Scolaris Content Display Scolaris Content Display

Antidepresivos para el tratamiento de los pacientes con depresión y dependencia del alcoholismo concomitantes

Contraer todo Desplegar todo

Referencias

Referencias de los estudios incluidos en esta revisión

Adamson 2015 {published data only}

Adamson SJ, Sellman JD, Foulds JA, Frampton CM, Deering D, Dunn A, et al. A randomized trial of combined citalopram and naltrexone for nonabstinent outpatients with co‐occurring alcohol dependence and major depression. Journal of Clinical Psychopharmacology 2015;35(2):143‐9. CENTRAL
Foulds JA, Douglas Sellman J, Adamson SJ, Boden JM, Mulder RT, Joyce PR. Depression outcome in alcohol dependent patients: an evaluation of the role of independent and substance‐induced depression and other predictors. Journal of Affective Disorders 2015;174:503‐10. CENTRAL
Foulds JA, Ton K, Kennedy MA, Adamson SJ, Mulder RT, Sellman JD. OPRM1 genotype and naltrexone response in depressed alcohol‐dependent patients. Pharmacogenetics and Genomics 2015;25(5):270‐3. CENTRAL

Altamura 1990 {published data only}

Altamura AC, Mauri MC, Girardi T, Panetta B. Alcoholism and depression: a placebo controlled study with viloxazine. International Journal of Clinical Pharmacology Research 1990;10(5):293‐8. CENTRAL

Altintoprak 2008 {published data only}

Altintoprak AE, Zorlu N, Coskunol H, Akdeniz F, Kitapcioglu G. Effectiveness and tolerability of mirtazapine and amitriptyline in alcoholic patients with co‐morbid depressive disorder: a randomized, double‐blind study. Human Psychopharmacology 2008;23(4):313‐9. CENTRAL

Butterworth 1971a {published data only}

Butterworth AT, Watts RD. Treatment of hospitalized alcoholics with doxepin and diazepam. A controlled study. Quarterly Journal of Studies on Alcohol 1971;32(1):78‐81. CENTRAL

Butterworth 1971b {published data only}

Butterworth AT. Depression associated with alcohol withdrawal. Imipramine therapy compared with placebo. Quarterly Journal of Studies on Alcohol 1971;32(2):343‐8. CENTRAL

Cocchi 1997 {published data only}

Cocchi R. Paroxetine vs amitryptiline in depressed alcoholics. European Neuropsychopharmacology 1997;7(Suppl 2):S254. CENTRAL

Cornelius 1997 {published data only}

Cornelius JC. Fluoxetine versus placebo in depressed alcoholics. Psychopharmacology. Psychopharmacology Bulletin 1994;30(4):661. CENTRAL
Cornelius JR, Salloum IM, Cornelius MD, Perel JM, Ehler JG, Jarrett PJ, et al. Preliminary report: double‐blind, placebo‐controlled study of fluoxetine in depressed alcoholics. Psychopharmacology Bulletin 1995;31(2):297‐303. CENTRAL
Cornelius JR, Salloum IM, Ehler JG, Jarrett PJ, Cornelius MD, Black A, et al. Double‐blind fluoxetine in depressed alcoholic smokers. Psychopharmacology Bulletin 1997;33(1):165‐70. CENTRAL
Cornelius JR, Salloum IM, Ehler JG, Jarrett PJ, Cornelius MD, Perel JM, et al. Fluoxetine in depressed alcoholics. A double‐blind, placebo‐controlled trial. Archives of General Psychiatry 1997;54(8):700‐5. CENTRAL
Cornelius JR, Salloum IM, Haskett RF, Daley DC, Cornelius MD, Thase ME, et al. Fluoxetine versus placebo in depressed alcoholics: a 1‐year follow‐up study. Addictive Behaviors 2000;25(2):307‐10. CENTRAL
Cornelius JR, Salloum IM, Thase ME, Haskett RF, Daley DC, Jones‐Barlock A, et al. Fluoxetine versus placebo in depressed alcoholic cocaine abusers. Psychopharmacology Bulletin 1998;34(1):117‐21. CENTRAL

Cornelius 2016 {published data only}

Cornelius J, Chung T, Douaihy A, Glance J. Mirtazapine pilot trial in youthful MDD/AUD subjects. American Journal on Addictions / American Academy of Psychiatrists in Alcoholism and Addictions 2017;26(3):250‐1. CENTRAL
Cornelius J, Chung T, Douaihy A, Glance J, Kmiec J, Wesesky M, et al. Double‐blind mirtazapine pilot trial in AUD/MDD. Alcoholism, Clinical and Experimental Research 2016;40:233. CENTRAL
Cornelius JR, Chung T, Douaihy AB, Kirisci L, Glance J, Kmiec J, et al. Mirtazapine in comorbid major depression and an alcohol use disorder: a double‐blind placebo‐controlled pilot trial. Psychiatry Research 2016;242:326‐30. [DOI: 10.1016/j.psychres.2016.06.005]CENTRAL
Cornelius JR, Chung TA, Wesesky MA, Douaihy A, Fitzgerald D, Kirisci L, et al. Combined young and older adult mirtazapine pilot trials in AUD/MDD. Alcoholism: Clinical and Experimental Research 2017;41:176A. CENTRAL

Gallant 1969 arm a {published data only}

Gallant DM, Bishop MP, Guerrero‐Figueroa R, Selby M, Phillips R. Doxepin versus diazepam: a controlled evaluation in 100 chronic alcoholic patients. Journal of Clinical Pharmacology and New Drugs 1969;9(1):57‐65. CENTRAL

Gallant 1969 arm b {published data only}

Gallant DM, Bishop MP, Guerrero‐Figueroa R, Selby M, Phillips R. Doxepin versus diazepam: a controlled evaluation in 100 chronic alcoholic patients. Journal of Clinical Pharmacology and New Drugs 1969;9(1):57‐65. CENTRAL

Gual 2003 {published data only}

Gual A, Balcells M, Torres M, Madrigal M, Diez T, Serrano L. Sertraline for the prevention of relapse in detoxicated alcohol dependent patients with a comorbid depressive disorder: a randomized controlled trial. Alcohol and Alcoholism (Oxford, Oxfordshire) 2003;38(6):619‐25. CENTRAL

Habrat 2006 {published data only}

Habrat B, Zaloga B. A double‐blind controlled study of the efficacy and acceptability of tianeptine in comparison with fluvoxamine in the treatment of depressed alcoholic patients. Psychiatria Polska 2006;40(3):579‐97. CENTRAL

Hernandez‐Avila 2004 {published data only}

Hernandez‐Avila CA, Modesto‐Lowe V, Feinn R, Kranzler HR. Nefazodone treatment of comorbid alcohol dependence and major depression. Alcoholism, Clinical and Experimental Research 2004;28(3):433‐40. CENTRAL

Kranzler 2006 arm A {published data only}

Kranzler HR, Mueller T, Cornelius J, Pettinati HM, Moak D, Martin PR, et al. Sertraline treatment of co‐occurring alcohol dependence and major depression. Journal of Clinical Psychopharmacology 2006;26(1):13‐20. CENTRAL

Kranzler 2006 arm B {published data only}

Kranzler HR, Mueller T, Cornelius J, Pettinati HM, Moak D, Martin PR, et al. Sertraline treatment of co‐occurring alcohol dependence and major depression. Journal of Clinical Psychopharmacology 2006;26(1):13‐20. CENTRAL

Krupitsky 1993 arm A {published data only}

Krupitsky EM, Burakov AM, Grinenko AIa, Borodkin IuS. Effect of pharmacotherapy of affective disorders on the psycho‐semantics of alcoholic patients. Zhurnal Nevrologii i Psikhiatrii Imeni S.S. Korsakova 1995;95(6):67‐71. CENTRAL
Krupitsky EM, Burakov AM, Ivanov VB, Karandashova GF, Lapin IP, Grinenko Ala, et al. The use of baclofen for treating affective disorders in alcoholism. Zhurnal Nevrologii i Psikhiatrii Imeni S.S. Korsakova 1994;94(1):57‐61. CENTRAL
Krupitsky EM, Burakov AM, Ivanov VB, Krandashova GF, Lapin IP, Grinenko AJa, et al. Baclofen administration for the treatment of affective disorders in alcoholic patients. Drug and Alcohol Dependence 1993;33(2):157‐63. CENTRAL

Krupitsky 1993 arm B {published data only}

Krupitsky EM, Burakov AM, Grinenko AIa, Borodkin IuS. Effect of pharmacotherapy of affective disorders on the psycho‐semantics of alcoholic patients. Zhurnal Nevrologii i Psikhiatrii Imeni S.S. Korsakova 1995;95(6):67‐71. CENTRAL
Krupitsky EM, Burakov AM, Ivanov VB, Karandashova GF, Lapin IP, Grinenko AIa, et al. The use of baclofen for treating affective disorders in alcoholism. Zhurnal Nevrologii i Psikhiatrii Imeni S.S. Korsakova 1994;94(1):57‐61. CENTRAL
Krupitsky EM, Burakov AM, Ivanov VB, Krandashova GF, Lapin IP, Grinenko AJa, et al. Baclofen administration for the treatment of affective disorders in alcoholic patients. Drug and Alcohol Dependence 1993;33(2):157‐63. CENTRAL

Krupitsky 2012 {published data only}

Krupitsky E, Yerish S, Kiselev A. Clinical trial of escitalopram for alcoholism comorbid with affective disorders. Alcoholism, Clinical and Experimental Research 2012;36:297A. CENTRAL
Krupitsky E, Yerish S, Kiselev A. Clinical trial of escitalopram for alcoholism comorbid with affective disorders. European Neuropsychopharmacology 2010;20(Suppl 3):S571. CENTRAL
Krupitsky EM, Yerish SM, Berntsev VA, Kiselev A, Alexandrovsky NA, Torban MN, et al. Double blind placebo controlled randomized clinical trial of escitalopram for alcoholism comorbid with affective disorders (depression and anxiety). ISBRA 2010 Poster Abstracts. 2010:167A. CENTRAL
Krupitsky EM, Yerish SM, Kiselev AS, Berntsev VA, Alexandrovsky NA, Torban MN, et al. A double blind, placebo controlled, randomized clinical trial of escitalopram for the treatment of affective disorders in alcohol dependent patients in early remission. The International Psychiatry and Behavioral Neurosciences Yearbook. Boutros N. Vol. II, New York (NY): Nova Science Publishers, 2012:239‐56. CENTRAL

Liappas 2005 arm A {published data only}

Liappas J, Paparrigopoulos T, Tzavellas E, Rabavilas A. Mirtazapine and venlafaxine in the management of collateral psychopathology during alcohol detoxification. Progress in Neuro‐psychopharmacology & Biological Psychiatry 2005;29(1):55‐60. CENTRAL

Liappas 2005 arm B {published data only}

Liappas J, Paparrigopoulos T, Tzavellas E, Rabavilas A. Mirtazapine and venlafaxine in the management of collateral psychopathology during alcohol detoxification. Progress in Neuro‐psychopharmacology & Biological Psychiatry 2005;29(1):55‐60. CENTRAL

Liappas 2005 arm C {published data only}

Liappas J, Paparrigopoulos T, Tzavellas E, Rabavilas A. Mirtazapine and venlafaxine in the management of collateral psychopathology during alcohol detoxification. Progress in Neuro‐psychopharmacology & Biological Psychiatry 2005;29(1):55‐60. CENTRAL

Lôo 1988 {published data only}

Lôo H, Malka R, Defrance R, Barrucand D, Benard JY, Niox‐Rivière H, et al. Tianeptine and amitriptyline. Controlled double‐blind trial in depressed alcoholic patients. Neuropsychobiology 1988;19(2):79‐85. CENTRAL

Mason 1996 {published data only}

Mason BJ, Kocsis JH. Desipramine treatment of alcoholism. Psychopharmacology Bulletin 1991;27(2):155‐61. CENTRAL
Mason BJ, Kocsis JH, Ritvo EC, Cutler RB. A double‐blind, placebo‐controlled trial of desipramine for primary alcohol dependence stratified on the presence or absence of major depression. JAMA 1996;275(10):761‐7. CENTRAL

McGrath 1996 {published data only}

McGrath PJ, Nunes EV, Stewart JW, Goldman D, Agosti V, Ocepek‐Welikson K, et al. Imipramine treatment of alcoholics with primary depression: a placebo‐controlled clinical trial. Archives of General Psychiatry 1996;53(3):232‐40. CENTRAL
McGrath PJ, Nunes EV, Stewart JW, Ocepek‐Welikson K, Quitkin FM. Antidepressant treatment of primary depression in alcoholics: a placebo‐controlled randomized clinical trial. Psychopharmacology Bulletin. 1995:598. CENTRAL

McLean 1986 {published data only}

McLean PC, Ancill RJ, Szulecka TK. Mianserin in the treatment of depressive symptoms in alcoholics. A double‐blind placebo controlled study using a computer delivered self‐rating scale. Psychiatria Polska 1986;20(6):417‐27. CENTRAL

Moak 2003 {published data only}

Moak DH, Anton RF, Latham PK, Voronin KE, Waid RL, Durazo‐Arvizu R. Sertraline and cognitive behavioral therapy for depressed alcoholics: results of a placebo‐controlled trial. Journal of Clinical Psychopharmacology 2003;23(6):553‐62. CENTRAL

Muhonen 2008 {published data only}

Muhonen LH, Lahti J, Alho H, Lönnqvist J, Haukka J, Saarikoski ST. Serotonin transporter polymorphism as a predictor for escitalopram treatment of major depressive disorder comorbid with alcohol dependence. Psychiatry Research 2011;186(1):53‐7. CENTRAL
Muhonen LH, Lahti J, Sinclair D, Lönnqvist J, Alho H. Treatment of alcohol dependence in patients with co‐morbid major depressive disorder ‐ predictors for the outcomes with memantine and escitalopram medication. Substance Abuse Treatment, Prevention, and Policy 2008;3:20. CENTRAL
Muhonen LH, Lönnqvist J, Juva K, Alho H. Double‐blind, randomized comparison of memantine and escitalopram for the treatment of major depressive disorder comorbid with alcohol dependence. Journal of Clinical Psychiatry 2008;69(3):392‐9. CENTRAL
Muhonen LH, Lönnqvist J, Lahti J, Alho H. Age at onset of first depressive episode as a predictor for escitalopram treatment of major depression comorbid with alcohol dependence. Psychiatry Research 2009;167(1‐2):115‐22. CENTRAL

Nunes 1993 {published data only}

Nunes EV, McGrath PJ, Quitkin FM, Stewart JP, Harrison W, Tricamo E, et al. Imipramine treatment of alcoholism with comorbid depression. American Journal of Psychiatry 1993;150(6):963‐5. CENTRAL

Pettinati 2001a {published data only}

Dundon W, Lynch KG, Pettinati HM, Lipkin C. Treatment outcomes in type A and B alcohol dependence 6 months after serotonergic pharmacotherapy. Alcoholism, Clinical and Experimental Research 2004;28(7):1065‐73. CENTRAL
Levin SC. The efficacy of sertraline treatment for subtypes of alcohol dependence: advancing individualized treatments. Dissertation Abstracts International 2010;71(4‐B2):2333. CENTRAL
Pettinati HM, Dundon W, Lipkin C. Gender differences in response to sertraline pharmacotherapy in Type A alcohol dependence. American Journal on Addictions / American Academy of Psychiatrists in Alcoholism and Addictions 2004;13(3):236‐47. CENTRAL
Pettinati HM, Volpicelli JR, Kranzler HR, Luck G, Rukstalis MR, Cnaan A. Sertraline treatment for alcohol dependence: interactive effects of medication and alcoholic subtype. Alcoholism, Clinical and Experimental Research 2000;24(7):1041‐9. CENTRAL
Pettinati HM, Volpicelli JR, Luck G, Kranzler HR, Rukstalis MR, Cnaan A. Double‐blind clinical trial of sertraline treatment for alcohol dependence. Journal of Clinical Psychopharmacology 2001;21(2):143‐53. CENTRAL

Pettinati 2010 arm A {published data only}

NCT00004554. Sertraline for alcohol dependence and depression. clinicaltrials.gov/ct2/show/NCT00004554 Date first received: 7 February 2000. CENTRAL
Pettinati HM, Leshner AR, Dundon WD. Sertraline treatment in subtypes of alcohol dependence: a replication. Alcoholism: Clinical and Experimental Research 2012;36:245A. CENTRAL
Pettinati HM, Oslin DW, Kampman KM, Dundon WD, Xie H, Gallis TL, et al. A double‐blind, placebo‐controlled trial combining sertraline and naltrexone for treating co‐occurring depression and alcohol dependence. American Journal of Psychiatry 2010;167(6):668‐75. CENTRAL
Roth TL, Mahoney EM, Dundon WD, Pettinati HM. Combination pharmacotherapy sertraline and naltrexone decreases suicidal ideation scores in co‐morbid depressed alcoholics. Alcoholism: Clinical and Experimental Research 2012;36:245A. CENTRAL

Pettinati 2010 arm B {published data only}

NCT00004554. Sertraline for alcohol dependence and depression. clinicaltrials.gov/ct2/show/NCT00004554 Date first received: 7 February 2000. CENTRAL
Pettinati HM, Leshner AR, Dundon WD. Sertraline treatment in subtypes of alcohol dependence: a replication. Alcoholism: Clinical and Experimental Research 2012;36:245A. CENTRAL
Pettinati HM, Oslin DW, Kampman KM, Dundon WD, Xie H, Gallis TL, et al. A double‐blind, placebo‐controlled trial combining sertraline and naltrexone for treating co‐occurring depression and alcohol dependence. American Journal of Psychiatry 210;167(6):668‐75. CENTRAL
Roth TL, Mahoney EM, Dundon WD, Pettinati HM. Combination pharmacotherapy sertraline and naltrexone decreases suicidal ideation scores in co‐morbid depressed alcoholics. Alcoholism: Clinical and Experimental Research 2012;36:245A. CENTRAL

Roy 1998 {published data only}

Roy A. Placebo‐controlled study of sertraline in depressed recently abstinent alcoholics. Biological Psychiatry 1998;44(7):633‐7. CENTRAL
Roy A. Treatment of depressed alcoholics. 150th Annual Meeting of the American Psychiatric Association; 1997 May 17‐22; San Diego (CA). 1997:151. CENTRAL

Roy‐Byrne 2000 {published data only}

Roy‐Byrne PP, Pages KP, Russo JE, Jaffe C, Blume AW, Kingsley E, et al. Nefazodone treatment of major depression in alcohol‐dependent patients: a double‐blind, placebo‐controlled trial. Journal of Clinical Psychopharmacology 2000;20(2):129‐36. CENTRAL

Referencias de los estudios excluidos de esta revisión

Anthenelli 2014 {published data only}

Anthenelli RM, Blom TJ, Heffner JL, Higley AE, Bekman NM, Doran N, et al. Central serotonergic & peripheral mechanisms underlie sex‐sensitive, stressor‐specific endocrine responses in alcohol dependent subjects and controls. Alcoholism: Clinical and Experimental Research 2014;38:85A. CENTRAL

Arnow 2015 {published data only}

Arnow BA, Blasey C, Williams LM, Palmer DM, Rekshan W, Schatzberg AF, et al. Depression subtypes in predicting antidepressant response: a report from the iSPOT‐D trial. American Journal of Psychiatry 2015;172(8):743‐50. CENTRAL

Balaratnasingam 2011 {published data only}

Balaratnasingam S, Janca A. Combining sertraline and naltrexone in the treatment of adults with comorbid depression and alcohol dependence. Current Psychiatry Reports 2011;13(4):245‐7. CENTRAL

Bandati 2013 {published data only}

Bandati A, Bandati AO, Albu A. Management of affective disorders in alcohol‐dependent patients. European Neuropsychopharmacology 2013;23:S564‐5. CENTRAL

Batki 2015 {published data only}

Batki S, Pennington D, Meyerhoff D, Spigelman I, Back S, Petrakis I. Pharmacologic therapies for alcohol use disorder and PTSD. Alcoholism, Clinical and Experimental Research 2015;39:289A. CENTRAL

Bowman 1966 {published data only}

Bowman EH, Thimann J. Treatment of alcoholism in the subacute stage. (A study of three active agents). Diseases of the Nervous System 1966;27(5):342‐6. CENTRAL

Brewer 2015 {published data only}

Brewer A, Thompson‐Lake DG, Mahoney JJ, Newton TF, De La Garza R. Evaluation of lisdexamfetamine alone and lisdexamfetamine + modafinil for cocaine use disorder. Drug and Alcohol Dependence 2015;146:e231. CENTRAL

Brown 2003 {published data only}

Brown ES, Bobadilla L, Nejtek VA, Perantie D, Dhillon H, Frol A. Open‐label nefazodone in patients with a major depressive episode and alcohol dependence. Progress in Neuro‐psychopharmacology & Biological Psychiatry 2003;27(4):681‐5. CENTRAL

Brunelin 2014 {published data only}

Brunelin J, Jalenques I, Trojak B, Attal J, Szekely D, Gay A, et al. The efficacy and safety of low frequency repetitive transcranial magnetic stimulation for treatment‐resistant depression: the results from a large multicenter French RCT. Brain Stimulation 2014;7(6):855‐63. CENTRAL

Charney 2015 {published data only}

Charney DA, Heath LM, Zikos E, Palacios‐Boix J, Gill KJ. Poorer drinking outcomes with citalopram treatment for alcohol dependence: a randomized, double‐blind, placebo‐controlled trial. Alcoholism, Clinical and Experimental Research 2015;39(9):1756‐65. CENTRAL

Charnoff 1967 {published data only}

Charnoff SM. Long‐term treatment of alcoholism with amitriptyline and emylcamate. A double‐blind evaluation. Quarterly Journal of Studies on Alcohol 1967;28(2):289‐94. CENTRAL

Chick 2004b {published data only}

Chick J, Aschauer H, Hornik K, Investigators' Group. Efficacy of fluvoxamine in preventing relapse in alcohol dependence: a one‐year, double‐blind, placebo‐controlled multicentre study with analysis by typology. Drug and Alcohol Dependence 2004;74(1):61‐70. CENTRAL

Clark 2003 {published data only}

Clark DB, Wood DS, Cornelius JR, Bukstein OG, Martin CS. Clinical practices in the pharmacological treatment of comorbid psychopathology in adolescents with alcohol use disorders. Journal of Substance Abuse Treatment 2003;25(4):293‐5. CENTRAL

Cornelius 1993 {published data only}

Cornelius JR, Salloum IM, Cornelius MD, Perel JM, Thase ME, Ehler JG, et al. Fluoxetine trial in suicidal depressed alcoholics. Psychopharmacology Bulletin 1993;29(2):195‐9. CENTRAL

Cornelius 2011 {published data only}

Cornelius JR, Douaihy A, Bukstein OG, Daley DC, Wood SD, Kelly TM, et al. Evaluation of cognitive behavioral therapy/motivational enhancement therapy (CBT/MET) in a treatment trial of comorbid MDD/AUD adolescents. Addictive Behaviors 2011;36(8):843‐8. CENTRAL

Cornelius 2012 {published data only}

Cornelius JR, Douaihy A, Clark DB. Mirtazapine pilot trial in depressed alcoholics. American Journal on Addictions / American Academy of Psychiatrists in Alcoholism and Addictions 2012;21(4):392. CENTRAL
Cornelius JR, Douaihy AB, Clark DB, Chung T, Wood DS, Daley D. Mirtazapine in comorbid major depression and alcohol dependence: an open‐label trial. Journal of Dual Diagnosis 2012;8(3):200‐4. CENTRAL

Davis 2005 {published data only}

Davis LL, Rush JA, Wisniewski SR, Rice K, Cassano P, Jewell ME, et al. Substance use disorder comorbidity in major depressive disorder: an exploratory analysis of the Sequenced Treatment Alternatives to Relieve Depression cohort. Comprehensive Psychiatry 2005;46(2):81‐9. CENTRAL
Davis LL, Wisniewski SR, Howland RH, Trivedi MH, Husain MM, Fava M, et al. Does comorbid substance use disorder impair recovery from major depression with SSRI treatment? An analysis of the STAR*D level one treatment outcomes. Drugs and Alcohol Dependence 2010;107(2‐3):161‐70. CENTRAL

Desai 1999 {published data only}

Desai N, Losardo M, Farrell D, Penk W, Petrakis I, Rounsaville B. The efficacy of sertraline versus fluoxetine in Veterans comorbid depression and alcohol dependence. Abstract. 1999:45. CENTRAL

Douglas 1996 {published data only}

Douglas MB, Fleming MF. Treating depression in alcoholics. Journal of Family Practice 1996;42(6):565‐6. CENTRAL

Eriksson 2001 {published data only}

Eriksson M, Berggren U, Blennow K, Fahlke C, Balldin J. Further investigation of citalopram on alcohol consumption in heavy drinkers: responsiveness possibly linked to the DRD2 A2/A2 genotype. Alcohol 2001;24(1):15‐23. CENTRAL

Farren 1999 {published data only}

Farren CK, O'Malley SS. Occurrence and management of depression in the context of naltrexone treatment of alcoholism. American Journal of Psychiatry 1999;156(8):1258‐62. CENTRAL

Foulds 2016 {published data only}

Foulds JA, Sellman JD, Mulder RT. Antidepressant therapy for depressed patients with an alcohol use disorder. Australian and New Zealand Journal of Psychiatry 2016;50(3):199‐200. CENTRAL

García‐Portilla 2005 {published data only}

García‐Portilla MP, Bascarán MT, Saiz PA, Mateos M, González‐Quirós M, Pérez P, et al. Effectiveness of venlafaxine in the treatment of alcohol dependence with comorbid depression. Actas Espanolas de Psiquiatria 2005;33(1):41‐5. CENTRAL

Glasner‐Edwards 2007 {published data only}

Glasner‐Edwards S, Tate SR, McQuaid JR, Cummins K, Granholm E, Brown SA. Mechanisms of action in integrated cognitive‐behavioral treatment versus twelve‐step facilitation for substance‐dependent adults with comorbid major depression. Journal of Studies on Alcohol and Drugs 2007;68(5):663‐72. CENTRAL

Gorelick 1992 {published data only}

Gorelick DA, Paredes A. Effect of fluoxetine on alcohol consumption in male alcoholics. Alcoholism, Clinical and Experimental Research 1992;16(2):261‐5. CENTRAL

Grelotti 2014 {published data only}

Grelotti DJ, Hammer GP, Dilley JW, Karasic DH, Sorensen JL, Bangsberg DR, et al. Does substance use compromise depression treatment? A randomized trial of homeless HIV+ persons. Topics in Antiviral Medicine 2014;22(e‐1):541‐2. CENTRAL

Han 2013 {published data only}

Han DH, Kim SM, Choi JE, Min KJ, Renshaw PF. Adjunctive aripiprazole therapy with escitalopram in patients with co‐morbid major depressive disorder and alcohol dependence: clinical and neuroimaging evidence. Journal of Psychopharmacology (Oxford, England) 2013;27(3):282‐91. CENTRAL

Hautzinger 2005 {published data only}

Hautzinger M, Wetzel H, Szegedi A, Scheurich A, Lörch B, Singer P, et al. Combination treatment with SSRI and cognitive behavior therapy for relapse prevention of alcohol‐dependent men. Results of a randomized, controlled multicenter therapeutic study. Nervenarzt 2005;76(3):295‐307. CENTRAL

Ionescu 2011 {published data only}

Ionescu D, Dehelean C, Funar TS, Dumache R. A comparative evaluation of therapy escitalopram versus memantine in the case of patients with alcohol dependence and co‐morbid major depressive disorder. Toxicology Letters 2011;205:S89. CENTRAL

Ivanets 1998 {published data only}

Ivanets NN. Treatment of alcoholism with antidepressant mianserin. 5th World Congress on Innovations in Psychiatry; 1998 19‐22 May; London. 1998:S27. CENTRAL
Ivanets NN, Anokhina IP, Kogan BM, Chirko VV, Nebarakova TP, Rusinov AV. The efficacy and mechanisms of action of lerivon in alcoholism. Zhurnal Nevrologii i Psikhiatrii Imeni S.S. Korsakova 1996;96(5):52‐8. CENTRAL

Janiri 1996 {published data only}

Janiri L, Gobbi G, Mannelli P, Pozzi G, Serretti A, Tempesta E. Effects of fluoxetine at antidepressant doses on short‐term outcome of detoxified alcoholics. International Clinical Psychopharmacology 1996;11(2):109‐17. CENTRAL

Janiri 1997 {published data only}

Janiri L, Hadjichristos A, Lombardi U, Rago R, Mannelli P, Tempesta E. SSRIs in alcoholism: fluvoxamine vs fluoxetine in alcoholic outpatients. Sixth World Congress of Biological Psychiatry; 1997 Jun 22‐27; Nice. 1997:35S. CENTRAL

Kalyoncu 2007 {published data only}

Kalyoncu OA, Mirsal H, Pektas O, Tan D, Beyazyurek M. The efficacy of venlafaxine on depressive symptoms of patients diagnosed with both alcohol use disorder and major depressive disorder. Journal of Dependence 2007;8(2):59‐65. CENTRAL

Kranzler 1995 {published data only}

Kranzler HR, Burleson JA, Korner P, Del Boca FK, Bohn MJ, Brown J, et al. Placebo‐controlled trial of fluoxetine as an adjunct to relapse prevention in alcoholics. American Journal of Psychiatry 1995;152(3):391‐7. CENTRAL

Kranzler 2011 {published data only}

Kranzler HR, Armeli S, Tennen H. Post‐treatment outcomes in a double‐blind, randomized trial of sertraline for alcohol dependence. Alcoholism, Clinical and Experimental Research 2012;36(4):739‐44. CENTRAL
Kranzler HR, Armeli S, Tennen H, Covault J, Feinn R, Arias AJ, et al. A double‐blind, randomized trial of sertraline for alcohol dependence: moderation by age of onset [corrected] and 5‐hydroxytryptamine transporter‐linked promoter region genotype. Journal of Clinical Psychopharmacology 2011;31(1):22‐30. CENTRAL

Krupitsky 2013 {published data only}

Krupitsky EM, Yerish SM, Rybakova KV, Kiselev AS. Single blind placebo controlled randomized clinical trial of trazodon for alcoholism comorbid with affective disorders. Alcoholism, Clinical and Experimental Research 2013;37:18A. CENTRAL

Krystal 2008 {published data only}

Krystal JH, Gueorguieva R, Cramer J, Collins J, Rosenheck R, VA CSP No. 425 Study Team. Naltrexone is associated with reduced drinking by alcohol dependent patients receiving antidepressants for mood and anxiety symptoms: results from VA Cooperative Study No. 425, "Naltrexone in the treatment of alcoholism". Alcoholism, Clinical and Experimental Research 2008;32(1):85‐91. CENTRAL

Labbate 2004 {published data only}

Labbate LA, Sonne SC, Randal CL, Anton RF, Brady KT. Does comorbid anxiety or depression affect clinical outcomes in patients with post‐traumatic stress disorder and alcohol use disorders?. Comprehensive Psychiatry 2004;45(4):304‐10. CENTRAL

Lee 2012 {published data only}

Lee Y, Han D, Kim S, Park D, Na C. Augmentation therapy of aripiprazole with escitalopram in patients with co‐occurrence of depression and alcohol dependence. European Neuropsychopharmacology 2012;22:S389. CENTRAL

Liappas 2004 {published data only}

Liappas J, Paparrigopoulos T, Malitas P, Tzavellas E, Christodoulou G. Mirtazapine improves alcohol detoxification. Journal of Psychopharmacology 2004;18(1):88‐93. CENTRAL

Macher 1991 {published data only}

Macher JP, Minot R, Duval F, Luthringer R, Toussaint M, Schaltenbrand N. Neuro‐electrophysiologic studies in abstinent and depressed alcoholic patients treated with tianeptine. Presse Medicale 1991;20(37):1853‐7. CENTRAL

Malka 1992 {published data only}

Malka R, Loo H, Ganry H, Souche A, Marey C, Kamoun A. Long‐term administration of tianeptine in depressed patients after alcohol withdrawal. British Journal of Psychiatry 1992;15 Suppl:66‐71. CENTRAL

Marey 1991 {published data only}

Marey C, Ganry H, Delalleau B, Kamoun A. Therapeutic effects of tianeptine in patients with depression and anxiety disorders with or without associated alcoholism. Presse Medicale 1991;20(37):1828‐36. CENTRAL

Mason 1999 {published data only}

Mason BJ, Salvato FR, Cutler RB, Cooper TB, Suckow RF, Williams LD. Naltrexone augmentation of sertraline in depressed alcoholics. 152nd Annual Meeting of the American Psychiatric Association; 1999 May 15‐20; Washington (DC). 1999. CENTRAL
Mason BJ, Salvato FR, Williams LD, Cutler RB. Naltrexone and/or sertraline in outpatients with alcoholism and depression. 11th European College of Neuropsychopharmacology Congress; 1998 Oct 31‐Nov 4; Paris. 1999. CENTRAL

Naranjo 1997 {published data only}

Naranjo CA, Bremner KE, Bazoon M, Turksen IB. Using fuzzy logic to predict response to citalopram in alcohol dependence. Clinical Pharmacology and Therapeutics 1997;62(2):209‐24. CENTRAL

Oslin 2005 {published data only}

Gopalakrishnan R, Ross J, O'Brien C, Oslin D. Course of late‐life depression with alcoholism following combination therapy. Journal of Studies on Alcohol 2009;70(2):237‐41. CENTRAL
Oslin DW. Treatment of late‐life depression complicated by alcohol dependence. American Journal of Geriatric Psychiatry 2005;13(6):491‐500. CENTRAL

Overall 1973 {published data only}

Overall JE, Brown D, Williams JD, Neill LT. Drug treatment of anxiety and depression in detoxified alcoholic patients. Archives of General Psychiatry 1973;29(2):218‐25. CENTRAL

Powell 1995 {published data only}

Powell BJ, Campbell JL, Landon JF, Liskow BI, Thomas HM, Nickel EJ, et al. A double‐blind, placebo‐controlled study of nortriptyline and bromocriptine in male alcoholics subtyped by comorbid psychiatric disorders. Alcoholism, Clinical and Experimental Research 1995;19(2):462‐8. CENTRAL

Ruiz‐Mellott 2005 {published data only}

Ruiz‐Mellott K, Poland R, Wilkins J. 5‐HTTLPR, antidepressant response, and alcohol use disorder. 158th Annual Meeting of the American Psychiatric Association; 2005 May 21‐26; Atlanta, GA. 2005:28. CENTRAL

Saatcioglu 2005 {published data only}

Saatcioglu O, Evren C, Erim R, Cakmak D. Venlafaxine treatment in alcohol dependants with comorbid depression. European Neuropsychopharmacology 2005;Suppl 3:S589. CENTRAL

Salloum 2011 {published data only}

Salloum IM, Cornelius JR, Douaihy A, Kirisci L. Gender difference in alcohol treatment response in patients with comorbid alcoholism and depression. Alcoholism, Clinical and Experimental Research 2011;35:19A. CENTRAL

Salloum 2013 {published data only}

Salloum IM, Cornelius JR, Douaihy A, Daley DC. Predicting medication adherence and treatment completion among depressed alcoholics. Alcoholism, Clinical and Experimental Research 2013;37:198A. CENTRAL

Schottenfeld 1989 {published data only}

Schottenfeld RS, O'Malley SS, Smith L, Rounsaville BJ, Jaffe JH. Limitation and potential hazards of MAOI's for the treatment of depressive symptoms in abstinent alcoholics. American Journal of Drug and Alcohol Abuse 1989;15(3):339‐44. CENTRAL

Shaw 1975 {published data only}

Shaw JA, Donley P, Morgan DW, Robinson JA. Treatment of depression in alcoholics. American Journal of Psychiatry 1975;132(6):641‐4. CENTRAL

Witte 2012 {published data only}

Witte J, Bentley K, Evins AE, Clain AJ, Baer L, Pedrelli P, et al. A randomized, controlled, pilot study of acamprosate added to escitalopram in adults with major depressive disorder and alcohol use disorder. Journal of Clinical Pharmacology 2012;32(6):787‐96. CENTRAL

Referencias de los estudios en espera de evaluación

Petrakis 2013 {published data only}

Petrakis I, Jane JS, O'Brien E, Keegan K, Ralevski E, Desai N. Effect of depression on outcomes in a clinical trial evaluating antidepressants +/‐ naltrexone for treatment of alcohol dependence and PTSD. Alcoholism, Clinical and Experimental Research 2013;37:19A. CENTRAL
Petrakis IL, Ralevski E, Desai N, Trevisan L, Gueorguieva R, Rounsaville B, et al. Noradrenergic vs serotonergic antidepressant with or without naltrexone for veterans with PTSD and comorbid alcohol dependence. Neuropsychopharmacology 2012;37(4):996‐1004. CENTRAL

Schifano 1993 {published data only}

Schifano F, Garofoli F. Dothiepin vs. S‐adenosylmethionine (SAMe) for the treatment of major depression in weaned alcoholics: a pilot study. European Neuropsychopharmacology 1993;3(3):330. CENTRAL

Anton 1996

Anton RR, Moak DH, Latham PK. The obsessive compulsive drinking scale: a new method of assessing outcome in alcoholism treatment studies. Archives of General Psychiatry 1996;53:225‐31.

APA 1980

American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 3rd Edition. Washington (DC): American Psychiatric Association, 1980.

APA 1987

American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders (DSM III‐R). Washington (DC): American Psychiatric Press, 1987.

APA 1994

American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 4th Edition. Washington (DC): American Psychiatric Press, 1994.

APA 2000

American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, Text Revision (DSM‐IV‐TR). Washington (DC): American Psychiatric Association, 2000.

APA 2013

American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 5th Edition. Arlington (VA): American Psychiatric Association, 2013.

Artigas 2002

Artigas F, Nutt DJ, Shelton R. Mechanism of action of antidepressants. Psychopharmacology Bulletin 2002;36(Suppl 2):123‐32.

Babor 1992

Babor TF, Hofmann M, DelBoca FK, Hesselbrock V, Meyer RE, Dolinsky ZS, et al. Types of alcoholics, I. Evidence for an empirically derived typology based on indicators of vulnerability and severity. Archives of General Psychiatry 1992;49:599‐608.

Beck 1961

Beck AT, Ward CH, Mendelson M, Mock J, Erbaugh J. An inventory for measuring depression. Archives of General Psychiatry 1961;4:561‐71.

Beck 1988

Beck AT, Epstein N, Brown G, Steer RA. An inventory for measuring clinical anxiety: psychometric properties. Journal of Consulting and Clinical Psychology 1988;56:893‐7.

Beck 1996

Beck AT, Steer RA, Brown GK. Beck Depression Inventory: Manual. 2nd Edition. San Antonio (TX): Harcourt Brace, 1996.

Brown 1995

Brown SA, Inaba RK, Gillin JC, Schuckit MA, Stewart MA, Irwin MR. Alcoholism and affective disorder: clinical course of depressive symptoms. American Journal of Psychiatry 1995;152:45‐52.

Busner 2009

Busner J, Targum SD, Miller DS. The Clinical Global Impressions scale: errors in understanding and use. Comprehensive Psychiatry 2009;50(3):257‐62.

Buysse 1989

Buysse DJ, Reynolds CF, Monk TH, Berman SR, Kupfer DJ. The Pittsburgh Index: a new instrument for psychiatric practice and research. Psychiatry Research 1989;28:193‐213.

Carboni 2004

Carboni E, Silvagni A. Dopamine reuptake by norepinephrine neurons: exception or rule?. Critical Reviews in Neurobiology 2004;16(1‐2):121‐8.

Carr 1981

Carr AC, Ancill RJ, Ghosh A, Margo A. Direct assessment of depression by microcomputer. A feasibility study. Acta Psychiatrica Scandinavica 1981;64(5):415‐22.

Chick 2004a

Chick J, Aschauer H, Hornik K. Efficacy of fluvoxamine in preventing relapse in alcohol dependence: a one‐year, double blind, placebo‐controlled Multicentre study with analysis by typology. Drug and Alcohol Dependence 2004;74:61‐70.

Cloninger 1988

Cloninger CR, Sigvardsson S, Gilligan SB, von Knorring AL, Reich T, Bohman M. Genetic heterogeneity and the classification of alcoholism. Advances in Alcohol and Substance Abuse 1988;7:3‐16.

Derogatis 1974

Derogatis LR, Lipman RS, Rickels K, Uhlenhuth EH, Covi L. The Hopkins Symptom Checklist (HSCL): a self‐report symptom inventory. Behavioral Science 1974;19(1):1‐15.

Edwards 1976

Edwards G, Gross MM. Alcohol dependence: provisional description of a clinical syndrome. BMJ 1976;1(6017):1058‐61.

Endicott 1976

Endicott J, Spitzer RL, Fleiss JL, Cohen J. The global assessment scales: a procedure for measuring overall severity of psychiatric disturbance. Archives of General Psychiatry 1976;35:837‐44.

Fillenbaum 1997

Fillenbaum GG, Beekly D, Edland SD, Hughes JP, Heyman A, van Belle G. Consortium to establish a registry for Alzheimer's disease: development, database structure, and selected findings. Topics in Health Information Management 1997;18(1):47‐58.

First 1995

First MB, Spitzer RL, Gibbon M, Williams JBW. Structured Clinical Interview for DSM‐IV Axis I Disorders, Patient Edition (SCID‐P), version 2. New York, New York State Psychiatric Institute, Biometrics Research1995.

Flannery 1999

Flannery BA, Volpicelli JR, Pettinati HM. Psychometric properties of the Penn Alcohol Craving Scale. Alcoholism, Clinical and Experimental Research 1999;23(8):1289‐95.

Foulds 2015

Foulds JA, Adamson SJ, Boden JM, Williman JA, Mulder RT. Depression in patients with alcohol use disorders: systematic review and meta‐analysis of outcomes for independent and substance‐induced disorders. Journal of Affective Disorders 2015;185:47‐59.

Goldman 1992

Goldman HH, Skodol AE, Lave TR. Revising axis V for DSM‐IV: a review of measures of social functioning. American Journal of Psychiatry 1992;149(9):1148‐56.

GRADE 2004

The GRADE working group. Grading quality of evidence and strength of recommendations. BMJ 2004;328:1490‐4.

Grant 1995

Grant BF, Harford TC. Comorbidity between DSM‐IV alcohol use disorders and major depression: results of a national survey. Drug and Alcohol Dependence 1995;39:197‐206.

Grant 2015

Grant BF, Goldstein RB, Saha TD, Chou SP, Jung J, Zhang H, et al. Epidemiology of DSM‐5 Alcohol Use Disorder: Results From the National Epidemiologic Survey on Alcohol and Related Conditions III. JAMA Psychiatry 2015;72(8):757‐66.

Guy 1975

Guy W. ECDEU Assessment Manual for Psychopharmacology. Rockville (MD): National Institute of Mental Health, 1975.

Guy 1976

Guy W. ECDEU Assessment Manual for Psychopharmacology. Washington (DC): DHEW Publication, 1976.

Guyatt 2008

Guyatt GH, Oxman AD, Vist GE, Kunz R, Falck‐Ytter Y, Alonso‐Coello P, et al. GRADE: an emerging consensus on rating quality of evidence and strength of recommendations. BMJ 2008;336(7560):924‐6.

Guyatt 2011

Guyatt GH, Oxman AD, Montori V, Vist G, Kunz R, Brozek J, et al. GRADE guidelines: 5. Rating the quality of evidence ‐ publication bias. Journal of Clinical Epidemiology 2011;64:1277‐82.

Hamilton 1959

Hamilton M. The assessment of anxiety states by rating. British Journal of Medical Psychology 1959;32:50‐5.

Hamilton 1960

Hamilton M. A rating scale for depression. Journal of Neurology and Psychiatry 1960;23:56‐61.

Hamilton 1967

Hamilton M. Development of a rating scale for primary depressive illness. British Journal of Social and Clinical Psychology 1967;6:278‐96.

Hamilton 1969

Hamilton M. Diagnosis and rating of anxiety. British Journal of Psychiatry 1969;Special publication:76‐9.

Hasin 1996

Hasin DS, Trautman KD, Miele GM, Samet S, Smith M, Endicott J. Psychiatric Research Interview for Substance and Mental disorders (PRISM): reliability for substance abusers. American Journal of Psychiatry 1996;153(9):1195‐201.

Hasin 2005

Hasin DS, Goodwin RD, Stinson FS, Grant BF. Epidemiology of major depressive disorder: results from the national epidemiologic survey on alcoholism and related conditions. Archives of General Psychiatry 2005;62(10):1097‐106.

Higgins 2011

Higgins JPT, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Iovieno 2011

Iovieno N, Tedeschini E, Bentley KH, Evins AE, Papakostas GI. Antidepressants for major depressive disorder and dysthymic disorder in patients with comorbid alcohol use disorders: a meta‐analysis of placebo‐controlled randomized trials. Journal of Clinical Psychiatry 2011;72(8):1144‐51.

Kadden 1992

Kadden R, Carroll K, Donovan D, Cooney N, Monti P. Cognitive‐behavioral coping skills therapy manual: a clinical research guide for therapists treating individuals with alcohol abuse and dependence. Vol. 3, Washington (DC): NIAAA Project MATCH Monograph Series, 1992.

Kranzler 1996

Kranzler HR, Burleson JA, Brown J, Babor TF. Fluoxetine treatment seems to reduce the beneficial effects of cognitive behavioral therapy in type B alcoholics. Alcoholism, Clinical and Experimental Research 1996;20:1534‐41.

Kranzler 2000

Kranzler HR, Modesto‐Lowe V, Van Kirk J. Naltrexone vs. nefazodone for treatment of alcohol dependence. A placebo‐controlled trial. Neuropsychopharmacology 2000;22(5):493‐503.

LeMoal 2007

Le Moal M, Koob GF. Drug addiction: pathways to the disease and pathophysiological perspectives. European Neuropsychopharmacology 2007;17:377‐93.

Levine 1986

Levine J, Schooler NR. SAFTEE: a technique for the systematic assessment of side effects in clinical trials. Psychopharmacology Bulletin 1986;22(2):343‐81.

Lingjaerde 1987

Lingjaerde O, Ahlfors UG, Bech P, Dencker SJ, Elgen K. The UKU side effect rating scale. Acta Psychiatrica Scandinavica 1987;76(suppl 334):1‐96.

McLellan 1992

McLellan AT, Kushner H, Metzger D, Peters R, Smith I, Grissom G, et al. The fifth edition of the Addiction Severity Index. Journal of Substance Abuse Treatment 1992;9(3):199‐213.

Miller 1992

Miller WR, Zweben A, DiClemente CC, Rychtarik RG. Motivational Enhancement Therapy Manual. Vol. 2, Washington (DC): Project MATCH Monograph Series. National Institute on Alcohol Abuse and Alcoholism, 1992.

Miller 1995

Miller WR, Tonigan JS, Longabaugh R. The Drinker Inventory of Consequences (DrInC): an instrument for assessing adverse consequences of alcohol abuse (Project MATCH Monograph Series). NIH Publication No. 95‐3911. Vol. 4, Rockville (MD): US Department of Health and Human Services, Public Health Service, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, 1995.

Moher 2001

Moher D, Jones A, Lepage L, CONSORT Group (Consolidated Standards for Reporting of Trials). The CONSORT statement: revised recommendations for improving the quality of reports of parallel‐group randomized trials. JAMA 2001;285:1992‐5.

Montgomery 1979

Montgomery SA, Åsberg M. A new depression scale designed to be sensitive to change. British Journal of Psychiatry 1979;134:382‐9.

Naranjo 2001

Naranjo CA, Knoke DM. The role of selective serotonin reuptake inhibitors in reducing alcohol consumption. Journal of Clinical Psychiatry 2001;62(Suppl 20):18‐25.

Nunes 2004

Nunes EV, Levin FR. Treatment of depression in patients with alcohol or other drug dependence: a meta‐analysis. JAMA 2004;29:1887‐96.

Nunes 2006

Nunes EV, Rounsaville BJ. Comorbidity of substance use with depression and other mental disorders: from Diagnostic and Statistical Manual of Mental Disorders, fourth edition (DSM‐IV) to DSM‐V. Addiction 2006;101:S89‐96.

O'Donnell 2011

O'Donnell JM, Shelton RC. Drug Therapy of Depression and Anxiety Disorders. In: Brunton LL, Chabner BA, Knollmann BC editor(s). Goodman & Gilman's The Pharmacological Basis of Therapeutics. 12th Edition. Columbus (OH): McGraw‐Hill, Medical Publishing Division, 2011:397‐415.

Overall 1962

Overall JE, Gorham DR. The Brief Psychiatric Rating Scale. Psychological Reports 1962;10:799‐812.

Pettinati 2000

Pettinati HM, Volpicelli JR, Kranzler HR, Luck G, Rukstalis MR, Cnaan A. Sertraline treatment for alcohol dependence: interactive effects of medication and alcoholic subtype. Alcoholism, Clinical and Experimental Research 2000;24(7):1041‐9.

Pettinati 2001b

Pettinati HM, Volpicelli JR, Luck G, Kranzler HR, Rukstalis MR, Cnaan A. Double‐blind clinical trial of sertraline treatment for alcohol dependence. Journal of Clinical Psychopharmacology 2001;21:143‐53.

Pettinati 2004

Pettinati HM. Antidepressant treatment of co‐occurring depression and alcohol dependence. Biological Psychiatry 2004;56:785‐92.

Pettinati 2013

Pettinati HM, O'Brien CP, Dundon WD. Current status of co‐occurring mood and substance use disorders: a new therapeutic target. American Journal of Psychiatry 2013;170(1):23‐30.

Raistrick 1994

Raistrick D, Bradshaw J, Tober G, Weiner J, Allison J, Healey C. Development of the Leeds Dependence Questionnaire (LDQ): a questionnaire to measure alcohol and opiate dependence in the context of a treatment evaluation package. Addiction 1994;89(5):563‐72.

Regier 1990

Regier DA, Farmer ME, Rae DS, Locke BZ, Keith D, Judd LL, et al. Comorbidity of mental disorders with alcohol and other drug abuse. JAMA 1990;264:2511‐8.

Rockliff 1971

Rockliff BW. A brief rating scale for antidepressant drug trials. Comprehensive Psychiatry 1971;12(2):122‐35.

Saunders 1993

Saunders JB, Aasland OG, Babor TF, de la Fuente JR, Grant M. Development of the Alcohol Use Disorders Identification Test (AUDIT): WHO collaborative project on early detection of persons with harmful alcohol consumption ‐ II. Addiction 1993;88(6):791‐804.

Schneider 2009

Schneider B. Substance use disorders and risk for completed suicide. Archives of Suicide Research 2009;13:303‐16.

Schuckit 1997

Schuckit MA, Tipp JE, Bergman M, Reich W, Hesselbrock VM, Smith TL. Comparison of induced and independent major depressive disorders in 2,945 alcoholics. American Journal of Psychiatry 1997;154:948‐57.

Schuckit 2006

Schuckit MA. Comorbidity between substance use disorders and psychiatric conditions. Addiction 2006;101(Suppl 1):76‐78.

Schünemann 2006

Schünemann HJ, Jaeschke R, Cook D, Bria W, El‐Solh A, Ernst A, et al. An official ATS statement: grading the quality of evidence and strength of recommendations in ATS guidelines and recommendations. American Journal of Respiratory and Critical Care Medicine 2006;174:605‐14.

Scott 1976

Scott J, Huskisson EC. Graphic representation of pain. Pain 1976;2:175‐84.

Selzer 1971

Selzer M. The Michigan Alcoholism Screening Test (MAST): the quest for a new diagnostic instrument. American Journal of Psychiatry 1971;127:1653‐8.

Sheehan 1998

Sheehan DV, Lecrubier Y, Sheehan KH, Amorim P, Janavs J, Weiller E, et al. The Mini‐International Neuropsychiatric Interview (M.I.N.I.): the development and validation of a structured diagnostic psychiatric interview for DSM‐IV and ICD‐10. Journal of Clinical Psychiatry 1998;59(Suppl 20):22‐33.

Shelton 1969

Shelton J, Hollister LE, Gocka EF. The drinking behavior interview (an attempt to quantify alcoholic impairment). Diseases of the Nervous System 1969;30(7):464‐7.

Sher 2005

Sher L, Oquendo MA, Galfalvy HC, Grunebaum MF, Burke AK, Zalsman G, et al. The relationship of aggression to suicidal behavior in depressed patients with a history of alcoholism. Addictive behaviours 2005;30:1144‐53.

Shirayama 2006

Shirayama Y, Chaki S. Neurochemistry of the nucleus accumbens and its relevance to depression and antidepressant action in rodents. Current Neuropharmacology 2006;4(4):277‐91.

Skinner 1982

Skinner HA, Allen BA. Alcohol dependence syndrome: measurement and validation. Journal of Abnormal Psychology 1982;91:199‐209.

Skinner 1984

Skinner HA, Horn JL. Alcohol dependence scale: user's guide. Toronto: Addiction Research Foundation1984.

Sobell 1992

Sobell L, Sobell M. Timeline follow‐back: a technique for assessing self‐reported alcohol consumption. In: Litten RZ, Allen JP editor(s). Measuring Alcohol Consumption. Totowa (NJ): The Humana Press, 1992:41‐72.

Spielberger 1970

Spielberger C, Gorsuch A, Lushene R. The State‐Trait Anxiety Inventory. Palo Alto (CA): Consulting Psychologists Press, 1970.

Spielberger 1983

Spielberger CD, Gorsuch RL, Lushene RE, Vagg PR, Jacobs GA. State‐Trait Anxiety Inventory for Adults. Palo Alto (CA): Mind Garden, 1983.

Spitzer 1992

Spitzer RL, Williams BW, Gibbon M, First MB. The Structured Clinical Interview for DSM‐III‐R (SCID). I: History, rationale, and description. Archives of General Psychiatry 1992;49:624‐9.

Stahl 2003

Stahl SM, Grady MM. Differences in mechanism of action between current and future antidepressants. Journal of Clinical Psychiatry 2003;64(Suppl 13):13‐7.

Torrens 2005

Torrens M, Fonseca F, Mateu G, Farré M. Efficacy of antidepressants in substance use disorders with and without comorbid depression. A systematic review and meta‐analysis. Drug and Alcohol Dependence 2005;78(1):1‐22.

Ware 1992

Ware JE, Sherbourne CD. The MOS 36‐item Short Form Health Survey I: conceptual framework and item selection. Medical Care 1992;30:473‐83.

WHO 1993

World Health Organization. The ICD 10. The ICD‐10 Classification of Mental and Behavioural Disorders. Diagnostic Criteria for Research. Geneva: WHO, 1993.

Wilcox 2004

Wilcox HC, Conner KR, Caine ED. Association of alcohol and drug use disorders and completed suicide: an empirical review of cohort studies. Drug and Alcohol Dependence 2004;76S:11‐9.

Zung 1965

Zung WW. A self‐rating depression scale. Archives of General Psychiatry 1965;12:63‐70.

Referencias de otras versiones publicadas de esta revisión

Pani 2010

Pani PP, Trogu E, Amato L, Davoli M. Antidepressants for the treatment of depression in alcohol dependent individuals. Cochrane Database of Systematic Reviews 2010, Issue 7. [DOI: 10.1002/14651858.CD008581]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Adamson 2015

Methods

Randomized, double‐blind, placebo‐controlled trial

Participants

138 depressed people with alcohol dependence (56 men and 82 women; mean (± SD) age 43.6 ± 9.1 years).

Inclusion criteria:

  • aged 17‐65 years

  • current DSM‐IV diagnoses of alcohol dependence and depression

  • MADRS score > 20

Exclusion criteria:

  • past regular intravenous drug use for > 2 weeks

  • recreational use of any opioid drugs in the previous 4 weeks or a current requirement for ongoing opioid use

  • psychosis, including psychotic delirium complicating alcohol or other drug withdrawal

  • mania or hypomania

  • significant current suicidality or homicidality

  • current severe psychiatric symptoms requiring hospitalization,

  • unstable physical disease

  • use of disulfiram, naltrexone, antidepressant, or mood stabilizing medication in past 4 weeks

  • serum AST, ALT, or GGT greater than 3 × the upper limit of laboratory reference range, or a bilirubin level > upper limit of reference range

  • pregnancy, breastfeeding, or unwillingness to use a reliable method of contraception in women of childbearing age

  • current or pending imprisonment

Participants with bipolar disorder were excluded.

Interventions

Drugs:

  • citalopram (up to 60 mg/day) + naltrexone (up to 100 mg/day) (73 participants; 29 men and 44 women)

  • placebo + naltrexone (up to 100 mg/day) (65 participants; 27 men and 38 women)

Psychotherapy: manualized clinical case management was delivered by experienced addiction clinicians.

Scheduled duration of treatment: 12 weeks

Sites: 7 addiction clinics spanning urban, provincial, and rural catchments in Australia.

Setting: outpatients

Route of administration: orally

Starting dose:

  • citalopram: 20 mg/day in week 1; if tolerated, dose then increased to 40 mg/day. After 6 weeks, dose could be further increased to 60 mg/day if participants remained depressed

  • naltrexone: 25 mg daily for 1 week, then increased to 50 mg in participants without significant adverse effects. Dose could be further increased to 75 mg or 100 mg after 6 weeks

Pattern of dose reduction: information not available

Outcomes

Depression:

  • final MADRS score

  • final SCL‐90 score

  • remission

Alcohol dependence:

  • rate of abstinent days

  • number of heavy drinking days per week (obtained from the rate of heavy drinking days)

  • number of drinks per drinking day

  • final LDQ score

Dropouts

Adverse effects

Notes

Baseline characteristics of participants

Depression:

  • primary depression (% of participants): 76.1%

  • duration (years): 19.3

  • MADRS score (mean ± SD): 31.0 ± 5.8

Alcohol dependence:

  • number of drinks per drinking day (mean ± SD): 14.3 ± 8.0

  • duration (years): 13.8

  • being actively drinking: participants were not required to be abstinent

Other psychiatric comorbidity: 47.1% of participants had current anxiety disorder.

Other substance‐use disorders: 14.5% of participants had current substance dependence.

Other characteristics of study

Other pharmacological treatment offered: all participants received naltrexone.

Funding sources: study funded by Health Research Council of New Zealand grant HRC 07/138.

Declaration of interest: authors declared no conflict of interest.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomization was performed using a computer‐generated random number table.

Allocation concealment (selection bias)

Low risk

Treatment allocation was conducted by an administrative staff member independent of study investigators or research clinicians, and the allocation sequence record was stored securely.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The investigators, doctors, participants, and any other staff members taking part in the experiment were unaware which of the groups any particular participant belonged to.

Blinding of outcome assessment (detection bias) objective

Low risk

No information on blinding of outcome assessors.

Blinding of outcome assessment (detection bias) subjective

Unclear risk

No information on blinding of outcome assessors.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing data were imputed using appropriate methods.

Selective reporting (reporting bias)

Unclear risk

Numbers of dropouts per group were missing.

Altamura 1990

Methods

Randomized, placebo‐controlled, double‐blind trial

Participants

30 people with alcohol dependence with dysthymia (24 men and 6 women; mean (± SD) age: 44.5 ± 2.6 years).

Inclusion criteria:

  • alcohol dependence and dysthymia according to DSM‐III‐R

  • HRSD score ≥ 18

Exclusion criteria:

  • diagnosis of cirrhosis

  • substance‐use disorders by other substances

  • relevant internal or neurological conditions

Participants with bipolar disorder: information not available.

Interventions

Drugs:

  • viloxazine (400 mg/day, in 4 daily administrations; 15 participants; information on number of men and women not available)

  • placebo (15 participants; information on number of men and women not available)

Psychotherapy: information not available

Scheduled duration of treatment: 12 weeks

Site: 1 centre, Department of Clinical Psychiatry, Policlinico, Milan, Italy

Setting: inpatient setting for first 4 weeks, then outpatients for following 8 weeks.

Route of administration: orally

Starting dose: information not available

Pattern of dose reduction: information not available

Outcomes

Depression:

  • final HRSD score (obtained from a figure)

Alcohol dependence: information not available

Dropouts

Adverse effects: information not available

Notes

Baseline characteristics of participants

Depression:

  • primary depression: "Patients were not depressed but affected by dysthymic disorder"

  • duration: information not available

  • HRSD score (mean ± SD): viloxazine = 26.7 ± 2.8; placebo = 25.6 ± 1.7

Alcohol dependence:

  • severity: information not available

  • duration of alcohol consumption (mean ± SD): 10.2 ± 1.3 years

  • being actively drinking: information not available

  • length of abstinence: information not available

Other psychiatric comorbidity: information not available

Other substance‐use disorders: participants with other substance‐use disorders were excluded.

Other characteristics of study

Other pharmacological treatment offered: information not available

Funding sources: information not available

Declaration of interest: information not available

Other information

Standard errors were converted into SDs.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random allocation stated but no further details provided.

Allocation concealment (selection bias)

Unclear risk

Method of concealment not described.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Information insufficient to permit judgement.

Blinding of outcome assessment (detection bias) objective

Low risk

No information on blinding of outcome assessors.

Blinding of outcome assessment (detection bias) subjective

Unclear risk

No information on blinding of outcome assessors.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Method to account for missing data not described. Intention‐to‐treat approach not reported. No high numbers of dropouts or unbalanced between groups.

Selective reporting (reporting bias)

High risk

DBI and DOTES scores are reported in 2 figures (figures 3 and 4 of the publication) in which the titles of the y axis do not correspond to those reported in the legends and in the text, and the positions of the points do not correspond to the values indicated in the y axes. Accordingly, these results were not included in the present meta‐analysis.

Altintoprak 2008

Methods

Double‐blind, randomized, comparative trial

Participants

44 depressed people with alcohol dependence (number of men and women: information not available; mean age: information not available). Sociodemographic characteristics available only for 36 participants (20 mirtazapine, 16 amitriptyline).

Inclusion criteria:

  • aged 18‐65 years

  • current DSM‐IV diagnoses of alcohol dependence and depressive disorder

  • HDRS score ≥ 14 after detoxification

Exclusion criteria:

  • serious physical illness

  • for women, lack of protection against pregnancy, pregnancy, or breastfeeding

  • other major psychiatric disorder on the DSM‐IV axis‐I other than depressive disorder

  • history of a psychiatric problem other than depressive disorder

  • organic brain diseases

  • history of hypersensitivity to mirtazapine or amitriptyline

  • other drug dependence and abuse, excluding nicotine and caffeine

  • consumption of alcohol during study

Participants with bipolar disorder were excluded.

Interventions

Drugs:

  • mirtazapine (30‐60 mg/day; 24 participants)

  • amitriptyline (100‐150 mg/day; 20 participants)

Psychotherapy: information not available.

Scheduled duration of treatment: 8 weeks.

Site: Ege University School of Medicine Hospital, Specialized Addiction Unit, Izmir, Turkey

Setting: inpatient

Route of administration: orally

Starting dose:

  • mirtazapine: 15 mg/day (increased to 30 mg/day at the third day; at end of first week, dose was increased to 45‐60 mg/day if severity of symptoms persisted)

  • amitriptyline 50 mg/day (increased to 100 mg/day at the third day; at end of first week, dose was increased to 125‐150 mg/day if severity of symptoms persisted)

Pattern of dose reduction: information not available

Outcomes

Depression:

  • final HRSD score

Alcohol dependence: data not available

Alcohol craving:

  • final score in a questionnaire prepared by authors

Dropouts: data not available

Adverse effects:

  • evaluated using the UKU scale

Bodyweight:

  • final value

Anxiety:

  • final STAI score

Notes

Baseline characteristics of participants

Depression:

  • primary depression (rate of participants): 34.1%

  • duration: information not available

  • HRSD score (mean ± SD): mirtazapine = 24.0 ± 4.4; amitriptyline = 23.7 ± 4.8

Alcohol dependence:

  • MAST score (mean ± SD): 38.9 ± 6.1

  • duration (mean ± SD): 12.1 ± 3.9 years

  • being actively drinking: people who consumed alcohol during study were excluded from study.

  • length of abstinence: 2 weeks

Anxiety:

  • STAI score (mean ± SD): mirtazapine = 51.8 ± 3.9; amitriptyline = 53.4 ± 4.5.

Global assessment: information not available.

Weight (mean ± SD):

  • mirtazapine = 75.9 ± 16.2 kg; amitriptyline = 71.4 ± 10.9 kg

Other psychiatric comorbidity: participants with other psychiatric disorders were excluded.

Other substance use disorders: participants with other substance use disorders were excluded.

Other characteristics of study

Other pharmacological treatment: no other pharmacological treatment was allowed.

Funding sources: not available.

Declaration of interest: not available.

Other information

After their inclusion in study, participants were admitted at a specialized department for alcohol detoxification on an inpatient basis. Alcohol consumption was prohibited during hospitalization and people who consumed alcohol during study were excluded from study. At end of alcohol detoxification treatment (approximately 10‐14 days), people were included in study.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random allocation stated. No further details provided.

Allocation concealment (selection bias)

Unclear risk

Method of concealment not described.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind stated. Medication and placebo prepared to appear identical ("Both the clinicians and patients were blind to the treatment. Drugs were given in identical‐looking opaque capsules").

Blinding of outcome assessment (detection bias) objective

Low risk

No information on the blinding of outcome assessors.

Blinding of outcome assessment (detection bias) subjective

Unclear risk

No information on the blinding of outcome assessors.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Intention‐to‐treat approach not used ("Dropouts were not included in the analysis due to missing data"). People who consumed alcohol during study were excluded by study.

Selective reporting (reporting bias)

High risk

People who consumed alcohol during study were excluded by study.

Butterworth 1971a

Methods

Randomized, double‐blind, comparative trial

Participants

39 people with alcohol dependence (all men; mean age: information not available) with a significant degree of anxious‐depressive symptomatology.

Inclusion criteria:

  • aged 20‐55 years

  • significant degree of anxiety and depression as determined by psychiatric interview

  • current diagnosis of alcohol dependence

Exclusion criteria:

  • physical brain

  • liver illnesses

  • psychotic disorder

Participants with bipolar disorder: information not available

Interventions

Drugs:

  • doxepin (75 mg/day; 20 participants, all men; mean age: 45 years)

  • diazepam (15 mg/day; 19 participants, all men; mean age: 41 years)

Psychotherapy: information not available

Scheduled duration of treatment: 3 weeks

Site: Alcoholism Treatment Service of East Louisiana State Hospital, Mandeville, LA, USA

Setting: inpatients

Route of administration: orally

Starting dose: information not available

Pattern of dose reduction: information not available

Outcomes

Depression:

  • final BPRS score

  • final ZUNG score

  • response

Alcohol dependence: data not available

Dropouts

Adverse effects

Notes

Baseline characteristics of participants

Depression:

  • primary depression: information not available

  • duration: information not available

  • BPRS score (mean): doxepin = 76.3; diazepam = 73.8

  • ZUNG score (mean): doxepin = 47.8; diazepam = 37.9

Alcohol dependence:

  • severity: information not available

  • being actively drinking: participants not actively drinking

Other psychiatric comorbidity: information not available

Other substance‐use disorders: information not available

Other characteristics of study

Other pharmacological treatment: other concomitant therapy not allowed

Funding source: medications were supplied by Laboratories of Pfizer Inc.

Declaration of interest: information not available

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random allocation stated. No further details provided.

Allocation concealment (selection bias)

Unclear risk

Method of concealment not described.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind stated. Medications prepared to appear identical. Evaluations conducted by 2 independent investigators and the results pooled.

Blinding of outcome assessment (detection bias) objective

Low risk

No information on blinding of outcome assessors.

Blinding of outcome assessment (detection bias) subjective

Unclear risk

No information on blinding of outcome assessors.

Incomplete outcome data (attrition bias)
All outcomes

High risk

No information on dropouts provided. Methods applied to account for missing data not described. Intention‐to‐treat approach not reported.

Selective reporting (reporting bias)

High risk

No information on dropouts provided. Methods applied to account for missing data not described. Intention‐to‐treat approach not reported.

Butterworth 1971b

Methods

Randomized, placebo‐controlled, double‐blind trial

Participants

40 depressed people with alcohol dependence (all men; mean age: majority aged 31‐50 years)

Inclusion criteria:

  • alcohol dependence requiring involuntarily admission for detoxification

  • diagnoses of depression according to clinical impression

  • LDRS score ≥ 12

Exclusion criteria:

  • hepatic disease

  • organic brain damage

  • psychosis

Participants with bipolar disorder: information not available

Interventions

Drugs:

  • imipramine (75‐200 mg/day; 20 participants)

  • placebo (20 participants)

Psychotherapy: none

Scheduled duration of treatment: 3 weeks

Site: Alcoholic Treatment Service, East Louisiana State Hospital, Jackson, LA, USA

Setting: inpatients for first 3‐4 days for treatment of alcohol withdrawal, then 3 weeks for trial

Route of administration: orally

Starting dose:

  • 75 mg/day

  • increased to maximum 200 mg/day according to individual requirements

  • reduced if indicated by adverse effects

Pattern of dose reduction: information not available

Outcomes

Depression:

  • difference between basal and final LRDS score

  • response

Alcohol dependence: data not available

Dropouts

Adverse effects

Notes

Baseline characteristics of participants

Depression:

  • primary depression: information not available

  • duration: information not available

  • after detoxification and washout, LDRS score (mean ± SD): imipramine = 16.0 ± 3.1; placebo = 15.6 ± 3.3.

Alcohol dependence:

  • duration: for 24 participants = 1‐5 years; for 16 participants ≥ 10 years;

  • severity: all participants required involuntarily admission for detoxification, many participants had been hospitalized repeatedly for detoxification, in some instances as many as 30 times;

  • being actively drinking: participants were abstinent;

  • length of abstinence: 0.5 weeks 3‐4 days for the treatment of alcohol withdrawal).

Other psychiatric comorbidity: information not available.

Other substance‐use disorders: information not available.

Other characteristics of study

Other pharmacological treatment offered: participants received pharmacological treatment to control the acute symptoms of alcohol withdrawal for 3‐4 days.

Funding sources: information not available

Declaration of interest: information not available

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random allocation stated. No further details provided.

Allocation concealment (selection bias)

Unclear risk

Method of concealment not described.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind stated. Medication and placebo prepared to appear identical. No specific reference made to blinding of participants and personnel.

Blinding of outcome assessment (detection bias) objective

Low risk

No information on the blinding of outcome assessors.

Blinding of outcome assessment (detection bias) subjective

Unclear risk

No information on the blinding of outcome assessors.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Methods applied to account for missing data not described. Intention‐to‐treat approach not reported. People who left study were replaced by other people ("One patients taking imipramine left the hospital ... and global evaluation were omitted. Two additional patients left without permission just after entering the trial, and were therefore replaced in the study. One had received six doses of imipramine and the other one placebo").

Selective reporting (reporting bias)

Unclear risk

Information insufficient to permit judgement.

Cocchi 1997

Methods

Randomized comparative trial

Participants

122 depressed people with alcohol dependence (95 men and 27 women; mean age: 42 years)

Inclusion criteria:

  • DSM‐IV diagnosis F10‐24

  • ZUNG score > 49

Exclusion criteria: information not available

Participants with bipolar disorder: information not available

Interventions

Drugs:

  • paroxetine (20 mg/day; 61 participants; 49 men and 12 women; age (mean ± SD) = 42.1 ± 11.5 years)

  • amitriptyline (25 mg/day; 61 participants; 46 men and 15 women; age (mean ± SD) = 42.2 ± 10.7 years)

Psychotherapy: information not available

Scheduled duration of treatment: 3‐4 weeks

Site: Alcohol Unit, casa di Cura Villa Silvia per malattie nervose e mentali, Senigallia, Italy

Setting: inpatients

Route of administration: orally

Starting dose: information not available

Pattern of dose reduction: information not available

Outcomes

Depression

  • final ZUNG score

  • response (according to ZUNG)

  • remission (according to ZUNG)

Alcohol dependence: data not available

Dropouts: data not available

Adverse effects: data not available

Notes

Baseline characteristics of participants

Depression:

  • primary depression: information not available

  • duration: information not available

  • ZUNG score (mean ± SD): paroxetine = 68.8 ± 9.0; amitriptyline = 63.3 ± 6.0

Alcohol dependence: data not available

Other psychiatric comorbidity: information not available

Other substance use disorders: information not available

Other characteristics of study

Other pharmacological treatment: information not available

Funding sources: information not available

Declaration of interest: information not available

Other information

Data on response and remission were excluded because evaluated using a self‐administered scale.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random allocation stated. No further details provided.

Allocation concealment (selection bias)

Unclear risk

Method of concealment not described.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No information on the design (double‐blind or open trial), on the preparation and appearance of medications, and on blinding of participants and personnel.

Blinding of outcome assessment (detection bias) objective

Low risk

No information on study design (double‐blind or open trial).

Blinding of outcome assessment (detection bias) subjective

High risk

No information on study design (double‐blind or open trial).

Incomplete outcome data (attrition bias)
All outcomes

High risk

Methods applied to account for missing data. Intention‐to‐treat approach not reported. No high number of dropouts or unbalanced between groups.

Selective reporting (reporting bias)

Unclear risk

Information insufficient to permit judgement.

Cornelius 1997

Methods

Randomized, double‐blind, placebo‐controlled trial

Participants

51 depressed people with alcohol dependence (26 men and 25 women; age (mean ± SD) = 34.8 ± 10.2 years)

Inclusion criteria:

  • current DSM‐III‐R diagnoses of depression and alcohol dependence

Exclusion criteria:

  • diagnosis of bipolar disorder, schizoaffective disorder, schizophrenia, or non‐alcohol substance dependence

  • hyperthyroidism or hypothyroidism

  • clinically significant medical diseases

  • pregnancy

  • mental retardation or cognitive impairment

  • use of antipsychotic or antidepressant medication in the previous month

Participants with bipolar disorder were excluded.

Interventions

Drugs:

  • fluoxetine (20 mg/day; 25 participants)

  • placebo (26 participants)

Psychotherapy:

  • weekly supportive psychotherapy sessions

  • weekly meetings with an attending psychiatrist with expertise in treating people with dual‐disorder

  • attendance at Alcoholics Anonymous was encouraged.

Scheduled duration of treatment: 12 weeks

Site: Western Psychiatric Institute and Clinic of the University of Pittsburgh, Pittsburgh, USA

Setting: inpatients for first 2 weeks of abstinence, then outpatients

Route of administration: orally

Starting dose:

  • 20 mg/day

  • increased to 40 mg/day after 2 weeks if substantial residual depressive symptoms persisted

Pattern of dose reduction: information not available

Outcomes

Depression:

  • difference between baseline and final HRSD score

  • difference between baseline and final BDI score

Alcohol dependence:

  • rate of abstinent days (obtained from cumulative number of drinking days)

  • number of abstinent participants

  • number of drinks per drinking day

  • number of heavy drinking days per week (obtained from the cumulative number of heavy drinking days)

  • time to first relapse (obtained from the number of weeks until first relapse)

Global assessment:

  • severity (difference between baseline and final GAS score)

Dropouts: information not available

Adverse effects: information not available

Notes

Baseline characteristics of participants

Depression:

  • primary depression: 100%

  • duration: information not available

  • after detoxification and washout, HRSD score (mean ± SD): fluoxetine = 19.2 ± 8.2; placebo = 17.9 ± 8.1

  • number of diagnostic criteria (mean ± SD): fluoxetine = 6.7 ± 1.1; placebo = 6.8 ± 1.1

  • current suicide ideation: fluoxetine = 92.0%; placebo = 88.5%

Alcohol dependence:

  • number of diagnostic criteria (mean ± SD): fluoxetine = 5.5 ± 1.6; placebo = 5.9 ± 1.8; range: 3.9‐7.7

  • duration: information not available

  • being actively drinking: participants were actively drinking

  • length of abstinence: 0

  • number of drinking days in past 90 days (mean ± SD): fluoxetine = 54.5 ± 29.2; placebo = 45.2 ± 28.9

  • number of days drinking to drunkenness in past 90 days (mean ± SD): fluoxetine = 40.1 ± 27.7; placebo = 32.0 ± 26.4

Other psychiatric comorbidity: participants with other mental disorders were excluded.

Other substance‐use disorders: participants with substance‐use disorders were excluded. Abuse of other substances was not an exclusionary criterion, provided that alcohol was the main substance of abuse.

Other characteristics of study

Other pharmacological treatment: other pharmacological treatments were not allowed.

Funding sources: work was supported by the National Institute on Alcohol Abuse and Alcoholism (grants AA09127 and AA10523), and by the Mental Health Clinical Research Center, Rockville, MD (grant MH30915).

Declarations of interest: information not available

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomization balanced for gender and race. It was not reported whether a computer‐generated list was used.

Allocation concealment (selection bias)

Unclear risk

Information insufficient to permit judgement. Method of concealment not described.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Medications were administered in identical opaque capsules. Substantial blood levels of fluoxetine were observed in more than 99% of participants assigned to fluoxetine. Not reported if blood analyses were made also to participants who received placebo.

Blinding of outcome assessment (detection bias) objective

Low risk

No information on the blinding of outcome assessors.

Blinding of outcome assessment (detection bias) subjective

Unclear risk

No information on the blinding of outcome assessors.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Intention‐to‐treat analysis and last point carried forward analysis applied.

Selective reporting (reporting bias)

Unclear risk

Information insufficient to permit judgement.

Cornelius 2016

Methods

Randomized, double‐blind, placebo‐controlled trial

Participants

14 depressed people with alcohol dependence (10 men and 4 women; mean age = 41.3 years)

Inclusion criteria:

  • aged 18‐55 years

  • current DSM‐IV diagnoses of depression and alcohol dependence

  • eligible for outpatient treatment

Exclusion criteria:

  • aged < 18 years or over 55 years

  • presence of psychotic symptoms or a diagnosis involving psychosis

  • receiving psychotropic medication in the prior month

  • current DSM diagnosis of dependence or abuse on substances other than alcohol, cannabis, nicotine, or caffeine

  • current significant medical or neurological condition

  • suicidal ideation in the last 3 months, or lifetime suicidal attempt

  • positive pregnancy test or breastfeeding

  • inability or unwillingness to use contraceptive methods

  • inability to read or understand study forms

  • pending incarceration

  • current participation in another research study

Participants with bipolar disorder: information not available

Interventions

Drugs:

  • mirtazapine (30 mg/day; 7 participants; 4 men and 3 women)

  • placebo (7 participants; 6 men and 1 woman)

Psychotherapy:

  • brief MET at each assessment

Scheduled duration of treatment: 12 weeks

Site: University of Pittsburgh, Western Psychiatric Institute and Clinic, Pittsburgh, USA

Setting: outpatients

Route of administration: orally

Starting dose:

  • 15 mg/day for the first 2 weeks

  • then 30 mg/day for 12 weeks

Pattern of dose reduction: information not available

Outcomes

Depression:

  • final BDI score

  • difference between baseline and final BDI score

Alcohol dependence:

  • number of drinking days per week

  • number of drinks per drinking days

  • number of drinks per week

  • number of heavy drinking days per week

Craving for alcohol:

  • final OCDS score

  • difference between baseline and final OCDS score

Dropouts

Adverse effects

Notes

Baseline characteristics of participants

Depression:

  • primary depression: information not available

  • duration: information not available

  • BDI score (mean ± SD): mirtazapine = 27.6 ± 7.7; placebo = 26.1 ± 11.7

Alcohol dependence:

  • severity: information not available

  • duration: information not available

  • number of drinks per drinking day (mean ± SD): 6.6 ± 2.0

  • being actively drinking: participants were not abstinent

Other psychiatric comorbidity: participants with other mental disorders were excluded.

Other substance use disorders: participants with substance use disorders were excluded.

Other characteristics of study

Other pharmacological treatment: participants did not receive other pharmacological treatments.

Funding sources: study received grants from the National Institute on Alcohol Abuse and Alcoholism (R21 AA022123, R21 AA022863, R01 AA013370, R01 AA015173, K24 AA15320) and from the National Institute on Drug Abuse (R01 DA019142, P50 DA05605, K02 DA017822).

Declarations of interest: information not available

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information about sequence generation process to permit judgement of low or high risk.

Allocation concealment (selection bias)

Unclear risk

Method of concealment not described.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Medications were identical in appearance (identical‐looking opaque capsules).

Blinding of outcome assessment (detection bias) objective

Low risk

Medications were identical in appearance (identical‐looking opaque capsules).

Blinding of outcome assessment (detection bias) subjective

Unclear risk

No information on the blinding of outcome assessors.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Intention‐to‐treat approach reported.

Selective reporting (reporting bias)

Low risk

No dropouts

Gallant 1969 arm a

Methods

Randomized, double‐blind, placebo‐controlled trial

Participants

76 people with alcohol dependence (all men; mean age = 42 years) in association with a predominant chronic anxiety or depressive reaction.

Inclusion criteria: information not available

Exclusion criteria:

  • psychotic disorders

Participants with bipolar disorder: information not available

Interventions

Drugs:

  • doxepin (150 mg/day, in 3 daily administrations; 24 participants; all men)

  • doxepin (75 mg/day, in 3 daily administrations; 23 participants; all men)

  • placebo (29 participants; all men)

Psychotherapy: information not available

Scheduled duration of treatment: 3 weeks

Site: Alcoholism Treatment Service of Southeast Louisiana Hospital, Mandeville, LA, USA

Setting: inpatients

Route of administration: orally

Starting dose: information not available

Pattern of dose reduction: information not available

Outcomes

Depression:

  • response

Alcohol dependence: data not available

Dropouts

Adverse effects

Notes

Baseline characteristics of participants included inGallant 1969 arm a; Gallant 1969 arm b

Depression:

  • primary depression: information not available

  • duration: information not available

  • severity: information not available

Alcohol dependence:

  • severity: information not available

  • being actively drinking: information not available

  • duration: information not available

Other psychiatric comorbidity: participants with other mental disorders were included.

Other substance use disorders: information not available

Other characteristics of study

Other pharmacological treatment offered: information not available

Funding source: the project was partially supported by PHS Grant MH‐03701‐08, Psychopharmacology Research Branch, NIMH.

Declarations of interest: information not available

Other information

In the original study, 100 participants were divided into 4 groups:

  • doxepin 150 mg/day (24 participants)

  • doxepin 75 mg/day (23 participants)

  • diazepam 15 mg/day (24 participants)

  • placebo (29 participants)

In the present meta‐analysis, participants were divided into 2 substudies:

  • Gallant 1969 arm a (76 participants), in which the 2 groups with different doses of doxepin were combined into a single group and compared to placebo group

  • Gallant 1969 arm b (71 participants), in which the 2 groups with different doses of doxepin were combined into a single group and compared to diazepam group

The first arm (Gallant 1969 arm a) was included in the 'Effects of interventions: Antidepressants versus placebo' comparison and the second arm (Gallant 1969 arm b) in the 'Effects of interventions: Antidepressants versus other medications' comparison.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information about sequence generation process to permit judgement of low or high risk.

Allocation concealment (selection bias)

Unclear risk

Method of concealment not described.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Medications were identical in appearance and were coded in accordance with double‐blind procedure to ensure that all personal involved in project remained blind as to which group any given participant belonged.

Blinding of outcome assessment (detection bias) objective

Low risk

No information on blinding of outcome assessors.

Blinding of outcome assessment (detection bias) subjective

Unclear risk

No information on blinding of outcome assessors.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Methods applied to account for missing data not described. Intention‐to‐treat approach not reported. However, there were no dropouts.

Selective reporting (reporting bias)

Unclear risk

Insufficient information to permit judgement. However, there were no dropouts.

Gallant 1969 arm b

Methods

Randomized, double‐blind, placebo‐controlled trial

Participants

71 people with alcohol dependence (all men; mean age: 42 years) in association with a predominant chronic anxiety or depressive reaction (diagnosis of depression was uncertain).

Inclusion criteria: information not available

Exclusion criteria:

  • psychotic disorders

Participants with bipolar disorder: information not available

Interventions

Drugs:

  • doxepin (150 mg/day, in 3 daily administrations; 24 participants; all men)

  • doxepin (75 mg/day, in 3 daily administrations; 23 participants; all men)

  • diazepam (24 participants; all men)

Psychotherapy: information not available

Scheduled duration of treatment: 3 weeks

Site: Alcoholism Treatment Service of Southeast Louisiana Hospital, Mandeville, LA, USA

Setting: inpatients

Route of administration: orally

Starting dose: information not available

Pattern of dose reduction: information not available

Outcomes

Depression:

  • response

Alcohol dependence: data not available (probably because of inpatient setting).

Dropouts

Adverse effects

Notes

Baseline characteristics of participants included inGallant 1969 arm a; Gallant 1969 arm b

Depression:

  • primary depression: information not available

  • duration: information not available

  • severity: information not available

Alcohol dependence:

  • severity: information not available

  • being actively drinking: information not available

  • duration: information not available

Other psychiatric comorbidity: participants with other mental disorders were included.

Other substance use disorders: information not available

Other characteristics of study

Other pharmacological treatment offered: information not available

Funding source: the project was partially supported by PHS Grant MH‐03701‐08, Psychopharmacology Research Branch, NIMH.

Declarations of interest: information not available

Other information

In the original study, 100 patients were divided into 4 groups:

  • doxepin 150 mg/day (24 participants)

  • doxepin 75 mg/day (23 participants)

  • diazepam 15 mg/day (24 participants)

  • placebo (29 participants)

In the present meta‐analysis, participants were divided into 2 substudies:

  • Gallant 1969 arm a (76 participants), in which the 2 groups with different doses of doxepin were combined into a single group and compared to placebo group

  • Gallant 1969 arm b (71 participants), in which the 2 groups with different doses of doxepin were combined into a single group and compared to diazepam group

The first arm (Gallant 1969 arm a) was included in the 'Effects of interventions: Antidepressants versus placebo' comparison and the second arm (Gallant 1969 arm b) in the 'Effects of interventions: Antidepressants versus other medications' comparison.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information on sequence generation process to permit judgement

Allocation concealment (selection bias)

Unclear risk

Method of concealment not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Medications were identical in appearance and were coded in accordance with double‐blind procedure to ensure that all personal involved in project remained blind as to which group any given participant belonged.

Blinding of outcome assessment (detection bias) objective

Low risk

No information on blinding of outcome assessors.

Blinding of outcome assessment (detection bias) subjective

Unclear risk

No information on blinding of outcome assessors.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Methods applied to account for missing data not described. Intention‐to‐treat approach not reported. However, there were no dropouts.

Selective reporting (reporting bias)

Unclear risk

Insufficient information to permit judgement.

Gual 2003

Methods

Randomized, double‐blind, placebo‐controlled trial

Participants

83 depressed people with alcohol dependence (44 men and 39 women; mean age = 47 years)

Inclusion criteria:

  • age ≥ 18 years

  • current DSM‐IV and ICD‐10 diagnoses of alcohol dependence and depression or dysthymia

Exclusion criteria:

  • women who were pregnant, breastfeeding, or who were of childbearing potential and were not using reliable contraception or who wished to become pregnant during study or within 1 month after study

  • psychiatric disorders apart from alcohol dependence and depressive symptoms

  • moderate or severe liver disease including active cirrhosis or acute hepatitis

  • high suicide risk

  • requiring therapy with additional psychotropic drugs, ECT, or intensive psychotherapy during study

  • history of convulsive disorders, cerebral organic disease, or laxative misuse within the 6 months prior to receiving test drug

  • had received therapy with depot neuroleptics during the 6 months prior to their inclusion in study

  • requiring medical treatment

  • history of failure on sertraline or any other serotonin reuptake selective inhibitor, either alone or combined with another therapy

  • people in whom sertraline therapy was contraindicated

  • other severe organic diseases

Participants with bipolar disorder were excluded.

Interventions

Drugs:

  • sertraline (50–150 mg/day; 44 participants)

  • placebo (39 participants)

Psychotherapy: information not available

Scheduled duration of treatment: 24 weeks

Site: Alcohol Unit of the Hospital ‘Clínico y Provincial' in Barcelona, Spain

Setting: outpatients

Route of administration: orally

Starting dose: 50 mg/day

Pattern of dose reduction: information not available

Outcomes

Depression:

  • final HRSD score (data obtained from a figure)

  • final MADRS score (data obtained from a figure)

  • response

  • remission

Alcohol dependence:

  • rate of abstinent days

  • number of heavy drinkers

  • time to first relapse

Dropouts

Adverse effects

Notes

Baseline characteristics of participants

Depression:

  • primary depression: information not available

  • duration: about 3 years for men; about 1 year for women

  • MADRS score (mean ± SD): sertraline = 22 ± 7; placebo = 23 ± 8

  • HRSD score (mean ± SD): sertraline = 14 ± 6, placebo = 13 ± 4

Alcohol dependence:

  • severity: information not available

  • duration: about 16 years

  • being actively drinking: participants had to be "abstinent for at least 2 weeks following detoxification" and "to have a negative drug and alcohol urine screen at inclusion"

  • length of abstinence: ≥ 2 weeks

Other psychiatric comorbidity: people with other mental disorders were excluded.

Other substance‐use disorders: people with substance‐use disorders were excluded.

Other characteristics of study

Other pharmacological treatment: other pharmacological treatments were not allowed.

Funding source: information not available

Declarations of interest: information not available

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Random allocation stated. The investigator did not have access to the randomization code.

Allocation concealment (selection bias)

Unclear risk

Method of concealment was not described.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Matching packets containing placebo were provided for all possible sertraline dose progressions, so that titration could be performed double blind. Medication was dispensed in bottles with MEMS caps, which contain an electronic monitoring device that records the date and time of bottle cap openings (Aprex Corp, San Diego, CA).

Blinding of outcome assessment (detection bias) objective

Low risk

No information on the blinding of outcome assessors.

Blinding of outcome assessment (detection bias) subjective

Unclear risk

No information on the blinding of outcome assessors.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Intention‐to‐treat method utilized in all statistical analyses.

Selective reporting (reporting bias)

Low risk

For alcohol consumption data, participants with missing assessments at last observation were treated as non‐abstinent. For depression scale scores, missing data were handled on the principle of last observation carried forward.

Habrat 2006

Methods

Randomized, double‐blind, comparative trial

Participants

286 depressed people with alcohol dependence (222 men and 64 women; mean age: information not available).

Inclusion criteria:

  • aged 21‐65 years

  • ICD‐10 diagnoses of alcohol dependence or alcohol abuse and depression or bipolar disorder

  • HRSD score ≥ 16

Exclusion criteria:

  • any condition potentially dangerous, e.g. suicidal thoughts, psychotic depression, or pregnancy

  • needs of treatment for other diseases

  • treatments in near past with drugs that interfere with drugs in research or influence investigated therapy

  • lack of conscious agreement

  • depression resistant to pharmacotherapy

Participants with bipolar disorder were included.

Interventions

Drugs:

  • tianeptine (37.5 mg/day; 146 participants; 109 men and 37 women; age: information not available)

  • fluvoxamine (100 mg/day; 140 participants; 113 men and 27 women; age: information not available)

Psychotherapy: information not available

Scheduled duration of treatment: 6 weeks (responders were proposed to continue the same treatment up to 12 weeks).

Site: Department of Substance Use Prevention and Treatment, Institute of Psychiatry and Neurology, Warsaw, Poland

Setting: outpatients

Starting dose: information not available

Pattern of dose reduction: information not available

Outcomes

Depression:

  • final HRSD score

  • response

Alcohol dependence: data not available

Craving for alcohol:

  • final OCDS score

Anxiety:

  • final HRSA score

Dropout

Adverse effects

Notes

Baseline characteristics of participants

Depression:

  • primary depression (rate of participants): 12.2%

  • duration: information not available

  • HRSD score (mean ± SD): tianeptine = 22.2 ± 4.9; fluvoxamine = 21.8 ± 4.2

Alcohol dependence:

  • alcohol dependence = 89.3%; alcohol abuse = 10.7%

  • severity: information not available

  • duration: information not available

  • being actively drinking: participants had to be abstinent

  • length of abstinence: ≥ 2 weeks

Craving for alcohol:

  • OCDS score (mean ± SD): tianeptine = 18.2 ± 7.3; fluvoxamine = 18.1 ± 7.7

Anxiety:

  • HRSA score (mean ± SD): tianeptine = 19.2 ± 6.2; fluvoxamine = 19.5 ± 6.9

Other psychiatric comorbidity: information not available

Other substance use disorders: information not available

Other characteristics of study

Other pharmacological treatment: other pharmacological treatments were not allowed.

Funding source: information not available

Declarations of interest: information not available

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information to permit judgement.

Allocation concealment (selection bias)

Unclear risk

Method of concealment was not described.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Blinding of participants and key study personnel ensured. Both drugs were blinded to participants.

Blinding of outcome assessment (detection bias) objective

Low risk

No information on the blinding of outcome assessors.

Blinding of outcome assessment (detection bias) subjective

Unclear risk

No information on the blinding of outcome assessors.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only full analysis set were evaluated by the present meta‐analysis.

Selective reporting (reporting bias)

Low risk

Only full analysis set were evaluated by the present meta‐analysis.

Hernandez‐Avila 2004

Methods

Randomized, double‐blind, placebo‐controlled trial

Participants

41 depressed people with alcohol dependence (20 men and 21 women; age (mean ± SD): 42.9 ± 8.6 years)

Inclusion criteria:

  • current DSM‐IV diagnoses of alcohol dependence and depression

  • HRSD score ≥ 17 with a score ≥ 1 on item 1

  • aged 21‐65 years

Exclusion criteria:

  • history of major medical or psychiatric problems other than major depression or an anxiety disorder

  • clinically significant baseline laboratory abnormalities or a positive pregnancy test

  • current DSM‐IV diagnosis of drug dependence other than for alcohol or nicotine

  • positive urine drug screen

  • being treated with disulfiram or naltrexone or with any psychotropic drug

  • being at serious suicide risk

Participants with bipolar disorder were excluded.

Interventions

Drugs:

  • nefazodone (200‐600 mg/day; 21 participants; 10 men and 11 women; age (mean ± SD): 43.1 ± 9.0 years)

  • placebo (20 participants; 10 men and 10 women; age (mean ± SD): 42.7 ± 8.4 years)

Psychotherapy:

  • participants received manual‐guided supportive psychotherapy at each study visit

Scheduled duration of treatment: 10 weeks

Site: Alcohol Research Center, Department of Psychiatry, University of Connecticut School of Medicine, Farmington, CT, USA

Setting: outpatients

Route of administration: orally

Starting dose:

  • 100 mg twice daily

  • then titrated up to a maximum dose of 300 mg twice daily

Pattern of dose reduction: information not available

Outcomes

Depression:

  • final HRSD score

Alcohol dependence:

  • rate of abstinent days (obtained from weekly drinking days)

  • number of abstinent participants

  • number of drinking days per week

  • number of drinks per week

  • number of heavy drinking days per week

  • final DrinC score

  • final GGT levels

Anxiety:

  • final STAI score

Sleep quality:

  • final PSQI score

Dropouts

Adverse effects

Notes

Baseline characteristics of participants

Depression:

  • primary depression: information not available

  • duration: information not available

  • HRSD score (mean ± SD): nefazodone = 16.3 ± 2.3; placebo = 17.3 ± 2.0

Alcohol dependence:

  • drinks per drinking days (mean ± SD): nefazodone = 6.4 ± 6.5; placebo = 8.4 ± 5.2

  • duration: information not available

  • being actively drinking: participants were not abstinent alcohol (their consumption had to be a mean ≥ 18 drinks per week for men or 14 drinks per week for women; heavy drinking (≥ 5 drinks for men and ≥ 4 drinks for women) on at least 1 day/week during the month preceding screening)

  • length of abstinence at entry: 0 weeks

Anxiety:

  • STAI score (mean ± SD): nefazodone = 51.1 ± 9.9; placebo = 47.9 ± 9.4

Quality of sleep

  • PSQI score (mean ± SD): nefazodone = 22.2 ± 4.5; placebo = 22.1 ± 3.9

Other psychiatric comorbidity: participants with other mental disorders were included.

Other substance use disorders: participants with substance use disorders were excluded.

Other characteristics of study

Other pharmacological treatment offered: other pharmacological treatments were not allowed.

Funding sources: study supported by NIH Grants P50‐AA03510, K24‐AA13736, and M01‐RR06192 and the Bristol‐Myers Squibb Co.

Declarations of interest: information not available

Other information

After a baseline assessment, participants entered a 1‐week single‐blind placebo treatment, followed by random assignment to nefazodone or placebo groups.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Assignment to treatment groups used an urn randomization procedure, which balanced group assignment on sex, age, educational level, percentage of heavy drinking days, and severity of depressive symptoms at the time of the initial assessment.

Allocation concealment (selection bias)

Low risk

Method of concealment not reported but unlikely that selection bias was introduced.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Blinding of participants and key study personnel ensured.

Blinding of outcome assessment (detection bias) objective

Low risk

No information provided on blinding of assessors.

Blinding of outcome assessment (detection bias) subjective

Unclear risk

No information provided on blinding of assessors.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Mixed model analysis used to examine variables measured at each visit during study. This procedure allows the inclusion of all cases (41 participants) by estimating individual trajectories even when other data points are missing because of participant attrition.

Selective reporting (reporting bias)

Low risk

All expected outcomes were reported.

Kranzler 2006 arm A

Methods

Randomized, double‐blind, placebo‐controlled trial

Participants

189 depressed people with alcohol dependence (123 men; 66 women; age (mean ± SD): placebo = 44.0 ± 8.0 years; sertraline = 41.7 ± 9.4 years)

Inclusion criteria:

  • aged 21‐65 years

  • current DSM‐IV diagnoses of alcohol dependence and depression (modified: to meet DSM‐IV criteria for depression, except that symptoms could have occurred during a period of heavy alcohol use)

  • HRSD score ≥ 17

Exclusion criteria:

  • pregnant, nursing, or of childbearing potential not using an effective method of contraception

  • clinically significant co‐occurring psychiatric or medical diagnoses, including dependence on any psychoactive substance other than alcohol or nicotine during the preceding year

  • current treatment with disulfiram, naltrexone, or psychotropic medication

  • serum aminotransferase levels or other measures of hepatic function that were > 250% of normal

  • significant suicidal risk

Participants with bipolar disorder were excluded.

Interventions

Drugs:

  • sertraline (up to 200 mg/day; 89 participants)

  • placebo (100 participants)

Psychotherapy: at each study visit, participants received supportive therapy delivered according to a manual developed specifically for study consisting in:

  • general support for abstinence

  • promotion of compliance

  • monitoring of medication adverse effects

Scheduled duration of treatment: for up to 10 weeks

Sites: 13 investigative sites in USA

Setting: outpatients

Route of administration: orally

Starting dose: 50 mg/day

Pattern of dose reduction:

  • participants who achieved a satisfactory therapeutic response and who wished to continue treatment beyond the end of week 10 were continued double‐blind on the same medication they were taking at the end of week 10 for an additional 14‐week period;

  • participants who did not continue in the extension study were tapered off medication by reducing the daily dose by one capsule every 2 to 3 days until the medication was completely discontinued.

Outcomes

Depression:

  • final HRSD score (obtained from a figure)

  • difference between baseline and final HRSD score

  • response

Alcohol dependence:

  • rate of abstinent days (obtained from a figure)

Dropout

Adverse effects

Notes

Baseline characteristics of participants

Depression:

  • primary depression (rate of participants): 100%

  • duration: information not available

  • number of DSM‐IV criteria (mean ± SD): sertraline = 6.7 ± 1.0; placebo = 6.8 ± 1.2

  • HRSD score (mean ± SD): sertraline = 20.3 ± 2.8; placebo = 20.9 ± 4.0

Alcohol dependence:

  • number of DSM‐IV criteria (mean ± SD): sertraline = 5.6 ± 0.9; placebo = 5.5 ± 0.9

  • number of drinks per week (mean ± SD): sertraline = 45.9 ± 32.2; placebo = 63.1 ± 44.4

  • duration (mean ± SD): sertraline = 11.9 ± 9.0 years; placebo = 11.9 ± 9.9 years

  • being actively drinking: participants had to have drunk a mean of 18 drinks weekly for men or 14 drinks weekly for women and at least 1 heavy drinking day per week during the month before screening

  • length of abstinence: at least 4 days with no heavy drinking and no more than 16 days of abstinence

Other psychiatric comorbidity: participants with other mental disorders were excluded.

Other substance use disorders: participants with substance use disorders were excluded.

Other characteristics of study

Other pharmacological treatment offered: other pharmacological treatments were not allowed.

Funding source: supported by Pfizer Pharmaceuticals. Manuscript preparation supported by NIH grant K24 AA13736.

Declarations of interest: information not available.

Other information

In the original study, 328 participants were divided into the 2 groups on whether initially elevated HRSD score declined with cessation of heavy drinking:

  • HRSD score ≥ 17 (189 participants);

  • HRSD score ≤ 16 (139 participants).

In the present meta‐analysis, participants were divided into 2 substudies:

Both the substudies (Kranzler 2006 arm A; Kranzler 2006 arm B) were included in the 'Effects of interventions: Antidepressants versus placebo' comparison. Unfortunately, the original study did not report the adverse events for the single substudies.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomization schedule.

Allocation concealment (selection bias)

Unclear risk

Assignment to medication group was done according to a computer‐generated randomization schedule for groups A and B, with the medication groups within each stratum balanced for recent outpatient/inpatient status.

Despite random assignment, participants who received placebo were older, reported more drinks per week during the pretreatment period, and had higher CGI depression scores at baseline.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Insufficient information to permit judgement.

Blinding of outcome assessment (detection bias) objective

Low risk

No information on the blinding of outcome assessors.

Blinding of outcome assessment (detection bias) subjective

Unclear risk

No information on the blinding of outcome assessors.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All analyses used an intention‐to‐treat approach.

Selective reporting (reporting bias)

Low risk

All expected outcomes were reported. However, some of them were reported only as a % reduction (e.g. BDI score).

Kranzler 2006 arm B

Methods

Randomized, double‐blind, placebo‐controlled trial

Participants

139 depressed people with alcohol dependence (86 men and 53 women; age (mean ± SD): placebo = 42.9 ± 9.2 years; sertraline = 41.8 ± 9.4 years)

Inclusion criteria:

  • aged 21‐65 years

  • current DSM‐IV diagnoses of alcohol dependence and depression (modified: to meet DSM‐IV criteria for depression, except that symptoms could have occurred during a period of heavy alcohol use)

  • HRSD score ≤ 16 with cessation of heavy drinking

Exclusion criteria:

  • pregnant, nursing, or women of childbearing potential not using an effective method of contraception

  • clinically significant co‐occurring psychiatric or medical diagnoses

  • including dependence on any psychoactive substance other than alcohol or nicotine during the preceding year

  • current treatment with disulfiram, naltrexone, or psychotropic medication

  • serum aminotransferase levels or other measures of hepatic function > 250% of normal

  • significant suicidal risk

Participants with bipolar disorder were excluded.

Interventions

Drugs:

  • sertraline (up to 200 mg/day; 70 participants)

  • placebo (69 participants)

Psychotherapy:

At each study visit, participants received supportive therapy delivered according to a manual developed specifically for study consisting of:

  • general support for abstinence;

  • promotion of compliance;

  • monitoring of medication adverse effects.

Scheduled duration of treatment: up to 10 weeks

Sites: 13 investigative sites in the USA

Setting: outpatients

Route of administration: orally

Starting dose: 50 mg/day

Pattern of dose reduction:

  • participants who achieved a satisfactory therapeutic response and who wished to continue treatment beyond the end of week 10 were continued double‐blind on the same medication they were taking at the end of week 10 for an additional 14‐week period;

  • participants who did not continue in the extension study were tapered off medication by reducing the daily dose by 1 capsule every 2 or 3 days until the medication was completely discontinued.

Outcomes

Depression:

  • final HRSD score (obtained from a figure)

  • difference between baseline and final HRSD score

  • response

Alcohol dependence:

  • rate of abstinent days (obtained from a figure)

Dropout

Adverse effects

Notes

Baseline characteristics of participants

Depression:

  • primary depression (rate of participants): 0%

  • duration: information not available

  • number of DSM‐IV criteria (mean ± SD): sertraline = 5.3 ± 1.3; placebo = 5.4 ± 1.1

  • HRSD score (mean ± SD): sertraline = 12.6 ± 2.8; placebo = 12.5 ± 2.9

Alcohol dependence:

  • number of DSM‐IV criteria (mean ± SD): sertraline = 4.6 ± 1.2; placebo = 4.5 ± 1.0

  • number of drinks per week (mean ± SD): sertraline = 54.4 ± 40.5; placebo = 46.8 ± 27.9

  • duration (mean ± SD): sertraline = 10.7 ± 8.1 years; placebo = 11.1 ± 8.5 years

  • being actively drinking: participants had to have drunk a mean of 18 drinks weekly for men or 14 drinks weekly for women and at least 1 heavy drinking day per week during the month before screening

  • length of abstinence: at least 4 days with no heavy drinking and no more than 16 days of abstinence

Other psychiatric comorbidity: participants with other mental disorders were excluded.

Other substance use disorders: participants with substance use disorders were excluded.

Other characteristics of study

Other pharmacological treatment offered: other pharmacological treatments were not allowed.

Funding source: study supported by Pfizer Pharmaceuticals. Manuscript preparation supported by NIH grant K24 AA13736.

Declarations of interest: information not available

Other information

In the original study, 328 participants were divided into the 2 groups on whether initially elevated HRSD score declined with cessation of heavy drinking:

  • HRSD score ≥ 17 (189 participants);

  • HRSD score ≤ 16 (139 participants).

In the present meta‐analysis, participants were divided into 2 substudies:

Both the substudies (Kranzler 2006 arm A; Kranzler 2006 arm B) were included in the 'Effects of interventions: Antidepressants versus placebo' comparison. Unfortunately, the original study did not report the adverse events for the single substudies.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomization schedule reported.

Allocation concealment (selection bias)

Unclear risk

The method of concealment was not described. Despite random assignment, participants who received placebo were older, reported more drinks per week during the pretreatment period, and had higher CGI depression scores at baseline.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No information provided.

Blinding of outcome assessment (detection bias) objective

Low risk

No information on the blinding of outcome assessors.

Blinding of outcome assessment (detection bias) subjective

Unclear risk

No information on the blinding of outcome assessors.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All analyses used an intention‐to‐treat approach. Weekly comparisons including only participants for whom data were available from that visit, whereas end of study analyses used last observation carried forward analysis.

Selective reporting (reporting bias)

Low risk

All expected outcomes were reported. However, some of them were reported only as a % reduction (e.g. BDI score).

Krupitsky 1993 arm A

Methods

Randomized, placebo‐controlled trial

Participants

41 people with alcohol dependence (number of men and women not available; mean age = 36‐37 years) with non‐severe affective disorders

Inclusion criteria: information not available

Exclusion criteria: information not available

Participants with bipolar disorder: information not available

Interventions

Drugs:

  • amitriptyline (dose: 75 mg/day; 18 participants; number of men and women not available; age (mean ± SD): 36.3 ± 1.9 years)

  • placebo (23 participants; number of men and women not available; age (mean ± SD): 37.3 ± 1.7 years)

Psychotherapy: information not available

Scheduled duration of treatment: 3 weeks

Site: Russia

Setting: inpatients

Route of administration: orally

Starting dose: information not available

Pattern of dose reduction: information not available

Outcomes

Depression:

  • final ZUNG score

  • final MMPI score

Alcohol dependence: no information provided

Anxiety:

  • final STAI score

  • final MMPI score

Dropouts: data not available

Adverse effects: information not provided

Notes

Baseline characteristics of participants

Depression:

  • primary depression (rate of participants): 100%

  • duration: information not available

  • ZUNG score (mean ± SD): amitriptyline = 55.6 ± 1.2; placebo = 55.3 ± 1.0

  • MMPI score (mean ± SD): amitriptyline = 82.1 ± 1.5; placebo = 81.3 ± 3.9

Alcohol dependence:

  • severity: information not available

  • duration: information not available

  • participants were abstinent for at least 3‐4 weeks

Anxiety:

  • STAI score (mean ± SD): amitriptyline = 51.9 ± 2.6; placebo = 52.5 ± 2.9

  • MMPI score (mean ± SD): amitriptyline = 25.5 ± 1.9; placebo = 28.0 ± 2.0

Other psychiatric comorbidity: information not available

Other substance use disorders: information not available

Other characteristics of study

Other pharmacological treatment offered: other treatments were not administrated during study

Funding sources: information not available.

Other information

In the original study, 90 people with alcohol dependence were randomly divided into 4 groups:

  • baclofen (37.5 mg/day, 29 participants)

  • diazepam (15 mg/day; 20 participants)

  • amitriptyline (75 mg/day, 18 participants)

  • placebo (23 participants)

In the present meta‐analysis, study was divided into 2 substudies:

The first substudy (Krupitsky 1993 arm A) was included in the 'Effects of interventions: Antidepressants versus placebo' comparison and the second substudy (Krupitsky 1993 arm B) in the 'Antidepressants versus other medications' comparison

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information about the sequence generation process to permit judgement.

Allocation concealment (selection bias)

Unclear risk

Method of concealment not described.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Not reported if it was a double‐ or single‐blind study.

Blinding of outcome assessment (detection bias) objective

Low risk

Not reported if it was a double‐ or single‐blind study.

Blinding of outcome assessment (detection bias) subjective

High risk

Not reported if it was a double‐ or single‐blind study.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Number of dropouts not reported.

Selective reporting (reporting bias)

Unclear risk

Insufficient information to permit judgement.

Krupitsky 1993 arm B

Methods

Randomized, controlled trial

Participants

38 people with alcohol dependence (number of men and women not available; mean age = 36‐37 years) with affective disorders not severe

Inclusion criteria: information not available

Exclusion criteria: information not available

Participants with bipolar disorder: information not available

Interventions

Drugs:

  • amitriptyline (75 mg/day; 18 participants; number of men and women not available; age (mean ± SD): 36.3 ± 1.9 years)

  • diazepam (15 mg/day; 20 participants; number of men and women not available; age (mean ± SD): 38.3 ± 1.8 years)

Psychotherapy: information not available

Scheduled duration of treatment: 3 weeks

Site: Russia

Setting: inpatients

Route of administration: orally

Starting dose: information not available

Pattern of dose reduction: information not available

Outcomes

Depression:

  • final ZUNG score

  • final MMPI score

Alcohol dependence: no information provided

Anxiety:

  • final STAI score

  • final MMPI score

Dropouts: data not available

Adverse effects: data not available

Notes

Baseline characteristics of participants

Depression:

  • primary depression (rate of participants): 100%

  • duration: information not available

  • ZUNG score (mean ± SD): amitriptyline = 55.6 ± 1.2; diazepam = 54.2 ± 0.6

  • MMPI score (mean ± SD): amitriptyline = 82.1 ± 1.5; diazepam =81.1 ± 3.3

Alcohol dependence:

  • severity: information not available

  • duration: information not available

  • being actively drinking: participants were abstinent for at least 3‐4 weeks

Anxiety:

  • STAI score (mean ± SD): amitriptyline = 51.9 ± 2.6; diazepam = 51.8 ± 1.5

  • MMPI score (mean ± SD): amitriptyline = 25.5 ± 1.9; diazepam = 27.0 ± 1.7

Other psychiatric comorbidity: information not available

Other substance use disorders: information not available

Other characteristics of study

Other pharmacological treatment offered: other treatments were not administrated during study

Funding sources: information not available

Other information

In the original study, 90 people with alcohol dependence were randomly divided into 4 groups:

  • baclofen (37.5 mg/day, 29 participants)

  • diazepam (15 mg/day; 20 participants)

  • amitriptyline (75 mg/day, 18 participants)

  • placebo (23 participants)

In the present meta‐analysis, study was divided into 2 substudies:

The first substudy (Krupitsky 1993 arm A) was included in the 'Effects of interventions: Antidepressants versus placebo' comparison and the second substudy (Krupitsky 1993 arm B) in the 'Antidepressants versus other medications' comparison.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information about sequence generation process to permit judgement.

Allocation concealment (selection bias)

Unclear risk

Method of concealment not described.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Not reported if it was a double‐ or single‐blind study.

Blinding of outcome assessment (detection bias) objective

Low risk

Not reported if it was a double‐ or single‐blind study.

Blinding of outcome assessment (detection bias) subjective

High risk

Not reported if it was a double‐ or single‐blind study.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Number of dropouts not reported.

Selective reporting (reporting bias)

Unclear risk

Insufficient information to permit judgement.

Krupitsky 2012

Methods

Randomized, double‐blind, placebo‐controlled trial

Participants

60 depressed people with alcohol dependence (47 men and 13 women; age (mean ± SD): escitalopram = 43.9 ± 1.1 years; placebo = 40.9 ± 1.3 years).

Inclusion criteria:

  • current ICD‐10 diagnoses of alcohol dependence and affective disorders (a depression episode, a moderate depression episode, or a recurrent depression disorder)

  • HRSD score: 7‐23

Exclusion criteria:

  • substance dependence besides alcohol or nicotine dependence

  • bipolar‐affective disorder, schizophrenia, or other psychotic or organic mental disorders

  • other psychotropic drug

  • severe medical illnesses

  • pregnancy

Participants with bipolar disorder were excluded.

Interventions

Drugs:

  • escitalopram (10 mg/day; 29 participants; 22 men and 7 women)

  • placebo (31 participants, 25 men and 6 women)

Psychotherapy:

  • medical management which included elements of cognitive behavioural psychotherapy, once a week

Scheduled duration of treatment: 13 weeks

Site: a single‐site at the Department of Narcology (Addiction Psychiatry) of the Bekhterev PsychoNeurological Research Institute, St Petersburg, Russia

Setting: outpatients

Route of administration: orally

Starting dose: information not available

Pattern of dose reduction: information not available

Outcomes

Depression:

  • final HRSD score

  • final MADRS score

  • final ZUNG score

Alcohol dependence:

  • heavy drinkers

  • time to first relapse

  • final GGT levels

Craving for alcohol:

  • final PACS score

  • final OCDS score

  • final VAS score

Global response:

  • response

  • final GAF score

Anxiety:

  • final HRSA score

  • final STAI score

Dropouts

Adverse effects

Notes

Baseline characteristics of participants

Depression:

  • primary depression (rate of participants): information not available

  • duration: information not available

  • severity: mild or moderate depression

Alcohol dependence:

  • duration (mean ± SD): escitalopram = 11.5 ± 1.5 years; placebo = 9.5 ± 1.4 years

  • being actively drinking: participants had to be abstinent at least 7 days and had a negative alcohol test on expired air

  • length of abstinence: 1 week

Other psychiatric comorbidity: participants with other mental disorders were excluded.

Other substance use disorders: participants with substance use disorders were excluded.

Other characteristics of study

Other pharmacological treatment offered: other pharmacological treatments were not allowed.

Funding source: information not available.

Declarations of interest: information not available.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomization was performed by means of generation random numbers in Excel.

Allocation concealment (selection bias)

Low risk

Sequentially numbered drug containers of identical appearance.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The investigators, doctors, participants, and any other staff members taking part in the experiment were unaware which of the groups any particular person belonged to.

Blinding of outcome assessment (detection bias) objective

Low risk

No information on blinding of outcome assessors.

Blinding of outcome assessment (detection bias) subjective

Unclear risk

No information on blinding of outcome assessors.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Methods: participants who relapsed to heavy drinking were excluded from study.

Results: the relatively small number of alcohol consumption days in the groups was due to participants who relapsed being excluded from trial.

Selective reporting (reporting bias)

High risk

Methods: participants who relapsed to heavy drinking were excluded from study.

Results: the relatively small number of alcohol consumption days in the groups was due to participants who relapsed being excluded from trial.

Liappas 2005 arm A

Methods

Randomized, single‐blind, comparative trial

Participants

30 people with alcohol dependence of an original group constituted by 60 participants (41 men and 19 women; mean age: 47 years)

Inclusion criteria:

  • current DSM‐IV diagnosis of alcohol abuse or alcohol dependence

  • aged 18‐70 years

Exclusion criteria:

  • DSM‐IV diagnosis of primary depression; secondary depression was not an exclusion criterion

  • serious physical illness

  • other pre‐ or coexisting major psychiatric disorder, i.e. any psychotic disorder and bipolar disorder

  • other drug abuse, excluding nicotine

Participants with bipolar disorder were excluded.

Interventions

Treatment:

  • mirtazapine (30‐60 mg/day, in 1‐2 divided doses per day; 20 participants; 13 men and 7 women) and psychotherapy

  • only psychotherapy (10 participants)

Psychotherapy: cognitive behavioural psychotherapy administered in individual sessions and family interventions, twice a week

Scheduled duration of treatment: 3 weeks

Site: Drug and Alcohol Addiction Clinic, Athens University Psychiatric Clinic, Eginition Hospital, Athens, Greece

Setting: inpatients for 1 week, then residential treatment

Route of administration: orally

Starting dose: information not available

Pattern of dose reduction: information not available

Outcomes

Depression:

  • final HRSD score

Alcohol dependence: no information available

Anxiety:

  • final HRSA score

Global assessment:

  • final GAS score

Dropouts

Adverse effects: data not available

Notes

Baseline characteristics of participants

Depression:

  • primary depression (rate of participants): 0%

  • duration: information not available

  • HRSD score (mean ± SD): mirtazepine = 37.9 ± 7.8; controls = 39

Alcohol dependence:

  • drinks per drinking days (mean ± SD): mirtazepine = 27.6 ± 18.5; controls = 22.1,

  • duration (mean ± SD): mirtazepine = 15 years; controls = 14 years,

  • being actively drinking: participants were detoxicated before treatment,

  • length of abstinence: 1 week

Anxiety:

  • HRSA score (mean ± SD): mirtazapine = 33.2 ± 12.6; controls = 33.

Other psychiatric comorbidity: participants with other mental disorders were excluded.

Other substance‐use disorders: participants with substance use disorders were excluded.

Other characteristics of study

Other pharmacological treatment offered: other pharmacological treatments were not allowed.

Funding source: information not available

Declarations of interest: information not available

Other information

In the original study, 60 participants were included into 4 groups:

  • only psychotherapy (20 participants)

  • mirtazapine plus psychotherapy (20 participants)

  • venlafaxine plus psychotherapy (20 participants)

Participants but not clinicians were blind to group status

In the present meta‐analysis, we divided the control group (only psychotherapy) into 2 smaller groups, and compared these 2 smaller groups to the 2 antidepressants, using 3 subgroups:

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer random sequence generation was used ("At the end of the first week, individuals were randomly/electronically allocated to one of the three groups").

Allocation concealment (selection bias)

Low risk

Computer sequence of allocation used.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Single‐blind study ("Patients but not clinicians were blind to group status").

Blinding of outcome assessment (detection bias) objective

Low risk

Single‐blind study ("Patients but not clinicians were blind to group status").

Blinding of outcome assessment (detection bias) subjective

High risk

Single‐blind study ("Patients but not clinicians were blind to group status").

Incomplete outcome data (attrition bias)
All outcomes

High risk

Dropouts were not included in the analysis due to missing data.

Selective reporting (reporting bias)

Unclear risk

Insufficient information to permit judgement.

Liappas 2005 arm B

Methods

Randomized, single‐blind, comparative trial

Participants

30 people with alcohol dependence of an original group constituted by 60 participants (41 men and 19 women; mean age: 47 years)

Inclusion criteria:

  • current DSM‐IV diagnosis of alcohol abuse or alcohol dependence

  • aged 18‐70 years

Exclusion criteria:

  • DSM‐IV diagnosis of primary depression; secondary depression was not an exclusion criterion

  • serious physical illness

  • other pre‐ or coexisting major psychiatric disorder, i.e. any psychotic disorder and bipolar disorder

  • other drug abuse, excluding nicotine

Participants with bipolar disorder were excluded.

Interventions

Treatment:

  • venlafaxine (150‐300 mg/day, in 1‐2 divided doses per day; 20 participants; 13 men and 7 women) and psychotherapy

  • only psychotherapy (10 participants)

Psychotherapy: cognitive behavioural psychotherapy was administered in individual sessions and family interventions, twice a week

Scheduled duration of treatment: 3 weeks

Site: Drug and Alcohol Addiction Clinic, Athens University Psychiatric Clinic, Eginition Hospital, Athens, Greece

Setting: inpatients for 1 week, then residential treatment

Route of administration: orally

Starting dose: information not available

Pattern of dose reduction: information not available

Outcomes

Depression:

  • final HRSD score

Alcohol dependence: no information available

Anxiety:

  • final HRSA score

Global assessment:

  • final GAS score

Dropouts

Adverse effects: data not available

Notes

Baseline characteristics of participants

Depression:

  • primary depression (rate of participants): 0%

  • duration: information not available

  • HRSD score (mean ± SD): venlafaxine = 41.9 ± 4.5; controls = 39

Alcohol dependence:

  • drinks per drinking days (mean ± SD): venlafaxine = 18.4 ± 6.2; controls = 22.1

  • duration (mean ± SD): venlafaxine = 17 years; controls = 14 years

  • being actively drinking: participants were detoxicated before treatment

  • length of abstinence: 1 week

Anxiety:

  • HRSA score (mean ± SD): venlafaxine = 36.6 ± 5.4; controls = 33

Other psychiatric comorbidity: participants with other mental disorders were excluded.

Other substance‐use disorders: participants with substance use disorders were excluded.

Other characteristics of study

Other pharmacological treatment offered: other pharmacological treatments were not allowed.

Funding source: information not available

Declarations of interest: information not available

Other information

In the original study, 60 participants were included into 4 groups:

  • only psychotherapy (20 participants)

  • mirtazapine plus psychotherapy (20 participants)

  • venlafaxine plus psychotherapy (20 participants)

Participants but not clinicians were blind to group status

In the present meta‐analysis, we divided the control group (only psychotherapy) into 2 smaller groups, and compared these 2 smaller groups to the 2 antidepressants, using 3 subgroups:

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer random sequence generation reported ("At the end of the first week, individuals were randomly/electronically allocated to one of the three groups")

Allocation concealment (selection bias)

Low risk

Computer sequence of allocation reported.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Single‐blind study ("Patients but not clinicians were blind to group status").

Blinding of outcome assessment (detection bias) objective

Low risk

Single‐blind study ("Patients but not clinicians were blind to group status").

Blinding of outcome assessment (detection bias) subjective

High risk

Single‐blind study ("Patients but not clinicians were blind to group status").

Incomplete outcome data (attrition bias)
All outcomes

High risk

Dropouts were not included in the analysis due to missing data.

Selective reporting (reporting bias)

Unclear risk

Insufficient information to permit judgement.

Liappas 2005 arm C

Methods

Randomized, single‐blind, comparative trial

Participants

40 people with alcohol dependence of an original group constituted by 60 participants (41 men and 19 women; mean age: 47 years)

Inclusion criteria:

  • current DSM‐IV diagnosis of alcohol abuse or alcohol dependence

  • aged 18‐70 years

Exclusion criteria:

  • DSM‐IV diagnosis of primary depression; secondary depression was not an exclusion criterion

  • serious physical illness

  • other pre‐ or coexisting major psychiatric disorder, i.e. any psychotic disorder and bipolar disorder

  • other drug abuse, excluding nicotine

Participants with bipolar disorder were excluded.

Interventions

Drugs:

  • mirtazapine (30‐60 mg/day, in 1‐2 divided doses per day, 20 participants; 13 men and 7 women) and psychotherapy

  • venlafaxine (150‐300 mg/day, in 1‐2 divided doses per day; 20 participants; 13 men and 7 women) and psychotherapy

Psychotherapy: cognitive behavioural psychotherapy was administered in individual sessions and family interventions, twice a week.

Scheduled duration of treatment: 3 weeks

Site: Drug and Alcohol Addiction Clinic, Athens University Psychiatric Clinic, Eginition Hospital, Athens, Greece

Setting: inpatients for 1 week, then residential treatment

Route of administration: orally

Starting dose: information not available

Pattern of dose reduction: information not available

Outcomes

Depression:

  • final HRSD score

Alcohol dependence: no information available

Anxiety:

  • final HRSA score

Global assessment:

  • final GAS score

Dropouts

Adverse effects: data not available

Notes

Baseline characteristics of participants

Depression:

  • primary depression (rate of participants): 0%

  • duration: information not available

  • HRSD score (mean ± SD): mirtazepine = 37.9 ± 7.8; venlafaxine = 41.9 ± 4.5

Alcohol dependence:

  • drinks per drinking days: mirtazepine = 27.6 ± 18.5; venlafaxine = 18.4 ± 6.2

  • duration: venlafaxine = 17 years; controls = 14 years

  • being actively drinking: participants were detoxicated before treatment

  • length of abstinence: 1 week

Anxiety:

  • HRSA score (mean ± SD): mirtazapine: 33.2 ± 12.6; venlafaxine = 36.6 ± 5.4

Other psychiatric comorbidity: participants with other mental disorders were excluded.

Other substance use disorders: participants with substance use disorders were excluded.

Other characteristics of study

Other pharmacological treatment offered: other pharmacological treatments were not allowed.

Funding source: information not available

Declarations of interest: information not available

Other information

In the original study, 60 participants were included into 4 groups:

  • only psychotherapy (20 participants)

  • mirtazapine plus psychotherapy (20 participants)

  • venlafaxine plus psychotherapy (20 participants)

Participants but not clinicians were blind to group status.

In the present meta‐analysis, we divided the control group (only psychotherapy) into 2 smaller groups, and compared these 2 smaller groups to the 2 antidepressants, using 3 subgroups:

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer random sequence generation reported ("At the end of the first week, individuals were randomly/electronically allocated to one of the three groups")

Allocation concealment (selection bias)

Low risk

Computer sequence of allocation reported.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Single‐blind study, and the outcomes are likely to be influenced by lack of blinding ("Patients but not clinicians were blind to group status").

Blinding of outcome assessment (detection bias) objective

Low risk

Single‐blind study, and the outcomes are likely to be influenced by lack of blinding ("Patients but not clinicians were blind to group status").

Blinding of outcome assessment (detection bias) subjective

High risk

Single‐blind study, and the outcomes are likely to be influenced by lack of blinding ("Patients but not clinicians were blind to group status").

Incomplete outcome data (attrition bias)
All outcomes

High risk

Dropouts were not included in the analysis due to missing data.

Selective reporting (reporting bias)

Unclear risk

Insufficient information to permit judgement.

Lôo 1988

Methods

Randomized, double‐blind, comparative trial

Participants

129 people with alcohol dependence with depression or dysthymic disorder (111 men and 18 women; mean age: approximately 38 years)

Inclusion criteria:

  • DSM‐III diagnoses of alcohol dependence or alcohol abuse and depression or dysthymia

  • MADRS score ≥ 20

Exclusion criteria:

  • persistent alcohol intoxication at time of trial

  • presence of psychotic traits

  • recent antidepressant or neuroleptic treatment

  • serious somatic illness

  • pregnancy or absence of contraception in a woman of childbearing potential

Participants with bipolar disorder: information not available

Interventions

Drugs:

  • tianeptine (37.5 mg/day; 64 participants; 57 men and 7 women; age (mean ± SD): 37.9 ± 1.0 years)

  • amitriptyline (75.0 mg/day; 65 participants; 54 men and 11 women)

Psychotherapy: information not available

Scheduled duration of treatment: 4‐8 weeks (depending on the centre concerned)

Sites: 7 centres, in France

Setting: unclear

Route of administration: orally

Starting dose: tianeptine = 37.5 mg/day; amitriptyline = 75.0 mg/day

Pattern of dose reduction: information not available

Outcomes

Depression:

  • final MADRS score (obtained from a figure)

  • final SCL‐90 (obtained from a figure)

  • response

Alcohol dependence: data not available

Anxiety:

  • final HRSA score (obtained from a figure)

Dropouts

Adverse effects

Notes

Baseline characteristics of participants

Depression:

  • primary depression (rate of participants): 100%

  • duration: information not available

  • MADRS score (mean ± SD): tianeptine = 30.0 ± 0.8; amitriptyline = 29.3 ± 0.8

Alcohol dependence:

  • duration: information not available

  • severity: information not available

  • being actively drinking: participants were withdrawn from alcohol 2‐5 weeks before inclusion

Other psychiatric comorbidity: information not available

Other substance use disorders: information not available

Other characteristics of study

Other pharmacological treatment offered: other pharmacological treatments were allowed.

Funding source: information not available

Declarations of interest: information not available

Other information

In the original study, the duration of the trial was 4‐8 weeks depending on the centre concerned. In the present meta‐analysis, only data of 4 weeks were analyzed.

Before the onset of the trial, participants received a pretreatment with a placebo for 3‐10 days to screen out placebo‐responder participants. After this period, participants received tianeptine or amitriptyline.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information to permit judgement.

Allocation concealment (selection bias)

Unclear risk

Method of concealment not described.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind stated. Medication and placebo prepared to appear identical. No specific reference made to blinding of participants and personnel.

Blinding of outcome assessment (detection bias) objective

Low risk

No information on the blinding of outcome assessors.

Blinding of outcome assessment (detection bias) subjective

Unclear risk

No information on the blinding of outcome assessors.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Intention‐to‐treat analysis not used. However, the numbers of dropouts were low (10/64, 12/65) and were not unbalanced between groups.

Selective reporting (reporting bias)

High risk

Several data were missing (final MADRS score for amitriptyline, adverse effects, and alcohol consumption).

Mason 1996

Methods

Randomized, double‐blind, placebo‐controlled trial

Participants

28 depressed people with alcohol dependence (24 men and 4 women; mean age = desimipramine: 36.0 ± 22 years; placebo = 41.0 ± 15.5 years)

Inclusion criteria:

  • current DSM‐III‐R diagnoses of alcohol dependence and depression

  • secondary depression

  • aged 18‐65 years

Exclusion criteria:

  • primary depression

  • instable medical illnesses

  • pregnancy

  • psychosis

  • suicidal ideation

  • cognitively impairment

  • dependence on any substance except alcohol or nicotine

Participants with bipolar disorder: information not available

Interventions

Drugs:

  • desimipramine (200 mg/day; 15 participants; 13 men and 2 women)

  • placebo (13 participants; 11 men and 2 women)

Psychotherapy: participants were encouraged to participate in Alcoholics Anonymous and any other psychosocial treatments.

Scheduled duration of treatment: 6 months

Sites: Department of the New York (NY) Hospital‐Cornell Medical Center, and the University of Miami (FL), School of Medicine, USA

Setting: outpatients

Route of administration: orally

Starting dose: medication was prescribed in divided doses for the first week, then changed to bedtime dosing

Pattern of dose reduction: information not available

Outcomes

Depression:

  • final HRSD score

  • difference between baseline and final HRSD score

  • response

Alcohol dependence:

  • number of heavy drinkers (obtained from a figure)

Dropouts

Adverse effects

Notes

Baseline characteristics of participants

Depression:

  • primary depression (rate of participants): 0%

  • HRSD score (mean ± SD): desimipramine = 20.5 ± 9.7; placebo = 19.0 ± 11.0

  • family history of depression: desimipramine = 41%; placebo = 83%

Alcohol dependence:

  • ADS score (mean ± SD): desimipramine = 23.5 ± 10.5; placebo = 23.0 ± 13.0

  • duration: information not available

  • being actively drinking: participants had to be abstinent (maximum = 3 months; minimum = 1 week)

  • number of drinks per drinking days (mean ± SD): desimipramine = 15.0 ± 15.0; placebo = 13.0 ± 6.7

  • family history of alcohol dependence: desimipramine = 66%; placebo = 66%

Other psychiatric comorbidity: participants with other mental disorders were excluded.

Other substance use disorders: participants with substance use disorders were excluded.

Other characteristics of study

Other pharmacological treatment offered: other pharmacological treatments were not allowed.

Funding source: grants from the National Institute on Alcohol Abuse and Alcoholism (AA06866 and AA08111)

Declarations of interest: information not available

Other information

Final HRSD score and difference between baseline and final HRSD score were excluded because they were expressed as medians and interquartile ranges.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information about sequence generation process to permit judgement.

Allocation concealment (selection bias)

Unclear risk

Method of concealment was not described.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind stated and blinding of key study personnel ensured by sentences of the evaluation of plasma desipramine concentration. "Plasma desipramine concentration was assessed and results were reviewed by a physician not involved in patient ratings to verify compliance and make dose recommendations. Equivalent dosing instructions were given by the nonblinded physician to blinded therapists for placebo‐treated patients to preserve the double‐blind study design."

Blinding of outcome assessment (detection bias) objective

Low risk

No information on the blinding of outcome assessors.

Blinding of outcome assessment (detection bias) subjective

Unclear risk

No information on the blinding of outcome assessors.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Intention to treat was not used. Participants who relapsed, who demonstrated non‐compliance, or who did not improve were removed from study.

Selective reporting (reporting bias)

High risk

Participants who relapsed, who demonstrated non‐compliance, or who did not improve were removed from study.

McGrath 1996

Methods

Randomized, double‐blind, placebo‐controlled trial

Participants

69 depressed people with alcohol dependence (34 men and 35 women; age (mean ± SD): imipramine = 37.4 ± 6.7 years; placebo = information not available (report stated "10.6 ± 9.1")).

Inclusion criteria:

  • current DSM‐III‐R diagnoses of depression (or dysthymia, or depressive disorder not otherwise specified) and alcohol dependence (or abuse)

  • primary depression

  • aged 18‐65 years

Exclusion criteria:

  • history of mania, psychosis, seizure disorder, severe current physical dependence on alcohol requiring inpatient detoxification, abstinence of 2 weeks' duration at baseline, or for current serious and unstable physical illnesses

  • dependence on another substance, apart from nicotine, within the last 6 months

  • women not using adequate contraception

Participants with bipolar disorder were excluded.

Interventions

Drugs:

  • imipramine (50‐300 mg/day; 36 participants; 19 men and 17 women)

  • placebo (33 participants; 15 men and 18 women)

Psychotherapy:

  • weekly individual relapse prevention counselling sessions;

  • attendance at Alcoholics Anonymous was strongly encouraged.

Scheduled duration of treatment: 12 weeks

Site: Depression Evaluation Service, New York State Psychiatric Institute, New York, NY, USA

Setting: outpatients

Route of administration: orally

Starting dose:

  • 50 mg/day

  • increased by 50 mg every 3‐5 days to a maximum dose of 300 mg

Pattern of dose reduction: information not available

Outcomes

Depression:

  • final HRSD score

  • response

Alcohol dependence:

  • rate of abstinent days (obtained from the rate of drinking days)

  • number of abstinent participants

  • number of drinks per drinking days

  • number of heavy drinking days per week (obtained from the rate of heavy drinking days)

Global response

Dropouts

Adverse effects: data not available

Notes

Baseline characteristics of participants

Depression:

  • primary depression (rate of participants): 100%

  • duration: information not available

  • HRSD score (mean ± SD): imipramine = 15.4 ± 5.2; placebo = 14.3 ± 5.2

Alcohol dependence:

  • age of onset (mean ± SD): imipramine = 28.6 ± 15.2 years; placebo = 25.7 ± 9.2 years

  • being actively drinking: participants excluded if their abstinence ≥ 2 weeks

  • number of drinks per drinking days (mean ± SD): imipramine = 9.1 ± 6.5; placebo = 11.4 ± 13.7

Other psychiatric comorbidity: history of hypomania was not exclusionary.

Other substance‐use disorders: history of current abuse of other substances was not exclusionary, provided that alcohol was clearly the main substance of abuse.

Other characteristics of study

Other pharmacological treatment offered: information not available

Funding source: grants from the National Institute on Alcohol Abuse and Alcoholism (AA9539), the state of New York, and the Mental Health Clinical Research Center (NIMH 30906). Medications were supplied by Ciba‐Geigy Corp.

Declarations of interest: information not available

Other information

Eligible participants were given single‐blind placebo for 1 week. Participants whose depression was not rated 'much improved' or 'very much improved' on the improvement item of the CGI scale for depression were randomized to receive placebo or imipramine.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information about sequence generation process to permit judgement.

Allocation concealment (selection bias)

Unclear risk

Method of concealment not described.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Double‐blind stated and medications prepared to appear identical. No specific reference made to blinding of participants and personnel. Plasma dosage of imipramine performed but no information on blinding of personnel provided.

Blinding of outcome assessment (detection bias) objective

Low risk

No information on the blinding of outcome assessors.

Blinding of outcome assessment (detection bias) subjective

Unclear risk

No information on the blinding of outcome assessors.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Intention‐to‐treat analyses used. "Ratings from the last observation were carried forward for those subjects who did not complete all 12 weeks of treatment. Intention‐to‐treat analyses included all randomized patients and carried last observation forward for dropouts and those who completed less than 12 weeks."

Selective reporting (reporting bias)

High risk

Treatment outcomes were reported only for completers.

McLean 1986

Methods

Randomized, double‐blind, placebo‐controlled trial

Participants

35 people with alcohol dependence (number of men and women: not available; mean age: information not available)

Inclusion criteria:

  • people with alcohol dependence with inability to restrict alcohol consumption, alcohol craving, and tolerance

  • most of the features of alcohol dependence described in Edwards 1976

  • score in a self‐rating scale derived by HRSD ≥ 17 within 4 days of detoxification with benzodiazepines

Exclusion criteria:

  • pregnant or lactating women

  • psychosis, or a severe organic or mental disease

Participants with bipolar disorder: information not available

Interventions

Drugs:

  • mianserin (60 mg/day; 17 participants)

  • placebo (18 participants)

Psychotherapy:

  • group meetings

  • individual counselling

  • relaxation

  • occupational therapy

Scheduled duration of treatment: 4 weeks

Site: Alcoholism Treatment Unit, Mapperley Hospital, Nottingham, UK

Setting: inpatients

Route of administration: orally

Starting dose:

  • 30 mg/day for 7 days

  • then 60 mg/day for the next 3 weeks

Pattern of dose reduction:

  • after 4 weeks of treatment, drug was withdrawn

Outcomes

Depression:

  • final score in a self‐rating scale derived by HRSD

  • difference between final and initial score in a self‐rating scale derived by HRSD

  • response (evaluated using a self‐rating scale derived by HRSD)

Alcohol dependence: data not available

Dropouts

Adverse effects

Notes

Baseline characteristics of participants

Depression:

  • primary depression: information not available

  • duration: information not available

  • score in a self‐rating scale derived by HRSD (mean ± SD): 27 ± 6.1

Alcohol dependence:

  • severity: information not available

  • being actively drinking: participants were rated within 4 days of admission

  • length of abstinence: information not available.

Other psychiatric comorbidity: sociopathic personality disorder was present in 6 participants; 3 were considered to have significant anxiety; there were no diagnoses of schizophrenia or psychotic illness.

Other substance‐use disorders: information not available

Other characteristics of study

Other pharmacological treatment offered:

  • disulfiram (18 participants)

  • chlordiazepoxide (10 participants)

  • benzodiazepines (8 participants)

Funding source: Bencard.

Declarations of interest: information not available

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random allocation stated. No further details provided.

Allocation concealment (selection bias)

Unclear risk

Method of concealment not described.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind stated. No information on blinding of participants and personnel and on the appearance of medications.

Blinding of outcome assessment (detection bias) objective

Low risk

No information on the blinding of outcome assessors.

Blinding of outcome assessment (detection bias) subjective

Unclear risk

No information on the blinding of outcome assessors.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Intention‐to‐treat analysis not used. However, the number of dropouts was low and balanced between groups.

Selective reporting (reporting bias)

High risk

Data of participants who dropped out of study were not included in results.

Moak 2003

Methods

Randomized, placebo‐controlled trial

Participants

82 depressed people with alcohol dependence (50 men and 32 women; age (mean ± SD): sertraline = 41 ± 11 years; placebo = 42 ± 10 years)

Inclusion criteria:

  • current DSM‐III‐R diagnoses of depression or dysthymic disorder and alcohol dependence or abuse

  • HRSD ≥ 17, both at screening and at the end of 1 week of single‐blind placebo

  • minimum of 40 standard drinks during month before study entry

Exclusion criteria:

  • any current psychoactive substance dependence other than nicotine

  • psychoactive substance abuse in month before study entry other than marijuana

  • current panic disorder or post‐traumatic stress disorder

  • lifetime history of bipolar affective or psychotic disorder

  • evidence of treatment‐resistant depression

  • significant current suicidal ideation or plan, homicidal ideation, unstable medical illness, or history of a seizure disorder

Participants with bipolar disorder were excluded.

Interventions

Drugs:

  • sertraline (up to 200 mg/day; 38 participants)

  • placebo (44 participants)

Psychotherapy:

  • weekly individual CBT according to Project MATCH guidelines

Scheduled duration of treatment: 12 weeks

Site: Alcohol Research Center, Center for Drug and Alcohol Programs, Charleston, SC, USA

Setting: outpatients

Route of administration: orally

Starting dose: 50 mg/day

Pattern of dose reduction: titrated back down 50 mg over 7‐day period

Outcomes

Depression:

  • final HRSD score

  • final BDI score

  • response (calculated from significant depression)

Alcohol dependence:

  • rate of abstinent days

  • number of drinks per drinking days

  • number of heavy drinkers (calculated from a figure on % without relapse)

Dropout

Adverse effects

Notes

Baseline characteristics of participants

Depression:

  • primary depression (rate of participants): 85.4%

  • duration: information not available

  • HRSD score (mean ± SD): sertraline = 19.4 ± 2.6; placebo = 18.8 ± 2.4

Alcohol dependence:

  • number of drinks per drinking days (mean ± SD): sertraline = 11.3 ± 5.2; placebo = 10.5 ± 4.5

  • duration (mean ± SD): sertraline = 10.9 ± 8.0 years; placebo = 12.0 ± 8.6 years

  • being actively drinking: participants had to have drunk a minimum of 40 standard drinks during the month before

  • length of abstinence: not available

Other psychiatric comorbidity: participants with other mental disorders were excluded.

Other substance use disorders: participants with substance use disorders were excluded.

Other characteristics of study

Other pharmacological treatment offered: other pharmacological treatments were not allowed.

Funding source: National Institute on Alcohol Abuse and Alcoholism (grant AA10476). Pfizer supplied study drug and matched placebo.

Declarations of interest: information not available

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Urn randomization used.

Allocation concealment (selection bias)

Unclear risk

Method of concealment not provided.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, placebo‐controlled medication design applied. Medications dispensed in identically tablets. No further details provided on blinding of participants and personnel.

Blinding of outcome assessment (detection bias) objective

Low risk

No information provided on blinding of assessors.

Blinding of outcome assessment (detection bias) subjective

Unclear risk

No information provided on blinding of assessors.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Intention‐to‐treat analysis used.

Selective reporting (reporting bias)

Low risk

All expected outcomes reported.

Muhonen 2008

Methods

Randomized, double‐blind, comparative trial

Participants

80 depressed people with alcohol dependence (44 men and 36 women; age (mean ± SD): memantine = 47.5 ± 8.3 years; escitalopram = 47.9 ± 8.3 years)

Inclusion criteria:

  • aged 26‐65 years

  • current DSM‐IV diagnoses of alcohol dependence and depression

Exclusion criteria:

  • other substance use dependence

  • schizophrenia or other psychotic disorder and bipolar I and II disorder

  • acute risk of suicide

  • pregnant or breastfeeding

  • severe untreated somatic problem or a serious liver dysfunction

  • mental disability

Participants with bipolar disorder were excluded.

Interventions

Drugs:

  • escitalopram (20 mg/day; 40 participants)

  • memantine (20 mg/day; 40 participants)

Psychotherapy: no psychosocial intervention was offered.

Scheduled duration of treatment: 26 weeks (6 months)

Sites: 3 centres, Helsinki, Finland, and Europe.

Setting: outpatients

Route of administration: orally

Starting dose:

  • 5 mg/day

  • increased at weekly intervals by 5 mg/day to 20 mg/day for both drugs.

Pattern of dose reduction: information not available

Outcomes

Depression:

  • final MADRS score

  • final BDI score

  • response (participants reporting that their depression was reduced)

Alcohol dependence:

  • number of abstinent participants

Anxiety:

  • final HRSA score

  • final BAI score

Cognitive functioning:

  • final MMSE score

  • final score in a retrieval wordlist

Quality of life:

  • final VAS score

  • final SOFAS score

Dropouts

Adverse effects

Notes

Baseline characteristics of participants

Depression:

  • primary depression: information not available

  • duration (mean ± SD): memantine = 1.9 ± 2.5 years; escitalopram = 3.9 ± 5.6 years

  • MADRS score (mean ± SD): memantine = 25.8 ± 4.4; escitalopram = 26.8 ± 4.1

Alcohol dependence:

  • AUDIT score (mean ± SD): memantine = 27.4 ± 7.1; escitalopram = 28.4 ± 6.4

  • number of heavy drinking days per week (mean ± SD): memantine = 2.9 ± 1.1; escitalopram = 3.1 ± 1.0

  • duration: participants had a history of heavy drinking for at least 10 years

  • being actively drinking (rate of participants): memantine = 43.6%; escitalopram = 42.5%

  • length of abstinence: abstinence not required but encouraged

Other psychiatric comorbidity: participants with other mental disorders were excluded.

Other substance use disorders: participants with substance use disorders were excluded.

Other characteristics of study

Other pharmacological treatment offered: other medications prescribed by the patient's physician were allowed, except other antidepressants.

Funding source: National Public Health Institute, the Finnish Foundation for Alcohol Research and Helsinki Health Center Research. Study medication provided by Lundbeck Oy Ab, Turku, Finland

Declaration of interest: no conflicts of interest.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

All participants meeting the inclusion criteria were randomly assigned by an independent person to escitalopram or memantine groups using a 1:1 ratio and random permuted blocks (Vassar Statistics randomizing algorithm).

Allocation concealment (selection bias)

Low risk

Randomization was concealed until study database was locked on by an independent clinical study monitor.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Study medication was double‐dummy packed: participant took 2 tablets every time, 1 of which was the active medicine and 1 was an identical placebo for the second medication. The medication was labelled and controlled by an independent supplier.

Blinding of outcome assessment (detection bias) objective

Low risk

Outcome analysis was performed by an independent source.

Blinding of outcome assessment (detection bias) subjective

Low risk

Outcome analysis was performed by an independent source.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Intention‐to‐treat analysis used.

Selective reporting (reporting bias)

Low risk

Data of all randomized participants were reported except for 1 participant due to an interrupted interview.

Nunes 1993

Methods

Randomized, double‐blind, placebo‐controlled trial

Participants

26 depressed people with alcohol dependence (number of men and women: data not available; mean age: data not available)

Inclusion criteria:

  • current DSM‐III‐R diagnoses of depressive disorders (or dysthymia, or depressive disorder not otherwise specified) and alcohol dependence (or abuse)

Exclusion criteria: information not available

Participants with bipolar disorder: information not available

Interventions

Drugs:

  • imipramine (dose: information not available; 13 participants)

  • or placebo (10 participants)

Psychotherapy: information not available

Scheduled duration of treatment: 6 months

Site: Depression Evaluation Service, New York State Psychiatric Institute, New York, NY, USA

Setting: outpatients

Route of administration: orally

Starting dose:

  • participants completed a previous open label study in which received a mean dose of 263 mg/day

Pattern of dose reduction: information not available

Outcomes

Depression: data not available

Alcohol dependence: data not available

Global response

Dropouts: data not available

Adverse effects: data not available

Notes

Baseline characteristics of participants

Depression:

  • primary depression (rate of participants): 100%

  • duration: information not available

  • CGI score: 'much improved' or 'very much improved' after 1‐week of single‐blind placebo treatment

Alcohol dependence:

  • severity: information not available

  • being actively drinking: participants were abstinent

  • length of abstinence: approximately 12 weeks

Other psychiatric comorbidity: information not available.

Other substance use disorders: information not available.

Other characteristics of study

Other pharmacological treatment offered: information not available.

Funding sources: supported in part by training grant MH‐15144 from NIMH, grants AA‐07688 and AA‐08030 from the National Institute on Alcohol Abuse and Alcoholism, and Scientist Development Award for Clinicians DA‐00154 from the National Institute on Drug Abuse. CIBA/Geigy provided imipramine and matching placebo.

Declarations of interest: information not available

Other information

Only data of the double‐blind trial were included in the present meta‐analysis.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Information insufficient to permit judgement.

Allocation concealment (selection bias)

Unclear risk

Information insufficient to permit judgement.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Information insufficient to permit judgement.

Blinding of outcome assessment (detection bias) objective

Low risk

No information on the blinding of outcome assessors.

Blinding of outcome assessment (detection bias) subjective

Unclear risk

No information on the blinding of outcome assessors.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Information insufficient to permit judgement.

Selective reporting (reporting bias)

High risk

Not all of study's prespecified outcomes were reported.

Pettinati 2001a

Methods

Randomized, double‐blind, placebo‐controlled trial

Participants

29 depressed people with alcohol dependence (number of men and women: data not available; mean age: data not available)

Inclusion criteria:

  • aged ≥ 18 years

  • DSM‐III‐R diagnoses of depression and alcohol dependence

Exclusion criteria:

  • current substance dependence other than alcohol or nicotine

  • serious or unstable physical illness

  • bipolar illness, dementia, or psychosis

  • need for other psychotropic medications

Participants with bipolar disorder were excluded.

Interventions

Drugs:

  • sertraline (up to 200 mg/day; 12 participants)

  • placebo (17 participants)

Psychotherapy: weekly individual cognitive‐behavioural therapy

Scheduled duration of treatment: 14 weeks

Sites: University of Pennsylvania and the Carrier Foundation, USA

Setting: outpatients

Starting dose: 50 mg/day

Pattern of dose reduction: tapering during the last 2 weeks of treatment

Outcomes

Depression:

  • final HRSD score

  • final BDI score

Alcohol dependence:

  • rate of abstinent days (obtained from the rate of drinking days)

  • number of abstinent participants

  • time to first relapse

Dropouts: information not available

Adverse effects: information not available

Notes

Baseline characteristics of participants

Depression:

  • primary depression: information not available

  • severity: information not available

  • duration: information not available

Alcohol dependence:

  • severity: information not available

  • duration: information not available

  • being actively drinking: participants had to be actively drinking in the past 30 days, seeking treatment for alcohol problems

Other psychiatric comorbidity: participants with other mental disorders were excluded.

Other substance use disorders: participants with substance use disorders were excluded.

Other characteristics of study

Other pharmacological treatment offered: other pharmacological treatments were not allowed.

Funding source: National Institute on Alcohol Abuse and Alcoholism (R01‐AA09544 and KO2‐AA00239) and by Veteran Affairs Medical Center. Pfizer Inc. provided sertraline and matching placebo.

Declarations of interest: information not available

Other information

In the original study participants were divided into 2 groups:

  • never depressed (47 participants)

  • with lifetime depression (53 participants; 26 men and 27 women; age (mean ± SD): sertraline = 42.3 ± 9.3 years; placebo = 43.8 ± 10.9 years)

Participants with lifetime depression were then divided into 2 subgroups:

  • with current depression (29 participants)

  • with only lifetime diagnosis of depression (24 participants)

In the meta‐analysis, we included only participants with current depression. Unfortunately, dropouts and adverse events were not provided for each subgroup.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Sequence generation referred to 1 randomization schedule for both sites.

Allocation concealment (selection bias)

Unclear risk

Method of concealment was not reported.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind stated and medications prepared to appear identical. No specific reference made to blinding of participants and personnel.

Blinding of outcome assessment (detection bias) objective

Low risk

No information on the blinding of outcome assessors.

Blinding of outcome assessment (detection bias) subjective

Unclear risk

No information on the blinding of outcome assessors.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Intention‐to‐treat analysis used.

Selective reporting (reporting bias)

Low risk

All expected outcomes reported.

Pettinati 2010 arm A

Methods

Randomized, double‐blind, placebo‐controlled trial

Participants

79 depressed people with alcohol dependence (49 men and 30 women; age (mean ± SD): sertraline = 43.9 ± 11.5 years; placebo = 43.4 ± 8.9 years)

Inclusion criteria:

  • current DSM‐IV diagnoses of depression and alcohol dependence

  • HRSD score ≥ 10

Exclusion criteria:

  • substance dependence besides alcohol or nicotine dependence

  • bipolar‐affective disorder, schizophrenia, or other psychotic or organic mental disorders

  • regular use of antidepressants

  • requiring psychiatric medications other than an antidepressant

  • severe medical illness

  • pregnant or breastfeeding

Participants with bipolar disorder were excluded.

Interventions

Drugs:

  • sertraline (200 mg/day; 40 participants)

  • placebo (39 participants)

Psychotherapy: weekly individual CBT

Scheduled duration of treatment: 14 weeks

Site: University of Pennsylvania Treatment Research Center, USA

Setting: outpatients

Route of administration: orally

Starting dose: 50 mg/day

Pattern of dose reduction:

  • in week 14, sertraline was reduced to 100 mg/day;

  • medications were completed by the last treatment day.

Outcomes

Depression:

  • final HRSD score

  • remission

Alcohol dependence:

  • number of abstinent participants

  • number of heavy drinkers (calculated from the figure on the rates of subjects without a heavy drinking day)

  • time to relapse

Dropouts

Adverse effects

Notes

Baseline characteristics of participants

Depression:

  • primary depression (rate of participants): 100%

  • duration: information not available

  • HRSD score (mean ± SD): sertraline = 23.4 ± 6.0; placebo = 22.9 ± 7.0

Alcohol dependence:

  • number of drinks per drinking days (mean ± SD): sertraline = 12.4 ± 5.6; placebo = 10.5 ± 5.9

  • duration (mean ± SD): sertraline = 21.7 ± 10.6 years; placebo = 19.3 ± 10.1 years

  • being actively drinking: participants had to consume a mean of ≥12 alcoholic drinks per week and on ≥ 40% of the 90 days before treatment

  • length of abstinence: 3 days

Other psychiatric comorbidity: participants with other mental disorders were excluded.

Other substance use disorders: participants with substance use disorders were excluded.

Other characteristics of study

Other pharmacological treatment offered: other pharmacological treatments were not allowed.

Funding source: National Institute on Alcohol Abuse and Alcoholism (grant R01‐AA09544‐10). Pfizer Inc., USA, Pharmaceutical Group provided sertraline and matching placebo.

Declaration of interest: the authors declared the grants received.

Other information

In the original study participants were divided into 4 groups:

  • sertraline (40 participants)

  • naltrexone (49 participants)

  • sertraline plus naltrexone (42 participants)

  • double placebo (39 participants)

In the meta‐analysis, data were analyzed and included in 2 substudies:

Both the substudies (Pettinati 2010 arm A; Pettinati 2010 arm B) were included in the 'Effects of interventions: Antidepressants versus placebo' comparison.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Urn randomization reported.

Allocation concealment (selection bias)

Low risk

Urn randomization used to evenly distribute participants across groups using 4 pretreatment variables: gender, regular smoking, HRSD scores at time of randomization, and drinking frequency.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Blinding of participants and key study personnel ensured, and unlikely that the blinding could have been broken.

Blinding of outcome assessment (detection bias) objective

Low risk

No information on the blinding of outcome assessors.

Blinding of outcome assessment (detection bias) subjective

Unclear risk

No information on the blinding of outcome assessors.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing data were imputed using appropriate methods.

Selective reporting (reporting bias)

Low risk

All expected outcomes reported.

Pettinati 2010 arm B

Methods

Randomized, double‐blind, placebo‐controlled trial

Participants

91 depressed people with alcohol dependence (57 men and 34 women; age (mean ± SD): sertraline plus naltrexone = 43.4 ± 10.2 years; naltrexone = 42.9 ± 8.1)

Inclusion criteria:

  • current DSM‐IV diagnoses of depression and alcohol dependence

  • HRSD score ≥ 10

Exclusion criteria:

  • substance dependence besides alcohol or nicotine dependence

  • bipolar‐affective disorder, schizophrenia, or other psychotic or organic mental disorders

  • regular use of antidepressants

  • requiring psychiatric medications other than an antidepressant

  • severe medical illness

  • pregnant or breastfeeding

Participants with bipolar disorder were excluded.

Interventions

Drugs:

  • sertraline (200 mg/day) plus naltrexone (100 mg/day) (42 participants)

  • naltrexone (100 mg/day; 49 participants)

Psychotherapy: weekly individual CBT

Scheduled duration of treatment: 14 weeks

Site: University of Pennsylvania Treatment Research Center, USA

Setting: outpatients

Route of administration: orally

Starting doses:

  • naltrexone 50 mg/day for the first 4 days

  • naltrexone 100 mg/day for other 3 days

  • naltrexone 100 mg/day plus sertraline 50 mg/day added every third day to 200 mg/day

Pattern of dose reduction:

  • in week 13, naltrexone was reduced to 50 mg/day and sertraline maintained at 200 mg/day;

  • in week 14, naltrexone was continued at 50 mg/day and sertraline reduced to 100 mg/day;

  • medications were completed by the last treatment day.

Outcomes

Depression:

  • final HRSD score

  • remission

Alcohol dependence:

  • number of abstinent participants

  • number of heavy drinkers (calculated from the figure on the rates of subjects without a heavy drinking day)

  • time to relapse

Dropouts

Adverse effects

Notes

Baseline characteristics of participants

Depression:

  • primary depression (rate of participants): 100%

  • duration: information not available

  • HRSD score (mean ± SD): sertraline plus naltrexone = 23.7 ± 6.7; naltrexone = 22.3 ± 5.7

Alcohol dependence:

  • number of drinks per drinking days (mean ± SD): sertraline plus naltrexone = 12.8 ± 9.2; naltrexone = 13.6 ± 6.9

  • duration (mean ± SD): sertraline plus naltrexone = 22.2 ± 10.5 years; naltrexone = 21.3 ± 8.3 years

  • being actively drinking: participants had to consume a mean of ≥12 alcoholic drinks per week and on ≥ 40% of the 90 days before treatment

  • length of abstinence: 3 days

Other psychiatric comorbidity: participants with other mental disorders were excluded.

Other substance use disorders: participants with substance use disorders were excluded.

Other characteristics of study

Other pharmacological treatment offered: other pharmacological treatments were not allowed.

Funding source: National Institute on Alcohol Abuse and Alcoholism (grant R01‐AA09544‐10). Pfizer Inc., USA, Pharmaceutical Group provided sertraline and matching placebo.

Declaration of interest: the authors declared the grants received.

Other information

In the original study participants were divided into 4 groups:

  • sertraline (40 participants)

  • naltrexone (49 participants)

  • sertraline plus naltrexone (42 participants)

  • double placebo (39 participants)

In the meta‐analysis, data were analyzed and included in 2 substudies:

Both the substudies (Pettinati 2010 arm A; Pettinati 2010 arm B) were included in the 'Effects of interventions: Antidepressants versus placebo' comparison.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Urn randomization.

Allocation concealment (selection bias)

Low risk

Urn randomization used to evenly distribute participants across groups using 4 pretreatment variables: gender, regular smoking, HRSD scores at the time of randomization, and drinking frequency.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Blinding of participants and key study personnel ensured, and unlikely that the blinding could have been broken.

Blinding of outcome assessment (detection bias) objective

Low risk

No information on the blinding of outcome assessors.

Blinding of outcome assessment (detection bias) subjective

Unclear risk

No information on the blinding of outcome assessors.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing data were imputed using appropriate methods.

Selective reporting (reporting bias)

Low risk

All expected outcomes reported.

Roy 1998

Methods

Randomized, double‐blind, placebo‐controlled trial

Participants

36 depressed people with alcohol dependence (33 men and 3 women; age (mean ± SD): sertraline = 40.5 ± 7.7 years; placebo = 41.2 ± 5.6 years)

Inclusion criteria:

  • current DSM‐III‐R diagnoses of depression and alcohol dependence

Exclusion criteria:

  • schizophrenia, bipolar disorder, obsessive‐compulsive disorder, dementia, other DSM‐III‐R major Axis I disorder

  • non‐alcohol substance dependence, epilepsy, organic mental disorder, or significant medical disorder

Participants with bipolar disorder were excluded.

Interventions

Drugs:

  • sertraline (100 mg/day; 18 participants)

  • placebo (18 participants)

Psychotherapy: information not available

Scheduled duration of treatment: 6 weeks

Site: Alcoholic Rehabilitation Unit of the Veterans Administration Medical Center, East Orange, New Jersey, USA

Setting: inpatients for the first 2 weeks then outpatients for 6 weeks

Route of administration: orally

Starting dose: information not available

Pattern of dose reduction: information not available

Outcomes

Depression:

  • final HRSD score (calculated from a figure)

  • final BDI score (calculated from a figure)

  • response

Alcohol dependence: information not available

Dropouts

Adverse effects: information not available.

Notes

Baseline characteristics of participants

Depression:

  • primary depression (rate of participants): 41.7%

  • duration (mean ± SD): sertraline = 5.2 ± 5.3 months; placebo = 8.8 ± 9.0 months

  • HRSD score (mean ± SD): sertraline = 25.3 ± 7.3; placebo = 20.2 ± 5.6

Alcohol dependence:

  • severity: information not available

  • duration (mean ± SD): sertraline = 16.3 ± 9.1 years; placebo = 19.4 ± 6.8 years

  • being actively drinking: participants had to be abstinent at least 2 weeks

  • length of abstinence (mean ± SD): sertraline = 4.8 ± 7.5 weeks; placebo = 4.7 ± 4.8 weeks

Other psychiatric comorbidity: participants with other mental disorders were excluded.

Other substance use disorders: participants with substance use disorders were excluded.

Abuse of other substances was not an exclusion.

Other characteristics of study

Other pharmacological treatment offered: participants who had received antidepressant medication in the previous month were excluded.

Funding source: information not available

Declarations of interest: information not available

Other information

Final HRSD and BDI scores and number of withdrawal for medical reasons were provided separately for participants with primary depression (arm A; 15 participants) and participants with secondary depression (arm B; 21 participants). For the other outcomes, data were provided for the entire sample of participants.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information about sequence generation process to permit judgement.

Allocation concealment (selection bias)

Unclear risk

Method of concealment was not provided.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Blinding of participants and key study personnel Information ensured but information insufficient to permit judgement.

Blinding of outcome assessment (detection bias) objective

Low risk

No information on the blinding of outcome assessors.

Blinding of outcome assessment (detection bias) subjective

Unclear risk

No information on the blinding of outcome assessors.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Intention‐to‐treat analysis referred to, but for participants who did not complete the study (dropouts), the last observation was not carried forward.

Selective reporting (reporting bias)

Low risk

All expected outcomes reported.

Roy‐Byrne 2000

Methods

Randomized, double‐blind, placebo‐controlled trial

Participants

64 depressed people with alcohol dependence (29 men and 35 women; age (mean ± SD): 40.2 ± 8.2 years)

Inclusion criteria:

  • aged 18‐55 years

  • current DSM‐III‐R diagnoses of depression and alcohol dependence

Exclusion criteria:

  • intravenous drug use

  • other drug use more than once per week

  • schizophrenia and bipolar disorder

  • active suicidal ideation with a plan

  • recent history of delirium tremens or alcohol‐withdrawal seizures

  • current treatment for depression or alcoholism

  • serious medical problems

  • treatment with medications that are contraindicated in combination with nefazodone

  • pregnancy

  • untreated hypothyroidism or hyperthyroidism

  • clinically significant live dysfunction, active cardiac or renal impairment

  • homelessness

Participants with bipolar disorder were excluded.

Interventions

Drugs:

  • nefazodone (200‐500 mg/day; 32 participants; 17 men and 15 women; age (mean ± SD): 40.9 ± 8.6 years)

  • placebo (32 participants; 12 men and 20 women; age (mean ± SD): 39.5 ± 7.9 years)

Psychotherapy: cognitive‐behavioural skills training and psychoeducational group for alcohol‐dependence and depression led by an experienced therapist; group sessions lasted 1 hour once per week.

Scheduled duration of treatment: 12 weeks

Site: Harborview Medical Center Outpatient Psychiatry Clinic, University of Washington, Seattle, USA

Setting: outpatients

Route of administration: orally

Starting dose:

  • 100 mg twice daily

  • additional 100 mg/day per week until 200 mg in the morning and 300 mg at night (500 mg total)

Pattern of dose reduction: information not available

Outcomes

Depression:

  • final HRSD score (obtained from a figure)

  • response

Alcohol dependence:

  • number of abstinent participants

  • number of drinks per drinking day (obtained from a figure)

Dropouts

Adverse effects

Notes

Baseline characteristics of participants

Depression:

  • primary depression (rate of participants): 100%

  • duration: information not available

  • HRSD score (mean ± SD): nefazodone = 23.1 ± 5.8; placebo = 24.8 ± 4.5

Alcohol dependence:

  • number of drinks per drinking day (mean ± SD): nefazodone = 11.0 ± 10.5; placebo = 8.5 ± 10.1

  • duration: information not available

  • being actively drinking: participants were asked to decrease or discontinue their drinking before randomization, but only 9.5% stopped drinking

  • length of abstinence: 0 weeks

Other psychiatric comorbidity: participants with other mental disorders were included.

Other substance use disorders: participants with substance use disorders were excluded.

Other pharmacological treatment offered: other pharmacological treatments were not allowed.

Funding source: supported in part by Bristol‐Meyers Squibb.

Declarations of interest: information not available.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information about sequence generation process to permit judgement.

Allocation concealment (selection bias)

Unclear risk

Method of concealment was not provided.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Blinding of participants and key study personnel ensured.

Blinding of outcome assessment (detection bias) objective

Low risk

Blinding of outcome assessment ensured.

Blinding of outcome assessment (detection bias) subjective

Low risk

Blinding of outcome assessment ensured.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing data were imputed using appropriate methods.

Selective reporting (reporting bias)

Low risk

All expected outcomes reported.

ADS: Alcohol Dependence Scale; ALT: alanine aminotransferase; AST: aspartate aminotransferase; AUDIT: Alcohol Use Disorders Identification Test; BAI: Beck Anxiety Inventory; BDI: Beck Depression Inventory; BPRS: Brief Psychiatric Rating Scale; CBT: cognitive behavioural therapy; CGI: Clinical Global Impression scale; DBI: Drinking Behaviour Interview; DrInC: Drinker Inventory of Consequences scale; DOTES: Dosage Record and Treatment Emergent Symptoms Scale; DSM: Diagnostic and Statistic Manual of Mental Disorders; DSM‐III‐R: Diagnostic and Statistic Manual of Mental Disorders III ‐ Revised; DSM‐IV: Diagnostic and Statistic Manual of Mental Disorders ‐ IV; ECT: electroconvulsive therapy; GAF: General Assessment of Functioning scale; GAS: Global Assessment Scale; GGT: γ‐glutamyltransferase; HRSA: Hamilton Rating Scale for Anxiety; HRSD: Hamilton Rating Scale for Depression; ICD: International Classification of Diseases; LDQ: Leeds Dependence Questionnaire; LDRS: Lehmann Depression Rating Scale; MADRS: Montgomery and Åsberg Depression Rating Scale; MAST: Michigan Alcoholism Screening Test; MET: Motivational Enhancement Therapy; MMPI: Minnesota Multiphasic Personality Inventory; MMSE: Mini‐Mental State Examination; OCDS: Obsessive‐Compulsive Drinking Scale; PACS: Penn Alcohol Craving scale; PSQI: Pittsburgh Sleep Quality Index; SCL‐90: Symptom Check List‐90; SD: standard deviation; SOFAS: The Social and Occupational Functioning Assessment Scale; STAI: State Trait Anxiety Inventory; UKU: Udvalg for Kliniske Undersogelser Side Effect Rating Scale; VAS: Visual Analogue Scale; ZUNG: Zung Self‐Assessment Depression Scale.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Anthenelli 2014

Type of participants not in the inclusion criteria: no depression

Arnow 2015

Type of participants not in the inclusion criteria: no alcohol dependence

Balaratnasingam 2011

Data of single group not available

Bandati 2013

Study design not in the inclusion criteria: no control group

Batki 2015

Type of participants and type of intervention not in the inclusion criteria: no depression, no use of antidepressant medications

Bowman 1966

Type of intervention not in the inclusion criteria: no antidepressant medications used

Brewer 2015

Type of participants and type of intervention not in the inclusion criteria: no depression, no alcohol dependence; no antidepressant medications used

Brown 2003

Study design not in the inclusion criteria: no control group

Brunelin 2014

Type of participants and type of intervention not in the inclusion criteria: no alcohol dependence; no antidepressant medications used

Charney 2015

Type of participants not in the inclusion criteria: only 22% of participants had depression (single data of these participants not available)

Charnoff 1967

Type of participants not in the inclusion criteria: no depression

Chick 2004b

Type of participants not in the inclusion criteria: no depression

Clark 2003

Study population not in the inclusion criteria: people aged < 18 years

Cornelius 1993

Study design not in the inclusion criteria: no control group

Cornelius 2011

Type of participants not in the inclusion criteria: people aged < 18 years

Cornelius 2012

Study design not in the inclusion criteria: no control group

Davis 2005

Study design not in the inclusion criteria: no control group

Desai 1999

Study design not in the inclusion criteria: no randomised trial

Douglas 1996

Study design not in the inclusion criteria: commentary on Mason 1996

Eriksson 2001

Type of participants not in the inclusion criteria: no alcohol dependence. Only 73% of recruited participants with alcohol dependence; data of these participants not reported.

Farren 1999

Study design not in the inclusion criteria: case report

Foulds 2016

Commentary

García‐Portilla 2005

Study design not in the inclusion criteria: no control group

Glasner‐Edwards 2007

Type of intervention not in the inclusion criteria: the name of the antidepressant medication used was not indicated

Gorelick 1992

Type of participants not in the inclusion criteria: no alcohol dependence

Grelotti 2014

Type of participants not in the inclusion criteria: no alcohol dependence

Han 2013

Study design not in the inclusion criteria: no control group for the antidepressant. Study compared the efficacy of aripripazole plus escitalopram to escitalopram in reducing the severity of depression and craving for alcohol in people with alcohol dependence and depression.

Hautzinger 2005

Type of participants not in the inclusion criteria: no depression

Ionescu 2011

Lack of information: number of patients for each group; time of collected data (1 month, 3 months, or 6 months?)

Ivanets 1998

Type of participants not in the inclusion criteria: no depression

Janiri 1996

Type of participants not in the inclusion criteria: no depression

Janiri 1997

Type of participants not in the inclusion criteria: no depression

Kalyoncu 2007

Study design not in the inclusion criteria: no control group

Kranzler 1995

Type of participants not in the inclusion criteria: no alcohol dependence. Only 14% of recruited participants with alcohol dependence; data of these participants not reported.

Kranzler 2011

Type of participants not in the inclusion criteria: no depression

Krupitsky 2013

Data on severity of depression, craving for alcohol, and consumption of alcohol were reported to be improved but not provided as score achieved in the different scales used

Krystal 2008

Type of intervention not in the inclusion criteria: the name of antidepressant medications was not indicated.

Labbate 2004

Study design not in the inclusion criteria. No control group for the antidepressant. Study compared the efficacy of sertraline in people with post‐traumatic stress disorder and alcohol dependence with comorbid anxiety or depression to that in people with post‐traumatic stress disorder and alcohol dependence but without comorbid anxiety or depression.

Lee 2012

Study design not in the inclusion criteria: no control group for the antidepressant. Study compared the efficacy of aripripazole plus escitalopram to escitalopram in reducing the severity of depression and craving for alcohol in people with alcohol dependence and depression.

Liappas 2004

Type of participants not in the inclusion criteria: no depression

Macher 1991

Study design not in the inclusion criteria: no control group

Malka 1992

Study design not in the inclusion criteria: no control group

Marey 1991

Study design not in the inclusion criteria: review

Mason 1999

Study design not in the inclusion criteria: no control group for the antidepressant. Study compared the efficacy of naltrexone plus sertraline to sertraline in reducing the severity of depression and alcohol consumption in people with alcohol dependence and depression.

Naranjo 1997

Study design not in the inclusion criteria: statistical elaboration of previous studies

Oslin 2005

Study design not in the inclusion criteria: no control group for the antidepressant. Study compared the efficacy of naltrexone plus sertraline to sertraline in reducing the severity of depression and alcohol consumption in people with alcohol dependence and depression.

Overall 1973

Type of participants not in the inclusion criteria: no depression

Powell 1995

Type of participants not in the inclusion criteria: no depression

Ruiz‐Mellott 2005

Study design not in the inclusion criteria: no control group

Saatcioglu 2005

Study design not in the inclusion criteria: no control group

Salloum 2011

Study design not in the inclusion criteria: no control group

Salloum 2013

Study design not in the inclusion criteria: no control group

Schottenfeld 1989

Study design not in the inclusion criteria: no control group

Shaw 1975

Study design not in the inclusion criteria: no control group for the antidepressant. Study compared the efficacy of chlordiazepoxide plus imipramine to placebo.

Witte 2012

Study design not in the inclusion criteria: no control group for the antidepressant. Study compared the efficacy of acamprosate plus escitalopram to escitalopram in reducing the severity of depression and alcohol consumption in people with alcohol dependence and depression.

Characteristics of studies awaiting assessment [ordered by study ID]

Petrakis 2013

Methods

Requested from the authors

Participants

Interventions

Outcomes

Notes

Schifano 1993

Methods

Requested from the authors

Participants

Interventions

Outcomes

Notes

Data and analyses

Open in table viewer
Comparison 1. Antidepressants versus placebo: all studies

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Depression severity: final score (interviewer‐rated scales) Show forest plot

14

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.1

Comparison 1 Antidepressants versus placebo: all studies, Outcome 1 Depression severity: final score (interviewer‐rated scales).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 1 Depression severity: final score (interviewer‐rated scales).

1.1 All studies

14

1074

Std. Mean Difference (IV, Random, 95% CI)

‐0.27 [‐0.49, ‐0.04]

1.2 Selective serotonin reuptake inhibitors (SSRIs)

10

881

Std. Mean Difference (IV, Random, 95% CI)

‐0.17 [‐0.40, 0.07]

1.3 5‐HT2 antagonists

2

97

Std. Mean Difference (IV, Random, 95% CI)

‐0.29 [‐0.69, 0.11]

2 Depression severity: final score (self‐administered scales) Show forest plot

8

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.2

Comparison 1 Antidepressants versus placebo: all studies, Outcome 2 Depression severity: final score (self‐administered scales).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 2 Depression severity: final score (self‐administered scales).

2.1 All studies

8

373

Std. Mean Difference (IV, Random, 95% CI)

‐0.29 [‐0.64, 0.07]

2.2 SSRIs

5

300

Std. Mean Difference (IV, Random, 95% CI)

‐0.25 [‐0.69, 0.18]

2.3 5‐HT2 antagonists

2

41

Std. Mean Difference (IV, Random, 95% CI)

0.02 [‐0.59, 0.64]

3 Depression severity: difference between basal and final score (interviewer‐rated scales) Show forest plot

5

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.3

Comparison 1 Antidepressants versus placebo: all studies, Outcome 3 Depression severity: difference between basal and final score (interviewer‐rated scales).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 3 Depression severity: difference between basal and final score (interviewer‐rated scales).

3.1 All studies

5

447

Std. Mean Difference (IV, Random, 95% CI)

0.15 [‐0.12, 0.42]

3.2 SSRIs

4

408

Std. Mean Difference (IV, Random, 95% CI)

0.07 [‐0.18, 0.32]

4 Depression severity: difference between basal and final score (self‐administered scales) Show forest plot

4

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.4

Comparison 1 Antidepressants versus placebo: all studies, Outcome 4 Depression severity: difference between basal and final score (self‐administered scales).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 4 Depression severity: difference between basal and final score (self‐administered scales).

4.1 All studies

4

121

Std. Mean Difference (IV, Random, 95% CI)

0.20 [‐0.16, 0.56]

4.2 SSRIs

2

80

Std. Mean Difference (IV, Random, 95% CI)

0.29 [‐0.16, 0.73]

4.3 5‐HT2 antagonists

2

41

Std. Mean Difference (IV, Random, 95% CI)

0.02 [‐0.59, 0.64]

5 Response to antidepressive treatment Show forest plot

10

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.5

Comparison 1 Antidepressants versus placebo: all studies, Outcome 5 Response to antidepressive treatment.

Comparison 1 Antidepressants versus placebo: all studies, Outcome 5 Response to antidepressive treatment.

5.1 All studies

10

805

Risk Ratio (M‐H, Random, 95% CI)

1.40 [1.08, 1.82]

5.2 Tricyclic antidepressants (TCAs)

4

212

Risk Ratio (M‐H, Random, 95% CI)

1.60 [1.09, 2.34]

5.3 SSRIs

5

529

Risk Ratio (M‐H, Random, 95% CI)

1.19 [0.87, 1.63]

6 Full remission of depression Show forest plot

4

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.6

Comparison 1 Antidepressants versus placebo: all studies, Outcome 6 Full remission of depression.

Comparison 1 Antidepressants versus placebo: all studies, Outcome 6 Full remission of depression.

6.1 All studies

4

372

Risk Ratio (M‐H, Random, 95% CI)

1.19 [0.77, 1.83]

6.2 SSRIs

3

308

Risk Ratio (M‐H, Random, 95% CI)

1.00 [0.74, 1.36]

7 Consumption of alcohol: abstinent days (%) Show forest plot

9

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.7

Comparison 1 Antidepressants versus placebo: all studies, Outcome 7 Consumption of alcohol: abstinent days (%).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 7 Consumption of alcohol: abstinent days (%).

7.1 All studies

9

821

Mean Difference (IV, Random, 95% CI)

1.34 [‐1.66, 4.34]

7.2 SSRIs

7

711

Mean Difference (IV, Random, 95% CI)

‐0.47 [‐3.20, 2.26]

8 Consumption of alcohol: abstinent participants (number) Show forest plot

7

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.8

Comparison 1 Antidepressants versus placebo: all studies, Outcome 8 Consumption of alcohol: abstinent participants (number).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 8 Consumption of alcohol: abstinent participants (number).

8.1 All studies

7

424

Risk Ratio (M‐H, Random, 95% CI)

1.71 [1.22, 2.39]

8.2 SSRIs

4

250

Risk Ratio (M‐H, Random, 95% CI)

1.66 [1.02, 2.68]

8.3 5‐HT2 antagonists

2

105

Risk Ratio (M‐H, Random, 95% CI)

1.62 [0.77, 3.39]

9 Consumption of alcohol: drinking days (per week) Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

Analysis 1.9

Comparison 1 Antidepressants versus placebo: all studies, Outcome 9 Consumption of alcohol: drinking days (per week).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 9 Consumption of alcohol: drinking days (per week).

9.1 All studies

2

55

Mean Difference (IV, Fixed, 95% CI)

‐1.15 [‐2.35, 0.05]

9.2 5‐HT2 antagonists

2

55

Mean Difference (IV, Fixed, 95% CI)

‐1.15 [‐2.35, 0.05]

10 Consumption of alcohol: drinks (per drinking days) Show forest plot

7

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.10

Comparison 1 Antidepressants versus placebo: all studies, Outcome 10 Consumption of alcohol: drinks (per drinking days).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 10 Consumption of alcohol: drinks (per drinking days).

10.1 All studies

7

451

Mean Difference (IV, Random, 95% CI)

‐1.13 [‐1.79, ‐0.46]

10.2 SSRIs

3

271

Mean Difference (IV, Random, 95% CI)

‐1.42 [‐2.58, ‐0.26]

10.3 5‐HT2 antagonists

3

111

Mean Difference (IV, Random, 95% CI)

‐1.06 [‐2.00, ‐0.11]

11 Consumption of alcohol: drinks (per week) Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

Analysis 1.11

Comparison 1 Antidepressants versus placebo: all studies, Outcome 11 Consumption of alcohol: drinks (per week).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 11 Consumption of alcohol: drinks (per week).

11.1 All studies

2

55

Mean Difference (IV, Fixed, 95% CI)

‐5.06 [‐12.30, 2.18]

11.2 5‐HT2 antagonists

2

55

Mean Difference (IV, Fixed, 95% CI)

‐5.06 [‐12.30, 2.18]

12 Consumption of alcohol: heavy drinking days (per week) Show forest plot

5

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.12

Comparison 1 Antidepressants versus placebo: all studies, Outcome 12 Consumption of alcohol: heavy drinking days (per week).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 12 Consumption of alcohol: heavy drinking days (per week).

12.1 All studies

5

313

Mean Difference (IV, Random, 95% CI)

‐0.33 [‐0.85, 0.20]

12.2 SSRIs

2

189

Mean Difference (IV, Random, 95% CI)

‐0.41 [‐1.09, 0.27]

12.3 5‐HT2 antagonists

2

55

Mean Difference (IV, Random, 95% CI)

‐0.43 [‐2.09, 1.22]

13 Consumption of alcohol: heavy drinkers (number) Show forest plot

9

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.13

Comparison 1 Antidepressants versus placebo: all studies, Outcome 13 Consumption of alcohol: heavy drinkers (number).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 13 Consumption of alcohol: heavy drinkers (number).

13.1 All studies

7

459

Risk Ratio (M‐H, Random, 95% CI)

0.78 [0.57, 1.07]

13.2 SSRIs

6

431

Risk Ratio (M‐H, Random, 95% CI)

0.87 [0.69, 1.11]

13.3 5‐HT2 antagonists

2

99

Risk Ratio (M‐H, Random, 95% CI)

1.78 [0.68, 4.67]

14 Consumption of alcohol: time to first relapse (days) Show forest plot

6

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.14

Comparison 1 Antidepressants versus placebo: all studies, Outcome 14 Consumption of alcohol: time to first relapse (days).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 14 Consumption of alcohol: time to first relapse (days).

14.1 All studies

6

348

Mean Difference (IV, Random, 95% CI)

2.54 [‐8.79, 13.87]

14.2 SSRIs

6

348

Mean Difference (IV, Random, 95% CI)

2.54 [‐8.79, 13.87]

15 Liver enzyme levels: γ‐glutamyltransferase (U/L) Show forest plot

2

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.15

Comparison 1 Antidepressants versus placebo: all studies, Outcome 15 Liver enzyme levels: γ‐glutamyltransferase (U/L).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 15 Liver enzyme levels: γ‐glutamyltransferase (U/L).

15.1 All studies

2

56

Mean Difference (IV, Random, 95% CI)

‐8.39 [‐26.47, 9.68]

16 Depression and alcohol: global response Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.16

Comparison 1 Antidepressants versus placebo: all studies, Outcome 16 Depression and alcohol: global response.

Comparison 1 Antidepressants versus placebo: all studies, Outcome 16 Depression and alcohol: global response.

16.1 All studies

3

152

Risk Ratio (M‐H, Random, 95% CI)

2.37 [1.34, 4.19]

16.2 TCAs

2

92

Risk Ratio (M‐H, Random, 95% CI)

2.09 [1.09, 4.02]

17 Acceptability: dropouts Show forest plot

17

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.17

Comparison 1 Antidepressants versus placebo: all studies, Outcome 17 Acceptability: dropouts.

Comparison 1 Antidepressants versus placebo: all studies, Outcome 17 Acceptability: dropouts.

17.1 All studies

17

1159

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.79, 1.22]

17.2 TCAs

4

216

Risk Ratio (M‐H, Random, 95% CI)

1.21 [0.48, 3.06]

17.3 SSRIs

8

759

Risk Ratio (M‐H, Random, 95% CI)

1.04 [0.79, 1.36]

17.4 5‐HT2 antagonists

4

154

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.38, 1.64]

18 Tolerability of treatment: adverse events Show forest plot

15

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.18

Comparison 1 Antidepressants versus placebo: all studies, Outcome 18 Tolerability of treatment: adverse events.

Comparison 1 Antidepressants versus placebo: all studies, Outcome 18 Tolerability of treatment: adverse events.

18.1 Withdrawal for medical reasons: all studies

10

947

Risk Ratio (M‐H, Random, 95% CI)

1.15 [0.65, 2.04]

18.2 Withdrawal for medical reasons: SSRIs

7

786

Risk Ratio (M‐H, Random, 95% CI)

1.10 [0.52, 2.32]

18.3 Withdrawal for medical reasons: TCAs

2

97

Risk Ratio (M‐H, Random, 95% CI)

0.91 [0.10, 8.41]

18.4 Total adverse events: all studies

5

644

Risk Ratio (M‐H, Random, 95% CI)

1.18 [0.97, 1.44]

18.5 Total adverse events: TCAs

2

115

Risk Ratio (M‐H, Random, 95% CI)

1.66 [1.13, 2.42]

18.6 Total adverse events: SSRIs

3

529

Risk Ratio (M‐H, Random, 95% CI)

1.06 [0.92, 1.23]

18.7 Dry mouth: all studies

2

132

Risk Ratio (M‐H, Random, 95% CI)

1.91 [0.96, 3.81]

18.8 Insomnia: all studies

4

564

Risk Ratio (M‐H, Random, 95% CI)

1.69 [1.02, 2.77]

18.9 Insomnia: SSRIs

2

469

Risk Ratio (M‐H, Random, 95% CI)

1.75 [1.04, 2.96]

18.10 Headache: all studies

3

470

Risk Ratio (M‐H, Random, 95% CI)

1.21 [0.89, 1.64]

18.11 Headache: SSRIs

2

414

Risk Ratio (M‐H, Random, 95% CI)

1.18 [0.87, 1.61]

18.12 Dizziness: all studies

2

139

Risk Ratio (M‐H, Random, 95% CI)

1.68 [0.42, 6.73]

18.13 Diarrhoea: all studies

2

139

Risk Ratio (M‐H, Random, 95% CI)

1.95 [0.37, 10.22]

18.14 Nausea: all studies

3

277

Risk Ratio (M‐H, Random, 95% CI)

1.46 [0.66, 3.23]

18.15 Nausea: SSRIs

2

221

Risk Ratio (M‐H, Random, 95% CI)

1.44 [0.62, 3.35]

18.16 Constipation: all studies

2

387

Risk Ratio (M‐H, Random, 95% CI)

1.70 [0.19, 15.64]

18.17 Total serious adverse events: all studies

7

774

Risk Ratio (M‐H, Random, 95% CI)

1.22 [0.80, 1.86]

18.18 Total serious adverse events: SSRIs

5

721

Risk Ratio (M‐H, Random, 95% CI)

1.22 [0.80, 1.86]

18.19 Worsening of clinical condition because of relapse: all studies

2

413

Risk Ratio (M‐H, Random, 95% CI)

2.81 [0.73, 10.87]

18.20 Worsening of clinical condition because of relapse: SSRIs

2

413

Risk Ratio (M‐H, Random, 95% CI)

2.81 [0.73, 10.87]

18.21 Depression: all studies

2

413

Risk Ratio (M‐H, Random, 95% CI)

2.31 [0.30, 17.69]

18.22 Depression: SSRIs

2

413

Risk Ratio (M‐H, Random, 95% CI)

2.31 [0.30, 17.69]

19 Suicide attempts Show forest plot

4

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.19

Comparison 1 Antidepressants versus placebo: all studies, Outcome 19 Suicide attempts.

Comparison 1 Antidepressants versus placebo: all studies, Outcome 19 Suicide attempts.

19.1 All studies

4

602

Risk Ratio (M‐H, Random, 95% CI)

1.33 [0.23, 7.61]

19.2 SSRIs

4

602

Risk Ratio (M‐H, Random, 95% CI)

1.33 [0.23, 7.61]

20 Secondary outcomes: craving Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

Analysis 1.20

Comparison 1 Antidepressants versus placebo: all studies, Outcome 20 Secondary outcomes: craving.

Comparison 1 Antidepressants versus placebo: all studies, Outcome 20 Secondary outcomes: craving.

20.1 All studies

2

29

Mean Difference (IV, Fixed, 95% CI)

1.00 [‐3.27, 5.27]

21 Secondary outcomes: severity of dependence Show forest plot

2

Std. Mean Difference (IV, Fixed, 95% CI)

Subtotals only

Analysis 1.21

Comparison 1 Antidepressants versus placebo: all studies, Outcome 21 Secondary outcomes: severity of dependence.

Comparison 1 Antidepressants versus placebo: all studies, Outcome 21 Secondary outcomes: severity of dependence.

21.1 All studies

2

168

Std. Mean Difference (IV, Fixed, 95% CI)

‐0.14 [‐0.44, 0.17]

22 Secondary outcomes: severity of anxiety Show forest plot

3

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

Analysis 1.22

Comparison 1 Antidepressants versus placebo: all studies, Outcome 22 Secondary outcomes: severity of anxiety.

Comparison 1 Antidepressants versus placebo: all studies, Outcome 22 Secondary outcomes: severity of anxiety.

22.1 All studies

3

97

Mean Difference (IV, Fixed, 95% CI)

‐6.31 [‐10.33, ‐2.28]

Open in table viewer
Comparison 2. Antidepressants versus psychotherapy

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Depression severity: final score Show forest plot

2

60

Mean Difference (IV, Random, 95% CI)

‐2.61 [‐6.92, 1.70]

Analysis 2.1

Comparison 2 Antidepressants versus psychotherapy, Outcome 1 Depression severity: final score.

Comparison 2 Antidepressants versus psychotherapy, Outcome 1 Depression severity: final score.

2 Global assessment: final score Show forest plot

2

60

Mean Difference (IV, Random, 95% CI)

5.92 [1.30, 10.54]

Analysis 2.2

Comparison 2 Antidepressants versus psychotherapy, Outcome 2 Global assessment: final score.

Comparison 2 Antidepressants versus psychotherapy, Outcome 2 Global assessment: final score.

3 Acceptability: dropouts Show forest plot

2

68

Risk Ratio (M‐H, Random, 95% CI)

1.43 [0.31, 6.54]

Analysis 2.3

Comparison 2 Antidepressants versus psychotherapy, Outcome 3 Acceptability: dropouts.

Comparison 2 Antidepressants versus psychotherapy, Outcome 3 Acceptability: dropouts.

Study flow diagram.
Figuras y tablas -
Figure 1

Study flow diagram.

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figuras y tablas -
Figure 2

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.
Figuras y tablas -
Figure 3

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Funnel plot of comparison: 1 Antidepressants versus placebo: all studies, outcome: 1.1 Depression severity: final score (interviewer‐rated scales).
Figuras y tablas -
Figure 4

Funnel plot of comparison: 1 Antidepressants versus placebo: all studies, outcome: 1.1 Depression severity: final score (interviewer‐rated scales).

Funnel plot of comparison: 1 Antidepressants versus placebo: all studies, outcome: 1.5 Response to antidepressive treatment.
Figuras y tablas -
Figure 5

Funnel plot of comparison: 1 Antidepressants versus placebo: all studies, outcome: 1.5 Response to antidepressive treatment.

Funnel plot of comparison: 1 Antidepressants versus placebo: all studies, outcome: 1.17 Acceptability: dropouts.
Figuras y tablas -
Figure 6

Funnel plot of comparison: 1 Antidepressants versus placebo: all studies, outcome: 1.17 Acceptability: dropouts.

Comparison 1 Antidepressants versus placebo: all studies, Outcome 1 Depression severity: final score (interviewer‐rated scales).
Figuras y tablas -
Analysis 1.1

Comparison 1 Antidepressants versus placebo: all studies, Outcome 1 Depression severity: final score (interviewer‐rated scales).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 2 Depression severity: final score (self‐administered scales).
Figuras y tablas -
Analysis 1.2

Comparison 1 Antidepressants versus placebo: all studies, Outcome 2 Depression severity: final score (self‐administered scales).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 3 Depression severity: difference between basal and final score (interviewer‐rated scales).
Figuras y tablas -
Analysis 1.3

Comparison 1 Antidepressants versus placebo: all studies, Outcome 3 Depression severity: difference between basal and final score (interviewer‐rated scales).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 4 Depression severity: difference between basal and final score (self‐administered scales).
Figuras y tablas -
Analysis 1.4

Comparison 1 Antidepressants versus placebo: all studies, Outcome 4 Depression severity: difference between basal and final score (self‐administered scales).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 5 Response to antidepressive treatment.
Figuras y tablas -
Analysis 1.5

Comparison 1 Antidepressants versus placebo: all studies, Outcome 5 Response to antidepressive treatment.

Comparison 1 Antidepressants versus placebo: all studies, Outcome 6 Full remission of depression.
Figuras y tablas -
Analysis 1.6

Comparison 1 Antidepressants versus placebo: all studies, Outcome 6 Full remission of depression.

Comparison 1 Antidepressants versus placebo: all studies, Outcome 7 Consumption of alcohol: abstinent days (%).
Figuras y tablas -
Analysis 1.7

Comparison 1 Antidepressants versus placebo: all studies, Outcome 7 Consumption of alcohol: abstinent days (%).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 8 Consumption of alcohol: abstinent participants (number).
Figuras y tablas -
Analysis 1.8

Comparison 1 Antidepressants versus placebo: all studies, Outcome 8 Consumption of alcohol: abstinent participants (number).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 9 Consumption of alcohol: drinking days (per week).
Figuras y tablas -
Analysis 1.9

Comparison 1 Antidepressants versus placebo: all studies, Outcome 9 Consumption of alcohol: drinking days (per week).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 10 Consumption of alcohol: drinks (per drinking days).
Figuras y tablas -
Analysis 1.10

Comparison 1 Antidepressants versus placebo: all studies, Outcome 10 Consumption of alcohol: drinks (per drinking days).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 11 Consumption of alcohol: drinks (per week).
Figuras y tablas -
Analysis 1.11

Comparison 1 Antidepressants versus placebo: all studies, Outcome 11 Consumption of alcohol: drinks (per week).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 12 Consumption of alcohol: heavy drinking days (per week).
Figuras y tablas -
Analysis 1.12

Comparison 1 Antidepressants versus placebo: all studies, Outcome 12 Consumption of alcohol: heavy drinking days (per week).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 13 Consumption of alcohol: heavy drinkers (number).
Figuras y tablas -
Analysis 1.13

Comparison 1 Antidepressants versus placebo: all studies, Outcome 13 Consumption of alcohol: heavy drinkers (number).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 14 Consumption of alcohol: time to first relapse (days).
Figuras y tablas -
Analysis 1.14

Comparison 1 Antidepressants versus placebo: all studies, Outcome 14 Consumption of alcohol: time to first relapse (days).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 15 Liver enzyme levels: γ‐glutamyltransferase (U/L).
Figuras y tablas -
Analysis 1.15

Comparison 1 Antidepressants versus placebo: all studies, Outcome 15 Liver enzyme levels: γ‐glutamyltransferase (U/L).

Comparison 1 Antidepressants versus placebo: all studies, Outcome 16 Depression and alcohol: global response.
Figuras y tablas -
Analysis 1.16

Comparison 1 Antidepressants versus placebo: all studies, Outcome 16 Depression and alcohol: global response.

Comparison 1 Antidepressants versus placebo: all studies, Outcome 17 Acceptability: dropouts.
Figuras y tablas -
Analysis 1.17

Comparison 1 Antidepressants versus placebo: all studies, Outcome 17 Acceptability: dropouts.

Comparison 1 Antidepressants versus placebo: all studies, Outcome 18 Tolerability of treatment: adverse events.
Figuras y tablas -
Analysis 1.18

Comparison 1 Antidepressants versus placebo: all studies, Outcome 18 Tolerability of treatment: adverse events.

Comparison 1 Antidepressants versus placebo: all studies, Outcome 19 Suicide attempts.
Figuras y tablas -
Analysis 1.19

Comparison 1 Antidepressants versus placebo: all studies, Outcome 19 Suicide attempts.

Comparison 1 Antidepressants versus placebo: all studies, Outcome 20 Secondary outcomes: craving.
Figuras y tablas -
Analysis 1.20

Comparison 1 Antidepressants versus placebo: all studies, Outcome 20 Secondary outcomes: craving.

Comparison 1 Antidepressants versus placebo: all studies, Outcome 21 Secondary outcomes: severity of dependence.
Figuras y tablas -
Analysis 1.21

Comparison 1 Antidepressants versus placebo: all studies, Outcome 21 Secondary outcomes: severity of dependence.

Comparison 1 Antidepressants versus placebo: all studies, Outcome 22 Secondary outcomes: severity of anxiety.
Figuras y tablas -
Analysis 1.22

Comparison 1 Antidepressants versus placebo: all studies, Outcome 22 Secondary outcomes: severity of anxiety.

Comparison 2 Antidepressants versus psychotherapy, Outcome 1 Depression severity: final score.
Figuras y tablas -
Analysis 2.1

Comparison 2 Antidepressants versus psychotherapy, Outcome 1 Depression severity: final score.

Comparison 2 Antidepressants versus psychotherapy, Outcome 2 Global assessment: final score.
Figuras y tablas -
Analysis 2.2

Comparison 2 Antidepressants versus psychotherapy, Outcome 2 Global assessment: final score.

Comparison 2 Antidepressants versus psychotherapy, Outcome 3 Acceptability: dropouts.
Figuras y tablas -
Analysis 2.3

Comparison 2 Antidepressants versus psychotherapy, Outcome 3 Acceptability: dropouts.

Summary of findings for the main comparison. Antidepressants compared to placebo: all studies for the treatment of people with co‐occurring depression and alcohol consumption

Antidepressants compared to placebo: all studies

Patient or population: people with co‐occurring depression and alcohol dependence

Settings: unknown
Intervention: antidepressants
Comparison: 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

Placebo

Antidepressants

Depression severity: final score (interviewer‐rated scales)

The mean depression: final score (interviewer‐rated scales) ‐ all studies in the intervention groups was

0.27 standard deviations lower (0.49 lower to 0.04 lower)

1074
(14 studies)

⊕⊕⊝⊝
Low1,2

Response to antidepressive treatment

Study population

RR 1.40
(1.08 to 1.82)

805
(10 studies)

⊕⊝⊝⊝
Very low3,4,5

481 per 1000

674 per 1000
(520 to 876)

392 per 1000

521 per 1000
(416 to 659)

Consumption of alcohol: abstinent days (%)

The mean alcohol: abstinent days (%) ‐ all studies in the intervention groups was

1.34 higher (1.66 lower to 4.34 higher)

821
(9 studies)

⊕⊕⊝⊝
Low6,7

Consumption of alcohol: abstinent participants (number)

Study population

RR 1.71
(1.22 to 2.39)

424
(7 studies)

⊕⊕⊕⊝
Moderate8,9

199 per 1000

340 per 1000
(243 to 476)

188 per 1000

321 per 1000
(229 to 449)

Consumption of alcohol: drinks (per drinking days)

The mean alcohol: drinks (per drinking days) ‐ all studies in the intervention groups was
1.13 lower (1.79 lower to 0.46 lower)

451
(7 studies)

⊕⊕⊕⊝
Moderate10

Acceptability: dropouts

Study population

RR 0.98
(0.79 to 1.22)

1159
(17 studies)

⊕⊕⊝⊝
Low11,12

334 per 1000

328 per 1000
(264 to 408)

307 per 1000

301 per 1000
(243 to 375)

Tolerability of treatment: withdrawal for medical reasons

Study population

RR 1.15
(0.65 to 2.04)

947
(10 studies)

⊕⊕⊝⊝
Low13,14

69 per 1000

80 per 1000
(45 to 141)

32 per 1000

37 per 1000
(21 to 65)

*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: We are very confident that the true effect lies close to that of the estimate of the effect.
Moderate: We are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low: Our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect
Very low: We have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1Nine studies at unclear risk for selection bias; two studies at high risk and six at unclear risk for performance bias; 13 studies at unclear risk for detection bias (subjective); one study at high risk and two at unclear risk for attrition bias; three studies at high risk and one at unclear risk for reporting bias.
2Significant heterogeneity: Tau² = 0.11; Chi² = 39.01, df = 13 (P = 0.0002); I² = 67%.
3Ten studies at unclear risk for selection bias; one study at high risk and five at unclear risk for performance bias; nine studies at unclear risk for detection bias (subjective); two studies at high risk and two at unclear risk for attrition bias; two studies at high risk and two at unclear risk for reporting bias.
4Significant heterogeneity: Tau² = 0.10; Chi² = 31.63, df = 9 (P = 0.0002); I² = 72%.
5Funnel plot showed asymmetry in favour of 'positive' trials.

6Seven studies with unclear risk of selection bias; one study at high risk and five studies at unclear risk for performance bias; all studies at unclear risk for detection bias (subjective); one study at high risk and two at unclear risk for reporting bias.
7Significant heterogeneity: Tau² = 9.16; Chi² = 39.42, df = 8 (P < 0.00001); I² = 80%.
8Four studies with unclear risk of selection bias; one study at high risk and two studies at unclear risk for performance bias; six studies at unclear risk for detection bias (subjective); one study at high risk and one at unclear risk for reporting bias.
9Total number of events was fewer than 300.
10Five studies with unclear risk of selection bias; one study at high risk and two studies at unclear risk for performance bias; six studies at unclear risk for detection bias (subjective); one study at high risk and two at unclear risk for reporting bias.
11Twelve studies with unclear risk of selection bias; one study at high risk and seven studies at unclear risk for performance bias; 16 studies at unclear risk for detection bias (subjective); four studies at high risk and three studies at unclear risk for attrition bias; five studies at high risk and two at unclear risk for reporting bias.
12Significant heterogeneity: Chi² = 23.80, df = 14 (P = 0.05); I² = 41%.
13Six studies at unclear risk for selection bias; one study at high risk and three at unclear risk for performance bias; nine studies at unclear risk for detection bias (subjective); two studies at high risk and one at unclear risk for attrition bias; three studies at high risk and two at unclear risk for reporting bias.
14Optimal information size not met.

Figuras y tablas -
Summary of findings for the main comparison. Antidepressants compared to placebo: all studies for the treatment of people with co‐occurring depression and alcohol consumption
Comparison 1. Antidepressants versus placebo: all studies

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Depression severity: final score (interviewer‐rated scales) Show forest plot

14

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

1.1 All studies

14

1074

Std. Mean Difference (IV, Random, 95% CI)

‐0.27 [‐0.49, ‐0.04]

1.2 Selective serotonin reuptake inhibitors (SSRIs)

10

881

Std. Mean Difference (IV, Random, 95% CI)

‐0.17 [‐0.40, 0.07]

1.3 5‐HT2 antagonists

2

97

Std. Mean Difference (IV, Random, 95% CI)

‐0.29 [‐0.69, 0.11]

2 Depression severity: final score (self‐administered scales) Show forest plot

8

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

2.1 All studies

8

373

Std. Mean Difference (IV, Random, 95% CI)

‐0.29 [‐0.64, 0.07]

2.2 SSRIs

5

300

Std. Mean Difference (IV, Random, 95% CI)

‐0.25 [‐0.69, 0.18]

2.3 5‐HT2 antagonists

2

41

Std. Mean Difference (IV, Random, 95% CI)

0.02 [‐0.59, 0.64]

3 Depression severity: difference between basal and final score (interviewer‐rated scales) Show forest plot

5

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

3.1 All studies

5

447

Std. Mean Difference (IV, Random, 95% CI)

0.15 [‐0.12, 0.42]

3.2 SSRIs

4

408

Std. Mean Difference (IV, Random, 95% CI)

0.07 [‐0.18, 0.32]

4 Depression severity: difference between basal and final score (self‐administered scales) Show forest plot

4

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

4.1 All studies

4

121

Std. Mean Difference (IV, Random, 95% CI)

0.20 [‐0.16, 0.56]

4.2 SSRIs

2

80

Std. Mean Difference (IV, Random, 95% CI)

0.29 [‐0.16, 0.73]

4.3 5‐HT2 antagonists

2

41

Std. Mean Difference (IV, Random, 95% CI)

0.02 [‐0.59, 0.64]

5 Response to antidepressive treatment Show forest plot

10

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

5.1 All studies

10

805

Risk Ratio (M‐H, Random, 95% CI)

1.40 [1.08, 1.82]

5.2 Tricyclic antidepressants (TCAs)

4

212

Risk Ratio (M‐H, Random, 95% CI)

1.60 [1.09, 2.34]

5.3 SSRIs

5

529

Risk Ratio (M‐H, Random, 95% CI)

1.19 [0.87, 1.63]

6 Full remission of depression Show forest plot

4

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

6.1 All studies

4

372

Risk Ratio (M‐H, Random, 95% CI)

1.19 [0.77, 1.83]

6.2 SSRIs

3

308

Risk Ratio (M‐H, Random, 95% CI)

1.00 [0.74, 1.36]

7 Consumption of alcohol: abstinent days (%) Show forest plot

9

Mean Difference (IV, Random, 95% CI)

Subtotals only

7.1 All studies

9

821

Mean Difference (IV, Random, 95% CI)

1.34 [‐1.66, 4.34]

7.2 SSRIs

7

711

Mean Difference (IV, Random, 95% CI)

‐0.47 [‐3.20, 2.26]

8 Consumption of alcohol: abstinent participants (number) Show forest plot

7

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

8.1 All studies

7

424

Risk Ratio (M‐H, Random, 95% CI)

1.71 [1.22, 2.39]

8.2 SSRIs

4

250

Risk Ratio (M‐H, Random, 95% CI)

1.66 [1.02, 2.68]

8.3 5‐HT2 antagonists

2

105

Risk Ratio (M‐H, Random, 95% CI)

1.62 [0.77, 3.39]

9 Consumption of alcohol: drinking days (per week) Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

9.1 All studies

2

55

Mean Difference (IV, Fixed, 95% CI)

‐1.15 [‐2.35, 0.05]

9.2 5‐HT2 antagonists

2

55

Mean Difference (IV, Fixed, 95% CI)

‐1.15 [‐2.35, 0.05]

10 Consumption of alcohol: drinks (per drinking days) Show forest plot

7

Mean Difference (IV, Random, 95% CI)

Subtotals only

10.1 All studies

7

451

Mean Difference (IV, Random, 95% CI)

‐1.13 [‐1.79, ‐0.46]

10.2 SSRIs

3

271

Mean Difference (IV, Random, 95% CI)

‐1.42 [‐2.58, ‐0.26]

10.3 5‐HT2 antagonists

3

111

Mean Difference (IV, Random, 95% CI)

‐1.06 [‐2.00, ‐0.11]

11 Consumption of alcohol: drinks (per week) Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

11.1 All studies

2

55

Mean Difference (IV, Fixed, 95% CI)

‐5.06 [‐12.30, 2.18]

11.2 5‐HT2 antagonists

2

55

Mean Difference (IV, Fixed, 95% CI)

‐5.06 [‐12.30, 2.18]

12 Consumption of alcohol: heavy drinking days (per week) Show forest plot

5

Mean Difference (IV, Random, 95% CI)

Subtotals only

12.1 All studies

5

313

Mean Difference (IV, Random, 95% CI)

‐0.33 [‐0.85, 0.20]

12.2 SSRIs

2

189

Mean Difference (IV, Random, 95% CI)

‐0.41 [‐1.09, 0.27]

12.3 5‐HT2 antagonists

2

55

Mean Difference (IV, Random, 95% CI)

‐0.43 [‐2.09, 1.22]

13 Consumption of alcohol: heavy drinkers (number) Show forest plot

9

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

13.1 All studies

7

459

Risk Ratio (M‐H, Random, 95% CI)

0.78 [0.57, 1.07]

13.2 SSRIs

6

431

Risk Ratio (M‐H, Random, 95% CI)

0.87 [0.69, 1.11]

13.3 5‐HT2 antagonists

2

99

Risk Ratio (M‐H, Random, 95% CI)

1.78 [0.68, 4.67]

14 Consumption of alcohol: time to first relapse (days) Show forest plot

6

Mean Difference (IV, Random, 95% CI)

Subtotals only

14.1 All studies

6

348

Mean Difference (IV, Random, 95% CI)

2.54 [‐8.79, 13.87]

14.2 SSRIs

6

348

Mean Difference (IV, Random, 95% CI)

2.54 [‐8.79, 13.87]

15 Liver enzyme levels: γ‐glutamyltransferase (U/L) Show forest plot

2

Mean Difference (IV, Random, 95% CI)

Subtotals only

15.1 All studies

2

56

Mean Difference (IV, Random, 95% CI)

‐8.39 [‐26.47, 9.68]

16 Depression and alcohol: global response Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

16.1 All studies

3

152

Risk Ratio (M‐H, Random, 95% CI)

2.37 [1.34, 4.19]

16.2 TCAs

2

92

Risk Ratio (M‐H, Random, 95% CI)

2.09 [1.09, 4.02]

17 Acceptability: dropouts Show forest plot

17

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

17.1 All studies

17

1159

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.79, 1.22]

17.2 TCAs

4

216

Risk Ratio (M‐H, Random, 95% CI)

1.21 [0.48, 3.06]

17.3 SSRIs

8

759

Risk Ratio (M‐H, Random, 95% CI)

1.04 [0.79, 1.36]

17.4 5‐HT2 antagonists

4

154

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.38, 1.64]

18 Tolerability of treatment: adverse events Show forest plot

15

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

18.1 Withdrawal for medical reasons: all studies

10

947

Risk Ratio (M‐H, Random, 95% CI)

1.15 [0.65, 2.04]

18.2 Withdrawal for medical reasons: SSRIs

7

786

Risk Ratio (M‐H, Random, 95% CI)

1.10 [0.52, 2.32]

18.3 Withdrawal for medical reasons: TCAs

2

97

Risk Ratio (M‐H, Random, 95% CI)

0.91 [0.10, 8.41]

18.4 Total adverse events: all studies

5

644

Risk Ratio (M‐H, Random, 95% CI)

1.18 [0.97, 1.44]

18.5 Total adverse events: TCAs

2

115

Risk Ratio (M‐H, Random, 95% CI)

1.66 [1.13, 2.42]

18.6 Total adverse events: SSRIs

3

529

Risk Ratio (M‐H, Random, 95% CI)

1.06 [0.92, 1.23]

18.7 Dry mouth: all studies

2

132

Risk Ratio (M‐H, Random, 95% CI)

1.91 [0.96, 3.81]

18.8 Insomnia: all studies

4

564

Risk Ratio (M‐H, Random, 95% CI)

1.69 [1.02, 2.77]

18.9 Insomnia: SSRIs

2

469

Risk Ratio (M‐H, Random, 95% CI)

1.75 [1.04, 2.96]

18.10 Headache: all studies

3

470

Risk Ratio (M‐H, Random, 95% CI)

1.21 [0.89, 1.64]

18.11 Headache: SSRIs

2

414

Risk Ratio (M‐H, Random, 95% CI)

1.18 [0.87, 1.61]

18.12 Dizziness: all studies

2

139

Risk Ratio (M‐H, Random, 95% CI)

1.68 [0.42, 6.73]

18.13 Diarrhoea: all studies

2

139

Risk Ratio (M‐H, Random, 95% CI)

1.95 [0.37, 10.22]

18.14 Nausea: all studies

3

277

Risk Ratio (M‐H, Random, 95% CI)

1.46 [0.66, 3.23]

18.15 Nausea: SSRIs

2

221

Risk Ratio (M‐H, Random, 95% CI)

1.44 [0.62, 3.35]

18.16 Constipation: all studies

2

387

Risk Ratio (M‐H, Random, 95% CI)

1.70 [0.19, 15.64]

18.17 Total serious adverse events: all studies

7

774

Risk Ratio (M‐H, Random, 95% CI)

1.22 [0.80, 1.86]

18.18 Total serious adverse events: SSRIs

5

721

Risk Ratio (M‐H, Random, 95% CI)

1.22 [0.80, 1.86]

18.19 Worsening of clinical condition because of relapse: all studies

2

413

Risk Ratio (M‐H, Random, 95% CI)

2.81 [0.73, 10.87]

18.20 Worsening of clinical condition because of relapse: SSRIs

2

413

Risk Ratio (M‐H, Random, 95% CI)

2.81 [0.73, 10.87]

18.21 Depression: all studies

2

413

Risk Ratio (M‐H, Random, 95% CI)

2.31 [0.30, 17.69]

18.22 Depression: SSRIs

2

413

Risk Ratio (M‐H, Random, 95% CI)

2.31 [0.30, 17.69]

19 Suicide attempts Show forest plot

4

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

19.1 All studies

4

602

Risk Ratio (M‐H, Random, 95% CI)

1.33 [0.23, 7.61]

19.2 SSRIs

4

602

Risk Ratio (M‐H, Random, 95% CI)

1.33 [0.23, 7.61]

20 Secondary outcomes: craving Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

20.1 All studies

2

29

Mean Difference (IV, Fixed, 95% CI)

1.00 [‐3.27, 5.27]

21 Secondary outcomes: severity of dependence Show forest plot

2

Std. Mean Difference (IV, Fixed, 95% CI)

Subtotals only

21.1 All studies

2

168

Std. Mean Difference (IV, Fixed, 95% CI)

‐0.14 [‐0.44, 0.17]

22 Secondary outcomes: severity of anxiety Show forest plot

3

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

22.1 All studies

3

97

Mean Difference (IV, Fixed, 95% CI)

‐6.31 [‐10.33, ‐2.28]

Figuras y tablas -
Comparison 1. Antidepressants versus placebo: all studies
Comparison 2. Antidepressants versus psychotherapy

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Depression severity: final score Show forest plot

2

60

Mean Difference (IV, Random, 95% CI)

‐2.61 [‐6.92, 1.70]

2 Global assessment: final score Show forest plot

2

60

Mean Difference (IV, Random, 95% CI)

5.92 [1.30, 10.54]

3 Acceptability: dropouts Show forest plot

2

68

Risk Ratio (M‐H, Random, 95% CI)

1.43 [0.31, 6.54]

Figuras y tablas -
Comparison 2. Antidepressants versus psychotherapy