Scolaris Content Display Scolaris Content Display

Pharmacothérapies pour la dépendance au cannabis

Esta versión no es la más reciente

Contraer todo Desplegar todo

Referencias

References to studies included in this review

Allsop 2014 {published data only}

Allsop DJ, Copeland J, Lintzeris N, Dunlop AJ, Montebello M, et al. Nabiximols as an agonist replacement therapy during cannabis withdrawal: a randomized clinical trial. JAMA Psychiatry 2014;71(3):281‐91. [DOI: 10.1001/jamapsychiatry.2013.3947]

Carpenter 2009 {published data only}

Carpenter KM, McDowell D, Brooks DJ, Cheng WY, Levin FR. A preliminary trial: double‐blind comparison of nefazodone, bupropion‐SR, and placebo in the treatment of cannabis dependence. American Journal on Addictions 2009;18(1):53‐64.

Cornelius 2010 {published data only}

Cornelius JR, Bukstein OG, Douaihy AB, Clark DB, Chung TA, et al. Double‐blind fluoxetine trial in comorbid MDD‐CUD youth and young adults. Drug and Alcohol Dependence 2010;112(1‐2):39‐45. [PsycINFO AN 2010‐26726‐005]

Frewen 2007 {published and unpublished data}

Frewen A, Montebello ME, Baillie A, Rea F. Effects of mirtazapine on withdrawal from dependent cannabis use. Proceedings of the 69th Annual Scientific Meeting of the College on Problems of Drug Dependence; June 16‐21; Quebec City, Canada 2007. 21. [MEDLINE: FREWEN2007]
Frewen AR. An examination of withdrawal symptoms and their relationship with outcomes in a combined behavioural and pharmacological intervention for dependent cannabis users. Thesis (Macquarie University)2009.

Gray 2012 {published data only}

Gray KM, Carpenter MJ, Baker NL, DeSantis SM, Kryway E, et al. A double‐blind randomized controlled trial of N‐acetylcysteine in cannabis‐dependent adolescents. American Journal of Psychiatry 2012;169(8):805‐12. [DOI: 10.1176/appi.ajp.2012.12010055; EMBASE: AN 2012469181]

Johnston 2012 {published data only}

Johnston J, Lintzeris N, McGregor I, Allsop DJ, Helliwell D, Winstock A. A double blind, randomised, placebo controlled trial of lithium carbonate for the management of cannabis withdrawal [conference abstract]. Drug and Alcohol Review 2013;32:43.
Johnston J, Lintzeris N, McGregor I, Guastella A, Allsop D, Helliwell D, Winstock A. Preliminary findings from a double blind, randomised, placebo controlled trial of lithium carbonate for the management of cannabis withdrawal [conference abstract]. Drug and Alcohol Review 2012;31:45.

Levin 2004 {published data only}

Levin FR, McDowell D, Evans SM, Nunes E, Akerele E, Donovan S, et al. Pharmacotherapy for marijuana dependence: a double‐blind, placebo‐controlled pilot study of divalproex sodium. American Journal on Addictions 2004;13:21‐32. [MEDLINE: AN 14766435]

Levin 2011 {published data only}

Levin FR, Mariani JJ, Brooks DJ, Pavlicova M, Cheng W, Nunes EV. Dronabinol for the treatment of cannabis dependence: A randomized, double‐blind, placebo‐controlled trial. Drug and Alcohol Dependence 2011;116:142‐50. [DOI: 10.1016/j.drugalcdep.2010.12.010; MEDLINE: AN 21310551]

Levin 2013 {published data only}

Levin FR, Mariani J, Brooks DJ, Pavlicova M, Nunes EV, et al. A randomized double‐blind, placebo‐controlled trial of venlafaxine‐extended release for co‐occurring cannabis dependence and depressive disorders. Addiction 2013;108(6):1084‐94. [DOI: 10.1111/add.12108; MEDLINE: AN 23297841]

Mason 2012 {published data only}

Mason BJ, Crean R, Goodell V, Light JM, Quello S, Shadan F, et al. A proof‐of‐concept randomized controlled study of gabapentin: effects on cannabis use, withdrawal and executive function deficits in cannabis‐dependent adults. Neuropsychopharmacology 2012;37:1689‐98. [DOI: 10.1038/npp.2012.14; MEDLINE: AN 22373942]

McRae‐Clark 2009 {published data only}

McRae‐Clark AL, Carter RE, Killeen TK, Carpenter MJ, Wahlquist AE, Simpson SA, Brady KT. A placebo‐controlled trial of buspirone for the treatment of marijuana dependence. Drug and Alcohol Dependence 2009;105:132‐8. [CENTRAL: CN‐00729425; DOI: 10.1016/j.drugalcdep.2009.06.022]

McRae‐Clark 2010 {published data only}

McRae‐Clark AL, Carter RE, Killeen TK, Carpenter MJ, White KG, Brady KT. A placebo‐controlled trial of atomoxetine in marijuana‐dependent individuals with attention deficit hyperactivity disorder. American Journal on Addictions 2010;19(6):481‐9.

Penetar 2012 {published data only}

Penetar DM, Looby AR, Ryan ET, Maywalt MA, Lukas SE. Bupropion reduces some of the symptoms of marihuana withdrawal in chronic marihuana users: A pilot study. Substance Abuse: Research and Treatment 2012;6:63‐71.

Weinstein 2014 {published data only}

Weinstein AM, Miller H, Bluvstein I, Rapoport E, Schreiber S, et al. Treatment of cannabis dependence using escitalopram in combination with cognitive‐behavior therapy: A double‐blind placebo‐controlled study. American Journal of Drug and Alcohol Abuse 2014;40(1):16‐22. [DOI: 10.3109/00952990.2013.819362; EMBASE: AN 2013816531]

References to studies excluded from this review

Akerele 2007 {published data only}

Akerele E, Levin FR. Comparison of olanzapine to risperidone in substance‐abusing individuals with schizophrenia. American Journal on Addictions 2007;16:260‐8. [DOI: 10.1080/10550490701389658]

Brown 2013 {published data only}

Brown PC, Budney AJ, Thostenson JD, Stanger C. Initiation of abstinence in adolescents treated for marijuana use disorders. Journal of Substance Abuse Treatment 2013;44(4):384‐90. [EMBASE: AN 2013109720]

Budney 2007 {published data only}

Budney AJ, Higgins ST, Radonovich KJ, Novy PL. Oral delta‐9‐tetrahydrocannabinol suppresses cannabis withdrawal symptoms. Drug and Alcohol Dependence 2007;86(1):22‐9. [CENTRAL: CN‐00590656]

Cooper 2013 {published data only}

Cooper ZD, Foltin RW, Hart CL, Vosburg SK, Comer SD, Haney M. A human laboratory study investigating the effects of quetiapine on marijuana withdrawal and relapse in daily marijuana smokers. Addiction Biology 2013;18(6):993‐1002. [EMBASE: AN 2013769387]

Cornelius 1999 {published data only}

Cornelius JR, Salloum IM, Haskett RF, Ehler JG, Jarrett PJ, Thase ME, Perel JM. Fluoxetine versus placebo for the marijuana use of depressed alcoholics. Addictive Behaviours 1999;24(1):111‐4.

Cornelius 2008 {published data only}

Cornelius JR, Chung T, Martin C, Wood DS, Clark DB. Cannabis withdrawal is common among treatment‐seeking adolescents with cannabis dependence and major depression, and is associated with rapid relapse to dependence. Addictive Behaviors 2008;33(11):1500‐5. [DOI: 10.1016/j.addbeh.2008.02.001; MEDLINE: 18313860]

Daynes 1994 {published data only}

Daynes G, Gillman MA. Psychotropic analgesic nitrous oxide prevents craving after withdrawal for alcohol, cannabis and tobacco. International Journal of Neuroscience 1994;76:13‐6. [MEDLINE: 7960461]

Findling 2009 {published data only}

Findling RL, Pagano ME, McNamara NK, Stansbrey RJ, Faber JE, Lingler J, et al. The short‐term safety and efficacy of fluoxetine in depressed adolescents with alcohol and cannabis use disorders: a pilot randomized placebo‐controlled trial. Child and Adolescent Psychiatry and Mental Health 2009;3:11. [CENTRAL: CN‐00754249; DOI: 10.1186/1753‐2000‐3‐11]
Hirschtritt ME, Pagano ME, Christian KM, McNamara NK, Stansbrey RJ, et al. Moderators of fluoxetine treatment response for children and adolescents with comorbid depression and substance use disorders. Journal of Substance Abuse Treatment 2012;42(4):366‐72. [DOI: 10.1016/j.jsat.2011.09.010; PsycINFO AN 2011‐27672‐001]

Geller 1998 {published data only}

Geller B, Cooper TB, Sun K, Zimermann B, Frazier J, et al. Double‐blind and placebo‐controlled study of lithium for adolescent bipolar disorders with secondary substance dependency. Journal of the American Academy of Child and Adolescent Psychiatry 1998;37(2):171‐8. [PsycINFO AN 1997‐39092‐009]
Geller B, Cooper TB, Watts HE, Cosby CM, Fox LW. Early findings from a pharmacokinetically designed double‐blind and placebo‐controlled study of lithium for adolescents comorbid with bipolar and substance dependency disorders. Progress in Neuro‐Psychopharmacology and Biological Psychiatry 1992;16(3):281‐99. [EMBASE: AN 1992154297]

Gillman 2006 {published data only}

Gillman MA, Harker N, Lichtigfeld FJ. Combined cannabis/methaqualone withdrawal treated with psychotropic analgesic nitrous oxide. International Journal of Neuroscience 2006;116:859‐69. [CENTRAL: CN‐00566721; DOI: 10.1080/00207450600753998]

Gray 2010 {published data only}

Gray KM, Watson NL, Carpenter MJ, Larowe SD. N‐Acetylcysteine (NAC) in young marijuana users: An open‐label pilot study. American Journal on Addictions 2010;19(2):187‐9. [DOI: 10.1111/j.1521‐0391.2009.00027.x; EMBASE: 2010123976]

Haney 2001 {published data only}

Haney M, Ward AS, Comer SD, Hart CL, Foltin RW, Fischman MW. Bupropion SR worsens mood during marijuana withdrawal in humans. Psychopharmacology 2001;155(2):171‐9.

Haney 2003 {published data only}

Haney M, Bisaga A, Foltin RW. Interaction between naltrexone and oral THC in heavy marijuana smokers. Psychopharmacology 2003;166:77‐85. [DOI: 10.1007/s00213‐002‐1279‐8]

Haney 2003a {published data only}

Haney M, Hart CL, Ward AS, Foltin RW. Nefazodone decreases anxiety during marijuana withdrawal in humans. Psychopharmacology 2003;165(2):157‐65.

Haney 2004 {published data only}

Haney M, Hart CL, Vosburg SK, Nasser J, Bennett A, Zubaran C, Foltin RW. Marijuana withdrawal in humans: effects of oral THC or divalproex. Neuropsychopharmacology 2004;29:158‐70.

Haney 2008 {published data only}

Haney M, Hart CL, Vosburg SK, Comer SD, Reed SC, Foltin RW. Effects of THC and lofexidine in a human laboratory model of marijuana withdrawal and relapse. Psychopharmacology 2008;197(1):157‐68.

Haney 2010 {published data only}

Haney M, Hart CL, Vosburg SK, Comer SD, Reed SC, Cooper ZD, Foltin RW. Effects of baclofen and mirtazapine on a laboratory model of marijuana withdrawal and relapse. Psychopharmacology 2010;211(2):233‐44. [CENTRAL: CN‐00760900; DOI: 10.1007/s00213‐010‐1888‐6]

Haney 2013 {published data only}

Haney M, Bedi G, Cooper Z, Vosburg S, Comer S, Foltin R. Nabilone decreases marijuana withdrawal and relapse in the human laboratory. Neuropsychopharmacology 2011;36:S177‐8. [DOI: 10.1038/npp.2011.291; EMBASE: AN 70607410]
Haney M, Cooper ZD, Bedi G, Vosburg SK, Comer SD, Foltin RW. Nabilone decreases marijuana withdrawal and a laboratory measure of marijuana relapse. Neuropsychopharmacology 2013;38(8):1557‐65. [DOI: 10.1038/npp.2013.54; MEDLINE: AN 23443718]

Haney 2013a {published data only}

Haney M, Cooper Z, Bedi G, Reed SC, Ramesh D, Foltin RW. Marijuana withdrawal and relapse in the human laboratory: Effect of zolpidem alone and in combination with nabilone. Neuropsychopharmacology 2013;38:S508‐9. [DOI: 10.1038/npp.2013.281; EMBASE: AN 71278673]

Hart 2002 {published data only}

Hart CL, Haney M, Ward AS, Fischman MW, Foltin RW. Effects of oral THC maintenance on smoked marijuana self‐administration. Drug and Alcohol Dependence 2002;67(3):301‐9. [MEDLINE: AN 12127201]

Imbert 2014 {published data only}

Imbert B, Labrune N, Lancon C, Simon N. Baclofen in the management of cannabis dependence syndrome. Therapeutic Advances in Psychopharmacology 2014;4(1):50‐2. [10.1177 / 2045125313512324]

Levin 2008 {published data only}

Levin FR, Kleber HD. Use of dronabinol for cannabis dependence: two case reports and review. American Journal on Addictions 2008;17:161‐4. [DOI: 10.1080/10550490701861177; MEDLINE: 18393061]

McRae 2006 {published data only}

McRae AL, Brady KT, Carter RE. Buspirone for treatment of marijuana dependence: a pilot study. American Journal on Addictions 2006;15:404. [DOI: 10.1080/10550490600860635; MEDLINE: 16966201]

Robinson 2006 {published data only}

Robinson DG, Woerner MG, Napolitano B, Patel RC, Sevy SM, et al. Randomized comparison of olanzapine versus risperidone for the treatment of first‐episode schizophrenia: 4‐month outcomes. American Journal of Psychiatry 2006;163(12):2096‐102. [DOI: 10.1176/appi.ajp.163.12.2096]
Sevy S, Robinson DG, Sunday S, Napolitano B, Miller R, et al. Olanzapine vs. risperidone in patients with first‐episode schizophrenia and a lifetime history of cannabis use disorders: 16‐week clinical and substance use outcomes. Psychiatry Research 2011;188(3):310‐4. [DOI: 10.1016/j.psychres.2011.05.001]

Subodh 2011 {published data only}

Nanjayya SB, Shivappa M, Chand PK, Murthy P, Benegal V. Baclofen in cannabis dependence syndrome. Biological Psychiatry 2010;68:e9‐e10. [DOI: 10.1016/j.biopsych.2010.03.033; MEDLINE: 20494335]
Subodh N, Chand P, Murthy P, Madhusudhan S, Benegal V. Baclofen in cannabis dependence syndrome ‐ a open‐label study and a brief follow up. European Psychiatry, Abstracts of the 19th European Congress of Psychiatry 2011;26 Suppl 1:110. [DOI: 10.1016/S0924‐9338(11)71821‐1]

Sugarman 2011 {published data only}

Sugarman DE, Poling J, Sofuoglu M. The safety of modafinil in combination with oral 9‐tetrahydrocannabinol in humans. Pharmacology, Biochemistry and Behavior 2011;98(1):94‐100. [DOI: 10.1016j.pbb.2010.12.013]

Tirado 2008 {published data only}

Tirado CF, Goldman M, Lynch K, Kampman KM, Obrien CP. Atomoxetine for treatment of marijuana dependence: A report on the efficacy and high incidence of gastrointestinal adverse effects in a pilot study. Drug and Alcohol Dependence 2008;94:254‐7. [DOI: 10.1016/j.drugalcdep.2007.10.020]

Vandrey 2011 {published data only}

Vandrey R, Smith MT, McCann UD, Budney AJ, Curran EM. Sleep disturbance and the effects of extended‐release zolpidem during cannabis withdrawal. Drug and Alcohol Dependence 2011;117:38‐44. [DOI: 10.1016/j.drugalcdep.2011.01.003]

Vandrey 2013 {published data only}

Vandrey R, Stitzer ML, Mintzer MZ, Huestis MA, Murray JA, Lee D. The dose effects of short‐term dronabinol (oral THC) maintenance in daily cannabis users. Drug and Alcohol Dependence 2013;128(1‐2):64‐70. [DOI: 10.1016/j.drugalcdep.2012.08.001; MEDLINE: AN 22921474]

Van Nimwegen 2008 {published data only}

Van Nimwegen LJ, De Haan L, Van Beveren NJM, Van Der Helm M, Van Den Brink W, Linszen D. Effect of olanzapine and risperidone on subjective well‐being and craving for cannabis in patients with schizophrenia or related disorders: A double‐blind randomized controlled trial. Canadian Journal of Psychiatry 2008;53(6):400‐5. [EMBASE: AN 2008305264]

Winstock 2009 {published data only}

Winstock AR, Lea T, Copeland J. Lithium carbonate in the management of cannabis withdrawal in humans: an open‐label study. Journal of Psychopharmacology 2009;23(1):84‐93. [CENTRAL: CN‐00714374; DOI: 10.1177/0269881108089584]

Additional references

Agrawal 2008

Agrawal A, Pergadia ML, Lynskey MT. Is there evidence for symptoms of cannabis withdrawal in the National Epidemiologic Survey of Alcohol and Related Conditions?. American Journal on Addictions2008; Vol. 17, issue 3:199‐208.

Beauvais 2004

Beauvais F, Jumper‐Thurman P, Helm H, Plested B, Burnside M. Surveillance of drug use among American Indian adolescents: patterns over 25 years. Journal of Adolescent Health2004; Vol. 34, issue 6:493‐500. [1054‐139X]

Benyamina 2008

Benyamina A, Lecacheux M, Blecha L, Reynaud M, Lukasiewcz M. Pharmacotherapy and psychotherapy in cannabis withdrawal and dependence. Expert Review of Neurotherapeutics March 2008;8(3):479‐91.

Budney 2003

Budney AJ, Moore BA, Vandrey RG, Hughes JR. The time course and significance of cannabis withdrawal. Journal of Abnormal Psychology2003; Vol. 112, issue 3:393‐402.

Budney 2006

Budney AJ, Hughes JR. The cannabis withdrawal syndrome. Current Opinion in Psychiatry2006; Vol. 19, issue 3:233‐8.

Budney 2007a

Budney AJ, Roffman R, Stephens RS, Walker D. Marijuana dependence and its treatment. Addiction Science & Clinical Practice2007; Vol. 4, issue 1:4‐16.

Chung 2008

Chung T, Martin CS, Cornelius JR, Clark DB. Cannabis withdrawal predicts severity of cannabis involvement at 1‐year follow‐up among treated adolescents. Addiction2008; Vol. 103, issue 5:787‐99.

Clough 2004

Clough AR, D'Abbs P, Cairney S, Gray D, Maruff P, Parker R, et al. Emerging patterns of cannabis and other substance use in Aboriginal communities in Arnhem Land, Northern Territory: a study of two communities. Drug and Alcohol Review2004; Vol. 23, issue 4:381‐90.

Copeland 2001

Copeland J, Swift W, Rees V. Clinical profile of participants in a brief intervention program for cannabis use disorder. Journal of Substance Abuse Treatment2001; Vol. 20, issue 1:45‐52.

Copeland 2014

Copeland J, Clement N, Swift W. Cannabis use, harms and the management of cannabis use disorder. Neuropsychiatry 2014;4(1):55‐63. [DOI: 10.2217/npy.13.90]

Copersino 2006

Copersino ML, Boyd SJ, Tashkin DP, Huestis MA, Heishman SJ, Dermand JC, et al. Cannabis withdrawal among non‐treatment‐seeking adult cannabis users. American Journal on Addictions2006; Vol. 15, issue 1:8‐14.

Danovitch 2012

Danovitch I, Gorelick DA. State of the art treatments for cannabis dependence. Psychiatric Clinics of North America 2012;35(2):309‐26. [DOI: 10.1016/j.psc.2012.03.003]

DSM‐5

American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition. dsm.psychiatryonline.org2013.

EMCDDA 2004

European Monitoring Centre for Drugs and Drug Addiction. Cannabis problems in context: understanding the increase in European treatment demands. http://www.emcdda.europa.eu/html.cfm/index34898EN.htmlNovember 2004, issue Selected Issue 2:81‐92.

EMCDDA Cannabis Drug Profile

European Monitoring Centre for Drugs and Drug Addiction. Cannabis Drug Profile. http://www.emcdda.europa.eu/publications/drug‐profiles/cannabis Page last updated:19 July 2010.

Gruber 2003

Gruber AJ, Pope HG, Hudson JI, Yurgelun‐Todd D. Attributes of long‐term heavy cannabis users: a case‐control study. Psychological Medicine2003; Vol. 33, issue 8:1415‐22.

Hall 2001

Hall W, Degenhardt L, Lynskey M. The health and psychological effects of cannabis use. 2nd Edition. Canberra, Australia: Commonwealth of Australia, 2001. [National Drug Strategy Monograph No. 44]

Hall 2009

Hall W, Degenhardt L. Adverse health effects of non‐medical cannabis use. Lancet2009; Vol. 374, issue 9698:1383‐91.

Hart 2005

Hart CL. Increasing treatment options for cannabis dependence: A review of potential pharmacotherapies. Drug and Alcohol Dependence2005; Vol. 80, issue 2:147‐59.

Hasin 2008

Hasin DS, Keyes KM, Alderson D, Wang S, Aharonovich E, Grant BF. Cannabis withdrawal in the United States: Results from NESARC. Journal of Clinical Psychiatry2008; Vol. 69, issue 9:1354‐63.

Higgins 2011

Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 [updated March 2011]. The Cochrane Collaboration 2011. www.cochrane‐handbook.org.

Large 2011

Large M, Sharma S, Compton MT, Slade T, Nielssen O. Cannabis use and earlier onset of psychosis. Archives of General Psychiatry 2011;68(6):555‐61.

Lev‐Ran 2013

Lev‐Ran S, Le Foll B, McKenzie K, George TP, Rehm J. Cannabis use and cannabis use disorders among individuals with mental illness. Comprehensive Psychiatry 2013;54:589‐98.

Lev‐Ran 2013a

Lev‐Ran S, Le Strat Y, Imtiaz S, Rehm J, Le Foll B. Gender differences in prevalence of substance use disorders among individuals with lifetime exposure to substances: Results from a large representative sample. American Journal on Addictions 2013;22:7‐13.

McClure 2012

McClure EA, Stitzer ML, Vandrey R. Characterizing smoking topography of cannabis in heavy users. Psychopharmacology 2012;220:309‐18.

Milin 2008

Milin R, Manion I, Dare G, Walker S. Prospective assessment of cannabis withdrawal in adolescents with cannabis dependence: A pilot study. Journal of the American Academy of Child & Adolescent Psychiatry2008; Vol. 47, issue 2:174‐9.

Minozzi 2010

Minozzi S, Davoli M, Bargagli M, Amato L, Vecchi S, Perucci C. An overview of systematic reviews on cannabis and psychosis: Discussing apparently conflicting results. Drug and Alcohol Review May 2010;29(3):304‐17.

Nordstrom 2007

Nordstrom BR, Levin FR. Treatment of cannabis use disorders: A review of the literature. American Journal on Addictions2007; Vol. 16:331‐42.

SAMHSA 2008

Substance Abuse and Mental Health Services Administration, Office of Applied Studies. Treatment Episode Data Set (TEDS): 1996‐2006. National Admissions to Substance Abuse Treatment Services. www.oas.samhsa.gov/copies.cfm. Rockville, MD, United States: Substance Abuse and Mental Health Services Administration, 2008. [DASIS Series: S‐43, DHHS Publication No. (SMA) 08‐4347]

Solowij 2008

Solowij N, Battisti R. The chronic effects of cannabis on memory in humans: A review. Current Drug Abuse Reviews 2008;1(1):81‐98.

Stead 2012

Stead LF, Perera R, Bullen C, Mant D, Hartmann‐Boyce J, Cahill K, Lancaster T. Nicotine replacement therapy for smoking cessation. Cochrane Database of Systematic Reviews 2012, Issue 11. [DOI: 10.1002/14651858.CD000146.pub4]

van der Pol 2014

van der Pol P, Liebregts N, Brunt T, van Amsterdam J, de Graaf R, et al. Cross‐sectional and prospective relation of cannabis potency, dosing and smoking behaviour with cannabis dependence: an ecological study. Addiction 2014;109:1101‐9.

Vandrey 2009

Vandrey R, Haney M. Pharmacotherapy for cannabis dependence: How close are we?. CNS Drugs2009; Vol. 23, issue 7:543‐53.

Volkow 2014

Volkow ND, Baler RD, Compton WM, Weiss SRB. Adverse health effects of marijuana use. New England Journal of Medicine 2014;370:2219‐27.

Wagner 2002

Wagner FA, Anthony JC. From first drug use to drug dependence; developmental periods of risk for dependence upon marijuana, cocaine, and alcohol. Neuropsychopharmacology2002; Vol. 26, issue 4:479‐88.

World Drug Report 2013

United Nations Office on Drugs and Crime. World Drug Report 2013. www.unodc.org.

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Allsop 2014

Methods

Double‐blind, randomised, placebo‐controlled trial. Setting: inpatient (two hospitals), New South Wales, Australia. Funding: research grant (Australian National Health and Medical Research Council), with study drugs provided by manufacturer (GW Pharmaceucticals, UK). Declaration of conflict of interest not published

Participants

N = 51 adults seeking treatment for cannabis use, dependent by DSM‐IV‐TR. Average age 35; 76% male; 53% unemployed; 25% married or in de facto relationship; on average using 23 g cannabis per day, average duration of use 20 years; 71% also nicotine dependent. Dependence on alcohol or other drugs except nicotine or caffeine and unstable medical or psychiatric conditions were exclusion criteria. Groups well matched apart from differences in baseline withdrawal score and disability scale scores

Interventions

(1) N = 27, nabiximols (cannabis extract, Sativex®), maximum dose 86.4 mg THC, 80 mg cannabidiol; 6 days medication, 3 days washout, or (2) N = 24, placebo. Cognitive‐behavioural therapy tailored to inpatient cannabis withdrawal as adjunct intervention. Total 9 days inpatient admission. Follow‐up interview after 28 days. Participants compensated AUD 40 for follow‐up interviews

Outcomes

Overall withdrawal score, irritability, craving, and depression reported as graphs and results of statistical analyses with imputation for missing data. Number retained in treatment at all time points, median days inpatient stay. Change in amount of cannabis use from baseline to 28‐day follow‐up

Notes

Withdrawal and craving assessed with Cannabis Withdrawal Scale (19 items on 11‐point Likert scale for the previous 24 hours). Drug use by modified timeline follow‐back

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "An independent statistician generated a randomization list for each site using random block sizes in Stata, version 11.1 ..."

Allocation concealment (selection bias)

Low risk

Comment: Method of allocation concealment not reported, but generation of lists by independent statistician and use of matching placebos would be expected to provide good control of bias

Blinding (subjective outcomes)
All outcomes

Low risk

Quote: "Patients, investigators, and outcome assessors were blind to treatment allocation until all research procedures were complete. Blinding was maintained by the use of a matched placebo ... The success of patient blinding was formally assessed before hospital discharge."

Blinding (objective outcomes)
All outcomes

Low risk

Quote: "Patients, investigators, and outcome assessors were blind to treatment allocation until all research procedures were complete. Blinding was maintained by the use of a matched placebo ... The success of patient blinding was formally assessed before hospital discharge."

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Statistical methods used to impute missing data and assess data as missing at random

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Carpenter 2009

Methods

Double‐blind, randomised, placebo‐controlled trial. Setting: outpatient clinic, New York, USA. Funding from research grant (NIDA). One author declared past associations with pharmaceutical companies

Participants

N = 106 participants seeking treatment for problems related to cannabis use, cannabis dependent by DSM‐IV and smoking at least 5 cannabis joints per week. Average age 32; 76% male (63% in bupropion group); 34% Caucasian, 28% Hispanic, 27% African‐American; 91% employed. Exclusion criteria for the trial included "significant and unstable psychiatric condition", “chronic organic mental disorder” and "DSM‐IV dependence criteria for another substance"

Interventions

Placebo for 1 week then (1) N = 36, oral nefazodone, 150 mg/day to maximum 600 mg/day (2) N = 40, oral bupropion‐SR 150 mg to maximum of 300 mg/day, or (3) N = 30, oral placebo for 10 weeks. Riboflavin added to medication to monitor adherence. All participants received placebo for 2 weeks after medication phase. Participants attended treatment clinic twice weekly (paid USD 5 for transport costs at each visit); medications dispensed weekly. Weekly individual psychosocial intervention based on coping skills as adjunct therapy. Scheduled duration 13 weeks

Outcomes

Number completing 13 weeks of study, number abstinent at week 10, dependence severity at baseline and week 10 (and improvement), withdrawal symptoms, sleep, HAM‐A at baseline and week 10. Total side effects during study

Notes

Cannabis use assessed by self‐report and urine toxicology of observed samples provided at each clinic visit, with a cut‐off of 100 ng/ml (rather than usual 50 ng/ml) to minimise false positives. Severity of dependence symptoms assessed by Clinical Global Impression (scores from 1 = no pathology, to 7 = extreme pathology). Sleep quality self‐reported once a week using the St Mary's Hospital Sleep Questionnaire. Irritability self‐reported every other week with the Snaith Irritability Scale (4 items each rated 0 to 3). Hamilton anxiety scale (14 items each rated 0 to 4) administered by clinician every other week. If either urine or self‐report data were missing for a given week, it was considered a non‐abstinent week

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "A research pharmacist who was independent of the research team, conducted the randomization"

Comment: Method of sequence generation not reported, but the involvement of an independent pharmacist would be expected to protect against bias

Allocation concealment (selection bias)

Low risk

Quote: "All capsules were prepared at the research pharmacy and looked identical for all three treatment conditions"

Comment: although not specifically stated, treatment allocation was likely to have been through medication provided by the research pharmacist making it unlikely that participants or investigators could foresee intervention assignment. Characteristics of participants in three groups similar, except significantly more females in bupropion group

Blinding (subjective outcomes)
All outcomes

Low risk

Study stated to have been conducted double‐blind, without specification as to whether participants, observers and treating personnel were all blinded to group allocation. However, the provision of active and placebo medications in identical capsules, and the use of urine screening to support self‐report data would be expected to be associated with a low risk of bias

Blinding (objective outcomes)
All outcomes

Low risk

Study conducted double‐blind and these outcomes less likely to be affected by knowledge of treatment allocation. The use of riboflavin to confirm medication adherence would help to reduce the risk of bias

Incomplete outcome data (attrition bias)
All outcomes

Low risk

There was substantial dropout from all three groups, with only 52 of 106 (49%) participants randomised completing the 10‐week medication phase and 43% completing the full 13‐week trial. Quote: "Survival analysis revealed no statistically significant group differences on treatment retention... there were no differences between those participants who completed the trial and those who did not on demographic indices or baseline substance use measures." Comment: Missing data on cannabis use regarded as indicative of “non‐abstinence”; statistical methods used to allow for missing data

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Cornelius 2010

Methods

Double‐blind, randomised, placebo‐controlled trial. One physician remained non‐blinded to handle any potential problems. Setting: outpatient clinic, Pittsburgh, USA. Funding from research grants (NIDA, NIAAA, Veterans Affairs). All authors declared no conflict of interest

Participants

N = 70 adolescents and young adults (aged 14 to 25 at baseline) with comorbid major depression and cannabis use disorder by DSM‐IV criteria. Average age 21.1; 61% male; 56% Caucasian, 37% African‐American; 94% cannabis dependent, using on average of 76% of days in prior month; 28.6% also alcohol dependent. Bipolar disorder, schizoaffective disorder, schizophrenia, substance abuse or dependence other than alcohol, nicotine or cannabis, history of IV drug use were exclusion criteria

Interventions

(1) N = 34, fluoxetine, 10 mg increasing to 20 mg/day after 2 weeks (2) N = 36, placebo. Nine sessions (delivered at each clinic visit) of manual‐based cognitive‐behavioural therapy for depression and cannabis use and motivation enhancement therapy for cannabis use as adjunct intervention. Scheduled duration 12 weeks

Outcomes

Severity of abuse or dependence (criteria count), days cannabis used in past week, number completing treatment

Notes

Depressive symptoms rated by observer with Hamilton Rating Scale for Depression and by participants with Beck Depression Inventory. Cannabis use behaviours assessed by timeline follow‐back method

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

“Patient randomization was conducted by urn randomization stratified by gender…”

Allocation concealment (selection bias)

Low risk

“Active medication and matching placebo were prepared by the research pharmacy…”

Blinding (subjective outcomes)
All outcomes

Low risk

Quote: "The study was conducted in a double‐blind fashion, though [one] physician ... remained non‐blinded in order to handle any problems which may have arisen." This suggests it is likely that participants, treating personnel and observers were all blind to group allocation

Blinding (objective outcomes)
All outcomes

Low risk

Study conducted double‐blind, as indicated above

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Authors note “low percentage of missing data”. Missing data handled by carrying forward last observation

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Frewen 2007

Methods

Double‐blind, randomised, placebo‐controlled trial. Setting: outpatient, Sydney, Australia. Funding: not reported. No declaration of conflict of interest made

Participants

N = 81 adults, seeking treatment for cannabis use, used cannabis in 72 hours prior to assessment interview, dependent by DSM‐IV‐TR in previous 3 months. Average age 31.4; 81% male; 78% Australian‐born; 64% employed; 92% living in stable accommodation; 63% not in a relationship. Average of 12 years cannabis use; 97% daily smokers; 63% daily tobacco smokers. Psychiatric or medical instability were exclusion criteria. Characteristics of participants similar to characteristics of general population seeking treatment for cannabis use

Interventions

1) Oral mirtazapine 30 mg/day or 2) placebo

Weekly individual cognitive‐behavioural therapy as adjunct intervention

Reimbursement of AUD 30 for expenses at the day 56 interview

Scheduled 4 weeks medication, with follow‐up 28 days later

Outcomes

Withdrawal symptoms in first seven days related to subsequent cannabis use for groups combined (effect of medication not considered in this analysis). Measures of sleep quality and disruption

Notes

Withdrawal symptoms measured daily for 14 days with the Marijuana Withdrawal Scale (32 items, rated from 0 = "none" to 3 = "severe").  

Cannabis use assessed with the drug scale from the Opiate Treatment Index.

Sleep problems recorded with the Karolinksa Sleep Questionnaire for 7 days, and the Pittsburgh Sleep Quality Index (24 items, global score 0 to 21, with higher scores indicative of poorer sleep) at baseline and days 28 and 56

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: “Participants were randomized ... using permuted block randomisation.”

Allocation concealment (selection bias)

Low risk

Quote: “Randomisation was independently assigned by pharmacy staff offsite.” " ... the placebo was identically matched in colour, shape, size and taste to the medication."

Comment: As independent pharmacy staff controlled the randomization process, it is likely to have prevented investigators and participants from foreseeing allocation assignment

Blinding (subjective outcomes)
All outcomes

Low risk

Quote: "All treating physicians, psychologists and research staff were blind to the randomisation until all participants had completed the final research interview."

Blinding (objective outcomes)
All outcomes

Low risk

Quote: "All treating physicians, psychologists and research staff were blind to the randomisation until all participants had completed the final research interview."

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Insufficient information available to form a view

Selective reporting (reporting bias)

Unclear risk

Limited study results available

Other bias

Low risk

None apparent

Gray 2012

Methods

Double‐blind, randomised, placebo‐controlled trial. Setting: outpatient, university clinic, South Carolina, USA. Funding: research grants (NIDA, National Center for Research Resources). Authors declared "no competing interests"

Participants

N = 116 adolescents (age 13 to 21), cannabis‐dependent and using cannabis regularly. Average age 18.9 years; 73% male; 83.5% Caucasian; 73.9% enrolled in school. Average 22.6 days with cannabis use in 30 days prior to baseline; 57% smoked tobacco; 13.8% had a psychiatric comorbidity. Dependence on other substances (except nicotine) and unstable psychiatric or medical illness were exclusion criteria

Interventions

(1) N = 58, N‐acetylcysteine 1200 mg twice daily or (2) N = 58, placebo. Twice‐weekly contingency management and weekly brief (10 minute) individual cessation counselling as adjunct therapies. Initial contingent reward USD 5 (cash) for both adherence and abstinence with amount increased by USD 2 for each successive visit; reward reset to baseline if conditions not met. Seen in clinic weekly. Follow‐up 4 weeks after treatment conclusion. Scheduled duration 8 weeks

Outcomes

Likelihood of negative urine test reported as odds ratio and 95% confidence interval. Occurrence of adverse events (number of events and number of participants). Proportion of medication doses consumed, discontinuation of medication due to adverse effects. Number completing treatment, median days in treatment, contingency rewards earned

Notes

Urine cannabinoid testing at all visits. Self‐reported cannabis use by timeline follow‐back. Medication diaries and weekly pill counts used to determine adherence. Participants lost to follow‐up or absent for visits were coded as having a positive urine test

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised in 1:1 parallel group allocation stratified by age and baseline cannabis use. No significant group differences at baseline suggesting appropriate sequence generation

Allocation concealment (selection bias)

Low risk

Quote: "university investigational drug service oversaw randomization, encased medication in identical‐appearing capsules, and dispensed them in weekly blister packs..."

Blinding (subjective outcomes)
All outcomes

Low risk

Quote: "Participants, investigators and clinical staff remained blind to treatment assignment throughout the study."

Blinding (objective outcomes)
All outcomes

Low risk

Quote: "Participants, investigators and clinical staff remained blind to treatment assignment throughout the study."

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing data and non‐attendance regarded as indicating non‐abstinence

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Johnston 2012

Methods

Double‐blind, randomised, placebo‐controlled trial. Setting: inpatient withdrawal unit; Sydney, Australia. Funding source not reported. No declaration of conflict of interest made

Participants

N = 38 cannabis dependent adults. No other participant characteristics reported

Interventions

(1) N = 19, lithium carbonate, 500 mg bd or (2) N = 19, placebo. Scheduled 7 days inpatient treatment. Follow‐up at 14, 30 and 90‐days post‐discharge

Outcomes

Withdrawal severity by Cannabis Withdrawal Scale; retention; number and severity of adverse effects

Notes

Conference abstracts only ‐ limited data

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random allocation stated; method of sequence generation not reported

Allocation concealment (selection bias)

Unclear risk

Random allocation stated; method of allocation concealment not reported

Blinding (subjective outcomes)
All outcomes

Unclear risk

Double‐blind stated, but adequacy of control for assessment of subjective outcomes (withdrawal severity) unclear

Blinding (objective outcomes)
All outcomes

Low risk

Double‐blind stated and these outcomes unlikely to be affected by awareness of group allocation

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Insufficient information reported to assess risk

Selective reporting (reporting bias)

Unclear risk

Insufficient information reported to assess risk

Other bias

Unclear risk

Insufficient information reported to assess risk

Levin 2004

Methods

Double‐blind, randomised, placebo‐controlled trial. Study included a cross‐over phase which was not included in this review due to substantial dropout (> 30%) in the first 2 weeks. Setting: outpatient with two clinic visits per week; New York, USA. Funding: Research grants (NIDA). Declaration of conflict of interest not published

Participants

N = 27 enrolled, N = 25 randomized; cannabis dependent by DSM‐IV, using daily. Average age 32; 92% male; 56% Caucasian, 20% Hispanic, 24% African American; average (± SD) joints smoked per week at baseline (1) 28.3 ± 23.2 (2) 19.4 ± 16.4. Dependence on other substances, except caffeine and nicotine, and psychiatric disorder requiring medical intervention were exclusion criteria

Interventions

Two‐week single‐blind placebo lead‐in phase, then (1) N = 13, oral divalproex sodium commenced at 500 mg/day, increasing to maximum of 2 g/day, depending on response, or (2) N = 12, placebo. Medication in 2 doses per day. Weekly individual cognitive‐behavioural relapse prevention therapy as adjunct. Scheduled duration 8 weeks (plus subsequent cross‐over phase that was excluded from this review)

Outcomes

Outcomes reported for N = 19 who completed 8 weeks of study: frequency and amount of cannabis use and craving score at baseline and weeks 7 and 8; number completing scheduled treatment; number with 2 or more weeks of assumed abstinence

Notes

Urine samples collected and analysed at each visit. Participants reported cannabis use and completed a visual analogue scale of intensity and desire for cannabis each week. Clinician‐rated global impression assessment for cannabis use completed weekly. "Strict abstinence" defined as at least one negative urine sample and no self‐reported cannabis use for that week. "Assumed abstinence" if patient reported no cannabis use and urine samples had THC‐COOH levels at least 50% below the previous week

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Twenty‐seven participants were enrolled and 25 were randomized." Comment: method of sequence generation not reported

Allocation concealment (selection bias)

Unclear risk

Quote: [participants] "...were randomly assigned to receive either divalproex or a matching placebo." Comment: method of allocation concealment not reported

Blinding (subjective outcomes)
All outcomes

Low risk

Quote: "Following randomization, patients received...either divalproex sodium or a placebo using a double‐blind design"

Comment: use of urine screening to support determination of "abstinence" would be expected to help reduce bias in these outcomes

Blinding (objective outcomes)
All outcomes

Low risk

Quote: "Following randomization, patients received...either divalproex sodium or a placebo using a double‐blind design"

Comment: these outcomes considered unlikely to be affected by knowledge of group allocation

Incomplete outcome data (attrition bias)
All outcomes

High risk

Rates of dropout were similar in the two groups, but there was no discussion of possible differences between those retained and those who dropped out of the study. Cannabis use outcomes were reported only for those who completed treatment

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

The cross‐over phase of the trial was excluded from analyses and this review due to high rates of dropout in the first two weeks

Levin 2011

Methods

Randomised, double‐blind, placebo‐controlled, trial. Randomisation after 1‐week placebo lead‐in phase. Those who used cannabis less than twice a week during the placebo lead‐in phase were not randomised. Setting: outpatient with clinic attendance twice weekly, New York, USA. Funding: research grant (NIDA). One author declared prior associations with pharmaceutical companies

Participants

N = 156 adults seeking outpatient treatment for problems related to cannabis use, dependent by DSM‐IV‐TR, using cannabis at least 5 days a week in prior 28 days. Average age 38; 82% male; 60% employed full‐time, 13% part‐time; 31% married. Significant psychiatric condition and dependence on other substances except nicotine were exclusion criteria. No significant group differences in demographic or clinical characteristics at baseline

Interventions

Placebo for 1 week, then 1) N = 79, oral dronabinol, commenced at 10 mg/day, titrated to 20 mg twice a day or the maximum tolerated, or 2) N = 77, placebo. Medication maintained to end of week 8 then tapered over 2 weeks. Weekly individual therapy based on coping skills plus motivational enhancement therapy as adjunct intervention. Participants earned vouchers with value increased by USD 1.50 for each consecutive visit, with value reset for non‐attendance, and USD 10 for returning their pill bottle and remaining medication. Maximum possible earnings were USD 570. Cash payments of USD 5 to 20 were made at each visit for transport costs

Outcomes

Number achieving 2 weeks abstinence in weeks 7 and 8 and median maximum consecutive days abstinence; number retained in study to week 8; average number of therapy sessions attended; number experiencing any adverse effects, requiring dose reduction, serious adverse events and number withdrawn due to adverse events; withdrawal scores reported as graph and results of statistical modelling; medication compliance

Notes

Cannabis use assessed by timeline follow‐back. Urine samples tested at each clinic visit for confirmation of self‐report. Withdrawal symptoms assessed twice a week using the Withdrawal Discomfort Score (10 items, scores 0‐30). Craving by Marijuana Craving Questionnaire with the 47‐item version completed once a month, and the 12‐item version weekly. Side effects assessed twice a week using the Modified Systematic Assessment for Treatment and Emergent Events (SAFTEE). Hamilton Anxiety and Depression scales used

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: “participants were randomized ...using a fixed block size of 4, stratified by joints used per week…and whether or not they were receiving a psychotropic medication.”

Allocation concealment (selection bias)

Low risk

Quote:“A research pharmacist, who was independent of the research team, conducted the randomization.”

Blinding (subjective outcomes)
All outcomes

Low risk

Quote: "Donabinol...or matching placebo...was prepared by the pharmacy...packaged in matching gelatin capsules with lactose filler and an equal amount of riboflavin. All capsules looked identical..."

Comment: double‐blind stated. Participants may have been able to distinguish the effects of dronabinol, but use of urine screening to support self‐report would be expected to reduce risk of bias

Blinding (objective outcomes)
All outcomes

Low risk

Double‐blind stated. Packaging of medication in identical capsules as above. Objective outcomes less likely to be influenced by awareness of group allocation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: “All analyses were conducted on the intent‐to‐treat population.” “...missing data in weeks 7 and 8 were scored as indicating cannabis use...”

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Levin 2013

Methods

Randomised, double‐blind, placebo‐controlled trial. Outpatient setting with twice weekly clinic attendance, New York, USA. Funding: research grants (NIDA). Two authors declared prior associations with pharmaceutical companies

Participants

N = 103 seeking treatment for problems related to cannabis use, cannabis dependence and major depressive disorder or dysthymia by DSM‐IV. Average age 35; 74% male; 40% working full‐time; 18% currently married; average 27.4 days of use in month prior to baseline. No significant group differences on demographic or clinical characteristics at baseline. Physical dependence on substances other than cannabis or nicotine was an exclusion criterion

Interventions

One‐week placebo lead‐in phase ‐ those who improved as assessed by Clinical Global Impression rating were not randomised. (1) N = 51, venlafaxine‐extended release, up to 375 mg on a fixed‐flexible schedule or (2) N = 52, placebo. Medication dose titrated over 3 weeks, then maintained for 8 weeks. Weekly individual cognitive behavioural therapy that primarily targeted cannabis use as adjunct intervention. Participants received USD 5 to 20 per visit for transport costs, and USD 10 per week if they returned their pill bottles and any remaining medication. Scheduled duration 12 weeks

Outcomes

Abstinence defined by 2 or more consecutive urine‐confirmed abstinent weeks. Improvement in depressive symptoms by Hamilton Depression Rating Scale

Notes

Cannabis use assessed by timeline follow‐back. Urine THC levels tested at each visit, with cut‐off of 100 ng/ml to decrease the probability of false positives. Side effects assessed weekly using the Modified Systematic Assessment for Treatment and Emergent Events

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "randomized at the end of the [placebo] lead‐in phase using a computer‐generated fixed‐block size of 4, with a 1:1 allocation ratio, and stratified by joints used per week...and severity of depression"

Comment: similarities of groups at baseline suggest adequate method of sequence generation

Allocation concealment (selection bias)

Low risk

Quote: "A research pharmacist, who was independent of the research team, conducted the randomization and maintained the allocation sequence." Venlafaxine or placebo "was prepared by the pharmacy...packaged in matching gelatin capsules with lactose filler."

Comment: allocation by pharmacy and identical appearance of medication and placebo would support adequate concealment of allocation

Blinding (subjective outcomes)
All outcomes

Low risk

Quote: "Participants, care providers and outcome assessors were kept blinded to the allocation."

Blinding (objective outcomes)
All outcomes

Low risk

Quote: "Participants, care providers and outcome assessors were kept blinded to the allocation."

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Patients who dropped out were significantly younger and less likely to be married, but rates of dropout were similar in the two arms. Those who dropped out without achieving 2 continuous weeks of abstinence were classified as not abstinent

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Mason 2012

Methods

Randomised, double‐blind, placebo controlled trial. Setting: outpatient with weekly clinic visits, California, USA. Funding: research grants (NIDA). One author declared past associations with pharmaceutical companies

Participants

N = 50 treatment‐seeking volunteers with current cannabis dependence by DSM‐IV, smoked cannabis at least once in week prior to randomisation. Average age 33.9 years, 88% male, average 11.6 years of daily cannabis use, smoking an average of 11.0 g/week; 62% employed full‐time; 40% married. Abuse or dependence on substances other than cannabis or nicotine, and significant psychiatric disorders were exclusion criteria. No significant group differences on demographic or clinical variables at baseline

Interventions

1) N = 25, oral gabapentin 300 mg, increasing to 1200 mg/day, or 2) N = 25, matched placebo. Abstinence‐oriented individual counselling weekly. Scheduled duration 12 weeks

Outcomes

Change in amount of cannabis use, frequency of use and withdrawal symptoms, as graphs and results of statistical tests. Number completing treatment

Notes

Cannabis use by weekly urine toxicology and self‐report by timeline follow‐back interview. Withdrawal symptoms by Marijuana Withdrawal Checklist. Marijuana Problems Scale completed at baseline and end of treatment (week 12)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: “subjects were randomly assigned … in a 1:1 ratio, on the basis of a computer‐generated randomization code.”

Allocation concealment (selection bias)

Low risk

Quote: “The randomization code was kept by the study pharmacist, who provided subjects with a 1‐week supply of medication in a blister card package at each weekly study visit…”

Comment: allocation by pharmacy and identical appearance of medication and placebo would support adequate concealment of allocation

Blinding (subjective outcomes)
All outcomes

Low risk

Quote: "Subjects, care providers, and those assessing outcomes were blinded to the identity of drug assignment. Gabapentin was purchased and over‐encapsulated to match placebo capsules."

Blinding (objective outcomes)
All outcomes

Low risk

Quote: "Subjects, care providers, and those assessing outcomes were blinded to the identity of drug assignment. Gabapentin was purchased and over‐encapsulated to match placebo capsules."

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

High rate of dropout. Extent of missing data, and adjustments for missing data unclear

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

McRae‐Clark 2009

Methods

Randomised, double‐blind, placebo‐controlled trial. N = 93 randomised; N = 34 did not receive study drug (21 failed to return for second baseline visit); analysis based on those randomised who received study drug and completed at least one post‐baseline visit. Setting: outpatient with clinic visits 1 to 2 times per week, South Carolina, USA. Funding: research grant (NIDA). Two authors declared past associations with pharmaceutical companies

Participants

N = 50 with current cannabis dependence by DSM‐IV. Average age 31.6; 90% male; 86% Caucasian; on average used cannabis on 89% of days prior to study entry, using average 3.8 g/day. Dependence on other substances except caffeine or nicotine, history of psychotic disorder, current major depression were exclusion criteria. Treatment groups similar on baseline characteristics

Interventions

(1) N = 23, oral buspirone, initiated at 5 mg twice a day, increased 5 to 10 mg every 3 to 4 days as tolerated to maximum 60 mg per day or (2) N = 27, placebo. Motivational interviewing (3 sessions) as adjunct intervention for first four weeks. Subjects received USD 10 for time and travel associated with study visits. Scheduled duration 12 weeks

Outcomes

Urinalysis data reported as per cent of screens that were negative, not participants with negative screens. Mean change in withdrawal score. Number experiencing any adverse effect. Number completing treatment. Change in reported cannabis use per using day, % days abstinent during study

Notes

Cannabis use by timeline follow‐back for 90 days prior to study entry, and weekly throughout the study. Craving by Marijuana Craving Questionnaire, withdrawal, by Marijuana Withdrawal Checklist. Urine drug screens at baseline and weekly during study. Side effects evaluated weekly with open‐ended questions. Adjustment for missing data by last observation carried forward

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Urn randomization ...was used to determine treatment assignment. Urn variables used were age ... gender, and [anxiety] score..."

Allocation concealment (selection bias)

Low risk

Quote: [participants] "Randomized at central pharmacy..." "Buspirone and placebo tablets were packaged in identical opaque gelatin capsules with cornstarch."

Blinding (subjective outcomes)
All outcomes

Low risk

Double‐blind stated. Urinalysis to support self‐report data would be expected to reduce bias, although authors noted some inconsistencies between urine screen and self‐report data

Blinding (objective outcomes)
All outcomes

Low risk

Double‐blind stated and these outcomes considered unlikely to be affected by awareness of group allocation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

High rate of dropout but statistical methods used to adjust for missing data (GEE modelling and last observation carried forward)

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

McRae‐Clark 2010

Methods

Randomised, double‐blind, placebo‐controlled trial; 78 participants were randomised but only 46 received study medication and only 38 returned for at least one post‐baseline assessment. Analyses based on this group. Setting: outpatient, South Carolina, USA. Funding: research grants (NIDA), with medication and placebo provided by manufacturer (Eli Lilly and Company). Two authors declared past associations with pharmaceutical companies

Participants

N = 38 adults, cannabis dependence and attention deficit hyperactivity disorder (with age of onset before 12 years of age) by DSM‐IV. Average age 29.9 years; 76% male; 92% Caucasian; used cannabis on average 87% of days prior to baseline, using average of 4.1 times per day. Dependence on other substances except caffeine or nicotine, and other psychiatric disorders were exclusion criteria. No significant group differences on baseline characteristics

Interventions

(1) N = 19, oral atomoxetine started at 25 mg, increased to 40 mg in week 2, and to 80 mg in week 3 as tolerated, with further increase to 100 mg/day in week 4 if required, or (2) N = 19, matching placebo. Motivational interviewing (3 sessions) as adjunct intervention. Nominal monetary reimbursement for completion of study assessments. Scheduled duration 12 weeks

Outcomes

Self‐reported cannabis use during week 12 (last observation carried forward for participants who did not complete the trial). Number completing treatment. Change in craving scores. Number experiencing adverse effects and type of adverse effects

Notes

Cannabis use self‐reported by timeline follow‐back weekly and assessed by Clinical Global Impression of Severity and Improvement Scales. Urine drug screens at baseline and then weekly. Medication side effects weekly by standard checklist. Craving by Marijuana Craving Questionnaire. Compliance assessed by patient report and pill count

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Simple randomization was used to assign treatments to participants using a 1:1 allocation ratio."

Allocation concealment (selection bias)

Low risk

Quote: "...participants were randomized at the central pharmacy..."

Blinding (subjective outcomes)
All outcomes

Low risk

Double‐blind stated. Use of matching capsules along with urine screening to validate self‐report data would be expected to reduce the risk of bias

Blinding (objective outcomes)
All outcomes

Low risk

Double‐blind stated and these outcomes unlikely to be affected by awareness of group allocation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

High rates of dropout in both groups. Last observation carried forward and statistical techniques used to allow for missing data

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Penetar 2012

Methods

Randomised, double‐blind, placebo‐controlled trial. Setting: outpatient with daily clinic attendance Monday to Friday, Harvard Medical School, USA. Funding: research grant (NIDA). Disclosures of interests according to ICMJE criteria were a requirement of publication

Participants

N = 22 treatment seeking, with cannabis abuse or dependence by DSM‐IV, with at least 3 years of heavy use (smoking on 5 or more days a week or more than 25 times per month) and with 2 or more negative symptoms in previous quit attempts. Demographic data were provided only for N = 9 who completed the study (5 male, average age 31.2 years, 7 met criteria for dependence). Abuse or dependence on any other drug (including nicotine) was an exclusion criterion

Interventions

Participants used cannabis as usual for 7 days then commenced 1) N = 10, oral bupropion‐SR (sustained release) 150 mg/day for days 1 to 3, then 150 mg twice a day or 2) N = 12, placebo. Cannabis use stopped on day 8 (Quit Day). Tobacco and caffeine continued throughout the study. Weekly individual motivational enhancement therapy (3 sessions) as adjunct intervention. Scheduled duration 21 days

Outcomes

Data reported as graphs and results of statistical tests. Relevant outcomes reported were completion of study, change in withdrawal discomfort and change in craving

Notes

Withdrawal by Marijuana Withdrawal Checklist (29 items each rated 0‐3). Withdrawal discomfort score calculated from 10 items (max score 30). Drug use, sleep and withdrawal recorded by participants in daily diary. With each medication administration participants consumed identical appearing capsule that contained riboflavin to measure compliance. Urine testing to confirm drug use

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random allocation to treatment group stated, but method of sequence generation not reported

Allocation concealment (selection bias)

Unclear risk

Method of allocation concealment not reported

Blinding (subjective outcomes)
All outcomes

Low risk

Double‐blind stated and "Bupropion tablets were repackaged into gelatin capsules...Placebo consisted of identical appearing gelatin capsules". Use of urine screening to verify self‐report expected to reduce risk of bias

Blinding (objective outcomes)
All outcomes

Low risk

Double‐blind stated, placebo used, and these outcomes less likely to be affected by awareness of group allocation

Incomplete outcome data (attrition bias)
All outcomes

High risk

High rate of dropout and demographics reported only for those who completed treatment. Unclear whether there were differences between the groups, or between those who did and did not complete the study. Unclear how missing data were handled

Selective reporting (reporting bias)

Unclear risk

Data on adverse effects not reported

Other bias

Low risk

None apparent

Weinstein 2014

Methods

Randomised, double‐blind, placebo‐controlled trial. Setting: outpatient, Tel Aviv, Israel. Funding: research grant (Israeli anti‐drug authority). Authors declared no conflict of interest

Participants

N = 52, regular cannabis users, dependent by DSM‐IV. Average age 32.7, 75% male. Dependence on other drugs or alcohol and significant psychiatric disorders were exclusion criteria

Interventions

One week "induction" with placebo, then (1) N = 26, escitalopram 10 mg/day, or (2) N = 26 placebo. Medication for 9 weeks, follow‐up sessions for further 14 weeks. Blinding broken after 9 weeks; participants able to continue open‐label escitalopram use. Participants instructed to stop cannabis use after 4 weeks of medication. Weekly (9 sessions) cognitive‐behaviour (relapse prevention) and motivation enhancement therapy in combination with medication. Scheduled duration 9 weeks

Outcomes

Number completing treatment, number abstinent, number reporting not taking medication, results of statistical analyses of withdrawal scores

Notes

Urine samples collected every second week. Questionnaires administered to assess anxiety and depression. Revised Clinical Institute Withdrawal Assessment Scale (CIWA) adapted for assessment of cannabis withdrawal (score of 10 or more indicated significant withdrawal)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "participants were blindly randomized..." Method of sequence generation not reported

Allocation concealment (selection bias)

Unclear risk

Quote: "participants were blindly randomized..." Method of allocation concealment not reported

Blinding (subjective outcomes)
All outcomes

Low risk

Double‐blind stated. Subjective outcomes not reported

Blinding (objective outcomes)
All outcomes

Low risk

Double‐blind stated and these outcomes unlikely to be affected by awareness of group allocation

Incomplete outcome data (attrition bias)
All outcomes

High risk

High (50%) rate of dropout. Those who did not complete study were younger, and more likely to be daily alcohol drinkers. Non‐completers marginally more depressed, but difference not statistically significant

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Akerele 2007

Participants were diagnosed with abuse or dependence on marijuana or cocaine. Data was reported separately for cocaine and marijuana use, but it was not possible to extract data just for those dependent on marijuana. All participants were diagnosed with schizophrenia; the management of substance use in the context of schizophrenia was the main focus of the study

Brown 2013

Secondary analysis of data from a randomised controlled trial comparing two behavioural interventions. No use of medications

Budney 2007

Laboratory study involving non‐treatment seeking cannabis users. Not all users were cannabis dependent, and participants were not trying to reduce their cannabis use

Cooper 2013

Laboratory study involving marijuana smokers who were not seeking treatment. Investigation of research model of withdrawal and relapse rather than treatment intervention

Cornelius 1999

Randomised controlled trial comparing fluoxetine and placebo for treatment of alcohol dependence with comorbid major depression. Effect on subgroup with diagnosed marijuana abuse considered as secondary analysis

Cornelius 2008

Reports cannabis withdrawal symptoms in participants entering two separate trials of fluoxetine. No treatment intervention for cannabis dependence

Daynes 1994

No treatment comparison. Unclear if participants are cannabis dependent. Insufficient information on participants and treatment regimes

Findling 2009

Randomised controlled trial comparing fluoxetine and placebo for treatment of depressive symptoms in adolescents with comorbid substance use disorder. Cannabis use reported by 88.2% of participants (41.2% dependent). The emphasis of this study is on the amelioration of depression. Outcome data not reported separately for the subset of cannabis‐dependent participants

Geller 1998

Randomised controlled trial comparing lithium and placebo for treatment of adolescents with bipolar disorder and comorbid substance use disorder. Majority of participants were polydrug users ‐ 2 of 25 were dependent on cannabis only

Gillman 2006

Reports the use of nitrous oxide for treatment of withdrawal associated with the smoking of methaqualone combined with cannabis. Unclear how many participants were cannabis dependent. All participants received placebo then analgesic nitrous oxide. Effectiveness assessed only in terms of improvement in withdrawal symptoms

Gray 2010

Open‐label single group study investigating the effectiveness of N‐Acetylcysteine in promoting cessation of cannabis use. No treatment comparison

Haney 2001

Comparison of bupropion and placebo in terms of effect on mood when administered in conjunction with active or placebo cannabis cigarettes. Laboratory study which aimed to assess the therapeutic potential of buproprion, but not a treatment intervention

Haney 2003

Investigation of mechanism of effects of cannabis through comparison of naltrexone and methadone, administered prior to oral THC, and different doses of oral THC administered in combination with naltrexone or placebo. No treatment intervention

Haney 2003a

Laboratory study comparing the effect of nefazodone (450mg/day) and placebo on the acute effects of cannabis, and on cannabis withdrawal symptoms. The study aimed to assess the therapeutic potential of nefazodone in cannabis withdrawal but was not a treatment intervention

Haney 2004

Two separate laboratory‐based studies, one assessing THC and the other divalproex, compared to placebo, in terms of effects on cannabis withdrawal. Studies aimed to assess the therapeutic potential of THC and divalproex but were not treatment interventions

Haney 2008

Laboratory study investigating the effect of lofexidine and THC (separately and in combination) compared with placebo on cannabis withdrawal symptoms and a model of cannabis relapse. The study aimed to test the therapeutic potential of lofexidine in cannabis withdrawal but was not a treatment intervention

Haney 2010

Controlled laboratory study investigating the effects of baclofen or mirtazapine on cannabis smoking, craving and withdrawal. Exploratory study of the potential therapeutic value of baclofen and mirtazapine, but not a treatment intervention

Haney 2013

Laboratory study with aim of assessing effect of nabilone on marijuana withdrawal symptoms, and laboratory measure of relapse. The study aimed to test the therapeutic potential of nabilone but was not a treatment intervention

Haney 2013a

Laboratory study investigating the effect of zolpidem and nabilone (separately and in combination) compared with placebo on marijuana withdrawal symptoms and a model of marijuana relapse. The study aimed to test the therapeutic potential of zolpidem in marijuana smokers but was not a treatment intervention

Hart 2002

Laboratory study assessing the effect of oral THC or placebo on smoking of marijuana. Aim of study was to explore therapeutic potential of THC, but not a treatment intervention

Imbert 2014

Reports single case involving the use of baclofen to manage cannabis dependence. No treatment comparison

Levin 2008

Not a controlled study. Two case studies and a review of the use of dronabinol for cannabis dependence

McRae 2006

Open label study of buspirone for treatment of cannabis dependence. No treatment comparison

Robinson 2006

Randomised controlled trial comparing olanzapine and risperidone for treatment of schizophrenia in people with a history of cannabis use disorders. Primary goal of treatment was management of schizophrenia. Comparison of substance use outcomes was secondary. Data on substance use was reported only for those who completed treatment

Subodh 2011

An open label study investigating the use of baclofen for the treatment of cannabis dependence. No treatment comparison

Sugarman 2011

Controlled study assessing the safety of modafinil in combination with THC. While the study contributes to assessment of the therapeutic potential of modafinil, this study did not involve a treatment intervention. Participants were occasional cannabis users (people who were heavy users or dependent were excluded)

Tirado 2008

An open label study investigating the use / effect of atomoxetine for the treatment of marijuana dependence. No treatment comparison

Van Nimwegen 2008

Randomised controlled trial comparing olanzapine and risperidone for treatment of schizophrenia. Majority of participants were not using cannabis and cannabis dependence was not assessed

Vandrey 2011

Cross‐over study comparing zolpidem and placebo during short (3‐day) periods of abstinence from cannabis in terms of sleep parameters. Not a full treatment intervention for cannabis dependence

Vandrey 2013

Comparison of dronabinol and placebo in terms of effect on cannabis withdrawal and subjective effects of smoked cannabis, but without providing a treatment intervention for cannabis dependence

Winstock 2009

An open label study investigating the use of lithium carbonate for the management of cannabis withdrawal. No treatment comparison

Data and analyses

Open in table viewer
Comparison 1. Active medication versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number abstinent at end of treatment Show forest plot

5

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.1

Comparison 1 Active medication versus placebo, Outcome 1 Number abstinent at end of treatment.

Comparison 1 Active medication versus placebo, Outcome 1 Number abstinent at end of treatment.

1.1 THC preparations

1

156

Risk Ratio (M‐H, Random, 95% CI)

1.14 [0.56, 2.30]

1.2 Mixed action antidepressants

2

179

Risk Ratio (M‐H, Random, 95% CI)

0.82 [0.12, 5.41]

1.3 SSRI antidepressants

1

52

Risk Ratio (M‐H, Random, 95% CI)

2.33 [0.68, 8.05]

1.4 Anticonvulsant and mood stabiliser

1

19

Risk Ratio (M‐H, Random, 95% CI)

1.08 [0.50, 2.34]

1.5 Buspirone

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

1.6 Atomoxetine

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

1.7 N‐acetylcysteine

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2 Number experiencing adverse effects Show forest plot

5

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.2

Comparison 1 Active medication versus placebo, Outcome 2 Number experiencing adverse effects.

Comparison 1 Active medication versus placebo, Outcome 2 Number experiencing adverse effects.

2.1 THC preparations

1

156

Risk Ratio (M‐H, Random, 95% CI)

1.15 [0.90, 1.46]

2.2 Mixed action antidepressants

1

76

Risk Ratio (M‐H, Random, 95% CI)

0.93 [0.55, 1.55]

2.3 SSRI antidepressants

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.4 Anticonvulsant and mood stabiliser

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.5 Buspirone

1

50

Risk Ratio (M‐H, Random, 95% CI)

1.23 [0.99, 1.53]

2.6 Atomoxetine

1

38

Risk Ratio (M‐H, Random, 95% CI)

1.18 [0.95, 1.46]

2.7 N‐acetylcysteine

1

116

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.59, 1.34]

3 Number withdrawn due to adverse effects Show forest plot

7

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.3

Comparison 1 Active medication versus placebo, Outcome 3 Number withdrawn due to adverse effects.

Comparison 1 Active medication versus placebo, Outcome 3 Number withdrawn due to adverse effects.

3.1 THC preparations

1

156

Risk Ratio (M‐H, Random, 95% CI)

0.97 [0.06, 15.31]

3.2 Mixed action antidepressants

2

179

Risk Ratio (M‐H, Random, 95% CI)

1.44 [0.11, 18.90]

3.3 SSRI antidepressants

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.4 Anticonvulsant and mood stabiliser

1

50

Risk Ratio (M‐H, Random, 95% CI)

1.0 [0.07, 15.12]

3.5 Buspirone

1

50

Risk Ratio (M‐H, Random, 95% CI)

1.17 [0.08, 17.74]

3.6 Atomoxetine

1

38

Risk Ratio (M‐H, Random, 95% CI)

3.0 [0.13, 69.31]

3.7 N‐acetylcysteine

1

116

Risk Ratio (M‐H, Random, 95% CI)

3.0 [0.12, 72.15]

4 Completion of treatment Show forest plot

12

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.4

Comparison 1 Active medication versus placebo, Outcome 4 Completion of treatment.

Comparison 1 Active medication versus placebo, Outcome 4 Completion of treatment.

4.1 THC preparations

2

207

Risk Ratio (M‐H, Random, 95% CI)

1.29 [1.08, 1.55]

4.2 Mixed action antidepressants

2

169

Risk Ratio (M‐H, Random, 95% CI)

0.93 [0.71, 1.21]

4.3 SSRI antidepressants

2

122

Risk Ratio (M‐H, Random, 95% CI)

0.82 [0.44, 1.53]

4.4 Anticonvulsant and mood stabiliser

2

75

Risk Ratio (M‐H, Random, 95% CI)

0.78 [0.42, 1.46]

4.5 Buspirone

1

50

Risk Ratio (M‐H, Random, 95% CI)

0.99 [0.56, 1.77]

4.6 Atomoxetine

1

38

Risk Ratio (M‐H, Random, 95% CI)

1.29 [0.60, 2.74]

4.7 N‐acetylcysteine

1

116

Risk Ratio (M‐H, Random, 95% CI)

1.12 [0.83, 1.51]

4.8 Bupropion

2

92

Risk Ratio (M‐H, Random, 95% CI)

1.06 [0.67, 1.67]

Study flow diagram.
Figuras y tablas -
Figure 1

Study flow diagram.

Methodological quality summary: review authors' judgements about each methodological quality item for each included study.
Figuras y tablas -
Figure 2

Methodological quality summary: review authors' judgements about each methodological quality item for each included study.

Methodological quality graph: review authors' judgements about each methodological quality item presented as percentages across all included studies.
Figuras y tablas -
Figure 3

Methodological quality graph: review authors' judgements about each methodological quality item presented as percentages across all included studies.

Comparison 1 Active medication versus placebo, Outcome 1 Number abstinent at end of treatment.
Figuras y tablas -
Analysis 1.1

Comparison 1 Active medication versus placebo, Outcome 1 Number abstinent at end of treatment.

Comparison 1 Active medication versus placebo, Outcome 2 Number experiencing adverse effects.
Figuras y tablas -
Analysis 1.2

Comparison 1 Active medication versus placebo, Outcome 2 Number experiencing adverse effects.

Comparison 1 Active medication versus placebo, Outcome 3 Number withdrawn due to adverse effects.
Figuras y tablas -
Analysis 1.3

Comparison 1 Active medication versus placebo, Outcome 3 Number withdrawn due to adverse effects.

Comparison 1 Active medication versus placebo, Outcome 4 Completion of treatment.
Figuras y tablas -
Analysis 1.4

Comparison 1 Active medication versus placebo, Outcome 4 Completion of treatment.

Summary of findings for the main comparison. Abstinence at end of treatment

Active medication compared with placebo

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

Active Medication

Number abstinent at end of treatment ‐ mixed action antidepressants

Study population

RR 0.82
(0.12 to 5.41)

179
(2 studies)

⊕⊝⊝⊝
very low1,2

250 per 1000

205 per 1000
(30 to 1000)

Moderate

233 per 1000

191 per 1000
(28 to 1000)

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: Confidence interval; RR: Risk ratio

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

1 Significant heterogeneity between studies
2 Studies small (< 300 participants in total)

Figuras y tablas -
Summary of findings for the main comparison. Abstinence at end of treatment
Summary of findings 2. Withdrawal due to adverse effects

Active medication compared with placebo

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

Active Medication

Number withdrawn due to adverse effects ‐ mixed action antidepressants

Study population

RR 1.44
(0.11 to 18.9)

179
(2 studies)

⊕⊝⊝⊝
very low1,2

11 per 1000

16 per 1000
(1 to 205)

Moderate

13 per 1000

19 per 1000
(1 to 246)

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: Confidence interval; RR: Risk ratio

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

1 Studies differ in direction of effect without significant heterogeneity
2 Very few events and small group sizes

Figuras y tablas -
Summary of findings 2. Withdrawal due to adverse effects
Summary of findings 3. Completion of treatment

Active medication compared with placebo

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

Active Medication

Completion of treatment ‐ THC preparations

Study population

RR 1.29
(1.08 to 1.55)

207
(2 studies)

⊕⊕⊕⊝
moderate2

614 per 1000

792 per 1000
(663 to 951)

Moderate

618 per 1000

797 per 1000
(667 to 958)

Completion of treatment ‐ mixed action antidepressants

Study population

RR 0.93
(0.71 to 1.21)

169
(2 studies)

⊕⊕⊕⊝
moderate2

573 per 1000

533 per 1000
(407 to 694)

Moderate

551 per 1000

512 per 1000
(391 to 667)

Completion of treatment ‐ SSRI antidepressants

Study population

RR 0.82
(0.44 to 1.53)

122
(2 studies)

⊕⊝⊝⊝
very low1,2,3

790 per 1000

648 per 1000
(348 to 1000)

Moderate

766 per 1000

628 per 1000
(337 to 1000)

Completion of treatment ‐ anticonvulsant and mood stabiliser

Study population

RR 0.78
(0.42 to 1.46)

75
(2 studies)

⊕⊝⊝⊝
very low2,3

405 per 1000

316 per 1000
(170 to 592)

Moderate

387 per 1000

302 per 1000
(163 to 565)

Completion of treatment ‐ atypical antidepressant (bupropion)

Study population

RR 1.06
(0.67 to 1.67)

92
(2 studies)

⊕⊝⊝⊝
very low2,3

429 per 1000

454 per 1000
(287 to 716)

Moderate

400 per 1000

424 per 1000
(268 to 668)

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: Confidence interval; RR: Risk ratio

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

1 Significant heterogeneity between studies
2 Studies small (< 300 participants in total)
3 One study at risk of attrition bias

Figuras y tablas -
Summary of findings 3. Completion of treatment
Comparison 1. Active medication versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number abstinent at end of treatment Show forest plot

5

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 THC preparations

1

156

Risk Ratio (M‐H, Random, 95% CI)

1.14 [0.56, 2.30]

1.2 Mixed action antidepressants

2

179

Risk Ratio (M‐H, Random, 95% CI)

0.82 [0.12, 5.41]

1.3 SSRI antidepressants

1

52

Risk Ratio (M‐H, Random, 95% CI)

2.33 [0.68, 8.05]

1.4 Anticonvulsant and mood stabiliser

1

19

Risk Ratio (M‐H, Random, 95% CI)

1.08 [0.50, 2.34]

1.5 Buspirone

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

1.6 Atomoxetine

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

1.7 N‐acetylcysteine

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2 Number experiencing adverse effects Show forest plot

5

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 THC preparations

1

156

Risk Ratio (M‐H, Random, 95% CI)

1.15 [0.90, 1.46]

2.2 Mixed action antidepressants

1

76

Risk Ratio (M‐H, Random, 95% CI)

0.93 [0.55, 1.55]

2.3 SSRI antidepressants

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.4 Anticonvulsant and mood stabiliser

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.5 Buspirone

1

50

Risk Ratio (M‐H, Random, 95% CI)

1.23 [0.99, 1.53]

2.6 Atomoxetine

1

38

Risk Ratio (M‐H, Random, 95% CI)

1.18 [0.95, 1.46]

2.7 N‐acetylcysteine

1

116

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.59, 1.34]

3 Number withdrawn due to adverse effects Show forest plot

7

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

3.1 THC preparations

1

156

Risk Ratio (M‐H, Random, 95% CI)

0.97 [0.06, 15.31]

3.2 Mixed action antidepressants

2

179

Risk Ratio (M‐H, Random, 95% CI)

1.44 [0.11, 18.90]

3.3 SSRI antidepressants

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.4 Anticonvulsant and mood stabiliser

1

50

Risk Ratio (M‐H, Random, 95% CI)

1.0 [0.07, 15.12]

3.5 Buspirone

1

50

Risk Ratio (M‐H, Random, 95% CI)

1.17 [0.08, 17.74]

3.6 Atomoxetine

1

38

Risk Ratio (M‐H, Random, 95% CI)

3.0 [0.13, 69.31]

3.7 N‐acetylcysteine

1

116

Risk Ratio (M‐H, Random, 95% CI)

3.0 [0.12, 72.15]

4 Completion of treatment Show forest plot

12

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

4.1 THC preparations

2

207

Risk Ratio (M‐H, Random, 95% CI)

1.29 [1.08, 1.55]

4.2 Mixed action antidepressants

2

169

Risk Ratio (M‐H, Random, 95% CI)

0.93 [0.71, 1.21]

4.3 SSRI antidepressants

2

122

Risk Ratio (M‐H, Random, 95% CI)

0.82 [0.44, 1.53]

4.4 Anticonvulsant and mood stabiliser

2

75

Risk Ratio (M‐H, Random, 95% CI)

0.78 [0.42, 1.46]

4.5 Buspirone

1

50

Risk Ratio (M‐H, Random, 95% CI)

0.99 [0.56, 1.77]

4.6 Atomoxetine

1

38

Risk Ratio (M‐H, Random, 95% CI)

1.29 [0.60, 2.74]

4.7 N‐acetylcysteine

1

116

Risk Ratio (M‐H, Random, 95% CI)

1.12 [0.83, 1.51]

4.8 Bupropion

2

92

Risk Ratio (M‐H, Random, 95% CI)

1.06 [0.67, 1.67]

Figuras y tablas -
Comparison 1. Active medication versus placebo