US20100292236A1 - 5-Membered Heterocyclic Amides And Related Compounds - Google Patents

5-Membered Heterocyclic Amides And Related Compounds Download PDF

Info

Publication number
US20100292236A1
US20100292236A1 US12/669,308 US66930808A US2010292236A1 US 20100292236 A1 US20100292236 A1 US 20100292236A1 US 66930808 A US66930808 A US 66930808A US 2010292236 A1 US2010292236 A1 US 2010292236A1
Authority
US
United States
Prior art keywords
alkyl
mono
amino
cycloalkyl
pain
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/669,308
Other languages
English (en)
Inventor
Hongbin Li
Jun Yuan
Rajagopal Bakthavatchalam
Kevin J. Hodgetts
Scott M. Capitosti
Jianmin Mao
David J. Wustrow
Qin Guo
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.)
Mindimmune Therapeutics Inc
Original Assignee
H Lundbeck AS
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 H Lundbeck AS filed Critical H Lundbeck AS
Priority to US12/669,308 priority Critical patent/US20100292236A1/en
Assigned to NEUROGEN CORPORATION reassignment NEUROGEN CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MAO, JIANMIN, BAKTHAVATCHALAM, RAJAGOPAL, CAPITOSTI, SCOTT M., GUO, QIN, HODGETTS, KEVIN J., LI, HONGBIN, WUSTROW, DAVID J., YUAN, JUN
Assigned to H. LUNDBECK A/S reassignment H. LUNDBECK A/S ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NEUROGEN CORPORATION
Publication of US20100292236A1 publication Critical patent/US20100292236A1/en
Assigned to MINDIMMUNE THERAPEUTICS, INC. reassignment MINDIMMUNE THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: H. LUNDBECK A/S
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • 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
    • A61P11/06Antiasthmatics
    • 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
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/06Antigout agents, e.g. antihyperuricemic or uricosuric agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • 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
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/14Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/20Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D277/32Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or 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
    • C07D277/56Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/02Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings
    • C07D333/04Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom
    • C07D333/26Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D333/38Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/04Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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
    • C07D417/14Heterocyclic 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 three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • This invention relates generally to 5-membered heterocyclic amides and related compounds that have useful pharmacological properties.
  • the invention further relates to the use of such compounds for treating conditions related to P2X 7 receptor activation, for identifying other agents that bind to P2X 7 receptor, and as probes for the detection and localization of P2X 7 receptors.
  • nociceptors A wide variety of physical and chemical stimuli induce activation of such neurons in mammals, leading to recognition of a potentially harmful stimulus. Inappropriate or excessive activation of nociceptors, however, can result in debilitating acute or chronic pain.
  • Neuropathic pain which typically results from damage to the nervous system, involves pain signal transmission in the absence of stimulus, pain from a normally innocuous stimulus (allodynia) and increased pain from a normally painful stimulus (hyperalgesia). In most instances, neuropathic pain is thought to occur because of sensitization in the peripheral and central nervous systems following initial damage to the peripheral system (e.g., via direct injury or systemic disease). Neuropathic pain is typically burning, shooting and unrelenting in its intensity and can sometimes be more debilitating than the initial injury or disease process that induced it.
  • neuropathic pain is generally suboptimal.
  • Opiates such as morphine
  • morphine are potent analgesics, but their usefulness is limited because of adverse side effects, such as physical addictiveness and withdrawal properties, as well as respiratory depression, mood changes, and decreased intestinal motility with concomitant constipation, nausea, vomiting, and alterations in the endocrine and autonomic nervous systems.
  • neuropathic pain is frequently non-responsive or only partially responsive to conventional opioid analgesic regimens, or to treatment with other drugs, such as gabapentin.
  • Treatments employing the N-methyl-D-aspartate antagonist ketamine or the alpha(2)-adrenergic agonist clonidine can reduce acute or chronic pain, and permit a reduction in opioid consumption, but these agents are often poorly tolerated due to side effects.
  • Transient inflammation is a beneficial mechanism that protects mammals from invading pathogens. Uncontrolled inflammation, however, causes tissue damage and pain and is the underlying cause of many illnesses, including asthma, as well as other allergic, infectious, autoimmune, degenerative, and idiopathic diseases. Existing treatments often exhibit low, delayed or only temporary efficacy, undesirable side-effects and/or a lack of selectivity.
  • new drugs that overcome one or more of the shortcomings of drugs currently used for immunosuppression or in the treatment or prevention of inflammatory disorders, including allergic disorders, autoimmune disorders, fibrogenic disorders, and neurodegenerative diseases, such as amyotrophic lateral sclerosis, Alzheimer's disease, and Huntington's disease.
  • the P2X 7 receptor is a ligand-gated ion channel that is activated by ATP and is present on a variety of cell types, including microglia in the central nervous system and cells involved in inflammation and immune system function, such as immune cells.
  • P2X 7 is involved in activation of lymphocytes and monocyte/macrophages leading to the increased release of pro-inflammatory cytokines (e.g., TNFalpha and IL-1beta) from these cells.
  • pro-inflammatory cytokines e.g., TNFalpha and IL-1beta
  • P2X 7 receptor antagonists may find use in the treatment and prophylaxis of pain, including acute, chronic and neuropathic pain, as well as a variety of other conditions including osteoarthritis, rheumatoid arthritis, arthrosclerosis, inflammatory bowel disease, Alzheimer's disease, traumatic brain injury, asthma, chronic obstructive pulmonary disease, and fibrosis of internal organs (e.g., interstitial fibrosis).
  • the present invention provides 5-membered heterocyclic amides and related compounds of Formula A:
  • the present invention provides 5-membered heterocyclic amides and related compounds of Formula I:
  • the present invention provides 5-membered heterocyclic amides and related compounds of Formula A that further satisfy Formula III:
  • the present invention provides 5-membered heterocyclic amides and related compounds of Formula A that further satisfy Formula IV or Formula V:
  • the present invention provides 5-membered heterocyclic amides and related compounds of Formula A that further satisfy Formula VI:
  • the present invention provides 5-membered heterocyclic amides and related compounds of Formula A that further satisfy Formula VII:
  • the present invention provides 5-membered heterocyclic amides and related compounds of Formula A that further satisfy Formula VIII:
  • the present invention provides 5-membered heterocyclic amides and related compounds of Formula A that further satisfy Formula IX:
  • 5-membered heterocyclic amides and related compounds of Formula A or I-IX, as well as other Formula (s) provided herein are P2X 7 receptor antagonists and exhibit an IC 50 value of no greater than 20 micromolar, 10 micromolar, 5 micromolar, 1 micromolar, 500 nanomolar, or 100 nanomolar in an in vitro assay for determination of P2X 7 receptor antagonist activity.
  • such P2X 7 receptor antagonists exhibit no detectable agonist activity in an in vitro assay of P2X 7 receptor activity (i.e., within an assay provided in Example 4, herein) at a concentration equal to the IC 50 , 10 times the IC 50 or 100 times the IC 50 and/or at a concentration of 2,500 nM.
  • 5-membered heterocyclic amides and related compounds provided herein are labeled with a detectable marker (e.g., radiolabeled or fluorescein conjugated).
  • a detectable marker e.g., radiolabeled or fluorescein conjugated
  • the present invention further provides, within other aspects, pharmaceutical compositions comprising at least one 5-membered heterocyclic amides or related compound provided herein in combination with a physiologically acceptable carrier or excipient.
  • methods for modulating (e.g., reducing) cellular P2X 7 receptor activation or activity, comprising contacting a cell (e.g., microglia, astrocyte or peripheral macrophage or monocyte) that expresses a P2X 7 receptor with at least one P2X 7 receptor modulator as described herein.
  • a cell e.g., microglia, astrocyte or peripheral macrophage or monocyte
  • P2X 7 receptor modulator as described herein.
  • Such contact may occur in vivo or in vitro and is generally performed using a concentration of P2X 7 receptor modulator that is sufficient to detectably alter P2X 7 receptor activity in vitro (as determined using an assay provided in Example 4).
  • the present invention further provides methods for treating a condition responsive to P2X 7 receptor modulation in a patient, comprising administering to the patient a therapeutically effective amount of at least one P2X 7 receptor antagonist as described herein.
  • methods for treating pain in a patient, comprising administering to a patient suffering from (or at risk for) pain a therapeutically effective amount of at least one P2X 7 receptor antagonist as described herein.
  • methods for treating inflammation in a patient, comprising administering to a patient suffering from (or at risk for) inflammation a therapeutically effective amount of at least one P2X 7 receptor antagonist as described herein.
  • Methods are further provided for treating neurological and/or neurodegenerative disorders, cardiovascular disorders, osteoarthritis, rheumatoid arthritis, arthrosclerosis, irritable bowel syndrome, inflammatory bowel disease, Alzheimer's disease, traumatic brain injury, asthma, chronic obstructive pulmonary disease, ocular conditions (e.g., glaucoma), cirrhosis, lupus, scleroderma, or fibrosis of internal organs (e.g., interstitial fibrosis) in a patient, comprising administering to a patient suffering from (or at risk for) one or more of the foregoing conditions a therapeutically effective amount of at least one P2X 7 receptor antagonist as described herein.
  • the present invention provides methods for inhibiting death of retinal ganglion cells in a patient, comprising administering to the patient a therapeutically effective amount of at least one P2X 7 receptor antagonist as described herein.
  • a P2X 7 receptor antagonist such as, but not limited to, a P2X 7 receptor antagonist as described herein.
  • Methods are further provided for identifying an agent that binds to P2X 7 receptor, comprising: (a) contacting P2X 7 receptor with a labeled compound that is a 5-membered heterocyclic amides or related compound as described herein under conditions that permit binding of the compound to P2X 7 receptor, thereby generating bound, labeled compound; (b) detecting a signal that corresponds to the amount of bound, labeled compound in the absence of test agent; (c) contacting the bound, labeled compound with a test agent; (d) detecting a signal that corresponds to the amount of bound labeled compound in the presence of test agent; and (e) detecting a decrease in signal detected in step (d), as compared to the signal detected in step (b).
  • the present invention provides methods for determining the presence or absence of P2X 7 receptor in a sample, comprising: (a) contacting a sample with a compound as described herein under conditions that permit modulation by the compound of P2X 7 receptor activity; and (b) detecting a signal indicative of a level of the compound modulating P2X 7 receptor activity.
  • the present invention also provides packaged pharmaceutical preparations, comprising: (a) a pharmaceutical composition as described herein in a container; and (b) instructions for using the composition to treat one or more conditions responsive to P2X 7 receptor modulation, such as pain, inflammation, a neurological or neurodegenerative disorder, a cardiovascular disorder, an ocular disorder or an immune system disorder, osteoarthritis rheumatoid arthritis, arthrosclerosis, inflammatory bowel disease, Alzheimer's disease, traumatic brain injury, asthma, chronic obstructive pulmonary disease, and/or fibrosis of internal organs such as interstitial fibrosis.
  • P2X 7 receptor modulation such as pain, inflammation, a neurological or neurodegenerative disorder, a cardiovascular disorder, an ocular disorder or an immune system disorder, osteoarthritis rheumatoid arthritis, arthrosclerosis, inflammatory bowel disease, Alzheimer's disease, traumatic brain injury, asthma, chronic obstructive pulmonary disease, and/or fibrosis of internal organs such as inter
  • the present invention provides methods for preparing the compounds disclosed herein, including the intermediates.
  • the present invention provides 5-membered heterocyclic amides and related compounds. Such compounds may be used in vitro or in vivo, to modulate P2X 7 receptor activity in a variety of contexts.
  • 5-membered heterocyclic amides and related compounds encompasses all compounds of Formula A, I, II, III, IV, V, VI, VII, VIII and/or IX, as well as compounds of other Formulas provided herein (including any enantiomers, racemates and stereoisomers) and pharmaceutically acceptable salts, solvates, amides and esters of such compounds.
  • a “pharmaceutically acceptable salt” of a compound recited herein is an acid or base salt that is suitable for use in contact with the tissues of human beings or animals without excessive toxicity or carcinogenicity, and preferably without irritation, allergic response, or other problem or complication.
  • Such salts include mineral and organic acid salts of basic residues such as amines, as well as alkali or organic salts of acidic residues such as carboxylic acids.
  • Specific pharmaceutically acceptable anions for use in salt formation include, but are not limited to, acetate, 2-acetoxybenzoate, ascorbate, benzoate, bicarbonate, bromide, calcium edetate, carbonate, chloride, citrate, dihydrochloride, diphosphate, ditartrate, edetate, estolate (ethylsuccinate), formate, fumarate, gluceptate, gluconate, glutamate, glycolate, glycollylarsanilate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroiodide, hydroxymaleate, hydroxynaphthoate, iodide, isethionate, lactate, lactobionate, malate, maleate, mandelate, methylbromide, methylnitrate, methylsulfate, mucate, napsylate, nitrate, pamoate, pantothenate, phenylacetate
  • pharmaceutically acceptable cations for use in salt formation include, but are not limited to ammonium, benzathine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine, procaine, and metals such as aluminum, calcium, lithium, magnesium, potassium, sodium and zinc.
  • a pharmaceutically acceptable acid or base salt can be synthesized from a parent compound that contains a basic or acidic moiety by any conventional chemical method.
  • such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, the use of nonaqueous media, such as ether, ethyl acetate, ethanol, methanol, isopropanol or acetonitrile, is preferred.
  • nonaqueous media such as ether, ethyl acetate, ethanol, methanol, isopropanol or acetonitrile
  • prodrugs of the compounds of the recited Formulas are provided herein.
  • a “prodrug” is a compound that may not fully satisfy the structural requirements of the compounds provided herein, but is modified in vivo, following administration to a patient, to produce a compound a formula provided herein.
  • a prodrug may be an acylated derivative of a compound as provided herein.
  • Prodrugs include compounds wherein hydroxy, amine or sulfhydryl groups are bonded to any group that, when administered to a mammalian subject, cleaves to form a free hydroxy, amino or sulfhydryl group, respectively.
  • Examples of prodrugs include, but are not limited to, acetate, formate, benzoate and peptide derivatives of alcohol and amine functional groups within the compounds provided herein.
  • Prodrugs of the compounds provided herein may be prepared by modifying functional groups present in the compounds in such a way that the modifications are cleaved in vivo to yield the parent compounds.
  • alkyl refers to a straight or branched chain saturated aliphatic hydrocarbon.
  • Alkyl groups include groups having from 1 to 8 carbon atoms (C 1 -C 8 alkyl), from 1 to 6 carbon atoms (C 1 -C 6 alkyl) and from 1 to 4 carbon atoms (C 1 -C 4 alkyl), such as methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, tert-butyl, pentyl, 2-pentyl, isopentyl, neopentyl, hexyl, 2-hexyl, 3-hexyl and 3-methylpentyl.
  • C 0 -C u alkyl refers to a single covalent bond (C 0 ) or an alkyl group having from 1 to n carbon atoms; for example “C 0 -C 4 alkyl” refers to a single covalent bond or a C 1 -C 4 alkyl group. In some instances, a substituent of an alkyl group is specifically indicated. For example, “hydroxyalkyl” refers to an alkyl group substituted with at least one —OH; “aminoalkyl” refers to an alkyl group substituted with at least one —NH 2 .
  • Alkenyl refers to straight or branched chain alkene groups, which comprise at least one unsaturated carbon-carbon double bond. Alkenyl groups include C 2 -C 8 alkenyl, C 2 -C 6 alkenyl and C 2 -C 4 alkenyl groups, which have from 2 to 8, 2 to 6 or 2 to 4 carbon atoms, respectively, such as ethenyl, allyl or isopropenyl. “Alkynyl” refers to straight or branched chain alkyne groups, which have one or more unsaturated carbon-carbon bonds, at least one of which is a triple bond.
  • Alkynyl groups include C 2 -C 8 alkynyl, C 2 -C 6 alkynyl and C 2 -C 4 alkynyl groups, which have from 2 to 8, 2 to 6 or 2 to 4 carbon atoms, respectively.
  • Alkylene refers to a divalent alkyl group, as defined above.
  • C 1 -C 2 alkylene is methylene or ethylene;
  • C 0 -C 4 alkylene is a single covalent bond or an alkylene group having 1, 2, 3 or carbon atoms;
  • C 0 -C 2 alkylene is a single covalent bond, methylene or ethylene.
  • a “C 1 -C 6 alkylene that is optionally modified by the replacement of a carbon-carbon single bond with a double or triple carbon-carbon bond” is a C 1 -C 6 alkylene group as described above, or a divalent C 2 -C 6 alkene or C 2 -C 6 alkyne.
  • a “cycloalkyl” is a group that comprises one or more saturated and/or partially saturated rings in which all ring members are carbon, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, adamantyl, mertanyl, and partially saturated variants of the foregoing, such as cyclohexenyl. Cycloalkyl groups do not comprise an aromatic ring or a heterocyclic ring. Certain cycloalkyl groups are C 3 -C 7 cycloalkyl, in which the cycloalkyl group contains a single ring having from 3 to 7 ring members, all of which are carbon.
  • a “(C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl” is a C 3 -C 7 cycloalkyl group linked via a single covalent bond or a C 1 -C 4 alkylene group.
  • cycloalkyl groups are described herein as resulting from two substituents (e.g., R 5 moieties) being taken together, along with any intervening atoms.
  • “Intervening atoms” in this context are any atoms that appear in the chain of covalently bonded atoms linking the two substituents. If the two substituents are attached to the same carbon atom, then the carbon atom to which they are attached is the only intervening atom. If the two substituents are attached to different atoms (typically carbon or nitrogen), then the atoms to which they are attached are intervening atoms, as are additional atoms (if any) that are needed to complete the chain of covalent linkages between the two substituents.
  • a “(C 4 -C 7 cycloalkyl)C 0 -C 4 alkylene” is a divalent (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl group that is linked via two single covalent bonds to two specified moieties.
  • one such single covalent bond is located on the cyclic portion and the other is located on the alkylene portion, if present; alternatively, if no alkylene group is present, both such single covalent bonds are located on different ring members.
  • one a (C 6 cycloalkyl)C 2 alkylene moiety is:
  • alkoxy is meant an alkyl group as described above attached via an oxygen bridge.
  • Alkoxy groups include C 1 -C 6 alkoxy and C 1 -C 4 alkoxy groups, which have from 1 to 6 or from 1 to 4 carbon atoms, respectively.
  • Methoxy, ethoxy, propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, n-pentoxy, 2-pentoxy, 3-pentoxy, isopentoxy, neopentoxy, hexoxy, 2-hexoxy, 3-hexoxy, and 3-methylpentoxy are representative alkoxy groups.
  • alkylthio refers to an alkyl group as described above attached via a sulfur bridge
  • alkenylthio refers to an alkenyl group as described above attached via a sulfur bridge
  • oxo is used herein to refer to an oxygen substituent of a carbon atom that results in the foil nation of a carbonyl group (C ⁇ O).
  • An oxo group that is a substituent of a nonaromatic carbon atom results in a conversion of —CH 2 — to —C( ⁇ ))—.
  • An oxo group that is a substituent of an aromatic carbon atom results in a conversion of —CH— to —C( ⁇ O)— and may result in a loss of aromaticity.
  • alkanoyl refers to an acyl group (e.g., —(C ⁇ O)-alkyl), in which carbon atoms are in a linear or branched alkyl arrangement and where attachment is through the carbon of the keto group.
  • Alkanoyl groups have the indicated number of carbon atoms, with the carbon of the keto group being included in the numbered carbon atoms.
  • a C 2 alkanoyl group is an acetyl group having the formula —(C ⁇ O)CH 3 .
  • Alkanoyl groups include, for example, C 1 -C 8 alkanoyl, C 1 -C 6 alkanoyl and C 1 -C 4 alkanoyl groups, which have from 1 to 8, from 1 to 6 or from 1 to 4 carbon atoms, respectively.
  • C 1 alkanoyl refers to —(C ⁇ O)H.
  • Alkyl ether refers to a linear or branched ether substituent (i.e., an alkyl group that is substituted with an alkoxy group).
  • Alkyl ether groups include C 2 -C 8 alkyl ether, C 2 -C 6 alkyl ether and C 2 -C 4 alkyl ether groups, which have 2 to 8, 6 or 4 carbon atoms, respectively.
  • a C 2 alkyl ether has the structure —CH 2 —O—CH 3 .
  • alkoxycarbonyl refers to an alkoxy group attached through a keto (—(C ⁇ O)—) bridge (i.e., a group having the general structure —C( ⁇ O)—O-alkyl).
  • Alkoxycarbonyl groups include C 1 -C 8 , C 1 -C 6 and C 1 -C 4 alkoxycarbonyl groups, which have from 1 to 8, 6 or 4 carbon atoms, respectively, in the alkyl portion of the group (i.e., the carbon of the keto bridge is not included in the indicated number of carbon atoms).
  • C 1 alkoxycarbonyl refers to —C( ⁇ O)—O—CH 3 ;
  • C 3 alkoxycarbonyl indicates —C( ⁇ O)—O—(CH 2 ) 2 CH 3 or —C( ⁇ O)—O—(CH)(CH 3 ) 2 .
  • Alkanoyloxy refers to an alkanoyl group linked via an oxygen bridge (i.e., a group having the general structure —O—C( ⁇ O)-alkyl).
  • Alkanoyloxy groups include C 1 -C 8 , C 1 -C 6 and C 1 -C 4 alkanoyloxy groups, which have from 1 to 8, 6 or 4 carbon atoms, respectively, in the alkyl portion of the group.
  • C 1 alkanoyloxy refers to —O—C( ⁇ O)—CH 3 .
  • alkanoylamino refers to an alkanoyl group linked via a nitrogen bridge (i.e., a group having the general structure —N(R)—C( ⁇ O)-alkyl), in which R is hydrogen or C 1 -C 6 alkyl.
  • Alkanoylamino groups include C 1 -C 8 , C 1 -C 6 and C 1 -C 4 alkanoylamino groups, which have from 1 to 8, 6 or 4 carbon atoms within the alkanoyl group, respectively, in the alkyl portion of the group.
  • Alkylsulfonyl refers to groups of the formula —(SO 2 )-alkyl, in which the sulfur atom is the point of attachment. Alkylsulfonyl groups include C 1 -C 6 alkylsulfonyl and C 1 -C 4 alkylsulfonyl groups, which have from 1 to 6 or from 1 to 4 carbon atoms, respectively. Methylsulfonyl is one representative alkylsulfonyl group.
  • C 1 -C 6 alkylsulfonylC 0 -C 4 alkyl is a C 1 -C 6 alkylsulfonyl moiety that is linked via a single covalent bond or a C 1 -C 4 alkylne group.
  • C 1 -C 4 haloalkylsulfonyl is an alkylsulfonyl group that has from 1 to 4 carbon atoms and is substituted with at least one halogen (e.g., trifluoromethylsulfonyl).
  • C 1 -C 6 alkylsulfonyloxy refers to a C 1 -C 6 alkylsulfonyl moiety that is linked via an oxygen bridge.
  • Alkylsulfonylamino refers to groups of the formula —N(R)—(SO 2 )-alkyl, in which R is hydrogen or C 1 -C 6 alkyl and the nitrogen atom is the point of attachment.
  • Alkylsulfonylamino groups include C 1 -C 6 alkylsulfonylamino and C 1 -C 4 alkylsulfonylamino groups, which have from 1 to 6 or 1 to 4 carbon atoms, respectively.
  • Methylsulfonylamino is a representative alkylsulfonylamino group.
  • C 1 -C 6 alkylsulfonylaminoC 0 -C 4 alkyl is a C 1 -C 0 alkylsulfonylamino moiety linked via a single covalent bond or a C 1 -C 4 alkylene group.
  • C 1 -C 6 haloalkylsulfonylamino is an alkylsulfonylamino group that has from 1 to 6 carbon atoms and is substituted with at least one halogen (e.g., trifluoromethylsulfonylamino).
  • Aminosulfonyl refers to groups of the formula —(SO 2 )—NH 2 , in which the sulfur atom is the point of attachment.
  • the term “mono- or di-(C 1 -C 6 alkyl)aminosulfonyl” refers to groups that satisfy the formula —(SO 2 )—NR 2 , in which the sulfur atom is the point of attachment, and in which one R is C 1 -C 6 alkyl and the other R is hydrogen or an independently chosen C 1 -C 6 alkyl.
  • Alkylaminoalkyl refers to an alkylamino group linked via an alkylene group (i.e., a group having the general structure -alkylene-NH-alkyl or -alkylene-N(alkyl)(alkyl)) in which each alkyl is selected independently from alkyl, cycloalkyl and (cycloalkyl)alkyl groups.
  • alkylene group i.e., a group having the general structure -alkylene-NH-alkyl or -alkylene-N(alkyl)(alkyl)
  • Alkylaminoalkyl groups include, for example, mono- and di-(C 1 -C 8 alkyl)aminoC 1 -C 8 alkyl, mono- and di-(C 1 -C 6 alkyl)aminoC 1 -C 6 alkyl and mono- and di-(C 1 -C 6 alkyl)aminoC 1 -C 4 alkyl.
  • “Mono- or di-(C 1 -C 6 alkyl)aminoC 0 -C 6 alkyl” refers to a mono- or di-(C 1 -C 6 alkyl)amino group linked via a single covalent bond or a C 1 -C 6 alkylene group.
  • alkyl as used in the terms “alkylamino” and “alkylaminoalkyl” differs from the definition of “alkyl” used for all other alkyl-containing groups, in the inclusion of cycloalkyl and (cycloalkyl)alkyl groups (e.g., (C 3 -C 7 cycloalkyl)C 0 -C 6 alkyl).
  • aminocarbonyl refers to an amide group (i.e., —(C ⁇ O)NH 2 ).
  • “Mono- or di-(C 1 -C 0 alkyl)aminocarbonyl” refers to groups of the formula —(C ⁇ O)—N(R) 2 , in which the carbonyl is the point of attachment, one R is C 1 -C 6 alkyl and the other R is hydrogen or an independently chosen C 1 -C 6 alkyl.
  • “Mono- or di-(C 1 -C 6 alkyl)aminocarbonylC 0 -C 4 alkyl” is an aminocarbonyl group in which one or both of the hydrogen atoms is replaced with C 1 -C 6 alkyl, and which is linked via a single covalent bond (i.e., mono- or di-(C 1 -C 6 alkyl)aminocarbonyl) or a C 1 -C 4 alkylene group (i.e., —(C 0 -C 4 alkyl)-(C ⁇ O)N(C 1 -C 6 alkyl) 2 ). If both hydrogen atoms are so replaced, the C 1 -C 6 alkyl groups may be the same or different.
  • aminosulfonyl refers to a sulfonamide group (i.e., —(SO 2 )NH 2 ).
  • “Mono- or di-(C 1 -C 8 alkyl)aminosulfonyl” refers to groups of the formula —(SO 2 )—N(R) 2 , in which the sulfur atom is the point of attachment, one R is C 1 -C 8 alkyl and the other R is hydrogen or an independently chosen C 1 -C 8 alkyl.
  • “Mono- or di-(C 1 -C 6 alkyl)aminosulfonylC 0 -C 4 alkyl” is an aminosulfonyl group in which one or both of the hydrogen atoms is replaced with C 1 -C 6 alkyl, and which is linked via a single covalent bond (i.e., mono- or di-(C 1 -C 6 alkyl)aminosulfonyl) or a C 1 -C 4 alkylene group (i.e., —(C 1 -C 4 alkyl)-(SO 2 )N(C 1 -C 6 alkyl) 2 ). If both hydrogen atoms are so replaced, the C 1 -C 6 alkyl groups may be the same or different.
  • halogen refers to fluorine, chlorine, bromine or iodine.
  • haloalkyl is an alkyl group that is substituted with 1 or more independently chosen halogens (e.g., “C 1 -C 6 haloalkyl” groups have from 1 to 6 carbon atoms).
  • haloalkyl groups include, but are not limited to, mono-, di- or tri-fluoromethyl; mono-, di- or tri-chloromethyl; mono-, di-, tri-, tetra- or penta-fluoroethyl; mono-, di-, tri-, tetra- or penta-chloroethyl; and 1,2,2,2-tetrafluoro-1-trifluoromethyl-ethyl.
  • Typical haloalkyl groups are trifluoromethyl and difluoromethyl.
  • haloalkoxy refers to a haloalkyl group as defined above that is linked via an oxygen bridge.
  • a dash (“-”) that is not between two letters or symbols is used to indicate a point of attachment for a substituent. For example, —CONH 2 is attached through the carbon atom.
  • a “carbocycle” or “carbocyclic group” comprises at least one ring formed entirely by carbon-carbon bonds (referred to herein as a carbocycle), and does not contain a heterocycle. Unless otherwise specified, each ring within a carbocycle may be independently saturated, partially saturated or aromatic, and is optionally substituted as indicated.
  • a carbocycle generally has from 1 to 3 fused, pendant or spiro rings and optionally further contains one or more alkylene bridges; carbocycles within certain embodiments have one ring or two fused rings. Typically, each ring contains from 3 to 8 ring members (i.e., C 3 -C 8 ); C 5 -C 7 rings are recited in certain embodiments.
  • Carbocycles comprising fused, pendant or spiro rings typically contain from 9 to 16 ring members.
  • Certain representative carbocycles are cycloalkyl as described above (e.g., cyclohexyl, cycloheptyl or adamantly).
  • Other carbocycles are aryl (i.e., contain at least one aromatic carbocyclic ring, with or without one or more additional aromatic and/or cycloalkyl rings).
  • Such aryl carbocycles include, for example, phenyl, naphthyl (e.g., 1-naphthyl and 2-naphthyl), fluorenyl, indanyl and 1,2,3,4-tetrahydronaphthyl.
  • carbocycles recited herein are phenyl groups linked via a single covalent bond or C 1 -C 2 alkylene group (e.g., benzyl, 1-phenyl-ethyl and 2-phenyl-ethyl), and are designated phenylC 0 -C 2 alkyl.
  • a “heterocycle” or “heterocyclic group” has from 1 to 3 fused, pendant or spiro rings, at least one of which is a heterocyclic ring (i.e., one or more ring atoms is a heteroatom independently chosen from O, S and N, with the remaining ring atoms being carbon). Additional rings, if present, may be heterocyclic or carbocyclic. Typically, a heterocyclic ring comprises 1, 2, 3 or 4 heteroatoms; within certain embodiments each heterocycle has 1 or 2 heteroatoms per ring.
  • Each heterocyclic ring generally contains from 3 to 8 ring members (rings having from 4 or 5 to 7 ring members are recited in certain embodiments) and heterocycles comprising fused, pendant or spiro rings typically contain from 9 to 14 ring members.
  • Certain heterocycles comprise a sulfur atom as a ring member; in certain embodiments, the sulfur atom is oxidized to SO or SO 2 .
  • a heterocycle may be a heterocycloalkyl group (i.e., each ring is saturated or partially saturated), such as a 4- to 7-membered heterocycloalkyl, which generally comprises 1, 2, 3 or 4 ring atoms that are independently chosen from C, O, N and S; or a heteroaryl group (i.e., at least one ring within the group is aromatic), such as a 5- to 10-membered heteroaryl (which may be monocyclic or bicyclic) or a 6-membered heteroaryl (e.g., pyridyl or pyrimidyl).
  • N-linked heterocyclic groups are linked via a component nitrogen atom.
  • a “heterocycleC 0 -C 4 alkyl” is a heterocyclic group linked via a single covalent bond or C 1 -C 4 alkylene group.
  • a “(4- to 7-membered heterocycloalkyl)C 1 -C 4 alkyl” is a heterocycloalkyl ring with from 4 to 7 ring members that is linked via a C 1 -C 4 alkylene group.
  • a “(4- to 7-membered heterocycloalkyl)C 0 -C 4 alkylene” is a divalent (4- to 7-membered heterocycloalkyl)C 0 -C 4 alkyl group that is linked via two single covalent bonds to two specified moieties.
  • one such single covalent bond is located on the cyclic portion and the other is located on the alkylene portion, if present; alternatively, if no alkylene group is present, both such single covalent bonds are located on different ring members.
  • R A if L is a (piperidinyl)C 2 alkylene, A is absent and M is —COOH, one R A moiety so formed is:
  • a “substituent,” as used herein, refers to a molecular moiety that is covalently bonded to an atom within a molecule of interest.
  • a ring substituent may be a moiety such as a halogen, alkyl group, haloalkyl group or other group that is covalently bonded to an atom (preferably a carbon or nitrogen atom) that is a ring member.
  • Substituents of aromatic groups are generally covalently bonded to a ring carbon atom.
  • substitution refers to replacing a hydrogen atom in a molecular structure with a substituent, such that the valence on the designated atom is not exceeded, and such that a chemically stable compound (i.e., a compound that can be isolated, characterized, and tested for biological activity) results from the substitution.
  • Groups that are “optionally substituted” are unsubstituted or are substituted by other than hydrogen at one or more available positions, typically 1, 2, 3, 4 or 5 positions, by one or more suitable groups (which may be the same or different).
  • Optional substitution is also indicated by the phrase “substituted with from 0 to X substituents,” where X is the maximum number of possible substituents.
  • Certain optionally substituted groups are substituted with from 0 to 2, 3 or 4 independently selected substituents (i.e., are unsubstituted or substituted with up to the recited maximum number of substituents).
  • Other optionally substituted groups are substituted with at least one substituent (e.g., substituted with from 1 to 2, 3 or 4 independently selected substituents).
  • P2X 7 receptor refers to any P2X7 receptor, preferably a mammalian receptor such as the human or rat P2X7 receptor disclosed in U.S. Pat. No. 6,133,434, as well as homologues thereof found in other species.
  • P2X 7 receptor modulator also referred to herein as a “modulator,” is a compound that modulates P2X 7 receptor activation and/or P2X 7 receptor-mediated activity (e.g., signal transduction).
  • P2X 7 receptor modulators specifically provided herein are compounds of Formula A and pharmaceutically acceptable salts, hydrates and esters thereof.
  • a modulator may be a P2X 7 receptor agonist or antagonist.
  • a modulator is considered an “antagonist” if it detectably inhibits P2X 7 receptor-mediated signal transduction (using, for example, a representative assay provided in Example 4); in general, such an antagonist inhibits P2X 7 receptor activation with a IC 50 value of less than 20 micromolar, preferably less than 10 micromolar, more preferably less than 5 micromolar, more preferably less than 1 micromolar, still more preferably less than 500 nanomolar, and most preferably less than 100 nanomolar within an assay provided in Example 4.
  • P2X 7 receptor antagonists include neutral antagonists and inverse agonists.
  • An “inverse agonist” of P2X 7 receptor is a compound that reduces the activity of P2X 7 receptor below its basal activity level in the absence of added ligand. Inverse agonists of P2X 7 receptor may also inhibit the activity of ligand at P2X 7 receptor and/or binding of ligand to P2X 7 receptor.
  • the basal activity of P2X 7 receptor, as well as a reduction in P2X 7 receptor activity due to the presence of P2X 7 receptor antagonist, may be determined from a calcium mobilization assay (e.g., the assay of Example 4).
  • a “neutral antagonist” of P2X 7 receptor is a compound that inhibits the activity of ligand at P2X 7 receptor, but does not significantly change the basal activity of the receptor (i.e., within a calcium mobilization assay as described in Example 4 performed in the absence of ligand, P2X 7 receptor activity is reduced by no more than 10%, preferably by no more than 5%, and more preferably by no more than 2%; most preferably, there is no detectable reduction in activity).
  • Neutral antagonists of P2X 7 receptor may inhibit the binding of ligand to P2X 7 receptor.
  • P2X 7 receptor agonist is a compound that elevates the activity of the P2X 7 receptor above the basal activity level of the receptor (i.e., enhances P2X 7 receptor activation and/or P2X 7 receptor-mediated activity, such as signal transduction).
  • P2X 7 receptor agonist activity may be detected using the representative assay provided in Example 4.
  • P2X 7 receptor agonists include ATP and 2′(3′)—O-(4-benzoyl-benzoyl)adenosine 5′-triphosephate (BzATP).
  • a “therapeutically effective amount” is an amount that, upon administration to a patient, results in a discernible patient benefit (e.g., provides detectable relief from at least one condition being treated). Such relief may be detected using any appropriate criteria, including alleviation of one or more symptoms such as pain.
  • a therapeutically effective amount or dose generally results in a concentration of compound in a body fluid (such as blood, plasma, serum, CSF, synovial fluid, lymph, cellular interstitial fluid, tears or urine) that is sufficient to alter P2X 7 receptor-mediated signal transduction (using an assay provided in Example 4). It will be apparent that the discernible patient benefit may be apparent after administration of a single dose, or may become apparent following repeated administration of the therapeutically effective dose according to a predetermined regimen, depending upon the indication for which the compound is administered.
  • statically significant is meant results varying from control at the p ⁇ 0.1 level of significance as measured using a standard parametric assay of statistical significance such as a student's T test.
  • a “patient” is any individual treated with a compound provided herein. Patients include humans, as well as other animals such as companion animals (e.g., dogs and cats) and livestock. Patients may be experiencing one or more symptoms of a condition responsive to P2X 7 receptor modulation or may be free of such symptom(s) (i.e., treatment may be prophylactic in a patient considered at risk for the development of such symptoms).
  • the present invention provides 5-membered heterocyclic amides and related compounds of Formula A and other formulas recited herein.
  • modulators that may be used in a variety of contexts, including in the treatment of conditions responsive to P2X 7 receptor modulation, such as pain.
  • Such modulators are also useful as probes for detection and localization of P2X 7 receptor and as standards in P2X 7 receptor-mediated signal transduction assays.
  • X is not absent (e.g., C 1 -C 3 alkylene, optionally substituted as described above).
  • R 2 in certain embodiments, represents zero substituents or 1 or 2 substituents independently chosen from halogen, C 1 -C 6 alkyl and C 1 -C 6 haloalkyl.
  • Representative subformulas of Formulas A, I and IIa-c include, for example:
  • each R 3 is independently hydrogen, halogen, cyano, amino, nitro, aminocarbonyl, aminosulfonyl, —COOH, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 haloalkyl, C 1 -C 6 hydroxyalkyl, C 1 -C 6 -aminoalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy, C 1 -C 6 alkanoyl, C 2 -C 6 alkyl ether, (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl, mono- or di-(C 1 -C 6 alkyl)amino, C 1 -C 6 alkylsulfonyl, C 1 -C 6 alkanoylamino, mono- or di-(C 1 -C 6 alkyl)aminocarbon
  • any CH or NH moiety at Z 1 , Z 2 , Z 3 , Z 4 , Z 5 and/or Z 6 may be substituted with a substituent represented by R A or R 2 , and that such substitution will result in the replacement of the hydrogen atom with a substituent represented by R A or R 2 .
  • each R 3 is independently hydrogen, halogen, C 1 -C 4 alkyl or C 1 -C 4 haloalkyl.
  • R A is phenylC 0 -C 2 alkyl, (5- or 6-membered heteroaryl)C 0 -C 4 alkyl, phenylC 0 -C 2 alkyl-T- or (5- or 6-membered heteroaryl)C 0 -C 4 alkyl-T-, wherein T is S or O; each of which is substituted with from 0 to 4 substituents independently chosen from:
  • R A represents pyrazole or imidazole
  • T is not O
  • R A is not unsubstituted benzyl.
  • R A is phenylC 0 -C 2 alkyl or (5- or 6-membered heteroaryl)C 0 -C 4 alkyl, optionally substituted as described above, such that R A is not unsubstituted benzyl.
  • R A moieties include, for example, (5- or 6-membered heteroaryl)C 0 -C 4 alkyl that is substituted with from 0 to 4 substituents independently chosen from oxo, amino, halogen, hydroxy, cyano, aminocarbonyl, aminosulfonyl, —COOH, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 1 -C 6 hydroxyalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy, C 1 -C 6 alkylthio, C 2 -C 6 alkyl ether, C 1 -C 6 alkanoylamino, mono- or di-(C 1 -C 6 alkyl)amino, C 1 -C 6 alkylsulfonyl, C 1 -C 6 alkylsulfonylamino, mono- or di-(C 1 -C 6 alkyl)aminosulfon
  • R A is C 3 -C 6 alkyl, C 2 -C 6 alkenyl, C 1 -C 6 alkoxy, C 1 -C 6 alkylthio, C 2 -C 6 alkenylthio, C 2 -C 6 alkyl ether, C 1 -C 6 alkoxycarbonyl, C 1 -C 6 alkylsulfonyl, C 1 -C 6 alkylsulfonylamino, mono- or di-C 1 -C 6 alkylaminosulfonyl, mono- or di-(C 1 -C 6 alkyl)aminoC 0 -C 4 alkyl, (4- to 7-membered heterocycloalkyl)C 0 -C 4 alkyl or (4- to 7-membered heterocycloalkyl)C 0 -C 4 alkyl
  • R A is not optionally substituted alkoxy.
  • R A is C 3 -C 6 alkyl, C 2 -C 6 alkyl ether, C 1 -C 4 alkoxycarbonyl, mono- or di-(C 1 -C 6 alkyl)aminoC 0 -C 4 alkyl, or (4- to 7-membered heterocycloalkyl)C 0 -C 4 alkyl; each of which is substituted with from 0 to 4 substituents independently chosen from amino, hydroxy, oxo, aminocarbonyl, aminosulfonyl, C 1 -C 6 alkyl, C 1 -C 6 hydroxyalkyl, C 2 -C 6 alkyl ether, mono- or di-(C 1 -C 6 alkyl)amino, mono- or di-(C 1 -C 6 alkyl)aminocarbonyl, C 1 -C 6 alkylsulfony
  • R A moieties include, for example, C 3 -C 6 alkyl, C 2 -C 6 alkyl ether, and mono- or di-(C 1 -C 6 alkyl)aminoC 0 -C 4 alkyl, each of which is substituted with from 1 to 4 substituents independently chosen from halogen, hydroxy, amino, oxo, aminocarbonyl, aminosulfonyl, —COOH, C 1 -C 6 alkoxy, mono- or di-(C 1 -C 6 alkyl)amino, C 1 -C 6 alkanoylamino, C 1 -C 6 alkylsulfonyl, C 1 -C 6 alkylsulfonyloxy, C 1 -C 6 alkylsulfonylamino, and 4- to 7-membered heterocycle.
  • R A is: (i) C 3 -C 6 alkyl that is substituted with amino, hydroxy or —COOH; or (ii) mono- or di-(C 1 -C 6 alkyl)aminoC 0 -C 2 alkyl that is substituted with from 0 to 2 substituents independently chosen from hydroxy, oxo, amino, —COOH and C 1 -C 4 alkylsulfonylamino.
  • R A is a group of the formula:
  • G is CH or N; s and t are independently 0, 1, 2, 3 or 4, such that the sum of s and t ranges from 2 to 5; and R 8 is: (i) hydrogen, aminocarbonyl, aminosulfonyl or —COOH; or (ii) C 1 -C 6 alkyl, mono- or di-(C 1 -C 6 alkyl)aminoC 0 -C 4 alkyl, C 1 -C 6 alkylsulfonylC 0 -C 4 alkyl, C 1 -C 6 alkylsulfonylaminoC 0 -C 4 alkyl, or 4- to 7-membered heterocycle; each of which is substituted with from 0 to 4 substituents independently chosen from halogen, hydroxy, amino, oxo, aminocarbonyl, aminosulfonyl, —COOH, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, mono- or di-(C 1
  • R 9 is: (i) C 1 -C 6 alkyl that is substituted with —COOH; or (ii) a 5- or 6-membered heteroaryl that is unsubstituted or substituted with 1 or 2 oxo; and R 10 represents one substituent chosen from: (i) —COOH; (ii) C 1 -C 6 alkyl that is substituted with —COOH; (iii) mono- or di-(C 1 -C 6 alkyl)aminoC 0 -C 2 alkyl, C 1 -C 6 alkylsulfonyl and C 1 -C 6 alkylsulfonylamino; each of which is substituted with from 0 to 3 substituents independently chosen from hydroxy, oxo and —COOH; and (iv) C 1 -C 6 haloalkylsulfonylamino.
  • R 1 represents from 0 to 2 substituents independently chosen from halogen, hydroxy, cyano, amino, nitro, aminocarbonyl, aminosulfonyl, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 haloalkyl, C 1 -C 6 hydroxyalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy, (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl, and mono- or di-(C 1 -C 6 alkyl)amino; and each R 5 is independently hydrogen, C 1 -C 6 alkyl, C 3 -C 7 cycloalkyl or phenyl; or two R 5 are taken together with the carbon atom or carbon atoms to which they are bound to form a C 3 -C 8 cycloalkyl.
  • Representative subformulas of Formula III include, for example:
  • each R 3 is independently hydrogen, halogen, cyano, amino, nitro, aminocarbonyl, aminosulfonyl, —COOH, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 haloalkyl, C 1 -C 6 hydroxyalkyl, C 1 -C 6 -aminoalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy, C 1 -C 6 alkanoyl, C 2 -C 6 alkyl ether, (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl, mono- or di-(C 1 -C 6 alkyl)amino, C 1 -C 6 alkylsulfonyl, C 1 -C 6 alkanoylamino, mono- or di-(C 1 -C 6 alkyl)aminocarbon
  • each R 3 is independently hydrogen, halogen, C 1 -C 4 alkyl or C 1 -C 4 haloalkyl.
  • R A is (5- or 6-membered heteroaryl)C 0 -C 4 alkyl or (5- or 6-membered heteroaryl)C 0 -C 4 alkyl-T-, wherein T is S or O; each of which is substituted with from 0 to 4 substituents independently chosen from: (i) oxo, amino, halogen, hydroxy, cyano, aminocarbonyl, aminosulfonyl and —COOH; and (ii) C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 1 -C 6 hydroxyalkyl, C 1 -C 6 haloalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy, C 1 -C 6 alkylthio, C 2 -C 6 alkenylthio, C 2 -C 6 alkyl ether, (mono- or di-C 1 -C 6 alkylamino
  • R A groups include (5- or 6-membered heteroaryl)C 0 -C 4 alkyl that is substituted with from 0 to 4 substituents independently chosen from oxo, amino, halogen, hydroxy, cyano, aminocarbonyl, aminosulfonyl, —COOH, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 1 -C 6 haloalkyl, C 1 -C 6 hydroxyalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy, C 1 -C 6 alkylthio, C 2 -C 6 alkyl ether, C 1 -C 6 alkanoylamino, mono- or di-(C 1 -C 6 alkyl)amino, C 1 -C 6 alkylsulfonyl, C 1 -C 6 alkylsulfonylamino, mono- or di-(C 1 -C 6 alkyl
  • Representative subformulas of Formula III include, for example:
  • each R 3 is independently hydrogen, halogen, cyano, amino, nitro, aminocarbonyl, aminosulfonyl, —COOH, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 haloalkyl, C 1 -C 6 hydroxyalkyl, C 1 -C 6 -aminoalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy, C 1 -C 6 alkanoyl, C 2 -C 6 alkyl ether, (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl, or mono- or di-(C 1 -C 6 alkyl)amino.
  • each R 3 is independently hydrogen, halogen, C 1 -C 4 alkyl or C 1 -C 4 haloalkyl
  • Representative subformulas of Formulas IV and V include, for example:
  • each R 3 is as described above.
  • each R 3 is independently hydrogen, halogen, cyano, amino, nitro, aminocarbonyl, aminosulfonyl, —COOH, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 haloalkyl, C 1 -C 6 hydroxyalkyl, C 1 -C 0 aminoalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy, C 1 -C 6 alkanoyl, C 2 -C 6 alkyl ether, (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl, or mono- or di-(C 1 -C 6 alkyl)amino.
  • each R 3 is independently hydrogen, halogen, C 1 -C 4 alkyl or C 1 -C 4 haloalkyl.
  • R A is (5- or 6-membered heteroaryl)C 0 -C 4 alkyl that is substituted with from 0 to 4 substituents independently chosen from: (i) oxo, amino, halogen, hydroxy, cyano, aminocarbonyl, aminosulfonyl and —COOH; and (ii) C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 1 -C 6 haloalkyl, C 1 -C 6 hydroxyalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy, C 1 -C 6 alkylthio, C 2 -C 6 alkyl ether, C 1 -C 6 alkanoylamino, mono- or di-(C 1 -C 6 alkyl)amino, C 1 -C 6 alkylsulfonyl, C 1 -C 6 alkyl
  • the (5- or 6-membered heteroaryl)C 0 -C 4 alkyl (which is unsubstituted or substituted as described above) is thiazolyl, pyrazolyl, pyridinyl, pyrimidinyl or pyridazinyl.
  • Y is phenyl or a 6-membered heteroaryl, each of which is substituted with from 0 to 3 substituents independently chosen from hydroxy, halogen, cyano, amino, nitro, oxo, aminocarbonyl, aminosulfonyl, —COOH, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 haloalkyl, C 1 -C 6 hydroxyalkyl, C 1 -C 6 -aminoalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy, C 2 -C 6 alkyl ether, C 1 -C 0 alkanoyl, C 1 -C 6 alkylsulfonyl, (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl,
  • R 1 represents from 0 to 3 substituents independently chosen from halogen, hydroxy, cyano, amino, nitro, aminocarbonyl, aminosulfonyl, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 haloalkyl, C 1 -C 6 hydroxyalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy, (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl, and mono- or di-(C 1 -C 0 alkyl)amino; and each R 5 is independently hydrogen, C 1 -C 6 alkyl, C 3 -C 7 cycloalkyl, phenyl, or taken together with another R 5 attached to the same carbon atom, and the carbon atom to which they are bound, to form a C 3 -C 8
  • R A is (5- or 6-membered heteroaryl)C 0 -C 4 alkyl that is substituted with from 0 to 4 substituents independently chosen from: (i) oxo, amino, halogen, hydroxy, cyano, aminocarbonyl, aminosulfonyl and —COOH; and (ii) C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 1 -C 6 haloalkyl, C 1 -C 6 hydroxyalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy, C 1 -C 6 alkylthio, C 2 -C 6 alkyl ether, C 1 -C 6 alkanoylamino, mono- or di-(C 1 -C 6 alkyl)amino, C 1 -C 6 alkylsulfonyl, C 1 -C 6 alkylsulfonylamino, mono- or di-(C 1
  • At least one R 5 is not hydrogen (e.g., both R 5 are taken together with the carbon atom to which they are bound to form a C 3 -C 8 cycloalkyl).
  • R 3 is hydrogen, halogen, C 1 -C 4 alkyl or C 1 -C 4 haloalkyl.
  • R 1 represents from 0 to 3 substituents independently chosen from halogen, hydroxy, cyano, amino, nitro, aminocarbonyl, aminosulfonyl, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 haloalkyl, C 1 -C 6 hydroxyalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy, (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl, and mono- or di-(C 1 -C 6 alkyl)amino; and each R 5 is independently hydrogen, C 1 -C 6 alkyl, C 3 -C 7 cycloalkyl or phenyl; or taken together with another R 5 moiety and the carbon atom to which they are bound to foam a C 3 -C 8 cycloalkyl.
  • R A is phenyl or 5- or 6-membered heteroaryl, each of which is substituted with from 0 to 4 substituents independently chosen from: (i) oxo, amino and halogen; and (ii) C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 1 -C 6 haloalkyl, C 1 -C 6 hydroxyalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy, C 2 -C 6 alkyl ether, mono- or di-(C 1 -C 6 alkyl)amino, and (4- to 7-membered heterocycle)C 0 -C 2 alkyl, each of which is substituted with from 0 to 4 substituents independently chosen from oxo, amino, halogen, hydroxy, cyano, C 1 -C 4 alkyl, C 2 -C 4 alkenyl and C 1 -C 4 haloalky
  • R A is 5- to 7-membered heterocycloalkyl that is substituted with from 0 to 4 substituents independently chosen from: (i) oxo, amino and halogen; and (ii) C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 1 -C 6 haloalkyl, C 1 -C 6 hydroxyalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy, C 2 -C 6 alkyl ether, mono- or di-(C 1 -C 6 alkyl)amino, and (4- to 7-membered heterocycle)C 0 -C 2 alkyl, each of which is substituted with from 0 to 4 substituents independently chosen from oxo, amino, halogen, hydroxy, cyano, C 1 -C 4 alkyl, C 2 -C 4 alkenyl and C 1 -C 4 haloalkyl.
  • one R 5 moiety is a N-linked 5- or 6-membered heterocycloalkyl group (e.g., piperazinyl, morpholinyl, thiomorpholinyl, piperidinyl or pyrrolidinyl), each of which is unsubstituted or substituted with one substituent such as hydroxy, halogen, aminocarbonyl, C 1 -C 4 alkoxy, mono- or di-(C 1 -C 4 alkyl)amino or 4- to 7-membered heterocycloalkyl.
  • one substituent such as hydroxy, halogen, aminocarbonyl, C 1 -C 4 alkoxy, mono- or di-(C 1 -C 4 alkyl)amino or 4- to 7-membered heterocycloalkyl.
  • V is absent or methylene.
  • Z 1 , Z 2 and Z 3 are independently N, NH, CH, S or O; such that no more than one of Z 1 , Z 2 and Z 3 is chosen from O and S. Certain such compounds satisfy one or more of the following formulas:
  • each R 3 is independently hydrogen, halogen, cyano, amino, nitro, aminocarbonyl, aminosulfonyl, —COOH, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 haloalkyl, C 1 -C 6 hydroxyalkyl, C 1 -C 6 -aminoalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy, C 1 -C 6 alkanoyl, C 2 -C 6 alkyl ether, (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl, mono- or di-(C 1 -C 6 alkyl)amino, C 1 -C 6 alkylsulfonyl, C 1 -C 6 alkanoylamino, mono- or di-(C 1 -C 6 alkyl)aminocarbon
  • Z 4 and Z 5 are independently CH, N, NH, O or S; and Z 6 is CH, S or O; such that no more than one of Z 4 , Z 5 and Z 6 is chosen from O and S. Certain such compounds further satisfy the formula:
  • R 3 is as described above.
  • each R 3 is independently hydrogen, halogen, C 1 -C 4 alkyl or C 1 -C 4 haloalkyl.
  • R A is as described above, or R A is C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 1 -C 6 alkoxy, C 1 -C 6 alkylthio, C 2 -C 6 alkenylthio, C 2 -C 6 alkyl ether, C 1 -C 6 alkoxycarbonyl, C 1 -C 6 alkylsulfonyl, C 1 -C 6 alkylsulfonylamino, mono- or di-C 1 -C 6 alkylaminosulfonyl, mono- or di-(C 1 -C 6 alkyl)aminoC 0 -C 4 alkyl, (4- to 7-membered heterocycloalky
  • R A is not optionally substituted C 1 -C 6 alkoxy.
  • R A is C 1 -C 6 alkyl, C 2 -C 6 alkyl ether, C 1 -C 4 alkoxycarbonyl, mono- or di-(C 1 -C 6 alkyl)aminoC 0 -C 4 alkyl, or (4- to 7-membered heterocycloalkyl)C 0 -C 4 alkyl; each of which is substituted with from 0 to 4 substituents independently chosen from amino, hydroxy, oxo, aminocarbonyl, aminosulfonyl, C 1 -C 6 alkyl, C 1 -C 6 hydroxyalkyl, C 2 -C 6 alkyl ether, mono- or di-(C 1 -C 6 alkyl)amino, mono- or di-(C 1 -C 6 alkyl)aminocarbonyl, C 1 -C 6 alkyl
  • R 11 and R 12 are taken together, with the nitrogen atom to which they are bound, to form a 4- to 7-membered heterocycloalkyl that is substituted with from 0 to 4 substituents independently chosen from hydroxy, halogen, cyano, amino, C 1 -C 6 alkyl, C 1 -C 6 hydroxyalkyl, C 1 -C 6 haloalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy, C 2 -C 6 alkyl ether, C 1 -C 6 alkanoylamino, mono- or di-(C 1 -C 6 alkyl)amino, C 1 -C 6 alkylsulfonyl, C 1 -C 6 alkylsulfonylamino, mono- or di-(C 1 -C 6 alkyl)aminosulfonyl, mono- or di-(C 1 -C 6 alkylamino)carbonyl, and 4-
  • R 5 moieties are taken together, with the carbon atom to which they are bound, to form a C 3 -C 8 cycloalkyl.
  • R 3 is halogen, cyano, C 1 -C 4 haloalkyl, phenyl or 5- or 6-membered heteroaryl; and/or Y is piperidinyl, piperazinyl, morpholinyl or thiomorpholinyl, each of which is substituted with from 0 to 2 substituents independently chosen from C 1 -C 6 alkyl.
  • Y is piperidinyl, piperazinyl, morpholinyl or thiomorpholinyl, each of which is substituted with from 0 to 2 substituents independently chosen from C 1 -C 6 alkyl.
  • R 13 is hydrogen or C 1 -C 4 alkyl.
  • R A is phenyl or 5- or 6-membered heteroaryl, each of which is substituted with from 0 to 4 substituents independently chosen from: (i) oxo, amino, and halogen; and (ii) C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 1 -C 6 haloalkyl, C 1 -C 6 hydroxyalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy, C 2 -C 6 alkyl ether, mono- or di-(C 1 -C 6 alkyl)amino, and (4- to 7-membered heterocycle)C 0 -C 2 alkyl, each of which is substituted with from 0 to 4 substituents independently chosen from oxo, amino, halogen, hydroxy, cyano, C 1 -C 4 alkyl, C 2 -C 4 alkenyl and C 1 -C 4 haloal
  • R A is 5- to 7-membered heterocycloalkyl that is substituted with from 0 to 4 substituents independently chosen from: (i) oxo, amino, and halogen; and (ii) C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 1 -C 6 haloalkyl, C 1 -C 6 hydroxyalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy, C 2 -C 6 alkyl ether, mono- or di-(C 1 -C 6 alkyl)amino, and (4- to 7-membered heterocycle)C 0 -C 2 alkyl, each of which is substituted with from 0 to 4 substituents independently chosen from oxo, amino, halogen, hydroxy, cyano, C 1 -C 4 alkyl, C 2 -C 4 alkenyl and C 1 -C 4 haloalkyl.
  • each R 3 is independently hydrogen, halogen, cyano, amino, nitro, aminocarbonyl, aminosulfonyl, —COOH, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 haloalkyl, C 1 -C 6 hydroxyalkyl, C 1 -C 6 -aminoalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy, C 1 -C 6 alkanoyl, C 2 -C 6 alkyl ether, (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl, mono- or di-(C 1 -C 6 alkyl)amino, C 1 -C 6 alkylsulfonyl, C 1 -C 6 alkanoylamino, mono- or di-(C 1 -C 6 alkyl)aminocarbon
  • R A moieties include, for example, phenylC 0 -C 2 alkyl, (5- or 6-membered heteroaryl)C 0 -C 4 alkyl, phenylC 0 -C 2 alkyl-T- or (5- or 6-membered heteroaryl)C 0 -C 4 alkyl-T-, wherein T is S or O; each of which is substituted with from 0 to 4 substituents independently chosen from: (i) oxo, amino, halogen, hydroxy, cyano, aminocarbonyl, aminosulfonyl and —COOH; and (ii) C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 1 -C 6 hydroxyalkyl, C 1 -C 6 haloalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy, C 1 -C 6 alkylthio, C 2 -C 6 alkenylthio
  • R 1 represents from 0 to 3 substituents independently chosen from halogen, hydroxy, cyano, amino, nitro, aminocarbonyl, aminosulfonyl, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 haloalkyl, C 1 -C 6 hydroxyalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy, (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl, and mono- or di-(C 1 -C 6 alkyl)amino.
  • Representative 5-membered heterocyclic amides and related compounds provided herein include, but are not limited to, those specifically described in Examples 1-3. It will be apparent that the specific compounds recited herein are representative only, and are not intended to limit the scope of the present invention. Further, as noted above, all compounds of the present invention may be present as a free acid or base, or as a pharmaceutically acceptable salt. In addition, other forms such as hydrates and prodrugs of such compounds are specifically contemplated by the present invention.
  • 5-membered heterocyclic amides and related compounds provided herein detectably alter (modulate) P2X 7 receptor activity, as determined using an assay such as an assay recited in Example 4, herein. Additional assays that may be used for this purpose include assays that measure IL-1 ⁇ release; assays that measure uptake of a membrane-impermeant fluorescent dye such as YO—PRO1; assays that measure lucifer yellow uptake; assays that measure ethidium bromide uptake; and assays that use calcium imaging to detect P2X 7 activity; all of which assays are well known in the art. Certain modulators provided herein detectably modulate P2X 7 receptor activity at micromolar concentrations, at nanomolar concentrations, or at subnanomolar concentrations.
  • IC 50 values for such compounds may be determined using a standard in vitro P2X 7 receptor-mediated calcium mobilization assay, as provided in Example 4. Briefly, cells expressing P2X 7 receptor are contacted with a compound of interest and with an indicator of intracellular calcium concentration (e.g., a membrane permeable calcium sensitivity dye such as Fluo-3, Fluo-4 or Fura-2 (Invitrogen, Carlsbad, Calif.), each of which produce a fluorescent signal when bound to Ca ++ ). Such contact is preferably carried out by one or more incubations of the cells in buffer or culture medium comprising either or both of the compound and the indicator in solution.
  • an indicator of intracellular calcium concentration e.g., a membrane permeable calcium sensitivity dye such as Fluo-3, Fluo-4 or Fura-2 (Invitrogen, Carlsbad, Calif.
  • P2X 7 receptor agonist e.g., ATP or 2′(3′)—O—(4-benzoyl-benzoyl)adenosine 5′-triphosephate at, for example, a concentration equal to the EC 50 concentration
  • fluorescence response is measured.
  • P2X 7 receptor antagonists When agonist-contacted cells are contacted with a compound that is a P2X 7 receptor antagonist, the fluorescence response is generally reduced by at least 20%, preferably at least 50% and more preferably at least 80%, as compared to cells that are contacted with the agonist in the absence of test compound.
  • P2X 7 receptor antagonists provided herein exhibit no detectable agonist activity an in vitro assay of P2X 7 receptor agonism at a concentration of compound equal to the IC 50 .
  • Certain such antagonists exhibit no detectable agonist activity an in vitro assay of P2X 7 receptor agonism at a concentration of compound that is 100-fold higher than the IC 50 .
  • P2X 7 receptor modulating activity may also, or alternatively, be assessed using an in vivo pain relief assay as provided in Example 5.
  • Modulators provided herein preferably have a statistically significant specific effect on P2X 7 receptor activity within such a functional assay.
  • preferred modulators are non-sedating.
  • a dose of modulator that is twice the minimum dose sufficient to provide analgesia in an animal model for determining pain relief causes only transient (i.e., lasting for no more than 1 ⁇ 2 the time that pain relief lasts) or preferably no statistically significant sedation in an animal model assay of sedation (using the method described by Fitzgerald et al. (1988) Toxicology 49 (2-3):433-9).
  • a dose that is five times the minimum dose sufficient to provide analgesia does not produce statistically significant sedation.
  • a modulator provided herein does not produce 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, more preferably less than 30 mg/kg).
  • compounds provided herein may be evaluated for certain pharmacological properties including, but not limited to, oral bioavailability (preferred compounds are orally bioavailable to an extent allowing for therapeutically effective concentrations of the compound to be achieved at oral doses of less than 140 mg/kg, preferably less than 50 mg/kg, more preferably less than 30 mg/kg, even more preferably less than 10 mg/kg, still more preferably less than 1 mg/kg and most preferably less than 0.1 mg/kg), toxicity (a preferred compound is nontoxic when a therapeutically effective amount is administered to a subject), side effects (a preferred compound produces side effects comparable to placebo when a therapeutically effective amount of the compound is administered to a subject), serum protein binding and in vitro and in vivo half-life (a preferred compound exhibits an in vivo half-life allowing for Q.I.D.
  • differential penetration of the blood brain barrier may be desirable for modulators used to treat pain or neurodegenerative disease by modulating CNS P2X 7 receptor activity such that total daily oral doses as described above provide such modulation to a therapeutically effective extent, while low brain levels of modulators used to treat peripheral nerve mediated pain or certain inflammatory diseases (e.g. rheumatoid arthritis) may be preferred (i.e., such doses do not provide brain (e.g., CSF) levels of the compound sufficient to significantly modulate P2X 7 receptor activity).
  • CNS P2X 7 receptor activity preferably T.I.D. dosing, more preferably B.I.D. dosing, and most preferably once-a-day dosing.
  • modulators used to treat pain or neurodegenerative disease by modulating CNS P2X 7 receptor activity such that total daily oral doses as described above provide such modulation to a therapeutically effective extent
  • low brain levels of modulators used to treat peripheral nerve mediated pain or certain inflammatory diseases e.g. rheum
  • Routine assays that are well known in the art may be used to assess these properties, and identify superior compounds for a particular use.
  • assays used to predict bioavailability include transport across human intestinal cell monolayers, including Caco-2 cell monolayers.
  • 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).
  • Serum protein binding may be predicted from albumin binding assays.
  • Compound half-life is inversely proportional to the frequency of dosage of a compound. In vitro half-lives of compounds may be predicted from assays of microsomal half-life as described, for example, within Example 7 of U.S. Patent Application Publication Number 2005/0070547.
  • nontoxic shall be understood in a relative sense and is intended to refer to any substance that has been approved by the United States Food and Drug Administration (“FDA”) for administration to mammals (preferably humans) or, in keeping with established criteria, is susceptible to approval by the FDA for administration to mammals (preferably humans).
  • FDA United States Food and Drug Administration
  • a highly preferred nontoxic compound generally satisfies one or more of the following criteria: (1) does not substantially inhibit cellular ATP production; (2) does not significantly prolong heart QT intervals; (3) does not cause substantial liver enlargement, or (4) does not cause substantial release of liver enzymes.
  • a compound that does not substantially inhibit cellular ATP production is a compound that satisfies the criteria set forth in Example 8 of U.S. Patent Application Publication Number 2005/0070547.
  • cells treated as described therein with 100 ⁇ M of such a compound exhibit ATP levels that are at least 50% of the ATP levels detected in untreated cells.
  • such cells exhibit ATP levels that are at least 80% of the ATP levels detected in untreated cells.
  • a compound that does not significantly prolong heart QT intervals is a compound that does not result in a statistically significant prolongation of heart QT intervals (as determined by electrocardiography) in guinea pigs, minipigs or dogs upon administration of a dose that yields a serum concentration equal to the EC 50 or IC 50 for the compound.
  • a dose of 0.01, 0.05, 0.1, 0.5, 1, 5, 10, 40 or 50 mg/kg administered parenterally or orally does not result in a statistically significant prolongation of heart QT intervals.
  • a compound does not cause substantial liver enlargement if daily treatment of laboratory rodents (e.g., mice or rats) for 5-10 days with a dose that yields a serum concentration equal to the EC 50 or IC 50 for the compound results in an increase in liver to body weight ratio that is no more than 100% over matched controls. In more highly preferred embodiments, such doses do not cause liver enlargement of more than 75% or 50% over matched controls. If non-rodent mammals (e.g., dogs) are used, such doses should not result in an increase of liver to body weight ratio of more than 50%, preferably not more than 25%, and more preferably not more than 10% over matched untreated controls. Preferred doses within such assays include 0.01, 0.05, 0.1, 0.5, 1, 5, 10, 40 or 50 mg/kg administered parenterally or orally.
  • a compound does not promote substantial release of liver enzymes if administration of twice the minimum dose that yields a serum concentration equal to the EC 50 or IC 50 at P2X 7 receptor for the compound does not elevate serum levels of ALT, LDH or AST in laboratory animals (e.g., rodents) by more than 100% over matched mock-treated controls. In more highly preferred embodiments, such doses do not elevate such serum levels by more than 75% or 50% over matched controls.
  • a compound does not promote substantial release of liver enzymes if, in an in vitro hepatocyte assay, concentrations (in culture media or other such solutions that are contacted and incubated with hepatocytes in vitro) that are equal to the EC 50 or IC 50 for the compound do not cause detectable release of any of such liver enzymes into culture medium above baseline levels seen in media from matched mock-treated control cells. In more highly preferred embodiments, there is no detectable release of any of such liver enzymes into culture medium above baseline levels when such compound concentrations are five-fold, and preferably ten-fold the EC 50 or IC 50 for the compound.
  • certain preferred compounds do not inhibit or induce microsomal cytochrome P450 enzyme activities, such as CYP1A2 activity, CYP2A6 activity, CYP2C9 activity, CYP2C19 activity, CYP2D6 activity, CYP2E1 activity or CYP3A4 activity at a concentration equal to the EC 50 or IC 50 at P2X 7 receptor for the compound.
  • microsomal cytochrome P450 enzyme activities such as CYP1A2 activity, CYP2A6 activity, CYP2C9 activity, CYP2C19 activity, CYP2D6 activity, CYP2E1 activity or CYP3A4 activity at a concentration equal to the EC 50 or IC 50 at P2X 7 receptor for the compound.
  • Certain preferred compounds are not clastogenic (e.g., as determined using a mouse erythrocyte precursor cell micronucleus assay, an Ames micronucleus assay, a spiral micronucleus assay or the like) at a concentration equal the EC 50 or IC 50 for the compound.
  • certain preferred compounds do not induce sister chromatid exchange (e.g., in Chinese hamster ovary cells) at such concentrations.
  • modulators provided herein may be isotopically-labeled or radiolabeled.
  • compounds may have one or more atoms replaced by an atom of the same element having an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes that can be present in the compounds provided herein include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine and chlorine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 15 N, 18 O, 17 O, 31 P, 32 P, 35 S, 18 F and 36 Cl.
  • substitution with heavy isotopes such as deuterium (i.e., 2 H) can afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements and, hence, may be preferred in some circumstances.
  • 5-membered heterocyclic amides and related compounds may generally be prepared using standard synthetic methods. Starting materials are commercially available from suppliers such as Sigma-Aldrich Corp. (St. Louis, Mo.), or may be synthesized from commercially available precursors using established protocols. By way of example, a synthetic route similar to that shown in any of the following Schemes may be used, together with synthetic methods known in the art of synthetic organic chemistry. In some cases, protecting groups may be required during preparation. Such protecting groups can be removed by methods well known to those of ordinary skill in the art, such as methods described in Greene and Wuts, “Protective Groups in Organic Synthesis” (2 nd Edition, John Wiley & Sons, 1991).
  • Scheme 6 illustrates the preparation of certain compounds provided herein that have a thiazole core.
  • Thiazole 5-carboxylic acid ester is made by condensing carbothioic acid amide 1 and 2-halogen-3-oxo-carboxylic acid ester. Hydrolysis of the ester followed by amination of the resulting acid generates the desired product.
  • Scheme 7 illustrates a general route for the preparation of certain compounds provided herein that have a thiazole core.
  • Condensation of thiourea and the 2-halogen-3-oxo-carboxylic acid ester generates the 2-aminothiazole, which is converted to the bromothiazole via Sandermyer reaction.
  • the bromothiazole can be either coupled to an organometallic compound under transition metal catalyzed reaction conditions, such as a boronic acid under Suzuki condition or an organostannane under Stille reaction condition, or condensed directly with a heterocycle to give the thiazole ester. Hydrolysis of the thiazole ester followed by amination of the resulting acid generates the desired product.
  • Scheme 8 illustrates a general method for preparing certain compounds provided herein that have a thiazole core. Protection of the 2-aminothiazole 5-carboxylic acid ester by t-Boc, followed by hydrolysis, amination, deprotection, and Sandermyer reaction generates the bromothiazole.
  • the desired product is prepared by coupling the bromothiazole with an organometallic compound under transition metal catalyzed reaction conditions or by condensing the bromothiazole directly with a heterocycle.
  • a thiophene carboxylic acid is reacted with an amine in the presence of a base such as TEA and a dehydrating agent such as 2-chloro-1,3-dimethylimidazolinium chloride in a solvent such as dimethylacetamide.
  • a base such as TEA
  • a dehydrating agent such as 2-chloro-1,3-dimethylimidazolinium chloride in a solvent such as dimethylacetamide.
  • Scheme 15 illustrates a general method for preparing compounds of Formula X. Curtius rearrangement of the acid generates the amino intermediate, which provides the compound of Formula X upon amination.
  • Scheme 16 illustrates a general method of preparing certain intermediates NH 2 —CH 2 —CHR 5 —Y of Formula X wherein R 5 is substituted phenyl or heteroaryl and Y ⁇ N—R 5b R 5b wherein R 5b is independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 7 cycloalkyl; or both R 5b are taken together to form a heterocycle.
  • Scheme 17 illustrates a general method of preparing certain intermediates NH 2 —CH 2 —CHR 5 —Y or NH 2 —CH 2 —CR 5 R 5 —Y of Formula X wherein Y is substituted phenyl or heteroaryl.
  • Alkylation of the starting acetonitrile with one equivalent of X—R 5 (X ⁇ Br or I), with base such as sodium hydride in a solvent such as THF-DMSO gives intermediate Y—X(R 5 )—CN.
  • Scheme 18 illustrates a general method of preparing certain intermediates NH 2 —CH 2 —CR 5 R 5 —Y of Formula X wherein R 5 is independently hydrogen, C 1 -C 6 alkyl, C 3 -C 7 cycloalkyl or phenyl; or both R 5 are taken together to form a C 3 -C 8 cycloalkyl or heterocycle, and Y ⁇ N—R 5b R 5b wherein R 5b is C 1 -C 6 alkyl, optionally substituted with halogen, hydroxyl, C 1 -C 4 alkyl, C 1 -C 4 alkoxy or CO 2 H, or both R5 b are taken together to form a 5- to 7-membered heterocycle.
  • a compound provided herein may contain one or more asymmetric carbon atoms, so that the compound can exist in different stereoisomeric forms.
  • Such forms can be, for example, racemates or optically active forms.
  • All stereoisomers are encompassed by the present invention. Nonetheless, it may be desirable to obtain single enantiomers (i.e., optically active forms).
  • Standard methods for preparing single enantiomers include asymmetric synthesis and resolution of the racemates. 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.
  • Compounds may be radiolabeled by carrying out their synthesis using precursors comprising at least one atom that is a radioisotope.
  • Each radioisotope is preferably carbon (e.g., 14 C), hydrogen (e.g., 3 H), sulfur (e.g., 35 S) or iodine (e.g., 125 I).
  • Tritium labeled compounds may also be prepared catalytically via platinum-catalyzed exchange in tritiated acetic acid, acid-catalyzed exchange in tritiated trifluoroacetic acid, or heterogeneous-catalyzed exchange with tritium gas using the compound as substrate.
  • Radiolabeled Compounds may be Conveniently Performed by a Radioisotope supplier specializing in custom synthesis of radiolabeled probe compounds.
  • compositions comprising one or more compounds provided herein, together with at least one physiologically acceptable carrier or excipient.
  • Pharmaceutical compositions may comprise, for example, one or more of water, buffers (e.g., sodium bicarbonate, neutral buffered saline or phosphate buffered saline), ethanol, mineral oil, vegetable oil, dimethylsulfoxide, carbohydrates (e.g., glucose, mannose, sucrose, starch, mannitol or dextrans), proteins, adjuvants, polypeptides or amino acids such as glycine, antioxidants, chelating agents such as EDTA or glutathione and/or preservatives.
  • other active ingredients may (but need not) be included in the pharmaceutical compositions provided herein.
  • compositions may be formulated for any appropriate manner of administration, including, for example, topical, oral, nasal, rectal or parenteral administration.
  • parenteral as used herein includes subcutaneous, intradermal, intravascular (e.g., intravenous), intramuscular, spinal, intracranial, intrathecal and intraperitoneal injection, as well as any similar injection or infusion technique.
  • compositions suitable for oral use are preferred. Such compositions include, for example, tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs.
  • pharmaceutical compositions may be formulated as a lyophilizate.
  • Formulation for topical administration may be preferred for certain conditions (e.g., in the treatment of skin conditions such as burns or itch).
  • Formulation for direct administration into the bladder may be preferred for treatment of urinary incontinence and overactive bladder.
  • Compositions intended for oral use may further comprise one or more components such as sweetening agents, flavoring agents, coloring agents and/or preserving agents in order to provide appealing and palatable preparations.
  • Tablets contain the active ingredient in admixture with physiologically acceptable excipients that are suitable for the manufacture of tablets.
  • excipients include, for example, inert diluents (e.g., calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate), granulating and disintegrating agents (e.g., corn starch or alginic acid), binding agents (e.g., starch, gelatin or acacia) and lubricating agents (e.g., magnesium stearate, stearic acid or talc). Tablets may be formed using standard techniques, including dry granulation, direct compression and wet granulation. The tablets may be uncoated or they may be coated by known techniques.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent (e.g., calcium carbonate, calcium phosphate or kaolin), or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium (e.g., peanut oil, liquid paraffin or olive oil).
  • an inert solid diluent e.g., calcium carbonate, calcium phosphate or kaolin
  • an oil medium e.g., peanut oil, liquid paraffin or olive oil
  • Aqueous suspensions contain the active material(s) in admixture with suitable excipients, such as suspending agents (e.g., sodium carboxymethylcellulose, methylcellulose, hydropropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia); and dispersing or wetting agents (e.g., naturally-occurring phosphatides such as lecithin, condensation products of an alkylene oxide with fatty acids such as polyoxyethylene stearate, condensation products of ethylene oxide with long chain aliphatic alcohols such as heptadecaethyleneoxycetanol, 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 such as polyethylene sorbitan monooleate).
  • suspending agents e.g., sodium carb
  • Aqueous suspensions may also comprise one or more preservatives, such as ethyl or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and/or one or more sweetening agents, such as sucrose or saccharin.
  • Oily suspensions may be formulated by suspending the active ingredient(s) in a vegetable oil (e.g., 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 such as beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and/or flavoring agents may be added to provide palatable oral preparations.
  • Such suspensions may be preserved by the addition of an anti-oxidant 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, a suspending agent and one or more preservatives.
  • a dispersing or wetting agent e.g., a suspending agent
  • Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, such as sweetening, flavoring and coloring agents, may also be present.
  • compositions may also be formulated as oil-in-water emulsions.
  • the oily phase may be a vegetable oil (e.g., olive oil or arachis oil), a mineral oil (e.g., liquid paraffin) or a mixture thereof.
  • Suitable emulsifying agents include naturally-occurring gums (e.g., gum acacia or gum tragacanth), naturally-occurring phosphatides (e.g., soy bean lecithin, and esters or partial esters derived from fatty acids and hexitol), anhydrides (e.g., sorbitan monoleate) and condensation products of partial esters derived from fatty acids and hexitol with ethylene oxide (e.g., polyoxyethylene sorbitan monoleate).
  • An emulsion may also comprise one or more sweetening and/or flavoring agents.
  • Syrups and elixirs may be formulated with sweetening agents, such as glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also comprise one or more demulcents, preservatives, flavoring agents and/or coloring agents.
  • sweetening agents such as glycerol, propylene glycol, sorbitol or sucrose.
  • Such formulations may also comprise one or more demulcents, preservatives, flavoring agents and/or coloring agents.
  • Formulations for topical administration typically comprise a topical vehicle combined with active agent(s), with or without additional optional components.
  • Suitable topical vehicles and additional components are well known in the art, and it will be apparent that the choice of a vehicle will depend on the particular physical form and mode of delivery.
  • Topical vehicles include water; organic solvents such as alcohols (e.g., ethanol or isopropyl alcohol) or glycerin; glycols (e.g., butylene, isoprene or propylene glycol); aliphatic alcohols (e.g., lanolin); mixtures of water and organic solvents and mixtures of organic solvents such as alcohol and glycerin; lipid-based materials such as fatty acids, acylglycerols (including oils, such as mineral oil, and fats of natural or synthetic origin), phosphoglycerides, sphingolipids and waxes; protein-based materials such as collagen and gelatin; silicone-based materials (both non-volatile and volatile); and hydrocarbon-based materials such as microsponges and polymer matrices.
  • organic solvents such as alcohols (e.g., ethanol or isopropyl alcohol) or glycerin
  • glycols e.g., butylene, isoprene or
  • a composition may further include one or more components adapted to improve the stability or effectiveness of the applied formulation, such as stabilizing agents, suspending agents, emulsifying agents, viscosity adjusters, gelling agents, preservatives, antioxidants, skin penetration enhancers, moisturizers and sustained release materials.
  • stabilizing agents such as stabilizing agents, suspending agents, emulsifying agents, viscosity adjusters, gelling agents, preservatives, antioxidants, skin penetration enhancers, moisturizers and sustained release materials.
  • stabilizing agents such as hydroxymethylcellulose or gelatin-microcapsules, liposomes, albumin microspheres, microemulsions, nanoparticles or nanocapsules.
  • a topical formulation may be prepared in any of a variety of physical forms including, for example, solids, pastes, creams, foams, lotions, gels, powders, aqueous liquids and emulsions.
  • the physical appearance and viscosity of such pharmaceutically acceptable forms can be governed by the presence and amount of emulsifier(s) and viscosity adjuster(s) present in the formulation.
  • Solids are generally firm and non-pourable and commonly are formulated as bars or sticks, or in particulate form; solids can be opaque or transparent, and optionally can contain solvents, emulsifiers, moisturizers, emollients, fragrances, dyes/colorants, preservatives and other active ingredients that increase or enhance the efficacy of the final product.
  • Creams and lotions are often similar to one another, differing mainly in their viscosity; both lotions and creams may be opaque, translucent or clear and often contain emulsifiers, solvents, and viscosity adjusting agents, as well as moisturizers, emollients, fragrances, dyes/colorants, preservatives and other active ingredients that increase or enhance the efficacy of the final product.
  • Gels can be prepared with a range of viscosities, from thick or high viscosity to thin or low viscosity.
  • These formulations may also contain solvents, emulsifiers, moisturizers, emollients, fragrances, dyes/colorants, preservatives and other active ingredients that increase or enhance the efficacy of the final product.
  • Liquids are thinner than creams, lotions, or gels and often do not contain emulsifiers.
  • Liquid topical products often contain solvents, emulsifiers, moisturizers, emollients, fragrances, dyes/colorants, preservatives and other active ingredients that increase or enhance the efficacy of the final product.
  • Suitable emulsifiers for use in topical formulations include, but are not limited to, ionic emulsifiers, cetearyl alcohol, non-ionic emulsifiers like polyoxyethylene oleyl ether, PEG-40 stearate, ceteareth-12, ceteareth-20, ceteareth-30, ceteareth alcohol, PEG-100 stearate and glyceryl stearate.
  • Suitable viscosity adjusting agents include, but are not limited to, protective colloids or non-ionic gums such as hydroxyethylcellulose, xanthan gum, magnesium aluminum silicate, silica, microcrystalline wax, beeswax, paraffin, and cetyl palmitate.
  • a gel composition may be formed by the addition of a gelling agent such as chitosan, methyl cellulose, ethyl cellulose, polyvinyl alcohol, polyquaterniums, hydroxyethylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose, carbomer or ammoniated glycyrrhizinate.
  • a gelling agent such as chitosan, methyl cellulose, ethyl cellulose, polyvinyl alcohol, polyquaterniums, hydroxyethylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose, carbomer or ammoniated glycyrrhizinate.
  • Suitable surfactants include, but are not limited to, nonionic, amphoteric, ionic and anionic surfactants.
  • dimethicone copolyol polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 80, lauramide DEA, cocamide DEA, and cocamide MEA, oleyl betaine, cocamidopropyl phosphatidyl PG-dimonium chloride, and ammonium laureth sulfate may be used within topical formulations.
  • Suitable preservatives include, but are not limited to, antimicrobials such as methylparaben, propylparaben, sorbic acid, benzoic acid, and formaldehyde, as well as physical stabilizers and antioxidants such as vitamin E, sodium ascorbate/ascorbic acid and propyl gallate.
  • Suitable moisturizers include, but are not limited to, lactic acid and other hydroxy acids and their salts, glycerin, propylene glycol, and butylene glycol.
  • Suitable emollients include lanolin alcohol, lanolin, lanolin derivatives, cholesterol, petrolatum, isostearyl neopentanoate and mineral oils.
  • Suitable fragrances and colors include, but are not limited to, FD&C Red No. 40 and FD&C Yellow No. 5.
  • Suitable additional ingredients include, but are not limited to, abrasives, absorbents, anti-caking agents, anti-foaming agents, anti-static agents, astringents (e.g., witch hazel, alcohol and herbal extracts such as chamomile extract), binders/excipients, buffering agents, chelating agents, film forming agents, conditioning agents, propellants, opacifying agents, pH adjusters and protectants.
  • An example of a suitable topical vehicle for formulation of a gel is: hydroxypropylcellulose (2.1%); 70/30 isopropyl alcohol/water (90.9%); propylene glycol (5.1%); and Polysorbate 80 (1.9%).
  • An example of a suitable topical vehicle for formulation as a foam is: cetyl alcohol (1.1%); stearyl alcohol (0.5%; Quaternium 52 (1.0%); propylene glycol (2.0%); Ethanol 95 PGF3 (61.05%); deionized water (30.05%); P75 hydrocarbon propellant (4.30%). All percents are by weight.
  • Typical modes of delivery for topical compositions include application using the fingers; application using a physical applicator such as a cloth, tissue, swab, stick or brush; spraying (including mist, aerosol or foam spraying); dropper application; sprinkling; soaking; and rinsing.
  • a pharmaceutical composition may be prepared as a sterile injectible aqueous or oleaginous suspension.
  • the compound(s) provided herein, depending on the vehicle and concentration used, can either be suspended or dissolved in the vehicle.
  • Such a composition may be formulated according to the known art using suitable dispersing, wetting agents and/or suspending agents such as those mentioned above.
  • suitable dispersing, wetting agents and/or suspending agents such as those mentioned above.
  • the acceptable vehicles and solvents that may be employed are water, 1,3-butanediol, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils may be 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 injectible compositions, and adjuvants such as local anesthetics, preservatives and/or buffering agents can be dissolved in the vehicle.
  • compositions may also be formulated as suppositories (e.g., for rectal administration).
  • Such 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 excipients include, for example, cocoa butter and polyethylene glycols.
  • compositions for inhalation typically can be provided in the form of a solution, suspension or emulsion that can be administered as a dry powder or in the form of an aerosol using a conventional propellant (e.g., dichlorodifluoromethane or trichlorofluoromethane).
  • a conventional propellant e.g., dichlorodifluoromethane or trichlorofluoromethane.
  • compositions may be formulated for release at a pre-determined rate.
  • Instantaneous release may be achieved, for example, via sublingual administration (i.e., administration by mouth in such a way that the active ingredient(s) are rapidly absorbed via the blood vessels under the tongue rather than via the digestive tract).
  • Controlled release formulations i.e., formulations such as a capsule, tablet or coated tablet that slows and/or delays release of active ingredient(s) following administration
  • a controlled release formulation comprises a matrix and/or coating that delays disintegration and absorption in the gastrointestinal tract (or implantation site) and thereby provides a delayed action or a sustained action over a longer period.
  • One type of controlled-release formulation is a sustained-release formulation, in which at least one active ingredient is continuously released over a period of time at a constant rate.
  • the therapeutic agent is released at such a rate that blood (e.g., plasma) concentrations are maintained within the therapeutic range, but below toxic levels, over a period of time that is at least 4 hours, preferably at least 8 hours, and more preferably at least 12 hours.
  • Such formulations may generally be prepared using well known technology and administered by, for example, oral, rectal or subcutaneous implantation, or by implantation at the desired target site.
  • Carriers for use within such formulations are biocompatible, and may also be biodegradable; preferably the formulation provides a relatively constant level of modulator release.
  • the amount of modulator contained within a sustained release formulation depends upon, for example, the site of implantation, the rate and expected duration of release and the nature of the condition to be treated or prevented.
  • Controlled release may be achieved by combining the active ingredient(s) with a matrix material that itself alters release rate and/or through the use of a controlled-release coating.
  • the release rate can be varied using methods well known in the art, including (a) varying the thickness or composition of coating, (b) altering the amount or manner of addition of plasticizer in a coating, (c) including additional ingredients, such as release-modifying agents, (d) altering the composition, particle size or particle shape of the matrix, and (e) providing one or more passageways through the coating.
  • the amount of modulator contained within a sustained release formulation depends upon, for example, the method of administration (e.g., the site of implantation), the rate and expected duration of release and the nature of the condition to be treated or prevented.
  • the matrix material which itself may or may not serve a controlled-release function, is generally any material that supports the active ingredient(s).
  • a time delay material such as glyceryl monosterate or glyceryl distearate may be employed.
  • Active ingredient(s) may be combined with matrix material prior to formation of the dosage form (e.g., a tablet).
  • active ingredient(s) may be coated on the surface of a particle, granule, sphere, microsphere, bead or pellet that comprises the matrix material. Such coating may be achieved by conventional means, such as by dissolving the active ingredient(s) in water or other suitable solvent and spraying.
  • additional ingredients are added prior to coating (e.g., to assist binding of the active ingredient(s) to the matrix material or to color the solution).
  • the matrix may then be coated with a barrier agent prior to application of controlled-release coating. Multiple coated matrix units may, if desired, be encapsulated to generate the final dosage form.
  • a controlled release is achieved through the use of a controlled release coating (i.e., a coating that permits release of active ingredient(s) at a controlled rate in aqueous medium).
  • the controlled release coating should be a strong, continuous film that is smooth, capable of supporting pigments and other additives, non-toxic, inert and tack-free.
  • Coatings that regulate release of the modulator include pH-independent coatings, pH-dependent coatings (which may be used to release modulator in the stomach) and enteric coatings (which allow the formulation to pass intact through the stomach and into the small intestine, where the coating dissolves and the contents are absorbed by the body).
  • pH dependent coatings include, for example, shellac, cellulose acetate phthalate, polyvinyl acetate phthalate, hydroxypropylmethylcellulose phthalate, methacrylic acid ester copolymers and zein.
  • the coating is a hydrophobic material, preferably used in an amount effective to slow the hydration of the gelling agent following administration.
  • Suitable hydrophobic materials include alkyl celluloses (e.g., ethylcellulose or carboxymethylcellulose), cellulose ethers, cellulose esters, acrylic polymers (e.g., poly(acrylic acid), poly(methacrylic acid), acrylic acid and methacrylic acid copolymers, methyl methacrylate copolymers, ethoxy ethyl methacrylates, cyanoethyl methacrylate, methacrylic acid alkamide copolymer, poly(methyl methacrylate), polyacrylamide, ammonio methacrylate copolymers, aminoalkyl methacrylate copolymer, poly(methacrylic acid anhydride) and glycidyl methacrylate copolymers) and mixtures of the foregoing.
  • Representative aqueous dispersions of ethylcellulose include, for example, AQUACOAT® (FMC Corp., Philadelphia, Pa.) and SURELEASE® (Colorcon, Inc., West Point, Pa.), both of which can be applied to the substrate according to the manufacturer's instructions.
  • Representative acrylic polymers include, for example, the various EUDRAGIT® (Rohm America, Piscataway, N.J.) polymers, which may be used singly or in combination depending on the desired release profile, according to the manufacturer's instructions.
  • Suitable plasticizers for alkyl celluloses include, for example, dibutyl sebacate, diethyl phthalate, triethyl citrate, tributyl citrate and triacetin.
  • Suitable plasticizers for acrylic polymers include, for example, citric acid esters such as triethyl citrate and tributyl citrate, dibutyl phthalate, polyethylene glycols, propylene glycol, diethyl phthalate, castor oil and triacetin.
  • Controlled-release coatings are generally applied using conventional techniques, such as by spraying in the form of an aqueous dispersion.
  • the coating may comprise pores or channels or to facilitate release of active ingredient. Pores and channels may be generated by well known methods, including the addition of organic or inorganic material that is dissolved, extracted or leached from the coating in the environment of use.
  • pore-forming materials include hydrophilic polymers, such as hydroxyalkylcelluloses (e.g., hydroxypropylmethylcellulose), cellulose ethers, synthetic water-soluble polymers (e.g., polyvinylpyrrolidone, cross-linked polyvinylpyrrolidone and polyethylene oxide), water-soluble polydextrose, saccharides and polysaccharides and alkali metal salts.
  • a controlled release coating may include one or more orifices, which may be formed my methods such as those described in U.S. Pat. Nos. 3,845,770; 4,034,758; 4,077,407; 4,088,864; 4,783,337 and 5,071,607. Controlled-release may also be achieved through the use of transdermal patches, using conventional technology (see, e.g., U.S. Pat. No. 4,668,232).
  • controlled release formulations may be found, for example, in U.S. Pat. Nos. 4,572,833; 4,587,117; 4,606,909; 4,610,870; 4,684,516; 4,777,049; 4,994,276; 4,996,058; 5,128,143; 5,202,128; 5,376,384; 5,384,133; 5,445,829; 5,510,119; 5,618,560; 5,643,604; 5,891,474; 5,958,456; 6,039,980; 6,143,353; 6,126,969; 6,156,342; 6,197,347; 6,387,394; 6,399,096; 6,437,000; 6,447,796; 6,475,493; 6,491,950; 6,524,615; 6,838,094; 6,905,709; 6,923,984; 6,923,988; and 6,911,217; each of which is hereby incorporated by reference for its teaching of the preparation of
  • a compound provided herein may be conveniently added to food or drinking water (e.g., for administration to non-human animals including companion animals (such as dogs and cats) and livestock).
  • Animal feed and drinking water compositions may be formulated so that the animal takes in an appropriate quantity of the composition along with its diet. It may also be convenient to present the composition as a premix for addition to feed or drinking water.
  • Compounds are generally administered in a therapeutically effective amount.
  • Preferred systemic doses are no higher than 50 mg per kilogram of body weight per day (e.g., ranging from about 0.001 mg to about 50 mg per kilogram of body weight per day), with oral doses generally being about 5-20 fold higher than intravenous doses (e.g., ranging from 0.01 to 40 mg per kilogram of body weight per day).
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage unit will vary depending, for example, upon the patient being treated, the particular mode of administration and any other co-administered drugs. Dosage units generally contain between from about 10 ⁇ g to about 500 mg of active ingredient. Optimal dosages may be established using routine testing, and procedures that are well known in the art.
  • compositions may be packaged for treating conditions responsive to P2X 7 receptor modulation (e.g., pain, inflammation, neurodegeneration or other condition described herein).
  • Packaged pharmaceutical compositions generally include (i) a container holding a pharmaceutical composition that comprises at least one modulator as described herein and (ii) instructions (e.g., labeling or a package insert) indicating that the contained composition is to be used for treating a condition responsive to P2X 7 receptor modulation in the patient.
  • P2X 7 receptor modulators provided herein may be used to alter activity and/or activation of P2X 7 receptors in a variety of contexts, both in vitro and in vivo.
  • P2X 7 receptor antagonists may be used to inhibit the binding of ligand agonist to P2X 7 receptor in vitro or in vivo.
  • such methods comprise the step of contacting a P2X 7 receptor with one or more P2X 7 receptor modulators provided herein, in the presence of ligand in aqueous solution and under conditions otherwise suitable for binding of the ligand to P2X 7 receptor.
  • the modulator(s) are generally present at a concentration that is sufficient to alter P2X 7 receptor-mediated signal transduction (using an assay provided in Example 4).
  • the P2X 7 receptor may be present in solution or suspension (e.g., in an isolated membrane or cell preparation), or in a cultured or isolated cell.
  • the P2X 7 receptor is expressed by a cell that is present in a patient, and the aqueous solution is a body fluid.
  • one or more modulators are administered to an animal in an amount such that the modulator is present in at least one body fluid of the animal at a therapeutically effective concentration that is 20 micromolar or less, 10 micromolar or less, 5 micromolar or less, or 1 micromolar or less.
  • such compounds may be administered at a therapeutically effective dose that is less than 20 mg/kg body weight, preferably less than 5 mg/kg and, in some instances, less than 1 mg/kg.
  • modulation may be achieved by contacting a P2X 7 receptor (either in vitro or in vivo) with one or more modulators provided herein under conditions suitable for binding of the modulator(s) to the receptor.
  • the modulator(s) are generally present at a concentration that is sufficient to alter P2X 7 receptor-mediated signal transduction as described herein.
  • the receptor may be present in solution or suspension, in a cultured or isolated cell preparation or in a cell within a patient. For example, the cell may be contacted in vivo in an animal.
  • Modulation of signal tranducing activity may be assessed by detecting an effect on calcium ion conductance (also referred to as calcium mobilization or flux). Modulation of signal transducing activity may alternatively be assessed by detecting an alteration of a symptom (e.g., pain or inflammation) of a patient being treated with one or more modulators provided herein.
  • a symptom e.g., pain or inflammation
  • P2X 7 receptor modulator(s) provided herein are preferably administered to a patient (e.g., a human) orally or topically, and are present within at least one body fluid of the animal while modulating P2X 7 receptor signal-transducing activity.
  • the present invention further provides methods for treating conditions responsive to P2X 7 receptor modulation.
  • treatment encompasses both disease-modifying treatment and symptomatic treatment, either of which may be prophylactic (i.e., before the onset of symptoms, in order to prevent, delay or reduce the severity of symptoms) or therapeutic (i.e., after the onset of symptoms, in order to reduce the severity and/or duration of symptoms).
  • a condition is “responsive to P2X 7 receptor modulation” if it is characterized by inappropriate activity of a P2X 7 receptor, regardless of the amount of P2X 7 agonist present locally, and/or if modulation of P2X 7 receptor activity results in alleviation of the condition or a symptom thereof.
  • Such conditions include, for example, pain, inflammation, cardiovascular disorders, neurodegenerative disorders and respiratory disorders (such as cough, asthma, chronic obstructive pulmonary disease, chronic bronchitis, cystic fibrosis and rhinitis, including allergic rhinitis, such as seasonal an perennial rhinitis, and non-allergic rhinitis), fibrosis as well as other conditions described in more detail below.
  • Such conditions may be diagnosed and monitored using criteria that have been established in the art.
  • Patients may include humans, domesticated companion animals and livestock, with dosages as described above.
  • Treatment regimens may vary depending on the compound used and the particular condition to be treated; however, for treatment of most disorders, a frequency of administration of 4 times daily or less is preferred. In general, a dosage regimen of 2 times daily is more preferred, with once a day dosing particularly preferred. For the treatment of acute pain, a single dose that rapidly reaches effective concentrations is desirable. It will be understood, however, that the specific dose level and treatment regimen 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 severity of the particular disease undergoing therapy. In general, the use of the minimum dose sufficient to provide effective therapy is preferred. Patients may generally be monitored for therapeutic effectiveness using medical or veterinary criteria suitable for the condition being treated or prevented.
  • Pain that may be treated using the modulators provided herein includes, for example, acute, chronic, inflammatory, and neuropathic pain.
  • Specific pain indications that may be treated as described herein include, but are not limited to, pain associated with osteoarthritis or rheumatoid arthritis; various neuropathic pain syndromes (such as post-herpetic neuralgia, trigeminal neuralgia, reflex sympathetic dystrophy, diabetic neuropathy, Guillian Bane syndrome, fibromyalgia, oral neuropathic pain, phantom limb pain, post-mastectomy pain, peripheral neuropathy, myofascial pain syndromes, MS-related neuropathy, HIV or AIDS-related neuropathy, and chemotherapy-induced and other iatrogenic neuropathies); visceral pain, (such as that associated with gastroesophageal reflux disease (GERD), irritable bowel syndrome, inflammatory bowel disease, pancreatitis, intestinal gas, gynecological disorders (e.g., menstrual pain
  • Further pain conditions that can be treated as described herein include Charcot's pains, ear pain, muscle pain, eye pain, orofacial pain (e.g., odontalgia), carpel tunnel syndrome, acute and chronic back pain (e.g., lower back pain), gout, scar pain, hemorrhoidal pain, dyspeptic pains, angina, nerve root pain, “non-painful” neuropathies, complex regional pain syndrome, homotopic pain and heterotopic pain—including pain associated with carcinoma, often referred to as cancer-associated pain (e.g., in patients with bone cancer), pain (and inflammation) associated with venom exposure (e.g., due to snake bite, spider bite, or insect sting) and trauma-associated pain (e.g., post-surgical pain, episiotomy pain, pain from cuts, musculoskeletal pain, bruises and broken bones, and burn pain, especially primary hyperalgesia associated therewith).
  • Additional pain conditions that may be treated as described herein include pain associated with autoimmune diseases or
  • Conditions associated with inflammation and/or immune system disorders include, but are not limited to, arthritis (including osteoarthritis, rheumatoid arthritis, psoriatic arthritis, Reiter's syndrome, gout, traumatic arthritis, rubella arthritis, rheumatoid spondylitis, gouty arthritis and juvenile arthritis); cystic fibrosis; uveitis; systemic lupus erythematosus (and associated glomerulonephritis); spondyloarthropathies; psoriasis; scleritis; allergic conditions (including allergic reactions, allergic rhinitis, allergic contact hypersensitivity, allergic dermatitis, eczema and contact dermatitis), reperfusion injury (e.g., cardiac and renal reperfusion injury), respiratory system disorders (including hyper-responsiveness of the airway, cough, asthma (e.g., to prevent or decrease the severity of both acute early phase asthma attack and the late phase
  • arthritis including osteoarthritis,
  • pathologic sequellae associated with insulin-dependent diabetes mellitus including diabetic retinopathy
  • lupus nephropathy including diabetic retinopathy
  • Heyman nephritis membranous nephritis and other forms of glomerulonephritis
  • macular degeneration e.g., contact sensitivity responses, and inflammation resulting from contact of blood with artificial surfaces as occurs, for example, during extracorporeal circulation of blood (e.g., during hemodialysis or via a heart-lung machine, for example, in association with vascular surgery such as coronary artery bypass grafting or heart valve replacement) such as extracorporeal post-dialysis syndrome, or in association with contact with other artificial vessel or container surfaces (e.g., ventricular assist devices, artificial heart machines, transfusion tubing, blood storage bags, plasmapheresis, plateletpheresis, and the like).
  • other artificial vessel or container surfaces e.g., ventricular assist devices, artificial heart machines, transfusion
  • modulators provided herein may be used within combination therapy for the treatment of conditions responsive to P2X 7 receptor modulation (e.g., conditions involving pain and/or inflammatory components).
  • conditions responsive to P2X 7 receptor modulation include, for example, autoimmune disorders and pathologic autoimmune responses known to have an inflammatory component including, but not limited to, arthritis (especially rheumatoid arthritis), psoriasis, Crohn's disease, lupus erythematosus, irritable bowel syndrome, tissue graft rejection, and hyperacute rejection of transplanted organs.
  • Other such conditions include trauma (e.g., injury to the head or spinal cord), cardio- and cerebro-vascular disease and certain infectious diseases.
  • a modulator is administered to a patient along with a second therapeutic agent (e.g., an analgesic and/or anti-inflammatory agent).
  • a second therapeutic agent e.g., an analgesic and/or anti-inflammatory agent.
  • the modulator and second therapeutic agent may be present in the same pharmaceutical composition, or may be administered separately in either order.
  • Anti-inflammatory agents include, for example, non-steroidal anti-inflammatory drugs (NSAIDs), non-specific and cyclooxygenase-2 (COX-2) specific cyclooxygenase enzyme inhibitors, gold compounds, corticosteroids, methotrexate, leflunomide, cyclosporine A, IM gold, minocycline, azathioprine, tumor necrosis factor (TNF) receptor antagonists, soluble TNF alpha receptor (etanercept), anti-TNF alpha antibodies (e.g., infliximab and adalimumab), anti-05 antibodies, interleukin-1 (IL-1) receptor antagonists (e.g., anakinra or IL-1 trap), IL-18 binding protein, CTLA4-Ig (e.g., abatacept), anti-human IL-6 receptor monoclonal antibody (e.g., tocilizumab), LFA-3-Ig fusion proteins (e.g., alefacept), L
  • NSAIDs include, but are not limited to, ibuprofen, flurbiprofen, naproxen or naproxen sodium, diclofenac, combinations of diclofenac sodium and misoprostol, sulindac, oxaprozin, diflunisal, piroxicam, indomethacin, etodolac, fenoprofen calcium, ketoprofen, sodium nabumetone, sulfasalazine, tolmetin sodium, and hydroxychloroquine.
  • COX cyclooxygenase
  • NSAIDs further include salicylates such as acetylsalicylic acid or aspirin, sodium salicylate, choline and magnesium salicylates, and salsalate, as well as corticosteroids such as cortisone, dexamethasone, methylprednisolone, prednisolone, prednisolone sodium phosphate, and prednisone.
  • salicylates such as acetylsalicylic acid or aspirin
  • sodium salicylate sodium salicylate
  • choline and magnesium salicylates sodium salicylate
  • salsalate sodium salicylate
  • corticosteroids such as cortisone, dexamethasone, methylprednisolone, prednisolone, prednisolone sodium phosphate, and prednisone.
  • Suitable dosages for P2X 7 receptor modulator within such combination therapy are generally as described above. Dosages and methods of administration of anti-inflammatory agents can be found, for example, in the manufacturer's instructions in the Physician's Desk Reference.
  • the combination administration of a modulator with an anti-inflammatory agent results in a reduction of the dosage of the anti-inflammatory agent required to produce a therapeutic effect (i.e., a decrease in the minimum therapeutically effective amount).
  • the dosage of anti-inflammatory agent in a combination or combination treatment method is less than the maximum dose advised by the manufacturer for administration of the anti-inflammatory agent without combination administration of a modulator.
  • this dosage is less than 3 ⁇ 4, even more preferably less than 1 ⁇ 2, and highly preferably, less than 1 ⁇ 4 of the maximum dose, while most preferably the dose is less than 10% of the maximum dose advised by the manufacturer for administration of the anti-inflammatory agent(s) when administered without combination administration of a modulator. It will be apparent that the dosage amount of modulator component of the combination needed to achieve the desired effect may similarly be reduced by the co-administraction of the anti-inflammatory agent.
  • the combination administration of a modulator with an anti-inflammatory agent is accomplished by packaging one or more modulators and one or more anti-inflammatory agents in the same package, either in separate containers within the package or in the same contained as a mixture of one or more modulators and one or more anti-inflammatory agents.
  • Preferred mixtures are formulated for oral administration (e.g., as pills, capsules, tablets or the like).
  • the package comprises a label bearing indicia indicating that the one or more modulators and one or more anti-inflammatory agents are to be taken together for the treatment of an inflammatory pain condition.
  • modulators provided herein may be used in combination with one or more additional pain relief medications.
  • Certain such medications are also anti-inflammatory agents, and are listed above.
  • Other such medications are analgesic agents, including narcotic agents which typically act at one or more opioid receptor subtypes (e.g., ⁇ , ⁇ and/or ⁇ ), preferably as agonists or partial agonists.
  • opioid receptor subtypes e.g., ⁇ , ⁇ and/or ⁇
  • Such agents include opiates, opiate derivatives and opioids, as well as pharmaceutically acceptable salts and hydrates thereof.
  • narcotic analgesics include, within preferred embodiments, alfentanil, alphaprodine, anileridine, bezitramide, buprenorphine, butorphanol, codeine, diacetyldihydromorphine, diacetylmorphine, dihydrocodeine, diphenoxylate, ethylmorphine, fentanyl, heroin, hydrocodone, hydromorphone, isomethadone, levomethorphan, levorphane, levorphanol, meperidine, metazocine, methadone, methorphan, metopon, morphine, nalbuphine, opium extracts, opium fluid extracts, powdered opium, granulated opium, raw opium, tincture of opium, oxycodone, oxymorphone, paregoric, pentazocine, pethidine, phenazocine, piminodine, propoxyphene, racemethorphan
  • narcotic analgesic agents include acetorphine, acetyldihydrocodeine, acetylmethadol, allylprodine, alphracetylmethadol, alphameprodine, alphamethadol, benzethidine, benzylmorphine, betacetylmethadol, betameprodine, betamethadol, betaprodine, clonitazene, codeine methylbromide, codeine-N-oxide, cyprenorphine, desomorphine, dextromoramide, diampromide, diethylthiambutene, dihydromorphine, dimenoxadol, dimepheptanol, dimethylthiamubutene, dioxaphetyl butyrate, dipipanone, drotebanol, ethanol, ethylmethylthiambutene, etonitazene, e
  • analgesic agents include, for example acetaminophen (paracetamol); aspirin and other NSAIDs described above; NR2B antagonists; bradykinin antagonists; anti-migraine agents; anticonvulsants such as oxcarbazepine and carbamazepine; antidepressants (such as TCAs, SSRIs, SNRIs, substance P antagonists, etc.); spinal blocks; pentazocine/naloxone; meperidine; levorphanol; buprenorphine; hydromorphone; fentanyl; sufentanyl; oxycodone; oxycodone/acetaminophen, nalbuphine and oxymorphone.
  • analgesic agents include CB2-receptor agonists, such as AM1241, capsaicin receptor antagonists and compounds that bind to the ⁇ 2 ⁇ subunit of voltage-gated calcium channels, such as gabapentin and pregabalin.
  • Representative anti-migraine agents for use in combination with a modulator provided herein include CGRP antagonists, capsaicin receptor antagonists, ergotamines and 5-HT 1 agonists, such as sumatripan, naratriptan, zolmatriptan and rizatriptan.
  • modulators provided herein may be used, for example, in the treatment of pulmonary disorders such as asthma, in combination with one or more beta(2)-adrenergic receptor agonists or leukotriene receptor antagonists (e.g., agents that inhibits the cysteinyl leukotriene CysLT 1 receptor).
  • CysLT 1 antagonists include montelukast, zafirlukast, and pranlukast.
  • Suitable dosages for P2X 7 receptor modulator within such combination therapy are generally as described above. Dosages and methods of administration of other pain relief medications can be found, for example, in the manufacturer's instructions in the Physician's Desk Reference. In certain embodiments, the combination administration of a modulator with one or more additional pain medications results in a reduction of the dosage of each therapeutic agent required to produce a therapeutic effect (e.g., the dosage or one or both agent may less than 3 ⁇ 4, less than 1 ⁇ 2, less than 1 ⁇ 4 or less than 10% of the maximum dose listed above or advised by the manufacturer).
  • compositions as described above may further comprise one or more additional medications as described above.
  • the additional medication is an analgesic.
  • packaged pharmaceutical preparations comprising one or more modulators and one or more additional medications (e.g., analgesics) in the same package.
  • Such packaged pharmaceutical preparations generally include (i) a container holding a pharmaceutical composition that comprises at least one modulator as described herein; (ii) a container holding a pharmaceutical composition that comprises at least one additional medication (such as a pain relief and/or anti-inflammatory medication) as described above and (iii) instructions (e.g., labeling or a package insert) indicating that the compositions are to be used simultaneously, separately or sequentially for treating or preventing a condition responsive to P2X 7 receptor modulation in the patient (such as a condition in which pain and/or inflammation predominates).
  • a condition responsive to P2X 7 receptor modulation such as a condition in which pain and/or inflammation predominates.
  • the present invention provides a variety of non-pharmaceutical in vitro and in vivo uses for the modulator compounds provided herein.
  • such compounds may be labeled and used as probes for the detection and localization of P2X 7 receptor (in samples such as cell preparations or tissue sections, preparations or fractions thereof).
  • modulators provided herein that comprise a suitable reactive group such as an aryl carbonyl, nitro or azide group
  • modulators provided herein may be used in photoaffinity labeling studies of receptor binding sites.
  • modulators provided herein may be used as positive controls in assays for receptor activity or as radiotracers (e.g., in receptor mapping procedures).
  • a modulator compound may be labeled using any of a variety of well known techniques (e.g., radiolabeled with a radionuclide such as tritium, as described herein), and used as a probe for receptor autoradiography (receptor mapping) of P2X 7 receptor in cultured cells or tissue samples, which may be performed as described by Kuhar in sections 8.1.1 to 8.1.9 of Current Protocols in Pharmacology (1998) John Wiley & Sons, New York, which sections are incorporated herein by reference.
  • receptor mapping procedures also include methods that can be used to characterize P2X 7 receptor in living subjects, such as positron emission tomography (PET) imaging or single photon emission computerized tomography (SPECT).
  • PET positron emission tomography
  • SPECT single photon emission computerized tomography
  • Mass spectroscopy data provided herein is Electrospray MS, obtained in positive ion mode. Unless otherwise specified, such data is obtained using a Micromass Time-of-Flight LCT (Waters Corp.; Milford, Mass.), equipped with a Waters 600 pump (Waters Corp.), Waters 996 photodiode array detector (Waters Corp.), and a Gilson 215 autosampler (Gilson, Inc.; Middleton, Wis.). MassLynxTM (Waters Corp.) version 4.0 software with OpenLynx Global ServerTM, OpenLynxTM and AutoLynxTM processing is used for data collection and analysis.
  • Micromass Time-of-Flight LCT Waters Corp.; Milford, Mass.
  • Waters 600 pump Waters Corp.
  • Waters 996 photodiode array detector Waters Corp.
  • Gilson 215 autosampler Gilson, Inc.; Middleton, Wis.
  • Sample volume of 1 microliter is injected onto a 50 ⁇ 4 6 mm Chromolith SpeedROD RP-18e column (Merck KGaA, Darmstadt, Germany), and eluted using a 2-phase linear gradient at a flow rate of 6 mL/min. Sample is detected using total absorbance count over the 220-340 nm UV range.
  • the elution conditions are: Mobile Phase A-95% water, 5% MeOH with 0.05% TFA; Mobile Phase B-5% water, 95% MeOH with 0.025% TFA.
  • the following gradient is used: 0-0.5 min 10-100% B, hold at 100% B to 1.2 min, return to 10% B at 1.21 min Inject to inject cycle is 2.15 min.
  • retention times are provided in minutes.
  • Example 4 illustrates the preparation of representative 5-membered heterocyclic amides and related compounds, as well as certain intermediates useful in the preparation of such compounds.
  • Mass spectroscopy characterization data is obtained as described above.
  • a “IC 50 ” after the mass spectroscopy data provided for a compound indicates that the IC 50 determined as described in Example 4A is 2 micromolar or less (i.e., the concentration of the compound that is required to provide a 50% decrease in the fluorescence response of cells exposed to 80 ⁇ M of (2′(3′)—O-(4-benzoyl-benzoyl)adenosine 5′-triphosephate is 2 micromolar or less).
  • BOP (18 mg, 0.04 mmol) is added to a solution of 1-(4-amino-5-fluoro-pyrimidin-2-yl)-3-trifluoromethyl-1H-pyrazole-4-carboxylic acid (10 mg, 0.03 mmol), (1-pyridin-3-yl-cyclohexyl)-methylamine (8 mg, 0.04 mmol), HOBt (2 mg, 0.01 mmol) and DIEA (1.7 ⁇ L, 0.1 mmol) in DMF (0.2 mL) at rt. The mixture is stirred for 4 h. H 2 O is added. The mixture is extracted with 5% MeOH in DCM and dried over Na 2 SO 4 .
  • Acetic anhydride (1.2 mL, 12.8 mmol) is added to a suspension of ethyl 3-hydroxy-1H-pyrazole-4-carboxylate (2.0 g, 12.8 mmol) in 30 mL of acetic acid.
  • the reaction mixture is stirred at RT for 2 h.
  • the solvent is removed in vacuo, and the remaining solid is washed with water, collected by vacuum filtration, and dried in vacuo to give the title compound as a white solid.
  • 1 H NMR 400 MHz, CDCl 3 ) ⁇ 8.45 (1H, s), 8.27 (1H, bs), 4.38 (2H, q), 2.64 (3H, s), 1.38 (3H, t).
  • a 1 N aqueous sodium hydroxide solution (10 mL) is added to a slurry of ethyl 3-methoxy-1-pyrimidin-2-yl-1H-pyrazole-4-carboxylate (1.1 mg, 4.43 mmol) in 30 mL of EtOH.
  • the reaction mixture becomes homogeneous and is stirred for 2 h at RT.
  • Most of the EtOH is removed in vacuo, and the resultant mixture is filtered. The collected solid is dried in vacuo to give the title compound as a white solid.
  • Potassium tert-butoxide (2.4 g, 21.23 mmol) is added to a mixture of ethyl 3-amino-1H-pyrazole-4-carboxylate (3.0 g, 19.3 mmol) and 2-chloropyrimidine (2.2 g, 19.3 mmol) in 50 mL of anhydrous dioxane.
  • the reaction mixture is heated to 100° C. and stirred for 2 h.
  • the solvent is removed in vacuo, and the residue is partitioned between EtOAc and water (50 mL each).
  • a 1 N aqueous sodium hydroxide solution (3.0 mL) is added to a suspension of ethyl 3-amino-1-pyrimidin-2-yl-1H-pyrazole-4-carboxylate (500 mg, 2.14 mmol) in 6.0 mL of EtOH.
  • the reaction mixture is heated to 50° C. for 16 h. After cooling to RT, water is added, and the mixture is extracted with EtOAc (3 ⁇ 20 mL).
  • the precipitated solid is collected by vacuum filtration and dried in vacuo to give the title compound as an off-white solid.
  • Step 1 Ethyl 3-chloro-1-pyrimidin-2-yl-1H-pyrazole-4-carboxylate and ethyl 5-chloro-1-pyrimidin-2-yl-1H-pyrazole-4-carboxylate
  • a solution of sodium nitrite (1.0 g, 15.1 mmol) in 5.0 mL of water is added dropwise to a solution of ethyl 3-amino-1-pyrimidin-2-yl-1H-pyrazole-4-carboxylate (2.7 g, 11.6 mmol) in 18 mL of conc. HCl at ⁇ 5° C. with stirring. After 1 h at 0° C., the reaction mixture is added dropwise to a suspension of copper (I) chloride (1.8 g, 18.6 mmol) in 18 mL of CHCl 3 at RT. After 1 h at RT, water (40 mL) and CHCl 3 (40 mL) are added, and the layers are separated.
  • a 1 N aqueous sodium hydroxide solution (5.0 mL) is added to a suspension of ethyl 3-chloro-1-pyrimidin-2-yl-1H-pyrazole-4-carboxylate (850 mg, 3.36 mmol) in 10.0 mL of EtOH.
  • the reaction mixture is stirred at RT for 1 h.
  • the precipitated solid is collected by vacuum filtration and dried in vacuo to give the title compound as a white solid.
  • a 1 N aqueous sodium hydroxide solution (2.0 mL) is added to a suspension of ethyl 5-chloro-1-pyrimidin-2-yl-1H-pyrazole-4-carboxylate (250 mg, 1.11 mmol) in 5.0 mL of EtOH.
  • the reaction mixture is stirred at RT for 1 h. Water is added, and the solution is washed with EtOAc.
  • the precipitated solid is collected by vacuum filtration and dried in vacuo to give the title compound as a white solid.
  • HBr (48% in H 2 O) is added to a solution of 2-amino-3-chloropyrazine (1.2 g, 9.26 mmol) and BrCH 2 CH(OEt) 2 (1.6 mL, 10 mmol) in MeOH and H 2 O (5 mL and 10 mL) at RT.
  • the mixture is stirred for 24 h at RT and then heated to 40° C. for 48 h.
  • the pH is adjusted to 7 with sat. Na 2 CO 3 .
  • the mixture is extracted with CH 2 Cl 2 .
  • the organic phase is dried over anhydrous Na 2 SO 4 .
  • the product is purified by silica gel column chromatography (4% MeOH in CH 2 Cl 2 ) to give the title compound as an off white solid.
  • *IC 50 for N-(adamantan-1-ylmethyl)-1-benzyl-1H-pyrazole-5-carboxamide).
  • This Example illustrates the synthesis of additional representative 5-membered heterocyclic amides and related compounds provided herein, as well as certain intermediates useful in the preparation of such compounds.
  • This compound is prepared essentially as described for 2-pyrimidin-2-yl-4-trifluoromethyl-thiazole-5-carboxylic acid, with readily apparent starting material modification.
  • This compound is prepared essentially as described for 2-pyrimidin-2-yl-4-trifluoromethyl-thiazole-5-carboxylic acid, with readily apparent starting material modification.
  • Step 1 4-(1-cyano-cycloheptyl)-piperazine-1-carboxylic acid tert-butyl ester
  • the amine (0.2 M in toluene; 0.10 mL) and acid (0.2 M in DMA; 0.12 mL) are added to a vial along with DMC 3 (0.2 M in ACN, freshly prepared; 0.2 mL) and TEA (0.3 M in toluene; 0.10 mL) are added to a vial and incubated at RT for 16 h.
  • the reaction mixture is then extracted with 1 N NaOH (0.5 mL) and EtOAc (0.5 mL).
  • the upper organic layer is removed and concentrated to dryness.
  • the residue is purified via solid phase extraction chromatography eluting with 25% MeOH/EtOAc (4.0 mL) to afford the title compound.
  • This Example illustrates representative calcium mobilization assays for use in evaluating test compounds for agonist and antagonist activity.
  • SH-SY5Y cells ATCC Number CRL-2266, (American Type Culture Collection, Manassas, Va.) are cultured under DMEM/High medium supplemented with 10% FBS, and 10 mM HEPES (Invitrogen Corp., Carlsbad, Calif.) in 5% CO 2 and at 37° C.
  • DMEM/High medium supplemented with 10% FBS, and 10 mM HEPES (Invitrogen Corp., Carlsbad, Calif.) in 5% CO 2 and at 37° C.
  • cells are plated at a density of 100,000 cells/well in a 96 well black/clear TC plate (Corning® Costar®, Sigma-Aldrich Co., St. Louis, Mo.).
  • the culture medium is removed and cells are incubated with 50 ⁇ L of 2.3 ⁇ M Fluo-4 AM dye (Invitrogen Corp.) in the assay solution (5 mM KCl, 9.6 mM NaH 2 PO 4 H 2 O, 25 mM HEPES, 280 mM sucrose, 5 mM glucose, and 0.5 mM CaCl 2 ; pH is adjusted to 7.4 with NaOH) for an hour at 37° C.
  • wells are rinsed once with 50 ⁇ L assay solution, and are then incubated for an hour at room temperature with 100 ⁇ L assay solution containing the test compound.
  • test compound generally ranges from 1 to 2500 nM; for positive control cells, no test compound is added. After the one hour incubation, plates are transferred to a FLIPR TETRA instrument (Molecular Devices, Sunnyvale, Calif.) for calcium mobilization analysis.
  • the data is analyzed as follows. First, the average maximum relative fluorescent unit (RFU) response from the negative control wells (no agonist) is subtracted from the maximum response detected for each of the other experimental wells. Second, average maximum RFU response is calculated for the positive control wells (agonist wells). Then, percent inhibition for each compound tested is calculated using the equation:
  • the % inhibition data is plotted as a function of test compound concentration and test compound IC 50 is determined using a linear regression in which x is 1n(concentration of test compound) and y is 1n(percent inhibition/(100 ⁇ percent inhibition). Data with a percent inhibition that is greater than 90% or less than 15% are rejected and are not used in the regression.
  • the IC 50 is e (-intercept/slope) .
  • a sigmoidal fit can be used, such as that achieved using KALEIDAGRAPH software (Synergy Software, Reading, Pa.), which determines the best fit of the data to the equation:
  • m 0 is the concentration of the agonist
  • m 1 is the maximum RFU
  • m 2 corresponds to the test compound IC 50 (the concentration required to provide a 50% decrease, relative to the response observed in the presence of agonist and without antagonist)
  • m 3 is the Hill coefficient.
  • the calculated IC 50 is preferably below 20 micromolar, more preferably below 10 micromolar, even more preferably below 5 micromolar and most preferably below 1 micromolar.
  • test compounds Similar assays are performed in the absence of added agonist for the determination of agonist activity of the test compounds.
  • the ability of a test compound to act as an agonist of P2X 7 receptor is determined by measuring the fluorescence response elicited by the test compound as a function of compound concentration.
  • P2X 7 receptor antagonists that exhibit no detectable agonist activity elicit no detectable fluorescence response at a concentration of 2,500 nM.
  • SH-SY5Y cells are cultured under DMEM/High medium supplemented with 10% FBS, and 10 mM HEPES (Invitrogen Corp., Carlsbad, Calif.) in 5% CO 2 and at 37° C., and are split onto 12 mm round Poly-D-Lysine (PDL) coated coverslips (BD Biosciences, San Jose, Calif.) in a 35 mm dish with a density of 130K cells/dish a day prior to the experiment.
  • Whole cell voltage clamp recordings are made with the Axopatch-200B amplifier (Axon Instruments, Foster City, Calif.).
  • the recording electrodes are pulled from borosilicate pipettes (World Precision Instruments, Sarasota, Fla.) on a horizontal puller (Sutter Instrument Model P-87) and have resistances ranging from 2 to 3 M ⁇ when backfilled with internal solution. All voltage protocols are generated using pClamp 8 (Axon Instruments) software. Data are digitized at 1 or 5 kHz and recorded onto a PC for further analysis. Data are analyzed using Clampfit (Axon Instruments), Excel (Microsoft, Redmond, Wash.), and Origin software (MicroCal, LLC; Northampton, Mass.). All whole-cell recordings are conducted at room temperature.
  • P2X 7 receptor is activated by 200 ⁇ M of P2X 7 agonist, BzATP. At a holding potential of ⁇ 80 mV, the activated inward current is recorded in the presence and absence of the test compound. Then, percent inhibition for each compound tested is calculated using the equation:
  • C is the concentration of the antagonist
  • N is the Hill coefficient
  • IC 50 represents the compound IC 50 value against P2X 7 receptors.
  • This Example illustrates a representative method for assessing the degree of pain relief provided by a test compound.
  • Animals are then subjected to the first baseline, second baseline and testing on consecutive days. For each baseline, the animal is restrained as in the habituation session and the paw tested using the paw pressure testing apparatus (Digital Randall Selitto, IITC Inc., Woodland Hills, Calif.). Animals are baselined and tested in groups of ten, each animal being tested once on the left and right hindpaws, followed by the next consecutive animal. This procedure is repeated three times for a total of three measurements on each hindpaw. If any individual read is drastically different (varies by more than about 100 g) from the other two on a given hindpaw, the hindpaw is retested a 4 th time, and the average of the three most consistent scores is used.
  • the paw pressure testing apparatus Digital Randall Selitto, IITC Inc., Woodland Hills, Calif.
  • Test compounds or vehicle may be administered by various routes at various timepoints prior to testing, but for any particular assay, the routes and timepoints are the same for animals in each treatment group administered test compound (a different dosage of test compound may be administered to each such group) and those in the treatment group administered vehicle control. If a compound is orally administered, the animals are food-deprived the evening before testing. As with the baseline, each hindpaw is tested three times and the results recorded for analysis.
  • Hypersensitivity of nociception values are calculated for each treatment group as the mean of the left foot gram force scores on test day (left foot only or LFO score). Statistical significance between treatment groups is determined by running an ANOVA on LFO scores followed with a least significant difference (LSD) post hoc test. A p ⁇ 0.05 is considered to be a statistically significant difference.
  • Compounds are said to relieve pain in this model if they result in a statistically significant reduction in hypersensitivity of nociception values compared to vehicle controls, determined as described above, when administered (0.01-50 mg/kg, orally, parenterally or topically) immediately prior to testing as a single bolus, or for several days: once or twice or three times daily prior to testing.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Pulmonology (AREA)
  • Pain & Pain Management (AREA)
  • Ophthalmology & Optometry (AREA)
  • Immunology (AREA)
  • Rheumatology (AREA)
  • Dermatology (AREA)
  • Psychiatry (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Hospice & Palliative Care (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Heterocyclic Compounds Containing Sulfur Atoms (AREA)
  • Thiazole And Isothizaole Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Medicinal Preparation (AREA)
  • Hydrogenated Pyridines (AREA)
US12/669,308 2007-07-19 2008-07-21 5-Membered Heterocyclic Amides And Related Compounds Abandoned US20100292236A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/669,308 US20100292236A1 (en) 2007-07-19 2008-07-21 5-Membered Heterocyclic Amides And Related Compounds

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US95062507P 2007-07-19 2007-07-19
US12/669,308 US20100292236A1 (en) 2007-07-19 2008-07-21 5-Membered Heterocyclic Amides And Related Compounds
PCT/US2008/070613 WO2009012482A2 (en) 2007-07-19 2008-07-21 5-membered heterocyclic amides and related compounds

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/070613 A-371-Of-International WO2009012482A2 (en) 2007-07-19 2008-07-21 5-membered heterocyclic amides and related compounds

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/480,979 Continuation US9133204B2 (en) 2007-07-19 2014-09-09 5-membered heterocyclic amides and related compounds

Publications (1)

Publication Number Publication Date
US20100292236A1 true US20100292236A1 (en) 2010-11-18

Family

ID=40260407

Family Applications (2)

Application Number Title Priority Date Filing Date
US12/669,308 Abandoned US20100292236A1 (en) 2007-07-19 2008-07-21 5-Membered Heterocyclic Amides And Related Compounds
US14/480,979 Expired - Fee Related US9133204B2 (en) 2007-07-19 2014-09-09 5-membered heterocyclic amides and related compounds

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/480,979 Expired - Fee Related US9133204B2 (en) 2007-07-19 2014-09-09 5-membered heterocyclic amides and related compounds

Country Status (29)

Country Link
US (2) US20100292236A1 (zh)
EP (1) EP2178865B1 (zh)
JP (2) JP2010533736A (zh)
KR (2) KR101593237B1 (zh)
CN (2) CN101711245B (zh)
AU (1) AU2008275891B2 (zh)
BR (1) BRPI0813426A2 (zh)
CA (1) CA2691507C (zh)
CO (1) CO6251318A2 (zh)
CY (1) CY1116809T1 (zh)
DK (1) DK2178865T3 (zh)
EA (1) EA017250B1 (zh)
ES (1) ES2551095T3 (zh)
HK (2) HK1142330A1 (zh)
HR (1) HRP20151052T1 (zh)
HU (1) HUE026446T2 (zh)
IL (1) IL202499A (zh)
MX (1) MX2009014159A (zh)
MY (1) MY154668A (zh)
NZ (1) NZ582057A (zh)
PH (1) PH12014501677A1 (zh)
PL (1) PL2178865T3 (zh)
PT (1) PT2178865E (zh)
RS (1) RS54303B1 (zh)
SG (1) SG187396A1 (zh)
SI (1) SI2178865T1 (zh)
UA (1) UA105758C2 (zh)
WO (1) WO2009012482A2 (zh)
ZA (1) ZA200908447B (zh)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8686018B2 (en) 2010-09-21 2014-04-01 Eisai R&D Management Co., Ltd. Pharmaceutical composition
US9487508B2 (en) * 2012-09-19 2016-11-08 Ironwood Pharmaceuticals, Inc. SGC stimulators
US9725414B2 (en) 2011-11-30 2017-08-08 Bayer Intellectual Property Gmbh Fungicidal N-bicycloalkyl and N-tricycloalkyl pyrazole-4-(thio)carboxamide derivatives
US11149010B2 (en) 2018-05-18 2021-10-19 Fujifilm Corporation Producing method for 3-difluoromethylpyrazole compound, producing method for 3-difluoromethylpyrazole-4-carboxylic acid compound, and pyrazolidine compound

Families Citing this family (65)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AR044519A1 (es) 2003-05-02 2005-09-14 Novartis Ag Derivados de piridin-tiazol amina y de pirimidin-tiazol amina
US8563573B2 (en) 2007-11-02 2013-10-22 Vertex Pharmaceuticals Incorporated Azaindole derivatives as CFTR modulators
US8940771B2 (en) 2007-12-20 2015-01-27 Novartis Ag Organic compounds
KR20110016955A (ko) * 2008-06-13 2011-02-18 바이엘 크롭사이언스 아게 살해충제로서의 신규 헤테로방향족 아미드 및 티오아미드
UA104147C2 (uk) 2008-09-10 2014-01-10 Новартис Аг Похідна піролідиндикарбонової кислоти та її застосування у лікуванні проліферативних захворювань
RU2011123647A (ru) 2008-11-10 2012-12-20 Вертекс Фармасьютикалз Инкорпорейтед Соединения, полезные в качестве ингибиторов atr киназы
SG172248A1 (en) 2008-12-19 2011-07-28 Vertex Pharma Pyrazine derivatives useful as inhibitors of atr kinase
US8293753B2 (en) 2009-07-02 2012-10-23 Novartis Ag Substituted 2-carboxamide cycloamino ureas
US8802868B2 (en) 2010-03-25 2014-08-12 Vertex Pharmaceuticals Incorporated Solid forms of (R)-1(2,2-difluorobenzo[D][1,3]dioxo1-5-yl)-N-(1-(2,3-dihydroxypropyl-6-fluoro-2-(1-hydroxy-2-methylpropan2-yl)-1H-Indol-5-yl)-Cyclopropanecarboxamide
MX353408B (es) 2010-04-22 2018-01-11 Vertex Pharma Proceso para producir compuestos de cicloalquilcarboxamido-indol.
CA2798763A1 (en) 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
EP2569287B1 (en) 2010-05-12 2014-07-09 Vertex Pharmaceuticals Inc. Compounds useful as inhibitors of atr kinase
US9062008B2 (en) 2010-05-12 2015-06-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
WO2011143399A1 (en) 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
EP2569284B1 (en) 2010-05-12 2015-07-08 Vertex Pharmaceuticals Incorporated 2-aminopyridine derivatives useful as inhibitors of atr kinase
WO2011143419A1 (en) 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated Pyrazines useful as inhibitors of atr kinase
NZ605627A (en) 2010-06-23 2015-06-26 Vertex Pharma Pyrrolo-pyrazine derivatives useful as inhibitors of atr kinase
EP2595962B1 (en) * 2010-07-23 2018-07-11 Solvay Sa Process for the preparation of esters of 1-substituted-3-fluoroalkyl-pyrazole-4-carboxylic acids
CN101921268B (zh) * 2010-08-27 2016-08-03 中山大学肿瘤防治中心 5-噻唑酰胺类化合物及生物学应用
JP2013249256A (ja) * 2010-09-15 2013-12-12 Astellas Pharma Inc 脂肪性肝疾患治療薬
WO2012138938A1 (en) 2011-04-05 2012-10-11 Vertex Pharmaceuticals Incorporated Aminopyrazine compounds useful as inhibitors of tra kinase
WO2012178125A1 (en) 2011-06-22 2012-12-27 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2012178123A1 (en) 2011-06-22 2012-12-27 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
EP2723747A1 (en) 2011-06-22 2014-04-30 Vertex Pharmaceuticals Inc. Compounds useful as inhibitors of atr kinase
MX2014000359A (es) * 2011-07-15 2014-05-01 Basf Se Metodos plaguicidas que utilizan compuestos de 3 - piridin tiazol sustituido y derivados para combatir las plagas de animales ii.
AR087274A1 (es) 2011-07-22 2014-03-12 Actelion Pharmaceuticals Ltd Derivados de amidas heterociclicas como antagonistas de receptores p2x7
US8846686B2 (en) 2011-09-30 2014-09-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
EP3878851A1 (en) 2011-09-30 2021-09-15 Vertex Pharmaceuticals Incorporated Process for making compounds useful as inhibitors of atr kinase
US8765751B2 (en) 2011-09-30 2014-07-01 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8853217B2 (en) 2011-09-30 2014-10-07 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
CN103957917A (zh) 2011-09-30 2014-07-30 沃泰克斯药物股份有限公司 用atr抑制剂治疗胰腺癌和非小细胞肺癌
WO2013071094A1 (en) 2011-11-09 2013-05-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
EP2776419B1 (en) 2011-11-09 2016-05-11 Vertex Pharmaceuticals Incorporated Pyrazine compounds useful as inhibitors of atr kinase
US8841337B2 (en) 2011-11-09 2014-09-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
EP2776420A1 (en) 2011-11-09 2014-09-17 Vertex Pharmaceuticals Incorporated Pyrazine compounds useful as inhibitors of atr kinase
US8846917B2 (en) 2011-11-09 2014-09-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
CN102603637A (zh) * 2012-01-18 2012-07-25 中国药科大学 吡唑化合物及其作为rtk和pi3k双重抑制剂的用途
NZ628910A (en) 2012-01-20 2016-02-26 Actelion Pharmaceuticals Ltd Heterocyclic amide derivatives as p2x7 receptor antagonists
PL2833973T3 (pl) 2012-04-05 2018-02-28 Vertex Pharmaceuticals Incorporated Związki użyteczne jako inhibitory kinazy ATR i ich terapie skojarzone
WO2013186089A2 (en) 2012-06-14 2013-12-19 Basf Se Pesticidal methods using substituted 3-pyridyl thiazole compounds and derivatives for combating animal pests
CN113243374B (zh) 2012-10-02 2022-04-26 拜耳农作物科学股份公司 作为杀虫剂的杂环化合物
EP2904406B1 (en) 2012-10-04 2018-03-21 Vertex Pharmaceuticals Incorporated Method for measuring atr inhibition mediated increases in dna damage
WO2014062604A1 (en) 2012-10-16 2014-04-24 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
TWI600654B (zh) 2012-11-19 2017-10-01 艾姆公司 用於治療寄生蟲疾病之化合物及組合物
US8871754B2 (en) 2012-11-19 2014-10-28 Irm Llc Compounds and compositions for the treatment of parasitic diseases
BR112015012454B1 (pt) 2012-12-07 2022-07-05 Vertex Pharmaceuticals Incorporated Compostos inibidores de atr quinase, seu uso e composição farmacêutica compreendendo os mesmos
WO2014091415A1 (en) 2012-12-12 2014-06-19 Actelion Pharmaceuticals Ltd Indole carboxamide derivatives as p2x7 receptor antagonists
EP2935211B1 (en) 2012-12-18 2016-11-09 Actelion Pharmaceuticals Ltd. Indole carboxamide derivatives as p2x7 receptor antagonists
CN104918617B (zh) 2013-01-22 2017-05-10 埃科特莱茵药品有限公司 作为p2x7受体拮抗剂的杂环酰胺衍生物
US9388198B2 (en) 2013-01-22 2016-07-12 Actelion Pharmaceuticals Ltd. Heterocyclic amide derivatives as P2X7 receptor antagonists
US9663519B2 (en) 2013-03-15 2017-05-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
SG11201604519PA (en) 2013-12-06 2016-07-28 Vertex Pharma 2-amino-6-fluoro-n-[5-fluoro-pyridin-3-yl]pyrazolo[1,5-a]pyrimidin-3-carboxamide compound useful as atr kinase inhibitor, its preparation, different solid forms and radiolabelled derivatives thereof
WO2015150300A1 (de) 2014-04-02 2015-10-08 Bayer Cropscience Ag N-(1-(hetero)aryl-1h-pyrazol-4-yl)-(hetero)arylamid-derivate und ihre verwendung als schädlingsbekämpfungsmittel
WO2015160787A1 (en) 2014-04-15 2015-10-22 Vertex Pharmaceuticals Incorporated Pharmaceutical compositions for the treatment of cystic fibrosis transmembrane conductance regulator mediated diseases
EP3152212B9 (en) 2014-06-05 2020-05-27 Vertex Pharmaceuticals Inc. Radiolabelled derivatives of a 2-amino-6-fluoro-n-[5-fluoro-pyridin-3-yl]- pyrazolo[1,5-a]pyrimidin-3-carboxamide compound useful as atr kinase inhibitor, the preparation of said compound and different solid forms thereof
WO2015195740A1 (en) 2014-06-17 2015-12-23 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of chk1 and atr inhibitors
EP3186229A4 (en) * 2014-07-31 2018-01-10 Dow AgroSciences LLC Process for the preparation of 3-(3-chloro-1h-pyrazol-1-yl)pyridine
EP3186230A4 (en) * 2014-07-31 2018-01-10 Dow AgroSciences, LLC Process for the preparation of 3-(3-chloro-1h-pyrazol-1-yl)pyridine
WO2016018442A1 (en) * 2014-07-31 2016-02-04 Dow Agrosciences Llc Process for the preparation of 3-(3-chloro-1h-pyrazol-1-yl)pyridine
US11464774B2 (en) 2015-09-30 2022-10-11 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of DNA damaging agents and ATR inhibitors
US9963462B2 (en) * 2015-09-30 2018-05-08 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften E.V. Sepiapterin reductase inhibitors
TW201722432A (zh) 2015-11-02 2017-07-01 H 朗德貝克公司 P2x受體之抑制劑
CN106187910A (zh) * 2016-07-07 2016-12-07 浙江大学 哒嗪类衍生物及其制备方法和用途
KR20220124176A (ko) 2019-12-06 2022-09-13 버텍스 파마슈티칼스 인코포레이티드 나트륨 채널의 조절제로서의 치환된 테트라하이드로푸란
AU2022284886A1 (en) 2021-06-04 2023-11-30 Vertex Pharmaceuticals Incorporated N-(hydroxyalkyl (hetero)aryl) tetrahydrofuran carboxamides as modulators of sodium channels

Citations (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2221647A1 (de) * 1971-05-05 1972-11-09 Uniroyal Inc 2-(3-Pyridyl)-thiazolabkoemmlinge und deren Verwendung als Insektizide
DE2331246A1 (de) * 1972-06-21 1974-01-17 Uniroyal Inc Antiinflammatorisch wirksame thiazolabkoemmlinge und diese enthaltende arzneipraeparate
WO1997005131A1 (en) * 1995-08-02 1997-02-13 J. Uriach & Cia. S.A. New carboxamides with antifungal activity
US5948777A (en) * 1997-03-18 1999-09-07 Smithkline Beecham Corporation Cannabinoid receptor agonists
US6323227B1 (en) * 1996-01-02 2001-11-27 Aventis Pharmaceuticals Products Inc. Substituted N-[(aminoiminomethyl or aminomethyl)phenyl]propyl amides
US6511998B2 (en) * 1999-05-12 2003-01-28 Cheryl P. Kordik Pyrazole carboxamides useful for the treatment of obesity and other disorders
US6596746B1 (en) * 1999-04-15 2003-07-22 Bristol-Myers Squibb Company Cyclic protein tyrosine kinase inhibitors
US20030207874A1 (en) * 2000-05-08 2003-11-06 Mcdonald Iain Mair Pyrazole derivatives and their use as gastrin and cholecystokin receptor ligands
WO2004041813A1 (en) * 2002-10-30 2004-05-21 Vertex Pharmaceuticals Incorporated Compositions useful as inhibitors of rock and other protein kinases
US20040167129A1 (en) * 2003-01-02 2004-08-26 Alexander Mayweg Novel CB 1 receptor inverse agonists
US20040220170A1 (en) * 2003-05-01 2004-11-04 Atkinson Robert N. Pyrazole-amides and sulfonamides as sodium channel modulators
US20040235926A1 (en) * 2003-05-07 2004-11-25 Pfizer Inc. Cannabinoid receptor ligands and uses thereof
US20050009827A1 (en) * 2003-05-19 2005-01-13 Aventis Pharma Deutschland Gmbh Triazole-derivatives as factor Xa inhibitors
US20050049288A1 (en) * 2003-06-26 2005-03-03 Fryszman Olga M. 5-MEMBERED HETEROCYCLE-BASED p38 KINASE INHIBITORS
US6864252B2 (en) * 2001-11-02 2005-03-08 Pfizer, Inc. Hydrazinyl and nitrogen oxide pyrazoles
US20050080087A1 (en) * 2003-10-10 2005-04-14 Annapurna Pendri Pyrazole derivatives as cannabinoid receptor modulators
US20050261290A1 (en) * 2004-05-06 2005-11-24 Hengmiao Cheng Novel compounds of proline and morpholine derivatives
US20060058302A1 (en) * 2001-11-12 2006-03-16 Pfizer Inc. Benzamide, heteroarylamide and reverse amides
US7125875B2 (en) * 1999-04-15 2006-10-24 Bristol-Myers Squibb Company Cyclic protein tyrosine kinase inhibitors
US7223782B2 (en) * 2001-11-01 2007-05-29 Icagen, Inc. Pyrazole-amides and -sulfonamides
US7491821B2 (en) * 2005-08-15 2009-02-17 Roche Palo Alto Llc Inhibitors of P2X3
US20090156615A1 (en) * 2006-06-05 2009-06-18 Natalie Dales Organic compounds

Family Cites Families (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SE9704544D0 (sv) 1997-12-05 1997-12-05 Astra Pharma Prod Novel compounds
SE9704545D0 (sv) * 1997-12-05 1997-12-05 Astra Pharma Prod Novel compounds
PE20011010A1 (es) * 1999-12-02 2001-10-18 Glaxo Group Ltd Oxazoles y tiazoles sustituidos como agonista del receptor activado por el proliferador de peroxisomas humano
WO2008011131A2 (en) 2006-07-21 2008-01-24 Takeda Pharmaceutical Company Limited Amide compounds
CN1732164A (zh) * 2002-10-30 2006-02-08 沃泰克斯药物股份有限公司 可用作rock及其他蛋白质激酶抑制剂的组合物
NZ539546A (en) 2002-11-26 2007-11-30 Pfizer Prod Inc Phenyl substituted piperidine compounds for use as PPAR activators
ATE355273T1 (de) * 2003-05-12 2006-03-15 Pfizer Prod Inc Benzamidinhibitoren des p2x7-rezeptors
EP1479679A1 (en) * 2003-05-19 2004-11-24 Aventis Pharma Deutschland GmbH Triazole-derivatives as factor Xa inhibitors
SE0302192D0 (sv) * 2003-08-08 2003-08-08 Astrazeneca Ab Novel compounds
RU2006115602A (ru) * 2003-10-08 2007-11-20 Вертекс Фармасьютикалз Инкорпорейтед (Us) Модуляторы переносчиков атф-связывающих кассет
EP1682127B1 (en) * 2003-11-14 2009-07-29 Vertex Pharmaceuticals Incorporated Thiazoles and oxazoles useful as modulators of atp-binding cassette transporters
WO2005061462A2 (en) 2003-12-19 2005-07-07 Neurogen Corporation Diaryl pyrazole derivatives and their use as neurokinin-3 receptor modulators
WO2005075435A1 (en) * 2004-01-30 2005-08-18 Vertex Pharmaceuticals Incorporated Modulators of atp-binding cassette transporters
RU2006138036A (ru) * 2004-03-30 2008-05-10 Чирон Корпорейшн (Us) Производные замещенного тиофена в качестве противораковых средств
DE102004051277A1 (de) * 2004-10-21 2006-04-27 Merck Patent Gmbh Heterocyclische Carbonylverbindungen
WO2006089076A2 (en) * 2005-02-18 2006-08-24 Neurogen Corporation Thiazole amides, imidazole amides and related analogues
JP2009514864A (ja) 2005-11-07 2009-04-09 アボット・ラボラトリーズ P2x7受容体拮抗薬および使用方法
CN101304975A (zh) 2005-11-09 2008-11-12 艾博特公司 P2x7受体拮抗剂及其用途
EP1989205B1 (en) 2006-01-23 2012-06-13 Vertex Pharmceuticals Incorporated Thiophene-carboxamides useful as inhibitors of protein kinases
MX2009003821A (es) * 2006-10-10 2009-05-25 Amgen Inc Compuestos de n-aril pirazol para usarse contra la diabetes.
KR20090094125A (ko) 2006-12-08 2009-09-03 엑셀리시스, 인코포레이티드 Lxr 및 fxr 조절자
CL2008000836A1 (es) 2007-03-26 2008-11-07 Actelion Pharmaceuticals Ltd Compuestos derivados de tiazolidina, antagonistas del receptor de orexina; composicion farmaceutica que los comprende; y su uso en el tratamiento de neurosis emocional, depresion grave, trastornos psicoticos, alzheimer, parkinson, dolor, entre otras.
WO2008138876A1 (en) 2007-05-10 2008-11-20 Glaxo Group Limited Pyrazole derivatives as p2x7 modulators

Patent Citations (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2221647A1 (de) * 1971-05-05 1972-11-09 Uniroyal Inc 2-(3-Pyridyl)-thiazolabkoemmlinge und deren Verwendung als Insektizide
DE2331246A1 (de) * 1972-06-21 1974-01-17 Uniroyal Inc Antiinflammatorisch wirksame thiazolabkoemmlinge und diese enthaltende arzneipraeparate
US3852293A (en) * 1972-06-21 1974-12-03 Uniroyal Inc 4-phenyl-2-(3-pyridyl)-thiazole carboxamides
WO1997005131A1 (en) * 1995-08-02 1997-02-13 J. Uriach & Cia. S.A. New carboxamides with antifungal activity
US6323227B1 (en) * 1996-01-02 2001-11-27 Aventis Pharmaceuticals Products Inc. Substituted N-[(aminoiminomethyl or aminomethyl)phenyl]propyl amides
US5948777A (en) * 1997-03-18 1999-09-07 Smithkline Beecham Corporation Cannabinoid receptor agonists
US6596746B1 (en) * 1999-04-15 2003-07-22 Bristol-Myers Squibb Company Cyclic protein tyrosine kinase inhibitors
US7125875B2 (en) * 1999-04-15 2006-10-24 Bristol-Myers Squibb Company Cyclic protein tyrosine kinase inhibitors
US6511998B2 (en) * 1999-05-12 2003-01-28 Cheryl P. Kordik Pyrazole carboxamides useful for the treatment of obesity and other disorders
US20030207874A1 (en) * 2000-05-08 2003-11-06 Mcdonald Iain Mair Pyrazole derivatives and their use as gastrin and cholecystokin receptor ligands
US7223782B2 (en) * 2001-11-01 2007-05-29 Icagen, Inc. Pyrazole-amides and -sulfonamides
US6864252B2 (en) * 2001-11-02 2005-03-08 Pfizer, Inc. Hydrazinyl and nitrogen oxide pyrazoles
US20060058302A1 (en) * 2001-11-12 2006-03-16 Pfizer Inc. Benzamide, heteroarylamide and reverse amides
US7235549B2 (en) * 2001-11-12 2007-06-26 Pfizer Inc. Benzamide, heteroarylamide and reverse amides
WO2004041813A1 (en) * 2002-10-30 2004-05-21 Vertex Pharmaceuticals Incorporated Compositions useful as inhibitors of rock and other protein kinases
US20040167129A1 (en) * 2003-01-02 2004-08-26 Alexander Mayweg Novel CB 1 receptor inverse agonists
US20040220170A1 (en) * 2003-05-01 2004-11-04 Atkinson Robert N. Pyrazole-amides and sulfonamides as sodium channel modulators
US20050020564A1 (en) * 2003-05-01 2005-01-27 Atkinson Robert N. Pyrazole-amides and sulfonamides as sodium channel modulators
US20040235926A1 (en) * 2003-05-07 2004-11-25 Pfizer Inc. Cannabinoid receptor ligands and uses thereof
US20050009827A1 (en) * 2003-05-19 2005-01-13 Aventis Pharma Deutschland Gmbh Triazole-derivatives as factor Xa inhibitors
US20050049288A1 (en) * 2003-06-26 2005-03-03 Fryszman Olga M. 5-MEMBERED HETEROCYCLE-BASED p38 KINASE INHIBITORS
US20050080087A1 (en) * 2003-10-10 2005-04-14 Annapurna Pendri Pyrazole derivatives as cannabinoid receptor modulators
US20050261290A1 (en) * 2004-05-06 2005-11-24 Hengmiao Cheng Novel compounds of proline and morpholine derivatives
US7491821B2 (en) * 2005-08-15 2009-02-17 Roche Palo Alto Llc Inhibitors of P2X3
US20090156615A1 (en) * 2006-06-05 2009-06-18 Natalie Dales Organic compounds

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8686018B2 (en) 2010-09-21 2014-04-01 Eisai R&D Management Co., Ltd. Pharmaceutical composition
US9000024B2 (en) 2010-09-21 2015-04-07 Eisai R&D Management Co., Ltd. Pharmaceutical composition
US9725414B2 (en) 2011-11-30 2017-08-08 Bayer Intellectual Property Gmbh Fungicidal N-bicycloalkyl and N-tricycloalkyl pyrazole-4-(thio)carboxamide derivatives
US9487508B2 (en) * 2012-09-19 2016-11-08 Ironwood Pharmaceuticals, Inc. SGC stimulators
US11149010B2 (en) 2018-05-18 2021-10-19 Fujifilm Corporation Producing method for 3-difluoromethylpyrazole compound, producing method for 3-difluoromethylpyrazole-4-carboxylic acid compound, and pyrazolidine compound

Also Published As

Publication number Publication date
SG187396A1 (en) 2013-02-28
EP2178865A4 (en) 2011-03-16
IL202499A0 (en) 2010-06-30
ZA200908447B (en) 2011-05-25
HK1142330A1 (zh) 2010-12-03
KR20100032861A (ko) 2010-03-26
CA2691507C (en) 2016-06-21
CN101711245A (zh) 2010-05-19
AU2008275891B2 (en) 2013-10-10
CO6251318A2 (es) 2011-02-21
KR101593237B1 (ko) 2016-02-12
CN104844578B (zh) 2018-06-19
BRPI0813426A2 (pt) 2014-12-23
HK1213879A1 (zh) 2016-07-15
PH12014501677B1 (en) 2015-08-03
CA2691507A1 (en) 2009-01-22
CY1116809T1 (el) 2017-03-15
PL2178865T3 (pl) 2015-12-31
WO2009012482A2 (en) 2009-01-22
US20150005293A1 (en) 2015-01-01
RS54303B1 (en) 2016-02-29
CN104844578A (zh) 2015-08-19
EA200971084A1 (ru) 2010-04-30
MX2009014159A (es) 2010-03-04
HUE026446T2 (en) 2016-05-30
UA105758C2 (uk) 2014-06-25
PT2178865E (pt) 2015-11-16
US9133204B2 (en) 2015-09-15
SI2178865T1 (sl) 2015-11-30
DK2178865T3 (en) 2015-10-19
JP2014058545A (ja) 2014-04-03
MY154668A (en) 2015-07-15
WO2009012482A3 (en) 2009-04-30
EP2178865B1 (en) 2015-08-19
PH12014501677A1 (en) 2015-08-03
EP2178865A2 (en) 2010-04-28
HRP20151052T1 (en) 2015-11-06
AU2008275891A1 (en) 2009-01-22
IL202499A (en) 2017-05-29
JP2010533736A (ja) 2010-10-28
JP5813734B2 (ja) 2015-11-17
NZ582057A (en) 2012-03-30
EA017250B1 (ru) 2012-11-30
ES2551095T3 (es) 2015-11-16
CN101711245B (zh) 2015-03-25
KR20150118197A (ko) 2015-10-21

Similar Documents

Publication Publication Date Title
US9133204B2 (en) 5-membered heterocyclic amides and related compounds
US8580812B2 (en) Heteroaryl amide analogues as P2X7 antagonists
US8431593B2 (en) Heteroaryl amide derivatives
US20120190680A1 (en) Heteroaryl Amide Analogues
AU2008286946B2 (en) Heteroaryl amide analogues
WO2009108551A2 (en) Heteroaryl amide analogues
Ihle et al. Heteroaryl amide analogues as P2X7 antagonists

Legal Events

Date Code Title Description
AS Assignment

Owner name: H. LUNDBECK A/S, DENMARK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NEUROGEN CORPORATION;REEL/FRAME:023763/0180

Effective date: 20090427

Owner name: NEUROGEN CORPORATION, CONNECTICUT

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LI, HONGBIN;YUAN, JUN;BAKTHAVATCHALAM, RAJAGOPAL;AND OTHERS;SIGNING DATES FROM 20070719 TO 20070726;REEL/FRAME:023763/0077

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: MINDIMMUNE THERAPEUTICS, INC., RHODE ISLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:H. LUNDBECK A/S;REEL/FRAME:046610/0343

Effective date: 20180608