Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Pharmacol Rev ; 2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38849155

RESUMO

Cannabis is one of the oldest and widely used substances in the world. Cannabinoids within the cannabis plant, known as phytocannabinoids, mediate cannabis' effects through interactions with the body's endogenous cannabinoid system. This endogenous system, the endocannabinoid system, has important roles in physical and mental health. These roles point to the potential to develop cannabinoids as therapeutic agents, while underscoring the risks related to interfering with the endogenous system during non-medical use. This scoping narrative review synthesizes the current evidence for both the therapeutic and adverse effects of the major (i.e., Δ9-tetrahydrocannabinol and cannabidiol) and lesser studied minor phytocannabinoids, from nonclinical to clinical research. We pay particular attention to the areas where evidence is well-established, including analgesic effects after acute exposures and neurocognitive risks after acute and chronic use. In addition, drug development considerations for cannabinoids as therapeutic agents within the United States are reviewed. The proposed clinical study design considerations encourage methodological standards for greater scientific rigor and reproducibility, ultimately, to extend our knowledge of the risks and benefits of cannabinoids for patients and providers. Significance Statement This work provides a review of prior research related to phytocannabinoids, including therapeutic potential and known risks in the context of drug development within the United States. We also provide study design considerations for future cannabinoid drug development.

2.
Psychol Med ; 54(2): 245-255, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37882050

RESUMO

Cannabis use is consistently associated with both increased incidence of frank psychotic disorders and acute exacerbations of psychotic symptoms in healthy individuals and people with psychosis spectrum disorders. Although there is uncertainty around causality, cannabis use may be one of a few modifiable risk factors for conversion to psychotic disorders in individuals with Clinical High Risk for Psychosis (CHR-P) syndromes, characterized by functionally impairing and distressing subthreshold psychotic symptoms. To date, few recommendations beyond abstinence to reduce adverse psychiatric events associated with cannabis use have been made. This narrative review synthesizes existing scientific literature on cannabis' acute psychotomimetic effects and epidemiological associations with psychotic disorders in both CHR-P and healthy individuals to bridge the gap between scientific knowledge and practical mental health intervention. There is compelling evidence for cannabis acutely exacerbating psychotic symptoms in CHR-P, but its impact on conversion to psychotic disorder is unclear. Current evidence supports a harm reduction approach in reducing frequency of acute psychotic-like experiences, though whether such interventions decrease CHR-P individuals' risk of conversion to psychotic disorder remains unknown. Specific recommendations include reducing frequency of use, lowering delta-9-tetrahydrocannabinol content in favor of cannabidiol-only products, avoiding products with inconsistent potency like edibles, enhancing patient-provider communication about cannabis use and psychotic-like experiences, and utilizing a collaborative and individualized therapeutic approach. Despite uncertainty surrounding cannabis' causal association with psychotic disorders, cautious attempts to reduce acute psychosis risk may benefit CHR-P individuals uninterested in abstinence. Further research is needed to clarify practices associated with minimization of cannabis-related psychosis risk.


Assuntos
Cannabis , Alucinógenos , Abuso de Maconha , Transtornos Psicóticos , Humanos , Adolescente , Cannabis/efeitos adversos , Redução do Dano , Abuso de Maconha/complicações , Transtornos Psicóticos/psicologia , Fatores de Risco , Agonistas de Receptores de Canabinoides
3.
Harm Reduct J ; 21(1): 82, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38622670

RESUMO

BACKGROUND: Cannabis use before the COVID-19 pandemic for many involved sharing prepared cannabis for inhalation, practices that were less prevalent during the pandemic. State-level COVID-19 containment policies may have influenced this decrease. This study examined the extent to which the intensity of state-level COVID-19 policies were associated with individual-level cannabis sharing. Findings have the potential to guide harm reduction policies for future respiratory pandemics and seasonal respiratory virus waves. METHODS: This study used cross-sectional individual-level data from the COVID-19 Cannabis Study, an anonymous U.S.-based web survey on cannabis use disseminated during the early phase of the pandemic (Full sample N = 1,883). We combined individual-level data with state-level policy data from Kaiser Family Foundation's State COVID-19 Data and Policy Actions for three time-points from June to August 2020 that overlapped with the survey period. Cannabis sharing was dichotomized as any versus no sharing. We adapted a previously published coding framework to score the intensity of COVID-19 policies implemented in each U.S. state and averaged the policy score across the time period. We then used Poisson regression models to quantify the associations of the average state-level COVID-19 policy score with cannabis sharing during the pandemic. RESULTS: Participants (n = 925) reporting using inhalation as a mode for cannabis use were included in this analysis. Most respondents were male (64.1%), non-Hispanic White (54.3%), with a mean age of 33.7 years (SD 8.8). A large proportion (74.9%) reported sharing cannabis during the pandemic. Those who shared cannabis more commonly lived in states with a lower average policy score (16.7, IQR 12.3-21.5) compared to those who did not share (18.6, IQR 15.3-25.3). In adjusted models, the prevalence ratio of any cannabis sharing per every 5-unit increase in the average COVID-19 policy score was 0.97 (95% CI 0.93, 1.01). CONCLUSIONS: Fewer individuals shared cannabis in states with more intense COVID-19 containment policies compared to those in states with less intense policies. Individuals who use cannabis may be willing to make changes to their behavior and may further benefit from specific and directed public health messaging to avoid sharing during respiratory infection outbreaks.


Assuntos
COVID-19 , Cannabis , Alucinógenos , Masculino , Humanos , Adulto , Feminino , Estudos Transversais , Pandemias , COVID-19/prevenção & controle , Políticas
4.
Addict Biol ; 28(7): e13301, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37369126

RESUMO

Preclinical studies have shown sex-based differences in the reinforcing effects of cannabinoid 1 receptor agonists such as delta-9-tetrahydrocannabinol (THC). This study sought to test whether these sex differences translate to humans by assessing the subjective and reinforcing effects of smoked cannabis in male and female volunteers. We pooled data (n = 68; 55M, 13F) from two within-subject randomized controlled trials of healthy, ≥weekly cannabis users comparing the subjective and reinforcing effects of smoked active (~25 mg THC) versus placebo cannabis (0-mg THC). Subjective ratings of drug effects and mood were measured using visual analogue scales, and reinforcing effects were measured with a cannabis self-administration task. Sex-dependent outcomes were explored using generalized linear mixed models. Under active cannabis conditions, female participants reported greater reductions from baseline in cannabis craving and significantly higher cannabis-specific ratings of strength, liking, willingness to take again and good effect, compared with males (interaction p < 0.05). Placebo and active cannabis were self-administered by 22% and 36% of male participants, respectively, and by 15% and 54% of female participants, respectively. Receipt of active cannabis significantly increased likelihood of self-administration (p = 0.011), but a sex difference was not detected (p = 0.176). Although females were more sensitive to certain positive subjective effects of active cannabis, they were not more likely than males to self-administer it. These findings highlight the need to test sex differences as a primary objective in experimental studies and may shed light on accelerated trajectories from initiation to cannabis use disorder observed among women.


Assuntos
Cannabis , Fumar Maconha , Feminino , Humanos , Masculino , Afeto , Agonistas de Receptores de Canabinoides/farmacologia , Ensaios Clínicos Controlados Aleatórios como Assunto
5.
Harm Reduct J ; 20(1): 23, 2023 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-36829150

RESUMO

BACKGROUND: Prior to the COVID-19 pandemic, cannabis use social practices often involved sharing prepared cannabis (joints/blunts/cigarettes) and cannabis-related paraphernalia. Previous studies have demonstrated that sharing paraphernalia for cannabis, tobacco, and crack cocaine is a risk factor for respiratory viral and bacterial infections. Although COVID-19 is a respiratory viral infection that spreads through droplets and airborne transmission, it is unclear if many individuals adopted harm reduction practices around sharing cannabis. This study: quantifies the prevalence of sharing prepared non-medical cannabis and cannabis-related paraphernalia reported before and during the pandemic; assesses changes in sharing of non-medical cannabis from before to during the pandemic; assess the association between frequency of non-medical cannabis use and sharing of cannabis during the pandemic; and describes how respondents obtained their cannabis and the reasons for changing their cannabis use during the pandemic to explain differences in sharing patterns. METHODS: This cross-sectional study used data collected from an anonymous, US-based web survey on cannabis-related behaviors from August to September 2020 (n = 1833). Participants were included if they reported using a mode of inhalation for non-medical cannabis consumption. We calculated proportional changes in sharing cannabis before/during the COVID-19 pandemic. Associations between frequency of cannabis use and cannabis sharing during the COVID-19 pandemic were assessed using logistic regression analysis. RESULTS: Overall, 1,112 participants reported non-medical cannabis use; 925 (83.2%) reported a mode of cannabis inhalation. More respondents reported no sharing during (24.9%) than before the pandemic (12.4%; p < 0.01); less respondents shared most of the time (19.5% before; 11.2% during; p < 0.01) and always during the pandemic (5.2% before; 3.1% during; p < 0.01). After adjusting for covariates, the odds of any sharing during the pandemic for those who reported ≥ weekly cannabis use was 0.53 (95% CI 0.38, 0.75) compared to those who reported ≤ monthly. CONCLUSIONS: Sharing of prepared cannabis and cannabis-related paraphernalia decreased during the COVID-19 pandemic compared to before the pandemic. This finding suggests potential risk mitigation strategies taken by participants for COVID-19 prevention either directly through behavior change or indirectly through adherence to COVID-19 prevention recommendations. Harm reduction messaging around sharing of cannabis during surges of COVID-19 or other respiratory infections may provide benefit in reducing infection among those who use cannabis, especially as cannabis use in the USA continues to increase.


Assuntos
COVID-19 , Cannabis , Humanos , Pandemias , Redução do Dano , Estudos Transversais
6.
Subst Use Misuse ; 58(9): 1080-1089, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37158563

RESUMO

BACKGROUND: Some patterns of cannabis use may presage risk for long-term negative effects. We examined associations between a novel adolescent cannabis misuse scale and early-adult life course outcomes. METHODS: We performed a secondary data analysis of a cohort of Los Angeles, CA high school students from grade 9 through age 21. Participants reported baseline individual demographic and family characteristics at grade 9, adolescent cannabis misuse (8-items) and alcohol misuse (12-items) at grade 10, and outcomes at age 21. We used multivariable regression to model the associations of cannabis misuse scale score with problem substance use (defined as any of: 30-day illegal drug use, 30-day use of another's prescription to get high, hazardous drinking) and several secondary outcomes (behavioral, mental health, academic, social determinants of health), adjusting for covariates. Parallel analyses were conducted for alcohol misuse. RESULTS: The 1,148 participants (86% retention) were 47% male, 90% Latinx, 87% US born, and 40% native English speakers. Approximately 11.4% and 15.9% of participants reported at least one item on the cannabis and alcohol misuse scales, respectively. At age 21, approximately 6.7% of participants reported problem substance use, which was associated with both Cannabis and Alcohol Misuse Scales (OR 1.31, 95%CI[1.16, 1.49] and OR 1.33, 95%CI[1.18, 1.49], respectively). Both scales were similarly associated with outcomes in all four categories. CONCLUSIONS: The Adolescent Cannabis Misuse Scale is a promising tool for identifying early patterns of substance use that predict future negative outcomes and enabling early intervention at a critical period in youth development.


Assuntos
Alcoolismo , Cannabis , Transtornos Relacionados ao Uso de Substâncias , Adulto , Humanos , Masculino , Adolescente , Adulto Jovem , Feminino , Saúde Mental , Determinantes Sociais da Saúde , Transtornos Relacionados ao Uso de Substâncias/psicologia
7.
Br J Clin Pharmacol ; 88(1): 347-355, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34223660

RESUMO

AIMS: Preclinical studies demonstrate that cannabidiol (CBD) elicits an antinociceptive response in animal models of neuropathic pain; in humans, limited data are available to support such analgesic effects. Few studies have examined CBD's analgesic effects when administered without other compounds, and little is known regarding dose-dependent effects in noncannabis users. METHODS: This double-blind, placebo-controlled, within-subject outpatient clinical laboratory study sought to determine the analgesic effects, abuse liability, safety and tolerability of acute CBD (0, 200, 400 and 800 mg orally) in healthy noncannabis-using volunteers (n = 17; 8 men, 9 women). Outcomes included experimental pain threshold and pain tolerance using the cold pressor test (CPT), subjective ratings of CPT painfulness and bothersomeness, subjective ratings of abuse liability and mood, and cardiovascular measures, which were assessed at baseline and several time points after drug administration. Data analyses included repeated measures analysis of variance (ANOVA) with planned comparisons. RESULTS: CBD failed to consistently affect pain threshold and tolerance in the CPT relative to placebo. All doses of CBD increased ratings of painfulness compared to placebo (P < .01). Further, CBD had dose-dependent, modest effects on mood and subjective drug effects associated with abuse liability. Oral CBD was safe and well tolerated, producing small decreases in blood pressure (P < .01). CONCLUSION: CBD did not elicit consistent dose-dependent analgesia and in fact increased pain on some measures. Future studies exploring CBD-induced pain relief should consider using a more extensive pain assessment paradigm in different participant populations.


Assuntos
Analgesia , Canabidiol , Analgésicos/efeitos adversos , Canabidiol/farmacologia , Método Duplo-Cego , Feminino , Humanos , Masculino , Dor/induzido quimicamente , Dor/tratamento farmacológico , Medição da Dor
8.
Addict Biol ; 27(4): e13183, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35754107

RESUMO

Attenuating enzymatic degradation of endocannabinoids (eCBs) by fatty acid amide hydrolase (FAAH) reduces cannabis withdrawal symptoms in preclinical and clinical studies. In mice, blocking cyclooxygenase-2 (COX-2) activity increases central eCB levels by inhibiting fatty acid degradation. This placebo-controlled study examined the effects of the FDA-approved COX-2 selective inhibitor, celecoxib, on cannabis withdrawal, 'relapse', and circulating eCBs in a human laboratory model of cannabis use disorder. Daily, nontreatment-seeking cannabis smokers (12M, 3F) completed a crossover study comprising two 11-day study phases (separated by >14 days for medication clearance). In each phase, the effects of daily BID placebo (0 mg) or celecoxib (200 mg) on cannabis (5.3% THC) intoxication, withdrawal symptoms (4 days of inactive cannabis self-administration) and 'relapse' (3 days of active cannabis self-administration following abstinence) were assessed. Outcome measures included mood, cannabis self-administration, sleep, food intake, cognitive performance, tobacco cigarette use and circulating eCBs and related lipids. Under placebo maintenance, cannabis abstinence produced characteristic withdrawal symptoms (negative mood, anorexia and dreaming) relative to cannabis administration and was associated with increased OEA (a substrate of FAAH) and oleic acid (metabolite of OEA), with no change in eCB levels. Compared to placebo, celecoxib improved subjective (but not objective) measures of sleep and did not affect mood or plasma levels of eCBs or associated lipids and increased cannabis craving. The overall absence of effects on cannabis withdrawal symptoms, self-administration or circulating eCBs relative to placebo, combined with an increase in cannabis craving, suggests celecoxib does not show promise as a potential pharmacotherapy for CUD.


Assuntos
Cannabis , Abuso de Maconha , Síndrome de Abstinência a Substâncias , Agonistas de Receptores de Canabinoides , Celecoxib/uso terapêutico , Estudos Cross-Over , Ciclo-Oxigenase 2/uso terapêutico , Dronabinol , Endocanabinoides , Humanos , Abuso de Maconha/psicologia , Recidiva , Fumantes , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Síndrome de Abstinência a Substâncias/psicologia
9.
Am J Drug Alcohol Abuse ; 48(3): 321-327, 2022 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-35130464

RESUMO

Background: During the SARS-CoV-2 pandemic, cannabis dispensaries in the United States (US) reported increased sales.Objectives: This study assessed changes in cannabis use before to during the pandemic; it was hypothesized that cannabis use increased during the pandemic.Methods: A US-based survey of adults assessed patterns of use before and during the pandemic. Participants recruited via social media from 8/2020 to 9/2020 self-reported medical and non-medical cannabis use. Use was categorized as ≤ monthly, weekly, and daily/almost daily; adjusted logistic regressions determined associations between cannabis use before the pandemic with change in frequency during the pandemic.Results: Of 1,886 respondents (63% male),1,113 reported non-medical and 603 medical use of cannabis. Those reporting ≤monthly non-medical use before the pandemic had higher odds of increasing use during the pandemic than those using weekly before the pandemic (AOR 3.2 [95% CI 2.2-4.5]). Those reporting ≤ monthly and daily medical cannabis use before the pandemic had higher odds of increasing use during the pandemic than those using weekly before the pandemic (AOR 2.3 [95% 1.3, 3.9]; AOR 2.4 [95% CI 1.2, 5.1] respectively).Conclusions: The most notable increases in cannabis use during the pandemic were among those who reported using cannabis least frequently before the pandemic (two to three times odds of increased use among ≤ monthly use compared to weekly). These findings have important implications for potential health consequences related to increased cannabis use both during and after the pandemic, even in populations thought to be protected by minimal use prior to the pandemic.


Assuntos
COVID-19 , Cannabis , Maconha Medicinal , Adulto , Feminino , Humanos , Masculino , Maconha Medicinal/uso terapêutico , Pandemias , SARS-CoV-2 , Estados Unidos/epidemiologia
10.
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
11.
Int J Clin Pract ; 75(8): e13871, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33249713

RESUMO

AIMS: Opioid misuse and overuse have contributed to a widespread overdose crisis and many patients and physicians are considering medical cannabis to support opioid tapering and chronic pain control. Using a five-step modified Delphi process, we aimed to develop consensus-based recommendations on: 1) when and how to safely initiate and titrate cannabinoids in the presence of opioids, 2) when and how to safely taper opioids in the presence of cannabinoids and 3) how to monitor patients and evaluate outcomes when treating with opioids and cannabinoids. RESULTS: In patients with chronic pain taking opioids not reaching treatment goals, there was consensus that cannabinoids may be considered for patients experiencing or displaying opioid-related complications, despite psychological or physical interventions. There was consensus observed to initiate with a cannabidiol (CBD)-predominant oral extract in the daytime and consider adding tetrahydrocannabinol (THC). When adding THC, start with 0.5-3 mg, and increase by 1-2 mg once or twice weekly up to 30-40 mg/day. Initiate opioid tapering when the patient reports a minor/major improvement in function, seeks less as-needed medication to control pain and/or the cannabis dose has been optimised. The opioid tapering schedule may be 5%-10% of the morphine equivalent dose (MED) every 1 to 4 weeks. Clinical success could be defined by an improvement in function/quality of life, a ≥30% reduction in pain intensity, a ≥25% reduction in opioid dose, a reduction in opioid dose to <90 mg MED and/or reduction in opioid-related adverse events. CONCLUSIONS: This five-stage modified Delphi process led to the development of consensus-based recommendations surrounding the safe introduction and titration of cannabinoids in concert with tapering opioids.


Assuntos
Canabinoides , Dor Crônica , Analgésicos Opioides , Dor Crônica/tratamento farmacológico , Consenso , Humanos , Qualidade de Vida
12.
Am J Drug Alcohol Abuse ; 47(5): 535-547, 2021 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-34280058

RESUMO

BACKGROUND: While males are more likely diagnosed with cannabis use disorder (CUD), females are more susceptible to developing and maintaining CUD. Yet, for both sexes, CUD is associated with high rates of comorbid mental illness (MI). OBJECTIVES: To identify and compare sex differences in the prevalence of comorbid CUD amongst individuals with/without MIs. METHODS: This systematic review generated pooled odds ratios (OR) and 95% confidence intervals (CI) from 37 studies (including clinical trials, cohort, and case-control studies) among individuals with and without MIs, quantifying sex differences in rates of comorbid CUD. A meta-analysis was also completed. RESULTS: In the CUD-only group, males were twice as likely to have CUD than females (OR = 2.0, CI = 1.9-2.1). Among MIs, males were more likely than females to have CUD comorbid with schizophrenia (OR ~2.6, CI = 2.5-2.7) and other psychotic, mood, and substance use disorders (1> OR <2.2, CI = 0.7-2.6). The reverse association (females > males) was observed for anxiety disorders and antisocial personality disorder (OR = 0.8, CI = 0.7-1.0). Among females, MIs increased the likelihood of having CUD, except for psychotic disorders and depression. A meta-analysis was inconclusive due to high heterogeneity across studies. Thus, comparisons across MI groups were not possible. CONCLUSION: While males are more likely to be diagnosed with CUD, there are important sex differences in the prevalence of CUD across MI diagnoses that should be taken into account when approaching CUD prevention and determining treatment efficacy.


Assuntos
Abuso de Maconha/epidemiologia , Transtornos Mentais/epidemiologia , Comorbidade , Feminino , Humanos , Masculino , Razão de Chances , Prevalência , Distribuição por Sexo , Fatores Sexuais
13.
Alcohol Alcohol ; 55(4): 416-423, 2020 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-32328657

RESUMO

BACKGROUND: Alcohol and cannabis are frequently co-used, as 20-50% of those who drink alcohol report co-using cannabis. This study is based on the argument that alcohol researchers should enroll cannabis users in human laboratory studies of alcohol use disorder (AUD) to strengthen generalizability. This study examines how heavy drinking cannabis users differ from non-cannabis using heavy drinkers. METHODS: In a community sample of non-treatment-seeking heavy drinkers (n = 551, 35% female), cannabis users were identified through: (a) self-reported cannabis use in the past 6 months and (b) positive urine toxicology test for tetrahydrocannabinol (THC). Cannabis users, identified as described previously, were compared with non-cannabis users on demographic and clinical characteristics. RESULTS: Those who endorsed cannabis use in the past 6 months reported more binge drinking days. Participants who tested positive for THC had higher Alcohol Use Disorder Identification Test scores and more binge drinking days. Younger age and being a tobacco smoker were associated with an increased likelihood of cannabis use in the past 6 months, whereas male gender and being a tobacco use were associated with a greater likelihood of testing positive for THC. Individuals with cannabis use disorder (CUD) endorsed more depression and anxiety and had higher AUD symptom counts than cannabis users without CUD. CONCLUSIONS: The inclusion of cannabis users in AUD samples allows for increased clinical severity. Excluding cannabis users from AUD studies may limit representativeness and expend unnecessary study resources. Lastly, tobacco use may explain a large portion of the effects of cannabis use on sample characteristics. SHORT SUMMARY: Alcohol and cannabis are frequently co-used substances. In a sample of non-treatment-seeking heavy drinkers (n = 551, 35% female), cannabis users reported higher alcohol use and higher likelihood of tobacco use than non-cannabis users. Including cannabis users in alcohol research studies will improve representativeness and likely increase clinical severity.


Assuntos
Alcoolismo/complicações , Alcoolismo/diagnóstico , Fumar Maconha/urina , Programas de Rastreamento/métodos , Sujeitos da Pesquisa , Adulto , Feminino , Humanos , Masculino , Autorrelato
14.
PLoS Med ; 16(11): e1002967, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31743343

RESUMO

BACKGROUND: Ecological research suggests that increased access to cannabis may facilitate reductions in opioid use and harms, and medical cannabis patients describe the substitution of opioids with cannabis for pain management. However, there is a lack of research using individual-level data to explore this question. We aimed to investigate the longitudinal association between frequency of cannabis use and illicit opioid use among people who use drugs (PWUD) experiencing chronic pain. METHODS AND FINDINGS: This study included data from people in 2 prospective cohorts of PWUD in Vancouver, Canada, who reported major or persistent pain from June 1, 2014, to December 1, 2017 (n = 1,152). We used descriptive statistics to examine reasons for cannabis use and a multivariable generalized linear mixed-effects model to estimate the relationship between daily (once or more per day) cannabis use and daily illicit opioid use. There were 424 (36.8%) women in the study, and the median age at baseline was 49.3 years (IQR 42.3-54.9). In total, 455 (40%) reported daily illicit opioid use, and 410 (36%) reported daily cannabis use during at least one 6-month follow-up period. The most commonly reported therapeutic reasons for cannabis use were pain (36%), sleep (35%), stress (31%), and nausea (30%). After adjusting for demographic characteristics, substance use, and health-related factors, daily cannabis use was associated with significantly lower odds of daily illicit opioid use (adjusted odds ratio 0.50, 95% CI 0.34-0.74, p < 0.001). Limitations of the study included self-reported measures of substance use and chronic pain, and a lack of data for cannabis preparations, dosages, and modes of administration. CONCLUSIONS: We observed an independent negative association between frequent cannabis use and frequent illicit opioid use among PWUD with chronic pain. These findings provide longitudinal observational evidence that cannabis may serve as an adjunct to or substitute for illicit opioid use among PWUD with chronic pain.


Assuntos
Dor Crônica/tratamento farmacológico , Maconha Medicinal/uso terapêutico , Transtornos Relacionados ao Uso de Opioides/epidemiologia , Adulto , Analgésicos/uso terapêutico , Analgésicos Opioides/uso terapêutico , Canadá , Cannabis , Feminino , Humanos , Estudos Longitudinais , Masculino , Fumar Maconha/epidemiologia , Pessoa de Meia-Idade , Dor/tratamento farmacológico , Manejo da Dor/métodos , Manejo da Dor/tendências , Estudos Prospectivos
15.
Addict Biol ; 24(4): 707-716, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-29659126

RESUMO

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.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Guanfacina/uso terapêutico , Abuso de Maconha , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Adulto , Afeto , Anorexia/etiologia , Anorexia/fisiopatologia , Pressão Sanguínea , Cannabis/efeitos adversos , Comportamento Alimentar , Feminino , Humanos , Humor Irritável , Masculino , Desempenho Psicomotor , Autoadministração , Sono , Síndrome de Abstinência a Substâncias/etiologia , Síndrome de Abstinência a Substâncias/fisiopatologia , Síndrome de Abstinência a Substâncias/psicologia , Adulto Jovem
16.
Addict Biol ; 24(5): 1044-1055, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30328665

RESUMO

Illicit drug use among aging cohorts is increasing, yet little is known about functional impairments in older drug users. Given the importance of social integration for aging and documented social decrements in cocaine users, we examined social function and its neurocognitive substrates in aging cocaine users relative to carefully matched non-cocaine users. Regular (≥twice/week), long-term (≥15 years) cocaine smokers 50-60 years old (COCs; n = 22; four women) and controls (CTRLs; n = 19; four women) underwent standardized probes of social reward and threat processing during functional magnetic resonance imaging and a behavioral facial affect recognition task. Self-report and peer-report of daily interpersonal function were also collected. COCs, and CTRLs reporting current marijuana or alcohol use, were tested after four drug-free inpatient days. COCs had pronounced problems in daily social function relative to CTRLs indicated by both their own and their peers' reports. Compared with CTRLs, COCs had stronger amygdala responses to social threat versus control stimuli, with no other differences in social processing or cognition. Aging cocaine users appear to have marked, generalized difficulties in 'real-world' interpersonal function but largely intact social processing on laboratory-based measures when compared with appropriately matched controls and tested under well-controlled conditions. Daily social difficulties may be related to transient factors such as acute/residual drug effects or cocaine-related changes in health behaviors (e.g. disrupted sleep and poor diet). These data suggest that interpersonal function may be a valid intervention target for aging cocaine users and warrants further study in older drug users.


Assuntos
Envelhecimento , Encéfalo/diagnóstico por imagem , Fumar Cocaína/psicologia , Transtornos Relacionados ao Uso de Cocaína/diagnóstico por imagem , Reconhecimento Facial , Recompensa , Habilidades Sociais , Afeto , Encéfalo/fisiopatologia , Estudos de Casos e Controles , Fumar Cocaína/fisiopatologia , Transtornos Relacionados ao Uso de Cocaína/fisiopatologia , Transtornos Relacionados ao Uso de Cocaína/psicologia , Expressão Facial , Feminino , Neuroimagem Funcional , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Motivação , Autorrelato , Comportamento Social
17.
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
18.
Am J Drug Alcohol Abuse ; 45(6): 580-595, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31687845

RESUMO

Background: Pain is the most frequent indication for which medical cannabis treatment is sought.Objectives: The clinical potential of cannabis and cannabis-derived products (CDPs) relies on their efficacy to treat an indication and potential adverse effects that impact outcomes, including abuse liability and neurocognitive effects. To ascertain the extent to which these effects impact therapeutic utility, studies investigating cannabis and CDPs for pain were reviewed for analgesic efficacy and assessments of abuse liability and neurocognitive effects.Methods: A comprehensive review of placebo-controlled studies investigating cannabis and CDP analgesia was performed. Methods and findings related to adverse effects, abuse liability, and neurocognitive effects were extracted.Results: Thirty-eight studies were reviewed; 29 assessed cannabis and CDPs for chronic pain, 1 for acute pain, and 8 used experimental pain tests. Most studies ascertained adverse effects through self-report (N = 27). Fewer studies specifically probed abuse liability (N = 7) and cognitive and psychomotor effects (N = 12). Many studies related to chronic and experimental pain (N = 18 and N = 5, respectively) found cannabis and CDPs to reduce pain. Overall, adverse effects were mild to moderate, and dose-related. Studies investigating the impact of cannabis and CDPs on abuse liability and neurocognitive endpoints were mostly limited to inhaled administration and confirmed dose-related effects.Conclusion: Few studies investigating cannabis and CDP analgesia assess abuse liability and cognitive endpoints, adverse effects that impact the long-term clinical utility of these drugs. Future studies should include these measures to optimize research and clinical care related to cannabis-based therapeutics.


Assuntos
Analgésicos não Narcóticos/uso terapêutico , Canabinoides/uso terapêutico , Dor Crônica/tratamento farmacológico , Maconha Medicinal/uso terapêutico , Dor Aguda/tratamento farmacológico , Canabidiol/uso terapêutico , Dronabinol/uso terapêutico , Humanos , Abuso de Maconha/etiologia , Testes de Estado Mental e Demência , Desempenho Psicomotor , Ensaios Clínicos Controlados Aleatórios como Assunto
20.
Curr Psychiatry Rep ; 18(5): 52, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27074934

RESUMO

Although several chemical structural classes of synthetic cannabinoids (SCs) were recently classified as Schedule I substances, rates of use and cases of serious toxic effects remain high. While case reports and media bring attention to severe SC toxicity, daily SC use resulting in dependence and withdrawal is a significant concern that is often overlooked when discussing the risks of these drugs. There is a rich literature on evidence-based approaches to treating substance use disorders associated with most abused drugs, yet little has been published regarding how to best treat symptoms related to SC dependence given its recency as an emerging clinically significant issue. This review provides a background of the pharmacology of SCs, recent findings of adverse effects associated with both acute intoxication and withdrawal as a consequence of daily use, and treatment approaches that have been implemented to address these issues, with an emphasis on pharmacotherapies for managing detoxification. In order to determine prevalence of use in cannabis smokers, a population at high risk for SC use, we obtained data on demographics of SC users, frequency of use, and adverse effects over a 3.5-year period (2012-2015) in the New York City metropolitan area, a region with a recent history of high SC use. While controlled studies on the physiological and behavioral effects of SCs are lacking, it is clear that risks associated with using these drugs pertain not only to the unpredictable and severe nature of acute intoxication but also to the effects of long-term, chronic use. Recent reports in the literature parallel findings from our survey, indicating that there is a subset of people who use SCs daily. Although withdrawal has not been systematically characterized and effective treatments have yet to be elucidated, some symptom relief has been reported with benzodiazepines and the atypical antipsychotic, quetiapine. Given the continued use and abuse of SCs, empirical studies characterizing (1) SCs acute effects, (2) withdrawal upon cessation of use, and (3) effective treatment strategies for SC use disorder are urgently needed.


Assuntos
Canabinoides/toxicidade , Drogas Ilícitas/toxicidade , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Humanos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa