Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
2.
Neuropharmacology ; 227: 109426, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36693562

ABSTRACT

The substantial challenges facing high and low dose psychedelic drug development to achieve regulatory approval have been documented in the scientific literature. These limitations have not deterred drug developers and social media from repeatedly misleading patients, the public and health professionals. Developing "micro doses" of psychedelics overcomes many of the scientific and regulatory challenges of high dose psychedelics. If micro-dosing could be shown to be efficacious and safe for long term use, it could be administered in the typical model for treatment of mental disorders. Such a model would be more cost effective than the high dose/intense psychotherapy model currently described and could be readily available to all individuals who need another medication option. Outpatient psychotherapeutic agents have a clear route for approval and would be unlikely to be burdened by the extensive Risks Evaluation and Mitigation Strategy needed for high dose use. There may be a different therapeutic role for both high and low dose psychedelic agents. This article is part of the Special Issue on "National Institutes of Health Psilocybin Research Speaker Series".


Subject(s)
Hallucinogens , Mental Disorders , Social Media , Humans , Hallucinogens/therapeutic use , Lysergic Acid Diethylamide/therapeutic use , Psilocybin/therapeutic use , Mental Disorders/drug therapy
3.
Front Pharmacol ; 12: 640241, 2021.
Article in English | MEDLINE | ID: mdl-33716753

ABSTRACT

Long term benefits following short-term administration of high psychedelic doses of serotonergic and dissociative hallucinogens, typified by psilocybin and ketamine respectively, support their potential as treatments for psychiatric conditions such as major depressive disorder. The high psychedelic doses induce perceptual experiences which are associated with therapeutic benefit. There have also been anecdotal reports of these drugs being used at what are colloquially referred to as "micro" doses to improve mood and cognitive function, although currently there are recognized limitations to their clinical and preclinical investigation. In the present studies we have defined a low dose and plasma exposure range in rats for both ketamine (0.3-3 mg/kg [10-73 ng/ml]) and psilocybin/psilocin (0.05-0.1 mg/kg [7-12 ng/ml]), based on studies which identified these as sub-threshold for the induction of behavioral stereotypies. Tests of efficacy were focused on depression-related endophenotypes of anhedonia, amotivation and cognitive dysfunction using low performing male Long Evans rats trained in two food motivated tasks: a progressive ratio (PR) and serial 5-choice (5-CSRT) task. Both acute doses of ketamine (1-3 mg/kg IP) and psilocybin (0.05-0.1 mg/kg SC) pretreatment increased break point for food (PR task), and improved attentional accuracy and a measure of impulsive action (5-CSRT task). In each case, effect size was modest and largely restricted to test subjects characterized as "low performing". Furthermore, both drugs showed a similar pattern of effect across both tests. The present studies provide a framework for the future study of ketamine and psilocybin at low doses and plasma exposures, and help to establish the use of these lower concentrations of serotonergic and dissociative hallucinogens both as a valid scientific construct, and as having a therapeutic utility.

4.
Prog Brain Res ; 259: 229-263, 2021.
Article in English | MEDLINE | ID: mdl-33541678

ABSTRACT

Nicotine use and dependence, typically achieved through cigarette smoking, but increasingly through vape products, is the leading cause of preventable death today. Despite a recognition that many current smokers would like to quit, the success rate at doing so is low and indicative of the persistent nature of nicotine dependence and the high urge to relapse. There are currently three main forms of pharmacotherapy approved as aids to treat nicotine dependence: a variety of nicotine replacement products (NRT's), the mixed NA/DA reuptake inhibitor bupropion (Zyban®), and the preferential nicotinic α4ß2 receptor agonist drug, varenicline (Chantix®); the latter being generally recognized to be the most effective. However, each of these approaches afford only limited efficacy, and various other pharmacological approaches are being explored. This chapter focusses on approaches targeted to the serotonin (5-HT) system, namely, selective serotonin reuptake inhibitors (SSRI's) which served a pioneer role in the investigation of serotoninergic modulators in human smoking cessation trials; and secondly drugs selectively interacting with the 5-HT2A and 5-HT2C receptor systems. From an efficacy perspective, measured as smoking abstinence, the 5-HT2A agonist psychedelics, namely psilocybin, seem to show the most promise; although as the article highlights, these findings are both preliminary and there are significant challenges to the route to approval, and therapeutic use of this class should they reach approval status. Additional avenues include 5-HT2C receptor agonists, which until recently was pioneered by lorcaserin, and 5-HT2A receptor antagonists represented by pimavanserin. Each of these approaches has distinct profiles across preclinical tests of nicotine dependence, and may have therapeutic potential. It is anticipated as diagnostic and predictive biomarkers emerge, they may provide opportunities for subject stratification and opportunities for personalizing smoking cessation treatment. The clinical assessment of SSRI, 5-HT2A and/or 5-HT2C receptor-based treatments may be best served by this process.


Subject(s)
Receptor, Serotonin, 5-HT2A , Receptor, Serotonin, 5-HT2C , Serotonin , Smoking Cessation , Tobacco Use Cessation Devices , Humans , Molecular Targeted Therapy , Nicotine , Varenicline
5.
J Clin Psychopharmacol ; 40(6): 568-578, 2020.
Article in English | MEDLINE | ID: mdl-33086237

ABSTRACT

BACKGROUND: Human abuse potential studies include multiple measures to assess the subjective effects of central nervous system-active drugs. In this retrospective analysis, measurement properties of commonly used measures were assessed, and factor analysis was conducted to identify a core battery of measures. METHODS: Measures of positive, negative and other effects, for example, bipolar "at-the-moment" Drug Liking visual analog scale (VAS), were derived for active controls and placebo from 19 studies in recreational drug users (N = 570). Distribution, placebo response, variability, convergent/discriminant validity, parameter effect sizes (eg, maximum effect [Emax], time-averaged area under the effect curve), and predictive validity were evaluated. A factor analysis was conducted with 9 studies. RESULTS: Most parameters were not normally distributed. Bipolar VAS exhibited the lowest variability. Drug Liking VAS Emax was very sensitive, showed large effect sizes (>1.0), and was moderately to strongly correlated with Emax of other positive effects measures (r > 0.5), but weaker with less specific scales (eg, high, Any Effects VAS); time-averaged area under the effect curve showed higher variability and lower effect sizes. Maximum effect at any dose (EmaxD) was significantly correlated with Emax across all selected measures and showed higher effect sizes. In the overall factor analysis, factors could be categorized into positive effects/euphoria (77% of variance), negative effects (17.9%), and pharmacologic effects (5%). For predictive validity, effect sizes for Drug Liking VAS Emax/EmaxD were moderately correlated with postmarket adverse events related to abuse (R = 0.52). CONCLUSIONS: A core battery of 7 subjective measures was proposed, with additional measures added based on pharmacologic effects.


Subject(s)
Behavior, Addictive/etiology , Central Nervous System Agents/adverse effects , Research Design , Substance-Related Disorders/etiology , Adult , Behavior, Addictive/diagnosis , Behavior, Addictive/psychology , Data Interpretation, Statistical , Factor Analysis, Statistical , Female , Humans , Male , Predictive Value of Tests , Psychiatric Status Rating Scales , Research Design/statistics & numerical data , Retrospective Studies , Risk Assessment , Risk Factors , Substance-Related Disorders/diagnosis , Substance-Related Disorders/psychology , Visual Analog Scale
6.
Epilepsy Behav ; 88: 162-171, 2018 11.
Article in English | MEDLINE | ID: mdl-30286443

ABSTRACT

RATIONALE: Treatment with a highly purified oral solution of cannabidiol (CBD), derived from the plant Cannabis sativa L., demonstrated some evidence of central nervous system (CNS)-related adverse events in patients enrolled in phase 3 trials for treatment of childhood-onset epilepsy. Cannabidiol was categorized as a Schedule 1 substance by the United States Drug Enforcement Administration; therefore, it was important to test CBD for human abuse potential. METHODS: This was a single-dose, randomized, double-blind, double-dummy, placebo- and active-controlled crossover trial. The abuse potential of single oral doses of plant-derived pharmaceutical formulations of highly purified CBD (Epidiolex®; 750 mg, 1500 mg, and 4500 mg) was compared with that of single oral doses of alprazolam (2 mg), dronabinol (10 mg and 30 mg), and placebo in healthy recreational polydrug users. The primary endpoint to assess abuse potential was the maximum effect (Emax) on Drug-Liking visual analog scale (VAS). Other measurements included Emax on Overall Drug-Liking VAS, Take Drug Again VAS, positive and negative effects, other subjective effects, and Drug Similarity VAS. Cognitive and psychomotor functions were assessed using the Divided Attention Test, the Hopkins Verbal Learning Test-Revised, and the Digit-Symbol Substitution Task. Pharmacokinetic parameters were determined for CBD and its major metabolites. Standard safety measures and adverse events were assessed. PRINCIPAL RESULTS: Of 95 eligible subjects, 43 qualified for the treatment phase, received at least 1 dose of investigational medicinal product, and were included in safety assessments; 35 subjects were included in the pharmacodynamic analysis. Subjects receiving alprazolam and dronabinol had significantly higher Drug-Liking Emax (P < 0.0001) compared with those receiving placebo, confirming study validity. Compared with placebo, Drug-Liking was not significantly different for subjects taking 750-mg CBD (P = 0.51). Drug-Liking Emax values for 1500-mg and 4500-mg CBD were significantly different from placebo (P = 0.04 and 0.002, respectively); however, the mean differences were <10 points on VAS compared with >18-point differences between positive controls and placebo. Alprazolam and dronabinol had significantly higher Drug-Liking, Overall-Liking, and Take Drug Again VAS Emax values compared with all doses of CBD (P ≤ 0.004). In contrast to alprazolam, CBD administration had no observable effect on cognitive/psychomotor tests. Pharmacokinetic parameters for CBD in this trial were consistent with previous studies. The majority of adverse events reported during the trial were of mild or moderate severity; no serious adverse events or deaths were reported. CONCLUSION: Administration of a therapeutic dose of CBD (750 mg) showed significantly low abuse potential in a highly sensitive population of polydrug users. Although high and supratherapeutic doses of CBD (1500 mg and 4500 mg, respectively) had detectable subjective effects compared with placebo; the effects were significantly lower than those observed with alprazolam and dronabinol.


Subject(s)
Cannabidiol/adverse effects , Illicit Drugs/adverse effects , Substance-Related Disorders/etiology , Adolescent , Adult , Cannabidiol/pharmacokinetics , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Illicit Drugs/pharmacokinetics , Male , Middle Aged , Risk Assessment , Substance-Related Disorders/prevention & control , Substance-Related Disorders/psychology , Young Adult
7.
Clin Pharmacol Ther ; 103(6): 924-935, 2018 06.
Article in English | MEDLINE | ID: mdl-29878319

ABSTRACT

The analgesic, sedative, antidepressant, euphoriant, intoxicating, and addictive properties of opium and its synthetic derivatives are well known and have been known for centuries. Hence, the current major public health problems due to excessive availability should be no surprise. What is unprecedented in the United States, and emerging elsewhere, is the extent of the profound consequences and complexity of addressing this public health crisis.


Subject(s)
Analgesics, Opioid/supply & distribution , Drug Overdose/epidemiology , Abuse-Deterrent Formulations/methods , Analgesics, Opioid/pharmacology , Analgesics, Opioid/therapeutic use , Chronic Pain/drug therapy , Comorbidity , Drug Discovery/organization & administration , Drug and Narcotic Control/organization & administration , Guideline Adherence , Health Personnel/education , Humans , Neonatal Abstinence Syndrome/epidemiology , Neonatal Abstinence Syndrome/therapy , Opioid-Related Disorders , Pharmacogenomic Testing/methods , Practice Guidelines as Topic , Public Health , United States/epidemiology
8.
J Clin Psychopharmacol ; 38(2): 113-118, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29360651

ABSTRACT

PURPOSE: This article discusses the conduct of a human abuse potential study as outlined in the Food and Drug Administration Final Guidance to Industry on Assessment of Abuse Potential. In addition, areas where alternative approaches should be considered are proposed. PROCEDURES: The design, end points, conduct, and interpretation of the human abuse potential study were reviewed, analyzed, and placed in the context of current scientific knowledge and best practices to mitigate regulatory risk and expedite drug development. FINDINGS: The guidance is based on regulatory needs and current scientific practices. However, the reliability and utility of such studies can be improved with better subject selection, data collection, standardization of data collection and staff training, and a better understanding of the measurement properties of the dependent measures. CONCLUSIONS: The guidance provides a useful framework for conduct of human abuse potential studies. However, design assumptions, poor choice of end points, failure to consider alternate approaches, and limited experience with interpretation can result in an inadequate study or one that does not fairly represent the abuse potential of a new chemical entity. Methodologic development is needed to strengthen the regulatory framework. The Food and Drug Administration or the National Institutes on Drug Abuse could take a targeted initiative to encourage this work.


Subject(s)
Drug Design , Drug Industry/standards , Guidelines as Topic/standards , Research Design/standards , Substance-Related Disorders , United States Food and Drug Administration/standards , Humans , United States
9.
Neuropharmacology ; 142: 116-134, 2018 11.
Article in English | MEDLINE | ID: mdl-29162429

ABSTRACT

Scientific curiosity and fascination have played a key role in human research with psychedelics along with the hope that perceptual alterations and heightened insight could benefit well-being and play a role in the treatment of various neuropsychiatric disorders. These motivations need to be tempered by a realistic assessment of the hurdles to be cleared for therapeutic use. Development of a psychedelic drug for treatment of a serious psychiatric disorder presents substantial although not insurmountable challenges. While the varied psychedelic agents described in this chapter share some properties, they have a range of pharmacologic effects that are reflected in the gradation in intensity of hallucinogenic effects from the classical agents to DMT, MDMA, ketamine, dextromethorphan and new drugs with activity in the serotonergic system. The common link seems to be serotonergic effects modulated by NMDA and other neurotransmitter effects. The range of hallucinogens suggest that they are distinct pharmacologic agents and will not be equally safe or effective in therapeutic targets. Newly synthesized specific and selective agents modeled on the legacy agents may be worth considering. Defining therapeutic targets that represent unmet medical need, addressing market and commercial issues, and finding treatment settings to safely test and use such drugs make the human testing of psychedelics not only interesting but also very challenging. This article is part of the Special Issue entitled 'Psychedelics: New Doors, Altered Perceptions'.


Subject(s)
Hallucinogens/pharmacology , Biomedical Research , Hallucinogens/adverse effects , Hallucinogens/pharmacokinetics , Hallucinogens/therapeutic use , Healthy Volunteers , Humans
10.
Epilepsy Behav ; 78: 194-201, 2018 01.
Article in English | MEDLINE | ID: mdl-29153631

ABSTRACT

BACKGROUND: Brivaracetam is a new antiepileptic drug indicated for adjunctive treatment of focal seizures in adults at a dose of 50-200mg/day taken in two equal doses. The objective of this study was to evaluate the abuse potential of brivaracetam compared with alprazolam (positive control), placebo, and levetiracetam. METHODS: This was a randomized, double-blind, triple-dummy, crossover study in healthy male and female recreational central nervous system (CNS) depressant users aged 18-55years, who could distinguish between the subjective effects of alprazolam 2mg and placebo. All participants received single doses of brivaracetam (50 [therapeutic dose], 200, 1000mg [supratherapeutic doses]), alprazolam (1.5, 3mg), placebo, and levetiracetam (4000mg) in random order each separated by 7-10days. Subjective Visual Analogue Scales (VAS) and Addiction Research Center Inventory (ARCI) scales were completed at intervals up to 24h postdose. Primary endpoints were Drug Liking (at this moment) VAS, Overall Drug Liking VAS, Feeling High VAS, and ARCI Pentobarbital Chlorpromazine Alcohol Group (PCAG, sedation) maximum effect (Emax). Maximum effect values on each scale were analyzed using a mixed-effect model (per protocol population, N=44). RESULTS: The maximum effect for both alprazolam doses was significantly greater versus placebo for six designated endpoints, confirming study validity. Drug Liking (at this moment) VAS Emax was significantly lower for brivaracetam 50mg than alprazolam (both doses); there were no significant differences between brivaracetam 200mg and alprazolam (both doses), and brivaracetam 1000mg and alprazolam 1.5mg. Brivaracetam 1000mg (supratherapeutic single dose) had significantly higher Drug Liking (at this moment) VAS Emax than alprazolam 3mg. Overall, Drug Liking VAS Emax for brivaracetam 50 and 200mg was not significantly different from alprazolam (both doses). Brivaracetam 1000mg had significantly higher Overall Drug Liking VAS Emax than alprazolam 1.5mg, but was not significantly different from alprazolam 3mg. Feeling High VAS Emax was lower versus alprazolam with brivaracetam 50 and 200mg, while brivaracetam 1000mg was comparable with alprazolam (both doses). Addiction Research Center Inventory PCAG Emax for brivaracetam (all doses) was significantly lower than alprazolam (both doses). On the secondary/supportive endpoints, compared with alprazolam, brivaracetam had fewer positive effects (ARCI Morphine Benzedrine Group [euphoria]; Good Drug Effects VAS [50mg]) and fewer negative effects (Bad Drug Effects VAS; ARCI Lysergic Acid Diethylamide [dysphoria]). Brivaracetam was not significantly different from alprazolam for Take Drug Again VAS (50, 200mg). For most endpoints, brivaracetam (50-200mg) was not significantly different from levetiracetam (4000mg). CONCLUSION: This study in healthy recreational CNS depressant users showed that single doses of brivaracetam 50mg (therapeutic single dose) had lower sedative, positive, and negative drug effects than alprazolam, while brivaracetam 200 and 1000mg (supratherapeutic single doses) were more similar to alprazolam. The subjective profile of brivaracetam appeared to be similar to that of levetiracetam, but further evaluation using a range of levetiracetam doses would be needed to confirm similar abuse potential.


Subject(s)
Alprazolam/pharmacology , Anticonvulsants/pharmacology , Central Nervous System Depressants/pharmacology , Hypnotics and Sedatives/pharmacology , Levetiracetam/pharmacology , Pyrrolidinones/pharmacology , Adolescent , Adult , Alprazolam/administration & dosage , Alprazolam/adverse effects , Anticonvulsants/administration & dosage , Anticonvulsants/adverse effects , Central Nervous System Depressants/administration & dosage , Central Nervous System Depressants/adverse effects , Cross-Over Studies , Double-Blind Method , Drug-Related Side Effects and Adverse Reactions , Euphoria , Female , Humans , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/adverse effects , Illicit Drugs/adverse effects , Levetiracetam/administration & dosage , Levetiracetam/adverse effects , Male , Middle Aged , Pyrrolidinones/administration & dosage , Pyrrolidinones/adverse effects , Young Adult
11.
Clin Pharmacol Drug Dev ; 7(6): 569-574, 2018 08.
Article in English | MEDLINE | ID: mdl-29024490

ABSTRACT

All drugs with central nervous system activity must undergo an assessment of their abuse potential, and these data must be included in a New Drug Application. Part of this assessment is an analysis of treatment-emergent adverse events that occur during clinical development. Using an iterative consensus strategy, we evaluated and grouped an available list of 213 flag terms for abuse potential from the Food and Drug Administration, into categories and assessed the relevance of the terms to primary abuse behavior. Consequences of abuse (28%) were most common, followed by diagnoses (19%), altered thoughts (18%), cognitive effects (10%), stimulation/anxiety (9%), central nervous system depression (9%), and mood elevation (1%). The vast majority of abuse potential-related terms reflects treatment-emergent adverse events, not behaviorally motivating features to abuse a drug. Almost 30% of terms are related to altered perception or altered cognition. These are serious consequences in the context of abusable psychoactive drugs. Only 20% of terms were rated as definitely or probably reflecting intrinsic behavioral reinforcing potential, and 30% were assessed as having weak predictive utility. Sponsors need to have an explicit strategy for collecting, interpreting, and analyzing abuse-related adverse event information completely and accurately.


Subject(s)
Central Nervous System Agents/adverse effects , Drug Development , Substance-Related Disorders , Humans , Safety , Substance-Related Disorders/etiology , Substance-Related Disorders/prevention & control , Substance-Related Disorders/psychology , United States , United States Food and Drug Administration
12.
J Clin Psychopharmacol ; 38(1): 11-18, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29215386

ABSTRACT

PURPOSE: This article brings to the attention of drug developers the Food and Drug Administration's (FDA's) recent final Guidance to Industry on Assessment of Abuse Potential and provides practical suggestions about compliance with the Guidance. PROCEDURES: The Guidance areas are reviewed, analyzed, and placed in the context of current scientific knowledge and best practices to mitigate regulatory risk. FINDINGS: The Guidance provides substantial new detail on what needs to be done at all stages of drug development for central nervous system-active drugs. However, because many psychopharmacologic agents have unique preclinical and clinical features, the plan for each agent needs to be not only carefully prepared but also reviewed and approved by the FDA. Examples are provided where assumptions about interpretation of the Guidance can delay development. CONCLUSIONS: If the expertise and experience needed for assessing abuse potential during drug development do not exist within a company, external preclinical and clinical expert should be involved. Consultation with the FDA is encouraged and important because the specific requirements for each drug will vary.


Subject(s)
Central Nervous System Agents/adverse effects , Drug Design , Drug Industry/legislation & jurisprudence , Risk Management/legislation & jurisprudence , Central Nervous System Agents/administration & dosage , Drug Industry/methods , Guideline Adherence , Guidelines as Topic , Humans , Substance-Related Disorders/prevention & control , United States , United States Food and Drug Administration
13.
Clin Pharmacol Ther ; 103(4): 561-564, 2018 04.
Article in English | MEDLINE | ID: mdl-28836272

ABSTRACT

Interest in the potential therapeutic benefits of psychedelic agents has recently increased. In addition to psilocybin, a wide variety of agents with psychedelic properties have been proposed and partially tested. However, the challenges of obtaining approval to market a restricted psychotomimetic agent are formidable.


Subject(s)
Drug Approval/methods , Drug Discovery , Hallucinogens , Risk Evaluation and Mitigation , Drug Discovery/ethics , Drug Discovery/methods , Hallucinogens/pharmacology , Hallucinogens/therapeutic use , Humans , Mental Disorders/drug therapy , Patient Selection
14.
J Clin Psychopharmacol ; 37(6): 675-683, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28926353

ABSTRACT

PURPOSE: This phase 1, randomized, double-blind, placebo- and active comparator-controlled crossover study assessed the abuse potential of the antiepileptic drug, lacosamide. METHODS: After a qualification phase, 38 healthy, recreational central nervous system-depressant users were randomized to treatment sequences comprising single oral therapeutic (200 mg) and supratherapeutic (800 mg) doses of lacosamide, alprazolam (1.5 and 3 mg), and placebo. Subjective effects were assessed for 24 hours following each dose using a range of scales, with a 5- to 9-day washout between treatments. FINDINGS: Mean subjective effects for 200 mg lacosamide were statistically similar to placebo and significantly lower than with alprazolam for most end points. Lacosamide 800 mg elicited transient, statistically significant positive effects compared with placebo, but also persistent Bad Drug Effects including statistically greater maximum effect (Emax) scores for Nausea and Dysphoria compared with other treatments (P < 0.0002). Consistent with this, the 800 mg lacosamide dose showed a significantly lower "at this moment" Drug Liking visual analog scale (VAS) Emax compared with 3 mg alprazolam, but was not different from 1.5 mg alprazolam (73.1/100, 85.4/100, and 78.9/100, respectively, where 50 is neutral). Overall Drug Liking VAS and Take Drug Again VAS Emax for 800 mg lacosamide were not significantly different from placebo and were lower than those for both alprazolam doses (P < 0.0001). IMPLICATIONS: These results suggest that in recreational central nervous system-depressant users, lacosamide has detectable abuse-related subjective effects, but a relatively low potential for abuse compared with alprazolam. These findings contributed toward placement of lacosamide into Schedule V of the US Controlled Substances Act.


Subject(s)
Acetamides/pharmacology , Alprazolam/pharmacology , Anticonvulsants/pharmacology , Central Nervous System Depressants/pharmacology , Drug-Related Side Effects and Adverse Reactions , Substance-Related Disorders , Acetamides/administration & dosage , Acetamides/adverse effects , Adult , Alprazolam/administration & dosage , Alprazolam/adverse effects , Anticonvulsants/administration & dosage , Anticonvulsants/adverse effects , Central Nervous System Depressants/administration & dosage , Central Nervous System Depressants/adverse effects , Cross-Over Studies , Double-Blind Method , Female , Humans , Lacosamide , Male , Middle Aged , Young Adult
15.
J Clin Psychopharmacol ; 36(4): 314-23, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27253658

ABSTRACT

Suvorexant is a dual orexin receptor antagonist approved in the United States and Japan for the treatment of insomnia at a maximum dose of 20 mg. This randomized double-blind crossover study evaluated the abuse potential of suvorexant in 36 healthy recreational polydrug users with a history of sedative and psychedelic drug use. Single doses of suvorexant (40, 80, and 150 mg: 2-7.5 × maximum dose), zolpidem (15 and 30 mg: 1.5-3 × maximum dose), and placebo were administered, with a 10-day washout between treatments. Subjective and objective measures, including visual analog scales (VASs), Addiction Research Center Inventory, and cognitive/psychomotor tests, were evaluated for 24-hour postdose. Suvorexant had significantly greater peak effects on "drug liking" VAS (primary endpoint) than placebo. Although effects of suvorexant on abuse potential measures were generally similar to zolpidem, they remained constant across doses, whereas zolpidem often had greater effects at higher doses. Suvorexant (all doses) had significantly fewer effects than zolpidem 30 mg on secondary measures, such as "high" VAS, Bowdle VAS, and Addiction Research Center Inventory morphine-benzedrine group. The overall incidence of abuse-related adverse events, such as euphoric mood and hallucination, was numerically lower with suvorexant than zolpidem. In agreement with its classification as a schedule IV drug, suvorexant demonstrated abuse potential, compared with placebo. The abuse potential was similar to zolpidem using certain measures, but with a reduced incidence of abuse-related adverse events. Although this suggests that the overall abuse liability of suvorexant may be lower than zolpidem, the actual abuse rates will be assessed with the postmarketing experience.


Subject(s)
Azepines/pharmacology , Euphoria/drug effects , Hallucinations/chemically induced , Hypnotics and Sedatives/pharmacology , Orexin Receptor Antagonists/pharmacology , Pyridines/pharmacology , Triazoles/pharmacology , Adult , Azepines/administration & dosage , Azepines/adverse effects , Cross-Over Studies , Double-Blind Method , Female , Humans , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/adverse effects , Illicit Drugs , Male , Middle Aged , Orexin Receptor Antagonists/administration & dosage , Orexin Receptor Antagonists/adverse effects , Prescription Drug Misuse , Pyridines/administration & dosage , Pyridines/adverse effects , Triazoles/administration & dosage , Triazoles/adverse effects , Zolpidem
16.
J Clin Psychopharmacol ; 35(4): 396-405, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26082975

ABSTRACT

UNLABELLED: Cocaine dependence presents a major public health issue, and to date, no pharmacotherapies are approved for its treatment. TV-1380 is a novel recombinant albumin-fused mutated butyrylcholinesterase (Albu-BChE) that has increased catalytic efficiency for cocaine compared with wild-type BChE and therefore has the potential to facilitate abstinence in cocaine-dependent subjects by decreasing exposure to cocaine and its reinforcing effects. METHODS: This randomized, double-blind, placebo-controlled, parallel-group study in nondependent cocaine users was conducted to evaluate the effect of a single intramuscular dose of Albu-BChE (50, 100, and 300 mg) on the pharmacokinetic and metabolic profile of intravenous cocaine infusions (40 mg) administered at baseline and at 24, 96, and 168 hours after Albu-BChE dosing, to assess safety of coadministering Albu-BChE and cocaine, and to explore the subjective responses to cocaine infusions after Albu-BChE dosing. RESULTS: Administration of Albu-BChE resulted in significant dose-dependent reductions in cocaine exposure (maximum concentration, area under the curve) and half-life. Effects were greatest at 24 hours after Albu-BChE dose, but were sustained up to 168 hours. Spearman correlations indicated a significant negative relationship between Albu-BChE concentration and cocaine clearance and exposure. Consistent with its mechanism of action, Albu-BChE also shifted cocaine metabolism toward preferential formation of ecgonine methyl ester. Administration of Albu-BChE was associated with modest decreases in subjective reports of feeling high and willingness to take cocaine again after cocaine infusion. Coadministration of Albu-BChE and cocaine was safe and well tolerated. CONCLUSIONS: Administration of Albu-BChE at single doses of 50, 100, and 300 mg safely resulted in long-lasting decreases in cocaine exposure in recreational cocaine users.


Subject(s)
Albumins/administration & dosage , Butyrylcholinesterase/administration & dosage , Butyrylcholinesterase/blood , Cocaine/administration & dosage , Cocaine/blood , Illicit Drugs/blood , Adolescent , Adult , Albumins/metabolism , Dose-Response Relationship, Drug , Double-Blind Method , Drug Interactions/physiology , Female , Humans , Injections, Intravenous , Male , Middle Aged , Young Adult
17.
J Clin Psychopharmacol ; 35(3): 242-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25928699

ABSTRACT

A novel clinical study design was used to evaluate the blockade of a selective short-acting µ-opioid agonist (remifentanil) in 24 opioid-experienced subjects. Samidorphan (3-carboxamido-4-hydroxynaltrexone) is a novel opioid modulator with µ-antagonist properties. Objective (pupil diameter) and subjective (visual analog scale) responses to repeated remifentanil and saline infusion challenges were assessed after single oral administration of placebo (day 1) and samidorphan (day 2). Complete blockade persisted with samidorphan for 24 hours for pupil miosis and 48 hours for the drug liking visual analog scale. Samidorphan effects persisted beyond measurable samidorphan exposure (t½ = 7 hours). Samidorphan was associated with complete blockade of remifentanil, and the duration supports daily administration. This study used a novel approach with multiple administrations of remifentanil to successfully demonstrate a durable effect with samidorphan and a rapid and potent blockade of physiological and subjective µ-opioid effects.


Subject(s)
Analgesics, Opioid/pharmacology , Naltrexone/analogs & derivatives , Narcotic Antagonists/pharmacology , Piperidines/pharmacology , Receptors, Opioid, mu/antagonists & inhibitors , Adult , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/antagonists & inhibitors , Analgesics, Opioid/pharmacokinetics , Double-Blind Method , Female , Humans , Male , Middle Aged , Miosis/chemically induced , Naltrexone/pharmacology , Piperidines/administration & dosage , Piperidines/antagonists & inhibitors , Piperidines/pharmacokinetics , Remifentanil , Visual Analog Scale , Young Adult
18.
19.
Neuropsychiatr Dis Treat ; 10: 1161-74, 2014.
Article in English | MEDLINE | ID: mdl-25061302

ABSTRACT

Pseudobulbar affect (PBA) is a common manifestation of brain pathology associated with many neurological diseases, including amyotrophic lateral sclerosis, Alzheimer's disease, stroke, multiple sclerosis, Parkinson's disease, and traumatic brain injury. PBA is defined by involuntary and uncontrollable expressed emotion that is exaggerated and inappropriate, and also incongruent with the underlying emotional state. Dextromethorphan/quinidine (DM/Q) is a combination product indicated for the treatment of PBA. The quinidine component of DM/Q inhibits the cytochrome P450 2D6-mediated metabolic conversion of dextromethorphan to its active metabolite dextrorphan, thereby increasing dextromethorphan systemic bioavailability and driving the pharmacology toward that of the parent drug and away from adverse effects of the dextrorphan metabolite. Three published efficacy and safety studies support the use of DM/Q in the treatment of PBA; significant effects were seen on the primary end point, the Center for Neurologic Study-Lability Scale, as well as secondary efficacy end points and quality of life. While concentration-effect relationships appear relatively weak for efficacy parameters, concentrations of DM/Q may have an impact on safety. Some special safety concerns exist with DM/Q, primarily because of the drug interaction and QT prolongation potential of the quinidine component. However, because concentrations of dextrorphan (which is responsible for many of the parent drug's side effects) and quinidine are lower than those observed in clinical practice with these drugs administered alone, some of the perceived safety issues may not be as relevant with this low dose combination product. However, since patients with PBA have a variety of other medical problems and are on numerous other medications, they may not tolerate DM/Q adverse effects, or may be at risk for drug interactions. Some caution is warranted when initiating DM/Q treatment, particularly in patients with underlying risk factors for torsade de pointes and in those receiving medications that may interact with DM/Q.

20.
Neuropharmacology ; 87: 81-90, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24793872

ABSTRACT

Psychoactive drugs that increase alertness, attention and concentration and energy, while also elevating mood, heart rate and blood pressure are referred to as stimulants. Despite some overlapping similarities, stimulants cannot be easily categorized by their chemical structure, mechanism of action, receptor binding profile, effects on monoamine uptake, behavioral pharmacology (e.g., effects on locomotion, temperature, and blood pressure), therapeutic indication or efficacy. Because of their abuse liability, a pre-market assessment of abuse potential is required for drugs that show stimulant properties; this review article focuses on the clinical aspects of this evaluation. This includes clinical trial adverse events, evidence of diversion or tampering, overdoses and the results of a human abuse potential study. While there are different types of human experimental studies that can be employed to evaluate stimulant abuse potential (e.g., drug discrimination, self-administration), only the human abuse potential study and clinical trial adverse event data are required for drug approval. The principal advances that have improved human abuse potential studies include using study enrichment strategies (pharmacologic qualification), larger sample sizes, better selection of endpoints and measurement strategies and more carefully considered interpretation of data. Because of the methodological advances, comparisons of newer studies with historical data is problematic and may contribute to a biased regulatory framework for the evaluation of newer stimulant-like drugs, such as A2 antagonists. This article is part of the Special Issue entitled 'CNS Stimulants'.


Subject(s)
Central Nervous System Stimulants/administration & dosage , Drug Evaluation/methods , Substance-Related Disorders , Clinical Trials as Topic , Humans , Self Administration , Substance-Related Disorders/diagnosis , Substance-Related Disorders/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...