Scolaris Content Display Scolaris Content Display

Intervenciones farmacológicas para la infección aguda por el virus de la hepatitis C

Contraer todo Desplegar todo

Referencias

References to studies included in this review

Calleri 1998 {published data only}

Calleri G, Colombatto P, Gozzelino M, Chieppa F, Romano P, Delmastro B, et al. Natural beta interferon in acute type‐C hepatitis patients: a randomized controlled trial. Italian Journal of Gastroenterology and Hepatology 1998;30(2):181‐4. CENTRAL

Csatary 1998 {published data only}

Csatary LK, Telegdy L, Gergely P, Bodey B, Bakacs T. Preliminary report of a controlled trial of MTH‐68/B virus vaccine treatment in acute B and C hepatitis: a phase II study. Anticancer Research 1998;18(2B):1279‐82. CENTRAL

Deterding 2013 {published data only}

Deterding K, Gruner N, Buggisch P, Wiegand J, Gale PR, Spengler U, et al. Delayed versus immediate treatment for patients with acute hepatitis C: a randomised controlled non‐inferiority trial. Lancet Infectious Diseases 2013;13(6):497‐506. CENTRAL

Genesca 1993 {published data only}

Genesca J. Interferon alfa in acute posttransfusion hepatitis C: a randomized, controlled trial. Gastroenterology 1992;103(5):1702‐3. CENTRAL
Genesca J, Esteban JI, Quer J, Viladomiu LL, Gonzalez A, Esteban R, et al. Hepatitis C virus markers in patients with acute post‐transfusion hepatitis treated with interferon alfa‐2b. Gut 1993;34(2 Suppl):S62‐3. CENTRAL

Hwang 1994 {published data only}

Hwang SJ, Lee SD, Chan CY, Lu RH, Lo KJ. A randomized controlled trial of recombinant interferon alpha‐2b in the treatment of Chinese patients with acute post‐transfusion hepatitis C. Journal of Hepatology 1994;21(5):831‐6. CENTRAL
Hwang SJ, Lee SD, Lee YH, Wu JC, Chan CY, Huang YS, et al. A randomized controlled clinical trial of recombinant interferon alpha‐2b in the treatment of acute post‐transfusion hepatitis C: a preliminary report. Journal of Gastroenterology and Hepatology 1993;8:S92‐8. CENTRAL

Lampertico 1994 {published data only}

Lampertico P, Rumi M, Romeo R, Craxi A, Soffredini R, Biassoni D, et al. A multicenter randomized controlled trial of recombinant interferon‐alpha 2b in patients with acute transfusion‐associated hepatitis C. Hepatology 1994;19(1):19‐22. CENTRAL
Lampertico P, Rumi MG, De FF, Romeo R, Soffredini R, Biassoni D, et al. Treatment of acute post‐transfusional non‐a, non‐b hepatitis with recombinant interferon alpha‐2b. A multicenter randomized controlled trial. Argomenti Gastroenterologia Clinica 1994;7(3):111‐5. CENTRAL

Omata 1991 {published data only}

Omata M, Yokosuka O, Takano S, Kato N, Hosoda K, Imazeki F, et al. Resolution of acute hepatitis C after therapy with natural beta interferon. Lancet 1991;338(8772):914‐5. CENTRAL

Omata 1994 {published data only}

Omata M, Takano S. A randomized, controlled trial of interferon‐beta treatment for acute hepatitis C. Proceedings of the international symposium on viral hepatitis and liver Disease. 1st Edition. Tokyo: Springer‐Verlag, 1994:601‐3. CENTRAL

Santantonio 2014 {published data only}

Santantonio T, Fasano M, Sagnelli E, Tundo P, Babudieri S, Fabris P, et al. Acute hepatitis C: a 24‐week course of pegylated interferon alpha‐2b versus a 12‐week course of pegylated interferon alpha‐2b alone or with ribavirin. Hepatology 2014;59(6):2101‐9. CENTRAL

Wang 2005 {published data only}

Wang C, Cook L, Krows M, Ng K, Hough E, Thiede H. Randomized trial of pegylated interferon for the treatment of acute HCV in Seattle injection drug users. Hepatology 2005;42(4 Suppl 1):673a. CENTRAL

Additional references

AASLD 2014

American Association for the Study of Liver Diseases, Infectious Diseases Society of America. Recommendations for testing, managing, and treating hepatitis C, 2014. www.hcvguidelines.org/fullreport (accessed 12 October 2014).

Anonymous 1981

Anonymous. Interferon production by genetic engineering. British Medical Journal (Clinical Research Ed.) 1981;282(6265):674‐5.

Bailon 2001

Bailon P, Palleroni A, Schaffer CA, Spence CL, Fung WJ, Porter JE, et al. Rational design of a potent, long‐lasting form of interferon: a 40 kDa branched polyethylene glycol‐conjugated interferon alpha‐2a for the treatment of hepatitis C. Bioconjugate Chemistry 2001;12(2):195‐202.

Beinhardt 2012

Beinhardt S, Aberle JH, Strasser M, Dulic‐Lakovic E, Maieron A, Kreil A, et al. Serum level of IP‐10 increases predictive value of IL28B polymorphisms for spontaneous clearance of acute HCV infection. Gastroenterology 2012;142(1):78‐85.

Beltrami 2000

Beltrami EM, Williams IT, Shapiro CN, Chamberland ME. Risk and management of blood‐borne infections in health care workers. Clinical Microbiology Reviews 2000;13(3):385‐407.

Bunchorntavakul 2015

Bunchorntavakul C, Jones LM, Kikuchi M, Valiga ME, Kaplan DE, Nunes FA, et al. Distinct features in natural history and outcomes of acute hepatitis C. Journal of Clinical Gastroenterology 2015;40(4):e31‐40.

Chaimani 2012

Chaimani A, Salanti G. Using network meta‐analysis to evaluate the existence of small‐study effects in a network of interventions. Research Synthesis Methods 2012;3(2):161‐76.

Chaimani 2013

Chaimani A, Higgins JP, Mavridis D, Spyridonos P, Salanti G. Graphical tools for network meta‐analysis in STATA. PLoS One 2013;8(10):e76654.

Chan 2013

Chan AW, Tetzlaff JM, Altman DG, Laupacis A, Gotzsche PC, Krleza‐Jeric K, et al. SPIRIT 2013 statement: defining standard protocol items for clinical trials. Annals of Internal Medicine 2013;158(3):200‐7.

Coilly 2014

Coilly A, Roche B, Dumortier J, Leroy V, Botta‐Fridlund D, Radenne S, et al. Safety and efficacy of protease inhibitors to treat hepatitis C after liver transplantation, a multicenter experience. Journal of Hepatology 2014;60(1):78‐86.

D'Amico 2006

D'Amico G, Garcia‐Tsao G, Pagliaro L. Natural history and prognostic indicators of survival in cirrhosis: a systematic review of 118 studies. Journal of Hepatology 2006;44(1):217‐31.

Del Re 2013

Del Re AC, Spielmans GI, Flückiger C, Wampold BE. Efficacy of new generation antidepressants: differences seem illusory. PLoS One 2013;8(6):e63509.

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.

Dias 2010

Dias S, Welton NJ, Caldwell DM, Ades AE. Checking consistency in mixed treatment comparison meta‐analysis. Statistics in Medicine 2010;29(7‐8):932‐44.

Dias 2012a

Dias S, Sutton AJ, Welton NJ, Ades AE. NICE DSU Technical Support Document 3: heterogeneity: subgroups, meta‐regression, bias and bias‐adjustment, September 2011 (last updated April 2012). www.nicedsu.org.uk/TSD3%20Heterogeneity.final%20report.08.05.12.pdf (accessed 27 March 2014).

Dias 2012b

Dias S, Welton NJ, Sutton AJ, Ades AE. NICE DSU Technical Support Document 1: introduction to evidence synthesis for decision making, April 2011 (last updated April 2012). www.nicedsu.org.uk/TSD1%20Introduction.final.08.05.12.pdf (accessed 27 March 2014).

Dias 2014a

Dias S, Welton NJ, Sutton AJ, Ades AE. NICE DSU Technical Support Document 2: a generalised linear modelling framework for pairwise and network meta‐analysis of randomised controlled trials, August 2011 (last updated April 2014). www.nicedsu.org.uk/TSD2%20General%20meta%20analysis%20corrected%2015April2014.pdf (accessed 8 October 2014).

Dias 2014b

Dias S, Welton NJ, Sutton AJ, Caldwell DM, Lu G, Ades AE. NICE DSU Technical Support Document 4: inconsistency in networks of evidence based on randomised controlled trials, May 2011 (last updated April 2014). www.nicedsu.org.uk/TSD4%20Inconsistency.final.15April2014.pdf (accessed 8 October 2014).

EASL 2014

European Association for Study of Liver. EASL clinical practice guidelines: management of hepatitis C virus infection. Journal of Hepatology 2014;60(2):392‐420.

Egger 1997

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

El Khoury 2012

El Khoury AC, Wallace C, Klimack WK, Razavi H. Economic burden of hepatitis C‐associated diseases: Europe, Asia Pacific, and the Americas. Journal of Medical Economics 2012;15(5):887‐96.

EuroQol 2014

EuroQol. About EQ‐5D, 2014. www.euroqol.org/about‐eq‐5d.html (accessed 8 October 2014).

Feld 2005

Feld JJ, Hoofnagle JH. Mechanism of action of interferon and ribavirin in treatment of hepatitis C. Nature 2005;436(7053):967‐72.

Fierer 2014

Fierer DS, Dieterich DT, Mullen MP, Branch AD, Uriel AJ, Carriero DC, et al. Telaprevir in the treatment of acute hepatitis C virus infection in HIV‐infected men. Clinical Infectious Diseases 2014;58(6):873‐9.

Furusyo 2013

Furusyo N, Ogawa E, Nakamuta M, Kajiwara E, Nomura H, Dohmen K, et al. Telaprevir can be successfully and safely used to treat older patients with genotype 1b chronic hepatitis C. Journal of Hepatology 2013;59(2):205‐12.

Global Burden of Hepatitis C Working Group 2004

Global Burden of Hepatitis C Working Group. Global burden of disease (GBD) for hepatitis C. Journal of Clinical Pharmacology 2004;44(1):20‐9.

Gluud 2014

Gluud C, Koretz R, Gurusamy K. Hepatitis C: a new direction, but an old story?. Lancet (London, England)2014; Vol. 383, issue 9935:2122‐3. [PUBMED: 24953469]

Gluud 2015

Gluud C, Nikolova D, Klingenberg SL. Cochrane Hepato‐Biliary. About The Cochrane Collaboration (Cochrane Review Groups (CRGs)) 2015, Issue 1. Art. No.: LIVER.

Grebely 2011

Grebely J, Matthews GV, Dore GJ. Treatment of acute HCV infection. Nature Reviews. Gastroenterology & Hepatology 2011;8(5):265‐74.

Grebely 2014

Grebely J, Page K, Sacks‐Davis R, van der Loeff MS, Rice TM, Bruneau J, et al. The effects of female sex, viral genotype, and IL28B genotype on spontaneous clearance of acute hepatitis C virus infection. Hepatology 2014;59(1):109‐20.

Guyatt 2011

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

Hajarizadeh 2012

Hajarizadeh B, Grebely J, Dore GJ. Case definitions for acute hepatitis C virus infection: a systematic review. Journal of Hepatology 2012;57(6):1349‐60.

Hezode 2013

Hezode C, Fontaine H, Dorival C, Larrey D, Zoulim F, Canva V, et al. Triple therapy in treatment‐experienced patients with HCV‐cirrhosis in a multicentre cohort of the French Early Access Programme (ANRS CO20‐CUPIC) ‐ NCT01514890. Journal of Hepatology 2013;59(3):434‐41.

Higgins 2011

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

Higgins 2012

Higgins JPT, Jackson D, Barrett JK, Lu G, Ades AE, White IR. Consistency and inconsistency in network meta‐analysis: concepts and models for multi‐arm studies. Research Synthesis Methods 2012;3(2):98‐110.

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, Pennsylvania: Barnett International/PAREXEL, 1997.

Jafari 2010

Jafari S, Copes R, Baharlou S, Etminan M, Buxton J. Tattooing and the risk of transmission of hepatitis C: a systematic review and meta‐analysis. International Journal of Infectious Diseases 2010;14(11):e928‐40.

Jakobsen 2014

Jakobsen JC, Wetterslev J, Winkel P, Lange T, Gluud C. Thresholds for statistical and clinical significance in systematic reviews with meta‐analytic methods. BMC Medical Research Methodology 2014;14(1):120.

Kamal 2008

Kamal SM. Acute hepatitis C: a systematic review. American Journal of Gastroenterology 2008;103(5):1283‐97.

Kenny‐Walsh 1999

Kenny‐Walsh E. Clinical outcomes after hepatitis C infection from contaminated anti‐D immune globulin. Irish Hepatology Research Group. New England Journal of Medicine 1999;340(16):1228‐33.

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.

Kleinman 2009

Kleinman SH, Lelie N, Busch MP. Infectivity of human immunodeficiency virus‐1, hepatitis C virus, and hepatitis B virus and risk of transmission by transfusion. Transfusion 2009;49(11):2454‐89.

Koretz 2015

Koretz RL, Lin KW, Ioannidis JP, Lenzer J. Is widespread screening for hepatitis C justified?. BMJ (Clinical Research Ed.) 2015;350:g7809.

Lavanchy 2011

Lavanchy D. Evolving epidemiology of hepatitis C virus. Clinical Microbiology and Infection 2011;17(2):107‐15.

Lehmann 2004

Lehmann M, Meyer MF, Monazahian M, Tillmann HL, Manns MP, Wedemeyer H. High rate of spontaneous clearance of acute hepatitis C virus genotype 3 infection. Journal of Medical Virology 2004;73(3):387‐91.

Lewis 2012

Lewis H, Cunningham M, Foster G. Second generation direct antivirals and the way to interferon‐free regimens in chronic HCV. Best Practice & Research Clinical Gastroenterology 2012;26(4):471‐85.

Loomba 2011

Loomba R, Rivera MM, McBurney R, Park Y, Haynes‐Williams V, Rehermann B, et al. The natural history of acute hepatitis C: clinical presentation, laboratory findings and treatment outcomes. Alimentary Pharmacology & Therapeutics 2011;33(5):559‐65.

Lu 2006

Lu G, Ades AE. Assessing evidence inconsistency in mixed treatment comparisons. Journal of the American Statistical Association 2006;101(474):447‐59.

Lundh 2017

Lundh A, Sismondo S, Lexchin J, Busuioc OA, Bero L. Industry sponsorship and research outcome. Cochrane Database of Systematic Reviews 2017, Issue 2. [DOI: 10.1002/14651858.MR000033.pub3]

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.

Maheshwari 2008

Maheshwari A, Ray S, Thuluvath PJ. Acute hepatitis C. Lancet 2008;372(9635):321‐32.

Martindale 2011

Sweetman S, editor. Martindale: the complete drug reference (online version), 37th edition, 2011. www.pharmpress.com/product/MC_MART/martindale‐the‐complete‐drug‐reference (accessed 23 September 2014).

Mills 2012

Mills EJ, Ioannidis JP, Thorlund K, Schünemann HJ, Puhan MA, Guyatt GH. How to use an article reporting a multiple treatment comparison meta‐analysis. JAMA 2012;308(12):1246‐53.

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.

Myers 2001

Myers RP, Regimbeau C, Thevenot T, Leroy V, Mathurin P, Opolon P, et al. Interferon for acute hepatitis C. Cochrane Database of Systematic Reviews 2001, Issue 4. [DOI: 10.1002/14651858.CD000369]

NCBI 2014a

NCBI. Hepatitis C, 2014. www.ncbi.nlm.nih.gov/mesh/68006526 (accessed 11 October 2014).

NCBI 2014b

NCBI. Interferons, 2014. www.ncbi.nlm.nih.gov/mesh/68007372 (accessed 11 October 2014).

Newell 1992

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

OpenBUGS 3.2.3 [Computer program]

Members of OpenBUGS Project Management Group. OpenBUGS. Version 3.2.3. Members of OpenBUGS Project Management Group, 2014.

Perry 2012

Perry CM. Telaprevir: a review of its use in the management of genotype 1 chronic hepatitis C. Drugs 2012;72(5):619‐41.

Poordad 2014

Poordad F, Hezode C, Trinh R, Kowdley KV, Zeuzem S, Agarwal K, et al. ABT‐450/r‐ombitasvir and dasabuvir with ribavirin for hepatitis C with cirrhosis. New England Journal of Medicine 2014;370(21):1973‐82.

Poynard 1997

Poynard T, Bedossa P, Opolon P. Natural history of liver fibrosis progression in patients with chronic hepatitis C. The OBSVIRC, METAVIR, CLINIVIR, and DOSVIRC groups. Lancet 1997;349(9055):825‐32.

Puhan 2014

Puhan MA, Schünemann HJ, Murad MH, Li T, Brignardello‐Petersen R, Singh JA, et al. A GRADE Working Group approach for rating the quality of treatment effect estimates from network meta‐analysis. BMJ (Clinical Research Ed.) 2014;349:g5630.

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.

Rodger 2000

Rodger AJ, Roberts S, Lanigan A, Bowden S, Brown T, Crofts N. Assessment of long‐term outcomes of community‐acquired hepatitis C infection in a cohort with sera stored from 1971 to 1975. Hepatology 2000;32(3):582‐7.

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.

Salanti 2011

Salanti G, Ades AE, Ioannidis JP. Graphical methods and numerical summaries for presenting results from multiple‐treatment meta‐analysis: an overview and tutorial. Journal of Clinical Epidemiology 2011;64(2):163‐71.

Salanti 2012

Salanti G. Indirect and mixed‐treatment comparison, network, or multiple‐treatments meta‐analysis: many names, many benefits, many concerns for the next generation evidence synthesis tool. Research Synthesis Methods 2012;3(2):80‐97.

Sangiovanni 2006

Sangiovanni A, Prati GM, Fasani P, Ronchi G, Romeo R, Manini M, et al. The natural history of compensated cirrhosis due to hepatitis C virus: a 17‐year cohort study of 214 patients. Hepatology 2006;43(6):1303‐10.

Savović 2012a

Savović 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: combined analysis of meta‐epidemiological studies. Health Technology Assessment 2012;16(35):1‐82.

Savović 2012b

Savović 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.

Schulz 2010

Schulz KF, Altman DG, Moher D. CONSORT 2010 statement: updated guidelines for reporting parallel group randomised trials. BMJ 2010;340:c332.

Seeff 2001

Seeff LB, Hollinger FB, Alter HJ, Wright EC, Cain CM, Buskell ZJ, et al. Long‐term mortality and morbidity of transfusion‐associated non‐A, non‐B, and type C hepatitis: a National Heart, Lung, and Blood Institute collaborative study. Hepatology 2001;33(2):455‐63.

Seeff 2009

Seeff LB. The history of the "natural history" of hepatitis C (1968‐2009). Liver International 2009;29(Suppl 1):89‐99.

Severini 1993

Severini TA. Bayesian interval estimates which are also confidence intervals. Journal of the Royal Statistical Society. Series B (Methodological) 1993;55(2):533‐40.

Singal 2013

Singal AK, Hmoud BS, Guturu P, Kuo YF. Outcome after liver transplantation for cirrhosis due to alcohol and hepatitis C: comparison to alcoholic cirrhosis and hepatitis C cirrhosis. Journal of Clinical Gastroenterology 2013;47(8):727‐33.

Smith 2014

Smith DB, Bukh J, Kuiken C, Muerhoff AS, Rice CM, Stapleton JT, et al. Expanded classification of hepatitis C virus into 7 genotypes and 67 subtypes: updated criteria and genotype assignment web resource. Hepatology 2014;59(1):318‐27.

Stata/SE 14.2 [Computer program]

StataCorp LP. Stata/SE 14.2 for Windows[64‐bit x86‐64]. Version 14. College Station: StataCorp LP, 2017.

Syriopoulou 2005

Syriopoulou V, Nikolopoulou G, Daikos GL, Theodoridou M, Pavlopoulou I, Nicolaidou P, et al. Mother to child transmission of hepatitis C virus: rate of infection and risk factors. Scandinavian Journal of Infectious Diseases 2005;37(5):350‐3.

Thorlund 2011

Thorlund K, Engstrøm J, Wetterslev J, Brok J, Imberger G, Gluud C. User manual for Trial Sequential Analysis (TSA). ctu.dk/tsa/files/tsa_manual.pdf 2011 (accessed 30 November 2016).

Thorlund 2012

Thorlund K, Mills EJ. Sample size and power considerations in network meta‐analysis. Systematic Reviews 2012;1:41.

Tohme 2010

Tohme RA, Holmberg SD. Is sexual contact a major mode of hepatitis C virus transmission?. Hepatology 2010;52(4):1497‐505.

TSA 2011 [Computer program]

Copenhagen Trial Unit. TSA ‐ Trial Sequential Analysis. Version 0.9 Beta. Copenhagen: Copenhagen Trial Unit, 2011.

Turner 2012

Turner RM, Davey J, Clarke MJ, Thompson SG, Higgins JP. Predicting the extent of heterogeneity in meta‐analysis, using empirical data from the Cochrane Database of Systematic Reviews. International Journal of Epidemiology 2012;41(3):818‐27.

Uemura 2012

Uemura T, Ramprasad V, Hollenbeak CS, Bezinover D, Kadry Z. Liver transplantation for hepatitis C from donation after cardiac death donors: an analysis of OPTN/UNOS data. American Journal of Transplantation 2012;12(4):984‐91.

van Valkenhoef 2012

van Valkenhoef G, Lu G, de Brock B, Hillege H, Ades AE, Welton NJ. Automating network meta‐analysis. Research Synthesis Methods 2012;3(4):285‐99.

Ware 2014

Ware JE. SF‐36® health survey update, 2014. www.sf‐36.org/tools/sf36.shtml (accessed on 8 October 2014).

Wawrzynowicz‐Syczewska 2004

Wawrzynowicz‐Syczewska M, Kubicka J, Lewandowski Z, Boron‐Kaczmarska A, Radkowski M. Natural history of acute symptomatic hepatitis type C. Infection 2004;32(3):138‐43.

Welsch 2012

Welsch C, Jesudian A, Zeuzem S, Jacobson I. New direct‐acting antiviral agents for the treatment of hepatitis C virus infection and perspectives. Gut 2012;61(Suppl 1):i36‐46.

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.

Wiese 2005

Wiese M, Grungreiff K, Guthoff W, Lafrenz M, Oesen U, Porst H. Outcome in a hepatitis C (genotype 1b) single source outbreak in Germany ‐ a 25‐year multicenter study. Journal of Hepatology 2005;43(4):590‐8.

Wood 2008

Wood L, Egger M, Gluud LL, Schulz KF, Jüni P, Altman DG, 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(7644):601‐5.

Xia 2008

Xia X, Luo J, Bai J, Yu R. Epidemiology of hepatitis C virus infection among injection drug users in China: systematic review and meta‐analysis. Public Health 2008;122(10):990‐1003.

Yang 2014

Yang LS, Shan LL, Saxena A, Morris DL. Liver transplantation: a systematic review of long‐term quality of life. Liver International 2014;34(9):1298‐313.

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Calleri 1998

Methods

Randomised clinical trial.

Participants

Country: Italy.

Number randomised: 40.

Post‐randomisation dropouts: 0 (0%).

Revised sample size: 40.

Mean age: 29 years.

Females: 6 (15%).

Genotype 1: not stated.

Genotype 3: not stated.

Other genotypes: not stated.

Mean follow‐up period in months (for all groups): 22.5.

Inclusion criteria

  • Elevated serum alanine aminotransferase levels (above 300 IU/L).

  • Seroconversion from negative to positive anti‐HCV.

Exclusion criteria

  • Other causes of acute liver damage.

  • Heavy alcohol intake (> 40 g/day).

  • Fulminant hepatitis.

  • History of chronic liver disease or severe non‐liver diseases (cancer, chronic renal failure, chronic heart failure).

  • HBV and HIV carriers.

  • Pregnant women.

Interventions

Participants were randomly assigned to 2 groups.

Group 1: interferon‐beta (n = 20).

Further details: interferon‐beta 3 MU IM once daily for 5 days then 3 times per day for 3 more weeks.

Group 2: no intervention (n = 20).

Duration of treatment: 1 month.

Outcomes

Sustained virological response.

Biochemical response.

Severity and frequency of adverse events.

Notes

6 participants were not randomised for logistical issues and they were allocated to the immediate treatment.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Randomized, computer generated according to the author's reply."

Allocation concealment (selection bias)

Unclear risk

Comment: information not available.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Comment: "different treatments" (author replies).

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Comment: "different treatments" (author replies).

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: no post‐randomisation dropouts.

Selective reporting (reporting bias)

Low risk

Comment: all important outcomes were reported.

For‐profit bias

Low risk

Quote: "Italian NHS".

Comment: according to the author's reply.

Other bias

Low risk

Comment: no other risk of bias.

Csatary 1998

Methods

Randomised clinical trial.

Participants

Country: Hungary, USA.

Number randomised: 41.

Post‐randomisation dropouts: 0 (0%).

Revised sample size: 41.

Mean age: not stated.

Females: not stated.

Genotype 1: not stated.

Genotype 3: not stated.

Other genotypes: not stated.

Mean follow‐up period in months (for all groups): 12.

Inclusion criteria

  • Anti‐HCV positive.

  • Hospitalisation because of jaundice, other clinical signs of acute hepatitis (fever, severe malaise, loss of appetite) and a 10‐ to 100‐fold elevation of alanine aminotransferase level.

Exclusion criteria

  • Positivity for HAV, HBV, Epstein‐Barr virus, cytomegalovirus.

  • HIV positivity.

  • Suspicion or evidence of alcohol‐induced or drug‐induced hepatitis.

  • Clinical or histological signs of chronic hepatitis as well as other chronic liver diseases.

  • Fulminant hepatitis.

  • Malignancies.

  • Underlying systemic disease.

  • Immunosuppressive treatment within 6 months.

  • Pregnancy.

  • Lack of compliance.

Interventions

Participants were randomly assigned to 2 groups.

Group 1: MTH‐68/B (n = 22).

Further details: MTH‐68/B (live attenuated infectious bursal disease virus (IBDV)) 4000 U/day for 1 week, then 3 times per week for 2 weeks, then once monthly for 6 months.

Group 2: no intervention (n = 19).

Duration of treatment: 6 months

Outcomes

Sustained virological response.

Frequency of adverse events.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Patients were randomly allocated to two groups."

Comment: further details not available.

Allocation concealment (selection bias)

Unclear risk

Comment: information not available.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Comment: no use of placebo.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: information not available.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: no post‐randomisation dropouts.

Selective reporting (reporting bias)

Low risk

Comment: all important outcomes were reported.

For‐profit bias

Unclear risk

Comment: information not available.

Other bias

Low risk

Comment: no other bias.

Deterding 2013

Methods

Randomised clinical trial.

Participants

Country: Germany.

Number randomised: 107.

Post‐randomisation dropouts: 0 (0%).

Revised sample size: 107.

Mean age: 39 years.

Females: 46 (43%).

Genotype 1: 73 (68.2%).

Genotype 3: 18 (16.8%).

Other genotypes: 10 (9.3%).

Mean follow‐up period in months (for all groups): 6.

Inclusion criteria

  • Aged > 18 years.

  • Acute HCV infection: seroconversion for antibodies against HCV documented OR proven/very likely exposure to HCV within the preceding 4 months combined with a serum ALT level of at least 10‐times the upper limit of normal range with no evidence of any pre‐existing disorder.

  • ≥ 1 of right upper abdominal pain, jaundice, influenza‐like symptoms, fatigue.

Exclusion criteria

  • People who had other causes of liver disease as assessed by standard clinical and laboratory criteria.

  • HBV infection.

  • HIV coinfection.

  • People with ongoing uncontrolled misuse of alcohol or IV drugs.

  • Autoimmune diseases.

  • Absolute neutrophil count < 1500 cells/mm3.

  • Thrombocytopenia (< 70000 cells/mm3).

  • Anaemia (< 11 g/dL in women and < 12 g/dL in men).

  • Decompensated liver disease.

  • Decompensated renal disease.

  • Decompensated thyroid disease.

  • Psychiatric conditions.

  • History of seizures.

  • Poorly controlled diabetes mellitus.

  • Ophthalmological disease.

  • Immunologically mediated disease.

  • History of chronic pulmonary disease or cardiac disease.

  • Pregnancy.

  • History of transplantation or malignancy.

Interventions

Participants were randomly assigned to 2 groups.

Group 1: immediate pegylated interferon‐alpha‐2b (n = 55).

Further details: immediate pegylated interferon‐alfa‐2b 1.5 μg/kg.

Group 2: delayed pegylated interferon‐alfa‐2b (n = 52).

Further details: delayed pegylated interferon‐alfa‐2b 1.5 μg/kg + ribavirin > 10.6 mg/kg.

Duration of treatment: 6 months.

Outcomes

Sustained virological response.

Biochemical response.

Severity and frequency of adverse events.

Analysis of responses to the respective treatment approaches according to severity of symptoms.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Patients were randomised via a web‐based randomisation service provided by the Hep‐Net Study House."

Allocation concealment (selection bias)

Unclear risk

Quote: "We used stratified block randomisation with block sizes of eight, independent across strata."

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Quote: "This investigator‐initiated study was designed as an open‐label, phase 3, multicentre study."

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Comment: outcome assessors blind according to the author reply.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: no post‐randomisation dropouts.

Selective reporting (reporting bias)

Low risk

Comment: all important outcomes were reported.

For‐profit bias

High risk

Quote: "The study was supported by the German Network of Competence on Viral Hepatitis (Hep‐Net, funded by the Federal Ministry of Education and Research). The study was also supported by a research grant from Essex Pharma, Schering‐Plough, and MSD. MSD provided study drugs and financial support."

Other bias

Low risk

Comment: no other bias.

Genesca 1993

Methods

Randomised clinical trial.

Participants

Country: Spain.

Number randomised: 28.

Post‐randomisation dropouts: 0 (0%).

Revised sample size: 28.

Mean age: not stated.

Females: not stated.

Genotype 1: not stated.

Genotype 3: not stated.

Other genotypes: not stated.

Mean follow‐up period in months (for all groups): 12 months.

Inclusion criteria

  • Acute HCV.

Interventions

Participants were randomly assigned to 2 groups.

Group 1: interferon‐alpha‐2b (n = 15).

Further details: interferon‐alpha‐2b 3 MU 3 times per week.

Group 2: no treatment (n = 13).

Duration of treatment: 3 months.

Outcomes

Sustained virological response.

Biochemical response.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "A randomised, controlled trial was undertaken."

Comment: further details not available.

Allocation concealment (selection bias)

Unclear risk

Comment: information not available.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Quote: "Twenty eight patients with acute HCV (15 treated and 13 controls) were included in the trial."

Comment: placebo not used.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: information not available.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: no post‐randomisation dropouts.

Selective reporting (reporting bias)

High risk

Comment: some important outcomes which would generally be assessed were not reported.

For‐profit bias

Unclear risk

Comment: information not available.

Other bias

Low risk

Comment: no other bias.

Hwang 1994

Methods

Randomised clinical trial.

Participants

Country: China.

Number randomised: 33.

Post‐randomisation dropouts: 0 (0%).

Revised sample size: 33.

Mean age: 54 years.

Females: 9 (27.3%).

Genotype 1: not stated.

Genotype 3: not stated.

Other genotypes: not stated.

Mean follow‐up period in months (for all groups): 12.

Inclusion criteria

  • People who received blood transfusion due to cardiovascular surgery, other operations, or upper gastrointestinal bleeding in Veterans General Hospital, Taipei.

  • Diagnosis of acute HCV infection: serum ALT elevated above 90 IU/L (twice the upper normal value) and the seroconversion of serum antibody to HCV (anti‐HCV) or serum HCV‐RNA after blood transfusion.

Exclusion criteria

  • Estimated survival < 6 months.

  • History of interferon treatment within 12 months prior to entering trial.

  • Presence of severe systemic diseases or malignancies.

  • Women of childbearing age not using contraception and breastfeeding mothers.

  • Age < 18 years.

  • Evidence of haematopoietic dysfunction.

  • Presence of decompensated liver conditions such as hepatic encephalopathy, ascites, or a serum bilirubin level > 4 mg/dL.

  • Presence of any concurrent illness that could interfere with the investigator's assessment in the treatment of hepatitis.

  • Psychiatric disorders.

Interventions

Participants were randomly assigned to 2 groups.

Group 1: interferon‐alpha‐2b (n = 16).

Further details: interferon‐alpha‐2b 3 MU 3 times per week.

Group 2: no treatment (n = 17).

Duration of treatment: 3 months.

Outcomes

Sustained virological response.

Frequency of adverse events.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "These patients were randomly allocated to either the IFN‐treated group or the control group by a random number table."

Allocation concealment (selection bias)

Unclear risk

Comment: information not available.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Quote: "Seventeen patients in the control group received no specific treatment."

Comment: placebo not used.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: information not available.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Quote: "Only one patient in the control group was unwilling to continue and withdrew from the study after 6 months of follow up."

Comment: there were post‐randomisation dropouts.

Selective reporting (reporting bias)

Low risk

Comment: all important outcomes were reported.

For‐profit bias

High risk

Quote: "This study was supported by grants from the National Science Council (NSC82‐0419‐B075‐092) and National Health Research Institutes (DOH‐83‐HR‐208), Republic of China. Drug supplied by pharm. company."

Other bias

Low risk

Comment: no other bias.

Lampertico 1994

Methods

Randomised clinical trial.

Participants

Country: Italy.

Number randomised: 41.

Post‐randomisation dropouts: 3 (7.3%).

Revised sample size: 38.

Mean age: 47 years.

Females: 19 (50%).

Genotype 1: not stated.

Genotype 3: not stated.

Other genotypes: not stated.

Mean follow‐up period in months (for all groups): 18.

Inclusion criteria

  • Post‐transfusion acute non‐A, non‐B/type C hepatitis: increase of serum ALT level to > 2.5 times the upper normal limit, on 2 separate occasions at least 2 weeks apart between 2 weeks and 6 months after transfusion.

Exclusion criteria

  • Aged > 60 years.

  • Pregnancy.

  • Previous transfusion with blood, fresh frozen plasma, or clotting factor concentrates.

  • Treatment with immunosuppressive drugs.

  • Malignant tumours.

  • Antibody to HIV.

Interventions

Participants were randomly assigned to 2 groups.

Group 1: interferon‐alpha‐2b (n = 22).

Further details: interferon‐alpha‐2b 3 MU IM 3 times per week.

Group 2: no treatment (n = 16).

Duration of treatment: 3 months.

Outcomes

Sustained virological response.

Biochemical response.

Mortality at the end of follow‐up (18 months).

Notes

Reasons for post‐randomisation dropouts: of the 48 participants enrolled in the study, 1 refused therapy and 2 untreated people were lost to follow‐up during month 1. 7 participants (16% of total) were thought to have been infected with a non‐A, non‐B, non‐C agent.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Comment: randomisation was computerised according to the author's reply.

Allocation concealment (selection bias)

Unclear risk

Comment: information not available.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Comment: multicentre, prospective, open, randomised study comparing interferon treatment and no treatment.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: information not available.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Comment: there were post‐randomisation dropouts.

Selective reporting (reporting bias)

High risk

Comment: some important outcomes which would generally be assessed were not reported.

For‐profit bias

High risk

Quote: "We thank Dr. Paola Mazzanti and Dr. Cristina Pintus (Schering‐Plough) for their assistance."

Other bias

Low risk

Comment: there was no other bias.

Omata 1991

Methods

Randomised clinical trial.

Participants

Country: Japan.

Number randomised: 27.

Post‐randomisation dropouts: 2 (7.4%).

Revised sample size: 25.

Mean age: 40 years.

Females: 14 (56%).

Genotype 1: not stated.

Genotype 3: not stated.

Other genotypes: not stated.

Mean follow‐up period in months (for all groups): 36.

Inclusion criteria

  • Serum ALT > 200 IU after transfusion or raised serum ALT without a history of taking hepatotoxic drug or of heavy alcohol intake.

  • Seronegativity for hepatitis B surface antigen, IgM antibody to HBV core protein, IgM‐HA antibody, HBV‐DNA, and autoimmune markers.

  • Liver histology compatible with the diagnosis of acute hepatitis.

Interventions

Participants were randomly assigned to 2 groups.

Group 1: interferon‐beta (n = 11).

Further details: interferon‐beta, 3 MU IV for 5 consecutive days in the first week, and then 3 times per week for the next 3 weeks.

Group 2: no treatment (n = 14).

Duration of treatment: 1 month.

Outcomes

Sustained virological response.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "25 patients with acute non‐A, non‐B hepatitis drew lots for allocation to treatment with interferon."

Allocation concealment (selection bias)

Low risk

Quote: "25 patients with acute non‐A, non‐B hepatitis drew lots for allocation to treatment with interferon."

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Comment: placebo not used.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: information not available.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Quote: "2 patients in the untreated group were lost to follow‐up."

Comment: there were post‐randomisation dropouts.

Selective reporting (reporting bias)

High risk

Comment: some important outcomes which would generally be assessed were not reported.

For‐profit bias

Unclear risk

Comment: this information was not available.

Other bias

Low risk

Comment: there was no other bias.

Omata 1994

Methods

Randomised clinical trial.

Participants

Country: Japan.

Number randomised: 25.

Post‐randomisation dropouts: 0 (0%).

Revised sample size: 25.

Mean age: 39 years.

Females: 14 (56%).

Genotype 1: not stated.

Genotype 3: not stated.

Other genotypes: not stated.

Mean follow‐up period in months (for all groups): 36.

Inclusion criteria

  • Serum ALT > 200 IU after transfusion, or elevated serum ALT without a history of transfusion, hepatotoxic drugs, or heavy alcohol intake.

  • Liver histology compatible with the diagnosis of acute hepatitis.

Exclusion criteria

  • Seronegativity for hepatitis B surface antigen, IgM antibody to HBV core protein, IgM anti‐HA, HBV‐DNA, and autoimmune markers.

Interventions

Participants were randomly assigned to 2 groups.

Group 1: interferon‐beta (n = 11).

Further details: interferon‐beta 3 MU IV for 5 days, then 3 times per week for 3 weeks.

Group 2: no treatment (n = 14).

Duration of treatment: 1 month.

Outcomes

Sustained virological response.

Fluctuation of ALT concentrations.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "25 patients randomly assigned to."

Comment: further details not available.

Allocation concealment (selection bias)

Unclear risk

Comment: information not available.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Comment: no use of placebo.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: information not available.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: there were no post‐randomisation dropouts.

Selective reporting (reporting bias)

High risk

Comment: some important outcomes which would generally be assessed were not reported.

For‐profit bias

Unclear risk

Comment: information not available.

Other bias

Low risk

Comment: there was no other bias.

Santantonio 2014

Methods

Randomised clinical trial.

Participants

Country: Italy.

Number randomised: 130.

Post‐randomisation dropouts: 0 (0%).

Revised sample size: 130.

Mean age: 34 years.

Females: 41 (31.5%).

Genotype 1: 53 (40.8%).

Genotype 3: 32 (24.6%).

Other genotypes: 45 (34.6%).

Mean follow‐up period in months (for all groups): 12.

Inclusion criteria

  • Aged 18 to 65 years.

  • Diagnosis of acute HCV infection: documented anti‐HCV seroconversion or, alternatively, abrupt increase of transaminases > 20 times the upper limit of the normal range.

  • Absence of other hepatitis viruses (HAV, HBV) or toxic hepatitis in previously healthy people.

  • HCV‐RNA positive.

  • Acute hepatitis C still viraemic after 12 weeks of observation from disease onset.

Exclusion criteria

  • Liver disease unrelated to HCV infection.

  • Haemoglobin < 12 g/dL in women and 13 g/dL in men.

  • White blood count < 3000/µL.

  • Platelets < 100,000/µL.

  • Pregnancy.

  • History of severe psychiatric disease.

  • Neurological disease.

  • Severe cardiac, gastrointestinal, and kidney disease.

  • Infection with HBV or HIV.

  • Positive antinuclear antibodies or antismooth muscle antibody (titre > 1/80), or both.

  • History of having received any systemic antineoplastic or immunomodulatory treatment in the previous 6 months.

  • History or other evidence of severe illness or any other conditions that would make people unsuitable for the study (alcohol intake at a daily dose > 40 g for males and > 30 g for females, thalassaemia, and dialysis).

  • People with ongoing drug abuse.

Interventions

Participants were randomly assigned to 3 groups.

Group 1: pegylated interferon‐alpha‐2b (n = 44).

Further details: pegylated interferon‐alpha‐2b 1.5 µg/kg/week (24 weeks).

Group 2: pegylated interferon‐alpha‐2b (n = 43).

Further details: pegylated interferon‐alpha‐2b 1.5 µg/kg/week (12 weeks).

Group 3: pegylated interferon‐alpha‐2b plus ribavirin (n = 43).

Further details: pegylated interferon‐alpha‐2b 1.5 µg/kg/week + ribavirin 10.6 mg/kg/day orally (12 weeks).

Duration of treatment: 12 to 24 weeks (see above).

Outcomes

Sustained virological response.

Virological responses after 2 weeks of treatment (very rapid virological response), after 4 weeks of treatment (rapid virological response), at the end of treatment (end‐of‐treatment virological response), and at 12 months' post‐treatment follow‐up (long‐term virological response).

ALT level normalisation at the end of treatment and at 6 and 12 months' post‐treatment follow‐up.

Safety (adverse events).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "The randomization list was generated centrally by an independent biostatistician using the Proc Plan of the SAS system (version 9.2; SAS Institute Inc., Cary, NC) and consisted of a computer‐generated treatment allocation list in blocks of 9 patients each."

Allocation concealment (selection bias)

Unclear risk

Comment: information not available.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Comment: different treatments, no placebo.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: information not available.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: "ITT analysis, Patients who discontinued the study for any reason before the 6‐month follow‐up visit were considered as nonresponders."

Comment: low for sustained virological response, high for 1‐year mortality because there were post‐randomisation dropouts.

Selective reporting (reporting bias)

High risk

Comment: adverse events not clearly reported.

For‐profit bias

High risk

Quote: "The study sponsor for drug supply and financial support was Schering‐Plough (now Merck) SpA, Milan, Italy."

Other bias

Low risk

Comment: there was no other bias.

Wang 2005

Methods

Randomised clinical trial.

Participants

Country: USA.

Number randomised: 21.

Post‐randomisation dropouts: 0 (0%).

Revised sample size: 21.

Mean age: not stated.

Females: not stated.

Genotype 1: 13 (61.9%).

Genotype 3: not stated.

Other genotypes: not stated.

Mean follow‐up period in months (for all groups): 6.

Inclusion criteria

  • Seroconversion to anti‐HCV positive within 6 months of screening in a previously seronegative IV drug users.

  • Detectable serum HCV.

  • No contraindications to pegylated interferon.

Interventions

Participants were randomly assigned to 2 groups.

Group 1: pegylated interferon‐alpha (n = 9).

Further details: pegylated interferon‐alpha (no further details of treatment regimen).

Group 2: no treatment (n = 12).

Duration of treatment: 6 months.

Outcomes

Sustained virological response was reported but was not reported in sufficient details to include for analysis.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: information not available.

Allocation concealment (selection bias)

Unclear risk

Comment: information not available.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Comment: information not available.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: information not available.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Comment: there were post‐randomisation dropouts.

Selective reporting (reporting bias)

High risk

Comment: some important outcomes which would generally be assessed were not reported.

For‐profit bias

Unclear risk

Comment: information not available.

Other bias

Low risk

Comment: there was no other bias.

ALT: alanine transaminase; DNA: deoxy ribonucleic acid; HAV: hepatitis A virus; HBV: hepatitis B virus; HCV: hepatitis C virus; HCV‐RNA: hepatitis C virus ribonucleic acid; IM: intramuscular; ITT: intention‐to‐treat analysis; IU: international units; IV: intravenous; MU: million units.

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 Serious adverse events (proportion) Show forest plot

4

Odds Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.1

Comparison 1 Intervention versus control, Outcome 1 Serious adverse events (proportion).

Comparison 1 Intervention versus control, Outcome 1 Serious adverse events (proportion).

1.1 Interferon‐beta versus no intervention

1

40

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.2 MTH‐68/B vaccine versus no intervention

1

41

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.3 Pegylated interferon‐alpha plus ribavirin versus pegylated interferon‐alpha

2

237

Odds Ratio (M‐H, Fixed, 95% CI)

1.72 [0.70, 4.21]

2 Serious adverse events (number) Show forest plot

4

Rate Ratio (Fixed, 95% CI)

Subtotals only

Analysis 1.2

Comparison 1 Intervention versus control, Outcome 2 Serious adverse events (number).

Comparison 1 Intervention versus control, Outcome 2 Serious adverse events (number).

2.1 Interferon‐beta versus no intervention

1

40

Rate Ratio (Fixed, 95% CI)

0.0 [0.0, 0.0]

2.2 MTH‐68/B vaccine versus no intervention

1

41

Rate Ratio (Fixed, 95% CI)

0.0 [0.0, 0.0]

2.3 Pegylated interferon‐alpha plus ribavirin versus pegylated interferon‐alpha

2

237

Rate Ratio (Fixed, 95% CI)

2.74 [1.40, 5.33]

3 Adverse events (proportion) Show forest plot

3

Odds Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.3

Comparison 1 Intervention versus control, Outcome 3 Adverse events (proportion).

Comparison 1 Intervention versus control, Outcome 3 Adverse events (proportion).

3.1 Interferon‐alpha versus no intervention

1

33

Odds Ratio (M‐H, Fixed, 95% CI)

203.0 [9.01, 4574.81]

3.2 Interferon‐beta versus no intervention

1

40

Odds Ratio (M‐H, Fixed, 95% CI)

27.88 [1.48, 526.12]

3.3 MTH‐68/B vaccine versus no intervention

1

41

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4 Adverse events (number) Show forest plot

2

Odds Ratio (Fixed, 95% CI)

Subtotals only

Analysis 1.4

Comparison 1 Intervention versus control, Outcome 4 Adverse events (number).

Comparison 1 Intervention versus control, Outcome 4 Adverse events (number).

4.1 Interferon‐beta versus no intervention

1

40

Odds Ratio (Fixed, 95% CI)

17.00 [0.98, 294.53]

4.2 MTH‐68/B vaccine versus no intervention

1

41

Odds Ratio (Fixed, 95% CI)

0.0 [0.0, 0.0]

5 Chronic HCV infection Show forest plot

9

Odds Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.5

Comparison 1 Intervention versus control, Outcome 5 Chronic HCV infection.

Comparison 1 Intervention versus control, Outcome 5 Chronic HCV infection.

5.1 Interferon‐alpha versus no intervention

3

99

Odds Ratio (M‐H, Random, 95% CI)

0.27 [0.09, 0.76]

5.2 Interferon‐beta versus no intervention

3

90

Odds Ratio (M‐H, Random, 95% CI)

0.07 [0.00, 1.24]

5.3 MTH‐68/B vaccine versus no intervention

1

41

Odds Ratio (M‐H, Random, 95% CI)

0.28 [0.05, 1.65]

5.4 Pegylated interferon‐alpha plus ribavirin versus pegylated interferon‐alpha

2

237

Odds Ratio (M‐H, Random, 95% CI)

0.86 [0.41, 1.79]

Study flow diagram.
Figuras y tablas -
Figure 1

Study flow diagram.

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figuras y tablas -
Figure 2

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.
Figuras y tablas -
Figure 3

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Trial Sequential Analysis of serious adverse events (proportion) for pegylated interferon‐alpha plus ribavirin versus pegylated interferon‐alpha performed using an alpha error of 2.5%, power of 90% (beta error of 10%), relative risk reduction (RRR) of 20%, control group proportion observed in trials (Pc = 7%), and observed heterogeneity in the trials (0%) shows that the accrued sample size was only a small fraction of the diversity‐adjusted required information size (DARIS); so the trial sequential monitoring boundaries were not drawn. The Z‐curve (blue line) does not cross the conventional boundaries (dotted green line). There was a high risk of random errors.
Figuras y tablas -
Figure 4

Trial Sequential Analysis of serious adverse events (proportion) for pegylated interferon‐alpha plus ribavirin versus pegylated interferon‐alpha performed using an alpha error of 2.5%, power of 90% (beta error of 10%), relative risk reduction (RRR) of 20%, control group proportion observed in trials (Pc = 7%), and observed heterogeneity in the trials (0%) shows that the accrued sample size was only a small fraction of the diversity‐adjusted required information size (DARIS); so the trial sequential monitoring boundaries were not drawn. The Z‐curve (blue line) does not cross the conventional boundaries (dotted green line). There was a high risk of random errors.

Trial Sequential Analysis of chronic hepatitis C virus for interferon‐alpha versus no intervention was performed using an alpha error of 1.6%, power of 90% (beta error of 10%), relative risk reduction (RRR) of 20%, control group proportion observed in trials (Pc = 84%; upper figure) and Pc = 20% (lower figure), and observed diversity in the trials (0%). The upper figure with Pc = 84% shows that the accrued sample size was only a small fraction of the diversity‐adjusted required information size (DARIS). The Z‐curve (blue line) crosses the conventional boundaries (dotted green line), but it does not cross any of the trial sequential monitoring boundaries (dotted red lines). The lower figure with Pc = 20% shows that the accrued sample size was so small that trial sequential monitoring boundaries were not drawn. There is a high risk of random errors.
Figuras y tablas -
Figure 5

Trial Sequential Analysis of chronic hepatitis C virus for interferon‐alpha versus no intervention was performed using an alpha error of 1.6%, power of 90% (beta error of 10%), relative risk reduction (RRR) of 20%, control group proportion observed in trials (Pc = 84%; upper figure) and Pc = 20% (lower figure), and observed diversity in the trials (0%). The upper figure with Pc = 84% shows that the accrued sample size was only a small fraction of the diversity‐adjusted required information size (DARIS). The Z‐curve (blue line) crosses the conventional boundaries (dotted green line), but it does not cross any of the trial sequential monitoring boundaries (dotted red lines). The lower figure with Pc = 20% shows that the accrued sample size was so small that trial sequential monitoring boundaries were not drawn. There is a high risk of random errors.

Trial Sequential Analysis of chronic hepatitis C virus for interferon‐beta versus no intervention performed using an alpha error of 1.6%, power of 90% (beta error of 10%), relative risk reduction (RRR) of 20%, control group proportion observed in the trials (Pc = 84%; upper figure) and a Pc of 20% (lower figure), and observed heterogeneity in the trials (84%) shows that the accrued sample size was only a small fraction of the diversity‐adjusted required information size (DARIS); so the trial sequential monitoring boundaries were not drawn. The Z‐curve (blue line) crosses the conventional boundaries (dotted green line). There is a high risk of random errors.
Figuras y tablas -
Figure 6

Trial Sequential Analysis of chronic hepatitis C virus for interferon‐beta versus no intervention performed using an alpha error of 1.6%, power of 90% (beta error of 10%), relative risk reduction (RRR) of 20%, control group proportion observed in the trials (Pc = 84%; upper figure) and a Pc of 20% (lower figure), and observed heterogeneity in the trials (84%) shows that the accrued sample size was only a small fraction of the diversity‐adjusted required information size (DARIS); so the trial sequential monitoring boundaries were not drawn. The Z‐curve (blue line) crosses the conventional boundaries (dotted green line). There is a high risk of random errors.

Trial Sequential Analysis of chronic hepatitis C virus infection for pegylated interferon‐alpha plus ribavirin versus pegylated interferon‐alpha performed using an alpha error of 1.6%, power of 90% (beta error of 10%), relative risk reduction (RRR) of 20% (top figure and bottom figure) and 10% (middle figure), control group proportion observed in the trials (Pc = 84%; top figure and middle figure) and Pc = 20% (bottom figure), and observed heterogeneity in the trials (0%) shows that the Z‐curve (blue line) has reached the zone of futility for a RRR of 20% (top figure). However, when a RRR of 10% or when a Pc = 20% was used, the accrued sample size was only a small fraction of the diversity adjusted required information size (DARIS); the Z‐curve (blue line) does not cross the conventional boundaries (dotted green line) or trial sequential monitoring boundaries (dotted red line) (middle figure). For a Pc = 20%, the accrued sample size was so small that the trial sequential monitoring boundaries were not drawn. There is a high risk of random errors.
Figuras y tablas -
Figure 7

Trial Sequential Analysis of chronic hepatitis C virus infection for pegylated interferon‐alpha plus ribavirin versus pegylated interferon‐alpha performed using an alpha error of 1.6%, power of 90% (beta error of 10%), relative risk reduction (RRR) of 20% (top figure and bottom figure) and 10% (middle figure), control group proportion observed in the trials (Pc = 84%; top figure and middle figure) and Pc = 20% (bottom figure), and observed heterogeneity in the trials (0%) shows that the Z‐curve (blue line) has reached the zone of futility for a RRR of 20% (top figure). However, when a RRR of 10% or when a Pc = 20% was used, the accrued sample size was only a small fraction of the diversity adjusted required information size (DARIS); the Z‐curve (blue line) does not cross the conventional boundaries (dotted green line) or trial sequential monitoring boundaries (dotted red line) (middle figure). For a Pc = 20%, the accrued sample size was so small that the trial sequential monitoring boundaries were not drawn. There is a high risk of random errors.

Comparison 1 Intervention versus control, Outcome 1 Serious adverse events (proportion).
Figuras y tablas -
Analysis 1.1

Comparison 1 Intervention versus control, Outcome 1 Serious adverse events (proportion).

Comparison 1 Intervention versus control, Outcome 2 Serious adverse events (number).
Figuras y tablas -
Analysis 1.2

Comparison 1 Intervention versus control, Outcome 2 Serious adverse events (number).

Comparison 1 Intervention versus control, Outcome 3 Adverse events (proportion).
Figuras y tablas -
Analysis 1.3

Comparison 1 Intervention versus control, Outcome 3 Adverse events (proportion).

Comparison 1 Intervention versus control, Outcome 4 Adverse events (number).
Figuras y tablas -
Analysis 1.4

Comparison 1 Intervention versus control, Outcome 4 Adverse events (number).

Comparison 1 Intervention versus control, Outcome 5 Chronic HCV infection.
Figuras y tablas -
Analysis 1.5

Comparison 1 Intervention versus control, Outcome 5 Chronic HCV infection.

Summary of findings for the main comparison. Intervention versus no intervention or control intervention (control) for acute hepatitis C infection: primary outcomes

Intervention versus no intervention or control intervention (control) for acute hepatitis C infection: primary outcomes

Patient or population: people with acute hepatitis C infection

Intervention: multiple

Control: multiple

Settings: secondary or tertiary care

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(trials)

Quality of the evidence
(GRADE)

Assumed risk

Corresponding risk

Control

Intervention

Mortality: there was 1 mortality within 6 months (in the pegylated interferon‐alpha group (1/95 = 1.1%). There was no mortality in the remaining groups. There was no further mortality in the trials which reported mortality until maximal follow‐up.

Serious adverse events: there were no serious adverse events in either group in the comparisons interferon‐beta versus control and MH‐68/B vaccine versus control. Trials in interferon‐alpha versus control did not report serious adverse events.

Serious adverse events (proportion) ‐ pegylated interferon‐alpha plus ribavirin versus pegylated interferon‐alpha

70 per 1000

115 per 1000
(50 to 242)

OR 1.72
(0.7 to 4.21)

237
(2 RCTs)

⊕⊝⊝⊝
Very low1,2,3

Serious adverse events (number) ‐ pegylated interferon‐alpha plus ribavirin versus pegylated interferon‐alpha

92 per 1000

251 per 1000
(128 to 488)

Rate ratio 2.74
(1.40 to 5.33)

237
(2 RCTs)

⊕⊝⊝⊝
Very low1,2

Adverse events (proportion) ‐ interferon‐alpha versus no intervention

10 per 1000

672 per 1000
(83 to 979)

OR 203
(9.01 to 4574.81)

33
(1 RCT)

⊕⊝⊝⊝
Very low1,2

Adverse events (proportion) ‐ interferon‐beta versus no intervention

10 per 1000

220 per 1000
(15 to 842)

OR 27.88
(1.48 to 526.12)

40
(1 RCT)

⊕⊝⊝⊝
Very low1,2

Adverse events: there were no adverse events in the comparison MTH‐68/B vaccine versus control. The number of adverse events was not reported for the comparison interferon‐alpha versus control. The proportion of people with adverse events and number of adverse events was not reported for the comparison pegylated interferon‐alpha plus ribavirin versus pegylated interferon‐alpha.

Adverse events (number) ‐ interferon‐beta versus no intervention

10 per 1000

147 per 1000
(10 to 748)

OR 17
(0.98 to 294.53)

40
(1 RCT)

⊕⊝⊝⊝
Very low1,2,3

Health‐related quality of life

None of the trials reported this outcome.

None of the trials reported health‐related quality of life, cirrhosis, decompensated liver disease, liver transplantation, or hepatocellular carcinoma.

*The basis for the assumed risk is the mean control group proportion (or control group rate) unless there were no events in the control group when the control group proportion (or control group rate) was considered as 1%. 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; OR: odds ratio; RCT: randomised clinical trial.

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 Downgraded 2 levels for high risk of bias.
2 Downgraded 1 level for small sample size (i.e. imprecision).
3 Downgraded 1 level for wide confidence intervals (i.e. imprecision).

Figuras y tablas -
Summary of findings for the main comparison. Intervention versus no intervention or control intervention (control) for acute hepatitis C infection: primary outcomes
Summary of findings 2. Intervention versus no intervention or control intervention (control) for acute hepatitis C infection: secondary outcomes

Intervention versus no intervention or control intervention (control) for acute hepatitis C infection: secondary outcomes

Patient or population: people with acute hepatitis C infection

Intervention: multiple (see below)

Control: multiple (see below)

Settings: secondary or tertiary care

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(trials)

Quality of the evidence
(GRADE)

Assumed risk

Corresponding risk

Control

Intervention

Liver transplantation

None of the trials reported this outcome.

Decompensated liver disease

None of the trials reported this outcome.

Cirrhosis

None of the trials reported this outcome.

Hepatocellular carcinoma

None of the trials reported this outcome.

Chronic HCV infection‐ interferon‐alpha versus no intervention

848 per 1000

601 per 1000
(334 to 809)

OR 0.27
(0.09 to 0.76)

99
(3 RCTs)

⊕⊝⊝⊝
Very low1,2

Chronic HCV infection‐ interferon‐beta versus no intervention

833 per 1000

259 per 1000
(0 to 861)

OR 0.07
(0 to 1.24)

90
(3 RCTs)

⊕⊝⊝⊝
Very low1,2,3,4

Chronic HCV infection‐ MTH‐68/B vaccine versus no intervention

263 per 1000

91 per 1000
(18 to 371)

OR 0.28
(0.05 to 1.65)

41
(1 RCT)

⊕⊝⊝⊝
Very low1,2,3

Chronic HCV infection‐ pegylated interferon‐alpha plus ribavirin versus pegylated interferon‐alpha

204 per 1000

181 per 1000
(95 to 315)

OR 0.86
(0.41 to 1.79)

237
(2 RCTs)

⊕⊝⊝⊝
Very low1,2,3

*The basis for the assumed risk is the mean control group proportion. 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).

†Chronic HCV infection was measured by absence of sustained virological response (i.e. the presence of circulating virus at least 6 months after cessation of treatment).

CI: confidence interval; HCV: hepatitis C virus; OR: odds ratio; RCT: randomised clinical trial.

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 Downgraded 2 levels for high risk of bias (i.e. within study risk of bias).
2 Downgraded 1 level for small sample size (i.e. imprecision).
3 Downgraded 1 level for wide confidence intervals (i.e. imprecision).
4 Downgraded 1 level (substantial heterogeneity in magnitude of effect) (i.e. heterogeneity).

Figuras y tablas -
Summary of findings 2. Intervention versus no intervention or control intervention (control) for acute hepatitis C infection: secondary outcomes
Table 1. Characteristics table

Study name

Intervention

Control

Period of follow‐up (months)

Randomisation

Blinding of participants and healthcare professionals

Blinding of outcome assessors

Missing outcome bias

Selective outcome reporting bias

For‐profit bias

Genesca 1993

Interferon‐alpha

No intervention

12

Unclear

High

Unclear

Low

High

Unclear

Hwang 1994

Interferon‐alpha

No intervention

12

Unclear

High

Unclear

High

Low

High

Lampertico 1994

Interferon‐alpha

No intervention

18

Unclear

High

Unclear

High

High

High

Omata 1991

Interferon‐beta

No intervention

36

Low

High

Unclear

High

High

Unclear

Omata 1994

Interferon‐beta

No intervention

36

Unclear

High

Unclear

Low

High

Unclear

Calleri 1998

Interferon‐beta

No intervention

22.5

Unclear

High

High

Low

Low

Low

Csatary 1998

MTH‐68/B vaccine

No intervention

12

Unclear

High

Unclear

Low

Low

Unclear

Wang 2005

Pegylated interferon‐alpha

No intervention

6

Unclear

Unclear

Unclear

High

High

Unclear

Deterding 2013

Pegylated interferon‐alpha

Pegylated interferon‐alpha plus ribavirin

6

Unclear

High

Low

High

Low

High

Santantonio 2014

Pegylated interferon‐alpha

Pegylated interferon‐alpha plus ribavirin

12

Unclear

High

Unclear

Low

High

High

Figuras y tablas -
Table 1. Characteristics table
Comparison 1. Intervention versus control

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Serious adverse events (proportion) Show forest plot

4

Odds Ratio (M‐H, Fixed, 95% CI)

Subtotals only

1.1 Interferon‐beta versus no intervention

1

40

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.2 MTH‐68/B vaccine versus no intervention

1

41

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.3 Pegylated interferon‐alpha plus ribavirin versus pegylated interferon‐alpha

2

237

Odds Ratio (M‐H, Fixed, 95% CI)

1.72 [0.70, 4.21]

2 Serious adverse events (number) Show forest plot

4

Rate Ratio (Fixed, 95% CI)

Subtotals only

2.1 Interferon‐beta versus no intervention

1

40

Rate Ratio (Fixed, 95% CI)

0.0 [0.0, 0.0]

2.2 MTH‐68/B vaccine versus no intervention

1

41

Rate Ratio (Fixed, 95% CI)

0.0 [0.0, 0.0]

2.3 Pegylated interferon‐alpha plus ribavirin versus pegylated interferon‐alpha

2

237

Rate Ratio (Fixed, 95% CI)

2.74 [1.40, 5.33]

3 Adverse events (proportion) Show forest plot

3

Odds Ratio (M‐H, Fixed, 95% CI)

Subtotals only

3.1 Interferon‐alpha versus no intervention

1

33

Odds Ratio (M‐H, Fixed, 95% CI)

203.0 [9.01, 4574.81]

3.2 Interferon‐beta versus no intervention

1

40

Odds Ratio (M‐H, Fixed, 95% CI)

27.88 [1.48, 526.12]

3.3 MTH‐68/B vaccine versus no intervention

1

41

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4 Adverse events (number) Show forest plot

2

Odds Ratio (Fixed, 95% CI)

Subtotals only

4.1 Interferon‐beta versus no intervention

1

40

Odds Ratio (Fixed, 95% CI)

17.00 [0.98, 294.53]

4.2 MTH‐68/B vaccine versus no intervention

1

41

Odds Ratio (Fixed, 95% CI)

0.0 [0.0, 0.0]

5 Chronic HCV infection Show forest plot

9

Odds Ratio (M‐H, Random, 95% CI)

Subtotals only

5.1 Interferon‐alpha versus no intervention

3

99

Odds Ratio (M‐H, Random, 95% CI)

0.27 [0.09, 0.76]

5.2 Interferon‐beta versus no intervention

3

90

Odds Ratio (M‐H, Random, 95% CI)

0.07 [0.00, 1.24]

5.3 MTH‐68/B vaccine versus no intervention

1

41

Odds Ratio (M‐H, Random, 95% CI)

0.28 [0.05, 1.65]

5.4 Pegylated interferon‐alpha plus ribavirin versus pegylated interferon‐alpha

2

237

Odds Ratio (M‐H, Random, 95% CI)

0.86 [0.41, 1.79]

Figuras y tablas -
Comparison 1. Intervention versus control