WO2005056524A2 - Agents therapeutiques pour traitement de la douleur - Google Patents

Agents therapeutiques pour traitement de la douleur Download PDF

Info

Publication number
WO2005056524A2
WO2005056524A2 PCT/US2004/041378 US2004041378W WO2005056524A2 WO 2005056524 A2 WO2005056524 A2 WO 2005056524A2 US 2004041378 W US2004041378 W US 2004041378W WO 2005056524 A2 WO2005056524 A2 WO 2005056524A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
halo
alkyl
membered
animal
Prior art date
Application number
PCT/US2004/041378
Other languages
English (en)
Other versions
WO2005056524A3 (fr
Inventor
Bin Shao
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.
Publication of WO2005056524A2 publication Critical patent/WO2005056524A2/fr
Publication of WO2005056524A3 publication Critical patent/WO2005056524A3/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems

Definitions

  • the present invention relates to Piperazine Compounds, compositions comprising an effective amount of Piperazine Compound and methods for treating or preventing a Condition such as pain, comprising administering to an animal in need thereof an effective amount of a Piperazine Compound.
  • Pain is the most common symptom for which patients seek medical advice and treatment. Pain can be acute or chronic. While acute pain is usually self-limited, chronic pain persists for 3 months or longer and can lead to significant changes in a patient's personality, lifestyle, functional ability and overall quality of life (K.M. Foley, Pain, in Cecil Textbook of Medicine 100-107 (J.C. Bennett and F. Plum eds., 20th ed. 1996)). Chronic pain can be classified as either nociceptive or neuropathic. 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 central nervous system and is maintained by aberrant somatosensory processing.
  • 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.
  • neuropathic pain which can be difficult to treat, has also been treated with anti-epileptics (e.g., gabapentin, carbamazepine, valproic acid, topiramate, phenytoin), NMDA antagonists (e.g., ketamine, dextromethorphan), topical lidocaine (for post-herpetic neuralgia), and tricyclic antidepressants (e.g., fluoxetine, sertraline and amitriptyline). Ul is uncontrollable urination, generally caused by bladder-detrusor-muscle instability. Ul affects people of all ages and levels of physical health, both in health care settings and in the community at large.
  • anti-epileptics e.g., gabapentin, carbamazepine, valproic acid, topiramate, phenytoin
  • NMDA antagonists e.g., ketamine, dextromethorphan
  • Physiologic bladder contraction results in large part from acetylcholine-induced stimulation of post-ganglionic muscarinic-receptor sites on bladder smooth muscle.
  • Treatments for Ul 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 anticholinergic
  • U.S. Patent No. 5,556,838 to Mayer et al. discloses the use of nontoxic NMDA-blocking agents co-administered with an addictive substance to prevent the development of tolerance or withdrawal symptoms.
  • U.S. Patent No. 5,574,052 to Rose et al. discloses co-administration of an addictive substance with an antagonist to partially block the pharmacological effects of the addictive substance.
  • 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.
  • Patent No. 5,556,837 to Nestler et al. discloses infusing BDNF or NT-4 growth factors to inhibit or reverse neurological adaptive changes that correlate with behavioral changes in an addicted individual.
  • 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 tolerance.
  • 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 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.
  • benzodiazepines are the most commonly used anti-anxiety agents for generalized anxiety disorder.
  • Benzodiazepines 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 of the mGluR5 receptor have also been shown to exert anxiolytic and anti-depressant activity in in vivo animal models (E.
  • 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 hyperplasia; 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 scarlatiniform rash, morbilliform rash, Stevens- ohnson syndrome, systemic lupus erythematosus, and hepatic necrosis; and hematological reactions such as red-cell aplasia, agranulocytosis, thrombocytopenia, aplastic anemia, and megaloblastic anemia.
  • Symptoms of strokes vary depending on what part of the brain is affected. Symptoms include loss of or abnormal sensations in an arm or leg or one side of the 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 of the 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.
  • 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. Tlie Merck Manual of Medical Information 352-355 (R. Berkow ed., 1997). Pruritus is an unpleasant sensation that prompts scratching. Conventionally, 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
  • X is -O-, -C(R (R 2 )-, -N(H , -N -GOalkyl)-,
  • Y and Z are, independently, -C(O)- or -C(R (R 2 )-; each R t and R 2 is independently: (a) -H, or (b) linear or branched -(CrC 6 )alkyl, ⁇ (C 3 -C 8 )cycloalkyl, phenyl or naphthyl, each of which is unsubstituted or substituted with one or more R 9 groups; m is 0, 1 or 2; R 3 and R 4 are independently -CH 2 -, -CH(CH 3 )- or -C(O)-; each R 5 is independently: (a) -halo, -CN, -OH, -NO 2 , -O(C 1 -C 6 )alkyl or -NH 2 ; or (b) -(C ⁇ -C ⁇ o)alkyl, -(C 2 -C 10 )alkenyl, -(C 2 -C 10 )alkyn
  • each R 9 is independently -(C 1 -C 6 )alkyl, -(C 2 -C 6 )alkenyl, -(C 2 -C 6 )alkynyl, -(C 3 - C 8 )cycloalkyl, -(C 5 -Cs)cycloalkenyl, -phenyl or -(3- to 5 ⁇ membered)heterocycle;
  • Rio is independently -H or -( -C ⁇ alkyl;
  • Rn and R 12 are taken together with the carbon to which they are attached to form a ⁇ (C 3 -C 8 )cycloalkyl group;
  • each R 13 is independently
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof is useful for treating or preventing pain, Ul, 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 further relates to a synthetic intermediate that is useful for making a Piperazine Compound.
  • the invention further relates to a method for making a compound of formula A':
  • n' is 0 or 1
  • Q is I, Br, Cl or F
  • W is -( - C 10 )alkyl, -(C 2 -C 10 )alkenyl, -(C 2 -C 10 )alkynyl, -(C 3 -C 8 )cycloalkyl, -(C 8 -C 14 )bicycloalkyl, -(C 8 -C 1 )tricycloalkyl, -(Cs-C 1 o)cycloalkenyl, -(C 8 -C 14 )bicycloalkenyl, -(C 8 - C 14 )tricycloalkenyl, -(3- to 7-membered)heterocycle,
  • 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 in an animal.
  • the invention also relates to methods for treating a Condition, comprising administering to an animal in need thereof an effective amount of a Piperazine Compound.
  • the invention also 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 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 kit may further comprise printed instructions for using the Piperazine Compound to treat or prevent any of the aforementioned Conditions.
  • each R 5 when present, can be bonded to either of the available carbons of the pyridine ring, i.e., as indicated in the figure above, the carbon atom at the 5-position or the carbon atom at the 6-position.
  • the Piperazine Compound has only one R5 group, and that R 5 group is attached to the 5-position carbon atom of the pyridine ring.
  • the Piperazine Compound has only one R 5 group, that R 5 group is attached to the 6-position carbon atom of the pyridine ring, and the 6-position R 5 group is selected from: (a) -halo, -CN, -OH, -NO 2 , -O(C 1 -C 6 )alkyl or -NH 2 , or (b) -(C ⁇ -C ⁇ o)alkyl, -(C 2 -C 10 )alkenyl, -(C 2 -C 10 )alkynyl, -(C 3 - C 8 )cycloalkyl, -(C 8 -C 14 )bicycloalkyl, -(C 8 -C 14 )tricycloalkyl, -(Cs- c cycloalkenyl, -(
  • the Piperazine Compound has only one R 5 group, that R5 group is attached to the 5-position carbon atom of the pyridine ring, and the 5-position R 5 group is selected from -(C 1 -C 1 o)alkyl, -(C 2 -C 1 o)alkenyl, -(C 2 - o)alkynyl, -(C 3 - C 8 )cycloalkyl, -(C 8 -C 1 )bicycloalkyl, -(C 8 -C 14 )tricycloalkyl, -(C5-C 1 o)cycloalkenyl, -(C 8 - C 1 )bicycloalkenyl, -(C 8 -C 1 )tricycloalkenyl, -(3- to 7-membered)heterocycle, -(7- to 10- membered)bicycloheterocycle, -phenyl, -naphth
  • n 2
  • an R 5 group is attached to the 6-position carbon atom of the pyridine ring, the 6-position R 5 group is selected from: (a) -halo, -CN, -OH, -NO 2 , -O(d-C 6 )alkyl or -NH 2 , or (b) -(C ⁇ -C 10 )alkyl, -(C 2 -C 10 )alkenyl, ⁇ (C 2 -C 10 )alkynyl, -(C 3 -
  • R is phenyl.
  • R 6 is benzo(l,3)dioxolyl.
  • X is -N(H)- or -N((C ⁇ -C4)alkyl)-.
  • X is O.
  • n is 0.
  • m is 0.
  • m is 1.
  • R 3 and 1 ⁇ are each -CH 2 -.
  • A is -C(O)-.
  • Z is -C(O)-.
  • Y is -C(O)-.
  • Y is -C(CH 3 ) 2 -.
  • A is -C(O)- and n is 0.
  • A is -C(O)-; n is 0; and R 3 and R 4 are independently -CH 2 - or -CH(CH 3 )-.
  • A is -C(O)-; n is 0; and R 3 and R 4 are each -CH 2 -.
  • A is -C(O)-; n is 2; and R 3 and R 4 are independently -CH 2 - or -CH(CH 3 )-.
  • A is -C(O)-; n is 2; and R 3 and R 4 are each -CH 2 -.
  • A is -C(O)-; m is 0; Z is -C(O)-; and X is -CH 2 -.
  • A is -C(O)-; m is 0; Z is -C(O)-; and X is O.
  • A is -C(O)-; m is 0; Y and Z are both -C(O)-; and X is -CH 2 -.
  • A is -C(O)-; m is 0; Y and Z are both -C(O)-; and X is O.
  • A is -C(O)-; m is 0; Y is -C(Ri)(R 2 )-; Z is -C(O)-; and X is -CH 2 -.
  • A is -C(O)-; m is 0; Y is -C(R 1 )(R 2 )-; Z is -C(O)-; and X is O.
  • R 3 is -CH(CH 3 )- and the R 3 carbon atom to which the methyl group is attached has the (R) configuration.
  • R 3 is -CH(CH 3 )- and the R 3 carbon atom to which the methyl group is attached has the (S) configuration.
  • R 4 is -CH(CH 3 )- and the R 4 carbon atom to which the methyl group is attached has the (R) configuration.
  • R is -CH(CH 3 )- and the R 4 carbon atom to which the methyl group is attached has the (S) configuration.
  • R 3 and I are each -CH(CH 3 )- and the R 3 and R 4 carbon atoms to which each methyl group is attached both have the (R) configuration.
  • R 3 and 1 ⁇ . are each -CH(CH 3 )- and the R 3 and R 4 carbon atoms to which each methyl group is attached both have the (S) configuration.
  • R 3 and I are each -CH(CH 3 )- and one of the R 3 and R 4 carbon atoms to which a methyl group is attached has the (S) configuration and the other has the (R) configuration.
  • R 3 and R are each -CH(CH 3 )- and the R 3 carbon atom to which a methyl group is attached has the (S) configuration and the R 4 carbon atom to which a methyl group is attached has the (R) configuration.
  • both R 3 and R 4 are each -CH(CH )- and the R 4 carbon atom to which a methyl group is attached has the (S) configuration and the R 3 carbon atom to which a methyl group is attached has the (R) configuration.
  • X is -C(R ⁇ )(R 2 )- and one of R and R 2 is hydrogen and the other is linear or branched -(C 1 -C 6 )alkyl, -(C 3 -C 8 )cycloalkyl, phenyl or naphthyl, each of which is unsubstituted or substituted with one or more R 9 groups, where the carbon atom to which Ri and R 2 are attached has the (R) configuration.
  • X is -C(R (R 2 )- and one of R ⁇ and R 2 is hydrogen and the other is linear or branched -(Cr C 6 )alkyl, -(C 3 -C 8 )cycloalkyl, phenyl or naphthyl, each of which is unsubstituted or substituted with one or more R 9 groups, where the carbon atom to which R and R 2 are attached has the (S) configuration.
  • Y is -C(R_)(R 2 )- and one of Ri and R 2 is hydrogen and the other is linear or branched -(C 1 -C 6 )alkyl, -(C 3 -C 8 )cycloalkyl, phenyl or naphthyl, each of which is unsubstituted or substituted with one or more R groups, where the carbon atom to which Ri and R 2 are attached has the (R) configuration.
  • Y is -C(R (R 2 )- and one of R ⁇ and R 2 is hydrogen and the other is linear or branched -(Q- C 6 )alkyl, -(C 3 -C 8 )cycloalkyl, phenyl or naphthyl, each of which is unsubstituted or substituted with one or more R groups, where the carbon atom to which R and R 2 are attached has the (S) configuration.
  • Z is -C(R_)(R 2 )- and one of Rt and R 2 is hydrogen and the other is linear or branched -(C 1 -C 6 )alkyl, -(C 3 -C 8 )cycloalkyl, phenyl or naphthyl, each of which is unsubstituted or substituted with one or more R groups, where the carbon atom to which Ri and R 2 are attached has the (R) configuration.
  • Z is -C(R 1 )(R 2 )- and one of R ⁇ and R 2 is hydrogen and the other is linear or branched -(Ci- C 6 )alkyl, -(C 3 -C 8 )cycloalkyl, phenyl or naphthyl, each of which is unsubstituted or substituted with one or more R 9 groups, where the carbon atom to which R ⁇ and R 2 are attached has the (S) configuration.
  • A is -CH(C ⁇ -C 4 alkyl)- and the carbon atom of A to which the C 1 -C 4 alkyl group is attached has the (R) configuration.
  • A is -CH(C 1 -C 4 alkyl)- and the carbon atom of A to which the C 1 -C 4 alkyl group is attached has the (S) configuration. In one embodiment, A is -CH(phenyl)- and the carbon atom of A to which the phenyl group is attached has the (R) configuration. In another embodiment, A is
  • each R 5 when present, can be bonded to either of the available carbons of the pyridine ring.
  • the Piperazine Compound has only one R 5 group, and that R 5 group is attached to the 5-position carbon atom of the pyridine ring.
  • the Piperazine Compound has only one R 5 group, that R 5 group is attached to the 6-position carbon atom of the pyridine ring, and the 6-position R 5 group is selected from: (a) -halo, -CN, -OH, -NO 2 , -O(C 1 -C 6 )alkyl or -NH 2 , or (b) -(C 1 -C 10 )alkyl, -(C 2 -C 10 )alkenyl, -(C 2 -C 10 )alkynyl, -(C 3 -
  • the Piperazine Compound has only one R 5 group, that R 5 group is attached to the 5-position carbon atom of the pyridine ring, and the 5-position R 5 group is selected from -(C 1 -C 1 o)alkyl, -(C 2 -Cio)alkenyl, -(C 2 -C 1 o)alkynyl, -(C 3 - C 8 )cycloalkyl, -(C 8 -C 14 )bicycloalkyl, -(C 8 -C 14 )tricycloalkyl, -(C 5 -C 10 )cycloalkenyl, -(C 8 - C 14 )bicycloalkenyl, -(C 8 -C 14 )tricycloalkenyl, -(3- to 7-membered)heterocycle, -(7- to 10- membered)bicycloheterocycle, -phenyl, -naph
  • an R 5 group is attached to the 6-position carbon atom of the pyridine ring, the 6-position R 5 group is selected from: (a) -halo, -CN, -OH, -NO , -O(C 1 -C 6 )alkyl or -NH 2 , or (b) -(C 1 -C 10 )alkyl, -(C 2 -C 10 )alkenyl, -(C 2 -C 10 )alkynyl, -(C 3 - C 8 )cycloalkyl, -(C 8 -C 1 )bicycloalkyl, -(C 8 -C 14 )tricycloalkyl, -(C 5 -C 10 )cycloalkenyl, -(C 8 - C 1 )bicycloalkenyl, -(C 8 -C 14 )tricycloalkenyl, -(3- to 7-membered)
  • R 6 is phenyl. In another embodiment, R 6 is benzo(l,3)dioxolyl. In another embodiment, X is -N(H)- or -N ⁇ -G alkyl)-. In another embodiment, n is 0. In another embodiment, R 3 and R 4 are each -CH 2 -. In another embodiment, A is -C(O) ⁇ . In another embodiment, A is -C(O)- and n is 0. In another embodiment, A is -C(O)-; n is 0; and R and R 4 are independently -CH 2 - or -CH(CH 3 )-.
  • each R 5 when present, can be bonded to either of the available carbons of the pyridine ring.
  • the Piperazine Compound has only one R 5 group, and that R 5 group is attached to the 5-position carbon atom of the pyridine ring.
  • the Piperazine Compound has only one R 5 group, that R 5 group is attached to the 6-position carbon atom of the pyridine ring, and the 6-position
  • R 5 group is selected from: (a) -halo, -CN, -OH, -NO 2 , -O(C r C 6 )alkyl or -NH 2 , or . (b) -(C_-C 10 )alkyl, -(C 2 -C 10 )alkenyl, -(C 2 -C 10 )alkynyl, -(C 3 - C 8 )cycloalkyl, -(C 8 -C 14 )bicycloalkyl, -(C 8 -C 14 )tricycloalkyl, -(C 5 -C 1 o)cycloalkenyl, -(C 8 - C 14 )bicycloalkenyl, -(C 8 -C 14 )tricycloalkenyl, -(3- to 7-membered)heterocycle, -(7- to 10- membered)bicycloheterocycle, -phenyl
  • the Piperazine Compound has only one R 5 group, that R 5 group is attached to the 5-position carbon atom of the pyridine ring, and the 5-position R 5 group is selected from -(C 1 -C 1 o)alkyl, -(C 2 -C 1 o)alkenyl, -(C 2 -C 1 o)alkynyl, -(C 3 - C 8 )cycloalkyl, -(C 8 -C 14 )bicycloalkyl, -(C 8 -C 14 )tricycloalkyl, -(C 5 -C 1 o)cycloalkenyl, -(C 8 - C 14 )bicycloalkenyl, -(C 8 -C 14 )tricycloalkenyl, -(3- to 7-membered)heterocycle, -(7- to 10- membered)bicycloheterocycle, -phenyl, -na
  • an R 5 group is attached to the 6-position carbon atom of the pyridine ring, the 6-position R 5 group is selected from: (a) -halo, -CN, -OH, -NO 2 , -O(C C 6 )alkyl or -NH 2 , or (b) -(C 1 -C 10 )alkyl, -(C 2 -C 10 )alkenyl, -(C 2 -C 10 )alkynyl, -(C 3 - C 8 )cycloalkyl, -(C 8 -C 1 )bicycloalkyl, -(C 8 -C 1 )tricycloalkyl, -(C5-Cio)cycloalkenyl, -(C 8 - C 14 )bicycloalkenyl, -(C 8 -C 1 )tricycloalkenyl, -(3- to 7-membered)hetero
  • R 6 is phenyl. In another embodiment, R 6 is benzo(l,3)dioxolyl. In another embodiment, X is -N(H)- or -N((C 1 -C 4 )alkyl)-. In another embodiment, n is 0. In another embodiment, R 3 and R 4 . are each -CH 2 -. In another embodiment, A is -C(O)-. In another embodiment, A is -C(O)- and n is 0. In another embodiment, A is -C(O)-; n is 0; and R 3 and R 4 are independently -CH 2 - or -CH(CH 3 )-.
  • A is -C(O)-; n is 0; and R 3 and R 4 are each -CH 2 -. In another embodiment, A is -C(O)-; n is 2; and R 3 and R 4 are independently -CH 2 - or -CH(CH 3 )-. In another embodiment, A is -C(O)-; n is 2; and R 3 and R 4 are each -CH 2 -. In another embodiment, A is -C(O)- and X is -CH 2 -. In another embodiment, A is -C(O)- and X is O.
  • Illustrative Piperazine Compounds of formula (I) are listed below in Tables 1-8.
  • 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 -( - C!o)alkyls include - so-propyl, -sec-butyl, -iso-butyl, -tert-butyl, -tso-pentyl, -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-dimethylhexy
  • -(C 1 -C 6 )alkyl means a saturated straight chain or branched non-cyclic hydrocarbon having from 1 to 6 carbon atoms.
  • Representative saturated straight chain -(CrC ⁇ alkyls include -methyl, -ethyl, -n-propyl, -n-butyl, -n-pentyl, and -n-hexyl.
  • Representative saturated branched -(C 1 -C 6 )alkyls include -.so-propyl, -.sec-butyl, -iso-butyl, -tert-butyl, -zso-pentyl, -2-methylbutyl, -3-methylbutyl, -2,2-dimethylbutyl, -2,3-dimethylbutyl, -2-methylpentyl, -3-methylpentyl, -4-methylpentyl and the like.
  • "-(d-C ⁇ alkyl” means a saturated straight chain or branched non-cyclic hydrocarbon having from 1 to 4 carbon atoms.
  • Representative saturated straight chain -( -G alkyls include -methyl, -ethyl, -n-propyl, and -n-butyl.
  • Representative saturated branched -( -G ⁇ alkyls include -.so-propyl, -sec-butyl, -iso-butyl, and -tert-butyl.
  • "-(C 2 -C 1 o)alkenyl” means a straight chain or branched non-cyclic hydrocarbon having from 2 to 10 carbon atoms and including at least one carbon-carbon double bond.
  • Representative straight chain and branched (C 2 -C 1 o)alkyls include 1- ⁇ entenyl, -2-pentenyl, -3 -methyl- 1-butenyl, -2-methyl-2-butenyl, -2,3-dimethyl-2-butenyl, -1-hexenyl, -2-hexenyl, -3-hexenyl, -1-heptenyl, -2-he ⁇ tenyl, -3-heptenyl, -1-octenyl, -2-octenyl, -3-octenyl, -1-nonenyl, -2-nonenyl, -3-nonenyl, -1-decenyl, -2-decenyl, -3-decenyl and the like.
  • -(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 -C6)alkenyls include -vinyl, -allyl, -1-butenyl, -2-butenyl, -iso-butylenyl, -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 1 o)alkynyl means a straight chain or branched non-cyclic hydrocarbon having from 2 to 10 carbon atoms and including at least one carbon-carbon triple bond.
  • Representative straight chain and branched -(C 2 -C 1 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 least 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 8 )cycloalkyl means a saturated cyclic hydrocarbon having from 3 to 8 carbon atoms.
  • Representative (C 3 -C 8 )cycloalkyls include -cyclopropyl, -cyclobutyl, -cyclopentyl, -cyclohexyl, -cycloheptyl, and -cyclooctyl.
  • "-(C 8 -C 1 )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 14 )bicycloalkyls include -indanyl, -1,2,3,4-tetrahydronaphthyl, -5,6,7,8- tetrahydronaphthyl, -perhydronaphthyl and the like.
  • "-(C 8 -C 1 )tricycloalkyl” means a tri-cyclic hydrocarbon ring system having from 8 to 14 carbon atoms and at least one saturated cyclic alkyl ring.
  • Representative -(C 8 - C 14 )tricycloalkyls include -pyrenyl, -1,2,3,4-tetrahydroanthracenyl,
  • -(C 5 -C 1 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 5 -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 14 )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 -(Cg-C 1 )bicycloalkenyls include -indenyl, -pentalenyl, -naphthalenyl, -azulenyl, -heptalenyl, -1,2,7,8-tetrahydronaphthalenyl and the like.
  • "-(C 8 -C 1 )tricycloalkeny ⁇ ” 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 ⁇ )tricycloalkenyls include -anthracenyl, -phenanthrenyl, -phenalenyl, -acenaphthalenyl, - ⁇ s-indacenyl, -s-indacenyl and the like.
  • "-(5- to 10-membered)heteroary_” 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 of the rings is replaced with a heteroatom independently selected from nitrogen, oxygen, and sulfur.
  • one of the -(5- to 10- membered)heteroaryl's rings contain at least one carbon atom.
  • both of the -(5- to 10-membered)heteroaryl's rings contain at least one carbon atom.
  • Representative (5- to 10-membered)heteroaryls include pyridyl, furyl, benzofuranyl, benzo(l,3)dioxolyl, thiophenyl, benzothiophenyl, quinolinyl, pyrrolyl, indolyl, oxazolyl, benzoxazolyl, imidazolyl, benzimidazolyl, thiazolyl, benzothiazolyl, isoxazolyl, pyrazolyl, isothiazolyl, pyridazinyl, pyrimidyl, pyrazinyl, triazinyl, cinnolinyl, phthalazinyl, quinazolinyl and the like.
  • “-(3- to 7-rnembered)heterocycle” or “-(3- to 7-membered)heterocyclo” means a 3- to 7-membered monocyclic heterocyclic ring which is either saturated, unsaturated, non-aromatic or aromatic.
  • a 3- or a 4-membered heterocycle can contain up to 3 heteroatoms
  • a 5-membered heterocycle can contain up to 4 heteroatoms
  • a 6-membered heterocycle can contain up to 6 heteroatoms
  • a 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 heterocycle can contain up to 3 heteroatoms and a 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, -benzo(l,3)dioxolyl and the like.
  • a benzo(l,3)dioxolyl group has the structure:
  • -(C 1 )aryl means a 14-membered aromatic carbocyclic moiety such as anthryl or phenanthryl.
  • -CH (halo) means a methyl group where one of the hydrogens of the 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 where two of the hydrogens of the methyl group have been replaced with a halogen.
  • Representative -CH(halo) 2 groups include -CHF 2 , -CHC1 2 , -CHBr 2 , -CHBrCl, -CHC1I and -CHI 2 .
  • "-C(halo) 3 " means a methyl group where each of the hydrogens of the 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.
  • phrases "pharmaceutically acceptable salt,” as used herein, is any pharmaceutically acceptable salt that can be prepared from a Piperazine Compound, including a salt formed from an acid and a basic functional group, such as a nitrogen, of one of the 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, p-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
  • an "animal” is defined herein to include any animal (e.g., human, cow, horse, sheep, pig, chicken, turkey, quail, cat, dog, mouse, rat, rabbit or guinea pig), in one embodiment a mammal such as a non-primate and a primate (e.g. , monkey or human), and in another embodiment a human.
  • a mammal such as a non-primate and a primate (e.g. , monkey or human)
  • the human is an infant, child, adolescent or adult.
  • the phrase "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.
  • Compound and at last one therapeutic agent affects the treatment or prevention of a Condition, the same Condition in one embodiment, different Conditions in another embodiment.
  • the term "adjunctively administered” refers to the administration of at last one therapeutic agent in addition to a Piperazine Compound, either simultaneously with the same or at intervals prior to, during, or following administration of the a Piperazine Compound to achieve the desired therapeutic or prophylactic effect.
  • a first group is "substituted with one or more" second groups, each of one or more of the 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.
  • a first group is substituted with only one second group.
  • Ul means urinary incontinence.
  • ALS means amyotrophic lateral sclerosis.
  • treatment of includes the amelioration or cessation a Condition or a symptom thereof.
  • treating includes inhibiting, for example, decreasing the overall frequency of episodes of a Condition or a symptom thereof.
  • prevention of 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.
  • the illustrative methods disclose one or more synthetic intermediates that are useful for making the Piperazine Compounds. All of the synthetic intermediates are also encompassed by the present invention.
  • Certain Piperazine Compounds can be prepared according to Scheme 1.
  • the reaction mixture is concentrated, e.g., on rotary evaporator, and the residue is purified, e.g., via preparative thin-layer chromatography ("TLC"), to yield a Piperazine Compound.
  • TLC preparative thin-layer chromatography
  • Piperazine Compounds can be obtained by reacting a diaminoalkyl compound of formula D with a (C 1 -C 1 o)alkyliodide, or a (C 2 -C ⁇ o)alkenyliodide or (C 2 - C 1 o)alkynyliodide in which the iodine atom is bonded to an sp 3 carbon atom, at low temperature, e.g., about 0°C to about -78°C, in the presence of a strong base, e.g., lithium di-.so-propylamide (“LDA”), optionally in hexamethylphosphoramide (“HMPA”) as shown below in Scheme 2, e.g., for a (C 1 -C 1 o)alkyliodide reactant.
  • a strong base e.g., lithium di-.so-propylamide (“LDA)
  • HMPA hexamethylphosphoramide
  • Piperazine Compounds where R 6 contains an sp or sp carbon atom bonded to the acetylene group can be obtained by reacting a compound of formula A with an aryl iodide, or with a (C 2 -C 1 o)alkenyliodide or (C 2 -C 1 o)alkynyliodide in which the iodine atom is bonded to an sp or sp 2 carbon atom, at room temperature, e.g., about 25°C, in ethyl acetate (“EtOAc”) in the presence of Pd(Ph 3 P) 2 (OAc) 2 , Cul and TEA, as shown below in Scheme 3, e.g., for an aryl iodide reagent containing an R ⁇ substituent.
  • EtOAc ethyl acetate
  • a compound of formula D' i.e., a compound of formula D where A is -C(O)-
  • a compound of formula G can be prepared by reacting a compound of formula G with propynoic acid in the presence of 1-hydroxybenzotriazolehydrate (“HOBT”) and 1,3-di-iso-propylcarbodiimide (“DIC”) as shown in Scheme 4.
  • HOBT 1-hydroxybenzotriazolehydrate
  • DIC 1,3-di-iso-propylcarbodiimide
  • R 3 , R 4 , R 5 , R 7 , R 8 , X, Y, Z, m and n are as defined above for the Piperazine Compounds of formula (I).
  • a general procedure for reacting a carboxylic acid with an amine is provided in F.M. Martin et al, Bioorg. Med. Chem. Lett. 2887-2892 (1999).
  • a compound of formula D' can also be prepared by reacting a compound of formula G with propynoic acid chloride in the presence of a tertiary amine, such as TEA, as shown in Scheme 5.
  • a protected, e.g., Boc-protected, compound of formula G can be prepared by reacting a 2-halo-substituted piperadine-containing compound of formula A with a protected, e.g., Boc-protected, compound of formula I in the presence of TEA, e.g., in chloroform at a temperature of about 50°C, as shown in Scheme 6.
  • R ls R 3 , R 4 , R 5 , R , R 8 , R 9 and n are as defined above for the Piperazine Compounds of formula (I) and L is H or a protecting group. Methods for using a protecting group are described below. Selective reduction of an anhydride with sodium borohydride can be performed to yield a compound of formula AC where n is 0 and/or AD where n is 0 directly by methods known in the art, e.g., by adapting the procedures provided in Can. J. Chem., 64(6): 1031-1035 (1986) or J. Med. Chem., 34(9):2726-2735 (1991).
  • selective partial reduction of an anhydride with an aluminum hydride or sodium borohydride can be performed by methods known in the art, e.g., by adapting the procedures provided in Australian J. Chem., 42(6):787-795 (1989) or Tetrahedron, 44(10):2903-2912 (1988).
  • Selective partial reduction is followed by Grignard addition, e.g., with Ri-MgCl, to yield a compound of formula AC and/or AD.
  • Grignard addition can be performed by methods known in the art, e.g., by adapting the procedures provided in Hecheng Huaxue, l(3):255-257 (1993) or Tetrahedron, 54:7033-7044 (1998).
  • a protected, e.g., Boc-protected, compound of formula B e.g., a compound of formula J or J' where A is -C(O)-or -C(C 1 -C alkyl)(C 1 -C 4 alkyl)-, respectively, can be prepared by reacting a protected, e.g., Boc-protected, compound of formula I' with R 6 -substituted propynoic acid or R 6 -substituted 3-chloro-3-dialkyl-prop-l-yne as shown in Scheme 8.
  • R 3 , R , R 6 , R 13 , R 14 and R 15 are as defined above for the Piperazine Compounds of formula (I).
  • a general procedure for reacting a carboxylic acid with an amine is provided in F.M. Martin et al, Bioorg. Med. Chem. Lett. 2887-2892 (1999) and a general procedure for reacting a halogenated propyne with an amine is provided in International Publication No. WO 03/004480. Methods for using a protecting group are described below.
  • Certain bicyclic compounds of formula A' i.e., compounds of formula A where n is 0 or 1, can be prepared as shown in Scheme 9 from a compound of formula BB, which is, in turn, prepared from a compound of formula BA, by performing a thermally assisted intramolecular Diels-Alder reaction.
  • R 1? R 2 , R 5 , R 7 , R 8 , R 9 , X, Y, Z and m are as defined above for the Piperazine Compounds of formula (I), n' is 0 or 1, Q is I, Br, Cl or F, and W is -(C 1 -C 1 o)alkyl, -(C 2 - C 10 )alkenyl, -(C 2 -C 10 )alkynyl, -(C 3 -C 8 )cycloalkyl, -(C 8 -C 1 )bicycloalkyl, -(C 8 - C 14 )tricycloalkyl, -(C5-C 1 o)cycloalkenyl, -(C 8 -C 14 )bicycloalkenyl, -(C 8 - C 14 )tricycloalkenyl, -(3- to 7-membered)heterocycle, -(7- to 10- member
  • M in nitrobenzene can be irradiated with microwave radiation to form a compound of formula A'.
  • a compound of formula BB is irradiated under conditions that are sufficient to make a compound of formula A'.
  • the microwave irradiation frequency is from about 1 GHz to about 10 GHz. In another embodiment, the microwave irradiation frequency is from about 2 GHz to about 10 GHz. hi another embodiment, the microwave irradiation frequency is about 2.45 GHz. In one embodiment, a microwave-emitting apparatus operating at a frequency of about 2.45
  • the microwave irradiation causes the solution, which can be contained in a non-metallic container, to heat from a temperature of about 25°C to a reaction temperature of from about 250°C to about 300°C.
  • the reaction temperature is from about 260°C to about 290°C.
  • the reaction temperature is from about 265°C to about 280°C.
  • the heating occurs over a period of about 2 minutes or less. In another embodiment, the heating occurs over a period of about 1.5 minutes or less.
  • the heating occurs over a period of about 1 minute or less.
  • the solution can be held at the reaction temperature with continued microwave irradiation for, in one embodiment, from about 0.5 to about 60 minutes, in another embodiment from about 0.5 to about 3 minutes, in another embodiment from about 1 to about 2 minutes, in another embodiment from about 30 to about 60 minutes, and in another embodiment from about 30 to about 50 minutes. Thereafter, irradiation can be stopped and the solution can be allowed to cool to about 25°C.
  • Appropriate reaction conditions can be established by monitoring the formation of the compound of formula A' through routine experimentation, e.g., using TLC.
  • a compound of formula A where n is 2 can be prepared from a compound of formula A', where n' is 0 or 1, by methods known in the art.
  • Representative compounds of formula A and/or A' e.g., compounds of formula M, Q, T, V and Y', can be prepared by the following Schemes 10-14.
  • Ri, R2, R 5 , R 7 , R 8 , R 9 , R_o, m and n are as defined above for the Piperazine Compounds of formula (I); n' is 0 or 1; each R' is independently
  • A, R 3 through R 8 and R ⁇ through R 15 are as defined above for the Piperazine Compounds of formula (I); n' is 0 or 1; and W is, independently, as defined above for the compound of formula BB.
  • the reaction of 1 equivalent of U' with 1 equivalent of B can be conducted at a temperature of about 0°C.
  • the reaction of 1 equivalent of U' with 2 equivalents of B can be conducted at a temperature of about 25°C. If any group of the compounds depicted in Schemes 1-15 above contain a -hydroxyl or -amino group, that -hydroxyl and/or -amino group can be protected using a suitable protecting group, using methods known to those skilled in the art, before the compound is reacted.
  • Suitable protecting groups for a hydroxyl group include, but are not limited to, methyl ether, methoxymethyl ether, methoxythiomethyl ether, 2-methoxyethoxymethyl ether, bis(2-chloroethoxy)ethyl ether, tetrahydropyranyl ether, tetrahydrothiopyranyl ether, 4-methoxytetrahydropyranyl ether, methoxytetrahydrothiopyranyl ether, tetrahydrofuranyl ether, tetrahydrothiofuranyl ether, 1-ethoxyethyl ether, 1 -methyl- 1-methoxyethyl ether, 2-(phenylselenyl ether), tert-butyl ether, allyl ether, benzyl ether, o-nitrobenzyl ether, triphenylmethyl ether, o-napthyldiphenylmethyl ether, /7-methoxy
  • Suitable protecting groups for an amino group include, but are not limited to, l,l-dimethyl-2,2,2-trichloroethyl carbamate, 1 -methyl- l-(4-biphenylyl)ethyl carbamate, 2-trimethylsilylethyl carbamate, 9-fluorenylmethyl carbamate, and tert-butyl carbamate (T.W.
  • 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.
  • Optical isomers of the Piperazine Compounds can be obtained by known techniques such as chiral chromatography or formation of diastereomeric salts from an optically active acid or base.
  • the Piperazine Compound can be replaced by an isotope of the 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. Examples of 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, Ul, 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, labor pain, pain from physical trauma, myocardial infarction pain, pancreatic pain, colic pain, post-operative pain, headache pain, muscle pain, arthritic pain, neuropathic pain, and pain associated with a periodontal disease, including gingivitis and periodontitis.
  • the Piperazine Compounds can also be used for treating or preventing pain associated with inflammation or with an inflammatory disease in an animal. Such pain can arise when there is an inflammation of the body tissue, and which can be a local inflammatory response and/or a systemic inflammation.
  • the Piperazine Compounds can be used to 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 31 :297-303 (1999)) including, but not limited to, transplantation of the heart, lung, liver, or kidney; chronic inflammatory diseases of the joints, including arthritis, rheumatoid arthritis, osteoarthritis and bone diseases associated with increased bone resorption; inflammatory lung diseases, such as asthma, adult respiratory distress syndrome, and chronic obstructive airway disease; inflammatory diseases of the eye, including corneal dystrophy, trachoma, onchocerciasis, uveitis, sympathetic ophthalmitis and endophthalmitis; chronic inflammatory diseases of the gum, including gingivitis and periodontitis; tuberculosis; leprosy; inflammatory diseases of the kidney, including uremic complications, glomerulonephritis and
  • the Piperazine Compounds can also be used for treating or preventing pain associated with inflammatory disease that can, for example, be a systemic inflammation of the 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 of the 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 administered as a treatment for cancer.
  • the Piperazine Compounds can be used to treat or prevent UL Examples of Ul treatable or preventable using the Piperazine Compounds include, but are not limited to, urge incontinence, stress incontinence, overflow incontinence, neurogenic incontinence, and total incontinence.
  • Ul 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.
  • 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 Explosive Disorder, Kleptomania, Pyromania, Pathological Gambling, Trichotillomania, and Impulse Control Disorder not otherwise specified (NOS).
  • Alcohol-related disorders include, but are not limited to, Alcohol-Induced
  • 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
  • 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
  • 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 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, Cocaine-Induced Psychotic Disorders with hallucinations, Cocaine-Induced Mood Disorder, Cocaine- Induced Anxiety Disorder, Cocaine-Induced Sexual Dysfunction, Cocaine-Induced Sleep Disorder, and Cocaine 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, Cocaine-Induced Psychotic Disorders with hallucinations, Cocaine-Induced Mood Disorder, Cocaine- Induced Anxiety Disorder, Cocaine-Induced Sexual Dysfunction, Cocaine-Induced Sleep Disorder, and Cocaine Related Disorder not otherwise specified
  • Hallucinogen-related disorders include, but are not limited to, Hallucinogen Dependence, Hallucinogen Abuse, Hallucinogen Intoxication, Hallucinogen Withdrawal, Hallucinogen Intoxication Delirium, Hallucinogen-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 Disorder, Hallucinogen Persisting Perception Disorder (Flashbacks), and Hallucinogen Related Disorder not otherwise specified (NOS).
  • Inhalant-related disorders include, but are not limited to, Inhalant Dependence, Hallucinogen Abuse, Hallucinogen Intoxication, Hallucinogen Withdrawal, Hallucinogen Intoxication Delirium, Hallucinogen-Induced Psychotic Disorder with delusions, Hallucinogen-Induced Psychotic Disorder
  • Opioid-related disorders include, but are not limited to, Opioid Dependence,
  • 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, 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, and fiberglass dermatitis.
  • a pruritic condition including but not limited to, pruritus caused by dry skin, scabies, dermatitis, herpetiformis, atopic dermatitis, pruritus vulvae et ani, malaria, insect bites, pediculosis, contact dermatitis, drug reactions, urticaria, urticarial eruptions of pregnancy
  • 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.
  • 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 (e. g. , inhibit) 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.
  • the Piperazine Compounds can be used to treat or prevent depression, including but not limited to, major depression and bipolar disorder.
  • the Piperazine Compounds are antagonists for mGluR5.
  • the invention 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.
  • 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, psychosis or schizophrenia.
  • the method is also useful for inhibiting mGluR5 function in a cell in vivo, in an animal, in a human in one embodiment, by contacting a cell, in an animal, with an effective amount of a Piperazine Compound.
  • 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, another embodiment, the method is useful for treating or preventing parkinsonism in an animal in need thereof. In another embodiment, 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 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 schizophrenia in an animal in need thereof. Examples of cells capable of expressing mGluR5 are neuronal and glial cells of the central nervous system, particularly the brain, especially in the nucleus accumbens.
  • the invention also relates to methods for inhibiting mGluRl function in a cell comprising contacting a cell capable of expressing mGluRl with an effective amount of a Piperazine Compound.
  • 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 a Condition.
  • 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 effective amount of a Piperazine Compound.
  • the method is useful for treating or preventing pain in an animal in need thereof.
  • the method is useful for treating or preventing Ul 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, hi another embodiment, the method is useful for treating or preventing anxiety in an animal in need thereof.
  • the method is useful for treating or preventing epilepsy in an animal in need thereof.
  • the method is useful for treating or preventing a seizure in an animal in need thereof.
  • the method is useful for treating or preventing stroke 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.
  • the method is useful for treating or preventing a cognitive disorder in an animal in need thereof.
  • 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. In another embodiment, the method is useful for treating or preventing ALS in an animal in need thereof. In another embodiment, the method is useful for treating or preventing dementia in an animal in need thereof. In another embodiment, the method is useful for treating or preventing retinopathy in an animal in need thereof. In another embodiment, the method is useful for treating or preventing a muscle spasm in an animal in need thereof. In another embodiment, the method is useful for treating or preventing a migraine in an animal in need thereof. In another embodiment, the method is useful for treating or preventing vomiting in an animal in need thereof, i another embodiment, 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.
  • cells capable of expressing mGluRl include, but are not limited to, cerebellar Purkinje neuron cells, Purkinje cell bodies (punctate), cells of spine(s) of the cerebellum; neurons and neurophil cells of olfactory-bulb glomeruli; cells of the superficial layer of the cerebral cortex; hippocampus cells; thalamus cells; superior colliculus cells; and spinal trigeminal nucleus cells.
  • cerebellar Purkinje neuron cells Purkinje cell bodies (punctate), cells of spine(s) of the cerebellum; neurons and neurophil cells of olfactory-bulb glomeruli; cells of the superficial layer of the cerebral cortex; hippocampus cells; thalamus cells; superior colliculus cells; and spinal trigeminal nucleus cells.
  • Methods for assaying cells that express mGluRl are known in the art.
  • the Piperazine Compounds are advantageously useful in veterinary and human medicine. As described above, the Piperazine Compounds are useful for treating or preventing a Condition in an animal in need thereof. When administered to an animal, the Piperazine Compounds can be 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 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, or intestinal mucosa) and can be adjunctively administered with another therapeutical agent. Administration can be systemic or local.
  • Various delivery systems are known, e.g., encapsulation in liposomes, microparticles, microcapsules or capsules, and can be used to administer the Piperazine
  • Methods of administration include, but are not limited to, intradermal, 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 can be at the discretion of the practitioner. In specific embodiments, it can be desirable to administer the Piperazine
  • 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.
  • the Piperazine 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 Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)). Other controlled- or sustained-release systems discussed in the review by Langer, Science 249:1527-1533 (1990) can be used.
  • a pump can be used (Langer, Science
  • polymeric materials can be used (See Medical Applications of Controlled Release (Langer and Wise eds., 1974); Controlled Drug Bioavailability, Drug Product Design and Performance (Smolen and Ball eds., 1984); Ranger and Peppas, J. Macromol Sci. Rev. Macromol Chem. 23:61 (1983); Levy et al, Science 228:190 (1985); During et al., Ann.
  • a controlled- or sustained-release system can be placed in proximity of a target of the Piperazine Compounds, e.g., the spinal column, brain, or gastrointestinal tract, thus requiring only a fraction of the 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 excipient 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 can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • the present compositions can take the form of solutions, suspensions, emulsions, tablets, pills, pellets, capsules, capsules containing liquids, powders, sustained-release formulations, suppositories, 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), incorporated 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.
  • the compositions can be coated to delay disintegration and absorption 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.
  • 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. Typically, compositions for intravenous administration comprise sterile isotonic aqueous buffer. Where necessary, 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 of the injection.
  • the ingredients can be 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 ampoule or sachette indicating the quantity of active agent.
  • the 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.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients can be mixed prior to administration.
  • Piperazine Compounds can be administered by controlled-release or sustained-release means or by delivery devices that are known to those skilled 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 incorporated herein by reference.
  • Such dosage forms can be used to provide controlled- or sustained-release of one or more active ingredients using, for example, hydroxypropylmethyl 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 in the art, including those described herein, can be readily selected for use with the active ingredients of the 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-release counterparts.
  • a controlled- or sustained- release composition comprises a minimal amount of a Piperazine Compound to treat or prevent the condition in a minimal amount of time.
  • Advantages of controlled- or sustained-release compositions include extended activity of the 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 of the 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 of the 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 of the 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 of the Condition and can be decided according to the judgment of a practitioner and/or each animal's circumstances. Suitable effective dosage amounts, however, range from about 0.01 mg/kg of body weight to about 2500 mg/kg of body weight, although they are typically about 100 mg/kg of body weight or less.
  • the effective dosage amount ranges from about 0.01 mg/kg of body weight to about 100 mg/kg of body weight of a Piperazine Compound, in another embodiment, from about 0.02 mg/kg of body weight to about 50 mg/kg of body weight, and in another embodiment, from about 0.025 mg/kg of body weight to about 20 mg/kg of body weight.
  • an effective dosage amount is administered about every 24 hours until the Condition is abated.
  • an effective dosage amount is administered about every 12 hours until the Condition is abated.
  • an effective dosage amount is administered about every 8 hours until the Condition is abated.
  • an effective dosage amount is administered about every 6 hours until the Condition is abated.
  • an effective dosage amount is administered about every 4 hours until the Condition is abated.
  • 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 mGluR5 or mGluRl 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 ⁇ g/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 comprising the Piperazine Compound is from about 0.01 ⁇ 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/kg of body weight to about 2500 mg/kg of body weight, although it typically ranges from about 100 mg/kg of body weight or less.
  • the effective dosage amount ranges from about 0.01 mg/kg of body weight to about 100 mg/kg of body weight of a Piperazine Compound, in another embodiment, from about 0.02 mg/kg of body weight to about 50 mg/kg of body weight and in another embodiment, from about 0.025 mg/kg of body weight to about 20 mg/kg of body weight.
  • an effective dosage amount is administered about every 24 h.
  • an effective dosage amount is administered about every 12 h. In another embodiment, an effective dosage amount is administered about every 8 h. In another embodiment, an effective dosage amount is administered about every 6 h. In another embodiment, an effective dosage amount is administered about every 4 h.
  • 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 in humans.
  • the present methods for treating or preventing a Condition in an animal in need thereof can further comprise adjunctively administering another therapeutic agent to the animal being administered a Piperazine Compound. In one embodiment, the other therapeutic agent is adjunctively 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.
  • Effective amounts of the other therapeutic agents are known to those skilled in the art. However, it is within the skilled artisan's purview to determine the other therapeutic agent's optimal effective- amount range. In one embodiment of the invention, where another therapeutic agent is adjunctively administered to an animal, the effective amount of the Piperazine Compound is less than its effective amount would be where the other therapeutic agent is not administered.
  • the effective amount of the other therapeutic agent is less than its effective amount would be where the Piperazine Compound is not administered, hi these cases, without being bound by theory, it is believed that the Piperazine Compound(s) and the other therapeutic agent(s) act synergistically to treat or prevent a Condition.
  • the other therapeutic agent can be, 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 Ca -channel blocker, an anticancer agent, an agent for treating or preventing one or more Conditions, and mixtures thereof.
  • Examples of useful opioid agonists include, but are not limited to, alfentanil, allylprodine, alphaprodine, anileridine, benzylmorphine, bezitramide, buprenorphine, butorphanol, clonitazene, codeine, desomorphine, dextromoramide, dezocine, diampromide, diamorphone, dihydrocodeine, dihydromorphine, dimenoxadol, dimepheptanol, dimethylthiambutene, dioxaphetyl butyrate, dipipanone, eptazocine, ethoheptazine, ethylmethylthiambutene, ethylmorphine, etonitazene fentanyl, heroin, hydrocodone, hydromorphone, hydroxypethidine, isomethadone, ketobemidone, levorphanol, levophenacylmorphan, lofentanil
  • the opioid agonist is selected from codeine, hydromorphone, hydrocodone, oxycodone, dihydrocodeine, dihydromorphine, morphine, tramadol, oxymorphone, 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, carprofen, oxaprozin, pramoprofen, muroprofen, trioxaprofen, suprofen, aminoprofen, tiaprofenic acid, fluprofen, bucloxic acid, indomethacin, sulindac, tolmetin, zomepirac, tiopinac, zidometacin, acemetacin, fentiazac, clidanac, oxpinac, mefenamic acid, meclofenamic acid, flufenamic acid, niflumic acid, tolfenamic
  • 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 (phenylbuty
  • useful antimigraine agents include, but are not limited to, alpiropride, bromocriptine, dihydroergotamine, dolasetron, ergocornine, ergocorninine, ergocryptine, ergonovine, ergot, ergotamine, flumedroxone acetate, fonazine, ketanserin, lisuride, lomerizine, methylergonovine, methysergide, metoprolol, naratriptan, oxetorone, pizotyline, propranolol, risperidone, rizatriptan, sumatriptan, timolol, trazodone, zolmitriptan, and mixtures thereof.
  • the other therapeutic agent can alternatively be an agent useful for reducing any potential side effect of a Piperazine Compounds.
  • the other therapeutic agent can be an antiemetic agent.
  • useful antiemetic agents include, but are not limited to, metoclopromide, domperidone, prochlorperazine, promethazine, chlorpromazine, 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, thiethylpera
  • ⁇ -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, 5-methyl-5-(3-phenanthryl)- hydantoin, 3-mefhyl-5-phenylhydantoin,
  • Examples of useful antidepressants include, but are not limited to, binedaline, caroxazone, citalopram, (S)-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
  • Ca -channel blockers include, but are not limited to, bepridil, clentiazem, diltiazem, fendiline, gallopamil, mibefradil, prenylamine, semotiadil, terodiline, verapamil, amlodipine, aranidipine, barnidipine, benidipine, cilnidipine, efonidipine, elgodipine, felodipine, isradipine, lacidipine, lercanidipine, manidipine, nicardipine, nifedipine, nilvadipine, nimodipine, nisoldipine, nitrendipine, cinnarizine, flunarizine, lidoflazine, lo erizine, 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, chlorambucil,
  • anti-cancer drugs include, but are not limited to, 20-epi-l,25 dihydroxyvitamin D3; 5-ethynyluracil; abiraterone; aclarubicin; acylfulvene; adecypenol; adozelesin; aldesleukin; ALL-TK antagonists; altretamine; ambamustine; amidox; amifostine; aminolevulinic acid; amrubicin; amsacrine; anagrelide; anastrozole; andrographolide; angiogenesis inhibitors; antagonist D; antagonist G; antarelix; anti- dorsalizing morphogenetic protein- 1; antiandrogen; antiestrogen; antineoplaston; antisense oligonucleotides; aphidicolin glycinate; apoptosis gene modulators; apoptosis regulators; apurinic acid; ara-CDP-DL-PTBA;
  • useful therapeutic agents for treating or preventing Ul include, but are not limited to, propantheline, imipramine, hyoscyamine, oxybutynin, and dicyclomine.
  • useful therapeutic agents for treating or preventing an addictive disorder include, but are not limited to, methadone, desipramine, amantadine, fluoxetine, buprenorphine, an opiate agonist, 3-phenoxypyridine, levomethadyl acetate hydrochloride, and serotonin antagonists.
  • Examples of useful therapeutic agents for treating or preventing 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 chlorpromazine 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 chlorpromazine hydrochloride, mesoridazine besylate, and thoridazine hydrochloride
  • thioxanthenes such as chloroprothixene and thiothixene hydrochloride
  • clozapine
  • Examples of useful therapeutic agents for treating or preventing Huntington's chorea include, but are not limited to, haloperidol and pimozide.
  • Examples of 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.
  • Examples of 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.
  • useful therapeutic agents for treating (e.g., inhibiting) 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 prochlorperazine, thiethylperazine, chlorpromazine, metoclopramide, and domperidone; glucocorticoids such as dexamethasone; and benzodiazepines such as lorazepam and alprazolam.
  • useful therapeutic agents for treating or preventing dyskinesia include, but are not limited to, reserpine 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, and venlafaxine; selective serotonin reuptake inhibitors such as fluoxetine, fluvoxamine, paroxetine, citalopram, (S)-citalopram 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, des
  • a Piperazine Compound and the other therapeutic agent can act additively or, in one embodiment, synergistically.
  • a Piperazine Compound is adjunctively administered with another therapeutic agent; for example, a composition comprising an effective amount of a Piperazine Compound, 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.
  • a composition of the 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 known for admixing a compound (or salt) and a pharmaceutically acceptable carrier or excipient. In one embodiment the composition is prepared such that the Piperazine Compound is present in the composition in an effective amount.
  • Kits The invention encompasses kits that can simplify the administration of a Piperazine Compound to an animal.
  • a typical kit of the 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 of the Piperazine Compound to treat a Condition.
  • the kit can also further comprise a unit dosage form of another therapeutic agent, for example, a second container containing an effective amount of the other therapeutic agent and a pharmaceutically acceptable carrier or excipient.
  • the kit comprises a container containing an effective amount of a Piperazine Compound, an effective amount of another therapeutic agent and a pharmaceutically acceptable carrier or excipient.
  • Kits of the invention can further comprise a device that is useful for administering the unit dosage forms.
  • a device include but are not limited to a syringe, a drip bag, a patch, an inhaler, and an enema bag.
  • the following examples are set forth to assist in understanding the invention and should not be construed as specifically limiting the invention described and claimed herein.
  • the examples disclose one or more synthetic intermediates that are useful for making the Piperazine Compounds. All of the synthetic intermediates are also encompassed by the present invention.
  • Such variations of the invention including the substitution of all equivalents now known or later developed, which would be within the purview of those skilled in the art, and changes in formulation or changes in experimental design, are to be considered to fall within the scope of the invention incorporated herein.
  • Examples 1-12 relate to the synthesis of illustrative Piperazine Compounds.
  • Compound 8 was prepared from Compound 6 using a procedure that was analogous to that used to prepare Compound 7 except that 3-butyn-l-ol (Aldrich Chemical) was used in place of propargyl alcohol.
  • Compound 15 was prepared by irradiating Compound 8 with microwave irradiation using a procedure that was analogous to that used to prepare Compound 14 except that, after the 1 minute warming period, the duration of microwave irradiation was about 45 minutes at about 265°C (31% yield). The structure of Compound 15 was confirmed by 1H NMR and mass spectrometry.
  • Compound 1441(a) was prepared from Compound 23 using a procedure that was analogous to that used in Example 1 to prepare Compound 1009(a) except that Compound 15 was used in place of Compound 14 (48% yield). The structure of Compound 1441(a) was confirmed by 1H NMR and mass spectrometry.
  • Compound 1011(a) was prepared from Compound 23 using a procedure that was analogous to that used in Example 1 to prepare Compound 1009(a) except that Compound 3 was used in place of Compound 14 (37% yield). The structure of Compound 1011(a) was confirmed by 1H NMR and mass spectrometry.
  • Compound 48 was prepared from Compound 21 using a procedure that was analogous to that used to prepare Compound 22 except that 4-methoxy phenyl-propynoic acid (Compound 47) was used in place of phenyl-propynoic acid (55% yield). The structure of Compound 48 was confirmed by 1H NMR and mass spectrometry.
  • Compound 1009(b) was prepared from Compound 14 using a procedure that was analogous to that used in Example 1 to prepare Compound 1009(a) except that Compound 27 was used in place of Compound 23 (56% yield). The structure of Compound 1009(b) was confirmed by 1H NMR and mass spectrometry.
  • Compound 1009(c) was prepared from Compound 14 using a procedure that was analogous to that used in Example 1 to prepare Compound 1009(a) except that Compound 29 was used in place of Compound 23 (63% yield). The structure of Compound 1009(c) was confirmed by 1H NMR and mass spectrometry.
  • Compound 9 was prepared from Compound 6 using a procedure that was analogous to that used to prepare Compound 7 except that the reaction temperature was about -78°C and 2-propynyl amine (Aldrich Chemical) was used in place of propargyl alcohol. The reaction was complete within one hour, as evidenced by LCMS, and quenched at about -78°C by addition of saturated aqueous NH C1. The structure of Compound 9 was confirmed by 1H NMR and mass spectrometry.
  • Compound 865(a) was prepared by irradiating Compound 36 with microwave irradiation using a procedure that was analogous to that used to prepare Compound 14 except that the temperature was about 280°C at the end of the 1 minute warming period and the duration of microwave irradiation was about 1.5 minutes at about 280°C.
  • the resulting mixture was poured onto a prepacked silica gel column of the type described previously in Example 1. Nitrobenzene was removed using DCM eluent, and a mixture of Compounds 865(a) and 16 were eluted using flash chromatography on the silica gel column with gradient elution from 2.5% methanol/DCM to 5% methanol/DCM.
  • Compound 11 was prepared from Compound 6 using a procedure that was analogous to that used to prepare Compound 9 except that N-methyl-2-propynyl-amine (Aldrich Chemical) was used in place of 2-propynyl amine.
  • the structure of Compound 11 was confirmed by 1H NMR and mass spectrometry. Two conformers of Compound 11 were observed in the former.
  • Compound 37 was prepared from Compound 23 using a procedure that was analogous to that used to prepare Compound 36 except that Compound 11 was used in place of Compound 9.
  • the structure of Compound 37 was confirmed by 1H NMR and mass spectrometry. Two conformers of Compound 37 were observed in the former.
  • Compound 937(a) was prepared by irradiating Compound 37 with microwave irradiation using a procedure that was analogous to that used to prepare Compound 14 (about 8.7% and 64% yield of Compounds 937(a) and 17, respectively). The resulting mixture was poured onto a prepacked silica gel column of the type described previously in Example 1. Nitrobenzene was removed using DCM eluent, and a mixture of
  • Compounds 17 and 18 were each prepared (isolated in 69% and 24% yields, respectively) by irradiating Compound 12 with microwave irradiation using a procedure that was analogous to that used to prepare Compound 14. The structures of Compounds 17 and 18 were confirmed by 1H NMR and mass spectrometry.
  • Compound 17: 1H NMR(CDC1 3 ): ⁇ (ppm): 8.51 (d, 7 4.8Hz, IH); 7.37(dt, 7
  • Compound 1753(a) was prepared from Compound 23 using a procedure that was analogous to that used in Example 1 to prepare Compound 1009(a) except that Compound 18 was used in place of Compound 14 (37% yield).
  • the structure of Compound 1753(a) was confirmed by 1H NMR and mass spectrometry.
  • MS m/z 375.2 (MH).
  • Compound 1753(c) was prepared using a procedure that was analogous to that used in Example 1 to prepare Compound 1009(a) except that Compounds 18 and 29 were used in place of Compounds 14 and 23, respectively (94% yield). The structure of Compound 1753(c) was confirmed by 1H NMR and mass spectrometry.
  • Compound 1753(b) was prepared using a procedure that was analogous to that used in Example 1 to prepare Compound 1009(a) except that Compounds 18 and 27 were used in place of Compounds 14 and 23, respectively (72% yield). The structure of Compound 1753(b) was confirmed by 1H NMR and mass spectrometry.
  • Compound 13 was prepared from Compound 6 using a procedure that was analogous to that used to prepare Compound 9 except that N,N-dimethyl-prop-2- ynylamine (Aldrich Chemical) was used in place of 2-propynyl amine.
  • the structure of Compound 13 was confirmed by 1H NMR and mass spectrometry.
  • Compound 13 1H NMR(CDC1 3 ): ⁇ (ppm): 8.80(s, IH); 6.04(bs, IH); 2.45(s, IH); 1.80(s, 6H). MS; 280(M+Na).
  • Compound 38 was prepared from Compound 23 using a procedure that was analogous to that used to prepare Compound 36 except that Compound 13 was used in place of Compound 9. The structure of Compound 38 was confirmed by 1H NMR and mass spectrometry. Two conformers of Compound 38 were observed in the former.
  • Compound 38 1H NMR(CDC1 3 ): ⁇ (ppm): 8.80(s, 0.4H); 8.38(s, IH); 7.54(m, 3.1H); 7.41(m, 5.6H); 3.93(m, 3.1H); 3.90(m, 3.1H); 3.79(s,5.6H); 2.45(s, 0.4H); 2.43(s, IH); 1.81(s, 1.4H); 1.76(s, 6H).
  • Compound 867(a) was prepared by irradiating Compound 38 with microwave irradiation using a procedure that was analogous to that used to prepare Compound 14. The resulting mixture was poured onto a prepacked silica gel column of the type described previously in Example 1. Nitrobenzene was washed off with DCM eluent and a mixture of Compounds 867(a) and 19 was purified using flash chromatography on the silica gel column with gradient elution from 2.5% methanol DCM to 5% methanol/DCM. Purification by preparative TLC (2.5% methanol DCM) provided Compound 867(a) (4% yield from Compound 13).
  • Compound 867(a) was confirmed by 1H NMR and mass spectrometry.
  • Compound 867(a): 1H NMR(CDC1 3 ): ⁇ (ppm): 8.32(d, 7 5.0 Hz, IH); 7.56(m,
  • Example 13 Binding of an Illustrative Piperazine Compound to rnGluRS The following assay demonstrates that Compound 1441(a), an illustrative
  • Piperazine Compound binds to mGluR5.
  • Cell Cultures Primary 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.
  • DMEM Dulbecco's Modified Eagle's Medium
  • 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 of the 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 KC1, 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).
  • CHO-rat mGluR5 cells/well are plated into 96 well plate (Costar 3409, Black, clear bottom, 96 well, tissue culture treated) for an about 16 hour incubation in Dulbecco's Modified Eagle's Medium (DMEM, pH 7.4) and supplemented with glutamine, 10% FBS, 1% Pen Strep, and 500 ⁇ g/mL Geneticin.
  • DMEM Dulbecco's Modified Eagle's Medium
  • DMEM Dulbecco's Modified Eagle's Medium
  • Pen Strep 500 ⁇ g/mL Geneticin.
  • 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 KC1, 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 KC1, 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 commercially
  • the plates containing the CHO-rat mGluR5 cells are then transferred to a FLIPR for the assessment of calcium mobilization flux in the presence of glutamate and in the presence or absence of test compounds.
  • DMSO solutions containing various concentrations of the 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 ⁇ M. Plate fluorescence is then monitored for an additional 60 seconds after agonist addition.
  • the final DMSO concentration in the assay is 1.0%.
  • Example 15 In Vivo Assays for Treatment or Prevention of Pain The following assays can be used to demonstrate that Piperazine Compounds are useful for treating or preventing pain.
  • Test Animals Each experiment uses rats weighing between 200-260 g at the start of the 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 of the thermal stimulus and the flick of the 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 of the maximal possible effect (% MPE), i.e., 20 seconds, is calculated as follows:
  • % MPE X 100 (20 s pre-administration latency)
  • FCA Freund's complete adjuvant
  • FCA-induced inflammation of the rat hind paw is associated with the development of persistent inflammatory mechanical hyperalgesia and provides reliable prediction of the anti-hyperalgesic action of clinically useful analgesic drugs (L. Bartho et al, "Involvement of Capsaicin-sensitive Neurones in Hyperalgesia and Enhanced Opioid Antinociception in Inflammation," Naunyn-Schmiedeberg's Archives of Pharmacol. 342:666-670 (1990)).
  • 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 of a control selected from Celebrex, indomethacin or naproxen; 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 Seltzer model To assess the actions of the Piperazine Compounds for the treatment or prevention of neuropathic pain either the Seltzer model or the Chung model can be used.
  • the Seltzer model 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 of the left sciatic nerve is performed under isoflurane/O 2 inhalation anaesthesia.
  • the left thigh of the 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 l ⁇ of the 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. Sham-treated rats undergo an identical surgical procedure except that the sciatic nerve is not manipulated.
  • 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, prior to surgery (baseline), then immediately prior to and 1, 3 and 5 hours after being administered a Piperazine Compound for the left rear paw of the animal.
  • 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," P ⁇ 50(3):355-363 (1992).
  • Response to Mechanical Stimuli as an Assessment of Mechanical Hyperalgesia 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.
  • 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 of the 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 of the paw are considered nociceptive behavioral responses.
  • Example 16 In Vivo Assays for Treatment or Prevention of Anxiety
  • 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 of the platform, at the crossroad of the 4 arms, facing one of the closed arms. Time spent in the open arms vs the closed arms and number of open arm entries during the testing period are recorded.
  • 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 40 x 30 x 40 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 niA. 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 corner of the test chamber following drug administration.
  • 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 of the 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.
  • an index of the rat's reactivity to each shock is scored on a 4 point scale. The total time spent immobile during the 15 min testing period is used as an index of general activity. The shock-probe burying test is described in D. Treit, 1985, supra.
  • Example 17 In Vivo Assays for Treatment or Prevention of an Addictive Disorder
  • the conditioned 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 Conditioned 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).
  • 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 During habituation, rats generally show no preference for either compartment. Following habituation, 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 + morphine.
  • carrier pre-treatment + carrier control group
  • Piperazine Compound pre-treatment + carrier carrier pre-treatment + morphine
  • Piperazine Compound pre-treatment + morphine During each conditioning session the rat is injected with one of the drug combinations and confined to one compartment for 30 min. On the following day, the rat receives a carrier + carrier treatment and is confined to the other large compartment.
  • 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.
  • the apparatus for 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 morphine. Rats are then presented with two levers, an "active" lever and an "inactive" lever.
  • Pressing of the active lever results in drug infusion on a fixed ratio 1 (FR1) 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 of the inactive lever results in infusion of excipient. 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.
  • FR1 ratio 1
  • Piperazine Compound blocks or reduces 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).
  • Example 18 Functional Assay for Characterizing mGluRl Antagonistic Properties
  • Functional assays for the characterization of mGluRl antagonistic properties are known in the art. For example, the following procedure can be used.
  • a CHO-rat mGluRl cell line is generated using cDNA encoding rat mGluRl receptor (M. Masu and S. Nakanishi, Nature 349:760-765 (1991)).
  • the cDNA encoding rat mGluRla receptor can be obtained from, e.g., Prof. S. Nakanishi (Kyoto, Japan).
  • CHO-rat mGluRl cells/well are plated into a COSTAR 3409, black, clear bottom, 96 well, tissue culture treated plate (commercially available from Fisher Scientific of Chicago, IL) and are incubated in Dulbecco's Modified Eagle's Medium (DMEM, pH 7.4) supplemented with glutamine, 10% FBS, 1% Pen/Strep, and 500 ⁇ g/mL Geneticin for about 12 hours.
  • DMEM Dulbecco's Modified Eagle's Medium
  • the CHO-rat mGluRl cells are then washed and treated with OPTIMEM medium (commercially available from Invitrogen, Carlsbad, CA) and incubated for a time period ranging from 1 to 4 hours prior to loading the cells with the dye FLUO-4.
  • OPTIMEM medium commercially available from Invitrogen, Carlsbad, CA
  • the cell plates are washed with loading buffer (127 mM NaCl, 5 mM KC1, 2 mM MgCl 2 , 700 ⁇ M, NaH 2 PO 4 , 2 mM CaCl 2 , 5 mMNaHCO 3 , 8 mM HEPES, and 10 mM glucose, pH 7.4) and incubated with 3 ⁇ M FLUO-4 in 0.1 mL loading buffer for 90 min.
  • the cells are then washed twice with 0.2 mL loading buffer, resuspended in 0.1 mL of loading buffer, and transferred to a FLIPR for measurement of calcium mobilization flux in the presence of glutamate and in the presence or absence of a Piperazine Compound.
  • DMSO solutions containing various concentrations of a Piperazine Compound ranging from about 50 ⁇ M to about 0.8 nM diluted in loading buffer (0.05 mL of a 4X dilution) are added to the cell plate and fluorescence is monitored for about 2 min.
  • 0.05 mL of a 4X glutamate solution (agonist) is then added to each well to provide a final glutamate concentration in each well of 10 ⁇ M and fluorescence is monitored for about one additional min.
  • the final DMSO concentration in the assay is 1%.

Abstract

L'invention concerne un composé représenté par la formule (I), (dans laquelle A, X, Y, Z, R3, R4, R5, R6, m et n sont définis dans le descriptif) ou un sel pharmaceutiquement acceptable de celui-ci (le composé ou le sel pharmaceutiquement acceptable étant un « composé de pipérazine ») ; des compositions pharmaceutiquement contenant une dose efficace d'un composé de pipérazine ; ainsi que des méthodes permettant de traiter la douleur ainsi que d'autres troubles, consistant à administrer à un animal nécessitant un tel traitement une dose efficace d'un composé de pipérazine.
PCT/US2004/041378 2003-12-09 2004-12-09 Agents therapeutiques pour traitement de la douleur WO2005056524A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US52858103P 2003-12-09 2003-12-09
US60/528,581 2003-12-09

Publications (2)

Publication Number Publication Date
WO2005056524A2 true WO2005056524A2 (fr) 2005-06-23
WO2005056524A3 WO2005056524A3 (fr) 2005-09-22

Family

ID=34676853

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/041378 WO2005056524A2 (fr) 2003-12-09 2004-12-09 Agents therapeutiques pour traitement de la douleur

Country Status (1)

Country Link
WO (1) WO2005056524A2 (fr)

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007079957A1 (fr) * 2005-12-28 2007-07-19 Grünenthal GmbH Amides de l'acide propiolique substitues et leur utilisation pour produire des medicaments
JP2010518104A (ja) * 2007-02-09 2010-05-27 アストラゼネカ・アクチエボラーグ アザ−イソインドロンおよび代謝型グルタミン酸レセプター増強剤−613としてのそれらの使用
WO2012074769A1 (fr) * 2010-12-02 2012-06-07 Eli Lilly And Company Composés 6-(pyridinylméthoxy)-pyrrolopyridines 3-substitués
US8440689B2 (en) 2009-12-23 2013-05-14 Takeda Pharmaceutical Company Limited Fused heteroaromatic pyrrolidinones
US9056873B2 (en) 2011-06-22 2015-06-16 Takeda Pharmaceutical Company Limited Substituted 6-aza-isoindolin-1-one derivatives
US9452990B2 (en) 2012-06-20 2016-09-27 Novartis Ag Complement pathway modulators and uses thereof
US9611267B2 (en) 2012-06-13 2017-04-04 Incyte Holdings Corporation Substituted tricyclic compounds as FGFR inhibitors
US9708318B2 (en) 2015-02-20 2017-07-18 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9745311B2 (en) 2012-08-10 2017-08-29 Incyte Corporation Substituted pyrrolo[2,3-b]pyrazines as FGFR inhibitors
US9801889B2 (en) 2015-02-20 2017-10-31 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9890156B2 (en) 2015-02-20 2018-02-13 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10040790B2 (en) 2013-04-19 2018-08-07 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US10213427B2 (en) 2010-12-22 2019-02-26 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US10253046B2 (en) 2015-02-27 2019-04-09 Nimbus Lakshmi TYK2 inhibitors and uses thereof
US10611762B2 (en) 2017-05-26 2020-04-07 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
US10851105B2 (en) 2014-10-22 2020-12-01 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
RU2738707C1 (ru) * 2020-06-02 2020-12-15 Федеральное государственное бюджетное образовательное учреждение высшего образования "Чувашский государственный университет имени И.Н. Ульянова" Способ получения производных фуро[3,4-с]пиридин-1(3Н)-онов
JP2021514362A (ja) * 2018-02-23 2021-06-10 ベイジン タイド ファーマシューティカル カンパニー リミテッドBeijing Tide Pharmaceutical Co., Ltd. 受容体阻害剤、同阻害剤を含む医薬組成物及びその使用
US11174257B2 (en) 2018-05-04 2021-11-16 Incyte Corporation Salts of an FGFR inhibitor
US11407750B2 (en) 2019-12-04 2022-08-09 Incyte Corporation Derivatives of an FGFR inhibitor
US11466004B2 (en) 2018-05-04 2022-10-11 Incyte Corporation Solid forms of an FGFR inhibitor and processes for preparing the same
US11607416B2 (en) 2019-10-14 2023-03-21 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11628162B2 (en) 2019-03-08 2023-04-18 Incyte Corporation Methods of treating cancer with an FGFR inhibitor
US11897891B2 (en) 2019-12-04 2024-02-13 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors
US11939331B2 (en) 2021-06-09 2024-03-26 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9266892B2 (en) 2012-12-19 2016-02-23 Incyte Holdings Corporation Fused pyrazoles as FGFR inhibitors
WO2021007269A1 (fr) 2019-07-09 2021-01-14 Incyte Corporation Hétérocycles bicycliques en tant qu'inhibiteurs de fgfr
WO2021076728A1 (fr) 2019-10-16 2021-04-22 Incyte Corporation Hétérocycles bicycliques en tant qu'inhibiteurs de fgfr

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5110927A (en) * 1987-12-31 1992-05-05 The United States Of America As Represented By The Department Of Health And Human Services Prazosin analog with increased selectivity and duration of action
EP0494604A1 (fr) * 1991-01-10 1992-07-15 Hoechst-Roussel Pharmaceuticals Incorporated 1-Pipérazinyl-2-butènes et 2-butynes, des intermédiaires, un procédé pour leur préparation et leur utilisation comme médicaments
EP0623616A1 (fr) * 1993-05-04 1994-11-09 Egis Gyogyszergyar Rt. Composés hétérocycliques et procédé pour leur préparation
EP0976753A1 (fr) * 1997-03-24 2000-02-02 Kyowa Hakko Kogyo Co., Ltd. DERIVES 1,2,4]TRIAZOLO 1,5-c]PYRIMIDINIQUES
WO2003022214A2 (fr) * 2001-09-06 2003-03-20 Millennium Pharmaceuticals, Inc. Composes a base de piperazine et d'homopiperazine

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5110927A (en) * 1987-12-31 1992-05-05 The United States Of America As Represented By The Department Of Health And Human Services Prazosin analog with increased selectivity and duration of action
EP0494604A1 (fr) * 1991-01-10 1992-07-15 Hoechst-Roussel Pharmaceuticals Incorporated 1-Pipérazinyl-2-butènes et 2-butynes, des intermédiaires, un procédé pour leur préparation et leur utilisation comme médicaments
EP0623616A1 (fr) * 1993-05-04 1994-11-09 Egis Gyogyszergyar Rt. Composés hétérocycliques et procédé pour leur préparation
EP0976753A1 (fr) * 1997-03-24 2000-02-02 Kyowa Hakko Kogyo Co., Ltd. DERIVES 1,2,4]TRIAZOLO 1,5-c]PYRIMIDINIQUES
WO2003022214A2 (fr) * 2001-09-06 2003-03-20 Millennium Pharmaceuticals, Inc. Composes a base de piperazine et d'homopiperazine

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
MARK H. NORMAN, DOUGLAS J. MINICK, GREG C. RIGDON: "Effect of Linking Bridge Modifications on the Antipsychotic Profile of Some Phthalimide and Isoindolinone Derivatives" J. MED. CHEM., vol. 39, 1996, pages 149-157, XP002330280 *

Cited By (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2009522219A (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 置換されたプロピオール酸アミド及び医薬の製造へのその使用
WO2007079957A1 (fr) * 2005-12-28 2007-07-19 Grünenthal GmbH Amides de l'acide propiolique substitues et leur utilisation pour produire des medicaments
JP2010518104A (ja) * 2007-02-09 2010-05-27 アストラゼネカ・アクチエボラーグ アザ−イソインドロンおよび代謝型グルタミン酸レセプター増強剤−613としてのそれらの使用
US9108970B2 (en) 2009-12-23 2015-08-18 Takeda Pharmaceutical Company Limited Fused heteroaromatic pyrrolidinones
US8440689B2 (en) 2009-12-23 2013-05-14 Takeda Pharmaceutical Company Limited Fused heteroaromatic pyrrolidinones
US9181255B2 (en) 2009-12-23 2015-11-10 Takeda Pharmaceutical Company Limited Fused heteroaromatic pyrrolidinones as SYK inhibitors
KR101547407B1 (ko) 2010-12-02 2015-08-25 일라이 릴리 앤드 캄파니 3-치환-6-(피리디닐메톡시)-피롤로피리딘 화합물
US8778946B2 (en) 2010-12-02 2014-07-15 Eli Lilly And Company 3-substituted-6-(pyridinylmethoxy)-pyrrolopyridine compounds
JP2013544836A (ja) * 2010-12-02 2013-12-19 イーライ リリー アンド カンパニー 3−置換−6−(ピリジニルメトキシ)−ピロロピリジン化合物
EA021781B1 (ru) * 2010-12-02 2015-08-31 Эли Лилли Энд Компани 3-замещенные 6-(пиридинилметокси)пирролопиридиновые соединения
CN103228656B (zh) * 2010-12-02 2015-09-30 伊莱利利公司 3-取代基-6-(吡啶基甲氧基)-吡咯并吡啶化合物
CN103228656A (zh) * 2010-12-02 2013-07-31 伊莱利利公司 3-取代基-6-(吡啶基甲氧基)-吡咯并吡啶化合物
WO2012074769A1 (fr) * 2010-12-02 2012-06-07 Eli Lilly And Company Composés 6-(pyridinylméthoxy)-pyrrolopyridines 3-substitués
US10213427B2 (en) 2010-12-22 2019-02-26 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US10813930B2 (en) 2010-12-22 2020-10-27 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US9663514B2 (en) 2011-06-22 2017-05-30 Takeda Pharmaceutical Company Limited Substituted 6-aza-isoindolin-1-one derivatives
US9056873B2 (en) 2011-06-22 2015-06-16 Takeda Pharmaceutical Company Limited Substituted 6-aza-isoindolin-1-one derivatives
US9611267B2 (en) 2012-06-13 2017-04-04 Incyte Holdings Corporation Substituted tricyclic compounds as FGFR inhibitors
US11840534B2 (en) 2012-06-13 2023-12-12 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
US10131667B2 (en) 2012-06-13 2018-11-20 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
US11053246B2 (en) 2012-06-13 2021-07-06 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
US9452990B2 (en) 2012-06-20 2016-09-27 Novartis Ag Complement pathway modulators and uses thereof
US9745311B2 (en) 2012-08-10 2017-08-29 Incyte Corporation Substituted pyrrolo[2,3-b]pyrazines as FGFR inhibitors
US10040790B2 (en) 2013-04-19 2018-08-07 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US10947230B2 (en) 2013-04-19 2021-03-16 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11530214B2 (en) 2013-04-19 2022-12-20 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US10450313B2 (en) 2013-04-19 2019-10-22 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US10851105B2 (en) 2014-10-22 2020-12-01 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10251892B2 (en) 2015-02-20 2019-04-09 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11014923B2 (en) 2015-02-20 2021-05-25 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10632126B2 (en) 2015-02-20 2020-04-28 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10738048B2 (en) 2015-02-20 2020-08-11 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11667635B2 (en) 2015-02-20 2023-06-06 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10214528B2 (en) 2015-02-20 2019-02-26 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9708318B2 (en) 2015-02-20 2017-07-18 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10016438B2 (en) 2015-02-20 2018-07-10 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11173162B2 (en) 2015-02-20 2021-11-16 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9801889B2 (en) 2015-02-20 2017-10-31 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9890156B2 (en) 2015-02-20 2018-02-13 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10968236B2 (en) 2015-02-27 2021-04-06 Nimbus Lakshmi, Inc. TYK2 inhibitors and uses thereof
US10253046B2 (en) 2015-02-27 2019-04-09 Nimbus Lakshmi TYK2 inhibitors and uses thereof
US10611762B2 (en) 2017-05-26 2020-04-07 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
US11472801B2 (en) 2017-05-26 2022-10-18 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
JP2021514362A (ja) * 2018-02-23 2021-06-10 ベイジン タイド ファーマシューティカル カンパニー リミテッドBeijing Tide Pharmaceutical Co., Ltd. 受容体阻害剤、同阻害剤を含む医薬組成物及びその使用
US11905274B2 (en) 2018-02-23 2024-02-20 Beijing Tide Pharmaceutical Co., Ltd. Receptor inhibitors, pharmaceutical compositions comprising the same and use thereof
US11174257B2 (en) 2018-05-04 2021-11-16 Incyte Corporation Salts of an FGFR inhibitor
US11466004B2 (en) 2018-05-04 2022-10-11 Incyte Corporation Solid forms of an FGFR inhibitor and processes for preparing the same
US11628162B2 (en) 2019-03-08 2023-04-18 Incyte Corporation Methods of treating cancer with an FGFR inhibitor
US11607416B2 (en) 2019-10-14 2023-03-21 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11407750B2 (en) 2019-12-04 2022-08-09 Incyte Corporation Derivatives of an FGFR inhibitor
US11897891B2 (en) 2019-12-04 2024-02-13 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors
RU2738707C1 (ru) * 2020-06-02 2020-12-15 Федеральное государственное бюджетное образовательное учреждение высшего образования "Чувашский государственный университет имени И.Н. Ульянова" Способ получения производных фуро[3,4-с]пиридин-1(3Н)-онов
US11939331B2 (en) 2021-06-09 2024-03-26 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors

Also Published As

Publication number Publication date
WO2005056524A3 (fr) 2005-09-22

Similar Documents

Publication Publication Date Title
KR100896497B1 (ko) 통증 치료에 유용한 치료제
US6864261B2 (en) Therapeutic agents useful for treating pain
KR100977242B1 (ko) 피페리딘 화합물 및 그들을 포함하는 약학적 조성물
WO2005056524A2 (fr) Agents therapeutiques pour traitement de la douleur
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
WO2004029031A2 (fr) Agents therapeutiques utiles dans le traitement de la douleur
EP1664016A2 (fr) Agents therapeutiques utiles pour traiter la douleur
EP1664041A1 (fr) Composes phenyl-carboxamide utiles pour traiter la douleur
WO2004002983A2 (fr) Agents therapeutiques utiles pour le traitement de la douleur
CA2534088C (fr) Composes pyridyle 2,3-substitues utiles dans le traitement de la douleur
WO2003093236A1 (fr) Composes 1-(pyrid-2-yl)-piperazine utilises en tant qu'inhibiteur du recepteur de glutamate metabotropique
KR100867188B1 (ko) 통증 치료에 유용한 피페라진
EP1542991A1 (fr) Composes de pyrimidine en tant qu'agents therapeutiques
WO2005007641A1 (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: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG 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 NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY 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: A2

Designated state(s): GM KE LS MW MZ NA 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 IS IT LT LU MC NL PL 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
122 Ep: pct application non-entry in european phase