Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Psychol Med ; 53(15): 7006-7024, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37671673

RESUMEN

Cannabis is well established to impact affective states, emotion and perceptual processing, primarily through its interactions with the endocannabinoid system. While cannabis use is quite prevalent in many individuals afflicted with psychiatric illnesses, there is considerable controversy as to whether cannabis may worsen these conditions or provide some form of therapeutic benefit. The development of pharmacological agents which interact with components of the endocannabinoid system in more localized and discrete ways then via phytocannabinoids found in cannabis, has allowed the investigation if direct targeting of the endocannabinoid system itself may represent a novel approach to treat psychiatric illness without the potential untoward side effects associated with cannabis. Herein we review the current body of literature regarding the various pharmacological tools that have been developed to target the endocannabinoid system, their impact in preclinical models of psychiatric illness and the recent data emerging of their utilization in clinical trials for psychiatric illnesses, with a specific focus on substance use disorders, trauma-related disorders, and autism. We highlight several candidate drugs which target endocannabinoid function, particularly inhibitors of endocannabinoid metabolism or modulators of cannabinoid receptor signaling, which have emerged as potential candidates for the treatment of psychiatric conditions, particularly substance use disorder, anxiety and trauma-related disorders and autism spectrum disorders. Although there needs to be ongoing clinical work to establish the potential utility of endocannabinoid-based drugs for the treatment of psychiatric illnesses, the current data available is quite promising and shows indications of several potential candidate diseases which may benefit from this approach.


Asunto(s)
Cannabis , Alucinógenos , Trastornos Mentales , Humanos , Endocannabinoides , Trastornos Mentales/tratamiento farmacológico , Ansiedad , Trastornos de Ansiedad , Agonistas de Receptores de Cannabinoides
2.
Artículo en Inglés | MEDLINE | ID: mdl-33677045

RESUMEN

The expanding legalization of cannabis across the United States is associated with increases in cannabis use, and accordingly, an increase in the number and severity of individuals with cannabis use disorder (CUD). The lack of FDA-approved pharmacotherapies and modest efficacy of psychotherapeutic interventions means that many of those who seek treatment for CUD relapse within the first few months. Consequently, there is a pressing need for innovative, evidence-based treatment development for CUD. Preliminary evidence suggests that repetitive transcranial magnetic stimulation (rTMS) may be a novel, non-invasive therapeutic neuromodulation tool for the treatment of a variety of substance use disorders (SUDs), including recently receiving FDA clearance (August 2020) for use as a smoking cessation aid in tobacco cigarette smokers. However, the potential of rTMS for CUD has not yet been reviewed. This paper provides a primer on therapeutic neuromodulation techniques for SUDs, with a particular focus on reviewing the current status of rTMS research in people who use cannabis. Lastly, future directions are proposed for rTMS treatment development in CUD, with suggestions for study design parameters and clinical endpoints based on current gold-standard practices for therapeutic neuromodulation research.


Asunto(s)
Encéfalo/fisiopatología , Abuso de Marihuana/terapia , Estimulación Magnética Transcraneal/métodos , Encéfalo/diagnóstico por imagen , Neuroimagen Funcional , Humanos , Abuso de Marihuana/diagnóstico por imagen , Abuso de Marihuana/fisiopatología , Resultado del Tratamiento
3.
Addict Biol ; 26(4): e12993, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33389797

RESUMEN

There are no FDA-approved treatments for cannabis use disorder (CUD). Preclinical research has shown that the 5HT-2C agonist lorcaserin attenuates cue-induced reinstatement of THC seeking and self-administration. The goal of this placebo-controlled, counterbalanced, within-subject human laboratory study was to examine lorcaserin's effects on cannabis intoxication and self-administration. Lorcaserin (10 mg BID) was administered during one of two 13-day inpatient phases and placebo during the other; each phase was separated by ≥7 days of washout. Inpatient phases comprised (1) standardized cannabis administration (7.0% THC) at no financial cost (intoxication), counterbalanced with (2) the option to self-administer cannabis following either 0 or 3 days of abstinence. Cognitive task performance, food intake, subjective ratings of drug effects, objective/subjective sleep measures, and tobacco cigarette use were also assessed. Fifteen normal-weight, daily cannabis users (4F, 11M) not seeking treatment for CUD completed the study. Lorcaserin significantly reduced cannabis self-administration following 0 and 3 days of cannabis abstinence and also reduced craving for cannabis during abstinence. Lorcaserin produced small but significant increases in positive cannabis ratings and body weight relative to placebo. Lorcaserin also reduced tobacco cigarette smoking on days of cannabis administration relative to placebo. During abstinence, subjective but not objective measures of sleep quality worsened during lorcaserin maintenance. Overall, lorcaserin's ability to decrease drug taking and cannabis craving in nontreatment-seeking cannabis users supports further investigation of 5HT-2C agonists as potential pharmacotherapies for CUD.


Asunto(s)
Benzazepinas/uso terapéutico , Abuso de Marihuana/tratamiento farmacológico , Fumar Marihuana/tratamiento farmacológico , Adulto , Afecto/efectos de los fármacos , Ansia/efectos de los fármacos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Autoadministración , Sueño/efectos de los fármacos , Calidad del Sueño , Adulto Joven
4.
Addict Biol ; 24(4): 765-776, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30378231

RESUMEN

Tobacco and cannabis co-users (T+CUs) have poor cannabis cessation outcomes, but the mechanisms underlying this are not well understood. This laboratory study examined the effects of (1) the partial nicotinic agonist, varenicline, on tobacco cessation among T+CUs, and (2) varenicline, alone, and when combined with the cannabinoid agonist nabilone, on cannabis withdrawal and a laboratory model of cannabis relapse. Non-treatment-seeking T+CUs were randomized to active-varenicline or placebo-varenicline, and completed a 15-day outpatient phase; varenicline was titrated to 1 mg BID during days 1-8, and participants were instructed to abstain from tobacco during days 9-15. Participants then moved inpatient for 16 days, where they continued their outpatient medication and tobacco abstinence. Inpatient testing included two, 8-day medication periods, where active-nabilone and placebo-nabilone were administered in counterbalanced order, and measures of acute cannabis effects (days 1-2), withdrawal (days 4-5) and 'relapse' (days 6-8) were collected. Participants in the active-varenicline group were more likely to achieve cotinine-verified tobacco abstinence during the outpatient period versus placebo-varenicline group (46 percent versus 24 percent, respectively), and also reported less mood disturbance and cigarette craving while inpatient. Active-nabilone attenuated cannabis withdrawal in both groups but did not affect cannabis relapse. Regression analyses revealed that two tobacco-related variables, i.e. age of first cigarette use, and cigarette craving while inpatient, were independent predictors of cannabis relapse outcomes. Thus, varenicline holds promise in this population, as a tool to examine the effects of tobacco abstinence on cannabis use outcomes, and as a component of smoking cessation treatments targeting T+CUs.


Asunto(s)
Fumar Cigarrillos/tratamiento farmacológico , Dronabinol/análogos & derivados , Abuso de Marihuana/tratamiento farmacológico , Agentes para el Cese del Hábito de Fumar/uso terapéutico , Cese del Hábito de Fumar , Síndrome de Abstinencia a Sustancias/fisiopatología , Vareniclina/uso terapéutico , Adulto , Fumar Cigarrillos/epidemiología , Comorbilidad , Dronabinol/uso terapéutico , Femenino , Humanos , Masculino , Abuso de Marihuana/epidemiología , Nicotina/efectos adversos , Agonistas Nicotínicos/efectos adversos , Síndrome de Abstinencia a Sustancias/etiología , Adulto Joven
5.
Addict Biol ; 24(4): 707-716, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-29659126

RESUMEN

The α2a-adrenergic agonist, lofexidine, reduced cannabis withdrawal-related sleep disruption in the laboratory, but side effects (e.g. fatigue, hypotension) limit its utility as a treatment for cannabis use disorder. This study tested the potential efficacy and tolerability of a daily bedtime administration of the FDA-approved α2a-adrenergic agonist, guanfacine, in a human laboratory model of cannabis use disorder. Daily, nontreatment-seeking cannabis smokers (13M, 2F) completed a within-subject study comprising two 9-day inpatient study phases. Each phase tested the effects of daily placebo or immediate-release guanfacine (2 mg) on cannabis intoxication (5.6 percent THC; 2 days), withdrawal (4 days of abstinence) and subsequent 'relapse' (3 days of cannabis self-administration). Ratings of mood, sleep, cardiovascular effects, food intake, psychomotor performance and cannabis self-administration were assessed. An outpatient phase preceded each inpatient phase for medication clearance or dose induction. Under placebo medication conditions, cannabis abstinence produced significant withdrawal, including irritability, sleep disruption and anorexia. Guanfacine reduced ratings of irritability and improved objective measures of sleep during cannabis withdrawal relative to placebo but did not reduce cannabis self-administration. Guanfacine was well tolerated with little evidence of fatigue and only small decreases in blood pressure: no dose was held due to hypotension. Thus, a single daily administration of guanfacine at bedtime improved sleep and mood during cannabis withdrawal relative to placebo. This positive signal supports further studies varying the guanfacine dose, formulation or frequency of administration, or combining it with other medications to increase the likelihood of having an impact on cannabis use.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/uso terapéutico , Guanfacina/uso terapéutico , Abuso de Marihuana , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Adulto , Afecto , Anorexia/etiología , Anorexia/fisiopatología , Presión Sanguínea , Cannabis/efectos adversos , Conducta Alimentaria , Femenino , Humanos , Genio Irritable , Masculino , Desempeño Psicomotor , Autoadministración , Sueño , Síndrome de Abstinencia a Sustancias/etiología , Síndrome de Abstinencia a Sustancias/fisiopatología , Síndrome de Abstinencia a Sustancias/psicología , Adulto Joven
6.
Cannabis Cannabinoid Res ; 4(2): 77-87, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32656342

RESUMEN

Introduction: Obsessive-compulsive disorder (OCD) is a disabling illness that is associated with significant functional impairment. Although evidence-based pharmacotherapies exist, currently available medications are ineffective in some patients and may cause intolerable side effects in others. There is an urgent need for new treatments. Discussion: A growing body of basic and clinical research has showed that the endocannabinoid system (ECS) plays a role in anxiety, fear, and repetitive behaviors. At the same time, some patients with OCD who smoke cannabis anecdotally report that it relieves their symptoms and mitigates anxiety, and several case reports describe patients whose OCD symptoms improved after they were treated with cannabinoids. Taken together, these findings suggest that the ECS could be a potential target for novel medications for OCD. In this study, we review evidence from both animal and human studies that suggests that the ECS may play a role in OCD and related disorders. We also describe findings from studies in which cannabinoid drugs were shown to impact symptoms of these conditions. Conclusions: An emerging body of evidence suggests that the ECS plays a role in OCD symptoms and may be a target for the development of novel medications. Further exploration of this topic through well-designed human trials is warranted.

7.
Psychopharmacology (Berl) ; 233(13): 2469-78, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27085870

RESUMEN

RATIONALE: Each year, over 300,000 individuals in the USA enter treatment for cannabis use disorder (CUD). The development of effective pharmacotherapy for CUD is a priority. OBJECTIVE: This placebo-controlled study examined the effects of zolpidem alone and in combination with nabilone on cannabis withdrawal and a laboratory measure of relapse. METHODS: Eleven daily, non-treatment-seeking cannabis users completed three, 8-day inpatient phases; each phase tested a different medication condition in counter-balanced order. On the first day of each phase, participants were administered placebo capsules t.i.d. and smoked experimenter-administered active cannabis (5.6 % Δ(9)-tetrahydrocannabinol (THC)). On days 2-8, the participants were administered capsules containing either placebo (0 mg at 0900, 1800, and 2300 hours), zolpidem (0 mg at 0900 and 1800, and 12.5 mg at 2300), or zolpidem (12.5 mg at 2300) and nabilone (3 mg at 0900 and 1800). Cannabis withdrawal, subjective capsule effects, and cognitive performance were examined on days 3-4, when only inactive cannabis (0.0 % THC) was available for self-administration. "Relapse" was measured on days 5-8, when participants could self-administer active cannabis. RESULTS: Both medication conditions decreased withdrawal-related disruptions in sleep, but only zolpidem in combination with nabilone decreased withdrawal-related disruptions in mood and food intake relative to placebo. Zolpidem in combination with nabilone, but not zolpidem alone, decreased self-administration of active cannabis. Zolpidem in combination with nabilone also produced small increases in certain abuse-related subjective capsule ratings, while zolpidem alone did not. Neither medication condition altered cognitive performance. CONCLUSIONS: Clinical testing of nabilone, either alone, or in combination with zolpidem is warranted.


Asunto(s)
Ansiolíticos/uso terapéutico , Dronabinol/análogos & derivados , Hipnóticos y Sedantes/farmacología , Abuso de Marihuana/tratamiento farmacológico , Piridinas/farmacología , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Adulto , Afecto/efectos de los fármacos , Cognición/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Dronabinol/farmacología , Quimioterapia Combinada , Ingestión de Alimentos/efectos de los fármacos , Femenino , Humanos , Masculino , Abuso de Marihuana/psicología , Fumar Marihuana/tratamiento farmacológico , Persona de Mediana Edad , Sueño/efectos de los fármacos , Síndrome de Abstinencia a Sustancias/psicología , Adulto Joven , Zolpidem
8.
Drug Alcohol Depend ; 154: 38-45, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26187456

RESUMEN

BACKGROUND: Evidence suggests that the cannabinoid system is involved in the maintenance of opioid dependence. We examined whether dronabinol, a cannabinoid receptor type 1 partial agonist, reduces opioid withdrawal and increases retention in treatment with extended release naltrexone (XR-naltrexone). METHODS: Opioid dependent participants were randomized to receive dronabinol 30mg/d (n=40) or placebo (n=20), under double-blind conditions, while they underwent inpatient detoxification and naltrexone induction. Before discharge all participants received an injection of XR-naltrexone, with an additional dose given four weeks later. Dronabinol or placebo was given while inpatient and for 5 weeks afterwards. The primary outcomes were the severity of opioid withdrawal, measured with the Subjective Opioid Withdrawal Scale, and retention in treatment at the end of the inpatient phase and at the end of the 8-week trial. RESULTS: The severity of opioid withdrawal during inpatient phase was lower in the dronabinol group relative to placebo group (p=0.006). Rates of successful induction onto XR-naltrexone (dronabinol 66%, placebo 55%) and completion of treatment (dronabinol 35%, placebo 35%) were not significantly different. Post hoc analysis showed that the 32% of participants who smoked marijuana regularly during the outpatient phase had significantly lower ratings of insomnia and anxiety and were more likely to complete the 8-week trial. CONCLUSION: Dronabinol reduced the severity of opiate withdrawal during acute detoxification but had no effect on rates of XR-naltrexone treatment induction and retention. Participants who elected to smoke marijuana during the trial were more likely to complete treatment regardless of treatment group assignment.


Asunto(s)
Preparaciones de Acción Retardada/uso terapéutico , Dronabinol/uso terapéutico , Naltrexona/administración & dosificación , Naltrexona/uso terapéutico , Trastornos Relacionados con Opioides/tratamiento farmacológico , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Adulto , Cannabinoides/uso terapéutico , Método Doble Ciego , Dronabinol/efectos adversos , Esquema de Medicación , Quimioterapia Combinada , Femenino , Humanos , Masculino , Antagonistas de Narcóticos/uso terapéutico , Cooperación del Paciente , Adulto Joven
9.
Neuropsychopharmacology ; 40(11): 2489-98, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25881117

RESUMEN

Given that cannabis use is increasing in the United States, pharmacological treatment options to treat cannabis use disorder are needed. Opioid antagonists modulate cannabinoid effects and may offer a potential approach to reducing cannabis use. In this double-blind, placebo-controlled human laboratory study, we assessed the effects of naltrexone maintenance on the reinforcing, subjective, psychomotor, and cardiovascular effects of active and inactive cannabis. Nontreatment-seeking, daily cannabis smokers were randomized to receive naltrexone (50 mg: n=18 M and 5 F) or placebo (0 mg; n=26 M and 2 F) capsules for 16 days. Before, during, and after medication maintenance, participants completed 10 laboratory sessions over 4-6 weeks, assessing cannabis' behavioral and cardiovascular effects. Medication compliance was verified by observed capsule administration, plasma naltrexone, and urinary riboflavin. Relative to placebo, maintenance on naltrexone significantly reduced both active cannabis self-administration and its positive subjective effects ('good effect'). Participants in the placebo group had 7.6 times (95% CI: 1.1-51.8) the odds of self-administering active cannabis compared with the naltrexone group. This attenuation of reinforcing and positive subjective effects also influenced cannabis use in the natural ecology. Naltrexone had intrinsic effects: decreasing ratings of friendliness, food intake, and systolic blood pressure, and increasing spontaneous reports of stomach upset and headache, yet dropout rates were comparable between groups. In summary, we show for the first time that maintenance on naltrexone decreased cannabis self-administration and ratings of 'good effect' in nontreatment-seeking daily cannabis smokers. Clinical studies in patients motivated to reduce their cannabis use are warranted to evaluate naltrexone's efficacy as a treatment for cannabis use disorder.


Asunto(s)
Fumar Marihuana/tratamiento farmacológico , Naltrexona/uso terapéutico , Antagonistas de Narcóticos/uso terapéutico , Adulto , Afecto/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Cannabis , Método Doble Ciego , Ingestión de Alimentos/efectos de los fármacos , Femenino , Humanos , Masculino , Fumar Marihuana/fisiopatología , Fumar Marihuana/psicología , Persona de Mediana Edad , Naltrexona/efectos adversos , Naltrexona/sangre , Antagonistas de Narcóticos/efectos adversos , Antagonistas de Narcóticos/sangre , Cooperación del Paciente , Distribución Aleatoria , Riboflavina/orina , Autoadministración , Adulto Joven
10.
Curr Addict Rep ; 1(2): 137-143, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24955304

RESUMEN

With large and increasing numbers of people using cannabis, the development of cannabis use disorder (CUD) is a growing public health concern. Despite the success of evidence-based psychosocial therapies, low rates of initial abstinence and high rates of relapse during and following treatment for CUD suggest a need for adjunct pharmacotherapies. Here we review the literature on medication development for the treatment of CUD, with a particular focus on studies published within the last three years (2010-2013). Studies in both the human laboratory and in the clinic have tested medications with a wide variety of mechanisms. In the laboratory, the following medication strategies have been shown to decrease cannabis withdrawal and self-administration following a period of abstinence (a model of relapse): the cannabinoid receptor agonist, nabilone, and the adrenergic agonist, lofexidine, alone and in combination with dronabinol (synthetic THC), supporting clinical testing of these medication strategies. Antidepressant, anxiolytic and antipsychotic drugs targeting monoamines (norepinephrine, dopamine, and serotonin) have generally failed to decrease withdrawal symptoms or laboratory measures of relapse. In terms of clinical trials, dronabinol and multiple antidepressants (fluoxetine, venlafaxine and buspirone) have failed to decrease cannabis use. Preliminary results from controlled clinical trials with gabapentin and N-acetylcysteine (NAC) support further research on these medication strategies. Data from open label and laboratory studies suggest lithium and oxytocin also warrant further testing. Overall, it is likely that different medications will be needed to target distinct aspects of problematic cannabis use: craving, ongoing use, withdrawal and relapse. Continued research is needed in preclinical, laboratory and clinical settings.

11.
Addict Biol ; 18(6): 993-1002, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22741619

RESUMEN

Marijuana withdrawal contributes to the high relapse rates in individuals seeking treatment for marijuana-use disorders. Quetiapine, an atypical antipsychotic, reduces characteristic symptoms of marijuana withdrawal in a variety of psychiatric conditions, including mood lability, sleep disruption and anorexia. This human laboratory study investigated the effectiveness of quetiapine to decrease marijuana withdrawal and relapse to marijuana use in non-treatment-seeking marijuana smokers. Volunteers were maintained on placebo or quetiapine (200 mg/day) in this double-blind, counter-balanced, within-subject study consisting of two 15-day medication phases, the last 8 days of which were in-patient. On the first in-patient day, active marijuana [6.2% delta (9)-tetrahydrocannabinol (THC)] was repeatedly smoked under controlled conditions. For the next 3 days, inactive marijuana (0.0% THC) was available for self-administration (withdrawal). On the subsequent 4 days, active marijuana (6.2% THC) was available for self-administration (relapse). Volunteers (n = 14) who smoked an average of 10 marijuana cigarettes/day, 7 days/week, completed the study. Under placebo, withdrawal was marked by increased subjective ratings of negative mood, decreased sleep quality, and decreased caloric intake and weight loss. Compared with placebo, quetiapine improved sleep quality, increased caloric intake and decreased weight loss. However, quetiapine increased marijuana craving and marijuana self-administration during the relapse phase. These data do not suggest that quetiapine shows promise as a potential treatment for marijuana dependence.


Asunto(s)
Anorexia/tratamiento farmacológico , Antipsicóticos/uso terapéutico , Dibenzotiazepinas/uso terapéutico , Dronabinol/efectos adversos , Trastornos del Sueño-Vigilia/tratamiento farmacológico , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Adulto , Afecto/efectos de los fármacos , Análisis de Varianza , Anorexia/inducido químicamente , Antipsicóticos/administración & dosificación , Antipsicóticos/farmacología , Dibenzotiazepinas/administración & dosificación , Dibenzotiazepinas/farmacología , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Femenino , Humanos , Masculino , Abuso de Marihuana/tratamiento farmacológico , Persona de Mediana Edad , Pruebas Neuropsicológicas/estadística & datos numéricos , Placebos , Desempeño Psicomotor/efectos de los fármacos , Fumarato de Quetiapina , Prevención Secundaria , Autoadministración/estadística & datos numéricos , Trastornos del Sueño-Vigilia/inducido químicamente , Pérdida de Peso/efectos de los fármacos , Adulto Joven
12.
Addict Biol ; 18(5): 872-81, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22260337

RESUMEN

Marijuana dependence is a substantial public health problem, with existing treatments showing limited efficacy. In laboratory and clinical studies, the cannabinoid receptor 1 agonist oral Δ9tetrahydrocannabinol (THC; dronabinol) has been shown to decrease marijuana withdrawal but not relapse. Dronabinol has poor bioavailability, potentially contributing to its failure to decrease relapse. The synthetic THC analogue, nabilone, has better bioavailability than dronabinol. We therefore aimed to characterize nabilone's behavioral and physiological effects across a range of acute doses in current marijuana smokers and compare these with dronabinol's effects. Participants (4 female; 10 male) smoking marijuana 6.6 (standard deviation = 0.7) days/week completed this outpatient, within-subjects, double-blind, randomized protocol. Over seven sessions, the time-dependent subjective, cognitive and cardiovascular effects of nabilone (2, 4, 6, 8 mg), dronabinol (10, 20 mg) and placebo were assessed. Nabilone (4, 6, 8 mg) and dronabinol (10, 20 mg) increased ratings of feeling a good effect, a strong effect and/or 'high' relative to placebo; nabilone had a slower onset of peak subjective effects than dronabinol. Nabilone (6, 8 mg) modestly lowered psychomotor speed relative to placebo and dronabinol. There were dose-dependent increases in heart rate after nabilone, and nabilone (2 mg) and dronabinol (10 mg) decreased systolic blood pressure. Thus, nabilone produced sustained, dose-related increases in positive mood, few cognitive decrements and lawful cardiovascular alterations. It had a longer time to peak effects than dronabinol, and effects were more dose-related, suggesting improved bioavailability. Nabilone was well tolerated by marijuana smokers, supporting further testing as a potential medication for marijuana dependence.


Asunto(s)
Agonistas de Receptores de Cannabinoides/uso terapéutico , Dronabinol/análogos & derivados , Dronabinol/uso terapéutico , Abuso de Marihuana/tratamiento farmacológico , Síndrome de Abstinencia a Sustancias/prevención & control , Adulto , Afecto/efectos de los fármacos , Análisis de Varianza , Disponibilidad Biológica , Presión Sanguínea/efectos de los fármacos , Agonistas de Receptores de Cannabinoides/farmacocinética , Agonistas de Receptores de Cannabinoides/farmacología , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Dronabinol/farmacocinética , Dronabinol/farmacología , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Placebos , Desempeño Psicomotor/efectos de los fármacos , Prevención Secundaria , Factores de Tiempo , Adulto Joven
13.
Psychopharmacology (Berl) ; 212(4): 675-86, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20824270

RESUMEN

RATIONALE: Dronabinol (Δ(9)tetrahydrocannabinol) is approved for HIV-related anorexia, yet, little is known about its effects in HIV-positive marijuana smokers. HIV-negative marijuana smokers require higher than recommended dronabinol doses to experience expected effects. OBJECTIVES: Employing a within-subjects, double-blind, placebo-controlled design, we assessed the effects of repeated high-dose dronabinol in HIV-positive marijuana smokers taking antiretroviral medication. METHODS: Participants (N = 7), who smoked marijuana 4.2 ± 2.3 days/week, resided in a residential laboratory for two 16-day stays, receiving dronabinol (10 mg QID) in one stay and placebo in the other. Efficacy was assessed with objectively verified food intake and body weight. Tolerability was measured with sleep, subjective, and cognitive assessments. For analyses, each inpatient stay was divided into two phases, days 1-8 and 9-16; we compared dronabinol's effects with placebo in each 8-day phase to investigate tolerance. RESULTS: Despite sustained increases in self-reported food cravings, dronabinol only increased caloric intake in the initial 8 days of dosing. Similarly, sleep quality was improved only in the first 8 days of dosing. Dronabinol's mood-enhancing effects were sustained across the 16-day inpatient stay. Dronabinol was well tolerated, causing few negative subjective or cognitive effects. CONCLUSIONS: In HIV-positive marijuana smokers, high dronabinol doses safely and effectively increased caloric intake. However, repeated high-dose dronabinol appeared to result in selective tolerance to these effects. These findings indicate that HIV-positive individuals who smoke marijuana may require higher dronabinol doses than are recommended by the FDA. Future research to establish optimal dosing regimens, and reduce the development of tolerance, is required.


Asunto(s)
Anorexia/tratamiento farmacológico , Estimulantes del Apetito/administración & dosificación , Dronabinol/administración & dosificación , Conducta Alimentaria/efectos de los fármacos , Infecciones por VIH/complicaciones , Fumar Marihuana/psicología , Adulto , Afecto/efectos de los fármacos , Anorexia/fisiopatología , Anorexia/psicología , Anorexia/virología , Estimulantes del Apetito/efectos adversos , Peso Corporal/efectos de los fármacos , Cognición/efectos de los fármacos , Método Doble Ciego , Dronabinol/efectos adversos , Tolerancia a Medicamentos , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Energía/efectos de los fármacos , Humanos , Masculino , Fumar Marihuana/fisiopatología , Ciudad de Nueva York , Efecto Placebo , Sueño/efectos de los fármacos , Conducta Social , Factores de Tiempo , Resultado del Tratamiento
14.
CNS Drugs ; 23(7): 543-53, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19552483

RESUMEN

Cannabis is the most widely used illicit drug in the world. Treatment admissions for cannabis use disorders have risen considerably in recent years, and the identification of medications that can be used to improve treatment outcomes among this population is a priority for researchers and clinicians. To date, several medications have been investigated for indications of clinically desirable effects among cannabis users (e.g. reduced withdrawal, attenuation of subjective or reinforcing effects, reduced relapse). Medications studied have included those: (i) known to be effective in the treatment of other drug use disorders; (ii) known to alleviate symptoms of cannabis withdrawal (e.g. dysphoric mood, irritability); or (iii) that directly affect endogenous cannabinoid receptor function. Results from controlled laboratory studies and small open-label clinical studies indicate that buspirone, dronabinol, fluoxetine, lithium and lofexidine may have therapeutic benefit for those seeking treatment for cannabis-related problems. However, controlled clinical trials have not been conducted and are needed to both confirm the potential clinical efficacy of these medications and to validate the laboratory models being used to study candidate medications. Although the recent increase in research towards the development of pharmacotherapy for cannabis use disorders has yielded promising leads, well controlled clinical trials are needed to support broad clinical use of these medications to treat cannabis use disorders.


Asunto(s)
Antipsicóticos/uso terapéutico , Ensayos Clínicos como Asunto , Abuso de Marihuana/tratamiento farmacológico , Serotoninérgicos/uso terapéutico , Humanos , Abuso de Marihuana/complicaciones
15.
Drug Alcohol Depend ; 102(1-3): 102-7, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19303723

RESUMEN

Studies using rodents have shown that behavioral responses to a stimulant are enhanced when the stimulant is given within the same context as previous stimulant administrations; this increase in effect related to context is often referred to as sensitization. We examined the role of environmental stimuli in modulating the subjective and cardiovascular effects of cocaine in humans (1) within a daily "binge" and (2) after cocaine abstinence. Ten non-treatment seeking users of smoked cocaine were admitted to the hospital for 17 consecutive days. Participants smoked cocaine (25mg/dose) under two counterbalanced conditions: paired stimuli (same stimuli presented each session) and unpaired stimuli (varied stimuli presented each session). Under each stimulus condition, participants had cocaine test sessions for three consecutive days, no sessions for the next 3 days, then another cocaine test session on the following day, for a total of eight test days. Stimulus condition had no effect on cardiovascular or subjective effects so data were analyzed as a function of repeated cocaine administration over 2 weeks. Maximal ratings on "good drug" and "drug rating" subjective effects clusters decreased over days of repeated cocaine exposure. In contrast, baseline and peak heart rate and systolic pressure increased over days of repeated cocaine administration. Thus, repeated administration of smoked cocaine to experienced cocaine users resulted in increases in baseline blood pressure and heart rate and modest decreases in positive subjective effects. These data indicate modest tolerance rather than sensitization to the positive subjective effects of cocaine with repeated exposure.


Asunto(s)
Trastornos Relacionados con Cocaína/fisiopatología , Trastornos Relacionados con Cocaína/psicología , Hemodinámica/efectos de los fármacos , Fumar/fisiopatología , Fumar/psicología , Estimulación Acústica , Administración por Inhalación , Adulto , Presión Sanguínea/efectos de los fármacos , Tolerancia a Medicamentos , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Estimulación Luminosa , Desempeño Psicomotor/efectos de los fármacos , Olfato , Encuestas y Cuestionarios
16.
Subst Abus ; 29(3): 17-29, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19042204

RESUMEN

Marijuana is the number one illicit drug of abuse worldwide and a major public health problem, especially in the younger population. The objective of this article is to update and review the state of the science and treatments available for marijuana dependence based on a pre-meeting workshop that was presented at ISAM 2006. At the workshop, several papers were presented addressing the neurobiology and pharmacology of marijuana and treatment approaches, both psychotherapy and medications, for marijuana withdrawal. Medicolegal and ethical issues concerning marijuana medical use were also discussed. Concise summaries of these presentations are incorporated in this article, which is meant to be an updated review of the state of the science. Major advances have been made in understanding the underpinning of marijuana dependence and the role of the CNS cannabinoid system, which is a major area for targeting medications to treat marijuana withdrawal and dependence, as well as other addictions. Behavioral therapies are efficacious for facilitating abstinence from marijuana. Nefazadone, Marinol, and buspirone are showing early positive signals for efficacy in ameliorating marijuana withdrawal symptoms. Effective psychotherapeutic approaches are available and promising medications studies need to be confirmed in outpatient trials. The next few years looking promising for translational research efforts to make treatment widely accessible to patients with marijuana dependence.


Asunto(s)
Encéfalo/efectos de los fármacos , Buspirona/uso terapéutico , Cannabis/efectos adversos , Dronabinol/uso terapéutico , Abuso de Marihuana/rehabilitación , Antagonistas de Narcóticos/uso terapéutico , Triazoles/uso terapéutico , Terapia Cognitivo-Conductual , Humanos , Abuso de Marihuana/terapia , Piperazinas , Facilitación Social
17.
Psychopharmacology (Berl) ; 197(1): 157-68, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18161012

RESUMEN

INTRODUCTION: Individuals seeking treatment for their marijuana use rarely achieve sustained abstinence. OBJECTIVES: The objectives of the study are to determine if THC, a cannabinoid agonist, and lofexidine, an alpha(2)-adrenergic receptor agonist, given alone and in combination, decreased symptoms of marijuana withdrawal and relapse, defined as a return to marijuana use after a period of abstinence. MATERIALS AND METHODS: Nontreatment-seeking, male volunteers (n = 8), averaging 12 marijuana cigarettes/day, were maintained on each of four medication conditions for 7 days: placebo, tetrahydrocannabinol (THC) (60 mg/day), lofexidine (2.4 mg/day), and THC (60 mg/day) combined with lofexidine (2.4 mg/day); each inpatient phase was separated by an outpatient washout phase. During the first three inpatient days, placebo marijuana was available for self-administration (withdrawal). For the next 4 days, active marijuana was available for self-administration (relapse). Participants paid for self-administered marijuana using study earnings. Self-administration, mood, task performance, food intake, and sleep were measured. RESULTS: THC reversed the anorexia and weight loss associated with marijuana withdrawal, and decreased a subset of withdrawal symptoms, but increased sleep onset latency, and did not decrease marijuana relapse. Lofexidine was sedating, worsened abstinence-related anorexia, and did not robustly attenuate withdrawal, but improved sleep and decreased marijuana relapse. The combination of lofexidine and THC produced the most robust improvements in sleep and decreased marijuana withdrawal, craving, and relapse in daily marijuana smokers relative to either medication alone. CONCLUSIONS: These data suggest the combination of lofexidine and THC warrant further testing as a potential treatment for marijuana dependence.


Asunto(s)
Agonistas alfa-Adrenérgicos/uso terapéutico , Cannabinoides/toxicidad , Clonidina/análogos & derivados , Dronabinol/uso terapéutico , Abuso de Marihuana/rehabilitación , Psicotrópicos/uso terapéutico , Síndrome de Abstinencia a Sustancias/rehabilitación , Agonistas alfa-Adrenérgicos/efectos adversos , Adulto , Afecto/efectos de los fármacos , Apetito/efectos de los fármacos , Atención/efectos de los fármacos , Clonidina/efectos adversos , Clonidina/uso terapéutico , Método Doble Ciego , Dronabinol/efectos adversos , Quimioterapia Combinada , Humanos , Masculino , Abuso de Marihuana/psicología , Recuerdo Mental/efectos de los fármacos , Desempeño Psicomotor/efectos de los fármacos , Psicotrópicos/efectos adversos , Prevención Secundaria , Sueño/efectos de los fármacos , Síndrome de Abstinencia a Sustancias/psicología
18.
Am J Drug Alcohol Abuse ; 32(4): 589-97, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17127547

RESUMEN

RATIONALE: Non-therapeutic research with drugs of abuse in humans is important for a more comprehensive understanding of substance abuse and for the development of more effective treatments. However, the administration of substances from drug classes with abuse potential to human volunteers raises ethical questions regarding potential risk to study volunteers. OBJECTIVE: The purpose of this study was to assess the psychosocial functioning and reported drug-taking behavior of volunteers before and after participating in a residential laboratory study, during which either marijuana, methamphetamine or zolpidem was administered. METHODS: Twenty-two volunteers were administered Addiction Severity Index (ASI) interviews at intake and approximately six months following their study participation. RESULTS: No significant differences between intake and follow-up assessments were found on any ASI composite or drug/alcohol-taking variable. CONCLUSION: These preliminary data suggest that participation in residential laboratory studies involving the administration of drugs from classes with abuse potential does not alter subsequent psychosocial functioning or reported drug use.


Asunto(s)
Hipnóticos y Sedantes/uso terapéutico , Abuso de Marihuana/psicología , Abuso de Marihuana/rehabilitación , Metanfetamina/uso terapéutico , Piridinas/uso terapéutico , Apoyo Social , Trastornos Relacionados con Sustancias/psicología , Trastornos Relacionados con Sustancias/rehabilitación , Alcoholismo/economía , Alcoholismo/psicología , Alcoholismo/rehabilitación , Costos y Análisis de Costo , Dopaminérgicos/economía , Dopaminérgicos/uso terapéutico , Humanos , Hipnóticos y Sedantes/economía , Abuso de Marihuana/economía , Metanfetamina/economía , New York , Piridinas/economía , Trastornos Relacionados con Sustancias/economía , Resultado del Tratamiento , Zolpidem
19.
Curr Psychiatry Rep ; 7(5): 360-6, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16216154

RESUMEN

A subset of marijuana smokers develop a cannabis use disorder and seek treatment for their marijuana use on their own initiative. A less well-known consequence of daily, repeated marijuana use is a withdrawal syndrome, characterized by a time-dependent constellation of symptoms: irritability, anxiety, marijuana craving, decreased quality and quantity of sleep, and decreased food intake. Treatment studies show that rates of continuous abstinence are low (comparable to relapse rates for other abused drugs), and more treatment options are needed. The objective of this review is to update clinicians on the current state of marijuana research and to describe features of marijuana withdrawal to facilitate the diagnosis and treatment of cannabis use disorders.


Asunto(s)
Cannabis/efectos adversos , Abuso de Marihuana/fisiopatología , Síndrome de Abstinencia a Sustancias/diagnóstico , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Ansiedad , Ingestión de Alimentos , Humanos , Genio Irritable , Trastornos del Sueño-Vigilia/etiología
20.
Neuropsychopharmacology ; 29(1): 158-70, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14560320

RESUMEN

Abstinence following daily marijuana use can produce a withdrawal syndrome characterized by negative mood (eg irritability, anxiety, misery), muscle pain, chills, and decreased food intake. Two placebo-controlled, within-subject studies investigated the effects of a cannabinoid agonist, delta-9-tetrahydrocannabinol (THC: Study 1), and a mood stabilizer, divalproex (Study 2), on symptoms of marijuana withdrawal. Participants (n=7/study), who were not seeking treatment for their marijuana use, reported smoking 6-10 marijuana cigarettes/day, 6-7 days/week. Study 1 was a 15-day in-patient, 5-day outpatient, 15-day in-patient design. During the in-patient phases, participants took oral THC capsules (0, 10 mg) five times/day, 1 h prior to smoking marijuana (0.00, 3.04% THC). Active and placebo marijuana were smoked on in-patient days 1-8, while only placebo marijuana was smoked on days 9-14, that is, marijuana abstinence. Placebo THC was administered each day, except during one of the abstinence phases (days 9-14), when active THC was given. Mood, psychomotor task performance, food intake, and sleep were measured. Oral THC administered during marijuana abstinence decreased ratings of 'anxious', 'miserable', 'trouble sleeping', 'chills', and marijuana craving, and reversed large decreases in food intake as compared to placebo, while producing no intoxication. Study 2 was a 58-day, outpatient/in-patient design. Participants were maintained on each divalproex dose (0, 1500 mg/day) for 29 days each. Each maintenance condition began with a 14-day outpatient phase for medication induction or clearance and continued with a 15-day in-patient phase. Divalproex decreased marijuana craving during abstinence, yet increased ratings of 'anxious', 'irritable', 'bad effect', and 'tired.' Divalproex worsened performance on psychomotor tasks, and increased food intake regardless of marijuana condition. Thus, oral THC decreased marijuana craving and withdrawal symptoms at a dose that was subjectively indistinguishable from placebo. Divalproex worsened mood and cognitive performance during marijuana abstinence. These data suggest that oral THC, but not divalproex, may be useful in the treatment of marijuana dependence.


Asunto(s)
Cannabis/efectos adversos , Dronabinol/uso terapéutico , Psicotrópicos/uso terapéutico , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Ácido Valproico/uso terapéutico , Adulto , Afecto/efectos de los fármacos , Análisis de Varianza , Anticonvulsivantes/uso terapéutico , Peso Corporal/efectos de los fármacos , Método Doble Ciego , Ingestión de Alimentos/efectos de los fármacos , Humanos , Masculino , Pruebas Neuropsicológicas , Desempeño Psicomotor/efectos de los fármacos , Sueño/efectos de los fármacos , Fumar/tratamiento farmacológico , Conducta Social , Encuestas y Cuestionarios , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA