US20090041800A1 - Methods for Treating Dependence - Google Patents

Methods for Treating Dependence Download PDF

Info

Publication number
US20090041800A1
US20090041800A1 US12/187,166 US18716608A US2009041800A1 US 20090041800 A1 US20090041800 A1 US 20090041800A1 US 18716608 A US18716608 A US 18716608A US 2009041800 A1 US2009041800 A1 US 2009041800A1
Authority
US
United States
Prior art keywords
cocaine
nepicastat
compound
dopamine
patient
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/187,166
Other languages
English (en)
Inventor
Tom Woiwode
Mark Moran
Lesley Pickford
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Biotie Therapies Inc
Original Assignee
Synosia Therapeutics Inc
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 Synosia Therapeutics Inc filed Critical Synosia Therapeutics Inc
Priority to US12/187,166 priority Critical patent/US20090041800A1/en
Assigned to SYNOSIA THERAPEUTICS, INC. reassignment SYNOSIA THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WOIWODE, TOM, MORAN, MARK, PICKFORD, LESLEY
Publication of US20090041800A1 publication Critical patent/US20090041800A1/en
Assigned to BIOTIE THERAPIES, INC. reassignment BIOTIE THERAPIES, INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: SYNOSIA THERAPUETICS, INC.
Assigned to BIOTIE THERAPIES, INC. reassignment BIOTIE THERAPIES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SYNOSIA THERAPEUTICS, INC.
Priority to US14/099,882 priority patent/US10561638B2/en
Priority to US16/736,609 priority patent/US20200383951A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/417Imidazole-alkylamines, e.g. histamine, phentolamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/08Drugs for disorders of the alimentary tract or the digestive system for nausea, cinetosis or vertigo; Antiemetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • A61P25/12Antiepileptics; Anticonvulsants for grand-mal
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/20Hypnotics; Sedatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/32Alcohol-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/34Tobacco-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/36Opioid-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/06Antiarrhythmics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • Certain methods of treating patients suffering from or susceptible to at least one symptom of abuse of, dependence on, or withdrawal from at least one substance with Compound A are also provided. Also provided are certain methods of treating at least one phase of substance dependence on at least one substance in patients and certain methods of treating at least one phase of cocaine dependence in patients.
  • Substance abuse and dependence are characterized by substance craving, seeking, and use with loss of control in limiting intake of the substance. These behaviors occur despite significant substance use related problems and at the expense of other behaviors.
  • substance alcohol or illicit drug
  • the dopamine ⁇ -hydroxylase (DBH) inhibitor disulfiram is the most effective pharmacologic treatment for cocaine dependence currently available.
  • DBH dopamine ⁇ -hydroxylase
  • disulfiram non-specifically inhibits several enzymes, including aldehyde dehydrogenase and plasma esterases.
  • Disulfiram and related compounds chelate copper, which is a necessary cofactor for a variety of enzymes, including aldehyde dehydrogenase, plasma esterases and DBH.
  • aldehyde dehydrogenase By inhibiting aldehyde dehydrogenase, disulfiram alters the metabolism of alcohol (ethanol), producing the disulfiram-ethanol reaction. This reaction consists of flushing, nausea, and hypotension.
  • disulfiram As a treatment of cocaine dependence.
  • treatment with disulfiram reduced the positive subjective effects produced by cocaine. Patients with comorbid alcohol and cocaine dependence had improved outcomes when treated with disulfiram, up to 500 mg.
  • buprenorphine-maintained opiate- and cocaine-dependent patients reduced cocaine use during treatment with disulfiram.
  • results from a large clinical trial suggested that disulfiram 250 mg per day was associated with reduced cocaine use compared to placebo, regardless of alcohol use pattern or type of psychotherapy provided.
  • 112 cocaine-dependent volunteers were randomized to placebo or disulfiram, and provided one of two psychotherapies.
  • Disulfiram treatment was associated with reduced cocaine use documented by the provision of fewer cocaine-positive urine samples compared to placebo treatment. The effect size was modest and this outcome remains to be replicated.
  • DBH norepinephrine
  • NE norepinephrine
  • DBH is expressed in noradrenergic neurons and is localized within synaptic vesicles and is released along with NE.
  • DBH can be measured in the plasma, and the concentration of DBH is highly heritable and variability in activity is largely accounted for by variability at the DBH locus.
  • the T variant ( ⁇ 1021 ⁇ COT) is associated with diminished DBH gene transcription and with lower DBH activity. This allele is fairly common. The frequency of the T allele is reported to be 20% among African-Americans, 22% among Northern European Americans and 16% among Japanese. The corresponding haplotype frequencies are 0.32, 0.34, and 0.09 for these populations, respectively.
  • Disulfiram more effectively reduces cocaine use in patients with the DBH C ⁇ T genotype associated with lower DBH activity. Presumably, disulfiram more completely inhibits DBH in those with lower DBH activity, so that disulfiram is more effective in those with the lower activity C ⁇ T genotype. The observation that disulfiram is more effective in patients with the low-activity DBH C ⁇ T genotype confirms that inhibition of DBH is a key mechanism of action for disulfiram as a therapy for cocaine dependence.
  • kits for treating patients suffering from or susceptible to at least one symptom of abuse of, dependence on, or withdrawal from at least one substance include administering to the patient a therapeutically effective amount of Compound A.
  • the methods include administering to the patient a therapeutically effective amount of Compound A.
  • the methods include administering to the patient a therapeutically effective amount of Compound A.
  • FIG. 1 shows the effect size achieved with sample sizes ranging from 5 to 15.
  • FIG. 2 shows the details of the individual enzymatic assays.
  • FIG. 3 shows a table with the affinities (IC50s or PKi) of nepicastat with DBH and a range of selected enzymes and receptors.
  • FIG. 4 shows the effects of nepicastat on % inhibition of enzyme activity.
  • FIG. 5 shows the urinary dopamine levels in normal volunteers after 24 hour treatment with nepicastat.
  • FIG. 6 shows the norepinephrine levels in the cortex in SHRs dosed with vehicle or varying doses of nepicastat.
  • FIG. 7 shows the dopamine levels in the cortex in SHRs dosed with vehicle or varying doses of nepicastat.
  • FIG. 8 shows the dopamine/norepinephrine ratio in the cortex in SHRs dosed with vehicle or varying doses of nepicastat.
  • FIG. 9 shows the norepinephrine levels in the mesenteric artery in SHRs dosed with vehicle or varying doses of nepicastat.
  • FIG. 10 shows the dopamine levels in the mesenteric artery in SHRs dosed with vehicle or varying doses of nepicastat.
  • FIG. 11 shows the dopamine/norepinephrine ratio in the mesenteric artery in SHRs dosed with vehicle or varying doses of nepicastat.
  • Compound A includes (S)-5-Aminomethyl-l-(5,7-difluoro-1,2,3,4-tetrahydronaphth-2-yl)-2,3-dihydro-2-thioxo-1H-imidazole, (R)-5 -Aminomethyl-1-(5,7-difluoro-1,2,3,4-tetrahydronaphth-2-yl)-2,3-dihydro-2-thioxo-1H-imidazole, and mixtures thereof, as well as pharmaceutically acceptable salts thereof, such as the hydrochloride salt.
  • nepicastat is used ((S)-5-Aminomethyl-1-(5,7-difluoro-1,2,3,4-tetrahydronaphth-2-yl)-2,3 -dihydro-2-thioxo-1H-imidazole hydrochloride).
  • Compound B refers to (R)-5-Aminomethyl-1-(5,7-difluoro-1,2,3,4-tetrahydronaphth-2-yl)-2,3 -dihydro-2-thioxo-1H-imidazole, as well as pharmaceutically acceptable salts thereof, such as the hydrochloride salt.
  • “Pharmaceutically acceptable salts” include, but are not limited to salts with inorganic acids, such as hydrochlorate, phosphate, diphosphate, hydrobromate, sulfate, sulfinate, nitrate, and like salts; as well as salts with an organic acid, such as malate, maleate, fumarate, tartrate, succinate, citrate, acetate, lactate, methanesulfonate, p-toluenesulfonate, 2-hydroxyethylsulfonate, benzoate, salicylate, stearate, and alkanoate such as acetate, HOOC—(CH2)n-COOH where n is 0-4, and like salts.
  • inorganic acids such as hydrochlorate, phosphate, diphosphate, hydrobromate, sulfate, sulfinate, nitrate, and like salts
  • an organic acid such as malate, maleate, fumarate, tartrate, succinate
  • the free base can be obtained by basifying a solution of the acid salt.
  • an addition salt particularly a pharmaceutically acceptable addition salt, may be produced by dissolving the free base in a suitable organic solvent and treating the solution with an acid, in accordance with conventional procedures for preparing acid addition salts from base compounds.
  • Those skilled in the art will recognize various synthetic methodologies that may be used to prepare non-toxic pharmaceutically acceptable addition salts.
  • patient refers to a mammal. In certain embodiments, the term “patient” refers to a human.
  • administer refers to either directly administering Compound A or a composition thereof to a patient.
  • treat refers to partially or completely alleviating, inhibiting, preventing, ameliorating and/or relieving the condition, or at least one symptom thereof.
  • uffer or “suffering” as used herein refers to one or more conditions that a patient has been diagnosed with, or is suspected to have.
  • susceptible refers to having a likelihood of being affected by at least one symptom of a condition.
  • substance abuse often involves symptoms of physical and/or psychological “dependence.” Also, when the substance of abuse is withdrawn from a dependent individual, the individual often develops certain symptoms including sleep and mood disturbance and intense craving of the substance of abuse, known as “withdrawal.”
  • the methods described herein encompass treatment of substance abuse itself, dependence, and also of withdrawal.
  • substance abuse can be defined with reference to criteria set forth in the Diagnostic and Statistical Manual of Mental Disorders, 4 th Ed. Text revision (2000) (“DSM-IV TR”), which was prepared by the Task Force on DSM-IV of the American Psychiatric Association.
  • DSM-IV TR Diagnostic and Statistical Manual of Mental Disorders, 4 th Ed. Text revision (2000)
  • substance abuse is defined as maladaptive pattern of substance abuse leading to clinically significant impairment or distress, as manifested by at least one of the following symptoms, occurring within a 12-month period: (1) recurrent substance use resulting in a failure to fulfill major role obligations at work, school, or home; (2) recurrent substance use in situations in which it is physically hazardous; (3) recurrent substance-related legal problems; and (4) continued substance use despite having persistent or recurrent social or interpersonal problems caused or exacerbated by the effects of the substance.
  • the DSM-IV TR requires that the symptoms of substance abuse have never met the criteria for substance dependence.
  • treatment of substance abuse with nepicastat reduces the amount or frequency of substance use in a patient.
  • treatment of substance abuse with Compound A in a patient reduces at least one DSM-IV TR symptom for substance abuse.
  • treatment with Compound A in a patient reduces at least one symptom of substance abuse which includes by way of example and without limitation at least one of euphoria, apathy, irritability, recklessness, poor judgment, compulsion, aggression, anger, craving for the substance being abused, and mood disorders.
  • treatment with Compound A reduces the substance craving induced by a stressful event in a patient.
  • the phrase “reduces a symptom” refers to reducing at least one of the frequency and amplitude of a symptom of a condition in a patient. In certain embodiments the patient enters remission and no longer experiences the symptom.
  • the phrase “increases a symptom” refers to increasing at least one of the frequency and amplitude of a symptom of a condition in a patient.
  • the term “substance dependence,” as used herein, can be defined with reference to criteria set forth in the DSM-IV TR.
  • the symptoms for substance dependence set forth in DSM-IV TR is a pattern of substance use, leading to clinically significant impairment or distress as manifested by at least three selected from the following group, occurring at any time within the same twelve month period: (1) tolerance as defined by either (a) a need for substantially increased amounts of the substance to achieve the desired effect; or (b) substantially diminished effect with continued use of the same amount of the substance; (2) withdrawal, as demonstrated by either (a) the characteristic withdrawal syndrome for the specific substance; or (b) the same, or a closely related substance is taken to relieve or avoid withdrawal symptoms; (3) the substance is often taken in larger amounts or over a longer period than was intended; (4) there is a persistent desire or unsuccessful efforts to cut down or control substance use; (5) a great deal of time is spent in activities to obtain the substance, use the substance, or recover from its effects; (6) important social, occupational or recreational activities are given up or reduced because of substance use; and
  • Substance dependence can be with physiological dependence, where evidence of tolerance or withdrawal is present, or without physiological dependence, where no evidence of tolerance or withdrawal is present.
  • Compound A treatment of substance dependence reduces the amount or frequency of substance use by a patient.
  • Compound A treatment of substance dependence reduces at least one DSM-IV TR symptom for substance dependence in a patient.
  • treatment with Compound A in a patient reduces at least one symptom of substance dependence which includes by way of example and without limitation at least one of euphoria, apathy, irritability, recklessness, poor judgment, compulsion, aggression, anger, craving for the substance depended upon, and mood disorders.
  • treatment with Compound A reduces the substance craving induced by a stressful event in a patient.
  • remission refers to a state during which the occurrence of at least one symptom of substance abuse or dependence has been reduced. In some embodiments, the term remission does not apply if the patient is on agonist therapy or in a controlled environment where access to the relevant substance is restricted. In some embodiments remission refers to a state during which the occurrence of at least one symptom of substance abuse or dependence does not occur. In some embodiments, remission refers to a state during which all symptoms of substance abuse or dependence have been reduced in a patient. In some embodiments, remission refers to a state during which no symptoms of substance abuse or dependence occur. In some embodiments, remission refers to a state during which substance use does not occur.
  • the remission is characterized by at least one of early full remission, early partial remission, sustained full remission, and sustained partial remission and only applies after none of the symptoms for substance abuse and dependence have been present for at least one month.
  • the definition of these four types of remission are based on the interval of time that has elapsed since the cessation of dependence (early versus sustained remission) and whether there is continued presence of at least one symptom of substance dependence or abuse (partial versus full remission).
  • the qualifier “early full remission” is used when for at least one month, but for less than twelve months, no symptom of substance dependence or substance abuse has been met.
  • the qualifier “early partial remission” is used when for at least one month but less than 12 months, at least one symptom of substance dependence or substance abuse has been met, but the criteria for substance dependence or substance abuse have not been met.
  • sustained full remission is used when none of the symptoms of substance dependence or substance abuse have been met at any time during a period of at least twelve months.
  • sustained partial remission is used when for at least twelve months, at least one symptom of substance dependence or substance abuse has been met, but the criteria for substance dependence or substance abuse have not been met.
  • Compound A treatment promotes remission in a patient. In some embodiments, Compound A treatment prolongs a period of remission period in a patient.
  • a stressful event can cause remission to end in a patient.
  • relapse occurs at the end of remission.
  • Compound A treatment reduces the likelihood that remission will end in a patient after a stressful event.
  • Compound A treatment promotes at least one of early partial remission, sustained full remission, sustained partial remission, and sustained full remission.
  • “Withdrawal” refers to a collection of symptoms that arise when administration of a relevant substance is reduced, delayed, or stopped.
  • the substance-specific symptoms of withdrawal can cause clinically significant distress or impairment in social, occupational or other important areas of functioning, for example. These symptoms are not due to a general medical condition and are not better accounted for by another mental disorder. Withdrawal usually, but not necessarily, is associated with substance dependence.
  • treatment with Compound A reduces at least one symptom of withdrawal in a patient.
  • withdrawal symptoms include for example and without limitation apathy, irritability, recklessness, poor judgment, compulsion, aggression, anger, substance craving, mood disorders, and sleep disorders.
  • treatment with Compound A reduces the substance craving induced by a stressful event in a patient.
  • substance dependence can be characterized by the presence of at least one of the following conditions characterized in the DSM-IV TR: Alcohol Abuse; Alcohol Dependence; Alcohol Intoxication; Alcohol Intoxication Delirium; Alcohol Withdrawal; Alcohol Withdrawal Delirium; Alcohol-Induced Anxiety Disorder; Alcohol-Induced Mood Disorder; Alcohol-Induced Persisting Amnestic Disorder; Alcohol-Induced Persisting Dementia; Alcohol-Induced Psychotic Disorder, With Delusions; Alcohol-Induced Psychotic Disorder, With Hallucinations; Alcohol-Induced Sexual Dysfunction; Alcohol-Induced Sleep Disorder; Alcohol-Related Disorder Not Otherwise Specified (NOS); Amphetamine Abuse; Amphetamine Dependence; Amphetamine Intoxication; Amphetamine Intoxication Delirium; Amphetamine Withdrawal; Amphetamine-Induced Anxiety Disorder; Amphetamine-Induced Mood Disorder; Amphetamine-Induced
  • on agonist therapy refers to being treated with an agonist for substance abuse, dependence, or withdrawal.
  • agonist refers to a factor including, but not limited to a chemical compound, such as a small molecule or a complex organic compound or a protein, that triggers a response in a patient that is at least one response or partial response of the substance being abused, depended upon, or withdrawn from by the patient.
  • Opioid Dependence On Agonist Therapy refers to Opioid Dependence on methadone therapy.
  • Withdrawal symptoms can arise upon reduction of any of a variety of substances.
  • the discontinued use of tobacco products all of which contain nicotine, typically results in the onset of nicotine withdrawal conditions.
  • Individuals often suffer the symptoms of nicotine withdrawal as a consequence of the discontinued use of tobacco in any form, including, but not limited to smoking of cigarette, cigar, or pipe tobacco, or the oral or intranasal ingestion of tobacco or chewing tobacco.
  • Such oral or intranasal tobacco includes, but is not limited to snuff and chewing tobacco.
  • the cessation of nicotine use or reduction in the amount of nicotine use is often followed within 24 hours by symptoms including dysphoric, depressed mood; light-headedness; insomnia; irritability, frustration or anger; anxiety; nervous tremor; difficulty concentrating; restlessness; decreased heart rate; increased appetite or weight gain; and the craving for tobacco or nicotine.
  • symptoms often cause clinically significant distress or impairment in social, occupational, or other important areas of functioning.
  • the methods described herein may be used to alleviate one or more symptoms attributed to nicotine withdrawal when such symptoms are not due to a general medical condition and are not better accounted for by another medical disorder.
  • the present method is also helpful to those who have replaced, or partially replaced, their use of tobacco with the use of nicotine replacement therapy. Thus, such patients can be assisted to reduce and even eliminate entirely their dependence on nicotine in all forms.
  • the discontinuing or reduction in administration of an opioid often results in the presence of a characteristic opioid withdrawal condition.
  • This withdrawal condition can also be precipitated by administration of an opioid antagonist such as naloxone or naltrexone after opioid use.
  • opioid antagonist such as naloxone or naltrexone after opioid use.
  • Opioid withdrawal is characterized by symptoms that are generally opposite to the opioid agonist effects. These withdrawal symptoms may include anxiety; restlessness; muscle aches, often in the back and legs; craving for opioids; irritability and increased sensitivity to pain; dysphoric mood; nausea or vomiting; lacrimation; rhinorrhoea; papillary dilation; piloerection; sweating; diarrhea; yawning; fever; and insomnia.
  • Ethanol withdrawal conditions are characterized by symptoms that begin when blood concentrations of ethanol decline sharply, within 4 to 12 hours after ethanol use has been stopped or reduced. These ethanol withdrawal symptoms include craving for ethanol; autonomic hyperactivity (such as sweating or pulse rate greater than 100); hand tremor; insomnia; nausea; vomiting; transient visual, tactile, or auditory hallucinations or illusions; psychomotor agitation; anxiety; and grand mal seizures. These symptoms often cause clinically significant distress or impairment in social, occupational, or other important areas of functioning.
  • the methods described herein may be used to alleviate one or more symptoms attributed to ethanol withdrawal when such symptoms are not due to a general medical condition and are not better accounted for by another medical disorder.
  • Cocaine abuse and dependence can cause cognitive, behavioral, and physiological symptoms. Symptoms of cocaine abuse and dependence can include varying degrees of attention deficit hyperactivity disorder and euphoria; increased energy, excitement, and sociability; less hunger and fatigue; a marked feeling of physical and mental strength; dysphoria; decreased sensation of pain; and craving for cocaine. Respiratory effects include symptoms such as bronchitis, shortness of breath, and chest pain, and cardiovascular effects include symptoms such as heart palpitations, arrhythmia, cardiomyopathy, and heart attacks. Symptoms also include dilated pupils, nausea, vomiting, headache, vertigo, anxiety, dizziness, psychosis, and confusion.
  • Compound A treatment reduces at least one symptom of cocaine abuse and dependence in a patient.
  • nepicstat treatment increases at least one negative subjective symptom of cocaine abuse and dependence.
  • Cocaine withdrawal symptoms can include a fatigue, lack of pleasure, depression, irritability, sleep disorders, increased appetite, pyschomotor retardation, agitation, extreme suspicion, and craving for cocaine.
  • Compound A treatment reduces at least one symptom of cocaine withdrawal.
  • Substance dependence can be characterized by the phases: acquisition, maintenance, extinction, and relapse.
  • acquisition refers to a phase of substance dependence in which dependence on the substance is initiated and acquired by a patient.
  • Compound A treatment inhibits the development of the acquisition phase in a patient.
  • Compound A treatment of the acquisition phase reduces at least one of the amount or frequency of substance use by a patient.
  • Compound A treatment of the acquisition phase reduces at least one DSM-IV symptom of substance abuse and dependence in a patient.
  • Compound A treatment of the acquisition phase reduces at least one symptom of substance abuse and dependence which include by way of example and without limitation at least one of euphoria, apathy, irritability, recklessness, poor judgment, compulsion, aggression, anger, craving for the substance being abused or depended upon, and mood disorders.
  • treatment with Compound A reduces the substance craving induced by a stressful event in a patient during the acquisition phase.
  • “Maintenance” refers to a phase of substance dependence in which there is stable administration to or use of the substance by a patient. In some embodiments, a 10% variance in at least one of the amount and frequency of substance use by a patient is considered a stable behavior. In some embodiments, Compound A treatment of the maintenance phase reduces at least one of the amount and frequency of substance use by a patient. In some embodiments, Compound A treatment of the maintenance phase reduces at least one DSM-IV symptom of substance abuse and dependence in a patient.
  • Compound A treatment of the maintenance phase reduces at least one symptom of substance abuse and dependence which includes by way of example and without limitation at least one of euphoria, apathy, irritability, recklessness, poor judgment, compulsion, aggression, anger, craving for the substance being abused or depended upon, and mood disorders.
  • treatment with Compound A reduces the substance craving induced by a stressful event in a patient during the maintenance phase.
  • “Extinction” refers to a phase of substance dependence in which the substance is not provided to a patient or a patient abstains from use of the substance. In some embodiments, the dependence on the substance is extinguished or reduced in the extinction phase. In some embodiments, at least one withdrawal symptom occurs in the extinction phase. In some embodiments, Compound A treatment promotes the development of the extinction phase in a patient. In some embodiments, Compound A treatment of the extinction phase reduces at least one DSM-IV symptom of substance abuse and dependence in a patient.
  • Compound A treatment during the extinction phase reduces at least one symptom of substance abuse and dependence which includes by way of example and without limitation at least one of euphoria, apathy, irritability, recklessness, poor judgment, compulsion, aggression, anger, craving for the substance being abused or depended upon, and mood disorders.
  • Compound A treatment reduces the withdrawal symptoms in a patient in the extinction phase.
  • treatment with Compound A reduces the substance craving induced by a stressful event in a patient in the extinction phase.
  • Relapse refers to recurrence of at least one symptom of substance abuse or dependence after a period of abstinence in a patient. In some embodiments, the relapse occurs at the end of remission. In some embodiments, a patient has undergone extinction training prior to relapse. In some embodiments, relapse occurs after drug priming, stress, or exposure to an environment related cue or stimulation that was previously associated with substance use. In some embodiments, Compound A treatment reduces the frequency of relapse in a patient. In some embodiments, Compound A treatment of the relapse phase reduces at least one DSM-IV symptom of substance abuse and dependence in a patient.
  • Compound A treatment of the relapse phase reduces at least one symptom of substance abuse and dependence which includes by way of example and without limitation at least one of euphoria, apathy, irritability, recklessness, poor judgment, compulsion, aggression, anger, craving for the substance being abused or depended upon, and mood disorders.
  • Compound A treatment reduces the withdrawal symptoms in a patient during the relapse phase.
  • treatment with Compound A reduces the substance craving induced by a stressful event in a patient during the relapse phase.
  • Treatment of substance abuse, dependence, and withdrawal may be conducted in stages.
  • an initial period of abstinence from substance use is preferred before induction of treatment with Compound A in a patient.
  • an initial low dose of Compound A is administered to a patient.
  • the amount of Compound A administered to a patient is escalated until a desired therapeutic response is observed.
  • the amount of Compound A is escalated in order to determine the optimal dose to treat the condition while minimizing symptoms, side effects, and cravings for the substance in a patient.
  • Compound A treatment promotes remission.
  • the dose of Compound A is unchanged or tapered off after remission is reached in a patient.
  • the methods include administering to the patient a therapeutically effective amount of Compound A.
  • the at least one substance is selected from a drug of abuse and a medication.
  • the drug of abuse is selected from a psychostimulant agent, an opioid, a hallucinogen, an inhalant, a sedative, a tranquilizer, a hypnotic, an anxiolytic, and an illicit substance.
  • the psychostimulant agent is a beta-phenylisopropylamine derivative.
  • the beta-phenylisopropylamine derivative is selected from amphetamine, dextroamphetamine, and methamphetamine.
  • the psychostimulant agent is selected from ecstasy, phenmetrazine, methylphenidate, diethylpropion, pemoline, mazindol, ( ⁇ ) cathione, and fenfluramine.
  • the opioid is selected from Lortab, Tramadol, heroin, methadone, hydrocodone, and oxycodone.
  • the hallucinogen is selected from psilocybin, a hallucinogenic mushroom, lysergic acid diethylamide (LSD), phencyclidine (PCP), and ketamine.
  • the inhalant is selected from benzene, toluene, o-xylene, m-xylene, p-xylene, ethylbenzene, fluorobenzene, o-difluorobenzene, 1,3,5-triflurobenzene, 1,2,4-trifluorobenzene, pentafluorotoluene, pentafluorobenzene, and perfluorobenzene.
  • the medication is selected from an anesthetic, an analgesic, an anticholinergic agent, an antihistamine, a muscle relaxant, a nonsteroidal anti-inflammatory medication, an over the counter medication, and an antidepressant medication.
  • the drug of abuse is cocaine, alcohol, caffeine, opium, cannabinoid, cannabis, benzodiazapine carisprodol, tobacco, nicotine, Vicodin, Lorcet, Percocet, Percodan, and Tylox.
  • the drug of abuse is cocaine and the Compound A reduces at least one symptom of cocaine abuse and dependence in the patient selected from attention deficit hyperactivity disorder; euphoria; increased energy, excitement and sociability; less hunger and fatigue; a marked feeling of physical and mental strength; decreased sensation of pain; bronchitis; shortness of breath; chest pain; heart palpitations; arrhythmia; cardiomyopathy; heart attack; dilated pupils; nausea; vomiting; headache; vertigo; dizziness; anxiety; pychosis; confusion; nasal irritation; nasal crusting; recurrent nosebleeds; nasal stuffiness; facial pain; dysphoria; and craving for cocaine.
  • the drug of abuse is cocaine and the Compound A increases at least one negative subjective symptom of cocaine abuse and dependence.
  • the drug of abuse is cocaine and the Compound A reduces at least one symptom of cocaine withdrawal selected from fatigue, lack of pleasure, depression, irritability, sleep disorders, increased appetite, pyschomotor retardation, agitation, extreme suspicion, and craving for cocaine.
  • the Compound A treatment improves a score of the patient on at least one of the attention deficit hyperactivity disorder IV rating scale (ADHD-IV), Hamilton Depression Scale (HAM-D), Hamilton Anxiety Scale (HAM-A), Beck Depression inventory (BDI), apathy scale from Neuropsychiatric Inventory, and a cognitive function rating scale.
  • ADHD-IV attention deficit hyperactivity disorder IV rating scale
  • HAM-D Hamilton Depression Scale
  • HAM-A Hamilton Anxiety Scale
  • BDI Beck Depression inventory
  • apathy scale from Neuropsychiatric Inventory and a cognitive function rating scale.
  • the cognitive function rating scale is selected from the Wechsler Adult Intelligence Scale-Revised (WAIS-R), Wechsler Memory Scale-Revised (WMS-R), Rey Auditory Verbal Learning Test (RAVLT, Trials I-VII), Rey Complex Figure Test (RCFT), and the Trail Making Test (TMT, Parts A and B).
  • WAIS-R Wechsler Adult Intelligence Scale-Revised
  • WMS-R Wechsler Memory Scale-Revised
  • RAVLT Rey Auditory Verbal Learning Test
  • RCFT Rey Complex Figure Test
  • TTT Trail Making Test
  • the Compound A reduces at least one symptom of substance abuse in the patient selected from recurrent substance use resulting in a failure to fulfill major role obligations at work, school, or home; recurrent substance use in situations in which it is physically hazardous; recurrent substance-related legal problems; and continued substance use despite having persistent or recurrent social or interpersonal problems caused or exacerbated by the effects of the substance.
  • the Compound A reduces at least one symptom of substance dependence in the patient selected from tolerance; withdrawal; the substance is often taken in larger amounts or over a longer period then was intended; there is a persistent desire and/or unsuccessful efforts to cut down or control substance use; a great deal of time is spent in at least one of activities to obtain the substance, use the substance, and recover from its effects; at least one of important social, occupational and recreational activities are given up and/or reduced because of substance use; and the substance use is continued despite knowledge of having a persistent and/or recurrent physical and/or psychological problem that is likely to have been caused or exacerbated by the substance.
  • the Compound A promotes remission in the patient.
  • the remission is characterized by at least one of early full remission, early partial remission, sustained full remission, and sustained partial remission.
  • the Compound A prolongs a period of remission in the patient.
  • the methods further include treatment with at least one of contingency management and cognitive behavioral therapy.
  • the methods further include co-administering a therapeutically effective amount of least one other agent selected from a selective serotonin reuptake inhibitor (SSRI), a serotonin-norepinephrine reuptake inhibitor (SNRI), a norepinephrine reuptake inhibitor (NRI), a norepinephrine-dopamine reuptake inhibitor (NDRI), a serotonin 5-hydroxytryptamine1A (5HT1A) antagonist, a dopamine ⁇ -hydroxylase inhibitor, an adenosine receptor antagonist, an adenosine A2A receptor antagonist, a monoamine oxidase inhibitor (MAOI), a monoamine oxidase B inhibitor, a sodium channel blocker, a calcium channel blocker, a central and peripheral alpha adrenergic receptor antagonist, a central alpha adrenergic agonist, a central or peripheral beta adrenergic receptor antagonist, a NK
  • the at least one other agent is a SSRI selected from paroxetine, sertraline, citalopram, escitalopram, and fluoxetine. In some embodiments, the at least one other agent is a SNRI selected from duloxetine, mirtazapine, and venlafaxine. In some embodiments, the at least one other agent is a NRI selected from bupropion and atomoxetine. In some embodiments, the at least other agent is the NDRI bupropion. In some embodiments, the at least one other agent is the dopamine ⁇ -hydroxylase inhibitor disulfiram. In some embodiments, the at least one other agent is the adenosine A2A receptor antagonist istradefylline.
  • the at least one other agent is a sodium channel blocker selected from lamotrigine, carbamazepine, oxcarbazepine, and valproate. In some embodiments, the at least one other agent is a calcium channel blocker selected from nimodopone, lamotrigine, and carbamazepine. In some embodiments, the at least one other agent is the central and peripheral alpha adrenergic receptor antagonist prazosin. In some embodiments, the at least one other agent is the central alpha adrenergic agonist clonidine. In some embodiments, the at least one other agent is the central or peripheral beta adrenergic receptor antagonist propranolol.
  • the at least one other agent is an atypical antidepressant/antipsychotic selected from bupropion, olanzepine, risperidone, and quetiapine.
  • the at least one other agent is a tricyclic selected from amitriptyline, amoxapine, desipramine, doxepin, imipramine, nortriptyline, protiptyline, and trimipramine.
  • the at least one other agent is an anticonvulsant selected from phenytoin, lamotrigine, carbamazepine, oxcarbazepine, valproate, topiramate, tiagabine, vigabatrin, and levetiracetam.
  • the at least one other agent is the glutamate antagonist topiramate.
  • the at least one other agent is a GABA agonist selected from baclofen, valproate, and topiramate.
  • the at least one other agent is the dopamine metabolism enzyme inhibitor carbidopa.
  • the at least one other agent is the partial dopamine D2 agonist aripiprazole.
  • the at least one other agent is an opioid receptor antagonist selected from naltrexone and naloxone. In some embodiments, the at least one other agent is a mood stabilizer selected from carbamazepine and lithium. In some embodiments, the at least one other agent is a direct or indirect dopamine agonist selected from dopamine, bromocriptine, pergolide, amantadine, mazindole, and methylphenidate. In some embodiments, the at least other agent is the partial 5HT1 agonist gepirone. In some embodiments, the at least other agent is the serotonin 5HT2 antagonist ritanserin. In some embodiments, the at least other agent is the opioid methadone.
  • the at least other agent is the partial opioid agonist buprenorphine. In some embodiments, the at least other agent is the partial nicotinic agonist champix. In some embodiments, the at least one other agent is an inhalant selected from benzene, toluene, o-xylene, m-xylene, p-xylene, ethylbenzene, fluorobenzene, o-difluorobenzene, 1,3,5-triflurobenzene, 1,2,4-trifluorobenzene, pentafluorotoluene, pentafluorobenzene, and perfluorobenzene.
  • the methods further include co-administering a therapeutically effective amount of least one other agent selected from benzodiazepine, levodopa, carisprodol, modafenil, acamprosate, gamma-butyrolactone, gamma-hydroxybutyrate, opium, psilopcybin, hallucinogenic mushroom, tobacco, and nicotine.
  • the Compound A is administered to the patient after a period of abstinence from substance use by the patient.
  • the therapeutically effective amount of Compound A in the patient is determined by escalating the amount of Compound A administered to the patient until a desired therapeutic response is observed.
  • the amount of Compound A is tapered off after remission is reached in the patient.
  • the amount of Compound A is unchanged after remission is reached in the patient.
  • the at least one phase of substance dependence is selected from acquisition, maintenance, extinction, and relapse.
  • the methods include administering to the patient a therapeutically effective amount of Compound A.
  • the Compound A inhibits the development of the acquisition phase in the patient.
  • the Compound A promotes the development of the extinction phase in the patient.
  • the Compound A reduces the frequency of relapse in the patient.
  • the at least one substance is selected from a drug of abuse and a medication.
  • the drug of abuse is selected from a psychostimulant agent, an opioid, a hallucinogen, an inhalant, a sedative, a tranquilizer, a hypnotic, an anxiolytic, and an illicit substance.
  • the psychostimulant agent is a beta-phenylisopropylamine derivative.
  • the beta-phenylisopropylamine derivative is selected from amphetamine, dextroamphetamine, and methamphetamine.
  • the psychostimulant agent is selected from ecstasy, phenmetrazine, methylphenidate, diethylpropion, pemoline, mazindol, ( ⁇ ) cathione, and fenfluramine.
  • the opioid is selected from Lortab, Tramadol, heroin, methadone, hydrocodone, and oxycodone.
  • the hallucinogen is selected from psilocybin, a hallucinogenic mushroom, lysergic acid diethylamide (LSD), phencyclidine (PCP), and ketamine.
  • the inhalant is selected from benzene, toluene, o-xylene, m-xylene, p-xylene, ethylbenzene, fluorobenzene, o-difluorobenzene, 1,3,5-triflurobenzene, 1,2,4-trifluorobenzene, pentafluorotoluene, pentafluorobenzene, and perfluorobenzene.
  • the medication is selected from an anesthetic, an analgesic, an anticholinergic agent, an antihistamine, a muscle relaxant, a nonsteroidal anti-inflammatory medication, an over the counter medication, and an antidepressant medication.
  • the drug of abuse is alcohol, caffeine, opium, cannabinoid, cannabis, benzodiazapine, carisprodol, tobacco, nicotine, Vicodin, Lorcet, Percocet, Percodan, and Tylox.
  • the Compound A treatment improves a score of the patient on at least one of the ADHD-IV, HAM-D, HAM-A, BDI, apathy scale from Neuropsychiatric Inventory, and a cognitive function rating scale.
  • the cognitive function rating scale is selected from the WAIS-R, WMS-R, RAVLT, Trials I-VII, RCFT, and TMT, Parts A and B.
  • the Compound A reduces in the patient at least one of the amount and frequency of use of the at least one substance by the patient. In some embodiments, the Compound A reduces in the patient at least one symptom of abuse of, dependence on, or withdrawal from the at least one substance. In some embodiments, the Compound A reduces at least one symptom of substance abuse in the patient selected from recurrent substance use resulting in a failure to fulfill major role obligations at work, school, or home; recurrent substance use in situations in which it is physically hazardous; recurrent substance-related legal problems; and continued substance use despite having persistent or recurrent social or interpersonal problems caused or exacerbated by the effects of the substance.
  • the Compound A reduces at least one symptom of substance dependence in the patient selected from tolerance; withdrawal; the substance is often taken in larger amounts or over a longer period then was intended; there is a persistent desire and/or unsuccessful efforts to cut down or control substance use; a great deal of time is spent in at least one of activities to obtain the substance, use the substance, and recover from its effects; at least one of important social, occupational and recreational activities are given up and/or reduced because of substance use; and the substance use is continued despite knowledge of having a persistent and/or recurrent physical and/or psychological problem that is likely to have been caused or exacerbated by the substance.
  • the Compound A promotes remission in the patient.
  • the remission is characterized by at least one of early full remission, early partial remission, sustained full remission, and sustained partial remission.
  • the Compound A prolongs a period of remission in the patient.
  • the methods further include treatment with at least one of contingency management and cognitive behavioral therapy.
  • the methods further include co-administering a therapeutically effective amount of least one other agent selected from a selective serotonin reuptake inhibitor (SSRI), a serotonin-norepinephrine reuptake inhibitor (SNRI), a norepinephrine reuptake inhibitor (NRI), a norepinephrine-dopamine reuptake inhibitor (NDRI), a serotonin 5-hydroxytryptamine1A (5HTIA) antagonist, a dopamine ⁇ -hydroxylase inhibitor, an adenosine receptor antagonist, an adenosine A2A receptor antagonist, a monoamine oxidase inhibitor (MAOI), a monoamine oxidase B inhibitor, a sodium channel blocker, a calcium channel blocker, a central and peripheral alpha adrenergic receptor antagonist, a central alpha adrenergic agonist, a central or peripheral beta adrenergic receptor antagonist, a NK
  • the at least one other agent is a SSRI selected from paroxetine, sertraline, citalopram, escitalopram, and fluoxetine. In some embodiments, the at least one other agent is a SNRI selected from duloxetine, mirtazapine, and venlafaxine. In some embodiments, the at least one other agent is a NRI selected from bupropion and atomoxetine. In some embodiments, the at least other agent is the NDRI bupropion. In some embodiments, the at least one other agent is the dopamine ⁇ -hydroxylase inhibitor disulfiram. In some embodiments, the at least one other agent is the adenosine A2A receptor antagonist istradefylline.
  • the at least one other agent is a sodium channel blocker selected from lamotrigine, carbamazepine, oxcarbazepine, and valproate. In some embodiments, the at least one other agent is a calcium channel blocker selected from nimodopone, lamotrigine, and carbamazepine. In some embodiments, the at least one other agent is the central and peripheral alpha adrenergic receptor antagonist prazosin. In some embodiments, the at least one other agent is the central alpha adrenergic agonist clonidine. In some embodiments, the at least one other agent is the central or peripheral beta adrenergic receptor antagonist propranolol.
  • the at least one other agent is an atypical antidepressant/antipsychotic selected from bupropion, olanzepine, risperidone, and quetiapine.
  • the at least one other agent is a tricyclic selected from amitriptyline, amoxapine, desipramine, doxepin, imipramine, nortriptyline, protiptyline, and trimipramine.
  • the at least one other agent is an anticonvulsant selected from phenytoin, lamotrigine, carbamazepine, oxcarbazepine, valproate, topiramate, tiagabine, vigabatrin, and levetiracetam.
  • the at least one other agent is the glutamate antagonist topiramate.
  • the at least one other agent is a GABA agonist selected from baclofen, valproate, and topiramate.
  • the at least one other agent is the dopamine metabolism enzyme inhibitor carbidopa.
  • the at least one other agent is the partial dopamine D2 agonist aripiprazole.
  • the at least one other agent is an opioid receptor antagonist selected from naltrexone and naloxone. In some embodiments, the at least one other agent is a mood stabilizer selected from carbamazepine and lithium. In some embodiments, the at least one other agent is a direct or indirect dopamine agonist selected from dopamine, bromocriptine, pergolide, amantadine, mazindole, and methylphenidate. In some embodiments, the at least other agent is the partial 5HT1 agonist gepirone. In some embodiments, the at least other agent is the serotonin 5HT2 antagonist ritanserin. In some embodiments, the at least other agent is the opioid methadone.
  • the at least other agent is the partial opioid agonist buprenorphine. In some embodiments, the at least other agent is the partial nicotinic agonist champix. In some embodiments, the at least one other agent is an inhalant selected from benzene, toluene, o-xylene, m-xylene, p-xylene, ethylbenzene, fluorobenzene, o-difluorobenzene, 1,3,5-triflurobenzene, 1,2,4-trifluorobenzene, pentafluorotoluene, pentafluorobenzene, and perfluorobenzene.
  • the methods further include co-administering a therapeutically effective amount of least one other agent selected from benzodiazepine, levodopa, carisprodol, modafenil, acamprosate, gamma-butyrolactone, gamma-hydroxybutyrate, opium, psilopcybin, hallucinogenic mushroom, tobacco, and nicotine.
  • the Compound A is administered to the patient after a period of abstinence from substance use by the patient.
  • the therapeutically effective amount of Compound A in the patient is determined by escalating the amount of Compound A administered to the patient until a desired therapeutic response is observed.
  • the amount of Compound A is tapered off after remission is reached in the patient.
  • the amount of Compound A is unchanged after remission is reached in the patient.
  • the at least one phase is selected from acquisition, maintenance, extinction, and relapse.
  • the methods include administering to the patient a therapeutically effective amount of Compound A.
  • the Compound A inhibits the development of the acquisition phase in the patient.
  • the Compound A promotes development of the extinction phase in the patient.
  • the Compound A reduces the frequency of relapse in the patient.
  • the Compound A reduces in the patient at least one symptom of abuse of, dependence on, or withdrawal from cocaine.
  • the Compound A reduces at least one symptom of cocaine abuse in the patient selected from recurrent cocaine use resulting in a failure to fulfill major role obligations at work, school, or home; recurrent cocaine use in situations in which it is physically hazardous; recurrent cocaine-related legal problems; and continued cocaine use despite having persistent or recurrent social or interpersonal problems caused or exacerbated by the effects of the cocaine.
  • the Compound A reduces at least one symptom of cocaine dependence in the patient selected from tolerance; withdrawal; the cocaine is often taken in larger amounts or over a longer period then was intended; there is a persistent desire or unsuccessful efforts to cut down or control cocaine use; a great deal of time is spent in activities to obtain the cocaine, use the cocaine, or recover from its effects; important social, occupational or recreational activities are given up or reduced because of cocaine use; and the cocaine use is continued despite knowledge of having a persistent or recurrent physical or psychological problem that is likely to have been caused or exacerbated by the cocaine.
  • the Compound A reduces at least one symptom of cocaine abuse and dependence selected from attention deficit hyperactivity disorder; euphoria; increased energy, excitement and sociability; less hunger and fatigue; a marked feeling of physical and mental strength; decreased sensation of pain; bronchitis; shortness of breath; chest pain; heart palpitations; arrhythmia; cardiomyopathy; heart attack; dilated pupils; nausea; vomiting; headache; vertigo; dizziness; anxiety; pychosis; confusion; nasal irritation; nasal crusting; recurrent nosebleeds; nasal stuffiness; facial pain; dysphoria; and craving for cocaine.
  • the Compound A increases at least one negative subjective symptom of cocaine abuse and dependence.
  • the Compound A reduces at least one symptom of cocaine withdrawal selected from fatigue, lack of pleasure, depression, irritability, sleep disorders, increased appetite, pyschomotor retardation, agitation, extreme suspicion, and craving for cocaine.
  • the Compound A improves a score of the patient on at least one of ADHD-IV, HAM-D, HAM-A, BDI, apathy scale from Neuropsychiatric Inventory, and a cognitive function rating scale.
  • the cognitive function rating scale is selected from WAIS-R, WMS-R, RAVLT, Trials I-VII, RCFT, and TMT, Parts A and B.
  • the Compound A reduces at least one of the amount and frequency of cocaine use by the patient. In some embodiments, the Compound A promotes remission in the patient. In some embodiments, the remission is characterized by at least one of early full remission, early partial remission, sustained full remission, and sustained partial remission. In some embodiments, the Compound A prolongs a period of remission in the patient. In some embodiments, the methods further include treatment with at least one of contingency management and cognitive behavioral therapy.
  • the methods further include co-administering a therapeutically effective amount of least one other agent selected from a selective serotonin reuptake inhibitor (SSRI), a serotonin-norepinephrine reuptake inhibitor (SNRI), a norepinephrine reuptake inhibitor (NRI), a norepinephrine-dopamine reuptake inhibitor (NDRI), a serotonin 5-hydroxytryptamine1A (5HT1A) antagonist, a dopamine ⁇ -hydroxylase inhibitor, an adenosine receptor antagonist, an adenosine A2A receptor antagonist, a monoamine oxidase inhibitor (MAOI), a monoamine oxidase B inhibitor, a sodium channel blocker, a calcium channel blocker, a central and peripheral alpha adrenergic receptor antagonist, a central alpha adrenergic agonist, a central or peripheral beta adrenergic receptor antagonist, a NK
  • the at least one other agent is a SSRI selected from paroxetine, sertraline, citalopram, escitalopram, and fluoxetine. In some embodiments, the at least one other agent is a SNRI selected from duloxetine, mirtazapine, and venlafaxine. In some embodiments, the at least one other agent is a NRI selected from bupropion and atomoxetine. In some embodiments, the at least other agent is the NDRI bupropion. In some embodiments, the at least one other agent is the dopamine ⁇ -hydroxylase inhibitor disulfiram. In some embodiments, the at least one other agent is the adenosine A2A receptor antagonist istradefylline.
  • the at least one other agent is a sodium channel blocker selected from lamotrigine, carbamazepine, oxcarbazepine, and valproate. In some embodiments, the at least one other agent is a calcium channel blocker selected from nimodopone, lamotrigine, and carbamazepine. In some embodiments, the at least one other agent is the central and peripheral alpha adrenergic receptor antagonist prazosin. In some embodiments, the at least one other agent is the central alpha adrenergic agonist clonidine. In some embodiments, the at least one other agent is the central or peripheral beta adrenergic receptor antagonist propranolol.
  • the at least one other agent is an atypical antidepressant/antipsychotic selected from bupropion, olanzepine, risperidone, and quetiapine.
  • the at least one other agent is a tricyclic selected from amitriptyline, amoxapine, desipramine, doxepin, imipramine, nortriptyline, protiptyline, and trimipramine.
  • the at least one other agent is an anticonvulsant selected from phenytoin, lamotrigine, carbamazepine, oxcarbazepine, valproate, topiramate, tiagabine, vigabatrin, and levetiracetam.
  • the at least one other agent is the glutamate antagonist topiramate.
  • the at least one other agent is a GABA agonist selected from baclofen, valproate, and topiramate.
  • the at least one other agent is the dopamine metabolism enzyme inhibitor carbidopa.
  • the at least one other agent is the partial dopamine D2 agonist aripiprazole.
  • the at least one other agent is an opioid receptor antagonist selected from naltrexone and naloxone. In some embodiments, the at least one other agent is a mood stabilizer selected from carbamazepine and lithium. In some embodiments, the at least one other agent is a direct or indirect dopamine agonist selected from dopamine, bromocriptine, pergolide, amantadine, mazindole, and methylphenidate. In some embodiments, the at least other agent is the partial 5HT1 agonist gepirone. In some embodiments, the at least other agent is the serotonin 5HT2 antagonist ritanserin. In some embodiments, the at least other agent is the opioid methadone.
  • the at least other agent is the partial opioid agonist buprenorphine. In some embodiments, the at least other agent is the partial nicotinic agonist champix. In some embodiments, the at least one other agent is an inhalant selected from benzene, toluene, o-xylene, m-xylene, p-xylene, ethylbenzene, fluorobenzene, o-difluorobenzene, 1,3,5-triflurobenzene, 1,2,4-trifluorobenzene, pentafluorotoluene, pentafluorobenzene, and perfluorobenzene.
  • the methods further include co-administering a therapeutically effective amount of least one other agent selected from benzodiazepine, levodopa, carisprodol, modafenil, acamprosate, gamma-butyrolactone, gamma-hydroxybutyrate, opium, psilopcybin, hallucinogenic mushroom, tobacco, and nicotine.
  • the Compound A is administered to the patient after a period of abstinence from cocaine use by the patient.
  • the therapeutically effective amount of Compound A in the patient is determined by escalating the amount of Compound A administered to the patient until a desired therapeutic response is observed.
  • the amount of Compound A is tapered off after remission from cocaine dependence is reached in the patient. In some embodiments, the amount of Compound A is unchanged after remission from cocaine dependence is reached in the patient. In some embodiments, the Compound A treats at least one symptom of abuse of, dependence on, or withdrawal from at least one secondary substance in the patient. In some embodiments, the at least one secondary substance is selected from a drug of abuse and a medication. In some embodiments, the drug of abuse is selected from a psychostimulant agent, an opioid, a hallucinogen, an inhalant, a sedative, a tranquilizer, a hypnotic, an anxiolytic, and an illicit substance.
  • the psychostimulant agent is a beta-phenylisopropylamine derivative.
  • the beta-phenylisopropylamine derivative is selected from amphetamine, dextroamphetamine, and methamphetamine.
  • the psychostimulant agent is selected from ecstasy, phenmetrazine, methylphenidate, diethylpropion, pemoline, mazindol, ( ⁇ ) cathione, and fenfluramine.
  • the opioid is selected from Lortab, Tramadol, heroin, methadone, hydrocodone, and oxycodone.
  • the hallucinogen is selected from psilocybin, a hallucinogenic mushroom, lysergic acid diethylamide (LSD), phencyclidine (PCP), and ketamine.
  • the inhalant is selected from benzene, toluene, o-xylene, m-xylene, p-xylene, ethylbenzene, fluorobenzene, o-difluorobenzene, 1,3,5-triflurobenzene, 1,2,4-trifluorobenzene, pentafluorotoluene, pentafluorobenzene, and perfluorobenzene.
  • the medication is selected from an anesthetic, an analgesic, an anticholinergic agent, an antihistamine, a muscle relaxant, a nonsteroidal anti-inflammatory medication, an over the counter medication, and an antidepressant medication.
  • the drug of abuse is alcohol, caffeine, opium, cannabinoid, cannabis, benzodiazapine, carisprodol, tobacco, nicotine, Vicodin, Lorcet, Percocet, Percodan, and Tylox.
  • compositions include acids, bases, enol ethers, and esters, esters, hydrates, solvates, and prodrug forms.
  • the derivative is selected such that its pharmokinetic properties are superior with respect to at least one characteristic to the corresponding neutral agent.
  • the Compound A may be derivatized prior to formulation.
  • a therapeutically effective amount of Compound A or a pharmaceutically acceptable derivative may vary widely depending on the severity of the addiction or dependence, the age and relative health of the subject, the potency of the compound used and other factors.
  • a therapeutically effective amount is from about 0.1 milligram per kg (mg/kg) body weight per day to about 50 mg/kg body weight per day. In other embodiments the amount is about 1.0 to about 10 mg/kg/day. Therefore, in certain embodiments a therapeutically effective amount for a 70 kg human is from about 7.0 to about 3500 mg/day, while in other embodiments it is about 70 to about 700 mg/day.
  • Compound A will be administered as pharmaceutical compositions by one of the following routes: oral, systemic (e.g., transdermal, intranasal or by suppository) or parenteral (e.g., intramuscular, intravenous or subcutaneous).
  • routes oral, systemic (e.g., transdermal, intranasal or by suppository) or parenteral (e.g., intramuscular, intravenous or subcutaneous).
  • compositions can, by way of example and without limitation, take the form of tablets, pills, capsules, semisolids, powders, sustained release formulations, solutions, suspensions, elixirs, aerosols, or any other appropriate composition and are comprised of, in general, Compound A in combination with at least one pharmaceutically acceptable excipient.
  • Acceptable excipients are, by way of example and without limitation, non-toxic, aid administration, and do not adversely affect the therapeutic benefit of the compound.
  • excipient may be, for example, any solid, liquid, semisolid or, in the case of an aerosol composition, gaseous excipient that is generally available to one of skill in the art.
  • Solid pharmaceutical excipients include by way of example and without limitation starch, cellulose, talc, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, magnesium stearate, sodium stearate, glycerol monostearate, sodium chloride, dried skim milk, and the like.
  • Liquid and semisolid excipients may be selected from for example and without limitation water, ethanol, glycerol, propylene glycol and various oils, including those of petroleum, animal, vegetable or synthetic origin (e.g., peanut oil, soybean oil, mineral oil, sesame oil, etc.).
  • Preferred liquid carriers particularly for injectable solutions, include by way of example and without limitation water, saline, aqueous dextrose and glycols.
  • Compressed gases may be used to disperse the compound in aerosol form.
  • Inert gases suitable for this purpose are by way of example and without limitation nitrogen, carbon dioxide, nitrous oxide, etc.
  • the pharmaceutical preparations can by way of example and without limitation, moreover, contain preservatives, solubilizers, stabilizers, wetting agents, emulsifiers, sweeteners, colorants, flavorants, salts for varying the osmotic pressure, buffers, masking agents or antioxidants. In certain embodiments, they can contain still other therapeutically valuable substances.
  • suitable pharmaceutical carriers and their formulations are described in A. R. Alfonso Remington's Pharmaceutical Sciences 1985, 17th ed. Easton, Pa.: Mack Publishing Company.
  • the amount of Compound A in the composition may vary widely depending for example, upon the type of formulation, size of a unit dosage, kind of excipients and other factors known to those of skill in the art of pharmaceutical sciences.
  • the final composition will comprise from 10% w to 90% w of the compound, preferably 25% w to 75% w, with the remainder being the excipient or excipients.
  • the pharmaceutical composition is administered in a single unit dosage form for continuous treatment or in a single unit dosage form ad libitum when relief of symptoms is specifically required.
  • nepicastat Following oral administration of [ 14 C]-nepicastat, the majority of the radioactivity in plasma was associated with nepicastat, an N-linked glucuronide of nepicastat (Metabolite 2, M2), and an unidentified polar fraction (M1). There was no significant accumulation of nepicastat with multiple dosing and T 1/2 was similar after single and multiple doses. T 1/2 was 10-14 hours.
  • the pharmacokinetics of nepicastat after a single 40 mg dose was compared in men and women.
  • the AUC in women was approximately 43% greater than in men and the Cmax approximately 23% greater in women than in men.
  • the T1/2 was longer in women than in men. Comparing the pharmacokinetics of nepicastat following 10 days of dosing with a 40 mg dose, the AUC was higher in healthy subjects than in patients with CHEF, with no difference in the T1/2. There was no significant accumulation with multiple dosing in either population.
  • nepicastat In a study designed to assess the effects of nepicastat on cognitive function, subjects treated with 5 or 40 mg of nepicastat demonstrated no significant impairment of mood, sleep, or cognition. In studies of thyroid uptake of 123 I, doses of 5, 40, and 100 mg of nepicastat demonstrated no differences from placebo. Reduction of uptake after a single 200 mg dose of nepicastat was significantly greater than placebo, but significantly less than that following a 10 mg dose of methimazole. In single-dose Phase I studies, doses from 5 to 800 mg (dose calculation based on the hydrochloride salt) of nepicastat were generally well tolerated in healthy men.
  • Treatment at each dose level are daily for 4 days, or well over 4 half-lives of nepicastat, which is 10 to 14 hours.
  • participants receive cocaine 0 mg, 10 mg, 20 mg, and 40 mg in that order.
  • Cocaine is administered at hourly intervals, providing sufficient time for both the cardiovascular and subjective effects to return to baseline.
  • Cardiovascular indices are carefully monitored using continuous EKG and frequent blood pressure during all procedures involving the administration of cocaine, and stopping parameters are in place so that cocaine is not be administered if cardiovascular indices exceed preset limits. Previous studies have shown that 6 doses of 32 mg cocaine given at 14-minute intervals is safe, and extending the inter-dose interval to 1 hour may enhance safety further.
  • Blood samples are collected for analysis of the pharmacokinetics of 10 mg cocaine administered on the 3 rd day of treatment with 0 mg nepicastat and again on the 3 rd day of treatment with 80 mg and 160 mg nepicastat.
  • the effects of nepicastat on the pharmacokinetics of cocaine are studied. Based on existing information, no interaction is expected.
  • Participants are required to have used cocaine by the IV route to avoid exposing participants to routes of administration that produce more intensive interoceptive effects.
  • the age criteria were selected primarily to avoid enrolling participants with undiagnosed cardiovascular disease. Participants with active HIV disease are excluded to avoid potential exacerbation of their underlying disease; participants with asymptomatic HIV are included because this group is at high risk for cocaine dependence. Participants with asthma (or who take asthma medications) are excluded due to potential adverse interactions between beta agonist medications and cocaine.
  • Cocaine produces prototypical stimulant effects by inhibiting the uptake of DA, NE, and serotonin into presynaptic storage granules.
  • Cocaine has a short elimination half-life, about 90 min.
  • the principal clinical effects of cocaine are psychomotor activation and increases in sympathetic tone, evident as increases in heart rate and blood pressure.
  • Cocaine is administered at up to 40 mg in single doses and up to 200 mg in self-administration sessions consisting of 10 doses of 20 mg administered at 13 min intervals. These doses are modest compared to amounts that participants in these studies have reported using daily; typical daily dosing patterns are on average 250 mg to 500 mg or more.
  • Cocaine is administered IV, so availability is complete.
  • Cocaine is metabolized primarily to benzoylecgonine by plasma esterases that are not known to be affected by nepicastat.
  • Benzyoleconine and other minor metabolites are excreted renally.
  • Ascending doses of nepicastat (0 mg, 80 mg, and 160 mg) are administered at 7 AM. Treatment at each dose level is continued for 4 days.
  • nepicastat is not an enzyme inhibitor, though pharmacokinetic assessment of the 10 mg does of cocaine administered on the 3 rd day of treatment at each dose level of nepicastat can confirm this. Because nepicastat reduces the synthesis of NE, the rewarding effects of cocaine may be lower during treatment with nepicastat. Because nepicastat increases plasma and brain concentrations of DA, DA-mediated side effects such as paranoia may occur. These symptoms were not observed during the trials for CHF, but stimulants were not administered in those studies.
  • participant Following consent, participants are required to submit a cocaine-positive urine sample for documentation of ongoing drug use. Some participants (limited by the number of devices available) are also asked to wear a telemetry device during screening and throughout the study that records heart rate and movement. Data from this device can identify drug use episodes based on changes in these parameters.
  • mice are conducted at approximately the same time of day for a given participant.
  • Cocaine is administered in an experimental room.
  • Cocaine is administered using a syringe pump, which administers the correct dose of cocaine or saline placebo over 2 minutes.
  • heart rate and blood pressure are monitored.
  • the Actiheart MiniMitter is used to measure heart rate and movement prior to admission in some volunteers (the number is limited by the number of devices available).
  • the MiniMitter attaches to the participants' skin using paste and non-invasively records EKG and movement for up to two weeks. The data can be downloaded to a PC for analysis later.
  • the MINI is a short, structured diagnostic interview developed in 1990 by psychiatrists and clinicians in the United States and Europe for DSM-IV TR and ICD-10 psychiatric disorders.
  • the MINI is the structured psychiatric interview of choice for psychiatric evaluation and outcome tracking in clinical psychopharmacology trials and epidemiological studies, and is the most widely used psychiatric structured diagnostic interview instrument in the world. This instrument can be used to determine whether the subject met DSM-IV TR criteria for drug dependence and to rule out any major psychiatric disorders (e.g., affective disorders, schizophrenia).
  • the Addiction Severity Index-Lite Clinical Factors (ASI-Lite CF) version is administered by a trained research staff member during screening.
  • the ASI-Lite is the interviewer's estimate of the severity of the participant's status in seven areas (medical, employment, drug use, alcohol use, legal, family/social, and psychological).
  • the Lite version is a shorter version of the ASI that still retains all questions used to calculate the ASI composite scores.
  • BDI Beck Depression Inventory
  • ADHD ADHD-IV rating scale
  • the apathy scale from the Neuropsychiatric Inventory are collected at baseline.
  • DNA is collected with buccal swabs applied to Whatman FTA cards. These cards allow safe and stable storage of biological samples for DNA extraction.
  • the anticipated yield of genomic DNA is 50-100 ⁇ g, which is adequate for over 500 genotype assays using currently available methods.
  • Genotypes are determined using 5′ Exonuclease-based (Taqman) genotyping assays. Assays are developed by Applied Biosystems (ABI; Assays by Design). Allele discrimination are performed using the ABI 3730 realtime PCR cycler.
  • Blood samples for analysis of the pharmacokinetics of cocaine are collected during treatment with 0 mg nepicastat (study day 1) and during treatment with 80 mg and 160 mg nepicastat (study days 4 and 8). Blood samples are collected at ⁇ 15, 20, 30, 40, 50, 60, 90, 120, 180, 240, 300, 360, 420 and 480 minutes following dosing of 10 mg cocaine on the 3 rd day of treatment with each dose level of nepicastat. Note that other doses of cocaine (0-40 mg) are administered on the 4 th day of treatment with each dose level of nepicastat, so the pharmacokinetic assessment does not interfere with the other assessments. Blood is collected and plasma separated and frozen at ⁇ 70° C. until analyzed.
  • DBH is stored in NE storage granules and is released along with NE. Plasma DBH thus gives a good index of enzymatic activity within the CNS. Blood is sampled daily at 10 AM (prior to cocaine/placebo dosing) and stored for subsequent analysis. DBH activity is measured by using the tyramine-octopamine method using a high performance liquid chromatographic-fluorometric system, as described previously. This allows examination of changes in DBH over time, providing an insight into the pharmacodynamics of nepicastat's inhibition of DBH. The BDI is administered repeatedly throughout the protocol to monitor changes in mood.
  • VAS computerized visual analogue scale
  • VAS measures are collected prior to cocaine administration and at 5, 10, 15, 20, 30, and 45 minutes following drug administration.
  • an infusion corresponding to a color (“blue” or “green”), and money.
  • the color corresponds to the dose (cocaine 0 mg or 20 mg) administered to the subject during the sample session.
  • participants make 10 choices for the infusion (cocaine 0 mg IV in one session and cocaine 20 mg in the other) or money.
  • the participant makes a series of choices between ascending value money options ($0.05, $0.05, $0.05, $0.05, $1, $4, $7, $10, $13, and $16) or cocaine (0 mg or 20 mg/IV/infusion) using a patient-controlled analgesia (PCA) pump.
  • PCA patient-controlled analgesia
  • Infusion choices are performed by the participant using the PCA button, while choices for money are indicated verbally to the investigator. Infusions take place over 2-min followed by a 3-min time-out period. As such, selections are made at 5-min intervals.
  • the table shows the Experimental choice sessions with 16 total participants.
  • a sample size of 12 in the nepicastat-treated group allows detection of medium to large effects, which is appropriate for an initial assessment.
  • the plot ( FIG. 1 ) shows the effect size achieved with sample sizes ranging from 5 to 15. Increasing the sample size above 12 would enhance analytical power to detect differences between the treatments but at increasing cost.
  • the placebo-treated group is included only to maintain the blind and is not intended to serve as a comparison group.
  • the analysis focuses primarily on the effects of nepicastat in the nepicastat-treated group.
  • the placebo-treated group is included primarily to maintain the blind.
  • Side effects and adverse events (AEs) are tabulated and compared across treatment conditions using ANOVA or Chi-square.
  • Subjective and cardiovascular effects produced by IV cocaine and placebo during treatment with nepicastat are compared to those produced during treatment with placebo using repeated measures (time being the repeated measure) analysis of variance (ANOVA), peak effect one-way ANOVA, and if indicated, area under the curve ANOVA.
  • Bovine and human dopamine ⁇ -hydroxylase activity were assayed by measuring the conversion of tyramine to octopamine.
  • Bovine adrenal dopamine ⁇ -hydroxylase was obtained from Sigma Chemicals (St Louis, Mo., USA) whereas human dopamine ⁇ -hydroxylase was purified from the culture medium of the neuroblastoma cell line SK—N—SH.
  • the assay was performed at pH 5.2 and 32° C. in a medium containing 0.125 M NaAc, 10 mM fumarate, 0.5-2 ⁇ M CuSO4, 0.1 mg.ml ⁇ 1 catalase, 0.1 mM tyramine and 4 mM ascorbate.
  • a substrate mixture containing catalase, tyramine and ascorbate was added to initiate the reaction (final volume of 200 ⁇ l).
  • Samples were incubated with or without the appropriate concentration of nepicastat or Compound B at 37° C. for 30 to 40 minutes.
  • the reaction was quenched by the stop solution containing 25 mM EDTA and 240 ⁇ M 3-hydroxytyramine (internal standard).
  • the samples were analyzed for octopamine by reverse phase high pressure liquid chromatography (HPLC) using ultraviolet-detection at 280 nM.
  • the HPLC chromatography run was carried out at the flow rate of 1 ml.min ⁇ 1 using a LiChroCART 125-4 RP-18 column and isocratic elution with 10 mM acidic acid, 10 mM 1-heptane sulfonic acid, 12 mM tetrabutyl ammonium phosphate and 10% methanol.
  • the remaining percent activity was calculated based on controls, corrected using internal standards and fitted to a non-linear four-parameter concentration-response curve.
  • Nepicastat S-enantiomer
  • Compound B R-enantiomer
  • IC 50 's for nepicastat were 8.5 ⁇ 0.8 nM and 9.0 ⁇ 0.8 nM for the bovine and human enzyme, respectively.
  • Compound B was slightly less potent (IC 50 's of 25.1 ⁇ 0.6 nM and 18.3 ⁇ 0.6 nM for the bovine and human enzyme, respectively) than nepicastat.
  • Nepicastat was shown to be a potent inhibitor of human and bovine dopamine ⁇ -hydroxylase in vitro. The inhibitory effects of the compound were stereospecific since the S-enantiomer (nepicastat) was marginally, but significantly, more potent than the R-enantiomer (Compound B).
  • nepicastat The activity of nepicastat at twelve selected enzymes and receptors was determined using established assays. A brief account of the principle underlying each of the enzymatic assays is given in FIG. 2 . Binding data were analyzed by iterative curve-fitting to a four parameter logistic equation. Ki values were calculated from IC 50 values using the Cheng-Prusoff equation. Enzyme inhibitory activity was expressed as IC 50 (concentration required to produce 50% inhibition of enzyme activity).
  • Nepicastat had negligible affinity (IC 50 s or Kis>10 82 M) for a range of other enzymes (tyrosine hydroxylase, acetyl CoA synthetase, acyl CoA-cholesterol acyl transferase, Ca 2+ /calmodulin protein kinase II, cyclooxygenase-I, HMG-CoA reductase, neutral endopeptidase, nitric oxide synthase, phosphodiesterase III, phospholipase A 2 , and protein kinase C) and neurotransmitter receptors ( ⁇ 1A , ⁇ 1B , ⁇ 2A , ⁇ 2B , ⁇ 1 and ⁇ 2 adrenoceptors, M 1 muscarinic receptors, D 1 and D 2 dopamine receptors, ⁇ opioid receptors, 5-HT 1A , 5-HT 2A , and 5-HT 2C serotonin receptors).
  • nepicastat (S)-5-aminomethyl-1-(5,7-difluoro-1,2,3,4-tetrahydronaphth-2-yl)-1,3-dihydroimidazole-2-thione hydrochloride
  • Compound B the corresponding R-enantiomer
  • mice Male SHRs (15-16 weeks old, Charles River, Wilmington, Mass., USA) were used in in vivo studies. On the day of the study, animals were weighed and randomly assigned to be dosed with either vehicle (control) or the appropriate dose of nepicastat (3, 10, 30 or 100 mg.kg ⁇ 1 , po) or Compound B (30 mg.kg ⁇ 1 , po) three consecutive times, twelve hours apart. At six hours after the third dose, the rats were anaesthetized with halothane, decapitated and tissues (cerebral cortex, mesenteric artery and left ventricle) were rapidly harvested, weighed, placed in iced perchloric acid (0.4 M), frozen in liquid nitrogen and stored at ⁇ 70° C. until subsequent analysis.
  • vehicle control
  • nepicastat 3, 10, 30 or 100 mg.kg ⁇ 1 , po
  • Compound B 30 mg.kg ⁇ 1 , po
  • tissues were homogenized by brief sonication and centrifuged at 13,000 rpm for 30 minutes at 4° C. The supernatant, spiked with 3,4-dihydroxybenzylamine (internal standard), was assayed for noradrenaline and dopamine by HPLC using electrochemical detection.
  • Basal tissue catecholamine content ( ⁇ g.g ⁇ 1 wet weight) in control animals were as follows : mesenteric artery (noradrenaline, 10.40 ⁇ 1.03; dopamine, 0.25 ⁇ 0.02), left ventricle (noradrenaline, 1.30 ⁇ 0.06; dopamine, 0.02 ⁇ 0.00) and cerebral cortex (noradrenaline, 0.76 ⁇ 0.03; dopamine, 0.14 ⁇ 0.01).
  • Nepicastat produced dose-dependent reduction in noradrenaline content and enhancement of dopamine content and dopamine/noradrenaline ratio in the three tissues which were studied.
  • mice Male beagle dogs (10-16 kg, Marshall Farms USA Inc, North Rose, N.Y., USA) were also used in in vivo studies. On the day of the study, dogs were weighed and randomly assigned to be orally dosed with either empty capsules (control) or the appropriate dose of nepicastat (0.05, 0.5, 1.5 or 5 mg.kg ⁇ 1 ; po, b.i.d.) for 5 days. At six hours following the first dose on day-5, the dogs were euthanized with pentobarbital and the tissues (cerebral cortex, renal artery, left ventricle) were rapidly harvested. The tissues were subsequently processed and analyzed for noradrenaline and dopamine.
  • nepicastat 0.05, 0.5, 1.5 or 5 mg.kg ⁇ 1 ; po, b.i.d.
  • Basal tissue catecholamine content ( ⁇ g.g ⁇ 1 wet weight) in control animals were as follows: renal artery (noradrenaline, 10.7 ⁇ 1.05; dopamine, 0.22 ⁇ 0.01), left ventricle (noradrenaline, 2.11 ⁇ 0.18; dopamine, 0.07 ⁇ 0.03) and cerebral cortex (noradrenaline, 0.26 ⁇ 0.02; dopamine, 0.03 ⁇ 0.00).
  • nepicastat produced a dose-dependent reduction in noradrenaline content and enhancement of dopamine content and dopamine/noradrenaline ratio in the three tissues which were studied.
  • Male beagle dogs were randomized to be orally dosed with either empty capsules (control) or nepicastat ( 2 mg.kg ⁇ 1 , po, b.i.d.) for 15 days.
  • Daily venous blood samples were drawn, six hours after the first dose, for measurement of plasma concentrations of dopamine and noradrenaline.
  • the samples were collected in tubes containing heparin and glutathione, centrifuged at ⁇ 4° C. and the separated plasma was stored at ⁇ 70° C. until analysis.
  • nepicastat 2 mg.kg ⁇ 1 , b.i.d, po
  • plasma noradrenaline and dopamine concentrations were 460.3 ⁇ 59.6 and 34.4 ⁇ 11.9 ⁇ g.ml ⁇ 1 , respectively, in the control group and 401.9 ⁇ 25.5 and 41.1 ⁇ 8.8 ⁇ g.ml ⁇ 1 , respectively, in the nepicastat-treated group.
  • nepicastat (2 mg.kg ⁇ 1 , b.i.d, po) produced significant decreases in plasma concentrations of noradrenaline and increases in plasma concentrations of dopamine and dopamine/noradrenaline ratio.
  • Inhibitory modulation of sympathetic nerve function is an attractive therapeutic strategy for the management of congestive heart failure, inasmuch as elevated activity of this system has been implicated in the progressive worsening of the disease.
  • the aim of this study was to pharmacologically characterize the effects of nepicastat, a compound which modulates noradrenaline synthesis in sympathetic nerves by inhibiting the enzyme dopamine ⁇ -hydroxylase.
  • Compound B was less potent than nepicastat which is consistent with the lower IC 50 of the former enantiomer for the enzyme.
  • dopamine/noradrenaline ratio was also elevated, there did not appear to be stoichiometric replacement of noradrenaline with dopamine. The most likely explanation for this finding is that tissue levels of dopamine may have been underestimated due to intraneuronal metabolism of dopamine.
  • nepicastat The ability of nepicastat to alter catecholamine levels in the cerebral cortex suggests that the drug does penetrate the blood brain barrier. In dogs, the magnitude of the changes in catecholamines in the cerebral cortex appeared comparable to those in peripheral tissues. In SHRs, however, nepicastat, at low doses ( ⁇ 10 mg.kg ⁇ 1 ), produced significant changes in noradrenaline and dopamine content in peripheral tissues without affecting catecholamines in the cerebral cortex. This suggests that, at least in SHRs, the drug does possess modest peripheral selectivity.
  • Plasma noradrenaline concentrations provide a useful measure of overall sympathetic nerve activity although this parameter may be influenced by alterations in neuronal uptake and metabolic clearance of the catecholamine.
  • Baseline concentrations of noradrenaline in the plasma were surprisingly elevated in the dogs and is, perhaps, a reflection of the initial stress induced by the phlebotomy blood-sampling procedure. Nevertheless, compared to the control group, nepicastat produced significant decreases in plasma noradrenaline concentrations consistent with reduced transmitter synthesis and release although an indirect effect, secondary to facilitation of neuronal uptake or metabolic clearance, cannot be discounted.
  • noradrenaline represents a small fraction of the total neuronal noradrenaline stores
  • an inhibitor of noradrenaline biosynthesis would affect noradrenaline release only after existing stores of the catecholamine have been sufficiently depleted. Accordingly, the decreases in plasma noradrenaline concentrations did not attain statistical significance until 4 days of dosing with nepicastat suggesting gradual modulation of the sympathetic nervous system.
  • Inhibitors of dopamine ⁇ -hydroxylase such as nepicastat
  • this class of drugs would attenuate, but not abolish, noradrenaline release and, second, they produce gradual modulation of the system thereby obviating the need for dose-titration.
  • Another advantage of nepicastat over ⁇ -blockers is that it enhances dopamine levels which, via agonism of dopamine receptors, may have salutary effects on renal function such as renal vasodilation, diuresis and natriuresis.
  • Nepicastat is a potent, selective and orally active inhibitor of dopamine ⁇ -hydroxylase which may be of value in the treatment of cardiovascular disorders associated with over-activation of the sympathetic nervous system.
  • nepicastat was based upon the chiral reduction of tetralone 3 (available from the AlCl 3 -catalyzed Friedel-Crafts reaction of 3,5-difluorophenylacetyl chloride with ethylene in CH 2 Cl 2 at 31 65° C.) under the conditions described by Terashima (LAH, ( ⁇ )-1R,2S-N-methylephedrine, 2-ethylaminopyridine) to give R-(+)-tetralol 4a (92-95% ee), that was converted to the R-(+)-mesylate, followed by reaction with sodium azide afforded a mixture (9:1) of azide and dihydronaphthalene 7.
  • tetralone 3 available from the AlCl 3 -catalyzed Friedel-Crafts reaction of 3,5-difluorophenylacetyl chloride with ethylene in CH 2 Cl 2 at 31 65° C.
  • Terashima LAH, ( ⁇
  • the azide was hydrogenated and the product treated with anhydrous HCl to give S-( ⁇ )-amine hydrochloride, converted by a Strecker reaction (formaldehyde bisulfite complex and KCN) to S-( ⁇ )-aminonitrile. Formation of the heterocycle was accomplished by sequential diformylation of aminonitrile followed by subsequent treatment with thiocyanic acid. Competing hydrolysis of the nitrile afforded comparable amounts of the primary amide. Reduction of nitrile to amine (93-96% ee) was accomplished using LAH in THF.
  • the enantiomer (91.6% ee) was available by the same above described route using (+)-1S,2R-N-methylephedrine as a chiral auxiliary in the Terashima reduction of ketone.
  • the absolute configuration of the chiral center was based upon literature precedence of the previously described S-( ⁇ )-2-tetralol.
  • UV spectra were recorded on a Varian Cary 3 UV-Visible spectrometer, Leeman Labs Inc. Optical rotations were measured in a Perkin-Elmer Model 141 polarimeter. Chiral HPLC measurements were performed on a Regis Chiral AGP column (4.6 ⁇ 100 mm) eluting with 2% acetonitrile-98% 20 mM KH 2 PO 4 (pH 4.7) at 1 mL/min at 20° C.
  • DBH activity was assayed by measuring the conversion of tyramine to octopamine.
  • Bovine DBH from adrenal glands was obtained from Sigma Chemical Co (St Louis, Mo.). Human secretory DBH was purified from the culture medium of the neuroblastoma cell line SK—N—SH. The assay was performed at pH 5.2 and 32° C. in 0.125 M NaOAc, 10 mM fumarate, 0.5-2 ⁇ M CUSO 4 , 0.1 mg/mL catalase, 0.1 mM tyramine and 4 mM ascorbate. In a typical assay, 0.5-1 milliunits of enzyme were added to the reaction mixture and then a substrate mixture containing catalase, tyramine and ascorbate was added to initiate the reaction (final volume of 200 ⁇ L).
  • FIG. 3 shows a table describing the interaction of nepicastat at DBH and a range of selected enzymes and receptors. Nepicastat showed weak affinity for a range of other enzymes and neurotransmitter receptors.
  • nepicastat to spontaneously hypertensive rats (SHRs) and normal dogs produced potent and dose-dependent increases in tissue dopamine (DA)/norepinephrine (NE) ratios in peripheral arteries (renal or mesenteric), left ventricle and cerebral cortex.
  • Chronic oral administration of nepicastat to normal dogs also produced sustained increases in the plasma DA/NE ratio.
  • acute oral administration of nepicastat produced dose-dependent and long-lasting (>4 h) antihypertensive effects and also attenuation of the pressor responses to pre-ganglionic sympathetic nerve stimulation. Serum T 3 and T 4 levels were unaffected by a dose (6.2 mg/kg, po, b.i.d.
  • nepicastat has been clinical evaluated for the treatment of congestive heart failure.
  • CHF Congestive heart failure
  • SNS sympathetic nervous system
  • RAS renin-angiotensin system
  • ACE angiotensin-converting enzyme
  • Inhibition of the SNS with ⁇ -adrenoceptor antagonists is a promising approach that is currently under clinical evaluation.
  • An alternative strategy to directly modulate the SNS is inhibition of norepinephrine (NE) biosynthesis via inhibition of dopamine ⁇ -hydroxylase (DBH), the enzyme responsible for conversion of NE to dopamine (DA).
  • Inhibition of DBH would be expected to reduce tissue levels of NE and elevate tissue levels of DA thereby increasing the tissue DA/NE ratio.
  • This approach has potential advantages over ⁇ -adrenoceptor antagonists, such as reduced stimulation of ⁇ -adrenoceptors and elevated DA levels that can produce renal vasodilation, natriuresis and diminished aldosterone release.
  • Previous DBH inhibitors, such as fusaric acid and SKF-102698 have drawbacks such as low potency and specificity, that have precluded their clinical development in heart failure.
  • Nepicastat was used in in vivo biochemical studies to study the effects in spontaneously hypertensive rats (SHRs) and normal beagle dogs.
  • SHRs spontaneously hypertensive rats
  • the animals were weighed and randomly assigned to receive either placebo (vehicle) or the appropriate dose of nepicastat.
  • Each rat was dosed orally three times, 12 h apart, beginning in the morning.
  • the rats were anesthetized with halothane, decapitated, and the tissues (cerebral cortex, mesenteric artery and left ventricle) were rapidly harvested, weighed, placed in iced 0.4 M perchloric acid, frozen in liquid nitrogen and stored at ⁇ 70° C. until analysis.
  • Tissue NE and DA concentrations were assayed by HPLC using electrochemical detection.
  • Male beagle dogs (10-16 kg, Marshall Farms USA Inc, North Rose, N.Y.) were used in the study. On the day of the study, dogs were randomly assigned to receive either placebo (empty capsule) or the appropriate dose of nepicastat. Each dog was dosed twice a day for 4.5 days. 6 h after the first dose on day 5, the dogs were euthanized with pentobarbital and the tissues (cerebral cortex. renal artery, left ventricle) harvested, weighed, placed in iced 0.4 M perchloric acid, frozen in liquid nitrogen and stored at ⁇ 70° C. until analysis. Tissue NE and DA concentrations were assayed by HPLC using electrochemical detection.
  • Oral administration of nepicastat produced dose-dependent increases in DA/NE ratios in the artery (mesenteric or renal), left ventricle and cerebral cortex in SHRs and dogs.
  • SKF-102698 (1) increased the DA/NE ratio by 5.5-fold, 3.5-fold and 2.7-fold, whereas nepicastat, at the same dose, increased the ratio by 8.3, 7.5 and 1.5 fold in the mesenteric artery, left ventricle and cerebral cortex, respectively.
  • nepicastat produces the expected biochemical effects in both SHRs and dogs but is more potent in the latter species. Furthermore, nepicastat is more potent than its Compound B and SKF-102698 (1) in SHRs.
  • nepicastat (14.5 day treatment) on the plasma DA/NE ratio were investigated in normal dogs. Animals were randomized to receive, orally, either placebo (empty capsule) or nepicastat (2 mg/kg, b.i.d) for 14.5 days. Daily blood samples were drawn, 6 h after the first dose, for the measurement of plasma concentrations of DA and NE. The samples were collected in tubes containing heparin and glutathione, centrifuged at ⁇ 4° C. and stored at ⁇ 70° C. until analysis.
  • nepicastat (2 mg/kg; b.i.d) produced a significant increase in the DA/NE ratio that attained its peak effect at approximately 6-7 days, then plateaued to a new steady-state between 7-14 days.
  • nepicastat The in vivo hemodynamic activity of nepicastat was further assessed in conscious, restrained SHRs, a model having high sympathetic drive to cardiovascular tissues. Hemodynamic study in SHRs. Male SHRs (15-16 week old) were used in the study. The animals were lightly anesthetized with ether and the left femoral artery and vein were catheterized for measurement of blood pressure and drug administration, respectively. The animals were placed in restrainers and allowed to recover for 30-40 min. After obtaining baseline measurements, the animals were treated, orally, with either vehicle or the appropriate dose of nepicastat and hemodynamic parameters were continuously recorded for 4 h.
  • the animals were then anesthetized with pentobarbital, placed on a heating pad (37° C.) and ventilated with a Harvard rodent ventilator. After administration of atropine (1 mg/kg, iv) and tubocurarine (1 mg/kg, iv), the animals were pithed through the orbit of the eye with a stainless steel rod. The pithing rod was stimulated electrically with 1 ms pulses of 80V at different frequencies (0. 15, 0.45, 1.5, 5, 15 Hz) to obtain frequency -pressor response curves.
  • Nepicastat produced significant (p ⁇ 0.05) lowering of mean arterial pressure at all doses and time points, except at 0.3 mg/kg (180, 210 and 240 min) and 1 mg/kg (30, 210 and 240 min).
  • the frequency-pressor response curve was shifted significantly (p ⁇ 0.05) to the right in a dose-dependent manner (maximum shift of ⁇ 5 fold in the frequency-response curve).
  • the heart rate response to PNS was not significantly affected.
  • Methimazole (1 mg/kg, po, b.i.d.), used as a positive control, caused a significant reduction in serum levels of T 3 (day 3, 31%, p ⁇ 0.05; days 7 and 9, 42% and 44%, p ⁇ 0.01) and T 4 (days 3 and 7, 46% and 58%, p ⁇ 0.01) 4 h post-dose, whereas nepicastat showed no significant effects throughout the study (days 3, 7 and 9). Both doses of nepicastat significantly raised the DAJNE ratio in the mesenteric artery (p ⁇ 0.01 relative to vehicle controls) but not in the cortex 4 h after the final dose on day 10.
  • nepicastat is a potent, selective and orally active inhibitor of DBH.
  • the compound is also devoid of significant behavioral effects in animal models and these findings will be the subject of a future publication.
  • Concentrations of dopamine and norepinephrine were determined in 942 samples of plasma collected from congestive heart failure (CHF) patients.
  • the objectives of the study were:
  • nepicastat 1. to evaluate the effects of various doses of nepicastat on transmyocardial (arterial-coronary sinus) and coronary sinus catecholamine levels after four weeks, and to evaluate the safety and tolerability of nepicastat over 12 weeks.
  • Hemodynamic parameters including cardiac output, systemic vascular resistance, MVO 2 , pulmonary artery pressures, and pulmonary artery wedge pressure after four weeks
  • Samples of blood were collected from patients from a peripheral vein, whilst they were supine, at 2 hours post-dose during weeks 4 and 12. Further samples from supine patients were collected on day 0 (i.e. the day prior to the start of dosing) at a time corresponding to 2 hours post-dose.
  • a group of patients underwent right heart and coronary sinus catheterization during week 4 at 2 hours post-dose and on day 0 (i.e. the day prior to the start of dosing) at a time corresponding to 2 hours post-dose.
  • Triplicate samples of blood were collected from the arterial vein and coronary sinus of these patients.
  • Concentrations of the free base of dopamine and norepinephrine were determined by a radioenzymatic method.
  • the method involves the incubation of the plasma samples with catechol-O-methyl transferase and tritiated S-adenosyl methionine.
  • the ⁇ -methylated catecholamines are extracted from the plasma by liquid/liquid extraction and then separated by thin layer chromatography.
  • the relevant bands for each catecholamine are marked and then scraped into scintillation vials for counting.
  • the quantitation limit of the method is 1 ⁇ g of dopamine or norepinephrine per mL of plasma.
  • the linear range is 1 to 333000 ⁇ g of dopamine or norepinephrine per mL of plasma using aliquots of 0.045 mL to 1 mL.
  • a pooled human plasma sample was used as the Quality Control sample (QC) and was analyzed in singlicate each day during routine use of the method to monitor the performance of the method.
  • QC Quality Control sample
  • Nepicastat was a potent inhibitor of both bovine and human DBH.
  • the IC 50 for nepicastat on human DBH was 9 nM (CL 6960), significantly lower than that for the DBH inhibitor SKF-102698 (85 nM).
  • the S enantiomer nepicastat was more potent than the R enantiomer (18 nM), denoted as Compound B.
  • nepicastat The binding affinity for nepicastat was screened at selected receptors. Nepicastat showed a binding affinity of less than 5.0 for M1, D1 and D2, and 5HT 1A, 2A and 2 C . The N-acetyl metabolite of nepicastat in rats and monkeys, showed a similar lack of binding affinity for these receptors. Thus nepicastat and its primary metabolite were not potent inhibitors for the receptors listed above.
  • SHRs spontaneously hypertensive rats
  • Wistar-Kyoto rats The aortic contractile response in vitro to phenylephrine is impaired in spontaneously hypertensive rats (SHRs) relative to normotensive Wistar-Kyoto rats.
  • SHRs spontaneously hypertensive rats
  • nepicastat 10 mg/kg, p.o.
  • nepicastat was an effective inhibitor of DBH in rats and dogs. Oral or intravenous administration resulted in a significant (p ⁇ 0.05) decrease in tissue norepinephrine, an increase in dopamine, and an increase in the dopamine/norepinephrine levels in the heart, mesenteric or renal artery, and the cerebral cortex in both species.
  • nepicastat significantly decreased norepinephrine and increased dopamine and the dopamine/norepinephrine ratio in the mesenteric artery from 0.5 to 4 hours following oral or i.v. administration at 6.2 mg/kg. Significant changes in these parameters were also observed in the left ventricle of male Sprague-Dawley rats 6 hours after the second of two i.v. injections (15 mg/kg) given 12 hours apart. The 24 hour time course of tissue catecholamines was studied in male SHRs following oral administration of either 10 or 30 mg/kg, respectively.
  • the increase in the dopamine/norepinephrine ratio was significant at 1 hour, and was long lasting (12 hours at 10 mg/kg, mesenteric artery, and 24 hours at 30 mg/kg, left ventricle).
  • Significant changes in mesenteric artery dopamine and norepinephrine levels were observed following 10 days of dosing to male Sprague-Dawley rats at 2.0 and 6.2 mg/kg p.o. b.i.d., with no significant effects observed in the cerebral cortex.
  • SHRs dosed at 1 or 10 mg/kg/d p.o. for either 7 or 25 days had significant increases in dopamine and the dopamine/norepinephrine ratio in the mesenteric artery and cerebral cortex.
  • nepicastat resulted in a significant decrease in norepinephrine and an elevation in dopamine and the dopamine/norepinephrine ratio in the mesenteric artery in rats with either acute or chronic (up to 25 days) dosing.
  • nepicastat in male SHRs and Sprague-Dawley rats were found to be dose responsive when assessed 6 hours following a single oral dose at 0.3, 1, 3, 10, 30, and 100 mg/kg.
  • SHRs there were significant changes in the dopamine/norepinephrine ratio in the mesenteric artery at doses of 0.3 mg/kg, in the left ventricle at 3.0 mg/kg, and in the cerebral cortex at 10 mg/kg.
  • Sprague-Dawley rats there were significant increases in the dopamine/norepinephrine ratio in the mesenteric artery at 3.0 mg/kg, in the left ventricle at 1.0 mg/kg, and in the cerebral cortex only at 100 mg/kg.
  • Nepicastat caused a significant dose dependent decrease in norepinephrine (10 mg/kg) and increase in dopamine (3.0 mg/kg) and the dopamine/norepinephrine ratio (3.0 mg/kg) in the left ventricle and mesenteric artery.
  • the effects of nepicastat on dopamine and norepinephrine concentrations, and the dopamine/norepinephrine ratio in the cerebral cortex were significant only at 30 and 100 mg/kg.
  • nepicastat was less potent in inhibiting DBH in the cerebral cortex of rats (60-100 mg/kg/d) than in the left ventricle and mesenteric artery (1-6 mg/kg/d).
  • Nepicastat (the S enantiomer) was significantly more potent then the R enantiomer in the left ventricle and mesenteric artery in SHRs after three doses given 12 hours apart (30 mg/kg p.o.).
  • nepicastat was significantly more potent than the DBH inhibitor SKF-102698 in decreasing norepinephrine and increasing dopamine and the dopamine/norepinephrine ratio in the left ventricle and mesenteric artery in SHRs after a single dose, or three doses at 30 mg/kg.
  • the potency relationships in the left ventricle and mesenteric artery resulting from these in vivo studies strongly parallel those obtained from in vitro studies using purified DBH (see above).
  • nepicastat had significantly less effects than SKF-102698 in decreasing norepinephrine levels and increasing dopamine levels in the cerebral cortex. Norepinephrine has been shown to stimulate the release of renin and increase plasma renin activity. It was therefore of interest to assess whether decreasing norepinephrine levels with nepicastat would result in a decrease in plasma renin activity. However, nepicastat (30 and 100 mg/kg/d p.o. for 5 days) did not alter plasma renin activity in male SHRs. Thus, nepicastat, when given at doses that lower tissue norepinephrine levels, does not alter plasma renin activity in SHRs.
  • Nepicastat caused a significant decrease in norepinephrine levels and an increase in the dopamine/norepinephrine ratio, but did not alter dopamine levels, in the mesenteric artery from male beagle dogs 5 hours after administration of 30 mg/kg intraduodenally.
  • nepicastat was a potent, orally active inhibitor of DBH in dogs at doses of at least 10 mg/kg/d.
  • Nepicastat has structural similarities to methimazole, a potent inhibitor of thyroid peroxidase in vivo. nepicastat at doses of 4 or 12.4 mg/kg/d, p.o. had no effect on serum levels of triiodothyramine or thyroxine in male Sprague-Dawley rats fed a low iodine diet and dosed for 10 days, while methimazole (2 mg/kg/d) significantly reduced serum levels of triiodothyramine or thyroxine. Thus, nepicastat, unlike methimazole, did not affect serum levels of triiodothyramine or thyroxine.
  • Nepicastat induced a significant antihypertensive effect for up to 4 hours in conscious, restrained SHRs (1.0-30 mg/kg, p.o.), and significantly reduced heart rate (10 and 30 mg/kg).
  • the antihypertensive effects of nepicastat in conscious, restrained SHRs (10 mg/kg, p.o.) were not attenuated by pretreatment with the dopamine receptor (DA-1) antagonist SCH-23390.
  • nepicastat (10 mg/kg) also reduced blood pressure 4 hours after dosing in conscious, restrained normotensive Wistar-Kyoto rats; however, the decrease in pressure was less ( ⁇ 13 mmHg) than with SHRs ( ⁇ 46 mmHg).
  • nepicastat causes a decrease in blood pressure in both SHRs and normotensive rats, though the antihypertensive effect is more pronounced in SHRs.
  • the antihypertensive effects in SHRs do not appear to be mediated via DA-1 receptors.
  • Nepicastat also significantly attenuated the hypertensive and tachycardic responses to preganglionic nerve stimulation in pithed SHRs 5 hours after dosing (3 mg/kg p.o.). Thus, nepicastat reduces the rise in blood pressure in response to sympathetic nerve stimulation.
  • Acute intravenous treatment of anesthetized SHRs with nepicastat (3.0 mg/kg, i.v.) decreased mean arterial pressure over a 3 hour period, but did not lower renal blood flow or alter urine production or urinary excretion of sodium or potassium.
  • the calculated renal vascular resistance was decreased following dosing.
  • nepicastat did not impair renal function in anesthetized SHRs, and did not decrease renal blood flow despite causing a decrease in arterial blood pressure.
  • nepicastat 1 and 10 mg/kg, p.o.
  • SHRs systolic blood pressure as measured by the tail cuff method.
  • nepicastat(10 mg/kg, p.o.) induced a significant antihypertensive effect when the rats were restrained and their blood pressure measured directly via an arterial cannulae.
  • 30 days of treatment of SHRs with nepicastat at 30 and 100 mg/kg/d resulted in a decrease in blood pressure and, when combined with enalapril, additional blood pressure decreases along with a reduction in left ventricular mass.
  • nepicastat had a greater blood pressure lowering effect in SHRs than in normotensive rats.
  • Nepicastat (30 mg/kg intraduodenally) did not significantly inhibit either the decrease in renal blood flow in response to direct renal nerve stimulation, or the increase in arterial blood pressure in response to carotid artery occlusion up to 5 hours after dosing in anesthetized male beagle dogs.
  • nepicastat caused a significant decrease in norepinephrine levels and an increase in the dopamine/norepinephrine ratio, but not dopamine levels, in the mesenteric artery 5 hours after dosing.
  • tissue norepinephrine levels were significantly reduced, there was no significant inhibition of sympathetically-evoked functional responses.
  • nepicastat When nepicastat was given to male beagle dogs for 4.5 days at 10 mg/kg/d there was no statistically significant decrease in the degree of blood pressure and heart rate increases in response to carotid artery occlusion in anesthetized animals. Nepicastat treatment significantly reduced the increase in heart rate in response to an i.v. tyramine challenge, but produced only slight and non-significant inhibition of blood pressure increases. Thus, chronic dosing with nepicastat at a dose that has been shown to result in a maximal decrease in tissue norepinephrine levels, does not have a major inhibitory effect on sympathetically-evoked functional responses.
  • Nepicastat caused no significant effects on gross motor behavior in mice following acute dosing at 1.0-30 mg/kg, p.o., and it did not effect locomotor activity in mice (10-100 mg/kg i.p.). Acute administration to rats did not effect locomotor activity or acoustic startle reactivity (3-100 mg/kg i.p.).
  • Nepicastat is a potent competitive inhibitor of human DBH in vitro, and in rats and dogs in vivo.
  • oral treatment with nepicastat resulted in significant evidence for DBH inhibition in the heart and mesenteric artery at a dose 6 mg/kg/d.
  • nepicastat showed some selectivity to the left ventricle and mesenteric artery relative to the cerebral cortex. No behavioral effects were observed with nepicastat in rats.
  • a plateau effect for DBH inhibition occurred at 10 mg/kg/d in the heart, renal artery and kidney.
  • Nepicastat significantly reduced the hypertensive response to sympathetic nerve stimulation in rats (3 mg/kg p.o.), and it significantly lowered blood pressure throughout the day when dosed once daily (30 mg/kg/d p.o.) for 30 days in SHRs.
  • nepicastat is a potent DBH inhibitor that modulates the action of the sympathetic nervous system.
  • the studies described here were designed to evaluate the pharmacokinetics of higher oral doses of nepicastat, to compare the pharmacokinetics in male and female rats, and to determine penetration of nepicastat into the CNS by quantitating levels of nepicastat in brain.
  • samples of blood were obtained by cardiac puncture with heparinized syringes, and plasma was prepared by centrifugation. Brains of rats were surgically excised, and all samples were frozen at ⁇ 20° C. until analysis.
  • the LC system used a Keystone Hypersil BDS 15 cm C 8 column at ambient temperature.
  • Mobile phase A was 12.5 mM potassium phosphate, pH 3.0, with 5 mM dodecanesulfonic acid and mobile phase B was acetonitrile.
  • Solvent composition was 40% B and was pumped at a flow rate of 1 ml/min. Detection was by UV absorption at 261 nm. Concentrations of analytes were determined from a standard curve generated from the analysis of plasma from untreated rats fortified with known concentrations of analyte. Plasma concentration data are expressed as ⁇ g (free base) per ml.
  • Brains were rinsed briefly with saline, blotted on a paper towel, then weighed (1.5-2.0 g). Internal standard was added (50 ⁇ l of methanol containing 20 ⁇ g/ml a monofluro analog of nepicastat), and brains were homogenized in 5 ml of 200 mM sodium phosphate, pH 7.0, containing 0.5 mg/ml dithiothreitol. Aliquots of homogenate (2 ml) were extracted with 10 ml of ethyl acetate/hexane (1/1, v/v). The organic phase was gently back extracted with 150 ⁇ l of 250 mM acetic acid.
  • Plasma half-life T 1/2
  • is the elimination rate constant determined by linear regression of the log plasma concentration vs. time data within the terminal linear portion of the data.
  • Areas under the plasma concentration vs. time curve AUC from zero to the time of the last quantifiable plasma concentrations were calculated by the trapezoidal rule.
  • AUC from zero to infinity AUC total was calculated as:
  • AUC total AUC (0 ⁇ C last )+C last / ⁇ where C last is the last quantifiable plasma concentration.
  • Concentrations of nepicastat in plasma of male rats given 10, 30, or 100 mg/kg single oral doses were obtained. Concentrations of nepicastat in plasma increased with increasing dose, and the relationship between AUC total and dose was linear. The elimination half-life appeared to increase slightly at higher doses (1.70, 2.09, and 3.88 hr following the 10, 30, and 100 mg/kg oral doses to male rats, respectively). Following a 30 mg/kg oral dose of nepicastat to female rats, the plasma AUC total of nepicastat was 77% higher in female rats than in male rats given an equivalent dose of nepicastat.
  • nepicastat in brain were initially lower than those in plasma (expressed as ⁇ g/ml). From 2 hr following dosing onward, however, concentrations of nepicastat in brain exceeded those in plasma.
  • Plasma levels of nepicastat in male rats increased linearly with increasing doses between 10 and 100 mg/kg, based on values of AUC total .
  • Plasma levels of nepicastat were higher in female rats than in male rats following a 30 mg/kg oral dose.
  • nepicastat in brain were initially lower than those in plasma, but from 2 hr onward, levels of nepicastat in brain were greater than in plasma.
  • nepicastat 10 mg/kg
  • dopamine and norepinephrine levels in the mesenteric artery following a single oral dose in spontaneously hypertensive rats.
  • Catecholamine levels were measured at 1, 2, 4, 6, 8, 12, 16, and 24 hours after a single oral administration of either nepicastat (10 mg/kg) or vehicle (dH 2 O; 10 ml/kg).
  • SHRS spontaneous hypertensive rats
  • Nepicastat was synthesized as the hydrochloride salt and nepicastat was dissolved in vehicle (dH 2 O) to yield an oral dose that could be administered in repeated volumes of 10 ml/kg. All doses of nepicastat were administered as free base equivalents and prepared the morning of administration.
  • Each treatment group was compared to vehicle at each time point.
  • a two way analysis of variance (ANOVA) with effects TRT, HARVEST and their interaction was performed.
  • a one way ANOVA with factor TRT was performed for each harvest time. Pairwise analyses between treated and vehicle animals, at each time point, were carried out using Fisher's LSD strategy to control the experiment-wise error rate. Norepinephrine values were significantly (p ⁇ 0.05) lower than vehicle only at the 4 hr time point. Levels were marginally (0.05 ⁇ p ⁇ 0.1) lower at the 6 hour time point. Dopamine levels were significantly (p ⁇ 0.05) higher than those of vehicle at the 2 and 6 hr harvest times. The dopamine/norepinephrine ratio was significantly (p ⁇ 0.05) greater than those of vehicle treated animals at the 1, 2, 4, 6 and 12 hour time points.
  • nepicastat had few statistically significant effects on mesenteric artery norepinephrine or dopamine levels following a single oral administration at 10 mg/kg in spontaneously hypertensive rats at 1, 2, 4, 6, 8, 12, 16 or 24 hours following dosing.
  • a consistent increase in the dopamine/norepinephrine ratios were observed across most of the first 12 hours of treatment.
  • no changes in any of the three parameters were observed.
  • nepicastat nepicastat
  • Animals received two intravenous (iv) administrations, 12 hours apart, of either vehicle (75% propylene glycol+25% DMSO; 1.0 ml/kg) or 15 mg/kg of nepicastat. Tissue norepinephrine and dopamine levels were measured six hours after the last compound administration.
  • Nepicastat was synthesized and was dissolved in the appropriate amount of vehicle (75% propylene glycol+25% DMSO) to obtain a dosing volume of 1.0 ml/kg. Nepicastat was administered as the free base equivalent and prepared the afternoon prior to the first administration.
  • Each rat was dosed iv in the tail vein the afternoon before harvest. The dosing was repeated 12 hours later the following morning. Six hours after the final administration rats were anesthetized with halothane, decapitated, and the left ventricle was rapidly harvested and weighed. The ventricle was placed in 1.0 ml iced 0.4 M perchloric acid. Tissues were immediately frozen in liquid nitrogen and stored at ⁇ 70° C. Tissue dopamine and norepinephrine concentrations were assayed by high performance liquid chromatography using electrochemical detection.
  • a one-way analysis of variance (ANOVA) with a main effect for treatment was performed for norepinephrine.
  • a Kiruskal-Wallis was performed for dopamine and their ratio primarily due to heterogeneous variances among treatment groups.
  • Subsequent pairwise comparisons between nepicastat treated rats and vehicle were performed using Fisher's LSD test.
  • a Bonferroni adjustment was performed on all p-values to ensure an overall experiment-wise type 1 error rate of 5%.
  • Nepicastat was dissolved in the appropriate amount of vehicle (dH 2 O for nepicastat and PEG 400:dH 2 O, 50:50 vol:vol for SKF-102698. Doses of 3, 10, 30, and 100 mg/kg of nepicastat, and 30 mg/kg SKF-102698 were prepared in 10.0 ml/kg dosing volumes.
  • SHRs spontaneously hypertensive rats
  • Animals were weighed and randomly assigned to one of the following treatment groups: 1) distilled water vehicle (dH 2 O), or nepicastat at 3, 10, 30, and 100 mg/kg, 2) Compound B at 30 mg/kg in distilled water, or 3) PEG 400:dH 2 O vehicle or SKF-102698 at 30 mg/kg.
  • Each rat was dosed orally (p.o., using a gavage needle) three times 12 hours apart, beginning in the morning.
  • rats were anesthetized with halothane, decapitated, and the cortex, mesenteric artery, and left ventricle were rapidly harvested, weighed, placed in iced 0.4 M perchioric acid, frozen in liquid nitrogen, and stored at ⁇ 70° C. Tissue dopamine and norepinephrine concentrations were assayed by high performance liquid chromatography and electrochemical detection.
  • the first series compared the rats treated with various doses of nepicastat, and Compound B at 30 mg/kg to the vehicle control animals.
  • a nonparametric one-way analysis of variance (ANOVA) with factor Dose and blocking factor Day was performed for each tissue and strain separately. Overall results are reported. Pairwise analysis between treated and controls at each dose were carried out using Dunnett's test to control the experiment-wise error rate.
  • the second statistical test compared SKF-102698 to the PEG-dH 2 O vehicle treated group using a nonparametric t-test.
  • the third statistical test compared Compound B to nepicastat at doses of 30 mg/kg using a nonparametric t-test.
  • a fourth statistical analysis compared nepicastat to SKF-102698 at doses of 30 mg/kg. Since two different vehicles were used, a linear contrast was developed which calculates the difference of differences as follows:
  • the dopamine concentration in the cerebral cortex was significantly (p ⁇ 0.05) greater, the norepinephrine concentration was significantly (p ⁇ 0.05) lower), and the dopamine/norepinephrine ratios significantly (p ⁇ 0.05) greater than vehicle at doses of 30 and 100 mg/kg of nepicastat.
  • Dopamine concentration in the left ventricle was significantly (p ⁇ 0.05) greater than vehicle at doses of 3, 10, 30 and 100 mg/kg.
  • Norepinephrine concentration was significantly (p ⁇ 0.05) lower than vehicle at doses of 10, 30 and 100 mg/kg.
  • the dopamine/norepinephrine ratio in the left ventricle was significantly (p ⁇ 0.05) greater than vehicle at doses of 3, 10, 30, and 100 mg/kg of nepicastat.
  • Dopamine concentration in the mesenteric artery of SHRs was significantly (p ⁇ 0.05) greater than vehicle at doses of 3, 10, 30 and 100 mg/kg.
  • Norepinephrine concentration was not significantly less (p>0.05) than vehicle at 10, 30, and 100 mg/kg.
  • the dopamine/norepinephrine ratios in the mesenteric artery were significantly (p ⁇ 0.05) greater than vehicle at all doses of nepicastat.
  • Compound B In the cerebral cortex, relative to treatment with vehicle, Compound B resulted in significant increase in both dopamine and norepinephrine (p ⁇ 0.01), and had no effect on the dopamine/norepinephrine ratio. Norepinephrine levels were significantly lower with nepicastat compared to Compound B (p ⁇ 0.01).
  • Compound B resulted in a significant increase in dopamine and the dopamine/norepinephrine ratio (p ⁇ 0.01), but did not significantly lower norepinephrine levels.
  • Nepicastat was significantly more effective (p ⁇ 0.01) than Compound B at lowering norepinephrine levels, and increasing dopamine and the dopamine/norepinephrine ratio.
  • Nepicastat was significantly more effective (p ⁇ 0.01) than Compound B at lowering norepinephrine levels, and increasing dopamine and the dopamine/norepinephrine ratio.
  • nepicastat Comparing nepicastat with SKF-102698 at 30 mg/kg in the cerebral cortex, dopamine concentration in the cortex was significantly greater (p ⁇ 0.01) than vehicle for SKF-102698 at a dose of 30 mg/kg. The increase above vehicle was greater for SKF-102698 than for nepicastat (p ⁇ 0.01). Norepinephrine concentration was significantly lower than vehicle for SKF-102698, and the decrease was greater for SKF-102698 than for nepicastat (p ⁇ 0.01).
  • the dopamine/norepinephrine ratios in the cortex were significantly (p ⁇ 0.01) greater than vehicle for SKF-102698, and the increase above vehicle was greater for SKF-102698 than for nepicastat (p ⁇ 0 01).
  • the dopamine concentration in the left ventricle was significantly greater (p ⁇ 0.01) than vehicle for SK-F102698, and the increase above vehicle was greater for nepicastat than for SKF-102698 (p ⁇ 0.01).
  • Norepinephrine concentration was not different from vehicle with SKF-102698 treatment, however treatment with nepicastat significantly lowered norepinephrine relative to vehicle more than SKF-102698 (p ⁇ 0.01).
  • the dopamine/norepinephrine ratios in the left ventricle were significantly (p ⁇ 0.05) greater than vehicle for SKF-102698, and the increase above vehicle was greater for nepicastat than for SKF-102698 (p ⁇ 0.05).
  • the dopamine concentration in the mesenteric artery was significantly greater than vehicle for SKF-102698, and the increase above vehicle was greater for NEPICASTAT than for SKF-102698.
  • Norepinephrine concentration was significantly lower than vehicle with SKF-102698 treatment, and treatment with nepicastat significantly lowered norepinephrine relative to vehicle more than SKF-102698.
  • the dopamine/norepinephrine ratios in the left ventricle were significantly greater than vehicle than for SKF-102698, and the increase above vehicle was greater for nepicastat than for SKF-102698.
  • nepicastat is a potent inhibitor of DBH in vivo in the mesenteric artery, left ventricle, and cerebral cortex of SHRs six hours after the third of three oral doses administered 12 hours apart.
  • the S enantiomer, nepicastat was more potent than the R enantiomer (Compound B) in all three tissues at 30 mg/kg.
  • nepicastat was more effective than SKF-102698 in the mesenteric artery and left ventricle, but less effective in the cerebral cortex, following three doses at 30 mg/kg administered over 24 hours.
  • Nepicastat was prepared and administered as the free base equivalent. Nepicastat and methimazole were dissolved in vehicle (66.7% propylene glycol:33.3% dH2O) to yield dosing solutions of appropriate concentrations so that all doses could be administered in a 1.0 ml/kg volume.
  • iodine deficient diet Purina, 5891C, Lot 1478, 0.066 ⁇ 0.042 mg iodine/kg sample
  • rats were anesthetized with halothane, decapitated, and the cortex, striatum, and mesenteric artery were harvested and weighed. Tissue samples were not harvested from the methimazole groups as they only served as positive controls for determination of thyroid function.
  • the mesenteric artery, cortex, and striatum were immediately placed in 0.4 M iced perchloric acid and analyzed for norepinephrine and dopamine levels the same day using HPLC.
  • Orbital blood samples were taken at day ⁇ 3, 0, 3, 7, and 9 (day 0 was the first day of dosing). Serum samples were analyzed for T 3 and T 4 levels using a radioimmunoassay.
  • Norepinephrine levels in the nepicastat treated animals were not significantly (p>0.05) different in the cortex compared to vehicle control at doses of 2.0 and 6.2 mg/kg.
  • Norepinephrine levels in the mesenteric artery were significantly (p ⁇ 0.05) lower at the 2.0 and 6.2 mg/kg dose groups, and norepinephrine levels in the striatum were marginally (p ⁇ O.1O) lower in both the 2.0 and 6.2 mg/kg dose groups, compared to vehicle control.
  • Dopamine levels in all three tissues were not significantly (p>0.05) different from vehicle control at either the 2.0 or 6.2 mg/kg dose group of nepicastat.
  • the dopamine/norepinephrine ratio of the cortex and striatum at 2.0 and 6.2 mg/kg nepicastat were not significantly (p>0.05) different from vehicle control, while the ratio of the mesenteric artery at both 2.0 and 6.2 mg/kg nepicastat were significantly (p ⁇ 0.05) higher than vehicle control.
  • nepicastat affected thyroid function by altering free T 3 or total T 4 levels in the rat serum.
  • T 4 levels of the methimazole treated animals were only marginally (p ⁇ 0.10) lower on day nine.
  • Nepicastat (2.0 or 6.2 mg/kg) did not cause any significant (p>0.05) changes in dopamine or the norepinephrine levels, or dopamine/norepinephrine ratio when compared to vehicle.
  • a marginally significant (p ⁇ 0.10) decrease in norepinephrine level was observed in the 6.2 mg/kg dose group, but no other significant changes were observed.
  • both 2.0 and 6.2 mg/kg of nepicastat produced significantly (p ⁇ 0.05) lower norepinephrine levels and significantly (p ⁇ 0.05) higher dopamine/norepinephrine ratios, compared to vehicle, with no significant changes observed in dopamine levels.
  • nepicastat appears to be an effective inhibitor of dopamine ⁇ -hydroxylase in vivo, with greater effect in the mesenteric artery than the cerebral cortex or striatum following 10 days of dosing in Sprague-Dawley rats.
  • kidney medulla and kidney cortex were prepared from dogs dosed with nepicastat.
  • Adult male beagle dogs were randomly assigned to four groups of 8 dogs per group and dosed by oral administration with nepicastat.
  • Nepicastat was delivered in doses of 5, 15 and 30 mg/kg placed in single capsules. Vehicle was an empty capsule.
  • Each dog received 2 doses daily, morning and afternoon (8-10 hours apart) for four days. On the fifth day, each dog received a single dose in the morning and the dogs were euthanized six hours after the last dose.
  • Samples of kidney medulla and kidney cortex were rapidly harvested, weighed, placed in cold 0.4 M perchloric acid, frozen in liquid nitrogen and stored at ⁇ 70° C.
  • NE norepinephrine
  • D dopamine
  • Each analyte determination was normalized to the weight of the tissue sample and expressed as ⁇ g of analyte per gram of tissue.
  • concentrations of dopamine, norepinephrine and the ratio of dopamine concentration to norepinephrine concentration (D/NE) were obtained for each dog.
  • D/NE ratio of dopamine concentration to norepinephrine concentration
  • Tissue samples were also taken from the dogs at the end of the study in case it was deemed necessary to analyze tissue catecholamines at a later point.
  • a final blood sample (10 ml) was taken.
  • Dogs were anesthetized with sodium pentobarbital (40 mg/kg, iv), placed on a necropsy table and euthanized with a second injection of pentobarbital (80 mg/kg,iv).
  • a rapid bilateral transthoracotomy and abdominal incision was performed.
  • Biopsies were taken from the renal artery and left ventricle. The skull was opened to expose the frontal lobe of the cerebral cortex and a biopsy was taken.
  • Tissue samples were weighed, placed on iced 0.4 M perchloric acid, frozen in liquid nitrogen and stored at ⁇ 70° C. until analyzed.
  • Plasma norepinephrine (NE), dopamine (DA) and epinephrine (EPI) were analyzed by HPLC using electrochemical detection. Plasma concentration of nepicastat was determined by HPLC using electrochemical detection.
  • the Box-Cox transformations indicated that the logarithm was an appropriate variance stabilizing transformation; hence all analyses were performed on the log-values.
  • the BQL (below quantitation limit) in the DA concentration of dog 1 at day 10 was set to 0; In (0) was set to missing.
  • the analysis was performed using a mixed model (using PROC MIXED) with the day and treatment categorical variables being fixed and the dog within treatment being a random factor. For the fixed effects, the interaction between the day and the treatment was included, since the difference between the drug and placebo groups varies from day to day. Contrasts were calculated using the CONTRAST statement, which correctly takes into account the error terms for each particular contrast. In particular, the contrasts comparing the treatment group to the drug group uses the dog mean square for its error term, while the comparisons used to establish steady state are all within dog comparisons, and require the error mean square.
  • the time period of steady state was calculated using the Helmert transformation (cf. SAS PROC GLM manual). These transformations compare each treatment mean with the average of the treatment means of the time points following.
  • the steady state period is defined to start at the first time point following the maximum time at which the Helmert contrast is statistically significant.
  • the slope of the analyte concentration during the steady state period also was calculated.
  • the slope during the steady state period was calculated for each dog individually, yielding one slope per animal. Univariate statistics on the slopes were then calculated, with Normal theory confidence intervals built on the mean slope, and the hypothesis of slope equaling zero was tested, and its Normal theory p-value was calculated. This slope analysis was used as the basis for determining whether the steady state period was a period of changing concentration.
  • nepicastat (2 mg/kg, b i d) produced significant decreases in plasma NE (2.1 fold) and EPI (1.91 fold) and significant increases in plasma DA (7.5 fold) and DA/NE ratio (13.6 fold).
  • the peak decreases in plasma NE and EPI were observed at day 6 and day 8, respectively, whereas the peak increases in plasma DA and DA/NE ratio were observed at day 7 and day 6, respectively.
  • the effects on plasma NE, DA and EPI attained steady-state at approximately 4, 8 and 6 days post-dose, respectively.
  • the changes in plasma DA and DA/NE ratio were significantly different from placebo on all days post-dose.
  • the changes in plasma NE were significantly different from placebo on days 4-9 and days 11-13 post dose.
  • the changes in plasma EPI were significantly different from placebo on days 7-9 and day 12 post-dose.
  • nepicastat (2 mg/kg, bid) produced significant plasma levels of the drug on all days. The peak levels were observed at 2 days post-dose. No significant levels of the N-acetyl metabolite of nepicastat were detected on any of the days.
  • nepicastat (2 mg/kg, bid, po) produced significant decreases in plasma NE and EPI and significant increases in plasma DA and DA/NE ratio. These changes reflect inhibition of the sympatho-adrenal system via inhibition of the enzyme dopamine ⁇ -hydroxylase.
  • Nepicastat was weighed and put into capsules (size 13—Torpac; East Hanover, N.J.) to yield doses of 5, 15, and 30 mg/kg per capsule (given b.i.d. to yield doses of 10, 30 and 60 mg/kg/day). The initial dog weight was used to determine the dose for each animal. Dogs receiving 0 mg/kg/day received empty capsules (placebo). All doses of nepicastat were administered as free base equivalents.
  • nepicastat at 0 mg/kg/day (placebo), 10 mg/kg/day (5 mg/kg b.i.d.), 30 mg/kg (15 mg/kg b.i.d.), or 60 mg/kg/day (30 mg/kg b.i.d.).
  • Dog numbers 1-16 were assigned as dose group A and dog numbers 17-32 as dose group B.
  • the terminal surgery for tissue harvest was performed over 2 days with 16 animals studied per day. Two or 3 days before the first compound administration each dog was weighed and skin regions overlying both cephalic, saphenous and jugular veins were shaved.
  • Dosing consisted of oral administration of one capsule with the second given 8-10 hr later. Dogs were dosed as scheduled on days 1-3. On day 4, prior to the AM dose, 3 ml of blood were obtained from a jugular vein for determination of baseline plasma compound levels. The dog was then administered the AM dose, and at 1, 2, 4 and 8 hr following the dose additional 3 ml blood samples were collected for determination of plasma compound levels. Blood samples were put into tubes containing heparin, centrifuged at 4° C. and stored at ⁇ 20° C. until analysis. The PM dose was then administered as scheduled. The AM dose was administered as scheduled on the days of surgery.
  • a final 3 ml blood sample was taken from the jugular vein for determination of plasma compound levels.
  • the dog was then anesthetized with pentobarbital Na (40 mg/kg), given i.v. in a cephalic or saphenous vein, and delivered to the necropsy room where an additional dose of pentobarbital Na was given ( ⁇ 80 mg/kg, iv).
  • pentobarbital Na 40 mg/kg
  • the left ventricle, renal artery, kidney, renal medulla, renal cortex and cerebral cortex were then rapidly harvested, weighed, put into 2 ml iced 0.4M perchloric acid, frozen in liquid nitrogen and stored at ⁇ 70° C. until analysis for catecholamines by HPLC using electrochemical detection.
  • tissue samples were divided into 2 portions, the second of which were immediately frozen in liquid nitrogen and stored at ⁇ 70° C. for determination of tissue compound levels.
  • a third transmural sample taken from the left ventricle was immediately frozen in liquid nitrogen and stored at ⁇ 70° C. for use in receptor binding studies.
  • Ventricles were homogenized in 50 mM Tris-HCl, 5 mM Na 2 EDTA buffer (pH 7.4 at 4° C.) using a Polytron P-10 tissue disrupter (setting 10, 2 ⁇ 15 second bursts). Homogenates were centrifuged at 500 ⁇ g for 10 minutes and the supernatants stored on ice. The pellets were washed by resuspension and centrifugation at 500 ⁇ g and the supernatants combined. The combined supernatants were centrifuged at 48,000 ⁇ g for 20 minutes.
  • Receptor densities were expressed, per mg protein, as mean for each treatment group.
  • Tissue catecholamine levels were analyzed by comparing nepicastat-treated groups with the placebo (control) treated groups.
  • a nonparametric one-way analysis-of-variance (ANOVA) with factor DOSE was performed for each tissue and each catecholamine measure separately. Pairwise analyses between treated and controls at each dose were carried out using Dunnett's test to control the experiment-wise error rate. Student-Neuman-Kuels and Fisher's LSD tests were performed as validation. Analysis of tissue and plasma compound levels were performed in 2 ways. First, individual t-tests were run to compare each dose level to a factored level of its partner dose for each parameter.
  • Dogs were orally administered 0, 5, 15, or 30 mg/kg nepicastat capsules b.i.d. to yield doses of 10, 30, and 60 mg/kg/day for 4.5 days and tissue was harvested 6 hr after the final administration.
  • nepicastat administered at doses of 10, 30 and 60 mg/kg/day significantly (p ⁇ 0.01) decreased norepinephrine levels by 86%, 81% and 85%, respectively.
  • Dopamine levels were significantly (p ⁇ 0.01) increased at doses of 10, 30 and 60 mg/kg/day by 180%, 273% and 268%, respectively.
  • norepinephrine was significantly (p ⁇ 0.01) reduced by 63% and the ratio significantly (p ⁇ 0.01) elevated by 86%, while dopamine levels marginally (0.05 ⁇ p ⁇ 0. 10) increased 174%, compared to placebo.
  • norepinephrine levels were significantly (p ⁇ 0.01) decreased by 85%, 58% and 79%, respectively in the left ventricle.
  • the dopamine/norepinephrine ratio significantly (p ⁇ 0.01) increased 852%, 279% and 607%, respectively, compared to placebo animals. No significant changes were observed in dopamine levels at doses of 10, 30, and 60 mg/kg/day nepicastat.
  • norepinephrine levels were significantly decreased (p ⁇ 0.01) by 86%, 66% and 85%, respectively, following doses of 10, 30 and 60 mg/kg/day nepicastat.
  • Dopamine levels were significantly (p ⁇ 0.01) increased 156%, 502% and 208%, respectively, at these doses.
  • the dopamine/norepinephrine ratio significantly (p ⁇ 0.01) increased by 1653%, 1440% and 1693%, respectively, at doses of 10, 30, and 60 mg/kg/day.
  • the dopamine/norepinephrine ratios were significantly (p ⁇ 0.01) increased by 555%, 636% and 677%, respectively, at doses of 10, 30 and 60 mg/kg/day nepicastat, compared to placebo.
  • Dopamine levels were significantly (p ⁇ 0.01) increased 522% at 30 mg/kg/day and marginally (0.05 ⁇ p ⁇ 0.10) increased by 150% and 156%, respectively, at 10 and 60 mg/kg/day.
  • Norepinephrine levels were significantly (p ⁇ 0.01) decreased 72% following administration of 10 mg/kg/day nepicastat, compared to placebo, and marginally (0.05 ⁇ p ⁇ 0. 10) decreased by 69% following 60 mg/kg/day.
  • Nepicastat was evaluated for its activity at a range of enzymes including tyrosine hydroxylase, NO synthase, phosphodiesterase III, phospholipase A 2 , neutral endopeptidase, Ca 2+ /calmodulin protein kinase II, acetyl CoA synthetase, acyl CoA-cholesterol acyl transferase, HMG-CoA reductase, protein kinase (non-selective) and cyclooxygenase-I. As shown in FIG. 4 , nepicastat had an IC 50 of >10 ⁇ M at all the 12 enzymes studied, and therefore it is a highly selective (>1000-fold) inhibitor of dopamine- ⁇ -hydroxylase.
  • Bovine DBH from adrenal glands was obtained from Sigma Chemicals (St. Louis, Mo.). Human secretory DBH was purified from the culture medium of the neuroblastoma cell line SK—N—SH and was used to obtain the inhibition data.
  • a lentil lectin-sepharose column containing 25 ml gel was prepared and equilibrated with 50 mM KH 2 PO 4 , pH 6.5, 0.5 M NaCl. The column was eluted with 35 ml of 10% methyl ⁇ , D-mannopyranoside in 50 mM KH 2 PO 4 , pH 6.5, 0.5 M NaCl at 0.5 ml/min.
  • An HPLC assay was used to measure DBH activity using tyramine and ascorbate as substrates. The method is based on the separation and quantitation of tyramine and octopamine by reverse phase HPLC chromatography (Feilchenfeld, N. B., Richter, H. & Waddell, W. H. (1982). Anal. Biochem: A time-resolved assay of dopamine ⁇ -hydroxylase activity utilizating high-pressure liquid chromatography. 122: 124-128.). The assay was performed at pH 5.2 and 37° C.
  • the samples (150 ⁇ l) were loaded to a Gilson autosampler and analyzed by HPLC using UV detection at 280 nm.
  • PC-1000 software (Thermo Separations products, Fremont, Calif.) was used for integration and data analysis.
  • the HPLC run was carried out at the flow rate of 1 ml/min using a LiChroCART 125-4 RP-18 column and isocratic elution with 10 mM acidic acid, 10 mM 1-heptanesulfonic acid, 12 mM tetrabutylammonium phosphate, and 10% methanol.
  • the remaining percent activity was calculated based on the control without inhibitor, corrected using internal standards and fitted to a nonlinear 4 parameter dose response curve to obtain the IC 50 values.
  • [ 14 C]Tyramine hydrochloride was purified by a C18 light load column (two columns combined into one) that was washed with 2 ml of MeOH, 2 ml of 50 mM KH 2 PO 4 , pH 2.3, 30% acetonitrile, and then 4 ml of 50 mM KH 2 PO 4 , pH 2.3.
  • a vacuum manifold (Speed Mate 30, from Applied Separations) was used to wash and elute the column by vacuum.
  • Enzyme Assay by Radioactive Method Enzymatic activity was assayed using [ 14 C]tyramine as substrate and a C18 column to separate the product. The assay was performed in 200 ml volume containing 100 mM NaAc, pH 5.2, 10 mM fumaric acid, 0.5 82 M CuSO 4 , 4 mM ascorbic acid, 0.1 mg/ml catalase and various concentrations of tyramine. The total counts of each reaction was ⁇ 150,000 cpm. Bovine DBH (0.18 ng for each reaction) was mixed with tyramine and inhibitor in the reaction buffer at 37° C. The reaction was initiated by the addition of ascorbate/catalase mixture and was incubated at 37° C. for 30 minutes.
  • the reaction was stopped by the addition of 100 ml of 25 mM EDTA, 50 mM KH 2 PO 4 , pH 2.3. Entire mixture was loaded to a C18 light load column (two combined into one) that was pre-washed with MeOH and equilibrated with 50 mM KH 2 PO 4 , pH 2.3. Elution into scintillation vials was carried out with 1 ml of KH 2 PO 4 , pH 2.3 buffer twice, followed by 2 ml of the same buffer. ReadySafe scintillation fluid (16 ml) was added to the scintillation vials and the samples were counted for 14 C radioactivity.
  • Nepicastat concentrations of 0, 1, 2, 4, 8 nM were used to study inhibition kinetics at the following tyramine concentrations: 0.5, 1, 2, 3, 4 mM.
  • the 14 C counts were identical in each reaction which was carried out as described above.
  • a blank control without the enzyme was used to obtain the background.
  • the data were corrected for background, converted to activity in nmol/min, and plotted (1/V vs 1/S).
  • Km′ was calculated from the slopes and Y intercepts and linear regression was used to obtain Ki value.
  • IC 50 values for SKF-102698, nepicastat and Compound B against human and bovine DBH were obtained using the HPLC assay at the substrate concentrations of 0.1 mM tyramine, 4 mM ascorbate at pH 5.2 and 37° C. All three compounds caused a dose-dependent inhibition of DBH activity on both bovine and human enzyme.
  • IC 50 values calculated for nepicastat, Compound B and SKF-102698 showed that the S enantiomer (nepicastat) was more potent than the R enantiomer (Compound B by 3-fold against bovine DBH and 2-fold) against the human enzyme.
  • Nepicastat was more potent than SKF-102698 by 8-fold against bovine enzyme, and 9-fold against human DBH.
  • a Km of 0.6 mM was determined from the Lineweaver-Burk plot. Nepicastat (1-8 nM) caused a major shift in Km, as would be predicted for a competitive inhibitor. The inhibition of bovine DBH by nepicastat appears to be competitive with tyramine. A Ki of 4.7 ⁇ 0.4 nM was calculated by linear regression.
  • Nepicastat was a potent inhibitor of both human and bovine DBH. It was 8-9-fold more potent than SKF-102698. nepicastat (the S enantiomer) is 2-3 fold more potent than Compound B (the R enantiomer). The inhibition of bovine DBH by nepicastat appeared to be competitive with tyramine, with a Ki of 4.7 ⁇ 0.4 nM.
  • nepicastat was determined in the bindings assays outlined using standard radioligand filtration binding methods were used.
  • Nepicastat had moderate affinity for alpha, receptors (pKi of 6.9-6.7). The affinity at all other receptors examined was relatively low (pKi ⁇ 6.2).
  • nepicastat formulation was prepared by mixing vehicle with nepicastat powder, followed by shaking.
  • the 6- and 20-mg/ml Nepicastat formulations were prepared by diluting the 60-mg/ml formulation with vehicle.
  • the reconstituted nepicastat formulations retained potency for the duration of use.
  • the aqueous vehicle and nepicastat formulations contained hydroxypropylmethylcellulose, benzyl alcohol, and polysorbate 80.
  • Dose selection was based on an acute toxicity study in which mice were administered single oral doses of 250, 1000, or 2500 mg/kg of nepicastat. Clinical signs of toxicity and death occurred at 1000 and 2500 mg/kg.
  • a single oral dose of vehicle or nepicastat formulation was administered by gavage to each mouse using a rodent intubator.
  • the oral route was selected because it is a proposed clinical route of administration.
  • Dose volumes were calculated on the basis of individual body weights recorded before dosing (body weight data are not tabulated in this report). Food and water were withheld from the mice 2.5 to 3.5 hours before dosing, instead of 1.5 hours as specified in the protocol. This deviation did not affect the integrity of the study.
  • mice in each treatment group were evaluated in groups of up to 3 over an interval of approximately 10 minutes each for clinical observations and protocol-specified behavioral tests.
  • One mouse in the 30-mg/kg group and 1 mouse in the 100-mg/kg group died after dosing and they were removed from the study.
  • Surviving mice were euthanatized and removed from the study at the end of the observation/testing period.
  • Rats Male male Sprague Dawley rats (250-350 g on study day ) were obtained from Charles Rivers Labs. Rats were housed under a normal light/dark cycle with lights on between 0900 Hrs. and 2100 Hrs. Animals were housed in pairs in standard metal wire cages, and food and water were allowed ad libitum.
  • the locomotor activity boxes consisted of a Plexiglas® box measuring 18′′ ⁇ 18′′ by 12′′ high. Surrounding the Plexiglas® boxes were Omnitech Digiscan Monitors (model # RXXCM 16) which consisted of a one inch ban of photobeams and photosensors numbering 32 per box. The number of photobeam breaks were analyzed by an Omnitech Digiscan Analyzer (model # DCM-8). The animals were tested in an enclosed room with a white noise generator running to mask extraneous noise.
  • SR-Lab San Diego Instruments, San Diego, Calif.
  • the rats were placed individually in a Plexiglas® cylinder (10 cm diameter) which is housed in a ventilated sound-attenuating enclosure.
  • Acoustic noise bursts (a broad band noise with a rise time and fall time of 1 msec) was presented via a speaker mounted 30 cm above the animal.
  • a piezoelectric accelerometer transforms the subject's movement into an arbitrary voltage on a scale of 0 to 4095.
  • each of seventy-two rats was placed in the startle apparatus, and after a 5 minute adaption period they were presented with an acoustic noise burst every 20 seconds for 15 minutes (45 startles total).
  • the average startle was calculated for each rat by taking the mean of startle number 11 through 45 (the first ten startles will be disregarded).
  • Sixty-four of these rats were then placed in one of eight treatment groups such that each group had similar mean startle values.
  • the eight treatment groups were as follows: SKF-102698 (100 mg/kg) and its vehicle (50% water/50% polyethylene glycol), clonidine (40 ⁇ g/kg), nepicastat (3, 10, 30 and 100 mg/kg), and their vehicle, dH 2 O. Previous work has shown that this matching procedure to be the most appropriate for startle since there is significant variability in startle response between rats, but a high degree of consistency within rats from one day to the next.
  • mice Each day after this testing procedure, eight rats (one rat from each of the eight treatment groups) was injected with their assigned drug treatment and immediately placed individually in a motor activity box. The rats motor activity was monitored for four hours. Next, the rats were placed in a transfer cage for fifteen minutes. At the beginning of this fifteen minutes the rat that has been assigned the clonidine treatment will receive another injection of 40 ⁇ g/kg. Next, the rats were placed in the startle apparatus, and after a five minute acclimation period they were presented with a 90 dB noise burst every minute for four hours.
  • the clonidine-treated group When compared to the vehicle-treated controls, the clonidine-treated group had significantly more horizontal activities at 2 and 2.5 hours, significantly more movements at 2 hours, and significantly less rest time at 2 hours (all p ⁇ 0.05). Note that the clonidine-treated group had significantly more rest time than the vehicle-treated controls at 1 hour (p ⁇ 0.05).
  • the SKF-102698-treated group When compared to the vehicle-treated controls, the SKF-102698-treated group had significantly less horizontal activities and significantly less movements at 2.5 hours (both p ⁇ 0.05). Note that the SKF-102698-treated group had significantly more movements than the vehicle-treated controls at 1.5 and 4 hours (both p ⁇ 0.05). No significant differences between SKF-102698 and vehicle were detected at any time examined in the rest time.
  • the horizontal activity and number of movements decreased for the first 2 hours and stayed low for the last 2 hours.
  • the rest time increased for the first 2 hours and remained elevated for the last 2 hours.
  • Nepicastat had no significant effects on the locomotor activity in rats. Animals treated with 3, 10, 30 or 100 mg/kg of nepicastat were not significantly different from the vehicle-treated controls at any time examined in the horizontal activity, no. of movements or rest time.
  • SKF-102698 (100 mg/kg) was not statistically significantly different from vehicle at any time for either startle response measurement.
  • Nepicastat administered at 3, 10, 30, or 100 mg/kg does not appear to effect the maximum or average startle response in rats at any time when compared to vehicle.
  • SKF-102698 behaved similarly to vehicle (PEG) for both startle responses at all times.
  • Clonidine successfully lowered both maximum and average startle response during earlier times, and behaved similarly to vehicle during later times.
  • the effects of chronic dosing of nepicastat in rats were examined. Between three and thirteen days prior to the first dosing day the rats were placed inside the startle apparatus and after a five minute acclimation period they were presented with a 118 dB noise burst on average once a minute (a variable inter-trial interval ranging between 30 and 90 seconds will be used) for 20 minutes. The startle responses were measured and a mean for the last twenty startle response was calculated for each rat.
  • the rats were randomly placed in one of the eight treatment groups (nepicastat, 5, 15 or 50 mg/kg, bid; SKF-102698, 50 mg/kg, bid; clonidine, 20 ⁇ g/kg, bid: d-amphetamine, 2 mg/kg, bid; dH 2 O or cyclodextrin (SKF-102698's vehicle).
  • Rats were dosed by oral gavage with a 10 ml/kg dosing volume. The rats were dosed in the morning and in the evening every day for ten day. The time in between morning and evening dosing will be between 6 and 10 hours. Previous work has shown that this matching procedure to be the most appropriate for acoustic startle reactivity since there is significant variability in startle response between rats, but a high degree of consistency within rats from one day to the next.
  • the animals were tested in an enclosed room with a white noise generator running. Motor activity tests were conducted immediately after the body core temperature reading taken on dosing day ten (about 3 hours and 35 minutes after the morning daily dose of nepicastat, and SKF-102698, and 20 minutes prior to the daily administration of clonidine and d-amphetamine on dosing day ten). Motor activity tests were run for one hour. A diagnostic program was run on each of the motor activity chambers prior to each test session to assure that the photo beams and light sensors were operating properly. Motor activity has been shown to be sensitive to changes in central dopamine levels (Dietze and Kuschinsky, 1994) which makes this behavioral test a potential sensitive assay to the effects of DBH inhibitors in-vivo. D-amphetamine was used as the positive control for this assay.
  • Rat body core temperatures were obtained by inserting the rectal probe 2 cm into the colon of each rat. Each rat's body core temperature was measured three times and the average of the three reading was calculated. Body core temperature readings were obtained immediately prior to the ten day chronic dosing schedule (to obtain a baseline), and three and half hour after the morning daily dose of nepicastat, and SKF-102698, and 15 minutes prior to the daily administration of clonidine and d-amphetamine, on dosing days one, five and ten. Body core temperature has been shown to be sensitive to both dopamine and norepinephrine levels, which makes this behavioral test a potential sensitive assay to the effects of DBH inhibitors in-vivo. Both clonidine (an alpha 2 agonist), and d-amphetamine (a dopamine releaser) were used as the positive controls for this assay.
  • a piezoelectric accelerometer attached below the plexiglas cylinder transduced the subject's movement into a voltage which was then rectified and digitized (on a scale from 0 to 4095) by a PC computer equipped with SR-Lab software and interface assembly.
  • a decibel meter was used to calibrate the speakers in each of the eight test station to ⁇ 1% of the mean.
  • a SR-Lab calibrating instrument was used to calibrate each of the eight startle detection apparatuses to ⁇ 2% of the mean.
  • Startle reactivity and pre-pulse inhibition tests were run concurrently immediately alter the motor activity test (about 4 hours and 40 minutes after the morning daily injection of nepicastat, and SKF-102698, and 10 minutes after a supplemental administration of clonidine and d-amphetamine on dosing day ten).
  • the startle reactivity and pre-pulse inhibition tests consisted of placing each rat individually into a SR-Lab test station and after a five minute acclimation period the rats were presented with one of three different types of noise bursts (and startle reaction measured) on average once a minute (a variable inter-trial interval ranging between 30 and 90 seconds was used) for an hour (60 total noise bursts and startle reactions).
  • the three different types of noise bursts consisted of a loud noise burst (118 dB), and a relatively quite noise burst (77 dB), the quite burst preceding the loud noise bursts by 100 msec (pre-pulse inhibition trial). These trials were presented in pseudo-random order. Pre-pulse inhibition has been shown to be sensitive to changes in mesolimbic dopamine levels. Furthermore, acoustic startle reactivity has also been shown to be sensitive to changes in dopamine and norepinephrine levels which makes these behavioral test a potential sensitive assay to the effects of DBH inhibitors in vivo. Clonidine and d-amphetamine served as the positive control for the acoustic startle reactivity and pre-pulse inhibition of acoustic startle tests.
  • Each rats spontaneous locomotion was obtained by calculating the total number of photobeams that the subject broke during the testing session.
  • the subject's reaction was measured during each trial for the 40 msec window after the stimulus was presented.
  • Each startle reaction was calculated by taking the avenge of 40 readings (one per millisecond) starting immediately after each noise burst.
  • Acoustic startle reactivity was calculated by determining the mean response for each subjects startle elicited by the 118 dB acoustic burst.
  • Pre-pulse inhibition values were calculated by subtracting the mean startle response elicited by the 77 dB pulse-118 dB pulse paired trial (pre-pulse inhibition trial described above) from the 118 dB alone trial and then dividing this value by the 118 db alone trial for each rat, i.e. ([118 dB trial value ⁇ pre-pulse inhibition trial value] ⁇ 118 db trial value). 26859/14442/DOCS/1 946886.3
  • Spontaneous motor activity was measured for each animal every 15 min for 1 hour. Each time block (every 15 min) was analyzed separately. Kiruskal-Wallis test (nonparametric technique) was performed to test for the difference between treatment groups. If the overall significant difference is not detected, Bonferroni's adjustment for multiple comparisons is then made.
  • AVGMEAN mean average voltage
  • RATIO mean percent prepulse inhibition
  • TRIALT trial type
  • Pre-pulse inhibition values were calculated by subtracting the mean startle response elicited by the 77 dB pulse-118 dB pulse paired trial (pre-pulse inhibition trial described above) from the 118 dB alone trial and then dividing this value by the 118 db alone trial for each rat, i.e. ([118 dB trial value ⁇ pre-pulse inhibition trial value] ⁇ 118 db trial value).
  • the average startle response and the percent prepulse inhibition were analyzed using Analysis of Variance.
  • the model included terms for treatment, animals nested within treatment, time and treatment by time interaction. Treatment effects were tested using the error term for animals nested within treatment. Overall treatment effects and treatment effects by time were studied. The method of Fisher's Least Significant Differences was used to adjust for multiple comparisons. If the overall treatment or treatment by time effects were not significant (p-value>0.05) then a Bonferroni adjustment was made. If the overall treatment effects were nonsignificant, then the adjustment was applied to the specific pairwise comparisons. Further, if the specific pairwise treatment effect was not significant (p-value >0.05), then the adjustment was also applied to the treatment effects within time. If both the overall treatment and treatment by time effects were not significant (p-value >0.05) then a Bonferroni adjustment was made for the individual comparisons within time and averaging over time.
  • the change from pre-dose in body weights was calculated for each animal for the analysis.
  • a repeated measures two-way ANOVA was used to test for the overall effects of treatment, time and treatment by time interaction.
  • One-way ANOVAS were then performed to test the treatment effect at each day.
  • the positive controls (d-amphetamine and clonidine) significantly increasing body core temperature on day one of the chronic dosing, but no other compound had any significant effect on body core temperature at any time.
  • the d-amphetamine group had significantly higher locomotor activity than the vehicle control at all times examined.
  • the clonidine group was not significantly different from the vehicle controls at any time examined.
  • the SKF-102698 50 mg/kg b.i.d. group had significantly lower locomotor activity than its vehicle control at the first 45 minutes (i.e. samples 1-3), but not significant after 45 minutes.
  • the clonidine group had just significantly higher percent prepulse inhibition than the vehicle control and were not significantly different from vehicle during times 3 and 4.
  • Neither d-amphetamine nor SKF-102698 was significantly different from their own vehicle at any time. None of the nepicastat dose groups were significantly different from dH 2 O at any time.
  • the SKF-102698 50 mg/kg b.i.d. group had significantly lower startle response compared to cyclodextrin, and also had significantly lower startle response as compared to the nepicastat 50 mg/kg b.i.d. group.
  • the SKF-102698 (50 mg/kg b.i.d.) group had significantly lower startle response than the cyclodextrin group at all times. During times 1 and 3, the nepicastat (50 mg/kg b.i.d.) group had significantly higher startle response than the SKF-102698( 50 mg/kg b.i.d.) group. No other significant differences were detected.
  • the d-amphetamine group had a significantly smaller change in body weight from pre-dose than the vehicle controls (p ⁇ 0.01).
  • the vehicle controls had a significantly greater increase from pre-dose in body weight than the amphetamine group at treatment days 4-10.
  • the clonidine group was not significantly different from the vehicle controls at any time examined.
  • the SKF-102698 (50 mg/kg b.i.d.) group showed a significantly smaller increase (p ⁇ 0.01) in body weight from pre-dose baseline than its vehicle control (SKF-vehicle).
  • the SKF-vehicle controls had a significantly greater increase from pre-dose in body weight than the SKF-102698 group at treatment days 2-10, except days 3 and 6. Importantly, there was no difference in changes in body weight between the SKF-vehicle and the vehicle control groups on any day.
  • MPTP 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
  • nepicastat Six squirrel monkeys, three non-lesioned and three lesioned (received 2 mg/kg MPTP 3 months prior), were used to study the optimal route of administration of nepicastat. Three different approaches were examined including (i) insertion into treats, (ii) oral syringe, and (ii) oral gavage. Insertion of nepicastat solution (5 mg/ml) into marshmallows was tested in 3 non-lesioned monkeys and proved to be a poor route of drug administration due to failure of animals to ingest treats probably due to adverse taste.
  • Oral syringe injection of nepicastat (0.5, 2, and 5 mg/kg) into the mouth of three non-lesioned and three lesioned monkeys was also not an acceptable route since animals tended to spit out the solution at the highest drug concentration.
  • Oral gavage administration was carried out in 3 MPTP-lesioned monkeys at the highest dose (5 mg/kg) and was well accepted.
  • nepicastat Six squirrel monkeys, three non-lesioned and three lesioned (received 2 mg/kg MPTP 3 months prior), were used to study safety and tolerability of nepicastat. Animals received nepicastat at a concentration of 0.5, 2.0, or 5.0 mg/kg twice daily, (10 am and 2 pm), for 5 days with a two-day washout between the different dose levels. Nepicastat was administered via oral syringe at the 0.5, 2.0 and 5.0 mg/kg doses and as oral gavage at the 5.0 mg/kg dose. Drug was well tolerated at the two lower doses. One non-lesioned monkey receiving 5.0 mg/kg had light beige colored loose stools on the final two days of administration that resolved upon one day withdrawal of drug.
  • Twenty four squirrel monkeys, fourteen females and ten males were used in a Parkinsonian model.
  • the twenty-four animals were randomly assigned to one of four treatment groups, with 6 animals per group.
  • the groups consisted of the following: Group A (6 animals) received placebo (water) treatment; Group B (5 animals) received drug nepicastat at 1 mg/kg/day (0.5 mg/kg twice daily); Group C (6 animals) received 4 mg/kg/day (2 mg/kg twice daily); and Group D (6 animals) received 10 mg/kg/day (5 mg/kg twice daily).
  • Group B one animal died acutely following MPTP-lesioning, and was not replaced.
  • IRAM infrared activity monitor
  • animals required additional doses of MPTP (2 mg/kg) to obtain a sufficient degree of lesioning to display parkinsonian symptoms, defined as an average total clinical rating score greater than 3. All animals received a final post-MPTP behavioral assessment (IRAM and CRS), within three weeks of starting the efficacy study. This final post-MPTP evaluation was used to establish a baseline clinical parkinsonian state and used as a pretreatment value for statistical analysis.
  • L-Dopa was administered at a concentration of either 2.5, 5, or 7.5 mg/kg by oral gavage twice daily (at 10 am and 2 pm) for 7 consecutive days. Behavior was determined by IRAM and CRS. Clinical rating was carried out 60 to 90 minutes following the 10 am morning dose on the last 4 days of treatment. Raters (one to three individuals) were blinded to the different treatment groups. IRAM assessment were preformed for 90 minutes immediately following drug administration at 2 pm on the last 2 to 5 days of drug treatment. There was a minimum 2 day washout period between each treatment dose.
  • Nepicastat or water (as placebo) was administered for 12 days following a minimum 2 day washout after L-Dopa dosing.
  • Drug was administered twice daily at 10 am and 2 pm by oral gavage.
  • Behavior was rated by IRAM and CRS. The CRS was conducted in the morning, 60 to 90 minutes after the 10 am dose of nepicastat on the last 5 days of drug treatment. Raters (one to three individuals) were blinded to the different treatment groups. IRAM assessments were preformed for 90 minutes immediately following drug administration at 2 pm on the last 5 days of drug treatment.
  • locomotor activity and clinical rating scores were monitored.
  • the average locomotor activity was calculated pre- and post-MPTP-lesioning for each animal.
  • the pre-MPTP-lesioning baseline was determined by averaging ten 1-hour monitoring sessions.
  • the post-MPTP (pre-treatment) behavioral assessment was obtained within three weeks of commencing the efficacy study.
  • the post-MPTP-lesioning locomotor activity was determined by averaging three to five 1-hour monitoring sessions (IRAMS). Activity monitoring was reported as “movements/10 minutes”. A higher score was considered a faster animal.
  • the Wilcoxon sign rank test was used to compare pre- and post-MPTP-lesioning activity for each group of animals (groups A through D).
  • IRAM Locomotor activity was monitored every 10 minutes for a minimum of 90 minutes following each drug level. A higher rating is considered a faster (less parkinsonian) animal.
  • Statistical analysis consisted of: (1) comparisons between the average CRS of placebo to nepicastat at 1, 4, and 10 mg/kg/day using the Kniskal-Wallis (non-parametric analysis of variance). This comparison was repeated using the average CRS for each experimental drug concentration corrected by the final post-MPTP ratings for each animal. The corrected clinical scores are clinical scores of experimental drug at each concentration as a ratio of post-MPTP clinical scores. (2) Pairwise comparisons between the average CRS post-MPTP lesioning (pre-treatment) to 2.5, 5.0, and 7.5 mg/kg L-Dopa and placebo treatment using Friedman's analysis (non-parametric analysis of variance, repeated measures). The same analysis was performed for nepicastat at concentrations of 1, 4, and 10 mg/kg. Dunnett's post hoc analysis for non-parametric data was performed when needed.
  • IRAM activity monitoring
  • CRS clinical rating scale
  • nepicastat Based on a non-pairwise comparative analysis of 4 different treatment groups (1,4, and 10 mg/kg of nepicastat and placebo), nepicastat produced no significant effect in parkinsonian symptoms compared to placebo (water treatment) in the MPTP-lesioned non-human primate model of PD. Based on a pairwise comparative analysis of animals, (animals of the same group examined pre and post treatment), nepicastat at 4 and 10 mg/kg/day concentrations showed a significant effect in parkinsonian symptoms compared to post-MPTP lesioning, (pre-treatment evaluation). Placebo had a borderline significant effect.
  • a pharmacokinetic study was carried out to determine the plasma concentration of nepicastat in the squirrel monkey. This study was carried out concurrently with the safety and tolerability study. Three MPTP-lesioned squirrel monkeys (#353, 358 and 374) were used. One milliliter of blood (drawn from the femoral vein of each animal) was collected for analysis. Nepicastat was administered at concentrations of 1, 4, and 10 mg/kg for 5 days with a 2-day washout between each drug concentration. Blood was collected for analysis 1 hour prior to the first dose to establish baseline and at 6 hours after this first drug dose of each of the different drug levels.
  • a second pharmacokinetic study was carried out to determine the steady-state plasma level of nepicastat. This study was carried out concomitantly with the efficacy study where animals were tested on each of three different drug concentrations for 12 days. One milliliter of blood was drawn from the femoral vein 6 hours after the first dose on day 1, then 6 hours after the first dose on day 7, and finally 6 hours after the first dose on day 12. Baseline plasma levels were determined on samples collected the week prior to drug dosing.
  • Nepicastat was dissolved in deionized water (vehicle) to a free base concentration of 1 mg/ml. Oral dosing volume for nepicastat or vehicle was 10 ml/kg.
  • SCH-23390 was dissolved in saline (vehicle) to a free base concentration of 0.2 mg/ml. Nepicastat or saline were administered intravenously as a bolus in a volume of 1.0 ml/kg followed by 0.2 ml flush of isotonic saline.
  • mice were randomly assigned to four treatment groups: vehicle (iv)/vehicle (po); vehicle (iv)/nepicastat (po); SCH-23390 (iv)/vehicle (po); or SCH-23390 (iv)/nepicastat (po).
  • vehicle (iv)/vehicle (po) vehicle (iv)/nepicastat (po); SCH-23390 (iv)/vehicle (po); or SCH-23390 (iv)/nepicastat (po).
  • each animal was anesthetized with halothane and euthanized via decapitation.
  • the cortex, left ventricle (apex), and mesenteric artery were dissected out, weighed, and fixed in 0.4 M perchloric acid. Tissues were then frozen in liquid nitrogen and stored at ⁇ 70° C. Biochemical analysis are performed on these tissues at a later date to determine catecholamine levels including dopamine and norepinephrine levels.
  • Blood pressure and heart rate were analyzed separately. The change from baseline for blood pressure and heart rate were analyzed by an analysis of variance (ANOVA) with effects for treatment, time, and their interaction. This analysis was performed both for the post-iv time period and for the post-oral time period. Further analyses were performed at each time by an ANOVA with a main effect for time. Pairwise comparisons were performed following each ANOVA by Fisher's LSD strategy with a Bonferroni correction when the overall treatment effect was not significant.
  • ANOVA analysis of variance
  • Intravenous treatment with SCH-23390 resulted in a significant decrease (p ⁇ 0.05) in heart rate during the post-oral period at 120 min and 240 min compared to vehicle control. Nepicastat did not decrease the heart rate as much as observed in vehicle treated animals. This was statistically significant (p ⁇ 0.05) at 150 and 180 min post dose. The large variability in heart rate observed over the course of this experiment should be noted.
  • Intravenous administration of SCH-23390 produced a small (5 ⁇ 1 mmHg) yet significant decrease (p ⁇ 0.05) in mean arterial pressure compared to animals that received vehicle during the 15 min post-iv period.
  • Oral treatment with nepicastat caused a significant decrease (p ⁇ 0.05) in mean arterial pressure by 30 min post dose which continued for the duration of the experiment.
  • Pretreatment with SCH-23390 did not significantly attenuate the antihypertensive effects observed with nepicastat administration alone.
  • mice Male Crl:COBS(WI)BR rats of 15 weeks old were used. Twenty-four rats were chronically implanted with telemetry implants (TA11PA-C40, Data Sciences, Inc., St. Paul, Minn.) for measurement of arterial blood pressure, heart rate and motor activity. The rat was anesthetized with pentobarbital sodium (60 mg/kg, ip) and its abdomen shaved. Under aseptic conditions, an incision was made on midline. The abdominal aorta was exposed, and cannulated with the catheter of a telemetry transmitter unit. After the transmitter was sutured to the abdominal musculature, the skin was closed. Each rat was allowed to recover for at least 2 weeks before being subjected to drug administration.
  • TA11PA-C40 Data Sciences, Inc., St. Paul, Minn.
  • the rats were randomly divided into 4 treatment groups: Vehicle (p.o.), Hydralazine (10 mg/kg, p.o.), nepicastat (30 mg/kg, p.o.), nepicastat (100 mg/kg, p.o.).
  • Vehicle p.o.
  • Hydralazine 10 mg/kg, p.o.
  • nepicastat 30 mg/kg, p.o.
  • nepicastat 100 mg/kg, p.o.
  • SBP Systolic blood pressure
  • DBP diastolic blood pressure
  • MBP mean blood pressure
  • HR heart rate
  • MA motor activity
  • a computerized data collection system was used to continuously collect data on SBP, DBP, MBP, HR, and MA. Data on each rat were collected every 5 min. for 10 sec. These were then averaged hourly and standard errors of the mean (SEM) calculated. All values were expressed as means i SEM. Statistical significance was defined as a p level of less than 0.05. Data on MBP, HR and MA were analyzed separately. Each analysis was done on 26 time points measured each day. A two-way ANOVA with main effects for treatment and time and their interaction was used. If an overall treatment effect or a significant interaction was detected, a series of one-way ANOVA at each time point would be performed. The pairwise comparisons at each time point were performed using Dunn's procedure. If no overall treatment effect was detected, then the pairwise difference from control would be performed by adjusting the critical value using a Bonferroni adjustment.
  • nepicastat at 30 mg/kg (all doses expressed hereafter are po) tended to slowly lower blood pressure but did not induce a consistent hypotensive effect on day 1.
  • a peak hypotensive effect of ⁇ 10 mmHg was observed on day 2 at hour 13.
  • Similar degrees of antihypertensive effects were induced throughout the study.
  • the compound induced a peak antihypertensive response of ⁇ 11 mmHg 22 hr after dosing on day 1 (p ⁇ 0.01).
  • MBP continued to decrease and reached its nadir of approximately ⁇ 17 mmHg on day 3 (p ⁇ 0.01). The MBP remained low throughout the study.
  • Hydralazine at 10 mg/kg caused an immediate hypotensive effect which subsided in 10 hr, and a maximal decrease of -24 mmHg (p ⁇ 0.01) in MBP was observed within 1 hr after dosing on day 1. Similar transient hypotensive effects were observed throughout the study.
  • Body weights were recorded daily. For body weight, a two-way ANOVA with respect to the changes from pre-dose was used to analyze overall effects for treatment, day, and treatment by day interaction. Then a one-way ANOVA was performed for each day, and pairwise comparisons for the drug-treated groups to the vehicle controls were made using Dunn's procedure and Fisher's LSD strategy to adjust for multiple comparisons. Compared to that treated with vehicle, none of the drug treatments had any effect on body weights (p ⁇ 0.05). Although treatment with nepicastat at 100 mg/kg tended to decrease body weight on day 3, it was not statistically significant.
  • Nepicastat reduces the conversion of dopamine to norepinephrine.
  • Basic tests for nepicastat activity measure the levels of plasma or urinary dopamine or the ratio of dopamine to norepinephrine.
  • Nepicastat treatment can increase plasma or urinary levels of dopamine or increase the dopamine/norepinephrine ratio in plasma or urine.
  • FIG. 5 Shown in FIG. 5 , are the levels in urinary dopamine levels in normal volunteers after 24 hour treatment with nepicastat.
  • CHF chronic heart failure
  • Dopamine/norepinephrine ratios in the brain of rodents treated with nepicastat were determined. Dopamine/norepinephrine ratios increased in the brains of rodents treated with nepicastat or disulfiram.
  • the delayed-matching-to-position (DMTP) test is used to examine the potential effects of drugs on short-term or working memory in rats.
  • nepicastat Prior to the commencement of testing nepicastat in the delayed matching to position study, pilot studies were performed with the aims of assessing the behavioural/physiological effects of both acute and repeated administration of the highest proposed dose of nepicastat (100 mg/kg p.o.) and establishing the maximum tolerated oral dose of physostigmine administered repeatedly.
  • nepicastat 100 mg/kg p.o. or vehicle was administered twice daily (06:00 and 18:00 h) for 10 days (once on day 11) to groups of 8 rats.
  • the weights of the animals were monitored throughout the study and, on day 5, the animals were observed ‘blind’ by an independent observer in order to assess any overt behavioural/physiological effects following repeated administration.
  • groups of 8 rats received vehicle or physostigmine (0.3, 1, 3 or 10 mg/kg p.o.) using the same dosing schedule. The weights of the animals were monitored throughout the study.
  • Nepicastat (30 or 100 mg/kg p.o. acutely) did not induce any overt behavioural/physiological changes. Similarly, there were no overt effects of repeated administration of nepicastat at a dose of 100 mg/kg p.o. However, in the latter study the drug-treated animals displayed a mean loss in body weight of 28 g after 11 days whereas controls had a mean increase in body weight of 1 g. Drug treated animals also became more irritable than controls when handled during the 11 day study.
  • nepicastat for the DMTP study was reduced to 30 mg/kg p.o. and a dose of 1 mg/kg p.o. of physostigmine was chosen for repeated administration.
  • nepicastat (1, 3, 10 and 30 mg/kg p.o. b.i.d.) or physostigmine (1.0 mg/kg p.o., b.i.d.) were examined across 10 successive days of testing in the DMTP task.
  • scopolamine Hbr 0.1 mg/kg, s.c., 30 mm pretreatment time.
  • the dose of scopolamine was selected on the basis of data from a pilot DMTP study in which 0.1 mg/kg of scopolamine was found to induce a significant impairment in choice accuracy.
  • scopolamine was treated with scopolamine.
  • Another group of rats received only vehicle treatment throughout the experiment.
  • the purpose of the final scopolamine test was to determine whether chronic administration of nepicastat or physostigmine would reverse a scopolamine induced impairment in choice accuracy in the DMTP task.
  • the dependent measures in the present study included percentage of correct choices, the latency to make choice responses and the number of trials which the animals were able to complete during the 70 min test sessions. Changes in the former measure may indicate changes in memory and/or attentional function whereas changes in the latter two measures may be indicative of other non-cognitive effects of the drugs.
  • Fifty-six male Sprague Dawley rats weighing between 200-290 g at the beginning of training, were used. They were housed in groups of four per cage and were each fed approximately 12-15 g of food per rat per day. This amount of food maintained the rats at approximately 85% of their free-feeding weight. Any animals which began to drop below this weight were given additional food. Water was freely available. The animals were maintained on a 12:12 hour light/dark cycle with the light period beginning at 6 a.m.
  • Rats were initially trained to retrieve Noyes 45 mg Formula ‘A’ food pellets from behind the magazine flap. Rats were then trained to press both the left and right levers to obtain food reward. Either the left or right lever was randomly presented during a 30 mm session. A response to the inserted lever resulted in the retraction of the lever, delivery of a food pellet and illumination of the magazine light. The magazine light remained on until the pellet was retrieved.
  • a response to the sample lever resulted in retraction of both levers, delivery of a food pellet and the illumination of the magazine light.
  • the magazine light remained on until the flap was pressed.
  • a response to the incorrect lever did not produce a food pellet and initiated a 10 s time-out (TO) period during which the houselight was extinguished.
  • TO time-out
  • a 30 s ITI was initiated before the commencement of a new trial.
  • the lever inserted as the sample lever was semi-randomly determined such that the right and lefthand levers were presented as the sample lever 8 times in a block of 16 trials.
  • a correction procedure was used throughout this and all subsequent training.
  • the lever to be inserted (left or right) as the sample lever was randomly determined by the computer on non-correction trials (i.e., the first trial of the session and trials that immediately followed a trial on which a correct choice occurred). Each time an incorrect response occurred, the lever that was not chosen (i.e., the ‘correct’ lever) was presented as the sample on the subsequent ‘correction’ trial.
  • These correction trials prevented position habits (i.e., always responding on either the left or right lever and achieving 50% correct). The number of correction trials was recorded, but only the data collected on the non-correction trials were used to evaluate the percentage of correct choices.
  • the rats received oral administration of either vehicle, physostigmine (Phys) or nepicastat (1, 3, 10 or 30 mg/kg). Due to the high degree of choice accuracy displayed by the vehicle treated animals during sessions 68 and 69, the 0, 8, 16 and 32s delay was also used during the final test session (Session 70) in which all but the Vehicle/Vehicle treated group received 0.1 mg/kg of scopolamine HBr administered s.c. 30 minutes prior to testing. The animals in the Vehicle/Vehicle group received a s.c. injection of saline 30 minutes prior to the final test session. Thus, the drug treatments administered to the seven groups during the 11 consecutive days of the present experiment were:
  • Session 60-69 Session 70 Group (0, 8, 16, and 32 s delay) (0, 4, 8, and 32 s delay) 1 Vehicle Vehicle/Vehicle 2 Vehicle Vehicle/Scopolamine 3 nepicastat 1.0 mg/kg nepicastat 1.0 mg/kg/Scopolamine 4 nepicastat 3.0 mg/kg nepicastat 3.0 mg/kg/Scopolamine 5 nepicastat 10 mg/kg nepicastat 10 mg/kg/Scopolamine 6 nepicastat 30 mg/kg nepicastat 30 mg/kg/Scopolamine 7 Physostigmine Physostigmine/Scopolamine
  • the data collected and analyzed in the present DMTP study include 1) the percentage of correct responses; 2) the latency between performance of a response to the sample lever and the performance of the choice response and 3) the total number of correction and non-correction trials completed. The former two dependent measures were collected for the non-correction trials only.
  • Physostigmine sulphate (1.0 mg/kg, supplied by RBI) and nepicastat (1, 3, 10 and 30 mg/kg, supplied by Roche) were administered p.o. twice daily starting at 6:00 am and 6:00 pm.
  • Scopolamine HBr (0.1 mg/kg, supplied by Sigma) was administered s.c. 30 minutes prior to the last test session.
  • Physostigmine and nepicastat were dissolved or suspended in distilled water and injected in a volume of 2.5 ml/kg.
  • Scopolamine HBr was dissolved in saline and injected in a volume of 1.0 ml/kg. All drug doses are expressed as base weight.
  • nepicastat induced a marked dose- and delay-dependent impairment in choice accuracy.
  • a post hoc Dunnett's test conducted at the 32 s delay found that both the 10 and the 30 mg/kg nepicastat treated groups showed impairments in percentage of correct choices relative to the vehicle treated animals.
  • n ⁇ 4 occurred in all the groups except for the Vehicle/Vehicle and the Vehicle/Scopolamine in which seven and four rats, respectively, completed trials at each of the four delays.
  • Nepicastat when administered alone does not appear to induce memory enhancing effects in the DMTP test. It is notable that the delay induced memory impairment observed in the vehicle treated control animals appeared to dissipate across the five blocks of testing. However, by the fifth block of testing the vehicle treated control animals were still showing a delay-dependent memory impairment with 100% choice accuracy at the 0 s delay and 80% choice accuracy at the 32 s delay. Thus, a ceiling effect in the performance of the vehicle treated animals at the 32 s delay was not observed.
  • nepicastat may have selective memory disrupting effects.
  • Physostigmine did not improve performance on any of the treatment days and actually produced a delay-independent impairment in choice accuracy during Block 3 of testing (Days 5 & 6).
  • the results from a scopolamine challenge test on day 11 in which the animals were co-administered scopolamine HBr (0.1 mg/kg) and nepicastat or physostigmine could not be analyzed due to the small number of subjects in the nepicastat and physostigmine treated groups that were able to perform the DMTP task.
  • nepicastat is capable of reversing some of the cognitive disruption induced by scopolamine, an effect that may be masked by other “non-cognitive” actions of the compound.
  • Nepicastat induced significant dose- and delay-dependent impairments in choice accuracy.
  • the animals treated with 10.0 mg/kg of nepicastat showed absolutely no impairments in choice accuracy at the 0, 8 and 16 s delays.
  • the animals in the 10.0 mg/kg nepicastat group were impaired relative to the vehicle treated group.
  • the group treated with the highest dose of 30.0 mg/kg of nepicastat showed no impairments in choice accuracy at the 0 s delay, a tendency to impaired choice accuracy at the 8 and 16 s delays and a significant impairment in choice accuracy relative to the vehicle treated groups at the 32 s delay.
  • the delay-dependent nature of these drug-induced impairments in choice accuracy suggests that the compound may be acting directly on short-term or working memory.
  • Nepicastat had a small effect on latency to complete trials which was apparent on the fourth block of training, during which the animals treated with 10.0 mg/kg of nepicastat took longer to completed the 32 s delay trials than the vehicle treated animals. This effect was not dose-dependent and was not observed in the group treated with 30.0 mg/kg.
  • Physostigmine did not improve performance of the rats in the DMTP test. In fact, the animals treated with physostigmine showed a significant impairment in percentage of correct choices during Block 3 of training. In contrast to the effects obtained with nepicastat, the impairment in choice accuracy induced by physostigmine was delay-independent: the interaction term from the analysis of variance did not approach statistical significance. Thus, the effects of physostigmine on response accuracy are likely to be secondary to behaviorally toxic effects of the drug when it is administered at this dose. The animals appeared to develop tolerance to these effects over the last two blocks of training during which the impairments in choice accuracy induced by physostigmine no longer reached statistical significance.
  • physostigmine did not appear to reverse the effects of scopolamine during the scopolamine test. It is possible that a different dose of physostigmine may have been effective against scopolamine. We have not previously attempted to reverse scopolamine with physostigmine using the present dosing regime and therefore have no historical data to compare with the present results. This lack of effect of physostigmine may be due to the fact that, compared with acute administration, a lower dose of physostigmine had to be employed for chronic administration. The animals would not have tolerated repeated administration of a higher dose of physostigmine (see results of the pilot study) which may be required to reverse the effects of scopolamine.
  • nepicastat did not appear to reverse the effects of scopolamine although it is interesting that the two animals treated with 30.0 mg/kg of nepicastat that were able to perform during the scopolamine test showed higher choice accuracy than any of the animals in any of the other scopolamine treated groups. Further research would be needed to determine unequivocally whether acute or chronic treatment with nepicastat can reverse the effects of scopolamine in this test.
  • Nepicastat appears to have specific memory-disrupting effects which are apparent after 8 days of dosing. Physostigmine did not improve performance on any of the treatment days and actually produced a delay-independent impairment in choice accuracy during Block 3 of testing (Days 5 & 6). The results from a scopolamine challenge test on day 11 in which the animals were co-administered scopolamine HBr (0.1 mg/kg) and nepicastat or physostigmine could not be analyzed due to the small number of subjects in the nepicastat and physostigmine treated groups that were able to perform the DMTP task.
  • nepicastat is capable of reversing some of the cognitive disruption induced by scopolamine, an effect that may be masked by other “non-cognitive” actions of the compound.
  • nepicastat induced dose- and delay-dependent impairments in choice accuracy.
  • nepicastat a selective dopamine ⁇ -hydroxylase inhibitor
  • SHRs acute studies in SHRs.
  • the antihypertensive effects of nepicastat were examined chronically in the same strain of rats.
  • angiotensin converting enzyme inhibitor enalapril effects of the treatments on the cardiac hypertrophy in SHRs were also examined.
  • SBP systolic blood pressure
  • DBP diastolic blood pressure
  • MBP mean blood pressure
  • HR heart rate
  • MA motor activity
  • the rats were sacrificed and the left ventricles were collected, weighted (wet weight), and lyophilized for at least 24 hr to obtain dry weights.
  • nepicastat and enalapril were prepared in water. All doses were given orally to the rat in 10 ml/kg and were expressed as free base equivalents. Enalapril (Vasotec®) was obtained commercially from a local pharmacy.
  • a computerized data collection system was used to continuously collect data on SBP, DBP, MBP, HR, and MA. Data on each rat were collected every 5 min. for 10 sec. These were then averaged hourly and standard errors of the mean (SEM) calculated. At the end of the treatment, left ventricular mass (dry and wet weights) were obtained. Body weights were recorded daily.
  • nepicastat for series I and II, Oral administration of nepicastat at 3 and 10 mg/kg (all doses expressed hereafter are po) did not significantly affect blood pressure on any of the 30 day treatment (data not shown).
  • nepicastat gradually lowered MBP on day 1 and continued to lower the MBP to a maximal of ⁇ 20 mmHg on day 3 (p ⁇ 0.01), with little recovery within 24 hr. Similar antihypertensive effects were induced throughout the study.
  • the compound induced a peak antihypertensive response of ⁇ 29 mmHg 21 hr after dosing on day 1 (p ⁇ 0.01).
  • MBP continued to decrease and reached its nadir of approximately ⁇ 42 mmHg on day 3 (p ⁇ 0.01). The MBP remained low throughout the study.
  • enalapril at 1 mg/kg did not affect HR
  • the compound at 10 mg/kg tended to induce a transient small tachycardia within 2 hr after dosing.
  • none of the treatments, i.e., enalapril (1 mg/kg), nepicastat (30 mg/kg) or the combination consistently affected HR.
  • the groups treated with nepicastat (15, 30 and 60 mg/kg) and enalapril (1 mg/kg) tended to exhibit lower HR than the group treated with enalapril (1 mg/kg) alone.
  • Nepicastat at 3-100 mg/kg did not affect the cardiac hypertrophy observed in SHRs (p>0.05).
  • enalapril at 1 mg/kg did not regress the hypertrophy, but the co-administration of enalapril (1 mg/kg) and nepicastat (30 mg/kg) significantly decreased the left ventricular mass of the SHRs (p ⁇ 0.01).
  • Series IV however, effects of co-administration of enalapril (1 mg/kg) and nepicastat at 15, 30 and 60 mg/kg on the left ventricular mass were not different from enalapril alone (p>0.05).
  • enalapril at 10 mg/kg significantly decreased (p ⁇ 0.05) or had no effect on the body weights of the rat.
  • treatment with enalapril at 1 mg/kg slightly decreased body weight
  • co-administration of enalapril (1 mg/kg) and nepicastat (30 and 60 mg/kg) slightly increased the body weights of the rat.
  • the pre-dose body weights of the rats treated with vehicle, enalapril, and nepicastat at 3 and 10 mg/kg were 387 ⁇ 11, 415 ⁇ 12, 407 ⁇ 4, and 415 ⁇ 12 g, respectively.
  • the pre-dose body weights of the rats treated with vehicle, enalapril, and nepicastat at 30 and 100 mg/kg were 399 ⁇ 10, 389 ⁇ 6, 389 ⁇ 9, and 401 ⁇ 10 g, respectively.
  • the pre-dose body weights of the rats treated with vehicle, enalapril, and nepicastat at 30 mg/kg without and with enalapril were 365 ⁇ 9, 371 ⁇ 8, 361 ⁇ 7, and 369 ⁇ 7 g, respectively.
  • the pre-dose body weights of the rats treated with enalapril alone and co-administrations of nepicastat at 15, 30, and 60 mg/kg were 357 ⁇ 6, 363 ⁇ 6, 347 ⁇ 8, and 346 ⁇ 8 g, respectively.
  • nepicastat The effects of 30-day chronic oral administration of nepicastat on blood pressure, heart rate, motor activity and left ventricular mass were evaluated in four series of experiments in spontaneously hypertensive rats (SHRs) with radio-telemetry implants.
  • SHRs spontaneously hypertensive rats
  • Daily treatment of nepicastat at 3 and 10 mg/kg (n 6) did not affect blood pressure.
  • nepicastat In groups treated with nepicastat at 3-10 mg/kg or enalapril at 1 mg/kg, no significant effects on heart rate were observed. The groups that received nepicastat at 30 or 100 mg/kg, however, exhibited slight bradycardia during the awake hours of the rat. In contrast, enalapril at 10 mg/kg induced a transient tachycardia. Co-administration of nepicastat (15, 30, and 60 mg/kg) and enalapril (1 mg/kg) tended to exhibit slower heart rate than enalapril (1 mg/kg) alone. In any of the treatment groups, no significant effect on motor activity was detected.
  • Beagle dogs were administered single intraduodenal doses of 0 (vehicle) or 60 mg/kg of nepicastat through an intraduodenal cannula.
  • the vehicle-control group consisted of 1 male and 1 female
  • the nepicastat-treatment group consisted of 2 males and 2 females.
  • Each animal was surgically instrumented while anesthetized with isoflurane gas.
  • the average blood pressure responses to intravenous doses of autonomic agents, norepinephrine (3 ⁇ g/kg), isoproterenol (0.3 ⁇ g/kg), and acetylcholine (10 ⁇ g/kg) were evaluated.
  • a single bolus dose of test formulation was then administered to each animal and the blood pressure responses to the autonomic agents were evaluated approximately 1, 2, and 3 hours after dosing.
  • each dog was euthanatized and removed from the study.
  • the dog was selected because it is commonly used to evaluate the effects of test compounds on hemodynamic parameters. Beagle dogs were obtained from Marshall Farms, Inc., North Rose, N.Y. Each dog was identified uniquely by an ear tattoo applied by the vendor. The animals were acclimated to laboratory conditions at least 3 weeks before dosing. During the acclimation period, the general condition of each animal was evaluated and those considered healthy were used. The dogs were randomly assigned to treatment groups; males were assigned odd numbers and females were assigned even numbers.
  • the dogs were housed individually in stainless steel cages identified with the study number, animal number, and tattoo number. The room housing the dogs was environmentally controlled. The cages were cleaned daily and the animals were transferred into sanitized cages every other week. Purina Certified Canine Chow® was offered once daily and water was provided ad libitum.
  • a 60-mg/ml suspension was prepared by mixing nepicastat powder with vehicle.
  • the constituted 60-mg/ml nepicastat formulation retained potency for the duration of use.
  • aqueous solutions of norepinephrine (60 ⁇ g/ml), isoproterenol (6 ⁇ g/ml), and acetylcholine (200 ⁇ g/ml) were prepared in sterile water.
  • a vehicle-control group of 1 male and 1 female were administered 1 ml/kg of vehicle and a nepicastat-treatment group of 2 males and 2 females were administered 1 ml/kg of a 60 mg/ml nepicastat solution.
  • the total dose of nepicastat administered to each animal was 60 mg/kg.
  • Dose selection was based on data from two studies with nepicastat. In an acute toxicity study in dogs, a single oral dose of 400 mg/kg resulted in transient clinical signs of toxicity. In a 1-month study, doses of 5, 20, or 80 mg/kg were administered to dogs orally once daily. Clinical signs of toxicity were present at 80 mg/kg/day.
  • a single intraduodenal dose of vehicle or nepicastat formulation was administered directly into the duodenum through an intraduodenal cannula.
  • the intraduodenal route was selected because the oral route is a proposed clinical route of administration of nepicastat.
  • Dose volumes were calculated on the basis of individual body weights recorded before dosing (data not tabulated in this report).
  • the dog being evaluated was euthanatized by an overdose of sodium pentobarbital (300 mg/kg, IV) and removed from the study.
  • the dogs were surgically instrumented according to procedures described in the protocol. Food was withheld from the animals overnight before surgical instrumentation. Each animal being evaluated was initially anesthetized by injecting (IV) a mixture of ketamine (10 mg/kg) and diazepam (0.5 mg/kg). Each animal was placed on a surgical table on top of a circulating warm-water pad to maintain body temperature and mechanically ventilated throughout the experiment. A surgical plane of anesthesia was maintained with isoflurane gas (1.5% to 2% of tidal volume delivered in oxygen at a flow rate of approximately 1.5 L/minute). Rectal body temperature was monitored only for use in measuring blood gas levels and the data are not presented in this report. External needle electrodes were placed subcutaneously to monitor a standard limb lead II electrocardiogram (ECG) for assessing anesthesia.
  • ECG electrocardiogram
  • the left femoral vein was cannulated and the tip of the polyethylene tubing was advanced into the vena cava for administration of autonomic agents.
  • the left femoral artery was cannulated using a polyethylene tube filled with 50 U/ml of heparin-saline solution.
  • the tip of the arterial cannula was advanced into the thoracic aorta and coupled to an external pressure transducer and systolic and diastolic aortic pressure were recorded.
  • Arterial blood samples were withdrawn from the arterial cannula for blood pH, PCO 2 , and PO 2 analyses.
  • a midline laparotomy was performed and the duodenum was isolated just caudal to the pyloric sphincter.
  • a needle was inserted into the duodenum and the tip of a saline-filled cannula was advanced through the needle and into the lumen for test formulation administration.
  • the needle was withdrawn from the incision site, the cannula was anchored into position, the cannula's stopcock was exteriorized outside of the abdomen, and the skin of the abdominal incision was reapposed.
  • the autonomic agents norepinephrine (3 ⁇ g/kg), isoproterenol (0.3 ⁇ g/kg), and acetylcholine (10 ⁇ g/kg), were administered intravenously by bolus injection (over approximately 15 seconds) using the femoral vein cannula with approximately 10 minutes between each dose. Following each administration of an agent, the cannula was flushed with 3 ml of water. Administration of the agents was repeated approximately 20 minutes after the first administration of acetylcholine.
  • each animal was dosed with vehicle or nepicastat.
  • the dose volume was 1 ml/kg given as a bolus directly into the duodenum using the intraduodenal cannula.
  • the intraduodenal cannula was flushed with 3 ml of vehicle solution.
  • administration of the autonomic agents was repeated with approximately 10 minutes between administration of each agent.
  • Aortic blood pressure, heart rate, and ECG parameters were continuously recorded directly on a polygraph recorder.
  • the blood pH, PCO 2 , and PO 2 values from the blood gas analyzer were manually recorded onto the polygraph chart at the approximate time at which the blood samples were withdrawn.
  • Heart rate, ECG, and blood gas parameters were used only for assessing the level of anesthesia and the stability of the animal preparation; these data are not presented in this report.
  • Systolic, diastolic, and mean aortic blood pressures were evaluated just before administration (baseline) and at the time of peak response to each agent (maximum change from baseline). Systolic, diastolic, and mean aortic blood pressures, and blood pH, PCO 2 , and PO 2 were evaluated before dosing and approximately 50, 110, and 170 minutes after dosing with test formulation.
  • the responses to norepinephrine were characterized by evaluating the mean aortic blood pressure just before and at the time of peak pressure increase for each norepinephrine administration.
  • the responses to isoproterenol and acetylcholine were characterized by evaluating the diastolic aortic blood pressure just before and at the time of peak pressure decrease for each isoproterenol and acetylcholine administration.
  • each dog was euthanatized by an overdose of sodium pentobarbital (approximately 300 mg/kg, IV) and removed from the study.
  • Surgically instrumented, anesthetized beagle dogs were administered a single intraduodenal dose of 60 mg/kg of nepicastat.
  • Blood pressure responses to intravenous doses of autonomic agents were evaluated before dosing and approximately 1, 2, and 3 hours after dosing. No treatment-related differences between predose and postdose responses to autonomic agents were present.
  • mice (30-40 g on study day ) were housed in groups of eight under a normal light/dark cycle with lights on between 0900 hr and 2100 hr. Food and water were allowed ad libitum. All animals were naive to drug treatment and behavioral testing. Each animal was only used once.
  • Locomotor activity was monitored in an automated 14 station activity monitoring system (San Diego Instrument Co.). Each station consisted of a clear perspex cage (25 cm ⁇ 45 cm ⁇ 20 cm; w ⁇ l ⁇ h) placed within a metal frame containing 3 photoemmitors and 3 photodetectors spaced equally along the length of the wall. The bottom of each cage was lightly covered in clean cedar bedding.
  • mice were placed in the testing room at least 1 hr prior to testing.
  • the mice were placed individually into one of the activity cages and allowed to explore for 30 min.
  • the mice were dosed intraperitoneally with either nepicastat (10, 30 and 100 mg/kg), SKF-102698 (30 and 100 mg/kg), cocaine (30 mg/kg) or vehicle and returned immediately to same cage.
  • motor activity was monitored for 180 minutes.
  • Activity counts and ambulations (defined as a break of 2 consecutive photobeams) for each animal were recorded every 30 minutes.
  • a repeated measures two-way analysis of variance was performed using the overall ranked data (nonparametric technique) to test for the overall effects of treatment, time interval and treatment by time interval interaction. At each time interval, a one-way ANOVA was performed to see at which of the intervals, in any, treatment effects existed. Pairwise comparisons were then performed at each time interval using Dunn's procedure and Fisher's LSD strategy to adjust for the problem of multiple comparisons.
  • the dose range was 3-100 mg/kg and was dissolved in dH 2 O and sonicated.
  • the dose range was 30-100 mg/kg.
  • cocaine hydrochloride the dose was 30 mg/kg.
  • Compounds were administered in a volume of 1 ml/100 g. All doses reported are represented as the free base, except for cocaine in which the salt weight was used.
  • Cocaine was effectively demonstrated as a locomotor stimulant at the dose of 30 mg/kg.
  • acute administration of nepicastat at doses of 3, 10, 30 or 100 mg/kg did not cause any significant change in total activity or the ambulations at any time interval, as compared to vehicle control.
  • SKF-102698 at doses of 30 and 100 mg/kg had no significant effects on total activity or ambulations at any time interval examined.
  • Acute dosing with the dopamine-p-hydroxylase inhibitor nepicastat has been shown to inhibit the enzyme in the mesenteric artery and left ventricle in spontaneously hypertensive rats. Changes in norepinephrine and dopamine levels in the spontaneous hypertensive rat brain cortex and mesenteric artery after 7 and 25 days of oral administration of 1 mg/kg or 10 mg/kg nepicastat were examined.
  • Nepicastat at 1 and 10 mg/kg was prepared in terms of the free base. The weighings were dissolved in vehicle (dH 2 O) to yield oral doses that could be administered in a volume of 10.0 ml/kg.
  • SHRs Male spontaneous hypertensive rats
  • the 10 mg/kg dose group had significantly (p ⁇ 0O1) lower norepinephrine levels and a significantly (p ⁇ 0.05) higher dopamine/norepinephrine ratio compared to the vehicle group.
  • dopamine levels compared to vehicle in either of the two treatment groups (1 or 10 mg/kg nepicastat), or in the norepinephrine levels or the dopamine/norepinephrine ratio of the 1 mg/kg nepicastat dose group, after seven days of treatment (FIGS. 6 -8).
  • cortex levels of dopamine in the 1 mg/kg nepicastat dose group were significantly (p ⁇ 0.05) higher compared to the vehicle group.
  • the cortex dopamine/norepinephrine ratio in this group was also significantly (p ⁇ 0.01) greater than the vehicle ratio.
  • the ratio of the 10 mg/kg nepicastat dose group was significantly greater (p ⁇ 0.05) compared to vehicle. Norepinephrine levels in either dose group were not significantly (p.>0.05) different than control, nor were the dopamine levels in the 10 mg/kg dose group ( FIGS. 6-8 ).
  • the 10 mg/kg dose group had significantly higher dopamine levels and dopamine/norepinephrine ratios compared to the vehicle group, but there were no differences in norepinephrine levels. None of the parameters measured were significantly (p ⁇ 0.05) different than control in the 1 mg/kg nepicastat dose group ( FIGS. 9-11 ).
  • Nepicastat administered orally for 7 and 25 days, significantly (p ⁇ 0.05) inhibited dopamine- ⁇ -hydroxylase in the cortex and mesenteric artery of spontaneously hypertensive rats (SHRs). Greater inhibition was seen with administration of 10 mg/kg nepicastat compared to 1 mg/kg, therefore the effects observed were dose dependent.

Landscapes

  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Epidemiology (AREA)
  • Addiction (AREA)
  • Psychiatry (AREA)
  • Pain & Pain Management (AREA)
  • Otolaryngology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Hospice & Palliative Care (AREA)
  • Pulmonology (AREA)
  • Cardiology (AREA)
  • Urology & Nephrology (AREA)
  • Psychology (AREA)
  • Anesthesiology (AREA)
  • Vascular Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicinal Preparation (AREA)
US12/187,166 2007-08-06 2008-08-06 Methods for Treating Dependence Abandoned US20090041800A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US12/187,166 US20090041800A1 (en) 2007-08-06 2008-08-06 Methods for Treating Dependence
US14/099,882 US10561638B2 (en) 2007-08-06 2013-12-06 Methods for treating dependence
US16/736,609 US20200383951A1 (en) 2007-08-06 2020-01-07 Methods for treating dependence

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US93532307P 2007-08-06 2007-08-06
US95655507P 2007-08-17 2007-08-17
US96059107P 2007-10-04 2007-10-04
US12/187,166 US20090041800A1 (en) 2007-08-06 2008-08-06 Methods for Treating Dependence

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/099,882 Continuation US10561638B2 (en) 2007-08-06 2013-12-06 Methods for treating dependence

Publications (1)

Publication Number Publication Date
US20090041800A1 true US20090041800A1 (en) 2009-02-12

Family

ID=40341713

Family Applications (3)

Application Number Title Priority Date Filing Date
US12/187,166 Abandoned US20090041800A1 (en) 2007-08-06 2008-08-06 Methods for Treating Dependence
US14/099,882 Expired - Fee Related US10561638B2 (en) 2007-08-06 2013-12-06 Methods for treating dependence
US16/736,609 Abandoned US20200383951A1 (en) 2007-08-06 2020-01-07 Methods for treating dependence

Family Applications After (2)

Application Number Title Priority Date Filing Date
US14/099,882 Expired - Fee Related US10561638B2 (en) 2007-08-06 2013-12-06 Methods for treating dependence
US16/736,609 Abandoned US20200383951A1 (en) 2007-08-06 2020-01-07 Methods for treating dependence

Country Status (16)

Country Link
US (3) US20090041800A1 (es)
EP (2) EP2182804B1 (es)
JP (5) JP2010535801A (es)
CN (2) CN106983747A (es)
AU (1) AU2008283903B2 (es)
BR (1) BRPI0815089A2 (es)
CA (1) CA2695372C (es)
CO (1) CO6260015A2 (es)
ES (1) ES2638190T3 (es)
IL (1) IL257418A (es)
MX (3) MX2010001390A (es)
NZ (1) NZ583192A (es)
PH (1) PH12015502210A1 (es)
RU (1) RU2491067C2 (es)
SG (2) SG183696A1 (es)
WO (1) WO2009021055A1 (es)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140100791A1 (en) * 2012-10-10 2014-04-10 Bio-Rad Laboratories, Inc. Patient-based results display
US9192570B2 (en) 2013-12-20 2015-11-24 AntiOP, Inc. Intranasal naloxone compositions and methods of making and using same
CN113017632A (zh) * 2021-03-17 2021-06-25 陈思 一种智慧校园心理咨询辅助方法及系统
US11116723B2 (en) 2017-08-20 2021-09-14 Formulex Pharma Innovations Ltd. Dry powder compositions for intranasal delivery
WO2022040137A1 (en) * 2020-08-17 2022-02-24 The Trustess Of Columbia Univeristy In The City Of New York Use of neuromelanin-sensitive mri as a biomarker of dopamine function
US11806320B2 (en) * 2020-02-19 2023-11-07 Endo Ventures Limited Isoproterenol compositions and methods
US11844859B2 (en) 2017-08-20 2023-12-19 Nasus Pharma Ltd. Dry powder compositions for intranasal delivery

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2491067C2 (ru) * 2007-08-06 2013-08-27 Байотай Терапис, Инк Способы лечения зависимости
WO2010124089A2 (en) * 2009-04-22 2010-10-28 Synosia Therapeutics, Inc. Methods for treating dependence
CA2919892C (en) 2013-08-12 2019-06-18 Pharmaceutical Manufacturing Research Services, Inc. Extruded immediate release abuse deterrent pill
WO2015095391A1 (en) 2013-12-17 2015-06-25 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US9492444B2 (en) 2013-12-17 2016-11-15 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
EP3169315B1 (en) 2014-07-17 2020-06-24 Pharmaceutical Manufacturing Research Services, Inc. Immediate release abuse deterrent liquid fill dosage form
WO2016064873A1 (en) 2014-10-20 2016-04-28 Pharmaceutical Manufacturing Research Services, Inc. Extended release abuse deterrent liquid fill dosage form
US20170235908A1 (en) * 2015-11-15 2017-08-17 Oriah Behaviorial Health, Inc. Systems and Methods for Managing and Treating Substance Abuse Addiction
GB2571696B (en) 2017-10-09 2020-05-27 Compass Pathways Ltd Large scale method for the preparation of Psilocybin and formulations of Psilocybin so produced
CN108066355A (zh) * 2017-12-28 2018-05-25 宁夏恩多芬科技有限公司 Nitrous Oxide作为治疗神经官能症的用途
FR3093419B1 (fr) * 2019-03-08 2021-06-18 Univ Grenoble Alpes Composition et procédé associé de mesure de l’observance thérapeutique
WO2020212952A1 (en) 2019-04-17 2020-10-22 Compass Pathfinder Limited Treatment of depression and other various disorders with psilocybin
PL3844147T3 (pl) * 2019-11-07 2022-07-18 Small Pharma Ltd Związki chemiczne
AU2021276656A1 (en) 2020-05-19 2022-11-24 Cybin Irl Limited Deuterated tryptamine derivatives and methods of use
CN112690771B (zh) * 2020-12-09 2022-05-24 华南理工大学 一种利用线性回归模型的人脸视频心率检测方法

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5593367A (en) * 1995-08-18 1997-01-14 Deere & Company Switchable ignition and starter control system
US5958896A (en) * 1997-08-08 1999-09-28 The Mclean Hospital Cytidine-containing and cytosine-containing compounds as treatments for stimulant exposure
US20030040015A1 (en) * 2001-03-07 2003-02-27 Kwang-Soo Kim Methods and reagents for identifying compounds and mutations that modulate dopamine beta-hydroxylase activity
US20060058336A1 (en) * 2003-02-27 2006-03-16 Shigetada Nakanishi Pharmaceutical composition for treatment of drug dependence
US20100105748A1 (en) * 2007-03-16 2010-04-29 David Weinshenker Methods and compositions for treatment of drug addiction

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE91891T1 (de) * 1987-10-07 1993-08-15 Matrix Technologies Inc Pharmazeutische zusammensetzung fuer die behandlung der kokainsucht.
WO1997030977A1 (en) * 1996-02-23 1997-08-28 Pharm-Eco Laboratories, Incorporated Selective d1 dopamine receptor agonists and partial agonists/antagonists
US6541520B1 (en) * 1998-08-05 2003-04-01 Brookhaven Science Associates Treatment of addiction and addiction-related behavior
JP2004519461A (ja) * 2001-01-17 2004-07-02 ハイチアム,インコーポレイテッド アルコール依存症治療用薬剤の製造時におけるフルマゼニルの使用
SE0104388D0 (sv) * 2001-12-27 2001-12-27 Pharmacia Ab New formulation and use and manufacture thereof
RU2252756C1 (ru) * 2003-12-23 2005-05-27 Хохлов Александр Петрович Препарат против алкоголизма и наркомании, способ получения такого препарата
US20090054414A1 (en) * 2007-07-23 2009-02-26 Synosia Therapeutics Rufinamide for the Treatment of Post-Traumatic Stress Disorder
AU2008279169A1 (en) * 2007-07-23 2009-01-29 Biotie Therapies, Inc 4-hydroxy-4-methyl-piperidine-1-carboxylic acid (4-methoxy-7-morpholin-4-yl-benzothiazol-2-yl)-amide for the treatment of post-traumatic stress disorder
US20090054403A1 (en) * 2007-07-23 2009-02-26 Synosia Therapeutics Treatment of Post-Traumatic Stress Disorder
RU2491067C2 (ru) 2007-08-06 2013-08-27 Байотай Терапис, Инк Способы лечения зависимости
US20100243466A1 (en) 2007-11-26 2010-09-30 Bridgestone Corporation Copper-zinc alloy electroplating bath and plating method using the copper-zinc alloy electroplating bath

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5593367A (en) * 1995-08-18 1997-01-14 Deere & Company Switchable ignition and starter control system
US5958896A (en) * 1997-08-08 1999-09-28 The Mclean Hospital Cytidine-containing and cytosine-containing compounds as treatments for stimulant exposure
US20030040015A1 (en) * 2001-03-07 2003-02-27 Kwang-Soo Kim Methods and reagents for identifying compounds and mutations that modulate dopamine beta-hydroxylase activity
US20060058336A1 (en) * 2003-02-27 2006-03-16 Shigetada Nakanishi Pharmaceutical composition for treatment of drug dependence
US20100105748A1 (en) * 2007-03-16 2010-04-29 David Weinshenker Methods and compositions for treatment of drug addiction

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140100791A1 (en) * 2012-10-10 2014-04-10 Bio-Rad Laboratories, Inc. Patient-based results display
US9804149B2 (en) * 2012-10-10 2017-10-31 Bio-Rad Laboratories, Inc. Patient-based results display
US9192570B2 (en) 2013-12-20 2015-11-24 AntiOP, Inc. Intranasal naloxone compositions and methods of making and using same
US9289425B2 (en) 2013-12-20 2016-03-22 AntiOP, Inc. Intranasal naloxone compositions and methods of making and using same
US11116723B2 (en) 2017-08-20 2021-09-14 Formulex Pharma Innovations Ltd. Dry powder compositions for intranasal delivery
US11202757B2 (en) * 2017-08-20 2021-12-21 Formulex Pharma Innovations Ltd. Dry powder compositions for intranasal delivery
US11331270B2 (en) 2017-08-20 2022-05-17 Nasus Pharma Ltd. Dry powder compositions for intranasal delivery
US11844859B2 (en) 2017-08-20 2023-12-19 Nasus Pharma Ltd. Dry powder compositions for intranasal delivery
US11806320B2 (en) * 2020-02-19 2023-11-07 Endo Ventures Limited Isoproterenol compositions and methods
WO2022040137A1 (en) * 2020-08-17 2022-02-24 The Trustess Of Columbia Univeristy In The City Of New York Use of neuromelanin-sensitive mri as a biomarker of dopamine function
CN113017632A (zh) * 2021-03-17 2021-06-25 陈思 一种智慧校园心理咨询辅助方法及系统

Also Published As

Publication number Publication date
EP2182804A4 (en) 2010-12-22
CA2695372C (en) 2013-01-22
US10561638B2 (en) 2020-02-18
IL257418A (en) 2018-04-30
JP2018154645A (ja) 2018-10-04
ES2638190T3 (es) 2017-10-19
JP2014148551A (ja) 2014-08-21
SG10201705968RA (en) 2017-08-30
BRPI0815089A2 (pt) 2014-09-30
CN101815438A (zh) 2010-08-25
NZ583192A (en) 2012-06-29
US20140099336A1 (en) 2014-04-10
MX2010001390A (es) 2010-08-02
CN106983747A (zh) 2017-07-28
EP3251670A1 (en) 2017-12-06
RU2491067C2 (ru) 2013-08-27
RU2010108249A (ru) 2011-09-20
CA2695372A1 (en) 2009-02-12
EP2182804B1 (en) 2017-05-24
CO6260015A2 (es) 2011-03-22
US20200383951A1 (en) 2020-12-10
EP2182804A1 (en) 2010-05-12
MX2022001510A (es) 2022-08-11
JP2010535801A (ja) 2010-11-25
SG183696A1 (en) 2012-09-27
MX357542B (es) 2018-07-13
PH12015502210A1 (en) 2017-04-10
AU2008283903B2 (en) 2014-01-16
JP2016210798A (ja) 2016-12-15
JP2020114883A (ja) 2020-07-30
AU2008283903A1 (en) 2009-02-12
WO2009021055A1 (en) 2009-02-12

Similar Documents

Publication Publication Date Title
US20200383951A1 (en) Methods for treating dependence
US20090054403A1 (en) Treatment of Post-Traumatic Stress Disorder
WO2010124089A2 (en) Methods for treating dependence
Renner et al. Pharmacokinetics and pharmacodynamics in clinical use of scopolamine
AU2018203524B2 (en) Methods for treating dependence
US20230382885A1 (en) Enantiomeric entactogen compositions and their use
US20020016334A1 (en) Pharmaceutical composition for the treatment of attention deficit hyperactivity disorder (ADHD)
RU2469715C2 (ru) Композиции (-)-e-10-oh-nt и способы их синтеза и применения
MXPA02007003A (es) Metodo para la ansiedad.
AU2014202047A1 (en) Methods for treating dependence
CA3067688A1 (en) Use of mavoglurant in the reduction of alcohol use or in preventing relapse into alcohol use
Docherty Investigations of Functional Subtypes of α1-and α2-Adrenoceptor

Legal Events

Date Code Title Description
AS Assignment

Owner name: SYNOSIA THERAPEUTICS, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:WOIWODE, TOM;MORAN, MARK;PICKFORD, LESLEY;REEL/FRAME:021482/0768;SIGNING DATES FROM 20080814 TO 20080821

AS Assignment

Owner name: BIOTIE THERAPIES, INC., CALIFORNIA

Free format text: CHANGE OF NAME;ASSIGNOR:SYNOSIA THERAPUETICS, INC.;REEL/FRAME:026783/0854

Effective date: 20110817

AS Assignment

Owner name: BIOTIE THERAPIES, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:SYNOSIA THERAPEUTICS, INC.;REEL/FRAME:026869/0614

Effective date: 20110817

STCB Information on status: application discontinuation

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