Next Article in Journal
Trial Sequential Analysis and Updated Meta-Analysis of Fluvoxamine on Clinical Deterioration in Adult Patients with Symptomatic COVID-19 Infection
Next Article in Special Issue
A Survey of Cannabis Use among Patients with Inflammatory Bowel Disease (IBD)
Previous Article in Journal
Energy Balance, Hormonal Status, and Military Performance in Strenuous Winter Training
Previous Article in Special Issue
The Rise and Rise of Medicinal Cannabis, What Now? Medicinal Cannabis Prescribing in Australia 2017–2022
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Evidence from Human Studies for Utilising Cannabinoids for the Treatment of Substance-Use Disorders: A Scoping Review with a Systematic Approach

by
Kayvan Ali Gharbi
1,2,*,
Yvonne Ann Bonomo
2,3 and
Christine Mary Hallinan
1,4,*
1
Department of General Practice, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
2
Department of Medicine, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
3
St Vincent’s Health—Department of Addiction Medicine, Fitzroy, VIC 3065, Australia
4
Health & Biomedical Research Information Technology Unit (HaBIC R2), Department of General Practice, Melbourne Medical School, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
*
Authors to whom correspondence should be addressed.
Int. J. Environ. Res. Public Health 2023, 20(5), 4087; https://doi.org/10.3390/ijerph20054087
Submission received: 21 November 2022 / Revised: 2 February 2023 / Accepted: 10 February 2023 / Published: 24 February 2023
(This article belongs to the Special Issue Cannabis, Cannabis-Based Products, and Cannabinoids for Medicinal Use)

Abstract

:
Substance-use disorders are pervasive, comorbid with a plethora of disease and possess limited treatment options. Medicinal cannabinoids have been proposed as a novel potential treatment based on preclinical/animal trials. The objective of this study was to examine the efficacy and safety of potential therapeutics targeting the endocannabinoid system in the treatment of substance-use disorders. We performed a scoping review using a systematic approach of systematic reviews, narrative reviews, and randomised control trials that utilised cannabinoids as treatment for substance-use disorders. For this scoping review we used the PRISMA guidelines, a framework for systematic reviews and meta-analyses, to inform our methodology. We conducted a manual search of Medline, Embase, and Scopus databases in July 2022. Of the 253 results returned by the databases, 25 studies including reviews were identified as relevant, from which 29 randomised controlled trials were derived and analysed via a primary study decomposition. This review captured a small volume of highly heterogenous primary literature investing the therapeutic effect of cannabinoids for substance-use disorders. The most promising findings appeared to be for cannabis-use disorder. Cannabidiol appeared to be the cannabinoid showing the most promise for the treatment of multiple-substance-use disorders.

1. Introduction

1.1. Substance-Use Disorders

Substance-use disorders are prevalent across Australia, with 3.3% of individuals aged between 16 and 85 years possessing a substance-use disorder of at least 12 months, with alcohol the principal substance of abuse at 2.5% [1]. Substance-use disorders are comorbid with both psychiatric (mood, psychotic, trauma, anxiety) [2] and physical disease (HIV, STIs, CVD, chronic pain, opportunistic infection) [3]. Mental health and substance-use disorders comprise 13% of Australia’s total burden of disease, making them the fourth most significant disease group [4]. Treatment options for substance-use disorders remain relatively limited, though there is strong evidence for medications (particularly agonist therapies) such as methadone and buprenorphine for opioid-use disorder [5], nicotine-replacement therapy for smoking cessation [6], and ‘anti-craving’ medications for alcohol-use disorder [7].The effectiveness of psychosocial interventions for substance-use disorders is mixed across the literature, with patient reluctance being an important contributor to the low utilisation of treatments considered effective [8]. Given the prevalence of substance-use disorders and their limited treatment options, efficacious therapeutics are needed. Medicinal cannabinoids have been proposed as a potential treatment option [9].

1.2. The Endocannabinoid System

The endocannabinoid system is a complex system of receptors, their ligands (endocannabinoids), and regulatory enzymes. There are two types of cannabinoid receptor (CB1R, CB2R) and two primary endocannabinoids: anandamide (ANA), also known as arachidonoylethanolamide (AEA), and 2-arachidonoylglycerol (2-AG). Each endocannabinoid is degraded by the enzymes fatty-acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL), respectively [10].
Preclinical studies of substance-use disorder treatments have applied both organic and synthetic molecules within the endocannabinoid system. Those at the stage of human trials include CB1 receptor agonists (tetrahydrocannabinol, dronabinol, nabiximols), receptor modulators (cannabidiol), enzyme inhibitors (FAAH Inhibitors), CB1 receptor inverse agonists (rimonabant, taranabant), and CB1 receptor antagonists (surinabant). Tetrahydrocannabinol (THC) is a naturally occurring exogenous cannabinoid that is orally bioavailable and the chief psychoactive component of cannabis acting at the CB1 receptor. Dronabinol is a synthetic enantiomer of THC. Cannabidiol (CBD) is an alternative cannabinoid with minimal direct action at receptors; nonetheless, it possesses a wide array of effects including allosteric modification of both endocannabinoid receptors [11] and opioid receptors [12], whilst inhibiting the hydrolysis and reuptake of AEA (increasing the availability of this CB1 receptor agonist) [13]. Nabiximols are a whole-plant-extract combined formulation of THC and CBD. They are typically delivered at an even dosage ratio in the form of an oromucosal spray that engenders a more predictable pharmacokinetic profile than oral dronabinol [14]. FAAH (fatty-acid amide hydrolase) inhibitors augment CB1 activation via increasing concentrations of the endocannabinoid AEA through inhibition of the enzyme FAAH thereby inhibiting degradation of AEA. CB1 inverse agonists and antagonists inhibit the downstream effects of endocannabinoids upon dopaminergic release in the nucleus accumbens [15], a mechanism central to the development of substance dependence [16].
Given growing preclinical evidence for several differing mechanisms of action of medicinal cannabinoids in the neurobiological pathways of substance-use disorders, findings from human studies were of interest. Hence, the objective of this scoping review is to synthesise primary research investigating the effect of cannabinoids on substance-use disorders to answer the following question: What is the efficacy and safety of potential therapeutics targeting the endocannabinoid system in the treatment of substance-use disorders in humans?

2. Materials and Methods

The review questions were decomposed using the PICOS search algorithm [17] to guide the development of search terms for the systematic review of reviews. The PICOS acronym refers to: P–participants; I–intervention; C–control; O–outcome; and S–study design (Table 1).

2.1. Search Strategy

For this systematic scoping review, we used a framework for systematic reviews and meta-analyses, the PRISMA guidelines, to inform our search methodology [18] and applied the Arksey and O’Malley [19,20,21] approach to performing a scoping review. In doing so we searched the Medline (Ovid), Embase (Ovid), and Scopus databases (Figure A1). An identical keyword search was applied to all three databases, with the added option of mesh terms leveraged in the Medline search. Studies published in English from January 2000 to July 2022 were considered for inclusion, as treatments that involved the use of cannabinoids for the treatment of substance-use disorders were not investigated prior to this time. Titles and abstracts of search results were screened independently by two researchers to determine whether they met the inclusion criteria. Accepted papers were retrieved, and further papers were excluded based on failure to meet inclusion criteria following full-text readings. Any additional papers referenced by these results meeting inclusion criteria were added to our search results.
The full-text articles that were identified for inclusion following the screening process were then independently critiqued by pairs of reviewers. A standardised format was used to extract and summarise their data according to PICOS [22]. Qualitative data synthesis was applied to each category of substance-use disorder. Due to the heterogeneity of outcome measures, no aggregated data analysis was performed.

2.2. Study Inclusion

Of the 243 articles identified in the search, 71 duplicate articles were removed using Endnote, and 128 were excluded following the screening of title and abstracts. This provided 44 potentially relevant full-text articles for screening prior to inclusion. Of these, 1 article was not able to be retrieved, and 21 were excluded as they did not meet criteria for study type, language, intervention intention, or were a non-clinical investigation (Table A1). After analysis of references across these 43 papers, an additional 1 SR and 2 NRs yielded were deemed as meeting inclusion criteria and incorporated into the search results, yielding a final total of 25 studies.
The search strategy and results are presented in the PRISMA flow chart found in Figure 1.
Adapted from: Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et. al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ 2021;372:n71. doi: 10.1136/bmj.n71. For more information, visit: http://www.prisma-statement.org/ (accessed on 24 August 2022).

3. Results

Of the 25 studies included in this review, 5 were systematic reviews, 14 were narrative reviews, and 6 were randomised controlled trials. The primary papers that were included in each of the systematic and narrative reviews captured by this study were evaluated and considered for inclusion in what is termed a ‘snowballing’ approach for identification of primary studies from systematic literature studies [23]. This yielded 23 further RCTs in addition to the 6 RCTs captured by the original search, yielding a total of 29 unique RCTs (Table 2).

3.1. Participants

The 29 unique RCTs included patients meeting criteria for a substance-use disorder (DSM-5), substance abuse, or dependence (DSM-IV) pertaining to the following substances: cannabis (13 RCTs), opioids (4 RCTs), cocaine (2 RCTs), nicotine (8 RCTs), and alcohol (2 RCTs).

3.2. Interventions

The 29 unique RCTs tested THC (3 RCTs), dronabinol (6 RCTs), nabiximols (5 RCTs), cannabidiol (7 RCTs), FAAH inhibitors (1 RCTs), rimonabant (5 RCT), surinabant (1 RCT), and taranabant (1 RCT). In 16 studies (55%) the endocannabinoid-based therapy was given in conjunction with a psychosocial intervention such as standard counselling and/or psychotherapy.
Table 3, Table 4, Table 5, Table 6 and Table 7 provide a summary of each paper that includes author names, title, date of publication, study type and study details including intervention type and duration, and outcome.
In this review, we categorised 29 research articles into five categories based on the type of substance-use disorder that was treated with medicinal cannabinoids. Due to the heterogeneity of outcome measures, no aggregated data analysis was performed.

3.3. Cannabis-Use Disorder

3.3.1. Tetrahydrocannabinol (THC)

There were three small trials (n = 7,8,8) examining THC for cannabis-use disorder. These were short duration (<7 days), within-subject studies trialling THC for attenuation of cannabis withdrawal severity and cravings. Daily 50 mg doses of THC significantly decreased a number of withdrawal symptoms including anxiety, mood, chills, sleep disturbance, and anorexia [51]. A study comparing 30 mg/90 mg THC to placebo yielded similar results with a dose-dependent relationship, such that 90 mg reduced symptom ratings to that as if regular smoking practice was maintained [52]. A trial combining THC with lofexidine (an alpha-2 receptor agonist aiming to attenuate known noradrenergic hyperactivity in cannabinoid withdrawal) versus THC monotherapy reported similar effect in attenuating withdrawal symptoms, as well as significant reduction in relapse with the combination treatment but not with THC monotherapy [53]. All studies noted excellent tolerability with few side effects including cognitive effects.

3.3.2. Dronabinol

Larger trials have examined dronabinol, a synthetic enantiomer of THC, for cannabis-use disorder. Two double-blinded RCTs in large populations (n = 156,122) of longer duration (9, 10 weeks) were conducted [54,56]. Both studies combined oral dronabinol (with additional lofexidine in the 2016 study) with Motivational Enhancement Therapy (MET) and Cognitive Behavioural Therapy (CBT) examining abstinence, cannabis use, and withdrawal severity. Although significant attenuation of withdrawal symptoms was observed, no significant difference in abstinence or cannabis use at two weeks was found versus placebo (although both groups showed reduction in cannabis use over the nine-week trial) [54]. No significant difference was found for combined dronabinol and lofexidine for withdrawal severity or abstinence [56].

3.3.3. Cannabidiol

A single-phase 2a RCT has examined the effect of oral CBD for four weeks for cannabis-use disorder [57]. Higher doses (400, 800 mg compared with 200 mg daily) were shown to significantly reduce cannabis use as assessed by urinary THC-COOH: creatinine concentrations and reported days of cannabis use per week (−0.48 and −0.27 days, respectively).

3.3.4. Nabiximols

Nabiximols were assessed in three small (n = 9,40,51) and one larger trial (n = 128). One study (n = 51) trialling 86.4 mg THC:80 mg CBD as a maximum daily dose for six days, showed withdrawal symptoms (measured by CWS (Cannabis Withdrawal Scale = 19 questions, each on a scale of 0–10)) significantly improved (mean 66% decrease from baseline levels) compared with placebo (mean 52% increase) [48]. However, no significant difference was found in cannabis use at follow-up 28 days later. The study with a fixed dose of 108 mg THC/100 mg CBD (n = 9) across eight weeks also reported attenuated withdrawal reducing the average CWS score to 10 from a baseline of 18 (with smoking as usual scoring 9). Effects on craving were not statistically significant [49]. The study of 40 patients given nabiximols (113.4 mg THC:105 mg CBD) also included MET/CBT for 12 weeks [50] but did not replicate findings described earlier. No significant differences in withdrawal scores or abstinence rates were found, but significant reduction in cannabis use across 12 weeks (−70.5% in nabiximols vs. −42.6% placebo) and cravings was found. The larger trial (n = 128) of 86.4 mg THC/80 mg CBD for 12 weeks also yielded non-significant findings for withdrawal symptoms, yet found significant reduction in craving and cannabis use across the trial (41.7% vs. 63.1% placebo days of usage across 12 weeks) [42]. A study following the same subjects three months after cessation of treatment found significant reduction in average number of days of cannabis use (−6.7 days in the previous 28 days) in addition to an increased proportion of subjects meeting criteria for abstinence (23% vs. 9% placebo) [43].

3.3.5. Fatty-Acid Amide Hydrolase Inhibitor—PF-04457845

One single-phase 2a RCT (n = 46) treatment with 4 mg PF-04457845 oral capsules for four weeks reported it to be safe and well tolerated, with cannabis withdrawal severity and cannabis usage (both self-reported and urine verified) both significantly reduced (61.2% reduction in urinary THC-COOH concentration) [58].

3.4. Opioid-Use Disorder

3.4.1. Dronabinol

Dronabinol (30 mg daily) for patients undergoing eight-day inpatient withdrawal and induction onto naltrexone continuing for five weeks post discharge, showed significant attenuation of the acute inpatient phase of withdrawal before naltrexone initiation (average SOWS (Subjective Opiate Withdrawal Scale = 16 questions, each on a scale of 0–4; mild withdrawal scores 1–10, moderate withdrawal scores 11–20, severe withdrawal scores 21–30) reduction of 11.34 vs. placebo) [44]. However, dronabinol did not improve symptoms of protracted low-grade withdrawal (insomnia, appetite, and low energy) that typically occur over the subsequent few weeks of outpatient treatment.
In a within-patient trial (n = 12) where a single dose of dronabinol (ranging from 5 to 40 mg, randomly assigned) was administered, subjects were assessed for physiological tolerability in one analysis [59] and withdrawal severity and psychomotor/cognitive effects in another [60]. Neither study found a difference compared to placebo for doses less than 20 mg. Larger doses did induce significant (albeit clinically moderate) withdrawal suppression; however, they also induced dose-dependent sustained symptomatic tachycardia and significant cognitive effects, specifically time estimation and continuous performance tasks.

3.4.2. Cannabidiol (CBD)

An RCT (n = 42) assessed CBD for its potential impacts on craving, anxiety, cognition, affect, and physiological markers in abstinent opioid dependent individuals for a week following a three-day treatment with daily 400 mg or 800 mg CBD [61]. Significant reductions in craving (VAS-C (Visual Analog Scale for Craving = line 10 cm in length with ‘no craving’ and ‘severe craving’ at the extremes) mean difference 0.44 and 0.23, respectively), anxiety (VAS-A (Visual Analog Scale for Anxiety = line 10 cm in length with ‘not anxious’ and ‘very anxious’ at the extremes) mean difference 0.48 0.24, respectively) and physiological markers (heart rate, cortisol) were found, with no significant effect upon cognition or affect.

3.5. Cocaine-Use Disorder

Cannabidiol

A double-blinded study (n = 31) trialling CBD (300 mg daily) for crack-cocaine (crack- cocaine is a free-base form of cocaine that can be smoked, formulated by cooking cocaine powder with baking soda then breaking it into small pieces called ‘rocks’) dependence for 10 days reduced cue-induced craving severity but was not statistically significant [46]. Nor were there significant findings for anxiety, depression, or sleep disturbance. An RCT (n = 50) treating outpatients for 12 weeks with CBD (800 mg daily) also found no significant reduction in cue-induced craving severity or in time to relapse [47].

3.6. Nicotine-Use Disorder

3.6.1. Rimonabant

Studies with Rimonabant and Tobacco Use (STRATUS) conducted multiple RCTs evaluating rimonabant for smoking cessation. STRATUS-EU and STRATUS-US were conducted with identical protocols and similar sample sizes (n = 784 and 783, respectively). Patients were treated with rimonabant (5 mg or 20 mg) daily for 10 weeks and followed up at 48 weeks for abstinence (both end of trial and prolonged), weight gain, and adverse events. STRATUS-META used only a 20 mg dose with no long-term follow-up after the 10-week intervention. A pooled analysis of these three trials showed 20 mg rimonabant significantly increased both end of treatment (OR = 1.6, 95% CI: 1.26, 2.12) and long-term (48 weeks) abstinence (OR = 1.50, 95% CI: 1.03, 2.17) [65]. Whilst a significantly greater likelihood of side effects including nausea (19.9% vs. 5.9% placebo) and anxiety (14.7% vs. 2.4% placebo) were found, no evidence of depressive symptoms was established. The STRATUS-WW (Worldwide) trial used a similar approach, demonstrating daily doses of 20 mg elicited significant improvement (RR for the 20 mg maintenance group was 1.29 (95% CI 1.06 to 1.57)) in the primary outcome of prevention of relapse to smoking.

3.6.2. Taranabant

Another CB1 inverse agonist, taranabant, was trialled for eight weeks (2 vs. 4 vs. 8 mg daily) for dependent cigarette smokers and failed to demonstrate effect on end-of-trial abstinence [67]. This trial recorded significant neuropsychiatric (depression, irritability) and gastrointestinal (nausea, vomiting, diarrhoea) side effects.

3.6.3. Surinabant

An RCT testing surinabant, a CB1 receptor antagonist and alternative to CB1 inverse agonists, used a similar protocol to the previously discussed taranabant trial and failed to have significant effect on abstinence over placebo [66]. Unlike the inverse agonists, Surinabant did not demonstrate significant neuropsychiatric side effects.

3.6.4. Cannabidiol

The effect of CBD upon self-reported cigarette use in dependent cigarette smokers was recorded in a one-week study where participants were given access to ad hoc usage of a CBD inhaler [68]. A significant reduction in cigarette use (~40%) both at the end of the treatment week and at the two-week follow-up was demonstrated in the absence of significant effect on craving or mood related side effects (sedation, depression, anxiety). An RCT trialling a single dose of 800 mg in dependent cigarette smokers observed that whilst it had non-significant effects on craving or withdrawal, it significantly reduced both attentional bias towards and subjective pleasantness of cigarette-related stimuli [69].

3.7. Alcohol-Use Disorder

Rimonabant
A proof-of-concept study (n = 258) tested the efficacy of rimonabant (2 × 10 mg daily) in relapse prevention for alcohol dependence for 12 weeks. Rimonabant was deemed tolerable but had no significant effect upon relapse to first drink, relapse to heavy drinking, or drinking frequency [45]. A second study applying the same dose for two weeks amongst non-treatment-seeking alcohol-dependent individuals reported similar findings, namely a non-significant effect upon self-reported (via telephone) alcohol consumption [62].

4. Discussion

4.1. Cannabis-Use Disorder

4.1.1. THC and Dronabinol

For cannabis-use disorder both THC and dronabinol (synthetic THC) appear to have benefit in attenuating withdrawal and cravings in a dose-dependent manner with good tolerability.
Neither cannabinoid, however, shows benefit in achieving abstinence from cannabis [29,30,31,32], which might have been expected given the success of opioid-substitution and nicotine-substitution treatments for their respective use disorders [5,6]. The dronabinol trials [54,56] cited cogent limitations accounting for their results. In the first, the authors postulated the short maintenance period (nine weeks) or the enrolment of non-treatment-seeking participants may have contributed to lack of abstinence. In the second, they suggested the failed result may have arisen because the intervention in this trial was introduced to induce abstinence, as opposed to prevent relapse in an already abstinent patient. Future studies of dronabinol for cannabis-use disorder could investigate an increased dose of dronabinol or, alternatively, other analogues of THC that have greater bioavailability and potency [56].

4.1.2. Cannabidiol (CBD)

Significant reduction in cannabis use with CBD [57] is promising, with 200 mg found ineffective and a marginal superiority of 400 mg over 800 mg indicating an inverted-U dose–response curve. Treatment duration of longer than four weeks, however, requires investigation. The mechanism of action of CBD therapy has been postulated to be via reduction of the impact of drug-related cues in attentional bias and craving [61,70]. Alternative mechanisms may include the effect of CBD on modulation of other comorbid psychiatric symptoms in cannabis-use disorder such as anxiety.

4.1.3. Nabiximols

Nabiximols show paradoxical findings. On one hand, Allsop et al., (2014) [48] and Trigo et al., (2016) [49] reported similar findings: a reduction in withdrawal severity but non-significant effects on long-term cannabis use. Conversely, Trigo et al., (2018) [50] and Lintzeris et al., (2019) [42] observed non-significant effects on withdrawal severity, yet a significant reduction in longer term cannabis use. Of note, both latter trials employed adjunctive psychotherapy in their intervention, and this may explain the longer-term improvements found. In the case of Trigo et al., (2018), the higher daily dosages (113.4 mg THC:105 mg CBD) may also have contributed to the beneficial effect on cannabis use, especially given the dose-dependent effect found. The high abstinence rate in the placebo group (>40%) indicates that the behavioural intervention was efficacious; therefore, future research should attempt to establish the role of nabiximols alone on abstinence. The question arises regarding why nabiximols had benefit in reducing cannabis use whilst fixed-dose dronabinol failed. Potential explanations include the flexible dose schedule, pharmacokinetic profile (higher bioavailability and more rapid onset of action), presence of additional whole-plant-extract components, and/or synergistic effects of THC with CBD [42].

4.1.4. Fatty-Acid Amide Hydrolase Inhibitor—PF-04457845

D’Souza et al., (2019) [58] demonstrated that FAAH inhibitors reduced both withdrawal severity and cannabis use in humans with limited psychoactive effects, suggesting that this approach could be more effective than simple direct CB1R agonism. This concurs with evidence showing that possessing a genetic variation of FAAH with reduced enzyme expression/activity confers a significantly lower likelihood of developing cannabis-use disorder than in wild-type carriers [71]. It also aligns with preclinical research showing a beneficial effect of FAAH inhibitors upon withdrawal severity [72]. Future research should compare the two approaches within the context of a larger sample in an outpatient setting.

4.2. Opioid-Use Disorder

4.2.1. Dronabinol

Dronabinol (30 mg) significantly reduced acute opioid withdrawal symptoms, but failed to have effect upon protracted low-grade withdrawal [44]. Potential explanations include that a threshold of symptom severity is necessary for dronabinol to be effective, a specificity of dronabinol for the symptoms of acute but not more chronic symptoms of opioid withdrawal, or participants developing tolerance to its effects after weeks of administration. Dose-dependent withdrawal suppression with dronabinol was found in other studies [59,60]; however, accompanying sustained tachycardia and anxiety/panic halted further investigation of dronabinol for opioid-use disorder.

4.2.2. Cannabidiol

CBD attenuates heroin-seeking behaviour in response to drug-associated cues in animals with a history of heroin self-administration [73]. This is clinically relevant as environmental cues are one of the strongest precipitators of craving, contributing to relapse. CBD’s effect upon cue-induced cravings, safe pharmacological profile, and lack of hedonic properties is promising as a potential treatment for opioid-use disorder [61]. Future studies with larger sample sizes, objective opioid measures, in addition to subjective metrics and longer duration are needed to establish the efficacy of CBD in relapse prevention for opioid-use disorder.

4.3. Cocaine-Use Disorder

Cannabidiol

Multiple preclinical trials suggested the onset and maintenance of cocaine addiction are reinforced by the dopaminergic neuro-transmission system [74] and withdrawal is associated with impaired dopamine function [75,76]. Preclinical studies of CB1 receptors in the ventral tegmental area demonstrate that CB1 agonists stimulate dopaminergic neurons causing an increase in extracellular dopamine levels in the nucleus accumbens [77,78] and therefore could perhaps disrupt the dopaminergic mechanisms of cocaine addiction.
Human trials have not, however, supported these hypotheses. Two trials explored CBD for cocaine-use disorder in both inpatients [46] and outpatients [47]. Neither context, higher doses (800 mg vs. 300 mg) nor longer treatment period (12 weeks vs. 10 days), showed benefit. No significant reduction in outcomes of cue-induced craving severity and time to relapse was found. Potential explanations for lack of effect include dose (CBD has complex dose–response curves), frequency of administration (e.g., twice-daily administration may be more effective given CBD’s 3 h peak plasma concentration), or that the mechanism of action of CBD is not as effective in the context of stimulants as for other substance-use disorders [47].

4.4. Nicotine-Use Disorder

4.4.1. Rimonabant

The STRATUS trials were promising for use of Rimonabant in smoking cessation. However, in June 2007 the FDA found significantly increased likelihood of suicidality for daily rimonabant (20 mg) taken for at least a year, leading Sanofi-Aventis to withdraw rimonabant [65]. Further trials evaluating other CB1 antagonists also ceased [79].

4.4.2. Taranabant

Taranabant, being related to rimonabant, demonstrated neuropsychiatric effects of a similar nature to rimonabant [67]. This illustrates a CB1 inverse agonist class effect and is therefore not suitable as a pharmacotherapy for substance-use disorders.

4.4.3. Surinabant

Surinabant, a CB1 receptor antagonist, showed no significant effect in nicotine-use disorder [66]. Preclinical trials demonstrated that while CB1 receptor activation is necessary for short-term induction of nicotine-incentive learning and reinforcement of drug-seeking behaviour in rats, other mechanisms become more significant for these behaviours after a few weeks [35]. Therefore, smokers who have been dependent for a matter of decades (as in this trial) perhaps lose sensitivity to endocannabinoid CB1 antagonism as an effective intervention.

4.4.4. Cannabidiol

The reduction in cigarette use in outpatients using an ad hoc CBD inhaler for one week was significant [68]. Reduction in both attentional bias and subjective pleasantness of cigarette-related stimuli was also demonstrated [69]. This supports the postulate that CBD exerts anti-addictive effects by minimising the effect of drug cues (incentive salience model of drug addiction), in addition to preventing the indexing of the reinforcing value of a drug through pleasure. No effect upon withdrawal or craving was demonstrated, and a significant limitation was that only a single dose was provided. Further studies with a longer treatment period and a range of repeated doses are needed.

4.5. Alcohol-Use Disorder

Rimonabant

Animal models show rimonabant decreases sensitivity to appetitive reinforcers [80] and reduces voluntary ethanol consumption [81,82]. Human trials have not, however, supported these findings [45,62]. A potential explanation for its limited effectiveness is that the higher doses used in animal studies enable near-complete receptor occupancy [62], yet higher doses of rimonabant in humans cannot be used because of the well documented adverse psychological effects.

4.6. Limitations

This review was limited by the vast heterogeneity across all primary studies included in this review. The populations varied in their specific diagnoses, treatment motivation, abstinence status, and degree of concurrent substance use. With respect to the intervention, there was significant variation in doses, adjunct interventions utilized, treatment setting, and duration. With respect to outcomes, differences included the specific outcomes assessed, how they were defined, follow-up duration, and the metric used to measure the outcomes. Most published reviews were inconclusive due to this heterogeneity, and quantitative data aggregation could not be undertaken.

4.7. Future Research Directions

The volume of research into endocannabinoids for substance-use disorders has failed to match the rapidly evolving public interest into their applications. Multiple states in the U.S. have decriminalized recreational use, and from a medical perspective the FDA has already approved multiple endocannabinoid formulations including Epidiolex (a CBD formulation) and Marinol (a dronabinol formulation). Given this rising tide and changing public opinion, the volume of primary research into the applications of endocannabinoids for substance-use disorders is set to increase. Such research will need to address the limitations in current research specified in 4.6 —this includes rigorous definitions of study populations, exploration of a range of dosages and adjunct interventions, and the application of standardised metrics for outcomes such as abstinence, withdrawal symptoms, and side effects.

4.8. Implications for Clinical Practice

The primary research thus far hints at the future potential for multiple endocannabinoid formulations across a range of substance-use disorders. Whilst there are many promising results, the paucity of human studies means that optimal dosages and treatment protocols are still yet to be researched and established. This is clinically critical not only from the perspective of optimising efficacy, but also in ensuring that side effects are either identified and characterised or alternatively deemed irreconcilable as in the case of rimonabant [79]. Therefore, given this absence in both volume and nuance of research it can be asserted that it is not yet appropriate to prescribe endocannabinoids as a clinician treating substance-use disorders. However, as future research moves to address these deficiencies and establish empirical validity, endocannabinoids will likely soon become a tool that is available to clinicians for both treating withdrawal and bolstering abstinence maintenance in the context of multiple-substance-use disorders. Therefore, it is worthwhile for such clinicians to vigilantly monitor the developing literature. Beyond efficacy, clinicians will next have to consider the socio-political dimension of their prescriptions including societal taboos and the interplay with the legal system (e.g., driving laws), and must be prepared to conduct evidence-based discussions with patients undoubtedly possessing a variety of preconceptions [83].

5. Conclusions

The body of evidence from human trials targeting the endocannabinoid system to treat substance-use disorders is not large and currently shows mixed results. The most promising research exists for cannabis-use disorder, indicating benefits of CB1R agonist therapy (dronabinol) for cannabis withdrawal. Potential application of nabiximols (whole-plant extract) and cannabidiol for reduction of cannabis use also exists. For opioids, direct CB1 agonist therapy at effective doses appears to induce intolerable side effects; however, CBD may have potential for reducing opioid cue-induced craving. Rimonabant was the only therapy that had been trialled in the context of alcohol-use disorder, but is not feasible because of significant psychotropic effects, hence its withdrawal from the market. The same applies for nicotine dependence. There is little promise thus far for the use of cannabinoids in cocaine-use disorder. Most of the examined studies, however, possessed small samples and multiple limitations; hence further studies of medicinal cannabinoids as a treatment option for substance-use disorders are needed moving into the future.

Author Contributions

Conceptualization, Y.A.B.; methodology, C.M.H.; software, K.A.G., Y.A.B. and C.M.H.; validation, K.A.G., Y.A.B. and C.M.H.; formal analysis, K.A.G. and Y.A.B.; investigation, K.A.G., Y.A.B. and C.M.H.; resources, Y.A.B. and C.M.H.; data curation, K.A.G., Y.A.B. and C.M.H.; writing—original draft preparation, K.A.G., Y.A.B. and C.M.H.; writing—review and editing, K.A.G., Y.A.B. and C.M.H.; visualization, K.A.G. and C.M.H.; supervision, Y.A.B. and C.M.H.; project administration, Y.A.B. and C.M.H.; funding acquisition, Y.A.B. All authors have read and agreed to the published version of the manuscript.

Funding

This review was supported by the Australian Centre for Cannabinoid Clinical and Research Excellence Funding: (ACRE), funded by the National Health and Medical Research Council (NHMRC) through the Centre of Research Excellence scheme (NHMRC CRE APP1135054).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All relevant data within the manuscript are available on request.

Conflicts of Interest

The authors declare no conflict of interest.

Appendix A

Figure A1. Search strategy.
Figure A1. Search strategy.
Ijerph 20 04087 g0a1
Table A1. Excluded studies.
Table A1. Excluded studies.
NO.AuthorReason for exclusion
1Bhardwaj et. al., 2018 [84]Ineligible study type—study protocol.
2Calpe-López et. al., 2019 [31]Nonclinical—pharmacological review of mechanisms.
3Calpe-López et. al., 2019 [31]Duplicate.
4Cohen et. al., 2020 [85]Not pertaining to SUD treatment, only PTSD.
5De Ternay et. al., 2019 [86]Animal studies—no human trials.
6George 2007 [87]Ineligible study type—book chapter.
7Janero 2012 [88]Ineligible study type—short survey.
8Janero & Makriyannis, 2007 [89]Nonclinical—pharmacological review of mechanisms.
9Khurana et. al., 2017 [90]Nonclinical—pharmacological review of mechanisms.
10Lake et. al., 2021 [91]Ineligible study type—cohort study.
11Lee et. al., 2017 [39]Nonclinical—pharmacological review of mechanisms.
12Luján & Valverde, 2020 [92]Nonclinical—pharmacological review of mechanisms.
13Mackie 2006 [93]Nonclinical—pharmacological review of mechanisms.
14Onaivi 2009 [94]Nonclinical—pharmacological review of mechanisms.
15Pietrzak et. al., 2011 [95]Non-English (Polish).
16Preedy 2017 [96]Ineligible study type—book chapter.
17Rodrigues et. al., 2020 [97]Animal studies—no human trials.
18Sholler et. al., 2020 [41]Nonclinical—pharmacological review of mechanisms.
19Śmiarowska et. al., 2022 [98]Nonclinical—pharmacological review of mechanisms.
20Weidenauer et. al., 2021 [99]Non-English (German).
21Yang et. al., 2012 [100]Animal studies—no human trials.

References

  1. Australian Bureau of Statistics. National Study of Mental Health and Wellbeing, 2020-21. Available online: https://www.abs.gov.au/statistics/health/mental-health/national-study-mental-health-and-wellbeing/latest-release (accessed on 21 September 2022).
  2. Kalin, N.H. Substance Use Disorders and Addiction: Mechanisms, Trends, and Treatment Implications. Am. J. Psychiatry 2020, 177, 1015–1018. [Google Scholar] [CrossRef]
  3. Commission on Narcotic Drugs: 65th Session. Comorbidities in Drug Use Disorders: No Wrong Door; United Nations Office on Drugs and Crime: Vienna, Austria, 2022. [Google Scholar]
  4. Australian Institute of Health and Welfare. Mental Health: Prevalence and Impact. 2022. Available online: https://www.aihw.gov.au/reports/mental-health-services/mental-health (accessed on 25 September 2022).
  5. National Institute on Drug Abuse. Effective Treatments for Opioid Addiction. 2022. Available online: https://nida.nih.gov/publications/effective-treatments-opioid-addiction (accessed on 16 September 2022).
  6. National Institute on Drug Abuse. Tobacco Addiction. 2022. Available online: https://nida.nih.gov/publications/principles-drug-addiction-treatment-research-based-guide-third-edition/evidence-based-approaches-to-drug-addiction-treatment/pharmacotherapies/tobacco-addiction (accessed on 16 September 2022).
  7. National Institute on Drug Abuse. Alcohol Addiction. 2022. Available online: https://nida.nih.gov/publications/principles-drug-addiction-treatment-research-based-guide-third-edition/evidence-based-approaches-to-drug-addiction-treatment/pharmacotherapies/alcohol (accessed on 20 September 2022).
  8. Hodgkin, D.; Connery, H.S. Effectiveness and Availability of Treatment for Substance Use Disorders. Oxf. Res. Encycl. Econ. Financ. 2018. [Google Scholar] [CrossRef]
  9. Sloan, M.E.; Gowin, J.L.; Ramchandani, V.A.; Hurd, Y.L.; Le Foll, B. The Endocannabinoid System as a Target for Addiction Treatment: Trials and Tribulations. Neuropharmacology 2017, 124, 73–83. [Google Scholar] [CrossRef]
  10. Sviženska, I.; Dubový, P.; Sulcova, A. Cannabinoid Receptors 1 and 2 (CB1 and CB2), Their Distribution, Ligands and Functional Involvement in Nervous System Structures—A Short Review. Pharmacol. Biochem. Behav. 2008, 90, 501–511. [Google Scholar] [CrossRef]
  11. Laprairie, R.B.; Bagher, A.M.; Kelly, M.E.M.; Denovan-Wright, E.M. Cannabidiol Is a Negative Allosteric Modulator of the Cannabinoid CB1 Receptor. Br. J. Pharmacol. 2015, 172, 4790–4805. [Google Scholar] [CrossRef] [Green Version]
  12. Kathmann, M.; Flau, K.; Redmer, A.; Tränkle, C.; Schlicker, E. Cannabidiol Is an Allosteric Modulator at Mu- and Delta-Opioid Receptors. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2006, 372, 354–361. [Google Scholar] [CrossRef]
  13. Bisogno, T.; Hanuš, L.; De Petrocellis, L.; Tchilibon, S.; Ponde, D.E.; Brandi, I.; Moriello, A.S.; Davis, J.B.; Mechoulam, R.; Di Marzo, V. Molecular Targets for Cannabidiol and Its Synthetic Analogues: Effect on Vanilloid VR1 Receptors and on the Cellular Uptake and Enzymatic Hydrolysis of Anandamide. Br. J. Pharmacol. 2001, 134, 845–852. [Google Scholar] [CrossRef]
  14. Karschner, E.L.; Darwin, W.D.; Goodwin, R.S.; Wright, S.; Huestis, M.A. Plasma Cannabinoid Pharmacokinetics Following Controlled Oral Δ9-Tetrahydrocannabinol and Oromucosal Cannabis Extract Administration. Clin. Chem. 2011, 57, 66–75. [Google Scholar] [CrossRef] [Green Version]
  15. Schlicker, E.; Kathmann, M. Modulation of Transmitter Release via Presynaptic Cannabinoid Receptors. Trends Pharmacol. Sci. 2001, 22, 565–572. [Google Scholar] [CrossRef]
  16. Volkow, N.D.; Fowler, J.S.; Wang, G.-J.; Swanson, J.M.; Telang, F. Dopamine in Drug Abuse and Addiction. Arch. Neurol. 2007, 64, 1575–1579. [Google Scholar] [CrossRef]
  17. Liberati, A.; Altman, D.G.; Tetzlaff, J.; Mulrow, C.; Gotzsche, P.C.; Ioannidis, J.P.A.; Clarke, M.; Devereaux, P.J.; Kleijnen, J.; Moher, D. The Prisma Statement for Reporting Systematic Reviews and Meta-Analyses of Studies That Evaluate Healthcare Interventions: Explanation and Elaboration. BMJ 2009, 339, b2700. [Google Scholar] [CrossRef] [Green Version]
  18. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The Prisma 2020 Statement: An Updated Guideline for Reporting Systematic Reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef]
  19. Arksey, H.; O’Malley, L. Scoping Studies: Towards a Methodological Framework. Int. J. Soc. Res. Methodol. 2005, 8, 19–32. [Google Scholar] [CrossRef] [Green Version]
  20. Hallinan, C.M.; Habibabadi, S.K.; Conway, M.; Bonomo, Y.A. Social Media Discourse and Internet Search Queries on Cannabis as a Medicine: A Systematic Scoping Review. PLoS ONE 2023, 18, e0269143. [Google Scholar] [CrossRef]
  21. Pawliuk, C.; Chau, B.; Rassekh, S.R.; McKellar, T.; Siden, H. Efficacy and Safety of Paediatric Medicinal Cannabis Use: A Scoping Review. Paediatr. Child Health 2020, 26, 228–233. [Google Scholar] [CrossRef]
  22. Amir-Behghadami, M.; Janati, A. Population, Intervention, Comparison, Outcomes and Study (PICOS) Design as a Framework to Formulate Eligibility Criteria in Systematic Reviews. Emerg. Med. J. 2020, 37, 387. [Google Scholar] [CrossRef]
  23. Wohlin, C.; Kalinowski, M.; Felizardo, K.R.; Mendes, E. Successful Combination of Database Search and Snowballing for Identification of Primary Studies in Systematic Literature Studies. Inf. Softw. Technol. 2022, 147, 106908. [Google Scholar] [CrossRef]
  24. Batalla, A.; Janssen, H.; Gangadin, S.S.; Bossong, M.G. The Potential of Cannabidiol as a Treatment for Psychosis and Addiction: Who Benefits Most? A Systematic Review. J. Clin. Med. 2019, 8, 1058. [Google Scholar] [CrossRef] [Green Version]
  25. Hoch, E.; Niemann, D.; von Keller, R.; Schneider, M.; Friemel, C.M.; Preuss, U.W.; Hasan, A.; Pogarell, O. Correction to: How effective and safe is medical cannabis as a treatment of mental disorders? A systematic review. Eur. Arch. Psychiatry Clin. Neurosci. 2019, 269, 995. [Google Scholar] [CrossRef] [Green Version]
  26. Paulus, V.; Billieux, J.; Benyamina, A.; Karila, L. Cannabidiol in the context of substance use disorder treatment: A systematic review. Addict. Behav. 2022, 132, 107360. [Google Scholar] [CrossRef]
  27. Pavel, A.; Paun, R.; Valentin, M.P. 316 The use of cannabidiol in treating psychiatric disorder: A systematic review. Eur. Neuropsychopharmacol. 2021, 44, S50. [Google Scholar] [CrossRef]
  28. Prud’Homme, M.; Cata, R.; Jutras-Aswad, D. Cannabidiol as an Intervention for Addictive Behaviors: A Systematic Review of the Evidence. Subst. Abus. Res. Treat. 2015, 9, 33–38. [Google Scholar] [CrossRef] [Green Version]
  29. Babalonis, S.; Walsh, S.L. Therapeutic potential of opioid/cannabinoid combinations in humans: Review of the evidence. Eur. Neuropsychopharmacol. 2020, 36, 206–216. [Google Scholar] [CrossRef]
  30. Beardsley, P.M.; Thomas, B.F.; McMahon, L.R. Cannabinoid CB1 receptor antagonists as potential pharmacotherapies for drug abuse disorders. Int. Rev. Psychiatry 2009, 21, 134–142. [Google Scholar] [CrossRef]
  31. Calpe-López, C.; García-Pardo, M.P.; Aguilar, M.A. Cannabidiol Treatment Might Promote Resilience to Cocaine and Methamphetamine Use Disorders: A Review of Possible Mechanisms. Molecules 2019, 24, 2583. [Google Scholar] [CrossRef] [Green Version]
  32. Chye, Y.; Christensen, E.; Solowij, N.; Yücel, M. The Endocannabinoid System and Cannabidiol’s Promise for the Treatment of Substance Use Disorder. Front. Psychiatry 2019, 10, 63. [Google Scholar] [CrossRef]
  33. Femenía, T.; Portillo, P.; Pérez-Ortiz, J.M.; Aracil-Fernández, A.; Rubio, G.; Manzanares, J. Opioid and Cannabinoid Systems as Therapeutic Targets for the Treatment of Alcohol Dependence: From Animal Models to Clinical Practice. Open Neuropsychopharmacol. J. 2009, 2, 53–63. [Google Scholar] [CrossRef] [Green Version]
  34. Fischer, B.; Kuganesan, S.; Gallassi, A.; Malcher-Lopes, R.; Brink, W.V.D.; Wood, E. Addressing the stimulant treatment gap: A call to investigate the therapeutic benefits potential of cannabinoids for crack-cocaine use. Int. J. Drug Policy 2015, 26, 1177–1182. [Google Scholar] [CrossRef]
  35. Le Foll, B.; Forget, B.; Aubin, H.-J.; Goldberg, S.R. Blocking cannabinoid CB1 receptors for the treatment of nicotine dependence: Iinsights from pre-clinical and clinical studies. Addict. Biol. 2008, 13, 239–252. [Google Scholar] [CrossRef]
  36. Galaj, E.; Xi, Z.-X. Potential of Cannabinoid Receptor Ligands as Treatment for Substance Use Disorders. CNS Drugs 2019, 33, 1001–1030. [Google Scholar] [CrossRef]
  37. Kleczkowska, P.; Smaga, I.; Filip, M.; Bujalska-Zadrozny, M. Cannabinoid Ligands and Alcohol Addiction: A Promising Therapeutic Tool or a Humbug? Neurotox. Res. 2015, 29, 173–196. [Google Scholar] [CrossRef] [Green Version]
  38. Kolongowski, B.; Tjiattas-Saleski, L. Cannabidiol: Background and Literature Review of Potential Treatments. Osteopat. Fam. Physician 2021, 16–23. [Google Scholar] [CrossRef]
  39. Lee, J.L.C.; Bertoglio, L.J.; Guimarães, F.S.; Stevenson, C.W. Cannabidiol regulation of emotion and emotional memory processing: Relevance for treating anxiety-related and substance abuse disorders. Br. J. Pharmacol. 2017, 174, 3242–3256. [Google Scholar] [CrossRef] [Green Version]
  40. Navarrete, F.; García-Gutiérrez, M.S.; Gasparyan, A.; Austrich-Olivares, A.; Manzanares, J. Role of Cannabidiol in the Therapeutic Intervention for Substance Use Disorders. Front. Pharmacol. 2021, 12, 626010. [Google Scholar] [CrossRef]
  41. Sholler, D.J.; Huestis, M.A.; Amendolara, B.; Vandrey, R.; Cooper, Z.D. Therapeutic potential and safety considerations for the clinical use of synthetic cannabinoids. Pharmacol. Biochem. Behav. 2020, 199, 173059. [Google Scholar] [CrossRef]
  42. Lintzeris, N.; Bhardwaj, A.; Mills, L.; Dunlop, A.; Copeland, J.; Mcgregor, I.; Bruno, R.; Gugusheff, J.; Phung, N.; Montebello, M.; et al. Nabiximols for the Treatment of Cannabis Dependence. JAMA Intern. Med. 2019, 179, 1242–1253. [Google Scholar] [CrossRef]
  43. Lintzeris, N.; Mills, L.; Dunlop, A.; Copeland, J.; Mcgregor, I.; Bruno, R.; Kirby, A.; Montebello, M.; Hall, M.; Jefferies, M.; et al. Cannabis Use in Patients 3 Months after Ceasing Nabiximols for the Treatment of Cannabis Dependence: Results from a Placebo-Controlled Randomised Trial. Drug Alcohol Depend. 2020, 215, 108220. [Google Scholar] [CrossRef]
  44. Bisaga, A.; Sullivan, M.A.; Glass, A.; Mishlen, K.; Pavlicova, M.; Haney, M.; Raby, W.N.; Levin, F.R.; Carpenter, K.M.; Mariani, J.J.; et al. The Effects of Dronabinol during Detoxification and the Initiation of Treatment with Extended Release Naltrexone. Drug Alcohol Depend. 2015, 154, 38–45. [Google Scholar] [CrossRef] [Green Version]
  45. Soyka, M.; Koller, G.; Schmidt, P.; Lesch, O.-M.; Leweke, M.; Fehr, C.; Gann, H.; Mann, K.F. Cannabinoid Receptor 1 Blocker Rimonabant (SR 141716) for Treatment of Alcohol Dependence. J. Clin. Psychopharmacol. 2008, 28, 317–324. [Google Scholar] [CrossRef]
  46. de Meneses-Gaya, C.; Crippa, J.A.; Hallak, J.E.; Miguel, A.Q.; Laranjeira, R.; Bressan, R.A.; Zuardi, A.W.; Lacerda, A.L. Cannabidiol for the Treatment of Crack-Cocaine Craving: An Exploratory Double-Blind Study. Braz. J. Psychiatry 2021, 43, 467–476. [Google Scholar] [CrossRef]
  47. Mongeau-Pérusse, V.; Brissette, S.; Bruneau, J.; Conrod, P.; Dubreucq, S.; Gazil, G.; Stip, E.; Jutras-Aswad, D. Cannabidiol as a Treatment for Craving and Relapse in Individuals with Cocaine Use Disorder: A Randomized Placebo-Controlled Trial. Addiction 2021, 116, 2431–2442. [Google Scholar] [CrossRef]
  48. Allsop, D.J.; Copeland, J.; Lintzeris, N.; Dunlop, A.J.; Montebello, M.; Sadler, C.; Rivas, G.R.; Holland, R.M.; Muhleisen, P.; Norberg, M.M.; et al. Nabiximols as an Agonist Replacement Therapy During Cannabis Withdrawal. JAMA Psychiatry 2014, 71, 281–291. [Google Scholar] [CrossRef] [Green Version]
  49. Trigo, J.M.; Lagzdins, D.; Rehm, J.; Selby, P.; Gamaleddin, I.; Fischer, B.; Barnes, A.J.; Huestis, M.A.; Le Foll, B. Effects of Fixed or Self-Titrated Dosages of Sativex on Cannabis Withdrawal and Cravings. Drug Alcohol Depend. 2016, 161, 298–306. [Google Scholar] [CrossRef] [Green Version]
  50. Trigo, J.M.; Soliman, A.; Quilty, L.C.; Fischer, B.; Rehm, J.; Selby, P.; Barnes, A.J.; Huestis, M.A.; George, T.P.; Streiner, D.L.; et al. Nabiximols Combined with Motivational Enhancement/Cognitive Behavioral Therapy for the Treatment of Cannabis Dependence: A Pilot Randomized Clinical Trial. PLoS ONE 2018, 13, e0190768. [Google Scholar] [CrossRef] [Green Version]
  51. Haney, M.; Hart, C.L.; Vosburg, S.K.; Nasser, J.; Bennett, A.; Zubaran, C.; Foltin, R.W. Marijuana Withdrawal in Humans: Effects of Oral THC or Divalproex. Neuropsychopharmacology 2003, 29, 158–170. [Google Scholar] [CrossRef] [Green Version]
  52. Budney, A.J.; Vandrey, R.G.; Hughes, J.R.; Moore, B.A.; Bahrenburg, B. Oral Delta-9-Tetrahydrocannabinol Suppresses Cannabis Withdrawal Symptoms. Drug Alcohol Depend. 2007, 86, 22–29. [Google Scholar] [CrossRef]
  53. Haney, M.; Hart, C.L.; Vosburg, S.K.; Comer, S.D.; Reed, S.C.; Foltin, R.W. Effects of THC and Lofexidine in a Human Laboratory Model of Marijuana Withdrawal and Relapse. Psychopharmacology 2007, 197, 157–168. [Google Scholar] [CrossRef] [Green Version]
  54. Levin, F.R.; Mariani, J.J.; Brooks, D.J.; Pavlicova, M.; Cheng, W.; Nunes, E. Dronabinol for the Treatment of Cannabis Dependence: A Randomized, Double-Blind, Placebo-Controlled Trial. Drug Alcohol Depend. 2011, 116, 142–150. [Google Scholar] [CrossRef] [Green Version]
  55. Vandrey, R.; Stitzer, M.L.; Mintzer, M.Z.; Huestis, M.A.; Murray, J.A.; Lee, D. The Dose Effects of Short-Term Dronabinol (Oral THC) Maintenance in Daily Cannabis Users. Drug Alcohol Depend. 2013, 128, 64–70. [Google Scholar] [CrossRef] [Green Version]
  56. Levin, F.R.; Mariani, J.J.; Pavlicova, M.; Brooks, D.; Glass, A.; Mahony, A.; Nunes, E.V.; Bisaga, A.; Dakwar, E.; Carpenter, K.M.; et al. Dronabinol and Lofexidine for Cannabis Use Disorder: A Randomized, Double-Blind, Placebo-Controlled Trial. Drug Alcohol Depend. 2016, 159, 53–60. [Google Scholar] [CrossRef] [Green Version]
  57. Freeman, T.P.; Hindocha, C.; Baio, G.; Shaban, N.D.C.; Thomas, E.M.; Astbury, D.; Freeman, A.M.; Lees, R.; Craft, S.; Morrison, P.D.; et al. Cannabidiol for the Treatment of Cannabis Use Disorder: A Phase 2a, Double-Blind, Placebo-Controlled, Randomised, Adaptive Bayesian Trial. Lancet Psychiatry 2020, 7, 865–874. [Google Scholar] [CrossRef]
  58. D’Souza, D.C.; Cortes-Briones, J.; Creatura, G.; Bluez, G.; Thurnauer, H.; Deaso, E.; Bielen, K.; Surti, T.; Radhakrishnan, R.; Gupta, A.; et al. Efficacy and Safety of a Fatty Acid Amide Hydrolase Inhibitor (PF-04457845) in the Treatment of Cannabis Withdrawal and Dependence in Men: A Double-Blind, Placebo-Controlled, Parallel Group, Phase 2a Single-Site Randomised Controlled Trial. Lancet Psychiatry 2019, 6, 35–45. [Google Scholar] [CrossRef]
  59. Jicha, C.J.; Lofwall, M.R.; Nuzzo, P.A.; Babalonis, S.; Elayi, S.C.; Walsh, S.L. Safety of Oral Dronabinol during Opioid Withdrawal in Humans. Drug Alcohol Depend. 2015, 157, 179–183. [Google Scholar] [CrossRef] [Green Version]
  60. Lofwall, M.R.; Babalonis, S.; Nuzzo, P.A.; Elayi, S.C.; Walsh, S.L. Opioid Withdrawal Suppression Efficacy of Oral Dronabinol in Opioid Dependent Humans. Drug Alcohol Depend. 2016, 164, 143–150. [Google Scholar] [CrossRef] [Green Version]
  61. Hurd, Y.L.; Spriggs, S.; Alishayev, J.; Winkel, G.; Gurgov, K.; Kudrich, C.; Oprescu, A.M.; Salsitz, E. Cannabidiol for the Reduction of Cue-Induced Craving and Anxiety in Drug-Abstinent Individuals With Heroin Use Disorder: A Double-Blind Randomized Placebo-Controlled Trial. Am. J. Psychiatry 2019, 176, 911–922. [Google Scholar] [CrossRef]
  62. George, D.T.; Herion, D.W.; Jones, C.L.; Phillips, M.J.; Hersh, J.; Hill, D.; Heilig, M.; Ramchandani, V.A.; Geyer, C.; Spero, D.E.; et al. Rimonabant (SR141716) Has No Effect on Alcohol Self-Administration or Endocrine Measures in Nontreatment-Seeking Heavy Alcohol Drinkers. Psychopharmacology 2009, 208, 37–44. [Google Scholar] [CrossRef] [Green Version]
  63. Rigotti, N.A.; Gonzales, D.; Dale, L.C.; Lawrence, D.; Chang, Y. A Randomized Controlled Trial of Adding the Nicotine Patch to Rimonabant for Smoking Cessation: Efficacy, Safety and Weight Gain. Addiction 2009, 104, 266–276. [Google Scholar] [CrossRef]
  64. Cahill, K.; Ussher, M.H. Cannabinoid Type 1 Receptor Antagonists for Smoking Cessation. Cochrane Database Syst. Rev. 2011, 2011, CD005353. [Google Scholar] [CrossRef]
  65. Robinson, J.D.; Cinciripini, P.M.; Karam-Hage, M.; Aubin, H.-J.; Dale, L.C.; Niaura, R.; Anthenelli, R.M. Pooled Analysis of Three Randomized, Double-Blind, Placebo Controlled Trials with Rimonabant for Smoking Cessation. Addict. Biol. 2017, 23, 291–303. [Google Scholar] [CrossRef]
  66. Tonstad, S.; Aubin, H.-J. Efficacy of a Dose Range of Surinabant, a Cannabinoid Receptor Blocker, for Smoking Cessation: A Randomized Controlled Clinical Trial. J. Psychopharmacol. 2012, 26, 1003–1009. [Google Scholar] [CrossRef]
  67. Morrison, M.F.; Ceesay, P.; Gantz, I.; Kaufman, K.D.; Lines, C.R. Randomized, controlled, double-blind trial of taranabant for smoking cessation. Psychopharmacology 2010, 209, 245–253. [Google Scholar] [CrossRef]
  68. Morgan, C.J.; Das, R.K.; Joye, A.; Curran, H.V.; Kamboj, S.K. Cannabidiol Reduces Cigarette Consumption in Tobacco Smokers: Preliminary Findings. Addict. Behav. 2013, 38, 2433–2436. [Google Scholar] [CrossRef]
  69. Hindocha, C.; Freeman, T.P.; Grabski, M.; Crudgington, H.; Davies, A.C.; Stroud, J.B.; Das, R.K.; Lawn, W.; Morgan, C.J.A.; Curran, H.V. The Effects of Cannabidiol on Impulsivity and Memory during Abstinence in Cigarette Dependent Smokers. Sci. Rep. 2018, 8, 1–7. [Google Scholar] [CrossRef] [Green Version]
  70. Hindocha, C.; Freeman, T.P.; Grabski, M.; Stroud, J.B.; Crudgington, H.; Davies, A.C.; Das, R.K.; Lawn, W.; Morgan, C.J.A.; Curran, H.V. Cannabidiol Reverses Attentional Bias to Cigarette Cues in a Human Experimental Model of Tobacco Withdrawal. Addiction 2018, 113, 1696–1705. [Google Scholar] [CrossRef] [Green Version]
  71. Chiang, K.P. Reduced Cellular Expression and Activity of the P129T Mutant of Human Fatty Acid Amide Hydrolase: Evidence for a Link between Defects in the Endocannabinoid System and Problem Drug Use. Hum. Mol. Genet. 2004, 13, 2113–2119. [Google Scholar] [CrossRef]
  72. Schlosburg, J.E.; Carlson, B.L.A.; Ramesh, D.; Abdullah, R.A.; Long, J.Z.; Cravatt, B.F.; Lichtman, A.H. Inhibitors of Endocannabinoid-Metabolizing Enzymes Reduce Precipitated Withdrawal Responses in THC-Dependent Mice. AAPS J. 2009, 11, 342–352. [Google Scholar] [CrossRef]
  73. Ren, Y.; Whittard, J.; Higuera-Matas, A.; Morris, C.V.; Hurd, Y.L. Cannabidiol, a Nonpsychotropic Component of Cannabis, Inhibits Cue-Induced Heroin Seeking and Normalizes Discrete Mesolimbic Neuronal Disturbances. J. Neurosci. 2009, 29, 14764–14769. [Google Scholar] [CrossRef] [Green Version]
  74. Peña, I.D.; Gevorkiana, R.; Shi, W.-X. Psychostimulants Affect Dopamine Transmission through Both Dopamine Transporter-Dependent and Independent Mechanisms. Eur. J. Pharmacol. 2015, 764, 562–570. [Google Scholar] [CrossRef] [Green Version]
  75. Conrad, K.; Ford, K.; Marinelli, M.; Wolf, M. Dopamine Receptor Expression and Distribution Dynamically Change in the Rat Nucleus Accumbens after Withdrawal from Cocaine Self-Administration. Neuroscience 2010, 169, 182–194. [Google Scholar] [CrossRef] [Green Version]
  76. Kramar, C.P.; I Chefer, V.; A Wise, R.; Medina, J.H.; Barbano, M.F. Dopamine in the Dorsal Hippocampus Impairs the Late Consolidation of Cocaine-Associated Memory. Neuropsychopharmacology 2014, 39, 1645–1653. [Google Scholar] [CrossRef] [Green Version]
  77. Gessa, G.; Melis, M.; Muntoni, A.; Diana, M. Cannabinoids Activate Mesolimbic Dopamine Neurons by an Action on Cannabinoid CB1 Receptors. Eur. J. Pharmacol. 1998, 341, 39–44. [Google Scholar] [CrossRef]
  78. Fanarioti, E.; Mavrikaki, M.; Panagis, G.; Mitsacos, A.; Nomikos, G.G.; Giompres, P. Behavioral and Neurochemical Changes in Mesostriatal Dopaminergic Regions of the Rat after Chronic Administration of the Cannabinoid Receptor Agonist WIN55,212-2. Int. J. Neuropsychopharmacol. 2015, 18, pyu097. [Google Scholar] [CrossRef]
  79. Jones, D. End of the Line for Cannabinoid Receptor 1 as an Anti-Obesity Target? Nat. Rev. Drug Discov. 2008, 7, 961–962. [Google Scholar] [CrossRef]
  80. Gardner, E. Endocannabinoid Signaling System and Brain Reward: Emphasis on Dopamine. Pharmacol. Biochem. Behav. 2005, 81, 263–284. [Google Scholar] [CrossRef]
  81. Lallemand, F.; De Witte, P. Ethanol Induces Higher BEC in CB1 Cannabinoid Receptor Knockout Mice While Decreasing Ethanol Preference. Alcohol Alcohol. 2004, 40, 54–62. [Google Scholar] [CrossRef]
  82. Economidou, D.; Mattioli, L.; Cifani, C.; Perfumi, M.; Massi, M.; Cuomo, V.; Trabace, L.; Ciccocioppo, R. Effect of the Cannabinoid CB1 Receptor Antagonist SR-141716A on Ethanol Self-Administration and Ethanol-Seeking Behaviour in Rats. Psychopharmacology 2005, 183, 394–403. [Google Scholar] [CrossRef]
  83. Hill, K.P.; Gold, M.S.; Nemeroff, C.B.; McDonald, W.; Grzenda, A.; Widge, A.S.; Rodriguez, C.; Kraguljac, N.V.; Krystal, J.H.; Carpenter, L.L. Risks and Benefits of Cannabis and Cannabinoids in Psychiatry. Am. J. Psychiatry 2022, 179, 98–109. [Google Scholar] [CrossRef]
  84. Bhardwaj, A.K.; Agonist Replacement for Cannabis Dependence (ARCD) study group; Allsop, D.J.; Copeland, J.; McGregor, I.S.; Dunlop, A.; Shanahan, M.; Bruno, R.; Phung, N.; Montebello, M.; et al. Randomised Controlled Trial (RCT) of Cannabinoid Replacement Therapy (Nabiximols) for the Management of Treatment-Resistant Cannabis Dependent Patients: A Study Protocol. BMC Psychiatry 2018, 18, 140. [Google Scholar] [CrossRef] [Green Version]
  85. Cohen, J.; Wei, Z.; Phang, J.; LaPrairie, R.B.; Zhang, Y. Cannabinoids as an Emerging Therapy for Posttraumatic Stress Disorder and Substance Use Disorders. J. Clin. Neurophysiol. 2020, 37, 28–34. [Google Scholar] [CrossRef]
  86. De Ternay, J.; Naassila, M.; Nourredine, M.; Louvet, A.; Bailly, F.; Sescousse, G.; Maurage, P.; Cottencin, O.; Carrieri, P.; Rolland, B. Therapeutic Prospects of Cannabidiol for Alcohol Use Disorder and Alcohol-Related Damages on the Liver and the Brain. Front. Pharmacol. 2019, 10, 627. [Google Scholar] [CrossRef]
  87. George, T.P. Medication Treatments for Nicotine Dependence; CRC/Taylor & Francis: Boca Raton, FL, USA, 2007. [Google Scholar]
  88. Janero, D.R. Cannabinoid-1 Receptor (CB1R) Blockers as Medicines: Beyond Obesity and Cardiometabolic Disorders to Substance Abuse/Drug Addiction with CB1R Neutral Antagonists. Expert Opin. Emerg. Drugs 2012, 17, 17–29. [Google Scholar] [CrossRef]
  89. Janero, D.R.; Makriyannis, A. Targeted Modulators of the Endogenous Cannabinoid System: Future Medications to Treat Addiction Disorders and Obesity. Curr. Psychiatry Rep. 2007, 9, 365–373. [Google Scholar] [CrossRef]
  90. Khurana, L.; Mackie, K.; Piomelli, D.; Kendall, D.A. Modulation of CB1 Cannabinoid Receptor by Allosteric Ligands: Pharmacology and Therapeutic Opportunities. Neuropharmacology 2017, 124, 3–12. [Google Scholar] [CrossRef] [Green Version]
  91. Lake, S.; Kerr, T.; Buxton, J.; Walsh, Z.; Cooper, Z.D.; Socías, M.E.; Fairbairn, N.; Hayashi, K.; Milloy, M.-J. The Cannabis-Dependent Relationship Between Methadone Treatment Dose and Illicit Opioid Use in a Community-Based Cohort of People Who Use Drugs. Cannabis Cannabinoid Res. 2021, 8, 155–165. [Google Scholar] [CrossRef]
  92. Luján, M.; Valverde, O. The Pro-Neurogenic Effects of Cannabidiol and Its Potential Therapeutic Implications in Psychiatric Disorders. Front. Behav. Neurosci. 2020, 14, 109. [Google Scholar] [CrossRef]
  93. Mackie, K. Cannabinoid Receptors as Therapeutic Targets. Annu. Rev. Pharmacol. Toxicol. 2006, 46, 101–122. [Google Scholar] [CrossRef] [Green Version]
  94. Onaivi, E.S. Cannabinoid Receptors in Brain. Pharmacogenetics, Neuropharmacology, Neurotoxicology, and Potential Therapeutic Applications. New Concepts Psychostimulant Induc. Neurotox. 2009, 88, 335–369. [Google Scholar] [CrossRef]
  95. Pietrzak, B.; Dunaj, A.; Piątkowska, K. The Role of the Cannabinoid System in the Pathogenesis and Treatment of Alcohol Dependence. Postępy Hig. I Med. Doświadczalnej 2011, 65, 606–615. [Google Scholar] [CrossRef]
  96. Preedy, V. Handbook of Cannabis and Related Pathologies; Elsevier: Amsterdam, The Netherlands, 2017. [Google Scholar]
  97. Rodrigues, L.A.; Caroba, M.E.S.; Taba, F.K.; Filev, R.; Gallassi, A.D. Evaluation of the Potential Use of Cannabidiol in the Treatment of Cocaine Use Disorder: A Systematic Review. Pharmacol. Biochem. Behav. 2020, 196, 172982. [Google Scholar] [CrossRef]
  98. Śmiarowska, M.; Białecka, M.; Machoy-Mokrzyńska, A. Cannabis and Cannabinoids: Pharmacology and Therapeutic Potential. Neurol. I Neurochir. Pol. 2022, 56, 4–13. [Google Scholar] [CrossRef]
  99. Weidenauer, A.; Sauerzopf, U.; Praschak-Rieder, N.; Keimpema, E.; Kasper, S.; Willeit, M. Cannabidiol in for the Treatment of Psychiatric Disorders: A New Hope or Smoke on the Water? J. Fur Neurol. Neurochir. Und Psychiatr. 2021, 22, 136–140. [Google Scholar]
  100. Yang, P.; Wang, L.; Xie, X.-Q. Latest Advances in Novel Cannabinoid CB2 Ligands for Drug Abuse and Their Therapeutic Potential. Future Med. Chem. 2012, 4, 187–204. [Google Scholar] [CrossRef] [Green Version]
Figure 1. PRISMA flow chart of narrative review search methods.
Figure 1. PRISMA flow chart of narrative review search methods.
Ijerph 20 04087 g001
Table 1. PICOS Search algorithm–scoping review using a systematic approach.
Table 1. PICOS Search algorithm–scoping review using a systematic approach.
Search ParameterInclusion Criterion
ParticipantsStudies were included if their participants met the criteria for a substance-use disorder (DSM-5), abuse, or dependence (DSM-IV).
InterventionsExperimental condition: any formulation with primary mechanism of action upon the endocannabinoid system (receptors, ligands, or enzymes) with the intention to treat a substance-use disorder, abuse, or dependence.
ControlPlacebo or any other intervention differing from the experimental condition.
OutcomeThe primary outcome was the effectiveness of the intervention for the treatment of any substance-use disorder.
Study designThe studies considered for inclusion in this review were systematic reviews (SRs), narrative reviews (NRs), and randomised-control trials (RCTs)
Table 2. Primary studies included in scoping review.
Table 2. Primary studies included in scoping review.
Primary
Papers
N = 29
Systematic Scoping Review Search Results
N = 25
Systematic Reviews
n = 5
Narrative Reviews
n = 14
Randomised Controlled Trials
n = 6
Batalla et. al. 2019,
[24]
Hoch
et. al., 2019
[25]
Paulus et. al., 2022
[26]
Pavel
et. al., 2021
[27]
Prud’hommeet. al., 2015
[28]
Babalonis
& Walsh 2020
[29]
Beardsley
et. al.,
2009
[30]
Calpe-López et al., 2019
[31]
Chye
et. al., 2019
[32]
Femenia et. al., 2009
[33]
Fischer et. al., 2015
[34]
Foll
et. al., 2008
[35]
Galaj
& Xi 2019
[36]
Kleczko-ska et. al., 2015
[37]
Kolongo-wski et. al., 2021
[38]
Lee
et. al., 2017
[39]
Navarrete
et. al.,
2021
[40]
Sholler
et. al., 2020
[41]
Sloan
et. al., 2017
[9]
Lintzeris et. al., 2019
[42]
Lintzeris et. al., 2020
[43]

Bisaga et. al., 2015
[44]
Soyka
et. al.,
2008
[45]
Meneses-Gaya et. al., 2021
[46]

Mongeau-Pérusse et. al., 2021
[47]
Allsop
et. al. 2014
[48]
Trigo
et. al., 2016
[49]
Trigo
et. al., 2018
[50]
Lintzeris
et. al., 2019
[42]
Lintzeris
et. al., 2020
[43]
Haney
et. al., 2003
[51]
Budney
et. al., 2007
[52]
Haney
et. al., 2007
[53]
Levin
et. al., 2011
[54]
Vandrey
et. al., 2013
[55]
Levin
et. al., 2016
[56]
Freeman
et. al., 2020
[57]
D’Souza
et. al., 2019
[58]
Bisaga
et. al., 2015
[44]
Jicha
et. al., 2015
[59]
Lofwall
et. al., 2016
[60]
Hurd
et. al., 2019
[61]
Soyka
et. al., 2008
[45]
George
et. al., 2009
[62]
Meneses-Gaya et. al., 2021
[46]
Mongeau-Pérusse et. al., 2021
[47]
Rigotti et. al., 2009
[63]
Cahill & Ussher 2011
[64]
Robinson et. al., 2017
[65]
Tonstad & Aubin 2012
[66]
Morrison
et. al., 2010
[67]
Morgan
et. al., 2013
[68]
Hindocha
et. al., 2018a
[69]
Hindocha
et. al., 2018b
[70]
Legend: Substance of use disorder—green = cannabis (13 RCTs); blue = opioids (4 RCTs); orange = alcohol (2 RCTs); red = cocaine (2 RCTs); purple = nicotine (8 RCTs).
Table 3. Primary Studies pertaining to Cannabis Use Disorder.
Table 3. Primary Studies pertaining to Cannabis Use Disorder.
StudyTypeNPopulationInterventionAdjunct
Intervention
DurationFollow UpComparatorOutcomesFindings
Nabiximols
Allsop et. al., 2014 [48]Randomised
Control Trial (RCT)
51Cannabis Dependence (DSM-IV), Treatment SeekingNabixi-mols,
oro-mucosal spray,
86.4 mg tetrahydrocannabinol (THC):80 mg can-nabidiol (CBD) (max dai-ly dose)
CBT Workbook,
Standard Detoxification Care
6 days28 daysPlacebo Primary:
Withdrawal Severity (CWS)

Secondary:
Cannabis Use (28 day follow-up)

Treatment Retention

↓ Withdrawal Severity

=Cannabis Use


↑ Treatment Retention
Trigo et. al., 2016 [49]RCT9Cannabis Dependence (DSM-IV), Non-Treatment SeekingNabiximols,
Oromucosal Spray,
100 mg CBD:108 mg THC (max daily dose)
n/a8 weeksn.a.PlaceboPrimary:
Withdrawal Severity (CWS)

Secondary:
Craving (MCQ)

↓ Withdrawal Severity (dose-dependent)

=Craving (MCQ)
Trigo et. al., 2018 [50]RCT40Cannabis Dependence (DSM-IV), Treatment SeekingNabiximols Oromucusal Spray,
113.4 mg THC:105 mg CBD (max daily dose)
MET
CBT
12 weeksn.a.PlaceboPrimary
Tolerability

Abstinence (EOT)

Secondary
Cannabis Use (days/week)

Withdrawal Severity (CWS)
Craving (MCQ)

=Tolerability =Abstinence

↓ Cannabis Use

= Withdrawal Severity

↓ Craving
Lintzeris et. al., 2019 [42]RCT128Cannabis Dependence (DSM-IV), Treatment SeekingNabiximols,
Oromucosal Spray,
80 mg CBD:86.4 mg THC (maximum daily doses),
CBT,
Case Management
12 weeksn.a.PlaceboPrimary:
Cannabis Use (days/trial)

Secondary:
Craving (MCQ)

Withdrawal Severity (CWS)

↓ Cannabis Use


= Craving

= Withdrawal Severity
Lintzeris et. al., 2020 [43]RCT128Cannabis Dependence (DSM-IV), Treatment SeekingNabiximols,
Oromucosal Spray,
80 mg CBD:86.4 mg THC (maximum daily doses),
CBT,
Case Management
12 weeks24weeksPlaceboPrimary:
Cannabis Use

Secondary:
Abstinence (previous 28 days)

↓ Cannabis Use


↑ Abstinence
Tetrahydrocannabinol (THC)
Haney et. al., 2003 [51]Placebo Controlled, Within Subject Study7Cannabis Users,
Non-Treatment Seeking
THC,
Oral Capsules,
5 × 10 mg
n/a 6 daysn.a.PlaceboPrimary:
Withdrawal Severity (Marijuana Withdrawal Checklist)

Secondary:
Craving

↓ Withdrawal Severity




↓ Craving
Budney et. al., 2007 [52]Placebo Controlled, Within Subject Study8Cannabis Dependence (DSM-IV), Non-Treatment SeekingTHC,
Oral Capsules,
30 mg vs. 90 mg
n/a5 daysn.a.PlaceboPrimary:
Withdrawal Severity (Marijuana Withdrawal Checklist)

Secondary:
Craving (Marijuana Craving Questionnaire)
↓ Withdrawal Severity (dose-dependent)




↓ Craving
Haney et. al., 2007 [53]Placebo Controlled, Within Subject Study8Cannabis Users,
Non-Treatment Seeking
THC 3x20 mg vs
Lofexidine 2.4 mg vs
THC + Lofexidine
n/a7 daysn.a.PlaceboPrimary:
Withdrawal Severity (Marijuana Withdrawal Checklist)

Secondary:
Relapse Cannabis Use



Craving (VAS)

↓ Withdrawal Severity (all combinations)



↓ Relapse Cannabis Use (Lofexidine, THC + Lofexidine)

↓ Craving (Lofexidine, THC + Lofexidine)
Dronabinol
Levin et. al., 2011 [54]RCT156Cannabis Dependence (DSM-IV),
Treatment Seeking
Dronabinol,
Oral Capsules,
2 × 20 mg
MET,
Relapse Prevention Therapy
9 weeksn.a.PlaceboPrimary:
Abstinence (2 weeks, EOT)

Secondary:
Cannabis Use (Self-Reported)

Withdrawal Severity (Withdrawal Discomfort Score)

=Abstinence


=Cannabis Use


↓ Withdrawal Severity
Vandrey et. al., 2013 [55]Placebo Controlled, Within Subject Study13Cannabis Dependence (DSM-IV),
Non-Treatment Seeking
Dronabinol,
Oral Capsules,
30 vs. 60 vs. 120 mg
n/a5 daysn.a.PlaceboPrimary:
Withdrawal Severity (Marijuana Withdrawal Checklist)

↓ Withdrawal Severity (dose-dependent)
Levin et. al., 2016 [56]RCT122Cannabis Dependence (DSM-IV),
Treatment Seeking
Dronabinol(3 × 20 mg) +Lofexidine(3 × 0.6 mg) MET,
Relapse Prevention Therapy
10 weeksn.a.PlaceboPrimary:
Abstinence (3 weeks, EOT)
Secondary:
Withdrawal Severity

=Abstinence


= Withdrawal Severity
Cannabidiol (CBD)
Freeman et. al., 2020 [57]Phase 2a, double-blind, placebo-controlled, randomized, adaptive Bayesian trial48CUD (DSM-V),Treatment SeekingCBD,
Oral Capsules,
200 vs. 400 vs. 800 mg
Motivational Interviewing4 weeksn.a.PlaceboPrimary:
Cannabis Use (urinary THC-COOH: creatinine conc)

Secondary:
Withdrawal Severity (Cannabis Withdrawal Scale)

↓ Cannabis Use (400 mg, 800 mg)


↓ Withdrawal
Severity (800 mg)
Fatty Acid Amide Hydrolase (FAAH) Inhibitor
D’Souza et. al., 2019 [58]Phase 2a, double-blind, placebo-controlled, randomized trial46Cannabis Dependence (DSM-IV), Treatment SeekingPF-04457845,Oral Capsules,
4 mg
n/a4 weeksn.a.PlaceboPrimary:
Cannabis Withdrawal Severity

Secondary:
Cannabis Use (Urine + Self-Reported)

↓ Cannabis Withdrawal


↓ Cannabis Use
Table 4. Primary Studies pertaining to Opioid Use Disorder.
Table 4. Primary Studies pertaining to Opioid Use Disorder.
StudyTypeNPopulationInterventionAdjunct
Intervention
DurationFollow UpComparatorOutcomesFindings
Dronabinol
Bisaga et. al., 2015 [44]RCT60Opioid Dependence (DSM-IV), Treatment SeekingDronabinol, Oral Capsule30 mgMET,
CBT,
Relapse Prevention Therapy
8 weeks.3 weeks.PlaceboPrimary
Withdrawal Severity (SOWS)

Naltrexone Treatment Retention

↓ Withdrawal Severity

= Naltrexone Treatment Retention
Jicha et. al., 2015 [59] Within Subject RCT12Opioid Dependence (DSM-IV), Non-Treatment SeekingDronabinol, Oral Capsule5 vs. 10 vs. 20 vs. 30 mg (40 mg discontinued)n/aSingle Dosen.a.Placebo, Oxycodone 30 vs. 60 mgPhysiological Tolerability↑ Heart Rate (>=20 mg)

= Physiological Parameters (<20 mg)
Lofwall et. al., 2016 [60]Within Subject RCT12Opioid Dependence (DSM-IV), Non-Treatment SeekingDronabinol, Oral Capsule5 vs. 10 vs. 20 vs. 30 mg (40 mg discontinued)n/aSingle Dosen.a.Placebo, Oxycodone 30 vs. 60 mgWithdrawal Severity (SOWS)

Psychomotor/Cognitive Effects
↓Withdrawal Severity (>=20 mg)

↑Psychomotor/Cognitive Effects
Cannabidiol (CBD)
Hurd et. al., 2019 [61]RCT42Opioid Dependence (DSM-IV)Cannabidiol, Oral Solution,
400,800 mg
n/a3 days7 daysPlaceboPrimary
Cue Induced Craving (HCQ)

Anxiety (VAS-A)

Secondary
Cognition

Affect

Physiological Markers (Heart Rate, Cortisol)

↓ Cue Induced Craving3

↓ Anxiety

=Cognition

=Affect

↓Physiological Markers (Heart Rate, Cortisol)
Table 5. Primary Studies pertaining to Alcohol Use Disorder.
Table 5. Primary Studies pertaining to Alcohol Use Disorder.
StudyTypeNPopulationInterventionAdjunct
Intervention
DurationFollow UpComparatorOutcomesFindings
Rimonabant
Soyka et. al., 2008 [45] Phase 2a RCT258Alcohol Dependence (DSM-IV), Recently DetoxifiedRimonabant, Oral Capsule,
2 × 10 mg
n/a12 weeksn.a.PlaceboPrimary
Relapse to First Drink

Relapse to Heavy Drinking

Secondary Alcohol Consumption

=Relapse to First Drink

=Relapse to Heavy Drinking

=Alcohol Consumption
George et. al., 2009 [62]Phase I/II RCT49Alcohol Dependence/Abuse (DSM-IV),
Non-Treatment Seeking
Rimonabant, Oral Capsule,
20 mg
n/a2 weeksn.a.PlaceboPrimary
Alcohol Consumption
=Alcohol Consumption
Table 6. Primary Studies pertaining to Cocaine Use Disorder.
Table 6. Primary Studies pertaining to Cocaine Use Disorder.
StudyTypeNPopulationInterventionAdjunct
Intervention
DurationFollow UpComparatorOutcomesFindings
Cannabidiol (CBD)
Meneses-Gaya et. al., 2021 [46]RCT31Crack-Cocaine Dependence (DSM-IV)CBD,
Oral Solution,
300 mg
n/a10 daysn.a.PlaceboPrimary
Cue Induced Craving Severity

=Cue Induced Craving Severity
Mongeau-Pérusse et. al., 2021 [47]Phase II RCT50Cocaine Use Disorder (DSM-VCBD,
Oral Solution,
800 mg
Group Therapy12 weeksn.a.PlaceboPrimary
Cue Induced Craving Severity

Secondary
Time to Relapse

=Cue Induced Craving Severity

=Time to Relapse
Table 7. Primary Studies pertaining to Nicotine Use Disorder.
Table 7. Primary Studies pertaining to Nicotine Use Disorder.
StudyTypeNPopulationInterventionAdjunct
Intervention
DurationFollow UpComparatorOutcomesFindings
Rimonabant
Rigotti et. al., 2009 [63]RCT755Nicotine Dependence (DSM-IV), Treatment SeekingRimonabant 20 mg
+ Nicotine Patch
Smoking Counselling10 weeks13 weeksRimonabant 20 mg
+ Placebo Patch
Primary
Abstinence (EOT, 4 Week Continuous)

Secondary
Point Prevalence Abstinence (weeks 9,24)

Sustained Abstinence (weeks 6-24)

Weight Change
↑ Abstinence (all measures)












=Weight Change
STRATUS-WW 2005 [64]Double-blind placebo-controlled parallel-assignment RCT5055Smokers (>10cpd), Treatment SeekingRimonabant5 vs. 20 mgBehavioural CounsellingPhase 1:
10 weeks

Phase 2:
42 weeks
104 weeksPlaceboPrimary
Relapse Prevention Rate

Secondary
Weight Change

↑ Relapse Prevention Rate (20 mg)

↓ Weight Gain (20 mg)
STRATUS-EU 2006
[64,65]
Double-blind placebo-controlled parallel-assignment RCT783Smokers (>10cpd), Treatment SeekingRimonabant5 vs. 20 mgBehavioural Counselling10 weeks48 weeksPlaceboPrimary
Abstinence at EOT (10 weeks) and Prolonged (48 weeks)

Secondary
Weight Gain

Adverse Events (GI Disturbance, Anxiety)

↑ Abstinence (EOT & Prolonged) (20 mg)


↓ Weight Gain (20 mg)

↑ Adverse Events (20 mg)
STRATUS-US 2006
[64,65]
Double-blind placebo-controlled parallel-assignment RCT784Smokers (>10cpd), Treatment SeekingRimonabant5 vs. 20 mgBehavioural Counselling10 weeks48 weeksPlaceboPrimary
Abstinence at EOT (10 weeks) and Prolonged (48 weeks)

Secondary
Weight Gain

Adverse Events (GI Disturbance, Anxiety)

↑ Abstinence (EOT & Prolonged) (20 mg)

↓ Weight Gain (20 mg)

↑ Adverse Events (20 mg)
STRATUS-META 2006
[65]
Double-blind placebo-controlled parallel-assignment RCT 530Smokers (>10cpd), Treatment SeekingRimonabant20mgBehavioural Counselling10 weeksn.a.PlaceboPrimary
Abstinence at EOT (10 weeks)

Secondary
Weight Gain

Adverse Events (GI Disturbance, Anxiety)

↑ Abstinence (EOT)

↓ Weight Gain

↑ Adverse Events
Surinabant
Tonstad & Aubin, 2012 [66]Double-blind placebo-controlled parallel-assignment RCT 810Smokers (>10cpdSurinabant 2.5 vs. 5 vs. 10 mgSmoking Cessation Counselling8 weeks6 weeksPlaceboPrimary
Abstinence (EOT, 4 weeks continuous)

Secondary
Weight Gain

Neuropsychiatric SE

=Abstinence


↓ Weight Gain

=Neuropsychiatric SE
Taranabant
Morrison et. al., 2010 [67]RCT317Dependent Cigarette SmokersTaranabant, Oral Capsules,
2 vs. 4 vs. 8 mg

+ Counselling
Smoking Cessation Counselling8 weeks6 weeksPlaceboPrimary
Abstinence (EOT, 4 weeks continuous)

Secondary
Weight Gain

Neuropsychiatric SE (Depression)

Gastrointestinal SE

=Abstinence


↓ Weight Gain

↑ Neuropsychiatric SE

↑Gastrointestinal SE
Cannabidiol (CBD)
Morgan et. al., 2013
[68]
RCT24Dependent Cigarette Smokers, Non-Treatment SeekingCBD,
Inhaler,
Ad Hoc Use
Smoking Cessation Counselling1 week2 weeksPlaceboPrimary
Cigarette Usage

Secondary
Craving

Mood Side Effects (Sedation, Depression, Anxiety)

↓ Cigarette Usage

=Craving

=Mood Side Effects
Hindocha et. al., 2018a
[69]
RCT Double Blind Cross-Over Design30Dependent Cigarette Smokers, Non Treatment SeekingCBD,
800 mg
n/aSingle Dosen.a.PlaceboPrimary
Attentional Bias to Cigarette Cues during Abstinence

Pleasantness of Cigarette Stimuli during Abstinence

Craving

Withdrawal

Side Effects

↓ Attentional Bias to Cigarette Cues during Abstinence

↓Pleasantness of Cigarette Stimuli during Abstinence

=Craving

=Withdrawal

=Side Effects
Hindocha et. al., 2018b
[70]
RCT Double Blind Cross-Over Design30Dependent Cigarette Smokers, Non Treatment SeekingCBD,
800 mg
n/aSingle Dosen.a.PlaceboVerbal and Spatial Working Memory

Impulsivity
=Verbal and Spatial Working Memory

=Impulsivity
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Gharbi, K.A.; Bonomo, Y.A.; Hallinan, C.M. Evidence from Human Studies for Utilising Cannabinoids for the Treatment of Substance-Use Disorders: A Scoping Review with a Systematic Approach. Int. J. Environ. Res. Public Health 2023, 20, 4087. https://doi.org/10.3390/ijerph20054087

AMA Style

Gharbi KA, Bonomo YA, Hallinan CM. Evidence from Human Studies for Utilising Cannabinoids for the Treatment of Substance-Use Disorders: A Scoping Review with a Systematic Approach. International Journal of Environmental Research and Public Health. 2023; 20(5):4087. https://doi.org/10.3390/ijerph20054087

Chicago/Turabian Style

Gharbi, Kayvan Ali, Yvonne Ann Bonomo, and Christine Mary Hallinan. 2023. "Evidence from Human Studies for Utilising Cannabinoids for the Treatment of Substance-Use Disorders: A Scoping Review with a Systematic Approach" International Journal of Environmental Research and Public Health 20, no. 5: 4087. https://doi.org/10.3390/ijerph20054087

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop