Cannabidiol as a treatment for craving and relapse in individuals with cocaine use disorder: a randomized placebo-controlled trial

Violaine Mongeau-Pérusse, Suzanne Brissette, Julie Bruneau, Patricia Conrod, Simon Dubreucq, Guillaume Gazil, Emmanuel Stip, Didier Jutras-Aswad, Violaine Mongeau-Pérusse, Suzanne Brissette, Julie Bruneau, Patricia Conrod, Simon Dubreucq, Guillaume Gazil, Emmanuel Stip, Didier Jutras-Aswad

Abstract

Background and aims: Cocaine use disorder (CUD) is a significant public health concern for which no efficacious pharmacological interventions are available. Cannabidiol (CBD) has attracted considerable interest as a promising treatment for addiction. This study tested CBD efficacy for reducing craving and preventing relapse in people with CUD.

Design: Single-site double-blind randomized controlled superiority trial comparing CBD with placebo.

Setting and participants: Centre Hospitalier de l'Université de Montréal, Canada. Seventy-eight adults (14 women) with moderate to severe CUD participated.

Intervention: Participants were randomly assigned (1 : 1) by stratified blocks to daily 800 mg CBD (n = 40) or placebo (n = 38). They first underwent an inpatient detoxification phase lasting 10 days. Those who completed this phase entered a 12-week outpatient follow-up.

Measurements: Primary outcomes were drug-cue-induced craving during detoxication and time-to-cocaine relapse during subsequent outpatient treatment.

Findings: During drug-cue exposure, craving scores [mean ± standard deviation (SD)] increased from baseline by 4.69 (2.89) versus 3.21 (2.78) points, respectively, in CBD (n = 36) and placebo (n = 28) participants [confidence interval (CI) = -0.33 to 3.04; P = 0.069; Bayes factor = 0.498]. All but three participants relapsed to cocaine by week 12 with similar risk for CBD (n = 34) and placebo (n = 27) participants (hazard ratio = 1.20, CI = 0.65-2.20, P = 0.51; Bayes factor = 0.152). CBD treatment was well tolerated and associated mainly with diarrhoea.

Conclusions: CBD did not reduce cocaine craving or relapse among people being treated for CUD.

Trial registration: ClinicalTrials.gov NCT02559167.

Keywords: Addiction; cannabidiol; cocaine; craving; human; relapse.

© 2021 The Authors. Addiction published by John Wiley & Sons Ltd on behalf of Society for the Study of Addiction.

Figures

FIGURE 1
FIGURE 1
Consolidated Standards of Reporting Trials (CONSORT) flow‐chart of participants with cocaine use disorder (CUD) involved in this trial. Participants are considered lost to follow‐up when they missed two consecutive visits. *Other reasons for ineligibility included men with fertility problems (n = 2), immunocompromised participants (n = 2) and not currently moderate or severe CUD according to the Diagnostic and Statistical Manual of Mental Disorders, 5th edition criteria (n = 1). **One participant's consent form was lost. When asked, this participant refused to re‐consent, which ended her participation. This was reported to the data safety and monitoring board who requested that no data from this participant be used in the study. CBD = cannabidiol; n = number of participants
FIGURE 2
FIGURE 2
Cocaine craving among treatment groups. Bar chart illustrating mean changes in craving scores on the 10‐point visual analogue scale in each treatment group following a cocaine, a stress and a neutral cue imagery‐induced craving session. Standard deviations are indicated on the bars with vertical lines. CBD = cannabidiol; n = number of participants
FIGURE 3
FIGURE 3
Time‐to‐cocaine‐relapse and cocaine use among treatment groups. (a) Kaplan–Meier curves illustrating the proportion of participants without cocaine relapse in each treatment group during the 12‐week follow‐up period (Phase II) together with the number of participants at risk of relapse. (b) Bar chart illustrating similar cocaine use in both treatment groups during the follow‐up period. Standard deviations are indicated on the bars with vertical lines. CBD = cannabidiol; n= number of participants
FIGURE 4
FIGURE 4
Participants’ CBD blood levels among treatment groups. Box and whisker plots illustrating minimum, first quartile, median, third quartile and maximum CBD concentrations in each treatment group. Mean and outlier values are marked with diamonds and circles, respectively. In the CBD group, 9.00 a.m. CBD blood concentration evolved from (mean ± standard deviation) 37.14 ± 14.54 ng/ml on day 8 to 67.75 ± 71.20 ng/ml on week 4 and 74.57 ± 130.33 ng/ml on week 12. At 1.00 p.m. on day 9, CBD blood concentration was 553.82 ± 379.13 ng/ml in the CBD group. The single participant treated with placebo who tested positive for CBD had a CBD blood concentration of 0.06 ng/ml. CBD = cannabidiol; D = day; n = number of participants analyzed, including those with no detectable CBD; W = week

References

    1. United Nations Office on Drugs Crime . Stimulants. World Drug Report 2019. Sales No. E.19.XI.8 Vienna, Austria: United Nations Publications; 2019, pp. 1–90.
    1. Farrell M., Martin N. K., Stockings E., Bórquez A., Cepeda J. A., Degenhardt L., et al. Responding to global stimulant use: challenges and opportunities. Lancet 2019; 394: 1652–1667.
    1. Butler A. J., Rehm J., Fischer B.Health outcomes associated with crack‐cocaine use: systematic review and meta‐analyses. Drug Alcohol Depend 2017; 180: 401–416.
    1. Arendt M., Munk‐Jørgensen P., Sher L., Jensen S. O.Mortality among individuals with cannabis, cocaine, amphetamine, MDMA, and opioid use disorders: a nationwide follow‐up study of Danish substance users in treatment. Drug Alcohol Depend 2011; 114: 134–139.
    1. Back S. E., Hartwell K., DeSantis S. M., Saladin M., McRae‐Clark A. L., Price K. L., et al. Reactivity to laboratory stress provocation predicts relapse to cocaine. Drug Alcohol Depend 2010; 106: 21–27.
    1. De Crescenzo F., Ciabattini M., D'Alo G. L., De Giorgi R., Del Giovane C., Cassar C., et al. Comparative efficacy and acceptability of psychosocial interventions for individuals with cocaine and amphetamine addiction: a systematic review and network meta‐analysis. PLOS Med 2018; 15: e1002715.
    1. Chan B., Kondo K., Freeman M., Ayers C., Montgomery J., Kansagara D.Pharmacotherapy for cocaine use disorder‐a systematic review and meta‐analysis. J Gen Intern Med 2019; 34: 2858–2873.
    1. Indave B. I., Minozzi S., Pani P. P., Amato L.Antipsychotic medications for cocaine dependence. Cochrane Database Syst Rev 2016; 3: Cd006306. 10.1002/14651858.CD006306.pub3
    1. Prud'homme M., Cata R., Jutras‐Aswad D.Cannabidiol as an intervention for addictive behaviors: a systematic review of the evidence. Subs Abuse Res Treat 2015. 10.4137/SART.S25081
    1. 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.
    1. Fischer B., Kuganesan S., Gallassi A., Malcher‐Lopes R., van den Brink W., 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.
    1. 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.
    1. 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: 172982.
    1. Chesney E., Oliver D., Green A., Sovi S., Wilson J., Englund A., et al. Adverse effects of cannabidiol: a systematic review and meta‐analysis of randomized clinical trials. Neuropsychopharmacology 2020; 45: 1799–1806.
    1. Vilela L. R., Gomides L. F., David B. A., Antunes M. M., Diniz A. B., Moreira F. A., et al. Cannabidiol rescues acute hepatic toxicity and seizure induced by cocaine. Mediators Inflamm 2015; 2015.
    1. Rong C., Lee Y., Carmona N. E., Cha D. S., Ragguett R.‐M., Rosenblat J. D., et al. Cannabidiol in medical marijuana: research vistas and potential opportunities. Pharmacol Res 2017; 121: 213–218.
    1. Preston K. L., Epstein D. H.Stress in the daily lives of cocaine and heroin users: relationship to mood, craving, relapse triggers, and cocaine use. Psychopharmacology 2011; 218: 29–37.
    1. Luján M. Á., Cantacorps L., Valverde O.The pharmacological reduction of hippocampal neurogenesis attenuates the protective effects of cannabidiol on cocaine voluntary intake. Addict Biol 2020; 25: e12778.
    1. Gonzalez‐Cuevas G., Martin‐Fardon R., Kerr T. M., Stouffer D. G., Parsons L. H., Hammell D. C., et al. Unique treatment potential of cannabidiol for the prevention of relapse to drug use: preclinical proof of principle. Neuropsychopharmacology 2018; 43: 2036.
    1. Hurd Y. L., Yoon M., Manini A. F., Hernandez S., Olmedo R., Ostman M., et al. Early phase in the development of cannabidiol as a treatment for addiction: opioid relapse takes initial center stage. Neurotherapeutics 2015; 12: 807–815.
    1. Hurd Y. L., Spriggs S., Alishayev J., Winkel G., Gurgov K., Kudrich C., et al. 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.
    1. Freeman T. P., Hindocha C., Baio G., Shaban N. D., Thomas E. M., Astbury 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.
    1. Hindocha C., Freeman T. P., Grabski M., Stroud J. B., Crudgington H., Davies A. C., et al. Cannabidiol reverses attentional bias to cigarette cues in a human experimental model of tobacco withdrawal. Addiction 2018; 113: 1696–1705.
    1. Meneses‐Gaya C. C. J., Hallak J. E., Miguel A. Q., Laranjeira R., Bressan R. A., Zuardi A. W., et al. Cannabidiol for the treatment of crack‐cocaine craving: an exploratory double‐blind study. Rev Bras Psiquiatr 2020. 10.1590/1516-4446-2020-1416
    1. First M. B.Structured clinical interview for the DSM (SCID). In: Cautin R. L., Lilienfeld S. O., editors. The Encyclopedia of Clinical Psychology; 2014, pp. 1–6. 10.1002/9781118625392.wbecp351
    1. Robinson S. M., Sobell L. C., Sobell M. B., Leo G. I.Reliability of the timeline followback for cocaine, cannabis, and cigarette use. Psychol Addict Behav 2014; 28: 154.
    1. Sheehan D., Janavs J., Baker R., Sheehan K., Knapp E., Sheehan M.The Mini‐International Neuropsychiatric Interview, Version 7.0 for DSM‐5 (MINI 7.0). Jacksonville, FL: Medical Outcomes Systems; 2014.
    1. Fox H. C., Garcia M., Kemp K., Milivojevic V., Kreek M. J., Sinha R.Gender differences in cardiovascular and corticoadrenal response to stress and drug cues in cocaine dependent individuals. Psychopharmacology 2006; 185: 348–357.
    1. Ferri C. P., Dunn J., Gossop M., Laranjeira R.Factors associated with adverse reactions to cocaine among a sample of long‐term, high‐dose users in Sao Paulo. Brazil Add Behav 2004; 29: 365–374.
    1. James K., Bloch D., Lee K., Kraemer H., Fuller index for assessing blindness in a multi‐centre clinical trial: disulfiram for alcohol cessation—a VA cooperative study. Stat Med 1996; 15: 1421–1434.
    1. Iffland K., Grotenhermen update on safety and side effects of cannabidiol: a review of clinical data and relevant animal studies. Cannabis Cannabinoid Res 2017; 2: 139–154.
    1. Sinha R.Modeling stress and drug craving in the laboratory: implications for addiction treatment development. Addict Biol 2009; 14: 84–98.
    1. Castells X., Casas M., Vidal X., Bosch R., Roncero C., Ramos‐Quiroga J. A., et al. Efficacy of central nervous system stimulant treatment for cocaine dependence: a systematic review and meta‐analysis of randomized controlled clinical trials. Addiction 2007; 102: 1871–1887.
    1. Levine J., Nina R.SAFTEE: a technique for the systematic assessment of side. Psychopharmacol Bull 1986; 22: 343.
    1. Lacroix C., Saussereau E.Fast liquid chromatography/tandem mass spectrometry determination of cannabinoids in micro volume blood samples after dabsyl derivatization. J Chromatogr B 2012; 905: 85–95.
    1. Sinha R., Talih M., Malison R., Cooney N., Anderson G. M., Kreek M. J.Hypothalamic–pituitary–adrenal axis and sympatho–adreno–medullary responses during stress‐induced and drug cue‐induced cocaine craving states. Psychopharmacology 2003; 170: 62–72.
    1. Jeffreys H. In Theory of Probability. Oxford Classic Texts in the Physical Sciences 3rd ed.. Oxford, United Kingdom: Oxford University Press; 1961, pp. 1–470.
    1. Beard E., Dienes Z., Muirhead C., West R.Using Bayes factors for testing hypotheses about intervention effectiveness in addictions research. Addiction 2016; 111: 2230–2247.
    1. Jones H. E., Johnson R. E., Bigelow G. E., Silverman K., Mudric T., Strain E. C.Safety and efficacy of L‐tryptophan and behavioral incentives for treatment of cocaine dependence: a randomized clinical trial. Am J Addict 2004; 13: 421–437.
    1. Linares I. M., Zuardi A. W., Pereira L. C., Queiroz R. H., Mechoulam R., Guimaraes F. S., et al. Cannabidiol presents an inverted U‐shaped dose–response curve in a simulated public speaking test. Rev Bras Psiquiatr 2019; 41: 9–14.
    1. Haney M., Malcolm R. J., Babalonis S., Nuzzo P. A., Cooper Z. D., Bedi G., et al. Oral cannabidiol does not alter the subjective, reinforcing or cardiovascular effects of smoked cannabis. Neuropsychopharmacology 2016; 41: 1974–1982.
    1. 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.
    1. Galaj E., Bi G.‐H., Yang H.‐J., Xi Z.‐X.Cannabidiol attenuates the rewarding effects of cocaine in rats by CB2, 5‐HT1A and TRPV1 receptor mechanisms. Neuropharmacology 2020; 167: 107740.

Source: PubMed

3
購読する