AU2002338424A1 - Benzimidazolone compounds - Google Patents

Benzimidazolone compounds

Info

Publication number
AU2002338424A1
AU2002338424A1 AU2002338424A AU2002338424A AU2002338424A1 AU 2002338424 A1 AU2002338424 A1 AU 2002338424A1 AU 2002338424 A AU2002338424 A AU 2002338424A AU 2002338424 A AU2002338424 A AU 2002338424A AU 2002338424 A1 AU2002338424 A1 AU 2002338424A1
Authority
AU
Australia
Prior art keywords
alkyl
benzimidazol
octahydro
piperidinyl
group
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.)
Granted
Application number
AU2002338424A
Other versions
AU2002338424B2 (en
Inventor
Zhengming Chen
R. Richard Goehring
Donald Kyle
Sam Victory
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.)
Euro Celtique SA
Original Assignee
Euro Celtique SA
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 Euro Celtique SA filed Critical Euro Celtique SA
Priority claimed from PCT/US2002/012375 external-priority patent/WO2002085361A1/en
Publication of AU2002338424A1 publication Critical patent/AU2002338424A1/en
Application granted granted Critical
Publication of AU2002338424B2 publication Critical patent/AU2002338424B2/en
Assigned to EURO-CELTIQUE S.A. reassignment EURO-CELTIQUE S.A. Alteration of Name(s) in Register under S187 Assignors: EURO-CELTIQUE, S.A.
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Description

BENZIMIDAZOLONE COMPOUNDS
This application claims priority from U.S. Provisional Application Serial No. 60/284,665, filed April 18, 2001, the disclosure of which is 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 K. 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 (ORLl) 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 K receptors had low affinity for the ORLl . 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 ORLl receptor. This ligand is a seventeen amino acid peptide structurally similar to members of the opioid peptide family.
The discovery of the ORLl 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 ORLl receptor.
It is an object of certain embodiments of the present invention to provide new compounds which exhibit affinity for the ORLl receptor and one or more of the μ, δ or K 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 ORLl 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 K 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, d and k 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 K 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, neuroprotective agents, anti- hypertensives, anti-anxioltics; agents for appetite control; hearing regulators; anti-tussives, anti-asthmatics, modulators of locomotor activity, modulators of learning and memory, regulators of neuro transmitter 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 K) 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 μ 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):
(I)
wherein
A is a saturated or partially saturated ring;
R is hydrogen, C,.10 alkyl, C3.|2 cycloalkyl, C3.12 cycloalkylC,.4alkyl-, C,.10 alkoxy, C3.|2 cycloalkoxy-, C,.,„ alkyl substituted with 1-3 halogen, C3.12 cycloalkyl substituted with 1-3 halogen, C3.,2 cycloalkylC,.4alkyl- substituted with 1-3 halogen, C,.,0 alkoxy substituted with 1-3 halogen, C3.12 cycloalkoxy- substituted with 1-3 halogen, -COON,, -C COOV„ -
CH2OH, -SO2Ν(V,)2 , hydroxyC^oalkyl-, hydroxyC3.10cycloalkyl-, cyanoC,.10alkyl-, cyanoC3. 10cycloalkyl-, -CON(V,)2, NH2SO2CMalkyl-, NH2SOCMalkyl-, sulfonylaminoC,.10alkyl-, diaminoalkyl-, -sulfonylC alkyl, a 6-membered heterocyclic ring, a 6-membered heteroaromatic ring, a 6-membered heterocyclicC alkyl-, a 6-membered heteroaromaticC,. 4alkyl-, a 6-membered aromatic ring, a 6-membered aromaticC alkyl-, a 5-membered heterocyclic ring optionally substituted with an oxo or thio, a 5-membered heteroaromatic ring, a 5-membered heterocyclicCMalkyl- optionally substituted with an oxo or thio, a 5- membered heteroaromaticC alkyl-, -C,.5(=O)W„ -CI.5(=NH)W„ -C,.sNHC(=O)W„ -C 5NHS(=O)2W„ -C,.jNHS(=O)W„ wherein W, is hydrogen, CM0 alkyl, C3.12 cycloalkyl, C,.,0 alkoxy, C3.12 cycloalkoxy, -CH2OH, amino, C|.4alkylamino-, diC1.4alkylamino-, or a 5- membered heteroaromatic ring optionally substituted with 1-3 lower alkyl; wherein each V, is independently selected from H, Ct.6 alkyl, C3.6 cycloalkyl, benzyl and phenyl; n is an integer from 0 to 3;
M,, M2, M3 and M4 are each independently N, NH, CH or CH2, up to a maximum of 3 N or NH;
D, B and C are independently hydrogen, C 0 alkyl, C3.,2 cycloalkyl, C,.,0 alkoxy, C3.12 cycloalkoxy, -CH2OH, -NHSO2, hydroxyC Oalkyl-, aminocarbonyl-, C,.4alkylaminocarbonyl-, diCMalkylaminocarbonyl-, acylamino-, acylaminoalkyl-, amide, sulfonylaminoC,.10alkyl-, or D- B can together form a C2.6 bridge, or B-C can together form a C3.7 bridge, or D-C can together form a C,.5 bridge;
Z is selected from the group consisting of a bond, straight or branched C,_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;
R, is selected from the group consisting of hydrogen, C,.10 alkyl, C3.12cycloalkyl, C2. 10alkenyl, amino, C,.10alkylamino-, C3.12cycloalkylamino-, -COOV,, -CMCOOV, , cyano, cyanoC,.10alkyl-, cyanoC3.10cycloalkyl-, NH2SO2-, NH2SO2C,.4alkyl-, NH2SOC1.4alkyl-, aminocarbonyl-, C,.4alkylaminocarbonyl-, diCMalkylaminocarbonyl-, 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 (II):
(II) wherein X, and X2 are independently selected from the group consisting of NH, O, S and CH2; and wherein said alkyl, cycloalkyl, alkenyl, C,.10alkylamino-, C3.12cycloalkylamino-, or benzyl of R, is optionally substituted with 1 -3 substituents selected from the group consisting of halogen, hydroxy, C,.l0 alkyl, CM0 alkoxy, nitro, trifluoromethyl-, cyano, - COOV,, -C COOV„ cyanoC Oalkyl-, -C,.s(=O)W„ -C,.5NHS(=O)2W„ -C,.5NHS(=O)W„ 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, C,.,0 alkyl-, C,.10 alkoxy-, and cyano; and wherein said C3.,2 cycloalkyl, C3.,2 cycloalkenyl, monocyclic, bicyclic or tricyclic aryl, heteroaryl ring, hetero-monocyclic ring, hetero-bicyclic ring system, or spiro ring system of the formula (II) is optionally substituted with 1-3 substituents selected from the group consisting* of halogen, C,.,0 alkyl, C,. 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, C,.,0 alkyl, CM0 alkoxy, and cyano;
R2 is selected from the group consisting of hydrogen, C,.,0 alkyl, C3.,2 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):
(IA)
wherein
A is a saturated or partially saturated ring; 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;
R is selected from the group consisting of hydrogen, C,.10 alkyl, C,.,0 alkoxy, and C3. ,2cycloalkyl;
R, is selected from the group consisting of hydrogen, C,.10alkyl, C3.,2cycloalkyl, C2.10alkenyl, amino, C,.,0alkylamino, C3.12cycloalkylamino, benzyl, C3.,2 cycloalkenyl, a monocyclic, bicyclic or tricyclic aryl or heteroaryl ring, a hetero-moncyclic ring, a heterobicyclic ring system, and a spiro ring system of the formula (II):
wherein X, 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, C,.10alkylamino, C3.,2cycloalkylamino, or benzyl is optionally substituted with 1-3 substituents selected from the group consisting of halogen, C,.10 alkyl, C,.,0 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,.,0 alkyl, C,.)0 alkoxy, and cyano; wherein said C3.,2 cycloalkyl, C3.12 cycloalkenyl, monocyclic, bicyclic or tricyclic aryl, heteroaryl ring, hetero-monocyclic ring, heterobicyclic ring system, and spiro ring system of the formula (II) are optionally substituted with 1 -3 substituents selected from the group consisting of halogen, C,.,0 alkyl, C,.,0 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,.,0 alkyl, C,. ,0 alkoxy, and cyano;
R2 is selected from the group consisting of hydrogen, C,.,0 alkyl, C3.12 cycloalkyl and halogen, said alkyl optionally substituted with an oxo group; and pharmaceutically acceptable salts thereof.
In certain preferred embodiments of formula (I) or (IA), the R, alkyl is methyl, ethyl, propyl, butyl, pentyl, or hexyl.
In certain preferred embodiments of formula (I) or (IA), the R, cycloalkyl is cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, or norbornyl.
In other preferred embodiments of formula (I) or (IA), the R, bicyclic ring system is naphthyl. In other preferred embodiments of formula (I) or (IA), the R, bicyclic ring system is tetrahydronaphthyl, or decahydronaphthyl and the R, tricyclic ring system is dibenzocycloheptyl. In other preferred embodiments R, is phenyl or benzyl.
In other preferred embodiments of formula (I) or (IA), the R, bicyclic aromatic ring is a 10-membered ring, preferably quinoline or naphthyl.
In other preferred embodiments of formula (I) or (IA), the R, bicyclic aromatic ring is a 9-membered ring, preferably indenyl.
In certain embodiments of formula (I) or (IA), Z is a bond, methyl, or ethyl.
In certain embodiments of formula (I) or (IA), 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) or (IA), n is 0.
In certain embodiments, A is a saturated ring.
In certain embodiments of formula (I) or (IA), X, and X2 are both O.
In certain embodiments of formula (I), R is -CH2C=ONH2, -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), ZR, 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 (I), at least one of ZR, or R is -CH2COOV„ tetrazolylmethyl-, cyanomethyl-, NH2SO2methyl-, NH2SOmethyl-, aminocarbonylmethyl-, C,. 4alkylaminocarbonylmethyl-, or diC,.4alkylaminocarbonylmethyl-.
In certain embodiments of formula (I), ZR, is 3,3 diphenylpropyl optionally substituted at the 3 carbon of the propyl with -COOV,, tetrazolylC0.4alkyl-, cyano-, aminocarbonyl-, C,_ 4alkylaminocarbonyl-, or diC,.4alkylaminocarbonyl-.
In alternate embodiments in formula (I) or (IA), ZR, can be the following
wherein
Y, is R3-(C,-C12)alkyl, R4-aryl, R5-heteroaryl, R6-(C3-C,2)cyclo-alkyl, R7-(C3- C7)heterocycloalkyl, -CO2(C,-C6)alkyl, CN or -C(O)NR8R9; Y2 is hydrogen or Y,; Y3 is hydrogen or (C,-C6)alkyl; or Y„ Y2 and Y3, together with the carbon to which they are attached, form one of the following structures:
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, (C,- C6)alkyl, -OR8, - (C,-C6)alkyl-ORg, -NR^and -(C C alkyl-NRgRg;
Rπ is 1 to 3 substituents independently selected from the group consisting of R10, -CF3, -OCF3, NO2 and halo, or Rn substituents on adjacent ring carbon atoms may together form a methylenedioxy or ethylenedioxy ring;
R8and R, are independently selected from the group consisting of hydrogen, (C,-C6) alkyl, (C3-C,2)cycloalkyl, aryl and aryl(C,-C6)alkyl;
R3 is 1 to 3 substituents independently selected from the group consisting of H, R4- aryl, RH -C12)cycloalkyl, R5-heteroaryl, R7-(C3 -C7)heterocycloalkyl, -NR8 R,, -OR,2and - S(O)0.2R,2; Ra, is 1 to 3 substituents independently selected from the group consisting of H, (C,- C6)alkyl, R4-aryl, -NRgR, , -OR12 and -SR12;
R4 is 1 to 3 substituents independently selected from the group consisting of hydrogen, halo, (C,- C6 )alkyl, R13 -aryl, (C3 - C12)cycloalkyl, -CN, -CF3, -OR8, -(C,-C6)alkyl-OR8, - OCF3, -NRgRg, -(C, - C6)alkyl -NR8R<„ -NHSO2R8, -SO2N(R14)2, -SO2R8, -SOR8, -SR8, -NO2, -CONRgR,, -NR9COR8, -CORg, -COCF3, -OCOR8, -OCO2R8, -COOR8, -(C,-C6)alkyl- NHCOOC(CH3)3, -(C,-C6)alkyl-NHCOCF3, -(C,-C6)alkyl-NHSO2-(C,-C6)alkyl, -(C,-C6)alkyl- NHCONH-(C,-C6)-alkyl and
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, (C,-C6)alkyl, R13-aryl, (C3-C12)cycloalkyl, -CN, -CF3, -OR8, -(C,-C6)alkyl-OR8, -OCF3,- NRg ,, -NHSO2R8, -SO2N(R14)2, -NO2, -CONRgRg, -COR8, -OCORg, -OCO2R8and -COOR8;
R7 is H, (C,-C6)alkyl, -OR8, -(C,-C6)alkyl-OR8, -NRgRg or
RI2 is H, (C,-C6)alkyl, R4-aryl, -(C,-C6)alkyl-OR8, -(C.-C alkyl-NRgR,, -(C,-C6)alkyl- SR8, or aryl (C,-C6)alkyl;
R,3 is 1-3 substituents independently selected from the group consisting of H, (C,- C6)alkyl, (C,-C6)alkoxy and halo;
R14 is independently selected from the group consisting of H, (C,-C6)alkyl and R,3- 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 benzo thiazolyl. 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, benzdioxole and benzothiophene.
As used herein, the term "heterocyclicC,.4alkyl", "heteroaromaticC,.4alkyl" and the like refer to the ring structure bonded to a C 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 Η, Η, 13C, l4C, 15N, 18O, 170, 31P, 32P, 35S, ,8F, 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 of the invention include: l-[l-benzyl-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H-benzimidazol-2-one; l-[l-(naphth-2-yl-methyl)-4-piperidinyl]-trans-l, 3,4,5, 6,7,8,9-octahydro-2H- benzimidazol-2-one;
l-[l-(3,3-Bis(phenyl)propyl)-4-piperidinyl]-trans-l, 3,4,5,6, 7,8,9-octahydro-2H- benzimidazol-2-one; l-[l-(4-propylcyclohexyl)-4-piperidinyl]-trans-l, 3,4,5, 6,7,8,9-octahydro-2H- benzimidazol-2-one; l-[l-(5-methylhex-2-yl)-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one; l-[l-(decahydro-2-naphthyl)-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one; l-[l-(cyclooctyl)-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H-benzimidazol-2- one; l-[l-[4-(l-methylethyl)-cyclohexyl]-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one; l-[l-(cyclooctylmethyl)-4-piperidinyl]-trans-l, 3,4,5,6,7,8, 9-octahydro-2H- benzimidazol-2-one;
3-ethyl-l-[l-(benzyl)-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H-benzimidazol- 2-one;
3-ethyl-l-[l-(naphth-2-yl-methyl)-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one ;
3 -ethyl- 1 - [ 1 -(3 ,3 -Bis(phenyl)propyl)-4-piperidinyl] -trans- 1 ,3 ,4,5 ,6,7,8 ,9-octahydro- 2H-benzimidazol-2-one;
3-ethyl-l-[l-(4-propylcyclohexyl)-4-piperidinyl]-trans- 1,3,4, 5,6,7,8, 9-octahydro-2H- benzimidazol-2-one;
3-ethyl-l-[l-(5-methylhex-2-yl)-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one;
3-ethyl-l-[l-(decahydro-2-naphthyl)-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro- 2H-benzimidazol-2-one;
3 -ethyl- 1 - [ 1 -(cyclooctyl)-4-piperidinyl]-trans- 1 ,3 ,4, 5 ,6,7, 8 ,9-octahydro-2H- benzimidazol-2-one;
3-ethyl-l-[l-(4-propylcyclohexyl)-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one;
3-ethyl-l-[l-(cyclooctylmethyl)-4-piperidinyl]-trans-l, 3,4,5,6,7,8, 9-octahydro-2H- benzimidazol-2-one; l-[l-benzyl-4-piperidinyl]-cis-l, 3,4,5,6,7,8, 9-octahydro-2H-benzimidazol-2-one; l-[l-(naphth-2-yl-methyl)-4-piperidinyl]-cis-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one; l-[l-(3,3-Bis(phenyl)propyl)-4-piperidinyl]-cis-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one; l-[l-(4-propylcyclohexyl)-4-piperidinyl]-cis-l, 3,4,5,6,7,8, 9-octahydro-2H- benzimidazol-2-one; l-[l-(5-methylhex-2-yl)-4-piperidinyl]-cis-l,3,4,5,6,7,8,9-octahydro-2H-benzimidazol- 2-one; l-[l-(decahydro-2-naphthyl)-4-piperidinyl]-cis-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one; l-[l-(cyclooctyl)-4-piperidinyl]-cis-l, 3,4,5,6, 7, 8,9-octahydro-2H-benzimidazol-2-one; l-[l-[4-(l-methylethyl)-cyclohexyl]-4-piperidinyl]-cis-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one; l-[l-(cyclooctylmethyl)-4-piperidinyl]-cis-l,3,4,5,6,7,8,9-octahydro-2H-benzimidazol- 2-one;
3-ethyl-l-[l-(benzyl)-4-piperidinyl]-cis-l, 3,4,5,6,7, 8, 9-octahydro-2H-benzimidazol-2- one;
3-ethyl-l-[l-(naphth-2-yl-methyl)-4-piperidinyl]-cis-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one;
3 -ethyl- 1 - [ 1 -(3 ,3 -Bis(phenyl)propyl)-4-piperidinyl] -cis- 1 ,3 ,4,5 ,6,7, 8,9-octahydro-2H- benzimidazol-2-one;
3-ethyl-l-[l-(4-propylcyclohexyl)-4-piperidinyl]-cis-l, 3,4,5, 6,7,8, 9-octahydro-2H- benzimidazol-2-one;
3-ethyl-l-[l-(5-methylhex-2-yl)-4-piperidinyl]-cis-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one;
3-ethyl-l-[l-(decahydro-2-naphthyl)-4-piperidinyl]-cis-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one;
3-ethyl-l-[l-(cyclooctyl)-4-piperidinyl]-cis-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one;
3-ethyl-l-[l-(4-propylcyclohexyl)-4-piperidinyl]-cis-l, 3,4,5,6,7,8, 9-octahydro-2H- benzimidazol-2-one;
3-ethyl-l-[l-(cyclooctylmethyl)-4-piperidinyl]-cis-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-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 ORLl 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 arid/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, papaveretum, 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. Patent 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 antifϊnflammatory 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 (Perry B. Molinhoff and Raymond W. Ruddon, Eds., Ninth Edition, 1996), and Glen R. Hanson Analgesic, Antipyretic and Anit-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, mephenytoin, mephobarbital, metharbital, methetoin, methsuximide, 5-methyl-5-(3- phenanthryl)-hydantoin, 3-methyl-5-phenylhydantoin, narcobarbital, nimetazepam, nitrazepam, oxcarbazepine, paramethadione, phenacemide, phenetharbital, pheneturide, phenobarbital, phensuximide, phenylmethylbarbituric acid, phenytoin, 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 perhexiline.
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; anthramycin; 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; flurocitabine; 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-nl ; 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-l,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; cyclopentanthraquinones; 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; fenretinide; 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; immunostimulant 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 inhibitor; 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 inhibitors; 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 Bl ; 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; sizofiran; 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; thrombopoietin; 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 g/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 K, 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, can be measured by one skilled in the art by an assay utilizing membranes from recombinant HEK-293 cells expressing the human opioid receptorlike 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 "trans" head groups.
NaBH(OAc)3
NaH, Etl DMF
Procedure:
A mixture of compound 1 (22.8 g, 200 mmol), compound 2 (19.9 g, 100 mmol) and Na(OAc)3BH (29.7 g, 140 mmol) in 300 mL 1 ,2-dichloroethane was stirred overnight. The reaction was quenched with aqueous K2CO3. The mixture was extracted with Et2O (3x), and the organic extracts were dried over K2CO3, filtered and the solvent was evaporated. The crude product was purified by column chromatography (10% Et3N, 40% EtOAc in hexane, then 10% Et3N, 90% EtOAc, and then 10% MeOH, 90% EtOAc) to give pure 3 as a solid (18.80 g, 63.6%).
MS: m/z 298.3 (M+l)
'H-NMR (CDC13): d 0.90 (m, 1H), 1.05-1.40 (m, 6H), 1.45 (s, 9H), 1.68-1.80 (m, 5H), 1.88 (m, 2 H), 2.05 (m, 1H), 2.15 (m, 1H), 2.30 (m, 1H), 2.70-2.90 (m, 3H), 4.00 (b, 2H).
To a solution of compound 3 (18.8 g, 63 mmol) in 50 mL of CH3CN was added 1,1 '- carbonyldiimidazole (12.82 g, 79 mmol). Gas was evolved and the mixture became a slurry in a few min and then a solid after 10 min. After 2 hr, the solid was dissolved in CH2C12 and washed with H2O, dried over K2CO3 and filtered. The solvent was evaporated to give 4 as a white solid (18.0 g, 88.5%)). This material was used directly in subsequent next steps. 'H-NMR (CDCI3): d 1.30-1.50 (m, 13H), 1.55-1.75 (m, 8H), 1.80 (m, 2H), 1.95 (m, 1H), 2.15 (m, 1H), 2.98 (m, 2H), 3.85 (m, 1H), 4.50 (bs, 1H).
To a solution of compound 4 (6.25 g, 19.3 mmol) in 70 mL of CH2C12 was added 20 mL of TFA. The mixture was stirred for 2 hr and then the solvent and TFA were evaporated. The residue was dissolved in CH2C12, washed with saturated aqueous K2CO3 solution. The organic layer was dried over K2CO3 and filtered. Evaporation of solvent gave 5 as a solid (3.50 g, 81.2%). MS: m/z 224.2 (M+l)
'H-NMR (CDCl3): d 1.30-1.50 (m, 4H), 1.60-1.85 (m, 6H), 1.90 (b, 1H), 1.25 (b, 1H), 1.65 (dq, 2H), 2.90-3.20 (m, 4H), 4.75 (m, 1H), 4.45 (b, 1H).
To a suspension of NaH (60% oil dispersion, 1.0 g, 24 mmol) in 30 mL of DMF was added compound 4 (6.46 g, 20 mmol) in 15 mL of DMF. After 5 minutes, Etl (3.70 g, 24 mmol) was added to the reaction mixture. The mixture was stirred overnight, quenched with H2O and extracted with CH2C12. The organic extracts were dried over K2CO3 and filtered. Evaporation of the solvent provided 6 (6.20 g, 88.2%). This material was used directly in the next step. LC: 94.6%
MS: m/z 352.2 (M+l)
'H -NMR (CDC13): d 1.07 (t, 3H), 1.30-1.40 (m, 4H), 1.50 (s, 9H), 1.60-1.77 (m,6H), 1.80 (m,
2H), 2.05 (m, 1H), 2.15 (m, 1H), 2.70-2.90 (m, 4H), 3.20 (m, 2H), 3.90 (m, 1H).
To a solution of compound 6 (6.10 g, 17.36 mmol) in 70 mL of CH2C12 was added 20 mL of TFA. The mixture was stirred for 2 hr and then the solvent and TFA were evaporated. The residue was dissolved in CH2C12, washed with saturated aqueous K2CO3 solution. The organic layer was dried over K2CO3 and filtered. Evaporation of solvent gave 7 as an oil (3.32 g, 76.1%). 'H-NMR (CDCI3): d 1.05 (t, 3H), 1.25-1.45 (m, 4H), 1.55-1.75 (m, 4H), 1.80 (m, 2H), 2.00 (b, 1H), 2.25 (b, 1H), 2.60-2.75 (m, 3H), 2.80 (m, 1H), 3.10 (t, 2H), 3.25 (m, 2H), 3.75 (m, 1H).
EXAMPLE 2 Synthesis of "cis" head groups.
12
11
Procedure:
In a manner similar to the preparation of 7, compound 12 was prepared. EXAMPLE 3 ATTACHMENT OF TAIL GROUPS
Tail groups were attached to the head groups according to the following procedures:
;
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 2x 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 animation:
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 2x 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: l-[l-benzyl-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H-benzimidazol-2-one
LC: 95%
MS: m/z
'H NMR (CDCI3): d 1.10 - 2.90 (m, 18H), 3.5 (s, 2H), 3.70 (m, IH), 7.25 (m, 5H).
l-[l-(naphth-2-yl-methyl)-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H-benzimidazol-2- one
MS: m/z 364.2 (M+l)
'H-NMR (CDCI3): d 1.35-2.00 (m, 11H), 2.10 (m, 2H), 2.35 (m, IH), 2.95-3.10 (m, 4H),
3.70 (s, 2H), 3.80 (m, IH), 4.40 (s, IH), 7.50 (m, 3H), 7.70 (s, IH), 7.80 (m, 3H).
l-[l-(3,3-Bis(phenyl)propyl)-4-piperidinyl]-trans-l, 3,4,5, 6,7, 8,9-octahydro-2H-benzimidazol-
2-one
MS: m/z 418.3 (M+l)
'H-NMR (CDCI3): d 1.30-2.00 (m, 15H), 2.28 (m, 4H), 2.80-3.05 (m, 3H), 3.75 (m, IH),
3.92 (t, lH), 4.40 (b, IH).
l-[l-(4-propylcyclohexyl)-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H-benzimidazol-2- one
MS: m/z 348.3 (M+l)
'H-NMR (CDCI3): d 0.85 (m 3H), 1.10-1.70 (m, 20H), 1.78 (m, 5H), 1.92 (m, IH), 2.10-2.40,
(m, 3H), 3.00 (m, 3H), 3.75 (m, IH), 4.38 (b, IH).
1 - [ 1 -(5 -methylhex-2-yl)-4-piperidinyl] -trans- 1 ,3 ,4,5 ,6,7,8,9-octahydro-2H-benzimidazol-2-one MS: m/z 322.3 (M+l)
'H-NMR (CDCI3): d 0.80-1.00 (m, 9H), 1.10-1.85 (m, 15H), 1.95 (m, IH), 2.20-2.45 (m, 3H), 2.52 (m, IH), 2.78 (m, 2H), 3.00 (m, 2H), 3.75 (m, IH), 4.40 (b, IH).
1 -[ 1 -(decahydro-2-naphthyl)-4-piperidinyl]-trans- 1 ,3,4,5,6,7,8,9-octahydro-2H-benzimidazol-2- one
MS: m/z 360.3 (M+l)
'H-NMR (CDCI3): d 0.80-1.10 (m, 2H), 1.20-2.00 (m, 25H), 2.20-2.60 (m, 4H), 2.85-3.15 (m,
4H), 3.75 (m, IH), 4.40 (b, IH). l-[l-(cyclooctyl)-4-piperidinyl]-trans-l, 3,4,5,6,7,8, 9-octahydro-2H-benzimidazol-2-one MS: m/z 334.3 (M+l)
'H-NMR (CDC13): d 1.20-1.90 (m, 24H), 1.95 (m, IH), 2.35 (m, 3H), 2.55 (m, IH), 2.80 (m, 2H), 3.00 (m, 2H), 3.75 (m, IH), 4.45 (s, IH).
l-[l -[4-(l-methylethyl)-cyclohexyl]-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one
MS: m/z 348.3 (M+l)
'H-NMR (CDCI3): d 0.85 (m, 6H), 0.90-2.00 (m, 21H), 2.05-2.40 (m, 4H), 3.00 (m, 4H), 3.75
(m, IH), 4.40 (b, IH).
I -[l-(cyclooctylmethyl)-4-piperidinyl] -trans- 1, 3,4,5, 6,7,8, 9-octahydro-2H-benzimidazol-2-one MS: m z 348.3 (M+l)
'H-NMR (CDCI3): d 1.20 (m, 2H), 1.30-1.70 (m, 19H), 1.78 (m, 4H), 1.95 (m, 3H), 2.05 (m, 2H), 2.3 (m, IH), 2.90 (m, 2H), 3.00 (m, 2H), 3.75 (m, IH), 4.40 (b, IH).
3-ethyl-l-[l-(benzyl)-4-piperidinyl]-trans-l, 3,4,5, 6,7,8,9-octahydro-2H-benzimidazol-2-one MS: m/z 342.2 (M+l)
'H-NMR (CDCI3): d 1.02 (t, 3H), 1.40 (m, 4H), 1.60-1.72 (m, 2H), 1.80 (m, 4H), 2.08 (m, 3H), 2.30 (m, IH), 2.75 (m, IH), 2.88 (m, 3H), 3.20 (m, 2H), 3.50 (s, 2H), 3.80 (m, IH), 7.25-7.35 (m, 5H).
3-ethyl- l -[l -(naphth-2-yl-methyl)-4-piperidinyl]-trans-l ,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one
MS: m/z 392.2
'H-NMR (CDCI3): d 1.05 (t, 3H), 1.30-1.90 (m, 8H), 2.00-2.15 (m, 3H), 2.30 (m, IH), 2.60 (m,
2H), 2.75 (m, IH), 2.90 (m, IH), 2.95 (m, 2H), 3.20 (m, 2H), 3.65 (s, 2H), 3.80 (m, IH), 7.48
(m, 3H), 7.70 (s, IH), 7.80 (m, 3H).
3-ethyl-l-[l-(3,3-Bis(phenyl)propyl)-4-ρiperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one
MS: m/z 446.3 (M+l) 'H-NMR (CDCI3): d 1.02 (t, 3H), 1.15 (m, IH), 1.35 (m, 4H), 1.65-1.85 (m, 5H), 1.90-2.05 (m, 3H), 2.25 (m, 4H), 2.6-2.78 (m, 2H), 2.80-2.95 (m, 3H), 3.20 (m, 2H), 3.75 (m, IH), 3.95 (m, IH), 7.15-7.30 (m, 10H).
3 -ethyl- 1-[1 -(4-propylcyclohexyl)-4-piperidinyl]-trans- 1 ,3,4, 5, 6,7,8, 9-octahydro-2H- benzimidazol-2-one
MS: m/z 376.3 (M+l)
'H-NMR (CDCI3): d 0.85 (m, 3H), 1.06 (t, 3H), 1.15-1.90 (m, 23H), 2.10 (m, IH), 2.10-2.35 (m,
4H), 2.74 (m, IH), 2.85 (m, IH), 2.99 (m, 2H), 3.23 (m, 2H), 3.75 (m, IH).
3-ethyl- 1 -[1 -(5-methylhex-2-yl)-4-piperidinyl]-trans-l ,3,4,5,6,7,8,9-octahydro-2H-benzimidazol-
2-one
MS: m/z 350.3 (M+l)
'H-NMR (CDCI3): d 0.85 (d, 6H), 0.96 (M, 3H), 1.05 (t, 3H), 1.10-1.45 (m, 8H), 1.51 (m, 2H),
1.70-1.85 (m, 5H), 2.10 (m, IH), 2.20-2.60 (m, 4H), 2.70-2.90 (m, 4H), 3.20 (m, 2H), 3.78 (m,
IH).
3-ethyl-l -[l -(decahydro-2-naphthyl)-4-piperidinyl]-trans-l ,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one
MS: m/z 388.3 (M+l)
'H-NMR (CDCI3): d 1.02 (m, 3H), 1.20-2.40 (m, 32H), 2.70-3.00 (m, 3H), 3.25 (m, 2H),*3.78
(m, IH).
3-ethyl-l-[l-(cyclooctyl)-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H-benzimidazol-2-one MS: m/z 362.3 (M+l)
'H-NMR (CDCI3): d 1.03 (m, 3H), 1.25-1.90 (m, 24H), 2.02 (m, IH), 2.30 (m, 3H), 2.60 (m, IH), 2.70-2.90 (m, 4H), 3.20 (m, 2H), 3.75 (m, IH).
3-ethyl- 1 -[ 1 -(4-propylcyclohexyl)-4-piperidinyl]-trans- 1 ,3, 4, 5, 6,7,8, 9-octahydro-2H- benzimidazol-2-one
MS: m/z 376.3 (M+l)
'H-NMR (CDCI3): d 0.85 (d, 6H), 1.05 (t, 3H), 1.25-1.90 (m, 20H), 1.99-2.35 (m, 5H), 2.73 (m,
IH), 2.85 (m, IH), 3.00 (m, 2H), 3.20 (m, 2H), 3.75 (m, IH). 3-ethyl-l-[l-(cyclooc1ylmethyl)-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H-benzimidazol-
2-one
MS: m/z 376.3 (M+l)
'H-NMR (CDCI3): d 1.05 (t, 3H), 1.10-2.10 (m, 30H), 2.27 (m, IH), 2.74 (m, IH), 2.85 (m, 3H),
3.20 (m, 2H), 3.75 (m, IH).
EXAMPLE 4
Nociceptin affinity at the ORLl receptor for preferred compounds was obtained using the following assay:
Membranes from recombinant HEK-293 cells expressing the human opioid receptorlike 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 x 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 20x 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 PRISMO, v. 3.0 and the results are set forth in table 1 below: Example 5
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 20x 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 for several compounds are set forth in table 2 below:

Claims (30)

  1. What is claimed is:
    A compound of the formula (I):
    wherein
    A is a saturated or partially saturated ring;
    R is hydrogen, C,.,0 alkyl, C3.12 cycloalkyl, C3.]2 cycloalkylC alkyl-, C,.,0 alkoxy, C3.,2 cycloalkoxy-, C,.,0 alkyl substituted with 1-3 halogen, C3.,2 cycloalkyl substituted with 1-3 halogen, C3.12 cycloalkylC alkyl- substituted with 1-3 halogen, C 0 alkoxy substituted with 1-3 halogen, C3.12 cycloalkoxy- substituted with 1-3 halogen, -COOV,, -C COOV„ -
    CH2OH, -SO2N(V,)2 , hydroxyC,.10alkyl-, hydroxyC3.,0cycloalkyl-, cyanoC,.ι0alkyl-, cyanoC3. 10cycloalkyl-, -CON(V,)2, NH2SO2CMalkyl-, NH2SOC,.4alkyl-, sulfonylaminoC,.10alkyl-, diaminoalkyl-, -sulfonylC alkyl, a 6-membered heterocyclic ring, a 6-membered heteroaromatic ring, a 6-membered heterocyclicCMalkyl-, a 6-membered heteroaromaticC,. 4alkyl-, a 6-membered aromatic ring, a 6-membered aromaticC,^ alkyl-, a 5-membered heterocyclic ring optionally substituted with an oxo or thio, a 5-membered heteroaromatic ring, a 5-membered heterocyclicCMalkyl- optionally substituted with an oxo or thio, a 5- membered heteroaromaticC,.4alkyl-, -C,.5(=O)W„ -C,.5(=NH)W„ -C,.5NHC(=O)W„ -C,. 5NHS(=O)2W„ -C,.5NHS(=O)W„ wherein W, is hydrogen, C,.,0 alkyl, C3.I2 cycloalkyl, C 0 alkoxy, C3.,2 cycloalkoxy, -CH2OH, amino, C alkylamino-, diC,.4alkylamino-, or a 5- membered heteroaromatic ring optionally substituted with 1-3 lower alkyl; wherein each V, is independently selected from H, C,.6 alkyl, C3.6 cycloalkyl, benzyl and phenyl; n is an integer from 0 to 3;
    M„ M2, M3 and M4 are each independently N, NH, CH or CH2 up to a maximum of 3 N or NH;
    D, B and C are independently hydrogen, C 0 alkyl, C3.I2 cycloalkyl, C,.,0 alkoxy, C3.,2 cycloalkoxy, -CH2OH, -NHSO2, hydroxyC,.10alkyl-, aminocarbonyl-, C,.4alkylaminocarbonyl-, diC,.4alkylaminocarbonyl-, acylamino-, acylaminoalkyl-, amide, sulfonylaminoC,.10alkyl-, or D- B can together form a C2.6 bridge, or B-C can together form a C3.7 bridge, or D-C can together form a C,.5 bridge;
    Z is selected from the group consisting of a bond, straight or branched C,.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;
    R, is selected from the group consisting of hydrogen, C,.,0 alkyl, C3.12cycloalkyl, C2. 10alkenyl, amino, CMOalkylamino-, C3.12cycloalkylamino-, -COOV,, -C COOV, , cyano, cyanoC,.,0alkyl-, cyanoC30cycloalkyl-, NH2SO2-, NH2SO2C,.4alkyl-, NH2SOC,.4alkyl-, aminocarbonyl-, C,.4alkylaminocarbonyl-, diCMalkylaminocarbonyl-, 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 (II):
    (II) wherein X, and X2 are independently selected from the group consisting of NH, O, S and CH2; and wherein said alkyl, cycloalkyl, alkenyl, C,.,0alkylamino-, C3.12cycloalkylamino-, or benzyl of R, is optionally substituted with 1 -3 substituents selected from the group consisting of halogen, hydroxy, C,.]0 alkyl, C,.,0 alkoxy, nitro, trifluoromethyl-, cyano, - COOV,, -C,.4COOV„ cyanoC Oalkyl-, -C,.5(=O)W„ -C,.5NHS(=O)2W„ -C,.5NHS(=O)W„ 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, C,.10 alkyl-, C,.10 alkoxy-, and cyano; and wherein said C3.12 cycloalkyl, C3-i2 cycloalkenyl, monocyclic, bicyclic or tricyclic aryl, heteroaryl ring, hetero-monocyclic ring, hetero-bicyclic ring system, or spiro ring system of the formula (II) is optionally substituted with 1-3 substituents selected from the group consisting of halogen, C,.,0 alkyl, C,. ,0 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, C,.,0 alkyl, C,.,0 alkoxy, and cyano;
    R2 is selected from the group consisting of hydrogen, C,.I0 alkyl, C3.,2 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.
  2. 2. A compound of claim 1, wherein R is -CH2C=ONH2, -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-.
  3. 3. A compound of claim 1, wherein ZR, is cyclohexylethyl-, cyclohexylmethyl-, cyclopentylmethyl-, dimethylcyclohexylmethyl-, phenylethyl-, pyrrolyitrifluoroethyl-, thienyltrifluoroethyl-, pyridylethyl-, cyclopentyl-, cyclohexyl-, methoxycyclohexyl-, tetrahydropyranyl-, propylpiperidinyl-, indolylmethyl-, pyrazoylpentyl-, thiazolylethyl-, phenyltrifluoroethyl-, hydroxyhexyl-, methoxyhexyl-, isopropoxybutyl-, hexyl-, or oxocanylpropyl-.
  4. 4. A compound of claim 1, wherein at least one of ZR, or R is -CH2COOV,, tetrazolylmethyl-, cyanomethyl-, NH2SO2methyl-, NH2SOmethyl-, aminocarbonylmethyl-, C,. 4alkylaminocarbonylmethyl-, or diC,.4alkylaminocarbonylmethyl-.
  5. 5. A compound of claim 1, wherein ZR, is 3,3 diphenylpropyl optionally substituted at the 3 carbon of the propyl with -COOV,, tetrazolylC0.4alkyl-, cyano-, aminocarbonyl-, C,_ 4alkylaminocarbonyl-, or diC,.4alkylaminocarbonyl-.
  6. 6. A compound of the formula (IA):
    (IA)
    wherein
    A is a saturated or partially saturated ring; 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;
    R is selected from the group consisting of hydrogen, C,.,0 alkyl, C,.10 alkoxy, and C3. ,2cycloalkyl;
    R, is selected from the group consisting of hydrogen, C,.10alkyl, C3.,2cycloalkyl, C2.,0alkenyl, amino, C,.,0alkylamino, C3.,2cycloalkylamino, benzyl, C3.12 cycloalkenyl, a monocyclic, bicyclic or tricyclic aryl or heteroaryl ring, a heteromonocyclic ring, a heterobicyclic ring system, and a spiro ring system of the formula (II):
    (II)
    wherein X, and X2 are independently selected from the group consisting of NH, O, S and CH2; wherein said alkyl, cycloalkyl, alkenyl, C,.,0alkylamino, C3.,2cycloalkylamino, or benzyl is optionally substituted with 1-3 substituents selected from the group consisting of halogen, C 0 alkyl, C 0 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,.,0 alkyl, C,.,0 alkoxy, and cyano; wherein said C3.,2 cycloalkyl, C3.,2 cycloalkenyl, monocyclic, bicyclic or tricyclic aryl, heteroaryl ring, heteromonocyclic ring, heterobicyclic ring system, and spiro ring system of the formula (II) are optionally substituted with 1-3 substituents selected from the group consisting of halogen, C,.,0 alkyl, C,.,0 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,.10 alkyl, C 0 alkoxy, and cyano;
    R2 is selected from the group consisting of hydrogen, C,.,0 alkyl, C3.12 cycloalkyl and halogen, said alkyl optionally substituted with an oxo group; or a pharmaceutically acceptable salt thereof.
  7. 7. A compound of claim 6, wherein R, is alkyl selected from the group consisting of methyl, ethyl, propyl, butyl, pentyl, and hexyl.
  8. 8. A compound of claim 6, wherein R, is cycloalkyl selected from the group consisting of cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, and norbornyl.
  9. 9. A compound of claim 6, wherein R, is tetrahydronaphthyl, decahydronaphthyl or dibenzocyclohepty 1.
  10. 10. A compound of claim 6, wherein R, is phenyl or benzyl.
  11. 11. A compound of claim 6, wherein R, is a bicyclic aromatic ring.
  12. 12. A compound of claim 11, wherein R, is indenyl, quinoline or naphthyl.
  13. 13. A compound of claim 6, wherein Z is a bond, methyl, or ethyl.
  14. 14. A compound of claim 6, wherein n is 0.
  15. 15. A compound of claim 6, wherein A is a saturated ring.
  16. 16. A compound of claim 6, wherein X, and X2 are both O.
  17. 17. A compound selected from the group consisting of: l-[l-benzyl-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H-benzimidazol-2-one; l-[l-(naphth-2-yl-methyl)-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one; l-[l-(3,3-Bis(phenyl)propyl)-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one; l-[l-(4-propylcyclohexyl)-4-piperidinyl]-trans-l, 3,4,5,6,7,8, 9-octahydro-2H- benzimidazol-2-one; l-[l-(5-methylhex-2-yl)-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one; l-[l-(decahydro-2-naphthyl)-4-piperidinyl]-trans-l, 3,4,5,6,7, 8,9-octahydro-2H- benzimidazol-2-one; l-[l-(cyclooctyl)-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H-benzimidazol-2- one; l-[l-[4-(l-methylethyl)-cyclohexyl]-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one; l-[l-(cyclooctylmethyl)-4-piperidinyl]-trans-l, 3,4,5, 6,7,8, 9-octahydro-2H- benzimidazol-2-one;
    3-ethyl-l-[l-(benzyl)-4-piperidinyl]-trans-l, 3,4,5,6,7^,8, 9-octahydro-2H-benzimidazol- 2-one;
    3-ethyl-l-[l-(naphth-2-yl-methyl)-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one;
    3-ethyl-l-[l-(3,3-Bis(phenyl)propyl)-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro- 2H-benzimidazol-2-one;
    3-ethyl-l-[l-(4-propylcyclohexyl)-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one;
    3-ethyl-l-[l-(5-methylhex-2-yl)-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one;
    3-ethyl- l-[l-(decahydro-2-naphthyl)-4-piperidinyl]-trans-l, 3,4,5,6,7,8, 9-octahydro- 2H-benzimidazol-2-one;
    3-ethyl-l-[l-(cyclooctyl)-4-piperidinyl]-trans-l, 3,4,5,6,7, 8, 9-octahydro-2H- benzimidazol-2-one;
    3-ethyl-l-[l-(4-propylcyclohexyl)-4-piperidinyl]-trans-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one;
    3-ethyl- l-[l-(cyclooctylmethyl)-4-piperidinyl]-trans-l, 3,4,5, 6,7,8,9-octahydro-2H- benzimidazol-2-one; l-[l-benzyl-4-piperidinyl]-cis-l, 3,4,5,6,7,8, 9-octahydro-2H-benzimidazol-2-one; l-[l-(naphth-2-yl-methyl)-4-piperidinyl]-cis-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one;
    1 - [ 1 -(3 ,3 -Bis(phenyl)propyl)-4-piperidinyl] -cis- 1 ,3 ,4,5 ,6,7, 8,9-octahydro-2H- benzimidazol-2-one; l-[l-(4-propylcyclohexyl)-4-piperidinyl]-cis-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2 -one ; l-[l-(5-methylhex-2-yl)-4-piperidinyl]-cis- 1,3,4,5,6,7, 8, 9-octahydro-2H-benzimidazol- 2-one; 1 -[1 -(decahydro-2-naphthyl)-4-piperidinyl]-cis-l ,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one; l-[l-(cyclooctyl)-4-piperidinyl]-cis-l,3,4,5,6,7,8,9-octahydro-2H-benzimidazol-2-one; l-[l-[4-(l-methylethyl)-cyclohexyl]-4-piperidinyl]-cis-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one; l-[l-(cyclooctylmethyl)-4-piperidinyl]-cis-l,3,4,5,6,7,8,9-octahydro-2H-benzimidazol- 2-one;
    3-ethyl- l-[l-(benzyl)-4-piperidinyl]-cis-l, 3,4,5,6,7, 8, 9-octahydro-2H-benzimidazol-2- one;
    3-ethyl-l-[l-(naphth-2-yl-methyl)-4-piperidinyl]-cis-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one;
    3 -ethyl- 1 - [ 1 -(3 ,3 -Bis(phenyl)propyl)-4-piperidinyl] -cis- 1 ,3 ,4, 5 ,6,7,8,9-octahydro-2H- benzimidazol-2-one;
    3-ethyl- l-[l-(4-propylcyclohexyl)-4-piperidinyl]-cis-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one;
    3-ethyl-l-[l-(5-methylhex-2-yl)-4-piperidinyl]-cis-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one;
    3-ethyl- 1 -[ 1 -(decahydro-2-naphthyl)-4-piperidinyl]-cis- 1 ,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one;
    3-ethyl-l-[l-(cyclooctyl)-4-piperidinyl]-cis-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one;
    3-ethyl- l-[l-(4-propylcyclohexyl)-4-piperidinyl]-cis-l, 3,4,5, 6,7, 8,9-octahydro-2H- benzimidazol-2-one;
    3-ethyl-l-[l-(cyclooctylmethyl)-4-piperidinyl]-cis-l,3,4,5,6,7,8,9-octahydro-2H- benzimidazol-2-one; and pharmaceutically acceptable salts thereof.
  18. 18. A pharmaceutical composition comprising a compound of claim 1 and at least one pharmaceutically acceptable excipient.
  19. 19. A method of treating pain comprising administering to a patient in need thereof, an effective amount of an analgesic compound according to claim 1.
  20. 20. A method of modulating a pharmacological response from the ORLl receptor comprising administering to a patient in need thereof, an effective amount of a compound according to claim 1.
  21. 21. A pharmaceutical composition comprising a compound of claim 6 and at least one pharmaceutically acceptable excipient.
  22. 22. A method of treating pain comprising administering to a patient in need thereof, an effective amount of an analgesic compound according to claim 6.
  23. 23. A method of modulating a pharmacological response from the ORLl receptor comprising administering to a patient in need thereof, an effective amount of a compound according to claim 6.
  24. 24. A compound of the formula (IA):
    (IA) wherein
    R is selected from the group consisting of hydrogen, C,.10 alkyl, C,.,0 alkoxy, and C3. 12cycloalkyl;
    R2 is selected from the group consisting of hydrogen, C,.,0 alkyl, C3.12 cycloalkyl and halogen, said alkyl optionally substituted with an oxo group; A is a saturated or partially saturated ring; n is an integer from 0 to 3; and ZR, is
    wherein
    Y, is R3-(C,-C,2)alkyl, R4-aryl, R5-heteroaryl, R6-(C3-C,2)cyclo-alkyl, R7-(C3- C7)heterocycloalkyl, -CO2(C,-C6)alkyl, CN or -C(O)NR8R9; Y2 is hydrogen or Y,; Y3 is hydrogen or (C,-C6)alkyl; or Y,, Y2 and Y3, together with the carbon to which they are attached, form one of the following structures:
    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;
    R,0 is 1 to 3 substituents independently selected from the group consisting of H, (C,- C6)alkyl, -OR8, - (C,-C6)alkyl-OR8, -NR^and -(C,-C6)alkyl-NR8R9;
    R, i is 1 to 3 substituents independently selected from the group consisting of R,0, -CF3, -OCF3, NO2 and halo, or Ru substituents on adjacent ring carbon atoms may together form a methylenedioxy or ethylenedioxy ring;
    R8and Rg are independently selected from the group consisting of hydrogen, (C,-C6) alkyl, (C3-C12)cycloalkyl, aryl and aryl(C,-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, -NR8 R,, -OR,2 and - S(O)0.2R,2;
    Rg is 1 to 3 substituents independently selected from the group consisting of H, (C,- C6)alkyl, R4-aryl, -NRgR, , -OR,2 and -SR12;
    R4 is 1 to 3 substituents independently selected from the group consisting of hydrogen, halo, (C,- C6 )alkyl, R13 -aryl, (C3 - C12)cycloalkyl, -CN, -CF3, -OR8, -(C,-C6)alkyl-OR8, - OCF3, -NRsR,, -(C, - C6)alkyl -NRgR,, -NHSO2R8, -SO2N(R,4)2, -SO2R8, -SOR8, -SR8, -NO2, -CONRgR,, -COR8, -COCF3, -OCOR8, -OCO2R8, -COOR8, -(C,-C6)alkyl- NHCOOC(CH3)3, -(C,-C6)alkyl-NHCOCF3, -(C,-C6)alkyl-NHSO2-(C,-C6)alkyl, -(C,-C6)alkyl- NHCONH-(C,-C6)-alkyl and
    -(CH2)f-N^N-R8 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, (C,-C6)alkyl, R13-aryl, (C3-C12)cycloalkyl, -CN, -CF3, -OR8, -(C,-C6)alkyl-OR8, -OCF3,- NRgRg, -(C C alkyl-NRgRg, -NHSO2R8, -SO2N(R,4)2, -NO2, -COR8, -OCOR8, -OCO2R8and -COOR8;
    R7 is H, (C,-C6)alkyl, -OR8) -(C,-C6)alkyl-OR8, -NRgR., or -(C,-C6)alkyl-NRgR9;
    R,2 is H, (C,-C6)alkyl, R4-aryl, -(C,-C6)alkyl-OR8, -(C,-C6)alkyl- SRg, or aryl (C,-C6)alkyl; R,3 is 1-3 substituents independently selected from the group consisting of H, (Cr C6)alkyl, (C,-C6)alkoxy and halo;
    R,4 is independently selected from the group consisting of H, (C,-C6)alkyl and R13- C6H4-CH2-; or a pharmaceutically acceptable salt thereof.
  25. 25. A pharmaceutical composition comprising a compound of claim 24 and at least one pharmaceutically acceptable excipient.
  26. 26. A method of treating pain comprising administering to a patient in need thereof, an effective amount of an analgesic compound according to claim 24.
  27. 27. A method of modulating a pharmacological response from the ORLl receptor comprising administering to a patient in need thereof, an effective amount of a compound according to claim 24.
  28. 28. 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 1.
  29. 29. 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 6.
  30. 30. 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 24.
AU2002338424A 2001-04-18 2002-04-18 Benzimidazolone compounds Ceased AU2002338424B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US28466501P 2001-04-18 2001-04-18
US60/284,665 2001-04-18
PCT/US2002/012375 WO2002085361A1 (en) 2001-04-18 2002-04-18 Benzimidazolone compounds

Publications (2)

Publication Number Publication Date
AU2002338424A1 true AU2002338424A1 (en) 2003-04-17
AU2002338424B2 AU2002338424B2 (en) 2005-04-21

Family

ID=23091051

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2002338424A Ceased AU2002338424B2 (en) 2001-04-18 2002-04-18 Benzimidazolone compounds

Country Status (13)

Country Link
US (2) US6872733B2 (en)
EP (3) EP1385518B1 (en)
JP (1) JP4401076B2 (en)
AT (2) ATE403431T1 (en)
AU (1) AU2002338424B2 (en)
CA (1) CA2444489C (en)
CY (2) CY1108456T1 (en)
DE (2) DE60228072D1 (en)
DK (2) DK1975164T3 (en)
ES (2) ES2312616T3 (en)
PT (2) PT1975164E (en)
SI (2) SI1385518T1 (en)
WO (1) WO2002085361A1 (en)

Families Citing this family (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003082333A1 (en) 2002-03-29 2003-10-09 Mitsubishi Pharma Corporation Remedy for sleep disturbance
US20100009928A1 (en) 2004-03-29 2010-01-14 Cheng Jin Q Compositions including triciribine and taxanes and methods of use thereof
US20100028339A1 (en) 2004-03-29 2010-02-04 Cheng Jin Q Compositions including triciribine and trastuzumab and methods of use thereof
DK2574341T3 (en) 2004-03-29 2017-06-26 Univ South Florida Effective treatment of tumors and cancer with triciribin phosphate
US20100009929A1 (en) 2004-03-29 2010-01-14 Cheng Jin Q Compositions including triciribine and bortezomib and derivatives thereof and methods of use thereof
US20100173864A1 (en) * 2004-03-29 2010-07-08 Cheng Jin Q Compositions including triciribine and one or more platinum compounds and methods of use thereof
US20110008327A1 (en) * 2004-03-29 2011-01-13 Cheng Jin Q Compositions including triciribine and epidermal growth factor receptor inhibitor compounds or salts thereof and methods of use thereof
EP1937670A1 (en) * 2005-09-30 2008-07-02 Glaxo Group Limited Benzimidazolones which have activity at m1 receptor
EA016286B1 (en) * 2005-09-30 2012-03-30 Глэксо Груп Лимитед Compounds which have activity at mreceptor and their uses in medicine
ES2391107T3 (en) * 2005-09-30 2012-11-21 Glaxo Group Limited Compounds that have activity in the M1 receptor and their uses in medicine
CN1870631B (en) * 2005-11-11 2010-04-14 华为技术有限公司 Gate control method of media gateway
TW200813018A (en) 2006-06-09 2008-03-16 Astrazeneca Ab Novel compounds
US20080051380A1 (en) 2006-08-25 2008-02-28 Auerbach Alan H Methods and compositions for treating cancer
CN101595100B (en) * 2007-01-16 2014-06-11 普渡制药公司 Heterocyclic-substituted piperidine as orl-1 ligands
GB0706188D0 (en) * 2007-03-29 2007-05-09 Glaxo Group Ltd Compounds
EP2433937B1 (en) 2007-08-31 2016-06-29 Purdue Pharma LP Substituted-quinoxaline-type-piperidine compounds and the uses thereof
US8119661B2 (en) * 2007-09-11 2012-02-21 Astrazeneca Ab Piperidine derivatives and their use as muscarinic receptor modulators
GB0718415D0 (en) * 2007-09-20 2007-10-31 Glaxo Group Ltd Compounds
PT2324013E (en) 2008-07-21 2012-12-24 Purdue Pharma Lp Substituted-quinoxaline-type bridged-piperidine compounds and the uses thereof
WO2014102594A2 (en) 2012-12-27 2014-07-03 Purdue Pharma L.P. Substituted benzimidazole-type piperidine compounds and uses thereof
WO2014102588A2 (en) 2012-12-27 2014-07-03 Purdue Pharma L.P. Indole and indoline-type piperidine compounds and uses thereof
WO2014102590A1 (en) 2012-12-27 2014-07-03 Purdue Pharma L.P. Substituted piperidin-4-amino-type 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

Family Cites Families (29)

* 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
DE2609645A1 (en) 1976-03-09 1977-09-15 Boehringer Sohn Ingelheim AMINOALKYLHETEROCYCLES
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
US4410528A (en) 1980-05-16 1983-10-18 Kyowa Hakko Kogyo Co., Ltd. Hypotensive piperidine derivatives
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
EP0755392A4 (en) 1994-04-14 1997-05-02 Merck & Co Inc Alpha1c adrenergic receptor antagonists
FR2722190B1 (en) 1994-07-05 1996-10-04 Sanofi Sa 1-BENZYL-1,3-DIHYDRO-2H-BENZIMIDAZOL-2-ONE DERIVATIVES, THEIR PREPARATION, 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
CA2224517A1 (en) 1995-06-12 1996-12-27 G.D. Searle & Co. Compositions comprising a cyclooxygenase-2 inhibitor and a 5-lipoxygenase inhibitor
WO1997010213A1 (en) 1995-09-13 1997-03-20 Kyowa Hakko Kogyo Co., Ltd. Phenylpiperidine derivatives
ATE341542T1 (en) 1996-04-19 2006-10-15 Neurosearch As 1-(4-PIPERIDYL)-BENZIMIDAZOLE WITH NEUROTROPHIC ACTIVITY
US6063796A (en) 1997-04-04 2000-05-16 Merck & Co., Inc. Somatostatin agonists
CA2291094C (en) 1997-05-30 2008-02-26 Banyu Pharmaceutical Co., Ltd. 2-oxoimidazole derivative
JPH10330377A (en) 1997-06-02 1998-12-15 Kyowa Hakko Kogyo Co Ltd Piperidine derivative
US6166037A (en) 1997-08-28 2000-12-26 Merck & Co., Inc. Pyrrolidine and piperidine modulators of chemokine receptor activity
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
ES2175925T3 (en) * 1998-01-19 2002-11-16 Pfizer COMPOUNDS 4- (2-CETO-1-BENZILIMIDAZOLINIL) PIPERIDINE AS ORL1 RECEIVER AGONISTS.
US6410561B1 (en) 1998-03-26 2002-06-25 Japan Tobacco Inc. Amide derivatives and nociceptin antagonists
EP1080091A1 (en) 1998-05-18 2001-03-07 Novo Nordisk A/S Novel 1,3,8-triazaspiro[4.5]decanones with high affinity for opioid receptor subtypes
ID29137A (en) 1998-07-27 2001-08-02 Schering Corp HIGH AFINITY LIGANS FOR ORL-1 NOSISEPTIN RECEPTORS
US6262066B1 (en) 1998-07-27 2001-07-17 Schering Corporation High affinity ligands for nociceptin receptor ORL-1
MA26659A1 (en) 1998-08-06 2004-12-20 Pfizer NOVEL BENZIMIDAZOLE DERIVATIVES, PHARMACEUTICAL COMPOSITIONS CONTAINING THEM AND PROCESS FOR THEIR PREPARATION.
SE9904652D0 (en) 1999-12-17 1999-12-17 Astra Pharma Prod Novel Compounds
CN1212839C (en) 2000-09-06 2005-08-03 奥索-麦克尼尔药品公司 Method for treating allergies using substituted pyrazoles

Similar Documents

Publication Publication Date Title
US6828440B2 (en) Spiroindene and spiroindane compounds
US7495109B2 (en) Nociceptin analogs
US6635653B2 (en) Spiropyrazole compounds
US7563809B2 (en) Nociceptin analogs
US20050192307A1 (en) Benzimidazolone compounds
AU2002256282A1 (en) Spiroindene and spiroindane compounds
AU2002311833A1 (en) Spiropyrazole compounds
AU2002307416A1 (en) Nociceptin analogs
AU2002338424A1 (en) Benzimidazolone compounds
AU2002303406A1 (en) Nociceptin analogs