Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
Psychopharmacology (Berl) ; 240(7): 1393-1415, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37157001

RESUMO

Cannabis self-administration studies may be helpful for identifying factors that influence cannabis consumption and subjective response to cannabis. Additionally, these paradigms could be useful for testing novel pharmacotherapies for cannabis use disorder. This scoping review aims to summarize the findings from existing ad libitum cannabis self-administration studies to determine what has been learned from these studies as well as their limitations. We examined studies that specifically examined cannabis smoking, focusing on subjective response and self-administration behavior (e.g., smoking topography). A systematic search was conducted using PubMed and Embase from inception to October 22, 2022. Our search strategy identified 26 studies (total N = 662, 79% male) that met our eligibility criteria. We found that tetrahydrocannabinol (THC) concentration significantly affected subjective response to cannabis in some but not all studies. In general, cannabis self-administration tended to be most intense at the beginning of the laboratory session and decreased in later parts of the session. There was limited data on cannabis self-administration in adults older than 55. Data on external validity and test-retest reliability were also limited. Addressing these limitations in future ad libitum cannabis self-administration studies could lead to more valid and generalizable paradigms, which in turn could be used to improve our understanding of cannabis use patterns and to help guide medication development for cannabis use disorder.


Assuntos
Cannabis , Alucinógenos , Abuso de Maconha , Fumar Maconha , Feminino , Humanos , Masculino , Agonistas de Receptores de Canabinoides , Dronabinol/farmacologia , Dronabinol/uso terapêutico , Alucinógenos/uso terapêutico , Abuso de Maconha/tratamento farmacológico , Reprodutibilidade dos Testes
2.
Addict Biol ; 27(1): e13085, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34390300

RESUMO

Disrupted brain gamma-aminobutyric acid (GABA)/glutamate homeostasis is a promising target for pharmacological intervention in co-occurring bipolar disorder (BD) and cannabis use disorder (CUD). Gabapentin is a safe and well-tolerated medication, FDA-approved to treat other neurological diseases, that restores GABA/glutamate homeostasis, with treatment studies supporting efficacy in treating CUD, as well as anxiety and sleep disorders that are common to both BD and CUD. The present manuscript represents the primary report of a randomized, double-blind, placebo-controlled, crossover (1-week/condition), multimodal-MRI (proton-MR spectroscopy, functional MRI) pilot study of gabapentin (1200 mg/day) in BD + CUD (n = 22). Primary analyses revealed that (1) gabapentin was well tolerated and adherence and retention were high, (2) gabapentin increased dorsal anterior cingulate cortex (dACC) and right basal ganglia (rBG) glutamate levels and (3) gabapentin increased activation to visual cannabis cues in the posterior midcingulate cortex (pMCC, a region involved in response inhibition to rewarding stimuli). Exploratory evaluation of clinical outcomes further found that in participants taking gabapentin versus placebo, (1) elevations of dACC GABA levels were associated with lower manic/mixed and depressive symptoms and (2) elevations of rBG glutamate levels and pMCC activation to cannabis cues were associated with lower cannabis use. Though promising, the findings from this study should be interpreted with caution due to observed randomization order effects on dACC glutamate levels and identification of statistical moderators that differed by randomization order (i.e. cigarette-smoking status on rBG glutamate levels and pMCC cue activation). Nonetheless, they provide the necessary foundation for a more robustly designed (urn-randomized, parallel-group) future study of adjuvant gabapentin for BD + CUD.


Assuntos
Transtorno Bipolar/tratamento farmacológico , Gabapentina/uso terapêutico , Ácido Glutâmico/efeitos dos fármacos , Abuso de Maconha/tratamento farmacológico , Ácido gama-Aminobutírico/efeitos dos fármacos , Adolescente , Adulto , Transtorno Bipolar/epidemiologia , Fumar Cigarros/epidemiologia , Método Duplo-Cego , Feminino , Gabapentina/administração & dosagem , Gabapentina/efeitos adversos , Giro do Cíngulo/efeitos dos fármacos , Humanos , Imageamento por Ressonância Magnética , Masculino , Abuso de Maconha/epidemiologia , Pessoa de Meia-Idade , Projetos Piloto , Espectroscopia de Prótons por Ressonância Magnética , Adulto Jovem
3.
Med Clin (Barc) ; 159(4): 183-186, 2022 08 26.
Artigo em Inglês, Espanhol | MEDLINE | ID: mdl-34756408

RESUMO

There are few studies in Spain on cannabinoid hyperemesis syndrome (CHS), as well as on the use of topical capsaicin as a treatment. METHODS: Retrospective study of patients over 14 years of age seen in a hospital emergency department during 2018 and 2019 with a diagnosis of CHS based on the following criteria: compatible clinical picture, cannabis use less than 48h and positive urine cannabis test. Epidemiological and clinical variables, attendance times and treatment (including use of topical capsaicin 0.075%) were collected. RESULTS: Fifty-nine attendances were studied, from 29 patients (4.4 cases/10,000 visits, 95% CI 2.8-4.7). Fifty per cent returned for CHS, differing only in more tobacco (P=.01) and cocaine (P=.031) use. Capsaicin was used in 74.6% of visits. The mean time to resolution of vomiting after application was 17.87min. CONCLUSIONS: Although probably underdiagnosed, CHS has a low incidence in the emergency department in Spain, with high patient recurrence. The use of capsaicin ointment is efficient and safe.


Assuntos
Canabinoides , Cannabis , Abuso de Maconha , Canabinoides/efeitos adversos , Cannabis/efeitos adversos , Capsaicina , Humanos , Incidência , Abuso de Maconha/complicações , Abuso de Maconha/tratamento farmacológico , Abuso de Maconha/epidemiologia , Estudos Retrospectivos , Síndrome , Vômito/induzido quimicamente , Vômito/tratamento farmacológico , Vômito/epidemiologia
4.
Drug Alcohol Depend ; 229(Pt B): 109111, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34655945

RESUMO

BACKGROUND: An efficacious pharmacotherapy for cannabis use disorder (CUD) has yet to be established. This study preliminarily evaluated the safety and efficacy of varenicline for CUD in a proof-of-concept clinical trial. METHODS: Participants in this 6-week randomized, placebo-controlled pilot trial received either varenicline (n = 35) or placebo (n = 37), added to a brief motivational enhancement therapy intervention. Outcomes included cannabis withdrawal, cannabis abstinence, urine cannabinoid levels, percent cannabis use days, and cannabis sessions per day. RESULTS: Both treatment groups noted significant decreases in self-reported cannabis withdrawal, percentage of days used, and use sessions per day during treatment compared to baseline. While this pilot trial was not powered to detect statistically significant between-group differences, participants randomized to varenicline evidenced numerically greater rates of self-reported abstinence at the final study visit [Week 6 intent-to-treat (ITT): Varenicline: 17.1% vs. Placebo: 5.4%; RR = 3.2 (95% CI: 0.7,14.7)]. End-of-treatment urine creatinine corrected cannabinoid levels were numerically lower in the varenicline group and higher in the placebo group compared to baseline [Change from baseline: Varenicline -1.7 ng/mg (95% CI: -4.1,0.8) vs. Placebo: 1.9 ng/mg (95% CI: -0.4,4.3); Δ = 3.5 (95% CI: 0.1,6.9)]. Adverse events related to study treatment did not reveal new safety signals. CONCLUSIONS: Findings support the feasibility of conducting clinical trials of varenicline as a candidate pharmacotherapy for CUD, and indicate that a full-scale efficacy trial, powered based on effect sizes and variability yielded in this study, is warranted.


Assuntos
Abuso de Maconha , Abandono do Hábito de Fumar , Método Duplo-Cego , Humanos , Abuso de Maconha/tratamento farmacológico , Projetos Piloto , Vareniclina/efeitos adversos
5.
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
6.
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
7.
Ann Intern Med ; 173(9): ITC65-ITC80, 2020 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-33137270

RESUMO

The past 2 decades have seen a revolution in legal access to cannabis, driven largely by activists and business interests. As a result, the population of cannabis users nationwide-especially daily users-has grown significantly. An estimated 4.5-7 million persons in the United States now meet criteria for cannabis use disorder annually. This article focuses on the effects of cannabis use, intoxication, and withdrawal while also reviewing the developmental pathways of cannabis use disorder as well as evidence-based pharmacologic and psychosocial treatments.


Assuntos
Abuso de Maconha/terapia , Fumar Maconha/efeitos adversos , Dor Crônica/tratamento farmacológico , Feminino , Humanos , Abuso de Maconha/complicações , Abuso de Maconha/tratamento farmacológico , Maconha Medicinal/uso terapêutico , Transtornos Mentais/complicações , Educação de Pacientes como Assunto , Gravidez , Psicoterapia , Infecções Respiratórias/complicações , Fatores de Risco , Convulsões/prevenção & controle , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Síndrome de Abstinência a Substâncias/etiologia , Síndrome de Abstinência a Substâncias/terapia , Síndrome , Vômito/etiologia
8.
Lancet Psychiatry ; 7(10): 865-874, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32735782

RESUMO

Background A substantial and unmet clinical need exists for pharmacological treatment of cannabis use disorders. Cannabidiol could offer a novel treatment, but it is unclear which doses might be efficacious or safe. Therefore, we aimed to identify efficacious doses and eliminate inefficacious doses in a phase 2a trial using an adaptive Bayesian design. METHODS: We did a phase 2a, double-blind, placebo-controlled, randomised, adaptive Bayesian trial at the Clinical Psychopharmacology Unit (University College London, London, UK). We used an adaptive Bayesian dose-finding design to identify efficacious or inefficacious doses at a-priori interim and final analysis stages. Participants meeting cannabis use disorder criteria from DSM-5 were randomly assigned (1:1:1:1) in the first stage of the trial to 4-week treatment with three different doses of oral cannabidiol (200 mg, 400 mg, or 800 mg) or with matched placebo during a cessation attempt by use of a double-blinded block randomisation sequence. All participants received a brief psychological intervention of motivational interviewing. For the second stage of the trial, new participants were randomly assigned to placebo or doses deemed efficacious in the interim analysis. The primary objective was to identify the most efficacious dose of cannabidiol for reducing cannabis use. The primary endpoints were lower urinary 11-nor-9-carboxy-δ-9-tetrahydrocannabinol (THC-COOH):creatinine ratio, increased days per week with abstinence from cannabis during treatment, or both, evidenced by posterior probabilities that cannabidiol is better than placebo exceeding 0·9. All analyses were done on an intention-to-treat basis. This trial is registered with ClinicalTrials.gov (NCT02044809) and the EU Clinical Trials Register (2013-000361-36). FINDINGS: Between May 28, 2014, and Aug 12, 2015 (first stage), 48 participants were randomly assigned to placebo (n=12) and to cannabidiol 200 mg (n=12), 400 mg (n=12), and 800 mg (n=12). At interim analysis, cannabidiol 200 mg was eliminated from the trial as an inefficacious dose. Between May 24, 2016, and Jan 12, 2017 (second stage), randomisation continued and an additional 34 participants were allocated (1:1:1) to cannabidiol 400 mg (n=12), cannabidiol 800 mg (n=11), and placebo (n=11). At final analysis, cannabidiol 400 mg and 800 mg exceeded primary endpoint criteria (0·9) for both primary outcomes. For urinary THC-COOH:creatinine ratio, the probability of being the most efficacious dose compared with placebo given the observed data was 0·9995 for cannabidiol 400 mg and 0·9965 for cannabidiol 800 mg. For days with abstinence from cannabis, the probability of being the most efficacious dose compared with placebo given the observed data was 0·9966 for cannabidiol 400 mg and 0·9247 for cannabidiol 800 mg. Compared with placebo, cannabidiol 400 mg decreased THC-COOH:creatinine ratio by -94·21 ng/mL (95% interval estimate -161·83 to -35·56) and increased abstinence from cannabis by 0·48 days per week (0·15 to 0·82). Compared with placebo, cannabidiol 800 mg decreased THC-COOH:creatinine ratio by -72·02 ng/mL (-135·47 to -19·52) and increased abstinence from cannabis by 0·27 days per week (-0·09 to 0·64). Cannabidiol was well tolerated, with no severe adverse events recorded, and 77 (94%) of 82 participants completed treatment. INTERPRETATION: In the first randomised clinical trial of cannabidiol for cannabis use disorder, cannabidiol 400 mg and 800 mg were safe and more efficacious than placebo at reducing cannabis use. FUNDING: Medical Research Council.


Assuntos
Canabidiol/administração & dosagem , Abuso de Maconha/tratamento farmacológico , Síndrome de Abstinência a Substâncias , Adolescente , Adulto , Teorema de Bayes , Canabidiol/efeitos adversos , Método Duplo-Cego , Dronabinol/urina , Feminino , Alucinógenos/urina , Humanos , Londres , Masculino , Fumar Maconha , Resultado do Tratamento , Adulto Jovem
9.
Drug Alcohol Depend ; 214: 108188, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32717502

RESUMO

INTRODUCTION: Opioid misuse has reached epidemic proportions among emerging adults in the U.S. To inform prevention efforts, this study examined adolescent factors related to alcohol and marijuana (AM) use that are associated with a higher or lower risk for opioid misuse during emerging adulthood. METHODS: We used 11 waves of survey data from a diverse California cohort (N = 6,509). Predictor variables from waves 1-7 (ages 11-17) included individual (resistance self-efficacy, positive expectancies) family (older sibling and important adult use), and peer (perceived norms, time spent with peers who use, peer approval) factors. Opioid misuse at wave 8 (mean age = 18.3) and wave 11 (mean age = 21.6) included heroin and nonmedical prescription drug use. RESULTS: Initial latent growth models (LGMs) indicated that nearly all intercepts and slopes for individual, family, and peer AM factors predicted opioid misuse at waves 8 and 11. These associations were reduced to non-significance after adjusting for prior other substance use with the exception of three intercepts: positive expectancies, peer approval, and older sibling use predicted a higher probability of opioid misuse at wave 8. CONCLUSIONS: Stronger AM positive expectancies, perceived peer approval of AM use, and older sibling AM use during adolescence are associated with a higher likelihood of opioid misuse during the transition to emerging adulthood. However, most adolescent factors were no longer associated with subsequent opioid misuse after adjusting for history of other substance use, highlighting the importance of considering the larger context of substance use in studies of opioid misuse among young people.


Assuntos
Transtornos Relacionados ao Uso de Opioides/epidemiologia , Adolescente , Comportamento do Adolescente , Adulto , Consumo de Bebidas Alcoólicas/epidemiologia , Analgésicos Opioides/uso terapêutico , Criança , Feminino , Heroína , Humanos , Masculino , Abuso de Maconha/tratamento farmacológico , Fumar Maconha/epidemiologia , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Grupo Associado , Uso Indevido de Medicamentos sob Prescrição/estatística & dados numéricos , Medicamentos sob Prescrição , Inquéritos e Questionários , Adulto Jovem
10.
Psychopharmacology (Berl) ; 237(5): 1507-1519, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32034447

RESUMO

RATIONALE: Given that tetrahydrocannabinol (THC) and nicotine have similar effects on negative affect (NA), we hypothesized that a 7-mg nicotine patch (NP) would reduce NA-related cannabis (CAN) withdrawal symptoms in cannabis-dependent (CD) individuals who were not nicotine dependent. OBJECTIVE: We sought to determine whether NP reduces NA across 15 days of CAN abstinence in two groups: non-tobacco smokers (NTS) and light tobacco smokers (LTS). METHODS: CD participants (N = 127; aged 18-35) who used CAN at least 5 times/week for the past 12 + months were randomized to (1) NP or (2) a placebo patch (PP) and received $300 for sustained biochemically verified CAN abstinence. Of those randomly assigned, 52 of 63 NP, and 56 of 64 PP maintained biochemically verified CAN abstinence and 51 NP and 50 PP participants complied with all aspects of the study. Affect and other withdrawal symptoms were measured every 48 h across 15 days of CAN abstinence. RESULTS: After controlling for age, tobacco use, baseline THC concentration, and baseline measurements of the dependent variable, NP reduced NA symptoms across the 15-day treatment relative to PP. Differences in NA and CAN withdrawal symptoms were not moderated by tobacco user status. CONCLUSIONS: The findings provide the first evidence that NP may be able to attenuate NA-related withdrawal symptoms in individuals with cannabis use disorder who are not heavy users of tobacco or nicotine. CLINICAL TRIALS REGISTRY: NCT01400243 http://www.clinicaltrials.gov.


Assuntos
Abuso de Maconha/tratamento farmacológico , Abuso de Maconha/psicologia , Nicotina/administração & dosagem , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Síndrome de Abstinência a Substâncias/psicologia , Dispositivos para o Abandono do Uso de Tabaco , Adolescente , Adulto , Feminino , Alucinógenos/uso terapêutico , Humanos , Masculino , Fumar Maconha/tratamento farmacológico , Fumar Maconha/psicologia , Abandono do Hábito de Fumar/métodos , Abandono do Hábito de Fumar/psicologia , Dispositivos para o Abandono do Uso de Tabaco/tendências , Adulto Jovem
11.
Clin Toxicol (Phila) ; 58(6): 471-475, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31482758

RESUMO

Introduction: Cannabinoid hyperemesis syndrome (CHS) is a disorder of cyclic and recurrent nausea, vomiting, and abdominal pain associated with high-frequency and extended-duration marijuana use. Standard antiemetic therapy is often ineffective; however, capsaicin, an agonist of transient receptor potential vanilloid 1 (TRPV1), has shown promise in treating CHS.Methods: This retrospective cohort analysis evaluated the safety and efficacy of topical capsaicin for patients presenting with CHS. The primary outcome was to assess if utilization of capsaicin for ED management of CHS decreased ED length of stay (LOS) as compared to a visit without capsaicin. Secondary outcomes included a cost analysis, use of rescue therapies, and adverse events.Results: Forty-three patients met the inclusion criteria within the study period. ED LOS was reduced with capsaicin by a median of 22 minutes (201 vs. 179 min, p = 0.33). Patients received fewer additional medications if capsaicin was utilized (4 vs. 3 doses, p = 0.015), and 67% of visits where capsaicin was utilized required no further treatment prior to discharge. Additionally, opioid usage was less when utilizing capsaicin (166.5 vs. 69 mg OME). Forty-two percent of patients did not have a repeat CHS presentation to the ED after receiving capsaicin for an additional three months after the study period ended. Total medication cost was minimally more expensive (median difference of $3.26) in the capsaicin group. There were no significant adverse events reported with capsaicin.Conclusion: There was no significant difference in ED LOS when capsaicin was utilized for CHS. However, there was a decrease in total medications administered and a reduction in opioid requirements. While medication costs for capsaicin visits were minimally more expensive, the utility of capsaicin as an over-the-counter (OTC) product may empower at home therapy with OTC products, decreasing potentially unnecessary healthcare encounters and costs.


Assuntos
Antieméticos/uso terapêutico , Canabinoides/efeitos adversos , Capsaicina/uso terapêutico , Serviço Hospitalar de Emergência , Abuso de Maconha/tratamento farmacológico , Fumar Maconha/efeitos adversos , Vômito/tratamento farmacológico , Dor Abdominal/tratamento farmacológico , Dor Abdominal/etiologia , Administração Tópica , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Antieméticos/administração & dosagem , Antieméticos/efeitos adversos , Capsaicina/administração & dosagem , Capsaicina/efeitos adversos , Colorado , Serviço Hospitalar de Emergência/estatística & dados numéricos , Humanos , Tempo de Internação , Abuso de Maconha/complicações , Pessoa de Meia-Idade , Náusea/tratamento farmacológico , Náusea/etiologia , Estudos Retrospectivos , Síndrome , Vômito/etiologia , Adulto Jovem
12.
Psychopharmacology (Berl) ; 237(2): 479-490, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31712969

RESUMO

RATIONALE: Depression is common among individuals with cannabis use disorder (CUD), particularly individuals who present to CUD treatment. Treatments that consider this comorbidity are essential. OBJECTIVES: The goal of this secondary analysis was to examine whether N-acetylcysteine (NAC) reduced depressive symptoms among adults (age 18-50) with CUD (N = 302) and whether the effect of NAC on cannabis cessation varied as a result of baseline levels of depression. Bidirectional associations between cannabis use amount and depression were also examined. METHODS: Data for this secondary analysis were from a National Drug Abuse Treatment Clinical Trials Network (NIDA CTN) multi-site clinical trial for CUD. Adults with CUD (N = 302) were randomized to receive 2400 mg of NAC daily or matched placebo for 12 weeks. All participants received abstinence-based contingency management. Cannabis quantity was measured by self-report, and weekly urinary cannabinoid levels (11-nor-9-carboxy-Δ9-tetrahydrocannabinol) confirmed abstinence. Depressive symptoms were measured by the Hospital Anxiety and Depression Scale. RESULTS: Depressive symptoms did not differ between the NAC and placebo groups during treatment. There was no significant interaction between treatment and baseline depression predicting cannabis abstinence during treatment. Higher baseline depression was associated with decreased abstinence throughout treatment and a significant gender interaction suggested that this may be particularly true for females. Cross-lagged panel models suggested that depressive symptoms preceded increased cannabis use amounts (in grams) during the subsequent month. The reverse pathway was not significant (i.e., greater cannabis use preceding depressive symptoms). CONCLUSIONS: Results from this study suggest that depression may be a risk factor for poor CUD treatment outcome and therefore should be addressed in the context of treatment. However, results do not support the use of NAC to concurrently treat co-occurring depressive symptoms and CUD in adults. TRIAL REGISTRATION: Clinicaltrials.gov: NCT01675661.


Assuntos
Acetilcisteína/uso terapêutico , Depressão/tratamento farmacológico , Depressão/psicologia , Abuso de Maconha/tratamento farmacológico , Abuso de Maconha/psicologia , Acetilcisteína/farmacologia , Adulto , Cannabis , Comorbidade , Depressão/epidemiologia , Método Duplo-Cego , Dronabinol/uso terapêutico , Feminino , Sequestradores de Radicais Livres/farmacologia , Sequestradores de Radicais Livres/uso terapêutico , Humanos , Masculino , Abuso de Maconha/epidemiologia , Motivação/efeitos dos fármacos , Motivação/fisiologia , Resultado do Tratamento , Adulto Jovem
13.
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
14.
Int J Drug Policy ; 69: 55-59, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31030906

RESUMO

BACKGROUND: Alternative cannabis regulation models are discussed and implemented worldwide. A baseline scenario under the assumption of no policy or market changes may prove useful to forecast cannabis use and treatment demand and evaluate changes in legislation. METHODS: Based on data of the Continuous Rolling Survey of Addictive Behaviours and Related Risks on cannabis use, age, gender and nationality from 2011 to 2015, we used general estimating equation analysis to model lifetime and 30-days prevalence from 2015 to 2045 in Switzerland accounting for demographic trends. RESULTS: Lifetime prevalence of cannabis use is projected to grow from 28.3% (CI 95% 27.8-28.8) in 2015 to 42.0% (CI 95% 41.0-43.0) in 2045. 30-days prevalence would increase slightly from 2.70% (CI 95% 2.53-2.88) to 3.39% (CI 95% 3.11-3.66). Due to population growth, absolute numbers with past 30-day cannabis use are estimated to increase from 202,784 (CI 95% 189,534-216,035) to 314,302 (CI 95% 288,504-340,100). Among those aged under 30 years no substantial change in lifetime and 30-days prevalence of cannabis use is projected. Larger changes are estimated to occur in the age group 30+. The mean age of past 30-day cannabis users would increase for men with Swiss nationality from 30.3 to 38.7 years. DISCUSSION: Population-based survey data and demographic projections can be used to develop baseline scenarios of future cannabis use. Assuming no changes in cannabis legislation, growing absolute numbers of users will likely increase treatment demand. Cannabis use is estimated to increase among the group aged >30 years, which is currently underrepresented in clinical treatment and research. Our findings highlight the need for prospective baseline scenarios to evaluate the impact of legislative changes on cannabis use. Moreover, in Switzerland effective prevention and treatment interventions for cannabis use disorders are required even if cannabis legislation remains unchanged.


Assuntos
Abuso de Maconha/epidemiologia , Fumar Maconha/tendências , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Usuários de Drogas , Feminino , Previsões , Humanos , Masculino , Abuso de Maconha/tratamento farmacológico , Abuso de Maconha/prevenção & controle , Pessoa de Meia-Idade , Prevalência , Inquéritos e Questionários , Suíça/epidemiologia , Adulto Jovem
15.
Cochrane Database Syst Rev ; 1: CD008940, 2019 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-30687936

RESUMO

BACKGROUND: Globally, cannabis use is prevalent and widespread. There are currently no pharmacotherapies approved for treatment of cannabis use disorders.This is an update of a Cochrane Review first published in the Cochrane Library in Issue 12, 2014. OBJECTIVES: To assess the effectiveness and safety of pharmacotherapies as compared with each other, placebo or no pharmacotherapy (supportive care) for reducing symptoms of cannabis withdrawal and promoting cessation or reduction of cannabis use. SEARCH METHODS: We updated our searches of the following databases to March 2018: the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, PsycINFO and Web of Science. SELECTION CRITERIA: Randomised controlled trials (RCTs) and quasi-RCTs involving the use of medications to treat cannabis withdrawal or to promote cessation or reduction of cannabis use, or both, in comparison with other medications, placebo or no medication (supportive care) in people diagnosed as cannabis dependent or who were likely to be dependent. DATA COLLECTION AND ANALYSIS: We used standard methodological procedures expected by Cochrane. MAIN RESULTS: We included 21 RCTs involving 1755 participants: 18 studies recruited adults (mean age 22 to 41 years); three studies targeted young people (mean age 20 years). Most (75%) participants were male. The studies were at low risk of performance, detection and selective outcome reporting bias. One study was at risk of selection bias, and three studies were at risk of attrition bias.All studies involved comparison of active medication and placebo. The medications were diverse, as were the outcomes reported, which limited the extent of analysis.Abstinence at end of treatment was no more likely with Δ9-tetrahydrocannabinol (THC) preparations than with placebo (risk ratio (RR) 0.98, 95% confidence interval (CI) 0.64 to 1.52; 305 participants; 3 studies; moderate-quality evidence). For selective serotonin reuptake inhibitor (SSRI) antidepressants, mixed action antidepressants, anticonvulsants and mood stabilisers, buspirone and N-acetylcysteine, there was no difference in the likelihood of abstinence at end of treatment compared to placebo (low- to very low-quality evidence).There was qualitative evidence of reduced intensity of withdrawal symptoms with THC preparations compared to placebo. For other pharmacotherapies, this outcome was either not examined, or no significant differences was reported.Adverse effects were no more likely with THC preparations (RR 1.02, 95% CI 0.89 to 1.17; 318 participants; 3 studies) or N-acetylcysteine (RR 0.94, 95% CI 0.71 to 1.23; 418 participants; 2 studies) compared to placebo (moderate-quality evidence). For SSRI antidepressants, mixed action antidepressants, buspirone and N-acetylcysteine, there was no difference in adverse effects compared to placebo (low- to very low-quality evidence).There was no difference in the likelihood of withdrawal from treatment due to adverse effects with THC preparations, SSRIs antidepressants, mixed action antidepressants, anticonvulsants and mood stabilisers, buspirone and N-acetylcysteine compared to placebo (low- to very low-quality evidence).There was no difference in the likelihood of treatment completion with THC preparations, SSRI antidepressants, mixed action antidepressants and buspirone compared to placebo (low- to very low-quality evidence) or with N-acetylcysteine compared to placebo (RR 1.06, 95% CI 0.93 to 1.21; 418 participants; 2 studies; moderate-quality evidence). Anticonvulsants and mood stabilisers appeared to reduce the likelihood of treatment completion (RR 0.66, 95% CI 0.47 to 0.92; 141 participants; 3 studies; low-quality evidence).Available evidence on gabapentin (anticonvulsant), oxytocin (neuropeptide) and atomoxetine was insufficient for estimates of effectiveness. AUTHORS' CONCLUSIONS: There is incomplete evidence for all of the pharmacotherapies investigated, and for many outcomes the quality of the evidence was low or very low. Findings indicate that SSRI antidepressants, mixed action antidepressants, bupropion, buspirone and atomoxetine are probably of little value in the treatment of cannabis dependence. Given the limited evidence of efficacy, THC preparations should be considered still experimental, with some positive effects on withdrawal symptoms and craving. The evidence base for the anticonvulsant gabapentin, oxytocin, and N-acetylcysteine is weak, but these medications are also worth further investigation.


Assuntos
Abuso de Maconha/tratamento farmacológico , Acetilcisteína/efeitos adversos , Acetilcisteína/uso terapêutico , Adulto , Anticonvulsivantes/efeitos adversos , Anticonvulsivantes/uso terapêutico , Antidepressivos/efeitos adversos , Antidepressivos/uso terapêutico , Buspirona/efeitos adversos , Buspirona/uso terapêutico , Dronabinol/efeitos adversos , Dronabinol/uso terapêutico , Feminino , Humanos , Masculino , Ensaios Clínicos Controlados Aleatórios como Assunto , Agonistas do Receptor de Serotonina/efeitos adversos , Agonistas do Receptor de Serotonina/uso terapêutico , Inibidores Seletivos de Recaptação de Serotonina/uso terapêutico , Adulto Jovem
16.
Addict Biol ; 24(4): 765-776, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30378231

RESUMO

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.


Assuntos
Fumar Cigarros/tratamento farmacológico , Dronabinol/análogos & derivados , Abuso de Maconha/tratamento farmacológico , Agentes de Cessação do Hábito de Fumar/uso terapêutico , Abandono do Hábito de Fumar , Síndrome de Abstinência a Substâncias/fisiopatologia , Vareniclina/uso terapêutico , Adulto , Fumar Cigarros/epidemiologia , Comorbidade , Dronabinol/uso terapêutico , Feminino , Humanos , Masculino , Abuso de Maconha/epidemiologia , Nicotina/efeitos adversos , Agonistas Nicotínicos/efeitos adversos , Síndrome de Abstinência a Substâncias/etiologia , Adulto Jovem
17.
Lancet Psychiatry ; 6(1): 35-45, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30528676

RESUMO

BACKGROUND: Cannabis is one of the most widely used drugs worldwide. Cannabis use disorder is characterised by recurrent use of cannabis that causes significant clinical and functional impairment. There are no approved pharmacological treatments for cannabis use disorder. One approach is to potentiate endocannabinoid signalling by inhibiting fatty acid amide hydrolase (FAAH), the enzyme that degrades the endocannabinoid anandamide. We aimed to test the efficacy and safety of the FAAH-inhibitor PF-04457845 in reduction of cannabis withdrawal and cannabis use in men who were daily cannabis users. METHODS: We did a double-blind, placebo-controlled, parallel group phase 2a trial at one site in men aged 18-55 years with cannabis dependence according to DSM-IV criteria (equivalent to cannabis use disorder in DSM-5). After baseline assessments, participants were randomly assigned (2:1) to receive PF-04457845 (4 mg per day) or placebo using a fixed block size of six participants, stratified by severity of cannabis use and desire to quit. Participants were admitted to hospital for 5 days (maximum 8 days) to achieve abstinence and precipitate cannabis withdrawal, after which they were discharged to continue the remaining 3 weeks of treatment as outpatients. The primary endpoints were treatment-related differences in cannabis withdrawal symptoms during hospital admission, and week 4 (end of treatment) self-reported cannabis use and urine THC-COOH concentrations in the intention-to-treat population. The study is registered at ClinicalTrials.gov, number NCT01618656. FINDINGS: Between Sept 12, 2012, and Jan 18, 2016, 46 men were randomly assigned to PF-04457845 and 24 to placebo. Adherence to study medication was 88%, as confirmed by video-calling and pill count, and corroborated by corresponding drug and anandamide concentrations in blood. Relative to placebo, treatment with PF-04457845 was associated with reduced symptoms of cannabis withdrawal (first day of treatment mean symptom score 11·00 [95% CI 7·78-15·57] vs 6·04 [4·43-8·24]; difference 4·96 [0·71-9·21]; padj=0·048; second day of treatment 11·74 [8·28-16·66] vs 6·02 [4·28-8·47]; difference 5·73 [1·13-10·32]; padj=0·035) and related mood symptoms during the inpatient phase. Additionally, treatment with PF-04457845 was associated with lower self-reported cannabis use at 4 weeks (mean 1·27 joints per day [95% CI 0·82-1·97] vs 0·40 [0·25-0·62]; difference 0·88 [0·29-1·46]; p=0·0003) and lower urinary THC-COOH concentrations (mean 657·92 ng/mL [95% CI 381·60-1134·30] vs 265·55 [175·60-401·57]; difference 392·37 [17·55-767·18)]; p=0·009). Eight (17%) patients in the PF-04457845 group and four (17%) in the placebo group discontinued during the treatment period. During the 4-week treatment phase, 20 (43%) of 46 participants in the PF-04457845 group and 11 (46%) of 24 participants in the placebo group had an adverse event. There were no serious adverse events. INTERPRETATION: PF-04457845, a novel FAAH inhibitor, reduced cannabis withdrawal symptoms and cannabis use in men, and might represent an effective and safe approach for the treatment of cannabis use disorder. FUNDING: United States National Institute of Drug Abuse (NIDA).


Assuntos
Cannabis , Abuso de Maconha/tratamento farmacológico , Piridazinas/administração & dosagem , Síndrome de Abstinência a Substâncias , Ureia/análogos & derivados , Adolescente , Adulto , Amidoidrolases , Método Duplo-Cego , Humanos , Masculino , Fumar Maconha , Pessoa de Meia-Idade , Resultado do Tratamento , Ureia/administração & dosagem , Adulto Jovem
18.
Drug Alcohol Depend ; 192: 59-66, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30218864

RESUMO

BACKGROUND: It is common for cannabis users to also use tobacco. While data suggest that tobacco users have more difficulty achieving cannabis cessation, secondary analyses of clinical trial data sets may provide insight into the moderating variables contributing to this relationship, as well as changes in tobacco use during cannabis treatment. Those were the aims of this secondary analysis. METHODS: The parent study was a multi-site trial of N-acetylcysteine for cannabis dependence conducted within the National Drug Abuse Treatment Clinical Trials Network. Participants were treatment-seeking adults (ages 18-50) who met criteria for cannabis dependence (N = 302). For cigarette smokers (n = 117), tobacco use was assessed via timeline follow-back and nicotine dependence was assessed via the Fagerström Test for Nicotine Dependence (FTND). Outcome measures included: 1) changes in tobacco use based on treatment assignment, nicotine dependence, and concurrent cannabis reduction/abstinence, and 2) independent associations between nicotine dependence and cannabis abstinence. RESULTS: Cigarette smokers accounted for 39% of the sample (117/302), with a median FTND score of 3.0 (10-point scale). Among those with lower baseline nicotine dependence scores, cigarette smoking was reduced in the active treatment group compared to placebo. Those with moderate/high levels of nicotine dependence showed slight increases in smoking following active treatment. Nicotine dependence did not affect cannabis cessation. CONCLUSIONS: Cigarette smoking during cannabis treatment was affected, but depended on baseline nicotine dependence severity, though dependence levels did not impact cannabis abstinence. Interventions that address both tobacco and cannabis are needed, especially due to an increasing prevalence of cannabis use.


Assuntos
Fumar Cigarros/epidemiologia , Abuso de Maconha/tratamento farmacológico , Abuso de Maconha/epidemiologia , Abandono do Hábito de Fumar/métodos , Tabagismo/epidemiologia , Acetilcisteína/uso terapêutico , Adolescente , Adulto , Fumar Cigarros/psicologia , Feminino , Humanos , Masculino , Abuso de Maconha/psicologia , Maconha Medicinal/uso terapêutico , Pessoa de Meia-Idade , Abandono do Hábito de Fumar/psicologia , Uso de Tabaco/epidemiologia , Uso de Tabaco/psicologia , Tabagismo/diagnóstico , Tabagismo/psicologia , Adulto Jovem
19.
Exp Clin Psychopharmacol ; 26(3): 310-319, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29863387

RESUMO

No medications are approved for cannabis use disorder (CUD). Gamma-aminobutyric acid (GABA) reuptake is modulated by cannabinoid (CB) receptor agonists, and there are shared effects between CB agonists and the GABA reuptake inhibitor tiagabine. This overlapping neuropharmacology suggested that tiagabine might be useful for CUD. The study determined the ability of tiagabine maintenance to reduce cannabis self-administration using a placebo-controlled, double-blind, counterbalanced, within-subjects design. Nontreatment-seeking daily cannabis users (N = 12; 3 female, 9 male) completed two 12-day outpatient maintenance phases (0 or 12 mg of tiagabine/day). Each phase consisted of a safety session, 7 maintenance days, and 4 experimental sessions. During experimental sessions, maintenance continued and participants completed two 2-day blocks of sampling and self-administration sessions to determine the reinforcing effects of smoked cannabis (0% and 5.9% Δ9-tetrahydrocannabinol). Naturalistic cannabis use, the subjective, performance and physiological response to cannabis, as well as side effects, sleep quality, craving, other self-reported substance use, and observer ratings were also measured. Cannabis functioned as a reinforcer and produced prototypical effects (e.g., increased heart rate and ratings of "high"), but tiagabine generally did not impact the effects of cannabis, or alter naturalistic use. Furthermore, tiagabine produced small, but significant, increases on 2 subscales of a Marijuana Craving Questionnaire, and reductions in both the amount of time slept in the past 24 hr and ratings of positive mood upon awakening. These human laboratory results from a sample of nontreatment-seeking cannabis users do not support the potential efficacy of 12 mg of tiagabine as a stand-alone pharmacotherapy for CUD. (PsycINFO Database Record


Assuntos
Fissura/efeitos dos fármacos , Abuso de Maconha/tratamento farmacológico , Abuso de Maconha/psicologia , Fumar Maconha/tratamento farmacológico , Fumar Maconha/psicologia , Tiagabina/administração & dosagem , Adulto , Afeto/efeitos dos fármacos , Afeto/fisiologia , Agonistas de Receptores de Canabinoides/administração & dosagem , Agonistas de Receptores de Canabinoides/farmacologia , Cannabis/efeitos adversos , Fissura/fisiologia , Método Duplo-Cego , Dronabinol/administração & dosagem , Feminino , Inibidores da Captação de GABA/administração & dosagem , Alucinógenos/administração & dosagem , Humanos , Masculino , Abuso de Maconha/diagnóstico , Uso da Maconha/psicologia , Reforço Psicológico , Autoadministração , Sono/efeitos dos fármacos , Sono/fisiologia , Inquéritos e Questionários
20.
Addict Behav ; 85: 26-30, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29803870

RESUMO

Relative to adults, adolescents are at greater risk of developing a cannabis use disorder (CUD) and risk may be exacerbated by co-occurring depressive symptoms. N-Acetylcysteine (NAC), an over-the-counter antioxidant, is thought to normalize glutamate transmission. Oxidative stress and glutamate transmission are disrupted in both depression and CUD. Thus, NAC may be particularly effective at promoting cannabis abstinence among adolescents with elevated depressive symptoms. Secondary analyses were conducted using a sub-sample of adolescents with CUD (N = 74) who participated in an 8-week randomized placebo-controlled clinical trial examining the efficacy of NAC for cannabis cessation. It was hypothesized that NAC would reduce severity of depressive symptoms, and that decreases depressive symptom severity would mediate decreases in positive weekly urine cannabinoid tests (11-nor-9-carboxy-Δ9-tetrahydrocannabinol). Additionally, it was expected that adolescents with greater severity of baseline depressive symptoms would be more likely to become abstinent when assigned NAC relative to placebo. Results from linear mixed models and generalized estimating equations did not suggest that NAC reduced severity of depressive symptoms, and the hypothesis that NAC's effect on cannabis cessation would be mediated by reduced depressive symptoms was not supported. However, an interaction between treatment condition and baseline severity of depressive symptoms as a predictor of weekly urine cannabinoid tests was significant, suggesting that NAC was more effective at promoting abstinence among adolescents with heightened baseline depressive symptoms. These secondary findings, though preliminary, suggest a need for further examination of the role of depressive symptoms in treatment of adolescent CUD with NAC.


Assuntos
Acetilcisteína/uso terapêutico , Depressão/psicologia , Sequestradores de Radicais Livres/uso terapêutico , Abuso de Maconha/tratamento farmacológico , Adolescente , Feminino , Humanos , Masculino , Abuso de Maconha/psicologia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA