Scolaris Content Display Scolaris Content Display

长期苯二氮卓类药物使用者停用苯二氮卓的药物干预

Contraer todo Desplegar todo

Referencias

References to studies included in this review

Ashton 1990 {published data only}

Ashton CH, Rawlins MD, Tyrer SP. A double‐blind placebo‐controlled study of buspirone in diazepam withdrawal in chronic benzodiazepine users. British Journal of Psychiatry 1990;157:232‐8. CENTRAL

Baandrup 2016 {published data only}

Baandrup L, Fagerlund B, Glenthoj B. Neurocognitive performance, subjective well‐being, and psychosocial functioning after benzodiazepine withdrawal in patients with schizophrenia or bipolar disorder: a randomized clinical trial of add‐on melatonin versus placebo. European Archives of Psychiatry and Clinical Neuroscience 2017;267(2):163‐71. [PUBMED: 27400927]CENTRAL
Baandrup L, Glenthoj BY, Jennum PJ. Objective and subjective sleep quality: melatonin versus placebo add‐on treatment in patients with schizophrenia or bipolar disorder withdrawing from long‐term benzodiazepine use. Psychiatry Research 2016;240:163‐9. [PUBMED: 27107670]CENTRAL
Baandrup L, Lindschou J, Winkel P, Gluud C, Glenthoj BY. Prolonged‐release melatonin versus placebo for benzodiazepine discontinuation in patients with schizophrenia or bipolar disorder: a randomised, placebo‐controlled, blinded trial. World Journal of Biological Psychiatry 2016;17(7):514‐24. [PUBMED: 26086792]CENTRAL

Cassano 1996 {published data only}

Cassano GB, Petracca A, Borghi C, Chiroli S, Didoni G, Garreau M. A randomized, double‐blind study of alpidem vs placebo in the prevention and treatment of benzodiazepine withdrawal syndrome. European Psychiatry 1996;11(2):93‐9. CENTRAL

Cialdella 2001 {published data only}

Cialdella P, Boissel JP, Belon P, the ASTRHO group. Homeopathic specialties as a substitute for benzodiazepines: a double‐blind vs. placebo study. Thérapie 2001;56:397‐402. CENTRAL

Di Costanzo 1992 {published data only}

Di Costanzo E, Rovea A. The prophylaxis of benzodiazepine withdrawal syndrome in the elderly: the effectiveness of carbamazepine. A double‐blind study vs. placebo. Minerva Psichiatrica 1992;33:301‐4. CENTRAL

Garfinkel 1999 {published data only}

Garfinkel D, Zisapel N, Wainstein J, Laudon M. Facilitation of benzodiazepine discontinuation by melatonin: a new clinical approach. Archives of Internal Medicine 1999;159(20):2456‐60. CENTRAL

Gerra 1993 {published data only}

Gerra G, Marcato A, Caccavari R, Fertonani‐Affini G, Fontanesi B, Zaimovic A, et al. Effectiveness of flumazenil (Ro 15‐1788) in the treatment of benzodiazepine withdrawal. Current Therapeutic Research ‐ Clinical and Experimental 1993;54(5):580‐7. CENTRAL

Gerra 2002 {published data only}

Gerra G, Zaimovic A, Giusti F, Moi G, Brewer C. Intravenous flumazenil versus oxazepam tapering in the treatment of benzodiazepine withdrawal: a randomized, placebo‐controlled study. Addiction Biology 2002;7(4):385‐95. CENTRAL

GlaxoSmithKline 2002 {unpublished data only}

GlaxoSmithKline. Clinical comparison of paroxetine and placebo on the symptoms emerging during the taper phase of a chronic benzodiazepine treatment, in patients suffering from a variety of anxiety disorders. GSK ‐ Clinical Study Register (www.gsk‐clinicalstudyregister.com)2002. CENTRAL

Hadley 2012 {published data only}

Hadley SJ, Mandel FS, Schweizer E. Switching from long‐term benzodiazepine therapy to pregabalin in patients with generalized anxiety disorder: a double‐blind, placebo‐controlled trial. Journal of Psychopharmacology 2012;26(4):461‐70. [DOI: 10.1177/0269881111405360]CENTRAL
Szczypa P, Hadley SJ, Donevan S, Mandel FS, Leon T. P.4.a.016 Switching from long‐term benzodiazepine therapy to pregabalin in patients with generalised anxiety disorder (GAD). European Neuropsychopharmacology 2009;19:S594‐5. [DOI: 10.1016/S0924‐977X(09)70952‐6]CENTRAL

Hantouche 1998 {published data only}

Hantouche EG, Guelfi JD, Comet D. Discontinuation of long‐term benzodiazepine use: double‐blind controlled study of α‐β L‐aspartate magnesium versus placebo in 144 chronic users of benzodiazepines [α‐β L‐aspartate de magnésium dans l'arrêt de la consommation chronique des benzodiazépines: étude contrôlée en double aveugle versus placebo]. L'encéphale 1998;XXIV:469‐79. CENTRAL
Hantouche EG, Jacob L, Comet D, Guelfi JD. Discontinuation of long‐term benzodiazepine use: predictive model of success in a double‐blind, controlled‐study. 150th Annual Meeting of the American Psychiatric Association, 1997 May 17‐22; San Diego, (CA). 1997. CENTRAL

Harrison‐Read 1996 {published data only}

Harrison‐Read PE, Tyrer P, Lawson C, Lack S, Fernandes C, File SE. Flumazenil‐precipitated panic and dysphoria in patients dependent on benzodiazepines: a possible aid to abstinence. Journal of Psychopharmacology 1996;10(2):89‐97. CENTRAL

Klein 1994 {published data only}

Klein E, Colin V, Stolk J, Lenox RH. Alprazolam withdrawal in patients with panic disorder and generalized anxiety disorder: vulnerability and effect of carbamazepine. American Journal of Psychiatry 1994;151(12):1760‐6. CENTRAL

Kornowski 2002 {published data only}

Kornowski J. The comparison between tianeptine and carbamazepine in benzodiazepines withdrawal syndrome. Psychiatria Polska 2002;6(Suppl):311‐8. CENTRAL

Lader 1987 {published data only}

Lader M, Olajide D. A comparison of buspirone and placebo in relieving benzodiazepine withdrawal symptoms. Journal of Clinical Psychopharmacology 1987;7(1):11‐5. CENTRAL

Lader 1993 {published data only}

Lader M, Farr I, Morton S. A comparison of alpidem and placebo in relieving benzodiazepine withdrawal symptoms. International Clinical Psychopharmacology 1993;8(1):31‐6. CENTRAL

Lecrubier 2005 {published data only}

Lecrubier Y, Fessard N. Benzodiazepine discontinuation in chronic users: a double‐blind trial of lithium gluconate vs placebo [Arrêt des benzodiazépines chez des consommateurs chronique: un essai en double insu du gluconate de lithium vs placebo]. Annales Médico Phychologiques 2005;163:24‐9. CENTRAL

Lemoine 2006 {published data only}

Lemoine P, Kermadi I, Garcia‐Acosta S, Garay RP, Dib M. Double‐blind, comparative study of cyamemazine vs. bromazepam in the benzodiazepine withdrawal syndrome. Progress in Neuro‐Psychopharmacology & Biological Psychiatry 2006;30(1):131‐7. CENTRAL

Mariani 2016 {published data only}

Mariani JJ, Malcolm RJ, Mamczur AK, Choi JC, Brady R, Nunes E, et al. Pilot trial of gabapentin for the treatment of benzodiazepine abuse or dependence in methadone maintenance patients. American Journal of Drug and Alcohol Abuse 2016;42(3):333‐40. [PUBMED: 26962719]CENTRAL

Mercier‐Guyon 2004 {published data only}

Mercier‐Guyon C, Chabannes JP, Saviuc P. The role of captodiamine in the withdrawal from long‐term benzodiazepine treatment. Current Medical Research and Opinion 2004;20(9):1347‐55. [DOI: 10.1185/030079904125004457]CENTRAL

Morton 1995 {published data only}

Morton S, Lader M. Buspirone treatment as an aid to benzodiazepine withdrawal. Journal of Psychopharmacology 1995;9(4):331‐5. CENTRAL

Nakao 2006 {published data only}

Nakao M, Takeuchi T, Nomura K, Teramoto T, Yano E. Clinical application of paroxetine for chronic benzodiazepine users at an internal medicine clinic. Therapeutic Research 2006;27(5):859‐67. CENTRAL
Nakao M, Takeuchi T, Nomura K, Teramoto T, Yano E. Clinical application of paroxetine for tapering benzodiazepine use in non‐major‐depressive outpatients visiting an internal medicine clinic. Psychiatry and Clinical Neurosciences 2006;60(5):605‐10. CENTRAL

Pat‐Horenczyk 1998 {published data only}

Pat‐Horenczyk R, Hacohen D, Herer P, Lavie P. The effects of substituting zopiclone in withdrawal from chronic use of benzodiazepine hypnotics. Psychopharmacology 1998;140(4):450‐7. [DOI: 10.1007/s002130050789]CENTRAL

Peles 2007 {published data only}

Peles E, Hetzroni T, Bar‐Hamburger R, Adelson M, Schreiber S. Melatonin for perceived sleep disturbances associated with benzodiazepine withdrawal among patients in methadone maintenance treatment: a double‐blind randomized clinical trial. Addiction 2007;102(12):1947‐53. CENTRAL

Rickels 1999 {published data only}

Rickels K, Schweizer E, Garcia Espana F, Case, G, DeMartinis N, Greenblatt D. Trazodone and valproate in patients discontinuing long‐term benzodiazepine therapy: effects on withdrawal symptoms and taper outcome. Psychopharmacology 1999;141(1):1‐5. CENTRAL

Rickels 2000 {published data only}

Rickels K, DeMartinis N, Garcia‐Espana F, Greenblatt DJ, Mandos LA, Rynn M. Imipramine and buspirone in treatment of patients with generalized anxiety disorder who are discontinuing long‐term benzodiazepine therapy. American Journal of Psychiatry 2000;157(12):1973‐9. CENTRAL

Romach 1998 {published data only}

Romach MK, Kaplan HL, Busto UE, Somer G, Sellers EM. A controlled trial of ondansetron, a 5‐HT3 antagonist, in benzodiazepine discontinuation. Journal of Clinical Psychopharmacology 1998;18(2):121‐31. CENTRAL

Rynn 2003 {published data only}

Rynn M, Garcia‐Espana F, Greenblatt DJ, Mandos LA, Schweizer E, Rickels K. Imipramine and buspirone in patients with panic disorder who are discontinuing long‐term benzodiazepine therapy. Journal of Clinical Psychopharmacology 2003;23(5):505‐8. CENTRAL

Saul 1989 {published data only}

Saul PA, Korlipara K, Presley P. A randomised, multicentre, double‐blind, comparison of atenolol and placebo in the control of benzodiazepine withdrawal symptoms. Acta Therapeutica 1989;15(2):117‐23. CENTRAL

Schweizer 1991 {published data only}

Schweizer E, Rickels, K, Case WG, Greenblatt DJ. Carbamazepine treatment in patients discontinuing long‐term benzodiazepine therapy. Effects on withdrawal severity and outcome. Archives of General Psychiatry 1991;48(5):448‐52. CENTRAL

Schweizer 1995 {published data only}

Schweizer E, Case WG, Garcia‐Espana F, Greenblatt DJ, Rickels K. Progesterone co‐administration in patients discontinuing long‐term benzodiazepine therapy: effects on withdrawal severity and taper outcome. Psychopharmacology 1995;117(4):424‐9. CENTRAL

Tyrer 1981 {published data only}

Tyrer P, Rutherford D, Huggett T. Benzodiazepine withdrawal symptoms and propranolol. Lancet 1981;1(8219):520‐2. CENTRAL

Tyrer 1996 {published data only}

Tyrer P, Ferguson B, Hallstrom C, Michie M, Tyrer S, Cooper S, et al. A controlled trial of dothiepin and placebo in treating benzodiazepine withdrawal symptoms. British Journal of Psychiatry 1996;168(4):457‐61. CENTRAL

Udelman 1990 {published data only}

Udelman HD, Udelman DL. Concurrent use of buspirone in anxious patients during withdrawal from alprazolam therapy. Journal of Clinical Psychiatry 1990;51 Suppl:46‐50. CENTRAL

Vissers 2007 {published data only}

Vissers FH, Knipschild PG, Crebolder HF. Is melatonin helpful in stopping the long‐term use of hypnotics? A discontinuation trial. Pharmacy World & Science 2007;29(6):641‐6. [DOI: 10.1007/s11096‐007‐9118‐y]CENTRAL

Vorma 2011 {published data only}

Vorma H, Katila H. Effect of valproate on benzodiazepine withdrawal severity in opioid‐dependent subjects: a pilot study. Heroin Addiction and Related Clinical Problems 2011;13(1):15‐20. CENTRAL

Zhang 2013 {published data only}

Zhang H, Jiang X, Ma M, Zhang J. A control study on treatment for benzodiazepine dependence with trazodone. Chinese Journal of Contemporary Neurology and Neurosurgery 2013;13(5):411‐5. CENTRAL

Zitman 2001 {published data only}

Zitman FG, Couvee JE. Chronic benzodiazepine use in general practice patients with depression: an evaluation of controlled treatment and taper‐off: report on behalf of the Dutch Chronic Benzodiazepine Working Group. British Journal of Psychiatry 2001;178:317‐24. CENTRAL

References to studies excluded from this review

Allain 1998 {published data only}

Allain H, Le Coz F, Borderies P, Schuck S, De La Giclais B, Patat A, et al. Use of zolpidem 10 mg as a benzodiazepine substitute in 84 patients with insomnia. Human Psychopharmacology 1998;13(8):551‐9. CENTRAL

Avedisova 2007 {published data only}

Avedisova AS, Iastrebov DV. Use of anxiolytic atarax as a substitutive drug for benzodiazepine tranquilizers. Zhurnal Nevrologii i psikhiatrii imeni S.S. Korsakova 2007;107(3):37‐41. CENTRAL

Bobes 2012 {published data only}

Bobes J, Rubio G, Teran A, Cervera G, Lopez‐Gomez V, Vilardaga I, et al. Pregabalin for the discontinuation of long‐term benzodiazepines use: an assessment of its effectiveness in daily clinical practice. European Psychiatry 2012;27(4):301‐7. CENTRAL

Bourgeois 2014 {published data only}

Bourgeois J, Elseviers MM, Van Bortel L, Petrovic M, Vander Stichele RH. Feasibility of discontinuing chronic benzodiazepine use in nursing home residents: a pilot study. European Journal of Clinical Pharmacology 2014;17(10):1251‐60. CENTRAL

Cantopher 1990 {published data only}

Cantopher T, Olivieri S, Cleave N, Edwards JG. Chronic benzodiazepine dependence. A comparative study of abrupt withdrawal under propranolol cover versus gradual withdrawal. British Journal of Psychiatry 1990;156:406‐11. CENTRAL

Cohen‐Mansfield 1999 {published data only}

Cohen‐Mansfield J, Lipson S, Werner P, Billig N, Taylor L, Woosley R. Withdrawal of haloperidol, thioridazine, and lorazepam in the nursing home: a controlled, double‐blind study. Archives of Internal Medicine 1999;159(15):1733‐40. CENTRAL

Declerck 1999 {published data only}

Declerck A, Smits M. Zolpidem, a valuable alternative to benzodiazepine hypnotics for chronic insomnia?. Journal of International Medical Research 1999;27(6):253‐63. CENTRAL

Emara 2009 {published data only}

Emara A, Elgharabawy R. Evaluation of escitalopram in the treatment of benzodiazepines withdrawal syndrome. Basic and Clinical Pharmacology and Toxicology 2009;105(Suppl 1):66. CENTRAL

Garcia‐Borreguero 1992 {published data only}

Garcia‐Borreguero D, Bronisch T, Apelt S, Yassouridis J, Emrich HM. Treatment of benzodiazepine withdrawal symptoms with carbamazepine. Pharmacopsychiatry 1992;25:100. CENTRAL

Hallstrom 1988 {published data only}

Hallstrom C, Crouch G, Robson M, Shine P. The treatment of tranquilizer dependence by propranolol. Postgraduate Medical Journal 1988;64 Suppl 2:40‐4. CENTRAL

Isaka 2009 {published data only}

Isaka M. Withdrawal symptoms of benzodiazepines in panic disorder patients' pharmacotherapy. Journal of the Osaka City Medical Center 2009;58(1‐2):11‐20. CENTRAL

Lahteenmaki 2014 {published data only}

Lahteenmaki R, Puustinen J, Vahlberg T, Lyles A, Neuvonen PJ, Partinen M, et al. Melatonin for sedative withdrawal in older patients with primary insomnia: a randomized double‐blind placebo‐controlled trial. British Journal of Clinical Pharmacology 2014;77(6):975‐85. CENTRAL

Lemoine 1997 {published data only}

Lemoine P, Touchon J, Billardon M. Comparison of 6 different methods for lorazepam withdrawal. A controlled study, hydroxyzine versus placebo. Encephale 1997;23(4):290‐9. CENTRAL

Lopatko 2006 {published data only}

Lopatko O, Morefield K, Danz C, Davies J, Ali R, White JM. Reducing benzodiazepine consumption in opioid maintenance therapy patients: A controlled clinical trial. Proceedings of the 68th Annual Scientific Meeting of the College on Problems of Drug Dependence. 2006:20. CENTRAL

Nakajima 2007 {published data only}

Nakajima S, Uchida H, Suzuki T, Tomita M, Tsunoda K, Kitta M, et al. An open‐label trial of discontinuing benzodiazepines in patients with chronic schizophrenia. Journal of Clinical Psychopharmacology 2007;27(4):401‐3. CENTRAL

Petrovic 2002 {published data only}

Petrovic M, Pevernagie D, Mariman A, Van Maele G, Afschrift M. Fast withdrawal from benzodiazepines in geriatric inpatients: a randomised double‐blind, placebo‐controlled trial. European Journal of Clinical Pharmacology 2002;57:759‐64. CENTRAL

Rocco 1992 {published data only}

Rocco PL, Giavedoni A, Pacella G. Withdrawal from benzodiazepines in a hospital setting: an open trial with buspirone. Current Therapeutic Research ‐ Clinical and Experimental 1992;52(3):386‐9. CENTRAL

Rubio 2009 {published data only}

Rubio G, Bobes J, Cervera G, Teran A, Perez M, Lopez‐Gomez V, et al. Benzodiazepines use withdrawal tapering gradually with pregabalin: findings from the medical practice. European Neuropsychopharmacology 2009;19:S652. CENTRAL

Saxon 1997 {published data only}

Saxon L, Hjemdahl P, Hiltunen AJ, Borg S. Effects of flumazenil in the treatment of benzodiazepine withdrawal ‐ a double‐blind pilot study. Psychopharmacology 1997;131(2):153‐60. CENTRAL

Shapiro 1995 {published data only}

Shapiro CM, Sherman D, Peck DF. Withdrawal from benzodiazepines by initially switching to zopiclone. European Psychiatry 1995;10:S145‐51. CENTRAL

Vescovi 1987 {published data only}

Vescovi PP, Gerra G, Ippolito L. Nicotinic acid effectiveness in the treatment of benzodiazepine withdrawal. Current Therapeutic Research ‐ Clinical and Experimental 1987;41(6):1017‐21. CENTRAL

Weizman 2003 {published data only}

Weizman T, Gelkopf M, Melamed Y, Adelson M, Bleich A. Treatment of benzodiazepine dependence in methadone maintenance treatment patients: a comparison of two therapeutic modalities and the role of psychiatric comorbidity. Australian and New Zealand Journal of Psychiatry 2003;37(4):458‐63. CENTRAL

Additional references

Ashton 2005

Ashton H. The diagnosis and management of benzodiazepine dependence. Current Opinion in Psychiatry 2005;18(3):249‐55. [PUBMED: 16639148]

Baandrup 2015

Baandrup L, Ebdrup BH, Lindschou J, Gluud C, Glenthøj BY. Pharmacological interventions for benzodiazepine discontinuation in chronic benzodiazepine users. Cochrane Database of Systematic Reviews 2015, Issue 1. [DOI: 10.1002/14651858.CD011481]

Bachhuber 2016

Bachhuber MA, Hennessy S, Cunningham CO, Starrels JL. Increasing benzodiazepine prescriptions and overdose mortality in the United States, 1996‐2013. American Journal of Public Health 2016;106(4):686‐8. [PUBMED: 26890165]

Baldwin 2013

Baldwin DS, Aitchison K, Bateson A, Curran HV, Davies S, Leonard B, et al. Benzodiazepines: risks and benefits. A reconsideration. Journal of Psychopharmacology 2013;27(11):967‐71.

Balshem 2011

Balshem H, Helfand M, Schunemann HJ, Oxman AD, Kunz R, Brozek J, et al. GRADE guidelines: 3. Rating the quality of evidence. Journal of Clinical Epidemiology 2011;64(4):401‐6.

Barker 2004

Barker MJ, Greenwood KM, Jackson M, Crowe SF. Cognitive effects of long‐term benzodiazepine use: a meta‐analysis. CNS Drugs 2004;18(1):37‐48.

Billioti 2012

Billioti de Gage S, Begaud B, Bazin F, Verdoux H, Dartigues JF, Peres K, et al. Benzodiazepine use and risk of dementia: prospective population based study. BMJ 2012;345(27):e6231.

Brok 2008

Brok J, Thorlund K, Gluud C, Wetterslev J. Trial sequential analysis reveals insufficient information size and potentially false positive results in many meta‐analyses. Journal of Clinical Epidemiology 2008;61(8):763‐9.

Brok 2009

Brok J, Thorlund K, Wetterslev J, Gluud C. Apparently conclusive meta‐analyses may be inconclusive ‐ Trial sequential analysis adjustment of random error risk due to repetitive testing of accumulating data in apparently conclusive neonatal meta‐analyses. International Journal of Epidemiology 2009;38(1):287‐98.

Buscemi 2007a

Buscemi N, Vandermeer B, Friesen C, Bialy L, Tubman M, Ospina M, et al. The efficacy and safety of drug treatments for chronic insomnia in adults: a meta‐analysis of RCTs. Journal of General Internal Medicine 2007;22(9):1335‐50.

Chan 2015

Chan AW, Tetzlaff JM, Altman DG, Laupacis A, Gotzsche PC, Krle A‐Jeric K, et al. SPIRIT 2013 Statement: defining standard protocol items for clinical trials. Revista Panamericana de Salud Publica(Pan American Journal of Public Health) 2015;38(6):506‐14. [PUBMED: 27440100]

CTU 2011

Copenhagen Trial Unit. TSA ‐ Trial Sequential Analysis. ctu.dk/tsa/ (accessed 25 March 2017).

Darker 2015

Darker CD, Sweeney BP, Barry JM, Farrell MF, Donnelly‐Swift E. Psychosocial interventions for benzodiazepine harmful use, abuse or dependence. The Cochrane Database of Systematic Reviews 2015, Issue 5. [DOI: 10.1002/14651858.CD009652.pub2]

Deeks 2011

Deeks JJ, Higgins JPT, Altman DG (editors). Chapter 9: Analysing data and undertaking meta‐analyses. In: Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Dell'osso 2013

Dell'osso B, Lader M. Do benzodiazepines still deserve a major role in the treatment of psychiatric disorders? A critical reappraisal. European Psychiatry: the Journal of the Association of European Psychiatrists 2013;28(1):7‐20. [PUBMED: 22521806]

DeMets 1987

DeMets DL. Methods for combining randomized clinical trials: strengths and limitations. Statistics in Medicine 1987;6(3):341‐50.

Denis 2006

Denis C, Fatseas M, Lavie E, Auriacombe M. Pharmacological interventions for benzodiazepine mono‐dependence management in outpatient settings. Cochrane Database of Systematic Reviews 2006, Issue 3. [DOI: 10.1002/14651858.CD005194.pub2]

DerSimonian 1986

DerSimonian R, Laird N. Meta‐analysis in clinical trials. Controlled Clinical Trials 1986;7(3):177‐88.

Egger 1997

Egger M, Davey Smith G, Schneider M, Minder C. Bias in meta‐analysis detected by a simple, graphical test. BMJ (Clinical Research Ed.) 1997;315(7109):629‐34.

Gallacher 2012

Gallacher J, Elwood P, Pickering J, Bayer A, Fish M, Ben‐Shlomo Y. Benzodiazepine use and risk of dementia: evidence from the Caerphilly Prospective Study (CaPS). Journal of Epidemiology and Community Health 2012;66(10):869‐73.

Gisev 2011

Gisev N, Hartikainen S, Chen TF, Korhonen M, Bell JS. Mortality associated with benzodiazepines and benzodiazepine‐related drugs among community‐dwelling older people in Finland: a population‐based retrospective cohort study. Canadian Journal of Psychiatry 2011;56(6):377‐81.

Glass 2005

Glass J, Lanctot KL, Herrmann N, Sproule BA, Busto UE. Sedative hypnotics in older people with insomnia: meta‐analysis of risks and benefits. BMJ (Clinical Research Ed.) 2005;331(7526):1169.

GRADE

ims.cochrane.org/revman/gradepro.

Hausken 2007

Hausken AM, Skurtveit S, Tverdal A. Use of anxiolytic or hypnotic drugs and total mortality in a general middle‐aged population. Pharmacoepidemiology and Drug Safety 2007;16(8):913‐8.

Higgins 2011

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

Huthwaite 2013

Huthwaite MA, Andersson V, Stanley J, Romans SE. Hypnosedative prescribing in outpatient psychiatry. International Clinical Psychopharmacology 2013;28(4):157‐63.

ICH GCP

International Council for Harmonisation (ICH) Topic E 6 (R1) Guideline for Good Clinical Practice. ichgcp.net/pdf/ich‐gcp‐en.pdf (accessed prior to 10 February 2018).

Ioannidis 2004

Ioannidis JP, Evans SJ, Gotzsche PC, O'Neill RT, Altman DG, Schulz K, et al. Better reporting of harms in randomized trials: an extension of the CONSORT statement. Annals of Internal Medicine 2004;141(10):781‐8. [PUBMED: 15545678]

Ioannidis 2009

Ioannidis JP. Adverse events in randomized trials: neglected, restricted, distorted, and silenced. Archives of Internal Medicine2009; Vol. 169, issue 19:1737‐9. [PUBMED: 19858427]

Islam 2014

Islam MM, Conigrave KM, Day CA, Nguyen Y, Haber PS. Twenty‐year trends in benzodiazepine dispensing in the Australian population. Internal Medicine Journal 2014;44(1):57‐64.

Jakobsen 2014

Jakobsen JC, Wetterslev J, Winkel P, Lange T, Gluud C. Thresholds for statistical and clinical significance in systematic reviews with meta‐analytic methods. BMC Medical Research Methodology 2014;14:120. [PUBMED: 25416419]

Jaussent 2013

Jaussent I, Ancelin ML, Berr C, Peres K, Scali J, Besset A, et al. Hypnotics and mortality in an elderly general population: a 12‐year prospective study. BMC Medicine 2013;11:212.

Jones 2014

Jones CM, Paulozzi LJ, Mack KA. Alcohol involvement in opioid pain reliever and benzodiazepine drug abuse‐related emergency department visits and drug‐related deaths ‐ United States, 2010. MMWR. Morbidity and Mortality Weekly Report 2014;63(40):881‐5. [PUBMED: 25299603]

Kripke 1998

Kripke DF, Klauber MR, Wingard DL, Fell RL, Assmus JD, Garfinkel L. Mortality hazard associated with prescription hypnotics. Biological Psychiatry 1998;43(9):687‐93.

Kripke 2012

Kripke DF, Langer RD, Kline LE. Hypnotics' association with mortality or cancer: a matched cohort study. BMJ Open 2012;2(1):e000850.

Lefebvre 2011

Lefebvre C, Manheimer E, Glanville. Chapter 6: Searching for studies. In: Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Lundh 2017

Lundh A, Lexchin J, Mintzes B, Schroll JB, Bero L. Industry sponsorship and research outcome. Cochrane Database of Systematic Reviews 2017, Issue 2. [DOI: 10.1002/14651858.MR000033.pub3; PUBMED: 28207928]

Macaskill 2001

Macaskill P, Walter SD, Irwig L. A comparison of methods to detect publication bias in meta‐analysis. Statistics in Medicine 2001;20(4):641‐54.

Mallon 2009

Mallon L, Broman JE, Hetta J. Is usage of hypnotics associated with mortality?. Sleep Medicine 2009;10(3):279‐86.

Manchikanti 2008

Manchikanti L, Singh A. Therapeutic opioids: a ten‐year perspective on the complexities and complications of the escalating use, abuse, and nonmedical use of opioids. Pain Physician 2008;11(2 Suppl):S63‐88. [PUBMED: 18443641]

Manchikanti 2012

Manchikanti L, Helm S, Fellows B, Janata JW, Pampati V, Grider JS, et al. Opioid epidemic in the United States. Pain Physician 2012;15(3 Suppl):ES9‐38. [PUBMED: 22786464]

O'Brien 2005

O'Brien CP. Benzodiazepine use, abuse, and dependence. Journal of Clinical Psychiatry 2005;66 Suppl 2:28‐33.

Parr 2009

Parr JM, Kavanagh DJ, Cahill L, Mitchell G, McD Young R. Effectiveness of current treatment approaches for benzodiazepine discontinuation: a meta‐analysis. Addiction 2009;104(1):13‐24.

Parrino 1996

Parrino L, Terzano MG. Polysomnographic effects of hypnotic drugs. A review. Psychopharmacology 1996;126(1):1‐16.

Paulose‐Ram 2007

Paulose‐Ram R, Safran MA, Jonas BS, Gu Q, Orwig D. Trends in psychotropic medication use among U.S. adults. Pharmacoepidemiology and Drug Safety 2007;16(5):560‐70.

RevMan 2014 [Computer program]

The Nordic Cochrane Centre, The Cochrane Collaboration. Review Manager (RevMan). Version 5.3. Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboration, 2014.

Schroll 2015

Schroll JB, Bero L. Regulatory agencies hold the key to improving Cochrane Reviews of drugs [editorial]. Cochrane Database of Systematic Reviews2015; Vol. 4. [DOI: 10.1002/14651858.ED000098; PUBMED: 25904511]

Schulz 2011

Schulz KF, Altman DG, Moher D. CONSORT 2010 statement: updated guidelines for reporting parallel group randomised trials. International Journal of Surgery (London, England) 2011;9(8):672‐7. [PUBMED: 22019563]

Skolnick 2018

Skolnick P. The opioid epidemic: crisis and solutions. Annual Review of Pharmacology and Toxicology 2018;58:143‐59. [PUBMED: 28968188]

Smink 2010

Smink BE, Egberts AC, Lusthof KJ, Uges DR, de Gier JJ. The relationship between benzodiazepine use and traffic accidents: a systematic literature review. CNS Drugs 2010;24(8):639‐53.

Sonnenberg 2012

Sonnenberg CM, Bierman EJ, Deeg DJ, Comijs HC, van Tilburg W, Beekman AT. Ten‐year trends in benzodiazepine use in the Dutch population. Social Psychiatry and Psychiatric Epidemiology 2012;47(2):293‐301.

Thorlund 2009

Thorlund K, Devereaux PJ, Wetterslev J, Guyatt G, Ioannidis JP, Thabane L, et al. Can trial sequential monitoring boundaries reduce spurious inferences from meta‐analyses?. International Journal of Epidemiology 2009;38(1):276‐86.

Thorlund 2010

Thorlund K, Anema A, Mills E. Interpreting meta‐analysis according to the adequacy of sample size. An example using isoniazid chemoprophylaxis for tuberculosis in purified protein derivative negative HIV‐infected individuals. Clinical Epidemiology 2010;2:57‐66.

Thorlund 2011a

Thorlund K, Imberger G, Walsh M, Chu R, Gluud C, Wetterslev J, et al. The number of patients and events required to limit the risk of overestimation of intervention effects in meta‐analysis ‐ a simulation study. PLoS ONE 2011;6(10):e25491.

Thorlund 2011b

Thorlund K, Engstrøm J, Wetterslev J, Brok J, Imberger G, Gluud C. User manual for Trial Sequential Analysis (TSA). ctu.dk/tsa/files/tsa_manual.pdf (accessed 19 March 2014).

Tsimtsiou 2009

Tsimtsiou Z, Ashworth M, Jones R. Variations in anxiolytic and hypnotic prescribing by GPs: a cross‐sectional analysis using data from the UK Quality and Outcomes Framework. British Journal of General Practice 2009;59(563):e191‐8.

Turner 2014

Turner RM, Jackson D, Wei Y, Thompson SG, Higgins JP. Predictive distributions for between‐study heterogeneity and simple methods for their application in Bayesian meta‐analysis. Statistics in Medicine 2014;34(6):984‐98. [PUBMED: 25475839]

Vinkers 2012

Vinkers CH, Olivier B. Mechanisms underlying tolerance after long‐term benzodiazepine use: a future for subtype‐selective GABA(A) receptor modulators?. Advances in Pharmacological Sciences 2012;2012:416864. [PUBMED: 22536226]

Voshaar 2006

Voshaar RC, Couvee JE, van Balkom AJ, Mulder PG, Zitman FG. Strategies for discontinuing long‐term benzodiazepine use: meta‐analysis. British Journal of Psychiatry 2006;189:213‐20.

Warner 2009

Warner M, Chen LH, Makuc DM. Increase in fatal poisonings involving opioid analgesics in the United States, 1999‐2006. NCHS Data Brief 2009, (22):1‐8. [PUBMED: 19796521]

Webster 2011

Webster LR, Cochella S, Dasgupta N, Fakata KL, Fine PG, Fishman SM, et al. An analysis of the root causes for opioid‐related overdose deaths in the United States. Pain Medicine (Malden, Mass.) 2011;12 Suppl 2:S26‐35. [PUBMED: 21668754]

Weich 2014

Weich S, Pearce HL, Croft P, Singh S, Crome I, Bashford J, et al. Effect of anxiolytic and hypnotic drug prescriptions on mortality hazards: retrospective cohort study. BMJ (Clinical Research Ed.) 2014;348:g1996.

Wetterslev 2008

Wetterslev J, Thorlund K, Brok J, Gluud C. Trial sequential analysis may establish when firm evidence is reached in cumulative meta‐analysis. Journal of Clinical Epidemiology 2008;61(1):64‐75.

Wetterslev 2009

Wetterslev J, Thorlund K, Brok J, Gluud C. Estimating required information size by quantifying diversity in random‐effects model meta‐analyses. BMC Medical Research Methodology 2009;9:86.

Wetterslev 2017

Wetterslev J, Jakobsen JC, Gluud C. Trial Sequential Analysis in systematic reviews with meta‐analysis. BMC Medical Research Methodology 2017;17(1):39. [PUBMED: 28264661]

Woolcott 2009

Woolcott JC, Richardson KJ, Wiens MO, Patel B, Marin J, Khan KM, et al. Meta‐analysis of the impact of 9 medication classes on falls in elderly persons. Archives of Internal Medicine 2009;169(21):1952‐60.

Wu 2009

Wu CS, Wang SC, Chang IS, Lin KM. The association between dementia and long‐term use of benzodiazepine in the elderly: nested case‐control study using claims data. American Journal of Geriatric Psychiatry 2009;17(7):614‐20.

Zandstra 2004

Zandstra SM, Van Rijswijk E, Rijnders CA, Van De Lisdonk EH, Bor JH, Van Weel C, et al. Long‐term benzodiazepine users in family practice: differences from short‐term users in mental health, coping behaviour and psychological characteristics. Family Practice 2004;21(3):266‐9.

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Ashton 1990

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 12 weeks

Single‐centre

Participants

Baseline characteristics

Buspirone

  • Years of benzodiazepine use, mean (SD): 9.5 (5.6)

  • Male, N (%): 3 (27.3)

  • Age, mean (SD): 39.8 (10.2)

  • Benzodiazepine dose (mg diazepam equivalents), mean (SD): 15.5 (8.2)

Placebo

  • Years of benzodiazepine use, mean (SD): 11.25 (4.47)

  • Male, N (%): 6 (50.0)

  • Age, mean (SD): 43.4 (10.9)

  • Benzodiazepine dose (mg diazepam equivalents), mean (SD): 7.5 (4.6)

Inclusion criteria: above 18 years of age, continuous benzodiazepine therapy for a minimum of 6 months, wish to withdraw from benzodiazepines

Exclusion criteria: use of other psychotropic medication, abuse of alcohol or drugs, major psychiatric or physical disease

Pretreatment group differences: Mean benzodiazepine dosage at baseline was 15.5 mg in buspirone group and 7.5 mg in placebo group.

Interventions

Benzodiazepine taper schedule: all participants switched to an equivalent dose of diazepam, stable dosage for 4 weeks, then taper with 25% each week for 4 weeks to 0, then 4 weeks without benzodiazepines.

  1. Benzodiazepine taper schedule + buspirone 5 mg 3 times a day (N = 11).

  2. Benzodiazepine taper schedule + placebo (N = 12).

Outcomes

  • Benzodiazepine withdrawal symptoms: Ashton scale

  • Benzodiazepine cessation

  • Relapse to benzodiazepine use

  • Anxiety: Hospital Anxiety Depression Scale

Identification

Sponsorship source: Bristol Myers CNS provided buspirone and placebo tablets and covered laboratory and administrative expenses.

Country: UK

Setting: Outpatients, participants referred from their GP, rapid benzodiazepine tapering regimen

Declarations of interest: Not mentioned

Author's name: Ashton CH

Institution: Department of Pharmacological Sciences, University of Newcastle upon Tyne, NE2 4HH

Email:

Address: Department of Pharmacological Sciences, University of Newcastle upon Tyne, NE2 4HH

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "patients were randomly assigned..."

Comment: Not further described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Comment: The study was carried out double‐blind using matching placebo tablets.

Quote: "double‐blind...either buspirone or matching placebo tablets..."

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

Comment: No actions to adjust for high dropout (64%) in the intervention group compared with the placebo group (8%).

Selective reporting (reporting bias)

Low risk

Comment: No protocol available, but no reason to suspect selective outcome reporting

Other bias

Unclear risk

Comment: The role of Bristol Myers insufficiently described.

Baandrup 2016

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 24 weeks

Single‐centre

Participants

Baseline characteristics

Pronlonged‐release melatonin (PRM)

  • Years of benzodiazepine use, mean (SD): 10.4 (7.7)

  • Male, N (%): 23 (55)

  • Age, mean (SD): 47.9 (8.7)

  • Benzodiazepine dose (mg diazepam equivalents), mean (SD): 24.5 (20.1)

Placebo

  • Years of benzodiazepine use, mean (SD): 10.5 (6.8)

  • Male, N (%): 25 (57)

  • Age, mean (SD): 49.4 (12.3)

  • Benzodiazepine dose (mg diazepam equivalents), mean (SD): 23.1 (14.1)

Inclusion criteria: Age 18 years or above, an ICD‐10 diagnosis of schizophrenia (F20), schizoaffective disorder (F25), or bipolar disorder (F31). Bipolar patients were required to be euthymic a the time of inclusion. Treatment with antipsychotic drug(s) for at least 3 months before inclusion, treatment with 1 or more benzodiazepine derivatives or benzodiazepine‐related drugs for at least 3 months before inclusion, fertile women: negative pregnancy test at baseline and the use of safe contraceptives (intrauterine devices or hormonal contraception) throughout the trial period, written informed consent.

Exclusion criteria: Known aggressive or violent behaviour, mental retardation, pervasive developmental disorder, or dementia, epilepsy, terminal illness, severe somatic comorbidity, or inability to understand Danish, allergy to compounds in the trial medication (melatonin, lactose, starch, gelatine, and talc), hepatic impairment, pregnancy or nursing, lack of informed consent.

Pretreatment group differences: None

Interventions

Benzodiazepine taper schedule: gradual reduction of usual benzodiazepine dosage (including benzodiazepine‐related drugs) at an approximate rate of 10% to 20% every second week.

  1. Benzodiazepine taper schedule + PRM 2 mg x 1 (N = 42).

  2. Benzodiazepine taper schedule + placebo (N = 44).

Outcomes

  • Benzodiazepine cessation

  • Benzodiazepine mean dose

  • SAEs

  • Non‐serious AEs

  • Discontinuation due to AEs

  • Subjective sleep quality

Identification

Sponsorship source: The Research Fund of the Mental Health Services of the Capital Region in Denmark financed the trial with a post doc grant and a grant for external randomisation and database management.

Country: Denmark

Setting: Mainly outpatients

Declarations of interest: None

Author's name: Baandrup L

Institution: Centre for Neuropsychiatric Schizophrenia Research (CNSR) & Centre for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), University of Copenhagen, Mental Health Centre Glostrup, Mental Health Services – Capital Region of Denmark, Glostrup

Email: [email protected]

Address: Mental Health Centre Glostrup, Mental Health Services – Capital Region of Denmark, DK‐2600 Glostrup

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Central randomisation was performed by the Copenhagen Trial Unit (CTU) with computer‐generated, permuted randomisation allocation sequence"

Allocation concealment (selection bias)

Low risk

Quote: "The allocation sequence and block sizes were kept unknown to the investigator. Allocation ratio was 1:1. The investigator contacted the CTU and provided a personal pin code, participant civil registration number, participant trial identification number, and the value of the stratification variable of benzodiazepine dosage (low (15 mg diazepam equivalents) or high (15 mg diazepam equivalents)) at baseline. Then the randomisation was announced as a trial medication container number and confirmation sent by e‐mail"

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Thus, the placebo was matched to the study medication for taste, smell, colour, size and solubility. CTU held the randomisation code and the trial was not unblinded until all data were registered, primary analyses finished and conclusions drawn"

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "Trial participants, staff, and outcome assessors were blinded to the allocated treatment. We maintained blinding using matching placebo and an independent unit to perform the randomisation and do the packaging and labelling of the trial medication. Both PRM and placebo were encapsulated in lactose‐ containing gelatine capsules to optimise the blinding"

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: Data complete for the primary outcome.

Selective reporting (reporting bias)

Low risk

Comment: Primary outcome, etc. reported in published trial protocol.

Other bias

Low risk

Comment: No other apparent source of bias

Cassano 1996

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 6 weeks (4 weeks double‐blind followed by 2 weeks single‐blind placebo)

Multicentre

Participants

Baseline characteristics

Alpidem

  • Male, N (%): 33 (37.9)

  • Age, mean (SD): 45.5 (11.3)

  • Years of benzodiazepine use, mean (SD): 4.7

Placebo

  • Male, N (%): 29 (33.7)

  • Age, mean (SD): 43.9 (11.4)

  • Years of benzodiazepine use, mean (SD): 5.1

Inclusion criteria: Outpatients with generalised anxiety disorder (GAD; DSM‐III‐R, item 300.02) or adjustment disorder with anxious mood (DSM‐III‐R, item 309.24). Consecutive patients of either sexes, aged between 18 and 60 years, taking non‐hypnotic benzodiazepines for anxiety as continuous course of therapy of at least 1 year duration, at a dose schedule corresponding to 30 mg or less of diazepam per day, were considered eligible.

Exclusion criteria: Montgomery–Åsberg Depression Rating Scale was administered to exclude depressed patients (total score > 18).

Pretreatment: No significant differences

Interventions

Intervention characteristics

Benzodiazepine taper schedule: all benzodiazepines abruptly discontinued at inclusion.

  1. Benzodiazepine taper schedule + alpidem 100 to 150 mg/d(N = 87).

  2. Benzodiazepine taper schedule + placebo(N = 86).

Outcomes

  • Anxiety: HAM‐A

  • Non‐serious adverse events

  • Withdrawal syndrome (clinical diagnosis)

Identification

Sponsorship source: No information

Country: Italy

Setting: Outpatients

Declarations of interest: Not mentioned

Author's name: Cassano GB

Institution: Clinica Psichiatrica, University degli Studi di Pisa

Email:

Address: Clinica Psichiatrica, University degli Studi di Pisa, Ospedale Santa Chiara, Via Roma 67, 56100 Pisa

Notes

The study lasted 6 weeks: a 4‐week comparative period (phase I) to prevent and treat benzodiazepine withdrawal symptoms (primary aim) was followed by a 2‐week single‐blind period with placebo (phase II) to monitor the occurrence of withdrawal symptoms after abrupt discontinuation of alpidem (secondary aim). 6 weeks was chosen as endpoint because alpidem is a Z‐drug. According to the review protocol, such studies are included if data are available on relevant outcomes AFTER withdrawal of the new benzodiazepine/Z‐drug, in this case after discontinuation of alpidem.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Quote: "The Italian multicentre (15 centres), double‐blind, randomised (versus placebo), parallel group study"

Comment: What has been done to ensure blinding of participants and study personnel is not described.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "The diagnosis of withdrawal symptoms was made by a respected academic expert, in blind conditions, on the basis of the definition in the protocol"

Comment: Done

Incomplete outcome data (attrition bias)
All outcomes

High risk

Comment: 10 (11.5%) discontinued in the alpidem group, 18 (21%) in the placebo group.

Selective reporting (reporting bias)

Low risk

Comment: Protocol published but could not be retrieved. No reason to suspect selective outcome reporting

Other bias

Unclear risk

Comment: Source of financing not described.

Cialdella 2001

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 1 month

Single‐centre

Participants

Baseline characteristics

Homéogène

  • Male, N (%): 4 (26.7)

  • Age, mean (SD): 52.9 (12.8)

  • Employed, N (%): 8 (53.3)

  • Benzodiazepine dose (diazepam equivalent), mean (SD): 4.5 (6.5)

Sédatif PC

  • Male, N (%): 4 (20.0)

  • Age, mean (SD): 50.7 (11.9)

  • Employed, N (%): 8 (40.0)

  • Benzodiazepine dose (diazepam equivalent), mean (SD): 4.2 (4.7)

Placebo

  • Male, N (%): 11 (42.3)

  • Age, mean (SD): 58.2 (15.3)

  • Employed, N (%): 7 (26.9)

  • Benzodiazepine dose (diazepam equivalent), mean (SD): 2.4 (2.6)

Inclusion criteria: At least 18 years of age, at least 3 months use of benzodiazepines at low dosage (max 10 mg/day diazepam equivalents), clinically stable for at least 1 month

Exclusion criteria: Severe insomnia, severe psychiatric disorders, alcohol or substance abuse disorder, previous seizures, current use of muscle relaxants, clonidine, or psychotropic drugs.

Pretreatment: Higher scores on somatic symptoms in Homéogène group

Interventions

Benzodiazepines substituted (no taper schedule) with study drug:

  1. Homéogène 6 tablets/day(N = 15)

  2. Sédatif PC 6 tablets/day(N = 20)

  3. placebo(N = 26)

Both experimental drugs were homeopathic drugs.

Outcomes

  • Benzodiazepine cessation

  • Hamilton Anxiety Rating Scale (HAM‐A)

  • Benzodiazepine Withdrawal Symptom Questionnaire (BWSQ)

Identification

Sponsorship source: Laboratoires Boiron, l'Agence Nationale de Valirisation de la Recherce

Country: France

Setting: Outpatients

Declarations of interest: Not mentioned

Author's name: Cialdella P

Institution: Service de Pharmacologie Clinique, Faculté RTH Laënnec

Email:

Address:

Notes

Homéogène and Sédatif groups were combined as 1 homeopathic drug group.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Comment: Described as double‐blind, but lacks a description of what have been done to ensure blinding of participants and study personnel

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: Not described. Insufficient information to permit judgement of low or high risk of bias

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Comment: 25% attrition. An ITT approach was used, but distribution of attrition between groups was not reported.

Selective reporting (reporting bias)

Low risk

Comment: No obvious selective outcome reporting

Other bias

Unclear risk

Comment: Role of funding source not described, both industry and publicly funded.

Di Costanzo 1992

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 4 weeks

Single‐centre

Participants

Baseline characteristics

Carbamazepine

  • Benzodiazepine dose (diazepam equivalent), mean (SD): 18.7 (7.9)

Placebo

  • Benzodiazepine dose (diazepam equivalent), mean (SD): 19.4 (9.5)

Inclusion criteria: > 60 years of age, GAD, benzodiazepine abuse, minimum duration of benzodiazepine treatment 6 months

Exclusion criteria: None described.

Pretreatment: No significant pretreatment differences

Interventions

Benzodiazepine taper schedule: 25% benzodiazepine dose reduction every week

  1. Benzodiazepine taper schedule + carbamazepine dose adjusted to serum level 6 to 8 mcg/mL(N = 15)

  2. Benzodiazepine taper schedule + placebo(N = 14)

Outcomes

  • Benzodiazepine withdrawal symptoms: Physician Withdrawal Checklist

  • Benzodiazepine cessation

  • Discontinuation due to adverse events

  • Anxiety: HAM‐A

  • Relapse to benzodiazepine use

  • Serious adverse events

  • Non‐serious adverse events

Identification

Sponsorship source: Not reported

Country: Italy

Setting: Outpatients

Declarations of interest: Not mentioned

Author's name: Di Constanzo E

Institution: Servizio Psichiatrico

Email:

Address: Viale Spellanzon, 55 31015 Conegliano

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Comment: Described as double‐blind, but what has been done to ensure blinding of participants and study personnel is not mentioned

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: Insufficient information to permit judgement of low or high risk

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: 4 (26.6%) and 3 (21.4%) participants did not complete benzodiazepine cessation but participated in the study.

Selective reporting (reporting bias)

Low risk

Comment: No indication of selective outcome reporting

Other bias

Low risk

Comment: No apparent other source of bias

Garfinkel 1999

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 6 weeks double‐blind, 6 weeks single‐blind

Single‐centre

Participants

Baseline characteristics

Controlled‐release melatonin

  • Male, N (%): 4 (22)

  • Age, mean (SD): 69 (11)

  • Number of benzodiazepine tablets, mean (SD): 1.08 (0.38)

Placebo

  • Male, N (%): 5 (31)

  • Age, mean (SD): 68 (16)

  • Number of benzodiazepine tablets, mean (SD): 1.23 (0.61)

Inclusion criteria: People with a daily use of benzodiazepines for more than 6 months, expressed willingness to discontinue the use, living independently

Exclusion criteria: Cognitive impairment, liver or renal disorders

Pretreatment: No significant differences

Interventions

Intervention characteristics

Benzodiazepine taper schedule: participants were encouraged to reduce their usual benzodiazepine therapy dosage 50% during week 2, 75% during weeks 3 and 4, and then to discontinue benzodiazepine therapy completely during weeks 5 and 6. Participants who did not succeed in stopping benzodiazepine therapy during period 1 were encouraged to further reduce benzodiazepine dosage 50%, 75%, and 100% during weeks 8, 9 and 10, 11 and 12, respectively.

  1. Benzodiazepine taper schedule + controlled‐release melatonin 2 mg/d (2 hours before bedtime)(N = 18)

  2. Benzodiazepine taper schedule + placebo(N = 16)

Outcomes

  • Benzodiazepine cessation

  • Sleep quality (scale 1 to 10, higher = better)

Identification

Sponsorship source: Neurim Pharmaceuticals sponsored study medication and study nurse; statistical evaluations performed independently.

Country: Israel

Setting: Outpatients, living independently

Declarations of interest: Not mentioned

Authors name: Doron Garfinkel

Institution: Department of Neurobiochemistry, Tel Aviv University

Email: [email protected]

Address: Tel Aviv 69978, Israel

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Subjects were randomised to receive either 2 mg of CRM therapy or a placebo that was identical in appearance"

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "Collection and entry of all data were completed before revealing the randomisation codes of the study"

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: All included participants analysed.

Selective reporting (reporting bias)

Low risk

Comment: No indications of selective reporting

Other bias

High risk

Comment: The trial was partly financed by a company with an interest in given result, the company's role in interpreting the data is not sufficiently described.

Gerra 1993

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group, stratifies for flunitrazepam/lormetazepam at baseline

Blinding: Single

Duration: 7 days

Single‐centre

Participants

Baseline characteristics

Not reported

Inclusion criteria: 18 to 40 years of age, flunitrazepam abuse at a dose of 10 to 12 mg/day (18 participants) or lormetazepam abuse at a dose of 8 to 10 mg/day (18 participants). All participants met the criteria of the DSM‐III‐R for benzodiazepine withdrawal syndrome. Abuse was defined as use for at least 9 months. Informed consent was obtained from all participants.

Excluded criteria: Psychiatric patients were not included in the study. Daily urine samples were taken to rule out the abuse of morphine, methadone, cocaine, amphetamine, barbiturates, cannabis, and ethanol during the study.

Pretreatment: Not reported

Interventions

Intervention characteristics

Benzodiazepine taper schedule: abrupt cessation

  1. Benzodiazepine taper schedule + flumazenil 0.5 mg IV x 4/d days 1 to 4 and 0.5 mg x 2/d days 5 to 7. N = 18 (9 flunitrazepam abusers and 9 lormetazepam abusers)

  2. Benzodiazepine taper schedule + placebo (saline solution). N = 18 (9 flunitrazepam abusers and 9 lormetazepam abusers)

Outcomes

  • Serious adverse events

  • Non‐serious adverse events

  • Anxiety, HAM‐D

  • Benzodiazepine withdrawal symptoms (score 0 to 45)

  • Discontinuation due to adverse events

  • Benzodiazepine cessation

Identification

Sponsorship source: Not reported

Country: Italy

Setting: Inpatients

Declarations of interest: Not mentioned

Author's name: Gilberto Gerra

Institution: University of Parma

Email:

Address: USL n. 4, Via Guasti S. Cecilia, 3, Parma 43100, Italy

Notes

Results were reported separately for flunitrazepam and lormetazepam users. To avoid including several comparisons from the same study, we only included results for the lormetazepam users in this meta‐analysis (flunitrazepam is now very seldom used in clinical practice and in many countries is no longer registered for use).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Insufficient information

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Quote: "Placebo groups B and D were only treated with saline solution for 7 days."

Comment: Only participants were blinded.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Comment: Study only described as single‐blinded, therefore probably not done.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: All participants analysed.

Selective reporting (reporting bias)

Low risk

Comment: No indication of selective outcome reporting

Other bias

Unclear risk

Comment: Insufficient information regarding sponsorship

Gerra 2002

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Single

Duration: 8 days

Single‐centre

Participants

Baseline characteristics

Flumazenil IV

  • Male, N (%): 9 (45)

  • Age, mean (SD): 35.9

Oxazepam tapering

  • Male, N (%): 11 (55)

  • Age, mean (SD): 38.2

Placebo

  • Male, N (%): 6 (60)

  • Age, mean (SD): 35.4

Inclusion criteria: History of benzodiazepine dependence according to DSM‐IV criteria.

Exclusion criteria: Severe chronic liver or renal diseases or other chronic physical disorders, recent onset of significant weight loss or gain, endocrinopathies, neurological disorders, immunopathy, in particular HIV disease, a positive family history of cardiovascular disease and hypertension, current abuse of illicit drugs and alcohol

Pretreatment: None in reported parameters

Interventions

Intervention characteristics

All participants received high doses of oxazepam (120 mg/day) during the last week before detoxification (pretreatment week).

  1. Benzodiazepine cessation + flumazenil 1.0 mg x 2 IV (N = 20)

  2. Oxazepam tapering + placebo (saline solution IV)(N = 20)

  3. Placebo + placebo(N = 10)

Outcomes

  • Benzodiazepine withdrawal symptoms: self reported withdrawal scores

  • Relapse to benzodiazepine use

Identification

Sponsorship source: Not mentioned

Country: Italy

Setting: Inpatients

Declarations of interest: Not mentioned

Author's name: Gilberto Gerra

Institution: Addiction Research Center, Ser.T., AUSL, Parma, Italy

Email: [email protected]

Address: Gilberto Gerra, Centro Studi Farmacotossicodipendenze, Ser.T., A.U.S.L., Via Spalato 2,43100 Parma, Italy

Notes

Only the comparison between flumazenil and placebo was considered relevant and included in the meta‐analysis, cf. Cochrane Handbook on multiple comparisons.

Rate of relapse NOT reported for the placebo group because: (quote) Long‐term outcome of group C (placebo) patients was not evaluated in comparison with A and B patients because they received low‐dose benzodiazepine treatment for 2 weeks, immediately after the detoxification procedure, for ethical reasons.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described

Quote: "The study was single‐blind, randomised and placebo‐controlled."

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Quote: "the trial was single‐blind, permitting direct clinical interventions in the case of dramatic withdrawal symptoms"

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Comment: Not done

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: Though not clearly described, judging from the text it appears that no participants withdrew during the 8‐day intervention trial.

Selective reporting (reporting bias)

Low risk

Comment: No reason to suspect selective outcome reporting

Other bias

Unclear risk

Comment: Funding not described.

GlaxoSmithKline 2002

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 12 weeks

Multicentre

Participants

Baseline characteristics

Paroxetine

  • Male, N (%): 10 (33)

  • Age, mean (SD): 51.8 (17.6)

Placebo

  • Male, N (%): 11 (46)

  • Age, mean (SD): 46.3 (17.9)

Inclusion criteria: Participants were males or females aged > 18 years suffering from 1 or more of the following anxiety disorders of non‐severe degree in axis I: panic attack disorder (with or without agoraphobia), GAD, social anxiety/social phobia or mixed anxiety and depression disorder with significant anxiety; people continuously treated with benzodiazepines (any) for at least 6 consecutive months prior to the screening visit at doses between 2 and 8 mg/day of lorazepam or equivalent; a total score ≤ 16 on the HAM‐A and MADRS at screening and baseline.

Exclusion criteria: People suffering (or diagnosed within the 6 months prior to screening) from 1 or more of the following conditions: major depressive episode; post‐traumatic stress disorder; obsessive‐compulsive disorder; eating behavioural disorders, people diagnosed with dysthymia or who had suffered from dysthymia in the 6 months prior to screening; people with a concomitant psychotic disorder, or history of psychotic disorder; people having a concomitant bipolar disorder or history of bipolar disorder, or having a cyclothymic disorder, or had suffered from it in the past; people who met DSM‐IV (protocol appendix O) criteria for substance (alcohol or drugs) abuse or dependence, except for benzodiazepine, within 6 months prior to screening; current suicidal or homicidal risk; and people who had electroconvulsive therapy in the 3 months prior to screening.

Pretreatment: No significant group differences

Interventions

Intervention characteristics

Benzodiazepine taper schedule: 4‐week open‐label run‐in period during which participants were switched from their original benzodiazepine to an equivalent dosage of chlordemethyldiazepam (between 2 and 8 mg/d). The taper schedule during the treatment phase not described.

  1. Benzodiazepine taper + paroxetine 10 mg/d for the first week, 10 to 20 mg/d during weeks 2 to 8, 20 mg/d during weeks 9 to 12(N = 30)

  2. Benzodiazepine taper + placebo(N = 24)

Outcomes

  • Serious adverse events

  • Anxiety: HAM‐A

  • Non‐serious adverse events

  • Relapse to benzodiazepine use

  • Benzodiazepine withdrawal symptoms: BWSQ

  • Benzodiazepine cessation

Identification

Sponsorship source: GlaxoSmithKline

Country: Italy

Setting: Outpatients

Declarations of interest: Not mentioned

Comments: Unpublished phase III study

Author's name: GlaxoSmithKline

Institution:

Email:

Address: Clinical Study Register (www.gskclinicalstudyregister.com) 2002

Notes

Change scores extracted, final scores not available. Standard deviation calculated from CI using the following formula:

SE = (upper limit – lower limit of CI)/3.92

Standard deviation σ = standard error x √n

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "12‐week double blind, multicentre, randomised, placebo‐controlled, parallel group"

Comment: Not described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Subjects were randomised to either paroxetine or placebo and entered the 12‐week double‐blind, randomised treatment phase. Dosage of paroxetine or matched placebo started with..."

Comment: Double‐blind and using matched placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: 2 versus 8 participants withdrew, but ITT analysis data extracted for this meta‐analysis.

Selective reporting (reporting bias)

High risk

Comment: No protocol available, benzodiazepine dose at follow‐up not described.

Other bias

High risk

Comment: Study funded by the study drug manufacturer, no information available on involvement in design, data collection, etc.

Hadley 2012

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 12 weeks (6 weeks during tapering and 6 weeks post‐tapering)

Multicentre

Participants

Baseline characteristics

Pregabalin

  • Male, N (%): 14 (25)

  • Age, mean (SD): 40.1 (10.6)

Placebo

  • Male, N (%): 16 (32)

  • Age, mean (SD): 43.5 (11.3)

Inclusion criteria: Adult outpatients aged 18 to 65 years were enrolled if they met DSM‐IV criteria for a primary lifetime diagnosis of GAD, and if they were receiving stable treatment with a benzodiazepine in daily doses of 1 to 4 mg/day (in alprazolam dose equivalents) for 8 to 52 weeks. A primary diagnosis of GAD was made, based on predominant clinical presentation, using the module P form of the Mini International Neuropsychiatric Interview (MINI)‐Plus version 5.0.0 (Sheehan et al, 1997). The current diagnosis of GAD could be sub‐threshold due to treatment.

Exclusion criteria: (1) women who were pregnant, lactating, or of childbearing potential who were not using a medically approved form of contraception; (2) 17‐item HAM‐D total score > 15; (3) a history of anxiolytic non‐response to benzodiazepines or pregabalin, or hypersensitivity to either class of drug; (4) they met DSM‐IV criteria in the past 6 months of major depressive disorder, dysthymia, social phobia, post‐traumatic stress disorder, body dysmorphic disorder, or eating disorder; (5) met DSM‐IV criteria in the past 5 years of schizophrenia, psychotic disorder, bipolar affective disorder, obsessive–compulsive disorder, substance dependence (excluding nicotine), or in the past year for substance abuse; (6) currently receiving cognitive behavioural therapy for GAD or other anxiety disorder; (7) a history of seizure disorder, except febrile seizures of childhood; (8) a history of neuropathic pain or narrow angle glaucoma; (9) receiving treatment with fluoxetine (in past 5 weeks) or any psychotropic other than benzodiazepines (in past 2 weeks), or electroconvulsive therapy (in past 6 months); (10) positive urine drug screen for amphetamines, barbiturates, ethanol, narcotics, non‐benzodiazepine sedatives and hypnotics, cocaine, phencyclidine, cannabinoids or other illegal or illicit drugs; (11) considered by the investigator to be at risk for suicide or aggressive behaviour; (12) any serious or uncontrolled medical illness in the opinion of the investigator that would render the person unsuitable for the study; or (13) creatinine clearance 60 mL/min.

Pretreatment: None

Interventions

Intervention characteristics

Benzodiazepine taper schedule: switch to equivalent dose alprazolam, 2‐week stabilisation phase before randomisation, 25% reduction per week, permitted up to 6 weeks to complete the alprazolam taper, after maintained 6 weeks on double‐blind treatment, then 1 week taper off study medication

  1. Benzodiazepine taper schedule + pregabalin 150 to 600 mg/d according to tolerability and efficacy(N = 56).

  2. Benzodiazepine taper schedule + placebo(N = 50).

Outcomes

  • Benzodiazepine cessation

  • Anxiety, HAM‐A

  • Non‐serious adverse events

  • Serious adverse events

  • Benzodiazepine withdrawal symptoms, PWC

Identification

Sponsorship source: Funded by Pfizer

Country: 20 investigational sites in Spain, Mexico, France, Italy, Costa Rica, the Czech Republic, and Guatemala

Setting: Outpatients

Declarations of interest: Dr Schweizer was at the time of the writing of the manuscript employee of Paladin Consulting Group Inc., which was a paid consultancy to Pfizer Inc. At the time the study was conducted and the paper was initially drafted, Dr Sallie J Hadley was an employee of Pfizer Inc. and owns stock in Pfizer. Dr Francine S Mandel was a full‐time employee of Pfizer Inc. Dr Edward Schweizer owns stock in Pfizer and has received payments for consulting and/or medical writing services from Alkermes, Bristol‐Myers Squibb, Sumitomo Dainippon Pharma, Eli Lilly, Memory Pharmaceuticals, Neurocrine Biosciences, and Pfizer Inc.

Author's name: Sallie J Hadley

Institution: Pfizer Inc., New York, NY, USA

Email: [email protected]

Address: Francine S Mandel, Pfizer Inc., 235 East 42nd Street, New York, NY, USA

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Quote: "Patients...were randomised on a one‐to‐one basis to 12 weeks of double‐blind treatment with either pregabalin or placebo"

Comment: Described as "double‐blind" but what has been done to ensure blinding of participants and study personnel is not mentioned.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

Comment: High attrition rate in both the pregabalin group (46.4%) and the placebo group (62.0%)

Selective reporting (reporting bias)

Low risk

Comment: No apparent selective outcome reporting

Other bias

High risk

Comment: Study funded by Pfizer, no indication of role of funding body in design, data collection, analysis, and interpretation of the data.

Hantouche 1998

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 3 months

Multicentre

Participants

Baseline characteristics

Magnesium aspartate

  • Male, N (%): 17 (29)

  • Age, mean (SD): 44.1 (10.8)

  • Employed, N (%): 45 (77)

Placebo

  • Male, N (%): 20 (27)

  • Age, mean (SD): 44.7 (12.1)

  • Employed, N (%): 55 (73)

Inclusion criteria: Outpatients, 18 to 65 years of age, chronic users of lorazepam, alprazolam, or bromazepam (> 6 months, regular dose => 3 mg lorazepam equivalents), benzodiazepines prescribed due to an anxious disorder now in remission defined as score on Hamilton Anxiety < 14 and Raskin‐Depression < 6, no major psychiatric disorder, at least 1 trial of unsuccessful benzodiazepine withdrawal, a wish to discontinue benzodiazepine use

Exclusion criteria: Severe hepatic or renal dysfunction, alcohol or substance use disorder, currently trying to discontinue use of tobacco, current psychotherapy, use of other psychotropics within 6 months, treatment with a magnesium salt or calcium within 1 month, regular use of magnesium aspartate during 1 month within the last 6 months

Pretreatment: No significant pretreatment group differences

Interventions

Benzodiazepine taper schedule: co‐administration of benzodiazepine and study drug for 1 month, gradual taper of benzodiazepine during the next month (50% of dosage for 2 weeks, 25% for 2 weeks, then stop), follow‐up during a third month after complete benzodiazepine discontinuation

  1. Benzodiazepine taper schedule + magnesium aspartate 2 capsules x 3 (300 mg magnesium/day)(N = 69)

  2. Benzodiazepine taper schedule + placebo(N = 75)

Outcomes

  • Benzodiazepine cessation

  • Anxiety

  • Non‐serious AEs

  • Relapse to benzodiazepine use

  • Discontinuation due to AEs

Identification

Sponsorship source: Not reported

Country: France

Setting: Outpatients

Declarations of interest: Not mentioned

Author's name: Hantouche EG

Institution: Département de Psychiatrie, Groupe Hospitaliers de la Pitrie‐Salpetriere

Email:

Address: 47, Boulevard de l'Hopital, 75013 Paris

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Comment: Described as double‐blind, what has been done to ensure blinding of participants and study personnel is not mentioned

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Comment: Not described

Selective reporting (reporting bias)

Low risk

Comment: No indications of reporting bias

Other bias

Low risk

Comment: No apparent other sources of bias

Harrison‐Read 1996

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 3 weeks

Single‐centre

Participants

Baseline characteristics

Flumazenil IV challenge

  • Male, N (%): 1 (25)

  • Age, mean (SD): 46

  • Years of benzodiazepine use, mean (SD): 8.25

Placebo

  • Male, N (%): 2 (33)

  • Age, mean (SD): 42.3

  • Years of benzodiazepine use, mean (SD): 8.5

Inclusion criteria: People were recruited to the study if they had been taking benzodiazepines in usual therapeutic doses (< 30 mg per day of diazepam or equivalent) for 3 months or more, and if they had experienced withdrawal problems on discontinuing medication.

Exclusion criteria: (i) regular intake of any other psychotropic medication, (ii) a diagnosis of schizophrenia, epilepsy, or cardiorespiratory disease

Pretreatment: No significant pretreatment differences

Interventions

Intervention characteristics

  1. Flumazenil IV challenge 1 mg injected over 30 s, followed by an individually tailored phased withdrawal schedule which, if followed correctly, would produce complete abstinence (100% dose reduction) after 3 weeks following the challenge test(N = 4)

  2. Placebo (vehicle solution alone) followed by identical benzodiazepine taper schedule(N = 6)

Outcomes

  • Benzodiazepine dose reduction of 70%

  • Serious adverse events

  • Benzodiazepine mean dose

  • Benzodiazepine withdrawal symptoms: BWSQ

  • Non‐serious adverse events

Identification

Sponsorship source: Roche Products Ltd supplied unmarked ampoules of flumazenil and vehicle solution and a grant towards the cost of the project.

Country: UK

Setting: Outpatients (inpatients when receiving flumazenil challenge)

Declarations of interest: Not mentioned

Comments: The study was approved by the local ethics committee but, owing to the unexpectedly severe reactions shown in some participants, it was felt to be unethical to continue with the study after 10 participants had been tested using the original protocol.

Author's name: Harrison‐Read PE

Institution: Academic Unit of Psychiatry, St Charles Hospital, Exmoor Street, London W10 6DZ

Email:

Address: Academic Unit of Psychiatry, St Charles Hospital, Exmoor Street, London W10 6DZ

Notes

Study discontinued due to unacceptable adverse effects (marked panic reaction in the 4 participants who received flumazenil), beginning within 30 seconds of the end of the injection.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "High risk and low risk subjects were allocated separately at random to placebo or flumazenil challenge by an independent pharmacist."

Comment: Description of how the sequence was generated was insufficient.

Allocation concealment (selection bias)

Low risk

Comment: Allocation was done by independent pharmacist.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "This ’challenge test’ was carried out double‐blind, with both subject and experimenter being unaware of the identify of the substance being injected"

Comment: Described as double‐blind and using placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Quote: "Immediately afterwards, the subject began filling in the BWSQ and the MRS, and then repeated these measures at 5, 15, 25, 35, 45 and 60 min post‐injection. Pulse and blood pressure were recorded as before"

Comment: Description is insufficient to judge the risk of bias.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: All randomised participants analysed.

Selective reporting (reporting bias)

Low risk

Comment: No reason to suspect selective outcome reporting

Other bias

High risk

Comment: As the reaction to acute challenge with flumazenil proved to be unexpectedly severe, the study was stopped after only 10 participants had been recruited for the study: 4 were allocated to the flumazenil group and 6 to the placebo group. Despite separately randomising high‐ and low‐risk participants, the early cessation of the study led to unequal distribution between the 2 treatment groups: 1 out of the 4 participants in the flumazenil group and 3 out of the 6 in the placebo group were high‐risk participants. In addition, the study was supported by a company.

Klein 1994

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: Approximately 5 weeks (dependent on duration of taper phase)

Single‐centre

Participants

Baseline characteristics

Carbamazepine: not available

Placebo: not available

Inclusion criteria: DSM‐III‐R diagnosis of panic disorder or generalised anxiety disorder

Exclusion criteria: 1) Lifetime history of psychotic disorder, 2) Bipolar disorder, 3) Seizure disorder, 4) Severe head trauma, 5) Major depression, 6) Abuse of alcohol or other substances, 7) Obsessive‐compulsive disorder, 8) PTSD, 9) Pregnancy, 10) Active systemic illness with chronic medication

Pretreatment: Reported to be non‐significant but not reported for the carbamazepine versus placebo group, only reported for the panic disorder versus the GAD group

Interventions

Intervention characteristics

Benzodiazepine taper schedule: 25% every third day

  1. Benzodiazepine taper schedule + carbamazepine 400 to 800 mg/d(N = 38)

  2. Benzodiazepine taper schedule + placebo(N = 34)

Outcomes

  • Benzodiazepine mean dose

  • Benzodiazepine cessation

Identification

Sponsorship source: Supported by the Upjohn Company

Country: Israel

Setting: Outpatients

Declarations of interest: Not mentioned

Author's name: Ehud Klein

Institution: Rambam Medical Center and University of Vermont

Email:

Address: Rambam Medical Center, Rapapport Faculty of Medicine, Technion‐IIT, Bat Galim, Haifa, Israel

Notes

Baseline characteristics for the carbamazepine versus placebo group were not reported, only for panic disorder group versus generalised anxiety disorder group. Same problem when reporting the results

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described in detail, but it is stated that randomisation was stratified by diagnosis and alprazolam daily dosage

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Patients entered the controlled portion of the study and were randomly assigned, in a double‐blind fashion, to receive either carbamazepine or placebo as adjunctive treatment...In order to maintain blindness of the study throughout the taper period, patients received a fixed number of capsules with a gradually increasing proportion of identical placebo capsules substituting for the alprazolam"

Comment: The use of carbamazepine versus placebo (the primary interest for the current review) was double‐blinded with identical placebo. The alprazolam taper was single‐blind, but these data are not considered here.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: Not sufficiently described

Incomplete outcome data (attrition bias)
All outcomes

High risk

Comment: Very high dropout rates (56% vs 71%), and no ITT analysis performed.

Selective reporting (reporting bias)

High risk

Comment: No reporting on benzodiazepine dosage or withdrawal symptoms

Other bias

High risk

Comment: Role of supporting company not described

Kornowski 2002

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 28 days

Single‐centre

Participants

Baseline characteristics

Carbamazepine

  • Age, mean (SD): 43.29 (6.24)

  • Years of benzodiazepine use, mean (SD): 7.63 (6.91)

  • Benzodiazepine dose (diazepam equivalent), median (range): 27.63 (20.1)

Tianeptine

  • Age, mean (SD): 44.79 (5.18)

  • Years of benzodiazepine use, mean (SD): 7.06 (6.12)

  • Benzodiazepine dose (diazepam equivalent), median (range): 28.45 (28.4)

Inclusion criteria: ICD‐10 criteria for benzodiazepine dependence, 18 to 65 years of age

Exclusion criteria: Previously treated with 1 or both of the experimental drugs, psychotic symptoms, not treated with other psychotropic drugs until 2 weeks before inclusion, pregnant or nursing, substance abuse, severe somatic illness

Pretreatment: No significant pretreatment differences

Interventions

Benzodiazepines substituted with

  1. Carbamazepine 600 mg/day(N = 24)

  2. Tianeptine 37.5 mg/day(N = 24)

Outcomes

  • Benzodiazepine cessation

  • Relapse to benzodiazepine use

  • Serious AEs

Identification

Sponsorship source: Not mentioned

Country: Poland

Setting: Inpatients

Declarations of interest: Not mentioned

Comments: No data reported for the outcomes, only overall results from statistical analyses.

Author's name: Kornowski J

Institution: Psychiatric Hospital in Starogard Gdansk

Email: [email protected]

Address: 83‐200 Starogard Gdansk

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Comment: Blinding not described, not possible to judge whether participants and personnel were blinded, also it is not stated if the study was open‐label.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: 4 (17%) dropped out in each group because they ingested benzodiazepines during the trial(detected by urine screen), but all participants were included in the statistical analyses.

Selective reporting (reporting bias)

Low risk

Comment: No apparent reporting bias

Other bias

Unclear risk

Comment: No apparent other sources of bias

Lader 1987

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 6 weeks

Single‐centre

Participants

Baseline characteristics

Only reported for the total sample: men: 41.7%; age: 39.1 years; years of benzodiazepine use: 8.4 years

Inclusion criteria: More than 6 months of benzodiazepine use, physically dependent, no requirements of further benzodiazepine treatment as deemed by mental state assessment

Exclusion criteria: Abuse of alcohol or other drugs

Pretreatment: Not described

Interventions

Intervention characteristics

Benzodiazepine taper schedule: 2 weeks on unchanged dosage, 2 weeks on halved benzodiazepine dosage, 2 weeks with no benzodiazepines (followed by 2 weeks with placebo in both groups and 2 weeks with no study medication)

  1. Benzodiazepine taper schedule + buspirone 10 to 30 mg/d(N = 13)

  2. Benzodiazepine taper schedule + placebo(N = 11)

Outcomes

  • Benzodiazepine withdrawal symptoms, Tranquilizer Withdrawal Rating Scale

  • Insomnia: Sleep rating scale

  • Anxiety: HAM‐A

  • Benzodiazepine cessation

Identification

Sponsorship source: Not mentioned

Country: UK

Setting: Outpatients

Declarations of interest: Not mentioned

Authors name: Malcolm Lader

Institution: Institute of Psychiatry, University of London

Email:

Address: Institute of Psychiatry, University of London, De Crespigny Park, Denmark Hill, London, SE5 8AF England

Notes

For reasons that are unclear, results are reported at week 3, i.e. after the first week of benzodiazepine reduction to half. That is, results are not available for week 6, when benzodiazepines have been tapered off. Figure 2 shows the temporal pattern for Hamilton Anxiety Scale (i.e. all time points available graphically) but only for the successful completers (5 buspirone, 6 placebo).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "During the first two weeks of withdrawal (3 and 4), buspirone or placebo was substituted for the benzodiazepine in an initial dosage of 5 mg (one capsule) twice daily, followed by 10 mg (two capsules) twice daily...The study was conducted double‐blind in that neither investigator nor patient knew whether placebo or buspirone was being administered during weeks 2 to 5"

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Comment: It is stated that "investigators" were blinded. Judged as done.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: Successful completers: no attrition bias

Selective reporting (reporting bias)

Unclear risk

Comment: No data on benzodiazepine dosage in the 2 groups, but the trial was designed to stop benzodiazepine use, and therefore dose reduction was not considered. However, the choice of using 3 weeks as primary time point does not seem justified.

Other bias

Low risk

Comment: No other apparent source of bias

Lader 1993

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 8 weeks

Single‐centre

Participants

Baseline characteristics

Alpidem (a Z‐drug)

  • Male, N (%): 4 (31)

Placebo

  • Male, N (%): 4 (33)

Inclusion criteria: Benzodiazepine use for more than 6 months, less than 30 mg/d diazepam equivalents, regarded as dependent (problems on previous attempts to lower the dosage), 18 to 65 years of age, within 20% of normal body weight

Exclusion criteria: Major physical or psychiatric illness, drug abusers, women of child‐bearing age unless on adequate contraception

Pretreatment: Not described

Interventions

Intervention characteristics

Benzodiazepine taper schedule: 2 weeks on unchanged dosage, 2 weeks on halved benzodiazepine dosage, 2 weeks with no benzodiazepines (followed by 2 weeks with halved dosage study medication and 2 weeks with no study medication)

  1. Benzodiazepine taper schedule + alpidem (a Z‐drug) 100 to 150 mg/d(N = 13)

  2. Benzodiazepine taper schedule + placebo(N = 12)

Outcomes

  • Benzodiazepine cessation

  • Anxiety, HAM‐A

  • Benzodiazepine withdrawal symptoms, Tranquilizer Withdrawal Rating Scale

Identification

Sponsorship source: Not described

Country: UK

Setting: Outpatients

Declarations of interest: Not mentioned

Author's name: Lader M

Institution: Institute of Psychiatry

Email:

Address: Institute of Psychiatry, De Crespigny Park, Denmark Hill, London SES 8AF, UK

Notes

Anxiety and benzodiazepine withdrawal symptoms: the results are only shown in graphic as mean values, SDs not reported. SDs for HAM‐A were therefore imputed from Cassano 1996, which is a similar trial also using alpidem to facilitate benzodiazepine withdrawal. It was not possible to impute SDs for benzodiazepine withdrawal symptoms because withdrawal symptoms in Cassano 1996 were reported as a dichotomised variable, whereas they were reported as a continuous variable in Lader 1993.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "The study was conducted double‐blind in that neither investigators nor patients knew whether placebo or alpidem was being administered during weeks 3‐8"

Comment: Study described as double‐blinded and using placebo.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Comment: It is stated that "investigators" were blinded. Judged as done.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: High dropout, however only completion could be extracted from the study, and this is not biased by the high dropout rate.

Selective reporting (reporting bias)

Low risk

Comment: No indications of selective reporting

Other bias

Low risk

Comment: No other apparent bias

Lecrubier 2005

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 16 weeks

Multicentre

Participants

Baseline characteristics

Lithium

  • Male, N (%): 46 (32)

  • Age, mean (SD): 49.3 (10.3)

  • Benzodiazepine dose (diazepam equivalent), mean (SD): 15.7 (7.0)

Placebo

  • Male, N (%): 30 (31)

  • Age, mean (SD): 47.6 (11.2)

  • Benzodiazepine dose (diazepam equivalent), mean (SD): 13.5 (5.2)

Inclusion criteria: Outpatients, 18 to 65 years old, receiving benzodiazepines for at least 6 months at a daily dose ranging from 10 to 40 mg diazepam or equivalent and wishing to withdraw benzodiazepine treatment

Exclusion criteria: Anxiety disorder with a score of 15 or above on the HAM‐A, major depressive disorder, social phobia, alcohol or substance abuse according to Mini International Neuropsychiatric Interview, and/or other serious pathology. Tranquilisers including antihistamines, hypnotics, anxiolytics, and lithium salts were not allowed.

Pretreatment: No significant pretreatment group differences

Interventions

Benzodiazepine taper schedule: 4 weeks stable benzodiazepine and lithium versus placebo, 4 weeks benzodiazepine withdrawal ‐ reduction with 50% every week, 8 weeks lithium maintenance

  1. Benzodiazepine taper schedule + lithium 0.84 mg/day(N = 146)

  2. Benzodiazepine taper schedule + placebo(N = 98)

Outcomes

  • Benzodiazepine cessation

  • Serious AEs

  • Non‐serious AEs

  • Discontinuation due to AEs

Identification

Sponsorship source: Not described

Country: France

Setting: Outpatients

Declarations of interest: Not mentioned

Author's name: Lecrubier

Institution: Inserm unité 302, service de psychiatrie AD

Email: [email protected]

Address: Hôpital Pitié‐Salpêtrière, 17, boulevard de l’hôpital, 75013 Paris, France

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "double‐blind, randomised study"

Comment: Not sufficiently described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Lithium gluconate and placebo were dispensed in vials and were indistinguishable in terms of appearance, taste and smell"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: Not sufficiently described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: 244 participants were randomised: 146 to lithium and 98 to placebo. Only participants entering the benzodiazepine tapering phase were analysed (136 participants allocated to lithium and 94 to placebo), thus attrition rate of 7% and 4%, respectively.

Selective reporting (reporting bias)

Unclear risk

Comment: Benzodiazepine dose at endpoint not reported, only participants who succeeded in discontinuing benzodiazepine usage.

Other bias

Low risk

Comment: No other apparent source of bias

Lemoine 2006

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 4 weeks

Multicentre

Participants

Baseline characteristics

Bromazepam

  • Male, N (%): 20 (24)

  • Age, mean (SD): 48.2 (11.1)

  • Years of benzodiazepine use, mean (SD): 4.0 (5.6)

  • Months of benzodiazepine use, mean (SD): 47.4 (67.7)

Cyamemazine

  • Male, N (%): 30 (39)

  • Age, mean (SD): 48.7 (10.2)

  • Years of benzodiazepine use, mean (SD): 4.5 (1.8)

  • Months of benzodiazepine use, mean (SD): 52.5 (21.4)

Inclusion criteria: Participants were aged 18 to 65 years, treated for anxiety for at least 3 months with benzodiazepines (bromazepam, lorazepam, alprazolam, or oxazepam) at a daily dose of 5 to 20 mg diazepam‐equivalent, and requiring a withdrawal. A < 18 score in the Hamilton Anxiety Rating Scale was required.

Exclusion criteria: Female patients were excluded if they were pregnant or likely to become so or if they were breastfeeding. Individuals incapable of completing a questionnaire or of properly giving informed consent were also excluded. In addition, current treatment with any psychotropic drug or any other central nervous system active medication was forbidden. The presence of comorbid depression was also an exclusion criterion.

Pretreatment: NS

Interventions

Intervention characteristics

  1. Abrupt benzodiazepine cessation + bromazepam 3 to 6 mg/d(N = 83)

  2. Abrupt benzodiazepine cessation + cyamemazine 25 to 50 mg/d(N = 77)

Outcomes

  • Anxiety: maximum amplitude of rebound (HAM‐A)

  • Non‐serious adverse events

  • Relapse to benzodiazepine use

  • Discontinuation due to adverse events

Identification

Sponsorship source: Not described. 1 of the authors affiliated with Sanofi‐Aventis.

Country: France

Setting: Outpatients

Declarations of interest: Not mentioned

Author's name: Patrick Lemoine

Institution: Unite´ Clinique de Psychiatrie Biologique, Bron

Email: [email protected]

Address: 46bis rue Gallie´ni, 91360 Villemoisson‐sur‐Orge, France

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Both drugs were administered in identical soft gelatin capsules"

Comment: Sufficient blinding

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: ITT analysis performed.

Selective reporting (reporting bias)

High risk

Comment: Protocol not available, unusual primary outcome (maximum amplitude of anxiety rebound).

Other bias

High risk

Comment: Role of Sanofi‐Aventis not described.

Mariani 2016

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 8 weeks

Single‐centre

Participants

Baseline characteristics

Gabapentin

  • Age, mean (SD): 40

  • Male, N (%): 6 (75)

Placebo

  • Age, mean (SD): 37

  • Male, N (%): 8 (73)

Inclusion criteria: Meeting DSM‐IV criteria for current benzodiazepine abuse or dependence and opioid dependence, and being treated for opioid dependence with methadone, 18 to 65 years of age

Exclusion criteria: (1) Any Axis I psychiatric disorder as defined by DSM‐IV‐TR that was unstable or would be disrupted by study medication or by an effort to discontinue benzodiazepines; (2) Acute physiological withdrawal or a history of seizures during alcohol or sedative‐hypnotic withdrawal; (3) Individuals with cocaine dependence as their primary substance use disorder diagnosis; (4) Individuals with unstable physical disorders or impaired kidney function; (5) Prescribed psychotropic medications other than methadone or medications prescribed for pain syndromes that would be disrupted by study medication or by an effort to discontinue benzodiazepines; (6) Anticonvulsants prescribed for pain syndromes; (7) Known sensitivity to gabapentin; (8) Individuals who had exhibited suicidal or homicidal behaviour within the past 2 years or had current active suicidal ideation; (9) Individuals physiologically dependent on any other drugs (excluding nicotine, caffeine, methadone); (10) Individuals currently prescribed gabapentin; and (11) Individuals requiring pharmacological detoxification from benzodiazepines in the past year and are unlikely to be able to tolerate taper off of benzodiazepines

Pretreament differences: None reported.

Interventions

Intervention characteristics

  1. Abrupt benzodiazepine cessation + gabapentin 1200 mg 3 times daily(N = 8)

  2. Abrupt benzodiazepine cessation (control group)(N = 11)

Outcomes

Benzodiazepine mean dose

Identification

Sponsorship source: Funding for this work was provided by National Institute on Drug Abuse grants K23‐ DA021209 (Mariani), P50‐DA09236 (Kleber), K24‐ DA022412 (Nunes), and K24 029647 (Levin).

Country: USA

Setting: Methadone maintenance outpatients

Declarations of interest: None

Authors name: John J Mariani

Institution: Division on Substance Abuse, New York State Psychiatric Institute, New York, NY, USA

Email: [email protected]

Address: New York State Psychiatric Institute, Division on Substance Abuse, 1051 Riverside Drive, Unit 66, New York, NY 10032, USA

Notes

Data not reported sufficiently, not possible to extract results relevant to this review. The author has not responded to our queries.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Randomisation method not described.

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Comment: All capsules were over‐capsulated with riboflavin to ensure compliance.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

Comment: Only 50% were retained in the study.

Selective reporting (reporting bias)

Low risk

Comment: Selective outcome reporting not evident.

Other bias

Low risk

Comment: No other apparent source of bias

Mercier‐Guyon 2004

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 6 weeks (2 weeks taper off, 4 weeks assessment, ends day 45)

Multicentre

Participants

Baseline characteristics

Captodiame

  • Age, mean (SD): 39.1 (1.3)

  • Male, N (%): 20 (50)

Placebo

  • Age, mean (SD): 41.9 (1.4)

  • Male, N (%): 20 (48.8)

Inclusion criteria: Participants aged 25 to 55 years who had been prescribed certain benzodiazepines (lorazepam, bromazepam, alprazolam, oxazepam, or clobazam) in the official recommended dose range for the treatment of an anxiety disorder for at least 6 months, stable benzodiazepine dosage over the 6‐month period. Since alertness was assessed with a driving simulation test, included participants were required to be in possession of a valid driving license for at least 5 years.

Exclusion criteria: People with a history of alcohol dependence in the previous 5 years were excluded, as were those consuming excessive quantities of alcohol as defined in the CAGE questionnaire. Proven consumption (either openly declared or detected by urine testing) of illicit psychotropic drugs (opiates, cocaine, cannabis, amphetamines) or of any other sedatives also constituted grounds for exclusion. Additionally, people with severe, unstable, or uncontrolled hepatic, renal, or cardiac insufficiency, with glaucoma or prostate hypertrophy, or with any psychiatric disease other than generalised anxiety disorders were also excluded. Female individuals who were pregnant or breastfeeding were excluded.

Pretreatment: No significant group differences

Interventions

Intervention characteristics

Benzodiazepine taper schedule: half dose first week of experimental treatment, a quarter dose the second week, then discontinuation on day 14

  1. Benzodiazepine taper schedule + captodiame 150 mg/d(N = 40)

  2. Benzodiazepine taper schedule + placebo(N = 41)

Outcomes

  • Benzodiazepine withdrawal symptoms, BWSQ

  • Non‐serious adverse events

  • Anxiety, Hamilton Anxiety Rating Scale

  • Serious adverse events

Identification

Sponsorship source: This study was funded by Laboratoires Bailly‐Creat, Paris, France, manufacturers of captodiame (Covatine), who financed the honoraria of the participating physicians and the statistical analysis.

Country: France

Setting: Outpatients

Declarations of interest: Not mentioned

Comments: Benzodiazepine dose during and after discontinuation not recorded/documented.

Authors name: Merzecier‐Guyon C

Institution: Centre d’Etudes et de Recherches en Médecine du Trafic, Annecy, France

Email: [email protected]

Address: Dr C Mercier‐Guyon, CERMT, BP 132, 74004 ANNECY Cedex, France

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not sufficiently described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Quote: "randomised, double‐blind, placebo‐controlled trial"

Comment: What has been done to ensure blinding of participants and study personnel is not described.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: All randomised participants analysed.

Selective reporting (reporting bias)

Unclear risk

Quote: "we have little information available on real benzodiazepine use during the discontinuation and follow‐up phases, which is the most relevant measure of successful benzodiazepine"

Comment: No information due to study design, not interpreted as being left out intentionally

Other bias

High risk

Quote: "This study was funded by Laboratoires Bailly‐Creat, Paris, France, manufacturers of captodiamine (Covatine), who financed the honoraria of the participating physicians and the statistical analysis."

Comment: No indication of sponsor's influence on study analysis, etc.; interpreted as high risk of bias

Morton 1995

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 16 weeks

Single‐centre

Participants

Baseline characteristics

Not described

Inclusion criteria: Referred to Benzodiazepine Withdrawal Clinic, benzodiazepines had been taken long term (> 6 months) at normal dose (< 30 mg/day of diazepam or equivalent), 18 to 70 years of age, body weight within normal limits

Exclusion criteria: Major physical or psychiatric illnesses, drug abuse, women of childbearing age unless taking effective contraceptive measures

Pretreatment: Not reported

Interventions

Intervention characteristics

Benzodiazepine taper schedule: 4 weeks buspirone/placebo stabilisation, 6 weeks tapering to zero, 4 weeks of benzodiazepine abstinence, buspirone/placebo halved in dosage and then stopped 2 weeks later

  1. Benzodiazepine taper schedule + buspirone flexible dosing, min 15 mg/d, mean 25 mg/day (N = 12)

  2. Benzodiazepine taper schedule + placebo(N = 12)

Outcomes

  • Benzodiazepine cessation

  • Anxiety, HAM‐A

  • Benzodiazepine withdrawal symptoms, benzodiazepine withdrawal profile

  • Non‐serious adverse events

Identification

Sponsorship source: This study was supported by a grant from Bristol‐Myers Squibb UK to the Institute of Psychiatry.

Country: UK

Setting: Outpatients

Declarations of interest: Not mentioned

Author's name: Morton S

Institution: Institute of Psychiatry, University of London

Email:

Address: Institute of Psychiatry, De Crespigny Park, London SE5 8AFUK

Notes

Means only given in figures (HAM‐A and benzodiazepine withdrawal symptoms), no SDs reported, not possible to impute in a methodologically valid way.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Half the patients were given buspirone in flexible dosage according to the usual criteria of clinical need, at a minimum of 15 mg/day in divided doses. The others received matching placebo in equivalent flexible dosage, again according to apparent clinical need...The study was conducted double‐blind with reference to whether buspirone or placebo was being administered in weeks 2‐18"

Comment: Done

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: No risk of bias regarding the main outcome (benzodiazepine cessation), but the secondary outcomes were only analysed for the participants who had discontinued treatment, i.e. half of the participants.

Selective reporting (reporting bias)

Low risk

Comment: No reason to suspect selective outcome reporting

Other bias

High risk

Comment: Financed by a grant from Bristol‐Myers Squibb, but further information on potential influence on design, etc. not provided.

Nakao 2006

Methods

Study design: Randomised controlled trial

Study grouping: Parallel

Blinding: None, open‐label

Duration: 8 weeks

Single‐centre

Participants

Baseline characteristics: Not reported

Inclusion criteria: The participant selection criteria were as follows: (i) those aged 20 to 70 years; (ii) those whose medical condition was stable and drug regimens unchanged for longer than 3 months; (iii) those who had been prescribed either alprazolam, bromazepam, etizolam, or lorazepam for at least 3 months prior to visiting the clinic; and (iv) those who were able to visit the clinic for an 8‐week intervention (or control) period.

Exclusion criteria: DSM‐IV major depression

Pretreatment: No baseline characteristics provided.

Interventions

Intervention characteristics

Benzodiazepine taper schedule: 8‐week gradual benzodiazepine discontinuation

  1. Benzodiazepine taper schedule + paroxetine 10 to 20 mg(N = 22)

  2. Benzodiazepine taper schedule (control group, no placebo)(N = 23)

Outcomes

  • Anxiety, HAM‐A

  • Benzodiazepine withdrawal symptoms, Benzodiazepine Withdrawal Symptom Questionnaire

Identification

Sponsorship source: Not described

Country: Japan

Setting: Outpatients

Declarations of interest: Not mentioned

Author's name: Nakao M

Institution: Division of Psychosomatic Medicine, Teikyo University Hospital

Email: [email protected]‐u.ac.jp

Address: Department of Hygiene and Public Health, Teikyo University School of Medicine, 2‐11‐1 Kaga, Itabashi‐ku, Tokyo, Japan

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Comment: No placebo: open‐label trial

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Comment: Not done

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: All randomised participants analysed.

Selective reporting (reporting bias)

Low risk

Comment: No protocol provided, but all relevant outcomes seem to be reported.

Other bias

Unclear risk

Comment: No other apparent biases, the funding of the study not described.

Pat‐Horenczyk 1998

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 4 weeks

Single‐centre

Participants

Baseline characteristics

Zopiclone

  • Age, mean (SD): 52.7 (6.05)

Flunitrazepam

  • Age, mean (SD): 49 (10.6)

Inclusion criteria: Long‐term usage of benzodiazepine hypnotics (range 6 months to 22 years), use of flunitrazepam for at least 3 months with stabilisation at a nightly dosage of 1 mg for at least 1 month before inclusion

Exclusion criteria: Other benzodiazepine consumption, use of psychotropic medications

Pretreatment: No significant group differences

Interventions

Intervention characteristics

Benzodiazepine taper schedule: during the first part of the study, participants were either switched gradually to 1) zopiclone (3.75 mg and then 7.5 mg) over a 2‐week period (N = 7), or 2) continued their usual treatment of flunitrazepam 1 mg (N = 11).

In the second part of the trial, the hypnotic (either zopiclone or flunitrazepam) was gradually withdrawn according to a 2‐step scheme over 2 weeks.

Outcomes

  • Benzodiazepine withdrawal symptoms, Ashton Withdrawal Symptom Checklist, the Benzodiazepine Withdrawal Scale, the Benzodiazepine Withdrawal Symptom Questionnaire

  • Relapse to benzodiazepine use

  • Insomnia: total sleep time (and a range of other polysomnographic measures)

Identification

Sponsorship source: This study was supported by Rhone‐Poulenc Rorer Ltd, France, and the Technion Sleep Medicine Center, Israel

Country: Israel

Setting: Outpatients

Declarations of interest: Not mentioned

Authors name: Ruth Pat‐Horenczyk

Institution: Technion Sleep Laboratory, Faculty of Medicine

Email:

Address: Technion Sleep Laboratory, Faculty of Medicine, Gutwirth Building, Technion‐Israel Institute of Technology, Haifa 32000, Israel

Notes

Data from the benzodiazepine withdrawal questionnaires were not reported, thus only data on benzodiazepine relapse and insomnia could be extracted from this trial.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "double‐blind randomised", "ZOP and FLU were encapsulated, and dummy placebo capsules were given to the patients who did not switch to ZOP, so that during the 5‐week period, all patients consumed", "two identical‐looking pills each night."

Comment: Double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: Not sufficiently described

Incomplete outcome data (attrition bias)
All outcomes

High risk

Comment: 20/24 participants completed the 5‐week withdrawal programme, however analysis was performed on women only due to uneven gender distribution between groups and high dropout rate among men.

All 5 male participants had been randomised to the zopiclone group, 3 of whom dropped out, and it was decided to perform the analyses on women only (n = 18).

Selective reporting (reporting bias)

Low risk

Comment: Unlikely

Other bias

High risk

Comment: Role of funding source not explicitly described.

Peles 2007

Methods

Study design: Randomised controlled trial

Study grouping: Cross‐over

Blinding: Double

Duration: 6 weeks

Single‐centre

Participants

Baseline characteristics

Not reported for each group because of cross‐over design.

Most (70%) of the 80 study participants were male. The mean age during study was 42.6 years, and the mean duration in MMT was 4.4 years. Almost half (48.8%) of the participants had other drug abuse in addition to benzodiazepines in the month prior to study entry. Specifically, 25 had positive urine for opiates, 12 for cocaine, 14 for cannabis, and 5 for amphetamines. With respect to lifetime psychiatric diagnosis, 9 participants (11.3%) had 1 of the psychotic disorders, 18 (22.5%) had an affective disorder, 8 (10%) had an adjustment disorder, 2 (2.5%) had an organic brain disorder, 38 (47.5%) had no DSM‐IV Axis I diagnosis (but all 38 had a DSM‐IV Axis II personality disorder), and 5 (6.3%) had no DSM‐IV Axis I or Axis II psychiatric diagnosis.

Inclusion criteria: All patients who were admitted to the MMT clinic between July 1993 and July 2004 were eligible for inclusion in the study. This MMT clinic receives patients who meet DSM‐IV criteria for opioid dependence and report self administration of illicit heroin for 1 year or more.

Exclusion criteria: None

Pretreatment: No significant group differences

Interventions

Intervention characteristics

Benzodiazepine taper schedule: run‐in phase: taper, until reaching 6 mg/day clonazepam or equivalent. Week 1 through 6: 0.5 mg/week dose reduction

  1. Benzodiazepine taper schedule + melatonin 5 mg/d (N = 40)

  2. Benzodiazepine taper schedule + placebo (N = 40)

Outcomes

  • Benzodiazepine cessation

  • Serious adverse events

  • Insomnia, PSQI

  • Relapse to benzodiazepine use

Identification

Sponsorship source: The study was supported (in part) by a grant from The Israel Anti Drug Authority.

Country: Israel

Setting: Outpatients in methadone maintenance treatment

Declarations of interest: Not mentioned

Author's name: Einat Peles

Institution: Adelson Clinic, Tel Aviv Sourasky Medical Center

Email: [email protected]

Address: Adelson Clinic, Tel Aviv Sourasky Medical Center, 1 Henrietta Szold Street, Tel Aviv 64924, Israel

Notes

Only data from the first period (first 6 weeks) of this cross‐over trial was included.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not sufficiently described

Allocation concealment (selection bias)

Low risk

Quote: "consecutive container numbers"

Comment: Done

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Quote: "codes for melatonin first/placebo first were known only to the pharmacist"

Comment: What has been done to ensure blinding of participants and study personnel is not described.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "The codes for melatonin first/placebo first were known only to the pharmacist who prepared the sequence in a random manner and identified it to us only at the end of the study"

Comment: Only pharmacist knew the code ‐ done.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Quote: "40 patients who started on melatonin and 40 patients who started on placebo. Sixty‐one patients (31 from the ‘melatonin‐first’ group and 30 from the ‘placebo‐first’ group) completed phase one (6 weeks). Forty‐four patients completed all 13 weeks of the study, with no differences between groups (60% of the 40 ‘melatonin‐first’ group and 50% of the 40 ‘placebo‐first’ group (P = 0.5)."

Comment: Unclear how the high dropout after 6 weeks affected the results

Selective reporting (reporting bias)

Unclear risk

Comment: The division of benzodiazepine continuers/discontinuers in the analysis seems to blur the effect of the study medication in itself.

Other bias

Low risk

Comment: No other apparent source of bias

Rickels 1999

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 13 weeks

Single‐centre

Participants

Baseline characteristics

No group difference. Only combined baseline characteristics reported:

  • Age, mean (SD): 47 (12)

  • Male, N (%): 38 (49)

  • Employed: 51%

  • Duration of benzodiazepine treatment, months: 83 (75)

  • Benzodiazepine daily dose, mg diazepam equivalents: 19 (16.7)

Inclusion criteria: Age range of 21 to 70 years, had to have been on continuous daily treatment with diazepam, lorazepam, or alprazolam for a minimum of 1 year, and needed to be able to provide written informed consent

Exclusion criteria: A screening medical history, physical examination, ECG, blood count, blood chemistry, urine analysis, and urine drug screens were performed to confirm each patient’s study eligibility.

Pretreatment: No significant group differences

Interventions

Intervention characteristics

Benzodiazepine taper schedule: gradual taper was initiated at approximately 25% reduction per week, with participants on lower therapeutic benzodiazepine doses possibly being tapered slightly faster, and participants on higher therapeutic doses being tapered slightly slower, but not longer than 6 weeks. After the taper was completed, participants were seen weekly for at least 5 weeks in order to determine their ability to stay off their benzodiazepine. During that time, participants continued to receive their double‐blind study medication. Study medication was discontinued at 5 weeks' post‐taper completion. From 5 to 12 weeks post‐taper, participants left the program and returned to their private physician for doctor’s choice management.

  1. Benzodiazepine taper schedule + valproate 500 to 2500 mg/day(N = 19)

  2. Benzodiazepine taper schedule + trazodone 100 to 500 mg/day(N = 41)

  3. Benzodiazepine taper schedule + placebo(N = 18)

Outcomes

  • Benzodiazepine withdrawal symptoms: Physician Withdrawal Checklist

  • Benzodiazepine cessation

  • Anxiety: HAM‐A

  • Non‐serious adverse events

  • Relapse to benzodiazepine use

  • Discontinuation due to adverse events

Identification

Sponsorship source: This study was supported by USPHS Research Grant MHO8957.

Country: USA

Setting: Outpatients

Declarations of interest: The study was supported by a US Public Health Service research grant

Author's name: Rickels K

Institution: Mood and Anxiety Disorders Section, Department of Psychiatry, University of Pennsylvania

Email:

Address: University Science Center, 3600 Market Street, Suite 803, Philadelphia, PA 19104‐2649, USA

Notes

We selected only the valproate vs placebo comparison for this meta‐analysis because we did not consider it relevant to combine the experimental intervention groups into a single group (cf. Cochrane Handbook 16.5.4 on how to include multiple groups from 1 study).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Quote: "randomly assigned under double blind conditions to study drug or placebo."

Comment: What has been done to ensure blinding of participants and study personnel is not described.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: Not sufficiently described

Incomplete outcome data (attrition bias)
All outcomes

High risk

Comment: 15 participants, 5 trazodone (12.2%), 5 valproate (26.3%), and 5 placebo (27.7%), dropped out during the pretreatment phase. The 15 dropouts were compared on a variety of demographic and illness variables with the 63 participants who entered taper, and no significant differences were present.

Selective reporting (reporting bias)

Low risk

Comment: No apparent selective outcome reporting

Other bias

Unclear risk

Comment: Groups were not of equal size. No argument is provided.

Rickels 2000

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 11 to 13 weeks

Single‐centre

Participants

Baseline characteristics

No significant differences between the groups; only data for the combined participant group reported:

  • Male, N (%): 59 (55)

  • Age, mean (SD): 48 (14)

  • Months of benzodiazepine use, mean (SD): 102 (92)

Inclusion criteria: Participants were required to have a diagnosis of generalised anxiety disorder according to DSM‐III‐R, to be at least 21 years old, and to have been taking diazepam, lorazepam, or alprazolam in therapeutic doses continuously for the past 12 months.

Exclusion criteria: Panic disorder diagnosis

Pretreatment: No significant group differences

Interventions

Intervention characteristics

Benzodiazepine taper schedule: 4‐week stabilisation phase, 4‐ to 6‐week taper phase: 25% reduction per week, 5‐week benzodiazepine‐free phase, the experimental drug continued for the first 3 weeks of the benzodiazepine‐free phase

  1. Benzodiazepine taper schedule + imipramine 180 mg/d(N = 23)

  2. Benzodiazepine taper schedule + buspirone 38 mg/d(N = 28)

  3. Benzodiazepine taper schedule + placebo(N = 24)

Outcomes

  • Benzodiazepine cessation

  • Non‐serious adverse events

  • Discontinuation due to adverse events

  • Serious adverse events

Identification

Sponsorship source: Supported by NIMH grant MH‐08957. Dr Greenblatt was supported by NIMH grant MH‐34223 and grant DA‐05258 from the National Institute on Drug Abuse. The medications used were provided by Bristol‐Myers Squibb CNS Group, Wallingford, CT.

Country: USA

Setting: Outpatients

Declarations of interest: Not mentioned

Authors name: Karl Rickels

Institution: Mood and Anxiety Disorders Section, Department of Psychiatry, University of Pennsylvania, Philadelphia

Email: [email protected]

Address: University Science Center, 3600 Market St., Suite 803, Philadelphia, PA 19104

Notes

Adverse events not reported appropriately for a meta‐analysis.

We selected only the imipramine versus placebo comparison for this meta‐analysis because we did not consider it relevant to combine the experimental intervention groups into a single group (cf. Cochrane Handbook 16.5.4 on how to include multiple groups from 1 study).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Medication was prepared double blind in identical capsules containing either 5 mg buspirone, 25 mg imipramine, or placebo"

Comment: Sufficiently done

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Comment: Not clearly described

Selective reporting (reporting bias)

Low risk

Comment: Probably not, relevant outcome measures

Other bias

Low risk

Comment: No other sources of bias evident.

Romach 1998

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 6 weeks

Single‐centre

Participants

Baseline characteristics

Ondansetron

  • Male, N (%): 29 (62)

  • Age, mean (SD): 46 (13)

  • Months of benzodiazepine use: 62 (53)

  • Benzodiazepine dose (diazepam equivalents), mean (SD): 16 (13)

Placebo

  • Male, N (%): 29 (58)

  • Age, mean (SD): 48 (13)

  • Months of benzodiazepine use: 74 (67)

  • Benzodiazepine dose (diazepam equivalents), mean (SD): 9 (7)

Inclusion criteria: DSM‐III‐R criteria for benzodiazepine dependence, a desire to discontinue use of benzodiazepines, daily use of alprazolam or lorazepam for > 3 months

Exclusion criteria: Dosage of lorazepam > 8 mg/day or alprazolam > 5 mg/day, psychoactive substance use disorder (other than benzodiazepines), using other prescribed psychotropic medications (other benzodiazepine, antidepressants, antipsychotics, or anticonvulsants), psychosis, moderate to severe major depression, significant cognitive impairment, or suicidal ideation. Serious medical illness, pregnancy, liver enzymes elevated more than 3 times the upper limit, past history of head trauma

Pretreatment: More anxiety patients in the placebo group

Interventions

Intervention characteristics

Benzodiazepine taper schedule: participants set their benzodiazepine tapering goals weekly with a study team member; the overall goal was benzodiazepine discontinuation within the treatment period (6 weeks).

  1. Benzodiazepine taper schedule + ondansetron 4 mg/d(N = 54)

  2. Benzodiazepine taper schedule + placebo(N = 54)

Outcomes

  • Benzodiazepine withdrawal symptoms, Clinical Institute Withdrawal Assessment Scale ‐ Benzodiazepines

  • Benzodiazepine mean dose

  • Anxiety, Symptom Checklist‐90 anxiety subscale

  • Non‐serious adverse events

  • Discontinuation due to adverse events

Identification

Sponsorship source: The study was supported in part by Glaxo Wellcome Canada.

Country: Canada

Setting: Outpatients

Declarations of interest: Not mentioned

Author's name: Myroslav Romach

Institution: Departments of Pharmacology, Medicine, and Psychiatry and Faculty of Pharmacy, University of Toronto and Addiction Research Foundation, Toronto, Ontario, Canada

Email:

Address: Women's College Hospital, Department of Psychiatry, 76 Grenville St., Toronto, Ontario, Canada M5S 1B2

Notes

Not possible to extract results from this study due to poor reporting

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Comment: Described as double‐blind and identical‐appearing capsules

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: Not specifically described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Comment: Out of 108 participants, only 11 (10%) participants dropped out. However, it is unclear if dropout was balanced between groups.

Selective reporting (reporting bias)

Low risk

Comment: No protocol available, but no reason to suspect selective outcome reporting.

Other bias

High risk

Comment: Role of medicinal company as funding source not sufficiently described.

Rynn 2003

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 13 weeks

Single‐centre

Participants

Baseline characteristics

No significant differences between groups; only data for the combined participant group reported:

  • Months of benzodiazepine use, mean (SD): 75 (64)

  • Male, N (%): 21 (52)

Inclusion criteria: A diagnosis of panic disorder according to the Diagnostic and Statistical Manual of Mental Disorders, Revised Third Edition, at least 21 years old, and have been taking diazepam, lorazepam, or alprazolam in therapeutic doses continuously for at least the past 12 months (5 mg diazepam was considered equivalent to 1 mg lorazepam and 0.5 mg alprazolam). Individuals must also have expressed a desire to stop benzodiazepine intake.

Exclusion criteria: Other psychotropic medication than benzodiazepines

Pretreatment: No significant group differences

Interventions

Intervention characteristics

Benzodiazepine taper schedule: after being kept on a stable benzodiazepine dose for 2 to 4 weeks within the therapeutic range during the screening period, participants were assigned to double‐blind treatment during which time their benzodiazepine intake was not altered. This pretreatment lasted 4 weeks, after which participants were tapered from their benzodiazepine dose over a 6‐week period. Benzodiazepine intake was reduced at the rate of 25% per week. Taper was followed by a 5‐week benzodiazepine‐free phase designed to prospectively assess the participant's clinical status in the initial period while being off benzodiazepines. Double‐blind study treatment was continued for the first 3 weeks of this phase. For the last 2 weeks, placebo was substituted for imipramine and buspirone.

  1. Benzodiazepine taper schedule + imipramine 180 mg/d(N = 18)

  2. Benzodiazepine taper schedule + buspirone 32 mg/d(N = 12)

  3. Benzodiazepine taper schedule + placebo(N = 10)

Outcomes

  • Benzodiazepine withdrawal symptoms, Physician Withdrawal Checklist, Covi Withdrawal Cluster of the Hopkins Symptom Checklist

  • Benzodiazepine cessation

  • Anxiety: HAM‐A (change from baseline)

  • Non‐serious adverse event

  • Discontinuation due to adverse events

Identification

Sponsorship source: This research was supported by NIMH grant MH‐08957. Bristol‐Myers Squibb CNS Group (Wallingford, CT) provided all double‐blinded medications.

Country: USA

Setting: Outpatients

Declarations of interest: Not mentioned

Authors name: Moira Rynn

Institution: Mood and Anxiety Disorders Section, Department of Psychiatry, University of Pennsylvania, Suite 670, 3535 Market Street, Philadelphia, PA 19104‐3309

Email: [email protected]

Address: Mood and Anxiety Disorders Section, Department of Psychiatry, University of Pennsylvania, Suite 670, 3535 Market Street, Philadelphia, PA 19104‐3309

Notes

Adverse events not appropriately described.
We included only the imipramine‐placebo comparison in the meta‐analysis because we did not consider it relevant to combine the experimental intervention groups into a single group (cf. Cochrane Handbook 16.5.4 on how to include multiple groups from 1 study).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Medication was prepared double blind in identical capsules"

Comment: Done

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: Double‐blind identical capsules ‐ doneNot described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: "Of 52 patients randomised to 3 treatment conditions, 12 patients did not complete the pretreatment phase and 40 patients entered taper. The 2 patient groups did not differ in any baseline demographic or clinical measures with 1 exception. Dropouts had lower BZ doses at baseline, ex‐ pressed in diazepam equivalents, than patients entering taper (12.1 ± 7.7 versus 25.7 ± 19.5; F = 5.52; df = 1.50; P &lt; 0.02). Dropouts also did not differ from taper patients in treatment assignment ( 2 = 0.69; df = 2; P = NS) or type of BZ at baseline ( 2 = 1.43; df = 2; P = NS). The main reason for dropping out of the program during the pre taper phase were adverse events (2 buspirone, 2 imipramine, and 1 placebo)."

Comment: Acceptable and no difference between groups

Selective reporting (reporting bias)

Low risk

Comment: Protocol not available but no obvious selective outcome reporting

Other bias

Low risk

Comment: Role of BMS only to provide double‐blinded study medication

Saul 1989

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 18 weeks

Multicentre

Participants

Baseline characteristics

Atenolol

  • Male, N (%): 19 (31)

  • Age, mean: 43.55

Placebo

  • Male, N (%): 22 (37)

  • Age, mean: 44.35

Inclusion criteria: 18 to 60 years old, daily use of benzodiazepines for at least 8 weeks, not more than 15 mg of diazepam

Exclusion criteria: Cerebrovascular or generalised vascular disease, heart block, thyrotoxicosis, premenstrual tension or other trigger of cyclical anxiety and depression, pregnancy, antihypertensive therapy or any drug likely to affect anxiety, and those for whom diazepam would be an unsuitable rescue

Pretreatment: None reported.

Interventions

Intervention characteristics

Benzodiazepine taper schedule: follow‐up visits at 4‐week intervals, participants should have stopped taking benzodiazepines by their 4th visit

  1. Benzodiazepine taper schedule + atenolol 50 mg/d(N = 62)

  2. Benzodiazepine taper schedule + placebo(N = 59)

Outcomes

  • Benzodiazepine consumption

  • Anxiety

  • Withdrawal symptoms

Identification

Sponsorship source: Not described

Country: UK

Setting: Outpatients

Declarations of interest: Not mentioned

Authors name: Saul PA

Institution: General Practitioners, Stuart Clinical Research Group

Email:

Address: P. A. Saul, 555 Chorley Old Road, Bolton, Lancashire, BL2 6AF, UK

Notes

None of the results were reported with mean and SD.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Comment: Described as double‐blind and matching placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

Comment: High dropout rate: 59 out of 121 withdrew (48.7%)

Selective reporting (reporting bias)

Low risk

Comment: No apparent selective outcome reporting

Other bias

Low risk

Comment: Apparently no other bias

Schweizer 1991

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Blinding

Duration: 8 to 10 weeks

Single‐centre

Participants

Baseline characteristics

No significant differences between the intervention groups, therefore combined group reported:

  • Male, N (%): 39 (48)

  • Age, mean (SD): 47 (15)

Inclusion criteria: 18 years or older and receiving a daily dose of benzodiazepine continuously for at least the past year. Individuals were entered directly into the study if they were taking diazepam, alprazolam, or lorazepam in a dose of 40 mg or less diazepam equivalents (5 mg of diazepam = 0.5 mg of alprazolam = 1.0 mg of lorazepam). 6 individuals who were receiving a different benzodiazepine were switched to diazepam in an equivalent dose and stabilised for 3 weeks before entry into the study.

Exclusion criteria: A history in the past year of alcohol or substance abuse or dependence, any acute or unstable medical condition, or not practicing adequate contraception

Pretreatment: No significant group differences

Interventions

Intervention characteristics

Benzodiazepine taper schedule: 1 to 2 weeks pretreatment, benzodiazepine taper was initiated at a rate of 25% per week and completed over 4 weeks. Once the taper phase was completed and the participant had discontinued benzodiazepine intake, treatment with carbamazepine or placebo was continued for 2 to 4 weeks, then discontinued abruptly.

  1. Benzodiazepine taper schedule + carbamazepine, 200 to 800 mg/d(N = 27) (only 19 entered the benzodiazepine taper phase)

  2. Benzodiazepine taper schedule + placebo(N = 28) (only 21 entered the benzodiazepine taper phase)

Outcomes

  • Benzodiazepine withdrawal symptoms: Patient Withdrawal Checklist

  • Benzodiazepine cessation

Identification

Sponsorship source: This investigation was supported by Public Health Service research grant MH‐08957, Washington, DC.

Country: USA

Setting: Outpatients

Declarations of interest: Not mentioned

Author's name: Edward Schweizer

Institution: Psychopharmacology Research Unit, Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia

Email:

Address: 203 Piersol Bldg, Hospital of the University of Pennsylvania, 3400 Spruce St, Philadelphia, PA 19104

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "random and double‐blind fashion", "Carbamazepine was provided in capsules that were identical to the placebo"

Comment: Done

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "Carbamazepine levels were obtained in 12 of 19 patients, with the treating psychiatrist kept blind to the results."

Comment: Judged as done since treating psychiatrist was kept blind

Incomplete outcome data (attrition bias)
All outcomes

High risk

Comment: Total dropout rate 15/55 (27%), 8 (30%) in the carbamazepine group and 7 (25%) in the placebo group

Selective reporting (reporting bias)

Low risk

Comment: No obvious selective outcome reporting

Other bias

Low risk

Comment: No other obvious sources of bias

Schweizer 1995

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 10 weeks

Single‐centre

Participants

Baseline characteristics

Not reported

Inclusion criteria: At least 18 years of age and taking diazepam, lorazepam, or alprazolam on a continuous daily basis for at least 1 year

Exclusion criteria: Individuals were excluded from the study if they had a history of alcohol or substance abuse or dependence in the past year, or if they had any acute or unstable medical condition. Men could be of any age, while women had to be at least 2 years' postmenopause, or to have undergone an ovariectomy.

Pretreatment: Unknown

Interventions

Intervention characteristics

Benzodiazepine taper schedule: 2 to 3 weeks pretreatment with experimental drug, taper at the rate of 25% per week, after completion of taper experimental drug was continued for 4 weeks, then abruptly discontinued

  1. Benzodiazepine taper schedule + progesterone minimum 1200 mg/d (up to 3600 mg as tolerated) (N = 30)

  2. Benzodiazepine taper schedule + placebo (N = 13)

Outcomes

  • Benzodiazepine withdrawal symptoms, Physician Withdrawal Checklist

  • Benzodiazepine cessation

  • Anxiety, HAM‐A

  • Non‐serious adverse events: sedation

Identification

Sponsorship source: This study was supported by USPHS Research Grant MHO‐8957.

Country: USA

Setting: Outpatients

Declarations of interest: Not mentioned

Author's name: Edward Schweizer

Institution: University Science Center, Suite 803, 3600 Market Street, Philadelphia, PA 19104‐2649, USA

Email:

Address: University Science Center, Suite 803, 3600 Market Street, Philadelphia, PA 19104‐2649, USA

Notes

Benzodiazepine withdrawal symptoms and anxiety not reported appropriately.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "under random, double‐blind conditions", "either micronized oral progesterone in 300 mg capsules or matched placebo"

Comment: Described as double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

Comment: 8 (27%) participants in progesterone group versus 1 (8%) participant in placebo group dropped out during the pretreatment phase (due to sedation as side effect).

Selective reporting (reporting bias)

Low risk

Comment: No apparent selective outcome reporting

Other bias

Low risk

Comment: No apparent other bias

Tyrer 1981

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 2 weeks

Multicentre

Participants

Baseline characteristics

Not reported

Inclusion criteria: Monotherapy with diazepam or lorazepam, medication use for at least 4 months regularly, and were thought not to require continued prescription

Exclusion criteria: Drugs other than benzodiazepines

Pretreatment: Not reported

Interventions

Intervention characteristics

Benzodiazepine taper schedule: abrupt cessation

  1. Benzodiazepines stopped and replaced by propranolol 20 mg x 3, increased to 40 mg x 3 if necessary(N = 20)

  2. Benzodiazepines stopped and replaced by placebo(N = 20)

Outcomes

  • Relapse to benzodiazepine use

  • Benzodiazepine withdrawal symptoms (authors' own scale, self rating of symptoms)

Identification

Sponsorship source: Not stated

Country: UK

Setting: General practice and outpatient psychiatric clinics

Declarations of interest: Not mentioned

Authors name: Peter Tyrer

Institution: Mapperley Hospital, Nottingham, and Poisons Unit, New Cross Hospital, London

Email:

Address: Mapperley Hospital, Porchester Road, Nottingham NG3 6AA, UK

Notes

Figure 1 reports withdrawal symptoms in each group, only mean score not SD, and no other measures from which the SD can be calculated. Since this was the only identified study using propranolol, and since the scale for withdrawal symptoms was not a validated scale used in other included studies, it was not possible to safely impute values for SD.

Otherwise only dropout rate was reported for each group (55% in placebo group and 36% in propranolol group) = patients returning to same benzodiazepine treatment as previously.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "If patients agreed to enter this double‐blind study their benzodiazepines were stopped and replaced by propranolol or placebo tablets of identical appearance"

Comment: Done

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Comment: Daily self ratings, no third party involved in outcome assessment

Incomplete outcome data (attrition bias)
All outcomes

High risk

Quote: "Of the 40 patients entering the study 18 (45%) dropped out during the two week period and took their benzodiazepine drugs again."

Comment: High attrition rate

Selective reporting (reporting bias)

Low risk

Comment: No obvious selective outcome reporting

Other bias

High risk

Quote: "I.C.I. Pharmaceuticals Division for providing the propranolol and placebo tablets."

Comment: Role, if any, in the analyses not described.

Tyrer 1996

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 12 weeks

Single‐centre

Participants

Baseline characteristics

Dosulepin

  • Years of benzodiazepine use, median: 10

  • Benzodiazepine dose (mg diazepam equivalent), mean: 8.0

Placebo

  • Years of benzodiazepine use, median: 10

  • Benzodiazepine dose (mg diazepam equivalent), mean: 8.3

Inclusion criteria: Use of benzodiazepines for at least 6 months and had tried unsuccessfully to reduce or stop benzodiazepines due to apparent withdrawal symptoms, no other medication, written consent

Exclusion criteria: Hypertension or the psychiatric diagnoses of major depressive disorder, a psychotic disorder, or melancholia

Pretreatment: Not reported

Interventions

Intervention characteristics

Benzodiazepine taper schedule: reduction of the initial dosage by 20% every 2 weeks with the intention of stopping benzodiazepines entirely at week 8

  1. Benzodiazepine taper schedule + dosulepin 150 mg/d(N = 41).

  2. Benzodiazepine taper schedule + placebo(N = 46).

Outcomes

  • Benzodiazepine cessation

  • Benzodiazepine withdrawal symptoms

  • Anxiety, HADS‐A

Identification

Sponsorship source: Research Department of Boots Drug Company funded the study.

Country: UK

Setting: Outpatients

Declarations of interest: Not mentioned

Authors name: Peter Tyrer

Institution: St Charles Hospital, London

Email:

Address: St Charles Hospital, London WlO 6DZ

Notes

For benzodiazepine withdrawal symptoms and anxiety, means were available from graphs only, but SDs were not reported. It was not possible to calculate SD from the presented data.

Adverse events insufficiently reported.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Randomisation process not described.

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Quote: "under cover of randomly allocated dothiepin or placebo tablets...administered using double‐blind procedure."

Comment: What has been done to ensure blinding of participants and study personnel is not described.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: Not sufficiently described

Incomplete outcome data (attrition bias)
All outcomes

High risk

Quote: "During the 14 weeks 45 patients (21 (51%) allocated to dothiepin and 24 (52%) to placebo) withdrew from the study."

Comment: High attrition rate

Selective reporting (reporting bias)

Low risk

Comment: No apparent selective outcome reporting

Other bias

High risk

Comment: Funded by a drug company. Role of funding source not described.

Udelman 1990

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 6 to 12 weeks depending on the alprazolam starting dose

Multicentre

Participants

Baseline characteristics

Buspirone

  • Male, N (%): 17 (47)

  • Age, mean (range): 40 (24 to 62)

  • Number of benzodiazepine compounds: 1 (alprazolam only)

Placebo

  • Male, N (%): 18 (50)

  • Age, mean (range): 44 (24 to 63)

  • Number of benzodiazepine compounds: 1 (alprazolam only)

Inclusion criteria: 18 to 70 years of age, primary clinical anxiety, alprazolam pharmacotherapy for at least 3 months 0.75 to 3 mg daily, good physical health

Exclusion criteria: Significant or uncontrolled organic disease, epilepsy or seizures, nursing/pregnant/not using contraceptive measures, substance use disorder, primary depression, panic disorder, psychosis, severe behaviour disorder, organic mental disorders, serious psychosomatic disorders, hypersensitivity to study drug, other drugs (psychotropics, beta‐blockers, carbamazepine, clonazepam) within 1 month before start of the study

Pretreatment: No significant pretreatment group differences

Interventions

Intervention characteristics

Benzodiazepine taper schedule: 2 weeks of concurrent treatment (study drug + stable benzodiazepine dosage), from the third week tapering of alprazolam 0.5 mg/day each week until the total daily dose was 1.5 mg, after which the rate of tapering was 0.25 mg/day each week; completion of the tapering process was to take from 2 to 8 weeks depending on the alprazolam starting dose. At completion, participants were to continue receiving the study drugs (buspirone or placebo) for an additional 2 weeks.

  1. Benzodiazepine taper schedule + buspirone 15 mg/d(N = 36)

  2. Benzodiazepine taper schedule + placebo(N = 36)

Outcomes

  • Benzodiazepine withdrawal symptoms: Abstinence Rating Scale

  • Benzodiazepine cessation

  • Adverse events

  • Anxiety: HAM‐A

  • Discontinuation due to adverse events

Identification

Sponsorship source: Bristol‐Myers Squibb

Country: USA

Setting: Outpatients

Declarations of interest: Not mentioned

Authors name: Harold D Udelman

Institution: Biomedical Stress Research Foundation, Phoenix, Arizona, USA

Email: Not available

Address: Biomedical Stress Research Foundation, 45 East Born Road, Phoenix, Arizona 85012

Notes

Means only given in figures (HAM‐A and benzodiazepine withdrawal symptoms), no SDs reported, not possible to impute in a methodologically valid way.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Patients meeting the selection criteria were randomly assigned to one of two groups to receive either buspirone or placebo...Both buspirone and placebo (indistinguishable in physical appearance) were to be administered at a fixed dose..."

Comment: Placebo was described as indistinguishable in physical appearance.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: Not sufficiently described

Incomplete outcome data (attrition bias)
All outcomes

High risk

Comment: High attrition rate in buspirone group (42%) and in placebo group (53%)

Selective reporting (reporting bias)

Low risk

Comment: Protocol not available, but no indication of selective outcome reporting.

Other bias

High risk

Comment: Role of funding pharmaceutical company not described.

Vissers 2007

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 16 weeks (10 weeks to taper off and 6 weeks post‐taper)

9 general practices

Participants

Baseline characteristics

Melatonin

  • Male, N (%): 6 (30)

  • Years of benzodiazepine use, 1 to 5 years: 7 (35%)

  • Years of benzodiazepine use, 6 to 9 years: 4 (20%)

  • Years of benzodiazepine use, >= 10 years: 9 (45%)

  • Age: < 50 years: 3 (15%), 50 to 59: 3 (15%), 60 to 69: 6 (30%), 70 to 79: 7 (35%), > 80: 1 (5%)

  • Benzodiazepine dose: low: 11 (55%), moderate: 4 (20%), high: 5 (25%)

Placebo

  • Male, N (%): 10 (56)

  • Years of benzodiazepine use, 1 to 5 years: 7 (39%)

  • Years of benzodiazepine use, 6 to 9 years: 4 (22%)

  • Years of benzodiazepine use, >= 10 years: 6 (34%)

  • Age: < 50 years: 3 (17%), 50 to 59: 3 (17%), 60 to 69: 7 (39%), 70 to 79: 4 (22%), > 80: 1 (5%)

  • Benzodiazepine dose: low: 14 (78%), moderate: 0, high: 5 (25%)

Inclusion criteria: Adult patients who used benzodiazepines as a sleeping medication for more than 3 months (defined as long‐term use) at a minimum of 3 days per week

Exclusion criteria: Use of more than 1 benzodiazepine at the same time, use of another type of sleep medication, use of stimulants and alcohol misuse (according to individual's GP), serious mental/somatic disease, or unfit to participate

Pretreatment: No significant pretreatment differences

Interventions

Intervention characteristics

Benzodiazepine taper schedule: the benzodiazepine dose was converted to an equivalent dose of diazepam, which was stabilised for 2 weeks and then further converted every 2 weeks to 75%, 50%, 25%, 12.5%, and 0% of the original dose.

  1. Benzodiazepine taper schedule + melatonin 5 mg (4 hours before bed) (N = 20)

  2. Benzodiazepine taper schedule + placebo (N = 18)

Outcomes

  • Benzodiazapine cessation

  • Alcohol consumption

  • Relapse to benzodiazepine use

  • Insomnia

Identification

Sponsorship source: Not described

Country: The Netherlands

Setting: Outpatients in GP

Declarations of interest: Not mentioned

Authors name: Vissers FHJA

Institution: Department of General Practice, Maastricht University

Email: [email protected]

Address: Department of General Practice, Maastricht University, P.O. Box 616, Maastricht 6200 MD, the Netherlands

Notes

Insomnia: the Sleep Wake Experience List: mean and SD not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not sufficiently described

Allocation concealment (selection bias)

Unclear risk

Comment: Not sufficiently described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Quote: "The patients, their GPs and the principal investigator were blinded for the study medication."

Comment: What has been done to ensure blinding of participants and study personnel is not described.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: Insufficient information to judge the risk of bias

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: All randomised participants were analysed.

Selective reporting (reporting bias)

Unclear risk

Comment: Benzodiazepine withdrawal symptoms ambiguously reported.

Other bias

Low risk

Comment: No other apparent biases, funding not reported

Vorma 2011

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: None ‐ open‐label

Duration: 3 weeks of inpatient treatment

Single‐centre

Participants

Baseline characteristics

Valproate

  • Male, N (%): 12 (86)

  • Age, mean (SD): 32 (6.7)

  • Employed, N (%): 0

  • Years of opioid use, mean (SD): 11 (5.5)

  • Years of benzodiazepine use, mean (SD): 12 (7.1)

  • Benzodiazepine dose, mg (diazepam equivalent), median (range): 60 (20 to 160)

Control

  • Male, N (%): 10 (62)

  • Age, mean (SD): 32 (5.3)

  • Employed, N (%): 3 (19)

  • Years of opioid use, mean (SD): 10 (4.6)

  • Years of benzodiazepine use, mean (SD): 9 (5.2)

  • Benzodiazepine dose, mg (diazepam equivalent), median (range): 30 (8 to 75)

Inclusion criteria: DSM‐IV criteria for opioid dependence and benzodiazepine dependence

Exclusion criteria: Pregnancy, active medical illnesses or severe mental disorders, history of convulsions, or unable to speak Finnish

Pretreatment: At baseline, the median diazepam‐equivalent dose was 60 mg daily (range 20 to 160 mg) in the valproate group and 30 mg (range 8 to 75 mg) in the control group.

Interventions

Intervention characteristics

Benzodiazepine taper schedule: reduction with 10 mg diazepam equivalents daily until 40 mg per day, after which reductions were 5 mg daily

  1. Benzodiazepine taper schedule + valproate 20 mg/kg(N = 14)

  2. Benzodiazepine taper schedule only (control group)(N = 16)

Outcomes

  • Benzodiazepine withdrawal symptoms, Clinical Institute Withdrawal Assessment Scale ‐ Benzodiazepines

  • Serious adverse events

Identification

Sponsorship source: The study was supported by Annual EVO Financing (special government subsidies) from the Department of Psychiatry, Helsinki University Central Hospital. No support was provided by any pharmaceutical company.

Country: Finland

Setting: Opioid maintenance treatment, inpatient setting, very rapid benzodiazepine‐tapering regimen

Declarations of interest: None

Author's name: Helena Vorma

Institution: Helsinki University Central Hospital, Department of Psychiatry

Email: [email protected]

Address: Helsinki University Hospital, Department of Psychiatry, P.O. Box 590, FI‐00029 HUS, Finland

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described

Allocation concealment (selection bias)

Low risk

Quote: "To prevent unequal treatment group sizes, we used block randomisation in blocks of six subjects. Sealed envelopes were used to keep the randomisation sequence unknown. The study was carried out as an open trial, with all outcome ratings assessed blindly to prevent detection bias."

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Comment: Not done, open‐label trial

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Comment: All outcome assessments were done blindly.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Quote: "Another 8 subjects discontinued participation in CIWA‐B ratings, but stayed in treatment."

Comment: Not described why and from which group, accounted for by LOCF, but difficult to judge if this might give rise to any kind of bias

Selective reporting (reporting bias)

Unclear risk

Comment: No protocol available; despite the rapid benzodiazepine taper regimen, it is remarkable that there is no indication of the benzodiazepine‐tapering success

Other bias

Unclear risk

Comment: Big difference in benzodiazepine dose between groups at baseline (valproate 60 mg/day, control group 30 mg/day)

Zhang 2013

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Unknown

Duration: 3 months

Single‐centre

Participants

Baseline characteristics

Trazodone

  • Male, N (%): 8 (40)

  • Age, mean (SD): 45.73 (9.51)

  • Years of benzodiazepine use, mean (SD): 2.20 (1.20)

Placebo

  • Male, N (%): 8 (44)

  • Age, mean (SD): 44.92 (9.41)

  • Years of benzodiazepine use, mean (SD): 2.20 (1.10)

Inclusion criteria: Benzodiazepine dependence syndrome (Criteria of Mental Disorders in China, Third Edition), insomnia

Exclusion criteria: Abuse of alcohol or other psychoactive drugs, other mental disorders, serious somatic illness, allergic to study medication, suicidal risk, pregnancy, breastfeeding, lack of consent

Pretreatment: No significant pretreatment group differences

Interventions

Benzodiazepine taper schedule: benzodiazepine reduced to half dosage, when participant has had stable sleep for 5 days, then dosage is halved again and so forth.

  1. Benzodiazepine taper schedule + trazodone 50 to 300 mg/day(N = 20)

  2. Benzodiazepine taper schedule + placebo(N = 18)

Outcomes

  • Benzodiazepine withdrawal symptoms: Withdrawal Symptoms Checklist

  • Anxiety: HAM‐A

Identification

Sponsorship source: Not reported

Country: China

Setting: Outpatients

Declarations of interest: Not mentioned

Author's name: Zhang Hong‐Ju

Institution: He'nan Provincial People's Hospital, Zhengzhou

Email: [email protected]

Address: Department of Neurology, He'nan Provincial People's Hospital, Zhengzhou 450003, He'nan, China

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: Not described

Allocation concealment (selection bias)

Unclear risk

Comment: Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Comment: Not described as blinded or open‐label

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: Not described as blinded or open‐label

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: Attrition: 2 of 38 (5%) participants

Selective reporting (reporting bias)

High risk

Comment: No data on use of benzodiazepine at follow‐up

Other bias

Low risk

Comment: No other apparent sources of bias

Zitman 2001

Methods

Study design: Randomised controlled trial

Study grouping: Parallel group

Blinding: Double

Duration: 12 weeks

Multicentre

Participants

Baseline characteristics

Paroxetine

  • Male, N (%): 19 (27)

  • Age, mean (range): 55 (24 to 84)

  • Years of benzodiazepine use, mean (range): 5.5 (0.3 to 27)

  • Benzodiazepine dose, mg (diazepam equivalent), median (range): 9 (1 to 30)

Placebo

  • Male, N (%): 36 (28)

  • Age, mean (range): 57 (25 to 84)

  • Years of benzodiazepine use, mean (range): 6.4 (0.3 to 25)

  • Benzodiazepine dose, mg (diazepam equivalent), median (range): 9 (0.5 to 60)

Inclusion criteria: Benzodiazepine use for at least 3 months, a diagnosis of major depressive disorder (DSM‐III‐R), at least 18 years of age, written informed consent

Exclusion criteria: Depression caused by organic factors, psychosis, schizophrenia, pregnancy, lactation, childbearing potential with a lack of adequate contraception, severe concomitant medical conditions, history of seizure disorders, use of other psychotropic medication during the 3 months prior to screening, clinically significant abnormalities in haematology or clinical chemistry, misuse of alcohol or illicit drugs, excessive use of benzodiazepines (more than 3 times the maximal dose), current suicidal risk

Pretreatment: No significant pretreatment group differences

Interventions

Intervention characteristics

Benzodiazepine taper schedule: weeks 1 to 4: transfer to diazepamweeks 5 to 10: constant dose; weeks 11 to 12: 25% reduction per week; weeks 13 to 14: 12.5% reduction in 4 steps to 0

  1. Benzodiazepine taper schedule + paroxetine 20 mg/d(N = 70)

  2. Benzodiazepine taper schedule + placebo(N = 129)

Outcomes

  • Benzodiazepine withdrawal symptoms, BWSQ

  • Benzodiazepine cessation

  • Serious adverse events

  • Anxiety (Spielberger State‐Trait Anxiety Inventory (STAI‐DY1 and 2))

  • Non‐serious adverse events

Identification

Sponsorship source: One of the authors was employed by SmithKline Beecham, which also funded the study.

Country: The Netherlands

Setting: Outpatients

Declarations of interest: Not mentioned

Authors name: Frans Zitman

Institution: Department of Psychiatry, Leiden and UMC Stat Radbond, Nijmegen

Email: [email protected]

Address: Department of Psychiatry, Leiden, BIP, P.O. Box 9600, 2300 RC Leiden

Notes

Anxiety: Not sufficiently reported

Withdrawal symptoms: Data not reported for placebo vs paroxetine, but for success vs no‐success groups.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "A randomisation list (1‐330) in blocks of six was obtained by using the blocks of six was obtained by using the random number generator of SPSS/PC+ (SPSS, 1997)."

Comment: Done

Allocation concealment (selection bias)

Low risk

Quote: "Based on this list, study medication (paroxetine, placebo) was blister packed and wrapped by Genfarma, The Netherlands. Blocks were sequentially distributed to GPs. Unused blocks were reallocated. The list was kept by the Medical Adviser on Safety of the medical department Adviser on Safety of the medical department of SmithKline Beecham, The Netherlands."

Comment: Done

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Quote: "Patients were randomised to 20 mg of paroxetine or placebo in a 1:2 double‐blind fashion"

Comment: What was done to ensure blinding of participants and personnel in terms of matching paroxetine/placebo is not described.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "After the database was closed and basic descriptive analyses were done, the actual codes were added to the database"

Comment: Done

Incomplete outcome data (attrition bias)
All outcomes

High risk

Comment: 50% completed the programme in the paroxetine group and 43% in the placebo group.

Selective reporting (reporting bias)

Unclear risk

Comment: Some outcome data not reported for randomisation groups, but for success and no‐success group instead.

Other bias

High risk

Comment: Role of funding pharmaceutical company not described.

AEs: adverse events
BWSQ: Benzodiazepine Withdrawal Symptom Questionnaire
CI: confidence interval
CIWA‐B: Clinical Institute Withdrawal Assessment Scale ‐ Benzodiazepines
DSM‐III‐R: Diagnostic and Statistical Manual of Mental Disorders, Revised 3rd Edition
DSM‐IV‐TR: Diagnostic and Statistical Manual of Mental Disorders, 4th Edition, Text Revision
ECG: electrocardiogram
GAD: generalised anxiety disorder
GP: general practitioner
HADS‐A Hospital Anxiety and Depression Scale ‐ Anxiety subscale
HAM‐A: Hamilton Anxiety Rating Scale
HAM‐D: Hamilton Depression Rating Scale
ICD‐10: International Statistical Classification of Diseases and Related Health Problems, 10th revision
ITT: intention‐to‐treat
IV: intravenous
LOCF: last observation carried forward
MADRS: Montgomery–Åsberg Depression Rating Scale
MMT: methadone maintenance treatment
NIMH: National Institute of Mental Health
NS: not specified
PSQI: Pittsburgh Sleep Quality Index
PTSD: post‐traumatic stress disorder
PWC: Physician Withdrawal Checklist
SAEs: serious adverse events
SD: standard deviation
SE: standard error

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Allain 1998

Wrong patient population: duration of benzodiazepine use above 1 month and no mention of dependence

Avedisova 2007

Wrong study design: not randomised

Bobes 2012

Wrong study design: uncontrolled, observational study

Bourgeois 2014

Wrong study design: uncontrolled, observational study

Cantopher 1990

Wrong study design: co‐intervention not delivered equally in both intervention groups

Cohen‐Mansfield 1999

Wrong study design: observational study

Declerck 1999

Wrong intervention: switch to benzodiazepine‐like drug without discontinuation

Emara 2009

Wrong study design: not randomised

Garcia‐Borreguero 1992

Wrong study design: not randomised

Hallstrom 1988

Wrong study design: co‐intervention not delivered equally in both intervention groups

Isaka 2009

Wrong patient population: not chronic benzodiazepine users

Lahteenmaki 2014

Wrong patient population: not chronic benzodiazepine users

Lemoine 1997

Wrong study design: co‐intervention not delivered equally in both intervention groups

Lopatko 2006

Wrong study design: not randomised

Nakajima 2007

Wrong study design: observational study

Petrovic 2002

Wrong study design: the study investigated gradual benzodiazepine withdrawal versus abrupt discontinuation

Rocco 1992

Wrong study design: not randomised

Rubio 2009

Wrong study design: observational study

Saxon 1997

Wrong study design: study not designed to evaluate benzodiazepine discontinuation

Shapiro 1995

Wrong intervention: switch to benzodiazepine‐like drug without discontinuation

Vescovi 1987

Wrong study design: study not designed to evaluate benzodiazepine discontinuation

Weizman 2003

Wrong study design: not randomised

Data and analyses

Open in table viewer
Comparison 1. Valproate versus placebo or no intervention

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation, end of intervention Show forest plot

1

27

Risk Ratio (M‐H, Random, 95% CI)

2.55 [1.08, 6.03]

Analysis 1.1

Comparison 1 Valproate versus placebo or no intervention, Outcome 1 Benzodiazepine discontinuation, end of intervention.

Comparison 1 Valproate versus placebo or no intervention, Outcome 1 Benzodiazepine discontinuation, end of intervention.

2 Relapse to benzodiazepine use, end of intervention Show forest plot

1

27

Risk Ratio (M‐H, Random, 95% CI)

0.31 [0.11, 0.90]

Analysis 1.2

Comparison 1 Valproate versus placebo or no intervention, Outcome 2 Relapse to benzodiazepine use, end of intervention.

Comparison 1 Valproate versus placebo or no intervention, Outcome 2 Relapse to benzodiazepine use, end of intervention.

3 Benzodiazepine discontinuation, longest follow‐up Show forest plot

1

24

Risk Ratio (M‐H, Random, 95% CI)

1.57 [0.80, 3.09]

Analysis 1.3

Comparison 1 Valproate versus placebo or no intervention, Outcome 3 Benzodiazepine discontinuation, longest follow‐up.

Comparison 1 Valproate versus placebo or no intervention, Outcome 3 Benzodiazepine discontinuation, longest follow‐up.

4 Relapse to benzodiazepine use, longest follow‐up Show forest plot

1

24

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.13, 1.39]

Analysis 1.4

Comparison 1 Valproate versus placebo or no intervention, Outcome 4 Relapse to benzodiazepine use, longest follow‐up.

Comparison 1 Valproate versus placebo or no intervention, Outcome 4 Relapse to benzodiazepine use, longest follow‐up.

5 Anxiety: HAM‐A (Hamilton Anxiety Rating Scale), end of intervention Show forest plot

1

27

Mean Difference (IV, Random, 95% CI)

‐0.40 [‐6.47, 5.67]

Analysis 1.5

Comparison 1 Valproate versus placebo or no intervention, Outcome 5 Anxiety: HAM‐A (Hamilton Anxiety Rating Scale), end of intervention.

Comparison 1 Valproate versus placebo or no intervention, Outcome 5 Anxiety: HAM‐A (Hamilton Anxiety Rating Scale), end of intervention.

6 Benzodiazepine withdrawal symptoms, end of intervention Show forest plot

2

56

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

‐0.15 [‐0.68, 0.37]

Analysis 1.6

Comparison 1 Valproate versus placebo or no intervention, Outcome 6 Benzodiazepine withdrawal symptoms, end of intervention.

Comparison 1 Valproate versus placebo or no intervention, Outcome 6 Benzodiazepine withdrawal symptoms, end of intervention.

6.1 Physician Withdrawal Checklist

1

27

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

‐0.01 [‐0.77, 0.74]

6.2 CIWA‐B (Clinical Institute Withdrawal Assessment Scale ‐ Benzodiazepines)

1

29

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

‐0.28 [‐1.01, 0.45]

7 Discontinuation due to adverse events Show forest plot

1

29

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Analysis 1.7

Comparison 1 Valproate versus placebo or no intervention, Outcome 7 Discontinuation due to adverse events.

Comparison 1 Valproate versus placebo or no intervention, Outcome 7 Discontinuation due to adverse events.

8 Serious adverse events Show forest plot

1

29

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Analysis 1.8

Comparison 1 Valproate versus placebo or no intervention, Outcome 8 Serious adverse events.

Comparison 1 Valproate versus placebo or no intervention, Outcome 8 Serious adverse events.

Open in table viewer
Comparison 2. Carbamazepine versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation, end of intervention Show forest plot

3

147

Risk Ratio (M‐H, Random, 95% CI)

1.33 [0.99, 1.80]

Analysis 2.1

Comparison 2 Carbamazepine versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.

Comparison 2 Carbamazepine versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.

2 Benzodiazepine withdrawal symptoms Show forest plot

2

76

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

‐1.14 [‐2.43, 0.16]

Analysis 2.2

Comparison 2 Carbamazepine versus placebo, Outcome 2 Benzodiazepine withdrawal symptoms.

Comparison 2 Carbamazepine versus placebo, Outcome 2 Benzodiazepine withdrawal symptoms.

2.1 Physician Withdrawal Checklist

1

36

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

‐1.82 [‐2.61, ‐1.03]

2.2 Patient Withdrawal Checklist

1

40

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

‐0.50 [‐1.13, 0.13]

3 Benzodiazepine discontinuation, longest follow‐up Show forest plot

1

40

Risk Ratio (M‐H, Random, 95% CI)

1.41 [0.86, 2.29]

Analysis 2.3

Comparison 2 Carbamazepine versus placebo, Outcome 3 Benzodiazepine discontinuation, longest follow‐up.

Comparison 2 Carbamazepine versus placebo, Outcome 3 Benzodiazepine discontinuation, longest follow‐up.

4 Relapse to benzodiazepine use Show forest plot

1

36

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.08, 1.44]

Analysis 2.4

Comparison 2 Carbamazepine versus placebo, Outcome 4 Relapse to benzodiazepine use.

Comparison 2 Carbamazepine versus placebo, Outcome 4 Relapse to benzodiazepine use.

5 Serious adverse events Show forest plot

1

36

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Analysis 2.5

Comparison 2 Carbamazepine versus placebo, Outcome 5 Serious adverse events.

Comparison 2 Carbamazepine versus placebo, Outcome 5 Serious adverse events.

6 Non‐serious adverse events Show forest plot

1

36

Risk Ratio (M‐H, Random, 95% CI)

7.0 [0.39, 126.48]

Analysis 2.6

Comparison 2 Carbamazepine versus placebo, Outcome 6 Non‐serious adverse events.

Comparison 2 Carbamazepine versus placebo, Outcome 6 Non‐serious adverse events.

7 Anxiety, HAM‐A Show forest plot

1

36

Mean Difference (IV, Random, 95% CI)

‐6.0 [‐9.58, ‐2.42]

Analysis 2.7

Comparison 2 Carbamazepine versus placebo, Outcome 7 Anxiety, HAM‐A.

Comparison 2 Carbamazepine versus placebo, Outcome 7 Anxiety, HAM‐A.

8 Discontinuation due to adverse events Show forest plot

1

36

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Analysis 2.8

Comparison 2 Carbamazepine versus placebo, Outcome 8 Discontinuation due to adverse events.

Comparison 2 Carbamazepine versus placebo, Outcome 8 Discontinuation due to adverse events.

Open in table viewer
Comparison 3. Lithium versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation Show forest plot

1

230

Risk Ratio (M‐H, Random, 95% CI)

1.05 [0.86, 1.28]

Analysis 3.1

Comparison 3 Lithium versus placebo, Outcome 1 Benzodiazepine discontinuation.

Comparison 3 Lithium versus placebo, Outcome 1 Benzodiazepine discontinuation.

2 Serious adverse events Show forest plot

1

230

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Analysis 3.2

Comparison 3 Lithium versus placebo, Outcome 2 Serious adverse events.

Comparison 3 Lithium versus placebo, Outcome 2 Serious adverse events.

3 Non‐serious adverse events Show forest plot

1

230

Risk Ratio (M‐H, Random, 95% CI)

1.06 [0.75, 1.49]

Analysis 3.3

Comparison 3 Lithium versus placebo, Outcome 3 Non‐serious adverse events.

Comparison 3 Lithium versus placebo, Outcome 3 Non‐serious adverse events.

4 Discontinuation due to adverse events Show forest plot

1

230

Risk Ratio (M‐H, Random, 95% CI)

1.38 [0.13, 15.03]

Analysis 3.4

Comparison 3 Lithium versus placebo, Outcome 4 Discontinuation due to adverse events.

Comparison 3 Lithium versus placebo, Outcome 4 Discontinuation due to adverse events.

Open in table viewer
Comparison 4. Pregabalin versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation, end of intervention Show forest plot

1

106

Risk Ratio (M‐H, Random, 95% CI)

1.44 [0.92, 2.25]

Analysis 4.1

Comparison 4 Pregabalin versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.

Comparison 4 Pregabalin versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.

2 Benzodiazepine withdrawal symptoms (Physician Withdrawal Checklist), end of intervention Show forest plot

1

106

Mean Difference (IV, Random, 95% CI)

‐3.1 [‐3.51, ‐2.69]

Analysis 4.2

Comparison 4 Pregabalin versus placebo, Outcome 2 Benzodiazepine withdrawal symptoms (Physician Withdrawal Checklist), end of intervention.

Comparison 4 Pregabalin versus placebo, Outcome 2 Benzodiazepine withdrawal symptoms (Physician Withdrawal Checklist), end of intervention.

3 Anxiety, HAM‐A, end of intervention Show forest plot

1

106

Mean Difference (IV, Random, 95% CI)

‐4.8 [‐5.28, ‐4.32]

Analysis 4.3

Comparison 4 Pregabalin versus placebo, Outcome 3 Anxiety, HAM‐A, end of intervention.

Comparison 4 Pregabalin versus placebo, Outcome 3 Anxiety, HAM‐A, end of intervention.

4 Serious adverse events Show forest plot

1

106

Risk Ratio (M‐H, Random, 95% CI)

0.67 [0.16, 2.85]

Analysis 4.4

Comparison 4 Pregabalin versus placebo, Outcome 4 Serious adverse events.

Comparison 4 Pregabalin versus placebo, Outcome 4 Serious adverse events.

5 Non‐serious adverse events Show forest plot

1

106

Risk Ratio (M‐H, Random, 95% CI)

1.08 [0.84, 1.40]

Analysis 4.5

Comparison 4 Pregabalin versus placebo, Outcome 5 Non‐serious adverse events.

Comparison 4 Pregabalin versus placebo, Outcome 5 Non‐serious adverse events.

6 Discontinuation due to adverse events Show forest plot

1

106

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.31, 2.59]

Analysis 4.6

Comparison 4 Pregabalin versus placebo, Outcome 6 Discontinuation due to adverse events.

Comparison 4 Pregabalin versus placebo, Outcome 6 Discontinuation due to adverse events.

Open in table viewer
Comparison 5. Captodiame versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine withdrawal symptoms, BWSQ (Benzodiazepine Withdrawal Symptom Questionnaire), end of intervention Show forest plot

1

81

Mean Difference (IV, Random, 95% CI)

1.00 [‐1.13, ‐0.87]

Analysis 5.1

Comparison 5 Captodiame versus placebo, Outcome 1 Benzodiazepine withdrawal symptoms, BWSQ (Benzodiazepine Withdrawal Symptom Questionnaire), end of intervention.

Comparison 5 Captodiame versus placebo, Outcome 1 Benzodiazepine withdrawal symptoms, BWSQ (Benzodiazepine Withdrawal Symptom Questionnaire), end of intervention.

2 Anxiety, HAM‐A, end of intervention Show forest plot

1

81

Mean Difference (IV, Random, 95% CI)

‐5.7 [‐6.05, ‐5.35]

Analysis 5.2

Comparison 5 Captodiame versus placebo, Outcome 2 Anxiety, HAM‐A, end of intervention.

Comparison 5 Captodiame versus placebo, Outcome 2 Anxiety, HAM‐A, end of intervention.

3 Serious adverse events Show forest plot

1

81

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Analysis 5.3

Comparison 5 Captodiame versus placebo, Outcome 3 Serious adverse events.

Comparison 5 Captodiame versus placebo, Outcome 3 Serious adverse events.

4 Non‐serious adverse events Show forest plot

1

81

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Analysis 5.4

Comparison 5 Captodiame versus placebo, Outcome 4 Non‐serious adverse events.

Comparison 5 Captodiame versus placebo, Outcome 4 Non‐serious adverse events.

Open in table viewer
Comparison 6. Paroxetine versus placebo or no intervention

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation, end of intervention Show forest plot

3

221

Risk Ratio (M‐H, Random, 95% CI)

1.45 [0.88, 2.39]

Analysis 6.1

Comparison 6 Paroxetine versus placebo or no intervention, Outcome 1 Benzodiazepine discontinuation, end of intervention.

Comparison 6 Paroxetine versus placebo or no intervention, Outcome 1 Benzodiazepine discontinuation, end of intervention.

2 Benzodiazepine withdrawal symptoms: BWSQ, end of intervention Show forest plot

2

99

Mean Difference (IV, Random, 95% CI)

‐3.57 [‐5.34, ‐1.80]

Analysis 6.2

Comparison 6 Paroxetine versus placebo or no intervention, Outcome 2 Benzodiazepine withdrawal symptoms: BWSQ, end of intervention.

Comparison 6 Paroxetine versus placebo or no intervention, Outcome 2 Benzodiazepine withdrawal symptoms: BWSQ, end of intervention.

3 Anxiety: HAM‐A, end of intervention Show forest plot

2

99

Mean Difference (IV, Random, 95% CI)

‐6.75 [‐9.64, ‐3.86]

Analysis 6.3

Comparison 6 Paroxetine versus placebo or no intervention, Outcome 3 Anxiety: HAM‐A, end of intervention.

Comparison 6 Paroxetine versus placebo or no intervention, Outcome 3 Anxiety: HAM‐A, end of intervention.

4 Benzodiazepine withdrawal symptoms: BWSQ, longest follow‐up: 6 months Show forest plot

1

54

Mean Difference (IV, Random, 95% CI)

‐0.13 [‐4.03, 3.77]

Analysis 6.4

Comparison 6 Paroxetine versus placebo or no intervention, Outcome 4 Benzodiazepine withdrawal symptoms: BWSQ, longest follow‐up: 6 months.

Comparison 6 Paroxetine versus placebo or no intervention, Outcome 4 Benzodiazepine withdrawal symptoms: BWSQ, longest follow‐up: 6 months.

5 Serious adverse events Show forest plot

2

176

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Analysis 6.5

Comparison 6 Paroxetine versus placebo or no intervention, Outcome 5 Serious adverse events.

Comparison 6 Paroxetine versus placebo or no intervention, Outcome 5 Serious adverse events.

6 Non‐serious adverse events Show forest plot

1

54

Risk Ratio (M‐H, Random, 95% CI)

1.33 [0.35, 5.03]

Analysis 6.6

Comparison 6 Paroxetine versus placebo or no intervention, Outcome 6 Non‐serious adverse events.

Comparison 6 Paroxetine versus placebo or no intervention, Outcome 6 Non‐serious adverse events.

Open in table viewer
Comparison 7. Tricyclic antidepressants versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation, end of intervention Show forest plot

2

105

Risk Ratio (M‐H, Random, 95% CI)

0.82 [0.52, 1.28]

Analysis 7.1

Comparison 7 Tricyclic antidepressants versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.

Comparison 7 Tricyclic antidepressants versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.

2 Anxiety: HAM‐A (change from baseline), end of intervention Show forest plot

2

66

Mean Difference (IV, Random, 95% CI)

‐10.38 [‐25.96, 5.20]

Analysis 7.2

Comparison 7 Tricyclic antidepressants versus placebo, Outcome 2 Anxiety: HAM‐A (change from baseline), end of intervention.

Comparison 7 Tricyclic antidepressants versus placebo, Outcome 2 Anxiety: HAM‐A (change from baseline), end of intervention.

3 Benzodiazepine discontinuation, longest follow‐up Show forest plot

1

47

Risk Ratio (M‐H, Random, 95% CI)

2.20 [1.27, 3.82]

Analysis 7.3

Comparison 7 Tricyclic antidepressants versus placebo, Outcome 3 Benzodiazepine discontinuation, longest follow‐up.

Comparison 7 Tricyclic antidepressants versus placebo, Outcome 3 Benzodiazepine discontinuation, longest follow‐up.

4 Benzodiazepine withdrawal symptoms (Physician Withdrawal Checklist), end of intervention Show forest plot

1

38

Mean Difference (IV, Random, 95% CI)

‐19.78 [‐20.25, ‐19.31]

Analysis 7.4

Comparison 7 Tricyclic antidepressants versus placebo, Outcome 4 Benzodiazepine withdrawal symptoms (Physician Withdrawal Checklist), end of intervention.

Comparison 7 Tricyclic antidepressants versus placebo, Outcome 4 Benzodiazepine withdrawal symptoms (Physician Withdrawal Checklist), end of intervention.

5 Relapse to benzodiazepine use, end of intervention Show forest plot

1

36

Risk Ratio (M‐H, Random, 95% CI)

2.0 [0.73, 5.47]

Analysis 7.5

Comparison 7 Tricyclic antidepressants versus placebo, Outcome 5 Relapse to benzodiazepine use, end of intervention.

Comparison 7 Tricyclic antidepressants versus placebo, Outcome 5 Relapse to benzodiazepine use, end of intervention.

6 Discontinuation due to adverse events Show forest plot

2

134

Risk Ratio (M‐H, Random, 95% CI)

1.16 [0.42, 3.21]

Analysis 7.6

Comparison 7 Tricyclic antidepressants versus placebo, Outcome 6 Discontinuation due to adverse events.

Comparison 7 Tricyclic antidepressants versus placebo, Outcome 6 Discontinuation due to adverse events.

Open in table viewer
Comparison 8. Alpidem versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation, end of intervention Show forest plot

1

25

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.17, 0.99]

Analysis 8.1

Comparison 8 Alpidem versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.

Comparison 8 Alpidem versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.

2 Withdrawal syndrome (clinical diagnosis), end of intervention Show forest plot

1

145

Risk Ratio (M‐H, Random, 95% CI)

4.86 [1.12, 21.14]

Analysis 8.2

Comparison 8 Alpidem versus placebo, Outcome 2 Withdrawal syndrome (clinical diagnosis), end of intervention.

Comparison 8 Alpidem versus placebo, Outcome 2 Withdrawal syndrome (clinical diagnosis), end of intervention.

3 Anxiety, HAM‐A, end of intervention Show forest plot

2

170

Mean Difference (IV, Random, 95% CI)

‐1.60 [‐4.64, 1.45]

Analysis 8.3

Comparison 8 Alpidem versus placebo, Outcome 3 Anxiety, HAM‐A, end of intervention.

Comparison 8 Alpidem versus placebo, Outcome 3 Anxiety, HAM‐A, end of intervention.

4 Relapse to benzodiazepine use, end of intervention Show forest plot

1

145

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.09, 1.20]

Analysis 8.4

Comparison 8 Alpidem versus placebo, Outcome 4 Relapse to benzodiazepine use, end of intervention.

Comparison 8 Alpidem versus placebo, Outcome 4 Relapse to benzodiazepine use, end of intervention.

5 Serious adverse events Show forest plot

1

25

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Analysis 8.5

Comparison 8 Alpidem versus placebo, Outcome 5 Serious adverse events.

Comparison 8 Alpidem versus placebo, Outcome 5 Serious adverse events.

6 Discontinuation due to adverse events Show forest plot

1

25

Risk Ratio (M‐H, Random, 95% CI)

0.46 [0.05, 4.46]

Analysis 8.6

Comparison 8 Alpidem versus placebo, Outcome 6 Discontinuation due to adverse events.

Comparison 8 Alpidem versus placebo, Outcome 6 Discontinuation due to adverse events.

Open in table viewer
Comparison 9. Buspirone versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation, end of intervention Show forest plot

4

143

Risk Ratio (M‐H, Random, 95% CI)

0.82 [0.49, 1.37]

Analysis 9.1

Comparison 9 Buspirone versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.

Comparison 9 Buspirone versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.

2 Anxiety: HAM‐A/Hospital Anxiety Depression Scale, end of intervention Show forest plot

2

41

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

0.18 [‐0.50, 0.86]

Analysis 9.2

Comparison 9 Buspirone versus placebo, Outcome 2 Anxiety: HAM‐A/Hospital Anxiety Depression Scale, end of intervention.

Comparison 9 Buspirone versus placebo, Outcome 2 Anxiety: HAM‐A/Hospital Anxiety Depression Scale, end of intervention.

3 Benzodiazepine withdrawal symptoms, end of intervention Show forest plot

1

17

Mean Difference (IV, Random, 95% CI)

4.69 [‐14.47, 23.85]

Analysis 9.3

Comparison 9 Buspirone versus placebo, Outcome 3 Benzodiazepine withdrawal symptoms, end of intervention.

Comparison 9 Buspirone versus placebo, Outcome 3 Benzodiazepine withdrawal symptoms, end of intervention.

4 Benzodiazepine discontinuation, longest follow‐up Show forest plot

1

23

Risk Ratio (M‐H, Random, 95% CI)

0.60 [0.34, 1.05]

Analysis 9.4

Comparison 9 Buspirone versus placebo, Outcome 4 Benzodiazepine discontinuation, longest follow‐up.

Comparison 9 Buspirone versus placebo, Outcome 4 Benzodiazepine discontinuation, longest follow‐up.

5 Benzodiazepine withdrawal symptoms, longest follow‐up Show forest plot

1

15

Mean Difference (IV, Random, 95% CI)

‐1.34 [‐14.31, 11.63]

Analysis 9.5

Comparison 9 Buspirone versus placebo, Outcome 5 Benzodiazepine withdrawal symptoms, longest follow‐up.

Comparison 9 Buspirone versus placebo, Outcome 5 Benzodiazepine withdrawal symptoms, longest follow‐up.

6 Anxiety, Hospital Anxiety Depression Scale, longest follow‐up Show forest plot

1

12

Mean Difference (IV, Random, 95% CI)

2.75 [‐2.83, 8.33]

Analysis 9.6

Comparison 9 Buspirone versus placebo, Outcome 6 Anxiety, Hospital Anxiety Depression Scale, longest follow‐up.

Comparison 9 Buspirone versus placebo, Outcome 6 Anxiety, Hospital Anxiety Depression Scale, longest follow‐up.

7 Discontinuation due to adverse events Show forest plot

1

72

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.01, 7.92]

Analysis 9.7

Comparison 9 Buspirone versus placebo, Outcome 7 Discontinuation due to adverse events.

Comparison 9 Buspirone versus placebo, Outcome 7 Discontinuation due to adverse events.

Open in table viewer
Comparison 10. Melatonin versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation, end of intervention Show forest plot

4

219

Risk Ratio (M‐H, Random, 95% CI)

1.20 [0.73, 1.96]

Analysis 10.1

Comparison 10 Melatonin versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.

Comparison 10 Melatonin versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.

2 Insomnia Show forest plot

3

150

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

‐1.23 [‐2.70, 0.23]

Analysis 10.2

Comparison 10 Melatonin versus placebo, Outcome 2 Insomnia.

Comparison 10 Melatonin versus placebo, Outcome 2 Insomnia.

2.1 PSQI (Pittsburgh Sleep Quality Index) global score (higher = worse), end of intervention

2

116

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

‐0.31 [‐0.92, 0.31]

2.2 Sleep quality (1 poorest, 10 excellent), end of intervention

1

34

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

‐3.34 [‐4.42, ‐2.26]

3 Discontinuation due to adverse events Show forest plot

2

120

Risk Ratio (M‐H, Random, 95% CI)

2.10 [0.20, 22.26]

Analysis 10.3

Comparison 10 Melatonin versus placebo, Outcome 3 Discontinuation due to adverse events.

Comparison 10 Melatonin versus placebo, Outcome 3 Discontinuation due to adverse events.

4 Benzodiazepine discontinuation, longest follow‐up Show forest plot

1

38

Risk Ratio (M‐H, Random, 95% CI)

1.03 [0.47, 2.27]

Analysis 10.4

Comparison 10 Melatonin versus placebo, Outcome 4 Benzodiazepine discontinuation, longest follow‐up.

Comparison 10 Melatonin versus placebo, Outcome 4 Benzodiazepine discontinuation, longest follow‐up.

5 Adverse events Show forest plot

1

86

Risk Ratio (M‐H, Random, 95% CI)

0.97 [0.52, 1.82]

Analysis 10.5

Comparison 10 Melatonin versus placebo, Outcome 5 Adverse events.

Comparison 10 Melatonin versus placebo, Outcome 5 Adverse events.

6 Relapse to benzodiazepine use, longest follow‐up Show forest plot

1

38

Risk Ratio (M‐H, Random, 95% CI)

1.8 [0.37, 8.68]

Analysis 10.6

Comparison 10 Melatonin versus placebo, Outcome 6 Relapse to benzodiazepine use, longest follow‐up.

Comparison 10 Melatonin versus placebo, Outcome 6 Relapse to benzodiazepine use, longest follow‐up.

Open in table viewer
Comparison 11. Flumazenil versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine withdrawal symptoms, end of intervention Show forest plot

3

58

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

‐0.95 [‐1.71, ‐0.19]

Analysis 11.1

Comparison 11 Flumazenil versus placebo, Outcome 1 Benzodiazepine withdrawal symptoms, end of intervention.

Comparison 11 Flumazenil versus placebo, Outcome 1 Benzodiazepine withdrawal symptoms, end of intervention.

2 Anxiety, HAM‐D (Hamilton Depression Rating Scale), end of intervention Show forest plot

1

18

Mean Difference (IV, Random, 95% CI)

‐1.3 [‐2.28, ‐0.32]

Analysis 11.2

Comparison 11 Flumazenil versus placebo, Outcome 2 Anxiety, HAM‐D (Hamilton Depression Rating Scale), end of intervention.

Comparison 11 Flumazenil versus placebo, Outcome 2 Anxiety, HAM‐D (Hamilton Depression Rating Scale), end of intervention.

3 Benzodiazepine mean dose, end of intervention Show forest plot

1

10

Mean Difference (IV, Random, 95% CI)

‐3.70 [‐22.06, 14.66]

Analysis 11.3

Comparison 11 Flumazenil versus placebo, Outcome 3 Benzodiazepine mean dose, end of intervention.

Comparison 11 Flumazenil versus placebo, Outcome 3 Benzodiazepine mean dose, end of intervention.

Open in table viewer
Comparison 12. Propranolol versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Relapse to benzodiazepine use, end of intervention: 2 weeks Show forest plot

1

40

Risk Ratio (M‐H, Random, 95% CI)

0.64 [0.31, 1.30]

Analysis 12.1

Comparison 12 Propranolol versus placebo, Outcome 1 Relapse to benzodiazepine use, end of intervention: 2 weeks.

Comparison 12 Propranolol versus placebo, Outcome 1 Relapse to benzodiazepine use, end of intervention: 2 weeks.

Open in table viewer
Comparison 13. Progesterone versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation, end of intervention Show forest plot

1

35

Risk Ratio (M‐H, Random, 95% CI)

1.15 [0.52, 2.54]

Analysis 13.1

Comparison 13 Progesterone versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.

Comparison 13 Progesterone versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.

2 Non‐serious adverse events Show forest plot

1

35

Risk Ratio (M‐H, Random, 95% CI)

3.13 [1.15, 8.54]

Analysis 13.2

Comparison 13 Progesterone versus placebo, Outcome 2 Non‐serious adverse events.

Comparison 13 Progesterone versus placebo, Outcome 2 Non‐serious adverse events.

Open in table viewer
Comparison 14. Magnesium aspartate versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation Show forest plot

1

144

Risk Ratio (M‐H, Random, 95% CI)

0.80 [0.66, 0.96]

Analysis 14.1

Comparison 14 Magnesium aspartate versus placebo, Outcome 1 Benzodiazepine discontinuation.

Comparison 14 Magnesium aspartate versus placebo, Outcome 1 Benzodiazepine discontinuation.

2 Anxiety Show forest plot

1

144

Mean Difference (IV, Random, 95% CI)

‐0.80 [‐2.73, 1.13]

Analysis 14.2

Comparison 14 Magnesium aspartate versus placebo, Outcome 2 Anxiety.

Comparison 14 Magnesium aspartate versus placebo, Outcome 2 Anxiety.

3 Relapse to benzodiazepine use Show forest plot

1

144

Risk Ratio (M‐H, Random, 95% CI)

0.93 [0.46, 1.87]

Analysis 14.3

Comparison 14 Magnesium aspartate versus placebo, Outcome 3 Relapse to benzodiazepine use.

Comparison 14 Magnesium aspartate versus placebo, Outcome 3 Relapse to benzodiazepine use.

4 Non‐serious adverse events Show forest plot

1

144

Risk Ratio (M‐H, Random, 95% CI)

0.49 [0.18, 1.35]

Analysis 14.4

Comparison 14 Magnesium aspartate versus placebo, Outcome 4 Non‐serious adverse events.

Comparison 14 Magnesium aspartate versus placebo, Outcome 4 Non‐serious adverse events.

5 Discontinuation due to adverse events Show forest plot

1

144

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.13, 1.18]

Analysis 14.5

Comparison 14 Magnesium aspartate versus placebo, Outcome 5 Discontinuation due to adverse events.

Comparison 14 Magnesium aspartate versus placebo, Outcome 5 Discontinuation due to adverse events.

Open in table viewer
Comparison 15. Homéogène 46/Sedatif PC (homeopathic drugs) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation Show forest plot

1

51

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.36, 1.70]

Analysis 15.1

Comparison 15 Homéogène 46/Sedatif PC (homeopathic drugs) versus placebo, Outcome 1 Benzodiazepine discontinuation.

Comparison 15 Homéogène 46/Sedatif PC (homeopathic drugs) versus placebo, Outcome 1 Benzodiazepine discontinuation.

Open in table viewer
Comparison 16. Carbamazepine versus tricyclic antidepressant

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation, end of intervention Show forest plot

1

48

Risk Ratio (M‐H, Random, 95% CI)

1.0 [0.78, 1.29]

Analysis 16.1

Comparison 16 Carbamazepine versus tricyclic antidepressant, Outcome 1 Benzodiazepine discontinuation, end of intervention.

Comparison 16 Carbamazepine versus tricyclic antidepressant, Outcome 1 Benzodiazepine discontinuation, end of intervention.

2 Relapse to benzodiazepine use Show forest plot

1

48

Risk Ratio (M‐H, Random, 95% CI)

1.0 [0.28, 3.54]

Analysis 16.2

Comparison 16 Carbamazepine versus tricyclic antidepressant, Outcome 2 Relapse to benzodiazepine use.

Comparison 16 Carbamazepine versus tricyclic antidepressant, Outcome 2 Relapse to benzodiazepine use.

3 Serious adverse events Show forest plot

1

48

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Analysis 16.3

Comparison 16 Carbamazepine versus tricyclic antidepressant, Outcome 3 Serious adverse events.

Comparison 16 Carbamazepine versus tricyclic antidepressant, Outcome 3 Serious adverse events.

Open in table viewer
Comparison 17. Cyamemazine versus bromazepam

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Relapse to benzodiazepine use, longest follow‐up Show forest plot

1

124

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.14, 0.78]

Analysis 17.1

Comparison 17 Cyamemazine versus bromazepam, Outcome 1 Relapse to benzodiazepine use, longest follow‐up.

Comparison 17 Cyamemazine versus bromazepam, Outcome 1 Relapse to benzodiazepine use, longest follow‐up.

2 Anxiety: Maximum amplitude of rebound (HAM‐A), end of intervention Show forest plot

1

160

Mean Difference (IV, Random, 95% CI)

0.50 [‐1.23, 2.23]

Analysis 17.2

Comparison 17 Cyamemazine versus bromazepam, Outcome 2 Anxiety: Maximum amplitude of rebound (HAM‐A), end of intervention.

Comparison 17 Cyamemazine versus bromazepam, Outcome 2 Anxiety: Maximum amplitude of rebound (HAM‐A), end of intervention.

3 Discontinuation due to adverse events Show forest plot

1

160

Risk Ratio (M‐H, Random, 95% CI)

2.87 [0.79, 10.44]

Analysis 17.3

Comparison 17 Cyamemazine versus bromazepam, Outcome 3 Discontinuation due to adverse events.

Comparison 17 Cyamemazine versus bromazepam, Outcome 3 Discontinuation due to adverse events.

4 Non‐serious adverse events Show forest plot

1

160

Risk Ratio (M‐H, Random, 95% CI)

1.68 [1.01, 2.78]

Analysis 17.4

Comparison 17 Cyamemazine versus bromazepam, Outcome 4 Non‐serious adverse events.

Comparison 17 Cyamemazine versus bromazepam, Outcome 4 Non‐serious adverse events.

Open in table viewer
Comparison 18. Zopiclone versus flunitrazepam

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Relapse to benzodiazepine use, longest follow‐up Show forest plot

1

18

Risk Ratio (M‐H, Random, 95% CI)

1.05 [0.23, 4.78]

Analysis 18.1

Comparison 18 Zopiclone versus flunitrazepam, Outcome 1 Relapse to benzodiazepine use, longest follow‐up.

Comparison 18 Zopiclone versus flunitrazepam, Outcome 1 Relapse to benzodiazepine use, longest follow‐up.

Study flow diagram.
Figuras y tablas -
Figure 1

Study flow diagram.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.
Figuras y tablas -
Figure 2

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figuras y tablas -
Figure 3

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

Trial Sequential Analysis of comparison: 2 Carbamazepine versus placebo, outcome: 2.1 Benzodiazepine discontinuation. Trial Sequential Analysis on benzodiazepine discontinuation in three trials was performed based on the proportion with benzodiazepine discontinuation in the control group set at 48%, a relative risk reduction (RRR) of 20%, a type I error of 2.5%, a type II error of 10% (90% power), and diversity of 36% as observed in the trials. The diversity‐adjusted required information size (DARIS) was 2109 participants, and the Trial Sequential Analysis‐adjusted confidence interval is 0.24 to 2.38. The blue line represents the cumulative Z‐score of the meta‐analysis. The green lines represent the conventional statistical boundaries of P = 5%. The cumulative Z‐curve (blue line) touches the conventional statistical boundaries, but does not cross the trial sequential monitoring boundaries, and the diversity‐adjusted required information size is not met, showing that insufficient information has been accrued.
Figuras y tablas -
Figure 4

Trial Sequential Analysis of comparison: 2 Carbamazepine versus placebo, outcome: 2.1 Benzodiazepine discontinuation. Trial Sequential Analysis on benzodiazepine discontinuation in three trials was performed based on the proportion with benzodiazepine discontinuation in the control group set at 48%, a relative risk reduction (RRR) of 20%, a type I error of 2.5%, a type II error of 10% (90% power), and diversity of 36% as observed in the trials. The diversity‐adjusted required information size (DARIS) was 2109 participants, and the Trial Sequential Analysis‐adjusted confidence interval is 0.24 to 2.38. The blue line represents the cumulative Z‐score of the meta‐analysis. The green lines represent the conventional statistical boundaries of P = 5%. The cumulative Z‐curve (blue line) touches the conventional statistical boundaries, but does not cross the trial sequential monitoring boundaries, and the diversity‐adjusted required information size is not met, showing that insufficient information has been accrued.

Trial Sequential Analysis of comparison: 6 Paroxetine versus placebo, outcome: 6.1 Benzodiazepine discontinuation. Trial Sequential Analysis on benzodiazepine discontinuation in three trials was performed based on the proportion with benzodiazepine discontinuation in the control group set at 48%, a relative risk reduction of 20%, a type I error of 2.5%, a type II error of 10% (90% power), and diversity of 86% as observed in the trials. The diversity‐adjusted required information size was 9448 participants, and the Trial Sequential Analysis‐adjusted confidence interval could not be estimated due to lack of information. The blue line represents the cumulative Z‐score of the meta‐analysis. The green lines represent the conventional statistical boundaries of P = 5%. The cumulative Z‐curve (blue line) does not cross the conventional statistical boundaries. The trial sequential monitoring boundaries and the diversity‐adjusted required information size are not shown as the accrued number of participants only amounted to 221/9448 (2.34%), showing that insufficient information has been accrued.
Figuras y tablas -
Figure 5

Trial Sequential Analysis of comparison: 6 Paroxetine versus placebo, outcome: 6.1 Benzodiazepine discontinuation. Trial Sequential Analysis on benzodiazepine discontinuation in three trials was performed based on the proportion with benzodiazepine discontinuation in the control group set at 48%, a relative risk reduction of 20%, a type I error of 2.5%, a type II error of 10% (90% power), and diversity of 86% as observed in the trials. The diversity‐adjusted required information size was 9448 participants, and the Trial Sequential Analysis‐adjusted confidence interval could not be estimated due to lack of information. The blue line represents the cumulative Z‐score of the meta‐analysis. The green lines represent the conventional statistical boundaries of P = 5%. The cumulative Z‐curve (blue line) does not cross the conventional statistical boundaries. The trial sequential monitoring boundaries and the diversity‐adjusted required information size are not shown as the accrued number of participants only amounted to 221/9448 (2.34%), showing that insufficient information has been accrued.

Trial Sequential Analysis of comparison: 6 Paroxetine versus placebo, outcome: 6.2 Benzodiazepine withdrawal symptoms Benzodiazepine Withdrawal Symptom Questionnaire (BWSQ). Trial Sequential Analysis on benzodiazepine withdrawal symptoms assessed with BWSQ assessing a minimal relevant clinical difference (MIREDIF) of 2.25 points, and a variance of 20 points (empirical data), was performed based on a type I error of 1.25%, a type II error of 10% (90% power), and diversity of 0%. The diversity‐adjusted required information size (DARIS) was 229 participants, and the Trial Sequential Analysis‐adjusted confidence interval is ‐7.18 to 0.05. The blue line represents the cumulative Z‐score of the meta‐analysis. The green lines represent the conventional statistical boundaries of P = 0.05. The red inward‐sloping lines represent the trial sequential monitoring boundaries. The cumulative Z‐curve touches the trial sequential monitoring boundaries, indicating that sufficient information was provided.
Figuras y tablas -
Figure 6

Trial Sequential Analysis of comparison: 6 Paroxetine versus placebo, outcome: 6.2 Benzodiazepine withdrawal symptoms Benzodiazepine Withdrawal Symptom Questionnaire (BWSQ). Trial Sequential Analysis on benzodiazepine withdrawal symptoms assessed with BWSQ assessing a minimal relevant clinical difference (MIREDIF) of 2.25 points, and a variance of 20 points (empirical data), was performed based on a type I error of 1.25%, a type II error of 10% (90% power), and diversity of 0%. The diversity‐adjusted required information size (DARIS) was 229 participants, and the Trial Sequential Analysis‐adjusted confidence interval is ‐7.18 to 0.05. The blue line represents the cumulative Z‐score of the meta‐analysis. The green lines represent the conventional statistical boundaries of P = 0.05. The red inward‐sloping lines represent the trial sequential monitoring boundaries. The cumulative Z‐curve touches the trial sequential monitoring boundaries, indicating that sufficient information was provided.

Trial Sequential Analysis of comparison: 6 Paroxetine versus placebo, outcome: 6.3 Anxiety, Hamilton Anxiety Rating Scale (HAM‐A). Trial Sequential Analysis on anxiety evaluated with HAM‐A assessing a minimal relevant clinical difference (MIREDIF) of 5 points, and a variance of 103 points, was performed based on a type I error of 1.25%, a type II error of 10% (90% power), and diversity of 0%. The diversity‐adjusted required information size (DARIS) was 236 participants, and the Trial Sequential Analysis‐adjusted confidence interval is ‐12.72 to ‐0.80. The blue line represents the cumulative Z‐score of the meta‐analysis. The green lines represent the conventional statistical boundaries of P = 0.05. The red inward‐sloping lines represent the trial sequential monitoring boundaries. The cumulative Z‐curve crosses the trial sequential monitoring boundaries, indicating that sufficient information was provided.
Figuras y tablas -
Figure 7

Trial Sequential Analysis of comparison: 6 Paroxetine versus placebo, outcome: 6.3 Anxiety, Hamilton Anxiety Rating Scale (HAM‐A). Trial Sequential Analysis on anxiety evaluated with HAM‐A assessing a minimal relevant clinical difference (MIREDIF) of 5 points, and a variance of 103 points, was performed based on a type I error of 1.25%, a type II error of 10% (90% power), and diversity of 0%. The diversity‐adjusted required information size (DARIS) was 236 participants, and the Trial Sequential Analysis‐adjusted confidence interval is ‐12.72 to ‐0.80. The blue line represents the cumulative Z‐score of the meta‐analysis. The green lines represent the conventional statistical boundaries of P = 0.05. The red inward‐sloping lines represent the trial sequential monitoring boundaries. The cumulative Z‐curve crosses the trial sequential monitoring boundaries, indicating that sufficient information was provided.

Trial Sequential Analysis of comparison: 7 Tricyclic antidepressants versus placebo, outcome: 7.1 Benzodiazepine discontinuation. Trial Sequential Analysis on benzodiazepine discontinuation in two trials was performed based on the proportion with benzodiazepine discontinuation in the control group set at 48%, a relative risk reduction (RRR) of 20%, a type I error of 2.5%, a type II error of 10% (90% power), and diversity of 0% as observed in the trials. The diversity‐adjusted required information size (DARIS) was 1343 participants, and the Trial Sequential Analysis‐adjusted confidence interval is 0.20 to 7.55. The blue line represents the cumulative Z‐score of the meta‐analysis. The green lines represent the conventional statistical boundaries of P = 5%. The cumulative Z‐curve (blue line) does not cross the conventional statistical boundaries or the trial sequential monitoring boundaries (red dotted lines), and the diversity‐adjusted required information size is not met, showing that insufficient information has been accrued.
Figuras y tablas -
Figure 8

Trial Sequential Analysis of comparison: 7 Tricyclic antidepressants versus placebo, outcome: 7.1 Benzodiazepine discontinuation. Trial Sequential Analysis on benzodiazepine discontinuation in two trials was performed based on the proportion with benzodiazepine discontinuation in the control group set at 48%, a relative risk reduction (RRR) of 20%, a type I error of 2.5%, a type II error of 10% (90% power), and diversity of 0% as observed in the trials. The diversity‐adjusted required information size (DARIS) was 1343 participants, and the Trial Sequential Analysis‐adjusted confidence interval is 0.20 to 7.55. The blue line represents the cumulative Z‐score of the meta‐analysis. The green lines represent the conventional statistical boundaries of P = 5%. The cumulative Z‐curve (blue line) does not cross the conventional statistical boundaries or the trial sequential monitoring boundaries (red dotted lines), and the diversity‐adjusted required information size is not met, showing that insufficient information has been accrued.

Trial Sequential Analysis of comparison: 8 Alpidem versus placebo, outcome: 8.3 Anxiety, Hamilton Anxiety Rating Scale (HAM‐A). Trial Sequential Analysis on anxiety evaluated with HAM‐A assessing a minimal relevant clinical difference (MIREDIF) of 5 points, and a variance of 103 points (empirical data), was performed based on a type I error of 1.25%, a type II error of 10% (90% power), and diversity of 0%. The diversity‐adjusted required information size (DARIS) was 235 participants, and the Trial Sequential Analysis‐adjusted confidence interval is ‐6.28 to 3.08. The blue line represents the cumulative Z‐score of the meta‐analysis. The green lines represent the conventional statistical boundaries of P = 0.05. The red inward‐sloping lines represent the trial sequential alpha‐spending monitoring boundaries, while the red outward‐sloping lines represent the beta‐spending (futility) boundaries. The cumulative Z‐curve crosses the beta‐spending (futility) boundaries, showing that an intervention effect, if any, is less than 5 points.
Figuras y tablas -
Figure 9

Trial Sequential Analysis of comparison: 8 Alpidem versus placebo, outcome: 8.3 Anxiety, Hamilton Anxiety Rating Scale (HAM‐A). Trial Sequential Analysis on anxiety evaluated with HAM‐A assessing a minimal relevant clinical difference (MIREDIF) of 5 points, and a variance of 103 points (empirical data), was performed based on a type I error of 1.25%, a type II error of 10% (90% power), and diversity of 0%. The diversity‐adjusted required information size (DARIS) was 235 participants, and the Trial Sequential Analysis‐adjusted confidence interval is ‐6.28 to 3.08. The blue line represents the cumulative Z‐score of the meta‐analysis. The green lines represent the conventional statistical boundaries of P = 0.05. The red inward‐sloping lines represent the trial sequential alpha‐spending monitoring boundaries, while the red outward‐sloping lines represent the beta‐spending (futility) boundaries. The cumulative Z‐curve crosses the beta‐spending (futility) boundaries, showing that an intervention effect, if any, is less than 5 points.

Trial Sequential Analysis of comparison: 9 Buspirone versus placebo, outcome: 9.1 Benzodiazepine discontinuation. Trial Sequential Analysis on benzodiazepine discontinuation in four trials was performed based on the proportion with benzodiazepine discontinuation in the control group set at 48%, a relative risk reduction of 20%, a type I error of 2.5%, a type II error of 10% (90% power), and diversity of 60% as observed in the trials. The diversity‐adjusted required information size was 3381 participants, and the Trial Sequential Analysis‐adjusted confidence interval could not be estimated due to lack of information. The blue line represents the cumulative Z‐score of the meta‐analysis. The green lines represent the conventional statistical boundaries of P = 5%. The cumulative Z‐curve (blue line) does not cross the conventional statistical boundaries. The trial sequential monitoring boundaries and the diversity‐adjusted required information size are not shown, as the accrued number of participants only amounted to 143/3381 (4.23%), showing that insufficient information has been accrued.
Figuras y tablas -
Figure 10

Trial Sequential Analysis of comparison: 9 Buspirone versus placebo, outcome: 9.1 Benzodiazepine discontinuation. Trial Sequential Analysis on benzodiazepine discontinuation in four trials was performed based on the proportion with benzodiazepine discontinuation in the control group set at 48%, a relative risk reduction of 20%, a type I error of 2.5%, a type II error of 10% (90% power), and diversity of 60% as observed in the trials. The diversity‐adjusted required information size was 3381 participants, and the Trial Sequential Analysis‐adjusted confidence interval could not be estimated due to lack of information. The blue line represents the cumulative Z‐score of the meta‐analysis. The green lines represent the conventional statistical boundaries of P = 5%. The cumulative Z‐curve (blue line) does not cross the conventional statistical boundaries. The trial sequential monitoring boundaries and the diversity‐adjusted required information size are not shown, as the accrued number of participants only amounted to 143/3381 (4.23%), showing that insufficient information has been accrued.

Trial Sequential Analysis of comparison: 10 Melatonin versus placebo, outcome: 10.1 Benzodiazepine discontinuation. Trial Sequential Analysis on benzodiazepine discontinuation in four trials was performed based on the proportion with benzodiazepine discontinuation in the control group set at 48%, a relative risk reduction (RRR) of 20%, a type I error of 2.5%, a type II error of 10% (90% power), and diversity of 61% as observed in the trials. The diversity‐adjusted required information size (DARIS) was 3438 participants, and the Trial Sequential Analysis‐adjusted confidence interval is 0.11 to 6.25. The blue line represents the cumulative Z‐score of the meta‐analysis. The green lines represent the conventional statistical boundaries of P = 5%. The cumulative Z‐curve (blue line) does not cross the conventional statistical boundaries or the trial sequential monitoring boundaries (red dotted lines), and the diversity‐adjusted required information size is not met, showing that insufficient information has been accrued.
Figuras y tablas -
Figure 11

Trial Sequential Analysis of comparison: 10 Melatonin versus placebo, outcome: 10.1 Benzodiazepine discontinuation. Trial Sequential Analysis on benzodiazepine discontinuation in four trials was performed based on the proportion with benzodiazepine discontinuation in the control group set at 48%, a relative risk reduction (RRR) of 20%, a type I error of 2.5%, a type II error of 10% (90% power), and diversity of 61% as observed in the trials. The diversity‐adjusted required information size (DARIS) was 3438 participants, and the Trial Sequential Analysis‐adjusted confidence interval is 0.11 to 6.25. The blue line represents the cumulative Z‐score of the meta‐analysis. The green lines represent the conventional statistical boundaries of P = 5%. The cumulative Z‐curve (blue line) does not cross the conventional statistical boundaries or the trial sequential monitoring boundaries (red dotted lines), and the diversity‐adjusted required information size is not met, showing that insufficient information has been accrued.

Comparison 1 Valproate versus placebo or no intervention, Outcome 1 Benzodiazepine discontinuation, end of intervention.
Figuras y tablas -
Analysis 1.1

Comparison 1 Valproate versus placebo or no intervention, Outcome 1 Benzodiazepine discontinuation, end of intervention.

Comparison 1 Valproate versus placebo or no intervention, Outcome 2 Relapse to benzodiazepine use, end of intervention.
Figuras y tablas -
Analysis 1.2

Comparison 1 Valproate versus placebo or no intervention, Outcome 2 Relapse to benzodiazepine use, end of intervention.

Comparison 1 Valproate versus placebo or no intervention, Outcome 3 Benzodiazepine discontinuation, longest follow‐up.
Figuras y tablas -
Analysis 1.3

Comparison 1 Valproate versus placebo or no intervention, Outcome 3 Benzodiazepine discontinuation, longest follow‐up.

Comparison 1 Valproate versus placebo or no intervention, Outcome 4 Relapse to benzodiazepine use, longest follow‐up.
Figuras y tablas -
Analysis 1.4

Comparison 1 Valproate versus placebo or no intervention, Outcome 4 Relapse to benzodiazepine use, longest follow‐up.

Comparison 1 Valproate versus placebo or no intervention, Outcome 5 Anxiety: HAM‐A (Hamilton Anxiety Rating Scale), end of intervention.
Figuras y tablas -
Analysis 1.5

Comparison 1 Valproate versus placebo or no intervention, Outcome 5 Anxiety: HAM‐A (Hamilton Anxiety Rating Scale), end of intervention.

Comparison 1 Valproate versus placebo or no intervention, Outcome 6 Benzodiazepine withdrawal symptoms, end of intervention.
Figuras y tablas -
Analysis 1.6

Comparison 1 Valproate versus placebo or no intervention, Outcome 6 Benzodiazepine withdrawal symptoms, end of intervention.

Comparison 1 Valproate versus placebo or no intervention, Outcome 7 Discontinuation due to adverse events.
Figuras y tablas -
Analysis 1.7

Comparison 1 Valproate versus placebo or no intervention, Outcome 7 Discontinuation due to adverse events.

Comparison 1 Valproate versus placebo or no intervention, Outcome 8 Serious adverse events.
Figuras y tablas -
Analysis 1.8

Comparison 1 Valproate versus placebo or no intervention, Outcome 8 Serious adverse events.

Comparison 2 Carbamazepine versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.
Figuras y tablas -
Analysis 2.1

Comparison 2 Carbamazepine versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.

Comparison 2 Carbamazepine versus placebo, Outcome 2 Benzodiazepine withdrawal symptoms.
Figuras y tablas -
Analysis 2.2

Comparison 2 Carbamazepine versus placebo, Outcome 2 Benzodiazepine withdrawal symptoms.

Comparison 2 Carbamazepine versus placebo, Outcome 3 Benzodiazepine discontinuation, longest follow‐up.
Figuras y tablas -
Analysis 2.3

Comparison 2 Carbamazepine versus placebo, Outcome 3 Benzodiazepine discontinuation, longest follow‐up.

Comparison 2 Carbamazepine versus placebo, Outcome 4 Relapse to benzodiazepine use.
Figuras y tablas -
Analysis 2.4

Comparison 2 Carbamazepine versus placebo, Outcome 4 Relapse to benzodiazepine use.

Comparison 2 Carbamazepine versus placebo, Outcome 5 Serious adverse events.
Figuras y tablas -
Analysis 2.5

Comparison 2 Carbamazepine versus placebo, Outcome 5 Serious adverse events.

Comparison 2 Carbamazepine versus placebo, Outcome 6 Non‐serious adverse events.
Figuras y tablas -
Analysis 2.6

Comparison 2 Carbamazepine versus placebo, Outcome 6 Non‐serious adverse events.

Comparison 2 Carbamazepine versus placebo, Outcome 7 Anxiety, HAM‐A.
Figuras y tablas -
Analysis 2.7

Comparison 2 Carbamazepine versus placebo, Outcome 7 Anxiety, HAM‐A.

Comparison 2 Carbamazepine versus placebo, Outcome 8 Discontinuation due to adverse events.
Figuras y tablas -
Analysis 2.8

Comparison 2 Carbamazepine versus placebo, Outcome 8 Discontinuation due to adverse events.

Comparison 3 Lithium versus placebo, Outcome 1 Benzodiazepine discontinuation.
Figuras y tablas -
Analysis 3.1

Comparison 3 Lithium versus placebo, Outcome 1 Benzodiazepine discontinuation.

Comparison 3 Lithium versus placebo, Outcome 2 Serious adverse events.
Figuras y tablas -
Analysis 3.2

Comparison 3 Lithium versus placebo, Outcome 2 Serious adverse events.

Comparison 3 Lithium versus placebo, Outcome 3 Non‐serious adverse events.
Figuras y tablas -
Analysis 3.3

Comparison 3 Lithium versus placebo, Outcome 3 Non‐serious adverse events.

Comparison 3 Lithium versus placebo, Outcome 4 Discontinuation due to adverse events.
Figuras y tablas -
Analysis 3.4

Comparison 3 Lithium versus placebo, Outcome 4 Discontinuation due to adverse events.

Comparison 4 Pregabalin versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.
Figuras y tablas -
Analysis 4.1

Comparison 4 Pregabalin versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.

Comparison 4 Pregabalin versus placebo, Outcome 2 Benzodiazepine withdrawal symptoms (Physician Withdrawal Checklist), end of intervention.
Figuras y tablas -
Analysis 4.2

Comparison 4 Pregabalin versus placebo, Outcome 2 Benzodiazepine withdrawal symptoms (Physician Withdrawal Checklist), end of intervention.

Comparison 4 Pregabalin versus placebo, Outcome 3 Anxiety, HAM‐A, end of intervention.
Figuras y tablas -
Analysis 4.3

Comparison 4 Pregabalin versus placebo, Outcome 3 Anxiety, HAM‐A, end of intervention.

Comparison 4 Pregabalin versus placebo, Outcome 4 Serious adverse events.
Figuras y tablas -
Analysis 4.4

Comparison 4 Pregabalin versus placebo, Outcome 4 Serious adverse events.

Comparison 4 Pregabalin versus placebo, Outcome 5 Non‐serious adverse events.
Figuras y tablas -
Analysis 4.5

Comparison 4 Pregabalin versus placebo, Outcome 5 Non‐serious adverse events.

Comparison 4 Pregabalin versus placebo, Outcome 6 Discontinuation due to adverse events.
Figuras y tablas -
Analysis 4.6

Comparison 4 Pregabalin versus placebo, Outcome 6 Discontinuation due to adverse events.

Comparison 5 Captodiame versus placebo, Outcome 1 Benzodiazepine withdrawal symptoms, BWSQ (Benzodiazepine Withdrawal Symptom Questionnaire), end of intervention.
Figuras y tablas -
Analysis 5.1

Comparison 5 Captodiame versus placebo, Outcome 1 Benzodiazepine withdrawal symptoms, BWSQ (Benzodiazepine Withdrawal Symptom Questionnaire), end of intervention.

Comparison 5 Captodiame versus placebo, Outcome 2 Anxiety, HAM‐A, end of intervention.
Figuras y tablas -
Analysis 5.2

Comparison 5 Captodiame versus placebo, Outcome 2 Anxiety, HAM‐A, end of intervention.

Comparison 5 Captodiame versus placebo, Outcome 3 Serious adverse events.
Figuras y tablas -
Analysis 5.3

Comparison 5 Captodiame versus placebo, Outcome 3 Serious adverse events.

Comparison 5 Captodiame versus placebo, Outcome 4 Non‐serious adverse events.
Figuras y tablas -
Analysis 5.4

Comparison 5 Captodiame versus placebo, Outcome 4 Non‐serious adverse events.

Comparison 6 Paroxetine versus placebo or no intervention, Outcome 1 Benzodiazepine discontinuation, end of intervention.
Figuras y tablas -
Analysis 6.1

Comparison 6 Paroxetine versus placebo or no intervention, Outcome 1 Benzodiazepine discontinuation, end of intervention.

Comparison 6 Paroxetine versus placebo or no intervention, Outcome 2 Benzodiazepine withdrawal symptoms: BWSQ, end of intervention.
Figuras y tablas -
Analysis 6.2

Comparison 6 Paroxetine versus placebo or no intervention, Outcome 2 Benzodiazepine withdrawal symptoms: BWSQ, end of intervention.

Comparison 6 Paroxetine versus placebo or no intervention, Outcome 3 Anxiety: HAM‐A, end of intervention.
Figuras y tablas -
Analysis 6.3

Comparison 6 Paroxetine versus placebo or no intervention, Outcome 3 Anxiety: HAM‐A, end of intervention.

Comparison 6 Paroxetine versus placebo or no intervention, Outcome 4 Benzodiazepine withdrawal symptoms: BWSQ, longest follow‐up: 6 months.
Figuras y tablas -
Analysis 6.4

Comparison 6 Paroxetine versus placebo or no intervention, Outcome 4 Benzodiazepine withdrawal symptoms: BWSQ, longest follow‐up: 6 months.

Comparison 6 Paroxetine versus placebo or no intervention, Outcome 5 Serious adverse events.
Figuras y tablas -
Analysis 6.5

Comparison 6 Paroxetine versus placebo or no intervention, Outcome 5 Serious adverse events.

Comparison 6 Paroxetine versus placebo or no intervention, Outcome 6 Non‐serious adverse events.
Figuras y tablas -
Analysis 6.6

Comparison 6 Paroxetine versus placebo or no intervention, Outcome 6 Non‐serious adverse events.

Comparison 7 Tricyclic antidepressants versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.
Figuras y tablas -
Analysis 7.1

Comparison 7 Tricyclic antidepressants versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.

Comparison 7 Tricyclic antidepressants versus placebo, Outcome 2 Anxiety: HAM‐A (change from baseline), end of intervention.
Figuras y tablas -
Analysis 7.2

Comparison 7 Tricyclic antidepressants versus placebo, Outcome 2 Anxiety: HAM‐A (change from baseline), end of intervention.

Comparison 7 Tricyclic antidepressants versus placebo, Outcome 3 Benzodiazepine discontinuation, longest follow‐up.
Figuras y tablas -
Analysis 7.3

Comparison 7 Tricyclic antidepressants versus placebo, Outcome 3 Benzodiazepine discontinuation, longest follow‐up.

Comparison 7 Tricyclic antidepressants versus placebo, Outcome 4 Benzodiazepine withdrawal symptoms (Physician Withdrawal Checklist), end of intervention.
Figuras y tablas -
Analysis 7.4

Comparison 7 Tricyclic antidepressants versus placebo, Outcome 4 Benzodiazepine withdrawal symptoms (Physician Withdrawal Checklist), end of intervention.

Comparison 7 Tricyclic antidepressants versus placebo, Outcome 5 Relapse to benzodiazepine use, end of intervention.
Figuras y tablas -
Analysis 7.5

Comparison 7 Tricyclic antidepressants versus placebo, Outcome 5 Relapse to benzodiazepine use, end of intervention.

Comparison 7 Tricyclic antidepressants versus placebo, Outcome 6 Discontinuation due to adverse events.
Figuras y tablas -
Analysis 7.6

Comparison 7 Tricyclic antidepressants versus placebo, Outcome 6 Discontinuation due to adverse events.

Comparison 8 Alpidem versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.
Figuras y tablas -
Analysis 8.1

Comparison 8 Alpidem versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.

Comparison 8 Alpidem versus placebo, Outcome 2 Withdrawal syndrome (clinical diagnosis), end of intervention.
Figuras y tablas -
Analysis 8.2

Comparison 8 Alpidem versus placebo, Outcome 2 Withdrawal syndrome (clinical diagnosis), end of intervention.

Comparison 8 Alpidem versus placebo, Outcome 3 Anxiety, HAM‐A, end of intervention.
Figuras y tablas -
Analysis 8.3

Comparison 8 Alpidem versus placebo, Outcome 3 Anxiety, HAM‐A, end of intervention.

Comparison 8 Alpidem versus placebo, Outcome 4 Relapse to benzodiazepine use, end of intervention.
Figuras y tablas -
Analysis 8.4

Comparison 8 Alpidem versus placebo, Outcome 4 Relapse to benzodiazepine use, end of intervention.

Comparison 8 Alpidem versus placebo, Outcome 5 Serious adverse events.
Figuras y tablas -
Analysis 8.5

Comparison 8 Alpidem versus placebo, Outcome 5 Serious adverse events.

Comparison 8 Alpidem versus placebo, Outcome 6 Discontinuation due to adverse events.
Figuras y tablas -
Analysis 8.6

Comparison 8 Alpidem versus placebo, Outcome 6 Discontinuation due to adverse events.

Comparison 9 Buspirone versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.
Figuras y tablas -
Analysis 9.1

Comparison 9 Buspirone versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.

Comparison 9 Buspirone versus placebo, Outcome 2 Anxiety: HAM‐A/Hospital Anxiety Depression Scale, end of intervention.
Figuras y tablas -
Analysis 9.2

Comparison 9 Buspirone versus placebo, Outcome 2 Anxiety: HAM‐A/Hospital Anxiety Depression Scale, end of intervention.

Comparison 9 Buspirone versus placebo, Outcome 3 Benzodiazepine withdrawal symptoms, end of intervention.
Figuras y tablas -
Analysis 9.3

Comparison 9 Buspirone versus placebo, Outcome 3 Benzodiazepine withdrawal symptoms, end of intervention.

Comparison 9 Buspirone versus placebo, Outcome 4 Benzodiazepine discontinuation, longest follow‐up.
Figuras y tablas -
Analysis 9.4

Comparison 9 Buspirone versus placebo, Outcome 4 Benzodiazepine discontinuation, longest follow‐up.

Comparison 9 Buspirone versus placebo, Outcome 5 Benzodiazepine withdrawal symptoms, longest follow‐up.
Figuras y tablas -
Analysis 9.5

Comparison 9 Buspirone versus placebo, Outcome 5 Benzodiazepine withdrawal symptoms, longest follow‐up.

Comparison 9 Buspirone versus placebo, Outcome 6 Anxiety, Hospital Anxiety Depression Scale, longest follow‐up.
Figuras y tablas -
Analysis 9.6

Comparison 9 Buspirone versus placebo, Outcome 6 Anxiety, Hospital Anxiety Depression Scale, longest follow‐up.

Comparison 9 Buspirone versus placebo, Outcome 7 Discontinuation due to adverse events.
Figuras y tablas -
Analysis 9.7

Comparison 9 Buspirone versus placebo, Outcome 7 Discontinuation due to adverse events.

Comparison 10 Melatonin versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.
Figuras y tablas -
Analysis 10.1

Comparison 10 Melatonin versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.

Comparison 10 Melatonin versus placebo, Outcome 2 Insomnia.
Figuras y tablas -
Analysis 10.2

Comparison 10 Melatonin versus placebo, Outcome 2 Insomnia.

Comparison 10 Melatonin versus placebo, Outcome 3 Discontinuation due to adverse events.
Figuras y tablas -
Analysis 10.3

Comparison 10 Melatonin versus placebo, Outcome 3 Discontinuation due to adverse events.

Comparison 10 Melatonin versus placebo, Outcome 4 Benzodiazepine discontinuation, longest follow‐up.
Figuras y tablas -
Analysis 10.4

Comparison 10 Melatonin versus placebo, Outcome 4 Benzodiazepine discontinuation, longest follow‐up.

Comparison 10 Melatonin versus placebo, Outcome 5 Adverse events.
Figuras y tablas -
Analysis 10.5

Comparison 10 Melatonin versus placebo, Outcome 5 Adverse events.

Comparison 10 Melatonin versus placebo, Outcome 6 Relapse to benzodiazepine use, longest follow‐up.
Figuras y tablas -
Analysis 10.6

Comparison 10 Melatonin versus placebo, Outcome 6 Relapse to benzodiazepine use, longest follow‐up.

Comparison 11 Flumazenil versus placebo, Outcome 1 Benzodiazepine withdrawal symptoms, end of intervention.
Figuras y tablas -
Analysis 11.1

Comparison 11 Flumazenil versus placebo, Outcome 1 Benzodiazepine withdrawal symptoms, end of intervention.

Comparison 11 Flumazenil versus placebo, Outcome 2 Anxiety, HAM‐D (Hamilton Depression Rating Scale), end of intervention.
Figuras y tablas -
Analysis 11.2

Comparison 11 Flumazenil versus placebo, Outcome 2 Anxiety, HAM‐D (Hamilton Depression Rating Scale), end of intervention.

Comparison 11 Flumazenil versus placebo, Outcome 3 Benzodiazepine mean dose, end of intervention.
Figuras y tablas -
Analysis 11.3

Comparison 11 Flumazenil versus placebo, Outcome 3 Benzodiazepine mean dose, end of intervention.

Comparison 12 Propranolol versus placebo, Outcome 1 Relapse to benzodiazepine use, end of intervention: 2 weeks.
Figuras y tablas -
Analysis 12.1

Comparison 12 Propranolol versus placebo, Outcome 1 Relapse to benzodiazepine use, end of intervention: 2 weeks.

Comparison 13 Progesterone versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.
Figuras y tablas -
Analysis 13.1

Comparison 13 Progesterone versus placebo, Outcome 1 Benzodiazepine discontinuation, end of intervention.

Comparison 13 Progesterone versus placebo, Outcome 2 Non‐serious adverse events.
Figuras y tablas -
Analysis 13.2

Comparison 13 Progesterone versus placebo, Outcome 2 Non‐serious adverse events.

Comparison 14 Magnesium aspartate versus placebo, Outcome 1 Benzodiazepine discontinuation.
Figuras y tablas -
Analysis 14.1

Comparison 14 Magnesium aspartate versus placebo, Outcome 1 Benzodiazepine discontinuation.

Comparison 14 Magnesium aspartate versus placebo, Outcome 2 Anxiety.
Figuras y tablas -
Analysis 14.2

Comparison 14 Magnesium aspartate versus placebo, Outcome 2 Anxiety.

Comparison 14 Magnesium aspartate versus placebo, Outcome 3 Relapse to benzodiazepine use.
Figuras y tablas -
Analysis 14.3

Comparison 14 Magnesium aspartate versus placebo, Outcome 3 Relapse to benzodiazepine use.

Comparison 14 Magnesium aspartate versus placebo, Outcome 4 Non‐serious adverse events.
Figuras y tablas -
Analysis 14.4

Comparison 14 Magnesium aspartate versus placebo, Outcome 4 Non‐serious adverse events.

Comparison 14 Magnesium aspartate versus placebo, Outcome 5 Discontinuation due to adverse events.
Figuras y tablas -
Analysis 14.5

Comparison 14 Magnesium aspartate versus placebo, Outcome 5 Discontinuation due to adverse events.

Comparison 15 Homéogène 46/Sedatif PC (homeopathic drugs) versus placebo, Outcome 1 Benzodiazepine discontinuation.
Figuras y tablas -
Analysis 15.1

Comparison 15 Homéogène 46/Sedatif PC (homeopathic drugs) versus placebo, Outcome 1 Benzodiazepine discontinuation.

Comparison 16 Carbamazepine versus tricyclic antidepressant, Outcome 1 Benzodiazepine discontinuation, end of intervention.
Figuras y tablas -
Analysis 16.1

Comparison 16 Carbamazepine versus tricyclic antidepressant, Outcome 1 Benzodiazepine discontinuation, end of intervention.

Comparison 16 Carbamazepine versus tricyclic antidepressant, Outcome 2 Relapse to benzodiazepine use.
Figuras y tablas -
Analysis 16.2

Comparison 16 Carbamazepine versus tricyclic antidepressant, Outcome 2 Relapse to benzodiazepine use.

Comparison 16 Carbamazepine versus tricyclic antidepressant, Outcome 3 Serious adverse events.
Figuras y tablas -
Analysis 16.3

Comparison 16 Carbamazepine versus tricyclic antidepressant, Outcome 3 Serious adverse events.

Comparison 17 Cyamemazine versus bromazepam, Outcome 1 Relapse to benzodiazepine use, longest follow‐up.
Figuras y tablas -
Analysis 17.1

Comparison 17 Cyamemazine versus bromazepam, Outcome 1 Relapse to benzodiazepine use, longest follow‐up.

Comparison 17 Cyamemazine versus bromazepam, Outcome 2 Anxiety: Maximum amplitude of rebound (HAM‐A), end of intervention.
Figuras y tablas -
Analysis 17.2

Comparison 17 Cyamemazine versus bromazepam, Outcome 2 Anxiety: Maximum amplitude of rebound (HAM‐A), end of intervention.

Comparison 17 Cyamemazine versus bromazepam, Outcome 3 Discontinuation due to adverse events.
Figuras y tablas -
Analysis 17.3

Comparison 17 Cyamemazine versus bromazepam, Outcome 3 Discontinuation due to adverse events.

Comparison 17 Cyamemazine versus bromazepam, Outcome 4 Non‐serious adverse events.
Figuras y tablas -
Analysis 17.4

Comparison 17 Cyamemazine versus bromazepam, Outcome 4 Non‐serious adverse events.

Comparison 18 Zopiclone versus flunitrazepam, Outcome 1 Relapse to benzodiazepine use, longest follow‐up.
Figuras y tablas -
Analysis 18.1

Comparison 18 Zopiclone versus flunitrazepam, Outcome 1 Relapse to benzodiazepine use, longest follow‐up.

Summary of findings for the main comparison. Valproate compared with placebo or no intervention for benzodiazepine discontinuation in chronic benzodiazepine users

Valproate compared with placebo or no intervention for benzodiazepine discontinuation in chronic benzodiazepine users

Patient or population: adults who withdraw from chronic benzodiazepine use
Settings: outpatients
Intervention: valproate
Comparison: placebo or no intervention

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 or no intervention

Valproate

Benzodiazepine discontinuation, end of intervention

Study population

RR 2.55
(1.08 to 6.03)

27
(1 study)

⊕⊝⊝⊝
very low1,2

The required information size of 1918 participants was not met.

679 per 1000

1000 per 1000
(142 to 1000)

Benzodiazepine discontinuation, longest follow‐up

Study population

RR 1.57
(0.80 to 3.09)

24
(1 study)

⊕⊝⊝⊝
very low1,2

500 per 1000

785 per 1000
(400 to 1000)

Benzodiazepine withdrawal symptoms, end of intervention

The mean benzodiazepine withdrawal symptoms in the intervention groups was
0.15 standard deviations lower
(0.68 lower to 0.37 higher).

56
(2 studies)

⊕⊝⊝⊝
very low3,4

SMD ‐0.15 (‐0.68 to 0.37).

As a rule of thumb, 0.2 represents a small effect, 0.5 a moderate effect, and 0.8 a large effect.

Benzodiazepine withdrawal symptoms, longest follow‐up

Not estimable

(0 study)

No included study measured this outcome.

*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: SMD: standardised mean difference

GRADE Working Group grades of evidence
High quality: We are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: 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 quality: Our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low quality: We have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1No details provided regarding random sequence generation, allocation concealment, and blinding, leading to unclear risk of selection bias, performance and detection bias (downgraded one level).
2Required information size not met (downgraded two levels due to serious imprecision: the sample size is far from the required one).
3Unclear risk of selection bias, attrition bias, reporting bias and high risk of performance bias (downgraded one level).
4Required information size not met (downgraded two levels due to serious imprecision).

Figuras y tablas -
Summary of findings for the main comparison. Valproate compared with placebo or no intervention for benzodiazepine discontinuation in chronic benzodiazepine users
Summary of findings 2. Carbamazepine compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users

Carbamazepine compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users

Patient or population: adults who withdraw from chronic benzodiazepine use
Settings: outpatients
Intervention: carbamazepine
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

Carbamazepine

Benzodiazepine discontinuation, end of intervention

Study population

RR 1.33
(0.99 to 1.8)

147
(3 studies)

⊕⊕⊝⊝
low1,2

Trial Sequential Analysis showed that only 7.0% of the required information size (2109) was reached, indicating that insufficient information has been obtained.

480 per 1000

638 per 1000
(475 to 864)

Benzodiazepine discontinuation, longest follow‐up

Study population

RR 1.41
(0.86 to 2.29)

40
(1 study)

⊕⊝⊝⊝
very low3,4

524 per 1000

739 per 1000
(450 to 1000)

Benzodiazepine withdrawal symptoms, end of intervention

The mean benzodiazepine withdrawal symptoms in the intervention groups was
1.14 standard deviations lower
(2.43 lower to 0.16 higher).

76
(2 studies)

⊕⊝⊝⊝
very low1,5,6

SMD ‐1.14 (‐2.43 to 0.16).

As a rule of thumb, 0.2 represents a small effect, 0.5 a moderate effect, and 0.8 a large effect.

Benzodiazepine withdrawal symptoms, longest follow‐up

Not estimable

(0 study)

No included study measured this outcome.

*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: SMD: standardised mean difference

GRADE Working Group grades of evidence
High quality: We are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: 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 quality: Our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low quality: We have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1Unclear risk of selection bias. One study with high risk of attrition, reporting, and other bias (downgraded one level).
2Required information size not met (downgraded one level due to imprecision).
3Unclear risk of selection and attrition bias (downgraded one level).
4Required information size not met, and 95% CI includes both no effect and appreciable benefit (downgraded two levels due to imprecision).
5Required information size not met (downgraded one level for imprecision).
6Significant heterogeneity (downgraded one level for inconsistency).

Figuras y tablas -
Summary of findings 2. Carbamazepine compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users
Summary of findings 3. Lithium compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users

Lithium compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users

Patient or population: adults who withdraw from chronic benzodiazepine use
Settings: outpatients
Intervention: lithium
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

Lithium

Benzodiazepine discontinuation, end of intervention

Study population

RR 1.05
(0.86 to 1.28)

230
(1 study)

⊕⊕⊝⊝
low1,2

The required information size of 1918 participants was not met.

617 per 1000

648 per 1000
(531 to 790)

Benzodiazepine discontinuation, longest follow‐up

Not estimable

(0 study)

No included studies measured this outcome.

Benzodiazepine withdrawal symptoms, end of intervention

Not estimable

(0 study)

No included studies measured this outcome.

Benzodiazepine withdrawal symptoms, longest follow‐up

Not estimable

(0 study)

No included study measured this outcome.

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

GRADE Working Group grades of evidence
High quality: We are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: 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 quality: Our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low quality: We have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1Unclear risk of selection, attrition, and reporting bias (downgraded one level).
2The required information size of 1918 participants was not met (downgraded one level due to imprecision).

Figuras y tablas -
Summary of findings 3. Lithium compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users
Summary of findings 4. Pregabalin compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users

Pregabalin compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users

Patient or population: adults who withdraw from chronic benzodiazepine use
Settings: outpatients
Intervention: pregabalin
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

Pregabalin

Benzodiazepine discontinuation, end of intervention

Study population

RR 1.44
(0.92 to 2.25)

106
(1 study)

⊕⊝⊝⊝
very low1,2

The required information size of 1918 participants was not met.

360 per 1000

518 per 1000
(331 to 810)

Benzodiazepine discontinuation, longest follow‐up

Not estimable

(0 study)

No included studies measured this outcome.

Benzodiazepine withdrawal symptoms, Physician Withdrawal Checklist (PWCL), end of intervention

The mean benzodiazepine withdrawal symptoms, PWCL, end of intervention in the intervention group was
3.10 lower
(3.51 to 2.69 lower).

106
(1 study)

⊕⊝⊝⊝
very low1,2

MD ‐3.10 (‐3.51 to ‐2.69)

Benzodiazepine withdrawal symptoms, longest follow‐up

Not estimable

(0 study)

No included study measured this outcome.

*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; MD: mean difference; RR: risk ratio

GRADE Working Group grades of evidence
High quality: We are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: 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 quality: Our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low quality: We have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1Unclear risk of selection bias and high risk of attrition and other bias (downgraded two levels).
2Required information size not met (downgraded one level due to imprecision).

Figuras y tablas -
Summary of findings 4. Pregabalin compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users
Summary of findings 5. Captodiame compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users

Captodiame compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users

Patient or population: adults who withdraw from chronic benzodiazepine use
Settings: outpatients
Intervention: captodiame
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

Captodiame

Benzodiazepine discontinuation, end of intervention

Not estimable

(0 study)

No included studies measured this outcome.

Benzodiazepine discontinuation, longest follow‐up

Not estimable

(0 study)

No included studies measured this outcome.

Benzodiazepine withdrawal symptoms, Benzodiazepine Withdrawal Symptom Questionnaire (BWSQ), end of intervention

The mean benzodiazepine withdrawal symptoms, BWSQ, end of intervention in the intervention group was
1.00 lower
(1.13 to 0.87 lower).

81
(1 study)

⊕⊝⊝⊝
very low1,2

MD ‐1.00 (‐1.13 to ‐0.87)

The required information size of 229 participants was not met.

Benzodiazepine withdrawal symptoms, longest follow‐up

Not estimable

(0 study)

No included study measured this outcome.

*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; MD: mean difference

GRADE Working Group grades of evidence
High quality: We are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: 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 quality: Our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low quality: We have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1Unclear risk of selection and reporting bias. High risk of other bias (downgraded one level).
2Required information size not met (downgraded two levels due to imprecision).

Figuras y tablas -
Summary of findings 5. Captodiame compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users
Summary of findings 6. Paroxetine compared with placebo or no intervention for benzodiazepine discontinuation in chronic benzodiazepine users

Paroxetine compared with placebo or no intervention for benzodiazepine discontinuation in chronic benzodiazepine users

Patient or population: adults who withdraw from chronic benzodiazepine use
Settings: outpatients
Intervention: paroxetine
Comparison: placebo or no intervention

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 or Control

Paroxetine

Benzodiazepine discontinuation, end of intervention

Study population

RR 1.45
(0.88 to 2.39)

221
(3 studies)

⊕⊝⊝⊝
very low1,2

Trial Sequential Analysis showed that only 2.34% of the required information size (9448) was reached, indicating that insufficient information has been obtained.

504 per 1000

731 per 1000
(444 to 1000)

Benzodiazepine discontinuation, longest follow‐up

Not estimable

(0 study)

No included study measured this outcome.

Benzodiazepine withdrawal symptoms, BWSQ, end of intervention

The mean benzodiazepine withdrawal symptoms, BWSQ, end of intervention in the intervention groups was
3.57 lower
(5.34 to 1.8 lower).

99
(2 studies)

⊕⊝⊝⊝
very low3,4

MD ‐3.57 (‐5.34 to ‐1.8). Trial Sequential Analysis showed that the required information size of 229 participants was not reached. However, the alpha‐spending boundaries for benefit were crossed, indicating that sufficient information was obtained, and the result was not due to random error.

Benzodiazepine withdrawal symptoms, BWSQ, longest follow‐up: 6 months

The mean benzodiazepine withdrawal symptoms, BWSQ, longest follow‐up: 6 months in the intervention group was
0.13 lower
(4.03 lower to 3.77 higher).

54
(1 study)

⊕⊝⊝⊝
very low5,6

MD ‐0.13 (‐4.03 to 3.77)

*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).
BWSQ: Benzodiazepine Withdrawal Symptom Questionnaire; CI: confidence interval; MD: mean difference; RR: risk ratio

GRADE Working Group grades of evidence
High quality: We are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: 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 quality: Our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low quality: We have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1Unclear risk of selection and attrition bias. High risk of performance, detection, reporting, and other bias (downgraded two levels).
2Required information size not met (downgraded one level due to imprecision).
3Unclear risk of selection bias. High risk of performance, detection, reporting, and other bias (downgraded two levels).
4The required information size was not met (downgraded one level due to imprecision).
5Unclear risk of selection bias. High risk of reporting and other bias (downgraded one level).
6Required information size not met (downgraded two levels due to imprecision).

Figuras y tablas -
Summary of findings 6. Paroxetine compared with placebo or no intervention for benzodiazepine discontinuation in chronic benzodiazepine users
Summary of findings 7. Tricyclic antidepressants compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users

Tricyclic antidepressants compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users

Patient or population: adults who withdraw from chronic benzodiazepine use
Settings: outpatients
Intervention: tricyclic 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

Tricyclic antidepressants

Benzodiazepine discontinuation, end of intervention

Study population

RR 0.82
(0.52 to 1.28)

105
(2 studies)

⊕⊝⊝⊝
very low1,2

Trial Sequential Analysis showed that only 7.82% of the required information size (1343) was reached, indicating that insufficient information has been obtained.

451 per 1000

370 per 1000
(235 to 577)

Benzodiazepine discontinuation, longest follow‐up

Study population

RR 2.2
(1.27 to 3.82)

47
(1 study)

⊕⊕⊝⊝
low3,4

375 per 1000

825 per 1000
(476 to 1000)

Benzodiazepine withdrawal symptoms, Physician Withdrawal Checklist, end of intervention

The mean benzodiazepine withdrawal symptoms in the intervention group was
19.78lower
(20.25 lower to 19.31 lower).

38

(1 study)

⊕⊝⊝⊝
very low4,5

MD ‐19.78 (‐20.25 to ‐19.31)

Benzodiazepine withdrawal symptoms, longest follow‐up

Not estimable

(0 study)

No included study measured this outcome.

*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; MD: mean difference; RR: risk ratio

GRADE Working Group grades of evidence
High quality: We are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: 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 quality: Our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low quality: We have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1Unclear risk of selection bias and high risk of attrition and other bias (downgraded one level).
2Required information size not met (downgraded one level due to imprecision).
3Unclear risk of selection and attrition bias (downgraded one level).
4Required information size not met (downgraded two levels due to imprecision).
5High risk of performance, detection, and reporting bias (downgraded two levels).

Figuras y tablas -
Summary of findings 7. Tricyclic antidepressants compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users
Summary of findings 8. Alpidem compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users

Alpidem compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users

Patient or population: adults who withdraw from chronic benzodiazepine use
Settings: outpatients
Intervention: alpidem
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

Alpidem

Benzodiazepine discontinuation, end of intervention

Study population

RR 0.41
(0.17 to 0.99)

25
(1 study)

⊕⊕⊝⊝
low1

The required information size of 1918 participants was not met.

750 per 1000

308 per 1000
(128 to 743)

Benzodiazepine discontinuation, longest follow‐up

Not estimable

(0 study)

No included study measured this outcome.

Withdrawal syndrome (clinical diagnosis), end of intervention

Study population

RR 4.86
(1.12 to 21.14)

145
(1 study)

⊕⊝⊝⊝
low2,3

29 per 1000

143 per 1000
(33 to 622)

Benzodiazepine withdrawal symptoms, longest follow‐up

Not estimable

(0 study)

No included study measured this outcome.

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

GRADE Working Group grades of evidence
High quality: We are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: 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 quality: Our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low quality: We have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1Required information size not met (downgraded two levels due to imprecision).
2Required information size not met (downgraded one level due to imprecision).

3Unclear risk of selection and other bias, high risk of attrition bias (downgraded one level)

Figuras y tablas -
Summary of findings 8. Alpidem compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users
Summary of findings 9. Buspirone compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users

Buspirone compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users

Patient or population: adults who withdraw from chronic benzodiazepine use
Settings: outpatients
Intervention: buspirone
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

Buspirone

Benzodiazepine discontinuation, end of intervention

Study population

RR 0.82
(0.49 to 1.37)

143
(4 studies)

⊕⊕⊝⊝
low1,2

Trial Sequential Analysis showed that only 4.23% of the required information size (3381) was reached, indicating that insufficient information has been obtained.

563 per 1000

462 per 1000
(276 to 772)

Benzodiazepine discontinuation, longest follow‐up

Study population

RR 0.60
(0.34 to 1.05)

23
(1 study)

⊕⊕⊝⊝
low2,3

917 per 1000

550 per 1000
(312 to 962)

Benzodiazepine withdrawal symptoms, end of intervention

The mean benzodiazepine withdrawal symptoms, end of intervention in the intervention groups was
4.69 higher
(14.47 lower to 23.85 higher).

17
(1 study)

⊕⊝⊝⊝
very low1,4

MD 4.69 (‐14.47 to 23.87)

Benzodiazepine withdrawal symptoms, longest follow‐up

The mean benzodiazepine withdrawal symptoms, longest follow‐up in the intervention groups was
1.34 lower
(14.31 lower to 11.63 higher).

15
(1 study)

⊕⊝⊝⊝
very low3,4

MD ‐1.34 (‐14.31 to 11.63)

*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; MD: mean difference; RR: risk ratio

GRADE Working Group grades of evidence
High quality: We are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: 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 quality: Our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low quality: We have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1Unclear risk of selection, performance, and reporting bias. High risk of attrition and other bias (downgraded one level).
2Required information size not met (downgraded one level due to imprecision).
3Unclear risk of selection and reporting bias. High risk of attrition bias (downgraded one level).
4Reguired information size not met (downgraded two levels due to serious imprecision).

Figuras y tablas -
Summary of findings 9. Buspirone compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users
Summary of findings 10. Melatonin compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users

Melatonin compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users

Patient or population: adults who withdraw from chronic benzodiazepine use
Settings: outpatients (3 studies), outpatients in methadone maintenance treatment (1 study)
Intervention: melatonin
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

Melatonin

Benzodiazepine discontinuation, end of intervention

Study population

RR 1.20
(0.73 to 1.96)

219
(4 studies)

⊕⊝⊝⊝
very low1,2

Trial Sequential Analysis showed that only 6.37% of the required information size (3438) was reached, indicating that insufficient information has been obtained.

417 per 1000

500 per 1000
(304 to 817)

Benzodiazepine discontinuation, longest follow‐up

Study population

RR 1.03
(0.47 to 2.27)

38
(1 study)

⊕⊝⊝⊝
very low2,3,4

389 per 1000

401 per 1000
(183 to 883)

Benzodiazpine withdrawal symptoms, end of intervention

Not estimable

(0 study)

No included study measured this outcome.

Benzodiazepine withdrawal symptoms, longest follow‐up

Not estimable

(0 study)

No included study measured this outcome.

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

GRADE Working Group grades of evidence
High quality: We are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: 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 quality: Our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low quality: We have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1Unclear risk of selection, attrition, and reporting bias. High risk of other bias (downgraded one level).
2Required information size not met, and the 95% CI includes both no effect and appreciable benefit (downgraded two levels due to imprecision).
3Unclear risk of selection and reporting bias (downgraded one level).
4Required information size not met (downgraded two levels due to imprecision).

Figuras y tablas -
Summary of findings 10. Melatonin compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users
Summary of findings 11. Flumazenil compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users

Flumazenil compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users

Patient or population: adults who withdraw from chronic benzodiazepine use
Settings: outpatients in methadone maintenance treatment (2 studies), outpatients (1 study)
Intervention: flumazenil
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

Flumazenil

Benzodiazepine discontinuation, end of intervention

Not estimable

(0 study)

No included study measured this outcome.

Benzodiazepine discontinuation, longest follow‐up

Not estimable

(0 study)

No included study measured this outcome.

Benzodiazepine withdrawal symptoms, end of intervention

The mean benzodiazepine withdrawal symptoms, end of intervention in the intervention groups was
0.95 standard deviations lower
(1.71 to 0.19 lower).

58
(3 studies)

⊕⊝⊝⊝
very low1,2

SMD ‐0.95 (‐1.71 to ‐0.19)

As a rule of thumb, 0.2 represents a small effect, 0.5 a moderate effect, and 0.8 a large effect.

Benzodiazepine withdrawal symptoms, longest follow‐up

Not estimable

(0 study)

No included study measured this outcome.

*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; SMD: standardised mean difference

GRADE Working Group grades of evidence
High quality: We are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: 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 quality: Our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low quality: We have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1Unclear risk of selection bias and high risk of performance, detection, and other bias (downgraded one level).
2Required information size not met (downgraded two levels due to imprecision).

Figuras y tablas -
Summary of findings 11. Flumazenil compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users
Summary of findings 12. Progesterone compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users

Progesterone compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users

Patient or population: adults who withdraw from chronic benzodiazepine use
Settings: outpatients
Intervention: progesterone
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

Progesterone

Benzodiazepine discontinuation, end of intervention

Study population

RR 1.15
(0.52 to 2.54)

35
(1 study)

⊕⊝⊝⊝
very low1,2

The required information size of 1918 participants was not met.

417 per 1000

479 per 1000
(217 to 1000)

Benzodiazepine discontinuation, longest follow‐up

Not estimable

(0 study)

No included study measured this outcome.

Benzodiazepine withdrawal symptoms, end of intervention

Not estimable

(0 study)

No included study measured this outcome.

Benzodiazepine withdrawal symptoms, longest follow‐up

Not estimable

(0 study)

No included study measured this outcome.

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

GRADE Working Group grades of evidence
High quality: We are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: 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 quality: Our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low quality: We have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1Unclear risk of selection and attrition bias (downgraded one level).
2Required information size not met, and the 95% CI includes both no effect and appreciable benefit (downgraded two levels due to imprecision).

Figuras y tablas -
Summary of findings 12. Progesterone compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users
Summary of findings 13. Magnesium aspartate compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users

Magnesium aspartate compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users

Patient or population: adults who withdraw from chronic benzodiazepine use
Settings: outpatients
Intervention: magnesium aspartate
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

Magnesium aspartate

Benzodiazepine discontinuation, end of intervention

Study population

RR 0.80
(0.66 to 0.96)

144
(1 study)

⊕⊝⊝⊝
very low1,2

The required information size of 1918 participants was not met.

853 per 1000

683 per 1000
(563 to 819)

Benzodiazepine discontinuation, longest follow‐up

Not estimable

(0 study)

No included study measured this outcome.

Benzodiazepine withdrawal symptoms, end of intervention

Not estimable

(0 study)

No included study measured this outcome.

Benzodiazepine withdrawal symptoms, longest follow‐up

Not estimable

(0 study)

No included study measured this outcome.

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

GRADE Working Group grades of evidence
High quality: We are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: 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 quality: Our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low quality: We have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1Unclear risk of selection, detection, and attrition bias (downgraded one level).
2Required information size not met (downgraded two levels due to imprecision).

Figuras y tablas -
Summary of findings 13. Magnesium aspartate compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users
Summary of findings 14. Homéogène 46/Sedatif PC (homeopathic drugs) compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users

Homéogène 46/Sedatif PC (homeopathic drugs) compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users

Patient or population: adults who withdraw from chronic benzodiazepine use
Settings: outpatients
Intervention: Homéogène 46/Sedatif PC (homeopathic drugs)
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

Homéogène 46/Sedatif PC (homeopathic drugs)

Benzodiazepine discontinuation, end of intervention

Study population

RR 0.79
(0.36 to 1.7)

51
(1 study)

⊕⊝⊝⊝
very low1,2

The required information size was not met.

381 per 1000

301 per 1000
(137 to 648)

Benzodiazepine discontinuation, longest follow‐up

Not estimable

(0 study)

No included study measured this outcome.

Benzodiazepine withdrawal symptoms, end of intervention

Not estimable

(0 study)

No included study measured this outcome.

Benzodiazepine withdrawal symptoms, longest follow‐up

Not estimable

(0 study)

No included study measured this outcome.

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

GRADE Working Group grades of evidence
High quality: We are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: 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 quality: Our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low quality: We have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1Unclear risk of selection, attrition, and other bias (downgraded one level).
2Required information size not met, and the 95% CI includes both no effect and appreciable benefit (downgraded two levels due to imprecision).

Figuras y tablas -
Summary of findings 14. Homéogène 46/Sedatif PC (homeopathic drugs) compared with placebo for benzodiazepine discontinuation in chronic benzodiazepine users
Summary of findings 15. Carbamazepine compared with tricyclic antidepressant for benzodiazepine discontinuation in chronic benzodiazepine users

Carbamazepine compared with tricyclic antidepressant for benzodiazepine discontinuation in chronic benzodiazepine users

Patient or population: adults who withdraw from chronic benzodiazepine use
Settings: outpatients
Intervention: carbamazepine
Comparison: tricyclic antidepressant

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No. of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Tricyclic antidepressant

Carbamazepine

Benzodiazepine discontinuation, end of intervention

Study population

RR 1.00
(0.78 to 1.29)

48
(1 study)

⊕⊕⊝⊝
low1,2

The required information size was not met.

833 per 1000

833 per 1000
(650 to 1000)

Benzodiazepine discontinuation, longest follow‐up

Not estimable

(0 study)

No included study measured this outcome.

Benzodiazepine withdrawal symptoms, end of intervention

Not estimable

(0 study)

No included study measured this outcome.

Benzodiazepine withdrawal symptoms, longest follow‐up

Not estimable

(0 study)

No included study measured this outcome.

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

GRADE Working Group grades of evidence
High quality: We are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: 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 quality: Our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low quality: We have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1Unclear risk of selection, detection, and attrition bias (downgraded one level).
2Required information size not met (downgraded one level due to imprecision).

Figuras y tablas -
Summary of findings 15. Carbamazepine compared with tricyclic antidepressant for benzodiazepine discontinuation in chronic benzodiazepine users
Comparison 1. Valproate versus placebo or no intervention

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation, end of intervention Show forest plot

1

27

Risk Ratio (M‐H, Random, 95% CI)

2.55 [1.08, 6.03]

2 Relapse to benzodiazepine use, end of intervention Show forest plot

1

27

Risk Ratio (M‐H, Random, 95% CI)

0.31 [0.11, 0.90]

3 Benzodiazepine discontinuation, longest follow‐up Show forest plot

1

24

Risk Ratio (M‐H, Random, 95% CI)

1.57 [0.80, 3.09]

4 Relapse to benzodiazepine use, longest follow‐up Show forest plot

1

24

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.13, 1.39]

5 Anxiety: HAM‐A (Hamilton Anxiety Rating Scale), end of intervention Show forest plot

1

27

Mean Difference (IV, Random, 95% CI)

‐0.40 [‐6.47, 5.67]

6 Benzodiazepine withdrawal symptoms, end of intervention Show forest plot

2

56

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

‐0.15 [‐0.68, 0.37]

6.1 Physician Withdrawal Checklist

1

27

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

‐0.01 [‐0.77, 0.74]

6.2 CIWA‐B (Clinical Institute Withdrawal Assessment Scale ‐ Benzodiazepines)

1

29

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

‐0.28 [‐1.01, 0.45]

7 Discontinuation due to adverse events Show forest plot

1

29

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

8 Serious adverse events Show forest plot

1

29

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 1. Valproate versus placebo or no intervention
Comparison 2. Carbamazepine versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation, end of intervention Show forest plot

3

147

Risk Ratio (M‐H, Random, 95% CI)

1.33 [0.99, 1.80]

2 Benzodiazepine withdrawal symptoms Show forest plot

2

76

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

‐1.14 [‐2.43, 0.16]

2.1 Physician Withdrawal Checklist

1

36

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

‐1.82 [‐2.61, ‐1.03]

2.2 Patient Withdrawal Checklist

1

40

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

‐0.50 [‐1.13, 0.13]

3 Benzodiazepine discontinuation, longest follow‐up Show forest plot

1

40

Risk Ratio (M‐H, Random, 95% CI)

1.41 [0.86, 2.29]

4 Relapse to benzodiazepine use Show forest plot

1

36

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.08, 1.44]

5 Serious adverse events Show forest plot

1

36

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

6 Non‐serious adverse events Show forest plot

1

36

Risk Ratio (M‐H, Random, 95% CI)

7.0 [0.39, 126.48]

7 Anxiety, HAM‐A Show forest plot

1

36

Mean Difference (IV, Random, 95% CI)

‐6.0 [‐9.58, ‐2.42]

8 Discontinuation due to adverse events Show forest plot

1

36

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 2. Carbamazepine versus placebo
Comparison 3. Lithium versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation Show forest plot

1

230

Risk Ratio (M‐H, Random, 95% CI)

1.05 [0.86, 1.28]

2 Serious adverse events Show forest plot

1

230

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3 Non‐serious adverse events Show forest plot

1

230

Risk Ratio (M‐H, Random, 95% CI)

1.06 [0.75, 1.49]

4 Discontinuation due to adverse events Show forest plot

1

230

Risk Ratio (M‐H, Random, 95% CI)

1.38 [0.13, 15.03]

Figuras y tablas -
Comparison 3. Lithium versus placebo
Comparison 4. Pregabalin versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation, end of intervention Show forest plot

1

106

Risk Ratio (M‐H, Random, 95% CI)

1.44 [0.92, 2.25]

2 Benzodiazepine withdrawal symptoms (Physician Withdrawal Checklist), end of intervention Show forest plot

1

106

Mean Difference (IV, Random, 95% CI)

‐3.1 [‐3.51, ‐2.69]

3 Anxiety, HAM‐A, end of intervention Show forest plot

1

106

Mean Difference (IV, Random, 95% CI)

‐4.8 [‐5.28, ‐4.32]

4 Serious adverse events Show forest plot

1

106

Risk Ratio (M‐H, Random, 95% CI)

0.67 [0.16, 2.85]

5 Non‐serious adverse events Show forest plot

1

106

Risk Ratio (M‐H, Random, 95% CI)

1.08 [0.84, 1.40]

6 Discontinuation due to adverse events Show forest plot

1

106

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.31, 2.59]

Figuras y tablas -
Comparison 4. Pregabalin versus placebo
Comparison 5. Captodiame versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine withdrawal symptoms, BWSQ (Benzodiazepine Withdrawal Symptom Questionnaire), end of intervention Show forest plot

1

81

Mean Difference (IV, Random, 95% CI)

1.00 [‐1.13, ‐0.87]

2 Anxiety, HAM‐A, end of intervention Show forest plot

1

81

Mean Difference (IV, Random, 95% CI)

‐5.7 [‐6.05, ‐5.35]

3 Serious adverse events Show forest plot

1

81

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

4 Non‐serious adverse events Show forest plot

1

81

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 5. Captodiame versus placebo
Comparison 6. Paroxetine versus placebo or no intervention

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation, end of intervention Show forest plot

3

221

Risk Ratio (M‐H, Random, 95% CI)

1.45 [0.88, 2.39]

2 Benzodiazepine withdrawal symptoms: BWSQ, end of intervention Show forest plot

2

99

Mean Difference (IV, Random, 95% CI)

‐3.57 [‐5.34, ‐1.80]

3 Anxiety: HAM‐A, end of intervention Show forest plot

2

99

Mean Difference (IV, Random, 95% CI)

‐6.75 [‐9.64, ‐3.86]

4 Benzodiazepine withdrawal symptoms: BWSQ, longest follow‐up: 6 months Show forest plot

1

54

Mean Difference (IV, Random, 95% CI)

‐0.13 [‐4.03, 3.77]

5 Serious adverse events Show forest plot

2

176

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

6 Non‐serious adverse events Show forest plot

1

54

Risk Ratio (M‐H, Random, 95% CI)

1.33 [0.35, 5.03]

Figuras y tablas -
Comparison 6. Paroxetine versus placebo or no intervention
Comparison 7. Tricyclic antidepressants versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation, end of intervention Show forest plot

2

105

Risk Ratio (M‐H, Random, 95% CI)

0.82 [0.52, 1.28]

2 Anxiety: HAM‐A (change from baseline), end of intervention Show forest plot

2

66

Mean Difference (IV, Random, 95% CI)

‐10.38 [‐25.96, 5.20]

3 Benzodiazepine discontinuation, longest follow‐up Show forest plot

1

47

Risk Ratio (M‐H, Random, 95% CI)

2.20 [1.27, 3.82]

4 Benzodiazepine withdrawal symptoms (Physician Withdrawal Checklist), end of intervention Show forest plot

1

38

Mean Difference (IV, Random, 95% CI)

‐19.78 [‐20.25, ‐19.31]

5 Relapse to benzodiazepine use, end of intervention Show forest plot

1

36

Risk Ratio (M‐H, Random, 95% CI)

2.0 [0.73, 5.47]

6 Discontinuation due to adverse events Show forest plot

2

134

Risk Ratio (M‐H, Random, 95% CI)

1.16 [0.42, 3.21]

Figuras y tablas -
Comparison 7. Tricyclic antidepressants versus placebo
Comparison 8. Alpidem versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation, end of intervention Show forest plot

1

25

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.17, 0.99]

2 Withdrawal syndrome (clinical diagnosis), end of intervention Show forest plot

1

145

Risk Ratio (M‐H, Random, 95% CI)

4.86 [1.12, 21.14]

3 Anxiety, HAM‐A, end of intervention Show forest plot

2

170

Mean Difference (IV, Random, 95% CI)

‐1.60 [‐4.64, 1.45]

4 Relapse to benzodiazepine use, end of intervention Show forest plot

1

145

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.09, 1.20]

5 Serious adverse events Show forest plot

1

25

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

6 Discontinuation due to adverse events Show forest plot

1

25

Risk Ratio (M‐H, Random, 95% CI)

0.46 [0.05, 4.46]

Figuras y tablas -
Comparison 8. Alpidem versus placebo
Comparison 9. Buspirone versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation, end of intervention Show forest plot

4

143

Risk Ratio (M‐H, Random, 95% CI)

0.82 [0.49, 1.37]

2 Anxiety: HAM‐A/Hospital Anxiety Depression Scale, end of intervention Show forest plot

2

41

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

0.18 [‐0.50, 0.86]

3 Benzodiazepine withdrawal symptoms, end of intervention Show forest plot

1

17

Mean Difference (IV, Random, 95% CI)

4.69 [‐14.47, 23.85]

4 Benzodiazepine discontinuation, longest follow‐up Show forest plot

1

23

Risk Ratio (M‐H, Random, 95% CI)

0.60 [0.34, 1.05]

5 Benzodiazepine withdrawal symptoms, longest follow‐up Show forest plot

1

15

Mean Difference (IV, Random, 95% CI)

‐1.34 [‐14.31, 11.63]

6 Anxiety, Hospital Anxiety Depression Scale, longest follow‐up Show forest plot

1

12

Mean Difference (IV, Random, 95% CI)

2.75 [‐2.83, 8.33]

7 Discontinuation due to adverse events Show forest plot

1

72

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.01, 7.92]

Figuras y tablas -
Comparison 9. Buspirone versus placebo
Comparison 10. Melatonin versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation, end of intervention Show forest plot

4

219

Risk Ratio (M‐H, Random, 95% CI)

1.20 [0.73, 1.96]

2 Insomnia Show forest plot

3

150

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

‐1.23 [‐2.70, 0.23]

2.1 PSQI (Pittsburgh Sleep Quality Index) global score (higher = worse), end of intervention

2

116

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

‐0.31 [‐0.92, 0.31]

2.2 Sleep quality (1 poorest, 10 excellent), end of intervention

1

34

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

‐3.34 [‐4.42, ‐2.26]

3 Discontinuation due to adverse events Show forest plot

2

120

Risk Ratio (M‐H, Random, 95% CI)

2.10 [0.20, 22.26]

4 Benzodiazepine discontinuation, longest follow‐up Show forest plot

1

38

Risk Ratio (M‐H, Random, 95% CI)

1.03 [0.47, 2.27]

5 Adverse events Show forest plot

1

86

Risk Ratio (M‐H, Random, 95% CI)

0.97 [0.52, 1.82]

6 Relapse to benzodiazepine use, longest follow‐up Show forest plot

1

38

Risk Ratio (M‐H, Random, 95% CI)

1.8 [0.37, 8.68]

Figuras y tablas -
Comparison 10. Melatonin versus placebo
Comparison 11. Flumazenil versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine withdrawal symptoms, end of intervention Show forest plot

3

58

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

‐0.95 [‐1.71, ‐0.19]

2 Anxiety, HAM‐D (Hamilton Depression Rating Scale), end of intervention Show forest plot

1

18

Mean Difference (IV, Random, 95% CI)

‐1.3 [‐2.28, ‐0.32]

3 Benzodiazepine mean dose, end of intervention Show forest plot

1

10

Mean Difference (IV, Random, 95% CI)

‐3.70 [‐22.06, 14.66]

Figuras y tablas -
Comparison 11. Flumazenil versus placebo
Comparison 12. Propranolol versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Relapse to benzodiazepine use, end of intervention: 2 weeks Show forest plot

1

40

Risk Ratio (M‐H, Random, 95% CI)

0.64 [0.31, 1.30]

Figuras y tablas -
Comparison 12. Propranolol versus placebo
Comparison 13. Progesterone versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation, end of intervention Show forest plot

1

35

Risk Ratio (M‐H, Random, 95% CI)

1.15 [0.52, 2.54]

2 Non‐serious adverse events Show forest plot

1

35

Risk Ratio (M‐H, Random, 95% CI)

3.13 [1.15, 8.54]

Figuras y tablas -
Comparison 13. Progesterone versus placebo
Comparison 14. Magnesium aspartate versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation Show forest plot

1

144

Risk Ratio (M‐H, Random, 95% CI)

0.80 [0.66, 0.96]

2 Anxiety Show forest plot

1

144

Mean Difference (IV, Random, 95% CI)

‐0.80 [‐2.73, 1.13]

3 Relapse to benzodiazepine use Show forest plot

1

144

Risk Ratio (M‐H, Random, 95% CI)

0.93 [0.46, 1.87]

4 Non‐serious adverse events Show forest plot

1

144

Risk Ratio (M‐H, Random, 95% CI)

0.49 [0.18, 1.35]

5 Discontinuation due to adverse events Show forest plot

1

144

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.13, 1.18]

Figuras y tablas -
Comparison 14. Magnesium aspartate versus placebo
Comparison 15. Homéogène 46/Sedatif PC (homeopathic drugs) versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation Show forest plot

1

51

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.36, 1.70]

Figuras y tablas -
Comparison 15. Homéogène 46/Sedatif PC (homeopathic drugs) versus placebo
Comparison 16. Carbamazepine versus tricyclic antidepressant

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Benzodiazepine discontinuation, end of intervention Show forest plot

1

48

Risk Ratio (M‐H, Random, 95% CI)

1.0 [0.78, 1.29]

2 Relapse to benzodiazepine use Show forest plot

1

48

Risk Ratio (M‐H, Random, 95% CI)

1.0 [0.28, 3.54]

3 Serious adverse events Show forest plot

1

48

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 16. Carbamazepine versus tricyclic antidepressant
Comparison 17. Cyamemazine versus bromazepam

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Relapse to benzodiazepine use, longest follow‐up Show forest plot

1

124

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.14, 0.78]

2 Anxiety: Maximum amplitude of rebound (HAM‐A), end of intervention Show forest plot

1

160

Mean Difference (IV, Random, 95% CI)

0.50 [‐1.23, 2.23]

3 Discontinuation due to adverse events Show forest plot

1

160

Risk Ratio (M‐H, Random, 95% CI)

2.87 [0.79, 10.44]

4 Non‐serious adverse events Show forest plot

1

160

Risk Ratio (M‐H, Random, 95% CI)

1.68 [1.01, 2.78]

Figuras y tablas -
Comparison 17. Cyamemazine versus bromazepam
Comparison 18. Zopiclone versus flunitrazepam

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Relapse to benzodiazepine use, longest follow‐up Show forest plot

1

18

Risk Ratio (M‐H, Random, 95% CI)

1.05 [0.23, 4.78]

Figuras y tablas -
Comparison 18. Zopiclone versus flunitrazepam