Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros

Métodos Terapêuticos e Terapias MTCI
País/Região como assunto
Intervalo de ano de publicação
1.
Addict Biol ; 26(4): e12993, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33389797

RESUMO

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.


Assuntos
Benzazepinas/uso terapêutico , Abuso de Maconha/tratamento farmacológico , Fumar Maconha/tratamento farmacológico , Adulto , Afeto/efeitos dos fármacos , Fissura/efeitos dos fármacos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Autoadministração , Sono/efeitos dos fármacos , Qualidade do Sono , Adulto Jovem
2.
Drug Alcohol Depend ; 218: 108366, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33153828

RESUMO

BACKROUND: Pharmacotherapy for cannabis use disorder (CUD) is an important unmet public health need. METHODS: In a 12-week randomized double-blind placebo-controlled trial, the efficacy of quetiapine (300 mg nightly) for the treatment of CUD was tested in 130 outpatients. Weekly cannabis use was categorized into three groups: heavy use (5-7 days), moderate use (2-4 days) and light use (0-1 days). RESULTS: At baseline both groups were considered heavy users (using days per week: median = 7.0; interquartile range (IQR): 6.5-7.0; daily dollar value: median = $121.4; IQR: 73.8-206.3). The week-by-treatment interaction was marginally significant (χ2(2) = 5.56, P = .06). With each week, the odds of moderate compared to heavy use significantly increased in the quetiapine group (OR=1.17, P < .0001), but not significantly in the placebo group (OR=1.05, P = .16). The odds of light versus heavy use did not significantly differ over time (P = .12). Treatment was also associated with reduced cannabis withdrawal symptoms by 10.4% each week (95% CI: 8.9-11.8). No serious adverse events occurred during the study and no evidence of development of a movement disorder was detected. Adverse effects were not significantly different between the quetiapine and placebo treatment arms. CONCLUSIONS: The use of quetiapine to treat CUD was associated with an increased likelihood of heavy frequency use transitioning to moderate use, but not light use. The clinical significance of reductions in cannabis use, short of abstinence warrants further study.


Assuntos
Antipsicóticos/uso terapêutico , Abuso de Maconha/tratamento farmacológico , Fumarato de Quetiapina/uso terapêutico , Adulto , Cannabis , Método Duplo-Cego , Feminino , Alucinógenos/uso terapêutico , Humanos , Masculino , Fumar Maconha/tratamento farmacológico , Pessoa de Meia-Idade , Pacientes Ambulatoriais , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Resultado do Tratamento
3.
Nicotine Tob Res ; 22(8): 1374-1382, 2020 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-31612956

RESUMO

INTRODUCTION: The co-use of cannabis and alcohol among tobacco-using youth is common. Alcohol co-use is associated with worse tobacco cessation outcomes, but results are mixed regarding the impact of cannabis on tobacco outcomes and if co-use leads to increased use of non-treated substances. This secondary analysis from a youth smoking cessation trial aimed to (1) evaluate the impact of cannabis or alcohol co-use on smoking cessation, (2) examine changes in co-use during the trial, and (3) explore secondary effects of varenicline on co-use. METHODS: The parent study was a 12-week, randomized clinical trial of varenicline for smoking cessation among youth (ages 14-21, N = 157; Mage = 19, 40% female; 76% White). Daily cigarette, cannabis, and alcohol use data were collected via daily diaries during treatment and Timeline Follow-back for 14 weeks post-treatment. RESULTS: Baseline cannabis co-users (68%) had double the odds of continued cigarette smoking throughout the trial compared with noncannabis users, which was pronounced in males and frequent cannabis users. Continued smoking during treatment was associated with higher probability of concurrent cannabis use. Baseline alcohol co-users (80%) did not have worse smoking outcomes compared with nonalcohol users, but continued smoking was associated with higher probability of concurrent drinking. Varenicline did not affect co-use. CONCLUSIONS: Inconsistent with prior literature, results showed that alcohol co-users did not differ in smoking cessation, whereas cannabis co-users had poorer cessation outcomes. Youth tobacco treatment would benefit from added focus on substance co-use, particularly cannabis, but may need to be tailored appropriately to promote cessation. IMPLICATIONS: Among youth cigarette smokers enrolled in a pharmacotherapy evaluation clinical trial, alcohol and/or cannabis co-use was prevalent. The co-use of cannabis affected smoking cessation outcomes, but more so for males and frequent cannabis users, whereas alcohol co-use did not affect smoking cessation. Reductions in smoking were accompanied by concurrent reductions in alcohol or cannabis use. Substance co-use does not appear to affect all youth smokers in the same manner and treatment strategies may need to be tailored appropriately for those with lower odds of smoking cessation.


Assuntos
Consumo de Bebidas Alcoólicas/tratamento farmacológico , Fumar Maconha/tratamento farmacológico , Agentes de Cessação do Hábito de Fumar/uso terapêutico , Abandono do Hábito de Fumar/métodos , Vareniclina/uso terapêutico , Adolescente , Adulto , Consumo de Bebidas Alcoólicas/epidemiologia , Feminino , Humanos , Masculino , Fumar Maconha/epidemiologia , Prevalência , South Carolina/epidemiologia , Adulto Jovem
4.
Psychiatry Res ; 280: 112523, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31450032

RESUMO

Whilst the effects of cannabis preceding psychosis onset are well established, an effect post-onset is less clear. Emerging evidence suggests that cannabis use is associated with increased relapse outcomes possibly because of determinants, antipsychotic treatment failure and medication adherence, that are not mutually exclusive. Due to the paucity of literature on antipsychotic treatment failure an association with cannabis remains conjectural. This review sought to summarise current evidence regarding the effect of cannabis use on antipsychotic treatment failure among users and non-users with psychosis. Ovid databases (Embase, Journals@Ovid Full Text, OvidMEDLINE® In-Process and Other Non-Indexed Citations and PsycINFO) were searched to identify relevant articles. Seven articles met eligibility criteria. Cannabis use was associated with the following deleterious outcomes increased: odds of non-remission, prescription of unique antipsychotic medications, cumulative prescription of Clozapine and poor treatment trajectories. One study reported similar life-time, but lower past-year, rates of cannabis use in those prescribed Clozapine. Another study reported differences between groups for chlorpromazine equivalent doses for long-term Olanzapine prescription. Improved methodologies are warranted due to a lack of well-designed prospective studies and heterogeneity of key variables. There remains, despite research paucity, the need to encourage early cannabis cessation and higher-quality research to inform clinical practice.


Assuntos
Antipsicóticos/uso terapêutico , Fumar Maconha/tratamento farmacológico , Fumar Maconha/psicologia , Transtornos Psicóticos/tratamento farmacológico , Transtornos Psicóticos/psicologia , Adulto , Estudos de Casos e Controles , Comorbidade , Feminino , Humanos , Masculino , Abuso de Maconha/tratamento farmacológico , Abuso de Maconha/epidemiologia , Abuso de Maconha/psicologia , Fumar Maconha/epidemiologia , Adesão à Medicação , Estudos Observacionais como Assunto/métodos , Estudos Prospectivos , Transtornos Psicóticos/epidemiologia , Ensaios Clínicos Controlados Aleatórios como Assunto/métodos , Falha de Tratamento
5.
Am J Addict ; 27(2): 101-107, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29457671

RESUMO

BACKGROUND AND OBJECTIVE: Many patients with cannabis use disorder (CUD) do not achieve or do not have abstinence as a goal of treatment, rather they reduce their use. Assessing outcome measures as they relate to functioning and reductions in cannabis use is an important area of study. Quality of life (QoL) shows promise as one such measure. Past studies have demonstrated gender differences in QoL and CUD. We aim to assess (1) the relationship between cannabis use and QoL and (2) gender effects in an outpatient medication treatment study for CUD. METHODS: Data from an 11-weeks, double-blind, placebo-controlled trial of lofexidine and dronabinol for CUD (n = 62) was analyzed. Pearson's correlations between baseline QoL as measured with the Quality of Life, Enjoyment, and Satisfaction Questionnaire-Short Form (QLES-Q-SF) and cannabis use assessed with modified timeline follow-back (TLFB) were examined. Multiple linear regression models of cannabis use on end of study QLES-Q-SF were analyzed, while adjusting for baseline QLES-Q-SF, study arm, and gender. Moderation effects with gender were also tested. RESULTS: No significant association between baseline cannabis use and QoL was found. End of study abstinence (F1,47 = 8.34, p = .006) and reduced proportion of using days (F1,47 = 9.48, p = .004) were each significantly associated with end of study QoL. Reduction in grams (F1,27 = 0.25, p = .62) was not associated with QoL at end of study. Gender was not a significant moderator. DISCUSSION AND CONCLUSIONS: Abstinence and lower frequency of use are associated with higher QoL, regardless of gender. SCIENTIFIC SIGNIFICANCE: This is the first time QoL has been demonstrated to change over the course of CUD medication treatment. QoL is an important outcome in CUD treatment. TRIAL REGISTRATION: NCT01020019. (Am J Addict 2018;27:101-107).


Assuntos
Clonidina/análogos & derivados , Dronabinol/administração & dosagem , Fumar Maconha , Qualidade de Vida , Redução do Consumo de Tabaco , Adulto , Clonidina/administração & dosagem , Método Duplo-Cego , Feminino , Humanos , Masculino , Abuso de Maconha/tratamento farmacológico , Abuso de Maconha/psicologia , Fumar Maconha/tratamento farmacológico , Fumar Maconha/psicologia , Pessoa de Meia-Idade , Motivação , Antagonistas de Entorpecentes/administração & dosagem , Fatores Sexuais , Redução do Consumo de Tabaco/métodos , Redução do Consumo de Tabaco/psicologia , Inquéritos e Questionários , Temperança , Resultado do Tratamento
6.
J Psychopharmacol ; 32(3): 353-356, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29039260

RESUMO

Cannabis use disorders are frequently comorbid in patients with a psychotic disorder and are associated with worse outcomes. To date there are no proven effective strategies to achieve cannabis abstinence in this population. An alternative for abstinence might be harm reduction, i.e. replacing the use of street cannabis with high tetrahydrocannabinol and low cannabidiol levels by medicinal cannabis variants with relatively low tetrahydrocannabinol and relatively high cannabidiol levels, thereby reducing the psychosis inducing effects of cannabis and enhancing the antipsychotic effects of cannabis. Here we present the data of a case series with seven inpatients diagnosed with a psychotic disorder and a treatment-resistant cannabis use disorder who received substitution therapy with a low tetrahydrocannabinol medicinal cannabis variant (Bedrolite). The results suggest that the low tetrahydrocannabinol medicinal cannabis variant Bedrolite is not effective in the treatment of inpatients with a psychotic disorder and comorbid cannabis use disorder. Bedrolite is thus not very likely to become an effective harm reduction strategy in these patients.


Assuntos
Canabidiol/uso terapêutico , Abuso de Maconha/tratamento farmacológico , Fumar Maconha/tratamento farmacológico , Maconha Medicinal/uso terapêutico , Transtornos Psicóticos/tratamento farmacológico , Adulto , Antipsicóticos , Cannabis , Comorbidade , Dronabinol/uso terapêutico , Feminino , Humanos , Pacientes Internados , Masculino , Pessoa de Meia-Idade , Adulto Jovem
7.
Drug Alcohol Depend ; 177: 249-257, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28623823

RESUMO

BACKGROUND: Cannabis use disorder (CUD) is a prevalent and impairing condition, and established psychosocial treatments convey limited efficacy. In light of recent findings supporting the efficacy of N-acetylcysteine (NAC) for CUD in adolescents, the objective of this trial was to evaluate its efficacy in adults. METHODS: In a 12-week double-blind randomized placebo-controlled trial, treatment-seeking adults ages 18-50 with CUD (N=302), enrolled across six National Drug Abuse Treatment Clinical Trials Network-affiliated clinical sites, were randomized in a 1:1 ratio to a 12-week course of NAC 1200mg (n=153) or placebo (n=149) twice daily. All participants received contingency management (CM) and medical management. The primary efficacy measure was the odds of negative urine cannabinoid tests during treatment, compared between NAC and placebo participants. RESULTS: There was not statistically significant evidence that the NAC and placebo groups differed in cannabis abstinence (odds ratio=1.00, 95% confidence interval 0.63-1.59, p=0.984). Overall, 22.3% of urine cannabinoid tests in the NAC group were negative, compared with 22.4% in the placebo group. Many participants were medication non-adherent; exploratory analysis within medication-adherent subgroups revealed no significant differential abstinence outcomes by treatment group. CONCLUSIONS: In contrast with prior findings in adolescents, there is no evidence that NAC 1200mg twice daily plus CM is differentially efficacious for CUD in adults when compared to placebo plus CM. This discrepant finding between adolescents and adults with CUD may have been influenced by differences in development, cannabis use profiles, responses to embedded behavioral treatment, medication adherence, and other factors.


Assuntos
Acetilcisteína/uso terapêutico , Abuso de Maconha/diagnóstico , Abuso de Maconha/tratamento farmacológico , Adolescente , Adulto , Cannabis , Método Duplo-Cego , Feminino , Sequestradores de Radicais Livres/uso terapêutico , Humanos , Masculino , Abuso de Maconha/psicologia , Fumar Maconha/tratamento farmacológico , Fumar Maconha/psicologia , Adesão à Medicação/psicologia , Sulpirida , Resultado do Tratamento , Adulto Jovem
8.
Psychopharmacology (Berl) ; 233(13): 2469-78, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27085870

RESUMO

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.


Assuntos
Ansiolíticos/uso terapêutico , Dronabinol/análogos & derivados , Hipnóticos e Sedativos/farmacologia , Abuso de Maconha/tratamento farmacológico , Piridinas/farmacologia , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Adulto , Afeto/efeitos dos fármacos , Cognição/efeitos dos fármacos , Relação Dose-Resposta a Droga , Método Duplo-Cego , Dronabinol/farmacologia , Quimioterapia Combinada , Ingestão de Alimentos/efeitos dos fármacos , Feminino , Humanos , Masculino , Abuso de Maconha/psicologia , Fumar Maconha/tratamento farmacológico , Pessoa de Meia-Idade , Sono/efeitos dos fármacos , Síndrome de Abstinência a Substâncias/psicologia , Adulto Jovem , Zolpidem
10.
Neuropsychopharmacology ; 40(11): 2489-98, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25881117

RESUMO

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.


Assuntos
Fumar Maconha/tratamento farmacológico , Naltrexona/uso terapêutico , Antagonistas de Entorpecentes/uso terapêutico , Adulto , Afeto/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Cannabis , Método Duplo-Cego , Ingestão de Alimentos/efeitos dos fármacos , Feminino , Humanos , Masculino , Fumar Maconha/fisiopatologia , Fumar Maconha/psicologia , Pessoa de Meia-Idade , Naltrexona/efeitos adversos , Naltrexona/sangue , Antagonistas de Entorpecentes/efeitos adversos , Antagonistas de Entorpecentes/sangue , Cooperação do Paciente , Distribuição Aleatória , Riboflavina/urina , Autoadministração , Adulto Jovem
11.
Psychopharmacology (Berl) ; 232(2): 343-53, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24998257

RESUMO

RATIONALE: One of the most often reported cognitive deficits of acute cannabis administration is an impaired recall of previously learned information. OBJECTIVE: The aim of the present study was to determine whether cannabis-induced memory impairment in humans is mediated via glutamatergic or cholinergic pathways. METHODS: Fifteen occasional cannabis users participated in a double-blind, placebo-controlled, six-way cross-over study. On separate test days, subjects received combinations of pretreatment (placebo, vardenafil 20 mg or rivastigmine 3 mg) and treatment (placebo or 1,376 mg cannabis/kg body weight). Cognitive tests were administered immediately after inhalation of treatment was finished and included measures of memory (visual verbal learning task, prospective memory test, Sternberg memory test), perceptual-motor control (critical tracking task), attention (divided attention task) and motor impulsivity (stop signal task). RESULTS: The results of this study demonstrate that subjects under the influence of cannabis were impaired in all memory tasks, in critical tracking, divided attention and the stop signal task. Pretreatment with rivastigmine attenuated the effect of cannabis on delayed recall and showed a trend towards significance on immediate recall. When cannabis was given in combination with vardenafil, there were no significant interaction effects in any of the tasks. CONCLUSIONS: The present data therefore suggest that acetylcholine plays an important role in cannabis-induced memory impairment, whereas similar results for glutamate have not been demonstrated in this study.


Assuntos
Cannabis , Imidazóis/uso terapêutico , Fumar Maconha/efeitos adversos , Fumar Maconha/metabolismo , Transtornos da Memória/etiologia , Transtornos da Memória/metabolismo , Fenilcarbamatos/uso terapêutico , Piperazinas/uso terapêutico , Acetilcolina/metabolismo , Adulto , Atenção/efeitos dos fármacos , Canabinoides/administração & dosagem , Canabinoides/sangue , Cognição/efeitos dos fármacos , Estudos Cross-Over , Método Duplo-Cego , Feminino , Ácido Glutâmico/metabolismo , Humanos , Masculino , Fumar Maconha/tratamento farmacológico , Fumar Maconha/psicologia , Memória/efeitos dos fármacos , Transtornos da Memória/tratamento farmacológico , Memória de Curto Prazo/efeitos dos fármacos , Estudos Prospectivos , Rivastigmina , Sulfonas/uso terapêutico , Triazinas/uso terapêutico , Dicloridrato de Vardenafila , Aprendizagem Verbal/efeitos dos fármacos , Adulto Jovem
12.
Drug Alcohol Depend ; 144: 42-6, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25283697

RESUMO

BACKGROUND AND AIMS: Cannabis-dependent participants with depressive disorder are less likely to achieve abstinence with venlafaxine-XR (VEN-XR) treatment. Individuals on VEN-XR reported more severe withdrawal, despite not reducing their smoking behavior. We hypothesized that withdrawal-like symptoms, likely medication side effects, led to continued marijuana smoking in this group. METHODS: We conducted a secondary analysis using Marijuana Withdrawal Checklist (MWC) scores and urine THC to test whether severity of withdrawal-like symptoms mediates the relationship between VEN-XR treatment and continued marijuana smoking. We included 103 participants (VEN-XR=51, Placebo=52). Marijuana use was dichotomized into smoking (THC>100 ng/ml) and non-smoking (THC ≤ 100 ng/ml) weeks. MWC scores were obtained weekly. We used three models in a regression based mediation analysis. RESULTS: The estimated risk of smoking marijuana was greater for individuals on VEN-XR in weeks 7-9, even when controlling for MWC scores (week 7 Risk Difference (RD)=0.11, p=0.034; week 8 RD=0.20, p=0.014), and higher scores mediated this effect. In weeks 10 and 11, the estimated effect was stronger (week 10 RD=0.03, p=0.380; week 11 RD=0.07, p=0.504), and worse withdrawal-like symptoms more fully accounted for continued marijuana smoking in the VEN-XR group, according to the models. CONCLUSIONS: Individuals treated with VEN-XR had more severe withdrawal-like symptoms, which mediated their continued marijuana smoking. Noradrenergic agents, such as VEN-XR, may negatively impact treatment outcomes in cannabis-dependent patients attempting to reduce or stop their use.


Assuntos
Antidepressivos de Segunda Geração/uso terapêutico , Cicloexanóis/uso terapêutico , Transtorno Depressivo/tratamento farmacológico , Fumar Maconha/tratamento farmacológico , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Adolescente , Adulto , Transtorno Depressivo/diagnóstico , Transtorno Depressivo/psicologia , Método Duplo-Cego , Feminino , Humanos , Masculino , Fumar Maconha/psicologia , Pessoa de Meia-Idade , Síndrome de Abstinência a Substâncias/complicações , Síndrome de Abstinência a Substâncias/psicologia , Resultado do Tratamento , Cloridrato de Venlafaxina
13.
Am J Drug Alcohol Abuse ; 40(4): 285-91, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24720376

RESUMO

BACKGROUND AND OBJECTIVES: Tobacco and cannabis use are both highly prevalent worldwide. Their co-use is also common in adults and adolescents. Despite this frequent co-occurrence, cessation from both substances is rarely addressed in randomized clinical trials. Given evidence that tobacco use may increase during cannabis cessation attempts, and additionally that tobacco users have poorer cannabis cessation outcomes, we explored tobacco outcomes, specifically cigarette smoking, from an adolescent cannabis cessation trial that tested the efficacy of N-acetylesteine (NAC). METHODS: Cannabis-dependent adolescents (ages 15-21; n = 116) interested in cannabis treatment were randomized to NAC (1200 mg bid) or matched placebo for 8 weeks. Participants did not need to be cigarette smokers or be interested in smoking cessation to qualify for inclusion. RESULTS: Approximately 59% of enrolled participants were daily and non-daily cigarette smokers, and only differed from non-smoking participants on the compulsion sub-scale of the Marijuana Craving Questionnaire. Among cigarette smokers who were retained in the study, there was no change in cigarettes per day for either NAC or placebo groups during the eight-week treatment phase. Being a cigarette smoker did not appear to influence the effects of NAC on cannabis abstinence, though there was a trend in the placebo group of poorer cannabis outcomes for cigarette smokers vs. non-smokers. CONCLUSIONS: No evidence was found of compensatory cigarette smoking during this cannabis cessation trial in adolescents. Further work assessing interventions to reduce both cannabis and tobacco use in this population is greatly needed.


Assuntos
Acetilcisteína/uso terapêutico , Abuso de Maconha/tratamento farmacológico , Fumar Maconha/tratamento farmacológico , Fumar , Adolescente , Feminino , Humanos , Masculino , Inquéritos e Questionários , Resultado do Tratamento , Adulto Jovem
14.
Port of Spain; Caribbean Public Health Agency; Feb. 2014. 29 p.
Monografia em Inglês | MedCarib | ID: med-17903

RESUMO

This Evidence Summary is prepared in response to a request from the Assistant Secretary-General, Human and Social Development, CARICOM. Given the debate internationally with respect to Cannabis sativa’s safety and efficacy, and given that CARPHA’s role is to provide direction in analyzing, defining and responding to public health priorities, we have interpreted this broad request as requiring our response to the following question: What is the current state of scientific knowledge regarding the effectiveness of marijuana and its chemical constituents in medical treatment in humans? We present a synthesis of research findings, of international relevance, that answers the specified question.


Assuntos
Cannabis/uso terapêutico , Cannabis/efeitos dos fármacos , Fumar Maconha/tratamento farmacológico , Efetividade
15.
Ther Drug Monit ; 36(2): 218-24, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24067260

RESUMO

BACKGROUND: Recently, high-dose oral synthetic delta-9-tetrahydrocannabinol (THC) was shown to alleviate cannabis withdrawal symptoms. The present data describe cannabinoid pharmacokinetics in chronic, daily cannabis smokers who received high-dose oral THC pharmacotherapy and later a smoked cannabis challenge. METHODS: Eleven daily cannabis smokers received 0, 30, 60, or 120 mg/d THC for four 5-day medication sessions, each separated by 9 days of ad libitum cannabis smoking. On the fifth day, participants were challenged with smoking one 5.9% THC cigarette. Plasma collected on the first and fifth days was quantified by two-dimensional gas chromatography mass spectrometer for THC, 11-hydroxy-THC (11-OH-THC), and 11-nor-9-carboxy-THC (THCCOOH). Linear ranges (ng/mL) were 0.5-100 for THC, 1-50 for 11-OH-THC, and 0.5-200 for THCCOOH. RESULTS: During placebo dosing, THC, 11-OH-THC, and THCCOOH concentrations consistently decreased, whereas all cannabinoids increased dose dependently during active dronabinol administration. THC increase over time was not significant after any dose, 11-OH-THC increased significantly during the 60- and 120-mg/d doses, and THCCOOH increased significantly only during the 120-mg/d dose. THC, 11-OH-THC, and THCCOOH concentrations peaked within 0.25 hours after cannabis smoking, except after 120 mg/d THC when THCCOOH peaked 0.5 hours before smoking. CONCLUSIONS: The significant withdrawal effects noted during placebo dronabinol administration were supported by significant plasma THC and 11-OH-THC concentration decreases. During active dronabinol dosing, significant dose-dependent increases in THC and 11-OH-THC concentrations support withdrawal symptom suppression. THC concentrations after cannabis smoking were only distinguishable from oral THC doses for 1 hour, too short a period to feasibly identify cannabis relapse. THCCOOH/THC ratios were higher 14 hours after overnight oral dronabinol abstinence but cannot distinguish oral THC dosing from the smoked cannabis intake.


Assuntos
Canabinoides/sangue , Canabinoides/uso terapêutico , Dronabinol/sangue , Dronabinol/uso terapêutico , Abuso de Maconha/sangue , Abuso de Maconha/tratamento farmacológico , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Adolescente , Adulto , Canabinoides/farmacocinética , Relação Dose-Resposta a Droga , Método Duplo-Cego , Dronabinol/farmacocinética , Feminino , Humanos , Masculino , Fumar Maconha/sangue , Fumar Maconha/tratamento farmacológico , Pessoa de Meia-Idade , Síndrome de Abstinência a Substâncias/sangue , Adulto Jovem
16.
Clin Neuropharmacol ; 36(3): 98-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23673914

RESUMO

Cannabis-induced psychotic symptoms (CIPSs) have both similarities and differences with positive symptoms of schizophrenia, and it remains unclear whether CIPSs result from dopaminergic mechanisms and can be treated with antipsychotics. We report the case of a 22-year-old male patient with ultrahigh risk criteria for psychosis, who reported cannabis addiction and recurrent CIPSs. Aripiprazole 10 mg/d could totally and durably suppress CIPSs in the patient, but had no effect on the smoking level. Treating CIPSs in ultrahigh risk individuals who cannot stop or refuse stopping cannabis might fit a harm-reduction strategy by preventing transition into psychosis.


Assuntos
Antipsicóticos/uso terapêutico , Abuso de Maconha/complicações , Abuso de Maconha/tratamento farmacológico , Fumar Maconha/efeitos adversos , Piperazinas/uso terapêutico , Psicoses Induzidas por Substâncias/tratamento farmacológico , Psicoses Induzidas por Substâncias/etiologia , Quinolonas/uso terapêutico , Aripiprazol , Humanos , Masculino , Abuso de Maconha/psicologia , Fumar Maconha/tratamento farmacológico , Adulto Jovem
17.
Neuropsychopharmacology ; 35(9): 1879-85, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20428110

RESUMO

Worldwide cannabis dependence is increasing, as is the concentration of Delta(9)-tetrahydrocannabinol (THC) in street cannabis. At the same time, the concentration of the second most abundant cannabinoid in street cannabis, cannabidiol (CBD), is decreasing. These two cannabinoids have opposing effects both pharmacologically and behaviorally when administered in the laboratory. No research has yet examined how the ratio of these constituents impacts on the appetitive/reinforcing effects of cannabis in humans. A total of 94 cannabis users were tested 7 days apart, once while non-intoxicated and once while acutely under the influence of their own chosen smoked cannabis on dependence-related measures. Using an unprecedented methodology, a sample of cannabis (as well as saliva) was collected from each user and analyzed for levels of cannabinoids. On the basis of CBD : THC ratios in the cannabis, individuals from the top and bottom tertiles were directly compared on indices of the reinforcing effects of drugs, explicit liking, and implicit attentional bias to drug stimuli. When intoxicated, smokers of high CBD : THC strains showed reduced attentional bias to drug and food stimuli compared with smokers of low CBD : THC. Those smoking higher CBD : THC strains also showed lower self-rated liking of cannabis stimuli on both test days. Our findings suggest that CBD has potential as a treatment for cannabis dependence. The acute modulation of the incentive salience of drug cues by CBD may possibly generalize to a treatment for other addictive disorders.


Assuntos
Comportamento Apetitivo/efeitos dos fármacos , Canabidiol/uso terapêutico , Dronabinol/administração & dosagem , Alucinógenos/administração & dosagem , Fumar Maconha/tratamento farmacológico , Fumar Maconha/psicologia , Adolescente , Análise de Variância , Canabidiol/administração & dosagem , Canabidiol/urina , Comportamento de Escolha/efeitos dos fármacos , Cromatografia Líquida/métodos , Dronabinol/urina , Feminino , Alucinógenos/urina , Humanos , Masculino , Abuso de Maconha , Fumar Maconha/urina , Espectrometria de Massas/métodos , Testes Neuropsicológicos , Tempo de Reação/efeitos dos fármacos , Adulto Jovem
18.
J Clin Pharmacol ; 42(S1): 34S-40S, 2002 11.
Artigo em Inglês | MEDLINE | ID: mdl-12412834

RESUMO

This article presents data from two avenues of marijuana research. First, the author shows that daily marijuana smoking in healthy individuals produces dependence, as demonstrated by withdrawal symptoms such as increased irritability and depression and decreased food intake. In addition, two antidepressant medications were evaluated to assess their potential effectiveness in the treatment of marijuana withdrawal symptoms: (1) sustained-release bupropion (0, 300 mg/day) and (2) nefazodone (0, 450 mg/day). Research participants were regular marijuana smokers who lived in a residential laboratory in groups of two to four. While inpatients, participants smoked active marijuana (2.8%-3.1% THC) repeatedly for 4 days, followed by 8 to 12 days of placebo marijuana (0.0% THC). Results show that during marijuana abstinence, (1) bupropion increased ratings of irritability, depression, and stomach pain and decreased food intake and sleep quality compared to placebo maintenance, and (2) nefazodone decreased anxiety during marijuana withdrawal but did not alter ratings of irritability and misery. Thus, neither medication showed promise as potential treatments for symptoms of marijuana withdrawal. The second avenue of research focused on the effect of cannabinoids in individuals with muscle mass loss, an indicator of wasting in HIV illness. Given that there are little scientific data contributing to the debates concerning medical marijuana, this study directly compared the effects of oral delta9-THC (0, 10, 20, 30 mg PO) to smoked marijuana (0.0%, 1.8%, 2.8%, 3.9% THC) in HIV + marijuana smokers with muscle mass loss (< 90% body cell mass/height). Multiple dimensions of human behavior were measured, including food intake, mood, and cognitive performance. Drugs were administered using a within-subject, double-blind, staggered, double-dummy design. Participants were free to self-select from a variety of foods throughout most of the session. Preliminary data (n = 9) suggest that oral THC was more effective at increasing food intake, but the volunteers "liked" the effects of smoked marijuana more than the effects of oral THC.


Assuntos
Dronabinol/uso terapêutico , Soropositividade para HIV/tratamento farmacológico , Fumar Maconha/efeitos adversos , Fumar Maconha/tratamento farmacológico , Bupropiona/uso terapêutico , Ingestão de Alimentos/efeitos dos fármacos , Humanos , Piperazinas , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Triazóis/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA