4-carboxylic acid (intermediate 1) To the stirred solution of ethyl 1 -(4-chlorophenyl)-1 H-1 ,2,3-triazole-4-carboxylate (12.75 mmol) in THF: H20 (1 :1 vol, 20 mL) was added NaOH (25.50 mmol) and the resulting mixture was heated to reflux for 4 h. The reaction mixture was cooled to a room temperature and THF was removed. The aqueous layer was separated, washed with ethyl acetate and acidified with dilute HCI to afford the title compound. Step 2): 1-(4-chlorophenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide (intermediate 2) Method A:
To a stirred solution of the compound obtained in step 1 [intermediate 1 ] (3.90 mmol) in acetone (10 mL) in an ice bath, was added trichloroacetonitrile (7.81 mmol), triphenyl phosphine (7.81 mmol) and sodium azide (4.68 mmol). The reaction mixture was stirred for 2 h at room temperature. On completion of reaction, the reaction was quenched with water (10 mL). The reaction mixture was extracted with ethyl acetate (25 mL). The organic layer was separated, washed with brine (1 0 mL), dried with Na2SO4, concentrated and purified using column chromatography (silica gel, ethyl acetate and petroleum ether gradient) to afford the title compound.
Method B:
To a stirred solution of the compound obtained in step 1 [intermediate 1 ] (2.23 mmol) in dry acetone (5 mL) was added sodium azide (4.47 mmol), triethylamine (4.47 mmol) at
0
0 C. Triphosgene (1 .1 1 mmol) dissolved in dry acetone (5 mL) was added to the resulting reaction mixture drop wise over a period of 15 minutes. The resulting reaction mixture was stirred at room temperature for 12 h. After completion of the reaction, the white precipitate was filtered out, and washed with acetone. The resulting organic layer was evaporated to afford a crude product, which was further purified using column chromatography (silica gel, ethyl acetate petroleum and ether gradient) to afford the title compound.
Step 3): 1-(1-(4-chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(4-morpholino piperidin -1 -yl)phenyl)urea (final compound)
The compound obtained in step 2 [intermediate 2] (0.339 mmol) and 2-(trifluoromethyl)- 3H-benzo[d]imidazol-5-amine (0.339 mmol) in toluene (3 mL) was refluxed for 5 h. The reaction mixture was allowed to cool to the room temperature. The precipitated compound was filtered, washed with petroleum ether and cold dichloromethane, and dried to afford the title compound.
1 H NMR (DMSO-d6, 300 MHz): δ 1 .64(m, 2H) 2.00 (m, 2H), 2.79-2.72(m, 2H), 3.43-3.33 (m, 2H), 3.64 (s, 4H), 7.17 (d, J= 8.7 Hz, 1 H), 7.59 (d, J= 8.7 Hz, 1 H), 7.67 (d, J= 8.4 Hz, 2H), 7.90 (s, 1 H), 8.00 (d, J= 8.4 Hz, 2H), 8.63 (s, 1 H), 8.96 (s, 1 H), 9.59 (s, 1 H); MS (M+H) 507.3. Example 12:
1- (4-([1 ,4'-bipiperidin]-1 '-yl)-3-cyanophenyl)-3-(1 -(4-chlorophenyl)-1 H-1 ,2,3-triazol- 4-yl)urea
The compound of example 12 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-chlorophenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 2- ([1 ,4'-bipiperidin]-1 '-yl)-5-aminobenzonitrile in step 3.
1 H NMR (DMSO-de, 300 MHz): δ 1 .42-1 .73 (m, 4H), 2.1 5-2.27 (m, 4H), 2.79-3.49 (m, 1 1 H), 6.81 (m, 1 H), 7.20 (d, J = 8.7 Hz, 1 H), 7.59-7.67 (m, 2H), 7.91 (s, 1 H), 7.97 (d, J = 8.4 Hz, 1 H), 8.62 (s, 1 H), 8.97 (s, 1 H), 8.98-9.1 2 (m, 1 H), 9.60 (s, 1 H); MS: m/z 505.2 (M+H)+.
Example 13:
2- (1 -(4-(3-(1 -(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)ureido)-2-cyanophenyl) piperidin-4-yl)ethyl diethyl phosphate
The compound of example 13 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 2-(1 -(4-amino-2-cyanophenyl)piperidin-4-yl)ethyl diethyl phosphate in step 3.
1 H NMR (DMSO-de, 300 MHz): δ 1 .25 (t, J = 7.2 Hz, 6H), 1 .32 (s, 9H), 1 .53-1 .63 (m, 4H), 1 .82 (d, J = 1 1 .4 Hz, 2H), 2.71 (t, J = 1 1 .1 Hz, 2H), 3.35-3.39 (m, 3H), 4.01 (qn, J = 7.2 Hz, 6H), 7.1 6 (d, J = 9.0 Hz, 1 H), 7.60 (d, J = 8.4 Hz, 3H), 7.82 (d, J = 8.4 Hz, 2H), 7.88 (s, 1 H), 8.52 (s, 1 H), 8.86 (s, 1 H), 9.51 (s, 1 H); MS: m/z 624.0 (M+H)+.
Example 14:
1 -(1 -(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(4-(2-methoxyethyl) piperidin-1-yl)phenyl)urea
The compound of example 14 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5-amino-2-(4-(2-methoxyethyl)piperidin-1 -yl)benzonitrile in step 3.
1 H NMR (DMSO-d6, 300 MHz): δ 1 .32 (s, 9H), 1 .60-1 .63 (m, 2H), 1 .90-1 .95 (m, 2H), 2.87 (t, J = 9.3 Hz, 2H), 3.26-3.56 (m, 10H), 7.15 (d, J = 8.7 Hz, 1 H), 7.58 (d, J = 9.0 Hz, 3H), 7.83 (d, J = 8.4 Hz, 2H), 7.88 (s, 1 H), 8.52 (s, 1 H), 8.87 (s, 1 H), 9.52 (s, 1 H); MS: m/z 502.2 (M+H)+. Example 15:
1-(1-(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(4-(4-morpholinopiperidin-1^ (trifluoromethyl)phenyl)urea
The compound of example 15 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 4-(4-morpholinopiperidin-1 -yl)-2-(trifluoromethyl)aniline in step 3.
1 H NMR (DMSO-d6): δ 1 .33 (s, 9H), 1 .49 (q, J = 6.3 Hz, 2H), 1 .88 (d, J = 7.2 Hz, 2H), 2.23-2.29 (m, 1 H), 2.47 (s, 4H), 2.72 (t, J= 7.2 Hz, 2H), 3.57 (s, 4H), 3.77 (d, J = 7.2 Hz, 2H), 7.1 1 (s, 1 H), 7.23 (d, J= 5.1 Hz, 1 H), 7.59 (d, J = 4.4 Hz, 2H), 7.61 (s, 1 H), 7.82 (d, J = 5.1 Hz, 2H), 8.04 (s, 1 H), 8.43 (s, 1 H), 9.86 (s, 1 H); MS: m/z 572.6 (M+H)+.
Example 16:
1- (3-cyano-4-(4-morpholinopiperidin-1-yl)phenyl)-3-(1-(4-morpholinophenyl)-1 H- 1 ,2,3-triazol-4-yl)urea
The compound of example 16 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-morpholinophenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5-amino-2-(4-morpholinopiperidin-1 -yl)benzonitrile in step 3.
1 H NMR (DMSO-d6, 300 MHz): δ 1 .56-1 .58 (m, 2H), 1 .89-1 .91 (m, 2H), 2.20-2.28 (m, 1 H), 2.45 (s, 4H), 2.75 (t, J = 6.9 Hz, 2H), 3.20 (s, 4H), 3.43 (d, J = 6.6 Hz, 2H), 3.59 (s, 4H), 3.76 (s, 4H), 7.1 1 (d, J =5.1 Hz, 2H), 7.15 (d, J = 5.4 Hz, 1 H), 7.57 (d, J = 5.1 Hz, 1 H), 7.75 (d, J = 5.1 Hz, 2H), 7.88 (s, 1 H), 8.43 (s, 1 H), 8.86 (s, 1 H), 9.45 (s, 1 H); MS: m/z 558.4 (M+H)+.
Example 17:
1 -(4-(1 ,1 ,1 ,3,3,3-hexaf luoro-2-hydroxypropan-2-yl)phenyl)-3-(1 -(4-isopropylphenyl) -1 H-1 ,2,3-triazol-4-yl)urea
The compound of example 17 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-isopropylphenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with
2- (4-aminophenyl)-1 ,1 ,1 ,3,3,3-hexafluoropropan-2-ol in step 3.
1 H NMR (DMSO-de, 300 MHz): δ 1 .25 (d, J = 4.2 Hz, 6H), 2.97-3.00 (m, 1 H), 7.46 (d, J = 4.8 Hz, 2H), 7.61 (s, 4H), 7.85 (d, J = 4.8 Hz, 2H), 8.53 (s, 1 H), 8.59 (s, 1 H), 9.03 (s, 1 H), 9.47 (s, 1 H); MS: m/z 488.1 (M+H)+. Example 18:
1-(1-(4-chlorophenyl)-5-methyl-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(4-morpholino piperidin-1 -yl)phenyl)urea
The compound of example 18 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-chlorophenyl)-5-methyl-1 H-1 ,2,3-triazole-4-carbonyl azide with 5-amino-2-(4-morpholinopiperidin-1 -yl)benzonitrile in step 3.
1 H NMR (DMSO-de, 300 MHz): δ 1 .21 (m, 2H), 1 .50-1 .53 (m, 2H), 2.16-2.20 (m, 4H), 2.69-2.76 (m, 2H), 3.37 (m, 3H), 3.56-3.57 (m, 7H), 7.14 (d, J= 8.7 Hz, 1 H), 7.48-7.69 (m, 5H), 8.62 (s, 1 H), 8.96 (s, 1 H); MS: m/z 521 .0 (M+H)+.
Example 19:
3-(3-(1 -(4-chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)ureido)-N-(4-hydroxy-3-methoxy benzyl)benzamide
The compound of example 19 was prepared analogous to the procedure described in example 1 1 by reaction of 1 1 -(4-chlorophenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with
3- amino-N-(4-hydroxy-3-methoxybenzyl)benzamide in step 3.
1 H NMR (DMSO-d6, 300 MHz): δ 3.74 (s, 3H), 4.36 (d, J= 5.7 Hz, 2H ), 6.71 (s, 2H), 6.90 (s, 1 H), 7.37-7.40 (m, 1 H), 7.48-7.50 (m, 1 H), 7.61 -7.66 (m, 3H), 7.96-8.01 (m, 3H), 8.64 (s, 1 H), 8.84 (s, 1 H), 8.91 (br s, 2H), 9.50 (s, 1 H); MS: m/z 490.7 (M-H).
Example 20:
4- (1 -(2-cyano-4-(3-(1 -(4-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)ureido) phenyl)piperidin-4-yl)butanoic acid
The compound of example 20 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 4-(1 -(4-amino-2-cyanophenyl)piperidin-4-yl)butanoic acid in step 3.
1 H NMR (DMSO-de, 300 MHz): δ 1 .22-1 .28 (m, 4H), 1 .54-1 .50 (m, 2H), 1 .75-1 .79 (m, 2H), 2.19-2.23 (m, 2H), 2.70 (t, J= 10.8 Hz, 2H), 3.26 (m, 3H), 7.14 (d, J= 9.00 Hz, 1 H), 7.57 (d, J= 7.2 Hz, 1 H), 7.88 (s, 1 H), 7.96 (d, J= 8.4 Hz, 2H), 8.22 (d, J= 8.1 Hz, 2H), 8.74 (s, 1 H), 8.90 (s, 1 H), 9.61 (s, 1 H), 12.03 (br s, 1 H); MS: m/z 542.0 (M+H).
Example 21 :
1-(3-cyano-4-(4-(pyrimidin-2-yl)piperazin-1-yl)phenyl)-3-(1-(4-(trifluoromethyl) phenyl)-1 H-1 ,2,3-triazol-4-yl)urea The compound of example 21 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5-amino-2-(4-(pyrimidin-2-yl)piperazin-1 -yl)benzonitrile in step 3.
1 H NMR (DMSO-de, 300 MHz): δ 3.19-3.24 (m, 4H), 3.91 (m, 4H), 6.74 (t, J= 4.5 Hz, 1 H), 7.21 (d, J= 9.0 Hz, 1 H), 7.62 (d, J= 6.9 Hz, 1 H),7.97 (d, J= 8.4 Hz, 3H), 8.22 (d, J= 8.4 Hz, 2H), 8.40 (d, J= 4.5 Hz, 2H), 8.76 (s, 1 H), 8.95 (s, 1 H), 9.65 (s, 1 H); MS: m/z 535.1 (M+H).
Example 22:
1-(1-(4-chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(piperazin-1-yl)phenyl) urea
The compound of example 22 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-chlorophenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5- amino-2-(piperazin-1 -yl)benzonitrile in step 3.
1 H NMR (DMSO-de, 300 MHz): δ 3.27 (m, 8H), 7.25 (d, J= 8.7 Hz, 1 H), 7.65 (d, J= 8.7 Hz, 3H), 7.94-7.99 (m, 3H), 8.61 (s, 1 H), 8.89 (s, 1 H), 9.22 (s, 1 H), 9.67 (s, 1 H); MS: m/z 423.1 (M+H).
Example 23:
1-(1-(4-chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(5-(4-morpholinopiperidin-1-yl) pyridin-2-yl)urea
The compound of example 23 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-chlorophenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5- (4-morpholinopiperidin-1 -yl)pyridin-2-amine in step 3.
1 H NMR (DMSO-de, 300 MHz): δ 1 .17(bs, 2H), 1 .34(m, 2H), 1 .81 (d, J=1 1 .4Hz, 2H), 2.46(bs, 3H), 2.74(t, J=12.0 Hz, 2H), 3.55(d, J=4.2Hz, 4H), 4.18(d, J=1 2.9 Hz, 2H), 6.81 (d, J=9.0 Hz, 1 H), 7.63(d, J=8.7 Hz, 2H), 7.67(d, J=2.7 Hz, 1 H), 7.96(d, J=8.7Hz, 2H), 8.16(d, J=2.4Hz, 1 H), 8.48(s, 1 H), 8.57(s, 1 H), 9.40 (s, 1 H); MS: m/z 483 (M+H). Example 24:
1-(1-(4-chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(4-cyano-5-(4-morpholinopiperidin -1- yl)pyridin-2-yl)urea
The compound of example 24 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-chlorophenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 2- amino-5-(4-morpholinopiperidin-1 -yl)isonicotinonitrile in step 3. 1 H NMR (DMSO-d6, 300 MHz): δ 1 .48(q, J=1 1 .7 Hz, 2H), 1 .86(d, J=1 1 .4Hz, 2H), 2.38(d, J=1 1 .4Hz, 1 H), 2.95(t, J= 12.3Hz, 2H), 3.57(s, 4H), 4.04(d, J=1 2.6Hz, 2H), 7.63(d, J=8.7Hz, 2H), 7.97(d, J=8.7Hz, 2H), 8.20(d, J=2.7Hz, 1 H), 8.44(d, J=2.4Hz, 1 H), 8.61 (s, 1 H), 8.86(s, 1 H), 9.70 (bs, 1 H); MS: m/z 508 (M+H).
Example 25:
1-(6-(4-morpholinopiperidin-1-yl)pyridin-3-yl)-3-(1-(3-(trifluoromethyl)pheny^ 1 ,2,3-triazol-4-yl)urea
The compound of example 25 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 6-(4-morpholinopiperidin-1 -yl)pyridin-3-amine in step 3.
1 H NMR (DMSO-d6, 300 MHz): δ 9.45 (s, 1 H), 8.78 (s, 1 H), 8.50 (s, 1 H), 8.34 (m, 2H), 8.18 (d, J=2.4Hz, 1 H), 7.84 (m, 2H), 7.69 (dd, J=2.7 & 6.3 Hz, 1 H), 6.85 (d, J= 9.0 Hz, 1 H), 4.22 (d, J=1 2.6 Hz, 2H), 3.56 (m, 4H), 2.78 (t, J=1 2.0 Hz, 2H), 2.46 (m, 4H), 2.34 (m, 1 H), 1 .84 (d, J= Hz, 2H), 1 .37 (m, 2H); MS: m/z 517 (M+1 ).
Example 26:
1-(5-(3-(1-(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)ureido)pyridin-2-yl)piperid 4-carboxylic acid
The compound of example 26 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 1 -(5-aminopyridin-2-yl)piperidine-4-carboxylic acid in step 3.
1 H NMR (DMSO-de, 300 MHz): δ 12.25 (bs, 1 H), 9.72 (s, 1 H), 9.21 ( s, 1 H), 8.51 (s, 1 H), 8.28 (s, 1 H), 7.84 (d, J=8.1 Hz, 3H), 7.58 (d, J=8.4 Hz, 2H), 7.26 (s, 1 H), 4.1 1 (d, J=12.9 Hz, 2H), 3.18 (m, 2H), 2.57 (m, 1 H), 1 .94 (d, J=1 1 .7 Hz, 2H), 1 .62 (m, 2H), 1 .31 (s, 9H); MS: m/z 464 (M+1 ).
Example 27:
1-(6-(4-(pyrimidin-2-yl)piperazin-1-yl)-5-(trifluoromethyl)pyridin-3-yl)-3-(1-(3- (trifluoromethyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)urea
The compound of example 27 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(3-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 6-(4-(pyrimidin-2-yl)piperazin-1 -yl)-5-(trifluoromethyl)pyridin-3-amine in step 3. 1H NMR (DMSO-d6, 300 MHz): δ 9.79 (s, 1H), 9.17 ( s, 1H), 8.88 (s, 1H), 8.55 (s, 1H), 8.47 (d, J=2.4 Hz, 1 H), 8.38 (m, 4H), 7.83 (m, 2H), 6.66 (t, J=4.8 Hz, 1 H), 3.85 (m, 4H), 3.18 (m, 4H); MS: m/z 579 (M+1). Example 28:
1-(5-chloro-6-(4-morpholinopiperidin-1-yl)pyridin-3-yl)-3-(1-(4-chlorophenyl)-1H- 1 ,2,3-triazol-4-yl)urea
The compound of example 28 was prepared analogous to the procedure described in example 11 by reaction of 1-(4-chlorophenyl)-1H-1,2,3-triazole-4-carbonyl azide with 5- chloro-6-(4-morpholinopiperidin-1-yl)pyridin-3-amine in step 3..
1H NMR (DMSO-de, 300 MHz): δ 9.63 (s, 1H), 8.89 (s, 1H), 8.63 (s, 1H), 8.23 (s, 1H), 8.08 (s, 1 H), 8.00 (d, J=8.4 Hz, 2H), 7.66 (d, J=8.1 Hz, 2H), 3.57 (m, 6H), 3.38 (m, 2H), 2.75 (m, 2H), 2.29 (m, 1H), 1.87 (m, 2H), 1.54 (m, 2H), 1.11 (m, 2H); MS: m/z 517 (M+1).
Example 29:
1-(1-(4-(tert-butyl)phenyl)-1H-1,2,3-triazol-4-yl)-3-(5-methyl-6-(4-morpholino piperidin-1-yl)pyridin-3-yl)urea
The compound of example 29 was prepared analogous to the procedure described in example 11 by reaction of 1-(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5-methyl-6-(4-morpholinopiperidin-1-yl)pyridin-3-amine in step 3.
1H NMR (DMSO-d6, 300 MHz): δ 9.45 (s, 1H), 8.66 (s, 1H), 8.51 (s, 1H), 8.16 (s, 1H), 7.85 (d, J=4.8 Hz, 2H), 7.70 (s, 1 H), 7.59 (d, J=4.8 Hz, 2H), 3.58 (m, 6H), 3.32 (m, 2H), 2.68 (t, J=7.2 Hz, 2H), 2.29 (s, 3H), 1.87 (m, 2H), 1.54 (m, 2H), 1.33 (s, 9H); MS: m/z 519 (M+1).
Example 30:
1-(1-(4-(tert-butyl)phenyl)-1H-1,2,3-triazol-4-yl)-3-(5-methyl-6-(4-(pyrimidin-2- yl)piperazin-1-yl)pyridin-3-yl)urea
The compound of example 30 was prepared analogous to the procedure described in example 11 by reaction of 1-(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5-methyl-6-(4-(pyrimidin-2-yl)piperazin-1 -yl)pyridin-3-amine in step 3.
1H NMR (DMSO-d6, 300 MHz): δ 9.47 (s, 1 H), 8.71 (s, 1 H), 8.39 (d, J=4.5 Hz, 3H), 8.19 (d, J=2.1 Hz, 1H), 7.85 (d, J=8.7 Hz, 2H), 7.76 (d, J=2.1 Hz, 1H), 7.59 (d, J=8.7 Hz, 2H), 6.66 (t, J=4.5 Hz, 1 H), 3.86 (m, 4H), 3.04 (m, 4H), 2.30 (s, 3H), 1 .32 (s, 9H); MS: m/z 513 (M+1 ).
Example 31 :
1-(1-(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(5-cyano-6-(4-(pyrimidin piperazin-1-yl)pyridin-3-yl)urea
The compound of example 31 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5-amino-2-(4-(pyrimidin-2-yl)piperazin-1 -yl)nicotinonitrile in step 3.
1 H NMR (DMSO-de, 300 MHz): δ 9.45 (s, 1 H), 8.92 (s, 1 H), 8.53 (s, 1 H), 8.50 (d, J=2.7 Hz, 1 H), 8.40 (d, J=4.5 Hz, 2H), 8.28 (d, J=2.7 Hz, 1 H), 7.85 (d, J=8.7 Hz, 2H), 7.60 (d, J=8.7 Hz, 2H), 6.69 (t, J=4.8 Hz, 1 H), 3.90 (m, 4H), 3.57 (m, 4H), 1 .32 (s, 9H); MS: m/z 524 (M+1 ). Example 32:
1-(6-(4-cyano-4-phenylpiperidin-1-yl)pyridin-3-yl)-3-(1-(3-(trifluoromethyl)phenyl)- 1 H-1 ,2,3-triazol-4-yl)urea
The compound of example 32 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(3-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 1 -(5-aminopyridin-2-yl)-4-phenylpiperidine-4-carbonitrile in step 3.
1 H NMR (DMSO-d6, 300 MHz): δ 9.49 (s, 1 H), 8.78 ( s, 1 H), 8.60 (s, 1 H), 8.32 (m, 2H), 8.25 (d, J=2.7 Hz, 1 H), 7.85 (t, J=6.3 Hz, 2H), 7.76 (dd, J=2.4 Hz, 1 H), 7.56 (d, J=7.5 Hz, 2H), 7.47 (t, J=7.2 Hz, 2H), 7.39 (m, 1 H), 6.98 (d, J=9.0 Hz, 1 H), 4.40 (d, J=1 3.8 Hz, 2H), 3.09 (t, J=12.3 Hz, 2H), 2.24 (d, J=13.2 Hz, 2H), 2.08 (t, J=12.3 Hz, 2H); MS: m/z 533 (M+1 ).
Example 33:
1-(3-cyano-4-(piperidin-1-yl)phenyl)-3-(1-(4-(trifluoromethyl)phenyl)-1 H-1 ,2,3- triazol-4-yl)urea
The compound of example 33 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5-amino-2-(piperidin-1 -yl)benzonitrile in step 3.
1 H NMR (DMSO-d6, 300 MHz): δ 1 .53 (s, 2H), 1 .67 (s, 4H), 3.02 (s, 4H), 7.15 (d, J=9.0 Hz, 1 H), 7.59 (m, 1 H), 7.88 (s, 1 H), 7.97 (d, J=8.4 Hz, 2H), 8.22 (d, J=8.1 Hz, 2H), 8.75 (s, 1 H), 8.88 (s, 1 H), 9.61 (s, 1 H); MS: m/z 454.2 (M-1 ). Example 34:
1-(3-cyano-4-(2-oxa-6-azaspiro[3.3]heptan-6-yl)phenyl)-3-(1-(4-(trifluoro
methyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)urea
The compound of example 34 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5-amino-2-(2-oxa-6-azaspiro[3.3]heptan-6-yl)benzonitrile in step 3.
1 H NMR (DMSO-de, 300 MHz): δ 4.23 (s, 4H), 4.72 ( s, 4H), 6.61 (d, J=9.0 Hz, 1 H), 7.47 (d, J=9.0 Hz, 1 H), 7.70 (s, 1 H), 7.97 (s, 1 H), 7.97 (d, J=8.4 Hz, 1 H), 8.22 (d, J=8.1 Hz, 2H), 8.72(d, J=12.3 Hz, 2H), 9.53 (s,1 H); MS: m/z 470.0 (M+1 ).
Example 35:
1-(1-(4-chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(4-(cyclopropylmethyl)pipera; 1-yl)phenyl)urea
The compound of example 35 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-chlorophenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5- amino-2-(4-(cyclopropylmethyl)piperazin-1 -yl)benzonitrile in step 3.
1 H NMR (DMSO-d6, 300 MHz): δ 0.37 (s, 2H), 0.66 (s, 2H), 0.85 (m, 1 H), 1 .08 (m, 2H), 3.14 (s, 4H), 3.50-3.68 (m, 4H), 7.27 (d, J=8.7 Hz, 1 H), 7.67 (d, J=8.7 Hz, 3H), 8.00 (m, 3H), 8.62 (s, 1 H), 9.00 (s, 1 H), 9.62 (s, 1 H); MS: m/z 475.9 (M-1 ).
Example 36:
1-(1-(4-chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(4-methoxypiperidin-1- yl)phenyl)urea
The compound of example 36 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-chlorophenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5- amino-2-(4-methoxypiperidin-1 -yl)benzonitrile in step 3.
1 H NMR (DMSO-de, 300 MHz): δ 1 .61 -1 .63 (m, 2H), 1 .97 (s, 2H), 2.87-2.90 (m, 2H), 3.24-3.26 (m, 2H), 3.28 (s, 3H), 3.38 (m, 1 H), 7.17 (d, J = 5.1 Hz, 1 H), 7.57 (d, J = 5.4 Hz, 1 H), 7.66(d, J = 5.1 Hz, 2H),7.88 (s, 1 H), 8.00 (d, J = 5.1 Hz, 2H), 8.63 (s, 1 H), 8.88 (s, 1 H), 9.56 (s, 1 H); MS: m/z 452.6 (M+H)+.
Example 37:
1-(3-cyano-4-(4-(3-hydroxyphenyl)piperazin-1-yl)phenyl)-3-(1-(p-tolyl)-1 H-1 ,2,3- triazol-4-yl)urea The compound of example 37 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(p-tolyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5-amino-2- (4-(3-hydroxyphenyl)piperazin-1 -yl)benzonitrile in step 3.
1 H NMR (DMSO-de, 300 MHz): δ 2.38 (s, 3H), 3.21 (s, 4H), 3.26 (m, 4H), 6.37 (s, 1 H), 6.45 (d, J=4.8 Hz, 1 H), 7.01 (m, 1 H), 7.23 (d, J=5.4 Hz, 1 H), 7.40(d, J=4.8 Hz, 2H), 7.64 (d, J=5.4 Hz, 1 H), 7.82 (d, J=4.8 Hz, 2H), 7.93 (s, 1 H), 8.53 (s, 1 H), 8.92 (s, 1 H), 9.1 7 (s, 1 H), 9.53 (s, 1 H); MS: m/z 495.3 (M+1 ).
Example 38:
1-(3-cyano-4-(4-morpholinopiperidin-1-yl)phenyl)-3-(1-(3-(trifluoromethyl)phenyl)- 1 H-1 ,2,3-triazol-4-yl)urea
The compound of example 38 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(3-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5-amino-2-(4-morpholinopiperidin-1 -yl)benzonitrile in step 3.
1 H NMR (DMSO-de, 300 MHz): δ 1 .58 (d, J=9.6 Hz, 2H), 1 .80-2.00 (s, 2H), 2.20-2.30 (m, 1 H), 2.38-2.60 (m, 4H), 2.74 (t, J=1 1 .4 Hz, 2H),3.33-3.58 (m, 2H), 3.58 (s, 4H), 7.1 6 (d, J=9.0 Hz, 1 H), 7.58 (d, J=7.2 Hz, 1 H), 7.84-7.89 (m, 3H), 8.29-8.34(m, 2H), 8.83 (s, 1 H), 8.90 (s, 1 H), 9.60 (s, 1 H); MS: m/z 541 .0 (M+1 ). Example 39:
1-(1-(4-chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(4-hydroxypiperidin-1- yl)phenyl)-1 ,3-dimethylurea
Step 1): Synthesis of 1-(1-(4-chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(4- hydroxypiperidin-1 -yl)phenyl)urea
The compound of step 1 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-chlorophenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5- amino-2-(4-hydroxypiperidin-1 -yl)benzonitrile in step 3. Step 2): Synthesis of 1-(1-(4-chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(4- hydroxypiperidin-1 -yl)phenyl)-1 ,3-dimethylurea
To the compound of step 1 in DMF was added excess of methyl iodide and potassium carbonate (2 equivalents). The resulting reaction mixture was heated at 50-70 °C for 12-16 h to afford title compound. 1 H NMR (DMSO-d6, 300 MHz): δ 1 .32 (s, 2H), 1 .33-1 .46 (m, 2H), 1 .70-1 .73 (m, 2H), 1 .63-1 .72 (m, 3H), 3.10 (s, 3H), 3.12 (s, 3H), 4.70 (brs, 1 H), 6.90 (d, J=9.0 Hz, 1 H), 7.18 (dd, J=10.5, 1 .8 Hz, 1 H), 7.35 (d, J=2.1 Hz, 1 H), 7.67 (d, J=8.7 Hz, 2H), 7.79 (d, J=8.7 Hz, 2H), 8.32 (s, 1 H); MS: m/z 466.1 (M+1 ).
Example 40:
1 -(4-(3-(1 -(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)ureido)-2-cyanophenyl)-4-(2- hydroxypropan-2-yl)piperidine
The compound of example 40 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5-amino-2-(4-(2-hydroxypropan-2-yl)piperidin-1 -yl)benzonitrile in step 3.
1 H NMR (DMSO-d6, 300 MHz): δ 1 .06 (s, 6H), 1 .30 (s, 9H), 1 .32-1 .40 (m, 2H), 1 .77- 1 .81 (m, 2H), 2.25-2.30 (m, 2H), 2.31 (s, 3H), 2.63-2.65 (m, 2H), 3.35-3.46 (m, 3H), 7.12 (d, 1 H, J = 9.0Hz), 7.56-7.59 (m, 3H), 7.81 -7.87 (m, 3H), 8.51 (s, 1 H), 8.87 (s, 1 H), 9.50 (s, 1 H); MS: m/z 502.2 [M +1 -(S03Me)]+.
Example 40A:
1 -(4-(3-(1 -(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)ureido)-2-cyanophenyl)-4-(2- hydroxypropan-2-yl)piperidin-1-ium methanesulfonate
The compound of example 40 (50 mg, 0.098 mmol) in dry ethyl acetate (2 ml.) was treated with methane sulphonic acid (9.4 mg, 0.098 mmol). The resulting reaction mixture was stirred for about 12-1 6 h at room temperature. Solvent was distilled off and residue was washed with ether and dried to afford methanesulfonate salt of example 40A.
Yield: 40 mg; 80 %. Example 41 :
1-(3-cyano-4-(piperazin-1 -yl)phenyl)-3-(1 -(4-(trifluoromethyl)phenyl)-1 H-1 ,2,3- triazol-4-yl)urea
The compound of example 41 was prepared analogous to the procedure described in example 1 1 by reaction of 1 -(4-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazole-4-carbonyl azide with 5-amino-2-(piperazin-1 -yl)benzonitrile in step 3.
1 H NMR (DMSO-d6, 300 MHz): δ 3.27-3.34 (m, 8H), 7.27 (d, J= 9.0 Hz, 1 H), 7.67 (d, J= 8.7 Hz, 1 H), 7.98-7.95 (m, 2H), 8.22 (d, J= 8.4 Hz, 2H), 8.74 (s, 1 H), 8.87 (s, 2H), 9.23 (s, 1 H), 9.74 (s, 1 H); MS: m/z 493.2 (M-HCI). Example 41 A:
1-(3-cyano-4-(piperazin-1-yl)phenyl)-3-(1-(4-(trifluoromethyl)phenyl)-1 H-1 ,2,3- triazol-4-yl)urea hydrochloride
The compound of example 41 was further treated with hydrochloric acid to obtain hydrochloric salt of compound of example 41 A.
Example 42:
1-(3-cyano-4-(4-morpholinopiperidin-1-yl)phenyl)-3-(3-(4-fluorophenyl)-5- methylisoxazol-4-yl)urea
Step 1) 3-(4-fluorophenyl)-5-methylisoxazole-4-carboxylic acid (intermediate 1)
To the stirred solution of ethyl 3-(4-fluorophenyl)-5-methylisoxazole-4-carboxylate (12.75 mmol) in THF: H2O (1 :1 vol, 20 mL) was added NaOH (25.50 mmol) and the resulting mixture was heated to reflux for 4 h. The reaction mixture was cooled to a room temperature and THF was removed. The aqueous layer was separated, washed with ethyl acetate and acidified with dilute HCI to afford the title compound.
Step 2): 3-(4-fluorophenyl)-5-methylisoxazole-4-carbonyl azide (intermediate 2) Method A:
To a stirred solution of the compound obtained in step 1 [intermediate 1 ] (3.90 mmol) in acetone (10 mL) in an ice bath, was added trichloroacetonitrile (7.81 mmol), triphenyl phosphine (7.81 mmol) and sodium azide (4.68 mmol). The reaction mixture was stirred for 2 h at room temperature. On completion of reaction, the reaction was quenched with water (10 mL). The reaction mixture was extracted with ethyl acetate (25 mL). The organic layer was separated, washed with brine (1 0 mL), dried with Na2SO4, concentrated and purified using column chromatography (silica gel, ethyl acetate and petroleum ether gradient) to afford the title compound.
Method B:
To a stirred solution of the compound obtained in step 1 [intermediate 1 ] (2.23 mmol) in dry acetone (5 mL) was added sodium azide (4.47 mmol), triethylamine (4.47 mmol) at
0
0 C. Triphosgene (1 .1 1 mmol) dissolved in dry acetone (5 mL) was added to the resulting reaction mixture drop wise over a period of 15 minutes. The resulting reaction mixture was stirred at room temperature for 12 h. After completion of the reaction, the white precipitate was filtered out, and washed with acetone. The resulting organic layer was evaporated to afford a crude product, which was further purified using column chromatography (silica gel, ethyl acetate and petroleum ether gradient) to afford the title compound.
Step 3): 1-(3-cyano-4-(4-morpholinopiperidin-1-yl)phenyl)-3-(3-(4-fluorophenyl)-5- methylisoxazol-4-yl)urea (final compound)
The compound obtained in step 2 [intermediate 2] (0.339 mmol) and 5-amino-2-(4- morpholinopiperidin-1 -yl)benzonitrile (0.339 mmol) in toluene (3 mL) was refluxed for 5 h. The reaction mixture was allowed to cool to the room temperature. The precipitated compound was filtered, washed with petroleum ether and cold dichloromethane, and dried to afford the title compound.
1 H NMR (DMSO-de, 300 MHz): δ 1 .53-1 .57 (m, 2H), 1 .91 (d, J= 1 1 .4 Hz, 2H), 2.27(m,2H) 2.36 (s, 2H), 2.67-2.75 (m, 2H), 3.58 (br s, 4H), 7.12 (d, J = 9.0 Hz, 1 H), 7.36 (t, J = 8.7 Hz, 2H), 7.55 (d, J = 8.7 Hz, 1 H), 7.76-7.80 (m, 3H), 7.96 (s, 1 H), 9.00 (s, 1 H); MS: m/z (M+H)+ 505.2.
Example 43:
1-(4-(tert-butyl) phenyl)-N-(3-cyano-4-(4-(2-hydroxyethyl) piperidin-1-yl) phenyl)- 1 H-1 , 2, 3-triazole-4-carboxamide
To a stirred solution of 1 -(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazole-4-carboxylic acid (0.214 mmol) in DMF (5 mL) was added EDC (45.1 mg, 0.235 mmol) and HOBt (36.0 mg, 0.235 mmol). The resulting reaction mixture was stirred for 30 min. 5-amino-2-(4-(2- hydroxyethyl)piperidin-1 -yl)benzonitrile (0.214 mmol) and triethylamine (0.1 1 9 mL, 0.855 mmol) was added to the reaction mixture. Resulting reaction mixture was stirred for 1 2-16 h at room temperature. After completion of the reaction, the solvent was evaporated and the crude product obtained was further purified using column chromatography (silica gel, methanol and dichloromethane gradient) to afford the title compound.
1 H NMR (DMSO-de, 300 MHz): δ 1 .22-1 .46 (m, 15H), 1 .76-1 .80 (m, 2H), 2.73 (t, 2H, J = 1 1 .4 Hz), 3.41 -3.50 (m, 4H), 4.38 (br s, 1 H), 7.18 (d, 1 H, J = 9.0 Hz), 7.63 (d, 2H, J = 8.4Hz), 7.90 (d, 2H, J = 8.7 Hz), 8.02 (d, 1 H, J = 7.2 Hz), 8.1 6 (br s, 1 H), 9.41 (s, 1 H), 10.78 (s, 1 H); MS: m/z 473.2 [M + H] +. Example 44:
1-(4-(tert-butyl) phenyl)-N-(3-cyano-4-(4-hydroxypiperidin-1-yl) phenyl)-1H-1, 2, 3- triazole-4-carboxamide
The compound of example 44 was prepared analogous to the procedure described in example 43 by reaction of 1-(4-(tert-butyl)phenyl)-1 H-1 ,2,3-triazole-4-carboxylic acid with 5-amino-2-(4-hydroxypiperidin-1 -yl)benzonitrile.
1H NMR (DMSO-de, 300 MHz): δ 1.34 (s, 9H), 1.57-1.59 (m, 2H), 1.87-1.99 (m, 2H), 2.89 (t, 2H, J = 6.0 Hz), 3.31-3.32 (m, 2H), 3.65 (br s, 1 H), 4.72 (d, 1 H, J = 2.4 Hz), 7.20 (d, 1H, J=5.4Hz), 7.64 (d, 2H, J=5.1 Hz), 7.92 (d,2H, J=5.1 Hz), 8.01 (d, 1 H, J = 5.4 Hz), 8.17 (br s, 1H), 9.42 (s, 1H), 10.78 (s, 1H); MS: m/z 445.3 [M+H]+.
Example 45:
1-(4-chlorophenyl)-N-(3-cyano-4-(4-morpholinopiperidin-1-yl)phenyl)-1H-1,2,3- triazole-4-carboxamide
The compound of example 45 was prepared analogous to the procedure described in example 43 by reaction of 1-(4-chlorophenyl)-1 H-1 ,2,3-triazole-4-carboxylic acid with 5- amino-2-(4-morpholinopiperidin-1-yl)benzonitrile.
1H NMR (DMSO-de, 300 MHz): δ 1.80-1.84 (m, 2H), 1.98 (d, J= 12 Hz, 2H), 2.39 (m, 1 H), 2.63 (s, 4H), 2.84 (t, J= 11.4 Hz, 2H), 3.67 (d, J= 11.4 Hz, 2H), 3.77 (s, 4H), 7.07 (d,J=9.0 Hz, 1H), 7.60 (d, J= 8.4 Hz, 2H ), 7.77 (d, J= 8.4 Hz, 3H), 8.05 (s, 1H), 8.58 (s, 1 H), 8.95 (s, 1 H) MS: m/z 490.2 (M-H)+.
Example 45A:
1-(4-chlorophenyl)-N-(3-cyano-4-(4-morpholinopiperidin-1-yl)phenyl)-1H-1,2,3- triazole-4-carboxamide hydrochloride
To the solution of compound of example 45 (0.015 g, 0.030 mmol) was added 10 mL ethyl acetate saturated in HCI and methanol (2 drops) with continuous stirring. The resulting reaction mixture was stirred for 10-12 h. The reaction solvent was decanted and triturated with hexane. The precipitate obtained was filtered and dried to afford the title compound.
1H NMR (DMSO-de, 300 MHz): δ 1.84 (m, 2H), 1.98-2.02 (m, 2H), 2.41 (m, 1 H), 2.64 (s, 4H), 2.84 (t, J=11.4 Hz, 2H), 3.67 (d, J=11.4 Hz, 2H), 3.77(s, 4H), 7.07 (d, J=8.7 Hz, 1H), 7.60 (d, J=8.4 Hz, 2H), 7.77 (d, J=8.4 Hz, 3H), 8.05 (s, 1H), 8.59 (s, 1H), 8.95 (s, 1 H); MS: m/z 492.80 [M+1 -(HCI)]. Example 46:
1-(4-chlorophenyl)-N-(3-cyano-4-(4-(pyrimidin-2-yl)piperazin-1-yl)phenyl)-1 H-1 ,2,3- triazole-4-carboxamide
The compound of example 46 was prepared analogous to the procedure described in example 43 by reaction of 1 -(4-chlorophenyl)-1 H-1 ,2,3-triazole-4-carboxylic acid with 5- amino-2-(4-(pyrimidin-2-yl)piperazin-1 -yl)benzonitrile.
1 H NMR (DMSO-de, 300 MHz): δ 3.18 (s, 4H), 3.93 (s, 4H), 6.68 (m, 1 H), 7.28 (d, J= 7.5 Hz, 1 H), 7.73-7.69 (m, 2H), 8.07 (d, J= 5.1 Hz, 3H), 8.24 (s, 1 H), 8.41 (d, J = 3.0 Hz, 2H), 9.50 (s, 1 H ), 10.86 (s, 1 H) MS: m/z 486.3 [M+H] +.
Example 47:
N-(3-cyano-4-(4-morpholinopiperidin-1 -yl) phenyl)-5-pentyl-1 -(3-(trifluoromethyl) phenyl)-1 H-1 ,2,3-triazole-4-carboxamide
The compound of example 47 was prepared analogous to the procedure described in example 43 by reaction of 5-pentyl-1 -(3-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazole-4- carboxylic acid with 5-amino-2-(4-morpholinopiperidin-1 -yl)benzonitrile.
1 H NMR (DMSO-d6, 500 MHz): δ 1 0.92 (s, 1 H), 7.97(s, 1 H), 7.95 (d, J=7.5Hz, 1 H), 7.90 (d, J=8Hz, 1 H), 7.84 (s, 1 H), 7.83(d, J=8.5Hz, 1 H), 7.72 (d, J=8.5Hz, 1 H), 7.64 (d, J=8.5Hz, 1 H), 7.20 (d, J= 9Hz, 1 H), 3.2 (s, 4H), 3.50 (d, J= 9Hz, 2H), 2.86-2.75 (m, 4H), 2.63(s, 4H), 1 .96-1 .94 (m, 2H), 1 .68-1 .58 (m, 4H), 1 .33-1 .23(m, 10H), 0.85 (t, J=7Hz, 3H); MS: m/z 596 (M+1 )+.
Example 48:
1-(4-chlorophenyl)-N-(3-cyano-4-(4-morpholinopiperidin-1-yl)phenyl)-1 H-1 ,2,3- triazole-5-carboxamide
The compound of example 48 was prepared analogous to the procedure described in example 43 by reaction of 1 -(4-chlorophenyl)-1 H-1 ,2,3-triazole-5-carboxylic acid with 5- amino-2-(4-morpholinopiperidin-1 -yl)benzonitrile.
1 H NMR (DMSO-d6, 300 MHz): δ 1 .85-1 .99 (m, 2H), 2.27 (d, J= 10.2 Hz, 2H), 2.79 (t, J= 1 1 .2Hz, 2H), 3.08-3.32 (m, 3H), 3.45-3.60 (m, 4H), 3.84-3.99 (m, 4H), 7.24 (d, J= 9.0 Hz, 1 H), 7.85 (d, J= 6.0 Hz, 1 H), 8.03 (s, 1 H), 8.62 (s, 1 H), 1 1 .1 1 (s, 1 H), 1 1 .26 (s, 1 H); MS: m/z 492.0 (M-HCI). Example 48 A:
1- (4-chlorophenyl)-N-(3-cyano-4-(4-morpholinopiperidin-1-yl)phenyl)-1 H-1 ,2,3- triazole-5-carboxamide hydrochloride
The compound of example 48 was further treated with hydrochloric acid to obtain hydrochloric salt of compound of example 48A.
Example 49:
N-(3-cyano-4-(4-morpholinopiperidin-1 -yl)phenyl)-3-(4-fluorophenyl)-4- methylisoxazole-5-carboxamide
To a stirred solution of 3-(4-fluorophenyl)-4-methylisoxazole-5-carboxylic acid (0.214 mmol) in DMF (5 mL) was added EDC (45.1 mg, 0.235 mmol) and HOBt (36.0 mg, 0.235 mmol). The resulting reaction mixture was stirred for 30 min. 5-amino-2-(4- morpholinopiperidin-1 -yl)benzonitrile (0.214 mmol) and triethylamine (0.1 19 mL, 0.855 mmol) was added to the reaction mixture. The resulting reaction mixture was stirred 12- 16 h at room temperature. After completion of the reaction, the solvent was evaporated and the crude product obtained was further purified using column chromatography (silica gel, methanol and dichloromethane gradient) to afford the title compound.
1 H NMR (CDCI3, 300 MHz): δ 7.70-7.63 (m, 3H), 7.31 -7.26 (m, 3H), 6.98-6.92 (m, 1 H), 3.76 (brs, 4H), 3.61 (d, J=1 1 .4 Hz, 2H), 2.79-2.75 (m, 5H), 2.63 (brs, 4H), 2.39-2.40 (m, 1 H), 2.20-1 .96 (m, 2H), 1 .80-1 .77 (m, 2H); MS: m/z 490.2 (M+1 )+.
Example 50:
3-(4-chlorophenyl)-N-(3-cyano-4-(4-morpholinopiperidin-1-yl)phenyl)isoxazole-5- carboxamide
The compound of example 50 was prepared analogous to the procedure described in example 49 by reaction of 3-(4-chlorophenyl)isoxazole-5-carboxylic acid with 5-amino-
2- (4-morpholinopiperidin-1 -yl)benzonitrile.
1 H NMR (CDCI3, 300 MHz): δ 1 .79-1 .86 (m, 2H), 1 .98-2.02 (m, 2H), 2.36-2.39 (m, 1 H), 2.62 (s, 4H), 2.86 (t, J = 1 1 .4 Hz, 2H), 3.69 (d, J= 1 1 .7 Hz, 2H), 3.76 (s, 4H), 7.05 (d, J = 9.0 Hz, 1 H), 7.34 (s, 1 H), 7.52 (d, J = 8.4 Hz, 2H), 7.75 (d, J = 8.7 Hz, 1 H), 7.82 (d, J = 8.4 Hz, 2H), 7.97 (s, 1 H), 8.22 (s, 1 H); ); MS: m/z 492.6 (M+H).
Example 51 :
N-(1 -(4-chlorophenyl)-5-methyl-1 H-pyrazol-4-yl)-3-cyano-4-(4-morpholinopiperidin -1-yl)benzenesulfonamide Step 1 ): Synthesis of 4-chloro-3-cyanobenzene-1-sulfonyl chloride (intermediate 1)
5-amino-2-chloro benzonitrile (0.59 g, 3.28 mmol) was added in one portion to the mixture of concentrated HCI (1 .0 mL) and glacial acetic acid (0.4 mL) in a two neck flask. The resulting reaction mixture was cooled to a temperature 0 °C. A solution of sodium nitrite [0.226 g, 3.28 mmol in water (0.4 mL)] was added drop wise to the reaction mixture at 0 °C. The reaction mixture was stirred for 1 h at 0 °C. Acetic acid (3 mL) was added to the reaction mixture with continuous stirring. Sulphur dioxide was introduced into the reaction mixture by a bubler tube. Cuprous chloride (0.325 g, 3.28 mmol) was added to the reaction mixture. The reaction mixture was cooled to 0 °C. The solid precipitate was filtered and washed with hexane and dried to afford the title compound;
1 H NMR (DMSO-de, 300 MHz): δ 7.85 (d, J=8.7 Hz, 1 H), 8.22 (m, 1 H), 8.35 (s, 1 H).
Step 2): Synthesis of_4-chloro-N-(1-(4-chlorophenyl)-5-methyl-1 H-pyrazol-4-yl)-3- cyanobenzenesulfonamide (Intermediate 2)
To the stirred solution of compound obtained in step 1 [intermediate 1 ] (0.193 g, 0.81 9 mmol) in dichloromethane (10 mL) was added 1 -(4-chlorophenyl)-5-methyl-1 H-pyrazol- 4-amine hydrochloride (0.200 g, 0.81 9 mmol) and triethylamine (0.166 g, 1 .639 mmol) at room temperature. After completion of the reaction, the reaction was quenched with water. The crude compound obtained was extracted with dichloromethane, dried over anhydrous Na2S04 and purified using column chromatography (silica gel, methanol and dichloromethane gradient) to afford the title compound.
1 H NMR (CDCI3, 300 MHz): δ 2.28 (s, 3H), 5.32 (s, 1 H), 6.13 (s, 1 H), 7.40 (d, J= 8.4Hz, 2H), 7.50 (d, J= 8.4 Hz, 2H), 7.71 (d, J= 8.4 Hz, 1 H), 7 .91 (m, 1 H), 8.13 (s, 1 H); MS: m/z 407.1 (M+1 )+.
Step 3): Synthesis of N-(1-(4-chlorophenyl)-5-methyl-1 H-pyrazol-4-yl)-3-cyano-4- (4-morpholinopiperidin-1 -yl)benzenesulfonamide (final compound)
To the stirred solution of compound obtained in step 2 [intermediate 2] (0.50 g, 1 .228 mmol) in acetonitrile (10 mL) was added morpholinopiperidine (0.209 g, 1 .228 mmol) and triethylamine (0.248 g, 2.455 mmol). The resulting reaction mixture was heated to 65-70 eC for about 1 2-16 h. The solvent was removed, and the residue was diluted with ethylacetate and filtered through celite. The organic layers obtained were combined and dried over anhydrous Na2S04. The solvent was removed and the crude product obtained was further purified using column chromatography (silica gel, methanol and dichloromethane gradient) to afford the title compound.
1 H NMR (CDCI3, 300 MHz): δ 1 .27-1 .31 (m, 2H), 1 .77-1 .81 (m, 2H), 2.00-2.04 (m, 2H), 2.29 (s, 3H), 2.58-2.66 (m, 4H), 3.03 (t, J= 1 1 .7 Hz, 2H), 3.79 (br s, 3H), 3.93 (d, J = 12.3 Hz, 2H), 5.97 (s, 1 H), 7.01 (d, J= 9.0 Hz, 2H), 7.14(s,1 H) 7.40 (d, J= 8.4 Hz, 2H, 7.49 (d, = J8.4 Hz, 2H), 7.77 (d, J=9.0 Hz,1 H) 7.97 (s, 1 H); MS: m/z 541 .1 (M+1 )+.
Example 52:
N-(4-(3-methoxyphenyl)pyrimidin-2-yl)-5-phenylthiazol-2-amine
To a stirred solution of 4-phenylthiazol-2-amine (0.453 mmol) in dioxane (10 mL) was added 2-chloro-4-(3-methoxyphenyl)pyrimidine (0.453 mmol), xantphos [4,5- Bis(diphenylphosphino)-9,9-dimethylxanthene] (0.045 mmol), cesium carbonate (0.906 mmol) and Tris(dibenzylidineacetone)dipd(0) (0.023 mmol). The resulting reaction mixture was refluxed for about 12-1 6 h. The solvent was evaporated, residue was diluted with water, extracted with dichloromethane, dried over anhydrous Na2S04 and purified using column chromatography (silica gel, methanol and dichloromethane gradient) to afford the title compound.
1 H NMR (DMSO-d6, 300 MHz): δ 3.90 (s, 3H), 7.1 6-7.18 (m, 1 H), 7.29-7.34 (m, 2H), 7.41 -7.46 (m, 2H), 7.52 (t, J=8.1 Hz, 1 H), 7.62 (s, 1 H), 7.67 (d, J=5.4Hz, 1 H), 7.87-7.95 (m, 3H), 8.72 (d, J=5.1 Hz, 1 H), 1 1 .9 (s, 1 H); MS: m/z: 361 .1 (M+1 ).
Example 53:
5-phenyl-N-(4-(4-(trifluoromethoxy)phenyl)pyrimidin-2-yl)thiazol-2-amine
The compound of example 53 was prepared analogous to the procedure described in example 52 by reaction of 4-phenylthiazol-2-amine with 2-chloro-4-(4- (trifluoromethoxy)phenyl)pyrimidine.
1 H NMR (DMSO-de, 300 MHz): δ 7.32 (d, J=7.2Hz, 1 H), 7.41 (t, J=7.5 Hz, 2H), 7.57- 7.61 (m, 3H), 7.65 (d, J= 5.1 Hz, 1 H), 7.93 (d, J= 7.2 Hz, 2H), 8.44 (d, J= 9.0 Hz, 2H), 8.73 (d, J=5.4 Hz, 1 H), 1 1 .96 (s, 1 H); MS: m/z 415.1 (M+1 ).
Example 54:
N-(4-(2-methoxyphenyl)pyrimidin-2-yl)-5-phenylthiazol-2-amine The compound of example 54 was prepared analogous to the procedure described in example 52 by reaction of 4-phenylthiazol-2-amine with 2-chloro-4-(2- methoxyphenyl)pyrimidine.
1 H NMR (DMSO-de, 300 MHz): δ 3.88 (s, 3H), 7.13-7.16 (m, 1 H), 7.1 8-7.22(m, 1 H), 7.28-7.31 (m, 1 H), 7.40 (t, J= 7.5 Hz, 2H), 7.49-7.56 (m, 3H), 7.91 (d, J= 7.5 Hz, 2H), 8.14 (d, J=6.9 Hz, 1 H), 1 1 .79 (s, 1 H); MS: m/z 361 .2 ( +1 ).
Example 55:
5-((6-fluorobenzo[d]thiazol-2-yl) amino)-2-(4-morpholinopiperidin-1-yl) benzo nitrile
The compound of example 55 was prepared analogous to the procedure described in example 52 by reaction of 6-fluorobenzo[d]thiazol-2-amine with 5-chloro-2-(4- morpholinopiperidin-1 -yl)benzonitrile.
1 H NMR (DMSO-de, 300 MHz): δ 1 .73-1 .84 (m, 2H), 2.22-2.28 (m, 2H), 2.76 (t, 2H, J = 1 1 .7 Hz), 3.12-3.24 (m, 3H), 3.51 -3.55 (m, 4H), 3.67 (t, 2H, J = 12.0 Hz), 4.03 (d, 2H, J = 1 2.6 Hz), 7.17-7.18 (m, 1 H), 7.26 (d, 1 H, J = 9.0 Hz), 7.60-7.64 (m, 1 H), 7.74-7.78 (m, 1 H), 7.81 -7.84 (m, 1 H), 8.22 (d, 1 H, J = 2.4 Hz), 10.65 (s, 1 H); MS: m/z 438.2 [M + HP- Example 56:
5-((4-(3, 5-dimethoxyphenyl) pyrimidin-2-yl) amino)-2-(4-morpholinopiperidin-1-yl) benzonitrile
To a stirred solution of 2-chloro-4-(3,5-dimethoxyphenyl)pyrimidine (0.199 mmol) and 5-amino-2-(4-morpholinopiperidin-1 -yl)benzonitrile (0.199 mmol) in N-methyl-2- pyrrolidinone (NMP, 1 mL) was added PTSA (0.1 99 mmol). The resulting reaction mixture was heated at 140 °C for 4 h. After completion of the reaction, the reaction mixture was cooled to the room temperature and quenched with ice water. The reaction mixture was extracted with ethyl acetate (1 0 mL). The organic layer was separated and washed with saturated NaHC03 solution (1 0 mL), water (10 mL) and brine (10 mL). The crude product was dried over anhydrous Na2S04, purified using using column chromatography (silica gel, methanol and dichloromethane gradient) using methanol and dichloromethane as eluent to afford the title compound.
1 H NMR (DMSO- d6, 300 MHz): δ 1 .55-1 .62 (m, 2H), 1 .88-1 .92 (m, 2H), 2.26-2.29 (m, 1 H), 2.49-2.51 (m, 2H), 2.74 (t, J= 1 1 .0 Hz, 2H), 3.32-3.44 (m, 4H), 3.58 (br s, 4H), 3.84 (s, 6H), 6.67 (s, 1 H), 7.1 6 (d, J= 9.0 Hz, 1 H), 7.31 (d, J= 2.1 Hz, 2H), 7.46 (d, J= 5.4 Hz, 1 H), 7.84 (d, J= 9.0 Hz, 1 H), 8.32 (br s, 1 H), 8.56 (d, J= 2.1 Hz, 1 H), 9.84 (s, 1 H); MS: m/z 501 .4 [M+H]+. Example 57:
5-((4-(3, 5-dimethoxyphenyl) pyrimidin-2-yl) amino)-2-(4-(hydroxymethyl) piperidin -1-yl) benzonitrile
The compound of example 57 was prepared analogous to the procedure described in example 56 by reaction of 2-chloro-4-(3,5-dimethoxyphenyl)pyrimidine with 5-amino-2- (4-(hydroxymethyl)piperidin-1 -yl)benzonitrile.
1 H NMR (DMSO-de, 300 MHz): δ 1 .22-1 .38 (m, 2H), 1 .71 -1 .80 (m, 2H), 1 .84-1 .94 (m, 1 H), 2.69-2.74 (m, 2H), 3.29-3.41 (m, 4H), 3.84 (s, 6H), 4.48-4.53 (m, 1 H), 6.67 (m, 1 H), 7.1 6 (d, J= 9.0 Hz, 1 H), 7.31 (d, J= 1 .8 Hz, 2H), 7.46 (d, J= 5.1 Hz, 1 H), 7.83 (d, J= 9.0 Hz, 1 H), 8.32 (br s, 1 H), 8.56 (d, J= 2.1 Hz, 1 H), 9.83 (s, 1 H); MS: 446.2 [M+H]+.
Example 58:
4-(2-methoxyphenyl)-N-(4-(4-(pyrimidin-2-yl)piperazin-1-yl)-3-(trifluoromethyl) phenyl) pyrimidin-2-amine
The compound of example 58 was prepared analogous to the procedure described in example 56 by reaction of 2-chloro-4-(2-methoxyphenyl)pyrimidine with 3- (trifluoromethyl)-4-(4-(pyrimidin-2-yl)piperazin-1 -yl)benzenamine.
1 H NMR (DMSO-de, 300 MHz): δ 2.72-2.87 (m, 2H), 3.82 (m, 7H), 6.64 (t, J= 4.8 Hz, 1 H), 7.10 (t, J= 7.5 Hz, 1 H), 7.21 (d, J= 8.4Hz, 1 H), 7.40 (d, J= 5.1 Hz, 1 H), 7.55-7.48 (m, 2H), 7.92-7.97 (m, 2H), 8.37-8.39 (m, 3H), 8.53 (d, J= 5.1 Hz, 1 H), 9.92 (s, 1 H); MS: m/z 508.2 [M+H]+.
Example 59:
1 ,1 ,1 ,3,3,3-hexafluoro-2-(4-((4-(3-methoxyphenyl)pyrimidin-2-yl)amino)phenyl) propan-2-ol
The compound of example 59 was prepared analogous to the procedure described in example 56 by reaction of 2-chloro-4-(3-methoxyphenyl)pyrimidine with 2-(4- aminophenyl)-1 ,1 ,1 ,3,3,3-hexafluoropropan-2-ol.
1 H NMR (DMSO-d6, 300 MHz): δ 3.85 (s, 3H), 7.1 1 -7.13 (m, 1 H), 7.44-7.48 (m, 2H), 7.60 (d, J= 8.4 Hz, 2H), 7.76 (d, J= 8.1 Hz, 2H), 7.96 (d, J= 8.7 Hz, 2H), 8.52 (s, 1 H), 8.58 (d, J= 5.4 Hz, 1 H), 9.92 (s, 1 H); MS: 444.1 [M+H]+. Example 60:
2-(4-hydroxypiperidin-1-yl)-5-((4-(3-methoxyphenyl)pyrimidin-2-yl)amino) benzonitrile
The compound of example 60 was prepared analogous to the procedure described in example 56 by reaction of 2-chloro-4-(3-methoxyphenyl)pyrimidine with 5-amino-2-(4- hydroxypiperidin-1 -yl)benzonitrile.
1 H NMR (DMSO-de, 300 MHz): δ 1 .59 (m, 2H), 1 .89 (m, 2H), 2.87 (m, 2H), 3.33(m, 2H), 3.64 (m, 1 H), 3.87 (s, 3H), 4.72 (d, J= 4.2 Hz, 1 H), 7.10-7.19 (m, 2H), 7.43-7.48 (m, 2H), 7.71 -7.73 (m, 2H), 7.85-7.89 (m, 1 H), 8.28 (d, J= 5.4 Hz, 1 H), 9.84 (s, 1 H); MS: m/z 402.2 [M+H]+.
Example 61 :
5-((4-morpholinopyrimidin-2-yl)amino)-2-(4-(pyridin-2-yl)piperazin-1-yl)
benzonitrile
The compound of example 61 was prepared analogous to the procedure described in example 56 by reaction of 2-chloro-4-morpholinopyrimidine with 5-amino-2-(4-(pyridin- 2-yl)piperazin-1 -yl)benzonitrile.
1 H NMR (CDCI3, 300 MHz): δ 9.25 (s, 1 H), 8.13-8.1 1 (m, 2H), 8.01 (d, J=6Hz, 1 H), 7.88 (dd, J=9.0, 2.7Hz, 1 H) ,7.57-7.52 (m, 1 H), 7.1 8 (d, J=9Hz,1 H), 6.89 (d, J=8.7Hz, 1 H), 6.68-6.64 (m, 1 H), 6.30 (d, J=6Hz, 1 H), 3.66-3.55 (m, 12H), 3.17 (bs,4H), MS: m/z 443 (M+1 ).
Example 62:
2-morpholino-5-((4-morpholinopyrimidin-2-yl)amino)benzonitrile
The compound of example 62 was prepared analogous to the procedure described in example 56 by reaction of 2-chloro-4-morpholinopyrimidine with 5-amino-2- morpholinobenzonitrile.
1 H NMR (DMSO-de, 300 MHz): δ 8.82 (s, 1 H), 7.95 (d, J=6Hz, 1 H), 7.55 (d, J=8.5Hz, 2H), 6.86 (d, J=9Hz ,2H), 6.19 (d, J=6Hz, 1 H), 3.72-3.73 (m, 4H), 3.66-3.65 (m, 4H), 3.54 (s, 4H), 3.00 (s, 4H).
Example 63:
5-((4-((2-morpholinoethyl)amino)pyrimidin-2-yl)amino)-2-(4-morpholinopiperidin- 1-yl)benzonitrile The compound of example 63 was prepared analogous to the procedure described in example 56 by reaction of 2-chloro-N-(2-morpholinoethyl)pyrimidin-4-amine with 5- amino-2-(4-morpholinopiperidin-1-yl)benzonitrile.
1H NMR (DMSO-de, 500 MHz): δ 9.08 (s, 1H), 8.18 (d, J=2.4Hz, 1H), 7.80-7.77 (m, 2H),7.06-7.03 (m, 2H), 5.95 (d, J=5.7Hz, 1 H), 3.57 (bs, 8H), 3.36-3.30 (m, 3H), 2.71 (t, J=11.4Hz, 2H), 2.47-2.22 (m, 9H), 1.88-1.84 (m, 2H),1.54-1.51 (m,2H); MS: m/z 493 (M+1).
Example 64:
5-((2-chloro-6-methoxyquinolin-4-yl) amino)-2-(4-morpholinopiperidin-1-yl) benzonitrile
The compound of example 64 was prepared analogous to the procedure described in example 56 by reaction of 2,4-dichloro-6-methoxyquinoline with 5-amino-2-(4- morpholinopiperidin-1-yl)benzonitrile.
1H NMR (DMSO-de, 300 MHz): δ 1.57-1.64 (m, 2H), 1.83-1.94 (m, 2H),2.27-2.34 (m, 2H), 2.83 (t, 2H, J= 11.4 Hz), 3.17-3.21 (m, 1H), 3.53-3.58 (m, 8H), 3.94 (s, 3H), 6.80 (br s, 1H), 7.26 (d, 1H, J= 9.0 Hz), 7.51-7.55 (m, 1H), 7.67 (d, 1H, J= 9.3 Hz), 7.90- 7.96 (m,2H), 8.05 (d, 1H, J=2.4 Hz), 10.06(s, 1H);MS:m/z 478.2 [M + H]+. Example 65:
2-(4-morpholinopiperidin-1-yl)-5-(quinolin-2-ylamino) benzonitrile
The compound of example 65 was prepared analogous to the procedure described in example 56 by reaction of 2-chloroquinoline with 5-amino-2-(4-morpholinopiperidin-1- yl)benzonitrile.
1H NMR (DMSO-de, 300 MHz): δ 1.56-1.60 (m, 2H), 1.89-1.93 (m, 2H), 2.23-2.28 (m, 2H), 2.72-2.79 (m, 3H), 3.42-3.46 (m, 4H), 3.59 (br s, 4H), 7.00 (d, 1H, J= 8.7 Hz), 7.19 (d, 1 H, J = 9.0 Hz), 7.30 (t, 1 H, J = 6.6 Hz), 7.58 (t, 1 H, J = 7.5 Hz), 7.66 (d, 1 H, J = 8.1 Hz), 7.73 (d, 1 H, J = 7.5 Hz), 7.98-8.02 (m, 1 H), 8.06 (d, 1 H, J = 9.0 Hz), 8.48 (d, 1 H, J = 2.4 Hz), 9.56 (s, 1 H); MS: m/z 414.2 [M + H] +.
Example 66:
5-((cyano(1-(3-(trifluoromethyl)phenyl)-1H-1,2,3-triazol-4-yl)methyl)amino)-2-(4-(2- hydroxypropan-2-yl)piperidin-1-yl)benzonitrile Step 1): Synthesis of (1-(3-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)methanol (intermediate 1)
To a solution of ethyl 1 -(3-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazole-4-carboxylate (17.53 mmol) in tetrahydrofuran (25 ml.) was added LAH (26.3 mmol). The resulting reaction mixture was stirred at room temperature. On completion of the reaction, the reaction was quenched with saturated solution of ammonium chloride. The reaction mixture was extracted with ethyl acetate. The organic layer was separated and washed with brine and water, and dried over anhydrous Na2SO4 to afford the title compound. 1 H NMR (DMSO-d6, 300 MHz): δ 8.89 (s, 1 H), 8.29 (m, 2H), 5.39 (t, J= 5.4 Hz, 1 H), 4.63 (d, J= 5.4 Hz, 2H).
Step-2: Synthesis of 1-(3-(trifluoromethyl) phenyl)-1 H-1 , 2, 3-triazole-4- carbaldehyde (intermediate 2)
To a stirred solution of the compound obtained in step 1 [intermediate 1 ] (1 1 .10 mmol) in chloroform (30 mL) was added manganese dioxide (1 1 .10 mmol). The resulting reaction mixture was heated at 70 °C for 3 h. On completion of the reaction, the reaction mixture was filtered through celite bed and residue was washed 2-3 times with chloroform. The combined filtrate was dried over anhydrous Na2SO4 to afford the title compound.
1 H NMR (DMSO-de, 300 MHz): δ 1 0.13 (s, 1 H), 9.75 (s, 1 H), 8.40-8.33 (m, 2H), 7.96- 7.86 (m, 2H).
Step-3: Synthesis of 5-((cyano(1-(3-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazol-4- yl)methyl)amino)-2-(4-(2-hydroxypropan-2-yl)piperidin-1-yl)benzonitrile (Final compound)
To a stirred solution of the compound obtained in step 2 [intermediate 2] (0.829 mmol) in tetrahydrofuran (20 mL) was added 5-amino-2-(4-(2-hydroxypropan-2-yl)piperidin-1 - yl)benzonitrile (0.829 mmol) and trifluoroacetic acid (0.035 mmol). The resulting reaction mixture was stirred at room temperature for 3 h. After the formation of imine compound, trimethylsilanecarbonitrile (1 .244 mmol) and tetrabutylammonium difluorotriphenylsilicate (1 .244 mmol) was added to the reaction mixture. The resulting reaction mixture was stirred for about 1 2-16 h. The solvent was removed and the crude residue was further purified using column chromatography (silica gel, methanol and dichloromethane gradient) to afford the title compound. 1 H NMR (DMSO-d6, 300 MHz): δ 9.24(s,1 H), 8.36 (s, 1 H), 8.34(d, J=7.5Hz, 1 H), 7.89- 7.84 (m, 2H), 7.23(s, 1 H), 7.13 (s, 2H), 6.94(d, J=9.3Hz, 1 H), 6.36 (d, J=9.6Hz, 1 H), 4.16 (s, 1 H), 2.64 (t, J=1 0.8Hz, 2H), 2.28(s, 1 H), 1 .81 (d, J=1 1 .7Hz, 2H), 1 .42-1 .23 (m, 4H), 1 .07(s, 6H); MS: m/z 51 0 (M+1 )+.
Example 67:
5-((cyano(1-(3-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)methyl)amino)-2-(4- (pyrimidin-2-yl)piperazin-1-yl)benzonitrile
The compound of example 67 was prepared analogous to the procedure described in example 66 by reaction of 1 -(3-(trifluoromethyl) phenyl)-1 H-1 , 2, 3-triazole-4- carbaldehyde with 5-amino-2-(4-(pyrimidin-2-yl)piperazin-1 -yl)benzonitrile in step 3. 1 H NMR (DMSO-d6, 300 MHz): δ 9.26(s, 1 H), 8.40-8.31 (m, 4H), 7.93 (d, J=8.1 Hz, 2H), 7.29 (s, 1 H), 7.18(s, 2H), 7.04 (d, J=9.6Hz, 1 H), 6.68 (t, J=4.8Hz, 1 H), 6.39 (d, J=6.3Hz, 1 H), 3.90(brs, 4H), 3.07 (t, J=4.5Hz, 4H); MS: m/z 531 (M+1 )+.
Example 68:
2-chloro-5-(((1-(4-chlorophenyl)-5-methyl-1 H-pyrazol-4-yl)methyl)amino) benzonitrile
Step 1): Synthesis of (1-(4-chlorophenyl)-5-methyl-1 H-pyrazol-4-yl)methanol (intermediate 1)
To a solution of ethyl 1 -(4-chlorophenyl)-5-methyl-1 H-pyrazole-4-carboxylate (17.53 mmol) in tetrahydrofuran (25 ml.) was added LAH (26.3 mmol). The resulting reaction mixture was stirred at room temperature. On completion of the reaction, the reaction was quenched with saturated solution of ammonium chloride. The reaction mixture was extracted with ethyl acetate. The organic layer was separated and washed with brine and water, and dried over anhydrous Na2SO4 to afford the title compound.
Step-2: Synthesis of 1-(4-chlorophenyl)-5-methyl-1 H-pyrazole-4-carbaldehyde (intermediate 2)
To a stirred solution of the compound obtained in step 1 [intermediate 1 ] (1 1 .10 mmol) in chloroform (30 ml.) was added manganese dioxide (1 1 .10 mmol). The resulting reaction mixture was heated at 70 °C for 3 h. On completion of the reaction, the reaction mixture was filtered through celite bed and residue was washed 2-3 times with chloroform. The combined filtrate was dried over anhydrous Na2SO4 to afford the title compound. Step-3: Synthesis of 2-chloro-5-(((1 -(4-chlorophenyl)-5-methyl-1 H-pyrazol-4- yl)methyl)amino) benzonitrile (Final compound)
To a stirred solution of the compound obtained in step 2 [intermediate 2] (0.829 mmol) in tetrahydrofuran (20 ml_) was added 5-amino-2-chlorobenzonitrile (0.829 mmol) and trifluoroacetic acid (0.035 mmol). The resulting reaction mixture was stirred at room temperature for 3 h. After the formation of imine compound, sodium borohydride was added to the reaction mixture. The resulting reaction mixture was stirred for about 12- 16 h. The solvent was removed and the crude residue was further purified using column chromatography (silica gel, methanol and dichloromethane gradient) to afford the title compound.
1 H NMR (CDCI3, 300 MHz): δ 2.34 (s, 3H), 3.99 (m, 1 H), 4.15 (s, 2H), 6.79-6.82 (m, 1 H), 6.88-6.92 (m, 1 H), 7.30 (s, 1 H), 7.42 (d, J= 8.7 Hz, 2H), 7.50(d, J= 8.7 Hz, 2H), 7.63 (s, 1 H); MS: m/z 357.4 [M+H]+. Example 69:
5-(((1-(4-chlorophenyl)-5-methyl-1 H-pyrazol-4-yl)methyl)amino)-2-(4-morpholino piperidin-1 -yl)benzonitrile
The compound of example 69 was prepared analogous to the procedure described in example 68 by reaction of 1 -(4-chlorophenyl)-5-methyl-1 H-pyrazole-4-carbaldehyde with 5-amino-2-(4-morpholinopiperidin-1 -yl)benzonitrile in step 3.
1 H NMR (CDCI3): δ 1 .77-1 .88 (m, 2H), 1 .96-2.00 (m, 2H), 2.33 (s, 3H), 2.36-2.43 (m, 1 H), 2.65-2.77 (m, 6H), 3.45 (d, J= 1 1 .1 Hz, 2H), 3.79 (s, 4H), 4.12 (s, 2H), 6.81 -6.86 (m, 2H), 6.97 (d, J= 8.7 Hz, 1 H), 7.41 (d, J = 8.4 Hz, 2H), 7.48 (d, J = 8.4 Hz, 2H), 7.62 (s, 1 H); MS: m/z 491 .6 (M+H)+.
Example 70:
N-(4-(1 ,1 ,1 ,3,3,3-Hexafluoro-2-hydroxypropan-2-yl)phenyl)-2-hydroxy-2-(1 -(3- (trif luoromethyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)acetamide
To a stirred solution of 2-hydroxy-2-(1 -(3-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazol-4- yl)acetic acid (0.214 mmol) in DMF (5 imL) was added EDC (45.1 mg, 0.235 mmol) and HOBt (36.0 mg, 0.235 mmol). The resulting reaction mixture was stirred for 30 min. 2- (4-aminophenyl)-1 ,1 ,1 ,3,3,3-hexafluoropropan-2-ol (0.214 mmol) and triethylamine (0.1 1 9 ml_, 0.855 mmol) was added to the reaction mixture. Resulting reaction mixture was stirred for 12-16 h at room temperature. After completion of the reaction, the solvent was evaporated and the crude product obtained was further purified using column chromatography (silica gel, methanol and dichloromethane gradient) to afford the title compound.
1 H NMR (DMSO-de, 500 MHz): δ 1 0.31 (s, 1 H), 9.01 (s, 1 H), 8.64 (s, 1 H), 8.32-8.28 (m, 2H)), 7.88-7.84 (m, 4H), 7.63 (d, J=3.8 Hz, 2H), 6.69 (d, J=5.5 Hz, 1 H), 5.42 (d, J=5.5 Hz, 1 H); MS: m/z 529.0 (M+1 ).
Example 71 :
N-(3-cyano-4-(4-methoxypiperidin-1-yl)phenyl)-2-hydroxy-2-(1 -(3 (trifluoromethyl) phenyl)-1 H-1 ,2,3-triazol-4-yl)acetamide
The compound of example 71 was prepared analogous to the procedure described example 70 by reaction of 2-hydroxy-2-(1 -(3-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazol yl)acetic acid with 5-amino-2-(4-methoxypiperidin-1 -yl)benzonitrile.
1 H NMR (DMSO-de, 500 MHz): δ 1 0.31 (s, 1 H), 9.01 (s, 1 H), 8.64 (s, 2H), 8.32-8.28 (m, 2H)) ,7.88-7.84 (m, 4H), 7.63 (d, J=8Hz, 2H), 6.89 (d, J=5.5Hz, 1 H), 5.42 (d, J=5.5Hz, 1 H), 3.27 (s, 3H), 2.92(t, J=9.5Hz, 2H), 1 .96 (bs, 2H), 1 .62(bs, 2H); MS: m/z 501 (M+1 )+.
Biological Assays The biological activity of the compounds of the present invention as IL-17 and TNF-a inhibitor can be confirmed by a number of biological assays known in the art. The exemplified biological assays, given below, have been carried out with certain representative compounds of the formula I (referred to as the test compounds) synthesized in the above examples.
Materials:
Cell- based assays: RPMI-1640 (AMIMED, Cat#1 -41 F50-1 ), MEM EBS (Amimed, Cat#1 -31 S22-I), Penicillin-Streptomycin Stabilized Solution (Sigma- Aldrich, Cat#P4333), FBS (Hyclone, Cat#SH30071 .03), Ficoll/Hypaque solution Histopaque- 1077 (Sigma, Cat#10771 ), DMSO (Sigma, Cat#D4540), PMA (Sigma-Aldrich, Cat#79346), lonomycin (Sigma, Cat#l0634), Human-IL-17 duoset, economy Pack, 45 plates (R&D systems, Cat#DY317E), EGTA (Sigma, Cat#E3889), β glycerophosphate (Sigma, Cat#G5422), 96 well white flat bottom plate (Nunc, Cat#136101 ), 96 flat bottom plates (NUNC, Cat#167008), Tritium (3H) Thymidine (Methyl-T)(BRIT, Cat#LCT 3) (Cat# = Catalogue number) Dose Preparation Test compounds are weighed and dissolved in required amount of DMSO to give a stock solution of 20 mg/mL and 20 mM respectively. Working stock of 200X was prepared for the test compounds according to procedure shown below.
Figure imgf000081_0001
Serial dilutions of 1 :10 (10 iL of working stock and 90 iL of DMSO) were prepared from each concentration of 20 mg/mL (mM) or 6 mg/mL (mM) in a log dose to give the next log dose working stock concentration.
Test System: Human peripheral blood cells isolated from healthy volunteers.
Methods
In vitro screening method to identify inhibitors of IL-17 and TNF-oc release
Isolation of peripheral blood mononuclear cells (PBMC): PBMC were obtained from healthy donors by centrifugation of heparinized venous blood over Ficoll/Hypaque solution (Histopaque-1077, Sigma). Mononuclear cells in the buffy layer were collected and washed three times in sterile PBS with 2 % FBS. Cells were suspended in RPMI- 1640 supplemented with 100 U/mL penicillin, 100 μς/ιηΙ. streptomycin and 1 0 % fetal bovine serum (FBS).
Cytokine release assay: The cells were uniformly plated in 96-well tissue culture plates (Nunc 167008) at a concentration of 1 x1 05 cells/well. After plating, different concentrations (0.003, 0.01 , 0.03, 0.1 , 0.3, 1 , 3 and 1 0 μg/mL) of the test compounds dissolved in dimethylsulfoxide (DMSO) were added to the cells and incubated at 37 °C for 30 min in 5 % CO2 atmosphere. The final concentration of DMSO was maintained at 0.5 %. The vehicle (0.5 % DMSO) was used as control. The cells were then stimulated with 25 ng/mL of PMA (P1585, SIGMA) and 1 μΜ ionomycin (SIGMA, I0634). The plates were incubated in 5 % CO2 air humidified atmosphere at 37 °C for 48 h. At the end of 48 h, the supernatant from the cells were collected and ELISA was performed to detect the level of IL-17 (R&D systems, DY31 7E) and TNF-a (BD Biosciences, 555212) in the supernatant.
Lymphocyte proliferation assay by 3H thymidine uptake: The cells were uniformly plated in 96-well tissue culture plates (Nunc 167008) at a concentration of 1 x1 05 cells/well. After plating, different concentrations (0.003, 0.01 , 0.03, 0.1 , 0.3, 1 , 3 and 1 0 μg/mL) of the test compounds dissolved in DMSO were added to the cells and incubated at 37 °C for 30 min in 5 % CO2 atmosphere. The final concentration of DMSO was maintained at 0.5 %. The vehicle (0.5 % DMSO) was used as control. The cells were then stimulated with 25 ng/mL of PMA (P1585, SIGMA) and 1 μΜ ionomycin (SIGMA, I0634). The plates were incubated in 5 % CO2 air humidified atmosphere at 37 °C for 48 h. At the end of 24 h, cells are pulsed with 0.5 μα/ well of 3[H]-Thymidine (BRIT, LCT3). The cells were washed twice with PBS to remove excess radioactivity. Radioactivity in the incorporated cells was captured on lysis with Microscint-40, Perkin Elmer and measured by using a liquid scintillation counter (TopCount, Perkin Elmer). The anti-proliferative effect of the test compounds was measured using the following formula.
Control (CPM) - Treated (CPM)
% Anti-proliferation = X 1 00
Control (CPM)
Controls consisted of PBMC with PMA and ionomycin (Stimulated) and PBMC with RPMI (Un-stimulated) The test compounds exhibited IL-17 and TNF-a inhibitory activity in the cytokine release assay in which the compounds were tested. The IC50 values for test compounds inhibiting IL-17 are given in Table 1 , whereas the IC50 values for test compounds inhibiting TNF-a is given in Table 2.
Table 1 : IC50 values of the test compounds (IL-17 inhibition)
Figure imgf000083_0001
IC50 range class
< 0.1 μΜ but < 1 μΜ
1 μΜ but < 10 μΜ
10 μΜ but < 1 000 μΜ Table 2: IC50 values of the test compounds (TNF-a inhibition)
Figure imgf000084_0001
Symbol IC50 range class
+++ < 0.1 μΜ but < 1 μΜ
++ >1 μΜ but < 10 μΜ
+ > 10 μΜ but < 1000 μΜ
Conclusion:
The IC50 values determined for the test compounds are indicative of their IL17 and TNF-a inhibitory activity.

Claims

We claim:
1 . A compound of formula I;
Figure imgf000085_0001
Formula I
wherein,
A is triazole, pyrazole, imidazole, oxazole, isoxazole, thiazole, pyrrole, a 5- or 6- membered saturated heterocyclic ring system or a heteroaryl ring system selected from:
Figure imgf000085_0002
wherein * indicates the point of attachment to L;
Ri and R3 are independently selected from the group consisting of hydrogen, hydroxy, halogen, cyano, nitro, (C C6)-alkyl, (CrC6)-alkoxy, (CrC6)-alkoxyalkyl, (Ci-C6)- alkylamino, (C3-Ci2)-cycloalkyl, 0-(C3-Ci2)-cycloalkyl, halo(C-|-C6)alkyl, halo(C C6)alkoxy, (C6-Ci4)-aryl, heterocyclyl, 0-(C6-Ci4)aryl, (C6-Ci4)ar(CrC6)alkyl, (C6- Ci4)ar(CrC6)alkyloxy, heteroaryl, C(0)Ra, C(0)ORa, S(0)nRa, NRaRb and C(0)NRaRb; n is an integer from 0 to 2; R2 is (CrC6)-alkyl, (C C6)-alkoxy, (C1-C6)-alkoxy(C1-C6)-alkyl, (C3-C12)-cycloalkyl, O- (C3-C12)-cycloalkyl, (C6-Ci4)-aryl, 0-(C6-C14)aryl, (Ce-C jartd-C^alkyl, (C6-Ci4)ar(C Ce)alkyloxy, heterocyclyl or heteroaryl;
R2 together with R-i can form a 5- or 6- membered saturated or unsaturated ring optionally containing 1 to 3 heteroatoms independently selected from the group consisting of O, N and S, which ring is unsubstituted or substituted with 1 to 5 groups independently selected from the group consisting of halogen, hydroxy, nitro, cyano, (C C6)-alkyl, (C C6)-alkoxy, (C C6)-alkoxyalkyl, (C C6)-alkylamino, (C3-Ci2)-cycloalkyl, O- (C3-Ci2)-cycloalkyl, halo(CrC6)alkyl, halo(CrC6)alkoxy, (C6-C )-aryl, 0-(C6-Cu)aryl, (C6-Ci4)ar(Ci-C6)alkyl, (C6-Ci4)ar(Ci-C6)alkyloxy, heterocyclyl, heteroaryl, C(0)Ra, C(0)ORa, S(0)nRa, NRaRb and C(0)NRaRb;
L is *N(R4)-C(0)-N(R5), *C(0)N(R5), *N(R4)-C(0), *CH(R6)-C(0)-N(R5), *CH(R6)N(R5), *N(R4)-S(0)2 or *NR4; wherein * indicates the point of attachment to A;
R4, R5 and R6 are independently selected from the group consisting of hydrogen, hydroxy, cyano, (CrC6)-alkyl, (CrC6)-alkoxy, (CrC6)-alkoxyalkyl, (CrC6)-alkylamino, (C3-Ci2)-cycloalkyl, 0-(C3-Ci2)-cycloalkyl, halo(CrC6)alkyl, halo(C C6)alkoxy, hydroxy d^-alkyl, C(0)Ra, C(0)ORa, S(0)nRa, P(0)n(0(Ci-C6)alkyl)2l O-P(O) (0(C C6)alkyl)2 and C(0)NRaRb;
Xi, X2, X and X5 are independently selected from N and C(R7); wherein R7 is hydrogen, halogen, hydroxy, nitro, cyano, (C-|-C6)-alkyl, (CrC6)-alkoxy, (C-|-C6)- alkoxy(CrC6)-alkyl, (CrC6)-alkylamino, (C3-Ci2)-cycloalkyl, halo(C C6)alkyl, halo(C C6)alkoxy, (C2-C6)-alkenyl, (C2-C6)-alkynyl, (C6-Ci4)-aryl, 0-(C6-Ci4)aryl, (C6-Ci4)ar(C C6)alkyl, (C6-Ci4)ar(Ci-C6)alkyloxy, heterocyclyl, O-heterocyclyl, heteroaryl, C(0)Ra, C(0)ORa, S(0)nRa, P(0)n(0(CrC6)alkyl)2, 0-P(0)(0(C C6)alkyl)2, NRaRb or C(0)NRaRb;
X3 is N or C(R8), wherein R8 is hydrogen, halogen, hydroxy, cyano, nitro, (CrC6)-alkyl, (CrC6)-alkoxy, (Ci-C6)-alkoxy(Ci-C6)alkyl, (C C6)-alkylamino, (C3-Ci2)-cycloalkyl, O- (C3-Ci2)-cycloalkyl, halo(C C6)alkyl, halo(C C6)alkoxy, (C6-Ci4)-aryl, 0-(C6-Ci4)aryl, (C6-Ci )ar(Ci-C6)alkyl, (C6-Ci )ar(CrC6)alkyloxy, heterocyclyl, O-heterocyclyl, heteroaryl, C(0)Ra, C(0)ORa, S(0)nRa, NRaRb or C(0)NRaRb; provided that no more than three of the X-i to X5 positions can be simultaneously N; when two or more of Xi , X2, X4 and X5 represent C( R7) then R7 groups on the two adjacent carbon atoms can form a 3-7 membered unsaturated or saturated fused ring optionally containing 1 to 3 heteroatoms independently selected from the group consisting of O, N and S; wherein the ring can be unsubstituted or substituted with 1 -3 groups of Rc; or
when any one of X2 and X4 represent C( R7) and X3 represents C(R8) then the R7 group together with the R8 group can form a 3-7 membered fused ring optionally containing 1 to 3 heteroatoms selected from O, N or S; wherein the ring can be unsubstituted or substituted with 1 -3 groups of Rc;
Ra and Rb are independently selected from the group consisting of hydrogen, (C-| -C6)- alkyl, (C3-Ci2)-cycloalkyl, halo(CrC6)alkyl, (C6-C )-aryl, (C6-Ci 4)ar(Ci -C6)alkyl, heterocyclyl, heteroaryl, and S(0)nRa; or
Ra and Rb, can form a 3-7 membered saturated or unsaturated ring optionally containing one or more heteroatoms selected from N, O or S, which is unsubstituted or substituted with one or more groups independently selected from the group consisting of halogen, hydroxy, cyano, nitro, (C C6)-alkyl, (C-i -C6)-alkoxy, (CrC6)-alkylamino, (C3- Ci2)-cycloalkyl, 0-(C3-Ci 2)-cycloalkyl, halo(C C6)alkyl, halo(CrC6)alkoxy, (C6-Ci4)-aryl, 0-(C6-C )aryl, (C6-Ci4)ar(Ci -C6)alkyl, heterocyclyl, heteroaryl, C(0)Ra, C(0)ORa, S(0)n Ra, P(0)n(0(Ci -C6)alkyl)2, 0-P(0)(0(C C6)alkyl)2, N RaRb and C(0) N RaRb; wherein:
each of the (Ci -C6)alkyl and (Ci -C6)-alkoxy can be unsubstituted or substituted with one or more groups independently selected from the group consisting of halogen, hydroxy, nitro, cyano, (CrC6)-alkoxy-(Rc)o-3, (C3-C12)-cycloalkyl-(Rc)0-3, 0-(C3-C12)-cycloalkyl- (Rc)o-3, halo(CrC6)alkyl, halo(CrC6)alkoxy, (C6-Ci4)-aryl-( Rc)o-3, O-(C6-Ci4)aryl-( Rc)0-3, (C6-Ci4)ar(Ci -C6)alkyl-(Rc)o-3, heterocyclyl-( Rc)o-3, heteroaryl-(Rc)0-3, C(0) Ra, C(0)ORa, S(0)n Ra, P(0)n(0(Ci -C6)alkyl)2j 0-P(0)(0(C C6)alkyl)2, N RaRb and C(0) N RaRb; heterocyclyl refers to a 3-1 2 membered saturated or partially unsaturated monocyclic or bicyclic ring system containing one to four identical or different heteroatoms independently selected from the group consisting of N, S and O; heteroaryl refers to a 5-10 membered aromatic monocyclic or bicyclic ring system containing one to four identical or different heteroatoms independently selected from the group consisting of N, S and O; each of the (C2-C6)-alkenyl, (C2-C6)-alkynyl, (C3-C12)-cycloalkyl, (C6-Ci4)-aryl, (C6- Ci )ar(CrC6)alkyl, heterocyclyl and heteroaryl can be unsubstituted or substituted with one or more groups independently selected from the group consisting of halogen, hydroxy, nitro, cyano, (CrC6)alkyl-(Rc)o-3, (Ci-C6)-alkoxy-(Rc)o-3, (C3-C 2)-cycloalkyl- (Rc)o-3, 0-(C3-Ci2)-cycloalkyl-(Rc)o-3, halo(CrC6)alkyl, halo(C C6)alkoxy, (C6-Ci4)-aryl- (Rc)o-3, 0-(C6-Ci4)aryl-(Rc)o-3, (C6-Ci4)ar(Ci-C6)alkyl-(Rc)o-3, heterocyclyl-(Rc)o-3, heteroaryl-(Rc)o-3, C(0)Ra, C(0)ORa, S(0)nRa, P(0)n(0(Ci-C6)alkyl)2, O-P(O) (0(C C6)alkyl)2, NRaRb and C(0)NRaRb; and
Rc is halogen, hydroxy, cyano, nitro, (Ci-C6)-alkyl-(Rc)o-i , (Ci-C6)-alkoxy-(Rc)o-i, (C3- Ci2)-cycloalkyl-(Rc)o-i , O-(C3-Ci2)-cycloalkyl-(Rc)0-i, hydroxy(Ci-C6)-alkyl-(Rc)0-i , halo(C C6)alkyl, halo(CrC6)alkoxy, (C6-C14)-aryl-(Rc)0-i, O-(C6-Ci4)aryl-(Rc)0-i, (C6- Ci4)ar(Ci-C6)alkyl-(Rc)o-i, heterocyclyl-(Rc)0-i , heteroaryl-(Rc)0-i, C(0)Ra, C(0)ORa, SRa, S(0)nRa, P(0)n(0(Ci-C6)alkyl)2j 0-P(0)(0(C C6)alkyl)2, NRaRb or C(0)NRaRb; and with the provisos that:
(c) when A is pyrazole then L is a group other than *C(0)N(R5) and *N(R4)-C(0); and
(d) when A is pyrazole and L is *N(R4)-C(0)-N(R5), then the said L group is not attached to 5-position of pyrazole;
or an isotopic form, a stereoisomer or a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
2. The compound according to claim 1 ,
wherein,
A is triazole, pyrazole, imidazole, oxazole, isoxazole, thiazole, pyrrole or a heteroaryl ring system selected from:
Figure imgf000089_0001
wherein * indicates the point of attachment to L; with the provisos that:
(c) when A is pyrazole then L is a group other than *C(0)N(R5) and *N(R4)-C(0); and
(d) when A is pyrazole and L is *N(R4)-C(0)-N(R5), then said L group is not attached to 5-position of pyrazole;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
3. The compound according to claim 1 or claim 2,
wherein,
A is triazole, pyrazole, imidazole, oxazole, isoxazole, thiazole or pyrrole;
with the provisos that:
(a) when A is pyrazole then L is a group other than *C(0)N(R5) and *N(R4)-C(0); and
(b) when A is pyrazole and L is *N(R4)-C(0)-N(R5), then said L group is not attached to 5-position of pyrazole;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
4. The compound according to any one of the claims 1 -3,
wherein,
A is triazole, pyrazole, isoxazole or thiazole; L is *N(R4)-C(0)-N(R5), *C(0)N(R5), *N(R4)-C(0), *CH(R6)-C(0)-N(R5), *CH(R6)N(R5), *N(R4)-S(0)2 or *NR4; wherein * indicates the point of attachment to A;
with the provisos that:
(c) when A is pyrazole then L is a group other than *C(0)N(R5) and *N(R4)-C(0); and
(d) when A is pyrazole and L is *N(R4)-C(0)-N(R5), then said L group is not attached to 5-position of pyrazole;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
5. The compound according to any one of the claims 1 -4,
wherein,
A is triazole;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
6. The compound according to any one of the claims 1 -4,
wherein,
A is pyrazole;
L is *N(R4)-C(0)-N(R5); *CH(R6)-C(0)-N(R5); *CH(R6)N(R5); *N(R4)-S(0)2 or *NR4; wherein * indicates the point of attachment to A;
with the proviso that when L is *N(R4)-C(0)-N(R5), then said L group is not attached to 5-position of pyrazole;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
7. The compound according to any one of the claims 1 -4,
wherein,
A is isoxazole;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
8. The compound according to any one of the claims 1 -4,
wherein,
A is thiazole; or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
9. The compound according to claim 1 or claim 2,
Figure imgf000091_0001
wherein * indicates the point of attachment to L;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
10. The compound according to any one of the claims 1 , 2 and 9,
wherein,
A is
Figure imgf000091_0002
wherein * indicates the point of attachment to L;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
1 1 . The compound according to any one of the claims 1 , 2 and 9,
wherein,
A is
Figure imgf000092_0001
wherein * indicates the point of attachment to L;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
12. The compound according to claim 1 or claim 2,
wherein,
each of Xi , X2, X4 and X5 is C(R7); and X3 is C(R8);
with the provisos that:
(c) when A is pyrazole then L is a group other than *C(0)N(R5) or *N(R4)-C(0); and
(d) when A is pyrazole and L is *N(R )-C(0)-N(R5), then said L group is not attached to 5-position of pyrazole;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
13. The compound according to any one of the claims 1 -4 and 12,
wherein,
A is triazole, pyrrazole, isoxazole or thiazole;
each of Xi , X2, X and X5 is C(R7); and X3 is C(R8);
with the provisos that:
(c) when A is pyrazole then L is a group other than *C(0)N(R5) or *N(R4)-C(0); and
(d) when A is pyrazole and L is *N(R4)-C(0)-N(R5), then said L group is not attached to 5-position of pyrazole;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
14. The compound according to any one of the claims 1 -4, 12 and 13,
wherein,
A is triazole or pyrazole;
L is *N(R4)-C(0)-N(R5); wherein * indicates the point of attachment to A; and
each of Xi , X2, X4 and X5 is C(R7); and X3 is C(R8); with the proviso that when A is pyrazole the said L group is not attached to 5-position of pyrazole;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
15. The compound according to any one of the claims 1 -5 and 12-14,
wherein,
A is triazole;
L is *N(R4)-C(0)-N(R5); *C(0)N(R5), *CH(R6)-C(0)-N(R5); *CH(R6)N(R5) or *N(R4)- S(0)2 ; wherein * indicates the point of attachment to A;
each of Xi , X2, X4 and X5 is C(R7); and X3 is C(R8);
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
16. The compound according to any one of the claims 1 -4, 6 and 1 2-14,
wherein,
A is pyrazole;
L is *N(R4)-C(0)-N(R5); N(R4)-S(0)2 or *CH(R6)N(R5); wherein * indicates the point of attachment to A;
each of Xi , X2, X4 and X5 is C(R7); and X3 is C(R8);
with the proviso that when A is pyrazole and L is *N(R4)-C(0)-N(R5), then said L group is not attached to 5-position of pyrazole;
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
17. The compound according to any one of the claims 1 , 2, 9, 10 and 12,
wherein,
A is
Figure imgf000093_0001
wherein * indicates the point of attachment to L; L is *N(R4)-C(0)-N(R5); *C(0)N(R5); *N(R4)-S(0)2 or *NR4; wherein * indicates the point of attachment to A;
each of Xi , X2, X4 and X5 is C(R7); and X3 is C(R8);
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
18. The compound according to any one of the claims 1 , 2, 9, 1 1 and 12,
wherein,
A is
Figure imgf000094_0001
wherein * indicates the point of attachment to L;
L is *N(R4)-C(0)-N(R5); *C(0)N(R5); N(R4)-S(0)2 or *NR4; wherein * indicates the point of attachment to A;
each of Xi , X2, X4 and X5 is C(R7); and X3 is C(R8);
or an isotopic form, a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
19. The compound according to claim 1 or claim 2,
wherein,
one of X-i , X2, X4 or X5 is N, while the remaining are C(R7); X3 is N or C(Rs); or any two of X-i, X2, X4 or X5 are N, while the remaining are C(R7); X3 is N or C(R8); with the provisos that:
(c) when A is pyrazole then L is a group other than *C(0)N(R5) or *N(R )-C(0); and
(d) when A is pyrazole and L is *N(R4)-C(0)-N(R5), then said L group is not attached to 5-position of pyrazole;
or an isotopic form, stereoisomer or a tautomer or a pharmaceutically acceptable salt, a solvate, a polymorph, a prodrug, N-oxide or S-oxide thereof.
20. The compound according to any one of the claims 1 to 1 9, wherein said compound is selected from 1 -(5-Methyl-1 -(4-(trifluoromethyl)phenyl)-1 H-pyrazol-4-yl)-3-(2-(trifluoromethyl)-1 H- benzo[d]imidazol-6-yl)urea;
1 -(4-(4-(4-Chlorophenyl)-4-hydroxypiperidin-1 -yl)-3-cyanophenyl)-3-(5-methyl-1 -(4- (trifluoromethyl)phenyl)-1 H-pyrazol-4-yl)urea;
1 -(3-Cyano-4-(4-(2-hydroxyethyl)piperidin-1 -yl)phenyl)-3-(5-methyl-1 -(4-
(trifluoromethyl)phenyl)-1 H-pyrazol-4-yl)urea;
1 -(5-Methyl-1 -phenyl-1 H-pyrazol-4-yl)-3-(naphthalen-1 -yl)urea;
1 -(1 -(4-Chlorophenyl)-5-methyl-1 H-pyrazol-4-yl)-3-(3-cyano-4-(4-morpholino piperidin
1 -yl) phenyl) urea;
1 -(3-Cyano-4-(4-morpholinopiperidin-1 -yl) phenyl)-3-(5-methyl-1 -(4-(trifluoromethyl) phenyl)-1 H-pyrazol-4-yl) urea;
1 -(3-Cyano-4-(piperazin-1 -yl) phenyl)-3-(5-methyl-1 -(4-(trifluoromethyl) phenyl)-1 H- pyrazol-4-yl) urea;
1 -(5-Chloro-6-morpholinopyridin-3-yl)-3-(5-methyl-1 -(4-(trifluoromethyl) phenyl)-1 H- pyrazol-4-yl) urea;
1 - (3-Cyano-4-(4-(4-fluoro-3-(trifluoromethyl) benzoyl) piperazin-1 -yl) phenyl)-3-(5- methyl-1 -(4-(trifluoromethyl)phenyl)-1 H-pyrazol-4-yl)urea;
1 -(3-Cyano-4-(4-morpholinopiperidin-1 -yl) phenyl)-1 -methyl-3-(5-methyl-1 -(4- (trifluoromethyl) phenyl)-1 H-pyrazol-4-yl) urea;
1 -(1 -(4-Chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(4-morpholinopiperidin -1 - yl)phenyl)urea;
1 -(4-([1 ,4'-Bipiperidin]-1 '-yl)-3-cyanophenyl)-3-(1 -(4-chlorophenyl)-1 H-1 ,2,3-triazol-4- yl)urea;
2- (1 -(4-(3-(1 -(4-(Tert-butyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)ureido)-2-cyanophenyl) piperidin-4-yl)ethyl diethyl phosphate;
1 -(1 -(4-(Tert-butyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(4-(2-methoxyethyl) piperidin-1 -yl)phenyl)urea;
1 -(1 -(4-(Tert-butyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(4-(4-morpholinopiperidin-1 -yl)-2- (trifluoromethyl)phenyl)urea;
1 -(3-Cyano-4-(4-morpholinopiperidin-1 -yl)phenyl)-3-(1 -(4-morpholinophenyl)-1 H-1 ,2,3 triazol-4-yl)urea;
1 -(4-(1 ,1 ,1 ,3,3,3-Hexafluoro-2-hydroxypropan-2-yl)phenyl)-3-(1 -(4-isopropylphenyl) - 1 H-1 ,2,3-triazol-4-yl)urea; 1 -(1 -(4-Chlorophenyl)-5-methyl-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(4-morpholino piperidin-1 -yl)phenyl)urea;
3- (3-(1 -(4-Chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)ureido)-N-(4-hydroxy-3-methoxy benzyl)benzamide;
4- (1 -(2-Cyano-4-(3-(1 -(4-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)ureido) phenyl)piperidin-4-yl)butanoic acid;
1 -(3-Cyano-4-(4-(pyrimidin-2-yl)piperazin-1 -yl)phenyl)-3-(1 -(4-(trifluoromethyl) phenyl)- 1 H-1 ,2,3-triazol-4-yl)urea;
1 -(1 -(4-Chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(piperazin-1 -yl)phenyl) urea; 1 -(1 -(4-Chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(5-(4-morpholinopiperidin-1 -yl) pyridin-2- yl)urea;
1 -(1 -(4-Chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(4-cyano-5-(4-morpholinopiperidin -1 - yl)pyridin-2-yl)urea;
1 -(6-(4-Morpholinopiperidin-1 -yl)pyridin-3-yl)-3-(1 -(3-(trifluoromethyl)phenyl)-1 H-1 ,2,3- triazol-4-yl)urea;
1 -(5-(3-(1 -(4-(Tert-butyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)ureido)pyridin-2-yl)piperidine-4- carboxylic acid;
1 -(6-(4-(Pyrimidin-2-yl)piperazin-1 -yl)-5-(trifluoromethyl)pyridin-3-yl)-3-(1 -(3- (trifluoromethyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)urea;
1 -(5-Chloro-6-(4-morpholinopiperidin-1 -yl)pyridin-3-yl)-3-(1 -(4-chlorophenyl)-1 H-1 ,2,3- triazol-4-yl)urea;
1 -(1 -(4-(Tert-butyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(5-methyl-6-(4-morpholino piperidin- 1 -yl)pyridin-3-yl)urea;
1 -(1 -(4-(Tert-butyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(5-methyl-6-(4-(pyrimidin-2- yl)piperazin-1 -yl)pyridin-3-yl)urea;
1 -(1 -(4-(Tert-butyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(5-cyano-6-(4-(pyrimidin-2- yl)piperazin-1 -yl)pyridin-3-yl)urea;
1 -(6-(4-Cyano-4-phenylpiperidin-1 -yl)pyridin-3-yl)-3-(1 -(3-(trifluoromethyl) phenyl)-1 H- 1 ,2,3-triazol-4-yl)urea;
1 -(3-Cyano-4-(piperidin-1 -yl)phenyl)-3-(1 -(4-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazol-4- yl)urea;
1 -(3-Cyano-4-(2-oxa-6-azaspiro[3.3]heptan-6-yl)phenyl)-3-(1 -(4-(trifluoro
methyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)urea; 1 -(1 -(4-Chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(4-(cyclopropylmethyl) piperazin-1 -yl)phenyl)urea;
1 -(1 -(4-Chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(4-methoxypiperidin-1 - yl)phenyl)urea;
1 -(3-Cyano-4-(4-(3- ydroxyphenyl)piperazin-1 -yl)phenyl)-3-(1 -(p-tolyl)-l H-1 ,2,3-triazol- 4-yl)urea;
1 -(3-Cyano-4-(4-morpholinopiperidin-1 -yl)phenyl)-3-(1 -(3-(trifluoromethyl) phenyl)-1 H- 1 ,2,3-triazol-4-yl)urea;
1 -(1 -(4-Chlorophenyl)-1 H-1 ,2,3-triazol-4-yl)-3-(3-cyano-4-(4-hydroxypiperidin-1 - yl)phenyl)-1 ,3-dimethylurea;
1 -(4-(3-(1 -(4-(Tert-butyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)ureido)-2-cyanophenyl)-4-(2- hydroxypropan-2-yl)piperidin-1 -ium methanesulfonate;
1 -(3-Cyano-4-(piperazin-1 -yl)phenyl)-3-(1 -(4-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazol-4- yl)urea hydrochloride;
1 -(3-Cyano-4-(4-morpholinopiperidin-1 -yl)phenyl)-3-(3-(4-fluorophenyl)-5- methylisoxazol-4-yl)urea;
1 -(4-(Tert-butyl) phenyl)-N-(3-cyano-4-(4-(2-hydroxyethyl) piperidin-1 -yl) phenyl)-1 H-1 , 2, 3-triazole-4-carboxamide;
1 -(4-(Tert-butyl) phenyl)-N-(3-cyano-4-(4-hydroxypiperidin-1 -yl) phenyl)-1 H-1 , 2, 3- triazole-4-carboxamide;
1 -(4-Chlorophenyl)-N-(3-cyano-4-(4-morpholinopiperidin-1 -yl)phenyl)-1 H-1 ,2,3-triazole- 4-carboxamide;
1 -(4-Chlorophenyl)-N-(3-cyano-4-(4-morpholinopiperidin-1 -yl)phenyl)-1 H-1 ,2,3-triazole-
4- carboxamide hydrochloride;
1 -(4-Chlorophenyl)-N-(3-cyano-4-(4-(pyrimidin-2-yl)piperazin-1 -yl)phenyl)-1 H-1 ,2,3- triazole-4-carboxamide;
N-(3-Cyano-4-(4-morpholinopiperidin-1 -yl) phenyl)-5-pentyl-1 -(3-(trifluoromethyl) phenyl)-1 H-1 ,2,3-triazole-4-carboxamide;
N-(3-Cyano-4-(4-morpholinopiperidin-1 -yl)phenyl)-3-(4-fluorophenyl)-4-rnethylisoxazole-
5- carboxamide;
3-(4-Chlorophenyl)-N-(3-cyano-4-(4-morpholinopiperidin-1 -yl)phenyl)isoxazole-5- carboxamide;
N-(1 -(4-Chlorophenyl)-5-methyl-1 H-pyrazol-4-yl)-3-cyano-4-(4-morpholino piperidin-1 - yl)benzenesulfonamide; N-(4-(3-Methoxyphenyl)pyrimidin-2-yl)-5-phenylthiazol-2-arnine;
5-Phenyl-N-(4-(4-(trifluoromethoxy)phenyl)pyrimidin-2-yl)thiazol-2-amine;
N-(4-(2-Methoxyphenyl)pyrimidin-2-yl)-5-phenylthiazol-2-arnine;
5-((6-Fluorobenzo[d]thiazol-2-yl) amino)-2-(4-morpholinopiperidin-1 -yl) benzonitrile; 5-((4-(3, 5-Dimethoxyphenyl) pyrimidin-2-yl) amino)-2-(4-morpholinopiperidin-1 -yl) benzonitrile;
5-((4-(3, 5-Dimethoxyphenyl) pyrimidin-2-yl) amino)-2-(4-(hydroxymethyl) piperidin-1 -yl) benzonitrile;
4- (2-Methoxyphenyl)-N-(4-(4-(pyrimidin-2-yl)piperazin-1 -yl)-3-(trifluoromethyl) phenyl) pyrimidin-2-amine;
1 ,1 ,1 ,3,3,3-Hexafluoro-2-(4-((4-(3-methoxyphenyl)pyrimidin-2-yl)amino)phenyl) propan- 2-ol;
2-(4-Hydroxypiperidin-1 -yl)-5-((4-(3-methoxyphenyl)pyrimidin-2-yl)amino) benzonitrile;
5- ((4-Morpholinopyrimidin-2-yl)amino)-2-(4-(pyridin-2-yl)piperazin-1 -yl)benzonitrile 2-morpholino-5-((4-morpholinopyrimidin-2-yl)amino)benzonitrile;
5-((4-((2-Morpholinoethyl)amino)pyrimidin-2-yl)amino)-2-(4-morpholino piperidin-1 - yl)benzonitrile;
5-((2-Chloro-6-methoxyquinolin-4-yl) amino)-2-(4-morpholinopiperidin-1 -yl) benzonitrile;
2-(4-Morpholinopiperidin-1 -yl)-5-(quinolin-2-ylamino) benzonitrile;
5-((Cyano(1 -(3-(trifluoromethyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)methyl)amino)-2-(4-(2- hydroxypropan-2-yl)piperidin-1 -yl)benzonitrile;
5-((Cyano(1 -(3-(trifluoromethyl)phenyl)-1 H-1 ,2,3-t azol-4-yl)methyl)amino)-2-(4- (pyrimidin-2-yl)piperazin-1 -yl)benzonitrile;
2-Chloro-5-(((1 -(4-chlorophenyl)-5-methyl-1 H-pyrazol-4-yl)methyl)amino) benzonitrile; 5-(((1 -(4-Chlorophenyl)-5-methyl-1 H-pyrazol-4-yl)methyl)amino)-2-(4-morpholino piperidin-1 -yl)benzonitrile;
Diethyl (((4-chloro-3-cyanophenyl)amino)(1 -(4-chlorophenyl)-5-methyl-1 H-pyrazol-4- yl)methyl)phosphonate;
N-(4-(1 ,1 ,1 ,3,3,3-Hexafluoro-2-hydroxypropan-2-yl)phenyl)-2-hydroxy-2-(1 -(3- (trifluoromethyl)phenyl)-1 H-1 ,2,3-triazol-4-yl)acetamide; or
N-(3-Cyano-4-(4-methoxypiperidin-1 -yl)phenyl)-2-hydroxy-2-(1 -(3 (trifluoromethyl) phenyl)-1 H-1 ,2,3-triazol-4-yl)acetamide; or
a stereoisomer, a tautomer or a pharmaceutically acceptable salt thereof.
21 . A pharmaceutical composition comprising a therapeutically effective amount of a compound as defined in any one of the claims 1 -20, or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof; and at least one pharmaceutically acceptable carrier or excipient.
22. A method for the treatment of a disease or disorder mediated by IL-17 or TNF-a, comprising administering to a subject in need thereof; a therapeutically effective amount of a compound as defined in any one of the claims 1 -20, or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof.
23. The method according to claim 22, wherein the disease or disorder is selected from the group consisting of an autoimmune or inflammatory disease or disorder; and a metabolic disease or disorder.
24. The method according to claim 23, wherein the autoimmune or inflammatory disease or disorder is selected from the group consisting of inflammatory bowel disease, rheumatoid arthritis, juvenile rheumatoid arthritis, psoriatic arthritis, osteoarthritis, refractory rheumatoid arthritis, chronic non-rheumatoid arthritis, ankylosing spondylitis, osteoporosis/bone resorption, chronic graft-versus-host disease, acute graft-versus-host disease, multiple sclerosis, systemic lupus erythematosus, Celiac Sprue, idiopathic thrombocytopenic thrombotic purpura, myasthenia gravis, Sjogren's syndrome, scleroderma, asthma, bronchitis, epidermal hyperplasia, Crohn's disease, atherosclerosis, septic shock syndrome, coronary heart disease, vasculitis, ulcerative colitis, psoriasis, adult respiratory distress syndrome, myolitis, polymyolitis, dermatomyolitis, polyarteritis nodossa, Wegener's granulomatosis, arteritis, polymyalgia rheumatica, sarcoidosis, sclerosis, primary biliary sclerosis, sclerosing cholangitis, dermatitis, atopic dermatitis, Still's disease, chronic obstructive pulmonary disease, Guillain-Barre disease, Type I diabetes, Graves' disease, Addison's disease, Raynaud's phenomenon, autoimmune hepatitis, psoriatic epidermal hyperplasia and delayed type hypersensitivity in skin disorders.
25. The method according to claim 23, wherein the metabolic disease or disorder is selected from the group consisting of obesity, dyslipidemia, hyperlipidemia, hypertriglyceridemia, low adiponectin level, dyslipoproteinemia, impaired glucose tolerance, insulin resistance, increase in HbA1 c (glycosylated haemoglobin) level, reduced metabolic activity and Type II diabetes.
26. A compound according to any one of the claims 1 -20, or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof; for use as an IL-1 7 inhibitor.
27. A compound according to any one of the claims 1 -20, or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof; for use as a TNF-a inhibitor.
28. A compound as defined in any one of the claims 1 -20, or a stereoisomer, a tautomer or a pharmaceutically acceptable salt or a solvate thereof; for use in the treatment of a disease or a disorder mediated by IL-1 7 or TNF-a.
29. The compound for the use according to claim 28, wherein the disease or disorder is selected from the group consisting of an autoimmune or inflammatory disease or disorder; and a metabolic disease or disorder.
30. The compound for the use according to claim 29, wherein the autoimmune or inflammatory disease or disorder is selected from the group consisting of inflammatory bowel disease, rheumatoid arthritis, juvenile rheumatoid arthritis, psoriatic arthritis, osteoarthritis, refractory rheumatoid arthritis, chronic non-rheumatoid arthritis, ankylosing spondylitis, osteoporosis/bone resorption, chronic graft-versus-host disease, acute graft-versus-host disease, multiple sclerosis, systemic lupus erythematosus, Celiac Sprue, idiopathic thrombocytopenic thrombotic purpura, myasthenia gravis, Sjogren's syndrome, scleroderma, asthma, bronchitis, epidermal hyperplasia, Crohn's disease, atherosclerosis, septic shock syndrome, coronary heart disease, vasculitis, ulcerative colitis, psoriasis, adult respiratory distress syndrome, myolitis, polymyolitis, dermatomyolitis, polyarteritis nodossa, Wegener's granulomatosis, arteritis, polymyalgia rheumatica, sarcoidosis, sclerosis, primary biliary sclerosis, sclerosing cholangitis, dermatitis, atopic dermatitis, Still's disease, chronic obstructive pulmonary disease, Guillain-Barre disease, Type I diabetes, Graves' disease, Addison's disease, Raynaud's phenomenon, autoimmune hepatitis, psoriatic epidermal hyperplasia and delayed type hypersensitivity in skin disorders.
31 . The compound for the use according to claim 29, wherein the metabolic disease or disorder is selected from the group consisting of obesity, dyslipidemia, hyperlipidemia, hypertriglyceridemia, low adiponectin level, dyslipoproteinemia, impaired glucose tolerance, insulin resistance, increase in HbA1 c (glycosylated haemoglobin) level, reduced metabolic activity and Type II diabetes.
32. Use of a compound as defined in any one of the claims 1 -20 or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof; in combination with one further therapeutically active agent; for the treatment of a disease or a disorder mediated by IL-1 7 or TNF-a.
PCT/IB2014/061299 2013-05-09 2014-05-08 Heterocyclyl compounds and uses thereof WO2014181287A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361821287P 2013-05-09 2013-05-09
US61/821,287 2013-05-09

Publications (1)

Publication Number Publication Date
WO2014181287A1 true WO2014181287A1 (en) 2014-11-13

Family

ID=51866863

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2014/061299 WO2014181287A1 (en) 2013-05-09 2014-05-08 Heterocyclyl compounds and uses thereof

Country Status (1)

Country Link
WO (1) WO2014181287A1 (en)

Cited By (86)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9328096B2 (en) 2014-05-07 2016-05-03 Pfizer Inc. Tropomyosin-related kinase inhibitors
US9434719B2 (en) 2013-03-14 2016-09-06 Novartis Ag 3-pyrimidin-4-yl-oxazolidin-2-ones as inhibitors of mutant IDH
US9452998B2 (en) 2014-08-06 2016-09-27 Novartis Ag Protein kinase C inhibitors and methods of their use
WO2016202935A1 (en) 2015-06-19 2016-12-22 Bayer Pharma Aktiengesellschaft Glucose transport inhibitors
WO2017112853A1 (en) * 2015-12-22 2017-06-29 Proteostasis Therapeutics, Inc. Methods of treating pulmonary diseases and disorders
US9708272B2 (en) 2014-08-29 2017-07-18 Tes Pharma S.R.L. Inhibitors of α-amino-β-carboxymuconic acid semialdehyde decarboxylase
US9745292B2 (en) 2014-03-13 2017-08-29 Proteostasis Therapeutics, Inc. Compounds, compositions, and methods for increasing CFTR activity
US9782408B2 (en) 2014-10-06 2017-10-10 Vertex Pharmaceuticals Incorporated Modulators of cystic fibrosis transmembrane conductance regulator
US9790219B2 (en) 2014-03-13 2017-10-17 Proteostasis Therapeutics, Inc. Compounds, compositions, and methods for increasing CFTR activity
US9815842B2 (en) 2014-05-28 2017-11-14 Novartis Ag Pyrazolo pyrimidine derivatives and their use as MALT1 inhibitors
US9932325B2 (en) 2016-06-16 2018-04-03 Denali Therapeutics Inc. Compounds, compositions, and methods
WO2018069222A1 (en) 2016-10-14 2018-04-19 Bayer Aktiengesellschaft Substituted 6-(1h-pyrazol-1-yl)pyrimidin-4-amine derivatives and uses thereof
US9951069B1 (en) 2017-01-11 2018-04-24 Rodin Therapeutics, Inc. Bicyclic inhibitors of histone deacetylase
US10030004B2 (en) 2014-01-01 2018-07-24 Medivation Technologies Llc Compounds and methods of use
CN108358869A (en) * 2018-03-27 2018-08-03 河北师范大学 A kind of N- benzothiazolyls benzsulfamide analog derivative, Preparation method and use
US10174014B2 (en) 2014-06-19 2019-01-08 Proteostasis Therapeutics, Inc. Compounds, compositions, and methods for increasing CFTR activity
US10220027B2 (en) 2011-07-13 2019-03-05 Gilead Sciences, Inc. FXR (NR1H4) binding and activity modulating compounds
US10273227B2 (en) 2017-04-27 2019-04-30 Astrazeneca Ab C5-anilinoquinazoline compounds and their use in treating cancer
US10308644B2 (en) 2016-12-22 2019-06-04 Incyte Corporation Heterocyclic compounds as immunomodulators
US10329286B2 (en) 2016-06-13 2019-06-25 Gilead Sciences, Inc. FXR (NR1H4) modulating compounds
US10344023B2 (en) 2014-12-23 2019-07-09 Proteostasis Therapeutics, Inc. Derivatives of 3-heteroarylisoxazol-5-carboxylic amide useful for the treatment of inter alia cystic fibrosis
WO2019138017A1 (en) * 2018-01-15 2019-07-18 Ucb Biopharma Sprl Fused imidazole derivatives as il-17 modulators
US10392378B2 (en) 2014-12-23 2019-08-27 Proteostasis Therapeutics, Inc. Derivatives of 5-phenyl- or 5-heteroarylathiazol-2-carboxylic amide useful for the treatment of inter alia cystic fibrosis
US10392372B2 (en) 2014-12-23 2019-08-27 Proteostasis Therapeutics, Inc. Compounds, compositions, and methods for increasing CFTR activity
CN110267964A (en) * 2016-12-13 2019-09-20 百时美施贵宝公司 As IL-12, IL-23 and/or the heteroaryl phosphine oxide compound of IFN α response regulation agent replaced through alkylamide
US10421756B2 (en) 2015-07-06 2019-09-24 Rodin Therapeutics, Inc. Heterobicyclic N-aminophenyl-amides as inhibitors of histone deacetylase
US10421730B2 (en) 2016-06-13 2019-09-24 Gilead Sciences, Inc. FXR (NR1H4) modulating compounds
WO2019190117A1 (en) * 2018-03-28 2019-10-03 한림제약(주) 2-cyanopyrimidin-4-yl carbamate or urea derivative or salt thereof, and pharmaceutical composition including same
US10450273B2 (en) 2016-08-29 2019-10-22 Novartis Ag N-(pyridin-2-yl)pyridine-sulfonamide derivatives and their use in the treatment of disease
US10548878B2 (en) 2015-07-24 2020-02-04 Proteostasis Therapeutics, Inc. Compounds, compositions, and methods of increasing CFTR activity
US10550106B2 (en) 2015-10-06 2020-02-04 Proteostasis Therapeutics, Inc. Compounds, compositions, and methods for modulating CFTR
US10570096B2 (en) 2015-11-26 2020-02-25 Novartis Ag Diamino pyridine derivatives
US10570115B2 (en) 2016-09-30 2020-02-25 Vertex Pharmaceuticals Incorporated Modulator of cystic fibrosis transmembrane conductance regulator, pharmaceutical compositions, methods of treatment, and process for making the modulator
US10618916B2 (en) 2018-05-11 2020-04-14 Incyte Corporation Heterocyclic compounds as immunomodulators
US10654829B2 (en) 2017-10-19 2020-05-19 Vertex Pharmaceuticals Incorporated Crystalline forms and compositions of CFTR modulators
US10662207B2 (en) 2016-04-07 2020-05-26 Proteostasis Therapeutics, Inc. Compounds, compositions, and methods for modulating CFTR
US10669271B2 (en) 2018-03-30 2020-06-02 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2020120141A1 (en) * 2018-12-11 2020-06-18 UCB Biopharma SRL Functionalised amine derivatives as il-17 modulators
US10738030B2 (en) 2016-03-31 2020-08-11 Vertex Pharmaceuticals Incorporated Modulators of cystic fibrosis transmembrane conductance regulator
US10738011B2 (en) 2014-12-23 2020-08-11 Proteostasis Therapeutics, Inc. Derivatives of 5-(hetero)arylpyrazol-3-carboxylic amide or 1-(hetero)aryltriazol-4-carboxylic amide useful for the treatment of inter alia cystic fibrosis
US10751417B2 (en) 2017-04-20 2020-08-25 Novartis Ag Sustained release delivery systems comprising traceless linkers
US10793547B2 (en) 2016-12-09 2020-10-06 Vertex Pharmaceuticals Incorporated Modulator of the cystic fibrosis transmembrane conductance regulator, pharmaceutical compositions, methods of treatment, and process for making the modulator
US10793544B2 (en) * 2016-09-01 2020-10-06 The Board Of Regents Of The University Of Texas System Disubstituted and trisubstituted 1,2,3-triazoles as Wnt inhibitors
US10793565B2 (en) 2016-12-22 2020-10-06 Incyte Corporation Heterocyclic compounds as immunomodulators
US10806785B2 (en) 2016-12-22 2020-10-20 Incyte Corporation Immunomodulator compounds and methods of use
US10899751B2 (en) 2016-06-21 2021-01-26 Proteostasis Therapeutics, Inc. Compounds, compositions, and methods for increasing CFTR activity
US10906888B2 (en) 2016-07-14 2021-02-02 Pfizer Inc. Pyrimidine carboxamides as inhibitors of Vanin-1 enzyme
US10919902B2 (en) 2015-07-06 2021-02-16 Alkermes, Inc. Hetero-halo inhibitors of histone deacetylase
WO2021066559A1 (en) * 2019-10-02 2021-04-08 Kainos Medicine, Inc. N-(1h-imidazol-2-yl)benzamide compound and pharmaceutical composition comprising the same as active ingredient
US11028080B2 (en) 2016-03-11 2021-06-08 Denali Therapeutics Inc. Substituted pyrimidines as LRKK2 inhibitors
WO2021214020A1 (en) 2020-04-24 2021-10-28 Bayer Aktiengesellschaft Substituted aminothiazoles as dgkzeta inhibitors for immune activation
US11168068B2 (en) 2016-07-18 2021-11-09 Janssen Pharmaceutica Nv Tau PET imaging ligands
US11179367B2 (en) 2018-02-05 2021-11-23 Vertex Pharmaceuticals Incorporated Pharmaceutical compositions for treating cystic fibrosis
WO2021255174A1 (en) * 2020-06-18 2021-12-23 Leo Pharma A/S Small molecule modulators of il-17
US11214565B2 (en) 2015-11-20 2022-01-04 Denali Therapeutics Inc. Compound, compositions, and methods
US11225473B2 (en) 2019-01-15 2022-01-18 Gilead Sciences, Inc. FXR (NR1H4) modulating compounds
US11225475B2 (en) 2017-08-07 2022-01-18 Alkermes, Inc. Substituted pyridines as inhibitors of histone deacetylase
US11253509B2 (en) 2017-06-08 2022-02-22 Vertex Pharmaceuticals Incorporated Methods of treatment for cystic fibrosis
CN114302876A (en) * 2019-09-06 2022-04-08 英夫拉索姆有限公司 NLRP3 inhibitor
US11299467B2 (en) 2017-07-21 2022-04-12 Antabio Sas Chemical compounds
US11389541B2 (en) 2018-10-03 2022-07-19 Novartis Ag Sustained delivery of angiopoetin-like 3 polypeptides
US11401279B2 (en) 2019-09-30 2022-08-02 Incyte Corporation Pyrido[3,2-d]pyrimidine compounds as immunomodulators
US11407749B2 (en) 2015-10-19 2022-08-09 Incyte Corporation Heterocyclic compounds as immunomodulators
US11414439B2 (en) 2018-04-13 2022-08-16 Vertex Pharmaceuticals Incorporated Modulators of cystic fibrosis transmembrane conductance regulator, pharmaceutical compositions, methods of treatment, and process for making the modulator
US11434201B2 (en) 2017-08-02 2022-09-06 Vertex Pharmaceuticals Incorporated Processes for preparing pyrrolidine compounds
US11465981B2 (en) 2016-12-22 2022-10-11 Incyte Corporation Heterocyclic compounds as immunomodulators
US11465985B2 (en) 2017-12-08 2022-10-11 Vertex Pharmaceuticals Incorporated Processes for making modulators of cystic fibrosis transmembrane conductance regulator
US11517564B2 (en) 2017-07-17 2022-12-06 Vertex Pharmaceuticals Incorporated Methods of treatment for cystic fibrosis
US11524005B2 (en) 2019-02-19 2022-12-13 Gilead Sciences, Inc. Solid forms of FXR agonists
US11529350B2 (en) 2019-07-03 2022-12-20 Sumitomo Pharma Oncology, Inc. Tyrosine kinase non-receptor 1 (TNK1) inhibitors and uses thereof
US11535615B2 (en) 2015-12-22 2022-12-27 Incyte Corporation Heterocyclic compounds as immunomodulators
RU2788740C2 (en) * 2018-03-28 2023-01-24 Ханлим Фармасьютикал Ко., Лтд. 2-cyanopyrimidine-4-yl carbamate, or a urea derivative, or its salt and a pharmaceutical composition containing them
US11572366B2 (en) 2015-11-19 2023-02-07 Incyte Corporation Heterocyclic compounds as immunomodulators
US11608337B2 (en) 2016-05-06 2023-03-21 Incyte Corporation Heterocyclic compounds as immunomodulators
US11613536B2 (en) 2016-08-29 2023-03-28 Incyte Corporation Heterocyclic compounds as immunomodulators
US11673883B2 (en) 2016-05-26 2023-06-13 Incyte Corporation Heterocyclic compounds as immunomodulators
US11691979B2 (en) 2020-04-30 2023-07-04 Janssen Pharmaceutica Nv Imidazopyridazines as modulators of IL-17
US11718605B2 (en) 2016-07-14 2023-08-08 Incyte Corporation Heterocyclic compounds as immunomodulators
US11753406B2 (en) 2019-08-09 2023-09-12 Incyte Corporation Salts of a PD-1/PD-L1 inhibitor
US11760756B2 (en) 2020-11-06 2023-09-19 Incyte Corporation Crystalline form of a PD-1/PD-L1 inhibitor
US11780836B2 (en) 2020-11-06 2023-10-10 Incyte Corporation Process of preparing a PD-1/PD-L1 inhibitor
US11833150B2 (en) 2017-03-28 2023-12-05 Gilead Sciences, Inc. Methods of treating liver disease
US11866451B2 (en) 2019-11-11 2024-01-09 Incyte Corporation Salts and crystalline forms of a PD-1/PD-L1 inhibitor
US11866434B2 (en) 2020-11-06 2024-01-09 Incyte Corporation Process for making a PD-1/PD-L1 inhibitor and salts and crystalline forms thereof
US11873309B2 (en) 2016-06-20 2024-01-16 Incyte Corporation Heterocyclic compounds as immunomodulators
US11932648B2 (en) 2021-06-28 2024-03-19 Blueprint Medicines Corporation CDK2 inhibitors

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000025780A1 (en) * 1998-10-29 2000-05-11 Bristol-Myers Squibb Company Compounds derived from an amine nucleus that are inhibitors of impdh enzyme
WO2004052280A2 (en) * 2002-12-10 2004-06-24 Imclone Systems Incorporated Anti-angiogenic compounds and their use in cancer treatment
WO2005000298A2 (en) * 2003-06-03 2005-01-06 Novartis Ag 5-membered heterocycle-based p-38 inhibitors
WO2006024034A1 (en) * 2004-08-25 2006-03-02 Targegen, Inc. Heterocyclic compounds and methods of use
WO2006124692A2 (en) * 2005-05-18 2006-11-23 Wyeth 3-cyanoquinoline inhibitors of tpl2 kinase and methods of making and using the same
WO2007061923A2 (en) * 2005-11-18 2007-05-31 Takeda San Diego, Inc. Glucokinase activators

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000025780A1 (en) * 1998-10-29 2000-05-11 Bristol-Myers Squibb Company Compounds derived from an amine nucleus that are inhibitors of impdh enzyme
WO2004052280A2 (en) * 2002-12-10 2004-06-24 Imclone Systems Incorporated Anti-angiogenic compounds and their use in cancer treatment
WO2005000298A2 (en) * 2003-06-03 2005-01-06 Novartis Ag 5-membered heterocycle-based p-38 inhibitors
WO2006024034A1 (en) * 2004-08-25 2006-03-02 Targegen, Inc. Heterocyclic compounds and methods of use
WO2006124692A2 (en) * 2005-05-18 2006-11-23 Wyeth 3-cyanoquinoline inhibitors of tpl2 kinase and methods of making and using the same
WO2007061923A2 (en) * 2005-11-18 2007-05-31 Takeda San Diego, Inc. Glucokinase activators

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 915361-49-8 *

Cited By (156)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10220027B2 (en) 2011-07-13 2019-03-05 Gilead Sciences, Inc. FXR (NR1H4) binding and activity modulating compounds
US10485795B2 (en) 2011-07-13 2019-11-26 Gilead Sciences, Inc. FXR (NR1H4) binding and activity modulating compounds
US9434719B2 (en) 2013-03-14 2016-09-06 Novartis Ag 3-pyrimidin-4-yl-oxazolidin-2-ones as inhibitors of mutant IDH
US10112931B2 (en) 2013-03-14 2018-10-30 Novartis Ag 3-pyrimidin-4-yl-oxazolidin-2-ones as inhibitors of mutant IDH
US9688672B2 (en) 2013-03-14 2017-06-27 Novartis Ag 3-pyrimidin-4-yl-oxazolidin-2-ones as inhibitors of mutant IDH
US11702401B2 (en) 2014-01-01 2023-07-18 Medivation Technologies Llc Compounds and methods of use
US10501436B2 (en) 2014-01-01 2019-12-10 Medivation Technologies Llc Compounds and methods of use
US11053216B2 (en) 2014-01-01 2021-07-06 Medivation Technologies Llc Compounds and methods of use
US10030004B2 (en) 2014-01-01 2018-07-24 Medivation Technologies Llc Compounds and methods of use
US9790219B2 (en) 2014-03-13 2017-10-17 Proteostasis Therapeutics, Inc. Compounds, compositions, and methods for increasing CFTR activity
US9745292B2 (en) 2014-03-13 2017-08-29 Proteostasis Therapeutics, Inc. Compounds, compositions, and methods for increasing CFTR activity
US10017503B2 (en) 2014-03-13 2018-07-10 Proteostasis Therapeutics, Inc. Compounds, compositions, and methods for increasing CFTR activity
US9328096B2 (en) 2014-05-07 2016-05-03 Pfizer Inc. Tropomyosin-related kinase inhibitors
US9815842B2 (en) 2014-05-28 2017-11-14 Novartis Ag Pyrazolo pyrimidine derivatives and their use as MALT1 inhibitors
US10442808B2 (en) 2014-05-28 2019-10-15 Novartis Ag Pyrazolo pyrimidine derivatives and their use as MALT1 inhbitors
US10738040B2 (en) 2014-06-19 2020-08-11 Proteostasis Therapeutics, Inc. Compounds, compositions, and methods for increasing CFTR activity
US10174014B2 (en) 2014-06-19 2019-01-08 Proteostasis Therapeutics, Inc. Compounds, compositions, and methods for increasing CFTR activity
US10508101B2 (en) 2014-08-06 2019-12-17 Novartis Ag Protein kinase C inhibitors and methods of their use
US9845309B2 (en) 2014-08-06 2017-12-19 Novartis Ag Protein kinase C inhibitors and methods of their use
US11505541B2 (en) 2014-08-06 2022-11-22 Novartis Ag Protein kinase C inhibitors and methods of their use
US9452998B2 (en) 2014-08-06 2016-09-27 Novartis Ag Protein kinase C inhibitors and methods of their use
US11059804B2 (en) 2014-08-06 2021-07-13 Novartis Ag Protein kinase C inhibitors and methods of their use
US11254644B2 (en) 2014-08-29 2022-02-22 Tes Pharma S.R.L. Inhibitors of alpha-amino-beta-carboxymuconic acid semialdehyde decarboxylase
US10513499B2 (en) 2014-08-29 2019-12-24 Tes Pharma S.R.L. Inhibitors of alpha-amino-beta-carboxymuconic acid semialdehyde decarboxylase
US9708272B2 (en) 2014-08-29 2017-07-18 Tes Pharma S.R.L. Inhibitors of α-amino-β-carboxymuconic acid semialdehyde decarboxylase
US9782408B2 (en) 2014-10-06 2017-10-10 Vertex Pharmaceuticals Incorporated Modulators of cystic fibrosis transmembrane conductance regulator
US10758534B2 (en) 2014-10-06 2020-09-01 Vertex Pharmaceuticals Incorporated Modulators of cystic fibrosis transmembrane conductance regulator
US11426407B2 (en) 2014-10-06 2022-08-30 Vertex Pharmaceuticals Incorporated Modulators of cystic fibrosis transmembrane conductance regulator
US10258624B2 (en) 2014-10-06 2019-04-16 Vertex Pharmaceuticals Incorporated Modulators of cystic fibrosis transmembrane conductance regulator
US10392378B2 (en) 2014-12-23 2019-08-27 Proteostasis Therapeutics, Inc. Derivatives of 5-phenyl- or 5-heteroarylathiazol-2-carboxylic amide useful for the treatment of inter alia cystic fibrosis
US10392372B2 (en) 2014-12-23 2019-08-27 Proteostasis Therapeutics, Inc. Compounds, compositions, and methods for increasing CFTR activity
US10344023B2 (en) 2014-12-23 2019-07-09 Proteostasis Therapeutics, Inc. Derivatives of 3-heteroarylisoxazol-5-carboxylic amide useful for the treatment of inter alia cystic fibrosis
US11098035B2 (en) 2014-12-23 2021-08-24 Proteostasis Therapeutics, Inc. Compounds, compositions, and methods for increasing CFTR activity
US10738011B2 (en) 2014-12-23 2020-08-11 Proteostasis Therapeutics, Inc. Derivatives of 5-(hetero)arylpyrazol-3-carboxylic amide or 1-(hetero)aryltriazol-4-carboxylic amide useful for the treatment of inter alia cystic fibrosis
WO2016202935A1 (en) 2015-06-19 2016-12-22 Bayer Pharma Aktiengesellschaft Glucose transport inhibitors
US11858939B2 (en) 2015-07-06 2024-01-02 Alkermes, Inc. Hetero-halo inhibitors of histone deacetylase
US10421756B2 (en) 2015-07-06 2019-09-24 Rodin Therapeutics, Inc. Heterobicyclic N-aminophenyl-amides as inhibitors of histone deacetylase
US10919902B2 (en) 2015-07-06 2021-02-16 Alkermes, Inc. Hetero-halo inhibitors of histone deacetylase
US10548878B2 (en) 2015-07-24 2020-02-04 Proteostasis Therapeutics, Inc. Compounds, compositions, and methods of increasing CFTR activity
US11083709B2 (en) 2015-07-24 2021-08-10 Proteostasis Therapeutics, Inc. Compounds, compositions, and methods of increasing CFTR activity
US10550106B2 (en) 2015-10-06 2020-02-04 Proteostasis Therapeutics, Inc. Compounds, compositions, and methods for modulating CFTR
US11136313B2 (en) 2015-10-06 2021-10-05 Proteostasis Therapeutics, Inc. Compounds, compositions, and methods for modulating CFTR
US11407749B2 (en) 2015-10-19 2022-08-09 Incyte Corporation Heterocyclic compounds as immunomodulators
US11572366B2 (en) 2015-11-19 2023-02-07 Incyte Corporation Heterocyclic compounds as immunomodulators
US11214565B2 (en) 2015-11-20 2022-01-04 Denali Therapeutics Inc. Compound, compositions, and methods
US11274080B2 (en) 2015-11-26 2022-03-15 Novartis Ag Diamino pyridine derivatives
US10570096B2 (en) 2015-11-26 2020-02-25 Novartis Ag Diamino pyridine derivatives
US11866435B2 (en) 2015-12-22 2024-01-09 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2017112853A1 (en) * 2015-12-22 2017-06-29 Proteostasis Therapeutics, Inc. Methods of treating pulmonary diseases and disorders
US11535615B2 (en) 2015-12-22 2022-12-27 Incyte Corporation Heterocyclic compounds as immunomodulators
US11840529B2 (en) 2016-03-11 2023-12-12 Denali Therapeutics Inc. Substituted pyrimidines as LRKK2 inhibitors
US11028080B2 (en) 2016-03-11 2021-06-08 Denali Therapeutics Inc. Substituted pyrimidines as LRKK2 inhibitors
US10738030B2 (en) 2016-03-31 2020-08-11 Vertex Pharmaceuticals Incorporated Modulators of cystic fibrosis transmembrane conductance regulator
US10662207B2 (en) 2016-04-07 2020-05-26 Proteostasis Therapeutics, Inc. Compounds, compositions, and methods for modulating CFTR
US11248010B2 (en) 2016-04-07 2022-02-15 Proteostasis Therapeutics, Inc. Compounds, compositions, and methods for modulating CFTR
US11608337B2 (en) 2016-05-06 2023-03-21 Incyte Corporation Heterocyclic compounds as immunomodulators
US11673883B2 (en) 2016-05-26 2023-06-13 Incyte Corporation Heterocyclic compounds as immunomodulators
US10981881B2 (en) 2016-06-13 2021-04-20 Gilead Sciences, Inc. FXR (NR1H4) modulating compounds
US10774054B2 (en) 2016-06-13 2020-09-15 Gilead Sciences, Inc. FXR (NR1H4) modulating compounds
US11247986B2 (en) 2016-06-13 2022-02-15 Gilead Sciences, Inc. FXR (NR1H4) modulating compounds
US10329286B2 (en) 2016-06-13 2019-06-25 Gilead Sciences, Inc. FXR (NR1H4) modulating compounds
US11739065B2 (en) 2016-06-13 2023-08-29 Gilead Sciences, Inc. FXR (NR1H4) modulating compounds
US10421730B2 (en) 2016-06-13 2019-09-24 Gilead Sciences, Inc. FXR (NR1H4) modulating compounds
US9932325B2 (en) 2016-06-16 2018-04-03 Denali Therapeutics Inc. Compounds, compositions, and methods
US11111235B2 (en) 2016-06-16 2021-09-07 Denali Therapeutics Inc. Compounds, compositions, and methods
US11834439B2 (en) 2016-06-16 2023-12-05 Denali Therapeutics Inc. Compounds, compositions, and methods
US10590114B2 (en) 2016-06-16 2020-03-17 Denali Therapautics Inc. Compounds, compositions, and methods
US11591316B2 (en) 2016-06-16 2023-02-28 Denali Therapeutics Inc. Compounds, compositions, and methods
US11873309B2 (en) 2016-06-20 2024-01-16 Incyte Corporation Heterocyclic compounds as immunomodulators
US10899751B2 (en) 2016-06-21 2021-01-26 Proteostasis Therapeutics, Inc. Compounds, compositions, and methods for increasing CFTR activity
US10906888B2 (en) 2016-07-14 2021-02-02 Pfizer Inc. Pyrimidine carboxamides as inhibitors of Vanin-1 enzyme
US11718605B2 (en) 2016-07-14 2023-08-08 Incyte Corporation Heterocyclic compounds as immunomodulators
US11168068B2 (en) 2016-07-18 2021-11-09 Janssen Pharmaceutica Nv Tau PET imaging ligands
US10450273B2 (en) 2016-08-29 2019-10-22 Novartis Ag N-(pyridin-2-yl)pyridine-sulfonamide derivatives and their use in the treatment of disease
US11613536B2 (en) 2016-08-29 2023-03-28 Incyte Corporation Heterocyclic compounds as immunomodulators
US11066369B2 (en) 2016-08-29 2021-07-20 Novartis Ag N-(pyridin-2-yl)pyridine-sulfonamide derivatives and their use in the treatment of disease
US10793544B2 (en) * 2016-09-01 2020-10-06 The Board Of Regents Of The University Of Texas System Disubstituted and trisubstituted 1,2,3-triazoles as Wnt inhibitors
US11186566B2 (en) 2016-09-30 2021-11-30 Vertex Pharmaceuticals Incorporated Modulator of cystic fibrosis transmembrane conductance regulator, pharmaceutical compositions, methods of treatment, and process for making the modulator
US10570115B2 (en) 2016-09-30 2020-02-25 Vertex Pharmaceuticals Incorporated Modulator of cystic fibrosis transmembrane conductance regulator, pharmaceutical compositions, methods of treatment, and process for making the modulator
US11208400B2 (en) 2016-10-14 2021-12-28 Bayer Aktiengesellschaft Substituted 6-(1H-pyrazol-1-yl)pyrimidin-4-amine derivatives and uses thereof
WO2018069222A1 (en) 2016-10-14 2018-04-19 Bayer Aktiengesellschaft Substituted 6-(1h-pyrazol-1-yl)pyrimidin-4-amine derivatives and uses thereof
US10793547B2 (en) 2016-12-09 2020-10-06 Vertex Pharmaceuticals Incorporated Modulator of the cystic fibrosis transmembrane conductance regulator, pharmaceutical compositions, methods of treatment, and process for making the modulator
US11453655B2 (en) 2016-12-09 2022-09-27 Vertex Pharmaceuticals Incorporated Modulator of the cystic fibrosis transmembrane conductance regulator, pharmaceutical compositions, methods of treatment, and process for making the modulator
CN110267964A (en) * 2016-12-13 2019-09-20 百时美施贵宝公司 As IL-12, IL-23 and/or the heteroaryl phosphine oxide compound of IFN α response regulation agent replaced through alkylamide
CN110267964B (en) * 2016-12-13 2022-05-03 百时美施贵宝公司 Substituted heteroaryl phosphine oxide compounds as IL-12, IL-23 and/or IFN alpha response modulators
US10800768B2 (en) 2016-12-22 2020-10-13 Incyte Corporation Heterocyclic compounds as immunomodulators
US11787793B2 (en) 2016-12-22 2023-10-17 Incyte Corporation Heterocyclic compounds as immunomodulators
US11339149B2 (en) 2016-12-22 2022-05-24 Incyte Corporation Heterocyclic compounds as immunomodulators
US10308644B2 (en) 2016-12-22 2019-06-04 Incyte Corporation Heterocyclic compounds as immunomodulators
US10806785B2 (en) 2016-12-22 2020-10-20 Incyte Corporation Immunomodulator compounds and methods of use
US11566026B2 (en) 2016-12-22 2023-01-31 Incyte Corporation Heterocyclic compounds as immunomodulators
US11465981B2 (en) 2016-12-22 2022-10-11 Incyte Corporation Heterocyclic compounds as immunomodulators
US10793565B2 (en) 2016-12-22 2020-10-06 Incyte Corporation Heterocyclic compounds as immunomodulators
US10519149B2 (en) 2017-01-11 2019-12-31 Rodin Therapeutics, Inc. Bicyclic inhibitors of histone deacetylase
US10793567B2 (en) 2017-01-11 2020-10-06 Rodin Therapeutics, Inc. Bicyclic inhibitors of histone deacetylase
US9951069B1 (en) 2017-01-11 2018-04-24 Rodin Therapeutics, Inc. Bicyclic inhibitors of histone deacetylase
US10696673B2 (en) 2017-01-11 2020-06-30 Rodin Therapeutics, Inc. Bicyclic inhibitors of histone deacetylase
US11225479B2 (en) 2017-01-11 2022-01-18 Alkermes, Inc. Bicyclic inhibitors of histone deacetylase
US11987580B2 (en) 2017-01-11 2024-05-21 Alkermes, Inc. Bicyclic inhibitors of histone deacetylase
US11286256B2 (en) 2017-01-11 2022-03-29 Alkermes, Inc. Bicyclic inhibitors of histone deacetylase
US11833150B2 (en) 2017-03-28 2023-12-05 Gilead Sciences, Inc. Methods of treating liver disease
US10751417B2 (en) 2017-04-20 2020-08-25 Novartis Ag Sustained release delivery systems comprising traceless linkers
US10829479B2 (en) 2017-04-27 2020-11-10 Astrazeneca Ab C5-anilinoquinazoline compounds and their use in treating cancer
US10273227B2 (en) 2017-04-27 2019-04-30 Astrazeneca Ab C5-anilinoquinazoline compounds and their use in treating cancer
US11253509B2 (en) 2017-06-08 2022-02-22 Vertex Pharmaceuticals Incorporated Methods of treatment for cystic fibrosis
US11517564B2 (en) 2017-07-17 2022-12-06 Vertex Pharmaceuticals Incorporated Methods of treatment for cystic fibrosis
US11299467B2 (en) 2017-07-21 2022-04-12 Antabio Sas Chemical compounds
US11434201B2 (en) 2017-08-02 2022-09-06 Vertex Pharmaceuticals Incorporated Processes for preparing pyrrolidine compounds
US11225475B2 (en) 2017-08-07 2022-01-18 Alkermes, Inc. Substituted pyridines as inhibitors of histone deacetylase
US11912702B2 (en) 2017-08-07 2024-02-27 Alkermes, Inc. Substituted pyridines as inhibitors of histone deacetylase
US11155533B2 (en) 2017-10-19 2021-10-26 Vertex Pharmaceuticals Incorporated Crystalline forms and compositions of CFTR modulators
US10654829B2 (en) 2017-10-19 2020-05-19 Vertex Pharmaceuticals Incorporated Crystalline forms and compositions of CFTR modulators
US11465985B2 (en) 2017-12-08 2022-10-11 Vertex Pharmaceuticals Incorporated Processes for making modulators of cystic fibrosis transmembrane conductance regulator
WO2019138017A1 (en) * 2018-01-15 2019-07-18 Ucb Biopharma Sprl Fused imidazole derivatives as il-17 modulators
US11472794B2 (en) 2018-01-15 2022-10-18 UCB Biopharma SRL Fused imidazole derivatives as IL-17 modulators
CN111601799A (en) * 2018-01-15 2020-08-28 Ucb生物制药有限责任公司 Fused imidazole derivatives as Il-17 modulators
JP2021510707A (en) * 2018-01-15 2021-04-30 ユーシービー バイオファルマ エスアールエル Condensed imidazole derivative as IL-17 modulator
JP7271557B2 (en) 2018-01-15 2023-05-11 ユーシービー バイオファルマ エスアールエル Fused Imidazole Derivatives as IL-17 Modulators
US11179367B2 (en) 2018-02-05 2021-11-23 Vertex Pharmaceuticals Incorporated Pharmaceutical compositions for treating cystic fibrosis
CN108358869A (en) * 2018-03-27 2018-08-03 河北师范大学 A kind of N- benzothiazolyls benzsulfamide analog derivative, Preparation method and use
CN108358869B (en) * 2018-03-27 2019-09-10 河北师范大学 A kind of N- benzothiazolyl benzsulfamide analog derivative, Preparation method and use
RU2788740C2 (en) * 2018-03-28 2023-01-24 Ханлим Фармасьютикал Ко., Лтд. 2-cyanopyrimidine-4-yl carbamate, or a urea derivative, or its salt and a pharmaceutical composition containing them
WO2019190117A1 (en) * 2018-03-28 2019-10-03 한림제약(주) 2-cyanopyrimidin-4-yl carbamate or urea derivative or salt thereof, and pharmaceutical composition including same
US11299478B2 (en) 2018-03-28 2022-04-12 Hanlim Pharmaceutical Co., Ltd. 2-cyanopyrimidin-4-yl carbamate or urea derivative or salt thereof, and pharmaceutical composition including same
JP2021519759A (en) * 2018-03-28 2021-08-12 ハンリム ファーマシューティカル カンパニー リミテッド 2-Cyanopyrimidine-4-ylcarbamate derivative or urea derivative or salt thereof and a pharmaceutical composition containing the same.
JP7271566B2 (en) 2018-03-28 2023-05-11 ハンリム ファーマシューティカル カンパニー リミテッド 2-cyanopyrimidin-4-yl carbamate derivative or urea derivative or salt thereof and pharmaceutical composition containing same
US11124511B2 (en) 2018-03-30 2021-09-21 Incyte Corporation Heterocyclic compounds as immunomodulators
US10669271B2 (en) 2018-03-30 2020-06-02 Incyte Corporation Heterocyclic compounds as immunomodulators
US11414439B2 (en) 2018-04-13 2022-08-16 Vertex Pharmaceuticals Incorporated Modulators of cystic fibrosis transmembrane conductance regulator, pharmaceutical compositions, methods of treatment, and process for making the modulator
US10618916B2 (en) 2018-05-11 2020-04-14 Incyte Corporation Heterocyclic compounds as immunomodulators
US10906920B2 (en) 2018-05-11 2021-02-02 Incyte Corporation Heterocyclic compounds as immunomodulators
US11414433B2 (en) 2018-05-11 2022-08-16 Incyte Corporation Heterocyclic compounds as immunomodulators
US11389541B2 (en) 2018-10-03 2022-07-19 Novartis Ag Sustained delivery of angiopoetin-like 3 polypeptides
CN113260418A (en) * 2018-12-11 2021-08-13 Ucb生物制药有限责任公司 Functionalized amine derivatives as IL-17 modulators
WO2020120141A1 (en) * 2018-12-11 2020-06-18 UCB Biopharma SRL Functionalised amine derivatives as il-17 modulators
US11225473B2 (en) 2019-01-15 2022-01-18 Gilead Sciences, Inc. FXR (NR1H4) modulating compounds
US11524005B2 (en) 2019-02-19 2022-12-13 Gilead Sciences, Inc. Solid forms of FXR agonists
US11529350B2 (en) 2019-07-03 2022-12-20 Sumitomo Pharma Oncology, Inc. Tyrosine kinase non-receptor 1 (TNK1) inhibitors and uses thereof
US11753406B2 (en) 2019-08-09 2023-09-12 Incyte Corporation Salts of a PD-1/PD-L1 inhibitor
CN114302876A (en) * 2019-09-06 2022-04-08 英夫拉索姆有限公司 NLRP3 inhibitor
US11401279B2 (en) 2019-09-30 2022-08-02 Incyte Corporation Pyrido[3,2-d]pyrimidine compounds as immunomodulators
AU2020360000B2 (en) * 2019-10-02 2023-04-20 Kainos Medicine, Inc. N-(1H-imidazol-2-yl)benzamide compound and pharmaceutical composition comprising the same as active ingredient
WO2021066559A1 (en) * 2019-10-02 2021-04-08 Kainos Medicine, Inc. N-(1h-imidazol-2-yl)benzamide compound and pharmaceutical composition comprising the same as active ingredient
RU2806754C1 (en) * 2019-10-02 2023-11-07 Кайнос Медсин, Инк. N-(1h-imidazol-2-yl)benzamide and pharmaceutical composition containing it as active ingredient
US11866451B2 (en) 2019-11-11 2024-01-09 Incyte Corporation Salts and crystalline forms of a PD-1/PD-L1 inhibitor
US11964953B2 (en) 2020-04-24 2024-04-23 Bayer Aktiengesellschaft Substituted aminothiazoles as DGKzeta inhibitors for immune activation
WO2021214020A1 (en) 2020-04-24 2021-10-28 Bayer Aktiengesellschaft Substituted aminothiazoles as dgkzeta inhibitors for immune activation
WO2021214019A1 (en) 2020-04-24 2021-10-28 Bayer Aktiengesellschaft Substituted aminothiazoles as dgkzeta inhibitors for immune activation
US11702422B2 (en) 2020-04-30 2023-07-18 Janssen Pharmaceutica Nv Imidazopyridazines as modulators of IL-17
US11691979B2 (en) 2020-04-30 2023-07-04 Janssen Pharmaceutica Nv Imidazopyridazines as modulators of IL-17
WO2021255174A1 (en) * 2020-06-18 2021-12-23 Leo Pharma A/S Small molecule modulators of il-17
US11866434B2 (en) 2020-11-06 2024-01-09 Incyte Corporation Process for making a PD-1/PD-L1 inhibitor and salts and crystalline forms thereof
US11760756B2 (en) 2020-11-06 2023-09-19 Incyte Corporation Crystalline form of a PD-1/PD-L1 inhibitor
US11780836B2 (en) 2020-11-06 2023-10-10 Incyte Corporation Process of preparing a PD-1/PD-L1 inhibitor
US11932648B2 (en) 2021-06-28 2024-03-19 Blueprint Medicines Corporation CDK2 inhibitors
US11970498B2 (en) 2021-06-28 2024-04-30 Blueprint Medicines Corporation CDK2 inhibitors

Similar Documents

Publication Publication Date Title
WO2014181287A1 (en) Heterocyclyl compounds and uses thereof
JP6918819B2 (en) Tricyclic compounds useful in the treatment of orthomyxovirus infection
AU2016256728B2 (en) Thiadiazole analogs thereof and methods for treating smn-deficiency-related-conditions
AP1122A (en) Therapeutically active compounds based on based on based on indazole bioisostere replacement of catechol in PDE4 inhibitors.
CN110156770B (en) Aminopyridine derivatives as inhibitors of TAM family kinases
JP5755741B2 (en) 4-Aminoquinazolin-2-yl-1-pyrazole-4-carboxylic acid compounds as prolyl hydroxylase inhibitors
MX2015001941A (en) 4-HETEROARYL SUBSTITUTED BENZOIC ACID COMPOUNDS AS RORgammaT INHIBITORS AND USES THEREOF.
WO2004020393A1 (en) Dibenzylamine compound and medicinal use thereof
CA2915561A1 (en) Novel substituted bicyclic compounds as bromodomain inhibitors
CA2918284A1 (en) Autotaxin inhibitors comprising a heteroaromatic ring-benzyl-amide-cycle core
JP2007515432A (en) (3,4-Disubstituted) propanecarboxylic acids as S1P (Edg) receptor agonists
KR20070057965A (en) Compounds for inflammation and immune-related uses
US10875863B2 (en) RIPK2 inhibitors and method of treating cancer with same
JP6500092B2 (en) Triazole compounds as T-type calcium channel blockers
EP3814335B1 (en) 4-substituted phenyl-1,3,5-triazine derivatives as modulators of trk receptors
TW202332436A (en) Therapeutic compounds
AU2007298539B2 (en) Pyridine derivatives for the treatment of metabolic disorders related to insulin resistance or hyperglycemia
JPH11240832A (en) Amide or amine derivative
CA3087807A1 (en) Inhibitors of indoleamine 2,3-dioxygenase and/or tryptophan 2,3-dioxygenase
EP3914256B1 (en) Gpr35 modulators
WO2015092118A1 (en) Spiro[cyclobutane-1,3&#39;-indolin]-2&#39;-one derivatives as bromodomain inhibitors
CN114341135A (en) Estrogen-related receptor alpha (ERR alpha) modulators
JPWO2019022235A1 (en) Alopecia areata therapeutic or preventive agent
CA2853485A1 (en) Compounds for inflammation and immune-related uses
JP2010180183A (en) New compound having pyrimidin-4(3h)-one structure and medicine containing the same

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: 14794356

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: 14794356

Country of ref document: EP

Kind code of ref document: A1