Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Drug Alcohol Depend ; 257: 111267, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38498958

ABSTRACT

BACKGROUND: Cannabis contains hundreds of chemical constituents beyond delta-9-tetrahydrocannabinol (THC), which is believed to drive most of its acute pharmacodynamic effects. The entourage effect theory asserts that non-THC constituents can impact acute cannabis effects, but few empirical studies have systematically evaluated this theory in humans. This study assessed whether the cannabis terpenoid d-limonene mitigates the acute anxiogenic effects of THC. METHODS: Twenty healthy adults completed nine, double-blind outpatient sessions in which they inhaled vaporized THC alone (15mg or 30mg), d-limonene alone (1mg or 5mg), the same doses of THC and d-limonene together, or placebo; a subset of participants (n=12) completed a tenth session in which 30mg THC+15mg d-limonene was administered. Outcomes included subjective drug effects, cognitive/psychomotor performance, vital signs, and plasma THC and d-limonene concentrations. RESULTS: When d-limonene was administered alone, pharmacodynamic outcomes did not differ from placebo. Administration of 15mg and 30mg THC alone produced subjective, cognitive, and physiological effects typical of acute cannabis exposure. Ratings of anxiety-like subjective effects qualitatively decreased as d-limonene dose increased and concurrent administration of 30mg THC+15mg d-limonene significantly reduced ratings of "anxious/nervous" and "paranoid" compared with 30mg THC alone. Other pharmacodynamic effects were unchanged by d-limonene. D-limonene plasma concentrations were dose orderly, and concurrent administration of d-limonene did not alter THC pharmacokinetics. CONCLUSIONS: D-limonene selectively attenuated THC-induced anxiogenic effects, suggesting this terpenoid could increase the therapeutic index of THC. Future research should determine whether this effect extends to oral dose formulations and evaluate the interactions between other cannabis terpenoids or cannabinoids and THC.


Subject(s)
Cannabinoids , Cannabis , Hallucinogens , Adult , Humans , Cannabis/adverse effects , Dronabinol/adverse effects , Limonene , Cannabinoid Receptor Agonists , Double-Blind Method , Plant Extracts
2.
Clin Pharmacol Ther ; 114(3): 693-703, 2023 09.
Article in English | MEDLINE | ID: mdl-37313955

ABSTRACT

Understanding cannabis-drug interactions is critical given regulatory changes that have increased access to and use of cannabis. Cannabidiol (CBD) and Δ-9-tetrahydrocannabinol (Δ9-THC), the most abundant phytocannabinoids, are in vitro reversible and time-dependent (CBD only) inhibitors of several cytochrome P450 (CYP) enzymes. Cannabis extracts were used to evaluate quantitatively potential pharmacokinetic cannabinoid-drug interactions in 18 healthy adults. Participant received, in a randomized cross-over manner (separated by ≥ 1 week), a brownie containing (i) no cannabis extract (ethanol/placebo), (ii) CBD-dominant cannabis extract (640 mg CBD + 20 mg Δ9-THC), or (iii) Δ9-THC-dominant cannabis extract (20 mg Δ9-THC and no CBD). After 30 minutes, participants consumed a cytochrome P450 (CYP) drug cocktail consisting of caffeine (CYP1A2), losartan (CYP2C9), omeprazole (CYP2C19), dextromethorphan (CYP2D6), and midazolam (CYP3A). Plasma and urine samples were collected (0-24 hours). The CBD + Δ9-THC brownie inhibited CYP2C19 > CYP2C9 > CYP3A > CYP1A2 (but not CYP2D6) activity, as evidenced by an increase in the geometric mean ratio of probe drug area under the plasma concentration-time curve (AUC) relative to placebo (AUCGMR ) of omeprazole, losartan, midazolam, and caffeine by 207%, 77%, 56%, and 39%, respectively. In contrast, the Δ9-THC brownie did not inhibit any of the CYPs. The CBD + Δ9-THC brownie increased Δ9-THC AUCGMR by 161%, consistent with CBD inhibiting CYP2C9-mediated oral Δ9-THC clearance. Except for caffeine, these interactions were well-predicted by our physiologically-based pharmacokinetic model (within 26% of observed interactions). Results can be used to help guide dose adjustment of drugs co-consumed with cannabis products and the dose of CBD in cannabis products to reduce interaction risk with Δ9-THC.


Subject(s)
Cannabidiol , Cannabinoids , Cannabis , Hallucinogens , Humans , Adult , Cannabinoids/pharmacology , Cytochrome P-450 CYP1A2 , Cytochrome P-450 CYP2C19 , Caffeine/pharmacokinetics , Midazolam/pharmacokinetics , Cytochrome P-450 CYP3A , Losartan , Cytochrome P-450 CYP2C9 , Cytochrome P-450 Enzyme System , Cytochrome P-450 CYP2D6 , Drug Interactions , Omeprazole/pharmacokinetics , Plant Extracts/pharmacokinetics , Dronabinol/pharmacology
3.
JAMA Netw Open ; 6(2): e2254752, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36780161

ABSTRACT

Importance: Controlled clinical laboratory studies have shown that cannabidiol (CBD) can sometimes attenuate or exacerbate the effects of Δ9-tetrahydrocannabinol (Δ9-THC). No studies have evaluated differences in pharmacokinetics (PK) of Δ9-THC and pharmacodynamics (PD) between orally administered cannabis extracts that vary with respect to Δ9-THC and CBD concentrations. Objective: To compare the PK and PD of orally administered Δ9-THC-dominant and CBD-dominant cannabis extracts that contained the same Δ9-THC dose (20 mg). Design, Setting, and Participants: This randomized clinical trial was a within-participant, double-blind, crossover study conducted from January 2021 to March 2022 at the Johns Hopkins University Behavioral Pharmacology Research Unit, Baltimore, MD. Eighteen healthy adults completed 3 randomized outpatient experimental test sessions that were each separated by at least 1 week. Interventions: Brownies containing (1) no cannabis extract (ie, placebo); (2) Δ9-THC-dominant extract (20 mg Δ9-THC with no CBD); and (3) CBD-dominant extract (20 mg Δ9-THC + 640 mg CBD) were administered to participants 30 minutes prior to administering a cytochrome P450 (CYP) probe drug cocktail, which consisted of 100 mg caffeine, 20 mg omeprazole, 25 mg losartan, 30 mg dextromethorphan, and 2 mg midazolam. Main Outcomes and Measures: Change-from-baseline plasma concentrations for Δ9-THC or Δ9-THC metabolites and scores for subjective drug effects, cognitive and psychomotor performance, and vital signs. The area under the plasma vs concentration vs time curve (AUC) and maximum plasma concentration (Cmax) were determined. Results: The participant cohort of 18 adults included 11 males (61.1%) and 7 females (38.9%) with a mean (SD) age of 30 (7) years who had not used cannabis for at least 30 days prior to initiation of the study (mean [SD] day since last cannabis use, 86 [66] days). The CYP cocktail + placebo brownie and the CYP cocktail did not affect any PD assessments. Relative to CYP cocktail + Δ9-THC, CYP cocktail + Δ9-THC + CBD produced a higher Cmax and area under the plasma concentration vs time curve for Δ9-THC, 11-OH-Δ9-THC, and Δ9-THC-COOH. The CYP cocktail + Δ9-THC + CBD increased self-reported anxiety, sedation, and memory difficulty, increased heart rate, and produced a more pronounced impairment of cognitive and psychomotor performance compared with both CYP cocktail + Δ9-THC and CYP cocktail + placebo. Conclusions and Relevance: In this randomized clinical trial of oral Δ9-THC and CBD, stronger adverse effects were elicited from a CBD-dominant cannabis extract compared with a Δ9-THC-dominant cannabis extract at the same Δ9-THC dose, which contradicts common claims that CBD attenuates the adverse effects of Δ9-THC. CBD inhibition of Δ9-THC and 11-OH-Δ9-THC metabolism is the likely mechanism for the differences observed. An improved understanding of cannabinoid-cannabinoid and cannabinoid-drug interactions are needed to inform clinical and regulatory decision-making regarding the therapeutic and nontherapeutic use of cannabis products. Trial Registration: clinicaltrials.gov Identifier: NCT04201197.


Subject(s)
Cannabidiol , Cannabis , Hallucinogens , Male , Female , Humans , Adult , Dronabinol , Cross-Over Studies , Cannabinoid Receptor Agonists , Double-Blind Method , Plant Extracts
5.
Cannabis Cannabinoid Res ; 7(2): 214-223, 2022 04.
Article in English | MEDLINE | ID: mdl-33998874

ABSTRACT

Introduction: Given the increasing availability and use of cannabis among individuals with post-traumatic stress disorder (PTSD) and the addition of PTSD as an eligible diagnosis in several U.S. medical cannabis programs, the efficacy of dispensary-obtained cannabis needs to be thoroughly examined. Materials and Methods: This prospective study assessed PTSD symptoms and functioning every 3 months over the course of a year in two samples of participants diagnosed with PTSD: (1) those with PTSD using dispensary-obtained cannabis (cannabis users) and (2) those with PTSD, who do not use cannabis (controls). Linear mixed-effects models and generalized estimating equations tested whether trajectories of symptoms differed between the two subsamples. Results: A total of 150 participants (mean [standard deviation] age, 50.67 [15.26] years; 73% male) were enrolled in the study. Over the course of 1 year, the cannabis users reported a greater decrease in PTSD symptom severity over time compared to controls [group×time interaction=-0.32 (95% confidence interval [CI]=-0.59 to -0.05, R2=0.13; t=-2.35, p=0.02). Participants who used cannabis were 2.57 times more likely to no longer meet DSM-5 criteria for PTSD at the end of the study observation period compared to participants who did not use cannabis (95% CI=1.12-6.07; p=0.03). Conclusions: This study provides evidence that the types of cannabis available in recreational and medical cannabis dispensaries might hold promise as an alternative treatment for PTSD. Randomized placebo-controlled trials are needed to assess safety and determine how different preparations of cannabis impact PTSD and functioning.


Subject(s)
Cannabis , Hallucinogens , Medical Marijuana , Stress Disorders, Post-Traumatic , Cannabis/adverse effects , Female , Hallucinogens/therapeutic use , Humans , Male , Medical Marijuana/adverse effects , Middle Aged , Prospective Studies , Stress Disorders, Post-Traumatic/drug therapy
6.
Pharmacol Biochem Behav ; 199: 173059, 2020 12.
Article in English | MEDLINE | ID: mdl-33086126

ABSTRACT

The phytocannabinoid Δ9-tetrahydrocannabinol (THC) was isolated and synthesized in the 1960s. Since then, two synthetic cannabinoids (SCBs) targeting the cannabinoid 1 (CB1R) and 2 (CB2R) receptors were approved for medical use based on clinical safety and efficacy data: dronabinol (synthetic THC) and nabilone (synthetic THC analog). To probe the function of the endocannabinoid system further, hundreds of investigational compounds were developed; in particular, agonists with (1) greater CB1/2R affinity relative to THC and (2) full CB1/2R agonist activity. This pharmacological profile may pose greater risks for misuse and adverse effects relative to THC, and these SCBs proliferated in retail markets as legal alternatives to cannabis (e.g., novel psychoactive substances [NPS], "Spice," "K2"). These SCBs were largely outlawed in the U.S., but blanket policies that placed all SCB chemicals into restrictive control categories impeded research progress into novel mechanisms for SCB therapeutic development. There is a concerted effort to develop new, therapeutically useful SCBs that target novel pharmacological mechanisms. This review highlights the potential therapeutic efficacy and safety considerations for unique SCBs, including CB1R partial and full agonists, peripherally-restricted CB1R agonists, selective CB2R agonists, selective CB1R antagonists/inverse agonists, CB1R allosteric modulators, endocannabinoid-degrading enzyme inhibitors, and cannabidiol. We propose promising directions for SCB research that may optimize therapeutic efficacy and diminish potential for adverse events, for example, peripherally-restricted CB1R antagonists/inverse agonists and biased CB1/2R agonists. Together, these strategies could lead to the discovery of new, therapeutically useful SCBs with reduced negative public health impact.


Subject(s)
Cannabinoids/therapeutic use , Patient Safety , Cannabinoid Receptor Agonists/pharmacology , Cannabinoids/adverse effects , Cannabinoids/chemical synthesis , Cannabinoids/isolation & purification , Cannabis/chemistry , Drug Development , Humans , Substance-Related Disorders/prevention & control
7.
Sleep Med Rev ; 53: 101339, 2020 10.
Article in English | MEDLINE | ID: mdl-32603954

ABSTRACT

Cannabinoids, including the two main phytocannabinoids Δ9-tetrahydrocannabinol (THC) and cannabidiol (CBD), are being increasingly utilised as pharmacological interventions for sleep disorders. THC and CBD are known to interact with the endocannabinoid and other neurochemical systems to influence anxiety, mood, autonomic function, and circadian sleep/wake cycle. However, their therapeutic efficacy and safety as treatments for sleep disorders are unclear. The current systematic review assessed the available evidence base using PubMed, Scopus, Web of Science, Embase, CINAHL and PsycInfo databases. A total of 14 preclinical studies and 12 clinical studies met inclusion criteria. Results indicated that there is insufficient evidence to support routine clinical use of cannabinoid therapies for the treatment of any sleep disorder given the lack of published research and the moderate-to-high risk of bias identified within the majority of preclinical and clinical studies completed to-date. Promising preliminary evidence provides the rationale for future randomised controlled trials of cannabinoid therapies in individuals with sleep apnea, insomnia, post-traumatic stress disorder-related nightmares, restless legs syndrome, rapid eye movement sleep behaviour disorder, and narcolepsy. There is a clear need for further investigations on the safety and efficacy of cannabinoid therapies for treating sleep disorders using larger, rigorously controlled, longer-term trials.


Subject(s)
Cannabinoids/therapeutic use , Sleep Wake Disorders/drug therapy , Animals , Humans , Rats , Sleep/drug effects , Sleep Initiation and Maintenance Disorders/drug therapy
8.
Drug Alcohol Depend ; 194: 500-517, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30530238

ABSTRACT

Cannabis use disorder (CUD) is prevalent and demand for treatment is increasing, yet few individuals engage in formal treatment and the efficacy of established interventions for CUD is modest. Existing clinical trials evaluating psychosocial and pharmacological treatments for CUD have incorporated a wide variety of measures for assessing cannabis use outcomes, including abstinence, self-reported frequency and quantity used, withdrawal, use/dependence severity, and other psychosocial outcomes. The heterogeneity of measures and outcomes has limited quantitative analyses of the comparative effectiveness of existing interventions. The purpose of this systematic review is to: 1) identify and characterize approaches for measuring cannabis use in existing CUD intervention trials, including abstinence, frequency and quantity of use, and 2) summarize measures used to assess treatment efficacy in other outcome domains (e.g., cannabis use severity, psychosocial functioning, cannabis withdrawal), and provide a platform for future research to evaluate which outcome measures are most likely to reflect treatment efficacy and clinically significant improvement in other outcome domains.


Subject(s)
Cannabis/adverse effects , Marijuana Abuse/therapy , Substance Withdrawal Syndrome/diagnosis , Humans , Marijuana Abuse/drug therapy , Marijuana Abuse/psychology , Self Report , Treatment Outcome
9.
Int Rev Psychiatry ; 30(3): 277-284, 2018 06.
Article in English | MEDLINE | ID: mdl-30179534

ABSTRACT

Accumulating evidence suggests that the endocannabinoid system is a promising target for the treatment of a variety of health conditions. Two paths of cannabinoid drug development have emerged. One approach is focused on developing medications that are directly derived from the cannabis plant. The other utilizes a single molecule approach whereby individual phytocannabinoids or novel cannabinoids with therapeutic potential are identified and synthesized for pharmaceutical development. This commentary discusses the unique challenges and merits of botanical vs single molecule cannabinoid drug development strategies, highlights how both can be impacted by legalization of cannabis via legislative processes, and also addresses regulatory and public health considerations that are important to consider as cannabinoid medicine advances as a discipline.


Subject(s)
Cannabinoids , Cannabis , Drug Development , Legislation, Drug , Plant Extracts , Humans
11.
Exp Clin Psychopharmacol ; 25(4): 295-302, 2017 08.
Article in English | MEDLINE | ID: mdl-28782982

ABSTRACT

The objective of the study was to describe self-report and objectively measured sleep characteristics of adult treatment-seeking cannabis users. Study participants (n = 87) were adults who were screened for a 12-week outpatient cannabis treatment research program in Baltimore, MD. Participants completed objective and self-report measures of sleep quality. Data were analyzed for the sample overall and after stratifying by sex (54 men, 33 women). Participants were primarily urban, socioeconomically disadvantaged African Americans. Participants were frequent, heavy cannabis users; among a subset of participants assessed, 76.7% used cannabis on the day/night of the assessment. Participants had low rates of other substance abuse and of psychiatric comorbidities. Polysomnography indicated 19.5% of participants received the recommended 7 to 9 hr of sleep, with women averaging more sleep than men. One third (31.0%) had sleep latencies >30 min, one half spent >30 min awake after sleep onset, and more than one half of the sample (55.2%) had sleep efficiency scores of <85%. Most participants met criteria for subthreshold (36.8%) or clinical insomnia (25.3%) on the Insomnia Severity Index, 77.0% had scores of >5 on the Pittsburgh Sleep Quality Index. Most had average scores on the Dysfunctional Beliefs and Attitudes About Sleep (DBAS) questionnaire (M = 51.1, SD = 18.8) that were higher than average among clinical insomnia patients. Women had higher DBAS scores than men. Most participants exhibited characteristics of disordered sleep, and sex differences were observed on polysomnography and self-report measures. Findings extend prior research concerning the association between cannabis use and disordered sleep. Data presented in this article come from Clinical Trial NCT01685073. (PsycINFO Database Record


Subject(s)
Marijuana Abuse/rehabilitation , Sleep Initiation and Maintenance Disorders/epidemiology , Sleep/physiology , Adult , Ambulatory Care , Baltimore , Female , Humans , Male , Outpatients , Polysomnography , Self Report , Sex Factors , Surveys and Questionnaires , Young Adult
12.
Drug Alcohol Depend ; 177: 249-257, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28623823

ABSTRACT

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


Subject(s)
Acetylcysteine/therapeutic use , Marijuana Abuse/diagnosis , Marijuana Abuse/drug therapy , Adolescent , Adult , Cannabis , Double-Blind Method , Female , Free Radical Scavengers/therapeutic use , Humans , Male , Marijuana Abuse/psychology , Marijuana Smoking/drug therapy , Marijuana Smoking/psychology , Medication Adherence/psychology , Sulpiride , Treatment Outcome , Young Adult
13.
Addict Behav ; 72: 14-20, 2017 09.
Article in English | MEDLINE | ID: mdl-28340421

ABSTRACT

INTRODUCTION: Medical cannabis is increasingly being used for a variety of health conditions as more states implement legislation permitting medical use of cannabis. Little is known about medical cannabis use patterns and motives among adults across the lifespan. METHODS: The present study examined data collected at a medical cannabis dispensary in San Francisco, California. Participants included 217 medical cannabis patients who were grouped into age-defined cohorts (younger: 18-30, middle-aged: 31-50, and older: 51-72). The age groups were compared on several measures of cannabis use, motives and medical conditions using one-way ANOVAs, chi-square tests and linear regression analyses. RESULTS: All three age groups had similar frequency of cannabis use over the past month; however, the quantity of cannabis used and rates of problematic cannabis use were higher among younger users relative to middle-aged and older adults. The association between age and problematic cannabis use was moderated by age of regular use initiation such that earlier age of regular cannabis use onset was associated with more problematic use in the younger users, but not among older users. Middle-aged adults were more likely to report using medical cannabis for insomnia, while older adults were more likely to use medical cannabis for chronic medical problems such as cancer, glaucoma and HIV/AIDS. Younger participants reported cannabis use when bored at a greater rate than middle-aged and older adults. CONCLUSIONS: Findings suggest that there is an age-related risk for problematic cannabis use among medical cannabis users, such that younger users should be monitored for cannabis use patterns that may lead to deleterious consequences.


Subject(s)
Medical Marijuana/therapeutic use , Motivation , Adaptation, Psychological , Adolescent , Adult , Age Factors , Age of Onset , Aged , Ambulatory Care/statistics & numerical data , Analysis of Variance , Female , Humans , Male , Marijuana Use/epidemiology , Marijuana Use/psychology , Mental Disorders/drug therapy , Middle Aged , Pain/prevention & control , San Francisco/epidemiology , Surveys and Questionnaires , Young Adult
14.
Drug Alcohol Depend ; 167: 199-206, 2016 Oct 01.
Article in English | MEDLINE | ID: mdl-27590742

ABSTRACT

BACKGROUND: Rates of young adult cannabis use are rising, perceived harm is at its historical nadir, and most users do not want to quit. Most studies evaluating effects of cannabis use in young adults are cross-sectional, limiting causal inference. A method to reliably induce abstinence periods in cannabis users would allow assessment of the effects of abstinence and resumption of use on a variety of outcomes in a within-subjects, repeated measures design. METHODS: We examined the efficacy and feasibility of a voucher-based contingency management procedure for incentivizing one month of continuous cannabis abstinence among young adults who reported at least weekly cannabis use, volunteered to participate in a laboratory study, and did not express a desire to discontinue cannabis use long-term. Continuous cannabis abstinence was reinforced with an escalating incentive schedule, and self-report of abstinence was confirmed by frequent quantitative assays of urine cannabis metabolite (THCCOOH) concentration. New cannabis use during the abstinence period was determined using an established algorithm of change in creatinine-adjusted cannabis metabolite concentrations between study visits. RESULTS: Thirty-eight young adults, aged 18-25 years, enrolled and 34 (89.5%) attained biochemically confirmed 30-day abstinence. Among those who attained abstinence, 93.9% resumed regular use within two-weeks of incentive discontinuation. CONCLUSION: Findings support the feasibility and efficacy of contingency management to elicit short-term, continuous cannabis abstinence among young adult, non-treatment seeking, regular cannabis users. Further work should test the effectiveness of this contingency management procedure for cannabis abstinence in periods longer than one month, which may be required to evaluate some effects of abstinence.


Subject(s)
Behavior Therapy , Marijuana Abuse/therapy , Marijuana Smoking/therapy , Motivation , Adolescent , Adult , Cross-Sectional Studies , Feasibility Studies , Female , Humans , Male , Marijuana Abuse/psychology , Marijuana Smoking/psychology , Treatment Outcome , Young Adult
15.
Exp Clin Psychopharmacol ; 23(6): 415-21, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26461168

ABSTRACT

Over 300,000 individuals enter treatment for cannabis-use disorders (CUDs) in the United States annually. Cannabis withdrawal is associated with poor CUD-treatment outcomes, but no prior studies have examined sex differences in withdrawal among treatment-seeking cannabis users. Treatment-seeking cannabis users (45 women and 91 men) completed a Marijuana Withdrawal Checklist (Budney, Novy, & Hughes, 1999, Budney, Moore, Vandrey, & Hughes, 2003) at treatment intake to retrospectively characterize withdrawal symptoms experienced during their most recent quit attempt. Scores from the 14-item Composite Withdrawal Discomfort Scale (WDS), a subset of the Marijuana Withdrawal Checklist that corresponds to valid cannabis withdrawal symptoms described in the Diagnostic and Statistical Manual of Mental Disorders (5th ed.; APA, 2013) were calculated. Demographic and substance-use characteristics, overall WDS scores, and scores on individual WDS symptoms were compared between women and men. Women had higher overall WDS scores than men, and women had higher scores than men on 6 individual symptoms in 2 domains, mood symptoms (i.e., irritability, restlessness, increased anger, violent outbursts), and gastrointestinal symptoms (i.e., nausea, stomach pain). Follow-up analyses isolating the incidence and severity of WDS symptoms demonstrated that women generally reported a higher number of individual withdrawal symptoms than men, and that they reported experiencing some symptoms as more severe. This is the first report to demonstrate that women seeking treatment for CUDs may experience more withdrawal then men during quit attempts. Prospective studies of sex differences in cannabis withdrawal are warranted.


Subject(s)
Cannabis/adverse effects , Irritable Mood/drug effects , Marijuana Abuse/therapy , Sex Characteristics , Substance Withdrawal Syndrome/diagnosis , Adult , Female , Humans , Incidence , Male , Marijuana Smoking , Middle Aged , Retrospective Studies , Severity of Illness Index , Substance Withdrawal Syndrome/epidemiology , Young Adult
16.
JMIR Res Protoc ; 4(3): e87, 2015 Jul 17.
Article in English | MEDLINE | ID: mdl-26187404

ABSTRACT

BACKGROUND: Cannabis is the most frequently used illicit substance in the United States resulting in high rates of cannabis use disorders. Current treatments for cannabis use are often met with high rates of lapse/relapse, tied to (1) behavioral health factors that impact cannabis use such as poor sleep, and (2) access, stigma, supply, and cost of receiving a substance use intervention. OBJECTIVE: This pilot study examined the feasibility, usability, and changes in cannabis use and sleep difficulties following mobile phone-delivered Cognitive Behavioral Therapy for Insomnia (CBT-I) in the context of a cannabis cessation attempt. METHODS: Four male veterans with DSM-5 cannabis use disorder and sleep problems were randomized to receive a 2-week intervention: CBT-I Coach mobile app (n=2) or a placebo control (mood-tracking app) (n=2). Cannabis and sleep measures were assessed pre- and post-treatment. Participants also reported use and helpfulness of each app. Changes in sleep and cannabis use were evaluated for each participant individually. RESULTS: Both participants receiving CBT-I used the app daily over 2 weeks and found the app user-friendly, helpful, and would use it in the future. In addition, they reported decreased cannabis use and improved sleep efficiency; one also reported increased sleep quality. In contrast, one participant in the control group dropped out of the study, and the other used the app minimally and reported increased sleep quality but also increased cannabis use. The mood app was rated as not helpful, and there was low likelihood of future participation. CONCLUSIONS: This pilot study examined the feasibility and initial patient acceptance of mobile phone delivery of CBT-I for cannabis dependence. Positive ratings of the app and preliminary reports of reductions in cannabis use and improvements in sleep are both encouraging and support additional evaluation of this intervention.

17.
Addict Behav ; 45: 191-4, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25697725

ABSTRACT

INTRODUCTION: This secondary analysis of data from a large, multi-site effectiveness trial (NCT01104805) sought to determine whether effects of a web-based behavioral treatment (Therapeutic Education System [TES]) differed by participants' self-identified primary drug of abuse. METHODS: The all-comers sample of individuals entering outpatient psychosocial counseling treatment for substance abuse (N=497) cited cannabis (22.9%; n=114), stimulants (34.4%, n=171), opioids (21.7%, n=108), or alcohol (20.9%, n=104) as their primary substance of abuse. Participants were randomly assigned to receive treatment-as-usual (TAU) with or without TES substituted for approximately 2h of usual counseling. Multivariate analyses of abstinence outcomes examined interactions of treatment effects with primary substance. RESULTS: Adjusted odds ratios (AORs) demonstrated that primary stimulant users receiving TES were more likely to be abstinent in the final four weeks of treatment compared to stimulant users receiving TAU (AOR=3.59, 95% CI=1.25-10.27). Adjusted odds ratios for alcohol (AOR=3.15, 95% CI=0.85-11.65) and cannabis (AOR=2.64, 95% CI=0.73-9.52) also were of similar magnitude to stimulants but did not reach significance. Abstinence among primary opioid users was not improved by the TES intervention (AOR=0.35, 95% CI=0.09-1.47). CONCLUSIONS: This study supports the TES web-delivered treatment as a viable intervention for the majority of substance users entering outpatient counseling treatment, with demonstrated effectiveness among stimulant users and promising effects in alcohol and cannabis users but little or no effect in primary opioid users. Web-delivered treatments hold promise for expanding the availability of effective behavioral interventions for the majority of substance use disorders.


Subject(s)
Alcoholism/rehabilitation , Central Nervous System Stimulants , Cognitive Behavioral Therapy/methods , Internet , Marijuana Abuse/rehabilitation , Opioid-Related Disorders/rehabilitation , Therapy, Computer-Assisted/methods , Adult , Counseling/methods , Female , Humans , Male , Odds Ratio , Substance-Related Disorders/rehabilitation , Treatment Outcome
18.
Contemp Clin Trials ; 39(2): 211-23, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25179587

ABSTRACT

Despite recent advances in behavioral interventions for cannabis use disorders, effect sizes remain modest, and few individuals achieve long-term abstinence. One strategy to enhance outcomes is the addition of pharmacotherapy to complement behavioral treatment, but to date no efficacious medications targeting cannabis use disorders in adults through large, randomized controlled trials have been identified. The National Institute on Drug Abuse Clinical Trials Network (NIDA CTN) is currently conducting a study to test the efficacy of N-acetylcysteine (NAC) versus placebo (PBO), added to contingency management, for cannabis cessation in adults (ages 18-50). This study was designed to replicate positive findings from a study in cannabis-dependent adolescents that found greater odds of abstinence with NAC compared to PBO. This paper describes the design and implementation of an ongoing 12-week, intent-to-treat, double-blind, randomized, placebo-controlled study with one follow-up visit four weeks post-treatment. Approximately 300 treatment-seeking cannabis-dependent adults will be randomized to NAC or PBO across six study sites in the United States. The primary objective of this 12-week study is to evaluate the efficacy of twice-daily orally-administered NAC (1200 mg) versus matched PBO, added to contingency management, on cannabis abstinence. NAC is among the first medications to demonstrate increased odds of abstinence in a randomized controlled study among cannabis users in any age group. The current study will assess the cannabis cessation efficacy of NAC combined with a behavioral intervention in adults, providing a novel and timely contribution to the evidence base for the treatment of cannabis use disorders.


Subject(s)
Acetylcysteine/therapeutic use , Marijuana Abuse/drug therapy , Research Design , Acetylcysteine/administration & dosage , Acetylcysteine/adverse effects , Adolescent , Adult , Double-Blind Method , Female , Genetic Testing , Humans , Male , Marijuana Abuse/epidemiology , Marijuana Abuse/genetics , Middle Aged , National Institute on Drug Abuse (U.S.) , Smoking/epidemiology , United States , Young Adult
19.
Exp Clin Psychopharmacol ; 22(6): 517-23, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25150537

ABSTRACT

Cannabis is the most commonly used illicit substance worldwide, and cannabis use disorders (CUDs) are correspondingly high. Increased demand for treatment and relatively low rates of positive clinical outcomes has led to a large scientific investment in the development of interventions for the treatment of CUD. Much of this research is conducted with cannabis users who are not seeking treatment at the time of study participation, and it is unknown whether these individuals are representative of those who seek treatment. This study contrasted samples of cannabis users participating in screening interviews for treatment and nontreatment research studies. Several differences between groups emerged: Treatment-seekers were more likely to be female (43% vs. 29%), older (33.4 vs. 29.7 years), and have longer cannabis use histories compared with non-treatment-seekers (p = .007). Treatment-seekers were more likely to report experiencing guilt after using cannabis and to feel that cannabis use has been a problem for them. Additionally, treatment-seekers reported a greater mean number of reasons for making a quit attempt, experiencing a greater number of withdrawal symptoms, and employing more coping strategies during prior quit attempts. Despite the aforementioned differences, the 2 groups were similar on several key characteristics, particularly with regards to current levels of cannabis use and related problems.


Subject(s)
Marijuana Abuse/therapy , Marijuana Smoking/adverse effects , Patient Acceptance of Health Care , Academic Medical Centers , Adolescent , Adult , Baltimore , Cohort Studies , Cross-Sectional Studies , Diagnostic and Statistical Manual of Mental Disorders , Female , Guilt , Humans , Male , Marijuana Abuse/physiopathology , Marijuana Abuse/psychology , Marijuana Smoking/psychology , Middle Aged , Outpatient Clinics, Hospital , Retrospective Studies , Self Report , Severity of Illness Index , Young Adult
20.
Drug Alcohol Depend ; 136: 162-5, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24412475

ABSTRACT

BACKGROUND: The use of cannabis for medical purposes is proliferating in the U.S., and PTSD is an explicitly approved condition for accessing medical cannabis in 5 states. Prior research suggests that people with PTSD often use cannabis to help cope with their condition, and that doing so results in more frequent and problematic cannabis use patterns. Specific coping motivations, such as sleep improvement, among medical cannabis users, have not been examined. METHODS: The present study evaluated specific coping use motivations, frequency of cannabis and alcohol use, and mental health among a convenience sample of patients (N=170) at a medical cannabis dispensary in California. RESULTS: Those with high PTSD scores were more likely to use cannabis to improve sleep, and for coping reasons more generally, compared with those with low PTSD scores. Cannabis use frequency was greater among those with high PTSD scores who used for sleep promoting purposes compared with those with low PTSD scores or those who did not use for sleep promoting purposes. CONCLUSIONS: Consistent with prior research, this study found increased rates of coping-oriented use of cannabis and greater frequency of cannabis use among medical users with high PTSD scores compared with low PTSD scores. In addition, sleep improvement appears to be a primary motivator for coping-oriented use. Additional research is needed to examine the health consequences of this pattern of cannabis use and whether alternative sleep promoting interventions (e.g. CBT-I) could reduce the reliance on cannabis for adequate sleep among those with PTSD.


Subject(s)
Medical Marijuana/therapeutic use , Sleep Wake Disorders/drug therapy , Sleep Wake Disorders/etiology , Stress Disorders, Post-Traumatic/complications , Stress Disorders, Post-Traumatic/drug therapy , Adaptation, Psychological , Adult , Alcohol Drinking/psychology , Data Interpretation, Statistical , Depression/psychology , Female , Humans , Male , Middle Aged , Motivation , Neuropsychological Tests , Sleep/drug effects , Sleep Wake Disorders/psychology , Stress Disorders, Post-Traumatic/psychology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL