US20040176443A1 - Capsaicin receptor ligands - Google Patents

Capsaicin receptor ligands Download PDF

Info

Publication number
US20040176443A1
US20040176443A1 US10/799,286 US79928604A US2004176443A1 US 20040176443 A1 US20040176443 A1 US 20040176443A1 US 79928604 A US79928604 A US 79928604A US 2004176443 A1 US2004176443 A1 US 2004176443A1
Authority
US
United States
Prior art keywords
alkyl
substituted
alkoxy
halo
occurrence
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/799,286
Inventor
Rajagopal Bakthavatchalam
Alan Hutchison
Robert DeSimone
Kevin Hodgetts
James Krause
Geoffrey White
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.)
Neurogen Corp
Original Assignee
Neurogen Corp
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 Neurogen Corp filed Critical Neurogen Corp
Priority to US10/799,286 priority Critical patent/US20040176443A1/en
Publication of US20040176443A1 publication Critical patent/US20040176443A1/en
Priority to US12/057,808 priority patent/US20090082362A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/72Nitrogen atoms
    • C07D213/74Amino or imino radicals substituted by hydrocarbon or substituted hydrocarbon radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/08Bronchodilators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/72Nitrogen atoms
    • C07D213/75Amino or imino radicals, acylated by carboxylic or carbonic acids, or by sulfur or nitrogen analogues thereof, e.g. carbamates
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/22Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the nitrogen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/02Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings
    • C07D241/10Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members
    • C07D241/14Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D241/20Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • This invention relates compounds that bind with high selectivity and high affinity to Vanilloid Receptors, especially Type I Vanilloid Receptors, also known as capsaicin receptors or VR1 Receptors.
  • the invention provides capsaicin receptor, preferably human VR1 receptor, antagonists that are not capsaicin analogs (e.g., they do not contain a phenyl ring with two oxygen atoms bound to two adjacent ring carbons), are free of agonist activity, and exhibit an unprecedented level of affinity for the VR1 receptor.
  • the invention provides aryl piperazines and related compounds that act as VR1 receptor ligands.
  • this invention relates to such VR1 receptor ligands, high affinity antagonists and pharmaceutical compositions comprising such compounds and to the use of such compounds in treatment of diseases and other health-related conditions. Additionally this invention relates to the use of aryl piperazines and related compounds as tool for the analysis of VR1 receptors and as probes for the quantitative measurement and localization of VR1 receptors in cell and tissue samples.
  • the sensation of pain can be triggered by any number of physical or chemical stimuli.
  • the peripheral terminals of a group of specialized small diameter sensory neurons, termed “nociceptors” mediate this response to a potentially harmful stimulus.
  • Capsaicin responses in isolated sensory neurons show dose-dependence and are also evoked by structural analogues of capsaicin that share a common vanilloid moiety.
  • the term vanilloid receptor (VR) was coined to describe the neuronal membrane recognition site for capsaicin and such related irritant compounds. It was postulated that the VR is a nonselective cation channel with a preference for calcium.
  • resiniferatoxin (RTX) a natural product of certain Euphorbia plants, was recognized as an ultrapotent VR agonist. Specific binding of 3H RTX provided the first unequivocal proof for the existence of a vanilloid receptor.
  • the capsaicin response is competitively inhibited (and thereby antagonized) by another capsaicin analog, capsazepine and is also inhibited by the non-selective cation channel blocker ruthenium red.
  • These antagonists bind to VR with no more than moderate affinity (i.e., with Ki values of no lower than 140 uM).
  • VR1 rat capsaicin receptor
  • the capsaicin receptor's channel opens in response to elevated temperatures (higher than about 45° C.).
  • Capsaicin and related compounds, as well as protons are stimuli that lower the threshold channel opening, so that in the presence of any of these stimuli the capsaicin receptor can be opened even at room temperature.
  • Opening of the capsaicin receptor channel is followed by the release of inflammatory peptides from neurons expressing the receptor and other nearby neurons, increasing the pain response.
  • the capsaicin receptor undergoes a rapid desensitization, possibly via phosphorylation of intracellular sites of the receptor.
  • Capsaicin and related VR1 agonist vanilloid compounds have enjoyed long-pharmaceutical use as topical anaesthetics. While such compounds initially cause a strong burning sensation, receptor desensitization provides pain relief.
  • the vanilloid compounds capsaicin and Resiniferatoxin act as potent and specific agonists of the capsaicin receptor.
  • Capsazepine which contains a phenyl ring with two oxygen atoms bound to two adjacent ring carbons and is therefore a capsaicin analog
  • Iodo-RTX is a capsaicin analog that has recently been reported to act as a high affinity antagonist.
  • the inorganic dye, Ruthenium red also antagonizes capsaicin responses of the receptor, albeit as a non-selective cation channel blocker.
  • This invention relates to VR1 receptor ligands, particularly VR1 receptor antagonists, and methods of using VR1 receptor antagonists for the treatment of neuropathic pain, peripheral-nerve-mediated pain, and pain, inflammatory and broncho-constriction symptoms resulting from exposure to capsacin-receptor-activating stimuli such as capsaicin and tear gas.
  • the invention provides novel chemical compounds that act as capsaicin receptor modulatory agents, some of which exhibit antagonist potency greater than that of any previously described VR1 receptor antagonist.
  • Compounds that act as capsaicin receptor antagonists and bind to capsaicin (preferably human VR1) receptors with K i values of less than 100 uM, as measured by a capsacin receptor binding assay, such as the assay given by Example 10, or that inhibit capsaicin activity in an assay for determination of capsaicin receptor antagonist effects (Example 11) with EC 50 values of less than or equal to 100 uM, are referred to herein as potent capsaicin receptor antagonists; such compounds that bind or antagonize with K 1 or EC 50 values of less than or equal to 10 uM are referred to herein as highly potent capsaicin receptor antagonists.
  • the invention provides methods of using the potent capsaicin receptor antagonist compounds of the invention for the treatment of symptoms resulting from exposure to painful capsaicin receptor activating stimuli.
  • the invention provides methods of treating subjects who have been exposed to capsaicin or have been burnt by heat, light, tear gas, or acid exposure, the methods comprising administering to such subjects an effective amount of a potent capsaicin receptor antagonist, preferably a highly potent. (high potency) capsaicin receptor antagonist, so that the subject's symptoms of pain or sensitivity are reduced.
  • Preferred compounds of the invention provide pain relief without loss of consciousness, and preferably without sedation, in such subjects that is equal to or grater than the degree of pain relief that can be provided to such subjects by morphine without loss of consciousness. Highly preferred compounds provide such pain relief while causing only transient (i.e., lasting for no more than one half the time that pain relief lasts) or no sedation (see Example 16 for sedation assay).
  • Subjects or patients referred to herein may be humans or non-human mammals including domestic companion animals (pets) and livestock animals, as discussed more fully below.
  • the invention provides methods of treating of neuropathic pain based on the unexpected finding that capsaicin receptor antagonists can alleviate such pain.
  • This invention also provides aryl piperazines and related compounds that bind with high affinity and high selectivity to capsaicin receptors, including human capsaicin receptors, also known as VR1 receptors.
  • the invention provides novel compounds of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, or Formula VII, Formula VIII, Formula IX and Formulae A-F shown below (the “compounds of the invention,” hereinafter Formulae I-IX and Formulae A-F), and pharmaceutical compositions comprising compounds of Formulae I-IX and Formulae A-F.
  • the invention further comprises methods of treating patients suffering from certain diseases or conditions, especially those involving pain or urinary incontinence, with an amount of a compound Formulae I-IX and Formulae A-F that is effective to improve the symptoms (e.g., reduce pain or reduce the frequency of urinary incontinence) of the disease or condition being treated.
  • this invention relates to the use of the compounds of the invention as reagents, standards, and probes for measurement, characterization and localization of capsaicin receptors, particularly VR1 receptors (e.g., in cells or tissues.
  • A is chosen from O, S, NR A , CR B R B ′, NR A CR B R B ′, CR B R B ′NR A , —CR A ⁇ CR B —, and C 3 H 4 ; where R A , R B , and R B ′ are independently selected, at each occurrence from hydrogen or alkyl;
  • Z is oxygen or sulfur
  • R 1 and R 2 independently represent hydrogen or lower alkyl
  • R 1 and R 2 are taken together to form a 5 to 8 membered nitrogen containing ring of the formula:
  • n 1, 2, or 3;
  • R 3 and R 4 are independently selected at each occurrence from the group consisting of hydrogen; halogen; hydroxy; amino; cyano; nitro; —COOH; —CHO, optionally substituted alkyl; optionally substituted alkenyl; optionally substituted alkynyl; optionally substituted alkoxy; optionally substituted mono or dialkylamino; optionally substituted alkylthio; optionally substituted alkyl ketone; optionally substituted alkylester; optionally substituted alkylsulfinyl; optionally substituted alkylsulfonyl; optionally substituted mono- or di-alkylcarboxamide; optionally substituted —S(O) n NHalkyl; optionally substituted —S(O) n N(alkyl)(alkyl); optionally substituted —NHC( ⁇ O)alkyl; optionally substituted —NC( ⁇ O)(alkyl)(alkyl); optionally substituted —NHC( ⁇
  • R 3 and R 4 not attached to the same carbon may be joined to form an optionally substituted aryl ring; a saturated or partially unsaturated carbocyclic ring of from 5 to 8 members, which carbocyclic ring is optionally substituted; or a saturated, partially unsaturated, or aromatic heterocyclic ring of from 5 to 8 members, which heterocyclic ring is optionally substituted and contains 1, 2, or 3 heteroatoms selected from N, O, and S; and
  • Ar 1 and Ar 2 are the same or different and independently represent optionally substituted cycloalkyl; an optionally substituted heterocycloalkyl ring of from 5 to 8 atoms, which heterocyloalkyl ring contains 1, 2, or 3 heteroatoms selected from N, O, and S; optionally substituted aryl having from 1 to 3 rings; or optionally substituted heteroaryl, said heteroaryl having from 1 to 3 rings, 5 to 8 ring members in each ring and, in at least one of said rings, from 1 to about 3 heteroatoms per ring selected from the group consisting of N, O, and S, and
  • n is independently chosen at each occurrence from 0, 1, and 2.
  • R 1 and R 2 are joined to form a 5 to 7-membered heterocycloalkyl ring, e.g. R 1 and R 2 may be joined to form a piperazine ring.
  • This 5 to 7-membered heterocycloalkyl ring is preferably unsubstituted or substituted at one or two positions with a C 1-6 alkyl group, such as methyl or ethyl.
  • the variable “Z” is preferably oxygen and the variable “A” is generally NH, CH ⁇ CH, or CH 2 NH.
  • Ar 1 and Ar 2 are preferably optionally substituted phenyl or optionally substituted pyridyl; optionally substituted 2-pyridyl is preferred for Ar 2 .
  • Substitutuents that may occur on Ar 1 and Ar 2 include, but are not limited to, butyl, isopropyl, trifluoromethyl, nitro, methyl, and halogen. Substitution at the 4 position of Ar 1 (when Ar 1 is phenyl or pyridyl) and substitution at the 3 position of Ar 2 (when Ar 2 is phenyl or pyridyl) are described in specific embodiments of the invention.
  • the invention is particularly directed to compounds of Formula I, in which R 1 and R 2 independently represent hydrogen or lower alkyl, e.g., C 1-6 alkyl. Such compound will be referred to as compounds of Formula IA.
  • Preferred compounds and pharmaceutically acceptable salts of Formula IA are those wherein:
  • R 3 and R 4 are independently chosen at each occurrence from the group consisting of hydrogen, halogen, cyano, nitro, haloalkyl, haloalkoxy, hydroxy, amino, alkyl substituted with 0-2 R 6 , alkenyl substituted with 0-2 R 6 ; alkynyl substituted with 0-2 R 6 ; alkoxy substituted with 0-2 R 6 , —NH(alkyl) substituted with 0-2 R 6 , —N(alkyl)(alkyl) where each alkyl is independently substituted with 0-2 R 6 , —XR 7 , and Y;
  • R 3 and R 4 not attached to the same carbon may be joined to form an aryl ring substituted with 0-3 R 6 , a saturated or partially unsaturated carbocyclic ring of from 5 to 8 members, which carbocyclic ring is substituted with 0-2 R 6 , or a saturated, partially unsaturated, or aromatic heterocyclic ring of from 5 to 8 members, which heterocyclic ring is substituted with 0-2 R 6 and contains 1, 2, or 3 heteroatoms selected from N, O, and S; and
  • Ar 1 and Ar 2 may be the same or different and are selected from the group consisting of cyclohexyl, cyclopentyl, piperidinyl, piperazinyl, phenyl, pyrrolyl, furanyl, thienyl, pyrazolyl, imidazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, triazolyl, tetrazolyl, pyridyl, pyrimidyl, pyrazinyl, benzimidazolyl, naphthyl, indolyl, isoindolyl, benzofuranyl, isobenzofuranyl, benzo[b]thiophenyl, benz[d]isoxazolyl, quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, and quinoxalinyl
  • Ar 1 and Ar 2 may be the same or different and represent a bicyclic oxygen-containing group of the formula:
  • R 5 optionally mono-, di-, or trisubstituted with R 5 , where L represents point of attachment and may be at any point on the benzene ring, and the oxygen-containing ring of the bicyclic oxygen-containing group consists of from 5 to 8 ring atoms, contains 1 or 2 oxygen atoms and remaining ring atoms are carbon;
  • R 5 is independently selected at each occurrence from the group consisting of halogen, cyano, nitro, haloalkyl, haloalkoxy, hydroxy, amino, alkyl substituted with 0-2 R 6 , alkenyl substituted with 0-2 R 6 , alkynyl substituted with 0-2 R 6 , alkoxy substituted with 0-2 R 6 , —NH(alkyl) substituted with 0-2 R 6 , —N(alkyl)(alkyl) where each alkyl is independently substituted with 0-2 R 6 , —XR 7 , and Y;
  • R 6 is independently selected at each occurrence from the group consisting of halogen, hydroxy, cyano, alkyl, alkoxy, —NH(alkyl), —N(alkyl)(alkyl), —S(O) n (alkyl), haloalkyl, haloalkoxy, CO(alkyl), CONH(alkyl), CON(alkyl 1 )(alkyl 2 ) where alkyl 1 and alkyl 2 may be joined to form a heterocycloalkyl ring of from 5 to 8 ring atoms and containing 1, 2, or 3 heteroatoms selected from N, O, and S, —XR 7 , and Y;
  • X is independently selected at each occurrence from the group consisting of —CH 2 —, —CHR 8 —, —O—, —S(O) n —, —NH—, —NR 8 —, —C( ⁇ O)—, —C( ⁇ O)O—, —C( ⁇ O)NH—, —C( ⁇ O)NR 8 —, —S(O) n NH—, —S(O) n NR 8 —, NHC( ⁇ O)—, —NR 8 C( ⁇ O)—, —NHS(O) n —, and —NR 8 S(O) n —;
  • R 7 and R 8 are independently selected at each occurrence from hydrogen
  • Y and Y′ are independently selected at each occurrence from 3- to 8-membered carbocyclic or heterocyclic groups which are saturated, unsaturated, or aromatic, which may be further substituted with one or more substituents independently selected from halogen, oxo, hydroxy, amino, nitro, cyano, alkyl, alkoxy, haloalkyl, haloalkoxy, mono- or dialkylamino, and alkylthio;
  • said 3- to 8-memberered heterocyclic groups contain one or more heteroatom(s) independently selected from N, O, and S;
  • n is independently chosen at each occurrence from 0, 1, and 2.
  • A, Z, R 3 , R 4 are as defined for Formula I or for Formula IB;
  • Ar 1 and Ar 2 are as defined for Formula I or for formula IB;
  • x is 1 or 3.
  • Preferred compounds and salts of Formula II are those in which
  • R A , R B , and R B ′ are independently selected at each occurrence from hydrogen or C 1-6 alkyl.
  • G, Q, T, and W are the same or different and represent N, CH, or CR 5 , where R 5 is as defined for Formula IB;
  • R A , R B , and R B ′ are independently selected at each occurrence from hydrogen or C 1-6 alkyl;
  • Z is oxygen or sulfur
  • R 3 and R 4 are as defined for Formula I or for Formula IB;
  • x is 1 or 3.
  • Z is S or O (preferably O);
  • R 3 , and R 4 is as defined for Formula I or Formula IB;
  • Ar 1 and Ar 2 may be the same or different and are selected from the group consisting of cyclohexyl, cyclopentyl, piperidinyl, piperazinyl, pyrrolyl, furanyl, thienyl, pyrazolyl, imidazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, triazolyl, tetrazolyl, pyridyl, pyrimidyl, pyrazinyl, benzimidazolyl, naphthyl, indolyl, isoindolyl, benzofuranyl, isobenzofuranyl, benzo[b]thiophenyl, benz[d]isoxazolyl, quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, and quinoxalinyl; wherein Ar 1
  • Ar 1 and Ar 2 may be the same or different and represent a bicyclic oxygen-containing group as described for Formula IB,
  • R 5 is independently selected at each occurrence from the group consisting of cyano, nitro, haloalkyl, haloalkoxy, hydroxy, amino, alkyl substituted with 0-2 R 6 , alkenyl substituted with 0-2 R 6 , alkynyl substituted with 0-2 R 6 , alkoxy substituted with 0-2 R 6 , —NH(alkyl) substituted with 0-2 R 6 , —N(alkyl)(alkyl) where each alkyl is independently substituted with 0-2 R 6 , —XR 7 , and Y;
  • R 9 is independently selected at each occurrence from the group consisting of cyano, nitro, haloalkoxy, hydroxy, amino, alkyl substituted with 0-2 R 6 , alkenyl substituted with 0-2 R 6 , alkynyl substituted with 0-2 R 6 , alkoxy substituted with 0-2 R 6 , —NH(alkyl) substituted with 0-2 R 6 , —N(alkyl)(alkyl) where each alkyl is independently substituted with 0-2 R 6 , —XR 7 , and Y; and
  • R 6 , R 7 , R 8 , X, Y and Y′ are as defined for Formula IB.
  • Another embodiment of the invention is directed to compounds of Formula V
  • G, Q, T, and W are the same or different and are selected from the group consisting of N, CH, and CR 5 , wherein T or W or both is N;
  • A, R 3 , and R 4 are as defined for Formula I or for Formula IB (preferably A is —CH ⁇ CH—, —CH 2 NH, NH, and R 3 and R 4 are hydrogen or C 1-6 alkyl);
  • Z is oxygen or sulfur (preferably oxygen);
  • R 5 represents 1 to 3 substituents and is independently selected at each occurrence from the group consisting of cyano, hydroxy, amino, C 3-6 alkyl substituted with 0-2 R 6 , C 2-6 alkenyl substituted with 0-2 R 6 , C 2-6 alkynyl substituted with 0-2 R 6 , C 3-6 alkoxy substituted with 0-2 R 6 , —NH(C 1-6 alkyl) substituted with 0-2 R 6 , —N(C 1-6 alkyl)(C 1-6 alkyl) where each alkyl is independently substituted with 0-2 R 6 , —XR 7 , and Y;
  • R 9 represents 0 to 3 substituents and is independently selected at each occurrence from the group consisting of halogen, cyano, nitro, halo(C 1-6 )alkyl, halo(C 1-6 )alkoxy, hydroxy, amino, C 1-6 alkyl substituted with 0-2 R 6 , C 2-6 alkenyl substituted with 0-2 R 6 , C 2-6 alkynyl substituted with 0-2 R 6 , C 1-6 alkoxy substituted with 0-2 R 6 , —NH(C 1-6 alkyl) substituted with 0-2 R 6 , —N(C 1-6 alkyl)(C 1-6 alkyl) where each C 1-6 alkyl is independently substituted with 0-2 R 6 , —XR 7 , and Y;
  • R 6 , R 7 , R 8 , X, Y, and Y′ are as defined for Formula IB.
  • the invention is particularly directed to compounds and salts of Formula V wherein G and Q are selected from the group consisting of CH and CR 5 .
  • the invention is also directed to compounds and salts of Formula V wherein G, Q, and W are independently selected at each occurrence from the group consisting of CH and CR 5 ; and T is N.
  • R 6 substituents are halogen, hydroxy, C 1-4 alkyl, C 1-4 alkoxy, —NH(C 1-4 alkyl), and —N(C 1-4 alkyl)(C 1-4 alkyl).
  • Still another embodiment of the invention is directed of compounds of Formula VI
  • A is selected from the group consisting of NH, —CH ⁇ CH—, and CH 2 NH;
  • R 4 is independently chosen from hydrogen and C 1-4 alkyl
  • R 5 represents 0 to 2 substituents and is independently chosen at each occurrence from the group consisting of halogen, cyano, nitro, halo(C 1-6 )alkyl, halo(C 1-6 )alkoxy, hydroxy, amino, C 1-6 alkyl substituted with 0-2 R 6 , C 2-6 alkenyl substituted with 0-2 R 6 , C 2-6 alkynyl substituted with 0-2 R 6 , C 1-6 alkoxy substituted with 0-2 R 6 , —NH(C 1-6 alkyl) substituted with 0-2 R 6 , —N(C 1-6 alkyl)(C 1-6 alkyl) where each C 1-6 alkyl is independently substituted with 0-2 R 6 ;
  • R 9 represents 0 to 2 substituents and is independently chosen at each occurrence from the group consisting of halogen, cyano, nitro, halo(C 1-6 )alkyl, halo(C 1-6 )alkoxy, hydroxy, amino, C 1-6 alkyl substituted with 0-2 R 6 , C 2-6 alkenyl substituted with 0-2 R 6 , C 2-6 alkynyl substituted with 0-2 R 6 , C 1-6 alkoxy substituted with 0-2 R 6 , —NH(C 1-6 alkyl) substituted with 0-2 R 6 , and —N(C 1-6 alkyl)(C 1-6 alkyl) where each C 1-6 alkyl is independently substituted with 0-2 R 6 ;
  • R 5A is independently selected from the group consisting of halogen, cyano, nitro, halo(C 1-6 )alkyl, halo(C 1-6 )alkoxy, hydroxy, amino, C 1-6 alkyl, C 1-6 alkoxy, —NH(C 1-6 alkyl), and —N(C 1-6 alkyl)(C 1-6 alkyl)
  • R 9B is independently selected from the group consisting of halogen, nitro, halo(C 1-6 )alkoxy, hydroxy, amino, C 1-6 alkyl, C 1-6 alkoxy, —NH(C 1-6 alkyl), and —N(C 1-6 alkyl)(C 1-6 alkyl); and
  • R 6 is independently selected at each occurrence the group consisting of halogen, hydroxy, C 1-4 alkyl, C 1-4 alkoxy, —NH(C 1-4 alkyl), and —N(C 1-4 alkyl)(C 1-4 alkyl)
  • R 3 , and R 4 are as defined for Formula I or for Formula IB;
  • R 5 is independently selected at each occurrence from the group consisting of cyano, nitro, haloalkyl, haloalkoxy, C 1-6 alkyl substituted with 0-2 R 6 , C 2-6 alkenyl substituted with 0-2 R 6 , C 2-6 alkynyl substituted with 0-2 R 6 , C 1-6 alkoxy substituted with 0-2 R 6 , —NH(C 1-6 alkyl) substituted with 0-2 R 6 , —N(C 1-6 alkyl)(C 1-6 alkyl) where each alkyl is independently substituted with 0-2 R 6 , —XR 7 , and Y;
  • R 9 represents 0-3 substituents and is independently selected at each occurrence from the group consisting of bromo, haloalkyl, haloalkoxy, hydroxy, C 2-6 alkyl substituted with 0-2 R 6 , C 2-6 alkenyl substituted with 0-2 R 6 , C 2-6 alkynyl substituted with 0-2 R 6 , C 2-6 alkoxy substituted with 0-2 R 6 , —NH(C 2-6 alkyl) substituted with 0-2 R 6 , —N(C 2-6 alkyl)(C 2-6 alkyl) where each C 2-6 alkyl is independently substituted with 0-2 R 6 , —XR 7 , and Y;
  • R 6 , R 7 , R 8 , X, Y, and Y′ are as defined for Formula IB.
  • Preferred compounds and salts of Formula VII include those wherein A is selected from NH, —CH ⁇ CH—, and CH 2 NH; and R 6 is independently selected at each occurrence from the group consisting of halogen, hydroxy, C 1-4 alkyl, C 1-4 alkoxy, —NH(C 1-4 alkyl), and —N(C 1-4 alkyl)(C 1-4 alkyl).
  • the invention includes compounds of Formula VIII
  • A is selected from the group consisting of NH, —CH ⁇ CH—, and CH 2 NH (NH is preferred);
  • R 4 is independently selected at each occurrence from hydrogen and C 1-4 alkyl
  • R 5 represents 0 to 2 substituents independently selected at each occurrence from the group consisting of halogen, cyano, nitro, halo(C 1-6 )alkyl, halo(C 1-6 )alkoxy, hydroxy, amino, C 1-6 alkyl substituted with 0-2 R 6 , C 2-6 alkenyl substituted with 0-2 R 6 , C 2-6 alkynyl substituted with 0-2 R 6 , C 1-6 alkoxy substituted with 0-2 R 6 , —NH(C 1-6 alkyl) substituted with 0-2 R 6 , and —N(C 1-6 alkyl)(C 1-6 alkyl) where each C 1-6 alkyl is independently substituted with 0-2 R 6 ;
  • R 9 represents 0 to 2 substituents and is independently selected at each occurrence from the group consisting of halogen, cyano, nitro, halo(C 1-6 )alkyl, halo(C 1-6 )alkoxy, hydroxy, amino, C 1-6 alkyl substituted with 0-2 R 6 , C 2-6 alkenyl substituted with 0-2 R 6 , C 2-6 alkynyl substituted with 0-2 R 6 , C 1-6 alkoxy substituted with 0-2 R 6 , —NH(C 1-6 alkyl) substituted with 0-2 R 6 , and —N(C 1-6 alkyl)(C 1-6 alkyl) where each C 1-6 alkyl is independently substituted with 0-2 R 6 ;
  • R 5A is independently selected from the group consisting of halogen, cyano, nitro, trifluoromethyl, trifluoromethoxy, hydroxy, amino, C 1-6 alkyl, C 1-6 alkoxy, —NH(C 1-6 alkyl), and —N(C 1-6 alkyl)(C 1 -C 6 alkyl);
  • R 9B is independently selected from the group consisting of trifluoromethoxy, hydroxy, C 2-6 alkyl, C 2-6 alkoxy, —NH(C 2-6 alkyl), and —N(C 2-6 alkyl)(C 2-6 alkyl); and
  • R 6 is independently selected at each occurrence from the group consisting of halogen, hydroxy, C 1-4 alkyl, C 1-4 alkoxy, —NH(C 1-4 alkyl), and —N(C 1-4 alkyl)(C 1-4 alkyl)
  • Preferred compound of Formula VIII are those wherein one of R 4 is hydrogen and the other is methyl.
  • a particularly preferred embodiment of the invention includes compounds of Formula IX
  • R 3 , and R 4 are as defined for Formula I or for Formula IB;
  • R 5 is selected from the group consisting of bromo, fluoro, iodo, halo(C 1-6 )alkyl, halo(C 3-6 )alkoxy, C 3-6 alkyl substituted with 0-3 R 6 , C 2-6 alkenyl substituted with 0-3 R 6 , C 2-6 alkynyl substituted with 0-3 R 6 , C 3-6 alkoxy substituted with 0-2 R 6 , (C 3-8 cycloalkyl)C 1-4 alkyl, —NH(C 1-6 alkyl) substituted with 0-2 R 6 , —N(C 1-6 alkyl)(C 1-6 alkyl) where each C 1-6 alkyl is substituted with 0-2 R 6 , Y, —(C ⁇ O)Y, —(CH 2 )Y, and —(CH(CN))Y;
  • R 9 is selected from the group consisting of halogen, cyano, —N(SO 2 C 1-6 alkyl)(SO 2 C 1-6 alkyl), —SO 2 NH 2 , halo (C 1-6 ) alkyl, halo(C 1-6 )alkoxy, C 1-6 alkyl substituted with 0-2 R 6 , C 2-6 alkenyl substituted with 0-2 R 6 , C 2-6 alkynyl substituted with 0-2 R 6 , C 1-6 alkoxy substituted with 0-2 R 6 , —NH(C 1-6 alkyl) substituted with 0-2 R 6 , —N(C 1-6 alkyl)(C 1-6 alkyl) where each C 1-6 alkyl is substituted with 0-2 R 6 ;
  • R 5B and R 9B each represent from 0 to 2 substituents and are independently selected at each occurrence from the group consisting of halogen, cyano, nitro, halo(C 1-6 )alkyl, halo(C 1-6 )alkoxy, hydroxy, amino, C 1-6 alkyl substituted with 0-2 R 6 , (C 3-8 cycloalkyl)C 1-4 alkyl substituted with 0-2 R 6 , C 2-6 alkenyl substituted with 0-2 R 6 , C 2-6 alkynyl substituted with 0-2 R 6 , C 1-6 alkoxy substituted with 0-2 R 6 , —NH(C 1-6 alkyl) substituted with 0-2 R 6 , and —N(C 1-6 alkyl)(C 1-6 alkyl) where each C 1-6 alkyl is independently substituted with 0-2 R 6 , and Y; and any two
  • R 5 and R 5B bound to adjacent atoms may be joined to form a C 3-8 cycloalkyl group or a heterocycloalkyl group, each of which is optionally substituted by from 1 to 5 substituents independently chosen from cyano, halogen, hydroxy, C 1-4 alkyl, C 1-4 alkoxy, —NH(C 1-4 alkyl), —N(C 1-4 alkyl)(C 1-4 alkyl), halo(C 1-4 )alkyl, and halo(C 1-4 )alkoxy, wherein the heterocycloalkyl group consists of from 4 to 8 atoms and contains 1, 2, or 3 heteroatoms selected from N, O, and S; and
  • R 6 , R 7 , R 8 , X, Y, and Y′ are as defined for Formula IB.
  • Preferred compounds and salts of Formula IX are those wherein A is O or NR A , wherein R A is hydrogen or methyl.
  • A is O or NR A , wherein R A is hydrogen or methyl
  • R 3 and R 4 are independently chosen at each occurrence from the group consisting of hydrogen, halogen, cyano, nitro, halo(C 1-6 )alkyl, halo(C 1-6 )alkoxy, hydroxy, amino, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 alkoxy, —NH(C 1-6 alkyl), and —N(C 1-6 alkyl)(C 1-6 alkyl).
  • A is O or NR A , wherein R A is hydrogen or methyl;
  • R 3 is hydrogen
  • R 4 is independently chosen at each occurrence from the group consisting of hydrogen, halogen, cyano, nitro, halo(C 1-6 )alkyl, halo(C 1-6 )alkoxy, hydroxy, amino, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 alkoxy, —NH(C 1-6 alkyl), and —N(C 1-6 alkyl)(C 1-6 alkyl).
  • Still more preferred compounds and salts of Formula IX are those wherein
  • A is O, NR A , wherein R A is hydrogen or methyl;
  • R 3 is hydrogen
  • R 4 is independently chosen at each occurrence from hydrogen and C 1-6 alkyl.
  • IX is the group wherein
  • A is NR A , wherein R A is hydrogen or methyl
  • R 3 is hydrogen
  • R 4 is independently chosen at each occurrence from hydrogen, halo(C 1-3 )alkyl, and C 1-6 alkyl, but more preferably R 4 is chosen from hydrogen and C 1-4 alkyl.
  • R 5 , R 5B , R 9 , and R 9B are as defined for Formula IX;
  • R 4 is independently chosen at each occurrence from hydrogen and C 1-4 alkyl.
  • R 5B and R 9B are independently chosen from hydrogen, halogen, cyano, nitro, halo(C 1-2 )alkyl, halo(C 1-2 )alkoxy, amino, C 1-4 alkyl, and C 1-2 alkoxy; and
  • R 10 is independently chosen at each occurrence from hydrogen, halogen, and C 1-4 alkyl.
  • Preferred compounds and salts of Formula IX-B are those wherein R 9 is selected from the group consisting of halogen, cyano, —N(SO 2 CH 3 ) 2 , —SO 2 NH 2 , halo(C 1-3 )alkyl, C 1-3 alkoxy, —NH(C 1-3 alkyl), and —N(C 1-3 alkyl)(C 1-3 alkyl).
  • R 5B and R 9B are independently chosen from hydrogen, halogen, cyano, nitro, halo(C 1-2 )alkyl, halo(C 1-2 )alkoxy, amino,
  • R 5B represents 0 or 1 substituents chosen from halogen, cyano, nitro, halo(C 1-2 )alkyl, halo(C 1-2 )alkoxy, amine, C 1-4 alkyl, and C 1-2 alkoxy; and
  • R 9B represents 0 or 1 substituents chosen from halogen, cyano, nitro, halo(C 1-2 )alkyl, and C 1-2 alkyl, and C 1-2 alkoxy.
  • the invention is further directed to compounds and salts of Formula IX-A and IX-B, wherein:
  • R 9 is selected from the group consisting of halogen, cyano, —N(SO 2 CH 3 ) 2 , —SO 2 NH 2 , halo(C 1-3 )alkyl, C 1-3 alkoxy, —NH(C 1-3 alkyl), and —N(C 1-3 alkyl)(C 1-3 alkyl);
  • R 5B represents 0 or 1 substituents chosen from halogen, cyano, nitro, halo(C 1-2 )alkyl, halo(C 1-2 )alkoxy, amino, C 1-4 alkyl, and C 1-2 alkoxy; and
  • R 9B represents 0 or 1 substituents chosen from halogen, cyano, nitro, halo(C 1-2 )alkyl, and C 1-2 alkyl, and C 1-2 alkoxy.
  • R 5 is selected from the group consisting of bromo, fluoro, iodo, halo(C 1-6 )alkyl, halo(C 3-6 )alkoxy, C 3-6 alkyl substituted with 0-3 R 6 , C 2-6 alkenyl substituted with 0-3 R 6 , Y. —(C ⁇ O)Y, —(CH 2 )Y, and —(CH(CN))Y;
  • R 9 is selected from the group consisting of halogen, cyano, —N(SO 2 CH 3 ) 2 , —SO 2 NH 2 , halo(C 1-2 )alkyl, C 1-3 alkoxy, —NH(C 1-6 alkyl), and —N(C 1-6 alkyl)(C 1-6 alkyl);
  • R 5B represents 0 or 1 substituents chosen from halogen, cyano, nitro, halo(C 1-2 )alkyl, halo(C 1-2 )alkoxy, amino, C 1-4 alkyl, and C 1-2 alkoxy; and
  • R 9B represents 0 or 1 substituents chosen from halogen, cyano, nitro, halo(C 1-2 )alkyl, and C 1-2 alkyl, and C 1-2 alkoxy.
  • R 6 for compounds and salts of this class are cyano, halogen, hydroxy, C 1-4 alkyl, C 1-4 alkoxy, —NH(C 1-4 alkyl), and —N(C 1-4 alkyl)(C 1-4 alkyl) and Y;
  • Y is independently selected at each occurrence from C 3-8 cycloalkyl, piperidinyl, piperazinyl, tetrahydropyranyl, dihydropyranyl, morpholinyl, thiomorpholinyl, phenyl, pyridyl, pyrazinyl, pyrimidinyl, thiazolyl, thienyl, and imidazolyl, each of which may be further substituted with one or more substituents independently selected from halogen, oxo, hydroxy, amino, nitro, cyano, C 1-4 alkyl, C 1-4 alkoxy, halo(C 1-4 )alkyl, halo(C 1-4 )alkoxy, mono- or di(C 1-4 )alkylamino, and C 1-4 alkylthio.
  • R 9 and R 9B for compounds of Formula IX-A are:
  • R 9 is cyano, trifluoromethyl, chloro, or iodo
  • R 9B is hydrogen
  • R 5 for compounds of Formula IX-A are isopropyl, t-butyl, 2-butyl, trifluoromethyl, cyclopentyl, cyclohexyl, and heptafluoropropyl.
  • the invention is particularly directed to compounds and pharmaceutically acceptable salts of Formula IB, Formula II, Formula III, Formula IV, Formula V, Formula VII and Formula IX in which:
  • R A , R B , and R B ′ are independently selected at each occurrence from hydrogen and C 1-6 alkyl; for the variables R 3 , R 4 , and R 5 haloalkyl is halo(C 1-6 )alkyl, i.e.
  • haloalkyl group having from 1 to 6 carbon atoms and from 1 to maximum allowable number of halogen substituents on those carbon atoms, haloalkoxy is halo(C 1-6 )alkoxy; alkyl is C 1-6 alkyl, alkenyl is C 2-6 alkenyl; alkynyl is C 2-6 alkynyl; alkoxy is C 1-6 alkoxy, —NH(alkyl) is —NH(C 1-6 alkyl), and —N(alkyl)(alkyl) is —N(C 1-6 alkyl)(C 1-6 alkyl),
  • R 6 , R 7 , R 8 , Y and Y′ alkyl is C 1-4 alkyl, alkoxy (or —O(alkyl) is C 1-4 alkoxy (or —O(C 1-4 alkyl)), —NHalkyl (or monoalkylamino) is —NH(C 1-4 alkyl)(or mono(C 1-4 alkyl)amino), —N(alkyl)(alkyl) (also dialkylamino) is —N(C 1-4 alkyl)(C 1-4 alkyl) (also di(C 1-4 alkyl) amino), —S(O) n alkyl is —S(O) n (C 1-4 alkyl), haloalkyl is halo(C 1-4 )alkyl, haloalkoxy is halo(C 1-4 )alkoxy, —CO(alkyl) is —CO(C 1-4 alkyl),
  • Preferred compounds and salts of Formula V, Formula VI, Formula VII and Formula IX are those wherein R 3 and R 4 are independently selected at each occurrence from the group consisting of hydrogen and C 1-6 alkyl. More preferred compounds and salts of Formula V are those wherein R 3 is hydrogen and the R 4 substituents present on the 3 and 5 positions of the piperazine ring are hydrogen and the R 4 substituents on the 2 and 6 position of the piperazine ring are independently hydrogen or C 1-4 alkyl.
  • R 3 is hydrogen and the R 4 substituents present on the 3 and 5 positions of the piperazine ring are hydrogen and the R 4 substituents on the 2 and 6 position of the piperazine ring are independently hydrogen or C 1-4 alkyl.
  • the invention particularly includes compounds Formula A-1, Formula B-1, Formula C-1, Formula D-1, Formula E-1, and Formula F-1
  • R 5 and R 9 are independently selected from the group consisting of halogen, cyano, nitro, halo(C 1-6 )alkyl, halo(C 1-6 )alkoxy, hydroxy, amino, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 alkoxy, —NH(C 1-6 alkyl, —N(C 1-6 alkyl)(C 1-6 alkyl), and C 3-8 cycloalkyl; and
  • R 5B and R 9B each represent up to 2 substituents independently selected at each occurrence from hydrogen, halogen, cyano, nitro, halo(C 1-2 )alkyl, halo(C 1-2 )alkoxy, hydroxy, amino, C 1-3 alkyl, C 1-3 alkoxy, —NH(C 1-3 alkyl), and —N(C 1-6 alkyl)(C 1-6 alkyl).
  • R 5 is C 3-6 alkyl; C 3-6 alkoxy; halo(C 1-3 )alkyl, halo(C 1-3 )alkoxy, or C 3-8 cycloalkyl;
  • R 9 is chloro or trifluomethyl
  • R 5B and R 9B are hydrogen.
  • diaryl piperazines and related compounds that bind with high affinity to capsaicin receptors, including human capsaicin receptors.
  • Compounds that bind with high affinity for the capsaicin receptor include compounds exhibit K i values of less than 10 uM, and preferably exhibit K i values of less than 1 uM, more preferably exhibit K i values of less than 100 nM, and most preferably exhibit K i values of less than 10 nM at the capsaicin receptors.
  • This invention also includes diaryl piperazines that bind with high selectivity to capsaicin receptor.
  • Compounds that exhibit high selectivity for the capsaicin receptor exhibit at least 20-fold, and preferably at least 100-fold greater affinity for the capsaicin receptor than for other cell surface receptors (e.g., NPY Y5 receptors, NPY Y1 receptors, GABA A receptors, MCH receptors, Bradykinin receptors, C5a receptors, androgen receptors, and the like).
  • NPY Y5 receptors e.g., NPY Y5 receptors, NPY Y1 receptors, GABA A receptors, MCH receptors, Bradykinin receptors, C5a receptors, androgen receptors, and the like.
  • the invention further comprises methods of treating patients in need of such treatment with an amount of a compound of the invention sufficient to alter the symptoms of a disorder responsive to capsaicin receptor modulation.
  • treatement encompases both disease modifying treatment and symptomatic treatment.
  • the diseases and/or disorders that can also be treated using compounds and compositions according to the invention include:
  • Chronic and acute pain conditions including toothache, postherpetic neuralgia, diabetic neuropathy, postmastectomy pain syndrome, stump pain (and phantom limb pain), reflex sympathetic dystrophy, trigeminal neuralgia, oral neuropathic pain, osteoarthritis, rheumatoid arthritis, fibromyalgia, Guillain-Barre syndrome, meralgia paresthetica, “burning-mouth” syndrome, and pain due to bilateral peripheral neuropathy.
  • Preferred pain conditions for treatment in accordance with the invention are neuropathic pain conditions, including causalgia (reflex sympathetic dystrophy—RSD, secondary to injury of a peripheral nerve; this type of pain is generally considered to be non-responsive or only partially responsive to conventional opioid analgesic regimens), neuritis—including, e.g., sciatic neuritis, peripheral neuritis, polyneuritis, optic neuritis, postfebrile neuritis, migrating neuritis, segmental neuritis, Gombault's neuritis, and neuronitis, and neuralgias—including those mentioned above and, e.g., cervicobrachial neuralgia, cranial neuralgia, geniculate neuralgia, glossopharyngial neuralgia, migranous neuralgia, idiopathic neuralgia, intercostals neuralgia, mammary neuralgia, mandibular joint neuralgia, Morton's neuralgia,
  • Additional pain conditions that can be treated in accordance with the invention include headache—particularly those involving peripheral nerve activity including, e.g., sinus, cluster (i.e., migranous neuralgia, supra) and some tension and migraine headache conditions—, labor pains, Charcot's pains, gas pains, menstrual pain, root pain, homotopic pain and heterotopic pain—including cancer associated pain, pain (and inflammation) associated with venom exposure, e.g., due to snake bite, spider bite, or insect sting, and traumatic, e.g., post-surgical pain and burn pain.
  • headache particularly those involving peripheral nerve activity including, e.g., sinus, cluster (i.e., migranous neuralgia, supra) and some tension and migraine headache conditions—, labor pains, Charcot's pains, gas pains, menstrual pain, root pain, homotopic pain and heterotopic pain—including cancer associated pain, pain (and inflammation) associated with venom exposure, e.g., due to snake bite, spider bite, or insect
  • a preferred condition that can be treated in accordance with the invention is pain (as well as broncho-constriction and inflammation) due to exposure (e.g., via ingestion, inhalation, or eye contact) of mucous membranes to capsacin and related irritants such as tear gas, hot peppers, or pepper spray.
  • Itching conditions including psoriatic pruritis, itch due to hemodyalisis, aguagenic pruritus, and itching associated with vulvar vestibulitis, contact dermatitis, insect bites and skin allergies.
  • Urinary incontenience including detrusor hyperflexia of spinal origin and bladder hypersentivity.
  • the invention also provides pharmaceutical compositions comprising compounds of the invention, including packaged pharmaceutical compositions for treating disorders responsive to capsaicin receptor modulation.
  • the packaged pharmaceutical compositions include a container holding a therapeutically effective amount of at least one capsaicin receptor modulator as described supra and instructions (e.g., labeling) indicating the contained capsaicin receptor ligand is to be used for treating a disorder responsive to capsaicin receptor modulation in the patient.
  • the present invention also pertains to methods of inhibiting the binding of vanilloid (capsaicin analog) compounds, such as capsaicin, olvanil and RTX, to capsaicin receptors, which methods involve contacting a compound of the invention with cells expressing capsaicin receptors, wherein the compound is present at a concentration sufficient to inhibit vanilloid binding to capsaicin receptors in vitro.
  • vanilloid capsaicin analog
  • the methods of the invention include inhibiting the binding of vanilloid compounds to capsaicin receptors in vivo, e.g., in a patient given an amount of a compound of Formulae I-IX and Formulae A-F that results in and in vivo concentration in a body fluid sufficient to inhibit the binding of capsaicin compounds to capsaicin receptors in vitro.
  • such methods are useful in treating the effects of tear gas, hot pepper or pepper spray exposure.
  • the amount of a compound that would be sufficient to inhibit the binding of a vanilloid compound to the capsaicin receptor may be readily determined via a capsaicin receptor binding assay, such as the assay described in Example 10 or by an assay of capsaicin receptor antagonism e.g, as in Example 11.
  • the capsaicin receptors used to determine in vitro binding may be obtained from a variety of sources, for example from preparations of mammalian dorsal root ganglion (DRG) or from cells expressing cloned rat or human capsaicin receptors.
  • DRG mammalian dorsal root ganglion
  • the present invention also pertains to methods for altering the signal-transducing activity, particularly the calcium ion conductance, mediated by capsaicin receptors, said method comprising exposing cells expressing such receptors to a solution comprising a compound of the, wherein the compound is present in the solution at a concentration sufficient to specifically alter the calcium conductance activity in response to capsaicin or RTX in vitro in cells expressing capsaicin receptors, preferred cells for this purpose are those that express high levels of capsaicin receptors (i.e., equal to or greater than the number of capsaicin receptors per cell found in rat DRG cells).
  • This method includes altering the signal-transducing activity of capsaicin receptors in vivo.
  • Such alterations are reductions of calcium flux.
  • the amount of a compound that would be sufficient to alter the signal-transducing activity of capsaicin receptors may be determined in vitro via a capsaicin receptor signal transduction assay, such as the calcium mobilization (conductance, flux) assay described in Example 11.
  • the amount of a compound that would be sufficient to alter the calcium conductance activity in response to capsaicin or RTX of capsaicin receptors may also be determined via an assay of capsaicin receptor mediated calcium conductance, such as an assay wherein the binding of capsaicin to a cell surface capsaicin receptor effects changes in the fluorescence of a calcium sensitive dye or in the expression of a calcium sensitive reporter gene.
  • the invention further provides:
  • a method of reducing the calcium conductance of a capsaicin receptor which method comprises:
  • a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound or salt of the invention; and a package comprising the pharmaceutical composition in a container and further comprising indicia comprising instructions for using the composition to either alleviate pain; or to treat a patient suffering from urinary incontinence or to alleviate symptoms of exposure to capsaicin or tear gas.
  • a compound or salt of the invention wherein, in an in vitro assay of capsaicin receptor antagonism, the compound or salt exhibits capsaicin receptor antagonist activity, but in an in vitro assay of capsaicin receptor agonism the compound does not exhibit detectable agonist activity.
  • a compound or salt of the invention wherein a dose of the compound or salt sufficient to provide analgesia in an animal model for determining pain relief does not produce sedation in an animal model assay of sedation.
  • a method of treating a mammal suffering from at least one symptom selected from the group consisting of symptoms of exposure to capsaicin, symptoms of burns or irritation due to exposure to heat, symptoms of burns or irritation due to exposure to light, symptoms of burns or irritation due to exposure to tear gas, and symptoms of burns or irritation due to exposure to acid comprising administering to the mammal a therapeutic dose of a compound that:
  • a) is a high potency capsaicin receptor antagonist in an in vitro assay of capsaicin receptor antagonism
  • the therapeutic dose contains an amount of the compound that is effective to reduce severity of at least one of the at least one symptom and preferably wherein the compound is a compound of the invention.
  • a method of treating a mammal suffering from neuropathic pain comprising administering to the mammal a therapeutic dose of a compound that is a capsaicin receptor antagonist, and in certain embodiments, wherein the compound is a compound of the invention.
  • a) is a high potency capsaicin receptor antagonist in an in vitro assay of capsaicin receptor antagonism
  • the therapeutic dose contains an amount of the compound that is effective to reduce the peripheral-nerve-mediated pain, and preferably wherein the pain is neuropathic pain and the compound is a compound of the invention, and preferably wherein the pain is associated with a condition selected from the group consisting of postmastectomy pain syndrome, stump pain, phantom limb pain, oral neuropathic pain, Charcot's pain, toothache, postherpetic neuralgia, diabetic neuropathy, reflex sympathetic dystrophy, trigeminal neuralgia, osteoarthritis, rheumatoid arthritis, fibromyalgia, Guillain-Barre syndrome, meralgia paresthetica, burning-mouth syndrome, bilateral peripheral neuropathy, causalgia, sciatic neuritis, peripheral neuritis, polyneuritis, optic neuritis, postfebrile neuritis, migrating neuritis, segmental neuritis, Gombault's neuritis, neuronitis, cervicobrach
  • a compound of the invention wherein the compound is not addictive.
  • capsaicin receptor ligands provided by this invention and labeled derivatives thereof are also useful as standards and reagents for determining the ability of a compound to bind to the capsaicin receptor and to act as an agonist, antagonist, inverse agonist, mixed agonist/antagonist or the like.
  • More particularly compounds of the invention may be used for demonstrating the presence of VR1 receptors or other capsaicin receptors in cell or tissue samples. This may be done by preparing a plurality of matched cell or tissue samples, at least one of which is prepared as an experimental sample and at least one of which is prepared as a control sample.
  • the experimental sample is prepared by contacting (under conditions that permit binding of capsaicin or RTX to capsaicin receptors within cell and tissue samples) at least one of the matched cell or tissue samples that has not previously been contacted with any compound or salt of the invention with an experimental solution comprising the detectably-labeled preparation of the selected compound or salt at a first measured molar concentration.
  • control sample is prepared by in the same manner as the experimental sample and is incubated in a solution that contains the same ingredients as the experimental solution but that also contains an unlabelled preparation of the same compound or salt of the invention at a molar concentration that is greater than the first measured molar concentration.
  • the experimental and control samples are then washed (using the same wash conditions) to remove unbound detectably-labeled compound.
  • the amount of detectably-labeled compound remaining bound to each sample is then measured and the amount of detectably-labeled compound in the experimental and control samples is compared.
  • a comparison that indicates the detection of a greater amount of detectable label in the at least one washed experimental sample than is detected in any of the at least one washed control samples demonstrates the presence of capsaicin receptors in that experimental sample.
  • the detectably-labeled compound used in this procedure may be labeled with any detectable label, such as a radioactive label, a biological tag such as biotin (which can be detected by binding to detectably-labeled avidin), an enzyme (e.g., alkaline phosphatase, beta galactosidase, or a like enzyme that can be detected its activity in a calorimetric, luminescent, or like assay) or a directly or indirectly luminescent label.
  • tissue sections are used in this procedure and the detectably-labeled compound is radiolabeled
  • the bound, labeled compound may be detected autoradiographically to generate an autoradiogram.
  • the amount of detectable label in an experimental or control sample may be measured by viewing the autoradiograms and comparing the exposure density of matched regions of the autoradiograms.
  • Labeled derivatives the capsaicin receptor ligands provided by this invention are also useful as radiotracers for positron emission tomography (PET) imaging or for single photon emission computerized tomography (SPECT) to characterize and localize capsaicin receptors in vivo.
  • PET positron emission tomography
  • SPECT single photon emission computerized tomography
  • the compounds herein described may have one or more asymmetric centers or planes.
  • Compounds of the present invention containing an asymmetrically substituted atom may be isolated in optically active or racemic forms. It is well known in the art how to prepare optically active forms, such as by resolution of racemic forms (racemates), by asymmetric synthesis, or by synthesis from optically active starting materials. Resolution of the racemates can be accomplished, for example, by conventional methods such as crystallization in the presence of a resolving agent, or chromatography, using, for example a chiral HPLC column.
  • substituted means that any one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valence is not exceeded, and that the substitution results in a stable compound.
  • the present invention is intended to include all isotopes of atoms occurring in the present compounds. Isotopes include those atoms having the same atomic number but different mass numbers, By way of general example and without limitation, isotopes of hydrogen include tritium and deuterium. Isotopes of carbon include 11 C, 13 C, and 14 C.
  • any variable occurs more than one time in any constituent or formula for a compound, its definition at each occurrence is independent of its definition at every other occurrence.
  • a group is shown to be substituted with 0-2 R*, then said group may optionally be substituted with up to two R* groups and R* at each occurrence is selected independently from the definition of R*.
  • combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
  • those substituents may be substituted by other than hydrogen at one or more available positions, typically 1 to 3 or 4 positions, by one or more groups, such as, halogen such as fluoro, chloro, bromo and iodo; cyano; hydroxyl; nitro; azido; alkanoyl such as a C 1-6 alkanoyl group such as acyl and the like; carboxamido; alkyl groups including those groups having 1 to about 12 carbon atoms, or 1, 2, 3, 4, 5, or 6 carbon atoms; alkenyl and alkynyl groups including groups having one or more unsaturated linkages and from 2 to about 12 carbon, or 2, 3, 4, 5 or 6 carbon atoms; alkoxy groups having those having one or more oxygen linkages and from 1 to about 12 carbon atoms, or 1, 2, 3, 4, 5 or 6
  • an Ar group being a substituted or unsubstituted biphenyl moiety
  • arylalkyl having 1 to 3 separate or fused rings and from 6 to about 18 carbon ring atoms, with benzyl being a preferred group
  • arylalkoxy having 1 to 3 separate or fused rings and from 6 to about 18 carbon ring atoms, with O-benzyl being a preferred group
  • a heteroaromatic or heteroalicyclic group having 1 to 3 separate or fused rings with 3 to about 8 members per ring and one or more N, O or S atoms, e.g.
  • alkyl is intended to include both branched, straight-chain, and cyclic alkyl groups, having the specified number of carbon atoms that may contain one or more double or triple bonds.
  • “Lower alkyl” denotes an alkyl group having from 1 to about 6 carbon atoms. Examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl, t-butyl, n-pentyl, and s-pentyl.
  • Preferred alkyl groups are C 1 -C 6 alkyl groups. Especially preferred alkyl groups are methyl, ethyl, propyl, butyl, 3-pentyl.
  • alkoxy As used herein, “alkoxy”, “C 1 -C 6 alkoxy”, or “lower alkoxy” in the present invention is meant an alkyl group attached through an oxygen bridge such as, for example, methoxy, ethoxy, propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, pentoxy, 2-pentyl, isopentoxy, neopentoxy, hexoxy, 2-hexoxy, 3-hexoxy, and 3-methylpentoxy.
  • “Lower alkoxy” denotes an alkyl group having from 1 to about 6 carbon atoms.
  • haloalkyl include, but are not limited to, trifluoromethyl, trichloromethyl, pentafluoroethyl, heptafluoropropyl, and pentachloroethyl.
  • carrier or “carbocyclic ring” is intended to mean any stable 3- to 7-membered monocyclic or bicyclic or 7- to 13-membered bicyclic or tricyclic moeity, any of which may be saturated, partially unsaturated, or aromatic.
  • carbocycles include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, adamantyl, cyclooctyl, [3.3.0]bicyclooctane, [4.3.0]bicyclononane, [4.4.0]bicyclodecane, [2.2.2]bicyclooctane, fluorenyl, phenyl, naphthyl, indanyl, adamantyl, and tetrahydronaphthyl.
  • carboxyaryl indicates aromatic groups containing only carbon. Such aromatic groups may be further substituted.
  • heterocyclic ring is intended to mean a stable 5- to 7-membered monocyclic or bicyclic or 7- to 10-membered bicyclic heterocyclic ring which is saturated, partially unsaturated, or unsaturated (aromatic), and which consists of carbon atoms and from 1 to 4 heteroatoms independently selected from the group consisting of N, O and S and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring.
  • the nitrogen and sulfur heteroatoms may optionally be oxidized.
  • the heterocyclic ring may be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure.
  • heterocyclic rings described herein may be substituted on carbon or on a nitrogen atom if the resulting compound is stable.
  • a nitrogen atom in the heterocycle may optionally be quaternized. It is preferred that when the total number of S and O atoms in the heterocycle exceeds 1, then these heteroatoms are not adjacent to one another. It is preferred that the total number of S and O atoms in the heterocycle is not more than 1.
  • heteroaryl is intended to mean a stable 5- to 7-membered monocyclic or bicyclic or 7- to 10-membered bicyclic heterocyclic aromatic ring which consists of carbon atoms and from 1 to 4 heteroatoms independently selected from the group consisting of N, O and S.
  • heterocycloalkyl is intended to mean a stable 5- to 7-membered monocyclic or bicyclic or 7- to 10-membered bicyclic heterocyclic saturated ring which consists of carbon atoms and from 1 to 4 heteroatoms independently selected from the group consisting of N, O and S.
  • heterocycles include, but are not limited to, acridinyl, azocinyl, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, NH-carbazolyl, carbolinyl, chromanyl, chromenyl, cinnolinyl, decahydroquinolinyl, 2H,6H-1,5,2-dithiazinyl, dihydrofuro[2,3-b]tetrahydrofuran, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, 1H-indazolyl, indolenyl, indolinyl, in
  • Preferred heterocycles include, but are not limited to, pyrrolyl, furanyl, thienyl, pyrazolyl, imidazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, triazolyl, tetrazolyl, pyridyl, pyrimidyl, pyrazinyl, benzimidazolyl, indolyl, isoindolyl, benzofuranyl, isobenzofuranyl, benzo[b]thiophenyl, benz[d]isoxazolyl, quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, or quinoxalinyl. Also included are fused ring and spiro compounds containing, for example, the above heterocycles.
  • bicyclic oxygen-containing group is meant to encompass a particular type of heteroaryl group of the formula:
  • L indicates the point of attachment of the group to the structure of Formulae I-IX and Formulae A-F.
  • the heterocyclic oxygen-containing ring has a total of from 5 to 7 members, and is saturated or unsaturated. Either ring of the bicyclic oxygen-containing group may be further substituted.
  • bicyclic oxygen-containing groups include any or all of the following structures:
  • prodrugs are intended to include any compounds that become compounds of Formulae I-IX and Formulae A-F when administered to a mammalian subject, e.g., upon metabolic processing of the prodrug.
  • prodrugs include, but are not limited to, acetate, formate and benzoate and like derivatives of functional groups (such as alcohol or amine groups) in the compounds of Formulae I-IX and Formulae A-F.
  • solvents that may be used to prepare solvates of the compounds of the invention, such as water, ethanol, mineral oil, vegetable oil, and dimethylsulfoxide.
  • the compounds of general Formulae I-IX and general Formulae A-F may be administered orally, topically, parenterally, e.g., by inhalation or spray or rectally in dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles. Oral administration in the form of a pill, capsule, elixir, syrup, lozenge, troche, or the like is particularly preferred.
  • parenteral as used herein includes subcutaneous injections, intradermal, intravascular (e.g., intravenous), intramuscular, spinal, intrathecal injection or like injection or infusion techniques.
  • a pharmaceutical formulation comprising a compound of general Formulae I-IX and general Formulae A-F and a pharmaceutically acceptable carrier.
  • One or more compounds of general Formulae I-IX and general Formulae A-F may be present in association with one or more non-toxic pharmaceutically acceptable carriers and/or diluents and/or adjuvants and if desired other active ingredients.
  • the pharmaceutical compositions containing compounds of general Formulae I-IX and general Formulae A-F may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs.
  • compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations.
  • Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients that are suitable for the manufacture of tablets.
  • excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc.
  • the tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monosterate or glyceryl distearate may be employed.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • water or an oil medium for example peanut oil, liquid paraffin or olive oil.
  • Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydropropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monoo
  • the aqueous suspensions may also contain one or more preservatives, for example ethyl, or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose or saccharin.
  • preservatives for example ethyl, or n-propyl p-hydroxybenzoate
  • coloring agents for example ethyl, or n-propyl p-hydroxybenzoate
  • flavoring agents for example ethyl, or n-propyl p-hydroxybenzoate
  • sweetening agents such as sucrose or saccharin.
  • Oily suspensions may be formulated by suspending the active ingredients in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol.
  • Sweetening agents such as those set forth above, and flavoring agents may be added to provide palatable oral preparations. These compositions may be preserved by the addition of an antioxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • a dispersing or wetting agent e.g., sodium EDTA
  • suspending agent e.g., sodium EDTA
  • preservatives e.g., sodium EDTA, sodium bicarbonate, sodium bicarbonate
  • compositions of the invention may also be in the form of oil-in-water emulsions.
  • the oily phase may be a vegetable oil, for example olive oil or arachis oil, or a mineral oil, for example liquid paraffin or mixtures of these.
  • Suitable emulsifying agents may be naturally-occurring gums, for example gum acacia or gum tragacanth, naturally-occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitil, anhydrides, for example sorbitan monoleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monoleate.
  • the emulsions may also contain sweetening and flavoring agents.
  • Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative and flavoring and coloring agents.
  • the pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be sterile injectable solution or suspension in a non-toxic parentally acceptable diluent or solvent, for example as a solution in 1,3-butanediol.
  • Suitable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • the compounds of general Formulae I-IX and general Formulae A-F may also be administered in the form of suppositories, e.g., for rectal or vaginal administration of the drug.
  • suppositories e.g., for rectal or vaginal administration of the drug.
  • These compositions can be prepared by mixing the drug with a suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • Such materials are cocoa butter and polyethylene glycols.
  • Compounds of general Formulae I-IX and general Formulae A-F may be administered parenterally in a sterile medium.
  • the drug depending on the vehicle and concentration used, can either be suspended or dissolved in the vehicle.
  • adjuvants such as local anesthetics, preservatives and buffering agents can be dissolved in the vehicle where desirable.
  • Typical subjects to which compounds of the invention may be administered will be mammals, particularly primates, especially humans.
  • mammals particularly primates, especially humans.
  • livestock such as cattle, sheep, goats, cows, swine and the like; poultry such as chickens, ducks, geese, turkeys, and the like; and other domesticated animals particularly pets (companion animals) such as dogs and cats.
  • rodents e.g. mice, rats, hamsters
  • rabbits, primates, and swine such as inbred pigs and the like.
  • the composition may also be added to the animal feed or drinking water. It will be convenient to formulate these animal feed and drinking water compositions so that the animal takes in an appropriate quantity of the composition along with its diet. It will also be convenient to present the composition as a premix for addition to the feed or drinking water.
  • dosage levels of the order of from about 0.01 mg to about 140 mg per kilogram of body weight per day are useful in the treatment of pain, urinary incontinence, or other of the above-indicated conditions (about 0.05 mg to about 7 g per human patient per day).
  • Preferred systemic doses for preferred high potency compounds of Formulae I-IX and Formulae A-F range from about 0.01 mg to about 50 mg per kilogram of body weight per subject per day, with oral doses generally being about 5-20 fold higher than intravenous doses.
  • the most highly preferred compounds of the invention are orally active (e.g., provide a reduction of pain or a reduction of frequency of urinary incontinence) at doses ranging from 0.05 to 40 mg per kilogram of body weight per subject per day.
  • Dosage unit forms will generally contain between from about 1 mg to about 500 mg of an active ingredient.
  • Frequency of dosage may also vary depending on the compound used and the particular disease treated. However, for treatment of most disorders, a dosage regimen (frequency of administration) of 4 times daily or less is preferred. For the treatment of chronic pain or urinary incontinence a dosage regimen of 2 times daily is more preferred and a frequency of administration of once a day is particularly preferred. For the treatment of acute pain a single dose that rapidly reaches effective concentrations is desirable.
  • Preferred compounds of the invention will have certain desirable pharmacological properties.
  • such properties include, but are not limited to high oral bioavailability, such that the preferred oral dosages and dosage forms discussed above can provide therapeutically effective levels of the compound in vivo, low serum protein binding and low first pass hepatic metabolism.
  • low toxicity, and desirable in vitro and in vivo half-lifes are desired. While penetration of the blood brain barrier for compounds used to treat CNS disorders is necessary, low brain levels of compounds used to treat peripheral disorders (such as urinary incontinence, or chronic or acute pain that does not originate from the CNS) are often preferred.
  • Laboratory assays may be used to predict these desirable pharmacological properties. The discussion that follows is supplemented by the detailed protocols of Example 16, infra. Assays used to predict bioavailability include transport across human intestinal cell monolayers, including. Caco-2 cell monolayers. Toxicity to cultured hepatocyctes may be used to predict compound toxicity, with non-toxic compounds being preferred. Penetration of the blood brain barrier of a compound in humans may be predicted from the brain levels of the compound in laboratory animals given the compound, e.g., intravenously.
  • Percentage of serum protein binding may be predicted from albumin binding assays. Examples of such assays are described in a review by Oravcová, et al. (Journal of Chromatography B (1996) volume 677, pages 1-27). Preferred compounds exhibit reversible serum protein binding. Preferably this binding is less than 99%, more preferably less than 95%, even more preferably less than 90%, and most preferably less than 80%.
  • Frequency of administration is generally inversely proportional to the in vivo half-life of a compound.
  • In vivo half-lives of compounds may be predicted from the results of assays, e.g., in vitro assays of microsomal half-life as described by Kuhnz and Gieschen ( Drug Metabolism and Disposition, 1998, volume 26, pages 1120-1127).
  • Preferred half-lives are those allowing for a preferred frequency of administration.
  • the compounds of the present invention can be prepared in a number of ways well known to those skilled in the art of organic synthesis.
  • compounds of the present invention can be synthesized using the methods described below, together with synthetic methods known in the art of synthetic organic chemistry, or variations thereon as appreciated by those skilled in the art. Preferred methods include but are not limited to those methods described below.
  • Compounds of the present invention i.e. urea or thiourea derivatives (VI) can be synthesized by following the steps outlined in Scheme 1.
  • Intermediate III can be obtained by treating I with II in the presence of a base (eg: K 2 CO 3 , Cs 2 CO 3 , NR 1 R 2 R 3 , NaOR, KOR) in an inert solvent such as N,N-dialkylformamide, N,N-dialkylacetamide, dialkylethers, cyclic ethers, DMSO, N-methyl-2-pyrrolidinone at temperatures ranging from ⁇ 78° C. to 200° C.
  • a base eg: K 2 CO 3 , Cs 2 CO 3 , NR 1 R 2 R 3 , NaOR, KOR
  • an inert solvent such as N,N-dialkylformamide, N,N-dialkylacetamide, dialkylethers, cyclic ethers, DMSO, N-methyl-2-pyrrolidinone at temperatures ranging from ⁇ 78° C. to 200° C.
  • Isocyanates or isothiocyanates of V can be obtained by treating compound of IV with phosgene, thiophosgene, carbonyldiimidazole in an inert solvent such as benzene, toluene at temperatures ranging from ⁇ 78° C. to 200° C.
  • the compound of present invention VI can be obtained by treating intermediates III with V in an organic solvent at temperatures ⁇ 78° C. to 200° C.
  • compound of VI can be prepared by treatment of intermediate VII with III in the presence of base such as triethylamine in an inert solvent such as chloroform at temperatures ranging from 78° C. to 200° C.
  • Carbamates or thiocarbamates (X) of the present invention can be synthesized by following the steps outlined in Scheme 2.
  • Compound of product X can be obtained by treatment with phenols (IX) with compound VIII in the presence of a base in an inert solvent at temperatures ranging from ⁇ 78° C. to 200° C.
  • Part B Synthesis of (4-sec-Butyl-phenyl)-carbamic acid phenyl ester:
  • NMR (CDCl 3 ): ⁇ 1.45-1.47 (d, 3H, J 1.7 Hz), 2.97-3.01 (t, 1H), 3.06-3.10 (m, 1H), 3.47-3.50 (m, 1H), 3.75-3.85 (m, 2H), 3.92-3.95 (m, 1H), 4.37-4.38 (m, 1H), 6.59 (bs, 1H), 6.88-6.91 (dd, 1H), 7.52-7.56 (m, 4H), 7.61-7.63 (dd, 1H), 8.19-8.21 (dd, 1H).
  • Part A Synthesis of (R)-4-(3-Chloro-pyridin-2-yl)-2-methylpiperazine-1-carbonyl chloride:
  • Part A Synthesis of 3-Methyl-1-(3-trifluoromethyl-pyridin-2-yl)-piperazine
  • Part B Synthesis of 4-(1-Trifluoromethyl-vinyl)-phenylamine
  • Part C Synthesis of 1-Isocyanato-4-(1-trifluoromethyl-vinyl)-benzene
  • Part D Synthesis of 2-Methyl-4-(3-trifluoromethyl-pyridin-2-yl)-piperazine-1-carboxylic acid [4-(1-trifluoromethyl-vinyl)-phenyl]-amide
  • Part A Synthesis of 4-(2,2,2-Trifluoro-1-methyl-ethyl)-phenylamine
  • Part B Synthesis of 1-Isocyanato-4-(2,2,2-trifluoro-1-methylethyl)-benzene
  • Part A Synthesis of 1-Isocyanato-4-(1,2,2,2-tetrafluoro-1-trifluoromethyl-ethyl)-benzene
  • the following assay is a standard assay of capsaicin receptor binding that may be used to determine the binding affinity of compounds for the capsaicin (VR1) receptor.
  • RTX Resiniferatoxin
  • a cDNA encoding the full length human capsaicin receptor (SEQ ID NO:1 or SEQ ID NO:2) is subcloned in the appropriate orientation for expression into an expression vector such as pcDNA3.1 (Invitrogen, Carlsbad, Calif.) or pUHG102-3 (Clontech, Palo Alto, Calif.) for recombinant expression in mammalian cells.
  • HEK293 Human embryonic kidney (HEK293) cells are transfected with a pcDNA3.1 expression consrtuct encoding the full length human capsaicin receptor (i.e. containing either the nucleotide sequence of SEQ ID NO:1 and SEQ ID NO:2) using standard methods. These transfected cells are selected for two weeks in media containing G418 (400 ug/ml) and then maintained as a pool of stably transfected cells.
  • G418 400 ug/ml
  • pUHG102 VR1 expression plasmids are transfected into Chinese Hamster Ovary (CHO) cells containing the pTet Off Regulator plasmid (Clontech). In these cells, expression of the pUHG plasmid is repressed in the presence of tetracycline but is induced upon removal of the antibiotic. Stable clones are isolated in culture medium containing puromycin (10 ug/ml) and maintained in medium supplemented with tetracycline (1 ug/ml). Cells utilized for assays are grown in culture medium without antibiotic for 48-72 hours prior to use.
  • cells are seeded in T175 cell culture flasks in media without antibiotics and grown to approximately 90% confluency. The flasks are then washed with PBS and harvested in PBS containing 5 mM EDTA. The cells are pelleted by gentle centrifugation and stored at ⁇ 80° C. until assayed.
  • Previously frozen cells are disrupted with the aid of a tissue homogenizer in ice-cold HEPES homogenization buffer (5 mM KCl 5, 5.8 mM NaCl, 0.75 mM CaCl 2 , 2 mM MgCl 2 , 320 mM sucrose, and 10 mM HEPES pH 7.4).
  • Tissue homogenates are first centrifuged for 10 min at 1000 ⁇ g (4° C.) to remove the nuclear fraction and debris and then the supernatant from the first centrifugation is further centrifuged for 30 min at 35,000 ⁇ g (4° C.) to obtain a partially purified membrane fraction.
  • Membranes are resuspended in the HEPES homogenization buffer prior to being assayed. An aliquot of this membrane homogenate is used to determine protein concentration via the Bradford method (BIO-RAD Protein Assay Kit, #500-0001, BIO-RAD, Hercules, Calif.).
  • Binding studies with [ 3 H]RTX are carried out essentially according to a published protocol (Szallasi and Blumberg, 1992, J. Pharmacol. Exp. Ter. 262: 883-888) in which non-specific RTX binding is reduced by adding bovine alpha, acid glycoprotein (100 ug per tube) after the binding reaction has been terminated. The homogenate is centrifuged as before and resuspended to a protein concentration of 333 ug/ml in homogenization buffer.
  • Binding assay mixtures were set up on ice and contained [ 3 H]RTX (specific activity 2200 mCi/ml), 2 ul non-radioactive ligands test compound, 0.25 mg/ml bovine serum albumin (Cohn fraction V), and 5 ⁇ 10 4 -1 ⁇ 10 5 VR1-transfected cells. The final volume was adjusted to 500 ul (competition binding assays) or 1,000 ul (saturation binding assays) with the ice-cold HEPES homogenization buffer solution (pH 7.4) described above. Non-specific binding was defined as that occurring in the presence of 1 uM non-radioactive RTX. For saturation binding, [ 3 H]RTX was added in the concentration range of 7-1,000 pM, using 1 to 2 dilutions. Typically 11 concentration points are collected per saturation binding curve.
  • K i values for capsaicin receptors of greater than 1 uM are generally less preferred as pharmaceutical agents
  • useful compounds of the invention exhibit K i values for capsaicin receptors of less than 4 uM
  • more preferred compounds exhibit K i values of less than 1 uM
  • even more preferred compounds exhibit K i values of less than 100 nM
  • more highly preferred compounds exhibit K i values of less than 50 nM
  • even more highly preferred compounds exhibit K i values of less than 25 nM
  • the most preferred compounds of the invention yield K i values of less than or about equal to 10 nM in this assay.
  • the following assay can be used to monitor the response of cells capsaicin receptors to capsaicin and other vanilloid ligands of the capsaicin receptor.
  • the assay can also be used to determine if test compounds act as agonists or antagonists of capsaicin receptors.
  • KRH Krebs-Ringer HEPES
  • Agonist e.g., olvanil, capsaicin, or RTX
  • FLUOROSKAN ASCENT Labsystems, Franklin, Mass.
  • FLIPR fluorometric imaging plate reader system, Molecular Devices, Sunnyvale, Calif.
  • agonist e.g. 25-50 nM capsaicin
  • KALEIDAGRAPH software was utilized to fit the data to the equation:
  • y is the maximum fluorescence signal
  • x is the concentration of the agonist or antagonist
  • a is the E max
  • b corresponds to the EC 50 or IC 50 value
  • c is the Hill coefficient.
  • capsaicin EC 50 An additional 20 ul of KRH buffer and 1 ul DMSO is added to each well of cells, prepared as described above. 100 ul capsaicin in KRH buffer is automatically transferred by the FLIPR instrument to each well. An 8-point concentration response curve, with final capsaicin concentrations of 1 nM to 3 uM, is used to determine capsaicin EC 50 .
  • Test compounds are dissolved in DMSO, diluted in 20 ul KRH buffer so that the final concentration of test compounds in the assay well is between 1 uM and 5 uM, and added to cells prepared as described above.
  • the 96 well plates containing prepared cells and test compounds are incubated in the dark, at room temperature for 0.5-6 hours. It is important that the incubation not continue beyond 6 hours.
  • 100 ul capsaicin in KRH buffer at twice the EC 50 concentration determined from the concentration response curve is automatically added by the FLIPR instrument to each well of the 96 well plate for a final sample volume of 200 ul and a final capsaicin concentration equal to the EC 50 .
  • the final concentration of test compounds in the assay wells is between 1 uM and 5 uM.
  • cells exposed to one EC 50 of capsaicin exhibit a fluorescence response of about 10,000 Relative Fluorescence Units.
  • Antagonists of the capsaicin receptor decrease this response by about 20%, preferably by about 50%, and most preferably by at least 80% as compared to matched control.
  • the concentration of antagonist required to provide a 50% decrease is the EC 50 for the antagonist (also referred to as the IC 50 )
  • the ability of a compound to act as an agonist of the capsaicin receptor may be determined by measuring the fluorescence response of cells expressing capsaicin receptors, using the methods described above, in the absence of capsaicin, RTX, or other known capsaicin receptor agonists.
  • Compounds that cause cells to exhibit fluorescence above background are capsaicin receptor agonists.
  • Highly preferred compounds of the invention are antagonists that are essentially free of agonist activity as demonstrated by the absence of detectable agonist activity in such an assay at compound concentrations below 4 nM, more preferably at concentrations below 10 uM and most preferably at concentrations less than or equal to 100 uM.
  • the following assay is a standard assay for NPY Y5 (neuropeptide Y receptor 5) receptor binding that may be used to determine the affinity of compounds for the NPY Y5 receptor.
  • NPY Y5 neuropeptide Y receptor 5
  • Expression of a recombinant human Y5 receptor in cultured cells and receptor binding assays using of membranes prepared from such cells has been described previously, e.g. in U.S. Pat. No. 5,602,024 at columns 17-20.
  • U.S. Pat. No. 5,602,024 is hereby incorporated by reference for its teachings regarding a recombinant human Y5 receptor, expression of this receptor in cultured cells, and receptor binding assays using membranes prepared from such cells.
  • Baculovirus-infected Sf9 cells expressing recombinant human NPY Y5 receptors are harvested at 48 hrs.
  • Sf9 cell pellets are resuspended in lysis buffer (20 mM Tris-HCL, 5 mM EDTA, 0.5 ug/ml leupeptin, 2 ug/ml Aprotinin, 200 uM PMSF, pH 7.4) and homogenized using a POLYTRON homogenizer (setting 3 for 25-30 seconds).
  • the homogenate was centrifuged (536 ⁇ g/10 minutes/4° C.) to pellet the nuclei.
  • the supernatant containing isolated membranes are decanted to a clean centrifuge tube, centrifuged (48,000 ⁇ g/30 minutes, 4° C.) and resuspended in 30 ml homogenization buffer.
  • membranes (10-25 ug) in 150 ul binding buffer are added to polypropylene tubes or 96-well deepwell plates containing [ 125 I]PYY (porcine, NEN, Boston, Mass.)/GTP.
  • Final concentration of [ 125 I]PYY is 30-35 pM/assay well; final concentration of GTP is 100 uM/well.
  • Nonspecific binding is determined in the presence of 1 uM NPY (human, American Peptide Co., Sunnyvale, Calif.) and accounts for less than 10% of total binding.
  • Test compounds at a concentration of 1-4 uM in 2 ul DMSO are added to the assay mixtures. Final assay volume is 250 ul.
  • test compounds are added at concentrations ranging from 10 ⁇ 12 to 10 ⁇ 6 M. Typically 11 concentration points are collected per saturation binding curve. Following a 2-hour incubation at room temperature, the assay reactions are terminated by rapid vacuum filtration. Samples are filtered over presoaked (in polyethyleneimine for 2 hours prior to use) GF/C WHATMAN filters and rinsed 2 times with 5 mls cold binding buffer without BSA. Remaining bound radioactivity is quantified by gamma counting. Ki values may be determined by the method described in Example 10.
  • Preferred compounds of the invention exhibit 10-fold greater affinity for the capsaicin receptor than for the chimeric NPY Y5 receptor, more preferred compounds of the invention also exhibit 100-fold greater affinity for the capsaicin receptor than for the chimeric NPY Y5 receptor, and still more preferred compounds of the invention also exhibit 1000-fold greater affinity for the capsaicin receptor than for the chimeric NPY Y5 receptor. Most highly preferred compounds of the invention do not exhibit detectable binding at the NPY 5 receptor.
  • the compounds of the invention are prepared as radiolabeled probes by carrying out their synthesis using precursors comprising at least one atom that is a radioisotope.
  • the radioisotope is preferably selected from of at least one of carbon (preferably 14 C), hydrogen (preferably 3 H), sulfur (preferably 35 S), or iodine (preferably 125 I).
  • Such radiolabeled probes are conveniently synthesized by a radioisotope supplier specializing in custom synthesis of radiolabeled probe compounds. Such suppliers include Amersham Corporation, Arlington Heights, Ill.; Cambridge Isotope Laboratories, Inc.
  • Tritium labeled probe compounds are also conveniently prepared catalytically via platinum-catalyzed exchange in tritiated acetic acid, acid-catalyzed exchange in tritiated trifluoroacetic acid, or heterogeneous-catalyzed exchange with tritium gas. Such preparations are also conveniently carried out as a custom radiolabeling by any of the suppliers listed in the preceding paragraph using the compound of the invention as substrate. In addition, certain precursors may be subjected to tritium-halogen exchange with tritium gas, tritium gas reduction of unsaturated bonds, or reduction using sodium borotritide, as appropriate.
  • Receptor autoradiography (receptor mapping) is carried out in vitro as described by Kuhar in sections 8.1.1 to 8.1.9 of Current Protocols in Pharmacology (1998) John Wiley & Sons, New York, using radiolabeled compounds of the invention prepared as described in the preceding Example.
  • the most preferred compounds of the invention are suitable for pharmaceutical use in treating human patients. Accordingly, such preferred compounds are non-toxic. They do not exhibit single or multiple dose acute or long-term toxicity, mutagenicity (e.g., as determined in a bacterial reverse mutation assay such as an Ames test), teratogenicity, tumorogenicity, or the like, and rarely trigger adverse effects (side effects) when administered at therapeutically effective dosages.
  • administering does not result in prolongation of heart QT intervals (i.e., as determined by electrocardiography, e.g., in guinea pigs, minipigs or dogs).
  • such doses of such preferred compounds When administered daily for 5 or preferably ten days, such doses of such preferred compounds also do not cause liver enlargement resulting in an increase of liver to body weight ratio of more than 100%, preferably not more than 75% and more preferably not more than 50% over matched controls in laboratory rodents (e.g., mice or rats). In another aspect such doses of such preferred compounds also preferably do not cause liver enlargement resulting in an increase of liver to body weight ratio of more than 50%, preferably preferably not more than 25%, and more preferably not more than 10% over matched untreated controls in dogs or other non-rodent mammals.
  • such doses of such preferred compounds also preferably do not promote the release of liver enzymes (e.g., ALT, LDH, or AST) from hepatocytes in vivo.
  • liver enzymes e.g., ALT, LDH, or AST
  • such doses do not elevate serum levels of such enzymes by more than 100%, preferably not by more than 75% and more preferably not by more than 50% over matched untreated controls in vivo in laboratory rodents.
  • concentrations in culture media or other such solutions that are contacted and incubated with cells in vitro; equivalent to two, fold, preferably five-fold, and most preferably ten-fold the minimum in vivo therapeutic concentration do not cause release of any of such liver enzymes from hepatocytes in vitro into culture medium above baseline levels seen in media from untreated cells.
  • preferred compounds of the invention exert their receptor-modulatory effects with high selectivity. This means that they do not bind to certain other receptors (other than capsaicin receptors) with high affinity, but rather only bind to, activate, or inhibit the activity of such other receptors with affinity constants of greater than 100 nanomolar, preferably greater than 1 micromolar (uM), more preferably greater than 10 uM and most preferably greater than 100 uM.
  • affinity constants of greater than 100 nanomolar, preferably greater than 1 micromolar (uM), more preferably greater than 10 uM and most preferably greater than 100 uM.
  • Such receptors preferably are selected from the group including ion channel receptors, including sodium ion channel receptors, neurotransmitter receptors such as alpha- and beta-adrenergic receptors, muscarinic receptors (particularly m1, m2, and m3 receptors), dopamine receptors, and metabotropic glutamate receptors; and also include histamine receptors and cytokine receptors, e.g., interleukin receptors, particularly IL-8 receptors.
  • ion channel receptors including sodium ion channel receptors, neurotransmitter receptors such as alpha- and beta-adrenergic receptors, muscarinic receptors (particularly m1, m2, and m3 receptors), dopamine receptors, and metabotropic glutamate receptors; and also include histamine receptors and cytokine receptors, e.g., interleukin receptors, particularly IL-8 receptors.
  • the group of other receptors to which preferred compounds do not bind with high affinity also includes GABA A receptors, bioactive peptide receptors (including NPY and VIP receptors), neurokinin receptors, bradykinin receptors (e.g., BK1 receptors and BK2 receptors), and hormone receptors (including androgen receptors, thyrotropin releasing hormone receptors and melanocyte-concentrating hormone receptors).
  • GABA A receptors include GABA A receptors, bioactive peptide receptors (including NPY and VIP receptors), neurokinin receptors, bradykinin receptors (e.g., BK1 receptors and BK2 receptors), and hormone receptors (including androgen receptors, thyrotropin releasing hormone receptors and melanocyte-concentrating hormone receptors).
  • Preferred compounds of the invention do not exhibit significant activity as sodium ion channel blockers, exhibiting less than 15 percent inhibition, and more preferably less than 10 percent inhibition, of sodium channel specific ligand (e.g., batrachotoxin, tetrodotoxin, or saxitoxin) binding when present at a concentration of 4 uM or less.
  • sodium channel specific ligand e.g., batrachotoxin, tetrodotoxin, or saxitoxin
  • Preferred compounds of the invention do not exhibit significant androgen bioactivity, more preferably they do not exhibit significant androgen antagonist activity, e.g., in vivo, in a Hershberger assay, or in vitro, in an assay such as that described by Nellemann et al., Toxicology 2001, 163(1):29-38. Preferred compounds of the invention exhibit less than a 15% inhibition, more preferably less than a 10%, and most preferably less than 5% inhibition of androgen receptor activation in this in vitro assay when present at concentrations of 4 uM or less.
  • significant activity results varying from control at the p ⁇ 0.1 level or more preferably at the p ⁇ 0.05 level of significance as measured using a standard parametric assay of statistical significance such as a student's T test.
  • Compound half-life values may be determined via the following standard liver microsomal half-life assay. Pooled Human liver microsomes are obtained from XenoTech LLC, 3800 Cambridge St., Kansas City, Kansas 66103 (catalog # H0610). Such liver microsomes may also be obtained, e.g., from In Vitro Technologies, Baltimore, Md. 21227, or from Tissue Transformation Technologies, Edison, N.J. 08837. Reactions are preformed as follows:
  • Phosphate buffer 19 mL 0.1 M NaH 2 PO 4 , 81 mL 0.1 Na 2 HPO 4 , adjusted to pH 7.4 with H 3 PO 4 .
  • CoFactor Mixture 16.2 mg NADP, 45.4 mg Glucose-6-phosphate in 4 mL 100 mM MgCl 2 .
  • Glucose-6-phosphate dehydrogenase 214.3 ul glucose-6-phosphate dehydrogenase suspension (Boehringer-Manheim catalog no. 0737224, distributed by Roche Molecular Biochemicals, Indianapolis, Ind. 46250) is diluted into 1285.7 ul distilled water.
  • 6 test reactions are prepared, each containing 25 ul microsomes, 5 ul of a 100 uM solution of test compound, and 399 ul 0.1 M phosphate buffer.
  • a seventh reaction is prepared as a positive control containing 25 ul microsomes, 399 ul 0.1 M phosphate buffer, and 5 ul of a 100 uM solution of a compound with known metabolic properties (e.g. DIAZEPAM or CLOZEPINE). Reactions are preincubated at 39° C. for 10 minutes.
  • 71 ul Starting Reaction Mixture is added to 5 of the 6 test reactions and to the positive control, 71 ul 100 mM MgCl 2 is added to the sixth test reaction, which is used as a negative control.
  • Preferred compounds of the invention exhibit in vitro t 1/2 values of greater than 10 minutes and less than 4 hours. Most preferred compounds of the invention exhibit in vitro half-life values of between 30 minutes and 1 hour in human liver microsomes.
  • MDCK cells ATCC no. CCL-34 (American Type Culture Collection, Manassas, Va.) are maintained in sterile conditions following the instructions in the ATCC production information sheet.
  • test compound or control sample Prior to assay 1 ul of test compound or control sample is pipetted into PACKARD (Meriden, Conn.) clear bottom 96-well plates. Test compounds and control samples are diluted in DMSO to give final concentration in the assay of 10 micromolar, 100 micromolar, or 200 micromolar. Control samples are drug or other compounds having known toxicity properties.
  • Confluent MDCK cells are trypsinized, harvested, and diluted to a concentration of 0.1 ⁇ 10 6 cells/ml with warm (37° C.) VITACELL Minimum Essential Medium Eagle (ATCC catalog # 30-2003). 100 ul of cells in medium is pipetted into each of all but five wells of each 96-well plate. Warm medium without cells (100 ul) is pipetted in the remaining five wells of each plate to provide standard curve control wells. These wells, to which no cells are added, are used to determine the standard curve. The plates are then incubated at 37° C. under 95% O 2 , 5% CO 2 for 2 hours with constant shaking. After incubation, 50 ul of mammalian cell lysis solution is added per well, the wells are covered with PACKARD TOPSEAL stickers, and plates are shaken at approximately 700 rpm on a suitable shaker for 2 minutes.
  • PACKARD substrate solution 50 ul is added to all wells. Wells are covered with PACKARD TOPSEAL stickers, and plates are shaken at approximately 700 rpm on a suitable shaker for 2 minutes. A white PACKARD sticker is attached to the bottom of each plate and samples are dark adapted by wrapping plates in foil and placing in the dark for 10 minutes. Luminescence is then measured at 22° C. using a luminescence counter, e.g. PACKARD TOPCOUNT Microplate Scintillation and Luminescense Counter or TECAN SPECTRAFLUOR PLUS.
  • a luminescence counter e.g. PACKARD TOPCOUNT Microplate Scintillation and Luminescense Counter or TECAN SPECTRAFLUOR PLUS.
  • Luminescence values at each drug concentration are compared to the values computed from the standard curve for that concentration.
  • Preferred test compounds exhibit luminescence values 80% or more of the standard, or preferably 90% or more of the standard, when a 10 micromolar (uM) concentration of the test compound is used.
  • uM micromolar
  • preferred test compounds exhibit luminescence values 50% or more of the standard, or more preferably 80% or more of the standard. Luminescence values less than 50% of the standard indicate a substantial decrease of ATP production.
  • Rats are tested for thermal (as described by Hargreaves K, Dubner R, Brown F, Flores C, Joris J. Pain. 1988 32(1):77-88) and mechanical (as described by Tal M, Eliav E. Pain. 1996 March;64(3):511-8) sensitivities on days 5, 6, and 7. Baseline data should be obtained for each animal prior to CFA injection.
  • Results are conveniently expressed as % of Maximum Potential Efficacy (MPE). 0% MPE is defined as analgesic effect of vehicle, 100% MPE is defined as an animal's return to baseline level of thermal or mechanical sensitivity.
  • This assay determines the effectiveness of compounds of Formulae I-IX and Formulae A-F in relieving at least one of the symptoms in an in vivo model of pain produced by spinal nerve ligation, namely mechanical allodynia.
  • Tactile allodynia is induced in rats using the procedures described by Bennet and Xie, Pain 1988, 33:87-107. Rats are anesthetized, e.g., with an intraperitoneal dose of pentobarbital sodium (65 mg/kg) with additional doses of anesthetic given as needed. The lateral aspect of each rat's hind limb is shaved and disinfected. Using aseptic technique, an incision is made on the lateral aspect of the hind limb at the mid-thigh level. The biceps femoris is bluntly dissected to expose the sciatic nerve.
  • each rat On one hind limb of each rat, four loosely tied ligatures are made around the sciatic nerve approximately 1-2 millimeters apart. On the other side of each rat, an identical dissection is performed except that the sciatic nerve is not ligated. The muscle is closed with a continuous suture pattern, and the skin is closed with wound clips or sutures.
  • Certain preferred compounds of Formulae I-IX and Formulae A-F are effective in reversing mechanical allodynia-like symptoms (i.e., rats treated with effective amounts of such compounds will require stimulation with a Von Frey filament of higher rigidity strength to provoke a positive allodynic response as compared to control untreated or vehicle treated rats) when tested by this method.
  • This assay determines the effectiveness of compounds in relieving one of the symptoms of neuropathic pain produced by unilateral mononeuropathy, namely cold allodynia.
  • Unilateral mononeuropathy is produced in rats using the Chronic Constriction Injury model performed essentially as described by Bennet and Xie, Pain 1988, 33:87-107. Rats are anesthetized. The lateral aspect of each rat's hind limb is shaved and disinfected. Using aseptic technique, an incision is made on the lateral aspect of the hind limb at the mid-thigh level. The biceps femoris is bluntly dissected to expose the sciatic nerve. On one hind limb of each rat, four loosely tied ligatures are made around the sciatic nerve approximately 1-2 millimeters apart. On the other side of each rat, an identical dissection is performed except that the sciatic nerve is not ligated. The muscle is closed with a continuous suture pattern, and the skin is closed with wound clips or sutures.
  • Rats demonstrating unilateral mononeuropathy are assessed for acute and chronic cold allodynia sensitivity. Each rat is placed individually into a chamber with a metal plate about 6 cm from the bottom. This chamber is filled with ice water to a depth of about 2.5 cm above the metal plate, with the temperature of the bath maintained at about zero to four degrees C. throughout the experiment. A timer is started, and the rat's response latency is measured to the nearest tenth of a second. A “response” is defined as a rapid withdrawal of the ligated hindpaw completely out of the water while the animal is stationary and not pivoting. An exaggerated limp while the animal is walking is not scored as a response. Maximum immersion time is 20 seconds with a 20-minute interval between trials.
  • the screening criteria are 1) the average of two trials is less than or equal to 13 seconds, and 2) there is consistency across the two trial scores. Animals are screened for hypersensitivity to cold on post-surgery days 4 through 10, and selected for inclusion in drug-response studies based on the criteria described above. The pre-dose screening values are used as the animal's baseline cold allodynia scores. For acute studies, the animals are tested for cold allodynia at 1, 3, and sometimes 5 hours post-dose.
  • This assay determines the effectiveness of compounds in relieving one of the symptoms of neuropathic pain produced by unilateral mononeuropathy, namely mechanical hyperalgesia.
  • a chronic constriction injury is produced by loosely ligating the right common sciatic nerve as described by Bennet and Xie, Pain 1988, 33:87-107.
  • the left common sciatic nerve is visualized, but not manipulated to produce sham conditions.
  • Rats having a chronic constriction injury are assessed for mechanical hyperalgesia to a pin-prick stimulus as described by Koch et al. Analgesia 1996, 2(3), 157-164. Rats are placed in individual compartments of a cage with a warmed, perforated metal floor. Hindpaw withdrawal duration is measured after a mild pinprick to the plantar surface of the ligated and sham hindpaws.
  • Preferred compounds of the invention produce a reduction of mechanical hyperalgesia (i.e., a reduction in the duration of hindpaw withdrawal) elicited by a pin-prick stimulus in rats with a chronic constriction injury at doses of 50 mg/kg or less when tested by this method.
  • mechanical hyperalgesia i.e., a reduction in the duration of hindpaw withdrawal
  • This assay determines the effectiveness of compounds in relieving one of the symptoms of neuropathic pain produced by unilateral mononeuropathy, namely thermal hyperalgesia.
  • Rats having had surgery as described in Example 18d are assessed for thermal hyperalgesia sensitivity at least 10 days post-surgery.
  • the rats are placed beneath inverted cages upon an elevated glass platform and a radiant heat source beneath the glass is aimed at the plantar hindpaw.
  • the duration of time before the hindpaw is withdrawn from the floor is measured to the nearest tenth of a second.
  • the cutoff time for the heat stimulus is 20 seconds, and the light is calibrated such that this stimulus duration does not burn or blister the skin.
  • Preferably about four latency measurements are taken for each hindpaw in each test session, alternating left and right hindpaws, with 5-minute intervals between tests. The times to withdrawal of each side are averaged and a difference score is obtained.
  • Preferred compounds of the invention produce an increase in the average time to withdrawal after oral administration of 50 mg/kg or less in this model.
  • Sedation may be determined using the method described by Fitzgerald et al., Toxicology 1988, 49(2-3)433-9.
  • Preferred compounds of the invention do not produce reproducible or significant sedation at intravenous doses of less than 25 mg/kg (preferably less than 10 mg/kg) or at oral doses of less than 140 mg/kg (preferably less than 50 mg/kg).

Abstract

Disclosed are diaryl piperazines and related compounds. These compounds are selective modulators of capsaicin receptors, including human capsaicin receptors, that are, therefore, useful in the treatment of a chronic and acute pain conditions, itch and urinary incontinence. Methods of treatment of such disorders and well as packaged pharmaceutical compositions are also provided. Compounds of the invention are also useful as probes for the localization of capsaicin receptors and as standards in assays for capsaicin receptor binding and capsaicin receptor mediated cation conductance. Methods of using the compounds in receptor localization studies are given.

Description

    RELATED APPLICATION INFORMATION
  • This application claims the benefit of priority under 35 U.S.C. section 119(e) to Provisional Patent Application 60/280,223, filed Mar. 30, 2001, 60/230,726, filed Sep. 7, 2000, and 60/219,529, filed Jul. 20, 2001. These applications are hereby incorporated by reference in their entirety.[0001]
  • BACKGOUND OF THE INVENTION
  • 1. Field of the Invention [0002]
  • This invention relates compounds that bind with high selectivity and high affinity to Vanilloid Receptors, especially Type I Vanilloid Receptors, also known as capsaicin receptors or VR1 Receptors. In an important aspect the invention provides capsaicin receptor, preferably human VR1 receptor, antagonists that are not capsaicin analogs (e.g., they do not contain a phenyl ring with two oxygen atoms bound to two adjacent ring carbons), are free of agonist activity, and exhibit an unprecedented level of affinity for the VR1 receptor. In another aspect, the invention provides aryl piperazines and related compounds that act as VR1 receptor ligands. In addition, this invention relates to such VR1 receptor ligands, high affinity antagonists and pharmaceutical compositions comprising such compounds and to the use of such compounds in treatment of diseases and other health-related conditions. Additionally this invention relates to the use of aryl piperazines and related compounds as tool for the analysis of VR1 receptors and as probes for the quantitative measurement and localization of VR1 receptors in cell and tissue samples. [0003]
  • 2. Background [0004]
  • The sensation of pain can be triggered by any number of physical or chemical stimuli. In mammals, the peripheral terminals of a group of specialized small diameter sensory neurons, termed “nociceptors” mediate this response to a potentially harmful stimulus. [0005]
  • In efforts to discover better analgesics for the treatment of both acute and chronic pain, and to develop treatments for various neuropathic pain states, considerable research has been focused on the molecular mechanism of nociception. The response to heat, low extracellular pH (acidity), or capsaicin (the compound responsible for the hotness of hot peppers) is characterized by the persistent activation of nociceptors. It has been shown that both heat and capsaicin are capable of activating dorsal root ganglion and trigeminal ganglion neurons via an influx of cations. Additionally, moderately acidic conditions produce this response and can also potentiate the response of nociceptors to heat and capsaicin. [0006]
  • Capsaicin responses in isolated sensory neurons show dose-dependence and are also evoked by structural analogues of capsaicin that share a common vanilloid moiety. The term vanilloid receptor (VR) was coined to describe the neuronal membrane recognition site for capsaicin and such related irritant compounds. It was postulated that the VR is a nonselective cation channel with a preference for calcium. In 1989, resiniferatoxin (RTX), a natural product of certain Euphorbia plants, was recognized as an ultrapotent VR agonist. Specific binding of 3H RTX provided the first unequivocal proof for the existence of a vanilloid receptor. The capsaicin response is competitively inhibited (and thereby antagonized) by another capsaicin analog, capsazepine and is also inhibited by the non-selective cation channel blocker ruthenium red. These antagonists bind to VR with no more than moderate affinity (i.e., with Ki values of no lower than 140 uM). [0007]
  • Interest in characterizing VRs led to the cloning of a functional rat capsaicin receptor (VR1), from a rat dorsal root ganglion cDNA library. A human version of VR1 has also been described, and the term VR1 is used herein to refer to either or both. [0008]
  • The capsaicin receptor's channel opens in response to elevated temperatures (higher than about 45° C.). Capsaicin and related compounds, as well as protons are stimuli that lower the threshold channel opening, so that in the presence of any of these stimuli the capsaicin receptor can be opened even at room temperature. [0009]
  • Opening of the capsaicin receptor channel is followed by the release of inflammatory peptides from neurons expressing the receptor and other nearby neurons, increasing the pain response. After initial activation by capsaicin the capsaicin receptor undergoes a rapid desensitization, possibly via phosphorylation of intracellular sites of the receptor. Capsaicin and related VR1 agonist vanilloid compounds have enjoyed long-pharmaceutical use as topical anaesthetics. While such compounds initially cause a strong burning sensation, receptor desensitization provides pain relief. [0010]
  • Localization of the capsaicin receptor in the dorsal root ganglion established this receptor as a leading target for analgesic discovery. Most currently marketed analgesic compounds act centrally, and often have side effects. Analgesic compounds that act peripherally are desirable for treating acute and chronic pain more effectively and with fewer side effects. Thus, compounds that interact with the capsaicin receptors, particularly antagonists of this receptor, which would not elicit the initial painful sensation of currently marketed capsaicin containing compounds, are desirable for the treatment of chronic and acute pain, itch, and urinary incontinence. [0011]
  • 3. Description of Related Art [0012]
  • The vanilloid compounds capsaicin and Resiniferatoxin (RTX) act as potent and specific agonists of the capsaicin receptor. Capsazepine (which contains a phenyl ring with two oxygen atoms bound to two adjacent ring carbons and is therefore a capsaicin analog) acts as a moderate affinity competitive capsaicin receptor antagonist. Iodo-RTX is a capsaicin analog that has recently been reported to act as a high affinity antagonist. The inorganic dye, Ruthenium red, also antagonizes capsaicin responses of the receptor, albeit as a non-selective cation channel blocker. For an extensive review of vanilloid receptor ligands see Szallasi and Blumberg, (Pharmacological Reviews (1999) 51(3): 159-211). [0013]
  • SUMMARY OF THE INVENTION
  • This invention relates to VR1 receptor ligands, particularly VR1 receptor antagonists, and methods of using VR1 receptor antagonists for the treatment of neuropathic pain, peripheral-nerve-mediated pain, and pain, inflammatory and broncho-constriction symptoms resulting from exposure to capsacin-receptor-activating stimuli such as capsaicin and tear gas. [0014]
  • In one aspect the invention provides novel chemical compounds that act as capsaicin receptor modulatory agents, some of which exhibit antagonist potency greater than that of any previously described VR1 receptor antagonist. Compounds that act as capsaicin receptor antagonists and bind to capsaicin (preferably human VR1) receptors with K[0015] i values of less than 100 uM, as measured by a capsacin receptor binding assay, such as the assay given by Example 10, or that inhibit capsaicin activity in an assay for determination of capsaicin receptor antagonist effects (Example 11) with EC50 values of less than or equal to 100 uM, are referred to herein as potent capsaicin receptor antagonists; such compounds that bind or antagonize with K1 or EC50 values of less than or equal to 10 uM are referred to herein as highly potent capsaicin receptor antagonists.
  • In an additional aspect, the invention provides methods of using the potent capsaicin receptor antagonist compounds of the invention for the treatment of symptoms resulting from exposure to painful capsaicin receptor activating stimuli. In particular, the invention provides methods of treating subjects who have been exposed to capsaicin or have been burnt by heat, light, tear gas, or acid exposure, the methods comprising administering to such subjects an effective amount of a potent capsaicin receptor antagonist, preferably a highly potent. (high potency) capsaicin receptor antagonist, so that the subject's symptoms of pain or sensitivity are reduced. Preferred compounds of the invention provide pain relief without loss of consciousness, and preferably without sedation, in such subjects that is equal to or grater than the degree of pain relief that can be provided to such subjects by morphine without loss of consciousness. Highly preferred compounds provide such pain relief while causing only transient (i.e., lasting for no more than one half the time that pain relief lasts) or no sedation (see Example 16 for sedation assay). Subjects or patients referred to herein may be humans or non-human mammals including domestic companion animals (pets) and livestock animals, as discussed more fully below. [0016]
  • In yet another aspect the invention provides methods of treating of neuropathic pain based on the unexpected finding that capsaicin receptor antagonists can alleviate such pain. [0017]
  • This invention also provides aryl piperazines and related compounds that bind with high affinity and high selectivity to capsaicin receptors, including human capsaicin receptors, also known as VR1 receptors. [0018]
  • Thus, the invention provides novel compounds of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, or Formula VII, Formula VIII, Formula IX and Formulae A-F shown below (the “compounds of the invention,” hereinafter Formulae I-IX and Formulae A-F), and pharmaceutical compositions comprising compounds of Formulae I-IX and Formulae A-F. [0019]
  • The invention further comprises methods of treating patients suffering from certain diseases or conditions, especially those involving pain or urinary incontinence, with an amount of a compound Formulae I-IX and Formulae A-F that is effective to improve the symptoms (e.g., reduce pain or reduce the frequency of urinary incontinence) of the disease or condition being treated. [0020]
  • Additionally this invention relates to the use of the compounds of the invention as reagents, standards, and probes for measurement, characterization and localization of capsaicin receptors, particularly VR1 receptors (e.g., in cells or tissues. [0021]
  • Accordingly, a broad aspect of the invention is directed to compounds of Formula I: [0022]
    Figure US20040176443A1-20040909-C00001
  • or the pharmaceutically acceptable salts thereof, [0023]
  • wherein: [0024]
  • A is chosen from O, S, NR[0025] A, CRBRB′, NRACRBRB′, CRBRB′NRA, —CRA═CRB—, and C3H4; where RA, RB, and RB′ are independently selected, at each occurrence from hydrogen or alkyl;
  • Z is oxygen or sulfur; [0026]
  • R[0027] 1 and R2 independently represent hydrogen or lower alkyl; or
  • R[0028] 1 and R2 are taken together to form a 5 to 8 membered nitrogen containing ring of the formula:
    Figure US20040176443A1-20040909-C00002
  • wherein n is 1, 2, or 3; [0029]
  • R[0030] 3 and R4 are independently selected at each occurrence from the group consisting of hydrogen; halogen; hydroxy; amino; cyano; nitro; —COOH; —CHO, optionally substituted alkyl; optionally substituted alkenyl; optionally substituted alkynyl; optionally substituted alkoxy; optionally substituted mono or dialkylamino; optionally substituted alkylthio; optionally substituted alkyl ketone; optionally substituted alkylester; optionally substituted alkylsulfinyl; optionally substituted alkylsulfonyl; optionally substituted mono- or di-alkylcarboxamide; optionally substituted —S(O)nNHalkyl; optionally substituted —S(O)nN(alkyl)(alkyl); optionally substituted —NHC(═O)alkyl; optionally substituted —NC(═O)(alkyl)(alkyl); optionally substituted —NHS(O)nalkyl; optionally substituted —NS(O)n(alkyl)(alkyl); optionally substituted saturated or partially unsaturated heterocycloalkyl of from 5 to 8 atoms, which saturated or partially unsaturated heterocycloalkyl contains 1, 2, or 3 heteroatoms selected from N, O, and S; optionally substituted aryl having from 1 to 3 rings; or optionally substituted heteroaryl, said heteroaryl having from 1 to 3 rings, 5 to 8 ring members in each ring and, in at least one of said rings, from 1 to about 3 heteroatoms per ring selected from the group consisting of N, O, and S;
  • or any two [0031]
  • R[0032] 3 and R4 not attached to the same carbon may be joined to form an optionally substituted aryl ring; a saturated or partially unsaturated carbocyclic ring of from 5 to 8 members, which carbocyclic ring is optionally substituted; or a saturated, partially unsaturated, or aromatic heterocyclic ring of from 5 to 8 members, which heterocyclic ring is optionally substituted and contains 1, 2, or 3 heteroatoms selected from N, O, and S; and
  • Ar[0033] 1 and Ar2 are the same or different and independently represent optionally substituted cycloalkyl; an optionally substituted heterocycloalkyl ring of from 5 to 8 atoms, which heterocyloalkyl ring contains 1, 2, or 3 heteroatoms selected from N, O, and S; optionally substituted aryl having from 1 to 3 rings; or optionally substituted heteroaryl, said heteroaryl having from 1 to 3 rings, 5 to 8 ring members in each ring and, in at least one of said rings, from 1 to about 3 heteroatoms per ring selected from the group consisting of N, O, and S, and
  • n is independently chosen at each occurrence from 0, 1, and 2. [0034]
  • In specific embodiments of the invention R[0035] 1 and R2 are joined to form a 5 to 7-membered heterocycloalkyl ring, e.g. R1 and R2 may be joined to form a piperazine ring. This 5 to 7-membered heterocycloalkyl ring is preferably unsubstituted or substituted at one or two positions with a C1-6 alkyl group, such as methyl or ethyl. The variable “Z” is preferably oxygen and the variable “A” is generally NH, CH═CH, or CH2NH. Ar1 and Ar2 are preferably optionally substituted phenyl or optionally substituted pyridyl; optionally substituted 2-pyridyl is preferred for Ar2. Substitutuents that may occur on Ar1 and Ar2 include, but are not limited to, butyl, isopropyl, trifluoromethyl, nitro, methyl, and halogen. Substitution at the 4 position of Ar1 (when Ar1 is phenyl or pyridyl) and substitution at the 3 position of Ar2 (when Ar2 is phenyl or pyridyl) are described in specific embodiments of the invention.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The invention is particularly directed to compounds of Formula I, in which R[0036] 1 and R2 independently represent hydrogen or lower alkyl, e.g., C1-6alkyl. Such compound will be referred to as compounds of Formula IA.
  • Preferred compounds and pharmaceutically acceptable salts of Formula IA are those wherein: [0037]
  • R[0038] 3 and R4 are independently chosen at each occurrence from the group consisting of hydrogen, halogen, cyano, nitro, haloalkyl, haloalkoxy, hydroxy, amino, alkyl substituted with 0-2 R6, alkenyl substituted with 0-2 R6; alkynyl substituted with 0-2 R6; alkoxy substituted with 0-2 R6, —NH(alkyl) substituted with 0-2 R6, —N(alkyl)(alkyl) where each alkyl is independently substituted with 0-2 R6, —XR7, and Y;
  • or any two [0039]
  • R[0040] 3 and R4 not attached to the same carbon may be joined to form an aryl ring substituted with 0-3 R6, a saturated or partially unsaturated carbocyclic ring of from 5 to 8 members, which carbocyclic ring is substituted with 0-2 R6, or a saturated, partially unsaturated, or aromatic heterocyclic ring of from 5 to 8 members, which heterocyclic ring is substituted with 0-2 R6 and contains 1, 2, or 3 heteroatoms selected from N, O, and S; and
  • Ar[0041] 1 and Ar2 may be the same or different and are selected from the group consisting of cyclohexyl, cyclopentyl, piperidinyl, piperazinyl, phenyl, pyrrolyl, furanyl, thienyl, pyrazolyl, imidazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, triazolyl, tetrazolyl, pyridyl, pyrimidyl, pyrazinyl, benzimidazolyl, naphthyl, indolyl, isoindolyl, benzofuranyl, isobenzofuranyl, benzo[b]thiophenyl, benz[d]isoxazolyl, quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, and quinoxalinyl, each of which is optionally mono-, di-, or trisubstituted with R5; or
  • Ar[0042] 1 and Ar2 may be the same or different and represent a bicyclic oxygen-containing group of the formula:
    Figure US20040176443A1-20040909-C00003
  • optionally mono-, di-, or trisubstituted with R[0043]   5, where L represents point of attachment and may be at any point on the benzene ring, and the oxygen-containing ring of the bicyclic oxygen-containing group consists of from 5 to 8 ring atoms, contains 1 or 2 oxygen atoms and remaining ring atoms are carbon;
  • R[0044] 5 is independently selected at each occurrence from the group consisting of halogen, cyano, nitro, haloalkyl, haloalkoxy, hydroxy, amino, alkyl substituted with 0-2 R6, alkenyl substituted with 0-2 R6, alkynyl substituted with 0-2 R6, alkoxy substituted with 0-2 R6, —NH(alkyl) substituted with 0-2 R6, —N(alkyl)(alkyl) where each alkyl is independently substituted with 0-2 R6, —XR7, and Y;
  • R[0045] 6 is independently selected at each occurrence from the group consisting of halogen, hydroxy, cyano, alkyl, alkoxy, —NH(alkyl), —N(alkyl)(alkyl), —S(O)n(alkyl), haloalkyl, haloalkoxy, CO(alkyl), CONH(alkyl), CON(alkyl1)(alkyl2) where alkyl1 and alkyl2 may be joined to form a heterocycloalkyl ring of from 5 to 8 ring atoms and containing 1, 2, or 3 heteroatoms selected from N, O, and S, —XR7, and Y;
  • X is independently selected at each occurrence from the group consisting of —CH[0046] 2—, —CHR8—, —O—, —S(O)n—, —NH—, —NR8—, —C(═O)—, —C(═O)O—, —C(═O)NH—, —C(═O)NR8—, —S(O)nNH—, —S(O)nNR8—, NHC(═O)—, —NR8C(═O)—, —NHS(O)n—, and —NR8S(O)n—;
  • R[0047] 7 and R8 are independently selected at each occurrence from hydrogen, and
  • straight, branched, and cyclic alkyl groups, and (cycloalkyl)alkyl groups, said straight, branched, and cyclic alkyl groups, and (cycloalkyl)alkyl groups consisting of 1 to 8 carbon atoms, and containing zero or one or more double or triple bonds, each of which 1 to 8 carbon atoms may be further substituted with one or more substituent(s) independently selected from oxo, hydroxy, halogen, amino, cyano, nitro, haloalkyl, haloalkoxy, —O(alkyl), —NH(alkyl), —N(alkyl)(alkyl), —NHC(O)(alkyl), —N(alkyl)C(O)(alkyl), —NHS(O)[0048]   n(alkyl), —S(O)n(alkyl), —S(O)nNH(alkyl), —S(O)nN(alkyl3)(alkyl4) where alkyl3 and alkyl4 may be joined to form a heterocycloalkyl ring consisting of from 5 to 8 ring atoms and containing 1, 2, or 3 heteroatoms selected from N, O, and S, and Y′;
  • Y and Y′ are independently selected at each occurrence from 3- to 8-membered carbocyclic or heterocyclic groups which are saturated, unsaturated, or aromatic, which may be further substituted with one or more substituents independently selected from halogen, oxo, hydroxy, amino, nitro, cyano, alkyl, alkoxy, haloalkyl, haloalkoxy, mono- or dialkylamino, and alkylthio; [0049]
  • wherein said 3- to 8-memberered heterocyclic groups contain one or more heteroatom(s) independently selected from N, O, and S; and [0050]  
  • n is independently chosen at each occurrence from 0, 1, and 2. [0051]
  • Such compounds and pharmaceutically acceptable salts thereof, will be referred to as compounds of Formula IB. [0052]
  • The invention is further directed to compounds of Formula II [0053]
    Figure US20040176443A1-20040909-C00004
  • and the pharmaceutically acceptable salts thereof, wherein: [0054]
  • A, Z, R[0055] 3, R4 are as defined for Formula I or for Formula IB;
  • Ar[0056] 1 and Ar2 are as defined for Formula I or for formula IB; and
  • x is 1 or 3. [0057]
  • Preferred compounds and salts of Formula II are those in which [0058]
  • R[0059] A, RB, and RB′ (which are contained in the definition of A) are independently selected at each occurrence from hydrogen or C1-6alkyl.
  • Other preferred compounds salts of Formula II are those in which Z is oxygen, and those in which Z is oxygen and A is NH. [0060]
  • The invention is further directed to compounds of Formula III [0061]
    Figure US20040176443A1-20040909-C00005
  • and the pharmaceutically acceptable salts thereof, wherein: [0062]
  • G, Q, T, and W are the same or different and represent N, CH, or CR[0063] 5, where R5 is as defined for Formula IB;
  • R[0064] A, RB, and RB′ are independently selected at each occurrence from hydrogen or C1-6alkyl;
  • Z is oxygen or sulfur; [0065]
  • R[0066] 3 and R4 are as defined for Formula I or for Formula IB; and
  • x is 1 or 3. [0067]
  • The invention also included compounds of Formula IV [0068]
    Figure US20040176443A1-20040909-C00006
  • and the pharmaceutically acceptable salts thereof, wherein: [0069]
  • Z is S or O (preferably O); [0070]
  • A, R[0071] 3, and R4 is as defined for Formula I or Formula IB;
  • Ar[0072] 1 and Ar2 may be the same or different and are selected from the group consisting of cyclohexyl, cyclopentyl, piperidinyl, piperazinyl, pyrrolyl, furanyl, thienyl, pyrazolyl, imidazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, triazolyl, tetrazolyl, pyridyl, pyrimidyl, pyrazinyl, benzimidazolyl, naphthyl, indolyl, isoindolyl, benzofuranyl, isobenzofuranyl, benzo[b]thiophenyl, benz[d]isoxazolyl, quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, and quinoxalinyl; wherein Ar1 is optionally mono-, di-, or trisubstituted with R5, and Ar2 is optionally mono-, di-, or trisubstituted with R9; or
  • Ar[0073]   1 and Ar2 may be the same or different and represent a bicyclic oxygen-containing group as described for Formula IB,
  • R[0074] 5 is independently selected at each occurrence from the group consisting of cyano, nitro, haloalkyl, haloalkoxy, hydroxy, amino, alkyl substituted with 0-2 R6, alkenyl substituted with 0-2 R6, alkynyl substituted with 0-2 R6, alkoxy substituted with 0-2 R6, —NH(alkyl) substituted with 0-2 R6, —N(alkyl)(alkyl) where each alkyl is independently substituted with 0-2 R6, —XR7, and Y;
  • R[0075] 9 is independently selected at each occurrence from the group consisting of cyano, nitro, haloalkoxy, hydroxy, amino, alkyl substituted with 0-2 R6, alkenyl substituted with 0-2 R6, alkynyl substituted with 0-2 R6, alkoxy substituted with 0-2 R6, —NH(alkyl) substituted with 0-2 R6, —N(alkyl)(alkyl) where each alkyl is independently substituted with 0-2 R6, —XR7, and Y; and
  • R[0076] 6, R7, R8, X, Y and Y′ are as defined for Formula IB.
  • Another embodiment of the invention is directed to compounds of Formula V [0077]
    Figure US20040176443A1-20040909-C00007
  • and the pharmaceutically acceptable salt thereof, wherein: [0078]
  • G, Q, T, and W are the same or different and are selected from the group consisting of N, CH, and CR[0079] 5, wherein T or W or both is N;
  • A, R[0080] 3, and R4 are as defined for Formula I or for Formula IB (preferably A is —CH═CH—, —CH2NH, NH, and R3 and R4 are hydrogen or C1-6 alkyl);
  • Z is oxygen or sulfur (preferably oxygen); [0081]
  • R[0082] 5 represents 1 to 3 substituents and is independently selected at each occurrence from the group consisting of cyano, hydroxy, amino, C3-6 alkyl substituted with 0-2 R6, C2-6 alkenyl substituted with 0-2 R6, C2-6 alkynyl substituted with 0-2 R6, C3-6 alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6, —N(C1-6alkyl)(C1-6alkyl) where each alkyl is independently substituted with 0-2 R6, —XR7, and Y;
  • R[0083] 9 represents 0 to 3 substituents and is independently selected at each occurrence from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl substituted with 0-2 R6, C2-6alkenyl substituted with 0-2 R6, C2-6alkynyl substituted with 0-2 R6, C1-6alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6, —N(C1-6alkyl)(C1-6alkyl) where each C1-6alkyl is independently substituted with 0-2 R6, —XR7, and Y;
  • R[0084] 6, R7, R8, X, Y, and Y′ are as defined for Formula IB.
  • The invention is particularly directed to compounds and salts of Formula V wherein G and Q are selected from the group consisting of CH and CR[0085] 5.
  • The invention is also directed to compounds and salts of Formula V wherein G, Q, and W are independently selected at each occurrence from the group consisting of CH and CR[0086] 5; and T is N.
  • For compounds of Formula V, particularly preferred R[0087] 6 substituents are halogen, hydroxy, C1-4 alkyl, C1-4 alkoxy, —NH(C1-4alkyl), and —N(C1-4 alkyl)(C1-4 alkyl).
  • Still another embodiment of the invention is directed of compounds of Formula VI [0088]
    Figure US20040176443A1-20040909-C00008
  • and the pharmaceutically acceptable salts thereof, wherein: [0089]
  • A is selected from the group consisting of NH, —CH═CH—, and CH[0090] 2NH;
  • R[0091] 4 is independently chosen from hydrogen and C1-4 alkyl;
  • R[0092] 5 represents 0 to 2 substituents and is independently chosen at each occurrence from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl substituted with 0-2 R6, C2-6alkenyl substituted with 0-2 R6, C2-6alkynyl substituted with 0-2 R6, C1-6alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6, —N(C1-6alkyl)(C1-6alkyl) where each C1-6alkyl is independently substituted with 0-2 R6;
  • R[0093] 9 represents 0 to 2 substituents and is independently chosen at each occurrence from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl substituted with 0-2 R6, C2-6alkenyl substituted with 0-2 R6, C2-6alkynyl substituted with 0-2 R6, C1-6alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6, and —N(C1-6alkyl)(C1-6alkyl) where each C1-6alkyl is independently substituted with 0-2 R6;
  • R[0094] 5A is independently selected from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6 alkyl, C1-6 alkoxy, —NH(C1-6 alkyl), and —N(C1-6 alkyl)(C1-6 alkyl)
  • R[0095] 9B is independently selected from the group consisting of halogen, nitro, halo(C1-6)alkoxy, hydroxy, amino, C1-6 alkyl, C1-6 alkoxy, —NH(C1-6 alkyl), and —N(C1-6 alkyl)(C1-6 alkyl); and
  • R[0096] 6 is independently selected at each occurrence the group consisting of halogen, hydroxy, C1-4alkyl, C1-4alkoxy, —NH(C1-4 alkyl), and —N(C1-4 alkyl)(C1-4 alkyl)
  • The invention is also directed to compounds of Formula VII [0097]
    Figure US20040176443A1-20040909-C00009
  • and the pharmaceutically acceptable salts thereof, wherein: [0098]
  • A, R[0099] 3, and R4 are as defined for Formula I or for Formula IB;
  • R[0100] 5 is independently selected at each occurrence from the group consisting of cyano, nitro, haloalkyl, haloalkoxy, C1-6 alkyl substituted with 0-2 R6, C2-6 alkenyl substituted with 0-2 R6, C2-6 alkynyl substituted with 0-2 R6, C1-6 alkoxy substituted with 0-2 R6, —NH(C1-6 alkyl) substituted with 0-2 R6, —N(C1-6 alkyl)(C1-6 alkyl) where each alkyl is independently substituted with 0-2 R6, —XR7, and Y;
  • R[0101] 9 represents 0-3 substituents and is independently selected at each occurrence from the group consisting of bromo, haloalkyl, haloalkoxy, hydroxy, C2-6 alkyl substituted with 0-2 R6, C2-6 alkenyl substituted with 0-2 R6, C2-6 alkynyl substituted with 0-2 R6, C2-6 alkoxy substituted with 0-2 R6, —NH(C2-6 alkyl) substituted with 0-2 R6, —N(C2-6 alkyl)(C2-6 alkyl) where each C2-6 alkyl is independently substituted with 0-2 R6, —XR7, and Y;
  • R[0102] 6, R7, R8, X, Y, and Y′ are as defined for Formula IB.
  • Preferred compounds and salts of Formula VII include those wherein A is selected from NH, —CH═CH—, and CH[0103] 2NH; and R6 is independently selected at each occurrence from the group consisting of halogen, hydroxy, C1-4 alkyl, C1-4 alkoxy, —NH(C1-4 alkyl), and —N(C1-4 alkyl)(C1-4 alkyl).
  • The invention includes compounds of Formula VIII [0104]
    Figure US20040176443A1-20040909-C00010
  • and the pharmaceutically acceptable salts thereof, wherein: [0105]
  • A is selected from the group consisting of NH, —CH═CH—, and CH[0106] 2NH (NH is preferred);
  • R[0107] 4 is independently selected at each occurrence from hydrogen and C1-4alkyl;
  • R[0108] 5 represents 0 to 2 substituents independently selected at each occurrence from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl substituted with 0-2 R6, C2-6alkenyl substituted with 0-2 R6, C2-6alkynyl substituted with 0-2 R6, C1-6alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6, and —N(C1-6alkyl)(C1-6alkyl) where each C1-6alkyl is independently substituted with 0-2 R6;
  • R[0109] 9 represents 0 to 2 substituents and is independently selected at each occurrence from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl substituted with 0-2 R6, C2-6alkenyl substituted with 0-2 R6, C2-6alkynyl substituted with 0-2 R6, C1-6alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6, and —N(C1-6alkyl)(C1-6alkyl) where each C1-6alkyl is independently substituted with 0-2 R6;
  • R[0110] 5A is independently selected from the group consisting of halogen, cyano, nitro, trifluoromethyl, trifluoromethoxy, hydroxy, amino, C1-6 alkyl, C1-6 alkoxy, —NH(C1-6 alkyl), and —N(C1-6 alkyl)(C1-C6 alkyl);
  • R[0111] 9B is independently selected from the group consisting of trifluoromethoxy, hydroxy, C2-6 alkyl, C2-6 alkoxy, —NH(C2-6 alkyl), and —N(C2-6 alkyl)(C2-6 alkyl); and
  • R[0112] 6 is independently selected at each occurrence from the group consisting of halogen, hydroxy, C1-4 alkyl, C1-4 alkoxy, —NH(C1-4 alkyl), and —N(C1-4 alkyl)(C1-4 alkyl)
  • Preferred compound of Formula VIII are those wherein one of R[0113] 4 is hydrogen and the other is methyl.
  • A particularly preferred embodiment of the invention includes compounds of Formula IX [0114]
    Figure US20040176443A1-20040909-C00011
  • and the pharmaceutically acceptable salts thereof, wherein: [0115]
  • A, R[0116] 3, and R4 are as defined for Formula I or for Formula IB;
  • R[0117] 5 is selected from the group consisting of bromo, fluoro, iodo, halo(C1-6)alkyl, halo(C3-6)alkoxy, C3-6alkyl substituted with 0-3 R6, C2-6alkenyl substituted with 0-3 R6, C2-6alkynyl substituted with 0-3 R6, C3-6alkoxy substituted with 0-2 R6, (C3-8cycloalkyl)C1-4alkyl, —NH(C1-6alkyl) substituted with 0-2 R6, —N(C1-6alkyl)(C1-6alkyl) where each C1-6alkyl is substituted with 0-2 R6, Y, —(C═O)Y, —(CH2)Y, and —(CH(CN))Y;
  • R[0118] 9 is selected from the group consisting of halogen, cyano, —N(SO2C1-6alkyl)(SO2C1-6alkyl), —SO2NH2, halo (C1-6) alkyl, halo(C1-6)alkoxy, C1-6alkyl substituted with 0-2 R6, C2-6alkenyl substituted with 0-2 R6, C2-6alkynyl substituted with 0-2 R6, C1-6alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6, —N(C1-6alkyl)(C1-6alkyl) where each C1-6alkyl is substituted with 0-2 R6;
  • R[0119] 5B and R9B each represent from 0 to 2 substituents and are independently selected at each occurrence from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl substituted with 0-2 R6, (C3-8cycloalkyl)C1-4alkyl substituted with 0-2 R6, C2-6alkenyl substituted with 0-2 R6, C2-6alkynyl substituted with 0-2 R6, C1-6alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6, and —N(C1-6alkyl)(C1-6alkyl) where each C1-6alkyl is independently substituted with 0-2 R6, and Y; and any two
  • R[0120] 5 and R5B bound to adjacent atoms may be joined to form a C3-8cycloalkyl group or a heterocycloalkyl group, each of which is optionally substituted by from 1 to 5 substituents independently chosen from cyano, halogen, hydroxy, C1-4alkyl, C1-4alkoxy, —NH(C1-4alkyl), —N(C1-4alkyl)(C1-4alkyl), halo(C1-4)alkyl, and halo(C1-4)alkoxy, wherein the heterocycloalkyl group consists of from 4 to 8 atoms and contains 1, 2, or 3 heteroatoms selected from N, O, and S; and
  • R[0121] 6, R7, R8, X, Y, and Y′ are as defined for Formula IB.
  • Preferred compounds and salts of Formula IX are those wherein A is O or NR[0122] A, wherein RA is hydrogen or methyl.
  • More preferred compounds and salts of Formula IX are those wherein [0123]
  • A is O or NR[0124] A, wherein RA is hydrogen or methyl; and
  • R[0125] 3 and R4 are independently chosen at each occurrence from the group consisting of hydrogen, halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6alkoxy, —NH(C1-6alkyl), and —N(C1-6alkyl)(C1-6alkyl).
  • Other preferred compounds and salts of Formula IX are those wherein: [0126]
  • A is O or NR[0127] A, wherein RA is hydrogen or methyl;
  • R[0128] 3 is hydrogen; and
  • R[0129] 4 is independently chosen at each occurrence from the group consisting of hydrogen, halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6alkoxy, —NH(C1-6alkyl), and —N(C1-6alkyl)(C1-6alkyl).
  • Still more preferred compounds and salts of Formula IX are those wherein [0130]
  • A is O, NR[0131] A, wherein RA is hydrogen or methyl;
  • R[0132] 3 is hydrogen; and
  • R[0133] 4 is independently chosen at each occurrence from hydrogen and C1-6alkyl.
  • Another group of preferred compounds and salts of Formula [0134]
  • IX is the group wherein [0135]
  • A is NR[0136] A, wherein RA is hydrogen or methyl;
  • R[0137] 3 is hydrogen; and
  • R[0138] 4 is independently chosen at each occurrence from hydrogen, halo(C1-3)alkyl, and C1-6alkyl, but more preferably R4 is chosen from hydrogen and C1-4 alkyl.
  • A particular class of compounds of Formula IX is represented by Formula IX-A [0139]
    Figure US20040176443A1-20040909-C00012
  • and the pharmaceutically acceptable salts thereof, wherein: [0140]
  • R[0141] 5, R5B, R9, and R9B are as defined for Formula IX; and
  • R[0142] 4 is independently chosen at each occurrence from hydrogen and C1-4alkyl.
  • Another class of compounds of Formula IX is represented by Formula IX-B: [0143]
    Figure US20040176443A1-20040909-C00013
  • and the pharmaceutically acceptably salts thereof, wherein: [0144]
  • R[0145] 5B and R9B are independently chosen from hydrogen, halogen, cyano, nitro, halo(C1-2)alkyl, halo(C1-2)alkoxy, amino, C1-4alkyl, and C1-2alkoxy; and
  • R[0146] 10 is independently chosen at each occurrence from hydrogen, halogen, and C1-4 alkyl.
  • Preferred compounds and salts of Formula IX-B are those wherein R[0147] 9 is selected from the group consisting of halogen, cyano, —N(SO2CH3)2, —SO2NH2, halo(C1-3)alkyl, C1-3alkoxy, —NH(C1-3alkyl), and —N(C1-3alkyl)(C1-3alkyl).
  • Other preferred compounds and salts of Formula IX-A and Formula IX-B are those wherein R[0148] 5B and R9B are independently chosen from hydrogen, halogen, cyano, nitro, halo(C1-2)alkyl, halo(C1-2)alkoxy, amino,
  • C[0149] 1-4alkyl, and C1-2alkoxy.
  • Still other preferred compounds and salts of Formula IX-A and Formula IX-B are those wherein: [0150]
  • R[0151] 5B represents 0 or 1 substituents chosen from halogen, cyano, nitro, halo(C1-2)alkyl, halo(C1-2)alkoxy, amine, C1-4alkyl, and C1-2alkoxy; and
  • R[0152] 9B represents 0 or 1 substituents chosen from halogen, cyano, nitro, halo(C1-2)alkyl, and C1-2alkyl, and C1-2alkoxy.
  • The invention is further directed to compounds and salts of Formula IX-A and IX-B, wherein: [0153]
  • R[0154] 9 is selected from the group consisting of halogen, cyano, —N(SO2CH3)2, —SO2NH2, halo(C1-3)alkyl, C1-3alkoxy, —NH(C1-3alkyl), and —N(C1-3alkyl)(C1-3alkyl);
  • R[0155] 5B represents 0 or 1 substituents chosen from halogen, cyano, nitro, halo(C1-2)alkyl, halo(C1-2)alkoxy, amino, C1-4alkyl, and C1-2alkoxy; and
  • R[0156] 9B represents 0 or 1 substituents chosen from halogen, cyano, nitro, halo(C1-2)alkyl, and C1-2alkyl, and C1-2alkoxy.
  • For Formula IX-A, preferred substituents are [0157]
  • R[0158] 5 is selected from the group consisting of bromo, fluoro, iodo, halo(C1-6)alkyl, halo(C3-6)alkoxy, C3-6alkyl substituted with 0-3 R6, C2-6alkenyl substituted with 0-3 R6, Y. —(C═O)Y, —(CH2)Y, and —(CH(CN))Y;
  • R[0159] 9 is selected from the group consisting of halogen, cyano, —N(SO2CH3)2, —SO2NH2, halo(C1-2)alkyl, C1-3alkoxy, —NH(C1-6alkyl), and —N(C1-6alkyl)(C1-6alkyl);
  • R[0160] 5B represents 0 or 1 substituents chosen from halogen, cyano, nitro, halo(C1-2)alkyl, halo(C1-2)alkoxy, amino, C1-4alkyl, and C1-2alkoxy; and
  • R[0161] 9B represents 0 or 1 substituents chosen from halogen, cyano, nitro, halo(C1-2)alkyl, and C1-2alkyl, and C1-2alkoxy.
  • Particularly preferred definitions of R[0162] 6 for compounds and salts of this class are cyano, halogen, hydroxy, C1-4alkyl, C1-4alkoxy, —NH(C1-4alkyl), and —N(C1-4alkyl)(C1-4alkyl) and Y; where
  • Y is independently selected at each occurrence from C[0163] 3-8 cycloalkyl, piperidinyl, piperazinyl, tetrahydropyranyl, dihydropyranyl, morpholinyl, thiomorpholinyl, phenyl, pyridyl, pyrazinyl, pyrimidinyl, thiazolyl, thienyl, and imidazolyl, each of which may be further substituted with one or more substituents independently selected from halogen, oxo, hydroxy, amino, nitro, cyano, C1-4alkyl, C1-4alkoxy, halo(C1-4)alkyl, halo(C1-4)alkoxy, mono- or di(C1-4)alkylamino, and C1-4alkylthio.
  • Particularly preferred definitions of R[0164] 9 and R9B for compounds of Formula IX-A are:
  • R[0165] 9 is cyano, trifluoromethyl, chloro, or iodo; and
  • R[0166] 9B is hydrogen.
  • Particularly preferred definitions of R[0167] 5 for compounds of Formula IX-A are isopropyl, t-butyl, 2-butyl, trifluoromethyl, cyclopentyl, cyclohexyl, and heptafluoropropyl.
  • The invention is particularly directed to compounds and pharmaceutically acceptable salts of Formula IB, Formula II, Formula III, Formula IV, Formula V, Formula VII and Formula IX in which: [0168]
  • R[0169] A, RB, and RB′ are independently selected at each occurrence from hydrogen and C1-6alkyl; for the variables R3, R4, and R5 haloalkyl is halo(C1-6)alkyl, i.e. a haloalkyl group having from 1 to 6 carbon atoms and from 1 to maximum allowable number of halogen substituents on those carbon atoms, haloalkoxy is halo(C1-6)alkoxy; alkyl is C1-6alkyl, alkenyl is C2-6alkenyl; alkynyl is C2-6alkynyl; alkoxy is C1-6alkoxy, —NH(alkyl) is —NH(C1-6alkyl), and —N(alkyl)(alkyl) is —N(C1-6alkyl)(C1-6alkyl),
  • for the variables R[0170] 6, R7, R8, Y and Y′ alkyl is C1-4alkyl, alkoxy (or —O(alkyl) is C1-4alkoxy (or —O(C1-4alkyl)), —NHalkyl (or monoalkylamino) is —NH(C1-4alkyl)(or mono(C1-4alkyl)amino), —N(alkyl)(alkyl) (also dialkylamino) is —N(C1-4alkyl)(C1-4alkyl) (also di(C1-4alkyl) amino), —S(O)nalkyl is —S(O)n(C1-4alkyl), haloalkyl is halo(C1-4)alkyl, haloalkoxy is halo(C1-4)alkoxy, —CO(alkyl) is —CO(C1-4alkyl), —CONH(alkyl) is —CONH(C1-4alkyl), and —CON(alkyl)(alkyl) is —CON(C1-4alkyl1)(C1-4alkyl2); NHC(O)(alkyl) is —NHC(O)(C1-4alkyl), —N(alkyl)C(O)(alkyl) is —N(C1-4alkyl)C(O)(alkyl), —NHS(O)n (alkyl) is —NHS(O)n(C1-4alkyl), —S(O)nNH(C1-4alkyl), —S(O)nN(alkyl)(alkyl) is —S(O)nN(C1-4alkyl)(C1-4alkyl); and alkylthio is C1-4alkylthio.
  • Preferred compounds and salts of Formula V, Formula VI, Formula VII and Formula IX are those wherein R[0171] 3 and R4 are independently selected at each occurrence from the group consisting of hydrogen and C1-6 alkyl. More preferred compounds and salts of Formula V are those wherein R3 is hydrogen and the R4 substituents present on the 3 and 5 positions of the piperazine ring are hydrogen and the R4 substituents on the 2 and 6 position of the piperazine ring are independently hydrogen or C1-4 alkyl. For this discussion thhe piperazine ring is numbered as follows:
    Figure US20040176443A1-20040909-C00014
  • Even more preferred compounds and salts of Formula V are those wherein R[0172] 4 is methyl at the 2 position of the piperazine ring and R3 and R4 are hydrogen at all other positions.
  • The invention particularly includes compounds Formula A-1, Formula B-1, Formula C-1, Formula D-1, Formula E-1, and Formula F-1 [0173]
    Figure US20040176443A1-20040909-C00015
  • and the pharmaceutically acceptable salts of Formula A-1, Formula B-1, Formula C-1, Formula D-1, Formula E-1, and Formula F-1 wherein: [0174]
  • R[0175] 5 and R9 are independently selected from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6alkoxy, —NH(C1-6alkyl, —N(C1-6alkyl)(C1-6alkyl), and C3-8 cycloalkyl; and
  • R[0176] 5B and R9B each represent up to 2 substituents independently selected at each occurrence from hydrogen, halogen, cyano, nitro, halo(C1-2)alkyl, halo(C1-2)alkoxy, hydroxy, amino, C1-3alkyl, C1-3alkoxy, —NH(C1-3alkyl), and —N(C1-6alkyl)(C1-6alkyl).
  • Especially preferred compounds and salts of Formula A-1, Formula B-1, Formula C-1, Formula D-1, Formula E-1, and Formula F-1 are those wherein: [0177]
  • R[0178] 5 is C3-6 alkyl; C3-6 alkoxy; halo(C1-3)alkyl, halo(C1-3)alkoxy, or C3-8 cycloalkyl;
  • R[0179] 9 is chloro or trifluomethyl; and
  • R[0180] 5B and R9B are hydrogen.
  • Representative compounds of the invention are shown in Table I below: [0181]
    TABLE I
    Figure US20040176443A1-20040909-C00016
    Figure US20040176443A1-20040909-C00017
    Figure US20040176443A1-20040909-C00018
    Figure US20040176443A1-20040909-C00019
    Figure US20040176443A1-20040909-C00020
    Figure US20040176443A1-20040909-C00021
  • In one aspect invention relates to diaryl piperazines and related compounds that bind with high affinity to capsaicin receptors, including human capsaicin receptors. Compounds that bind with high affinity for the capsaicin receptor include compounds exhibit K[0182] i values of less than 10 uM, and preferably exhibit Ki values of less than 1 uM, more preferably exhibit Ki values of less than 100 nM, and most preferably exhibit Ki values of less than 10 nM at the capsaicin receptors. This invention also includes diaryl piperazines that bind with high selectivity to capsaicin receptor. Compounds that exhibit high selectivity for the capsaicin receptor exhibit at least 20-fold, and preferably at least 100-fold greater affinity for the capsaicin receptor than for other cell surface receptors (e.g., NPY Y5 receptors, NPY Y1 receptors, GABAA receptors, MCH receptors, Bradykinin receptors, C5a receptors, androgen receptors, and the like).
  • Without wishing to be bound to any particular theory of operation, it is believed that the interaction of the compounds of Formulae I-IX and Formulae A-F with the capsaicin receptor results in the pharmaceutical utility of these compounds. [0183]
  • The invention further comprises methods of treating patients in need of such treatment with an amount of a compound of the invention sufficient to alter the symptoms of a disorder responsive to capsaicin receptor modulation. Thus, as used herein, the term treatement encompases both disease modifying treatment and symptomatic treatment. [0184]
  • The diseases and/or disorders that can also be treated using compounds and compositions according to the invention (which are examples of disorders responsive to capsaicin receptor modulation) include: [0185]
  • Chronic and acute pain conditions, including toothache, postherpetic neuralgia, diabetic neuropathy, postmastectomy pain syndrome, stump pain (and phantom limb pain), reflex sympathetic dystrophy, trigeminal neuralgia, oral neuropathic pain, osteoarthritis, rheumatoid arthritis, fibromyalgia, Guillain-Barre syndrome, meralgia paresthetica, “burning-mouth” syndrome, and pain due to bilateral peripheral neuropathy. Preferred pain conditions for treatment in accordance with the invention are neuropathic pain conditions, including causalgia (reflex sympathetic dystrophy—RSD, secondary to injury of a peripheral nerve; this type of pain is generally considered to be non-responsive or only partially responsive to conventional opioid analgesic regimens), neuritis—including, e.g., sciatic neuritis, peripheral neuritis, polyneuritis, optic neuritis, postfebrile neuritis, migrating neuritis, segmental neuritis, Gombault's neuritis, and neuronitis, and neuralgias—including those mentioned above and, e.g., cervicobrachial neuralgia, cranial neuralgia, geniculate neuralgia, glossopharyngial neuralgia, migranous neuralgia, idiopathic neuralgia, intercostals neuralgia, mammary neuralgia, mandibular joint neuralgia, Morton's neuralgia, nasociliary neuralgia, occipital neuralgia, red neuralgia, Sluder's neuralgia, splenopalatine neuralgia, supraorbital neuralgia and vidian neuralgia. Additional pain conditions that can be treated in accordance with the invention include headache—particularly those involving peripheral nerve activity including, e.g., sinus, cluster (i.e., migranous neuralgia, supra) and some tension and migraine headache conditions—, labor pains, Charcot's pains, gas pains, menstrual pain, root pain, homotopic pain and heterotopic pain—including cancer associated pain, pain (and inflammation) associated with venom exposure, e.g., due to snake bite, spider bite, or insect sting, and traumatic, e.g., post-surgical pain and burn pain. A preferred condition that can be treated in accordance with the invention is pain (as well as broncho-constriction and inflammation) due to exposure (e.g., via ingestion, inhalation, or eye contact) of mucous membranes to capsacin and related irritants such as tear gas, hot peppers, or pepper spray. [0186]
  • Itching conditions, including psoriatic pruritis, itch due to hemodyalisis, aguagenic pruritus, and itching associated with vulvar vestibulitis, contact dermatitis, insect bites and skin allergies. [0187]
  • Urinary incontenience, including detrusor hyperflexia of spinal origin and bladder hypersentivity. [0188]
  • The invention also provides pharmaceutical compositions comprising compounds of the invention, including packaged pharmaceutical compositions for treating disorders responsive to capsaicin receptor modulation. The packaged pharmaceutical compositions include a container holding a therapeutically effective amount of at least one capsaicin receptor modulator as described supra and instructions (e.g., labeling) indicating the contained capsaicin receptor ligand is to be used for treating a disorder responsive to capsaicin receptor modulation in the patient. [0189]
  • The present invention also pertains to methods of inhibiting the binding of vanilloid (capsaicin analog) compounds, such as capsaicin, olvanil and RTX, to capsaicin receptors, which methods involve contacting a compound of the invention with cells expressing capsaicin receptors, wherein the compound is present at a concentration sufficient to inhibit vanilloid binding to capsaicin receptors in vitro. The methods of the invention include inhibiting the binding of vanilloid compounds to capsaicin receptors in vivo, e.g., in a patient given an amount of a compound of Formulae I-IX and Formulae A-F that results in and in vivo concentration in a body fluid sufficient to inhibit the binding of capsaicin compounds to capsaicin receptors in vitro. In one embodiment, such methods are useful in treating the effects of tear gas, hot pepper or pepper spray exposure. The amount of a compound that would be sufficient to inhibit the binding of a vanilloid compound to the capsaicin receptor may be readily determined via a capsaicin receptor binding assay, such as the assay described in Example 10 or by an assay of capsaicin receptor antagonism e.g, as in Example 11. The capsaicin receptors used to determine in vitro binding may be obtained from a variety of sources, for example from preparations of mammalian dorsal root ganglion (DRG) or from cells expressing cloned rat or human capsaicin receptors. [0190]
  • The present invention also pertains to methods for altering the signal-transducing activity, particularly the calcium ion conductance, mediated by capsaicin receptors, said method comprising exposing cells expressing such receptors to a solution comprising a compound of the, wherein the compound is present in the solution at a concentration sufficient to specifically alter the calcium conductance activity in response to capsaicin or RTX in vitro in cells expressing capsaicin receptors, preferred cells for this purpose are those that express high levels of capsaicin receptors (i.e., equal to or greater than the number of capsaicin receptors per cell found in rat DRG cells). This method includes altering the signal-transducing activity of capsaicin receptors in vivo. Preferably such alterations are reductions of calcium flux. The amount of a compound that would be sufficient to alter the signal-transducing activity of capsaicin receptors may be determined in vitro via a capsaicin receptor signal transduction assay, such as the calcium mobilization (conductance, flux) assay described in Example 11. The amount of a compound that would be sufficient to alter the calcium conductance activity in response to capsaicin or RTX of capsaicin receptors may also be determined via an assay of capsaicin receptor mediated calcium conductance, such as an assay wherein the binding of capsaicin to a cell surface capsaicin receptor effects changes in the fluorescence of a calcium sensitive dye or in the expression of a calcium sensitive reporter gene. [0191]
  • The invention further provides: [0192]
  • A method of reducing the calcium conductance of a capsaicin receptor, which method comprises: [0193]
  • contacting a first solution comprising a fixed concentration of a capsaicin receptor agonist and a compound or salt of the invention with a cell expressing the capsaicin receptor, wherein the compound or salt is present in the solution at a concentration sufficient to produce a detectable reduction of the calcium mobilization effects of the capsaicin receptor agonist when tested in an in vitro assay in which cells expressing a capsaicin receptor are contacted with a second solution comprising the fixed concentration of capsaicin receptor agonist and the compound or salt and the same method wherein: the cell expressing the capsaicin receptor is a neuronal cell that is contacted in vivo in an animal, and wherein the first solution is a body fluid of said animal; or the animal is a human patient. [0194]
  • A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound or salt of the invention; and a package comprising the pharmaceutical composition in a container and further comprising indicia comprising instructions for using the composition to either alleviate pain; or to treat a patient suffering from urinary incontinence or to alleviate symptoms of exposure to capsaicin or tear gas. [0195]
  • A compound or salt of the invention wherein, in an in vitro assay of capsaicin receptor antagonism, the compound or salt exhibits capsaicin receptor antagonist activity, but in an in vitro assay of capsaicin receptor agonism the compound does not exhibit detectable agonist activity. [0196]
  • A compound or salt of the invention wherein a dose of the compound or salt sufficient to provide analgesia in an animal model for determining pain relief does not produce sedation in an animal model assay of sedation. [0197]
  • A method of treating a mammal suffering from at least one symptom selected from the group consisting of symptoms of exposure to capsaicin, symptoms of burns or irritation due to exposure to heat, symptoms of burns or irritation due to exposure to light, symptoms of burns or irritation due to exposure to tear gas, and symptoms of burns or irritation due to exposure to acid, the method comprising administering to the mammal a therapeutic dose of a compound that: [0198]
  • a) is a high potency capsaicin receptor antagonist in an in vitro assay of capsaicin receptor antagonism, [0199]
  • b) exhibits no detectable agonist activity in an in vitro assay of capsaicin receptor agonism, [0200]
  • c) is not a capsaicin analog, and [0201]
  • d) when administered to an animal in an animal model assay of sedation, at five times the minimum dosage needed to provide analgesia in an animal model for determining pain relief, does not cause sedation, [0202]
  • wherein the therapeutic dose contains an amount of the compound that is effective to reduce severity of at least one of the at least one symptom and preferably wherein the compound is a compound of the invention. [0203]
  • A method of treating a mammal suffering from neuropathic pain, the method comprising administering to the mammal a therapeutic dose of a compound that is a capsaicin receptor antagonist, and in certain embodiments, wherein the compound is a compound of the invention. [0204]
  • A method of treating a mammal suffering from peripheral-nerve-mediated pain, e.g., neuropathic pain, the method comprising administering to the mammal a therapeutic dose of a compound that is a capsaicin receptor antagonist, wherein the compound: [0205]
  • a) is a high potency capsaicin receptor antagonist in an in vitro assay of capsaicin receptor antagonism, [0206]
  • b) exhibits no detectable agonist activity in an in vitro assay of capsaicin receptor agonism, [0207]
  • c) is not a capsaicin analog, and [0208]
  • d) when administered to an animal in an animal model assay of sedation, at five times the minimum dosage needed to provide analgesia in an animal model for determining pain relief, does not cause sedation, [0209]
  • wherein the therapeutic dose contains an amount of the compound that is effective to reduce the peripheral-nerve-mediated pain, and preferably wherein the pain is neuropathic pain and the compound is a compound of the invention, and preferably wherein the pain is associated with a condition selected from the group consisting of postmastectomy pain syndrome, stump pain, phantom limb pain, oral neuropathic pain, Charcot's pain, toothache, postherpetic neuralgia, diabetic neuropathy, reflex sympathetic dystrophy, trigeminal neuralgia, osteoarthritis, rheumatoid arthritis, fibromyalgia, Guillain-Barre syndrome, meralgia paresthetica, burning-mouth syndrome, bilateral peripheral neuropathy, causalgia, sciatic neuritis, peripheral neuritis, polyneuritis, optic neuritis, postfebrile neuritis, migrating neuritis, segmental neuritis, Gombault's neuritis, neuronitis, cervicobrachial neuralgia, cranial neuralgia, geniculate neuralgia, glossopharyngial neuralgia, migranous neuralgia, idiopathic neuralgia, intercostals neuralgia, mammary neuralgia, mandibular joint neuralgia, Morton's neuralgia, nasociliary neuralgia, occipital neuralgia, red neuralgia, Sluder's neuralgia, splenopalatine neuralgia, supraorbital neuralgia, vidian neuralgia, sinus headache, tension headache, labor, childbirth, intestinal gas, menstruation, cancer, and trauma. [0210]
  • A compound of the invention, wherein the compound is not addictive. [0211]
  • The capsaicin receptor ligands provided by this invention and labeled derivatives thereof are also useful as standards and reagents for determining the ability of a compound to bind to the capsaicin receptor and to act as an agonist, antagonist, inverse agonist, mixed agonist/antagonist or the like. [0212]
  • More particularly compounds of the invention may be used for demonstrating the presence of VR1 receptors or other capsaicin receptors in cell or tissue samples. This may be done by preparing a plurality of matched cell or tissue samples, at least one of which is prepared as an experimental sample and at least one of which is prepared as a control sample. The experimental sample is prepared by contacting (under conditions that permit binding of capsaicin or RTX to capsaicin receptors within cell and tissue samples) at least one of the matched cell or tissue samples that has not previously been contacted with any compound or salt of the invention with an experimental solution comprising the detectably-labeled preparation of the selected compound or salt at a first measured molar concentration. The control sample is prepared by in the same manner as the experimental sample and is incubated in a solution that contains the same ingredients as the experimental solution but that also contains an unlabelled preparation of the same compound or salt of the invention at a molar concentration that is greater than the first measured molar concentration. [0213]
  • The experimental and control samples are then washed (using the same wash conditions) to remove unbound detectably-labeled compound. The amount of detectably-labeled compound remaining bound to each sample is then measured and the amount of detectably-labeled compound in the experimental and control samples is compared. A comparison that indicates the detection of a greater amount of detectable label in the at least one washed experimental sample than is detected in any of the at least one washed control samples demonstrates the presence of capsaicin receptors in that experimental sample. [0214]
  • The detectably-labeled compound used in this procedure may be labeled with any detectable label, such as a radioactive label, a biological tag such as biotin (which can be detected by binding to detectably-labeled avidin), an enzyme (e.g., alkaline phosphatase, beta galactosidase, or a like enzyme that can be detected its activity in a calorimetric, luminescent, or like assay) or a directly or indirectly luminescent label. When tissue sections are used in this procedure and the detectably-labeled compound is radiolabeled, the bound, labeled compound may be detected autoradiographically to generate an autoradiogram. When autoradiography is used, the amount of detectable label in an experimental or control sample may be measured by viewing the autoradiograms and comparing the exposure density of matched regions of the autoradiograms. [0215]
  • Labeled derivatives the capsaicin receptor ligands provided by this invention are also useful as radiotracers for positron emission tomography (PET) imaging or for single photon emission computerized tomography (SPECT) to characterize and localize capsaicin receptors in vivo. [0216]
  • Definitions [0217]
  • The compounds herein described may have one or more asymmetric centers or planes. Compounds of the present invention containing an asymmetrically substituted atom may be isolated in optically active or racemic forms. It is well known in the art how to prepare optically active forms, such as by resolution of racemic forms (racemates), by asymmetric synthesis, or by synthesis from optically active starting materials. Resolution of the racemates can be accomplished, for example, by conventional methods such as crystallization in the presence of a resolving agent, or chromatography, using, for example a chiral HPLC column. Many geometric isomers of olefins, C═N double bonds, and the like can also be present in the compounds described herein, and all such stable isomers are contemplated in the present invention. Cis and trans geometric isomers of the compounds of the present invention are described and may be isolated as a mixture of isomers or as separated isomeric forms. All chiral (enantiomeric and diastereomeric), and racemic forms, as well as all geometric isomeric forms of a structure are intended, unless the specific stereochemistry of isomeric form is specifically indicated. [0218]
  • The term “substituted,” as used herein, means that any one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valence is not exceeded, and that the substitution results in a stable compound. The present invention is intended to include all isotopes of atoms occurring in the present compounds. Isotopes include those atoms having the same atomic number but different mass numbers, By way of general example and without limitation, isotopes of hydrogen include tritium and deuterium. Isotopes of carbon include [0219] 11C, 13C, and 14C.
  • When any variable occurs more than one time in any constituent or formula for a compound, its definition at each occurrence is independent of its definition at every other occurrence. Thus, for example, if a group is shown to be substituted with 0-2 R*, then said group may optionally be substituted with up to two R* groups and R* at each occurrence is selected independently from the definition of R*. Also, combinations of substituents and/or variables are permissible only if such combinations result in stable compounds. [0220]
  • As indicated above, various substituents of the various formulae are “optionally substituted”, including Ar[0221] 1, Ar2, R3 and R4 of Formulae I-IX and Formulae A-F, and such substituents as recited in the sub-formulae such as Formula Ia and the like. When substituted, those substituents (e.g., C1-6 alkyl, n, Ar1, Ar2, R1, R2, R3, and R4) may be substituted by other than hydrogen at one or more available positions, typically 1 to 3 or 4 positions, by one or more groups, such as, halogen such as fluoro, chloro, bromo and iodo; cyano; hydroxyl; nitro; azido; alkanoyl such as a C1-6 alkanoyl group such as acyl and the like; carboxamido; alkyl groups including those groups having 1 to about 12 carbon atoms, or 1, 2, 3, 4, 5, or 6 carbon atoms; alkenyl and alkynyl groups including groups having one or more unsaturated linkages and from 2 to about 12 carbon, or 2, 3, 4, 5 or 6 carbon atoms; alkoxy groups having those having one or more oxygen linkages and from 1 to about 12 carbon atoms, or 1, 2, 3, 4, 5 or 6 carbon atoms; aryloxy such as phenoxy; alkylthio groups including those moieties having one or more thioether linkages and from 1 to about 12 carbon atoms, or 1, 2, 3, 4, 5 or 6 carbon atoms; alkylsulfinyl groups including those moieties having one or more sulfinyl linkages and from 1 to about 12 carbon atoms, or 1, 2, 3, 4, 5, or 6 carbon atoms, alkylsulfonyl groups including those moieties having one or more sulfonyl linkages and from 1 to about 12 carbon atoms, or 1, 2, 3, 4, 5, or 6 carbon atoms; aminoalkyl groups such as groups having one or more N atoms and from 1 to about 12 carbon atoms, or 1, 2, 3, 4, 5 or 6 carbon atoms; carbocyclic aryl having 6 or more carbons, particularly phenyl (e.g. an Ar group being a substituted or unsubstituted biphenyl moiety); arylalkyl having 1 to 3 separate or fused rings and from 6 to about 18 carbon ring atoms, with benzyl being a preferred group; arylalkoxy having 1 to 3 separate or fused rings and from 6 to about 18 carbon ring atoms, with O-benzyl being a preferred group; or a heteroaromatic or heteroalicyclic group having 1 to 3 separate or fused rings with 3 to about 8 members per ring and one or more N, O or S atoms, e.g. coumarinyl, quinolinyl, pyridyl, pyrazinyl, pyrimidyl, furyl, pyrrolyl, thienyl, thiazolyl, oxazolyl, imidazolyl, indolyl, benzofuranyl, benzothiazolyl, tetrahydrofuranyl, tetrahydropyranyl, piperidinyl, morpholino and pyrrolidinyl.
  • As used herein, “alkyl” is intended to include both branched, straight-chain, and cyclic alkyl groups, having the specified number of carbon atoms that may contain one or more double or triple bonds. “Lower alkyl” denotes an alkyl group having from 1 to about 6 carbon atoms. Examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl, t-butyl, n-pentyl, and s-pentyl. Preferred alkyl groups are C[0222] 1-C6 alkyl groups. Especially preferred alkyl groups are methyl, ethyl, propyl, butyl, 3-pentyl.
  • As used herein, “alkoxy”, “C[0223] 1-C6 alkoxy”, or “lower alkoxy” in the present invention is meant an alkyl group attached through an oxygen bridge such as, for example, methoxy, ethoxy, propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, pentoxy, 2-pentyl, isopentoxy, neopentoxy, hexoxy, 2-hexoxy, 3-hexoxy, and 3-methylpentoxy. “Lower alkoxy” denotes an alkyl group having from 1 to about 6 carbon atoms.
  • “Haloalkyl” is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms, substituted with 1 or more halogen (for example —C[0224] vFw where v=1 to 3 and w=1 to (2v+1). Examples of haloalkyl include, but are not limited to, trifluoromethyl, trichloromethyl, pentafluoroethyl, heptafluoropropyl, and pentachloroethyl.
  • As used herein, “carbocycle” or “carbocyclic ring” is intended to mean any stable 3- to 7-membered monocyclic or bicyclic or 7- to 13-membered bicyclic or tricyclic moeity, any of which may be saturated, partially unsaturated, or aromatic. Examples of such carbocycles include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, adamantyl, cyclooctyl, [3.3.0]bicyclooctane, [4.3.0]bicyclononane, [4.4.0]bicyclodecane, [2.2.2]bicyclooctane, fluorenyl, phenyl, naphthyl, indanyl, adamantyl, and tetrahydronaphthyl. [0225]
  • As used herein, the term “carbocyclic aryl” indicates aromatic groups containing only carbon. Such aromatic groups may be further substituted. [0226]
  • As used herein, the term “heterocyclic ring” is intended to mean a stable 5- to 7-membered monocyclic or bicyclic or 7- to 10-membered bicyclic heterocyclic ring which is saturated, partially unsaturated, or unsaturated (aromatic), and which consists of carbon atoms and from 1 to 4 heteroatoms independently selected from the group consisting of N, O and S and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring. The nitrogen and sulfur heteroatoms may optionally be oxidized. The heterocyclic ring may be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure. The heterocyclic rings described herein may be substituted on carbon or on a nitrogen atom if the resulting compound is stable. A nitrogen atom in the heterocycle may optionally be quaternized. It is preferred that when the total number of S and O atoms in the heterocycle exceeds 1, then these heteroatoms are not adjacent to one another. It is preferred that the total number of S and O atoms in the heterocycle is not more than 1. As used herein, the term “heteroaryl” is intended to mean a stable 5- to 7-membered monocyclic or bicyclic or 7- to 10-membered bicyclic heterocyclic aromatic ring which consists of carbon atoms and from 1 to 4 heteroatoms independently selected from the group consisting of N, O and S. It is preferred that the total number of S and O atoms in the aromatic heterocycle is not more than 1. The term “heterocycloalkyl” is intended to mean a stable 5- to 7-membered monocyclic or bicyclic or 7- to 10-membered bicyclic heterocyclic saturated ring which consists of carbon atoms and from 1 to 4 heteroatoms independently selected from the group consisting of N, O and S. Examples of heterocycles include, but are not limited to, acridinyl, azocinyl, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, NH-carbazolyl, carbolinyl, chromanyl, chromenyl, cinnolinyl, decahydroquinolinyl, 2H,6H-1,5,2-dithiazinyl, dihydrofuro[2,3-b]tetrahydrofuran, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, 1H-indazolyl, indolenyl, indolinyl, indolizinyl, indolyl, 3H-indolyl, isobenzofuranyl, isochromanyl, isoindazolyl, isoindolinyl, isoindolyl, isoquinolinyl, isothiazolyl, isoxazolyl, morpholinyl, naphthyridinyl, octahydroisoquinolinyl, oxadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl; 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, oxazolidinyl, oxazolyl, oxazolidinyl, pyrimidinyl, phenanthridinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, phenoxathiinyl, phenoxazinyl, phthalazinyl, piperazinyl, piperidinyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridooxazole, pyridoimidazole, pyridothiazole, pyridinyl, pyridyl, pyrimidinyl, pyrrolidinyl, pyrrolinyl, 2H-pyrrolyl, pyrrolyl, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl, quinuclidinyl, tetrahydrofuranyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, 6H-1,2,5-thiadiazinyl, 1,2,3-thiadiazolyl, 1,2,4-thiadiazolyl, 1,2,5-thiadiazolyl, 1,3,4thiadiazolyl, thianthrenyl, thiazolyl, thienyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl, thiophenyl, triazinyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,2,5-triazolyl, 1,3,4-triazolyl, and xanthenyl. [0227]
  • Preferred heterocycles include, but are not limited to, pyrrolyl, furanyl, thienyl, pyrazolyl, imidazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, triazolyl, tetrazolyl, pyridyl, pyrimidyl, pyrazinyl, benzimidazolyl, indolyl, isoindolyl, benzofuranyl, isobenzofuranyl, benzo[b]thiophenyl, benz[d]isoxazolyl, quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, or quinoxalinyl. Also included are fused ring and spiro compounds containing, for example, the above heterocycles. [0228]
  • The term “bicyclic oxygen-containing group” is meant to encompass a particular type of heteroaryl group of the formula: [0229]
    Figure US20040176443A1-20040909-C00022
  • where L indicates the point of attachment of the group to the structure of Formulae I-IX and Formulae A-F. The heterocyclic oxygen-containing ring has a total of from 5 to 7 members, and is saturated or unsaturated. Either ring of the bicyclic oxygen-containing group may be further substituted. [0230]
  • Examples of bicyclic oxygen-containing groups include any or all of the following structures: [0231]
    Figure US20040176443A1-20040909-C00023
  • Pharmaceutical Preparations [0232]
  • Those skilled in the art will recognize various synthetic methodologies that may be employed to prepare non-toxic pharmaceutically acceptable prodrugs of the compounds encompassed by Formulae I-IX and Formulae A-F, which prodrugs are encompassed by the present invention. “Prodrugs” are intended to include any compounds that become compounds of Formulae I-IX and Formulae A-F when administered to a mammalian subject, e.g., upon metabolic processing of the prodrug. Examples of prodrugs include, but are not limited to, acetate, formate and benzoate and like derivatives of functional groups (such as alcohol or amine groups) in the compounds of Formulae I-IX and Formulae A-F. Those skilled in the art will recognize a wide variety of non-toxic pharmaceutically acceptable solvents that may be used to prepare solvates of the compounds of the invention, such as water, ethanol, mineral oil, vegetable oil, and dimethylsulfoxide. [0233]
  • The compounds of general Formulae I-IX and general Formulae A-F may be administered orally, topically, parenterally, e.g., by inhalation or spray or rectally in dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles. Oral administration in the form of a pill, capsule, elixir, syrup, lozenge, troche, or the like is particularly preferred. The term parenteral as used herein includes subcutaneous injections, intradermal, intravascular (e.g., intravenous), intramuscular, spinal, intrathecal injection or like injection or infusion techniques. In addition, there is provided a pharmaceutical formulation comprising a compound of general Formulae I-IX and general Formulae A-F and a pharmaceutically acceptable carrier. One or more compounds of general Formulae I-IX and general Formulae A-F may be present in association with one or more non-toxic pharmaceutically acceptable carriers and/or diluents and/or adjuvants and if desired other active ingredients. The pharmaceutical compositions containing compounds of general Formulae I-IX and general Formulae A-F may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs. [0234]
  • Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients that are suitable for the manufacture of tablets. These excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc. The tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monosterate or glyceryl distearate may be employed. [0235]
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil. [0236]
  • Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydropropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monooleate. The aqueous suspensions may also contain one or more preservatives, for example ethyl, or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose or saccharin. [0237]
  • Oily suspensions may be formulated by suspending the active ingredients in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavoring agents may be added to provide palatable oral preparations. These compositions may be preserved by the addition of an antioxidant such as ascorbic acid. [0238]
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example sweetening, flavoring and coloring agents, may also be present. [0239]
  • Pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil, for example olive oil or arachis oil, or a mineral oil, for example liquid paraffin or mixtures of these. Suitable emulsifying agents may be naturally-occurring gums, for example gum acacia or gum tragacanth, naturally-occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitil, anhydrides, for example sorbitan monoleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monoleate. The emulsions may also contain sweetening and flavoring agents. [0240]
  • Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative and flavoring and coloring agents. The pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be sterile injectable solution or suspension in a non-toxic parentally acceptable diluent or solvent, for example as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables. [0241]
  • The compounds of general Formulae I-IX and general Formulae A-F may also be administered in the form of suppositories, e.g., for rectal or vaginal administration of the drug. These compositions can be prepared by mixing the drug with a suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials are cocoa butter and polyethylene glycols. [0242]
  • Compounds of general Formulae I-IX and general Formulae A-F may be administered parenterally in a sterile medium. The drug, depending on the vehicle and concentration used, can either be suspended or dissolved in the vehicle. Advantageously, adjuvants such as local anesthetics, preservatives and buffering agents can be dissolved in the vehicle where desirable. [0243]
  • Typical subjects to which compounds of the invention may be administered will be mammals, particularly primates, especially humans. For veterinary applications, a wide variety of subjects will be suitable, e.g. livestock such as cattle, sheep, goats, cows, swine and the like; poultry such as chickens, ducks, geese, turkeys, and the like; and other domesticated animals particularly pets (companion animals) such as dogs and cats. For diagnostic or research applications, a wide variety of mammals will be suitable subjects including rodents (e.g. mice, rats, hamsters), rabbits, primates, and swine such as inbred pigs and the like. [0244]
  • For administration to non-human animals, the composition may also be added to the animal feed or drinking water. It will be convenient to formulate these animal feed and drinking water compositions so that the animal takes in an appropriate quantity of the composition along with its diet. It will also be convenient to present the composition as a premix for addition to the feed or drinking water. [0245]
  • For systemic (as opposed to local or topical) administration, dosage levels of the order of from about 0.01 mg to about 140 mg per kilogram of body weight per day are useful in the treatment of pain, urinary incontinence, or other of the above-indicated conditions (about 0.05 mg to about 7 g per human patient per day). Preferred systemic doses for preferred high potency compounds of Formulae I-IX and Formulae A-F range from about 0.01 mg to about 50 mg per kilogram of body weight per subject per day, with oral doses generally being about 5-20 fold higher than intravenous doses. The most highly preferred compounds of the invention are orally active (e.g., provide a reduction of pain or a reduction of frequency of urinary incontinence) at doses ranging from 0.05 to 40 mg per kilogram of body weight per subject per day. [0246]
  • The amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. Dosage unit forms will generally contain between from about 1 mg to about 500 mg of an active ingredient. [0247]
  • Frequency of dosage may also vary depending on the compound used and the particular disease treated. However, for treatment of most disorders, a dosage regimen (frequency of administration) of 4 times daily or less is preferred. For the treatment of chronic pain or urinary incontinence a dosage regimen of 2 times daily is more preferred and a frequency of administration of once a day is particularly preferred. For the treatment of acute pain a single dose that rapidly reaches effective concentrations is desirable. [0248]
  • It will be understood, however, that the specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, and rate of excretion, drug combination and the nature and severity of the particular disease or condition undergoing treatment. [0249]
  • Preferred compounds of the invention will have certain desirable pharmacological properties. For systemic administration such properties include, but are not limited to high oral bioavailability, such that the preferred oral dosages and dosage forms discussed above can provide therapeutically effective levels of the compound in vivo, low serum protein binding and low first pass hepatic metabolism. For all types of administration low toxicity, and desirable in vitro and in vivo half-lifes are desired. While penetration of the blood brain barrier for compounds used to treat CNS disorders is necessary, low brain levels of compounds used to treat peripheral disorders (such as urinary incontinence, or chronic or acute pain that does not originate from the CNS) are often preferred. [0250]
  • Laboratory assays may be used to predict these desirable pharmacological properties. The discussion that follows is supplemented by the detailed protocols of Example 16, infra. Assays used to predict bioavailability include transport across human intestinal cell monolayers, including. Caco-2 cell monolayers. Toxicity to cultured hepatocyctes may be used to predict compound toxicity, with non-toxic compounds being preferred. Penetration of the blood brain barrier of a compound in humans may be predicted from the brain levels of the compound in laboratory animals given the compound, e.g., intravenously. [0251]
  • Percentage of serum protein binding may be predicted from albumin binding assays. Examples of such assays are described in a review by Oravcová, et al. (Journal of Chromatography B (1996) volume 677, pages 1-27). Preferred compounds exhibit reversible serum protein binding. Preferably this binding is less than 99%, more preferably less than 95%, even more preferably less than 90%, and most preferably less than 80%. [0252]
  • Frequency of administration is generally inversely proportional to the in vivo half-life of a compound. In vivo half-lives of compounds may be predicted from the results of assays, e.g., in vitro assays of microsomal half-life as described by Kuhnz and Gieschen ([0253] Drug Metabolism and Disposition, 1998, volume 26, pages 1120-1127). Preferred half-lives are those allowing for a preferred frequency of administration.
  • Preparation of Compounds [0254]
  • The compounds of the present invention can be prepared in a number of ways well known to those skilled in the art of organic synthesis. By way of example, compounds of the present invention can be synthesized using the methods described below, together with synthetic methods known in the art of synthetic organic chemistry, or variations thereon as appreciated by those skilled in the art. Preferred methods include but are not limited to those methods described below. Compounds of the present invention i.e. urea or thiourea derivatives (VI) can be synthesized by following the steps outlined in Scheme 1. [0255]
    Figure US20040176443A1-20040909-C00024
  • Intermediate III can be obtained by treating I with II in the presence of a base (eg: K[0256] 2CO3, Cs2CO3, NR1R2R3, NaOR, KOR) in an inert solvent such as N,N-dialkylformamide, N,N-dialkylacetamide, dialkylethers, cyclic ethers, DMSO, N-methyl-2-pyrrolidinone at temperatures ranging from −78° C. to 200° C. Isocyanates or isothiocyanates of V can be obtained by treating compound of IV with phosgene, thiophosgene, carbonyldiimidazole in an inert solvent such as benzene, toluene at temperatures ranging from −78° C. to 200° C. The compound of present invention VI can be obtained by treating intermediates III with V in an organic solvent at temperatures −78° C. to 200° C. Alternatively compound of VI can be prepared by treatment of intermediate VII with III in the presence of base such as triethylamine in an inert solvent such as chloroform at temperatures ranging from 78° C. to 200° C.
  • Carbamates or thiocarbamates (X) of the present invention can be synthesized by following the steps outlined in Scheme 2. [0257]
    Figure US20040176443A1-20040909-C00025
  • Intermediate III can be converted to VIII (A=O, S, L=halogen, imidazole) upon treatment with phosgene, thiophosgene or carbonyldiimidazoles. Compound of product X can be obtained by treatment with phenols (IX) with compound VIII in the presence of a base in an inert solvent at temperatures ranging from −78° C. to 200° C. [0258]
  • Those having skill in the art will recognize that the starting materials may be varied and additional steps employed to produce compounds encompassed by the present inventions, as demonstrated by the following examples. Unless otherwise specified all reagents and solvent are of standard commercial grade and are used without further purification. [0259]
  • The disclosures in this application of all articles and references, including patents, are incorporated herein by reference. [0260]
  • The invention is illustrated further by the following examples, which are not to be construed as limiting the invention in scope or spirit to the specific procedures described in them. Those having skill in the art will recognize that the starting materials may be varied and additional steps employed to produce compounds encompassed by the present inventions, as demonstrated by the following examples. In some cases, protection of certain reactive functionalities may be necessary to achieve some of the above transformations. In general, such need for protecting groups, as well as the conditions necessary to attach and remove such groups, will be apparent to those skilled in the art of organic synthesis. [0261]
  • EXAMPLES Example 1 (R)-4-(3-Chloro-pyridin-2-yl)-2-methyl-piperazine-1-carboxylic acid (4-sec-butyl-phenyl)-amide
  • Part A: Synthesis of (R)-1-(3-Chloro-pyridin-2-yl)-3-methylpiperazine: [0262]
    Figure US20040176443A1-20040909-C00026
  • Dissolve 2,3-dichloropyridine (8.5 g, 0.057 moles) and (R)-(−)-2-methylpiperazine (5.75 g, 0.057 moles) in N,N-dimethylacetamide (125.0 mL) under nitrogen atmosphere. Add anhydrous powdered K[0263] 2CO3 (23.75 g, 0.172 moles) to this mixture and stir at 135-140° C. for 48 h. New spot noticed in TLC (5% MeOH/CHCl3/1% NEt3) along with absence of starting materials. Cool the reaction mixture to room temperature, dilute with water (400 mL), extract with EtOAc (3×200 mL) and wash the combined organic extract with brine (2×150 mL). Dry over MgSO4, concentrate under vacuum to afford crude product (20.0 g) as orange yellow liquid. Distil the crude under high vacuum to afford pyridylpiperazine derivative as yellow viscous oil (10 g, bp 112-115° C./0.1 torr). NMR (CDCl3): δ 1.1-1.12 (d, 3H, J=1.6 Hz), 2.50-2.53 (t, 1H), 2.83-2.87 (m, 1H), 3.06-3.08 (m, 3H), 3.67-3.75 (m, 2H), 6.80-6.82(dd, 1H), 7.56-7.58 (dd, 1H), 8.17-8.18 (dd, 1H).
  • Part B: Synthesis of (4-sec-Butyl-phenyl)-carbamic acid phenyl ester: [0264]
    Figure US20040176443A1-20040909-C00027
  • Dissolve 4-isobutylaniline (4.5 g, 0.03 moles) in pyridine (30 mL) under nitrogen at room temperature. Add drop wise phenyl chloroformate (3.75 mL, 0.03 moles) to the reaction mixture at room temperature. Stir the mixture for 3 days and new spot noticed in TLC (30% EtOAc/hexane). Evaporate the reaction mixture under vacuo, partition between EtOAc and water (200 mL), wash several times with brine, dry (MgSO[0265] 4) and concentrate in vacuo. Purify the crude by flash column chromatography on a silica gel using (10% EtOAc/hexanes) to afford white solid.
  • Part C: Title Compound: [0266]
    Figure US20040176443A1-20040909-C00028
  • Dissolve Part A material of Example 1 (212 mg, 1.0 mmole) with Part B material of Example 1 (269 mg, 1.0 mmole) in CHCl[0267] 3 (10 mL) under nitrogen at room temperature. Add triethylamine (202 mg, 2.0 mmol) to the mixture and reflux for 4 hours. Cool the reaction mixture to room temperature, wash with 2N aq. NaOH, water and dry (MgSO4). Evaporate the dried extract in vacuo and purify by flash column chromatography on a silica gel using CHCl3 to afford white solid.
  • NMR (CDCl[0268] 3): δ 0.78-0.83. (t, 3H), 1.19-1.22 (d, 3H, J=2.2 Hz), 1.43-1.45 (d, 3H, J=2.3 Hz), 1.51-1.61 (m, 2H), 2.51-2.58 (m, 1H), 2.92-3.05 (m, 2H), 3.41-3.50 (m, 1H), 3.73-3.94 (m, 3H), 4.35 (m, 1H), 6.33 (bs, 1H), 6.86-6.90 (dd, 1H), 7.09-7.13 (m, 2H), 7.26-7.30 (m, 2H), 7.60-7.63 (dd, 1H), 8.18-8.20 (dd, 1H).
  • Example 2 (R)-(−)-4-(3-Chloro-pyridin-2-yl)-2-methyl-piperazine-1-carboxylic acid (4-trifluoromethyl-phenyl)-amide
  • [0269]
    Figure US20040176443A1-20040909-C00029
  • Dissolve Part A material of example 1 (0.2756 g, 1.3 mmoles) in toluene (1.5 mL) under nitrogen at room temperature. Add drop wise 4-trifluoromethylphenyl isocyanate (0.2431 g, 1.3 mmoles) dissolved in toluene (50 mL) to the mixture over a period of 30 mins and stir at room temperature for 3 hours. Evaporate the solvent from reaction mixture under vacuum to afford colorless oil. Crystallize the oil from 1:1 Et[0270] 2O/hexane (2.0 mL) to afford white solid.
  • NMR (CDCl[0271] 3): δ 1.45-1.47 (d, 3H, J=1.7 Hz), 2.97-3.01 (t, 1H), 3.06-3.10 (m, 1H), 3.47-3.50 (m, 1H), 3.75-3.85 (m, 2H), 3.92-3.95 (m, 1H), 4.37-4.38 (m, 1H), 6.59 (bs, 1H), 6.88-6.91 (dd, 1H), 7.52-7.56 (m, 4H), 7.61-7.63 (dd, 1H), 8.19-8.21 (dd, 1H).
  • Mass spectrum (ESI): 399.3 (M+H). [0272]
  • Analysis calcd. for C[0273] 18H18ClF3N4O: C, 54.21; H, 4.55; Cl, 8.89; F, 14.29; N, 14.05. Found: C, 54.47; H, 4.36; Cl, 8.50; F, 14.99; N, 13.94
  • Example 3 (R)-3-Chloro-pyridin-2-yl)-2-methyl-piperazine-1-carboxylic acid 4-tert-butyl-phenyl ester
  • Part A: Synthesis of (R)-4-(3-Chloro-pyridin-2-yl)-2-methylpiperazine-1-carbonyl chloride: [0274]
    Figure US20040176443A1-20040909-C00030
  • Dissolve Part A material of Example 1 (1.06 g, 5.0 mmole)) in CH[0275] 2Cl2 (50 mL) and saturated NaHCO3 (50 mL) under nitrogen at room temperature. Add drop wise 20% COCl2 in toluene (5.0 mL) at room temperature and stir overnight. Separate the organic layer, extract the aq. layer with CH2Cl2 (2×15 mL) and dry (MgSO4). Evaporate the organic layer under vacuo to afford yellow oil.
  • Part B: Title Compound: [0276]
    Figure US20040176443A1-20040909-C00031
  • Dissolve Part A material of Example 3 (136 g, 0.5 mmole)) in pyridine (2.0 mL) under nitrogen at room temperature. Add 4-tert.butylphenol to the reaction mixture at room temperature and stir overnight. Evaporate the reaction mixture under vacuo, partition between water/CH[0277] 2Cl2 (20 mL) and dry (MgSO4). Evaporate the organic layer under vacuo and purify by flash column chromatography on silica gel using 15% EtOAc/hexane to afford colorless oil.
  • NMR (CDCl[0278] 3): δ 1.28-1.31 (3S, 9H), 1.35-1.48 (m, 3H), 2.96-3.11 (m, 2H), 3.49 (m, 1H), 3.72-3.80 (m, 2H), 4.13-4.24 (m, 1H), 4.55-4.60 (m, 1H), 6.74-6.92 (m, 2H), 7.04-7.07 (m, 1H), 7.23-7.26 (m, 1H), 7.36-7.38 (m, 1H), 7.61-7.64 (m, 1H), 8.19-8.22 (m, 1H).
  • Mass spectrum (ESI): 388.2 (M+H). [0279]
  • Example 4 2-Methyl-4-(3-trifluoromethyl-pyridin-2-yl)-piperazine-1-carboxylic acid [4-(2,2,2-trifluoro-1-methyl-ethyl)-phenyl]-amide
  • Part A: Synthesis of 3-Methyl-1-(3-trifluoromethyl-pyridin-2-yl)-piperazine [0280]
    Figure US20040176443A1-20040909-C00032
  • Dissolve 2-chloro-3-trifluoromethylpyridine (5.4 g, 0.03 moles) and (R)-(−)-2-methylpiperazine (3.0 g, 0.03 moles) in N,N-dimethylacetamide (100.0 mL) under nitrogen atmosphere. Add anhydrous powdered K[0281] 2CO3 (12.4 g, 0.09 moles) to this mixture and stir at 135-140° C. for 24 h. New spot noticed in TLC (5% MeOH/CHCl3/1% NEt3) along with absence of starting materials. Cool the reaction mixture to room temperature, dilute with water (300 mL), extract with EtOAc (3×200 mL) and wash the combined organic extract with brine (2×150 mL). Dry over MgSO4, concentrate under vacuum to afford crude product as orange yellow liquid. Purify by flash column chromatography using 1% MeOH/CHCl3 to afford yellow viscous oil.
  • Part B: Synthesis of 4-(1-Trifluoromethyl-vinyl)-phenylamine [0282]
    Figure US20040176443A1-20040909-C00033
  • Dissolve 4-(4,4,5,5-tetramethyl-1,2,3-dioxaborolan-2-yl)aniline (2.19 g, 0.01 moles) and 2-Bromo-3,3,3-trifluoro-propene (2.61 g, 0.015 moles) in 1:1 THF/1,2-dimethoxyethane (30 mL) and cooled in an ice bath under nitrogen atmosphere. Add PdCl[0283] 2[(PPh3)2] (210 mg, 3 mol %) and AsPh3 (459 mg, 15 mol %) to the reaction mixture followed by dropwise addition of 2.0 N Aq. NaOH (20 mL). Stirred the resultant mixture at room temp for 1 h followed by 70° C. for 15 h. Add additional 1.5 eq. of 2-Bromo-3,3,3-trifluoro-propene (2.61 g) to the reaction mixture and continued at 70° C. for 6 h. Evaporate the reaction mixture under vacuo, dissolve the residue in water/EtOAc (100 mL each), separate the organic layer, extract the aq. layer with EtOAc (2×100 mL), combine the organic layers and dry with MgSO4. Filter the dried extract, evaporate under vacuo and purify the crude by flash column chromatography on a silica gel using CHCl3 to afford yellow oil.
  • Part C: Synthesis of 1-Isocyanato-4-(1-trifluoromethyl-vinyl)-benzene [0284]
    Figure US20040176443A1-20040909-C00034
  • Cool 20% phosgene in toluene (5.0 mL) to −40° C. under N2 atmosphere. Dissolve Part B material of Example 4 (0.47 g, 2.5 mmoles) in toluene and add dropwise to the cooled stirred solution. Stir at −40° C. for 30 mins followed by room temperature for 1 h and then at reflux for 1 h. Concentrate in vacuo to afford orange yellow liquid. [0285]
  • Part D: Synthesis of 2-Methyl-4-(3-trifluoromethyl-pyridin-2-yl)-piperazine-1-carboxylic acid [4-(1-trifluoromethyl-vinyl)-phenyl]-amide [0286]
    Figure US20040176443A1-20040909-C00035
  • Dissolve Part A material of example 4 (0.123 g, 0.5 mmoles) in toluene (1.0 mL) under nitrogen at room temperature. Add drop wise Part C material of Example 4 (0.106 g, 0.5 mmoles) dissolved in toluene (1.0 mL) to the mixture over a period of 5 mins and stir at room temperature for 20 hours. Evaporate the solvent from reaction mixture under vacuum and purify by flash column chromatography using CHCl[0287] 3 to afford yellow oil.
  • Part E: Title Compound: [0288]
    Figure US20040176443A1-20040909-C00036
  • Dissolve Part D material of example 4 (0.120 g, 0.262 mmoles) in EtOH(25.0 mL) at room temperature. Add 5% Pd/C (30 mg) and hydrogenate at 5 atm. of H[0289] 2 for 5 hours at room temperature. Filter the catalyst, evaporate the solvent from reaction mixture under vacuum and purify by PTLC using 5% MeOH/CHCl3 to afford yellow oil.
  • NMR (CDCl[0290] 3): δ 1.38-1.41 (d, 3H), 1.45-1.47 (d, 3H), 3.05-3.11 (m, 1H), 3.22-3.62 (m, 4H), 3.85-3.90 (m, 1H), 4.35-4.42 (m, 1H), 6.40 (s, 1H), 7.05-7.10 (m, 1H), 7.21-7.40 (m, 5H), 7.95-7.97 (d, 1H), 8.42-8.46 (d, 1H).
  • Mass spectrum (ESI): 461.3 (M+H). [0291]
  • Example 5 4-(3-Chloro-pyridin-2-yl)-2-methyl-piperazine-1-carboxylic acid [4-(2,2,2-trifluoro-1-methyl-ethyl)-phenyl]-amide
  • Part A: Synthesis of 4-(2,2,2-Trifluoro-1-methyl-ethyl)-phenylamine [0292]
    Figure US20040176443A1-20040909-C00037
  • Dissolve Part B material of example 4 (0.375 g, 0.262 mmoles) in MeOH (25.0 mL) at room temperature. Add raney Ni (500 mg) and hydrogenate at 40 atm. of H[0293] 2 for 20 hours at room temperature. Filter the catalyst, evaporate the solvent from reaction mixture under vacuum and purify by flash column chromatography using CHCl3 to afford yellow oil.
  • Part B: Synthesis of 1-Isocyanato-4-(2,2,2-trifluoro-1-methylethyl)-benzene [0294]
    Figure US20040176443A1-20040909-C00038
  • Cool 20% phosgene in toluene (2.0 mL) to −40° C. under N2 atmosphere. Dissolve Part A material of Example 5 (0.189 g, 1.0 mmole) in toluene and add drop wise to the cooled stirred solution. Stir at −40° C. for 30 mins followed by room temperature for 3 h and then at reflux for 18 h. Concentrate in vacuo to afford yellow liquid. [0295]
  • Part C: Title Compound: [0296]
    Figure US20040176443A1-20040909-C00039
  • Dissolve Part A material of example 1 (0.169 g, 0.795 mmoles) in toluene (1.0 mL) under nitrogen at room temperature. Add drop wise Part B material of Example 5 (0.171 g, 0.795 mmoles) dissolved in toluene (1.0 mL) to the mixture over a period of 5 mins and stir at room temperature for 20 hours. Evaporate the solvent from reaction mixture under vacuum and—purify by PTLC using 5% MeOH/CHCl[0297] 3 to afford white amorphous powder.
  • NMR (CDCl[0298] 3): δ 1.44-1.49 (2d, 6H), 2.94-3.11 (m, 2H), 3.36-3.51(m, 2H), 3.74-3.94 (m, 3H), 4.35-4.37 (m, 1H), 6.41 (s, 1H), 6.87-6.90 (dd, 1H), 7.23-7.26 (m, 2H), 7.36-7.38 (m, 2H), 7.607.63 (dd, 1H), 8.19-8.20 (dd, 1H).
  • Mass spectrum (ESI): 427.3 (M+H). [0299]
  • Example 6 (R)-4-(3-Chloro-pyridin-2-yl)-2-methyl-piperazine-1-carboxylic acid [4-(1,2,2,2-tetrafluoro-1-trifluoromethylethyl)-phenyl]-amide
  • Part A: Synthesis of 1-Isocyanato-4-(1,2,2,2-tetrafluoro-1-trifluoromethyl-ethyl)-benzene [0300]
    Figure US20040176443A1-20040909-C00040
  • Cool 20% phosgene in toluene (20.0 mL) to −40° C. under N2 atmosphere. Dissolve 4-heptafluoroisopropylaniline (2.0 g, 7.7 mmoles; see: EP 1006102 for aniline preparation) in toluene (5.0 mL) and add drop wise to the cooled stirred solution. Stir at −40° C. for 30 mins followed by room temperature for 2 h and then at reflux for 4 h. Concentrate in vacuo to afford yellow liquid. [0301]
  • Part B: Title Compound: [0302]
    Figure US20040176443A1-20040909-C00041
  • Dissolve Part A material of example 1 (0.106 g, 0.795 mmoles) in toluene (1.0 mL) under nitrogen at room temperature. Add drop wise Part A material of Example 6 (0.144 g, 0.5 mmoles) dissolved in toluene (1.0 mL) to the mixture over a period of 5 mins and stir at room temperature for 20 hours. Evaporate the solvent from reaction mixture under vacuum and purify by PTLC using 5% MeOH/CHCl[0303] 3 to afford white amorphous powder.
  • NMR (CDCl[0304] 3): δ 1.44-1.46 (d, 3H, J=1.6 Hz), 2.93-3.10 (m, 2H), 3.45-3.52(m, 1H), 3.74-3.78 (m, 2H), 3.91-3.94 (m, 1H), 4.37-4.38 (m, 1H), 6.60 (s, 1H), 6.87-6.90 (dd, 1H), 7.52-7.60 (m, 4H), 7.61-7.63 (m, 1H), 8.18-8.20(dd, 1H).
  • Mass spectrum (ESI): 499.2 (M+H). [0305]
  • Example 7 4-(3-Chloro-pyridin-2-yl)-2-methyl-piperazine-1-carbothioic acid (4-isopropyl-phenyl)-amide
  • [0306]
    Figure US20040176443A1-20040909-C00042
  • Dissolve Part A material of example 1 (0.212 g, 1.0 mmoles) in toluene (1.0 mL) under nitrogen at room temperature. Add drop wise 4-isopropylisothiocyanate (0.177 g, 0.5 mmoles) dissolved in toluene (1.0 mL) to the mixture over a period of 5 mins and stir at room temperature for 20 hours. Evaporate the solvent from reaction mixture under vacuum and purify by flash column chromatography using CHCl[0307] 3 to afford white solid (mp 49-51° C.).
  • NMR (CDCl[0308] 3): δ 1.22-1.25(d, 3H), 1.41-1.43 (d, 3H), 2.80-3.20 (m, 3H), 3.45-3.60(m, 1H), 3.65-3.80(m, 2H), 4.35-4.39 (m, 1H), 5.05-5.20 (m, 1H), 6.85-6.90 (s, 1H), 7.15-7.35 (m, 5H), 7.42-7.44(d, 1H), 8.18-8.20 (d, 1H).
  • Mass spectrum (ESI): 387.2 (M+H). [0309]
  • Example 8 Synthesis of 4-(3-Trifluoromethyl-2-pyridinyl)-N-(3-methoxy-4-hydroxyphenylmethyl)-1-piperazinecarboxamide
  • [0310]
    Figure US20040176443A1-20040909-C00043
  • A quantity of 0.2 mL of a 0.2 M isocyante solution in dichloroethane is treated with 0.26 mL of a 0.2 mL solution of piperazine in 95:5 toluene: N-Methyl Morphine at 60° C. for 16 hr. The resulting reaction solution was cooled to room temperature. To the resulting solution is added 1 drop of amino propyl morpholine and warmed to 60° C. for an additional hour. The resulting mixture is cooled to room temperature and chromatographed SiO[0311] 2 with ethyl acetate to afford 10 mg 63% of the title compound (Compound 1). MS m/z 410.16 found: 411, 433 Na adduct. Capsaicin rececptor Ki: 366 nM
  • Example 9
  • Additional Compounds [0312]
  • Using variations of the methods given in Schemes 1 and 2, and Examples 1-8 that will be readily apparent to one skilled in the art of organic synthesis the compounds list in Tables II, III and IV are prepared. Commercial grade reagents are used without further purification in the preparation of these compound [0313]
    TABLE II
    Figure US20040176443A1-20040909-C00044
    Compound R R1 R2 A X Calc Found Activity Chemical Name
    2 4-Butyl H H NH 3-NO2 383 384, * 4-(3-Nitro-2-pyridinyl)-N-[4-(n-
    406 Na butyl)phenyl]-1-piperazinecarboxamide
    adduct
    3 4-Butyl H H NH 3-CF3 406 407, * 4-(3-Trifluoromethyl-2-pyridinyl)-N-[4-
    429 Na (n-butyl)phenyl]-1-piperazinecarboxamide
    adduct
    4 4- H H NH 3-Me 338 339, * 4-(3-Methyl-2-pyridinyl)-N-[4-
    Isopropyl 361 Na (isopropyl)phenyl]-1-
    adduct Piperazinecarboxamide
    5 4-Butyl H H NH 3-Me 352 353, NA 4-(3-Methyl-2-pyridinyl)-N-[4-(n-
    375 Na butyl)phenyl]-1-piperazinecarboxamide
    adduct
    6 4- H H NH 3-CF3 352 353, * 4-(3-Trifluoromethyl-2-pyridinyl)-N-[4-
    Isopropyl 375 Na (isopropyl)phenyl]-1-
    adduct piperazinecarboxamide
    7 4- H H NH 3-Cl,5- 427 427, * 4-(3-Chloro-5-trifluoromethyl-2-
    Isopropyl CF3 449 Na pyridinyl)-N-[4-(isopropyl)phenyl]-1-
    adduct piperazinecarboxamide
    8 4- H H NH 3-Cl 359 359, * 4-(3-Chloro-2-pyridinyl)-N-[4-
    Isopropyl 381 Na (isopropyl)phenyl]-1-
    adduct piperazinecarboxamide
    9 4- H H NH 3,5- 393 393, * 4-(3,5-Dichloro-2-pyridinyl)-N-[4-
    Isopropyl diCl 415 Na (isopropyl)phenyl]-1-
    adduct piperazinecarboxamide
    10 4- H H CH═CH 3-CF3 * 1-(3-Methyl-2-pyridinyl)-3-(4-
    Isopropyl trifluoromethyl phenyl)-prop-2-en-1-one
    11 4-CF3 H H CH═CH 3-Me * 1-(3-Trifluoromethyl-2-pyrindinyl)-3-(4-
    isopropylphenyl)-prop-2-en-1-one
    12 4- H H NH 3-CN 349 * 4-(3-Cyano-2-pyridinyl)-N-[4-
    Isopropyl (isopropyl)phenyl]-1-
    piperazinecarboxamide
    13 4- Me H NH 3-Cl 373 373, NA 4-(3-Chloro-2-pyridinyl)-N-[4-
    Isopropyl 395 Na (isopropyl)phenyl]-2-methyl-1-
    adduct piperazinecarboxamide
    14 4- R- H NH 3-Cl 373 373, * 4-(3-Chloro-2-pyridinyl)-N-[4-
    Isopropyl Me 395 Na (isopropyl)phenyl]-1-
    adduct piperazinecarboxamide
    15 4- S- H NH 3-Cl 373 373, NA 4-(3-Chloro-2-pyridinyl)-N-[4-
    Isopropyl Me 395 Na (isopropyl)phenyl]-2-methylthio-1-
    adduct piperazinecarboxamide
    16 4-CF3 H H NH 3,5- 373 NA 4-(3,5-Dichloro-2-pyridinyl)-N-[4-
    diCl (isopropyl)phenyl]-1-
    piperazinecarboxamide
  • [0314]
    TABLE III
    Cmp. # STRUCTURE IUPAC Name EC50
    17
    Figure US20040176443A1-20040909-C00045
    N-(4-tert-butylphenyl)-4- (3-chloropyridin-2- yl)piperazine-1-carboxamide *
    18
    Figure US20040176443A1-20040909-C00046
    (2R)-4-(3-chloropyridin-2- yl)-N-(4-cyclohexylphenyl)-2- methylpiperazine-1-carboxamide *
    19
    Figure US20040176443A1-20040909-C00047
    (2R)-4-(3-chloropyridin-2- yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]- 2-methylpiperazine-1-carboxamide
    20
    Figure US20040176443A1-20040909-C00048
    (2R)-4-(3-chloropyridin-2- yl)-2-methyl-N-[4- (trifluoromethyl)phenyl]pierazine- 1-carboxamide *
    21
    Figure US20040176443A1-20040909-C00049
    (2R)-N-(4-tert-butylphenyl)- 4-(3-chloropyridin-2-yl)-2- methylpiperazine-1-carboxamide *
    22
    Figure US20040176443A1-20040909-C00050
    (2R)-4-(3-chloropyridin-2- yl)-N-(4-isopropylphenyl)-2- methylpiperazine-1-carboxamide *
    23
    Figure US20040176443A1-20040909-C00051
    (2S)-4-(3-chloropyridin-2- yl)-N-(4-trifluoromethylphenyl)-2- methylpiperazine-1-carboxamide *
    24
    Figure US20040176443A1-20040909-C00052
    (2S)-N-(4-tert-butylphenyl)- 4-(3-chloropyridin-2-yl)-2- methylpiperazine-1-carboxamide *
    25
    Figure US20040176443A1-20040909-C00053
    (2S)-4-(3-chloropyridin-2- yl)-N-(4-isopropylphenyl)-2- methylpiperazine-1-carboxamide *
    26
    Figure US20040176443A1-20040909-C00054
    (2R)-4-(3-chloropyridin-2- yl)-2-methyl-N-(4-piperidin-1- ylphenyl)piperazine-1-carboxamide *
    27
    Figure US20040176443A1-20040909-C00055
    (2R)-4-(3-chloropyridin-2- yl)-N-[2-fluoro-4- (trifluoromethyl)phenyl]-2- methylpiperazine-1-carboxamide *
    28
    Figure US20040176443A1-20040909-C00056
    (2R)-2-methyl-N-[4- (trifluoromethyl)phenyl]- 4-[3-(trifluoromethyl)pyridin- 2-yl]piperazine-1-carboxamide *
    29
    Figure US20040176443A1-20040909-C00057
    (2R)-N-(4-tert-butylphenyl)- 2-methyl-4-[3-(trifluoromethyl)- pyridin-2-yl]piperazine-1- carboxamide *
    30
    Figure US20040176443A1-20040909-C00058
    (2R)-N-(4-isopropylphenyl)- 2-methyl-4-[3-(trifluoromethyl)- pyridin-2-yl]piperazine-1- carboxamide *
    31
    Figure US20040176443A1-20040909-C00059
    4-(3-chloropyridin-2-yl)- 2,6-dimethyl-N-[4-(trifluoromethyl)- phenyl]piperazine-1-carboxamide *
    32
    Figure US20040176443A1-20040909-C00060
    N-(4-tert-butylphenyl)-4- (3-chloropyridin-2-yl)-2,6- dimethylpiperazine-1-carboxamide *
    33
    Figure US20040176443A1-20040909-C00061
    4-(3-chloropyridin-2-yl)- N-(4-isopropylphenyl)-2,6- dimethylpiperazine-1-carboxamide *
    34
    Figure US20040176443A1-20040909-C00062
    (2R)-N-(4-cyclohexylphenyl)- 2-methyl-4-[3-(trifluoromethyl)- pyridin-2-yl]piperazine-1- carboxamide *
    35
    Figure US20040176443A1-20040909-C00063
    4-(3-chloropyridin-2-yl)- N-(4-cyclohexylphenyl)- 2,6-dimethylpiperazine-1- carboxamide *
    36
    Figure US20040176443A1-20040909-C00064
    (2R)-4-(3-chloropyridin-2- yl)-N-(4-cyclopentylphenyl)-2- methylpiperazine-1-carboxamide *
    37
    Figure US20040176443A1-20040909-C00065
    (2R)-N-(4-cyclopentylphenyl)- 2-methyl-4-[3-(trifluoromethyl)- pyridin-2-yl]piperazine-1- carboxamide *
    38
    Figure US20040176443A1-20040909-C00066
    (2R)-4-isoquinolin-1-yl-2- methyl-N-[4-(trifiuoromethyl)- phenyl]piperazine-1-carboxamide *
    39
    Figure US20040176443A1-20040909-C00067
    (2R)-N-(4-tert-butylphenyl)- 4-isoquinolin-1-yl-2- methylpiperazine-1-carboxamide *
    40
    Figure US20040176443A1-20040909-C00068
    (2R)-N-(4-isopropylphenyl)-4- isoquinolin-1-yl-2- methylpiperazine-1-carboxamide *
    41
    Figure US20040176443A1-20040909-C00069
    (2R)-N-(4-cyclopentylphenyl)- 4-isoquinolin-1-yl-2- methylpiperazine-1-carboxamide *
    42
    Figure US20040176443A1-20040909-C00070
    (2R)-N-(4-cyclohexylphenyl)- 4-isoquinolin-1-yl-2- methylpiperazine-1-carboxamide *
    43
    Figure US20040176443A1-20040909-C00071
    (2R)-N-(4-tert-butylphenyl)- 4-[3-(dimethylamino)pyridin-2-yl]- 2-methylpiperazine-1-carboxamide *
    44
    Figure US20040176443A1-20040909-C00072
    (2R)-4-[3-(dimethylamino)- pyridin-2-yl]-2-methyl-N-[4- (trifluoromethyl)phenyl]- piperazine-1-carboxamide *
    45
    Figure US20040176443A1-20040909-C00073
    (2R)-N-(4-tert-butylphenyl)-4- (3-methoxypyridin-2-yl)-2- methylpiperazine-1-carboxamide *
    46
    Figure US20040176443A1-20040909-C00074
    (2R)-4-(3-methoxypyridin- 2-yl)-2-methyl-N-[4- (trifluoromethyl)phenyl]- piperazine-1-carboxamide
    47
    Figure US20040176443A1-20040909-C00075
    (2R)-N-(4-cyclohexylphenyl)- 4-(3-methoxypyridin-2-yl)-2- methylpiperazine-1-carboxamide *
    48
    Figure US20040176443A1-20040909-C00076
    (2R)-4-(3-chloropyridin-2- yl)-N-[4-(3,6-dihydro-2H- pyran-4-yl)phenyl]-2- methylpiperazine-1-carboxamide *
    49
    Figure US20040176443A1-20040909-C00077
    (2R)-4-(3-chloropyridin-2- yl)-2-methyl-N-(4-tetrahydro- 2H-pyran-4-ylphenyl)piperazine- 1-carboxamide *
    50
    Figure US20040176443A1-20040909-C00078
    (2R)-4-(3-chloropyridin-2- yl)-N-[4-(4-hydroxytetrahydro-2H- pyran-4-yl)phenyl]-2- methylpiperazine-1-carboxamide *
    51
    Figure US20040176443A1-20040909-C00079
    (2R)-N-[4-(4-hydroxytetra- hydro-2H-pyran-4-yl)phenyl]-2- methyl-4-[3-(trifluoromethyl)- pyridin-2-yl]piperazine-1- carboxamide *
    52
    Figure US20040176443A1-20040909-C00080
    (2R)-4-(3-chloropyridin-2- yl)-2-methyl-N-[4-(2-methyl-1,3- thiazol-4-yl)phenyl]piperazine-1- carboxamide *
    53
    Figure US20040176443A1-20040909-C00081
    (2R)-4-(3-chloropyridin-2- yl)-N-[4-(2-ethyl-1,3-thiazol- 4-yl)phenyl]-2-methyl- piperazine-1-carboxamide *
    54
    Figure US20040176443A1-20040909-C00082
    (2R)-4-(3-chloropyridin-2- yl)-N-[4-(2-methoxy-1,1- dimethylethyl)phenyl]-2- methylpiperazine-1-carboxamide *
    55
    Figure US20040176443A1-20040909-C00083
    (2R)-N-[4-(2-methoxy-1,1- dimethylethyl)phenyl]-2- methyl-4-[3-(trifluoromethyl)- pyridin-2-yl]piperazine-1- carboxamide *
    56
    Figure US20040176443A1-20040909-C00084
    (2R)-4-(3-chloropyridin-2- yl)-N-[4-(1-cyano-1-methyl- ethyl)phenyl]-2-methyl- piperazine-1-carboxamide *
    57
    Figure US20040176443A1-20040909-C00085
    (2R)-N-[4-(1-cyano-1-methylethyl)- phenyl]-2-methyl-4-[3- (trifluoromethyl)pyridin-2- yl]piperazine-1-carboxamide *
    58
    Figure US20040176443A1-20040909-C00086
    N-(4-tert-butylphenyl)-4- (3-chloropyridin-2-yl)-2- ethylpiperazine-1-carboxamide *
    59
    Figure US20040176443A1-20040909-C00087
    4-(3-chloropyridin-2-yl)- 2-ethyl-N-[4-(trifluoromethyl)- phenyl]piperazine-1-carboxamide *
    60
    Figure US20040176443A1-20040909-C00088
    4-(3-chloropyridin-2-yl)- 2-ethyl-N-(4-isopropylphenyl)- piperazine-1-carboxamide *
    61
    Figure US20040176443A1-20040909-C00089
    N-(4-tert-butylphenyl)-2- ethyl-4-[3-(trifluoromethyl)- pyridin-2-yl]piperazine-1- carboxamide *
    62
    Figure US20040176443A1-20040909-C00090
    2-ethyl-N-[4-(trifluoro- methyl)phenyl]-4-[3- (trifluoromethyl)pyridin-2- yl]piperazine-1-carboxamide *
    63
    Figure US20040176443A1-20040909-C00091
    2-ethyl-N-(4-isopropylphenyl)- 4-[3-(trifluoromethyl)pyridin- 2-yl]piperazine-1-carboxamide *
    64
    Figure US20040176443A1-20040909-C00092
    2-tert-butyl-N-(4-tert- butylphenyl)-4-(3-chloropyridin- 2-yl)piperazine-1-carboxamide *
    65
    Figure US20040176443A1-20040909-C00093
    2-tert-butyl-4-(3- chloropyridin-2-yl)-N-[4- (trifluoromethyl)phenyl]- piperazine-1-carboxamide
    66
    Figure US20040176443A1-20040909-C00094
    2-tert-butyl-4-(3- chloropyridin-2-yl)-N-(4- isopropylphenyl)piperazine-1- carboxamide
    67
    Figure US20040176443A1-20040909-C00095
    2-tert-butyl-N-(4-tert- butylphenyl)-4-[3- (trifluoromethyl)pyridin- 2-yl]piperazine-1-carboxamide
    68
    Figure US20040176443A1-20040909-C00096
    2-tert-butyl-N-[4- (trifluoromethyl)phenyl]- 4-[3-(trifluoromethyl)- pyridin-2-yl]piperazine-1- carboxamide *
    69
    Figure US20040176443A1-20040909-C00097
    N-(4-tert-butylphenyl)-4- (3-chloropyridin-2-yl)-2- isopropylpiperazine-1- carboxamide *
    70
    Figure US20040176443A1-20040909-C00098
    4-(3-chloropyridin-2-yl)- 2-isopropyl-N-[4-(trifluoro- methyl)phenyl]piperazine- 1-carboxamide
    71
    Figure US20040176443A1-20040909-C00099
    4-(3-chloropyridin-2-yl)- 2-isopropyl-N-(4-isopropyl- phenyl)piperazine-1-carboxamide
    72
    Figure US20040176443A1-20040909-C00100
    N-(4-tert-butylphenyl)-2- isopropyl-4-[3-(trifluoro- methyl)pyridin-2-yl]- piperazine-1-carboxamide *
    73
    Figure US20040176443A1-20040909-C00101
    2-isopropyl-N-[4-(trifluoro- methyl)phenyl]-4-[3-(tri- fluoromethyl) pyridin-2-yl]- piperazine-1-carboxamide *
    74
    Figure US20040176443A1-20040909-C00102
    2-isopropyl-N-(4-isopropyl- phenyl)-4-[3-(trifluoromethyl)- pyridin-2-yl]piperazine-1- carboxamide *
    75
    Figure US20040176443A1-20040909-C00103
    (2R)-4-(3-fluoropyridin-2- yl)-2-methyl-N-[4-(trifluoro- methyl)phenyl]piperazine- 1-carboxamide *
    76
    Figure US20040176443A1-20040909-C00104
    (2R)-N-(4-tert-butylphenyl)- 4-(3-fluoropyridin-2-yl)-2- methylpiperazine-1-carboxamide *
    77
    Figure US20040176443A1-20040909-C00105
    (2R)-4-(3-fluoropyridin-2- yl)-N-(4-isopropylphenyl)- 2-methylpiperazine-1-carboxamide *
    78
    Figure US20040176443A1-20040909-C00106
    (2R)-N-(4-cyclohexylphenyl)- 4-(3-fluoropyridin-2-yl)-2- methylpiperazine-1-carboxamide *
    79
    Figure US20040176443A1-20040909-C00107
    (2R)-N-(4-cyclopentylphenyl)- 4-(3-fluoropyridin-2-yl)-2- methylpiperazine-1-carboxamide *
    80
    Figure US20040176443A1-20040909-C00108
    N-(4-chlorophenyl)-4-(6- chloropyridin-2-yl)piperazine- 1-carboxamide
    81
    Figure US20040176443A1-20040909-C00109
    4-(6-chloropyridin-2-yl)- N-phenylpiperazine-1- carboxamide
    82
    Figure US20040176443A1-20040909-C00110
    (2R)-N-(4-tert-butylphenyl)- 4-(3-cyanopyridin-2-yl)-2- methylpiperazine-1-carboxamide *
    83
    Figure US20040176443A1-20040909-C00111
    (2R)-4-(3-cyanopyridin-2- yl)-2-methyl-N-[4-(trifluoro- methyl)phenyl]- piperazine-1-carboxamide *
    84
    Figure US20040176443A1-20040909-C00112
    (2R)-2-methyl-4-(6-methyl- pyridin-2-yl)-N-[4-(tri- fluoromethyl)phenyl]- piperazine-1-carboxamide
    85
    Figure US20040176443A1-20040909-C00113
    (2R)-4-(6-methoxypyridin- 2-yl)-2-methyl-N-[4- (trifluoromethyl)phenyl]- piperazine-1-carboxamide
    86
    Figure US20040176443A1-20040909-C00114
    (2R)-N-(4-tert-butylphenyl)- 2-methyl-4-(6-methylpyridin-2- yl)piperazine-1-carboxamide *
    87
    Figure US20040176443A1-20040909-C00115
    (2R)-N-(4-tert-butylphenyl)- 4-(6-methoxypyridin-2-yl)-2- methylpiperazine-1-carboxamide
    88
    Figure US20040176443A1-20040909-C00116
    (2R)-N-(4-isopropylphenyl)- 2-methyl-4-(6-methylpyridin-2- yl)piperazine-1-carboxamide
    89
    Figure US20040176443A1-20040909-C00117
    (2R)-N-(4-isopropylphenyl)- 4-(6-methoxypyridin-2-yl)-2- methylpiperazine-1-carboxamide
    90
    Figure US20040176443A1-20040909-C00118
    (2R)-N-(4-cyclopentylphenyl)- 2-methyl-4-(6-methylpyridin-2- yl)piperazine-1-carboxamide
    91
    Figure US20040176443A1-20040909-C00119
    (2R)-N-(4-cyclopentylphenyl)- 4-(6-methoxypyridin-2-yl)-2- methylpiperazine-1-carboxamide
  • [0315]
    TABLE IV
    Cpd. # STRUCTURE IUPAC Name EC50
    93
    Figure US20040176443A1-20040909-C00120
    4-(3-chloropyridin-2-yl)- N-[5-(trifluoromethyl)pyridin- 2-yl]piperazine-1-carboxamide *
    94
    Figure US20040176443A1-20040909-C00121
    (2R)-4-(3-chloropyridin- 2-yl)-2-methyl-N-[5- (trifluoromethyl)pyridin- 2-yl]piperazine-1-carboxamide *
    95
    Figure US20040176443A1-20040909-C00122
    (2R)-N-(4-tert-butylphenyl)- 4-(3-chloropyrazin-2-yl)-2- methylpiperazine-1-carboxamide *
    96
    Figure US20040176443A1-20040909-C00123
    (2R)-4-(3-chloropyrazin- 2-yl)-N-(4-isopropylphenyl)-2- methylpiperazine-1-carboxamide *
    97
    Figure US20040176443A1-20040909-C00124
    (2R)-4-(3-chloropyrazin- 2-yl)-2-methyl-N-[4- (trifluoromethyl)phenyl]- piperazine-1-carboxamide *
    98
    Figure US20040176443A1-20040909-C00125
    (2R)-4-(3-chloropyridin- 2-yl)-2-methyl-N-[6- (trifluoromethyl)pyridin- 3-yl]piperazine-1-carboxamide *
    99
    Figure US20040176443A1-20040909-C00126
    (2R)-N-(4-tert-butyl- cyclohexyl)-4-(3-chloropyridin- 2-yl)-2-methylpiperazine-1- carboxamide *
    100
    Figure US20040176443A1-20040909-C00127
    (2R)-4-(3-chloropyridin- 2-yl)-N-(4-isopropylcyclohexyl)- 2-methylpiperazine-1- carboxamide *
    101
    Figure US20040176443A1-20040909-C00128
    (2R)-N-(4-isopropyl- cyclohexyl)-2-methyl-4-[3- (trifluoromethyl)pyridin- 2-yl]piperazine-1-carboxamide *
    102
    Figure US20040176443A1-20040909-C00129
    (2R)-4-isoquinolin-1-yl- 2-methyl-N-[4-(trifluoro- methyl)phenyl]piperazine-1-carboxamide
    103
    Figure US20040176443A1-20040909-C00130
    (2R)-N-(4-tert-butylphenyl)- 4-isoquinolin-1-yl-2- methylpiperazine-1-carboxamide
    104
    Figure US20040176443A1-20040909-C00131
    (2R)-N-(4-isopropylphenyl)- 4-isoquinolin-1-yl-2- methylpiperazine-1-carboxamide
    105
    Figure US20040176443A1-20040909-C00132
    (2R)-N-(4-cyclopentyl- phenyl)-4-isoquinolin-1-yl-2- methylpiperazine-1-carboxamide
    106
    Figure US20040176443A1-20040909-C00133
    (2R)-N-(4-cyclohexylphenyl)- 4-isoquinolin-1-yl-2- methylpiperazine-1-carboxamide
  • [0316]
    TABLE V
    Cmp # Structure Name EC50
    107
    Figure US20040176443A1-20040909-C00134
    N-(4-chlorophenyl)-4- [4-(trifluoromethyl) pyridin-2-yl]piperazine-1-carboxamide *
    108
    Figure US20040176443A1-20040909-C00135
    N-[4-(trifluoromethoxy)- phenyl]-4-[4-(trifluoro- methyl)pyridin-2-yl]- piperazine-1-carboxamide *
    109
    Figure US20040176443A1-20040909-C00136
    N-(3-chlorophenyl)-4- [4-(trifluoromethyl)- pyridin-2-yl]piperazine- 1-carboxamide *
    110
    Figure US20040176443A1-20040909-C00137
    N-[3-(trifluoromethyl)- phenyl]-4-[3-(trifluoromethyl)- pyridin-2-yl]piperazine-1- carboxamide *
    111
    Figure US20040176443A1-20040909-C00138
    N-(4-methylphenyl)-4-[3- (trifluoromethyl)pyridin-2- yl]piperazine-1-carboxamide
    112
    Figure US20040176443A1-20040909-C00139
    N-(3-bromophenyl)-4-[3- (trifluoromethyl)pyridin-2- yl]piperazine-1-carboxamide
    113
    Figure US20040176443A1-20040909-C00140
    N-(3-methoxyphenyl)-4- [3-(trifluoromethyl)pyridin- 2-yl]piperazine-1-carboxamide *
    114
    Figure US20040176443A1-20040909-C00141
    4-(5-nitropyridin-2-yl)- N-[4-(trifluoromethoxy)phenyl]piperazine-1-carboxamide
    115
    Figure US20040176443A1-20040909-C00142
    N-(1-naphthyl)-4-[3- (trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide
    116
    Figure US20040176443A1-20040909-C00143
    N-(3-nitrophenyl)-4-[3- (trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide *
    117
    Figure US20040176443A1-20040909-C00144
    N-[4-(trifluoromethoxy)- phenyl]-4-[3-(trifluoro- methyl)pyridin-2-yl]- piperazine-1-carboxamide *
    118
    Figure US20040176443A1-20040909-C00145
    N-(4-chloro-3-nitrophenyl)- 4-[3-(trifluoromethyl)- pyridin-2-yl]piperazine-1- carboxamide *
    119
    Figure US20040176443A1-20040909-C00146
    N-(3,5-dichlorophenyl)-4- [3-(trifluoromethyl)- pyridin-2-yl] piperazine-1- carboxamide *
    120
    Figure US20040176443A1-20040909-C00147
    (2R)-4-(3-chloropyridin-2- yl)-N-{4-[cyano(phenyl)- methyl]phenyl}-2- methylpiperazin-1-carboxamide *
    121
    Figure US20040176443A1-20040909-C00148
    (2R)-4-(3-chloropyridin-2- yl)-2-methyl-N-[3-methyl-4- (trifluoromethyl)phenyl]- piperazine-1-carboxamide *
    122
    Figure US20040176443A1-20040909-C00149
    (2R)-4-(3-fluoropyridin-2- yl)-2-methyl-N-[3-methyl-4- (trifluoromethyl)phenyl]- piperazine-1-carboxamide *
    123
    Figure US20040176443A1-20040909-C00150
    (2R)-4-{3-[bis(methyl- sulfonyl)amino]pyridin-2- yl}-N-(4-tert-butylphenyl)- 2-methylpiperazine-1-carboxamide *
    124
    Figure US20040176443A1-20040909-C00151
    (2R)-2-methyl-N-[3-methyl- 4-(trifluoromethyl)phenyl]- 4-[3-(trifluoromethyl)- pyridin-2-yl] piperazine-1- carboxamide *
    125
    Figure US20040176443A1-20040909-C00152
    (2R)-4-(3-chloropyridin-2- yl)-2-methyl-N-{4-[1- (trifluoromethyl)vinyl]phenyl} piperazine-1- carboxamide *
    126
    Figure US20040176443A1-20040909-C00153
    (2R)-2-methyl-4-[3- (trifluoromethyl)pyridin-2-yl]- N-{4-[1-(trifluoromethyl)- vinyl] phenyl}piperazine- 1-carboxamide *
    127
    Figure US20040176443A1-20040909-C00154
    (2R)-4-(3-fluoropyridin- 2-yl)-2-methyl-N-{4-[1- (trifluoromethyl)vinyl]phenyl}piperazine-1- carboxamide *
    128
    Figure US20040176443A1-20040909-C00155
    (2R)-N-(4-sec-butylphenyl)- 4-(3-fluoropyridin-2-yl)-2- methylpiperazine-1-carboxamide *
    129
    Figure US20040176443A1-20040909-C00156
    (2R)-2-methyl-N-[4-(2,2,2- trifluoro-1-methylethyl)phenyl]- 4-[3-(trifluoromethyl)pyridin- 2-yl]piperazine-1-carboxamide *
    130
    Figure US20040176443A1-20040909-C00157
    (2R)-4-(3-fluoropyridin-2- yl)-2-methyl-N-[4-(2,2,2- trifluoro-1-methylethyl)phenyl]- piperazine-1-carboxamide *
    131
    Figure US20040176443A1-20040909-C00158
    (2R)-4-(3-chloro-5- nitropyridin-2-yl)-2-methyl-N- [4-(trifluoromethyl)phenyl]- piperazine-1-carboxamide *
    132
    Figure US20040176443A1-20040909-C00159
    (2R)-4-(5-amino-3- chloropyridin-2-yl)-2-methyl- N-[4-(trifluoromethyl)phenyl]- piperazine-1-carboxamide *
    133
    Figure US20040176443A1-20040909-C00160
    (2R)-4-(3-fluoropyridin- 2-yl)-N-[3-fluoro-4- (trifluoromethyl) phenyl]-2-methylpiperazine- 1-carboxamide *
    134
    Figure US20040176443A1-20040909-C00161
    (2R)-N-[3-fluoro-4- (trifluoromethyl)phenyl]- 2-methyl-4-[3-(trifluoro- methyl) pyridin-2-yl]- piperazine-1-carboxamide *
    135
    Figure US20040176443A1-20040909-C00162
    (2R)-4-(3-chloropyridin- 2-yl)-2-methyl-N-[4-(2,2,2- trifluoro-1-methylethyl)- phenyl]piperazine-1-carboxamide *
    136
    Figure US20040176443A1-20040909-C00163
    (2R)-4-(3-chloropyridin- 2-yl)-2-methyl-N-(2,2,4,4- tetrafluoro-4H-1,3-benzodioxin- 6-yl)piperazine-1-carboxamide *
    137
    Figure US20040176443A1-20040909-C00164
    (2R)-4-(3-fluoropyridin- 2-yl)-2-methyl-N-(2,2,4,4- tetrafluoro-4H-1,3-benzodioxin- 6-yl)piperazine-1-carboxamide *
    138
    Figure US20040176443A1-20040909-C00165
    (2R)-2-methyl-N-(2,2,4,4- tetrafluoro-4H-1,3-benzodioxin- 6-yl)-4-[3-(trifluoromethyl)- pyridin-2-yl]piperazine-1- carboxamide *
    139
    Figure US20040176443A1-20040909-C00166
    (2R)-4-[3-(aminosulfonyl)- pyridin-2-yl]-N-(4-tert- butylphenyl)-2-methylpiperazine- 1-carboxamide *
    140
    Figure US20040176443A1-20040909-C00167
    (2R)-N-(4-benzoylphenyl)-4- (3-chloropyridin-2-yl)-2- methylpiperazine-1-carboxamide *
    141
    Figure US20040176443A1-20040909-C00168
    (2R)-4-(3-chloropyridin-2- yl)-N-(4-iodophenyl)-2- methylpiperazine-1-carboxamide *
    142
    Figure US20040176443A1-20040909-C00169
    (2R)-4-(3-chloropyridin-2- yl)-N-(9H-fluoren-2-yl)-2- methylpiperazine-1-carboxamide *
    143
    Figure US20040176443A1-20040909-C00170
    (2R)-N-(9H-fluoren-2- yl)-2-methyl-4-[3-(trifluoro- methyl) pyridin-2-yl]- piperazine-1-carboxamide *
    144
    Figure US20040176443A1-20040909-C00171
    (2R)-4-[3-cyano-6- (trifluoromethyl)pyridin-2-yl]- 2-methyl-N-[4-(trifluoromethyl) phenyl]piperazine-1-carboxamide
    145
    Figure US20040176443A1-20040909-C00172
    (2R)-N-(4-tert-butyl- phenyl)-4-[3-cyano-6- (trifluoromethyl)pyridin-2-yl]- 2-methylpiperazine-1-carboxamide
    146
    Figure US20040176443A1-20040909-C00173
    (2R)-4-[3-cyano-6- (trifluoromethyl)pyridin-2-yl]- N-(4-cyclopentylphenyl)-2- methylpiperazine-1-carboxamide
    147
    Figure US20040176443A1-20040909-C00174
    (2R)-4-[3-cyano-6- (trifluoromethyl)pyridin-2-yl]- N-(4-cyclohexylphenyl)-2- methylpiperazine-1-carboxamide
    148
    Figure US20040176443A1-20040909-C00175
    (2R)-4-(3-chloropyridin- 2-yl)-2-methyl-N-{4-[2,2,2- trifluoro-1,1-bis(trifluoro- methyl)ethyl]phenyl}piperazine-1-carboxamide *
    149
    Figure US20040176443A1-20040909-C00176
    (2R)-2-methyl-N-{4- [2,2,2-trifluoro-1,1-bis(tri- fluoromethyl)ethyl]phenyl}- 4-[3-(trifluoromethyl)pyridin- 2-yl]piperazine-1-carboxamide *
    150
    Figure US20040176443A1-20040909-C00177
    (2R)-4-(3-chloropyridin-2- yl)-N-(3-iodophenyl)-2-methyl- piperazine-1-carboxamide *
    151
    Figure US20040176443A1-20040909-C00178
    (2R)-4-(3-fluoropyridin-2- yl)-N-(3-iodophenyl)-2- methylpiperazine-1-carboxamide *
    152
    Figure US20040176443A1-20040909-C00179
    (2R)-N-(4-butylphenyl)- 4-(3-chloropyridin-2-yl)-2- methylpiperazine-1-carboxamide *
    153
    Figure US20040176443A1-20040909-C00180
    2-(fluoromethyl)-N-[4- (trifluoromethyl)phenyl]-4-[3- (trifluoromethyl)pyridin-2- yl]piperazine-1-carboxamide *
    154
    Figure US20040176443A1-20040909-C00181
    (2R)-4-(3-chloropyridin-2- yl)-2-methyl-N-[4-methyl-3-(tri- fluoromethyl)phenyl]piperazine- 1-carboxamide *
    155
    Figure US20040176443A1-20040909-C00182
    (2R)-2-methyl-N-[4- methyl-3-(trifluoromethyl)- phenyl]-4-[3-(trifluoro- methyl)pyridin-2-yl]- piperazine-1-carboxamide *
    156
    Figure US20040176443A1-20040909-C00183
    (2R)-N-[4-bromo-3- (trifluoromethyl)phenyl]-4- (3-chloropyridin-2-yl)-2- methylpiperazine-1-carboxamide *
    157
    Figure US20040176443A1-20040909-C00184
    (2R)-N-[4-bromo-3- (trifluoromethyl)phenyl]-2- methyl-4-[3-(trifluoromethyl) pyridin-2-yl]piperazine-1- carboxamide *
    158
    Figure US20040176443A1-20040909-C00185
    (2R)-4-(3-chloropyridin- 2-yl)-N-[4-chloro-3-(trifluoro- methyl) phenyl]-2- methylpiperazine-1-carboxamide *
    159
    Figure US20040176443A1-20040909-C00186
    (2R)-4-(3-chloropyridin- 2-yl)-N-[4-fluoro-3-(trifluoro- methyl) phenyl]-2- methylpiperazine-1-carboxamide *
    160
    Figure US20040176443A1-20040909-C00187
    (2R)-N-[4-chloro-3-(trifluoro- methyl)phenyl]-2-methyl-4-[3- (trifluoromethyl) pyridin-2- yl]piperazine-1-carboxamide *
    161
    Figure US20040176443A1-20040909-C00188
    (2R)-N-[4-fluoro-3-(trifluoro- methyl) phenyl]-2-methyl-4-[3- (trifluoromethyl) pyridin-2-yl]piperazine-1-carboxamide *
    162
    Figure US20040176443A1-20040909-C00189
    (2R)-4-(3-chloropyridin-2-yl)-2- methyl-N-{4-[1,2,2,2-tetrafluoro- 1-(trifluoromethyl)ethyl]phenyl}- piperazine-1-carboxamide *
    163
    Figure US20040176443A1-20040909-C00190
    (2R)-2-methyl-N-{4-[1,2,2,2- tetrafluoro-1-(trifluoromethyl) ethyl]phenyl}-4-[3- (trifluoromethyl)pyridin-2-yl]- piperazine-1-carboxamide *
  • [0317]
    TABLE VI
    Cmp.
    # Structure NAME EC50
    164
    Figure US20040176443A1-20040909-C00191
    (2R)-4-(3- chloropyrazin-2-yl)-2- methyl-N-{4-[1,2,2,2- tetrafluoro-1- (trifluoromethyl) ethyl]phenyl}piperazine-1- carboxamide *
    165
    Figure US20040176443A1-20040909-C00192
    (2R)-4-(3- chloropyrazin-2-yl)-2- methyl-N-{4- cyclopentyl-phenyl}piperazine-1- carboxamide *
    166
    Figure US20040176443A1-20040909-C00193
    (2R)-4-(3- chloropyrazin-2-yl)-2- methyl-N-{4- cyclohexyl-phenyl}piperazine-1- carboxamide *
    167
    Figure US20040176443A1-20040909-C00194
    *
    168
    Figure US20040176443A1-20040909-C00195
    *
    169
    Figure US20040176443A1-20040909-C00196
    *
    170
    Figure US20040176443A1-20040909-C00197
    *
    171
    Figure US20040176443A1-20040909-C00198
    *
    172
    Figure US20040176443A1-20040909-C00199
    *
    173
    Figure US20040176443A1-20040909-C00200
    *
    174
    Figure US20040176443A1-20040909-C00201
  • Example 10
  • Capsaicin Receptor Binding Assay [0318]
  • The following assay is a standard assay of capsaicin receptor binding that may be used to determine the binding affinity of compounds for the capsaicin (VR1) receptor. [0319]
  • Materials. [[0320] 3H]Resiniferatoxin (RTX; 37 Ci/mmol) was synthesized by and obtained from the Chemical Synthesis and Analysis Laboratory, National Cancer Institute-Frederick Cancer Research and Development Center, Frederick, Md. [3H] RTX may also be obtained from commercial vendors, e.g., Amersham Pharmacia Biotech, Inc. 800 Centennial Avenue, P.O. Box 1327, Piscataway, N.J. 08855 (code TRK 1069). Nonradioactive RTX may be purchased from Alexis Corp. (San Diego, Calif.) and capsazepine from RBI (Natick, Mass.).
  • Molecular Biology [0321]
  • A cDNA encoding the full length human capsaicin receptor (SEQ ID NO:1 or SEQ ID NO:2) is subcloned in the appropriate orientation for expression into an expression vector such as pcDNA3.1 (Invitrogen, Carlsbad, Calif.) or pUHG102-3 (Clontech, Palo Alto, Calif.) for recombinant expression in mammalian cells. [0322]
  • Cell Culture. Human embryonic kidney (HEK293) cells are transfected with a pcDNA3.1 expression consrtuct encoding the full length human capsaicin receptor (i.e. containing either the nucleotide sequence of SEQ ID NO:1 and SEQ ID NO:2) using standard methods. These transfected cells are selected for two weeks in media containing G418 (400 ug/ml) and then maintained as a pool of stably transfected cells. [0323]
  • pUHG102 VR1 expression plasmids are transfected into Chinese Hamster Ovary (CHO) cells containing the pTet Off Regulator plasmid (Clontech). In these cells, expression of the pUHG plasmid is repressed in the presence of tetracycline but is induced upon removal of the antibiotic. Stable clones are isolated in culture medium containing puromycin (10 ug/ml) and maintained in medium supplemented with tetracycline (1 ug/ml). Cells utilized for assays are grown in culture medium without antibiotic for 48-72 hours prior to use. For radioligand binding experiments, cells are seeded in T175 cell culture flasks in media without antibiotics and grown to approximately 90% confluency. The flasks are then washed with PBS and harvested in PBS containing 5 mM EDTA. The cells are pelleted by gentle centrifugation and stored at −80° C. until assayed. [0324]
  • Membrane Preparations [0325]
  • Previously frozen cells are disrupted with the aid of a tissue homogenizer in ice-cold HEPES homogenization buffer (5 mM KCl 5, 5.8 mM NaCl, 0.75 mM CaCl[0326] 2, 2 mM MgCl2, 320 mM sucrose, and 10 mM HEPES pH 7.4). Tissue homogenates are first centrifuged for 10 min at 1000×g (4° C.) to remove the nuclear fraction and debris and then the supernatant from the first centrifugation is further centrifuged for 30 min at 35,000×g (4° C.) to obtain a partially purified membrane fraction. Membranes are resuspended in the HEPES homogenization buffer prior to being assayed. An aliquot of this membrane homogenate is used to determine protein concentration via the Bradford method (BIO-RAD Protein Assay Kit, #500-0001, BIO-RAD, Hercules, Calif.).
  • Radioligand Binding [0327]
  • Binding studies with [[0328] 3H]RTX are carried out essentially according to a published protocol (Szallasi and Blumberg, 1992, J. Pharmacol. Exp. Ter. 262: 883-888) in which non-specific RTX binding is reduced by adding bovine alpha, acid glycoprotein (100 ug per tube) after the binding reaction has been terminated. The homogenate is centrifuged as before and resuspended to a protein concentration of 333 ug/ml in homogenization buffer. Binding assay mixtures were set up on ice and contained [3H]RTX (specific activity 2200 mCi/ml), 2 ul non-radioactive ligands test compound, 0.25 mg/ml bovine serum albumin (Cohn fraction V), and 5×104-1×105 VR1-transfected cells. The final volume was adjusted to 500 ul (competition binding assays) or 1,000 ul (saturation binding assays) with the ice-cold HEPES homogenization buffer solution (pH 7.4) described above. Non-specific binding was defined as that occurring in the presence of 1 uM non-radioactive RTX. For saturation binding, [3H]RTX was added in the concentration range of 7-1,000 pM, using 1 to 2 dilutions. Typically 11 concentration points are collected per saturation binding curve.
  • Competition binding assays were performed in the presence of 60 pM [[0329] 3H]RTX and various concentrations of competing ligands. The binding reactions are initiated by transferring the assay mixtures into a 37° C. water bath and are terminated following a 60 min incubation period by cooling the tubes on ice. Membrane-bound RTX is separated from free, as well as any alpha, acid glycoprotein-bound RTX, by filtration onto WALLAC glass fiber filters (PERKIN-ELMER, Gaithersburg, Md.) which are pre-soaked with 1.0% PEI (polyethyleneimine) for 2 hours prior to use. Filters are allowed to dry overnight then counted in a WALLAC 1205 BETA PLATE counter after addition of WALLAC BETA SCINT scintillation fluid.
  • Equilibrium binding parameters were determined by fitting the allosteric Hill equation to the measured values with the aid of the computer program FIT P (Biosoft, Ferguson, Mo.) as described previously (Szallasi, et al. (1993) [0330] J. Pharmacol. Exp. Ther. 266:678-683)).
  • Though compounds exhibiting K[0331] i values for capsaicin receptors of greater than 1 uM are generally less preferred as pharmaceutical agents, useful compounds of the invention exhibit Ki values for capsaicin receptors of less than 4 uM, more preferred compounds exhibit Ki values of less than 1 uM, even more preferred compounds exhibit Ki values of less than 100 nM, more highly preferred compounds exhibit Ki values of less than 50 nM, even more highly preferred compounds exhibit Ki values of less than 25 nM and the most preferred compounds of the invention yield Ki values of less than or about equal to 10 nM in this assay.
  • Example 11
  • Calcium Mobilization Assay [0332]
  • The following assay can be used to monitor the response of cells capsaicin receptors to capsaicin and other vanilloid ligands of the capsaicin receptor. The assay can also be used to determine if test compounds act as agonists or antagonists of capsaicin receptors. [0333]
  • Cells transfected with expression plasmids (as described in Example 10) and thereby expressing the human capsaicin receptor are seeded and grown to 70-90% confluency in FALCON black-walled, clear-bottomed 96-well plates (#3904, BECTON-DICKINSON, Franklin Lakes, N.J.). The culture media are emptied from the 96 well plates and FLUO-3 AM calcium sensitive dye (Molecular Probes, Eugene, Oreg.) is added to each well (dye solution: 1 mg FLUO-3 AM, 440 uL DMSO and 440 ul 20% pluronic acid in DMSO, diluted 1:4, 50 ul diluted solution per well). Plates are covered with aluminum foil and incubated at 37° C. for 1-2 hours in an environment containing 5% CO[0334] 2. After the incubation the dye is emptied from the plates, and the cells are washed once with Krebs-Ringer HEPES (KRH) buffer (25 mM HEPES, 5 mM KCl, 0.96 mM NaH2PO4, 1 mM MgSO4, 2 mM CaCl2, 5 mM glucose, 1 mM probenecid, pH 7.4), and resuspended in KRH buffer.
  • Agonist (e.g., olvanil, capsaicin, or RTX)-induced calcium mobilization is monitored using either FLUOROSKAN ASCENT (Labsystems, Franklin, Mass.) or FLIPR (fluorometric imaging plate reader system, Molecular Devices, Sunnyvale, Calif.) instruments. Similarly, varying concentrations of the antagonists ruthenium red or capsazepine are added to cells concurrently with agonist (e.g., 25-50 nM capsaicin). For agonist-induced calcium responses, data obtained between 30 and 60 seconds after agonist application are used to generate the EC[0335] 50 values. KALEIDAGRAPH software (Synergy Software, Reading, Pa.) was utilized to fit the data to the equation:
  • y=a*(1/(1+(b/x)c)
  • to determine the EC[0336] 50 for the response. In this equation, y is the maximum fluorescence signal, x is the concentration of the agonist or antagonist, a is the Emax, b corresponds to the EC50 or IC50 value, and finally, c is the Hill coefficient.
  • Assay for Determination of Capsaicin Receptor Antagonist Effects [0337]
  • In order to measure the ability of a test compound to antagonize (inhibit) the response of cells expressing capsaicin receptors to capsaicin or other vanilloid agonist, the EC[0338] 50 Of capsaicin is first determined.
  • An additional 20 ul of KRH buffer and 1 ul DMSO is added to each well of cells, prepared as described above. 100 ul capsaicin in KRH buffer is automatically transferred by the FLIPR instrument to each well. An 8-point concentration response curve, with final capsaicin concentrations of 1 nM to 3 uM, is used to determine capsaicin EC[0339] 50.
  • Test compounds are dissolved in DMSO, diluted in 20 ul KRH buffer so that the final concentration of test compounds in the assay well is between 1 uM and 5 uM, and added to cells prepared as described above. The 96 well plates containing prepared cells and test compounds are incubated in the dark, at room temperature for 0.5-6 hours. It is important that the incubation not continue beyond 6 hours. Just prior to determining the fluorescence response, 100 ul capsaicin in KRH buffer at twice the EC[0340] 50 concentration determined from the concentration response curve is automatically added by the FLIPR instrument to each well of the 96 well plate for a final sample volume of 200 ul and a final capsaicin concentration equal to the EC50. The final concentration of test compounds in the assay wells is between 1 uM and 5 uM. Typically cells exposed to one EC50 of capsaicin exhibit a fluorescence response of about 10,000 Relative Fluorescence Units. Antagonists of the capsaicin receptor decrease this response by about 20%, preferably by about 50%, and most preferably by at least 80% as compared to matched control. The concentration of antagonist required to provide a 50% decrease is the EC50 for the antagonist (also referred to as the IC50)
  • Equilibrium binding parameters may be determined as described in Example 10. [0341]
  • Assay for Determination of Capsaicin Receptor Agonist Effects [0342]
  • The ability of a compound to act as an agonist of the capsaicin receptor may be determined by measuring the fluorescence response of cells expressing capsaicin receptors, using the methods described above, in the absence of capsaicin, RTX, or other known capsaicin receptor agonists. Compounds that cause cells to exhibit fluorescence above background are capsaicin receptor agonists. Highly preferred compounds of the invention are antagonists that are essentially free of agonist activity as demonstrated by the absence of detectable agonist activity in such an assay at compound concentrations below 4 nM, more preferably at concentrations below 10 uM and most preferably at concentrations less than or equal to 100 uM. [0343]
  • Example 12
  • NPY Y5 Receptor Binding Assay [0344]
  • The following assay is a standard assay for NPY Y5 (neuropeptide Y receptor 5) receptor binding that may be used to determine the affinity of compounds for the NPY Y5 receptor. Expression of a recombinant human Y5 receptor in cultured cells and receptor binding assays using of membranes prepared from such cells has been described previously, e.g. in U.S. Pat. No. 5,602,024 at columns 17-20. U.S. Pat. No. 5,602,024 is hereby incorporated by reference for its teachings regarding a recombinant human Y5 receptor, expression of this receptor in cultured cells, and receptor binding assays using membranes prepared from such cells. [0345]
  • Cell Culture [0346]
  • Baculovirus-infected Sf9 cells expressing recombinant human NPY Y5 receptors are harvested at 48 hrs. [0347]
  • Membrane Preparation [0348]
  • Sf9 cell pellets are resuspended in lysis buffer (20 mM Tris-HCL, 5 mM EDTA, 0.5 ug/ml leupeptin, 2 ug/ml Aprotinin, 200 uM PMSF, pH 7.4) and homogenized using a POLYTRON homogenizer (setting 3 for 25-30 seconds). The homogenate was centrifuged (536×g/10 minutes/4° C.) to pellet the nuclei. The supernatant containing isolated membranes are decanted to a clean centrifuge tube, centrifuged (48,000×g/30 minutes, 4° C.) and resuspended in 30 ml homogenization buffer. This centrifugation and resuspension step is repeated twice. The final pellet is resuspended in ice cold Dulbecco's PBS containing 5 mM EDTA and stored in aliquots at −80° C. until needed. The protein concentration of the resulting membrane preparation is measured using the Bradford Protein assay, as described in Example 3. By this measure, a 1-liter culture of cells typically yields 100-125 mg of total membrane protein. [0349]
  • Thawed Sf9 membranes are washed with PBS and resuspended by Dounce homogenization (tight pestle) in binding buffer (50 mM Tris-HCl, 5 mM KCl, 120 mM NaCl, 2 mM CaCl[0350] 2, 0.1% BSA, pH 7.4).
  • For competition binding analysis, membranes (10-25 ug) in 150 ul binding buffer are added to polypropylene tubes or 96-well deepwell plates containing [[0351] 125I]PYY (porcine, NEN, Boston, Mass.)/GTP. Final concentration of [125I]PYY is 30-35 pM/assay well; final concentration of GTP is 100 uM/well. Nonspecific binding is determined in the presence of 1 uM NPY (human, American Peptide Co., Sunnyvale, Calif.) and accounts for less than 10% of total binding. Test compounds at a concentration of 1-4 uM in 2 ul DMSO are added to the assay mixtures. Final assay volume is 250 ul. For competition analysis test compounds are added at concentrations ranging from 10−12 to 10−6 M. Typically 11 concentration points are collected per saturation binding curve. Following a 2-hour incubation at room temperature, the assay reactions are terminated by rapid vacuum filtration. Samples are filtered over presoaked (in polyethyleneimine for 2 hours prior to use) GF/C WHATMAN filters and rinsed 2 times with 5 mls cold binding buffer without BSA. Remaining bound radioactivity is quantified by gamma counting. Ki values may be determined by the method described in Example 10.
  • Preferred compounds of the invention exhibit 10-fold greater affinity for the capsaicin receptor than for the chimeric NPY Y5 receptor, more preferred compounds of the invention also exhibit 100-fold greater affinity for the capsaicin receptor than for the chimeric NPY Y5 receptor, and still more preferred compounds of the invention also exhibit 1000-fold greater affinity for the capsaicin receptor than for the chimeric NPY Y5 receptor. Most highly preferred compounds of the invention do not exhibit detectable binding at the NPY 5 receptor. [0352]
  • Example 13
  • Preparation of Radiolabeled Probe Compounds of the Invention [0353]
  • The compounds of the invention are prepared as radiolabeled probes by carrying out their synthesis using precursors comprising at least one atom that is a radioisotope. The radioisotope is preferably selected from of at least one of carbon (preferably [0354] 14C), hydrogen (preferably 3H), sulfur (preferably 35S), or iodine (preferably 125I). Such radiolabeled probes are conveniently synthesized by a radioisotope supplier specializing in custom synthesis of radiolabeled probe compounds. Such suppliers include Amersham Corporation, Arlington Heights, Ill.; Cambridge Isotope Laboratories, Inc. Andover, Mass.; SRI International, Menlo Park, Calif.; Wizard Laboratories, West Sacramento, Calif.; ChemSyn Laboratories, Lexena, KS; American Radiolabeled Chemicals, Inc., St. Louis, Mo.; and Moravek Biochemicals Inc., Brea, Calif.
  • Tritium labeled probe compounds are also conveniently prepared catalytically via platinum-catalyzed exchange in tritiated acetic acid, acid-catalyzed exchange in tritiated trifluoroacetic acid, or heterogeneous-catalyzed exchange with tritium gas. Such preparations are also conveniently carried out as a custom radiolabeling by any of the suppliers listed in the preceding paragraph using the compound of the invention as substrate. In addition, certain precursors may be subjected to tritium-halogen exchange with tritium gas, tritium gas reduction of unsaturated bonds, or reduction using sodium borotritide, as appropriate. [0355]
  • Examples 14a and 14b
  • Preparation of Radiolabeled Aryl Piperazine Capsaicin Receptor Ligands [0356]
  • Example 14a
  • [0357]
    Figure US20040176443A1-20040909-C00202
  • Example 14b
  • [0358]
    Figure US20040176443A1-20040909-C00203
  • Example 15
  • Receptor Autoradiography [0359]
  • Receptor autoradiography (receptor mapping) is carried out in vitro as described by Kuhar in sections 8.1.1 to 8.1.9 of Current Protocols in Pharmacology (1998) John Wiley & Sons, New York, using radiolabeled compounds of the invention prepared as described in the preceding Example. [0360]
  • Example 16
  • Additional Aspects of Preferred Compounds of the Invention [0361]
  • The most preferred compounds of the invention are suitable for pharmaceutical use in treating human patients. Accordingly, such preferred compounds are non-toxic. They do not exhibit single or multiple dose acute or long-term toxicity, mutagenicity (e.g., as determined in a bacterial reverse mutation assay such as an Ames test), teratogenicity, tumorogenicity, or the like, and rarely trigger adverse effects (side effects) when administered at therapeutically effective dosages. [0362]
  • Preferably, administration of such preferred compounds of the invention at certain doses (i.e., doses yielding therapeutically effective in vivo concentrations or preferably doses of 0.01, 0.05, 0.1, 0.5, 1, 5, 10, 40, or 50 mg/kg administered parenterally or prefrerably orally) does not result in prolongation of heart QT intervals (i.e., as determined by electrocardiography, e.g., in guinea pigs, minipigs or dogs). When administered daily for 5 or preferably ten days, such doses of such preferred compounds also do not cause liver enlargement resulting in an increase of liver to body weight ratio of more than 100%, preferably not more than 75% and more preferably not more than 50% over matched controls in laboratory rodents (e.g., mice or rats). In another aspect such doses of such preferred compounds also preferably do not cause liver enlargement resulting in an increase of liver to body weight ratio of more than 50%, preferably preferably not more than 25%, and more preferably not more than 10% over matched untreated controls in dogs or other non-rodent mammals. [0363]
  • In yet another aspect such doses of such preferred compounds also preferably do not promote the release of liver enzymes (e.g., ALT, LDH, or AST) from hepatocytes in vivo. Preferably such doses do not elevate serum levels of such enzymes by more than 100%, preferably not by more than 75% and more preferably not by more than 50% over matched untreated controls in vivo in laboratory rodents. Similarly, concentrations (in culture media or other such solutions that are contacted and incubated with cells in vitro; equivalent to two, fold, preferably five-fold, and most preferably ten-fold the minimum in vivo therapeutic concentration do not cause release of any of such liver enzymes from hepatocytes in vitro into culture medium above baseline levels seen in media from untreated cells. [0364]
  • Because side effects are often due to undesirable receptor activation or antagonism, preferred compounds of the invention exert their receptor-modulatory effects with high selectivity. This means that they do not bind to certain other receptors (other than capsaicin receptors) with high affinity, but rather only bind to, activate, or inhibit the activity of such other receptors with affinity constants of greater than 100 nanomolar, preferably greater than 1 micromolar (uM), more preferably greater than 10 uM and most preferably greater than 100 uM. Such receptors preferably are selected from the group including ion channel receptors, including sodium ion channel receptors, neurotransmitter receptors such as alpha- and beta-adrenergic receptors, muscarinic receptors (particularly m1, m2, and m3 receptors), dopamine receptors, and metabotropic glutamate receptors; and also include histamine receptors and cytokine receptors, e.g., interleukin receptors, particularly IL-8 receptors. The group of other receptors to which preferred compounds do not bind with high affinity also includes GABA[0365] A receptors, bioactive peptide receptors (including NPY and VIP receptors), neurokinin receptors, bradykinin receptors (e.g., BK1 receptors and BK2 receptors), and hormone receptors (including androgen receptors, thyrotropin releasing hormone receptors and melanocyte-concentrating hormone receptors).
  • Example 16a
  • Sodium Ion Channel and Anti-Androgen Activity Criteria [0366]
  • Preferred compounds of the invention do not exhibit significant activity as sodium ion channel blockers, exhibiting less than 15 percent inhibition, and more preferably less than 10 percent inhibition, of sodium channel specific ligand (e.g., batrachotoxin, tetrodotoxin, or saxitoxin) binding when present at a concentration of 4 uM or less. [0367]
  • Preferred compounds of the invention do not exhibit significant androgen bioactivity, more preferably they do not exhibit significant androgen antagonist activity, e.g., in vivo, in a Hershberger assay, or in vitro, in an assay such as that described by Nellemann et al., [0368] Toxicology 2001, 163(1):29-38. Preferred compounds of the invention exhibit less than a 15% inhibition, more preferably less than a 10%, and most preferably less than 5% inhibition of androgen receptor activation in this in vitro assay when present at concentrations of 4 uM or less.
  • By significant activity is meant results varying from control at the p<0.1 level or more preferably at the p<0.05 level of significance as measured using a standard parametric assay of statistical significance such as a student's T test. [0369]
  • Example 16b
  • Microsomal In Vitro Half-Life [0370]
  • Compound half-life values (t[0371] 1/2 values) may be determined via the following standard liver microsomal half-life assay. Pooled Human liver microsomes are obtained from XenoTech LLC, 3800 Cambridge St., Kansas City, Kansas 66103 (catalog # H0610). Such liver microsomes may also be obtained, e.g., from In Vitro Technologies, Baltimore, Md. 21227, or from Tissue Transformation Technologies, Edison, N.J. 08837. Reactions are preformed as follows:
  • Reagents: [0372]
  • Phosphate buffer: 19 mL 0.1 M NaH[0373] 2PO4, 81 mL 0.1 Na2HPO4, adjusted to pH 7.4 with H3PO4.
  • CoFactor Mixture: 16.2 mg NADP, 45.4 mg Glucose-6-phosphate in 4 mL 100 mM MgCl[0374] 2.
  • Glucose-6-phosphate dehydrogenase: 214.3 ul glucose-6-phosphate dehydrogenase suspension (Boehringer-Manheim catalog no. 0737224, distributed by Roche Molecular Biochemicals, Indianapolis, Ind. 46250) is diluted into 1285.7 ul distilled water. [0375]
  • Starting Reaction Mixture: 3 mL CoFactor Mixture, 1.2 mL Glucose-6-phosphate dehydrogenase. [0376]
  • Reaction: [0377]
  • 6 test reactions are prepared, each containing 25 ul microsomes, 5 ul of a 100 uM solution of test compound, and 399 ul 0.1 M phosphate buffer. A seventh reaction is prepared as a positive control containing 25 ul microsomes, 399 ul 0.1 M phosphate buffer, and 5 ul of a 100 uM solution of a compound with known metabolic properties (e.g. DIAZEPAM or CLOZEPINE). Reactions are preincubated at 39° C. for 10 minutes. 71 ul Starting Reaction Mixture is added to 5 of the 6 test reactions and to the positive control, 71 ul 100 mM MgCl[0378] 2 is added to the sixth test reaction, which is used as a negative control. At each time point (0, 1, 3, 5, and 10 minutes) 75 ul of each reaction mix is pipetted into a well of a 96-well deep-well plate containing 75 ul ice-cold acetonitrile. Samples are vortexed and centrifuged 10 minutes at 3500 rpm (Sorval T 6000D centrifuge, H1000B rotor). 75 ul of supernatant from each reaction is transferred to a well of a 96-well plate containing 150 ul of a 0.5 uM solution of a compound with a known LCMS profile (internal standard) per well. LCMS analysis of each sample is carried out and the amount of unmetabolized test compound is measured as AUC, compound concentration vs time is plotted, and the t1/2 value of the test compound is extrapolated.
  • Preferred compounds of the invention exhibit in vitro t[0379] 1/2 values of greater than 10 minutes and less than 4 hours. Most preferred compounds of the invention exhibit in vitro half-life values of between 30 minutes and 1 hour in human liver microsomes.
  • Example 16c
  • MDCK Toxicity Assay [0380]
  • Compounds causing acute cytotoxicity will produce a substantial decrease of ATP production by Madin Darby canine kidney (MDCK) cells in the following assay. Preferred compounds of the invention will not. [0381]
  • MDCK cells, ATCC no. CCL-34 (American Type Culture Collection, Manassas, Va.) are maintained in sterile conditions following the instructions in the ATCC production information sheet. The PACKARD, (Meriden, Conn.) ATP-LITE-M Luminescent ATP detection kit, product no. 6016941, allows measurement ATP production in MDCK cells. [0382]
  • Prior to assay 1 ul of test compound or control sample is pipetted into PACKARD (Meriden, Conn.) clear bottom 96-well plates. Test compounds and control samples are diluted in DMSO to give final concentration in the assay of 10 micromolar, 100 micromolar, or 200 micromolar. Control samples are drug or other compounds having known toxicity properties. [0383]
  • Confluent MDCK cells are trypsinized, harvested, and diluted to a concentration of 0.1×10[0384] 6 cells/ml with warm (37° C.) VITACELL Minimum Essential Medium Eagle (ATCC catalog # 30-2003). 100 ul of cells in medium is pipetted into each of all but five wells of each 96-well plate. Warm medium without cells (100 ul) is pipetted in the remaining five wells of each plate to provide standard curve control wells. These wells, to which no cells are added, are used to determine the standard curve. The plates are then incubated at 37° C. under 95% O2, 5% CO2 for 2 hours with constant shaking. After incubation, 50 ul of mammalian cell lysis solution is added per well, the wells are covered with PACKARD TOPSEAL stickers, and plates are shaken at approximately 700 rpm on a suitable shaker for 2 minutes.
  • During the incubation, PACKARD ATP LITE-M reagents are allowed to equilibrate to room temperature. Once equilibrated the lyophilized substrate solution is reconstituted in 5.5 mls of substrate buffer solution (from kit). Lyophilized ATP standard solution is reconstituted in deionized water to give a 10 mM stock. For the five control wells, 10 ul of serially diluted PACKARD standard is added to each of the five standard curve control wells to yield a final concentration in each subsequent well of 200 nM, 100 nM, 50 nM, 25 nM, and 12.5 nM. [0385]
  • PACKARD substrate solution (50 ul) is added to all wells. Wells are covered with PACKARD TOPSEAL stickers, and plates are shaken at approximately 700 rpm on a suitable shaker for 2 minutes. A white PACKARD sticker is attached to the bottom of each plate and samples are dark adapted by wrapping plates in foil and placing in the dark for 10 minutes. Luminescence is then measured at 22° C. using a luminescence counter, e.g. PACKARD TOPCOUNT Microplate Scintillation and Luminescense Counter or TECAN SPECTRAFLUOR PLUS. [0386]
  • Luminescence values at each drug concentration are compared to the values computed from the standard curve for that concentration. Preferred test compounds exhibit luminescence values 80% or more of the standard, or preferably 90% or more of the standard, when a 10 micromolar (uM) concentration of the test compound is used. When a 100 uM concentration of the test compound is used, preferred test compounds exhibit luminescence values 50% or more of the standard, or more preferably 80% or more of the standard. Luminescence values less than 50% of the standard indicate a substantial decrease of ATP production. [0387]
  • Example 18
  • Animal Models for Determining Pain Relief and Sedation [0388]
  • The following experimental protocols can be used to determine the degree of pain relief and sedation provided by compounds of the invention, e.g., in comparison to pain relief and sedation provided by morphine or pain relief by ibuprofen. [0389]
  • Example 18a
  • CFA Arthritis Model [0390]
  • Male SD rats are injected with 200 ml of CFA (0.1 mg heat killed and dried [0391] M. tuberculosis/ml) in the hind paw (100 ml on the dorsal and 100 ml on the plantar surface of the paw) essentially as described by Bertorelli R, Corradini L, Rafiq K, Tupper J, Calo G, Ongini E., Br J. Pharmacol. 1999 128(6):1252-8 and by Stein C, Millan M J, Herz A. Pharmacol Biochem Behav. 1988 31(2):455-51.
  • Rats are tested for thermal (as described by Hargreaves K, Dubner R, Brown F, Flores C, Joris J. [0392] Pain. 1988 32(1):77-88) and mechanical (as described by Tal M, Eliav E. Pain. 1996 March;64(3):511-8) sensitivities on days 5, 6, and 7. Baseline data should be obtained for each animal prior to CFA injection.
  • On day 7, animals are treated orally with a compound of the invention, morphine or vehicle (2% vitamin E-TPGS) 1 hour prior to testing. Note: an oral dose of 5 mg/kg morphine has sedative effects. [0393]
  • Results are conveniently expressed as % of Maximum Potential Efficacy (MPE). 0% MPE is defined as analgesic effect of vehicle, 100% MPE is defined as an animal's return to baseline level of thermal or mechanical sensitivity. [0394]
  • Example 18b
  • Mechanical Allodynia [0395]
  • This assay determines the effectiveness of compounds of Formulae I-IX and Formulae A-F in relieving at least one of the symptoms in an in vivo model of pain produced by spinal nerve ligation, namely mechanical allodynia. [0396]
  • Tactile allodynia is induced in rats using the procedures described by Bennet and Xie, Pain 1988, 33:87-107. Rats are anesthetized, e.g., with an intraperitoneal dose of pentobarbital sodium (65 mg/kg) with additional doses of anesthetic given as needed. The lateral aspect of each rat's hind limb is shaved and disinfected. Using aseptic technique, an incision is made on the lateral aspect of the hind limb at the mid-thigh level. The biceps femoris is bluntly dissected to expose the sciatic nerve. On one hind limb of each rat, four loosely tied ligatures are made around the sciatic nerve approximately 1-2 millimeters apart. On the other side of each rat, an identical dissection is performed except that the sciatic nerve is not ligated. The muscle is closed with a continuous suture pattern, and the skin is closed with wound clips or sutures. [0397]
  • Mechanical sensitivity is assessed using a procedure described by Chaplan et al. J. Neurosci. Methods 1994, 53:55-63. A series of Von Frey filaments of varying rigidity strength (typically eight filaments in the series) are applied to the plantar surface of the hind paw ipsilaterial to the ligations with just enough force to bend the filament. The filaments are held in this position for no more than three seconds or until a positive allodynic response is displayed by the rat. A positive allodynic response consists of lifting the affected paw followed immediately by licking or shaking of the paw. The order and frequency with which the individual filaments are applied are determined by using Dixon up-down method. Testing is initiated with the middle hair of the series with subsequent filaments being applied in consecutive fashion, ascending or descending, depending on whether a negative or positive response, respectively, is obtained with the initial filament. [0398]
  • Certain preferred compounds of Formulae I-IX and Formulae A-F are effective in reversing mechanical allodynia-like symptoms (i.e., rats treated with effective amounts of such compounds will require stimulation with a Von Frey filament of higher rigidity strength to provoke a positive allodynic response as compared to control untreated or vehicle treated rats) when tested by this method. [0399]
  • Example 18c
  • Cold Allodynia [0400]
  • This assay determines the effectiveness of compounds in relieving one of the symptoms of neuropathic pain produced by unilateral mononeuropathy, namely cold allodynia. [0401]
  • Unilateral mononeuropathy is produced in rats using the Chronic Constriction Injury model performed essentially as described by Bennet and Xie, Pain 1988, 33:87-107. Rats are anesthetized. The lateral aspect of each rat's hind limb is shaved and disinfected. Using aseptic technique, an incision is made on the lateral aspect of the hind limb at the mid-thigh level. The biceps femoris is bluntly dissected to expose the sciatic nerve. On one hind limb of each rat, four loosely tied ligatures are made around the sciatic nerve approximately 1-2 millimeters apart. On the other side of each rat, an identical dissection is performed except that the sciatic nerve is not ligated. The muscle is closed with a continuous suture pattern, and the skin is closed with wound clips or sutures. [0402]
  • Rats demonstrating unilateral mononeuropathy are assessed for acute and chronic cold allodynia sensitivity. Each rat is placed individually into a chamber with a metal plate about 6 cm from the bottom. This chamber is filled with ice water to a depth of about 2.5 cm above the metal plate, with the temperature of the bath maintained at about zero to four degrees C. throughout the experiment. A timer is started, and the rat's response latency is measured to the nearest tenth of a second. A “response” is defined as a rapid withdrawal of the ligated hindpaw completely out of the water while the animal is stationary and not pivoting. An exaggerated limp while the animal is walking is not scored as a response. Maximum immersion time is 20 seconds with a 20-minute interval between trials. The screening criteria are 1) the average of two trials is less than or equal to 13 seconds, and 2) there is consistency across the two trial scores. Animals are screened for hypersensitivity to cold on post-surgery days 4 through 10, and selected for inclusion in drug-response studies based on the criteria described above. The pre-dose screening values are used as the animal's baseline cold allodynia scores. For acute studies, the animals are tested for cold allodynia at 1, 3, and sometimes 5 hours post-dose. [0403]
  • Although this acute cold allodynia assay is generally less preferred for demonstrating efficacy of compounds of the invention, when tested in this assay, certain preferred compounds of Formulae I-IX and Formulae A-F demonstrate anti-allodynic effects (increases in response latency) at doses of less than 50 mg/kg. [0404]
  • Example 18d
  • Mechanical Hyperalgesia [0405]
  • This assay determines the effectiveness of compounds in relieving one of the symptoms of neuropathic pain produced by unilateral mononeuropathy, namely mechanical hyperalgesia. [0406]
  • A chronic constriction injury is produced by loosely ligating the right common sciatic nerve as described by Bennet and Xie, Pain 1988, 33:87-107. The left common sciatic nerve is visualized, but not manipulated to produce sham conditions. [0407]
  • The rats having a chronic constriction injury are assessed for mechanical hyperalgesia to a pin-prick stimulus as described by Koch et al. Analgesia 1996, 2(3), 157-164. Rats are placed in individual compartments of a cage with a warmed, perforated metal floor. Hindpaw withdrawal duration is measured after a mild pinprick to the plantar surface of the ligated and sham hindpaws. [0408]
  • Preferred compounds of the invention produce a reduction of mechanical hyperalgesia (i.e., a reduction in the duration of hindpaw withdrawal) elicited by a pin-prick stimulus in rats with a chronic constriction injury at doses of 50 mg/kg or less when tested by this method. [0409]
  • Example 18e
  • Thermal Hyperalgesia [0410]
  • This assay determines the effectiveness of compounds in relieving one of the symptoms of neuropathic pain produced by unilateral mononeuropathy, namely thermal hyperalgesia. [0411]
  • Rats having had surgery as described in Example 18d are assessed for thermal hyperalgesia sensitivity at least 10 days post-surgery. The rats are placed beneath inverted cages upon an elevated glass platform and a radiant heat source beneath the glass is aimed at the plantar hindpaw. [0412]
  • The duration of time before the hindpaw is withdrawn from the floor is measured to the nearest tenth of a second. The cutoff time for the heat stimulus is 20 seconds, and the light is calibrated such that this stimulus duration does not burn or blister the skin. Preferably about four latency measurements are taken for each hindpaw in each test session, alternating left and right hindpaws, with 5-minute intervals between tests. The times to withdrawal of each side are averaged and a difference score is obtained. [0413]
  • Preferred compounds of the invention produce an increase in the average time to withdrawal after oral administration of 50 mg/kg or less in this model. [0414]
  • Example 18f
  • Sedation [0415]
  • Sedation may be determined using the method described by Fitzgerald et al., [0416] Toxicology 1988, 49(2-3)433-9. Preferred compounds of the invention do not produce reproducible or significant sedation at intravenous doses of less than 25 mg/kg (preferably less than 10 mg/kg) or at oral doses of less than 140 mg/kg (preferably less than 50 mg/kg).

Claims (198)

What is claimed is:
1. A compound of the formula:
Figure US20040176443A1-20040909-C00204
or a pharmaceutically acceptable salt thereof,
wherein:
A is absent or is selected from the group consisting of O, S, NRA, CRBRB′, NRACRBRB′, CRBRB′NRA, —CRA═CRB—, and C3H4; where RA, RB, and RB′ are independently selected at each occurrence from hydrogen or alkyl;
Z is oxygen or sulfur;
R1 and R2 independently represent hydrogen or alkyl;
R3 and R4 are independently selected at each occurrence from the group consisting of hydrogen; halogen; hydroxy; amino; cyano; nitro; —COOH; —CHO, optionally substituted alkyl; optionally substituted alkenyl; optionally substituted alkynyl; optionally substituted alkoxy; optionally substituted mono or dialkylamino; optionally substituted alkylthio; optionally substituted alkyl ketone; optionally substituted alkylester; optionally substituted alkylsulfinyl; optionally substituted alkylsulfonyl; optionally substituted mono- or di-alkylcarboxamide; optionally substituted —S(O)nNHalkyl; optionally substituted —S(O)nN(alkyl)(alkyl); optionally substituted —NHC(═O) alkyl; optionally substituted —NC(═O)(alkyl)(alkyl); optionally substituted —NHS(O)nalkyl; optionally substituted —NS(O)n(alkyl)(alkyl); optionally substituted saturated or partially unsaturated heterocycloalkyl of from 5 to 8 atoms, which saturated or partially unsaturated heterocycloalkyl contains 1, 2, or 3 heteroatoms selected from N, O, and S; optionally substituted aryl having from 1 to 3 rings; or optionally substituted heteroaryl, said heteroaryl having from 1 to 3 rings, 5 to 8 ring members in each ring and, in at least one of said rings, from 1 to about 3 heteroatoms per ring selected from the group consisting of N, O, and S;
or any two
R3 and R4 not attached to the same carbon may be joined to form an optionally substituted aryl ring; a saturated or partially unsaturated carbocyclic ring of from 5 to 8 members, which carbocyclic ring is optionally substituted; or a saturated, partially unsaturated, or aromatic heterocyclic ring of from 5 to 8 members, which heterocyclic ring is optionally substituted and contains 1, 2, or 3 heteroatoms selected from N, O, and S; and
Ar1 and Ar2 are the same or different and independently represent optionally substituted cycloalkyl; an optionally substituted heterocycloalkyl ring of from 5 to 8 atoms, which heterocyloalkyl ring contains 1, 2, or 3 heteroatoms selected from N, O, and S; optionally substituted aryl having from 1 to 3 rings; or optionally substituted heteroaryl, said heteroaryl having from 1 to 3 rings, 5 to 8 ring members in each ring and, in at least one of said rings, from 1 to about 3 heteroatoms per ring selected from the group consisting of N, O, and S, and
n is independently chosen at each occurrence from 0, 1, and 2.
2. A compound or salt according to claim 1, wherein:
R3 and R4 are independently chosen at each occurrence from the group consisting of hydrogen, halogen, cyano, nitro, haloalkyl, haloalkoxy, hydroxy, amino, alkyl substituted with 0-2 R6, alkenyl substituted with 0-2 R6; alkynyl substituted with 0-2 R6; alkoxy substituted with 0-2 R6, —NH(alkyl) substituted with 0-2 R6, —N(alkyl)(alkyl) where each alkyl is independently substituted with 0-2 R6, —XR7, and Y;
or any two
R3 and R4 not attached to the same carbon may be joined to form an aryl ring substituted with 0-3 R6, a saturated or partially unsaturated carbocyclic ring of from 5 to 8 members, which carbocyclic ring is substituted with 0-2 R6, or a saturated, partially unsaturated, or aromatic heterocyclic ring of from 5 to 8 members, which heterocyclic ring is substituted with 0-2 R6 and contains 1, 2, or 3 heteroatoms selected from N, O, and S;
Ar1 and Ar2 may be the same or different and are selected from the group consisting of cyclohexyl, cyclopentyl, piperidinyl, piperazinyl, phenyl, pyrrolyl, furanyl, thienyl, pyrazolyl, imidazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, triazolyl, tetrazolyl, pyridyl, pyrimidyl, pyrazinyl, benzimidazolyl, naphthyl, indolyl, isoindolyl, benzofuranyl, isobenzofuranyl, benzo[b]thiophenyl, benz[d]isoxazolyl, quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, and quinoxalinyl, each of which is optionally mono-, di-, or trisubstituted with R5; or
Ar1 and Ar2 may be the same or different and represent a bicyclic oxygen-containing group of the formula:
Figure US20040176443A1-20040909-C00205
 optionally mono-, di-, or trisubstituted with R5, where L represents point of attachment and may be at any point on the benzene ring, and the oxygen-containing ring of the bicyclic oxygen-containing group consists of from 5 to 8 ring atoms, contains 1 or 2 oxygen atoms and remaining ring atoms are carbon;
R5 is independently selected at each occurrence from the group consisting of halogen, cyano, nitro, haloalkyl, haloalkoxy, hydroxy, amino, alkyl substituted with 0-2 R6, alkenyl substituted with 0-2 R6, alkynyl substituted with 0-2 R6, alkoxy substituted with 0-2 R6, —NH(alkyl) substituted with 0-2 R6, —N(alkyl)(alkyl) where each alkyl is independently substituted with 0-2 R6, —XR7, and Y;
R6 is independently selected at each occurrence from the group consisting of halogen, hydroxy, cyano, alkyl, alkoxy, —NH(alkyl), —N(alkyl)(alkyl), —S(O)n(alkyl), haloalkyl, haloalkoxy, CO(alkyl), CONH(alkyl), CON(alkyl1)(alkyl2) where alkyl1 and alkyl2 may be joined to form a heterocycloalkyl ring of from 5 to 8 ring atoms and containing 1, 2, or 3 heteroatoms selected from N, O, and S, —XR7, and Y;
X is independently selected at each occurrence from the group consisting of —CH2—, —CHR8—, —O—, —S(O)n—, —NH—, —NR8—, —C(═O)—, —C(═O)O—, —C(═O)NH—, —C(═O)NR8—, —S(O)nNH—, —S(O)nNR8—, NHC(═O)—, —NR8C(═O)—, —NHS(O)n—, and —NR8S(O)n—;
R7 and R8 are independently selected at each occurrence from hydrogen, and straight, branched, and cyclic alkyl groups, and (cycloalkyl)alkyl groups, said straight, branched, and cyclic alkyl groups, and (cycloalkyl)alkyl groups consisting of 1 to 8 carbon atoms, and containing zero or one or more double or triple bonds, each of which 1 to 8 carbon atoms may be further substituted with one or more substituent(s) independently selected from oxo, hydroxy, halogen, amino, cyano, nitro, haloalkyl, haloalkoxy, —O(alkyl),
 —NH(alkyl), —N(alkyl)(alkyl), —NHC(O)(alkyl), —N(alkyl)C(O)(alkyl), —NHS(O)n(alkyl),
 —S(O)n(alkyl), —S(O)nNH(alkyl), —S(O)nN (alkyl3)(alkyl4) where alkyl3 and alkyl4 may be joined to form a heterocycloalkyl ring consisting of from 5 to 8 ring atoms and containing 1, 2, or 3 heteroatoms selected from N, O, and S, and Y′;
Y and Y′ are independently selected at each occurrence from 3- to 8-membered carbocyclic or heterocyclic groups which are saturated, unsaturated, or aromatic, which may be further substituted with one or more substituents independently selected from halogen, oxo, hydroxy, amino, nitro, cyano, alkyl, alkoxy, haloalkyl, haloalkoxy, mono- or dialkylamino, and alkylthio;
 wherein said 3- to 8-memberered heterocyclic groups contain one or more heteroatom(s) independently selected from N, O, and S; and
n is independently chosen at each occurrence from 0, 1, and 2.
3. A compound or salt according to claim 1, wherein:
RA, RB, and RB′ are independently selected at each occurrence from hydrogen and C1-6alkyl;
R3 and R4 are independently chosen at each occurrence from the group consisting of hydrogen, halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl substituted with 0-2 R6, C2-6alkenyl substituted with 0-2 R6; C2-6alkynyl substituted with 0-2 R6; C1-6alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6, —N(C1-6alkyl)(C1-6alkyl) where each C1-6alkyl is independently substituted with 0-2 R6,
 —XR7, and Y;
or any two
R3 and R4 not attached to the same carbon may be joined to form an aryl ring substituted with 0-3 R6, a saturated or partially unsaturated carbocyclic ring of from 5 to 8 members, which carbocyclic ring is substituted with 0-2 R6, or a saturated, partially unsaturated, or aromatic heterocyclic ring of from 5 to 8 members, which heterocyclic ring is substituted with 0-2 R6 and contains 1, 2, or 3 heteroatoms selected from N, O, and S;
Ar1 and Ar2 may be the same or different and are selected from the group consisting of cyclohexyl, cyclopentyl, piperidinyl, piperazinyl, phenyl, pyrrolyl, furanyl, thienyl, pyrazolyl, imidazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, triazolyl, tetrazolyl, pyridyl, pyrimidyl, pyrazinyl, benzimidazolyl, naphthyl, indolyl, isoindolyl, benzofuranyl, isobenzofuranyl, benzo[b]thiophenyl, benz[d]isoxazolyl, quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, and quinoxalinyl, each of which is optionally mono-, di-, or trisubstituted with R5; or
Ar1 and Ar2 may be the same or different and represent a bicyclic oxygen-containing group of the formula:
Figure US20040176443A1-20040909-C00206
 optionally mono-, di-, or trisubstituted with R5, where L represents point of attachment and may be at any point on the benzene ring, and the oxygen-containing ring of the bicyclic oxygen-containing group consists of from 5 to 8 ring atoms, contains 1 or 2 oxygen atoms and remaining ring atoms are carbon;
R5 is independently selected at each occurrence from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl substituted with 0-2 R6, C2-6alkenyl substituted with 0-2 R6, C2-6alkynyl substituted with 0-2 R6, C1-6alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6, —N(C1-6alkyl)(C1-6alkyl) where each C1-6alkyl is independently substituted with 0-2 R6, —XR7, and Y;
R6 is independently selected at each occurrence from the group consisting of halogen, hydroxy, cyano, C1-4alkyl, C1-4alkoxy, —NH(C1-4alkyl), —N(C1-4alkyl)(C1-4alkyl), —S(O)n(C1-4alkyl), halo(C1-4)alkyl, halo(C1-4)alkoxy, CO(C1-4alkyl), CONH(C1-4alkyl), CON(C1-4alkyl1)(C1-4alkyl2) where alkyl1 and alkyl2 may be joined to form a heterocycloalkyl ring of from 5 to 8 ring atoms and containing 1, 2, or 3 heteroatoms selected from N, O, and S, —XR7, and Y;
X is independently selected at each occurrence from the group consisting of —CH2—, —CHR8—, —O—, —S(O)—, —NH—, —NR8—, —C(═O)—, —C(═O)O—, —C(═O)NH—, —C(═O)NR8—, —S(O)nNH—, —S(O)nNR8—, NHC(═O)—, —NR8C(═O)—, —NHS(O)n—, and —NR8S(O)n—;
R7 and R8 are independently selected at each occurrence from hydrogen, and straight, branched, and cyclic alkyl groups, and (cycloalkyl)alkyl groups, said straight, branched, and cyclic alkyl groups, and (cycloalkyl)alkyl groups consisting of 1 to 8 carbon atoms, and containing zero or one or more double or triple bonds, each of which 1 to 8 carbon atoms may be further substituted with one or more substituent(s) independently selected from oxo, hydroxy, halogen, amino, cyano, nitro, haloalkyl, haloalkoxy, —O(C1-4alkyl), —NH(C1-4alkyl), —N(C1-4alkyl)(C1-4alkyl) —NHC(O)(C1-4alkyl), —N(C1-4alkyl)C(O)(C1-4alkyl), —NHS(O)n(C1-4alkyl), —S(O)n(C1-4alkyl), —S(O)nNH(C1-4alkyl), —S(O)nN(C1-4alkyl3)(C1-4alkyl4) where C1-4alkyl3 and C1-4alkyl4 may be joined to form a heterocycloalkyl ring consisting of from 5 to 8 ring atoms and containing 1, 2, or 3 heteroatoms selected from N, O, and S, and Y′;
Y and Y′ are independently selected at each occurrence from 3- to 8-membered carbocyclic or heterocyclic groups which are saturated, unsaturated, or aromatic, which may be further substituted with one or more substituents independently selected from halogen, oxo, hydroxy, amino, nitro, cyano, C1-4alkyl, C1-4alkoxy, halo(C1-4)alkyl, halo(C1-4)alkoxy, mono- or di(C1-4)alkylamino, and C1-4alkylthio;
 wherein said 3- to 8-memberered heterocyclic groups contain one or more heteroatom(s) independently selected from N, O, and S; and
n is independently chosen at each occurrence from 0, 1, and 2.
4. A compound of the formula:
Figure US20040176443A1-20040909-C00207
or a pharmaceutically acceptable salt thereof, wherein:
A is absent or is selected from the group consisting of O, S, NRA, CRBRB′, NRACRBRB′, CRBRB′NRA, —CRA═CRB—, and C3H4; where RA, RB, and RB′ are independently selected at each occurrence from hydrogen or alkyl;
Z is oxygen or sulfur;
R3 and R4 are independently selected at each occurrence from the group consisting of hydrogen; halogen; hydroxy; amino; cyano; nitro; —COOH; —CHO, optionally substituted alkyl; optionally substituted alkenyl; optionally substituted alkynyl; optionally substituted alkoxy; optionally substituted mono or dialkylamino; optionally substituted alkylthio; optionally substituted alkyl ketone; optionally substituted alkylester; optionally substituted alkylsulfinyl; optionally substituted alkylsulfonyl; optionally substituted mono- or di-alkylcarboxamide; optionally substituted —S(O)nNHalkyl; optionally substituted —S(O)nN(alkyl)(alkyl); optionally substituted —NHC(═O) alkyl; optionally substituted —NC(═O)(alkyl)(alkyl); optionally substituted —NHS(O)nalkyl; optionally substituted —NS(O)n(alkyl)(alkyl); optionally substituted saturated or partially unsaturated heterocycloalkyl of from 5 to 8 atoms, which saturated or partially unsaturated heterocycloalkyl contains 1, 2, or 3 heteroatoms selected from N, O, and S; optionally substituted aryl having from 1 to 3 rings; and optionally substituted heteroaryl, said heteroaryl having from 1 to 3 rings, 5 to 8 ring members in each ring and, in at least one of said rings, from 1 to about 3 heteroatoms per ring selected from the group consisting of N, O, and S;
or any two
R3 and R4 not attached to the same carbon may be joined to form an optionally substituted aryl ring, a saturated or partially unsaturated carbocyclic ring of from 5 to 8 members, which carbocyclic ring is optionally substituted, or a saturated, partially unsaturated, or aromatic heterocyclic ring of from 5 to 8 members, which heterocyclic ring is optionally substituted and contains 1, 2, or 3 heteroatoms selected from N, O, and S;
Ar1 and Ar2 may be the same or different and are selected from the group consisting of cyclohexyl, cyclopentyl, piperidinyl, piperazinyl, phenyl, pyrrolyl, furanyl, thienyl, pyrazolyl, imidazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, triazolyl, tetrazolyl, pyridyl, pyrimidyl, pyrazinyl, benzimidazolyl, naphthyl, indolyl, isoindolyl, benzofuranyl, isobenzofuranyl, benzo[b]thiophenyl, benz[d]isoxazolyl, quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, and quinoxalinyl, each of which is optionally mono-, di-, or trisubstituted with R5; or
Ar1 and Ar2 may be the same or different and represent a bicyclic oxygen-containing group of the formula:
Figure US20040176443A1-20040909-C00208
 optionally mono-, di-, or trisubstituted with R5, where L represents point of attachment and may be at any point on the benzene ring, and the oxygen-containing ring of the bicyclic oxygen-containing group consists of from 5 to 8 ring atoms, contains 1 or 2 oxygen atoms and remaining ring atoms are carbon;
R5 is independently selected at each occurrence from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl substituted with 0-2 R6, C2-6alkenyl substituted with 0-2 R6, C2-6alkynyl substituted with 0-2 R6, C1-6alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6, —N(C1-6alkyl)(C1-6alkyl) where each C1-6alkyl is independently substituted with 0-2 R6, —XR7, and Y;
R6 is independently selected at each occurrence from the group consisting of halogen, hydroxy, cyano, C1-4alkyl, C1-4alkoxy, —NH(C1-4alkyl), —N(C1-4alkyl)(C1-4alkyl), —S(O)n(C1-4alkyl) halo(C1-4)alkyl, halo(C1-4)alkoxy, CO(C1-4alkyl), CONH(C1-4alkyl), CON(C1-4alkyl1)(C1-4alkyl2) where alkyl1 and alkyl2 may be joined to form a heterocycloalkyl ring of from 5 to 8 ring atoms and containing 1, 2, or 3 heteroatoms selected from N, O, and S, —XR7, and Y;
X is independently selected at each occurrence from the group consisting of —CH2—, —CHR8—, —O—, —S(O)n—, —NH—, —NR8—, —C(═O)—, —C(═O)O—, —C(═O)NH—, —C(═O)NR8—, —S(O)nNH—, —S(O)nNR8—, NHC(═O)—, —NR8C(═O)—, —NHS(O)n—, and —NR8S(O)n—;
R7 and R8 are independently selected at each occurrence from hydrogen, and straight, branched, and cyclic alkyl groups, and (cycloalkyl)alkyl groups, said straight, branched, and cyclic alkyl groups, and (cycloalkyl)alkyl groups consisting of 1 to 8 carbon atoms, and containing zero or one or more double or triple bonds, each of which 1 to 8 carbon atoms may be further substituted with one or more substituent(s) independently selected from oxo, hydroxy, halogen, amino, cyano, nitro, haloalkyl, haloalkoxy, —O(alkyl), —NH(alkyl), —N(alkyl)(alkyl), —NHC(O)(alkyl), —N(alkyl)C(O)(alkyl), —NHS(O)n(alkyl), —S(O)n(alkyl), —S(O)nNH(alkyl), —S(O)nN(alkyl3)(alkyl4) where alkyl3 and alkyl4 may be joined to form a heterocycloalkyl ring consisting of from 5 to 8 ring atoms and containing 1, 2, or 3 heteroatoms selected from N, O, and S, and Y′;
Y and Y′ are independently selected at each occurrence from 3- to 8-membered carbocyclic or heterocyclic groups which are saturated, unsaturated, or aromatic, which may be further substituted with one or more substituents independently selected from halogen, oxo, hydroxy, amino, nitro, cyano, alkyl, alkoxy, haloalkyl, haloalkoxy, mono- or dialkylamino, and alkylthio;
 wherein said 3- to 8-memberered heterocyclic groups contain one or more heteroatom(s) independently selected from N, O, and S;
 n is independently chosen at each occurrence from 0, 1, and 2; and
 x is 1 or 3.
5. A compound or salt according to claim 4, wherein:
RA, RB, and RB′ are independently selected at each occurrence from hydrogen or C1-6alkyl;
R3 and R4 are independently chosen at each occurrence from the group consisting of hydrogen, halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl substituted with 0-2 R6, C2-6alkenyl substituted with 0-2 R6; C2-6alkynyl substituted with 0-2 R6; C1-6alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6, —N(C1-6alkyl)(C1-6alkyl) where each C1-6alkyl is independently substituted with 0-2 R6,
 —XR7, and Y;
or any two
R3 and R4 not attached to the same carbon may be joined to form an aryl ring substituted with 0-3 R6, a saturated or partially unsaturated carbocyclic ring of from 5 to 8 members, which carbocyclic ring is substituted with 0-2 R6, or a saturated, partially unsaturated, or aromatic heterocyclic ring of from 5 to 8 members, which heterocyclic ring is substituted with 0-2 R6 and contains 1, 2, or 3 heteroatoms selected from N, O, and S;
Ar1 and Ar2 may be the same or different and are selected from the group consisting of cyclohexyl, cyclopentyl, piperidinyl, piperazinyl, phenyl, pyrrolyl, furanyl, thienyl, pyrazolyl, imidazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, triazolyl, tetrazolyl, pyridyl, pyrimidyl, pyrazinyl, benzimidazolyl, naphthyl, indolyl, isoindolyl, benzofuranyl, isobenzofuranyl, benzo[b]thiophenyl, benz[d]isoxazolyl, quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, and quinoxalinyl, each of which is optionally mono-, di-, or trisubstituted with R5; or
Ar1 and Ar2 may be the same or different and represent a bicyclic oxygen-containing group of the formula:
Figure US20040176443A1-20040909-C00209
 optionally mono-, di-, or trisubstituted with R5, where L represents point of attachment and may be at any point on the benzene ring, and the oxygen-containing ring of the bicyclic oxygen-containing group consists of from 5 to 8 ring atoms, contains 1 or 2 oxygen atoms and remaining ring atoms are carbon;
R5 is independently selected at each occurrence from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl substituted with 0-2 R6, C2-6alkenyl substituted with 0-2 R6, C2-6alkynyl substituted with 0-2 R6, C1-6alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6, —N(C1-6alkyl)(C1-6alkyl) where each C1-6alkyl is independently substituted with 0-2 R6, —XR7, and Y;
R6 is independently selected at each occurrence from the group consisting of halogen, hydroxy, cyano, C1-4alkyl, C1-4alkoxy, —NH(C1-4alkyl), —N(C1-4alkyl)(C1-4alkyl), —S(O)(C1-4alkyl), halo(C1-4)alkyl, halo(C1-4)alkoxy, CO(C1-4alkyl), CONH(C1-4alkyl), CON(C1-4alkyl1)(C1-4alkyl2) where alkyl1 and alkyl2 may be joined to form a heterocycloalkyl ring of from 5 to 8 ring atoms and containing 1, 2, or 3 heteroatoms selected from N, O, and S, —XR7, and Y;
X is independently selected at each occurrence from the group consisting of —CH2—, —CHR8—, —O—, —S(O)n—, —NH—, —NR8—, —C(═O)—, —C(═O)O—, —C(═O)NH—, —C(═O)NR8—, —S(O)nNH—, —S(O)nNR8—, NHC(═O)—, —NR8C(═O)—, —NHS(O)n—, and —NR8S(O)n—;
R7 and R8 are independently selected at each occurrence from hydrogen, and straight, branched, and cyclic alkyl groups, and (cycloalkyl)alkyl groups, said straight, branched, and cyclic alkyl groups, and (cycloalkyl)alkyl groups consisting of 1 to 8 carbon atoms, and containing zero or one or more double or triple bonds, each of which 1 to 8 carbon atoms may be further substituted with one or more substituent(s) independently selected from oxo, hydroxy, halogen, amino, cyano, nitro, haloalkyl, haloalkoxy, —O(alkyl), —NH(alkyl), —N(alkyl)(alkyl), —NHC(O)(C1-4alkyl), —N(C1-4alkyl) C(O)(C1-4alkyl), —NHS(O)n(C1-4alkyl) —S(O)n(C1-4alkyl), —S(O)nNH(C1-4alkyl), —S(O)nN(C1-4alkyl3)(C1-4alkyl4) where C1-4alkyl3 and C1-4alkyl4 may be joined to form a heterocycloalkyl ring consisting of from 5 to 8 ring atoms and containing 1, 2, or 3 heteroatoms selected from N, O, and S, and Y′;
Y and Y′ are independently selected at each occurrence from 3- to 8-membered carbocyclic or heterocyclic groups which are saturated, unsaturated, or aromatic, which may be further substituted with one or more substituents independently selected from halogen, oxo, hydroxy, amino, nitro, cyano, C1-4alkyl, C1-4alkoxy, halo(C1-4)alkyl, halo(C1-4)alkoxy, mono- or di(C1-4)alkylamino, and C1-4alkylthio;
 wherein said 3- to 8-memberered heterocyclic groups contain one or more heteroatom(s) independently selected from N, O, and S;
n is independently chosen at each occurrence from 0, 1, and 2; and
x is 1 or 3.
6. A compound or salt according to claim 4, wherein Z is oxygen.
7. A compound or salt according to claim 4 of the formula:
Figure US20040176443A1-20040909-C00210
or a pharmaceutically acceptable salt thereof, wherein:
G, Q, T, and W are the same or different and represent N, CH, or CR5;
RA, RB, and RB′ are independently selected at each occurrence from hydrogen or C1-6alkyl;
Z is oxygen or sulfur;
R3 and R4 are independently selected at each occurrence from the group consisting of hydrogen; halogen; hydroxy; amino; cyano; nitro; —COOH; —CHO, optionally substituted alkyl; optionally substituted alkenyl; optionally substituted alkynyl; optionally substituted alkoxy; optionally substituted mono or dialkylamino; optionally substituted alkylthio; optionally substituted alkyl ketone; optionally substituted alkylester; optionally substituted alkylsulfinyl; optionally substituted alkylsulfonyl; optionally substituted mono- or di-alkylcarboxamide; optionally substituted —S(O)nNHalkyl; optionally substituted —S(O)nN(alkyl)(alkyl); optionally substituted —NHC(═O)alkyl; optionally substituted —NC(═O)(alkyl)(alkyl); optionally substituted —NHS(O)nalkyl; optionally substituted —NS(O)n(alkyl)(alkyl); optionally substituted saturated or partially unsaturated heterocycloalkyl of from 5 to 8 atoms, which saturated or partially unsaturated heterocycloalkyl contains 1, 2, or 3 heteroatoms selected from N, O, and S; optionally substituted aryl having from 1 to 3 rings; and optionally substituted heteroaryl, said heteroaryl having from 1 to 3 rings, 5 to 8 ring members in each ring and, in at least one of said rings, from 1 to about 3 heteroatoms per ring selected from the group consisting of N, O, and S;
or any two
R3 and R4 not attached to the same carbon may be joined to form an optionally substituted aryl ring; a saturated or partially unsaturated carbocyclic ring of from 5 to 8 members, which carbocyclic ring is optionally substituted; or a saturated, partially unsaturated, or aromatic heterocyclic ring of from 5 to 8 members, which heterocyclic ring is optionally substituted and contains 1, 2, or 3 heteroatoms selected from N, O, and S;
R5 represents 0 to 3 substituents on each of the aryl rings on which it occurs and is independently chosen at each occurrence from the group consisting of halogen, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl substituted with 0-2 R6, C2-6alkenyl substituted with 0-2 R6, C2-6alkynyl substituted with 0-2 R6, C1-6alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6, —N(C1-6alkyl) C1-6alkyl) where each C1-6alkyl is independently substituted with 0-2 R6, —XR7, and Y;
R6 is independently selected at each occurrence from the group consisting of halogen, hydroxy, cyano, alkyl, alkoxy, —NH(alkyl), —N(alkyl)(alkyl), —S(O)n(alkyl), haloalkyl, haloalkoxy, CO(alkyl), CONH(alkyl), CON(alkyl1)(alkyl2) where alkyl1 and alkyl2 may be joined to form a heterocycloalkyl ring of from 5 to 8 ring atoms and containing 1, 2, or 3 heteroatoms selected from N, O, and S, —XR7, and Y;
X is independently selected at each occurrence from the group consisting of —CH2—, —CHR8—, —O—, —S(O)n—, —NH—, —NR8—, —C(═O)—, —C(═O)O—, —C(═O)NH—, —C(═O)NR8—, —S(O)nNH—, —S(O)nNR8—, NHC(═O)—, —NR8C(═O)—, —NHS(O)n—, and —NR8S(O)n—;
R7 and R8 are independently selected at each occurrence from hydrogen, and straight, branched, and cyclic alkyl groups, and (cycloalkyl)alkyl groups, said straight, branched, and cyclic alkyl groups, and (cycloalkyl)alkyl groups-consisting of 1 to 8 carbon atoms, and containing zero or one or more double or triple bonds, each of which 1 to 8 carbon atoms may be further substituted with one or more substituent(s) independently selected from oxo, hydroxy, halogen, amino, cyano, nitro, haloalkyl, haloalkoxy, —O(alkyl), —NH(alkyl), —N(alkyl)(alkyl), —NHC(O)(alkyl), —N(alkyl)C(O)(alkyl), —NHS(O)n(alkyl), —S(O)n(alkyl), —S(O)nNH(alkyl), —S(O)nN(alkyl3)(alkyl4) where alkyl3 and alkyl4 may be joined to form a heterocycloalkyl ring consisting of from 5 to 8 ring atoms and containing 1, 2, or 3 heteroatoms selected from N, O, and S, and Y′;
Y and Y′ are independently selected at each occurrence from 3- to 8-membered carbocyclic or heterocyclic groups which are saturated, unsaturated, or aromatic, which may be further substituted with one or more substituents independently selected from halogen, oxo, hydroxy, amino, nitro, cyano, alkyl, alkoxy, haloalkyl, haloalkoxy, mono- or dialkylamino, and alkylthio;
 wherein said 3- to 8-memberered heterocyclic groups contain one or more heteroatom(s) independently selected from N, O, and S;
n is independently chosen at each occurrence from 0, 1, and 2; and
x is 1 or 3.
8. A compound of the formula:
Figure US20040176443A1-20040909-C00211
or a pharmaceutically acceptable salt thereof, wherein:
A is absent or is selected from the group consisting of O, S, NRA, CRBRB′, NRACRBRB′, CRBRB′NRA, —CRA═CRB—, and C3H4; where RA, RB, and RB′ are independently selected at each occurrence from hydrogen or alkyl;
R3 and R4 are independently selected at each occurrence from the group consisting of hydrogen; halogen; hydroxy; amino; cyano; nitro; —COOH; —CHO, optionally substituted alkyl; optionally substituted alkenyl; optionally substituted alkynyl; optionally substituted alkoxy; optionally substituted mono or dialkylamino; optionally substituted alkylthio; optionally substituted alkyl ketone; optionally substituted alkylester; optionally substituted alkylsulfinyl; optionally substituted alkylsulfonyl; optionally substituted mono- or di-alkylcarboxamide; optionally substituted —S(O)nNHalkyl; optionally substituted —S(O)nN (alkyl)(alkyl); optionally substituted —NHC(═O) alkyl; optionally substituted —NC(═O)(alkyl)(alkyl); optionally substituted —NHS(O)nalkyl; optionally substituted —NS(O)n(alkyl)(alkyl); optionally substituted saturated or partially unsaturated heterocycloalkyl of from 5 to 8 atoms, which saturated or partially unsaturated heterocycloalkyl contains 1, 2, or 3 heteroatoms selected from N, O, and S; optionally substituted aryl having from 1 to 3 rings; and optionally substituted heteroaryl, said heteroaryl having from 1 to 3 rings, 5 to 8 ring members in each ring and, in at least one of said rings, from 1 to about 3 heteroatoms per ring selected from the group consisting of N, O, and S;
or any two
R3 and R4 not attached to the same carbon may be joined to form an optionally substituted aryl ring; a saturated or partially unsaturated carbocyclic ring of from 5 to 8 members, which carbocyclic ring is optionally substituted; or a saturated, partially unsaturated, or aromatic heterocyclic ring of from 5 to 8 members, which heterocyclic ring is optionally substituted and contains 1, 2, or 3 heteroatoms selected from N, O, and S;
Ar1 and Ar2 may be the same or different and are selected from the group consisting of cyclohexyl, cyclopentyl, piperidinyl, piperazinyl, pyrrolyl, furanyl, thienyl, pyrazolyl, imidazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, triazolyl, tetrazolyl, pyridyl, pyrimidyl, pyrazinyl, benzimidazolyl, naphthyl, indolyl, isoindolyl, benzofuranyl, isobenzofuranyl, benzo[b]thiophenyl, benz[d]isoxazolyl, quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, and quinoxalinyl; wherein Ar1 is optionally mono-, di-, or trisubstituted with R5, and Ar2 is optionally mono-, di-, or trisubstituted with R9; or
Ar1 and Ar2 may be the same or different and represent a bicyclic oxygen-containing group of the formula:
Figure US20040176443A1-20040909-C00212
 optionally mono-, di-, or trisubstituted with R5, where L represents point of attachment and may be at any point on the benzene ring, and the oxygen-containing ring of the bicyclic oxygen-containing group consists of from 5 to 8 ring atoms, contains 1 or 2 oxygen atoms and remaining ring atoms are carbon;
R5 is independently selected at each occurrence from the group consisting of cyano, nitro, haloalkyl, haloalkoxy, hydroxy, amino, alkyl substituted with 0-2 R6, alkenyl substituted with 0-2 R6, alkynyl substituted with 0-2 R6, alkoxy substituted with 0-2 R6, —NH(alkyl) substituted with 0-2 R6, —N(alkyl)(alkyl) where each alkyl is independently substituted with 0-2 R6, —XR7, and Y;
R9 is independently selected at each occurrence from the group consisting of cyano, nitro, haloalkoxy, hydroxy, amino, alkyl substituted with 0-2 R6, alkenyl substituted with 0-2 R6, alkynyl substituted with 0-2 R6, alkoxy substituted with 0-2 R6, —NH(alkyl) substituted with 0-2 R6, —N(alkyl)(alkyl) where each alkyl is independently substituted with 0-2 R6, —XR7, and Y;
R6 is independently selected at each occurrence from the group consisting of halogen, hydroxy, cyano, alkyl, alkoxy, —NH(alkyl), —N(alkyl)(alkyl), —S(O)n(alkyl), haloalkyl, haloalkoxy, CO(alkyl), CONH(alkyl), CON(alkyl1)-(alkyl2) where alkyl1 and alkyl2 may be joined to form a heterocycloalkyl ring of from 5 to 8 ring atoms and containing 1, 2, or 3 heteroatoms selected from N, O, and S, —XR7, and Y;
X is independently selected at each occurrence from the group consisting of —CH2—, —CHR8—, —O—, —S(O)n—, —NH—, —NR8—, —C(═O)—, —C(═O)O—, —C(═O)NH—, —C(═O)NR8—, —S(O)nNH—, —S(O)nNR8—, NHC(═O)—, —NR8C(═O)—, —NHS(O)n—, and —NR8S(O)n—;
R7 and R8 are independently selected at each occurrence from hydrogen, and straight, branched, and cyclic alkyl groups, and (cycloalkyl)alkyl groups, said straight, branched, and cyclic alkyl groups, and (cycloalkyl)alkyl groups consisting of 1 to 8 carbon atoms, and containing zero or one or more double or triple bonds, each of which 1 to 8 carbon atoms may be further substituted with one or more substituent(s) independently selected from oxo, hydroxy, halogen, amino, cyano, nitro, haloalkyl, haloalkoxy, —O(alkyl), —NH(alkyl), —N(alkyl)(alkyl), —NHC(O)(alkyl), —N(alkyl)C(O)(alkyl), —NHS(O)n(alkyl), —S(O)n(alkyl), —S(O)nNH(alkyl), —S(O)nN(alkyl3)(alkyl4) where alkyl3 and alkyl4 may be joined to form a heterocycloalkyl ring consisting of from 5 to 8 ring atoms and containing 1, 2, or 3 heteroatoms selected from N, O, and S, and Y′;
Y and Y′ are independently selected at each occurrence from 3- to 8-membered carbocyclic or heterocyclic groups which are saturated, unsaturated, or aromatic, which may be further substituted with one or more substituents independently selected from halogen, oxo, hydroxy, amino, nitro, cyano, alkyl, alkoxy, haloalkyl, haloalkoxy, mono- or dialkylamino, and alkylthio;
 wherein said 3- to 8-memberered heterocyclic groups contain one or more heteroatom(s) independently selected from N, O, and S; and
n is independently chosen at each occurrence from 0, 1, and 2.
9. A compound of the formula:
Figure US20040176443A1-20040909-C00213
 or a pharmaceutically acceptable salt thereof, wherein:
G, Q, T, and W are the same or different and are selected from the group consisting of N, CH, and CR5, wherein T or W or both is N;
A is absent or is selected from the group consisting of O, S, NRA, CRBRB′, NRACRBRB′, CRBRB′NRA, —CRA═CRB—, and C3H4; where RA, RB, and RB′ are independently selected at each occurrence from hydrogen or alkyl;
Z is oxygen or sulfur;
R3 and R4 are independently selected at each occurrence from the group consisting of hydrogen; halogen; hydroxy; amino; cyano; nitro; —COOH; —CHO, optionally substituted C1-6alkyl; optionally substituted C2-6alkenyl; optionally substituted C2-6alkynyl; optionally substituted C1-6alkoxy; optionally substituted mono or di(C1-6)alkylamino; optionally substituted C1-6alkylthio; optionally substituted C1-6alkyl ketone; optionally substituted C1-6alkylester; optionally substituted C1-6alkylsulfinyl; optionally substituted C1-6alkylsulfonyl; optionally substituted mono- or di(C1-6)alkylcarboxamide; optionally substituted —S(O)nNH C1-6alkyl; optionally substituted —S(O)nN(C1-6alkyl)(C1-6alkyl); optionally substituted —NHC(═O) C1-6alkyl; optionally substituted —NC(═O)(C1-6alkyl)(C1-6alkyl); optionally substituted —NHS(O)nC1-6alkyl; optionally substituted —NS(O)n(C1-6alkyl)(C1-6alkyl); optionally substituted saturated or partially unsaturated heterocycloalkyl of from 5 to 8 atoms, which saturated or partially unsaturated heterocycloalkyl contains 1, 2, or 3 heteroatoms selected from N, O, and S; optionally substituted aryl having from 1 to 3 rings; and optionally substituted heteroaryl, said heteroaryl having from 1 to 3 rings, 5 to 8 ring members in each ring and, in at least one of said rings, from 1 to about 3 heteroatoms per ring selected from the group consisting of N, O, and S;
or any two
R3 and R4 not attached to the same carbon may be joined to form an optionally substituted aryl ring; a saturated or partially unsaturated carbocyclic ring of from 5 to 8 members, which carbocyclic ring is optionally substituted; or a saturated, partially unsaturated, or aromatic heterocyclic ring of from 5 to 8 members, which heterocyclic ring is optionally substituted and contains 1, 2, or 3 heteroatoms selected from N, O, and S;
R5 represents 1 to 3 substituents and is independently selected at each occurrence from the group consisting of cyano, hydroxy, amino, C3-6 alkyl substituted with 0-2 R6, C2-6 alkenyl substituted with 0-2 R6, C2-6 alkynyl substituted with 0-2 R6, C3-6 alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6, —N(C1-6alkyl)(C1-6alkyl) where each alkyl is independently substituted with 0-2 R6, —XR7, and Y;
R9 represents 0 to 3 substituents and is independently selected at each occurrence from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl substituted with 0-2 R6, C2-6alkenyl substituted with 0-2 R6, C2-6alkynyl substituted with 0-2 R6, C1-6alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6, —N(C1-6alkyl)(C1-6alkyl) where each C1-6alkyl is independently substituted with 0-2 R6, —XR7, and Y;
R6 is independently selected at each occurrence from the group consisting of halogen, hydroxy, cyano, C1-4alkyl, C1-4alkoxy, —NH(C1-4alkyl), —N(C1-4alkyl)(C1-4alkyl), —S(O)n(C1-4alkyl), halo(C1-4)alkyl, halo(C1-4)alkoxy, CO(C1-4alkyl), CONH(C1-4alkyl), CON(C1-4alkyl1)(C1-4alkyl2) where alkyl1 and alkyl2 may be joined to form a heterocycloalkyl ring of from 5 to 8 ring atoms and containing 1, 2, or 3 heteroatoms selected from N, O, and S, —XR7, and Y;
X is independently selected at each occurrence from the group consisting of —CH2—, —CHR8—, —O—, —S(O)n—, —NH—, —NR8—, —C(═O)—, —C(═O)O—, —C(═O)NH—, —C(═O)NR8—, —S(O)nNH—, —S(O)nNR8—, NHC(═O)—, —NR8C(═O)—, —NHS(O)n—, and —NR8S(O)n—;
R7 and R8 are independently selected at each occurrence from hydrogen, and straight, branched, and cyclic alkyl groups, and (cycloalkyl)alkyl groups, said straight, branched, and cyclic alkyl groups, and (cycloalkyl)alkyl groups consisting of 1 to 8 carbon atoms, and containing zero or one or more double or triple bonds, each of which 1 to 8 carbon atoms may be further substituted with one or more substituent(s) independently selected from oxo, hydroxy, halogen, amino, cyano, nitro, haloalkyl, haloalkoxy, —O(C1-4alkyl), —NH(C1-4alkyl), —N(C1-4alkyl)(C1-4alkyl), —NHC(O)(C1-4alkyl), —N(C1-4alkyl) C(O)(C1-4alkyl), —NHS(O)n(C1-4alkyl), —S(O)n(C1-4alkyl), —S(O)nNH(C1-4alkyl), —S(O)nN(C1-4alkyl3)(C1-4alkyl4) where C1-4alkyl3 and C1-4alkyl4 may be joined to form a heterocycloalkyl ring consisting of from 5 to 8 ring atoms and containing 1, 2, or 3 heteroatoms selected from N, O, and S, and Y′;
Y and Y′ are independently selected at each occurrence from 3- to 8-membered carbocyclic or heterocyclic groups which are saturated, unsaturated, or aromatic, which may be further substituted with one or more substituents independently selected from halogen, oxo, hydroxy, amino, nitro, cyano, C1-4alkyl, C1-4alkoxy, halo(C1-4)alkyl, halo(C1-4)alkoxy, mono- or di(C1-4)alkylamino, and C1-4alkylthio; wherein said 3- to 8-memberered heterocyclic groups contain one or more heteroatom(s) independently selected from N, O, and S; and
n is independently chosen at each occurrence from 0, 1, and 2.
10. A compound according to claim 9, which is 4-(3-Chloro-2-pyridinyl)-N-[4(isopropyl)phenyl]-2-methylthio-1-piperazinecarboxamide, or a pharmaceutically acceptable salt thereof.
11. A compound according to claim 9, wherein R3 and R4 are independently selected at each occurrence from the group consisting of hydrogen and C1-6 alkyl.
12. A compound according to claim 11, wherein G and Q are selected from the group consisting of CH and CR5.
13. A compound according to claim 11, wherein G, Q, and W are independently selected at each occurrence from the group consisting of CH and CR5; and T is N.
14. A compound according to claim 13 wherein R3 and R4 are hydrogen; and A is selected from the group consisting of NH, —CH═CH—, and —CH2NH—.
15. A compound or salt according to claim 14, wherein R6 is independently selected at each occurrence from the group consisting of halogen, hydroxy, C1-4 alkyl, C1-4alkoxy, —NH(C1-4alkyl), and —N(C1-4 alkyl)(C1-4 alkyl).
16. A compound according to claim 14, which is 4-(3-Trifluoromethyl-2-pyridinyl)-N-(3-methoxy-4-hydroxyphenylmethyl)-1-piperazinecarboxamide, or a pharmaceutically acceptable salt thereof.
17. A compound according to claim 14, which is 4-(3-Nitro-2-pyridinyl)-N-[4-(n-butyl)phenyl]-1-piperazinecarboxamide, or a pharmaceutically acceptable salt thereof.
18. A compound according to claim 14, which is 4-(3-Trifluoromethyl-2-pyridinyl)-N-[4-(n-butyl)phenyl]-1-piperazinecarboxamide, or a pharmaceutically acceptable salt thereof.
19. A compound according to claim 14, which is 4-(3-Methyl-2-pyridinyl)-N-[4-(isopropyl)phenyl]-1-piperazinecarboxamide, or a pharmaceutically acceptable salt thereof.
20. A compound according to claim 14, which is 4-(3-Methyl-2-pyridinyl)-N-[4-(n-butyl)phenyl]-1-piperazinecarboxamide, or a pharmaceutically acceptable salt thereof.
21. A compound according to claim 14, which is 4-(3-Chloro-5-trifluoromethyl-2-pyridinyl)-N-[4-(isopropyl)phenyl]-1-piperazinecarboxamide, or a pharmaceutically acceptable salt thereof.
22. A compound according to claim 14, which is 4-(3-Chloro-2-pyridinyl)-N-[4-(isopropyl)phenyl]-1-piperazinecarboxamide, or a pharmaceutically acceptable salt thereof.
23. A compound according to claim 14, which is 4-(3,5-Dichloro-2-pyridinyl)-N-[4-(isopropyl)phenyl]-1-piperazinecarboxamide, or a pharmaceutically acceptable salt thereof.
24. A compound according to claim 13, which is 4-(3-Cyano-2-pyridinyl)-N-[4-(isopropyl)phenyl]-1-piperazinecarboxamide, or a pharmaceutically acceptable salt thereof.
25. The compound according to claim 13, which is 4-(3-Chloro-2-pyridinyl)-N-[4-(isopropyl)phenyl]-2-methyl-1-piperazinecarboxamide.
26. A compound of the formula:
Figure US20040176443A1-20040909-C00214
or a pharmaceutically acceptable salt thereof, wherein:
A is selected from the group consisting of NH, —CH═CH—, and CH2NH;
R4 is independently chosen from hydrogen and C1-4 alkyl;
R5 represents 0 to 2 substituents and is independently chosen at each occurrence from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl substituted with 0-2 R6, C2-6alkenyl substituted with 0-2 R6, C2-6alkynyl substituted with 0-2 R6, C1-6alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6,
 —N(C1-6alkyl)(C1-6alkyl) where each C1-6 alkyl is independently substituted with 0-2 R6;
R9 represents 0 to 2 substituents and is independently chosen at each occurrence from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl substituted with 0-2 R6, C2-6alkenyl substituted with 0-2 R6, C2-6alkynyl substituted with 0-2 R6, C1-6alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6, and —N(C1-6alkyl)(C1-6alkyl) where each C1-6alkyl is independently substituted with 0-2 R6;
R5A is independently selected from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6 alkyl, C1-6 alkoxy, —NH(C1-6 alkyl), and —N(C1-6 alkyl)(C1-6 alkyl);
R9B is independently selected from the group consisting of halogen, nitro, halo(C1-6)alkoxy, hydroxy, amino, C1-6 alkyl, C1-6 alkoxy, —NH(C1-6 alkyl), and —N(C1-6 alkyl)(C1-6 alkyl) and
R6 is independently selected at each occurrence the group consisting of halogen, hydroxy, C1-4alkyl, C1-4alkoxy, —NH(C1-4 alkyl), and —N(C1-4 alkyl)(C1-4 alkyl).
27. A compound of the formula:
Figure US20040176443A1-20040909-C00215
or a pharmaceutically acceptable salt thereof, wherein:
A is selected from the group consisting of a single bond, S, NRA, CHRB, NRACHRB, CHRBNRA, —CRA═CRB—, and C3H4; where RA and RB are independently selected at each occurrence from the group consisting of hydrogen and alkyl;
R3 and R4 are independently selected at each occurrence from the group consisting of hydrogen; halogen; hydroxy; amino; cyano; nitro; —COOH; —CHO, optionally substituted alkyl; optionally substituted alkenyl; optionally substituted alkynyl; optionally substituted alkoxy; optionally substituted mono or dialkylamino; optionally substituted alkylthio; optionally substituted alkyl ketone; optionally substituted alkylester; optionally substituted alkylsulfinyl; optionally substituted alkylsulfonyl; optionally substituted mono- or di-alkylcarboxamide; optionally substituted —S(O)nNHalkyl; optionally substituted —S(O)nN (alkyl)(alkyl); optionally substituted —NHC(═O) alkyl; optionally substituted —NC(═O)(alkyl)(alkyl); optionally substituted —NHS(O)nalkyl; optionally substituted NS(O)n(alkyl) (alkyl); optionally substituted saturated or partially unsaturated heterocycloalkyl of from 5 to 8 atoms, which saturated or partially unsaturated heterocycloalkyl contains 1, 2, or 3 heteroatoms selected from N, O, and S; optionally substituted aryl having from 1 to 3 rings; and optionally substituted heteroaryl, said heteroaryl having from 1 to 3 rings, 5 to 8 ring members in each ring and, in at least one of said rings, from 1 to about 3 heteroatoms per ring selected from the group consisting of N, O, and S;
or any two
R3 and R4 not attached to the same carbon may be joined to form an optionally substituted aryl ring; a saturated or partially unsaturated carbocyclic ring of from 5 to 8 members, which carbocyclic ring is optionally substituted; or a saturated, partially unsaturated, or aromatic heterocyclic ring of from 5 to 8 members, which heterocyclic ring is optionally substituted and contains 1, 2, or 3 heteroatoms selected from N, O, and S;
R5 is independently selected at each occurrence from the group consisting of cyano, nitro, haloalkyl, haloalkoxy, C1-6 alkyl substituted with 0-2 R6, C2-6 alkenyl substituted with 0-2 R6, C2-6 alkynyl substituted with 0-2 R6, C1-6 alkoxy substituted with 0-2 R6, —NH(C1-6 alkyl) substituted with 0-2 R6, —N(C1-6 alkyl)(C1-6 alkyl) where each alkyl is independently substituted with 0-2 R6, —XR7, and Y;
R9 represents 0-3 substituents and is independently selected at each occurrence from the group consisting of bromo, haloalkyl, haloalkoxy, hydroxy, C2-6 alkyl substituted with 0-2 R6, C2-6 alkenyl substituted with 0-2 R6, C2-6 alkynyl substituted with 0-2 R6, C2-6 alkoxy substituted with 0-2 R6, —NH(C2-6 alkyl) substituted with 0-2 R6, —N(C2-6 alkyl)(C2-6 alkyl) where each C2-6 alkyl is independently substituted with 0-2 R6, —XR7, and Y;
R6 is independently selected at each occurrence from the group consisting of halogen, hydroxy, cyano, alkyl, alkoxy, —NH(alkyl), —N(alkyl)(alkyl), —S(O)n(alkyl), haloalkyl, haloalkoxy, CO(alkyl), CONH(alkyl), CON(alkyl1)(alkyl2) where alkyl1 and alkyl2 may be joined to form a heterocycloalkyl ring of from 5 to 8 ring atoms and containing 1, 2, or 3 heteroatoms selected from N, O, and S, —XR7, and Y;
X is independently selected at each occurrence from the group consisting of —CH2—, —CHR8—, —O—, —S(O)n—, —NH—, —NR8—, —C(═O)—, —C(═O)O—, —C(═O)NH—, —C(═O)NR8—, —S(O)nNH—, —S(O)nNR8—, NHC(═O)—, —NR8C(═O)—, —NHS(O)n—, and —NR8S(O)n—;
R7 and R8 are independently selected at each occurrence from straight, branched, and cyclic alkyl groups, and (cycloalkyl)alkyl groups, said straight, branched, and cyclic alkyl groups, and (cycloalkyl)alkyl groups consisting of 3 to 8 carbon atoms, and containing zero or one or more double or triple bonds, each of which 3 to 8 carbon atoms may be further substituted with one or more substituent(s) independently selected from oxo, hydroxy, halogen, amino, cyano, nitro, haloalkyl, haloalkoxy, —O(alkyl),
 —NH(alkyl), —N(alkyl)(alkyl), —NHC(O)(alkyl), —N(alkyl)C(O)(alkyl), —NHS(O)n(alkyl),
 —S(O)n(alkyl), —S(O)nNH(alkyl), —S(O)nN(alkyl3)(alkyl4) where alkyl3 and alkyl4 may be joined to form a heterocycloalkyl ring consisting of from 5 to 8 ring atoms and containing 1, 2, or 3 heteroatoms selected from N, O, and S, and Y′;
Y and Y′ are independently selected at each occurrence from 3- to 8-membered carbocyclic or heterocyclic groups which are saturated, unsaturated, or aromatic, which may be further substituted with one or more substituents independently selected from halogen, oxo, hydroxy, amino, nitro, cyano, alkyl, alkoxy, haloalkyl, haloalkoxy, mono- or dialkylamino, and alkylthio;
 wherein said 3- to 8-memberered heterocyclic groups contain one or more heteroatom(s) independently selected from N, O, and S; and
n is independently chosen at each occurrence from 0, 1, and 2.
28. A compound or salt according to claim 27 in which R3 and R4 are independently selected at each occurrence from the group consisting of hydrogen and C1-6 alkyl.
29. A compound or salt according to claim 27, wherein A is selected from the group consisting of NH, —CH═CH—, and CH2NH; R3 is hydrogen and R4 is independently chosen at each occurrence from hydrogen and methyl; and R6 is independently selected at each occurrence from the group consisting of halogen, hydroxy, C1-4 alkyl, C1-4 alkoxy, —NH(C1-4 alkyl), and —N(C1-4 alkyl)(C1-4 alkyl).
30. A compound of the formula:
Figure US20040176443A1-20040909-C00216
or a pharmaceutically acceptable salt thereof, wherein:
A is selected from the group consisting of NH, —CH═CH—, and CH2NH;
R4 is independently selected at each occurrence from hydrogen and C1-4alkyl;
R5 represents 0 to 2 substituents independently selected at each occurrence from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl substituted with 0-2 R6, C2-6alkenyl substituted with 0-2 R6, C2-6alkynyl substituted with 0-2 R6, C1-6alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6, and —N(C1-6alkyl)(C1-6alkyl) where each C1-6alkyl is independently substituted with 0-2 R6;
R9 represents 0 to 2 substituents and is independently selected at each occurrence from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl substituted with 0-2 R6, C2-6alkenyl substituted with 0-2 R6, C2-6alkynyl substituted with 0-2 R6, C1-6alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6, and —N(C1-6alkyl)(C1-6alkyl) where each C1-6alkyl is independently substituted with 0-2 R6;
R5A is independently selected from the group consisting of halogen, cyano, nitro, trifluoromethyl, trifluoromethoxy, hydroxy, amino, C1-6 alkyl, C1-6 alkoxy, —NH(C1-6 alkyl), and —N(C1-6 alkyl)(C1-C6 alkyl);
R9B is independently selected from the group consisting of trifluoromethoxy, hydroxy, C2-6 alkyl, C2-6 alkoxy, —NH(C2-6 alkyl), and —N(C2-6 alkyl)(C2-6 alkyl); and
R6 is independently selected at each occurrence from the group consisting of halogen, hydroxy, C1-4 alkyl, C1-4 alkoxy, —NH(C1-4 alkyl), and —N(C1-4 alkyl)(C1-4 alkyl).
31. A compound of the formula:
Figure US20040176443A1-20040909-C00217
or a pharmaceutically acceptable salt thereof, wherein the compound or pharmaceutically acceptable salt thereof exhibits an EC50 or Ki of 1 micromolar or less in a standard assay of capsaicin receptor mediated calcium mobilization; and wherein
A is absent or is selected from the group consisting of O, S, NRA, CRBRB′, NRACRBRB′,
 CRBRB′NRA, —CRA=CRB—, and C3H4; where RA, RB, and RB′ are independently selected at each occurrence from hydrogen or C1-6 alkyl;
Z is oxygen or sulfur;
R1 and R2 independently represent hydrogen or C1-6 alkyl; or
R1 and R2 are taken together to form a 5 to 8 membered nitrogen-containing ring of the formula:
Figure US20040176443A1-20040909-C00218
wherein x is 1, 2, or 3;
R3 and R4 are independently chosen at each occurrence from the group consisting of hydrogen, halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl substituted with 0-2 R6, C2-6alkenyl substituted with 0-2 R6; C2-6alkynyl substituted with 0-2 R6; C1-6alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6, —N(C1-6alkyl)(C1-6alkyl) where each C1-6alkyl is independently substituted with 0-2 R6,
 —XR7, and Y;
or any two
R3 and R4 not attached to the same carbon may be joined to form an aryl ring substituted with 0-3 R6, a saturated or partially unsaturated carbocyclic ring of from 5 to 8 members, which carbocyclic ring is substituted with 0-2 R6, or a saturated, partially unsaturated, or aromatic heterocyclic ring of from 5 to 8 members, which heterocyclic ring is substituted with 0-2 R6 and contains 1, 2, or 3 heteroatoms selected from N, O, and S;
Ar1 and Ar2 may be the same or different and are selected from the group consisting of cyclohexyl, cyclopentyl, piperidinyl, piperazinyl, phenyl, pyrrolyl, furanyl, thienyl, pyrazolyl, imidazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, triazolyl, tetrazolyl, pyridyl, pyrimidyl, pyrazinyl, benzimidazolyl, naphthyl, indolyl, isoindolyl, benzofuranyl, isobenzofuranyl, benzo[b]thiophenyl, benz[d]isoxazolyl, quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, and quinoxalinyl, each of which is optionally mono-, di-, or trisubstituted with R5; or
Ar1 and Ar2 may be the same or different and represent a bicyclic oxygen-containing group of the formula:
Figure US20040176443A1-20040909-C00219
 optionally mono-, di-, or trisubstituted with R5, where L represents point of attachment and may be at any point on the benzene ring, and the oxygen-containing ring of the bicyclic oxygen-containing group consists of from 5 to 8 ring atoms, contains 1 or 2 oxygen atoms and remaining ring atoms are carbon;
R5 is independently selected at each occurrence from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl substituted with 0-2 R6, C2-6alkenyl substituted with 0-2 R6, C2-6alkynyl substituted with 0-2 R6, C1-6alkoxy substituted with 0-2 R6,
 —NH(C1-6alkyl) substituted with 0-2 R6, —N(C1-6alkyl)(C1-6alkyl) where each C1-6alkyl is independently substituted with 0-2 R6, —XR7, and Y;
R6 is independently selected at each occurrence from the group consisting of halogen, hydroxy, cyano, C1-4alkyl, C1-4alkoxy, —NH(C1-4alkyl), —N(C1-4alkyl)(C1-4alkyl), —S(O)n(C1-4alkyl), halo(C1-4)alkyl, halo(C1-4)alkoxy, CO(C1-4alkyl), CONH(C1-4alkyl), CON(C1-4alkyl1)(C1-4alkyl2) where alkyl1 and alkyl2 may be joined to form a heterocycloalkyl ring of from 5 to 8 ring atoms and containing 1, 2, or 3 heteroatoms selected from N, O, and S, —XR7, and Y;
X is independently selected at each occurrence from the group consisting of —CH2—, —CHR8—, —O—, —S(O)n—, —NH—, —NR8—, —C(═O)—, —C(═O)O—, —C(═O)NH—, —C(═O)NR8—, —S(O)NH—, —S(O)nNR8—, NHC(═O)—, —NR8C(═O)—, —NHS(O)n—, and —NR8S(O)n—;
R7 and R8 are independently selected at each occurrence from hydrogen, and straight, branched, and cyclic alkyl groups, and (cycloalkyl)alkyl groups, said straight, branched, and cyclic alkyl groups, and (cycloalkyl)alkyl groups consisting of 1 to 8 carbon atoms, and containing zero or one or more double or triple bonds, each of which 1 to 8 carbon atoms may be further substituted with one or more substituent(s) independently selected from oxo, hydroxy, halogen, amino, cyano, nitro, haloalkyl, haloalkoxy, —O(C1-4alkyl),
 —NH(C1-4alkyl), —N(C1-4alkyl)(C1-4alkyl), —NHC(O)(C1-4alkyl) —N(C1-4alkyl) C(O)n(C1-4alkyl), —NHS(O)n(C1-4alkyl), —S(O)n(C1-4alkyl), —S(O)nNH(C1-4alkyl),
 —S(O)nN(C1-4alkyl3)(C1-4alkyl4) where C1-4alkyl3 and C1-4alkyl4 may be joined to form a heterocycloalkyl ring consisting of from 5 to 8 ring atoms and containing 1, 2, or 3 heteroatoms selected from N, O, and S, and Y′;
Y and Y′ are independently selected at each occurrence from 3 to 8-membered carbocyclic or heterocyclic groups which are saturated, unsaturated, or aromatic, which may be further substituted with one or more substituents independently selected from halogen, oxo, hydroxy, amino, nitro, cyano, C1-4alkyl, C1-4alkoxy, halo(C1-4)alkyl, halo(C1-4)alkoxy, mono- or di(C1-4)alkylamino, and C1-4alkylthio;
 wherein said 3- to 8-memberered heterocyclic groups contain one or more heteroatom(s) independently selected from N, O, and S; and
n is independently chosen at each occurrence from 0, 1, and 2.
32. A compound of the Formula A, Formula B, Formula C, Formula D, Formula E, or Formula F:
Figure US20040176443A1-20040909-C00220
 or a pharmaceutically acceptable salt of Formula A, Formula B, Formula C, Formula D, Formula E, or Formula F, wherein
A represents NH or O;
R3 and R4 are independently chosen at each occurrence from the group consisting of hydrogen, halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl substituted with 0-2 R6, C2-6alkenyl substituted with 0-2 R6; C2-6alkynyl substituted with 0-2 R6; C1-6alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6, —N(C1-6alkyl)(C1-6alkyl) where each C1-6alkyl is independently substituted with 0-2 R6,
 —XR7, and Y;
or any two
R3 and R4 not attached to the same carbon may be joined to form an aryl ring substituted with 0-3 R6, a saturated or partially unsaturated carbocyclic ring of from 5 to 8 members, which carbocyclic ring is substituted with 0-2 R6, or a saturated, partially unsaturated, or aromatic heterocyclic ring of from 5 to 8 members, which heterocyclic ring is substituted with 0-2 R6 and contains 1, 2, or 3 heteroatoms selected from N, O, and S;
R5 and R9 each represent from 1 to 3 substituents and are independently selected at each occurrence from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl substituted with 0-2 R6, C2-6alkenyl substituted with 0-2 R6, C2-6alkynyl substituted with 0-2 R6, C1-6alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6, —N(C1-6alkyl)(C1-6alkyl) where each C1-6alkyl is independently substituted with 0-2 R6, —XR7, and Y;
R6 is independently selected at each occurrence from the group consisting of halogen, hydroxy, cyano, C1-4alkyl, C1-4alkoxy, —NH(C1-4alkyl), —N(C1-4alkyl)(C1-4alkyl), —S(O)n(C1-4alkyl) halo(C1-4) alkyl, halo(C1-4) alkoxy, CO(C1-4alkyl), CONH(C1-4alkyl), CON(C1-4alkyl1)(C1-4alkyl2) where alkyl1 and alkyl2 may be joined to form a heterocycloalkyl ring of from 5 to 8 ring atoms and containing 1, 2, or 3 heteroatoms selected from N, O, and S, —XR7, and Y;
X is independently selected at each occurrence from the group consisting of —CH2—, —CHR8—, —O—, —S(O)n—, —NH—, —NR8—, —C(═O)—, —C(═O)O—, —C(═O)NH—, —C(═O)NR8—, —S(O)nNH—, —S(O)nNR8—, NHC(═O)—, —NR8C(═O)—, —NHS(O)n—, and —NR8S(O)n—;
R7 and R8 are independently selected at each occurrence from hydrogen, and straight, branched, and cyclic alkyl groups, and (cycloalkyl)alkyl groups, said straight, branched, and cyclic alkyl groups, and (cycloalkyl)alkyl groups consisting of 1 to 8 carbon atoms, and containing zero or one or more double or triple bonds, each of which 1 to 8 carbon atoms may be further substituted with one or more substituent(s) independently selected from oxo, hydroxy, halogen, amino, cyano, nitro, haloalkyl, haloalkoxy, —O(C1-4alkyl)-NH(C1-4alkyl), —N(C1-4alkyl)(C1-4alkyl) —NHC(O)(C1-4alkyl), —N(C1-4alkyl)C(O)(C1-4alkyl) —NHS(O)n(C1-4alkyl), —S(O)n(C1-4alkyl), —S(O)nNH(C1-4alkyl), —S(O)nN(C1-4alkyl3)(C1-4alkyl4) where C1-4alkyl3 and C1-4alkyl4 may be joined to form a heterocycloalkyl ring consisting of from 5 to 8 ring atoms and containing 1, 2, or 3 heteroatoms selected from N, O, and S, and Y′;
Y and Y′ are independently selected at each occurrence from 3- to 8-membered carbocyclic or heterocyclic groups which are saturated, unsaturated, or aromatic, which may be further substituted with one or more substituents independently selected from halogen, oxo, hydroxy, amino, nitro, cyano, C1-4alkyl, C1-4alkoxy, halo(C1-4)alkyl, halo(C1-4)alkoxy, mono- or di(C1-4)alkylamino, and C1-4alkylthio;
 wherein said 3- to 8-memberered heterocyclic groups contain one or more heteroatom(s) independently selected from N, O, and S; and
n is independently chosen at each occurrence from 0, 1, and 2.
33. A compound or salt according to claim 32, wherein A represents NH.
34. A compound or salt according to claim 32, wherein:
A represents NH; and
R3 and R4 are independently chosen at each occurrence from the group consisting of hydrogen, halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6alkoxy substituted with 0-2 R6, —NH(C1-6alkyl), and —N(C1-6alkyl)(C1-6alkyl).
35. A compound or salt according to claim 32, wherein:
A represents NH;
R3 represents hydrogen; and
R4 is independently chosen at each occurrence from hydrogen and C1-6 alkyl.
36. A compound or salt according to claim 32, wherein:
A represents NH;
R3 represents hydrogen; and
R4 is independently chosen at each occurrence from hydrogen and methyl.
37. A compound or salt according to claim 32, wherein:
A represents NH;
R3 represents hydrogen;
R4 is independently chosen at each occurrence from hydrogen and methyl; and
R5 and R9 each represent from 1 to 3 substituents independently selected at each occurrence from the group consisting of halogen, cyano, nitro, halo(C1-16)alkyl, halo(C1-6)alkoxy, hydroxy; amino, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6alkoxy, —NH(C1-6alkyl, —N(C1-6alkyl)(C1-6alkyl), and C3-8 cycloalkyl.
38. A compound or salt according to claim 37 of the Formula A-1
Figure US20040176443A1-20040909-C00221
R5 and R9 are independently selected from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6alkoxy, —NH(C1-6alkyl, —N(C1-6alkyl)(C1-6alkyl), and C3-8 cycloalkyl; and
R5B and R9B each represent up to 2 substituents independently selected at each occurrence from hydrogen, halogen, cyano, nitro, halo(C1-2)alkyl, halo(C1-2)alkoxy, hydroxy, amino, C1-3alkyl, C1-3alkoxy, —NH(C1-3alkyl), and —N(C1-6alkyl)(C1-6alkyl).
39. A compound or salt according to claim 38, wherein:
R5 is C3-6 alkyl; C3-6 alkoxy; halo(C1-3)alkyl, halo(C1-3)alkoxy, or C3-8 cycloalkyl;
R9 is chloro or trifluoromethyl; and
R5B and R9B are hydrogen.
40. A compound or salt according to claim 37 of Formula B-1
Figure US20040176443A1-20040909-C00222
wherein
R5 and R9 are independently selected from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6alkoxy, —NH(C1-6alkyl, —N(C1-6alkyl)(C1-6alkyl), and C3-8 cycloalkyl; and
R5B and R9B each represent up to 2 substituents independently selected at each occurrence from hydrogen, halogen, cyano, nitro, halo(C1-2)alkyl, halo(C1-2)alkoxy, hydroxy, amino, C1-3alkyl, C1-3alkoxy, —NH(C1-3alkyl), and —N(C1-6alkyl)(C1-6alkyl).
41. A compound or salt according to claim 40, wherein:
R5 is C3-6 alkyl; C3-6 alkoxy; halo(C1-3)alkyl, halo(C1-3)alkoxy, or C3-8 cycloalkyl;
R9 is chloro or trifluoromethyl; and
R5B and R9B are hydrogen.
42. A compound or salt according to claim 37 of Formula C-1:
Figure US20040176443A1-20040909-C00223
wherein:
R5 and R9 are independently selected from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6alkoxy, —NH(C1-6alkyl, —N(C1-6alkyl)(C1-6alkyl), and C3-8 cycloalkyl; and
R5B and R9B each represent up to 2 substituents independently selected at each occurrence from hydrogen, halogen, cyano, nitro, halo(C1-2)alkyl, halo(C1-2)alkoxy, hydroxy, amino, C1-3alkyl, C1-3alkoxy, —NH(C1-3alkyl), and —N(C1-6alkyl)(C1-6alkyl).
43. A compound or salt according to claim 42, wherein:
R5 is C3-6 alkyl; C3-6 alkoxy; halo(C1-3)alkyl, halo(C1-3)alkoxy, or C3-8 cycloalkyl;
R9 is chloro or trifluoromethyl; and
R5B and R9B are hydrogen.
44. A compound or salt according to claim 37 of Formula D-1
Figure US20040176443A1-20040909-C00224
wherein:
R5 and R9 are independently selected from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6alkoxy, —NH(C1-6alkyl, —N(C1-6alkyl)(C1-6alkyl); and C3-8 cycloalkyl; and
R5B and R9B each represent up to 2 substituents independently selected at each occurrence from hydrogen, halogen, cyano, nitro, halo(C1-2)alkyl, halo(C1-2)alkoxy, hydroxy, amino, C1-3alkyl, C1-3alkoxy, —NH(C1-3alkyl), and —N(C1-6alkyl)(C1-6alkyl).
45. A compound or salt according to claim 44, wherein:
R5 is C3-6 alkyl; C3-6 alkoxy; halo(C1-3)alkyl, halo(C1-3)alkoxy, or C3-8 cycloalkyl;
R9 is chloro or trifluoromethyl; and
R5B and R9B are hydrogen.
46. A compound or salt according to claim 37, of Formula E-1
Figure US20040176443A1-20040909-C00225
wherein:
R5 and R9 are independently selected from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6alkoxy, —NH(C1-6alkyl, —N(C1-6alkyl)(C1-6alkyl), and C3-8 cycloalkyl; and
R5B and R9B each represent up to 2 substituents independently selected at each occurrence from hydrogen, halogen, cyano, nitro, halo(C1-2)alkyl, halo(C1-2)alkoxy, hydroxy, amino, C1-3alkyl, C1-3alkoxy, —NH(C1-3alkyl), and —N(C1-6alkyl)(C1-6alkyl).
47. A compound or salt according to claim 46, wherein:
R5 is C3-6 alkyl; C3-6 alkoxy; halo(C1-3)alkyl, halo(C1-3)alkoxy, or C3-8 cycloalkyl;
R9 is chloro or trifluoromethyl; and
R5B and R9B are hydrogen.
48. A compound of salt according to claim 37 of Formula F-1
Figure US20040176443A1-20040909-C00226
wherein:
R5 and R9 are independently selected from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6alkoxy, —NH(C1-6alkyl, —N(C1-6alkyl)(C1-6alkyl), and C3-8 cycloalkyl; and
R5B and R9B each represent up to 2 substituents independently selected at each occurrence from hydrogen, halogen, cyano, nitro, halo(C1-2)alkyl, halo(C1-2)alkoxy, hydroxy, amino, C1-3alkyl, C1-3alkoxy, —NH(C1-3alkyl), and —N(C1-6alkyl)(C1-6alkyl).
49. A compound or salt according to claim 47, wherein:
R5 is C3-6 alkyl; C3-6 alkoxy; halo(C1-3)alkyl, halo(C1-3)alkoxy, or C3-8 cycloalkyl;
R9 is chloro or trifluoromethyl; and
R5B and R9B are hydrogen.
50. A compound of the Formula:
Figure US20040176443A1-20040909-C00227
or a pharmaceutically acceptable salt thereof, wherein:
A is absent or is selected from the group consisting of O, S, NRA, CRBRB′, NRACRBRB′, CRBRB′NRA, —CRA═CRB—, and C3H4; where RA, RB, and RB′ are independently selected at each occurrence from hydrogen or C1-6 alkyl;
R3 and R4 are independently chosen at each occurrence from the group consisting of hydrogen, halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl substituted with 0-2 R6, C2-6alkenyl substituted with 0-2 R6; C2-6alkynyl substituted with 0-2 R6; C1-6alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6, —N(C1-16alkyl)(C1-6alkyl) where each C1-6alkyl is independently substituted with 0-2 R6,
 —XR7, and Y;
or any two
R3 and R4 not attached to the same carbon may be joined to form an aryl ring substituted with 0-3 R6, a saturated or partially unsaturated carbocyclic ring of from 5 to 8 members, which carbocyclic ring is substituted with 0-2 R6, or a saturated, partially unsaturated, or aromatic heterocyclic ring of from 5 to 8 members, which heterocyclic ring is substituted with 0-2 R6 and contains 1, 2, or 3 heteroatoms selected from N, O, and S;
R5 is selected from the group consisting of bromo, fluoro, iodo, halo(C1-6)alkyl, halo(C3-6)alkoxy, C3-6alkyl substituted with 0-3 R6, C2-6alkenyl substituted with 0-3 R6, C2-6alkynyl substituted with 0-3 R6, C3-6alkoxy substituted with 0-2 R6, (C3-8cycloalkyl)C1-4alkyl, —NH(C1-6alkyl) substituted with 0-2 R6, —N(C1-6alkyl)(C1-6alkyl) where each C1-6alkyl is substituted with 0-2 R6, Y, —(C═O)Y, —(CH2)Y, and —(CH(CN))Y;
R9 is selected from the group consisting of halogen, cyano, —N(SO2C1-6alkyl)(SO2C1-6alkyl), —SO2NH2, halo (C1-6) alkyl, halo(C1-16)alkoxy, C1-6alkyl substituted with 0-2 R6, C2-6alkenyl substituted with 0-2 R6, C2-6alkynyl substituted with 0-2 R6, C1-6alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6,
 —N(C1-6alkyl)(C1-6alkyl) where each C1-6alkyl is substituted with 0-2 R6;
R5B and R9B each represent from 0 to 2 substituents and are independently selected at each occurrence from the group consisting of halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl substituted with 0-2 R6, (C3-8cycloalkyl)C1-4alkyl substituted with 0-2 R6, C2-6alkenyl substituted with 0-2 R6, C2-6alkynyl substituted with 0-2 R6, C1-6alkoxy substituted with 0-2 R6, —NH(C1-6alkyl) substituted with 0-2 R6, and —N(C1-16alkyl)(C1-6alkyl) where each C1-6alkyl is independently substituted with 0-2 R6, and Y; and any two
R5 and R5B bound to adjacent atoms may be joined to form a C3-8cycloalkyl group or a heterocycloalkyl group, each of which is optionally substituted by from 1 to 5 substituents independently chosen from cyano, halogen, hydroxy, C1-4alkyl, C1-4alkoxy, —NH(C1-4alkyl), —N(C1-4alkyl)(C1-4alkyl) halo(C1-4)alkyl, and halo(C1-4)alkoxy, wherein the heterocycloalkyl group consists of from 4 to 8 atoms and contains 1, 2, or 3 heteroatoms selected from N, O, and S;
R6 is independently selected at each occurrence from the group consisting of cyano, halogen, hydroxy, C1-4alkyl, C1-4alkoxy, —NH(C1-4alkyl), —N(C1-4alkyl)(C1-4alkyl), —S(O)n(C1-4alkyl) halo(C1-4)alkyl, halo(C1-4)alkoxy, CO(C1-4alkyl), CONH(C1-4alkyl), CON(C1-4alkyl1)(C1-4alkyl2) where alkyl1 and alkyl2 may be joined to form a heterocycloalkyl ring of from 5 to 8 ring atoms and containing 1, 2, or 3 heteroatoms selected from N, O, and S, —XR7, and Y;
X is independently selected at each occurrence from the group consisting of —CH2—, —CHR8—, —O—, —S(O)n—, —NH—, —NR8—, C(═O)—, —C(═O)NH—, —C(═O)NR8—, —S(O)nNH—, S(O)nNR8—, NHC(═O)—, —NR8C(═O)—, —NHS(O)n—, and —NR8S(O)n—;
R7 and R8 are independently selected at each occurrence from hydrogen, and straight, branched, and cyclic alkyl groups, and (cycloalkyl)alkyl groups, said straight, branched, and cyclic alkyl groups, and (cycloalkyl)alkyl groups consisting of 1 to 8 carbon atoms, and containing zero or one or more double or triple bonds, each of which 1 to 8 carbon atoms may be further substituted with one or more substituent(s) independently selected from oxo, hydroxy, halogen, amino, cyano, nitro, haloalkyl, haloalkoxy, —O(C1-4alkyl), —NH(C1-4alkyl), —N(C1-4alkyl)(C1-4alkyl), —NHC(O)(C1-4alkyl), —N(C1-4alkyl)C(O)(C1-4alkyl), —NHS(O)n(C1-4alkyl), —S(O)n(C1-4alkyl), —S(O)nNH(C1-4alkyl), —S(O)nN(C1-4alkyl3)(C1-4alkyl4) where C1-4alkyl3 and C1-4alkyl4 may be joined to form a heterocycloalkyl ring consisting of from 5 to 8 ring atoms and containing 1, 2, or 3 heteroatoms selected from N, O, and S, and Y′;
Y and Y′ are independently selected at each occurrence from 3- to 8-membered carbocyclic or heterocyclic groups which are saturated, unsaturated, or aromatic, which may be further substituted with one or more substituents independently selected from halogen, oxo, hydroxy, amino, nitro, cyano, C1-4alkyl, C1-4alkoxy, halo(C1-4)alkyl, halo(C1-4)alkoxy, mono- or di(C1-4)alkylamino, and C1-4alkylthio;
 wherein said 3- to 8-memberered heterocyclic groups contain one or more heteroatom(s) independently selected from N, O, and S; and
n is independently chosen at each occurrence from 0, 1, and 2.
51. A compound or salt according to claim 50, wherein:
A is O or NRA, wherein RA is hydrogen or methyl.
52. A compound or salt according to claim 50, wherein:
A is O or NRA, wherein RA is hydrogen or methyl; and
R3 and R4 are independently chosen at each occurrence from the group consisting of hydrogen, halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6alkoxy, —NH(C1-6alkyl), and —N(C1-6alkyl)(C1-6alkyl).
53. A compound or salt according to claim 50, wherein:
A is O or NRA, wherein RA is hydrogen or methyl;
R3 is hydrogen; and
R4 is independently chosen at each occurrence from the group consisting of hydrogen, halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6alkoxy, —NH(C1-6alkyl), and —N(C1-6alkyl)(C1-6alkyl).
54. A compound or salt according to claim 50, wherein:
A is O or NRA, wherein RA is hydrogen or methyl;
R3 is hydrogen; and
R4 is independently chosen at each occurrence from hydrogen and C1-6alkyl.
55. A compound or salt according to claim 50, wherein:
A is NRA, wherein RA is hydrogen or methyl;
R3 is hydrogen; and
R4 is independently chosen at each occurrence from hydrogen, halo(C1-3)alkyl, and C1-6alkyl.
56. A compound or salt according to claim 50, wherein:
A is NRA, wherein RA is hydrogen or methyl;
R3 is hydrogen; and
R4 is independently chosen at each occurrence from hydrogen and C1-4alkyl.
57. A compound or salt according to claim 50 of the Formula
Figure US20040176443A1-20040909-C00228
wherein:
R4 is independently chosen at each occurrence from hydrogen and C1-4alkyl.
58. A compound or salt according to claim 57 of the formula:
Figure US20040176443A1-20040909-C00229
wherein
R5B and R9B are independently chosen from hydrogen, halogen, cyano, nitro, halo(C1-2)alkyl, halo(C1-2)alkoxy, amino, C1-4alkyl, and C1-2alkoxy; and
R10 is independently chosen at each occurrence from hydrogen, halogen, and C1-4 alkyl.
59. A compound or salt according to claim 58 wherein:
R9 is selected from the group consisting of halogen, cyano, —N(SO2CH3)2, —SO2NH2, halo(C1-3)alkyl, C1-3alkoxy, —NH(C1-3alkyl), and —N(C1-3alkyl)(C1-3alkyl).
60. A compound or salt according to claim 57, wherein:
R5B and R9B are independently chosen from hydrogen, halogen, cyano, nitro, halo(C1-2)alkyl, halo(C1-2)alkoxy, amino, C1-4alkyl, and C1-2alkoxy.
61. A compound or salt according to claim 57, wherein:
R5B represents 0 or 1 substituents chosen from halogen, cyano, nitro, halo(C1-2)alkyl, halo(C1-2)alkoxy, amino, C1-4alkyl, and C1-2alkoxy; and
R9B represents 0 or 1 substituents chosen from halogen, cyano, nitro, halo(C1-2)alkyl, and C1-2alkyl, and C1-2alkoxy.
62. A compound or salt according to claim 57, wherein:
R9 is selected from the group consisting of halogen, cyano, —N(SO2CH3)2, —SO2NH2, halo(C1-3)alkyl, C1-3alkoxy, —NH(C1-3alkyl), and —N(C1-3alkyl)(C1-3alkyl);
R5B represents 0 or 1 substituents chosen from halogen, cyano, nitro, halo(C1-2)alkyl, halo(C1-2)alkoxy, amino, C1-4alkyl, and C1-2alkoxy; and
R9B represents 0 or 1 substituents chosen from halogen, cyano, nitro, halo(C1-2)alkyl, and C1-2alkyl, and C1-2alkoxy.
63. A compound or salt according to claim 57, wherein:
R5 is selected from the group consisting of bromo, fluoro, iodo, halo(C1-6)alkyl, halo(C3-6)alkoxy, C3-6alkyl substituted with 0-3 R6, C2-alkenyl substituted with 0-3 R6, Y, —(C═O)Y, —(CH2)Y, and —(CH(CN))Y;
R9 is selected from the group consisting of halogen, cyano, —N(SO2CH3)2, —SO2NH2, halo(C1-2)alkyl,
 C1-3alkoxy, —NH(C1-6alkyl), and —N(C1-6alkyl)(C1-6alkyl);
R5B represents 0 or 1 substituents chosen from halogen, cyano, nitro, halo(C1-2)alkyl, halo(C1-2)alkoxy, amino, C1-4alkyl, and C1-2alkoxy; and
R9B represents 0 or 1 substituents chosen from halogen, cyano, nitro, halo(C1-2)alkyl, and C1-2alkyl, and C1-2alkoxy.
64. A compound or salt according to claim 63, wherein:
R6 is independently selected at each occurrence from the group consisting of cyano, halogen, hydroxy, C1-4alkyl, C1-4alkoxy, —NH(C1-4alkyl), and —N(C1-4alkyl)(C1-4alkyl) and Y; and
Y is independently selected at each occurrence from C3-8 cycloalkyl, piperidinyl, piperazinyl, tetrahydropyranyl, dihydropyranyl, morpholinyl, thiomorpholinyl, phenyl, pyridyl, pyrazinyl, pyrimidinyl, thiazolyl, thienyl, and imidazolyl, each of which may be further substituted with one or more substituents independently selected from halogen, oxo, hydroxy, amino, nitro, cyano, C1-4alkyl, C1-4alkoxy, halo(C1-4)alkyl, halo(C1-4)alkoxy, mono- or di(C1-4)alkylamino, and C1-4alkylthio.
65. A compound or salt according to claim 63, wherein:
R9 is cyano, trifluoromethyl, chloro, or iodo; and
R9B is hydrogen.
66. A compound according to claim 50, which is N-(4-tert-butylphenyl)-4-(3-chloropyridin-2-yl)piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
67. A compound according to claim 50, which is (2R)-4-(3-chloropyridin-2-yl)-N-(4-cyclohexylphenyl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
68. A compound according to claim 50, which is (2R)-4-(3-chloropyridin-2-yl)-2-methyl-N-[4-(trifluoromethyl)phenyl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
69. A compound according to claim 50, which is (2R)-N-(4-tert-butylphenyl)-4-(3-chloropyridin-2-yl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
70. A compound according to claim 50, which is (2R)-4-(3-chloropyridin-2-yl)-N-(4-isopropylphenyl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
71. A compound according to claim 50, which is (2S)-4-(3-chloropyridin-2-yl)-N-(4-trifluoromethylphenyl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
72. A compound according to claim 50, which is (2S)-N-(4-tert-butylphenyl)-4-(3-chloropyridin-2-yl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
73. A compound according to claim 50, which is (2S)-4-(3-chloropyridin-2-yl)-N-(4-isopropylphenyl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
74. A compound according to claim 50, which is (2R)-4-(3-chloropyridin-2-yl)-2-methyl-N-(4-piperidin-1-ylphenyl)piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
75. A compound according to claim 50, which is (2R)-4-(3-chloropyridin-2-yl)-N-[2-fluoro-4-(trifluoromethyl)phenyl]-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
76. A compound according to claim 50, which is (2R)-2-methyl-N-[4-(trifluoromethyl)phenyl]-4-[3-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
77. A compound according to claim 50, which is (2R)-N-(4-tert-butylphenyl)-2-methyl-4-[3-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
78. A compound according to claim 50, which is (2R)-N-(4-isopropylphenyl)-2-methyl-4-[3-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
79. A compound according to claim 50, which is 4-(3-chloropyridin-2-yl)-2,6-dimethyl-N-[4-(trifluoromethyl)phenyl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
80. A compound according to claim 50, which is N-(4-tert-butylphenyl)-4-(3-chloropyridin-2-yl)-2,6-dimethylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
81. A compound according to claim 50, which is 4-(3-chloropyridin-2-yl)-N-(4-isopropylphenyl)-2,6-dimethylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
82. A compound according to claim 50, which is (2R)-N-(4-cyclohexylphenyl)-2-methyl-4-[3-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
83. A compound according to claim 50, which is 4-(3-chloropyridin-2-yl)-N-(4-cyclohexylphenyl)-2,6-dimethylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
84. A compound according to claim 50, which is (2R)-4-(3-chloropyridin-2-yl)-N-(4-cyclopentylphenyl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
85. A compound according to claim 50, which is (2R)-N-(4-cyclopentylphenyl)-2-methyl-4-[3-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
86. A compound according to claim 50, which is (2R)-N-(4-tert-butylphenyl)-4-[3-(dimethylamino)pyridin-2-yl]-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
87. A compound according to claim 50, which is (2R)-4-[3-(dimethylamino)pyridin-2-yl]-2-methyl-N-[4-(trifluoromethyl)phenyl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
88. A compound according to claim 50, which is (2R)-N-(4-tert-butylphenyl)-4-(3-methoxypyridin-2-yl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
89. A compound according to claim 50, which is (2R)-4-(3-methoxypyridin-2-yl)-2-methyl-N-[4-(trifluoromethyl)phenyl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
90. A compound according to claim 50, which is (2R)-N-(4-cyclohexylphenyl)-4-(3-methoxypyridin-2-yl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
91. A compound according to claim 50, which is (2R)-4-(3-chloropyridin-2-yl)-N-[4-(3,6-dihydro-2H-pyran-4-yl)phenyl]-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
92. A compound according to claim 50, which is (2R)-4-(3-chloropyridin-2-yl)-2-methyl-N-(4-tetrahydro-2H-pyran-4-ylphenyl)piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
93. A compound according to claim 50, which is (2R)-4-(3-chloropyridin-2-yl)-N-[4-(4-hydroxytetrahydro-2H-pyran-4-yl)phenyl]-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
94. A compound according to claim 50, which is (2R)-N-[4-(4-hydroxytetrahydro-2H-pyran-4-yl)phenyl]-2-methyl-4-[3-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
95. A compound according to claim 50, which is (2R)-4-(3-chloropyridin-2-yl)-2-methyl-N-[4-(2-methyl-1,3-thiazol-4-yl)phenyl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
96. A compound according to claim 50, which is (2R)-4-(3-chloropyridin-2-yl)-N-[4-(2-ethyl-1,3-thiazol-4-yl)phenyl]-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
97. A compound according to claim 50, which is (2R)-4-(3-chloropyridin-2-yl)-N-[4-(2-methoxy-1,1-dimethylethyl)phenyl]-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
98. A compound according to claim 50, which is (2R)-N-[4-(2-methoxy-1,1-dimethylethyl)phenyl]-2-methyl-4-[3-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
99. A compound according to claim 50, which is (2R)-4-(3-chloropyridin-2-yl)-N-[4-(1-cyano-1-methylethyl)phenyl]-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
100. A compound according to claim 50, which is (2R)-N-[4-(1-cyano-1-methylethyl)phenyl]-2-methyl-4-[3-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
101. A compound according to claim 50, which is N-(4-tert-butylphenyl)-4-(3-chloropyridin-2-yl)-2-ethylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
102. A compound according to claim 50, which is 4-(3-chloropyridin-2-yl)-2-ethyl-N-[4-(trifluoromethyl)phenyl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
103. A compound according to claim 50, which is 4-(3-chloropyridin-2-yl)-2-ethyl-N-(4-isopropylphenyl)piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
104. A compound according to claim 50, which is N-(4-tert-butylphenyl)-2-ethyl-4-[3-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
105. A compound according to claim 50, which is 2-ethyl-N-[4-(trifluoromethyl)phenyl]-4-[3-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
106. A compound according to claim 50, which is 2-ethyl-N-(4-isopropylphenyl)-4-[3-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
107. A compound according to claim 50, which is 2-tert-butyl-N-(4-tert-butylphenyl)-4-(3-chloropyridin-2-yl)piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
108. A compound according to claim 50, which is 2-tertbutyl-N-[4-(trifluoromethyl)phenyl]-4-[3-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
109. A compound according to claim 50, which is N-(4-tert-butylphenyl)-4-(3-chloropyridin-2-yl)-2-isopropylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
110. A compound according to claim 50, which is N-(4-tert-butylphenyl)-2-isopropyl-4-[3-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
111. A compound according to claim 50, which is 2-isopropyl-N-[4-(trifluoromethyl)phenyl]-4-[3-(trifluoromethyl) pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
112. A compound according to claim 50, which is 2-isopropyl-N-(4-isopropylphenyl)-4-[3-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
113. A compound according to claim 50, which is (2R)-4-(3-fluoropyridin-2-yl)-2-methyl-N-[4-(trifluoromethyl)phenyl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
114. A compound according to claim 50, which is (2R)-N-(4-tert-butylphenyl)-4-(3-fluoropyridin-2-yl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
115. A compound according to claim 50, which is (2R)-4-(3-fluoropyridin-2-yl)-N-(4-isopropylphenyl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
116. A compound according to claim 50, which is (2R)-N-(4-cyclohexylphenyl)-4-(3-fluoropyridin-2-yl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
117. A compound according to claim 50, which is (2R)-N-(4-cyclopentylphenyl)-4-(3-fluoropyridin-2-yl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
118. A compound according to claim 50, which is (2R)-N-(4-tert-butylphenyl)-4-(3-cyanopyridin-2-yl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
119. A compound according to claim 50, which is (2R)-4-(3-cyanopyridin-2-yl)-2-methyl-N-[4-(trifluoromethyl)phenyl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
120. A compound according to claim 50, which is (2R)-4-(3-chloropyridin-2-yl)-N-{4-[cyano(phenyl)methyl]phenyl}-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
121. A compound according to claim 50, which is (2R)-4-(3-chloropyridin-2-yl)-2-methyl-N-[3-methyl-4-(trifluoromethyl)phenyl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
122. A compound according to claim 50, which is (2R)-4-(3-fluoropyridin-2-yl)-2-methyl-N-[3-methyl-4-(trifluoromethyl)phenyl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
123. A compound according to claim 50, which is (2R)-4-{3-[bis(methylsulfonyl)amino]pyridin-2-yl}-N-(4-tert-butylphenyl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
124. A compound according to claim 50, which is (2R)-2-methyl-N-[3-methyl-4-(trifluoromethyl)phenyl]-4-[3-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
125. A compound according to claim 50, which is (2R)-4-(3-chloropyridin-2-yl)-2-methyl-N-{4-[1-(trifluoromethyl)vinyl]phenyl} piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
126. A compound according to claim 50, which is (2R)-2-methyl-4-[3-(trifluoromethyl)pyridin-2-yl]-N-{4-[1-(trifluoromethyl)vinyl]phenyl}piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
127. A compound according to claim 50, which is (2R)-4-(3-fluoropyridin-2-yl)-2-methyl-N-{4-[1-(trifluoromethyl)vinyl]phenyl}piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
128. A compound according to claim 50, which is (2R)-N-(4-sec-butylphenyl)-4-(3-fluoropyridin-2-yl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
129. A compound according to claim 50, which is (2R)-2-methyl-N-[4-(2,2,2-trifluoro-1-methylethyl)phenyl]-4-[3-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
130. A compound according to claim 50, which is (2R)-4-(3-fluoropyridin-2-yl)-2-methyl-N-[4-(2,2,2-trifluoro-1-methylethyl)phenyl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
131. A compound according to claim 50, which is (2R)-4-(3-chloro-5-nitropyridin-2-yl)-2-methyl-N-[4-(trifluoromethyl)phenyl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
132. A compound according to claim 50, which is (2R)-4-(5-amino-3-chloropyridin-2-yl)-2-methyl-N-[4-(trifluoromethyl)phenyl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
133. A compound according to claim 50, which is (2R)-4-(3-fluoropyridin-2-yl)-N-[3-fluoro-4-(trifluoromethyl)phenyl]-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
134. A compound according to claim 50, which is (2R)-N-[3-fluoro-4-(trifluoromethyl)phenyl]-2-methyl-4-[3-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
135. A compound according to claim 50, which is (2R)-4-(3-chloropyridin-2-yl)-2-methyl-N-[4-(2,2,2-trifluoro-1-methylethyl)phenyl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
136. A compound according to claim 50, which is (2R)-4-(3-chloropyridin-2-yl)-2-methyl-N-(2,2,4,4-tetrafluoro-4H-1,3-benzodioxin-6-yl)piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
137. A compound according to claim 50, which is (2R)-4-(3-fluoropyridin-2-yl)-2-methyl-N-(2,2,4,4-tetrafluoro-4H-1,3-benzodioxin-6-yl)piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
138. A compound according to claim 50, which is (2R)-2-methyl-N-(2,2,4,4-tetrafluoro-4H-1,3-benzodioxin-6-yl)-4-[3-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
139. A compound according to claim 50, which is (2R)-4-[3-(aminosulfonyl)pyridin-2-yl]-N-(4-tert-butylphenyl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
140. A compound according to claim 50, which is (2R)-N-(4-benzoylphenyl)-4-(3-chloropyridin-2-yl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
141. A compound according to claim 50, which is (2R)-4-(3-chloropyridin-2-yl)-N-(4-iodophenyl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
142. A compound according to claim 50, which is (2R)-4-(3-chloropyridin-2-yl)-2-methyl-N-{4-[2,2,2-trifluoro-1,1-bis(trifluoromethyl)ethyl]phenyl}piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
143. A compound according to claim 50, which is (2R)-2-methyl-N-{4-[2,2,2-trifluoro-1,1-bis(trifluoromethyl)ethyl]phenyl}-4-[3-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
144. A compound according to claim 50, which is (2R)-N-(4-butylphenyl)-4-(3-chloropyridin-2-yl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
145. A compound according to claim 50, which is 2-(fluoromethyl)-N-[4-(trifluoromethyl)phenyl]-4-[3-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
146. A compound according to claim 50, which is (2R)-N-[4-bromo-3-(trifluoromethyl)phenyl]-4-(3-chloropyridin-2-yl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
147. A compound according to claim 50, which is (2R)-N-[4-bromo-3-(trifluoromethyl)phenyl]-2-methyl-4-[3-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
148. A compound according to claim 50, which is (2R)-4-(3-chloropyridin-2-yl)-N-[4-fluoro-3-(trifluoromethyl)phenyl]-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
149. A compound according to claim 50, which is (2R)-N-[4-fluoro-3-(trifluoromethyl)phenyl]-2-methyl-4-[3-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
150. A compound according to claim 50, which is (2R)-4-(3-chloropyridin-2-yl)-2-methyl-N-{4-[1,2,2,2-tetrafluoro-1-(trifluoromethyl)ethyl]phenyl}piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
151. A compound according to claim 50, which is (2R)-2-methyl-N-{4-[1,2,2,2-tetrafluoro-1-(trifluoromethyl)ethyl]phenyl}-4-[3-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
152. A compound according to claim 40, which is (2R)-N-(4-tert-butylphenyl)-4-(3-chloropyrazin-2-yl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
153. A compound according to claim 40, which is (2R)-4-(3-chloropyrazin-2-yl)-N-(4-isopropylphenyl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
154. A compound according to claim 40, which is (2R)-4-(3-chloropyrazin-2-yl)-2-methyl-N-[4-(trifluoromethyl)phenyl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
155. A compound according to claim 40, which is (2R)-4-(3-chloropyrazin-2-yl)-2-methyl-N-{4-[1,2,2,2-tetrafluoro-1-(trifluoromethyl)ethyl]phenyl}piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
156. A compound according to claim 40, which is (2R)-4-(3-chloropyrazin-2-yl)-2-methyl-N-{4-cyclopentyl-phenyl}piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
157. A compound according to claim 40, which is (2R)-4-(3-chloropyrazin-2-yl)-2-methyl-N-{4-cyclohexyl-phenyl}piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
158. A compound according to claim 42, which is 4-(3-chloropyridin-2-yl)-N-[5-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
159. A compound according to claim 42, which is (2R)-4-(3-chloropyridin-2-yl)-2-methyl-N-[5-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
160. A compound according to claim 42, which is (2R)-4-(3-chloropyridin-2-yl)-2-methyl-N-[6-(trifluoromethyl)pyridin-3-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
161. A compound of the Formula:
Figure US20040176443A1-20040909-C00230
or a pharmaceutically acceptable salt thereof, wherein:
A is absent or is selected from the group consisting of O, S, NRA, CRBRB′, NRACRBRB′, CRBRB′NRA, —CRA═CRB—, and C3H4; where RA, RB, and RB′ are independently selected at each occurrence from hydrogen or C1-6 alkyl;
R3 and R4 are independently chosen at each occurrence from the group consisting of hydrogen, halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6alkoxy, —NH(C1-6alkyl), and —N(C1-6alkyl)(C1-6alkyl)
R5 is selected from the group consisting of halogen, halo(C1-6)alkyl, C3-6alkyl substituted with 0-3 R6, C2-6alkenyl substituted with 0-3 R6, (C3-8cycloalkyl)C1-4alkyl substituted with 0-3 R6, and Y;
R5B and R9B each represent from 0 to 2 substituents and are independently chosen from halogen, cyano, nitro, halo(C1-2)alkyl, halo(C1-2)alkoxy, amino, C1-4alkyl, and C1-2alkoxy;
R6 is independently selected at each occurrence from the group consisting of cyano, halogen, hydroxy, C1-4alkyl, C1-4alkoxy, —NH(C1-4alkyl), and —N(C1-4alkyl)(C1-4alkyl) and Y;
Y is independently selected at each occurrence from C3-8 cycloalkyl, piperidinyl, piperazinyl, tetrahydropyranyl, dihydropyranyl, morpholinyl, thiomorpholinyl, phenyl, pyridyl, pyrazinyl, pyrimidinyl, thiazolyl, thienyl, and imidazolyl, each of which may be further substituted with one or more substituents independently selected from halogen, oxo, hydroxy, amino, nitro, cyano, C1-4alkyl, C1-4alkoxy, halo(C1-4)alkyl, halo(C1-4)alkoxy, mono- or di(C1-4)alkylamino, and C1-4alkylthio.
162. A compound or salt according to claim 161 of the Formula:
Figure US20040176443A1-20040909-C00231
wherein
R4 is independently selected at each occurrence from hydrogen and C1-4alkyl.
163. A compound or salt according to claim 162, wherein:
R5 is selected from the group consisting of halo(C1-6)alkyl, C3-6alkyl, (C3-8cycloalkyl)C1-4alkyl, and Y;
R5B and R9B each represent from 0 to 1 substituents and are independently chosen from halogen, cyano, nitro, halo(C1-2)alkyl, halo(C1-2)alkoxy, amino, C1-4alkyl, and C1-2alkoxy;
Y is selected from C3-8 cycloalkyl, piperidinyl, piperazinyl, tetrahydropyranyl, dihydropyranyl, morpholinyl, thiomorpholinyl, phenyl, pyridyl, pyrazinyl, pyrimidinyl, thiazolyl, thienyl, and imidazolyl.
164. A compound according to claim 161, which is (2R)-4-isoquinolin-1-yl-2-methyl-N-[4-(trifluoromethyl)phenyl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
165. A compound according to claim 161, which is (2R)-N-(4-tert-butylphenyl)-4-isoquinolin-1-yl-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
166. A compound according to claim 161, which is (2R)-N-(4-isopropylphenyl)-4-isoquinolin-1-yl-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
167. A compound according to claim 161, which is (2R)-N-(4-cyclopentylphenyl)-4-isoquinolin-1-yl-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
168. A compound according to claim 161, which is (2R)-N-(4-cyclohexylphenyl)-4-isoquinolin-1-yl-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
169. A compound of the Formula:
Figure US20040176443A1-20040909-C00232
or a pharmaceutically acceptable salt thereof, wherein:
A is absent or is selected from the group consisting of O, S, NRA, CRBRB′, NRACRBRB′, CRBRB′NRA, —CRA═CRB—, and C3H4; where RA, RB, and RB′ are independently selected at each occurrence from hydrogen or C1-6 alkyl;
R3 and R4 are independently chosen at each occurrence from the group consisting of hydrogen, halogen, cyano, nitro, halo(C1-6)alkyl, halo(C1-6)alkoxy, hydroxy, amino, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C1-6alkoxy, —NH(C1-6alkyl), and —N(C1-6alkyl)(C1-6alkyl);
R5B, R5C, and R9B each represent from 0 to 2 substituents and are independently chosen from halogen, cyano, nitro, halo(C1-2)alkyl, halo(C1-2)alkoxy, amino, C1-4alkyl, and C1-2alkoxy; and
R9 is selected from the group consisting of halogen, cyano, —N(SO2CH3) 2, —SO2NH2, halo(C1-3)alkyl, C1-3alkoxy, —NH(C1-3alkyl), and —N(C1-3alkyl)(C1-3alkyl)
170. A compound or salt according to claim 169 of the Formula:
Figure US20040176443A1-20040909-C00233
wherein
R4 is independently selected at each occurrence from hydrogen and C1-4alkyl.
171. A compound or salt according to claim 170, wherein:
R9 is selected from the group consisting of halogen and halo(C1-2)alkyl; and
R5B and R9B each represent from 0 to 1 substituents and are independently chosen from halogen, cyano, nitro, halo(C1-2)alkyl, halo(C1-2)alkoxy, amino, C1-4alkyl, and C1-2alkoxy.
172. A compound according to claim 169, which is (2R)-4-(3-chloropyridin-2-yl)-N-(9H-fluoren-2-yl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
173. A compound according to claim 169, which is (2R)-N-(9H-fluoren-2-yl)-2-methyl-4-[3-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
174. A compound according to claim 38, which is (2R)-N-(4-tert-butylcyclohexyl)-4-(3-chloropyridin-2-yl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
175. A compound according to claim 38, which is (2R)-4-(3-chloropyridin-2-yl)-N-(4-isopropylcyclohexyl)-2-methylpiperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
176. A compound according to claim 38, which is (2R)-N-(4-isopropylcyclohexyl)-2-methyl-4-[3-(trifluoromethyl)pyridin-2-yl]piperazine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
177. A method of reducing the calcium conductance of a capsaicin receptor, which method comprises: contacting a first solution comprising a fixed concentration of a capsaicin receptor agonist and a compound or salt of claim 50 with a cell expressing the capsaicin receptor, wherein the compound or salt is present in the solution at a concentration sufficient to produce a detectable reduction of the calcium mobilization effects of the capsaicin receptor agonist when tested in an in vitro assay in which cells expressing a capsaicin receptor are contacted with a second solution comprising the fixed concentration of capsaicin receptor agonist and the compound or salt.
178. The method of claim 177 wherein the cell expressing the capsaicin receptor is a neuronal cell that is contacted in vivo in an animal, and wherein the first solution is a body fluid of said animal.
179. The method of claim 177 wherein the animal is a human patient.
180. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound or salt of claim 50.
181. A package comprising a pharmaceutical composition of claim 180 in a container and further comprising indicia comprising instructions for using the composition to alleviate pain.
182. A package comprising a pharmaceutical composition of claim 180 in a container and further comprising indicia comprising instructions for using the composition to treat a patient suffering from urinary incontinence.
183. A package comprising a pharmaceutical composition of claim 180 in a container and further comprising indicia comprising instructions for using the composition to alleviate symptoms of exposure to capsaicin or tear gas.
184. A compound or salt of claim 50 wherein, in an in vitro assay of capsaicin receptor antagonism, the compound or salt exhibits capsaicin receptor antagonist activity, but in an in vitro assay of capsaicin receptor agonism the compound does not exhibit detectable agonist activity.
185. A compound or salt of claim 50 wherein a dose of the compound or salt that is twice the minimum dose sufficient to provide analgesia in an animal model for determining pain relief does not produce sedation in an animal model assay of sedation.
186. A method of treating a mammal suffering from at least one symptom selected from the group consisting of symptoms of exposure to capsaicin, symptoms of burns or irritation due to exposure to heat, symptoms of burns or irritation due to exposure to light, symptoms of burns, bronchoconstriction or irritation due to exposure to tear gas, and symptoms of burns or irritation due to exposure to acid, the method comprising administering to the mammal a therapeutic dose of a compound that is a high potency capsaicin receptor antagonist in an in vitro assay of capsaicin receptor antagonism, is not a capsaicin analog; wherein the therapeutic dose contains an amount of the compound that is effective to reduce severity of at least one of said at least one symptom.
187. The method of claim 186 wherein the compound is a compound or salt of any of claims 1-176.
188. A method of treating a mammal suffering from neuropathic pain, the method comprising administering to the mammal a therapeutically effective amount of a compound that is a high potency capsaicin receptor antagonist in an in vitro assay of capsaicin receptor antagonism.
189. A method of treating a mammal suffering from peripheral-nerve-mediated pain, the method comprising administering to the mammal a therapeutic dose of a compound that is a capsaicin receptor antagonist, wherein the compound is a high potency capsaicin receptor antagonist in an in vitro assay of capsaicin receptor antagonism and is not a capsaicin analog,
wherein the therapeutic dose contains an amount of the compound that is effective to reduce the peripheral-nerve-mediated pain.
190. The method of claim 189 wherein the compound is a compound or salt of claim 50.
191. The method of claim 189 wherein the pain is neuropathic pain.
192. The method of claim 190 wherein the pain is associated with a condition selected from the group consisting of postmastectomy pain syndrome, stump pain, phantom limb pain, oral neuropathic pain, Charcot's pain, toothache, venomous snake bite, spider bite, insect sting, postherpetic neuralgia, diabetic neuropathy, reflex sympathetic dystrophy, trigeminal neuralgia, osteoarthritis, rheumatoid arthritis, fibromyalgia, Guillain-Barre syndrome, meralgia paresthetica, burning-mouth syndrome, bilateral peripheral neuropathy, causalgia, sciatic neuritis, peripheral neuritis, polyneuritis, optic neuritis, postfebrile neuritis, migrating neuritis, segmental neuritis, Gombault's neuritis, neuronitis, cervicobrachial neuralgia, cranial neuralgia, geniculate neuralgia, glossopharyngial neuralgia, migranous neuralgia, idiopathic neuralgia, intercostals neuralgia, mammary neuralgia, mandibular joint neuralgia, Morton's neuralgia, nasociliary neuralgia, occipital neuralgia, red neuralgia, Sluder's neuralgia, splenopalatine neuralgia, supraorbital neuralgia, vidian neuralgia, sinus headache, tension headache, labor, childbirth, intestinal gas, menstruation, cancer, and trauma.
193. A compound or salt of claim 50 wherein the compound or salt is not addictive.
194. The use of a compound according to claim 1, 4, 9, 31, or 50 for the manufacture of a medicament for the treatment of pain.
195. The use of a compound according to claim 1, 4, 9, 31, or 50 for the manufacture of a medicament for the treatment of neuropathic pain.
196. The use of a compound according to claim 1, 4, 9, 31, or 50 for the manufacture of a medicament for the treatment of the pain associated with a condition selected from the group consisting of postmastectomy pain syndrome, stump pain, phantom limb pain, oral neuropathic pain, Charcot's pain, toothache, venomous snake bite, spider bite, insect sting, postherpetic neuralgia, diabetic neuropathy, reflex sympathetic dystrophy, trigeminal neuralgia, osteoarthritis, rheumatoid arthritis, fibromyalgia, Guillain-Barre syndrome, meralgia paresthetica, burning-mouth syndrome, bilateral peripheral neuropathy, causalgia, sciatic neuritis, peripheral neuritis, polyneuritis, optic neuritis, postfebrile neuritis, migrating neuritis, segmental neuritis, Gombault's neuritis, neuronitis, cervicobrachial neuralgia, cranial neuralgia, geniculate neuralgia, glossopharyngial neuralgia, migranous neuralgia, idiopathic neuralgia, intercostals neuralgia, mammary neuralgia, mandibular joint neuralgia, Morton's neuralgia, nasociliary neuralgia, occipital neuralgia, red neuralgia, Sluder's neuralgia, splenopalatine neuralgia, supraorbital neuralgia, vidian neuralgia, sinus headache, tension headache, labor, childbirth, intestinal gas, menstruation, cancer, and trauma.
197. A compound of the Formula
Figure US20040176443A1-20040909-C00234
or a pharmaceutically acceptable salt thereof wherein:
R4 is methyl or hydrogen;
R9B represents 0-2 substituents independently chosen from: halogen, cyano, nitro, halo(C1-2)alkyl, halo(C1-2)alkoxy, amino, C1-4alkyl, and C1-2alkoxy; and
Ar1 is 2,4-dichlorphenyl or 3-nitro-4-chlorophenyl.
198. A compound of the Formula
Figure US20040176443A1-20040909-C00235
or a pharmaceutically acceptable salt thereof wherein:
R9 is chloro or trifluoromethyl; and
R5B and R9B independently represent from 0-2 substituents on each of the rings on which they occur and are independently chosen from: halogen, cyano, nitro, halo(C1-2)alkyl, halo(C1-2)alkoxy, amino, C1-4alkyl, and C1-2alkoxy.
US10/799,286 2000-07-20 2004-03-12 Capsaicin receptor ligands Abandoned US20040176443A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US10/799,286 US20040176443A1 (en) 2000-07-20 2004-03-12 Capsaicin receptor ligands
US12/057,808 US20090082362A1 (en) 2000-07-20 2008-03-28 Use of capsaicin receptor antagonists to treat symptoms of tear gas exposure

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US21952900P 2000-07-20 2000-07-20
US23072600P 2000-09-07 2000-09-07
US28022301P 2001-03-30 2001-03-30
US09/910,442 US6723730B2 (en) 2000-07-20 2001-07-20 Capsaicin receptor ligands
US10/799,286 US20040176443A1 (en) 2000-07-20 2004-03-12 Capsaicin receptor ligands

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/910,442 Continuation US6723730B2 (en) 2000-07-20 2001-07-20 Capsaicin receptor ligands

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/057,808 Continuation US20090082362A1 (en) 2000-07-20 2008-03-28 Use of capsaicin receptor antagonists to treat symptoms of tear gas exposure

Publications (1)

Publication Number Publication Date
US20040176443A1 true US20040176443A1 (en) 2004-09-09

Family

ID=27396663

Family Applications (3)

Application Number Title Priority Date Filing Date
US09/910,442 Expired - Fee Related US6723730B2 (en) 2000-07-20 2001-07-20 Capsaicin receptor ligands
US10/799,286 Abandoned US20040176443A1 (en) 2000-07-20 2004-03-12 Capsaicin receptor ligands
US12/057,808 Abandoned US20090082362A1 (en) 2000-07-20 2008-03-28 Use of capsaicin receptor antagonists to treat symptoms of tear gas exposure

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/910,442 Expired - Fee Related US6723730B2 (en) 2000-07-20 2001-07-20 Capsaicin receptor ligands

Family Applications After (1)

Application Number Title Priority Date Filing Date
US12/057,808 Abandoned US20090082362A1 (en) 2000-07-20 2008-03-28 Use of capsaicin receptor antagonists to treat symptoms of tear gas exposure

Country Status (12)

Country Link
US (3) US6723730B2 (en)
EP (1) EP1301484A2 (en)
JP (1) JP2004525071A (en)
KR (1) KR20030024799A (en)
CN (1) CN1443170A (en)
AU (2) AU2001280667B2 (en)
BR (1) BR0112631A (en)
CA (1) CA2415606A1 (en)
IL (1) IL153884A0 (en)
MX (1) MXPA03000458A (en)
NZ (1) NZ523526A (en)
WO (1) WO2002008221A2 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060035939A1 (en) * 2004-07-14 2006-02-16 Japan Tobacco Inc. 3-Aminobenzamide compounds and inhibitors of vanilloid receptor subtype 1 (VR1) activity
US20060035882A1 (en) * 2004-07-15 2006-02-16 Japan Tobacco Inc. Condensed benzamide compounds and inhibitors of vanilloid receptor subtype 1 (vr1) activity
US20070149517A1 (en) * 2005-12-28 2007-06-28 Japan Tobacco Inc. 3,4-dihydrobenzoxazine compounds and inhibitors of vanilloid receptor subtype 1 (vri) activity
US20090163508A1 (en) * 2007-10-10 2009-06-25 Takeda Pharmaceutical Company Limited Amide compound
US20100120862A1 (en) * 2008-10-24 2010-05-13 Laykea Tafesse Monocyclic compounds and their use as trpv1 ligands
US20100130499A1 (en) * 2008-10-24 2010-05-27 Laykea Tafesse Bicycloheteroaryl compounds and their use as trpv1 ligands
WO2016100161A1 (en) * 2014-12-19 2016-06-23 Merck Sharp & Dohme Corp. Ethyldiamine orexin receptor antagonists

Families Citing this family (144)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MXPA03000458A (en) * 2000-07-20 2004-06-02 Neurogen Corp Capsaicin receptor ligands.
WO2002016319A1 (en) 2000-08-21 2002-02-28 Pacific Corporation Novel thiourea compounds and the pharmaceutical compositions containing the same
AU2001280229B2 (en) 2000-08-21 2006-12-07 Pacific Corporation Novel thiourea derivatives and the pharmaceutical compositions containing the same
US7867516B2 (en) 2001-01-29 2011-01-11 Shionogi & Co., Ltd. Medicinal preparation containing 5-methyl-1-phenyl-2-(1h)-pyridone as active ingredient
US7208497B2 (en) * 2001-07-02 2007-04-24 Novo Nordisk A/S Substituted piperazines and diazepanes
PE20030417A1 (en) 2001-09-13 2003-08-06 Smithkline Beecham Plc DERIVATIVES OF UREA AS ANTAGONISTS OF THE VAINILLOID RECEPTOR
PL371587A1 (en) * 2002-01-17 2005-06-27 Neurogen Corporation Substituted quinazolin-4-ylamine analogues as modulators of capsaicin receptors
DK1472225T3 (en) * 2002-02-01 2010-08-09 Euro Celtique Sa 2-Piperazine pyridines useful for the treatment of pain
WO2003068749A1 (en) * 2002-02-15 2003-08-21 Glaxo Group Limited Vanilloid receptor modulators
US7074805B2 (en) 2002-02-20 2006-07-11 Abbott Laboratories Fused azabicyclic compounds that inhibit vanilloid receptor subtype 1 (VR1) receptor
DE60325834D1 (en) * 2002-02-20 2009-03-05 Abbott Lab CONDENSED AZABICYCLIC COMPOUNDS AS INHIBITORS OF VANILLOID RECEPTOR 1 (VR1)
US6974818B2 (en) * 2002-03-01 2005-12-13 Euro-Celtique S.A. 1,2,5-thiadiazol-3-YL-piperazine therapeutic agents useful for treating pain
GB0206876D0 (en) * 2002-03-22 2002-05-01 Merck Sharp & Dohme Therapeutic agents
US6864261B2 (en) 2002-05-02 2005-03-08 Euro-Celtique S.A. Therapeutic agents useful for treating pain
CA2486092A1 (en) 2002-05-17 2003-11-27 Janssen Pharmaceutica N.V. Aminotetralin-derived urea modulators of vanilloid vr1 receptor
US7589199B2 (en) 2002-06-12 2009-09-15 Chemocentryx, Inc. Substituted piperazines
ATE438401T1 (en) 2002-06-12 2009-08-15 Chemocentryx Inc 1-ARYL-4-SUBSTITUTED PIPERAZINE DERIVATIVES FOR USE AS CCR1 ANTAGONISTS FOR THE TREATMENT OF INFLAMMATION AND IMMUNE DISEASES
US7842693B2 (en) 2002-06-12 2010-11-30 Chemocentryx, Inc. Substituted piperazines
US7183411B2 (en) 2002-07-12 2007-02-27 Janssen Pharmaceutica N.V. Naphthol, quinoline and isoquinoline-derived urea modulators of vanilloid VR1 receptor
GB0221157D0 (en) * 2002-09-12 2002-10-23 Glaxo Group Ltd Novel treatment
US7157462B2 (en) * 2002-09-24 2007-01-02 Euro-Celtique S.A. Therapeutic agents useful for treating pain
AU2003273856A1 (en) * 2002-09-24 2004-04-19 Bayer Healthcare Ag Vr1 antagonists for the treatment of urological disorders
MXPA05003948A (en) * 2002-10-17 2005-06-17 Amgen Inc Benzimidazole derivatives and their use as vanilloid receptor ligands.
GB0226724D0 (en) 2002-11-15 2002-12-24 Merck Sharp & Dohme Therapeutic agents
PT1569896E (en) 2002-12-06 2007-11-15 Xention Ltd Tetrahydro-naphthalene derivatives
EP1569926A1 (en) 2002-12-13 2005-09-07 Neurogen Corporation Carboxylic acid, phosphate or phosphonate substituted quinazolin-4-ylamine analogues as capsaicin receptor modulators
AU2003300791A1 (en) * 2002-12-13 2004-07-09 Neurogen Corporation Combination therapy for the treatment of pain
US20060100245A1 (en) * 2002-12-19 2006-05-11 Neurogen Corporation Substituted biphenyl-4-carboxylic acid arylamide analogues
GB0229808D0 (en) 2002-12-20 2003-01-29 Glaxo Group Ltd Novel compositions
US7582635B2 (en) 2002-12-24 2009-09-01 Purdue Pharma, L.P. Therapeutic agents useful for treating pain
EP1818333A1 (en) * 2003-02-10 2007-08-15 Amgen Inc. Vanilloid receptor ligands and their use in treatments
US7265138B2 (en) * 2003-02-10 2007-09-04 Amgen Inc. Vanilloid receptor ligands and their use in treatments
US6933311B2 (en) * 2003-02-11 2005-08-23 Abbott Laboratories Fused azabicyclic compounds that inhibit vanilloid receptor subtype 1 (VR1) receptor
ATE450533T1 (en) * 2003-02-14 2009-12-15 Glaxo Group Ltd CARBOXAMIDE DERIVATIVES
JP2006519826A (en) * 2003-03-06 2006-08-31 グラクソ グループ リミテッド Heterocyclic urea derivatives for the treatment of pain
WO2004078744A2 (en) * 2003-03-07 2004-09-16 Glaxo Group Limited Urea derivatives and their use as vanilloid receptor antagonists in the treatment of pain
GB0305426D0 (en) * 2003-03-08 2003-04-16 Glaxo Group Ltd Novel compounds
EP1608373A4 (en) * 2003-03-19 2010-09-29 Exelixis Inc Tie-2 modulators and methods of use
JP2006522815A (en) * 2003-04-08 2006-10-05 アルゴルクス ファーマスーティカルズ,インク Production and purification of synthetic capsaicin
WO2004103954A1 (en) 2003-05-20 2004-12-02 Ajinomoto Co., Inc. Amide derivative
US7585878B2 (en) 2003-06-12 2009-09-08 Astellas Pharma Inc. Benzamide derivative or salt thereof
CN1820001A (en) * 2003-07-10 2006-08-16 神经能质公司 Substituted heterocyclic diarylamine analogues
US7129235B2 (en) 2003-07-11 2006-10-31 Abbott Laboratories Amides useful for treating pain
US20050009841A1 (en) * 2003-07-11 2005-01-13 Zheng Guo Zhu Novel amides useful for treating pain
TW200510373A (en) 2003-07-14 2005-03-16 Neurogen Corp Substituted quinolin-4-ylamine analogues
CA2531619A1 (en) * 2003-07-16 2005-01-27 Neurogen Corporation Biaryl piperazinyl-pyridine analogues
DK1867644T3 (en) 2003-07-24 2009-09-21 Euro Celtique Sa Heteroberl-tetrahydropyridyl compounds useful for treating or preventing pain
PL1942106T3 (en) 2003-08-01 2012-02-29 Euro Celtique Sa Therapeutic agents useful for treating pain
EP1651638A1 (en) * 2003-08-08 2006-05-03 Janssen Pharmaceutica N.V. Pyridyl piperazinyl ureas
GB0319150D0 (en) 2003-08-14 2003-09-17 Glaxo Group Ltd Novel compounds
JP2007520444A (en) * 2003-09-09 2007-07-26 ニューロジェン・コーポレーション Substituted bicyclic quinazolin-4-ylamine derivatives
ES2321719T3 (en) * 2003-10-01 2009-06-10 Xention Limited DERIVATIVES OF TETRAHIDRO-NAFTALENO AND UREA.
JP4935073B2 (en) 2003-10-14 2012-05-23 味の素株式会社 Ether derivatives
GB0324159D0 (en) 2003-10-15 2003-11-19 Glaxo Group Ltd Novel compounds
GB0325287D0 (en) * 2003-10-29 2003-12-03 Merck Sharp & Dohme Therapeutic agents
WO2005047280A1 (en) * 2003-11-10 2005-05-26 Merck Sharp & Dohme Limited Substituted nitrogen-containing six-membered amino-heterocycles as vanilloid-1 receptor antagonists for treating pain
US7435831B2 (en) 2004-03-03 2008-10-14 Chemocentryx, Inc. Bicyclic and bridged nitrogen heterocycles
US7435830B2 (en) 2004-03-03 2008-10-14 Chemocentryx, Inc. Bicyclic and bridged nitrogen heterocycles
GB0412769D0 (en) 2004-06-08 2004-07-07 Novartis Ag Organic compounds
EP1771162B1 (en) * 2004-07-19 2013-06-05 Xenia Pharma Capsaicin inhibitors for the treatment of obesity-related disorders
MY145822A (en) 2004-08-13 2012-04-30 Neurogen Corp Substituted biaryl piperazinyl-pyridine analogues
ES2359840T5 (en) * 2004-09-20 2015-03-25 Xenon Pharmaceuticals Inc. Pyridine derivatives for inhibition of human stearoyl-CoA desaturase
AR051202A1 (en) 2004-09-20 2006-12-27 Xenon Pharmaceuticals Inc HETEROCICLIC DERIVATIVES AND THEIR USE AS INHIBITORS OF ESTEAROIL-COA DESATURASA
US7592343B2 (en) 2004-09-20 2009-09-22 Xenon Pharmaceuticals Inc. Pyridazine-piperazine compounds and their use as stearoyl-CoA desaturase inhibitors
WO2006038041A1 (en) * 2004-10-08 2006-04-13 Merck Sharp & Dohme Limited Besylate salts of six-membered amino-heterocycles as vanilloid-1 receptor antagonists for treating pain
JP2008521815A (en) * 2004-11-24 2008-06-26 アルゴルクス ファーマスーティカルズ,インク Capsinoid gel preparation and use thereof
HUE033441T2 (en) 2004-12-30 2017-11-28 Janssen Pharmaceutica Nv 4-(benzyl)-piperazine-1-carboxylic acid phenylamide derivatives and related compounds as modulators of fatty acid amide hydrolase (faah) for the treatment of anxiety, pain and other conditions
EP1686176A1 (en) 2005-01-28 2006-08-02 Icon Genetics AG Production of antibodies in plants with plus-sense single-stranded viral RNA vectors
KR20060087386A (en) 2005-01-28 2006-08-02 주식회사 대웅제약 Novel benzoimidazole derivatives and a pharmaceutical composition comprising the same
US7576099B2 (en) 2005-02-28 2009-08-18 Renovis, Inc. Amide derivatives as ion-channel ligands and pharmaceutical compositions and methods of using the same
ES2552804T3 (en) 2005-05-04 2015-12-02 Evotec Ag Condensed heterocyclic compounds, and their compositions and uses
DE102005023943A1 (en) 2005-05-20 2006-11-23 Grünenthal GmbH Pentafluorosulfanyl-substituted compound and its use for the preparation of medicaments
EP2540296A1 (en) 2005-06-03 2013-01-02 Xenon Pharmaceuticals Inc. Arminothiazole derivatives as human stearoyl-coa desaturase inhibitors
EP1739179A1 (en) 2005-06-30 2007-01-03 Octapharma AG Serum-free stable transfection and production of recombinant human proteins in human cell lines
WO2007005510A1 (en) 2005-06-30 2007-01-11 Janssen Pharmaceutica N.V. N-heteroarylpiperazinyl ureas as modulators of fatty acid amide hydrolase
JP2008545010A (en) * 2005-06-30 2008-12-11 プロシディオン・リミテッド G protein-coupled receptor agonist
US20070049578A1 (en) * 2005-08-24 2007-03-01 Astrazeneca Ab Cinnamamide compounds as metabotropic glutamate receptor antagonists
US7842703B2 (en) 2005-10-07 2010-11-30 Glenmark Pharmaceuticals S.A. Substituted benzofused derivatives and their use as vanilloid receptor ligands
DE102005062991A1 (en) * 2005-12-28 2007-07-05 Grünenthal GmbH New N-thiazolylalkyl-substituted propiolamide derivatives, useful for treatment and prevention of e.g. pain, anxiety and panic attacks, are inhibitors of the mGluR5 receptor
CA2646023A1 (en) * 2006-03-10 2007-09-20 Neurogen Corporation Piperazinyl oxoalkyl tetrahydroisoquinolines and related analogues
EP2046787B1 (en) 2006-08-01 2011-04-06 Glaxo Group Limited Pyrazolo[3,4-b]pyridine compounds, and their use as pde4 inhibitors
JP2010501592A (en) 2006-08-25 2010-01-21 アボット・ラボラトリーズ Indazole derivatives inhibiting TRPV1 and uses thereof
US20080153845A1 (en) * 2006-10-27 2008-06-26 Redpoint Bio Corporation Trpv1 antagonists and uses thereof
MX2009006730A (en) 2006-12-20 2009-06-30 Abbott Lab Antagonists of the trpv1 receptor and uses thereof.
US8653091B2 (en) 2007-04-02 2014-02-18 Evotec Ag Pyrid-2yl fused heterocyclic compounds, and compositions and uses thereof
ES2427999T3 (en) 2007-04-17 2013-11-05 Evotec Ag 2-Cyanophenyl-condensed heterocyclic compounds and compositions and uses thereof
US20090176796A1 (en) * 2007-04-27 2009-07-09 Purdue Pharma L. P. Trpv1 antagonists including amide substituent and uses thereof
AU2011226773C1 (en) * 2007-04-27 2012-07-26 Purdue Pharma L.P. TRPV1 antagonists and uses thereof
JP5372913B2 (en) 2007-04-27 2013-12-18 パーデュー、ファーマ、リミテッド、パートナーシップ Effective treatment for pain
CN101835750B (en) 2007-08-22 2013-07-17 阿斯利康(瑞典)有限公司 Cyclopropyl amide derivatives '978
PE20091199A1 (en) 2007-09-06 2009-09-12 Glaxo Group Ltd PIPERAZINE DERIVATIVE HAVING AFFINITY FOR THE HISTAMINE H3 RECEPTOR
WO2009081222A1 (en) 2007-12-21 2009-07-02 Glenmark Pharmaceuticals, S.A. Substituted tricyclic pyridine or pyrimidine vanilloid receptor ligands
WO2009117626A2 (en) 2008-03-20 2009-09-24 Abbott Laboratories Methods for making central nervous system agents that are trpv1 antagonists
JP5372138B2 (en) 2008-04-18 2013-12-18 テウン ファーマシューティカル カンパニー,リミテッド Novel benzoxazine benzimidazole derivative, pharmaceutical composition containing the same, and use thereof
US8546388B2 (en) * 2008-10-24 2013-10-01 Purdue Pharma L.P. Heterocyclic TRPV1 receptor ligands
BRPI0921496A2 (en) 2008-11-04 2016-01-19 Chemocentryx Inc compound, pharmaceutical composition, and methods for treating disease or disorder, for inhibiting binding of chemokines to a receptor, for imaging an said tumor, organ or tissue, and for detecting elevated levels of cxcr7 in a sample.
US8853202B2 (en) 2008-11-04 2014-10-07 Chemocentryx, Inc. Modulators of CXCR7
GB0902173D0 (en) 2009-02-10 2009-03-25 Takeda Pharmaceutical Compounds and their use
TW201039825A (en) 2009-02-20 2010-11-16 Astrazeneca Ab Cyclopropyl amide derivatives 983
US20120121515A1 (en) 2009-03-13 2012-05-17 Lenny Dang Methods and compositions for cell-proliferation-related disorders
JP5745505B2 (en) 2009-05-07 2015-07-08 グリュネンタール・ゲゼルシャフト・ミト・ベシュレンクテル・ハフツング Substituted phenylureas and substituted phenylamides as vanilloid receptor ligands
US8946204B2 (en) 2009-05-07 2015-02-03 Gruenenthal Gmbh Substituted phenylureas and phenylamides as vanilloid receptor ligands
CA2766882C (en) 2009-06-29 2016-11-22 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions
CA2793836C (en) 2009-10-21 2020-03-24 Agios Pharmaceuticals, Inc. Methods and compositions for cell-proliferation-related disorders
EP3561077B1 (en) 2009-10-21 2022-12-21 Les Laboratoires Servier Methods for cell-proliferation-related disorders
JP2013520413A (en) 2010-02-18 2013-06-06 アストラゼネカ・アクチエボラーグ Process for producing cyclopropylamide derivatives and related intermediates
UA108233C2 (en) 2010-05-03 2015-04-10 Fatty acid amide hydrolysis activity modulators
US8921373B2 (en) 2010-06-22 2014-12-30 Shionogi & Co., Ltd. Compounds having TRPV1 antagonistic activity and uses thereof
AR082498A1 (en) * 2010-08-20 2012-12-12 Gruenenthal Gmbh CYCLIC DERIVATIVES OF CARBOXAMIDE AND UREA SUBSTITUTED AS LEGANDS OF THE VAINYLOID RECEIVER
US8609672B2 (en) 2010-08-27 2013-12-17 University Of The Pacific Piperazinylpyrimidine analogues as protein kinase inhibitors
WO2012045729A1 (en) 2010-10-05 2012-04-12 Glaxo Group Limited Imidazo [1, 2 -a] pyridine and pyrazolo [1, 5 -a] pyridine derivatives as trpv1 antagonists
KR101293384B1 (en) 2010-10-13 2013-08-05 주식회사 대웅제약 Novel pyridyl benzoxazine derivatives, pharmaceutical composition comprising the same, and use thereof
EP2642998B1 (en) 2010-11-24 2020-09-16 The Trustees of Columbia University in the City of New York A non-retinoid rbp4 antagonist for treatment of age-related macular degeneration and stargardt disease
WO2012072512A1 (en) 2010-11-29 2012-06-07 Glaxo Group Limited N-cyclobutyl-imidazopyridine or -pyrazolopyridine carboxamides as trpv1 antagonists
EP2697221A1 (en) 2011-04-11 2014-02-19 Glaxo Group Limited N- cyclobutyl - imidazopyridine - methylamine as trpv1 antagonists
CN103764147B (en) 2011-05-03 2018-05-22 安吉奥斯医药品有限公司 For the pyruvate kinase activator for the treatment of
JP6130827B2 (en) * 2011-05-17 2017-05-17 塩野義製薬株式会社 Heterocyclic compounds
CN102827073A (en) 2011-06-17 2012-12-19 安吉奥斯医药品有限公司 Therapeutically active compositions and application methods thereof
CN103814020B (en) * 2011-06-17 2017-07-14 安吉奥斯医药品有限公司 Therapeutic activity composition and their application method
CN102827170A (en) 2011-06-17 2012-12-19 安吉奥斯医药品有限公司 Active treatment compositions and use method thereof
SG195136A1 (en) 2011-06-22 2013-12-30 Purdue Pharma Lp Trpv1 antagonists including dihydroxy substituent and uses thereof
AU2012293417A1 (en) 2011-08-10 2013-05-02 Purdue Pharma L.P. TRPV1 antagonists including dihydroxy substituent and uses thereof
EP3406608B8 (en) 2012-01-06 2022-01-19 Les Laboratoires Servier Therapeutically active compounds and their methods of use
US9474779B2 (en) 2012-01-19 2016-10-25 Agios Pharmaceuticals, Inc. Therapeutically active compositions and their methods of use
US9333202B2 (en) 2012-05-01 2016-05-10 The Trustees Of Columbia University In The City Of New York Non-retinoid antagonists for treatment of age-related macular degeneration and stargardt disease
EA201590053A1 (en) 2012-06-20 2015-08-31 Новартис Аг MODULATORS OF THE COMPLEMENT SYSTEM AND THEIR APPLICATION
CA2888360A1 (en) 2012-10-15 2014-04-24 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions
CA2892042C (en) 2012-11-29 2022-06-14 Chemocentryx, Inc. Cxcr7 antagonists
US9579324B2 (en) 2013-07-11 2017-02-28 Agios Pharmaceuticals, Inc Therapeutically active compounds and their methods of use
WO2015003360A2 (en) 2013-07-11 2015-01-15 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
WO2015003355A2 (en) 2013-07-11 2015-01-15 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
EP3019480B1 (en) 2013-07-11 2020-05-06 Agios Pharmaceuticals, Inc. 2,4- or 4,6-diaminopyrimidine compounds as idh2 mutants inhibitors for the treatment of cancer
US20150031627A1 (en) 2013-07-25 2015-01-29 Agios Pharmaceuticals, Inc Therapeutically active compounds and their methods of use
JP6564380B2 (en) 2013-09-20 2019-08-21 ユニバーシティ オブ ピッツバーグ − オブ ザ コモンウェルス システム オブ ハイヤー エデュケイション Compounds for treating prostate cancer
US9968595B2 (en) 2014-03-14 2018-05-15 Agios Pharmaceuticals, Inc. Pharmaceutical compositions of therapeutically active compounds
CA2989111C (en) 2015-06-11 2023-10-03 Agios Pharmaceuticals, Inc. Methods of using pyruvate kinase activators
CN105295896B (en) * 2015-09-11 2017-12-29 浙江大学 The capsaicine fluorescence probe and its synthetic method of a kind of specific marker and application
JP6871920B2 (en) 2015-10-15 2021-05-19 アジオス ファーマシューティカルズ, インコーポレイテッド Combination therapy to treat malignant tumors
UA123400C2 (en) 2015-10-15 2021-03-31 Аджиос Фармасьютікалз, Інк. Combination therapy for treating malignancies
CA2937365C (en) 2016-03-29 2018-09-18 F. Hoffmann-La Roche Ag Granulate formulation of 5-methyl-1-phenyl-2-(1h)-pyridone and method of making the same
BR112020007818A2 (en) 2017-10-27 2020-10-20 Boehringer Ingelheim International Gmbh carbonyl pyridine derivatives and their therapeutic uses as trpc6 inhibitors
US10980788B2 (en) 2018-06-08 2021-04-20 Agios Pharmaceuticals, Inc. Therapy for treating malignancies
MA54465A (en) 2018-12-12 2021-10-20 Chemocentryx Inc CXCR7 INHIBITORS FOR THE TREATMENT OF CANCER
DE102022104759A1 (en) 2022-02-28 2023-08-31 SCi Kontor GmbH Co-crystal screening method, in particular for the production of co-crystals

Citations (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4424205A (en) * 1982-03-18 1984-01-03 The Procter & Gamble Company Hydroxyphenylacetamides having analgesic and anti-irritant activity
US4439606A (en) * 1982-05-06 1984-03-27 American Cyanamid Company Antiatherosclerotic 1-piperazinecarbonyl compounds
US4450272A (en) * 1982-05-06 1984-05-22 American Cyanamid Company Antiatherosclerotic 1-piperazine-thicarboxamides
US4659710A (en) * 1985-04-17 1987-04-21 Ss Pharmaceutical Co., Ltd. 1,7-Naphthyridine derivatives and pharmaceutical compositions
US4681897A (en) * 1984-01-16 1987-07-21 The Procter & Gamble Company Pharmaceutical products providing enhanced analgesia
US5021438A (en) * 1988-03-18 1991-06-04 Bayer Aktiengesellschaft 1,3,4,5-tetrahydrobenz(c,d)indoles
US5021450A (en) * 1989-05-30 1991-06-04 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services New class of compounds having a variable spectrum of activities for capsaicin-like responses, compositions and uses thereof
US5290816A (en) * 1988-10-24 1994-03-01 The United States Of America As Represented By The Secretary Of Health And Human Services Composition of resiniferatoxin and analogues thereof to cause sensory afferent C-Fiber and thermoregulatory desensitization
US5354747A (en) * 1993-06-16 1994-10-11 G. D. Searle & Co. 2-, 3-, 4-, 5-, 6-, 7-, 8-, 9- and/or 10-substituted dibenzoxazepine and dibenzthiazepine compounds, pharmaceutical compositions and methods of use
US5430033A (en) * 1993-04-27 1995-07-04 John Wyeth & Brother Ltd. Piperazine derivatives
US5442064A (en) * 1992-10-12 1995-08-15 Dr. Karl Thomae Gmbh Carboxylic acid derivatives, pharmaceutical compositions containing these compounds and processes for preparing them
US5461047A (en) * 1993-06-16 1995-10-24 G. D. Searle & Co. 2-,3-,4-,5-,6-,7-,8-,9- and/or 10-substituted dibenzoxazepine and dibenzthiazepine compounds, pharmaceutical compositions and methods of use
US5756504A (en) * 1994-03-24 1998-05-26 Merck & Co., Inc. Tocolytic oxytocin receptor antagonists
US5789412A (en) * 1994-07-20 1998-08-04 Pierre Fabre Medicament Piperazides derived from arylpiperazine, processes for their preparation, their use as medicaments and pharmaceutical compositions comprising them
US5792768A (en) * 1993-03-30 1998-08-11 Merck, Sharp & Dohme Limited Antipsychotic benzimidazole derivatives
US5840720A (en) * 1995-10-23 1998-11-24 Tong-Ho Lin 4-O and 5-aminomethylation of synthetic capsaicin derivatives, a new discovery of capsaicin antagonist
US5962532A (en) * 1997-03-13 1999-10-05 Campbell; James N. Therapeutic method with capsaicin and capsaicin analogues
US6028195A (en) * 1996-06-29 2000-02-22 Samjin Pharmaceutical Co., Ltd. Piperazine derivatives and process for the preparation thereof
US6169088B1 (en) * 1996-01-10 2001-01-02 Kyowa Hakko Kogyo Co., Ltd. 1,3 diazines with platelet-derived growth factor receptor inhibitory activity
US6248788B1 (en) * 1996-11-06 2001-06-19 The Regents Of The University Of California Therapeutic method with capsaicin and capasicin analogs
US6329395B1 (en) * 1998-06-08 2001-12-11 Schering Corporation Neuropeptide Y5 receptor antagonists
US6437147B1 (en) * 2000-03-17 2002-08-20 Novo Nordisk Imidazole compounds
US6476076B1 (en) * 1999-02-22 2002-11-05 Pacific Corporation Vanilloid analogues containing resinferatoxin pharmacophores as potent vanilloid receptor agonists and analgesics, compositions and uses thereof
US6723730B2 (en) * 2000-07-20 2004-04-20 Neurogen Corporation Capsaicin receptor ligands
US20040106616A1 (en) * 2002-01-17 2004-06-03 Rajagopal Bakthavatchalam Substituted quinazolin-4-ylamine analogues

Family Cites Families (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4656174A (en) 1982-07-24 1987-04-07 Pfizer Inc. Quinoline therapeutic agents
CA2020437A1 (en) * 1989-07-05 1991-01-06 Yoshihide Fuse Cinnamamide derivative
JPH03275657A (en) * 1989-07-05 1991-12-06 Kanegafuchi Chem Ind Co Ltd Cinnamic acid amide derivative
US5639754A (en) 1994-07-12 1997-06-17 Janssen Pharmaceutica N.V. Urea and thiourea derivatives of azolones
CN1128139C (en) 1995-01-11 2003-11-19 三进制药株式会社 New piperazine derivatives and its prepn.
JPH10195037A (en) * 1996-02-15 1998-07-28 Tanabe Seiyaku Co Ltd Phenol derivative
CA2197364A1 (en) * 1996-02-15 1997-08-16 Toshikazu Suzuki Phenol compound and process for preparing the same
WO1998020867A1 (en) * 1996-11-15 1998-05-22 The United States Of America, Represented By The Secretary, Department Of Health And Human Services Pharmaceutical compositions containing vanilloid agonists in combination with vanilloid antagonists
JPH10245357A (en) * 1997-03-03 1998-09-14 Yakult Honsha Co Ltd Phenylpropenone and medicine containing the same
EP1019367A1 (en) 1997-08-05 2000-07-19 Novo Nordisk A/S Derivatives of 2,5- and 3,5-disubstituted anilines, their preparation and use
US6423716B1 (en) 1998-03-31 2002-07-23 Kyowa Hakko Kogyo Co., Ltd. Nitrogenous heterocyclic compounds
HUP0103796A3 (en) * 1998-06-08 2002-04-29 Schering Corp Neuropeptide y5 receptor antagonists and medicaments containing them
EP1122242B1 (en) 1998-09-22 2008-01-16 Astellas Pharma Inc. Cyanophenyl derivatives
SK5282001A3 (en) 1998-11-06 2002-01-07 Knoll Gmbh Tricyclic pyrazole derivatives
AU763030B2 (en) 1999-03-03 2003-07-10 Samjin Pharmaceutical Co., Ltd. Piperazine derivatives and process for the preparation thereof
US6713473B1 (en) 1999-04-20 2004-03-30 Meiji Seika Kaisha, Ltd. Tricyclic compounds
EP1313477A4 (en) 2000-07-15 2004-03-03 Smithkline Beecham Corp Compounds and methods

Patent Citations (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4424205A (en) * 1982-03-18 1984-01-03 The Procter & Gamble Company Hydroxyphenylacetamides having analgesic and anti-irritant activity
US4439606A (en) * 1982-05-06 1984-03-27 American Cyanamid Company Antiatherosclerotic 1-piperazinecarbonyl compounds
US4450272A (en) * 1982-05-06 1984-05-22 American Cyanamid Company Antiatherosclerotic 1-piperazine-thicarboxamides
US4681897A (en) * 1984-01-16 1987-07-21 The Procter & Gamble Company Pharmaceutical products providing enhanced analgesia
US4812446A (en) * 1984-01-16 1989-03-14 The Procter & Gamble Company Pharmaceutical products providing enhanced analgesia
US4659710A (en) * 1985-04-17 1987-04-21 Ss Pharmaceutical Co., Ltd. 1,7-Naphthyridine derivatives and pharmaceutical compositions
US5021438A (en) * 1988-03-18 1991-06-04 Bayer Aktiengesellschaft 1,3,4,5-tetrahydrobenz(c,d)indoles
US5290816A (en) * 1988-10-24 1994-03-01 The United States Of America As Represented By The Secretary Of Health And Human Services Composition of resiniferatoxin and analogues thereof to cause sensory afferent C-Fiber and thermoregulatory desensitization
US5021450A (en) * 1989-05-30 1991-06-04 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services New class of compounds having a variable spectrum of activities for capsaicin-like responses, compositions and uses thereof
US5442064A (en) * 1992-10-12 1995-08-15 Dr. Karl Thomae Gmbh Carboxylic acid derivatives, pharmaceutical compositions containing these compounds and processes for preparing them
US5792768A (en) * 1993-03-30 1998-08-11 Merck, Sharp & Dohme Limited Antipsychotic benzimidazole derivatives
US5430033A (en) * 1993-04-27 1995-07-04 John Wyeth & Brother Ltd. Piperazine derivatives
US5461047A (en) * 1993-06-16 1995-10-24 G. D. Searle & Co. 2-,3-,4-,5-,6-,7-,8-,9- and/or 10-substituted dibenzoxazepine and dibenzthiazepine compounds, pharmaceutical compositions and methods of use
US5354747A (en) * 1993-06-16 1994-10-11 G. D. Searle & Co. 2-, 3-, 4-, 5-, 6-, 7-, 8-, 9- and/or 10-substituted dibenzoxazepine and dibenzthiazepine compounds, pharmaceutical compositions and methods of use
US5756504A (en) * 1994-03-24 1998-05-26 Merck & Co., Inc. Tocolytic oxytocin receptor antagonists
US5789412A (en) * 1994-07-20 1998-08-04 Pierre Fabre Medicament Piperazides derived from arylpiperazine, processes for their preparation, their use as medicaments and pharmaceutical compositions comprising them
US5840720A (en) * 1995-10-23 1998-11-24 Tong-Ho Lin 4-O and 5-aminomethylation of synthetic capsaicin derivatives, a new discovery of capsaicin antagonist
US6169088B1 (en) * 1996-01-10 2001-01-02 Kyowa Hakko Kogyo Co., Ltd. 1,3 diazines with platelet-derived growth factor receptor inhibitory activity
US6028195A (en) * 1996-06-29 2000-02-22 Samjin Pharmaceutical Co., Ltd. Piperazine derivatives and process for the preparation thereof
US6248788B1 (en) * 1996-11-06 2001-06-19 The Regents Of The University Of California Therapeutic method with capsaicin and capasicin analogs
US5962532A (en) * 1997-03-13 1999-10-05 Campbell; James N. Therapeutic method with capsaicin and capsaicin analogues
US6329395B1 (en) * 1998-06-08 2001-12-11 Schering Corporation Neuropeptide Y5 receptor antagonists
US6476076B1 (en) * 1999-02-22 2002-11-05 Pacific Corporation Vanilloid analogues containing resinferatoxin pharmacophores as potent vanilloid receptor agonists and analgesics, compositions and uses thereof
US6437147B1 (en) * 2000-03-17 2002-08-20 Novo Nordisk Imidazole compounds
US6723730B2 (en) * 2000-07-20 2004-04-20 Neurogen Corporation Capsaicin receptor ligands
US20040106616A1 (en) * 2002-01-17 2004-06-03 Rajagopal Bakthavatchalam Substituted quinazolin-4-ylamine analogues

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060035939A1 (en) * 2004-07-14 2006-02-16 Japan Tobacco Inc. 3-Aminobenzamide compounds and inhibitors of vanilloid receptor subtype 1 (VR1) activity
US20100022523A1 (en) * 2004-07-14 2010-01-28 Japan Tobacco Inc. 3-aminobenzamide compounds and vanilloid receptor subtype 1 (vr1) inhibitors
US20060035882A1 (en) * 2004-07-15 2006-02-16 Japan Tobacco Inc. Condensed benzamide compounds and inhibitors of vanilloid receptor subtype 1 (vr1) activity
US8008292B2 (en) 2004-07-15 2011-08-30 Japan Tobacco Inc. Condensed benzamide compounds and inhibitors of vanilloid receptor subtype 1 (VR1) activity
US7906508B2 (en) 2005-12-28 2011-03-15 Japan Tobacco Inc. 3,4-dihydrobenzoxazine compounds and inhibitors of vanilloid receptor subtype 1 (VRI) activity
US20070149517A1 (en) * 2005-12-28 2007-06-28 Japan Tobacco Inc. 3,4-dihydrobenzoxazine compounds and inhibitors of vanilloid receptor subtype 1 (vri) activity
US20110212952A1 (en) * 2005-12-28 2011-09-01 Japan Tobacco Inc. 3,4-dihydrobenzoxazine compounds and inhibitors of vanilloid receptor subtype 1 (vri) activity
US20090163508A1 (en) * 2007-10-10 2009-06-25 Takeda Pharmaceutical Company Limited Amide compound
US20100130499A1 (en) * 2008-10-24 2010-05-27 Laykea Tafesse Bicycloheteroaryl compounds and their use as trpv1 ligands
US20100120862A1 (en) * 2008-10-24 2010-05-13 Laykea Tafesse Monocyclic compounds and their use as trpv1 ligands
US8703962B2 (en) 2008-10-24 2014-04-22 Purdue Pharma L.P. Monocyclic compounds and their use as TRPV1 ligands
US8759362B2 (en) 2008-10-24 2014-06-24 Purdue Pharma L.P. Bicycloheteroaryl compounds and their use as TRPV1 ligands
WO2016100161A1 (en) * 2014-12-19 2016-06-23 Merck Sharp & Dohme Corp. Ethyldiamine orexin receptor antagonists
US10011595B2 (en) 2014-12-19 2018-07-03 Merck Sharp & Dohme Corp. Ethyldiamine orexin receptor antagonists

Also Published As

Publication number Publication date
KR20030024799A (en) 2003-03-26
WO2002008221A2 (en) 2002-01-31
AU2001280667B2 (en) 2007-08-02
IL153884A0 (en) 2003-07-31
US20090082362A1 (en) 2009-03-26
BR0112631A (en) 2003-09-23
WO2002008221A3 (en) 2002-07-11
EP1301484A2 (en) 2003-04-16
US6723730B2 (en) 2004-04-20
MXPA03000458A (en) 2004-06-02
AU8066701A (en) 2002-02-05
CN1443170A (en) 2003-09-17
US20020132853A1 (en) 2002-09-19
NZ523526A (en) 2004-10-29
CA2415606A1 (en) 2002-01-31
JP2004525071A (en) 2004-08-19

Similar Documents

Publication Publication Date Title
US6723730B2 (en) Capsaicin receptor ligands
AU2001280667A1 (en) Capsaicin receptor ligands
EP1751113B1 (en) Urea antagonists of p2y1 receptor useful in the treatment of thrombotic conditions
KR100554988B1 (en) 2-Amino-Nicotinamide Derivatives and Their Use as VEGF-Receptor Tyrosine Kinase Inhibitors
EP1565452B1 (en) 3-substituted-6-aryl pyridines as ligands of c5a receptors
KR101458006B1 (en) Arylamide derivatives as ttx-s blockers
US9133122B2 (en) Amide compounds, compositions and uses thereof
US20150353538A1 (en) Compounds and therapeutic uses thereof
KR102067235B1 (en) Amide derivatives as ttx-s blockers
KR20150118964A (en) Amides as modulators of sodium channels
KR20150112031A (en) Pyridone amides as modulators of sodium channels
KR20150114536A (en) Quinoline and quinazoline amides as modulators of sodium channels
WO2002080928A1 (en) N-substituted nonaryl-heterocyclo amidyl nmda/nr2b antagonists
KR20140071518A (en) Sulfamoyl benzoic acid derivatives as trpm8 antagonists
US20130317027A1 (en) Compounds and therapeutic uses thereof
EA025322B1 (en) Dihydro-benzo-oxazine and dihydro-pyrido-oxazine derivatives
BG108490A (en) 5-substituted-2-arylpyridines as crf1 modulators
JP5220407B2 (en) 1-Aryl-4-substituted isoquinolines
KR20130122531A (en) Substituted 6-methylnicotinamides as mglur5 positive allosteric modulators
KR19980702030A (en) 5- (4-substituted piperidinyl-1) -3-aryl-pentanoic acid derivatives that are antagonists of the tachykinin receptor
KR20210110288A (en) Substituted aryl compounds, and methods for their preparation and uses thereof
CZ2004156A3 (en) The title is not available
US7115664B2 (en) Peptidomimetic ligands for cellular receptors and ion channels
EA038129B1 (en) Pyrazolylaminobenzimidazole derivatives as jak inhibitors
TW202204343A (en) Substituted 3-phenoxyazetidin-1-yl-pyrazines

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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