US20090253727A1 - Nociceptin analogs - Google Patents

Nociceptin analogs Download PDF

Info

Publication number
US20090253727A1
US20090253727A1 US12/485,612 US48561209A US2009253727A1 US 20090253727 A1 US20090253727 A1 US 20090253727A1 US 48561209 A US48561209 A US 48561209A US 2009253727 A1 US2009253727 A1 US 2009253727A1
Authority
US
United States
Prior art keywords
alkyl
group
piperidinyl
cycloalkyl
tetrahydro
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/485,612
Inventor
R. Richard Goehring
Zhengming Chen
John Whitehead
Parviz Gharagozloo
Sam Victory
Donald Kyle
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Purdue Pharma LP
Original Assignee
Purdue Pharma LP
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Purdue Pharma LP filed Critical Purdue Pharma LP
Priority to US12/485,612 priority Critical patent/US20090253727A1/en
Publication of US20090253727A1 publication Critical patent/US20090253727A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond

Definitions

  • Chronic pain is a major contributor to disability and is the cause of an untold amount of suffering.
  • the successful treatment of severe and chronic pain is a primary goal of the physician with opioid analgesics being preferred drugs.
  • This ligand is a seventeen amino acid peptide structurally similar to members of the opioid peptide family.
  • ORL1 receptor presents an opportunity in drug discovery for novel compounds which can be administered for pain management or other syndromes modulated by this receptor.
  • the compounds of the present invention are useful for modulating a pharmacodynamic response from one or more opioid receptors (ORL-1, ⁇ , ⁇ and ⁇ ) centrally and/or peripherally.
  • the response can be attributed to the compound stimulating (agonist) or inhibiting (antagonist) the one or more receptors.
  • Certain compounds can stimulate one receptor (e.g., a 1 agonist) and inhibit a different receptor (e.g., an ORL-1 antagonist).
  • the present invention in certain embodiments comprises compounds having the general formula (I):
  • W is hydrogen, C 1-10 alkyl, C 3-12 cycloalkyl, C 3-12 cycloalkylC 1-4 alkyl-, C 1-10 alkoxy, C 3-12 cycloalkoxy-, C 1-10 alkyl substituted with 1-3 halogen, C 3-12 cycloalkyl substituted with 1-3 halogen, C 3-12 cycloalkylC 1-4 alkyl-substituted with 1-3 halogen, C 1-10 alkoxy substituted with 1-3 halogen, C 3-12 cycloalkoxy-substituted with 1-3 halogen, —COOV 1 , —C 1-4 COOV 1 , —CH 2 OH, —SO 2 N(V 1 ) 2 , hydroxyC 3-10 alkyl-, hydroxyC 3-10 cycloalkyl-, cyanoC 1-10 alkyl-, cyanoC 3-10 cycloalkyl-, —CON(V 1 ) 2 , NH 2
  • each V 1 is independently selected from H, C 1-6 alkyl, C 3-6 cycloalkyl, benzyl and phenyl;
  • Q is a 5-8 membered cycloalkyl, 5-8 membered heterocyclic or a 6 membered aromatic or heteroaromatic group
  • each n is independently an integer from 0 to 3;
  • A, B and C are independently hydrogen, C 1-10 alkyl, C 3-12 cycloalkyl, C 1-10 alkoxy, C 3-12 cycloalkoxy, —CH 2 OH, —NHSO 2 , hydroxyC 1-10 alkyl-, aminocarbonyl-, C 1-4 alkylaminocarbonyl-, diC 1-4 alkylaminocarbonyl-, acylamino-, acylaminoalkyl-, amide, sulfonylaminoC 1-10 alkyl-, or A-B can together form a C 2-6 bridge, or B-C can together form a C 3-7 bridge, or A-C can together form a C 1-5 bridge;
  • Z is selected from the group consisting of a bond, straight or branched C 1-6 alkylene, —NH—, —CH 2 O—, —CH 2 NH—, —CH 2 N(CH 3 )—, —NHCH 2 —, —CH 2 CONH—, —NHCH 2 CO—, —CH 2 CO—, —COCH 2 —, —CH 2 COCH 2 —, —CH(CH 3 )—, —CH ⁇ , —O— and —HC ⁇ CH—, wherein the carbon and/or nitrogen atoms are unsubstituted or substituted with one or more lower alkyl, hydroxy, halo or alkoxy group;
  • R 1 is selected from the group consisting of hydrogen, C 1-10 alkyl, C 3-12 cycloalkyl, C 2-10 alkenyl, amino, C 1-10 alkylamino-, C 3-12 cycloalkylamino-, —COOV 1 , —C 1-4 COOV 1 , cyano, cyanoC 1-10 alkyl-, cyanoC 3-10 cycloalkyl-, NH 2 SO 2 —, NH 2 SO 2 C 1-4 alkyl-, NH 2 SOC 1-4 alkyl-, aminocarbonyl-, C 1-4 alkylaminocarbonyl-, diC 1-4 alkylaminocarbonyl-, benzyl, C 3-12 cycloalkenyl-, a monocyclic, bicyclic or tricyclic aryl or heteroaryl ring, a hetero-monocyclic ring, a hetero-bicyclic ring system, and a spiro ring system of the formula (III):
  • X 1 and X 2 are independently selected from the group consisting of NH, O, S and CH 2 ; and wherein said alkyl, cycloalkyl, alkenyl, C 1-10 alkylamino-, C 3-12 cycloalkylamino-, or benzyl of R 1 is optionally substituted with 1-3 substituents selected from the group consisting of halogen, hydroxy, C 1-10 alkyl, C 1-10 alkoxy, nitro, trifluoromethyl-, cyano, —COOV 1 , —C 1-4 COOV 1 , cyanoC 1-10 alkyl-, —C 1-5 ( ⁇ O)W 1 , —C 1-5 NHS( ⁇ O) 2 W 1 , —C 1-5 NHS( ⁇ O)W 1 , a 5-membered heteroaromaticC 0-4 alkyl-, phenyl, benzyl, benzyloxy, said phenyl, benzyl, and benzyloxy
  • R 2 is selected from the group consisting of hydrogen, C 1-10 alkyl, C 3-12 cycloalkyl- and halogen, said alkyl or cycloalkyl optionally substituted with an oxo, amino, alkylamino or dialkylamino group;
  • the present invention in certain embodiments comprises compounds having the formula (IA):
  • each n is independently an integer from 0 to 3;
  • Z is selected from the group consisting of a bond, —CH 2 —, —NH—, —CH 2 O—, —CH 2 CH 2 —, —CH 2 NH—, —CH 2 N(CH 3 )—, —NHCH 2 —, —CH 2 CONH—, —NHCH 2 CO—, —CH 2 CO—, —COCH 2 —, —CH 2 COCH 2 —, —CH(CH 3 )—, —CH ⁇ , and —HC ⁇ CH—, wherein the carbon and/or nitrogen atoms are unsubstituted or substituted with a lower alkyl, halogen, hydroxy or alkoxy group;
  • R 1 is selected from the group consisting of hydrogen, C 1-10 alkyl, C 3-12 cycloalkyl, C 2-10 alkenyl, amino, C 1-10 allylamino, C 3-12 cycloalkylamino, benzyl, C 3-12 cycloalkenyl, a monocyclic, bicyclic or tricyclic aryl or heteroaryl ring, a hetero-monocyclic ring, a hetero-bicyclic ring system, and a spiro ring system of the formula (III):
  • X 1 and X 2 are independently selected from the group consisting of NH, O, S and CH 2 ;
  • said monocyclic aryl is preferably phenyl
  • bicyclic aryl is preferably naphthyl
  • alkyl, cycloalkyl, alkenyl, C 1-10 alkylamino, C 3-12 cycloalkylamino, or benzyl is optionally substituted with 1-3 substituents selected from the group consisting of halogen, C 1-10 alkyl, C 1-10 alkoxy, nitro, trifluoromethyl, cyano, phenyl, benzyl, benzyloxy, said phenyl, benzyl, and benzyloxy optionally being substituted with 1-3 substituents selected from the group consisting of halogen, C 1-10 alkyl, C 1-10 alkoxy, and cyano;
  • C 3-12 cycloalkyl, C 3-12 cycloalkenyl, monocyclic, bicyclic or tricyclic aryl, heteroaryl ring, hetero-monocyclic ring, hetero-bicyclic ring system, and spiro ring system of the formula (III) are optionally substituted with 1-3 substituents selected from the group consisting of halogen, C 1-10 alkyl, C 1-10 alkoxy, nitro, trifluoromethyl, phenyl, benzyl, phenyloxy and benzyloxy, wherein said phenyl, benzyl, phenyloxy and benzyloxy are optionally substituted with 1-3 substituents selected from the group consisting of halogen, C 1-10 alkyl, C 1-10 alkoxy, and cyano;
  • R 2 is selected from the group consisting of hydrogen, C 1-10 alkyl, C 3-12 cycloalkyl and halogen, said alkyl optionally substituted with an oxo group;
  • the present invention in certain embodiments comprises compounds having the general formula (II):
  • W is hydrogen, C 1-10 alkyl, C 3-12 cycloalkyl, C 3-12 cycloalkylC 1-4 alkyl-, C 1-10 alkoxy, C 3-12 cycloalkoxy-, C 1-10 alkyl substituted with 1-3 halogen, C 3-12 cycloalkyl substituted with 1-3 halogen, C 3-12 cycloalkylC 1-4 alkyl-substituted with 1-3 halogen, C 1-10 alkoxy substituted with 1-3 halogen, C 3-12 cycloalkoxy-substituted with 1-3 halogen, —COOV 1 , —C 1-4 COOV 1 , —CH 2 OH, —SO 2 N(V 1 ) 2 , hydroxyC 1-10 alkyl-, hydroxyC 3-10 cycloalkyl-, cyanoC 1-10 alkyl-, cyanoC 3-10 cycloalkyl-, —CON(V 1 ) 2 , NH 2
  • each V 1 is independently selected from H, C 1-6 alkyl, C 3-6 cycloalkyl, benzyl and phenyl;
  • Q is a 5-8 membered cycloalkyl, 5-8 membered heterocyclic or a 6 membered aromatic or heteroaromatic group
  • n is an integer from 0 to 3;
  • A, B and C are independently hydrogen, C 1-10 allyl, C 3-12 cycloalkyl, C 1-10 alkoxy, C 3-12 cycloalkoxy, —CH 2 OH, —NHSO 2 , hydroxyC 1-10 alkyl-, aminocarbonyl-, C 1-4 alkylaminocarbonyl-, diC 1-4 alkylaminocarbonyl-, acylamino-, acylaminoalkyl-, amide, sulfonylaminoC 1-10 alkyl-, or A-B can together form a C 2-6 bridge, or B-C can together form a C 3-7 bridge, or A-C can together form a C 1-5 bridge;
  • Z is selected from the group consisting of a bond, straight or branched C 1-6 alkylene, —NH—, —CH 2 O—, —CH 2 NH—, —CH 2 N(CH 3 )—, —NHCH 2 —, —CH 2 CONH—, —NHCH 2 CO—, —CH 2 CO—, —COCH 2 —, —CH 2 COCH 2 —, —CH(CH 3 )—, —CH ⁇ , —O— and —HC ⁇ CH—, wherein the carbon and/or nitrogen atoms are unsubstituted or substituted with one or more lower alkyl, hydroxy, halo or alkoxy group;
  • R 1 is selected from the group consisting of hydrogen, C 1-10 alkyl, C 3-12 cycloalkyl, C 2-10 alkenyl, amino, C 1-10 alkylamino-, C 3-12 cycloalkylamino-, —COOV 1 , —C 1-4 COOV 1 , cyano, cyanoC 1-10 alkyl-, cyanoC 3-10 cycloalkyl-, NH 2 SO 2 —, NH 2 SO 2 C 1-4 allyl-, NH 2 SOC 1-4 alkyl-, aminocarbonyl-, C 1-4 alkylaminocarbonyl-, diC 1-4 alkylaminocarbonyl-, benzyl, C 3-12 cycloalkenyl-, a monocyclic, bicyclic or tricyclic aryl or heteroaryl ring, a hetero-monocyclic ring, a hetero-bicyclic ring system, and a spiro ring system of the formula (III):
  • X 1 and X 2 are independently selected from the group consisting of NH, O, S and CH 2 ; and wherein said alkyl, cycloalkyl, alkenyl, C 1-10 alkylamino-, C 3-12 cycloalkylamino-, or benzyl of R 1 is optionally substituted with 1-3 substituents selected from the group consisting of halogen, hydroxy, C 1-10 alkyl, C 1-10 alkoxy, nitro, trifluoromethyl-, cyano, —COOV 1 , —C 1-4 COOV 1 , cyanoC 1-10 alkyl-, —C 1-5 ( ⁇ O)W 1 , —C 1-5 NHS( ⁇ O) 2 W 1 , —C 1-5 NHS( ⁇ O)W 1 , a 5-membered heteroaromaticC 0-4 alkyl-, phenyl, benzyl, benzyloxy, said phenyl, benzyl, and benzyloxy
  • R 2 is selected from the group consisting of hydrogen, C 1-10 alkyl, C 3-12 cycloalkyl- and halogen, said alkyl or cycloalkyl optionally substituted with an oxo, amino, alkylamino or dialkylamino group;
  • the present invention in certain embodiments comprises compounds having the formula (IIA) as follows:
  • n is an integer from 0 to 3;
  • Z is selected from the group consisting of a bond, —CH 2 —, —NH—, —CH 2 O—, —CH 2 CH 2 —, —CH 2 NH—, —CH 2 N(CH 3 )—, —NHCH 2 —, —CH 2 CONH—, —NHCH 2 CO—, —CH 2 CO—, —COCH 2 —, —CH 2 COCH 2 —, —CH(CH 3 )—, —CH ⁇ , and —HC ⁇ CH—, wherein the carbon and/or nitrogen atoms are unsubstituted or substituted with a lower alkyl, halogen, hydroxy or alkoxy group;
  • R 1 is selected from the group consisting of hydrogen, C 1-10 alkyl, C 3-12 cycloalkyl, C 2-10 alkenyl, amino, C 1-10 alkylamino, C 3-12 cycloalkylamino, benzyl, C 3-12 cycloalkenyl, a monocyclic, bicyclic or tricyclic aryl or heteroaryl ring, a hetero-monocyclic ring, a hetero-bicyclic ring system, and a spiro ring system of the formula (III):
  • X 1 and X 2 are independently selected from the group consisting of NH, O, S and CH 2 ;
  • said monocyclic aryl is preferably phenyl
  • bicyclic aryl is preferably naphthyl
  • alkyl, cycloalkyl, alkenyl, C 1-10 alkylamino, C 3-12 cycloalkylamino, or benzyl is optionally substituted with 1-3 substituents selected from the group consisting of halogen, C 1-10 alkyl, C 1-10 alkoxy, nitro, trifluoromethyl, cyano, phenyl, benzyl, benzyloxy, said phenyl, benzyl, and benzyloxy optionally being substituted with 1-3 substituents selected from the group consisting of halogen, C 1-10 alkyl, C 1-10 alkoxy, and cyano;
  • C 3-12 cycloalkyl, C 3-12 cycloalkenyl, monocyclic, bicyclic or tricyclic aryl, heteroaryl ring, hetero-monocyclic ring, hetero-bicyclic ring system, and spiro ring system of the formula (III) are optionally substituted with 1-3 substituents selected from the group consisting of halogen, C 1-10 alkyl, C 1-10 alkoxy, nitro, trifluoromethyl, phenyl, benzyl, phenyloxy and benzyloxy, wherein said phenyl, benzyl, phenyloxy and benzyloxy are optionally substituted with 1-3 substituents selected from the group consisting of halogen, C 1-10 alkyl, C 1-10 alkoxy, and cyano;
  • R 2 is selected from the group consisting of hydrogen, C 1-10 alkyl, C 3-12 cycloalkyl and halogen, said alkyl optionally substituted with an oxo group;
  • Q is phenyl or a 6 membered heteroaromatic group containing 1-3 nitrogen atoms.
  • the R 1 alkyl is methyl, ethyl, propyl, butyl, pentyl, or hexyl.
  • the R 1 cycloalkyl is cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, or norbornyl.
  • the R 1 bicyclic ring system is naphthyl. In other preferred embodiments of formula (I), (II), (IA) or (IIA), the R 1 bicyclic ring system is tetrahydronaphthyl, or decahydronaphthyl and the R 1 tricyclic ring system is dibenzocycloheptyl. In other preferred embodiments R 1 is phenyl or benzyl.
  • the R 1 bicyclic aromatic ring is a 10-membered ring, preferably quinoline or naphthyl.
  • the R 1 bicyclic aromatic ring is a 9-membered ring, preferably indenyl.
  • Z is a bond, methyl, or ethyl.
  • the Z group is maximally substituted as not to have any hydrogen substitution on the base Z group. For example, if the base Z group is —CH 2 —, substitution with two methyl groups would remove hydrogens from the —CH 2 — base Z group.
  • n 0.
  • X 1 and X 2 are both O.
  • W is —CH 2 C( ⁇ O)NH 2 , —C(NH)NH 2 , pyridylmethyl, cyclopentyl, cyclohexyl, furanylmethyl, —C( ⁇ O)CH 3 , —CH 2 CH 2 NHC( ⁇ O)CH 3 , —SO 2 CH 3 , CH 2 CH 2 NHSO 2 CH 3 , furanylcarbonyl-, methylpyrrolylcarbonyl-, diazolecarbonyl-, azolemethyl-, trifluoroethyl-, hydroxyethyl-, cyanomethyl-, oxo-oxazolemethyl-, or diazolemethyl-.
  • ZR 1 is cyclohexylethyl-, cyclohexylmethyl-, cyclopentylmethyl-, dimethylcyclohexylmethyl-, phenylethyl-, pyrrolyltrifluoroethyl-, thienyltrifluoroethyl-, pyridylethyl-, cyclopentyl-, cyclohexyl-, methoxycyclohexyl-, tetrahydropyranyl-, propylpiperidinyl-, indolylmethyl-, pyrazoylpentyl-, thiazolylethyl-, phenyltrifluoroethyl-, hydroxyhexyl-, methoxyhexyl-, isopropoxybutyl-, hexyl-, or oxocanylpropyl-.
  • At least one of ZR 1 or W is —CH 2 COOV 1 , tetrazolylmethyl-, cyanomethyl-, NH 2 SO 2 methyl-, NH 2 SOmethyl-, aminocarbonylmethyl-, C 1-4 alkylaminocarbonylmethyl-, or diC 1-4 alkylaminocarbonylmethyl-.
  • ZR 1 is 3,3 diphenylpropyl optionally substituted at the 3 carbon of the propyl with —COOV 1 , tetrazolylC 1-4 alkyl-, cyano-, aminocarbonyl-, C 1-4 alkylaminocarbonyl-, or diC 1-4 alkylaminocarbonyl-.
  • ZR 1 in formula (I), (II), (IA) or (IIA) can be
  • Y 1 is R 3 —(C 1 -C 12 )alkyl, R 4 -aryl, R 5 -heteroaryl, R 6 —(C 3 -C 12 )cyclo-alkyl, R 7 —(C 3 -C 7 )heterocycloalkyl, —CO 2 (C 1 -C 6 )alkyl, CN or —C(O)NR 8 R 9 ;
  • Y 2 is hydrogen or Y;
  • Y 3 is hydrogen or (C 1 -C 6 )alkyl; or Y 1 , Y 2 and Y 3 , together with the carbon to which they are attached, form one of the following structures:
  • R 10 is 1 to 3 substituents independently selected from the group consisting of H, (C 1 -C 6 )alkyl, —OR 8 , —(C 1 -C 6 )alkyl-OR 8 , —NR 8 R 9 and —(C 1 -C 6 )alkyl-NR 8 R 9 ;
  • R 11 is 1 to 3 substituents independently selected from the group consisting of R 10 , —CF 3 , —OCF 3 , NO 2 and halo, or R 11 substituents on adjacent ring carbon atoms may together form a methylenedioxy or ethylenedioxy ring;
  • R 8 and R 9 are independently selected from the group consisting of hydrogen, (C 1 -C 6 ) alkyl, (C 3 -C 12 )cycloalkyl, aryl and aryl(C 1 -C 6 )alkyl;
  • R 3 is 1 to 3 substituents independently selected from the group consisting of H, R 4 -aryl, R 6 —(C 3 -C 12 )cycloalkyl, R 5 -heteroaryl, R 7 —(C 3 -C 7 )heterocycloalkyl, —NR 8 R 9 , —OR 12 and —S(O) 0-2 R 12 ;
  • R 6 is 1 to 3 substituents independently selected from the group consisting of H, (C 1 -C 6 )alkyl, R 4 -aryl, —NR 8 R 9 , —OR 12 and —SR 12 ;
  • R 4 is 1 to 3 substituents independently selected from the group consisting of hydrogen, halo, (C 1 -C 6 )alkyl, R 13 -aryl, (C 3 -C 12 )cycloalkyl, —CN, —CF 3 , —OR 8 , —(C 1 -C 6 )alkyl-OR 8 , —OCF 3 , —NR 8 R 9 , —(C 1 -C 6 )alkyl —NR 8 R 9 , —NHSO 2 R 8 , —SO 2 N(R 14 ) 2 , —SO 2 R 8 , —SOR 8 , —SR 8 , —NO 2 , —CONR 8 R 9 , —NR 9 COR 8 , —COR 8 , —COCF 3 , —OCOR 8 , —OCO 2 R 8 , —COOR 8 , —(C 1 -C 6 )alkyl-NHCOOC(CH
  • R 4 substituents on adjacent ring carbon atoms may together form a methylenedioxy or ethylenedioxy ring;
  • R 5 is 1 to 3 substituents independently selected from the group consisting of hydrogen, halo, (C 1 -C 6 )alkyl, R 13 -aryl, (C 3 -C 12 )cycloalkyl, —CN, —CF 3 , —OR 8 , —(C 1 -C 6 )alkyl-OR 8 , —OCF 3 , —NR 8 R 9 , —(C 1 -C 6 )alkyl-NR 8 R 9 , —NHSO 2 R 8 , —SO 2 N(R 14 ) 2 , —NO 2 , —CONR 8 R 9 , —NR 9 COR 8 , —COR 8 , —OCOR 8 , —OCO 2 R 8 and —COOR 8 ;
  • R 7 is H, (C 1 -C 6 )alkyl, —OR 8 , —(C 1 -C 6 )alkyl-OR 8 , —NR 8 R 9 or —(C 1 -C 6 )alkyl-NR 8 R 9 ;
  • R 12 is H, (C 1 -C 6 )alkyl, R 4 -aryl, —(C 1 -C 6 )alkyl-OR 8 , —(C 1 -C 6 )alkyl-NR 8 R 9 , —(C 1 -C 6 )alkyl-SR 8 , or aryl(C 1 -C 6 )alkyl;
  • R 13 is 1-3 substituents independently selected from the group consisting of H, (C 1 -C 6 )alkyl, (C 1 -C 6 )alkoxy and halo;
  • R 14 is independently selected from the group consisting of H, (C 1 -C 6 )alkyl and R 13 —C 6 H 4 —CH 2 —.
  • alkyl means a linear or branched saturated aliphatic hydrocarbon group having a single radical and 1-10 carbon atoms.
  • alkyl groups include methyl, propyl, isopropyl, butyl, n-butyl, isobutyl, sec-butyl, tert-butyl, and pentyl.
  • a branched alkyl means that one or more alkyl groups such as methyl, ethyl or propyl, replace one or both hydrogens in a —CH 2 — group of a linear alkyl chain.
  • lower alkyl means an alkyl of 1-3 carbon atoms.
  • alkoxy means an “alkyl” as defined above connected to an oxygen radical.
  • cycloalkyl means a non-aromatic mono- or multicyclic hydrocarbon ring system having a single radical and 3-12 carbon atoms.
  • exemplary monocyclic cycloalkyl rings include cyclopropyl, cyclopentyl, and cyclohexyl.
  • Exemplary multicyclic cycloalkyl rings include adamantyl and norbornyl.
  • alkenyl means a linear or branched aliphatic hydrocarbon group containing a carbon-carbon double bond having a single radical and 2-10 carbon atoms.
  • alkenyl groups include ethenyl, 1- and 2-propenyl, 1-, 2- and 3-butenyl, 3-methylbut-2-enyl, 2-propenyl, heptenyl, octenyl and decenyl.
  • cycloalkenyl means a non-aromatic monocyclic or multicyclic hydrocarbon ring system containing a carbon-carbon double bond having a single radical and 3 to 12 carbon atoms.
  • exemplary monocyclic cycloalkenyl rings include cyclopropenyl, cyclopentenyl, cyclohexenyl or cycloheptenyl.
  • An exemplary multicyclic cycloalkenyl ring is norbornenyl.
  • aryl means a carbocyclic aromatic ring system containing one, two or three rings which may be attached together in a pendent manner or fused, and containing a single radical.
  • exemplary aryl groups include phenyl, naphthyl and acenaphthyl.
  • heterocyclic means cyclic compounds having one or more heteroatoms (atoms other than carbon) in the ring, and having a single radical.
  • the ring may be saturated, partially saturated or unsaturated, and the heteroatoms may be selected from the group consisting of nitrogen, sulfur and oxygen.
  • saturated heterocyclic radicals include saturated 3 to 6-membered hetero-monocyclic groups containing 1 to 4 nitrogen atoms, such as pyrrolidinyl, imidazolidinyl, piperidino, piperazinyl; saturated 3- to 6-membered hetero-monocyclic groups containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms, such as morpholinyl; saturated 3- to 6-membered hetero-monocyclic groups containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms, such as thiazolidinyl.
  • partially saturated heterocyclic radicals include dihydrothiophene, dihydropyran, and dihydrofuran.
  • Other heterocyclic groups can be 7 to 10 carbon rings substituted with heteroatoms such as oxocanyl and thiocanyl.
  • the sulfur can be a sulfur dioxide such as thiocanyldioxide.
  • heteroaryl means unsaturated heterocyclic radicals, wherein “heterocyclic” is as previously described.
  • exemplary heteroaryl groups include unsaturated 3 to 6 membered hetero-monocyclic groups containing 1 to 4 nitrogen atoms, such as pyrrolyl, pyridyl, pyrimidyl, and pyrazinyl; unsaturated condensed heterocyclic groups containing 1 to 5 nitrogen atoms, such as indolyl, quinolyl and isoquinolyl; unsaturated 3 to 6-membered hetero-monocyclic groups containing an oxygen atom, such as furyl; unsaturated 3 to 6 membered hetero-monocyclic groups containing a sulfur atom, such as thienyl; unsaturated 3 to 6 membered hetero-monocyclic groups containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms, such as oxazolyl; unsaturated condensed heterocyclic groups containing 1 to 2 oxygen atoms and
  • heteroaryl also includes unsaturated heterocyclic radicals, wherein “heterocyclic” is as previously described, in which the heterocyclic group is fused with an aryl group, in which aryl is as previously described.
  • fused radicals include benzofuran, benzodioxole and benzothiophene.
  • heterocyclicC 1-4 alkyl refers to the ring structure bonded to a C 1-4 alkyl radical.
  • the term “patient” includes a human or an animal such as a companion animal or livestock.
  • halogen includes fluoride, bromide, chloride, iodide or alabamide.
  • the invention disclosed herein is meant to encompass all pharmaceutically acceptable salts thereof of the disclosed compounds.
  • the pharmaceutically acceptable salts include, but are not limited to, metal salts such as sodium salt, potassium salt, cesium salt and the like; alkaline earth metals such as calcium salt, magnesium salt and the like; organic amine salts such as triethylamine salt, pyridine salt, picoline salt, ethanolamine salt, triethanolamine salt, dicyclohexylamine salt, N,N′-dibenzylethylenediamine salt and the like; inorganic acid salts such as hydrochloride, hydrobromide, sulfate, phosphate and the like; organic acid salts such as formate, acetate, trifluoroacetate, maleate, fumarate, tartrate and the like; sulfonates such as methanesulfonate, benzenesulfonate, p-toluenesulfonate, and the like; amino acid salts such as arginate
  • Prodrugs are considered to be any covalently bonded carriers which release the active parent drug in vivo.
  • the invention disclosed herein is also meant to encompass the in vivo metabolic products of the disclosed compounds. Such products may result for example from the oxidation, reduction, hydrolysis, amidation, esterification and the like of the administered compound, primarily due to enzymatic processes. Accordingly, the invention includes compounds produced by a process comprising contacting a compound of this invention with a mammal for a period of time sufficient to yield a metabolic product thereof. Such products typically are identified by preparing a radiolabelled compound of the invention, administering it parenterally in a detectable dose to an animal such as rat, mouse, guinea pig, monkey, or to man, allowing sufficient time for metabolism to occur and isolating its conversion products from the urine, blood or other biological samples.
  • the invention disclosed herein is also meant to encompass the disclosed compounds being isotopically-labelled by having one or more atoms replaced by an atom having a different atomic mass or mass number.
  • isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine and chlorine, such as 2 H, 3 H, 13 C, 14 C, 15 N, 18 O, 17 O, 31 P, 32 P, 35 S, 18 F, and 36 Cl, respectively.
  • Some of the compounds disclosed herein may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms.
  • the present invention is also meant to encompass all such possible forms as well as their racemic and resolved forms and mixtures thereof.
  • the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended to include both E and Z geometric isomers. All tautomers are intended to be encompassed by the present invention as well
  • stereoisomers is a general term for all isomers of individual molecules that differ only in the orientation of their atoms in space. It includes enantiomers and isomers of compounds with more than one chiral center that are not mirror images of one another (diastereomers).
  • chiral center refers to a carbon atom to which four different groups are attached.
  • enantiomer or “enantiomeric” refers to a molecule that is nonsuperimposeable on its mirror image and hence optically active wherein the enantiomer rotates the plane of polarized light in one direction and its mirror image rotates the plane of polarized light in the opposite direction.
  • racemic refers to a mixture of equal parts of enantiomers and which is optically inactive.
  • resolution refers to the separation or concentration or depletion of one of the two enantiomeric forms of a molecule.
  • modulate means the mediation of a pharmacodynamic response (e.g., analgesia) in a subject from (i) inhibiting or activating the receptor, or (ii) directly or indirectly affecting the normal regulation of the receptor activity.
  • pharmacodynamic response e.g., analgesia
  • Compounds which modulate the receptor activity include agonists, antagonists, mixed agonists/antagonists and compounds which directly or indirectly affect regulation of the receptor activity.
  • Certain preferred compounds according to the invention of formulae (I) and (IA) include:
  • Certain preferred compounds according to the invention of formula (II) and (IIA) include:
  • the compounds of the present invention can be administered to anyone requiring modulation of the opioid and ORL1 receptors. Administration may be orally, topically, by suppository, inhalation, or parenterally.
  • the present invention also encompasses all pharmaceutically acceptable salts of the foregoing compounds.
  • acid addition salts of the presently claimed compounds may be prepared by reaction of the compounds with the appropriate acid via a variety of known methods.
  • Various oral dosage forms can be used, including such solid forms as tablets, gelcaps, capsules, caplets, granules, lozenges and bulk powders and liquid forms such as emulsions, solution and suspensions.
  • the compounds of the present invention can be administered alone or can be combined with various pharmaceutically acceptable carriers and excipients known to those skilled in the art, including but not limited to diluents, suspending agents, solubilizers, binders, disintegrants, preservatives, coloring agents, lubricants and the like.
  • Liquid oral dosage forms include aqueous and nonaqueous solutions, emulsions, suspensions, and solutions and/or suspensions reconstituted from non-effervescent granules, containing suitable solvents, preservatives, emulsifying agents, suspending agents, diluents, sweeteners, coloring agents, and flavoring agents.
  • the compounds of the present invention may be injected parenterally, they may be, e.g., in the form of an isotonic sterile solution.
  • the compounds of the present invention when the compounds of the present invention are to be inhaled, they may be formulated into a dry aerosol or may be formulated into an aqueous or partially aqueous solution.
  • dosage forms may provide an immediate release of the compound in the gastrointestinal tract, or alternatively may provide a controlled and/or sustained release through the gastrointestinal tract.
  • controlled and/or sustained release formulations are well known to those skilled in the art, and are contemplated for use in connection with the formulations of the present invention.
  • the controlled and/or sustained release may be provided by, e.g., a coating on the oral dosage form or by incorporating the compound(s) of the invention into a controlled and/or sustained release matrix.
  • the formulation for parenteral administration may be in the form of suspensions, solutions, emulsions in oily or aqueous vehicles, and such formulations may further comprise pharmaceutically necessary additives such as stabilizing agents, suspending agents, dispersing agents, and the like.
  • the compounds of the invention may also be in the form of a powder for reconstitution as an injectable formulation.
  • the compounds of the present invention can be used in combination with at least one other therapeutic agent.
  • Therapeutic agents include, but are not limited to, ⁇ -opioid agonists; non-opioid analgesics; non-steroid antiinflammatory agents; Cox-II inhibitors; antiemetics; ⁇ -adrenergic blockers; anticonvulsants; antidepressants; Ca2+-channel blockers; anticancer agent and mixtures thereof.
  • the compounds of the present invention can be formulated in a pharmaceutical dosage form in combination with a ⁇ -opioid agonist.
  • ⁇ -opioid agonists which may be included in the formulations of the present invention include but are not limited to include alfentanil, allylprodine, alphaprodine, anileridine, benzylmorphine, bezitramide, buprenorphine, butorphanol, clonitazene, codeine, desomorphine, dextromoramide, dezocine, diampromide, diamorphone, dihydrocodeine, dihydromorphine, dimenoxadol, dimepheptanol, dimethylthiambutene, dioxaphetyl butyrate, dipipanone, eptazocine, ethoheptazine, ethylmethylthiambutene, ethylmorphine, etonitazene fentany
  • the ⁇ -opioid agonist is selected from codeine, hydromorphone, hydrocodone, oxycodone, dihydrocodeine, dihydromorphine, morphine, tramadol, oxymorphone, pharmaceutically acceptable salts thereof, and mixtures thereof.
  • the medicament comprises a mixture of a Cox-II inhibitor and an inhibitor of 5-lipoxygenase for the treatment of pain and/or inflammation.
  • a Cox-II inhibitor and an inhibitor of 5-lipoxygenase for the treatment of pain and/or inflammation.
  • Suitable Cox-II inhibitors and 5-lipoxygenase inhibitors, as well as combinations thereof are described in U.S. Pat. No. 6,136,839, which is hereby incorporated by reference in its entirety.
  • Cox-II inhibitors include, but are not limited to rofecoxib (Vioxx), celecoxib (Celebrex), DUP-697, flosulide, meloxicam, 6-MNA, L-745337, nabumetone, nimesulide, NS-398, SC-5766, T-614, L-768277, GR-253035, JTE-522, RS-57067-000, SC-58125, SC-078, PD-138387, NS-398, flosulide, D-1367, SC-5766, PD-164387, etoricoxib, valdecoxib and parecoxib or pharmaceutically acceptable salts, enantiomers or tautomers thereof.
  • non-opioid analgesics e.g., non-steroidal anti-inflammatory agents, including aspirin, ibuprofen, diclofenac, naproxen, benoxaprofen, flurbiprofen, fenoprofen, flubufen, ketoprofen, indoprofen, piroprofen, carprofen, oxaprozin, pramoprofen, muroprofen, trioxaprofen, suprofen, aminoprofen, tiaprofenic acid, fluprofen, bucloxic acid, indomethacin, sulindac, tolmetin, zomepirac, tiopinac, zidometacin, acemetacin, fentiazac, clidanac, oxpinac, mefenamic acid, meclofenamic acid, flufenamic
  • non-steroidal anti-inflammatory agents including aspirin
  • non-opioid analgesics which may be included in the dosage forms of the present invention include the following, non-limiting, chemical classes of analgesic, antipyretic, nonsteroidal antiinflammatory drugs: salicylic acid derivatives, including aspirin, sodium salicylate, choline magnesium trisalicylate, salsalate, diflunisal, salicylsalicylic acid, sulfasalazine, and olsalazin; para-aminophennol derivatives including acetaminophen; indole and indene acetic acids, including indomethacin, sulindac, and etodolac; heteroaryl acetic acids, including tolmetin, diclofenac, and ketorolac; anthranilic acids (fenamates), including mefenamic acid, and meclofenamic acid; enolic acids, including oxicams (piroxicam, tenoxicam), and pyrazolidinedi
  • the compounds of the present invention can be formulated in a pharmaceutical dosage form in combination with antimigraine agents.
  • Antimigraine agents include, but are not limited to, alpiropride, dihydroergotamine, dolasetron, ergocornine, ergocorninine, ergocryptine, ergot, ergotamine, flumedroxone acetate, fonazine, lisuride, lomerizine, methysergide oxetorone, pizotyline, and mixtures thereof.
  • the other therapeutic agent can also be an adjuvant to reduce any potential side effects such as, for example, an antiemetic agent.
  • Suitable antiemetic agents include, but are not limited to, metoclopromide, domperidone, prochlorperazine, promethazine, chlorpromazine, trimethobenzamide, ondansetron, granisetron, hydroxyzine, acethylleucine monoethanolamine, alizapride, azasetron, benzquinamide, bietanautine, bromopride, buclizine, clebopride, cyclizine, dimenhydrinate, diphenidol, dolasetron, meclizine, methallatal, metopimazine, nabilone, oxyperndyl, pipamazine, scopolamine, sulpiride, tetrahydrocannabinols, thiethylperazine, thioproperazine, trop
  • the compounds of the present invention can be formulated in a pharmaceutical dosage form in combination with ⁇ -adrenergic blockers.
  • Suitable ⁇ -adrenergic blockers include, but are not limited to, acebutolol, alprenolol, amosulabol, arotinolol, atenolol, befunolol, betaxolol, bevantolol, bisoprolol, bopindolol, bucumolol, bufetolol, bufuralol, bunitrolol, bupranolol, butidrine hydrochloride, butofilolol, carazolol, carteolol, carvedilol, celiprolol, cetamolol, cloranolol, dilevalol, epanolol, esmolol, indenolol, labetalol
  • the compounds of the present invention can be formulated in a pharmaceutical dosage form in combination with anticonvulsants.
  • Suitable anticonvulsants include, but are not limited to, acetylpheneturide, albutoin, aloxidone, aminoglutethimide, 4-amino-3-hydroxybutyric acid, atrolactamide, beclamide, buramate, calcium bromide, carbamazepine, cinromide, clomethiazole, clonazepam, decimemide, diethadione, dimethadione, doxenitroin, eterobarb, ethadione, ethosuximide, ethotoin, felbamate, fluoresone, gabapentin, 5-hydroxytryptophan, lamotrigine, magnesium bromide, magnesium sulfate, mephenyloin, mephobarbital, metharbital, methetoin, methsuximide, 5-methyl-5-(3-phenyl
  • the compounds of the present invention can be formulated in a pharmaceutical dosage form in combination with antidepressants.
  • Suitable antidepressants include, but are not limited to, binedaline, caroxazone, citalopram, dimethazan, fencamine, indalpine, indeloxazine hydrocholoride, nefopam, nomifensine, oxitriptan, oxypertine, paroxetine, sertraline, thiazesim, trazodone, benmoxine, iproclozide, iproniazid, isocarboxazid, nialamide, octamoxin, phenelzine, cotinine, rolicyprine, rolipram, maprotiline, metralindole, mianserin, mirtazepine, adinazolam, amitriptyline, amitriptylinoxide, amoxapine,
  • the compounds of the present invention can be formulated in a pharmaceutical dosage form in combination with Ca2+-channel blockers.
  • Suitable Ca2+-channel blockers include, but are not limited to, bepridil, clentiazem, diltiazem, fendiline, gallopamil, mibefradil, prenylamine, semotiadil, terodiline, verapamil, amlodipine, aranidipine, barnidipine, benidipine, cilnidipine, efonidipine, elgodipine, felodipine, isradipine, lacidipine, lercanidipine, manidipine, nicardipine, nifedipine, nilvadipine, nimodipine, nisoldipine, nitrendipine, cinnarizine, flunarizine, lidoflazine, lomerizine,
  • the compounds of the present invention can be formulated in a pharmaceutical dosage form in combination with anticancer agents.
  • Suitable anticancer agents include, but are not limited to, acivicin; aclarubicin; acodazole hydrochloride; acronine; adozelesin; aldesleukin; altretamine; ambomycin; ametantrone acetate; aminoglutethimide; amsacrine; anastrozole; antlramycin; asparaginase; asperlin; azacitidine; azetepa; azotomycin; batimastat; benzodepa; bicalutamide; bisantrene hydrochloride; bisnafide dimesylate; bizelesin; bleomycin sulfate; brequinar sodium; bropirimine; busulfan; cactinomycin; calusterone; caracemide; carbetimer; carboplatin; car
  • anti-cancer drugs include, but are not limited to: 20-epi-1,25 dihydroxyvitamin D3; 5-ethynyluracil; abiraterone; aclarubicin; acylfulvene; adecypenol; adozelesin; aldesleukin; ALL-TK antagonists; altretamine; ambamustine; amidox; amifostine; aminolevulinic acid; amrubicin; amsacrine; anagrelide; anastrozole; andrographolide; angiogenesis inhibitors; antagonist D; antagonist G; antarelix; anti-dorsalizing morphogenetic protein-1; antiandrogen, prostatic carcinoma; antiestrogen; antineoplaston; antisense oligonucleotides; aphidicolin glycinate; apoptosis gene modulators; apoptosis regulators; apurinic acid; ara-CDP-DL-PTBA;
  • the compounds of the present invention and the other therapeutic agent can act additively or, more preferably, synergistically.
  • a composition comprising a compounds of the present invention is administered concurrently with the administration of another therapeutic agent, which can be part of the same composition or in a different composition from that comprising the compounds of the present invention.
  • a composition comprising the compounds of the present invention is administered prior to or subsequent to administration of another therapeutic agent.
  • the compounds of the present invention when administered, e.g., via the oral, parenteral or topical routes to mammals, can be in a dosage in the range of about 0.01 mg/kg to about 3000 mg/kg body weight of the patient per day, preferably about 0.01 mg/kg to about 1000 mg/kg body weight per day administered singly or as a divided dose.
  • variations will necessarily occur depending upon the weight and physical condition (e.g., hepatic and renal function) of the subject being treated, the affliction to be treated, the severity of the symptoms, the route of administration, the frequency of the dosage interval, the presence of any deleterious side-effects, and the particular compound utilized, among other things.
  • the compounds of the present invention preferably have a binding affinity K i for the human ORL-1 receptor of about 500 nM or less; 100 nM or less; 50 nM or less; 20 nM or less or 5 nM or less.
  • the binding affinity K i can be measured by one skilled in the art by an assay utilizing membranes from recombinant HEK-293 cells expressing the human opioid receptor-like receptor (ORL-1) as described below.
  • 1,2-Phenylenediamine 1 160 g, 1.50 mol
  • 4-oxo-piperidine-tert-butylester 2 100 g, 0.50 mol
  • 1,2-dichloroethane 2.0 L
  • Acetic acid 31.6 mL
  • sodium triacetoxyborohydride 148 g, 0.70 mol
  • the solvent was evaporated and the residue partitioned between ether and 1 M acetic acid.
  • the organic layer was separated and washed with 1M acetic acid (3 ⁇ ) followed by sodium bicarbonate solution (1 ⁇ ).
  • the diamine 3 (5.4 g, 18.6 mmol) was dissolved in dry pyridine (30 mL). Sulfamide (3.58 g, 37.2 mmol) was added and the mixture heated to reflux for 2 h. The mixture was cooled to room temperature and the solvent evaporated to dryness. The residue was partitioned between dichloromethane:methanol (10:1, 500 mL) and 0.1 M hydrochloric acid (500 mL), the organic phase separated, washed with brine (500 mL), dried over MgSO 4 , filtered and the solvent evaporated. The residue was triturated with ethyl acetate to give pure 4 as a pale pink solid (5.15 g, 79%).
  • Tail Groups to the “5-Membered SO 2 ” and “6-Membered SO 2 ” Head Groups
  • Tail groups were attached to the head groups according to the following procedures:
  • Nociceptin affinity at the ORL1 receptor for preferred compounds was obtained using the following assay:
  • Membranes from recombinant HEK-293 cells expressing the human opioid receptor-like receptor (ORL-1) were prepared by lysing cells in ice-cold hypotonic buffer (2.5 mM MgCl 2 , 50 mM HEPES, pH 7.4) (10 ml/10 cm dish) followed by homogenization with a tissue grinder/teflon pestle. Membranes were collected by centrifugation at 30,000 ⁇ g for 15 min at 4° C. and pellets resuspended in hypotonic buffer to a final concentration of 1-3 mg/ml. Protein concentrations were determined using the BioRad protein assay reagent with bovine serum albumen as standard. Aliquots of the ORL-1 receptor membranes were stored at ⁇ 80° C.
  • ORL-1 membrane solution was prepared by sequentially adding final concentrations of 0.066 mg/ml ORL-1 membrane protein, 10 mg/ml saponin, 3 mM GDP and 0.20 nM [ 35 S]GTPgS to binding buffer (100 mM NaCl, 10 mM MgCl 2 , 20 mM HEPES, pH 7.4) on ice.
  • the prepared membrane solution (190 ml/well) was transferred to 96-shallow well polypropylene plates containing 10 ml of 20 ⁇ concentrated stock solutions of agonist prepared in DMSO. Plates were incubated for 30 min at room temperature with shaking.
  • Reactions were terminated by rapid filtration onto 96-well Unifilter GF/B filter plates (Packard) using a 96-well tissue harvester (Brandel) and followed by three filtration washes with 200 ml ice-cold binding buffer (10 mM NaH 2 PO 4 , 10 mM Na 2 HPO 4 , pH 7.4). Filter plates were subsequently dried at 50° C. for 2-3 hours. Fifty ml/well scintillation cocktail (BetaScint; Wallac) was added and plates were counted in a Packard Top-Count for 1 ml/well.
  • BetaScint BetaScint; Wallac
  • Tail groups were attached to the quinoline head groups according to the following procedures:
  • Nociceptin affinity at the ORL1 receptor for preferred compounds was obtained using the following assay:
  • Membranes from recombinant HEK-293 cells expressing the human opioid receptor-like receptor (ORL-1) were prepared by lysing cells in ice-cold hypotonic buffer (2.5 mM MgCl 2 , 50 mM HEPES, pH 7.4) (10 ml/10 cm dish) followed by homogenization with a tissue grinder/teflon pestle. Membranes were collected by centrifugation at 30,000 ⁇ g for 15 min at 4° C. and pellets resuspended in hypotonic buffer to a final concentration of 1-3 mg/ml. Protein concentrations were determined using the BioRad protein assay reagent with bovine serum albumen as standard. Aliquots of the ORL-1 receptor membranes were stored at ⁇ 80° C.
  • ORL-1 membrane solution was prepared by sequentially adding final concentrations of 0.066 mg/ml ORL-1 membrane protein, 10 mg/ml saponin, 3 mM GDP and 0.20 nM [ 35 S]GTPgS to binding buffer (100 mM NaCl, 10 mM MgCl 2 , 20 mM HEPES, pH 7.4) on ice.
  • the prepared membrane solution (190 ml/well) was transferred to 96-shallow well polypropylene plates containing 10 ml of 20 ⁇ concentrated stock solutions of agonist prepared in DMSO. Plates were incubated for 30 min at room temperature with shaking.
  • Reactions were terminated by rapid filtration onto 96-well Unifilter GF/B filter plates (Packard) using a 96-well tissue harvester (Brandel) and followed by three filtration washes with 200 ml ice-cold binding buffer (10 mM NaH 2 PO 4 , 10 mM Na 2 HPO 4 , pH 7.4). Filter plates were subsequently dried at 50° C. for 2-3 hours. Fifty ml/well scintillation cocktail (BetaScint; Wallac) was added and plates were counted in a Packard Top-Count for 1 min/well.
  • BetaScint BetaScint; Wallac
  • Mu opioid receptor membrane solution was prepared by sequentially adding final concentrations of 0.075 ⁇ g/ ⁇ l of the desired membrane protein, 10 ⁇ g/ml saponin, 3 ⁇ M GDP and 0.20 nM [ 35 S]GTP ⁇ S to binding buffer (100 mM NaCl, 10 mM MgCl 2 , 20 mM HEPES, pH 7.4) on ice.
  • binding buffer 100 mM NaCl, 10 mM MgCl 2 , 20 mM HEPES, pH 7.4
  • the prepared membrane solution (190 ⁇ l/well) was transferred to 96-shallow well polypropylene plates containing 10 ⁇ l of 20 ⁇ concentrated stock solutions of agonist prepared in DMSO. Plates were incubated for 30 min at room temperature with shaking.
  • Reactions were terminated by rapid filtration onto 96-well Unifilter GF/B filter plates (Packard) using a 96-well tissue harvester (Brandel) and followed by three filtration washes with 200 ⁇ l ice-cold binding buffer (10 mM NaH 2 PO 4 , 10 mM Na 2 HPO 4 , pH 7.4). Filter plates were subsequently dried at 50° C. for 2-3 hours. Fifty ⁇ l/well scintillation cocktail (MicroScint20, Packard) was added and plates were counted in a Packard Top-Count for 1 min/well.

Abstract

A compound of the having the general formula (I) or general formula (II):
Figure US20090253727A1-20091008-C00001
wherein
  • Z, A, B, C, R1, R2, Q, W, and n are as described herein.

Description

  • This application claims priority from U.S. Provisional Application Ser. Nos. 60/284,674 and 60/284,676, both filed Apr. 18, 2001, the disclosures of which are hereby incorporated by reference.
  • BACKGROUND OF THE INVENTION
  • Chronic pain is a major contributor to disability and is the cause of an untold amount of suffering. The successful treatment of severe and chronic pain is a primary goal of the physician with opioid analgesics being preferred drugs.
  • Until recently, there was evidence of three major classes of opioid receptors in the central nervous system (CNS), with each class having subtype receptors. These receptor classes were designated as μ, δ and κ. As opiates had a high affinity to these receptors while not being endogenous to the body, research followed in order to identify and isolate the endogenous ligands to these receptors. These ligands were identified as enkephalins, endorphins and dynorphins.
  • Recent experimentation has led to the identification of a cDNA encoding an opioid receptor-like (ORL1) receptor with a high degree of homology to the known receptor classes. This newly discovered receptor was classified as an opioid receptor based only on structural grounds, as the receptor did not exhibit pharmacological homology. It was initially demonstrated that non-selective ligands having a high affinity for μ, δ and κ receptors had low affinity for the ORL1. This characteristic, along with the fact that an endogenous ligand had not yet been discovered, led to the term “orphan receptor”.
  • Subsequent research led to the isolation and structure of the endogenous ligand of the ORL1 receptor. This ligand is a seventeen amino acid peptide structurally similar to members of the opioid peptide family.
  • The discovery of the ORL1 receptor presents an opportunity in drug discovery for novel compounds which can be administered for pain management or other syndromes modulated by this receptor.
  • All documents cited herein, including the foregoing, are incorporated by reference in their entireties for all purposes.
  • OBJECTS AND SUMMARY OF THE INVENTION
  • It is accordingly an object of certain embodiments of the present invention to provide new compounds which exhibit affinity for the ORL1 receptor.
  • It is an object of certain embodiments of the present invention to provide new compounds which exhibit affinity for the ORL1 receptor and one or more of the μ, δ or κ receptors.
  • It is an object of certain embodiments of the present invention to provide new compounds for treating a patient suffering from chronic or acute pain by administering a compound having affinity for the ORL1 receptor.
  • It is an object of certain embodiments of the present invention to provide new compounds which have agonist activity at the μ, δ and κ receptors which is greater than compounds currently available e.g. morphine.
  • It is an object of certain embodiments of the present invention to provide methods of treating chronic and acute pain by administering compounds which have agonist activity at the μ, δ and κ receptors which is greater than compounds currently available.
  • It is an object of certain embodiments of the present invention to provide methods of treating chronic and acute pain by administering non-opioid compounds which have agonist activity at the μ, δ and κ receptors and which produce less side effects than compounds currently available.
  • It is an object of certain embodiments of the present invention to provide compounds useful as analgesics, anti-inflammatories, diuretics, anesthetics and neuroprotective agents, anti-hypertensives, anti-anxiolytics; agents for appetite control; hearing regulators; anti-tussives, anti-asthmatics, modulators of locomotor activity, modulators of learning and memory, regulators of neurotransmitter and hormone release, kidney function modulators, anti-depressants, agents to treat memory loss due to Alzheimer's disease or other dementias, anti-epileptics, anti-convulsants, agents to treat withdrawal from alcohol and drugs of addiction, agents to control water balance, agents to control sodium excretion and agents to control arterial blood pressure disorders and methods for administering said compounds.
  • The compounds of the present invention are useful for modulating a pharmacodynamic response from one or more opioid receptors (ORL-1, μ, δ and κ) centrally and/or peripherally. The response can be attributed to the compound stimulating (agonist) or inhibiting (antagonist) the one or more receptors. Certain compounds can stimulate one receptor (e.g., a 1 agonist) and inhibit a different receptor (e.g., an ORL-1 antagonist).
  • Other objects and advantages of the present invention will become apparent from the following detailed description thereof. The present invention in certain embodiments comprises compounds having the general formula (I):
  • Figure US20090253727A1-20091008-C00002
  • wherein W is hydrogen, C1-10 alkyl, C3-12 cycloalkyl, C3-12 cycloalkylC1-4alkyl-, C1-10 alkoxy, C3-12 cycloalkoxy-, C1-10 alkyl substituted with 1-3 halogen, C3-12 cycloalkyl substituted with 1-3 halogen, C3-12 cycloalkylC1-4alkyl-substituted with 1-3 halogen, C1-10 alkoxy substituted with 1-3 halogen, C3-12 cycloalkoxy-substituted with 1-3 halogen, —COOV1, —C1-4COOV1, —CH2OH, —SO2N(V1)2, hydroxyC3-10alkyl-, hydroxyC3-10cycloalkyl-, cyanoC1-10alkyl-, cyanoC3-10cycloalkyl-, —CON(V1)2, NH2SO2C1-4alkyl-, NH2SOC1-4alkyl-, sulfonylaminoC1-10alkyl-, diaminoalkyl-, -sulfonylC1-4alkyl, a 6-membered heterocyclic ring, a 6-membered heteroaromatic ring, a 6-membered heterocyclicC1-4alkyl-, a 6-membered heteroaromaticC1-4alkyl-, a 6-membered aromatic ring, a 6-membered aromaticC1-4 alkyl-, a 5-membered heterocyclic ring optionally substituted with an oxo or thio, a 5-membered heteroaromatic ring, a 5-membered heterocyclicC1-4alkyl-optionally substituted with an oxo or thio, a 5-membered heteroaromaticC1-4alkyl-, —C1-5(═O)W1, —C1-5(═NH)W1, —C1-5NHC(═O)W1, —C1-5NHS(═O)2W1, —C1-5NHS(═O)W1, wherein W1 is hydrogen, C1-10 alkyl, C3-12 cycloalkyl, C1-10 alkoxy, C3-12 cycloalkoxy, —CH2OH, amino, C1-4alkylamino-, diC1-4alkylamino-, or a 5-membered heteroaromatic ring optionally substituted with 1-3 lower alkyl;
  • wherein each V1 is independently selected from H, C1-6 alkyl, C3-6 cycloalkyl, benzyl and phenyl;
  • Q is a 5-8 membered cycloalkyl, 5-8 membered heterocyclic or a 6 membered aromatic or heteroaromatic group;
  • each n is independently an integer from 0 to 3;
  • A, B and C are independently hydrogen, C1-10 alkyl, C3-12 cycloalkyl, C1-10 alkoxy, C3-12 cycloalkoxy, —CH2OH, —NHSO2, hydroxyC1-10alkyl-, aminocarbonyl-, C1-4alkylaminocarbonyl-, diC1-4alkylaminocarbonyl-, acylamino-, acylaminoalkyl-, amide, sulfonylaminoC1-10alkyl-, or A-B can together form a C2-6 bridge, or B-C can together form a C3-7 bridge, or A-C can together form a C1-5 bridge;
  • Z is selected from the group consisting of a bond, straight or branched C1-6 alkylene, —NH—, —CH2O—, —CH2NH—, —CH2N(CH3)—, —NHCH2—, —CH2CONH—, —NHCH2CO—, —CH2CO—, —COCH2—, —CH2COCH2—, —CH(CH3)—, —CH═, —O— and —HC═CH—, wherein the carbon and/or nitrogen atoms are unsubstituted or substituted with one or more lower alkyl, hydroxy, halo or alkoxy group;
  • R1 is selected from the group consisting of hydrogen, C1-10 alkyl, C3-12cycloalkyl, C2-10alkenyl, amino, C1-10alkylamino-, C3-12cycloalkylamino-, —COOV1, —C1-4COOV1, cyano, cyanoC1-10alkyl-, cyanoC3-10cycloalkyl-, NH2SO2—, NH2SO2C1-4alkyl-, NH2SOC1-4alkyl-, aminocarbonyl-, C1-4alkylaminocarbonyl-, diC1-4alkylaminocarbonyl-, benzyl, C3-12cycloalkenyl-, a monocyclic, bicyclic or tricyclic aryl or heteroaryl ring, a hetero-monocyclic ring, a hetero-bicyclic ring system, and a spiro ring system of the formula (III):
  • Figure US20090253727A1-20091008-C00003
  • wherein X1 and X2 are independently selected from the group consisting of NH, O, S and CH2; and wherein said alkyl, cycloalkyl, alkenyl, C1-10alkylamino-, C3-12cycloalkylamino-, or benzyl of R1 is optionally substituted with 1-3 substituents selected from the group consisting of halogen, hydroxy, C1-10 alkyl, C1-10 alkoxy, nitro, trifluoromethyl-, cyano, —COOV1, —C1-4COOV1, cyanoC1-10alkyl-, —C1-5(═O)W1, —C1-5NHS(═O)2W1, —C1-5NHS(═O)W1, a 5-membered heteroaromaticC0-4alkyl-, phenyl, benzyl, benzyloxy, said phenyl, benzyl, and benzyloxy optionally being substituted with 1-3 substituents selected from the group consisting of halogen, C1-10 alkyl-, C1-10 alkoxy-, and cyano; and wherein said C3-12 cycloalkyl, C3-12 cycloalkenyl, monocyclic, bicyclic or tricyclic aryl, heteroaryl ring, hetero-monocyclic ring, hetero-bicyclic ring system, or spiro ring system of the formula (III) is optionally substituted with 1-3 substituents selected from the group consisting of halogen, C1-10 alkyl, C1-10 alkoxy, nitro, trifluoromethyl-, phenyl, benzyl, phenyloxy and benzyloxy, wherein said phenyl, benzyl, phenyloxy or benzyloxy is optionally substituted with 1-3 substituents selected from the group consisting of halogen, C1-10 alkyl, C1-10 alkoxy, and cyano;
  • R2 is selected from the group consisting of hydrogen, C1-10 alkyl, C3-12 cycloalkyl- and halogen, said alkyl or cycloalkyl optionally substituted with an oxo, amino, alkylamino or dialkylamino group;
  • and pharmaceutically acceptable salts thereof and solvates thereof.
  • The present invention in certain embodiments comprises compounds having the formula (IA):
  • Figure US20090253727A1-20091008-C00004
  • wherein
  • each n is independently an integer from 0 to 3;
  • Z is selected from the group consisting of a bond, —CH2—, —NH—, —CH2O—, —CH2CH2—, —CH2NH—, —CH2N(CH3)—, —NHCH2—, —CH2CONH—, —NHCH2CO—, —CH2CO—, —COCH2—, —CH2COCH2—, —CH(CH3)—, —CH═, and —HC═CH—, wherein the carbon and/or nitrogen atoms are unsubstituted or substituted with a lower alkyl, halogen, hydroxy or alkoxy group;
  • R1 is selected from the group consisting of hydrogen, C1-10alkyl, C3-12cycloalkyl, C2-10alkenyl, amino, C1-10allylamino, C3-12cycloalkylamino, benzyl, C3-12 cycloalkenyl, a monocyclic, bicyclic or tricyclic aryl or heteroaryl ring, a hetero-monocyclic ring, a hetero-bicyclic ring system, and a spiro ring system of the formula (III):
  • Figure US20090253727A1-20091008-C00005
  • wherein X1 and X2 are independently selected from the group consisting of NH, O, S and CH2;
  • wherein said monocyclic aryl is preferably phenyl;
  • wherein said bicyclic aryl is preferably naphthyl;
  • wherein said alkyl, cycloalkyl, alkenyl, C1-10alkylamino, C3-12cycloalkylamino, or benzyl is optionally substituted with 1-3 substituents selected from the group consisting of halogen, C1-10 alkyl, C1-10 alkoxy, nitro, trifluoromethyl, cyano, phenyl, benzyl, benzyloxy, said phenyl, benzyl, and benzyloxy optionally being substituted with 1-3 substituents selected from the group consisting of halogen, C1-10 alkyl, C1-10 alkoxy, and cyano;
  • wherein said C3-12 cycloalkyl, C3-12 cycloalkenyl, monocyclic, bicyclic or tricyclic aryl, heteroaryl ring, hetero-monocyclic ring, hetero-bicyclic ring system, and spiro ring system of the formula (III) are optionally substituted with 1-3 substituents selected from the group consisting of halogen, C1-10 alkyl, C1-10 alkoxy, nitro, trifluoromethyl, phenyl, benzyl, phenyloxy and benzyloxy, wherein said phenyl, benzyl, phenyloxy and benzyloxy are optionally substituted with 1-3 substituents selected from the group consisting of halogen, C1-10 alkyl, C1-10 alkoxy, and cyano;
  • R2 is selected from the group consisting of hydrogen, C1-10 alkyl, C3-12 cycloalkyl and halogen, said alkyl optionally substituted with an oxo group;
  • and pharmaceutically acceptable salts thereof and solvates thereof.
  • The present invention in certain embodiments comprises compounds having the general formula (II):
  • Figure US20090253727A1-20091008-C00006
  • wherein W is hydrogen, C1-10 alkyl, C3-12 cycloalkyl, C3-12 cycloalkylC1-4alkyl-, C1-10 alkoxy, C3-12 cycloalkoxy-, C1-10 alkyl substituted with 1-3 halogen, C3-12 cycloalkyl substituted with 1-3 halogen, C3-12 cycloalkylC1-4alkyl-substituted with 1-3 halogen, C1-10alkoxy substituted with 1-3 halogen, C3-12 cycloalkoxy-substituted with 1-3 halogen, —COOV1, —C1-4COOV1, —CH2OH, —SO2N(V1)2, hydroxyC1-10alkyl-, hydroxyC3-10cycloalkyl-, cyanoC1-10alkyl-, cyanoC3-10cycloalkyl-, —CON(V1)2, NH2SO2C1-4alkyl-, NH2SOC1-4alkyl-, sulfonylaminoC1-10alkyl-, diaminoalkyl-, -sulfonylC1-4alkyl, a 6-membered heterocyclic ring, a 6-membered heteroaromatic ring, a 6-membered heterocyclicC1-4alkyl-, a 6-membered heteroaromaticC1-4alkyl-, a 6-membered aromatic ring, a 6-membered aromaticC1-4 alkyl-, a 5-membered heterocyclic ring optionally substituted with an oxo or thio, a 5-membered heteroaromatic ring, a 5-membered heterocyclicC1-4alkyl- optionally substituted with an oxo or thio, a 5-membered heteroaromaticC1-4alkyl-, —C1-5(═O)W1, —C1-5(═NH)W1, —C1-5NHC(═O)W1, —C1-5NHS(═O)2W1, —C1-5NHS(═O)W1, wherein W1 is hydrogen, C1-10 alkyl, C3-12 cycloalkyl, C1-10 alkoxy, C3-12 cycloalkoxy, —CH2OH, amino, C1-4alkylamino-, diC1-4alkylamino-, or a 5-membered heteroaromatic ring optionally substituted with 1-3 lower alkyl;
  • wherein each V1 is independently selected from H, C1-6 alkyl, C3-6 cycloalkyl, benzyl and phenyl;
  • Q is a 5-8 membered cycloalkyl, 5-8 membered heterocyclic or a 6 membered aromatic or heteroaromatic group;
  • n is an integer from 0 to 3;
  • A, B and C are independently hydrogen, C1-10 allyl, C3-12 cycloalkyl, C1-10 alkoxy, C3-12 cycloalkoxy, —CH2OH, —NHSO2, hydroxyC1-10alkyl-, aminocarbonyl-, C1-4alkylaminocarbonyl-, diC1-4alkylaminocarbonyl-, acylamino-, acylaminoalkyl-, amide, sulfonylaminoC1-10alkyl-, or A-B can together form a C2-6 bridge, or B-C can together form a C3-7 bridge, or A-C can together form a C1-5 bridge;
  • Z is selected from the group consisting of a bond, straight or branched C1-6 alkylene, —NH—, —CH2O—, —CH2NH—, —CH2N(CH3)—, —NHCH2—, —CH2CONH—, —NHCH2CO—, —CH2CO—, —COCH2—, —CH2COCH2—, —CH(CH3)—, —CH═, —O— and —HC═CH—, wherein the carbon and/or nitrogen atoms are unsubstituted or substituted with one or more lower alkyl, hydroxy, halo or alkoxy group;
  • R1 is selected from the group consisting of hydrogen, C1-10 alkyl, C3-12cycloalkyl, C2-10alkenyl, amino, C1-10alkylamino-, C3-12cycloalkylamino-, —COOV1, —C1-4COOV1, cyano, cyanoC1-10alkyl-, cyanoC3-10cycloalkyl-, NH2SO2—, NH2SO2C1-4allyl-, NH2SOC1-4alkyl-, aminocarbonyl-, C1-4alkylaminocarbonyl-, diC1-4alkylaminocarbonyl-, benzyl, C3-12 cycloalkenyl-, a monocyclic, bicyclic or tricyclic aryl or heteroaryl ring, a hetero-monocyclic ring, a hetero-bicyclic ring system, and a spiro ring system of the formula (III):
  • Figure US20090253727A1-20091008-C00007
  • wherein X1 and X2 are independently selected from the group consisting of NH, O, S and CH2; and wherein said alkyl, cycloalkyl, alkenyl, C1-10alkylamino-, C3-12cycloalkylamino-, or benzyl of R1 is optionally substituted with 1-3 substituents selected from the group consisting of halogen, hydroxy, C1-10 alkyl, C1-10 alkoxy, nitro, trifluoromethyl-, cyano, —COOV1, —C1-4COOV1, cyanoC1-10alkyl-, —C1-5(═O)W1, —C1-5NHS(═O)2W1, —C1-5NHS(═O)W1, a 5-membered heteroaromaticC0-4alkyl-, phenyl, benzyl, benzyloxy, said phenyl, benzyl, and benzyloxy optionally being substituted with 1-3 substituents selected from the group consisting of halogen, C1-10 alkyl-, C1-10 alkoxy-, and cyano; and wherein said C3-12 cycloalkyl, C3-12 cycloalkenyl, monocyclic, bicyclic or tricyclic aryl, heteroaryl ring, hetero-monocyclic ring, hetero-bicyclic ring system, or spiro ring system of the formula (III) is optionally substituted with 1-3 substituents selected from the group consisting of halogen, C1-10 alkyl, C1-10 alkoxy, nitro, trifluoromethyl-, phenyl, benzyl, phenyloxy and benzyloxy, wherein said phenyl, benzyl, phenyloxy or benzyloxy is optionally substituted with 1-3 substituents selected from the group consisting of halogen, C1-10 alkyl, C1-10 alkoxy, and cyano;
  • R2 is selected from the group consisting of hydrogen, C1-10 alkyl, C3-12 cycloalkyl- and halogen, said alkyl or cycloalkyl optionally substituted with an oxo, amino, alkylamino or dialkylamino group;
  • and pharmaceutically acceptable salts thereof and solvates thereof.
  • The present invention in certain embodiments comprises compounds having the formula (IIA) as follows:
  • Figure US20090253727A1-20091008-C00008
  • wherein
  • n is an integer from 0 to 3;
  • Z is selected from the group consisting of a bond, —CH2—, —NH—, —CH2O—, —CH2CH2—, —CH2NH—, —CH2N(CH3)—, —NHCH2—, —CH2CONH—, —NHCH2CO—, —CH2CO—, —COCH2—, —CH2COCH2—, —CH(CH3)—, —CH═, and —HC═CH—, wherein the carbon and/or nitrogen atoms are unsubstituted or substituted with a lower alkyl, halogen, hydroxy or alkoxy group;
  • R1 is selected from the group consisting of hydrogen, C1-10alkyl, C3-12cycloalkyl, C2-10alkenyl, amino, C1-10alkylamino, C3-12cycloalkylamino, benzyl, C3-12 cycloalkenyl, a monocyclic, bicyclic or tricyclic aryl or heteroaryl ring, a hetero-monocyclic ring, a hetero-bicyclic ring system, and a spiro ring system of the formula (III):
  • Figure US20090253727A1-20091008-C00009
  • wherein X1 and X2 are independently selected from the group consisting of NH, O, S and CH2;
  • wherein said monocyclic aryl is preferably phenyl;
  • wherein said bicyclic aryl is preferably naphthyl;
  • wherein said alkyl, cycloalkyl, alkenyl, C1-10alkylamino, C3-12cycloalkylamino, or benzyl is optionally substituted with 1-3 substituents selected from the group consisting of halogen, C1-10 alkyl, C1-10 alkoxy, nitro, trifluoromethyl, cyano, phenyl, benzyl, benzyloxy, said phenyl, benzyl, and benzyloxy optionally being substituted with 1-3 substituents selected from the group consisting of halogen, C1-10 alkyl, C1-10 alkoxy, and cyano;
  • wherein said C3-12 cycloalkyl, C3-12 cycloalkenyl, monocyclic, bicyclic or tricyclic aryl, heteroaryl ring, hetero-monocyclic ring, hetero-bicyclic ring system, and spiro ring system of the formula (III) are optionally substituted with 1-3 substituents selected from the group consisting of halogen, C1-10 alkyl, C1-10 alkoxy, nitro, trifluoromethyl, phenyl, benzyl, phenyloxy and benzyloxy, wherein said phenyl, benzyl, phenyloxy and benzyloxy are optionally substituted with 1-3 substituents selected from the group consisting of halogen, C1-10 alkyl, C1-10 alkoxy, and cyano;
  • R2 is selected from the group consisting of hydrogen, C1-10 alkyl, C3-12 cycloalkyl and halogen, said alkyl optionally substituted with an oxo group;
  • and pharmaceutically acceptable salts thereof and solvates thereof.
  • In certain preferred embodiments of formula (I) or (II), Q is phenyl or a 6 membered heteroaromatic group containing 1-3 nitrogen atoms.
  • In certain preferred embodiments of formula (I), (II), (IA) or (IIA), the R1 alkyl is methyl, ethyl, propyl, butyl, pentyl, or hexyl.
  • In certain preferred embodiments of formula (I), (II), (IA) or (IIA), the R1 cycloalkyl is cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, or norbornyl.
  • In other preferred embodiments of formula (I), (II), (IA) or (IIA), the R1 bicyclic ring system is naphthyl. In other preferred embodiments of formula (I), (II), (IA) or (IIA), the R1 bicyclic ring system is tetrahydronaphthyl, or decahydronaphthyl and the R1 tricyclic ring system is dibenzocycloheptyl. In other preferred embodiments R1 is phenyl or benzyl.
  • In other preferred embodiments of formula (J), (II), (IA) or (IIA), the R1 bicyclic aromatic ring is a 10-membered ring, preferably quinoline or naphthyl.
  • In other preferred embodiments of formula (I), (II), (IA) or (IIA), the R1 bicyclic aromatic ring is a 9-membered ring, preferably indenyl.
  • In certain embodiments of formula (I), (II), (IA) or (IIA), Z is a bond, methyl, or ethyl.
  • In certain embodiments of formula (I), (II), (IA) or (IIA), the Z group is maximally substituted as not to have any hydrogen substitution on the base Z group. For example, if the base Z group is —CH2—, substitution with two methyl groups would remove hydrogens from the —CH2— base Z group.
  • In other preferred embodiments of formula (I), (II), (IA) or (IIA), n is 0.
  • In certain embodiments of formula (I), (II), (IA) or (IIA), X1 and X2 are both O.
  • In certain embodiments of formula (J) or (II), W is —CH2C(═O)NH2, —C(NH)NH2, pyridylmethyl, cyclopentyl, cyclohexyl, furanylmethyl, —C(═O)CH3, —CH2CH2NHC(═O)CH3, —SO2CH3, CH2CH2NHSO2CH3, furanylcarbonyl-, methylpyrrolylcarbonyl-, diazolecarbonyl-, azolemethyl-, trifluoroethyl-, hydroxyethyl-, cyanomethyl-, oxo-oxazolemethyl-, or diazolemethyl-.
  • In certain embodiments of formula (I) or (II), ZR1 is cyclohexylethyl-, cyclohexylmethyl-, cyclopentylmethyl-, dimethylcyclohexylmethyl-, phenylethyl-, pyrrolyltrifluoroethyl-, thienyltrifluoroethyl-, pyridylethyl-, cyclopentyl-, cyclohexyl-, methoxycyclohexyl-, tetrahydropyranyl-, propylpiperidinyl-, indolylmethyl-, pyrazoylpentyl-, thiazolylethyl-, phenyltrifluoroethyl-, hydroxyhexyl-, methoxyhexyl-, isopropoxybutyl-, hexyl-, or oxocanylpropyl-.
  • In certain embodiments of formula (J) or (II), at least one of ZR1 or W is —CH2COOV1, tetrazolylmethyl-, cyanomethyl-, NH2SO2methyl-, NH2SOmethyl-, aminocarbonylmethyl-, C1-4alkylaminocarbonylmethyl-, or diC1-4alkylaminocarbonylmethyl-.
  • In certain embodiments of formula (I) or (II), ZR1 is 3,3 diphenylpropyl optionally substituted at the 3 carbon of the propyl with —COOV1, tetrazolylC1-4alkyl-, cyano-, aminocarbonyl-, C1-4alkylaminocarbonyl-, or diC1-4alkylaminocarbonyl-.
  • In alternate embodiments ZR1 in formula (I), (II), (IA) or (IIA) can be
  • Figure US20090253727A1-20091008-C00010
  • wherein
  • Y1 is R3—(C1-C12)alkyl, R4-aryl, R5-heteroaryl, R6—(C3-C12)cyclo-alkyl, R7—(C3-C7)heterocycloalkyl, —CO2(C1-C6)alkyl, CN or —C(O)NR8R9; Y2 is hydrogen or Y; Y3 is hydrogen or (C1-C6)alkyl; or Y1, Y2 and Y3, together with the carbon to which they are attached, form one of the following structures:
  • Figure US20090253727A1-20091008-C00011
  • wherein r is 0 to 3; w and u are each 0-3, provided that the sum of w and u is 1-3; c and d are independently 1 or 2; s is 1 to 5; and ring E is a fused R4-phenyl or R5-heteroaryl ring;
  • R10 is 1 to 3 substituents independently selected from the group consisting of H, (C1-C6)alkyl, —OR8, —(C1-C6)alkyl-OR8, —NR8R9 and —(C1-C6)alkyl-NR8R9;
  • R11 is 1 to 3 substituents independently selected from the group consisting of R10, —CF3, —OCF3, NO2 and halo, or R11 substituents on adjacent ring carbon atoms may together form a methylenedioxy or ethylenedioxy ring;
  • R8 and R9 are independently selected from the group consisting of hydrogen, (C1-C6) alkyl, (C3-C12)cycloalkyl, aryl and aryl(C1-C6)alkyl;
  • R3 is 1 to 3 substituents independently selected from the group consisting of H, R4-aryl, R6—(C3-C12)cycloalkyl, R5-heteroaryl, R7—(C3-C7)heterocycloalkyl, —NR8R9, —OR12 and —S(O)0-2R12;
  • R6 is 1 to 3 substituents independently selected from the group consisting of H, (C1-C6)alkyl, R4-aryl, —NR8R9, —OR12 and —SR12;
  • R4 is 1 to 3 substituents independently selected from the group consisting of hydrogen, halo, (C1-C6)alkyl, R13-aryl, (C3-C12)cycloalkyl, —CN, —CF3, —OR8, —(C1-C6)alkyl-OR8, —OCF3, —NR8R9, —(C1-C6)alkyl —NR8R9, —NHSO2R8, —SO2N(R14)2, —SO2R8, —SOR8, —SR8, —NO2, —CONR8R9, —NR9COR8, —COR8, —COCF3, —OCOR8, —OCO2R8, —COOR8, —(C1-C6)alkyl-NHCOOC(CH3)3, —(C1-C6)alkyl-NHCOCF3, —(C1-C6)alkyl-NHSO2—(C1-C6)alkyl, —(C1-C6)alkyl-NHCONH—(C1-C6)-alkyl and
  • Figure US20090253727A1-20091008-C00012
  • wherein f is 0 to 6; or R4 substituents on adjacent ring carbon atoms may together form a methylenedioxy or ethylenedioxy ring;
  • R5 is 1 to 3 substituents independently selected from the group consisting of hydrogen, halo, (C1-C6)alkyl, R13-aryl, (C3-C12)cycloalkyl, —CN, —CF3, —OR8, —(C1-C6)alkyl-OR8, —OCF3, —NR8R9, —(C1-C6)alkyl-NR8R9, —NHSO2R8, —SO2N(R14)2, —NO2, —CONR8R9, —NR9COR8, —COR8, —OCOR8, —OCO2R8 and —COOR8;
  • R7 is H, (C1-C6)alkyl, —OR8, —(C1-C6)alkyl-OR8, —NR8R9 or —(C1-C6)alkyl-NR8R9;
  • R12 is H, (C1-C6)alkyl, R4-aryl, —(C1-C6)alkyl-OR8, —(C1-C6)alkyl-NR8R9, —(C1-C6)alkyl-SR8, or aryl(C1-C6)alkyl;
  • R13 is 1-3 substituents independently selected from the group consisting of H, (C1-C6)alkyl, (C1-C6)alkoxy and halo;
  • R14 is independently selected from the group consisting of H, (C1-C6)alkyl and R13—C6H4—CH2—.
  • As used herein, the term “alkyl” means a linear or branched saturated aliphatic hydrocarbon group having a single radical and 1-10 carbon atoms. Examples of alkyl groups include methyl, propyl, isopropyl, butyl, n-butyl, isobutyl, sec-butyl, tert-butyl, and pentyl. A branched alkyl means that one or more alkyl groups such as methyl, ethyl or propyl, replace one or both hydrogens in a —CH2— group of a linear alkyl chain. The term “lower alkyl” means an alkyl of 1-3 carbon atoms.
  • The term “alkoxy” means an “alkyl” as defined above connected to an oxygen radical.
  • The term “cycloalkyl” means a non-aromatic mono- or multicyclic hydrocarbon ring system having a single radical and 3-12 carbon atoms. Exemplary monocyclic cycloalkyl rings include cyclopropyl, cyclopentyl, and cyclohexyl. Exemplary multicyclic cycloalkyl rings include adamantyl and norbornyl.
  • The term “alkenyl” means a linear or branched aliphatic hydrocarbon group containing a carbon-carbon double bond having a single radical and 2-10 carbon atoms.
  • A “branched” alkenyl means that one or more alkyl groups such as methyl, ethyl or propyl replace one or both hydrogens in a —CH2— or —CH=linear alkenyl chain. Exemplary alkenyl groups include ethenyl, 1- and 2-propenyl, 1-, 2- and 3-butenyl, 3-methylbut-2-enyl, 2-propenyl, heptenyl, octenyl and decenyl.
  • The term “cycloalkenyl” means a non-aromatic monocyclic or multicyclic hydrocarbon ring system containing a carbon-carbon double bond having a single radical and 3 to 12 carbon atoms. Exemplary monocyclic cycloalkenyl rings include cyclopropenyl, cyclopentenyl, cyclohexenyl or cycloheptenyl. An exemplary multicyclic cycloalkenyl ring is norbornenyl.
  • The term “aryl” means a carbocyclic aromatic ring system containing one, two or three rings which may be attached together in a pendent manner or fused, and containing a single radical. Exemplary aryl groups include phenyl, naphthyl and acenaphthyl.
  • The term “heterocyclic” means cyclic compounds having one or more heteroatoms (atoms other than carbon) in the ring, and having a single radical. The ring may be saturated, partially saturated or unsaturated, and the heteroatoms may be selected from the group consisting of nitrogen, sulfur and oxygen. Examples of saturated heterocyclic radicals include saturated 3 to 6-membered hetero-monocyclic groups containing 1 to 4 nitrogen atoms, such as pyrrolidinyl, imidazolidinyl, piperidino, piperazinyl; saturated 3- to 6-membered hetero-monocyclic groups containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms, such as morpholinyl; saturated 3- to 6-membered hetero-monocyclic groups containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms, such as thiazolidinyl. Examples of partially saturated heterocyclic radicals include dihydrothiophene, dihydropyran, and dihydrofuran. Other heterocyclic groups can be 7 to 10 carbon rings substituted with heteroatoms such as oxocanyl and thiocanyl. When the heteroatom is sulfur, the sulfur can be a sulfur dioxide such as thiocanyldioxide.
  • The term “heteroaryl” means unsaturated heterocyclic radicals, wherein “heterocyclic” is as previously described. Exemplary heteroaryl groups include unsaturated 3 to 6 membered hetero-monocyclic groups containing 1 to 4 nitrogen atoms, such as pyrrolyl, pyridyl, pyrimidyl, and pyrazinyl; unsaturated condensed heterocyclic groups containing 1 to 5 nitrogen atoms, such as indolyl, quinolyl and isoquinolyl; unsaturated 3 to 6-membered hetero-monocyclic groups containing an oxygen atom, such as furyl; unsaturated 3 to 6 membered hetero-monocyclic groups containing a sulfur atom, such as thienyl; unsaturated 3 to 6 membered hetero-monocyclic groups containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms, such as oxazolyl; unsaturated condensed heterocyclic groups containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms, such as benzoxazolyl; unsaturated 3 to 6 membered hetero-monocyclic groups containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms, such as thiazolyl; and unsaturated condensed heterocyclic group containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms, such as benzothiazolyl. The term “heteroaryl” also includes unsaturated heterocyclic radicals, wherein “heterocyclic” is as previously described, in which the heterocyclic group is fused with an aryl group, in which aryl is as previously described. Exemplary fused radicals include benzofuran, benzodioxole and benzothiophene.
  • As used herein, the term “heterocyclicC1-4alkyl”, “heteroaromaticC1-4alkyl” and the like refer to the ring structure bonded to a C1-4 alkyl radical.
  • All of the cyclic ring structures disclosed herein can be attached at any point where such connection is possible, as recognized by one skilled in the art.
  • As used herein, the term “patient” includes a human or an animal such as a companion animal or livestock.
  • As used herein, the term “halogen” includes fluoride, bromide, chloride, iodide or alabamide.
  • The invention disclosed herein is meant to encompass all pharmaceutically acceptable salts thereof of the disclosed compounds. The pharmaceutically acceptable salts include, but are not limited to, metal salts such as sodium salt, potassium salt, cesium salt and the like; alkaline earth metals such as calcium salt, magnesium salt and the like; organic amine salts such as triethylamine salt, pyridine salt, picoline salt, ethanolamine salt, triethanolamine salt, dicyclohexylamine salt, N,N′-dibenzylethylenediamine salt and the like; inorganic acid salts such as hydrochloride, hydrobromide, sulfate, phosphate and the like; organic acid salts such as formate, acetate, trifluoroacetate, maleate, fumarate, tartrate and the like; sulfonates such as methanesulfonate, benzenesulfonate, p-toluenesulfonate, and the like; amino acid salts such as arginate, asparginate, glutamate and the like.
  • The invention disclosed herein is also meant to encompass all prodrugs of the disclosed compounds. Prodrugs are considered to be any covalently bonded carriers which release the active parent drug in vivo.
  • The invention disclosed herein is also meant to encompass the in vivo metabolic products of the disclosed compounds. Such products may result for example from the oxidation, reduction, hydrolysis, amidation, esterification and the like of the administered compound, primarily due to enzymatic processes. Accordingly, the invention includes compounds produced by a process comprising contacting a compound of this invention with a mammal for a period of time sufficient to yield a metabolic product thereof. Such products typically are identified by preparing a radiolabelled compound of the invention, administering it parenterally in a detectable dose to an animal such as rat, mouse, guinea pig, monkey, or to man, allowing sufficient time for metabolism to occur and isolating its conversion products from the urine, blood or other biological samples.
  • The invention disclosed herein is also meant to encompass the disclosed compounds being isotopically-labelled by having one or more atoms replaced by an atom having a different atomic mass or mass number. Examples of isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine and chlorine, such as 2H, 3H, 13C, 14C, 15N, 18O, 17O, 31P, 32P, 35S, 18F, and 36Cl, respectively. Some of the compounds disclosed herein may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms. The present invention is also meant to encompass all such possible forms as well as their racemic and resolved forms and mixtures thereof. When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended to include both E and Z geometric isomers. All tautomers are intended to be encompassed by the present invention as well
  • As used herein, the term “stereoisomers” is a general term for all isomers of individual molecules that differ only in the orientation of their atoms in space. It includes enantiomers and isomers of compounds with more than one chiral center that are not mirror images of one another (diastereomers).
  • The term “chiral center” refers to a carbon atom to which four different groups are attached.
  • The term “enantiomer” or “enantiomeric” refers to a molecule that is nonsuperimposeable on its mirror image and hence optically active wherein the enantiomer rotates the plane of polarized light in one direction and its mirror image rotates the plane of polarized light in the opposite direction.
  • The term “racemic” refers to a mixture of equal parts of enantiomers and which is optically inactive.
  • The term “resolution” refers to the separation or concentration or depletion of one of the two enantiomeric forms of a molecule.
  • The term “modulate” as used herein with respect to the ORL-1 receptor means the mediation of a pharmacodynamic response (e.g., analgesia) in a subject from (i) inhibiting or activating the receptor, or (ii) directly or indirectly affecting the normal regulation of the receptor activity. Compounds which modulate the receptor activity include agonists, antagonists, mixed agonists/antagonists and compounds which directly or indirectly affect regulation of the receptor activity.
  • Certain preferred compounds according to the invention of formulae (I) and (IA) include:
    • 1-[1-(naphth-2-yl-methyl)-4-piperidinyl]-2,1,3-benzothiadiazol-2,2-dione;
    • 1-[1-(p-phenylbenzyl)-4-piperidinyl]-2,1,3-benzothiadiazol-2,2-dione;
    • 1-[1-(3,3-Bis(phenyl)propyl)-4-piperidinyl]-2,1,3-benzothiadiazol-2,2-dione;
    • 1-[1-(p-benzyloxybenzyl)-4-piperidinyl]-2,1,3-benzothiadiazol-2,2-dione;
    • 1-[1-(4-propylcyclohexyl)-4-piperidinyl]-2,1,3-benzothiadiazol-2,2-dione;
    • 1-[1-(5-methylhex-2-yl)-4-piperidinyl]-2,1,3-benzothiadiazol-2,2-dione;
    • 1-[1-[4-(2-propyl)-cyclohexyl]-4-piperidinyl]-2,1,3-benzothiadiazol-2,2-dione;
    • 1-[1-(cyclooctylmethyl)-4-piperidinyl]-2,1,3-benzothiadiazol-2,2-dione;
    • 1-[1-(1,2,3,4-tetrahydro-2-naphthyl)-4-piperidinyl]-2,1,3-benzothiadiazol-2,2-dione;
    • 1-[1-(decahydro-2-naphthyl)-4-piperidinyl]-2,1,3-benzothiadiazol-2,2-dione;
    • 1-[1-(1,3,-dihydroinden-2-yl)-4-piperidinyl]-2,1,3-benzothiadiazol-2,2-dione;
    • 1-[1-(cyclooctyl)-4-piperidinyl]-2,1,3-benzothiadiazol-2,2-dione;
    • 1-[1-(naphth-2-yl-methyl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione;
    • 1-[1-(p-benzyloxybenzyl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione;
    • 1-[1-(p-phenylbenzyl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione;
    • 1-[1-(decahydro-2-naphthyl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione;
    • 1-[1-[4-(2-propyl)-cyclohexyl]-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione;
    • 1-[1-(4-propylcyclohexyl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione;
    • 1-[1-(benzyl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione;
    • 1-[1-(10,11-Dihydro-5H-dibenzo[a,d]-cyclohepten-5-yl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione;
    • 1-[1-(1,2,3,4 tetrahydro-2-naphthyl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione;
    • 1-[1-(5-methylhex-2-yl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione;
    • 1-[1-(norbornan-2-yl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione;
    • 1-[1-(cyclooctyl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione;
    • 1-[1-(1,3-dihydroinden-2-yl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione;
    • 1-[1-(3,3-Bis(phenyl)propyl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione; and
  • pharmaceutically acceptable salts thereof and solvates thereof.
  • Other preferred compounds of formula (I) include:
    • 3-butyl-1-[1-[4-(2-propyl)-cyclohexyl]-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione;
    • 3-acetamido-1-[1-[4-(2-propyl)-cyclohexyl]-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione;
    • 3-(2-methanesulfonamido)-1-[1-[4-(2-propyl)-cyclohexyl]-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione;
    • 3-methoxycarbonylmethyl-1-[1-[4-(2-propyl)-cyclohexyl]-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione;
    • 3-cyanomethyl-1-[1-[4-(2-propyl)-cyclohexyl]-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione;
    • 3-(2-hydroxyethyl)-1-[1-[4-(2-propyl)-cyclohexyl]-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione;
    • 3-butoxycarbonylmethyl-1-[1-[4-(2-propyl)-cyclohexyl]-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione; and
  • pharmaceutically acceptable salts thereof and solvates thereof.
  • Certain preferred compounds according to the invention of formula (II) and (IIA) include:
    • 1,2,3,4-tetrahydro-1-[1-(naphth-2-yl-methyl)-4-piperidinyl]-quinolin-2-one;
    • 1,2,3,4-tetrahydro-1-[1-(p-phenylbenzyl)-4-piperidinyl]-quinolin-2-one;
    • 1,2,3,4-tetrahydro-1-[1-[4,4-bis(4-fluorophenyl)butyl]-4-piperidinyl]-quinolin-2-one;
    • 1,2,3,4-tetrahydro-1-[1-(p-benzyloxybenzyl)-4-piperidinyl]-quinolin-2-one;
    • 1,2,3,4-tetrahydro-1-[1-(1,2,3,4-tetrahydro-2-naphthyl)-4-piperidinyl]-quinolin-2-one-;
    • 1,2,3,4-tetrahydro-1-[1-(4-propyl-cyclohexyl)-4-piperidinyl]-quinolin-2-one;
    • 1,2,3,4-tetrahydro-1-[1-(5-methylhex-2-yl)-4-piperidinyl]-quinolin-2-one;
    • 1,2,3,4-tetrahydro-1-[1-(norbornan-2-yl)-4-piperidinyl]-quinolin-2-one;
    • 1,2,3,4-tetrahydro-1-[1-(decahydro-2-naphthyl)-4-piperidinyl]-quinolin-2-one;
    • 1,2,3,4-tetrahydro-1-[1-(10,11-dihydro-5H-dibenzo[a,d]-cyclohepten-5-yl)-4-piperidinyl]-quinolin-2-one;
    • 1,2,3,4-tetrahydro-1-[1-(3,3-diphenylpropyl)-4-piperidinyl]-quinolin-2-one;
    • 1,2,3,4-tetrahydro-1-[1-(cyclooctyl)-4-piperidinyl]-quinolin-2-one;
    • 1,2,3,4-tetrahydro-1-[1-[4-(1-methylethyl)-cyclohexyl]-4-piperidinyl]-quinolin-2-one;
    • 1,2,3,4-tetrahydro-1-[1-(1,3-dihydroinden-2-yl)-4-piperidinyl]-quinolin-2-one;
    • 1,2,3,4-tetrahydro-1-[1-(cyclooctylmethyl)-4-piperidinyl]-quinolin-2-one; and
  • pharmaceutically acceptable salts thereof and solvates thereof.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The compounds of the present invention can be administered to anyone requiring modulation of the opioid and ORL1 receptors. Administration may be orally, topically, by suppository, inhalation, or parenterally.
  • The present invention also encompasses all pharmaceutically acceptable salts of the foregoing compounds. One skilled in the art will recognize that acid addition salts of the presently claimed compounds may be prepared by reaction of the compounds with the appropriate acid via a variety of known methods.
  • Various oral dosage forms can be used, including such solid forms as tablets, gelcaps, capsules, caplets, granules, lozenges and bulk powders and liquid forms such as emulsions, solution and suspensions. The compounds of the present invention can be administered alone or can be combined with various pharmaceutically acceptable carriers and excipients known to those skilled in the art, including but not limited to diluents, suspending agents, solubilizers, binders, disintegrants, preservatives, coloring agents, lubricants and the like.
  • When the compounds of the present invention are incorporated into oral tablets, such tablets can be compressed, tablet triturates, enteric-coated, sugar-coated, film-coated, multiply compressed or multiply layered. Liquid oral dosage forms include aqueous and nonaqueous solutions, emulsions, suspensions, and solutions and/or suspensions reconstituted from non-effervescent granules, containing suitable solvents, preservatives, emulsifying agents, suspending agents, diluents, sweeteners, coloring agents, and flavoring agents. When the compounds of the present invention are to be injected parenterally, they may be, e.g., in the form of an isotonic sterile solution. Alternatively, when the compounds of the present invention are to be inhaled, they may be formulated into a dry aerosol or may be formulated into an aqueous or partially aqueous solution.
  • In addition, when the compounds of the present invention are incorporated into oral dosage forms, it is contemplated that such dosage forms may provide an immediate release of the compound in the gastrointestinal tract, or alternatively may provide a controlled and/or sustained release through the gastrointestinal tract. A wide variety of controlled and/or sustained release formulations are well known to those skilled in the art, and are contemplated for use in connection with the formulations of the present invention. The controlled and/or sustained release may be provided by, e.g., a coating on the oral dosage form or by incorporating the compound(s) of the invention into a controlled and/or sustained release matrix.
  • Specific examples of pharmaceutically acceptable carriers and excipients that may be used to formulate oral dosage forms, are described in the Handbook of Pharmaceutical Excipients, American Pharmaceutical Association (1986). Techniques and compositions for making solid oral dosage forms are described in Pharmaceutical Dosage Forms: Tablets (Lieberman, Lachman and Schwartz, editors) 2nd edition, published by Marcel Dekker, Inc. Techniques and compositions for making tablets (compressed and molded), capsules (hard and soft gelatin) and pills are also described in Remington's Pharmaceutical Sciences (Arthur Osol, editor), 1553B1593 (1980). Techniques and composition for making liquid oral dosage forms are described in Pharmaceutical Dosage Forms: Disperse Systems, (Lieberman, Rieger and Banker, editors) published by Marcel Dekker, Inc.
  • When the compounds of the present invention are incorporated for parenteral administration by injection (e.g., continuous infusion or bolus injection), the formulation for parenteral administration may be in the form of suspensions, solutions, emulsions in oily or aqueous vehicles, and such formulations may further comprise pharmaceutically necessary additives such as stabilizing agents, suspending agents, dispersing agents, and the like. The compounds of the invention may also be in the form of a powder for reconstitution as an injectable formulation.
  • In certain embodiments, the compounds of the present invention can be used in combination with at least one other therapeutic agent. Therapeutic agents include, but are not limited to, μ-opioid agonists; non-opioid analgesics; non-steroid antiinflammatory agents; Cox-II inhibitors; antiemetics; β-adrenergic blockers; anticonvulsants; antidepressants; Ca2+-channel blockers; anticancer agent and mixtures thereof.
  • In certain embodiments, the compounds of the present invention can be formulated in a pharmaceutical dosage form in combination with a μ-opioid agonist. μ-opioid agonists, which may be included in the formulations of the present invention include but are not limited to include alfentanil, allylprodine, alphaprodine, anileridine, benzylmorphine, bezitramide, buprenorphine, butorphanol, clonitazene, codeine, desomorphine, dextromoramide, dezocine, diampromide, diamorphone, dihydrocodeine, dihydromorphine, dimenoxadol, dimepheptanol, dimethylthiambutene, dioxaphetyl butyrate, dipipanone, eptazocine, ethoheptazine, ethylmethylthiambutene, ethylmorphine, etonitazene fentanyl, heroin, hydrocodone, hydromorphone, hydroxypethidine, isomethadone, ketobemidone, levorphanol, levophenacylmorphan, lofentanil, meperidine, meptazinol, metazocine, methadone, metopon, morphine, myrophine, nalbuphine, narceine, nicomorphine, norlevorphanol, normethadone, nalorphine, normorphine, norpipanone, opium, oxycodone, oxymorphone, papavereturn, pentazocine, phenadoxone, phenomorphan, phenazocine, phenoperidine, piminodine, piritramide, proheptazine, promedol, properidine, propiram, propoxyphene, sufentanil, tilidine, tramadol, pharmaceutically acceptable salts thereof, and mixtures thereof.
  • In certain preferred embodiments, the μ-opioid agonist is selected from codeine, hydromorphone, hydrocodone, oxycodone, dihydrocodeine, dihydromorphine, morphine, tramadol, oxymorphone, pharmaceutically acceptable salts thereof, and mixtures thereof.
  • In another embodiment of the invention, the medicament comprises a mixture of a Cox-II inhibitor and an inhibitor of 5-lipoxygenase for the treatment of pain and/or inflammation. Suitable Cox-II inhibitors and 5-lipoxygenase inhibitors, as well as combinations thereof are described in U.S. Pat. No. 6,136,839, which is hereby incorporated by reference in its entirety. Cox-II inhibitors include, but are not limited to rofecoxib (Vioxx), celecoxib (Celebrex), DUP-697, flosulide, meloxicam, 6-MNA, L-745337, nabumetone, nimesulide, NS-398, SC-5766, T-614, L-768277, GR-253035, JTE-522, RS-57067-000, SC-58125, SC-078, PD-138387, NS-398, flosulide, D-1367, SC-5766, PD-164387, etoricoxib, valdecoxib and parecoxib or pharmaceutically acceptable salts, enantiomers or tautomers thereof.
  • The compounds of the present invention can also be combined in dosage forms with non-opioid analgesics, e.g., non-steroidal anti-inflammatory agents, including aspirin, ibuprofen, diclofenac, naproxen, benoxaprofen, flurbiprofen, fenoprofen, flubufen, ketoprofen, indoprofen, piroprofen, carprofen, oxaprozin, pramoprofen, muroprofen, trioxaprofen, suprofen, aminoprofen, tiaprofenic acid, fluprofen, bucloxic acid, indomethacin, sulindac, tolmetin, zomepirac, tiopinac, zidometacin, acemetacin, fentiazac, clidanac, oxpinac, mefenamic acid, meclofenamic acid, flufenamic acid, niflumic acid tolfenamic acid, diflurisal, flufenisal, piroxicam, sudoxicam or isoxicam, pharmaceutically acceptable salts thereof, and mixtures thereof. Other suitable non-opioid analgesics which may be included in the dosage forms of the present invention include the following, non-limiting, chemical classes of analgesic, antipyretic, nonsteroidal antiinflammatory drugs: salicylic acid derivatives, including aspirin, sodium salicylate, choline magnesium trisalicylate, salsalate, diflunisal, salicylsalicylic acid, sulfasalazine, and olsalazin; para-aminophennol derivatives including acetaminophen; indole and indene acetic acids, including indomethacin, sulindac, and etodolac; heteroaryl acetic acids, including tolmetin, diclofenac, and ketorolac; anthranilic acids (fenamates), including mefenamic acid, and meclofenamic acid; enolic acids, including oxicams (piroxicam, tenoxicam), and pyrazolidinediones (phenylbutazone, oxyphenthartazone); and alkanones, including nabumetone. For a more detailed description of the NSAIDs that may be included within the medicaments employed in the present invention, see Paul A. Insel Analgesic-Antipyretic and Antiinflammatory Agents and Drugs Employed in the treatment of Gout in Goodman & Gilman's The Pharmacological Basis of Therapeutics, 617-57 (Peny B. Molinhoff and Raymond W. Ruddon, Eds., Ninth Edition, 1996), and Glen R. Hanson Analgesic, Antipyretic and Anti-Inflammatory Drugs in Remington: The Science and Practice of Pharmacy Vol II, 1196-1221 (A. R. Gennaro, Ed. 19th Ed. 1995) which are hereby incorporated by reference in their entireties.
  • In certain embodiments, the compounds of the present invention can be formulated in a pharmaceutical dosage form in combination with antimigraine agents. Antimigraine agents include, but are not limited to, alpiropride, dihydroergotamine, dolasetron, ergocornine, ergocorninine, ergocryptine, ergot, ergotamine, flumedroxone acetate, fonazine, lisuride, lomerizine, methysergide oxetorone, pizotyline, and mixtures thereof.
  • The other therapeutic agent can also be an adjuvant to reduce any potential side effects such as, for example, an antiemetic agent. Suitable antiemetic agents include, but are not limited to, metoclopromide, domperidone, prochlorperazine, promethazine, chlorpromazine, trimethobenzamide, ondansetron, granisetron, hydroxyzine, acethylleucine monoethanolamine, alizapride, azasetron, benzquinamide, bietanautine, bromopride, buclizine, clebopride, cyclizine, dimenhydrinate, diphenidol, dolasetron, meclizine, methallatal, metopimazine, nabilone, oxyperndyl, pipamazine, scopolamine, sulpiride, tetrahydrocannabinols, thiethylperazine, thioproperazine, tropisetron, and mixtures thereof.
  • In certain embodiments, the compounds of the present invention can be formulated in a pharmaceutical dosage form in combination with β-adrenergic blockers. Suitable β-adrenergic blockers include, but are not limited to, acebutolol, alprenolol, amosulabol, arotinolol, atenolol, befunolol, betaxolol, bevantolol, bisoprolol, bopindolol, bucumolol, bufetolol, bufuralol, bunitrolol, bupranolol, butidrine hydrochloride, butofilolol, carazolol, carteolol, carvedilol, celiprolol, cetamolol, cloranolol, dilevalol, epanolol, esmolol, indenolol, labetalol, levobunolol, mepindolol, metipranolol, metoprolol, moprolol, nadolol, nadoxolol, nebivalol, nifenalol, nipradilol, oxprenolol, penbutolol, pindolol, practolol, pronethalol, propranolol, sotalol, sulfinalol, talinolol, tertatolol, tilisolol, timolol, toliprolol, and xibenolol.
  • In certain embodiments, the compounds of the present invention can be formulated in a pharmaceutical dosage form in combination with anticonvulsants. Suitable anticonvulsants include, but are not limited to, acetylpheneturide, albutoin, aloxidone, aminoglutethimide, 4-amino-3-hydroxybutyric acid, atrolactamide, beclamide, buramate, calcium bromide, carbamazepine, cinromide, clomethiazole, clonazepam, decimemide, diethadione, dimethadione, doxenitroin, eterobarb, ethadione, ethosuximide, ethotoin, felbamate, fluoresone, gabapentin, 5-hydroxytryptophan, lamotrigine, magnesium bromide, magnesium sulfate, mephenyloin, mephobarbital, metharbital, methetoin, methsuximide, 5-methyl-5-(3-phenantlu-yl)-hydantoin, 3-methyl-5-phenylhydantoin, narcobarbital, nimetazepam, nitrazepam, oxcarbazepine, paramethadione, phenacemide, phenetharbital, pheneturide, phenobarbital, phensuximide, phenylmethylbarbituric acid, phenyloin, phethenylate sodium, potassium bromide, pregabaline, primidone, progabide, sodium bromide, solanum, strontium bromide, suclofenide, sulthiame, tetrantoin, tiagabine, topiramate, trimethadione, valproic acid, valpromide, vigabatrin, and zonisamide.
  • In certain embodiments, the compounds of the present invention can be formulated in a pharmaceutical dosage form in combination with antidepressants. Suitable antidepressants include, but are not limited to, binedaline, caroxazone, citalopram, dimethazan, fencamine, indalpine, indeloxazine hydrocholoride, nefopam, nomifensine, oxitriptan, oxypertine, paroxetine, sertraline, thiazesim, trazodone, benmoxine, iproclozide, iproniazid, isocarboxazid, nialamide, octamoxin, phenelzine, cotinine, rolicyprine, rolipram, maprotiline, metralindole, mianserin, mirtazepine, adinazolam, amitriptyline, amitriptylinoxide, amoxapine, butriptyline, clomipramine, demexiptiline, desipramine, dibenzepin, dimetacrine, dothiepin, doxepin, fluacizine, imipramine, imipramine N-oxide, iprindole, lofepramine, melitracen, metapramine, nortriptyline, noxiptilin, opipramol, pizotyline, propizepine, protriptyline, quinupramine, tianeptine, trimipramine, adrafinil, benactyzine, bupropion, butacetin, dioxadrol, duloxetine, etoperidone, febarbamate, femoxetine, fenpentadiol, fluoxetine, fluvoxamine, hematoporphyrin, hypericin, levophacetoperane, medifoxamine, milnacipran, minaprine, moclobemide, nefazodone, oxaflozane, piberaline, prolintane, pyrisuccideanol, ritanserin, roxindole, rubidium chloride, sulpiride, tandospirone, thozalinone, tofenacin, toloxatone, tranylcypromine, L-tryptophan, venlafaxine, viloxazine, and zimeldine.
  • In certain embodiments, the compounds of the present invention can be formulated in a pharmaceutical dosage form in combination with Ca2+-channel blockers. Suitable Ca2+-channel blockers include, but are not limited to, bepridil, clentiazem, diltiazem, fendiline, gallopamil, mibefradil, prenylamine, semotiadil, terodiline, verapamil, amlodipine, aranidipine, barnidipine, benidipine, cilnidipine, efonidipine, elgodipine, felodipine, isradipine, lacidipine, lercanidipine, manidipine, nicardipine, nifedipine, nilvadipine, nimodipine, nisoldipine, nitrendipine, cinnarizine, flunarizine, lidoflazine, lomerizine, bencyclane, etafenone, fantofarone, and perhexyline.
  • In certain embodiments, the compounds of the present invention can be formulated in a pharmaceutical dosage form in combination with anticancer agents. Suitable anticancer agents include, but are not limited to, acivicin; aclarubicin; acodazole hydrochloride; acronine; adozelesin; aldesleukin; altretamine; ambomycin; ametantrone acetate; aminoglutethimide; amsacrine; anastrozole; antlramycin; asparaginase; asperlin; azacitidine; azetepa; azotomycin; batimastat; benzodepa; bicalutamide; bisantrene hydrochloride; bisnafide dimesylate; bizelesin; bleomycin sulfate; brequinar sodium; bropirimine; busulfan; cactinomycin; calusterone; caracemide; carbetimer; carboplatin; carmustine; carubicin hydrochloride; carzelesin; cedefingol; chlorambucil; cirolemycin; cisplatin; cladribine; crisnatol mesylate; cyclophosphamide; cytarabine; dacarbazine; dactinomycin; daunorubicin hydrochloride; decitabine; dexormaplatin; dezaguanine; dezaguanine mesylate; diaziquone; docetaxel; doxorubicin; doxorubicin hydrochloride; droloxifene; droloxifene citrate; dromostanolone propionate; duazomycin; edatrexate; eflornithine hydrochloride; elsamitrucin; enloplatin; enpromate; epipropidine; epirubicin hydrochloride; erbulozole; esorubicin hydrochloride; estramustine; estramustine phosphate sodium; etanidazole; etoposide; etoposide phosphate; etoprine; fadrozole hydrochloride; fazarabine; fenretinide; floxuridine; fludarabine phosphate; fluorouracil; fluorocitabine; fosquidone; fostriecin sodium; gemcitabine; gemcitabine hydrochloride; hydroxyurea; idarubicin hydrochloride; ifosfamide; ilmofosine; interleukin II (including recombinant interleukin II, or rIL2), interferon alfa-2a; interferon alfa-2b; interferon alfa-n1; interferon alfa-n3; interferon beta-I a; interferon gamma-I b; iproplatin; irinotecan hydrochloride; lanreotide acetate; letrozole; leuprolide acetate; liarozole hydrochloride; lometrexol sodium; lomustine; losoxantrone hydrochloride; masoprocol; maytansine; mechlorethamine hydrochloride; megestrol acetate; melengestrol acetate; melphalan; menogaril; mercaptopurine; methotrexate; methotrexate sodium; metoprine; meturedepa; mitindomide; mitocarcin; mitocromin; mitogillin; mitomalcin; mitomycin; mitosper; mitotane; mitoxantrone hydrochloride; mycophenolic acid; nocodazole; nogalamycin; ormaplatin; oxisuran; paclitaxel; pegaspargase; peliomycin; pentamustine; peplomycin sulfate; perfosfamide; pipobroman; piposulfan; piroxantrone hydrochloride; plicamycin; plomestane; porfimer sodium; porfiromycin; prednimustine; procarbazine hydrochloride; puromycin; puromycin hydrochloride; pyrazofurin; riboprine; rogletimide; safingol; safingol hydrochloride; semustine; simtrazene; sparfosate sodium; sparsomycin; spirogermanium hydrochloride; spiromustine; spiroplatin; streptonigrin; streptozocin; sulofenur; talisomycin; tecogalan sodium; tegafur; teloxantrone hydrochloride; temoporfin; teniposide; teroxirone; testolactone; thiamiprine; thioguanine; thiotepa; tiazofurin; tirapazamine; toremifene citrate; trestolone acetate; triciribine phosphate; trimetrexate; trimetrexate glucuronate; triptorelin; tubulozole hydrochloride; uracil mustard; uredepa; vapreotide; verteporfin; vinblastine sulfate; vincristine sulfate; vindesine; vindesine sulfate; vinepidine sulfate; vinglycinate sulfate; vinleurosine sulfate; vinorelbine tartrate; vinrosidine sulfate; vinzolidine sulfate; vorozole; zeniplatin; zinostatin; zorubicin hydrochloride. Other anti-cancer drugs include, but are not limited to: 20-epi-1,25 dihydroxyvitamin D3; 5-ethynyluracil; abiraterone; aclarubicin; acylfulvene; adecypenol; adozelesin; aldesleukin; ALL-TK antagonists; altretamine; ambamustine; amidox; amifostine; aminolevulinic acid; amrubicin; amsacrine; anagrelide; anastrozole; andrographolide; angiogenesis inhibitors; antagonist D; antagonist G; antarelix; anti-dorsalizing morphogenetic protein-1; antiandrogen, prostatic carcinoma; antiestrogen; antineoplaston; antisense oligonucleotides; aphidicolin glycinate; apoptosis gene modulators; apoptosis regulators; apurinic acid; ara-CDP-DL-PTBA; arginine deaminase; asulacrine; atamestane; atrimustine; axinastatin 1; axinastatin 2; axinastatin 3; azasetron; azatoxin; azatyrosine; baccatin III derivatives; balanol; batimastat; BCR/ABL antagonists; benzochlorins; benzoylstaurosporine; beta lactam derivatives; beta-alethine; betaclamycin B; betulinic acid; bFGF inhibitor; bicalutamide; bisantrene; bisaziridinylspermine; bisnafide; bistratene A; bizelesin; breflate; bropirimine; budotitane; buthionine sulfoximine; calcipotriol; calphostin C; camptothecin derivatives; canarypox IL-2; capecitabine; carboxamide-amino-triazole; carboxyamidotriazole; CaRest M3; CARN 700; cartilage derived inhibitor; carzelesin; casein kinase inhibitors (ICOS); castanospermine; cecropin B; cetrorelix; chlorlns; chloroquinoxaline sulfonamide; cicaprost; cis-porphyrin; cladribine; clomifene analogues; clotrimazole; collismycin A; collismycin B; combretastatin A4; combretastatin analogue; conagenin; crambescidin 816; crisnatol; cryptophycin 8; cryptophycin A derivatives; curacin A; cyclopentantlraquinones; cycloplatam; cypemycin; cytarabine ocfosfate; cytolytic factor; cytostatin; dacliximab; decitabine; dehydrodidemnin B; deslorelin; dexamethasone; dexifosfamide; dexrazoxane; dexverapamil; diaziquone; didemnin B; didox; diethylnorspermine; dihydro-5-azacytidine; dihydrotaxol, 9-; dioxamycin; diphenyl spiromustine; docetaxel; docosanol; dolasetron; doxifluridine; droloxifene; dronabinol; duocarmycin SA; ebselen; ecomustine; edelfosine; edrecolomab; eflornithine; elemene; emitefur; epirubicin; epristeride; estramustine analogue; estrogen agonists; estrogen antagonists; etanidazole; etoposide phosphate; exemestane; fadrozole; fazarabine; felmetinide; filgrastim; finasteride; flavopiridol; flezelastine; fluasterone; fludarabine; fluorodaunorunicin hydrochloride; forfenimex; formestane; fostriecin; fotemustine; gadolinium texaphyrin; gallium nitrate; galocitabine; ganirelix; gelatinase inhibitors; gemcitabine; glutathione inhibitors; hepsulfam; heregulin; hexamethylene bisacetamide; hypericin; ibandronic acid; idarubicin; idoxifene; idramantone; ilmofosine; ilomastat; imidazoacridones; imiquimod; immunostimulan-t peptides; insulin-like growth factor-1 receptor inhibitor; interferon agonists; interferons; interleukins; iobenguane; iododoxorubicin; ipomeanol, 4-; iroplact; irsogladine; isobengazole; isohomohalicondrin B; itasetron; jasplakinolide; kahalalide F; lamellarin-N triacetate; lanreotide; leinamycin; lenograstim; lentinan sulfate; leptolstatin; letrozole; leukemia inhibiting factor; leukocyte alpha interferon; leuprolide+estrogen+progesterone; leuprorelin; levamisole; liarozole; linear polyamine analogue; lipophilic disaccharide peptide; lipophilic platinum compounds; lissoclinamide 7; lobaplatin; lombricine; lometrexol; lonidamine; losoxantrone; lovastatin; loxoribine; lurtotecan; lutetium texaphyrin; lysofylline; lytic peptides; maitansine; mannostatin A; marimastat; masoprocol; maspin; matrilysin inhibitors; matrix metalloproteinase inhibitors; menogaril; merbarone; meterelin; methioninase; metoclopramide; MIF inhibitor; mifepristone; miltefosine; mirimostim; mismatched double stranded RNA; mitoguazone; mitolactol; mitomycin analogues; mitonafide; mitotoxin fibroblast growth factor-saporin; mitoxantrone; mofarotene; molgramostim; monoclonal antibody, human chorionic gonadotrophin; monophosphoryl lipid A+myobacterium cell wall sk; mopidamol; multiple drug resistance gene inhibitor; multiple tumor suppressor 1-based therapy; mustard anticancer agent; mycaperoxide B; mycobacterial cell wall extract; myriaporone; N-acetyldinaline; N-substituted benzamides; nafarelin; nagrestip; naloxone+pentazocine; napavin; naphterpin; nartograstim; nedaplatin; nemorubicin; neridronic acid; neutral endopeptidase; nilutamide; nisamycin; nitric oxide modulators; nitroxide antioxidant; nitrullyn; O6-benzylguanine; octreotide; okicenone; oligonucleotides; onapristone; ondansetron; ondansetron; oracin; oral cytokine inducer; ormaplatin; osaterone; oxaliplatin; oxaunomycin; paclitaxel; paclitaxel analogues; paclitaxel derivatives; palauamine; palmitoylrhizoxin; pamidronic acid; panaxytriol; panomifene; parabactin; pazelliptine; pegaspargase; peldesine; pentosan polysulfate sodium; pentostatin; pentrozole; perflubron; perfosfamide; perillyl alcohol; phenazinomycin; phenylacetate; phosphatase inhibitors; picibanil; pilocarpine hydrochloride; pirarubicin; piritrexim; placetin A; placetin B; plasminogen activator ihlaibitor; platinum complex; platinum compounds; platinum-triamine complex; porfimer sodium; porfiromycin; prednisone; propyl bis-acridone; prostaglandin J2; proteasome inhibitors; protein A-based immune modulator; protein kinase C inhibitor; protein kinase C inhibitors, microalgal; protein tyrosine phosphatase inhibitors; purine nucleoside phosphorylase ihliibitors; purpurins; pyrazoloacridine; pyridoxylated hemoglobin polyoxyethylene conjugate; raf antagonists; raltitrexed; ramosetron; ras farnesyl protein transferase inhibitors; ras inhibitors; ras-GAP inhibitor; retelliptine demethylated; rhenium Re 186 etidronate; rhizoxin; ribozymes; RII retinamide; rogletimide; rohitukine; romurtide; roquinimex; rubiginone B1; ruboxyl; safingol; saintopin; SarCNU; sarcophytol A; sargramostim; Sdi 1 mimetics; semustine; senescence derived inhibitor 1; sense oligonucleotides; signal transduction inhibitors; signal transduction modulators; single chain antigen binding protein; sizofuran; sobuzoxane; sodium borocaptate; sodium phenylacetate; solverol; somatomedin binding protein; sonermin; sparfosic acid; spicamycin D; spiromustine; splenopentin; spongistatin 1; squalamine; stem cell inhibitor; stem-cell division inhibitors; stipiamide; stromelysin inhibitors; sulfinosine; superactive vasoactive intestinal peptide antagonist; suradista; suramin; swainsonine; synthetic glycosaminoglycans; tallimustine; tamoxifen methiodide; tauromustine; tazarotene; tecogalan sodium; tegafur; tellurapyrylium; telomerase inhibitors; temoporfin; temozolomide; teniposide; tetrachlorodecaoxide; tetrazomine; thaliblastine; thiocoraline; thirombopoietin; thrombopoietin mimetic; thymalfasin; thymopoietin receptor agonist; thymotrinan; thyroid stimulating hormone; tin ethyl etiopurpurin; tirapazamine; titanocene bichloride; topsentin; toremifene; totipotent stem cell factor; translation inhibitors; tretinoin; triacetyluridine; triciribine; trimetrexate; triptorelin; tropisetron; turosteride; tyrosine kinase inhibitors; tyrphostins; UBC inhibitors; ubenimex; urogenital sinus-derived growth inhibitory factor; urokinase receptor antagonists; vapreotide; variolin B; vector system, erythrocyte gene therapy; velaresol; veramine; verdins; verteporfin; vinorelbine; vinxaltine; vitaxin; vorozole; zanoterone; zeniplatin; zilascorb; and zinostatin stimalamer.
  • The compounds of the present invention and the other therapeutic agent can act additively or, more preferably, synergistically. In a preferred embodiment, a composition comprising a compounds of the present invention is administered concurrently with the administration of another therapeutic agent, which can be part of the same composition or in a different composition from that comprising the compounds of the present invention. In another embodiment, a composition comprising the compounds of the present invention is administered prior to or subsequent to administration of another therapeutic agent.
  • The compounds of the present invention when administered, e.g., via the oral, parenteral or topical routes to mammals, can be in a dosage in the range of about 0.01 mg/kg to about 3000 mg/kg body weight of the patient per day, preferably about 0.01 mg/kg to about 1000 mg/kg body weight per day administered singly or as a divided dose. However, variations will necessarily occur depending upon the weight and physical condition (e.g., hepatic and renal function) of the subject being treated, the affliction to be treated, the severity of the symptoms, the route of administration, the frequency of the dosage interval, the presence of any deleterious side-effects, and the particular compound utilized, among other things.
  • The compounds of the present invention preferably have a binding affinity Ki for the human ORL-1 receptor of about 500 nM or less; 100 nM or less; 50 nM or less; 20 nM or less or 5 nM or less. The binding affinity Ki can be measured by one skilled in the art by an assay utilizing membranes from recombinant HEK-293 cells expressing the human opioid receptor-like receptor (ORL-1) as described below.
  • The following examples illustrate various aspects of the present invention, and are not to be construed to limit the claims in any manner whatsoever.
  • Example 1 Synthesis of “5-Membered SO2” Head Groups
  • Figure US20090253727A1-20091008-C00013
  • Procedure:
  • 1,2-Phenylenediamine 1 (160 g, 1.50 mol) and 4-oxo-piperidine-tert-butylester 2 (100 g, 0.50 mol) were dissolved in 1,2-dichloroethane (2.0 L) with stirring. Acetic acid (31.6 mL) was added, followed by sodium triacetoxyborohydride (148 g, 0.70 mol) and the resulting mixture stirred at room temperature for 18 hr. The solvent was evaporated and the residue partitioned between ether and 1 M acetic acid. The organic layer was separated and washed with 1M acetic acid (3×) followed by sodium bicarbonate solution (1×). The aqueous phases were back extracted with ether (1×) and the combined organic extracts dried over MgSO4, filtered and the solvent evaporated to give an orange gum. Addition of 600 mL of ether/hexane (1:1) followed by seeding induced crystallization. After 15 min. the mixture was filtered, washed with 300 mL of ether/hexane (1:1) to give pure 3 as a white solid (79.2 g, 53%).
  • m.p.=107.1-107.6° C. 1H-NMR (CDCl3): d 1.30 (bd, 2H), 1.40 (s, 9H), 1.95 (bd, 2H), 3.90 (bt, 2H), 3.25 (b, 2H), 3.30 (m, 1H), 3.95 (b, 2H), 6.60-6.80 (m, 4H).
  • The diamine 3 (5.4 g, 18.6 mmol) was dissolved in dry pyridine (30 mL). Sulfamide (3.58 g, 37.2 mmol) was added and the mixture heated to reflux for 2 h. The mixture was cooled to room temperature and the solvent evaporated to dryness. The residue was partitioned between dichloromethane:methanol (10:1, 500 mL) and 0.1 M hydrochloric acid (500 mL), the organic phase separated, washed with brine (500 mL), dried over MgSO4, filtered and the solvent evaporated. The residue was triturated with ethyl acetate to give pure 4 as a pale pink solid (5.15 g, 79%).
  • m.p.=204.7°-205.4° C. 1H-NMR (DMSO): d 1.30 (s, 9H), 1.80-1.95 (m, 4H), 2.80 (b, 2H), 3.95 (m, 2H), 4.10 (m, 1H), 6.70-7.00 (m, 4H), 11.25 (bs, 1H).
  • Compound 4 (5.15 g, 14.57 mmol) was suspended in 100 mL of ethyl acetate, 20 mL of a 1:1 mixture of concentrated hydrochloric acid/ethyl acetate was added and the suspension stirred at room temperature for 2 h. The mixture was filtered and the filtrate washed with ethyl acetate to give pure 5 (HCl salt) as a pale pink solid (3.82 g, 91%).
  • 1H-NMR (DMSO) (HCl salt): d 2.10 (m, 2H), 2.50 (m, 2H), 3.12 (m, 2H), 3.52 (m, 2H), 4.40 (m, 1H), 5.80 (b, 1H), 6.90-7.20 (m, 3H), 7.35 (m, 1H), 9.20 (b, 1H), 9.40 (b, 1H), 11.70 (b, 1H). Elemental analysis: C11H15N3O2S.HCl.0.75H2O requires: C, 43.56; H, 5.81; N, 13.85. found: C, 43.90; H, 5.78; N, 13.51.
  • Example 2 Synthesis of “6-Membered SO2” Head Groups
  • Figure US20090253727A1-20091008-C00014
  • Procedure:
  • Anthranilamide 6 (15.0 g, 110.1 mmol) and N-benzyl-4-piperidone (20.96 g, 110.1 mmol) were dissolved in glacial acetic acid (150 mL) with stirring under nitrogen over 15 min. Sodium triacetoxyborohydride (35.14 g, 165.8 mmol) was added in portions and the resulting mixture stirred at room temperature for 60 hr. The mixture was poured into water and extracted with ethyl acetate (1×). The ethyl acetate extract was back extracted with water (3×). The combined aqueous extracts were carefully basified with sodium hydroxide pellets to pH 12 and the mixture filtered to give a white solid that was triturated with acetone to give 7 as a white crystalline solid (19.8 g, 58%).
  • mp=249.8-250.7° C. (dec.).
  • To a suspension of lithium aluminium hydride (3.58 g, 96.7 mmol) in 50 mL of dry dioxane was added dropwise a suspension of 7 (10.0 g, 32.3 mmol) in 100 mL of dry dioxane. The resulting mixture was stirred for 1 hr at room temperature then heated to reflux overnight. The mixture was cooled to room temperature and cautiously quenched with water over 1 hr. Magnesium sulfate (ca 20 g) was added, the mixture filtered through Celite, and the filter cake washed with dichloromethane. The filtrate was dried over MgSO4, filtered and evaporated. The residue was triturated with ether to give pure 8 as a white solid (6.70 g, 71%).
  • mp=116-118° C. 1H-NMR (CDCl3): d 1.60 (m, 2H), 2.10 (m, 2H), 2.25 (bt, 2H), 2.85 (b, 2H), 3.40 (m, 1H), 3.60 (s, 2H), 3.91 (s, 2H), 6.63 (m, 2H), 7.05 (d, 1H), 7.20 (m, 1H), 7.25-7.4 (m, 5H).
  • To a solution of 8 (7.00 g, 23.7 mmol) in 50 mL of pyridine was added sulfamide (4.55 g, 47.4 mmol) and the resulting solution heated to reflux for 18 hr. After cooling to room temperature the mixture was evaporated to give a brown gum. This was partitioned between chloroform and 1M potassium carbonate solution and the organic phase separated. The aqueous phase was extracted with chloroform (2×) and the combined organic extracts dried over MgSO4, filtered and evaporated. The residue was chromatographed to give a pale yellow foam (6.00 g). This was dissolved in 60 mL of ethyl acetate and 4 mL of a 1:1 mixture of concentrated hydrochloric acid: ethyl acetate was added. The mixture was allowed to stand for 30 min until complete crystallization had occurred. The mixture was filtered and the solid washed with ethyl acetate to give pure 9 as a white crystalline solid (6.28 g, 67%).
  • mp=248-249.9° C. 1H-NMR (DMSO) (HCl salt): d 2.05 (m, 2H), 2.32 (m, 2H), 3.12 (m, 2H), 3.40 (b, 2H), 4.30 (b, 3H), 4.45 (b, 2H), 7.10-7.30 (m, 4H), 7.40-7.60 (m, 5H), 7.85 (m, 1H).
  • Compound 9 (4.0 g, 11 mmol) was hydrogenated over Pd/C in 100 mL of methanol/water (3:1) for 24 hr. Filtration and evaporation gave a residue that was triturated with ethyl acetate/methanol (1:1) to give pure 10 as a white crystalline solid (2.72 g, 89%).
  • MS: m/z 268.1 (M+1). 1H-NMR (CDCl3) (HCl salt): d 2.00 (m, 4H), 2.70 (m, 2H), 3.2 (m, 2H), 4.15 (m, 1H), 4.52 (s, 2H), 7.15-7.35 (m, 5H). Elemental analysis: C12H17N3O2S.HCl.0.4H2O requires: C, 46.34; H, 6.09; N, 13.51. found: C, 46.36; H, 5.88; N, 13.37.
  • Example 3
  • Attachments of Tail Groups to the “5-Membered SO2” and “6-Membered SO2” Head Groups
  • Tail groups were attached to the head groups according to the following procedures:
  • Figure US20090253727A1-20091008-C00015
  • General Procedure for Alkylation:
  • To a solution of the amine (1 eq) and triethylamine (1 eq) in dimethylformamide, was added 1 eq of alkyl bromide or chloride in one portion. The mixture was stirred and heated at 80° C. over night. TLC indicated the reaction was complete. The reaction was quenched by the addition of water followed by 1 N NaOH to pH 10. The mixture was extracted 2× with Et2O. The combined organic extracts were dried over potassium carbonate and the solvent evaporated, followed by chromatography to give the pure product.
  • General Procedure for Reductive Amination:
  • To a mixture of ketone or aldehyde (1 eq), amine (1 eq), and acetic acid (1 eq) in methanol, was added sodium cyanoborohydride (1.4 eq) in one portion. The mixture was stirred over night at room temperature. TLC indicated the reaction was complete. The reaction was quenched by the addition of water followed by 1 N NaOH to pH 10. The mixture was extracted 2× with Et2O. The combined organic extracts were dried over potassium carbonate and the solvent evaporated, followed by chromatography to give the pure product. The following compounds were prepared by attaching the tail groups using the general procedures described:
  • 1-[1-(naphth-2-yl-methyl)-4-piperidinyl]-2,1,3-benzothiadiazol-2,2-dione
  • LC: 93.2%. MS: m/z 394.2 (M+1). 1H-NMR (DMSO): d 1.95 (b, 2H), 2.22 (m, 2H), 2.40 (b, 2H), 3.10 (m, 2H), 3.80-4.00 (b, 3H), 6.60-6.80 (m, 4H), 7.50 (m, 3H), 7.90 (m, 4H), 10.8 (b, 1H).
  • 1-[1-(p-phenylbenzyl)-4-piperidinyl]-2,1,3-benzothiadiazol-2,2-dione
  • LC: 96.4%. MS: m/z 420.6 (M+1). 1H-NMR (CDCl3): d 2.07 (m, 2H), 2.30 (m, 2H), 2.48 (m, 2H), 3.15 (m, 2H), 3.65 (s, 2H), 3.90 (m, 1H), 6.78-6.95 (m, 4H), 7.30-7.60 (m, 9H).
  • 1-[1-(3,3-Bis(phenyl)propyl)-4-piperidinyl]-2,1,3-benzothiadiazol-2,2-dione
  • LC: 100%. MS: m/z 448.2 (M+1). 1H-NMR (DMSO): d 2.00 (b, 2H), 2.20-2.40 (m, 4H), 2.60-2.85 (m, 4H), 3.20-3.50 (m, 2H), 3.90 (bt, 1H), 4.00 (t, 1H), 6.40-6.60 (m, 4H), 7.18 (m, 2H), 7.25-7.40 (m, 8H).
  • 1-[1-(p-benzyloxybenzyl)-4-piperidinyl]-2,1,3-benzothiadiazol-2,2-dione
  • LC: 98%. MS: m/z 450.7 (M+1). 1H-NMR (CDCl3): d 2.23 (m, 2H), 2.75 (m, 2H), 2.90 (m, 2H), 3.62 (s, 2H), 3.85 (m, 2H), 4.12 (m, 1H), 5.10 (s, 2H), 6.90-7.48 (m, 13H).
  • 1-[1-(1,2,3,4-tetrahydro-2-naphthyl)-4-piperidinyl]-2,1,3-benzothiadiazol-2,2-dione
  • LC: 100%. MS: m/z 384.6 (M+1). 1H-NMR (DMSO-d6): d 1.20-2.60 (m, 10H), 2.75-3.10 (m, 5H), 3.90 (m, 1H), 6.40 (b, 2H), 6.55 (b, 2H), 7.10 (b, 4H).
  • 1-[1-(4-propylcyclohexyl)-4-piperidinyl]-2,1,3-benzothiadiazol-2,2-dione
  • LC: 92.7%. MS: m/z 378.3 (M+1). 1H-NMR (DMSO-d6): d 0.85 (t, 3H), 1.15 (m, 1H), 1.30 (m, 4H), 1.35-1.55 (m, 2H), 1.55-1.65 (m, 4H), 1.70 (b, 1H), 1.85 (b, 1H), 1.90-2.10 (m, 2H), 2.35-2.50 (b, 2H), 3.00 (b, 3H), 3.55 (b, 2H), 3.92 (m, 1H), 6.30 (m, 2H), 6.50 (m, 2H), 10.1 (b, 1H).
  • 1-[1-(5-methylhex-2-yl)-4-piperidinyl]-2,1,3-benzothiadiazol-2,2-dione
  • LC: 100%. MS: m/z 352.3 (M+1). 1H-NMR (DMSO): d 0.85 (m, 6H), 1.10-1.30 (m, 6H), 1.40 (m, 1H), 1.50 (m, 1H), 1.70 (b, 1H), 2.05 (bd, 2H), 2.45 (m, 2H), 2.90-3.20 (m, 3H), 3.90-4.10 (m, 2H), 6.30 (m, 2H), 6.50 (m, 2H).
  • 1-[1-(decahydro-2-naphthyl)-4-piperidinyl]-2,1,3-benzothiadiazol-2,2-dione
  • LC: 95%. MS: m/z 390.7 (M+1). 1H-NMR (DMSO-d6): d 0.8-2.10 (m, 20H), 2.90-3.60 (m, 5H), 3.95 (m, 11H), 6.30 (b, 2H), 6.50 (b, 2H).
  • 1-[1-(cyclooctyl)-4-piperidinyl]-2,1,3-benzothiadiazol-2,2-dione
  • LC: 100%. MS: m/z 364.3 (M+1). 1H-NMR (DMSO-d6): d 1.30-2.10 (m, 18H), 2.35-2.55 (m, 2H), 2.90-3.40 (m, 3H), 4.00 (m, 1H), 6.30 (m, 2H), 6.50 (m, 2H).
  • 1-[1-[4-(2-propyl)-cyclohexyl]-4-piperidinyl]-2,1,3-benzothiadiazol-2,2-dione
  • LC: 95.1%. MS: m/z 378.3 (M+1). 1H-NMR (DMSO-d6): d 0.80-0.95 (m, 6H), 1.03 (b, 1H), 1.15 (m, 1H), 1.30-1.50 (m, 2H), 1.55-1.90 (m, 6H), 2.05 (b, 2H), 2.30-2.50 (m, 2H), 2.80-3.20 (b, 3H), 3.40-3.60 (b, 2H), 3.95 (m, 1H), 6.38 (m, 2H), 6.45 (m, 2H), 7.00 (b, 1H).
  • 1-[1-(1,3,-dihydroinden-2-yl)-4-piperidinyl]-2,1,3-benzothiadiazol-2,2-dione
  • LC: 96.5%. MS: m/z 370.6 (M+1). 1H-NMR (DMSO-d6): d 2.00 (b, 2H), 2.30 (m, 2H), 2.70 (m, 2H), 3.10 (m, 2H), 3.20-3.60 (m, 5H), 3.90 (m, 1H), 6.40-6.65 (m, 4H), 7.10-7.30 (m, 4H).
  • 1-[1-(cyclooctylmethyl)-4-piperidinyl]-2,1,3-benzothiadiazol-2,2-dione
  • LC: 100%. MS: m/z 392.7 (M+15). 1H-NMR (MeOH): d 1.30-1.80 (m, 14H), 2.05 (m, 1H), 2.22 (m, 2H), 2.50 (m, 2H), 2.85 (m, 2H), 3.00 (m, 2H), 3.60 (m, 2H), 4.30 (m, 1H), 4.50 (s, 2H), 7.10-7.40 (m, 4H).
  • 1-[1-(benzyl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione
  • 1H-NMR (DMSO-d6) of HCl Salt form: d 2.05 (m, 2H), 2.32 (m, 2H), 3.12 (m, 2H), 3.40 (b, 2H), 4.30 (b, 3H), 4.45 (b, 2H), 7.10-7.30 (m, 4H), 7.40-7.60 (m, 5H), 7.85 (m, 1H).
  • 1-[1-(naphth-2-yl-methyl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione
  • LC: 100%. MS: m/z 408.3 (M+1) 1H-NMR (CDCl3): d 1.95 (b, 2H), 2.20 (m, 4H), 3.05 (m, 2H), 3.70 (s, 2H), 4.10 (m, 1H), 4.50 (s, 2H), 7.10 (m, 2H), 7.20 (m, 1H), 7.30 (m, 1H), 7.45 (m, 3H), 7.75 (s, 1H), 7.85 (m, 3H).
  • 1-[1-(p-phenylbenzyl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione
  • LC: 92.3%. MS: m/z 434.1 (M+1). 1H-NMR (CDCl3): d 1.95-2.30 (m, 6H), 3.10 (b, 2H), 3.65 (s, 2H), 4.10 (M, 1H), 4.50 (s, 2H), 7.10-7.70 (m, 13H).
  • 1-[1-(10,11-Dihydro-5H-dibenzo[a,d]-cyclohepten-5-yl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione
  • LC: 100%. MS: m/z 482.2. 1H-NMR (DMSO): d 1.75 (b, 4H), 1.92 (m, 2H), 2.60-2.80 (m, 4H), 3.82 (m, 3H), 4.00 (s, 1H), 4.30 (s, 2H), 7.00-7.40 (M, 12H).
  • 1-[1-(3,3-Bis(phenyl)propyl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione
  • LC: 100%. MS: m/z 461.7. 1H-NMR (DMSO): d 1.75-2.00 (m, 6H), 2.17 (b, 4H), 2.82 (m, 2H), 3.85 (m, 1H), 3.95 (m, 1H), 4.35 (s, 2H), 7.05-7.20 (m, 5H), 7.22-7.35 (m, 9H).
  • 1-[1-p-benzyloxybenzyl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione
  • LC: 100%. MS: m/z 464 (M+1). 1H-NMR (CDCl3): d 1.90 (m, 2H), 2.10 (m, 4H), 2.98 (m, 2H), 3.45 (s, 2H), 4.10 (m, 1H), 4.45 (s, 2H), 5.10 (s, 2H), 6.90 (d, 2H), 7.10-7.50 (m, 11H).
  • 1-[1-(1,2,3,4-tetrahydronaphthyl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione
  • LC: 100%. MS: m/z 398.5 (M+1). 1H-NMR (MeOH-d4): d 1.75-3.45 (m, 14H), 3.60 (m, 1H), 4.30 (m, 1H), 4.50 (s, 2H), 7.10-7.40 (m, 8H).
  • 1-[1-(4-propylcyclohexyl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione
  • LC: 95.1%. MS: m/z 391.9. 1H-NMR (CDCl3): d 0.90 (m, 4H), 1.20 (m, 1H), 1.30 (m, 5H), 1.60-1.95 (m, 6H), 2.00 (m, 2H), 2.17 (m, 2H), 2.30-2.50 (m, 3H), 3.20 (m, 2H), 4.10 (m, 1H), 4.50 (s, 2H), 7.15 (m, 2H), 7.23 (d, 1H), 7.35 (t, 1H).
  • 1-[1-(5-methylhex-2-yl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione
  • LC: 77.0%. MS: m/z 366.2 (M+1). 1H-NMR (CDCl3): d 0.85 (d, 6H), 0.95 (d, 3H), 1.15-2.15 (m, 9H), 2.22-2.60 (m, 3H), 2.90 (m, 2H), 4.01 (m, 1H), 4.50 (s, 2H), 7.08-7.40 (m, 4H).
  • 1-[1-(norbornan-2-yl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione
  • LC: 100%. MS: m/z 362.6 (M+1). 1H-NMR (CDCl3): d 1.10 (m, 1H), 1.40-1.75 (m, 6H), 2.05 (m, 1H), 2.15 (m, 2H), 2.30-2.55 (m, 3H), 2.60 (b, 1H), 2.80 (m, 2H), 3.10 (m, 1H), 3.51 (m, 2H), 4.25 (m, 1H), 4.50 (s, 2H), 7.20-7.40 (m, 4H).
  • 1-[1-(decahydro-2-naphthyl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione
  • LC: 100%. MS: m/z 404.2 (M+1). 1H-NMR (DMSO): d 1.15-1.78 (m, 16H), 1.85 (m, 1H), 1.90-2.10 (m, 3H), 2.32 (m, 2H), 2.51 (m, 1H), 2.98 (m, 2H), 4.05 (m, 1H), 4.50 (s, 2H), 7.10-7.35 (m, 4H).
  • 1-[1-(cyclooctyl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione
  • LC: 96.0%. MS: m/z 378.5 (M+1). 1H-NMR (MeOH): d 1.5-2.05 (m, 14H), 2.30 (m, 2H), 2.50 (m, 2H), 3.30 (m, 2H), 3.52 (m, 3H), 4.35 (m, 1H), 4.50 (s, 2H), 7.10-7.40 (m, 4H).
  • 1-[1-[4-(2-propyl)-cyclohexyl]-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione
  • LC: 100%. MS: m/z 392.2 (M+1). 1H-NMR (CDCl3): d 0.90 (m, 5H), 1.12 (m, 1H), 1.25 (m, 2H), 1.47 (b, 2H), 1.65 (m, 2H), 1.80-2.00 (m, 3H), 2.18 (m, 3H), 2.54 (m, 2H), 3.08 (m, 3H), 3.50 (m, 2H), 4.29 (m, 1H), 4.53 (s, 2H), 7.10-7.30 (m, 3H), 7.40 (t, 1H).
  • 1-[1-(1,3-dihydroinden-2-yl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione
  • LC: 100%. MS: m/z 384.3 (M+1). 1H-NMR (DMSO): d 1.78-2.10 (m, 6H), 2.70 (m, 2H), 2.90-3.10 (m, 5H), 3.89 (m, 1H), 4.35 (s, 2H), 7.00-7.25 (m, 8H).
  • 3-butyl-1-[1-[4-(2-propyl)-cyclohexyl]-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione
  • LC: 100%. MS: 448.4. H-NMR (DMSO): 7.35 t (1H); 7.11-7.23 m (3H); 6.60 s (2H); 4.48 s (2H); 3.88-3.97 m (1H); 3.25 d (2H); 2.95 t (2H); 2.41 bs (1H); 2.25 bs (2H); 1.87 m (4H); 1.65 m (7H); 1.51 m (2H); 1.33 m (4H); 0.88 t (3H); 0.81 d (6H).
  • 3-acetamido-1-[1-[4-(2-propyl)-cyclohexyl]-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione
  • LC: 100%. MS: 449.2. H-NMR (CDCl3): 7.48 s (1H); 7.35 t (1H); 7.25-7.33 m (3H); 6.55 s (2H); 4.51 s (2H); 3.90 m (1H); 3.56 s (2H); 3.18 d (2H); 2.45 bs (1H); 2.33 bs (2H); 1.75-1.95 m (4H); 1.50-1.68 m (6H); 1.44 m (1H); 1.33 m (1H); 1.12 m (1H); 0.78 d (6H).
  • 3-(2-methanesulfonamido)-1-[1-[4-(2-propyl)-cyclohexyl]-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione
  • LC: 100%. MS: 513.2. H-NMR (DMSO): 7.09-7.30 m (4H); 6.58 s (2H); 4.54 s (2H); 4.46 d (2H); 3.99 m (4H); 3.12 m (5H); 2.88 s (3H); 2.33 bs (3H); 1.98 s (3H); 1.89 m (5H); 1.34 m (6H); 1.18 m (4H); 1.16 t (2H); 0.82 d (6H).
  • 3-methoxycarbonylmethyl-1-[1-[4-(2-propyl)-cyclohexyl]-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione
  • LC: 100%. MS: 464.2. H-NMR (DMSO): 7.41 m (1H); 7.33 m (3H); 6.67 s (4H); 4.65 s (2H); 3.95 s (3H); 3.65 s (3H); 2.49 m (1H); 2.38 m (2H); 2.05 m (2H); 1.77 m (2H); 1.56 m (6H); 1.35 m (2H); 1.25 m (1H); 0.80 d (6H).
  • 3-cyanomethyl-1-[1-[4-(2-propyl)-cyclohexyl]-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione
  • LC: 100%. MS: 431.3. H-NMR (DMSO): 7.10-7.48 m (4H); 4.60 s (2H); 4.45 s (2H); 3.77 m (1H); 2.91 d (2H); 2.25 m (1H); 1.90 t (2H); 1.85 d (2H); 1.50-1.66 m (7H); 1.21-1.44 m (5H); 1.07 m (1H); 0.82 d (6H).
  • 3-(2-hydroxyethyl)-1-[1-[4-(2-propyl)-cyclohexyl]-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione
  • LC: 100%. MS: 436.2. H-NMR (CDCl3): 7.38 t (1H); 7.22 m (3H); 6.67 s (2H); 4.55 s (2H); 3.95 m (1H); 3.62 m (2H); 3.11 m (4H); 2.44 bs (1H); 2.35 bs (2H); 1.85 bs (4H); 1.55-1.65 m (5H); 1.48 m (2H); 1.33 m (2H); 1.12 m (1H); 0.82 d (6H)
  • 3-butoxycarbonylmethyl-1-[1-[4-(2-propyl)-cyclohexyl]-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione
  • LC: 100%. MS: 506.2. H-NMR (DMSO): 7.31 m (1H); 7.22 m (3H); 4.50 s (2H); 3.71 s (3H); 2.90 d (2H); 2.22 m (1H); 1.98 t (2H); 1.80 d (2H); 1.50-1.70 m (4H); 1.44-1.58 m (12H); 1.28 m (5H); 1.01 m (1H) 0.81 d (6H)
  • Other compounds within the scope of formula (J) or (IA) of the present invention can be synthesized by analogous techniques.
  • Example 4
  • Nociceptin affinity at the ORL1 receptor for preferred compounds was obtained using the following assay:
  • Membranes from recombinant HEK-293 cells expressing the human opioid receptor-like receptor (ORL-1) (Receptor Biology) were prepared by lysing cells in ice-cold hypotonic buffer (2.5 mM MgCl2, 50 mM HEPES, pH 7.4) (10 ml/10 cm dish) followed by homogenization with a tissue grinder/teflon pestle. Membranes were collected by centrifugation at 30,000×g for 15 min at 4° C. and pellets resuspended in hypotonic buffer to a final concentration of 1-3 mg/ml. Protein concentrations were determined using the BioRad protein assay reagent with bovine serum albumen as standard. Aliquots of the ORL-1 receptor membranes were stored at −80° C.
  • Functional SGTPgS binding assays were conducted as follows. ORL-1 membrane solution was prepared by sequentially adding final concentrations of 0.066 mg/ml ORL-1 membrane protein, 10 mg/ml saponin, 3 mM GDP and 0.20 nM [35S]GTPgS to binding buffer (100 mM NaCl, 10 mM MgCl2, 20 mM HEPES, pH 7.4) on ice. The prepared membrane solution (190 ml/well) was transferred to 96-shallow well polypropylene plates containing 10 ml of 20× concentrated stock solutions of agonist prepared in DMSO. Plates were incubated for 30 min at room temperature with shaking. Reactions were terminated by rapid filtration onto 96-well Unifilter GF/B filter plates (Packard) using a 96-well tissue harvester (Brandel) and followed by three filtration washes with 200 ml ice-cold binding buffer (10 mM NaH2PO4, 10 mM Na2HPO4, pH 7.4). Filter plates were subsequently dried at 50° C. for 2-3 hours. Fifty ml/well scintillation cocktail (BetaScint; Wallac) was added and plates were counted in a Packard Top-Count for 1 ml/well.
  • Data was analyzed using the curve fitting functions in GraphPad PRISMÔ, v. 3.0 and the results are set forth in table 1 below:
  • TABLE 1
    Nociceptin Affinity
    calc Ki
    Compound (nM)
    1-1-[1-(naphth-2-yl-methyl)-4-piperidinyl]-2,1,3- >10,000
    benzothiadiazin-2,2-dione;
    1-1-[1-(p-phenylbenzyl)-4-piperidinyl]-2,1,3-benzothiadiazin- >10,000
    2,2-dione;
    1-1-[1-(3,3-Bis(phenyl)propyl)-4-piperidinyl]-2,1,3- 4461
    benzothiadiazin-2,2-dione;
    1-1-[1-(p-benzyloxybenzyl)-4-piperidinyl]-2,1,3- 7114
    benzothiadiazin-2,2-dione;
    1-1-[1-(cyclooctylmethyl)-4-piperidinyl]-2,1,3-benzothiadiazin- 49
    2,2-dione;
    1-[1-(4-propylcyclohexyl)-4-piperidinyl]-2,1,3-benzothiadiazol- 5102
    2,2-dione;
    1-[1-(5-methylhex-2-yl)-4-piperidinyl]-2,1,3-benzothiadiazol- 3592
    2,2-dione;
    1-[1-[4-(2-propyl)-cyclohexyl]-4-piperidinyl]-2,1,3- 357
    benzothiadiazol-2,2-dione;
    1-[1-(1,2,3,4-tetrahydro-2-naphthyl)-4-piperidinyl]-2,1,3- 3454
    benzothiadiazol-2,2-dione;
    1-[1-(decahydro-2-naphthyl)-4-piperidinyl]-2,1,3- 225
    benzothiadiazol-2,2-dione;
    1-[1-(1,3,-dihydroinden-2-yl)-4-piperidinyl]-2,1,3- 5670
    benzothiadiazol-2,2-dione;
    1-[1-(cyclooctyl)-4-piperidinyl]-2,1,3-benzothiadiazol-2,2- 2297
    dione;
    1-[1-(naphth-2-yl-methyl)-4-piperidinyl]-2,1,3-benzothiadiazin- 609
    2,2-dione
    1-[1-(p-benzyloxybenzyl)-4-piperidinyl]-2,1,3-benzothiadiazin- 1878
    2,2-dione
    1-[1-(p-phenylbenzyl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2- 9535
    dione
    1-[1-(decahydro-2-naphthyl)-4-piperidinyl]-2,1,3- 28.7
    benzothiadiazin-2,2-dione
    1-[1-[4-(2-propyl)-cyclohexyl]-4-piperidinyl]-2,1,3- 35.3
    benzothiadiazin-2,2-dione
    1-1-[1-(4-propylcyclohexyl)-4-piperidinyl]-2,1,3- 231
    benzothiadiazin-2,2-dione
    1-1-[1-(benzyl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2-dione; 3900
    1-1-[1-(10,11-Dihydro-5H-dibenzo [a,d]-cyclohepten-5-yl)-4- 505
    piperidinyl]-2,1,3-benzothiadiazin-2,2-dione;
    1-1-[1-(1,2,3,4-tetrahydronaphthyl)-4-piperidinyl]-2,1,3- 2614
    benzothiadiazin-2,2-dione;
    1-1-[1-(5-methylhex-2-yl)-4-piperidinyl]-2,1,3-benzothiadiazin- 40
    2,2-dione;
    1-1-[1-(norbornan-2-yl)-4-piperidinyl]-2,1,3-benzothiadiazin- 6329
    2,2-dione;
    1-1-[1-(cyclooctyl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2- 187
    dione;
    1-1-[1-(1,3-dihydroinden-2-yl)-4-piperidinyl]-2,1,3- 46
    benzothiadiazin-2,2-dione.
    1-[1-(3,3-Bis(phenyl)propyl)-4-piperidinyl]-2,1,3- 83
    benzothiadiazin-2,2-dione
    3-butyl-1-[1-[4-(2-propyl)-cyclohexyl]-4-piperidinyl]-2,1,3- 18
    benzothiadiazin-2,2-dione
    3-acetamido-1-[1-[4-(2-propyl)-cyclohexyl]-4-piperidinyl]- 3.9
    2,1,3-benzothiadiazin-2,2-dione
    3-(2-methanesulfonamido)-1-[1-[4-(2-propyl)-cyclohexyl]-4- 5
    piperidinyl]-2,1,3-benzothiadiazin-2,2-dione
    3-methoxycarbonylmethyl-1-[1-[4-(2-propyl)-cyclohexyl]-4- 41
    piperidinyl]-2,1,3-benzothiadiazin-2,2-dione
    3-cyanomethyl-1-[1-[4-(2-propyl)-cyclohexyl]-4-piperidinyl]- 33
    2,1,3-benzothiadiazin-2,2-dione
    3-(2-hydroxyethyl)-1-[1-[4-(2-propyl)-cyclohexyl]-4- 7.4
    piperidinyl]-2,1,3-benzothiadiazin-2,2-dione
    3-butoxycarbonylmethyl-1-[1-[4-(2-propyl)-cyclohexyl]-4- 63
    piperidinyl]-2,1,3-benzothiadiazin-2,2-dione
  • Example 5 Synthesis of Quinoline Head Groups
  • Figure US20090253727A1-20091008-C00016
  • Procedure:
  • To a solution of 1 (5 g, 25.1 mmol) in 125 mL of MeOH at room temperature was added NaBH4 (1.9 g, 50.2 mmol) portionwise and the reaction mixture was stirred for 2 hr. The mixture was evaporated to dryness and sat. NH4Cl solution was added. The mixture was extracted with EtOAc (5×). The combined organic extracts were dried over K2CO3, filtered and evaporated under reduced pressure to give the crude product 2 as a white solid. This material was used directly in the next step without further purification.
  • 1H-NMR (CDCl3): d 1.40-1.55 (m, 11H), 1.85 (m, 2H), 3.00 (m, 2H), 3.85 (m, 3H).
  • To a solution of 2 (5.05 g, 25.1 mmol) in 50 mL of THF at 0° C. was added Et3N (5.25 mL, 37.6 mmol) and CH3SO2CJ (2.14 mL, 27.6 mmol). The reaction was stirred at room temperature for 2 hr. The resulting mixture was diluted with ether, washed with sat. NH4Cl solution (3×), dried over MgSO4 and evaporated to give the crude product as a solid. Trituration with hexane gave 3 as an off-white solid (6.30 g, 90% for two steps).
  • 1H-NMR (CDCl3): d 1.45 (s, 9H), 1.80 (m, 2H), 1.95 (m, 2H), 3.02 (s, 3H), 3.30 (m, 2H), 3.72 (m, 2H), 4.90 (m, 1H).
  • To a solution of compound 4 (5.00 g, 34.0 mmol) in 500 mL of xylene was added NaH (1.63 g, 40.8 mmol) at room temperature. After the addition, the mixture was heated to reflux for 2 hr. After cooling to room temperature 3 (11.39 g, 40.8 mmol) was added in one portion. The reaction mixture was heated to gentle reflux and maintained for 20 hr. The cooled solution was partitioned between brine and EtOAc. The layers were separated and the aqueous layer extracted with EtOAc (1×). The combined organic extracts were dried over MgSO4, filtered and evaporated to give the crude 5 as an oil, which was used directly to the next step without purification.
  • A mixture of concentrated HCl in EtOAc (150 ml, 1:10) was added to compound 5 (11.2 g, 34.0 mmol) at room temperature. The reaction was monitored by TLC. When the reaction was complete, water and EtOAc were added, the layers separated and the aqueous layer was washed with EtOAc (1×). The organic washings were discarded and the aqueous layer was basified with K2CO3 and extracted with EtOAc (3×). The organic extracts were dried over MgSO4, filtered and evaporated to give the crude product that was purified by column chromatography to give 6 (4.30 g, 55% for 2 steps) as a clear glass.
  • 1H-NMR (CDCl3): d 1.75 (m, 2H), 2.47-2.88 (m, 8H), 3.20 (m, 2H), 4.35 (m, 1H), 7.02 (m, 1H), 7.12-7.25 (m, 3H).
  • Example 6 Attachment of Tail Groups
  • Tail groups were attached to the quinoline head groups according to the following procedures:
  • Figure US20090253727A1-20091008-C00017
  • General Procedure for Alkylation:
  • To a solution of the amine (1 eq) and triethylamine (1 eq) in dimethylformamide, was added 1 eq of alkyl bromide or chloride in one portion. The mixture was stirred and heated at 80° C. over night. TLC indicated the reaction was complete. The reaction was quenched by the addition of water followed by 1 N NaOH to pH 10. The mixture was extracted 2× with Et2O. The combined organic extracts were dried over potassium carbonate and the solvent evaporated, followed by chromatography to give the pure product.
  • General Procedure for Reductive Amination:
  • To a mixture of ketone or aldehyde (1 eq), amine (1 eq), and acetic acid (1 eq) in methanol, was added sodium cyanoborohydride (1.4 eq) in one portion. The mixture was stirred over night at room temperature. TLC indicated the reaction was complete. The reaction was quenched by the addition of water followed by 1 N NaOH to pH 10. The mixture was extracted 2× with Et2O. The combined organic extracts were dried over potassium carbonate and the solvent evaporated, followed by chromatography to give the pure product.
  • The following compounds were prepared by attaching the tail groups using the general procedures described:
  • 1,2,3,4-tetrahydro-1-[1-(naphth-2-yl-methyl)-4-piperidinyl]-quinolin-2-one
  • LC: 97.4%. MS: m/z 371.2 (M+1). 1H-NMR (CDCl3): d 1.70 (m, 2H), 2.20 (m, 2H), 2.55 (m, 2H), 2.68 (m, 2H), 2.82 (m, 2H), 3.05 (b, 2H), 3.70 (s, 2H), 4.32 (m, 1H), 7.02 (m, 1H), 7.15 (d, 1H), 7.20-7.30 (m, 2H), 7.42 (m, 2H), 7.51 (d, 1H), 7.75 (s, 1H), 7.85 (m, 3H).
  • 1,2,3,4-tetrahydro-1-[1-(p-phenylbenzyl)-4-piperidinyl]-quinolin-2-one
  • LC: 100%. MS: 1-m/z 397.2 (M+1). 1H-NMR (CDCl3): d 1.75 (b, 2H), 2.20 (m, 2H), 2.60 (m, 2H), 2.72 (m, 2H), 2.85 (m, 2H), 3.08 (b, 2H), 3.62 (s, 2H), 4.35 (m, 1H), 7.00-7.70 (m, 13H).
  • 1,2,3,4-tetrahydro-1-[1-[4,4-bis(4-fluorophenyl)butyl]-4-piperidinyl]-quinolin-2-one
  • LC: 93.6%. MS: m/z 475.3 (M+1). 1H-NMR (CDCl3): d 1.50 (m, 2H), 1.75 (m, 2H), 2.05 (m, 4H), 2.4 (m, 2H), 2.65 (m, 4H), 2.80 (m, 2H), 3.01 (m, 2H), 3.90 (t, 1H), 4.35 (m, 1H), 6.90-7.35 (m, 12H).
  • 1,2,3,4-tetrahydro-1-[1-(p-benzyloxybenzyl)-4-piperidinyl]-quinolin-2-one
  • LC: 100%. MS: m/z 427.2 (M+1). 1H-NMR (CDCl3): d 1.75 (m, 2H), 2.10 (m, 2H), 2.78-2.90 (m, 4H), 2.85 (m, 2H), 3.05 (m, 2H), 3.50 (s, 2H), 4.40 (m, 1H), 5.05 (s, 2H), 7.00-7.60 (m, 13H).
  • 1,2,3,4-tetrahydro-1-[1-(1,2,3,4-tetrahydro-2-naphthyl)-4-piperidinyl]-quinolin-2-one
  • LC: 98.8%. MS: m/z 361.2 (M+1). 1H-NMR (CDCl3): d 1.88-2.12 (m, 3H), 2.45 (m, 1H), 2.65 (m, 2H), 2.80-2.35 (m, 8H), 3.50-3.75 (m, 3H), 3.90 (m, 2H), 4.80 (m, 1H), 7.05-7.55 (m, 8H).
  • 1,2,3,4-tetrahydro-1-[1-(4-propyl-cyclohexyl)-4-piperidinyl]-quinolin-2-one
  • LC: 100%. MS: m/z 355.2 (M+1). 1H-NMR (CDCl3): d 0.80-2.25 (m, 18H), 3.55-3.70 (m, 2H), 2.85-3.30 (m, 6H), 3.45-3.80 (m, 4H), 7.00-7.50 (m, 4H).
  • 1,2,3,4-tetrahydro-1-[1-(5-methylhex-2-yl)-4-piperidinyl]-quinolin-2-one
  • LC: 100%. MS: m/z 329.6 (M+1). 1H-NMR (CDCl3): d 0.95 (m, 6H), 1.25-1.35 (m, 3H), 1.42 (d, 3H), 1.53-1.70 (m, 2H), 2.10 (m, 2H), 3.60 (m, 2H), 2.85-3.55 (m, 12H), 7.05-7.40 (m, 4H).
  • 1,2,3,4-tetrahydro-1-[1-(norbornan-2-yl)-4-piperidinyl]-quinolin-2-one
  • LC: 100%. MS: m/z 325.2 (M+1). 1H-NMR (CDCl3): d 1.22 (m, 1H), 1.40-1.65 (m, 5H), 1.80-2.10 (m, 5H), 2.30 (b, 1H), 2.50 (m, 2H), 2.61 (b, 1H), 2.85 (m, 2H), 2.90-3.20 (m, 4H), 3.50 (m, 2H), 4.45 (m, 1H), 7.02 (t, 1H), 7.22 (d, 1H), 7.28-7.40 (m, 2H).
  • 1,2,3,4-tetrahydro-1-[1-(decahydro-2-naphthyl)-4-piperidinyl]-quinolin-2-one
  • LC: 100%. MS: m/z 367.2 (M+1). 1H-NMR (CDCl3): d 1.20-2.15 (m, 12H), 3.63 (m, 2H), 2.85-3.75 (m, 14H), 4.25-4.45 (m, 2H), 7.05-7.45 (m, 4H).
  • 1,2,3,4-tetrahydro-1-[1-(10,11-dihydro-5H-dibenzo[a.d]-cyclohepten-5-yl)-4-piperidinyl]-quinolin-2-one
  • LC: 100%. 1H-NMR (CDCl3): d 1.62 (b, 2H), 2.00 (m, 2H), 2.51-2.70 (m, 4H), 3.85 (m, 6H), 4.00 (s, 1H), 4.05-4.25 (m, 3H), 7.00-7.30 (m, 12H).
  • 1,2,3,4-tetrahydro-1-[1-(3,3-diphenylpropyl)-4-piperidinyl]-quinolin-2-one
  • LC: 99.3%. MS: m/z 425.3 (M+1). 1H-NMR (CDCl3): d 1.70 (b, 2H), 2.05 (m, 2H), 2.30 (m, 4H), 2.55-2.70 (m, 4H), 2.80 (m, 2H), 3.02 (b, 2H), 4.02 (m, 1H), 4.30 (m, 1H), 7.00 (m, 1H), 7.12-7.35 (m, 13H).
  • 1,2,3,4-tetrahydro-1-[1-(cyclooctyl)-4-piperidinyl]-quinolin-2-one
  • LC: 100%. MS: m/z 341.2 (M+1). 1H-NMR (CDCl3): d 1.36-1.76 (m, 114H), 1.78-1.89 (m, 2H), 1.92-1.95 (m, 2H), 1.98-2.09 (m, 2H), 2.58-2.62 (m, 2H), 2.79-2.82 (m, 2H), 3.00-3.08 (m, 3H), 3.28-3.42 (m, 3H), 4.90-4.98 (m, 1H), 7.05 (t, 1H), 7.14 (d, 1H), 7.40 (d, 1H), 7.51 (t, 1H).
  • 1,2,3,4-tetrahydro-1-[1-[4-(1-methylethyl)-cyclohexyl]-4-piperidinyl]-quinolin-2-one
  • LC: 100%. MS: m/z 355.2 (M+1). 1H-NMR (CDCl3): d 0.90-1.05 (m, 6H), 1.05-2.30 (m, 13H), 2.60 (m, 2H), 2.80-3.80 (m, 6H), 4.35 (m, 2H), 4.55 (m, 1H), 7.05-7.45 (m, 4H).
  • 1,2,3,4-tetrahydro-1-[1-(1,3-dihydroinden-2-yl)-4-piperidinyl]-quinolin-2-one
  • LC: 90.4%. MS: m/z 347.2 (M+1). 1H-NMR (CDCl3): d 1.90 (b, 2H), 2.60 (m, 2H), 2.70-3.00 (m, 6H), 3.25 (m, 4H), 3.40 (b, 2H), 3.80 (m, 1H), 4.70 (m, 1H), 7.00-7.40 (m, 8H).
  • 1,2,3,4-tetrahydro-1-[1-(cyclooctylmethyl)-4-piperidinyl]-quinolin-2-one
  • LC: 100%. MS: m/z 355.3 (M+1). 1H-NMR (CDCl3): d 1.15-1.28 (m, 2H), 1.39-1.78 (M, 15H), 1.98-2.10 (m, 4H), 2.51-2.68 (m, 4H), 2.79 (t, 2H), 2.98 (d, 2H), 4.21-4.31 (m, 1H), 6.95-7.01 (m, 1H), 7.11-7.14 (m, 1H), 7.20-7.24 (m, 2H).
  • Other compounds within the scope of formula (II) or (IIA) of the present invention can be synthesized by analogous techniques.
  • Example 7
  • Nociceptin affinity at the ORL1 receptor for preferred compounds was obtained using the following assay:
  • Membranes from recombinant HEK-293 cells expressing the human opioid receptor-like receptor (ORL-1) (Receptor Biology) were prepared by lysing cells in ice-cold hypotonic buffer (2.5 mM MgCl2, 50 mM HEPES, pH 7.4) (10 ml/10 cm dish) followed by homogenization with a tissue grinder/teflon pestle. Membranes were collected by centrifugation at 30,000×g for 15 min at 4° C. and pellets resuspended in hypotonic buffer to a final concentration of 1-3 mg/ml. Protein concentrations were determined using the BioRad protein assay reagent with bovine serum albumen as standard. Aliquots of the ORL-1 receptor membranes were stored at −80° C.
  • Functional SGTPgS binding assays were conducted as follows. ORL-1 membrane solution was prepared by sequentially adding final concentrations of 0.066 mg/ml ORL-1 membrane protein, 10 mg/ml saponin, 3 mM GDP and 0.20 nM [35S]GTPgS to binding buffer (100 mM NaCl, 10 mM MgCl2, 20 mM HEPES, pH 7.4) on ice. The prepared membrane solution (190 ml/well) was transferred to 96-shallow well polypropylene plates containing 10 ml of 20× concentrated stock solutions of agonist prepared in DMSO. Plates were incubated for 30 min at room temperature with shaking. Reactions were terminated by rapid filtration onto 96-well Unifilter GF/B filter plates (Packard) using a 96-well tissue harvester (Brandel) and followed by three filtration washes with 200 ml ice-cold binding buffer (10 mM NaH2PO4, 10 mM Na2HPO4, pH 7.4). Filter plates were subsequently dried at 50° C. for 2-3 hours. Fifty ml/well scintillation cocktail (BetaScint; Wallac) was added and plates were counted in a Packard Top-Count for 1 min/well.
  • Data was analyzed using the curve fitting functions in GraphPad PRISMÔ, v. 3.0 and the results are set forth in table 2 below:
  • TABLE 2
    Nociceptin Affinity
    calc Ki
    Compound (nM)
    1,2,3,4-tetrahydro-1-[1-(naphth-2-yl-methyl)-4-piperidinyl]- 3389
    quinolin-2-one
    1,2,3,4-tetrahydro-1-[1-(p-phenylbenzyl)-4-piperidinyl]- >10,000
    quinolin-2-one
    1,2,3,4-tetrahydro-1-[1-[4,4-bis(4-fluorophenyl)butyl]-4- 2898
    piperidinyl]-quinolin-2-one
    1,2,3,4-tetrahydro-1-[1-(p-benzyloxybenzyl)-4-piperidinyl]- 3502
    quinolin-2-one
    1,2,3,4-tetrahydro-1-[1-(1,2,3,4-tetrahydro-2-naphthyl)-4- 176
    piperidinyl]-quinolin-2-one-
    1,2,3,4-tetrahydro-1-[1-(4-propyl-cyclohexyl)-4-piperidinyl]- 1257
    quinolin-2-one
    1,2,3,4-tetrahydro-1-[1-(5-methylhex-2-yl)-4-piperidinyl]- 505
    quinolin-2-one
    1,2,3,4-tetrahydro-1-[1-(norbornan-2-yl)-4-piperidinyl]- 781
    quinolin-2-one
    1,2,3,4-tetrahydro-1-[1-(decahydro-2-naphthyl)-4-piperidinyl]- 105
    quinolin-2-one
    1,2,3,4-tetrahydro-1-[1-(10,11-dihydro-5H-dibenzo[a.d]- 95
    cyclohepten-5-yl)-4-piperidinyl]-quinolin-2-one;
    1,2,3,4-tetrahydro-1-[1-(3,3-diphenylpropyl)-4-piperidinyl]- 71
    quinolin-2-one
    1,2,3,4-tetrahydro-1-[1-[4-(1-methylethyl)-cyclohexyl]-4- 80
    piperidinyl]-quinolin-2-one
    1,2,3,4-tetrahydro-1-[1-(1,3-dihydroinden-2-yl)-4-piperidinyl]- 71
    quinolin-2-one
    1,2,3,4-tetrahydro-1-[1-(cyclooctyl)-4-piperidinyl]-quinolin-2- 14
    one
  • Example 8
  • Affinity at the μ receptor for compounds was obtained according to the following assay:
  • Mu opioid receptor membrane solution was prepared by sequentially adding final concentrations of 0.075 μg/μl of the desired membrane protein, 10 μg/ml saponin, 3 μM GDP and 0.20 nM [35S]GTPγS to binding buffer (100 mM NaCl, 10 mM MgCl2, 20 mM HEPES, pH 7.4) on ice. The prepared membrane solution (190 μl/well) was transferred to 96-shallow well polypropylene plates containing 10 μl of 20× concentrated stock solutions of agonist prepared in DMSO. Plates were incubated for 30 min at room temperature with shaking. Reactions were terminated by rapid filtration onto 96-well Unifilter GF/B filter plates (Packard) using a 96-well tissue harvester (Brandel) and followed by three filtration washes with 200 μl ice-cold binding buffer (10 mM NaH2PO4, 10 mM Na2HPO4, pH 7.4). Filter plates were subsequently dried at 50° C. for 2-3 hours. Fifty μl/well scintillation cocktail (MicroScint20, Packard) was added and plates were counted in a Packard Top-Count for 1 min/well.
  • Data were analyzed using the curve fitting functions in GraphPad PRISM™, v. 3.0 and the results of several compounds are set forth in table 3 below:
  • TABLE 3
    Mu Receptor Affinity
    calc Ki
    Compound (nM)
    1-[1-(decahydro-2-naphthyl)-4-piperidinyl]-2,1,3- 91.7
    benzothiadiazin-2,2-dione
    1-[1-[4-(2-propyl)-cyclohexyl]-4-piperidinyl]-2,1,3- 207
    benzothiadiazin-2,2-dione
    1-1-[1-(cyclooctyl)-4-piperidinyl]-2,1,3-benzothiadiazin-2,2- 1376
    dione;
    1-1-[1-(1,3-dihydroinden-2-yl)-4-piperidinyl]-2,1,3- 660
    benzothiadiazin-2,2-dione.
    1,2,3,4-tetrahydro-1-[1-(cyclooctyl)-4-piperidinyl]-quinolin-2- 86
    one;
    1,2,3,4-tetrahydro-1-[1-(5-methylhex-2-yl)-4-piperidinyl]- 156
    quinolin-2-one
    1,2,3,4-tetrahydro-1-[1-(norbornan-2-yl)-4-piperidinyl]- 957
    quinolin-2-one
    1,2,3,4-tetrahydro-1-[1-(decahydro-2-naphthyl)-4-piperidinyl]- 341
    quinolin-2-one

Claims (31)

1-32. (canceled)
33. A compound of the formula (II):
Figure US20090253727A1-20091008-C00018
wherein W is hydrogen, C1-10 alkyl, C3-12 cycloalkyl, C3-12 cycloalkylC1-4alkyl-, C1-10 alkoxy, C3-12 cycloalkoxy-, C1-10 alkyl substituted with 1-3 halogen, C3-12 cycloalkyl substituted with 1-3, halogen, C3-12 cycloalkylC1-4alkyl-substituted with 1-3 halogen, C1-10 alkoxy substituted with 1-3 halogen, C3-12 cycloalkoxy- substituted with 1-3 halogen, —COOV1, —C1-4COOV1, —CH2OH, —SO2N(V1)2, hydroxyC1-10alkyl-, hydroxyC3-10cycloalkyl-, cyanoC1-10alkyl-, cyanoC3-10cycloalkyl-, —CON(V1)2, NH2SO2C1-4alkyl-, NH2SOC1-4alkyl-, sulfonylaminoC1-10alkyl-, diaminoalkyl-, -sulfonylC1-4alkyl, a 6-membered heterocyclic ring, a 6-membered heteroaromatic ring, a 6-membered heterocyclicC1-4alkyl-, a 6-membered heteroaromaticC1-4alkyl-, a 6-membered aromatic ring, a 6-membered aromaticC1-4 alkyl-, a 5-membered heterocyclic ring optionally substituted with an oxo or thio, a 5-membered heteroaromatic ring, a 5-membered heterocyclicC1-4alkyl- optionally substituted with an oxo or thio, a 5-membered heteroaromaticC1-4alkyl-, —C1-5(═O)W1, —C0-5(═NH)W1, —C1-5NHC(═O)W1, —C1-5NHS(═O)2W1, —C1-5NHS(═O)W1, wherein W1 is hydrogen, C1-10 alkyl, C3-12 cycloalkyl, C1-10alkoxy, C3-12 cycloalkoxy, —CH2OH, amino, C1-4alkylamino-, diC1-4alkylamino-, or a 5-membered heteroaromatic ring optionally substituted with 1-3 lower alkyl;
wherein each V1 is independently selected from 1, C1-6 alkyl, C3-6 cycloalkyl, benzyl and phenyl;
Q is a 6 membered aromatic group;
n is an integer from 0 to 3,
A, B and C are independently hydrogen, C1-10 alkyl, C3-12 cycloalkyl, C1-10alkoxy, C3-12 cycloalkoxy, —CH2OH, —NHSO2, hydroxyC1-10alkyl-, aminocarbonyl-, C1-4alkylaminocarbonyl-, diC1-4alkylaminocarbonyl-, acylamino-, acylaminoalkyl-, amide, sulfonylaminoC1-10alkyl-, or A-B can together form a C2-6 bridge, or B-C can together form a C3-7 bridge, or A-C can together form a C1-5 bridge;
Z is selected from the group consisting of a bond, straight or branched C1-6 alkylene, —NH—, —CH2O—, —CH2NH—, —CH2N(CH3)—, —NHCH2—, —CH2CONH—, —NHCH2CO—, —CH2CO—, —COCH2—, —CH2COCH2—, —CH(CH3)—, —CH═, —O— and —HC═CH—, wherein the carbon and/or nitrogen atoms are unsubstituted or substituted with one or more lower alkyl, hydroxy, halo or alkoxy group;
R1 is selected from the group consisting of hydrogen, C1-10alkyl, C3-12cycloalkyl, C2-10alkenyl, amino, C1-10alkylamino-, C3-12cycloalkylamino-, —COOV1, —C1-4COOV1, cyano, cyanoC1-10alkyl-, cyanoC3-10cycloalkyl-, NH2SO2—, NH2SO2C1-4alkyl-, NH2SOC1-4alkyl-, aminocarbonyl-, C1-4alkylaminocarbonyl-, diC1-4alkylaminocarbonyl-, benzyl, C3-12 cycloalkenyl-, a monocyclic, bicyclic or tricyclic aryl or heteroaryl ring, a hetero-monocyclic ring, a hetero-bicyclic ring system, and a spiro ring system of the formula (III):
Figure US20090253727A1-20091008-C00019
wherein X1 and X2 are independently selected from the group consisting of NH, O, S and CH2;
wherein said allyl, cycloalkyl, alkenyl, C1-10alkylamino-, C3-12cycloalkylamino-, or benzyl of R1 is optionally substituted with 1-3 substituents selected from the group consisting of halogen, hydroxy, C1-10 alkyl, C1-10 alkoxy, nitro, trifluoromethyl-, cyano, —COOV1, —C1-4COOV1, cyanoC1-10alkyl-, —C1-5(═O)W1, —C1-5NHS(═O)2W1, —C1-5NHS(═O)W1, a 5-membered heteroaromaticC0-4alkyl-, phenyl, benzyl, benzyloxy, said phenyl, benzyl, and benzyloxy optionally being substituted with 1-3 substituents selected from the group consisting of halogen, C1-10 alkyl-, C1-10 alkoxy-, and cyano; and wherein said C3-12 cycloalkyl, C3-12 cycloalkenyl, monocyclic, bicyclic or tricyclic aryl, heteroaryl ring, hetero-monocyclic ring, hetero-bicyclic ring system, or spiro ring system of the formula (III) is optionally substituted with 1-3 substituents selected from the group consisting of halogen, C1-10 alkyl, C1-10 alkoxy, nitro, trifluoromethyl-, phenyl, benzyl, phenyloxy and benzyloxy, wherein said phenyl, benzyl, phenyloxy or benzyloxy is optionally substituted with 1-3 substituents selected from the group consisting of halogen, C1-10 alkyl, C1-10 alkoxy, and cyano;
R2 is selected from the group consisting of hydrogen, C1-10 alkyl, C3-12 cycloalkyl and halogen, said alkyl or cycloalkyl optionally substituted with an oxo, amino, alkylamino or dialkylamino group;
or a pharmaceutically acceptable salt thereof or solvate thereof.
34. (canceled)
35. The compound of claim 33, wherein W is selected from the group consisting of —CH2C(═O)NH2, —C(NH)NH2, pyridylmethyl, cyclopentyl, cyclohexyl, furanylmethyl, —C(O)CH3, —CH2CH2NHC═OCH3, —SO2CH3, CH2CH2NHSO2CH3, furanylcarbonyl-, methylpyrrolylcarbonyl-, diazolecarbonyl-, azolemethyl-, trifluoroethyl-, hydroxyethyl-, cyano methyl-, oxo-oxazolemethyl-, and diazolemethyl-.
36. The compound of claim 33, wherein ZR1 is selected from the group consisting of cyclohexylethyl-, cyclohexylmethyl-, cyclopentylmethyl-, dimethylcyclohexylmethyl-, phenylethyl-, pyrrolyltrifluoroethyl-, thienyltrifluoroethyl-, pyridylethyl-, cyclopentyl-, cyclohexyl-, methoxycyclohexyl-, tetrahydropyranyl-, propylpiperidinyl-, indolylmethyl-, pyrazoylpentyl-, thiazolylethyl-, phenyltrifluoroethyl-, hydroxyhexyl-, methoxyhexyl-, isopropoxybutyl-, hexyl-, and oxocanylpropyl-.
37. The compound of claim 33, wherein at least one of ZR1 or W is selected from the group consisting of CH2COOV1, tetrazolylmethyl-, cyanomethyl-, NH2SO2methyl-, NH2SOmethyl-, aminocarbonylmethyl-, C1-4alKylaminocarbonylmethyl-, and diC1-4alkylaminocarbonylmethyl-.
38. The compound of claim 33, wherein ZR1 is 3,3 diphenylpropyl optionally substituted at the 3 carbon of the propyl with —COOH, —COOV1, tetrazolylC0-4alkyl-, cyano-, aminocarbonyl-, C1-4alkylaminocarbonyl-, or diC1-4alkylaminocarbonyl-.
39. A compound of the formula (IIA):
Figure US20090253727A1-20091008-C00020
wherein
n is an integer from 0 to 3;
Z is selected from the group consisting of a bond, —CH2—, —NH—, —CH2O—, —CH2CH2—, —CH2NH—, —CH2N(CH3)—, —NHCH2—, —CH2CONH—, —NHCH2CO—, —CH2CO—, —COCH2—, —CH2COCH2—, —CH(CH3)—, —CH═, and —HC═CH—, wherein the carbon and/or nitrogen atoms are unsubstituted or substituted with a lower alkyl, halogen, hydroxy or alkoxy group;
R1 is selected from the group consisting of hydrogen, C1-10alkyl, C3-12cycloalkyl, C2-10alkenyl, amino, C1-10alkylamino, C3-12cycloalkylamino, benzyl, C3-12 cycloalkenyl, a monocyclic, bicyclic or tricyclic aryl or heteroaryl ring, a hetero-monocyclic ring, a hetero-bicyclic ring system, and a spiro ring system of the formula (III):
Figure US20090253727A1-20091008-C00021
wherein X1 and X2 are independently selected from the group consisting of NH, O, S and CH2;
wherein said alkyl, cycloalkyl, alkenyl, C1-10 alkylamino, C3-12cycloalkylamino, or benzyl is optionally substituted with 1-3 substituents selected from the group consisting of halogen, C1-10 alkyl, C1-10 alkoxy, nitro, trifluoromethyl, cyano, phenyl, benzyl, benzyloxy, said phenyl, benzyl, and benzyloxy optionally being substituted with 1-3 substituents selected from the group consisting of halogen, C1-10 alkyl, C1-10 alkoxy, and cyano;
wherein said C3-12 cycloalkyl, C3-12 cycloalkenyl, monocyclic, bicyclic or tricyclic aryl, heteroaryl ring, hetero-monocyclic ring, hetero-bicyclic ring system, and spiro ring system of the formula (II) are optionally substituted with 1-3 substituents selected from the group consisting of halogen, C1-10 alkyl, C1-10 alkoxy, nitro, trifluoromethyl, phenyl, benzyl, phenyloxy and benzyloxy, wherein said phenyl, benzyl, phenyloxy and benzyloxy are optionally substituted with 1-3 substituents selected from the group consisting of halogen, C1-10 alkyl, C1-10 alkoxy, and cyano;
R2 is selected from the group consisting of hydrogen, C1-10 alkyl, C3-12 cycloalkyl and halogen, said alkyl optionally substituted with an oxo group;
or a pharmaceutically acceptable salt thereof.
40. A compound of claim 39, wherein R1 is alkyl selected from the group consisting; of methyl, ethyl, propyl, butyl, pentyl and hexyl.
41. A compound of claim 39, wherein R1 is cycloalkyl selected from the group consisting of cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, and norbornyl.
42. A compound of claim 39, wherein R1 is tetrahydronaphthyl, decahydronaphthyl or dibenzocycloheptyl.
43. A compound of claim 39, wherein R1 is phenyl or benzyl.
44. A compound of claim 39, wherein R1 is a bicyclic aromatic ring.
45. A compound of claim 44, wherein said bicyclic aromatic ring is indenyl, quinoline or naphthyl.
46. A compound of claim 39, wherein Z is a bond, methyl, or ethyl.
47. A compound of claim 39, wherein n is 0.
48. A compound of claim 39, wherein X1 and X2 are both O.
49. A compound selected from the group consisting of:
1,2,3,4-tetrahydro-1-[1-(naphth-2-yl-methyl)-4-piperidinyl]-quinolin-2-one;
1,2,3,4-tetrahydro-1-[1-(p-phenylbenzyl)-4-piperidinyl]-quinolin-2-one;
1,2,3,4-tetrahydro-1-[1-[4,4-bis(4-fluorophenyl)butyl]-4-piperidinyl]-quinolin-2-one;
1,2,3,4-tetrahydro-1-[1-(p-benzyloxybenzyl)-4-piperidinyl]-quinolin-2-one;
1, 2,3,4-tetrahydro-1-[1-(1,2,3,4-tetrahydro-2-naphthyl)-4-piperidinyl]-quinolin-2-one;
1,2,3,4-tetrahydro-1-[1-(4-propyl-cyclohexyl)-4-piperidinyl]-quinolin-2-one;
1,2,3,4-tetrahydro-1-[1-(5-methylhex-2-yl)-4-piperidinyl]-quinolin-2-one;
1,2,3,4-tetrahydro-1-[1-(norbornan-2-yl)-4-piperidinyl]-quinolin-2-one;
1,2,3,4-tetrahydro-1-[1-(decahydro-2-naphthyl)-4-piperidinyl]-quinolin-2-one;
1,2,3,4-tetrahydro-1-[1-(10,11-dihydro-5H-dibenzo[a.d]-cyclohepten-5-yl)-4-piperidinyl]-quinolin-2-one;
1,2,3,4-tetrahydro-1-[1-(3,3-diphenylpropyl)-4-piperidinyl]-quinolin-2-one;
1,2,3,4-tetrahydro-1-[1-(cyclooctyl)-4-piperidinyl]-quinolin-2-one;
1,2,3,4-tetrahydro-1-[1-[4-(1-methylethyl)-cyclohexyl]-4-piperidinyl]-quinolin-2-one;
1,2,3,4-tetrahydro-1-[1-(1,3-dihydroinden-2-yl)-4-piperidinyl]-quinolin-2-one;
1,2,3,4-tetrahydro-1-[1-(cyclooctylmethyl)-4-piperidinyl]-quinolin-2-one;
and pharmaceutically acceptable salts thereof.
50. A pharmaceutical composition comprising a compound of claim 33 and at least one pharmaceutically acceptable excipient.
51. A method of treating pain comprising administering to a patient in need thereof, an effective amount of an analgesic compound according to claim 33.
52. A method of modulating a pharmacological response from the ORL1 receptor comprising administering to a patient in need thereof, an effective amount of a compound according to claim 33.
53. A pharmaceutical composition comprising a compound of claim 39 and at least one pharmaceutically acceptable excipient.
54. A method of treating pain comprising administering to a patient in need thereof, an effective amount of an analgesic compound according to claim 39.
55. A method of modulating a pharmacological response from the ORL1 receptor comprising administering to a patient in need thereof an effective amount of a compound according to claim 39.
56. A compound of the formula (IIA):
Figure US20090253727A1-20091008-C00022
wherein
R2 is selected from the group consisting of hydrogen, C1-10 alkyl, C3-12 cycloalkyl and halogen, said alkyl is optionally substituted with an oxo group;
n is an integer from 0 to 3;
and ZR1 is
Figure US20090253727A1-20091008-C00023
wherein Y1 is R3—(C1-C12)alkyl, R4-aryl, R5-heteroaryl, R6—(C3-C12)cyano-alkyl, R7—(C3-C7) heterocycloalkyl, —CO2(C1-C6)alkyl, CN or —C(O)NR8R9; Y2 is hydrogen or Y; Y3 is hydrogen or (C1-C6)alkyl; or Y1, Y2 and Y3, together with the carbon to which they are attached, form one of the following structures:
Figure US20090253727A1-20091008-C00024
wherein r is 0 to 3; c and d are independently 1 or 2; s is 1 to 5; and ring E is a fused R4-phenyl or R5-heteroaryl ring;
R10 is 1 to 3 substituents independently selected from the-group consisting of H, (C1-C6)alkyl, —OR9, —(C1-C6)alkyl-OR8, —NR8R9 and —(C1-C6)allyl-NR8R9;
R11 is 1 to 3 substituents independently selected from the group consisting of R10, —CF3, —OCF3, NO2 and halo, or R11 substituents on adjacent ring carbon atoms may together form a methylenedioxy or ethylenedioxy ring;
R8 and R9 are independently selected from the group consisting of hydrogen, (C1-C6) alkyl, (C3-C12) cycloalkyl, aryl and aryl(C1-C6)alkyl;
R3 is 1 to 3 substituents independently selected from the group consisting of H, R4-aryl, R6—(C3-C12) cycloalkyl, R5-heteroaryl, R7—(C3-C7)heterocycloalkyl, —NR8R9, —OR12 and —S(O)0-2R12,
R6 is 1 to 3 substituents independently selected from the group consisting of H, (C1-C6)alkyl, R4-aryl, —NR8R9, —OR12 and —SR12;
R4 is 1 to 3 substituents independently selected from the group consisting of hydrogen, halo, (C1-C6)alkyl, R13-aryl, (C3-C12)cycloalkyl, —CN, —CF3, —OR8, —(C1-C6)alkyl-OR8, —OCF3, NR8R9, —(C1-C6)alkyl-NR8R9, —NHSO2R8, —SO2N(R14)2, —SO2R5, —SOR9, —SR5, —NO2, —CONR8R9, —NR9COR8, —COR8, —COCF3, —OCOR8, —OCO2R8, —COOR8, —(C1-C6)allyl-NHCOOC(CH3)3, —(C1-C6)alkyl-NHCOCF3, —(C1-C6)alkyl-NHSO2—(C1-C6) alkyl, —(C1-C6)alkyl-NHCONH—(C1-C6)-alkyl and
Figure US20090253727A1-20091008-C00025
wherein f is 0 to 6; or R4 substituents on adjacent ring carbon atoms may together form a methylenedioxy or ethylenedioxy ring;
R5 is 1 to 3 substituents independently selected from the group consisting of hydrogen, halo, (C1-C6)alkyl, R13-aryl, (C3-C12)cycloalkyl, —CN, —CF3, —OR9, —(C1-C6)alkyl-OR8, —OCF3, —NR8R9, —(C1-C6)alkyl-NR8R9, —NHSO2R8, —SO2N(R14)2, —NO2, —CONR8R9, —NR9COR8, —COR8, —OCOR8, —OCO2R8 and —COOR8;
R7 is H, (C1-C6)alkyl, —OR8, —(C1-C6)alkyl-OR8, —NR8R9 or —(C1-C6)alkyl-NR8R9;
R12 is H-1, (C1-C6)alkyl, R4-aryl, —(C1-C6)alkyl-OR9, —(C1-C6)alkyl-NR8R9, —(C1-C6)alkyl-SR8, or aryl(C1-C6) alkyl;
R13 is 1-3 substituents independently selected from the group consisting of H, (C1-C6)alkyl, (C1-C6) alkoxy and halo;
R14 is independently selected from the group consisting of H, (C1-C6)alkyl and R13—C6H4—CH2—;
or a pharmaceutically acceptable salt thereof.
57. A pharmaceutical composition comprising a compound of claim 56 and at least one pharmaceutically acceptable excipient.
58. A method of treating pain comprising administering to a patient in need thereof, an effective amount of an analgesic compound according to claim 56.
59. A method of modulating a pharmacological response from the ORL1 receptor comprising administering to a patient in need thereof, an effective amount of a compound according to claim 56.
60. A method of modulating a pharmacological response from an opioid receptor comprising administering to a patient in need thereof, an effective amount of a compound according to claim 33.
61. A method of modulating a pharmacological response from an opioid receptor comprising administering to a patient in need thereof, an effective amount of a compound according to claim 39.
62. A method of modulating a pharmacological response from an opioid receptor comprising administering to a patient in need thereof, an effective amount of a compound according to claim 56.
US12/485,612 2001-04-18 2009-06-16 Nociceptin analogs Abandoned US20090253727A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/485,612 US20090253727A1 (en) 2001-04-18 2009-06-16 Nociceptin analogs

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US28467601P 2001-04-18 2001-04-18
US28467401P 2001-04-18 2001-04-18
US10/126,507 US6861421B2 (en) 2001-04-18 2002-04-18 Nociceptin analogs
US11/069,728 US7563809B2 (en) 2001-04-18 2005-03-01 Nociceptin analogs
US12/485,612 US20090253727A1 (en) 2001-04-18 2009-06-16 Nociceptin analogs

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/069,728 Division US7563809B2 (en) 2001-04-18 2005-03-01 Nociceptin analogs

Publications (1)

Publication Number Publication Date
US20090253727A1 true US20090253727A1 (en) 2009-10-08

Family

ID=26962736

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/126,507 Expired - Lifetime US6861421B2 (en) 2001-04-18 2002-04-18 Nociceptin analogs
US11/069,728 Expired - Fee Related US7563809B2 (en) 2001-04-18 2005-03-01 Nociceptin analogs
US12/485,612 Abandoned US20090253727A1 (en) 2001-04-18 2009-06-16 Nociceptin analogs

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US10/126,507 Expired - Lifetime US6861421B2 (en) 2001-04-18 2002-04-18 Nociceptin analogs
US11/069,728 Expired - Fee Related US7563809B2 (en) 2001-04-18 2005-03-01 Nociceptin analogs

Country Status (14)

Country Link
US (3) US6861421B2 (en)
EP (2) EP1379252A4 (en)
JP (2) JP4380992B2 (en)
KR (3) KR100729242B1 (en)
CN (1) CN100441182C (en)
AU (2) AU2002303406B2 (en)
BR (1) BR0209128A (en)
CA (1) CA2444108C (en)
HK (1) HK1070573A1 (en)
HU (1) HUP0402510A3 (en)
IL (2) IL158486A0 (en)
MX (1) MXPA03009602A (en)
RU (1) RU2268883C2 (en)
WO (1) WO2002085291A2 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014106238A1 (en) * 2012-12-31 2014-07-03 Fang, Qun, Kevin Heterocyclic compounds and methods of use thereof
US10118927B2 (en) 2012-12-27 2018-11-06 Purdue Pharma L.P. Substituted piperidin-4-amino-type compounds and uses thereof
US10793528B2 (en) 2016-01-13 2020-10-06 Grünenthal GmbH 3-((hetero-)aryl)-8-amino-2-oxo-1,3-diaza-spiro-[4.5]-decane derivatives
US10793556B2 (en) 2016-01-13 2020-10-06 Gruenenthal Gmbh 8-amino-2-oxo-1,3-diaza-spiro-[4.5]-decane derivatives
US10807988B2 (en) 2016-01-13 2020-10-20 Grünenthal GmbH 3-((hetero-)aryl)-8-amino-2-oxo-1,3-diaza-spiro-[4.5]-decane derivatives
US10807989B2 (en) 2016-01-13 2020-10-20 Grünenthal GmbH 3-(carboxymethyl)-8-amino-2-oxo-1,3-diaza-spiro-[4.5]-decane derivatives
US10829480B2 (en) 2016-01-13 2020-11-10 Gruenenthal Gmbh 3-((hetero-)aryl)-alkyl-8-amino-2-oxo-l,3-diaza-spiro-[4.5]-decane derivatives

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7501417B2 (en) * 2002-03-13 2009-03-10 Janssen Pharmaceutica, N.V. Aminocarbonyl-derivatives as novel inhibitors of histone deacetylase
JP4056977B2 (en) 2002-03-29 2008-03-05 田辺三菱製薬株式会社 Sleep disorder drug
WO2008073958A2 (en) 2006-12-12 2008-06-19 Wyeth Substituted benzothiadiazinedioxide derivatives and methods of their use
CL2007003590A1 (en) * 2006-12-12 2008-02-29 Wyeth Corp COMPOUNDS DERIVED FROM ARIL SULFAMIDA; PREPARATION PROCEDURE; PHARMACEUTICAL COMPOSITION THAT INCLUDES SUCH COMPOUNDS; AND ITS USE IN PREVENTION AND TREATMENT OF VASOMOTRIC SYMPTOMS, SEXUAL DYSFUNCTION, GASTROINTESTINAL DISORDERS, TRANST
NZ601683A (en) * 2007-01-16 2012-11-30 Shionogi & Co Heterocyclic-substituted piperidine compounds for modulating ORL-1 receptor function in a cell
WO2009027820A2 (en) 2007-08-31 2009-03-05 Purdue Pharma L.P. Substituted-quinoxaline-type-piperidine compounds and the uses thereof
AR072578A1 (en) 2008-07-21 2010-09-08 Purdue Pharma Lp COMPOSED PIPERIDINE BRIDGES REPLACED TYPE QUINOXALINE AND USES OF THE SAME
JP5872585B2 (en) * 2010-12-22 2016-03-01 パーデュー、ファーマ、リミテッド、パートナーシップ Phosphorus-substituted quinoxaline-type piperidine compounds and uses thereof
WO2014102589A1 (en) 2012-12-27 2014-07-03 Purdue Pharma L.P. Quinazolin-4(3h)-one-type piperidine compounds and uses thereof
US9963458B2 (en) 2012-12-27 2018-05-08 Purdue Pharma L.P. Indole and indoline-type piperidine compounds and uses thereof
WO2014102594A2 (en) 2012-12-27 2014-07-03 Purdue Pharma L.P. Substituted benzimidazole-type piperidine compounds and uses thereof

Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3161645A (en) * 1962-12-18 1964-12-15 Res Lab Dr C Janssen N V 1-(1-aroylpropyl-4-piperidyl)-2-benzimidazolinones and related compounds
US3318900A (en) * 1964-05-06 1967-05-09 Janssen Pharmaceutica Nv Derivatives of benzimidazolinyl piperidine
US3325499A (en) * 1964-11-02 1967-06-13 Mcneilab Inc 1-(1-hydrocarbyl-4-piperdyl)-2-indolinone
US4215119A (en) * 1976-03-09 1980-07-29 Boehringer Ingelheim Gmbh Aminoalkyl-substituted benzimidazolidin-2-ones
US4329353A (en) * 1980-10-22 1982-05-11 Janssen Pharmaceutica, N.V. 1-(4-Aryl-cyclohexyl)piperidine derivatives, method of use thereof and pharmaceutical compositions thereof
US4344945A (en) * 1980-03-10 1982-08-17 Kyowa Hakko Kogyo Co., Ltd. Piperidine derivatives
US4410528A (en) * 1980-05-16 1983-10-18 Kyowa Hakko Kogyo Co., Ltd. Hypotensive piperidine derivatives
US5574044A (en) * 1994-10-27 1996-11-12 Merck & Co., Inc. Muscarine antagonists
US5661169A (en) * 1994-07-05 1997-08-26 Sanofi 1-benzyl-1,3-dihydro-2H-benzimidazol-2-one derivatives, their preparation and the pharmaceutical compositions containing them
US5760054A (en) * 1994-04-14 1998-06-02 Merck & Co., Inc. Alpha IC adrenergic receptor antagonists
US5767118A (en) * 1994-10-26 1998-06-16 Merck & Co., Inc. 4-Heterocyclic peperidines promote release of growth hormone
US6063796A (en) * 1997-04-04 2000-05-16 Merck & Co., Inc. Somatostatin agonists
US6166037A (en) * 1997-08-28 2000-12-26 Merck & Co., Inc. Pyrrolidine and piperidine modulators of chemokine receptor activity
US6172067B1 (en) * 1998-08-06 2001-01-09 Pfizer Inc. 2-substituted-1-piperidyl benzimidazole compounds as ORL1-receptor agonists

Family Cites Families (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0029707B1 (en) * 1979-11-21 1984-02-01 Kyowa Hakko Kogyo Co., Ltd Novel piperidine derivatives, method for the preparation thereof and pharmaceutical compositions containing them
JPS5759889A (en) * 1980-09-30 1982-04-10 Kyowa Hakko Kogyo Co Ltd Novel piperidine derivative
JPS58180481A (en) 1982-04-15 1983-10-21 Kyowa Hakko Kogyo Co Ltd Novel piperidine derivative
JPS60120872A (en) * 1983-12-01 1985-06-28 Kyowa Hakko Kogyo Co Ltd Novel heterocyclic compound and cardiotonic agent
EP0382185B1 (en) * 1989-02-10 1994-06-15 Otsuka Pharmaceutical Co., Ltd. Carbostyril derivatives
WO1995019773A1 (en) * 1992-10-07 1995-07-27 Merck & Co., Inc. Carbostyril oxytocin receptor antagonists
ATE216580T1 (en) * 1993-07-16 2002-05-15 Merck & Co Inc BENZOXAZINONE AND BENZOPYRIMIDINONE PIPERIDINYL COMPOUNDS AS TOCOLYTIC OXYTOCIN RECEPTOR ANTAGONISTS
JPH11507670A (en) 1995-06-12 1999-07-06 ジー.ディー.サール アンド カンパニー Treatment of inflammation and inflammation-related diseases with a combination of a cyclooxygenase-2 inhibitor and a 5-lipoxygenase inhibitor
CA2231773A1 (en) 1995-09-13 1997-03-20 Shigeru Aono Phenylpiperidine derivative
DE69736776T2 (en) 1996-04-19 2007-01-18 Neurosearch A/S 1- (4-PIPERIDYL) -BENZIMIDAZOLE WITH NEUROTROPHIC ACTIVITY
KR20010020172A (en) 1997-05-30 2001-03-15 나가사까 겐지로 2-oxoimidazole derivatives
JPH10330377A (en) * 1997-06-02 1998-12-15 Kyowa Hakko Kogyo Co Ltd Piperidine derivative
SI0921125T1 (en) 1997-12-05 2002-04-30 F. Hoffmann-La Roche Ag 1,3,8-Triazaspiro(4,5)decan-4-on derivatives
US6166209A (en) 1997-12-11 2000-12-26 Hoffmann-La Roche Inc. Piperidine derivatives
BR9907104A (en) 1998-01-19 2000-10-24 Pfizer 4- (2-Keto-1-benzimidazolinyl) piperidine compounds as orl1 receptor agonists
AU754716B2 (en) 1998-03-26 2002-11-21 Japan Tobacco Inc. Amide derivatives and nociceptin antagonists
JP2002515503A (en) 1998-05-18 2002-05-28 ノボ ノルディスク アクティーゼルスカブ Novel 1,3,8-triazaspiro [4,5] decanone with high affinity for opioid receptor subtypes
US6262066B1 (en) 1998-07-27 2001-07-17 Schering Corporation High affinity ligands for nociceptin receptor ORL-1
ID29137A (en) 1998-07-27 2001-08-02 Schering Corp HIGH AFINITY LIGANS FOR ORL-1 NOSISEPTIN RECEPTORS
JP2001044213A (en) * 1999-07-30 2001-02-16 Hitachi Cable Ltd Epitaxial wafer for hetero-bipolar transistor
SE9904652D0 (en) * 1999-12-17 1999-12-17 Astra Pharma Prod Novel Compounds
CA2400640A1 (en) 2000-02-18 2001-08-23 Meiji Seika Kaisha, Ltd. Phenoxyalkylamine derivatives useful as opioid .delta. receptor ligands
BRPI0113286B8 (en) * 2000-08-14 2021-05-25 Johnson & Johnson substituted pyrazoles and pharmaceutical composition comprising the same.
MXPA03001963A (en) 2000-09-06 2004-03-18 Johnson & Johnson A method for treating allergies using substituted pyrazoles.
JP2007009500A (en) * 2005-06-30 2007-01-18 Nissei Ltd Operating method of puzzle circulating type parking facility

Patent Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3161645A (en) * 1962-12-18 1964-12-15 Res Lab Dr C Janssen N V 1-(1-aroylpropyl-4-piperidyl)-2-benzimidazolinones and related compounds
US3318900A (en) * 1964-05-06 1967-05-09 Janssen Pharmaceutica Nv Derivatives of benzimidazolinyl piperidine
US3325499A (en) * 1964-11-02 1967-06-13 Mcneilab Inc 1-(1-hydrocarbyl-4-piperdyl)-2-indolinone
US4215119A (en) * 1976-03-09 1980-07-29 Boehringer Ingelheim Gmbh Aminoalkyl-substituted benzimidazolidin-2-ones
US4344945A (en) * 1980-03-10 1982-08-17 Kyowa Hakko Kogyo Co., Ltd. Piperidine derivatives
US4410528A (en) * 1980-05-16 1983-10-18 Kyowa Hakko Kogyo Co., Ltd. Hypotensive piperidine derivatives
US4329353A (en) * 1980-10-22 1982-05-11 Janssen Pharmaceutica, N.V. 1-(4-Aryl-cyclohexyl)piperidine derivatives, method of use thereof and pharmaceutical compositions thereof
US5760054A (en) * 1994-04-14 1998-06-02 Merck & Co., Inc. Alpha IC adrenergic receptor antagonists
US5661169A (en) * 1994-07-05 1997-08-26 Sanofi 1-benzyl-1,3-dihydro-2H-benzimidazol-2-one derivatives, their preparation and the pharmaceutical compositions containing them
US5767118A (en) * 1994-10-26 1998-06-16 Merck & Co., Inc. 4-Heterocyclic peperidines promote release of growth hormone
US5574044A (en) * 1994-10-27 1996-11-12 Merck & Co., Inc. Muscarine antagonists
US6063796A (en) * 1997-04-04 2000-05-16 Merck & Co., Inc. Somatostatin agonists
US6166037A (en) * 1997-08-28 2000-12-26 Merck & Co., Inc. Pyrrolidine and piperidine modulators of chemokine receptor activity
US6172067B1 (en) * 1998-08-06 2001-01-09 Pfizer Inc. 2-substituted-1-piperidyl benzimidazole compounds as ORL1-receptor agonists

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10118927B2 (en) 2012-12-27 2018-11-06 Purdue Pharma L.P. Substituted piperidin-4-amino-type compounds and uses thereof
WO2014106238A1 (en) * 2012-12-31 2014-07-03 Fang, Qun, Kevin Heterocyclic compounds and methods of use thereof
JP2016508142A (en) * 2012-12-31 2016-03-17 サノビオン・ファーマシューティカルズ・インコーポレイテッドSunovion Pharmaceuticals Inc. Heterocyclic compounds and methods of use thereof
US9884844B2 (en) 2012-12-31 2018-02-06 Sunovion Pharmaceuticals, Inc. Heterocyclic compounds and methods of use thereof
US10793528B2 (en) 2016-01-13 2020-10-06 Grünenthal GmbH 3-((hetero-)aryl)-8-amino-2-oxo-1,3-diaza-spiro-[4.5]-decane derivatives
US10793556B2 (en) 2016-01-13 2020-10-06 Gruenenthal Gmbh 8-amino-2-oxo-1,3-diaza-spiro-[4.5]-decane derivatives
US10807988B2 (en) 2016-01-13 2020-10-20 Grünenthal GmbH 3-((hetero-)aryl)-8-amino-2-oxo-1,3-diaza-spiro-[4.5]-decane derivatives
US10807989B2 (en) 2016-01-13 2020-10-20 Grünenthal GmbH 3-(carboxymethyl)-8-amino-2-oxo-1,3-diaza-spiro-[4.5]-decane derivatives
US10829480B2 (en) 2016-01-13 2020-11-10 Gruenenthal Gmbh 3-((hetero-)aryl)-alkyl-8-amino-2-oxo-l,3-diaza-spiro-[4.5]-decane derivatives

Also Published As

Publication number Publication date
HK1070573A1 (en) 2005-06-24
HUP0402510A3 (en) 2012-08-28
MXPA03009602A (en) 2004-04-02
WO2002085291A2 (en) 2002-10-31
RU2268883C2 (en) 2006-01-27
JP2009280603A (en) 2009-12-03
KR20050118315A (en) 2005-12-16
CN1568188A (en) 2005-01-19
KR100722207B1 (en) 2007-05-29
JP2005506302A (en) 2005-03-03
US7563809B2 (en) 2009-07-21
RU2003133454A (en) 2005-05-27
KR100855204B1 (en) 2008-09-01
KR20060129103A (en) 2006-12-14
CA2444108C (en) 2012-07-24
EP1379252A4 (en) 2005-11-02
CN100441182C (en) 2008-12-10
KR20030096326A (en) 2003-12-24
US6861421B2 (en) 2005-03-01
WO2002085291A3 (en) 2003-02-20
KR100729242B1 (en) 2007-06-15
HUP0402510A2 (en) 2005-04-28
CA2444108A1 (en) 2002-10-31
EP1997818A3 (en) 2009-04-08
AU2006202794A1 (en) 2006-07-20
US20030013874A1 (en) 2003-01-16
AU2002303406B2 (en) 2006-03-30
JP4380992B2 (en) 2009-12-09
IL158486A (en) 2010-11-30
IL158486A0 (en) 2004-05-12
BR0209128A (en) 2005-11-01
EP1997818A2 (en) 2008-12-03
EP1379252A2 (en) 2004-01-14
US20050148580A1 (en) 2005-07-07

Similar Documents

Publication Publication Date Title
US8252815B2 (en) Nociceptin analogs
US7563809B2 (en) Nociceptin analogs
US6828440B2 (en) Spiroindene and spiroindane compounds
US6635653B2 (en) Spiropyrazole compounds
US20050192307A1 (en) Benzimidazolone compounds
AU2002338424A1 (en) Benzimidazolone compounds
AU2002303406A1 (en) Nociceptin analogs

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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