Scolaris Content Display Scolaris Content Display

Buprenorfina para el tratamiento de la abstinencia de opiáceos

Contraer todo Desplegar todo

Referencias

References to studies included in this review

Amass 1994 {published data only}

Amass L, Bickel WK, Higgins ST, Hughes JR. A preliminary investigation of outcome following gradual or rapid buprenorphine detoxification. Journal of Addictive Diseases 1994;13(3):33‐45. [MEDLINE: 95252233]CENTRAL

Assadi 2004 {published data only}

Assadi SM, Hafezi M, Mokri A, Razzaghi EM, Ghaeli P. Opioid detoxification using high doses of buprenorphine in 24 hours: a randomised, double‐blind, controlled clinical trial. Journal of Substance Abuse Treatment 2004;27(1):75‐82. CENTRAL

Bickel 1988 {published data only}

Bickel WK, Johnson RE, Stitzer ML, Bigelow GE, Liebson IA, Jasinski DR. A clinical trial of buprenorphine: I. Comparison with methadone in the detoxification of heroin addicts. II. Examination of its opioid blocking properties. NIDA Research Monograph 1987;76:182‐8. CENTRAL
Bickel WK, Stitzer ML, Bigelow GE, Liebson IA, Jasinski DR, Johnson RE. A clinical trial of buprenorphine: comparison with methadone in the detoxification of heroin addicts. Clinical Pharmacology & Therapeutics 1988;43(1):72‐8. [MEDLINE: 88081294]CENTRAL

Cheskin 1994 {published and unpublished data}

Cheskin LJ, Fudala PJ, Johnson RE. A controlled comparison of buprenorphine and clonidine for acute detoxification from opioids. Drug and Alcohol Dependence 1994;36(2):115‐21. [MEDLINE: 95154178]CENTRAL

Collins 2005 {published data only}

Collins ED, Kleber HD, Whittington RA, Heitler NE. Anesthesia‐assisted vs buprenorphine‐ or clonidine‐assisted heroin detoxification and naltrexone induction. A randomised trial. JAMA 2005;294(8):903‐13. CENTRAL
Collins ED, Whittington RA, Heitler NE, Kleber HD. A randomised comparison of anaesthesia‐assisted heroin detoxification with buprenorphine‐ and clonidine‐assisted detoxifications. Drug and Alcohol Dependence 2002;66(Suppl):S35. CENTRAL

Hopper 2005 {published data only}

Hopper JA, Wu J, Martus W, Pierre JD. A randomized trial of one‐day vs. three‐day buprenorphine inpatient detoxification protocols for heroin dependence. Journal of Opioid Management 2005;1(1):31‐5. CENTRAL

Hussain 2015 {published data only}

Hussain SS, Farhat S, Rather YH, Abbas Z. Comparative trial to study the effectiveness of clonidine hydrochloride and buprenorphine‐naloxone in opioid withdrawal: a hospital based study. Journal of Clinical and Diagnostic Research 2015;9(1):FC01‐FC4. CENTRAL

Janiri 1994 {published data only}

Janiri L, Mannelli P, Persico AM, Serretti A, Tempesta E. Opiate detoxification of methadone maintenance patients using lefetamine, clonidine and buprenorphine. Drug and Alcohol Dependence 1994;36(2):139‐45. [MEDLINE: 95154181]CENTRAL
Janiri L, Mannelli P, Serretti A, Tempesta E. Low‐dose buprenorphine detoxification in long term methadone addicts. Regulatory Peptides 1994;Suppl(1):289‐90. CENTRAL

Ling 2005 {published data only}

Amass L, Ling W, Freese TE, Reiber C, Annon JJ, Cohen AJ, et al. Bringing buprenorphine‐naloxone detoxification to community treatment providers: the NIDA Clinical Trials Network Field Experience. American Journal on Addictions 2004;13(Suppl):S42‐66. CENTRAL
Hillhouse M, Domier CP, Chim D, Ling W. Provision of ancillary medications during buprenorphine detoxification does not improve treatment outcomes. Journal of Addictive Diseases 2010;29(1):23‐9. CENTRAL
Ling W, Amass L, Shoptaw S, Annon JJ, Hillhouse M, et al. A multi‐centre randomised trial of buprenorphine‐naloxone versus clonidine for opioid detoxification: findings from the National Institute on Drug Abuse Clinical Trials Network. Addiction 2005;100(8):1090‐100. CENTRAL
Ziedonis DM, Amass L, Steinberg M, Woody G, Krejci J, Annon JJ, et al. Predictors of outcome for short‐term medically supervised opioid withdrawal during a randomized, multicenter trial of buprenorphine‐naloxone and clonidine in the NIDA clinical trials network drug and alcohol dependence. Drug and Alcohol Dependence 2009;99(1‐3):28‐36. CENTRAL

Ling 2009 {published data only}

Brown ES, Tirado C, Minhajuddin A, Hillhouse M, Adinoff B, Ling W, et al. Association of race and ethnicity with withdrawal symptoms, attrition, opioid use, and side‐effects during buprenorphine therapy. Journal of Ethnicity in Substance Abuse 2010;9(2):106‐14. CENTRAL
Hillhouse M, Canamar CP, Doraimani G, Thomas C, Hasson A, Ling W. Participant characteristics and buprenorphine dose. American Journal of Drug and Alcohol Abuse 2011;37(5):453‐9. CENTRAL
Hillhouse M, Canamar CP, Ling W. Predictors of outcome after short‐term stabilization with buprenorphine. Journal of Substance Abuse Treatment 2013;44(3):336‐42. CENTRAL
Ling W, Hillhouse M, Domier C, Doraimani G, Hunter J, Thomas C, et al. Buprenorphine tapering schedule and illicit opioid use. Addiction 2009;104(2):256‐65. CENTRAL
Nielsen S, Hillhouse M, Thomas C, Hasson A, Ling W. A comparison of buprenorphine taper outcomes between prescription opioid and heroin users. Journal of Addiction Medicine 2013;7(1):33‐8. CENTRAL
Saleh MI. Modeling longitudinal changes in buprenorphine treatment outcome for opioid dependence. Pharmacopsychiatry 2014;47(7):251‐8. CENTRAL
Saleh MI. Predictors of long term opioid withdrawal outcome after short‐term stabilization with buprenorphine. European Review for Medical and Pharmacological Sciences 2014;18(24):3935‐42. CENTRAL

Lintzeris 2002 {published data only}

Lintzeris N, Bell J, Bammer G, Jolley DJ, Rushworth L. A randomised controlled trial of buprenorphine in the management of short‐term ambulatory heroin withdrawal. Addiction 2002;97(11):1395‐404. CENTRAL

Marsch 2005 {published data only}

Marsch LA, Bickel WK, Badger GJ, Stothart ME, Quesnel KJ, Stanger C, et al. Comparison of pharmacological treatments for opioid‐dependent adolescents: a randomized controlled trial. Archives of General Psychiatry 2005;62(10):1157‐64. CENTRAL
Moore SK, Marsch LA, Badger GJ, Solhkhah R, Hofstein Y. Improvement in psychopathology among opioid‐dependent adolescents during behavioral‐pharmacological treatment. Journal of Addiction Medicine 2011;5(4):264‐71. CENTRAL
Motamed M, Marsch LA, Solhkhah R, Bickel WK, Badger GJ. Differences in treatment outcomes between prescription opioid‐dependent and heroin‐dependent adolescents. Journal of Addiction Medicine 2008;2(3):158‐64. CENTRAL

Marsch 2016 {published data only}

Marsch LA, Moore SK, Borodovsky JT, Solhkhah R, Badger GJ, et al. A randomized controlled trial of buprenorphine taper duration among opioid‐dependent adolescents and young adults. Addiction 2016;111(8):1406‐15. [DOI: 10.1111]CENTRAL

Nigam 1993 {published and unpublished data}

Nigam AK, Ray R, Tripathi BM. Buprenorphine in opiate withdrawal: a comparison with clonidine. Journal of Substance Abuse Treatment 1993;10(4):391‐4. [MEDLINE: 94016726]CENTRAL

O'Connor 1997 {published data only}

O'Connor PG, Carroll KM, Shi JM, Schottenfeld RS, Kosten TR, Rounsaville BJ. Three methods of opioid detoxification in a primary care setting. A randomized trial. Annals of Internal Medicine 1997;127(7):526‐30. [MEDLINE: 97443112]CENTRAL
Shi JM, O'Connor PG, Constantino JA, Carroll KM, Schottenfeld RS, Rounsaville BJ. Three methods of ambulatory opiate detoxification: preliminary results of a randomised clinical trial. NIDA Research Monograph 1993;132:309. CENTRAL

Oreskovich 2005 {published data only}

Oreskovich MR, Saxon AJ, Ellis ML, Malte CA, Reoux JP, Knox PC. A double‐blind, double‐dummy, randomized, prospective pilot study of the partial Mu opiate agonist, buprenorphine, for acute detoxification from heroin. Drug and Alcohol Dependence 2005;77(1):71‐9. [Embase 2004536231]CENTRAL

Petitjean 2002 {published and unpublished data}

Petitjean S, von Bardeleben U, Weber M, Ladewig D. Buprenorphine versus methadone in opiate detoxification: preliminary results. Drug and Alcohol Dependence 2002;66(Suppl):S138. CENTRAL

Ponizovsky 2006 {published data only}

Ponizovsky AM, Grinshpoon A, Margolis A, Cohen R, Rosca P. Well‐being, psychosocial factors, and side‐effects among heroin‐dependent inpatients after detoxification using buprenorphine versus clonidine. Addictive Behaviors 2006;31(11):2002‐13. CENTRAL

Raistrick 2005 {published data only}

Raistrick D, West D, Finnegan O, Thistlewaite G, Brearley R, Banbery J. A comparison of buprenorphine and lofexidine for community opiate detoxification: results from a randomized controlled trial. Addiction 2005;100(12):1860‐7. CENTRAL

Schneider 2000 {published data only}

Paetzold W, Eronat V, Seifert J, Holze I, Emrich HM, Schneider U. Detoxification of poly‐substance abusers with buprenorphine. Effects on affect, anxiety, and withdrawal symptoms [Detoxifikation polytoxikomaner Patienten mit Buprenorphin. Auswirkungen auf Affektivitat, Angst und Entzugssymptomaik]. Nervenarzt 2000;71(9):722‐9. [MEDLINE: 20497356]CENTRAL
Schneider U, Paetzold W, Eronat V, Huber TJ, Seifert J, Wiese B, et al. Buprenorphine and carbamazepine as a treatment for detoxification of opiate addicts with multiple drug misuse: a pilot study. Addiction Biology 2000;5:65‐9. CENTRAL

Seifert 2002 {published data only}

Seifert J, Metzner C, Paetzold W, Borsutzky M, Ohlmeier M, Passie T, et al. Mood and affect during detoxification of opiate addicts: a comparison of buprenorphine versus methadone. Addiction Biology 2005;10(2):157‐64. CENTRAL
Seifert J, Metzner C, Paetzold W, Borsutzky M, Passie T, Rollnik J, et al. Detoxification of opiate addicts with multiple drug abuse: a comparison of buprenorphine vs. methadone. Pharmacopsychiatry 2002;35(5):159‐64. [MEDLINE: 12237786]CENTRAL

Sigmon 2013 {published data only}

Dunn KE, Saulsgiver KA, Miller ME, Nuzzo PA, Sigmon SC. Characterizing opioid withdrawal during double‐blind buprenorphine detoxification. Drug and Alcohol Dependence 2015;151:47‐55. [DOI: 10.1016/j.drugalcdep.2015.02.033]CENTRAL
Patrick ME, Dunn KE, Badger GJ, Heil SH, Higgins ST, Sigmon SC. Spontaneous reductions in smoking during double‐blind buprenorphine detoxification. Addictive Behaviors 2014;39(9):1353‐6. CENTRAL
Sigmon SC, Dunn KE, Saulsgiver K, Patrick ME, Badger GJ, Heil SH, et al. A randomized, double‐blind evaluation of buprenorphine taper duration in primary prescription opioid abusers. JAMA Psychiatry 2013;70(12):1347‐54. CENTRAL

Steinmann 2008 {published data only}

Steinmann C, Artmann S, Henneberg B, Paul HW. Should methadone or buprenorphine be preferred for opiate detoxification? [Methadon‐racemat versus buprenorphin zur stationaren entgiftungsbehandlung opiatabhangiger]. Psychiatrische Praxis 2007;34(1):S103‐5. CENTRAL
Steinmann C, Artmann S, Schachtschneider A, Paul HW. Methadone versus buprenorphine for inpatient detoxification treatment [Methadon‐Racemat versus Buprenorphin zur stationaren Entgiftungsbehandlung]. Sucht 2008;54(4):217‐21. CENTRAL

Umbricht 2003 {published data only}

Umbricht A, Hoover DR, Tucker MJ, Leslie JM, Chaisson RE, Preston KL. Opioid detoxification with buprenorphine, clonidine, or methadone in hospitalized heroin‐dependent patients with HIV infection. Drug and Alcohol Dependence 2003;69(3):263‐72. CENTRAL

Wang 1996 {published data only}

Wang RI, Young LD. Double‐blind controlled detoxification from buprenorphine. NIDA Research Monograph 1996;162:114. CENTRAL

Wright 2011 {published data only}

Sheard L, Wright NMJ, Adams CE, Bound N, Rushforth B, Hart R, et al. The Leeds Evaluation of Efficacy of Detoxification Study (LEEDS) Prisons Project Study: protocol for a randomised controlled trial comparing methadone and buprenorphine for opiate detoxification. Trials 2009;10:53. CENTRAL
Wright NMJ, Sheard L, Adams CE, Rushforth BJ, Harrison W, Bound N, et al. Comparison of methadone and buprenorphine for opiate detoxification (LEEDS trial): a randomised controlled trial. British Journal of General Practice 2011;61(593):e772‐80. CENTRAL

Ziaaddini 2012 {published data only}

Ziaaddini H, Nasirian M, Nakhaee N. A comparison of the efficacy of buprenorphine and clonidine in detoxification of heroin‐dependents and the following maintenance treatment. Addiction and Health 2010;2(1‐2):18‐24. CENTRAL
Ziaaddini H, Nasirian M, Nakhaee N. Comparison of the efficacy of buprenorphine and clonidine in detoxification of opioid‐dependents. Addiction and Health 2012;4(3‐4):79‐86. CENTRAL

References to studies excluded from this review

Blondell 2008 {published data only}

Blondell RD, Frydrych L, Arber BC, Bashaw HL, Vexler A, Purdy CH, et al. A randomized trial of extended buprenorphine detoxification for opioid dependency. Journal of Addiction Medicine 2008;2(3):139‐46. CENTRAL

Breen 2003 {published data only}

Breen CL, Harris SJ, Lintzeris N, Mattick RP, Hawken L, Bell J, et al. Cessation of methadone maintenance treatment using buprenorphine: transfer from methadone to buprenorphine and subsequent buprenorphine reductions. Drug and Alcohol Dependence 2003;71(1):49‐55. CENTRAL

Fiellin 2014 {published data only}

Fiellin D, Cutter CJ, Moore BA, Barry D, O'Connor P, Schottenfeld RS. Primary care buprenorphine detoxification vs. maintenance for prescription opioid dependence. Drug and Alcohol Dependence 2015;146(12):e277. CENTRAL
Fiellin DA, Schottenfeld RS, Cutter CJ, Moore BA, Barry DT, O'Connor PG. Primary care‐based buprenorphine taper vs maintenance therapy for prescription opioid dependence: a randomized clinical trial. JAMA Internal Medicine 2014;174(12):1947‐54. CENTRAL

Fingerhood 2001 {published data only}

Fingerhood MI, Thompson MR, Jasinski DR. A comparison of clonidine and buprenorphine in the outpatient treatment of opiate withdrawal. Substance Abuse 2001;22(3):193‐9. CENTRAL

Frownfelter 2001 {published data only}

Frownfelter J, Hopper JA. Rapid opioid detoxification using buprenorphine and naltrexone. Drug and Alcohol Dependence 2001;63(Suppl 1):S50. CENTRAL

Gandhi 2003 {published data only}

Gandhi DH, Jaffe JH, McNary S, Kavanagh GJ, Hayes M, Currens M. Short‐term outcomes after brief ambulatory opioid detoxification with buprenorphine in young heroin users. Addiction 2003;98(4):453‐62. [MEDLINE: 12653815]CENTRAL

Gibson 2003 {published data only}

Doran CM, Shanahan M, Bell J, Gibson A. A cost‐effectiveness analysis of buprenorphine‐assisted heroin withdrawal. Drug and Alcohol Review 2004;23(2):171‐5. CENTRAL
Gibson AE, Doran CM, Bell JR, Ryan A, Lintzeris N. A comparison of buprenorphine treatment in clinic and primary care settings: A randomised trial. Medical Journal of Australia 2003;179(1):38‐42. CENTRAL

Gonzalez 2015 {published data only}

Gonzalez G, DiGirolamo G, Kolodziej ME, Smelson D, Romero‐Gonzalez M. Memantine improves buprenorphine treatment for opioid‐dependent young adults. Drug and Alcohol Dependence 2015;146:e124. CENTRAL
Gonzalez G, DiGirolamo G, Kolodziej ME, Smelson D, Romero‐Gonzalez M. Memantine improves buprenorphine treatment for opioid‐dependent young adults. Drug and Alcohol Dependence 2015;156:243‐253. CENTRAL

Hao 2000 {published data only}

Hao W, Zhao M. A comparative clinical study of the effect of WeiniCom, a Chinese herbal compound, on alleviation of withdrawal symptoms and craving for heroin in detoxification treatment. Journal of Psychoactive Drugs 2000;32(3):277‐84. [MEDLINE: 20513856]CENTRAL

Harris 2003 {published data only}

Harris L. A comparison of buprenorphine with lofexidine in the rapid opiate detoxification of patients with opiate dependency receiving treatment in a community setting. Addiction Biology 2003;8:243. CENTRAL

He 1999 {published data only}

He M, Zhang H, Tang J, Tang Z, Yang D, Chen X, et al. A comparative study of buprenorphine plus naloxone vs opium tincture in the detoxification treatment of heroin dependence. Chinese Journal of Clinical Psychology 1999;4(1):10‐5. CENTRAL

Jain 2011 {published data only}

Jain K, Jain R, Dhawan A. A double‐blind, double‐dummy, randomized controlled study of memantine versus buprenorphine in naloxone‐precipitated acute withdrawal in heroin addicts. Journal of Opioid Management 2011;7(1):11‐20. CENTRAL

Jia 2001 {published data only}

Jia Y. Effect of methadone plus buprenorphine on the alleviation of heroin withdrawal symptoms. Chinese Journal of Clinical Psychology 2001;9(1):60‐2. CENTRAL

Johnson 1989 {published data only}

Johnson RE, Fudala PJ, Collins CC, Jaffe JH. Outpatient maintenance/detoxification comparison of methadone and buprenorphine. NIDA Research Monograph 1989;95:384. [MEDLINE: 90348833]CENTRAL

Johnson 1992a {published data only}

Johnson RE, Fudala PJ, Cheskin LR. A comparison of buprenorphine to clonidine for rapid inpatient opiate detoxification. Clinical Pharmacology & Therapeutics 1992;51(2):167. CENTRAL

Johnson 1992b {published data only}

Johnson RE, Jaffe JH, Fudala PJ. A controlled trial of buprenorphine treatment for opioid dependence. JAMA 1992;267(20):2750‐5. CENTRAL

Johnson 1996 {published data only}

Johnson RE, Eissenberg T, Alim TN, Lindquist T, Vocci FJ, Risher‐Flowers DL, et al. Comparison of the withdrawal syndromes following abrupt termination of buprenorphine and methadone. NIDA Research Monograph 1996;162:200. CENTRAL

Katz 2009 {published data only}

Katz EC, Schwartz RP, King S, Highfield DA, O'Grady KE, Billings T, et al. Brief vs extended buprenorphine detoxification in a community treatment program: engagement and short‐term outcomes. American Journal of Drug and Alcohol Abuse 2009;35(2):63‐7. CENTRAL

Kosten 1988 {published data only}

Kosten TR, Kleber HD. Buprenorphine detoxification from opioid dependence: a pilot study. Life Sciences 1988;42(6):635‐41. [MEDLINE: 88121171]CENTRAL

Kosten 1991 {published data only}

Kosten TR, Krystal JH, Charney DS, Price LH, Morgan CH, Kleber HD. Opioid antagonist challenges in buprenorphine maintained patients. Drug and Alcohol Dependence 1990;25(1):73‐8. [MEDLINE: 90214491]CENTRAL
Kosten TR, Krystal JH, Charney DS, Price LH, Morgan CH, Kleber HD. Rapid detoxification from opioid dependence. American Journal of Psychiatry 1989;146(10):1349. [MEDLINE: 89390747]CENTRAL
Kosten TR, Morgan C, Kleber HD. Phase II clinical trials of buprenorphine: detoxification and induction onto naltrexone. NIDA Research Monograph 1992;121:101‐19. [MEDLINE: 93024845]CENTRAL
Kosten TR, Morgan C, Kleber HD. Treatment of heroin addicts using buprenorphine. American Journal of Drug and Alcohol Abuse 1991;17(2):119‐28. [MEDLINE: 91320874]CENTRAL

Law 2002 {published data only}

Law FD, Myles JS, Nutt DJ. BD RCT of Suboxone (buprenorphine/naloxone) vs. methadone/lofexidine for stabilization and withdrawal of low‐dose outpatient opiate addicts. Drug and Alcohol Dependence 2002;66(Suppl 1):S100. CENTRAL
Myles J, Law F, Raybould T. A double‐blind randomised controlled trial of buprenorphine/naloxone (suboxone) versus methadone/lofexidine for the detoxification of opiate‐dependent addicts. Drug and Alcohol Dependence 2000;60(Suppl 1):S156. CENTRAL

Liebschutz 2014 {published data only}

Cushman PA, Anderson BJ, Moreau M, Stein MD, Liebschutz JM. Buprenorphine initiation and linkage to outpatient buprenorphine treatment does not reduce frequency of injection drug use for inpatient opioid‐dependent injection drug users: results of a randomized clinical trial. Journal of General Internal Medicine 2015;30:S114‐S5. CENTRAL
Liebschutz J, Crooks D, Herman D, Anderson B, Meshesha L, Dossabhoy S, et al. Initiating buprenorphine maintenance for opiate‐dependent hospitalized patients: a randomized controlled trial. Journal of General Internal Medicine 2013;28:S108‐9. CENTRAL
Liebschutz JM, Crooks D, Herman D, Anderson B, Tsui J, Meshesha LZ, et al. Buprenorphine treatment for hospitalized, opioid‐dependent patients: a randomized clinical trial. JAMA Internal Medicine 2014;174(8):1369‐76. CENTRAL

Lintzeris 2003 {published data only}

Lintzeris N, Bammer G, Rushworth L, Jolley DJ, Whelan G. Buprenorphine dosing regime for inpatient heroin withdrawal: a symptom‐triggered dose titration study. Drug and Alcohol Dependence 2003;70(3):287‐94. CENTRAL

Liu 1997 {published data only}

Liu ZM, Cai ZJ, Wang XP, Ge Y, Li CM. Rapid detoxification of heroin dependence by buprenorphine. Acta Pharmacologica Sinica 1997;18(2):112‐4. CENTRAL

Maayan 2008 {published data only}

Maayan R, Touati‐Werner D, Shamir D, Yadid G, Friedman A, Eisner D, et al. The effect of DHEA complementary treatment on heroin addicts participating in a rehabilitation program: a preliminary study. European Neuropsychopharmacology 2008;18(6):406‐13. CENTRAL

Meade 2010 {published data only}

Meade CS, Lukas SE, McDonald LJ, Fitzmaurice GM, Eldridge JA, Merrill N, et al. A randomized trial of transcutaneous electric acupoint stimulation as adjunctive treatment for opioid detoxification. Journal of Substance Abuse Treatment 2010;38(1):12‐21. CENTRAL

Montoya 1994 {published data only}

Montoya ID, Mann DJ, Ellison PA, Lange WR, Preston KL. Inpatient medically supervised opioid withdrawal, with buprenorphine alone and in combination with naltrexone. Clinical Pharmacology and Therapeutics 1994;55(2):131. CENTRAL

Pjrek 2012 {published data only}

Pjrek E, Frey R, Naderi‐Heiden A, Strnad A, Kowarik A, Kasper S, et al. Actigraphic measurements in opioid detoxification with methadone or buprenorphine. Journal of Clinical Psychopharmacology 2012;32(1):75‐82. CENTRAL

Pycha 1994 {published and unpublished data}

Pycha C, Resnick RB, Galanter M. Buprenorphine: rapid and slow dose‐reductions for heroin detoxification. NIDA Research Monograph 1994;141:453. CENTRAL

Reed 2007 {published data only}

Reed LJ, Glasper A, de Wet CJ, Bearn J, Gossop M. Comparison of buprenorphine and methadone in the treatment of opiate withdrawal: possible advantages of buprenorphine for the treatment of opiate‐benzodiazepine codependent patients?. Journal of Clinical Psychopharmacology 2007;27(2):188‐92. CENTRAL

Resnick 1992 {published data only}

Resnick RB, Galanter M, Pycha C, Cohen A, Grandison P, Flood N. Buprenorphine: an alternative to methadone for heroin dependence treatment. Psychopharmacology Bulletin 1992;28(1):109‐13. [MEDLINE: 92302380]CENTRAL

Rosen 1995 {published data only}

Rosen M, Kosten TR. Detoxification and induction onto naltrexone. In: Cowan A, Lewis JW editor(s). Buprenorphine: Combatting Drug Abuse with a Unique Opioid. New York: Wiley‐Liss, 1995. CENTRAL

Rosenthal 2013 {published data only}

Rosenthal RN, Ling W, Casadonte P, Vocci F, Bailey GL, Kampman K, et al. Buprenorphine implants for treatment of opioid dependence: randomized comparison to placebo and sublingual buprenorphine/naloxone. Addiction 2013;108(12):2141‐9. CENTRAL

Sanders 2013 {published data only}

Sanders NC, Mancino MJ, Gentry WB, Guise JB, Bickel WK, Thostenson J, et al. Randomized, placebo‐controlled pilot trial of gabapentin during an outpatient, buprenorphine‐assisted detoxification procedure. Experimental and Clinical Psychopharmacology 2013;21(4):294‐302. CENTRAL

Sheard 2009 {published data only}

Sheard L, Adams CE, Wright NMJ, El‐Sayeh H, Dalton R, Tompkins CNE. The Leeds Evaluation of Efficacy of Detoxification Study (LEEDS) prisons project pilot study: protocol for a randomised controlled trial comparing dihydrocodeine and buprenorphine for opiate detoxification. Trials 2007;8:1. CENTRAL
Sheard L, Wright NMJ, El‐Sayeh HG, Adams CE, Li R, Tompkins CNE. The Leeds Evaluation of Efficacy of Detoxification Study (LEEDS) prisons project: a randomised controlled trial comparing dihydrocodeine and buprenorphine for opiate detoxification. Substance Abuse Treatment, Prevention, and Policy 2009;4:1. CENTRAL

Sigmon 2004 {published data only}

Sigmon SC, Wong CJ, Chausmer AL, Liebson IA, Bigelow GE. Evaluation of an injection depot formulation of buprenorphine: placebo comparison. Addiction 2004;99(11):1439‐49. CENTRAL

Sullivan 2015 {published data only}

Sullivan MA, Bisaga A, Carpenter K, Mariani JJ, Mishlen K, Nunes EV. Long‐acting injectable naltrexone induction: a randomized trial of outpatient opioid detoxification with naltrexone vs buprenorphine. Drug and Alcohol Dependence 2015;146:e112. CENTRAL

Tompkins 2014 {published data only}

Tompkins D, Smith MT, Mintzer MZ, Campbell CM, Strain EC. A double blind, within subject comparison of spontaneous opioid withdrawal from buprenorphine versus morphine. Journal of Pharmacology and Experimental Therapeutics 2014;348(2):217‐26. CENTRAL

Umbricht 1999 {published data only}

Umbricht A, Montoya ID, Hoover DR, Demuth KL, Chiang CT, Preston KL. Naltrexone shortened opioid detoxification with buprenorphine. Drug and Alcohol Dependence 1999;56(3):181‐90. CENTRAL

Weiss 2010 {published data only}

Weiss RD, Potter JS, Fiellin DA, Byrne M, Connery HS, Dickinson W, et al. Adjunctive counseling during brief and extended buprenorphine‐naloxone treatment for prescription opioid dependence: a 2‐phase randomized controlled trial. Archives of General Psychiatry 2011;68(12):1238‐46. CENTRAL
Weiss RD, Potter JS, Provost SE, Huang Z, Jacobs P, Hasson A, et al. A multi‐site, two‐phase, Prescription Opioid Addiction Treatment Study (POATS): Rationale, design, and methodology. Contemporary Clinical Trials 2010;31(2):189‐99. CENTRAL

White 2001 {published data only}

White R, Alcorn R, Feinmann C. Two methods of community detoxification from opiates: an open‐label comparison of lofexidine and buprenorphine. Drug and Alcohol Dependence 2001;65(1):77‐83. [MEDLINE: 11714592]CENTRAL

Williams 2002 {published data only}

Williams H, Remedios A, Oyefeso A, Bennett J. Buprenorphine detoxification treatment for heroin dependence: a preliminary experience in an outpatient setting. Irish Journal of Psychological Medicine 2002;19(3):80‐3. CENTRAL

Woody 2008 {published data only}

Hill KP, Bennett HE, Griffin ML, Connery HS, Fitzmaurice GM, Subramaniam G, et al. Association of cannabis use with opioid outcomes among opioid‐dependent youth. Drug and Alcohol Dependence 2013;132(1‐2):342‐5. CENTRAL
Meade CS, Weiss RD, Fitzmaurice GM, Poole SA, Subramaniam GA, Patkar AA, et al. HIV risk behavior in treatment‐seeking opioid‐dependent youth: results from a NIDA clinical trials network multisite study. Journal of Acquired Immune Deficiency Syndromes 2010;55(1):65‐72. CENTRAL
Polsky D, Glick HA, Yang J, Subramaniam GA, Poole SA, Woody GE. Cost‐effectiveness of extended buprenorphine‐naloxone treatment for opioid‐dependent youth: data from a randomized trial. Addiction 2010;105(9):1616‐24. CENTRAL
Subramaniam GA, Warden D, Minhajuddin A, Fishman MJ, Stitzer ML, Adinoff B, et al. Predictors of abstinence: National Institute of Drug Abuse multisite buprenorphine/naloxone treatment trial in opioid‐dependent youth. Journal of the American Academy of Child and Adolescent Psychiatry 2011;50(11):1120‐8. CENTRAL
Warden D, Subramaniam GA, Carmody T, Woody GE, Minhajuddin A, Poole SA, et al. Predictors of attrition with buprenorphine/naloxone treatment in opioid dependent youth. Addictive Behaviors 2012;37(9):1046‐53. CENTRAL
Wilcox CE, Bogenschutz MP, Nakazawa M, Woody GE. Compensation effects on clinical trial data collection in opioid‐dependent young adults. American Journal of Drug and Alcohol Abuse 2012;38(1):81‐6. CENTRAL
Woody GE, Poole SA, Subramaniam G, Dugosh K, Bogenschutz M, Abbott P, et al. Extended vs short‐term buprenorphine‐naloxone for treatment of opioid‐addicted youth. JAMA 2008;300(17):2003‐11. CENTRAL

Wright 2007 {published data only}

Oldham NS, Wright NM, Adams CE, Sheard L, Tompkins CN. The Leeds Evaluation of Efficacy of Detoxification Study (LEEDS) project: an open‐label pragmatic randomised control trial comparing the efficacy of differing therapeutic agents for primary care detoxification from either street heroin or methadone [ISRCTN07752728]. BMC Family Practice 2004;5(1):9. CENTRAL
Wright NM, Sheard L, Tompkins CN, Adams CE, Allgar VL, Oldham NS. Buprenorphine versus dihydrocodeine for opiate detoxification in primary care: a randomised controlled trial. BMC Family Practice 2007;8:3. CENTRAL

Zhang 1993 {published data only}

Zhang R, Li J, Wan W. Comparative study of buprenorphine and methadone in the treatment of heroin addicts. Chinese Journal of Neurological and Mental Diseases 1993;19(5):295‐96. CENTRAL

Amato 2011

Amato L, Minozzi S, Davoli M, Vecchi S, Ferri M, Mayet S. Psychosocial and pharmacological treatments versus pharmacological treatments for opioid detoxification. Cochrane Database of Systematic Reviews 2011, Issue 9. [DOI: 10.1002/14651858.CD005031.pub2]

Amato 2013

Amato L, Davoli M, Ferri M, Ali R. Methadone at tapered doses for the management of opioid withdrawal. Cochrane Database of Systematic Reviews 2013, Issue 2. [DOI: 10.1002/14651858.CD003409.pub4]

Ang‐Lee 2006

Ang‐Lee K, Oreskovich MR, Saxon AJ, Jaffe C, Meredith C, Ellis ML, et al. Single dose of 24 milligrams of buprenorphine for heroin detoxification: an open‐label study of five inpatients. Journal of Psychoactive Drugs 2006;38(4):505‐12. CENTRAL

Atkins 2004

Atkins D, Best D, Briss PA, Eccles M, Falck‐Ytter Y, Flottorp S, et al. GRADE working group. Grading quality of evidence and strength of recommendations. BMJ 2004;328(7454):1490‐4.

Banys 1994

Banys P, Clark HW, Tusel DJ, Sees K, Stewart P, Mongan L, et al. An open trial of low dose buprenorphine in treating methadone withdrawal. Journal of Substance Abuse Treatment 1994;11(1):9‐15. [MEDLINE: 94260563]CENTRAL

Broers 2000

Broers B, Giner F, Dumont P, Mino A. Inpatient opiate detoxification in Geneva: follow‐up at 1 and 6 months. Drug and Alcohol Dependence 2000;58(1):85‐92.

Cameron 2001

Cameron D, Allen D, Galway K. A pilot study of the effectiveness of buprenorphine and methadone as detoxication agents when choice is given to the consumer. Journal of Substance Use 2001;6(2):101‐9. CENTRAL

Chawarski 2005

Chawarski MC, Moody DE, Pakes J, O'Connor PG, Schottenfeld RS. Buprenorphine tablet versus liquid: a clinical trial comparing plasma levels, efficacy and symptoms. Journal of Substance Abuse Treatment 2005;29:307‐12.

Chiang 2003

Chiang CN, Hawks RL. Pharmacokinetics of the combination tablet of buprenorphine and naloxone. Drug and Alcohol Dependence 2003;70(Suppl):S39‐S47.

Day 2005

Day E, Ison J, Strang J. Inpatient versus other settings for detoxification for opioid dependence. Cochrane Database of Systematic Reviews 2005, Issue 2. [DOI: 10.1002/14651858.CD004580.pub2]

Elkader 2005

Elkader A, Sproule B. Buprenorphine: clinical pharmacokinetics in the treatment of opioid dependence. Clinical Pharmacokinetics 2005;44(7):661‐80.

Farrell 1994

Farrell M. Opiate withdrawal. Addiction 1994;89(11):1471‐5.

Gossop 1988

Gossop M. Clonidine and the treatment of the opiate withdrawal syndrome. Drug and Alcohol Dependence 1988;21(3):253‐9.

Gossop 1989a

Gossop M, Green L, Phillips G, Bradley B. Lapse, relapse and survival among opiate addicts after treatment. A prospective follow‐up study. British Journal of Psychiatry 1989;154:348‐53.

Gossop 1989b

Gossop M, Griffiths P, Bradley B, Strang J. Opiate withdrawal symptoms in response to 10 day and 21 day methadone withdrawal programmes. British Journal of Psychiatry 1989;154:360‐3. [MEDLINE: 90089752]

Gowing 2009

Gowing L, Ali R, White J. Opioid antagonists with minimal sedation for the management of opioid withdrawal. Cochrane Database of Systematic Reviews 2009, Issue 4. [DOI: 10.1002/14651858.CD002021.pub3]

Gowing 2010

Gowing LR, Ali RL, White JM. Opioid antagonists under heavy sedation or anaesthesia for opioid withdrawal. Cochrane Database of Systematic Reviews 2010, Issue 2. [DOI: 10.1002/14651858.CD002022.pub3]

Gowing 2014

Gowing L, Ali R, Dunlop A, Farrell M, Lintzeris N. National Guidelines for Medication‐Assisted Treatment of Opioid Dependence. Commonwealth of Australia, 2014. www.nationaldrugstrategy.gov.au/internet/drugstrategy/Publishing.nsf/content/ng‐mat‐op‐dep accessed prior to 18 January 2017. [Online ISBN: 978‐1‐74241‐945‐9]

Gowing 2015

Gowing LR, Ali RL, Allsop S, Marsden J, Turf EE, West R, et al. Global statistics on addictive behaviours: 2014 status report. Addiction 2015;110(6):904‐19. [DOI: 10.1111/add.12899]

Gowing 2016

Gowing LR, Farrell M, Ali RL, White JM. Alpha2‐adrenergic agonists for the management of opioid withdrawal. Cochrane Database of Systematic Reviews 2016, Issue 5. [DOI: 10.1002/14651858.CD002024.pub5]

GRADEpro GDT [Computer program]

GRADE Working Group, McMaster University. GRADEpro GDT. Version accessed prior to 17 January 2017. Hamilton (ON): GRADE Working Group, McMaster University, 2014.

Guyatt 2008a

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

Guyatt 2008b

Guyatt GH, Oxman AD, Kunz R, Vist GE, Falck‐Ytter Y, Schünemann HJ, GRADE Working Group. Rating quality of evidence and strength of recommendations: What is "quality of evidence" and why is it important to clinicians?. BMJ 2008;336(7651):955‐8.

Guyatt 2011

Guyatt G, Oxman AD, Akl EA, Kunz R, Vist G, Brozek J, et al GRADE working group. GRADE guidelines 1. Introduction‐GRADE evidence profiles and summary of findings tables. Journal of Clinical Epidemiology 2011;64(4):383‐94.

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 www.cochrane‐handbook.org.

Jaffe 1997

Jaffe JH, Knapp CM, Ciraulo DA. Opiates: Clinical Aspects. In: Lowinson JH, Ruiz P, Millman RB, Langrod JG editor(s). Substance Abuse: A Comprehensive Textbook. 3rd Edition. Baltimore: Williams & Wilkins, 1997.

Johnson 2003

Johnson RE, Strain EC, Amass L. Buprenorphine: how to use it right. Drug and Alcohol Dependence 2003;70(Suppl 2):S59‐S77.

Kleber 1982

Kleber HD, Riordan CE. The treatment of narcotic withdrawal: a historical review. Journal of Clinical Psychiatry 1982;43(6):30‐4.

Kutz 2002

Kutz I, Reznik V. Heroin detoxification with a single high dose of buprenorphine. Israel Journal of Psychiatry and Related Sciences 2002;39(2):113‐9. CENTRAL

Lanier 2007

Lanier RK, Umbricht A, Harrison JA, Nuwayser ES, Bigelow GE. Evaluation of a transdermal buprenorphine formulation in opioid detoxification. Addiction 2007;102(10):1648‐56. CENTRAL

Lanier 2008

Lanier RK, Umbricht A, Harrison JA, Nuwayser ES, Bigelow GE. Opioid detoxification via single 7‐day application of a buprenorphine transdermal patch: an open‐label evaluation. Psychopharmacology (Berlin) 2008;198(2):149‐58. CENTRAL

Larance 2011

Larance B, Degenhardt L, Lintzeris N, Bell J, Winstock A, Dietze P, et al. Post‐marketing surveillance of buprenorphine‐naloxone in Australia: diversion, injection and adherence with supervised dosing. Drug and Alcohol Dependence 2011;118(2‐3):265‐73.

Lintzeris 2013

Lintzeris N, Leung SY, Dunlop AJ, Larance B, White N, Rivas GR, et al. A randomised controlled trial of sublingual buprenorphine‐naloxone film versus tablets in the management of opioid dependence. Drug and Alcohol Dependence 2013;131(1‐2):119‐26.

Lipton 1983

Lipton D, Maranda M. Detoxification from heroin dependency: an overview of method and effectiveness. Advances in Alcohol and Substance Abuse 1983;2(1):31‐55.

Mark 2001

Mark TL, Woody GE, Juday T, Kleber HD. The economic costs of heroin addiction in the United States. Drug and Alcohol Dependence 2001;61(2):195‐206. [MEDLINE: 20578850]

Mattick 1996

Mattick RP, Hall W. Are detoxification programmes effective?. Lancet 1996;347(8994):97‐100.

Mattick 2014

Mattick RP, Breen C, Kimber J, Davoli M. Buprenorphine maintenance versus placebo or methadone maintenance for opioid dependence. Cochrane Database of Systematic Reviews 2014, Issue 2. [DOI: 10.1002/14651858.CD002207.pub4]

Minozzi 2014

Minozzi S, Amato L, Davoli M. Detoxification treatments for opiate dependent adolescents. Cochrane Database of Systematic Reviews 2014, Issue 4. [DOI: 10.1002/14651858.CD006749.pub3]

Phillips 1986

Phillips GT, Gossop M, Bradley B. The influence of psychological factors on the opiate withdrawal syndrome. British Journal of Psychiatry 1986;149:235‐8. [MEDLINE: 87050435]

Preston 1985

Preston KL, Bigelow GE. Pharmacological advances in addiction treatment. International Journal of the Addictions 1985;20(6‐7):845‐67.

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.

Satel 1993

Satel SL, Kosten TR, Schuckit MA, Fischman MW. Should protracted withdrawal from drugs be included in DSM‐IV?. American Journal of Psychiatry 1993;150(5):695‐704.

Sobel 2004

Sobel B‐FX, Sigmon SC, Walsh SL, Johnson RE, Liebson IA, Nuwayser ES, et al. Open‐label trial of an injection depot formulation of buprenorphine in opioid detoxification. Drug and Alcohol Dependence 2004;73(1):11‐22. CENTRAL

Strain 2004

Strain EC, Moody DE, Stoller KB, Walsh SL, Bigelow GE. Relative bioavailability of different buprenorphine formulations under chronic dosing conditions. Drug and Alcohol Dependence 2004;74:37‐43.

Strang 1999

Strang J, Griffiths P, Powis B, Gossop M. Heroin chasers and heroin injectors: differences observed in a community sample in London, UK. American Journal on Addictions 1999;8(2):148‐60. [MEDLINE: 99293529]

Tetrault 2009

Tetrault JM, O'Connor PG. Management of opioid intoxication and withdrawal. In: Ries RK, Fiellin DA, Miller SC, Siatz R editor(s). Principles of Addiction Medicine. 4th Edition. Philadelphia: Wolters Kluwer/Lippincott Williams & Wilkins, 2009:589‐606.

Vaillant 1988

Vaillant GE. What can long‐term follow‐up teach us about relapse and prevention of relapse in addiction?. British Journal of Addiction 1988;83(10):1147‐57.

WHO 2009

World Health Organization. Guidelines for the Psychosocially Assisted Pharmacological Treatment of Opioid Dependence. Geneva: World Health Organization, 2009. [ISBN 978 92 4 154754 3]

References to other published versions of this review

Gowing 2004

Gowing L, Ali R, White JM. Buprenorphine for the management of opioid withdrawal. Cochrane Database of Systematic Reviews 2004, Issue 4. [DOI: 10.1002/14651858.CD002025.pub4]

Gowing 2006

Gowing L, Ali R, White JM. Buprenorphine for the management of opioid withdrawal. Cochrane Database of Systematic Reviews 2006, Issue 2. [DOI: 10.1002/14651858.CD002025.pub4]

Gowing 2009a

Gowing L, Ali R, White JM, Mbewe D. Buprenorphine for the management of opioid withdrawal. Cochrane Database of Systematic Reviews 2009, Issue 3. [DOI: 10.1002/14651858.CD002025.pub4]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Amass 1994

Methods

Randomised, double‐blind, placebo‐controlled study

Participants

Setting: outpatient clinic, USA

Participants: N = 8, opioid dependent by DSM‐III‐R, using unspecified opioid drugs

Group sizes: group 1: N = 3; group 2: N = 5

Groups similar in withdrawal symptoms and positive urine samples during maintenance phase

Average age 39.4 years

87.5% male

Average duration opioid use 18.1 years, 87.5% reported i.v. use. One required benzodiazepine detoxification prior to study

Interventions

Buprenorphine solution (or placebo) administered sublingually, plus placebo pills during dose taper Buprenorphine increased to 8 mg/d over first three days, maintained for 28 days, then tapered. Group 1: 36 days (1 mg/4 days then 0.5 mg in last 4 days)

Group 2: 12 days (50% every 4 days)

Daily clinic attendance

Both groups received behavioural counselling (2‐3 sessions of 1 h each). Vouchers for recreational items given for negative urine samples

Outcomes

Change in withdrawal scores per day, % opioid‐negative urine samples, number in treatment for full 9 weeks, % continuously abstinent by week

Notes

Withdrawal rated by participants (20 signs and symptoms in prior 24 h, rated 0 (none) to 9 (severe), max score 180) and observers (8 signs, rated 0 (not at all) to 3 (severe), max score 24)

Observed urine and breath alcohol samples 3 times a week. Missed urine samples considered positive Source of funding for trial not reported; report preparation supported by government (NIDA) grants

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "subjects were assigned randomly . . . by drawing lots"

Allocation concealment (selection bias)

Unclear risk

Method of concealment not reported

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

Low risk

Double‐blind stated, with use of placebos. Quote: "Nursing staff who were blind to the treatment conditions completed an observer rating scale"

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

Double‐blind stated, with use of placebos, and these outcomes unlikely to be affected by awareness of treatment allocation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: "Missed (urine) samples were considered opioid positive for the purposes of data analyses." "A repeated measures analysis of variance (ANOVA) was performed to examine subject‐ and observer‐rated withdrawal, and mean percentage opioid‐negative urine samples across the treatment groups"

Comment: this approach less susceptible to bias due to missing data

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Assadi 2004

Methods

Randomised double‐blind, placebo‐controlled trial. Concurrent dependence on alcohol exclusion criterion

Participants

Setting: inpatient treatment, addiction ward of psychiatric hospital, Tehran, Iran

Participants: N = 40, opioid dependent by DSM‐IV, 68% using heroin, 32% opium

Group sizes: N = 20 in each group

Baseline characteristics of groups similar

Average age 31.5

39/40 male

Average duration opioid use 9 years; 50% using by injection or multiple routes; 30% employed, 55% married

Interventions

Buprenorphine, i.m. with:

Group 1: 4 × 1.5 mg between 12 pm and 6 pm day 1, 4 × 1.5 mg between 6 am and 12 pm day 2

Group 2: 2 × 1.5 mg day 1, tapered to 2 × 0.3 mg day 5

Adjunct medications (indomethacin, trazodone, chlorpromazine, hyoscine) as required. Relapse prevention treatment using naltrexone as aftercare

Outcomes

Withdrawal scores, days in treatment, number completing treatment

Notes

Withdrawal assessed once per day by participants (SOWS) and observer (OOWS)

Study conducted without financial support

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "randomly assigned . . . using a computer‐generated list of random numbers"

Allocation concealment (selection bias)

Unclear risk

Method of concealment not reported

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

Low risk

Patients and staff with patient contact, and person performing clinical research assessments all blind to group allocation. Placebo used

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

Patients and staff with patient contact, and person performing clinical research assessments all blind to group allocation. Placebo used

Incomplete outcome data (attrition bias)
All outcomes

Low risk

10% dropout in course of study. Missing data replaced by carrying last observation forward

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Bickel 1988

Methods

Randomised, double‐blind, placebo‐controlled trial

Participants

Setting: outpatient clinic, USA

Participants: N = 45, opioid dependent, using opiates other than methadone, seeking outpatient detoxification, eligible for methadone maintenance treatment (evidence of recent injecting use part of eligibility criteria)

Group sizes: group 1: N = 22; group 2: N = 23, but most analyses based on N = 17 from group 1 and N = 14 from group 2who remained in treatment at least 6 weeks

Groups similar on demographics

Average age 30 years

All male

Duration of heroin use, mean ± SD: group 1 6.7 ± 1.9 years; group 2: 9.2 ± 6.1 years

Interventions

Treatment regimens involved 3 weeks stabilisation, 4 weeks dose reduction, 6 weeks placebo dosing with:

Group 1: buprenorphine, sublingual solution, 2 mg/d or

Group 2: methadone, 30 mg/d, oral

Daily clinic attendance

All had access to supportive counselling. Scheduled 90‐day treatment

Outcomes

Mean duration in treatment, number in treatment for 6 weeks or more, graphs of retention, mean withdrawal scores and opiate positive or missing urine to week 6

Notes

Withdrawal rated by subjects 3 times a week (2 scales, each 20 symptoms rated 0‐9)

Urine samples collected according to random schedule

Study supported by government grants and pharmaceutical company

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Patients were randomised, stratifying for race"; method of sequence generation not specified

Allocation concealment (selection bias)

Unclear risk

Method of concealment not reported

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

Low risk

Quote: "Neither the subjects nor the nurses had information about which medication was active nor did they have any information about the dose‐reduction schedule."

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

Quote: "Neither the subjects nor the nurses had information about which medication was active nor did they have any information about the dose‐reduction schedule."

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Rapid dropout from week 6, towards the end of the dose taper of the two medications, but dropout similar in two groups. Retention main outcome reported after week 6 when dropout > 30%

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Cheskin 1994

Methods

Randomised, double‐blind, placebo‐controlled trial

Participants

Setting: inpatient, closed research ward, USA

Participants: N = 25 heroin users, with evidence of recent injecting use

Group sizes: group 1: N = 12; group 2: N = 13. Analysis primarily on basis of 18 who completed treatment

Group similar on demographic characteristics

Average age 35 years

44% male

Average duration heroin use 11.5 years

Interventions

Morphine used to attenuate withdrawal on admission. Subsequent treatment with:

Group 1: buprenorphine, sublingual solution, maximum of 2 mg/dose, total 17 mg over 3 days

Group 2: clonidine, maximum 0.3 mg/dose, total 2.7 mg over 5 days

Individual counselling twice weekly

Total study period 18 days. Placebo administered after completion of active medication phase

Outcomes

Graphs of mean 'urge' and 'need' for an opioid, mean withdrawal score, number completing 10 days of treatment, reasons for cessation

Notes

Visual analogue scales for rating 'urge' and 'need' for an opioid and subjective response to medication

Addiction Research Centre scales assessed opioid‐like euphoria, apathetic sedation and dysphoria

Daily urine screening

Study funded by US government research programme

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "patients were randomly assigned" with stratification based on response to naloxone challenge test. Method of sequence generation not reported

Allocation concealment (selection bias)

Unclear risk

Method of concealment not reported

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

Low risk

Quote: "double‐blind, double‐dummy . . . design"; extended treatment period to maintain blind

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

Quote: "double‐blind, double‐dummy . . . design"

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Analysis of withdrawal based on participants who completed the study. The significant difference in the first 3 days of treatment would be expected to be increased, not decreased, if dropouts had been included.

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Collins 2005

Methods

Randomised controlled, open‐label study

Participants

Setting: clinical research centre, New York, USA. Inpatient care for 72 h of withdrawal, outpatient for 12 weeks naltrexone maintenance treatment

Participants: N = 106, treatment seeking, heroin dependent by DSM‐IV

Group sizes: group 1: N = 35; group 2: N = 37; group 3: N = 34

Groups similar on demographic and clinical characteristics

Average age 36

72% male

40% using by injection; 36% currently married or cohabiting; 56% currently employed. Major psychiatric illness, active medical illness, dependence on alcohol or drugs other than heroin were exclusion criteria.

Interventions

Group 1 (excluded from review): nalmefene 4 mg i.v. over 30 min, naltrexone 50 mg via nasogastric tube, under propofol anaesthesia (4‐6 hours). Various adjunct medications, including octreotide

Group 2: buprenorphine (sublingual, preparation not specified), 8 mg (single dose) day 1, naltrexone 12.5 mg afternoon of day 2, 25 mg 12 hours later, then 50 mg/d

Group 3: clonidine, max 1.2 mg/d, discharged day 3, naltrexone 12.5 mg day 7, 25 mg day 8, then 50 mg/d. Clonidine and various adjunct medications available to all groups. Naltrexone maintenance and relapse prevention psychotherapy as aftercare

Outcomes

Graphs of withdrawal severity,.mean weeks in treatment. Number of participants completing inpatient phase, receiving at least one dose of naltrexone, receiving full 50 mg dose of naltrexone. Number retained in treatment over 12 weeks. Number retained 12 weeks who provided 2 or less opiate positive urine samples. Number experiencing serious adverse events

Notes

Withdrawal assessed by SOWS, OOWS, and Clinical Institute Narcotic Assessment, 4 times a day during inpatient phase

Study funded entirely by grants from US National Institutes of Health

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: ". . .using random, computer‐generated assignments, with stratification by sex. . . . In addition, the Berger‐Exner test was used to confirm that no selection bias in enrolment occurred."

Allocation concealment (selection bias)

Low risk

Quote: "All staff remained unaware of the randomisation sequence"

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

High risk

Patients were not blinded to treatment − sham anaesthesia not ethical. Unclear whether observers were blinded to treatment.

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

These outcomes unlikely to be affected by knowledge of treatment

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Losses to follow‐up similar in three groups and main outcomes reported not sensitive to missing data

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Unclear risk

Enrollment stopped at 106 participants (target 150) "because actual differences in withdrawal severity scores and treatment retention were smaller than anticipated, leading to an impractically large recalculated sample size"

Comment: it seems unlikely that the stopping of enrolment resulted in bias

Hopper 2005

Methods

Randomised controlled (open‐label) trial

Participants

Setting: residential therapeutic community, USA

Participants: N = 20, heroin dependent by DSM‐IV

Group sizes: N = 10 in each group

Groups similar on demographics and drug use

Average age 47 years

65% male

35% using heroin by injection (rest using nasally), 60% also cocaine dependent; 15% married, 20% employed in prior 30 days

Interventions

Buprenorphine/naloxone sublingual tablet, two dose regimens:

Group 1: 8 mg plus 24 mg after 30 min on day 1

Group 2: 8 mg day 1, 16 mg day 2, 8 mg day 3

Adjunct medications available as needed

Detoxification and monitoring over 7 days; follow‐up evaluations on days 14‐17

Outcomes

Number completing 7 days of detoxification programme, number completing follow‐up evaluation, mean daily withdrawal score, use of adjunct medication

Notes

Withdrawal assessed daily by Clinical Opiate Withdrawal Scale (COWS)

Urine testing 4 times in 7 days of detoxification

Source of funding government grants

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "patients were randomly assigned in an open‐label fashion"; method of sequence generation not reported.

Allocation concealment (selection bias)

High risk

Quote: "patients were randomly assigned in an open‐label fashion".

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

Unclear risk

Staff who assessed withdrawal symptoms daily were not aware of group assignments, but participants were probably aware of treatment allocation.

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

Treating staff were not aware of group assignments and these outcomes considered unlikely to be affected by lack of blinding

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No significant difference in dropouts between groups

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

High risk

All participants were resident in a therapeutic community. This may have resulted in selection of a highly motivated group who would have done well whatever the treatment.

Hussain 2015

Methods

Randomised controlled, open‐label trial

Participants

Setting: inpatient treatment facility, Srinagar, India

Participants: N = 58, opioid dependent by DSM‐IV, treatment seeking. Use of substances of abuse other than opioids was an exclusion criterion

Group sizes: N = 29 in each group

Groups similar on demographic characteristics

Average age 27 years

Sex not reported (probably all male)

Average 6 years of abuse; 69% employed, 26% married

Interventions

Group 1: buprenorphine/naloxone, sublingual, 4.0/1.0 mg on day 1, 8.0/2.0 mg/d on days 2‐4, 4.0/1.0 mg on days days 5‐7, 2.0/0.5 mg on days 8‐10

Group 2: clonidine, oral, 0.1 mg day 1, increasing to 0.2 mg on days 2‐4, tapered from day 5, ceased on day 10

Both groups received ancillary medications on demand

Outcomes

Average daily withdrawal score, average daily craving score, participants experiencing adverse effects, number completing study

Notes

Withdrawal assessed twice daily with Clinical Opiate Withdrawal Scale (COWS). Craving assessed with visual analogue scale (VAS)

Source of funding not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "participants were randomised into two groups using computer generated random numbers".

Allocation concealment (selection bias)

Unclear risk

Method of allocation concealment not reported

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

High risk

Quote: "open labelled study and blinding was not done"

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

These outcomes are unlikely to be affected by knowledge of the treatment group.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Low rate of attrition, and same in both groups

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Janiri 1994

Methods

Randomised double‐blind, placebo‐controlled trial

Participants

Setting: inpatient, hospital drug dependence unit, Italy

Participants: N = 39 from MMT requiring detoxification

Group sizes: N = 13 in each group. Analysis based on 32/39 who completed treatment

Groups stated to not differ on psychosocial and drug history data

Average age 26 years

59% male

Average duration opioid dependence 7.5 years, methadone maintenance 3.4 years; 56% using methadone only, 43% using methadone and heroin; polydrug use an exclusion criterion; 41% in stable job

Interventions

All stabilised on methadone, mean 23.7mg/d, tapered to 10 mg/d before study commenced Detoxification managed with:

Group 1: buprenorphine intramuscularly, 0.9 mg days 1 and 2, 0.45 mg day 3, 0.15 mg day 4

Group 2: Clonidine, 0.3 to 0.9 mg/d intramuscularly, 6 days or

Group 3 (excluded from review): Lefetamine, 60‐240 mg/d intramuscularly, 6 days. This group excluded from this review.

Clonidine and lefetamine groups given 5 mg methadone on days 1 and 2; oral and i.m. placebos used to maintain blind, no other drugs allowed.

Outcomes

Graphs of total, objective, subjective and psychological withdrawal scores, adjusted for baseline scores Number completing treatment

Notes

Withdrawal assessed by scale of Bruno and Ferracuti (12 objective, 10 subjective, 5 psychological items, each rated 0‐5). Urine screening used

Source of funding not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "patients were assigned randomly to one of three groups"

Comment: Method of sequence generation not reported but groups similar at baseline

Allocation concealment (selection bias)

Unclear risk

Method of allocation concealment not reported

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

Low risk

Quote: withdrawal "was rated by a trained psychiatrist, who was blind to study conditions".

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

Double‐blind stated and these outcomes unlikely to be affected by knowledge of intervention

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: "The 7 patients who dropped out were equally distributed among the three groups."

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Ling 2005

Methods

Randomised controlled open‐label trials − separate trials in outpatient and inpatient settings

Participants

Setting: inpatient and outpatient, in various community treatment programmes in USA

Participants: N = 113 (inpatient) and N = 231 (outpatient), heroin dependent by DSM‐IV

Group sizes: group 1: N = 77; group 2: N = 36 for inpatient study, group 1: N = 157; group 2: N = 74 for outpatient study

Groups similar on demographics, drug use history, except cannabis use with group 1 13.9%; group 2 2.6% meeting criteria for cannabis dependence

Average age 36 (inpatient) and 39 (outpatient)

68% male

Duration of heroin use more than 9 years for outpatient study, less than 7 years for inpatient study. 65% in buprenorphine groups injecting users, 70%‐94% across all groups worked full‐ or part‐time. Those codependent on other drugs not excluded

Interventions

Group 1: buprenorphine/naloxone sublingual tablet, first dose when in mild withdrawal, 4 mg day 1, with further 4 mg 1‐2 hours later, increasing to 16 mg day 3, tapering to 2 mg day 12/13

Group 2: clonidine, 0.05‐0.1 mg every 4‐6 hours day 1, plus transdermal patch from day 2, patch only from day 3, discontinued by day 13

Ancillary medications and counselling available. Medications administered daily. Scheduled duration 13 days

Outcomes

Success, defined as retention for entire study duration and opioid‐free urine on last day. Summary withdrawal, calculated as average of COWS scores across available observations. Average craving score.

Notes

Observers completed COWS (11 signs) daily. Participants completed Adjective Rating Scale for Withdrawal (16 items). Craving assessed by Visual Analogue Scale. Daily urine samples.

Source of funding: series of grants from US government (NIDA)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "patients were assigned randomly to bup‐nx or clonidine conditions using a 2:1 ratio in favour of buprenorphine"

Comment: based on contacts with the research group, we consider it likely that the method of sequence generation was adequate.

Allocation concealment (selection bias)

Low risk

Method of concealment not reported, but based on our knowledge of the research group, we consider it likely that concealment of allocation would have been achieved.

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

High risk

No blinding

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

No blinding, but these outcomes unlikely to be affected by knowledge of group allocation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Significantly more dropouts from clonidine groups, but method of analysis (ANCOVA) adjusted for missing data

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Unclear risk

Quote: "the NIDA Data and Safety Monitoring Board (DSMB) recommended that the study be halted prior to collecting the full complement of subjects for the following reasons: the large enrolment status, the consistency of findings overwhelmingly favourable toward the bup‐nx condition and the consideration that additional participant enrolment would not yield meaningful new information.

Comment: while early termination of studies is associated with some risk of bias, the extent of recruitment achieved is likely to have minimised this risk

Ling 2009

Methods

Randomised controlled, open‐label study. Randomisation stratified by buprenorphine dose at end of stabilisation (8, 16 or 24 mg)

Participants

Setting: outpatient, 11 treatment programmes, USA

Participants: N = 516, dependent on opioids other than methadone

Group sizes: group 1: N = 255; group 2: N = 261

No significant differences between groups in demographics or drug use at baseline

Average age 36 years

33% female

About 8 years of lifetime heroin use; exclusion criteria included positive urine test for methadone or benzodiazepine, and dependence on alcohol or any drug other than opioids; 35% unemployed in month prior to baseline

Interventions

Buprenorphine/naloxone sublingual tablet (Suboxone), 4‐week stabilisation with flexible dosing for 3 weeks to establish optimal dose, fixed at that dose for week 4, then tapered:

Group 1: over 7 days

Group 2: over 28 days

Psychosocial services as usual for both groups. Once weekly clinic attendance

Maximum 5 months covering screening, induction/stabilisation, taper and follow‐up

Outcomes

Mean withdrawal and craving scores at assessment time points, number in treatment at end of taper, number with opioid‐free urine samples at end of taper and at follow‐up

Notes

Participants rated withdrawal with Adjective Rating Scale (16 items each rated 0‐9). Clinicians rated withdrawal with COWS (range 0‐48). Visual analogue scale for overall withdrawal, craving and medication effectiveness. Weekly data collection to week 8. Incentive payments for each 'milestone' visit

Source of funding: series of grants from US government (NIDA)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "participants were assigned randomly. . . stratified by maintenance dose at the end of the stabilization phase".

Comment: This sort of randomisation would be expected to be computerised.

Allocation concealment (selection bias)

High risk

Quote: "Each treatment site had three sets of randomisation cards (one for each stabilisation dose) prepared in advance. Based on stabilisation dose, the top card in the appropriate stack was selected for the participant."

Comment: Concealment of allocation was unlikely meaning the sequence could be easily manipulated

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

High risk

Study conducted open label

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

Study open label but these outcomes unlikely to be affected by awareness of treatment allocation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No significant differences in baseline characteristics of those who remained in the study to the end of the taper and those who dropped out

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Lintzeris 2002

Methods

Randomised controlled, open‐label study

Participants

Setting: outpatient, specialist clinic, Australia

Participants: N = 114 heroin users, dependent by DSM‐IV

Group sizes: group 1: N = 58; group 2: N = 56

Groups similar on demographics and drug use

Average age 30 years

65% male

About 7 years since first regular heroin use, 90% injecting users; 39% employed full‐ or part‐time, 25% married/de facto. Some polydrug use, but significant other drug dependence an exclusion criterion

Interventions

Group 1: buprenorphine, sublingual tablet, supervised single daily dose around 6 mg/d, adjusted to symptoms, ceased day 5

Group 2: clonidine 0.1‐0.15 mg 4 times a day as required plus symptomatic medications

Daily clinic attendance for medication

Both groups received supportive counselling during withdrawal. Naltrexone, substitution treatment or counselling available as aftercare. Scheduled duration 8 days

Outcomes

Withdrawal score, treatment retention, time in treatment, days of heroin use, adverse effects, engagement in postwithdrawal treatment

Notes

Withdrawal assessed by SOWS and OOWS (Handelsman)

Urine screening used

Source of funding: government, buprenorphine provided by manufacturer

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "a computerized schedule was developed . . . using the technique of dynamic balanced randomisation . . . balancing treatment allocation within each site and across the study as a whole."

Allocation concealment (selection bias)

Low risk

Quote: "randomisation was conducted by an independent organization"

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

High risk

Quote: "patients, treatment providers and outcome assessors (were) aware of group allocation."

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

Quote: "patients, treatment providers and outcome assessors (were) aware of group allocation" but these outcomes unlikely to be affected by knowledge of group allocation.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Difference in dropout cannot account for significant differences between buprenorphine and clonidine treatment

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Marsch 2005

Methods

Randomised double‐blind, placebo‐controlled trial. Participants stratified by sex and past‐month route of opiate use (injection or intranasal)

Participants

Setting: outpatient, university‐based research clinic, USA

Participants: N = 36 adolescents (aged 13‐18 years), opiate dependent by DSM‐IV

Group sizes: N = 18 in each group

Groups similar on demographics and clinical criteria at baseline

Average age 17

39% male

Average age 15 years at first opiate use; 53% primarily heroin users, 36% injecting users

Interventions

Group 1: buprenorphine, sublingual tablets, commenced at 6 or 8 mg/day according to body weight and opiate use, decreased 2 mg every 7 days

Group 2: clonidine, 0.1 mg transdermal patches, 1 day 1, 2 days 2‐6, with optional 3rd days 4‐6, tapered after 7 days. Scheduled treatment duration 28 days

Behavioural counselling 3 times a week and incentives contingent on opiate abstinence. All offered naltrexone as aftercare

Outcomes

Number completing scheduled treatment. % urine tests opioid negative. Number initiating treatment with naltrexone

Notes

Withdrawal not assessed. Urine screening used

Funding support from government (NIDA) and university grants

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Participants were randomly assigned"; method not reported.

Allocation concealment (selection bias)

Unclear risk

Method of concealment not reported

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

Low risk

Quote: "double‐blind, double‐dummy design"

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

Quote: "double‐blind, double‐dummy design"

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Retention is major outcome. Analyses of secondary outcomes based on data from first week when retention still high in both groups

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Marsch 2016

Methods

Randomised controlled, double‐blind trial

Participants

Setting: Outpatient, in hospital‐based research clinics in New York City, USA

Participants: N=53 adolescents (age 16‐24), opioid‐dependent by DSM‐IV

Group sizes: (1) N=28 (2) N=25

Groups similar, except more participants in group (1) were nicotine dependent.

Average age 20.5 years

58% male

58% using iv, 81% primary heroin users. Pregnancy, active significant psychiatric disorder, serious medical condition were exclusion criteria.

Interventions

Commenced on sublingual tablets containing buprenorphine only, initial dose 6 or 8 mg based on bodyweight and drug use, additional 2, 4, 6 or 8 mg if withdrawal symptoms after 1 hour. Switched to sublingual buprenorphine/naloxone when stable (after 2 days). Taper adjusted to starting dose to cease after (1) 28 or (2) 56 days. Initially daily clinic attendance required; after 20 enrolments participants able to earn take home doses and clinic attendance required 2‐3 times weekly.

All received behavioural counselling and opioid abstinence incentives as adjunct therapies.

Outcomes

Abstinence as percentage of scheduled urine tests documented as negative (missed urine samples assumed to be positive)

Retention as number of days with attendance at scheduled visits.

Notes

Urine samples at intake and then randomly at scheduled visits.

Source of funding: grant from US government (NIDA); study medication provided by manufacturer.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "...participants were assigned randomly" "A minimum allocation procedure was used [to balance] groups on participant characteristics likely to influence treatment outcomes.

Allocation concealment (selection bias)

Unclear risk

Method of allocation concealment not reported.

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

Low risk

Double‐blind stated. Quote: "...each participant received the same number of identical‐appearing tablets ... throughout the trial".

No subjective outcomes reported.

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

Double‐blind stated. Quote: "...each participant received the same number of identical‐appearing tablets ... throughout the trial".

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No missing data

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Nigam 1993

Methods

Randomised controlled trial

Participants

Setting: inpatient, hospital, New Delhi, India

Participants: N = 72, opioid dependent by DSM‐III‐R. Published analysis based on 44 (of 72 recruited) who completed treatment. Additional information provided by authors

Group sizes: group 1: N = 34; group 2: N = 38

Groups similar on demographics, drug use history, withdrawal score at entry

Average age 29

All males

Duration heroin use 4‐5 years, 90% heroin users (1.5 g/day), rest opium users; no intravenous users; polydrug use an exclusion criterion; 64% married, 92% employed

Interventions

Group 1: buprenorphine, initial dose 0.6 mg/d, maximum 1.2 mg/d, sublingual tablet, in 3 divided doses

Group 2: clonidine, initial dose 0.3 mg/d, maximum 0.9 mg/d, oral, in 3 divided doses.

Nitrazepam as adjunct medication. Scheduled duration 10 days

Outcomes

Mean daily withdrawal scores. Side effects. Completion rates from personal communication

Notes

Withdrawal assessed daily using Subjective and Objective Opiate Withdrawal Scales (Handelsman). Urine screening used only for some participants

Source of funding not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Authors advised that subjects were randomly assigned but did not specify the method of sequence generation.

Allocation concealment (selection bias)

Unclear risk

Method of concealment not reported

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

High risk

Authors advised study not double‐blind

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

These outcomes unlikely to be affected by knowledge of group allocation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropout similar in two groups; withdrawal score comparisons based on those who completed treatment

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

O'Connor 1997

Methods

Randomised, double‐blind clinical trial

Participants

Setting: outpatient, primary care clinic, USA

Participants: N = 162, heroin dependent.

Group sizes: group 1: N = 55; group 2: N = 54; group 3: N = 53

Groups similar on sociodemographic and clinical characteristics

Age range 18 to 50 years (average not reported)

71% male

Average duration of heroin use > 7 years, polydrug users not excluded; 35% employed; 62% married

Interventions

Group 1: clonidine 0.1‐0.2 mg every 4 h as needed days 1‐7. 50 mg naltrexone day 8

Group 2 (excluded from review): clonidine as in group 1, 12.5 mg naltrexone day 1, increasing to 50 mg day 3

Group 3: buprenorphine 3 mg sublingually (preparation not specified) days 1‐3, then clonidine as in group 1, 25 mg naltrexone day 4, 50 mg day 5

Oxazepam, ibuprofen, ketorolac, prochlorperazine as adjunct medication

Daily clinic attendance except weekends. Participants referred for further treatment following trial

Outcomes

Mean overall and peak withdrawal scores. Number retained in treatment for 8 days. Number achieving 50 mg maintenance dose naltrexone

Notes

Withdrawal assessment primarily subjective symptoms

Funding support: government grant (NIDA)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "participants were randomly assigned"; method of sequence generation not reported.

Allocation concealment (selection bias)

Unclear risk

Method of concealment not reported

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

Unclear risk

Quote: "staff and patients were blinded to the protocols".

Comment: double‐blind possibly compromised by differential symptoms

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

Quote: "staff and patients were blinded to the protocols"

Comment: double‐blind possibly compromised by differential symptoms, but these outcomes unlikely to be affected by knowledge of treatment allocation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: "Retention did not differ significantly among the groups"

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

Outcome definition adjusted for differences in medication duration

Oreskovich 2005

Methods

Double‐blind, double‐dummy, randomised controlled trial

Participants

Setting: inpatient detoxification facility affiliated with University of Washington, USA

Participants: N = 30 heroin dependent by DSM‐IV, in moderate withdrawal at time of randomisation. One participant (clonidine group) discontinued due to protocol violation − main analysis on N = 29, complete demographic data on N = 28

Group sizes: group 1: N = 10; group 2: N = 10; group 3: N = 9

Groups similar on main demographic and clinical characteristics

Average age 36

63% male

Average duration of heroin use 6.5 to 10.3 years; polydrug users excluded if medically supervised withdrawal required for other drug dependence; unemployed group 1 10%, group 2 50%, group 3 25%

Interventions

Buprenorphine, sublingual tablet (Subutex), or placebo in two dose regimens:

Group 1: high dose: 8‐8‐8‐4‐2 mg/d or

Group 2: low dose: 2‐4‐8‐4‐2 mg/d on days 1‐5

Compared with group 3:clonidine, oral, 0.2‐0.3 mg four times a day for 5 days

Ancillary medications available

No specific behavioural therapies during withdrawal, but participants assessed for placement in outpatient treatment after completion of the study

Outcomes

Number achieving suppression of withdrawal; mean withdrawal score; chlordiazepoxide use; mean hours to discharge; doses of medication (or placebo) withheld due to hypotension

Notes

Withdrawal assessed by COWS, Adjective Rating Scale for Withdrawal, and Visual Analogue Craving Scale

Funding support from University of Washington. Buprenorphine and placebo provided by pharmaceutical company

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "randomly assigned . . . by a computerised random numbers generator"

Allocation concealment (selection bias)

Low risk

Quote: "(allocation) by the offsite research pharmacist"

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

Low risk

Double‐blind, double‐dummy stated

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

Double‐blind, double‐dummy stated

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Statistical methods used to adjust for missing data

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Petitjean 2002

Methods

Randomised, controlled (open‐label) study

Participants

Setting: inpatient, specialist unit, Switzerland

Participants: N = 37, opioid dependent by ICD‐10, using heroin or methadone

Group sizes: group 1: N = 19; group 2: N = 18

Groups similar on sociodemographics

Average age 32 years

78% male

Those with concurrent alcohol or benzodiazepine dependence were detoxified before entering study

Interventions

Medication tailored to declared amount of heroin use and withdrawal signs:

Group 1: buprenorphine, sublingual tablets, 8 mg/70 kg in 2 daily doses to max 16 mg/70 kg, reduced in 2 mg steps over average 12 days

Group 2: oral methadone, 40 mg/70kg to max 60 mg/70kg, reduced in 10 mg steps to 30 mg/70kg, then 5 mg steps, over total 15 days on average

Outcomes

Withdrawal severity, length of stay, completion rate

Notes

Withdrawal assessed by Short Opiate Withdrawal Scale. Urine testing used

Source of funding not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Patients were randomly assigned". Insufficient information reported on characteristics of groups to form a view on adequacy of sequence generation

Allocation concealment (selection bias)

Unclear risk

Method of concealment not reported

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

High risk

No blinding

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

These outcomes unlikely to be affected by lack of blinding

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropout evenly distributed between groups

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Ponizovsky 2006

Methods

Randomised, open‐label, controlled trial

Participants

Setting: inpatient, 2 separate specialist units, Jerusalem, Israel

Participants: N = 200 heroin dependent by ICD‐10

Group sizes: N = 100 in each group

Detailed demographics reported only for those who completed detoxification, but authors stated there were no significant differences in sociodemographic characteristics between completers and non‐completers within each group

Majority (> 60%) in each group aged between 25 and 44 years

86% male

Dependence on benzodiazepines or alcohol an exclusion criterion. 19% married, 88% unemployed

Interventions

Group 1: buprenorphine (Subutex) 10 mg day 1 with extra if needed, tapered to finish day 10

Group 2: clonidine, 0.6 mg/d in divided doses, tapered from day 5

10 days detoxification followed by relapse prevention programme

Outcomes

Number completing treatment. Mean adverse effects score. Adverse effects, well‐being assessed only for those who completed 10‐day detoxification

Notes

Withdrawal severity not reported

Study supported in part by pharmaceutical company, and by government

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "we used a random number generator"

Allocation concealment (selection bias)

Unclear risk

Method of allocation not reported. Groups treated in separate hospitals.

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

High risk

No blinding

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

These outcomes unlikely to be affected by lack of blinding

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Significantly more dropout from clonidine group. It is unclear how this difference in dropout may have affected the adverse symptoms mean score, which is the only outcome relevant to this review

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Raistrick 2005

Methods

Open‐label, randomised controlled trial

Participants

Setting: outpatient, specialist clinic, Leeds, UK

Participants: N = 210 heroin dependent by ICD‐10. Data also reported for N = 271 who opted not to be allocated randomly and chose 1 of the 2 treatments

Group sizes: group 1: N = 107; group 2: N = 103

Groups similar on baseline variables (but limited data reported)

Average age 28 years

75% male

Interventions

Group 1: buprenorphine, sublingual tablet, 4 mg day 1, 6‐8 mg day 2, 6 mg day 3, then tapered and ceased day 7; naltrexone offered 2 days after last buprenorphine

Group 2: lofexidine, 0.4 mg 4‐hourly as needed for 4 days; co‐phenotrope, hyoscine, chlordiazepoxide, chlorpromazine as needed day 1, naltrexone 25 mg day 4

Daily clinic attendance

Outcomes

Mean daily withdrawal severity, completion rate, abstinence at 1 month

Notes

Withdrawal assessed by Short Opiate Withdrawal Scale. Urine screening on completion

Funding from treatment service with contribution from pharmaceutical company but without input to study design, conduct, or report

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "participants were allocated randomly"; method not reported

Allocation concealment (selection bias)

Low risk

Quote: "predetermined random sequence . . . held by an administrator"

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

Unclear risk

Quote: "open‐label randomised controlled trial" and "The researcher was blind to . . . the treatment regimen given"

Comment: this suggests that observers, but not participants or treating staff were blind to group allocation. This introduces some risk of bias for measures completed by participants.

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

These outcomes unlikely to be affected by lack of blinding

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Peak withdrawal scores recorded on day 2 when missing data similar in both groups (20% and 22%). No other outcome data affected by differences in dropout rates

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

More than half eligible participants chose not to enter the trial, but data reported for these cohorts and outcomes were similar to the groups randomised

Schneider 2000

Methods

Randomised controlled open‐label trial

Participants

Setting: inpatient detoxification ward, Hannover, Germany

Participants: N = 27 methadone and/or heroin users, dependent by DSM‐IV

Group sizes: group 1: N = 15; group 2: N = 12

Groups similar on sociodemographics and drug use history

Average age 31 years

89% male

Average duration opioid use 11.9 ± 5.4 years (group 1); 8.7 ± 5.8 years (group 2). All polydrug users. 1st inpatient detoxification for around half of the participants.

Interventions

Group 1: buprenorphine, 3 mg/d for 7 days, then tapered and ceased day 11

Group 2: oxazepam, 90 mg/d for 7 days then tapered and ceased day 15

Both groups received carbamazepine, 900 mg/d for 7 days, then tapered and ceased day 20. Total 21‐day treatment

Outcomes

Graph of withdrawal scores. Number giving 'drug not effective' as reason for dropout. Number NOT completing treatment

Notes

Withdrawal assessed using Short Opiate Withdrawal Scale. Urine screening used

Source of funding: Government grant. Buprenorphine provided by pharmaceutical company

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Patients were randomly assigned"; method not reported

Allocation concealment (selection bias)

Unclear risk

Method of concealment not reported

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

High risk

Open‐label stated (authors suggest that medication effects would have revealed group allocation)

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

These outcomes unlikely to be affected by lack of blinding

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: "the difference in the dropout rate between the two treatment strategies was not significant."

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Seifert 2002

Methods

Double‐dummy randomised controlled trial

Participants

Setting: inpatient treatment in ward for drug and alcohol abuse, Hannover, Germany

Participants: N = 26, opioid dependent (methadone or heroin) by DSM‐IV

Group sizes: group 1: 14; group 2: 12

Groups similar on demographics and drug use history, except 6/14 in group 1 and 10/12 in group 2 abused cannabis in addition to opioids

Average age 32 years

85% male

Average duration of opioid abuse about 10 years First inpatient detoxification for 19%. All polydrug users

Interventions

All given 25 mg methadone, 400 mg carbamazepine on admission then:

Group 1: buprenorphine, 4 mg/d for 3 days then tapered to cease day 10, or

Group 2: L‐methadone, 20 mg day 1, tapered to cease day 10

All received carbamazepine, 900 mg/d for 6 days, then tapered to cease day 14

Outcomes

Withdrawal severity, completion rate

Notes

Withdrawal assessed by Short Opiate Withdrawal Scale. Observed urine samples weekly

Source of funding not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Patients were randomly assigned"; method not reported

Allocation concealment (selection bias)

Unclear risk

Method of concealment not reported

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

Unclear risk

Quote: "double‐dummy, parallel group study design" "Buprenorphine placebo was obtained from the pharmacy"

Comment: it is unclear whether participants, treating personnel and observers were all blind to group allocation

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

These outcomes unlikely to be affected by blinding

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: "the difference in the dropout rate was not significant"

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Sigmon 2013

Methods

Randomised controlled trial, double‐blind, double dummy, with stabilisation dose, abstinence during stabilisation, cocaine use, gender, alcohol dependence, and current pain as stratification variables

Participants

Setting: outpatient, research clinic, Vermont, USA

Participants: N = 70, opioid dependent by DSM‐IV

Group sizes: group 1: N = 24; group 2: N = 24; group 3: N = 22

Baseline characteristics of groups similar

Average age 27 years

69% male

Average 4.9 years of opioid abuse; primary drug of abuse oxycodone for 57%, buprenorphine for 39%; 16% used primary by intravenous injection

94% white, 63% employed full‐time, 24% ever married

Interventions

Stabilised on buprenorphine/naloxone for 2 weeks, then dose tapered:

Group 1: over 1 week

Group 2: over 2 weeks

Group 3: over 4 weeks

Participants received 5.5 sublingual tablets at each visit containing a combination of either active/placebo buprenorphine so participants and staff were blind to dose, taper and commencement of naltrexone

At completion of buprenorphine taper, all groups were commenced on naltrexone, 12.5 mg day 1, 25 mg days 2 and 3, 50 mg day 4, then 50 mg/d

All participants received behavioural therapy based on Community Reinforcement Approach

Participants visited the clinic daily in weeks 1‐5, then 3 times a week during weeks 6‐12

Outcomes

Participants abstinent from opioids, retention in treatment, participants receiving naltrexone, withdrawal scores

Notes

Urine samples were collected three times a week. Withdrawal and craving were assessed by visual analogue scales, and the CINA

Source of funding government grant (NIDA); buprenorphine and placebo provided by manufacturer through NIDA

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Participants . . . were then randomly assigned . . . Stratification variables included stabilization dose, illicit opioid abstinence during stabilization, past‐month cocaine use, sex, current alcohol dependence,and current chronic pain not reported."

Comment: method of sequence generation not reported, but computerisation is likely given the number of stratification variables used

Allocation concealment (selection bias)

Low risk

Quote: "Doses were prepared by the hospital’s investigational pharmacy".

Comment: although method of allocation concealment not specifically reported, the preparation of identical medications by the hospital pharmacy makes it likely that concealment was adequate

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

Low risk

Study stated as double‐blind with placebos used so that "neither the participants nor the staff knew the doses received, the duration of taper and the point at which naltrexone therapy began."

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

Study double‐blind and double‐dummy

Incomplete outcome data (attrition bias)
All outcomes

High risk

Analysis of withdrawal severity based on a subset of participants (n = 28, 40% of all participants) who were verified to be abstinent from opioids while the buprenorphine dose was being tapered

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Steinmann 2008

Methods

Randomised controlled trial

Participants

Setting: inpatient, detoxification unit, Wasserburg, Germany

Participants: N = 60

Group sizes: group 1: N = 30; group 2: N = 30

Groups similar on demographics and drug use at baseline

Average age 27 years

83% male, duration of heroin use about 7 years

Interventions

Group 1: methadone, 60 mg/d, tapered 2.5‐5 mg/d.

Group 2: buprenorphine, 12‐16 mg/d, tapered 0.8‐1.2 mg/d.

Study duration 28 days

Outcomes

Craving, withdrawal, use of additional medication, number completing treatment

Notes

Withdrawal assessed with COWS and tailored instrument (24 items)

Source of funding not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "patients were randomly assigned"; method not reported

Allocation concealment (selection bias)

Unclear risk

Method of concealment not reported

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

High risk

"Due to technical reasons" there was no blinding

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

These outcomes unlikely to be affected by lack of blinding

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: "The two groups of patients did not differ . . . in the rate at which they successfully completed the trial."

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Umbricht 2003

Methods

Randomised, double‐blind clinical trial, with stratification on withdrawal severity, pain, CD4 T‐cell count

Participants

Setting: inpatient, AIDS service, USA

Participants: N = 55, HIV positive, opioid‐dependent by self‐report and physical exam, hospitalised for acute medical illness

Group sizes: group 1: N = 21; group 2: N = 16; group 3. N = 18

Those in group 1 more likely to have been admitted for fever/cellulitis. Groups otherwise similar

Average age 40 years

62% male

Duration of heroin use around 18 years. Concurrent alcohol dependence, enrolment in MMT exclusion criteria.

Interventions

All stabilised with morphine 10 mg i.m. every 4 h as needed up to 6 h prior to enrolment in study then 3 day taper with:

Group 1: buprenorphine 0.6 mg i.m. every 4 h day 1, every 6 h day 2, every 8 h day 3,

Group 2: oral clonidine, 0.2 mg loading dose, 0.1 mg every 4 h day 1, every 6 h day 2, every 8 h day 3, or

Group 3: oral methadone, 30 mg day 1, 20 mg day 2, 10 mg day 3. All received clonidine transdermal patch day 4

No adjunct treatment for withdrawal

Outcomes

Withdrawal severity, completion rate, adverse effects, use of supplemental morphine for pain

Notes

Withdrawal assessed by Short Opiate Withdrawal Scale (participants) and Objective Opiate Withdrawal Scale (observers)

Source of funding: government grant (NIDA)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "patients . . . were randomly assigned"; "patients were stratified on four characteristics". Method of sequence generation was not reported

Allocation concealment (selection bias)

Unclear risk

Method of concealment not reported

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

Unclear risk

Quote: "To maintain the blind, one active medication and two inactive medications were administered to all participants."

Comment: It remains unclear whether participants, treating personnel and observers were all blind to group allocation

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

These outcomes unlikely to be affected by knowledge of treatment allocation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Statistical methods allowed for missing data and variation in time of assessment

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

Wang 1996

Methods

Double‐blind controlled trial, with 14/23 participants randomly allocated

Participants

Setting: not stated − probably outpatient, Milwaukee, USA

Participants: N = 23 who had completed maintenance phase of preliminary trial of buprenorphine

Group sizes: group 1: N = 14 (N = 7 randomly allocated); group 2: N = 7

Note: this review only used data for the participants who were randomly allocated

Participant characteristics not reported (conference abstract only information available)

Interventions

Buprenorphine 8 mg/d, tapered:

Group 1: rapidly (14 days)

Group 2: slowly (8 weeks)

Placebo to total 12 weeks

Outcomes

Descriptive comments on withdrawal; completion rate

Notes

Withdrawal rating scale not used. Urine screening not reported

Source of funding: government grant (NIDA)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "The first 14 (patients) were randomized into two groups"

Comment: method of allocation not reported. Data reported separately for the 14 participants who were allocated randomly, and the remaining 9 participants who were all allocated to group 1.

Allocation concealment (selection bias)

Unclear risk

Method of concealment not reported

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

High risk

Double‐blind only for those allocated randomly

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

These outcomes unlikely to be affected by lack of blinding

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Completion of withdrawal treatment is the only outcome reported

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Unclear risk

Unable to assess (conference abstract only report available)

Wright 2011

Methods

Randomised, open‐label, controlled trial

Participants

Setting: healthcare services in high, but not maximum, security prisons (2 male, 1 female), England

Participants: prisoners using illicit opioids, wanting to detoxify and remain abstinent, in custody at least 28 days

Group sizes: group 1: N = 148; group 2: N = 141

Groups similar on baseline characteristics

Median age 31 years

Median duration of opioid use 10 years; 53% used primarily by injection

Interventions

Group 1: buprenorphine, sublingual (probably tablets) starting dose 8 mg, tapered to 0.4 mg at day 20

Group 2: methadone (oral, 1 mg/1 ml mixture), starting dose 30 mg, tapered to 2 mg (day 20)

Outcomes

Opioid abstinence at 8 days, 1, 3 and 6 months after detoxification. Urinalysis for opioid abstinence at 8 days postdetoxification. Factors predictive of abstinence

Notes

Withdrawal severity not assessed

Source of funding: government grant

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Randomisation sequence (with random block size) was generated using Microsoft
Excel RAND function."

Allocation concealment (selection bias)

Low risk

Quote: "Sealed, opaque, consecutively numbered envelopes concealing the name of the allocated intervention were prepared by a researcher who had no contact with participants."

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

Low risk

Study undertaken open‐label because "blinded study would have necessitated commercial funding to develop dummy preparations . . . [and] there is also potential bias in study findings where there is a commercial funder". Because it was open‐label, subjective outcomes were not used.

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

Study undertaken open‐label, but these outcomes at less risk of bias due to awareness of treatment allocation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Loss to follow‐up at primary outcome (abstinence at 8 days postdetoxification) similar for two groups: group 1: 24%; group 2: 29%

Selective reporting (reporting bias)

Unclear risk

Data collected on adverse effects but not reported

Other bias

Low risk

None apparent

Ziaaddini 2012

Methods

Randomised, double‐blind, controlled trial

Participants

Setting: inpatient psychiatric hospital, Kerman, Iran

Participants: opioid dependent by DSM‐IV, randomly selected from individuals referred for detoxification

Group sizes: group 1: N = 14; group 2: N = 21

Employed: 42.9% (group 1); 71.4% (group 2); groups otherwise similar

Average age 25 years

All male

34% married

Interventions

Group 1: buprenorphine, sublingual tablet (plus clonidine placebo), 2 mg day 1, 4 mg day 2, 6 mg day 3, 4 mg day 4, 2 mg day 5 with additional 2‐4 mg if required for severe withdrawal

Group 2: clonidine, oral tablets, 0.4 mg day 1, 0.6 mg days 2 and 3, 0.2 mg days 4 and 5, plus additional 0.2‐0.4 mg/d if required for severe withdrawal

Actual doses administered not reported

Outcomes

Average change in withdrawal and craving scores on days 1, 2, 3, and 5. Total days in treatment and days receiving naltrexone at follow‐up

Notes

Withdrawal rated by 'psychiatric technician' with COWS (11 items rated 0‐4), and by participants with ARWS (Adjective Rating Withdrawal Scale, 16 items rated 0‐9). Craving assessed with visual analogue scale

Funding source intramural

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "participants were randomly allocated to . . . detoxification group."

Method of sequence generation not explained

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding (performance bias and detection bias)
Subjective outcomes ‐ intensity of withdrawal, adverse effects)

Low risk

Double‐blind stated, and placebos used to maintain blind. Quote: "All drugs and placebos were assigned a code and kept by a person who was not involved in the study"

Blinding (performance bias and detection bias)
Objective outcomes ‐ duration of treatment, completion of treatment

Low risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: "All patients took naltrexone at the end of the detoxification period"

Comment: this review focuses on the detoxification period, and if all participants completed detoxification there should be no incomplete data

Selective reporting (reporting bias)

Low risk

None apparent

Other bias

Low risk

None apparent

COWS: Clinical Opiate Withdrawal Scale; DSM‐IV: Diagnostic and Statistical Manual of Mental Health ‐ 4th edition; DSM‐III‐R: Diagnostic and Statistical Manual of Mental Health ‐ 3rd edition, Revised; i.m.: intramuscular; i.v.: intravenous; MMT: methadone maintenance treatment; OOWS: Objective Opiate Withdrawal Scale; SOWS: Subjective Opiate Withdrawal Scale.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Blondell 2008

Randomised controlled trial comparing effect of 2 different periods of extended buprenorphine prescription following inpatient detoxification. No data on detoxification outcomes defined for this review. Not primarily about withdrawal management

Breen 2003

Comparison of 3 different regimens for transferring participants from methadone to buprenorphine, with subsequent tapering of buprenorphine dose. No comparison during withdrawal phase and insufficient data on outcomes of this phase.

Fiellin 2014

Randomised controlled trial comparing buprenorphine/naloxone for withdrawal management and substitution treatment. Treatment comparison is not one of the modalities included in the review

Fingerhood 2001

Clonidine or buprenorphine offered consecutively for management of opioid withdrawal. Not randomised controlled trial

Frownfelter 2001

Investigation of buprenorphine as an adjunct to antagonist‐induced withdrawal. The groups differed only in the timing of naltrexone administration. Treatment comparison not one defined by inclusion criteria

Gandhi 2003

Randomised controlled trial comparing 2 different dose regimens of buprenorphine for management of opioid withdrawal. Groups combined for analysis of postdetoxification outcomes. No data on detoxification phase

Gibson 2003

Comparison of outcomes of opioid withdrawal managed with buprenorphine in specialist clinic or primary health setting. Treatment comparison not defined by inclusion criteria

Gonzalez 2015

Randomised controlled trial comparing memantine and placebo as adjuncts to buprenorphine for management of opioid withdrawal. Not one of the comparisons specified for this review

Hao 2000

Treatment comparison (WeiniCom, a Chinese herbal preparation with complex pharmacology) is not one of the modalities defined by the inclusion criteria.

Harris 2003

Comparison of buprenorphine and lofexidine for management of opioid withdrawal. Not a controlled study − partly retrospective; insufficient outcome data (conference abstract only)

He 1999

Controlled study comparing buprenorphine plus naloxone from day 5, with tincture of opium. Unclear whether group allocation was random. Treatment comparison (tincture of opium) is not one of the modalities defined by the inclusion criteria.

Jain 2011

Comparison of memantine and placebo in conjunction with buprenorphine in terms of capacity to ameliorate withdrawal induced with naloxone. Study more about pharmacology of memantine than management of withdrawal

Jia 2001

Randomised controlled trial comparing methadone followed by buprenorphine versus buprenorphine alone for managing opioid withdrawal. Primary treatment approach unclear − it appears that some participants in buprenorphine only group received methadone during first few days of treatment. Comparison is not one specified for this review

Johnson 1989

Preliminary report of trial comparing methadone and buprenorphine for the treatment of opioid dependence. Insufficient outcome data

Johnson 1992a

Comparison of buprenorphine and clonidine for management of opioid withdrawal. Insufficient data − conference abstract only. Probably preliminary report of Cheskin 1994

Johnson 1992b

Randomised controlled trial comparing buprenorphine and methadone (60 or 20 mg/d) for 17 weeks of maintenance followed by 8 weeks withdrawal. Comparison not one specified for this review. Focus on maintenance rather than management of withdrawal

Johnson 1996

Study of withdrawal symptoms following abrupt cessation of methadone or buprenorphine. Focus on pharmacological properties rather than management of withdrawal. Insufficient data − conference abstract only

Katz 2009

Comparison of brief (5‐day) and extended (30‐day) buprenorphine treatment for outpatient management of opioid withdrawal. Data drawn from 2 previous studies comparing counselling approaches as adjuncts to buprenorphine. Not randomised controlled trial

Kosten 1988

Open label study comparing 3 different dose regimens of buprenorphine in terms of illicit opiate use and withdrawal symptoms during 30 days at a fixed dose. Buprenorphine abruptly discontinued after 30 days but no data reported on this withdrawal phase. Study more about substitution treatment than management of withdrawal

Kosten 1991

Reports use of buprenorphine for 30 days as transition between methadone or heroin and naltrexone, with comparison of reactions to naloxone and naltrexone. No treatment comparison for withdrawal phase

Law 2002

Participants stabilised on methadone or buprenorphine then detoxified by combination of tapered methadone plus lofexidine, or tapered buprenorphine. Insufficient data (conference abstract, preliminary report)

Liebschutz 2014

Randomised controlled trial comparing 5‐day buprenorphine detoxification with stabilisation on buprenorphine in hospital and linkage to outpatient opioid substitution treatment. Comparison not one specified for this review.

Lintzeris 2003

Investigation of doses of buprenorphine required to suppress opioid withdrawal symptoms. No treatment comparison

Liu 1997

Comparison of different dose regimens of buprenorphine, with starting dose determined by duration and dose of heroin use and severity of dependence. Not a randomised controlled trial

Maayan 2008

Randomised controlled trial comparing DHEA and placebo as adjuncts to buprenorphine for management of opioid withdrawal. Analysis largely in terms of factors affecting response to DHEA treatment. Insufficient outcome data, and not primarily a study of buprenorphine

Meade 2010

Randomised controlled trial comparing active and sham transcutaneous electric acupoint stimulation (TEAS) as an adjunct to inpatient detoxification managed with buprenorphine. Focus of study on drug use following detoxification. Insufficient data on withdrawal episode

Montoya 1994

Comparison of naltrexone‐buprenorphine combination with buprenorphine only for managing opioid withdrawal. Investigation of pharmacology of buprenorphine and naltrexone rather than management of withdrawal

Pjrek 2012

Medication selection based on judgement of treating clinician. Not randomised controlled trial. Focus on sleep parameters − insufficient data on outcomes defined for this review

Pycha 1994

Comparison of 2 rates of reduction of buprenorphine dose for management of opioid withdrawal. Only information available is conference abstract − contact author advised study was never fully written up. Insufficient information on medications and characteristics of patients, insufficient outcome data

Reed 2007

Participants able to choose buprenorphine or methadone for management of opioid withdrawal

Resnick 1992

Randomised controlled trial comparing buprenorphine detoxification and continuation of buprenorphine substitution treatment for opioid dependence. Comparison not one of the modalities defined by the inclusion criteria

Rosen 1995

Participants maintained on buprenorphine for 30 days then given challenges with naltrexone or naloxone as process of easing transition to naltrexone maintenance treatment. No treatment comparison

Rosenthal 2013

Randomised controlled trial comparing active buprenorphine/naloxone implants with placebo implants and with sublingual buprenorphine/naloxone tablets. Opiate withdrawal and craving were assessed in the context of initiation of substitution (maintenance) treatment rather than managing withdrawal

Sanders 2013

Randomised controlled trial comparing gabapentin and placebo as adjuncts to buprenorphine for treatment of opioid withdrawal. Intervention is not one defined by the inclusion criteria for this review

Sheard 2009

Comparison of buprenorphine and dihydrocodeine for management of opioid withdrawal in prison setting. Comparison not one of the modalities defined by inclusion criteria

Sigmon 2004

Randomised placebo‐controlled trial of depot preparation of buprenorphine. Focus is on effectiveness in providing opioid blockade and suppression of withdrawal, but not full withdrawal intervention

Sullivan 2015

Randomised controlled trial comparing 2 different buprenorphine regimens to manage induction onto naltrexone treatment. Intervention not one defined by inclusion criteria for this review. Preliminary report (conference abstract) − insufficient outcome data

Tompkins 2014

Cross‐over study assessing withdrawal syndrome following cessation of morphine or buprenorphine. No intervention to manage withdrawal

Umbricht 1999

Comparison of buprenorphine‐naltrexone combination with buprenorphine followed by naltrexone for management of opioid withdrawal. Treatment comparison not one of the modalities defined by the inclusion criteria

Weiss 2010

Randomised controlled trial comparing adjunct therapies with phases of buprenorphine maintenance and dose taper. Comparison not one defined by the inclusion criteria for this review

White 2001

Comparison of buprenorphine and lofexidine for management of opioid withdrawal. Mixed retrospective and prospective data collection − not a controlled study

Williams 2002

Reports the use of buprenorphine for management of opioid withdrawal in an outpatient setting. No treatment comparison

Woody 2008

Comparison of 12 week buprenorphine maintenance and 14‐day detox in terms of positive urine samples and retention in treatment for opioid dependence. Comparison is not one of the modalities defined by the inclusion criteria

Wright 2007

Randomised controlled trial comparing buprenorphine and dihydrocodeine for management of opioid withdrawal in primary care setting. Comparison not one of the modalities defined by the inclusion criteria.

Zhang 1993

Comparison of buprenorphine and methadone for management of opioid withdrawal. Insufficient information on treatment regimens and outcomes

Data and analyses

Open in table viewer
Comparison 1. Buprenorphine versus methadone

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mean days in treatment Show forest plot

2

82

Mean Difference (IV, Random, 95% CI)

1.30 [‐8.11, 10.72]

Analysis 1.1

Comparison 1 Buprenorphine versus methadone, Outcome 1 Mean days in treatment.

Comparison 1 Buprenorphine versus methadone, Outcome 1 Mean days in treatment.

2 Completion of withdrawal treatment Show forest plot

5

457

Risk Ratio (M‐H, Random, 95% CI)

1.04 [0.91, 1.20]

Analysis 1.2

Comparison 1 Buprenorphine versus methadone, Outcome 2 Completion of withdrawal treatment.

Comparison 1 Buprenorphine versus methadone, Outcome 2 Completion of withdrawal treatment.

Open in table viewer
Comparison 2. Buprenorphine versus clonidine

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mean peak withdrawal score Show forest plot

6

521

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

‐0.43 [‐0.74, ‐0.13]

Analysis 2.1

Comparison 2 Buprenorphine versus clonidine, Outcome 1 Mean peak withdrawal score.

Comparison 2 Buprenorphine versus clonidine, Outcome 1 Mean peak withdrawal score.

2 Mean overall withdrawal score Show forest plot

7

902

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

‐0.43 [‐0.58, ‐0.28]

Analysis 2.2

Comparison 2 Buprenorphine versus clonidine, Outcome 2 Mean overall withdrawal score.

Comparison 2 Buprenorphine versus clonidine, Outcome 2 Mean overall withdrawal score.

3 Mean days in treatment Show forest plot

4

558

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

0.92 [0.57, 1.27]

Analysis 2.3

Comparison 2 Buprenorphine versus clonidine, Outcome 3 Mean days in treatment.

Comparison 2 Buprenorphine versus clonidine, Outcome 3 Mean days in treatment.

3.1 Inpatient setting

3

213

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

1.00 [0.33, 1.67]

3.2 Outpatient setting

2

345

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

0.82 [0.57, 1.06]

4 Number experiencing adverse effects Show forest plot

3

493

Risk Ratio (M‐H, Random, 95% CI)

0.93 [0.70, 1.26]

Analysis 2.4

Comparison 2 Buprenorphine versus clonidine, Outcome 4 Number experiencing adverse effects.

Comparison 2 Buprenorphine versus clonidine, Outcome 4 Number experiencing adverse effects.

4.1 Inpatient

2

148

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.12, 1.36]

4.2 Outpatient

2

345

Risk Ratio (M‐H, Random, 95% CI)

1.02 [0.66, 1.58]

5 Number with treatment stopped due to adverse effects Show forest plot

3

134

Risk Ratio (M‐H, Random, 95% CI)

0.20 [0.04, 1.15]

Analysis 2.5

Comparison 2 Buprenorphine versus clonidine, Outcome 5 Number with treatment stopped due to adverse effects.

Comparison 2 Buprenorphine versus clonidine, Outcome 5 Number with treatment stopped due to adverse effects.

5.1 Inpatient

3

134

Risk Ratio (M‐H, Random, 95% CI)

0.20 [0.04, 1.15]

5.2 Outpatient

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

6 Number completing withdrawal treatment Show forest plot

11

1264

Risk Ratio (M‐H, Random, 95% CI)

1.59 [1.23, 2.06]

Analysis 2.6

Comparison 2 Buprenorphine versus clonidine, Outcome 6 Number completing withdrawal treatment.

Comparison 2 Buprenorphine versus clonidine, Outcome 6 Number completing withdrawal treatment.

6.1 Inpatient

6

539

Risk Ratio (M‐H, Random, 95% CI)

1.74 [1.05, 2.89]

6.2 Outpatient

6

725

Risk Ratio (M‐H, Random, 95% CI)

1.45 [1.12, 1.88]

Open in table viewer
Comparison 3. Rapid versus slow dose taper

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Completion of withdrawal treatment Show forest plot

6

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 3.1

Comparison 3 Rapid versus slow dose taper, Outcome 1 Completion of withdrawal treatment.

Comparison 3 Rapid versus slow dose taper, Outcome 1 Completion of withdrawal treatment.

1.1 Inpatient

2

60

Risk Ratio (M‐H, Random, 95% CI)

1.0 [0.84, 1.18]

1.2 Outpatient

4

647

Risk Ratio (M‐H, Random, 95% CI)

0.86 [0.44, 1.70]

2 Number abstinent at completion of dose taper Show forest plot

5

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 3.2

Comparison 3 Rapid versus slow dose taper, Outcome 2 Number abstinent at completion of dose taper.

Comparison 3 Rapid versus slow dose taper, Outcome 2 Number abstinent at completion of dose taper.

2.1 Inpatient

1

40

Risk Ratio (M‐H, Random, 95% CI)

1.0 [0.81, 1.23]

2.2 Outpatient

4

610

Risk Ratio (M‐H, Random, 95% CI)

0.94 [0.39, 2.24]

Study flow diagram.
Figuras y tablas -
Figure 1

Study flow diagram.

Methodological quality graph: review authors' judgements about each methodological quality item presented as percentages across all included studies.
Figuras y tablas -
Figure 2

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

Methodological quality summary: review authors' judgements about each methodological quality item for each included study.
Figuras y tablas -
Figure 3

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

Comparison 1 Buprenorphine versus methadone, Outcome 1 Mean days in treatment.
Figuras y tablas -
Analysis 1.1

Comparison 1 Buprenorphine versus methadone, Outcome 1 Mean days in treatment.

Comparison 1 Buprenorphine versus methadone, Outcome 2 Completion of withdrawal treatment.
Figuras y tablas -
Analysis 1.2

Comparison 1 Buprenorphine versus methadone, Outcome 2 Completion of withdrawal treatment.

Comparison 2 Buprenorphine versus clonidine, Outcome 1 Mean peak withdrawal score.
Figuras y tablas -
Analysis 2.1

Comparison 2 Buprenorphine versus clonidine, Outcome 1 Mean peak withdrawal score.

Comparison 2 Buprenorphine versus clonidine, Outcome 2 Mean overall withdrawal score.
Figuras y tablas -
Analysis 2.2

Comparison 2 Buprenorphine versus clonidine, Outcome 2 Mean overall withdrawal score.

Comparison 2 Buprenorphine versus clonidine, Outcome 3 Mean days in treatment.
Figuras y tablas -
Analysis 2.3

Comparison 2 Buprenorphine versus clonidine, Outcome 3 Mean days in treatment.

Comparison 2 Buprenorphine versus clonidine, Outcome 4 Number experiencing adverse effects.
Figuras y tablas -
Analysis 2.4

Comparison 2 Buprenorphine versus clonidine, Outcome 4 Number experiencing adverse effects.

Comparison 2 Buprenorphine versus clonidine, Outcome 5 Number with treatment stopped due to adverse effects.
Figuras y tablas -
Analysis 2.5

Comparison 2 Buprenorphine versus clonidine, Outcome 5 Number with treatment stopped due to adverse effects.

Comparison 2 Buprenorphine versus clonidine, Outcome 6 Number completing withdrawal treatment.
Figuras y tablas -
Analysis 2.6

Comparison 2 Buprenorphine versus clonidine, Outcome 6 Number completing withdrawal treatment.

Comparison 3 Rapid versus slow dose taper, Outcome 1 Completion of withdrawal treatment.
Figuras y tablas -
Analysis 3.1

Comparison 3 Rapid versus slow dose taper, Outcome 1 Completion of withdrawal treatment.

Comparison 3 Rapid versus slow dose taper, Outcome 2 Number abstinent at completion of dose taper.
Figuras y tablas -
Analysis 3.2

Comparison 3 Rapid versus slow dose taper, Outcome 2 Number abstinent at completion of dose taper.

Summary of findings for the main comparison. Buprenorphine vs methadone for managing opioid withdrawal

Buprenorphine versus methadone for managing opioid withdrawal

Patient or population: Adults with opioid dependence
Setting: Inpatient or outpatient
Intervention: Buprenorphine
Comparison: Methadone

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Quality of the evidence
(GRADE)

Risk with methadone

Risk with buprenorphine

Mean days in treatment

The mean days in treatment in the methadone group was 10.8 or 48.5 days.

The mean days in treatment in the buprenorphine group was 1.3 more (8.1 less to 10.7 more) than in the methadone group.

82
(2 RCTs)

⊕⊕⊝⊝
Lowa,b

Completion of withdrawal treatment

Study population

RR 1.04
(0.91 to 1.20)

457
(5 RCTs)

⊕⊕⊕⊝
Moderateb

528 per 1000

549 per 1000
(481 to 634)

Moderate

534 per 1000

555 per 1000
(486 to 641)

*The risk in the intervention group (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; RCT: randomised controlled trial; RR: risk ratio; OR: odds 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.

aStudies were conducted in different settings (inpatient and outpatient).
bSmall number of participants (N < 300).

Figuras y tablas -
Summary of findings for the main comparison. Buprenorphine vs methadone for managing opioid withdrawal
Summary of findings 2. Buprenorphine vs alpha2‐adrenergic agonists for managing opioid withdrawal

Buprenorphine vs alpha2‐adrenergic agonists for managing opioid withdrawal

Patient or population: Adults or adolescents with opioid dependence
Setting: Inpatient or outpatient
Intervention: Buprenorphine
Comparison: Alpha2‐adrenergic agonists (clonidine or lofexidine)

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Quality of the evidence
(GRADE)

Risk with alpha2‐adrenergic agonists

Risk with buprenorphine

Mean peak withdrawal score, assessed with various scales

Not able to be summarised due to different means of assessment

The mean peak withdrawal score was lower in the buprenorphine group, indicating less severity, with a standardised mean difference of 0.43, which is a moderate effect size

SMD ‐0.43 (‐0.74 to ‐0.13)

521
(6 RCTs)

⊕⊝⊝⊝
Very lowa,b,c,d

Mean overall withdrawal score, with various methods of assessment

Not able to be summarised due to different means of assessment

The mean overall withdrawal score was lower in the buprenorphine group, indicating less severity, with a standardised mean difference of 0.43, which is a moderate effect size

SMD ‐0.43 (‐0.58 to ‐0.28)

902
(7 RCTs)

⊕⊕⊕⊝
Moderatea

Mean days in treatment (with varying scheduled duration)

Mean days in treatment ranged across studies from 21% to 70% of scheduled treatment duration

Mean days in treatment ranged across studies from 25% to 97% of scheduled treatment duration, with a standardised mean difference of 0.92, which is a large effect size.

SMD 0.92 (0.57 to 1.27)

558
(4 RCTs)

⊕⊕⊕⊝
Moderatec,d

Number experiencing adverse effects

Study population

RR 0.93
(0.70 to 1.26)

493
(3 RCTs)

⊕⊝⊝⊝
Very lowa,b,c

289 per 1000

269 per 1000
(202 to 364)

Moderate

127 per 1000

118 per 1000
(89 to 160)

Number with treatment stopped due to adverse effects

Study population

RR 0.20
(0.04 to 1.15)

134
(3 RCTs)

⊕⊕⊝⊝
Lowb

90 per 1000

18 per 1000
(4 to 103)

Moderate

79 per 1000

16 per 1000
(3 to 91)

Number completing withdrawal treatment

Study population

RR 1.59
(1.23 to 2.06)

1264
(11 RCTs)

⊕⊕⊕⊝
Moderated

453 per 1000

721 per 1000
(558 to 934)

Moderate

536 per 1000

852 per 1000
(659 to 1000)

*The risk in the intervention group (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; RCT: randomised controlled trial; RR: risk ratio; OR: odds 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.

aRisk of bias in blinding of subjective outcomes.
bSmall number of events (n < 300).
cDifferent treatment settings (outpatient and inpatient).
dHeterogeneity > 60%.

Figuras y tablas -
Summary of findings 2. Buprenorphine vs alpha2‐adrenergic agonists for managing opioid withdrawal
Summary of findings 3. Rapid vs slow taper of buprenorphine dose for managing opioid withdrawal

Rapid versus slow taper of buprenorphine dose for managing opioid withdrawal

Patient or population: Adults or adolescents with opioid dependence
Setting: Inpatient or outpatient
Intervention: Rapid
Comparison: Slow dose taper

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Quality of the evidence
(GRADE)

Risk with slow dose taper

Risk with rapid taper

Completion of withdrawal treatment − Inpatient

Study population

RR 1.00
(0.84 to 1.18)

60
(2 RCTs)

⊕⊕⊝⊝
Lowa

900 per 1000

900 per 1000
(756 to 1000)

Moderate

900 per 1000

900 per 1000
(756 to 1000)

Completion of withdrawal treatment − Outpatient

Study population

RR 0.86
(0.44 to 1.70)

647
(4 RCTs)

⊕⊝⊝⊝
Very lowb,c

594 per 1000

511 per 1000
(261 to 1000)

Moderate

636 per 1000

547 per 1000
(280 to 1000)

Number abstinent at completion of dose taper − Inpatient

Study population

RR 1.00
(0.81 to 1.23)

40
(1 RCT)

⊕⊕⊝⊝
Lowa,d

900 per 1000

900 per 1000
(729 to 1000)

Moderate

900 per 1000

900 per 1000
(729 to 1000)

Number abstinent at completion of dose taper − Outpatient

Study population

RR 0.94
(0.39 to 2.24)

619
(4 RCTs)

⊕⊝⊝⊝
Very lowb,c

320 per 1000

301 per 1000
(125 to 717)

Moderate

337 per 1000

317 per 1000
(131 to 755)

*The risk in the intervention group (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;RCT: randomised controlled trial; RR: risk ratio; OR: odds 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.

aSmall number of events (n < 300).
bHigh degree of heterogeneity (>75%).
cMost studies small.
dOnly one study.

Figuras y tablas -
Summary of findings 3. Rapid vs slow taper of buprenorphine dose for managing opioid withdrawal
Comparison 1. Buprenorphine versus methadone

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mean days in treatment Show forest plot

2

82

Mean Difference (IV, Random, 95% CI)

1.30 [‐8.11, 10.72]

2 Completion of withdrawal treatment Show forest plot

5

457

Risk Ratio (M‐H, Random, 95% CI)

1.04 [0.91, 1.20]

Figuras y tablas -
Comparison 1. Buprenorphine versus methadone
Comparison 2. Buprenorphine versus clonidine

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mean peak withdrawal score Show forest plot

6

521

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

‐0.43 [‐0.74, ‐0.13]

2 Mean overall withdrawal score Show forest plot

7

902

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

‐0.43 [‐0.58, ‐0.28]

3 Mean days in treatment Show forest plot

4

558

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

0.92 [0.57, 1.27]

3.1 Inpatient setting

3

213

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

1.00 [0.33, 1.67]

3.2 Outpatient setting

2

345

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

0.82 [0.57, 1.06]

4 Number experiencing adverse effects Show forest plot

3

493

Risk Ratio (M‐H, Random, 95% CI)

0.93 [0.70, 1.26]

4.1 Inpatient

2

148

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.12, 1.36]

4.2 Outpatient

2

345

Risk Ratio (M‐H, Random, 95% CI)

1.02 [0.66, 1.58]

5 Number with treatment stopped due to adverse effects Show forest plot

3

134

Risk Ratio (M‐H, Random, 95% CI)

0.20 [0.04, 1.15]

5.1 Inpatient

3

134

Risk Ratio (M‐H, Random, 95% CI)

0.20 [0.04, 1.15]

5.2 Outpatient

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

6 Number completing withdrawal treatment Show forest plot

11

1264

Risk Ratio (M‐H, Random, 95% CI)

1.59 [1.23, 2.06]

6.1 Inpatient

6

539

Risk Ratio (M‐H, Random, 95% CI)

1.74 [1.05, 2.89]

6.2 Outpatient

6

725

Risk Ratio (M‐H, Random, 95% CI)

1.45 [1.12, 1.88]

Figuras y tablas -
Comparison 2. Buprenorphine versus clonidine
Comparison 3. Rapid versus slow dose taper

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Completion of withdrawal treatment Show forest plot

6

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 Inpatient

2

60

Risk Ratio (M‐H, Random, 95% CI)

1.0 [0.84, 1.18]

1.2 Outpatient

4

647

Risk Ratio (M‐H, Random, 95% CI)

0.86 [0.44, 1.70]

2 Number abstinent at completion of dose taper Show forest plot

5

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 Inpatient

1

40

Risk Ratio (M‐H, Random, 95% CI)

1.0 [0.81, 1.23]

2.2 Outpatient

4

610

Risk Ratio (M‐H, Random, 95% CI)

0.94 [0.39, 2.24]

Figuras y tablas -
Comparison 3. Rapid versus slow dose taper