WO2003055848A2 - Urea derivatives as vr1- antagonists - Google Patents

Urea derivatives as vr1- antagonists Download PDF

Info

Publication number
WO2003055848A2
WO2003055848A2 PCT/EP2002/014216 EP0214216W WO03055848A2 WO 2003055848 A2 WO2003055848 A2 WO 2003055848A2 EP 0214216 W EP0214216 W EP 0214216W WO 03055848 A2 WO03055848 A2 WO 03055848A2
Authority
WO
WIPO (PCT)
Prior art keywords
phenyl
urea
alkyl
hydroxyethyl
biphenyl
Prior art date
Application number
PCT/EP2002/014216
Other languages
French (fr)
Other versions
WO2003055848A3 (en
Inventor
Takeshi Yura
Muneto Mogi
Yuka Ikegami
Tsutomu Masuda
Toshio Kokubo
Klaus Urbahns
Nagahiro Yoshida
Makiko Marumo
Masahiro Shiroo
Masaomi Tajimi
Keisuke Takeshita
Toshiya Moriwaki
Yasuhiro Tsukimi
Original Assignee
Bayer Healthcare Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from JP2001395033A external-priority patent/JP2003192660A/en
Priority claimed from JP2001395032A external-priority patent/JP2003192659A/en
Application filed by Bayer Healthcare Ag filed Critical Bayer Healthcare Ag
Priority to AU2002358700A priority Critical patent/AU2002358700A1/en
Priority to CA002469967A priority patent/CA2469967A1/en
Priority to US10/499,785 priority patent/US20050154230A1/en
Priority to JP2003556380A priority patent/JP2005513154A/en
Priority to EP02793004A priority patent/EP1461311A2/en
Publication of WO2003055848A2 publication Critical patent/WO2003055848A2/en
Publication of WO2003055848A3 publication Critical patent/WO2003055848A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/02Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings
    • C07D333/04Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom
    • C07D333/26Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur 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
    • C07D333/30Hetero atoms other than halogen
    • C07D333/36Nitrogen atoms
    • 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
    • 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
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • A61P29/02Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID] without antiinflammatory effect
    • 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
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • 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
    • C07C275/04Derivatives 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 acyclic carbon atoms
    • C07C275/20Derivatives 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 acyclic carbon atoms of an unsaturated carbon skeleton
    • C07C275/24Derivatives 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 acyclic carbon atoms of an unsaturated carbon skeleton containing six-membered aromatic rings
    • 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
    • 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
    • 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
    • C07C275/32Derivatives 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 singly-bound oxygen atoms
    • 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
    • C07C275/40Derivatives 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 nitrogen atoms not being part of nitro or nitroso groups
    • 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
    • C07C275/42Derivatives 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 carboxyl groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/23Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and nitrogen atoms, not being part of nitro or nitroso groups, bound to the same carbon skeleton
    • C07C323/39Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and nitrogen atoms, not being part of nitro or nitroso groups, bound to the same carbon skeleton at least one of the nitrogen atoms being part of any of the groups, X being a hetero atom, Y being any atom
    • C07C323/43Y being a hetero atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/56Ring systems containing three or more rings
    • C07D209/80[b, c]- or [b, d]-condensed
    • C07D209/82Carbazoles; Hydrogenated carbazoles
    • C07D209/88Carbazoles; Hydrogenated carbazoles 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 carbon atoms of the ring system
    • 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
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/02Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with only hydrogen atoms or radicals containing only carbon and hydrogen atoms, directly attached to carbon atoms of the nitrogen-containing ring; Alkylene-bis-isoquinolines
    • C07D217/06Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with only hydrogen atoms or radicals containing only carbon and hydrogen atoms, directly attached to carbon atoms of the nitrogen-containing ring; Alkylene-bis-isoquinolines with the ring nitrogen atom acylated by carboxylic or carbonic acids, or with sulfur or nitrogen analogues thereof, e.g. carbamates
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/54Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings condensed with carbocyclic rings or ring systems
    • C07D231/56Benzopyrazoles; Hydrogenated benzopyrazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/12Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly or doubly bound nitrogen atoms
    • C07D295/135Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly or doubly bound nitrogen atoms with the ring nitrogen atoms and the substituent nitrogen atoms separated by carbocyclic rings or by carbon chains interrupted by carbocyclic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/34Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D307/38Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D307/52Radicals substituted by nitrogen atoms not forming part of a nitro radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D317/00Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms
    • C07D317/02Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 2
    • C07D317/06Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 2 condensed with carbocyclic rings or ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D317/00Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms
    • C07D317/08Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3
    • C07D317/44Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3 ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D317/46Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3 ortho- or peri-condensed with carbocyclic rings or ring systems condensed with one six-membered ring
    • C07D317/48Methylenedioxybenzenes or hydrogenated methylenedioxybenzenes, unsubstituted on the hetero ring
    • C07D317/50Methylenedioxybenzenes or hydrogenated methylenedioxybenzenes, unsubstituted on the hetero ring with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to atoms of the carbocyclic ring
    • C07D317/58Radicals substituted by nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/02Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings
    • C07D333/04Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom
    • C07D333/06Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to the ring carbon atoms
    • C07D333/14Radicals substituted by singly bound hetero atoms other than halogen
    • C07D333/20Radicals substituted by singly bound hetero atoms other than halogen by nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2602/00Systems containing two condensed rings
    • C07C2602/02Systems containing two condensed rings the rings having only two atoms in common
    • C07C2602/04One of the condensed rings being a six-membered aromatic ring
    • C07C2602/10One of the condensed rings being a six-membered aromatic ring the other ring being six-membered, e.g. tetraline
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2603/00Systems containing at least three condensed rings
    • C07C2603/02Ortho- or ortho- and peri-condensed systems
    • C07C2603/04Ortho- or ortho- and peri-condensed systems containing three rings
    • C07C2603/06Ortho- or ortho- and peri-condensed systems containing three rings containing at least one ring with less than six ring members
    • C07C2603/10Ortho- or ortho- and peri-condensed systems containing three rings containing at least one ring with less than six ring members containing five-membered rings
    • C07C2603/12Ortho- or ortho- and peri-condensed systems containing three rings containing at least one ring with less than six ring members containing five-membered rings only one five-membered ring
    • C07C2603/18Fluorenes; Hydrogenated fluorenes

Definitions

  • the present invention relates to an urea derivative, which is useful as an active ingredient of pharmaceutical preparations.
  • the urea derivatives of the present invention have vanilloid receptor (VRl) antagonistic activity, and can be used for the prophylaxis and treatment of diseases associated with VRl activity, in particular for the treatment of urge urinary incontinence, overactive bladder, chronic pain, neuropathic pain, postoperative pain, rheumatoid arthritic pain, neuralgia, neuropathies, algesia, nerve injury, ischaemia, neurodegeneration, stroke, incontinence and/or inflammatory disorders.
  • VRl vanilloid receptor
  • Vanilloid compounds are characterized by the presence of vanillyl group or a functionally equivalent group.
  • vanilloid compounds or vanilloid receptor modulators are vanillin (4-hydroxy-3-methoxy-benzaldehyde), guaiacol (2- methoxy-phenol), zingerone (4-/4-hydroxy-3-methoxyphenyl/-2-butanon), eugenol(2-methoxy4-/2-propenyl/phenol), and capsaicin (8-methy-N-vanillyl-6- noneneamide).
  • capsaicin the main pungent ingredient in "hot” chili peppers, is a specific neurotoxin that desensitizes C-fiber afferent neurons.
  • Capsaicin interacts with vanilloid receptors (VRl), which are predominantly expressed in cell bodies of dorsal root ganglia (DRG) or nerve endings of afferent sensory fibers including C- fiber nerve endings [Tominaga M, Caterina MJ, Malmberg AB, Rosen TA, Gilbert H, Skinner K, Raumann BE, Basbaum AI, Julius D: The cloned capsaicin receptor integrates multiple pain-producing stimuli. Neuron. 21 : 531-543, 1998].
  • VRl vanilloid receptors
  • the VRl receptor was recently cloned [Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD, Julius D: Nature 389: 816-824, (1997)] and identified as a nonselective cation channel with six transmembrane domains that is structurally related to the TRP (transient receptor potential) channel family. Binding of capsaicin to VRl allows sodium, calcium and possibly potassium ions to flow down their concentration gradients, causing initial depolarization and release of neurotrans- mitters from the nerve terminals. VRl can therefore be viewed as a molecular integrator of chemical and physical stimuli that elicit neuronal signals in a pathological conditions or diseases.
  • antagonists of the VRl receptor can be used for prophylaxis and treatment of the condition and diseases including chronic pain, neuropathic pain, postoperative pain, rheumatoid arthritic pain, neuralgia, neuropathies, algesia, nerve injury, ischaemia, neurodegeneration, stroke, incontinence, inflammatory disorders, urge urinary incontinence (UUI), and/or overactive bladder.
  • condition and diseases including chronic pain, neuropathic pain, postoperative pain, rheumatoid arthritic pain, neuralgia, neuropathies, algesia, nerve injury, ischaemia, neurodegeneration, stroke, incontinence, inflammatory disorders, urge urinary incontinence (UUI), and/or overactive bladder.
  • WO 2000/50387 discloses the compounds having a vanilloid agonist activity represented by the general formula:
  • X is an oxygen or sulfur atom
  • a p is -NHCH 2 - or -CH 2 -;
  • R a is a substituted or unsubstituted C alkyl group, or R al CO-;
  • R al is an alkyl group having 1 to 18 carbon atoms, an alkenyl group having 2 to 18 carbon atoms, or substituted or unsubstituted aryl group having 6 to 10 carbon atoms;
  • R is a hydrogen atom, an alkyl group having 1 to 6 carbon atoms, an alkoxy group having 1 to 6 carbon atoms, a haloalkyl group having 1 to 6 carbon atoms or a halogen atom;
  • R is a hydrogen atom, an alkyl group having 1 to 4 carbon atom, an aminoalkyl, a diacid monoester or .-alkyl acid; and the asteric mark * indicates a chiral carbon atom, and their pharmaceutically acceptable salts.
  • WO 2000/61581 discloses amine derivatives represented by the general formula:
  • R ⁇ R represent (F, F), (CF 3 , H), or (iPr, iPr)
  • WO 2000/75106 discloses the compounds represented by the general formula:
  • R is hydrogen, C M 2 alkyl, C . 8 cycloalkyl, or the like, and R is amino- C ⁇ - 6 alkyl, aminocarbonyl-C ⁇ . 6 alkyl, or hydroxyaminocarbonyl C ⁇ - 6 alkyl;
  • R 90 and R 91 are independently selected from the group consisting of H, C ⁇ _ 6 alkyl, Ci- ⁇ alkylthio, C ⁇ . 6 alkoxy, fluoro, chloro, bromo, iodo, and nitro;
  • VRl activity in particular for the treatment of urge urinary incontinence and/or overactive bladder have been desired.
  • This invention is to provide urea derivatives of the formula (I), their tautomeric and stereoisomeric form, and salts thereof: wherein
  • X is C ⁇ - 6 alkyl substituted by phenyl or naphthyl (wherein said phenyl and naphthyl are optionally substituted by R 11 , R 12 and R 13 ), aryl or heterocyclic ring ,
  • aryl and heterocyclic ring are optionally substituted by R 1 1 , R 12 and R 13 and are selected from the group consisting of phenyl, naphthyl, pyridyl, carbazolyl, fluorenyl, thienyl, pyrimidyl, benzodioxolyl, indazolyl, and quinolyl,
  • R , R and R independently represent hydrogen, halogen, C ⁇ . 6 alkyl, mono-, di-, or tri- halogen substituted C ⁇ _ 6 alkyl, nitro, cyano, C ⁇ _ 6 alkoxy, hydroxy, piperidino, furyl, thienyl, benzyloxy, anilino, naphthyl, d. 6 alkylcarbamoyl, carbamoyl, carboxyl, amino, C ⁇ . 6 alkylamino, di(C ⁇ - 6 alkyl)amino, C ⁇ . 6 alkoxy- carbonyl, benzyl, phenoxy, C ⁇ .
  • substituents are each different or identical and selected from the group consisting of hydrogen, halogen, C ⁇ . 6 alkyl, C ⁇ . 6 alkoxy, pyridyl, mono-, di-, or tri- halogen substituted C ⁇ - 6 alkyl, nitro, cyano, benzyloxy, thienyl, C ⁇ _ 6 alkanoyl, C ⁇ - 6 alkoxycarbonyl, C ⁇ - 6 alkylthio, di(C ⁇ _ 6 alkyl)amino, and C ⁇ . 6 alkylamino, mono, di, or tri halogen substituted C ⁇ . 6 alkyloxy;
  • R 1 is hydrogen
  • R 2 is hydrogen
  • R 3 is hydrogen, or
  • R 2 and R 3 together form -(CH 2 ) m - (wherein m represents 1, 2, 3 or 4), or R 1 and R 3 together form -(CH 2 ) n - (wherein n represents 1, 2, or 3);
  • R 4 is hydrogen, halogen, C). 6 alkoxy, hydroxy, C ⁇ . 6 alkoxy substituted benzyloxy, sulfamoyl, C ⁇ - 6 alkylsulfamoyl, di(C ⁇ - 6 alkyl)sulfamoyl, di (C ⁇ - 6 alkyl)aminoCi_ 6 alkylene sulfamoyl, hydroxy C ⁇ . 6 alkyl piperazinosulfonyl, C ⁇ - 6 alkylsulfonylamino, nitro, amino, C ⁇ _ 6 alkanoylamino, C ⁇ _ 6 alkoxyC ⁇ _ 6 alkyleneoxy,
  • R 5 is hydrogen, halogen, C ⁇ _ 6 alkoxy, hydroxy, C ⁇ - 6 alkoxy substituted benzyloxy, sulfamoyl, C ⁇ . 6 alkylsulfamoyl, di (C ⁇ _ 6 alkyl)sulfamoyl, di(C ⁇ . 6 alkyl)amino C ⁇ - 6 alkylene sulfamoyl, hydroxy C ⁇ _ 6 alkyl piperazinosulfonyl,
  • R 4 and R 5 together form -O-(CH 2 )-O-;
  • R 6 is hydrogen, halogen, C ⁇ _ 6 alkyl, mono-, di-, or tri- halogen substituted C ⁇ . 6 alkyl, nitro, cyano, C ⁇ _ 6 alkoxy, hydroxy, C ⁇ . 6 alkylcarbamoyl, carbamoyl, carboxyl, amino, C ⁇ _ 6 alkylamino, di(C ⁇ - 6 alkyl) amino, C ⁇ - 6 alkoxycarbonyl, phenyl, benzyl, phenoxy, halogen substituted phenoxy, C ⁇ - 6 alkylthio, C ⁇ _ 6 alkanoyl, C ⁇ . 6 alkanoylamino, hydroxy substituted C ⁇ - 6 alkyl, mono-, di-, or tri- halogen substituted C ⁇ - 6 alkoxy.
  • Alkyl per se and "alk” and "alkyl” in alkoxy, alkanoyl, alkylthio, alkylamino, alkyl- aminocarbonyl, alkylaminosulphonyl, alkylsulphonylamino, alkoxycarbonyl, alkoxy- carbonylamino, alkylcarbamoyl and alkanoylamino represent a linear or branched alkyl radical having generally 1 to 6, preferably 1 to 4 and particularly preferably 1 to 3 carbon atoms, representing illustratively and preferably methyl, ethyl, n-propyl, isopropyl, tert-butyl, n-pentyl and n-hexyl.
  • Alkoxy illustratively and preferably represents methoxy, ethoxy, n-propoxy, iso- propoxy, tert-butoxy, n-pentoxy and n-hexoxy.
  • Alkanoyl illustratively and preferably represents acetyl and propanoyl.
  • Alkylamino represents an alkylamino radical having one or two (independently selected) alkyl substituents, illustratively and preferably representing methylamino, ethylamino, n-propylamino, isopropylamino, tert-butylamino, n-pentylamino, n- hexyl-amino, N,N-dimethylamino, N,N-diethylamino, N-ethyl-N-methylamino, N- methyl-N-n-propylamino, N-isopropyl-N-n-propylamino, N-t-butyl-N-methylamino, N-ethyl-N-n-pentylamino and N-n-hexyl-N-methylamino.
  • Alkylaminocarbonyl or alkylcarbamoyl represents an alkylaminocarbonyl radical having one or two (independently selected) alkyl substituents, illustratively and preferably representing methylaminocarbonyl, ethylaminocarbonyl, n-propylamino- carbonyl, isopropylamino-carbonyl, tert-butylaminocarbonyl, n-pentylamino- carbonyl, n-hexylaminocarbonyl, N,N-dimethylaminocarbonyl, N,N-diethylamino- carbonyl, N-ethyl-N-methylaminocarbonyl, N-methyl-N-n-propylaminocarbonyl, N- isopropyl-N-n-propylaminocarbonyl, N-t-butyl-N-methylaminocarbonyl, N-ethyl-N- n-
  • Alkoxycarbonyl illustratively and preferably represents methoxycarbonyl, ethoxy- carbonyl, n-propoxycarbonyl, isopropoxycarbonyl, tert-butoxycarbonyl, n-pentoxy- carbonyl and n-hexoxycarbonyl.
  • Alkoxycarbonylamino illustratively and preferably represents methoxycarbonylamino, ethoxycarbonylamino, n-propoxycarbonylamino, isopropoxycarbonylamino, tert-butoxycarbonylamino, n-pentoxycarbonylamino and n-hexoxycarbonylamino.
  • Alkanoylamino illustratively and preferably represents acetylamino and ethyl- carbonylamino.
  • Halogen represents fluorine, chlorine, bromine and iodine.
  • Aryl per se and in arylamino and in arylcarbonyl represents a mono- to tricyclic aromatic carbocyclic radical having generally 6 to 14 carbon atoms, and more preferably from 6-10 carbon atoms, optionally substituted with one or more substituents.
  • aryl radicals include, but are not limited to phenyl, naphthyl, indenyl, azulenyl, fluorenyl, anthracenyl, biphenyl, fluorenonyl and the like.
  • Heterocyclic ring refers to a 3- to 15-membered ring radical which consists of carbon atoms and from one to five heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur.
  • the heterocyclic ring radical may be a monocyclic, bicyclic or tricyclic ring system, which may include fused or bridged ring systems and may be partially or fully saturated or aromatic.
  • Such rings include, but are not limited to thienyl, benzothienyl, furanyl, benzofuranyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, isothiazolyl, thiazolyl, oxazolyl, isoxazolyl, triazolyl, tetrazolyl, imidazolyl, thiadiazolyl, benzothiadiazolyl, oxadiazolyl, benzothiazolyl, indolyl, carbazolyl, quinolinyl, isoquinolinyl, benzodioxolyl, indazolyl, indazolinolyl and the like
  • This invention is also to provide a method for treating or preventing a disorder or disease associated with VRl activity in a human or animal subject, comprising administering to said subject a therapeutically effective amount of the urea derivative shown in the formula (I), its tautomeric or stereoisomeric form, or a physiologically acceptable salt thereof.
  • this invention is to provide a use of the urea derivative shown in the formula (I), its tautomeric or stereoisomeric form, or a physiologically acceptable salt thereof in the preparation of a medicament.
  • said medicament is suitable for treating or preventing a disorder or disease associated with VRl activity.
  • the compounds of the present invention surprisingly show excellent VRl activity. They are, therefore, suitable for the production of medicament or medical composition, which may be useful to treat VRl related diseases.
  • the urea derivatives of the present invention inhibit VRl , they are useful for treatment and prophylaxis of diseases as follows:
  • the compounds of formula (I) are those wherein:
  • Y is I-i;
  • X is phenyl optionally substituted by R 11 , R 12 and R 13 , phenyl C ⁇ _ 6 alkyl (wherein said phenyl is optionally substituted by R u , R 12 and R 13 ), or naphthyl optionally substituted by R 11 , R 12 and R 13 ,
  • R 1 1 , R 12 and R 13 independently represent hydrogen, halogen, C ⁇ _ 6 alkyl, mono-, di-, or tri- halogen substituted C].
  • the compounds of formula (I) are those wherein:
  • Y is I-i
  • R 1 is hydrogen
  • R is hydrogen
  • R is hydrogen
  • the compounds of formula (I) are those wherein:
  • Y is I-i
  • X is phenyl optionally substituted by R 11 , R 12 and R 13 , phenyl C ⁇ . 6 alkyl (wherein said phenyl is optionally substituted by R 11 , R 12 and R 13 ), or naphthyl optionally substituted by R 1 ' , R 12 and R 13 ,
  • R 11 , R 12 and R 13 independently represent hydrogen, halogen, C ⁇ . 6 alkyl, mono-, di-, or tri- halogen substituted C ⁇ . 6 alkyl, nitro, C ⁇ . 6 alkoxy, C ⁇ - 6 alkoxycarbonyl, phenoxy, C ⁇ - 6 alkylthio, or C ⁇ - 6 alkanoyl.
  • R is hydrogen; and R and R together form -(CH2) m - (wherein m represents 1, 2, 3 or 4).
  • the compounds of formula (I) are those wherein: Y is I-i;
  • R 1 and R 3 together form -(CH 2 ) n - (wherein n represents 1, 2, or 3) and
  • R is hydrogen
  • urea derivative of formula (I) can be those wherein:
  • Y is I-ii
  • R 6 is hydrogen, halogen, C]. 6 alkyl, mono-, di-, or tri- halogen substituted C ⁇ . 6 alkyl, phenyl, or C ⁇ - 6 alkoxy.
  • the compounds of formula (I) are those wherein:
  • X is C ⁇ - 6 alkyl substituted by phenyl or naphthyl (wherein said phenyl and naphthyl are optionally substituted by R 11 , R 12 and R 13 ), aryl or Heterocyclic ring ,
  • aryl and Heterocyclic ring are optionally substituted by R ⁇ , R 12 and R 13 and are selected from the group consisting of phenyl, naphthyl, pyridyl, carbazolyl, fluorenyl, thienyl, benzodioxolyl, indazolyl, and quinolyl,
  • R 6 is hydrogen, halogen, C ⁇ . 6 alkyl, mono-, di-, or tri- halogen substituted C]. 6 alkyl, phenyl, or C ⁇ - 6 alkoxy.
  • the preferable compounds of the present invention are as follows:
  • the medicaments of the present invention further comprise one or more pharmaceutically acceptable carrier and/or excipients.
  • the compound of the formula (I) of the present invention can be, but not limited to be, prepared by either of the methods [A], [B] and [C] below.
  • one or more of the substituents, such as amino group, carboxyl group, and hydroxyl group of the compounds used as starting materials or intermediates are ad- vantageously protected by a protecting group known to those skilled in the art. Examples of the protecting groups are described in
  • the compound [I-a] wherein X and R 6 are the same as defined above can be prepared by the reaction of a substituted 2-(2-aminophenyl)ethanol[II] (wherein R 6 is the same as defined above) and isocyanate of the formula [VI] (wherein X is the same as defined above).
  • the reaction may be carried out in a solvent including, for instance, halogenated hydrocarbons such as dichloromethane, chloroform and 1 ,2-dichloroethane; ethers such as diethyl ether, isopropyl ether, dioxane and tetrahydrofuran (THF) and 1,2- dimethoxyethane; aromatic hydrocarbons such as benzene, toluene and xylene; nitriles such as acetonitrile; amides such as N,N-dimethylformamide (DMF), N,N- dimethylacetamide (DMAC) and N-methylpyrrolidone (NMP); urea such as 1,3- dimethyl-2-imidazolidinone (DMI); sulfoxides such as dimethylsulfoxide (DMSO); and others.
  • halogenated hydrocarbons such as dichloromethane, chloroform and 1 ,2-dichloroethane
  • ethers
  • the reaction temperature can be optionally set depending on the compounds to be reacted.
  • the reaction temperature is usually, but not limited to, about 30 °C to 100 °C.
  • the reaction may be conducted for, usually, 30 minutes to 48 hours and preferably 1 to 24 hours.
  • the compound [I-b], [I-c] and [I-d] wherein X, R 4 and R 5 are the same as defined above, can be prepared using substituted benzylamines [III], substituted tetrahydro- isoquinolines [IV] and substituted tetrahydro-naphthalenylamine [V] as starting material, respectively, by the same method as for the compound [I-a].
  • the compound [I-a] (wherein X and R 6 are the same as defined above) can be prepared by reacting a substituted 2-(2-aminophenyl)ethanol[II] and carbamate of the formula [VII] (wherein X is the same as defined above and Y represents phenyl).
  • the reaction may be carried out in a solvent including, for instance, halogenated hydrocarbons such as dichloromethane, chloroform and 1,2-dichloroethane; ethers such as diethyl ether, isopropyl ether, dioxane and tetrahydrofuran (THF) and 1,2- dimethoxy ethane; aromatic hydrocarbons such as benzene, toluene and xylene; nitriles such as acetonitrile; amides such as N, N-dimethylformamide (DMF), N, N- dimethylacetamide (DMAC) and N-methylpyrrolidone(NMP); urea such as 1,3- dimethyl-2-imidazolidinone (DMI); sulfoxides such as dimethylsulfoxide (DMSO); and others.
  • the reaction temperature can be optionally set depending on the compounds to be reacted.
  • the reaction temperature is usually, but not limited to, about 20 °C
  • the compound [I-b], [I-c] and [I-d] wherein X, R 4 and R 5 are the same as defined above, can be prepared using substituted benzylamines [III], substituted tetrahydro- isoquinolines [IV] and substituted tetrahydro-naphthalenylamine [V] as starting material, respectively, by the same method as for the compound [I-a].
  • the compound [I-a] can be prepared by reacting amine of the formula [VIII] (wherein X is the same as defined above) and l, -carbonyldi(l,2,4-triazole) (CDT)[IX], and then adding substituted 2-(2-aminophenyl)ethanol[II] to the reaction mixture.
  • the reaction may be carried out in a solvent including, for instance, halogenated hydrocarbons such as dichloromethane, chloroform and 1,2-dichloroethane; ethers such as diethyl ether, isopropyl ether, dioxane and tetrahydrofuran (THF) and 1,2- dimethoxyethane; aromatic hydrocarbons such as benzene, toluene and xylene; nitriles such as acetonitrile; amides such as N, N-dimethylformamide (DMF), N, N- dimethylacetamide(DMAC) and N-methylpyrrolidone (NMP); urea such as 1,3- dimethyl-2-imidazolidinone(DMI); sulfoxides such as dimethylsulfoxide(DMSO); and others.
  • halogenated hydrocarbons such as dichloromethane, chloroform and 1,2-dichloroethane
  • ethers such as dieth
  • the reaction temperature can be optionally set depending on the compounds to be reacted.
  • the reaction temperature is usually, but not limited to, about 20 °C to 50 °C.
  • the reaction may be conducted for, usually, 30 minutes to 10 hours and preferably 1 to 24 hours.
  • the compound [I-b], [I-c] and [I-d] wherein X, R 4 and R 5 are the same as defined above, can be prepared using substituted benzylamines [III], substituted tetrahydro- isoquinolines [IV] and substituted tetrahydro-naphthalenylamine [V] as starting material, respectively, by the same method as for the compound [I-a].
  • substituted 2-(2-aminophenyl)ethanols [II], substituted benzylamines [III], substituted tetrahydroisoquinolines [IV], substituted tetrahydro-naphthalenylamine [V], Isocyanates [VI], carbamates [VII], amine [VIII] and CDT [IX] are commercially available or can be prepared by the use of known techniques or by method described in the examples.
  • Typical salts of the compound shown by the formula (I) include salts prepared by reaction of the compounds of the present invention with a mineral or organic acid, or an organic or inorganic base. Such salts are known as acid addition and base addition salts, respectively.
  • Acids to form acid addition salts include inorganic acids such as, without limitation, sulfuric acid, phosphoric acid, hydrochloric acid, hydrobromic acid, hydriodic acid and the like, and organic acids, such as, without limitation, p-toluenesulfonic acid, methanesulfonic acid, oxalic acid, p-bromophenylsulfonic acid, carbonic acid, succinic acid, citric acid, benzoic acid, acetic acid, and the like.
  • inorganic acids such as, without limitation, sulfuric acid, phosphoric acid, hydrochloric acid, hydrobromic acid, hydriodic acid and the like
  • organic acids such as, without limitation, p-toluenesulfonic acid, methanesulfonic acid, oxalic acid, p-bromophenylsulfonic acid, carbonic acid, succinic acid, citric acid, benzoic acid, acetic acid, and the like.
  • Base addition salts include those derived from inorganic bases, such as, without limitation, ammonium hydroxide, alkaline metal hydroxide, alkaline earth metal hydroxides, carbonates, bicarbonates, and the like, and organic bases, such as, without limitation, ethanolamine, triethylamine, tris(hydroxymethyl)aminomethane, and the like.
  • inorganic bases include, sodium hydroxide, potassium hydroxide, potassium carbonate, sodium carbonate, sodium bicarbonate, potassium bicarbonate, calcium hydroxide, calcium carbonate, and the like.
  • the compound of the present invention or a salts thereof, depending on its substituents, may be modified to form lower alkylesters or known other esters; and/or hydrates or other solvates. Those esters, hydrates, and solvates are included in the scope of the present invention.
  • the compound of the present invention may be administered in oral forms, such as, without limitation normal and enteric coated tablets, capsules, pills, powders, granules, elixirs, tinctures, solution, suspensions, syrups, solid and liquid aerosols and emulsions. They may also be administered in parenteral forms, such as, without limitation, intravenous, intraperitoneal, subcutaneous, intramuscular, and the like forms, well-known to those of ordinary skill in the pharmaceutical arts.
  • the compounds of the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using trans- dermal delivery systems well-known to those of ordinary skilled in the art.
  • the dosage regimen with the use of the compounds of the present invention is selected by one of ordinary skill in the arts, in view of a variety of factors, including, without limitation, age, weight, sex, and medical condition of the recipient, the severity of the condition to be treated, the route of administration, the level of metabolic and excretory function of the recipient, the dosage form employed, the particular compound and salt thereof employed.
  • the compounds of the present invention are preferably formulated prior to administration together with one or more pharmaceutically-acceptable excipients.
  • Excipients are inert substances such as, without limitation carriers, diluents, flavoring agents, sweeteners, lubricants, solubilizers, suspending agents, binders, tablet disintegrating agents and encapsulating material.
  • compositions of the present invention are pharmaceutical formulation comprising a compound of the invention and one or more pharmaceutically- acceptable excipients that are compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • Pharmaceutical formulations of the invention are prepared by combining a therapeutically effective amount of the compounds of the invention together with one or more pharmaceutically-acceptable excipients therefore.
  • the active ingredient may be mixed with a diluent, or enclosed within a carrier, which may be in the form of a capsule, sachet, paper, or other container.
  • the carrier may serve as a diluent, which may be solid, semi-solid, or liquid material which acts as a vehicle, or can be in the form of tablets, pills powders, lozenges, elixirs, suspensions, emulsions, solutions, syrups, aerosols, ointments, containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions and sterile packaged powders.
  • a diluent which may be solid, semi-solid, or liquid material which acts as a vehicle, or can be in the form of tablets, pills powders, lozenges, elixirs, suspensions, emulsions, solutions, syrups, aerosols, ointments, containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions and sterile packaged powders.
  • the active ingredient may be combined with an oral, and non-toxic, pharmaceutically-acceptable carrier, such as, without limitation, lactose, starch, sucrose, glucose, sodium carbonate, mannitol, sorbitol, calcium carbonate, calcium phosphate, calcium sulfate, methyl cellulose, and the like; together with, optionally, disintegrating agents, such as, without limitation, maize, starch, methyl cellulose, agar, bentonite, xanthan gum, alginic acid, and the like; and optionally, binding agents, for example, without limitation, gelatin, natural sugars, beta-lactose, corn sweeteners, natural and synthetic gums, acacia, tragacanth, sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes, and the like; and, optionally, lubricating agents, for example, without limitation, magnesium stearate, sodium stearate, stearic acid, sodium oleate, sodium benzoate
  • the carrier may be a finely divided solid which is in admixture with the finely divided active ingredient.
  • the active ingredient may be mixed with a carrier having binding properties in suitable proportions and compacted in the shape and size desired to produce tablets.
  • the powders and tablets preferably contain from about 1 to about 99 weight percent of the active ingredient which is the novel composition of the present invention.
  • Suitable solid carriers are magnesium carboxy- methyl cellulose, low melting waxes, and cocoa butter.
  • Sterile liquid formulations include suspensions, emulsions, syrups and elixirs.
  • the active ingredient can be dissolved or suspended in a pharmaceutically acceptable carrier, such as sterile water, sterile organic solvent, or a mixture of both sterile water and sterile organic solvent.
  • a pharmaceutically acceptable carrier such as sterile water, sterile organic solvent, or a mixture of both sterile water and sterile organic solvent.
  • the active ingredient can also be dissolved in a suitable organic solvent, for example, aqueous propylene glycol.
  • Other compositions can be made by dispersing the finely divided active ingredient in aqueous starch or sodium carboxymethyl cellulose solution or in a suitable oil.
  • the formulation may be in unit dosage form, which is a physically discrete unit containing a unit dose, suitable for administration in human or other mammals.
  • a unit dosage form can be a capsule or tablets, or a number of capsules or tablets.
  • An "unit dose" is a predetermined quantity of the active compound of the present invention, calculated to produce the desired therapeutic effect, in association with one or more excipients.
  • the quantity of active ingredient in a unit dose may be varied or adjusted from about 0.1 to about 1000 milligrams or more according to the particular treatment involved.
  • Typical oral dosages of the present invention when used for the indicated effects, will range from about O.Olmg /kg/day to about 100 mg/kg/day, preferably from 0.1 mg/kg/day to 30 mg/kg/day, and most preferably from about 0.5 mg/kg/day to about 10 mg/kg/day.
  • parenteral administration it has generally proven advantageous to administer quantities of about 0.001 to lOOmg /kg/day, preferably from 0.01 mg/kg/day to 1 mg/kg/day.
  • the compounds of the present invention may be administered in a single daily dose, or the total daily dose may be administered in divided doses, two, three, or more times per day. Where delivery is via transdermal forms, of course, administration is continuous.
  • Human vanilloid receptor (hVRl) cDNA was cloned from libraries of axotomized dorsal root ganglia (WO2000/29577). The cloned hVRl cDNA was constructed with pcDNA3 vector and transfected into a CHOluc9aeq cell line. The cell line contains aequorin and CRE-luciferase reporter genes as read-out signals.
  • the transfectants were cloned by limiting dilution in selection medium (DMEM/F12 medium (Gibco BRL) supplemented with 10% FCS, 1.4 mM Sodium pyruvate, 20 mM HEPES, 0.15% Sodium bicarbonate, 100 U/ml penicillin, 100 ⁇ g/ml streptomycin, 2 mM glutamine, non-essential amino acids and 2 mg/ml G418). Ca 2+ influx was examined in the capsaicin-stimulated clones. A high responder clone was selected and used for further experiments in the project.
  • the human VRl-CHOluc9aeq cells were maintained in the selection medium and passaged every 3-4 days at l-2.5xl0 5 cells/flask (75 mm 2 ).
  • Photonics for 60 sec after the stimulation with 10 nM capsaicin. Integral R was calculated and compared with controls.
  • DRG was removed. DRG was incubated with 0.1 % trypsin (Gibco BRL) in PBS(-) (Gibco BRL) for 30 min at 37°C, then a half volume of fetal calf serum (FCS) was added and the cells were spun down. The DRG neuron cells were resuspended in Ham F12/5% FCS/5% horse serum (Gibco BRL) and dispersed by repeated pipetting and passing through 70 ⁇ m mesh (Falcon). The culture plate was incubated for 3 hrs at 37°C to remove contaminating Schwann cells.
  • Gibco BRL 0.1 % trypsin
  • FCS fetal calf serum
  • Non-adherent cells were recovered and further cultured in laminin-coated 384 well plates (Nunc) at lxlO 4 cells/50 ⁇ l/well for 2 days in the presence of 50 ng/ml recombinant rat NGF (Sigma) and 50 ⁇ M 5-fluoro- deoxyuridine (Sigma).
  • DRG neuron cells were washed twice with HBSS supplemented with 17 mM HEPES (pH 7.4) and 0.1% BSA. After incubating with 2 ⁇ M fluo-3AM (Molecular Probe), 0.02% PF127 (Gibco BRL) and 1 mM probenecid (Sigma) for 40 min at 37°C, cells were washed 3 times. The cells were in- cubated with VRl antagonists or vehicle (dimethylsulphoxide) and then with
  • capsaicin vehicle: 80% saline, 10% EtOH, and 10%
  • Tween 80 1 ⁇ M capsaicin (vehicle: 80% saline, 10% EtOH, and 10%) Tween 80).
  • One of the preparations made from the same animal was served as a control while the others were used for evaluating compounds.
  • Ratio of each capsaicin-induced contraction to the internal standard i.e. KCl-induced contraction
  • Human P2X1 -transfected CHOluc9aeq cell line was established and maintained in Dulbecco's modified Eagle's medium (DMEM/F12) supplemented with 7.5% FCS,
  • P2X1 receptor agonist-mediated increases in cytosolic Ca + levels were measured using a fluorescent Ca 2+ chelating dye, Fluo-3 AM (Molecular Probes).
  • the plate- attached cells were washed twice with washing buffer (HBSS, 17 mM HEPES-KOH (pH 7.4), 0.1%) BSA and 0.5 units/ml apyrase), and incubated in 40 ⁇ l of loading buffer (1 ⁇ M Fluo-3 AM, 1 mM probenecid, 1 ⁇ M cyclosporin A, 0.01% pluronic (Molecular Probes)in washing buffer) for 1 hour in a dark place.
  • mice Female Sprague-Dawley rats (200 ⁇ 250 g / Charles River Japan) were used.
  • Rats were anesthetized by intraperitoneal administration of urethane (Sigma) at
  • the bladder catheter was connected via T-tube to a pressure transducer (Viggo- Spectramed Pte Ltd, DT-XXAD) and a microinjection pump (TERUMO). Saline was infused at room temperature into the bladder at a rate of 2.4 ml/hr. Intravesical pressure was recorded continuously on a chart pen recorder (Yokogawa). At least three reproducible micturition cycles, corresponding to a 20-minute period, were recorded before a test compound administration and used as baseline values. (4) Administration of test compounds and stimulation of bladder with capsaicin
  • the saline infusion was stopped before administrating compounds.
  • a testing compound dissolved in the mixture of ethanol, Tween 80 (ICN Biomedicals Inc.) and saline (1 : 1 : 8, v/v/v) was administered intraarterially at 10 mg/kg. 2min after the administration of the compound 10 ⁇ g of capsaicin (Nacalai Tesque) dissolved in ethanol was administered intraarterially.
  • capsaicin-induced intravesical pressure were analyzed from the cystometry data.
  • the capsaicin-induced bladder pressures were compared with the maximum bladder pressure during micturition without the capsaicin stimulation.
  • the testing compounds-mediated inhibition of the increased bladder pressures was evaluated using Student's t-test. A probability level less than 5% was accepted as significant difference.
  • IC 50 A 0.1 ⁇ M ⁇ B 0.5 ⁇ M ⁇ C 1 ⁇ M ⁇ D
  • the compounds of the present invention also show excellent selectivity, and strong activity in other assays (2)-(4) described above .
  • Starting material B was prepared by the same method as for starting material A, using isovanillin instead of vanillin. 6-methoxy-l, 2, 3,4-tetrahydro-7-isoquinolinol (0.03g, 35%). [Starting compound C]
  • Step 1 To a suspension of 3-hydroxy-4-methoxybenzyl alcohol (2.00 g, 13.0 mmol) and K 2 CO 3 (2.13 g, 13.6 mmol) in acetone (80 ml) was added methoxybenz- ylchloride (2.13 g, 13.6 mmol). The reaction mixture was stirred at 60 °C for 16 hrs. The mixture was concentrated under reduced pressure and the residue was dissolved in AcOEt/water. Extraction was carried out with AcOEt and the organic layer was washed with brine, dried over Na 2 SO 4 and then concentrated under reduced pressure to give ⁇ 4-methoxy-3-[(4- methoxybenzyl)oxy]phenyl ⁇ methanol (quantitative yield).
  • Step 2 To a mixture of ⁇ 4-methoxy-3-[(4-methoxybenzyl)oxy]phenyl ⁇ methanol
  • Step 3 To a solution of 4-(azidomethyl)-l-methoxy-2-[(4-methoxybenz- yl)oxy]benzene (1.00 g, 3.3 mmol) in THF (33 ml) was added triphen- ylphosphine (2.63 g, 10.0 mmol) and water (0.25 ml) at room temperature. The reaction mixture was stirred at room temperature for 16 hrs and then concentrated under reduced pressure. The residue 4-(aminomethyl)-l- methoxy-2-[(4-methoxybenzyl)oxy]benzene was used in the reaction with isocyanates following method A without any further purification.
  • Step 1 To a solution of 3-hydroxy-4-methoxybenzaldehyde (3.00 g, 19.7 mmol) and imidazole (1.61 g, 23.7 mmol) in DMF (40 ml) was added t-butyldi- methylsilylchloride (3.12 g, 20.7 mmol) at 0°C. The reaction mixture was stirred at room temperature for 4 hrs and then diluted with diethylether. The organic layer was washed with brine, dried over Na 2 SO 4 and then concentrated under reduced pressure. The residue product was used in the next step without any further purification.
  • Step 2 To a solution of 3- ⁇ [tert-butyl(dimethyl)silyl]oxy ⁇ -4-methoxybenzaldehyde (5.25 g, 19.7 mmol) was added NaBH 4 (0.75 g, 19.7 mmol) and the reaction mixture was stirred at room temperature for 16 hrs. Saturated NH 4 C1 solution was added and the solvent was removed under reduced pressure. The residue was extracted with AcOEt and the organic layer was washed with brine and dried over Na 2 SO 4 and then concentrated under reduced pressure.
  • Step 3 To a mixture of (3- ⁇ [tert-butyl(dimethyl)silyl]oxy ⁇ -4-methoxyphenyl)meth- anol (1.00 g, 3.7 mmol) and l,8-diazabicyclo[5.4.0]undec-7-ene (0.60 g, 3.9 mmol) in toluene (18 ml) was added diphenylphosphinyl azide (1.08 g, 3.9 mmol) at 0°C. The mixture was stirred at room temperature for 4 hrs. Water was added and extraction was carried out with AcOEt. The organic layer was washed with brine, dried over Na SO 4 and concentrated under reduced pressure.
  • Step 4 To a solution of [5-(azidomethyl)-2-methoxyphenoxy](tert-butyl) dimethyl- silane (1.09 g, 3.7 mmol) in AcOEt (20 ml) was added 10% Pd/C (0.10 g) and the reaction mixture was stirred at room temperature for one day under a hydrogen atmosphere. The catalyst was removed by filtration and the filtrate was concentrated under reduced pressure. The residue was washed with diisopropyl ether and hexane to give (3- ⁇ [tert-butyl(dimeth- yl)silyl]oxy ⁇ -4-methoxyphenyl)methanamine which was used in the next step without any further purification.
  • Step 1 To a solution of 3-hydroxybenzonitrile (5.00 g, 42.0 mmol) and N,N-diiso- propylefhylamine (8.14 g, 63.0 mmol) in CH 2 CI 2 (100 ml) was added chlorodimethyl ether (4.06 g, 50.4 mmol) at 0°C. The reaction temperature was allowed to rise to room temperature over 30 minutes. The mixture was then stirred at room temperature for 3 hrs. The mixture was then washed with water, dried over Na 2 SO 4 and then concentrated under reduced pressure. 3-(methoxymethoxy)benzonitrile (4.24 g, 62%) was obtained as clear oil.
  • Step 2 To a cooled (0°C) suspension of lithiumaluminiumhydride (0.84 g,
  • Step 1 A mixture of 7-methoxy-l-tetraline (5.00 g, 28.4 mmol), hydroxylamine hydrochloride (5.92 g, 85.1 mmol) and potassium carbonate 12.94 g, 93.6 mmol) in methanol (100 ml) was heated to reflux and stirred for 16 hrs. The reaction mixture was cooled to room temperature and the solvent was removed under reduced pressure. Water was added to the residue and extraction was carried out with AcOEt. The organic layer was dried over Na 2 SO 4 and then concentrated to give 7-methoxy-3,4-dihydro-l(2H)- naphthalenone oxime (5.51 g, quantitative).
  • Step 2 To a suspension of Pd/C (10%>) in methanol (10 ml) was added a catalytic amount of acetic acid and 7-methoxy-3,4-dihydro-l(2H)-naphthalenone oxime (2.00 g, 10.5 mmol). The mixture was stirred under a hydrogen atmosphere at room temperature for 16 hrs. The Pd catalyst was removed and the filtrate was concentrated under reduced pressure. Water was added to the residue and extraction was carried out with AcOEt. The organic layer was dried over Na SO 4 and then concentrated to give of 7-methoxy-l,2,3,4- tetrahydro-1-naphthalenamine ( 2.00 g, quantitative).
  • Step 3 To a solution of 7-methoxy-l,2,3,4-tetrahydro-l-naphthalenamine (0.20 g,l.l mmol) in CH2CI 2 (5 ml) was added boron tribromide (1.5 ml, 1M solution in CH 2 C1 2 ) at 0°C. Water was then added to the reaction mixture and extraction was carried out with AcOEt. The organic layer was dried over Na 2 SO 4 , concentrated under reduced pressure to give 8-amino- 5,6,7,8-tetrahydro-2-naphthalenol (0.18 g, 98%)
  • Step 1 To a stirred mixture of [Pd(PPh 3 ) 4 ] (0.069 g, 0.06 mmol), K 3 PO 4 (0.636 g, 3.00 mmol) and 4-iodonitrobenzene (0.498 g, 2.00 mmol) in DMF was added phenylboronic acid (0.243 g, 2.00 mmol) and the mixture was stirred at 100 °C for 6 hrs. The reaction mixture was then washed with water and dried over MgSO 4 . The solution was concentrated under reduced pressure and the resulting residue was purified by silica gel column chromatography (5% AcOEt-Hexane) to give 4-nitro-l,l'-biphenyl (0.28 g, 69%).
  • Step 2 To a solution of 4-nitro-l,l'-biphenyl (0.275 g, 1.40 mmol) in ethanol
  • Step 1 To a suspension of 60% sodium hydride in THF/DMF (30 ml, 1 :1) was added dimethyl malonate(2.000 g, 9.57 mmol) at 0 °C. The mixture was allowed to warm to room temperature and stirred for another 30 minutes. 4- fluoro-3 -nitro benzotrifluoride was added and the reaction mixture was stirred for 16 hrs at room temperature. A saturated NH C1 solution was added the mixture was extracted with AcOEt. The organic layer was washed with brine and dried over Na 2 SO 4 .
  • Step 2 A mixture of 2-[2-nitro-4-(trifluoromethyl)phenyl]malonate (1.780 g, 5.55 mmol), LiCl (0.47 g, 11.11 mmol) in DMSO/water (DMSO 10 ml, water 0.10 ml) was heated to 100 °C and stirred for 5 hrs. After cooling to room temperature, AcOEt was added and the solution was washed with brine. The organic layer was washed with brine and dried over Na 2 SO 4 .and then concentrated under reduced pressure. The solution was concentrated under reduced pressure and the resulting residue was triturated with ethyl ether/hexane. Collected to give methyl [2-nitro-4-(trifluoromethyl)phen- yl]acetate (0.546 g, 37%).
  • Step 3 To a solution of methyl [2-nitro-4-(trifluoromethyl)phenyl]acetate (0.546 g, 2.07 mmol) in CH 2 C1 2 (25 ml) was added a 0.9M hexane solution of
  • Step 4 To a solution of 2-[2-nitro-4-(trifluoromethyl)phenyl]ethanol in methanol (20 ml) was added Pd C (0.050 g, 10%). The solution was stirred at room temperature under a hydrogen atmosphere for 20 hrs. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give 2-[2-amino-4-(trifluoromethyl)phenyl]ethanol. The obtained product was used as starting material without any further purification. [Starting compound M]
  • Step 2 A mixture of 2-(5-fluoro-2-nitrophenyl)ethanol (0.123 g, 0.664 mmol), Fe powder (0.300 g, 5.37 mmol) and NH 4 C1 (0.100 g, 1.86 mmol) in EtOH/Water (EtOH 8 ml, water 0.4 ml) was stirred at 90°C for 1 hr. After cooling to room temperature, AcOEt was added and the mixture was filtered through a celite pad. The filtrate was concentrated and the residue was dissolved in AcOEt, washed with water and then brine and dried over MgSO 4 .
  • EtOH 8 ml EtOH/Water
  • This example was performed according to said method A.
  • This example was performed according to the general method A.
  • This example was performed according to said method B.
  • a mixture of 4-(aminomethyl)-2-methoxyphenol hydrochloride ( 50.0 mg, 0.26 mmol) and phenyl 4'-chloro-l,l'-biphenyl-3-ylcarbamate (85.4 mg, 0.26 mmol) in DMSO (0.5 ml) was heated to 90°C and stirred for 16 hrs. Water was then added and extraction was carried out with AcOEt. The organic layer was dried over
  • This example was performed according to the general method B.
  • This example was performed according to the general method C.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Pain & Pain Management (AREA)
  • Urology & Nephrology (AREA)
  • Rheumatology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cardiology (AREA)
  • Vascular Medicine (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Heterocyclic Compounds Containing Sulfur Atoms (AREA)
  • Quinoline Compounds (AREA)
  • Other In-Based Heterocyclic Compounds (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Heterocyclic Compounds That Contain Two Or More Ring Oxygen Atoms (AREA)
  • Furan Compounds (AREA)
  • Pyridine Compounds (AREA)
  • Indole Compounds (AREA)

Abstract

This invention relates to urea derivatives of the formula (I), its tautomeric or stereoisomeric form, or a salt thereof: (I) wherein Y is R1- R6 and X have the same meanings given in the description, which is useful as an active ingredient of pharmaceutical preparations. The urea derivatives of the present invention has an excellent activity as VR1 antagonist and useful for the prophylaxis and treatment of urge urinary incontinence, overactive bladder, chronic pain, neuropathic pain, postoperative pain, rheumatoid arthritic pain, neuralgia, neuropathies, algesia, nerve injury, ischaemia, neurodegeneration, stroke, incontinence and/or inflammatory disorders.

Description

UREA DERIVATIVES
DETAILED DESCRIPTION OF INVENTION
TECHNICAL FIELD
The present invention relates to an urea derivative, which is useful as an active ingredient of pharmaceutical preparations. The urea derivatives of the present invention have vanilloid receptor (VRl) antagonistic activity, and can be used for the prophylaxis and treatment of diseases associated with VRl activity, in particular for the treatment of urge urinary incontinence, overactive bladder, chronic pain, neuropathic pain, postoperative pain, rheumatoid arthritic pain, neuralgia, neuropathies, algesia, nerve injury, ischaemia, neurodegeneration, stroke, incontinence and/or inflammatory disorders.
BACKGROUND ART
Vanilloid compounds are characterized by the presence of vanillyl group or a functionally equivalent group. Examples of several vanilloid compounds or vanilloid receptor modulators are vanillin (4-hydroxy-3-methoxy-benzaldehyde), guaiacol (2- methoxy-phenol), zingerone (4-/4-hydroxy-3-methoxyphenyl/-2-butanon), eugenol(2-methoxy4-/2-propenyl/phenol), and capsaicin (8-methy-N-vanillyl-6- noneneamide).
Among others, capsaicin, the main pungent ingredient in "hot" chili peppers, is a specific neurotoxin that desensitizes C-fiber afferent neurons. Capsaicin interacts with vanilloid receptors (VRl), which are predominantly expressed in cell bodies of dorsal root ganglia (DRG) or nerve endings of afferent sensory fibers including C- fiber nerve endings [Tominaga M, Caterina MJ, Malmberg AB, Rosen TA, Gilbert H, Skinner K, Raumann BE, Basbaum AI, Julius D: The cloned capsaicin receptor integrates multiple pain-producing stimuli. Neuron. 21 : 531-543, 1998]. The VRl receptor was recently cloned [Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD, Julius D: Nature 389: 816-824, (1997)] and identified as a nonselective cation channel with six transmembrane domains that is structurally related to the TRP (transient receptor potential) channel family. Binding of capsaicin to VRl allows sodium, calcium and possibly potassium ions to flow down their concentration gradients, causing initial depolarization and release of neurotrans- mitters from the nerve terminals. VRl can therefore be viewed as a molecular integrator of chemical and physical stimuli that elicit neuronal signals in a pathological conditions or diseases.
There are abundant of direct or indirect evidence that shows the relation between VRl activity and diseases such as pain, ischaemia, and inflammatory (e.g., WO 99/00115 and 00/50387). Further, it has been demonstrated that VRl transduce reflex signals that are involved in the overactive bladder of patients who have damaged or abnormal spinal reflex pathways [De Groat WC: A neurologic basis for the overactive bladder. Urology 50 (6A Suppl): 36-52, 1997]. Desensitisation of the afferent nerves by depleting neurotransmitters using VRl agonists such as capsaicin has been shown to give promising results in the treatment of bladder dysfunction associated with spinal cord injury and multiple sclerosis [(Maggi CA: Therapeutic potential of capsaicin-like molecules - Studies in animals and humans. Life Sciences
51 : 1777-1781, 1992) and (DeRidder D; Chandiramani V; Dasgupta P; VanPoppel H; Baert L; Fowler CJ: Intravesical capsaicin as a treatment for refractory detrusor hyperreflexia: A dual center study with long-term followup. J. Urol. 158: 2087- 2092, 1997)].
It is anticipated that antagonism of the VRl receptor would lead to the blockage of neurotransmitter release, resulting in prophylaxis and treatment of the condition and diseases associated with VRl activity.
It is therefore expected that antagonists of the VRl receptor can be used for prophylaxis and treatment of the condition and diseases including chronic pain, neuropathic pain, postoperative pain, rheumatoid arthritic pain, neuralgia, neuropathies, algesia, nerve injury, ischaemia, neurodegeneration, stroke, incontinence, inflammatory disorders, urge urinary incontinence (UUI), and/or overactive bladder.
WO 2000/50387 discloses the compounds having a vanilloid agonist activity represented by the general formula:
Figure imgf000004_0001
wherein;
X is an oxygen or sulfur atom; Ap is -NHCH2- or -CH2-;
Ra is a substituted or unsubstituted C alkyl group, or RalCO-;
wherein Ral is an alkyl group having 1 to 18 carbon atoms, an alkenyl group having 2 to 18 carbon atoms, or substituted or unsubstituted aryl group having 6 to 10 carbon atoms;
R is a hydrogen atom, an alkyl group having 1 to 6 carbon atoms, an alkoxy group having 1 to 6 carbon atoms, a haloalkyl group having 1 to 6 carbon atoms or a halogen atom;
R is a hydrogen atom, an alkyl group having 1 to 4 carbon atom, an aminoalkyl, a diacid monoester or .-alkyl acid; and the asteric mark * indicates a chiral carbon atom, and their pharmaceutically acceptable salts.
WO 2000/61581 discloses amine derivatives represented by the general formula:
Figure imgf000005_0001
wherein (R\ R") represent (F, F), (CF3, H), or (iPr, iPr)
as useful agents for diabetes, hyperlipemia, arteriosclerosis and cancer.
WO 2000/75106 discloses the compounds represented by the general formula:
Figure imgf000005_0002
wherein Z represents
Figure imgf000006_0001
in which R is hydrogen, CM 2 alkyl, C .8 cycloalkyl, or the like, and R is amino- Cι-6 alkyl, aminocarbonyl-Cι.6 alkyl, or hydroxyaminocarbonyl Cι-6 alkyl; and
R90 and R91 are independently selected from the group consisting of H, Cι_6 alkyl, Ci- β alkylthio, Cι.6 alkoxy, fluoro, chloro, bromo, iodo, and nitro;
as useful agents for treating MMP-mediated diseases in mammals.
However, none of these reference discloses simple urea derivatives having pharmaceutical activity.
The development of a compound having effective VRl antagonistic activity and the use of such compound for the prophylaxis and treatment of diseases associated with
VRl activity, in particular for the treatment of urge urinary incontinence and/or overactive bladder have been desired.
SUMMARY OF THE INVENTION
This invention is to provide urea derivatives of the formula (I), their tautomeric and stereoisomeric form, and salts thereof:
Figure imgf000007_0001
wherein
Y is
Figure imgf000007_0002
X is Cι-6 alkyl substituted by phenyl or naphthyl (wherein said phenyl and naphthyl are optionally substituted by R11, R12 and R13), aryl or heterocyclic ring ,
wherein said aryl and heterocyclic ring are optionally substituted by R1 1, R12 and R13 and are selected from the group consisting of phenyl, naphthyl, pyridyl, carbazolyl, fluorenyl, thienyl, pyrimidyl, benzodioxolyl, indazolyl, and quinolyl,
1 1 1 I T in which R , R and R independently represent hydrogen, halogen, Cι.6 alkyl, mono-, di-, or tri- halogen substituted Cι_6 alkyl, nitro, cyano, Cι_6 alkoxy, hydroxy, piperidino, furyl, thienyl, benzyloxy, anilino, naphthyl, d.6 alkylcarbamoyl, carbamoyl, carboxyl, amino, Cι.6 alkylamino, di(Cι-6 alkyl)amino, Cι.6 alkoxy- carbonyl, benzyl, phenoxy, Cι.6 alkyl substituted phenoxy, pyridyl, halogen substituted phenoxy, Cι.6 alkylthio, Cι_6 alkanoyl, C].6 alkanoylamino, hydroxy substituted Cι-6 alkyl, mono-, di-, or tri- halogen substituted Cι.6 alkyloxy, or phenyl optionally substituted by one to three substituents,
in which the substituents are each different or identical and selected from the group consisting of hydrogen, halogen, Cι.6 alkyl, Cι.6 alkoxy, pyridyl, mono-, di-, or tri- halogen substituted Cι-6 alkyl, nitro, cyano, benzyloxy, thienyl, Cι_6alkanoyl, Cι-6 alkoxycarbonyl, Cι-6 alkylthio, di(Cι_6 alkyl)amino, and Cι.6 alkylamino, mono, di, or tri halogen substituted Cι.6 alkyloxy;
R1 is hydrogen, R2 is hydrogen,
R3 is hydrogen, or
R2 and R3 together form -(CH2)m- (wherein m represents 1, 2, 3 or 4), or R1 and R3 together form -(CH2)n- (wherein n represents 1, 2, or 3);
R4 is hydrogen, halogen, C).6 alkoxy, hydroxy, Cι.6 alkoxy substituted benzyloxy, sulfamoyl, Cι-6 alkylsulfamoyl, di(Cι-6 alkyl)sulfamoyl, di (Cι-6 alkyl)aminoCi_6 alkylene sulfamoyl, hydroxy Cι.6 alkyl piperazinosulfonyl, Cι-6 alkylsulfonylamino, nitro, amino, Cι_6 alkanoylamino, Cι_6 alkoxyCι_6 alkyleneoxy,
R5 is hydrogen, halogen, Cι_6 alkoxy, hydroxy, Cι-6 alkoxy substituted benzyloxy, sulfamoyl, Cι.6 alkylsulfamoyl, di (Cι_6 alkyl)sulfamoyl, di(Cι.6 alkyl)amino Cι-6alkylene sulfamoyl, hydroxy Cι_6 alkyl piperazinosulfonyl,
Cι-6 alkylsulfonylamino, nitro, amino, Cι_6 alkanoylamino,
Figure imgf000008_0001
alkoxyCι-6 alkyleneoxy,
or
R4 and R5 together form -O-(CH2)-O-; and
R6 is hydrogen, halogen, Cι_6 alkyl, mono-, di-, or tri- halogen substituted Cι.6 alkyl, nitro, cyano, Cι_6 alkoxy, hydroxy, Cι.6 alkylcarbamoyl, carbamoyl, carboxyl, amino, Cι_6 alkylamino, di(Cι-6 alkyl) amino, Cι-6 alkoxycarbonyl, phenyl, benzyl, phenoxy, halogen substituted phenoxy, Cι-6 alkylthio, Cι_6 alkanoyl, Cι.6 alkanoylamino, hydroxy substituted Cι-6 alkyl, mono-, di-, or tri- halogen substituted Cι-6 alkoxy.
The urea derivatives of formula (I), their tautomeric and stereoisomeric form, and salts thereof surprisingly show excellent VRl antagonistic activity. They are, therefore suitable especially for the prophylaxis and treatment of diseases associated with VRl activity, in particular for the treatment of urge urinary incontinence and/or overactive bladder.
Alkyl per se and "alk" and "alkyl" in alkoxy, alkanoyl, alkylthio, alkylamino, alkyl- aminocarbonyl, alkylaminosulphonyl, alkylsulphonylamino, alkoxycarbonyl, alkoxy- carbonylamino, alkylcarbamoyl and alkanoylamino represent a linear or branched alkyl radical having generally 1 to 6, preferably 1 to 4 and particularly preferably 1 to 3 carbon atoms, representing illustratively and preferably methyl, ethyl, n-propyl, isopropyl, tert-butyl, n-pentyl and n-hexyl.
Alkoxy illustratively and preferably represents methoxy, ethoxy, n-propoxy, iso- propoxy, tert-butoxy, n-pentoxy and n-hexoxy.
Alkanoyl illustratively and preferably represents acetyl and propanoyl.
Alkylamino represents an alkylamino radical having one or two (independently selected) alkyl substituents, illustratively and preferably representing methylamino, ethylamino, n-propylamino, isopropylamino, tert-butylamino, n-pentylamino, n- hexyl-amino, N,N-dimethylamino, N,N-diethylamino, N-ethyl-N-methylamino, N- methyl-N-n-propylamino, N-isopropyl-N-n-propylamino, N-t-butyl-N-methylamino, N-ethyl-N-n-pentylamino and N-n-hexyl-N-methylamino.
Alkylaminocarbonyl or alkylcarbamoyl represents an alkylaminocarbonyl radical having one or two (independently selected) alkyl substituents, illustratively and preferably representing methylaminocarbonyl, ethylaminocarbonyl, n-propylamino- carbonyl, isopropylamino-carbonyl, tert-butylaminocarbonyl, n-pentylamino- carbonyl, n-hexylaminocarbonyl, N,N-dimethylaminocarbonyl, N,N-diethylamino- carbonyl, N-ethyl-N-methylaminocarbonyl, N-methyl-N-n-propylaminocarbonyl, N- isopropyl-N-n-propylaminocarbonyl, N-t-butyl-N-methylaminocarbonyl, N-ethyl-N- n-pentylamino-carbonyl and N-n-hexyl-N-methylaminocarbonyl.
Alkoxycarbonyl illustratively and preferably represents methoxycarbonyl, ethoxy- carbonyl, n-propoxycarbonyl, isopropoxycarbonyl, tert-butoxycarbonyl, n-pentoxy- carbonyl and n-hexoxycarbonyl. Alkoxycarbonylamino illustratively and preferably represents methoxycarbonylamino, ethoxycarbonylamino, n-propoxycarbonylamino, isopropoxycarbonylamino, tert-butoxycarbonylamino, n-pentoxycarbonylamino and n-hexoxycarbonylamino.
Alkanoylamino illustratively and preferably represents acetylamino and ethyl- carbonylamino.
Halogen represents fluorine, chlorine, bromine and iodine.
Aryl per se and in arylamino and in arylcarbonyl represents a mono- to tricyclic aromatic carbocyclic radical having generally 6 to 14 carbon atoms, and more preferably from 6-10 carbon atoms, optionally substituted with one or more substituents. Examples of aryl radicals include, but are not limited to phenyl, naphthyl, indenyl, azulenyl, fluorenyl, anthracenyl, biphenyl, fluorenonyl and the like.
Heterocyclic ring refers to a 3- to 15-membered ring radical which consists of carbon atoms and from one to five heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. The heterocyclic ring radical may be a monocyclic, bicyclic or tricyclic ring system, which may include fused or bridged ring systems and may be partially or fully saturated or aromatic. Examples of such rings include, but are not limited to thienyl, benzothienyl, furanyl, benzofuranyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, isothiazolyl, thiazolyl, oxazolyl, isoxazolyl, triazolyl, tetrazolyl, imidazolyl, thiadiazolyl, benzothiadiazolyl, oxadiazolyl, benzothiazolyl, indolyl, carbazolyl, quinolinyl, isoquinolinyl, benzodioxolyl, indazolyl, indazolinolyl and the like
This invention is also to provide a method for treating or preventing a disorder or disease associated with VRl activity in a human or animal subject, comprising administering to said subject a therapeutically effective amount of the urea derivative shown in the formula (I), its tautomeric or stereoisomeric form, or a physiologically acceptable salt thereof.
Further this invention is to provide a use of the urea derivative shown in the formula (I), its tautomeric or stereoisomeric form, or a physiologically acceptable salt thereof in the preparation of a medicament. Preferably, said medicament is suitable for treating or preventing a disorder or disease associated with VRl activity.
The compounds of the present invention surprisingly show excellent VRl activity. They are, therefore, suitable for the production of medicament or medical composition, which may be useful to treat VRl related diseases.
More specifically, since the urea derivatives of the present invention inhibit VRl , they are useful for treatment and prophylaxis of diseases as follows:
urinary incontinence, overactive bladder, chronic pain, neuropathic pain, postoperative pain, rheumatoid arthritic pain, neuralgia, neuropathies, algesia, nerve injury, ischaemia, neurodegeneration, stroke, incontinence and inflammatory disorders
In one embodiment, the compounds of formula (I) are those wherein:
Y is I-i; X is phenyl optionally substituted by R11, R12 and R13, phenyl Cι_6 alkyl (wherein said phenyl is optionally substituted by Ru, R12 and R13), or naphthyl optionally substituted by R11, R12 and R13,
in which R1 1, R12 and R13 independently represent hydrogen, halogen, Cι_6 alkyl, mono-, di-, or tri- halogen substituted C].6 alkyl, nitro, Cι.6 alkoxy, Cι-6 alkoxycarbonyl, phenoxy, Cι_6 alkylthio, or Cι-6 alkanoyl.
In another embodiment, the compounds of formula (I) are those wherein:
Y is I-i;
R1 is hydrogen;
R is hydrogen; and
R is hydrogen.
In another embodiment, the compounds of formula (I) are those wherein:
Y is I-i;
X is phenyl optionally substituted by R11, R12 and R13, phenyl Cι.6 alkyl (wherein said phenyl is optionally substituted by R11, R12 and R13), or naphthyl optionally substituted by R1 ' , R12 and R13,
in which R11, R12 and R13 independently represent hydrogen, halogen, Cι.6 alkyl, mono-, di-, or tri- halogen substituted Cι.6 alkyl, nitro, Cι.6 alkoxy, Cι-6 alkoxycarbonyl, phenoxy, Cι-6 alkylthio, or Cι-6 alkanoyl.
R is hydrogen; and R and R together form -(CH2)m- (wherein m represents 1, 2, 3 or 4).
In another embodiment, the compounds of formula (I) are those wherein: Y is I-i;
R1 and R3 together form -(CH2)n- (wherein n represents 1, 2, or 3) and
R is hydrogen.
alternatively, the urea derivative of formula (I) can be those wherein:
Y is I-ii;
R6 is hydrogen, halogen, C].6 alkyl, mono-, di-, or tri- halogen substituted Cι.6 alkyl, phenyl, or Cι-6 alkoxy.
In another embodiment, the compounds of formula (I) are those wherein:
Figure imgf000013_0001
X is Cι-6 alkyl substituted by phenyl or naphthyl (wherein said phenyl and naphthyl are optionally substituted by R11, R12 and R13), aryl or Heterocyclic ring ,
wherein said aryl and Heterocyclic ring are optionally substituted by Rπ, R12 and R13 and are selected from the group consisting of phenyl, naphthyl, pyridyl, carbazolyl, fluorenyl, thienyl, benzodioxolyl, indazolyl, and quinolyl,
R6 is hydrogen, halogen, Cι.6 alkyl, mono-, di-, or tri- halogen substituted C].6 alkyl, phenyl, or Cι-6 alkoxy.
The preferable compounds of the present invention are as follows:
N-(4-hydroxy-3-methoxybenzyl)-N'-(4-isopropylphenyl)urea; N-(4-hydroxy-3 -methoxybenzy l)-N'-( 1 -naphthyl)urea;
N-(3,4-dichlorophenyl)-N'-(4-hydroxy-3-methoxybenzyl)urea;
N-(3-chloro-4-methylphenyl)-N'-(4-hydroxy-3 methoxybenzyl)urea;
N-(4-hydroxy-3-methoxybenzyl)-N'-(4-phenoxyphenyl)urea;
N-[2-chloro-5-(trifluoromethyl)phenyl]-N'-(4-hydroxy-3- methoxybenzyl)urea;
N-(3-chlorophenyl)-N'-(4-hydroxy-3-methoxybenzyl)urea;
N-(4-chlorophenyl)-N'-(4-hydroxy-3-methoxybenzyl)urea;
N-[4-chloro-3-(trifluoromethyl)phenyl]-N'-(4-hydroxy-3- methoxybenzyl)urea; N-(4'-chloro-l,r-biphenyl-3-yl)-N'-(4-hydroxy-3-metoxybenzyl)urea;
N-[2-(2-hydroxyethyl)phenyl]-N'-[4'-(methylsulfanyl)-l,r-biphenyl-3- yl]urea;
N-[2-(2-hydroxyethyl)phenyl]-N'-(4'-nitro-l,l'-biphenyl-3-yl)urea;
N-(4'-acetyl-l,l'-biphenyl-3-yl)-N'-[2-(2-hydroxyethyl)phenyl]urea; Ethyl3'-[({[2-(2hydroxyethyl)phenyl]amino}carbonyl)amino]
-1,1 '-biphenyl-4-carboxylate;
N-[2-(2-hydroxyethyl)phenyl]-N'-[2'-(trifluoromethyl)-l,r-biphenyl-3- yl]urea;
N-(2'-chloro- 1 , 1 '-biphenyl-3 -yl)-N'- [2-(2-hydroxyethyl)phenyl]urea; N-[2-(2-hydroxyethyl)phenyl]-N'-[3-(l -naphthyl)phenyl]urea; N-[2-(2-hydroxyethyl)phenyl]-N'-[4'-(trifluoromethyl)- 1 , 1 '-biphenyl-3- yl]urea;
N-(4',6-dichloro-l,r-biphenyl-3-yl)-N'-[2-(2-hydroxyethyl)phenyl]urea;
N-(2',5'-dichloro-l,l'-biphenyl-3-yl)-N'-[2-(2-hydroxyethyl)phenyl]urea; N-(2',4'-dichloro-l,l'-biphenyl-3-yl)-N'-[2-(2-hydroxyethyl)phenyl]urea;
N-(3',4'-difluoro-l,l'-biphenyl-3-yl)-N'-[2-(2-hydroxyethyl)phenyl]urea;
N-(4'-fluoro- 1 , 1 '-biphenyl-3-yl)-N'-[2-(2-hydroxyethyl)phenyl]urea;
N-[2-(2-hydroxyethyl)phenyl]-N'-(3 '-nitro- l,l'-biphenyl-3-yl)urea;
N-[4'-(benzyloxy)-3'-fluoro-l,r-biphenyl-3-yl]-N,-[2-(2- hydroxyethyl)phenyl]urea;
N-(4'-chloro-l,l'-biphenyl-3-yl)-N'-[2-(2-hydroxyethyl)phenyl]urea;
N-(2',5'-dimethyl-l,l'-biphenyl-3-yl)-N'-[2-(2-hydroxyethyl)phenyl]urea;
N-[2-(2-hydroxyethyl)phenyl]-N'-[4'-(trifluoromethoxy)-l, -biphenyl-3- yljurea; N-(4*-chloro-l,l'-biphenyl-3-yl)-N'-[2-(2-hydroxyethyl)-3- methoxypheny 1] urea;
N-(3'-fluoro-l,l'-biphenyl-3-yl)-N'-[2-(2-hydroxyethyl)phenyl]urea;
N-(3 '-chloro- 1 , 1 '-biphenyl-3 -yl)-N'- [2-(2-hydroxyethyl)phenyl]urea;
N-(2',5'-difluoro-l,l'-biphenyl-3-yl)-N,-[2-(2-hydroxyethyl)ρhenyl]urea; and N-(3'-chloro-4'-fluoro-l,r-biphenyl-3-yl)-N'-[2-(2-hydroxyethyl)phenyl]urea.
Preferably, the medicaments of the present invention further comprise one or more pharmaceutically acceptable carrier and/or excipients.
EMBODIMENT OF THE INVENTION
The compound of the formula (I) of the present invention can be, but not limited to be, prepared by either of the methods [A], [B] and [C] below. In some embodiments, one or more of the substituents, such as amino group, carboxyl group, and hydroxyl group of the compounds used as starting materials or intermediates are ad- vantageously protected by a protecting group known to those skilled in the art. Examples of the protecting groups are described in
"Protective Groups in Organic Synthesis (3rd Edition)" by Greene and Wuts, John Wiley and Sons, New York 1999.
[Method A]
Figure imgf000016_0001
The compound [I-a] wherein X and R6 are the same as defined above, can be prepared by the reaction of a substituted 2-(2-aminophenyl)ethanol[II] (wherein R6 is the same as defined above) and isocyanate of the formula [VI] (wherein X is the same as defined above). The reaction may be carried out in a solvent including, for instance, halogenated hydrocarbons such as dichloromethane, chloroform and 1 ,2-dichloroethane; ethers such as diethyl ether, isopropyl ether, dioxane and tetrahydrofuran (THF) and 1,2- dimethoxyethane; aromatic hydrocarbons such as benzene, toluene and xylene; nitriles such as acetonitrile; amides such as N,N-dimethylformamide (DMF), N,N- dimethylacetamide (DMAC) and N-methylpyrrolidone (NMP); urea such as 1,3- dimethyl-2-imidazolidinone (DMI); sulfoxides such as dimethylsulfoxide (DMSO); and others.
The reaction temperature can be optionally set depending on the compounds to be reacted. The reaction temperature is usually, but not limited to, about 30 °C to 100 °C. The reaction may be conducted for, usually, 30 minutes to 48 hours and preferably 1 to 24 hours.
The compound [I-b], [I-c] and [I-d] wherein X, R4 and R5 are the same as defined above, can be prepared using substituted benzylamines [III], substituted tetrahydro- isoquinolines [IV] and substituted tetrahydro-naphthalenylamine [V] as starting material, respectively, by the same method as for the compound [I-a].
[Method B]
Figure imgf000018_0001
M [VII] [I-d]
Alternatively, the compound [I-a] (wherein X and R6 are the same as defined above) can be prepared by reacting a substituted 2-(2-aminophenyl)ethanol[II] and carbamate of the formula [VII] (wherein X is the same as defined above and Y represents phenyl).
The reaction may be carried out in a solvent including, for instance, halogenated hydrocarbons such as dichloromethane, chloroform and 1,2-dichloroethane; ethers such as diethyl ether, isopropyl ether, dioxane and tetrahydrofuran (THF) and 1,2- dimethoxy ethane; aromatic hydrocarbons such as benzene, toluene and xylene; nitriles such as acetonitrile; amides such as N, N-dimethylformamide (DMF), N, N- dimethylacetamide (DMAC) and N-methylpyrrolidone(NMP); urea such as 1,3- dimethyl-2-imidazolidinone (DMI); sulfoxides such as dimethylsulfoxide (DMSO); and others. The reaction temperature can be optionally set depending on the compounds to be reacted. The reaction temperature is usually, but not limited to, about 20 °C to 100 °C. The reaction may be conducted for, usually, 30 minutes to 40 hours and preferably 1 to 24 hours.
The compound [I-b], [I-c] and [I-d] wherein X, R4 and R5 are the same as defined above, can be prepared using substituted benzylamines [III], substituted tetrahydro- isoquinolines [IV] and substituted tetrahydro-naphthalenylamine [V] as starting material, respectively, by the same method as for the compound [I-a].
[Method C]
Figure imgf000019_0001
[I-d]
The compound [I-a] can be prepared by reacting amine of the formula [VIII] (wherein X is the same as defined above) and l, -carbonyldi(l,2,4-triazole) (CDT)[IX], and then adding substituted 2-(2-aminophenyl)ethanol[II] to the reaction mixture. The reaction may be carried out in a solvent including, for instance, halogenated hydrocarbons such as dichloromethane, chloroform and 1,2-dichloroethane; ethers such as diethyl ether, isopropyl ether, dioxane and tetrahydrofuran (THF) and 1,2- dimethoxyethane; aromatic hydrocarbons such as benzene, toluene and xylene; nitriles such as acetonitrile; amides such as N, N-dimethylformamide (DMF), N, N- dimethylacetamide(DMAC) and N-methylpyrrolidone (NMP); urea such as 1,3- dimethyl-2-imidazolidinone(DMI); sulfoxides such as dimethylsulfoxide(DMSO); and others.
The reaction temperature can be optionally set depending on the compounds to be reacted. The reaction temperature is usually, but not limited to, about 20 °C to 50 °C. The reaction may be conducted for, usually, 30 minutes to 10 hours and preferably 1 to 24 hours.
The compound [I-b], [I-c] and [I-d] wherein X, R4 and R5 are the same as defined above, can be prepared using substituted benzylamines [III], substituted tetrahydro- isoquinolines [IV] and substituted tetrahydro-naphthalenylamine [V] as starting material, respectively, by the same method as for the compound [I-a].
The substituted 2-(2-aminophenyl)ethanols [II], substituted benzylamines [III], substituted tetrahydroisoquinolines [IV], substituted tetrahydro-naphthalenylamine [V], Isocyanates [VI], carbamates [VII], amine [VIII] and CDT [IX] are commercially available or can be prepared by the use of known techniques or by method described in the examples.
When the compound shown by the formula (I) or a salt thereof has tautomeric isomers and/or stereoisomers (e.g., geometrical isomers and conformational isomers), each of their separated isomer and mixtures are also included in the scope of the present invention. When the compound shown by the formula (I) or a salt thereof has an asymmetric carbon in the structure, their optically active compounds and racemic mixtures are also included in the scope of the present invention.
Typical salts of the compound shown by the formula (I) include salts prepared by reaction of the compounds of the present invention with a mineral or organic acid, or an organic or inorganic base. Such salts are known as acid addition and base addition salts, respectively.
Acids to form acid addition salts include inorganic acids such as, without limitation, sulfuric acid, phosphoric acid, hydrochloric acid, hydrobromic acid, hydriodic acid and the like, and organic acids, such as, without limitation, p-toluenesulfonic acid, methanesulfonic acid, oxalic acid, p-bromophenylsulfonic acid, carbonic acid, succinic acid, citric acid, benzoic acid, acetic acid, and the like.
Base addition salts include those derived from inorganic bases, such as, without limitation, ammonium hydroxide, alkaline metal hydroxide, alkaline earth metal hydroxides, carbonates, bicarbonates, and the like, and organic bases, such as, without limitation, ethanolamine, triethylamine, tris(hydroxymethyl)aminomethane, and the like. Examples of inorganic bases include, sodium hydroxide, potassium hydroxide, potassium carbonate, sodium carbonate, sodium bicarbonate, potassium bicarbonate, calcium hydroxide, calcium carbonate, and the like.
The compound of the present invention or a salts thereof, depending on its substituents, may be modified to form lower alkylesters or known other esters; and/or hydrates or other solvates. Those esters, hydrates, and solvates are included in the scope of the present invention.
The compound of the present invention may be administered in oral forms, such as, without limitation normal and enteric coated tablets, capsules, pills, powders, granules, elixirs, tinctures, solution, suspensions, syrups, solid and liquid aerosols and emulsions. They may also be administered in parenteral forms, such as, without limitation, intravenous, intraperitoneal, subcutaneous, intramuscular, and the like forms, well-known to those of ordinary skill in the pharmaceutical arts. The compounds of the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using trans- dermal delivery systems well-known to those of ordinary skilled in the art.
The dosage regimen with the use of the compounds of the present invention is selected by one of ordinary skill in the arts, in view of a variety of factors, including, without limitation, age, weight, sex, and medical condition of the recipient, the severity of the condition to be treated, the route of administration, the level of metabolic and excretory function of the recipient, the dosage form employed, the particular compound and salt thereof employed.
The compounds of the present invention are preferably formulated prior to administration together with one or more pharmaceutically-acceptable excipients. Excipients are inert substances such as, without limitation carriers, diluents, flavoring agents, sweeteners, lubricants, solubilizers, suspending agents, binders, tablet disintegrating agents and encapsulating material.
Yet another embodiment of the present invention is pharmaceutical formulation comprising a compound of the invention and one or more pharmaceutically- acceptable excipients that are compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. Pharmaceutical formulations of the invention are prepared by combining a therapeutically effective amount of the compounds of the invention together with one or more pharmaceutically-acceptable excipients therefore. In making the compositions of the present invention, the active ingredient may be mixed with a diluent, or enclosed within a carrier, which may be in the form of a capsule, sachet, paper, or other container. The carrier may serve as a diluent, which may be solid, semi-solid, or liquid material which acts as a vehicle, or can be in the form of tablets, pills powders, lozenges, elixirs, suspensions, emulsions, solutions, syrups, aerosols, ointments, containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions and sterile packaged powders.
For oral administration, the active ingredient may be combined with an oral, and non-toxic, pharmaceutically-acceptable carrier, such as, without limitation, lactose, starch, sucrose, glucose, sodium carbonate, mannitol, sorbitol, calcium carbonate, calcium phosphate, calcium sulfate, methyl cellulose, and the like; together with, optionally, disintegrating agents, such as, without limitation, maize, starch, methyl cellulose, agar, bentonite, xanthan gum, alginic acid, and the like; and optionally, binding agents, for example, without limitation, gelatin, natural sugars, beta-lactose, corn sweeteners, natural and synthetic gums, acacia, tragacanth, sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes, and the like; and, optionally, lubricating agents, for example, without limitation, magnesium stearate, sodium stearate, stearic acid, sodium oleate, sodium benzoate, sodium acetate, sodium chloride, talc, and the like.
In powder forms, the carrier may be a finely divided solid which is in admixture with the finely divided active ingredient. The active ingredient may be mixed with a carrier having binding properties in suitable proportions and compacted in the shape and size desired to produce tablets. The powders and tablets preferably contain from about 1 to about 99 weight percent of the active ingredient which is the novel composition of the present invention. Suitable solid carriers are magnesium carboxy- methyl cellulose, low melting waxes, and cocoa butter.
Sterile liquid formulations include suspensions, emulsions, syrups and elixirs. The active ingredient can be dissolved or suspended in a pharmaceutically acceptable carrier, such as sterile water, sterile organic solvent, or a mixture of both sterile water and sterile organic solvent. The active ingredient can also be dissolved in a suitable organic solvent, for example, aqueous propylene glycol. Other compositions can be made by dispersing the finely divided active ingredient in aqueous starch or sodium carboxymethyl cellulose solution or in a suitable oil.
The formulation may be in unit dosage form, which is a physically discrete unit containing a unit dose, suitable for administration in human or other mammals. A unit dosage form can be a capsule or tablets, or a number of capsules or tablets. An "unit dose" is a predetermined quantity of the active compound of the present invention, calculated to produce the desired therapeutic effect, in association with one or more excipients. The quantity of active ingredient in a unit dose may be varied or adjusted from about 0.1 to about 1000 milligrams or more according to the particular treatment involved.
Typical oral dosages of the present invention, when used for the indicated effects, will range from about O.Olmg /kg/day to about 100 mg/kg/day, preferably from 0.1 mg/kg/day to 30 mg/kg/day, and most preferably from about 0.5 mg/kg/day to about 10 mg/kg/day. In the case of parenteral administration, it has generally proven advantageous to administer quantities of about 0.001 to lOOmg /kg/day, preferably from 0.01 mg/kg/day to 1 mg/kg/day. The compounds of the present invention may be administered in a single daily dose, or the total daily dose may be administered in divided doses, two, three, or more times per day. Where delivery is via transdermal forms, of course, administration is continuous.
EXAMPLES
The present invention will be described as a form of examples, but they should by no means be construed as defining the metes and bounds of the present invention.
In the examples below, all quantitative data, if not stated otherwise, relate to percentages by weight.
Mass spectra were obtained using electrospray (ES) ionization techniques (micro- mass Platform LC). Melting points are uncorrected. Liquid Chromatography - Mass spectroscopy (LC-MS) data were recorded on a Micromass Platform LC with Shimadzu Phenomenex ODS column(4.6 mm X 30 mm) flushing a mixture of acetonitrile-water (9:1 to 1 :9) at 1 ml/min of the flow rate. TLC was performed on a precoated silica gel plate (Merck silica gel 60 F-254). Silica gel (WAKO-gel C-200 (75-150 μm)) was used for all column chromatography separations. All chemicals were reagent grade and were purchased from Sigma-Aldrich, Wako pure chemical industries, Ltd., Tokyo kasei kogyo co. Ltd., Arch corporation.
The effect of the present compounds were examined by the following assays and pharmacological tests.
[Measurement of capsaicin-induced Ca2+ influx in the human VRl-transfected CHO cell line] (Assay 1)
(1) Establishment of the human VRl-CHOluc9aeq cell line
Human vanilloid receptor (hVRl) cDNA was cloned from libraries of axotomized dorsal root ganglia (WO2000/29577). The cloned hVRl cDNA was constructed with pcDNA3 vector and transfected into a CHOluc9aeq cell line. The cell line contains aequorin and CRE-luciferase reporter genes as read-out signals. The transfectants were cloned by limiting dilution in selection medium (DMEM/F12 medium (Gibco BRL) supplemented with 10% FCS, 1.4 mM Sodium pyruvate, 20 mM HEPES, 0.15% Sodium bicarbonate, 100 U/ml penicillin, 100 μg/ml streptomycin, 2 mM glutamine, non-essential amino acids and 2 mg/ml G418). Ca2+ influx was examined in the capsaicin-stimulated clones. A high responder clone was selected and used for further experiments in the project. The human VRl-CHOluc9aeq cells were maintained in the selection medium and passaged every 3-4 days at l-2.5xl05 cells/flask (75 mm2).
(2) Measurement of Ca2+ influx using FDSS-3000
Human VRl-CHOluc9aeq cells were suspended in a culture medium which is the same as the selection medium except for G418 and seeded at a density of 1,000 cells per well into 384-well plates (black walled clear-base / Nalge Nunc International). Following the culture for 48 hrs the medium was changed to 2 μM Fluo-3 AM (Molecular Probes) and 0.02% Puronic F-127 in assay buffer (Hank's balanced salt solution (HBSS), 17 mM HEPES (pH7.4), 1 mM Probenecid, 0.1 % BSA) and the cells were incubated for 60 min at 25°C. After washing twice with assay buffer the cells were incubated with a test compound or vehicle for 20 min at 25 °C. Mobilization of cytoplasmic Ca2+ was measured by FDSS-3000 (λex=488nm, λem=540nm / Hamamatsu
Photonics) for 60 sec after the stimulation with 10 nM capsaicin. Integral R was calculated and compared with controls.
[Measurement of the capsaicin-induced Ca2+ influx in primary cultured rat dorsal root ganglia neurons] (Assay 2)
( 1 ) Preparation of rat dorsal root ganglia neurons
New born Wister rats (5-11 days) were sacrificed and dorsal root ganglia
(DRG) was removed. DRG was incubated with 0.1 % trypsin (Gibco BRL) in PBS(-) (Gibco BRL) for 30 min at 37°C, then a half volume of fetal calf serum (FCS) was added and the cells were spun down. The DRG neuron cells were resuspended in Ham F12/5% FCS/5% horse serum (Gibco BRL) and dispersed by repeated pipetting and passing through 70 μm mesh (Falcon). The culture plate was incubated for 3 hrs at 37°C to remove contaminating Schwann cells. Non-adherent cells were recovered and further cultured in laminin-coated 384 well plates (Nunc) at lxlO4 cells/50 μl/well for 2 days in the presence of 50 ng/ml recombinant rat NGF (Sigma) and 50 μM 5-fluoro- deoxyuridine (Sigma).
(2) Ca2+ mobilization assay
DRG neuron cells were washed twice with HBSS supplemented with 17 mM HEPES (pH 7.4) and 0.1% BSA. After incubating with 2 μM fluo-3AM (Molecular Probe), 0.02% PF127 (Gibco BRL) and 1 mM probenecid (Sigma) for 40 min at 37°C, cells were washed 3 times. The cells were in- cubated with VRl antagonists or vehicle (dimethylsulphoxide) and then with
1 μM capsaicin in FDSS-6000 (λeχ=480nm, λem=520nm / Hamamatsu Photonics). The fluorescence changes at 480nm were monitored for 2.5 min. Integral R was calculated and compared with controls.
[Organ bath assay to measure the capsaicin-induced bladder contraction] (Assay 3)
Male Wistar rats (10 week old) were anesthetized with ether and sacrificed by dislocating the necks. The whole urinary bladder was excised and placed in oxygenated Modified Krebs-Henseleit solution (pH 7.4) of the following compo- sition (112mM NaCl, 5.9mM KCl, 1.2mM MgCl2, 1.2mM NaH2PO4, 2mM CaCl2,
2.5mM NaHCO , 12mM glucose). Contractile responses of the urinary bladder were studied as described previously [Maggi CA et al: Br.J.Pharmacol. 108: 801-805, 1993]. Isometric tension was recorded under a load of 1 g using longitudinal strips of rat detrusor muscle. Bladder strips were equilibrated for 60 min before each stimulation. Contractile response to 80 mM KCl was determined at 15 min intervals until reproducible responses were obtained. The response to KCl was used as an internal standard to evaluate the maximal response to capsaicin. The effects of the compounds were investigated by incubating the strips with compounds for 30 min prior to the stimulation with 1 μM capsaicin (vehicle: 80% saline, 10% EtOH, and 10%) Tween 80). One of the preparations made from the same animal was served as a control while the others were used for evaluating compounds. Ratio of each capsaicin-induced contraction to the internal standard (i.e. KCl-induced contraction) was calculated and the effects of the test compounds on the capsaicin-induced contraction were evaluated.
[Measurement of Ca2+ influx in the human P2Xl-transfected CHO cell line]
( 1 ) Preparation of the human P2X 1 -transfected CHOluc9aeq cell line
Human P2X1 -transfected CHOluc9aeq cell line was established and maintained in Dulbecco's modified Eagle's medium (DMEM/F12) supplemented with 7.5% FCS,
20 mM HEPES-KOH (pH 7.4), 1.4 mM sodium pyruvate, 100 U/ml penicillin, lOO μg/ml streptomycin, 2 mM glutamine (Gibco BRL) and 0.5 Units/ml apyrase (grade I, Sigma). The suspended cells were seeded in each well of 384-well optical bottom black plates (Nalge Nunc International) at 3 x 103 / 50 μl / well. The cells were cultured for following 48 hrs to adhere to the plates.
(2) Measurement of the intracellular Ca 2+ levels
P2X1 receptor agonist-mediated increases in cytosolic Ca + levels were measured using a fluorescent Ca2+ chelating dye, Fluo-3 AM (Molecular Probes). The plate- attached cells were washed twice with washing buffer (HBSS, 17 mM HEPES-KOH (pH 7.4), 0.1%) BSA and 0.5 units/ml apyrase), and incubated in 40 μl of loading buffer (1 μM Fluo-3 AM, 1 mM probenecid, 1 μM cyclosporin A, 0.01% pluronic (Molecular Probes)in washing buffer) for 1 hour in a dark place. The plates were washed twice with 40 μl washing buffer and 35 μl of washing buffer were added in each well with 5 μl of test compounds or 2 ',J '-o-(2,4,6-trinitrophenyl) adenosine 5'- triphpsphate (Molecular Probes) as a reference. After further incubation for 10 minutes in dark 200 nM α,β-methylene ATP agonist was added to initiate the Ca2+ mobilization. Fluorescence intensity was measured by FDSS-6000 (λex=410nm, λem =510nm / Hamamatsu Photonics) at 250 msec intervals. Integral ratios were calculated from the data and compared with that of a control.
[Measurement of capsaicin-induced bladder contraction in anesthetized rats] (Assay
4)
(1) Animals
Female Sprague-Dawley rats (200~250 g / Charles River Japan) were used.
(2) Catheter implantation
Rats were anesthetized by intraperitoneal administration of urethane (Sigma) at
1.2 g/kg. The abdomen was opened through a midline incision, and a polyethylene catheter (BECTON DICKINSON, PE50) was implanted into the bladder through the dome. In parallel, the inguinal region was incised, and a polyethylene catheter (Hibiki, size 5) filled with 2 IU / ml of heparin (Novo Heparin, Aventis Pharma) in saline (Otsuka) was inserted into a common iliac artery.
(3) Cystometric investigation
The bladder catheter was connected via T-tube to a pressure transducer (Viggo- Spectramed Pte Ltd, DT-XXAD) and a microinjection pump (TERUMO). Saline was infused at room temperature into the bladder at a rate of 2.4 ml/hr. Intravesical pressure was recorded continuously on a chart pen recorder (Yokogawa). At least three reproducible micturition cycles, corresponding to a 20-minute period, were recorded before a test compound administration and used as baseline values. (4) Administration of test compounds and stimulation of bladder with capsaicin
The saline infusion was stopped before administrating compounds. A testing compound dissolved in the mixture of ethanol, Tween 80 (ICN Biomedicals Inc.) and saline (1 : 1 : 8, v/v/v) was administered intraarterially at 10 mg/kg. 2min after the administration of the compound 10 μg of capsaicin (Nacalai Tesque) dissolved in ethanol was administered intraarterially.
(5) Analysis of cystometry parameters
Relative increases in the capsaicin-induced intravesical pressure were analyzed from the cystometry data. The capsaicin-induced bladder pressures were compared with the maximum bladder pressure during micturition without the capsaicin stimulation. The testing compounds-mediated inhibition of the increased bladder pressures was evaluated using Student's t-test. A probability level less than 5% was accepted as significant difference.
Results of IC 0 of capsaicin-induced Ca2+ influx in the human VRl -transfected CHO cell line are shown in Examples and tables of the Examples below. The data corresponds to the compounds as yielded by solid phase synthesis and thus to levels of purity of about 40 to 90%. For practical reasons, the compounds are grouped in four classes of activity as follows:
IC50 = A 0.1μM < B 0.5 μM < C 1 μM < D
The compounds of the present invention also show excellent selectivity, and strong activity in other assays (2)-(4) described above . Preparing method of starting compounds:
[Starting compound A]
7-methoxy-l, 2,3»4-tetrahydro-6-isoquinolinol
Figure imgf000031_0001
An ethanol (15 ml) solution of aminoacetaldehyde diethyl acetal (2.66 g, 20.0 mmol) and vanillin (3.04 g, 20.0 mmol) was added to a suspension of platinum (prepared by reduction of 0.2 g of platinum oxide) in ethanol (20 ml). The mixture was stirred under a hydrogen atmosphere at room temperature for 4 hrs. The catalyst was removed and the solvent was evaporated under reduced pressure. The residue was dissolved in 6N HC1 (150 ml) and Pd/C (2.0g, 10%) was added. The reaction mixture was stirred under a hydrogen atmosphere at room temperature for 16 hrs. The catalyst was removed by filtration and the filtrate was concentrated under reduced pressure. The residue was collected and washed with ethanol to give 7- methoxy-1, 2,3,4-tetrahydro-6-isoquinolinol (0/75 g, 25%>).
[Starting compound B]
6-methoxy-l , 2,3,4-tetrahydro-7-isoquinolinol
Figure imgf000031_0002
Starting material B was prepared by the same method as for starting material A, using isovanillin instead of vanillin. 6-methoxy-l, 2, 3,4-tetrahydro-7-isoquinolinol (0.03g, 35%). [Starting compound C]
7-nitro-l, 2,3»4-tetrahydro-l-naphthalenamine
Figure imgf000032_0001
A mixture of 7-nitro-l -tetralone (1.91 g, 10.0 mmol), titanium (IV)tetraisopropoxide (5.9 ml, 20.0 mmol), ammonium chloride (1.07 g, 20.0 mmol) and triethylamine (2.8 ml, 20.0 mmol) in ethanol (20 ml) was stirred for 16 hrs at room temperature. Sodium tetrahydroborate (0.57 g, 15.0 mmol) was added and the reaction mixture was stirred for another 7 hrs at room temperature. 2M aqueous ammonia (30 ml) was added and after filtration of the inorganic precipitate, extraction was carried out with diethylether. The organic layer was then extracted with 2M HC1. The HC1 solution was washed with diethylether and then treated with 2M NaOH. Extraction with diethylether was carried out. The organic layer was washed with brine, dried over Na2SO4 and then concentrated to give 7-nitro-l, 2, 3, 4-tetrahydro-l-naphthalenamine
(0.25 g, 20%)
[Starting compound D] 4-(aminomethyl)-l-methoxy-2-[(4-methoxybenzyl) oxyjbenzene
Figure imgf000033_0001
Step 1 : To a suspension of 3-hydroxy-4-methoxybenzyl alcohol (2.00 g, 13.0 mmol) and K2CO3 (2.13 g, 13.6 mmol) in acetone (80 ml) was added methoxybenz- ylchloride (2.13 g, 13.6 mmol). The reaction mixture was stirred at 60 °C for 16 hrs. The mixture was concentrated under reduced pressure and the residue was dissolved in AcOEt/water. Extraction was carried out with AcOEt and the organic layer was washed with brine, dried over Na2SO4 and then concentrated under reduced pressure to give {4-methoxy-3-[(4- methoxybenzyl)oxy]phenyl}methanol (quantitative yield).
Step 2: To a mixture of {4-methoxy-3-[(4-methoxybenzyl)oxy]phenyl}methanol
(1.00 g, 3.7 mmol) and l,8-diazabicyclo[5.4.0]undec-7-ene (0.61 g, 4.0 mmol) in toluene (18 ml) was added diphenylphosphinyl azide (1.10 g, 4.0 mmol) at 0°C. The mixture was stirred at room temperature for 4 hrs. Water was added and extraction was carried out with AcOEt. The organic layer was washed with brine, dried over Na2SO4 and concentrated under reduced pressure. The residue was passed through a silica gel plug (hexane: AcOEt = 1 :1) and the filtrate was concentrated under reduced pressure to give 4-(azidomethyl)-l-methoxy-2-[(4-methoxybenz- yl)oxy]benzene (1.00 g, 92%)which was used for the next step without any further purification.
Step 3: To a solution of 4-(azidomethyl)-l-methoxy-2-[(4-methoxybenz- yl)oxy]benzene (1.00 g, 3.3 mmol) in THF (33 ml) was added triphen- ylphosphine (2.63 g, 10.0 mmol) and water (0.25 ml) at room temperature. The reaction mixture was stirred at room temperature for 16 hrs and then concentrated under reduced pressure. The residue 4-(aminomethyl)-l- methoxy-2-[(4-methoxybenzyl)oxy]benzene was used in the reaction with isocyanates following method A without any further purification.
[Starting compound E] (3-{[tert-butyl(dimethyl)silyl]oxy}-4-methoxyphenyl)methanamine
Figure imgf000034_0001
Step 1: To a solution of 3-hydroxy-4-methoxybenzaldehyde (3.00 g, 19.7 mmol) and imidazole (1.61 g, 23.7 mmol) in DMF (40 ml) was added t-butyldi- methylsilylchloride (3.12 g, 20.7 mmol) at 0°C. The reaction mixture was stirred at room temperature for 4 hrs and then diluted with diethylether. The organic layer was washed with brine, dried over Na2SO4 and then concentrated under reduced pressure. The residue product was used in the next step without any further purification.
Step 2: To a solution of 3-{[tert-butyl(dimethyl)silyl]oxy}-4-methoxybenzaldehyde (5.25 g, 19.7 mmol) was added NaBH4 (0.75 g, 19.7 mmol) and the reaction mixture was stirred at room temperature for 16 hrs. Saturated NH4C1 solution was added and the solvent was removed under reduced pressure. The residue was extracted with AcOEt and the organic layer was washed with brine and dried over Na2SO4 and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (Hexane:AcOEt = 9:1 - 3:1) to give (3-{[tert-butyl(dimethyl)silyl]oxy}-4- methoxyphenyl)methanol (4.51 g, 85%).
Step 3: To a mixture of (3-{[tert-butyl(dimethyl)silyl]oxy}-4-methoxyphenyl)meth- anol (1.00 g, 3.7 mmol) and l,8-diazabicyclo[5.4.0]undec-7-ene (0.60 g, 3.9 mmol) in toluene (18 ml) was added diphenylphosphinyl azide (1.08 g, 3.9 mmol) at 0°C. The mixture was stirred at room temperature for 4 hrs. Water was added and extraction was carried out with AcOEt. The organic layer was washed with brine, dried over Na SO4 and concentrated under reduced pressure. The residue was passed through a silica gel plug (hexane: AcOEt = 1 :1) and the filtrate was concentrated under reduced pressure to give [5-(azidomethyl)-2-methoxyphenoxy](tert-butyl)dimeth- ylsilane (1.09 g, quantitative) which was used for the next step without any further purification.
Step 4: To a solution of [5-(azidomethyl)-2-methoxyphenoxy](tert-butyl) dimethyl- silane (1.09 g, 3.7 mmol) in AcOEt (20 ml) was added 10% Pd/C (0.10 g) and the reaction mixture was stirred at room temperature for one day under a hydrogen atmosphere. The catalyst was removed by filtration and the filtrate was concentrated under reduced pressure. The residue was washed with diisopropyl ether and hexane to give (3-{[tert-butyl(dimeth- yl)silyl]oxy}-4-methoxyphenyl)methanamine which was used in the next step without any further purification.
[Starting compound F] [3-(methoxymethoxy)phenyl]methanamine
Figure imgf000036_0001
Step 1 : To a solution of 3-hydroxybenzonitrile (5.00 g, 42.0 mmol) and N,N-diiso- propylefhylamine (8.14 g, 63.0 mmol) in CH2CI2 (100 ml) was added chlorodimethyl ether (4.06 g, 50.4 mmol) at 0°C. The reaction temperature was allowed to rise to room temperature over 30 minutes. The mixture was then stirred at room temperature for 3 hrs. The mixture was then washed with water, dried over Na2SO4 and then concentrated under reduced pressure. 3-(methoxymethoxy)benzonitrile (4.24 g, 62%) was obtained as clear oil.
Step 2: To a cooled (0°C) suspension of lithiumaluminiumhydride (0.84 g,
22.1 mmol) in THF (50 ml) was added drop wise a solution of 3-(meth- oxymethoxy) benzonitrile (3.00 g, 18.4 mmol) in THF (10 ml). The reaction mixture was stirred at 0°C for 1 hr and then at room temperature for 3 hrs. A 5 N NaOH solution was added dropwise at 0°C and the resulting precipitate was filtered off. The filtrate was concentrated under reduced pressure and the residue was dissolved in AcOEt. This was washed with water, dried over Na2SO4, and then concentrated under reduced pressure to give [3-(methoxymethoxy)phenyl]methanamine (1.78 g, 58%). [Starting compound G] 8-amino-5,6,7,8-tetrahydro-2-naphthalenol
Figure imgf000037_0001
Step 1 : A mixture of 7-methoxy-l-tetraline (5.00 g, 28.4 mmol), hydroxylamine hydrochloride (5.92 g, 85.1 mmol) and potassium carbonate 12.94 g, 93.6 mmol) in methanol (100 ml) was heated to reflux and stirred for 16 hrs. The reaction mixture was cooled to room temperature and the solvent was removed under reduced pressure. Water was added to the residue and extraction was carried out with AcOEt. The organic layer was dried over Na2SO4 and then concentrated to give 7-methoxy-3,4-dihydro-l(2H)- naphthalenone oxime (5.51 g, quantitative).
Step 2: To a suspension of Pd/C (10%>) in methanol (10 ml) was added a catalytic amount of acetic acid and 7-methoxy-3,4-dihydro-l(2H)-naphthalenone oxime (2.00 g, 10.5 mmol). The mixture was stirred under a hydrogen atmosphere at room temperature for 16 hrs. The Pd catalyst was removed and the filtrate was concentrated under reduced pressure. Water was added to the residue and extraction was carried out with AcOEt. The organic layer was dried over Na SO4 and then concentrated to give of 7-methoxy-l,2,3,4- tetrahydro-1-naphthalenamine ( 2.00 g, quantitative). Step 3: To a solution of 7-methoxy-l,2,3,4-tetrahydro-l-naphthalenamine (0.20 g,l.l mmol) in CH2CI2 (5 ml) was added boron tribromide (1.5 ml, 1M solution in CH2C12) at 0°C. Water was then added to the reaction mixture and extraction was carried out with AcOEt. The organic layer was dried over Na2SO4, concentrated under reduced pressure to give 8-amino- 5,6,7,8-tetrahydro-2-naphthalenol (0.18 g, 98%)
[Starting compound H] l-(3'-amino-l,l'-biphenyl-4-yl)ethanone
Figure imgf000038_0001
To a stirred solution of 3-bromoaniline (0.344 g, 2.00 mmol) and [Pd(PPh3) ] (0.069 g, 0.06 mmol) in DMF was added a 2N solution of sodium carbonate (1.5 ml). 4-acetylphenylboronic acid (0.656 g, 4.00 mmol) was added and the mixture was stirred at 90 °C for 16 hrs. The reaction mixture was then washed with water and dried over MgSO4. The solution was concentrated under reduced pressure and the resulting residue was purified by preparative thin layer chromatography on silica gel (CHCI3) to give l-(3'-amino-l,l'-biphenyl-4-yl)ethanone (0.25 g, 60 %).
[Starting compound I] 4-amino-l,l'-biphenyl
Figure imgf000039_0001
Step 1: To a stirred mixture of [Pd(PPh3)4] (0.069 g, 0.06 mmol), K3PO4 (0.636 g, 3.00 mmol) and 4-iodonitrobenzene (0.498 g, 2.00 mmol) in DMF was added phenylboronic acid (0.243 g, 2.00 mmol) and the mixture was stirred at 100 °C for 6 hrs. The reaction mixture was then washed with water and dried over MgSO4. The solution was concentrated under reduced pressure and the resulting residue was purified by silica gel column chromatography (5% AcOEt-Hexane) to give 4-nitro-l,l'-biphenyl (0.28 g, 69%).
Step 2: To a solution of 4-nitro-l,l'-biphenyl (0.275 g, 1.40 mmol) in ethanol
(30 ml) was added Pd/C (0.050 g, 10% with 51.5% water) and the mixture was stirred at room temperature under a hydrogen atmosphere for 5 hrs. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure to give 4-amino-l,l'-biphenyl (0.21g, 88%) [Starting compound J] 3'-methoxy-l,l'-biphenyl-3-amine
Figure imgf000040_0001
To a stirred solution of 3-bromoanisole (0.374 g, 2.00 mmol) and [Pd(PPh3)4] (0.069 g, 0.06 mmol) in DMF was added a 2N solution of sodium carbonate (1.5 ml). 3-aminophenylboronic acid (0.548 g, 4.00 mmol) was added and the mixture was stirred at 90 °C for 16 hrs. The reaction mixture was then washed with water and dried over MgSO4. The solution was concentrated under reduced pressure and the resulting residue was purified by preparative thin layer chromatography on silica gel (CHC13, IPE:Hexane = 1:1) to give 3'-methoxy-l,l'-biphenyl-3-amine (0.28 g, 92 %).
[Starting compound K] 3-(2-thienyl)aniline
Figure imgf000040_0002
To a stirred mixture of [Pd(PPh3)4] (0.069 g, 0.06 mmol), K3PO4 (0.636 g, 3.00 mmol) and 2-bromothiophene (0.343 g, 2.00 mmol) in DMF was added 3- nitrophenylboronic acid (0.335 g, 2.00 mmol) and the mixture was stirred at 100 °C for 6 hrs. The reaction mixture was then washed with water and dried over MgSO4.
The solution was concentrated under reduced pressure and the resulting residue was dissolved in ethanol (30 ml). Pd C (0.050 g, 10% with 51.5% water) was added and the reaction mixture was stirred at room temperature under a hydrogen atmosphere for 5 hrs. The reaction mixture was filtered and the filtrate was concentrated to give 3-(2-thienyl)aniline (0.35 g, 86 %). [Starting compound L]
2- [2-amino-4-(trifluoromethyl)phenyl] ethanol
Figure imgf000041_0001
Step 1 : To a suspension of 60% sodium hydride in THF/DMF (30 ml, 1 :1) was added dimethyl malonate(2.000 g, 9.57 mmol) at 0 °C. The mixture was allowed to warm to room temperature and stirred for another 30 minutes. 4- fluoro-3 -nitro benzotrifluoride was added and the reaction mixture was stirred for 16 hrs at room temperature. A saturated NH C1 solution was added the mixture was extracted with AcOEt. The organic layer was washed with brine and dried over Na2SO4. The solution was concentrated under reduced pressure and the resulting residue was purified by silica gel column chromatography (hexane: AcOEt = 7:1-3:1) to give dimethyl 2-[2- nitro-4-(trifluoromethyl)phenyl]malonate (1.784 g, 58%).
Step 2: A mixture of 2-[2-nitro-4-(trifluoromethyl)phenyl]malonate (1.780 g, 5.55 mmol), LiCl (0.47 g, 11.11 mmol) in DMSO/water (DMSO 10 ml, water 0.10 ml) was heated to 100 °C and stirred for 5 hrs. After cooling to room temperature, AcOEt was added and the solution was washed with brine. The organic layer was washed with brine and dried over Na2SO4.and then concentrated under reduced pressure. The solution was concentrated under reduced pressure and the resulting residue was triturated with ethyl ether/hexane. Collected to give methyl [2-nitro-4-(trifluoromethyl)phen- yl]acetate (0.546 g, 37%).
Step 3: To a solution of methyl [2-nitro-4-(trifluoromethyl)phenyl]acetate (0.546 g, 2.07 mmol) in CH2C12 (25 ml) was added a 0.9M hexane solution of
DIBAH (6.90 ml) at -78°C. The reaction temperature was allowed to rise to 0°C and was stirred for 2 hrs. The reaction was then quenched with iPrOH/H O and diluted with AcOEt. Siθ2 was added to the mixture and stirring was continued for another 1 hr. The mixture was passed through a celite pad and the filtrate was concentrated under reduced pressure. The obtained crude residue (0.454 g, 93%>) was used in the next step without any further purification.
Step 4: To a solution of 2-[2-nitro-4-(trifluoromethyl)phenyl]ethanol in methanol (20 ml) was added Pd C (0.050 g, 10%). The solution was stirred at room temperature under a hydrogen atmosphere for 20 hrs. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give 2-[2-amino-4-(trifluoromethyl)phenyl]ethanol. The obtained product was used as starting material without any further purification. [Starting compound M]
2-(5-fluoro-2-aminophenyl)ethanol
Figure imgf000043_0001
Step 1: To a stirred mixture of 5-fluoro-2-nitrotoluene (0.300 g, 1.93 mmol) and paraformaldehyde (0.023 g, 0.77 mmol) in DMSO (3.0 ml) was added sodium phenoxide trihydrate (0.010 g, 0.06 mmol). The reacting mixture was heated to 60°C and stirred for 1 hr. The resulting mixture was diluted with AcOEt and washed with dil. HC1, water and then brine. The organic layer was dried over Na2SO4. The solution was concentrated under reduced pressure and the resulting residue was purified by silica gel column chromatography (hexane:AcOEt = 3:1) to give 2-(5-fluoro-2-nitro- phenyl)ethanol.
Step 2: A mixture of 2-(5-fluoro-2-nitrophenyl)ethanol (0.123 g, 0.664 mmol), Fe powder (0.300 g, 5.37 mmol) and NH4C1 (0.100 g, 1.86 mmol) in EtOH/Water (EtOH 8 ml, water 0.4 ml) was stirred at 90°C for 1 hr. After cooling to room temperature, AcOEt was added and the mixture was filtered through a celite pad. The filtrate was concentrated and the residue was dissolved in AcOEt, washed with water and then brine and dried over MgSO4. The solution was concentrated under reduced pressure and the resulting residue was purified by silica gel column chromatography (hexane:AcOEt = 1 :2) to give 2-(5-fluoro-2-aminophenyl)ethanol. (0.09 g, 87%)
Other starting materials are commercially available or can be prepared according to methods reported in the literature.
Example 1-1; N-(4-hydroxy-3-methoxybenzyl)-N'-(4-isopropylphenyl)urea
Figure imgf000044_0001
This example was performed according to said method A.
To a stirred solution of 4-(aminomethyl)-2-methoxyphenol hydrochloride (50.0 mg, 0.26 mmol) and triethylamine (26.68 mg, 0.26 mmol) in 1,4-dioxane (1.5 ml) was added a solution of 4-isopropylphenyl isocyanate (38.3 mg, 0.24 mmol) in 1,4- dioxane (1.4 mL) at room temperature. The reaction mixture was warmed to 50 °C, and stirred for 20 hrs at the same temperature. The solvent was removed under reduced pressure, and the residue was purified by preparative thin layer chromatography (MeOH:CHC13 = 1:20) to give N-(4-hydroxy-3-methoxybenzyl)- N'-(4-isopropylphenyl)urea (21 mg, 25%).
mp 156 °C;
Molecular weight 314.39
Activity grade:A Example 1-2; N-(3,4-dichlorophenyl)-N'-[2-(2-hydroxyethyl)phenyl]urea
Figure imgf000045_0001
This example was performed according to the general method A.
A solution of 2-(2-aminophenyl)ethanol (30.0 mg, 0.22 mmol) and 3,4-dichloro- phenylisocyanate (41.1 mg, 0.22 mmol) in 1,4-dioxane (2.0 mL) was stirred at 50 °C for 18 hrs. The reaction mixture was cooled to room temperature and diluted with diisopropylether. The precipitate was collected and then washed with 'Pr2O to give N-(3,4-dichlorophenyl)-N'-[2-(2-hydroxyethyl)phenyl]urea (48.9 mg, 69%). mp 188-190 °C; Molecular weight 325.20 Activity grade: A
Example 2-1;
N-(4'-chloro-l,r-biphenyI-3-yl)-N'-(4-hydroxy-3 methoxybenzyl)urea
Figure imgf000045_0002
This example was performed according to said method B. A mixture of 4-(aminomethyl)-2-methoxyphenol hydrochloride ( 50.0 mg, 0.26 mmol) and phenyl 4'-chloro-l,l'-biphenyl-3-ylcarbamate (85.4 mg, 0.26 mmol) in DMSO (0.5 ml) was heated to 90°C and stirred for 16 hrs. Water was then added and extraction was carried out with AcOEt. The organic layer was dried over
Na2SO4 and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (AcOEt:hexane = 2:3) to give N-(4'-chloro-l,l'- biphenyl-3-yl)-N'-(4-hydroxy-3 mefhoxybenzyl)urea (65.0 mg, 64%>) m.p. 153.4°C Molecular weight 382.85
Activity grade:A
Example 2-2; N-[2-(2-hydroxyethyl)phenyl]-N'-[3(trifluoromethoxy)phenyl]urea
Figure imgf000046_0001
This example was performed according to the general method B.
A solution of 2-(2-aminophenyl)ethanol (80.1 mg, 0.58 mmol) and phenyl 3-(tri- fluoromethoxy)phenylcarbamate (165.3 mg, 0.56 mmol) in DMSO (2.0 mL) was stirred at 90 °C for 1 hr. The reaction mixture was cooled to room temperature and diluted with AcOEt. The solution was washed with IN HC1, IN NaOH and brine dried over Na2SO4. The solution was then concentrated under reduced pressure, and the residue was triturated with diisopropylether to give N-[2-(2-hydroxy- ethyl)phenyl]-N'-[3(trifluoromethoxy)phenyl]urea (70.5 mg, 37%).
mp 160-161 °C; Molecular weight 340.30 Activity grade: A
Example 3-1; N-(l,l*-biphenyl-3-yl)-N'-[2-(2-hydroxyethyl)phenyl]urea
Figure imgf000047_0001
This example was performed according to the general method C.
To a solution of l,l'-biphenyl-3 -amine (37.0 mg, 0.22 mmol) in THF (2.0 ml) was added l'-carbonyldi(l,2,4-triazole) (35.9mg, 0.22 mmol). 2-(2-aminophenyl)ethanol (30.0 mg, 0.22 mmol) was added and the mixture was stirred at 55°C for 18 hrs.
After cooling to room temperature, the mixture was diluted with water and ethylalcohol and the resulting precipitate was collected and washed to give N-(l,l'- biphenyl-3-yl)-N'-[2-(2-hydroxyethyl)phenyl]urea (20.8 mg, 29%).
mp 196-198 °C
Molecular weight 332.41 MS (M+H):333 Activity grade: A According to procedures similar to any one of the Examples 1 to 3 above, the following compounds were synthesized and tested.
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001

Claims

(1) Am urea derivative of the formula (I), its tautomeric or stereoisomeric form, or a salt thereof:
Figure imgf000113_0001
wherein Y is
Figure imgf000113_0002
X is Cι-6 alkyl substituted by phenyl or naphthyl (wherein said phenyl and naphthyl are optionally substituted by RA, R12 and R13), aryl or heterocyclic ring ,
wherein said aryl and heterocyclic ring are optionally substituted by R11, R12 and R13 and are are selected from the group consisting of phenyl, naphthyl, pyridyl, carbazolyl, fluorenyl, thienyl, pyrimidyl, benzodioxolyl, indazolyl, and quinolyl,
in which RA 12 and R13 independently represent hydrogen, halogen, Cι-6 alkyl, mono-, di-, or tri- halogen substituted Cι-6 alkyl, nitro, cyano, Ci-β alkoxy, hydroxy, piperidino, furyl, thienyl, benzyloxy, anilino, naphthyl, Cι_6 alkylcarbamoyl, carbamoyl, carboxyl, amino, Cι-6 alkylamino, di(Cι.6 alkyl)amino, C].6 alkoxycarbonyl, benzyl, phenoxy, Cι-6 alkyl substituted phenoxy, pyridyl, halogen substituted phenoxy, Cι_6 alkylthio, Cι_6 alkanoyl, Cι.6 alkanoylamino, hydroxy substituted Cι.6 alkyl, mono-, di-, or tri- halogen substituted G-6 alkyloxy, or phenyl optionally substituted by one to three substituents,
in which the substituents are each different or identical and selected from the group consisting of hydrogen, halogen, Cι_6 alkyl, Cι-6 alkoxy, pyridyl, mono-, di-, or tri- halogen substituted Q-β alkyl, nitro, cyano, benzyloxy, thienyl, G_6alkanoyl, G.6 alkoxycarbonyl, Cι.6 alkylthio, di(G-6 alkyl)amino, and G_6 alkylamino, mono, di, or tri halogen substituted Cι-6 alkyloxy; R1 is hydrogen,
R2 is hydrogen, R is hydrogen, or
R2 and R3 together form ~{CΗ._)m- (wherein m represents 1 , 2, 3 or 4), or
R1 and R3 together form -(CH2)n- (wherein n represents 1, 2, or 3);
R4 is hydrogen, halogen, Cι-6 alkoxy, hydroxy, G.6 alkoxy substituted benzyloxy, sulfamoyl, G-6 alkylsulfamoyl, di(G.6 alkyl)sulfamoyl, di(Cι.6 alkyl)amino G-6 alkylene sulfamoyl, hydroxy Q-6 alkyl piperazinosulfonyl, C].6 alkylsulfonylamino, nitro, amino, Cι-6 alkanoylamino, Cι-6 alkoxyC].6 alkyleneoxy,
R5 is hydrogen, halogen, Cι-6 alkoxy, hydroxy, Q-6 alkoxy substituted benzyloxy, sulfamoyl, Cue alkylsulfamoyl, di(Cι_6 alkyl)sulfamoyl, di(Cι.6 alkyl)amino G-6 alkylene sulfamoyl, hydroxy Q-6 alkyl piperazinosulfonyl, G.6 alkylsulfonylamino, nitro, amino, G-6 alkanoylamino, Cι.6 alkoxyCι.6 alkyleneoxy, or
R4 and R5 together form -O-(CH2)-O-; and
R6 is hydrogen, halogen, Q-6 alkyl, mono-, di-, or tri- halogen substituted Cι-6 alkyl, nitro, cyano, G_6 alkoxy, hydroxy, G.6 alkylcarbamoyl, carbamoyl, carboxyl, amino, Cι-6 alkylamino, di(Cι-6 alkyl)amino, G-6 alkoxycarbonyl, phenyl, benzyl, phenoxy, halogen substituted phenoxy, G-6 alkylthio, G-6 alkanoyl, G-6 alkanoylamino, hydroxy substituted G-6 alkyl, mono-, di-, or tri- halogen substituted G-6 alkoxy.
(2) The urea derivative of the formula (I), its tautomeric or stereoisomeric form, or a salt thereof as claimed in claim 1, wherein
Figure imgf000115_0001
X is phenyl optionally substituted by R11, R12 and R13, phenyl G.6 alkyl (wherein said phenyl is optionally substituted by RA 12 and R13), or naphthyl optionally substituted by R11, R12 and R13,
in which RA, R12 and R13 independently represent hydrogen, halogen, G-6 alkyl, mono-, di-, or tri- halogen substituted G.6 alkyl, nitro, G_ 6 alkoxy, G.6 alkoxycarbonyl, phenoxy, G-6 alkylthio, or G-6 alkanoyl.
(3) The urea derivative of the formula (I), its tautomeric or stereoisomeric form, or a salt thereof as claimed in claim 1 , wherein
Figure imgf000115_0002
R1 is hydrogen;
R is hydrogen; and R is hydrogen.
(4) The urea derivative of the formula (I), its tautomeric or .stereoisomeric form, or a salt thereof as claimed in claim 1, wherein Y is
Figure imgf000116_0001
X is phenyl optionally substituted by RA R12 and R13, phenyl G-6 alkyl (wherein said phenyl is optionally substituted by RA R12 and R13), or naphthyl optionally substituted by R11, R12 and R13, in which Ru, R12 and R13 independently represent hydrogen, halogen, G_6 alkyl, mono-, di-, or tri- halogen substituted G-6 alkyl, nitro, G-6 alkoxy, G-6 alkoxycarbonyl, phenoxy, G-6 alkylthio, or G.6 alkanoyl.
R1 is hydrogen; and
R2 and R3 together form -(CH2)m- (wherein m represents 1, 2, 3 or 4).
(5) The urea derivative of the formula (I), its tautomeric or stereoisomeric form, or a salt thereof as claimed in claim 1 , wherein
Y is
Figure imgf000116_0002
1 "\
R and R together form -(CH2)n- (wherein n represents 1, 2, or 3); and R2 is hydrogen.
(6) The urea derivative of the formula (I), its tautomeric or stereoisomeric form, or a salt thereof as claimed in claim 1 , wherein Y is
R is hydrogen, halogen, G-6 alkyl, mono-, di-, or tri- halogen substituted G-6 alkyl, phenyl or G-6 alkoxy.
(7) The urea derivative of the formula (I), its tautomeric or stereoisomeric form, or a salt thereof as claimed in claim 1 , wherein Y is
Figure imgf000117_0002
X is C ι-6 alkyl substituted by phenyl or naphthyl (wherein said phenyl and naphthyl are optionally substituted by RA, R12 and R13), aryl or heterocyclic ring ,
wherein said aryl and heterocyclic ring are optionally substituted by R11, R12 and R13 and are selected from the group consisting of phenyl, naphthyl, pyridyl, carbazolyl, fluorenyl, thienyl, benzodioxolyl, indazolyl, and quinolyl, in which RA, R12 and R13 independently represent hydrogen, halogen, G-6 alkyl, mono-, di-, or tri- halogen substituted G.6 alkyl, nitro, cyano, G-6 alkoxy, hydroxy, piperidino, furyl, thienyl, benzyloxy, anilino, naphthyl, di( G.6 alkyl)amino, G-6 alkoxycarbonyl, benzyl, phenoxy, G-6 alkyl substituted phenoxy, pyridyl, halogen substituted phenoxy, G-6 alkylthio, G_6 alkanoyl, G-6 alkanoylamino, hydroxy substituted G-6 alkyl, mono-, di-, or tri- halogen substituted G_6 alkyloxy,
or phenyl optionally substituted by one to three substituents, in which the substituents are each different or identical and selected from the group consisting of hydrogen, halogen, G-6 alkyl, G-6 alkoxy, pyridyl, mono-, di-, or tri- halogen substituted G-6 alkyl, nitro, cyano, benzyloxy, thienyl, G-6 alkanoyl, G-6 alkoxycarbonyl, G_6 alkylthio, di(G-6 alkyl)amino, G-6 alkylamino, and mono-, di- or tri- halogen substituted G-6 alkyloxy; and R6 is hydrogen, halogen, G-6 alkyl, mono-, di-, or tri- halogen substituted G-6 alkyl, phenyl or G_6 alkoxy.
(8) The urea derivative of the formula (I), its tautomeric or stereoisomeric form, or a salt thereof as claimed in claim 1, wherein said urea derivative of the formula (I) is selected from the group consisting of:
N-(4-hydroxy-3-methoxybenzyl)-N'-(4-isopropylphenyl)urea; N-(4-hydroxy-3-methoxybenzyl)-N'-(l -naphthyl)urea;
N-(3,4-dichlorophenyl)-N'-(4-hydroxy-3-methoxybenzyl)urea;
N-(3 -chloro-4-methylphenyl)-N'-(4-hydroxy-3 methoxybenzyl)urea;
N-(4-hydroxy-3-methoxybenzyl)-N'-(4-phenoxyphenyl)urea;
N-[2-chloro-5-(trifluoromethyl)phenyl]-N'-(4-hydroxy-3- methoxybenzyl)urea;
N-(3-chlorophenyl)-N'-(4-hydroxy-3-methoxybenzyl)urea; N-(4-chlorophenyl)-N'-(4-hydroxy-3-methoxybenzyl)urea;
N-[4-chloro-3-(trifluoromethyl)phenyl]-N'-(4-hydroxy-3- methoxybenzyl)urea;
N-(4'-chloro- 1 , 1 '-biphenyl-3-yl)-N'-(4-hydroxy-3-metoxybenzyl)urea; N-[2-(2-hydroxyethyl)phenyl]-N'-[4'-(methylsulfanyl)-l , 1 '-biphenyl-3- yljurea;
N-[2-(2-hydroxyethyl)phenyl]-N'-(4'-nitro- 1 , 1 '-biphenyl-3-yl)urea;
N-(4'-acetyl- 1 , 1 '-biphenyl-3-yl)-N'-[2-(2-hydroxyethyl)phenyl]urea;
Ethyl3 '-[( { [2-(2hydroxyethyl)phenyl] amino } carbonyl)amino] -l,l'-biphenyl-4-carboxylate;
N-[2-(2-hydroxyethyl)phenyl]-N'-[2'-(trifluoromethyl)-l,l'-biphenyl-3- yljurea;
N-(2'-chloro-l,r-biphenyl-3-yl)-N'-[2-(2-hydroxyethyl)phenyl]urea;
N-[2-(2-hydroxyethyl)phenyl]-N'-[3-(l-naphthyl)phenyl]urea; N-[2-(2-hydroxyethyl)ρhenyl]-N'-[4,-(trifluoromethyl)-l,l'-biρhenyl-3- yl]urea;
N-(4',6-dichloro- 1 , 1 '-biphenyl-3-yl)-N'-[2-(2-hydroxyethyl)phenyl]urea;
N-(2',5'-dichloro-l,l'-biphenyl-3-yl)-N'-[2-(2-hydroxyethyl)phenyl]urea;
N-(2',4'-dichloro-l ,l'-biρhenyl-3-yl)-N,-[2-(2-hydroxyethyl)phenyl]urea; N-(3',4'-difluoro-l,r-biphenyl-3-yl)-N'-[2-(2-hydroxyethyl)phenyl]urea;
N-(4'-fluoro- 1 , 1 '-biphenyl-3-yl)-N'-[2-(2-hydroxyethyl)phenyl]urea;
N-[2-(2-hydroxyethyl)phenyl]-N'-(3'-nitro- 1 , 1 '-biphenyl-3-yl)urea;
N-[4'-(benzyloxy)-3'-fluoro-l,l'-biphenyl-3-yl]-N'-[2-(2- hydroxyethyl)phenyl]urea; N-(4'-chloro-l,l'-biphenyl-3-yl)-N'-[2-(2-hydroxyethyl)phenyl]urea;
N-(2',5'-dimethyl-l,l'-biphenyl-3-yl)-N'-[2-(2-hydroxyethyl)ρhenyl]urea;
N-[2-(2-hydroxyethyl)phenyl]-N'-[4,-(trifluoromethoxy)-l,r-biphenyl-3- yljurea;
N-(4'-chloro- 1 , 1 '-biphenyl-3 -yl)-N'- [2-(2-hydroxyethyl)-3 -methoxy- phenyl]urea;
N-(3'-fluoro-l,l'-biphenyl-3-yl)-N'-[2-(2-hydroxyethyl)phenyl]urea; N-(3'-chloro-l,l'-biρhenyl-3-yl)-N'-[2-(2-hydroxyethyl)phenyl]urea; N-(2',5 '-difluoro- 1 , 1 '-biphenyl-3 -yl)-N'- [2-(2-hydroxyethyl)phenyl]urea; and N-(3'-chloro-4'-fluoro-l, -biphenyl-3-yl)-N'-[2-(2-hydroxyethyl)phenyl]urea.
(9) An urea derivative of the formula (I), its tautomeric or stereoisomeric form, or a salt thereof as claimed in claims 1 for the treatment and/or prophylaxis of diseases.
(10) A medicament comprising the urea derivative, its tautomeric or stereoiso- meric form, or a physiologically acceptable salt thereof as claimed in claim 1 as an active ingredient.
(11) The medicament as claimed in claim 10, further comprising one or more pharmaceutically acceptable excipients.
(12) The medicament as claimed in claim 10, wherein the urea derivative, its tautomeric or stereoisomeric form, or a physiologically acceptable salt thereof is a VRl antagonist.
(13) The medicament as claimed in claim 10 for treatment and/or prophylaxis of a disease selected from the group consisting of urinary incontinence, overactive bladder, chronic pain, neuropathic pain, postoperative pain, rheumatoid arthritic pain, neuralgia, neuropathies, algesia, nerve injury, ischaemia, neuro- degeneration, stroke, incontinence and inflammatory disorders.
(14) An agent to treat or prevent urological disorder; comprising the urea derivative, its tautomeric or stereoisomeric form, or a physiologically acceptable salt thereof as claimed in claim 1 as an active ingredient.
(15) An agent to treat or prevent of urinary incontinence, overactive bladder, chronic pain, neuropathic pain, postoperative pain, rheumatoid arthritic pain, neuralgia, neuropathies, algesia, nerve injury, ischaemia, neurodegeneration, stroke, incontinence and inflammatory disorders; comprising the urea derivative, its tautomeric or stereoisomeric form, or a physiologically acceptable salt thereof as claimed in claim 1 as an active ingredient.
(16) A method for treating or preventing disorder or disease associated with NR1 activity in a human or animal subject, comprising administering to said subject a therapeutically effective amount of the urea derivative, its tautomeric or stereoisomeric form, or a physiologically acceptable salt thereof as claimed in claim 1.
(17) The method of claim 16, wherein said disorder or disease is a urological disorder or disease.
(18) The method of claim 16, wherein said disorder or disease is selected from the group consisting of urinary incontinence, overactive bladder, chronic pain, neuropathic pain, postoperative pain, rheumatoid arthritic pain, neuralgia, neuropathies, algesia, nerve injury, ischaemia, neurodegeneration, stroke, incontinence and inflammatory disorders.
(19) The method of claim 16, wherein said urea derivative , its tautomeric or stereoisomeric form, or a physiologically acceptable salt thereof is administered with one or more pharmaceutically acceptable excipients.
(20) Use of the urea derivative, its tautomeric or stereoisomeric form, or a physiologically acceptable salt thereof as claimed in claim 1 in the preparation of a medicament.
(21) Use of urea derivative, its tautomeric or stereoisomeric form, or a physiologically acceptable salt thereof as claimed in claim 1 in the preparation of a medicament for treating or preventing disorder or disease associated with VRl activity.
(22) The use of claim 21, wherein said disorder or disease is urological disorder or disease.
(23) The use of claim 21, wherein said disorder or disease is selected from the group consisting of urinary incontinence, overactive bladder, chronic pain, neuropathic pain, postoperative pain, rheumatoid arthritic pain, neuralgia, neuropathies, algesia, nerve injury, ischaemia, neurodegeneration, stroke, incontinence and inflammatory disorders.
(24) The use of claim 21, wherein said urea derivative, its tautomeric or stereoisomeric form, or a physiologically acceptable salt thereof is formulated with one or more pharmaceutically acceptable excipients.
(25) Process for controlling urological disorders in humans and animals by administrating of a VRl antagonisticly effective amount of at least one compound as claimed in claim 1.
PCT/EP2002/014216 2001-12-26 2002-12-13 Urea derivatives as vr1- antagonists WO2003055848A2 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
AU2002358700A AU2002358700A1 (en) 2001-12-26 2002-12-13 Urea derivatives as vr1- antagonists
CA002469967A CA2469967A1 (en) 2001-12-26 2002-12-13 Urea derivatives as vr1- antagonists
US10/499,785 US20050154230A1 (en) 2001-12-26 2002-12-13 Urea derivatives
JP2003556380A JP2005513154A (en) 2001-12-26 2002-12-13 Urea derivatives
EP02793004A EP1461311A2 (en) 2001-12-26 2002-12-13 Urea derivatives as vr1-antagonists

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
JP2001-395032 2001-12-26
JP2001395033A JP2003192660A (en) 2001-12-26 2001-12-26 Urea derivative
JP2001395032A JP2003192659A (en) 2001-12-26 2001-12-26 Phenylurea derivative
JP2001-395033 2001-12-26

Publications (2)

Publication Number Publication Date
WO2003055848A2 true WO2003055848A2 (en) 2003-07-10
WO2003055848A3 WO2003055848A3 (en) 2003-10-23

Family

ID=26625299

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2002/014216 WO2003055848A2 (en) 2001-12-26 2002-12-13 Urea derivatives as vr1- antagonists

Country Status (6)

Country Link
US (1) US20050154230A1 (en)
EP (1) EP1461311A2 (en)
JP (1) JP2005513154A (en)
AU (1) AU2002358700A1 (en)
CA (1) CA2469967A1 (en)
WO (1) WO2003055848A2 (en)

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004110986A1 (en) * 2003-06-12 2004-12-23 Astellas Pharma Inc. Benzamide derivative or salt thereof
WO2005005392A1 (en) * 2003-07-07 2005-01-20 Ionix Pharmaceuticals Limited Azacyclic compounds as inhibitors of sensory neurone specific channels
WO2005040100A1 (en) * 2003-10-15 2005-05-06 Bayer Healthcare Ag Tetrahydro-naphthalene and urea derivatives
FR2870846A1 (en) * 2004-05-25 2005-12-02 Sanofi Synthelabo TETRAHYDROISOQUINOLYLSULFONAMIDE DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC USE
US7015233B2 (en) 2003-06-12 2006-03-21 Abbott Laboratories Fused compounds that inhibit vanilloid subtype 1 (VR1) receptor
WO2007002635A2 (en) 2005-06-27 2007-01-04 Bristol-Myers Squibb Company C-linked cyclic antagonists of p2y1 receptor useful in the treatment of thrombotic conditions
WO2007013811A2 (en) * 2005-07-29 2007-02-01 Quest International Services B.V. N-vanillyl urea derivatives as flavour modulating substances
JP2007523888A (en) * 2003-11-08 2007-08-23 バイエル・ヘルスケア・アクチェンゲゼルシャフト Tetrahydro-quinolinyl urea derivatives
US7432275B2 (en) 2002-12-13 2008-10-07 Neurogen Corporation Carboxylic acid, phosphate or phosphonate substituted quinazolin-4-ylamine analogues as capsaicin receptor modulators
US7550499B2 (en) 2004-05-12 2009-06-23 Bristol-Myers Squibb Company Urea antagonists of P2Y1 receptor useful in the treatment of thrombotic conditions
US7566712B2 (en) 2003-07-16 2009-07-28 Neurogen Corporation Biaryl piperazinyl-pyridine analogues
CN100562513C (en) 2003-06-12 2009-11-25 安斯泰来制药有限公司 Benzamide derivatives or its salt
US7645778B2 (en) 2005-01-19 2010-01-12 Bristol-Myers Squibb Company Heteroaryl compounds as P2Y1 receptor inhibitors
US7714002B2 (en) 2005-06-27 2010-05-11 Bristol-Myers Squibb Company Carbocycle and heterocycle antagonists of P2Y1 receptor useful in the treatment of thrombotic conditions
US7728008B2 (en) 2005-06-27 2010-06-01 Bristol-Myers Squibb Company N-linked heterocyclic antagonists of P2Y1 receptor useful in the treatment of thrombotic conditions
US7728005B2 (en) 2003-10-14 2010-06-01 Ajinomoto Co., Inc. Ether derivative
US7767705B2 (en) 2006-08-25 2010-08-03 Abbott Laboratories Compounds that inhibit TRPV1 and uses thereof
US7816382B2 (en) 2005-06-27 2010-10-19 Bristol-Myers Squibb Company Linear urea mimics antagonists of P2Y1 receptor useful in the treatment of thrombotic condition
US7915261B2 (en) 2005-02-17 2011-03-29 Astellas Pharma, Inc. Pyridyl non-aromatic nitrogen-containing heterocyclic-1-carboxylate compound
US7960569B2 (en) 2006-10-17 2011-06-14 Bristol-Myers Squibb Company Indole antagonists of P2Y1 receptor useful in the treatment of thrombotic conditions
US8030504B2 (en) 2006-12-20 2011-10-04 Abbott Laboratories Antagonists of the TRPV1 receptor and uses thereof
US8110687B2 (en) 2005-12-08 2012-02-07 Millennium Pharmaceuticals, Inc. Bicyclic compounds with kinase inhibitory activity
US8232411B2 (en) 2008-03-20 2012-07-31 Abbott Laboratories Methods for making central nervous system agents that are TRPV1 antagonists
US8664245B2 (en) 2009-06-26 2014-03-04 Sanofi Fumarate salts of a histamine H3 receptor antagonist
US8748615B2 (en) 2010-03-05 2014-06-10 Sanofi Process for the preparation of 2-(cyclohexylmethyl)-N-{2-[(2S)-1-methylpyrrolidin-2-yl]ethyl}-1,2,3,4-tetrahydroisoquinoline-7-sulfonamide
US8759361B2 (en) 2006-08-23 2014-06-24 Neurogen Corporation 2-phenoxy pyrimidinone analogues
WO2015042071A1 (en) * 2013-09-19 2015-03-26 Allergan, Inc. Diphenyl urea derivatives as formyl peptide receptor modulators
US9809544B2 (en) 2014-10-24 2017-11-07 Ono Pharmaceutical Co., Ltd. KCNQ2-5 channel activator
DE102022104759A1 (en) 2022-02-28 2023-08-31 SCi Kontor GmbH Co-crystal screening method, in particular for the production of co-crystals

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5430943B2 (en) * 2006-01-25 2014-03-05 シンタ ファーマシューティカルズ コーポレーション Substituted aromatic compounds for inflammation and immune related applications
US20080153845A1 (en) * 2006-10-27 2008-06-26 Redpoint Bio Corporation Trpv1 antagonists and uses thereof
CN102070525B (en) * 2010-12-24 2013-04-24 中国药科大学 Tetrahydroisoquinoline derivatives and preparation method and application thereof
CN110621656A (en) * 2017-05-12 2019-12-27 研究三角协会 Diaryl ureas as CB1 allosteric modulators
US11919895B2 (en) * 2019-10-25 2024-03-05 Saint Louis University GPR183 antagonists for the treatment of pain

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000050387A1 (en) * 1999-02-22 2000-08-31 Pacific Corporation Vanilloid analogues containing resiniferatoxin pharmacophores as potent vanilloid receptor agonists and analgesics, compositions and uses thereof
WO2002016318A1 (en) * 2000-08-21 2002-02-28 Pacific Corporation Novel thiourea derivatives and the pharmaceutical compositions containing the same
WO2002072536A1 (en) * 2001-03-09 2002-09-19 Smithkline Beecham P.L.C. Urea derivatives having vanilloid receptor (vr1) antagonist activity
WO2003014064A1 (en) * 2001-07-31 2003-02-20 Bayer Healthcare Ag Naphthylurea and naphthylacetamide derivatives as vanilloid receptor 1 (vr1) antagonists

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000050387A1 (en) * 1999-02-22 2000-08-31 Pacific Corporation Vanilloid analogues containing resiniferatoxin pharmacophores as potent vanilloid receptor agonists and analgesics, compositions and uses thereof
WO2002016318A1 (en) * 2000-08-21 2002-02-28 Pacific Corporation Novel thiourea derivatives and the pharmaceutical compositions containing the same
WO2002072536A1 (en) * 2001-03-09 2002-09-19 Smithkline Beecham P.L.C. Urea derivatives having vanilloid receptor (vr1) antagonist activity
WO2003014064A1 (en) * 2001-07-31 2003-02-20 Bayer Healthcare Ag Naphthylurea and naphthylacetamide derivatives as vanilloid receptor 1 (vr1) antagonists

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
DI MARZO V ET AL.: "A Structure/Activity Relationship Study on Arvanil, an Endocannabinoid and Vanilloid Hybrid" THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS, vol. 300, no. 3, 2002, pages 984-991, XP001145825 *
KLOPMAN G & LI J-Y: "Quantitative structure-agonist activity relationship of capsaicin analogues" JOURNAL OF COMPUTER-AIDED MOLECULAR DESIGN, vol. 9, no. 3, 1995, pages 283-294, XP009008828 *

Cited By (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7432275B2 (en) 2002-12-13 2008-10-07 Neurogen Corporation Carboxylic acid, phosphate or phosphonate substituted quinazolin-4-ylamine analogues as capsaicin receptor modulators
WO2004110986A1 (en) * 2003-06-12 2004-12-23 Astellas Pharma Inc. Benzamide derivative or salt thereof
US8487116B2 (en) 2003-06-12 2013-07-16 Abbvie Inc. Fused compounds that inhibit vanilloid receptor subtype 1 (VR1) receptor
US8071762B2 (en) 2003-06-12 2011-12-06 Abbott Laboratories Fused compounds that inhibit vanilloid receptor subtype 1 (VR1) receptor
US7585878B2 (en) 2003-06-12 2009-09-08 Astellas Pharma Inc. Benzamide derivative or salt thereof
US8026256B2 (en) 2003-06-12 2011-09-27 Abbott Laboratories Fused compounds that inhibit vanilloid subtype 1 (VR1) receptor
AU2004247559B2 (en) * 2003-06-12 2009-08-27 Astellas Pharma Inc. Benzamide derivative or salt thereof
US7855198B2 (en) 2003-06-12 2010-12-21 Astellas Pharma Inc. Benzamide derivative or salt thereof
US7015233B2 (en) 2003-06-12 2006-03-21 Abbott Laboratories Fused compounds that inhibit vanilloid subtype 1 (VR1) receptor
CN100562513C (en) 2003-06-12 2009-11-25 安斯泰来制药有限公司 Benzamide derivatives or its salt
WO2005005392A1 (en) * 2003-07-07 2005-01-20 Ionix Pharmaceuticals Limited Azacyclic compounds as inhibitors of sensory neurone specific channels
US7566712B2 (en) 2003-07-16 2009-07-28 Neurogen Corporation Biaryl piperazinyl-pyridine analogues
US7728005B2 (en) 2003-10-14 2010-06-01 Ajinomoto Co., Inc. Ether derivative
JP2007509846A (en) * 2003-10-15 2007-04-19 バイエル・ヘルスケア・アクチェンゲゼルシャフト Tetrahydro-naphthalene and urea derivatives
WO2005040100A1 (en) * 2003-10-15 2005-05-06 Bayer Healthcare Ag Tetrahydro-naphthalene and urea derivatives
JP2007523888A (en) * 2003-11-08 2007-08-23 バイエル・ヘルスケア・アクチェンゲゼルシャフト Tetrahydro-quinolinyl urea derivatives
US7550499B2 (en) 2004-05-12 2009-06-23 Bristol-Myers Squibb Company Urea antagonists of P2Y1 receptor useful in the treatment of thrombotic conditions
US7833999B2 (en) 2004-05-25 2010-11-16 Sanofi-Aventis Tetrahydroisoquinoline sulfonamide derivatives, the preparation thereof, and the use of the same in therapeutics
NO338104B1 (en) * 2004-05-25 2016-08-01 Sanofi Aventis Tetrahydroisoquinoline-sulfonamide derivatives, pharmaceutical compositions containing them and their use as therapeutic agents
US8524700B2 (en) 2004-05-25 2013-09-03 Sanofi Tetrahydroisoquinoline sulfonamide derivatives, the preparation thereof, and the use of the same in therapeutics
FR2870846A1 (en) * 2004-05-25 2005-12-02 Sanofi Synthelabo TETRAHYDROISOQUINOLYLSULFONAMIDE DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC USE
US8273733B2 (en) 2004-05-25 2012-09-25 Sanofi Tetrahydroisoquinoline sulfonamide derivatives, the preparation thereof, and the use of the same in therapeutics
WO2005118547A1 (en) 2004-05-25 2005-12-15 Sanofi-Aventis Tetrahydroisoquinoline sulfonamide derivatives, the preparation thereof, and the use of the same in therapeutics
EA010234B1 (en) * 2004-05-25 2008-06-30 Санофи-Авентис Tetrahydroisoquinoline sulfonamide derivatives, the preparation thereof, and the use of the same in therapeutics
US7645778B2 (en) 2005-01-19 2010-01-12 Bristol-Myers Squibb Company Heteroaryl compounds as P2Y1 receptor inhibitors
US7915261B2 (en) 2005-02-17 2011-03-29 Astellas Pharma, Inc. Pyridyl non-aromatic nitrogen-containing heterocyclic-1-carboxylate compound
US7919495B2 (en) 2005-02-17 2011-04-05 Astellas Pharma, Inc. Pyridyl non-aromatic nitrogen-containing heterocyclic-1-carboxylate compound
US7919494B2 (en) 2005-02-17 2011-04-05 Astellas Pharma, Inc. Pyridyl non-aromatic nitrogen-containing heterocyclic-1-carboxylate compound
WO2007002635A3 (en) * 2005-06-27 2007-03-29 Bristol Myers Squibb Co C-linked cyclic antagonists of p2y1 receptor useful in the treatment of thrombotic conditions
US7816382B2 (en) 2005-06-27 2010-10-19 Bristol-Myers Squibb Company Linear urea mimics antagonists of P2Y1 receptor useful in the treatment of thrombotic condition
US7700620B2 (en) 2005-06-27 2010-04-20 Bristol-Myers Squibb Company C-linked cyclic antagonists of P2Y1 receptor useful in the treatment of thrombotic conditions
WO2007002635A2 (en) 2005-06-27 2007-01-04 Bristol-Myers Squibb Company C-linked cyclic antagonists of p2y1 receptor useful in the treatment of thrombotic conditions
US7714002B2 (en) 2005-06-27 2010-05-11 Bristol-Myers Squibb Company Carbocycle and heterocycle antagonists of P2Y1 receptor useful in the treatment of thrombotic conditions
US7728008B2 (en) 2005-06-27 2010-06-01 Bristol-Myers Squibb Company N-linked heterocyclic antagonists of P2Y1 receptor useful in the treatment of thrombotic conditions
US8329718B2 (en) 2005-06-27 2012-12-11 Bristol-Myers Squibb Company N-linked heterocyclic antagonists of P2Y1 receptor useful in the treatment of thrombotic conditions
WO2007013811A3 (en) * 2005-07-29 2007-05-10 Quest Int Serv Bv N-vanillyl urea derivatives as flavour modulating substances
WO2007013811A2 (en) * 2005-07-29 2007-02-01 Quest International Services B.V. N-vanillyl urea derivatives as flavour modulating substances
US8110687B2 (en) 2005-12-08 2012-02-07 Millennium Pharmaceuticals, Inc. Bicyclic compounds with kinase inhibitory activity
US8759361B2 (en) 2006-08-23 2014-06-24 Neurogen Corporation 2-phenoxy pyrimidinone analogues
US7767705B2 (en) 2006-08-25 2010-08-03 Abbott Laboratories Compounds that inhibit TRPV1 and uses thereof
US8815930B2 (en) 2006-08-25 2014-08-26 Abbvie Inc. Compounds that inhibit TRPV1 and uses thereof
US7960569B2 (en) 2006-10-17 2011-06-14 Bristol-Myers Squibb Company Indole antagonists of P2Y1 receptor useful in the treatment of thrombotic conditions
US8030504B2 (en) 2006-12-20 2011-10-04 Abbott Laboratories Antagonists of the TRPV1 receptor and uses thereof
US8350083B2 (en) 2006-12-20 2013-01-08 Abbvie Inc. Antagonists of the TRPV1 receptor and uses thereof
US8232411B2 (en) 2008-03-20 2012-07-31 Abbott Laboratories Methods for making central nervous system agents that are TRPV1 antagonists
US8664245B2 (en) 2009-06-26 2014-03-04 Sanofi Fumarate salts of a histamine H3 receptor antagonist
US8748615B2 (en) 2010-03-05 2014-06-10 Sanofi Process for the preparation of 2-(cyclohexylmethyl)-N-{2-[(2S)-1-methylpyrrolidin-2-yl]ethyl}-1,2,3,4-tetrahydroisoquinoline-7-sulfonamide
US8779145B2 (en) 2010-03-05 2014-07-15 Sanofi Process for the preparation of 2-(cyclohexylmethyl)-N-{2-[(2S)-1-methylpyrrolidin-2-yl]ethyl}-1,2,3,4-tetrahydroisoquinoline
WO2015042071A1 (en) * 2013-09-19 2015-03-26 Allergan, Inc. Diphenyl urea derivatives as formyl peptide receptor modulators
US9809544B2 (en) 2014-10-24 2017-11-07 Ono Pharmaceutical Co., Ltd. KCNQ2-5 channel activator
US10196358B2 (en) 2014-10-24 2019-02-05 Ono Pharmaceutical Co., Ltd. KCNQ2-5 channel activator
US10676438B2 (en) 2014-10-24 2020-06-09 Ono Pharmaceutical Co., Ltd. KCNQ2-5 channel activator
DE102022104759A1 (en) 2022-02-28 2023-08-31 SCi Kontor GmbH Co-crystal screening method, in particular for the production of co-crystals

Also Published As

Publication number Publication date
JP2005513154A (en) 2005-05-12
CA2469967A1 (en) 2003-07-10
US20050154230A1 (en) 2005-07-14
AU2002358700A8 (en) 2003-07-15
WO2003055848A3 (en) 2003-10-23
EP1461311A2 (en) 2004-09-29
AU2002358700A1 (en) 2003-07-15

Similar Documents

Publication Publication Date Title
WO2003055848A2 (en) Urea derivatives as vr1- antagonists
US20050119304A1 (en) Urea derivatives
EP1414788A1 (en) Naphthylurea and naphthylacetamide derivatives as vanilloid receptor 1 (vr1) antagonists
US20090209514A1 (en) Tetrahydro-Naphthalene Derivatives as Vanilloid Receptor Antagonists
US20100137361A1 (en) Tetrahydro-Quinolinylurea Derivatives
EP1506167B1 (en) Hydroxy tetrahydro-naphthalenylurea derivatives
EP1687262B1 (en) Bicyclic amide, carbamate or urea derivatives as vanilloid receptor modulators
JP2003192659A (en) Phenylurea derivative
JP2003192660A (en) Urea derivative
EP1594836B1 (en) Hydroxy-tetrahydro-naphthalenylurea derivatives
KR101000688B1 (en) Hydroxy Tetrahydro-Naphthalenylurea Derivatives

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

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

AL Designated countries for regional patents

Kind code of ref document: A2

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

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

Ref document number: 2002793004

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2469967

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2003556380

Country of ref document: JP

WWP Wipo information: published in national office

Ref document number: 2002793004

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 10499785

Country of ref document: US