WO2003093236A1 - Composes 1-(pyrid-2-yl)-piperazine utilises en tant qu'inhibiteur du recepteur de glutamate metabotropique - Google Patents

Composes 1-(pyrid-2-yl)-piperazine utilises en tant qu'inhibiteur du recepteur de glutamate metabotropique Download PDF

Info

Publication number
WO2003093236A1
WO2003093236A1 PCT/US2003/013964 US0313964W WO03093236A1 WO 2003093236 A1 WO2003093236 A1 WO 2003093236A1 US 0313964 W US0313964 W US 0313964W WO 03093236 A1 WO03093236 A1 WO 03093236A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
halo
pharmaceutically acceptable
alkyl
acceptable salt
Prior art date
Application number
PCT/US2003/013964
Other languages
English (en)
Inventor
Parviz Gharagozloo
Khondaker Islam
Donald J. Kyle
Qun Sun
Laykea Tafesse
John William Frank Whitehead
Ji Yang
Xiaoming Zhou
Original Assignee
Euro-Celtique, S.A.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Euro-Celtique, S.A. filed Critical Euro-Celtique, S.A.
Priority to AU2003228857A priority Critical patent/AU2003228857A1/en
Publication of WO2003093236A1 publication Critical patent/WO2003093236A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/72Nitrogen atoms
    • C07D213/74Amino or imino radicals substituted by hydrocarbon or substituted hydrocarbon radicals
    • 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
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/79Acids; Esters
    • C07D213/80Acids; Esters in position 3
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/84Nitriles
    • C07D213/85Nitriles in position 3
    • 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
    • C07D401/14Heterocyclic 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
    • 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

Definitions

  • the present invention relates to Piperazine Compounds, compositions comprising a Piperazine Compound and methods for treating or preventing a condition such as pain, urinary incontinence (UI), an addictive disorder, Parkinson's disease, parkinsonism, anxiety, epilepsy, stroke, a seizure, a pruritic condition, psychosis, a cognitive disorder, a memory deficit, restricted brain function, Huntington's chorea, amyotrophic lateral sclerosis (ALS), dementia, retinopathy, a muscle spasm, a migraine, vomiting, dyskinesia or depression comprising administering to the animal in an animal in need thereof an effective amount of a Piperazine Compound.
  • UI urinary incontinence
  • UI urinary incontinence
  • an addictive disorder such as depression
  • Parkinson's disease parkinsonism
  • anxiety epilepsy
  • stroke a seizure, a pruritic condition
  • psychosis a cognitive disorder
  • a memory deficit such as Huntington's chorea
  • Nociceptive pain includes tissue injury-induced pain and inflammatory pain such as that associated with arthritis.
  • Neuropathic pain is caused by damage to the peripheral or cental nervous system and is maintained by aberrant somatosensory processing.
  • Group I mGluRs mGluRl and mGluR5
  • NR1 vaniloid receptors
  • Inhibiting mGluRl or mGluR5 reduces pain, as shown by in vivo treatment with antibodies selective for either mGluRl or mGluR5, where neuropathic pain in rats was attenuated (M.E. Fundytus et al, NeuroReport 9:731-735 (1998)). It has also been shown that antisense oligonucleotide knockdown of mGluRl alleviates both neuropathic and inflammatory pain (M.E. Fundytus et al, British Journal of Pharmacology 132:354-367 (2001); M.E. Fundytus et al, Pharmacology, Biochemsitry & Behavior 73:401-410 (2002)).
  • Nociceptive pain has been traditionally managed by administering non- opioid analgesics, such as acetylsalicylic acid, choline magnesium trisalicylate, acetaminophen, ibuprofen, fenoprofen, diflusinal, and naproxen; or opioid analgesics, including morphine, hydromorphone, methadone, levorphanol, fentanyl, oxycodone, and oxymorphone.
  • opioid analgesics including morphine, hydromorphone, methadone, levorphanol, fentanyl, oxycodone, and oxymorphone.
  • NMDA antagonists e.g. ketamine, dextromethorphan
  • topical lidocaine for post-herpetic neuralgia
  • tricyclic antidepressants e.g. fluoxetine, sertraline and amitriptyline.
  • UI uncontrollable urination, generally caused by bladder-detrusor- muscle instability.
  • UI affects people of all ages and levels of physical health, both in health care settings and in the community at large. At present, UI afflicts 15-30% of elderly people living at home, one-third of those living in acute-care settings, and at least one-half of those living in long-term care institutions (R.M. Resnick, Lancet 346:94 (1995)). Persons having UI are predisposed to also having urinary-tract infections, pressure ulcers, perineal rashes and urosepsis.
  • UI is associated with embarrassment, social stigmatization, depression and a risk of institutionalization (Herzo et al, Annu. Rev. Gerontol. Geriatr. 9:74 (1989)). Economically, the costs of UI are great; in the United States alone, health-care costs associated with UI are over $15 billion per annum.
  • Physiologic bladder contraction results in large part from acetylcholine- induced stimulation of post-ganglionic muscarinic-receptor sites on bladder smooth muscle.
  • Treatments for UI include the administration of drugs having bladder-relaxant properties, which help to control bladder-detrusor-muscle overactivity.
  • anticholinergics such as propantheline bromide and glycopyrrolate
  • smooth-muscle relaxants such as a combination of racemic oxybutynin and dicyclomine or an anticholmergic
  • drugs can cause physical and/or psychological addiction.
  • the most well known types of these drugs include opiates, such as heroin, opium, and morphine; sympathomimetics, including cocaine and amphetamines; sedative-hypnotics, including alcohol, benzodiazepines and barbiturates; and nicotine, which has effects similar to opioids and sympathomimetics.
  • Drug addiction is characterized by a craving or compulsion for taking the drug and an inability to limit its intake. Additionally, drug dependence is associated with drug tolerance, the loss of effect ofthe drug following repeated administration, and withdrawal, the appearance of physical and behavioral symptoms when the drug is not consumed. Sensitization occurs if repeated administration of a drug leads to an increased response to each dose.
  • U.S. Patent No. 5,075,341 to Mendelson et al. discloses the use of a mixed opiate agonist/antagonist to treat cocaine and opiate addiction.
  • U.S. Patent No. 5,232,934 to Downs discloses administration of 3-phenoxypyridine to treat addiction.
  • U.S. Patents No. 5,039,680 and 5,198,459 to Imperato et al. disclose using a serotonin antagonist to treat chemical addiction.
  • U.S. Patent. No. 5,762,925 to Sagan discloses implanting encapsulated adrenal medullary cells into an animal's central nervous system to inhibit the development of opioid intolerance.
  • U.S. Patent No. 6,204,284 to Beer et al. discloses racemic ( ⁇ )-l-(3,4-dichlorophenyl)-3-azabicyclo[3.1.0]hexane for use in the prevention or relief of a withdrawal syndrome resulting from addiction to drugs and for the treatment of chemical dependencies. Glutamate release is enhanced during opioid withdrawal (K.
  • Parkinson's disease is a clinical syndrome comprising bradykinesia (slowness and poverty of movement), muscular rigidity, resting tremor (which usually abates during voluntary movement), and an impairment of postural balance leading to disturbance of gait and falling.
  • the features of Parkinson's disease are a loss of pigmented, dopaminergic neurons ofthe substantia nigra pars compacta and the appearance of intracellular inclusions known as Lewy bodies (Goodman and Gillman 's The Pharmaceutical Basis of Therapeutics 506 (9 th ed. 1996)). Without treatment, Parkinson's disease progresses to a rigid akinetic state in which patients are incapable of caring for themselves. Death frequently results from complications of immobility, including aspiration pneumonia or pulmonary embolism.
  • Drugs commonly used for the treatment of Parkinson's disease include carbidopa/levodopa, pergolide, bromocriptine, selegiline, amantadine, and trihexyphenidyl hydrochloride.
  • Anxiety is a fear, apprehension, or dread of impending danger often accompanied by restlessness, tension, tachycardia, and dyspnea.
  • Other symptoms commonly associated with anxiety include depression, especially accompanied with dysthymic disorder (chronic "neurotic" depression); panic disorder; agoraphobia and other specific phobias; eating disorders; and many personality disorders. Often anxiety is unattached to a clearly identified treatable primary illness. If a primary illness is found, however, it can be desirable to deal with the anxiety at the same time as the primary illness.
  • benzodiazepines are the most commonly used anti-anxiety agents for generalized anxiety disorder. Benzodiazepines, however, carry the risk of producing impairment of cognition and skilled motor functions, particularly in the elderly, which can result in confusion, delerium, and falls with fractures. Sedatives are also commonly prescribed for treating anxiety.
  • the azapirones such as buspirone, are also used to treat moderate anxiety. The azapirones, however, are less useful for treating severe anxiety accompanied with panic attacks.
  • Antagonists ofthe mGluR5 receptor have also been shown to exert anxiolytic and anti-depressant activity in in vivo animal models (E. Tatarczynska et al, Br. J. Pharmacol. 132(7): 1423-1430 (2001) and PJ.M. Will et al, Trends in Pharmacological Sciences 22(7):331-37 (2001)).
  • Epilepsy is a disorder characterized by the tendency to have recurring seizures.
  • the etiology commonly consists of lesions in some part ofthe cortex, such as a tumor; developmental malformation; or damage due to trauma or stroke. In some cases the etiology is genetic.
  • An epileptic seizure can be triggered by repetitive sounds, flashing lights, video games, or touching certain parts ofthe body.
  • Epilepsy is typically treated with anti-seizure drugs. In epilepsy cases, where anti-seizure drugs are ineffective, and the defect in the brain is isolated to a small area ofthe brain, surgical removal of that part ofthe brain can be helpful in alleviating the seizures. In patients who have several sources for the seizures or who have seizures that spread quickly to all parts ofthe brain, surgical removal ofthe nerve fibers that connect the two sides ofthe brain can be helpful.
  • a seizure is the result of abnormal electrical discharge in the brain.
  • the discharge can involve a small area ofthe brain and lead to the person only noticing an odd taste or smell or it can involve a large area ofthe brain and lead to convulsions, i.e., a seizure that causes jerking and spasms ofthe muscles throughout the body. Convulsions can also result in brief attacks of altered consciousness and loss of consciousness, muscle control, or bladder control.
  • a seizure is often preceded by an aura, i.e., unusual sensation of smell, taste, or vision or an intense feeling that a seizure is about to begin.
  • a seizure typically lasts for about 2 to 5 minutes. When the seizure ends the person can have headache, sore muscles, unusual sensations, confusion, and profound fatigue (postictal state). Usually the person cannot remember what happened during the seizure.
  • Examples of drugs for treating a seizure and epilepsy include carbamazepine, ethosuximide, gabapentin, lamotrigine, phenobarbital, phenytoin, primidone, valproic acid, trimethadione, benzodiazepines, ⁇ -vinyl GABA, acetazolamide, and felbamate.
  • Anti-seizure drugs can have side effects such as drowsiness; hyperactivity; hallucinations; inability to concentrate; central and peripheral nervous system toxicity, such as nystagmus, ataxia, diplopia, and vertigo; gingival hype ⁇ lasia; gastrointestinal disturbances such as nausea, vomiting, epigastric pain, and anorexia; endocrine effects such as inhibition of antidiuretic hormone, hyperglycemia, glycosuria, osteomalacia; and hypersensitivity such as scarlatimform rash, morbilliform rash, Stevens- Johnson syndrome, systemic lupus erythematosus, and hepatic necrosis; and hematological reactions such as red-cell aplasia, agranulocytosis, thrombocytopenia, aplastic anemia, and megaloblastic anemia.
  • a stroke or cerebro vascular accident is the death of brain tissue (cerebral infarction) resulting from the lack of blood flow and insufficient oxygen to the brain.
  • a stroke can be either ischemic or hemorrhagic.
  • ischemic stroke blood supply to the brain is cut off because of atherosclerosis or a blood clot that has blocked a blood vessel.
  • hemorrhagic stroke a blood vessel bursts preventing normal blood flow and allowing blood to leak into an area ofthe brain and destroying it. Most strokes develop rapidly and cause brain damage within minutes. In some cases, however, strokes can continue to worsen for several hours or days.
  • Symptoms of strokes vary depending on what part ofthe brain is effected. Symptoms include loss or abnormal sensations in an arm or leg or one side ofthe body, weakness or paralysis of an arm or leg or one side of the body, partial loss of vison or hearing, double vision, dizziness, slurred speech, difficulty in thinking ofthe appropriate word or saying it, inability to recognize parts of the body, unusual movements, loss of bladder control, imbalance, and falling, and fainting.
  • the symptoms can be permanent and can be associated with coma or stupor. Strokes can cause edema or swelling ofthe brain which can further damage brain tissue. For persons suffering from a stroke, intensive rehabilitation can help overcome the disability caused by impairment of brain tissue.
  • Rehabilitation trains other parts ofthe brain to assume the tasks previously performed by the damaged part.
  • drugs for treating strokes include anticoagulants such as heparin, drugs that break up clots such as streptokinase or tissue plasminogen activator, and drugs that reduce swelling such as mannitol or corticosteroids.
  • anticoagulants such as heparin
  • drugs that break up clots such as streptokinase or tissue plasminogen activator
  • drugs that reduce swelling such as mannitol or corticosteroids.
  • Pruritus is an unpleasant sensation that prompts scratching. Pruritus can be attributed to dry skin, scabies, dermatitis, herpetiformis, atopic dermatitis, pruritus vulvae et ani, malaria, insect bites, pediculosis, contact dermatitis, drug reactions, urticaria, urticarial eruptions of pregnancy, psoriasis, lichen planus, lichen simplex chronicus, exfoliative dermatitis, folliculitis, bullous pemphigoid, or fiberglass dermatitis.
  • pruritus is treated by phototherapy with ultraviolet B or PUVA or with therapeutic agents such as naltrexone, nalmefene, danazol, and tricyclic antidepressants.
  • mGluR5 metabotropic glutamate receptor 5
  • the present invention encompasses compounds having the formula (I):
  • A is -C(O)-, -C(S)-, -CH(C ⁇ -C 4 alkyl)-, -C(C ⁇ -C 4 alky ⁇ )(C*-C 4 alkyl)-, -CH(phenyl)- or -C(phenyl) 2 -, each phenyl independently being unsubstituted or substituted with one or more R 7 groups; each R-.
  • R 2 is -(CrC ⁇ o)alkyl, -(C 2 -C ⁇ 0 )alkenyl, -(C 2 -C, 0 )alkynyl, -(C 3 - C ⁇ o)cycloalkyl, -(C 8 -C ⁇ 4 )bicycloalkyl, -(C 8 -C ⁇ 4 )tricycloalkyl, -(C 5 -C ⁇ o)cycloalkenyl, -(C 8 -C ⁇ 4 )bicycloalkenyl, -(C 8 -C ⁇ 4 )bicycloalkenyl, -(C 8 -C ⁇ 4 )
  • R 2 is -phenyl, -naphthyl or -(C ⁇ 4 )aryl, each of which is unsubstituted or substituted with one or more R 5 groups, or
  • each R 5 is independently -(C ⁇ -C 6 )alkyl, -O(C ⁇ -C 6 )alkyl, -(C 2 -C 6 )alkenyl,
  • the present invention also encompasses compounds having the formula (I), and pharmaceutically acceptable salts thereof, wherein: A is -C(O)-, -C(S)-, -CH 2 -, -CH(C,-C 4 alkyl)-, -C(C,-C 4 alkyl)(C*-C 4 alkyl)-, -CH(phenyl)- or -C(phenyl) -, each phenyl independently being unsubstituted or substituted with one or more R 7 groups; each Ri is independently -H, -(C--C 3 )alkyl, -O(C ⁇ -C 3 alkyl), -halo, -OCF 3 , -NO , -OH, -CN, -S(O) 2 R-j, -C(O)OR-t, -OC(O)R4, -NH 2 or -NHR4;
  • R 2 is -(Ci-C 10 )alkyl, -(C 2 -C, 0 )alkenyl, -(C 2 -C 10 )alkynyl, -(C 3 - C ⁇ o)cycloalkyl, -(C 8 -C ⁇ 4 )bicycloalkyl, -(C 8 -C ⁇ )tricycloalkyl, -(C 5 -C ⁇ 0 )cycloalkenyl, -(C 8 -C ⁇ 4 )bicycloalkenyl or -(C 8 -C ⁇ 4 )tricycloalkenyl, each of which is unsubstituted or substituted with one or more R 3 groups, or R2 is -phenyl, -naphthyl or -(C* )aryl, each of which is unsubstituted or substituted with one or more R 5 groups; each R 3 is independently -CN, -OH, -hal
  • each R 5 is independently -(C**-C 6 )alkyl, -O(C ⁇ -C 6 )alkyl, -(C 2 -C 6 )alkenyl, -(C 2 -C 6 )alkynyl, -(C -C 8 )cycloalkyl, -(C 5 -C 8 )cycloalkenyl, -phenyl, -(3- to 5- membered)heterocycle, -C(halo) 3 or -CH(halo) 2 ; each R 5 is independently -(C**-C 6 )alkyl, -O(C ⁇ -C 6 )alkyl, -(C 2 -C 6 )alkenyl, -(C 2 -C 6 )alkynyl, -(C -C 8 )cycloalkyl, -(C 5 -C 8 )cycloalkenyl, -phenyl, -(C 3
  • the present invention also encompasses compounds having the formula (I), and pharmaceutically acceptable salts thereof, wherein: A is -C(O)-, -C(S)-, -CH(C ⁇ -C 4 alkyl)- or -C(C*-C 4 alkyl)(C C 4 alkyl)-; each Ri is independently -(C ⁇ -C 3 )alkyl, -halo, -NO , -OH, or -CN; m is O or 1; n is an integer from 1-4; R 2 is -(C ⁇ -C ⁇ o)alkyl, -(C 2 -C 10 )alkenyl, -(C 2 -C ⁇ o)alkynyl, -(C 3 -C ⁇ o)cycloalkyl, -(C 8 -C ⁇ 4 )bicycloalkyl, -(C 8 -C ⁇ 4 )tricycloalkyl, -(C 5 -C ⁇ o)cycloalken
  • R 2 is -phenyl, -naphthyl or -(C* 4 )aryl, each of which is unsubstituted or substituted with one or more R 5 groups, or
  • each R is independently -H, -(C ⁇ -C 6 )alkyl, -(C 2 -C 6 )alkenyl, -(C 2 - C 6 )alkynyl, -(C 3 -C 8 )cycloalkyl, -(C 5 -C 8 )cycloalkenyl, -phenyl, -(3- to 5- membered)heterocycle, -C(halo) 3 or -CH(halo) 2 ; each R 5 is independently -(C ⁇ -C 6 )alkyl, -(C 2 -C 6 )alkenyl, -(C 2 -C 6 )alkenyl, -(C 2 -C 6 )
  • A is -C(O)-, -C(S)-, -CH 2 -, -CH(d-C 4 alkyl)- or -C(C C 4 alkyl)(C . -C 4 alkyl)-; each R t is independently -(C- ⁇ -C )alkyl, -halo, -NO 2 , -OH or -CN; m is O or l; n is an integer from 1-4;
  • R 2 is -(C ⁇ -C 10 )alkyl, -(C 2 -C ⁇ 0 )alkenyl, -(C 2 -C ⁇ o)alkynyl, -(C 3 - C ⁇ o)cycloalkyl, -(C 8 -C ⁇ 4 )bicycloalkyl, -(C 8 -C 14 )tricycloalkyl, -(C 5 -C ⁇ o)cycloalkenyl, -(C 8 -C* )bicycloalkenyl or -(C 8 -C ⁇ 4 )tricycloalkenyl, each of which is unsubstituted or substituted with one or more R 3 groups, or
  • the present invention also encompasses compounds having the formula (la):
  • A is -C(O)-, -C(S)-, -CH(C ⁇ -C 4 alkyl)- or -C(C C 4 alkyl)(C ⁇ -C 4 alkyl)-; each Ri is independently -(C ⁇ -C 3 )alkyl, -halo, -NO 2 , -OH or -CN; n is an integer from 1-4;
  • R 2 is -(C ⁇ -C ⁇ o)alkyl, -(C 2 -C ⁇ 0 )alkenyl, -(C 2 -C 10 )alkynyl, -(C 3 -C ⁇ o)cycloalkyl, -(C 8 -C ⁇ 4 )bicycloalkyl, -(C 8 -C ⁇ 4 )tricycloalkyl, -(C 5 -
  • eachR 5 is independently -(C ⁇ -C 6 )alkyl, -(C2-C 6 )alkenyl, -(C 2 -C 6 )alkynyl, -(C 3 -C 8 )cycloalkyl, -(C 5 -C 8 )cycloalkenyl, -phenyl, -(3- to 5- membered)heterocycle, -C(halo) 3 or -CH(halo) 2 ; eachR 5 is independently -(C ⁇ -C 6 )alkyl, -(C2-C 6 )alkenyl, -(C 2 -C 6 )alkynyl, -(C 3 -C 8 )cycloalkyl, -(C 5 -C 8 )cycloalkenyl, -phenyl, -(3- to 5-membered)heterocycle, -C(halo) 3 , -CH(halo) 2 ,
  • the present invention also encompasses compounds having the formula (la), and pharmaceutically acceptable salts thereof, wherein:
  • A is -C(O)-, -C(S)-, -CH 2 -, -CH(C ⁇ -C 4 alkyl)- or -C(C r C 4 alkyl)(C ⁇ -C 4 alkyl)-; each Ri is independently -(Ci -C 3 )alkyl, -halo, -NO 2 , -OH or -CN; n is an integer from 1-4;
  • R 2 is -(C ⁇ -C ⁇ o)alkyl, -(C 2 -C, 0 )alkenyl, -(C 2 -C, 0 )alkvnyl, -(C 3 - C ⁇ o)cycloalkyl, -(C 8 -C ⁇ )bicycloalkyl, -(C 8 -C ⁇ 4 )tricycloalkyl, -(C 5 -C ⁇ o)cycloalkenyl, -(C 8 -C ⁇ )bicycloalkenyl or -(C 8 -C ⁇ )tricycloalkenyl, each of which is unsubstituted or substituted with one or more R 3 groups, or
  • R 2 is -phenyl, -naphthyl or -(C ⁇ 4 )aryl, each of which is unsubstituted or substituted with one or more R 5 groups; 03/093236
  • a compound of formula (I), (la) or a pharmaceutically acceptable salt thereof is useful for treating or preventing pain, UI, an addictive disorder, Parkinson's disease, parkinsonism, anxiety, epilepsy, stroke, a seizure, a pruritic condition, psychosis, a cognitive disorder, a memory deficit, restricted brain function, Huntington's chorea, ALS, dementia, retinopathy, a muscle spasm, a migraine, vomiting, dyskinesia or depression (each being a "Condition”) in an animal.
  • the invention also relates to compositions comprising an effective amount of a Piperazine Compound and a pharmaceutically acceptable carrier or excipient.
  • the compositions are useful for treating or preventing a Condition.
  • the invention further relates to methods for treating a Condition, comprising administering to an animal in need thereof an effective amount of a Piperazine Compound.
  • the invention further relates to methods for preventing a Condition, comprising administering to an animal in need thereof an effective amount of a Piperazine Compound.
  • the invention still further relates to methods for inhibiting mGluR5 function in a cell, comprising contacting a cell capable of expressing mGluR5 with an effective amount of a Piperazine Compound.
  • the invention still further relates to methods for inhibiting metabotropic glutamate receptor 1 ("mGluRl") function in a cell, comprising contacting a cell capable of expressing mGluRl with an effective amount of a Piperazine Compound.
  • the invention still further relates to a method for preparing a composition comprising the step of admixing a Piperazine Compound and a pharmaceutically acceptable carrier or excipient.
  • the invention still further relates to a kit comprising a container containing an effective amount of a Piperazine Compound.
  • the invention also relates to a kit comprising a container containing an effective amount of a Piperazine Compound and instructions for using the Piperazine Compound to treat or prevent a Condition.
  • FIG. 1 is a plot of calcium mobilization as measured by calcium fluoresence against log [Piperazine Compound AA] for the dose dependent inhibition of glutamate induced calcium mobilization in rat astrocytes in the presence of 10 ⁇ M glutamate.
  • the present invention encompasses compounds of Formula (I):
  • A is -C(O)-, -C(S)-, -CH(C ⁇ -C 4 alkyl)-, -C(C ⁇ -C alkyl)(C ⁇ -C 4 alkyl)- -CH ⁇ henyl)- or -C(phenyl) 2 -, each phenyl independently being unsubstituted or substituted with one or more R 7 groups; each Ri is independently -H, -(C ⁇ -C 3 )alkyl, -O(C ⁇ -C 3 alkyl), -halo, -CF , -OCF 3 , -NO 2 , -OH, -CN, -S(O) 2 R-t, -C(O)OR 4 , -OC(O)Rt, -NH 2 , -NHR4 or -N(R,) 2 ;
  • R 2 is -(C ⁇ -C ⁇ o)alkyl, -(C 2 -C 10 )alkenyl, -(C 2 -C ⁇ o)alkynyl, -(C 3 - C ⁇ o)cycloalkyl, -(C 8 -C ⁇ 4 )bicycloalkyl, -(C 8 -C] 4 )tricycloalkyl, -(C 5 -C ⁇ o)cycloalkenyl, -(C 8 -C ⁇ 4 )bicycloalkenyl, -(C 8 -C ⁇ 4 )tricycloalkenyl, -(3- to 7-membered)heterocycle or -(7- to 10-membered)bicycloheterocycle, each of which is unsubstituted or substituted with one or more R 3 groups, or
  • each t is independently -H, -(C ⁇ -C 6 )alkyl, -(C 2 -C 6 )alkenyl, -(C 2 - C 6 )alkynyl, -(C -C 8 )cycloalkyl, -(C 5 -C 8 )cycloalkenyl, -phenyl, -(3- to 5- membered)heterocycle, -C(halo) 3 or -CH(halo) 2 ; each R 5 is independently -(C ⁇ -C 6 )alkyl, -O(C ⁇ -C 6 )alkyl, -(C 2 -C 6 )alkenyl,
  • the present invention also encompasses compounds having the formula (I), and pharmaceutically acceptable salts thereof, wherein:
  • A is -C(O)-, -C(S)-, -CH(C ⁇ -C 4 alkyl)-, -C(C ⁇ -C 4 alkyl)(C ⁇ -C 4 alkyl)-, -CH ⁇ henyl)- or -C(phenyl) 2 -, each phenyl independently being unsubstituted or substituted with one or more R 7 groups, each Ri is independently -H, -(C ⁇ -C 3 )alkyl, -O(C ⁇ -C 3 alkyl), -halo, -OCF 3 , -NO 2 , -OH, -CN, -S(O) 2 Rt, -C(O)OR t , -OC(O)R t , -NH 2 or -NHR t ; R 2 is -(d-C ⁇ o)alkyl, -(C 2 -C ⁇ 0 )alkenyl, -(C 2 -d o
  • R 2 is -phenyl, -naphthyl or -(C ⁇ 4 )aryl, each of which is unsubstituted or substituted with one or more R 5 groups, or
  • each t is independently -H, -(C ⁇ -C 6 )alkyl, -(C 2 -C 6 )alkenyl, -(C 2 - C 6 )alkynyl, -(C 3 -C 8 )cycloalkyl, -(C 5 -C 8 )cycloalkenyl, -phenyl, -(3- to 5- membered)heterocycle, -C(halo) 3 or -CH(halo)2; each R 5 is independently -(C ⁇ -C 6 )alkyl, -O(C ⁇ -C 6 )alkyl, -(C 2 -C 6 )alkenyl,
  • the present invention also encompasses compounds having the formula (I), and pharmaceutically acceptable salts thereof, wherein: A is -C(O)-, -C(S)-, -CH 2 -, -CH(C ⁇ -C 4 alkyl)-, -C(C ⁇ -C 4 alkyl)(C ⁇ -C 4 alkyl)-, -CH ⁇ henyl)- or -C(phenyl) 2 -, each phenyl independently being unsubstituted or substituted with one or more R 7 groups; each Ri is independently -H, -(C ⁇ -C 3 )alkyl, -O(C ⁇ -C 3 alkyl), -halo, -CF 3 , -OCF 3 , -NO 2 , -OH, -CN, -S(O) 2 Rt, -C(O)ORt, -OC(O)R,, -NH 2 , -NHR, or -N(R-,) 2 ;
  • R 2 is -(C 1 -C,o)alkyl, -(C 2 -C, o )alkenyl, -(C 2 -C ⁇ 0 )alkynyl, -(C 3 - C ⁇ o)cycloalkyl, -(C 8 -C ⁇ 4 )bicycloalkyl, -(C 8 -C ⁇ 4 )tricycloalkyl, -(C 5 -C ⁇ o)cycloalkenyl, -(C 8 -C ⁇ 4 )bicycloalkenyl or -(C 8 -C ⁇ 4 )tricycloalkenyl, each of which is unsubstituted or substituted with one or more R 3 groups, or R 2 is -phenyl, -naphthyl or -(C ⁇ 4 )aryl, each of which is unsubstituted or substituted with one or more R 5 groups; each R 3 is independently -CN, -OH,
  • the present invention also encompasses compounds having the formula (I), and pharmaceutically acceptable salts thereof, wherein:
  • A is -C(O)-, -C(S)-, -CH 2 -, -CH(C ⁇ -C 4 alkyl)-, -C(C ⁇ -C 4 alkyl)(C ⁇ -C 4 alkyl)-, -CH ⁇ henyl)- or -C(phenyl) 2 -, each phenyl independently being unsubstituted or substituted with one or more R 7 groups; each Ri is independently -H, -(C ⁇ -C 3 )alkyl, -O(C ⁇ -C 3 alkyl), -halo, -OCF 3 , -NO 2 , -OH, -CN, -S(O) 2 Rt, -C(O)OR t , -OC(O)R t , -NH 2 or -NHR-t;
  • R 2 is -(C ⁇ -C ⁇ o)alkyl, -(C 2 -C, 0 )alkenyl, -(C 2 -C 10 )a]kynyl, -(C 3 - C ⁇ o)cycloalkyl, -(C 8 -C )bicycloalkyl, -(C 8 -C ⁇ 4 )tricycloalkyl, -(C 5 -C ⁇ o)cycloalkenyl, -(C 8 -C ⁇ 4 )bicycloalkenyl or -(C 8 -C ⁇ )tricycloalkenyl, each of which is unsubstituted or substituted with one or more R 3 groups, or
  • A is -C(O)-, -C(S)-, -CH(C ⁇ -C 4 alkyl)- or -C(C ⁇ -C 4 alkyl)(C ⁇ -C 4 alkyl)-; each Ri is independently -(C ⁇ -C 3 )alkyl, -halo, -NO 2 , -OH, or -CN; m is O or 1; n is an integer from 1-4;
  • R 2 is -H, -(C ⁇ -C ⁇ o)alkyl, -(C 2 -C ⁇ o)alkenyl, -(C 2 -C, 0 )alkynyl, -(C 3 -C ⁇ 0 )cycloalkyl, -(C 8 -C ⁇ 4 )bicycloalkyl, -(C 8 -C] 4 )tricycloalkyl, -(C 5 -C ⁇ o)cycloalkenyl, -(C 8 -C 14 )bicycloalkenyl, -(C 8 -C ⁇ 4 )tricycloalkenyl, -(3- to 7- membered)heterocycle, or -(7- to 10-membered)bicycloheterocycle, each of which is unsubstituted or substituted with one or more R 3 groups, or
  • each t is independently -H, -(C ⁇ -C 6 )alkyl, -(C 2 -C 6 )alkenyl, -(C 2 - C 6 )alkynyl, -(C 3 -C 8 )cycloalkyl, -(C 5 -C 8 )cycloalkenyl, -phenyl, -(3- to 5- membered)heterocycle, -C(halo) or -CH(halo) 2 ; each R 5 is independently -(C ⁇ -C 6 )alkyl, -(C 2 -C 6 )alkenyl, -(C 2 -C 6 )alkyn
  • the present invention also encompasses compounds having the formula (I), and pharmaceutically acceptable salts thereof, wherein:
  • A is -C(O)-, -C(S)-, -CH(C ⁇ -C 4 alkyl)- or -C(C ⁇ -C 4 alky ⁇ )(d-C 4 alkyl)-; each R x is independently -(d-C 3 )alkyl, -halo, -NO 2 , -OH, or -CN; m is 0 or 1 ; n is an integer from 1-4;
  • R 2 is -(C ⁇ -C ⁇ o)alkyl, -(C 2 -C ⁇ o)alkenyl, -(C 2 -C, 0 )alkynyl, -(C 3 -C] 0 )cycloalkyl, -(C 8 -C ⁇ 4 )bicycloalkyl, -(C 8 -C ⁇ 4 )tricyclo alkyl, -(C 5 -C ⁇ o)cycloalkenyl, -(C 8 -C ⁇ 4 )bicycloalkenyl, -(C 8 -C ⁇ 4 )tricycloalkenyl, -(3- to 7- membered)heterocycle, or -(7- to 10-membered)bicycloheterocycle, each of which is unsubstituted or substituted with one or more R 3 groups, or
  • A is -C(O)-.
  • n is 1; Ri is substituted at the 3-position ofthe pyridyl ring and is -CH 3 , -halo, -NO 2 , -OH or -CN; and R 2 is -phenyl, -naphthyl or - (C ⁇ 4 )aryl, each which is unsubstituted or substituted with one or more R 5 groups.
  • n is 1; Ri is substituted at the 3-position ofthe pyridyl ring and is -CH , -halo, -NO 2j -OH or -CN; and R 2 is -(C ⁇ -C ⁇ o)alkyl, which is unsubstituted or substituted with one or more R 3 groups.
  • A is -C(O)-; n is 1; Ri is substituted at the 3- position ofthe pyridyl ring and is -CH 3 , -halo, -NO 2 , -OH, or -CN; and R 2 is -phenyl, -naphthyl, -(C ⁇ 4 )aryl or -(5- to 10-membered)heteroaryl, each which is unsubstituted or substituted with one or more R 5 groups.
  • A is -C(O)-; n is 1; Ri is -NO 2 and substituted at the 3-position ofthe pyridyl ring; and R 2 is phenyl, which is unsubstituted or substituted with one or more R 5 groups.
  • A is -C(O)-, n is 1, Ri is -NO 2 and substituted at the 3-position ofthe pyridyl ring, and R 2 is unsubstituted phenyl.
  • A is -C(O)-, n is 1, Ri is -CH , and R 2 is unsubstituted phenyl.
  • A is -C(O)-; n is 1 ; Ri is -NO 2 , -halo or -CN, each of which is substituted at the 3-position ofthe pyridyl ring; and R 2 is unsubstituted phenyl.
  • n 0.
  • m is 1. In another embodiment each R 6 is -CH 3 .
  • the present invention also encompasses compounds having the formula (I), and pharmaceutically acceptable salts thereof, wherein:
  • A is -C(O)-, -C(S)-, -CH 2 -, -CH(C ⁇ -C 4 alkyl)- or -C(C ⁇ -C 4 alkyl)(C ⁇ -C 4 alkyl)-; each R t is independently -(C,-C 3 )alkyl, -halo, -NO 2 , -OH or -CN; is 0 or 1 ; n is an integer from 1-4;
  • R 2 is -H, -(C ⁇ _C ⁇ o)alkyl, -(C 2 -C ⁇ o)alkenyl, -(C 2 --C ⁇ 0 )alkynyl, -(C 3 - C ⁇ o)cycloalkyl, -(C 8 -C ⁇ 4 )bicycloalkyl, -(C 8 -C ⁇ 4 )tricycloalkyl, -(C 5 -C ⁇ o)cycloalkenyl, -(C 8 -C ⁇ )bicycloalkenyl or -(C 8 -C ⁇ 4 )tricycloalkenyl, each of which is unsubstituted or substituted with one or more R 3 groups, or
  • the present invention also encompasses compounds having the formula (I), and pharmaceutically acceptable salts thereof, wherein: A is -C(O)-, -C(S)-, -CH 2 -, -CH(C,-C 4 alkyl)- or -C(C ⁇ -C 4 alkyl)(C ⁇ -C 4 alkyl)-; each Ri is independently -(C ⁇ -C 3 )alkyl, -halo, -NO 2 , -OH or -CN; m is 0 or 1; n is an integer from 1-4; R 2 is -(C ⁇ -C ⁇ o)alkyl, -(C 2 -C ⁇ o)alkenyl, -(C 2 -C ⁇ 0 )alkynyl, -(C 3 -
  • A is -CH 2 -, n is 1, Ri is -NO 2 and substituted at the 3-position ofthe pyridyl ring, and R 2 is unsubstituted phenyl.
  • A is -CH 2 -, n is 2, an Ri group is an -NO 2 substituted at the 3-position ofthe pyridyl ring and the other Ri group is a -OH substituted at the 6-position ofthe pyridyl ring, and R 2 is unsubstituted phenyl.
  • A is -CH 2 -, n is 1, Ri is -CN and substituted at the 3-position ofthe pyridyl ring, and R 2 is unsubstituted phenyl.
  • A is -CH 2 -, n is 1, R, is -Cl and substituted at the 3-position ofthe pyridyl ring, and R 2 is unsubstituted phenyl.
  • n 0.
  • m is 1. In another embodiment each Re is -CH 3 .
  • each Re can be on any carbon ofthe piperazine ring.
  • the Piperazine Compounds have only one Re group, and that Re group is attached to a carbon atom adjacent to the nitrogen atom attached to the pyridinyl group.
  • the Piperazine Compound has only one Re group, and that R 6 group is attached to a carbon atom adjacent to the nitrogen atom attached to the A group.
  • two Re groups are on a single atom ofthe piperazine ring.
  • an Re group is attached to a carbon atom adjacent to the nitrogen atom attached to the pyridinyl group and another R 6 group is attached to a carbon atom adjacent to the nitrogen atom attached to the A group.
  • the Piperazine Compound has two Re groups, each being attached to a different carbon atom adjacent to a nitrogen atom attached to the pyridinyl group, hi another embodiment, the Piperazine Compound has two Re groups, each being attached to a different carbon atom adjacent to a nitrogen atom attached to the A group.
  • the Piperazine Compound has one or two Re groups
  • the carbon atom to which an Re group is attached has the (R configuration.
  • the Piperazine Compound has one or two Re groups
  • the carbon atom to which the Re group is attached has the (S) configuration
  • the Piperazine Compound has one or two Re groups, and at least one ofthe carbon atoms to which an Re group is attached has the (R) configuration.
  • the Piperazine Compound has one or two Re groups, and at least one ofthe carbon atoms to which an Re group is attached has the (S) configuration.
  • the Piperazine Compound has one or two Re groups, an R 6 group is attached to a carbon atom adjacent to a nitrogen atom attached to the pyridinyl group, and the carbon to which the R 6 group is attached is in the (R) configuration.
  • the Piperazine Compound has one or two Re groups, an Re group is attached to a carbon atom adjacent to a nitrogen attached to the pyridinyl group, the carbon to which the Re group is attached is in the (R) configuration, and Re is -(C ⁇ -C 3 )alkyl.
  • the Piperazine Compound has one or two Re groups, an Re group is attached to a carbon atom adjacent to a nitrogen attached to the pyridinyl group, the carbon to which the R ⁇ group is attached is in the (R) configuration, and Re is -CH 3 .
  • the Piperazine Compound has one or two Re groups, an ⁇ group is attached to a carbon atom adjacent to a nitrogen attached to the pyridinyl group, the carbon to which the Re group is attached is in the (R) configuration, and Re is -CH 2 OH.
  • the Piperazine Compound has one or two Re groups, an R 6 group is attached to a carbon atom adjacent to a nitrogen attached to the pyridinyl group, the carbon to which the Re group is attached is in the (R) configuration, and Re is -CH 2 CH 3 .
  • the Piperazine Compound has one or two Re groups, an Re group is attached to a carbon atom adjacent to a nitrogen atom attached to the A group, and the carbon to which the Re group is attached is in the (R) configuration.
  • the Piperazine Compound has one or two Re groups, an Re group is attached to a carbon atom adjacent to a nitrogen attached to the A group, the carbon to which the Re group is attached is in the (R) configuration, and Re is -(Ci- C )alkyl.
  • the Piperazine Compound has one or two Re groups, an Re group is attached to a carbon atom adjacent to a nitrogen attached to the A group, the carbon to which the Re group is attached is in the (R) configuration, and Re is -CH 3 .
  • the Piperazine Compound has one or two Re groups, an Re group is attached to a carbon atom adjacent to a nitrogen attached to the A group, the carbon to which the Re group is attached is in the (R) configuration, and Re is -CH 2 OH.
  • the Piperazine Compound has one or two Re groups, an R group is attached to a carbon atom adjacent to a nitrogen attached to the A group, the carbon to which the Re group is attached is in the (R) configuration, and Re is -CH 2 CH 3 .
  • the Piperazine Compound has one or two Re groups, an Re group is attached to a carbon atom adjacent to a nitrogen atom attached to the pyridinyl group, and the carbon to which the R 6 group is attached is in the (S) configuration.
  • the Piperazine Compound has one or two Re groups, an Re group is attached to a carbon atom adjacent to a nitrogen attached to the pyridinyl group, the carbon to which the Re group is attached is in the (S) configuration, and Re is -(C ⁇ -C 3 )alkyl.
  • the Piperazine Compound has one or two Re groups, an Re group is attached to a carbon atom adjacent to a nitrogen attached to the pyridinyl group, the carbon to which the Re group is attached is in the (S) configuration, and Re is -CH 3 .
  • the Piperazine Compound has one or two Re groups, an R 6 group is attached to a carbon atom adjacent to a nitrogen attached to the pyridinyl group, the carbon to which the Re group is attached is in the (S) configuration, and Re is -CH 2 OH.
  • the Piperazine Compound has one or two Re groups, an Re group is attached to a carbon atom adjacent to a nitrogen attached to the pyridinyl group, the carbon to which the Re group is attached is in the (S) configuration, and R ⁇ is -CH2CH 3 .
  • the Piperazine Compound has one or two Re groups, an R ⁇ group is attached to a carbon atom adjacent to a nitrogen atom attached to the A group, and the carbon to which the Re group is attached is in the (S) configuration.
  • the Piperazine Compound has one or two Re groups, an Re group is attached to a carbon atom adjacent to a nitrogen attached to the A group, the carbon to which the Re group is attached is in the (S) configuration, and Re is -(C ⁇ -C 3 )alkyl.
  • the Piperazine Compound has one or two R ⁇ groups, an Re group is attached to a carbon atom adjacent to a nitrogen attached to the A group, the carbon to which the Re group is attached is in the (S) configuration, and Re is -CH 3 .
  • the Piperazine Compound has one or two R 6 groups, an Re group is attached to a carbon atom adjacent to a nitrogen attached to the A group, the carbon to which the Re group is attached is in the (S) configuration, and R 6 is -CH 2 OH.
  • the Piperazine Compound has one or two Re groups, a Re group is attached to a carbon atom adjacent to a nitrogen attached to the A group, the carbon to which the Re group is attached is in the (S) configuration, and R ⁇ is -CH 2 CH 3 .
  • the Piperazine Compound has only one Re group, the Re group is attached to a carbon atom adjacent to a nitrogen atom attached to the pyridinyl group, and the carbon to which the Re group is attached is in the (R) configuration.
  • the Piperazine Compound has only one Re group, the Re group is attached to a carbon atom adjacent to a nitrogen attached to the pyridinyl group, the carbon to which the Re group is attached is in the (R) configuration, and Re is -(d-C 3 )alkyl.
  • the Piperazine Compound has only one Re group, the Re group is attached to a carbon atom adjacent to a nitrogen attached to the pyridinyl group, the carbon to which the Re group is attached is in the (R) configuration, and Re is -CH 3 .
  • the Piperazine Compound has only one Re group, the Re group is attached to a carbon atom adjacent to a nitrogen attached to the pyridinyl group, the carbon to which the Re group is attached is in the (R) configuration, and Re is -CH 2 OH.
  • the Piperazine Compound has only one Re group, the R ⁇ group is attached to a carbon atom adjacent to a nitrogen attached to the pyridinyl group, the carbon to which the Re group is attached is in the (R) configuration, and R 6 is -CH 2 CH 3 .
  • the Piperazine Compound has only one R 6 group, the Re group is attached to a carbon atom adjacent to a nitrogen atom attached to the A group, and the carbon to which the Re group is attached is in the (R) configuration.
  • the Piperazine Compound has only one Re group, the Re group is attached to a carbon atom adjacent to a nitrogen attached to the A group, the carbon to which the Re group is attached is in the (R) configuration, and Re is -(C ⁇ -C 3 )alkyl.
  • the Piperazine Compound has only one Re group, the Re group is attached to a carbon atom adjacent to a nitrogen attached to the A group, the carbon to which the R 6 group is attached is in the (R) configuration, and Re is -CH 3 .
  • the Piperazine Compound has only one Re group, the Re group is attached to a carbon atom adjacent to a nitrogen attached to the A group, the carbon to which the Re group is attached is in the (R) configuration, and Re is -CH 2 OH.
  • the Piperazine Compound has only one Re group, the Re group is attached to a carbon atom adjacent to a nitrogen attached to the A group, the carbon to which the Re group is attached is in the (R) configuration, and Re is -CH 2 CH 3 .
  • the Piperazine Compound has only one R ⁇ group, the Re group is attached to a carbon atom adjacent to a nitrogen atom attached to the pyridinyl group, and the carbon to which the Re group is attached is in the (S) configuration.
  • the Piperazine Compound has only one Re group, the Re group is attached to a carbon atom adjacent to a nitrogen attached to the pyridinyl group, the carbon to which the R ⁇ group is attached is in the (S) configuration, and Re is -(C ⁇ -C 3 )alkyl.
  • the Piperazine Compound has only one Re group, the Re group is attached to a carbon atom adjacent to a nitrogen attached to the pyridinyl group, the carbon to which the Re group is attached is in the (S) configuration, and R ⁇ is -CH 3 .
  • the Piperazine Compound has only one Re group, the Re group is attached to a carbon atom adjacent to a nitrogen attached to the pyridinyl group, the carbon to which the Re group is attached is in the (S) configuration, and Re is - CH 2 OH.
  • the Piperazine Compound has only one R ⁇ group, the Re group is attached to a carbon atom adjacent to a nitrogen attached to the pyridinyl group, the carbon to which the R 6 group is attached is in the (S) configuration, and Re is -CH 2 CH 3 .
  • the Piperazine Compound has only one Re group, the R 6 group is attached to a carbon atom adjacent to a nitrogen atom attached to the A group, and the carbon to which the Re group is attached is in the (S) configuration.
  • the Piperazine Compound has only one Re group, the Re group is attached to a carbon atom adjacent to a nitrogen attached to the A group, the carbon to which the Re group is attached is in the (S) configuration, and Re is -(C ⁇ -C 3 )alkyl.
  • the Piperazine Compound has only one R 6 group, the Re group is attached to a carbon atom adjacent to a nitrogen attached to the A group, the carbon to which the Re group is attached is in the (S) configuration, and Re is -CH 3 .
  • the Piperazine Compound has only one Re group, the Re group is attached to a carbon atom adjacent to a nitrogen attached to the A group, the carbon to which the Re group is attached is in the (S) configuration, and Re is -CH 2 OH.
  • the Piperazine Compound has only one Re group, the R group is attached to a carbon atom adjacent to a nitrogen attached to the A group, the carbon to which the Re group is attached is in the (R) configuration, and Re is -CH 2 CH 3 .
  • the Re group is attached to a carbon atom adjacent to a nitrogen attached to the A group. In another embodiment, the Re group is attached to a carbon atom adjacent to a nitrogen attached to the A group and the Re group is a -CH 3 . In another embodiment, the Re group is attached to a carbon atom adjacent to a nitrogen attached to the A group and the R ⁇ group is a -CH 2 CH 3 . In another embodiment, the Re group is attached to a carbon atom adjacent to a nitrogen attached to the A group and the carbon to which the Re group is attached is in the (R) configuration.
  • the Re group is attached to a carbon atom adjacent to a nitrogen attached to the A group, the carbon to which the R 6 group is attached is in the (R) configuration, and the Re group is a -CH 3 .
  • the Re group is attached to a carbon atom adjacent to a nitrogen attached to the A group, the carbon to which the Re group is attached is in the (R) configuration, and the R ⁇ group is a - CH CH 3 .
  • A is -C(O)-; n is 2; an R, is substituted at the 4- position ofthe pyridinyl ring (denoted hereinafter for convenience as “Ri' ”) and is -CH 3 , -OCH 3 or -halo; the other Rj is substituted at the 6-position ofthe pyridinyl ring (denoted hereinafter for convenience as “Ri”' ”) and is -H or -CH 3 ;
  • R 2 is -phenyl or -pyridyl, each which is unsubstituted or substituted with one or more R 5 or R 5 ' groups, as described above, and R 6 is -H, -CH 3 or -CH 2 OH and is attached to a carbon atom adjacent to the nitrogen atom attached to the A group.
  • A is -C(O)-; n is 2; Ri' is -CH 3 , -OCH 3 or -Cl; Ri'" is -H or -CH 3 ; R 2 is -phenyl or -pyridyl, each which is unsubstituted or substituted with one or more R 5 or R 5 ' groups, as described above; and R is -H, -CH 3 or -CH 2 OH and is attached to a carbon atom adjacent to the nitrogen atom attached to the A group.
  • A is -C(O)-; n is 2; Ri' is -CH 3 , -OCH 3 or -halo; Ri'" is -H or -CH 3 ; R 2 is -phenyl or -pyridyl, each which is unsubstituted or substituted with one or more R or R 5 ' groups, as described above, selected from -halo and -CH 3 ; and R 6 is -H, -CH 3 or -CH 2 OH and is attached to a carbon atom adjacent to the nitrogen atom attached to the A group.
  • A is -C(O)-; n is 2; Ri' is -CH 3 , -OCH 3 or -Cl; Ri'" is -H or -CH 3 ; R 2 is -phenyl or -pyridyl, each which is unsubstituted or substituted with one or more R 5 or R 5 ' groups, as described above, selected from -F, -Cl and -CH 3 ; and Re is -H, -CH 3 or -CH 2 OH and is attached to a carbon atom adjacent to the nitrogen atom attached to the A group.
  • A is -C(O)-; n is 2; Ri' is -CH 3 , -OCH 3 or -Cl; Ri'" is -H or -CH 3 ; R 2 is -phenyl which is unsubstituted or substituted with one R 5 group that is para to the phenyl group's point of attachment to the triple bond and is selected from -F and -OCH 3 ; and Re is -H, -CH or -CH 2 OH and is attached to a carbon atom adjacent to the nitrogen atom attached to the A group.
  • A is -C(O)-; n is 2; Ri' is -CH 3 , -OCH 3 or -Cl; Ri'" is -H or -CH 3 ; R 2 is -pyridyl which is attached at its 2-position and is unsubstituted or substituted with one R 5 ' group at the 5-position ofthe -pyridyl group's point of attachment to the triple bond and is selected from -F, -Cl and -CH 3 ; and Re is -H, -CH 3 or -CH 2 OH and is attached to a carbon atom adjacent to the nitrogen atom attached to the A group.
  • A is -C(O)-; n is 2; Ri' is -CH 3 , -OCH 3 or -Cl; Ri'" is -H or -CH 3 ; R2 is -pyridyl which is attached at its 3-position and is unsubstituted or substituted with one R 5 ' group at the 6-position ofthe -pyridyl group's point of attachment to the triple bond and is selected from -F, -Cl and -CH 3 ; and Re is -H, -CH 3 or i -CH 2 OH and is attached to a carbon atom adjacent to the nitrogen atom attached to the A group.
  • the present invention also encompasses compounds of Formula (la):
  • A is -C(O)-, -C(S)-, -CH(C,-C 4 alkyl)- or -C(C * ⁇ -C 4 alkyl)(d-C 4 alkyl)-; each Ri is independently -(C ⁇ -C 3 )alkyl, -halo, -NO 2 , -OH or -CN; n is an integer from 1-4; R 2 is -H, -(C ⁇ -C ⁇ o)alkyl, -(C 2 -d 0 )alkenyl, -(C 2 -C ⁇ o)alkynyl,
  • the present invention also encompasses compounds having the formula (la), and pharmaceutically acceptable salts thereof, wherein:
  • A is -C(O)-, -C(S)-, -CH(C ⁇ -C 4 alkyl)- or -C(C ⁇ -C 4 alkyl)(C C 4 alkyl)-; each Ri is independently -(C ⁇ -C 3 )alkyl, -halo, -NO 2 , -OH or -CN; n is an integer from 1-4;
  • R 2 is -(C ⁇ -C ⁇ o)alkyl, -(C 2 -C 10 )alkenyl, -(C 2 -C ⁇ 0 )alkynyl, -(C 3 -C ⁇ o)cycloalkyl, -(C 8 -C ⁇ 4 )bicycloalkyl, -(C 8 -C ⁇ 4 )tricycloalkyl, -(C 5 -
  • R 2 is -phenyl, -naphthyl or -(C ⁇ 4 )aryl, each of which is unsubstituted or substituted with one or more R groups, or
  • each R 5 ' is independently -(C]-C 6 )alkyl, -(C 2 -C 6 )alkenyl, -(C 2 -Ce)alkynyl, -(C 3 -C 8 )cycloalkyl, -(C 5 -C 8 )cycloalkenyl, -phenyl, -(3- to 5-membered)heterocycle, -C(halo) 3 , -CH(
  • A is -C(O)-; n is 1; Ri is substituted at the 3- position ofthe pyridyl ring and is -CH 3 , -halo, -NO 2 , -OH, or -CN; and R 2 is -phenyl, -naphthyl, -(C ⁇ 4 )aryl or -(5- to 10-membered)heteroaryl, each which is unsubstituted or substituted with one or more R 5 groups.
  • A is -C(O)-; n is 1; Ri is -NO 2 and substituted at the 3-position ofthe pyridyl ring; and R2 is phenyl, which is unsubstituted or substituted with one or more R 5 groups.
  • A is -C(O)-, n is 1, Rj is -NO2 and substituted at the 3-position ofthe pyridyl ring, and R 2 is unsubstituted phenyl.
  • A is -C(O)-, n is 1, Ri is -CH 3 , and R 2 is unsubstituted phenyl.
  • A is -C(O)-; n is 1; R ⁇ is -NO2, -halo or -CN, each of which is substituted at the 3-position ofthe pyridyl ring; and R 2 is unsubstituted phenyl.
  • the present invention also encompasses compounds having the formula
  • A is -C(O)-, -C(S)-, -CH 2 -, -CH(C ⁇ -C 4 alkyl)- or -C(C ⁇ -C 4 alkyl) (C ⁇ -C 4 alkyl)-; each Ri is independently -(C ⁇ -C 3 )alkyl, -halo, -NO 2 , -OH or -CN; n is an integer from 1-4;
  • R 2 is -H, -(d-C ⁇ o)alkyl, -(C 2 -C 10 )alkenyl, -(C 2 -C 10 )alkynyl, -(C 3 - C ⁇ o)cycloalkyl, -(C 8 -C ⁇ 4 )bicycloalkyl, -(C 8 -C )tricycloalkyl, -(C -C ⁇ o)cycloalkenyl, -(C 8 -C ⁇ 4 )bicycloalkenyl or -(C 8 -C ⁇ )tricycloalkenyl, each of which is unsubstituted or substituted with one or more R 3 groups, or R 2 is -phenyl, -naphthyl or -(C ⁇ )aryl, each of which is unsubstituted or substituted with one or more R 5 groups; each R 3 is independently -CN, -OH, -halo, -
  • each R is independently -H, -(C ⁇ -C 6 )alkyl, -(C 2 -C 6 )alkenyl, -(C 2 - C 6 )alkynyl, -(C -C 8 )cycloalkyl, -(C 5 -C 8 )cycloalkenyl, -phenyl, -(3- to 5- membered)heterocycle, -C(halo) or -CH(halo) 2 ; and each R 5 is independently -(C ⁇ -Ce)alkyl, -(C 2 -Ce)alkenyl, -(C 2 -C 6 )alkynyl, -(C -C 8 )cycloalkyl, -(C 5 -C 8 )cycloalkenyl, -phenyl, -(3- to 5-membered)heterocycle, -C(halo) 3 , -CH(halo) 2
  • the present invention also encompasses compounds having the formula (la), and pharmaceutically acceptable salts thereof, wherein:
  • A is -C(O)-, -C(S)-, -CH 2 -, -CH(C ⁇ -C 4 alkyl)- or -C(d-C 4 alkyl)(C ⁇ -C 4 alkyl)-; each Ri is independently -(C ⁇ -C 3 )alkyl, -halo, -NO2, -OH or -CN; n is an integer from 1-4;
  • R 2 is -(d-C,o)alkyl, -(C 2 -C ⁇ 0 )alkenyl, -(C 2 -C 10 )alkynyl, -(C 3 - C ⁇ o)cycloalkyl, -(C 8 -Ci 4 )bicycloalkyl, -(C 8 -C ⁇ 4 )tricycloalkyl, -(C 5 -C ⁇ o)cycloalkenyl, -(C 8 -C ⁇ 4 )bicycloalkenyl or -(C 8 -Ci 4 )tricycloalkenyl, each of which is unsubstituted or substituted with one or more R 3 groups, or
  • A is -CH 2 -, n is 1, Ri is -NO 2 and substituted at the 3-position ofthe pyridyl ring, and R 2 is unsubstituted phenyl.
  • A is -CH 2 -, n is 2, Ri is an -NO 2 substituted at the 3-position ofthe pyridyl ring, Ri'" is a -OH and R 2 is unsubstituted phenyl.
  • A is -CH 2 -, n is 1 , Ri is -CN and substituted at the 3-position ofthe pyridyl ring, and R 2 is unsubstituted phenyl.
  • A is -CH 2 -, n is 1, Ri is -Cl and substituted at the 3-position ofthe pyridyl ring, and R 2 is unsubstituted phenyl.
  • the compound has a -CH 3 at a position para to the 6-membered ring's point of attachment to the triple bond; the compound has a -CF 3 at a position para to the 6-membered ring's point of attachment to the triple bond; the compound has a -F at a position para to the 6-membered ring's point of attachment to the triple bond; the compound has a -Cl at a position para to the 6- membered ring's point of attachment to the triple bond; the compound has a -NO 2 at a position para to the 6-membered ring's point of attachment to the triple bond; the compound has a -C(O)CH 3 at a position para to the 6-membered ring's point of attachment to the triple bond; the compound has a -C(CH 3 ) at a position para to the
  • -(C ⁇ -C ⁇ o)alkyl means a saturated straight chain or branched non-cyclic hydrocarbon having from 1 to 10 carbon atoms.
  • Representative saturated straight chain -(C ⁇ -C ⁇ o)alkyls include -methyl, -ethyl, -n-propyl, -n-butyl, -n-pentyl, -n-hexyl, -n-heptyl, -n-octyl, -n-nonyl, and -n-decyl.
  • Representative saturated branched -(Ci- C ⁇ o)alkyls include -isopropyl, -sec-butyl, -isobutyl, -tert-butyl, -isopentyl, -2-methylbutyl, -3-methylbutyl, -2,2-dimethylbutyl, -2,3-dimethylbutyl, -2-methylpentyl, -3-methylpentyl, -4-methylpentyl, -2-methylhexyl, -3-methylhexyl, -4-methylhexyl, -5-methylhexyl, -2,3-dimethylbutyl, -2,3-dimethylpentyl, -2,4-dimethylpentyl, -2,3-dimethylhexyl, -2,4-dimethylhexyl, -2,3-dimethylhexyl, -2,4-dimethylhexyl,
  • -(C ⁇ -C 6 )alkyl means a saturated straight chain or branched non-cyclic hydrocarbon having from 1 to 6 carbon atoms.
  • Representative saturated straight chain -(C ⁇ -C 6 )alkyls include -methyl, -ethyl, -n-propyl, -n-butyl, -n-pentyl, and -n-hexyl.
  • saturated branched -( -C ⁇ alkyls include -isopropyl, -sec-butyl, -isobutyl, -tert-butyl, -isopentyl, -2-methylbutyl, -3-methylbutyl, -2,2-dimethylbutyl, -2,3-dimethylbutyl, -2-methylpentyl, -3-methylpentyl, -4-methylpentyl and the like.
  • "-(C ⁇ -C 4 )alkyl” means a saturated straight chain or branched non-cyclic hydrocarbon having from 1 to 4 carbon atoms.
  • Representative saturated straight chain -(C ⁇ -C 4 )alkyls include -methyl, -ethyl, -n-propyl, and -n-butyl.
  • Representative saturated branched -(C ⁇ . -C 4 )alkyls include -isopropyl, -sec-butyl, -isobutyl, and -tert-butyl.
  • -(C ⁇ -C 3 )alkyl means a saturated straight chain or branched non-cyclic hydrocarbon having from 1 to 3 carbon atoms.
  • Representative saturated straight chain -(C ⁇ -C 3 )alkyls include -methyl, -ethyl, and -n-propyl.
  • a representative saturated branched -(C ⁇ -C 3 )alkyl is -isopropyl.
  • -(C 2 -C ⁇ o)alkenyl means a straight chain or branched nonfrom 2 to 10 carbon atoms and including at least one carbon-carbon double bond.
  • -(C 2 -C 6 )alkenyl means a straight chain or branched non-cyclic hydrocarbon having from 2 to 6 carbon atoms and including at least one carbon-carbon double bond.
  • Representative straight chain and branched (C 2 -C 6 )alkenyls include -vinyl, -allyl, -1-butenyl, -2-butenyl, -isobutylenyl, -1-pentenyl, -2-pentenyl, -3-methyl- 1-butenyl, -2-methyl-2-butenyl, -2,3-dimethyl-2-butenyl, -1-hexenyl, -2-hexenyl, -3-hexenyl and the like.
  • -(C 2 -C ⁇ o)alkynyl means a straight chain or branched non-cyclic hydrocarbon having from 2 to 10 carbon atoms and including at lease one carbon-carbon triple bond.
  • Representative straight chain and branched -(C 2 -C ⁇ o)alkynyls include - acetylenyl, -propynyl, -1-butynyl, -2-butynyl, -1-pentynyl, -2-pentynyl, -3-methyl-l- butynyl, -4-pentynyl, -1-hexynyl, -2-hexynyl, -5-hexynyl, -1-heptynyl, -2-heptynyl, -6-heptynyl, -1-octynyl, -2-octynyl, -7-octynyl, -1-
  • -(C 2 -C 6 )alkynyl means a straight chain or branched non-cyclic hydrocarbon having from 2 to 6 carbon atoms and including at lease one carbon-carbon triple bond.
  • Representative straight chain and branched (C 2 -C 6 )alkynyls include -acetylenyl, -propynyl, -1-butynyl, -2-butynyl, -1-pentynyl, -2-pentynyl, -3-methyl-l- butynyl, -4-pentynyl, -1-hexynyl, -2-hexynyl, -5-hexynyl and the like.
  • -(C 3 -C-io)cycloalkyl means a saturated cyclic hydrocarbon having from 3 to 10 carbon atoms.
  • Representative (C 3 -C*o)cycloalkyls include -cyclopropyl, -cyclobutyl, -cyclopentyl, -cyclohexyl, -cycloheptyl, -cyclooctyl, -cyclononyl, and -cyclodecyl.
  • -(C -C 8 )cycloalkyl means a saturated cyclic hydrocarbon having from 3 to 8 carbon atoms.
  • Representative (C -C 8 )cycloalkyls include -cyclopropyl, -cyclobutyl, -cyclopentyl, -cyclohexyl, -cycloheptyl, and -cyclooctyl.
  • -(C 8 -C ⁇ )bicycloalkyl means a bi-cyclic hydrocarbon ring system having from 8 to 14 carbon atoms and at least one saturated cyclic alkyl ring.
  • Representative -(C 8 -C ⁇ 4 )bicycloalkyls include -indanyl, -1,2,3,4-tetrahydronaphthyl, -5,6,7,8-tetrahydronaphthyl, -perhydronaphthyl and the like.
  • -(C 8 -C ⁇ 4 )tricycloalkyl means a tri-cyclic hydrocarbon ring system having from 8 to 14 carbon atoms and at least one saturated ring.
  • Representative -(C 8 - C ⁇ 4 )tricycloalkyls include -pyrenyl, -1,2,3,4-tetrahydroanthracenyl, -perhydroanthracenyl, -aceanthreneyl, -1,2,3,4-tetrahydropenanthrenyl, -5,6,7,8-tetrahydrophenanthrenyl, -perhydrophenanthrenyl and the like.
  • "-(C 5 -C ⁇ o)cycloalkenyl” means a cyclic non-aromatic hydrocarbon having at least one carbon-carbon double bond in the cyclic system and from 5 to 10 carbon atoms.
  • Representative (C -C ⁇ o)cycloalkenyls include -cyclopentenyl, -cyclopentadienyl, -cyclohexenyl, -cyclohexadienyl,-cycloheptenyl, -cycloheptadienyl, -cycloheptatrienyl, -cyclooctenyl, -cyclooctadienyl, -cyclooctatrienyl, -cyclooctatetraenyl, -cyclononenyl, -cyclononadienyl, -cyclodecenyl, -cyclodecadienyl and the like.
  • -(C 5 -C 8 )cycloalkenyl means a cyclic non-aromatic hydrocarbon having at least one carbon-carbon double bond in the cyclic system and from 5 to 8 carbon atoms.
  • Representative (C 5 -C 8 )cycloalkenyls include -cyclopentenyl, -cyclopentadienyl, -cyclohexenyl, -cyclohexadienyl, -cycloheptenyl, -cycloheptadienyl, -cycloheptatrienyl, -cyclooctenyl, -cyclooctadienyl, -cyclooctatrienyl, -cyclooctatetraenyl and the like.
  • -(C 8 -C ⁇ 4 )bicycloalkenyl means a bi-cyclic hydrocarbon ring system having at least one carbon-carbon double bond in each ring and from 8 to 14 carbon atoms.
  • Representative -(C 8 -C ⁇ 4 )bicycloalkenyls include -indenyl, -pentalenyl, -naphthalenyl, -azulenyl, -heptalenyl, -1,2,7,8-tetrahydronaphthalenyl and the like.
  • -(C 8 -C ⁇ 4 )tricycloalkenyl means a tri-cyclic hydrocarbon ring system having at least one carbon-carbon double bond in each ring and from 8 to 14 carbon atoms.
  • Representative -(C 8 -C ⁇ 4 )tricycloalkenyls include -anthracenyl, -phenanthrenyl, -phenalenyl, -acenaphthalenyl, - ⁇ s-indacenyl, -.s-indacenyl and the like.
  • -(5- to 10-membered)heteroaryl means an aromatic heterocycle ring of 5 to 10 members, including both mono- and bicyclic ring systems, where at least one carbon atom of one or both ofthe rings is replaced with a heteroatom independently selected from nitrogen, oxygen, and sulfur.
  • One or both ofthe -(5- to 10- membered)heteroaryl's rings contain at least one carbon atom.
  • Representative (5- to 10- membered)heteroaryls include pyridyl, furyl, benzofuranyl, thiophenyl, benzothiophenyl, quinolinyl, pyrrolyl, indolyl, oxazolyl, benzoxazolyl, imidazolyl, benzimidazolyl, thiazolyl, benzothiazolyl, isoxazolyl, pyrazolyl, isothiazolyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, cinnolinyl, phthalazinyl, quinazolinyl and the like.
  • a 3- or a 4-membered -(3- to 7- membered)heterocycle can contain up to 3 heteroatoms
  • a 5-membered -(3- to 7- membered)heterocycle can contain up to 4 heteroatoms
  • a 6-membered -(3- to 7- membered)heterocycle can contain up to 6 heteroatoms
  • a 7-membered -(3- to 7- membered)heterocycle can contain up to 7 heteroatoms.
  • Each heteroatom is independently selected from nitrogen, which can be quatemized; oxygen; and sulfur, including sulfoxide and sulfone.
  • the -(3- to 7-membered)heterocycle can be attached via any heteroatom or carbon atom.
  • Representative -(3- to 7-membered)heterocycles include pyridyl, furyl, thiophenyl, pyrrolyl, oxazolyl, imidazolyl, thiazolyl, isoxazolyl, pyrazolyl, isothiazolyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, morpholinyl, pyrrolidinonyl, pyrrolidinyl, piperidinyl, piperazinyl, hydantoinyl, valerolactamyl, oxiranyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydropyrindinyl, tetrahydropyrimidinyl, te
  • -(3- to 5-membered)heterocycle or "-(3- to 5-membered)heterocyclo” means a 3- to 5-membered monocyclic heterocyclic ring which is either saturated, unsaturated, non-aromatic or aromatic.
  • a 3- or 4-membered -(3- to 5- membered)heterocycle can contain up to 3 heteroatoms and a 5-membered -(3- to 5- membered)heterocycle can contain up to 4 heteroatoms.
  • Each heteroatom is independently selected from nitrogen, which can be quatemized; oxygen; and sulfur, including sulfoxide and sulfone.
  • the -(3- to 5-membered)heterocycle can be attached via any heteroatom or carbon atom.
  • Representative -(3- to 5-membered)heterocycles include furyl, thiophenyl, pyrrolyl, oxazolyl, imidazolyl, thiazolyl, isoxazolyl, pyrazolyl, isothiazolyl, triazinyl, pyrrolidinonyl, pyrrolidinyl, hydantoinyl, oxiranyl, oxetanyl, tetrahydrofuranyl, tetrahydrothiophenyl and the like.
  • “-(7- to 10-membered)bicycloheterocycle” or “-(7- to 10- membered)bicycloheterocyclo” means a 7- to 10-membered bicyclic, heterocyclic ring having a saturated, unsaturated, non-aromatic or aromatic group.
  • a -(7- to 10- membered)bicycloheterocycle contains from 1 to 4 heteroatoms independently selected from nitrogen, which can be quatemized; oxygen; and sulfur, including sulfoxide and sulfone.
  • the (7- to 10-membered)bicycloheterocycle can be attached via any heteroatom or carbon atom.
  • Representative -(7- to 10-membered)bicycloheterocycles include -quinolinyl, -isoquinolinyl, -chromonyl, -coumarinyl, -indolyl, -indolizinyl, -benzo[b]furanyl, -benzo[b]thiophenyl, -indazolyl, -purinyl, -4H-quinolizinyl, -isoquinolyl, -quinolyl, -phthalazinyl, -naphthyridinyl, -carbazolyl, - ⁇ -carbolinyl, 1,3-benzodioxole and the like.
  • -(C ⁇ 4 )aryl means a 14-membered aromatic carbocyclic moiety such as anthryl and phenanthryl.
  • -CH 2 (halo) means a methyl group wherein one ofthe hydrogens ofthe methyl group has been replaced with a halogen.
  • Representative -CH 2 (halo) groups include -CH 2 F, -CH 2 C1, -CH 2 Br and -CH 2 I.
  • -CH(halo) 2 means a methyl group wherein two ofthe hydrogens ofthe methyl group have been replaced with a halogen.
  • Representative -CH(halo) 2 groups include -CFfF 2 , -CHC1 2 , -CHBr 2 , CHBrCl, CHC1I and -CHI 2 .
  • -C(halo) 3 means a methyl group wherein each ofthe hydrogens ofthe methyl group has been replaced with a halogen.
  • Representative -C(halo) 3 groups include -CF 3 , -CF 2 C1, -CC1 3 , -CBr 3 , -CFBr 2 and -CI 3 .
  • "-Halogen” or “-halo” means -F, -Cl, -Br or -I.
  • animal includes, but is not limited to, a cow, monkey, horse, sheep, pig, chicken, turkey, quail, cat, dog, mouse, rat, rabbit, guinea pig and human.
  • pharmaceutically acceptable salt is a salt formed from an acid and a basic nitrogen group of one ofthe Piperazine Compounds.
  • Illustrative salts include, but are not limited, to sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucuronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, ?-toluenesulfonate and pamoate (i.e., l,l'-methylene-bis-(2-hydroxy-3-naphthoate)) salts.
  • sulfate citrate, acetate, oxalate, chlor
  • Suitable bases include, but are not limited to, hydroxides of alkali metals such as sodium, potassium, and lithium; hydroxides of alkaline earth metal such as calcium and magnesium; hydroxides of other metals, such as aluminum and zinc; ammonia and organic amines, such as unsubstituted or hydroxy-substituted mono-, di- or trialkylamines; dicyclohexylamine; tributyl amine; pyridine; N-methyl-N-ethylamine; diethylamine; triethylamine; mono-, bis- or tris-(2-hydroxy-lower alkyl amines), such as mono-, bis- or tris-(2-hydroxyethyl)amine, 2-hydroxy-tert-butylamine or tris
  • phrases "effective amount" when used in connection with a Piperazine Compound means an amount effective for: (a) treating or preventing a Condition; or (b) inhibiting mGluR5 or mGluRl function in a cell.
  • phrases "effective amount" when used in connection with another therapeutic agent means an amount for providing the therapeutic effect ofthe therapeutic agent.
  • each of one or more ofthe first group's hydrogen atoms is replaced with a second group.
  • a first group is substituted with up to three second groups.
  • a first group is substituted with one or two second groups. In another embodiment, a first group is substituted with only one second group.
  • UI means urinary incontinence.
  • ALS means amyotrophic lateral sclerosis.
  • treatment of and “treating” includes the amelioration or cessation a Condition or a symptom thereof.
  • prevention of and “preventing” includes the avoidance of the onset of a Condition or a symptom thereof.
  • Piperazine Compounds can be made using conventional organic syntheses and/or by the following illustrative methods.
  • Piperazine Compounds can be obtained by reacting a compound of formula A with an alkyl iodide, R 2 I, at low temperature, e.g., about 0°C to about -78 °C, in the presence of lithium diisopropylamide (“LDA”) in hexamethylphosphoramide (“HMPA”) as shown below in Scheme A:
  • LDA lithium diisopropylamide
  • HMPA hexamethylphosphoramide
  • Piperazine Compounds can also be obtained by reacting a compound of formula A with an aryl iodide at room temperature in ethyl acetate in the presence of Pd(Ph 3 P) 2 OAc 2 , Cul, and Et 3 N, as shown below in Scheme B: Scheme B
  • the compound of formula A can be prepared by reacting a compound of formula B with propynoic acid in the presence of 1-hydroxybenzotriazolehydrate (“HOBT”) and 1,3-diisopro ⁇ ylcarbodiimide (“DIC”) as shown below in Scheme C:
  • HOBT 1-hydroxybenzotriazolehydrate
  • DIC 1,3-diisopro ⁇ ylcarbodiimide
  • the compound of formula A can also be prepared by reacting a compound of formula B with propynoyl chloride in the presence of tertiary amine, such as triethylamine, as shown below in Scheme D: Scheme D
  • the compound of formula B can be prepared by reacting a 2-halo- substituted pyridine of formula C with piperazine D in chloroform, in the presence of triethylamine (TEA), at a temperature of 50°C as shown below in Scheme E:
  • substituted 2-halo-pyridines C are commercially available or can be prepared by methods well known to those skilled in the art.
  • Piperazine Compounds containing R 2 groups other than the R 2 groups exemplified in Schemes A through D can be prepared using analogous methods. Certain Piperazine Compounds may have asymmetric centers and therefore exist in different enantiomeric and diastereomeric forms.
  • a Piperazine Compound can be in the form of an optical isomer or a diastereomer. Accordingly, the invention encompasses Piperazine Compounds and their uses as described herein in the form of their optical isomers, diasteriomers and mixtures thereof, including a racemic mixture.
  • Compound can be replaced by an isotope ofthe hydrogen, carbon or other atoms.
  • Such compounds which are encompassed by the present invention, are useful as research and diagnostic tools in metabolism pharmacokinetic studies and in binding assays.
  • the Piperazine Compounds are administered to an animal in need of treatment or prevention of a Condition.
  • an effective amount of a Piperazine Compound can be used to treat or prevent any condition treatable or preventable by inhibiting mGluR5.
  • conditions that are treatable or preventable by inhibiting mGluR5 include, but are not limited to, pain, an addictive disorder, Parkinson's disease, parkinsonism, anxiety, a pruritic condition, and psychosis.
  • an effective amount of a Piperazine Compound can be used to treat or prevent any condition treatable or preventable by inhibiting mGluRl.
  • conditions that are treatable or preventable by inhibiting mGluRl include, but are not limited to, pain, UI, an addictive disorder, Parkinson's disease, parkinsonism, anxiety, epilepsy, a seizure, stroke, a pruritic condition, psychosis, a cognitive disorder, a memory deficit, restricted brain function, Huntington's chorea, ALS, dementia, retinopathy, a muscle spasm, a migraine, vomiting, dyskinesia and depression.
  • the Piperazine Compounds can be used to treat or prevent acute or chronic pain.
  • Examples of pain treatable or preventable using the Piperazine Compounds include, but are not limited to, cancer pain, central pain, labor pain, myocardial infarction pain, pancreatic pain, colic pain, post-operative pain, headache pain, muscle pain, pain associated with intensive care, arthritic pain, neuropathic pain, and pain associated with a periodontal disease, including gingivitis and periodontitis.
  • the Piperazine Compounds can also be used for inhibiting, preventing, or treating pain associated with inflammation or with an inflammatory disease in an animal.
  • the pain to be inhibited, treated or prevented may be associated with inflammation associated with an inflammatory disease, which can arise where there is an inflammation ofthe body tissue, and which can be a local inflammatory response and/or a systemic inflammation.
  • the Piperazine Compounds can be used to inhibit, treat, or prevent pain associated with inflammatory diseases including, but not limited to: organ transplant rejection; reoxygenation injury resulting from organ transplantation (see Grupp et al, J. Mol. Cell Cardiol.
  • the Piperazine Compounds can also be used for inhibiting, treating, or preventing pain associated with inflammatory disease that can, for example, be a systemic inflammation ofthe body, exemplified by gram-positive or gram negative shock, hemorrhagic or anaphylactic shock, or shock induced by cancer chemotherapy in response to pro- inflammatory cytokines, e.g., shock associated with pro-inflammatory cytokines.
  • inflammatory disease can, for example, be a systemic inflammation ofthe body, exemplified by gram-positive or gram negative shock, hemorrhagic or anaphylactic shock, or shock induced by cancer chemotherapy in response to pro- inflammatory cytokines, e.g., shock associated with pro-inflammatory cytokines.
  • shock can be induced, e.g., by a chemotherapeutic agent that is adminstered as a treatment for cancer.
  • the Piperazine Compounds can be used to treat or prevent UI.
  • UI treatable or preventable using the Piperazine Compounds include, but are not limited to, urge incontinence, stress incontinence, overflow incontinence, neurogenic incontinence, and total incontinence.
  • the Piperazine Compounds can be used to treat or prevent an addictive disorder, including but not limited to, an eating disorder, an impulse-control disorder, an alcohol-related disorder, a nicotine-related disorder, an amphetamine-related disorder, a cannabis-related disorder, a cocaine-related disorder, an hallucinogen-related disorder, an inhalant-related disorders, and an opioid-related disorder, all of which are further sub- classified as listed below.
  • an addictive disorder including but not limited to, an eating disorder, an impulse-control disorder, an alcohol-related disorder, a nicotine-related disorder, an amphetamine-related disorder, a cannabis-related disorder, a cocaine-related disorder, an hallucinogen-related disorder, an inhalant-related disorders, and an opioid-related disorder, all of which are further sub- classified as listed below.
  • Eating disorders include, but are not limited to, Bulimia Nervosa, Nonpurging Type; Bulimia Nervosa, Purging Type; Anorexia; and Eating Disorder not otherwise specified (NOS).
  • Impulse control disorders include, but are not limited to, Intermittent
  • Alcohol-related disorders include, but are not limited to, Alcohol-Induced Psychotic Disorder with delusions, Alcohol Abuse, Alcohol Intoxication, Alcohol Withdrawal, Alcohol Intoxication Delirium, Alcohol Withdrawal Delirium,
  • Alcohol-Induced Persisting Dementia Alcohol-Induced Persisting Amnestic Disorder, Alcohol Dependence, Alcohol-Induced Psychotic Disorder with hallucinations, Alcohol-Induced Mood Disorder, Alcohol-Induced Anxiety Disorder, Alcohol-Induced Sexual Dysfunction, Alcohol-Induced Sleep Disorder, Alcohol-Related Disorder not otherwise specified (NOS), Alcohol Intoxication, and Alcohol Withdrawal.
  • Nicotine-related disorders include, but are not limited to, Nicotine Dependence, Nicotine Withdrawal, and Nicotine-Related Disorder not otherwise specified (NOS).
  • Amphetamine-related disorders include, but are not limited to, Amphetamine Dependence, Amphetamine Abuse, Amphetamine Intoxication, Amphetamine Withdrawal, Amphetamine intoxication Delirium, Amphetamine-Induced Psychotic Disorder with delusions, Amphetamine-Induced Psychotic Disorders with hallucinations, Amphetamine-Induced Mood Disorder, Amphetamine-Induced Anxiety Disorder, Amphetamine-Induced sexual Dysfunction, Amphetamine-Induced Sleep Disorder, and Amphetamine Related Disorder not otherwise specified (NOS).
  • Cannabis-related disorders include, but are not limited to, Cannabis Dependence, Cannabis Abuse, Cannabis Intoxication, Cannabis Intoxication Delirium, Cannabis-Induced Psychotic Disorder with delusions, Cannabis-Induced Psychotic
  • Cannabis-Induced Anxiety Disorder a Disorder with hallucinations, Cannabis-Induced Anxiety Disorder, and Cannabis Related Disorder not otherwise specified (NOS).
  • Cocaine-related disorders include, but are not limited to, Cocaine Dependence, Cocaine Abuse, Cocaine Intoxication, Cocaine Withdrawal, Cocaine Intoxication Delirium, Cocaine-Induced Psychotic Disorder with delusions,
  • Hallucinogen-related disorders include, but are not limited to,
  • Hallucinogen-Induced Sleep Hallucinogen Dependence, Hallucinogen Abuse, Hallucinogen Intoxication, Hallucinogen Withdrawal, Hallucinogen Intoxication Delirium, Haljucinogen-Induced Psychotic Disorder with delusions, Hallucinogen-Induced Psychotic Disorders with hallucinations, Hallucinogen-Induced Mood Disorder, Hallucinogen-Induced Anxiety Disorder, Hallucinogen-Induced Sexual Dysfunction, Hallucinogen-Induced Sleep
  • Inhalant-related disorders include, but are not limited to, Inhalant Dependence, Inhalant Abuse, Inhalant intoxication, Inhalant Intoxication Delirium, Inhalant-Induced Psychotic Disorder with delusions, Inhalant-Induced Psychotic
  • Opioid-related disorders include, but are not limited to, Opioid Dependence, Opioid Abuse, Opioid Intoxication, Opioid Intoxication Delirium, Opioid-Induced Psychotic Disorder with delusions, Opioid-Induced Psychotic Disorder with hallucinations, Opioid-Induced Anxiety Disorder, Opioid Withdrawal, and Opioid Related Disorder not otherwise specified (NOS).
  • the Piperazine Compounds can be used to treat or prevent Parkinson's disease and parkinsonism and the symptoms associated with Parkinson's disease and parkinsonism, including but not limited to, bradykinesia, muscular rigidity, resting tremor, and impairment of postural balance.
  • the Piperazine Compounds can be used to treat or prevent generalized anxiety or severe anxiety and the symptoms associated with anxiety, including but not limited to, restlessness, tension, tachycardia, dyspnea, depression including chronic "neurotic" depression, panic disorder, agoraphobia and other specific phobias, eating disorders, and personality disorders.
  • the Piperazine Compounds can be used to treat or prevent epilepsy, including but not limited to, partial epilepsy, generalized epilepsy, and the symptoms associated with epilepsy, including but not limited to, simple partial seizures, jacksonian seizures, complex partial (psychomotor) seizures, convulsive seizures (grand mal or tonic-clonic seizures), petit mal (absence) seizures, and status epilepticus.
  • the Piperazine Compounds can be used to treat or prevent a seizure, including but not limited to, infantile spasms, febrile seizures, and epileptic seizures.
  • the Piperazine Compounds can be used to treat or prevent strokes, including but not limited to, ischemic strokes and hemorrhagic strokes.
  • the Piperazine Compounds can be used to treat or prevent a pruritic condition, including but not limited to, pruritus caused by dry skin, scabies, dermatitis, he ⁇ etiformis, atopic dermatitis, pruritus vulvae et ani, malaria, insect bites, pediculosis, contact dermatitis, drug reactions, urticaria, urticarial eruptions of pregnancy, psoriasis, lichen planus, lichen simplex chronicus, exfoliative dermatitis, folliculitis, bullous pemphigoid, or fiberglass dermatitis.
  • a pruritic condition including but not limited to, pruritus caused by dry skin, scabies, dermatitis, he ⁇ etiformis, atopic dermatitis, pruritus vulvae et ani, malaria, insect bites, pediculosis, contact dermatitis, drug reactions, urticaria, ur
  • the Piperazine Compounds can be used to treat or prevent psychosis, including but not limited to, schizophrenia, including paranoid schizophrenia, hebephrenic or disorganized schizophrenia, catatonic schizophrenia, undifferentiated schizophrenia, negative or deficit subtype schizophrenia, and non-deficit schizophrenia; a delusional disorder, including erotomanic subtype delusional disorder, grandiose subtype delusional disorder, ashamed subtype delusional disorder, persecutory subtype delusional disorder, and somatic subtype delusional disorder; and brief psychosis.
  • schizophrenia including paranoid schizophrenia, hebephrenic or disorganized schizophrenia, catatonic schizophrenia, undifferentiated schizophrenia, negative or deficit subtype schizophrenia, and non-deficit schizophrenia
  • a delusional disorder including erotomanic subtype delusional disorder, grandiose subtype delusional disorder, ashamed subtype delusional disorder, persecutory subtype delusional disorder, and somatic subtype delusional disorder
  • psychosis including but not
  • the Piperazine Compounds can be used to treat or prevent a cognitive disorder, including but not limited to, delirium and dementia such as multi-infarct dementia, dementia pugilistica, dementia caused by AIDS, and dementia caused by Alzheimer's disease.
  • a cognitive disorder including but not limited to, delirium and dementia such as multi-infarct dementia, dementia pugilistica, dementia caused by AIDS, and dementia caused by Alzheimer's disease.
  • the Piperazine Compounds can be used to treat or prevent a memory deficiency, including but not limited to, dissociative amnesia and dissociative fugue.
  • the Piperazine Compounds can be used to treat or prevent restricted brain function, including but not limited to, that caused by surgery or an organ transplant, restricted blood supply to the brain, a spinal cord injury, a head injury, hypoxia, cardiac arrest, or hypoglycemia.
  • the Piperazine Compounds can be used to treat or prevent Huntington's chorea.
  • the Piperazine Compounds can be used to treat or prevent ALS.
  • the Piperazine Compounds can be used to treat or prevent retinopathy, including but not limited to, arteriosclerotic retinopathy, diabetic arteriosclerotic retinopathy, hypertensive retinopathy, non-proliferative retinopathy, and proliferative retinopathy.
  • the Piperazine Compounds can be used to treat or prevent a muscle spasm.
  • the Piperazine Compounds can be used to treat or prevent a migraine.
  • the Piperazine Compounds can be used to treat or prevent vomiting, including but not limited to, nausea vomiting, dry vomiting (retching), and regurgitation.
  • the Piperazine Compounds can be used to treat or prevent dyskinesia, including but not limited to, tardive dyskinesia and biliary dyskinesia.
  • Piperazine Compounds can be used to treat or prevent depression, including but not limited to, major depression and bipolar disorder. Without wishing to be bound by theory, Applicants believe that the
  • Piperazine Compounds are antagonists for mGluR5.
  • the invention also relates to methods for inhibiting mGluR5 function in a cell comprising contacting a cell capable of expressing mGluR5 with an amount ofa Piperazine Compound effective to inhibit mGluR5 function in the cell.
  • This method can be used in vitro, for example, as an assay to select cells that express mGluR5 and, accordingly, are useful as part of an assay to select compounds useful for treating or preventing pain, an addictive disorder, Parkinson's disease, parkinsonism, anxiety, a pruritic condition or psychosis.
  • the method is also useful for inhibiting mGluR5 function in a cell in vivo, in an animal, a human in one embodiment, by contacting a cell, in an animal, with an amount of a Piperazine Compound effective to inhibit mGluR5 function in the cell.
  • the method is useful for treating or preventing pain in an animal in need thereof.
  • the method is useful for treating or preventing an addictive disorder in an animal in need thereof.
  • the method is useful for treating or preventing Parkinson's disease in an animal in need thereof.
  • the method is useful for treating or preventing parkinsonism in an animal in need thereof.
  • the method is useful for treating or preventing anxiety in an animal in need thereof.
  • the method is useful for treating or preventing a pruritic condition in an animal in need thereof.
  • the method is useful for treating or preventing psychosis in an animal in need thereof.
  • Examples of cells capable of expressing mGluR5 are neuronal and glial cells ofthe central nervous system, particularly the brain, especially in the nucleus accumbens. Methods for assaying cells that express mGluR5 are well known in the art.
  • the invention also relates to methods for inhibiting mGluRl function in a cell comprising contacting a cell capable of expressing mGluRl with an amount ofa Piperazine Compound effective to inhibit mGluRl function in the cell.
  • This method can be used in vitro, for example, as an assay to select cells that express mGluRl and, accordingly, are useful as part of an assay to select compounds useful for treating or preventing pain, UI, an addictive disorder, Parkinson's disease, parkinsonism, anxiety, epilepsy, a seizure, stroke, a pruritic condition, psychosis, a cognitive disorder, a memory deficit, restricted brain function, Huntington's chorea, ALS, dementia, retinopathy, a muscle spasm, a migraine, vomiting, dyskinesia or depression.
  • the method is also useful for inhibiting mGluRl function in a cell in vivo, in an animal, a human in one embodiment, by contacting a cell, in an animal, with an amount ofa Piperazine Compound effective to inhibit mGluRl function in the cell.
  • the method is useful for treating or preventing pain in an animal in need thereof.
  • the method is useful for treating or preventing UI in an animal in need thereof.
  • the method is useful for treating or preventing an addictive disorder in an animal in need thereof.
  • the method is useful for treating or preventing Parkinson's disease in an animal in need thereof.
  • the method is useful for treating or preventing parkinsonism in an animal in need thereof.
  • the method is useful for treating or preventing anxiety in an animal in need thereof. In another embodiment, the method is useful for treating or preventing epilepsy in an animal in need thereof. In another embodiment, the method is useful for treating or preventing a seizure in an animal in need thereof. In another embodiment, the method is useful for treating or preventing stroke in an animal in need thereof. In another embodiment, the method is useful for treating or preventing a pruritic condition in an animal in need thereof. In another embodiment, the method is useful for treating or preventing psychosis in an animal in need thereof. In another embodiment, the method is useful for treating or preventing a cognitive disorder in an animal in need thereof. In another embodiment, the method is useful for treating or preventing a memory deficit in an animal in need thereof.
  • the method is useful for treating or preventing restricted brain function in an animal in need thereof.
  • the method is useful for treating or preventing Huntington's chorea in an animal in need thereof.
  • the method is useful for treating or preventing ALS -nan animal in need thereof.
  • the method is useful for treating or preventing dementia in an animal in need thereof, ha another embodiment, the method is useful for treating or preventing retinopathy in an animal in need thereof.
  • the method is useful for treating or preventing a muscle spasm in an animal in need thereof.
  • the method is useful for treating or preventing a migraine in an animal in need thereof.
  • the method is useful for treating or preventing vomiting in an animal in need thereof.
  • the method is useful for treating or preventing dyskinesia in an animal in need thereof.
  • the method is useful for treating or preventing depression in an animal in need thereof.
  • Examples of cells capable of expressing mGluRl include, but are not limited to, cerebellar Purkinje neuron cells, Purkinje cell bodies (punctate), cells of spine(s) ofthe cerebellum; neurons and neurophil cells of olfactory-bulb glomeruli; cells ofthe superficial layer ofthe cerebral cortex; hippocampus cells; thalamus cells; superior colliculus cells; and spinal trigeminal nucleus cells. Methods for assaying cells that express mGluRl are well known in the art.
  • Compounds are useful for treating or preventing a Condition in an animal in need thereof.
  • the Piperazine Compounds When administered to an animal, the Piperazine Compounds are administered as a component of a composition that comprises a pharmaceutically acceptable carrier or excipient.
  • the present compositions, which comprise a Piperazine Compound can be administered orally.
  • the Piperazine Compounds ofthe invention can also be administered by any other convenient route, for example, by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral, rectal, and intestinal mucosa, etc.) and can be administered together with another biologically active agent. Administration can be systemic or local. Narious delivery systems are known, e.g., encapsulation in liposomes, microparticles, microcapsules, capsules, etc., and can be used to administer the Piperazine Compound.
  • Methods of administration include, but are not limited to, intraderrnal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, oral, sublingual, intracerebral, intravaginal, transdermal, rectal, by inhalation, or topical, particularly to the ears, nose, eyes, or skin.
  • the mode of administration is left to the discretion ofthe practitioner. In most instances, administration will result in the release ofthe Piperazine Compounds into the bloodstream.
  • This can be achieved, for example, and not by way of limitation, by local infusion during surgery, topical application, e.g., in conjunction with a wound dressing after surgery, by injection, by means of a catheter, by means of a suppository or enema, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers.
  • Compounds into the central nervous system or gastrointestinal tract by any suitable route including intraventricular, intrathecal, and epidural injection, and enema.
  • Intraventricular injection can be facilitated by an intraventricular catheter, for example, attached to a reservoir, such as an Ommaya reservoir.
  • Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent, or via perfusion in a fluorocarbon or synthetic pulmonary surfactant.
  • Compounds can be formulated as a suppository, with traditional binders and excipients such as triglycerides.
  • the Piperazine Compounds can be delivered in a vesicle, in particular a liposome (see Langer, Science 249:1527-1533 (1990) and Treat et al, Liposomes in the Therapy of Infectious Disease and Cancer 317-327 and 353-365
  • Piperazine Compounds can be delivered in a controlled-release system or sustained-release system (see, e.g., Goodson, in
  • a pump can be used (Langer,
  • polymeric materials can be used (see Medical Applications of Controlled Release (Langer and Wise eds., 1974); Controlled Drug
  • a controlled- or sustained- release system can be placed in proximity of a target ofthe Piperazine Compounds, e.g., the spinal column, brain, or gastrointestinal tract, thus requiring only a fraction ofthe systemic dose.
  • the present compositions can optionally comprise a suitable amount of a pharmaceutically acceptable excipient so as to provide the form for proper administration to the animal.
  • Such pharmaceutical excipients can be liquids, such as water and oils, including those of petroleum, animal, vegetable, or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like.
  • the pharmaceutical excipients can be saline, gum acacia, gelatin, starch paste, talc, keratin, colloidal silica, urea and the like.
  • auxiliary, stabilizing, thickening, lubricating, and coloring agents can be used.
  • the pharmaceutically acceptable excipients are sterile when administered to an animal. Water, and in one embodiment physiological saline, is a particularly useful excipient when the Piperazine Compound is administered intravenously.
  • Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid excipients, particularly for injectable solutions.
  • suitable pharmaceutical excipients also include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like.
  • the present compositions if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • compositions can take the form of solutions, suspensions, emulsion, tablets, pills, pellets, capsules, capsules containing liquids, powders, sustained-release formulations, suppositories, emulsions, aerosols, sprays, suspensions, or any other form suitable for use.
  • the composition is in the form of a capsule (see e.g., U.S. Patent No. 5,698,155).
  • suitable pharmaceutical excipients are described in Remington 's Pharmaceutical Sciences 1447- 1676 (Alfonso R. Gennaro ed., 19th ed. 1995), inco ⁇ orated herein by reference.
  • the Piperazine Compounds are formulated in accordance with routine procedures as a composition adapted for oral administration to human beings.
  • Compositions for oral delivery can be in the form of tablets, lozenges, aqueous or oily suspensions, granules, powders, emulsions, capsules, syrups, or elixirs, for example.
  • Orally administered compositions can contain one or more agents, for example, sweetening agents such as fructose, aspartame or saccharin; flavoring agents such as peppermint, oil of wintergreen, or cherry; coloring agents; and preserving agents, to provide a pharmaceutically palatable preparation.
  • compositions can be coated to delay disintegration and abso ⁇ tion in the gastrointestinal tract thereby providing a sustained action over an extended period of time.
  • Selectively permeable membranes surrounding an osmotically active driving compound are also suitable for orally administered compositions. In these latter platforms, fluid from the environment surrounding the capsule is imbibed by the driving compound, which swells to displace the agent or agent composition through an aperture.
  • These delivery platforms can provide an essentially zero order delivery profile as opposed to the spiked profiles of immediate release formulations.
  • a time-delay material such as glycerol monostearate or glycerol stearate can also be used.
  • Oral compositions can include standard excipients such as mannitol, lactose, starch, magnesium stearate, sodium saccharin, cellulose, and magnesium carbonate. In one embodiment, the excipients are of pharmaceutical grade.
  • the Piperazine Compounds can be formulated for intravenous administration.
  • compositions for intravenous administration comprise sterile isotonic aqueous buffer.
  • the compositions can also include a solubilizing agent.
  • Compositions for intravenous administration can optionally include a local anesthetic such as lidocaine to lessen pain at the site ofthe injection.
  • the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampule or sachette indicating the quantity of active agent.
  • Piperazine Compounds are to be administered by infusion, they can be dispensed, for example, with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the Piperazine Compounds are admimstered by injection, an ampule of sterile water for injection or saline can be provided so that the ingredients can be mixed prior to administration.
  • the Piperazine Compounds can be administered by controlled-release or sustained-release means or by delivery devices that are well known to those of ordinary skill in the art. Examples include, but are not limited to, those described in U.S. Patent Nos.: 3,845,770; 3,916,899; 3,536,809; 3,598,123; 4,008,719; 5,674,533; 5,059,595; 5,591,767; 5,120,548; 5,073,543; 5,639,476; 5,354,556; and 5,733,566, each of which is inco ⁇ orated herein by reference.
  • Such dosage forms can be used to provide controlled- or sustained-release of one or more active ingredients using, for example, hydropropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or a combination thereof to provide the desired release profile in varying proportions.
  • Suitable controlled- or sustained-release formulations known to those of ordinary skill in the art, including those described herein, can be readily selected for use with the active ingredients ofthe invention.
  • the invention thus encompasses single unit dosage forms suitable for oral administration such as, but not limited to, tablets, capsules, gelcaps, and caplets that are adapted for controlled- or sustained-release.
  • Controlled- or sustained-release pharmaceutical compositions can have a common goal of improving drug therapy over that achieved by their non-controlled or non-sustained counte ⁇ arts.
  • a controlled- or sustained-release composition comprises a minimal amount of a Piperazine Compound to cure or control the condition in a minimum amount of time.
  • Advantages of controlled- or sustained- release compositions include extended activity ofthe drug, reduced dosage frequency, and increased patient compliance.
  • controlled- or sustained-release compositions can favorably affect the time of onset of action or other characteristics, such as blood levels ofthe Piperazine Compound, and can thus reduce the occurrence of adverse side effects.
  • Controlled- or sustained-release compositions can initially release an amount of a Piperazine Compound that promptly produces the desired therapeutic or prophylactic effect, and gradually and continually release other amounts ofthe
  • Piperazine Compound to maintain this level of therapeutic or prophylactic effect over an extended period of time.
  • the Piperazine Compound can be released from the dosage form at a rate that will replace the amount of Piperazine Compound being metabolized and excreted from the body.
  • Controlled- or sustained-release of an active ingredient can be stimulated by various conditions, including but not limited to, changes in pH, changes in temperature, concentration or availability of enzymes, concentration or availability of water, or other physiological conditions or compounds.
  • the amount ofthe Piperazine Compound that is effective in the treatment or prevention of a Condition can be determined by standard clinical techniques.
  • in vitro or in vivo assays can optionally be employed to help identify optimal dosage ranges.
  • the precise dose to be employed will also depend on the route of administration, and the seriousness ofthe Condition and should be decided according to the judgment ofthe practitioner and each patient's circumstances in view of published clinical studies. Suitable effective dosage amounts, however, range from about 10 micrograms to about 2500 milligrams about every 4 h, although they are typically about 100 mg or less.
  • the effective dosage amount ranges from about 0.01 milligrams to about 100 milligrams of a Piperazine Compound about every 4 h, in another embodiment, about 0.020 milligrams to about 50 milligrams about every 4 h, and in another embodiment, about 0.025 milligrams to about 20 milligrams about every 4 h.
  • the effective dosage amounts described herein refer to total amounts administered; that is, if more than one Piperazine Compound is administered, the effective dosage amounts correspond to the total amount administered.
  • the amount effective for inhibiting the receptor function in a cell will typically range from about 0.01 ⁇ g/L to about 5 mg/L, in one embodiment, from about 0.01 ⁇ g/L to about 2.5 mg/L, in another embodiment, from about 0.01 ⁇ g/L to about 0.5 mg/L, and in another embodiment, from about 0.01 jug/L to about 0.25 mg/L of a solution or suspension of a pharmaceutically acceptable carrier or excipient.
  • the volume of solution or suspension is from about 1 ⁇ L to about 1 mL. In another embodiment, the volume of solution or suspension is about 200 ⁇ L.
  • the amount effective for inhibiting the receptor function in a cell will typically range from about 0.01 mg to about 100 mg/kg of body weight per day, in one embodiment, from about 0.1 mg to about 50 mg/kg body weight per day, and in another embodiment, from about 1 mg to about 20 mg/kg of body weight per day.
  • the Piperazine Compounds can be assayed in vitro or in vivo for the desired therapeutic or prophylactic activity prior to use in humans. Animal model systems can be used to demonstrate safety and efficacy.
  • the present methods for treating or preventing a Condition in an animal in need thereof can further comprise administering to the animal being administered a Piperazine Compound another therapeutic agent.
  • the other therapeutic agent is administered in an effective amount.
  • the present methods for inhibiting mGluR5 function in a cell capable of expressing mGluR5 can further comprise contacting the cell with an effective amount of another therapeutic agent.
  • the present methods for inhibiting mGluRl function in a cell capable of expressing mGluRl can further comprise contacting the cell with an effective amount of another therapeutic agent.
  • the other therapeutic agent includes, but is not limited to, an opioid agonist, a non-opioid analgesic, a non-steroidal anti-inflammatory agent, an antimigraine agent, a Cox-II inhibitor, an antiemetic, a -adrenergic blocker, an anticonvulsant, an antidepressant, a Ca2+-channel blocker, an anticancer agent, an agent for treating or preventing one or more Conditions, and mixtures thereof.
  • Effective amounts ofthe other therapeutic agents are well known to those skilled in the art. However, it is well within the skilled artisan's purview to determine the other therapeutic agent's optimal effective-amount range. In one embodiment ofthe invention, where another therapeutic agent is administered to an animal, the effective amount ofthe Piperazine Compound is less than its effective amount would be where the other therapeutic agent is not administered. In this case, without being bound by theory, it is believed that the Piperazine Compounds and the other therapeutic agent act synergistically to treat or prevent a Condition.
  • Examples of useful opioid agonists include, but are not limited to, alfentanil, allylprodine, alphaprodine, anileridine, benzylmo ⁇ hine, bezitramide, bupreno ⁇ hine, buto ⁇ hanol, clonitazene, codeine, desomo ⁇ hine, dextromoramide, dezocine, diampromide, diamo ⁇ hone, dihydrocodeine, dihydromo ⁇ hine, dimenoxadol, dimepheptanol, dimethylthiambutene, dioxaphetyl butyrate, dipipanone, eptazocine, ethoheptazine, ethylmethylthiambutene, ethylmo ⁇ hine, etonitazene fentanyl, heroin, hydrocodone, hydromo ⁇ hone, hydroxypethidine, isomethadone, ketobemidone, levo
  • the opioid agonist is selected from codeine, hydromo ⁇ hone, hydrocodone, oxycodone, dihydrocodeine, dihydromo ⁇ hine, mo ⁇ hine, tramadol, oxymo ⁇ hone, pharmaceutically acceptable salts thereof, and mixtures thereof.
  • non-steroidal anti-inflammatory agents such as aspirin, ibuprofen, diclofenac, naproxen, benoxaprofen, flurbiprofen, fenoprofen, flubufen, ketoprofen, indoprofen, piroprofen, ca ⁇ rofen, oxaprozin, pramoprofen, muroprofen, trioxaprofen, suprofen, aminoprofen, tiaprofenic acid, fluprofen, bucloxic acid, indomethacin, sulindac, tolmetin, zomepirac, tiopinac, zidometacin, acemetacin, fentiazac, clidanac, oxpinac, mefenamic acid, meclofenamic acid, flufenamic acid, niflumic acid, tolfenamic
  • non-steroidal anti-inflammatory agents such aspirin,
  • non-opioid analgesics include the following, non-limiting, chemical classes of analgesic, antipyretic, non-steroidal anti-inflammatory drugs: salicylic acid derivatives, including aspirin, sodium salicylate, choline magnesium trisalicylate, salsalate, diflunisal, salicylsalicylic acid, sulfasalazine, and olsalazin; para-aminophennol derivatives including acetaminophen and phenacetin; indole and indene acetic acids, including indomethacin, sulindac, and etodolac; heteroaryl acetic acids, including tolmetin, diclofenac, and ketorolac; anthranilic acids (fenamates), including mefenamic acid and meclofenamic acid; enolic acids, including oxicams (piroxicam, tenoxicam), and pyrazolidinediones (phenyl
  • useful antimigraine agents include, but are not limited to, alpiropride, dihydroergotamine, dolasetron, ergocornine, ergocorninine, ergocryptine, ergot, ergotamine, flumedroxone acetate, fonazine, lisuride, lomerizine, mefhysergide oxetorone, pizotyline, and mixtures thereof.
  • the other therapeutic agent can also be an agent useful for reducing any potential side effects of a Piperazine Compounds.
  • the other therapeutic agent can be an antiemetic agent.
  • useful antiemetic agents include, but are not limited to, metoclopromide, domperidone, prochlo ⁇ erazine, promethazine, chlo ⁇ romazine, trimethobenzamide, odansetron, granisetron, hydroxyzine, acetylleucine monoethanolamine, alizapride, azasetron, benzquinamide, bietanautine, bromopride, buclizine, clebopride, cyclizine, dimenhydrinate, diphenidol, dolasetron, meclizine, methallatal, metopimazine, nabilone, oxyperndyl, pipamazine, scopolamine, sulpiride, tetrahydrocannabinol, thiethylperazine, thioproperazine, tropisetron, and mixtures thereof.
  • ⁇ -adrenergic blockers include, but are not limited to, acebutolol, alprenolol, amosulabol, arotinolol, atenolol, befunolol, betaxolol, bevantolol, bisoprolol, bopindolol, bucumolol, bufetolol, bufuralol, bunitrolol, bupranolol, butidrine hydrochloride, butofilolol, carazolol, carteolol, carvedilol, celiprolol, cetamolol, cloranolol, dilevalol, epanolol, esmolol, indenolol, labetalol, levobunolol, mepindolol, metipranolol, metoprolol, moprol
  • useful anticonvulsants include, but are not limited to, acetylpheneturide, albutoin, aloxidone, aminoglutethimide, 4-amino-3-hydroxybutyric acid, atrolactamide, beclamide, buramate, calcium bromide, carbamazepine, cinromide, clomethiazole, clonazepam, decimemide, diethadione, dimethadione, doxenitroin, eterobarb, ethadione, ethosuximide, ethotoin, felbamate, fluoresone, gabapentin, 5-hydroxytryptophan, lamotrigine, magnesium bromide, magnesium sulfate, mephenytoin, mephobarbital, metharbital, methetoin, methsuximide,
  • Examples of useful antidepressants include, but are not limited to, binedaline, caroxazone, citalopram, dimethazan, fencamine, indalpine, indeloxazine hydrocholoride, nefopam, nomifensine, oxitriptan, oxypertine, paroxetine, sertraline, thiazesim, trazodone, benmoxine, iproclozide, iproniazid, isocarboxazid, nialamide, octamoxin, phenelzine, cotinine, rolicyprine, rolipram, maprotiline, metralindole, mianserin, mirtazepine, adinazolam, amitriptyline, amitriptylinoxide, amoxapine, butriptyline, clomipramine, demexiptiline, desipramine, dibenz
  • Ca2+-channel blockers examples include, but are not limited to, bepridil, clentiazem, diltiazem, fendiline, gallopamil, mibefradil, prenylamine, semotiadil, terodiline, verapamil, amlodipine, aranidipine, barnidipine, benidipine, cilnidipine, efonidipine, elgodipine, felodipine, isradipine, lacidipine, lercanidipine, manidipine, nicardipine, nifedipine, nilvadipine, nimodipine, nisoldipine, nitrendipine, cinnarizine, flunarizine, lidoflazine, lomerizine, bencyclane, etafenone, fantofarone, and perhexiline.
  • useful anticancer agents include, but are not limited to, acivicin, aclarubicin, acodazole hydrochloride, acronine, adozelesin, aldesleukin, altretamine, ambomycin, ametantrone acetate, aminoglutethimide, amsacrine, anastrozole, anthramycin, asparaginase, asperlin, azacitidine, azetepa, azotomycin, batimastat, benzodepa, bicalutamide, bisantrene hydrochloride, bisnafide dimesylate, bizelesin, bleomycin sulfate, brequinar sodium, bropirimine, busulfan, cactinomycin, calusterone, caracemide, carbetimer, carboplatin, carmustine, carubicin hydrochloride, carzelesin, cedefingol, chlorambucii,
  • anti-cancer drugs include, but are not limited to, 20-epi-l,25 dihydroxyvitamin D3; 5-ethynyluracil; abiraterone; aclarubicin; acylfulvene; adecypenol; adozelesin; aldesleukin; ALL-TK antagonists; altretamine; ambamustine; amidox; amifostine; aminolevulinic acid; amrubicin; amsacrine; anagrelide; anastrozole; andrographolide; angiogenesis inhibitors; antagonist D; antagonist G; antarelix; anti-dorsalizing mo ⁇ hogenetic protein- 1; antiandrogen, prostatic carcinoma; antiestrogen; antineoplaston; antisense oligonucleotides; aphidicolin glycinate; apoptosis gene modulators; apoptosis regulators; apurinic acid; ara-CDP-
  • BCR/ABL antagonists benzochlorins; benzoylstaurosporine; beta lactam derivatives; beta-alethine; betaclamycin B; betulinic acid; bFGF inhibitor; bicalutamide; bisantrene; bisaziridinylspermine; bisnafide; bistratene A; bizelesin; breflate; bropirimine; budotitane; buthionine sulfoximine; calcipotriol; calphostin C; camptothecin derivatives; canarypox IL-2; capecitabine; carboxamide-amino-triazole; carboxyamidotriazole;
  • useful therapeutic agents for treating or preventing an addictive disorder include, but are not limited to, methadone, desipramine, amantadine, fluoxetine, bupreno ⁇ hine, an opiate agonist, 3-phenoxypyridine, levomethadyl acetate hydrochloride, and serotonin antagonists.
  • Parkinson's disease and parkinsonism include, but are not limited to, carbidopa/levodopa, pergolide, bromocriptine, ropinirole, pramipexole, entacapone, tolcapone, selegiline, amantadine, and trihexyphenidyl hydrochloride.
  • useful therapeutic agents for treating or preventing anxiety include, but are not limited to, benzodiazepines, such as alprazolam, brotizolam, chlordiazepoxide, clobazam, clonazepam, clorazepate, demoxepam, diazepam, estazolam, flumazenil, flurazepam, halazepam, lorazepam, midazolam, nitrazepam, nordazepam, oxazepam, prazepam, quazepam, temazepam, and triazolam; non- benzodiazepine agents, such as buspirone, gepirone, ipsapirone, tiospirone, zolpicone, zolpidem, and zaleplon; tranquilizers, such as barbituates, e.g., amobarbital, aprobarbital, butabarbital, butalbital, me
  • useful therapeutic agents for treating or preventing epilepsy include, but are not limited to, carbamazepine, ethosuximide, gabapentin, lamotrigine, phenobarbital, phenytoin, primidone, valproic acid, trimethadione, benzodiazepines, gabapentin, lamotrigine, ⁇ -vinyl GABA, acetazolamide, and felbamate.
  • useful therapeutic agents for treating or preventing a seizure include, but are not limited to, carbamazepine, ethosuximide, gabapentin, lamotrigine, phenobarbital, phenytoin, primidone, valproic acid, trimethadione, benzodiazepines, gabapentin, lamotrigine, ⁇ -vinyl GABA, acetazolamide, and felbamate.
  • useful therapeutic agents for treating or preventing stroke include, but are not limited to, anticoagulants such as heparin, agents that break up clots such as streptokinase or tissue plasminogen activator, agents that reduce swelling such as mannitol or corticosteroids, and acetylsalicylic acid.
  • anticoagulants such as heparin
  • agents that break up clots such as streptokinase or tissue plasminogen activator
  • agents that reduce swelling such as mannitol or corticosteroids
  • acetylsalicylic acid acetylsalicylic acid
  • useful therapeutic agents for treating or preventing a pruritic condition include, but are not limited to, naltrexone; nalmefene; danazol; tricyclics such as amitriptyline, imipramine, and doxepin; antidepressants such as those given below; menthol; camphor; phenol; pramoxine; capsaicin; tar; steroids; and antihistamines.
  • useful therapeutic agents for treating or preventing psychosis include, but are not limited to, phenothiazines such as chlo ⁇ romazine hydrochloride, mesoridazine besylate, and thoridazine hydrochloride; thioxanthenes such as chloroprothixene and thiothixene hydrochloride; clozapine; risperidone; olanzapine; quetiapine; quetiapine fumarate; haloperidol; haloperidol decanoate; loxapine succinate; molindone hydrochloride; pimozide; and ziprasidone.
  • phenothiazines such as chlo ⁇ romazine hydrochloride, mesoridazine besylate, and thoridazine hydrochloride
  • thioxanthenes such as chloroprothixene and thiothixene hydroch
  • Huntington's chorea include, but are not limited to, haloperidol and pimozide.
  • useful therapeutic agents for treating or preventing ALS include, but are not limited to, baclofen, neurotrophic factors, riluzole, tizanidine, benzodiazepines such as clonazepan and dantrolene.
  • useful therapeutic agents for treating or preventing cognitive disorders include, but are not limited to, agents for treating or preventing dementia such as tacrine; donepezil; ibuprofen; antipsychotic drugs such as thioridazine and haloperidol; and antidepressant drugs such as those given below.
  • useful therapeutic agents for treating or preventing a migraine include, but are not limited to, sumatriptan; methysergide; ergotamine; caffeine; and beta-blockers such as propranolol, verapamil, and divalproex.
  • Examples of useful therapeutic agents for treating or preventing vomiting include, but are not limited to, 5-HT 3 receptor antagonists such as odansetron, dolasetron, granisetron, and tropisetron; dopamine receptor antagonists such as prochlo ⁇ erazine, thiethylperazine, chlo ⁇ romazine, metoclopramide, and domperidone; glucocorticoids such as dexamethasone; and benzodiazepines such as lorazepam and alprazolam.
  • 5-HT 3 receptor antagonists such as odansetron, dolasetron, granisetron, and tropisetron
  • dopamine receptor antagonists such as prochlo ⁇ erazine, thiethylperazine, chlo ⁇ romazine, metoclopramide, and domperidone
  • glucocorticoids such as dexamethasone
  • benzodiazepines such as lorazep
  • Examples of useful therapeutic agents for treating or preventing dyskinesia include, but are not limited to, rese ⁇ ine and tetrabenazine.
  • useful therapeutic agents for treating or preventing depression include, but are not limited to, tricyclic antidepressants such as amitryptyline, amoxapine, bupropion, clomipramine, desipramine, doxepin, imipramine, maprotiline, nefazadone, nortriptyline, protriptyline, trazodone, trimipramine, a ⁇ d venlafaxine; selective serotonin reuptake inhibitors such as fluoxetine, fluvoxamine, paroxetine, and setraline; monoamine oxidase inhibitors such as isocarboxazid, pargyline, phenelzine, and tranylcypromine; and psychostimulants such as dextroamphetamine and methylphenidate.
  • tricyclic antidepressants such as amitryptyline, amoxapine, bupropion, clomipramine, desipramine, doxepin,
  • a Piperazine Compound and the other therapeutic agent can act additively or, in one embodiment, synergistically.
  • a Piperazine Compound is administered concurrently with another therapeutic agent.
  • a composition comprising an effective amount of a Piperazine Compound and an effective amount of another therapeutic agent can be administered.
  • a composition comprising an effective amount of a Piperazine Compound and a different composition comprising an effective amount of another therapeutic agent can be concurrently administered.
  • an effective amount of a Piperazine Compound is administered prior or subsequent to administration of an effective amount of another therapeutic agent.
  • the Piperazine Compound is administered while the other therapeutic agent exerts its therapeutic effect, or the other therapeutic agent is administered while the Piperazine Compound exerts its preventative or therapeutic effect for treating or preventing a Condition.
  • a composition ofthe invention is prepared by a method comprising admixing a Piperazine Compound and pharmaceutically acceptable salt and a pharmaceutically acceptable carrier or excipient. Admixing can be accomplished using methods well known for admixing a compound (or salt) and a pharmaceutically acceptable carrier or excipient. In one embodiment the Piperazine Compound or the pharmaceutically acceptable salt ofthe Compound is present in the composition in an effective amount.
  • kits that can simplify the administration of a
  • a typical kit ofthe invention comprises a unit dosage form of a Piperazine Compound.
  • the unit dosage form is a container, which can be sterile, containing an effective amount of a Piperazine Compound and a pharmaceutically acceptable carrier or excipient.
  • the kit can further comprise a label or printed instructions instructing the use ofthe Piperazine Compound to treat a Condition.
  • the kit can also further comprise a unit dosage form of another therapeutic agent, for example, a container containing an effective amount ofthe other therapeutic agent.
  • the kit comprises a container containing an effective amount ofa Piperazine Compound and an effective amount of another therapeutic agent. Examples of other therapeutic agents include, but are not limited to, those listed above.
  • Kits ofthe invention can further comprise a device that is useful for administering the unit dosage forms.
  • a device includes, but are not limited to, a syringe, a drip bag, a patch, an inhaler, and an enema bag.
  • Examples 1-68 relate to the synthesis of illustrative Piperazine
  • Phenylpropynoic acid I (1.9 g, 13 mmol) was dissolved in 75 mL anhydrous CH 2 C1 2 and oxalyl chloride (3.8 mL, 43 mmol) was added followed by 2 drops of dimethylformamide. The resulting mixture was protected from exposure to moisture with a drying tube and stirred at room temperature for 2 hours. The solution was then evaporated to dryness to afford compound J. Compound J was used in final step C without further purification.
  • Compound AX was prepared by reacting l-(6-methyl-3-nitro-pyridin-2- yl)-piperazine with compound I using a procedure analogous to that used to make Compound AB (Example 6.2). HPLC analysis showed that the purity of Compound AX was greater than 97%. The structure of Compound AX, was confirmed by 1H NMR and MS analysis.
  • Compounds AE and AY can be prepared by reacting l-(3-chloro-pyridin- 2-yl)-piperazine (for Compound AE) or l-(4-methyl-3-nitro-pyridin-2-yl)-piperazine (for Compound AY) with compound I using a procedure analogous to that used to make Compound AB (Example 6.2).
  • Compounds AQ, AW and BB were prepared by reacting compound M with l-(3-chloro-pyridin-2-yl)-piperazine (for Compound AQ), l-(4-methyl-3-nitro- pyridin-2-yl)-piperazine (for Compound AW), or l-(6-methyl-3-cyano-pyridin-2-yl)- piperazine (for Compound BB) using a procedure analogous to that used to make Compound AO (Example 4).
  • the structure of Compounds AQ, AW and BB were confirmed by 1H NMR and MS analysis.
  • Compounds AZ and BA can be prepared by reacting 4-phenyl-3-butyn-2- one and l-(3-nitro-pyridin-2-yl)-piperazine (for Compound AZ) or l-(3-cyano-pyridin-2- yl)-piperazine (for Compound BA) using a procedure analogous to that used to make Compound AO (Example 4) as depicted below.
  • Compounds BC and BD were prepared by reacting compound H (prepared according to the method described in Step A of Example 1) with the appropriately substituted phenylacetylene and paraformaldehyde in dioxane at 70° C in the presence of CuCl.
  • the structure of Compounds BC and BD was confirmed by 1H NMR.
  • Compound BE was prepared by reacting compound N (prepared according to the method described in Step A of Example 1) with compound I in the presence of HOBT and DIC in methylene chloride at 70° C. The structure of Compound BE was confirmed by 1H NMR.
  • Compound BF was prepared by reacting compound O (prepared according to the method described in Step A of Example 1) with compound I in the presence of HOBT and DIC in methylene chloride at 70°C. The structure of Compound BF was confirmed by 1H NMR.
  • Example 11 Synthesis of Compound BG Compound BG was prepared according to Example 10, except that 3- iodopyridine was used in place of compound Q.
  • Compound BG was prepared from compound P according to Example 10, except that 3-bromopyridine was used in place of compound Q.
  • Compound BG: 1H NMR (CDCI 3 ) 8.40 (dd, IH, J 1.7 and 4.6 Hz), 8.22
  • Compound BH was prepared according to Example 10, except that 2- iodopyrazine was used in place of compound Q.
  • Compound BJ was prepared according to Example 10, except that 2- bromo-3-methylpyridine was used in place of compound Q.
  • Example 16 Synthesis of Compound BK Compound BK was prepared according to Example 10, except that 2- iodo-6-methylpyridine was used in place of compound Q.
  • Compound BN was prepared according to Example 10, except that 4- iodopyridine was used in place of compound Q.
  • Compound BO was prepared according to Example 10, except that 5- iodo-2-methoxypyridine was used in place of compound Q.
  • Example 21 Synthesis of Compound BP
  • Compound BP was prepared according to Example 10, except that 2- fluoro-5 -iodopyridine was used in place of compound Q.
  • Compound CK was prepared from compound P according to Example 10, except that 4-bromoanisole was used in place of compound Q.
  • Compound 100 was prepared according to Example 1, except that 2- chloropyridine was used in place of compound E.
  • Compound 100 1H NMR (CDC1 3 ) 8.25 (d, IH), 7.55-7.65 (m, 3H), 7.40-
  • Compound BW was prepared from compound P according to Example 10, except that 3-bromo-4-fluorotoluene was used in place of compound Q.
  • Compound BQ was prepared from compound P according to Example 10, except that 4-bromo-l,2-(methylenedioxy)benzene was used in place of compound Q.
  • Compound BQ 1H NMR (CDC1 3 ) 8.40 (d, IH), 8.23 (d, IH), 7.20 (d, IH), 7.00 (s, IH), 6.80-6.90 (m, IH), 6.75 (d, IH), 6.05 (s, 2H), 4.05 (dd, 2H), 3.80 (dd, 2H), 3.70-3.80 (m, 4H).
  • Compound BV was prepared according to Example 7, except that compound H was used in place of compound N and 2-hexynoic acid was used in place of compound I.
  • Compound CH was prepared according to Example 1, except that 2- chloro-3-trifluoromethylpyridine was used in place of compound E.
  • Compound 160 was prepared according to Example 1, except that 2- chloro-4-methylpyridine was used in place of compound E. _.-
  • Compound DB was prepared according to Example 1, except that 2- chloro-6-nitro ⁇ yridine was used in place of compound E.
  • Compound DE was prepared according to Example 7, except that compound H was used in place of compound N and 2-butynoic acid was used in place of compound I.
  • Compound BU was prepared according to Example 7, except that compound H was used in place of compound N and 2-heptynoic acid was used in place of compound I.
  • Example 41 Synthesis of Compound BS Compound BS was prepared accordmg to Example 7, except that compound H was used in place of compound N and 2-nonynoic acid was used in place of compound I.
  • Compound BX was prepared according to Example 10, except that 3,5- difluoroiodobenzene was used in place of compound Q.
  • Compound BY was prepared according to Example 10, except that 2,4- dimethoxyiodobenzene was used in place of compound Q.
  • Example 44 Synthesis of Compound DF
  • Compound DF was prepared according to Example 10, except that 3- fluoroiodobenzene was used in place of compound Q.
  • Compound BZ was prepared according to Example 10, except that 2- chloro-5-iodotoluene was used in place of compound Q.
  • Compound DI was prepared according to Example 10, except that 3- trifluoromethoxyiodobenzene was used in place of compound Q.
  • Compound CB was prepared according to Example 10, except that 5- chloro-2-methoxyiodobenzene was used in place of compound Q .
  • Compound CC was prepared according to Example 10, except that 2- fluoro-5-iodotoluene was used in place of compound Q.
  • Compound CD was prepared according to Example 10, except that 2,5- difluoroiodobenzene was used in place of compound Q.
  • Compound DN was prepared according to Example 10, except that 4-tert- butyliodobenzene was used in place of compound Q.
  • Example 57 Synthesis of Compound CE
  • Compound CE was prepared according to Example 10, except that 3- chloro-2-fluoroiodobenzene was used in place of compound Q.
  • Compound CJ was prepared from compound P according to Example 10, except that 3-iodoanisole was used in place of compound Q.
  • Compound CL was prepared from compound P according to Example 10, except that 2-iodotoluene was used in place of compound Q.
  • Compound CM was prepared from compound P according to Example 10, except that 4-iodotoluene was used in place of compound Q.
  • Example 62 Synthesis of Compound CP
  • Compound CP was prepared from compound P according to Example 10, except that 3-iodotoluene was used in place of compound Q.
  • Compound DQ was prepared according to Example 10, except that 2- fluoro-4-iodopyridine was used in place of compound Q.
  • Compound DS was prepared according to Example 10, except that 2- iodopyridine was used in place of compound Q.
  • Example 67 Synthesis of Compound DT
  • Compound DT was prepared according to Example 10, except that 3- iodopyridine was used in place of compound Q.
  • glial cultures were prepared from cortices of Sprague-Dawley 18 days old embryos. The cortices were dissected and then dissociated by trituration. The resulting cell homogenate was plated onto poly-D-lysine precoated T175 flasks (BIOCOAT, commercially available from Becton Dickinson and Company Inc. of Franklin Lakes, NJ) in Dulbecco's Modified Eagle's Medium (“DMEM,” pH 7.4), buffered with 25 mM HEPES, and supplemented with 15% fetal calf serum ("FCS,” commercially available from Hyclone Laboratories Inc. of Omaha, NE ), and incubated at 37°C and 5% CO 2 .
  • BIOCOAT poly-D-lysine precoated T175 flasks
  • DMEM Dulbecco's Modified Eagle's Medium
  • FCS fetal calf serum
  • FCS supplementation was reduced to 10%.
  • oligodendrocytes and microglia were removed by strongly tapping the sides of the flasks.
  • secondary astrocytes cultures were established by subplating onto 96 poly-D-lysine precoated T175 flasks (BIOCOAT) at a density of 65,000 cells/well in DMEM and 10% FCS.
  • the astrocytes were washed with serum free medium and then cultured in DMEM, without glutamate, supplemented with 0.5% FCS, 20 mM HEPES, 10 ng/mL epidermal growth factor ("EGF"), 1 mM sodium pyruvate, and IX penicillin/streptomycin at pH 7.5 for 3 to 5 days at 37°C and 5% CO 2.
  • DMEM serum free medium
  • FCS 20 mM HEPES
  • EGF epidermal growth factor
  • IX penicillin/streptomycin IX penicillin/streptomycin at pH 7.5 for 3 to 5 days at 37°C and 5% CO 2.
  • the procedure allows the expression ofthe mGluR5 receptor by astrocytes, as demonstrated by S. Miller et al, J. Neuroscience 15(9):6103-6109 (1995).
  • Assay Protocol After 3-5 days incubation with EGF, the astrocytes were washed with 127 mM NaCl, 5 mM KCl, 2 mM MgCl 2 , 700 mM NaH 2 PO 4 , 2 mM CaCl 2 , 5 mM NaHCO 3 , 8 mM HEPES, 10 mM Glucose at pH 7.4 ("Assay Buffer") and loaded with the dye Fluo-4 (commercially available from Molecular Probes Inc. of Eugene, OR) using 0.1 mL of Assay Buffer containing Fluo-4 (3 mM final).
  • Figure 1 represents a typical dose response curve, i.e., a single experiment, for Compound AA. In each experiment each data point was determined two times. These results show that Compound AA, an illustrative Piperazine Compound, binds to the mGluR5 receptor.
  • the following assay can be used to demonstrate that Piperazine Compounds bind to and modulate the activity of mGluR5 and, accordingly, are useful for treating or preventing, e.g., pain.
  • CHO-rat mGluR5 cells/well are plated into 96 well plate (Costar 3409, Black, clear bottom, 96 well, tissue culture treated) for an overnight incubation in Dulbecco's Modified Eagle's Medium (DMEM, pH 7.4) and supplemented with glutamine, 10% FBS, 1% Pen Strep, and 500ug/mL Geneticin.
  • DMEM Dulbecco's Modified Eagle's Medium
  • CHO-rat mGluR5 cells are washed and treated with Optimem medium and incubated for 1-4 hours prior to loading cells.
  • Cell plates are then washed with loading buffer (127 mM NaCl, 5 mM KCl, 2 mM MgCl 2 , 700 ⁇ M Na H 2 PO 4 , 2 mM CaCl 2 , 5 mM NaHCO 3 , 8 mM Hepes, and 10 mM glucose, pH 7.4) and then incubated with 3 ⁇ M Fluo 4 (commercially available from Molecular probes Inc. of Eugene, OR) in 0.1 mL of loading buffer. After 90 minutes of dye loading, the cells are then washed twice with 0.2 mL loading buffer and resuspended in 0.1 mL loading buffer.
  • loading buffer 127 mM NaCl, 5 mM KCl, 2 mM MgCl 2 , 700 ⁇ M Na H 2 PO 4 , 2 mM CaCl 2 , 5 mM NaHCO 3 , 8 mM Hepes, and 10 mM glucose, pH 7.4
  • 3 ⁇ M Fluo 4
  • the plates containing the CHO-rat mGluR5 cells are then transferred to a Fluorometric Imaging Plate Reader (FLIPR) (commercially available from Molecular Devices Corporation of Sunnyvale, CA) for the assessment of calcium mobilization flux in the presence of glutamate and in the presence or absence of test compounds.
  • FLIPR Fluorometric Imaging Plate Reader
  • DMSO solutions containing various concentrations ofthe test compound diluted in loading buffer 0.05 mL of 4X dilutions for the competition curves
  • fluorescence is monitored for 2 minutes.
  • 0.05 mL of 4X glutamate solution (agonist) is then added to each well to provide a final glutamate concentration in each well of 10 uM.
  • Test Animals Each experiment uses rats weighing between 200-260 g at the start ofthe experiment. The rats are group-housed and have free access to food and water at all times, except prior to oral administration of a Piperazine Compound when food is removed for 16 hours before dosing.
  • a control group acts as a comparison to rats treated with a Piperazine Compound. The control group is administered the carrier for the Piperazine Compound. The volume of carrier administered to the control group is the same as the volume of carrier and Piperazine Compound administered to the test group.
  • Tail flick latencies are defined as the interval between the onset ofthe thermal stimulus and the flick ofthe tail. Animals not responding within 20 seconds are removed from the tail flick unit and assigned a withdrawal latency of 20 seconds. Tail flick latencies are measured immediately before (pre-treatment) and 1, 3, and 5 hours following administration of a Piperazine Compound. Data are expressed as tail flick latency(s) and the percentage ofthe maximal possible effect (%> MPE), i.e., 20 seconds, is calculated as follows:
  • Acute pain can also be assessed by measuring the animal's response to noxious mechanical stimuli by determining the paw withdrawal threshold (PWT), as described below.
  • PWT paw withdrawal threshold
  • FCA Freund's complete adjuvant
  • the left hind paw of each animal is administered a 50 ⁇ L intraplantar injection of 50% FCA. 24 hour post injection, the animal is assessed for response to noxious mechanical stimuli by determining the PWT, as described below. Rats are then administered a single injection of 1, 3, 10 or 30 mg/Kg of either a Piperazine Compound, 30 mg/Kg indomethacin or carrier. Responses to noxious mechanical stimuli are then determined 1, 3, 5 and 24 hours post administration. Percentage reversal of hyperalgesia for each animal is defined as:
  • the partial sciatic nerve ligation model of neuropathic pain is used to produce neuropathic hyperalgesia in rats (Z. Seltzer et al, "A Novel Behavioral Model of Neuropathic Pain Disorders Produced in Rats by Partial Sciatic Nerve Injury," Pain 43:205-218 (1990)). Partial ligation ofthe left sciatic nerve is performed under isoflurane/O 2 inhalation anaesthesia.
  • the left thigh ofthe rat is shaved and the sciatic nerve exposed at high thigh level through a small incision and is carefully cleared of surrounding connective tissues at a site near the trocanther just distal to the point at which the posterior biceps semitendinosus nerve branches off of the common sciatic nerve.
  • a 7-0 silk suture is inserted into the nerve with a 3/8 curved, reversed-cutting mini-needle and tightly ligated so that the dorsal 1/3 to Vz ofthe nerve thickness is held within the ligature.
  • the wound is closed with a single muscle suture (4-0 nylon (Vicryl)) and vetbond tissue glue. Following surgery, the wound area is dusted with antibiotic powder.
  • the spinal nerve ligation model of neuropathic pain is used to produce mechanical hyperalgesia, thermal hyperalgesia and tactile allodynia in rats.
  • Surgery is performed under isoflurane/O 2 inhalation anaesthesia. Following induction of anaesthesia a 3 cm incision is made and the left paraspinal muscles are separated from the spinous process at the L 4 - S 2 levels. The L 6 transverse process is carefully removed with a pair of small rongeurs to identify visually the L 4 - L 6 spinal nerves.
  • the left L 5 (or L 5 and L 6 ) spinal nerve(s) is isolated and tightly ligated with silk thread.
  • a complete hemostasis is confirmed and the wound is sutured using non- absorbable sutures, such as nylon sutures or stainless steel staples.
  • Sham-treated rats undergo an identical surgical procedure except that the spinal nerve(s) is not manipulated. Following surgery animals are weighed, administered a subcutaneous (s.c.) injection of saline or ringers lactate, the wound area is dusted with antibiotic powder and they are kept on a warm pad until they recover from the anesthesia. Animals are then returned to their home cages until behavioral testing begins. The animals are assessed for response to noxious mechanical stimuli by determining PWT, as described below, immediately prior to and 1 , 3 and 5 hours after being administered a Piperazine
  • the animal can also be assessed for response to noxious thermal stimuli or for tactile allodynia, as described below.
  • the Chung model for neuropathic pain is described in S.H. Kim, "An Experimental Model for Peripheral Neuropathy Produced by Segmental Spinal Nerve Ligation in the Rat," Pain 50(3):355-363 (1992).
  • the paw pressure assay can be used to assess mechanical hyperalgesia.
  • hind paw withdrawal thresholds (PWT) to a noxious mechanical stimulus are determined using an analgesymeter (Model 7200, commercially available from Ugo Basile of Italy) as described in C. Stein, "Unilateral Inflammation ofthe Hindpaw in Rats as a Model of Prolonged Noxious Stimulation: Alterations in Behavior and Nociceptive Thresholds," Pharmacology Biochemistry and Behavior 31:451-455 (1988).
  • the maximum weight that can be applied to the hind paw is set at 250 g and the end point is taken as complete withdrawal ofthe paw.
  • PWT is determined once for each rat at each time point and only the affected (ipsilateral) paw is tested.
  • the plantar test can be used to assess thermal hyperalgesia.
  • hind paw withdrawal latencies to a noxious thermal stimulus are determined using a plantar test apparatus (commercially available from Ugo Basile of Italy) following the technique described by K. Hargreaves et al, "A New and Sensitive Method for Measuring Thermal Nociception in Cutaneous Hyperalgesia," Pain 32(l):77-88 (1988).
  • the maximum exposure time is set at 32 seconds to avoid tissue damage and any directed paw withdrawal from the heat source is taken as the end point. Three latencies are determined at each time point and averaged. Only the affected (ipsilateral) paw is tested.
  • Tactile Allodynia To assess tactile allodynia, rats are placed in clear, plexiglass compartments with a wire mesh floor and allowed to habituate for a period of at least 15 minutes. After habituation, a series of von Frey monofilaments are presented to the plantar surface ofthe left (operated) foot of each rat. The series of von Frey monofilaments consists of six monofilaments of increasing diameter, with the smallest diameter fiber presented first. Five trials are conducted with each filament with each trial separated by approximately 2 minutes. Each presentation lasts for a period of 4-8 seconds or until a nociceptive withdrawal behavior is observed. Flinching, paw withdrawal or licking ofthe paw are considered nociceptive behavioral responses.
  • the following assays can be used to demonstrate that Piperazine Compounds are useful for treating or preventing anxiety.
  • the elevated plus maze test or the shock-probe burying test can be used to assess the anxiolytic activity of Piperazine Compounds in rats or mice.
  • the Elevated Plus Maze Test The elevated plus maze consists of a platform with 4 arms, two open and two closed (50 x 10 x 50 cm enclosed with an open roof). Rats (or mice) are placed in the center ofthe platform, at the crossroad ofthe 4 arms, facing one ofthe closed arms. Time spent in the open arms vs the closed arms and number of open arm entries during the testing period are recorded. This test is conducted prior to drug administration and again after drug administration. Test results are expressed as the mean time spent in open arms and the mean number of entries into open arms. Known anxiolytic drugs increase both the time spent in open arms and number of open arm entries. The elevated plus maze test is described in D. Treit, "Animal Models for the Study of Anti-anxiety Agents: A Review," Neuroscience & Biobehavioral Reviews 9(2):203-222 (1985).
  • the Shock-Probe Burying Test For the shock-probe burying test the testing apparatus consists of a plexiglass box measuring 40x30x40 cm, evenly covered with approximately 5 cm of bedding material (odor absorbent kitty litter) with a small hole in one end through which a shock probe (6.5 cm long and 0.5 cm in diameter) is inserted. The plexiglass shock probe is helically wrapped with two copper wires through which an electric current is administered. The current is set at 2 mA. Rats are habituated to the testing apparatus for 30 min on 4 consecutive days without the shock probe in the box. On test day, rats are placed in one comer ofthe test chamber following drug administration.
  • bedding material odor absorbent kitty litter
  • the probe is not electrified until the rat touches it with its snout or fore paws, at which point the rat receives a brief 2 mA shock.
  • the 15 min testing period begins once the rat receives its first shock and the probe remains electrified for the remainder ofthe testing period.
  • the shock elicits burying behavior by the rat.
  • the duration of time the rat spends spraying bedding material toward or over the probe with its snout or fore paws is measured as well as the number of contact-induced shocks the rat receives from the probe.
  • Known anxiolytic drugs reduce the amount of burying behavior.
  • the following assays can be used to demonstrate that Piperazine Compounds are useful for treating or preventing an addictive disorder.
  • the condition place preference test or drug self-administration test can be used to assess the ability of Piperazine Compounds to attenuate the rewarding properties of known drugs of abuse.
  • the Condition Place Preference Test The apparatus for the conditioned place preference test consists of two large compartments (45 x 45 x 30 cm) made of wood with a plexiglass front wall. These two large compartments are distinctly different. Doors at the back of each large compartment lead to a smaller box (36 x 18 x 20 cm) box made of wood, painted grey, with a ceiling of wire mesh.
  • the two large compartments differ in terms of shading (white vs black), level of illumination (the plexiglass door of the white compartment is covered with aluminum foil except for a window of 7 x 7 cm), texture (the white compartment has a 3 cm thick floor board (40 x 40 cm) with nine equally spaced 5 cm diameter holes and the black has a wire mesh floor), and olfactory cues (saline in the white compartment and 1 mL of 10% acetic acid in the black compartment). On habituation and testing days, the doors to the small box remain open, giving the rat free access to both large compartments.
  • the first session that a rat is placed in the apparatus is a habituation session and entrances to the smaller grey compartment remain open giving the rat free access to both large compartments.
  • rats generally show no preference for either compartment.
  • rats are given 6 conditioning sessions. Rats are divided into 4 groups: carrier pre-treatment + carrier (control group), Piperazine Compound pre-treatment + carrier, carrier pre-treatment + morphine, Piperazine Compound pre-treatment + mo ⁇ hine.
  • carrier pre-treatment + carrier control group
  • Piperazine Compound pre-treatment + carrier carrier pre-treatment + morphine
  • Piperazine Compound pre-treatment + mo ⁇ hine are divided into 4 groups: carrier pre-treatment + carrier (control group), Piperazine Compound pre-treatment + carrier, carrier pre-treatment + morphine, Piperazine Compound pre-treatment + mo ⁇ hine.
  • the rat is injected with one ofthe drug combinations and confined to one compartment for 30 min.
  • the rat receives a carrier + carrier treatment and is confined to the other large
  • Each rat receives three conditioning sessions consisting of 3 drug combination-compartment and 3 carrier-compartment pairings.
  • the order of injections and the drug/compartment pairings are counterbalanced within groups.
  • rats are injected prior to testing (30 min to 1 hour) with either morphine or carrier and the rat is placed in the apparatus, the doors to the grey compartment remain open and the rat is allowed to explore the entire apparatus for 20 min.
  • the time spent in each compartment is recorded.
  • Known drugs of abuse increase the time spent in the drug-paired compartment during the testing session.
  • the Piperazine Compound blocks the acquisition of morphine conditioned place preference (reward)
  • Data will be analyzed as time spent in each compartment (drug combination-paired vs carrier-paired).
  • the experiment is repeated with a minimum of 3 doses of a Piperazine Compound.
  • the Drug Self- Administration Test is a standard commercially available operant conditioning chamber. Before drug trials begin rats are trained to press a lever for a food reward. After stable lever pressing behavior is acquired, rats are tested for acquisition of lever pressing for drug reward.
  • Rats are implanted with chronically indwelling jugular catheters for i.v. administration of compounds and are allowed to recover for 7 days before training begins. Experimental sessions are conducted daily for 5 days in 3 hour sessions. Rats are trained to self-administer a known drug of abuse, such as mo ⁇ hine. Rats are then presented with two levers, an "active" lever and an “inactive” lever. Pressing ofthe active lever results in drug infusion on a fixed ratio 1 (FRl) schedule (i.e., one lever press gives an infusion) followed by a 20 second time out period (signaled by illumination of a light above the levers). Pressing ofthe inactive lever results in infusion of excipient.
  • FRl ratio 1
  • Training continues until the total number of morphine infusions stabilizes to within ⁇ 10% per session. Trained rats are then used to evaluate the effect of Piperazine Compounds pre-treatment on drug self-administration. On test day, rats are pre-treated with a Piperazine Compound or excipient and then are allowed to self- administer drug as usual. If the Piperazine Compound blocks the rewarding effects of morphine, rats pre-treated with the Piperazine Compound will show a lower rate of responding compared to their previous rate of responding and compared to excipient pre- treated rats. Data is analyzed as the change in number of drug infusions per testing session (number of infusions during test session - number of infusions during training session).
  • Compounds bind to and modulate the activity of mGluR5 and, accordingly, are useful for treating or preventing, e.g., pain.
  • Functional assays for the characterization of mGluRl antagonistic properties are well known in the art. For example, the following procedure can be used.
  • cDNA encoding rat mGluRl a receptor is obtained from, e.g., Prof. S.
  • Nakanishi (Kyoto, Japan). It is transiently transfected into HEK-EBNA cells using a procedure described by Schlaeger et al, New Dev. New Appl. Anim. Cell Techn., Proc. ESACT Meet., 15 th a (1998), 105-112 and 117-120.
  • [Ca 2+ ] measurements are performed on mGluRl a transfected HEK-EBNA cells after incubation ofthe cells with Fluo-3 AM (0.5 ⁇ M final concentration) for 1 hour at 37°C followed by 4 washes with assay buffer (DMEM supplemented with Hank's salt and 20 mM HEPES.
  • Assay buffer DMEM supplemented with Hank's salt and 20 mM HEPES.
  • [Ca 2+ ] measurements are done using a flurometric imaging plate reader, e.g., FLIPR. 10 ⁇ M glutamate as agonist is used to evaluate the potency ofthe antagonists.
  • Increasing concentrations of antagonists are applied to the cells 5 minutes prior to application ofthe agonist.
  • the inhibition (antagonists) curves are fitted with appropriate software, for example, the four-parameter logistic equation giving IC 50 and Hill coefficient using the iterative nonlinear curve fitting software Origin from Microcal Software Inc., Northampton, MA.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Neurosurgery (AREA)
  • Engineering & Computer Science (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

L'invention concerne un composé de formule (I), dans laquelle A, R1, R2, R6, m et n sont décrits, ou un de ses sels pharmaceutiquement acceptable (un « composé de pipérazine »). L'invention concerne des compositions pharmaceutiques comprenant un composé de pipérazine, ainsi que des méthodes pour traiter la douleur, l'incontinence urinaire (UI), un trouble d'accoutumance, la maladie de Parkinson, le parkinsonisme, l'anxiété, l'épilepsie, un accident vasculaire cérébral, une crise d'épilepsie, des états pathologiques prurigineux, la psychose, un trouble cognitif, un déficit de la mémoire, des fonctions cérébrales limitées, la chorée de Huntington, la sclérose latérale amyotrophique, la démence, la rétinopathie, un spasme musculaire, une migraine, la dyskinésie et la dépression chez un animal comprenant l'administration à un animal nécessitant le traitement une quantité efficace d'un composé de pipérazine.
PCT/US2003/013964 2002-05-02 2003-05-02 Composes 1-(pyrid-2-yl)-piperazine utilises en tant qu'inhibiteur du recepteur de glutamate metabotropique WO2003093236A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2003228857A AU2003228857A1 (en) 2002-05-02 2003-05-02 1-(pyrid-2-yl)-piperazine compounds as metabotropic glutamate receptor inhibitor

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US37680302P 2002-05-02 2002-05-02
US60/376,803 2002-05-02
US10/000,000 2002-07-02

Publications (1)

Publication Number Publication Date
WO2003093236A1 true WO2003093236A1 (fr) 2003-11-13

Family

ID=29401406

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2003/013964 WO2003093236A1 (fr) 2002-05-02 2003-05-02 Composes 1-(pyrid-2-yl)-piperazine utilises en tant qu'inhibiteur du recepteur de glutamate metabotropique

Country Status (1)

Country Link
WO (1) WO2003093236A1 (fr)

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004067002A2 (fr) * 2003-01-30 2004-08-12 Recordati Ireland Limited Antagonistes selectifs du recepteur mglu5 destines au traitement de dysfonctionnements neuromusculaires du tractus urinaire inferieur
WO2005007641A1 (fr) * 2003-07-03 2005-01-27 Euro-Celtique S.A. Derives de 2-pyridine alkyne servant au traitement de la douleur
WO2005035500A2 (fr) * 2003-10-09 2005-04-21 Euro-Celtique S.A. Agents therapeutiques servant a traiter la douleur
WO2005058361A1 (fr) * 2003-12-19 2005-06-30 Astrazeneca Ab Utilisation d'antagonistes du recepteur mglur1 pour le traitement de reflux gastrooesophagien pathologique
WO2005080363A1 (fr) * 2004-02-18 2005-09-01 Astrazeneca Ab Composes acetyleniques de piperazine et leur utilisation en tant qu'antagonistes du recepteur metabotrope du glutamate
WO2006002981A1 (fr) * 2004-07-05 2006-01-12 Grünenthal GmbH 1-propiolylpiperazines substituees presentant une affinite pour le recepteur mglur5, utilisees pour le traitement de douleurs
WO2006109817A1 (fr) * 2005-04-06 2006-10-19 Banyu Pharmaceutical Co., Ltd. Dérivé de substituée pipérazine en 1,4
WO2006123257A2 (fr) 2005-05-18 2006-11-23 Addex Pharma Sa Derives de pyrrole utilises en tant que modulateurs allosteriques positifs des recepteurs de glutamate metabotropique
WO2007021573A1 (fr) * 2005-08-15 2007-02-22 Astrazeneca Ab Pipérazines substituées en tant qu'antagonistes du récepteur métabotropique du glutamate
DE102005062991A1 (de) * 2005-12-28 2007-07-05 Grünenthal GmbH Substituierte Thiazole und ihre Verwendung zur Herstellung von Arzneimitteln
WO2007079959A1 (fr) * 2005-12-28 2007-07-19 Grünenthal GmbH Amides de l'acide propiolique substitues et leur utilisation pour produire des analgesiques
WO2007087135A2 (fr) * 2006-01-17 2007-08-02 Astrazeneca Ab PIPERAZINES ET PIPERIDINES EN TANT QUE POTENTIALISATEURS mGluR5
WO2009015897A1 (fr) * 2007-08-02 2009-02-05 Recordati Ireland Limited Nouveaux composés hétérocycliques servant d'antagonistes du mglu5
JP2010520876A (ja) * 2007-03-09 2010-06-17 アストラゼネカ・アクチエボラーグ ピペラジンおよびピペリジンmGluR5増強剤
WO2011109398A2 (fr) 2010-03-02 2011-09-09 President And Fellows Of Harvard College Procédés et compositions pour le traitement du syndrome d'angelman et des troubles du spectre autistique
US8063216B2 (en) 2003-12-29 2011-11-22 The Trustees Of Columbia University In The City Of New York Photolabile compounds
US8101637B2 (en) 2004-06-17 2012-01-24 Addex Pharma Sa Alkynyl derivatives as modulators of metatropic glutamate receptors
WO2014117919A1 (fr) * 2013-02-04 2014-08-07 Merck Patent Gmbh Modulateurs allostériques positifs de mglur3
US9744236B2 (en) 2009-06-26 2017-08-29 The Trustees Of Columbia University In The City Of New York Photolabile compounds
DE102022104759A1 (de) 2022-02-28 2023-08-31 SCi Kontor GmbH Co-Kristall-Screening Verfahren, insbesondere zur Herstellung von Co-Kristallen

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4367335A (en) * 1981-08-03 1983-01-04 Mead Johnson & Company Thiazolidinylalkylene piperazine derivatives
JPS6310786A (ja) * 1986-07-01 1988-01-18 Sumitomo Pharmaceut Co Ltd 新規イミド誘導体およびその製造法
US5440048A (en) * 1991-01-10 1995-08-08 Hoechst-Roussel Pharmaceuticals Inc 1-piperazinyl-2-butenes and -2-butynes
WO1999002497A2 (fr) * 1997-07-11 1999-01-21 Novartis Ag Derives de pyridine

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4367335A (en) * 1981-08-03 1983-01-04 Mead Johnson & Company Thiazolidinylalkylene piperazine derivatives
JPS6310786A (ja) * 1986-07-01 1988-01-18 Sumitomo Pharmaceut Co Ltd 新規イミド誘導体およびその製造法
US5440048A (en) * 1991-01-10 1995-08-08 Hoechst-Roussel Pharmaceuticals Inc 1-piperazinyl-2-butenes and -2-butynes
WO1999002497A2 (fr) * 1997-07-11 1999-01-21 Novartis Ag Derives de pyridine

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
ISHIZUMI, KIKUO ET AL: "Synthesis and anxiolytic activity of N-substituted cyclic imides (1R*,2S*,3R*,4S*)-N-[4-[4-(2-pyrimidinyl)-1-piperazinyl]butyl]-2,3- bicyclo[2.2.1]heptanedicarboximide (tandospirone) and related compounds", CHEMICAL & PHARMACEUTICAL BULLETIN (1991), 39(9), 2288-300, XP002103452 *

Cited By (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004067002A2 (fr) * 2003-01-30 2004-08-12 Recordati Ireland Limited Antagonistes selectifs du recepteur mglu5 destines au traitement de dysfonctionnements neuromusculaires du tractus urinaire inferieur
WO2004067002A3 (fr) * 2003-01-30 2004-11-25 Recordati Chem Pharm Antagonistes selectifs du recepteur mglu5 destines au traitement de dysfonctionnements neuromusculaires du tractus urinaire inferieur
WO2005007641A1 (fr) * 2003-07-03 2005-01-27 Euro-Celtique S.A. Derives de 2-pyridine alkyne servant au traitement de la douleur
WO2005035500A2 (fr) * 2003-10-09 2005-04-21 Euro-Celtique S.A. Agents therapeutiques servant a traiter la douleur
WO2005035500A3 (fr) * 2003-10-09 2005-05-19 Euro Celtique Sa Agents therapeutiques servant a traiter la douleur
WO2005058361A1 (fr) * 2003-12-19 2005-06-30 Astrazeneca Ab Utilisation d'antagonistes du recepteur mglur1 pour le traitement de reflux gastrooesophagien pathologique
US8063216B2 (en) 2003-12-29 2011-11-22 The Trustees Of Columbia University In The City Of New York Photolabile compounds
US9273032B2 (en) 2003-12-29 2016-03-01 The Trustees Of Columbia University In The City Of New York Photolabile compounds
US9688708B2 (en) 2003-12-29 2017-06-27 The Trustees Of Columbia University In The City Of New York Photolabile compounds
WO2005080363A1 (fr) * 2004-02-18 2005-09-01 Astrazeneca Ab Composes acetyleniques de piperazine et leur utilisation en tant qu'antagonistes du recepteur metabotrope du glutamate
US8883826B2 (en) 2004-06-17 2014-11-11 Addex Pharma Sa Alkynyl derivatives as modulators of metabotropic glutamate receptors
US8674106B2 (en) 2004-06-17 2014-03-18 Addex Pharma Sa Alkynyl derivatives as modulators of metabotropic glutamate receptors
US8101637B2 (en) 2004-06-17 2012-01-24 Addex Pharma Sa Alkynyl derivatives as modulators of metatropic glutamate receptors
DE102004032567A1 (de) * 2004-07-05 2006-03-02 Grünenthal GmbH Substituierte 1-Propiolyl-piperazine
US7300939B2 (en) 2004-07-05 2007-11-27 Gruenenthal Gmbh Substituted 1-propiolylpiperazine compounds, their preparation and use
JP2008505149A (ja) * 2004-07-05 2008-02-21 グリューネンタール・ゲゼルシャフト・ミット・ベシュレンクテル・ハフツング 置換1−プロピオリルピペラジン
WO2006002981A1 (fr) * 2004-07-05 2006-01-12 Grünenthal GmbH 1-propiolylpiperazines substituees presentant une affinite pour le recepteur mglur5, utilisees pour le traitement de douleurs
WO2006109817A1 (fr) * 2005-04-06 2006-10-19 Banyu Pharmaceutical Co., Ltd. Dérivé de substituée pipérazine en 1,4
US8101618B2 (en) 2005-04-06 2012-01-24 Msd K.K. 1,4-substituted piperazine derivatives
WO2006123257A2 (fr) 2005-05-18 2006-11-23 Addex Pharma Sa Derives de pyrrole utilises en tant que modulateurs allosteriques positifs des recepteurs de glutamate metabotropique
WO2006123257A3 (fr) * 2005-05-18 2007-05-03 Addex Pharmaceuticals Sa Derives de pyrrole utilises en tant que modulateurs allosteriques positifs des recepteurs de glutamate metabotropique
EA014081B1 (ru) * 2005-05-18 2010-08-30 Аддекс Фарма Са Производные пиррола в качестве положительных аллостерических модуляторов метаботропных глутаматных рецепторов
WO2007021573A1 (fr) * 2005-08-15 2007-02-22 Astrazeneca Ab Pipérazines substituées en tant qu'antagonistes du récepteur métabotropique du glutamate
DE102005062991A1 (de) * 2005-12-28 2007-07-05 Grünenthal GmbH Substituierte Thiazole und ihre Verwendung zur Herstellung von Arzneimitteln
WO2007079960A1 (fr) * 2005-12-28 2007-07-19 Grünenthal GmbH Thiazoles substitues et leur utilisation pour produire des medicaments
US7871999B2 (en) 2005-12-28 2011-01-18 Gruenenthal Gmbh Substituted thiazoles and their use for producing drugs
US8008304B2 (en) 2005-12-28 2011-08-30 Grüenthal GmbH Substituted propiolic acid amides and their use for producing drugs
WO2007079959A1 (fr) * 2005-12-28 2007-07-19 Grünenthal GmbH Amides de l'acide propiolique substitues et leur utilisation pour produire des analgesiques
JP2009522222A (ja) * 2005-12-28 2009-06-11 グリュネンタール・ゲゼルシャフト・ミト・ベシュレンクテル・ハフツング 置換チアゾールおよび医薬の製造へのその使用
JP2009524702A (ja) * 2006-01-17 2009-07-02 アストラゼネカ・アクチエボラーグ mGluR5増強因子としてのピペラジン類及びピペリジン類
US7935703B2 (en) 2006-01-17 2011-05-03 Astrazeneca Ab Piperazines and piperidines as Mglur5 potentiators
WO2007087135A2 (fr) * 2006-01-17 2007-08-02 Astrazeneca Ab PIPERAZINES ET PIPERIDINES EN TANT QUE POTENTIALISATEURS mGluR5
WO2007087135A3 (fr) * 2006-01-17 2007-11-01 Astrazeneca Ab PIPERAZINES ET PIPERIDINES EN TANT QUE POTENTIALISATEURS mGluR5
JP2010520876A (ja) * 2007-03-09 2010-06-17 アストラゼネカ・アクチエボラーグ ピペラジンおよびピペリジンmGluR5増強剤
US8518916B2 (en) 2007-08-02 2013-08-27 Recordati Ireland Limited Heterocyclic derivatives as M-GLU5 antagonists
EA018328B1 (ru) * 2007-08-02 2013-07-30 Рекордати Айерленд Лимитед Новые гетероциклические соединения в качестве антагонистов метаботропных глутаматных рецепторов 5-го подтипа (мглу5)
WO2009015897A1 (fr) * 2007-08-02 2009-02-05 Recordati Ireland Limited Nouveaux composés hétérocycliques servant d'antagonistes du mglu5
JP2010535165A (ja) * 2007-08-02 2010-11-18 レコルダーティ アイルランド リミテッド mGlu5拮抗薬としての新規複素環系化合物
CN101821256A (zh) * 2007-08-02 2010-09-01 瑞蔻达蒂爱尔兰有限公司 作为mGlu5拮抗剂的新型杂环化合物
US9744236B2 (en) 2009-06-26 2017-08-29 The Trustees Of Columbia University In The City Of New York Photolabile compounds
WO2011109398A2 (fr) 2010-03-02 2011-09-09 President And Fellows Of Harvard College Procédés et compositions pour le traitement du syndrome d'angelman et des troubles du spectre autistique
WO2014117919A1 (fr) * 2013-02-04 2014-08-07 Merck Patent Gmbh Modulateurs allostériques positifs de mglur3
CN104955807A (zh) * 2013-02-04 2015-09-30 默克专利股份公司 mGluR3的正变构调节剂
JP2016506938A (ja) * 2013-02-04 2016-03-07 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツングMerck Patent Gesellschaft mit beschraenkter Haftung mGluR3の正のアロステリックモジュレーター
AU2014211727B2 (en) * 2013-02-04 2018-07-26 Prexton Therapeutics Sa Positive allosteric modulators of mGluR3
US10059671B2 (en) 2013-02-04 2018-08-28 Prexton Therapeutics Sa Positive allosteric modulators of mGluR3
DE102022104759A1 (de) 2022-02-28 2023-08-31 SCi Kontor GmbH Co-Kristall-Screening Verfahren, insbesondere zur Herstellung von Co-Kristallen

Similar Documents

Publication Publication Date Title
US6864261B2 (en) Therapeutic agents useful for treating pain
CA2533509C (fr) Composes de piperidine et compositions pharmaceutiques les contenant
KR100896497B1 (ko) 통증 치료에 유용한 치료제
EP1583763A1 (fr) Derives de benzoazolylpiperazine presentant une activite antagoniste vis-a-vis de mglur1 et de mglur5
WO2004011441A1 (fr) Derives de pyridazinylpiperazine pour traiter la douleur
EP1562936A2 (fr) Derives de piperazine therapeutiques
WO2005056524A2 (fr) Agents therapeutiques pour traitement de la douleur
WO2004002983A2 (fr) Agents therapeutiques utiles pour le traitement de la douleur
EP1664041A1 (fr) Composes phenyl-carboxamide utiles pour traiter la douleur
EP1664016A2 (fr) Agents therapeutiques utiles pour traiter la douleur
WO2003093236A1 (fr) Composes 1-(pyrid-2-yl)-piperazine utilises en tant qu'inhibiteur du recepteur de glutamate metabotropique
CA2534088C (fr) Composes pyridyle 2,3-substitues utiles dans le traitement de la douleur
KR100867188B1 (ko) 통증 치료에 유용한 피페라진
EP1542991A1 (fr) Composes de pyrimidine en tant qu'agents therapeutiques
EP1641775A1 (fr) Derives de 2-pyridine alkyne servant au traitement de la douleur
WO2005035500A2 (fr) Agents therapeutiques servant a traiter la douleur

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

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

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP

DPE2 Request for preliminary examination filed before expiration of 19th month from priority date (pct application filed from 20040101)