US20140235642A1 - Trpm8 antagonists and their use in treatments - Google Patents

Trpm8 antagonists and their use in treatments Download PDF

Info

Publication number
US20140235642A1
US20140235642A1 US14/263,143 US201414263143A US2014235642A1 US 20140235642 A1 US20140235642 A1 US 20140235642A1 US 201414263143 A US201414263143 A US 201414263143A US 2014235642 A1 US2014235642 A1 US 2014235642A1
Authority
US
United States
Prior art keywords
methyl
trifluoro
alk
pyridin
phenyl
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US14/263,143
Inventor
James Brown
Jian J. Chen
Vijay Keshav Gore
Scott Harried
Daniel B. Horne
Matthew R. Kaller
Qingyian Liu
Holger Monenschein
Thomas T. Nguyen
Nobuko Nishimura
Wenge Zhong
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Amgen Inc
Original Assignee
Amgen Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Amgen Inc filed Critical Amgen Inc
Priority to US14/263,143 priority Critical patent/US20140235642A1/en
Publication of US20140235642A1 publication Critical patent/US20140235642A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/24Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D213/44Radicals substituted by doubly-bound oxygen, sulfur, or nitrogen atoms, or by two such atoms singly-bound to the same carbon atom
    • C07D213/53Nitrogen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • 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
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C275/00Derivatives of urea, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups
    • C07C275/28Derivatives of urea, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups having nitrogen atoms of urea groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton
    • C07C275/30Derivatives of urea, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups having nitrogen atoms of urea groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton being further substituted by halogen atoms, or by nitro or nitroso groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/24Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D213/36Radicals substituted by singly-bound nitrogen atoms
    • C07D213/40Acylated substituent nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom 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
    • C07D213/72Nitrogen atoms
    • C07D213/75Amino or imino radicals, acylated by carboxylic or carbonic acids, or by sulfur or nitrogen analogues thereof, e.g. carbamates
    • 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
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur 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
    • 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

Definitions

  • the present invention relates to compounds that have TRPM8 antagonist properties and are useful in preparing medicaments and compositions and in treating diseases and conditions such as those mediated by TRPM8.
  • the compounds and compositions may be used to treat various diseases or conditions modulated by TRPM8 such as, but not limited to, migraines and neuropathic pain.
  • Cold sensation is derived from activation of the somatosensory system by a cold stimulus.
  • Calcium imaging and patch clamp experiments in dissociated trigeminal and dorsal root ganglia neurons have revealed cold stimuli induced calcium influx, suggesting the direct opening of a calcium-permeable ion channels by cold (Thut et al., 2003; Reid, 2005).
  • a recently cloned non-selective cation channel, TRPM8 (transient receptor potential melastatin 8) or trp-p8 is activated by cold stimulus of 10 to 24° C.
  • TRPM8 is also activated by compounds that elicit cool sensation such as menthol, icilin (AG-3-5) (McKemy et al., 2002), and the endogenous lipid PIP 2 (Rohacs et al., 2005).
  • TRPM8 is highly expressed in sensory neurons of the trigeminal and dorsal root ganglia (McKemy et al., 2002; Peier et al., 2002; Thut et al., 2003).
  • TRPM8 is also expressed in nerve fibers innervating urinary bladder in guinea pigs (Tsukimi et al., 2005) and humans (Mukerji et al., 2006) and believed to contribute to the bladder hypersensitivity.
  • TRPM8 Activation mechanism of TRPM8 by menthol and icilin appears to differ. Icilin requires calcium for robust activation of TRPM8, whereas menthol and cold do not (Chuang et al., 2004). Typically, activation by all these agonists follows a period of calcium-dependent desensitization.
  • TRPM8 is shown to mediate the analgesia by agonists such as menthol and icilin (by desensitization of the receptor) during experimental neuropathic pain in rodents (Proudfoot et al., 2006). Further, attenuation of cold sensation and cold allodynia after chronic constriction injury model of neuropathic pain in TRPM8 knockout mice (Colburn et al., 2007; Dhaka et al., 2007) suggests that antagonists of TRPM8 may be considered as pain therapeutics for chemotherapy-induced pain, neuropathic pain and bladder disorders.
  • Mint oil that contains menthol an agonist of TRPM8 has been reported to alleviate pain in post-herpetic neuralgia (Davies et al., 2002), a neuropathic pain condition. Furthermore, oral or intracerebroventricular injection of menthol decreased nociceptive responses to hot-plate test and acetic acid-induced writhing in mice (Galeotti et al., 2002). These responses are believed to be mediated by the activation and desensitization of the TRPM8. These observations and the knockout mice studies indicate that TRPM8 modulation by antagonists might be beneficial for patients experiencing neuropathic pain.
  • TRPM8 antagonist compounds that can be used to treat diseases and conditions mediated by TRPM8 such as, but not limited to, migraines and neuropathic pain and those other conditions described herein.
  • the present invention comprises a new class of compounds useful in the treatment of diseases, such as TRPM8-mediated diseases and other maladies, such as inflammatory or neuropathic pain and diseases involving sensory nerve function such as asthma, rheumatoid arthritis, osteoarthritis, inflammatory bowel disorders, urinary incontinence, migraine and psoriasis.
  • the compounds of the invention are useful for the treatment of acute, inflammatory and neuropathic pain, dental pain, general headache, migraine, cluster headache, mixed-vascular and non-vascular syndromes, tension headache, general inflammation, arthritis, rheumatic diseases, osteoarthritis, inflammatory bowel disorders, anxiety, depression, inflammatory eye disorders, inflammatory or unstable bladder disorders, psoriasis, skin complaints with inflammatory components, chronic inflammatory conditions, inflammatory pain and associated hyperalgesia and allodynia, neuropathic pain and associated hyperalgesia and allodynia, diabetic neuropathy pain, causalgia, sympathetically maintained pain, deafferentation syndromes, asthma, epithelial tissue damage or dysfunction, herpes simplex, disturbances of visceral motility at respiratory, genitourinary, gastrointestinal or vascular regions, wounds, burns, allergic skin reactions, pruritus, vitiligo, general gastrointestinal disorders, gastric ulceration, duodenal ulcers, diarrhea, gastric lesions
  • the invention also comprises pharmaceutical compositions comprising the compounds, methods for the treatment of TRPM8-receptor-mediated diseases, such as inflammatory or neuropathic pain, asthma, rheumatoid arthritis, osteoarthritis, inflammatory bowel disorders, urinary incontinence, migraine and psoriasis diseases, using the compounds and compositions of the invention, and intermediates and processes useful for the preparation of the compounds of the invention.
  • TRPM8-receptor-mediated diseases such as inflammatory or neuropathic pain, asthma, rheumatoid arthritis, osteoarthritis, inflammatory bowel disorders, urinary incontinence, migraine and psoriasis diseases
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , Y, X 1 , X 2 , m and n are defined below.
  • the invention provides a compound of Formula I having the structure:
  • n 0, 1, 2 or 3;
  • n 0 or 1
  • X 1 is C(R 4 ) or N;
  • X 2 is CH or N
  • Y is NH, NR 1a , or O
  • Z is O or S
  • R 1 is C 1-6 alk, —(C ⁇ O)—O—C 1-6 alk, or a direct-bonded, C 1-6 alk-linked, C 1-2 alkO-linked, —C( ⁇ O)—O linked, —C( ⁇ O)— linked, saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 heteroatoms selected from N, O and S, the C 1-6 alk, the C 1-6 alk of the C 1-6 alk-link, and the monocyclic or bicyclic ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C 1-6 alk, C 1-6 alkOH, C 1-6 alk-C( ⁇ O)R a , C 1-6 alk-C( ⁇ O)OR a , C 1-4 haloalk, cyano,
  • R 2 is H, halo, cyano, R c , —C( ⁇ O)R b , —C( ⁇ O)OR b , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R b , —OC( ⁇ O)NR a R a , —OC 2-6 alkNR a R a , —OC 2-6 alkOR a , —SR a , —S( ⁇ O)R b , —S( ⁇ O) 2 R b , —S( ⁇ O) 2 NR a R a , —NR a R a , —N(R a )C( ⁇ O)R b , —N(R a )C( ⁇ O)OR b , —N(R a )C( ⁇ O)NR a R a
  • R 3 is H, C 1-8 alk, C 1-8 alkOH, C 1-4 haloalk, halo, cyano, —C( ⁇ O)R b , —C( ⁇ O)OR b , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —ORa, —OC( ⁇ O)R b , —OC( ⁇ O)NR a R a , —OC 2-6 alkNR a R a , —OC 2-6 alkOR a , —SR a , —S( ⁇ O)R b , —S( ⁇ O) 2 R b , —S( ⁇ O) 2 NR a R a , —NR a R a , —N(R a )C( ⁇ O)R b , —N(R a )C( ⁇ O)OR b , —N(R
  • R 5 is independently, in each instance, Cl, Br, F, CH 3 , CF 3 , or OR a ;
  • R 6 is F, CF 3 , C 1-6 alk, or OR a ;
  • R a is independently, at each instance, H or R b ;
  • R b is independently, at each instance, phenyl, benzyl or C 1-6 alk, the phenyl, benzyl and C 1-6 alk being substituted by 0, 1, 2 or 3 substituents selected from halo, oxo, C 1-4 alk, C 1-3 haloalk, —OC 1-4 alk, —OH, —NH 2 , —OC 1-4 alk, —OC 1-4 haloalk, —NHC 1-4 alk, and —N(C 1-4 alk)C 1-4 alk; and
  • R c is independently, at each instance, a saturated, partially saturated or unsaturated 4-, 5- or 6-membered monocyclic ring containing 0, 1, 2, 3 or 4 heteroatoms selected from N, O and S, wherein the C 1-6 alkyl and ring are substituted by 0 or 1 oxo groups substituted by 0, 1, 2 or 3 substituents selected from C 1-8 alk, C 1-4 haloalk, halo and cyano.
  • Y is selected from NH or O.
  • Y is NH whereas in other such embodiments, Y is O.
  • the compound of Formula I has the Formula IA:
  • the compound of Formula I has the Formula IB:
  • Y is NH or O.
  • the compound of Formula I has the Formula IC:
  • n 0, 1, 2 or 3;
  • n 0 or 1
  • X 1 is C(R 4 ) or N;
  • X 2 is CH or N
  • R 1 is selected from C 1-6 alk or a direct-bonded, C 1-2 alk-linked, C 1-2 alkO-linked, saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 heteroatoms selected from N, O and S, but containing no more than one O or S atom, the C 1-6 alk and ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C 1-6 alk, C 1-4 haloalk, cyano, nitro, —C( ⁇ O)R a , —C( ⁇ O)OR a , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R a , —OC( ⁇ O)NR
  • R 2 is selected from H, halo, cyano, R c , —C( ⁇ O)R b , —C( ⁇ O)OR b , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R b , —OC( ⁇ O)NR a R a , —OC 2-6 alkNR a R a , —OC 2-6 alkOR a , —SR a , —S( ⁇ O)R b , —S( ⁇ O) 2 R b , —S( ⁇ O) 2 NR a R a , —NR a R a , —N(R a )C( ⁇ O)R b , —N(R a )C( ⁇ O)OR b , —N(R a )C( ⁇ O)NR a R
  • R 3 is H, C 1-8 alk, C 1-4 haloalk, halo, cyano, —C( ⁇ O)R b , —C( ⁇ O)OR b , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R b , —OC( ⁇ O)NR a R a , —OC 2-6 alkNR a R a , —OC 2-6 alkOR a , —SR a , —S( ⁇ O)R b , —S( ⁇ O) 2 R b , —S( ⁇ O) 2 NR a R a , —NR a R a , —N(R a )C( ⁇ O)R b , —N(R a )C( ⁇ O)OR b , —N(R a )C(
  • R 4 is independently, at each instance, H, C 1-6 alk, —C 1-3 haloalk, —OC 1-6 alk, —OC 1-3 haloalk, —N(C 1-6 alk)C 1-6 alk, —NHC 1-6 alk, —NC( ⁇ O)C 1-6 alk, —N(C 1-6 alk)C 1-6 alk, F, Cl, Br, CN, OH or NH 2 ; or R 3 and R 4 together form a four-atom unsaturated bridge containing 0 or 1 N atoms, wherein the bridge is substituted by 0, 1 or 2 R 5 substituents;
  • R 5 is independently, in each instance, F, CH 3 or CF 3 ;
  • R 6 is F
  • R a is independently, at each instance, H or R b ;
  • R b is independently, at each instance, phenyl, benzyl or C 1-6 alk, the phenyl, benzyl and C 1-6 alk being substituted by 0, 1, 2 or 3 substituents selected from halo, oxo, C 1-4 alk, C 1-3 haloalk, —OC 1-4 alk, —OH, —NH 2 , —OC 1-4 alk, —OC 1-4 haloalk, —NHC 1-4 alk, and —N(C 1-4 alk)C 1-4 alk; and
  • R c is independently, at each instance, a saturated, partially saturated or unsaturated 4-, 5- or 6-membered monocyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, wherein the C 1-6 alkyl and ring are substituted by 0 or 1 oxo groups substituted by 0, 1, 2 or 3 substituents selected from C 1-8 alk, C 1-4 haloalk, halo and cyano.
  • the compound of Formula I has the Formula ID:
  • Y is NH or O.
  • the invention provides a compound or tautomer of any of the embodiments in a neutral form. In some such embodiments, the invention provides a compound of any of the embodiments in a neutral form.
  • the invention provides a pharmaceutically-acceptably salt of the compound or a pharmaceutically acceptable salt of the tautomer of any one of the embodiments. In other embodiments, the invention provides a pharmaceutically-acceptably salt of the compound of any one of the embodiments. In some such embodiments, the salt is a trifluoroacetate or a bis trifluoroacetate salt.
  • compositions that include the compound of any one of the embodiments or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof.
  • Such compositions typically include a pharmaceutically-acceptable diluent or carrier.
  • the invention further provides methods of treating acute, inflammatory and neuropathic pain, dental pain, general headache, migraine, cluster headache, mixed-vascular and non-vascular syndromes, tension headache, general inflammation, arthritis, rheumatic diseases, osteoarthritis, inflammatory bowel disorders, depression, anxiety, inflammatory eye disorders, inflammatory or unstable bladder disorders, psoriasis, skin complaints with inflammatory components, chronic inflammatory conditions, inflammatory pain and associated hyperalgesia and allodynia, neuropathic pain and associated hyperalgesia and allodynia, diabetic neuropathy pain, causalgia, sympathetically maintained pain, deafferentation syndromes, asthma, epithelial tissue damage or dysfunction, herpes simplex, disturbances of visceral motility at respiratory, genitourinary, gastrointestinal or vascular regions, wounds, burns, allergic skin reactions, pruritus, vitiligo, general gastrointestinal disorders, gastric ulceration, duodenal ulcers, diarrhea, gastric lesions induced by necrotising agents
  • Such methods typically include administering the compound according to any of the embodiments or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof to the subject.
  • the subject is suffering from neuropathic pain whereas in other such embodiments, the subject is suffering from migraine pain.
  • the invention also provides the use of the compound of any of the embodiments or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof in the preparation of a medicament.
  • the invention still further provides the use of the compound of any of the embodiments or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof for treating acute, inflammatory and neuropathic pain, dental pain, general headache, migraine, cluster headache, mixed-vascular and non-vascular syndromes, tension headache, general inflammation, arthritis, rheumatic diseases, osteoarthritis, inflammatory bowel disorders, depression, anxiety, inflammatory eye disorders, inflammatory or unstable bladder disorders, psoriasis, skin complaints with inflammatory components, chronic inflammatory conditions, inflammatory pain and associated hyperalgesia and allodynia, neuropathic pain and associated hyperalgesia and allodynia, diabetic neuropathy pain, causalgia, sympathetically maintained pain, deafferentation syndromes, asthma, epithelial tissue damage or dysfunction, herpes simplex, disturbances of visceral motility at respiratory, genitourinary, gastrointestinal or vascular regions, wounds, burns
  • the invention also provides the compound according to any of the embodiments described herein or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof for treating acute, inflammatory and neuropathic pain, dental pain, general headache, migraine, cluster headache, mixed-vascular and non-vascular syndromes, tension headache, general inflammation, arthritis, rheumatic diseases, osteoarthritis, inflammatory bowel disorders, depression, anxiety, inflammatory eye disorders, inflammatory or unstable bladder disorders, psoriasis, skin complaints with inflammatory components, chronic inflammatory conditions, inflammatory pain and associated hyperalgesia and allodynia, neuropathic pain and associated hyperalgesia and allodynia, diabetic neuropathy pain, causalgia, sympathetically maintained pain, deafferentation syndromes, asthma, epithelial tissue damage or dysfunction, herpes simplex, disturbances of visceral motility at respiratory, genitourinary, gastrointestinal or vascular regions, wounds, burns
  • the compound of any of the embodiments or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof is for treating neuropathic pain in a subject. In other such embodiments, the compound of any of the embodiments or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof is for treating migraine pain in a subject
  • the compounds of this invention may have in general several asymmetric centers and are typically depicted in the form of racemic mixtures. This invention is intended to encompass racemic mixtures, partially racemic mixtures and separate enantiomers and diasteromers.
  • C ⁇ - ⁇ alk means an alkyl group comprising a minimum of ⁇ and a maximum of ⁇ carbon atoms in a branched, cyclical or linear relationship or any combination of the three, wherein ⁇ and ⁇ represent integers.
  • the alkyl groups described in this section may also contain one or two double or triple bonds.
  • a designation of C 0 alk indicates a direct bond.
  • Examples of C 1-6 alkyl include, but are not limited to the following:
  • C ⁇ - ⁇ alkyl and “C ⁇ - ⁇ cycloalkyl” relate to acyclic saturated alkyls and cyclic saturated alkyls, respectively.
  • Benzo group alone or in combination, means the divalent radical C 4 H 4 ⁇ , one representation of which is —CH ⁇ CH—CH ⁇ CH—, that when vicinally attached to another ring forms a benzene-like ring—for example tetrahydronaphthylene, indole and the like.
  • oxo and thioxo represent the groups ⁇ O (as in carbonyl) and ⁇ S (as in thiocarbonyl), respectively.
  • cyano refers to a nitrile group which may be written as —C ⁇ N.
  • Halo or “halogen” means a halogen atoms selected from F, Cl, Br and I.
  • C V-W haloalk means an alk group, as described above, wherein any number—at least one—of the hydrogen atoms attached to the alk chain are replaced by F, Cl, Br or I.
  • the group N(R a )R a and the like include substituents where the two R a groups together form a ring, optionally including a N, O or S atom, and include groups such as:
  • N(C ⁇ - ⁇ alk)C ⁇ - ⁇ alk wherein ⁇ and ⁇ are as defined above, include substituents where the two C ⁇ - ⁇ alk groups together form a ring, optionally including a N, O or S atom, and include groups such as:
  • Heterocycle means a ring comprising at least one carbon atom and at least one other atom selected from N, O and S. Examples of heterocycles that may be found in the claims include, but are not limited to, the following:
  • “Pharmaceutically-acceptable salt” means a salt prepared by conventional means, and are well known by those skilled in the art.
  • the “pharmacologically acceptable salts” include basic salts of inorganic and organic acids, including but not limited to hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, malic acid, acetic acid, trifluoroacetic acid, oxalic acid, tartaric acid, citric acid, lactic acid, fumaric acid, succinic acid, maleic acid, salicylic acid, benzoic acid, phenylacetic acid, mandelic acid and the like.
  • suitable pharmaceutically acceptable cation pairs for the carboxy group are well known to those skilled in the art and include alkaline, alkaline earth, ammonium, quaternary ammonium cations and the like.
  • pharmaceutically acceptable salts see infra and Berge et al., J. Pharm. Sci. 66:1 (1977).
  • “Saturated, partially-saturated or unsaturated” includes substituents saturated with hydrogens, substituents completely unsaturated with hydrogens and substituents partially saturated with hydrogens.
  • leaving group generally refers to groups readily displaceable by a nucleophile, such as an amine, a thiol or an alcohol nucleophile. Such leaving groups are well known in the art. Examples of such leaving groups include, but are not limited to, N-hydroxysuccinimide, N-hydroxybenzotriazole, halides, triflates, tosylates and the like. Preferred leaving groups are indicated herein where appropriate.
  • Protecting group generally refers to groups well known in the art which are used to prevent selected reactive groups, such as carboxy, amino, hydroxy, mercapto and the like, from undergoing undesired reactions, such as nucleophilic, electrophilic, oxidation, reduction and the like. Preferred protecting groups are indicated herein where appropriate. Examples of amino protecting groups include, but are not limited to, aralkyl, substituted aralkyl, cycloalkenylalkyl and substituted cycloalkenyl alkyl, allyl, substituted allyl, acyl, alkoxycarbonyl, aralkoxycarbonyl, silyl and the like.
  • aralkyl examples include, but are not limited to, benzyl, ortho-methylbenzyl, trityl and benzhydryl, which can be optionally substituted with halogen, alkyl, alkoxy, hydroxy, nitro, acylamino, acyl and the like, and salts, such as phosphonium and ammonium salts.
  • aryl groups include phenyl, naphthyl, indanyl, anthracenyl, 9-(9-phenylfluorenyl), phenanthrenyl, durenyl and the like.
  • cycloalkenylalkyl or substituted cycloalkylenylalkyl radicals preferably have 6-10 carbon atoms, include, but are not limited to, cyclohexenyl methyl and the like.
  • Suitable acyl, alkoxycarbonyl and aralkoxycarbonyl groups include benzyloxycarbonyl, t-butoxycarbonyl, iso-butoxycarbonyl, benzoyl, substituted benzoyl, butyryl, acetyl, trifluoroacetyl, trichloro acetyl, phthaloyl and the like.
  • a mixture of protecting groups can be used to protect the same amino group, such as a primary amino group can be protected by both an aralkyl group and an aralkoxycarbonyl group.
  • Amino protecting groups can also form a heterocyclic ring with the nitrogen to which they are attached, for example, 1,2-bis(methylene)benzene, phthalimidyl, succinimidyl, maleimidyl and the like and where these heterocyclic groups can further include adjoining aryl and cycloalkyl rings.
  • the heterocyclic groups can be mono-, di- or tri-substituted, such as nitrophthalimidyl.
  • Amino groups may also be protected against undesired reactions, such as oxidation, through the formation of an addition salt, such as hydrochloride, toluenesulfonic acid, trifluoroacetic acid and the like.
  • an addition salt such as hydrochloride, toluenesulfonic acid, trifluoroacetic acid and the like.
  • Many of the amino protecting groups are also suitable for protecting carboxy, hydroxy and mercapto groups.
  • aralkyl groups are also suitable groups for protecting hydroxy and mercapto groups, such as tert-butyl.
  • Silyl protecting groups are silicon atoms optionally substituted by one or more alkyl, aryl and aralkyl groups. Suitable silyl protecting groups include, but are not limited to, trimethylsilyl, triethylsilyl, triisopropylsilyl, tert-butyldimethylsilyl, dimethylphenylsilyl, 1,2-bis(dimethylsilyl)benzene, 1,2-bis(dimethylsilyl)ethane and diphenylmethylsilyl. Silylation of an amino groups provide mono- or di-silylamino groups. Silylation of aminoalcohol compounds can lead to a N,N,O-trisilyl derivative.
  • silyl function from a silyl ether function is readily accomplished by treatment with, for example, a metal hydroxide or ammonium fluoride reagent, either as a discrete reaction step or in situ during a reaction with the alcohol group.
  • Suitable silylating agents are, for example, trimethylsilyl chloride, tert-butyl-dimethylsilyl chloride, phenyldimethylsilyl chloride, diphenylmethyl silyl chloride or their combination products with imidazole or DMF.
  • Methods for silylation of amines and removal of silyl protecting groups are well known to those skilled in the art.
  • Methods of preparation of these amine derivatives from corresponding amino acids, amino acid amides or amino acid esters are also well known to those skilled in the art of organic chemistry including amino acid/amino acid ester or aminoalcohol chemistry.
  • Protecting groups are removed under conditions which will not affect the remaining portion of the molecule. These methods are well known in the art and include acid hydrolysis, hydrogenolysis and the like.
  • a preferred method involves removal of a protecting group, such as removal of a benzyloxycarbonyl group by hydrogenolysis utilizing palladium on carbon in a suitable solvent system such as an alcohol, acetic acid, and the like or mixtures thereof.
  • a t-butoxycarbonyl protecting group can be removed utilizing an inorganic or organic acid, such as HCl or trifluoroacetic acid, in a suitable solvent system, such as dioxane or methylene chloride. The resulting amino salt can readily be neutralized to yield the free amine.
  • Carboxy protecting group such as methyl, ethyl, benzyl, tert-butyl, 4-methoxyphenylmethyl and the like, can be removed under hydrolysis and hydrogenolysis conditions well known to those skilled in the art.
  • Prodrugs of the compounds of this invention are also contemplated by this invention.
  • a prodrug is an active or inactive compound that is modified chemically through in vivo physiological action, such as hydrolysis, metabolism and the like, into a compound of this invention following administration of the prodrug to a patient.
  • the suitability and techniques involved in making and using prodrugs are well known by those skilled in the art.
  • For a general discussion of prodrugs involving esters see Svensson and Tunek Drug Metabolism Reviews 165 (1988) and Bundgaard Design of Prodrugs, Elsevier (1985).
  • Examples of a masked carboxylate anion include a variety of esters, such as alkyl (for example, methyl, ethyl), cycloalkyl (for example, cyclohexyl), aralkyl (for example, benzyl, p-methoxybenzyl), and alkylcarbonyloxyalkyl (for example, pivaloyloxymethyl).
  • esters such as alkyl (for example, methyl, ethyl), cycloalkyl (for example, cyclohexyl), aralkyl (for example, benzyl, p-methoxybenzyl), and alkylcarbonyloxyalkyl (for example, pivaloyloxymethyl).
  • Amines have been masked as arylcarbonyloxymethyl substituted derivatives which are cleaved by esterases in vivo releasing the free drug and formaldehyde (Bungaard J. Med. Chem. 2503 (1989
  • drugs containing an acidic NH group such as imidazole, imide, indole and the like, have been masked with N-acyloxymethyl groups (Bundgaard Design of Prodrugs, Elsevier (1985)). Hydroxy groups have been masked as esters and ethers.
  • EP 039,051 (Sloan and Little, Apr. 11, 1981) discloses Mannich-base hydroxamic acid prodrugs, their preparation and use.
  • One aspect of the current invention relates to compounds having the general structure:
  • n 0, 1, 2 or 3;
  • n 0 or 1
  • X 1 is C(R 4 ) or N;
  • X 2 is C or N
  • Y is NH or O
  • R 1 is selected from C 1-6 alk or a direct-bonded, C 1-2 alk-linked, C 1-2 alkO-linked, saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, but containing no more than one O or S atom, the C 1-6 alk and ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C 1-6 alk, C 1-4 haloalk, cyano, nitro, —C( ⁇ O)R a , —C( ⁇ O)OR a , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R a , —OC( ⁇ O)NR
  • R 2 is selected from H, halo, cyano, R c , —C( ⁇ O)R b , —C( ⁇ O)OR b , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R b , —OC( ⁇ O)NR a R a , —OC 2-6 alkNR a R a , —OC 2-6 alkOR a , —SR a , —S( ⁇ O)R b , —S( ⁇ O) 2 R b , —S( ⁇ O) 2 NR a R a , —NR a R a , —N(R a )C( ⁇ O)R b , —N(R a )C( ⁇ O)OR b , —N(R a )C( ⁇ O)NR a R
  • R 3 is H, C 1-8 alk, C 1-4 haloalk, halo, cyano, —C( ⁇ O)R b , —C( ⁇ O)OR b , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R b , —OC( ⁇ O)NR a R a , —OC 2-6 alkNR a R a , —OC 2-6 alkOR a , —SR a , —S( ⁇ O)R b , —S( ⁇ O) 2 R b , —S( ⁇ O) 2 NR a R a , —NR a R a , —N(R a )C( ⁇ O)R b , —N(R a )C( ⁇ O)OR b , —N(R a )C(
  • R 4 is independently, at each instance, H, C 1-6 alk, —C 1-3 haloalk, —OC 1-6 alk, —OC 1-3 haloalk, —N(C 1-6 alk)C 1-6 alk, —NHC 1-6 alk, —NC( ⁇ O)C 1-6 alk, —N(C 1-6 alk)C 1-6 alk, F, Cl, Br, CN, OH or NH 2 ; or R 3 and R 4 together form a four-atom unsaturated bridge containing 0 or 1 N atoms, wherein the bridge is substituted by 0, 1 or 2 R 5 substituents;
  • R 5 is independently, in each instance, F, CH 3 or CF 3 ;
  • R 6 is F
  • R a is independently, at each instance, H or R b ;
  • R b is independently, at each instance, phenyl, benzyl or C 1-6 alk, the phenyl, benzyl and C 1-6 alk being substituted by 0, 1, 2 or 3 substituents selected from halo, oxo, C 1-4 alk, C 1-3 haloalk, —OC 1-4 alk, —OH, —NH 2 , —OC 1-4 alk, —OC 1-4 haloalk, —NHC 1-4 alk, and —N(C 1-4 alk)C 1-4 alk; and
  • R c is independently, at each instance, a saturated, partially saturated or unsaturated 4-, 5- or 6-membered monocyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, wherein the C 1-6 alkyl and ring are substituted by 0 or 1 oxo groups substituted by 0, 1, 2 or 3 substituents selected from C 1-8 alk, C 1-4 haloalk, halo and cyano.
  • Another aspect of the current invention relates to compounds having the general structure:
  • n 0, 1, 2 or 3;
  • n 0 or 1
  • X 1 is C(R 4 ) or N;
  • X 2 is C or N
  • R 1 is selected from C 1-6 alk or a direct-bonded, C 1-2 alk-linked, C 1-2 alkO-linked, saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, but containing no more than one O or S atom, the C 1-6 alk and ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C 1-6 alk, C 1-4 haloalk, cyano, nitro, —C( ⁇ O)R a , —C( ⁇ O)OR a , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R a , —OC( ⁇ O)NR
  • R 2 is selected from H, halo, cyano, R c , —C( ⁇ O)R b , —C( ⁇ O)OR b , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R b , —OC( ⁇ O)NR a R a , —OC 2-6 alkNR a R a , —OC 2-6 alkOR a , —SR a , —S( ⁇ O)R b , —S( ⁇ O) 2 R b , —S( ⁇ O) 2 NR a R a , —NR a R a , —N(R a )C( ⁇ O)R b , —N(R a )C( ⁇ O)OR b , —N(R a )C( ⁇ O)NR a R
  • R 3 is H, C 1-8 alk, C 1-4 haloalk, halo, cyano, —C( ⁇ O)R b , —C( ⁇ O)OR b , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R b , —OC( ⁇ O)NR a R a , —OC 2-6 alkNR a R a , —OC 2-6 alkOR a , —SR a , —S( ⁇ O)R b , —S( ⁇ O) 2 R b , —S( ⁇ O) 2 NR a R a , —NR a R a , —N(R a )C( ⁇ O)R b , —N(R a )C( ⁇ O)OR b , —N(R a )C(
  • R 4 is independently, at each instance, H, C 1-6 alk, —C 1-3 haloalk, —OC 1-6 alk, —OC 1-3 haloalk, —N(C 1-6 alk)C 1-6 alk, —NHC 1-6 alk, —NC( ⁇ O)C 1-6 alk, —N(C 1-6 alk)C 1-6 alk, F, Cl, Br, CN, OH or NH 2 ; or R 3 and R 4 together form a four-atom unsaturated bridge containing 0 or 1 N atoms, wherein the bridge is substituted by 0, 1 or 2 R 5 substituents;
  • R 5 is independently, in each instance, F, CH 3 or CF 3 ;
  • R 6 is F
  • R a is independently, at each instance, H or R b ;
  • R b is independently, at each instance, phenyl, benzyl or C 1-6 alk, the phenyl, benzyl and C 1-6 alk being substituted by 0, 1, 2 or 3 substituents selected from halo, oxo, C 1-4 alk, C 1-3 haloalk, —OC 1-4 alk, —OH, —NH 2 , —OC 1-4 alk, —OC 1-4 haloalk, —NHC 1-4 alk, and —N(C 1-4 alk)C 1-4 alk; and
  • R c is independently, at each instance, a saturated, partially saturated or unsaturated 4-, 5- or 6-membered monocyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, wherein the C 1-6 alkyl and ring are substituted by 0 or 1 oxo groups substituted by 0, 1, 2 or 3 substituents selected from C 1-8 alk, C 1-4 haloalk, halo and cyano.
  • Another aspect of the current invention relates to compounds having the general structure:
  • n 0, 1, 2 or 3;
  • n 0 or 1
  • X 1 is C(R 4 ) or N;
  • R 1 is selected from C 1-6 alk or a direct-bonded, C 1-2 alk-linked, C 1-2 alkO-linked, saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, but containing no more than one O or S atom, the C 1-6 alk and ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C 1-6 alk, C 1-4 haloalk, cyano, nitro, —C( ⁇ O)R a , —C( ⁇ O)OR a , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R a , —OC( ⁇ O)NR
  • R 2 is selected from H, halo, cyano, R c , —C( ⁇ O)R b , —C( ⁇ O)OR b , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R b , —OC( ⁇ O)NR a R a , —OC 2-6 alkNR a R a , —OC 2-6 alkOR a , —SR a , —S( ⁇ O)R b , —S( ⁇ O) 2 R b , —S( ⁇ O) 2 NR a R a , —NR a R a , —N(R a )C( ⁇ O)R b , —N(R a )C( ⁇ O)OR b , —N(R a )C( ⁇ O)NR a R
  • R 3 is H, C 1-8 alk, C 1-4 haloalk, halo, cyano, —C( ⁇ O)R b , —C( ⁇ O)OR b , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R b , —OC( ⁇ O)NR a R a , —OC 2-6 alkNR a R a , —OC 2-6 alkOR a , —SR a , —S( ⁇ O)R b , —S( ⁇ O) 2 R b , —S( ⁇ O) 2 NR a R a , —NR a R a , —N(R a )C( ⁇ O)R b , —N(R a )C( ⁇ O)OR b , —N(R a )C(
  • R 4 is independently, at each instance, H, C 1-6 alk, —C 1-3 haloalk, —OC 1-6 alk, —OC 1-3 haloalk, —N(C 1-6 alk)C 1-6 alk, —NHC 1-6 alk, —NC( ⁇ O)C 1-6 alk, —N(C 1-6 alk)C 1-6 alk, F, Cl, Br, CN, OH or NH 2 ; or R 3 and R 4 together form a four-atom unsaturated bridge containing 0 or 1 N atoms, wherein the bridge is substituted by 0, 1 or 2 R 5 substituents;
  • R 5 is independently, in each instance, F, CH 3 or CF 3 ;
  • R 6 is F
  • R a is independently, at each instance, H or R b ;
  • R b is independently, at each instance, phenyl, benzyl or C 1-6 alk, the phenyl, benzyl and C 1-6 alk being substituted by 0, 1, 2 or 3 substituents selected from halo, oxo, C 1-4 alk, C 1-3 haloalk, —OC 1-4 alk, —OH, —NH 2 , —OC 1-4 alk, —OC 1-4 haloalk, —NHC 1-4 alk, and —N(C 1-4 alk)C 1-4 alk; and
  • R c is independently, at each instance, a saturated, partially saturated or unsaturated 4-, 5- or 6-membered monocyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, wherein the C 1-6 alkyl and ring are substituted by 0 or 1 oxo groups substituted by 0, 1, 2 or 3 substituents selected from C 1-8 alk, C 1-4 haloalk, halo and cyano.
  • Another aspect of the current invention relates to compounds having the general structure:
  • n 0, 1, 2 or 3;
  • n 0 or 1
  • X 2 is C or N
  • R 1 is selected from C 1-6 alk or a direct-bonded, C 1-2 alk-linked, C 1-2 alkO-linked, saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, but containing no more than one O or S atom, the C 1-6 alk and ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C 1-6 alk, C 1-4 haloalk, cyano, nitro, —C( ⁇ O)R a , —C( ⁇ O)OR a , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R a , —OC( ⁇ O)NR
  • R 2 is selected from H, halo, cyano, R c , —C( ⁇ O)R b , —C( ⁇ O)OR b , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R b , —OC( ⁇ O)NR a R a , —OC 2-6 alkNR a R a , —OC 2-6 alkOR a , —SR a , —S( ⁇ O)R b , —S( ⁇ O) 2 R b , —S( ⁇ O) 2 NR a R a , —NR a R a , —N(R a )C( ⁇ O)R b , —N(R a )C( ⁇ O)OR b , —N(R a )C( ⁇ O)NR a R
  • R 3 is H, C 1-8 alk, C 1-4 haloalk, halo, cyano, —C( ⁇ O)R b , —C( ⁇ O)OR b , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R b , —OC( ⁇ O)NR a R a , —OC 2-6 alkNR a R a , —OC 2-6 alkOR a , —SR a , —S( ⁇ O)R b , —S( ⁇ O) 2 R b , —S( ⁇ O) 2 NR a R a , —NR a R a , —N(R a )C( ⁇ O)R b , —N(R a )C( ⁇ O)OR b , —N(R a )C(
  • R 4 is independently, at each instance, H, C 1-6 alk, —C 1-3 haloalk, —OC 1-6 alk, —OC 1-3 haloalk, —N(C 1-6 alk)C 1-6 alk, —NHC 1-6 alk, —NC( ⁇ O)C 1-6 alk, —N(C 1-6 alk)C 1-6 alk, F, Cl, Br, CN, OH or NH 2 ;
  • R 5 is F, CH 3 or CF 3 ;
  • R 6 is F
  • R a is independently, at each instance, H or R b ;
  • R b is independently, at each instance, phenyl, benzyl or C 1-6 alk, the phenyl, benzyl and C 1-6 alk being substituted by 0, 1, 2 or 3 substituents selected from halo, oxo, C 1-4 alk, C 1-3 haloalk, —OC 1-4 alk, —OH, —NH 2 , —OC 1-4 alk, —OC 1-4 haloalk, —NHC 1-4 alk, and —N(C 1-4 alk)C 1-4 alk; and
  • R c is independently, at each instance, a saturated, partially saturated or unsaturated 4-, 5- or 6-membered monocyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, wherein the C 1-6 alkyl and ring are substituted by 0 or 1 oxo groups substituted by 0, 1, 2 or 3 substituents selected from C 1-8 alk, C 1-4 haloalk, halo and cyano.
  • Another aspect of the current invention relates to compounds having the general structure:
  • n 0, 1, 2 or 3;
  • R 1 is a direct-bonded, partially-saturated or unsaturated 5- or 6-membered monocyclic ring containing 1 or 2 atoms selected from N, O and S, but containing no more than one O or S atom, the C 1-6 alk and ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C 1-6 alk, C 1-4 haloalk and cyano;
  • R 2 is selected from F and CF 3 ;
  • R 3 is CH 3 , CF 3 , F or Cl
  • R 4 is H or F
  • R 6 is F
  • R a is independently, at each instance, H or R b ;
  • R b is independently, at each instance, phenyl, benzyl or C 1-6 alk, the phenyl, benzyl and C 1-6 alk being substituted by 0, 1, 2 or 3 substituents selected from halo, oxo, C 1-4 alk, C 1-3 haloalk, —OC 1-4 alk, —OH, —NH 2 , —OC 1-4 alk, —OC 1-4 haloalk, —NHC 1-4 alk, and —N(C 1-4 alk)C 1-4 alk; and
  • R c is independently, at each instance, a saturated, partially saturated or unsaturated 4-, 5- or 6-membered monocyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, wherein the C 1-6 alkyl and ring are substituted by 0 or 1 oxo groups substituted by 0, 1, 2 or 3 substituents selected from C 1-8 alk, C 1-4 haloalk, halo and cyano.
  • Another aspect of the current invention relates to compounds having the general structure:
  • n 0, 1, 2 or 3;
  • R 1 is a direct-bonded, partially-saturated or unsaturated 5- or 6-membered monocyclic ring containing 1 or 2 atoms selected from N, O and S, but containing no more than one O or S atom, the C 1-6 alk and ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C 1-6 alk, C 1-4 haloalk and cyano; and
  • R 2 is selected from F and CF 3 .
  • Another aspect of the current invention relates to compounds having the general structure:
  • n 0, 1, 2 or 3;
  • n 0 or 1
  • X 1 is C(R 4 ) or N;
  • X 2 is C or N
  • Y is NH or O
  • R 1 is selected from C 1-6 alk or a direct-bonded, C 1-2 alk-linked, C 1-2 alkO-linked, saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, but containing no more than one O or S atom, the C 1-6 alk and ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C 1-6 alk, C 1-4 haloalk, cyano, nitro, —C( ⁇ O)R a , —C( ⁇ O)OR a , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R a , —OC( ⁇ O)NR
  • R 2 is selected from H, halo, cyano, R c , —C( ⁇ O)R b , —C( ⁇ O)OR b , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R b , —OC( ⁇ O)NR a R a , —OC 2-6 alkNR a R a , —OC 2-6 alkOR a , —SR a , —S( ⁇ O)R b , —S( ⁇ O) 2 R b , —S( ⁇ O) 2 NR a R a , —NR a R a , —N(R a )C( ⁇ O)R b , —N(R a )C( ⁇ O)OR b , —N(R a )C( ⁇ O)NR a R
  • R 3 is H, C 1-8 alk, C 1-4 haloalk, halo, cyano, —C( ⁇ O)R b , —C( ⁇ O)OR b , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R b , —OC( ⁇ O)NR a R a , —OC 2-6 alkNR a R a , —OC 2-6 alkOR a , —SR a , —S( ⁇ O)R b , —S( ⁇ O) 2 R b , —S( ⁇ O) 2 NR a R a , —NR a R a , —N(R a )C( ⁇ O)R b , —N(R a )C( ⁇ O)OR b , —N(R a )C(
  • R 4 is independently, at each instance, H, C 1-6 alk, —C 1-3 haloalk, —OC 1-6 alk, —OC 1-3 haloalk, —N(C 1-6 alk)C 1-6 alk, —NHC 1-6 alk, —NC( ⁇ O)C 1-6 alk, —N(C 1-6 alk)C 1-6 alk, F, Cl, Br, CN, OH or NH 2 ;
  • R 5 is F, CH 3 or CF 3 ;
  • R 6 is F
  • R a is independently, at each instance, H or R b ;
  • R b is independently, at each instance, phenyl, benzyl or C 1-6 alk, the phenyl, benzyl and C 1-6 alk being substituted by 0, 1, 2 or 3 substituents selected from halo, oxo, C 1-4 alk, C 1-3 haloalk, —OC 1-4 alk, —OH, —NH 2 , —OC 1-4 alk, —OC 1-4 haloalk, —NHC 1-4 alk, and —N(C 1-4 alk)C 1-4 alk; and
  • R c is independently, at each instance, a saturated, partially saturated or unsaturated 4-, 5- or 6-membered monocyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, wherein the C 1-6 alkyl and ring are substituted by 0 or 1 oxo groups substituted by 0, 1, 2 or 3 substituents selected from C 1-8 alk, C 1-4 haloalk, halo and cyano.
  • R 1 is C 1-6 alk substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C 1-6 alk, C 1-4 haloalk, cyano, nitro, —C( ⁇ O)R a , —C( ⁇ O)OR a , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R a , —OC( ⁇ O)NR a R a , —OC( ⁇ O)N(R a )S( ⁇ O) 2 R a , —OC 2-6 alkNR a R a , —OC 2-6 alkOR a , —SR a , —S( ⁇ O)R a , —S( ⁇ O) 2 R a , —S( ⁇ O) 2 NR a R
  • R 1 is C 1-2 alk-linked saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, but containing no more than one O or S atom, the ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C 1-6 alk, C 1-4 haloalk, cyano, nitro, —C( ⁇ O)R a , —C( ⁇ O)OR a , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R a , —OC( ⁇ O)NR a R a , —OC( ⁇ O)N(R a )S( ⁇ O) 2 R a , —OC 2-6
  • R 1 is a direct-bonded saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, but containing no more than one O or S atom, the ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C 1-6 alk, C 1-4 haloalk, cyano, nitro, —C( ⁇ O)R a , —C( ⁇ O)OR a , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R a , —OC( ⁇ O)NR a R a , —OC( ⁇ O)N(R a
  • R 1 is a direct-bonded partially-saturated or unsaturated 5- or 6-membered monocyclic ring containing 1, 2, 3 or 4 atoms selected from N, O and S, but containing no more than one O or S atom, the ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C 1-6 alk, C 1-4 haloalk, cyano, nitro, —C( ⁇ O)R a , —C( ⁇ O)OR a , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R a , —OC( ⁇ O)NR a R a , —OC( ⁇ O)N(R a )S( ⁇ O) 2 R a , —OC 2-6 alkNR a R a , —
  • R 1 is a direct-bonded partially-saturated or unsaturated 6-membered monocyclic ring containing 1 or 2 N atoms, substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C 1-6 alk, C 1-4 haloalk, cyano and —OR a .
  • R 1 is a direct-bonded unsaturated 10-membered bicyclic ring containing 1 or 2 N atoms, substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C 1-6 alk, C 1-4 haloalk, cyano and —OR a .
  • Another aspect of the invention relates to a method of treating acute, inflammatory and neuropathic pain, dental pain, general headache, migraine, cluster headache, mixed-vascular and non-vascular syndromes, tension headache, general inflammation, arthritis, rheumatic diseases, osteoarthritis, inflammatory bowel disorders, depression, anxiety, inflammatory eye disorders, inflammatory or unstable bladder disorders, psoriasis, skin complaints with inflammatory components, chronic inflammatory conditions, inflammatory pain and associated hyperalgesia and allodynia, neuropathic pain and associated hyperalgesia and allodynia, diabetic neuropathy pain, causalgia, sympathetically maintained pain, deafferentation syndromes, asthma, epithelial tissue damage or dysfunction, herpes simplex, disturbances of visceral motility at respiratory, genitourinary, gastrointestinal or vascular regions, wounds, burns, allergic skin reactions, pruritus, vitiligo, general gastrointestinal disorders, gastric ulceration, duodenal ulcers, diarrhea, gastric lesions
  • Another aspect of the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound according to claim 1 and a pharmaceutically-acceptable diluent or carrier.
  • Another aspect of the invention relates to the use of a compound according to any of the above embodiments as a medicament.
  • Another aspect of the invention relates to the use of a compound according to any of the above embodiments in the manufacture of a medicament for the treatment of acute, inflammatory and neuropathic pain, dental pain, general headache, migraine, cluster headache, mixed-vascular and non-vascular syndromes, tension headache, general inflammation, arthritis, rheumatic diseases, osteoarthritis, inflammatory bowel disorders, anxiety, depression, inflammatory eye disorders, inflammatory or unstable bladder disorders, psoriasis, skin complaints with inflammatory components, chronic inflammatory conditions, inflammatory pain and associated hyperalgesia and allodynia, neuropathic pain and associated hyperalgesia and allodynia, diabetic neuropathy pain, causalgia, sympathetically maintained pain, deafferentation syndromes, asthma, epithelial tissue damage or dysfunction, herpes simplex, disturbances of visceral motility at respiratory, genitourinary, gastrointestinal or vascular regions, wounds, burns, allergic skin reactions, pruritus, vitiligo, general gastrointestinal disorders
  • the invention provides a compound of Formula I having the structure:
  • n 0, 1, 2 or 3;
  • n 0 or 1
  • X 1 is C(R 4 ) or N;
  • X 2 is CH or N
  • Y is NH, NR 1a , or O
  • Z is O or S
  • R 1 is C 1-6 alk, —(C ⁇ O)—O—C 1-6 alk, or a direct-bonded, C 1-6 alk-linked, C 1-2 alkO-linked, —C( ⁇ O)—O linked, —C( ⁇ O)— linked, saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 heteroatoms selected from N, O and S, the C 1-6 alk, the C 1-6 alk of the C 1-6 alk-link, and the monocyclic or bicyclic ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C 1-6 alk, C 1-6 alkOH, C 1-6 alk-C( ⁇ O)R a , C 1-6 alk-C( ⁇ O)OR a , C 1-4 haloalk, cyano,
  • R 2 is H, halo, cyano, R c , —C( ⁇ O)R b , —C( ⁇ O)OR b , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R b , —OC( ⁇ O)NR a R a , —OC 2-6 alkNR a R a , —OC 2-6 alkOR a , —SR a , —S( ⁇ O)R b , —S( ⁇ O) 2 R b , —S( ⁇ O) 2 NR a R a , —NR a R a , —N(R a )C( ⁇ O)R b , —N(R a )C( ⁇ O)OR b , —N(R a )C( ⁇ O)NR a R a
  • R 3 is H, C 1-8 alk, C 1-8 alkOH, C 1-4 haloalk, halo, cyano, —C( ⁇ O)R b , —C( ⁇ O)OR b , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R b , —OC( ⁇ O)NR a R a , —OC 2-6 alkNR a R a , —OC 2-6 alkOR a , —SR a , —S( ⁇ O)R b , —S( ⁇ O) 2 R b , —S( ⁇ O) 2 NR a R a , —NR a R a , —N(R a )C( ⁇ O)R b , —N(R a )C( ⁇ O)OR b , —N(
  • R 4 is independently, at each instance, H, C 1-6 alk, —C 1-3 haloalk, —OC 1-6 alk, —OC 1-3 haloalk, —N(C 1-6 alk)C 1-6 alk, —NHC 1-6 alk, —NC( ⁇ O)C 1-6 alk, —N(C 1-6 alk)C 1-6 alk, F, Cl, Br, CN, OH or NH 2 ; or R 3 and R 4 together form a four-atom unsaturated bridge containing 0 or 1 N atoms, wherein the bridge is substituted by 0, 1 or 2 R 5 substituents;
  • R 5 is independently, in each instance, Cl, Br, F, CH 3 , CF 3 , or OR a ;
  • R 6 is F, CF 3 , C 1-6 alk, or OR a ;
  • R a is independently, at each instance, H or R b ;
  • R b is independently, at each instance, phenyl, benzyl or C 1-6 alk, the phenyl, benzyl and C 1-6 alk being substituted by 0, 1, 2 or 3 substituents selected from halo, oxo, C 1-4 alk, C 1-3 haloalk, —OC 1-4 alk, —OH, —NH 2 , —OC 1-4 alk, —OC 1-4 haloalk, —NHC 1-4 alk, and —N(C 1-4 alk)C 1-4 alk; and
  • R c is independently, at each instance, a saturated, partially saturated or unsaturated 4-, 5- or 6-membered monocyclic ring containing 0, 1, 2, 3 or 4 heteroatoms selected from N, O and S, wherein the C 1-6 alkyl and ring are substituted by 0 or 1 oxo groups substituted by 0, 1, 2 or 3 substituents selected from C 1-8 alk, C 1-4 haloalk, halo and cyano.
  • Y is NH or O.
  • n 0, 1, 2 or 3;
  • n 0 or 1
  • X 1 is C(R 4 ) or N;
  • X 2 is CH or N
  • R 1 is selected from C 1-6 alk or a direct-bonded, C 1-2 alk-linked, C 1-2 alkO-linked, saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 heteroatoms selected from N, O and S, but containing no more than one O or S atom, the C 1-6 alk and ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C 1-6 alk, C 1-4 haloalk, cyano, nitro, —C( ⁇ O)R a , —C( ⁇ O)OR a , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R a , —OC( ⁇ O)NR
  • R 2 is selected from H, halo, cyano, R c , —C( ⁇ O)R b , —C( ⁇ O)OR b , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R b , —OC( ⁇ O)NR a R a , —OC 2-6 alkNR a R a , —OC 2-6 alkOR a , —SR a , —S( ⁇ O)R b , —S( ⁇ O) 2 R b , —S( ⁇ O) 2 NR a R a , —NR a R a , —N(R a )C( ⁇ O)R b , —N(R a )C( ⁇ O)OR b , —N(R a )C( ⁇ O)NR a R
  • R 3 is H, C 1-8 alk, C 1-4 haloalk, halo, cyano, —C( ⁇ O)R b , —C( ⁇ O)OR b , —C( ⁇ O)NR a R a , —C( ⁇ NR a )NR a R a , —OR a , —OC( ⁇ O)R b , —OC( ⁇ O)NR a R a , —OC 2-6 alkNR a R a , —OC 2-6 alkOR a , —SR a , —S( ⁇ O)R b , —S( ⁇ O) 2 R b , —S( ⁇ O) 2 NR a R a , —NR a R a , —N(R a )C( ⁇ O)R b , —N(R a )C( ⁇ O)OR b , —N(R a )C(
  • R 4 is independently, at each instance, H, C 1-6 alk, —C 1-3 haloalk, —OC 1-6 alk, —OC 1-3 haloalk, —N(C 1-6 alk)C 1-6 alk, —NHC 1-6 alk, —NC( ⁇ O)C 1-6 alk, —N(C 1-6 alk)C 1-6 alk, F, Cl, Br, CN, OH or NH 2 ; or R 3 and R 4 together form a four-atom unsaturated bridge containing 0 or 1 N atoms, wherein the bridge is substituted by 0, 1 or 2 R 5 substituents;
  • R 5 is independently, in each instance, F, CH 3 or CF 3 ;
  • R 6 is F
  • R a is independently, at each instance, H or R b ;
  • R b is independently, at each instance, phenyl, benzyl or C 1-6 alk, the phenyl, benzyl and C 1-6 alk being substituted by 0, 1, 2 or 3 substituents selected from halo, oxo, C 1-4 alk, C 1-3 haloalk, —OC 1-4 alk, —OH, —NH 2 , —OC 1-4 alk, —OC 1-4 haloalk, —NHC 1-4 alk, and —N(C 1-4 alk)C 1-4 alk; and
  • R c is independently, at each instance, a saturated, partially saturated or unsaturated 4-, 5- or 6-membered monocyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, wherein the C 1-6 alkyl and ring are substituted by 0 or 1 oxo groups substituted by 0, 1, 2 or 3 substituents selected from C 1-8 alk, C 1-4 haloalk, halo and cyano.
  • Y is NH or O.
  • R 1 is a saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 heteroatoms selected from N, O and S, wherein the monocyclic or bicyclic ring is independently substituted by 0, 1, 2 or 3 substituents.
  • R 1 is selected from phenyl, pyridyl, pyridinonyl, piperidinonyl, pyrimidinyl, tetradyrofuranyl, tetrahydropyranyl, oxetanyl, furanyl, tetrahydrothiophenyl, thiophenyl, isoxazolyl, pyrrolidinonyl, piperidinyl, cyclohexyl, cyclohexanonyl, quinolinyl, isoquinolinyl, tetrahydronaphthalenyl, dihydroindenyl, indolinonyl, indolinyl, or benzofuranyl independently substituted by 0, 1, 2, or 3 substituents.
  • R 1 is a C 1-6 alk-linked, C 1-2 alkO-linked, —C( ⁇ O)—O linked, or —C( ⁇ O)— linked, saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 heteroatoms selected from N, O and S, wherein the monocyclic or bicyclic ring is substituted by 0, 1, 2 or 3 substituents.
  • R 1 is a —C 1-2 alk-linked saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 heteroatoms selected from N, O and S, wherein the monocyclic or bicyclic ring is substituted by 0, 1, 2 or 3 substituents.
  • a pharmaceutical composition comprising the compound according to any one of embodiments 1-4 or 9-69 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, and a pharmaceutically-acceptable diluent or carrier.
  • CDI carbonyldiimidazole
  • DABCO 1,4-diazabicyclo[2.2.2]octane
  • DCM diichloromethane
  • DIPEA diisopropyl ethylamine
  • DMSO dimethyl sulfoxide
  • n-BuLi n-butyllithium
  • t-BuLi t-butyllithium
  • TFA trifluoroacetic acid
  • h hour min—min rt—room temperature (22-25° C.)
  • h hour min—min rt—room temperature (22-25° C.)
  • h hour min—min rt—room temperature (22-25° C.)
  • h hour min—min rt—room temperature (22-25° C.)
  • grams ⁇ g micrograms mg milligrams ⁇ moL micromolars
  • Diarylamines used for the synthesis of compounds of the present invention were prepared as described in Scheme 1.
  • 2-Formylpyridines of the formula (1) were treated with 2-methylpropane-2-sulfinamide and copper sulfate in DCM to give 2-methyl-N-(pyridin-2-ylmethylene)propane-2-sulfinamides of the formula (2a).
  • the compounds of formula (2a) were treated with aryl or heteroaryl metal halides of formula (3) at low temperature to give sulfinamides of the formula (4).
  • Hydrolysis of sulfinamides (4) with hydrochloric acid in MeOH gives diaryl amines of formula (5a).
  • diaryl amines of formula (5a) An alternative approach to diaryl amines of formula (5a) is shown in Scheme 2.
  • Aryl or heteroaryl aldehydes of the formula (6) were treated with 2-methylpropane-2-sulfinamide and copper sulfate in DCM to give sulfinimines of the formula (7).
  • the compounds of formula (7) were treated with aryl or heteroaryl metal halides of formula (8) at low temperature to give sulfinamides of the formula (4).
  • Hydrolysis of sulfinamides (4) with hydrochloric acid in MeOH gives diaryl amines of formula (5a).
  • the vial was allowed to cool, diluted with hexanes (5 mL) and loaded directly to a normal phase silica gel column (80 g ISCO, 0 to 40% EtOAc in hexanes) to provide (S)-tert-butyl ((3-(prop-1-yn-1-yl)pyridin-2-yl)(4-(trifluoromethyl)phenyl)methyl)carbamate as a white solid.
  • the resulting product was dissolved in DMSO/MeOH (1/1) (2.0 mL) and loaded on a Gilson HPLC system for purification using a MeCN/H 2 O 0.1% TFA gradient and Phenomenx Gemini Axia-5 ⁇ C-18 column (150 ⁇ 30 mm). The solvent was removed from the pure fractions in the GENEVAC and the residue was dried under high vacuum to yield the title compound as an amorphous off-white solid.
  • reaction was then diluted with DMF (2 mL), filtered through a syringe filter, and purified by reverse phase HPLC (Phenomenx Gemini AxiaTM-5 ⁇ C-18 column (150 ⁇ 30 mm) 10-100% MeCN/0.1% TFA in H 2 O). The product-containing fractions were combined, and the solvent was removed by lyophilization to give the title compound as a white solid.
  • phenyl(4-(trifluoromethyl)phenyl)methanone (1.0105 g, 4.039 mmol) and methoxyamine (0.4208 g, 5.038 mmol) were dissolved in pyridine (10 mL). The mixture was stirred at rt overnight. Additional methoxyamine (0.1625 g) was added, and the solution was stirred at rt for 7 h. EtOAc was added and the organic phase was washed with water, 1 N HCl (2 ⁇ ), water, and then with brine. The organic phase was dried over sodium sulfate, filtered and concentrated in vacuo to afford a clear oil. The oil thus obtained was purified by Biotage (EtOAc/hexanes 0-20%) to afford the title compound (1.12 g, 99.4%) as a clear oil.
  • phenyl(4-(trifluoromethyl)phenyl)methanone O-methyl oxime (0.1541 g, 0.552 mmol) and Pd/C (10 wt %, 0.0345 g, 0.324 mmol) were mixed into MeOH (2 mL).
  • the reaction mixture was evacuated under vacuum and refilled with hydrogen (2 ⁇ ).
  • the mixture was hydrogenated under balloon pressure of hydrogen at rt for 45 min.
  • 2 M NH 3 in MeOH (1 mL) was added and the hydrogenation was resumed under balloon pressure of hydrogen for 1 h.
  • the Pd catalyst was removed via filtration through a pad of Celite® brand filter agent.
  • the resulting mixture was stirred at rt for 16 h.
  • the reaction was then heated at 70° C. for 5 h.
  • the reaction was next treated with another equivalent of catalyst, ligand and zinc reagent and heating was continued. After a further 5 days of heating, the reaction was filtered through a plug of silica gel and the plug washed with 50% EtOAc/hexanes.
  • Triethylborane (0.1 mL, 0.1 mmol, 1 M solution in hexane) was added to a DCM (5 mL) solution of N-allyl iodoacetamide (0.225 g, 1.0 mmol) and boron trifluoride dihydrate (0.25 g, 3.0 mmol) at rt. The resulting mixture was then stirred at this temperature for 2 h. The solution was then concentrated under reduced pressure, and the residue was refluxed gently with hydrochloric acid (20 mL, 1 N aqueous) for 3 h. The resulting mixture was then concentrated in vacuo, and the residue was dissolved in anhydrous MeOH (15 mL).
  • reaction product was adsorbed onto a plug of silica gel and chromatographed through a Redi-Sep® pre-packed silica gel column (4 g), eluting with 70% to 90% EtOAc in hexane, to provide the title compound as a white solid.
  • Absolute stereochemistries were determined by comparison of either (I) quantum-mechanically predicted optical rotation values (Stephens, P. J. et. al, J. Phys. Chem. A 2001, 105, 5356) or (II) VCD spectra (Stephens, P. J. et. al, Chirality 2008, 20, 643) to those measured experimentally (or, in the case of Example 28, both).
  • a single-crystal X-ray structure of (S)-tert-butyl((3-bromopyridin-2-yl)(4-(trifluoromethyl)phenyl)methyl)carbamate (Intermediate 40 Step 5; Example 173) provided confirmation of the absolute stereochemistry.
  • a stable Chinese hamster ovary cell line expressing human TRPM8 was generated using tetracycline inducible T-RExTM expression system from Invitrogen, Inc. (Carlsbad, Calif.). In order to enable a luminescence readout based on intracellular increase in calcium (Le Poul et al., 2002), the cell line was also co-transfected with pcDNA3.1 plasmid containing jelly fish aequorin cDNA. Twenty four hours before the assay, cells were seeded in 96-well plates and TRPM8 expression was induced with 0.5 ⁇ g/mL tetracycline.
  • the total number of jumps was recorded during the 10 minutes post-icilin administration based on the number of photocell beam breaks from the vertical array of open field boxes (Kinder Scientific) while movement of the mice was restricted within a clear Plexiglas® cylinder 9.5 cm diameter ⁇ 30 cm height. Resultant data is shown in Table 10.
  • LABORAS automation Methodris
  • CPT Cold Pressor Test
  • the cold pressor test was developed as a method to measure blood pressure response following exposure to a cold stimulus and has been used over 70 years in the diagnosis of hypertension and other cardiac autonomic disorders (Hines and Brown 1936).
  • the CPT is typically performed by immersing a subject's hand into ice water (0-5° C.) which triggers, through a vascular sympathetic activation of afferent pain and temperature neurons, an increase in blood pressure.
  • this test has also been utilized in rat to delineate the medullary and spinal pathways mediating the cardio-vascular responses to cold pressor test and to identify neurotransmitters in these pathways (Sapru N et al 2008) or to characterize analgesic compounds (Koltzenburg M et al 2006 and Player M R et al 2011).
  • TRPM8 antagonists can be evaluated in rat CPT to determine whether TRPM8 antagonists would attenuate the increase in blood pressure resulting from exposure to cold stimulation of the paws and ventral half of the body.
  • Male Sprague-Dawley rats weighing 350-450 g can be instrumented with a unilateral carotid artery-cannula connected to a transducer for measuring blood pressure using a Digi-Med Blood Pressure Analyzer, Model 400 Animals can be orally administrated with Vehicle (2% HPMC 1% Tween 80 pH 2.2 with MSA) or test compounds at 120 minutes prior to cold challenge and anesthetized with sodium pentobarbital at 60 mg/kg ip at 100 minutes prior to cold.
  • Vehicle 2% HPMC 1% Tween 80 pH 2.2 with MSA
  • Plasma can be collected through artery catheter immediately after CPT for pk analysis and IC50/90 determination.
  • CCI chronic constriction injury
  • Behavioral testing A behavioral test can be performed to estimate cold-induced ongoing pain as previously described (Choi et al., 1994).
  • the rat can be placed under a transparent plastic cover on an aluminum plate (IITC PE34, Woodland, Calif.) which can be kept at a cold temperature (5 ⁇ 0.5° C.). After 2 minutes of adaptation, the cumulative duration of time that the rat lifts its foot off the plate for the next 5 minutes can be measured. Foot lifts associated with locomotion or grooming are not counted. Seven to 9 days after the CCI surgery, baseline of the cold-induced ongoing pain can be measured. Any rat showing a cold-induced ongoing pain less than 100 sec out of 300 sec observation period can be eliminated from the study.
  • test compound Twenty four hours after the baseline measurement, test compound, positive control, morphine (2 mg/kg, Sigma, St. Louis) or a vehicle (saline or 2% HPMC/1% Tween 80) can be administered orally (test compound) or subcutaneously (morphine). Two hrs (test compound) or 30 mins (morphine) after the drug administration, the cold-induced ongoing pain can be measured again.
  • spinal nerve ligation surgery can be performed as previously described (Kim & Chung, 1992). Briefly, under gaseous anesthesia with a mixture of isoflurane (3% for induction and 2% for maintenance) in O 2 , the spinal nerve injury can be produced by ligating the left L5 and L6 spinal nerves taking special care to avoid any possible damage to the L4 spinal nerve or surrounding area. Additional treatments can be performed to increase the development of mechanical allodynia. First, L5 spinal nerve can be cut approximately 1 mm distal to the suture as described by Li et al. (2000).
  • the L4 spinal nerve can be lightly manipulated by slightly stretching it with a fine hooked glass rod and gently sliding the hook back and forth 20 times along the nerve as described by Lee et al. (2003).
  • the whole surgery procedure from anesthesia to the clipping of the incised skin can take at most 15 minutes.
  • the von Frey hair can be presented perpendicular to the plantar surface with sufficient force to cause slight bending, and held for approximately 3-4 seconds. Abrupt withdrawal of the foot (paw flinching, shaking or licking for more than 1 sec.) can be recorded as a response. Any rat showing a mechanical threshold of more than 3.16 g or less than 0.7 g after surgery can be eliminated from the study. After measuring basal threshold, test compound, positive control gabapentin (Sigma, St. Louis) or a vehicle (saline or 2% HPMC/1% Tween 80) can be administered orally (test compound) or intraperitoneally (gabapentin). The measurement of the tactile threshold can be reassessed at 1.5 and 2 hrs after drug administration.
  • TRPM8-receptor-diseases such as acute, inflammatory and neuropathic pain, dental pain, general headache, migraine, cluster headache, mixed-vascular and non-vascular syndromes, tension headache, general inflammation, arthritis, rheumatic diseases, osteoarthritis, inflammatory bowel disorders, inflammatory eye disorders, inflammatory or unstable bladder disorders, psoriasis, skin complaints with inflammatory components, chronic inflammatory conditions, inflammatory pain and associated hyperalgesia and allodynia, neuropathic pain and associated hyperalgesia and allodynia, diabetic neuropathy pain, causalgia, sympathetically maintained pain, deafferentation syndromes, asthma, epithelial tissue damage or dysfunction, herpes simplex, disturbances of visceral motility at respiratory, genitourinary, gastrointestinal or vascular regions, wounds, burns, allergic skin reactions, pruritus, vitiligo, general gastrointestinal disorders, gastric ulceration, duodenal ulcers, diarrhea, gastric lesions
  • Treatment of diseases and disorders herein is intended to also include the prophylactic administration of a compound of the invention, a pharmaceutical salt thereof, or a pharmaceutical composition of either to a subject (i.e., an animal, preferably a mammal, most preferably a human) believed to be in need of preventative treatment, such as, for example, pain, inflammation and the like.
  • a subject i.e., an animal, preferably a mammal, most preferably a human
  • preventative treatment such as, for example, pain, inflammation and the like.
  • the dosage regimen for treating TRPM8-receptor-mediated diseases, cancer, and/or hyperglycemia with the compounds of this invention and/or compositions of this invention is based on a variety of factors, including the type of disease, the age, weight, sex, medical condition of the patient, the severity of the condition, the route of administration, and the particular compound employed. Thus, the dosage regimen may vary widely, but can be determined routinely using standard methods. Dosage levels of the order from about 0.01 mg to 30 mg per kilogram of body weight per day, preferably from about 0.1 mg to 10 mg/kg, more preferably from about 0.25 mg to 1 mg/kg are useful for all methods of use disclosed herein.
  • the pharmaceutically active compounds of this invention can be processed in accordance with conventional methods of pharmacy to produce medicinal agents for administration to patients, including humans and other mammals.
  • the pharmaceutical composition may be in the form of, for example, a capsule, a tablet, a suspension, or liquid.
  • the pharmaceutical composition is preferably made in the form of a dosage unit containing a given amount of the active ingredient.
  • these may contain an amount of active ingredient from about 1 to 2000 mg, preferably from about 1 to 500 mg, more preferably from about 5 to 150 mg.
  • a suitable daily dose for a human or other mammal may vary widely depending on the condition of the patient and other factors, but, once again, can be determined using routine methods.
  • the active ingredient may also be administered by injection as a composition with suitable carriers including saline, dextrose, or water.
  • suitable carriers including saline, dextrose, or water.
  • the daily parenteral dosage regimen will be from about 0.1 to about 30 mg/kg of total body weight, preferably from about 0.1 to about 10 mg/kg, and more preferably from about 0.25 mg to 1 mg/kg.
  • Injectable preparations such as sterile injectable aqueous or oleaginous suspensions, may be formulated according to the known are using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1,3-butanediol.
  • a non-toxic parenterally acceptable diluent or solvent for example as a solution in 1,3-butanediol.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed, including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • Suppositories for rectal administration of the drug can be prepared by mixing the drug with a suitable non-irritating excipient such as cocoa butter and polyethylene glycols that are solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum and release the drug.
  • a suitable non-irritating excipient such as cocoa butter and polyethylene glycols that are solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum and release the drug.
  • a suitable topical dose of active ingredient of a compound of the invention is 0.1 mg to 150 mg administered one to four, preferably one or two times daily.
  • the active ingredient may comprise from 0.001% to 10% w/w, e.g., from 1% to 2% by weight of the formulation, although it may comprise as much as 10% w/w, but preferably not more than 5% w/w, and more preferably from 0.1% to 1% of the formulation.
  • Formulations suitable for topical administration include liquid or semi-liquid preparations suitable for penetration through the skin (e.g., liniments, lotions, ointments, creams, or pastes) and drops suitable for administration to the eye, ear, or nose.
  • liquid or semi-liquid preparations suitable for penetration through the skin e.g., liniments, lotions, ointments, creams, or pastes
  • drops suitable for administration to the eye, ear, or nose e.g., liniments, lotions, ointments, creams, or pastes
  • the compounds of this invention are ordinarily combined with one or more adjuvants appropriate for the indicated route of administration.
  • the compounds may be admixed with lactose, sucrose, starch powder, cellulose esters of alkanoic acids, stearic acid, talc, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulfuric acids, acacia, gelatin, sodium alginate, polyvinyl-pyrrolidine, and/or polyvinyl alcohol, and tableted or encapsulated for conventional administration.
  • the compounds of this invention may be dissolved in saline, water, polyethylene glycol, propylene glycol, ethanol, corn oil, peanut oil, cottonseed oil, sesame oil, tragacanth gum, and/or various buffers.
  • Other adjuvants and modes of administration are well known in the pharmaceutical art.
  • the carrier or diluent may include time delay material, such as glyceryl monostearate or glyceryl distearate alone or with a wax, or other materials well known in the art.
  • the pharmaceutical compositions may be made up in a solid form (including granules, powders or suppositories) or in a liquid form (e.g., solutions, suspensions, or emulsions).
  • the pharmaceutical compositions may be subjected to conventional pharmaceutical operations such as sterilization and/or may contain conventional adjuvants, such as preservatives, stabilizers, wetting agents, emulsifiers, buffers etc.
  • Solid dosage forms for oral administration may include capsules, tablets, pills, powders, and granules.
  • the active compound may be admixed with at least one inert diluent such as sucrose, lactose, or starch.
  • Such dosage forms may also comprise, as in normal practice, additional substances other than inert diluents, e.g., lubricating agents such as magnesium stearate.
  • the dosage forms may also comprise buffering agents. Tablets and pills can additionally be prepared with enteric coatings.
  • Liquid dosage forms for oral administration may include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art, such as water. Such compositions may also comprise adjuvants, such as wetting, sweetening, flavoring, and perfuming agents.
  • optical isomers can be obtained by resolution of the racemic mixtures according to conventional processes, e.g., by formation of diastereoisomeric salts, by treatment with an optically active acid or base.
  • appropriate acids are tartaric, diacetyltartaric, dibenzoyltartaric, ditoluoyltartaric, and camphorsulfonic acid and then separation of the mixture of diastereoisomers by crystallization followed by liberation of the optically active bases from these salts.
  • a different process for separation of optical isomers involves the use of a chiral chromatography column optimally chosen to maximize the separation of the enantiomers.
  • Still another available method involves synthesis of covalent diastereoisomeric molecules by reacting compounds of the invention with an optically pure acid in an activated form or an optically pure isocyanate.
  • the synthesized diastereoisomers can be separated by conventional means such as chromatography, distillation, crystallization or sublimation, and then hydrolyzed to deliver the enantiomerically pure compound.
  • the optically active compounds of the invention can likewise be obtained by using active starting materials. These isomers may be in the form of a free acid, a free base, an ester or a salt.
  • the compounds of this invention may exist as isomers, that is compounds of the same molecular formula but in which the atoms, relative to one another, are arranged differently.
  • the alkylene substituents of the compounds of this invention are normally and preferably arranged and inserted into the molecules as indicated in the definitions for each of these groups, being read from left to right.
  • substituents are reversed in orientation relative to the other atoms in the molecule. That is, the substituent to be inserted may be the same as that noted above except that it is inserted into the molecule in the reverse orientation.
  • these isomeric forms of the compounds of this invention are to be construed as encompassed within the scope of the present invention.
  • the compounds of the present invention can be used in the form of salts derived from inorganic or organic acids.
  • the salts include, but are not limited to,
  • the basic nitrogen-containing groups can be quaternized with such agents as lower alkyl halides, such as methyl, ethyl, propyl, and butyl chloride, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides, and others. Water or oil-soluble or dispersible products are thereby obtained.
  • lower alkyl halides such as methyl, ethyl, propyl, and butyl chloride, bromides and iodides
  • dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates
  • long chain halides such as de
  • organic acids such as oxalic acid, maleic acid, succinic acid and citric acid.
  • Other examples include salts with alkali metals or alkaline earth metals, such as sodium, potassium, calcium or magnesium or with organic bases.
  • esters of a carboxylic acid or hydroxyl containing group including a metabolically labile ester or a prodrug form of a compound of this invention.
  • a metabolically labile ester is one which may produce, for example, an increase in blood levels and prolong the efficacy of the corresponding non-esterified form of the compound.
  • a prodrug form is one which is not in an active form of the molecule as administered but which becomes therapeutically active after some in vivo activity or biotransformation, such as metabolism, for example, enzymatic or hydrolytic cleavage.
  • esters for example, methyl, ethyl
  • cycloalkyl for example, cyclohexyl
  • aralkyl for example, benzyl, p-methoxybenzyl
  • alkylcarbonyloxyalkyl for example, pivaloyloxymethyl
  • Amines have been masked as arylcarbonyloxymethyl substituted derivatives which are cleaved by esterases in vivo releasing the free drug and formaldehyde (Bungaard J. Med. Chem. 2503 (1989)). Also, drugs containing an acidic NH group, such as imidazole, imide, indole and the like, have been masked with N-acyloxymethyl groups (Bundgaard Design of Prodrugs, Elsevier (1985)). Hydroxy groups have been masked as esters and ethers.
  • EP 039,051 (Sloan and Little, Apr. 11, 1981) discloses Mannich-base hydroxamic acid prodrugs, their preparation and use.
  • Esters of a compound of this invention may include, for example, the methyl, ethyl, propyl, and butyl esters, as well as other suitable esters formed between an acidic moiety and a hydroxyl containing moiety.
  • Metabolically labile esters may include, for example, methoxymethyl, ethoxymethyl, iso-propoxymethyl, ⁇ -methoxyethyl, groups such as ⁇ -((C 1 -C 4 )alkyloxy)ethyl, for example, methoxyethyl, ethoxyethyl, propoxyethyl, iso-propoxyethyl, etc.; 2-oxo-1,3-dioxolen-4-ylmethyl groups, such as 5-methyl-2-oxo-1,3,dioxolen-4-ylmethyl, etc.; C 1 -C 3 alkylthiomethyl groups, for example, methylthiomethyl, ethylthiomethyl,
  • the compounds of the invention may exist as crystalline solids which can be crystallized from common solvents such as ethanol, N,N-dimethylformamide, water, or the like.
  • crystalline forms of the compounds of the invention may exist as polymorphs, solvates and/or hydrates of the parent compounds or their pharmaceutically acceptable salts. All of such forms likewise are to be construed as falling within the scope of the invention.
  • the compounds of the invention can be administered as the sole active pharmaceutical agent, they can also be used in combination with one or more compounds of the invention or other agents.
  • the therapeutic agents can be formulated as separate compositions that are given at the same time or different times, or the therapeutic agents can be given as a single composition.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Pulmonology (AREA)
  • Biomedical Technology (AREA)
  • Pain & Pain Management (AREA)
  • Dermatology (AREA)
  • Diabetes (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Virology (AREA)
  • Rheumatology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Endocrinology (AREA)
  • Urology & Nephrology (AREA)
  • Immunology (AREA)
  • Oncology (AREA)
  • Cardiology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Emergency Medicine (AREA)
  • Obesity (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Psychiatry (AREA)
  • Hematology (AREA)
  • Reproductive Health (AREA)
  • Communicable Diseases (AREA)
  • Molecular Biology (AREA)

Abstract

Compounds of Formula I are useful as antagonists of TRPM8. Such compounds are useful in treating a number of TRPM8 mediated disorders and conditions and may be used to prepare medicaments and pharmaceutical compositions useful for treating such disorders and conditions. Examples of such disorders include, but are not limited to, migraines and neuropathic pain. Compounds of Formula I have the following structure:
Figure US20140235642A1-20140821-C00001
where the definitions of the variables are provided herein.

Description

    CROSS REFERENCES TO RELATED APPLICATIONS
  • This application is a divisional of, and claims priority to, U.S. patent application Ser. No. 13/529,880, filed on Jun. 21, 2012, pending, which claims the benefit of, and priority to, U.S. Provisional Application No. 61/500,835, filed on Jun. 24, 2011, which is hereby incorporated by reference in its entirety and for all purposes as if fully set forth herein.
  • FIELD OF THE INVENTION
  • The present invention relates to compounds that have TRPM8 antagonist properties and are useful in preparing medicaments and compositions and in treating diseases and conditions such as those mediated by TRPM8. The compounds and compositions may be used to treat various diseases or conditions modulated by TRPM8 such as, but not limited to, migraines and neuropathic pain.
  • BACKGROUND OF THE INVENTION
  • Cold sensation is derived from activation of the somatosensory system by a cold stimulus. Calcium imaging and patch clamp experiments in dissociated trigeminal and dorsal root ganglia neurons have revealed cold stimuli induced calcium influx, suggesting the direct opening of a calcium-permeable ion channels by cold (Thut et al., 2003; Reid, 2005). A recently cloned non-selective cation channel, TRPM8 (transient receptor potential melastatin 8) or trp-p8 (identified as a prostate-specific gene, up-regulated in prostate cancer and other malignancies, (Tsavaler et al., 2001)) is activated by cold stimulus of 10 to 24° C. temperature (McKemy et al., 2002; Peier et al., 2002). In addition, TRPM8 is also activated by compounds that elicit cool sensation such as menthol, icilin (AG-3-5) (McKemy et al., 2002), and the endogenous lipid PIP2 (Rohacs et al., 2005). Correlating with the cold sensitivity of both A delta and C-fibers, TRPM8 is highly expressed in sensory neurons of the trigeminal and dorsal root ganglia (McKemy et al., 2002; Peier et al., 2002; Thut et al., 2003). TRPM8 is also expressed in nerve fibers innervating urinary bladder in guinea pigs (Tsukimi et al., 2005) and humans (Mukerji et al., 2006) and believed to contribute to the bladder hypersensitivity.
  • Activation mechanism of TRPM8 by menthol and icilin appears to differ. Icilin requires calcium for robust activation of TRPM8, whereas menthol and cold do not (Chuang et al., 2004). Typically, activation by all these agonists follows a period of calcium-dependent desensitization. The domain swap analysis of chicken and rat TRPM8 and further mutational studies revealed that determinants of icilin sensitivity map to a region of TRPM8 that corresponds to the capsaicin binding site in TRPV1 transmembrane domain 3 to 4 region (Chuang et al., 2004).
  • Cold allodynia and mechanical hyperalgesia are associated with neuropathic pain in humans and in rodent models of neuropathic and chemotherapy-induced pain. TRPM8 is shown to mediate the analgesia by agonists such as menthol and icilin (by desensitization of the receptor) during experimental neuropathic pain in rodents (Proudfoot et al., 2006). Further, attenuation of cold sensation and cold allodynia after chronic constriction injury model of neuropathic pain in TRPM8 knockout mice (Colburn et al., 2007; Dhaka et al., 2007) suggests that antagonists of TRPM8 may be considered as pain therapeutics for chemotherapy-induced pain, neuropathic pain and bladder disorders.
  • Mint oil that contains menthol, an agonist of TRPM8 has been reported to alleviate pain in post-herpetic neuralgia (Davies et al., 2002), a neuropathic pain condition. Furthermore, oral or intracerebroventricular injection of menthol decreased nociceptive responses to hot-plate test and acetic acid-induced writhing in mice (Galeotti et al., 2002). These responses are believed to be mediated by the activation and desensitization of the TRPM8. These observations and the knockout mice studies indicate that TRPM8 modulation by antagonists might be beneficial for patients experiencing neuropathic pain.
  • A need exists for TRPM8 antagonist compounds that can be used to treat diseases and conditions mediated by TRPM8 such as, but not limited to, migraines and neuropathic pain and those other conditions described herein.
  • SUMMARY OF THE INVENTION
  • The present invention comprises a new class of compounds useful in the treatment of diseases, such as TRPM8-mediated diseases and other maladies, such as inflammatory or neuropathic pain and diseases involving sensory nerve function such as asthma, rheumatoid arthritis, osteoarthritis, inflammatory bowel disorders, urinary incontinence, migraine and psoriasis. In particular, the compounds of the invention are useful for the treatment of acute, inflammatory and neuropathic pain, dental pain, general headache, migraine, cluster headache, mixed-vascular and non-vascular syndromes, tension headache, general inflammation, arthritis, rheumatic diseases, osteoarthritis, inflammatory bowel disorders, anxiety, depression, inflammatory eye disorders, inflammatory or unstable bladder disorders, psoriasis, skin complaints with inflammatory components, chronic inflammatory conditions, inflammatory pain and associated hyperalgesia and allodynia, neuropathic pain and associated hyperalgesia and allodynia, diabetic neuropathy pain, causalgia, sympathetically maintained pain, deafferentation syndromes, asthma, epithelial tissue damage or dysfunction, herpes simplex, disturbances of visceral motility at respiratory, genitourinary, gastrointestinal or vascular regions, wounds, burns, allergic skin reactions, pruritus, vitiligo, general gastrointestinal disorders, gastric ulceration, duodenal ulcers, diarrhea, gastric lesions induced by necrotising agents, hair growth, vasomotor or allergic rhinitis, bronchial disorders or bladder disorders. Accordingly, the invention also comprises pharmaceutical compositions comprising the compounds, methods for the treatment of TRPM8-receptor-mediated diseases, such as inflammatory or neuropathic pain, asthma, rheumatoid arthritis, osteoarthritis, inflammatory bowel disorders, urinary incontinence, migraine and psoriasis diseases, using the compounds and compositions of the invention, and intermediates and processes useful for the preparation of the compounds of the invention.
  • The compounds of the invention are represented by the following general structure:
  • Figure US20140235642A1-20140821-C00002
  • or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, R5, R6, Y, X1, X2, m and n are defined below.
  • In one aspect, the invention provides a compound of Formula I having the structure:
  • Figure US20140235642A1-20140821-C00003
  • or a pharmaceutically-acceptable salt thereof, a tautomer thereof, a pharmaceutically-acceptable salt of the tautomer, a stereoisomer thereof, or a mixture thereof, wherein:
  • m is 0, 1, 2 or 3;
  • n is 0 or 1;
  • X1 is C(R4) or N;
  • X2 is CH or N;
  • Y is NH, NR1a, or O;
  • Z is O or S;
  • R1 is C1-6alk, —(C═O)—O—C1-6alk, or a direct-bonded, C1-6alk-linked, C1-2alkO-linked, —C(═O)—O linked, —C(═O)— linked, saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 heteroatoms selected from N, O and S, the C1-6alk, the C1-6alk of the C1-6alk-link, and the monocyclic or bicyclic ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C1-6alk, C1-6alkOH, C1-6alk-C(═O)Ra, C1-6alk-C(═O)ORa, C1-4haloalk, cyano, nitro, —C(═O)Ra, —C(═O)ORa, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Ra, —OC(═O)NRaRa, —OC(═O)N(Ra)S(═O)2Ra, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, ═S, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRaRa, —S(═O)2N(Ra)C(═O)Ra, —S(═O)2N(Ra)C(═O)ORa, —S(═O)2N(Ra)C(═O)NRaRa, —NRaRa, —N(Ra)C(═O)Ra, —N(Ra)C(═O)ORa, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Ra, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa, wherein the ring is additionally substituted by 0 or 1 directly bonded, SO2 linked, C(═O) linked or CH2 linked saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic ring containing 0, 1, 2, 3 or 4 heteroatoms selected from N, O and S, and substituted by 0, 1, 2 or 3 groups selected from halo, C1-6alk, C1-4haloalk, cyano, oxo, nitro, —C(═O)Ra, —C(═O)ORa, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Ra, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRaRa, —NRaRa, and —N(Ra)C(═O)Ra; or R1a and R1, together with the N atom to which they are attached, form a saturated, partially-saturated or unsaturated 4-, 5-, 6-, or 7-membered monocyclic or a 9- or 10-membered bicylic ring containing 0, 1, or 2 additional heteroatoms independently selected from N, O, and S, wherein the ring formed by R1a and R1 is substituted with 0, 1, or 2 substituents independently selected from halo, oxo, C1-6alk, C1-6alkOH, C1-6alk-C(═O)Ra, C1-6alk-C(═O)ORa, C1-4haloalk, cyano, nitro, —C(═O)Ra, —C(═O)ORa, or —C(═O)NRaRa;
  • R2 is H, halo, cyano, Rc, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa; or R2 is C1-6alk substituted by 0, 1, 2 or 3 substituents selected from C1-4haloalk, halo, cyano, nitro, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa, or R2 is C1-6alk substituted by 0, 1, 2 or 3 halo substituents and additionally substituted by 0 or 1 substituents selected from Rc;
  • R3 is H, C1-8alk, C1-8alkOH, C1-4haloalk, halo, cyano, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa or —NRaC2-6alkORa;
  • R4 is independently, at each instance, H, C1-6alk, —C1-3haloalk, —OC1-6alk, —OC1-3haloalk, —N(C1-6alk)C1-6alk, —NHC1-6alk, —NC(═O)C1-6alk, —N(C1-6alk)C1-6alk, F, Cl, Br, CN, OH or NH2; or R3 and R4 together form a four-atom unsaturated bridge containing 0 or 1 N atoms, wherein the bridge is substituted by 0, 1 or 2 R5 substituents;
  • R5 is independently, in each instance, Cl, Br, F, CH3, CF3, or ORa;
  • R6 is F, CF3, C1-6alk, or ORa;
  • Ra is independently, at each instance, H or Rb;
  • Rb is independently, at each instance, phenyl, benzyl or C1-6alk, the phenyl, benzyl and C1-6alk being substituted by 0, 1, 2 or 3 substituents selected from halo, oxo, C1-4alk, C1-3haloalk, —OC1-4alk, —OH, —NH2, —OC1-4alk, —OC1-4haloalk, —NHC1-4alk, and —N(C1-4alk)C1-4alk; and
  • Rc is independently, at each instance, a saturated, partially saturated or unsaturated 4-, 5- or 6-membered monocyclic ring containing 0, 1, 2, 3 or 4 heteroatoms selected from N, O and S, wherein the C1-6alkyl and ring are substituted by 0 or 1 oxo groups substituted by 0, 1, 2 or 3 substituents selected from C1-8alk, C1-4haloalk, halo and cyano. In some such embodiments, Y is selected from NH or O. In some such embodiments, Y is NH whereas in other such embodiments, Y is O.
  • In one embodiment, the compound of Formula I has the Formula IA:
  • Figure US20140235642A1-20140821-C00004
  • In another embodiment, the compound of Formula I has the Formula IB:
  • Figure US20140235642A1-20140821-C00005
  • where Y is NH or O.
  • In another embodiment, the compound of Formula I has the Formula IC:
  • Figure US20140235642A1-20140821-C00006
  • or any pharmaceutically-acceptable salt thereof, wherein:
  • m is 0, 1, 2 or 3;
  • n is 0 or 1;
  • X1 is C(R4) or N;
  • X2 is CH or N;
  • R1 is selected from C1-6alk or a direct-bonded, C1-2alk-linked, C1-2alkO-linked, saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 heteroatoms selected from N, O and S, but containing no more than one O or S atom, the C1-6alk and ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C1-6alk, C1-4haloalk, cyano, nitro, —C(═O)Ra, —C(═O)ORa, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Ra, —OC(═O)NRaRa, —OC(═O)N(Ra)S(═O)2Ra, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRaRa, —S(═O)2N(Ra)C(═O)Ra, —S(═O)2N(Ra)C(═O)ORa, —S(═O)2N(Ra)C(═O)NRaRa, —NRaRa, —N(Ra)C(═O)Ra, —N(Ra)C(═O)ORa, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Ra, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa, wherein the ring is additionally substituted by 0 or 1 directly bonded, SO2 linked, C(═O) linked or CH2 linked saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic ring substituted by 0, 1, 2 or 3 groups selected from halo, C1-6alk, C1-4haloalk, cyano, nitro, —C(═O)Ra, —C(═O)ORa, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Ra, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRaRa, —NRaRa, and —N(Ra)C(═O)Ra;
  • R2 is selected from H, halo, cyano, Rc, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa; or R2 is C1-6alk substituted by 0, 1, 2 or 3 substituents selected from C1-4haloalk, halo, cyano, nitro, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa, or R2 is C1-6alk substituted by 0, 1, 2 or 3 halo substituents and additionally substituted by 0 or 1 substituents selected from Rc;
  • R3 is H, C1-8alk, C1-4haloalk, halo, cyano, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa or —NRaC2-6alkORa;
  • R4 is independently, at each instance, H, C1-6alk, —C1-3haloalk, —OC1-6alk, —OC1-3haloalk, —N(C1-6alk)C1-6alk, —NHC1-6alk, —NC(═O)C1-6alk, —N(C1-6alk)C1-6alk, F, Cl, Br, CN, OH or NH2; or R3 and R4 together form a four-atom unsaturated bridge containing 0 or 1 N atoms, wherein the bridge is substituted by 0, 1 or 2 R5 substituents;
  • R5 is independently, in each instance, F, CH3 or CF3;
  • R6 is F;
  • Ra is independently, at each instance, H or Rb;
  • Rb is independently, at each instance, phenyl, benzyl or C1-6alk, the phenyl, benzyl and C1-6alk being substituted by 0, 1, 2 or 3 substituents selected from halo, oxo, C1-4alk, C1-3haloalk, —OC1-4alk, —OH, —NH2, —OC1-4alk, —OC1-4haloalk, —NHC1-4alk, and —N(C1-4alk)C1-4alk; and
  • Rc is independently, at each instance, a saturated, partially saturated or unsaturated 4-, 5- or 6-membered monocyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, wherein the C1-6alkyl and ring are substituted by 0 or 1 oxo groups substituted by 0, 1, 2 or 3 substituents selected from C1-8alk, C1-4haloalk, halo and cyano.
  • In another embodiment, the compound of Formula I has the Formula ID:
  • Figure US20140235642A1-20140821-C00007
  • where Y is NH or O.
  • In some embodiments, the invention provides a compound or tautomer of any of the embodiments in a neutral form. In some such embodiments, the invention provides a compound of any of the embodiments in a neutral form.
  • In other embodiments, the invention provides a pharmaceutically-acceptably salt of the compound or a pharmaceutically acceptable salt of the tautomer of any one of the embodiments. In other embodiments, the invention provides a pharmaceutically-acceptably salt of the compound of any one of the embodiments. In some such embodiments, the salt is a trifluoroacetate or a bis trifluoroacetate salt.
  • The invention also provides pharmaceutical compositions that include the compound of any one of the embodiments or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof. Such compositions typically include a pharmaceutically-acceptable diluent or carrier.
  • The invention further provides methods of treating acute, inflammatory and neuropathic pain, dental pain, general headache, migraine, cluster headache, mixed-vascular and non-vascular syndromes, tension headache, general inflammation, arthritis, rheumatic diseases, osteoarthritis, inflammatory bowel disorders, depression, anxiety, inflammatory eye disorders, inflammatory or unstable bladder disorders, psoriasis, skin complaints with inflammatory components, chronic inflammatory conditions, inflammatory pain and associated hyperalgesia and allodynia, neuropathic pain and associated hyperalgesia and allodynia, diabetic neuropathy pain, causalgia, sympathetically maintained pain, deafferentation syndromes, asthma, epithelial tissue damage or dysfunction, herpes simplex, disturbances of visceral motility at respiratory, genitourinary, gastrointestinal or vascular regions, wounds, burns, allergic skin reactions, pruritus, vitiligo, general gastrointestinal disorders, gastric ulceration, duodenal ulcers, diarrhea, gastric lesions induced by necrotising agents, hair growth, vasomotor or allergic rhinitis, bronchial disorders or bladder disorders in a subject. Such methods typically include administering the compound according to any of the embodiments or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof to the subject. In some such methods the subject is suffering from neuropathic pain whereas in other such embodiments, the subject is suffering from migraine pain.
  • The invention also provides the use of the compound of any of the embodiments or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof in the preparation of a medicament.
  • The invention still further provides the use of the compound of any of the embodiments or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof for treating acute, inflammatory and neuropathic pain, dental pain, general headache, migraine, cluster headache, mixed-vascular and non-vascular syndromes, tension headache, general inflammation, arthritis, rheumatic diseases, osteoarthritis, inflammatory bowel disorders, depression, anxiety, inflammatory eye disorders, inflammatory or unstable bladder disorders, psoriasis, skin complaints with inflammatory components, chronic inflammatory conditions, inflammatory pain and associated hyperalgesia and allodynia, neuropathic pain and associated hyperalgesia and allodynia, diabetic neuropathy pain, causalgia, sympathetically maintained pain, deafferentation syndromes, asthma, epithelial tissue damage or dysfunction, herpes simplex, disturbances of visceral motility at respiratory, genitourinary, gastrointestinal or vascular regions, wounds, burns, allergic skin reactions, pruritus, vitiligo, general gastrointestinal disorders, gastric ulceration, duodenal ulcers, diarrhea, gastric lesions induced by necrotising agents, hair growth, vasomotor or allergic rhinitis, bronchial disorders or bladder disorders in a subject. In some such embodiments, the use is for treating neuropathic pain whereas in other such embodiments, the use is for treating migraine.
  • The invention also provides the compound according to any of the embodiments described herein or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof for treating acute, inflammatory and neuropathic pain, dental pain, general headache, migraine, cluster headache, mixed-vascular and non-vascular syndromes, tension headache, general inflammation, arthritis, rheumatic diseases, osteoarthritis, inflammatory bowel disorders, depression, anxiety, inflammatory eye disorders, inflammatory or unstable bladder disorders, psoriasis, skin complaints with inflammatory components, chronic inflammatory conditions, inflammatory pain and associated hyperalgesia and allodynia, neuropathic pain and associated hyperalgesia and allodynia, diabetic neuropathy pain, causalgia, sympathetically maintained pain, deafferentation syndromes, asthma, epithelial tissue damage or dysfunction, herpes simplex, disturbances of visceral motility at respiratory, genitourinary, gastrointestinal or vascular regions, wounds, burns, allergic skin reactions, pruritus, vitiligo, general gastrointestinal disorders, gastric ulceration, duodenal ulcers, diarrhea, gastric lesions induced by necrotising agents, hair growth, vasomotor or allergic rhinitis, bronchial disorders or bladder disorders in a subject. In some such embodiments, the compound of any of the embodiments or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof is for treating neuropathic pain in a subject. In other such embodiments, the compound of any of the embodiments or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof is for treating migraine pain in a subject
  • The foregoing merely summarizes certain aspects of the invention and is not intended, nor should it be construed, as limiting the invention in any way. All patents, patent applications and other publications recited herein are hereby incorporated by reference in their entirety.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The compounds of this invention may have in general several asymmetric centers and are typically depicted in the form of racemic mixtures. This invention is intended to encompass racemic mixtures, partially racemic mixtures and separate enantiomers and diasteromers.
  • Unless otherwise specified, the following definitions apply to terms found in the specification and claims:
  • “Cα-βalk” means an alkyl group comprising a minimum of α and a maximum of β carbon atoms in a branched, cyclical or linear relationship or any combination of the three, wherein α and β represent integers. The alkyl groups described in this section may also contain one or two double or triple bonds. A designation of C0alk indicates a direct bond. Examples of C1-6alkyl include, but are not limited to the following:
  • Figure US20140235642A1-20140821-C00008
  • Where the term “Cα-βalkyl” and “Cα-βcycloalkyl” are used, they relate to acyclic saturated alkyls and cyclic saturated alkyls, respectively.
  • “Benzo group”, alone or in combination, means the divalent radical C4H4═, one representation of which is —CH═CH—CH═CH—, that when vicinally attached to another ring forms a benzene-like ring—for example tetrahydronaphthylene, indole and the like.
  • The terms “oxo” and “thioxo” represent the groups ═O (as in carbonyl) and ═S (as in thiocarbonyl), respectively.
  • The term “cyano” refers to a nitrile group which may be written as —C≡N. “Halo” or “halogen” means a halogen atoms selected from F, Cl, Br and I. “CV-Whaloalk” means an alk group, as described above, wherein any number—at least one—of the hydrogen atoms attached to the alk chain are replaced by F, Cl, Br or I.
  • The group N(Ra)Ra and the like include substituents where the two Ra groups together form a ring, optionally including a N, O or S atom, and include groups such as:
  • Figure US20140235642A1-20140821-C00009
  • The group N(Cα-βalk)Cα-βalk, wherein α and β are as defined above, include substituents where the two Cα-βalk groups together form a ring, optionally including a N, O or S atom, and include groups such as:
  • Figure US20140235642A1-20140821-C00010
  • “Heterocycle” means a ring comprising at least one carbon atom and at least one other atom selected from N, O and S. Examples of heterocycles that may be found in the claims include, but are not limited to, the following:
  • Figure US20140235642A1-20140821-C00011
    Figure US20140235642A1-20140821-C00012
  • “Pharmaceutically-acceptable salt” means a salt prepared by conventional means, and are well known by those skilled in the art. The “pharmacologically acceptable salts” include basic salts of inorganic and organic acids, including but not limited to hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, malic acid, acetic acid, trifluoroacetic acid, oxalic acid, tartaric acid, citric acid, lactic acid, fumaric acid, succinic acid, maleic acid, salicylic acid, benzoic acid, phenylacetic acid, mandelic acid and the like. When compounds of the invention include an acidic function such as a carboxy group, then suitable pharmaceutically acceptable cation pairs for the carboxy group are well known to those skilled in the art and include alkaline, alkaline earth, ammonium, quaternary ammonium cations and the like. For additional examples of “pharmacologically acceptable salts,” see infra and Berge et al., J. Pharm. Sci. 66:1 (1977).
  • “Saturated, partially-saturated or unsaturated” includes substituents saturated with hydrogens, substituents completely unsaturated with hydrogens and substituents partially saturated with hydrogens.
  • “Leaving group” generally refers to groups readily displaceable by a nucleophile, such as an amine, a thiol or an alcohol nucleophile. Such leaving groups are well known in the art. Examples of such leaving groups include, but are not limited to, N-hydroxysuccinimide, N-hydroxybenzotriazole, halides, triflates, tosylates and the like. Preferred leaving groups are indicated herein where appropriate.
  • “Protecting group” generally refers to groups well known in the art which are used to prevent selected reactive groups, such as carboxy, amino, hydroxy, mercapto and the like, from undergoing undesired reactions, such as nucleophilic, electrophilic, oxidation, reduction and the like. Preferred protecting groups are indicated herein where appropriate. Examples of amino protecting groups include, but are not limited to, aralkyl, substituted aralkyl, cycloalkenylalkyl and substituted cycloalkenyl alkyl, allyl, substituted allyl, acyl, alkoxycarbonyl, aralkoxycarbonyl, silyl and the like. Examples of aralkyl include, but are not limited to, benzyl, ortho-methylbenzyl, trityl and benzhydryl, which can be optionally substituted with halogen, alkyl, alkoxy, hydroxy, nitro, acylamino, acyl and the like, and salts, such as phosphonium and ammonium salts. Examples of aryl groups include phenyl, naphthyl, indanyl, anthracenyl, 9-(9-phenylfluorenyl), phenanthrenyl, durenyl and the like. Examples of cycloalkenylalkyl or substituted cycloalkylenylalkyl radicals, preferably have 6-10 carbon atoms, include, but are not limited to, cyclohexenyl methyl and the like. Suitable acyl, alkoxycarbonyl and aralkoxycarbonyl groups include benzyloxycarbonyl, t-butoxycarbonyl, iso-butoxycarbonyl, benzoyl, substituted benzoyl, butyryl, acetyl, trifluoroacetyl, trichloro acetyl, phthaloyl and the like. A mixture of protecting groups can be used to protect the same amino group, such as a primary amino group can be protected by both an aralkyl group and an aralkoxycarbonyl group. Amino protecting groups can also form a heterocyclic ring with the nitrogen to which they are attached, for example, 1,2-bis(methylene)benzene, phthalimidyl, succinimidyl, maleimidyl and the like and where these heterocyclic groups can further include adjoining aryl and cycloalkyl rings. In addition, the heterocyclic groups can be mono-, di- or tri-substituted, such as nitrophthalimidyl. Amino groups may also be protected against undesired reactions, such as oxidation, through the formation of an addition salt, such as hydrochloride, toluenesulfonic acid, trifluoroacetic acid and the like. Many of the amino protecting groups are also suitable for protecting carboxy, hydroxy and mercapto groups. For example, aralkyl groups. Alkyl groups are also suitable groups for protecting hydroxy and mercapto groups, such as tert-butyl.
  • Silyl protecting groups are silicon atoms optionally substituted by one or more alkyl, aryl and aralkyl groups. Suitable silyl protecting groups include, but are not limited to, trimethylsilyl, triethylsilyl, triisopropylsilyl, tert-butyldimethylsilyl, dimethylphenylsilyl, 1,2-bis(dimethylsilyl)benzene, 1,2-bis(dimethylsilyl)ethane and diphenylmethylsilyl. Silylation of an amino groups provide mono- or di-silylamino groups. Silylation of aminoalcohol compounds can lead to a N,N,O-trisilyl derivative. Removal of the silyl function from a silyl ether function is readily accomplished by treatment with, for example, a metal hydroxide or ammonium fluoride reagent, either as a discrete reaction step or in situ during a reaction with the alcohol group. Suitable silylating agents are, for example, trimethylsilyl chloride, tert-butyl-dimethylsilyl chloride, phenyldimethylsilyl chloride, diphenylmethyl silyl chloride or their combination products with imidazole or DMF. Methods for silylation of amines and removal of silyl protecting groups are well known to those skilled in the art. Methods of preparation of these amine derivatives from corresponding amino acids, amino acid amides or amino acid esters are also well known to those skilled in the art of organic chemistry including amino acid/amino acid ester or aminoalcohol chemistry.
  • Protecting groups are removed under conditions which will not affect the remaining portion of the molecule. These methods are well known in the art and include acid hydrolysis, hydrogenolysis and the like. A preferred method involves removal of a protecting group, such as removal of a benzyloxycarbonyl group by hydrogenolysis utilizing palladium on carbon in a suitable solvent system such as an alcohol, acetic acid, and the like or mixtures thereof. A t-butoxycarbonyl protecting group can be removed utilizing an inorganic or organic acid, such as HCl or trifluoroacetic acid, in a suitable solvent system, such as dioxane or methylene chloride. The resulting amino salt can readily be neutralized to yield the free amine. Carboxy protecting group, such as methyl, ethyl, benzyl, tert-butyl, 4-methoxyphenylmethyl and the like, can be removed under hydrolysis and hydrogenolysis conditions well known to those skilled in the art.
  • It should be noted that compounds of the invention may contain groups that may exist in tautomeric forms, such as cyclic and acyclic amidine and guanidine groups, heteroatom substituted heteroaryl groups (Y′=O, S, NR), and the like, which are illustrated in the following examples:
  • Figure US20140235642A1-20140821-C00013
  • and though one form is named, described, displayed and/or claimed herein, all the tautomeric forms are intended to be inherently included in such name, description, display and/or claim.
  • Prodrugs of the compounds of this invention are also contemplated by this invention. A prodrug is an active or inactive compound that is modified chemically through in vivo physiological action, such as hydrolysis, metabolism and the like, into a compound of this invention following administration of the prodrug to a patient. The suitability and techniques involved in making and using prodrugs are well known by those skilled in the art. For a general discussion of prodrugs involving esters see Svensson and Tunek Drug Metabolism Reviews 165 (1988) and Bundgaard Design of Prodrugs, Elsevier (1985). Examples of a masked carboxylate anion include a variety of esters, such as alkyl (for example, methyl, ethyl), cycloalkyl (for example, cyclohexyl), aralkyl (for example, benzyl, p-methoxybenzyl), and alkylcarbonyloxyalkyl (for example, pivaloyloxymethyl). Amines have been masked as arylcarbonyloxymethyl substituted derivatives which are cleaved by esterases in vivo releasing the free drug and formaldehyde (Bungaard J. Med. Chem. 2503 (1989)). Also, drugs containing an acidic NH group, such as imidazole, imide, indole and the like, have been masked with N-acyloxymethyl groups (Bundgaard Design of Prodrugs, Elsevier (1985)). Hydroxy groups have been masked as esters and ethers. EP 039,051 (Sloan and Little, Apr. 11, 1981) discloses Mannich-base hydroxamic acid prodrugs, their preparation and use.
  • The specification and claims contain listing of species using the language like “selected from . . . and . . . ” and “is . . . or . . . ” (sometimes referred to as Markush groups). When this language is used in this application, unless otherwise stated it is meant to include the group as a whole, or any single members thereof, or any subgroups thereof. The use of this language is merely for shorthand purposes and is not meant in any way to limit the removal of individual elements or subgroups as needed.
  • One aspect of the current invention relates to compounds having the general structure:
  • Figure US20140235642A1-20140821-C00014
  • or any pharmaceutically-acceptable salt thereof, wherein:
  • m is 0, 1, 2 or 3;
  • n is 0 or 1;
  • X1 is C(R4) or N;
  • X2 is C or N;
  • Y is NH or O;
  • R1 is selected from C1-6alk or a direct-bonded, C1-2alk-linked, C1-2alkO-linked, saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, but containing no more than one O or S atom, the C1-6alk and ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C1-6alk, C1-4haloalk, cyano, nitro, —C(═O)Ra, —C(═O)ORa, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Ra, —OC(═O)NRaRa, —OC(═O)N(Ra)S(═O)2Ra, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRaRa, —S(═O)2N(Ra)C(═O)Ra, —S(═O)2N(Ra)C(═O)ORa, —S(═O)2N(Ra)C(═O)NRaRa, —NRaRa, —N(Ra)C(═O)Ra, —N(Ra)C(═O)ORa, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Ra, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa, wherein the ring is additionally substituted by 0 or 1 directly bonded, SO2 linked, C(═O) linked or CH2 linked saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic ring substituted by 0, 1, 2 or 3 groups selected from halo, C1-6alk, C1-4haloalk, cyano, nitro, —C(═O)Ra, —C(═O)ORa, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Ra, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRaRa, —NRaRa, and —N(Ra)C(═O)Ra;
  • R2 is selected from H, halo, cyano, Rc, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa; or R2 is C1-6alk substituted by 0, 1, 2 or 3 substituents selected from C1-4haloalk, halo, cyano, nitro, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa, or R2 is C1-6alk substituted by 0, 1, 2 or 3 halo substituents and additionally substituted by 0 or 1 substituents selected from Rc;
  • R3 is H, C1-8alk, C1-4haloalk, halo, cyano, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa or —NRaC2-6alkORa;
  • R4 is independently, at each instance, H, C1-6alk, —C1-3haloalk, —OC1-6alk, —OC1-3haloalk, —N(C1-6alk)C1-6alk, —NHC1-6alk, —NC(═O)C1-6alk, —N(C1-6alk)C1-6alk, F, Cl, Br, CN, OH or NH2; or R3 and R4 together form a four-atom unsaturated bridge containing 0 or 1 N atoms, wherein the bridge is substituted by 0, 1 or 2 R5 substituents;
  • R5 is independently, in each instance, F, CH3 or CF3;
  • R6 is F;
  • Ra is independently, at each instance, H or Rb;
  • Rb is independently, at each instance, phenyl, benzyl or C1-6alk, the phenyl, benzyl and C1-6alk being substituted by 0, 1, 2 or 3 substituents selected from halo, oxo, C1-4alk, C1-3haloalk, —OC1-4alk, —OH, —NH2, —OC1-4alk, —OC1-4haloalk, —NHC1-4alk, and —N(C1-4alk)C1-4alk; and
  • Rc is independently, at each instance, a saturated, partially saturated or unsaturated 4-, 5- or 6-membered monocyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, wherein the C1-6alkyl and ring are substituted by 0 or 1 oxo groups substituted by 0, 1, 2 or 3 substituents selected from C1-8alk, C1-4haloalk, halo and cyano.
  • Another aspect of the current invention relates to compounds having the general structure:
  • Figure US20140235642A1-20140821-C00015
  • or any pharmaceutically-acceptable salt thereof, wherein:
  • m is 0, 1, 2 or 3;
  • n is 0 or 1;
  • X1 is C(R4) or N;
  • X2 is C or N;
  • R1 is selected from C1-6alk or a direct-bonded, C1-2alk-linked, C1-2alkO-linked, saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, but containing no more than one O or S atom, the C1-6alk and ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C1-6alk, C1-4haloalk, cyano, nitro, —C(═O)Ra, —C(═O)ORa, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Ra, —OC(═O)NRaRa, —OC(═O)N(Ra)S(═O)2Ra, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRaRa, —S(═O)2N(Ra)C(═O)Ra, —S(═O)2N(Ra)C(═O)ORa, —S(═O)2N(Ra)C(═O)NRaRa, —NRaRa, —N(Ra)C(═O)Ra, —N(Ra)C(═O)ORa, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Ra, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa, wherein the ring is additionally substituted by 0 or 1 directly bonded, SO2 linked, C(═O) linked or CH2 linked saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic ring substituted by 0, 1, 2 or 3 groups selected from halo, C1-6alk, C1-4haloalk, cyano, nitro, —C(═O)Ra, —C(═O)ORa, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Ra, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRaRa, —NRaRa, and —N(Ra)C(═O)Ra;
  • R2 is selected from H, halo, cyano, Rc, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa; or R2 is C1-6alk substituted by 0, 1, 2 or 3 substituents selected from C1-4haloalk, halo, cyano, nitro, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa, or R2 is C1-6alk substituted by 0, 1, 2 or 3 halo substituents and additionally substituted by 0 or 1 substituents selected from Rc;
  • R3 is H, C1-8alk, C1-4haloalk, halo, cyano, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa or —NRaC2-6alkORa;
  • R4 is independently, at each instance, H, C1-6alk, —C1-3haloalk, —OC1-6alk, —OC1-3haloalk, —N(C1-6alk)C1-6alk, —NHC1-6alk, —NC(═O)C1-6alk, —N(C1-6alk)C1-6alk, F, Cl, Br, CN, OH or NH2; or R3 and R4 together form a four-atom unsaturated bridge containing 0 or 1 N atoms, wherein the bridge is substituted by 0, 1 or 2 R5 substituents;
  • R5 is independently, in each instance, F, CH3 or CF3;
  • R6 is F;
  • Ra is independently, at each instance, H or Rb;
  • Rb is independently, at each instance, phenyl, benzyl or C1-6alk, the phenyl, benzyl and C1-6alk being substituted by 0, 1, 2 or 3 substituents selected from halo, oxo, C1-4alk, C1-3haloalk, —OC1-4alk, —OH, —NH2, —OC1-4alk, —OC1-4haloalk, —NHC1-4alk, and —N(C1-4alk)C1-4alk; and
  • Rc is independently, at each instance, a saturated, partially saturated or unsaturated 4-, 5- or 6-membered monocyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, wherein the C1-6alkyl and ring are substituted by 0 or 1 oxo groups substituted by 0, 1, 2 or 3 substituents selected from C1-8alk, C1-4haloalk, halo and cyano.
  • Another aspect of the current invention relates to compounds having the general structure:
  • Figure US20140235642A1-20140821-C00016
  • or any pharmaceutically-acceptable salt thereof, wherein:
  • m is 0, 1, 2 or 3;
  • n is 0 or 1;
  • X1 is C(R4) or N;
  • R1 is selected from C1-6alk or a direct-bonded, C1-2alk-linked, C1-2alkO-linked, saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, but containing no more than one O or S atom, the C1-6alk and ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C1-6alk, C1-4haloalk, cyano, nitro, —C(═O)Ra, —C(═O)ORa, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Ra, —OC(═O)NRaRa, —OC(═O)N(Ra)S(═O)2Ra, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRaRa, —S(═O)2N(Ra)C(═O)Ra, —S(═O)2N(Ra)C(═O)ORa, —S(═O)2N(Ra)C(═O)NRaRa, —NRaRa, —N(Ra)C(═O)Ra, —N(Ra)C(═O)ORa, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Ra, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa, wherein the ring is additionally substituted by 0 or 1 directly bonded, SO2 linked, C(═O) linked or CH2 linked saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic ring substituted by 0, 1, 2 or 3 groups selected from halo, C1-6alk, C1-4haloalk, cyano, nitro, —C(═O)Ra, —C(═O)ORa, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Ra, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRaRa, —NRaRa, and —N(Ra)C(═O)Ra;
  • R2 is selected from H, halo, cyano, Rc, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa; or R2 is C1-6alk substituted by 0, 1, 2 or 3 substituents selected from C1-4haloalk, halo, cyano, nitro, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa, or R2 is C1-6alk substituted by 0, 1, 2 or 3 halo substituents and additionally substituted by 0 or 1 substituents selected from Rc;
  • R3 is H, C1-8alk, C1-4haloalk, halo, cyano, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa or —NRaC2-6alkORa;
  • R4 is independently, at each instance, H, C1-6alk, —C1-3haloalk, —OC1-6alk, —OC1-3haloalk, —N(C1-6alk)C1-6alk, —NHC1-6alk, —NC(═O)C1-6alk, —N(C1-6alk)C1-6alk, F, Cl, Br, CN, OH or NH2; or R3 and R4 together form a four-atom unsaturated bridge containing 0 or 1 N atoms, wherein the bridge is substituted by 0, 1 or 2 R5 substituents;
  • R5 is independently, in each instance, F, CH3 or CF3;
  • R6 is F;
  • Ra is independently, at each instance, H or Rb;
  • Rb is independently, at each instance, phenyl, benzyl or C1-6alk, the phenyl, benzyl and C1-6alk being substituted by 0, 1, 2 or 3 substituents selected from halo, oxo, C1-4alk, C1-3haloalk, —OC1-4alk, —OH, —NH2, —OC1-4alk, —OC1-4haloalk, —NHC1-4alk, and —N(C1-4alk)C1-4alk; and
  • Rc is independently, at each instance, a saturated, partially saturated or unsaturated 4-, 5- or 6-membered monocyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, wherein the C1-6alkyl and ring are substituted by 0 or 1 oxo groups substituted by 0, 1, 2 or 3 substituents selected from C1-8alk, C1-4haloalk, halo and cyano.
  • Another aspect of the current invention relates to compounds having the general structure:
  • Figure US20140235642A1-20140821-C00017
  • or any pharmaceutically-acceptable salt thereof, wherein:
  • m is 0, 1, 2 or 3;
  • n is 0 or 1;
  • X2 is C or N;
  • R1 is selected from C1-6alk or a direct-bonded, C1-2alk-linked, C1-2alkO-linked, saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, but containing no more than one O or S atom, the C1-6alk and ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C1-6alk, C1-4haloalk, cyano, nitro, —C(═O)Ra, —C(═O)ORa, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Ra, —OC(═O)NRaRa, —OC(═O)N(Ra)S(═O)2Ra, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRaRa, —S(═O)2N(Ra)C(═O)Ra, —S(═O)2N(Ra)C(═O)ORa, —S(═O)2N(Ra)C(═O)NRaRa, —NRaRa, —N(Ra)C(═O)Ra, —N(Ra)C(═O)ORa, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Ra, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa, wherein the ring is additionally substituted by 0 or 1 directly bonded, SO2 linked, C(═O) linked or CH2 linked saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic ring substituted by 0, 1, 2 or 3 groups selected from halo, C1-6alk, C1-4haloalk, cyano, nitro, —C(═O)Ra, —C(═O)ORa, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Ra, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRaRa, —NRaRa, and —N(Ra)C(═O)Ra;
  • R2 is selected from H, halo, cyano, Rc, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa; or R2 is C1-6alk substituted by 0, 1, 2 or 3 substituents selected from C1-4haloalk, halo, cyano, nitro, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa, or R2 is C1-6alk substituted by 0, 1, 2 or 3 halo substituents and additionally substituted by 0 or 1 substituents selected from Rc;
  • R3 is H, C1-8alk, C1-4haloalk, halo, cyano, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa or —NRaC2-6alkORa;
  • R4 is independently, at each instance, H, C1-6alk, —C1-3haloalk, —OC1-6alk, —OC1-3haloalk, —N(C1-6alk)C1-6alk, —NHC1-6alk, —NC(═O)C1-6alk, —N(C1-6alk)C1-6alk, F, Cl, Br, CN, OH or NH2;
  • R5 is F, CH3 or CF3;
  • R6 is F;
  • Ra is independently, at each instance, H or Rb;
  • Rb is independently, at each instance, phenyl, benzyl or C1-6alk, the phenyl, benzyl and C1-6alk being substituted by 0, 1, 2 or 3 substituents selected from halo, oxo, C1-4alk, C1-3haloalk, —OC1-4alk, —OH, —NH2, —OC1-4alk, —OC1-4haloalk, —NHC1-4alk, and —N(C1-4alk)C1-4alk; and
  • Rc is independently, at each instance, a saturated, partially saturated or unsaturated 4-, 5- or 6-membered monocyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, wherein the C1-6alkyl and ring are substituted by 0 or 1 oxo groups substituted by 0, 1, 2 or 3 substituents selected from C1-8alk, C1-4haloalk, halo and cyano.
  • Another aspect of the current invention relates to compounds having the general structure:
  • Figure US20140235642A1-20140821-C00018
  • or any pharmaceutically-acceptable salt thereof, wherein:
  • m is 0, 1, 2 or 3;
  • R1 is a direct-bonded, partially-saturated or unsaturated 5- or 6-membered monocyclic ring containing 1 or 2 atoms selected from N, O and S, but containing no more than one O or S atom, the C1-6alk and ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C1-6alk, C1-4haloalk and cyano;
  • R2 is selected from F and CF3;
  • R3 is CH3, CF3, F or Cl;
  • R4 is H or F;
  • R6 is F;
  • Ra is independently, at each instance, H or Rb;
  • Rb is independently, at each instance, phenyl, benzyl or C1-6alk, the phenyl, benzyl and C1-6alk being substituted by 0, 1, 2 or 3 substituents selected from halo, oxo, C1-4alk, C1-3haloalk, —OC1-4alk, —OH, —NH2, —OC1-4alk, —OC1-4haloalk, —NHC1-4alk, and —N(C1-4alk)C1-4alk; and
  • Rc is independently, at each instance, a saturated, partially saturated or unsaturated 4-, 5- or 6-membered monocyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, wherein the C1-6alkyl and ring are substituted by 0 or 1 oxo groups substituted by 0, 1, 2 or 3 substituents selected from C1-8alk, C1-4haloalk, halo and cyano.
  • Another aspect of the current invention relates to compounds having the general structure:
  • Figure US20140235642A1-20140821-C00019
  • or any pharmaceutically-acceptable salt thereof, wherein:
  • m is 0, 1, 2 or 3;
  • R1 is a direct-bonded, partially-saturated or unsaturated 5- or 6-membered monocyclic ring containing 1 or 2 atoms selected from N, O and S, but containing no more than one O or S atom, the C1-6alk and ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C1-6alk, C1-4haloalk and cyano; and
  • R2 is selected from F and CF3.
  • Another aspect of the current invention relates to compounds having the general structure:
  • Figure US20140235642A1-20140821-C00020
  • or any pharmaceutically-acceptable salt thereof, wherein:
  • m is 0, 1, 2 or 3;
  • n is 0 or 1;
  • X1 is C(R4) or N;
  • X2 is C or N;
  • Y is NH or O;
  • R1 is selected from C1-6alk or a direct-bonded, C1-2alk-linked, C1-2alkO-linked, saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, but containing no more than one O or S atom, the C1-6alk and ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C1-6alk, C1-4haloalk, cyano, nitro, —C(═O)Ra, —C(═O)ORa, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Ra, —OC(═O)NRaRa, —OC(═O)N(Ra)S(═O)2Ra, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRaRa, —S(═O)2N(Ra)C(═O)Ra, —S(═O)2N(Ra)C(═O)ORa, —S(═O)2N(Ra)C(═O)NRaRa, —NRaRa, —N(Ra)C(═O)Ra, —N(Ra)C(═O)ORa, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Ra, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa, wherein the ring is additionally substituted by 0 or 1 directly bonded, SO2 linked, C(═O) linked or CH2 linked saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic ring substituted by 0, 1, 2 or 3 groups selected from halo, C1-6alk, C1-4haloalk, cyano, nitro, —C(═O)Ra, —C(═O)ORa, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Ra, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRaRa, —NRaRa, and —N(Ra)C(═O)Ra;
  • R2 is selected from H, halo, cyano, Rc, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa; or R2 is C1-6alk substituted by 0, 1, 2 or 3 substituents selected from C1-4haloalk, halo, cyano, nitro, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa, or R2 is C1-6alk substituted by 0, 1, 2 or 3 halo substituents and additionally substituted by 0 or 1 substituents selected from Rc;
  • R3 is H, C1-8alk, C1-4haloalk, halo, cyano, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa or —NRaC2-6alkORa;
  • R4 is independently, at each instance, H, C1-6alk, —C1-3haloalk, —OC1-6alk, —OC1-3haloalk, —N(C1-6alk)C1-6alk, —NHC1-6alk, —NC(═O)C1-6alk, —N(C1-6alk)C1-6alk, F, Cl, Br, CN, OH or NH2;
  • R5 is F, CH3 or CF3;
  • R6 is F;
  • Ra is independently, at each instance, H or Rb;
  • Rb is independently, at each instance, phenyl, benzyl or C1-6alk, the phenyl, benzyl and C1-6alk being substituted by 0, 1, 2 or 3 substituents selected from halo, oxo, C1-4alk, C1-3haloalk, —OC1-4alk, —OH, —NH2, —OC1-4alk, —OC1-4haloalk, —NHC1-4alk, and —N(C1-4alk)C1-4alk; and
  • Rc is independently, at each instance, a saturated, partially saturated or unsaturated 4-, 5- or 6-membered monocyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, wherein the C1-6alkyl and ring are substituted by 0 or 1 oxo groups substituted by 0, 1, 2 or 3 substituents selected from C1-8alk, C1-4haloalk, halo and cyano.
  • In another embodiment, in conjunction with any above or below embodiments, R1 is C1-6alk substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C1-6alk, C1-4haloalk, cyano, nitro, —C(═O)Ra, —C(═O)ORa, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Ra, —OC(═O)NRaRa, —OC(═O)N(Ra)S(═O)2Ra, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRaRa, —S(═O)2N(Ra)C(═O)Ra, —S(═O)2N(Ra)C(═O)ORa, —S(═O)2N(Ra)C(═O)NRaRa, —NRaRa, —N(Ra)C(═O)Ra, —N(Ra)C(═O)ORa, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Ra, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa.
  • In another embodiment, in conjunction with any above or below embodiments, R1 is C1-2alk-linked saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, but containing no more than one O or S atom, the ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C1-6alk, C1-4haloalk, cyano, nitro, —C(═O)Ra, —C(═O)ORa, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Ra, —OC(═O)NRaRa, —OC(═O)N(Ra)S(═O)2Ra, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRaRa, —S(═O)2N(Ra)C(═O)Ra, —S(═O)2N(Ra)C(═O)ORa, —S(═O)2N(Ra)C(═O)NRaRa, —NRaRa, —N(Ra)C(═O)Ra, —N(Ra)C(═O)ORa, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Ra, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa.
  • In another embodiment, in conjunction with any above or below embodiments, R1 is a direct-bonded saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, but containing no more than one O or S atom, the ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C1-6alk, C1-4haloalk, cyano, nitro, —C(═O)Ra, —C(═O)ORa, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Ra, —OC(═O)NRaRa, —OC(═O)N(Ra)S(═O)2Ra, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRaRa, —S(═O)2N(Ra)C(═O)Ra, —S(═O)2N(Ra)C(═O)ORa, —S(═O)2N(Ra)C(═O)NRaRa, —NRaRa, —N(Ra)C(═O)Ra, —N(Ra)C(═O)ORa, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Ra, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa, wherein the ring is additionally substituted by 0 or 1 directly bonded, SO2 linked, C(═O) linked or CH2 linked saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic ring substituted by 0, 1, 2 or 3 groups selected from halo, C1-6alk, C1-4haloalk, cyano, nitro, —C(═O)Ra, —C(═O)ORa, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Ra, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRaRa, —NRaRa, and —N(Ra)C(═O)Ra.
  • In another embodiment, in conjunction with any above or below embodiments, R1 is a direct-bonded partially-saturated or unsaturated 5- or 6-membered monocyclic ring containing 1, 2, 3 or 4 atoms selected from N, O and S, but containing no more than one O or S atom, the ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C1-6alk, C1-4haloalk, cyano, nitro, —C(═O)Ra, —C(═O)ORa, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Ra, —OC(═O)NRaRa, —OC(═O)N(Ra)S(═O)2Ra, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRaRa, —S(═O)2N(Ra)C(═O)Ra, —S(═O)2N(Ra)C(═O)ORa, —S(═O)2N(Ra)C(═O)NRaRa, —NRaRa, —N(Ra)C(═O)Ra, —N(Ra)C(═O)ORa, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Ra, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa.
  • In another embodiment, in conjunction with any above or below embodiments, R1 is a direct-bonded partially-saturated or unsaturated 6-membered monocyclic ring containing 1 or 2 N atoms, substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C1-6alk, C1-4haloalk, cyano and —ORa.
  • In another embodiment, in conjunction with any above or below embodiments, R1 is a direct-bonded unsaturated 10-membered bicyclic ring containing 1 or 2 N atoms, substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C1-6alk, C1-4haloalk, cyano and —ORa.
  • Another aspect of the invention relates to a method of treating acute, inflammatory and neuropathic pain, dental pain, general headache, migraine, cluster headache, mixed-vascular and non-vascular syndromes, tension headache, general inflammation, arthritis, rheumatic diseases, osteoarthritis, inflammatory bowel disorders, depression, anxiety, inflammatory eye disorders, inflammatory or unstable bladder disorders, psoriasis, skin complaints with inflammatory components, chronic inflammatory conditions, inflammatory pain and associated hyperalgesia and allodynia, neuropathic pain and associated hyperalgesia and allodynia, diabetic neuropathy pain, causalgia, sympathetically maintained pain, deafferentation syndromes, asthma, epithelial tissue damage or dysfunction, herpes simplex, disturbances of visceral motility at respiratory, genitourinary, gastrointestinal or vascular regions, wounds, burns, allergic skin reactions, pruritus, vitiligo, general gastrointestinal disorders, gastric ulceration, duodenal ulcers, diarrhea, gastric lesions induced by necrotising agents, hair growth, vasomotor or allergic rhinitis, bronchial disorders or bladder disorders, comprising the step of administering a compound as described above.
  • Another aspect of the invention relates to a pharmaceutical composition comprising a compound according to claim 1 and a pharmaceutically-acceptable diluent or carrier.
  • Another aspect of the invention relates to the use of a compound according to any of the above embodiments as a medicament.
  • Another aspect of the invention relates to the use of a compound according to any of the above embodiments in the manufacture of a medicament for the treatment of acute, inflammatory and neuropathic pain, dental pain, general headache, migraine, cluster headache, mixed-vascular and non-vascular syndromes, tension headache, general inflammation, arthritis, rheumatic diseases, osteoarthritis, inflammatory bowel disorders, anxiety, depression, inflammatory eye disorders, inflammatory or unstable bladder disorders, psoriasis, skin complaints with inflammatory components, chronic inflammatory conditions, inflammatory pain and associated hyperalgesia and allodynia, neuropathic pain and associated hyperalgesia and allodynia, diabetic neuropathy pain, causalgia, sympathetically maintained pain, deafferentation syndromes, asthma, epithelial tissue damage or dysfunction, herpes simplex, disturbances of visceral motility at respiratory, genitourinary, gastrointestinal or vascular regions, wounds, burns, allergic skin reactions, pruritus, vitiligo, general gastrointestinal disorders, gastric ulceration, duodenal ulcers, diarrhea, gastric lesions induced by necrotising agents, hair growth, vasomotor or allergic rhinitis, bronchial disorders or bladder disorders.
  • ADDITIONAL EMBODIMENTS
  • The embodiments listed below are presented in numbered form for convenience and are in addition to the embodiments described above.
  • 1. In a first additional embodiment, the invention provides a compound of Formula I having the structure:
  • Figure US20140235642A1-20140821-C00021
  • or a pharmaceutically-acceptable salt thereof, a tautomer thereof, a pharmaceutically-acceptable salt of the tautomer, a stereoisomer thereof, or a mixture thereof, wherein:
  • m is 0, 1, 2 or 3;
  • n is 0 or 1;
  • X1 is C(R4) or N;
  • X2 is CH or N;
  • Y is NH, NR1a, or O;
  • Z is O or S;
  • R1 is C1-6alk, —(C═O)—O—C1-6alk, or a direct-bonded, C1-6alk-linked, C1-2alkO-linked, —C(═O)—O linked, —C(═O)— linked, saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 heteroatoms selected from N, O and S, the C1-6alk, the C1-6alk of the C1-6alk-link, and the monocyclic or bicyclic ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C1-6alk, C1-6alkOH, C1-6alk-C(═O)Ra, C1-6alk-C(═O)ORa, C1-4haloalk, cyano, nitro, —C(═O)Ra, —C(═O)ORa, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Ra, —OC(═O)NRaRa, —OC(═O)N(Ra)S(═O)2Ra, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, ═S, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRaRa, —S(═O)2N(Ra)C(═O)Ra, —S(═O)2N(Ra)C(═O)ORa, —S(═O)2N(Ra)C(═O)NRaRa, —NRaRa, —N(Ra)C(═O)Ra, —N(Ra)C(═O)ORa, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Ra, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa, wherein the ring is additionally substituted by 0 or 1 directly bonded, SO2 linked, C(═O) linked or CH2 linked saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic ring containing 0, 1, 2, 3 or 4 heteroatoms selected from N, O and S, and substituted by 0, 1, 2 or 3 groups selected from halo, C1-6alk, C1-4haloalk, cyano, oxo, nitro, —C(═O)Ra, —C(═O)ORa, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Ra, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRaRa, —NRaRa, and —N(Ra)C(═O)Ra; or R1a and R1, together with the N atom to which they are attached, form a saturated, partially-saturated or unsaturated 4-, 5-, 6-, or 7-membered monocyclic or a 9- or 10-membered bicyclic ring containing 0, 1, or 2 additional heteroatoms independently selected from N, O, and S, wherein the ring formed by R1a and R1 is substituted with 0, 1, or 2 substituents independently selected from halo, oxo, C1-6alk, C1-6alkOH, C1-6alk-C(═O)Ra, C1-6alk-C(═O)ORa, C1-4haloalk, cyano, nitro, —C(═O)Ra, —C(═O)ORa, or —C(═O)NRaRa;
  • R2 is H, halo, cyano, Rc, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa; or R2 is C1-6alk substituted by 0, 1, 2 or 3 substituents selected from C1-4haloalk, halo, cyano, nitro, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa, or R2 is C1-6alk substituted by 0, 1, 2 or 3 halo substituents and additionally substituted by 0 or 1 substituents selected from Rc;
  • R3 is H, C1-8alk, C1-8alkOH, C1-4haloalk, halo, cyano, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa or —NRaC2-6alkORa;
  • R4 is independently, at each instance, H, C1-6alk, —C1-3haloalk, —OC1-6alk, —OC1-3haloalk, —N(C1-6alk)C1-6alk, —NHC1-6alk, —NC(═O)C1-6alk, —N(C1-6alk)C1-6alk, F, Cl, Br, CN, OH or NH2; or R3 and R4 together form a four-atom unsaturated bridge containing 0 or 1 N atoms, wherein the bridge is substituted by 0, 1 or 2 R5 substituents;
  • R5 is independently, in each instance, Cl, Br, F, CH3, CF3, or ORa;
  • R6 is F, CF3, C1-6alk, or ORa;
  • Ra is independently, at each instance, H or Rb;
  • Rb is independently, at each instance, phenyl, benzyl or C1-6alk, the phenyl, benzyl and C1-6alk being substituted by 0, 1, 2 or 3 substituents selected from halo, oxo, C1-4alk, C1-3haloalk, —OC1-4alk, —OH, —NH2, —OC1-4alk, —OC1-4haloalk, —NHC1-4alk, and —N(C1-4alk)C1-4alk; and
  • Rc is independently, at each instance, a saturated, partially saturated or unsaturated 4-, 5- or 6-membered monocyclic ring containing 0, 1, 2, 3 or 4 heteroatoms selected from N, O and S, wherein the C1-6alkyl and ring are substituted by 0 or 1 oxo groups substituted by 0, 1, 2 or 3 substituents selected from C1-8alk, C1-4haloalk, halo and cyano.
  • 2. The compound of embodiment 1 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein the compound of Formula I has the Formula IA:
  • Figure US20140235642A1-20140821-C00022
  • 3. The compound of embodiment 1 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein the compound of Formula I has the Formula IB:
  • Figure US20140235642A1-20140821-C00023
  • wherein Y is NH or O.
  • 4. The compound of embodiment 3 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein the compound of Formula I has the Formula IC:
  • Figure US20140235642A1-20140821-C00024
  • or any pharmaceutically-acceptable salt thereof, wherein:
  • m is 0, 1, 2 or 3;
  • n is 0 or 1;
  • X1 is C(R4) or N;
  • X2 is CH or N;
  • R1 is selected from C1-6alk or a direct-bonded, C1-2alk-linked, C1-2alkO-linked, saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 heteroatoms selected from N, O and S, but containing no more than one O or S atom, the C1-6alk and ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C1-6alk, C1-4haloalk, cyano, nitro, —C(═O)Ra, —C(═O)ORa, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Ra, —OC(═O)NRaRa, —OC(═O)N(Ra)S(═O)2Ra, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRaRa, —S(═O)2N(Ra)C(═O)Ra, —S(═O)2N(Ra)C(═O)ORa, —S(═O)2N(Ra)C(═O)NRaRa, —NRaRa, —N(Ra)C(═O)Ra, —N(Ra)C(═O)ORa, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Ra, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa, wherein the ring is additionally substituted by 0 or 1 directly bonded, SO2 linked, C(═O) linked or CH2 linked saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic ring substituted by 0, 1, 2 or 3 groups selected from halo, C1-6alk, C1-4haloalk, cyano, nitro, —C(═O)Ra, —C(═O)ORa, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Ra, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRaRa, —NRaRa, and —N(Ra)C(═O)Ra;
  • R2 is selected from H, halo, cyano, Rc, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa; or R2 is C1-6alk substituted by 0, 1, 2 or 3 substituents selected from C1-4haloalk, halo, cyano, nitro, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa, or R2 is C1-6alk substituted by 0, 1, 2 or 3 halo substituents and additionally substituted by 0 or 1 substituents selected from Rc;
  • R3 is H, C1-8alk, C1-4haloalk, halo, cyano, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa or —NRaC2-6alkORa;
  • R4 is independently, at each instance, H, C1-6alk, —C1-3haloalk, —OC1-6alk, —OC1-3haloalk, —N(C1-6alk)C1-6alk, —NHC1-6alk, —NC(═O)C1-6alk, —N(C1-6alk)C1-6alk, F, Cl, Br, CN, OH or NH2; or R3 and R4 together form a four-atom unsaturated bridge containing 0 or 1 N atoms, wherein the bridge is substituted by 0, 1 or 2 R5 substituents;
  • R5 is independently, in each instance, F, CH3 or CF3;
  • R6 is F;
  • Ra is independently, at each instance, H or Rb;
  • Rb is independently, at each instance, phenyl, benzyl or C1-6alk, the phenyl, benzyl and C1-6alk being substituted by 0, 1, 2 or 3 substituents selected from halo, oxo, C1-4alk, C1-3haloalk, —OC1-4alk, —OH, —NH2, —OC1-4alk, —OC1-4haloalk, —NHC1-4alk, and —N(C1-4alk)C1-4alk; and
  • Rc is independently, at each instance, a saturated, partially saturated or unsaturated 4-, 5- or 6-membered monocyclic ring containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, wherein the C1-6alkyl and ring are substituted by 0 or 1 oxo groups substituted by 0, 1, 2 or 3 substituents selected from C1-8alk, C1-4haloalk, halo and cyano.
  • 5. The compound of embodiment 4 or the pharmaceutically-acceptable salt thereof
  • 6. The compound of embodiment 1 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein the compound of Formula I has the Formula ID:
  • Figure US20140235642A1-20140821-C00025
  • wherein Y is NH or O.
  • 7. The compound of embodiment 1 or embodiment 2, wherein Y is NR1a.
  • 8. The compound of embodiment 7, wherein R1a and R1, together with the N to which they are attached, form a 6 membered unsaturated ring.
  • 9. The compound of embodiment 8, wherein R1a and R1, together with the N to which they are attached, form a morpholine ring, a piperidine ring, or a piperazine ring.
  • 10. The compound of embodiment 1 or embodiment 2 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R1 is a saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 heteroatoms selected from N, O and S, wherein the monocyclic or bicyclic ring is independently substituted by 0, 1, 2 or 3 substituents.
  • 11. The compound of embodiment 10 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R1 is selected from phenyl, pyridyl, pyridinonyl, piperidinonyl, pyrimidinyl, tetradyrofuranyl, tetrahydropyranyl, oxetanyl, furanyl, tetrahydrothiophenyl, thiophenyl, isoxazolyl, pyrrolidinonyl, piperidinyl, cyclohexyl, cyclohexanonyl, quinolinyl, isoquinolinyl, tetrahydronaphthalenyl, dihydroindenyl, indolinonyl, indolinyl, or benzofuranyl independently substituted by 0, 1, 2, or 3 substituents.
  • 12. The compound of embodiment 11 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R1 is substituted with 0, 1, 2, or 3 substituents independently selected from F, Cl, Br, I, CN, CF3, NO2, oxo, C1-6alk, OCH3, OCH2CH3, COCH3, CO2H, CO2CH3, CO2CH2CH3, SCH3, SO2CH3, oxadiazolyl, a methyl-substituted oxadiazolyl, or oxadiazolonyl.
  • 13. The compound of embodiment 1 or embodiment 2 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R1 is C1-6alk, wherein the C1-6alk is independently substituted by 0, 1, 2 or 3 substituents.
  • 14. The compound of embodiment 13 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R1 is substituted with 0, 1, 2, or 3 substituents independently selected from F, Cl, Br, I, CN, CF3, NO2, oxo, C1-6alk, OCH3, OCH2CH3, COCH3, CO2H, CO2CH3, CO2CH2CH3, SCH3, SO2CH3, oxadiazolyl, a methyl-substituted oxadiazolyl, or oxadiazolonyl.
  • 15. The compound of embodiment 1 or embodiment 2 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R1 is a C1-6alk-linked, C1-2alkO-linked, —C(═O)—O linked, or —C(═O)— linked, saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 heteroatoms selected from N, O and S, wherein the monocyclic or bicyclic ring is substituted by 0, 1, 2 or 3 substituents.
  • 16. The compound of embodiment 15 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R1 is a —C1-2alk-linked saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 heteroatoms selected from N, O and S, wherein the monocyclic or bicyclic ring is substituted by 0, 1, 2 or 3 substituents.
  • 17. The compound of embodiment 1 or embodiment 2 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R1 is a group of formula
  • Figure US20140235642A1-20140821-C00026
    Figure US20140235642A1-20140821-C00027
    Figure US20140235642A1-20140821-C00028
    Figure US20140235642A1-20140821-C00029
    Figure US20140235642A1-20140821-C00030
    Figure US20140235642A1-20140821-C00031
    Figure US20140235642A1-20140821-C00032
    Figure US20140235642A1-20140821-C00033
    Figure US20140235642A1-20140821-C00034
    Figure US20140235642A1-20140821-C00035
  • and the symbol
    Figure US20140235642A1-20140821-P00001
    , when drawn across a bond, indicates the point of attachment to the rest of the molecule.
  • 18. The compound of embodiment 1 or claim 2 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R1 is a group of formula
  • Figure US20140235642A1-20140821-C00036
  • and the symbol
    Figure US20140235642A1-20140821-P00001
    , when drawn across a bond, indicates the point of attachment to the rest of the molecule.
  • 19. The compound of embodiment 1 or embodiment 2 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R1 is
  • Figure US20140235642A1-20140821-C00037
  • and the symbol
    Figure US20140235642A1-20140821-P00001
    , when drawn across a bond, indicates the point of attachment to the rest of the molecule.
  • 20. The compound of embodiment 1 or embodiment 2 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R1 is
  • Figure US20140235642A1-20140821-C00038
  • and the symbol
    Figure US20140235642A1-20140821-P00001
    , when drawn across a bond, indicates the point of attachment to the rest of the molecule.
  • 21. The compound of embodiment 1 or embodiment 2 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R1 is
  • Figure US20140235642A1-20140821-C00039
  • and the symbol
    Figure US20140235642A1-20140821-P00001
    , when drawn across a bond, indicates the point of attachment to the rest of the molecule.
  • 22. The compound of any one of embodiments 1-4 or 9-21 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein Y is NH.
  • 23. The compound of any one of embodiments 1-4 or 9-21 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein Y is O.
  • 24. The compound of any one of embodiments 1-4 or 9-23 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R5 is independently, in each instance F or CF3.
  • 25. The compound of embodiment 24 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R5 is F.
  • 26. The compound of any one of embodiments 1-4 or 9-25 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R6 is F or CF3.
  • 27. The compound of embodiment 26 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R6 is CF3.
  • 28. The compound of any one of embodiments 1-4 or 9-27 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein n is 1.
  • 29. The compound of any one of embodiments 1-4 or 9-27 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein m is 0 or 1.
  • 30. The compound of embodiment 29 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein m is 1.
  • 31. The compound of embodiment 29 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein m is 0.
  • 32. The compound of any one of embodiments 1-4 or 9-27 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein n is 0.
  • 33. The compound of any one of embodiments 1-4 or 9-32 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R2 is —H, halo, or C1-6alk substituted by 0, 1, 2 or 3 halo substituents.
  • 34. The compound of embodiment 33 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R2 is —H, —F, —Br, —CF3, or C1-6alk.
  • 35. The compound of embodiment 34 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R2 is —F, —Br, —CF3, or C1-6alk.
  • 36. The compound of embodiment 35 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R2 is —F.
  • 37. The compound of embodiment 35 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R2 is —CF3.
  • 38. The compound of any one of embodiments 1-4 or 9-32 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R2 is —CH3, —CH2CH3, —CH2CH2CH3, —C(CH3)3, —CH(CH3)2, —CH2—C(CH3)3, —CH═CH2, —CH2CH═CH2, —C≡C—CH3, or a group of formula or
  • Figure US20140235642A1-20140821-C00040
  • and the symbol
    Figure US20140235642A1-20140821-P00001
    , when drawn across a bond, indicates the point of attachment to the rest of the molecule.
  • 39. The compound of any one of embodiments 1-4 or 9-38 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R3 is H, C1-8alk, C1-4haloalk, halo, or —ORa.
  • 40. The compound of embodiment 39 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R3 is H, —CH3, —CH2CH3, F, Cl, —OCH3, —OCF3, or —CF3.
  • 41. The compound of embodiment 40 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R3 is —CH3, —CH2CH3, F, Cl, —OCH3, —OCF3, or —CF3.
  • 42. The compound of embodiment 41 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R3 is —OCF3 or —CF3.
  • 43. The compound of embodiment 42 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R3 is —OCF3.
  • 44. The compound of embodiment 42 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R3 is —CF3.
  • 45. The compound of embodiment 40 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R3 is —H.
  • 46. The compound of any one of embodiments 1-4 or 9-45 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R4 is H.
  • 47. The compound of any one of embodiments 1-4 or 9-45 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R4 is F, Cl, C1-6alk, —OC1-6alk, or —C1-3haloalk.
  • 48. The compound of embodiment 47 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R4 is F, Cl, CF3, CH3, or OCH3.
  • 49. The compound of embodiment 48 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R4 is F.
  • 50. The compound of any one of embodiments 1-4 or 9-38 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein R3 and R4 together form a four-atom unsaturated bridge containing 0 or 1 N atoms, wherein the bridge is substituted by 0, 1 or 2 R5 substituents.
  • 51. The compound of any one of embodiments 1-4 or 9-50 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein X2 is N.
  • 52. The compound of any one of embodiments 1-4 or 9-50 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein X2 is CH.
  • 53. The compound of any one of embodiments 1-4 or 9-52 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein X1 is N.
  • 54. The compound of any one of embodiments 1-4 or 9-52 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein X1 is C(R4).
  • 55. The compound of embodiment 54 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein X1 is CH.
  • 56. The compound of any one of embodiments 1-4 or 9-50 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein X1 is C(R4) and X2 is N.
  • 57. The compound of any one of embodiments 1-4 or 9-50 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein X1 is N and X2 is N.
  • 58. The compound of embodiment 1 or embodiment 2 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, wherein X2 is N; R2 is F or CF3; m is 0; X1 is CH; R4 is F or H; R3 is CF3 or OCF3; Y is NH; Z is O; and R1 is
  • Figure US20140235642A1-20140821-C00041
  • and the symbol
    Figure US20140235642A1-20140821-P00001
    , when drawn across a bond, indicates the point of attachment to the rest of the molecule.
  • 59. The compound of embodiment 1, wherein the compound is
    • (S)-1-((3-fluoro-4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)-methyl)-3-(pyridin-3-yl)urea;
    • (S)-1-((4-methoxy-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-(pyridin-3-yl)-urea;
    • (S)-1-((3-chlorophenyl)-(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-(pyridin-3-yl)-urea;
    • (S)-1-((3-methoxy-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-(pyridin-3-yl)-urea;
    • (S)-1-((3-fluoro-4-methoxy-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-(pyridin-3-yl)-urea;
    • (S)-1-(pyridin-3-yl)-3-(quinolin-3-yl(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-((3-methylphenyl)-(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-(pyridin-3-yl)-urea;
    • (S)-1-((3-fluorophenyl)-(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-(pyridin-3-yl)-urea;
    • (S)-1-((3-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-(pyridin-3-yl)-urea;
    • (S)-1-((5-chloroquinolin-3-yl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-(pyridin-3-yl)-urea;
    • (S)-1-(naphthalen-2-yl(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-(pyridin-3-yl)-urea;
    • (S)-1-((8-chloroquinolin-3-yl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-(pyridin-3-yl)-urea;
    • (S)-1-((3,4-dichlorophenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-(pyridin-3-yl)-urea;
    • (S)-1-((8-fluoroquinolin-3-yl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-(pyridin-3-yl)-urea;
    • (S)-1-((3-fluoropyridin-2-yl)(4-(trifluoro-methoxy)-phenyl)-methyl)-3-(pyridin-3-yl)-urea;
    • (S)-1-((8-methoxy-quinolin-3-yl)-(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-(pyridin-3-yl)-urea;
    • (S)-1-(pyridin-3-yl)-3-(quinolin-6-yl(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-((7-methoxy-quinolin-3-yl)-(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-(pyridin-3-yl)-urea;
    • (S)-1-((4-fluorophenyl)-(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-(pyridin-3-yl)-urea;
    • (S)-1-(phenyl(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-(pyridin-3-yl)-urea;
    • 1-((4-fluoro-3-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-(pyridin-3-yl)-urea;
    • 1-((4-fluoro-3-(trifluoro-methyl)-phenyl)(3-fluoropyridin-2-yl)methyl)-3-(pyridin-3-yl)-urea;
    • 1-((3-chloro-5-fluorophenyl)-(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-(pyridin-3-yl)-urea;
    • 1-((3-chloro-5-fluorophenyl)-(3-fluoro-pyridin-2-yl)-methyl)-3-(pyridin-3-yl)urea;
    • 1-(pyridin-3-yl)-3-((3-(trifluoro-methyl)-pyridin-2-yl)(6-(trifluoro-methyl)-pyridin-3-yl)-methyl)urea;
    • (S)-1-((4-chlorophenyl)-(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-(pyridin-3-yl)-urea;
    • (S)-1-((4-ethylphenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-(pyridin-3-yl)-urea;
    • (S)-1-((3-bromopyridin-2-yl)(4-(trifluoro-methyl)-phenyl)-methyl)-3-(pyridin-3-yl)-urea;
    • (S)-1-((3-fluoro-4-(trifluoro-methyl)-phenyl)(3-fluoropyridin-2-yl)methyl)-3-(pyridin-3-yl)-urea;
    • (S)-1-(pyridin-2-yl(4-(trifluoro-methyl)-phenyl)-methyl)-3-(pyridin-3-yl)-urea;
    • (S)-1-phenyl-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-isopropyl-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • 1-((S)-1-(naphthalen-1-yl)ethyl)-3-((S)-(4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(3-phenylpropyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(phenyl formate)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(2-(benzo[d][1,3]dioxol-5-yl)-ethyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(3-bromophenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-ethyl 2-(3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)ureido)-acetate;
    • (S)-1-(benzofuran-5-yl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(4-cyanophenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-((3-bromopyridin-2-yl)(4-(trifluoro-methyl)-phenyl)-methyl)-3-(tert-butyl)urea;
    • (S)-1-(tert-butyl)-3-((3-(prop-1-yn-1-yl)pyridin-2-yl)(4-(trifluoro-methyl)-phenyl)-methyl)urea;
    • (S)-1-((3-allylpyridin-2-yl)(4-(trifluoro-methyl)-phenyl)-methyl)-3-(pyridin-3-yl)urea;
    • (S)-1-(pyridin-3-yl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(tert-butyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(4-cyanophenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(4-fluoro-phenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(2,4-difluoro-phenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(3,5-difluoro-phenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(2,6-difluoro-phenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-((3-(prop-1-yn-1-yl)-pyridin-2-yl)(4-(trifluoro-methyl)-phenyl)-methyl)-3-(pyridin-3-yl)-urea;
    • (S)-1-(3-methoxy-phenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(3-(methylsulfonyl)phenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • 1-(tetrahydro-2H-pyran-3-yl)-3-((S)-(4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(3,5-dimethylisoxazol-4-yl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • 1-(tetrahydrofuran-3-yl)-3-((S)-(4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(pyrimidin-5-yl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(quinolin-6-yl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(quinolin-3-yl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(quinolin-4-yl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • 1-((S)-(3-((R)-2,2-dimethylcyclopropyl)pyridin-2-yl)(4-(trifluoro-methyl)-phenyl)-methyl)-3-(pyridin-3-yl)-urea;
    • (S)-1-((3-neopentylpyridin-2-yl)(4-(trifluoro-methyl)-phenyl)-methyl)-3-(pyridin-3-yl)-urea;
    • (S)-1-(2,6-dibromo-4-fluorophenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • 1-((3-Sulfonylcyclopentyl)methyl)-3-((S)-(4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • 1-((S)-3-methylbutan-2-yl)-3-((S)-(4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-ethyl 3-(3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)ureido)-propanoate;
    • (S)—N-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methylcarbamoyl)benzamide;
    • (S)-ethyl 4-(3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)ureido)-butanoate;
    • (S)-1-(3-methyl-5-phenylisoxazol-4-yl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • 1-((R)-1-phenylethyl)-3-((S)-(4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(2-tert-butylphenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(3-fluoro-benzyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(2-chlorobenzyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(3,4-dimethoxy-phenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-benzyl 4-(3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)ureido)-piperidine-1-carboxylate;
    • (S)-1-(2,6-dichloropyridin-4-yl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(3-(5-methyl-1,2,4-oxadiazol-3-yl)phenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-methyl 4-(methylthio)-2-(3-((S)-(4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)ureido)-butanoate;
    • (S)-methyl 2-(3-((S)-(4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)ureido)-propanoate;
    • 1-((S)-1-(4-methoxy-phenyl)ethyl)-3-((S)-(4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)—N-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)methylcarbamoyl)ethylamide;
    • (S)-1-(2,3-dimethoxy-phenethyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • 1-((S)-hexan-2-yl)-3-((S)-(4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • 1-((S)-1-(3-methoxy-phenyl)ethyl)-3-((S)-(4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(2-methylbenzyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(3-acetylphenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(2-fluoro-benzyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(2,6-diethylphenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(2-ethyl-6-methylphenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-ethyl 3-(3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)ureido)-benzoate;
    • (S)-1-(2-ethyl-6-isopropylphenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(2-isopropyl-6-methylphenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(3,5-dimethoxy-phenethyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-ethyl 2-(3-((S)-(4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)ureido)-propanoate;
    • (S)-1-(3,4-dimethoxy-phenethyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-methyl 3-methyl-2-(3-((S)-(4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)ureido)-butanoate;
    • (S)-1-(3-cyanophenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • ethyl 3-methyl-2-(3-((S)-(4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)ureido)-butanoate;
    • (3S)-methyl 3-methyl-2-(3-((S)-(4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)ureido)-pentanoate;
    • (S)-1-(3-nitrophenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • 1-(1-(4-bromophenyl)-ethyl)-3-((S)-(4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-methyl 3-(3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)ureido)-benzoate;
    • (S)-dimethyl 5-(3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)ureido)-isophthalate;
    • (S)-butyl 2-(3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)ureido)-acetate;
    • (S)-1-(2-(3-(prop-1-en-2-yl)phenyl)-propan-2-yl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(3,5-dimethylphenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • 1-((S)-1-phenylethyl)-3-((S)-(4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(2,5-dimethylphenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-benzyl-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(2,5-dimethoxy-phenethyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-(3,4,5-trimethoxy-benzyl)urea;
    • (S)-1-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-(3,4,5-trimethoxy-phenyl)urea;
    • (S)-1-(thiophen-2-yl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • 1-((2-bromopyridin-3-yl)(4-(trifluoro-methyl)-phenyl)-methyl)-3-(pyridin-3-yl)urea;
    • (S)-1-((2-bromophenyl)-(4-(trifluoro-methyl)-phenyl)-methyl)-3-(pyridin-3-yl)urea;
    • (S)-1-((2-bromophenyl)-(4-(trifluoro-methyl)-phenyl)-methyl)-3-tert-butylurea;
    • (R)-1-((3-fluoro-4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-phenyl)-methyl)-3-(pyridin-3-yl)-urea;
    • (S)-methyl 2-(3-((3-fluoro-pyridin-2-yl)(4-(trifluoro-methyl)-phenyl)-methyl)ureido)-benzoate;
    • (S)-1-(pyridin-3-yl)-3-((4-(trifluoro-methyl)-phenyl)(4-(trifluoro-methyl)-pyridin-3-yl)-methyl)urea;
    • (S)-1-((3-fluoro-4-(trifluoro-methyl)-phenyl)(2-(trifluoro-methyl)-phenyl)-methyl)-3-(pyridin-3-yl)-urea;
    • (S)-1-(pyridin-3-yl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-thiourea;
    • (S)-1-tert-butyl-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-thiourea;
    • (S)-1-(3-iodophenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(4-bromobenzyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(3-ethoxyphenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(cyclohexylmethyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • 1-((R)-1-(3-methoxy-phenyl)ethyl)-3-((S)-(4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • 1-((S)-2,3-dihydro-1H-inden-1-yl)-3-((S)-(4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • 1-((R)-1-(4-methoxy-phenyl)ethyl)-3-((S)-(4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-(2,4,4-trimethylpentan-2-yl)urea;
    • (S)-1-(2,6-dichlorophenethyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(2-ethoxybenzyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(4-chlorobenzyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(2,4-dichlorobenzyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-methyl 3-phenyl-2-(3-((S)-(4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)ureido)-propanoate;
    • (S)-1-(2-(methylthio)-phenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(4-acetylphenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(4-fluoro-3-nitrophenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(4-methyl-3-nitrophenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(2-methoxy-4-nitrophenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea
    • (S)-1-(2-methyl-3-nitrophenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(5-methyl-2-nitrophenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(4-fluoro-benzyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(o-tolyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(2-fluoro-5-methylphenyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(3-methylbenzyl)-3-((4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)urea;
    • (S)-1-(4-(5-oxo-4,5-dihydro-1,2,4-oxadiazol-3-yl)phenyl)-3-((4-(trifluoromethyl)-phenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)urea;
    • (S)-1-((3-fluoropyridin-2-yl)(4-(trifluoro-methyl)-phenyl)-methyl)-3-(2-oxoindolin-5-;
    • 1-((S)-(3-(prop-1-yn-1-yl)-pyridin-2-yl)(4-(trifluoro-methyl)-phenyl)-methyl)-3-((S)-1,1,1-trifluoro-propan-2-yl)-urea;
    • 1-((S)-(3-fluoro-4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-((S)-1,1,1-trifluoro-propan-2-yl)-urea;
    • 1-((S)-(3-fluoro-4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-((R)-1,1,1-trifluoro-propan-2-yl)-urea;
    • 1-((S)-(4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-((S)-1,1,1-trifluoro-propan-2-yl)-urea;
    • 1-((S)-(2-bromophenyl)-(4-(trifluoro-methyl)-phenyl)-methyl)-3-((S)-1,1,1-trifluoro-propan-2-yl)-urea;
    • (S)-1-((3-fluoro-4-(trifluoro-methyl)-phenyl)(3-(trifluoro-methyl)-pyridin-2-yl)-methyl)-3-(pyrimidin-5-yl)urea;
    • 1-((S)-(3-fluoropyridin-2-yl)(4-(trifluoro-methyl)-phenyl)-methyl)-3-((S)-1,1,1-trifluoro-propan-2-yl)-urea;
    • 1-((S)-(3-bromopyridin-2-yl)(4-(trifluoro-methyl)-phenyl)-methyl)-3-((S)-1,1,1-trifluoro-propan-2-yl)-urea;
    • 1-((S)-(3-allylpyridin-2-yl)(4-(trifluoro-methyl)-phenyl)-methyl)-3-((S)-1,1,1-trifluoro-propan-2-yl)-urea;
    • (S)-1-(2-methoxypyrimidin-5-yl)-3-((4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)urea;
    • (S)-1-((3-fluoro-pyridin-2-yl)(4-(trifluoro-methyl)phenyl)-methyl)-3-(2-methoxypyrimidin-5-yl)-urea;
    • (S)-1-((3-fluoro-pyridin-2-yl)(4-(trifluoro-methyl)phenyl)-methyl)-3-(1-methyl-2-oxo-1,2-dihydropyridin-4-yl)urea;
    • (S)-1-((3-fluoro-pyridin-2-yl)(4-(trifluoro-methyl)phenyl)-methyl)-3-(1-methyl-6-oxo-1,6-dihydropyridin-3-yl)urea;
    • (S)-1-((3-fluoro-pyridin-2-yl)(4-(trifluoro-methyl)phenyl)-methyl)-3-(2-oxo-1,2-dihydropyridin-3-yl)urea;
    • (S)-1-((3-fluoro-pyridin-2-yl)(4-(trifluoro-methyl)phenyl)-methyl)-3-(2-oxo-1,2-dihydropyridin-4-yl)urea;
    • (S)-1-((3-fluoro-pyridin-2-yl)(4-(trifluoro-methyl)phenyl)-methyl)-3-(6-oxo-1,6-dihydropyridin-3-yl)urea;
    • (S)-1-((3-fluoro-pyridin-2-yl)(4-(trifluoro-methyl)phenyl)-methyl)-3-(2-oxoindolin-6-yl)urea;
    • (S)-1-((3-propylpyridin-2-yl)(4-(trifluoromethyl)phenyl)methyl)-3-(pyridin-3-yl)urea;
    • (S)-tert-butyl (pyridin-2-yl(4-(trifluoromethyl)phenyl)methyl)carbamate;
    • (S)-ethyl ((4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)carbamate;
    • 1-(4-fluorophenyl)-3-(phenyl(4-(trifluoromethyl)phenyl)methyl)urea;
    • (S)-benzyl (pyridin-2-yl(4-(trifluoromethyl)phenyl)methyl)carbamate 2,2,2-trifluoroacetate; or
    • (S)-tert-butyl ((3-bromopyridin-2-yl)(4-(trifluoromethyl)phenyl)methyl)carbamate; or
      the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof
  • 60. The compound of embodiment 1, wherein the compound is
    • (S)-tert-butyl ((3-fluoro-4-(trifluoromethoxy)phenyl)(3-fluoropyridin-2-yl)methyl)carbamate;
    • (S)-isopropyl ((3-fluoro-4-(trifluoromethoxy)phenyl)(3-fluoropyridin-2-yl)methyl)carbamate;
    • (S)-tetrahydro-2H-pyran-4-yl ((3-fluoro-4-(trifluoromethoxy)phenyl)(3-fluoropyridin-2-yl)methyl)carbamate;
    • (S)-oxetan-3-yl ((3-fluoro-4-(trifluoromethoxy)phenyl)(3-fluoropyridin-2-yl)methyl)carbamate;
    • 6-oxopiperidin-3-yl ((S)-(3-fluoro-4-(trifluoromethoxy)phenyl)(3-fluoropyridin-2-yl)methyl)carbamate;
    • (S)-1-(quinolin-5-yl)-3-((4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)urea;
    • (S)-4-(3-((3-fluoro-4-(trifluoromethoxy)phenyl)(3-fluoropyridin-2-yl)methyl)ureido)benzoic acid;
    • (S)-1-(2-chlorophenyl)-3-((4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)urea (2,2,2-trifluoro-acetate);
    • (S)-methyl 2-methyl-3-(3-((4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)ureido)benzoate (2,2,2-trifluoro-acetate);
    • (S)-1-(2-methoxy-5-methylphenyl)-3-((4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)urea (2,2,2-trifluoro-acetate);
    • (S)-1-(3,4-dichlorobenzyl)-3-((4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)urea (2,2,2-trifluoro-acetate);
    • 1-(1,2,3,4-tetrahydronaphthalen-1-yl)-3-((S)-(4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)urea (2,2,2-trifluoro-acetate);
    • (S)-1-mesityl-3-((4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)urea (2,2,2-trifluoro-acetate);
    • (S)-ethyl 4-(3-((4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)ureido)benzoate (2,2,2-trifluoro-acetate); or
    • (S)-1-((3-fluoro-4-(trifluoromethoxy)phenyl)(3-fluoropyridin-2-yl)methyl)-3-(pyridin-3-yl)urea; or
      the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof
  • 61. The compound of embodiment 1, wherein the compound is
    • (S)-1-((3-fluoro-4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)-3-(pyridin-3-yl)urea;
    • (S)-1-(pyridin-3-yl)-3-((4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)urea;
    • (S)-1-((3-(prop-1-yn-1-yl)pyridin-2-yl)(4-(trifluoromethyl)phenyl)methyl)-3-(pyridin-3-yl)urea;
    • 1-((S)-(3-((R)-2,2-dimethylcyclopropyl)pyridin-2-yl)(4-(trifluoromethyl)phenyl)methyl)-3-(pyridin-3-yl)urea;
    • (S)-1-((3-fluoro-4-(trifluoromethyl)phenyl)(3-fluoropyridin-2-yl)methyl)-3-(pyridin-3-yl)urea; or
    • 1-((S)-(3-fluoro-4-(trifluoro-methyl)phenyl)-(3-(trifluoro-methyl)pyridin-2-yl)methyl)-3-((S)-1,1,1-trifluoropropan-2-yl)urea; or
      the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof
  • 62. The compound of embodiment 1, wherein the compound is (S)-1-((3-fluoro-4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)-methyl)-3-(pyridin-3-yl)urea or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof
  • 63. The compound of embodiment 1, wherein the compound is (S)-1-(pyridin-3-yl)-3-((4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)urea or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof.
  • 64. The compound of embodiment 1, wherein the compound is (S)-1-((3-(prop-1-yn-1-yl)pyridin-2-yl)(4-(trifluoromethyl)phenyl)methyl)-3-(pyridin-3-yl)urea or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof
  • 65. The compound of embodiment 1, wherein the compound is 1-((S)-(3-((R)-2,2-dimethylcyclopropyl)pyridin-2-yl)(4-(trifluoromethyl)phenyl)methyl)-3-(pyridin-3-yl)urea or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof
  • 66. The compound of embodiment 1, wherein the compound is (S)-1-((3-fluoro-4-(trifluoromethyl)phenyl)(3-fluoropyridin-2-yl)methyl)-3-(pyridin-3-yl)urea or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof
  • 67. The compound of embodiment 1, wherein the compound is 1-((S)-(3-fluoro-4-(trifluoro-methyl)phenyl)-(3-(trifluoro-methyl)pyridin-2-yl)methyl)-3-((S)-1,1,1-trifluoropropan-2-yl)urea or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof
  • 68. The compound of embodiment 1, wherein the compound is (S)-1-((3-fluoro-4-(trifluoromethoxy)phenyl)(3-fluoropyridin-2-yl)methyl)-3-(pyridin-3-yl)urea or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof
  • 69. The compound of embodiment 1, wherein the compound is (S)—N-((3-fluoro-4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)-morpholine-4-carboxamide or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof
  • 70. The compound or tautomer of any one of embodiments 1-4 or 9-69 in a neutral form.
  • 71. The compound of any one of embodiments 1-4 or 9-69 in a neutral form.
  • 72. The pharmaceutically-acceptably salt of the compound or the pharmaceutically acceptable salt of the tautomer of any one of embodiments 1-4 or 9-69.
  • 73. The pharmaceutically-acceptably salt of the compound of any one of embodiments 1-4 or 9-69.
  • 74. The pharmaceutically-acceptable salt of the compound of embodiment 73, wherein the salt is a trifluoroacetate or bis trifluoroacetate salt.
  • 75. A pharmaceutical composition comprising the compound according to any one of embodiments 1-4 or 9-69 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof, and a pharmaceutically-acceptable diluent or carrier.
  • 76. A method of treating acute, inflammatory and neuropathic pain, dental pain, general headache, migraine, cluster headache, mixed-vascular and non-vascular syndromes, tension headache, general inflammation, arthritis, rheumatic diseases, osteoarthritis, inflammatory bowel disorders, depression, anxiety, inflammatory eye disorders, inflammatory or unstable bladder disorders, psoriasis, skin complaints with inflammatory components, chronic inflammatory conditions, inflammatory pain and associated hyperalgesia and allodynia, neuropathic pain and associated hyperalgesia and allodynia, diabetic neuropathy pain, causalgia, sympathetically maintained pain, deafferentation syndromes, asthma, epithelial tissue damage or dysfunction, herpes simplex, disturbances of visceral motility at respiratory, genitourinary, gastrointestinal or vascular regions, wounds, burns, allergic skin reactions, pruritus, vitiligo, general gastrointestinal disorders, gastric ulceration, duodenal ulcers, diarrhea, gastric lesions induced by necrotising agents, hair growth, vasomotor or allergic rhinitis, bronchial disorders or bladder disorders in a subject, the method comprising administering the compound according to any one of embodiments 1-4 or 9-69 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof to the subject.
  • 77. The method of embodiment 76, wherein the subject is suffering from neuropathic pain.
  • 78. The method of embodiment 76, wherein the subject is suffering from migraine pain.
  • 79. The use of the compound according to any one of embodiments 1-4 or 9-69 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof in the preparation of a medicament.
  • 80. The use of the compound according to any one of embodiments 1-4 or 9-69 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof for treating acute, inflammatory and neuropathic pain, dental pain, general headache, migraine, cluster headache, mixed-vascular and non-vascular syndromes, tension headache, general inflammation, arthritis, rheumatic diseases, osteoarthritis, inflammatory bowel disorders, depression, anxiety, inflammatory eye disorders, inflammatory or unstable bladder disorders, psoriasis, skin complaints with inflammatory components, chronic inflammatory conditions, inflammatory pain and associated hyperalgesia and allodynia, neuropathic pain and associated hyperalgesia and allodynia, diabetic neuropathy pain, causalgia, sympathetically maintained pain, deafferentation syndromes, asthma, epithelial tissue damage or dysfunction, herpes simplex, disturbances of visceral motility at respiratory, genitourinary, gastrointestinal or vascular regions, wounds, burns, allergic skin reactions, pruritus, vitiligo, general gastrointestinal disorders, gastric ulceration, duodenal ulcers, diarrhea, gastric lesions induced by necrotising agents, hair growth, vasomotor or allergic rhinitis, bronchial disorders or bladder disorders in a subject.
  • 81. The use of embodiment 80, wherein the use is for treating neuropathic pain.
  • 82. The use of embodiment 80, wherein the use is for treating migraine.
  • 83. The compound according to any one of embodiments 1-4 or 9-69 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof for treating acute, inflammatory and neuropathic pain, dental pain, general headache, migraine, cluster headache, mixed-vascular and non-vascular syndromes, tension headache, general inflammation, arthritis, rheumatic diseases, osteoarthritis, inflammatory bowel disorders, depression, anxiety, inflammatory eye disorders, inflammatory or unstable bladder disorders, psoriasis, skin complaints with inflammatory components, chronic inflammatory conditions, inflammatory pain and associated hyperalgesia and allodynia, neuropathic pain and associated hyperalgesia and allodynia, diabetic neuropathy pain, causalgia, sympathetically maintained pain, deafferentation syndromes, asthma, epithelial tissue damage or dysfunction, herpes simplex, disturbances of visceral motility at respiratory, genitourinary, gastrointestinal or vascular regions, wounds, burns, allergic skin reactions, pruritus, vitiligo, general gastrointestinal disorders, gastric ulceration, duodenal ulcers, diarrhea, gastric lesions induced by necrotising agents, hair growth, vasomotor or allergic rhinitis, bronchial disorders or bladder disorders in a subject.
  • 84. The compound of embodiment 83 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof for treating neuropathic pain in a subject.
  • 85. The compound of embodiment 83 or the pharmaceutically-acceptable salt thereof, the tautomer thereof, the pharmaceutically-acceptable salt of the tautomer, or the mixture thereof for treating migraine in a subject.
  • EXAMPLES
  • Unless otherwise noted, all materials were obtained from commercial suppliers and used without further purification. All parts are by weight and temperatures are in degrees centigrade unless otherwise indicated. All microwave assisted reactions were conducted with a Smith Synthesizer from Biotage. Mass spectral data was determined by electrospray ionization technique. All examples were purified to >90% purity as determined by high-performance liquid chromatography. Unless otherwise stated, reactions were run at room temperature.
  • The following abbreviations are used:
  • CDI—carbonyldiimidazole
    DABCO—1,4-diazabicyclo[2.2.2]octane
    DCM—dichloromethane
    DIPEA—diisopropyl ethylamine
    DMSO—dimethyl sulfoxide
  • DMF—N,N-dimethylformamide
  • DPPA—diphenylphosphoryl azide
    THF—tetrahydrofuran
    Et2O—diethyl ether
    EtOAc—ethyl acetate
    EtOH—ethyl alcohol
    MeCN—acetonitrile
    MeOH—methyl alcohol
  • NBS—N-bromosuccinimide
  • n-BuLi—n-butyllithium
    t-BuLi—t-butyllithium
    TFA—trifluoroacetic acid
    h—hour
    min—min
    rt—room temperature (22-25° C.)
    mL milliliters
    μL microliters
    g grams
    μg micrograms
    mg milligrams
    μmoL micromolars
  • General Method of Preparation
  • The compounds described herein are prepared using techniques known to one skilled in the art through the reaction sequences depicted in schemes 1-4 as well as by other methods. Furthermore, in the following schemes, where specific acids, bases, reagents, coupling agents, solvents, etc. are mentioned, it is understood that other suitable acids, bases, reagents, coupling agents, solvents, etc. may be used and are included within the scope of the present invention.
  • Diarylamines used for the synthesis of compounds of the present invention were prepared as described in Scheme 1. 2-Formylpyridines of the formula (1) were treated with 2-methylpropane-2-sulfinamide and copper sulfate in DCM to give 2-methyl-N-(pyridin-2-ylmethylene)propane-2-sulfinamides of the formula (2a). The compounds of formula (2a) were treated with aryl or heteroaryl metal halides of formula (3) at low temperature to give sulfinamides of the formula (4). Hydrolysis of sulfinamides (4) with hydrochloric acid in MeOH gives diaryl amines of formula (5a).
  • Figure US20140235642A1-20140821-C00042
  • An alternative approach to diaryl amines of formula (5a) is shown in Scheme 2. Aryl or heteroaryl aldehydes of the formula (6) were treated with 2-methylpropane-2-sulfinamide and copper sulfate in DCM to give sulfinimines of the formula (7). The compounds of formula (7) were treated with aryl or heteroaryl metal halides of formula (8) at low temperature to give sulfinamides of the formula (4). Hydrolysis of sulfinamides (4) with hydrochloric acid in MeOH gives diaryl amines of formula (5a).
  • Figure US20140235642A1-20140821-C00043
  • The methods described in Scheme 1 and 2 can be adapted to provide an asymmetric syntheses using the appropriate (R)- or (S)-2-methylpropane-2-sulfinamides to give sulfinimines of the formula (2b) or (2c). Subsequent aryl metal addition and hydrolysis gave chiral amines of formula (5b) or (5c).
  • Figure US20140235642A1-20140821-C00044
  • The coupling reaction of diarylamines of formula (5a-c) with the various isocyanates (9) or amines (10) can be performed as shown in Scheme 4 to afford compounds of the present invention (Formula (I)).
  • Figure US20140235642A1-20140821-C00045
  • Experimental Procedures for Intermediates
  • Figure US20140235642A1-20140821-C00046
  • Figure US20140235642A1-20140821-C00047
  • Intermediate 1: (S)-(4-(Trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methanamine hydrochloride
  • Figure US20140235642A1-20140821-C00048
  • Step 1. (S,E)-2-Methyl-N-((3-(trifluoromethyl)pyridin-2-yl)methylene)propane-2-sulfinamide
  • To a solution of 3-(trifluoromethyl)picolinaldehyde (Frontier Scientific, 9.80 g, 56.0 mmol) and DCM (50 mL) was added (S)-2-methylpropane-2-sulfinamide (AK Scientific, 10.3 g, 85.0 mmol) and copper(II) sulfate (35.3 g, 221 mmol). After 1.5 h at rt, the reaction was filtered through a pad of Celite® brand filter agent and the pad of Celite® filter agent was rinsed with DCM. The filtrate was concentrated in vacuo to give a dark green oil. The oil thus obtained was loaded onto a silica gel column and eluted with 30% EtOAc in hexanes to give (S,E)-2-methyl-N-((3-(trifluoromethyl)pyridin-2-yl)methylene)propane-2-sulfinamide (13.2 g, 47.5 mmol, 85.0% yield), as a golden oil. 1H NMR (300 MHz, CDCl3) δ ppm 9.02 (d, J=4.3 Hz, 1H), 8.70 (d, J=1.3 Hz, 1H), 8.38 (d, J=7.7 Hz, 1H), 7.79 (dd, J=7.9, 4.8 Hz, 1H), 1.18 (s, 9H). MS (ESI pos. ion) m/z: 279.1 (M+H).
  • Figure US20140235642A1-20140821-C00049
  • Step 2. (S)-2-Methyl-N—((S)-(4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)propane-2-sulfinamide
  • To an oven dried flask containing magnesium (3.46 g, 143 mmol) and Et2O (120 mL) was added diisobutylaluminum hydride (0.950 mL, 0.950 mmol), and 1-bromo-4-(trifluoromethyl)benzene (1.0 mL, 7.3 mmol) was added dropwise. The solution was allowed to stir for ˜20 min during which time the reaction mixture changed from clear to a brownish tint. The reaction was placed in an ice bath and the remaining 1-bromo-4-(trifluoromethyl)benzene (11.5 mL, 83.7 mmol) was added dropwise over 20 minutes. In a separate flask, a solution of (S,E)-2-methyl-N-((3-(trifluoromethyl)pyridin-2-yl)methylene)propane-2-sulfinamide (13.22 g, 47.5 mmol) and THF (80 mL) was cooled to −78° C. for 10 min, and the Grignard solution was added to the solution of sulfinamide over 30 min. After 1 h, the reaction was quenched with saturated aqueous potassium sodium tartrate (10 mL). The reaction was poured into H2O (150 mL). The entire solution was filtered through a pad of Celite® brand filter agent and the Celite® filter agent was rinsed liberally with THF and EtOAc. The resulting filtrate was separated and the organic layers were concentrated in vacuo to give the product as a dark orange oil. The resulting oil was adsorbed onto a plug of silica gel and chromatographed through a Redi-Sep® pre-packed silica gel column (120 g), eluting with 0% to 40% EtOAc in hexanes, to provide (S)-2-methyl-N—((S)-(4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)propane-2-sulfinamide (14.83 g, 34.9 mmol, 77% yield) as a golden oil. 1H NMR (600 MHz, DMSO-d6) δ ppm 8.93 (d, J=4.8 Hz, 1H), 7.25 (d, J=7.8 Hz, 1H), 7.71-7.67 (m, 2H), 7.61-7.59 (m, 1H), 7.54 (d, J=8.4 Hz, 2H), 6.08 (d, J=9 Hz, 1H), 5.90 (d, J=9 Hz, 1H), 1.20 (s, 9H). MS (ESI pos. ion) m/z: 425.1 (M+H).
  • Figure US20140235642A1-20140821-C00050
  • Step 3. (S)-(4-(Trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methanamine hydrochloride
  • To a cooled (0° C.) stirring solution of ((S)-2-methyl-N—((S)-(4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)propane-2-sulfonamide (27 g, 63 mmol) in Et2O (270 mL) was added 4.0 M HCl in 1,4-dioxane (157 mL, 630 mmol, 10 equiv.) at 0° C. The reaction mixture was then stirred for 30 min at the same temperature. The reaction progress was monitored by TLC (50% EtOAc in petroleum ether). After completion of the reaction, the reaction mixture was concentrated under reduced pressure and triturated with Et2O to get a white solid which was filtered and dried to give (S)-(4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methanamine hydrochloride (14 g, 39 mmol, 70% yield) as a white solid. 1H NMR (600 MHz, DMSO-d6) δ ppm 9.26 (s, 3H), 9.08 (d, J=4.2 Hz, 1H), 8.35 (d, J=7.8 Hz, 1H), 7.82-7.77 (m, 3H), 7.67 (d, J=8.4 Hz, 2H), 5.94 (s, 1H). MS (ESI pos. ion) m/z: 321.1 (M+H) for free base.
  • Figure US20140235642A1-20140821-C00051
  • Figure US20140235642A1-20140821-C00052
  • Intermediate 2: (S)-(4-(Trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methanamine hydrochloride
  • Figure US20140235642A1-20140821-C00053
  • Step 1. 3-Fluoropicolinaldehyde
  • To a stirred solution of DABCO (262.4 g, 2342 mmol) in anhydrous Et2O (2.1 L) at −25° C. in a 10 L 3-neck round bottom flask was added n-BuLi (2.5 M in hexane, 938 mL, 2342 mmol). The mixture was stirred between −25° C. to −10° C. for 45 min. and then cooled to −70° C. To the above solution, was added 3-fluoropyridine (206.7 g, 2129 mmol) dropwise, and the reaction mixture was stirred between −70° C. to −60° C. for 1.5 h before DMF (344 mL, 4258 mmol) was added. The progress of reaction was monitored by TLC (5% EtOAc in Petroleum ether). After 1 h stirring at −70° C., water (800 mL) was added and the reaction was allowed to warm to rt. The layers were separated, and the aqueous layer was extracted with DCM (5×1 L). The combined organic layers were washed with brine and dried over sodium sulphate. After removal of solvent, the residue was purified by silica gel chromatography using a gradient of EtOAc in hexane to give 3-fluoropicolinaldehyde as a pale yellow oil. 1H-NMR (400 MHz, CDCl3): δ ppm 10.21 (s, 1H), 8.63 (t, J=2.2 Hz, 1H), 7.54-7.57 (m, 2H). MS (ESI pos. ion) m/z: 126.0 (M+H).
  • Figure US20140235642A1-20140821-C00054
  • Step 2. (S,E)-N-((3-Fluoropyridin-2-yl)methylene)-2-methylpropane-2-sulfinamide
  • A mixture of 3-fluoropicolinaldehyde (300 g, 2400 mmol), copper sulfate (572 g, 3600 mmol) and (S)-2-methylpropane-2-sulfinamide (319 g, 2640 mmol) in DCM (3 L) in a 10 L 3-neck round bottom flask was stirred for 3 h at rt. The progress of reaction was monitored by TLC (30% EtOAc in petroleum ether). After completion of reaction, the solid was filtered off and the filtrate was concentrated under vacuum. The residue was purified by column chromatography using silica (60-120 mesh) with 20% EtOAc in n-hexane as eluent to give (S,E)-N-((3-fluoropyridin-2-yl)methylene)-2-methylpropane-2-sulfinamide as a yellow oil. 1H-NMR (400 MHz, CDCl3): δ ppm 8.89 (s, 1H), 8.64 (s, 1H), 7.55 (t, J=8.4 Hz, 1H), 7.46 (d, J=3.6 Hz, 1H), 1.29 (s, 9H). MS (ESI pos. ion) m/z: 155.0 (M—O and t-Bu).
  • Figure US20140235642A1-20140821-C00055
  • Step-3. (S)—N—((S)-(3-Fluoropyridin-2-yl)(4-(trifluoromethyl)phenyl)methyl)-2-methylpropane-2-sulfinamide
  • To a stirred suspension of magnesium (170 g, 2365 mmol) in THF (1.35 L), was added 4-bromobenzotrifluoride (532 g, 2365 mmol). Stirring was continued for 4 h (cautious: slightly exothermic, cooled with a water bath if needed). The solution was cannulated to a stirred solution of (S,E)-N-((3-fluoropyridin-2-yl)methylene)-2-methylpropane-2-sulfinamide (270 g, 1182 mmol) in THF (1.3 L) at −78° C. dropwise. Stirring was continued for 1 h. The progress of reaction was monitored by TLC (50% EtOAc in petroleum ether). After completion of the reaction, the reaction mixture was quenched with saturated aqueous NH4Cl (2.5 L), and the solution extracted with Et2O (5×500 mL). The organic layers were combined, dried over Na2SO4, concentrated, and purified by column chromatography using silica (100-200 mesh) with 25-30% EtOAc in petroleum ether as eluent to give (S)—N—((S)-(3-fluoropyridin-2-yl)(4(trifluoromethyl)phenyl)methyl)-2-methylpropane-2-sulfinamide as a brown oil. 1H-NMR (400 MHz, CDCl3): δ ppm 8.45 (d, J=3.6 Hz, 1H), 7.73-7.78 (m, 1H), 7.71 (d, J=8.4 Hz, 2H), 7.63 (m, 2H), 7.43-7.48 (m, 1H), 6.23 (d, J=6.8 Hz, 1H), 5.99 (d, J=6.8 Hz, 1H), 1.36 (s, 9H). MS (ESI pos. ion) m/z: 375.1 (M—O and t-Bu).
  • Figure US20140235642A1-20140821-C00056
  • Step 4. (S)-(3-Fluoropyridin-2-yl)(4-(trifluoromethyl)phenyl)methanamine
  • To a cooled (0° C.) stirring solution of (S)—N—((S)-(3-fluoropyridin-2-yl)(4-(trifluoromethyl)phenyl)methyl)-2-methylpropane-2-sulfinamide (108 g, 288.8 mmol) in DCM:EtOH (1:1, 1080 mL), was added saturated HCl in 1,4-dioxane (216 mL). Stirring was continued for 2 h at 0° C. The progress of reaction was monitored by TLC (100% EtOAc). After completion of the reaction, the reaction mixture was concentrated and triturated with Et2O to give a white solid which was filtered and dried to give (S)-(3-fluoropyridin-2-yl)(4-(trifluoromethyl)phenyl)methanamine hydrochloride as a white solid. 1H-NMR (400 MHz, CDCl3) δ ppm 9.23 (s, 3H), 8.60 (d, J=4.8 Hz, 1H), 7.87 (d, J=1.2 Hz, 1H), 7.84 (d, J=8.4 Hz, 2H), 7.72 (d, J=8.4 Hz, 2H), 7.59-7.63 (m, 1H), 6.09 (s, 1H). MS (ESI pos. ion) m/z: 270.1 (M+H).
  • General Procedure for Preparation of Diarylmethanamines (Intermediates 3-40)
  • Additional diarylmethanamines were prepared as described in Scheme 5, Steps 2-3; substituting the appropriate starting materials. Variations in methods applied in Scheme 5, Step 2 of the various intermediate syntheses are elaborated below. The amine intermediates were isolated as either the hydrochloride salts or as the free bases.
  • Method A:
  • Figure US20140235642A1-20140821-C00057
  • Intermediate 3: (S)—N—((S)-(4-Methoxyphenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)-2-methylpropane-2-sulfinamide
  • Magnesium metal (0.095 g, 3.91 mmol) was activated using a crystal of iodine prior to addition of THF (1 mL). 1-Bromo-4-methoxybenzene (0.400 g, 2.139 mmol) was added, and the reaction was left without stirring for 5 minutes after which initiation was observed. Additional THF (15 mL) was added, and the resulting mixture was stirred for 2 hours. Next, (S,E)-2-methyl-N-((3-(trifluoromethyl)pyridin-2-yl)methylene)propane-2-sulfinamide (0.500 g, 1.797 mmol) was added, and the mixture was stirred for 10 minutes. The reaction was quenched by addition of saturated aqueous NH4Cl (10 mL). H2O (100 mL) and EtOAc (150 mL) were added, and the phases were mixed and separated. The organic layer was dried with magnesium sulfate and evaporated to dryness under reduced pressure. Purification using silica chromatography (hexane to EtOAc gradient) gave (S)—N—((S)-(4-methoxyphenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)-2-methylpropane-2-sulfinamide.
  • The sulfinamide prepared as described above was then subjected to hydrolysis conditions similar to those described above in Scheme 5, Step 3 to give Intermediate 3 in Table 1 below.
  • Method B:
  • Figure US20140235642A1-20140821-C00058
  • Intermediate 4: (S)—N—((S)-(3,4-Dichlorophenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)-2-methylpropane-2-sulfinamide
  • (S,E)-2-Methyl-N-((3-(trifluoromethyl)pyridin-2-yl)methylene)propane-2-sulfinamide (0.493 g, 1.772 mmol) was dissolved in dry THF (10 mL) and cooled in an ice bath. 3,4-Dichlorophenylmagnesium bromide (Aldrich, 0.5 M solution in THF, 4.0 mL, 2.0 mmol) was added, and the reaction mixture was stirred for 5 minutes. Saturated aqueous NH4Cl (10 mL), H2O (100 mL) and EtOAc (100 mL) were added, and the phases were mixed and separated. The organic layer was dried with magnesium sulfate and evaporated to dryness under reduced pressure. Purification using silica chromatography (hexane to EtOAc gradient) gave (S)—N—((S)-(3,4-dichlorophenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)-2-methylpropane-2-sulfinamide.
  • The sulfinamide prepared as described above was then subjected to hydrolysis conditions similar to those described above in Scheme 5, Step 3 to give Intermediate 4 in Table 1 below.
  • Method C:
  • Figure US20140235642A1-20140821-C00059
  • Intermediate 8: (S)—N—((S)-(3-Fluoropyridin-2-yl)(4-(trifluoromethoxy)phenyl)methyl)-2-methylpropane-2-sulfinamide
  • 1-Iodo-4-(trifluoromethoxy)benzene (1.00 g, 3.47 mmol) was dissolved in dry THF (10 mL) and cooled in an ice bath. Isopropylmagnesium chloride, lithium chloride complex (14% solution in THF, Aldrich, 3.07 mL, 2.82 mmol) was added, and the mixture was stirred for 10 min. A solution of (S,E)-N-((3-fluoropyridin-2-yl)methylene)-2-methylpropane-2-sulfinamide (0.643 g, 2.82 mmol) in dry THF (10 mL) was added, and the reaction was stirred. After 50 minutes, the reaction was quenched by addition of saturated aqueous NH4Cl (10 mL). H2O (100 mL) and EtOAc (150 mL) were added, and the phases were mixed and separated. The organic layer was dried with magnesium sulfate and evaporated to dryness under reduced pressure. Purification using silica chromatography (hexane to EtOAc gradient) gave the desired (S)—N—((S)-(3-fluoropyridin-2-yl)(4-(trifluoromethoxy)phenyl)methyl)-2-methylpropane-2-sulfinamide.
  • The sulfinamide was then subjected to hydrolysis conditions similar to those described above in Scheme 6, Step 4 to give Intermediate 8 in Table 1 below.
  • TABLE 1
    Diarylmethanamines prepared analogous to Scheme 5 and 6 (Intermediates 3-39).
    Aryl Halide Mol.
    Inter- or Grignard Formula
    mediate Method in Step 2 Structure Name (Mol. Wt.)
     3 A
    Figure US20140235642A1-20140821-C00060
    Figure US20140235642A1-20140821-C00061
    (S)-(4- methoxy- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methanamine hydrochloride C14H13F3N2O (282.26)
     4 B
    Figure US20140235642A1-20140821-C00062
    Figure US20140235642A1-20140821-C00063
    (S)-(3,4- dichlorophenyl) (3-(trifluoro- methyl)- pyridin-2-yl)- methanamine C13H9Cl2F3N2 (321.13)
     5 A
    Figure US20140235642A1-20140821-C00064
    Figure US20140235642A1-20140821-C00065
    (S)-(3-fluoro- 4-methoxy- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methanamine C14H12F4N2O (330.25)
     62 A
    Figure US20140235642A1-20140821-C00066
    Figure US20140235642A1-20140821-C00067
    (4-fluoro-3- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methanamine C14H9F7N2 (338.22)
     7 A
    Figure US20140235642A1-20140821-C00068
    Figure US20140235642A1-20140821-C00069
    (S)-(3-fluoro- 4-(trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methanamine C14H9F7N2 (338.22)
     8 C
    Figure US20140235642A1-20140821-C00070
    Figure US20140235642A1-20140821-C00071
    (S)-(3-fluoro- pyridin-2-yl)- (4-(trifluoro- methoxy)- phenyl)- methanamine C14H10F4N2O (286.22)
     9 A
    Figure US20140235642A1-20140821-C00072
    Figure US20140235642A1-20140821-C00073
    (4-fluoro-3- (trifluoro- methyl)- phenyl)(3- fluoropyridin- 2-yl)- methanamine C14H9F7N2 (338.22)
    10 C
    Figure US20140235642A1-20140821-C00074
    Figure US20140235642A1-20140821-C00075
    (S)-(4- chlorophenyl) (3-(trifluoro- methyl)- pyridin-2-yl)- methanamine C13H10ClF3N (286.68)
    11 C
    Figure US20140235642A1-20140821-C00076
    Figure US20140235642A1-20140821-C00077
    (S)-(8- chloroquinolin- 3-yl)(3- (trifluoro- methyl)- pyridin-2-yl)- methanamine C16H11ClF3N (337.73)
    12 C
    Figure US20140235642A1-20140821-C00078
    Figure US20140235642A1-20140821-C00079
    (S)-(7- methoxy- quinolin-3- yl)(3- (trifluoro- methyl)- pyridin-2-yl)- methanamine C17H14F3N3O (333.31)
    13 C
    Figure US20140235642A1-20140821-C00080
    Figure US20140235642A1-20140821-C00081
    (S)-(5- chloroquinolin- 3-yl)(3- (trifluoro- methyl)- pyridin-2-yl)- methanamine C16H11ClF3N3 (337.73)
    14 C
    Figure US20140235642A1-20140821-C00082
    Figure US20140235642A1-20140821-C00083
    (S)-quinolm- 3-yl(3- (trifluoro- methyl)- pyridin-2-yl)- methanamine C16H12F3N3 (303.28)
    15 A
    Figure US20140235642A1-20140821-C00084
    Figure US20140235642A1-20140821-C00085
    (S)-(3- chlorophenyl) (3-(trifluoro- methyl)- pyridin-2-yl)- methanamine C13H10ClF3N2 (286.68)
    162 A
    Figure US20140235642A1-20140821-C00086
    Figure US20140235642A1-20140821-C00087
    (3-chloro-5- fluoro- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methanamine C13H9ClF4N2 (304.67)
    17 A
    Figure US20140235642A1-20140821-C00088
    Figure US20140235642A1-20140821-C00089
    (S)- naphthalen-2- yl(3- (trifluoro- methyl)- pyridin-2-yl)- methanamine C17H13F3N2 (302.29)
    18 B
    Figure US20140235642A1-20140821-C00090
    Figure US20140235642A1-20140821-C00091
    (S)-(3-fluoro- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methanamine C13H10F4N2 (270.23)
    19 A
    Figure US20140235642A1-20140821-C00092
    Figure US20140235642A1-20140821-C00093
    (S)-(3- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methanamine C14H10F6N2 (320.23)
    20 C
    Figure US20140235642A1-20140821-C00094
    Figure US20140235642A1-20140821-C00095
    (S)-(8-fluoro- quinolin-3- yl)(3- (trifluoro- methyl)- pyridin-2-yl)- methanamine C16H11F4N3 (321.27)
    21 B
    Figure US20140235642A1-20140821-C00096
    Figure US20140235642A1-20140821-C00097
    (S)-m-tolyl(3- (trifluoro- methyl)- pyridin-2-yl)- methanamine C14H13F3N2 (266.26)
    22 C
    Figure US20140235642A1-20140821-C00098
    Figure US20140235642A1-20140821-C00099
    (S)-quinolin- 6-yl(3- (trifluoro- methyl)- pyridin-2-yl)- methanamine C16H12F3N3 (303.28)
    23 C
    Figure US20140235642A1-20140821-C00100
    Figure US20140235642A1-20140821-C00101
    (S)-(5- chloropyridin- 2-yl)(3- (trifluoro- methyl)- pyridin-2-yl)- methanamine C12H9ClF3N3 (287.67)
    24 C
    Figure US20140235642A1-20140821-C00102
    Figure US20140235642A1-20140821-C00103
    (S)-(8- methoxy- quinolin-3- yl)(3- (trifluoro- methyl)- pyridin-2-yl)- methanamine C14H10F6N2 (333.31)
    25 B
    Figure US20140235642A1-20140821-C00104
    Figure US20140235642A1-20140821-C00105
    (S)-(3- methoxy- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methanamine C14H13F3N2O (282.26)
    262 A
    Figure US20140235642A1-20140821-C00106
    Figure US20140235642A1-20140821-C00107
    (3- bromopyridin- 4-yl)(4- (trifluoro- methyl)- phenyl)- methanamine C13H10BrF3N2 (331.13)
    27 B
    Figure US20140235642A1-20140821-C00108
    Figure US20140235642A1-20140821-C00109
    (S)-(4-fluoro- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methanamine C13H10F4N2 (270.23)
    28 B
    Figure US20140235642A1-20140821-C00110
    Figure US20140235642A1-20140821-C00111
    (S)-phenyl(3- (trifluoro- methyl)- pyridin-2-yl)- methanamine C13H11F3N2 (252.24)
    29 B
    Figure US20140235642A1-20140821-C00112
    Figure US20140235642A1-20140821-C00113
    (S)-(4- ethylphenyl)- (3-(trifluoro- methyl)- pyridin-2-yl)- methanamine C15H15F3N2 (280.29)
    30 A
    Figure US20140235642A1-20140821-C00114
    Figure US20140235642A1-20140821-C00115
    (R)-(3-fluoro- 4-(trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- phenyl)- methanamine C15H10F7N (337.24)
    312 A
    Figure US20140235642A1-20140821-C00116
    Figure US20140235642A1-20140821-C00117
    (2- bromopyridin- 3-yl)(4- (trifluoro- methyl)- phenyl)- methanamine C13H10BrF3N2 (331.13)
    32 A
    Figure US20140235642A1-20140821-C00118
    Figure US20140235642A1-20140821-C00119
    (S)-(3-fluoro- 4-(trifluoro- methyl)- phenyl)(3- fluoropyridin- 2-yl)- methanamine C13H9F5N2 (288.22)
    332 A
    Figure US20140235642A1-20140821-C00120
    Figure US20140235642A1-20140821-C00121
    (3-(trifluoro- methyl)- pyridin-2-yl)- (6-(trifluoro- methyl)- pyridin-2-yl)- methanamine C13H9F6N3 (321.22)
    342 A
    Figure US20140235642A1-20140821-C00122
    Figure US20140235642A1-20140821-C00123
    (3-(trifluoro- methyl)- pyridin-2-yl)- (6-(trifluoro- methyl)- pyridin-3-yl)- methanamine C13H9F6N3 (321.22)
    352 A
    Figure US20140235642A1-20140821-C00124
    Figure US20140235642A1-20140821-C00125
    (3-chloro-5- fluoro- phenyl)(3- fluoropyridin- 2-yl)- methanamine C12H9ClF2N (254.66)
    361 A
    Figure US20140235642A1-20140821-C00126
    Figure US20140235642A1-20140821-C00127
    (S)-(2- bromophenyl) (4-(trifluoro- methyl)- phenyl)- methanamine C14H11BrF3N (330.14)
    371 A
    Figure US20140235642A1-20140821-C00128
    Figure US20140235642A1-20140821-C00129
    (S)-(3-fluoro- 4-(trifluoro- methyl)- phenyl)(2- (trifluoro- methyl)- phenyl)- methanamine C15H10F7N (337.24)
    381 A
    Figure US20140235642A1-20140821-C00130
    Figure US20140235642A1-20140821-C00131
    (S)-(4- (trifluoro- methyl)- phenyl)(4- (trifluoro- methyl)- pyridin-3-yl)- methanamine C14H10F6N2 (230.23)
    39 A
    Figure US20140235642A1-20140821-C00132
    Figure US20140235642A1-20140821-C00133
    (S)-(3-fluoro-4- (trifluoromethoxy) phenyl)(3- fluoropyridin- 2- yl)methanamine C13H9F5N2O (304.22)
    1These amines were prepared employing (R)-2-methylpropane-2-sulfinamide in the first step (Scheme 5). For reversal of stereochemistry observed in the Ellman sulfonylimine chemistry observed with 2-pyridyl substrates, see Kuduk, S. D.; DiPardo, R. M.; Chang, R. K.; Ng, C.; Bock, M. G. Tetrahedron Lett. 2004, 45, 6641-6643.
    2Prepared employing racemic 2-methylpropane-2-sulfinamide in the first step (Scheme 5)
  • Figure US20140235642A1-20140821-C00134
  • Figure US20140235642A1-20140821-C00135
  • Intermediate 40: (S)-(3-(Prop-1-yn-1-yl)pyridin-2-yl)(4-(trifluoromethyl)phenyl)methanamine hydrochloride
  • Figure US20140235642A1-20140821-C00136
  • Step-1. 3-Bromo-2-dibromethyl-pyridine
  • To a solution of 3-bromo-picoline (25 g, 0.145 mol) in CCl4 was added NBS (51.66 g, 0.29 mol) and benzoylperoxide (2.5 g, 0.018 mol). The resulting mixture was then gradually heated to reflux for 30 h. The reaction mixture was cooled to rt, the succinamide was filtered off, and the filtrate was concentrated under reduced pressure. The resulting residue was purified by flash column chromatography using silica (100-200 mesh) with EtOAc:hexane (1:9) as eluent to furnish 40.0 g pure 3-bromo-2-dibromomethyl-pyridine. 1H-NMR (400 MHz, CDCl3) δ ppm 7.67 (d, 1H), 7.86 (d, 1H), 7.15 (t, 2H).
  • Figure US20140235642A1-20140821-C00137
  • Step-2. 3-Bromo-pyridine-2-carbaldehyde
  • A suspension of 3-bromo-2-dibromomethyl-pyridine (10.0 g, 30.32 mmol) in morpholine (30.0 mL) was heated at 60° C. for 1 h. The reaction mixture was then cooled to rt and diluted with EtOAc (200 mL) followed by adjustment to pH 4 by adding citric acid (40.0 g). The reaction mixture was extracted with EtOAc (3×200 mL). The combined organic layers were dried over Na2SO4 and concentrated under reduced pressure. The resulting residue was purified by column chromatography using silica (100-200 mesh) and 3% EtOAc in hexane as eluent to give 3-bromo-pyridine-2-carbaldehyde. 1H-NMR (400 MHz, CDCl3) δ ppm 10.23 (s, 1H), 8.75 (d, 1H), 8.03 (d, 1H), 7.32 (t, 1H).
  • Figure US20140235642A1-20140821-C00138
  • Step 3. (S,E)-N-((3-Bromopyridin-2-yl)methylene)-2-methylpropane-2-sulfinamide
  • A mixture of 3-bromo piconaldehyde (10 g, 53.8 mmol), copper sulfate (3.98 mL, 81 mmol) and (S)-2-methylpropane-2-sulfinamide (6.84 g, 56.4 mmol) in DCM (100 mL) was stirred at rt overnight. The solid was filtered off and the filtrate concentrated under vacuum. The residue thus obtained was purified by column chromatography using silica (100-200 mesh) with 20% EtOAc in n-hexane as eluent to give (S,E)-N-((3-bromopyridin-2-yl)methylene)-2-methylpropane-2-sulfinamide as yellow oil. 1H-NMR (400 MHz, CDCl3): δ ppm 9.0 (s, 1H), 8.75 (d, 1H), 7.97 (d, 1H), 7.32 (t, 1H), 5.2 (d, 1H), 1.3 (s, 9H).
  • Figure US20140235642A1-20140821-C00139
  • Step 4. 2-Methyl-propane-2-sulfinic acid ((3-bromo-pyridin-2-yl)-(4-trifluoromethyl-phenyl)-methyl)-amide
  • To a stirred suspension of magnesium, (2.143 g, 88 mmol) in THF (50 mL) was added 4-bromobenzotrifluoride (5.06 mL, 36.2 mmol). Stirring was continued for 4 h (cautious: slightly exothermic, cooled with an water bath if needed). The solution was decanted from the resulting mixture and added to a stirred solution of N-(3-bromopyridin-2-yl)methylene)-2-methylpropane-2-sulfonamide (5.1 g, 17.64 mmol) in THF (100 mL) at −78° C. dropwise. Stirring was continued for another hour after the addition, and then the reaction was quenched with saturated aqueous NH4Cl, extracted with ether (3×20 mL), dried over Na2SO4, concentrated, and purified by column chromatography using silica (100-200 mesh) with 5% EtOAc in hexane as eluent to give the title compound as a brown oil.
  • Figure US20140235642A1-20140821-C00140
  • Step 5. (S)-tert-Butyl((3-bromopyridin-2-yl)(4-(trifluoromethyl)phenyl)methyl)carbamate
  • To a cooled (0° C.) stirring solution of 2-methyl-propane-2-sulfinic acid ((3-bromo-pyridin-2-yl)-(4-trifluoromethyl-phenyl)-methyl)-amide (5.0 g, 11.49 mmol) in DCM/EtOH (1:1, 60 mL), was added 4.0 M HCl in 1,4-dioxane (14.36 mL, 57.4 mmol). Stirring was continued for 2 h and then DIPEA (10.00 mL, 57.4 mmol) was added, followed by di-tert-butyl dicarbonate (4.00 mL, 17.23 mmol) addition. The resulting mixture was stirred at rt overnight, taken up in H2O, extracted in DCM (3×100 mL), dried over Na2SO4 and concentrated under reduced pressure. The residue thus obtained was purified by silica gel (100-200 mesh) column chromatography using 5% EtOAc in hexane as eluent to give the title compound as a white solid. 1H-NMR (400 MHz, DMSO-d6) δ ppm 8.5 (dd, 1H), 7.81 (dd, 1H), 7.52 (s, 4H), 7.2 (dd, 1H) 6.51 (d, 1H), 6.32 (d. 1H), 1.41 (s, 9H). MS (ESI pos. ion) m/z: 431.2, 433.2 (M+H).
  • Figure US20140235642A1-20140821-C00141
  • Step 6. (S)-(3-Bromopyridin-2-yl)(4-(trifluoromethyl)phenyl)methanamine hydrochloride
  • To a solution of (S)-tert-butyl((3-bromopyridin-2-yl)(4-(trifluoromethyl)phenyl)methyl)carbamate (2.0 g, 4.64 mmol) in MeOH (10 mL) was added hydrogen chloride (3.48 mL, 13.91 mmol) (4.0 M in 1,4-dioxane). The reaction was then stirred for 27 h at rt under N2 and then the reaction was concentrated in vacuo to give the amine (1.7 g) as a white solid which was used without further purification in subsequent steps. MS (ESI pos. ion) m/z: 331.0, 332.9 (M+H).
  • Figure US20140235642A1-20140821-C00142
  • Intermediate 41: (S)-(3-(Prop-1-yn-1-yl)pyridin-2-yl)(4-(trifluoromethyl)phenyl)methanamine hydrochloride
  • Figure US20140235642A1-20140821-C00143
  • Step 1. (S)-tert-Butyl ((3-(prop-1-yn-1-yl)pyridin-2-yl)(4-(trifluoromethyl)phenyl)methyl)carbamate
  • To a microwave vial containing (S)-tert-butyl ((3-bromopyridin-2-yl)(4-(trifluoromethyl)phenyl)methyl)carbamate (500 mg, 1.159 mmol) and a stir bar was added 1,4-dioxane (6 mL). To this solution was added tetrakis(triphenylphosphine)palladium(0) (67.0 mg, 0.058 mmol, 0.05 equiv.) and tributyl(prop-1-yn-1-yl)stannane (458 mg, 1.391 mmol, 1.2 equiv.). The vial was capped and irradiated in a microwave at 120° C. for 20 min. The vial was allowed to cool, diluted with hexanes (5 mL) and loaded directly to a normal phase silica gel column (80 g ISCO, 0 to 40% EtOAc in hexanes) to provide (S)-tert-butyl ((3-(prop-1-yn-1-yl)pyridin-2-yl)(4-(trifluoromethyl)phenyl)methyl)carbamate as a white solid.
  • Figure US20140235642A1-20140821-C00144
  • Step 2. (S)-(3-(Prop-1-yn-1-yl)pyridin-2-yl)(4-(trifluoromethyl)phenyl)methanamine hydrochloride
  • To a round bottom flask containing (S)-tert-butyl ((3-(prop-1-yn-1-yl)pyridin-2-yl)(4-(trifluoromethyl)phenyl)methyl)carbamate (400 mg, 1.025 mmol) was added DCM (8 mL). The resulting mixture was stirred at 23° C. for 2 min. Hydrogen chloride (4 mL, 4 N in 1,4-dioxane) was then added via syringe. The reaction was then stirred for 3 h and then the volatiles were removed via rotary evaporator. The solid was placed on high vacuum overnight to give (S)-(3-(prop-1-yn-1-yl)pyridin-2-yl)(4-(trifluoromethyl)phenyl)methanamine hydrochloride as a white solid.
  • Figure US20140235642A1-20140821-C00145
  • Intermediate 42: (1S)-(3-(2,2-Dimethylcyclopropyl)pyridin-2-yl)(4-(trifluoromethyl)phenyl)methanamine hydrochloride
  • A mixture of (S)-tert-butyl (3-bromopyridin-2-yl)(4-(trifluoromethyl)phenyl)methylcarbamate (1.50 g, 3.48 mmol), potassium (2,2-dimethylcyclopropyl)trifluoroborate (0.857 g, 4.87 mmol), potassium phosphate (2.58 g, 12.17 mmol), dicyclohexyl(2′,6′-dimethoxybiphenyl-2-yl)phosphine (0.314 g, 0.765 mmol), and palladium acetate (0.094 g, 0.417 mmol) in toluene/H2O (10:1, 11 mL) was heated at 100° C. for 24 h. The reaction mixture was then cooled, diluted with H2O, and extracted with EtOAc (3×). The combined extracts were dried over MgSO4, concentrated and purified by ISCO (silica gel, 10% EtOAc/hexanes) to give a colorless oil which was dissolved in DCM (3 mL) and then 4 mL of 4 M HCl in 1,4-dioxane was added. The solution was stirred at rt overnight and concentrated to dryness to give the title compound as an off white solid. MS (ESI pos. ion) m/z: 321.0 (M+H).
  • Figure US20140235642A1-20140821-C00146
  • Intermediate 43: (S)-(3-Allylpyridin-2-yl)(4-(trifluoromethyl)phenyl)methanamine hydrochloride
  • A mixture of (S)-tert-butyl (3-bromopyridin-2-yl)(4-(trifluoromethyl)phenyl)methylcarbamate (1.00 g, 2.319 mmol), allylboronic acid pinacol ester (0.507 mL, 3.01 mmol), cesium fluoride (0.171 mL, 4.64 mmol), and (Ph3P)4Pd (0.536 g, 0.464 mmol) in 1,4-dioxane (10 mL) was heated by microwave at 125° C. in 30 min. The mixture was then cooled, taken up in H2O, extracted with Et2O (3×), dried over MgSO4, concentrated and purified by ISCO (0-50% EtOAc/hexanes). The residue was dissolved in DCM (10 mL) and HCl in 4 M 1,4-dioxane (4.64 mL, 18.55 mmol) was added. Stirring was continued for 2 h, and the solution was then concentrated to dryness to give the title compound as a white solid. MS (ESI pos. ion) m/z: 293.0 (M+H).
  • Figure US20140235642A1-20140821-C00147
  • Intermediate 44: (S)-(3-Neopentylpyridin-2-yl)(4-(trifluoromethyl)phenyl)methanamine hydrochloride
  • A mixture of (S)-tert-butyl (3-bromopyridin-2-yl)(4-(trifluoromethyl)phenyl)methylcarbamate (0.500 g, 1.159 mmol), bis(tri-t-butylphosphine)palladium (0) (0.119 g, 0.232 mmol), and neopentylzinc(II) bromide (4.75 mL, 2.377 mmol) in THF (5 mL) was heated to 135° C. by microwave and stirred for 30 min. The reaction mixture was cooled, quenched with saturated aqueous NH4Cl, and extracted with EtOAc (3×). The extracts were dried over MgSO4, concentrated, and purified by ISCO (0-40% EtOAc/hyexanes) to give the carbamate intermediate. The carbamate was dissolved in DCM (5 mL) and 4 M HCl in 1,4-dioxane (2 mL) was added. The reaction mixture was then stirred at rt overnight and concentrated to dryness to give the title compound as a white solid. MS (ESI pos. ion) m/z: 323.0 (M+H).
  • Figure US20140235642A1-20140821-C00148
  • Intermediate 45: (S)-Pyridin-2-yl(4-(trifluoromethyl)phenyl)methanamine bis(2,2,2-trifluoroacetate)
  • Figure US20140235642A1-20140821-C00149
  • Step 1. (S)-tert-Butyl (pyridin-2-yl(4-(trifluoromethyl)phenyl)methyl)carbamate
  • To a solution of (S)-tert-butyl ((3-bromopyridin-2-yl)(4-(trifluoromethyl)phenyl)methyl)carbamate (Intermediate 40, Step 5) (2.24 g, 5.19 mmol) in MeOH (20 mL) was added palladium hydroxide, (20 wt % Pd (dry basis) on carbon, wet, degussa type) (0.365 g, 2.60 mmol). The resulting mixture was then stirred at rt under H2 (1 atm) overnight. The mixture was next filtered through Celite® brand filter agent and the Celite® filter agent was washed with a solution of MeOH/EtOAc (1:1, 3×20 mL). The combined filtrates were concentrated and dried to give the desired product as a yellow oil, which was used in the next step without further purification.
  • Figure US20140235642A1-20140821-C00150
  • Step 2. (S)-Pyridin-2-yl(4-(trifluoromethyl)phenyl)methanamine bis(2,2,2-trifluoroacetate)
  • To a solution of (S)-tert-butyl (pyridin-2-yl(4-(trifluoromethyl)phenyl)methyl)carbamate (1.83 g, 5.19 mmol) in DCM (12 mL) was added TFA (3.86 mL, 51.9 mmol). The resulting mixture was then stirred at rt for 1 h. The mixture was then concentrated and dried to give the title compound as a yellow oil which was used without further purification in the next step.
  • Figure US20140235642A1-20140821-C00151
  • Intermediate 46: (S)-[2,3′-Bipyridin]-2′-yl(4-(trifluoromethyl)phenyl)-methanaminedihydrochloride
  • A mixture of (S)-tert-butyl (3-bromopyridin-2-yl)(4-(trifluoromethyl)phenyl)methylcarbamate (Intermediate 40, Step 5) (0.711 g, 1.649 mmol), 2-(tributylstannyl)pyridine (0.910 g, 2.473 mmol), and (Ph3P)4Pd (0.381 g, 0.330 mmol) in 1,4-dioxane (10 mL) was heated by microwave at 125° C. for 30 min. The reaction mixture was then cooled, concentrated, and purified by ISCO (silica gel, 0-60% EtOAc/hexanes). The residue was dissolved in DCM (3 mL) and hydrogen chloride (3.30 mL, 13.19 mmol, 4.0 M in 1,4-dioxane) was added. After stirring for 2 h, the mixture was concentrated to dryness to give the title compound. MS (ESI pos. ion) m/z: 330.0 (M+H).
  • EXAMPLES
  • Figure US20140235642A1-20140821-C00152
  • Example 1 (S)-1-((3-Fluoro-4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)-pyridin-2-yl)methyl)-3-(pyridin-3-yl)urea bis(2,2,2-trifluoroacetate)
  • To a solution of (S)-(3-fluoro-4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methanamine (Intermediate 7) (566 mg, 1.673 mmol) and DIPEA (0.291 mL, 1.673 mmol) in DCM (5 mL) was added 3-isocyanatopyridine (201 mg, 1.673 mmol). The resulting mixture was stirred at rt for 1 h. The reaction was then concentrated to 1.0 mL, diluted with DMF (2.0 mL), filtered through a syringe filter, and purified by preparative reverse phase HPLC [Phenomenx Gemini Axia™-5μ C-18 column (150×30 mm), 10-100% MeCN/0.1% TFA in H2O]. The product-containing fractions were combined and the solvent was removed by lyophilization to give (S)-1-((3-fluoro-4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)-3-(pyridin-3-yl)urea bis(2,2,2-trifluoroacetate) as a white solid. 1H NMR (300 MHz, d4-MeOH): δ ppm 9.15 (br. s., 1H), 8.93 (d, J=4.1 Hz, 1H), 8.37 (br. s., 1H), 8.29-8.15 (m, 2H), 7.85 (dd, J=5.6, 8.3 Hz, 1H), 7.70-7.54 (m, 2H), 7.47-7.26 (m, 2H), 6.59 (s, 1H). MS (ESI pos. ion) m/z: 458.9 (M+H).
  • Figure US20140235642A1-20140821-C00153
  • General Urea Formation Procedure for Examples (2-147)
  • To a solution of amine (0.156 mmol), DIPEA (0.080 mL, 0.468 mmol, 3.0 equiv.) in DCM or DMF (1 mL) at rt was added the corresponding isocyanate (0.156 mmol). The reaction was then stirred 1 h at rt. Next, the reaction was diluted with DMF (1 mL), filtered through a syringe filter, and then it was purified by silica gel chromatography to provide the title compounds. Alternatively, the compounds were purified by preparative reverse phase HPLC [Phenomenx Gemini Axia™-5μ C-18 column (150×30 mm), 10-100% MeCN/0.1% TFA in H2O]. The product-containing fractions were combined and the solvent removed by lyophilization to provide the target compound as the TFA salts; or the product was dissolved in MeOH (1 mL) and washed through PL-HCO3 MP-resin, and the resin was further washed with MeOH (2×0.4 mL). The combined filtrates were then concentrated and dried in vacuo to give the title compounds as free bases; or the product containing fractions were concentrated, the solids dissolved in DCM and the organic layer extracted with saturated aqueous NaHCO3, the organic layer was dried, and concentrated to provide the title compounds as free bases. Compounds prepared using this general method are shown in Table 2.
  • TABLE 2
    Examples 2-147 prepared via urea formation analogous to Scheme 8.
    MS
    (ESI,
    Amine positive
    Ex. Inter- Isocyanate Product Product ion)
    # mediate Structure Structure Name M + H
    2 3
    Figure US20140235642A1-20140821-C00154
    Figure US20140235642A1-20140821-C00155
    (S)-1-((4- methoxy- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)-3- (pyridin-3-yl)- urea 403.1
    3 15
    Figure US20140235642A1-20140821-C00156
    Figure US20140235642A1-20140821-C00157
    (S)-1-((3- chlorophenyl)- (3-(trifluoro- methyl)- pyridin-2-yl)- methyl)-3- (pyridin-3-yl)- urea 407.0
    4 25
    Figure US20140235642A1-20140821-C00158
    Figure US20140235642A1-20140821-C00159
    (S)-1-((3- methoxy- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)-3- (pyridin-3-yl)- urea 403.1
    5 5
    Figure US20140235642A1-20140821-C00160
    Figure US20140235642A1-20140821-C00161
    (S)-1-((3- fluoro-4- methoxy- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)-3- (pyridin-3-yl)- urea 421.1
    6 14
    Figure US20140235642A1-20140821-C00162
    Figure US20140235642A1-20140821-C00163
    (S)-1-(pyridin- 3-yl)-3- (quinolin-3- yl(3-(trifluoro- methyl)- pyridin-2-yl)- methyl)urea 424.1
    7 21
    Figure US20140235642A1-20140821-C00164
    Figure US20140235642A1-20140821-C00165
    (S)-1-((3- methylphenyl) (3-(trifluoro- methyl)- pyridin-2-yl)- methyl)-3- (pyridin-3-yl)- urea 387.0
    8 18
    Figure US20140235642A1-20140821-C00166
    Figure US20140235642A1-20140821-C00167
    (S)-1-((3- fluorophenyl)- (3-(trifluoro- methyl)- pyridin-2-yl)- methyl)-3- (pyridin-3-yl)- urea 391.0
    9 19
    Figure US20140235642A1-20140821-C00168
    Figure US20140235642A1-20140821-C00169
    (S)-1-((3- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)-3- (pyridin-3-yl)- urea 441.0
    10 13
    Figure US20140235642A1-20140821-C00170
    Figure US20140235642A1-20140821-C00171
    (S)-1-((5- chloroquinolin- 3-yl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)-3- (pyridin-3-yl)- urea 458.0
    11 17
    Figure US20140235642A1-20140821-C00172
    Figure US20140235642A1-20140821-C00173
    (S)-1- (naphthalen-2- yl(3-(trifluoro- methyl)- pyridin-2-yl)- methyl)-3- (pyridin-3-yl)- urea 423.0
    12 11
    Figure US20140235642A1-20140821-C00174
    Figure US20140235642A1-20140821-C00175
    (S)-1-((8- chloroquinolin- 3-yl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)-3- (pyridin-3-yl)- urea 458.0
    13 4
    Figure US20140235642A1-20140821-C00176
    Figure US20140235642A1-20140821-C00177
    (S)-1-((3,4- dichlorophenyl) (3-(trifluoro- methyl)- pyridin-2-yl)- methyl)-3- (pyridin-3-yl)- urea 440.9
    14 20
    Figure US20140235642A1-20140821-C00178
    Figure US20140235642A1-20140821-C00179
    (S)-1-((8- fluoro- quinolin-3-yl)- (3-(trifluoro- methyl)- pyridin-2-yl)- methyl)-3- (pyridin-3-yl)- urea 442.0
    15 8
    Figure US20140235642A1-20140821-C00180
    Figure US20140235642A1-20140821-C00181
    (S)-1-((3- fluoropyridin- 2-yl)(4- (trifluoro- methoxy)- phenyl)- methyl)-3- (pyridin-3-yl)- urea 407.0
    16 24
    Figure US20140235642A1-20140821-C00182
    Figure US20140235642A1-20140821-C00183
    (S)-1-((8- methoxy- quinolin-3-yl)- (3-(trifluoro- methyl)- pyridin-2-yl)- methyl)-3- (pyridin-3-yl)- urea 454.0
    17 22
    Figure US20140235642A1-20140821-C00184
    Figure US20140235642A1-20140821-C00185
    (S)-1-(pyridin- 3-yl)-3- (quinolin-6- yl(3-(trifluoro- methyl)- pyridm-2-yl)- methyl)urea 424.1
    18 12
    Figure US20140235642A1-20140821-C00186
    Figure US20140235642A1-20140821-C00187
    (S)-1-((7- methoxy- quinolin-3-yl)- (3-(trifluoro- methyl)- pyridin-2-yl)- methyl)-3- (pyridin-3-yl)- urea 454.0
    19 27
    Figure US20140235642A1-20140821-C00188
    Figure US20140235642A1-20140821-C00189
    (S)-1-((4- fluorophenyl)- (3-(trifluoro- methyl)- pyridin-2-yl)- methyl)-3- (pyridin-3-yl)- urea 391.0
    20 28
    Figure US20140235642A1-20140821-C00190
    Figure US20140235642A1-20140821-C00191
    (S)-1- (phenyl(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)-3- (pyridin-3-yl)- urea 373.1
    21 6
    Figure US20140235642A1-20140821-C00192
    Figure US20140235642A1-20140821-C00193
    1-((4-fluoro-3- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)-3- (pyridin-3-yl)- urea (2,2,2- trifluoro- acelate) 459.0
    22 9
    Figure US20140235642A1-20140821-C00194
    Figure US20140235642A1-20140821-C00195
    1-((4-fluoro-3- (trifluoro- methyl)- phenyl)(3- fluoropyridin- 2-yl)methyl)- 3-(pyridin-3- yl)urea (2,2,2- trifluoro- acetate) 409.0
    23 16
    Figure US20140235642A1-20140821-C00196
    Figure US20140235642A1-20140821-C00197
    1-((3-chloro- 5-fluoro- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)-3- (pyridin-3-yl)- urea (2,2,2- trifluoro- acetate) 424.8
    24 35
    Figure US20140235642A1-20140821-C00198
    Figure US20140235642A1-20140821-C00199
    1-((3-chloro- 5-fluoro- phenyl)(3- fluoropyridin- 2-yl)methyl)- 3-(pyridin-3- yl)urea (2,2,2- trifluoro- acetate) 374.9
    25 34
    Figure US20140235642A1-20140821-C00200
    Figure US20140235642A1-20140821-C00201
    1-(pyridin-3- yl)-3-((3- (trifluoro- methyl)- pyridin-2-yl)- (6-(trifluoro- methyl)- pyridin-3-yl)- methyl)urea (2,2,2- trifluoro- acetate) 441.9
    26 10
    Figure US20140235642A1-20140821-C00202
    Figure US20140235642A1-20140821-C00203
    (S)-1-((4- chlorophenyl)- (3-(trifluoro- methyl)- pyridin-2-yl)- methyl)-3- (pyridin-3-yl)- urea 407.0
    27 29
    Figure US20140235642A1-20140821-C00204
    Figure US20140235642A1-20140821-C00205
    (S)-1-((4- ethylphenyl)- (3-(trifluoro- methyl)- pyridin-2-yl)- methyl)-3- (pyridin-3-yl)- urea 401.1
    28 40
    Figure US20140235642A1-20140821-C00206
    Figure US20140235642A1-20140821-C00207
    (S)-1-((3- bromopyridin- 2-yl)(4- (trifluoro- methyl)- phenyl)- methyl)-3- (pyridin-3-yl)- urea 452.0
    29 32
    Figure US20140235642A1-20140821-C00208
    Figure US20140235642A1-20140821-C00209
    (S)-1-((3- fluoro-4- (trifluoro- methyl)- phenyl)(3- fluoropyridin- 2-yl)methyl)- 3-(pyridin-3- yl)urea 409.0
    30 45
    Figure US20140235642A1-20140821-C00210
    Figure US20140235642A1-20140821-C00211
    (S)-1-(pyridin- 2-yl(4- (trifluoro- methyl)- phenyl)- methyl)-3- (pyridin-3-yl)- urea 373.0
    31 1
    Figure US20140235642A1-20140821-C00212
    Figure US20140235642A1-20140821-C00213
    (S)-1-phenyl- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea 440.0
    32 1
    Figure US20140235642A1-20140821-C00214
    Figure US20140235642A1-20140821-C00215
    (S)-1- isopropyl-3- ((4-(trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea 406.0
    33 1
    Figure US20140235642A1-20140821-C00216
    Figure US20140235642A1-20140821-C00217
    1-((S)-1- (naphthalen-1- yl)ethyl)-3- ((S)-(4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 518.0
    34 1
    Figure US20140235642A1-20140821-C00218
    Figure US20140235642A1-20140821-C00219
    (S)-1-(3- phenylpropyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 482.0
    35 1
    Figure US20140235642A1-20140821-C00220
    Figure US20140235642A1-20140821-C00221
    (S)-1-(phenyl formate)-3- ((4-(trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 484.0
    36 1
    Figure US20140235642A1-20140821-C00222
    Figure US20140235642A1-20140821-C00223
    (S)-1-(2- (benzo[d][l,3] dioxol-5-yl)- ethyl)-3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 512.0
    37 1
    Figure US20140235642A1-20140821-C00224
    Figure US20140235642A1-20140821-C00225
    (S)-1-(3- bromophenyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 519.9
    38 1
    Figure US20140235642A1-20140821-C00226
    Figure US20140235642A1-20140821-C00227
    (S)-ethyl 2-(3- ((4-(trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)- ureido)acetate (2,2,2- trifluoro- acetate) 450.0
    39 1
    Figure US20140235642A1-20140821-C00228
    Figure US20140235642A1-20140821-C00229
    (S)-1- (benzofuran- 5-yl)-3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 480.0
    40 40
    Figure US20140235642A1-20140821-C00230
    Figure US20140235642A1-20140821-C00231
    (S)-1-(4- cyanophenyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea 476.0
    41 40
    Figure US20140235642A1-20140821-C00232
    Figure US20140235642A1-20140821-C00233
    (S)-1-((3- bromopyridin- 2-yl)(4- (trifluoro- methyl)- phenyl)- methyl)-3- (tert-butyl)- urea 430.0
    42 41
    Figure US20140235642A1-20140821-C00234
    Figure US20140235642A1-20140821-C00235
    (S)-1-(tert- butyl)-3-((3- (prop-1-yn-1- yl)pyridin-2- yl)(4- (trifluoro- methyl)- phenyl)- methyl)urea 390.0
    43 43
    Figure US20140235642A1-20140821-C00236
    Figure US20140235642A1-20140821-C00237
    (S)-1-((3- allylpyridin-2- yl)(4- (trifluoro- methyl)- phenyl)- methyl)-3- (pyridin-3-yl)- urea 413.0
    44 1
    Figure US20140235642A1-20140821-C00238
    Figure US20140235642A1-20140821-C00239
    (S)-1-(pyridin- 3-yl)-3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea 441.0
    45 1
    Figure US20140235642A1-20140821-C00240
    Figure US20140235642A1-20140821-C00241
    (S)-1-(tert- butyl)-3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea 420.0
    46 1
    Figure US20140235642A1-20140821-C00242
    Figure US20140235642A1-20140821-C00243
    (S)-1-(4- cyanophenyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea 465.0
    47 1
    Figure US20140235642A1-20140821-C00244
    Figure US20140235642A1-20140821-C00245
    (S)-1-(4- fluorophenyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea 458.0
    48 1
    Figure US20140235642A1-20140821-C00246
    Figure US20140235642A1-20140821-C00247
    (S)-1-(2,4- difluoro- phenyl)-3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea 476.0
    49 1
    Figure US20140235642A1-20140821-C00248
    Figure US20140235642A1-20140821-C00249
    (S)-1-(3,5- difluoro- phenyl)-3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea 476.0
    50 1
    Figure US20140235642A1-20140821-C00250
    Figure US20140235642A1-20140821-C00251
    (S)-1-(2,6- difluoro- phenyl)-3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea 476.0
    51 41
    Figure US20140235642A1-20140821-C00252
    Figure US20140235642A1-20140821-C00253
    (S)-1-((3- (prop-1-yn-1- yl)pyridin-2- yl)(4- (trifluoro- methyl)- phenyl)- methyl)-3- (pyridin-3-yl)- urea 411.0
    52 1
    Figure US20140235642A1-20140821-C00254
    Figure US20140235642A1-20140821-C00255
    (S)-1-(3- methoxy- phenyl)-3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 470.0
    53 1
    Figure US20140235642A1-20140821-C00256
    Figure US20140235642A1-20140821-C00257
    (S)-1-(3- (methylsulfonyl) phenyl)-3- ((4-(trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea 518.0
    54 1
    Figure US20140235642A1-20140821-C00258
    Figure US20140235642A1-20140821-C00259
    1-(tetrahydro- 2H-pyran-3- yl)-3-((S)-(4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea 448.0
    55 1
    Figure US20140235642A1-20140821-C00260
    Figure US20140235642A1-20140821-C00261
    (S)-1-(3,5- dimethylisoxazol- 4-yl)-3- ((4-(trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea 459.0
    56 1
    Figure US20140235642A1-20140821-C00262
    Figure US20140235642A1-20140821-C00263
    1- (tetrahydrofuran- 3-yl)-3- ((S)-(4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea 434.0
    57 1
    Figure US20140235642A1-20140821-C00264
    Figure US20140235642A1-20140821-C00265
    (S)-1- (pyrimidin-5- yl)-3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea 442.0
    58 1
    Figure US20140235642A1-20140821-C00266
    Figure US20140235642A1-20140821-C00267
    (S)-1- (quinolin-6- yl)-3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea 491.0
    59 1
    Figure US20140235642A1-20140821-C00268
    Figure US20140235642A1-20140821-C00269
    (S)-1- (quinolin-3- yl)-3-((4- (trifluoro- methyl)-
    Figure US20140235642A1-20140821-P00899
    491.0
    60 1
    Figure US20140235642A1-20140821-C00270
    Figure US20140235642A1-20140821-C00271
    (S)-1- (quinolin-4- yl)-3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea 491.0
    61 42
    Figure US20140235642A1-20140821-C00272
    Figure US20140235642A1-20140821-C00273
    1-((S)-(3-((R)- 2,2- dimethylcyclo- propyl)- pyridin-2-yl)- (4-(trifluoro- methyl)- phenyl)- methyl)-3- (pyridin-3-yl)- urea 441.0
    62 44
    Figure US20140235642A1-20140821-C00274
    Figure US20140235642A1-20140821-C00275
    (S)-1-((3- neopentylpyridin- 2-yl)(4- (trifluoro- methyl)- phenyl)- methyl)-3- (pyridin-3-yl)- urea 443.0
    63 1
    Figure US20140235642A1-20140821-C00276
    Figure US20140235642A1-20140821-C00277
    (S)-1-(2,6- dibromo-4- fluorophenyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 614.0
    64 1
    Figure US20140235642A1-20140821-C00278
    Figure US20140235642A1-20140821-C00279
    1-((3- Sulfonylcyclo- pentyl)- methyl)-3- ((S)-(4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 496.0
    65 1
    Figure US20140235642A1-20140821-C00280
    Figure US20140235642A1-20140821-C00281
    1-((S)-3- methylbutan- 2-yl)-3-((S)- (4-(trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 434.0
    66 1
    Figure US20140235642A1-20140821-C00282
    Figure US20140235642A1-20140821-C00283
    (S)-ethyl 3-(3- ((4-(trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)- ureido)- propanoate (2,2,2- trifluoro- acetate) 464.0
    67 1
    Figure US20140235642A1-20140821-C00284
    Figure US20140235642A1-20140821-C00285
    (S)-N-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methylcarbamoyl) benzamide (2,2,2- trifluoro- acetate) 468.0
    68 1
    Figure US20140235642A1-20140821-C00286
    Figure US20140235642A1-20140821-C00287
    (S)-ethyl 4-(3- ((4-(trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)- ureido)- butanoate (2,2,2- trifluoro- acetate) 478.0
    69 1
    Figure US20140235642A1-20140821-C00288
    Figure US20140235642A1-20140821-C00289
    (S)-1-(3- methyl-5- phenylisoxazol- 4-yl)-3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 521.0
    70 1
    Figure US20140235642A1-20140821-C00290
    Figure US20140235642A1-20140821-C00291
    1-((R)-1- phenylethyl)- 3-((S)-(4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 468.0
    71 1
    Figure US20140235642A1-20140821-C00292
    Figure US20140235642A1-20140821-C00293
    (S)-1-(2-tert- butylphenyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 496.0
    72 1
    Figure US20140235642A1-20140821-C00294
    Figure US20140235642A1-20140821-C00295
    (S)-1-(3- fluorobenzyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 472.0
    73 1
    Figure US20140235642A1-20140821-C00296
    Figure US20140235642A1-20140821-C00297
    (S)-1-(2- chlorobenzyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 488.0
    74 1
    Figure US20140235642A1-20140821-C00298
    Figure US20140235642A1-20140821-C00299
    (S)-1-(3,4- dimethoxy- phenyl)-3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 500.0
    75 1
    Figure US20140235642A1-20140821-C00300
    Figure US20140235642A1-20140821-C00301
    (S)-benzyl 4- (3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)- ureido)- piperidine-1- carboxylate (2,2,2- trifluoro- acetate) 581.0
    76 1
    Figure US20140235642A1-20140821-C00302
    Figure US20140235642A1-20140821-C00303
    (S)-1-(2,6- dichloropyridin- 4-yl)-3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 509.0
    77 1
    Figure US20140235642A1-20140821-C00304
    Figure US20140235642A1-20140821-C00305
    (S)-1-(3-(5- methyl-1,2,4- oxadiazol-3- yl)phenyl)-3- ((4-(trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 522.0
    78 1
    Figure US20140235642A1-20140821-C00306
    Figure US20140235642A1-20140821-C00307
    (S)-methyl 4- (methylthio)- 2-(3-((S)-(4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)- ureido)- butanoate (2,2,2- trifluoro- acetate) 510.0
    79 1
    Figure US20140235642A1-20140821-C00308
    Figure US20140235642A1-20140821-C00309
    (S)-methyl 2- (3-((S)-(4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)- ureido)- propanoate (2,2,2- trifluoro- acetate) 450.0
    80 1
    Figure US20140235642A1-20140821-C00310
    Figure US20140235642A1-20140821-C00311
    1-((S)-1-(4- methoxy- phenyl)ethyl)- 3-((S)-(4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 498.0
    81 1
    Figure US20140235642A1-20140821-C00312
    Figure US20140235642A1-20140821-C00313
    (S)-N-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methylcarbamoyl)- ethylamide (2,2,2- trifluoro- acetate) 436.0
    82 1
    Figure US20140235642A1-20140821-C00314
    Figure US20140235642A1-20140821-C00315
    (S)-1-(2,3- dimethoxy- phenethyl)-3- ((4-(trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 528.0
    83 1
    Figure US20140235642A1-20140821-C00316
    Figure US20140235642A1-20140821-C00317
    1-((S)-hexan- 2-yl)-3-((S)- (4-(trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 448.0
    84 1
    Figure US20140235642A1-20140821-C00318
    Figure US20140235642A1-20140821-C00319
    1-((S)-1-(3- methoxy- phenyl)ethyl)- 3-((S)-(4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 498.0
    85 1
    Figure US20140235642A1-20140821-C00320
    Figure US20140235642A1-20140821-C00321
    (S)-1-(2- methylbenzyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 468.0
    86 1
    Figure US20140235642A1-20140821-C00322
    Figure US20140235642A1-20140821-C00323
    (S)-1-(3- acetylphenyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 482.0
    87 1
    Figure US20140235642A1-20140821-C00324
    Figure US20140235642A1-20140821-C00325
    (S)-1-(2- fluorobenzyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 472.0
    88 1
    Figure US20140235642A1-20140821-C00326
    Figure US20140235642A1-20140821-C00327
    (S)-1-(2,6- diethylphenyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 496.0
    89 1
    Figure US20140235642A1-20140821-C00328
    Figure US20140235642A1-20140821-C00329
    (S)-1-(2-ethyl- 6- methylphenyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 482.0
    90 1
    Figure US20140235642A1-20140821-C00330
    Figure US20140235642A1-20140821-C00331
    (S)-ethyl 3-(3- ((4-(trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)- ureido)- benzoate (2,2,2- trifluoro- acetate) 512.0
    91 1
    Figure US20140235642A1-20140821-C00332
    Figure US20140235642A1-20140821-C00333
    (S)-1-(2-ethyl- 6- isopropylphenyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 510.0
    92 1
    Figure US20140235642A1-20140821-C00334
    Figure US20140235642A1-20140821-C00335
    (S)-1-(2- isopropyl-6- methylphenyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 496.0
    93 1
    Figure US20140235642A1-20140821-C00336
    Figure US20140235642A1-20140821-C00337
    (S)-1-(3,5- dimethoxy- phenethyl)-3- ((4-(trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 528.0
    94 1
    Figure US20140235642A1-20140821-C00338
    Figure US20140235642A1-20140821-C00339
    (S)-ethyl 2-(3- ((S)-(4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)- ureido)- propanoate (2,2,2- trifluoro- acetate) 464.0
    95 1
    Figure US20140235642A1-20140821-C00340
    Figure US20140235642A1-20140821-C00341
    (S)-1-(3,4- dimethoxy- phenethyl)-3- ((4-(trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 528.0
    96 1
    Figure US20140235642A1-20140821-C00342
    Figure US20140235642A1-20140821-C00343
    (S)-methyl 3- methyl-2-(3- ((S)-(4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)- ureido)- butanoate (2,2,2- trifluoro- acetate) 478.0
    97 1
    Figure US20140235642A1-20140821-C00344
    Figure US20140235642A1-20140821-C00345
    (S)-1-(3- cyanophenyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 465.0
    98 1
    Figure US20140235642A1-20140821-C00346
    Figure US20140235642A1-20140821-C00347
    ethyl 3- methyl-2-(3- ((S)-(4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)- ureido)- butanoate (2,2,2- trifluoro- acetate) 492.0
    99 1
    Figure US20140235642A1-20140821-C00348
    Figure US20140235642A1-20140821-C00349
    (3S)-methyl 3- methyl-2-(3- ((S)-(4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)- ureido)- pentanoate (2,2,2- trifluoro- acetate) 492.0
    100 1
    Figure US20140235642A1-20140821-C00350
    Figure US20140235642A1-20140821-C00351
    (S)-1-(3- nitrophenyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 485.0
    101 1
    Figure US20140235642A1-20140821-C00352
    Figure US20140235642A1-20140821-C00353
    1-(1-(4- bromophenyl) ethyl)-3-((S)- (4-(trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 546.0
    102 1
    Figure US20140235642A1-20140821-C00354
    Figure US20140235642A1-20140821-C00355
    (S)-methyl 3- (3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)- ureido)- benzoate (2,2,2- trifluoro- acetate) 498.0
    103 1
    Figure US20140235642A1-20140821-C00356
    Figure US20140235642A1-20140821-C00357
    (S)-dimethyl 5-(3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)- ureido)- isophthalate (2,2,2- trifluoro- acetate) 556.0
    104 1
    Figure US20140235642A1-20140821-C00358
    Figure US20140235642A1-20140821-C00359
    (S)-butyl 2-(3- ((4-(trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)- ureido)acetate (2,2,2- trifluoro- acetate) 478.0
    105 1
    Figure US20140235642A1-20140821-C00360
    Figure US20140235642A1-20140821-C00361
    (S)-1-(2-(3- (prop-1-en-2- yl)phenyl)- propan-2-yl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 522.0
    106 1
    Figure US20140235642A1-20140821-C00362
    Figure US20140235642A1-20140821-C00363
    (S)-1-(3,5- dimethylphen yl)-3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 468.0
    107 1
    Figure US20140235642A1-20140821-C00364
    Figure US20140235642A1-20140821-C00365
    1-((S)-1- phenylethyl)- 3-((S)-(4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 468.0
    108 1
    Figure US20140235642A1-20140821-C00366
    Figure US20140235642A1-20140821-C00367
    (S)-1-(2,5- dimethylphenyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 468.0
    109 1
    Figure US20140235642A1-20140821-C00368
    Figure US20140235642A1-20140821-C00369
    (S)-1-benzyl- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 454.0
    110 1
    Figure US20140235642A1-20140821-C00370
    Figure US20140235642A1-20140821-C00371
    (S)-1-(2,5- dimethoxy- phenethyl)-3- ((4-(trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 528.0
    111 1
    Figure US20140235642A1-20140821-C00372
    Figure US20140235642A1-20140821-C00373
    (S)-1-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)-3- (3,4,5- trimethoxy- benzyl)urea (2,2,2- trifluoro- acetate) 544.0
    112 1
    Figure US20140235642A1-20140821-C00374
    Figure US20140235642A1-20140821-C00375
    (S)-1-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)-3- (3,4,5- trimethoxy- phenyl)urea (2,2,2- trifluoro- acetate) 530.0
    113 1
    Figure US20140235642A1-20140821-C00376
    Figure US20140235642A1-20140821-C00377
    (S)-1- (thiophen-2- yl)-3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 446.0
    114 31
    Figure US20140235642A1-20140821-C00378
    Figure US20140235642A1-20140821-C00379
    1-((2- bromopyridin- 3-yl)(4- (trifluoro- methyl)- phenyl)- methyl)-3- (pyridin-3-yl)- urea (2,2,2- trifluoro- acetate) 450.9 452.9
    115 36
    Figure US20140235642A1-20140821-C00380
    Figure US20140235642A1-20140821-C00381
    (S)-1-((2- bromophenyl) (4-(trifluoro- methyl)- phenyl)- methyl)-3- (pyridin-3-yl)- urea 2,2,2- trifluoro- acetate (2,2,2- trifluoro- acetate) 449.9 451.9
    116 36
    Figure US20140235642A1-20140821-C00382
    Figure US20140235642A1-20140821-C00383
    (S)-1-((2- bromophenyl) (4-(trifluoro- methyl)- phenyl)- methyl)-3-tert- butylurea 428.9 431.0
    117 30
    Figure US20140235642A1-20140821-C00384
    Figure US20140235642A1-20140821-C00385
    (R)-1-((3- fluoro-4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- phenyl)- methyl)-3- (pyridin-3-yl)- urea (2,2,2- trifluoro- acetate) 458.0
    118 2
    Figure US20140235642A1-20140821-C00386
    Figure US20140235642A1-20140821-C00387
    (S)-methyl 2- (3-((3-fluoro- pyridin-2-yl)- (4-(trifluoro- methyl)- phenyl)- methyl)- ureido)- benzoate 448.0
    119 38
    Figure US20140235642A1-20140821-C00388
    Figure US20140235642A1-20140821-C00389
    (S)-1-(pyridin- 3-yl)-3-((4- (trifluoro- methyl)- phenyl)(4- (trifluoro- methyl)- pyridin-3-yl)- methyl)urea bis(2,2,2- trifluoro- acetate) 440.9
    120 26
    Figure US20140235642A1-20140821-C00390
    Figure US20140235642A1-20140821-C00391
    1-((3- bromopyridin- 4-yl)(4- (trifluoro- methyl)- phenyl)- methyl)-3- (pyridin-3-yl)- urea (2,2,2- trifluoro- acetate) 450.9 452.9
    121 37
    Figure US20140235642A1-20140821-C00392
    Figure US20140235642A1-20140821-C00393
    (S)-1-((3- fluoro-4- (trifluoro- methyl)- phenyl)(2- (trifluoro- methyl)- phenyl)- methyl)-3- (pyridin-3-yl)- urea 458.0
    122 1
    Figure US20140235642A1-20140821-C00394
    Figure US20140235642A1-20140821-C00395
    (S)-1-(pyridin- 3-yl)-3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)- thiourea 457.0
    123 1
    Figure US20140235642A1-20140821-C00396
    Figure US20140235642A1-20140821-C00397
    (S)-1-tert- butyl-3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)- thiourea 436.0
    124 1
    Figure US20140235642A1-20140821-C00398
    Figure US20140235642A1-20140821-C00399
    (S)-1-(3- iodophenyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 566.0
    125 1
    Figure US20140235642A1-20140821-C00400
    Figure US20140235642A1-20140821-C00401
    (S)-1-(4- bromobenzyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 532.0 534.0
    126 1
    Figure US20140235642A1-20140821-C00402
    Figure US20140235642A1-20140821-C00403
    (S)-1-(3- ethoxyphenyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 484.1
    127 1
    Figure US20140235642A1-20140821-C00404
    Figure US20140235642A1-20140821-C00405
    (S)-1- (cyclohexylmethyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 460.2
    128 1
    Figure US20140235642A1-20140821-C00406
    Figure US20140235642A1-20140821-C00407
    1-((R)-1-(3- methoxy- phenyl)ethyl)- 3-((S)-(4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 498.1
    129 1
    Figure US20140235642A1-20140821-C00408
    Figure US20140235642A1-20140821-C00409
    1-((S)-2,3- dihydro-1H- inden-1-yl)-3- ((S)-(4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 480.1
    130 1
    Figure US20140235642A1-20140821-C00410
    Figure US20140235642A1-20140821-C00411
    1-((R)-1-(4- methoxy- phenyl)ethyl)- 3-((S)-(4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 498.1
    131 1
    Figure US20140235642A1-20140821-C00412
    Figure US20140235642A1-20140821-C00413
    (S)-1-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)-3- (2,4,4- trimethylpentan- 2-yl)urea (2,2,2- trifluoro- acetate) 476.2
    132 1
    Figure US20140235642A1-20140821-C00414
    Figure US20140235642A1-20140821-C00415
    (S)-1-(2,6- dichlorophenethyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 536.1
    133 1
    Figure US20140235642A1-20140821-C00416
    Figure US20140235642A1-20140821-C00417
    (S)-1-(2- ethoxybenzyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 498.1
    134 1
    Figure US20140235642A1-20140821-C00418
    Figure US20140235642A1-20140821-C00419
    (S)-1-(4- chlorobenzyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 488.1
    135 1
    Figure US20140235642A1-20140821-C00420
    Figure US20140235642A1-20140821-C00421
    (S)-1-(2,4- dichlorobenzyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 522.0
    136 1
    Figure US20140235642A1-20140821-C00422
    Figure US20140235642A1-20140821-C00423
    (S)-methyl 3- phenyl-2-(3- ((S)-(4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)- ureido)- propanoate (2,2,2- trifluoro- acetate) 526.1
    137 1
    Figure US20140235642A1-20140821-C00424
    Figure US20140235642A1-20140821-C00425
    (S)-1-(2- (methylthio)- phenyl)-3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 486.1
    138 1
    Figure US20140235642A1-20140821-C00426
    Figure US20140235642A1-20140821-C00427
    (S)-1-(4- acetylphenyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 482.1
    139 1
    Figure US20140235642A1-20140821-C00428
    Figure US20140235642A1-20140821-C00429
    (S)-1-(4- fluoro-3- nitrophenyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 503.1
    140 1
    Figure US20140235642A1-20140821-C00430
    Figure US20140235642A1-20140821-C00431
    (S)-1-(4- methyl-3- nitrophenyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 499.1
    141 1
    Figure US20140235642A1-20140821-C00432
    Figure US20140235642A1-20140821-C00433
    (S)-1-(2- methoxy-4- nitrophenyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 515.1
    142 1
    Figure US20140235642A1-20140821-C00434
    Figure US20140235642A1-20140821-C00435
    (S)-1-(2- methyl-3- nitrophenyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 499.1
    143 1
    Figure US20140235642A1-20140821-C00436
    Figure US20140235642A1-20140821-C00437
    (S)-1-(5- methyl-2- nitrophenyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 499.1
    144 1
    Figure US20140235642A1-20140821-C00438
    Figure US20140235642A1-20140821-C00439
    (S)-1-(4- fluorobenzyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 472.1
    145 1
    Figure US20140235642A1-20140821-C00440
    Figure US20140235642A1-20140821-C00441
    (S)-1-(o- tolyl)-3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 454.1
    146 1
    Figure US20140235642A1-20140821-C00442
    Figure US20140235642A1-20140821-C00443
    (S)-1-(2- fluoro-5- methylphenyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 472.1
    147 1
    Figure US20140235642A1-20140821-C00444
    Figure US20140235642A1-20140821-C00445
    (S)-1-(3- methylbenzyl)- 3-((4- (trifluoro- methyl)- phenyl)(3- (trifluoro- methyl)- pyridin-2-yl)- methyl)urea (2,2,2- trifluoro- acetate) 468.1
    Figure US20140235642A1-20140821-P00899
    indicates data missing or illegible when filed
  • Figure US20140235642A1-20140821-C00446
  • Example 148 (S)-1-(4-(5-Oxo-4,5-dihydro-1,2,4-oxadiazol-3-yl)phenyl)-3-((4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)urea
  • A 25 mL round-bottomed flask containing a solution of 3-(4-aminophenyl)-1,2,4-oxadiazol-5(4H)-one 2,2,2-trifluoroacetate (0.107 g, 0.367 mmol) and CDI (0.140 g, 0.863 mmol) in anhydrous DCM (3.5 mL) was treated with DIPEA (0.200 mL, 1.150 mmol) and stirred for 3.5 h at rt. A solution of (S)-(4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methanamine hydrochloride (Intermediate 1) (0.118 g, 0.368 mmol) in anhydrous DCM (3.5 mL) was added followed by DIPEA (0.200 mL, 1.150 mmol), and the reaction mixture was stirred at rt for 12 h. The reaction mixture was then diluted with DCM (10 mL) and water (5 mL) and then absorbed onto a Varian Chem Elut (Diatomaceous Earth) cartridge. The organic filtrates were collected and concentrated to dryness. The resulting product was dissolved in DMSO/MeOH (1/1) (2.0 mL) and loaded on a Gilson HPLC system for purification using a MeCN/H2O 0.1% TFA gradient and Phenomenx Gemini Axia-5μ C-18 column (150×30 mm). The solvent was removed from the pure fractions in the GENEVAC and the residue was dried under high vacuum to yield the title compound as an amorphous off-white solid. 1H NMR (400 MHz, DMSO-d6): δ ppm 12.76 (s, 1H), 9.24 (s, 1H), 8.98 (d, J=3.9 Hz, 1H), 8.33-8.21 (m, 1H), 7.78-7.61 (m, 6H), 7.59-7.46 (m, 4H), 6.51 (d, J=8.4 Hz, 1H) MS (ESI pos. ion) m/z: 524.2 (M+H).
  • Figure US20140235642A1-20140821-C00447
  • General Urea Formation Procedure for Examples
  • To a solution of amine (0.341 mmol) in DCM (2.0 mL) was added CDI (55.4 mg, 0.341 mmol). The solution was then stirred for 0.5 h at rt. The reaction was next treated with a solution of a second amine (selected from Intermediates 1-46) and DIPEA (0.075 mL, 0.429 mmol) in DCM (1.0 mL). After 1 h, the reaction was concentrated in vacuo. The product was then purified by either reverse phase HPLC (0-100% MeCN/(0.1% TFA in H2O) or silica gel chromatography to provide the target compounds as either TFA salts or free bases. Compounds prepared using this general method (Scheme 9) are shown in Table 3.
  • TABLE 3
    Examples 149-158 prepared via urea formation analogous to Scheme 9.
    Amine MS (ESI,
    Amine Coupling positive
    Inter- Partner Product Product ion)
    Ex # mediate Structure Structure Name M + H
    149 2
    Figure US20140235642A1-20140821-C00448
    Figure US20140235642A1-20140821-C00449
    (S)-1-((3- fluoropyridin- 2-yl)(4- (trifluoro- methyl)- phenyl)- methyl)-3-(2- oxoindolin-5- 445.1
    150 41
    Figure US20140235642A1-20140821-C00450
    Figure US20140235642A1-20140821-C00451
    1-((S)-(3- (prop-1-yn-1- yl)pyridin-2- yl)(4-(trifluoro- methyl)-phenyl)- methyl)-3- ((S)-1,1,1-trifluoro- propan-2-yl)-urea 430.1
    151 7
    Figure US20140235642A1-20140821-C00452
    Figure US20140235642A1-20140821-C00453
    1-((S)-(3-fluoro-4- (trifluoro- methyl)- phenyl)(3-(trifluoro- methyl)-pyridin-2-yl)- methyl)-3- ((S)-1,1,1-trifluoro- propan-2-yl)-urea (2,2,2-trifluoro- acetate) 478.0
    152 7
    Figure US20140235642A1-20140821-C00454
    Figure US20140235642A1-20140821-C00455
    1-((S)-(3- fluoro-4-(trifluoro- methyl)-phenyl)(3- (trifluoro- methyl)-pyridin- 2-yl)-methyl)-3- ((R)-1,1,1-trifluoro- propan-2-yl)-urea (2,2,2-trifluoro- acetate) 478.0
    153 1
    Figure US20140235642A1-20140821-C00456
    Figure US20140235642A1-20140821-C00457
    1-((S)-(4- (trifluoro- methyl)-phenyl)(3- (trifluoro- methyl)-pyridin- 2-yl)-methyl)-3- ((S)-1,1,1-trifluoro- propan-2-yl)-urea (2,2,2-trifluoro- acetate) 460.0
    154 36
    Figure US20140235642A1-20140821-C00458
    Figure US20140235642A1-20140821-C00459
    1-((S)-(2- bromophenyl) (4-(trifluoro- methyl)-phenyl)- methyl)-3- ((S)-1,1,1-trifluoro- propan-2-yl)-urea 468.9 470.9
    155 7
    Figure US20140235642A1-20140821-C00460
    Figure US20140235642A1-20140821-C00461
    (S)-1-((3- fluoro-4-(trifluoro- methyl)-phenyl)(3- (trifluoro- methyl)-pyridin- 2-yl)-methyl)-3- (pyrimidin-5-yl)urea (2,2,2-trifluoro- acetate) 460.0
    156 2
    Figure US20140235642A1-20140821-C00462
    Figure US20140235642A1-20140821-C00463
    1-((S)-(3- fluoropyridin- 2-yl)(4-(trifluoro- methyl)-phenyl)- methyl)-3- ((S)-1,1,1-trifluoro- propan-2-yl)-urea 410.0
    157 40
    Figure US20140235642A1-20140821-C00464
    Figure US20140235642A1-20140821-C00465
    1-((S)-(3- bromopyridin- 2-yl)(4-(trifluoro- methyl)-phenyl)- methyl)-3- ((S)-1,1,1-trifluoro- propan-2-yl)-urea 470.0
    158 43
    Figure US20140235642A1-20140821-C00466
    Figure US20140235642A1-20140821-C00467
    1-((S)-(3- allylpyridin-2- yl)(4-(trifluoro- methyl)-phenyl)- methyl)-3- ((S)-l,l,l-trifluoro- propan-2-yl)-urea 432.0
  • Figure US20140235642A1-20140821-C00468
  • Example 159 (S)-1-(2-Methoxypyrimidin-5-yl)-3-((4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)urea
  • To a solution of 2-methoxypyrimidine-5-carboxylic acid (57 mg, 0.370 mmol), DIPEA (0.161 mL, 0.925 mmol), and toluene (3 mL) was added DPPA (0.104 mL, 0.481 mmol). The reaction was stirred at 80° C. for 2 h. (S)-(4-(Trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methanamine hydrochloride (Intermediate 1) (134 mg, 0.376 mmol) was then added as a solid in one portion. After 16 h, the reaction was concentrated in vacuo and the crude product was adsorbed onto a plug of silica gel and chromatographed through a Redi-Sep® pre-packed silica gel column (12 g), eluting with 0-100% EtOAc in hexane, to provide the title compound as an off-white solid. 1H NMR (300 MHz, CDCl3): δ ppm 8.96 (s, 2H), 8.90 (d, J=3.8 Hz, 1H), 8.22 (d, J=7.5 Hz, 1H), 8.05 (d, J=6.9 Hz, 1H), 7.63-7.45 (m, 6H), 6.85 (d, J=7.7 Hz, 1H), 4.07 (s, 3H). MS (ESI pos. ion) m/z: 472.0 (M+H).
  • Figure US20140235642A1-20140821-C00469
  • General Urea Formation Procedure for Examples (160-166)
  • To a solution of a carboxylic acid (0.370 mmol), DIPEA (0.925 mmol, 2.5 equiv.) in toluene (3 mL) was added DPPA (0.104 mL, 0.481 mmol, 1.3 equiv.). The reaction was then stirred at 80° C. for 2 hours. An amine (Intermediates 1-46) (0.376 mmol, 1.0 equiv.) was then added. The resulting reaction mixture was then stirred for 16 h at rt. The reaction was then concentrated, and the product was purified by either reverse phase HPLC (0-100% MeCN/(0.1% TFA in H2O) or silica gel chromatography to provide the target compounds as either TFA salts or free bases. Compounds prepared using this general method (Scheme 10) are shown in Table 4.
  • TABLE 4
    Examples 160-166 prepared via urea formation analogous to Scheme 10.
    MS (ESI,
    Amine positive
    Inter- Acid Product Product ion)
    Ex # mediate Structure Structure Name M + H
    160 2
    Figure US20140235642A1-20140821-C00470
    Figure US20140235642A1-20140821-C00471
    (S)-1-((3- fluoropyridin- 2-yl)(4- (trifluoro- methyl)- phenyl)- methyl)-3-(2- methoxy- pyrimidin-5- yl)urea 422.0
    161 2
    Figure US20140235642A1-20140821-C00472
    Figure US20140235642A1-20140821-C00473
    (S)-1-((3- fluoropyridin- 2-yl)(4- (trifluoro- methyl)- phenyl)- methyl)-3-(1- methyl-2-oxo- 1,2- dihydropyridin- 4-yl)urea 421.1
    162 2
    Figure US20140235642A1-20140821-C00474
    Figure US20140235642A1-20140821-C00475
    (S)-1-((3- fluoropyridin- 2-yl)(4- (trifluoro- methyl)- phenyl)- methyl)-3-(1- methyl-6-oxo- 1,6- dihydropyridin- 3-yl)urea 421.1
    163 2
    Figure US20140235642A1-20140821-C00476
    Figure US20140235642A1-20140821-C00477
    (S)-1-((3- fluoropyridin- 2-yl)(4- (trifluoro- methyl)- phenyl)- methyl)-3-(2- oxo-1,2- dihydropyridin- 3-yl)urea 407.1
    164 2
    Figure US20140235642A1-20140821-C00478
    Figure US20140235642A1-20140821-C00479
    (S)-1-((3- fluoropyridin- 2-yl)(4- (trifluoro- methyl)- phenyl)- methyl)-3-(2- oxo-1,2- dihydropyridin- 4-yl)urea 407.1
    165 2
    Figure US20140235642A1-20140821-C00480
    Figure US20140235642A1-20140821-C00481
    (S)-1-((3- fluoropyridin- 2-yl)(4- (trifluoro- methyl)- phenyl)- methyl)-3-(6- oxo-1,6- dihydropyridin- 3-yl)urea 407.1
    166 2
    Figure US20140235642A1-20140821-C00482
    Figure US20140235642A1-20140821-C00483
    (S)-1-((3- fluoropyridin- 2-yl)(4- (trifluoro- methyl)- phenyl)- methyl)-3-(2- oxoindolin-6- yl)urea 445.1
  • ADDITIONAL EXAMPLES
  • Figure US20140235642A1-20140821-C00484
  • Example 167 (S)—N-((3-Fluoro-4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)morpholine-4-carboxamide 2,2,2-trifluoroacetate
  • To a solution of (S)-(3-fluoro-4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methanamine (Intermediate 7) (100 mg, 0.296 mmol) and DIPEA (0.101 mL, 0.591 mmol) in DCM (1.0 mL) was added morpholine-4-carbonyl chloride (66.3 mg, 0.443 mmol). The resulting reaction mixture was stirred at rt for 19 h. The reaction was then diluted with DMF (2 mL), filtered through a syringe filter, and purified by reverse phase HPLC (Phenomenx Gemini Axia™-5μ C-18 column (150×30 mm) 10-100% MeCN/0.1% TFA in H2O). The product-containing fractions were combined, and the solvent was removed by lyophilization to give the title compound as a white solid. 1H NMR (300 MHz, d4-MeOH) δ ppm 8.66-8.43 (m, 1H), 7.84 (d, J=7.9 Hz, 1H), 7.38-7.12 (m, 2H), 7.07-6.83 (m, 2H), 6.26 (s, 1H), 3.35 (peak obscured by solvent), 3.08 (br. s., 4H), 2.98 (br. s., 2H). MS (ESI pos. ion) m/z: 452.0 (M+H).
  • Figure US20140235642A1-20140821-C00485
  • Example 168 (S)-1-((3-Propylpyridin-2-yl)(4-(trifluoromethyl)phenyl)methyl)-3-(pyridin-3-yl)urea
  • A solution of (S)-1-((3-allylpyridin-2-yl)(4-(trifluoromethyl)phenyl)methyl)-3-(pyridin-3-yl)urea (Example 43) (0.055 g, 0.133 mmol) and Pd (10 wt % on carbon, 0.020 g, 0.133 mmol) in MeOH was hydrogenated under H2 (1 atm) at rt for 4 h. The catalyst was filtered off, and the filtrate was concentrated to give the title compound as a white solid. 1H NMR (300 MHz, d4-MeOH): δ ppm 8.60-8.38 (m, 2H), 8.12 (d, J=4.4 Hz, 1H), 7.93 (d, J=8.3 Hz, 1H), 7.74-7.43 (m, 5H), 7.40-7.21 (m, 2H), 6.42 (s, 1H), 2.89-2.51 (m, 2H), 1.74-1.35 (m, 2H), 0.95 (t, J=7.2 Hz, 3H). MS (ESI pos. ion) m/z: 415.0 (M+H).
  • Figure US20140235642A1-20140821-C00486
  • Example 169 (S)-tert-Butyl (pyridin-2-yl(4-(trifluoromethyl)phenyl)methyl)carbamate 2,2,2-trifluoroacetate
  • To a flame-dried round bottom flask was added (5)-tert-butyl ((3-bromopyridin-2-yl)(4-(trifluoromethyl)phenyl)methyl)carbamate (Intermediate 40, Step 5) (195 mg, 0.452 mmol) and THF (2 mL). The solution was cooled in a dry ice/acetone bath for 5 minutes and then it was treated with 1.7 M t-BuLi in pentane (0.6 mL, 1.020 mmol) and allowed to stir for 1 min. The solution was then treated dropwise with acetone (0.10 mL, 1.362 mmol). The solution was then allowed to warm to rt as the cooling bath warmed. After 5 minutes, LC-MS indicated that the major components were starting bromide and des-bromide product. The reaction was concentrated in vacuo, taken up in MeOH (3 mL) and purified by reverse-phase preparative HPLC (Shimadzu) on a Phenomenex Gemini column (10 micron, C18, 110 Å, Axia, 100×50 mm) eluting at 90 mL/min with a linear gradient of 20% to 90% MeCN (0.1% TFA) in water (0.1% TFA) over 10 minutes to give the title compound as a white solid after lyophilization. 1H NMR (300 MHz, CDCl3) δ ppm 8.76 (d, J=5.1 Hz, 1H), 8.11-7.90 (m, 1H), 7.68-7.39 (m, 6H), 6.88-6.63 (m, 1H), 6.26-6.01 (m, 1H), 1.42 (s, 9H). MS (ESI pos. ion) m/z: 353.0 (M+H).
  • Figure US20140235642A1-20140821-C00487
  • Example 170 (S)-Ethyl ((4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)carbamate
  • To a mixture of (S)-(4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methanamine (Intermediate 1) (0.0610 g, 0.190 mmol) and DIPEA (0.066 mL, 0.381 mmol) in MeCN (1 mL) was added ethyl carbonochloridate (0.027 mL, 0.286 mmol). The mixture was then stirred at rt overnight. The mixture was diluted with saturated aqueous NaHCO3 and extracted with DCM. The organic phase was dried over Na2SO4 and concentrated in vacuo. The resulting product was purified by silica gel chromatography: 5-50% EtOAc-hexanes to give the title compound as a colorless oil. 1H NMR (400 MHz, CDCl3) δ ppm 8.83 (d, J=4.30 Hz, 1H), 7.99 (d, J=7.24 Hz, 1H), 7.46-7.60 (m, 4H), 7.40 (dd, J=7.73, 4.79 Hz, 1H), 6.54 (d, J=7.43 Hz, 1H), 6.41 (d, J=8.22 Hz, 1H), 4.11 (q, J=7.17 Hz, 2H), 1.17-1.30 (m, 3H). MS (ESI pos. ion) m/z: 393.0 (M+H).
  • Figure US20140235642A1-20140821-C00488
  • Example 171 1-(4-Fluorophenyl)-3-(phenyl(4-(trifluoromethyl)phenyl)methyl)urea
  • Figure US20140235642A1-20140821-C00489
  • Step 1. Phenyl(4-(trifluoromethyl)phenyl)methanone
  • A solution of 4-(trifluoromethyl)benzhydrol (1.836 g, 7.3 mmol) in anhydrous DCM (65 mL) was treated with manganese(IV) oxide (<5 micron, activated) (5.7 g, 66 mmol). The resulting suspension was stirred at rt for 12 days. The catalyst was removed by filtration through a Celite® brand filter agent pad. The filtrate was concentrated, and the resulting solid was dried under high vacuum to afford the title compound as a white solid.
  • Figure US20140235642A1-20140821-C00490
  • Step 2. Phenyl(4-(trifluoromethyl)phenyl)methanone O-methyl oxime
  • To a 100-mL round-bottomed flask, phenyl(4-(trifluoromethyl)phenyl)methanone (1.0105 g, 4.039 mmol) and methoxyamine (0.4208 g, 5.038 mmol) were dissolved in pyridine (10 mL). The mixture was stirred at rt overnight. Additional methoxyamine (0.1625 g) was added, and the solution was stirred at rt for 7 h. EtOAc was added and the organic phase was washed with water, 1 N HCl (2×), water, and then with brine. The organic phase was dried over sodium sulfate, filtered and concentrated in vacuo to afford a clear oil. The oil thus obtained was purified by Biotage (EtOAc/hexanes 0-20%) to afford the title compound (1.12 g, 99.4%) as a clear oil.
  • Figure US20140235642A1-20140821-C00491
  • Step 3. 1-(4-Fluorophenyl)-3-(phenyl(4-(trifluoromethyl)phenyl)methyl)urea
  • To a 5 mL microwave vial, phenyl(4-(trifluoromethyl)phenyl)methanone O-methyl oxime (0.1541 g, 0.552 mmol) and Pd/C (10 wt %, 0.0345 g, 0.324 mmol) were mixed into MeOH (2 mL). The reaction mixture was evacuated under vacuum and refilled with hydrogen (2×). The mixture was hydrogenated under balloon pressure of hydrogen at rt for 45 min. Next, 2 M NH3 in MeOH (1 mL) was added and the hydrogenation was resumed under balloon pressure of hydrogen for 1 h. The Pd catalyst was removed via filtration through a pad of Celite® brand filter agent. Evaporation of solvent resulted in 0.1375 g of clear oil. The oil thus obtained was dissolved in DCM (5 mL) and 4-fluorophenyl isocyanate (0.0800 mL, 0.662 mmol) was added. The mixture was then stirred at rt for 1 h. Next, the reaction mixture was purified by Biotage™ (0-50% EtOAc/hexanes) twice to obtain the title compound as a white solid. 1H NMR (300 MHz, DMSO) δ ppm 8.52 (s, 1H), 7.72 (d, J=8.2 Hz, 2H), 7.54 (d, J=8.2 Hz, 2H), 7.40-7.23 (m, 8H), 7.06 (dd, J=8.6, 8.2 Hz, 2H), 6.05 (d, J=7.2 Hz, 1H). MS (ESI pos. ion) m/z: 389.0 (M+H).
  • Figure US20140235642A1-20140821-C00492
  • Example 172 (S)-Benzyl (pyridin-2-yl(4-(trifluoromethyl)phenyl)methyl)carbamate 2,2,2-trifluoroacetate
  • Figure US20140235642A1-20140821-C00493
  • Step 1. (S)-Benzyl ((3-bromopyridin-2-yl)(4-(trifluoromethyl)phenyl)methyl)carbamate
  • To an ice bath cooled solution of (S)-(3-bromopyridin-2-yl)(4-(trifluoromethyl)phenyl)methanamine hydrochloride (Intermediate 40) (1.20 g, 2.2 mmol), DIPEA (1.50 mL, 8.6 mmol), and DCM (20 mL) was added benzyl chloroformate (0.40 mL, 2.8 mmol). The resulting mixture was allowed to warm to rt as the bath warmed. After 16 h, the reaction was eluted through a cartridge of silica gel (25 g) with DCM. The filtrate was concentrated in vacuo to give the title compound as a light yellow oil. MS (ESI pos. ion) m/z: 464.9, 466.9 (M+H).
  • Figure US20140235642A1-20140821-C00494
  • Step 2. (S)-Benzyl (pyridin-2-yl(4-(trifluoromethyl)phenyl)methyl)carbamate 2,2,2-trifluoroacetate
  • To a round bottom flask was added (S)-benzyl ((3-bromopyridin-2-yl)(4-(trifluoromethyl)phenyl)methyl)carbamate (1.20 g, 1995 μmol), Pd2(dba)3 (183 mg, 200 μmol), 2-dicyclohexylphosphino-2′,4′,6′-triisopropylbiphenyl (XPhos) (190 mg, 399 μmol), and THF (20 mL). To this mixture was added (2-(1,3-dioxolan-2-yl)ethyl)zinc(II) bromide (0.5M in Et2O, 10 mL, 5 mmol). The resulting mixture was stirred at rt for 16 h. The reaction was then heated at 70° C. for 5 h. The reaction was next treated with another equivalent of catalyst, ligand and zinc reagent and heating was continued. After a further 5 days of heating, the reaction was filtered through a plug of silica gel and the plug washed with 50% EtOAc/hexanes. The filtrate was concentrated in vacuo and adsorbed onto a plug of silica gel and purified by silica gel chromatography (40 g SiO2 0-25% EtOAc/hexanes) to provide (S)-benzyl (3-((1,3-dioxolan-2-yl)methyl)pyridin-2-yl)(4-(trifluoromethyl)phenyl)methylcarbamate (175 mg, 19% yield) as a brown solid. Further purification of mixed fractions by reverse-phase preparative HPLC ((Shimadzu, Phenomenex Gemini column (5 micron, C18, 110 Å, Axia, 100×50 mm)) eluting at 90 mL/min with an linear gradient of 10-70% MeCN (0.1% TFA) in water (0.1% TFA) over 20 minutes gave the title compound as an off-white fluffy powder after lyophilization. 1H NMR (300 MHz, CDCl3) δ ppm 12.32 (br. s., 1H), 8.76 (d, J=4.4 Hz, 1H), 8.09 (t, J=7.0 Hz, 1H), 7.76-7.53 (m, 4H), 7.45 (d, J=8.0 Hz, 2H), 7.33 (br. s., 5H), 6.25 (d, J=6.9 Hz, 1H), 5.22-5.02 (m, 2H). MS (ESI pos. ion) m/z: 387.0 (M+H).
  • Figure US20140235642A1-20140821-C00495
  • Example 173 (S)-tert-Butyl ((3-bromopyridin-2-yl)(4-(trifluoromethyl)phenyl)methyl)carbamate
  • The title compound was synthesized as described for Intermediate 40, Step 5. 1H-NMR (300 MHz, DMSO-d6): δ ppm 8.60 (d, J=3.7 Hz, 1H), 8.10 (dd, J=8.0, 1.5 Hz, 1H) 7.63-7.79 (m, 3H), 7.54 (d, J=7.5 Hz, 2H), 7.32 (dd, J=8.0, 4.5 Hz, 1H), 6.31 (d, J=8.5 Hz, 1H), 1.37 (s, 9H). MS (ESI pos. ion) m/z: 431.2, 433.2 (M+H).
  • Figure US20140235642A1-20140821-C00496
  • Example 174 (S)-tert-Butyl ((3-fluoro-4-(trifluoromethoxy)phenyl)(3-fluoropyridin-2-yl)methyl)carbamate
  • Di-tert-butyl dicarbonate (0.066 mL, 0.29 mmol) was added to a stirred solution of (S)-(3-fluoro-4-(trifluoromethoxy)phenyl)(3-fluoropyridin-2-yl)methanamine hydrochloride (Intermediate 39) (0.1 g, 0.29 mmol) in a 25 mL single neck rb flask in THF (5.0 mL) and saturated aqueous NaHCO3 (2.0 mL) solution at ambient temperature. The reaction mixture was stirred at rt for 3 h. After completion of the reaction (monitored by TLC, 50% EtOAc in hexane), water (20 mL) was added to the reaction mixture, the product was extracted with Et2O (20 mL×2), and the combined organic layers were washed with water. The organic layer was dried over anhydrous sodium sulfate and concentrated to give a residue which was purified by prep TLC using 10% EtOAc in hexane as eluent to give the title compound (0.04 g) as a sticky liquid. 1H-NMR (400 MHz, DMSO-d6): δ ppm 8.41 (d, J=4.4 Hz, 1H), 7.93 (d, J=8.4 Hz, 1H), 7.75-7.71 (m, 1H), 7.55-7.51 (m, 2H), 7.46-7.42 (m, 1H), 7.30 (d, J=8.0 Hz, 1H), 6.19 (d, J=8.8 Hz, 1H), 1.37 (s, 9H). MS (ESI pos. ion) m/z: 405.1 (M+H).
  • Figure US20140235642A1-20140821-C00497
  • General Carbamate Formation Procedure for Examples (175-177)
  • To a stirred solution of an alcohol (1.0 eq) in DCM (10 volumes) at 0° C., was added DIPEA (1.5 eq) followed by triphosgene (1.0 eq). After stirring the resulting solution at the same temperature for 1 h, a solution of (S)-(3-fluoro-4-(trifluoromethoxy)phenyl)(3-fluoropyridin-2-yl)methanamine hydrochloride (1.0 eq) (Intermediate 39) in DCM (10 volumes) and DIPEA (1.5 eq) was added dropwise to the reaction. The reaction mixture was stirred at the same temperature for 0.5 h-2 h. After completion of the reaction (monitored by TLC, 50% EtOAc in hexane), water was added to the reaction mixture, and the mixture was extracted with Et2O. The combined organic layers were washed with water. The organic layer was dried, filtered and concentrated to give a residue. The residue was purified by column chromatography over silica gel (60-120 mesh) using 5-20% EtOAc in hexane as eluent to give examples 175-177 as solids.
  • TABLE 5
    Examples 175-177 prepared via carbamate formation analogous to Scheme 11.
    MS (ESI,
    positive
    Product Product ion)
    Ex # Alcohol Structure Name M + H
    175 propan-2-ol
    Figure US20140235642A1-20140821-C00498
    (S)-isopropyl ((3- fluoro-4-(trifluoro- methoxy)phenyl)(3- fluoropyridin-2- yl)methyl)carbamate 391.1
    176 tetrahydro- 2H-pyran- 4-ol
    Figure US20140235642A1-20140821-C00499
    (S)-tetrahydro-2H- pyran-4-yl ((3- fluoro-4-(trifluoro- methoxy)phenyl)(3- fluoropyridin-2- yl)methyl)carbamate 433.1
    177 oxetan-3-ol
    Figure US20140235642A1-20140821-C00500
    (S)-oxetan-3-yl ((3- fluoro-4-(trifluoro- methoxy)phenyl)(3- fluoropyridin-2- yl)methyl)carbamate 405.0
  • Figure US20140235642A1-20140821-C00501
  • Example 178 6-Oxopiperidin-3-yl ((S)-(3-fluoro-4-(trifluoromethoxy)phenyl)(3-fluoropyridin-2-yl)methyl)carbamate
  • Figure US20140235642A1-20140821-C00502
  • Step 1. 2-Iodoacetyl Chloride
  • Thionyl chloride (0.1 mL, 1.32 mmol) was added dropwise to 2-iodoacetic acid (0.2 g, 1.1 mmol) at 0° C. The cooling bath was removed, and the reaction mixture was stirred at rt overnight with the exclusion of moisture and light. After completion of the reaction (monitored by TLC, 5% EtOAc in hexane), the excess thionyl chloride was evaporated under reduced pressure to give the title compound as a pink oil (0.2 g crude). The intermediate thus obtained was taken on to the next step without further purification.
  • Figure US20140235642A1-20140821-C00503
  • Step 2. N-Allyl-2-iodoacetamide
  • To a stirred solution of allyl amine (0.07 mL, 0.98 mmol) in DCM (2.0 mL) was added triethylamine (0.17 mL, 1.2 mmol) at rt. The resulting mixture was treated dropwise with a solution of 2-iodoacetyl chloride (0.2 g, 0.98 mmol) in DCM (2 mL) at 0° C. under nitrogen atmosphere. The reaction mixture was then stirred at the same temperature for an additional 2 h. After completion of the reaction (monitored by TLC, 30% EtOAc in hexane), water (20 mL) was added to the reaction mixture, and the aqueous layer extracted with DCM (20 mL×2). The combined organic layers were dried, filtered and concentrated to give a residue. The residue was purified by column chromatography over silica gel (60 120 mesh) using 15-30% EtOAc in hexane as eluent to give the title compound (0.03 g, 14%) as a white solid.
  • Figure US20140235642A1-20140821-C00504
  • Step 3. 5-Hydroxypiperidin-2-one
  • Triethylborane (0.1 mL, 0.1 mmol, 1 M solution in hexane) was added to a DCM (5 mL) solution of N-allyl iodoacetamide (0.225 g, 1.0 mmol) and boron trifluoride dihydrate (0.25 g, 3.0 mmol) at rt. The resulting mixture was then stirred at this temperature for 2 h. The solution was then concentrated under reduced pressure, and the residue was refluxed gently with hydrochloric acid (20 mL, 1 N aqueous) for 3 h. The resulting mixture was then concentrated in vacuo, and the residue was dissolved in anhydrous MeOH (15 mL). Potassium carbonate (0.55 g, 4 mmol) was added, and the mixture was stirred at rt overnight. After evaporation of the solvent under reduced pressure, the resulting product was purified by flash chromatography on silica gel with chloroform-MeOH (4:1) as the eluent to afford the title compound as a white solid. 1H-NMR (400 MHz, CD3OD): δ ppm 3.94-3.98 (m, 1H), 3.30 (dd, J=12.8, 4.0 Hz, 1H), 3.07 (dd, J=4.8, 1.2 Hz, 1H), 2.35-2.44 (m, 1H), 2.16-2.23 (m, 1H), 1.79-1.87 (m, 2H).
  • Figure US20140235642A1-20140821-C00505
  • Step 4. 6-Oxopiperidin-3-yl ((S)-(3-fluoro-4-(trifluoromethoxy)phenyl)(3-fluoropyridin-2-yl)methyl)carbamate
  • To a solution of 5-hydroxypiperidin-2-one (0.2 g, 1.74 mmol) in DCM (10.0 mL) at 0° C., was added DIPEA (0.45 mL, 2.61 mmol) followed by triphosgene (0.52 g, 1.74 mmol). The reaction was then stirred for 3 h at rt and then added via cannula to a solution of (S)-(3-fluoro-4-(trifluoromethoxy)phenyl)(3-fluoropyridin-2-yl)methanamine hydrochloride (Intermediate 39) (0.59 g, 1.74 mmol) in DCM (10.0 mL) and DIPEA (0.45 mL, 2.61 mmol). The resulting reaction mixture was stirred at the same temperature for 2 h. After completion of the reaction (monitored by TLC, 50% EtOAc in hexane), saturated aqueous ammonium chloride was added to the reaction mixture, and the mixture was extracted with DCM (10 mL×2). The combined organic layers were washed with water, dried, filtered, and concentrated to give a residue. The residue was purified by prep HPLC [Agilent Zorbax XDB-C18 column (21×150 mm, 5 microns); gradient elution, 20 mL/min, 20-70% (1:1 CAN:MeOH):0.01% TFA in H2O] to give the title compound (0.04 g) as a white solid. 1H-NMR (400 MHz, DMSO-d6): δ ppm 8.50 (d, J=4.4 Hz, 1H), 7.77-7.82 (m, 1H), 7.55-7.57 (m, 1H), 7.48-7.51 (m, 1H), 7.38-7.43 (m, 2H), 7.24 (d, J=8.4 Hz, 1H), 6.64 (d, J=7.2 Hz, 1H), 6.32 (d, J=8.4 Hz, 1H) 4.52-4.53 (m, 1H), 3.24-3.28 (m, 2H), 2.44-2.47 (m, 2H), 2.10-2.20 (m, 1H), 1.65-1.75 (m, 1H). MS (ESI pos. ion) m/z: 445.9 (M+H).
  • Figure US20140235642A1-20140821-C00506
  • Example 179 (S)-1-(Quinolin-5-yl)-3-((4-(trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methyl)urea
  • A mixture of quinoline-5-carboxylic acid (0.054 g, 0.312 mmol), DIPEA (0.054 mL, 0.312 mmol), and DPPA (0.067 mL, 0.312 mmol) in 1,4-dioxane (2 mL) was stirred at rt for 1 h. (S)-(4-(Trifluoromethyl)phenyl)(3-(trifluoromethyl)pyridin-2-yl)methanamine hydrochloride (Intermediate 1) (0.100 g, 0.312 mmol) was added, and the resulting mixture was heated at 85° C. for 24 h. The reaction mixture was cooled, concentrated, and purified by ISCO (Silica gel, 0-75% EtOAc/hexanes) to give the title compound. 1H-NMR (300 MHz, MeOH-d4): δ ppm 8.93 (d, J=4.5 Hz, 1H), 8.82 (d, J=3.5 Hz, 1H), 8.46 (d, J=8.5 Hz, 1H), 8.18 (d, J=8.0 Hz, 1H), 7.85 (d, J=7.3 Hz, 1H), 7.75-7.83 (m, 1H), 7.71 (d, J=7.7 Hz, 1H), 7.54-7.68 (m, 5H), 7.51 (dd, J=8.6, 4.2 Hz, 1H), 6.67 (s, 1H). MS (ESI pos. ion) m/z: 491.0 (M+H).
  • Figure US20140235642A1-20140821-C00507
  • Example 180 (S)-4-(3-((3-Fluoro-4-(trifluoromethoxy)phenyl)(3-fluoropyridin-2-yl)methyl)ureido)benzoic acid
  • Figure US20140235642A1-20140821-C00508
  • Step 1. (S)-Ethyl 4-(3-((3-fluoro-4-(trifluoromethoxy)phenyl)(3-fluoropyridin-2-yl)methyl)ureido)benzoate
  • To a solution of (S)-(3-fluoro-4-(trifluoromethoxy)phenyl)(3-fluoropyridin-2-yl)methanamine hydrochloride (Intermediate 39) (112 mg, 0.329 mmol) and DCM (3 mL) was added ethyl 4-isocyanatobenzoate (65 mg, 0.340 mmol) and N-ethyl-N-isopropylpropan-2-amine (0.172 mL, 0.986 mmol). The solution was stirred at rt. After 20 minutes, the reaction product was adsorbed onto a plug of silica gel and chromatographed through a Redi-Sep® pre-packed silica gel column (4 g), eluting with 70% to 90% EtOAc in hexane, to provide the title compound as a white solid.
  • Figure US20140235642A1-20140821-C00509
  • Step 2. (S)-4-(3-((3-Fluoro-4-(trifluoromethoxy)phenyl)(3-fluoropyridin-2-yl)methyl)ureido)benzoic acid
  • To a solution of (S)-ethyl 4-(3-((3-fluoro-4-(trifluoromethoxy)phenyl)(3-fluoropyridin-2-yl)methyl)ureido)benzoate (109 mg, 0.220 mmol) in THF (3 mL) and MeOH (1 mL) was added 1 M aqueous LiOH (4 mL, 4.00 mmol). The resulting mixture was then stirred at rt. After stirring for 24 h, the reaction was acidified with 1 N HCl to ˜pH 7. The aqueous solution was extracted with DCM (3×10 mL). The combined DCM layers were washed with brine, dried over MgSO4, and concentrated in vacuo to give the title compound as an off-white solid. 1H-NMR (300 MHz, MeOH-d4): δ ppm 8.48 (d, J=4.7 Hz, 1H), 7.83-7.98 (m, 2H), 7.62 (ddd, J=9.8, 8.5, 1.3 Hz, 1H), 7.27-7.51 (m, 6H), 6.40 (d, J=1.5 Hz, 1H). MS (ESI pos. ion) m/z: 445.9 (M+H).
  • TABLE 6
    Examples 181-187 prepared via urea formation analogous to Scheme 8.
    MS (ESI,
    Amine positive
    Inter- Isocyanate Product Product ion)
    Ex. # mediate Structure Structure Name M + H
    181 1
    Figure US20140235642A1-20140821-C00510
    Figure US20140235642A1-20140821-C00511
    (S)-1-(2- chlorophenyl)- 3-((4- (trifluorometh- yl)phenyl)(3- (trifluorometh- yl)pyridin-2- yl)methyl)urea (2,2,2-trifluoro- acetate) 473.8
    182 1
    Figure US20140235642A1-20140821-C00512
    Figure US20140235642A1-20140821-C00513
    (S)-methyl 2- methyl-3-(3-((4- (trifluorometh- yl)phenyl)(3- (trifluorometh- yl)pyridin-2- yl)meth- yl)ureido)ben- zoate (2,2,2-trifluoro- acetate) 511.4
    183 1
    Figure US20140235642A1-20140821-C00514
    Figure US20140235642A1-20140821-C00515
    (S)-1-(2- methoxy-5- methylphenyl)- 3-((4- (trifluorometh- yl)phenyl)(3- (trifluorometh- yl)pyridin-2- yl)methyl)urea (2,2,2-trifluoro- acetate) 483.4
    184 1
    Figure US20140235642A1-20140821-C00516
    Figure US20140235642A1-20140821-C00517
    (S)-1-(3,4- dichlorobenzyl)- 3-((4- (trifluorometh- yl)phenyl)(3- (trifluorometh- yl)pyridin-2- yl)methyl)urea (2,2,2-trifluoro- acetate) 522.3
    185 1
    Figure US20140235642A1-20140821-C00518
    Figure US20140235642A1-20140821-C00519
    1-(1,2,3,4- tetrahydro- naphthalen-1-yl)- 3-((S)-(4- (trifluorometh- yl)phenyl)(3- (trifluorometh- yl)pyridin-2- yl)methyl)urea (2,2,2-trifluoro- acetate) 493.4
    186 1
    Figure US20140235642A1-20140821-C00520
    Figure US20140235642A1-20140821-C00521
    (S)-1-mesityl- 3-((4- (trifluorometh- yl)phenyl)(3- (trifluorometh- yl)pyridin-2- yl)methyl)urea (2,2,2-trifluoro- acetate) 481.4
    187 1
    Figure US20140235642A1-20140821-C00522
    Figure US20140235642A1-20140821-C00523
    (S)-ethyl 4-(3- ((4-(trifluorometh- yl)phenyl)(3- (trifluorometh- yl)pyridin-2- yl)meth- yl)ureido)ben- zoate (2,2,2-trifluoro- acetate) 511.4
    188 39
    Figure US20140235642A1-20140821-C00524
    Figure US20140235642A1-20140821-C00525
    (S)-1-((3- fluoro-4- (trifluorometh- oxy)phenyl)(3- fluoropyridin- 2-yl)methyl)- 3-(pyridin-3- yl)urea 425.0
  • TABLE 7
    1HNMR Data for Selected Examples 1-188
    Freq.,
    Ex. # Solvent 1HNMR Data (δ ppm)
    1 300 MHz 9.15 (br. s, 1H), 8.93 (d, J = 4.1 Hz, 1H), 8.37 (br. s,
    d4-MeOH 1H), 8.15-8.29 (m, 2H), 7.85 (dd, J = 8.3, 5.6 Hz, 1H),
    7.54-7.70 (m, 2H), 7.26-7.47 (m, 2H), 6.59 (s, 1H)
    2 400 8.65 (br. s., 1H), 8.14-8.32 (m, 3H), 8.08 (d, J = 7.04
    MHz, Hz, 1H), 7.90 (d, J = 7.04 Hz, 1H), 7.06-7.36 (m, 5H),
    CDCl3 6.72 (d, J = 7.43 Hz, 2H), 6.53 (d, J = 7.24 Hz, 1H),
    3.68 (s, 3H)
    3 400 8.64 (d, J = 4.3 Hz, 1H), 8.40 (s, 1H), 8.27 (d, J = 2.4
    MHz, Hz, 1H), 8.2 (dd, J = 4.7, 1.0 Hz, 1H), 8.07-8.14 (m,
    CDCl3 1H), 7.93 (dd, J = 8.2, 1.2 Hz, 1H), 7.28-7.35 (m, 2H),
    7.20-7.25 (m, 1H), 7.14-7.20 (m, 2H), 7.07-7.14 (m,
    2H), 6.55 (d, J = 8.2 Hz, 1H)
    4 400 8.62 (d, J = 4.3 Hz, 1H), 8.46 (br. s., 1H), 8.27 (br. s,
    MHz, 1H), 8.18 (br. s, 1H), 8.09 (d, J = 9.0 Hz, 1H), 7.89 (d,
    CDCl3 J = 7.4 Hz, 1H), 7.26 (dd, J = 7.6, 5.1 Hz, 1H), 7.05-
    7.21 (m, 3H), 6.87-6.95 (m, 2H), 6.68 (dd, J = 7.3,
    1.9 Hz, 1H), 6.57 (d, J = 8.2 Hz, 1H), 3.66 (s, 3H)
    5 400 10.89 (br. s., 1H), 9.19 (br. s., 1H), 8.78 (d, J = 2.9 Hz,
    MHz, 1H), 8.69 (d, J = 6.7 Hz, 1H), 8.13 (br. s., 1H), 7.90
    CDCl3 (dd, J = 15.6, 7.9 Hz, 2H), 7.63 (br. s., 1H), 7.23-7.37
    (m, 1H), 7.04-7.20 (m, 2H), 6.78 (t, J = 8.4 Hz, 1H),
    6.46 (d, J = 7.8 Hz, 1H), 3.76 (s, 3H)
    6 400 8.99 (s, 1H), 8.94 (d, J = 1.8 Hz, 1H), 8.35 (d, J = 4.5
    MHz, Hz, 1H), 8.27 (d, J = 2.4 Hz, 1H), 8.06 (d, J = 4.7 Hz,
    CDCl3 1H), 7.96-8.01 (m, 2H), 7.86-7.97 (m, 1H), 7.72-7.82
    (m, 1H), 7.43-7.64 (m, 3H), 7.33-7.43 (m, 1H), 7.12
    (dd, J = 7.8, 4.7 Hz, 1H), 7.04 (dd, J = 8.4, 4.7 Hz,
    1H), 6.71 (d, J = 8.0 Hz, 1H)
    7 400 8.70 (d, J = 4.5 Hz, 1H), 8.14-8.28 (m, 2H), 8.08
    MHz, (d, J = 8.2 Hz, 1H), 7.93 (d, J = 7.8 Hz, 1H), 7.86
    CDCl3 (s, 1H), 7.32 (dd, J = 7.8, 4.9 Hz, 1H), 7.06-7.20
    (m, 4H), 6.92-7.04 (m, 2H), 6.56 (d, J = 8.2 Hz, 1H),
    2.22 (s, 3H)
    8 400 8.65 (d, J = 4.5 Hz, 1H), 8.33 (s, 1H), 8.28 (d, J = 2.0
    MHz, Hz, 1H), 8.19 (d, J = 4.7 Hz, 1H), 8.10 (d, J = 8.4 Hz,
    CDCl3 1H), 7.93 (d, J = 7.8 Hz, 1H), 7.31 (dd, J = 7.8, 4.9
    Hz, 1H), 7.07-7.21 (m, 4H), 6.97-7.07 (m, 1H), 6.78-
    6.88 (m, 1H), 6.57 (d, J = 8.0 Hz, 1H)
    9 400 8.70 (d, J = 4.1 Hz, 1H), 8.28 (br. s., 1H), 8.18 (d, J =
    MHz, 3.7 Hz, 1H), 8.01-8.11 (m, 2H), 7.95 (d, J = 7.8 Hz,
    CDCl3 1H), 7.52-7.66 (m, 2H), 7.40-7.48 (m, 1H), 7.30-7.38
    (m, 2H), 7.08-7.21 (m, 2H), 6.62 (d, J = 8.0 Hz, 1H)
    10 400 9.09 (d, J = 1.8 Hz, 1H), 8.64 (d, J = 4.5 Hz, 1H), 8.46
    MHz, (br. s., 2H), 8.35 (d, J = 2.2 Hz, 1H), 8.17 (d, J = 4.7
    CDCl3 Hz, 1H), 8.11 (d, J = 8.4 Hz, 1H), 7.88-7.99 (m, 2H),
    7.48-7.57 (m, 2H), 7.39 (d, J = 8.0 Hz, 1H), 7.31 (dd,
    J = 7.8, 4.9 Hz, 1H), 7.17 (dd, J = 8.4, 4.7 Hz, 1H),
    6.83 (d, J = 8.0 Hz, 1H)
    11 400 MHz 8.92 (d, J = 4.3 Hz, 1H), 8.53 (d, J = 2.2 Hz, 1H), 8.09-
    d4-MeOH 8.16 (m, 2H), 7.93-8.00 (m, 1H), 7.72-7.84 (m, 4H),
    7.57 (dd, J = 8.6, 1.0 Hz, 1H), 7.53 (dd, J = 7.8, 4.9 Hz,
    1H), 7.41-7.48 (m, 2H), 7.29 (dd, J = 8.3, 4.8 Hz, 1H),
    6.74 (s, 1H)
    12 400 9.11 (d, J = 2.0 Hz, 1H), 8.94 (s, 1H), 8.43 (d, J = 4.1
    MHz, Hz, 1H), 8.36 (d, J = 2.4 Hz, 1H), 8.15 (dd, J = 4.7,
    CDCl3 1.4 Hz, 1H), 8.03-8.11 (m, 2H), 7.85 (dd, J = 7.8,
    1.0 Hz, 1H), 7.76 (dd, J = 7.5, 1.1 Hz, 1H), 7.61 (dd,
    J = 8.2, 1.0 Hz, 1H), 7.55 (d, J = 8.0 Hz, 1H), 7.40
    (t, J = 7.9 Hz, 1H), 7.23 (dd, J = 7.9, 4.8 Hz, 1H),
    7.13 (dd, J = 8.4, 4.7 Hz, 1H), 6.77 (d, J = 8.0 Hz, 1H)
    13 400 MHz 8.94 (d, J = 4.3 Hz, 1H), 8.54 (d, J = 2.5 Hz, 1H), 8.19
    d4-MeOH (dd, J = 8.2, 0.8 Hz, 1H), 8.15 (dd, J = 4.7, 1.0 Hz, 1H),
    7.91-7.99 (m, 1H), 7.59 (dd, J = 7.9, 4.8 Hz, 1H), 7.55
    (d, J = 2.0 Hz, 1H), 7.47 (d, J = 8.4 Hz, 1H), 7.27-7.37
    (m, 2H), 6.53 (s, 1H)
    14 400 9.07 (br. s, 1H), 8.96 (s, 1H), 8.36-8.46 (m, 2H), 8.16
    MHz, (d, J = 4.5 Hz, 1H), 8.02-8.11 (m, 2H), 7.86 (d, J = 8.0
    CDCl3 Hz, 1H), 7.55 (d, J = 8.0 Hz, 1H), 7.46-7.51 (m, 1H),
    7.41 (td, J = 7.8, 4.9 Hz, 1H), 7.32 (dd, J = 10.4, 7.8
    Hz, 1H), 7.23 (dd, J = 7.8, 4.9 Hz, 1H), 7.14 (dd, J =
    8.3, 4.8 Hz, 1H), 6.76 (d, J = 7.8 Hz, 1H)
    15 400 8.56 (s, 1H) 8.38 (d, J = 2.4 Hz, 1H), 8.19-8.29 (m,
    MHz, 2H), 8.03-8.13 (m, 1H), 7.46 (d, J = 7.2 Hz, 1H),
    CDCl3 7.29-7.42 (m, 3H), 7.13-7.21 (m, 2H), 7.05 (s, 1H),
    7.03 (s, 1H), 6.44 (dd, J = 7.4, 1.2 Hz, 1H)
    16 400 9.33 (s, 1H), 9.06 (d, J = 2.0 Hz, 1H), 8.37 (d, J = 2.4
    MHz, Hz, 1H), 8.23 (d, J = 4.5 Hz, 1H), 8.13 (dd, J = 4.7,
    CDCl3 1.2 Hz, 1H), 8.03-8.10 (m, 1H), 7.91 (d, J = 1.4 Hz,
    1H), 7.82 (d, J = 7.4 Hz, 1H), 7.61 (d, J = 8.2 Hz, 1H),
    7.37-7.44 (m, 1H), 7.23-7.29 (m, 1H), 7.12 (td, J =
    8.6, 4.8 Hz, 2H), 6.97 (d, J = 7.6 Hz, 1H), 6.80 (d, J =
    8.2 Hz, 1H), 3.87 (s, 3H)
    17 400 8.83 (dd, J = 4.1, 1.4 Hz, 1H), 8.60 (s, 1H), 8.51 (d,
    MHz, J = 4.7 Hz, 1H), 8.36 (d, J = 2.4 Hz, 1H), 8.15-8.22
    CDCl3 (m, 1H), 8.05-8.11 (m, 1H), 8.02 (d, J = 8.2 Hz, 1H),
    7.91 (d, J = 8.8 Hz, 1H), 7.87 (d, J = 8.0 Hz, 1H), 7.73
    (s, 1H), 7.68 (dd, J = 8.9, 1.5 Hz, 1H), 7.28-7.37 (m,
    2H), 7.23 (dd, J = 7.8, 4.9 Hz, 1H), 7.14 (dd, J = 8.4,
    4.7 Hz, 1H), 6.76 (d, J = 8.0 Hz, 1H)
    18 400 8.94 (s, 1H), 8.77 (s, 1H), 8.50 (d, J = 4.3 Hz, 1H),
    MHz, 8.34 (br. s, 1H), 8.16 (d, J = 4.5 Hz, 1H), 8.09 (d, J =
    CDCl3 8.0 Hz, 1H), 7.96 (s, 1H), 7.87 (d, J = 7.8 Hz, 1H),
    7.54 (d, J = 9.0 Hz, 1H), 7.43 (d, J = 8.0 Hz, 1H), 7.19-
    7.30 (m, 2H), 7.08-7.17 (m, 2H), 6.75 (d, J = 7.8 Hz,
    1H), 3.81 (s, 3H)
    19 400 8.71 (s, 1H), 8.61 (d, J = 4.1 Hz, 1H), 8.32 (d, J = 2.4
    MHz, Hz, 1H), 8.18 (dd, J = 4.7, 1.0 Hz, 1H), 8.03-8.10 (m,
    CDCl3 1H), 7.90 (dd, J = 7.8, 0.8 Hz, 1H), 7.22-7.36 (m, 4H),
    7.14 (dd, J = 8.3, 4.8 Hz, 1H), 6.8-6.9 (m, 2H), 6.54
    (d, J = 8.0 Hz, 1H)
    20 400 8.74 (s, 1H), 8.59 (d, J = 4.1 Hz, 1H), 8.27 (br. s., 1H),
    MHz, 8.17 (d, J = 3.72 Hz, 1H), 8.03-8.10 (m, 1H), 7.87 (dd,
    CDCl3 J = 8.0, 1.0 Hz, 1H), 7.20-7.35 (m, 4H), 7.04-7.19 (m,
    4H), 6.59 (d, J = 8.22 Hz, 1H)
    21 300 MHz 9.14 (s, 1H), 8.96 (d, J = 4.7 Hz, 1H), 8.36 (d, J = 5.4
    d4-MeOH Hz, 1H), 8.21 (d, J = 7.9 Hz, 2H), 7.80-7.87 (m, 1H),
    7.68-7.78 (m, 2H), 7.59-7.64 (m, 1H), 7.31 (t, J = 9.4
    Hz, 1H), 6.58 (s, 1H)
    22 300 MHz 9.21 (s, 1H), 8.52 (d, J = 4.5 Hz, 1H), 8.39 (d, J = 5.4
    d4-MeOH Hz, 1H), 8.26 (d, J = 7.6 Hz, 1H), 7.90 (dd, J = 5.6,
    8.3 Hz, 1H), 7.68-7.80 (m, 2H), 7.64 (t, J = 9.0 Hz,
    1H), 7.44-7.50 (m, 1H), 7.32 (t, J = 9.4 Hz, 1H), 6.43
    (s, 1H)
    23 300 MHz 9.15 (br. s., 1H), 8.96 (d, J = 4.7 Hz, 1H), 8.37 (d, J =
    d4-MeOH 5.4 Hz, 1H), 8.22 (d, J = 7.5 Hz, 2H), 7.84 (dd, J =
    5.8, 8.4 Hz, 1H), 7.62 (dd, J = 4.8, 6.9 Hz, 1H), 7.25
    (s, 1H), 7.05-7.18 (m, 2H), 6.52 (s, 1H)
    24 300 MHz 9.08-9.24 (m, 1H), 8.51 (d, J = 4.2 Hz, 1H), 8.38 (d, J =
    d4-MeOH 5.3 Hz, 1H), 8.25 (d, J = 7.9 Hz, 1H), 7.87 (dd, J = 8.6,
    5.6 Hz, 1H), 7.57-7.76 (m, 1H), 7.38-7.56 (m, 1H),
    7.28 (s, 1H), 7.13 (d, J = 8.6 Hz, 2H), 6.37 (s, 1H)
    25 300 MHz 9.11 (br. s., 1H), 8.98 (d, J = 1.2 Hz, 1H), 8.79 (br. s.,
    d4-MeOH 1H), 8.35 (dd, J = 4.0, 1.1 Hz, 1H), 8.14-8.30 (m, 2H),
    8.01-8.12 (m, 1H), 7.72-7.90 (m, 2H), 7.53-7.72 (m,
    1H), 6.66 (br. s., 1H)
    26 400 8.77 (s, 1H), 8.63 (d, J = 4.3 Hz, 1H), 8.41 (d, J = 2.0
    MHz, Hz, 1H), 8.18 (d, J = 4.1 Hz, 1H), 8.02-8.10 (m, 1H),
    CDCl3 7.90 (d, J = 7.8 Hz, 1H), 7.35 (d, J = 8.2 Hz, 1H),
    7.22-7.31 (m, 3H), 7.06-7.17 (m, 3H), 6.54 (d, J =
    8.0 Hz, 1H)
    27 400 9.00 (s, 1H), 8.96 (d, J = 4.4 Hz, 1H), 8.47 (d, J = 2.0
    MHz, Hz, 1H), 8.22 (d, J = 7.6 Hz, 1H), 8.11 (d, J = 4.0 Hz,
    d6-DMSO 1H), 7.86 (d, J = 8.0 Hz, 1H), 7.58-7.64 (m, 1H),
    7.49 (d, J = 8.4 Hz, 1H) 7.14-7.26 (m, 5H), 6.40 (d, J =
    8.8 Hz, 1H), 2.27 (q, J = 7.6 Hz, 2H), 1.13 (t, J = 7.6
    Hz, 3H)
    28 400 MHz 8.67 (d, J = 4.3 Hz, 1H), 8.54 (s, 1H), 8.14 (d, J = 4.7
    d4-MeOH Hz, 1H), 8.06 (d, J = 8.0 Hz, 1H), 7.96 (d, J = 8.2 Hz,
    1H), 7.63 (s, 4H), 7.26-7.40 (m, 2H), 6.61 (s, 1H)
    29 300 8.48 (br. s., 1H), 8.35 (d, J = 4.1 Hz, 1H), 8.26 (d, J =
    MHz, 4.1 Hz, 1H), 8.10 (d, J = 8.2 Hz, 1H), 7.62 (br. s., 1H),
    CDCl3 7.50 (t, J = 7.6 Hz, 1H), 7.41 (t, J = 8.3 Hz, 1H), 7.22-
    7.34 (m, 5H), 6.42 (d, J = 6.3 Hz, 1H)
    30 300 8.51 (d, J = 3.7 Hz, 1H), 8.41 (d, J = 2.2 Hz, 1H), 8.27
    MHz, (d, J = 4.0 Hz, 1H) 7.96 (d, J = 8.2 Hz, 1H), 7.65 (t, J =
    CDCl3 8.5 Hz, 1H), 7.53 (d, J = 8.3 Hz, 2H), 7.47 (d, J = 8.3
    Hz, 2H), 7.17-7.24 (m, 3H), 7.11 (s, 1H), 6.11 (d, J =
    6.3 Hz, 1H)
    31 400 8.65 (d, J = 3.3 Hz, 1H), 7.91 (d, J = 7.6 Hz, 1H), 7.35-
    MHz, 7.49 (m, 4H), 7.27-7.35 (m, 1H), 7.20 (br. s, 4H), 7.00
    CDCl3 (br. s, 1H), 6.73-6.86 (m, 2H), 6.60 (d, J = 8.0 Hz, 1H)
    32 400 8.77-8.84 (m, 1H), 7.75-8.03 (m, 1H), 7.49-7.55 (m,
    MHz, 2H), 7.43-7.48 (m, 2H), 7.36-7.42 (m, 1H), 6.49-6.66
    CDCl3 (m, 1H), 5.98-6.14 (m, 1H), 4.02-4.22 (m, 1H), 3.74-
    3.95 (m, 1H), 1.12 (d, J = 6.5 Hz, 6H)
    33 300 MHz 8.70-8.88 (m, 1H), 8.08 (d, J = 7.7 Hz, 1H), 8.14 (d, J =
    d4-MeOH 7.7 Hz, 1H), 7.86 (d, J = 6.7 Hz, 1H), 7.75 (d, J = 8.2
    Hz, 1H), 7.32-7.62 (m, 9H), 6.55 (s, 1H), 5.55-5.74
    (m, 1H), 1.55 (d, J = 6.7 Hz, 3H)
    34 300 MHz 8.86 (d, J = 4.2 Hz, 1H), 8.14 (d, J = 7.7 Hz, 1H), 7.43-
    d4-MeOH 7.66 (m, 5H), 7.02-7.31 (m, 5H), 6.55 (s, 1H), 3.12
    (t, J = 6.9 Hz, 2H), 2.60 (t, J = 7.7 Hz, 2H), 1.75 (quin,
    J = 7.3 Hz, 2H)
    35 300 MHz 9.53 (d, J = 8.0 Hz, 1H), 8.88 (d, J = 4.2 Hz, 1H), 8.16
    d4-MeOH (d, J = 7.6 Hz, 1H), 7.49-7.69 (m, 5H), 7.41 (t, J = 7.7
    Hz, 2H), 7.27 (t, J = 7.3 Hz, 1H), 7.18 (d, J = 7.6 Hz,
    2H), 6.65 (d, J = 4.8 Hz, 1H)
    36 300 MHz 8.85 (d, J = 4.2 Hz, 1H), 8.16 (d, J = 8.0 Hz, 1H), 7.39-
    d4-MeOH 7.67 (m, 5H), 6.58-6.76 (m, 3H), 6.55 (s, 1H), 5.89
    (s, 2H), 2.67 (t, J = 6.9 Hz, 2H)
    37 300 MHz 8.90 (d, J = 4.5 Hz, 1H), 8.17 (d, J = 7.5 Hz, 1H), 7.68
    d4-MeOH (s, 1H), 7.49-7.65 (m, 5H), 7.18-7.30 (m, 1H), 7.03-
    7.18 (m, 2H), 6.60 (s, 1H)
    38 300 MHz 8.86 (br. s., 1H), 8.15 (d, J = 7.5 Hz, 1H), 7.37-7.71
    d4-MeOH (m, 5H), 6.55 (s, 1H), 4.15 (q, J = 6.7 Hz, 2H), 3.87
    (br. s., 2H), 1.22 (t, J = 7.0 Hz, 3H)
    39 300 MHz 8.90 (d, J = 5.3 Hz, 1H), 8.17 (d, J = 7.5 Hz, 1H), 7.49-
    d4-MeOH 7.74 (m, 7H), 7.37 (d, J = 9.1 Hz, 1H), 7.10-7.24 (m,
    1H), 6.75 (s, 1H), 6.64 (s, 1H)
    40 300 MHz 8.66 (d, J = 3.8 Hz, 1H), 8.05 (d, J = 7.2 Hz, 1H), 7.62
    d4-MeOH (s, 4H), 7.58 (d, J = 2.3 Hz, 4H), 7.30 (dd, J = 8.2, 4.7
    Hz, 1H), 6.60 (s, 1H)
    41 300 MHz 8.58 (d, J = 4.5 Hz, 1H), 8.00 (d, J = 8.0 Hz, 1H), 7.42-
    d4-MeOH 7.67 (m, 4H), 7.23 (dd, J = 8.1, 4.6 Hz, 1H), 6.42-6.61
    (m, 1H), 1.28 (s, 9H)
    42 300 MHz 8.47 (dd, J = 4.8, 1.6 Hz, 1H), 7.73 (dd, J = 7.9, 1.6 Hz,
    d4-MeOH 1H), 7.56 (s, 4H), 7.23 (dd, J = 7.7, 4.8 Hz, 1H), 6.54
    (s, 1H), 2.14 (s, 3H), 1.28 (s, 9H)
    43 300 MHz 8.52-8.54 (m, 2H), 8.13 (d, J = 4.7 Hz, 1H), 7.94 (d,
    d4-MeOH J = 8.4 Hz, 1H), 7.65 (d, J = 7.6 Hz, 1H), 7.60 (d, J =
    8.2 Hz, 2H), 7.53 (d, J = 8.2 Hz, 2H), 7.28-7.36 (m,
    2H), 6.42 (s, 1H), 5.91 (ddt, J = 16.8, 10.3, 6.3 Hz, 1H),
    5.08 (d, J = 10.2 Hz, 1H), 4.95-5.05 (m, 1H), 3.39-3.63
    (m, 2H)
    44 400 MHz 8.93 (d, J = 4.1 Hz, 1H), 8.53 (s, 1H), 8.19 (d, J = 8.0
    d4-MeOH Hz, 1H), 8.14 (d, J = 4.5 Hz, 1H), 7.96 (d, J = 8.2 Hz,
    1H), 7.63 (d, J = 8.2 Hz, 2H), 7.58 (d, J = 8.0 Hz, 3H),
    7.27-7.37 (m, 1H), 6.63 (s, 1H)
    45 300 MHz 8.86 (d, J = 4.5 Hz, 1H), 8.15 (d, J = 8.0 Hz, 1H),
    d4-MeOH 7.56-7.63 (m, 2H), 7.47-7.56 (m, 3H), 6.53 (s, 1H),
    1.29 (s, 9H)
    46 400 MHz 8.93 (d, J = 4.1 Hz, 1H), 8.19 (d, J = 8.0 Hz, 1H), 7.43-
    d4-MeOH 7.68 (m, 9H), 6.62 (s, 1H)
    47 400 MHz 8.92 (d, J = 4.1 Hz, 1H), 8.18 (d, J = 8.0 Hz, 1H), 7.62
    d4-MeOH (d, J = 8.0 Hz, 2H), 7.57 (d, J = 8.0 Hz, 3H), 7.29-7.39
    (m, 2H), 6.99 (t, J = 8.3 Hz, 2H), 6.62 (s, 1H)
    48 400 MHz 8.91 (d, J = 4.3 Hz, 1H), 8.18 (d, J = 8.0 Hz, 1H), 7.93
    d4-MeOH (q, J = 8.0 Hz, 1H), 7.60-7.66 (m, 2H), 7.53-7.60 (m,
    3H), 6.92-7.02 (m, 1H), 6.88 (t, J = 8.5 Hz, 1H), 6.63
    (s, 1H)
    49 300 MHz 8.92 (d, J = 4.4 Hz, 1H), 8.19 (d, J = 8.0 Hz, 1H), 7.60-
    d4-MeOH 7.68 (m, 2H), 7.52-7.60 (m, 3H), 7.03 (d, J = 7.7 Hz,
    2H), 6.61 (s, 1H), 6.51 (t, J = 9.2 Hz, 1H)
    50 300 MHz 8.92 (d, J = 4.5 Hz, 1H), 8.18 (d, J = 7.9 Hz, 1H), 7.44-
    d4-MeOH 7.68 (m, 5H), 7.14-7.33 (m, 1H), 6.92-7.06 (m, 2H),
    6.62 (s, 1H)
    51 300 MHz 8.47-8.60 (m, 2H), 8.14 (d, J = 4.4 Hz, 1H), 7.96 (d, J =
    d4-MeOH 8.3 Hz, 1H), 7.79 (d, J = 7.9 Hz, 1H), 7.55-7.69 (m, 4H),
    7.21-7.38 (m, 2H), 6.61 (s, 1H), 2.17 (s, 3H)
    52 300 MHz 8.89 (d, J = 4.4 Hz, 1H), 8.16 (d, J = 7.9 Hz, 1H), 7.41-
    d4-MeOH 7.66 (m, 5H), 7.00-7.18 (m, 2H), 6.80 (d, J = 8.2 Hz,
    1H), 6.61 (s, 1H), 6.53 (d, J = 8.2 Hz, 1H), 3.74 (s, 3H)
    53 300 MHz 8.91 (d, J = 4.4 Hz, 1H), 8.17 (d, J = 7.9 Hz, 1H), 8.09
    d4-MeOH (s, 1H), 7.41-7.66 (m, 8H), 6.62 (s, 1H), 3.09 (s, 3H)
    54 300 MHz 8.86 (d, J = 4.5 Hz, 1H), 8.14 (d, J = 8.0 Hz, 1H),
    d4-MeOH 7.41-7.66 (m, 5H), 6.54 (s, 1H), 3.76 (d, J = 11.3 Hz,
    1H), 3.57-3.72 (m, 2H), 3.41-3.57 (m, 1H), 3.07-3.27
    (m, 1H), 1.88 (br. s., 1H), 1.66-1.81 (m, 1H), 1.39-1.66
    m, 2H)
    55 300 MHz 8.90 (d, J = 4.2 Hz, 1H), 8.17 (d, J = 8.0 Hz, 1H), 7.47-
    d4-MeOH 7.69 (m, 5H), 6.58 (s, 1H), 2.24 (s, 3H), 2.08 (s, 3H)
    56 300 MHz 8.86 (d, J = 4.7 Hz, 1H), 8.14 (d, J = 7.9 Hz, 1H), 7.33-
    d4-MeOH 7.64 (m, 5H), 6.54 (s, 1H), 4.23 (br. s., 1H), 3.66-3.95
    (m, 3H), 3.42-3.59 (m, 1H), 2.04-2.31 (m, 1H), 1.56-
    1.85 (m, 1H)
    57 300 MHz 8.92 (d, J = 4.4 Hz, 1H), 8.85 (s, 2H), 8.73 (s, 1H), 8.18
    d4-MeOH (d, J = 7.9 Hz, 1H), 7.59 (q, J = 8.1 Hz, 5H), 6.61 (s, 1H)
    58 400 MHz 8.94 (br. s., 1H), 8.69 (br. s., 1H), 8.22 (t, J = 9.8 Hz,
    d4-MeOH 2H), 8.12 (s, 1H), 7.92 (d, J = 9.0 Hz, 1H), 7.52-7.70
    (m, 6H), 7.37-7.52 (m, 1H), 6.68 (s, 1H)
    59 300 MHz 8.93 (d, J = 4.4 Hz, 1H), 8.64-8.77 (m, 1H), 8.45 (s, 1H),
    d4-MeOH 8.18 (d, J = 7.9 Hz, 1H), 7.92 (d, J = 8.3 Hz, 1H), 7.81
    (d, J = 7.9 Hz, 1H), 7.47-7.68 (m, 7H), 6.67 (s, 1H)
    60 300 MHz 8.95 (d, J = 4.5 Hz, 1H), 8.62 (d, J = 5.4 Hz, 1H),
    d4-MeOH 8.09-8.30 (m, 3H), 7.96 (d, J = 8.5 Hz, 1H), 7.74
    (t, J = 7.5 Hz, 1H), 7.50-7.69 (m, 6H), 6.70 (s, 1H)
    61 400 MHz 8.47-8.57 (m, 2H), 8.13 (d, J = 4.7 Hz, 1H), 7.95 (d, J =
    d4-MeOH 8.4 Hz, 1H), 7.46-7.67 (m, 5H), 7.19-7.37 (m, 2H),
    6.51 (s, 1H), 1.70 (t, J = 7.0 Hz, 1H), 1.51 (s, 3H), 0.90
    (d, J = 7.0 Hz, 2H), 0.81 (s, 3H)
    62 300 MHz 8.52 (br. s., 2H), 8.12 (d, J = 4.4 Hz, 1H), 7.93 (d, J =
    d4-MeOH 7.2 Hz, 1H), 7.45-7.69 (m, 5H), 7.16-7.38 (m, 2H),
    6.54 (s, 1H), 2.93 (d, J = 13.9 Hz, 1H), 2.54 (d, J = 14.2
    Hz, 1H), 1.00 (s, 9H)
    114 300 MHz 10.66 (br. s., 1H), 9.31 (d, J = 8.6 Hz, 1H), 8.78 (s, 1H),
    CDCl3 8.32 (d, J = 4.7 Hz, 1H), 8.01 (d, J = 4.8 Hz, 1H), 7.71-
    7.77 (m, 2H), 7.60 (d, J = 8.3 Hz, 2H), 7.46 (d, J = 8.3
    Hz, 2H), 7.29-7.33 (m, 1H), 6.50 (d, J = 7.5 Hz, 1H)
    115 300 MHz 9.19 (s, 1H), 8.40 (d, J = 5.4 Hz, 1H), 8.31 (d, J = 8.6
    d4-MeOH Hz, 1H), 7.90 (dd, J = 8.4, 5.8 Hz, 1H), 7.68 (d, J = 7.7
    Hz, 3H), 7.49 (d, J = 8.0 Hz, 2H), 7.34-7.45 (m, 2H),
    7.22-7.32 (m, 1H), 6.56 (s, 1H)
    116 300 MHz 7.62-7.64 (m, 3H), 7.35-7.47 (m, 3H), 7.28-7.35 (m,
    d4-MeOH 1H), 7.17-7.28 (m, 1H), 6.38 (s, 1H), 1.32 (s, 9H)
    117 300 MHz 9.15 (d, J = 2.3 Hz, 1H), 8.39 (d, J = 5.4 Hz, 1H), 8.30
    d4-MeOH (ddd, J = 8.7, 2.4, 1.2 Hz, 1H), 7.87 (dd, J = 8.7, 5.5 Hz,
    1H), 7.64-7.78 (m, 3H), 7.56-7.64 (m, 2H), 7.27-7.42
    (m, 2H), 6.31 (s, 1H)
    118 300 MHz 7.16 (d, J = 4.5 Hz, 1H), 6.89 (d, J = 9.2 Hz, 1H), 6.67
    d4-MeOH (dd, J = 8.0, 1.5 Hz, 1H), 6.22-6.39 (m, 5H), 6.05-6.22
    (m, 2H), 5.65-5.78 (m, 1H), 5.22 (s, 1H), 2.61 (s, 3H)
    119 300 MHz 7.67 (d, J = 2.0 Hz, 1H), 7.24-7.39 (m, 2H), 6.90 (d, J =
    d4-MeOH 5.1 Hz, 1H), 6.79 (dd, J = 8.8, 1.2 Hz, 1H), 6.39 (dd, J =
    8.6, 5.6 Hz, 1H), 6.31 (d, J = 5.3 Hz, 1H), 6.22 (d, J =
    8.2 Hz, 2H), 5.98 (d, J = 8.2 Hz, 2H), 5.13 (s, 1H)
    120 300 10.75 (s, 1H), 9.18 (d, J = 9.2 Hz, 1H), 8.86 (br. s., 1H),
    MHz, 8.78 (br. s., 1H), 8.62 (br. s., 1H), 8.04 (d, J = 4.8 Hz,
    CDCl3 1H), 7.72 (d, J = 9.9 Hz, 1H), 7.77 (d, J = 8.6 Hz, 1H),
    7.62 (m, J = 8.0 Hz, 2H), 7.55 (d, J = 7.9 Hz, 1H), 7.46
    (m, J = 7.9 Hz, 2H), 6.47 (d, J = 6.6 Hz, 1H)
    121 400 8.18 (d, J = 2.4 Hz, 1H), 7.96-8.07 (m, 2H), 7.87-7.92
    MHz, (m, 1H), 7.68 (d, J = 7.8 Hz, 1H), 7.33-7.54 (m, 3H),
    CDCl3 7.02-7.19 (m, 4H), 6.51 (d, J = 6.7 Hz, 1H), 6.31
    (d, J = 6.9 Hz, 1H)
    122 300 MHz 8.94 (d, J = 4.5 Hz, 1H), 8.64 (d, J = 1.9 Hz, 1H), 8.28
    d4-MeOH (d, J = 4.8 Hz, 1H), 8.18 (t, J = 9.0 Hz, 2H), 7.55-7.70
    (m, 5H), 7.32-7.50 (m, 2H)
    123 300 MHz 8.90 (d, J = 4.7 Hz, 1H), 8.16 (d, J = 7.9 Hz, 1H), 7.47-
    d4-MeOH 7.65 (m, 5H), 7.43 (s, 1H), 1.48 (s, 9H)
    149 400 10.16 (s, 1H), 8.75 (s, 1H), 8.50 (d, J = 4.5 Hz, 1H),
    MHz, 7.78 (t, J = 9.2 Hz, 1H), 7.67-7.74 (m, 2H), 7.53-7.58
    d6-DMSO (m, 2H), 7.53-7.44 (m, 2H), 7.30 (s, 1H), 7.04-7.14
    (m, 1H), 6.66 (d, J = 8.4 Hz, 1H), 6.38 (d, J = 8.2 Hz,
    1H), 3.40 (br. s., 2H)
    150 300 8.52 (dd, J = 5.1, 1.5 Hz, 1H), 7.86 (dd, J = 7.8, 1.5
    MHz, Hz, 1H), 7.56 (d, J = 8.3 Hz, 2H), 7.49 (d, J = 8.5 Hz,
    CDCl3 2H), 7.34 (dd, J = 7.9, 5.1 Hz, 1H), 7.23 (br. s., 1H),
    6.64 (br. s., 1H), 5.60 (br. s., 1H), 4.48 (qd, J = 16.5,
    7.2 Hz, 1H), 2.13 (s, 3H), 1.27 (d, J = 7.0 Hz, 3H)
    151 300 MHz 8.87 (d, J = 4.1 Hz, 1H), 8.05-8.30 (m, 1H), 7.60-7.66
    d4-MeOH (m, 1H), 7.52-7.60 (m, 1H), 7.26 (d, J = 9.8 Hz, 2H),
    6.53 (s, 1H), 4.42 (dt, J = 14.5, 7.3 Hz, 1H), 1.26 (d,
    J = 7.0 Hz, 3H)
    152 300 MHz 8.88 (d, J = 4.5 Hz, 1H), 8.17 (d, J = 7.9 Hz, 1H), 7.47-
    d4-MeOH 7.70 (m, 2H), 7.18-7.41 (m, 2H), 6.51 (s, 1H), 4.40
    (dt, J = 14.5, 7.3 Hz, 1H), 1.27 (d, J = 7.0 Hz, 3H)
    153 300 MHz 8.88 (dd, J = 4.8, 0.9 Hz, 1H), 8.17 (dd, J = 8.0, 1.0 Hz,
    d4-MeOH 1H), 7.58-7.65 (m, 2H), 7.55 (dd, J = 8.0, 4.8 Hz, 1H),
    7.45-7.52 (m, 2H), 6.57 (s, 1H), 4.42 (dt, J = 14.4, 7.3
    Hz, 1H), 1.27 (d, J = 6.9 Hz, 3H)
    154 300 MHz 7.63 (d, J = 7.7 Hz, 3H), 7.32-7.46 (m, 3H), 7.17-7.32
    d4-MeOH (m, 2H), 6.44 (s, 1H), 4.45 (dt, J = 14.7, 7.2 Hz, 1H),
    1.29 (d, J = 7.0 Hz, 3H)
    155 300 9.27 (br. s., 2H), 8.83-8.90 (m, 2H), 8.75 (s, 1H), 8.36
    MHz, (br. s., 2H), 8.04 (d, J = 7.9 Hz, 2H), 7.46-7.54 (m, 2H),
    CDCl3 7.36-7.39 (m, 1H), 7.22-7.30 (m, 1H), 6.56 (s, 1H)
    156 300 8.33 (d, J = 4.7 Hz, 1H), 7.52 (d, J = 8.5 Hz, 2H), 7.46
    MHz, (d, J = 8.3 Hz, 2H), 7.38 (t, J = 8.3 Hz, 2H), 7.23-7.29
    CDCl3 (m, 1H), 6.80 (d, J = 7.2 Hz, 1H), 6.39 (dd, J = 7.2, 1.8
    Hz, 1H), 4.84 (d, J = 9.5 Hz, 1H), 4.44-4.62 (m, 1H),
    1.25 (d, J = 6.9 Hz, 3H)
    157 300 MHz 8.63 (d, J = 4.4 Hz, 1H), 8.03 (d, J = 8.0 Hz, 1H), 7.51-
    d4-MeOH 7.65 (m, 4H), 7.28 (dd, J = 8.1, 4.6 Hz, 1H), 6.54 (s,
    1H), 4.41 (dt, J = 14.5, 7.3 Hz, 1H), 1.27 (d, J = 7.0
    Hz, 3H)
    158 400 MHz 8.37 (d, J = 4.7 Hz, 1H), 7.52 (d, J = 7.6 Hz, 1H), 7.47
    d4-MeOH (m, J = 8.2 Hz, 2H), 7.27-7.39 (m, 2H), 7.18 (dd, J =
    7.8, 4.7 Hz, 1H), 6.26 (s, 1H), 5.79 (ddt, J = 16.8, 10.3,
    6.3, 1H), 4.86-5.04 (m, 2H), 4.29 (dt, J = 14.5, 7.3 Hz,
    1H), 3.27-3.45 (m, 2H), 1.14 (d, J = 7.0 Hz, 3H)
    159 300 8.81 (d, J = 4.1 Hz, 1H), 8.52 (s, 2H), 8.01 (d, J = 8.0
    MHz, Hz, 1H), 7.53 (d, J = 8.3 Hz, 1H), 7.42-7.49 (m, 3H),
    CDCl3 6.67 (d, J = 8.0 Hz, 1H), 6.61 (d, J = 8.0 Hz, 1H), 6.23
    (s, 1H), 3.98 (s, 3H)
    160 300 8.55 (s, 2H), 8.36 (d, J = 4.5 Hz, 1H), 7.49-7.57 (m,
    MHz, 4H), 7.40 (t, J = 8.0 Hz, 1H), 7.27-7.31 (m, 1H), 7.05
    CDCl3 (d, J = 7.0 Hz, 1H), 6.64 (s, 1H), 6.44 (dd, J = 7.0, 1.9
    Hz, 1H), 3.99 (s, 3H)
    161 400 9.16 (s, 1H), 8.50 (d, J = 4.7 Hz, 1H), 7.75-7.86 (m,
    MHz, 2H), 7.72 (d, J = 8.2 Hz, 2H), 7.46-7.57 (m, 4H), 6.40
    d6-DMSO (d, J = 2.3 Hz, 1H), 6.34 (d, J = 7.4 Hz, 1H), 6.20 (dd,
    J = 7.4, 2.3 Hz, 1H), 3.30 (s, 3H)
    162 400 8.44-8.53 (m, 2H), 7.81 (d, J = 2.7 Hz, 1H), 7.74-7.78
    MHz, (m, 1H), 7.67-7.74 (m, J = 8.2 Hz, 2H), 7.59 (d, J = 8.0
    d6-DMSO Hz, 1H), 7.52-7.56 (m, J = 8.0 Hz, 2H), 7.48 (td, J =
    8.5, 4.4 Hz, 1H), 7.25 (dd, J = 9.7, 2.8 Hz, 1H), 6.29-
    6.38 (m, 2H), 3.36 (s, 3H)
    163 400 11.72 (br. s., 1H), 8.80 (s, 1H), 8.39-8.58 (m, 2H), 7.96
    MHz, (dd, J = 7.2, 1.6 Hz, 1H), 7.64-7.81 (m, 3H), 7.57 (d, J =
    d6-DMSO 8.0 Hz, 2H), 7.45 (dt, J = 8.5, 4.4 Hz, 1H), 6.86-6.99 (m,
    1H), 6.41 (d, J = 8.4 Hz, 1H), 6.12 (t, J = 6.9 Hz, 1H)
    164 400 10.96 (br. s., 1H), 9.15 (s, 1H), 8.50 (d, J = 4.5 Hz, 1H),
    MHz, 7.66-7.87 (m, 4H), 7.42-7.59 (m, 3H), 7.18 (d, J = 7.2
    d6-DMSO Hz, 1H), 6.34 (d, J = 2.2 Hz, 2H), 6.15 (dd, J = 7.2, 2.0
    Hz, 1H)
    165 400 11.17 (br. s., 1H), 8.42-8.59 (m, 2H), 7.77 (t, J = 9.2
    MHz, Hz, 1H), 7.71 (d, J = 8.2 Hz, 2H), 7.60-7.51 (m, 4H),
    d6-DMSO 7.48 (dt, J = 8.5, 4.4 Hz, 1H), 7.28 (dd, J = 9.6, 2.9 Hz,
    1H), 6.35 (d, J = 8.0 Hz, 1H), 6.29 (d, J = 9.6 Hz, 1H)
    166 400 10.39 (s, 1H), 9.09 (s, 1H), 8.64 (d, J = 4.7 Hz, 1H),
    MHz, 7.92 (t, J = 9.2 Hz, 1H), 7.85 (d, J = 8.2 Hz, 2H), 7.56-
    d6-DMSO 7.76 (m, 5H), 7.31 (d, J = 1.6 Hz, 1H), 7.15 (d, J = 8.0
    Hz, 1H), 6.84 (dd, J = 8.0, 2.0 Hz, 1H), 6.51 (d, J = 7.8
    Hz, 1H), 3.48 (s, 2H)
    175 400 8.41 (d, J = 4.0 Hz, 1H), 8.17 (d, J = 8.4 Hz, 1H), 7.71-
    MHz, 7.76 (m, 1H), 7.52-7.56 (m, 2H), 7.43-7.47 (m, 1H),
    d6-DMSO 7.31 (d, J = 8.4 Hz, 1H), 6.24 (d, J = 8.4 Hz, 1H), 4.74-
    4.77 (m, 1H), 1.16 (d, J = 5.6 Hz, 6H)
    176 400 8.41-8.43 (d, J = 0.8 Hz, 1H), 7.38-7.43 (m, 1H), 7.28-
    MHz, 7.32 (m, 1H), 7.22-7.25 (m, 3H), 6.87 (d, J = 7.2 Hz,
    CDCl3 1H), 6.19 (d, J = 6.4 Hz, 1H), 4.82-4.84 (m, 1H), 3.89-
    3.93 (m, 2H), 3.47-3.53 (m, 2H), 1.90-1.92 (m, 2H),
    1.70-1.64 (m, 2H)
    177 400 8.61 (d, J = 8.4 Hz, 1H), 8.43 (d, J = 4.4 Hz, 1H), 7.72-
    MHz, 7.74 (m, 1H), 7.52-7.56 (m, 2H), 7.45-7.48 (m, 1H),
    d6-DMSO 7.32 (d, J = 8.4 Hz, 1H), 6.23 (d, J = 8.8 Hz, 1H), 5.28-
    5.29 (m, 1H), 4.72-4.75 (m, 2H), 4.42-4.47 (m, 2H)
    188 300 MHz 8.55 (d, J = 2.3 Hz, 1H), 8.50 (d, J = 4.7 Hz, 1H), 8.15
    d4-MeOH (dd, J = 4.8, 1.4 Hz, 1H), 7.96 (ddd, J = 8.4, 2.6, 1.5 Hz,
    1H), 7.64 (ddd, J = 9.7, 8.5, 1.2 Hz, 1H), 7.27-7.51 (m,
    5H), 6.41 (d, J = 1.6 Hz, 1H)
  • Stereochemistry
  • Absolute stereochemistries, where noted, were determined by comparison of either (I) quantum-mechanically predicted optical rotation values (Stephens, P. J. et. al, J. Phys. Chem. A 2001, 105, 5356) or (II) VCD spectra (Stephens, P. J. et. al, Chirality 2008, 20, 643) to those measured experimentally (or, in the case of Example 28, both). A single-crystal X-ray structure of (S)-tert-butyl((3-bromopyridin-2-yl)(4-(trifluoromethyl)phenyl)methyl)carbamate (Intermediate 40 Step 5; Example 173) provided confirmation of the absolute stereochemistry.
  • Computed and observed optical rotation for a subset of examples in this invention are shown in Table 8. Optical rotations were measured in CHCl3 at room temperature using a Perkin-Elmer digital polarimeter at 589 nm (sodium D line) in a 1.0 dm cell.
  • TABLE 8
    Computed and Measured Optical Rotation for Absolute
    Stereochemistry Assignment
    Computed Measured Computed Measured
    Example Rotation Rotation Example Rotation Rotation
    28 (S) = + +105° 44 (S) = + +87°
    30 (S) = +  +62° 51 (S) = + +32°
  • Assays Luminescence Readout Assay for Measuring Intracellular Calcium.
  • A stable Chinese hamster ovary cell line expressing human TRPM8 was generated using tetracycline inducible T-REx™ expression system from Invitrogen, Inc. (Carlsbad, Calif.). In order to enable a luminescence readout based on intracellular increase in calcium (Le Poul et al., 2002), the cell line was also co-transfected with pcDNA3.1 plasmid containing jelly fish aequorin cDNA. Twenty four hours before the assay, cells were seeded in 96-well plates and TRPM8 expression was induced with 0.5 μg/mL tetracycline. On the day of the assay, culture media was removed and cells were incubated with assay buffer (Ham's F12 containing 30 mM HEPES) that contained 15 μM coelenterazine (P.J.K, Germany) for 2 h. Potential antagonists were added 2.5 min prior to the addition of agonist, 1 μM icilin, 100 μM L-menthol, or 1 min prior to the addition of cold buffer (<10° C.). The luminescence was measured by a CCD camera based FLASH-luminometer built by Amgen, Inc. A cooling device attached to FLASH luminometer was used for cold activation. Compound activity was calculated using either GraphPad Prism 4.01 (GraphPad Software Inc, San Diego, Calif.) or Genedata Screener.
  • TABLE 9
    hTRPMS IC50s for Examples 1-188
    IC50
    Example (μM)
    1 0.023
    2 0.230
    3 0.172
    4 1.230
    5 0.145
    6 0.259
    7 0.723
    8 0.593
    9 0.175
    10 0.172
    11 0.101
    12 0.088
    13 0.051
    14 0.533
    15 0.015
    16 2.260
    17 0.811
    18 0.308
    19 0.382
    20 1.310
    21 0.178
    22 0.201
    23 0.255
    24 0.674
    25 0.216
    26 0.248
    27 0.041
    28 0.067
    29 0.017
    30 0.097
    31 0.081
    32 0.066
    33 1.900
    34 0.127
    35 1.710
    36 0.390
    37 4.080
    38 1.500
    39 4.090
    40 2.510
    41 0.122
    42 0.107
    43 0.022
    44 0.023
    45 0.053
    46 3.560
    47 0.098
    48 2.280
    49 0.558
    50 0.209
    51 0.037
    52 0.061
    53 0.050
    54 0.397
    55 0.050
    56 0.676
    57 0.167
    58 0.562
    59 0.640
    60 0.182
    61 0.062
    62 1.930
    63 0.866
    64 5.650
    65 0.141
    66 0.905
    67 0.352
    68 0.881
    69 0.777
    70 0.781
    71 5.710
    72 0.163
    73 0.332
    74 0.112
    75 0.670
    76 0.294
    77 0.235
    78 1.230
    79 1.500
    80 0.399
    81 0.496
    82 0.559
    83 0.115
    84 0.767
    85 0.405
    86 0.031
    87 0.199
    88 0.108
    89 0.330
    90 0.646
    91 0.670
    92 0.713
    93 1.440
    94 1.710
    95 1.920
    96 0.884
    97 0.080
    98 2.220
    99 0.712
    100 0.142
    101 0.633
    102 0.262
    103 0.527
    104 0.604
    105 0.545
    106 0.494
    107 0.123
    108 0.829
    109 0.110
    110 0.190
    111 2.380
    112 0.287
    113 0.175
    114 0.886
    115 0.154
    116 0.133
    117 1.750
    118 0.162
    119 0.375
    120 2.850
    121 0.084
    122 0.221
    123 0.190
    124 0.708
    125 0.561
    126 3.140
    127 0.657
    128 4.060
    129 2.000
    130 3.790
    131 0.666
    132 0.673
    133 1.590
    134 1.070
    135 0.829
    136 4.050
    137 0.156
    138 3.720
    139 1.310
    140 1.300
    141 0.360
    142 2.180
    143 1.300
    144 0.541
    145 0.474
    146 3.770
    147 3.830
    148 1.610
    149 0.055
    150 0.048
    151 0.039
    152 0.070
    153 0.051
    154 0.900
    155 0.062
    156 0.119
    157 0.059
    158 0.028
    159 0.407
    160 1.650
    161 1.250
    162 0.491
    163 0.076
    164 2.880
    165 3.210
    166 0.086
    167 0.328
    168 0.067
    169 0.663
    170 0.147
    171 0.915
    172 0.360
    173 0.299
    174 0.474
    175 0.068
    176 0.211
    177 0.517
    178 2.060
    179 0.077
    180 3.240
    181 0.142
    182 0.425
    183 0.560
    184 0.583
    185 0.739
    186 1.110
    187 1.550
    188 0.007
  • Icilin Biochemical Challenge Models Inhibition of Icilin Induced Jumping in Mice
  • Example compounds at doses ranging from 0.01 to 10 mg/kg were administered to male C57BL/6 mice (18-25 g, Taconic, n=10/treatment) 1 hour before icilin to assess the ability to block the spontaneous jumps induced by icilin (i.p. suspended in 100% PEG400 at 20 mg/kg, 5 mL/kg). The total number of jumps was recorded during the 10 minutes post-icilin administration based on the number of photocell beam breaks from the vertical array of open field boxes (Kinder Scientific) while movement of the mice was restricted within a clear Plexiglas® cylinder 9.5 cm diameter×30 cm height. Resultant data is shown in Table 10.
  • TABLE 10
    Dose % Inhibition of
    Example (mg/kg) Mouse Jumping
    1 10 75
    29 10 96
    44 10 74
    51 10 76
    61 10 90
    151 3 30
  • Inhibition of Icilin Induced Shaking in Rats
  • Example compounds at doses ranging from 0.01 to 3 mg/kg (p.o, suspended in 5% Tween80/Oralplus or suspended in 2% HPMC-1% Tween-80 pH2.2 with MSA, 5 mL/kg) were administered to male Sprague Dawley rats (200-300 g, Harlan, n=6-8/treatment) 2 hours before icilin to assess the ability to block the spontaneous wet-dog shake phenomena induced by icilin (i.p., suspended in 100% PEG400 at 0.5 mg/kg, 1 mL/kg or p.o., suspended in 2% HPMC-1% Tween-80 at 3 mg/kg, 2.5 mL/kg). Spontaneous wet-dog shakes were counted manually by two blinded observers or using LABORAS automation (Metris) for 30 minutes post-icilin dosing.
  • Cold Pressor Test (CPT) as a Translatable PD Model for TRPM8
  • The cold pressor test (CPT) was developed as a method to measure blood pressure response following exposure to a cold stimulus and has been used over 70 years in the diagnosis of hypertension and other cardiac autonomic disorders (Hines and Brown 1936). In healthy human subjects, the CPT is typically performed by immersing a subject's hand into ice water (0-5° C.) which triggers, through a vascular sympathetic activation of afferent pain and temperature neurons, an increase in blood pressure. With some modifications, this test has also been utilized in rat to delineate the medullary and spinal pathways mediating the cardio-vascular responses to cold pressor test and to identify neurotransmitters in these pathways (Sapru N et al 2008) or to characterize analgesic compounds (Koltzenburg M et al 2006 and Player M R et al 2011).
  • TRPM8 antagonists can be evaluated in rat CPT to determine whether TRPM8 antagonists would attenuate the increase in blood pressure resulting from exposure to cold stimulation of the paws and ventral half of the body. Male Sprague-Dawley rats weighing 350-450 g can be instrumented with a unilateral carotid artery-cannula connected to a transducer for measuring blood pressure using a Digi-Med Blood Pressure Analyzer, Model 400 Animals can be orally administrated with Vehicle (2% HPMC 1% Tween 80 pH 2.2 with MSA) or test compounds at 120 minutes prior to cold challenge and anesthetized with sodium pentobarbital at 60 mg/kg ip at 100 minutes prior to cold. Blood pressure can be recorded for 5 minutes for pre-cold baseline and additional 5 minutes during immersion of the paws and ventral half of body in ice water. Percent inhibition attributed to treatment with test compound can then determined using the following formula: [1−(cold evoked change in MBP/cold evoked change in MBP post-vehicle)]×100. Plasma can be collected through artery catheter immediately after CPT for pk analysis and IC50/90 determination.
  • REFERENCES
    • Hines, E A and Brown G E. The cold pressor test for measuring the reactability of the blood pressure. Am. Heart J. 1936, 11:1-9
    • Nakamura T, Kawabe K, and Sapru H N. Cold pressor test in the rat: medullary and spinal pathways and neurotransmitters. Am J Physiol Heart Circ Physiol 2008, 295:H1780-H1787
    • Koltzenburg M, Pokorny R, Gasser U and Richarz U. Differential sensitivity of three experimental pain models in detecting the analgesic effects of transdermal fentanyl and buprenorphine. Pain 2006, 126:165-174
    • Parks D, Parsons W, Colburn R, Meegala S, Ballentine S, Illig C, Qin N, Liu Y, Hutchinson T, Lubin M, Stone D, Baker J, Schneider C, Ma J, Damiano B, Flores C, and Player M. Design and optimization of benzimidazole-containing transient receptor potentiate melastatin 8 (TRPM8) antagonists. J. Med. Chem. 2011, 54:233-247
    CCI Model
  • Surgery—A chronic constriction injury (CCI) can be produced as previously described (Bennett & Xie, 1988). Under gaseous anesthesia with a mixture of isoflurane (3% for induction and 2% for maintenance) in O2, the sciatic nerve can be exposed at the mid-thigh level proximal to the sciatic trifurcation. Four chromic gut ligatures (4-0) can be tied loosely around nerve, 1-2 mm apart such that the vascular supply will not be compromised.
  • Behavioral testing—A behavioral test can be performed to estimate cold-induced ongoing pain as previously described (Choi et al., 1994). The rat can be placed under a transparent plastic cover on an aluminum plate (IITC PE34, Woodland, Calif.) which can be kept at a cold temperature (5±0.5° C.). After 2 minutes of adaptation, the cumulative duration of time that the rat lifts its foot off the plate for the next 5 minutes can be measured. Foot lifts associated with locomotion or grooming are not counted. Seven to 9 days after the CCI surgery, baseline of the cold-induced ongoing pain can be measured. Any rat showing a cold-induced ongoing pain less than 100 sec out of 300 sec observation period can be eliminated from the study. Twenty four hours after the baseline measurement, test compound, positive control, morphine (2 mg/kg, Sigma, St. Louis) or a vehicle (saline or 2% HPMC/1% Tween 80) can be administered orally (test compound) or subcutaneously (morphine). Two hrs (test compound) or 30 mins (morphine) after the drug administration, the cold-induced ongoing pain can be measured again.
  • Chung Model
  • Surgery—Spinal nerve ligation surgery can be performed as previously described (Kim & Chung, 1992). Briefly, under gaseous anesthesia with a mixture of isoflurane (3% for induction and 2% for maintenance) in O2, the spinal nerve injury can be produced by ligating the left L5 and L6 spinal nerves taking special care to avoid any possible damage to the L4 spinal nerve or surrounding area. Additional treatments can be performed to increase the development of mechanical allodynia. First, L5 spinal nerve can be cut approximately 1 mm distal to the suture as described by Li et al. (2000). Second, immediately after ligation and cut, the L4 spinal nerve can be lightly manipulated by slightly stretching it with a fine hooked glass rod and gently sliding the hook back and forth 20 times along the nerve as described by Lee et al. (2003). The whole surgery procedure from anesthesia to the clipping of the incised skin can take at most 15 minutes.
  • Behavioral testing —Two weeks later, mechanical sensitivity can be measured by determining the median 50% foot withdrawal threshold for von Frey filaments using the up-down method (Chaplan et al., 1994). The rats can be placed under a plastic cover (9×9×20 cm) on a metal mesh floor. The area tested consists of the middle glabrous area between the footpads of the plantar surface of the hind paw. The plantar area can be touched with a series of 9 von Frey hairs with approximately exponentially incremental bending forces (von Frey values: 3.61, 3.8, 4.0, 4.2, 4.41, 4.6, 4.8, 5.0 and 5.2; equivalent to: 0.41, 0.63, 1.0, 1.58, 2.51, 4.07, 6.31, 10 and 15.8 g). The von Frey hair can be presented perpendicular to the plantar surface with sufficient force to cause slight bending, and held for approximately 3-4 seconds. Abrupt withdrawal of the foot (paw flinching, shaking or licking for more than 1 sec.) can be recorded as a response. Any rat showing a mechanical threshold of more than 3.16 g or less than 0.7 g after surgery can be eliminated from the study. After measuring basal threshold, test compound, positive control gabapentin (Sigma, St. Louis) or a vehicle (saline or 2% HPMC/1% Tween 80) can be administered orally (test compound) or intraperitoneally (gabapentin). The measurement of the tactile threshold can be reassessed at 1.5 and 2 hrs after drug administration.
    Data—Since the von Frey filament set is calibrated on a logarithmic scale by the vendor (Stoelting) and our selection of 9 filaments for the up-down method is also based on near equal logarithmic intervals (Dixon et al., 1980), data can be treated using logarithmic values in every aspect (statistical treatment as well as plotting). However, an equivalent gram value scale is labeled on the Y-axis of the figures for convenience. Data are expressed as mean±standard error of the mean (S.E.M.).
  • For the treatment of TRPM8-receptor-diseases, such as acute, inflammatory and neuropathic pain, dental pain, general headache, migraine, cluster headache, mixed-vascular and non-vascular syndromes, tension headache, general inflammation, arthritis, rheumatic diseases, osteoarthritis, inflammatory bowel disorders, inflammatory eye disorders, inflammatory or unstable bladder disorders, psoriasis, skin complaints with inflammatory components, chronic inflammatory conditions, inflammatory pain and associated hyperalgesia and allodynia, neuropathic pain and associated hyperalgesia and allodynia, diabetic neuropathy pain, causalgia, sympathetically maintained pain, deafferentation syndromes, asthma, epithelial tissue damage or dysfunction, herpes simplex, disturbances of visceral motility at respiratory, genitourinary, gastrointestinal or vascular regions, wounds, burns, allergic skin reactions, pruritus, vitiligo, general gastrointestinal disorders, gastric ulceration, duodenal ulcers, diarrhea, gastric lesions induced by necrotising agents, hair growth, vasomotor or allergic rhinitis, bronchial disorders or bladder disorders, the compounds of the present invention may be administered orally, parentally, by inhalation spray, rectally, or topically in dosage unit formulations containing conventional pharmaceutically acceptable carriers, adjuvants, and vehicles. The term parenteral as used herein includes, subcutaneous, intravenous, intramuscular, intrasternal, infusion techniques or intraperitoneally.
  • Treatment of diseases and disorders herein is intended to also include the prophylactic administration of a compound of the invention, a pharmaceutical salt thereof, or a pharmaceutical composition of either to a subject (i.e., an animal, preferably a mammal, most preferably a human) believed to be in need of preventative treatment, such as, for example, pain, inflammation and the like.
  • The dosage regimen for treating TRPM8-receptor-mediated diseases, cancer, and/or hyperglycemia with the compounds of this invention and/or compositions of this invention is based on a variety of factors, including the type of disease, the age, weight, sex, medical condition of the patient, the severity of the condition, the route of administration, and the particular compound employed. Thus, the dosage regimen may vary widely, but can be determined routinely using standard methods. Dosage levels of the order from about 0.01 mg to 30 mg per kilogram of body weight per day, preferably from about 0.1 mg to 10 mg/kg, more preferably from about 0.25 mg to 1 mg/kg are useful for all methods of use disclosed herein.
  • The pharmaceutically active compounds of this invention can be processed in accordance with conventional methods of pharmacy to produce medicinal agents for administration to patients, including humans and other mammals.
  • For oral administration, the pharmaceutical composition may be in the form of, for example, a capsule, a tablet, a suspension, or liquid. The pharmaceutical composition is preferably made in the form of a dosage unit containing a given amount of the active ingredient. For example, these may contain an amount of active ingredient from about 1 to 2000 mg, preferably from about 1 to 500 mg, more preferably from about 5 to 150 mg. A suitable daily dose for a human or other mammal may vary widely depending on the condition of the patient and other factors, but, once again, can be determined using routine methods.
  • The active ingredient may also be administered by injection as a composition with suitable carriers including saline, dextrose, or water. The daily parenteral dosage regimen will be from about 0.1 to about 30 mg/kg of total body weight, preferably from about 0.1 to about 10 mg/kg, and more preferably from about 0.25 mg to 1 mg/kg.
  • Injectable preparations, such as sterile injectable aqueous or oleaginous suspensions, may be formulated according to the known are using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed, including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables.
  • Suppositories for rectal administration of the drug can be prepared by mixing the drug with a suitable non-irritating excipient such as cocoa butter and polyethylene glycols that are solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum and release the drug.
  • A suitable topical dose of active ingredient of a compound of the invention is 0.1 mg to 150 mg administered one to four, preferably one or two times daily. For topical administration, the active ingredient may comprise from 0.001% to 10% w/w, e.g., from 1% to 2% by weight of the formulation, although it may comprise as much as 10% w/w, but preferably not more than 5% w/w, and more preferably from 0.1% to 1% of the formulation.
  • Formulations suitable for topical administration include liquid or semi-liquid preparations suitable for penetration through the skin (e.g., liniments, lotions, ointments, creams, or pastes) and drops suitable for administration to the eye, ear, or nose.
  • For administration, the compounds of this invention are ordinarily combined with one or more adjuvants appropriate for the indicated route of administration. The compounds may be admixed with lactose, sucrose, starch powder, cellulose esters of alkanoic acids, stearic acid, talc, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulfuric acids, acacia, gelatin, sodium alginate, polyvinyl-pyrrolidine, and/or polyvinyl alcohol, and tableted or encapsulated for conventional administration. Alternatively, the compounds of this invention may be dissolved in saline, water, polyethylene glycol, propylene glycol, ethanol, corn oil, peanut oil, cottonseed oil, sesame oil, tragacanth gum, and/or various buffers. Other adjuvants and modes of administration are well known in the pharmaceutical art. The carrier or diluent may include time delay material, such as glyceryl monostearate or glyceryl distearate alone or with a wax, or other materials well known in the art.
  • The pharmaceutical compositions may be made up in a solid form (including granules, powders or suppositories) or in a liquid form (e.g., solutions, suspensions, or emulsions). The pharmaceutical compositions may be subjected to conventional pharmaceutical operations such as sterilization and/or may contain conventional adjuvants, such as preservatives, stabilizers, wetting agents, emulsifiers, buffers etc.
  • Solid dosage forms for oral administration may include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound may be admixed with at least one inert diluent such as sucrose, lactose, or starch. Such dosage forms may also comprise, as in normal practice, additional substances other than inert diluents, e.g., lubricating agents such as magnesium stearate. In the case of capsules, tablets, and pills, the dosage forms may also comprise buffering agents. Tablets and pills can additionally be prepared with enteric coatings.
  • Liquid dosage forms for oral administration may include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art, such as water. Such compositions may also comprise adjuvants, such as wetting, sweetening, flavoring, and perfuming agents.
  • Compounds of the present invention can possess one or more asymmetric carbon atoms and are thus capable of existing in the form of optical isomers as well as in the form of racemic or non-racemic mixtures thereof. The optical isomers can be obtained by resolution of the racemic mixtures according to conventional processes, e.g., by formation of diastereoisomeric salts, by treatment with an optically active acid or base. Examples of appropriate acids are tartaric, diacetyltartaric, dibenzoyltartaric, ditoluoyltartaric, and camphorsulfonic acid and then separation of the mixture of diastereoisomers by crystallization followed by liberation of the optically active bases from these salts. A different process for separation of optical isomers involves the use of a chiral chromatography column optimally chosen to maximize the separation of the enantiomers. Still another available method involves synthesis of covalent diastereoisomeric molecules by reacting compounds of the invention with an optically pure acid in an activated form or an optically pure isocyanate. The synthesized diastereoisomers can be separated by conventional means such as chromatography, distillation, crystallization or sublimation, and then hydrolyzed to deliver the enantiomerically pure compound. The optically active compounds of the invention can likewise be obtained by using active starting materials. These isomers may be in the form of a free acid, a free base, an ester or a salt.
  • Likewise, the compounds of this invention may exist as isomers, that is compounds of the same molecular formula but in which the atoms, relative to one another, are arranged differently. In particular, the alkylene substituents of the compounds of this invention, are normally and preferably arranged and inserted into the molecules as indicated in the definitions for each of these groups, being read from left to right. However, in certain cases, one skilled in the art will appreciate that it is possible to prepare compounds of this invention in which these substituents are reversed in orientation relative to the other atoms in the molecule. That is, the substituent to be inserted may be the same as that noted above except that it is inserted into the molecule in the reverse orientation. One skilled in the art will appreciate that these isomeric forms of the compounds of this invention are to be construed as encompassed within the scope of the present invention.
  • The compounds of the present invention can be used in the form of salts derived from inorganic or organic acids. The salts include, but are not limited to,
  • the following: acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphorsulfonate, digluconate, cyclopentanepropionate, dodecylsulfate, ethanesulfonate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, fumarate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, maleate, methansulfonate, nicotinate, 2-naphthalenesulfonate, oxalate, palmoate, pectinate, persulfate, 2-phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, tosylate, mesylate, and undecanoate. Also, the basic nitrogen-containing groups can be quaternized with such agents as lower alkyl halides, such as methyl, ethyl, propyl, and butyl chloride, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides, and others. Water or oil-soluble or dispersible products are thereby obtained.
  • Examples of acids that may be employed to from pharmaceutically acceptable acid addition salts include such inorganic acids as hydrochloric acid, sulfuric acid and phosphoric acid and such organic acids as oxalic acid, maleic acid, succinic acid and citric acid. Other examples include salts with alkali metals or alkaline earth metals, such as sodium, potassium, calcium or magnesium or with organic bases.
  • Also encompassed in the scope of the present invention are pharmaceutically acceptable esters of a carboxylic acid or hydroxyl containing group, including a metabolically labile ester or a prodrug form of a compound of this invention. A metabolically labile ester is one which may produce, for example, an increase in blood levels and prolong the efficacy of the corresponding non-esterified form of the compound. A prodrug form is one which is not in an active form of the molecule as administered but which becomes therapeutically active after some in vivo activity or biotransformation, such as metabolism, for example, enzymatic or hydrolytic cleavage. For a general discussion of prodrugs involving esters see Svensson and Tunek Drug Metabolism Reviews 165 (1988) and Bundgaard Design of Prodrugs, Elsevier (1985). Examples of a masked carboxylate anion include a variety of esters, such as alkyl (for example, methyl, ethyl), cycloalkyl (for example, cyclohexyl), aralkyl (for example, benzyl, p-methoxybenzyl), and alkylcarbonyloxyalkyl (for example, pivaloyloxymethyl). Amines have been masked as arylcarbonyloxymethyl substituted derivatives which are cleaved by esterases in vivo releasing the free drug and formaldehyde (Bungaard J. Med. Chem. 2503 (1989)). Also, drugs containing an acidic NH group, such as imidazole, imide, indole and the like, have been masked with N-acyloxymethyl groups (Bundgaard Design of Prodrugs, Elsevier (1985)). Hydroxy groups have been masked as esters and ethers. EP 039,051 (Sloan and Little, Apr. 11, 1981) discloses Mannich-base hydroxamic acid prodrugs, their preparation and use. Esters of a compound of this invention, may include, for example, the methyl, ethyl, propyl, and butyl esters, as well as other suitable esters formed between an acidic moiety and a hydroxyl containing moiety. Metabolically labile esters, may include, for example, methoxymethyl, ethoxymethyl, iso-propoxymethyl, α-methoxyethyl, groups such as α-((C1-C4)alkyloxy)ethyl, for example, methoxyethyl, ethoxyethyl, propoxyethyl, iso-propoxyethyl, etc.; 2-oxo-1,3-dioxolen-4-ylmethyl groups, such as 5-methyl-2-oxo-1,3,dioxolen-4-ylmethyl, etc.; C1-C3 alkylthiomethyl groups, for example, methylthiomethyl, ethylthiomethyl, isopropylthiomethyl, etc.; acyloxymethyl groups, for example, pivaloyloxymethyl, α-acetoxymethyl, etc.; ethoxycarbonyl-1-methyl; or α-acyloxy-α-substituted methyl groups, for example α-acetoxyethyl.
  • Further, the compounds of the invention may exist as crystalline solids which can be crystallized from common solvents such as ethanol, N,N-dimethylformamide, water, or the like. Thus, crystalline forms of the compounds of the invention may exist as polymorphs, solvates and/or hydrates of the parent compounds or their pharmaceutically acceptable salts. All of such forms likewise are to be construed as falling within the scope of the invention.
  • While the compounds of the invention can be administered as the sole active pharmaceutical agent, they can also be used in combination with one or more compounds of the invention or other agents. When administered as a combination, the therapeutic agents can be formulated as separate compositions that are given at the same time or different times, or the therapeutic agents can be given as a single composition.
  • The foregoing is merely illustrative of the invention and is not intended to limit the invention to the disclosed compounds. Variations and changes which are obvious to one skilled in the art are intended to be within the scope and nature of the invention which are defined in the appended claims.
  • From the foregoing description, one skilled in the art can easily ascertain the essential characteristics of this invention, and without departing from the spirit and scope thereof, can make various changes and modifications of the invention to adapt it to various usages and conditions.

Claims (1)

What is claimed is:
1. A method of treating acute, inflammatory and neuropathic pain, dental pain, general headache, migraine, cluster headache, mixed-vascular and non-vascular syndromes, tension headache, general inflammation, arthritis, rheumatic diseases, osteoarthritis, inflammatory bowel disorders, depression, anxiety, inflammatory eye disorders, inflammatory or unstable bladder disorders, psoriasis, skin complaints with inflammatory components, chronic inflammatory conditions, inflammatory pain and associated hyperalgesia and allodynia, neuropathic pain and associated hyperalgesia and allodynia, diabetic neuropathy pain, causalgia, sympathetically maintained pain, deafferentation syndromes, asthma, epithelial tissue damage or dysfunction, herpes simplex, disturbances of visceral motility at respiratory, genitourinary, gastrointestinal or vascular regions, wounds, burns, allergic skin reactions, pruritus, vitiligo, general gastrointestinal disorders, gastric ulceration, duodenal ulcers, diarrhea, gastric lesions induced by necrotising agents, hair growth, vasomotor or allergic rhinitis, bronchial disorders or bladder disorders in a subject, the method comprising: administering to the subject a compound of Formula I or a pharmaceutically-acceptable salt thereof, a tautomer thereof, a pharmaceutically-acceptable salt of the tautomer, a stereoisomer thereof or a mixture thereof, wherein the compound of Formula I has the structure:
Figure US20140235642A1-20140821-C00526
wherein:
m is 0, 1, 2 or 3;
n is 0 or 1;
X1 is C(R4) or N;
X2 is CH or N;
Y is NH, NR1a, or O;
Z is O or S;
R1 is C1-6alk, —(C═O)—O—C1-6alk, or a direct-bonded, C1-6alk-linked, C1-2alkO-linked, —C(═O)—O linked, —C(═O)— linked, saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic or 7-, 8-, 9-, 10- or 11-membered bicyclic ring containing 0, 1, 2, 3 or 4 heteroatoms selected from N, O and S, the C1-6alk, the C1-6alk of the C1-6alk-link, and the monocyclic or bicyclic ring being substituted by 0, 1, 2 or 3 substituents independently selected from halo, oxo, C1-6alk, C1-6alkOH, C1-6alk-C(═O)Ra, C1-6alk-C(═O)ORa, C1-4haloalk, cyano, nitro, —C(═O)Ra, —C(═O)ORa, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Ra, —OC(═O)NRaRa, —OC(═O)N(Ra)S(═O)2Ra, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, ═S, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRaRa, —S(═O)2N(Ra)C(═O)Ra, —S(═O)2N(Ra)C(═O)ORa, —S(═O)2N(Ra)C(═O)NRaRa, —NRaRa, —N(Ra)C(═O)Ra, —N(Ra)C(═O)ORa, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Ra, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa, wherein the ring is additionally substituted by 0 or 1 directly bonded, SO2 linked, C(═O) linked or CH2 linked saturated, partially-saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered monocyclic ring containing 0, 1, 2, 3 or 4 heteroatoms selected from N, O and S, and substituted by 0, 1, 2 or 3 groups selected from halo, C1-6alk, C1-4haloalk, cyano, oxo, nitro, —C(═O)Ra, —C(═O)ORa, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Ra, —SRa, —S(═O)Ra, —S(═O)2Ra, —S(═O)2NRaRa, —NRaRa, and —N(Ra)C(═O)Ra; or R1a and R1, together with the N atom to which they are attached, form a saturated, partially-saturated or unsaturated 4-, 5-, 6-, or 7-membered monocyclic or a 9- or 10-membered bicylic ring containing 0, 1, or 2 additional heteroatoms independently selected from N, O, and S, wherein the ring formed by R1a and R1 is substituted with 0, 1, or 2 substituents independently selected from halo, oxo, C1-6alk, C1-6alkOH, C1-6alk-C(═O)Ra, C1-6alk-C(═O)ORa, C1-4haloalk, cyano, nitro, —C(═O)Ra, —C(═O)ORa, or —C(═O)NRaRa;
R2 is H, halo, cyano, Rc, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa; or R2 is C1-6alk substituted by 0, 1, 2 or 3 substituents selected from C1-4haloalk, halo, cyano, nitro, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa and —NRaC2-6alkORa, or R2 is C1-6alk substituted by 0, 1, 2 or 3 halo substituents and additionally substituted by 0 or 1 substituents selected from Rc;
R3 is H, C1-8alk, C1-8alkOH, C1-4haloalk, halo, cyano, —C(═O)Rb, —C(═O)ORb, —C(═O)NRaRa, —C(═NRa)NRaRa, —ORa, —OC(═O)Rb, —OC(═O)NRaRa, —OC2-6alkNRaRa, —OC2-6alkORa, —SRa, —S(═O)Rb, —S(═O)2Rb, —S(═O)2NRaRa, —NRaRa, —N(Ra)C(═O)Rb, —N(Ra)C(═O)ORb, —N(Ra)C(═O)NRaRa, —N(Ra)C(═NRa)NRaRa, —N(Ra)S(═O)2Rb, —N(Ra)S(═O)2NRaRa, —NRaC2-6alkNRaRa or —NRaC2-6alkORa;
R4 is independently, at each instance, H, C1-6alk, —C1-3haloalk, —OC1-6alk, —OC1-3haloalk, —N(C1-6alk)C1-6alk, —NHC1-6alk, —NC(═O)C1-6alk, —N(C1-6alk)C1-6alk, F, Cl, Br, CN, OH or NH2; or R3 and R4 together form a four-atom unsaturated bridge containing 0 or 1 N atoms, wherein the bridge is substituted by 0, 1 or 2 R5 substituents;
R5 is independently, in each instance, Cl, Br, F, CH3, CF3, or ORa;
R6 is F, CF3, C1-6alk, or ORa;
Ra is independently, at each instance, H or Rb;
Rb is independently, at each instance, phenyl, benzyl or C1-6alk, the phenyl, benzyl and C1-6alk being substituted by 0, 1, 2 or 3 substituents selected from halo, oxo, C1-4alk, C1-3haloalk, —OC1-4alk, —OH, —NH2, —OC1-4alk, —OC1-4haloalk, —NHC1-4alk, and —N(C1-4alk)C1-4alk; and
Rc is independently, at each instance, a saturated, partially saturated or unsaturated 4-, 5- or 6-membered monocyclic ring containing 0, 1, 2, 3 or 4 heteroatoms selected from N, O and S, wherein the C1-6alkyl and ring are substituted by 0 or 1 oxo groups substituted by 0, 1, 2 or 3 substituents selected from C1-8alk, C1-4haloalk, halo and cyano.
US14/263,143 2011-06-24 2014-04-28 Trpm8 antagonists and their use in treatments Abandoned US20140235642A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/263,143 US20140235642A1 (en) 2011-06-24 2014-04-28 Trpm8 antagonists and their use in treatments

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201161500835P 2011-06-24 2011-06-24
US13/529,880 US8778941B2 (en) 2011-06-24 2012-06-21 TRPM8 antagonists and their use in treatments
US14/263,143 US20140235642A1 (en) 2011-06-24 2014-04-28 Trpm8 antagonists and their use in treatments

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US13/529,880 Division US8778941B2 (en) 2011-06-24 2012-06-21 TRPM8 antagonists and their use in treatments

Publications (1)

Publication Number Publication Date
US20140235642A1 true US20140235642A1 (en) 2014-08-21

Family

ID=46384522

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/529,880 Active US8778941B2 (en) 2011-06-24 2012-06-21 TRPM8 antagonists and their use in treatments
US14/263,143 Abandoned US20140235642A1 (en) 2011-06-24 2014-04-28 Trpm8 antagonists and their use in treatments

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US13/529,880 Active US8778941B2 (en) 2011-06-24 2012-06-21 TRPM8 antagonists and their use in treatments

Country Status (7)

Country Link
US (2) US8778941B2 (en)
EP (1) EP2723718A1 (en)
JP (1) JP2014517074A (en)
AU (1) AU2012272898A1 (en)
CA (1) CA2839703A1 (en)
MX (1) MX2013015058A (en)
WO (1) WO2012177896A1 (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20140045507A (en) 2011-06-24 2014-04-16 암젠 인크 Trpm8 antagonists and their use in treatments
US8952009B2 (en) 2012-08-06 2015-02-10 Amgen Inc. Chroman derivatives as TRPM8 inhibitors
CN105263901B (en) * 2013-05-08 2017-06-23 橘生药品工业株式会社 α substituted glycyl amine derivatives
US10781310B2 (en) 2019-02-08 2020-09-22 Covestro Llc Polymer polyol stabilizers
CN116134018A (en) 2020-06-17 2023-05-16 默沙东有限责任公司 2-oxoimidazolidine-4-carboxamide as NAV1.8 inhibitor

Family Cites Families (177)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2483250A (en) 1949-09-27 Tt a it tt
DE2417763A1 (en) 1974-04-11 1975-10-30 Bayer Ag CARBON ACID AMIDE, THE PROCESS FOR THEIR MANUFACTURING AND THEIR USE AS A MEDICINAL PRODUCT
DE2428673A1 (en) 1974-06-14 1976-01-02 Bayer Ag CARBON ACID AMIDE, THE PROCESS FOR THEIR MANUFACTURING AND THEIR USE AS A MEDICINAL PRODUCT
PT72878B (en) 1980-04-24 1983-03-29 Merck & Co Inc Process for preparing mannich-base hydroxamic acid pro-drugs for the improved delivery of non-steroidal anti-inflammatory agents
DE3020458C2 (en) 1980-05-29 1986-07-31 Herzog, Thomas, Prof. Dr., 3500 Kassel Curtain wall, building or decorative panel
US5081131A (en) 1986-01-13 1992-01-14 American Cyanamid Company Omega-((hetero)alkyl)benz(cd)-indol-2-amines
US5223499A (en) 1989-05-30 1993-06-29 Merck & Co., Inc. 6-amino substituted imidazo[4,5-bipyridines as angiotensin II antagonists
AU640016B2 (en) 1990-05-30 1993-08-12 Imperial Chemical Industries Plc Hydroquinazoline derivatives
US5380721A (en) 1990-09-10 1995-01-10 Sterling Winthrop Inc. Aryl-fused and hetaryl-fused-2,4-diazepine and 2,4-diazocine antiarrhythmic agents
ES2036926B1 (en) 1991-08-08 1994-01-16 Uriach & Cia Sa J "PROCEDURE FOR THE OBTAINING OF DERIVATIVES OF (2-ALKYL-3-PIRIDIL) METHYLPIPERAZINE".
JP2600644B2 (en) 1991-08-16 1997-04-16 藤沢薬品工業株式会社 Thiazolyl benzofuran derivative
DE4135474A1 (en) 1991-10-28 1993-04-29 Bayer Ag 2-AMINOMETHYL-chromans
DE4138819A1 (en) 1991-11-26 1993-05-27 Basf Ag HYDROXYPYRIDONE CARBONIC ACID AMIDE, THEIR PREPARATION AND USE
GB9127041D0 (en) 1991-12-20 1992-02-19 Fujisawa Pharmaceutical Co New use
FR2692895A1 (en) 1992-06-26 1993-12-31 Adir New 1-oxo-tetra:hydro 2,3,7-tri:aza:phenalene derivs. - inhibitors of platelet aggregation for treating e.g. thrombosis or pulmonary embolism, also new tetra:hydro-quinolone intermediates
DE4304455A1 (en) 1993-02-15 1994-08-18 Bayer Ag Heterocyclic substituted phenyl-cyclohexane-carboxylic acid derivatives
CA2135536A1 (en) 1993-11-12 1995-05-13 Hironori Kurita Glucosamine derivative, process for preparing the same and synthetic intermediate thereof
SK79797A3 (en) 1994-12-23 1997-11-05 Thomae Gmbh Dr K Piperazine derivatives, medicaments containing the same, their use and process for preparing the same
JPH11501934A (en) 1995-03-14 1999-02-16 シャスカン,エドワード・ジー A composition comprising nicotinylalanine and an inhibitor of glycine conjugation or vitamin B6
US5891872A (en) 1995-04-07 1999-04-06 Schering Corporation Tricyclic compounds
MY114302A (en) 1995-04-19 2002-09-30 Ihara Chemical Ind Co Benzylsulfide derivative, process for its production and pesticide
DE19518681A1 (en) 1995-05-22 1996-11-28 Bayer Ag Process for the preparation of 5,6-dihydro-1,3-oxazines
DE19523087A1 (en) 1995-06-26 1997-01-02 Bayer Ag 1,3-Oxa (thia) tin derivatives
IL118631A (en) 1995-06-27 2002-05-23 Tanabe Seiyaku Co Pyridazinone derivatives and processes for their preparation
GB9518552D0 (en) 1995-09-11 1995-11-08 Fujisawa Pharmaceutical Co New heterocyclic compounds
EP0858457A1 (en) 1995-10-20 1998-08-19 Dr. Karl Thomae GmbH 5-membered heterocycles, pharmaceutical agents containing said compounds and the use thereof and methods of producing them
GB9525828D0 (en) 1995-12-18 1996-02-21 Bayer Ag Use of hetarylacetic acid derivatives
US5977090A (en) 1996-09-27 1999-11-02 Guilford Pharmaceuticals Inc. Pharmaceutical compositions and methods of treating compulsive disorders using NAALADase inhibitors
US6268384B1 (en) 1997-08-29 2001-07-31 Vertex Pharmaceuticals Incorporated Compounds possessing neuronal activity
US6075029A (en) 1998-01-02 2000-06-13 Cell Therapeutics, Inc. Xanthine modulators of metabolism of cellular P-450
CA2318184C (en) 1998-01-21 2008-11-18 Banyu Pharmaceutical Co., Ltd. Substituted 5-(2,2-difluoro-1,3-benzodioxol-5-yl) cyclopentenopyridine derivative as selective endothelin antagonists
EP1082095A1 (en) 1998-06-03 2001-03-14 GPI NIL Holdings, Inc. N-oxide of heterocyclic ester, amide, thioester or ketone hair growth compositions and uses
US5910595A (en) 1998-06-26 1999-06-08 Salsbury Chemicals, Inc. Process for preparing oximidazoles
US6617351B1 (en) 1998-07-31 2003-09-09 Eli Lilly And Company Amide, carbamate, and urea derivatives
US6166028A (en) 1998-12-09 2000-12-26 American Home Products Corporation Diaminopuridine-containing thiourea inhibitors of herpes viruses
AU1684400A (en) 1998-12-11 2000-07-03 Sankyo Company Limited Substituted benzylamines
WO2000039102A1 (en) 1998-12-23 2000-07-06 Du Pont Pharmaceuticals Company THROMBIN OR FACTOR Xa INHIBITORS
US6200993B1 (en) 1999-05-05 2001-03-13 Merck Frosst Canada & Co. Heterosubstituted pyridine derivatives as PDE4 inhibitors
CZ290178B6 (en) 1999-07-22 2002-06-12 Sankyo Company Limited Cyclobutene derivatives
GB9919588D0 (en) 1999-08-18 1999-10-20 Hoechst Schering Agrevo Gmbh Fungicidal compounds
BR0013593A (en) 1999-09-01 2002-05-07 Unilever Nv Method for bleaching fabric stains
CA2383596A1 (en) 1999-09-01 2001-03-08 Unilever Plc Composition and method for bleaching a substrate
BR0013592A (en) 1999-09-01 2002-05-07 Unilever Nv Commercial packaging for bleaching fabric stains in an aqueous washing liquor, and using it
US7291641B2 (en) 1999-10-11 2007-11-06 Societe De Conseils De Recherches Et D'applications Scientifiques (S.C.R.A.S.) Derivatives of heterocycles with 5 members, their preparation and their use as medicaments
CA2325358C (en) 1999-11-10 2005-08-02 Pfizer Products Inc. 7-¬(4'-trifluoromethyl-biphenyl-2-carbonyl)amino|-quinoline-3-carboxylic acid amides, and methods of inhibiting the secretion of apolipoprotein b
IL139450A0 (en) 1999-11-10 2001-11-25 Pfizer Prod Inc Methods of administering apo b-secretion/mtp inhibitors
KR100518142B1 (en) 1999-11-10 2005-10-04 다케다 야쿠힌 고교 가부시키가이샤 5-membered n-heterocyclic compounds with hypoglycemic and hypolipidemic activity
TW574193B (en) 1999-12-03 2004-02-01 Astrazeneca Ab Novel phenalkyloxy-phenyl derivatives, pharmaceutical composition containing the same and their uses
YU41202A (en) 1999-12-28 2005-03-15 Pfizer Products Inc. Non-peptidyl inhibitors of vla-4 dependent cell binding useful in treating inflammatory, autoimmune and respiratory diseases
CA2398728A1 (en) 2000-01-28 2001-08-02 Ivars Kalvins Novel melanocortin receptor agonists and antagonists
EP1254115A2 (en) 2000-02-11 2002-11-06 Bristol-Myers Squibb Company Cannabinoid receptor modulators, their processes of preparation, and use of cannabinoid receptor modulators for treating respiratory and non-respiratory diseases
MXPA02010608A (en) 2000-04-27 2003-05-14 Abbott Lab Substituted phenyl farnesyltransferase inhibitors.
US20020019527A1 (en) 2000-04-27 2002-02-14 Wei-Bo Wang Substituted phenyl farnesyltransferase inhibitors
JP2001354563A (en) 2000-06-09 2001-12-25 Sankyo Co Ltd Medicine comprising substituted benzylamines
US6448281B1 (en) 2000-07-06 2002-09-10 Boehringer Ingelheim (Canada) Ltd. Viral polymerase inhibitors
US6417367B1 (en) 2000-08-11 2002-07-09 Pfizer Inc. Methods of making quinoline amides
US20030050211A1 (en) 2000-12-14 2003-03-13 Unilever Home & Personal Care Usa, Division Of Conopco, Inc. Enzymatic detergent compositions
WO2002051396A1 (en) 2000-12-26 2002-07-04 Sankyo Company, Limited Pharmaceutical compositions containing cyclobutene derivatives
JP2002302439A (en) 2001-01-19 2002-10-18 Sankyo Co Ltd Ileum-type bile acid transporter inhibitor containing cyclobutene derivative
US20040106635A1 (en) 2001-03-29 2004-06-03 Iwao Takamuro Spiroisoquinoline compound, a method for preparing the same and an intermediate thereof
DE10121003A1 (en) 2001-04-28 2002-12-19 Aventis Pharma Gmbh Anthranilic acid amides, processes for their preparation, their use as medicaments and pharmaceutical preparations containing them
MY130373A (en) 2001-10-29 2007-06-29 Malesci Sas Linear basic compounds having nk-2 antagonist activity and formulations thereof
ES2303558T3 (en) 2001-11-27 2008-08-16 F. Hoffmann-La Roche Ag DERIVATIVES OF BENZOTIAZOL.
CA2468716A1 (en) 2001-11-28 2003-06-05 Fujisawa Pharmaceutical Co., Ltd. Heterocyclic amide compounds as apolipoprotein b inhibitors
TWI261053B (en) 2001-12-06 2006-09-01 Novartis Ag Amidoacetonitrile compounds, their preparation, compositions and use as pesticides
SE0104248D0 (en) * 2001-12-14 2001-12-14 Astrazeneca Ab Method of treatment
ATE392897T1 (en) 2002-02-04 2008-05-15 Hoffmann La Roche QUINOLINE DERIVATIVES AS NPY ANTAGONISTS
IL164212A0 (en) 2002-03-28 2005-12-18 Applied Research Systems Thiazolidine carboxamide derivatives, their preparation and pharmaceutical compositions containing them
EP1496892B1 (en) 2002-04-11 2011-01-26 Merck Sharp & Dohme Corp. 1h-benzo(f)indazol-5-yl derivatives as selective glucocorticoid receptor modulators
FR2838739B1 (en) 2002-04-19 2004-05-28 Sanofi Synthelabo N- [PHENYL (PIPERIDIN-2-YL) METHYL) BENZAMIDE DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC APPLICATION
PL372936A1 (en) 2002-04-26 2005-08-08 Ishihara Sangyo Kaisha, Ltd. Pyridine compounds or salts thereof and herbicides containing the same
US7375093B2 (en) 2002-07-05 2008-05-20 Intrexon Corporation Ketone ligands for modulating the expression of exogenous genes via an ecdysone receptor complex
FR2842805A1 (en) 2002-07-29 2004-01-30 Sanofi Synthelabo N- [PHENYL (PIPERIDIN-2-YL) METHYL] BENZAMIDE DERIVATIVES, THEIR PREPARATION AND APPLICATION THERAPEUTICS
GB0221697D0 (en) 2002-09-18 2002-10-30 Unilever Plc Novel compouds and their uses
WO2004029204A2 (en) 2002-09-27 2004-04-08 Merck & Co., Inc. Substituted pyrimidines
JP2004115450A (en) 2002-09-27 2004-04-15 Tanabe Seiyaku Co Ltd Medicinal composition
WO2004039795A2 (en) 2002-10-29 2004-05-13 Fujisawa Pharmaceutical Co., Ltd. Amide compounds for the treatment of hyperlipidemia
ATE479683T1 (en) 2002-11-05 2010-09-15 Glaxo Group Ltd ANTIBACTERIAL AGENTS
JP4261884B2 (en) 2002-11-26 2009-04-30 株式会社ネオス Water-soluble metalworking fluid composition
JP2004203871A (en) 2002-12-13 2004-07-22 Yamanouchi Pharmaceut Co Ltd Medicinal composition
WO2004058755A2 (en) 2002-12-24 2004-07-15 Biofocus Plc Compound libraries of imidazo[1,5-a]pyridin-3-yl derivatives and related heterobicycles for targetting compounds capable of binding to g-protein coupled receptors
US7601868B2 (en) 2003-02-12 2009-10-13 Takeda Pharmaceutical Company Limited Amine derivative
WO2004099148A1 (en) 2003-05-09 2004-11-18 Pharmacia & Upjohn Company Llc Substituted pyrimidine derivatives
EP1660454A1 (en) 2003-07-07 2006-05-31 Vernalis (R&D) Limited Azacyclic compounds as inhibitors of sensory neurone specific channels
CA2573951A1 (en) 2003-07-18 2005-01-27 Virochem Pharma Inc. Spiro compounds and methods for the modulation of chemokine receptor activity
US7094791B2 (en) 2003-07-31 2006-08-22 Avalon Pharmaceuticals, Inc. Derivatives of 3-hydroxy-pyrrole-2,4-dicarboxylic acid and uses thereof
EP1664042A1 (en) 2003-09-03 2006-06-07 Galapagos N.V. IMIDAZO 1,5-a PYRIDINE OR IMIDAZO 1,5-a PIPERIDINE DERIVATIVES AND THEIR USE FOR THE PREPARATION OF MEDICAMENT AGAINST 5-HT2A RECEPTOR-RELATED DISORDERS
GB0320983D0 (en) 2003-09-08 2003-10-08 Biofocus Plc Compound libraries
CA2579609A1 (en) 2003-09-10 2005-03-17 Virochem Pharma Inc. Spirohydantoin compounds and methods for the modulation of chemokine receptor activity
WO2005042524A1 (en) 2003-10-30 2005-05-12 Virochem Pharma Inc. Pyridine carboxamide and methods for inhibiting hiv integrase
JP2007520543A (en) 2004-02-04 2007-07-26 ファイザー・プロダクツ・インク Substituted quinoline compounds
GB0403578D0 (en) 2004-02-18 2004-03-24 Biofocus Discovery Ltd Compounds which interact with the G-protein coupled receptor family
WO2005085227A1 (en) 2004-03-02 2005-09-15 Smithkline Beecham Corporation Inhibitors of akt activity
CN1972914A (en) 2004-03-31 2007-05-30 詹森药业有限公司 Non-imidazole heterocyclic compounds as histamine H3-receptor ligands
DE102004022897A1 (en) 2004-05-10 2005-12-08 Bayer Cropscience Ag Azinyl-imidazoazines
US7550499B2 (en) 2004-05-12 2009-06-23 Bristol-Myers Squibb Company Urea antagonists of P2Y1 receptor useful in the treatment of thrombotic conditions
WO2005115374A1 (en) 2004-05-29 2005-12-08 7Tm Pharma A/S Crth2 receptor ligands for therapeutic use
DE102004039876A1 (en) 2004-06-23 2006-01-26 Lanxess Deutschland Gmbh Preparation of fluorinated 1,3-benzodioxanes
DE102004031323A1 (en) 2004-06-29 2006-01-19 Bayer Cropscience Ag Substituted Pyridazincarboxamide and derivatives thereof
MX2007002410A (en) 2004-08-30 2007-05-07 Neuromed Pharmaceuticals Ltd Urea derivatives as calcium channel blockers.
AR050926A1 (en) 2004-09-03 2006-12-06 Astrazeneca Ab BENZAMIDE DERIVATIVES AS INHIBITORS OF HISTONADESACETILASE (HDAC)
KR100677149B1 (en) 2004-11-12 2007-02-02 삼성전자주식회사 Ink composition
JPWO2006057448A1 (en) 2004-11-26 2008-06-05 武田薬品工業株式会社 Arylalkanoic acid derivatives
EP1831194A4 (en) 2004-12-21 2009-12-02 Astrazeneca Ab HETEROCYCLIC MCHr1 ANTAGONISTS AND THEIR USE IN THERAPY
US20060173183A1 (en) 2004-12-31 2006-08-03 Alantos Pharmaceuticals, Inc., Multicyclic bis-amide MMP inhibitors
EP1833832A1 (en) 2004-12-31 2007-09-19 GPC Biotech AG Napthyridine compounds as rock inhibitors
US20080146612A1 (en) 2005-01-27 2008-06-19 Astrazeneca Ab Novel Biaromatic Compounds, Inhibitors of the P2X7-Receptor
WO2006094246A2 (en) 2005-03-03 2006-09-08 Sirtris Pharmaceuticals, Inc. N-arylmethyl benzamide sirtuin modulators
JP5007788B2 (en) 2005-05-16 2012-08-22 日産化学工業株式会社 Dihydroazole-substituted benzamide compounds and pest control agents
US20070155738A1 (en) 2005-05-20 2007-07-05 Alantos Pharmaceuticals, Inc. Heterobicyclic metalloprotease inhibitors
AU2006252768A1 (en) 2005-06-02 2006-12-07 Bayer Cropscience Ag Phenylalkyl substituted heteroaryl devivatives
JP5340729B2 (en) 2005-06-13 2013-11-13 メルク シャープ エンド ドーム リミテッド Therapeutic agent
ES2525217T3 (en) 2005-06-27 2014-12-19 Exelixis Patent Company Llc LXR modulators based on imidazole
CA2609319C (en) 2005-07-15 2014-02-04 Nissan Chemical Industries, Ltd. Thiophene compounds and thrombopoietin receptor activators
CN100494167C (en) 2005-08-02 2009-06-03 天津大学 Anti-hepatitis B virus tertiary amine oxide and preparation method and uses for manufacturing same
WO2007017093A1 (en) 2005-08-04 2007-02-15 Bayer Healthcare Ag Substituted 2-benzyloxy-benzoic acid amide derivatives
EP1764098A1 (en) 2005-09-17 2007-03-21 Speedel Experimenta AG Diaminoalcohols derivatives for the treatment of Alzheimer, malaria, HIV
CA2628875A1 (en) 2005-11-10 2007-05-18 Bayer Healthcare Ag Diaryl ureas for treating diabetic neuropathy
JP5075832B2 (en) 2005-11-10 2012-11-21 バイエル・ファルマ・アクチェンゲゼルシャフト Diarylureas for treating pulmonary hypertension
US7888376B2 (en) 2005-11-23 2011-02-15 Bristol-Myers Squibb Company Heterocyclic CETP inhibitors
DE102005059479A1 (en) 2005-12-13 2007-06-14 Merck Patent Gmbh hydroxyquinoline
CA2633411A1 (en) 2005-12-15 2007-06-21 Bayer Healthcare Ag Diaryl ureas for treating inflammatory skin, eye and/or ear diseases
JP2009524585A (en) 2005-12-15 2009-07-02 バイエル・ヘルスケア・アクチェンゲゼルシャフト Diarylureas for treating viral infections
TWI412322B (en) 2005-12-30 2013-10-21 Du Pont Isoxazolines for controlling invertebrate pests
CN101410392A (en) 2006-01-27 2009-04-15 阿斯利康(瑞典)有限公司 Amide substituted quinolines
WO2007097470A2 (en) 2006-02-23 2007-08-30 Takeda Pharmaceutical Company Limited Fused nitrogen- comprising heterocyclic compound
DE102006012251A1 (en) 2006-03-15 2007-11-08 Grünenthal GmbH Substituted 4-aminoquinazoline derivatives and their use for the preparation of medicaments
CA2652926A1 (en) 2006-05-26 2007-12-06 Bayer Healthcare Llc Drug combinations with substituted diaryl ureas for the treatment of cancer
GB0611152D0 (en) 2006-06-06 2006-07-19 Ucb Sa Therapeutic agents
WO2008003746A1 (en) 2006-07-06 2008-01-10 Bayer Cropscience Sa N-(4-pyridin-2-ylbutyl) carboxamide derivatives, their process of preparation and their use as fungicides
EP2079699A1 (en) 2006-07-25 2009-07-22 Envivo Pharmaceuticals, Inc. Quinoline derivatives
US7834023B2 (en) 2006-09-20 2010-11-16 Portola Pharmaceuticals, Inc. Substituted dihydroquinazolines as platelet ADP receptor inhibitors
WO2008056687A1 (en) 2006-11-09 2008-05-15 Daiichi Sankyo Company, Limited Novel spiropiperidine derivative
WO2008063667A1 (en) 2006-11-20 2008-05-29 Alantos Pharmaceuticals Holding, Inc. Heterotricyclic metalloprotease inhibitors
AU2007321922A1 (en) 2006-11-20 2008-05-29 Alantos Pharmaceuticals Holding, Inc. Heterobicyclic matrix metalloprotease inhibitors
WO2008073825A1 (en) 2006-12-08 2008-06-19 Exelixis, Inc. Lxr and fxr modulators
CA2673736A1 (en) 2006-12-21 2008-07-03 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
US20080293711A1 (en) 2007-03-08 2008-11-27 Clark Michael P Chemokine receptor modulators
JP2010523663A (en) 2007-04-11 2010-07-15 メルク・シャープ・エンド・ドーム・コーポレイション CGRP receptor antagonist with tertiary amide, sulfonamide, carbamic acid and urea end groups
US8993588B2 (en) 2007-06-05 2015-03-31 Merck Sharp & Dohme Corp. Carboxamide heterocyclic CGRP receptor antagonists
JP2010533159A (en) 2007-07-09 2010-10-21 アストラゼネカ アクチボラグ Compound 947
US20110028507A1 (en) 2007-08-10 2011-02-03 Crystalgenomics, Inc. Pyridine derivatives and methods of use thereof
JP2010535773A (en) 2007-08-10 2010-11-25 グラクソスミスクライン エルエルシー Nitrogen-containing bicyclic chemicals for treating viral infections
US20090082358A1 (en) 2007-09-20 2009-03-26 Nobuko Nishimura Vanilloid receptor ligands and their use in treatments
US8476297B2 (en) 2007-12-04 2013-07-02 Amgen Inc. TRP-M8 receptor ligands and their use in treatments
JPWO2009072621A1 (en) 2007-12-07 2011-04-28 日産化学工業株式会社 Substituted dihydroazole compounds and pest control agents
CN102036981B (en) 2008-03-18 2015-04-08 默沙东公司 Substituted 4-hydroxypyrimidine-5-carboxamides
CN101981025A (en) 2008-03-28 2011-02-23 万有制药株式会社 Diarylmethylamide derivative having antagonistic activity on melanin-concentrating hormone receptor
CA2720530A1 (en) 2008-04-11 2009-10-15 Merck Serono S.A. Sulfonamides
DE102008019838A1 (en) 2008-04-19 2009-12-10 Boehringer Ingelheim International Gmbh New arylsulfonylglycine derivatives, their preparation and their use as pharmaceuticals
EP2299813B1 (en) 2008-06-12 2015-12-16 Merck Sharp & Dohme Corp. Branched 3- and 6-substituted quinolines as cgrp receptor antagonists
US20110263657A1 (en) 2008-06-25 2011-10-27 Bayer Schering Pharma Aktiengesellschaft Diaryl ureas for treating heart failure
CN101343313B (en) 2008-09-03 2011-11-16 南京农业大学 Glucose dipeptide compounds, preparation method and application thereof
TWI389913B (en) 2008-09-08 2013-03-21 Lg Life Sciences Ltd Fused heterocyclic compound
WO2010028175A1 (en) 2008-09-08 2010-03-11 The Board Of Trustees Of The Leland Stanford Junior University Modulators of aldehyde dehydrogenase activity and methods of use thereof
WO2010046780A2 (en) 2008-10-22 2010-04-29 Institut Pasteur Korea Anti viral compounds
TW201030001A (en) 2008-11-14 2010-08-16 Amgen Inc Pyridine and pyrimidine derivatives as phosphodiesterase 10 inhibitors
UY32525A (en) 2009-04-03 2010-10-29 Astrazeneca Ab COMPOUNDS THAT HAVE AGONISTIC ACTIVITY OF THE GLUCOCORTICOSTEROID RECEPTOR
MX2011012712A (en) 2009-05-29 2012-01-30 Raqualia Pharma Inc Aryl substituted carboxamide derivatives as calcium or sodium channel blockers.
JP5384733B2 (en) 2009-06-02 2014-01-08 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Derivatives of 6,7-dihydro-5H-imidazo [1,2-a] imidazole-3-carboxylic acid amide
EP2459187B1 (en) 2009-07-29 2021-01-06 Olsen, Elise Compositions and methods for inhibiting hair growth
DE102009028929A1 (en) 2009-08-27 2011-07-07 Bayer Schering Pharma Aktiengesellschaft, 13353 Heterocyclic-substituted 2-acetamido-5-aryl-1,2,4-triazolones and their use
UY32940A (en) 2009-10-27 2011-05-31 Bayer Cropscience Ag AMIDAS REPLACED WITH HALOGENO RENT AS INSECTICIDES AND ACARICIDES
MX2012007872A (en) 2010-01-06 2012-10-03 Joseph P Errico Methods and compositions of targeted drug development.
WO2011094890A1 (en) 2010-02-02 2011-08-11 Argusina Inc. Phenylalanine derivatives and their use as non-peptide glp-1 receptor modulators
US20110212969A1 (en) 2010-02-26 2011-09-01 Millennium Pharmaceuticals, Inc. Substituted hydroxamic acids and uses thereof
WO2011142359A1 (en) 2010-05-10 2011-11-17 日産化学工業株式会社 Spiro compound and drug for activating adiponectin receptor
WO2011146300A1 (en) 2010-05-17 2011-11-24 Merck Sharp & Dohme Corp. Novel prolylcarboxypeptidase inhibitors
CN103025723A (en) 2010-05-27 2013-04-03 拜尔农作物科学股份公司 Pyridinylcarboxylic acid derivatives as fungicides
WO2012052540A1 (en) 2010-10-21 2012-04-26 Universitaet Des Saarlandes Selective cyp11b1 inhibitors for the treatment of cortisol dependent diseases
CN108484589A (en) 2010-11-05 2018-09-04 赛诺米克斯公司 The useful compound of conditioning agent as TRPM8
EP2649075B1 (en) 2010-12-08 2018-04-25 The U.S.A. As Represented By The Secretary, Department Of Health And Human Services Substituted pyrazolopyrimidines as glucocerebrosidase activators
EP2651412A4 (en) 2010-12-14 2014-08-13 Beth Israel Hospital Androgen receptor inhibitors and methods of use thereof
WO2012079624A1 (en) 2010-12-15 2012-06-21 Ppg Europe Bv Drier composition and use thereof
WO2012083190A1 (en) 2010-12-17 2012-06-21 The Rockefeller University Insect odorant receptor antagonists
CN103635472B (en) 2011-02-28 2018-01-12 阵列生物制药公司 Serine/threonine kinase inhibitor
WO2012120398A1 (en) 2011-03-04 2012-09-13 Pfizer Limited Aryl substituted carboxamide derivatives as trpm8 modulators

Also Published As

Publication number Publication date
MX2013015058A (en) 2014-01-20
US20130158034A1 (en) 2013-06-20
WO2012177896A1 (en) 2012-12-27
EP2723718A1 (en) 2014-04-30
CA2839703A1 (en) 2012-12-27
US8778941B2 (en) 2014-07-15
JP2014517074A (en) 2014-07-17
AU2012272898A1 (en) 2013-04-11

Similar Documents

Publication Publication Date Title
US9096527B2 (en) TRPM8 antagonists and their use in treatments
US7579347B2 (en) Vanilloid receptor ligands and their use in treatments
AU738037B2 (en) Nicotinamide derivatives
AU2003233671B8 (en) Calcium receptor modulating arylalkylamines
JP5542809B2 (en) Organic compounds
US20050165015A1 (en) Vanilloid receptor ligands and their use in treatments
US8778941B2 (en) TRPM8 antagonists and their use in treatments
US7553848B2 (en) Vanilloid receptor ligands and their use in treatments
JP2007524673A (en) Vanilloid receptor ligands and their use in therapy
CZ315196A3 (en) Sulfonamide derivatives and pharmaceutical composition containing thereof
US20050165028A1 (en) Vanilloid receptor ligands and their use in treatments
EA012589B1 (en) Derivatives of 4-(benzyl)piperazine carboxylic acid phenylamide and associated compounds as fatty acid amide hydrolase inhibitors for the treatment of anxiety, pain and other disorders
WO2014025651A1 (en) Chroman derivatives as trpm8 inhibitors
US8476297B2 (en) TRP-M8 receptor ligands and their use in treatments
US8952009B2 (en) Chroman derivatives as TRPM8 inhibitors

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION