Scolaris Content Display Scolaris Content Display

Antiviral prophylaxis for the prevention of chronic hepatitis C virus in patients undergoing liver transplantation

Contraer todo Desplegar todo

Referencias

References to studies included in this review

Belli 2001 {published data only}

Belli LS, Alberti AB, Rondinara GF, de Carlis L, Corti A, Mazza E, et al. Early ribavirin treatment and avoidance of corticosteroids in hepatitis C virus (HCV)‐positive liver transplant recipients: interim report of a prospective randomized trial. Transplantation Proceedings2001; Vol. 33, issue 1‐2:1353‐4.

Chalasani 2005 {published data only}

Alpert E, Levy GA, Marotta P, Deschenes M, Yoshida E, Peck‐Radosavljevic M, et al. Peginterferon alfa‐2a for hepatitis C after liver transplantation: two randomized, controlled trials (vol 41, pg 289, 2004). Hepatology 2005;42(2):506.
Chalasani N, Manzarbeitia C, Ferenci P, Vogel W, Fontana RJ, Voigt M, et al. Peginterferon alfa‐2a for hepatitis C after liver transplantation: two randomized, controlled trials. Hepatology 2005;41(2):289‐98.
Manzarbeitia C, Tepermann L, Chalasani N, Sheiner P, Wiesner R, Marks IM, et al. 40 KDA peginterferon alfa‐2a (PEGASYS) as a prophylaxis against hepatitis C infection recurrence after liver transplantation (LT): preliminary results of a randomized multicenter trial (abstract). Hepatology 2001;34(4 Pt 2):406A.
Vogel W, Ferenci P, Fontana R, Arbor A, Saab S, LaBrecque D, et al. Peginterferon alfa‐2A (40 kd) (PEGASYS) in liver transplant recipients with established recurrent hepatitis C: interim results of an ongoing randomized multicenter trial [abstract]. Hepatology 2002;36(4 Pt 2):312A.

Charlton 2007 {published data only}

Bzowej N, Nelson DR, Terrault NA, Everson GT, Teng LL, Prabhakar A, et al. PHOENIX: a randomized controlled trial of peginterferon alfa‐2a plus ribavirin as a prophylactic treatment after liver transplantation for hepatitis C virus. Liver Transplantation 2011;17(5):528‐38.
Bzowej N, Nelson DR, Terrault NA, Everson GT, Teng LL, Prabhakar A, et al. Phoenix: a randomized controlled trial of peginterferon alfa‐2a/ribavirin prophylactic treatment after liver transplant for hepatitis C. Liver Transplantation 2010;16(Suppl 1):S118.
Bzowej NH, Nelson D, Terrault N, Everson GT, Teng LL, Prabhakar A, et al. A randomized controlled trial of the efficacy, tolerability, and safety of prophylactic treatment with peginterferon alfa‐2a plus ribavirin after orthotopic liver transplantation (OLT) for hepatitis C: The PHOENIX Study. Hepatology 2009;50(4 Suppl):547a.
Bzowej NH, Nelson D, Thommes JA, Hamzeh FM, Charlton M. A randomized controlled trial of prophylactically administered peginterferon alfa‐2A plus ribavirin vs no prophylaxis following orthotopic liver transplantation (OLT) for hepatitis C: a report of initial safety and tolerability. Hepatology2006; Vol. 44, issue 4 Suppl 1:188A.
Charlton MR, Bzowej N, Rossi S, Nelson DR. Prophylactic peginterferon alfa‐2a/ribavirin vs no prophylaxis following orthotopic liver transplantation (OLT) for hepatitis C: 24‐week virologic and safety responses. Hepatology 2007;46(4 Suppl 1):244a.

Chung 2013 {published data only}

Chung RT, Gordon FD, Curry MP, Schiano TD, Emre S, Corey K, et al. Human monoclonal antibody MBL‐HCV1 delays HCV viral rebound following liver transplantation: a randomized controlled study. American Journal of Transplantation 2013;13(4):1047‐54.

Davis 2005 {published data only}

Davis GL, Nelson DR, Terrault N, Pruett TL, Schiano TD, Fletcher CV, et al. A randomized, open‐label study to evaluate the safety and pharmacokinetics of human hepatitis C immune globulin (Civacir) in liver transplant recipients. Liver Transplantation 2005;11(8):941‐9.

Mazzaferro 2003 {published data only}

Mazzaferro V, Schiavo M, Caccamo L, Tagger A, Morabito A, Lavezzo B, et al. Prospective randomized trial on early treatment of HCV infection after liver transplantation in HCV‐RNA positive patients. Liver Transplantation 2003;9(6):C36.

Reddy 2002 {published data only}

Reddy R, Fried M, Dickson R, Martin P, Schiff E, Torres M, et al. Interferon alfa‐2b and ribavirin vs. placebo as early treatment in patients transplanted for hepatitis C end‐stage liver disease: results of a multicenter, randomized trial. Gastroenterology 2002;122(4 Suppl 1):A632.

Schiano 2006 {published data only}

Schiano TD, Charlton M, Younossi Z, Galun E, Pruett T, Tur‐Kaspa R, et al. Monoclonal antibody HCV‐Ab(XTL)68 in patients undergoing liver transplantation for HCV: results of a phase 2 randomized study. Liver Transplantation 2006;12(9):1381‐9.

Sheiner 1998 {published data only}

Sheiner PA, Boros P, Klion FM, Thung SN, Schluger LK, Lau JYN, et al. The efficacy of prophylactic interferon alfa‐2b in preventing recurrent hepatitis C after liver transplantation. Hepatology 1998;28(3):831‐8.
Sheiner PA, Boros P, Thung SN, Klion FM, Emre S, Guy SR, et al. Prophylactic interferon‐alpha2b reduces the incidence of recurrent hepatitis after liver transplantation for hepatitis C. 16th Annual Meeting of American Society of Transplant Physicians (ASTP). 1997:262.

Shergill 2005 {published data only}

Shergill AK, Khalili M, Straley S, Bollinger K, Roberts JP, Ascher NA, et al. Applicability, tolerability and efficacy of preemptive antiviral therapy in hepatitis C‐infected patients undergoing liver transplantation. American Journal of Transplantation 2005;5(1):118‐24.
Terrault NA, Khalili M, Straley S, Bollinger K, Bass N, Roberts JP, et al. Efficacy and tolerability of preemptive interferon (IFN) versus IFN plus ribavirin (RBV) treatment in hepatitis C virus (HCV) infected liver transplant recipients [AASLD abstract]. Hepatology 2003;38(4 Suppl 1):158a‐9a.

Singh 1998 {published data only}

Singh N, Gayowski T, Wannstedt CF, Shakil AO, Wagener MM, Fung JJ, et al. Interferon‐alpha for prophylaxis of recurrent viral hepatitis C in liver transplant recipients: a prospective, randomized, controlled trial. Transplantation1998; Vol. 65, issue 1:82‐6.

Willems 2002 {published data only}

Willems B, Ede M, Marotta P, Wall W, Greig P, Lilly L, et al. Anti‐HCV human immunoglobulins for the prevention of graft infection in HCV‐related liver transplantation, a pilot study [abstract]. Journal of Hepatology 2002;36(Suppl 1):32.

References to studies excluded from this review

Beckebaum 2003 {published data only}

Beckebaum S, Cicinnati VR, Karliova M, Dirsch O, Erim Y, Frilling A, et al. Daily interferon alpha‐2B and ribavirin combination therapy for liver transplant patients with chronic hepatitis C infection. Transplantation Proceedings 2003;35(6):2080‐1.

Boillot 1995 {published data only}

Boillot O, Berger F, Rasolofo E, Mion F, Chevallier P, Gille D, et al. Effects of early interferon alfa therapy for hepatitis C virus infection recurrence after liver transplantation. Transplantation Proceedings1995; Vol. 27, issue 4:2501.

Casanovas 2004 {published data only}

Casanovas TT, Casais ALA, Samuel D, Bizollon T, Trepo C. Chronic hepatitis C after liver transplantation: a randomized study (5) (multiple letters). Gastroenterology 2004;126(1):373‐4.

Castedal 2003 {published data only}

Castedal M, Siewert DA, Olausson M, Friman S. Combination therapy of interferon alpha‐2B and ribavirin for recurrent hepatitis C after liver transplantation. Transplantation Proceedings 2003;35(2):820‐1.

Catalano 2003 {published data only}

Catalano G, Urbani L, Oliveri F, Iaria G, Biancofiore G, Mosca F, et al. Recurrence of hepatitis C in liver transplants from elderly donors aged more than 75 years. Transplantation Proceedings 2003;35(3):1034.

Ceccherini 2003 {published data only}

Ceccherini NL, Giannotti A, Malizia T, Ciccorossi P, Olivieri F, Vanni M, et al. Recurrence of HCV infection in liver transplant patients: evaluation of IgM anti‐HCV and IgM anti‐CMV. Transplantation Proceedings 2003;35(3):1030‐1.

Charlton 2002 {published data only}

Charlton M. Pre‐emptive treatment of recurrent hepatitis C infection. Liver Transplantation 2002;8(10 Suppl 1):S50‐S54.

Everson 2013 {published data only}

Everson GT, Terrault N, Lok AS, Brown RS, Saab S, Shiffman ML, et al. Interim analysis of a controlled trial of pre‐transplant peginterferon alfa‐2b/ribavirin (PEG/RBV) to prevent recurrent hepatitis C virus (HCV) infection after liver transplantation (LT) in the adult‐to‐adult liver transplantation (A2ALL) Study. Hepatology 2009;50(4 Suppl):302a.
Everson GT,  Terrault NA,  Lok AS,  Rodrigo del R,  Brown RS,  Saab S,  et al. A randomized controlled trial of pretransplant antiviral therapy to prevent recurrence of hepatitis C after liver transplantation. Hepatology 2013;57(5):1752‐62.

Garcia‐Retortillo 2004 {published data only}

Garcia‐Retortillo M, Forns X. Prevention and treatment of hepatitis C virus recurrence after liver transplantation. Journal of Hepatology 2004;41(1):2‐10.

Mazzaferro 1997 {published data only}

Mazzaferro V, Regalia E, Pulvirenti A, Tagger A, Andreola S, Pasquali M, et al. Prophylaxis against HCV recurrence after liver transplantation: effect of interferon and ribavirin combination. Transplantation Proceedings 1997;29(1‐2):519‐21.

Roche 2011 {published data only}

Roche B, Samuel D. Is early antiviral therapy for recurrent hepatitis C after liver transplantation superior to later treatment? The answer is no. Liver Transplantation 2011;17(5):488‐91.

Samuel 2004a {published data only}

Samuel D. Hepatitis C, interferon, and risk of rejection after liver transplantation. Liver Transplantation 2004;10(7):868‐71.

Samuel 2004b {published data only}

Samuel D, Bizollon T, Trepo C. Chronic hepatitis C after liver transplantation: a randomized study ‐ reply. Gastroenterology2004; Vol. 126, issue 1:373‐4.

Taltavull 2004 {published data only}

Taltavull TC, Alvarez LAC. Chronic hepatitis C after liver transplantation: a randomized study. Gastroenterology 2004;126(1):373.

Afdhal 2004

Afdhal NH, Dieterich DT, Pockros PJ, Schiff ER, Shiffman ML, Sulkowski MS, et al. Epoetin alfa maintains ribavirin dose in HCV‐infected patients: a prospective, double‐blind, randomized controlled study. Gastroenterology 2004;126(5):1302‐11.

Berenguer 2003

Berenguer M, Crippin J, Gish R, Bass N, Bostrom A, Netto G, et al. A model to predict severe HCV‐related disease following liver transplantation. Hepatology 2003;38(1):34‐41.

Bombuy 2004

Bombuy E, Fondevila C, Rodriguez‐Laiz G, Ferrer J, Amador A, Valentini M, et al. Ischemic preconditioning in adult living donor liver transplantation, a pilot study [EASL abstract]. Journal of Hepatology 2004;40(Suppl 1):39.

Brok 2008

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

Brok 2009

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

Brok 2010

Brok J, Gluud LL, Gluud C. Ribavirin plus interferon versus interferon for chronic hepatitis C. Cochrane Database of Systematic Reviews 2010, Issue 1. [DOI: 10.1002/14651858.CD005445.pub2]

Cameron 2006

Cameron AM, Ghobrial RM, Hiatt JR, Carmody IC, Gordon SA, Farmer DG, et al. Effect of nonviral factors on hepatitis C recurrence after liver transplantation. Annals of Surgery 2006;244(4):563‐71.

Cescon 2006

Cescon M, Grazi GL, Grassi A, Ravaioli M, Vetrone G, Ercolani G, et al. Effect of ischemic preconditioning in whole liver transplantation from deceased donors. A pilot study. Liver Transplantation 2006;12(4):628‐35.

Corno 2006

Corno V, Colledan M, Dezza MC, Guizzetti M, Lucianetti A, Maldini G, et al. Extended right split liver graft for primary transplantation in children and adults. Transplantation International 2006;19(6):492‐9.

CTU 2011

Copenhagen Trial Unit. TSA ‐ Trial Sequential Analysis, 2011. ctu.dk/tsa/ (accessed 25 November 2013).

DeMets 1987

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

DerSimonian 1986

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

Dieterich 2003

Dieterich DT, Wasserman R, Brau N, Hassanein TI, Bini EJ, Bowers PJ, et al. Once‐weekly epoetin alfa improves anemia and facilitates maintenance of ribavirin dosing in hepatitis C virus‐infected patients receiving ribavirin plus interferon alfa. American Journal of Gastroenterology 2003;98(11):2491‐9.

Eason 2001

Eason JD, Loss GE, Blazek J, Nair S, Mason AL. Steroid‐free liver transplantation using rabbit antithymocyte globulin induction: results of a prospective randomized trial. Liver Transplantation 2001;7(8):693‐7.

Egger 1997

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

Forman 2002

Forman LM, Lewis JD, Berlin JA, Feldman HI, Lucey MR. The association between hepatitis C infection and survival after orthotopic liver transplantation. Gastroenterology 2002;122(4):889‐96.

Ghobrial 1999

Ghobrial RM, Farmer DG, Baquerizo A, Colquhoun S, Rosen HR, Yersiz H, et al. Orthotopic liver transplantation for hepatitis C: outcome, effect of immunosuppression, and causes of retransplantation during an 8‐year single‐center experience. Annals of Surgery 1999;229(6):824‐31; discussion 831‐3.

Gluud 2007

Gluud C, Brok J, Gong Y, Koretz RL. Hepatology may have problems with putative surrogate outcome measures. Journal of Hepatology 2007;46(4):734‐42.

Gluud 2013

Gluud C, Nikolova D, Klingenberg SL, Alexakis N, Als‐Nielsen B, Colli A, et al. Cochrane Hepato‐Biliary Group. About The Cochrane Collaboration (Cochrane Review Groups (CRGs)). 2013, Issue 5. Art. No.: LIVER.

Gurusamy 2009

Gurusamy KS, Osmani B, Xirouchakis E, Burroughs AK, Davidson BR. Antiviral therapy for recurrent liver graft infection with hepatitis C virus. Cochrane Database of Systematic Reviews 2009, Issue 1. [DOI: 10.1002/14651858.CD006803.pub2]

Gurusamy 2013

Gurusamy KS, Wilson E, Koretz RL, Allen VB, Davidson BR, Burroughs AK, et al. Is sustained virological response a marker of treatment efficacy in patients with chronic hepatitis C viral infection with no response or relapse to previous antiviral intervention?. PLoS One 2013;in press.

Higgins 2002

Higgins JPT, Thompson SG. Quantifying heterogeneity in a meta‐analysis. Statistics in Medicine 2002;21(11):1539‐58.

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.

ICH‐GCP 1997

International Conference on Harmonisation Expert Working Group. International conference on harmonisation of technical requirements for registration of pharmaceuticals for human use. ICH harmonised tripartite guideline. Guideline for good clinical practice CFR & ICH Guidelines. Vol. 1, PA 19063‐2043, USA: Barnett International/PAREXEL, 1997.

Jain 2002

Jain A, Kashyap R, Demetris AJ, Eghstesad B, Pokharna R, Fung JJ. A prospective randomized trial of mycophenolate mofetil in liver transplant recipients with hepatitis C. Liver Transplantation 2002;8(1):40‐6.

Kamath 2001

Kamath PS, Wiesner RH, Malinchoc M, Kremers W, Therneau TM, Kosberg CL, et al. A model to predict survival in patients with end‐stage liver disease. Hepatology 2001;33(2):464‐70.

Kjaergard 2001

Kjaergard LL, Villumsen J, Gluud C. Reported methodologic quality and discrepancies between large and small randomized trials in meta‐analyses. Annals of Internal Medicine 2001;135(11):982‐9.

Koneru 2005

Koneru B, Fisher A, He Y, Klein KM, Skurnick J, Wilson DJ, et al. Ischemic preconditioning in deceased donor liver transplantation: a prospective randomized clinical trial of safety and efficacy. Liver Transplantation 2005;11(2):196‐202.

Lim 2006

Lim SG, Wai CT, Da Costa M, Sutedja DS, Lee YM, Lee KH, et al. Referral patterns and waiting times for liver transplantation in Singapore. Singapore Medical Journal 2006;47(7):599‐603.

Lundh 2012

Lundh A, Sismondo S, Lexchin J, Busuioc OA, Bero L. Industry sponsorship and research outcome. Cochrane Database of Systematic Reviews 2012, Issue 12. [DOI: 10.1002/14651858.MR000033.pub2]

Macaskill 2001

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

Moher 1998

Moher D, Pham B, Jones A, Cook DJ, Jadad AR, Moher M, et al. Does quality of reports of randomised trials affect estimates of intervention efficacy reported in meta‐analyses?. Lancet 1998;352(9128):609‐13.

Newell 1992

Newell DJ. Intention‐to‐treat analysis: implications for quantitative and qualitative research. International Journal of Epidemiology 1992;21(5):837‐41.

NHS UK Transplant

NHS UK Transplant ‐ activity report 2011‐2012. www.organdonation.nhs.uk/statistics/transplant_activity_report/current_activity_reports/ukt/liver_activity.pdf (accessed 25 November 2013).

OPTN/SRTR 2005

The Organ Procurement and Transplant Network/Scientific Registry of Transplant Recipients 2009 annual report. www.ustransplant.org/annual_reports/current/905_li.pdf (accessed 25 November 2013).

Parmar 1998

Parmar MK, Torri V, Stewart L. Extracting summary statistics to perform meta‐analyses of the published literature for survival endpoints. Statistics in Medicine 1998;17(24):2815‐34. [PUBMED: 9921604]

RevMan 2012 [Computer program]

The Nordic Cochrane Centre, The Cochrane Collaboration. Review Manager (RevMan). Version 5.2. Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboration, 2012.

Royle 2003

Royle P, Milne R. Literature searching for randomized controlled trials used in Cochrane reviews: rapid versus exhaustive searches. International Journal of Technology Assessment in Health Care 2003;19(4):591‐603.

Savovic 2012

Savovic J, Jones HE, Altman DG, Harris RJ, Jüni P, Pildal J, et al. Influence of reported study design characteristics on intervention effect estimates from randomized, controlled trials. Health Technology Assessment 2012;16(35):1‐82.

Savovic 2012a

Savovic J, Jones HE, Altman DG, Harris RJ, Jüni P, Pildal J, et al. Influence of reported study design characteristics on intervention effect estimates from randomized, controlled trials. Annals of Internal Medicine 2012;157(6):429‐38.

Schulz 1995

Schulz KF, Chalmers I, Hayes RJ, Altman DG. Empirical evidence of bias. Dimensions of methodological quality associated with estimates of treatment effects in controlled trials. JAMA 1995;273(5):408‐12.

Sharvadze 2006

Sharvadze L, Tsertsvadze T, Gochitashvili N, Kakabadze T, Dolmazashvili E. IFN/RBV treatment induced anemia and its correction with epoetin alpha in patients with hepatitis C. Georgian Medical News 2006;137:62‐5.

Sharvadze 2007

Sharvadze L, Gochitashvili N, Tophuria A, Bolokadze N, Tsertsvadze T. IFN/RBV treatment induced neutropenia and its correction with neupogen in patients with hepatitis C. Georgian Medical News 2007;147:52‐5.

Shiffman 2006

Shiffman ML, Saab S, Feng S, Abecassis MI, Tzakis AG, Goodrich NP, et al. Liver and intestine transplantation in the United States, 1995‐2004. American Journal of Transplantation 2006;6(5 Pt 2):1170‐87.

Sugo 2003

Sugo H, Balderson GA, Crawford DH, Fawcett J, Lynch SV, Strong RW, et al. The influence of viral genotypes and rejection episodes on the recurrence of hepatitis C after liver transplantation. Surgery Today 2003;33(6):421‐5.

Thorlund 2009

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

Thorlund 2010

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

Thorlund 2011

Thorlund K, Engstrøm J, Wetterslev J, Brok J, Imberger G, Gluud C. User manual for Trial Sequential Analysis (TSA), 2011. ctu.dk/tsa/files/tsa_manual.pdf (accessed 25 November 2013).

Wetterslev 2008

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

Wetterslev 2009

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

Wood 2008

Wood L, Egger M, Gluud LL, Schulz KF, Jüni P, Altman GD, et al. Empirical evidence of bias in treatment effect estimates in controlled trials with different interventions and outcomes: meta‐epidemiological study. BMJ (Clinical Research Ed.) 2008;336:601‐5.

Yilmaz 2007

Yilmaz N, Shiffman ML, Stravitz RT, Sterling RK, Luketic VA, Sanyal AJ, et al. A prospective evaluation of fibrosis progression in patients with recurrent hepatitis C virus following liver transplantation. Liver Transplantation 2007;13(7):975‐83.

References to other published versions of this review

Gurusamy 2010

Gurusamy KS, Tsochatzis E, Davidson BR, Burroughs AK. Antiviral prophylactic intervention for chronic hepatitis C virus in patients undergoing liver transplantation. Cochrane Database of Systematic Reviews 2010, Issue 12. [DOI: 10.1002/14651858.CD006573.pub2]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Belli 2001

Methods

Randomised clinical trial.

Participants

Country: Italy.
Sample size: 19.
Post‐randomisation drop‐out: unclear.
Revised sample size: 19.
Females: not stated.
Mean age: not stated.
Genotype 1 (intervention): not stated.
Genotype 1 (control): not stated.
Growth factors for bone marrow suppression: no.
Inclusion criteria:
1. Liver transplantation for HCV infection.
Exclusion criteria:
1. Early transplant deaths.

Interventions

The participants were randomly assigned to 1 of 2 groups.
Group 1: ribavirin (n = 11).
Further details: 400 to 600 mg/day orally.
Group 2: control (n = 8).

Timing of commencement of treatment: as soon as patients could tolerate food.

Outcomes

Graft rejection and adverse effects of drug.

Notes

Reasons for post‐randomisation drop‐out: initially 37 patients were randomised to 1 of 3 groups. Only 2 groups were included for this review. Overall, 7 patients died. It is not clear how many died in each group. Hence, these data could not be used in our review.

Attempted to contact the authors in September 2010. No replies were received.

Consent for liver donation: not stated.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: this information was not available.

Allocation concealment (selection bias)

Unclear risk

Comment: this information was not available.

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Comment: this information was not available.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Comment: post‐randomisation drop‐outs could be related to the outcomes.

Selective reporting (reporting bias)

High risk

Comment: important outcomes such as mortality and retransplantation were not reported.

Free from source of funding bias?

Unclear risk

Comment: this information was not available.

Chalasani 2005

Methods

Randomised clinical trial.

Participants

Country: USA.
Sample size: 54.
Post‐randomisation drop‐out: 0 (0%).
Revised sample size: 54.
Females: 11 (20.4%).
Mean age: 53 years.
Genotype 1 (intervention): 19 (73.1%).
Genotype 1 (control): 21 (75%).
Growth factors for bone marrow suppression: no.
Inclusion criteria:
1. HCV infected liver transplant recipients.
2. Alanine aminotransferase > 1.5 times before liver transplantation.
3. No histological evidence of rejection at 3 weeks after transplantation.
Exclusion criteria:
1. Prior interferon therapy.
2. Neutrophil count 1500/μL; haemoglobin < 10 g/dL.
3. Creatinine > 2.0 mg/dL.
4. Cirrhosis.
5. Cholestatic fibrosing hepatitis.
6. Uncontrolled epilepsy.
7. Alcohol or drug abuse within 1 year of entry.
8. Severe psychiatric illness.
9. Immune disorder.
10. Chronic obstructive pulmonary disease.
11. Cardiac disease.
12. Poorly controlled thyroid disease.

Interventions

The participants were randomly assigned to 1 of 2 groups.
Group 1: pegylated interferon alpha 2a (n = 26).
Further details: 180 μg/weekly SC for 48 weeks.
Group 2: control (n = 28).

Timing of commencement of treatment: 3 weeks after liver transplantation.

Outcomes

Mortality, graft rejection, and adverse effects.

Notes

Attempted to contact the authors in September 2010. No replies were received.

Consent for liver donation: not stated.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: this information was not available.

Allocation concealment (selection bias)

Unclear risk

Comment: this information was not available.

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Comment: this information was not available.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Comment: fibrosis and activity scores reported only in 12 patients in the intervention group and 11 patients in the control group.

Selective reporting (reporting bias)

High risk

Comment: important outcomes such as retransplantation were not reported.

Free from source of funding bias?

High risk

Quote: "supported by a grant from Roche Laboratories Inc., Nutley, NJ".

Charlton 2007

Methods

Randomised clinical trial.

Participants

Country: USA.
Sample size: 115.
Post‐randomisation drop‐out: 0 (0%).
Revised sample size: 115.
Females: 22 (19.1%).
Mean age: 59 years.
Genotype 1 (intervention): 43 (78.2%).
Genotype 1 (control): 48 (80%).
Growth factors for bone marrow suppression: no.
Inclusion criteria:
1. Patients at 10 to 26 weeks of liver transplantation without histological recurrence.

Interventions

The participants were randomly assigned to 1 of 2 groups.
Group 1: pegylated interferon alpha 2a plus ribavirin (n = 54).
Further details: interferon: 135 μg/week for 4 weeks followed by 180 μg/week for 44 weeks; ribavirin: 400 mg/day escalating to 1200 mg/day for 48 weeks.
Group 2: control (n = 48).

Timing of commencement of treatment: 10 to 26 weeks after liver transplantation.

Outcomes

Mortality, graft rejection, and adverse effects.

Notes

Reason for post‐randomisation drop‐outs: not stated.

Attempted to contact the authors in September 2010. No replies were received.

Consent for liver donation: not stated.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: this information was not available.

Allocation concealment (selection bias)

Unclear risk

Comment: this information was not available.

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Comment: this information was not available.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Comment: post‐randomisation dropouts could be related to outcomes.

Selective reporting (reporting bias)

High risk

Comment: important outcomes such as retransplantation were not reported.

Free from source of funding bias?

High risk

Quote: "Grant/Research Support: Roche".

Chung 2013

Methods

Randomised clinical trial.

Participants

Country: USA.
Number randomised: 13.
Post‐randomisation drop‐outs: 2 (15.4%).
Revised sample size: 11.
Females: 2 (18.2%).

Mean age: 59 years.
Genotype 1 (intervention): 6 (100%).
Genotype 1 (control): 5 (100%).
Growth factors for bone marrow suppression: not applicable.
Inclusion criteria:
1. ≥ 18 years old.
2. HCV genotype 1a‐infected.
3. Undergoing liver transplantation from deceased or living‐related donors.
Exclusion criteria:
1. HIV or HBV co‐infection.
2. Antiviral drugs or immunoglobulin within 90 days.
3. Hepatocellular carcinoma outside the Milan criteria.
4. Personal or family history of deep venous thrombosis or pulmonary embolism.
5. Creatinine > 2.5 mg/dL for ≥ 6 months.
6. Retransplantation or planned combined organ transplant.
7. Receipt of an allograft donated after cardiac death or from an HBV‐ or HCV‐infected donor. 

Interventions

The participants were randomly assigned to 1 of 2 groups.
Group 1: human monoclonal antibody (MBL‐HCV1) (n = 6).
Further details: 50 mg/kg; 3 infusions on the day of liver transplantation (1‐4 hours before anhepatic phase), during anhepatic phase, and 8 hours post‐reperfusion; daily infusions between day 1 and day 7 post‐liver transplant; and final infusion on day 14 post‐liver transplantation.
Group 2: placebo (n = 5).
Further details: normal saline.

Timing of commencement of treatment: day of liver transplantation.

Outcomes

Mortality and adverse effects.

Notes

Reasons for post‐randomisation drop‐outs: did not undergo transplantation (n = 1); did not receive treatment (n = 1).

Attempted to contact the authors in February 2013. No replies were received.

Consent for liver donation: not stated. The study report stated that ethical approval was obtained for the research.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: this information was not available.

Allocation concealment (selection bias)

Unclear risk

Comment: this information was not available.

Blinding (performance bias and detection bias)
All outcomes

Low risk

Quote: "This randomized, double‐blind, placebo‐controlled trial was conducted at eight transplant centers between August 2010 and June 2011….The study sponsor, investigators, subjects and laboratory personnel were blinded to treatment assignment".

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Comment: there were post‐randomisation drop‐outs.

Selective reporting (reporting bias)

High risk

Comment: important outcomes such as retransplantation were not reported.

Free from source of funding bias?

High risk

Quote: "This study was funded by MassBiologics, University of Massachusetts, Boston, MA. CTSA grant (UL1TR000067) awarded to Mt. Sinai School of Medicine for CRC research support. RTC was supported in part by NIH DK078772".

Davis 2005

Methods

Randomised clinical trial.

Participants

Country: USA.
Sample size: 18.
Post‐randomisation drop‐out: 0 (0%).
Revised sample size: 18.
Females: 7 (38.9%).
Mean age: 54 years.
Genotype 1 (intervention 1): 5 (83.3%).
Genotype 1 (intervention 2): 4 (66.7%).
Genotype 1 (control): 5 (83.3%).
Growth factors for bone marrow suppression: no.
Inclusion criteria:
1. Adults with decompensated chronic hepatitis C who were listed for liver transplant.
Exclusion criteria:
1. Immunoglobulin A deficient.
2. History of prior adverse reactions to immune globulin.
3. Hepatitis B surface antigen or anti‐HIV‐positive.
4. Extrahepatic malignancy.
5. Receiving cancer chemotherapy.
6. Renal insufficiency.
7. Previously received an organ transplant.
8. Received a graft from an HCV‐positive donor.
9. Received any antiviral agents for hepatitis C in the previous 3 months.

Interventions

The participants were randomly assigned to 1 of 3 groups.
Group 1: human hepatitis C antibody‐enriched immune globulin (n = 6).
Further details: 17 intravenous infusions of 200 mg/kg starting at the time of reperfusion of graft until 14 weeks after transplant.
Group 2: human hepatitis C antibody‐enriched immune globulin (n = 6).
Further details: 17 intravenous infusions of 75 mg/kg starting at the time of reperfusion of graft until 14 weeks after transplant.
Group 3: control (n = 6).

Timing of commencement of treatment: reperfusion of graft during liver transplantation.

Outcomes

Mortality, retransplantation, and fibrosis worsening.

Notes

Attempted to contact the authors in September 2010. Authors provided replies related to randomisation.

Consent for liver donation: not stated. The study report stated that ethical approval was obtained for the research.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "The randomization sequence was computer generated" (author replies).

Allocation concealment (selection bias)

Low risk

Quote: "Each site held a sequence in sealed envelopes held by the pharmacy" (author replies).

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Comment: this information was not available.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: there were no post‐randomisation drop‐outs.

Selective reporting (reporting bias)

Low risk

Comment: important outcomes such as mortality and retransplantation were reported.

Free from source of funding bias?

High risk

Quote: "C.V.S. (one of the authors) is an employee of Nabi Biopharmaceuticals".

Mazzaferro 2003

Methods

Randomised clinical trial.

Participants

Country: Italy.
Sample size: 63.
Post‐randomisation drop‐out: 0 (0%).
Revised sample size: 63.
Females: 15 (23.8%).
Mean age: 53 years.
Genotype 1 (intervention): not stated.
Genotype 1 (control): not stated.
Overall genotype 1 (individual groups not stated): 42 (66.7%).
Growth factors for bone marrow suppression: no.
Inclusion criteria:
1. Liver transplant recipients within 4 weeks after transplant for HCV cirrhosis.
Exclusion criteria:
1. Recurrent hepatitis.
2. Rejection.

Interventions

The participants were randomly assigned to 1 of 3 groups.
Group 1: ribavirin plus interferon (n = 22).
Further details: ribavirin 10 mg/kg/day; interferon 3 million units IM 3 times weekly (duration not stated).
Group 2: interferon (n = 21).
Further details: 3 million units IM 3 times weekly (duration not stated).
Group 3: control (n = 20).

Timing of commencement of treatment: within 4 weeks after liver transplantation.

Outcomes

Graft rejection.

Notes

Attempted to contact the authors in September 2010. No replies were received.

Consent for liver donation: not stated.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: this information was not available.

Allocation concealment (selection bias)

Unclear risk

Comment: this information was not available.

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Comment: this information was not available.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: there were no post‐randomisation drop‐outs.

Selective reporting (reporting bias)

High risk

Comment: important outcomes such as mortality and retransplantation were not reported.

Free from source of funding bias?

Unclear risk

Comment: this information was not available.

Reddy 2002

Methods

Randomised clinical trial.

Participants

Country: USA.
Sample size: 32.
Post‐randomisation drop‐out: not stated.
Revised sample size: 32.
Females: 13 (40.6%).
Mean age: 50 years.
Genotype 1 (intervention): not stated.
Genotype 1 (control): not stated.
Growth factors for bone marrow suppression: no.
Inclusion criteria:
1. Liver transplantation for HCV infection.

Interventions

The participants were randomly assigned to 1 of 2 groups.
Group 1: interferon alpha 2b plus ribavirin (n = 21).
Further details: interferon: 1.5 million units increased to 3 million units 3 times weekly SC; ribavirin 400 mg increased to 1000 mg/day.
Group 2: control (n = 11).

Timing of commencement of treatment: 2‐4 weeks after liver transplantation.

Outcomes

Adverse effects.

Notes

Attempted to contact the authors in September 2010. No replies were received.

Consent for liver donation: not stated.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: this information was not available.

Allocation concealment (selection bias)

Unclear risk

Comment: this information was not available.

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Comment: this information was not available.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Comment: this information was not available.

Selective reporting (reporting bias)

High risk

Comment: important outcomes such as mortality and retransplantation were not reported.

Free from source of funding bias?

Unclear risk

Comment: this information was not available.

Schiano 2006

Methods

Randomised clinical trial.

Participants

Country: USA/ Israel.
Sample size: 24.
Post‐randomisation drop‐out: 0 (0%).
Revised sample size: 24.
Females: 1 (4.2%).
Mean age: 50.8 years.
Genotype 1 (intervention 1): 7 (87.5%).
Genotype 1 (intervention 2): 9 (81.8%).
Genotype 1 (control): 5 (100%).
Growth factors for bone marrow suppression: no.
Inclusion criteria:
1. 18‐65 years of age.
2. Primary liver transplantation for HCV infection.
3. HCV RNA positive before liver transplantation by HCV RNA concentration above the local limit of detection within 1 year.
4. Acceptable methods of contraception.
Exclusion criteria:
1. Previous liver transplant recipients.
2. Pregnant or breast‐feeding.
3. HIV infection or chronic HBV infection within 1 year.
4. Concurrent transplantation in addition to liver transplantation.

Interventions

The participants were randomly assigned to 1 of 3 groups.
Group 1: HCV‐antibody 68 (high dose) (n = 8).
Further details: 120 or 240 or 480 mg intravenous starting from the anhepatic phase until 8 weeks after transplantation.
Group 2: HCV‐antibody 68 (low dose) (n = 11).
Further details: 20 or 40 or 80 mg intravenous starting from the anhepatic phase until 8 weeks after transplantation.
Group 3: placebo (n = 5).

Timing of commencement of treatment: anhepatic phase of liver transplantation.

Outcomes

Mortality.

Notes

Attempted to contact the authors in September 2010. No replies were received.

Consent for liver donation: not stated. The study report stated that ethical approval was obtained for the research.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: this information was not available.

Allocation concealment (selection bias)

Unclear risk

Comment: this information was not available.

Blinding (performance bias and detection bias)
All outcomes

Low risk

Quote: "This was a multicenter, randomised, double‐blind, placebo‐controlled, dose‐escalation trial".

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: there were no post‐randomisation drop‐outs.

Selective reporting (reporting bias)

High risk

Comment: important outcomes such as retransplantation were not reported.

Free from source of funding bias?

High risk

Quote: "Supported by XTL Biopharmaceuticals Ltd., the developer of HCV‐AbXTL68".

Sheiner 1998

Methods

Randomised clinical trial.

Participants

Country: USA, Israel.
Sample size: 86.
Post‐randomisation drop‐out: 5 (5.8%).
Revised sample size: 81.
Females: not stated.
Mean age: not stated.
Genotype 1 (intervention): 21 (60.0%).
Genotype 1 (control): 19 (41.3%).
Growth factors for bone marrow suppression: no.
Inclusion criteria:
1. Liver transplantation with positive serology for hepatitis C antibody.
2. Age > 18 years.
Exclusion criteria:
1. Retransplantation for recurrent hepatitis C.
2. Transplantation for hepatitis C with hepatocellular carcinoma requiring adjuvant chemotherapy (ie, tumour 0.5 cm or with vascular invasion).
3. Positive serology for hepatitis B surface antigen.
4. Platelet count < 50,000/mm3by day 14 after transplantation.

Interventions

The participants were randomly assigned to 1 of 2 groups.
Group 1: interferon alpha 2b (n = 35).
Further details: 3 million units SC 3 times weekly for 52 weeks.
Group 2: control (n = 46).

Timing of commencement of treatment: within 2 weeks after liver transplantation.

Outcomes

Mortality, retransplantation, graft rejection, and recurrence of viral hepatitis.

Notes

Reasons for post‐randomisation drop‐out: low white cell count (n = 3); adjuvant chemotherapy (n = 1); refusal to enter (n = 1).

Attempted to contact the authors in September 2010. No replies were received.

Consent for liver donation: not stated. The study report stated that ethical approval was obtained for the research.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: this information was not available.

Allocation concealment (selection bias)

Unclear risk

Comment: this information was not available.

Blinding (performance bias and detection bias)
All outcomes

High risk

Quote: "Protocol biopsies were also reviewed by three pathologists blinded to study group".
Comment: the other outcomes were not assessed by blinded observers.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Comment: post‐randomisation drop‐outs could be related to the outcomes.

Selective reporting (reporting bias)

Low risk

Comment: important outcomes such as mortality and retransplantation were reported.

Free from source of funding bias?

High risk

Quote: "Supported in part by Ortho Biotech, Raritan, NJ".

Shergill 2005

Methods

Randomised clinical trial.

Participants

Country: USA.
Sample size: 44.
Post‐randomisation drop‐out: 0 (0%).
Revised sample size: 44.
Females: not stated.
Mean age: not stated.
Genotype 1 (intervention): not stated.
Genotype 1 (control): not stated.
Growth factors for bone marrow suppression: yes.
Inclusion criteria:
1. ≥ 18 years of age.
2. Pretransplant diagnosis of HCV‐associated cirrhosis.
3. Clinical stability with white cell count > 3.0/mm3.
4. Haemoglobin > 10.0 g/dL.
5. Platelets > 45,000/mm3.
6. Creatinine < 1.5 mg/dL.
7. International Normalised Ratio < 2.0.
8. Aspartate aminotransferase < 200 IU.
9. Alanine aminotransferase < 200 IU.
10. Total bilirubin < 2.5 mg/dL.
Exclusion criteria:
1. Serum anti‐HIV or hepatitis B surface antigen positive prior to transplantation.
2. Acute rejection (by clinical and histological criteria) within 14 days of consent.
3. Vascular and biliary complications post‐transplantation resulting in need for interventions.
4. Current untreated infection.
5. Abnormal thyroid‐stimulating hormone.
6. Medically uncontrolled psychosis or depression.
7. History of haemoglobinopathies or any cause of chronic haemolysis.
8. Clinically significant retinopathy.
9. Uncontrolled diabetes mellitus.
10. Inability to practice effective contraception (male or female) during the treatment period.
11. Medical history of concomitant autoimmune disease.
12. Continued need for intensive care unit support or unstable cardiopulmonary status including myocardial infarction within preceding 4 weeks.

Interventions

The participants were randomly assigned to 1 of 2 groups.
Group 1: ribavirin plus interferon (n = 22).
Further details: ribavirin: 400 mg escalated to 1000 to 1200 mg for a total period of 48 weeks; interferon: interferon alpha 2b or pegylated interferon alpha 2b (initial patients treated with interferon and later patients treated with pegylated interferon). Interferon started at 1.5 million units daily and increased to 3 million units daily after 2 weeks if tolerated. After 8 weeks, 3 million units 3 times weekly. Pegylated interferon: 1.5 μg/kg/week.
Group 2: interferon (n = 22).

Timing of commencement of treatment: 2 to 6 weeks after liver transplantation.

Outcomes

None of the outcomes of interest for this review were reported in this trial.

Notes

Attempted to contact the authors in September 2010. No replies were received.

Consent for liver donation: not stated.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: this information was not available.

Allocation concealment (selection bias)

Unclear risk

Comment: this information was not available.

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Comment: this information was not available.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: there were no post‐randomisation drop‐outs.

Selective reporting (reporting bias)

High risk

Comment: important outcomes such as mortality and retransplantation were not reported.

Free from source of funding bias?

High risk

Quote: "The study was supported by Fugisawa Healthcare, Inc., and the UCSF Liver Center P30 DK26743 Clinical and Translational Core".

Singh 1998

Methods

Randomised clinical trial.

Participants

Country: USA.
Sample size: 24.
Post‐randomisation drop‐out: 0 (0%).
Revised sample size: 24.
Females: 0 (0%).
Mean age: 46 years.
Genotype 1 (intervention): 9 (75.0%).
Genotype 1 (control): 11 (91.7%).
Growth factors for bone marrow suppression: no.
Inclusion criteria:
1. Liver transplantation for end‐stage liver disease due to HCV.

Interventions

The participants were randomly assigned to 1 of 2 groups.
Group 1: interferon alpha (n = 12).
Further details: 3 million units 3 times weekly for 6 months.
Group 2: control (n = 12).

Timing of commencement of treatment: 2 weeks after liver transplantation.

Outcomes

Mortality, retransplantation, graft rejection, and recurrence.

Notes

Attempted to contact the authors in September 2010. No replies were received.

Consent for liver donation: not stated. The study report stated that ethical approval was obtained for the research.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: this information was not available.

Allocation concealment (selection bias)

Unclear risk

Comment: this information was not available.

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Comment: this information was not available.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: there were no post‐randomisation drop‐outs.

Selective reporting (reporting bias)

Low risk

Comment: important outcomes such as mortality and retransplantation were reported.

Free from source of funding bias?

Unclear risk

Comment: this information was not available.

Willems 2002

Methods

Randomised clinical trial.

Participants

Country: Canada.
Sample size: 16.
Post‐randomisation drop‐out: not stated.
Revised sample size: 16.
Females: not stated.
Mean age: not stated.
Genotype 1 (intervention): not stated.
Genotype 1 (control): not stated.
Growth factors for bone marrow suppression: no.
Inclusion criteria:
1. Patients undergoing liver transplantation for end‐stage post‐HCV cirrhosis.

Interventions

The participants were randomly assigned to 1 of 3 groups.
Group 1: human hepatitis C antibody‐enriched immune globulin (n = 6).
Further details: starting at anhepatic phase with 1500 mg and maintained at 250 mg twice weekly for 48 weeks.
Group 2: human hepatitis C antibody‐enriched immune globulin (n = 3).
Further details: starting at anhepatic phase with 500 mg and maintained at 83 mg twice weekly for 48 weeks.
Group 3: placebo (n = 7).

Timing of commencement of treatment: anhepatic phase of liver transplantation.

Outcomes

None of the outcomes of interest for this review were reported in this trial.

Notes

Attempted to contact the authors in September 2010. No replies were received.

Consent for liver donation: not stated.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: this information was not available.

Allocation concealment (selection bias)

Unclear risk

Comment: this information was not available.

Blinding (performance bias and detection bias)
All outcomes

Low risk

Quote: "Twenty‐six HCV‐RNA positive OLT [orthotopic liver transplantation] candidates were randomly assigned at the time of transplantation to one of three treatment schedules in a double‐blind fashion".

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Comment: this information was not available.

Selective reporting (reporting bias)

High risk

Comment: important outcomes such as mortality and retransplantation were not reported.

Free from source of funding bias?

Unclear risk

Comment: this information was not available.

HBV: hepatitis B virus; HCV: hepatitis C virus; HIV: human immunodeficiency virus; IM: intramuscular; RNA: ribonucleic acid; SC: subcutaneous.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Beckebaum 2003

Not a randomised clinical trial.

Boillot 1995

Not a randomised clinical trial.

Casanovas 2004

Comment on a trial of antiviral therapy in patients with recurrent hepatitis C virus infection after liver transplantation.

Castedal 2003

Not a randomised clinical trial.

Catalano 2003

Not a randomised clinical trial.

Ceccherini 2003

Not a randomised clinical trial.

Charlton 2002

Editorial.

Everson 2013

No separate data for patients involved in the randomised clinical trial.

Garcia‐Retortillo 2004

Review.

Mazzaferro 1997

Not a randomised clinical trial.

Roche 2011

Editorial.

Samuel 2004a

Editorial.

Samuel 2004b

Comments on trial in patients with recurrent hepatitis C virus infection after liver transplantation.

Taltavull 2004

Comment on a trial of antiviral therapy in patients with recurrent hepatitis C virus infection after liver transplantation.

Data and analyses

Open in table viewer
Comparison 1. Intervention versus control

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 90‐day mortality Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.1

Comparison 1 Intervention versus control, Outcome 1 90‐day mortality.

Comparison 1 Intervention versus control, Outcome 1 90‐day mortality.

1.1 Interferon vs. no intervention

1

81

Risk Ratio (M‐H, Fixed, 95% CI)

1.31 [0.41, 4.19]

1.2 Pegylated interferon plus ribavirin vs. no intervention

1

115

Risk Ratio (M‐H, Fixed, 95% CI)

1.82 [0.46, 7.25]

1.3 HCV antibody vs. placebo

3

53

Risk Ratio (M‐H, Fixed, 95% CI)

0.69 [0.15, 3.11]

1.4 HCV antibody (high dose) vs. HCV antibody (low dose)

2

31

Risk Ratio (M‐H, Fixed, 95% CI)

2.75 [0.30, 25.35]

2 Mortality at maximal follow‐up Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.2

Comparison 1 Intervention versus control, Outcome 2 Mortality at maximal follow‐up.

Comparison 1 Intervention versus control, Outcome 2 Mortality at maximal follow‐up.

2.1 Interferon vs. no intervention

2

105

Risk Ratio (M‐H, Fixed, 95% CI)

0.86 [0.36, 2.08]

2.2 Pegylated interferon vs. no intervention

1

54

Risk Ratio (M‐H, Fixed, 95% CI)

0.54 [0.05, 5.59]

3 Mortality (hazard ratio) Show forest plot

1

Hazard Ratio (Fixed, 95% CI)

Subtotals only

Analysis 1.3

Comparison 1 Intervention versus control, Outcome 3 Mortality (hazard ratio).

Comparison 1 Intervention versus control, Outcome 3 Mortality (hazard ratio).

3.1 Interferon vs. no intervention

1

Hazard Ratio (Fixed, 95% CI)

0.45 [0.13, 1.56]

4 90‐day retransplantation Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.4

Comparison 1 Intervention versus control, Outcome 4 90‐day retransplantation.

Comparison 1 Intervention versus control, Outcome 4 90‐day retransplantation.

4.1 HCV antibody vs. placebo

1

18

Risk Ratio (M‐H, Fixed, 95% CI)

1.71 [0.09, 32.93]

4.2 HCV antibody (high dose) vs. HCV antibody (low dose)

1

12

Risk Ratio (M‐H, Fixed, 95% CI)

3.0 [0.15, 61.74]

5 Retransplantation at maximal follow‐up Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.5

Comparison 1 Intervention versus control, Outcome 5 Retransplantation at maximal follow‐up.

Comparison 1 Intervention versus control, Outcome 5 Retransplantation at maximal follow‐up.

5.1 Interferon vs. no intervention

2

105

Risk Ratio (M‐H, Fixed, 95% CI)

1.17 [0.22, 6.20]

6 Serious adverse events Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.6

Comparison 1 Intervention versus control, Outcome 6 Serious adverse events.

Comparison 1 Intervention versus control, Outcome 6 Serious adverse events.

6.1 Pegylated interferon vs. no intervention

1

54

Risk Ratio (M‐H, Fixed, 95% CI)

0.97 [0.47, 2.00]

6.2 Pegylated interferon plus ribavirin vs. no intervention

1

115

Risk Ratio (M‐H, Fixed, 95% CI)

1.79 [1.03, 3.12]

6.3 HCV antibody vs. placebo

1

11

Risk Ratio (M‐H, Fixed, 95% CI)

0.21 [0.03, 1.31]

7 Anaemia Show forest plot

4

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.7

Comparison 1 Intervention versus control, Outcome 7 Anaemia.

Comparison 1 Intervention versus control, Outcome 7 Anaemia.

7.1 Interferon plus ribavirin vs. no intervention

1

32

Risk Ratio (M‐H, Fixed, 95% CI)

13.64 [0.88, 210.72]

7.2 Pegylated interferon vs. no intervention

1

54

Risk Ratio (M‐H, Fixed, 95% CI)

0.43 [0.09, 2.03]

7.3 Pegylated interferon plus ribavirin vs. no intervention

1

100

Risk Ratio (M‐H, Fixed, 95% CI)

11.07 [1.51, 81.47]

7.4 Ribavirin vs. no intervention

1

19

Risk Ratio (M‐H, Fixed, 95% CI)

6.75 [0.41, 110.01]

8 Leukopenia Show forest plot

4

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.8

Comparison 1 Intervention versus control, Outcome 8 Leukopenia.

Comparison 1 Intervention versus control, Outcome 8 Leukopenia.

8.1 Interferon vs. no intervention

1

24

Risk Ratio (M‐H, Fixed, 95% CI)

5.0 [0.27, 94.34]

8.2 Pegylated interferon vs. no intervention

1

54

Risk Ratio (M‐H, Fixed, 95% CI)

1.29 [0.45, 3.73]

8.3 Pegylated interferon plus ribavirin vs. no intervention

1

100

Risk Ratio (M‐H, Fixed, 95% CI)

3.98 [1.22, 12.98]

8.4 HCV monoclonal antibody vs. placebo

1

11

Risk Ratio (M‐H, Fixed, 95% CI)

0.21 [0.03, 1.31]

9 Graft rejection requiring retransplantation Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.9

Comparison 1 Intervention versus control, Outcome 9 Graft rejection requiring retransplantation.

Comparison 1 Intervention versus control, Outcome 9 Graft rejection requiring retransplantation.

9.1 Interferon vs. no intervention

1

31

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.2 Interferon plus ribavirin vs. no intervention

1

32

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.3 Ribavirin plus interferon vs. interferon

1

43

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10 Graft rejection requiring steroids or equivalent drugs Show forest plot

4

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.10

Comparison 1 Intervention versus control, Outcome 10 Graft rejection requiring steroids or equivalent drugs.

Comparison 1 Intervention versus control, Outcome 10 Graft rejection requiring steroids or equivalent drugs.

10.1 Interferon vs. no intervention

3

136

Risk Ratio (M‐H, Fixed, 95% CI)

1.02 [0.68, 1.51]

10.2 Interferon plus ribavirin vs. no intervention

1

32

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.3 Pegylated interferon vs. no intervention

1

54

Risk Ratio (M‐H, Fixed, 95% CI)

0.81 [0.20, 3.27]

10.4 Ribavirin plus interferon vs. interferon

1

43

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11 Graft rejection (others) Show forest plot

4

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.11

Comparison 1 Intervention versus control, Outcome 11 Graft rejection (others).

Comparison 1 Intervention versus control, Outcome 11 Graft rejection (others).

11.1 Interferon vs. no intervention

1

31

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.2 Interferon plus ribavirin vs. no intervention

1

32

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.3 Pegylated interferon vs. no intervention

1

54

Risk Ratio (M‐H, Fixed, 95% CI)

0.21 [0.01, 4.28]

11.4 Pegylated interferon plus ribavirin vs. no intervention

1

115

Risk Ratio (M‐H, Fixed, 95% CI)

1.64 [0.49, 5.49]

11.5 Ribavirin vs. no intervention

1

19

Risk Ratio (M‐H, Fixed, 95% CI)

1.09 [0.23, 5.09]

11.6 Ribavirin plus interferon vs. interferon

1

43

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12 Fibrosis worsening Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.12

Comparison 1 Intervention versus control, Outcome 12 Fibrosis worsening.

Comparison 1 Intervention versus control, Outcome 12 Fibrosis worsening.

12.1 HCV antibody vs. placebo

1

18

Risk Ratio (M‐H, Fixed, 95% CI)

0.86 [0.17, 4.43]

12.2 HCV antibody (high dose) vs. HCV antibody (low dose)

1

12

Risk Ratio (M‐H, Fixed, 95% CI)

0.33 [0.02, 6.86]

13 Recurrence (hazard ratio) Show forest plot

2

Hazard Ratio (Fixed, 95% CI)

Subtotals only

Analysis 1.13

Comparison 1 Intervention versus control, Outcome 13 Recurrence (hazard ratio).

Comparison 1 Intervention versus control, Outcome 13 Recurrence (hazard ratio).

13.1 Interferon vs. no intervention

2

Hazard Ratio (Fixed, 95% CI)

0.87 [0.61, 1.24]

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.

Trial sequential analysis of 90‐day mortality (hepatitis C virus antibody versus placebo) 
 The diversity‐adjusted required information size (DARIS) was calculated with 10,577 patients, based on the proportion of patients in the control group with the outcome of 6.3%, a relative risk reduction of 20%, an alpha of 5%, a beta of 20%, and a diversity of 0%. To account for zero event groups, a continuity correction of 0.01 was used in the calculation of the cumulative Z‐curve (blue line). After accruing 53 participants in three trials, only 0.5% of the DARIS was reached. Accordingly, the trial sequential analysis does not show the required information size and the trial sequential monitoring boundaries. As shown, the conventional boundaries (dotted red line) have also not been crossed by the cumulative Z‐curve.
Figuras y tablas -
Figure 4

Trial sequential analysis of 90‐day mortality (hepatitis C virus antibody versus placebo)
The diversity‐adjusted required information size (DARIS) was calculated with 10,577 patients, based on the proportion of patients in the control group with the outcome of 6.3%, a relative risk reduction of 20%, an alpha of 5%, a beta of 20%, and a diversity of 0%. To account for zero event groups, a continuity correction of 0.01 was used in the calculation of the cumulative Z‐curve (blue line). After accruing 53 participants in three trials, only 0.5% of the DARIS was reached. Accordingly, the trial sequential analysis does not show the required information size and the trial sequential monitoring boundaries. As shown, the conventional boundaries (dotted red line) have also not been crossed by the cumulative Z‐curve.

Trial sequential analysis of 90‐day mortality (hepatitis C virus antibody (high dose) versus hepatitis C virus antibody (low dose)) 
 The diversity‐adjusted required information size (DARIS) was calculated with 11,338 patients, based on the proportion of patients in the control group with the outcome of 5.9%, a relative risk reduction of 20%, an alpha of 5%, a beta of 20%, and a diversity of 0%. To account for zero event groups, a continuity correction of 0.01 was used in the calculation of the cumulative Z‐curve (blue line). After accruing 31 participants in two trials, only 0.27% of the DARIS has been reached. Accordingly, the trial sequential analysis does not show the required information size and the trial sequential monitoring boundaries. As shown, the conventional boundaries (dotted red line) have also not been crossed by the cumulative Z‐curve.
Figuras y tablas -
Figure 5

Trial sequential analysis of 90‐day mortality (hepatitis C virus antibody (high dose) versus hepatitis C virus antibody (low dose))
The diversity‐adjusted required information size (DARIS) was calculated with 11,338 patients, based on the proportion of patients in the control group with the outcome of 5.9%, a relative risk reduction of 20%, an alpha of 5%, a beta of 20%, and a diversity of 0%. To account for zero event groups, a continuity correction of 0.01 was used in the calculation of the cumulative Z‐curve (blue line). After accruing 31 participants in two trials, only 0.27% of the DARIS has been reached. Accordingly, the trial sequential analysis does not show the required information size and the trial sequential monitoring boundaries. As shown, the conventional boundaries (dotted red line) have also not been crossed by the cumulative Z‐curve.

Trial sequential analysis of mortality at maximal follow‐up (interferon versus no intervention) 
 The diversity‐adjusted required information size (DARIS) was calculated with 3796 patients, based on the proportion of patients in the control no intervention group with the outcome of 17.2%, a relative risk reduction of 20%, an alpha of 5%, a beta of 20%, and a diversity of 8.54%. To account for zero event groups, a continuity correction of 0.01 was used in the calculation of the cumulative Z‐curve (blue line). After accruing 105 participants in two trials, only 2.77% of the DARIS has been reached. Accordingly, the trial sequential analysis does not show the required information size and the trial sequential monitoring boundaries. As shown, the conventional boundaries (dotted red line) have also not been crossed by the cumulative Z‐curve.
Figuras y tablas -
Figure 6

Trial sequential analysis of mortality at maximal follow‐up (interferon versus no intervention)
The diversity‐adjusted required information size (DARIS) was calculated with 3796 patients, based on the proportion of patients in the control no intervention group with the outcome of 17.2%, a relative risk reduction of 20%, an alpha of 5%, a beta of 20%, and a diversity of 8.54%. To account for zero event groups, a continuity correction of 0.01 was used in the calculation of the cumulative Z‐curve (blue line). After accruing 105 participants in two trials, only 2.77% of the DARIS has been reached. Accordingly, the trial sequential analysis does not show the required information size and the trial sequential monitoring boundaries. As shown, the conventional boundaries (dotted red line) have also not been crossed by the cumulative Z‐curve.

Trial sequential analysis of retransplantation at maximal follow‐up (interferon versus no intervention) 
 The diversity‐adjusted required information size (DARIS) was calculated with 20,141 patients, based on the proportion of patients in the no intervention control group with the outcome of 3.4%, a relative risk reduction of 20%, an alpha of 5%, a beta of 20%, and a diversity of 8.54%. To account for zero event groups, a continuity correction of 0.01 was used in the calculation of the cumulative Z‐curve (blue line). After accruing 105 participants in two trials, only 0.52% of the DARIS has been reached. Accordingly, the trial sequential analysis does not show the required information size and the trial sequential monitoring boundaries. As shown, the conventional boundaries (dotted red line) have also not been crossed by the cumulative Z‐curve.
Figuras y tablas -
Figure 7

Trial sequential analysis of retransplantation at maximal follow‐up (interferon versus no intervention)
The diversity‐adjusted required information size (DARIS) was calculated with 20,141 patients, based on the proportion of patients in the no intervention control group with the outcome of 3.4%, a relative risk reduction of 20%, an alpha of 5%, a beta of 20%, and a diversity of 8.54%. To account for zero event groups, a continuity correction of 0.01 was used in the calculation of the cumulative Z‐curve (blue line). After accruing 105 participants in two trials, only 0.52% of the DARIS has been reached. Accordingly, the trial sequential analysis does not show the required information size and the trial sequential monitoring boundaries. As shown, the conventional boundaries (dotted red line) have also not been crossed by the cumulative Z‐curve.

Trial sequential analysis of graft rejection requiring steroids or equivalent drugs (interferon versus no intervention) 
 The diversity‐adjusted required information size (DARIS) was calculated with 1079 patients, based on the proportion of patients in the no intervention control group with the outcome of 41.2%, a relative risk reduction of 20%, an alpha of 5%, a beta of 20%, and a diversity of 8.54%. To account for zero event groups, a continuity correction of 0.01 was used in the calculation of the cumulative Z‐curve (blue line). After accruing 136 participants in three trials, only 12.60% of the DARIS has been reached. Accordingly, the trial sequential analysis does not show the futility area. Neither the trial sequential boundaries (continuous red line) nor the conventional boundaries (dotted red line) have been crossed by the cumulative Z‐curve.
Figuras y tablas -
Figure 8

Trial sequential analysis of graft rejection requiring steroids or equivalent drugs (interferon versus no intervention)
The diversity‐adjusted required information size (DARIS) was calculated with 1079 patients, based on the proportion of patients in the no intervention control group with the outcome of 41.2%, a relative risk reduction of 20%, an alpha of 5%, a beta of 20%, and a diversity of 8.54%. To account for zero event groups, a continuity correction of 0.01 was used in the calculation of the cumulative Z‐curve (blue line). After accruing 136 participants in three trials, only 12.60% of the DARIS has been reached. Accordingly, the trial sequential analysis does not show the futility area. Neither the trial sequential boundaries (continuous red line) nor the conventional boundaries (dotted red line) have been crossed by the cumulative Z‐curve.

Comparison 1 Intervention versus control, Outcome 1 90‐day mortality.
Figuras y tablas -
Analysis 1.1

Comparison 1 Intervention versus control, Outcome 1 90‐day mortality.

Comparison 1 Intervention versus control, Outcome 2 Mortality at maximal follow‐up.
Figuras y tablas -
Analysis 1.2

Comparison 1 Intervention versus control, Outcome 2 Mortality at maximal follow‐up.

Comparison 1 Intervention versus control, Outcome 3 Mortality (hazard ratio).
Figuras y tablas -
Analysis 1.3

Comparison 1 Intervention versus control, Outcome 3 Mortality (hazard ratio).

Comparison 1 Intervention versus control, Outcome 4 90‐day retransplantation.
Figuras y tablas -
Analysis 1.4

Comparison 1 Intervention versus control, Outcome 4 90‐day retransplantation.

Comparison 1 Intervention versus control, Outcome 5 Retransplantation at maximal follow‐up.
Figuras y tablas -
Analysis 1.5

Comparison 1 Intervention versus control, Outcome 5 Retransplantation at maximal follow‐up.

Comparison 1 Intervention versus control, Outcome 6 Serious adverse events.
Figuras y tablas -
Analysis 1.6

Comparison 1 Intervention versus control, Outcome 6 Serious adverse events.

Comparison 1 Intervention versus control, Outcome 7 Anaemia.
Figuras y tablas -
Analysis 1.7

Comparison 1 Intervention versus control, Outcome 7 Anaemia.

Comparison 1 Intervention versus control, Outcome 8 Leukopenia.
Figuras y tablas -
Analysis 1.8

Comparison 1 Intervention versus control, Outcome 8 Leukopenia.

Comparison 1 Intervention versus control, Outcome 9 Graft rejection requiring retransplantation.
Figuras y tablas -
Analysis 1.9

Comparison 1 Intervention versus control, Outcome 9 Graft rejection requiring retransplantation.

Comparison 1 Intervention versus control, Outcome 10 Graft rejection requiring steroids or equivalent drugs.
Figuras y tablas -
Analysis 1.10

Comparison 1 Intervention versus control, Outcome 10 Graft rejection requiring steroids or equivalent drugs.

Comparison 1 Intervention versus control, Outcome 11 Graft rejection (others).
Figuras y tablas -
Analysis 1.11

Comparison 1 Intervention versus control, Outcome 11 Graft rejection (others).

Comparison 1 Intervention versus control, Outcome 12 Fibrosis worsening.
Figuras y tablas -
Analysis 1.12

Comparison 1 Intervention versus control, Outcome 12 Fibrosis worsening.

Comparison 1 Intervention versus control, Outcome 13 Recurrence (hazard ratio).
Figuras y tablas -
Analysis 1.13

Comparison 1 Intervention versus control, Outcome 13 Recurrence (hazard ratio).

Summary of findings for the main comparison. Antiviral prophylaxis for the prevention of chronic hepatitis C virus in patients undergoing liver transplantation (mortality)

Mortality

Patient or population: patients undergoing liver transplantation for chronic hepatitis C viral infection.
Settings: tertiary.
Comparisons: shown in table.

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Assumed risk

Corresponding risk

Control

Intervention

Interferon vs. control

90‐day mortality

109 per 1000

142 per 1000
(45 to 455)

RR 1.31
(0.41 to 4.19)

81
(1 study)

⊕⊝⊝⊝
very low1,2

Mortality at maximal follow‐up

172 per 1000

148 per 1000
(62 to 359)

RR 0.86
(0.36 to 2.08)

105
(2 studies)

⊕⊝⊝⊝
very low1,2

Mortality (HR)
Follow‐up: mean 12 months

100 per 1000

46 per 1000
(14 to 152)

HR 0.45
(0.13 to 1.56)

81
(1 study)

⊕⊝⊝⊝
very low1,2

Pegylated interferon vs. control

Mortality at maximal follow‐up

71 per 1000

39 per 1000
(4 to 399)

RR 0.54
(0.05 to 5.59)

54
(1 study)

⊕⊝⊝⊝
very low1,2

Pegylated interferon plus ribavirin vs. control

90‐day mortality

50 per 1000

91 per 1000
(23 to 362)

RR 1.82
(0.46 to 7.25)

115
(1 study)

⊕⊝⊝⊝
very low1,2

HCV antibody vs. placebo

90‐day mortality

62 per 1000

43 per 1000
(9 to 194)

RR 0.69
(0.15 to 3.11)

53
(3 studies)

⊕⊝⊝⊝
very low1,2

HCV antibody (high dose) vs. HCV antibody (low dose)

90‐day mortality

59 per 1000

162 per 1000
(18 to 1000)

RR 2.75
(0.3 to 25.35)

31
(2 studies)

⊕⊝⊝⊝
very low1,2

*The basis for the assumed risk is the control group risk across studies. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: confidence interval; HCV: hepatitis C virus; RR: risk ratio; HR: hazard ratio.

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

1 The trial(s) was (were) of high risk of bias.
2 The CIs overlapped 1 and either 0.75 or 1.25, or both. The number of events in the intervention and control group was fewer than 300.

Figuras y tablas -
Summary of findings for the main comparison. Antiviral prophylaxis for the prevention of chronic hepatitis C virus in patients undergoing liver transplantation (mortality)
Summary of findings 2. Antiviral prophylaxis for the prevention of chronic hepatitis C virus in patients undergoing liver transplantation (retransplantation)

Retransplantation

Patient or population: patients undergoing liver transplantation for chronic hepatitis C viral infection.
Settings: tertiary.
Comparisons: shown in table.

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Assumed risk

Corresponding risk

Control

Intervention

Interferon vs. control

Retransplantation at maximal follow‐up

34 per 1000

40 per 1000
(8 to 214)

RR 1.17
(0.22 to 6.2)

105
(2 studies)

⊕⊝⊝⊝
very low1,2

HCV antibody vs. placebo

90‐day retransplantation

30 per 1000

51 per 1000
(3 to 988)

RR 1.71
(0.09 to 32.93)

18
(1 study)

⊕⊝⊝⊝
very low1,2

HCV antibody (high dose) vs. HCV antibody (low dose)

90‐day retransplantation

30 per 1000

90 per 1000
(5 to 1000)

RR 3
(0.15 to 61.74)

12
(1 study)

⊕⊝⊝⊝
very low1,2

*The basis for the assumed risk for interferon versus control was the control group risk in the studies. There were no events in the control group for the other two comparisons. So, the control group risk in the interferon versus control comparison was used as the assumed risk. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: confidence interval; HCV: hepatitis C virus; RR: risk ratio.

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

1 The trial(s) was (were) of high risk of bias.
2 The CIs overlapped 1 and either 0.75 or 1.25, or both. The number of events in the intervention and control group was fewer than 300.

Figuras y tablas -
Summary of findings 2. Antiviral prophylaxis for the prevention of chronic hepatitis C virus in patients undergoing liver transplantation (retransplantation)
Comparison 1. Intervention versus control

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 90‐day mortality Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

1.1 Interferon vs. no intervention

1

81

Risk Ratio (M‐H, Fixed, 95% CI)

1.31 [0.41, 4.19]

1.2 Pegylated interferon plus ribavirin vs. no intervention

1

115

Risk Ratio (M‐H, Fixed, 95% CI)

1.82 [0.46, 7.25]

1.3 HCV antibody vs. placebo

3

53

Risk Ratio (M‐H, Fixed, 95% CI)

0.69 [0.15, 3.11]

1.4 HCV antibody (high dose) vs. HCV antibody (low dose)

2

31

Risk Ratio (M‐H, Fixed, 95% CI)

2.75 [0.30, 25.35]

2 Mortality at maximal follow‐up Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

2.1 Interferon vs. no intervention

2

105

Risk Ratio (M‐H, Fixed, 95% CI)

0.86 [0.36, 2.08]

2.2 Pegylated interferon vs. no intervention

1

54

Risk Ratio (M‐H, Fixed, 95% CI)

0.54 [0.05, 5.59]

3 Mortality (hazard ratio) Show forest plot

1

Hazard Ratio (Fixed, 95% CI)

Subtotals only

3.1 Interferon vs. no intervention

1

Hazard Ratio (Fixed, 95% CI)

0.45 [0.13, 1.56]

4 90‐day retransplantation Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

4.1 HCV antibody vs. placebo

1

18

Risk Ratio (M‐H, Fixed, 95% CI)

1.71 [0.09, 32.93]

4.2 HCV antibody (high dose) vs. HCV antibody (low dose)

1

12

Risk Ratio (M‐H, Fixed, 95% CI)

3.0 [0.15, 61.74]

5 Retransplantation at maximal follow‐up Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

5.1 Interferon vs. no intervention

2

105

Risk Ratio (M‐H, Fixed, 95% CI)

1.17 [0.22, 6.20]

6 Serious adverse events Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

6.1 Pegylated interferon vs. no intervention

1

54

Risk Ratio (M‐H, Fixed, 95% CI)

0.97 [0.47, 2.00]

6.2 Pegylated interferon plus ribavirin vs. no intervention

1

115

Risk Ratio (M‐H, Fixed, 95% CI)

1.79 [1.03, 3.12]

6.3 HCV antibody vs. placebo

1

11

Risk Ratio (M‐H, Fixed, 95% CI)

0.21 [0.03, 1.31]

7 Anaemia Show forest plot

4

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

7.1 Interferon plus ribavirin vs. no intervention

1

32

Risk Ratio (M‐H, Fixed, 95% CI)

13.64 [0.88, 210.72]

7.2 Pegylated interferon vs. no intervention

1

54

Risk Ratio (M‐H, Fixed, 95% CI)

0.43 [0.09, 2.03]

7.3 Pegylated interferon plus ribavirin vs. no intervention

1

100

Risk Ratio (M‐H, Fixed, 95% CI)

11.07 [1.51, 81.47]

7.4 Ribavirin vs. no intervention

1

19

Risk Ratio (M‐H, Fixed, 95% CI)

6.75 [0.41, 110.01]

8 Leukopenia Show forest plot

4

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

8.1 Interferon vs. no intervention

1

24

Risk Ratio (M‐H, Fixed, 95% CI)

5.0 [0.27, 94.34]

8.2 Pegylated interferon vs. no intervention

1

54

Risk Ratio (M‐H, Fixed, 95% CI)

1.29 [0.45, 3.73]

8.3 Pegylated interferon plus ribavirin vs. no intervention

1

100

Risk Ratio (M‐H, Fixed, 95% CI)

3.98 [1.22, 12.98]

8.4 HCV monoclonal antibody vs. placebo

1

11

Risk Ratio (M‐H, Fixed, 95% CI)

0.21 [0.03, 1.31]

9 Graft rejection requiring retransplantation Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

9.1 Interferon vs. no intervention

1

31

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.2 Interferon plus ribavirin vs. no intervention

1

32

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.3 Ribavirin plus interferon vs. interferon

1

43

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10 Graft rejection requiring steroids or equivalent drugs Show forest plot

4

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

10.1 Interferon vs. no intervention

3

136

Risk Ratio (M‐H, Fixed, 95% CI)

1.02 [0.68, 1.51]

10.2 Interferon plus ribavirin vs. no intervention

1

32

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.3 Pegylated interferon vs. no intervention

1

54

Risk Ratio (M‐H, Fixed, 95% CI)

0.81 [0.20, 3.27]

10.4 Ribavirin plus interferon vs. interferon

1

43

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11 Graft rejection (others) Show forest plot

4

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

11.1 Interferon vs. no intervention

1

31

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.2 Interferon plus ribavirin vs. no intervention

1

32

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.3 Pegylated interferon vs. no intervention

1

54

Risk Ratio (M‐H, Fixed, 95% CI)

0.21 [0.01, 4.28]

11.4 Pegylated interferon plus ribavirin vs. no intervention

1

115

Risk Ratio (M‐H, Fixed, 95% CI)

1.64 [0.49, 5.49]

11.5 Ribavirin vs. no intervention

1

19

Risk Ratio (M‐H, Fixed, 95% CI)

1.09 [0.23, 5.09]

11.6 Ribavirin plus interferon vs. interferon

1

43

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12 Fibrosis worsening Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

12.1 HCV antibody vs. placebo

1

18

Risk Ratio (M‐H, Fixed, 95% CI)

0.86 [0.17, 4.43]

12.2 HCV antibody (high dose) vs. HCV antibody (low dose)

1

12

Risk Ratio (M‐H, Fixed, 95% CI)

0.33 [0.02, 6.86]

13 Recurrence (hazard ratio) Show forest plot

2

Hazard Ratio (Fixed, 95% CI)

Subtotals only

13.1 Interferon vs. no intervention

2

Hazard Ratio (Fixed, 95% CI)

0.87 [0.61, 1.24]

Figuras y tablas -
Comparison 1. Intervention versus control