Scolaris Content Display Scolaris Content Display

Les antiviraux à action directe pour l'hépatite C chronique.

Contraer todo Desplegar todo

Referencias

References to studies included in this review

ADVANCE 2011a1 {published data only}

Brogan AJ, Talbird SE, Thompson JR, Miller JD, Rubin J, Deniz B. Cost‐effectiveness of telaprevir combination therapy for chronic hepatitis C. PLoS ONE 2014;9(3):e90295. CENTRAL
Curtis S, Cure S, Gavart S, Dearden L, Fleischmann J, Ouwens M, et al. The cost‐effectiveness of telaprevir (TVR) in combination with pegylated interferon‐alfa and ribavirin (PR) for the treatment of genotype 1 chronic hepatitis C patients. Journal of Hepatology2012; Vol. 56, issue Suppl 2:S434. CENTRAL
Di Bisceglie AM, Dusheiko GM, Muir AJ, Zeuzem S, Marcellin P, Bzowej NH. Telaprevir in combination with peginterferon alfa‐2a and ribavirin: analyses of pre‐defined subpopulations in the phase 3 ADVANCE trial. Gastroenterology 2011;140(5 Suppl 1):S908. CENTRAL
Dusheiko GM, Bengtsson L, Adda N, Kauffman R, Jacobson IM. Telaprevir in combination with peginterferon and ribavirin in genotype 1 HCV treatment‐naive patients: final results of phase 3 Advance study. Gut 2011;60(Suppl 2):A32‐3. CENTRAL
Everson G, Dusheiko G, Pol S, Nelson D, Muir A, Andreone P, et al. Telaprevir in combination with peginterferon alfa‐2a and ribavirin increased sustained virologic response in genotype 1 chronic HCV and allowed for shorter treatment duration in treatment‐naive and prior relapser patients. American Journal of Gastroenterology 2011;106(2S):S113‐4. CENTRAL
Jacobson IM, McHutchison JG, Dusheiko G, Di Bisceglie AM, Reddy KR, Bzowej NH, et al. Telaprevir for previously untreated chronic hepatitis C virus infection. New England Journal of Medicine 2011;364(25):2405‐16. CENTRAL
Jacobson IM, McHutchison JG, Dusheiko GM, Di Bisceglie AM, Reddy R, Bzowej NH, et al. Telaprevir in combination with peginterferon and ribavirin in genotype 1 HCV treatment‐naive patients: final results of phase 3 ADVANCE study. Hepatology 2010;52(Suppl S1):427A. CENTRAL
Kieffer TL, Bartels DJ, Sullivan J, Adiwijaya BS, Zhang EZ, Tigges A, et al. Clinical virology results from telaprevir phase 3 study ADVANCE. Hepatology 2010;52(Suppl S1):879A. CENTRAL
Luo D, Zeuzem S, Jacobson IM, Sherman K, Adda N, Wright CI, et al. High concordance between SVR12 and SVR24 in patients receiving telaprevir plus peginterferon and ribavirin in three phase iii clinical trials: Advance, Illuminate and Realize. Journal of Hepatology 2012;56(Suppl 2):S446‐7. CENTRAL
Marcellin P, Jacobson IM, Zeuzem S, Bzowej N, Muir AJ, Dusheiko G. Telaprevir in combination with peginterferon alfa‐2a and ribavirin: analyses of pre‐defined subpopulations in the phase 3 ADVANCE trial. Journal of Hepatology 2011;54(Suppl 1):S183‐4. CENTRAL
Rizzetto M, Andreone P, Colombo M, Scuteri A, Ciancio A, Smedile A, et al. ADVANCE study: final results of the phase III trial with telaprevir in combination with PEG‐IFN and RBV in treatment‐naive genotype 1 HCV. Digestive and Liver Disease 2011;43(Suppl 2):S69. CENTRAL
Sulkowski MS, Reddy R, Afdhal NH, Di Bisceglie AM, Zeuzem S, Poordad F, et al. Anemia had no effect on efficacy outcomes in treatment‐naive patients who received telaprevir‐based regimen in the Advance and Illuminate phase 3 studies. Journal of Hepatology 2011;54(Suppl 1):S195. CENTRAL

ADVANCE 2011a2 {published data only}

Brogan AJ, Talbird SE, Thompson JR, Miller JD, Rubin J, Deniz B. Cost‐effectiveness of telaprevir combination therapy for chronic hepatitis C. PLoS ONE 2014;9(3):e90295. CENTRAL
Curtis S, Cure S, Gavart S, Dearden L, Fleischmann J, Ouwens M, et al. The cost‐effectiveness of telaprevir (TVR) in combination with pegylated interferon‐alfa and ribavirin (PR) for the treatment of genotype 1 chronic hepatitis C patients. Journal of Hepatology2012; Vol. 56, issue Suppl 2:S434. CENTRAL
Di Bisceglie AM, Dusheiko GM, Muir AJ, Zeuzem S, Marcellin P, Bzowej NH. Telaprevir in combination with peginterferon alfa‐2a and ribavirin: analyses of pre‐defined subpopulations in the phase 3 ADVANCE trial. Gastroenterology 2011;140(5 Suppl 1):S908. CENTRAL
Dusheiko GM, Bengtsson L, Adda N, Kauffman R, Jacobson IM. Telaprevir in combination with peginterferon and ribavirin in genotype 1 HCV treatment‐naive patients: final results of phase 3 Advance study. Gut 2011;60(Suppl 2):A32‐3. CENTRAL
Everson G, Dusheiko G, Pol S, Nelson D, Muir A, Andreone P, et al. Telaprevir in combination with peginterferon alfa‐2a and ribavirin increased sustained virologic response in genotype 1 chronic HCV and allowed for shorter treatment duration in treatment‐naive and prior relapser patients. American Journal of Gastroenterology 2011;106(2S):S113‐4. CENTRAL
Jacobson IM, McHutchison JG, Dusheiko G, Di Bisceglie AM, Reddy KR, Bzowej NH, et al. Telaprevir for previously untreated chronic hepatitis C virus infection. New England Journal of Medicine 2011;364(25):2405‐16. CENTRAL
Jacobson IM, McHutchison JG, Dusheiko GM, Di Bisceglie AM, Reddy R, Bzowej NH, et al. Telaprevir in combination with peginterferon and ribavirin in genotype 1 HCV treatment‐naive patients: final results of phase 3 ADVANCE study. Hepatology 2010;52(Suppl S1):427A. CENTRAL
Kieffer TL, Bartels DJ, Sullivan J, Adiwijaya BS, Zhang EZ, Tigges A, et al. Clinical virology results from telaprevir phase 3 study ADVANCE. Hepatology 2010;52(Suppl S1):879A. CENTRAL
Luo D, Zeuzem S, Jacobson IM, Sherman K, Adda N, Wright CI, et al. High concordance between SVR12 and SVR24 in patients receiving telaprevir plus peginterferon and ribavirin in three phase iii clinical trials: Advance, Illuminate and Realize. Journal of Hepatology 2012;56(Suppl 2):S446‐7. CENTRAL
Marcellin P, Jacobson IM, Zeuzem S, Bzowej N, Muir AJ, Dusheiko G. Telaprevir in combination with peginterferon alfa‐2a and ribavirin: analyses of pre‐defined subpopulations in the phase 3 ADVANCE trial. Journal of Hepatology 2011;54(Suppl 1):S183‐4. CENTRAL
Rizzetto M, Andreone P, Colombo M, Scuteri A, Ciancio A, Smedile A, et al. ADVANCE study: final results of the phase III trial with telaprevir in combination with PEG‐IFN and RBV in treatment‐naive genotype 1 HCV. Digestive and Liver Disease 2011;43(Suppl 2):S69. CENTRAL
Sulkowski MS, Reddy R, Afdhal NH, Di Bisceglie AM, Zeuzem S, Poordad F, et al. Anemia had no effect on efficacy outcomes in treatment‐naive patients who received telaprevir‐based regimen in the Advance and Illuminate phase 3 studies. Journal of Hepatology 2011;54(Suppl 1):S195. CENTRAL

Anderson 2014a1 {published data only}

Anderson RT, Baran RW, Erickson P, Revicki DA, Dietz B, Gooch K. Psychometric evaluation of the hepatitis C virus patient‐reported outcomes (HCV‐PRO) instrument: validity, responsiveness, and identification of the minimally important difference in a phase 2 clinical trial. Quality of Life Research 2014;23(3):877‐86. CENTRAL
Gaultier I, Cohen DE, Bhathena A, Idler K, Larsen LM, Podsadecki T, et al. The effect of IL28B polymorphisms on virologic response to treatment with pegylated interferon alpha‐2A and ribavirin (SOC) added to ABT‐450/ritonavir (ABT‐450/R), ABT‐333, or ABT‐072. Journal of Hepatology 2011;54(Suppl 1):S523. CENTRAL
Pilot‐Matias TJ, Tripathi RL, Dekhtyar T, Menon RM, Gaultier IA, Cohen DE, et al. Genotypic and phenotypic characterization of NS3 variants selected in HCV‐infected patients treated with ABT‐450. Journal of Hepatology 2011;54(Suppl 1):S485‐6. CENTRAL

Anderson 2014a2 {published data only}

Anderson RT, Baran RW, Erickson P, Revicki DA, Dietz B, Gooch K. Psychometric evaluation of the hepatitis C virus patient‐reported outcomes (HCV‐PRO) instrument: validity, responsiveness, and identification of the minimally important difference in a phase 2 clinical trial. Quality of Life Research 2014;23(3):877‐86. CENTRAL
Gaultier I, Cohen DE, Bhathena A, Idler K, Larsen LM, Podsadecki T, et al. The effect of IL28B polymorphisms on virologic response to treatment with pegylated interferon alpha‐2A and ribavirin (SOC) added to ABT‐450/ritonavir (ABT‐450/R), ABT‐333, or ABT‐072. Journal of Hepatology 2011;54(Suppl 1):S523. CENTRAL
Pilot‐Matias TJ, Tripathi RL, Dekhtyar T, Menon RM, Gaultier IA, Cohen DE, et al. Genotypic and phenotypic characterization of NS3 variants selected in HCV‐infected patients treated with ABT‐450. Journal of Hepatology 2011;54(Suppl 1):S485‐6. CENTRAL

Anderson 2014a3 {published data only}

Anderson RT, Baran RW, Erickson P, Revicki DA, Dietz B, Gooch K. Psychometric evaluation of the hepatitis C virus patient‐reported outcomes (HCV‐PRO) instrument: validity, responsiveness, and identification of the minimally important difference in a phase 2 clinical trial. Quality of Life Research 2014;23(3):877‐86. CENTRAL
Gaultier I, Cohen DE, Bhathena A, Idler K, Larsen LM, Podsadecki T, et al. The effect of IL28B polymorphisms on virologic response to treatment with pegylated interferon alpha‐2A and ribavirin (SOC) added to ABT‐450/ritonavir (ABT‐450/R), ABT‐333, or ABT‐072. Journal of Hepatology 2011;54(Suppl 1):S523. CENTRAL
Pilot‐Matias TJ, Tripathi RL, Dekhtyar T, Menon RM, Gaultier IA, Cohen DE, et al. Genotypic and phenotypic characterization of NS3 variants selected in HCV‐infected patients treated with ABT‐450. Journal of Hepatology 2011;54(Suppl 1):S485‐6. CENTRAL

Anderson 2014a4 {published data only}

Anderson RT, Baran RW, Erickson P, Revicki DA, Dietz B, Gooch K. Psychometric evaluation of the hepatitis C virus patient‐reported outcomes (HCV‐PRO) instrument: validity, responsiveness, and identification of the minimally important difference in a phase 2 clinical trial. Quality of Life Research 2014;23(3):877‐86. CENTRAL
Gaultier I, Cohen DE, Bhathena A, Idler K, Larsen LM, Podsadecki T, et al. The effect of IL28B polymorphisms on virologic response to treatment with pegylated interferon alpha‐2A and ribavirin (SOC) added to ABT‐450/ritonavir (ABT‐450/R), ABT‐333, or ABT‐072. Journal of Hepatology 2011;54(Suppl 1):S523. CENTRAL
Pilot‐Matias TJ, Tripathi RL, Dekhtyar T, Menon RM, Gaultier IA, Cohen DE, et al. Genotypic and phenotypic characterization of NS3 variants selected in HCV‐infected patients treated with ABT‐450. Journal of Hepatology 2011;54(Suppl 1):S485‐6. CENTRAL

Anderson 2014a5 {published data only}

Anderson RT, Baran RW, Erickson P, Revicki DA, Dietz B, Gooch K. Psychometric evaluation of the hepatitis C virus patient‐reported outcomes (HCV‐PRO) instrument: validity, responsiveness, and identification of the minimally important difference in a phase 2 clinical trial. Quality of Life Research 2014;23(3):877‐86. CENTRAL
Gaultier I, Cohen DE, Bhathena A, Idler K, Larsen LM, Podsadecki T, et al. The effect of IL28B polymorphisms on virologic response to treatment with pegylated interferon alpha‐2A and ribavirin (SOC) added to ABT‐450/ritonavir (ABT‐450/R), ABT‐333, or ABT‐072. Journal of Hepatology 2011;54(Suppl 1):S523. CENTRAL
Pilot‐Matias TJ, Tripathi RL, Dekhtyar T, Menon RM, Gaultier IA, Cohen DE, et al. Genotypic and phenotypic characterization of NS3 variants selected in HCV‐infected patients treated with ABT‐450. Journal of Hepatology 2011;54(Suppl 1):S485‐6. CENTRAL

Anderson 2014a6 {published data only}

Anderson RT, Baran RW, Erickson P, Revicki DA, Dietz B, Gooch K. Psychometric evaluation of the hepatitis C virus patient‐reported outcomes (HCV‐PRO) instrument: validity, responsiveness, and identification of the minimally important difference in a phase 2 clinical trial. Quality of Life Research 2014;23(3):877‐86. CENTRAL
Gaultier I, Cohen DE, Bhathena A, Idler K, Larsen LM, Podsadecki T, et al. The effect of IL28B polymorphisms on virologic response to treatment with pegylated interferon alpha‐2A and ribavirin (SOC) added to ABT‐450/ritonavir (ABT‐450/R), ABT‐333, or ABT‐072. Journal of Hepatology 2011;54(Suppl 1):S523. CENTRAL
Pilot‐Matias TJ, Tripathi RL, Dekhtyar T, Menon RM, Gaultier IA, Cohen DE, et al. Genotypic and phenotypic characterization of NS3 variants selected in HCV‐infected patients treated with ABT‐450. Journal of Hepatology 2011;54(Suppl 1):S485‐6. CENTRAL

Anderson 2014a7 {published data only}

Anderson RT, Baran RW, Erickson P, Revicki DA, Dietz B, Gooch K. Psychometric evaluation of the hepatitis C virus patient‐reported outcomes (HCV‐PRO) instrument: validity, responsiveness, and identification of the minimally important difference in a phase 2 clinical trial. Quality of Life Research 2014;23(3):877‐86. CENTRAL
Gaultier I, Cohen DE, Bhathena A, Idler K, Larsen LM, Podsadecki T, et al. The effect of IL28B polymorphisms on virologic response to treatment with pegylated interferon alpha‐2A and ribavirin (SOC) added to ABT‐450/ritonavir (ABT‐450/R), ABT‐333, or ABT‐072. Journal of Hepatology 2011;54(Suppl 1):S523. CENTRAL
Pilot‐Matias TJ, Tripathi RL, Dekhtyar T, Menon RM, Gaultier IA, Cohen DE, et al. Genotypic and phenotypic characterization of NS3 variants selected in HCV‐infected patients treated with ABT‐450. Journal of Hepatology 2011;54(Suppl 1):S485‐6. CENTRAL

Anderson 2014a8 {published data only}

Anderson RT, Baran RW, Erickson P, Revicki DA, Dietz B, Gooch K. Psychometric evaluation of the hepatitis C virus patient‐reported outcomes (HCV‐PRO) instrument: validity, responsiveness, and identification of the minimally important difference in a phase 2 clinical trial. Quality of Life Research 2014;23(3):877‐86. CENTRAL
Gaultier I, Cohen DE, Bhathena A, Idler K, Larsen LM, Podsadecki T, et al. The effect of IL28B polymorphisms on virologic response to treatment with pegylated interferon alpha‐2A and ribavirin (SOC) added to ABT‐450/ritonavir (ABT‐450/R), ABT‐333, or ABT‐072. Journal of Hepatology 2011;54(Suppl 1):S523. CENTRAL
Pilot‐Matias TJ, Tripathi RL, Dekhtyar T, Menon RM, Gaultier IA, Cohen DE, et al. Genotypic and phenotypic characterization of NS3 variants selected in HCV‐infected patients treated with ABT‐450. Journal of Hepatology 2011;54(Suppl 1):S485‐6. CENTRAL

Anonymous (PPI‐461) 2011a1 {unpublished data only}

NCT01247194. A phase 1b study of PPI‐461 in patients with HCV genotype 1. clinicaltrials.gov/ct2/show/study/NCT01247194?term=NCT01247194&rank=1 (first received 22 November 2010). CENTRAL

Anonymous (PPI‐461) 2011a2 {unpublished data only}

NCT01247194. A phase 1b study of PPI‐461 in patients with HCV genotype 1. clinicaltrials.gov/ct2/show/study/NCT01247194?term=NCT01247194&rank=1 (first received 22 November 2010). CENTRAL

Anonymous (PPI‐461) 2011a3 {unpublished data only}

NCT01247194. A phase 1b study of PPI‐461 in patients with HCV genotype 1. clinicaltrials.gov/ct2/show/study/NCT01247194?term=NCT01247194&rank=1 (first received 22 November 2010). CENTRAL

ASPIRE 2014 {published data only}

Ferenci P, Fried M, Poordad F, Zeuzem S, Lenz O, Ouwerkerk‐Mahadevan S, et al. Safety of simeprevir (SMV) with peg Ifnalpha‐2a/ribavirin (PR) in HCV genotype 1‐infected patients: PILLAR and ASPIRE trials. Swiss Medical Weekly 2013;143(Suppl 199):15S. CENTRAL
Lenz O, Fevery B, Vijgen L, Verbeeck J, Peeters M, Beumont‐Mauviel M, et al. TMC435 in patients infected with HCV genotype 1 who have failed previous pegylated interferon/ribavirin treatment: virologic analyses of the ASPIRE trial. Journal of Hepatology 2012;56(Suppl S2):S5. CENTRAL
Scott J, Rosa K, Fu M, Cerri K, Peeters M, Beumont M, et al. Fatigue during treatment for hepatitis C virus: results of self‐reported fatigue severity in two Phase IIb studies of simeprevir treatment in patients with hepatitis C virus genotype 1 infection. BMC Infectious Diseases 2014;14:465. CENTRAL
Scott J, Rosa K, Fu M, Cerri K, Peeters M, Beumont‐Mauviel M, et al. Fatigue severity scale: reliability, validity and interpretation of change‐evidence from two clinical trials in patients with chronic HCV infection. Value in Health 2013;16(3):A216. CENTRAL
Scott J, Rosa K, Zeuzem S, Beumont‐Mauviel M, Peeters M, Cerri, K, et al. Adding simeprevir (TMC435) to pegylated interferon/ribavirin does not increase patient reported fatigue in treatment‐experienced patients with chronic HCV infection: results from the ASPIRE trial. Journal of Hepatology 2013;58(Suppl S1):S371. CENTRAL
Zeuzem S, Berg T, Gane E, Ferenci P, Foster GR, Fried MW, et al. Simeprevir increases rate of sustained virologic response among treatment‐experienced patients with HCV genotype‐1 infection: a phase IIb trial. Gastroenterology 2014;146(2):430‐41. CENTRAL
Zeuzem S, Foster GR, Fried MW, Hezode C, Hirschfield GM, Nikitin I, et al. The ASPIRE trial: TMC435 in treatment‐experienced patients with genotype‐1 HCV infection who have failed previous PEGIFN/RBV treatment. Journal of Hepatology 2011;54(Suppl 1):S546. CENTRAL

ATLAS 2013 {published data only}

Marcellin P, Cooper C, Balart L, Larrey D, Box T, Yoshida E, et al. Randomized controlled trial of danoprevir plus peginterferon alfa‐2a and ribavirin in treatment‐naïve patients with hepatitis C virus genotype 1 infection. Gastroenterology 2013;145:790‐800. CENTRAL
Marcellin P, Cooper C, Balart LA, Larrey DG, Box TD, Yoshida E, et al. High sustained virological response rates with response‐guided danoprevir plus peginterferon alfa‐2a (40KD) and ribavirin in treatment‐naive, HCV genotype 1 patients: ATLAS study final results. Journal of Hepatology 2012;56(Suppl S2):S472. CENTRAL
Terrault N, Cooper C, Balart LA, Larrey DG, Box TD, Yoshida E, et al. High sustained virologic response (SVR24) rates with response‐guided danoprevir (DNV; RG7227) plus pegIfn alpha‐2A (40KD) and ribavirin (P/R) in treatment‐naive HCV genotype 1 (G1) patients: results from the ATLAS study. Hepatology 2011;54(S1):398A‐9A. CENTRAL
Terrault N, Cooper C, Balart LA, Larrey DG, Box TD, Yoshida EM, et al. Phase II randomised, partially‐blind, parallel‐group study of oral danoprevir (RG7227) with PegIFNα‐2a (Pegasys) plus ribavirin (Copegus) in treatment‐naive genotype 1 patients with CHC: results of planned week 12 interim analysis of the ATLAS study. Hepatology 2010;52(Suppl S1):335A. CENTRAL

Bacon 2011a1 {published data only}

Bacon BR, Bruno S, Schiff ER, Kwo PY, Buti M, Pedicone L, et al. Predictors of sustained virologic response (SVR) among poor interferon (IFN) responders when boceprevir (BOC) is added to peginterferon alfa‐2b/ribavirin (PR). Hepatology 2011;54(S1):376A‐7A. CENTRAL
Bacon BR, Gordon SC, Lawitz E, Marcellin P, Vierling JM, Zeuzem S, et al. Boceprevir for previously treated chronic HCV genotype 1 infection. New England Journal of Medicine 2011;364(13):1207‐17. CENTRAL
Bacon BR, Gordon SC, Lawitz E, Marcellin P, Vierling JM, Zeuzem S, et al. HCV RESPOND‐2 final results: high sustained virologic response among genotype 1 previous non‐responders and relapsers to peginterferon/ribavirin when re‐treated with boceprevir plus PEGINTRON (peginterferon alfa‐2b)/ribavirin. Hepatology 2010;52(Suppl S1):430A. CENTRAL
Barnard RJ, Howe JA, Ogert RA, Zeuzem S, Poordad F, Gordon SC, et al. Analysis of boceprevir resistance associated amino acid variants (RAVs) in two phase 3 boceprevir clinical studies. Virology 2013;444(1‐2):329‐36. CENTRAL
Bognar FA, Hu KQ, Thompson S, Pedicone L, Wahl J, Lin SG. Boceprevir (BOC) plus peginterferon alfa‐2b/ ribavirin (PR) in the treatment of chronic hepatitis C virus genotype‐1 (HCV‐G1) infected Asian patients in the SPRINT‐1, SPRINT‐2 and RESPOND‐2 trials. Journal of Gastroenterology and Hepatology 2012;27(Suppl 4):161. CENTRAL
Brass C, Barnard RJO, Howe JA, Ogert RA, Ralston R, Boparai N, et al. Sustained virologic response and boceprevir resistance‐associated variants observed in patients infected with HCV genotpype Ia/Ib when treated with boceprevir plus peginterferon alfa‐2b/ribavirin. Journal of Hepatology 2011;54(Suppl 1):S471‐2. CENTRAL
Bruno S, Vierling JM, Esteban R, Nyberg LM, Tanno H, Albrecht JK, et al. Boceprevir in addition to standard of care enhanced SVR in hepatitis C virus (HCV) genotype‐1 with advanced fibrosis/cirrhosis: subgroup analysis of sprint‐2 and respond‐2 studies. Journal of Hepatology 2011;54(Suppl 1):S4‐5. CENTRAL
Bruno S, Vierling JM, Esteban R, Nyberg LM, Tanno H, Goodman Z, et al. Efficacy and safety of boceprevir plus peginterferon‐ribavirin in patients with HCV G1 infection and advanced fibrosis/cirrhosis. Journal of Hepatology 2013;58(3):479‐87. CENTRAL
Chhatwal J, Ferrante SA, Brass C, El Khoury AC, Burroughs M, Bacon B, et al. Cost‐effectiveness of boceprevir in patients previously treated for chronic hepatitis C genotype 1 infection in the United States. Value in Health 2013;16(6):973‐86. CENTRAL
Chhatwal J, Ferrante SA, Dasbach EJ, El Khoury A, Burroughs M, Bacon B, et al. Projecting the long‐term clinical impact of boceprevir in patients with chronic hepatitis C genotype‐1 who failed prior treatment with peginterferon/ribavirin. Journal of Hepatology 2011;54:S164. CENTRAL
Chhatwal J, Ferrante SA, Dasbach EJ, Khoury A, Brass CA, Burroughs M, et al. Cost‐effectiveness of boceprevir use in patients with chronic hepatitis C genotype‐1 who failed prior treatment with peginterferon/ribavirin. Hepatology 2011;54(4 (Suppl)):801A‐2A. CENTRAL
Chhatwal J, Lundberg J, Ferrante S, El Khourhy AC, Oksanen A, Elbasha EH. Cost‐effectiveness of boceprevir in the treatment of chronic Hepatitis C genotype 1 in Sweden. Journal of Hepatology 2012;56(Suppl 2):S386‐7. CENTRAL
Gordon SC, Sulkowski MS, Jacobson IM, Vierling JM, Davis GL, Afdhal NH, et al. Reasons for screen failure in 2 phase III studies of boceprevir with peginterferon/ribavirin in patients with chronic HCV genotype 1 infection (sprint‐2 and respond‐2). Hepatology 2011;54(Suppl S1):848A‐9A. CENTRAL
Lenz O, Fevery B, Vijgen L, Verbeeck J, Peeters M, Beumont M, et al. TMC435 in combination with peginterferon alpha‐2A/ribavirin in treatment‐naïve patients infected with HCV genotype 1: virology analysis of the PILLAR study. Hepatology 2011;54(4 (Suppl)):985A. CENTRAL
Marcellin P, Vierling JM, Alric L, Pirisi M, Tran A, Albrecht JK, et al. Efficacy among North American and European HCV genotype‐1 treatment‐naive and previous non‐responders and relapsers to peginterferon/ribavirin when treated with boceprevir plus peginterferon alfa‐2b/ribavirin. Journal of Hepatology 2011;54(Suppl 1):S184. CENTRAL
Sulkowski MS, Bacon BR, Gordon SC, Lawitz E, Marcellin P, Vierling JM, et al. Boceprevir combined with peginterferon alfa‐2b/ribavirin for previous treatment failure patients with hepatitis C virus genotype 1: RESPOND‐2 final results. Journal of the International Association of Physicians in AIDS Care 2011;10(3):198. CENTRAL
Zeuzem S, Vierling JM, Esteban R, Gibas AL, Pol S, Boparai N, et al. Predictors of sustained virologic response among genotype 1 previous non‐responders and relapsers to peginterferon/ribavirin when re‐treated with boceprevir plus peginterferon alfa‐2b/ribavirin. Journal of Hepatology 2011;54(Suppl 1):S198‐9. CENTRAL

Bacon 2011a2 {published data only}

Bacon BR, Bruno S, Schiff ER, Kwo PY, Buti M, Pedicone L, et al. Predictors of sustained virologic response (SVR) among poor interferon (IFN) responders when boceprevir (BOC) is added to peginterferon alfa‐2b/ribavirin (PR). Hepatology 2011;54(S1):376A‐7A. CENTRAL
Bacon BR, Gordon SC, Lawitz E, Marcellin P, Vierling JM, Zeuzem S, et al. Boceprevir for previously treated chronic HCV genotype 1 infection. New England Journal of Medicine 2011;364(13):1207‐17. CENTRAL
Bacon BR, Gordon SC, Lawitz E, Marcellin P, Vierling JM, Zeuzem S, et al. HCV RESPOND‐2 final results: high sustained virologic response among genotype 1 previous non‐responders and relapsers to peginterferon/ribavirin when re‐treated with boceprevir plus PEGINTRON (peginterferon alfa‐2b)/ribavirin. Hepatology 2010;52(Suppl S1):430A. CENTRAL
Barnard RJ, Howe JA, Ogert RA, Zeuzem S, Poordad F, Gordon SC, et al. Analysis of boceprevir resistance associated amino acid variants (RAVs) in two phase 3 boceprevir clinical studies. Virology 2013;444(1‐2):329‐36. CENTRAL
Bognar FA, Hu KQ, Thompson S, Pedicone L, Wahl J, Lin SG. Boceprevir (BOC) plus peginterferon alfa‐2b/ ribavirin (PR) in the treatment of chronic hepatitis C virus genotype‐1 (HCV‐G1) infected Asian patients in the SPRINT‐1, SPRINT‐2 and RESPOND‐2 trials. Journal of Gastroenterology and Hepatology 2012;27(Suppl 4):161. CENTRAL
Brass C, Barnard RJO, Howe JA, Ogert RA, Ralston R, Boparai N, et al. Sustained virologic response and boceprevir resistance‐associated variants observed in patients infected with HCV genotpype Ia/Ib when treated with boceprevir plus peginterferon alfa‐2b/ribavirin. Journal of Hepatology 2011;54(Suppl 1):S471‐2. CENTRAL
Bruno S, Vierling JM, Esteban R, Nyberg LM, Tanno H, Albrecht JK, et al. Boceprevir in addition to standard of care enhanced SVR in hepatitis C virus (HCV) genotype‐1 with advanced fibrosis/cirrhosis: subgroup analysis of sprint‐2 and respond‐2 studies. Journal of Hepatology 2011;54(Suppl 1):S4‐5. CENTRAL
Bruno S, Vierling JM, Esteban R, Nyberg LM, Tanno H, Goodman Z, et al. Efficacy and safety of boceprevir plus peginterferon‐ribavirin in patients with HCV G1 infection and advanced fibrosis/cirrhosis. Journal of Hepatology 2013;58(3):479‐87. CENTRAL
Chhatwal J, Ferrante SA, Brass C, El Khoury AC, Burroughs M, Bacon B, et al. Cost‐effectiveness of boceprevir in patients previously treated for chronic hepatitis C genotype 1 infection in the United States. Value in Health 2013;16(6):973‐86. CENTRAL
Chhatwal J, Ferrante SA, Dasbach EJ, El Khoury A, Burroughs M, Bacon B, et al. Projecting the long‐term clinical impact of boceprevir in patients with chronic hepatitis C genotype‐1 who failed prior treatment with peginterferon/ribavirin. Journal of Hepatology 2011;54:S164. CENTRAL
Chhatwal J, Ferrante SA, Dasbach EJ, Khoury A, Brass CA, Burroughs M, et al. Cost‐effectiveness of boceprevir use in patients with chronic hepatitis C genotype‐1 who failed prior treatment with peginterferon/ribavirin. Hepatology 2011;54(4 (Suppl)):801A‐2A. CENTRAL
Chhatwal J, Lundberg J, Ferrante S, El Khourhy AC, Oksanen A, Elbasha EH. Cost‐effectiveness of boceprevir in the treatment of chronic Hepatitis C genotype 1 in Sweden. Journal of Hepatology 2012;56(Suppl 2):S386‐7. CENTRAL
Gordon SC, Sulkowski MS, Jacobson IM, Vierling JM, Davis GL, Afdhal NH, et al. Reasons for screen failure in 2 phase III studies of boceprevir with peginterferon/ribavirin in patients with chronic HCV genotype 1 infection (sprint‐2 and respond‐2). Hepatology 2011;54(Suppl S1):848A‐9A. CENTRAL
Marcellin P, Vierling JM, Alric L, Pirisi M, Tran A, Albrecht JK, et al. Efficacy among North American and European HCV genotype‐1 treatment‐naive and previous non‐responders and relapsers to peginterferon/ribavirin when treated with boceprevir plus peginterferon alfa‐2b/ribavirin. Journal of Hepatology 2011;54(Suppl 1):S184. CENTRAL
Sulkowski MS, Bacon BR, Gordon SC, Lawitz E, Marcellin P, Vierling JM, et al. Boceprevir combined with peginterferon alfa‐2b/ribavirin for previous treatment failure patients with hepatitis C virus genotype 1: RESPOND‐2 final results. Journal of the International Association of Physicians in AIDS Care 2011;10(3):198. CENTRAL
Zeuzem S, Vierling JM, Esteban R, Gibas AL, Pol S, Boparai N, et al. Predictors of sustained virologic response among genotype 1 previous non‐responders and relapsers to peginterferon/ribavirin when re‐treated with boceprevir plus peginterferon alfa‐2b/ribavirin. Journal of Hepatology 2011;54(Suppl 1):S198‐S9. CENTRAL

Basu 2014a {published data only}

Basu P, Shah N, Aloysius M. Simeprevir and sofosbuvir with modified doses of ribavirin (RBV) therapy on telaprevir‐experienced, coinfected (with HIV) cirrhotics with chronic hepatitis C (CHC): a randomized, open‐label, clinical pilot study, STOP C, interim results. American Journal of Gastroenterology 2014;109(Suppl. 2):S177. CENTRAL
Basu P, Shah NJ, Aloysius MM, Brown RS. Interferon ineligible naive chronic hepatitis C genotype I subjects treated with simeprevir and sofosbuvir in special population (psychiatric). A clinical pilot study; Inspire C study; interim results. HPB: the Official Journal of the International Hepato Pancreato Biliary Association 2015;17(Suppl S2):46. CENTRAL

Bavisotto 2007 {published data only}

Bavisotto L, Wang CC, Jacobson IM, Marcellin P, Zeuzem S, Lawitz EJ, et al. Antiviral, pharmacokinetic and safety data for GS‐9190, a non‐nucleoside HCV NS5b polymerase inhibitor, in a phase‐1 trial in HCV genotype 1 infected subjects. Hepatology 2007;46(4 (Suppl 1)):255A. CENTRAL
Harris J, Bae A, Sun SC, Svarovskaia ES, Miller MD, Mo H, et al. Antiviral response and resistance analysis of treatment‐naive HCV infected subjects receiving single and multiple doses of GS ‐ 9190. Hepatology 2010;52(Suppl S1):722A. CENTRAL

Benhamou 2013a1 {published data only}

Benhamou Y, Moussalli J, Ratziu V, Lebray P, De Backer K, De Meyer S, et al. Telaprevir activity in treatment‐naive patients infected hepatitis C virus genotype 4: a randomized trial. Journal of Infectious Diseases 2013;208(6):1000‐7. CENTRAL
Benhamou Y, Moussalli J, Ratziu V, Lebray P, Gysen V, de Backer K, et al. Results of a proof of concept study (C210) of telaprevir monotherapy and in combination with peginterferon ALFA‐2A and ribavirin in treatment‐naive genotype 4 HCV patients. Journal of Hepatology 2009;50(Suppl 1):S6. CENTRAL
Benhamou Y, Moussalli J, Ratziu V, Lebray P, de Backer K, Ghys A, et al. Activity of telaprevir monotherapy or in combination with peginterferon‐alfa‐2a and ribavirin in treatment‐naive genotype 4 hepatitis C patients: final results of study C210. Hepatology 2010;52(Suppl S1):719A. CENTRAL
De Meyer S, Ghys A, Dierynck I, Beumont M, Luo D, Picchio G. Virologic characterization of genotype 4 hepatitis C virus variants in patients treated with telaprevir. Virology Journal 2014;11:93. CENTRAL

Benhamou 2013a2 {published data only}

Benhamou Y, Moussalli J, Ratziu V, Lebray P, De Backer K, De Meyer S, et al. Telaprevir activity in treatment‐naive patients infected hepatitis C virus genotype 4: a randomized trial. Journal of Infectious Diseases 2013;208(6):1000‐7. CENTRAL
Benhamou Y, Moussalli J, Ratziu V, Lebray P, Gysen V, de Backer K, et al. Results of a proof of concept study (C210) of telaprevir monotherapy and in combination with peginterferon ALFA‐2A and ribavirin in treatment‐naive genotype 4 HCV patients. Journal of Hepatology 2009;50(Suppl 1):S6. CENTRAL
Benhamou Y, Moussalli J, Ratziu V, Lebray P, de Backer K, Ghys A, et al. Activity of telaprevir monotherapy or in combination with peginterferon‐alfa‐2a and ribavirin in treatment‐naive genotype 4 hepatitis C patients: final results of study C210. Hepatology 2010;52(Suppl S1):719A. CENTRAL
De Meyer S, Ghys A, Dierynck I, Beumont M, Luo D, Picchio G. Virologic characterization of genotype 4 hepatitis C virus variants in patients treated with telaprevir. Virology Journal 2014;11:93. CENTRAL

Boehringer Ingelheim 2010a {published data only}

Boehringer Ingelheim. Safety, antiviral activity, and pharmcokinetics of multiple rising oral doses of BI201334 NA in treatment‐naive patients with chronic hepatitis C infection for 14 days monotherapy followed by combination with pegylated interferon and ribavirin for an additional 14 days (double‐blind, placebo controlled); and in treatment experienced patients with chronic hepatitis C infection for 28 days as combination therapy with pegylated interferon and ribavirin (open label). www.trials.boehringer‐ingelheim.com/public/trial_results_documents/1220/1220.2_U10‐2363‐01_PE_DS_DR.pdf (accessed 30 March 2016). CENTRAL

Boehringer Ingelheim 2010b {published data only}

Boehringer Ingelheim. A randomised, double‐blind, placebo controlled trial with 200 mg BILN 2061ZW given p.o. at two consecutive days bid to investigate the antiviral efficacy, pharmacokinetics, safety in patients with cirrhosis and chronic hepatitis C. www.trials.boehringer‐ingelheim.com/public/trial_results_documents/605/605.9_U03‐1671.pdf (accessed prior to 27 March 2017. CENTRAL

Bronowicki 2013a1 {published data only}

Bronowicki JP, Pol S, Thuluvath P, Larrey D, Martorell CT, Rustgi VK, et al. Asunaprevir (ASV; BMS‐650032), an NS3 protease inhibitor, in combination with peginterferon and ribavirin in treatment‐naive patients with genotype 1 chronic hepatitis C infection. Journal of Hepatology 2012;56(Suppl 2):S431‐2. CENTRAL
Bronowicki JP, Pol S, Thuluvath PJ, Larrey D, Martorell CT, Rustgi VK, et al. BMS‐650032, an NS3 inhibitor, in combination with peginterferon alpha‐2a and ribavirin in treatment‐naive subjects with genotype 1 chronic hepatitis C infection. Journal of Hepatology 2011;54:S472. CENTRAL
Bronowicki JP, Pol S, Thuluvath PJ, Larrey D, Martorell CT, Rustgi VK, et al. Randomized study of asunaprevir plus pegylated interferon‐alpha and ribavirin for previously untreated genotype 1 chronic hepatitis C. Antiviral Therapy 2013;18(7):885‐93. CENTRAL

Bronowicki 2013a2 {published data only}

Bronowicki JP, Pol S, Thuluvath P, Larrey D, Martorell CT, Rustgi VK, et al. Asunaprevir (ASV; BMS‐650032), an NS3 protease inhibitor, in combination with peginterferon and ribavirin in treatment‐naive patients with genotype 1 chronic hepatitis C infection. Journal of Hepatology 2012;56(Suppl 2):S431‐2. CENTRAL
Bronowicki JP, Pol S, Thuluvath PJ, Larrey D, Martorell CT, Rustgi VK, et al. BMS‐650032, an NS3 inhibitor, in combination with peginterferon alpha‐2a and ribavirin in treatment‐naive subjects with genotype 1 chronic hepatitis C infection. Journal of Hepatology 2011;54:S472. CENTRAL
Bronowicki JP, Pol S, Thuluvath PJ, Larrey D, Martorell CT, Rustgi VK, et al. Randomized study of asunaprevir plus pegylated interferon‐alpha and ribavirin for previously untreated genotype 1 chronic hepatitis C. Antiviral Therapy 2013;18(7):885‐93. CENTRAL

Bronowicki 2013a3 {published data only}

Bronowicki JP, Pol S, Thuluvath P, Larrey D, Martorell CT, Rustgi VK, et al. Asunaprevir (ASV; BMS‐650032), an NS3 protease inhibitor, in combination with peginterferon and ribavirin in treatment‐naive patients with genotype 1 chronic hepatitis C infection. Journal of Hepatology 2012;56(Suppl 2):S431‐2. CENTRAL
Bronowicki JP, Pol S, Thuluvath PJ, Larrey D, Martorell CT, Rustgi VK, et al. BMS‐650032, an NS3 inhibitor, in combination with peginterferon alpha‐2a and ribavirin in treatment‐naive subjects with genotype 1 chronic hepatitis C infection. Journal of Hepatology 2011;54:S472. CENTRAL
Bronowicki JP, Pol S, Thuluvath PJ, Larrey D, Martorell CT, Rustgi VK, et al. Randomized study of asunaprevir plus pegylated interferon‐alpha and ribavirin for previously untreated genotype 1 chronic hepatitis C. Antiviral Therapy 2013;18(7):885‐93. CENTRAL

Bronowicki 2014 {published data only}

Bronowicki J‐P, Ratziu V, Gadano A, Thuluvath Paul J, Bessone F, Martorell Claudia T, et al. Randomized trial of asunaprevir plus peginterferon alfa and ribavirin for previously untreated genotype 1 or 4 chronic hepatitis C. Journal of Hepatology 2014;61(6):1220‐7. CENTRAL
Bronowicki JP, Pol S, Thuluvath P, Larrey D, Martorell CT, Rustgi VK, et al. Asunaprevir (ASV; BMS‐650032), an NS3 protease inhibitor, in combination with peginterferon and ribavirin in treatment‐naive patients with genotype 1 chronic hepatitis C infection. Journal of Hepatology 2012;56(Suppl 2):S431‐S2. CENTRAL
Bronowicki JP, Ratziu V, Gadano A, Thuluvath PJ, Bessone F, Martorell CT, et al. Asunaprevir with peginterferon‐alfa and ribavirin in treatment‐naive patients with genotype‐1 or ‐4 chronic hepatitis C: SVR24 results from a randomized phase 2B study (AI447016). Journal of Hepatology 2013;58:S571‐2. CENTRAL

C‐EDGE CO STAR 2015 {published data only}

Dore G, Altice F, Litwin AH, Dalgard O, Gane EJ, Shibolet O, et al. C‐edge Co‐star: efficacy of grazoprevir and elbasvir in persons who inject drugs (PWID) receiving opioid agonist therapy. Hepatology 2015;62(Suppl S1):227A‐8A. CENTRAL

C‐EDGE TN 2015 {published data only}

Arduino JM, Wang Y, Brown DD, Khawaja S, Martinez E, Butterton JR, et al. C‐EDGE TN: Impact of 12‐week oral regimen of grazoprevir (GZR, MK‐5172)/Elbasvir (EBR, MK‐8742) on patient‐reported outcomes (PROs) in treatment‐naive patients with chronic hepatitis C virus (HCV) genotype (GT) 1, 4, or 6 infection. Hepatology 2015;62(Suppl S1):565A‐566A. CENTRAL
Zeuzem S, Ghalib R, Reddy K, Pockros PJ, Ben Ari ZIV, Zhao YUE, et al. Grazoprevir‐elbasvir combination therapy for treatment‐naive cirrhotic and noncirrhotic patients with chronic hepatitis C virus genotype 1, 4, or 6 infection: a randomized trial. Annals of Internal Medicine 2015;163(1):1‐13. CENTRAL
Zeuzem S, Ghalib R, Reddy KR, Pockros PJ, Ari ZB, Zhao Y, et al. The phase 3 C‐EDGE treatment‐naive (TN) study of a 12‐week oral regimen of grazoprevir (GZR, mk‐5172)/ elbasvir (EBR, mk‐8742) in patients with chronic HCV genotype (GT) 1, 4, or 6 infection. Journal of Hepatology 2015;62(Suppl S2):S213. CENTRAL

Chandra 2006a {published data only}

Chandra P, Raible D, Harper D, Speth J, Villano S, Bichier G. Antiviral activity of the non‐nucleoside polymerase inhibitor, HCV‐796, in patients with chronic hepatitis C virus: preliminary results from a randomized, double‐blind, placebo‐controlled, ascending multiple dose study ‐ first article. Gastroenterology 2006;130(4 (Suppl 2)):A748. CENTRAL

COMMAND‐1 2015a1 {published data only}

Hezode C, Hirschfield GM, Ghesquiere W, Sievert W, Rodriguez‐Torres M, Shafran S, et al. BMS‐790052, a NS5A replication complex inhibitor, combined with peginterferon alfa‐2a and ribavirin in treatment‐naive HCV‐genotype 1 or 4 patients: phase 2b AI444010 study interim week 12 results. Hepatology 2011;54(S1):474A‐5A. CENTRAL
Hezode C, Hirschfield GM, Ghesquiere W, Sievert W, Rodriguez‐Torres M, Shafran S, et al. Daclatasvir (BMS‐790052), a NS5A replication complex inhibitor, combined with peginterferon‐alfa‐2a and ribavirin in treatment‐naive HCV genotype 1 or 4 subjects: phase 2b AI444010 study interim week 24 results. Hepatology International 2012;6(1):167. CENTRAL
Hezode C, Hirschfield GM, Ghesquiere W, Sievert W, Rodriguez‐Torres M, Shafran SD, et al. Daclatasvir plus peginterferon alfa and ribavirin for treatment‐naive chronic hepatitis C genotype 1 or 4 infection: a randomised study. Gut 2015;64(6):948‐56. CENTRAL

COMMAND‐1 2015a2 {published data only}

Hezode C, Hirschfield GM, Ghesquiere W, Sievert W, Rodriguez‐Torres M, Shafran S, et al. BMS‐790052, a NS5A replication complex inhibitor, combined with peginterferon alfa‐2a and ribavirin in treatment‐naive HCV‐genotype 1 or 4 patients: phase 2b AI444010 study interim week 12 results. Hepatology 2011;54(S1):474A‐5A. CENTRAL
Hezode C, Hirschfield GM, Ghesquiere W, Sievert W, Rodriguez‐Torres M, Shafran S, et al. Daclatasvir (BMS‐790052), a NS5A replication complex inhibitor, combined with peginterferon‐alfa‐2a and ribavirin in treatment‐naive HCV genotype 1 or 4 subjects: phase 2b AI444010 study interim week 24 results. Hepatology International 2012;6(1):167. CENTRAL
Hezode C, Hirschfield GM, Ghesquiere W, Sievert W, Rodriguez‐Torres M, Shafran SD, et al. Daclatasvir plus peginterferon alfa and ribavirin for treatment‐naive chronic hepatitis C genotype 1 or 4 infection: a randomised study. Gut 2015;64(6):948‐56. CENTRAL

CONCERTO‐1 2015 {published data only}

Hayashi N, Izumi N, Kumada H, Okanoue T, Tsubouchi H, Yatsuhashi H, et al. Simeprevir with peginterferon/ribavirin for treatment‐naive hepatitis C genotype 1 patients in Japan: CONCERTO‐1, a phase III trial. Journal of Hepatology 2014;61(2):219‐27. CENTRAL

Cooper 2009 {published data only}

Cooper C, Lawitz EJ, Ghali P, Rodriguez‐Torres M, Anderson FH, Lee SS, et al. Antiviral activity of the non‐nucleoside polymerase inhibitor, VCH‐759, in chronic hepatitis C patients: results from a randomized, double‐blind, placebo‐controlled, ascending multiple dose study. Hepatology 2007;46(4 (Suppl 1)):864A. CENTRAL
Cooper C, Lawitz EJ, Ghali P, Rodriguez‐Torres M, Anderson FH, Lee SS, et al. Evaluation of VCH‐759 monotherapy in hepatitis C infection. Journal of Hepatology 2009;51(1):39‐46. CENTRAL

Dauphine 2015a1 {published data only}

Everson G, Cooper C, Hezode C, Shiffman ML, Yoshida E, Beltran‐Jaramillo T, et al. Dauphine: a randomized phase II study of danoprevir/ritonavir plus peginterferon alpha‐2a/ribavirin in HCV genotypes 1 or 4. Liver International 2015;35(1):108‐19. CENTRAL
Everson G, Cooper C, Hezode C, Shiffman ML, Yoshida E, Beltran‐Jaramillo T, et al. Rapid and sustained achievement of undetectable HCV RNA during treatment with ritonavir‐boosted danoprevir/peg‐ifna‐2a/RBV in HCV genotype 1 or 4 patients: dauphine week 12 interim analysis. Journal of Hepatology 2012;56(Suppl 2):S466. CENTRAL
Hezode C, Shiffman ML, Cooper C, Everson GT, Marcellin P, Rodriguez‐Torres M, et al. Ritonavir‐boosted danoprevir plus peg‐IFNalpha‐2a/ribavirin (P/R) demonstrates up to 100% SVR24 with 12 or 24 weeks of total treatment in treatment‐naive patients with HCV genotype 4 infection in the DAUPHINE study. Hepatology 2012;56(S1):557A. CENTRAL
Le Pogam S, Navarro M, Bu L, Voulgari A, Illnicka M, Yan JM, et al. Low rate of on‐treatment resistance to danoprevir boosted by ritonavir (DNVR) combined with Peg‐IFNa‐2a/ribavirin: 12 week interim analysis from Dauphine study. Journal of Hepatology 2012;56(Suppl 2):S472. CENTRAL

Dauphine 2015a2 {published data only}

Everson G, Cooper C, Hezode C, Shiffman ML, Yoshida E, Beltran‐Jaramillo T, et al. Dauphine: a randomized phase II study of danoprevir/ritonavir plus peginterferon alpha‐2a/ribavirin in HCV genotypes 1 or 4. Liver International 2015;35(1):108‐19. CENTRAL
Everson G, Cooper C, Hezode C, Shiffman ML, Yoshida E, Beltran‐Jaramillo T, et al. Rapid and sustained achievement of undetectable HCV RNA during treatment with ritonavir‐boosted danoprevir/peg‐ifna‐2a/RBV in HCV genotype 1 or 4 patients: dauphine week 12 interim analysis. Journal of Hepatology 2012;56(Suppl 2):S466. CENTRAL
Hezode C, Shiffman ML, Cooper C, Everson GT, Marcellin P, Rodriguez‐Torres M, et al. Ritonavir‐boosted danoprevir plus peg‐IFNalpha‐2a/ribavirin (P/R) demonstrates up to 100% SVR24 with 12 or 24 weeks of total treatment in treatment‐naive patients with HCV genotype 4 infection in the DAUPHINE study. Hepatology 2012;56(S1):557A. CENTRAL
Le Pogam S, Navarro M, Bu L, Voulgari A, Illnicka M, Yan JM, et al. Low rate of on‐treatment resistance to danoprevir boosted by ritonavir (DNVR) combined with peg‐IFNa‐2a/ribavirin: 12 week interim analysis from Dauphine study. Journal of Hepatology 2012;56(Suppl 2):S472. CENTRAL

Dauphine 2015a3 {published data only}

Everson G, Cooper C, Hezode C, Shiffman ML, Yoshida E, Beltran‐Jaramillo T, et al. Dauphine: a randomized phase II study of danoprevir/ritonavir plus peginterferon alpha‐2a/ribavirin in HCV genotypes 1 or 4. Liver International 2015;35(1):108‐19. CENTRAL
Everson G, Cooper C, Hezode C, Shiffman ML, Yoshida E, Beltran‐Jaramillo T, et al. Rapid and sustained achievement of undetectable HCV RNA during treatment with ritonavir‐boosted danoprevir/peg‐ifna‐2a/RBV in HCV genotype 1 or 4 patients: dauphine week 12 interim analysis. Journal of Hepatology 2012;56(Suppl 2):S466. CENTRAL
Hezode C, Shiffman ML, Cooper C, Everson GT, Marcellin P, Rodriguez‐Torres M, et al. Ritonavir‐boosted danoprevir plus peg‐IFNalpha‐2a/ribavirin (P/R) demonstrates up to 100% SVR24 with 12 or 24 weeks of total treatment in treatment‐naive patients with HCV genotype 4 infection in the DAUPHINE study. Hepatology 2012;56(S1):557A. CENTRAL
Le Pogam S, Navarro M, Bu L, Voulgari A, Illnicka M, Yan JM, et al. Low rate of on‐treatment resistance to danoprevir boosted by ritonavir (DNVR) combined with peg‐IFNa‐2a/ribavirin: 12 week interim analysis from Dauphine study. Journal of Hepatology 2012;56(Suppl 2):S472. CENTRAL

Dauphine 2015a4 {published data only}

Everson G, Cooper C, Hezode C, Shiffman ML, Yoshida E, Beltran‐Jaramillo T, et al. Dauphine: a randomized phase II study of danoprevir/ritonavir plus peginterferon alpha‐2a/ribavirin in HCV genotypes 1 or 4. Liver International 2015;35(1):108‐19. CENTRAL
Everson G, Cooper C, Hezode C, Shiffman ML, Yoshida E, Beltran‐Jaramillo T, et al. Rapid and sustained achievement of undetectable HCV RNA during treatment with ritonavir‐boosted danoprevir/peg‐ifna‐2a/RBV in HCV genotype 1 or 4 patients: dauphine week 12 interim analysis. Journal of Hepatology 2012;56(Suppl 2):S466. CENTRAL
Hezode C, Shiffman ML, Cooper C, Everson GT, Marcellin P, Rodriguez‐Torres M, et al. Ritonavir‐boosted danoprevir plus peg‐IFNalpha‐2a/ribavirin (P/R) demonstrates up to 100% SVR24 with 12 or 24 weeks of total treatment in treatment‐naive patients with HCV genotype 4 infection in the DAUPHINE study. Hepatology 2012;56(S1):557A. CENTRAL
Le Pogam S, Navarro M, Bu L, Voulgari A, Illnicka M, Yan JM, et al. Low rate of on‐treatment resistance to danoprevir boosted by ritonavir (DNVR) combined with Peg‐IFNa‐2a/ribavirin: 12 week interim analysis from Dauphine study. Journal of Hepatology 2012;56(Suppl 2):S472. CENTRAL

De Bruijne 2010a1 {published data only}

De Bruijne J, Bergmann JF, Reesink HW, Weegink CJ, Molenkamp R, Schinkel J, et al. Antiviral activity of narlaprevir combined with ritonavir and pegylated interferon in chronic hepatitis C patients. Hepatology 2010;52(5):1590‐9. CENTRAL
De Bruijne J, Bergmann JF, Weegink CJ, Molenkamp R, Schinkel J, Treitel MA, et al. Narlaprevir and peginterferon alfa‐2b for 2 weeks in chronic hepatitis C genotype 1 patients, followed by peginterferon alfa‐2b and ribavirin for 24/48 weeks: final results. Journal of Hepatology 2010;52:S290‐1. CENTRAL
Hotho DM, De Bruijne J, Spaan M, Treitel MA, Boonstra A, De Knegt RJ, et al. Sustained virologic response after therapy with the HCV protease inhibitor narlaprevir in combination with peginterferon and ribavirin is durable through long‐term follow‐up. Journal of Viral Hepatitis 2013;20(4):e78‐81. CENTRAL

De Bruijne 2010a2 {published data only}

De Bruijne J, Bergmann JF, Reesink HW, Weegink CJ, Molenkamp R, Schinkel J, et al. Antiviral activity of narlaprevir combined with ritonavir and pegylated interferon in chronic hepatitis C patients. Hepatology 2010;52(5):1590‐9. CENTRAL
De Bruijne J, Bergmann JF, Weegink CJ, Molenkamp R, Schinkel J, Treitel MA, et al. Narlaprevir and peginterferon alfa‐2b for 2 weeks in chronic hepatitis C genotype 1 patients, followed by peginterferon alfa‐2b and ribavirin for 24/48 weeks: final results. Journal of Hepatology 2010;52:S290‐1. CENTRAL
Hotho DM, De Bruijne J, Spaan M, Treitel MA, Boonstra A, De Knegt RJ, et al. Sustained virologic response after therapy with the HCV protease inhibitor narlaprevir in combination with peginterferon and ribavirin is durable through long‐term follow‐up. Journal of Viral Hepatitis 2013;20(4):e78‐81. CENTRAL

Detishin 2011 {published data only}

Detishin V, Haazen W, Hooijmaijers R, Ruppert M, Bol K, Robison H, et al. Final results of the pharmacokinetics, efficacy, and safety/tolerability of 400 and 600 mg once‐daily dosing of ACH‐1625 (HCV NS3 protease inhibitor) in HCV genotype 1. Journal of Hepatology 2011;54(Suppl S1):S186‐7. CENTRAL

Dore 2015a1 {published data only}

Dore GJ, Lawitz E, Hezode C, Shafran SD, Ramji A, Tatum HA, et al. Daclatasvir plus peginterferon and ribavirin is noninferior to peginterferon and ribavirin alone, and reduces the duration of treatment for HCV genotype 2 or 3 infection. Gastroenterology 2015;148(2):355‐66. CENTRAL

Dore 2015a2 {published data only}

Dore GJ, Lawitz E, Hezode C, Shafran SD, Ramji A, Tatum HA, et al. Daclatasvir plus peginterferon and ribavirin is noninferior to peginterferon and ribavirin alone, and reduces the duration of treatment for HCV genotype 2 or 3 infection. Gastroenterology 2015;148(2):355‐66. CENTRAL

DRAGON 2014a1 {published data only}

Hayashi N, Seto C, Kato M, Komada Y, Goto S. Once‐daily simeprevir (TMC435) with peginterferon/ribavirin for treatment‐naive hepatitis C genotype 1‐infected patients in Japan: the DRAGON study. Journal of Gastroenterology 2014;49(1):138‐47. CENTRAL

DRAGON 2014a2 {published data only}

Hayashi N, Seto C, Kato M, Komada Y, Goto S. Once‐daily simeprevir (TMC435) with peginterferon/ribavirin for treatment‐naive hepatitis C genotype 1‐infected patients in Japan: the DRAGON study. Journal of Gastroenterology 2014;49(1):138‐47. CENTRAL

DRAGON 2014a3 {published data only}

Hayashi N, Seto C, Kato M, Komada Y, Goto S. Once‐daily simeprevir (TMC435) with peginterferon/ribavirin for treatment‐naive hepatitis C genotype 1‐infected patients in Japan: the DRAGON study. Journal of Gastroenterology 2014;49(1):138‐47. CENTRAL

DRAGON 2014a4 {published data only}

Hayashi N, Seto C, Kato M, Komada Y, Goto S. Once‐daily simeprevir (TMC435) with peginterferon/ribavirin for treatment‐naive hepatitis C genotype 1‐infected patients in Japan: the DRAGON study. Journal of Gastroenterology 2014;49(1):138‐47. CENTRAL

Erhardt 2009 {published data only}

Erhardt A, Deterding K, Benhamou Y, Reiser M, Forns X, Pol S, et al. Safety, pharmacokinetics and antiviral effect of BILB 1941, a novel hepatitis C virus RNA polymerase inhibitor, after 5 days oral treatment. Antiviral Therapy 2009;14(1):23‐32. CENTRAL

Feld 2014 {published data only}

Feld JJ, Kowdley KV, Coakley E, Sigal S, Nelson D, Crawford D, et al. Sapphire I: Phase 3 placebo‐controlled study of interferon‐free, 12‐week regimen of ABT‐450/R/ABT‐267, ABT‐333, and ribavirin in 631 treatment‐naive adults with hepatitis Cvirus genotype 1. Journal of Hepatology 2014;60(1 Suppl 1):S25. CENTRAL
Feld JJ, Kowdley KV, Coakley E, Sigal S, Nelson DR, Crawford D, et al. Treatment of HCV with ABT‐450/r‐ombitasvir and dasabuvir with ribavirin. New England Journal of Medicine 2014;370(17):1594‐603. CENTRAL
Kowdley KV, Feld JJ, Coakley E, Sigal S, Nelson DR, Crawford D, et al. Sapphire I: Phase 3 placebo‐controlled study of interferon‐free, 12‐week regimen of ABT‐450/R/ABT‐267, ABT‐333, and ribavirin in 631 treatment‐naive adults with hepatitis C virus genotype 1. Gastroenterology 2014;146(5 Suppl 1):S912‐S3. CENTRAL

Feld 2015 {published data only}

Feld JJ, Jacobson IM, Hezode C, Asselah T, Ruane PJ, Gruener N, et al. Sofosbuvir and velpatasvir for HCV genotype 1, 2, 4, 5, and 6 infection. New England Journal of Medicine 2015;373(27):2599‐607. CENTRAL

FISSION 2013 {published data only}

Lawitz E, Mangia A, Wyles D, Rodriguez‐Torres M, Hassanein T, Gordon Stuart C, et al. Sofosbuvir for previously untreated chronic hepatitis C infection. New England Journal of Medicine 2013;368(20):1878‐87. CENTRAL
Younossi ZM, Stepanova M, Henry L, Gane E, Jacobson IM, Lawitz E, et al. Minimal impact of sofosbuvir and ribavirin on health related quality of life in chronic hepatitis C (CH‐C). Journal of Hepatology 2014;60(4):741‐7. [PUBMED: 24333184]CENTRAL

Flamm 2013 {published data only}

Flamm S, Lawitz E, Jacobson I, Rubin R, Bourliere M, Hezode C, et al. High sustained virologic response (SVR) among genotype 1 previous non‐responders and relapsers to peginterferon/ribavirin when re‐treated with boceprevir (BOC) plus peginterferon alfa‐2A/ribavirin. Journal of Hepatology 2011;54(Suppl 1):S541‐2. CENTRAL
Flamm SL, Lawitz E, Jacobson I, Bourliere M, Hezode C, Vierling JM, et al. Boceprevir with peginterferon alfa‐2a‐ribavirin is effective for previously treated chronic hepatitis C genotype 1 infection. Clinical Gastroenterology and Hepatology 2013;11(1):81‐7. CENTRAL
Howe JA, Ogert RA, Barnard RJ, Hazuda D, Pedicone L, Brass CA. Analysis of sustained viral response and boceprevir resistance following combination treatment with boceprevir plus peginterferon alfa‐2A/ribavirin in HCV genotype 1 prior relapsers and non‐responders. Hepatology 2011;54(S1):443A. CENTRAL

Forestier 2007 {published data only}

Forestier N, Reesink HW, Weegink CJ, McNair L, Kieffer TL, Chu HM, et al. Antiviral activity of telaprevir (VX‐950) and peginterferon alfa‐2a in patients with hepatitis C. Hepatology 2007;46(3):640‐8. CENTRAL
Forestier N, Sarrazin C, Friedrich‐Rust M, Weegink C, Jansen P, Reesink H, et al. Status of patients with chronic hepatitis C after a 14‐day pre‐treatment with Telaprevir (VX‐950) followed by an anti‐viral therapy with Peg‐interferon‐alpha‐2a (Peg‐IFN‐2a) and Ribavirin (RBV). Zeitschrift fur Gastroenterologie 2007;45(8):841. CENTRAL
Forestier N, Sarrazin C, Friedrich‐Rust M, Zeuzem S. Status of patients with chronic hepatitis C after a pretreatment period of 14 days with Telaprevir (VX‐950) und a following antiviral therapy with Peg‐interferon‐alfa‐2a (Peg‐IFN‐2s) and ribavirin (RBV). Zeitschrift fur Gastroenterologie 2008;46(1):145. CENTRAL
Forestier N, Weegink CJ, Purdy S, McNair L, Jansen PL, Zeuzem S, et al. Current status of subjects receiving peg‐interferon‐alfa‐2A (peg‐ifn) and ribavirin (RBV) after a 14‐day study of the hepatitis C protease inhibitor telaprevir (VX‐950), with peg‐ifn. Hepatology 2006;44(4 (Suppl 1)):614A‐5A. CENTRAL
Weegink CJ, Forestier N, Jansen PL, Zeuzem S, Reesink HW. Final results of patients receiving peg‐interferon‐alfa‐2a (Peg‐IFN) and ribavirin (RBV) after a 14‐day study of the hepatitis C protease inhibitor telaprevir (VX‐950), with Peg‐IFN. Hepatology 2007;46(4 (Suppl 1)):819A. CENTRAL

Forestier 2011a1 {published data only}

Forestier N, Larrey D, Guyader D, Marcellin P, Rouzier R, Patat A, et al. Treatment of chronic hepatitis C patients with the NS3/4A protease inhibitor danoprevir (ITMN‐191/RG7227) leads to robust reductions in viral RNA: a phase 1b multiple ascending dose study. Journal of Hepatology 2011;54(6):1130‐6. CENTRAL
Forestier N, Larrey DG, Guyader D, Marcellin P, Rouzier R, Patat AA, et al. Treatment of chronic hepatitis C virus (HCV) genotype 1 patients with the NS3/4A protease inhibitor ITMN‐191 leads to rapid reductions in plasma HCV RNA: results of a phase 1b multiple ascending dose (MAD) study. Hepatology 2008;48(4 (Suppl)):1132A. CENTRAL
Moucari R, Forestier N, Larrey D, Guyader D, Couzigou P, Benhamou Y, et al. Danoprevir, an HCV NS3/4A protease inhibitor, improves insulin sensitivity in patients with genotype 1 chronic hepatitis C. Gut 2010;59(12):1694‐8. CENTRAL

Forestier 2011a2 {published data only}

Forestier N, Larrey D, Guyader D, Marcellin P, Rouzier R, Patat A, et al. Treatment of chronic hepatitis C patients with the NS3/4A protease inhibitor danoprevir (ITMN‐191/RG7227) leads to robust reductions in viral RNA: a phase 1b multiple ascending dose study. Journal of Hepatology 2011;54(6):1130‐6. CENTRAL
Forestier N, Larrey DG, Guyader D, Marcellin P, Rouzier R, Patat AA, et al. Treatment of chronic hepatitis C virus (HCV) genotype 1 patients with the NS3/4A protease inhibitor ITMN‐191 leads to rapid reductions in plasma HCV RNA: results of a phase 1b multiple ascending dose (MAD) study. Hepatology 2008;48(4 (Suppl)):1132A. CENTRAL
Moucari R, Forestier N, Larrey D, Guyader D, Couzigou P, Benhamou Y, et al. Danoprevir, an HCV NS3/4A protease inhibitor, improves insulin sensitivity in patients with genotype 1 chronic hepatitis C. Gut 2010;59(12):1694‐8. CENTRAL

Forestier 2011b {published data only}

Forestier N, Larrey D, Marcellin P, Benhamou Y, Guyader D, Bradford W, et al. Antiviral activity and safety of ITMN‐191 in combination with peginterferon alfa‐2a and ribavirin in patients with chronic hepatitis C virus (HCV). Journal of Hepatology 2009;50(Suppl 1):S35. CENTRAL
Forestier N, Larrey D, Marcellin P, Guyader D, Patat A, Rouzier R, et al. Antiviral activity of danoprevir (ITMN‐191/RG7227) in combination with pegylated interferon alpha‐2a and ribavirin in patients with hepatitis C. Journal of Infectious Diseases 2011;204(4):601‐8. CENTRAL

Forns 2014 {published data only}

Forns X, Lawitz E, Zeuzem S, Gane E, Bronowicki JP, Andreone P, et al. Simeprevir with peginterferon and ribavirin leads to high rates of SVR in patients with HCV genotype 1 who relapsed after previous therapy: a phase 3 trial. Gastroenterology 2014;146(7):1669‐79. CENTRAL
Forns X, Lawitz E, Zeuzem S, Gane EJ, Bronowicki JP, Andreone P, et al. Simeprevir (TMC435) with peg‐interferon alpha‐2a/ribavirin for treatment of chronic HCV genotype 1 infection in patients who relapsed after previous interferon‐based therapy: efficacy and safety in patient sub‐populations in the PROMISE phase III trial. Hepatology 2013;58(4 SUPPL. 1):737A‐8A. CENTRAL
Gane EJ, Forns X, Lawitz E, Zeuzem S, Bronowicki JP, Andreone P, et al. Simeprevir (TMC435) with peg‐interferon alpha‐2a/ribavirin for treatment of chronic HCV genotype 1 infection in patients who relapsed after previous interferon‐based therapy: efficacy in patients with genotype 1b HCV in the PROMISE phase III trial. Hepatology International 2014;8(1 Suppl 1):S181. CENTRAL
Lawitz E, Forns X, Zeuzem S, Gane E, Bronowicki JP, Andreone P, et al. Simeprevir (TMC435) with peginterferon/ribavirin for treatment of chronic HCV genotype 1 infection in patients who relapsed after previous interferon‐based therapy: results from PROMISE, a phase III trial. Gastroenterology 2013;144(5 Suppl 1):S151. CENTRAL

Foster 2011a1 {published data only}

De Meyer S, Ghys A, Foster GR, Beumont M, Van Baelen B, Lin TI, et al. Analysis of genotype 2 and 3 hepatitis C virus variants in patients treated with telaprevir demonstrates a consistent resistance profile across genotypes. Journal of Viral Hepatitis 2013;20(6):395‐403. CENTRAL
De Meyer S, Ghys A, Foster GR, Beumont‐Mauviel M, Van Baelen B, Lin TI, et al. Analyses of genotype 2/3 HCV variants in patients treated with telaprevir in study C209 showed that the telaprevir resistance profile appears to be consistent across genotypes. Antiviral Therapy 2010;15(Suppl 2):A118. CENTRAL
Foster GR, Hezode C, Bronowicki JP, Carosi G, Weiland O, Verlinden L, et al. Activity of telaprevir alone or in combination with peginterferon alfa‐2a and ribavirin in treatment‐naive genotype 2 and 3 hepatitis‐C patients: final results of STUDY C209. Journal of Hepatology 2010;52(Suppl 1):S27. CENTRAL
Foster GR, Hézode C, Bronowicki JP, Carosi G, Weiland O, Verlinden L, et al. Telaprevir alone or with peginterferon and ribavirin reduces HCV RNA in patients with chronic genotype 2 but not genotype 3 infections. Gastroenterology 2011;141(3):881‐9. CENTRAL

Foster 2011a2 {published data only}

De Meyer S, Ghys A, Foster GR, Beumont M, Van Baelen B, Lin TI, et al. Analysis of genotype 2 and 3 hepatitis C virus variants in patients treated with telaprevir demonstrates a consistent resistance profile across genotypes. Journal of Viral Hepatitis 2013;20(6):395‐403. CENTRAL
De Meyer S, Ghys A, Foster GR, Beumont‐Mauviel M, Van Baelen B, Lin TI, et al. Analyses of genotype 2/3 HCV variants in patients treated with telaprevir in study C209 showed that the telaprevir resistance profile appears to be consistent across genotypes. Antiviral Therapy 2010;15(Suppl 2):A118. CENTRAL
Foster GR, Hezode C, Bronowicki JP, Carosi G, Weiland O, Verlinden L, et al. Activity of telaprevir alone or in combination with peginterferon alfa‐2a and ribavirin in treatment‐naive genotype 2 and 3 hepatitis‐C patients: final results of STUDY C209. Journal of Hepatology 2010;52(Suppl 1):S27. CENTRAL
Foster GR, Hézode C, Bronowicki JP, Carosi G, Weiland O, Verlinden L, et al. Telaprevir alone or with peginterferon and ribavirin reduces HCV RNA in patients with chronic genotype 2 but not genotype 3 infections. Gastroenterology 2011;141(3):881‐9. CENTRAL

Foster 2015a1 {published data only}

Foster GR, Afdhal N, Roberts SK, Brau N, Gane EJ, Pianko S, et al. Sofosbuvir and velpatasvir for HCV genotype 2 and 3 infection. New England Journal of Medicine 2015;373(27):2608‐17. CENTRAL

Foster 2015a2 {published data only}

Foster GR, Afdhal N, Roberts SK, Brau N, Gane EJ, Pianko S, et al. Sofosbuvir and velpatasvir for HCV genotype 2 and 3 infection. New England Journal of Medicine 2015;373(27):2608‐17. CENTRAL

Fried 2013 {published data only}

Aerssens J, Fanning G, Scholliers A, Lenz O, Peeters M, De Smedt G, et al. Impact of IL28B genotype and pretreatment serum IP‐10 in treatment‐naive genotype‐1 HCV patients treated with TMC435 in combination with peginterferona‐2A and ribavirin in PILLAR study. Journal of Hepatology 2011;54(Suppl S1):S5‐6. CENTRAL
Fried MW, Buti M, Dore GJ, Ferenci P, Jacobson I, Marcellin P, et al. Efficacy and safety of TMC435 in combination with peginterferon alfa‐2a and ribavirin in treatment‐naive genotype‐1 HCV patients: 24‐week interim results from the PILLAR study. Hepatology 2010;52(Suppl S1):403A. CENTRAL
Fried MW, Buti M, Dore GJ, Flisiak R, Ferenci P, Jacobson I, et al. Once‐daily simeprevir (TMC435) with pegylated interferon and ribavirin in treatment‐naïve genotype 1 hepatitis C: the randomized PILLAR study. Hepatology 2013;58(6):1918‐29. CENTRAL
Scott J, Gilles L, Fu M, Brohan E, Amatya R, Jessner W, et al. Patients with chronic hepatitis C virus treated with simeprevir added to peginterferon and ribavirin experienced less time with fatigue, depressive symptoms, and functional limitations: results from patients in the QUEST‐1, QUEST‐2, and promise studies. Value in Health 2013;16(7):A362. CENTRAL
Scott J, Gilles L, Fu M, Brohan E, Panter C, Arbuckle R, et al. Simeprevir added to peginterferon and ribavirin lessens time with fatigue, depressive symptoms and functional limitations in patients with chronic hepatitis C compared with peginterferon and ribavirin: results from 1161 patients in the QUEST‐1, QUEST‐2 and PROMISE studies. Journal of Viral Hepatitis 2015;22(8):639‐50. CENTRAL
Scott J, Rosa K, Fu M, Cerri K, Peeters M, Beumont‐Mauviel M, et al. Improved SVR with simeprevir (TMC435) associated with reduced time with patient‐reported fatigue in treatment‐naive, HCV‐infected patients in the PILLAR phase IIb trial. Journal of Hepatology 2013;58(Suppl 1):S371‐2. CENTRAL

Fundamental 2014a1 {published data only}

Buti M, Flisiak R, Kao JH, Chuang WL, Streinu‐Cercel A, Tabak F, et al. Alisporivir with peginterferon/ribavirin in patients with chronic hepatitis C genotype 1 infection who failed to respond to or relapsed after prior interferon‐based therapy: FUNDAMENTAL, a phase II trial. Journal of Viral Hepatitis 2014;22(7):596‐606. [DOI: 10.1111/jvh.12360]CENTRAL
Chuang WL, Kao JH, Sheen IS, Hsu SJ, Hung CH, Zekry A, et al. Superior SVR24 rates with alisporivir (ALV) plus peg‐interferon/ribavirin (P/R) in chronic HCV hepatitis (CHC) genotype 1 (G1) prior P/R treatment failures: final results of the fundamental study. Hepatology International 2014;8(1 Suppl 1):S206. CENTRAL

Fundamental 2014a2 {published data only}

Buti M, Flisiak R, Kao JH, Chuang WL, Streinu‐Cercel A, Tabak F, et al. Alisporivir with peginterferon/ribavirin in patients with chronic hepatitis C genotype 1 infection who failed to respond to or relapsed after prior interferon‐based therapy: FUNDAMENTAL, a phase II trial. Journal of Viral Hepatitis 2014;22(7):596‐606. [DOI: 10.1111/jvh.12360]CENTRAL
Chuang WL, Kao JH, Sheen IS, Hsu SJ, Hung CH, Zekry A, et al. Superior SVR24 rates with alisporivir (ALV) plus peg‐interferon/ribavirin (P/R) in chronic HCV hepatitis (CHC) genotype 1 (G1) prior P/R treatment failures: Final results of the fundamental study. Hepatology International 2014;8(1 Suppl 1):S206. CENTRAL

Fundamental 2014a3 {published data only}

Buti M, Flisiak R, Kao JH, Chuang WL, Streinu‐Cercel A, Tabak F, et al. Alisporivir with peginterferon/ribavirin in patients with chronic hepatitis C genotype 1 infection who failed to respond to or relapsed after prior interferon‐based therapy: FUNDAMENTAL, a phase II trial. Journal of Viral Hepatitis 2014;22(7):596‐606. [DOI: 10.1111/jvh.12360]CENTRAL
Chuang WL, Kao JH, Sheen IS, Hsu SJ, Hung CH, Zekry A, et al. Superior SVR24 rates with alisporivir (ALV) plus peg‐interferon/ribavirin (P/R) in chronic HCV hepatitis (CHC) genotype 1 (G1) prior P/R treatment failures: Final results of the fundamental study. Hepatology International 2014;8(1 Suppl 1):S206. CENTRAL

Gane 2008 {published data only}

Gane EJ, Rodriguez‐Torres M, Nelson DR, Jacobson IM, McHutchison JG, Jeffers L, et al. Antiviral activity of the HCV nucleoside polymerase inhibitor R7128 in HCV genotype 2 and 3 prior non‐responders: interim results of R7128 1500mg BID with PEG‐IFN and ribavirin for 28 days. Hepatology 2008;48(4 Suppl):1024A. CENTRAL
Lalezari J, Gane E, Rodriguez‐Torres M, DeJesus E, Nelson D, Everson G, et al. Potent antiviral activity of the HCV nucleoside polymerase inhibitor R7128 with peg‐IFN and ribavirin: interim results of R7128 500MG bid for 28 days. Journal of Hepatology 2008;48(Suppl 2):S29. CENTRAL

Gane 2010 {published data only}

Chu T, Kulkarni R, Gane EJ, Roberts SK, Stedman C, Angus P, et al. The effect of host IL28B genotype on early viral kinetics during interferon‐free treatment in patients with chronic hepatitis C (CHC). Journal of Hepatology 2011;54(Suppl 1):S521‐2. CENTRAL
Gane EJ, Roberts SK, Stedman C, Angus PW, Ritchie B, Elston R, et al. First‐in‐man demonstration of potent antiviral activity with a nucleoside polymerase (R7128) and protease (R7227/ITMN‐191) inhibitor combination in HCV: safety, pharmacokinetics, and virologic results from INFORM‐1. Journal of Hepatology 2009;50(Suppl 1):S380. CENTRAL
Gane EJ, Roberts SK, Stedman CA, Angus PW, Ritchie B, Elston R, et al. Combination therapy with a nucleoside polymerase (R7128) and protease (R7227/ITMN ‐ 191) inhibitor in HCV: safety, pharmacokinetics, and virologic results from INFORM‐1. Hepatology 2009;50(4 Suppl):394A‐5A. CENTRAL
Gane EJ, Roberts SK, Stedman CAM, Angus PW, Ritchie B, Elston ROB, et al. Oral combination therapy with a nucleoside polymerase inhibitor (RG7128) and danoprevir for chronic hepatitis C genotype 1 infection (INFORM‐1): a randomised, double‐blind, placebo‐controlled, dose‐escalation trial. Lancet 2010;376(9751):1467‐75. CENTRAL
Le Pogam S, Yan JM, Chhabra M, Ilnicka M, Kang H, Kosaka A, et al. Characterization of hepatitis C virus (HCV) quasispecies dynamics upon short‐term dual therapy with the HCV NS5B nucleoside polymerase inhibitor mericitabine and the NS3/4 protease inhibitor danoprevir. Antimicrobial Agents and Chemotherapy 2012;56(11):5494‐502. CENTRAL
Lim LY, Kwo PY. A glimpse at the future of hepatitis C therapy: The INFORM trial. Hepatology 2011;54(1):360‐3. CENTRAL
Morcos PN, Kulkarni R, Ipe D, Jumbe S, Tran J, Bradford WZ, et al. Pharmacokinetics/pharmacodynamics (PK/PD) of combination R7227 and R7128 therapy from INFORM‐1 demonstrates similar early HCV viral dynamics when R7227 is combined with either PEG ‐ IFN/ribavirin (SOC) or R7128. Hepatology 2009;50(4 Suppl):1041A‐2A. CENTRAL

Gane 2011 {published data only}

Gane E, Rouzier R, Stedman C, Wiercinska‐Drapalo A, Horban A, Chang L, et al. Ritonavir boosting of low dose RG7227/ITMN‐191, HCV NS3/4 a protease inhibitor, results in robust reduction in HCV RNA at lower exposures than provided by unboosted regimens. Journal of Hepatology 2010;52(Suppl 1):S16‐7. CENTRAL
Gane EJ, Rouzier R, Stedman C, Wiercinska‐Drapalo A, Horban A, Chang L, et al. Antiviral activity, safety, and pharmacokinetics of danoprevir/ritonavir plus PEG‐IFN alpha‐2a/RBV in hepatitis C patients. Journal of Hepatology 2011;55(5):972‐9. CENTRAL
Morcos PN, Chang L, Kulkarni R, Giraudon M, Shulman N, Brennan BJ, et al. A randomised study of the effect of danoprevir/ritonavir or ritonavir on substrates of cytochrome P450 (CYP) 3A and 2C9 in chronic hepatitis C patients using a drug cocktail. European Journal of Clinical Pharmacology 2013;69(11):1939‐49. CENTRAL

Gane 2015 {published data only}

Gane E, Schwabe C, Mader M, Suri V, Donohue M, Huang M, et al. Sustained virologic response after ACH‐3102 and sofosbuvir treatment for 8 or 6 weeks: A phase 2 "proxy" study. Journal of Hepatology 2015;62(Suppl S2):S266. CENTRAL

Gardner 2014a {published data only}

Gardner S, Cutrell AMY, Elko‐Simms C, Adkison K, Hamatake R, Walker J, et al. A double‐blind, randomized, placebo‐controlled study to assess the safety, antiviral activity and pharmacokinetics of GSK2336805 when given as monotherapy and in combination with peginterferon alfa‐2a and ribavirin in hepatitis C virus genotype 1‐infected treatment‐naive subjects. Liver International 2014;34(6):e89‐e95. CENTRAL

GlaxoSmithKline 2014 {published data only}

GlaxoSmithKline plc. A randomized, single blind, dose escalation, placebo‐controlled study to assess the safety, pharmacokinetics, and antiviral activity of repeat doses of GSK2878175 in subjects with chronic hepatitis C. www.gsk‐clinicalstudyregister.com/files2/116976‐Clinical‐Study‐Result‐Summary.pdf (accessed 25 January 2016). CENTRAL

Goldwater 2010 {published data only}

Goldwater R, DeMicco MP, Zong J, Chittick GE, Yuen GJ, West S, et al. Safety, pharmacokinetics, and antiviral activity of single oral doses of the HCV NS3 protease inhibitor GS 9256. Hepatology 2010;52(Suppl S1):717A. CENTRAL

HALLMARK‐DUAL 2014 {published data only}

Jacobson I, Kumada H, Chayama K, Dore G, Pol S, Zeuzem S, et al. Safety and tolerability of daclatasvir (DCV) in patients with chronic HCV infection. Hepatology International 2015;9(1 Suppl):S278‐9. CENTRAL
Manns M, Pol S, Jacobson IM, Marcellin P, Gordon SC, Peng CY, et al. All‐oral daclatasvir plus asunaprevir for hepatitis C virus genotype 1b: a multinational, phase 3, multi cohort study. Lancet 2014;384(9954):1597‐605. CENTRAL

Han 2014 {published data only}

Han KH, Helmond FA, Paik SW, Han SY, Heo J, Tak WY. Boceprevir plus peginterferon alfa and ribavirin for Korean patients with chronic hepatitis C virus genotype 1 infection and previous treatment failure. Hepatology International 2014;8(1 Suppl 1):S222‐3. CENTRAL

Hezode 2009 {published data only}

Bronowicki JP, Hezode C, Bengtsson L, Pol S, Bourliere M, Serfaty L, et al. 100% SVR in IL28B CC patients treated with 12 weeks of telaprevir, peginterferon and ribavirin in the PROVE2 trial. Journal of Hepatology 2012;56(Suppl 2):S430‐1. CENTRAL
Dusheiko GM, Hezode C, Pol S, Goeser T, Bronowicki JP, Bourliere M, et al. Treatment of chronic hepatitis C with telaprevir (TVR) in combination with peginterferon‐alfa‐2a with or without ribavirin: further interim analysis results of the PROVE2 study. Journal of Hepatology 2008;48(Suppl 2):S26. CENTRAL
Hezode C, Ferenci P, Dusheiko Geoffrey M, Tran A, Grange J‐D, Mathurin P, et al. Prove2 Study: treatment of chronic hepatitis C with telaprevir (Tvr) in combination with peginterferon‐alfa‐2a with or without ribavirin, interim analysis results. Gastroenterology 2008;134(4, Suppl. 1):A755. CENTRAL
Hezode C, Forestier N, Dusheiko G, Ferenci P, Pol S, Goeser T, et al. Telaprevir and peginterferon with or without ribavirin for chronic HCV infection. New England Journal of Medicine 2009;360(18):1839‐50. CENTRAL
Muir A, Poordad F, Sheikh A, Elkashab M, Brennan R, Ankoma‐Sey V, et al. SVR4 results for the combination of ACH‐3102 and sovaprevir, with ribavirin, in subjects with genotype 1 chronic hepatitis C infection. Hepatology International 2014;8(1 Suppl):s395. CENTRAL
Pawlotsky J, Kieffer T, Zhou Y, Zhang E, Marcial M, Byrn R, et al. Evaluation of viral variants during a phase 2 study (PROVE2) of telaprevir with peginterferon alfa‐2A and ribavirin in treatment‐naïve HCV genotype 1‐infected patients. Hepatology 2007;46(4 Suppl 1):862A. CENTRAL
Pawlotsky JM, Ferenci P, Dusheiko GM, Alves K, Kauffman R, Zeuzem S. PROVE2: a phase 2b study of telaprevir combined with peginterferon‐alfa‐2a with or without ribavirin for treatment‐naive patients with chronic hepatitis C. Hepatology International 2009;3(1):169. CENTRAL
Zeuzem S, Hezode C, Ferenci P, Dusheiko GM, Alves K, Bengtsson L, et al. Telaprevir in combination with peginterferon‐alfa‐2a with or without ribavirin in the treatment of chronic hepatitis C: final results of the PROVE2 study. Hepatology 2008;48(4 Suppl):418A‐9A. CENTRAL
Zeuzem S, Hezode C, Ferenci P, Dusheiko GM, Pol S, Goeser T, et al. PROVE2: phase II study of VX950 (telaprevir) in combination with peginterferon alfa2A with or without ribavirin in subjects with chronic hepatitis C; first interim analysis. Hepatology 2007;46(4 Suppl 1):268A‐9A. CENTRAL

Hinrichsen 2004 {published data only}

Hinrichsen H, Benhamou Y, Wedemeyer H, Reiser M, Sentjens RE, Calleja JL, et al. Short‐term antiviral efficacy of BILN 2061, a hepatitis C virus serine protease inhibitor, in hepatitis C genotype 1 patients. Gastroenterology 2004;127:1347–55. CENTRAL
Wedemeyer H, Erhardt A, Schmiegel W, Hinrichsen H, Chaves R, Yong CL, et al. Safety and antiviral effect of BILN 2061, a novel HCV serine protease inhibitor, after oral treatment over 2 days in patients with chronic hepatitis C, genotype 1, and liver cirrhosis. Hepatology 2003;38(4 Suppl 1):297A. CENTRAL

Hoeben 2015a1 {published data only}

Hoeben E, Viberg A, Petersson K, Lee M, Vanwelkenhuysen I, Witek J, et al. Simeprevir exposure in Asian treatment naive patients with chronic hepatitis C virus genotype 1 infection results from a population pharmacokinetic model in the phase III Tiger study. Hepatology International 2015;9(1 Suppl):S71. CENTRAL
Wei L, Han T, Yang D, Heo J, Shang J, Cheng J. Simeprevir plus peginterferon ribavirin in treatment naive patients with chronic hepatitis C virus genotype 1 infection results from the phase III Tiger study conducted in East Asian patients living in China and Korea. Hepatology International 2015;9(1 Suppl):S61. CENTRAL

Hoeben 2015a2 {published data only}

Hoeben E, Viberg A, Petersson K, Lee M, Vanwelkenhuysen I, Witek J, et al. Simeprevir exposure in Asian treatment naive patients with chronic hepatitis C virus genotype 1 infection results from a population pharmacokinetic model in the phase III Tiger study. Hepatology International 2015;9(1 Suppl):S71. CENTRAL
Wei L, Han T, Yang D, Heo J, Shang J, Cheng J. Simeprevir plus peginterferon ribavirin in treatment naive patients with chronic hepatitis C virus genotype 1 infection results from the phase III Tiger study conducted in East Asian patients living in China and Korea. Hepatology International 2015;9(1 Suppl):S61. CENTRAL

Hotho 2012 {published data only}

Hotho D, De Bruijne J, O'Farrell A, Boyea T, Li J, Weegink CJ, et al. Accelerated clinical trial design to assess the safety, tolerability and anti‐viral activity of PHX1766, a novel HCV NS3/4A protease inhibitor, in healthy volunteers and chronic hepatitis C patients. Hepatology 2009;50(4 Suppl):1031A‐2A. CENTRAL
Hotho Daphne M, De Bruijne J, O'Farrell A, Boyea T, Li J, Bracken M, et al. Pharmacokinetics and antiviral activity of PHX1766, a novel HCV protease inhibitor, using an accelerated Phase I study design. Antiviral Therapy 2012;17(2):365‐75. CENTRAL

Isakov 2016 {published data only}

Isakov V, Nikitin I, Chulanov V, Ogurtsov P, Lukyanova E, Long J, et al. Boceprevir plus peginterferon/ribavirin for treatment of chronic hepatitis C in Russia. World Journal of Hepatology 2016;8(6):331‐9. CENTRAL

Izumi 2014a1 {published data only}

Izumi N, Asahina Y, Yokosuka O, Imazeki F, Kawada N, Tamori A, et al. Combination therapy of treatment‐naive and nonresponder patients with HCV genotype 1 infection with BMS‐790052, an NS5A replication complex inhibitor, in combination with peginterferon alfa‐2a and ribavirin. Hepatology 2011;54(S1):1439A‐40A. CENTRAL
Izumi N, Yokosuka O, Kawada N, Osaki Y, Yamamoto K, Sata M, et al. Daclatasvir combined with peginterferon alfa‐2a and ribavirin in Japanese patients infected with hepatitis C genotype 1. Antiviral Therapy 2014;19(5):501‐10. CENTRAL
McPhee F, Hernandez D, Zhou N, Yu F, Ueland J, Monikowski A, et al. Virological escape in HCV genotype‐1‐infected patients receiving daclatasvir plus ribavirin and peginterferon alfa‐2a or alfa‐2b. Antiviral Therapy 2014;19(5):479‐90. CENTRAL
Suzuki F, Chayama K, Kawakami Y, Toyota J, Karino Y, Mochida S, et al. BMS‐790052, AN NS5A replication complex inhibitor, in combination with peginterferon alpha‐2B and ribavirin in Japanese treatment naive and nonresponder patients with chronic HCV genotype 1 infection. Hepatology 2011;54(S1):1441A. CENTRAL
Suzuki F, Chayama K, Kawakami Y, Toyota J, Karino Y, Mochida S, et al. Daclatasvir (BMS‐790052), an NS5A replication complex inhibitor, in combination with peginterferon alpha‐2b and ribavirin in Japanese treatment‐naive and nonresponder patients with chronic HCV genotype 1 infection. Hepatology International 2012;6(1):161‐2. CENTRAL
Suzuki F, Toyota J, Ikeda K, Chayama K, Mochida S, Hayashi N, et al. A randomized trial of daclatasvir with peginterferon alfa‐2b and ribavirin for HCV genotype 1 infection. Antiviral Therapy 2014;19(5):491‐9. CENTRAL

Izumi 2014a2 {published data only}

Izumi N, Asahina Y, Yokosuka O, Imazeki F, Kawada N, Tamori A, et al. Combination therapy of treatment‐naive and nonresponder patients with HCV genotype 1 infection with BMS‐790052, an NS5A replication complex inhibitor, in combination with peginterferon alfa‐2a and ribavirin. Hepatology 2011;54(S1):1439A‐40A. CENTRAL
Izumi N, Yokosuka O, Kawada N, Osaki Y, Yamamoto K, Sata M, et al. Daclatasvir combined with peginterferon alfa‐2a and ribavirin in Japanese patients infected with hepatitis C genotype 1. Antiviral Therapy 2014;19(5):501‐10. CENTRAL
McPhee F, Hernandez D, Zhou N, Yu F, Ueland J, Monikowski A, et al. Virological escape in HCV genotype‐1‐infected patients receiving daclatasvir plus ribavirin and peginterferon alfa‐2a or alfa‐2b. Antiviral Therapy 2014;19(5):479‐90. CENTRAL
Suzuki F, Chayama K, Kawakami Y, Toyota J, Karino Y, Mochida S, et al. BMS‐790052, AN NS5A replication complex inhibitor, in combination with peginterferon alpha‐2B and ribavirin in Japanese treatment naive and nonresponder patients with chronic HCV genotype 1 infection. Hepatology 2011;54(S1):1441A. CENTRAL
Suzuki F, Chayama K, Kawakami Y, Toyota J, Karino Y, Mochida S, et al. Daclatasvir (BMS‐790052), an NS5A replication complex inhibitor, in combination with peginterferon alpha‐2b and ribavirin in Japanese treatment‐naive and nonresponder patients with chronic HCV genotype 1 infection. Hepatology International 2012;6(1):161‐2. CENTRAL
Suzuki F, Toyota J, Ikeda K, Chayama K, Mochida S, Hayashi N, et al. A randomized trial of daclatasvir with peginterferon alfa‐2b and ribavirin for HCV genotype 1 infection. Antiviral Therapy 2014;19(5):491‐9. CENTRAL

Jacobson 2010 {published data only}

Jacobson I, Pockros P, Lalezari J, Lawitz E, Rodriguez‐Torres M, DeJesus E, et al. Antiviral activity of filibuvir in combination with pegylated interferon alfa‐2A and ribavirin for 28 days in treatment naive patients chronically infected with HCV genotype 1. Journal of Hepatology 2009;50(Suppl 1):S382. CENTRAL
Jacobson I, Pockros PJ, Lalezari J, Lawitz E, Rodriguez‐Torres M, DeJesus E, et al. Virologic response rates following 4 weeks of filibuvir in combination with pegylated interferon alfa‐2A and ribavirin in chronically‐infected HCV genotype‐1 patients. Journal of Hepatology 2010;52(Suppl 1):S464. CENTRAL
Mori J, Hammond JL, Srinivasan S, Jagannatha S, Ryst ECC. Genotypic characterisation of filibuvir (PF‐00868554 ) resistance in patients receiving four weeks co‐administration of filibuvir with pegIFN/RBV. Hepatology 2010;52(Suppl S1):722A. CENTRAL
Mori J, Hammond JL, Srinivasan S, Jagannatha S, Van der Ryst E. Genotypic characterisation of filibuvir (PF‐00868554) resistance in patients receiving four weeks co‐administration of filibuvir with PEGIFN/RBV (12 week analysis). Journal of Hepatology 2010;52(Suppl 1):S15. CENTRAL

Jacobson 2014 {published data only}

Jacobson I, Dore GJ, Foster GR, Fried MW, Radu M, Rafalskiy VV, et al. Simeprevir (TMC435) with peginterferon/ribavirin for chronic HCV genotype‐1 infection in treatment‐naive patients: results from QUEST‐1, a phase III trial. Journal of Hepatology 2013;58(Suppl 1):S574. CENTRAL
Jacobson IM, Dore GJ, Foster GR, Fried MW, Radu M, Rafalsky VV, et al. Simeprevir with pegylated interferon alfa 2a plus ribavirin in treatment‐naive patients with chronic hepatitis C virus genotype 1 infection (QUEST‐1): a phase 3, randomised, double‐blind, placebo‐controlled trial. Lancet 2014;384(9941):403‐13. CENTRAL

JUMP‐C 2013 {published data only}

Pockros PJ, Jensen D, Tsai N, Taylor R, Ramji A, Cooper C, et al. JUMP‐C: a randomized trial of mericitabine plus pegylated interferon alpha‐2a/ribavirin for 24 weeks in treatment‐naïve HCV genotype 1/4 patients. Hepatology 2013;58(2):514‐23. CENTRAL

Kwo 2010a1 {published data only}

Kwo P, Lawitz E, McCone J, Schiff E, Vierling J, Pound D, et al. HCV Sprint‐1 final results: SVR 24 from a phase 2 study of boceprevir plus pegintron (peginterferon ALFA‐2B)/ribavirin in treatment‐naive subjects with genotype‐1 chronic hepatitis C. Journal of Hepatology 2009;50(Suppl 1):S4. CENTRAL
Kwo PJ, Lawitz EJ, McCone J, Schiff ER, Vierling JM, Pound D, et al. Efficacy of boceprevir, an NS3 protease inhibitor, in combination with peginterferon alfa‐2b and ribavirin in treatment‐naive patients with genotype 1 hepatitis C infection (SPRINT‐1): an open‐label, randomised, multicentre phase 2 trial. Lancet 2010;376(9742):705‐16. CENTRAL
Ogert RA, Howe JA, Vierling JM, Kwo PY, Lawitz EJ, McCone J, et al. Resistance‐associated amino acid variants associated with boceprevir plus pegylated interferon‐alpha2b and ribavirin in patients with chronic hepatitis C in the SPRINT‐1 trial. Antiviral Therapy 2013;18(3):387‐97. CENTRAL
Poordad F, Vierling JM, Esteban R, Kwo PY, Long J, Chaudhri EI, et al. Hemogloblin decline during lead‐in phase as an early predictor of anemia after the addition of boceprevir: a retrospective analysis of HCV SPRINT‐1. Hepatology 2010;52(Suppl S1):770A. CENTRAL
Vierling JM, Kwo PY, Lawitz E, McCone J, Schiff ER, Pound D, et al. Frequencies of resistance‐associated amino acid variants following combination treatment with boceprevir plus PEGINTRON (peginterferon alfa‐2b)/ribavirin in patients with chronic hepatitis C (CHC), genotype 1 (G1). Hepatology 2010;52(Suppl S1):702A. CENTRAL

Kwo 2010a2 {published data only}

Kwo P, Lawitz E, McCone J, Schiff E, Vierling J, Pound D, et al. HCV sprint‐1 final results: SVR 24 from a phase 2 study of boceprevir plus pegintron (peginterferon ALFA‐2B)/ribavirin in treatment‐naive subjects with genotype‐1 chronic hepatitis C. Journal of Hepatology 2009;50(Suppl 1):S4. CENTRAL
Kwo PJ, Lawitz EJ, McCone J, Schiff ER, Vierling JM, Pound D, et al. Efficacy of boceprevir, an NS3 protease inhibitor, in combination with peginterferon alfa‐2b and ribavirin in treatment‐naive patients with genotype 1 hepatitis C infection (SPRINT‐1): an open‐label, randomised, multicentre phase 2 trial. Lancet 2010;376(9742):705‐16. CENTRAL
Ogert RA, Howe JA, Vierling JM, Kwo PY, Lawitz EJ, McCone J, et al. Resistance‐associated amino acid variants associated with boceprevir plus pegylated interferon‐alpha2b and ribavirin in patients with chronic hepatitis C in the SPRINT‐1 trial. Antiviral Therapy 2013;18(3):387‐97. CENTRAL
Poordad F, Vierling JM, Esteban R, Kwo PY, Long J, Chaudhri EI, et al. Hemogloblin decline during lead‐in phase as an early predictor of anemia after the addition of boceprevir: a retrospective analysis of HCV SPRINT‐1. Hepatology 2010;52(Suppl S1):770A. CENTRAL
Vierling JM, Kwo PY, Lawitz E, McCone J, Schiff ER, Pound D, et al. Frequencies of resistance‐associated amino acid variants following combination treatment with boceprevir plus PEGINTRON (peginterferon alfa‐2b)/ribavirin in patients with chronic hepatitis C (CHC), genotype 1 (G1). Hepatology 2010;52(Suppl S1):702A. CENTRAL

Kwo 2010a3 {published data only}

Kwo P, Lawitz E, McCone J, Schiff E, Vierling J, Pound D, et al. HCV Sprint‐1 final results: SVR 24 from a phase 2 study of boceprevir plus pegintron (peginterferon ALFA‐2B)/ribavirin in treatment‐naive subjects with genotype‐1 chronic hepatitis C. Journal of Hepatology 2009;50(Suppl 1):S4. CENTRAL
Kwo PJ, Lawitz EJ, McCone J, Schiff ER, Vierling JM, Pound D, et al. Efficacy of boceprevir, an NS3 protease inhibitor, in combination with peginterferon alfa‐2b and ribavirin in treatment‐naive patients with genotype 1 hepatitis C infection (SPRINT‐1): an open‐label, randomised, multicentre phase 2 trial. Lancet 2010;376(9742):705‐16. CENTRAL
Ogert RA, Howe JA, Vierling JM, Kwo PY, Lawitz EJ, McCone J, et al. Resistance‐associated amino acid variants associated with boceprevir plus pegylated interferon‐alpha2b and ribavirin in patients with chronic hepatitis C in the SPRINT‐1 trial. Antiviral Therapy 2013;18(3):387‐97. CENTRAL
Poordad F, Vierling JM, Esteban R, Kwo PY, Long J, Chaudhri EI, et al. Hemogloblin decline during lead‐in phase as an early predictor of anemia after the addition of boceprevir: a retrospective analysis of HCV SPRINT‐1. Hepatology 2010;52(Suppl S1):770A. CENTRAL
Vierling JM, Kwo PY, Lawitz E, McCone J, Schiff ER, Pound D, et al. Frequencies of resistance‐associated amino acid variants following combination treatment with boceprevir plus PEGINTRON (peginterferon alfa‐2b)/ribavirin in patients with chronic hepatitis C (CHC), genotype 1 (G1). Hepatology 2010;52(Suppl S1):702A. CENTRAL

Kwo 2010a4 {published data only}

Kwo P, Lawitz E, McCone J, Schiff E, Vierling J, Pound D, et al. HCV Sprint‐1 final results: SVR 24 from a phase 2 study of boceprevir plus pegintron (peginterferon ALFA‐2B)/ribavirin in treatment‐naive subjects with genotype‐1 chronic hepatitis C. Journal of Hepatology 2009;50(Suppl 1):S4. CENTRAL
Kwo PJ, Lawitz EJ, McCone J, Schiff ER, Vierling JM, Pound D, et al. Efficacy of boceprevir, an NS3 protease inhibitor, in combination with peginterferon alfa‐2b and ribavirin in treatment‐naive patients with genotype 1 hepatitis C infection (SPRINT‐1): an open‐label, randomised, multicentre phase 2 trial. Lancet 2010;376(9742):705‐16. CENTRAL
Ogert RA, Howe JA, Vierling JM, Kwo PY, Lawitz EJ, McCone J, et al. Resistance‐associated amino acid variants associated with boceprevir plus pegylated interferon‐alpha2b and ribavirin in patients with chronic hepatitis C in the SPRINT‐1 trial. Antiviral Therapy 2013;18(3):387‐97. CENTRAL
Poordad F, Vierling JM, Esteban R, Kwo PY, Long J, Chaudhri EI, et al. Hemogloblin decline during lead‐in phase as an early predictor of anemia after the addition of boceprevir: a retrospective analysis of HCV SPRINT‐1. Hepatology 2010;52(Suppl S1):770A. CENTRAL
Vierling JM, Kwo PY, Lawitz E, McCone J, Schiff ER, Pound D, et al. Frequencies of resistance‐associated amino acid variants following combination treatment with boceprevir plus PEGINTRON (peginterferon alfa‐2b)/ribavirin in patients with chronic hepatitis C (CHC), genotype 1 (G1). Hepatology 2010;52(Suppl S1):702A. CENTRAL

Lalezari 2011 {published data only}

Lalezari JP, Hazan L, Kankam M, Lawitz E, Poordad FF, Araya V, et al. High rapid virologic response (RVR) with ach‐1625 daily dosing plus pegifn‐alpha 2a/RBV in a 28‐day phase 2a trial. Hepatology 2011;54(Suppl S1):992A‐3A. CENTRAL

Lalezari 2012 {published data only}

Lalezari J, Asmuth D, Casiro A, Vargas H, Lawrence S, Dubuc‐Patrick G, et al. Short‐term monotherapy with IDX184, a liver‐targeted nucleotide polymerase inhibitor, in patients with chronic hepatitis C virus infection. Antimicrobial Agents and Chemotherapy 2012;56(12):6372‐8. CENTRAL

Lalezari 2013 {published data only}

Lalezari J, Box T, O'Riordan W, Mehra P, Nguyen T, Poordad F, et al. IDX184 in combination with pegylated interferon‐alpha2a and ribavirin for 2 weeks in treatment‐naive patients with chronic hepatitis C. Antiviral Therapy 2013;18(6):755‐64. CENTRAL
Lalezari J, Poordad F, Mehra P, Nguyen T, Dejesus E, Godofsky E, et al. Antiviral activity, pharmacokinetics and safety of IDX184 in combination with pegylated interferon (pegIFN) and ribavirin (RBV) in treatment‐naive HCV genotype 1‐infected subjects. Journal of Hepatology 2010;52(Suppl S1):S469. CENTRAL
Lalezari JP, O'Riordan W, Poordad F, Nguyen TT, Patrick GD, Chen J, et al. A phase IIA study of IDX184 in combination with pegylated interferon (PEGIFN) and ribavirin (RBV) in treatment‐naive HCV genotype 1‐infected subjects. Hepatology 2010;52(Suppl S1):337A. CENTRAL

Larrey 2012 {published data only}

Larrey D, Lohse A, De Ledinghen V, Trepo C, Gerlach T, Zarski JP, et al. 4 week therapy with the non‐nucleosidic polymerase inhibitor BI207127 in combination with peginterferon‐alfa2A and ribavirin in treatment naive and treatment experienced chronic HCV GT1 patients. Journal of Hepatology 2010;52(Suppl S1):S466. CENTRAL
Larrey D, Lohse AW, De Ledinghen V, Trepo C, Gerlach T, Zarski JP, et al. Rapid and strong antiviral activity of the non‐nucleosidic NS5B polymerase inhibitor BI 207127 in combination with peginterferon alfa 2a and ribavirin. Journal of Hepatology 2012;57(1):39‐46. CENTRAL

Larrey 2013 {published data only}

Lagace L, Cartier M, Laflamme G, Lawetz C, Marquis M Triki I, et al. Genotypic and phenotypic analysis of the NS5B polymerase region from viral isolates of HCV chronically infected patients treated with BI 207127 for 5‐days monotherapy. Hepatology 2010;52(Suppl S1):1205A‐6A. CENTRAL
Larrey D, Lohse AW, Trepo C, Bronowicki JP, Arasteh K, Bourliere M, et al. Antiviral effect, safety, and pharmacokinetics of five‐day oral administration of deleobuvir (BI 207127), an investigational hepatitis C virus RNA polymerase inhibitor, in patients with chronic hepatitis C. Antimicrobial Agents and Chemotherapy 2013;57(10):4727‐35. CENTRAL
Larrey DG, Benhamou Y, Lohse AW, Trepo C, Moelleken C, Bronowicki J, et al. BI 207127 is a potent HCV RNA polymerase inhibitor during 5 days monotherapy in patients with chronic hepatitis C. Hepatology 2009;50(4):1044A. CENTRAL

Lawitz 2008 {published data only}

Lawitz EJ, Sulkowski MS, Jacobson IM, Faruqui S, Kraft WK, Maliakkal B, et al. Safety, tolerability and antiviral activity of MK‐7009, a novel inhibitor of the hepatitis C virus NS3/4A protease, in patients with chronic HCV genotype 1 infection. Hepatology 2008;48(4 Suppl):403A‐4A. CENTRAL

Lawitz 2009 {published data only}

Lawitz E, Cooper C, Rodriguez‐Torres M, Ghalib R, Lalonde R, Sheikh A, et al. Safety, tolerability and antiviral activity of VCH‐916, a novel non‐nucleoside HCV polymerase inhibitor in patients with chronic HCV genotype‐1 infection. Journal of Hepatology 2009;50(Suppl S1):S37. CENTRAL

Lawitz 2010a {published data only}

Lawitz EJ, Marbury TC, Vince BD, Grunenberg N, Rodriguez‐Torres M, De Micco MP, et al. Dose‐ranging, three‐day monotherapy study of the HCV NS3 protease inhibitor GS‐9256. Journal of Hepatology 2010;52(Suppl 1):S466‐S7. CENTRAL

Lawitz 2010b {published data only}

Lawitz E, Lalezari JP, Rodriguez‐Torres M, Kowdley KV, Nelson D, DeJesus E, et al. Clinical synergy of an anti‐HCV nucleoside analog with SOC: viral kinetics of PSI‐7977 with SOC. Hepatology 2010;52(Suppl S1):1205A. CENTRAL

Lawitz 2010c {published data only}

Lawitz E, Rodriguez‐Torres M, Rustgi VK, Hassanein T, Rahimy MH, Crowley CA, et al. Safety and antiviral activity of ANA598 in combination with pegylated interferon alpha2A plus ribavirin in treatment‐naive genotype‐1 chronic HCV patients. Hepatology 2010;52(Suppl S1):334A‐5A. CENTRAL
Lawitz E, Rodriquez‐Torres M, Rustgi VK, Hassanein T, Rahimy MH, Crowley CA, et al. Safety and antiviral activity of ANA598 in combination with pegylated interferon alpha2a plus ribavirin in treatment‐naive genotype‐1 chronic HCV patients. Journal of Hepatology 2010;52(Suppl S1):S467. CENTRAL
Muir AJ, Lawitz E, Rodriguez‐Torres M, Rustgi VK, Hassanein T, Appleman JR, et al. IL28B polymorphism and kinetics of antiviral activity for ANA598 in combination with pegylated interferon α2A plus ribavirin in treatment‐naïve genotype‐1 chronic HCV patients. Hepatology 2010;52(Suppl S1):1200A. CENTRAL

Lawitz 2011a {published data only}

Lawitz E, Gaultier I, Poordad F, Cohen DE, Menon R. ABT‐450/ritonavir (ABT‐450/R) combined with pegylated interferon alpha‐2a and ribavirin (SOC) after 3‐day monotherapy in genotype 1 HCV‐infected treatment‐naive subjects: 12‐week interim efficacy and safety results. Journal of Hepatology 2011;54(Suppl 1):S482. CENTRAL
Lawitz E, Gaultier I, Poordad F, DeJesus E, Kowdley KV, Sepulveda G, et al. 4‐week virologic response and safety of ABT‐450 given with low‐dose ritonavir (ABT‐450/r) in combination with pegylated interferon alpha‐2a and ribavirin (SOC) after 3‐day monotherapy in genotype 1 (GT1) HCV infected treatment‐naive subjects. Hepatology 2010;52(Suppl S1):878A. CENTRAL

Lawitz 2011b {published data only}

Lawitz E, Jacobson I, Godofsky E, Foster GR, Flisiak R, Bennett M, et al. A phase 2b trial comparing 24 to 48 weeks treatment with tegobuvir (GS‐9190)/PEG/RBV to 48 weeks treatment with PEG/RBV for chronic genotype 1 HCV infection. Journal of Hepatology 2011;54(Suppl S1):S181. CENTRAL

Lawitz 2012a {published data only}

Lawitz EJ, Gruener D, Hill JM, Marbury T, Moorehead L, Mathias A, et al. A phase 1, randomized, placebo‐controlled, 3‐day, dose‐ranging study of GS‐5885, an NS5A inhibitor, in patients with genotype 1 hepatitis C. Journal of Hepatology 2012;57(1):24‐31. CENTRAL

Lawitz 2012b {published data only}

Lawitz E, Hill J, Marbury T, Hazan L, Gruener D, Webster L, et al. GS‐6620, a liver‐targeted nucleotide prodrug, exhibits antiviral activity and favorable safety profile over 5 days in treatment naive chronic HCV genotype 1 subjects. Journal of Hepatology 2012;56(Suppl S2):S470‐1. CENTRAL

Lawitz 2013a1 {published data only}

Lawitz E, Lalezari JP, Hassanein T, Kowdley KV, Poordad FF, Sheikh AM, et al. Sofosbuvir in combination with peginterferon alfa‐2a and ribavirin for non‐cirrhotic, treatment‐naive patients with genotypes 1, 2, and 3 hepatitis C infection: a randomised, double‐blind, phase 2 trial. Lancet Infectious Diseases 2013;13(5):401‐8. CENTRAL

Lawitz 2013a2 {published data only}

Lawitz E, Lalezari JP, Hassanein T, Kowdley KV, Poordad FF, Sheikh AM, et al. Sofosbuvir in combination with peginterferon alfa‐2a and ribavirin for non‐cirrhotic, treatment‐naive patients with genotypes 1, 2, and 3 hepatitis C infection: a randomised, double‐blind, phase 2 trial. Lancet Infectious Diseases 2013;13(5):401‐8. CENTRAL

Lawitz 2013b {published data only}

Lawitz E, Hill JM, Marbury TC, Rodriguez‐Torres M, DeMicco MP, Quesada J, et al. Three‐day, dose‐ranging study of the HCV NS3 protease inhibitor GS‐9451. Hepatology 2010;52(Suppl S1):714A. CENTRAL
Lawitz EJ, Hill JM, Marbury T, Demicco MP, Delaney W, Yang J, et al. A phase I, randomized, placebo‐controlled, 3‐day, ascending‐dose study of GS‐9451, an NS3/4a protease inhibitor, in genotype 1 hepatitis C patients. Antiviral Therapy 2013;18(3):311‐9. CENTRAL

Lawitz 2013c {published data only}

Barnard R, Hwang PMT, Bhanja S, Campbell H, Strizki J, Cheney C, et al. Resistance analysis of cirrhotic treatment‐experienced genotype 1 patients in a study of MK‐7009 in combination with pegylated interferon/ribavirin. Journal of Hepatology 2013;58(Suppl S1):S487. CENTRAL
Elbasha E, El Khoury A, Mobashery N. Projected long‐term impact of MK‐7009 (vaniprevir) for previously treated chronic HCV genotype 1 infection. Hepatology International 2013;7(1 Suppl):S354. CENTRAL
Gao W, Caro L, Anderson M, Hwang P, Zhou A, Su J, et al. Vaniprevir (MK‐7009) demonstrates higher exposures in treatment‐experienced genotype (GT) 1 cirrhotic than non‐cirrhotic HCV‐infected patients. Hepatology 2013;58(Suppl S1):739A. CENTRAL
Lawitz E, Rodriguez‐Torres M, Stoehr A, Gane EJ, Serfaty L, Bhanja S, et al. A phase 2B study of MK‐7009 (vaniprevir) in patients with genotype 1 HCV infection who have failed previous pegylated interferon and ribavirin treatment. Journal of Hepatology 2013;59(1):11‐7. CENTRAL
Mobashery N, Wong P, Zhang S, Bhanja S, Warner A, Shaw PM, et al. Impact of IL28B genotype on virologic response in prior treatment failure patients who received MK‐7009 in combination with peginterferon and ribavirin. Journal of Hepatology 2012;56(Suppl S2):S467. CENTRAL

Lawitz 2013d {published data only}

Lawitz E, Rodriguez‐Torres M, Denning JM, Albanis E, Cornpropst M, Berrey MM, et al. Pharmacokinetics, pharmacodynamics, and tolerability of GS‐9851, a nucleotide analog polymerase inhibitor, following multiple ascending doses in patients with chronic hepatitis C infection. Antimicrobial Agents and Chemotherapy 2013;57(3):1209‐17. CENTRAL

Lawitz 2013e {published data only}

Lawitz E, Sulkowski M, Jacobson IRA, Kraft Walter K, Maliakkal B, Al‐Ibrahim M, et al. Characterization of vaniprevir, a hepatitis C virus NS3/4A protease inhibitor, in patients with HCV genotype 1 infection: safety, antiviral activity, resistance, and pharmacokinetics. Antiviral Research 2013;99(3):214‐20. CENTRAL

Lawitz 2013f {published data only}

Lawitz E, Hill J, Vince B, Murillo A, Gruener D, Marbury T, et al. ACH‐2684 demonstrates potent viral suppression in genotype 1 hepatitis C patients with and without cirrhosis: safety, pharmacokinetic, and viral kinetic analysis. Journal of Hepatology 2013;58(Suppl S1):S347. CENTRAL

Lawitz 2014a {published data only}

Lawitz E, Poordad F, Hyland RH, Wang J, Pang PS, Symonds WT, et al. High rates of SVR in patients with genotype 1 HCV infection and cirrhosis after treatment with ledipasvir/sofosbuvir+ribavirin or ledipasvir/sofosbuvir+ GS‐9669 for 8 weeks. Hepatology 2014;59(Suppl S1):1143A. CENTRAL

Lawitz 2015 {published data only}

Lawitz E, Freilich B, Link J, German P, Mo H, Han L, et al. A phase 1, randomized, dose‐ranging study of GS‐5816, a once‐daily NS5A inhibitor, in patients with genotype 1‐4 hepatitis C virus. Journal of Viral Hepatitis 2015;22(12):1011‐9. [PUBMED: 26183611]CENTRAL
Lawitz E, Glass SJ, Gruener D, Freilich B. Hill JM, Link JO, et al. GS‐5816, a once‐daily NS5A inhibitor, demonstrates potent antiviral activity in patients with genotype 1, 2, 3, or 4 HCV infection in a 3‐day monotherapy study. Hepatology 2013;58(Suppl S1):731A. CENTRAL

Liu 2015a {published data only}

Liu R, Curry S, McMonagle P, Nachbar RB, Pak I, Jumes P, et al. Resistance analysis of genotype‐1 and ‐3 HCV‐infected patients receiving MK‐8742, a HCV NS5A inhibitor with potent antiviral activity. Hepatology 2013;58(Suppl S1):435A. CENTRAL
Liu R, Curry S, McMonagle P, Yeh WW, Ludmerer SW, Jumes PA, et al. Susceptibilities of genotype 1a, 1b, and 3 hepatitis C virus variants to the NS5A inhibitor elbasvir. Antimicrobial Agents and Chemotherapy 2015;59(11):6922‐9. CENTRAL
Yeh WW, Lipardi C, Jumes P, De Lepeleire IM, Van Den Bulk N, Caro L, et al. MK‐8742, a HCV NS5A inhibitor with a broad spectrum of HCV genotypic activity, demonstrates potent antiviral activity in genotype‐1 and ‐3 HCV‐infected patients. Hepatology 2013;58(Suppl S1):438A‐9A. CENTRAL

Mallalieu 2014 {published data only}

Lawitz E, Rodriguez‐Torres M, DeMicco M, Nguyen T, Godofsky E, Appleman J, et al. Antiviral activity of ANA598, a potent non‐nucleoside polymerase inhibitor, in chronic hepatitis C patients. Journal of Hepatology 2009;50(Suppl 1):S384. CENTRAL
Mallalieu NL, Rahimy MH, Crowley CA, Appleman JR, Smith PF, Freddo JL. Pharmacokinetics and pharmacodynamics of setrobuvir, an orally administered hepatitis C virus non‐nucleoside analogue inhibitor. Clinical Therapeutics 2014;36(12):2047‐63. CENTRAL

Manns 2011 {published data only}

Manns MP, Bourliere M, Benhamou Y, Pol S, Bonacini M, Berg T. Safety and antiviral activity of BI201335, a new HCV NS3 protease inhibitor, in treatment‐naive patients with chronic hepatitis C genotype‐1 infection given as monotherapy and in combination with peginterferon alfa 2a (P) and ribavirin (R). Hepatology 2008;48(4 Suppl):1023A. CENTRAL
Manns MP, Bourliere M, Benhamou Y, Pol S, Bonacini M, Trepo C, et al. Potency, safety, and pharmacokinetics of the NS3/4A protease inhibitor BI201335 in patients with chronic HCV genotype‐1 infection. Journal of Hepatology 2011;54(6):1114‐22. CENTRAL

Manns 2012a1 {published data only}

Lawitz E, Sulkowski M, Jacobson IRA, Kraft WK, Maliakkal B, Al‐Ibrahim M, et al. Characterization of vaniprevir, a hepatitis C virus NS3/4A protease inhibitor, in patients with HCV genotype 1 infection: safety, antiviral activity, resistance, and pharmacokinetics. Antiviral Research 2013;99(3):214‐20. CENTRAL
Manns MP, Gane E, Rodriguez‐Torres M, Stoehr A, Yeh C‐T, Marcellin P, et al. Vaniprevir with pegylated interferon alpha‐2a and ribavirin in treatment‐naive patients with chronic hepatitis C: a randomized phase II study. Hepatology 2012;56(3):884‐93. CENTRAL
Manns MP, Gane E, Rodriguez‐Torres M, Stoehr A, Yeh C‐T, Wiedmann R, et al. MK‐7009 significantly improves rapid viral response (RVR) in combination with pegylated interferon ALFA‐2A and ribavirin in patients with chronic hepatitis C (CHC) genotype 1 infection. Journal of Hepatology 2009;50(Suppl S1):S384. CENTRAL
Manns MP, Gane E, Rodriguez‐Torres M, Stoehr A, Yeh CT, Marcellin P, et al. Vaniprevir with pegylated interferon alpha‐2a and ribavirin in treatment‐naive patients with chronic hepatitis C: a randomized phase II study. Hepatology 2012;56(3):884‐93. CENTRAL
Manns MP, Gane EJ, Rodriguez‐Torres M, Stoehr AD, Yeh C, Marcellin P, et al. Early viral response (EVR) rates in treatment ‐ naive patients with chronic hepatitis C (CHC) genotype 1 infection treated with MK‐7009, a novel NS3/4a protease inhibitor, in combination with pegylated interferon alfa‐2a and ribavirin for 28 days. Hepatology 2009;50(4 Suppl):332A‐3A. CENTRAL
Manns MP, Gane EJ, Rodriguez‐Torres M, Stoehr AD, Yeh C, Marcellin P, et al. Sustained viral response (SVR) rates in genotype 1 treatment‐naive patients with chronic hepatitis C (CHC) infection treated with vaniprevir (MK‐7009), A NS3/4A protease inhibitor, in combination with pegylated interferon alfa‐2a and ribavirin for 28 days. Hepatology 2010;52(Suppl S1):361A. CENTRAL

Manns 2012a2 {published data only}

Lawitz E, Sulkowski M, Jacobson IRA, Kraft WK, Maliakkal B, Al‐Ibrahim M, et al. Characterization of vaniprevir, a hepatitis C virus NS3/4A protease inhibitor, in patients with HCV genotype 1 infection: safety, antiviral activity, resistance, and pharmacokinetics. Antiviral Research 2013;99(3):214‐20. CENTRAL
Manns MP, Gane E, Rodriguez‐Torres M, Stoehr A, Yeh C‐T, Marcellin P, et al. Vaniprevir with pegylated interferon alpha‐2a and ribavirin in treatment‐naive patients with chronic hepatitis C: a randomized phase II study. Hepatology 2012;56(3):884‐93. CENTRAL
Manns MP, Gane E, Rodriguez‐Torres M, Stoehr A, Yeh C‐T, Wiedmann R, et al. MK‐7009 significantly improves rapid viral response (RVR) in combination with pegylated interferon ALFA‐2A and ribavirin in patients with chronic hepatitis C (CHC) genotype 1 infection. Journal of Hepatology 2009;50(Suppl S1):S384. CENTRAL
Manns MP, Gane E, Rodriguez‐Torres M, Stoehr A, Yeh CT, Marcellin P, et al. Vaniprevir with pegylated interferon alpha‐2a and ribavirin in treatment‐naive patients with chronic hepatitis C: a randomized phase II study. Hepatology 2012;56(3):884‐93. CENTRAL
Manns MP, Gane EJ, Rodriguez‐Torres M, Stoehr AD, Yeh C, Marcellin P, et al. Early viral response (EVR) rates in treatment ‐ naive patients with chronic hepatitis C (CHC) genotype 1 infection treated with MK‐7009, a novel NS3/4a protease inhibitor, in combination with pegylated interferon alfa‐2a and ribavirin for 28 days. Hepatology 2009;50(4 Suppl):332A‐3A. CENTRAL
Manns MP, Gane EJ, Rodriguez‐Torres M, Stoehr AD, Yeh C, Marcellin P, et al. Sustained viral response (SVR) rates in genotype 1 treatment‐naive patients with chronic hepatitis C (CHC) infection treated with vaniprevir (MK‐7009), A NS3/4A protease inhibitor, in combination with pegylated interferon alfa‐2a and ribavirin for 28 days. Hepatology 2010;52(Suppl S1):361A. CENTRAL

Manns 2012a3 {published data only}

Lawitz E, Sulkowski M, Jacobson IRA, Kraft WK, Maliakkal B, Al‐Ibrahim M, et al. Characterization of vaniprevir, a hepatitis C virus NS3/4A protease inhibitor, in patients with HCV genotype 1 infection: safety, antiviral activity, resistance, and pharmacokinetics. Antiviral Research 2013;99(3):214‐20. CENTRAL
Manns MP, Gane E, Rodriguez‐Torres M, Stoehr A, Yeh C‐T, Marcellin P, et al. Vaniprevir with pegylated interferon alpha‐2a and ribavirin in treatment‐naive patients with chronic hepatitis C: a randomized phase II study. Hepatology 2012;56(3):884‐93. CENTRAL
Manns MP, Gane E, Rodriguez‐Torres M, Stoehr A, Yeh C‐T, Wiedmann R, et al. MK‐7009 significantly improves rapid viral response (RVR) in combination with pegylated interferon ALFA‐2A and ribavirin in patients with chronic hepatitis C (CHC) genotype 1 infection. Journal of Hepatology 2009;50(Suppl S1):S384. CENTRAL
Manns MP, Gane E, Rodriguez‐Torres M, Stoehr A, Yeh CT, Marcellin P, et al. Vaniprevir with pegylated interferon alpha‐2a and ribavirin in treatment‐naive patients with chronic hepatitis C: a randomized phase II study. Hepatology 2012;56(3):884‐93. CENTRAL
Manns MP, Gane EJ, Rodriguez‐Torres M, Stoehr AD, Yeh C, Marcellin P, et al. Early viral response (EVR) rates in treatment ‐ naive patients with chronic hepatitis C (CHC) genotype 1 infection treated with MK‐7009, a novel NS3/4a protease inhibitor, in combination with pegylated interferon alfa‐2a and ribavirin for 28 days. Hepatology 2009;50(4 Suppl):332A‐3A. CENTRAL
Manns MP, Gane EJ, Rodriguez‐Torres M, Stoehr AD, Yeh C, Marcellin P, et al. Sustained viral response (SVR) rates in genotype 1 treatment‐naive patients with chronic hepatitis C (CHC) infection treated with vaniprevir (MK‐7009), A NS3/4A protease inhibitor, in combination with pegylated interferon alfa‐2a and ribavirin for 28 days. Hepatology 2010;52(Suppl S1):361A. CENTRAL

Manns 2012a4 {published data only}

Lawitz E, Sulkowski M, Jacobson IRA, Kraft WK, Maliakkal B, Al‐Ibrahim M, et al. Characterization of vaniprevir, a hepatitis C virus NS3/4A protease inhibitor, in patients with HCV genotype 1 infection: safety, antiviral activity, resistance, and pharmacokinetics. Antiviral Research 2013;99(3):214‐20. CENTRAL
Manns MP, Gane E, Rodriguez‐Torres M, Stoehr A, Yeh C‐T, Marcellin P, et al. Vaniprevir with pegylated interferon alpha‐2a and ribavirin in treatment‐naive patients with chronic hepatitis C: a randomized phase II study. Hepatology 2012;56(3):884‐93. CENTRAL
Manns MP, Gane E, Rodriguez‐Torres M, Stoehr A, Yeh C‐T, Wiedmann R, et al. MK‐7009 significantly improves rapid viral response (RVR) in combination with pegylated interferon ALFA‐2A and ribavirin in patients with chronic hepatitis C (CHC) genotype 1 infection. Journal of Hepatology 2009;50(Suppl S1):S384. CENTRAL
Manns MP, Gane E, Rodriguez‐Torres M, Stoehr A, Yeh CT, Marcellin P, et al. Vaniprevir with pegylated interferon alpha‐2a and ribavirin in treatment‐naive patients with chronic hepatitis C: a randomized phase II study. Hepatology 2012;56(3):884‐93. CENTRAL
Manns MP, Gane EJ, Rodriguez‐Torres M, Stoehr AD, Yeh C, Marcellin P, et al. Early viral response (EVR) rates in treatment ‐ naive patients with chronic hepatitis C (CHC) genotype 1 infection treated with MK‐7009, a novel NS3/4a protease inhibitor, in combination with pegylated interferon alfa‐2a and ribavirin for 28 days. Hepatology 2009;50(4 Suppl):332A‐3A. CENTRAL
Manns MP, Gane EJ, Rodriguez‐Torres M, Stoehr AD, Yeh C, Marcellin P, et al. Sustained viral response (SVR) rates in genotype 1 treatment‐naive patients with chronic hepatitis C (CHC) infection treated with vaniprevir (MK‐7009), A NS3/4A protease inhibitor, in combination with pegylated interferon alfa‐2a and ribavirin for 28 days. Hepatology 2010;52(Suppl S1):361A. CENTRAL

Manns 2014a {published data only}

Manns M, Marcellin P, Poordad F, De Araujo ESA, Buti M, Horsmans Y, et al. Simeprevir with pegylated interferon alfa 2a or 2b plus ribavirin in treatment‐naive patients with chronic hepatitis C virus genotype 1 infection (QUEST‐2): a randomised, double‐blind, placebo‐controlled phase 3 trial. Lancet 2014;384(9941):414‐26. CENTRAL
Poordad F, Manns MP, Marcellin P, Araujo ESA, Buti M, Horsmans Y, et al. Simeprevir (TMC435) with peginterferon/ribavirin for treatment of chronic HCV genotype‐1 infection in treatment‐naive patients: results from QUEST‐2, a phase III trial. Gastroenterology 2013;144(5 Suppl 1):S151. CENTRAL

Marcellin 2013a {published data only}

Marcellin P, Popa S, Berliba A, Boyer N, Streinu‐Cercel A, Tong M, et al. ALS‐2200, a novel once‐daily nucleotide HCV polymerase inhibitor, demonstrates potent antiviral activity in treatment naive GT1 chronic hepatitis C patients. Hepatology International 2013;7(1 Suppl):S342. CENTRAL

Marcellin 2013b {published data only}

Marcellin P, Manns MP, Janczewska E, Muir AJ, Wu X, Trenkle JD, et al. 12 week response‐guided treatment with the NS5A inhibitor, GS‐5885, the NS3 protease inhibitor, GS‐9451, plus pegylated interferon/ribavirin in treatment naive genotype 1 hepatitis C infected patients. Journal of Hepatology 2013;58(Suppl S1):S355. CENTRAL

MATTERHORN 2015a1 {published data only}

Feld JJ, Jacobson I, Jensen DM, Foster GR, Pol S, Tam E, et al. Randomized study of danoprevir/ritonavir‐based therapy for HCV genotype 1 patients with prior partial or null responses to peginterferon/ribavirin. Journal of Hepatology 2015;62(2):294‐302. CENTRAL
Song ZZ. Randomized study of danoprevir/ritonavir‐based therapy for HCV genotype 1 patients with prior partial or null responses to peginterferon/ribavirin. Journal of Hepatology 2015;63(3):769‐70. CENTRAL

MATTERHORN 2015a2 {published data only}

Feld JJ, Jacobson I, Jensen DM, Foster GR, Pol S, Tam E, et al. Randomized study of danoprevir/ritonavir‐based therapy for HCV genotype 1 patients with prior partial or null responses to peginterferon/ribavirin. Journal of Hepatology 2015;62(2):294‐302. CENTRAL
Song ZZ. Randomized study of danoprevir/ritonavir‐based therapy for HCV genotype 1 patients with prior partial or null responses to peginterferon/ribavirin. Journal of Hepatology 2015;63(3):769‐70. CENTRAL

McHutchison 2009 {published data only}

Everson GT, Gordon SC, Jacobson I, Kauffman RS, McNair L, Muir A, et al. PROVE 1: subgroup analysis of a phase 2 study of telaprevir with peginterferon alfa‐2a and ribavirin in treatment‐naive subjects with hepatitis C. Hepatology International 2009;3:167. CENTRAL
McHutchison JG, Everson GT, Gordon SC, Jacobson I, Kauffman R, McNair L, et al. PROVE1: results from a phase 2 study of telaprevir with peginterferon alfa‐2a and ribavirin in treatment‐naive subjects with hepatitis C. Journal of Hepatology 2008;48(Suppl S2):S4. CENTRAL
McHutchison JG, Everson GT, Gordon SC, Jacobson IM, Sulkowski M, Kauffman R, et al. Telaprevir with peginterferon and ribavirin for chronic HCV genotype 1 infection. New England Journal of Medicine 2009;360(18):1827‐38. CENTRAL

McHutchison 2010 {published data only}

Everson GT, Gordon SC, Jacobson I, Kauffman RS, McNair L, Muir A, et al. PROVE 1: subgroup analysis of a phase 2 study of telaprevir with peginterferon alfa‐2a and ribavirin in treatment‐naive subjects with hepatitis C. Hepatology International 2009;3:167. CENTRAL
Manns M, Muir A, Adda N, Jacobson I, Afdhal N, Heathcote J, et al. Telaprevir in hepatitis C genotype‐1‐infected patients with prior non‐response, viral breakthrough or relapse to peginterferonalfa‐2A/B and ribavirin therapy: SVR results of the PROVE3 study. Journal of Hepatology 2009;50(Suppl 1):S379. CENTRAL
McHutchinson JG, Manns MP, Muir AJ, Terrault NA, Jacobson IM, Afdhal NH, et al. Telaprevir for previously treated chronic HCV infection. New England Journal of Medicine 2010;362(14):1292‐303. CENTRAL
McHutchinson JG, Shiffman ML, Terrault N, Manns MP, Di Biscegli AM, Jacobson IM, et al. Interim results of PROVE 3: a randomized, controlled, phase 2b study of telaprevir (TVR) with peginterferon‐alfa‐2a (P) and/or ribavirin (R) in subjects with hepatitis C genotype 1 who failed to achieve an SVR with a prior course of PR therapy. Hepatology International 2009;3(1):25. CENTRAL
McHutchison JG, Manns MP, Muir A, Terrault N, Jacobson IM, Afdhal NH, et al. PROVE3 final results and 1‐year durability of SVR with telaprevir‐based regimen in hepatitis C genotype 1‐infected patients with prior non‐response, viral breakthrough or relapse to peginterferon‐alfa‐2a/b and ribavirin therapy. Hepatology 2009;50(4 Suppl):334A‐5A. CENTRAL
McHutchison JG, Shiffman ML, Terrault N, Manns MP, Di Bisceglie AM, Jacobson IM, et al. A phase 2b study of telaprevir with peginterferon‐alfa‐2a and ribavirin in hepatitis C genotype 1 null and partial responders and relapsers following a prior course of peginterferon‐alfa‐2a/b and ribavirin therapy: PROVE3 interim results. Hepatology 2008;48(4 Suppl):269A‐70A. CENTRAL

Mostafa 2015 {published data only}

Mostafa I, Hassan M, Ibrahim AM, Khalafalla O, Essawy F, Abdelazem A, et al. Pilot study to determine the efficacy and safety of combining boceprevir with peginterferon alfa 2B and ribavirin in treatment naive patients with genotype 4 chronic hepatitis C infection. Hepatology International 2015;9(1 Suppl):S259. CENTRAL

Muir 2014 {published data only}

Muir A, Poordad F, Sheikh A, Elkashab M, Brennan R, Ankoma‐Sey V, et al. SVR4 results for the combination of ACH‐3102 and Sovaprevir, with ribavirin, in patients with genotype 1 chronic hepatitis C infection. 23rd Annual Conference of APASL, Brisbane, Australia. 2014. CENTRAL

Nelson 2011 {published data only}

Lawitz E, Lalezari JP, Hassanein T, Kowdley KV. Once daily PSI‐7977 plus PEG/RBV in treatment naive patients with HCV GT1: robust end of treatment response rates are sustained posttreatment. Hepatology 2011;54(Suppl S1):472A. CENTRAL
Nelson DR, Lalezari J, Lawitz E, Hassanein T, Kowdley K, Poordad F, et al. Once daily PSI‐7977 plus PEG‐IFN/RBV in HCV GT1: 98% rapid virologic response, complete early virologic response: the proton study. Journal of Hepatology 2011;54(Suppl 1):S544. CENTRAL

Nelson 2012a1 {published data only}

Nelson DR, Zeuzem S, Andreone P, Ferenci P, Herring R, Jensen DM, et al. Balapiravir plus peginterferon alfa‐2a (40KD)/ribavirin in a randomized trial of hepatitis C genotype 1 patients. Annals of Hepatology 2012;11(1):15‐31. CENTRAL

Nelson 2012a2 {published data only}

Nelson DR, Zeuzem S, Andreone P, Ferenci P, Herring R, Jensen DM, et al. Balapiravir plus peginterferon alfa‐2a (40KD)/ribavirin in a randomized trial of hepatitis C genotype 1 patients. Annals of Hepatology 2012;11(1):15‐31. CENTRAL

Nelson 2012a3 {published data only}

Nelson DR, Zeuzem S, Andreone P, Ferenci P, Herring R, Jensen DM, et al. Balapiravir plus peginterferon alfa‐2a (40KD)/ribavirin in a randomized trial of hepatitis C genotype 1 patients. Annals of Hepatology 2012;11(1):15‐31. CENTRAL

Nelson 2012a4 {published data only}

Nelson DR, Zeuzem S, Andreone P, Ferenci P, Herring R, Jensen DM, et al. Balapiravir plus peginterferon alfa‐2a (40KD)/ribavirin in a randomized trial of hepatitis C genotype 1 patients. Annals of Hepatology 2012;11(1):15‐31. CENTRAL

Nelson 2012a5 {published data only}

Nelson DR, Zeuzem S, Andreone P, Ferenci P, Herring R, Jensen DM, et al. Balapiravir plus peginterferon alfa‐2a (40KD)/ribavirin in a randomized trial of hepatitis C genotype 1 patients. Annals of Hepatology 2012;11(1):15‐31. CENTRAL

Nelson 2012a6 {published data only}

Nelson DR, Zeuzem S, Andreone P, Ferenci P, Herring R, Jensen DM, et al. Balapiravir plus peginterferon alfa‐2a (40KD)/ribavirin in a randomized trial of hepatitis C genotype 1 patients. Annals of Hepatology 2012;11(1):15‐31. CENTRAL

Nelson 2012b {published data only}

Nelson DR, Lawitz E, Bain V, Gitlin N, Hawkins T, Marotta P, et al. High SVR12 with 16 weeks of tegobuvir and GS‐9256 with peginterferon‐alfa 2A and ribavirin in treatment‐naïve genotype 1 HCV patients. Journal of Hepatology 2012;56(Suppl 2):S6‐7. CENTRAL

Nettles 2010 {published data only}

Nettles RE, Chien C, Chung E, Persson A, Gao M, Belema M, et al. MS‐790052 is a first‐in class potent Hepatitis C Virus (HCV) NS5A inhibitor for patients with chronic HCV infection: results from a proof‐of‐concept study. Hepatology International 2010;4(1):184. CENTRAL

Nettles 2011a1 {published data only}

Nettles R, Sevinsky H, Chung E, Burt D, Xiao H, Marbury TC, et al. BMS‐790052, a first‐in class potent hepatitis C virus NS5A inhibitor, demonstrates multiple dose proof‐of‐concept in subjects with chronic GT1 HCV infection. Hepatology 2010;52(Suppl S1):1214A‐5A. CENTRAL
Nettles RE, Gao M, Bifano M, Chung E, Persson A, Marbury TC, et al. Multiple ascending dose study of BMS‐790052, a nonstructural protein 5A replication complex inhibitor, in patients infected with hepatitis C virus genotype 1. Hepatology 2011;54(6):1956‐65. CENTRAL

Nettles 2011a2 {published data only}

Nettles R, Sevinsky H, Chung E, Burt D, Xiao H, Marbury TC, et al. BMS‐790052, a first‐in class potent hepatitis C virus NS5A inhibitor, demonstrates multiple dose proof‐of‐concept in subjects with chronic GT1 HCV infection. Hepatology 2010;52(Suppl S1):1214A‐5A. CENTRAL
Nettles RE, Gao M, Bifano M, Chung E, Persson A, Marbury TC, et al. Multiple ascending dose study of BMS‐790052, a nonstructural protein 5A replication complex inhibitor, in patients infected with hepatitis C virus genotype 1. Hepatology 2011;54(6):1956‐65. CENTRAL

Nettles 2011a3 {published data only}

Nettles R, Sevinsky H, Chung E, Burt D, Xiao H, Marbury TC, et al. BMS‐790052, a first‐in class potent hepatitis C virus NS5A inhibitor, demonstrates multiple dose proof‐of‐concept in subjects with chronic GT1 HCV infection. Hepatology 2010;52(Suppl S1):1214A‐5A. CENTRAL
Nettles RE, Gao M, Bifano M, Chung E, Persson A, Marbury TC, et al. Multiple ascending dose study of BMS‐790052, a nonstructural protein 5A replication complex inhibitor, in patients infected with hepatitis C virus genotype 1. Hepatology 2011;54(6):1956‐65. CENTRAL

Nettles 2011a4 {published data only}

Nettles R, Sevinsky H, Chung E, Burt D, Xiao H, Marbury TC, et al. BMS‐790052, a first‐in class potent hepatitis C virus NS5A inhibitor, demonstrates multiple dose proof‐of‐concept in subjects with chronic GT1 HCV infection. Hepatology 2010;52(Suppl S1):1214A‐5A. CENTRAL
Nettles RE, Gao M, Bifano M, Chung E, Persson A, Marbury TC, et al. Multiple ascending dose study of BMS‐790052, a nonstructural protein 5A replication complex inhibitor, in patients infected with hepatitis C virus genotype 1. Hepatology 2011;54(6):1956‐65. CENTRAL

Nettles 2011a5 {published data only}

Nettles R, Sevinsky H, Chung E, Burt D, Xiao H, Marbury TC, et al. BMS‐790052, a first‐in class potent hepatitis C virus NS5A inhibitor, demonstrates multiple dose proof‐of‐concept in subjects with chronic GT1 HCV infection. Hepatology 2010;52(Suppl S1):1214A‐5A. CENTRAL
Nettles RE, Gao M, Bifano M, Chung E, Persson A, Marbury TC, et al. Multiple ascending dose study of BMS‐790052, a nonstructural protein 5A replication complex inhibitor, in patients infected with hepatitis C virus genotype 1. Hepatology 2011;54(6):1956‐65. CENTRAL

Nettles 2011a6 {published data only}

Nettles R, Sevinsky H, Chung E, Burt D, Xiao H, Marbury TC, et al. BMS‐790052, a first‐in class potent hepatitis C virus NS5A inhibitor, demonstrates multiple dose proof‐of‐concept in subjects with chronic GT1 HCV infection. Hepatology 2010;52(Suppl S1):1214A‐5A. CENTRAL
Nettles RE, Gao M, Bifano M, Chung E, Persson A, Marbury TC, et al. Multiple ascending dose study of BMS‐790052, a nonstructural protein 5A replication complex inhibitor, in patients infected with hepatitis C virus genotype 1. Hepatology 2011;54(6):1956‐65. CENTRAL

Nishiguchi 2014a1 {published data only}

Nishiguchi S, Kuboki M, Sakai Y, Aizawa N, Iwata K, Ikeda N, et al. All treated Japanese genotype 1 patients with IL28B TT allele achieved SVR by 4 weeks faldaprevir and 48 weeks PegIFN/ ribavirin. Hepatology International 2013;7(1 Suppl):S393. CENTRAL
Nishiguchi S, Kuboki M, Sakai Y, Aizawa N, Takashima T, Iwata K. 100% SVR in Japanese genotype 1 patients with IL28B TT allele treated with 4 weeks faldaprevir and 48 weeks PegIFN/ribavirin. Hepatology International 2013;7(1 Suppl):S372‐3. CENTRAL
Nishiguchi S, Sakai Y, Kuboki M, Tsunematsu S, Urano Y, Sakamoto W, et al. Safety and efficacy of faldaprevir with pegylated interferon alfa‐2a and ribavirin in Japanese patients with chronic genotype‐1 hepatitis C infection. Liver International 2014;34(1):78‐88. CENTRAL

Nishiguchi 2014a2 {published data only}

Nishiguchi S, Kuboki M, Sakai Y, Aizawa N, Iwata K, Ikeda N, et al. All treated Japanese genotype 1 patients with IL28B TT allele achieved SVR by 4 weeks faldaprevir and 48 weeks PegIFN/ ribavirin. Hepatology International 2013;7(1 Suppl):S393. CENTRAL
Nishiguchi S, Kuboki M, Sakai Y, Aizawa N, Takashima T, Iwata K. 100% SVR in Japanese genotype 1 patients with IL28B TT allele treated with 4 weeks faldaprevir and 48 weeks PegIFN/ribavirin. Hepatology International 2013;7(1 Suppl):S372‐3. CENTRAL
Nishiguchi S, Sakai Y, Kuboki M, Tsunematsu S, Urano Y, Sakamoto W, et al. Safety and efficacy of faldaprevir with pegylated interferon alfa‐2a and ribavirin in Japanese patients with chronic genotype‐1 hepatitis C infection. Liver International 2014;34(1):78‐88. CENTRAL

OPERA 2011a1 {published data only}

Manns M, Reesink H, Berg T, Dusheiko G, Flisiak R, Marcellin P, et al. Rapid viral response of once‐daily TMC435 plus pegylated interferon/ribavirin in hepatitis C genotype‐1 patients: a randomized trial. Antiviral Therapy 2011;16(7):1021‐33. CENTRAL

OPERA 2011a2 {published data only}

Manns M, Reesink H, Berg T, Dusheiko G, Flisiak R, Marcellin P, et al. Rapid viral response of once‐daily TMC435 plus pegylated interferon/ribavirin in hepatitis C genotype‐1 patients: a randomized trial. Antiviral Therapy 2011;16(7):1021‐33. CENTRAL

OPERA 2011a3 {published data only}

Manns M, Reesink H, Berg T, Dusheiko G, Flisiak R, Marcellin P, et al. Rapid viral response of once‐daily TMC435 plus pegylated interferon/ribavirin in hepatitis C genotype‐1 patients: a randomized trial. Antiviral Therapy 2011;16(7):1021‐33. CENTRAL

OPERA 2011a4 {published data only}

Manns M, Reesink H, Berg T, Dusheiko G, Flisiak R, Marcellin P, et al. Rapid viral response of once‐daily TMC435 plus pegylated interferon/ribavirin in hepatitis C genotype‐1 patients: a randomized trial. Antiviral Therapy 2011;16(7):1021‐33. CENTRAL

OPERA 2011a5 {published data only}

Manns M, Reesink H, Berg T, Dusheiko G, Flisiak R, Marcellin P, et al. Rapid viral response of once‐daily TMC435 plus pegylated interferon/ribavirin in hepatitis C genotype‐1 patients: a randomized trial. Antiviral Therapy 2011;16(7):1021‐33. CENTRAL

OPERA 2011a6 {published data only}

Manns M, Reesink H, Berg T, Dusheiko G, Flisiak R, Marcellin P, et al. Rapid viral response of once‐daily TMC435 plus pegylated interferon/ribavirin in hepatitis C genotype‐1 patients: a randomized trial. Antiviral Therapy 2011;16(7):1021‐33. CENTRAL

Pasquinelli 2012a1 {published data only}

Pasquinelli C, Eley T, Villegas C, Sandy K, Mathias E, Wendelburg P, et al. Safety, tolerability, pharmacokinetics and antiviral activity following single and multiple‐dose administration of BMS‐650032, a novel HCV NS3 inhibitor, in subjects with chronic genotype 1 HCV infection. Hepatology International 2010;4(1):184. CENTRAL
Pasquinelli C, Eley T, Villegas C, Sandy K, Mathias E, Wendelburg P, et al. Safety, tolerability, pharmacokinetics and antiviral activity following single‐and multiple‐dose administration of BMS‐650032, a novel HCV NS3 inhibitor, in subjects with chronic genotype 1 HCV infection. Hepatology 2009;50(4 Suppl):411A‐2A. CENTRAL
Pasquinelli C, McPhee F, Eley T, Villegas C, Sandy K, Sheridan P, et al. Single‐ and multiple‐ascending‐dose studies of the NS3 protease inhibitor asunaprevir in subjects with or without chronic hepatitis C. Antimicrobial Agents and Chemotherapy 2012;56(4):1838‐44. CENTRAL

Pasquinelli 2012a2 {published data only}

Pasquinelli C, Eley T, Villegas C, Sandy K, Mathias E, Wendelburg P, et al. Safety, tolerability, pharmacokinetics and antiviral activity following single and multiple‐dose administration of BMS‐650032, a novel HCV NS3 inhibitor, in subjects with chronic genotype 1 HCV infection. Hepatology International 2010;4(1):184. CENTRAL
Pasquinelli C, Eley T, Villegas C, Sandy K, Mathias E, Wendelburg P, et al. Safety, tolerability, pharmacokinetics and antiviral activity following single‐and multiple‐dose administration of BMS‐650032, a novel HCV NS3 inhibitor, in subjects with chronic genotype 1 HCV infection. Hepatology 2009;50(4 Suppl):411A‐2A. CENTRAL
Pasquinelli C, McPhee F, Eley T, Villegas C, Sandy K, Sheridan P, et al. Single‐ and multiple‐ascending‐dose studies of the NS3 protease inhibitor asunaprevir in subjects with or without chronic hepatitis C. Antimicrobial Agents and Chemotherapy 2012;56(4):1838‐44. CENTRAL

Pearlman 2014 {published data only}

Pearlman BL, Ehleben C. Hepatitis C genotype 1 virus with low viral load and rapid virologic response to peginterferon/ribavirin obviates a protease inhibitor. Hepatology 2014;59(1):71‐7. CENTRAL
Wang AS, Pfeiffer RM, Morgan TR, O'Brien TR. Re: Pearlman and Ehleben ‐ hepatitis C genotype 1 virus with low viral load and rapid virologic response to peginterferon/ribavirin obviates a protease inhibitor. Hepatology 2014;59(6):2423–4. CENTRAL

Pearlman 2015 {published data only}

Pearlman BL, Ehleben C, Perrys M. The combination of simeprevir and sofosbuvir is more effective than that of peginterferon, ribavirin, and sofosbuvir for patients with hepatitis C‐related Child's class A cirrhosis. Gastroenterology 2015;148(4):762. CENTRAL

Petry 2011 {published data only}

Petry AS, Fraser IP, O’Mara E, Van Dyck K, Nachbar RB, De Lepeleire IM, et al. Safety and antiviral activity of MK‐5172, a next generation HCV NS3/4A protease inhibitor with a broad HCV genotypic activity spectrum and potent activity against known resistance mutants, in genotype 1 and 3 HCV infected patients. Hepatology 2011;54(4 Suppl):531A. CENTRAL

Pockros 2008a1 {published data only}

Nelson D, Pockros PJ, Godofsky E, Rodriguez‐Torres M, Everson G, Fried M, et al. High end‐of‐treatment response (84%) after 4 weeks of R1626, peginterferon alfa‐2A (40KD) and ribavirin followed by a further 44 weeks of peginterferon alfa‐2A and ribavirin. Journal of Hepatology 2008;48(Suppl 2):S371. CENTRAL
Pockros PJ, Nelson D, Godofsky E, Rodriguez‐Torres M, Everson GT, Fried MW, et al. R1626 plus peginterferon alfa‐2a provides potent suppression of hepatitis C virus RNA and significant antiviral synergy in combination with ribavirin. Hepatology 2008;48(2):385‐97. CENTRAL

Pockros 2008a2 {published data only}

Nelson D, Pockros PJ, Godofsky E, Rodriguez‐Torres M, Everson G, Fried M, et al. High end‐of‐treatment response (84%) after 4 weeks of R1626, peginterferon alfa‐2A (40KD) and ribavirin followed by a further 44 weeks of peginterferon alfa‐2A and ribavirin. Journal of Hepatology 2008;48(Suppl 2):S371. CENTRAL
Pockros PJ, Nelson D, Godofsky E, Rodriguez‐Torres M, Everson GT, Fried MW, et al. R1626 plus peginterferon alfa‐2a provides potent suppression of hepatitis C virus RNA and significant antiviral synergy in combination with ribavirin. Hepatology 2008;48(2):385‐97. CENTRAL

Pockros 2008a3 {published data only}

Nelson D, Pockros PJ, Godofsky E, Rodriguez‐Torres M, Everson G, Fried M, et al. High end‐of‐treatment response (84%) after 4 weeks of R1626, peginterferon alfa‐2A (40KD) and ribavirin followed by a further 44 weeks of peginterferon alfa‐2A and ribavirin. Journal of Hepatology 2008;48(Suppl 2):S371. CENTRAL
Pockros PJ, Nelson D, Godofsky E, Rodriguez‐Torres M, Everson GT, Fried MW, et al. R1626 plus peginterferon alfa‐2a provides potent suppression of hepatitis C virus RNA and significant antiviral synergy in combination with ribavirin. Hepatology 2008;48(2):385‐97. CENTRAL

Pockros 2009 {published data only}

Pockros P, Rodriguez‐Torres M, Villano S, Maller E, Chojkier M. A phase 2, randomized study of HCV‐796 in combination with pegylated‐interferon (PEG) plus ribavirin (RBV) versus peg plus RBV in hepatitis C virus genotype‐1 infection. Journal of Hepatology 2009;50(Suppl S1):S7. CENTRAL

Pol 2012 {published data only}

Pol S, Ghalib RH, Rustgi VK, Martorell C, Everson GT, Tatum HA, et al. Daclatasvir for previously untreated chronic hepatitis C genotype‐1 infection: a randomised, parallel‐group, double‐blind, placebo‐controlled, dose‐finding, phase 2a trial. Lancet Infectious Diseases 2012;12(9):671‐7. CENTRAL

Pol 2013 {published data only}

Pol S, Jablkowski M, Trenkle JD, Kanwar B, Bekele BN, Subramanian M, et al. Antiviral efficacy of the NS3 protease inhibitor, GS‐9451, non‐nucleoside NS5b inhibitor, tegobuvir, and pegylated interferon plus ribavirin in treatment‐naive genotype 1 hepatitis C infected patients. Journal of Hepatology 2013;58(Suppl S1):S31. CENTRAL

Poordad 2007 {published data only}

Poordad F, Lawitz EJ, Gitlin N, Rodriguez‐Torres M, Box T, Nguyen T, et al. Efficacy and safety of valopicitabine in combination with pegylated interferon (Peg IFN) and ribavirin (RBV) in patients with chronic hepatitis C. Hepatology 2007;46(4 Suppl 1):866A. CENTRAL

Poordad 2011a1 {published data only}

Albrecht J, Vierling J, Kwo P, Lawitz E, McCone J, Schiff E. Frequencies of resistance‐associated amino acid variants (RAV) following treatment with boceprevir (BOC) plus peginterferon alfa‐2b and ribavirin (P/R). Hepatology International 2011;5(1):261‐2. CENTRAL
Barnard RJO, Pedicone LD, Chaudhri E, Tong X, Qiu P, Brass CA. Frequencies of resistance‐associated amino acid variants following combination treatment with boceprevir (BOC) plus Pegintron (peglnterferon alfa‐2b) and ribavirin (P/R) in patients with chronic hepatitis C (CHC), genotype 1 (G1). Journal of the International Association of Physicians in AIDS Care 2011;10(3):199‐200. CENTRAL
Bronowicki J, McCone J, Bacon BR, Bruno S, Manns MP, Sulkowski MS, et al. Response‐guided therapy (RGT) with boceprevir (BOC) + peginterferon alfa‐2b/ribavirin (P/R) for treatment‐naive patients with hepatitis C virus (HCV) genotype (G) 1 was similar to a 48‐wk fixed‐duration regimen with BOC + P/R in SPRINT‐2. Hepatology 2010;52(Suppl S1):881A. CENTRAL
Charlton M. Boceprevir (Victrelis) for HCV: V is for victory and very complex. Hepatology 2011;54(5):1882‐6. CENTRAL
Manns MP, Markova AA, Serrano BC, Cornberg M. Phase III results of boceprevir in treatment naive patients with chronic hepatitis C genotype 1. Liver International 2012;32(Suppl 1):27‐31. CENTRAL
Manns MP, McCone J, Davis M, Shiffman ML, Rossaro L, Bourliere M, et al. Safety benefits of response‐guided therapy with boceprevir (BOC) plus peginterferon alfa‐2B/ribavirin (PR) in previously untreated patients with HCV genotype 1 infection. Hepatology 2011;54(4 Suppl):813A. CENTRAL
McCone J, Jacobson IM, Bacon BR, Pedicone L, Goteti VS, Burroughs M, et al. Treatment‐naive black patients treated with boceprevir combined with peginterferon alfa‐2b + ribavirin: results from HCV sprint‐2. Hepatology 2011;54(4 Suppl):822A‐3A. CENTRAL
Poordad F, McCone J, Bacon BR, Bruno S, Manns MP, Sulkowski MS, et al. Boceprevir (BOC) combined with peginterferon alfa‐2b/ribavirin (P/R) for treatment‐naive patients with hepatitis C virus (HCV) genotype (G) 1: SPRINT‐2 final results. Hepatology 2010;52(Suppl S1):402A. CENTRAL
Poordad F, McCone J, Bacon BR, Bruno S, Manns MP, Sulkowski MS, et al. Boceprevir for untreated chronic HCV genotype 1 infection. New England Journal of Medicine 2011;364(13):1195‐206. CENTRAL
Reddy KR, Bruno S, Rossaro L, Ravendhran N, Pauly MP, Boparai N, et al. Predictors of sustained virologic response among treatment‐naive patients with hepatitis C virus genotype 1 when treated with boceprevir (BOC) plus peginterferon alfa‐2B/ribavirin (PR). Journal of Hepatology 2011;54(Suppl 1):S190. CENTRAL
Sulkowski MS, Bacon BR, Gordon SC, Lawitz E, Marcellin P, Vierling JM, et al. Boceprevir combined with peginterferon alfa‐2b/ribavirin for previous treatment failure patients with hepatitis C virus genotype 1: RESPOND‐2 final results. Journal of the International Association of Physicians in AIDS Care 2011;10(3):197‐8. CENTRAL
Sulkowski MS, Poordad F, Manns MP, Bronowicki JP, Rajender RK, Harrison SA, et al. Anemia during treatment with peginterferon alfa‐2b/ribavirin and boceprevir: analysis from the serine protease inhibitor therapy 2 (SPRINT‐2) trial. Hepatology 2013;57(3):974‐84. CENTRAL

Poordad 2011a2 {published data only}

Bronowicki J, McCone J, Bacon BR, Bruno S, Manns MP, Sulkowski MS, et al. Response‐guided therapy (RGT) with boceprevir (BOC) + peginterferon alfa‐2b/ribavirin (P/R) for treatment‐naive patients with hepatitis C virus (HCV) genotype (G) 1 was similar to a 48‐wk fixed‐duration regimen with BOC + P/R in SPRINT‐2. Hepatology 2010;52(Suppl S1):881A. CENTRAL
Charlton M. Boceprevir (Victrelis) for HCV: V is for victory and very complex. Hepatology 2011;54(5):1882‐6. CENTRAL
Manns MP, Markova AA, Serrano BC, Cornberg M. Phase III results of boceprevir in treatment naive patients with chronic hepatitis C genotype 1. Liver International 2012;32(Suppl 1):27‐31. CENTRAL
Manns MP, McCone J, Davis M, Shiffman ML, Rossaro L, Bourliere M, et al. Safety benefits of response‐guided therapy with boceprevir (BOC) plus peginterferon alfa‐2B/ribavirin (PR) in previously untreated patients with HCV genotype 1 infection. Hepatology 2011;54(4 Suppl):813A. CENTRAL
McCone J, Jacobson IM, Bacon BR, Pedicone L, Goteti VS, Burroughs M, et al. Treatment‐naive black patients treated with boceprevir combined with peginterferon alfa‐2b + ribavirin: results from HCV sprint‐2. Hepatology 2011;54(4 Suppl):822A‐3A. CENTRAL
Poordad F, McCone J, Bacon BR, Bruno S, Manns MP, Sulkowski MS, et al. Boceprevir (BOC) combined with peginterferon alfa‐2b/ribavirin (P/R) for treatment‐naive patients with hepatitis C virus (HCV) genotype (G) 1: SPRINT‐2 final results. Hepatology 2010;52(Suppl S1):402A. CENTRAL
Poordad F, McCone J, Bacon BR, Bruno S, Manns MP, Sulkowski MS, et al. Boceprevir for untreated chronic HCV genotype 1 infection. New England Journal of Medicine 2011;364(13):1195‐206. CENTRAL
Reddy KR, Bruno S, Rossaro L, Ravendhran N, Pauly MP, Boparai N, et al. Predictors of sustained virologic response among treatment‐naive patients with hepatitis C virus genotype 1 when treated with boceprevir (BOC) plus peginterferon alfa‐2B/ribavirin (PR). Journal of Hepatology 2011;54(Suppl 1):S190. CENTRAL
Sulkowski MS, Bacon BR, Gordon SC, Lawitz E, Marcellin P, Vierling JM, et al. Boceprevir combined with peginterferon alfa‐2b/ribavirin for previous treatment failure patients with hepatitis C virus genotype 1: RESPOND‐2 final results. Journal of the International Association of Physicians in AIDS Care 2011;10(3):197‐8. CENTRAL
Sulkowski MS, Poordad F, Manns MP, Bronowicki JP, Rajender RK, Harrison SA, et al. Anemia during treatment with peginterferon alfa‐2b/ribavirin and boceprevir: analysis from the serine protease inhibitor therapy 2 (SPRINT‐2) trial. Hepatology 2013;57(3):974‐84. CENTRAL

POSITRON 2013 {published data only}

Jacobson IM, Gordon SC, Kowdley KV, Yoshida EM, Rodriguez‐Torres M, Sulkowski MS, et al. Sofosbuvir for hepatitis C genotype 2 or 3 in patients without treatment options. New England Journal of Medicine 2013;368(20):1867‐77. CENTRAL
Younossi ZM, Stepanova M, Henry L, Gane E, Jacobson IM, Lawitz E, et al. Minimal impact of sofosbuvir and ribavirin on health related quality of life in chronic hepatitis C (CH‐C). Journal of Hepatology 2014;60(4):741‐7. [PUBMED: 24333184]CENTRAL

Reddy 2007 {published data only}

Reddy R, Rodriguez‐Torres M, Gane E, Robson R, Lalezari J, Everson GT, et al. Antiviral activity, pharmacokinetics, safety, and tolerability of R7128, a novel nucleoside HCV RNA polymerase inhibitor, following multiple, ascending, oral doses in patients with HCV genotype 1 infection who have failed prior interferon therapy. Hepatology 2007;46(Suppl S1):862A‐3A. CENTRAL

Reesink 2006 {published data only}

Reesink HW, Zeuzem S, Van Vliet A, McNair L, Purdy S, Chu H, et al. Initial results of a phase 1b, multiple‐dose study of VX‐950, a hepatitis C virus protease inhibitor. Gastroenterology 2005;128(4 Suppl 2):A697. CENTRAL
Reesink HW, Zeuzem S, Weegink CJ, Forestier N, Van Vliet A, Van De Wetering De Rooij J, et al. Rapid decline of viral RNA in hepatitis C patients treated with VX‐950: a phase Ib, placebo‐controlled, randomized study. Gastroenterology 2006;131(4):997‐1002. CENTRAL

Reiser 2005 {published data only}

Reiser M, Hinrichsen H, Benhamou Y, Reesink HW, Wedemeyer H, Avendano C, et al. Antiviral efficacy of NS3‐serine protease inhibitor BILN‐2061 in patients with chronic genotype 2 and 3 hepatitis C. Hepatology (Baltimore, Md.) 2005;41(4):832‐5. CENTRAL

Rodriguez‐Torres 2008 {published data only}

Rodriguez‐Torres M, Lalezari J, Gane EJ, DeJesus E, Nelson DR, Everson GT, et al. Potent antiviral response to the HCV nucleoside polymerase inhibitor R7128 for 28 days with peg‐ifn and ribavirin: subanalysis by race/ethnicity, weight and HCV genotype. Hepatology 2008;48(4 Suppl):1160A. CENTRAL

Rodriguez‐Torres 2010 {published data only}

Rodriguez‐Torres M, Lawitz E, Conway B, Kaita K, Sheikh AM, Ghalib R, et al. Safety and antiviral activity of the HCV non‐nucleoside polymerase inhibitor VX‐222 in treatment‐naive genotype 1 HCV infected patients. Journal of Hepatology 2010;52(Suppl S1):S14. CENTRAL

Rodriguez‐Torres 2011a1 {published data only}

Rodriguez‐Torres M, Lawitz E, Hazan L, Barry AN, Wenzel ED, Alam J. Antiviral activity and safety of INX‐08189, a nucleotide polymerase inhibitor, following 7‐days of oral therapy in naive genotype‐1 chronic HCV patients. Hepatology 2011;54(Suppl S1):535A. CENTRAL

Rodriguez‐Torres 2011a2 {published data only}

Rodriguez‐Torres M, Lawitz E, Denning J, Cornpropst M, Albanis E, Symonds W, et al. PSI‐352938, a novel purine nucleotide analog, exhibits potent antiviral activity and no evidence of resistance in patients with HCV genotype 1 over 7 days. Journal of Hepatology 2011;54(Suppl S1):S488. CENTRAL

Rodriguez‐Torres 2013 {published data only}

Rodriguez‐Torres M, Lawitz E, Kowdley KV, Nelson DR, Dejesus E, McHutchison JG, et al. Sofosbuvir (GS‐7977) plus peginterferon/ribavirin in treatment‐naïve patients with HCV genotype 1: a randomized, 28‐day, dose‐ranging trial. Journal of Hepatology 2013;58(4):663‐8. CENTRAL

Rodriguez‐Torres 2014a1 {published data only}

Rodriguez‐Torres M, Stoehr A, Gane EJ, Serfaty L, Lawitz E, Zhou A, et al. Combination of vaniprevir with peginterferon and ribavirin significantly increases the rate of SVR in treatment‐experienced patients with chronic HCV genotype 1 infection and cirrhosis. Clinical Gastroenterology and Hepatology 2014;12(6):1029‐37.e5. CENTRAL

Rodriguez‐Torres 2014a2 {published data only}

Rodriguez‐Torres M, Stoehr A, Gane EJ, Serfaty L, Lawitz E, Zhou A, et al. Combination of vaniprevir with peginterferon and ribavirin significantly increases the rate of SVR in treatment‐experienced patients with chronic HCV genotype 1 infection and cirrhosis. Clinical Gastroenterology and Hepatology 2014;12(6):1029‐37.e5. CENTRAL

Rodriguez‐Torres 2014a3 {published data only}

Rodriguez‐Torres M, Stoehr A, Gane EJ, Serfaty L, Lawitz E, Zhou A, et al. Combination of vaniprevir with peginterferon and ribavirin significantly increases the rate of SVR in treatment‐experienced patients with chronic HCV genotype 1 infection and cirrhosis. Clinical Gastroenterology and Hepatology 2014;12(6):1029‐37.e5. CENTRAL

Rodriguez‐Torres 2014a4 {published data only}

Rodriguez‐Torres M, Stoehr A, Gane EJ, Serfaty L, Lawitz E, Zhou A, et al. Combination of vaniprevir with peginterferon and ribavirin significantly increases the rate of SVR in treatment‐experienced patients with chronic HCV genotype 1 infection and cirrhosis. Clinical Gastroenterology and Hepatology 2014;12(6):1029‐37.e5. CENTRAL

Rodriguez‐Torres 2014b1 {published data only}

Rodriguez‐Torres M, Yoshida EM, Marcellin P, Srinivasan S, Purohit VS, Wang C, et al. A phase 2 study of filibuvir in combination with pegylated IFN alfa and ribavirin for chronic HCV. Annals of Hepatology 2014;13(4):364‐75. CENTRAL

Rodriguez‐Torres 2014b2 {published data only}

Rodriguez‐Torres M, Yoshida EM, Marcellin P, Srinivasan S, Purohit VS, Wang C, et al. A phase 2 study of filibuvir in combination with pegylated IFN alfa and ribavirin for chronic HCV. Annals of Hepatology 2014;13(4):364‐75. CENTRAL

Rodriguez‐Torres 2015 {published data only}

Rodriguez‐Torres M, Glass S, Hill J, Freilich B, Hassman D, Di Bisceglie A, et al. The pan‐genotypic NS3/4A protease inhibitor GS‐9857 demonstrates potent antiviral activity in patients infected with HCV genotype 1, 2, 3 or 4 in a 3‐day monotherapy study. Journal of Hepatology 2015;62(Suppl S2):S682. CENTRAL

Sarrazin 2007 {published data only}

Sarrazin C, Rouzier R, Wagner F, Forestier N, Larrey D, Gupta SK. SCH 503034, a novel hepatitis C virus protease inhibitor, plus pegylated interferon alpha‐2b for genotype 1 nonresponders. Gastroenterology 2007;132(4):1270‐8. CENTRAL
Vermehren J, Susser S, Karey U, Forestier N, Lange CM, Hughes EA, et al. Clonal analysis of mutations selected in the HCV NS3 protease domain of genotype 1 non‐responders sequentially treated with boceprevir (SCH503034) and/or pegylated interferon alfa‐2b (PEG‐IFN α‐2b). Hepatology 2009;50(4 Suppl):1040A‐1A. CENTRAL

Schiff 2008 {published data only}

Schiff E, Poordad F, Jacobson I, Flamm S, Bacon B, Lawitz E, et al. Boceprevir (B) combination therapy in null responders (NR): response dependent on interferon responsiveness. Journal of Hepatology 2008;48(Suppl 2):S46. CENTRAL

Silva 2013a1 {published data only}

Silva MO, Treitel M, Graham DJ, Curry S, Frontera MJ, McMonagle P, et al. Antiviral activity of boceprevir monotherapy in treatment‐naive subjects with chronic hepatitis C genotype 2/3. Journal of Hepatology 2013;59(1):31‐7. CENTRAL

Silva 2013a2 {published data only}

Silva MO, Treitel M, Graham DJ, Curry S, Frontera MJ, McMonagle P, et al. Antiviral activity of boceprevir monotherapy in treatment‐naive subjects with chronic hepatitis C genotype 2/3. Journal of Hepatology 2013;59(1):31‐7. CENTRAL

Silva 2013a3 {published data only}

Silva MO, Treitel M, Graham DJ, Curry S, Frontera MJ, McMonagle P, et al. Antiviral activity of boceprevir monotherapy in treatment‐naive subjects with chronic hepatitis C genotype 2/3. Journal of Hepatology 2013;59(1):31‐7. CENTRAL

Sims 2014 {published data only}

Sims KD, Lemm J, Eley T, Liu M, Berglind A, Sherman D, et al. Randomized, placebo‐controlled, single‐ascending‐dose study of BMS‐791325, a hepatitis C virus (HCV) NS5B polymerase inhibitor, in HCV genotype 1 infection. Antimicrobial Agents and Chemotherapy 2014;58(6):3496‐503. CENTRAL

STARTVerso‐1 2015a1 {published data only}

Asselah T, Jensen DM, Foster G, Sulkowski M, Ouzan D, Morano L, et al. Faldaprevir plus pegylated interferon/ribavirin did not increase anaemia compared with pegylated interferon/ribavirin in HCV genotype‐1, treatment‐naive patients: pooled analysis of phase III studies. Gastroenterology 2014;146(5 Suppl 1):S‐977. CENTRAL
Asselah T, Jensen DM, Foster G, Sulkowski M, Ouzan D, Morano L, et al. Virological response in treatment‐naive patients with chronic HCV genotype‐1 infection receiving faldaprevir plus pegylated interferon alpha‐2a and ribavirin is unaffected by ribavirin dose reduction. Gastroenterology 2014;146(5 Suppl 1):S975‐6. CENTRAL
Berger K, Sarrazin C, Ferenci P, Jensen DM, Jacobson IM, Stern JO, et al. NS3 Q80K did not impact efficacy or treatment‐emergent resistance patterns in HCV genotype 1‐infected patients receiving faldaprevir + peginterferon/ribavirin in three phase III trials. Journal of Hepatology 2014;60(1 Suppl S):S497. CENTRAL
Ferenci P, Asselah T, Foster GR, Zeuzem S, Sarrazin C, Moreno C, et al. Faldaprevir plus pegylated interferon alfa‐2A and ribavirin in chronic HCV genotype‐1 treatment‐naive patients: final results from STARTVerso1, a randomised, double‐blind, placebo‐controlled phase III trial. Journal of Gastroenterology and Hepatology 2013;28(Suppl 2):157‐8. CENTRAL
Ferenci P, Asselah T, Foster GR, Zeuzem S, Sarrazin C, Moreno C, et al. STARTVerso1: a randomized trial of faldaprevir plus pegylated interferon/ribavirin for chronic HCV genotype‐1 infection. Journal of Hepatology 2015;62(6):1246‐55. CENTRAL
Ferenci P, Dieterich D, Zeuzem S, Mantry P, Sarrazin C, Crespo J, et al. Early stopping rules for faldaprevir plus pegylated interferon alpha‐2a and ribavirin in treatment‐naive patients: exploratory study of pooled data from phase III trials. Journal of Hepatology 2014;60(1 Suppl 1):S452‐S3. CENTRAL
Foster GR, Cooper C, Dieterich D, Ferenci P, Crespo J, Diago M, et al. Pharmacokinetic‐response analysis of faldaprevir in treatment‐naive patients with chronic HCV genotype‐1 infection: a pooled analysis of two phase III trials. Journal of Hepatology 2014;60(1 Suppl 1):S454‐5. CENTRAL
Jensen DM, Asselah T, Dieterich DT, Foster GR, Sulkowski MS, Zeuzem S, et al. A pooled analysis of two randomized, double‐blind placebo‐controlled phase III trials (STARTVerso1 & 2) of faldaprevir plus pegylated interferon alfa‐2a and ribavirin in treatment‐naive patients with chronic hepatitis C genotype‐1 infection. Hepatology 2013;58(Suppl 1):734A‐5A. CENTRAL
Jensen DM, Foster G, Ferenci P, Cooper C, Dieterich DT, Sulkowski M, et al. Faldaprevir plus pegylated interferon alfa‐2a and ribavirin in treatment‐naive patients with chronic hepatitis C genotype‐1 infection: a pooled analysis of two randomized, double‐blind placebo‐controlled phase III trials (STARTVerso 1 & 2). Gastroenterology 2014;146(5 Suppl 1):S974. CENTRAL
Moreno C, Ferenci P, Asselah T, Foster G, Zeuzem S, Sarrazin C, et al. Faldaprevir plus pegylated interferon alfa‐2a and ribavirin in chronic HCV genotype‐1 treatment‐naive patients: final results from STARTVerso1: a randomized, double‐blind, placebo‐controlled, phase 3 trial. American Journal of Gastroenterology 2013;108(Suppl 1):S145. CENTRAL
Yoshida EM, Asselah T, Moreno C, Maevskaya MV, Kao JH, Crespo J, et al. Subgroup analyses and baseline predictors of response with faldaprevir plus pegylated interferon alfa‐2a and ribavirin in treatment‐naive patients with chronic hepatitis C genotype‐1 infection: a pooled analysis of STARTVerso1 and 2. Hepatology 2013;58(Suppl S1):751A‐2A. CENTRAL
Zeuzem S, Jensen DM, Carvalho A, Diago M, Feinman SV, Kowdley KV, et al. No effect of faldaprevir on renal function in treatment‐naive patients with chronic HCV genotype‐1 infection: pooled data from two phase III trials. Journal of Hepatology 2014;60(1 Suppl S):S451. CENTRAL

STARTVerso‐1 2015a2 {published data only}

Asselah T, Jensen DM, Foster G, Sulkowski M, Ouzan D, Morano L, et al. Faldaprevir plus pegylated interferon/ribavirin did not increase anaemia compared with pegylated interferon/ribavirin in HCV genotype‐1, treatment‐naive patients: pooled analysis of phase III studies. Gastroenterology 2014;146(5 Suppl 1):S‐977. CENTRAL
Asselah T, Jensen DM, Foster G, Sulkowski M, Ouzan D, Morano L, et al. Virological response in treatment‐naive patients with chronic HCV genotype‐1 infection receiving faldaprevir plus pegylated interferon alpha‐2a and ribavirin is unaffected by ribavirin dose reduction. Gastroenterology 2014;146(5 Suppl 1):S975‐6. CENTRAL
Berger K, Sarrazin C, Ferenci P, Jensen DM, Jacobson IM, Stern JO, et al. NS3 Q80K did not impact efficacy or treatment‐emergent resistance patterns in HCV genotype 1‐infected patients receiving faldaprevir + peginterferon/ribavirin in three phase III trials. Journal of Hepatology 2014;60(1 Suppl S):S497. CENTRAL
Ferenci P, Asselah T, Foster GR, Zeuzem S, Sarrazin C, Moreno C, et al. Faldaprevir plus pegylated interferon alfa‐2A and ribavirin in chronic HCV genotype‐1 treatment‐naive patients: final results from STARTVerso1, a randomised, double‐blind, placebo‐controlled phase III trial. Journal of Gastroenterology and Hepatology 2013;28(Suppl 2):157‐8. CENTRAL
Ferenci P, Asselah T, Foster GR, Zeuzem S, Sarrazin C, Moreno C, et al. STARTVerso1: a randomized trial of faldaprevir plus pegylated interferon/ribavirin for chronic HCV genotype‐1 infection. Journal of Hepatology 2015;62(6):1246‐55. CENTRAL
Ferenci P, Dieterich D, Zeuzem S, Mantry P, Sarrazin C, Crespo J, et al. Early stopping rules for faldaprevir plus pegylated interferon alpha‐2a and ribavirin in treatment‐naive patients: exploratory study of pooled data from phase III trials. Journal of Hepatology 2014;60(1 Suppl 1):S452‐S3. CENTRAL
Foster GR, Cooper C, Dieterich D, Ferenci P, Crespo J, Diago M, et al. Pharmacokinetic‐response analysis of faldaprevir in treatment‐naive patients with chronic HCV genotype‐1 infection: a pooled analysis of two phase III trials. Journal of Hepatology 2014;60(1 Suppl 1):S454‐5. CENTRAL
Jensen DM, Asselah T, Dieterich DT, Foster GR, Sulkowski MS, Zeuzem S, et al. A pooled analysis of two randomized, double‐blind placebo‐controlled phase III trials (STARTVerso1 & 2) of faldaprevir plus pegylated interferon alfa‐2a and ribavirin in treatment‐naive patients with chronic hepatitis C genotype‐1 infection. Hepatology 2013;58(Suppl 1):734A‐5A. CENTRAL
Jensen DM, Foster G, Ferenci P, Cooper C, Dieterich DT, Sulkowski M, et al. Faldaprevir plus pegylated interferon alfa‐2a and ribavirin in treatment‐naive patients with chronic hepatitis C genotype‐1 infection: a pooled analysis of two randomized, double‐blind placebo‐controlled phase III trials (STARTVerso 1 & 2). Gastroenterology 2014;146(5 Suppl 1):S974. CENTRAL
Moreno C, Ferenci P, Asselah T, Foster G, Zeuzem S, Sarrazin C, et al. Faldaprevir plus pegylated interferon alfa‐2a and ribavirin in chronic HCV genotype‐1 treatment‐naive patients: final results from STARTVerso1: a randomized, double‐blind, placebo‐controlled, phase 3 trial. American Journal of Gastroenterology 2013;108(Suppl 1):S145. CENTRAL
Yoshida EM, Asselah T, Moreno C, Maevskaya MV, Kao JH, Crespo J, et al. Subgroup analyses and baseline predictors of response with faldaprevir plus pegylated interferon alfa‐2a and ribavirin in treatment‐naive patients with chronic hepatitis C genotype‐1 infection: a pooled analysis of STARTVerso1 and 2. Hepatology 2013;58(Suppl S1):751A‐2A. CENTRAL
Zeuzem S, Jensen DM, Carvalho A, Diago M, Feinman SV, Kowdley KV, et al. No effect of faldaprevir on renal function in treatment‐naive patients with chronic HCV genotype‐1 infection: pooled data from two phase III trials. Journal of Hepatology 2014;60(1 Suppl S):S451. CENTRAL

STARTverso‐2 2014a1 {published data only}

Asselah T, Jensen DM, Foster G, Sulkowski M, Ouzan D, Morano L, et al. Faldaprevir plus pegylated interferon/ribavirin did not increase anaemia compared with pegylated interferon/ribavirin in HCV genotype‐1, treatment‐naive patients: pooled analysis of phase III studies. Gastroenterology 2014;146(5 (Suppl 1)):S‐977. CENTRAL
Asselah T, Jensen DM, Foster G, Sulkowski M, Ouzan D, Morano L, et al. Virological response in treatment‐naive patients with chronic HCV genotype‐1 infection receiving faldaprevir plus pegylated interferon alpha‐2a and ribavirin is unaffected by ribavirin dose reduction. Gastroenterology 2014;146(5 (Suppl 1)):S975‐6. CENTRAL
Berger K, Sarrazin C, Ferenci P, Jensen DM, Jacobson IM, Stern JO, et al. NS3 Q80K did not impact efficacy or treatment‐emergent resistance patterns in HCV genotype 1‐infected patients receiving faldaprevir + peginterferon/ribavirin in three phase III trials. Journal of Hepatology 2014;60(1):S497. CENTRAL
Ferenci P, Dieterich D, Zeuzem S, Mantry P, Sarrazin C, Crespo J, et al. Early stopping rules for faldaprevir plus pegylated interferon alpha‐2a and ribavirin in treatment‐naive patients: exploratory study of pooled data from phase III trials. Journal of Hepatology 2014;60(1 (Suppl 1)):S452‐3. CENTRAL
Foster GR, Cooper C, Dieterich D, Ferenci P, Crespo J, Diago M, et al. Pharmacokinetic‐response analysis of faldaprevir in treatment‐naive patients with chronic HCV genotype‐1 infection: a pooled analysis of two phase III trials. Journal of Hepatology 2014;60(1 (Suppl 1)):S454‐5. CENTRAL
Jensen DM, Asselah T, Dieterich DT, Foster GR, Sulkowski MS, Zeuzem S, et al. A pooled analysis of two randomized, double‐blind placebo‐controlled phase III trials (STARTVerso1&2) of faldaprevir plus pegylated interferon alfa‐2a and ribavirin in treatment‐naive patients with chronic hepatitis C genotype‐1 infection. Hepatology 2013;58(Suppl 1):734A‐5A. CENTRAL
Jensen DM, Foster G, Ferenci P, Cooper C, Dieterich DT, Sulkowski M, et al. Faldaprevir plus pegylated interferon alfa‐2a and ribavirin in treatment‐naive patients with chronic hepatitis C genotype‐1 infection: a pooled analysis of two randomized, double‐blind placebo‐controlled phase iii trials (STARTVerso1&2). Gastroenterology 2014;146(5 (Suppl 1)):S974. CENTRAL
Yoshida EM, Asselah T, Moreno C, Maevskaya MV, Kao JH, Crespo J, et al. Subgroup analyses and baseline predictors of response with faldaprevir plus pegylated interferon alfa‐2a and ribavirin in treatment‐naive patients with chronic hepatitis C genotype‐1 infection: a pooled analysis of STARTVerso1 and 2. Hepatology 2013;58(4 Suppl):751A‐2A. CENTRAL
Zeuzem S, Jensen DM, Carvalho A, Diago M, Feinman SV, Kowdley KV, et al. No effect of faldaprevir on renal function in treatment‐naive patients with chronic HCV genotype‐1 infection: pooled data from two phase III trials. Journal of Hepatology 2014;60(1 (Suppl 1)):S451. CENTRAL

STARTverso‐2 2014a2 {published data only}

Asselah T, Jensen DM, Foster G, Sulkowski M, Ouzan D, Morano L, et al. Faldaprevir plus pegylated interferon/ribavirin did not increase anaemia compared with pegylated interferon/ribavirin in HCV genotype‐1, treatment‐naive patients: pooled analysis of phase III studies. Gastroenterology 2014;146(5 (Suppl 1)):S‐977. CENTRAL
Asselah T, Jensen DM, Foster G, Sulkowski M, Ouzan D, Morano L, et al. Virological response in treatment‐naive patients with chronic HCV genotype‐1 infection receiving faldaprevir plus pegylated interferon alpha‐2a and ribavirin is unaffected by ribavirin dose reduction. Gastroenterology 2014;146(5 (Suppl 1)):S975‐6. CENTRAL
Berger K, Sarrazin C, Ferenci P, Jensen DM, Jacobson IM, Stern JO, et al. NS3 Q80K did not impact efficacy or treatment‐emergent resistance patterns in HCV genotype 1‐infected patients receiving faldaprevir + peginterferon/ribavirin in three phase III trials. Journal of Hepatology 2014;60(1):S497. CENTRAL
Ferenci P, Dieterich D, Zeuzem S, Mantry P, Sarrazin C, Crespo J, et al. Early stopping rules for faldaprevir plus pegylated interferon alpha‐2a and ribavirin in treatment‐naive patients: exploratory study of pooled data from phase III trials. Journal of Hepatology 2014;60(1 (Suppl 1)):S452‐3. CENTRAL
Foster GR, Cooper C, Dieterich D, Ferenci P, Crespo J, Diago M, et al. Pharmacokinetic‐response analysis of faldaprevir in treatment‐naive patients with chronic HCV genotype‐1 infection: a pooled analysis of two phase III trials. Journal of Hepatology 2014;60(1 (Suppl 1)):S454‐5. CENTRAL
Jensen DM, Asselah T, Dieterich DT, Foster GR, Sulkowski MS, Zeuzem S, et al. A pooled analysis of two randomized, double‐blind placebo‐controlled phase III trials (STARTVerso1&2) of faldaprevir plus pegylated interferon alfa‐2a and ribavirin in treatment‐naive patients with chronic hepatitis C genotype‐1 infection. Hepatology 2013;58(Suppl 1):734A‐5A. CENTRAL
Jensen DM, Foster G, Ferenci P, Cooper C, Dieterich DT, Sulkowski M, et al. Faldaprevir plus pegylated interferon alfa‐2a and ribavirin in treatment‐naive patients with chronic hepatitis C genotype‐1 infection: a pooled analysis of two randomized, double‐blind placebo‐controlled phase iii trials (STARTVerso1&2). Gastroenterology 2014;146(5 (Suppl 1)):S974. CENTRAL
Yoshida EM, Asselah T, Moreno C, Maevskaya MV, Kao JH, Crespo J, et al. Subgroup analyses and baseline predictors of response with faldaprevir plus pegylated interferon alfa‐2a and ribavirin in treatment‐naive patients with chronic hepatitis C genotype‐1 infection: a pooled analysis of STARTVerso1 and 2. Hepatology 2013;58(4 Suppl):751A‐2A. CENTRAL
Zeuzem S, Jensen DM, Carvalho A, Diago M, Feinman SV, Kowdley KV, et al. No effect of faldaprevir on renal function in treatment‐naive patients with chronic HCV genotype‐1 infection: pooled data from two phase III trials. Journal of Hepatology 2014;60(1 (Suppl 1)):S451. CENTRAL

STARTverso‐3 2013a1 {published data only}

Asselah T, Zehnter E, Agarwal K, Sakai Y, Yatsuhashi H, Willems B, et al. Pharmacokinetic‐response analysis of faldaprevir in patients with chronic HCV genotype‐1 infection with prior relapse. Journal of Hepatology 2014;60(1 Suppl S):S458‐9. CENTRAL
Berger K, Sarrazin C, Ferenci P, Jensen DM, Jacobson IM, Stern JO, et al. NS3 Q80K did not impact efficacy or treatment‐emergent resistance patterns in HCV genotype 1‐infected patients receiving faldaprevir+peginterferon/ribavirin in three phase III trials. Journal of Hepatology 2014;60(1 Suppl S):S497. CENTRAL
Jacobson IM, Asselah T, Ferenci P, Foster GR, Jensen DM, Negro F, et al. STARTVerso3: a randomized, double‐blind, placebo‐controlled phase III trial of faldaprevir in combination with pegylated interferon a lf a‐2a and ribavirin in treatment‐experienced patients with chronic hepatitis C genotype‐1 infection. Hepatology 2013;48(S1):742A‐3A. CENTRAL
Jacobson IM, Ferenci P, Foster GR, Jensen DM, Negro F, Mantry P, et al. STARTVerso3: a randomized, double‐blind, placebo controlled phase III trial of faldaprevir in combination with pegylated interferon alpha‐2a and ribavirin in treatment experienced patients with chronic hepatitis C genotype‐1 infection. Hepatology International 2014;8(1 Suppl):S213‐4. CENTRAL

STARTverso‐3 2013a2 {published data only}

Asselah T, Zehnter E, Agarwal K, Sakai Y, Yatsuhashi H, Willems B, et al. Pharmacokinetic‐response analysis of faldaprevir in patients with chronic HCV genotype‐1 infection with prior relapse. Journal of Hepatology 2014;60(1 Suppl S):S458‐9. CENTRAL
Berger K, Sarrazin C, Ferenci P, Jensen DM, Jacobson IM, Stern JO, et al. NS3 Q80K did not impact efficacy or treatment‐emergent resistance patterns in HCV genotype 1‐infected patients receiving faldaprevir+peginterferon/ribavirin in three phase III trials. Journal of Hepatology 2014;60(1 Suppl S):S497. CENTRAL
Jacobson IM, Asselah T, Ferenci P, Foster GR, Jensen DM, Negro F, et al. STARTVerso3: a randomized, double‐blind, placebo‐controlled phase III trial of faldaprevir in combination with pegylated interferon a lf a‐2a and ribavirin in treatment‐experienced patients with chronic hepatitis C genotype‐1 infection. Hepatology 2013;48(S1):742A‐3A. CENTRAL
Jacobson IM, Ferenci P, Foster GR, Jensen DM, Negro F, Mantry P, et al. STARTVerso3: a randomized, double‐blind, placebo controlled phase III trial of faldaprevir in combination with pegylated interferon alpha‐2a and ribavirin in treatment experienced patients with chronic hepatitis C genotype‐1 infection. Hepatology International 2014;8(1 Suppl):S213‐4. CENTRAL

STARTverso‐3 2013a3 {published data only}

Asselah T, Zehnter E, Agarwal K, Sakai Y, Yatsuhashi H, Willems B, et al. Pharmacokinetic‐response analysis of faldaprevir in patients with chronic HCV genotype‐1 infection with prior relapse. Journal of Hepatology 2014;60(1 Suppl S):S458‐9. CENTRAL
Berger K, Sarrazin C, Ferenci P, Jensen DM, Jacobson IM, Stern JO, et al. NS3 Q80K did not impact efficacy or treatment‐emergent resistance patterns in HCV genotype 1‐infected patients receiving faldaprevir+peginterferon/ribavirin in three phase III trials. Journal of Hepatology 2014;60(1 Suppl S):S497. CENTRAL
Jacobson IM, Asselah T, Ferenci P, Foster GR, Jensen DM, Negro F, et al. STARTVerso3: a randomized, double‐blind, placebo‐controlled phase III trial of faldaprevir in combination with pegylated interferon a lf a‐2a and ribavirin in treatment‐experienced patients with chronic hepatitis C genotype‐1 infection. Hepatology 2013;48(S1):742A‐3A. CENTRAL
Jacobson IM, Ferenci P, Foster GR, Jensen DM, Negro F, Mantry P, et al. STARTVerso3: a randomized, double‐blind, placebo controlled phase III trial of faldaprevir in combination with pegylated interferon alpha‐2a and ribavirin in treatment experienced patients with chronic hepatitis C genotype‐1 infection. Hepatology International 2014;8(1 Suppl):S213‐4. CENTRAL

STARTverso‐4 2015 {published data only}

Dieterich D, Nelson M, Soriano V, Arasteh K, Guardiola JM, Rockstroh JK, et al. Faldaprevir and pegylated interferon alpha‐2a/ribavirin in individuals co‐infected with hepatitis C virus genotype‐1 and HIV. AIDS 2015;29(5):571‐81. [PUBMED: 25710287]CENTRAL

Sulkowski 2013a {published data only}

Sherman KE, Rockstroh JK, Dieterich DT, Soriano V, Girard PM, McCallister S, et al. Telaprevir combination with peginterferon alfa‐2A/ribavirin in HCV/HIV coinfected patients: 24‐week treatment interim analysis. Hepatology 2011;54(4 Suppl):1431A‐2A. CENTRAL
Sulkowski MS, Sherman KE, Dieterich DT, Bsharat M, Mahnke L, Rockstroh JK, et al. Combination therapy with telaprevir for chronic hepatitis C virus genotype 1 infection in patients with HIV: a randomized trial. Annals of Internal Medicine 2013;159(2):86‐96. CENTRAL
Sulkowski MS, Sherman KE, Soriano V, Rockstroh J, Dieterich DT, Girard PM, et al. Telaprevir in combination with peginterferon alfa‐2a/ribavirin in HCV/HIV co‐infected patients: SVR24 final study results. Hepatology 2012;56(S1):219A. CENTRAL

Sulkowski 2013b {published data only}

Sulkowski M, Pol S, Mallolas J, Fainboim H, Cooper C, Slim J, et al. Boceprevir versus placebo with pegylated interferon alfa‐2b and ribavirin for treatment of hepatitis C virus genotype 1 in patients with HIV: a randomised, double‐blind, controlled phase 2 trial. Lancet Infectious Diseases 2013;13(7):597‐605. CENTRAL

Sulkowski 2013c {published data only}

Kukolj G, Bethell R, Cartier M, Cote‐Martin A, Lagace L, Marguis M, et al. Characterization of HCV NS3 variants that emerged during virologic breakthrough and relapse from BI 201335 phase II SILEN‐C2 study in PEGIFN/RBV treatment‐experienced patients. Journal of Hepatology 2012;56(Suppl S2):S469. CENTRAL
Sulkowski MS, Asselah T, Ferenc P, Stern JO, Kukolj G, Boecher WO, et al. Treatment with the second generation HCV protease inhibitor BI201335 results in high and consistent SVR rates ‐ results from silen‐C1 in treatment‐naive patients across different baseline factors. Hepatology 2011;54(Suppl S1):473A. CENTRAL
Sulkowski MS, Asselah T, Lalezari J, Ferenci P, Fainboim H, Leggett B, et al. Faldaprevir combined with pegylated interferon alfa‐2a and ribavirin in treatment‐naïve patients with chronic genotype 1 HCV: SILEN‐C1 trial. Hepatology 2013;57(6):2143‐54. CENTRAL
Sulkowski MS, Ceasu E, Asselah T, Caruntu FA, Lalezari J, Ferenci P, et al. Silen‐C1: sustained virologic response (SVR) and safety of BI201335 combined with peginterferon alfa‐2a and ribavirin (P/R) in treatment‐naive patients with chronic genotype 1 HCV Infection. Journal of Hepatology 2011;54(Suppl 1):S27. CENTRAL

Sullivan 2012 {published data only}

Sullivan GJ, Rodriques‐Torres M, Lawitz E, Poordad F, Kapoor M, Campbell A, et al. ABT‐267 combined with pegylated interferon alpha‐ 2a/ribavirin in genotype 1 (GT1) HCV‐infected treatment naive subjects: 12 week antiviral and safety analysis. Journal of Hepatology 2012;56(Suppl S2):S480. CENTRAL

Tanwandee 2012 {published data only}

Tanwandee T, Luscombe C, Ewart G, Wilkinson J, Miller M, Peters MG, et al. High sustained viral response with a HCV p7 inhibitor, BIT225: antiviral activity and tolerability of BIT225 plus pegylated interferon alfa 2b and weight‐based ribavirin for 28 days in HCV treatment‐naive patients. Hepatology 2012;56(S1):1530A. CENTRAL

Tatum 2015a1 {published data only}

McPhee F, Falk P, Fracasso P, Lemm J, Liu M, Kirk M, et al. Characterization of viral escape in HCV genotype 1‐infected patients treated with BMS‐791325 and pegylated interferon‐ALFA and ribavirin. Journal of Hepatology 2012;56(Suppl S2):S473. CENTRAL
Tatum H, Thuluvath P, Lawitz E, Martorell CT, Demicco M, Cohen S, et al. A phase 2A study of BMS‐791325, an NS5B polymerase inhibitor, with peginterferon alfa‐2A and ribavirin in treatment‐naive patients with genotype 1 chronic hepatitis C infection. Journal of Hepatology 2012;56(Suppl S2):S460. CENTRAL
Tatum H, Thuluvath PJ, Lawitz E, Martorell C, Demicco M, Cohen S, et al. A randomized, placebo‐controlled study of the NS5B inhibitor beclabuvir with peginterferon/ribavirin for HCV genotype 1. Journal of Viral Hepatitis 2015;22(8):658‐64. CENTRAL
Tatum HA, Thuluvath PJ, Lawitz E, Martorell C, Cohen SM, Rustgi VK, et al. Safety and efficacy of BMS‐791325, a non‐nucleoside NS5B polymerase inhibitor, combined with peginterferon alfa‐2a and ribavirin in treatment‐naive patients infected with hepatitis C virus genotype 1. Hepatology 2013;58(S1):759A‐60A. CENTRAL

Tatum 2015a2 {published data only}

McPhee F, Falk P, Fracasso P, Lemm J, Liu M, Kirk M, et al. Characterization of viral escape in HCV genotype 1‐infected patients treated with BMS‐791325 and pegylated interferon‐ALFA and ribavirin. Journal of Hepatology 2012;56(Suppl S2):S473. CENTRAL
Tatum H, Thuluvath P, Lawitz E, Martorell CT, Demicco M, Cohen S, et al. A phase 2A study of BMS‐791325, an NS5B polymerase inhibitor, with peginterferon alfa‐2A and ribavirin in treatment‐naive patients with genotype 1 chronic hepatitis C infection. Journal of Hepatology 2012;56(Suppl S2):S460. CENTRAL
Tatum H, Thuluvath PJ, Lawitz E, Martorell C, Demicco M, Cohen S, et al. A randomized, placebo‐controlled study of the NS5B inhibitor beclabuvir with peginterferon/ribavirin for HCV genotype 1. Journal of Viral Hepatitis 2015;22(8):658‐64. CENTRAL
Tatum HA, Thuluvath PJ, Lawitz E, Martorell C, Cohen SM, Rustgi VK, et al. Safety and efficacy of BMS‐791325, a non‐nucleoside NS5B polymerase inhibitor, combined with peginterferon alfa‐2a and ribavirin in treatment‐naive patients infected with hepatitis C virus genotype 1. Hepatology 2013;58(S1):759A‐60A. CENTRAL

Vierling 2011 {published data only}

Vierling JM, Poordad FF, Lawitz E, Ghalib RH, Lee WM, Ravendhran N, et al. Once daily narlaprevir (NVR; SCH 900518) and ritonavir (RTV) in combination with peginterferon alfa‐2B/ribavirin (PR) for 12 weeks plus 12 weeks pr in treatment‐naïve patients with HCV genotype 1 (G1): SVR results from next‐1, a phase 2 study. Hepatology 2011;54(Suppl S1):1437A‐8A. CENTRAL

Villano 2007 {published data only}

Villano S, Raible D, Harper D, Chandra P, Bazisotto L, Bichier G. Phase 1 evaluation of antiviral activity of the non‐nucleoside polymerase inhibitor, HCV‐796, in combination with different pegylated interferons in treatment‐naive patients with chronic HCV. Hepatology 2007;46(4 Suppl 1):815A. CENTRAL

Vince 2014 {published data only}

Vince B, Hill JM, Lawitz EJ, O'Riordan W, Webster LR, Gruener DM, et al. A randomized, double‐blind, multiple‐dose study of the pan‐genotypic NS5A inhibitor samatasvir in patients infected with hepatitis C virus genotype 1, 2, 3 or 4. Journal of Hepatology 2014;60(5):920‐7. CENTRAL

Wedemeyer 2013 {published data only}

Chen YC, Moreira S, Ipe D, Kulkarni R, Passe S, Zhu Y, et al. The effect of host il28b genotype and mericitabine (MCB; RG7128) dose on early viral kinetics following administration of MCB plus pegifn alpha‐2a plus ribavirin (P/R) in patients with chronic hepatitis C (CHC). Hepatology 2011;54(Suppl S1):553A. CENTRAL
Jensen DM, Wedemeyer H, Herring RW, Ferenci P, Ma MM, Zeuzem S, et al. High rates of early viral response, promising safety profile and lack of resistance‐related breakthrough in HCV GT 1/4 patients treated with RG7128 plus PegIFN alfa‐2a (40KD)/RBV: planned week 12 interim analysis from the PROPEL study. Hepatology 2010;52(Suppl S1):360A. CENTRAL
Wedemeyer H, Jensen D, Herring R, Ferenci P, Ma MM, Zeuzem S, et al. Efficacy and safety of mericitabine (MCB) in combination with Peg‐Ifn alpha‐2A/RBV in G1/4 treatment naive HCV patients: final analysis from the PROPEL study. Journal of Hepatology 2012;56(Suppl S2):S481‐2. CENTRAL
Wedemeyer H, Jensen D, Herring R, Ferenci P, Ma MM, Zeuzem S, et al. PROPEL: a randomized trial of mericitabine plus peginterferon alpha‐2a/ribavirin therapy in treatment‐naive HCV genotype 1/4 patients. Hepatology 2013;58(2):524‐37. CENTRAL

Wilfret 2013 {published data only}

Spreen W, Wilfret DA, Bechtel J, Adkison KK, Lou Y, Jones L, et al. GSK2336805 HCV NS5A inhibitor demonstrates potent antiviral activity in chronic genotype 1 infection; results from a first time in human (FTIH) single and repeat dose study. Hepatology 2011;54(Suppl S1):400A‐1A. CENTRAL
Wilfret DA, Walker J, Adkinson KK, Jones LA, Lou Y, Gan J, et al. Safety, tolerability, pharmacokinetics, and antiviral activity OFGSK2336805, an inhibitor of hepatitis C virus (HCV) NA5A, in healthy subjects and subjects chronically infected with HCV genotype 1. Antimicrobial Agents and Chemotherapy 2013;57(10):5037‐44. CENTRAL

Younossi 2015 {published data only}

Younossi Z, Stepanova M, Pol S, Bronowicki JP, Carrieri P, Bourliere M. The impact of ledipasvir (LDV)/sofosbuvir (SOF) combination on health‐related quality of life (HRQL) and patient‐reported outcomes (PROS) in cirrhotic patients with chronic hepatitis C (CH‐C): the SIRIUS study. Journal of Hepatology 2015;62(Suppl S2):S591. CENTRAL

Zeuzem 2011a {published data only}

Berg T, Andreone P, Pol S, Roberts SK, Younossi ZM, Diago M, et al. Predictors of virologic response with telaprevir‐based combination treatment in HCV genotype 1‐infected patients with prior peginterferon/ribavirin treatment failure: post‐hoc analysis of the phase III REALIZE study. Hepatology 2011;54(S1):375A‐6A. CENTRAL
De Meyer S, Dierynck I, Ghys A, Beumont M, Daems B, Van Baelen B, et al. Characterization of telaprevir treatment outcomes and resistance in patients with prior treatment failure: results from the REALIZE trial. Hepatology 2012;56(6):2106‐15. CENTRAL
Foster GR, Jacobson IM, Dore GJ, Fried M, Manns M, Marcellin P, et al. Simeprevir (TMC435) with peginterferon/ribavirin for treatment of chronic HCV genotype 1 infection in treatment‐naive European patients in the QUEST‐1 and QUEST‐2 phase III trials. Journal of Hepatology 2014;60(1 Suppl 1):S456. CENTRAL
Foster GR, Zeuzem S, Andreone P, Pol S, Lawitz EJ, Diago M, et al. Subanalyses of the telaprevir lead‐in arm in the REALIZE study: response at week 4 is not a substitute for prior null response categorization. Journal of Hepatology 2011;54(Suppl 1):S3‐S4. CENTRAL
Foster GR, Zeuzem S, Andreone P, Pol S, Lawitz EJ, Diago M, et al. Sustained virologic response rates with telaprevir by response after 4 weeks of lead‐in therapy in patients with prior treatment failure. Journal of Hepatology 2013;58(3):488‐94. CENTRAL
Pol S, Aerssens J, Zeuzem S, Andreone P, Lawitz EJ, Roberts S, et al. Similar SVR rates in IL28B CC, CT OR TT prior relapser, partial‐ or null‐responder patients treated with telaprevir/peginterferon/ribavirin: retrospective analysis of the REALIZE study. Journal of Hepatology 2011;54(Suppl 1):S6‐7. CENTRAL
Pol S, Roberts SK, Andreone P, Younossi ZM, Diago M, Lawitz E, et al. Efficacy and safety of telaprevir‐based regimens in cirrhotic patients with HCV genotype 1 and prior peginterferon/ribavirin treatment failure: subanalysis of the REALIZE phase III study. Hepatology 2011;54(S1):374A‐5A. CENTRAL
Roberts SK, Andreone P, Pol S, Younossi ZM, Diago M, Lawitz E, et al. Impact of anemia and ribavirin dose reduction on SVR to a telaprevir‐based regimen in patients with HCV genotype 1 and prior peginterferon/ribavirin treatment failure in the phase III REALIZE study. Hepatology 2011;54(4 Suppl):1007A‐8A. CENTRAL
Tuefferd M, Palescandolo E, Vijgen L, Colombo M, Roberts S, Wedemeyer H, et al. ITPA gene variants and anemia during telaprevir/peginterferon/ribavirin combination therapy in patients with chronic hepatitis C infection. Journal of Hepatology 2014;60(1 Suppl S):S490. CENTRAL
Witek J. Risk factors predictive of anemia development during telaprevir plus peginterferon/ribavirin therapy in treatment‐experienced patients. Journal of Hepatology 2014;60(6):1112‐7. CENTRAL
Younossi Z, Negro F, Serfaty L, Pol S, Diago M, Zeuzem S, et al. Homeostasis model assessment of insulin resistance does not seem to predict response to telaprevir in chronic hepatitis C in the REALIZE trial. Hepatology 2013;58(6):1897‐906. CENTRAL
Zeuzem S, Andreone P, Pol S, Lawitz E, Diago M, Roberts S, et al. Telaprevir for retreatment of HCV infection. New England Journal of Medicine 2011;364(25):2417‐28. CENTRAL
Zeuzem S, Andreone P, Pol S, Lawitz EJ, Diago M, Roberts S, et al. REALIZE trial final results: telaprevir‐based regimen for genotype 1 hepatitis C virus infection in patients with prior null response, partial response or relapse to peginterferon/ribavirin. Journal of Hepatology 2011;54(Suppl S1):S3. CENTRAL

Zeuzem 2014a {published data only}

Liu Y, Larsen L, Bourliere M, Baran R, Juday T. Ombitasvir/paritaprevir/ritonavir and dasabuvir with ribavirin (RBV) has mild impact on health‐related quality of life (HRQoL) compared with placebo during 12‐week treatment in treatment‐experienced adults with chronic hepatitis C (CHC). Journal of Hepatology 2015;62(Suppl S2):S661. CENTRAL
Zeuzem S, Jacobson IM, Baykal T, Marinho RT, Poordad F, Bourliere M, et al. Retreatment of HCV with ABT‐450/r‐ombitasvir and dasabuvir with ribavirin. New England Journal of Medicine 2014;370(17):1604‐14. CENTRAL

References to studies excluded from this review

AGATE‐I 2015 {published data only}

Asselah T, Hassanein TI, Qaqish RB, Feld JJ, Hezode C, Zeuzem S, et al. Efficacy and safety of ombitasvir/paritaprevir/ritonavir co‐administered with ribavirin in adults with genotype 4 chronic hepatitis C infection and cirrhosis (AGATE‐I). Hepatology 2015;62(Suppl S1):563A‐4A. CENTRAL

ALLY 2015 {published data only}

Wyles D, Ruane P, Sulkowski M, Dieterich D, Luetkemeyer A, Morgan T, et al. Daclatasvir in combination with sofosbuvir for HIV/HCV coinfection: ALLY‐2 study. Topics in Antiviral Medicine2015; Vol. 23, issue e‐1:62. CENTRAL

ANNAPURNA 2013 {published data only}

Jensen DM, Brunda M, Elston R, Gane EJ, George J, Glavini K, et al. Interferon‐free regimen containing setrobuvir (STV) in combination with ritonavir‐boosted danoprevir (DNVr) and ribavirin (R) with or without mericitabine (MCB) in HCV genotype (G)1 treatment‐naive patients: interim SVR4 results from the ANNAPURNA study. Hepatology 2013;58(S1):741A. CENTRAL

APRICOT 2004 {published data only}

APRICOT Study Group. APRICOT Study evaluated. Surprisingly high virologic response in HIV‐HCV coinfection. MMW Fortschritte der Medizin 2004;146(Spec No 1):74‐5. CENTRAL

ATOMIC 2013 {published data only}

Kowdley KV, Lawitz E, Crespo I, Hassanein T, Davis MN, Demicco M, et al. Sofosbuvir with pegylated interferon alfa‐2a and ribavirin for treatment‐naive patients with hepatitis C genotype‐1 infection (ATOMIC): an open‐label, randomised, multicentre phase 2 trial. Lancet 2013;381(9883):2100‐7. CENTRAL

ATTAIN 2015 {published data only}

Akpo EIH, Sbarigia U, Wan G, Kleintjens J. Determinant factors of the direct medical costs associated with genotype 1 hepatitis C infection in treatment‐experienced patients. Drugs in R&D 2015;15(4):335‐49. CENTRAL
Reddy KR, Zeuzem S, Zoulim F, Weiland O, Horban A, Stanciu C. A phase III randomised, double‐blind study to evaluate the efficacy, safety and tolerability of simeprevir vs telaprevir in combination with pegylated interferon and ribavirin in chronic hepatitis C virus genotype 1 treatment‐experienced patients: the ATTAIN study. Hepatology International 2014;8(Suppl 1):S397. CENTRAL
Reddy KR, Zeuzem S, Zoulim F, Weiland O, Horban A, Stanciu C, et al. Simeprevir versus telaprevir with peginterferon and ribavirin in previous null or partial responders with chronic hepatitis C virus genotype 1 infection (ATTAIN): A randomised, double‐blind, non‐inferiority phase 3 trial. Lancet Infectious Diseases 2015;15(1):27‐35. CENTRAL
Scott J, Cerri K, Sbarigia U, Corbett C, Fu M, Jessner W. Simeprevir with PegIFN/ribavirin for chronic HCV infection shortens time with patient reported symptoms and impairment in QoL: ATTAIN study results. Journal of Viral Hepatitis 2014;21(Suppl S2):25. CENTRAL
Scott J, Corbett C, Gilles L, Wan G, Sbarigia U, Jessner W. Impact of simeprevir versus telaprevir triple therapy for chronic HCV infection on patient‐reported outcomes in prior non‐responders to peginterferon/ribavirin results from the phase III attain study. Value in Health 2014;17(7):A682. CENTRAL

AVIATOR 2015 {published data only}

Poordad F, Agarwal K, Younes Z, Cohen D, Xie WG, Podsadecki T. Low relapse rate leads to high concordance of sustained virologic response (SVR) at 12 weeks with SVR at 24 weeks after treatment with abt‐450/ritonavir, ombitasvir, and dasabuvir plus ribavirin in subjects with chronic hepatitis C virus genotype 1 infection in the aviator study. Clinical Infectious Diseases 2015;60(4):608‐10. CENTRAL
Szeinbach SL, Baran RW. Treatment satisfaction in clinical trial (AVIATOR) patients treated with interferon‐free, oral DAA regimens: external validation of the hepatitis C virus (HCV) treatment satisfaction (HCVTSat) instrument. Gastroenterology 2014;146(5 Suppl 1):S‐977. CENTRAL

Basu 2014b {published data only}

Basu P, Shah N, Aloysius M. Simeprevir and sofosbuvir with modified doses of ribavirin (RBV) therapy on telaprevir‐experienced, coinfected (with HIV) cirrhotics with chronic hepatitis C (CHC): a randomized, open‐label, clinical pilot study, STOP C, interim results. American Journal of Gastroenterology 2014;109(Suppl 2):S177. CENTRAL

Bathgate 2011 {published data only}

Bathgate A. Boceprevir for previously treated chronic hepatitis C virus genotype I infection. Journal of the Royal College of Physicians of Edinburgh 2011;41(2):122‐3. CENTRAL

Bognar 2011 {published data only}

Bognar F. Projecting the clinical impact of therapeutic regimens including boceprevir in previously untreated adult subjects with chronic hepatitis C genotype 1. Journal of Gastroenterology and Hepatology 2011;26(Suppl 5):19. CENTRAL

Bourgeois 2015 {published data only}

Bourgeois S, Nevens F, Moreno C, Van Vlierberghe H, Arasteh K, Horsmans Y, et al. Efficacy safety and pharmacokinetics of 12 weeks of simeprevir in combination with TMC647055 ritonavir and JNJ 56914845 in genotype 1 hepatitis C virus infected patients. Hepatology International 2015;9(1 Suppl):S57‐8. CENTRAL

Chandra 2006b {published data only}

Chandra P, Raible D, Moyer L, Harper D, Speth J, Villano S, et al. Safety and pharmacokinetics of the non‐nucleoside polymerase inhibitor, HCV‐796: results of a randomized, double‐blind, placebo‐controlled, ascending single‐dose study in healthy subjects. Journal of Hepatology 2006;44(2 Suppl):S208‐9. CENTRAL

CONCISE 2013 {published data only}

Nelson DR, Poordad F, Feld JJ, Fried MW, Jacobson IM, Pockros PJ, et al. High SVR rates (SVR4) for 12‐week total telaprevir combination therapy in IL28B CC treatment‐naives and prior relapsers with G1 chronic hepatitis C: concise interim analysis. Journal of Hepatology 2013;58(Suppl S1):S362. CENTRAL

COSMOS 2014 {published data only}

Lawitz E, Sulkowski MS, Ghalib R, Rodriguez‐Torres M, Younossi ZM, Corregidor A, et al. Simeprevir plus sofosbuvir, with or without ribavirin, to treat chronic infection with hepatitis C virus genotype 1 in non‐responders to pegylated interferon and ribavirin and treatment‐naive patients: the COSMOS randomised study. Lancet 2014;384(9956):1756‐65. [PUBMED: 25078309]CENTRAL

C‐SURFER 2015 {published data only}

Elbasha EH, Ferrante S, Agarwal E, Greaves W, Nwankwo C. Projected clinical and economic impact of grazoprevir (GZR, MK‐5172)/elbasvir (EBR, MK‐8742) for chronic HCV genotype 1 infection in chronic kidney disease. Value in Health 2015;18(7):A510. CENTRAL
Roth D, Nelson D, Bruchfeld A, Liapakis A, Silva M, Monsour Jr H, et al. C‐surfer: grazoprevir plus elbasvir in treatment‐naive and treatment‐experienced patients with hepatitis C virus genotype 1 infection and chronic kidney disease. Journal of Hepatology 2015;62(Suppl S2):S263‐4. CENTRAL
Roth D, Nelson DR, Bruchfeld A, Liapakis A, Silva M, Monsour H, et al. Grazoprevir plus elbasvir in treatment‐naive and treatment‐experienced patients with hepatitis C virus genotype 1 infection and stage 4‐5 chronic kidney disease (the C‐SURFER study): a combination phase 3 study. Lancet 2015;386(10003):1537‐45. CENTRAL

C‐WORTHY 2015 {published data only}

Lawitz E, Gane E, Pearlman B, Tam E, Ghesquiere W, Guyader D, et al. Efficacy and safety of 12 weeks versus 18 weeks of treatment with grazoprevir (MK‐5172) and elbasvir (MK‐8742) with or without ribavirin for hepatitis C virus genotype 1 infection in previously untreated patients with cirrhosis and patients with previous null response with or without cirrhosis (C‐WORTHY): a randomised, open‐label phase 2 trial. Lancet 2015;385(9973):1075‐86. CENTRAL
Lawitz E, Vierling JM, Murillo A, Kugelmas M, Gerstoft J, Winkle P, et al. High efficacy and safety of the all‐oral combination regimen, MK‐5172/MK‐8742 +/‐ RBV for 12 weeks in HCV genotype 1 infected patients: the C‐WORTHY study. Hepatology 2013;48(S1):244A‐5A. CENTRAL
Sulkowski M, Hezode C, Gerstoft J, Vierling JM, Mallolas J, Pol S, et al. Efficacy and safety of 8 weeks versus 12 weeks of treatment with grazoprevir (MK‐5172) and elbasvir (MK‐8742) with or without ribavirin in patients with hepatitis C virus genotype 1 mono‐infection and HIV/hepatitis C virus co‐infection (C‐WORTHY): a randomised, open‐label phase 2 trial. Lancet 2015;385(9973):1087‐97. CENTRAL
Sulkowski M, Mallolas J, Bourliere M, Gerstoft J, Shibolet O, Nahass R, et al. On‐treatment viral response to MK‐5172/MK‐8742 +/‐ RBV for 12 weeks in HCV/HIV‐coinfected patients. Topics in Antiviral Medicine2014; Vol. 22, issue e‐1:324‐5. CENTRAL

Di Bisceglie 2014 {published data only}

Di Bisceglie AM, Sulkowski M, Gane E, Jacobson IM, Nelson D, DeSouza C, et al. VX‐222, a non‐nucleoside NS5B polymerase inhibitor, in telaprevir‐based regimens for genotype 1 hepatitis C virus infection. European Journal of Gastroenterology & Hepatology 2014;26(7):761‐73. CENTRAL

Dore 2014 {published data only}

Dore GJ, Jacobson IM, Foster GR, Fried M, Manns M, Marcellin P, et al. Simeprevir (TMC435) with peg‐interferon/ribavirin for treatment of chronic HCV genotype 1 infection in treatment naive patients: efficacy in patients with genotype 1b HCV in the QUEST‐1 and‐2 phase III trials. Hepatology International 2014;8(1 Suppl 1):S195. CENTRAL

Dusheiko 2015 {published data only}

Dusheiko GM, Manns MP, Vierling JM, Reddy KR, Sulkowski MS, Kwo PY, et al. Safety and tolerability of grazoprevir/elbasvir in patients with chronic hepatitis C (HCV) infection: integrated analysis of phase 2‐3 trials. Hepatology 2015;62(Suppl S1):562A. CENTRAL

Ferenci 2014 {published data only}

Ferenci P, Bernstein D, Lalezari J, Cohen D, Luo Y, Cooper C, et al. ABT‐450/r‐ombitasvir and dasabuvir with or without ribavirin for HCV. New England Journal of Medicine 2014;370(21):1983‐92. CENTRAL

Ferrante 2011a {published data only}

Ferrante SA, Chhatwal J, Dasbach EJ, Sniukiene V, Poordad F, Bronowicki JP. Projecting the clinical impact of therapeutic regimens including boceprevir in previously untreated adult subjects with chronic hepatitis C genotype 1. Gastroenterology 2011;140(5 Suppl 1):S909. CENTRAL

Ferrante 2011b {published data only}

Ferrante SA, Chhatwal J, Elbasha E, Dasbach EJ, Khoury A, Poordad F, et al. Cost‐effectiveness of boceprevir based regimens in previously untreated adult subjects with chronic hepatitis C genotype 1. Hepatology 2011;54(4 Suppl):795A‐796A. CENTRAL

Ferrante 2013 {published data only}

Ferrante SA, Chhatwal J, Brass CA, El Khoury AC, Poordad F, Bronowicki JP, et al. Boceprevir for previously untreated patients with chronic hepatitis C Genotype 1 infection: a US‐based cost‐effectiveness modelling study. BMC Infectious Diseases 2013;13:190. CENTRAL

Foster 2010 {published data only}

Foster GF, Dusheiko G, Hezode C, Marcellin P, McHutchinson J, Beumont M, et al. Final results from telaprevir phase II studies in genotype 1 treatment‐naive or experienced subjects with chronic hepatitis C. Gut 2010;59(Suppl 2):A5. CENTRAL

FOURward 2014 {published data only}

Sulkowski MS, Flamm SL, Kayali Z, Lawitz E, Kwo PY, McPhee F, et al. Short‐duration therapy with daclatasvir/asunaprevir/beclabuvir fixed‐dose combination plus sofosbuvir in patients with chronic hepatitis C genotype 1 (FOURward study). Hepatology 2015;62(Suppl S1):556A. CENTRAL

FUSION 2013 {published data only}

Jacobson IM, Gordon SC, Kowdley KV, Yoshida EM, Rodriguez‐Torres M, Sulkowski MS, et al. Sofosbuvir for hepatitis C genotype 2 or 3 in patients without treatment options. New England Journal of Medicine 2013;368(20):1867‐77. CENTRAL

Gardner 2014b {published data only}

Gardner S, Cutrell AMY, Elko‐Simms C, Adkison K, Hamatake R, Walker J, et al. A double‐blind, randomized, placebo‐controlled study to assess the safety, antiviral activity and pharmacokinetics of GSK2336805 when given as monotherapy and in combination with peginterferon alfa‐2a and ribavirin in hepatitis C virus genotype 1‐infected treatment‐naive subjects. Liver International 2014;34(6):e89‐95. CENTRAL

HCVerso 1 2014 {published data only}

Sarrazin C, Castelli F, Puoti M, Shiffman ML, Preotescu L, Forns X, et al. HCVerso1: a phase III study of faldaprevir (FDV) plus deleobuvir (DBV) and ribavirin (RBV) for chronic HCV genotype (GT)‐1b infection in treatment‐naive patients. Hepatology 2014;59(Suppl S1):1150A. [DOI: http://dx.doi.org/10.1002/hep.27533]CENTRAL

HCVerso 2 2014 {published data only}

Nelson DR, Andreone P, Colombo M, Calinas F, Olveira A, Delwaide J, et al. HCVerso2: a phase III study of faldaprevir (FDV) plus deleobuvir (DBV) and ribavirin (RBV) for chronic HCV genotype (GT)‐1b infection in treatment‐naive patients including those ineligible for pegylated interferon (PegIFN). Hepatology 2014;59(Suppl S1):1155A‐6A. [DOI: http://dx.doi.org/10.1002/hep.27533]CENTRAL

ION‐3 2014 {published data only}

Kowdley KV, Gordon SC, Reddy KR, Rossaro L, Bernstein DE, Lawitz E, et al. Ledipasvir and sofosbuvir for 8 or 12 weeks for chronic HCV without cirrhosis. New England Journal of Medicine 2014;370(20):1879‐88. CENTRAL

Jacobson 2013 {published data only}

Jacobson IM, Dore GJ, Foster GR, Fried MW, Manns MP, Marcellin P, et al. Simeprevir (TMC435) with peginterferon/ribavirin for treatment of chronic HCV genotype 1 infection in treatment‐naïve patients: efficacy in difficult‐to‐treat patient sub‐populations in the QUEST 1 and 2 phase III trials. Hepatology 2013;58(S1):756A‐7A. CENTRAL

Kawada 2015 {published data only}

Kawada N, Suzuki F, Karino Y, Chayama K, Itoh Y, Okanoue T, et al. Efficacy, safety and pharmacokinetics of grazoprevir (MK‐5172) and elbasvir (MK‐8742) In hepatitis C genotype 1 infected non‐cirrhotic Japanese patients (phase 2 portion in phase 2/3 combined study). Hepatology 2015;62(Suppl S1):559A. CENTRAL

Liu 2015b {published data only}

Liu Y, Bernstein D, Larsen L, Luo Y, Xie Y, Juday T. Ribavirin does not impact health‐related quality of life (HRQoL) in patients on ombitasvir/paritaprevir/ritonavir and dasabuvir at the end of 12‐week treatment in treatment‐naive adults with genotype 1a (GT1a) chronic hepatitis C. Value in Health 2015;18(3):A227. CENTRAL

Lok 2010 {published data only}

Lok AS, Gardiner DF, Lawitz E, Martorell C, Everson GT, Ghalib RH, et al. Combination therapy with BMS‐790052 and BMS‐650032 alone or with pegIFN/RBV results in undetectable HCV RNA through 12 weeks of therapy in HCV genotype 1 null responders. Hepatology 2010;52(Suppl S1):877A. CENTRAL

Lok 2011 {published data only}

Lok A, Gardiner D, Lawitz E, Martorell C, Everson G, Ghalib R, et al. Quadruple therapy with BMS‐790052, BMS‐650032 and PEG‐IFN/RBV for 24 weeks results in 100% SVR12 in HCV genotype 1 null responders. Journal of Hepatology 2011;54(Suppl 1):S536. CENTRAL

Lok 2012a {published data only}

Lok A, Gardiner D, Hezode C, Lawitz E, Bourliere M, Everson G, et al. Confirmation that quadruple therapy with daclatasvir (NS5A inhibitor), asunaprevir (NS3 inhibitor) and peginterferon/ribavirin results in high rate of svr4 in HCV genotype 1 null responders. Journal of Hepatology 2012;56(Suppl 2):S557. CENTRAL

Lok 2012b {published data only}

Lok AS, Gardiner DF, Hezode C, Lawitz E, Bourliere M, Everson GT, et al. Sustained virologic response in chronic HCV genotype (GT) 1‐infected null responders with combination of daclatasvir (DCV; NS5A Inhibitor) and asunaprevir (ASV; NS3 Inhibitor) with or without peginterferon alfa‐2a/ribavirin (PEG/RBV). Hepatology 2012;56(S1):230A‐1A. CENTRAL

Lok 2014 {published data only}

Lok AS, Gardiner DF, Hezode C, Lawitz EJ, Bourliere M, Everson GT, et al. Randomized trial of daclatasvir and asunaprevir with or without PegIFN/RBV for hepatitis C virus genotype 1 null responders. Journal of Hepatology 2014;60(3):490‐9. CENTRAL

MALACHITE‐I 2016 {published data only}

Conway B, Janczewska EWA, Luo YAN, Curescu M, Greenbloom S, Streinu‐Cercel A, et al. Malachite‐I: phase 3b trial of ombitasvir/paritaprevir/r and dasabuvir plus /‐ ribavirin or telaprevir plus peginterferon/ribavirin in treatment‐naive adults with HCV genotype 1. Gastroenterology 2015;148(4 Suppl 1):S1001‐S2. CENTRAL
Dore GJ, Conway B, Luo YAN, Janczewska EWA, Knysz B, Liu YAN, et al. Efficacy and safety of ombitasvir/paritaprevir/r and dasabuvir compared to IFN‐containing regimens in genotype 1 HCV patients: the malachite‐I/II trials. Journal of Hepatology 2016;64(1):19‐28. CENTRAL
Liu Y, Luo Y, Liu X, Sullivan D, Juday TR, Conway B, et al. Better work productivity and activity in patients on ombitasvir/ paritaprevir/ritonavir and dasabuvir with or without ribavirin (3D+RBV or 3D) in treatment‐naive adults with genotype 1 (GT1) chronic hepatitis C. Hepatology 2015;62(Suppl S1):802A. CENTRAL

MALACHITE‐II 2016 {published data only}

Dore G, Knysz B, Luo Y, Janczewska E, Streinu‐Cercel A, Caruntu FA, et al. Malachite‐II: phase 3B trial of ombitasvir/paritaprevir/R and dasabuvir + ribavirin or telaprevir + peginterferon/ribavirin in peginterferon/ribavirin treatment‐experienced adults with HCV genotype 1. Journal of Hepatology 2015;62(Suppl S2):S656‐7. CENTRAL
Dore GJ, Conway B, Luo YAN, Janczewska EWA, Knysz B, Liu YAN, et al. Efficacy and safety of ombitasvir/paritaprevir/r and dasabuvir compared to IFN‐containing regimens in genotype 1 HCV patients: the malachite‐I/II trials. Journal of Hepatology 2016;64(1):19‐28. CENTRAL

Manns 2014b {published data only}

Manns MP, McCone J, Davis MN, Rossaro L, Schiff E, et al. Overall safety profile of boceprevir plus peginterferon alfa‐2b and ribavirin in patients with chronic hepatitis C genotype 1: a combined analysis of 3 phase 2/3 clinical trials. Liver International 2014;34(5):707‐19. CENTRAL

Manns 2015 {published data only}

Manns M, Forns X, Samuel D, Denning J, Arterburn S, Brandt‐Sarif T, et al. Ledipasvir/sofosbuvir with ribavirin is safe and efficacious in decompensated and post liver transplantation patients with HCV infection: preliminary results of the prospective SOLAR 2 trial. Journal of Hepatology 2015;62(Suppl S2):S187‐8. CENTRAL

Mendez 2014 {published data only}

Mendez P, Suzuki Y, Ikeda K, Toyota J, Karino Y, Kawakami Y, et al. Safety of asunaprevir (ASV) in the all‐oral dual combination with daclatasvir (DCV) or in combination with peginterferon/ribavirin (pegIFN/RBV) for chronic HCV infection. Hepatology International 2014;59(Suppl S1):S186‐7. CENTRAL

Mizokami 2015 {published data only}

Mizokami M, Yokosuka O, Takehara T, Sakamoto N, Korenaga M, Mochizuki H, et al. Ledipasvir and sofosbuvir fixed‐dose combination with and without ribavirin for 12 weeks in treatment‐naive and previously treated Japanese patients with genotype 1 hepatitis C: an open‐label, randomised, phase 3 trial. Lancet Infectious Diseases 2015;15(6):645‐53. CENTRAL

Molina 2015 {published data only}

Molina JM, Orkin C, Iser DM, Zamora FX, Nelson M, Stephan C, et al. Sofosbuvir plus ribavirin for treatment of hepatitis C virus in patients co‐infected with HIV (PHOTON‐2): a multicentre, open‐label, non‐randomised, phase 3 study. Lancet 2015;385(9973):1098‐106. CENTRAL

Muir 2011 {published data only}

Muir AJ, Poordad FF, McHutchison JG, Shiffman ML, Berg T, Ferenci P, et al. Retreatment with telaprevir combination therapy in hepatitis C patients with well‐characterized prior treatment response. Hepatology 2011;54(5):1538‐46. CENTRAL

Muir 2015 {published data only}

Muir A, Poordad F, Lalezari J, Dore G, Hezode C, Ramji A, et al. Unity‐2: daclatasvir/asunaprevir/beclabuvir +/‐ RBV for HCV genotype 1 with cirrhosis. Topics in Antiviral Medicine 2015;23(e‐1):301. CENTRAL
Muir A, Poordad F, Lalezari J, Everson G, Dore G, Kwo P, et al. Fixed dose oral combination therapy with daclatasvir asunaprevir beclabuvir‐ribavirin for patients with chronic HCV genotype 1 infection and compensated cirrhosis unity 2 phase 3 results. Hepatology International 2015;9(1 Suppl 1):S47. CENTRAL
Muir AJ, Poordad F, Lalezari J, Everson G, Dore GJ, Herring R, et al. Daclatasvir in combination with asunaprevir and beclabuvir for hepatitis C virus genotype 1 infection with compensated cirrhosis. JAMA 2015;313(17):1736‐44. CENTRAL

NEUTRINO 2013 {published data only}

Lawitz E, Mangia A, Wyles D, Rodriguez‐Torres M, Hassanein T, Gordon Stuart C, et al. Sofosbuvir for previously untreated chronic hepatitis C infection. New England Journal of Medicine 2013;368(20):1878‐87. CENTRAL

Nishiguchi 2014b {published data only}

Nishiguchi S, Sakai Y, Kuboki M, Tsunematsu S, Urano Y, Sakamoto W, et al. Safety and efficacy of faldaprevir with pegylated interferon alfa‐2a and ribavirin in Japanese patients with chronic genotype‐1 hepatitis C infection. Liver International 2014;34(1):78‐88. CENTRAL

Nomura 2014 {published data only}

Nomura H, Miyagi Y, Tanimoto H, Kawano A, Yamashita N. Weight loss during telaprevir‐based triple therapy due to telaprevir‐induced appetite loss. Internal Medicine 2014;53(22):2567‐73. CENTRAL

NUCLEAR 2013 {published data only}

Lawitz EJ, Rodriguez‐Torres M, Denning J, Mathias A, Mo H, Gao B, et al. All‐oral therapy with nucleotide inhibitors sofosbuvir and GS‐0938 for 14 days in treatment‐naive genotype 1 hepatitis C (nuclear). Journal of Viral Hepatitis 2013;20(10):699‐707. CENTRAL

OPTIMIST‐1 2015 {published data only}

Kwo P, Gitlin N, Nahass R, Bernstein D, Rojter S, Schiff E, et al. A phase 3, randomised, open‐label study to evaluate the efficacy and safety of 8 and 12 weeks of simeprevir (SMV) plus sofosbuvir (SOF) in treatment‐naive and ‐experienced patients with chronic HCV genotype 1 infection without cirrhosis: OPTIMIST‐1. Journal of Hepatology 2015;62(Suppl S2):S270. CENTRAL

OPTIMIZE 2013 {published data only}

Buti M, Agarwal K, Horsmans Y, Sievert W, Janczewska E, Zeuzem S, et al. Telaprevir twice daily is noninferior to telaprevir every 8 hours for patients with chronic hepatitis C. Gastroenterology 2013;146(3):744‐53. [DOI: 10.1053/j.gastro.2013.11.047]CENTRAL
Buti M, Agarwal K, Horsmans Y, Sievert W, Zeuzem S, Nyberg L, et al. Efficacy of telaprevir dosed twice daily versus every 8 hours by IL28B genotype: results from phase III OPTIMIZE study. Journal of Hepatology 2013;58(Suppl1):S326. CENTRAL

Poordad 2014 {published data only}

Forns X, Poordad F, Pedrosa M, Berenguer M, Wedemeyer H, Ferenci P, et al. Ombitasvir/paritaprevir/r, dasabuvir and ribavirin for cirrhotic HCV patients with thrombocytopaenia and hypoalbuminaemia. Liver International 2015;35(11):2358‐62. CENTRAL
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. CENTRAL
Wedwemeyer H, Berg T, Flamm SL, Foster GR, Craxi A, Larrey D, et al. Improvement in liver function and non‐invasive estimates of liver fibrosis 48 weeks after treatment with ombitasvir/paritaprevir/r, dasabuvir and ribavirin in HCV genotype 1 patients with cirrhosis. Journal of Hepatology 2015;62(Suppl S2):S637‐8. CENTRAL

Proulx 2008 {published data only}

Proulx L, Bourgault B, Chauret N, Larouche R, Tanguay M, Thibert R. Results of a safety, tolerability and pharmacokinetic phase I study of VCH‐916, a novel polymerase inhibitor for HCV, following single ascending doses in healthy volunteers. Journal of Hepatology 2008;48(Suppl S2):S320. CENTRAL

Reddy 2011 {published data only}

Reddy KR, Nunes F, Balart LA, Sjogren R, Pedicone L, Burroughs M, et al. An evaluation of neutropenia in the pivotal studies of boceprevir (BOC) plus PegInterferon alfa‐2B/ribavirin (PR). Hepatology 2011;54(4 Suppl):814A. CENTRAL

Serfaty 2012 {published data only}

Carosi G, Colombo M, Spinetti A, Zaltron S, Serfaty L, Marcellin P, et al. Impact of insulin resistance on antiviral efficacy of telaprevir‐based regimen in treatment‐naive genotype 1 HCV: results from C208 sub‐analysis. Digestive and Liver Disease 2011;43(Suppl 2):S84. CENTRAL
Colombo M, Rumi MG, Carosi G, Puoti M, Marcellin P, Forns X, et al. Telaprevir (TVR) q8h or q12h combined with either peginterferon (PEG‐IFN, P) alfa‐2a or alfa‐2b and ribavirin (RBV, r) in treatment‐naive genotype 1 hepatitis C: final results of the randomized, open‐label, multicenter phase 2 study C208. Digestive and Liver Disease 2010;42(Suppl 1):S6‐7. CENTRAL
Forns X, Marcellin P, Ferenci P, Goser T, Nevens F, Carosi G, et al. On‐treatment response‐guided therapy with telaprevir q8h or q12h combined with peginterferon alfa‐2a or peginterferon alfa‐2b and ribavirin in treatment‐naive genotype 1 hepatitis C (study c208). Journal of Hepatology 2010;52(Suppl S1):S26. CENTRAL
Forns X, Marcellin P, Goeser T, Ferenci P, Nevens F, Carosi G, et al. Phase 2 study of telaprevir administered q8h or q12h with peginterferon‐alfa‐2a or ‐alfa‐2b and ribavirin in treatment‐naive subjects with genotype 1 hepatitis C: week 4 interim results. Hepatology 2008;48(4):1136A‐7A. CENTRAL
Marcellin P, Forns X, Goeser T, Ferenci P, Nevens F, Carosi G, et al. On‐treatment response‐guided therapy with telaprevir Q8h or Q12h combined with peginterferon alfa‐2a or peginterferon alfa‐2b and ribavirin in treatment‐naive genotype 1 hepatitis C (Study C208). Gastroenterology 2010;138(5 Suppl 1):S847. CENTRAL
Marcellin P, Forns X, Goeser T, Ferenci P, Nevens F, Carosi G, et al. Virological analysis of patients receiving telaprevir administered q8h or q12h with peginterferon‐alfa‐2a or ‐alfa‐ 2b and ribavirin in treatment ‐ naive patients with genotype 1 hepatitis C: study C208. Hepatology 2009;50(4 Suppl):395A. CENTRAL
Marcellin P, Forns X, Goser T, Ferenci P, Nevens F, Carosi G, et al. Telaprevir Q8H or Q12H combined with either peginterferon (PEG‐IFN, P) alfa‐2a or alfa‐2b and ribavirin (RBV, R) in treatment‐naive genotype 1 hepatitis C patients: Final results of the randomized, open‐label, multicenter phase 2 study C208. Hepatology International 2010;4(1):61. CENTRAL
Serfaty L, Forns X, Goeser T, Ferenci P, Nevens F, Carosi G, et al. Insulin resistance and response to telaprevir plus peginterferon a and ribavirin in treatment‐naive patients infected with HCV genotype 1. Gut 2012;61(10):1473‐80. CENTRAL

Sulkowski 2011 {published data only}

Sulkowski MS, Poordad F, Manns MP, Bronowicki JP, Reddy KR, Harrison SA, et al. Anemia during treatment with peginterferon alfa‐2b/ribavirin with or without boceprevir is associated with higher SVR rates: analysis of previously untreated and previous‐treatment‐failure patients. Journal of Hepatology 2011;54(Suppl 1):S194‐5. CENTRAL

Sulkowski 2012a {published data only}

Sulkowski MS, Roberts S, Afdhal N, Andreone P, Diago M, Pol S, et al. Ribavirin dose modification in treatment‐naive and previously treated patients who received telaprevir combination treatment: no impact on sustained virologic response in phase 3 studies. Journal of Hepatology 2012;56(Suppl S2):S459‐60. CENTRAL

Sulkowski 2012b {published data only}

Sulkowski MS, Rodriguez‐Torres M, Lawitz E, Shiffman ML Pol S, Herring RW, et al. Complete SVR4 rates in treatment‐naive HCV genotype 1a and 1b patients who achieved vRVR with an interferon‐free all‐oral regimen. Hepatology 2012;56(S1):298A. CENTRAL

Sulkowski 2013d {published data only}

Sulkowski M, Bourliere M, Bronowicki JP, Streinu‐Cercel A, Preotescu L, Asselah T, et al. Silen‐C2: early antiviral activity and safety of BI 201335 combined with peginterferon alfa‐2A and ribavirin (PegIfn/Rbv) in chronic HCV genotype‐1 patients with non‐response to PegIfn/Rbv. Journal of Hepatology 2010;52(Suppl S1):S462‐3. CENTRAL
Sulkowski MS, Bourliere M, Bronowicki JP, Asselah T, Pawlotsky JM, Shafran SD, et al. Faldaprevir combined with peginterferon alfa‐2a and ribavirin in chronic hepatitis C virus genotype‐1 patients with prior nonresponse: SILEN‐C2 trial. Hepatology 2013;57(6):2155‐63. CENTRAL
Sulkowski MS, Bourliere M, Bronowicki JP, Streinu‐Cercel A, Preotescu L, Asselah T, et al. Silen‐C2: sustained virologic response (SVR) and safety of BI201335 combined with peginterferon alfa‐2A and ribavirin (P/R) in chronic HCV genotype‐1 patients with non‐response to P/R. Journal of Hepatology 2011;54(Suppl S1):S30. CENTRAL

Sulkowski 2014 {published data only}

Sulkowski MS, Gardiner DF, Rodriguez‐Torres M, Reddy KR, Hassanein T, Jacobson I, et al. Daclatasvir plus sofosbuvir for previously treated or untreated chronic HCV infection. New England Journal of Medicine 2014;370(3):211‐21. CENTRAL

Zeuzem 2012 {published data only}

Zeuzem S, Soriano V, Asselah T, Bronowicki JP, Ceausu E, Lohse AW, et al. Virologic response to an interferon‐free regimen of BI201335 and BI207127, with and without ribavirin, in treatment‐naive patients with chronic genotype‐1 HCV infection: week 12 interim results of the sound‐C2 study. Hepatology 2011;54(4 Suppl):1436A. CENTRAL
Zeuzem S, Soriano V, Asselah T, Bronowicki JP, Lohse A, Mullhaupt B, et al. SVR4 and SVR12 with an interferon‐free regimen of BI201335 and BI207127, +/‐ribavirin, in treatment‐naive patients with chronic genotype‐1 HCV infection: interim results of sound‐C2. Journal of Hepatology 2012;56(Suppl S2):S45. CENTRAL

Zeuzem 2013 {published data only}

Zeuzem S, Asselah T, Angus P, Zarski JP, Larrey D, Muellhaupt B, et al. Faldaprevir (BI201335), deleobuvir (BI207127) and ribavirin oral therapy for treatment‐naive HCV genotype 1: SOUND‐C1 final results. Antiviral Therapy 2013;18(8):1015‐9. CENTRAL
Zeuzem S, Asselah T, Angus PW, Zarski JP, Larrey DG, Mullhaupt B, et al. High sustained virologic response following interferon‐free treatment of chronic HCV GT1 infection for 4 weeks with HCV protease inhibitor BI201335, polymerase inhibitor BI207127 and ribavirin, followed by BI201335 and pegifn/ribavirin the sound‐C1 study. Hepatology 2011;54(S1):486A‐7A. CENTRAL

Zeuzem 2014b {published data only}

Younossi ZM, Stepanova M, Zeuzem S, Dusheiko G, Esteban R, Hezode C, et al. Patient‐reported outcomes assessment in chronic hepatitis C treated with sofosbuvir and ribavirin: The VALENCE study. Journal of Hepatology 2014;61(2):228‐34. CENTRAL
Zeuzem S, Dusheiko GM, Salupere R, Mangia A, Flisiak R, Hyland RH, et al. Sofosbuvir + ribavirin for 12 or 24 weeks for patients with HCV genotype 2 or 3: the VALENCE trial. Hepatology 2013;58(Suppl S1):733A‐4A. CENTRAL
Zeuzem S, Dusheiko GM, Salupere R, Mangia A, Flisiak R, Hyland RH, et al. Sofosbuvir and ribavirin in HCV genotypes 2 and 3. New England Journal of Medicine 2014;370(21):1993‐2001. CENTRAL

References to ongoing studies

Izumi 2012 {published data only}

Izumi N, LaTaillade M, Chayama K, Toyota J, Mochida S, Kawada N, et al. First report of peginterferon lambda/ribavirin in combination with either daclatasvir or asunaprevir in HCV genotype 1 Japanese subjects: early sustained virologic response (SVR4) results from the D‐LITE Japanese sub‐study. Hepatology 2012;56(S1):310A. CENTRAL

Lawitz 2014b {published data only}

Lawitz E, Rodriguez‐Torres M, Nguyen T, Sheikh A, Tobias H, Galati J, et al. A phase II study of samatasvir (IDX719) in combination with simeprevir and ribavirin in treatment‐naive HCV‐infected subjects with genotypes 1B and 4 (helix‐1 study). Journal of Hepatology 2014;60(1 Suppl S):S495‐6. CENTRAL

Afdhal 2014

Afdhal N, Zeuzem S, Kwo P, Chojkier M, Gitlin N, Puoti M, et al. Ledipasvir and sofosbuvir for untreated HCV genotype 1 infection. New England Journal of Medicine 2014;370(20):1889‐98.

Bacon 2011

Bacon BR, Gordon SC, Lawitz E, Marcellin P, Vierling JM, Zeuzem S, et al. Boceprevir for previously treated chronic HCV genotype 1 infection. New England Journal of Medicine 2011;364(13):1207‐17.

Balshem 2011

Balshem H, Helfand M, Schunemann HJ, Oxman AD, Kunz R, Brozek J, et al. GRADE guidelines: 3. Rating the quality of evidence. Journal of Clinical Epidemiology 2011;64(4):401‐6. [PUBMED: 21208779]

Basile 2014

Basile N. Sofosbuvir‐ribavirin duo for chronic hepatitis C. Connecticut Medicine 2014;78(6):355‐6.

Berger 2012

Berger CT, Kim AY. IL28B polymorphisms as a pre‐treatment predictor of response to HCV treatment. Infectious Disease Clinics of North America 2012;26(4):863‐77. [DOI: 10.1016/j.idc.2012.08.010]

Brok 2009

Brok J, Gluud LL, Gluud C. Ribavirin monotherapy for chronic hepatitis C. Cochrane Database of Systematic Reviews 2009, Issue 4. [DOI: 10.1002/14651858.CD005527.pub2]

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]

CDCP 1998

Centers for Disease Control and Prevention. Recommendations for prevention and control of hepatitis C virus (HCV) infection and HCV‐related chronic disease. MMWR. Recommendations and Reports: Morbidity and Mortality Weekly Report 1998;47(RR‐19):1‐39.

Chan 2013

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

Childs‐Kean 2015

Childs‐Kean LM, Hand EO. Simeprevir and sofosbuvir for treatment of chronic hepatitis C infection. Clinical Therapeutics 2015;37(2):243‐67.

Choo 1989

Choo QL, Kuo G, Weiner AJ, Overby LR, Bradley DW, Houghton M. Isolation of a cDNA clone derived from a blood‐borne non‐A, non‐B viral hepatitis genome. Science 1989;244(4902):359‐62.

Ciani 2017

Ciani O, Buyse M, Drummond M, Rasi G, Saad ED, Taylor RS. Time to review the role of surrogate end points in health policy: state of the art and the way forward. Value Health 2017;20(3):487‐95.

Clark 2012

Clark V, Nelson DR. The role of ribavirin in direct acting antiviral drug regimens for chronic hepatitis C. Liver International 2012;32(1):103‐7.

Coco 2014

Coco B, Caraceni P, Aghemo A, Bitetto D, Bruno R, Ciancio A, et al. Triple therapy with first‐generation protease inhibitors for patients with genotype 1 chronic hepatitis C: recommendations of the Italian association for the study of the liver (AISF). Digestive and Liver Disease 2014;46(1):18‐24.

Conteduca 2014

Conteduca V, Sansonno D, Russi S, Pavone F, Dammacco F. Therapy of chronic hepatitis C virus infection in the era of direct‐acting and host‐targeting antiviral agents. Journal of Infection 2014;68(1):1‐20.

Conti 2016

Conti F, Buonfiglioli F, Scuteri A, Crespi C, Bolondi L, Caraceni P, et al. Early occurrence and recurrence of hepatocellular carcinoma in HCV‐related cirrhosis treated with direct‐acting antivirals. Journal of Hepatology 2016;65(4):727‐33.

Deeks 2011

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

Dubuisson 2014

Dubuisson J, Cosset FL. Virology and cell biology of the hepatitis C virus life cycle: an update. Journal of Hepatology 2014;61(1 Suppl):S3‐S13.

EASL 2015

European Association for Study of Liver. EASL Recommendations on Treatment of Hepatitis C. Journal of Hepatology 2015;63(1):199‐236.

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‐Serag 2016

El‐Serag HB, Kanwal F, Richardson P, Kramer J. Risk of hepatocellular carcinoma after sustained virological response in veterans with hepatitis C virus infection. Hepatology 2016;64(1):130‐7.

Elbaz 2015

Elbaz T, Mohamed El‐Kassas M, Esmat G. New era for management of chronic hepatitis C virus using direct antiviral agents: a review. Journal of Advanced Research 2015;6(3):301–10.

Ermis 2015

Ermis F, Senocak Tasci E. New treatment strategies for hepatitis C infection. World Journal of Hepatology 2015;7(17):2100‐9.

FiercePharma 2017

Engeneering & Biotechnology News. FiercePharma, annual Reports, company websites, SEC Filings and press releases 2017.

Flemming 1996

Fleming TR, DeMets DL. Surrogate end points in clinical trials: Are we being misled?. Annals of Internal Medicine 1996, 125(7):605‐613. .

Flemming 2012

Fleming TR, Powers JH. Biomarkers and surrogate endpoints in clinical trials. Statistics in medicine 2012, 31(25):2973‐2984. .

Garattini 2016

Garattini S, Jakobsen JC, Wetterslev J, Bertele V, Banzi R, Rath A, et al. Evidence‐based clinical practice: overview of threats to the validity of evidence and how to minimise them. European Journal of Internal Medicine 2016;32:13‐21.

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 2015

Gluud C, Nikolova D, Klingenberg SL. Cochrane Hepato‐Biliary Group. About Cochrane (Cochrane Review Groups (CRGs)) 2016, Issue 2. Art. No.: LIVER Vol. http://hbg.cochrane.org.

Gowan 2014

Gower E, Estes C, Blach S, Razavi‐Shearer K, Razavi H. Global epidemiology and genotype distribution of the hepatitis C virus infection. Journal of Hepatology 2014;61(1):45‐57.

Guyatt 2011a

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. [PUBMED: 21195583]

Guyatt 2011b

Guyatt GH, Oxman AD, Kunz R, Atkins D, Brozek J, Vist G, et al. GRADE guidelines: 2. Framing the question and deciding on important outcomes. Journal of Clinical Epidemiology 2011;64(4):395‐400. [PUBMED: 21194891]

Guyatt 2011c

Guyatt GH, Oxman AD, Vist G, Kunz R, Brozek J, Alonso‐Coello P, et al. GRADE guidelines: 4. Rating the quality of evidence‐‐study limitations (risk of bias). Journal of Clinical Epidemiology 2011;64(4):407‐15. [PUBMED: 21247734]

Guyatt 2011d

Guyatt GH, Oxman AD, Montori V, Vist G, Kunz R, Brozek J, et al. GRADE guidelines: 5. Rating the quality of evidence‐‐publication bias. Journal of Clinical Epidemiology 2011;64(12):1277‐82. [PUBMED: 21802904]

Guyatt 2011e

Guyatt GH, Oxman AD, Kunz R, Brozek J, Alonso‐Coello P, Rind D, et al. GRADE guidelines 6. Rating the quality of evidence‐‐imprecision. Journal of Clinical Epidemiology 2011;64(12):1283‐93. [PUBMED: 21839614]

Guyatt 2011f

Guyatt GH, Oxman AD, Kunz R, Woodcock J, Brozek J, Helfand M, et al. GRADE guidelines: 7. Rating the quality of evidence‐‐inconsistency. Journal of Clinical Epidemiology 2011;64(12):1294‐302. [PUBMED: 21803546]

Guyatt 2011g

Guyatt GH, Oxman AD, Kunz R, Woodcock J, Brozek J, Helfand M, et al. GRADE guidelines: 8. Rating the quality of evidence‐‐indirectness. Journal of Clinical Epidemiology 2011;64(12):1303‐10. [PUBMED: 21802903]

Guyatt 2011h

Guyatt GH, Oxman AD, Sultan S, Glasziou P, Akl EA, Alonso‐Coello P, et al. GRADE guidelines: 9. Rating up the quality of evidence. Journal of Clinical Epidemiology 2011;64(12):1311‐6. [PUBMED: 21802902]

Guyatt 2013a

Guyatt G, Oxman AD, Sultan S, Brozek J, Glasziou P, Alonso‐Coello P, et al. GRADE guidelines: 11. Making an overall rating of confidence in effect estimates for a single outcome and for all outcomes. Journal of Clinical Epidemiology 2013;66(2):151‐7. [PUBMED: 22542023]

Guyatt 2013b

Guyatt GH, Oxman AD, Santesso N, Helfand M, Vist G, Kunz R, et al. GRADE guidelines: 12. Preparing summary of findings tables‐binary outcomes. Journal of Clinical Epidemiology 2013;66(2):158‐72. [PUBMED: 22609141]

Guyatt 2013c

Guyatt GH, Thorlund K, Oxman AD, Walter SD, Patrick D, Furukawa TA, et al. GRADE guidelines: 13. Preparing summary of findings tables and evidence profiles‐continuous outcomes. Journal of Clinical Epidemiology 2013;66(2):173‐83. [PUBMED: 23116689]

Götte 2016

Götte M, Feld JJ. Direct‐acting antiviral agents for hepatitis C: structural and mechanistic insights. Nature Reviews. Gastroenterology & Hepatology 2016;13:338–51.

Harbord 2006

Harbord RM, Egger M, Sterne JA. A modified test for small‐study effects in meta‐analyses of controlled trials with binary endpoints. Statistics in Medicine 2006;25(20):3443‐57.

Hauser 2014

Hauser G, Awad T, Brok J, Thorlund K, Štimac D, Mabrouk M, et al. Peginterferon plus ribavirin versus interferon plus ribavirin for chronic hepatitis C. Cochrane Database of Systematic Reviews 2014, Issue 2. [DOI: 10.1002/14651858.CD005441.pub3]

Higgins 2003

Higgins JPT, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta‐analyses. BMJ 2003;327:557‐60.

Higgins 2011a

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

Higgins 2011b

Higgins JPT, Altman DG, Sterne JAC (editors). Chapter 8: Assessing risk of bias in included studies. In: Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Higgins 2011c

Higgins JPT, Deeks JJ, Altman DG (editors). Chapter 16: Special topics in statistics. In: Higgins JPT, Green S (editors), Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from www.handbook.cochrane.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, Philadelphia (PA): Barnett International/PAREXEL, 1997.

Jacobson 2011

Jacobson IM, McHutchison JG, Dusheiko G, Di Bisceglie AM, Reddy KR, Bzowej NH, et al. Telaprevir for previously untreated chronic hepatitis C virus infection. New England Journal of Medicine 2011;364(25):2405‐16.

Jakobsen 2014a

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.

Jakobsen 2014b

Jakobsen JC, Wetterslev J, Winkel P, Lange T, Gluud C. The thresholds for statistical and clinical significance ‐ a five‐step procedure for evaluation of intervention effects in randomised clinical trials. BMC Medical Research Methodology 2014;14:34.

Jakobsen 2016a

Jakobsen JC, Wetterslev J, Lange T, Gluud C. Viewpoint: taking into account risks of random errors when analysing multiple outcomes in systematic reviews [editorial]. Cochrane Database of Systematic Reviews2016, issue 3:10.1002/14651858.ED000111. [DOI: 10.1002/14651858.ED000111]

Jakobsen 2016b

Jakobsen JC, Nielsen EE, Feinberg J, Fobian K, Katakam KK, Hauser G, et al. Direct‐acting antivirals for chronic hepatitis C. Cochrane Database of Systematic Reviews 2016, Issue 4. [DOI: 10.1002/14651858.CD012143]

Kemp 2017

Kemp R, Prasad V. Surrogate endpoints in oncology: when are they acceptable for regulatory and clinical decisions, and are they currently overused?. BMC Medicine 2017, 15(1):134. .

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.

Koretz 2013

Koretz RL, Pleguezuelo M, Arvaniti V, Barrera BP, Ciria R, Gurusamy KS, et al. Interferon for interferon nonresponding and relapsing patients with chronic hepatitis C. Cochrane Database of Systematic Reviews 2013, Issue 1. [DOI: 10.1002/14651858.CD003617.pub2]

Koretz 2015

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

Koretz 2016

Koretz, R. Direct‐acting antivirals: do they work beyond sustained virological response in chronic hepatitis C patients? [Conference proceeding at the International Liver Congress 2016, Barcelona]. EASL. 2016; Vol. ilc‐congress.eu/wp‐content/uploads/2016/programme_book/EASL_ILC%20scientific%20programme%202016_Web.pdf.

Lang 2013

Lang D, Liang HJ, Li D, Wei X, Ma L, Jia Z. The efficacy and safety of telaprevir‐based regimens for treating chronic hepatitis C virus genotype 1 infection: a meta‐analysis of randomized trials. Internal Medicine 2013;52(6):653‐60.

Lawitz 2013

Lawitz E, Mangia A, Wyles D, Rodriguez‐Torres M, Hassanein T, Gordon SC, et al. Sofosbuvir for previously untreated chronic hepatitis C infection. New England Journal of Medicine 2013;368(20):1878‐87.

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]

Manns 2006

Manns MP, Wedemeyer H, Cornberg M. Treating viral hepatitis C: efficacy, side effects, and complications. Gut 2006;55(9):1350‐9.

MCDC 2015

GBD 2013 Mortality and Causes of Death Collaborators. Global, regional, and national age‐sex specific all‐cause and cause‐specific mortality for 240 causes of death, 1990‐2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet 2015;385(9963):117‐71.

Messina 2015

Messina JP, Humphreys I, Flaxman A, Brown A, Cooke GS, Pybus OG, et al. Global distribution and prevalence of hepatitis C virus genotypes. Hepatology 2015;61(1):77‐87.

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.

Moher 2009

Moher D, Liberati A, Tetzlaff J, Altman DG, The PRISMA Group (2009). Preferred Reporting Items for Systematic Reviews and Meta‐Analyses: The PRISMA Statement.. PLoS Medicine 2009;6(7):e1000097. [DOI: 10.1371/journal.pmed1000097]

Mok 2005

Mok J, Pembrey L, Tovo PA, Newell ML, European Paediatric Hepatitis C Virus Network. When does mother to child transmission of hepatitis C virus occur?. Archives of Disease in Childhood. Fetal and Neonatal Edition 2005;90(2):156‐60.

Mustafa 2013

Mustafa RA, Santesso N, Brozek J, Akl EA, Walter SD, Norman G, et al. The GRADE approach is reproducible in assessing the quality of evidence of quantitative evidence syntheses. Journal of Clinical Epidemiology 2013;66(7):736‐42; quiz 742.e1‐5. [PUBMED: 23623694]

NICE 2015a

NICE. Interferon alfa (pegylated and non‐pegylated) and ribavirin for the treatment of chronic hepatitis C. www.nice.org.uk/guidance/ta75NICE.org 2015 (accessed 2 March 2016).

NICE 2015b

NICE. Sofosbuvir for treating chronic hepatitis C. www.nice.org.uk/guidance/ta330/chapter/2‐The‐technology 2015 (accessed 2 March 2016).

Pockros 2015

Pockros PJ. Direct‐acting antivirals for the treatment of hepatitis C virus infection. www.uptodate.com/contents/direct‐acting‐antivirals‐for‐the‐treatment‐of‐hepatitis‐c‐virus‐infection?source=search_result&search=Direct‐acting+antivirals+for+the+treatment+of+hepatitis+C+virus+infection&selectedTitle=1˜150 2015 (accessed 2 March 2016).

Poordad 2011

Poordad F, McCone J, Bacon BR, Bruno S, Manns MP, Sulkowski MS, et al. SPRINT‐2 Investigators. Boceprevir for untreated chronic HCV genotype 1 infection. New England Journal of Medicine 2011;364(13):1195‐206.

Poordad 2012

Poordad F, Dieterich D. Treating hepatitis C: current standard of care and emerging direct‐acting antiviral agents. Journal of Viral Hepatitis 2012;19(7):449‐64.

Reig 2016

Reig M, Marino Z, Perello C, Inarrairaegui M, Ribeiro A, Lens S, et al. Unexpected high rate of early tumor recurrence in patients with HCV‐related HCC undergoing interferon‐free therapy. J Hepatol. 2016;65(4):719‐26. .

Reig 2017

Reig M, Boix L, Marino Z, Torres F, Forns X, Bruix J. Liver Cancer Emergence Associated with Antiviral Treatment: An Immune Surveillance Failure?. Semin Liver Dis. 2017;37(2):109‐18. .

RevMan 2014 [Computer program]

Nordic Cochrane Centre, The Cochrane Collaboration. Review Manager 5 (RevMan 5). Version 5.3. Copenhagen: Nordic Cochrane Centre, The Cochrane Collaboration, 2014.

Righi 2015

Righi E, Londero A, Carnelutti A, Baccarani U, Bassetti M. Impact of new treatment options for hepatitis C virus infection in liver transplantation. World Journal of Gastroenterology 2015;21(38):10760‐75.

Roche 2015

Roche B, Coilly A, Roque‐Afonso AM, Samuel D. Interferon‐free hepatitis C treatment before and after liver transplantation: the role of HCV drug resistance. Viruses 2015;7(9):5155‐68.

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. Annals of Internal Medicine 2012;157(6):429‐38.

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. Health Technology Assessment 2012;16(35):1‐82.

Scheel 2013

Scheel TK, Rice CM. Understanding the hepatitis C virus life cycle paves the way for highly effective therapies. Nature Medicine 2013;19(7):837‐49.

Schultz 2010

Schulz KF, Altman DG, Moher D. CONSORT 2010 statement: updated guidelines for reporting parallel group randomized trials. Annals of Internal Medicine 2010;152(11):726‐32.

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. Journal of the American Medical Association 1995;273(5):408‐12.

Schünemann 2011

Schünemann HJ, Oxman AD, Higgins JPT, Vist GE, Glasziou P, Guyatt GH. Chapter 11: Presenting results and ‘Summary of findings' tables. In: Higgins JPT, Green S (editors), Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Smith‐Palmer 2015

Smith‐Palmer J, Cerri K, Valentine W. Achieving sustained virologic response in hepatitis C: a systematic review of the clinical, economic and quality of life benefits. BMC Infectious Diseases 2015;15(19).

Spradling 2012

Spradling PR, Rupp L, Moorman AC, Lu M, Teshale EH, Gordon SC, et al. Chronic Hepatitis Cohort Study Investigators. Hepatitis B and C virus infection among 1.2 million persons with access to care: factors associated with testing and infection prevalence. Clinical Infectious Diseases 2012;55(8):1047‐55.

Sterne 2011

Sterne JAC, Egger M, Moher D (editors). Chapter 10: Addressing reporting biases. In: Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Intervention. Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Sweeting 2004

Sweeting MJ, Sutton AJ, Lambert PC. What to add to nothing? Use and avoidance of continuity corrections in meta‐analysis of sparse data. Statistics in Medicine 2004;23(9):1351‐75.

Thein 2008

Thein HH, Yi Q, Dore GJ, Krahn MD. Estimation of stage‐specific fibrosis progression rates in chronic hepatitis C virus infection: a meta‐analysis and meta‐regression.. Hepatology 2008;48(2):418.

Thomas 2009

Thomas DL, Thio CL, Martin MP, Qi Y, Ge D, O'Huigin C, et al. Genetic variation in IL28B and spontaneous clearance of hepatitis C virus. Nature 2009;461(7265):798–801.

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 2 March 2016).

TSA 2011 [Computer program]

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

Wandeler 2015

Wandeler G, Dufour JF, Bruggmann P, Rauch A. Hepatitis C: a changing epidemic. Swiss Medical Weekly 2015;145:w14093.

Wang 2017

Wang C, Ji D, Chen J, Shao Q, Li B, Liu J, Wu V, Wong A, Wang Y, Zhang X, Lu L, Wong C. Tsang S, Zhanf Z, Sun J, Hou J, Chen G, Lau G. Hepatitis due to reactivation of hepatitis B virus in endemic areas among patients with hepatitis C treated with direct‐acting antiviral agents. Clin Gastroenterol Hepatol 2017; 15:132‐6 .

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.

WHO 2014

WHO. Hepatitis C Fact sheet N°164. www.who.int/mediacentre/factsheets/fs164/en/ 2015 (accessed 2 March 2016).

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;366:601‐5.

Wyles 2015

Wyles DL, Ruane PJ, Sulkowski MS, Dieterich D, Luetkemeyer A, Morgan TR, et al. Daclatasvir plus sofosbuvir for HCV in patients coinfected with HIV‐1. New England Journal of Medicine 2015;373(8):714‐25.

Characteristics of studies

Characteristics of included studies [ordered by study ID]

ADVANCE 2011a1

Methods

Randomised phase III clinical trial

Participants

1088 participants

Countries: Europe and USA

Inclusion criteria: participants with HCV genotype 1 infection who had not received previous treatment 18‐70 years of age and had HCV genotype 1 infection with evidence of chronic hepatitis, as confirmed by means of a liver biopsy within 1 year before screening for the study; people with compensated liver cirrhosis were eligible.

Exclusion criteria: advanced liver disease, co‐infection with HBV or HIV, HCC, other clinical relevant comorbidity. ALT> 5 x the ULN, total bilirubin > 2 mL/dL, albumin < 3.5 g/dL, international normalised ratio > 1.7, platelets < 90 x 109, haemoglobin < 12 g/dL (women) or < 13 g/dL (men).

Interventions

Experimental group 1: telaprevir (orally at a dose of 750 mg every 8 h) and peg‐IFN α‐2a (by subcutaneous injection at a dose of 180 μg per week) and RBV (orally at a dose of 1000 mg per day (in participants who weighed < 75 kg) or 1200 mg per day (in participants who weighed ≥ 75 kg)) for the entire 12 weeks followed by 4 weeks of placebo and peg‐IFN–RBV (T12PR group).

Experimental group 2: telaprevir (orally at a dose of 750 mg every 8 h) and peg‐IFN α‐2a (by subcutaneous injection at a dose of 180 μg per week) and RBV (orally at a dose of 1000 mg per day (in participants who weighed < 75 kg) or 1200 mg per day (in participants who weighed ≥ 75 kg) for 8 weeks and placebo with peg‐IFN–RBV for 4 weeks (T8PR group).

Control group: placebo with peg‐IFN–RBV for 12 weeks, followed by 36 weeks of peg‐IFN–RBV.

Participants in the T12PR and T8PR groups who met the criteria for an extended RVR (defined as undetectable HCV RNA at weeks 4 and 12) received 12 additional weeks of treatment with peg‐IFN–RBV alone, for a total treatment period of 24 weeks. Participants in the T12PR and T8PR groups who had detectable HCV RNA either at week 4 or at week 12 received 36 additional weeks of treatment with peg‐IFN–RBV, for a total treatment period of 48 weeks. The group receiving peg‐IFN α‐2a and RBV alone (PR group) received placebo plus peg‐IFN– RBV for 12 weeks, followed by peg‐IFN–RBV alone for 36 additional weeks.

Co‐intervention: peg‐IFN (subcutaneously at 180 µg/week) and RBV orally twice daily dosed according to body weight.

Outcomes

HCV RNA, safety assessment

Notes

We emailed Jacobson and colleagues on 21 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as being double‐blinded but it was unclear how the blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Described as being double‐blinded but it was unclear how the blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Low risk

< 5% dropped out

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol were assessed

Vested‐interest bias

High risk

The trial was funded by Bristol‐Myers Squibb

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

ADVANCE 2011a2

Methods

For characteristics see ADVANCE 2011a2

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as being double‐blinded but it was unclear how the blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Described as being double‐blinded but it was unclear how the blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Low risk

< 5% dropped out

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol were assessed

Vested‐interest bias

High risk

The trial was funded by Bristol‐Myers Squibb

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Anderson 2014a1

Methods

Randomised clinical trial

Participants

74 participants were randomised

Sex: 58 men, 16 women

Mean age: 50.2

Inclusion criteria: treatment‐naive adults 18‐65 years of age with chronic HCV genotype 1 infection for > 6 months before study enrolment, with a BMI > 18 and < 35 kg/m2. Chronic HCV infection was defined as 1 of the following: detectable HCV RNA or reactive HCV antibody > 6 months before enrolment; reactive antibody for HCV before screening and a liver biopsy > 6 months before enrolment demonstrating pathology consistent with HCV infection; and reactive HCV antibody or detectable HCV RNA before screening with an HCV risk factor (e.g. unsafe injection practices, blood transfusion before June 1992, receipt of clotting factor before 1987) that had emerged > 6 months before enrolment. In addition, participants had a liver biopsy result with histology consistent with HCV‐induced liver damage and with no evidence of cirrhosis or liver pathology due to any cause other than chronic HCV within the 3‐year period before study enrolment and participants had plasma HCV RNA level > 100.000 IU/mL at screening.

Exclusion criteria: participants with METAVIR fibrosis score of 3 or 4 on liver biopsy, a positive test result for hepatitis B surface antigen or anti‐HIV antibodies, a history of major depression within the 2 years before enrolment, or unresolved clinically significant diseases other than HCV were excluded from participation.

Interventions

Experimental group:

  1. ABT‐450/r 50/100 mg once a day + peg‐IFN/RBV

  2. ABT‐450/r 100/100 mg once a day + peg‐IFN/RBV

  3. ABT‐450/r 200/100 mg once a day + peg‐IFN/RBV

  4. ABT‐072 100 mg once a day + peg‐IFN/RBV

  5. ABT‐072 300 mg once a day + peg‐IFN/RBV

  6. ABT‐072 600 mg once a day + peg‐IFN/RBV

  7. ABT‐333 400 mg twice a day + peg‐IFN/RBV

  8. ABT‐333 800 mg twice a day + peg‐IFN/RBV

Control group: placebo + peg‐IFN/RBV

Co‐intervention: peg‐IFN and RBV

Participants were treated with ABT‐450/r, ABT‐333, or ABT‐072 monotherapy for 3 days, followed by 81 days (12 weeks minus 3 days of monotherapy) of ABT‐450/r, ABT‐333, or ABT‐072 combined with pegylated IFN/RBV (peg‐IFN/RBV), followed by 36 weeks of peg‐IFN/RBV alone.

Outcomes

Primary outcomes: maximal change from baseline in HCV RNA levels, maximum plasma concentration (Cmax) of ABT‐450, time to maximum plasma concentration (Tmax) of ABT‐450, area under the plasma concentration‐time curve from 0‐24 h (AUC24) post‐dose of ABT‐450, maximum plasma concentration (Cmax) of ritonavir, time to maximum plasma concentration (Tmax) of ritonavir, area under the plasma concentration‐time curve from 0‐24 h (AUC24) post‐dose of ritonavir, maximum plasma concentration (Cmax) of ABT‐072, time to maximum plasma concentration (Tmax) of ABT‐072, area under the plasma concentration‐time curve from 0‐24 h (AUC24) post‐dose of ABT‐072, maximum plasma concentration (Cmax) of ABT‐333, time to maximum plasma concentration (Tmax) of ABT‐333, area under the plasma concentration‐time curve from 0‐12 h (AUC12) post‐dose of abt‐333.

Secondary outcomes: percentage of participants with rapid virologic response (RVR) at week 4, percentage of participants with partial early virologic response (EVR) at week 12, Ppercentage of participants with complete early virologic response (cEVR) at week 12.

Notes

We emailed Anderson and colleagues on 20 April 2016 for unpublished data and additional information regarding allocation concealment, random sequence generation, and blinding of outcome but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Investigators and participants were blinded to the study drug treatment regimen, but it was not stated how the blinding was maintained.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% of participants did not complete the study (19%), according to study protocol

Selective reporting (reporting bias)

Low risk

A protocol was found (NCT01074008)

Vested‐interest bias

High risk

This study was funded by AbbVie

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Anderson 2014a2

Methods

For characteristics see Anderson 2014a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Investigators and participants were blinded to the study drug treatment regimen, but it was not stated how the blinding was maintained.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% of participants did not complete the study (19%), according to study protocol

Selective reporting (reporting bias)

Low risk

A protocol was found (NCT01074008)

Vested‐interest bias

High risk

This study was funded by AbbVie

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Anderson 2014a3

Methods

For characteristics see Anderson 2014a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Investigators and participants were blinded to the study drug treatment regimen, but it was not stated how the blinding was maintained.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% of participants did not complete the study (19%), according to study protocol

Selective reporting (reporting bias)

Low risk

A protocol was found (NCT01074008)

Vested‐interest bias

High risk

This study was funded by AbbVie

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Anderson 2014a4

Methods

For characteristics see Anderson 2014a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Investigators and participants were blinded to the study drug treatment regimen, but it was not stated how the blinding was maintained.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% of participants did not complete the study (19%), according to study protocol

Selective reporting (reporting bias)

Low risk

A protocol was found (NCT01074008)

Vested‐interest bias

High risk

This study was funded by AbbVie

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Anderson 2014a5

Methods

For characteristics see Anderson 2014a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Investigators and participants were blinded to the study drug treatment regimen, but it was not stated how the blinding was maintained.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% of participants did not complete the study (19%), according to study protocol

Selective reporting (reporting bias)

Low risk

A protocol was found (NCT01074008)

Vested‐interest bias

High risk

This study was funded by AbbVie

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Anderson 2014a6

Methods

For characteristics see Anderson 2014a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Investigators and participants were blinded to the study drug treatment regimen, but it was not stated how the blinding was maintained.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% of participants did not complete the study (19%), according to study protocol

Selective reporting (reporting bias)

Low risk

A protocol was found (NCT01074008)

Vested‐interest bias

High risk

This study was funded by AbbVie

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Anderson 2014a7

Methods

For characteristics see Anderson 2014a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Investigators and participants were blinded to the study drug treatment regimen, but it was not stated how the blinding was maintained.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% of participants did not complete the study (19%), according to study protocol

Selective reporting (reporting bias)

Low risk

A protocol was found (NCT01074008)

Vested‐interest bias

High risk

This study was funded by AbbVie

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Anderson 2014a8

Methods

For characteristics see Anderson 2014a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Investigators and participants were blinded to the study drug treatment regimen, but it was not stated how the blinding was maintained.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% of participants did not complete the study (19%), according to study protocol

Selective reporting (reporting bias)

Low risk

A protocol was found (NCT01074008)

Vested‐interest bias

High risk

This study was funded by AbbVie

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Anonymous (PPI‐461) 2011a1

Methods

Randomised clinical trial

Participants

24 treatment‐naive participants were randomised

Inclusion criteria: 18‐65 years male and female, genotype 1, treatment‐naive. Female participants must be surgically sterile or 2 years post‐menopausal and are required to take a pregnancy test. BMI 18‐32 kg/m2, chronically infected with HCV genotype 1. Serum HCV RNA > 5 log 10 IU/mL. No previous treatment with IFNIFN, peg‐IFN, RBV or any investigational HCV antiviral agents. No history or signs of decompensated liver disease. No known history of cirrhosis, no co‐infection with HBV or HIV. No history of any medical condition that may interfere with absorption, distribution or elimination of study drug or with the clinical and laboratory assessments in this study. No history of alcohol abuse, or illicit drug use within 2 years prior to screen or enrolment in a methadone maintenance programme (unless he/she has been enrolled in the programme for at least 3 months with good compliance, stable psychosocial circumstances and no known current risks for recidivism).

Interventions

Experimental group: 50 mg PPI‐461 once a day, 100 mg PPI‐461 once a day, 200 mg PPI‐461 once a day for 3 days

Control group: placebo

2 weeks' follow‐up

Co‐intervention: none

Outcomes

Primary outcomes: safety and tolerability as measured by clinical AE and laboratory assessments (time frame: up to study day 16, 14 days after the last dose of PPI‐461). Antiviral effects of PPI‐461 measured by HCV RNA levels and pharmacokinetics measured by plasma concentrations of PPI‐461 concentrations.

Notes

This is an unpublished study, only results from 2 abstracts

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial is described as double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No main publication, no drop‐outs

Selective reporting (reporting bias)

Unclear risk

Outcomes are only published in the protocol

Vested‐interest bias

High risk

Lead sponsor is Presidio Pharmaceuticals

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Anonymous (PPI‐461) 2011a2

Methods

For characteristics see Anonymous (PPI‐461) 2011a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial is described as double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No main publication, no drop‐outs

Selective reporting (reporting bias)

Unclear risk

Outcomes are only published in the protocol

Vested‐interest bias

High risk

Lead sponsor is Presidio Pharmaceuticals

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Anonymous (PPI‐461) 2011a3

Methods

For characteristics see Anonymous (PPI‐461) 2011a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial is described as double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No main publication, no drop‐outs

Selective reporting (reporting bias)

Unclear risk

Outcomes are only published in the protocol

Vested‐interest bias

High risk

Lead sponsor is Presidio Pharmaceuticals

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

ASPIRE 2014

Methods

Randomised phase IIb clinical trial

Participants

462 participants

Location: Europe and USA

Inclusion criteria: participants infected with HCV genotype 1 who had failed to respond to previous peg‐IFN/RBV treatment, adult participants, aged 18‐70 years, chronically infected with HCV genotype 1 and with plasma HCV RNA > 10,000 IU/mL at screening. All participants had to have received at least 1 prior course of peg‐IFN/RBV for 12 consecutive weeks and not discontinued therapy due to tolerability.

Exclusion criteria: decompensated liver disease, any other liver disease of non‐HCV aetiology, and infection/co‐infection with nongenotype 1 HCV.

Interventions

Experimental group:

  1. simeprevir 100 mg plus peg‐IFN/RBV 12 weeks followed by 36 weeks of peg‐IFN/RBV

  2. simeprevir 150 mg plus peg‐IFN/RBV 12 weeks followed by 36 weeks of peg‐IFN/RBV

  3. simeprevir 100 mg plus peg‐IFN/RBV 24 weeks followed by 24 weeks of peg‐IFN/RBV

  4. simeprevir 150 mg plus peg‐IFN/RBV 24 weeks followed by 24 weeks of peg‐IFN/RBV

  5. simeprevir 100 mg plus peg‐IFN/RBV 48 weeks

  6. simeprevir 150 mg plus peg‐IFN/RBV 48 weeks

Control group: 48 weeks of simeprevir‐matched placebo plus peg‐IFN/RBV

Co‐intervention: peg‐IFN (subcutaneously at 180 µg/week) and RBV orally (1000 mg or 1200 mg/day, depending on body weight). For all participants, the 48‐week treatment period was followed by post‐treatment follow‐up for up to 72 weeks after treatment initiation.

Outcomes

HCV RNA, safety assessment

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The trial used a computer random‐generation code

Allocation concealment (selection bias)

Low risk

The trial used a interactive voice‐response system

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Low risk

< 5% dropped out

Selective reporting (reporting bias)

Low risk

All outcomes stated on ClinicalTrials.gov were reported (NCT00980330)

Vested‐interest bias

High risk

The trial was funded by Bristol‐Myers Squibb

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

ATLAS 2013

Methods

Randomised phase II clinical trial

Participants

225 participants

Inclusion criteria: HCV treatment‐naïve adults aged 18 years or older with serologic evidence of chronic HCV genotype 1 infection, a serum HCV RNA level 50,000 IU/mL, and an absence of advanced fibrosis or cirrhosis (METAVIR score of F3‐4).

Exclusion criteria: participants infected with HCV non‐1 genotypes or co‐infected with HBV or with HIV were excluded, as were participants with liver disease attributable to a cause other than HCV infection, cardiac or renal disease, severe psychiatric disease, uncontrolled seizures, severe retinopathy, immunologically‐mediated disease, poorly controlled diabetes, or who were pregnant or breastfeeding. Participants were also excluded if they had a haemoglobin concentration < 11 g/dL (women), or < 12 g/dL (men); neutrophil count < 1.5 x 109 cells/L; platelet count < 90 x 109 cells/L; serum creatinine concentration > 1.5 times the ULN); or BMI (calculated as kg/m2) < 18 or > 36. The use of agents that could interfere with the metabolism of danoprevir was prohibited.

Interventions

Experimental group:

  1. dareprevir (orally at a dose of 300 mg every 8 h) and peg‐IFN α‐2a (by subcutaneous injection at a dose of 180 μg per week) and RBV (orally at a dose of 1000 mg per day (in participants who weighed < 75 kg) or 1200 mg per day (in participants who weighed ≥ 75 kg)) for the entire 12 weeks

  2. dareprevir (orally at a dose of 600 mg every 12 hours) and peg‐IFN α‐2a (by subcutaneous injection at a dose of 180 μg per week) and RBV (orally at a dose of 1000 mg per day (in participants who weighed < 75 kg) or 1200 mg per day (in participants who weighed ≥ 75 kg)) for the entire 12 weeks

  3. dareprevir (orally at a dose of 900 mg every 12 h) and peg‐IFN α‐2a (by subcutaneous injection at a dose of 180 μg per week) and RBV (orally at a dose of 1000 mg per day (in participants who weighed < 75 kg) or 1200 mg per day (in participants who weighed ≥ 75 kg or more)) for the entire 12 weeks

Control group: placebo with peg‐IFN–RBV for 24 or 48 weeks

Co‐intervention: peg‐IFN (subcutaneously at 180 µg/week) and RBV orally twice daily dosed according to body weight

Outcomes

HCV RNA (SVR), safety assessment

Notes

we emailed Marcellin and colleagues on 27 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The trial used a computer‐generated randomisation code

Allocation concealment (selection bias)

Low risk

Interactive voice/web response system

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as "partial‐blind labeling"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol were assessed (NCT00963885)

Vested‐interest bias

High risk

The trial was funded by Bristol‐Myers Squibb

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Bacon 2011a1

Methods

Parallel‐group, randomised, placebo‐controlled, double‐blind study (RESPOND‐2)(NCT00708500)

Participants

403 participants

Inclusion criteria: chronic hepatitis C infection genotype 1 HCV RNA ≥ 10,000 IU/mL, demonstrated responsiveness to IFN (minimum duration of therapy 12 weeks); non‐response defined as a decrease in HCV RNA of at least 2 log10 IU/mL by week 12, but with detectable HCV RNA during the therapy period; relapse defined as an undetectable HCV RNA at end of treatment, but without subsequent attainment of SVR. A liver biopsy with histology consistent with chronic hepatitis C, age ≥ 18 years, weight between 40‐125 kg, signed informed consent, acceptable method of contraception for the participant and participant's partner(s) for at least 2 weeks before day 1 and continue until at least 6 months after treatment termination.

Exclusion criteria: Hepatitis B infection, HIV infection, other causes of liver disease, decompensated liver disease, uncontrolled diabetes mellitus, a severe psychiatric disorder, active substance abuse, active or suspected malignancy, or a history of malignancy within last 5 years, pregnant or nursing women, severe AE during prior treatment, seizure disorder, cerebrovascular diseases, cardiovascular disease, autoimmune diseases, prior organ transplantation, haemoglobinopathies, coagulopathies, abnormal levels of serum bilirubin, albumin, and creatinine, haemoglobin < 120 g/L (women) and < 130 g/L (men), neutrophil count < 1500/mm3, platelet count < 100,000/mm3.

Group 1: 80 participants

Age, mean (years): 52.9

Sex: 58 men (72%), 22 women (28%)

Race, n(%): white: 62(84), black: 12(15), other: 1(1)

Region, n(%): North America: 51(64), European Union: 29(36). Latin America: 0.

BMI, mean ± SD (kg/m2): 28.2 ± 4.3

HCV subtype, n(%): 1a: 46(58), 1b: 34(42), missing data: 0

HCV RNA > 800,000 IU/mL, n(%): 65(81)

METAVIR fibrosis score, n(%): 0, 1, or 2: 61(76), 3 or 4: 15(19)

Cirrhosis, n(%): 10(12)

Previous therapy, n(%): peg‐IFN alpha‐2a: 42(53), peg‐IFN alpha‐2b: 38(48)

Prior non‐response, n(%): 29(36)

Prior relapse, n(%): 51(64)

Group 2: 162 participants

Age, mean (years): 52.9

Sex: 98 men (60%), 64 women (40%)

Race, n(%): white: 142(88), black: 18(11), other: 2(1).

Region, n(%): North America: 115(71), European Union: 46(28), Latin America: 1(1).

BMI, mean ± SD (kg/m2): 28.8 ± 4.6

HCV subtype, n(%): 1a: 94(58), 1b: 66(41), missing data: 2(1)

HCV RNA > 800,000 IU/mL, n(%): 147(91)

METAVIR fibrosis score, n(%): 0, 1, or 2: 117(72), 3 or 4: 32(20)

Cirrhosis, n(%): 17(10)

Previous therapy, n(%): peg‐IFN alpha‐2a: 79(49), peg‐IFN alpha‐2b: 83(51)

Prior non‐response, n(%): 57(35)

Prior relapse, n(%): 105(65)

Group 3: 161 participants

Age, mean (yr.): 52.3

Sex: 112 men (70%), 49 women (30%)

Race, n(%): white: 135(84), black: 19(12), other: 7(4)

Region, n(%): North America: 119(74), European Union: 42(26), Latin America: 0.

BMI, mean ± SD (kg/m2): 28.2 ± 4.6

HCV subtype, n(%): 1a: 96(60), 1b: 61(38), missing data, 4(2)

HCV RNA > 800,000 IU/mL, n(%): 141(88)

METAVIR fibrosis score, n(%): 0, 1, or 2: 119(74), 3 or 4: 31(20)

Cirrhosis, n(%): 17(10)

Previous therapy, n(%): pegIFN alpha‐2a: 79(49), peg‐IFN alpha‐2b: 83(51)

Prior non‐response, n(%): 57(35)

Prior relapse, n(%): 105(65)

Interventions

Experimental group:

Group 2: oral boceprevir 800 mg thrice‐daily to be taken in with food and with an interval of 7‐9 h, in 4 capsules of 200 mg each, beginning at week 5 for a total of 32 weeks (if HCV RNA undetectable at week 8 and 12, treatment was terminated at week 36; if HCV RNA detectable at week 8 participants received placebo + peg‐IFN + RBV for an additional 12 weeks).

Group 3: oral boceprevir 800 mg thrice‐daily to be taken in with food and with an interval of 7‐9 h, in 4 capsules of 200 mg each, beginning at week 5 for a total of 44 weeks

Control group:

Group 1: boceprevir‐matched placebo beginning at week 5 for a total of 44 weeks.

Co‐interventions:

Group 1 and 3: peg‐IFN alpha‐2b 1.5 μg/kg body weight subcutaneously once weekly and weight‐based oral RBV at a divided daily dose of 600 to 1400 mg for a total of 48 weeks

Group 2: peg‐IFN alpha‐2b 1.5 μg/kg body weight subcutaneously once weekly and weight‐based oral RBV at a divided daily dose of 600 to 1400 mg for 36 weeks (if HCV RNA undetectable at week 8 and 12), and for 48 weeks if (HCV RNA detectable at week 8, but undetectable at week 12).

Outcomes

Primary outcome: achievement of SVR (undetectable HCV RNA at week 24).

Secondary outcome: achievement of SVR in randomised participants who received at least 1 dose of experimental study drug or placebo. Proportion of participants with EVR (undetectable HCV RNA at week 2, 4, 8, or 12) who achieved SVR. Proportion of participants with undetectable HCV RNA at week 12. Proportion of participants with undetectable HCV RNA at 72 weeks after randomisation.

Notes

Group 2 received a similar, but not equal co‐intervention as Groups 1 and 3.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random sequence

Allocation concealment (selection bias)

Low risk

Allocation of participants through interactive voice‐response system in a 1:2:2 ratio

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

A boceprevir‐matched placebo was used.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It was not mentioned if the outcome assessors were blinded.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Treatment discontinuation due to AE was 2% to 12%. Seems no other drop‐outs occurred.

Selective reporting (reporting bias)

Low risk

A study protocol was published prior to randomisation (NCT00708500). All pre‐specified outcomes were reported on.

Vested‐interest bias

High risk

Trial was sponsored by a pharmaceutical company (Schering‐Plough/Merck). The company was directly involved in trial design and managing, data analysis, and writing of article.

Other bias

Low risk

Seems there were no other potential sources of bias.

Bacon 2011a2

Methods

For characteristics see Bacon 2011a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random sequence

Allocation concealment (selection bias)

Low risk

Allocation of participants through interactive voice‐response system in a 1:2:2 ratio

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

A boceprevir‐matched placebo was used.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It was not mentioned if the outcome assessors were blinded.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Treatment discontinuation due to AE was 2% to 12%. Seems no other drop‐outs occurred.

Selective reporting (reporting bias)

Low risk

A study protocol was published prior to randomisation (NCT00708500). All pre‐specified outcomes were reported on.

Vested‐interest bias

High risk

Trial was sponsored by a pharmaceutical company (Schering‐Plough/Merck). The company was directly involved in trial design and managing, data analysis, and writing of article.

Other bias

Low risk

Seems there were no other potential sources of bias.

Basu 2014a

Methods

Randomised clinical trial

Participants

60 adult participants

Sex: not described

Mean age: not described

Inclusion criteria: chronic hepatitis C and with a psychiatric disorder (n = 60, schizophrenia 20/60 (33.3%)), major depression 15/60 (25%), bipolar disorder 20/60 (33.3%), and prior suicidal attempts with depression 5/60 (8.3%).

Exclusion criteria: Renal failure with CrCl < 30, sickle cell, thalassaemic syndromes, haemolytic syndrome, co‐infections (HBV, HIV), or CHF NYHA Stage IV.

Interventions

Experimental group:

Group 1: simeprevir 150 mg and RBV 1000 mg daily

Group 3: simeprevir 150 mg and vitamin D 5000 mg daily.

Control group: placebo and RBV 1000 mg daily

Co‐intervention: Sofosbuvir 400 mg

Outcomes

Antiviral effect

Notes

Email was sent to Basu and colleagues on 06 June 2016 for additional information on allocation sequence generation and concealment, blinding, incomplete outcome data, protocol, full publication, study sponsor, death, SAE, SVR but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The trial is described as open‐label

Blinding of outcome assessment (detection bias)
All outcomes

High risk

The trial is described as open‐label

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It is unclear how many participants dropped out

Selective reporting (reporting bias)

Unclear risk

No protocol could be found

Vested‐interest bias

Unclear risk

It was unclear how the trial was funded

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Bavisotto 2007

Methods

Randomised clinical trial

Participants

68 participants

Sex: 20 men, 11 women (only reported in Bavisotto trial)

Mean age: 43.6 years

Country: USA

Inclusion criteria: Chronically infected with HCV genotype 1 (genotype‐1) without cirrhosis. 18‐60 years of age and HCV treatment‐naive.

Interventions

Experimental group: ascending doses of GS‐9190 (40, 120, 240, 240‐with food, or 480 mg) orally for 8 days.

Control group: placebo orally for 8 days.

Outcomes

Adverse events, GS‐9190 concentration, HCV RNA

Notes

No data could be used.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described

Selective reporting (reporting bias)

Unclear risk

No protocol could be found

Vested‐interest bias

High risk

Sponsored by Gilead

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Benhamou 2013a1

Methods

Randomised clinical trial

Participants

24 participants

Sex: 15 men, 9 women

Median age: 45.5 years

Country: France

Inclusion criteria: chronic G4 HCV infection. HCV‐infected treatment‐naive participants aged 18–65 years and in good health (except for chronic G4 HCV infection) were eligible if they had a plasma HCV RNA load of > 10,000 IU/mL, an absolute neutrophil count of ≥ 1500 neutrophils/mm3, and a platelet count of ≥ 100,000 platelets/mm3.

Exclusion criteria: contraindications to IFN (peg‐IFN in particular) or RBV treatment; history or evidence of cirrhosis, end‐stage liver disease, or decompensated liver disease (as shown by screening laboratory results); HIV or HBV co‐infection; history of alcohol or illicit drug use; and pregnancy/current breast‐feeding

Interventions

Experimental group 1: oral 750 mg of telaprevir 3 times daily for 2 weeks

Experimental group 2: oral 750 mg of telaprevir 3 times daily for 2 weeks + peg‐IFN α‐2a 180 μg once weekly, and RBV 1000–1200 mg/day (weight‐based)

Control group: placebo + peg‐IFN α‐2a 180 μg once weekly, and RBV 1000–1200 mg/day (weight‐based) for 2 weeks

Co‐intervention: after the 2 weeks of treatment, all participants received peg‐IFN α‐2a 180 μg once weekly, and RBV 1000–1200 mg/day (weight based) (48 weeks for experimental group 1, and 46 weeks for experimental group 2 and control group)

Outcomes

Efficacy assessment, virology assessment, safety and pharmacokinetic assessment

Notes

NCT00580801

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described adequately (computer‐based)

Allocation concealment (selection bias)

Unclear risk

Not described adequately (computer‐based)

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was only partially blinded. The participants in the telaprevir group without peg‐IFN and RBV were not blinded

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was only partially blinded. The participants in the telaprevir group without peg‐IFN and RBV were not blinded

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

< 5% dropped out (1 person)

Selective reporting (reporting bias)

Unclear risk

No predefined outcomes were stated in the protocol (NCT00580801)

Vested‐interest bias

High risk

The trial was funded by Janssen Pharmaceuticals and Vertex Pharmaceuticals

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Benhamou 2013a2

Methods

For characteristics see Benhamou 2013a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described adequately (computer‐based)

Allocation concealment (selection bias)

Unclear risk

Not described adequately (computer‐based)

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was only partially blinded. The participants in the telaprevir group without peg‐IFN and RBV were not blinded

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was only partially blinded. The participants in the telaprevir group without peg‐IFN and RBV were not blinded

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

< 5% dropped out (1 person)

Selective reporting (reporting bias)

Unclear risk

No predefined outcomes were stated in the protocol (NCT00580801)

Vested‐interest bias

High risk

The trial was funded by Janssen Pharmaceuticals and Vertex Pharmaceuticals.

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Boehringer Ingelheim 2010a

Methods

Randomised clinical trial

Participants

34 adult participants

Inclusion criteria: chronic HCV infection with genotype 1 (1a, 1b or mixed 1a/1b), with an HCV VL ≥ 100,000 IU/ml at screening. For treatment‐naive participants, no prior therapy with IFN, peg‐IFN, or RBV was allowed. For treatment‐experienced participants, virological failure with peg‐IFN/RBV treatment was to be confirmed. treatment‐experienced participants without cirrhosis required histological evidence within 24 months prior to trial enrolment of chronic necroinflammatory activity or the presence of fibrosis; treatment‐experienced participants with compensated cirrhosis required histological evidence of cirrhosis due to HCV infection, without evidence of decompensation.

Exclusion criteria: HCV infection of mixed genotype or had been treated previously with at least one dose of any protease.

Interventions

Experimental group: oral BI 201335 NA, 20 mg, 48 mg, 120 mg, or 240 mg once daily.

Control group: placebo.

Co‐intervention: peg‐IFN/RBV.

Outcomes

Virological response, pharmacokinetics, safety

Notes

Unpublished data only

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as being blinded, but it was unclear how this was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as being blinded, but it was unclear how this was performed

Incomplete outcome data (attrition bias)
All outcomes

Low risk

< 5% dropped out

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

High risk

The trial was funded by Boehringer Ingelheim

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Boehringer Ingelheim 2010b

Methods

Randomised clinical trial

Participants

10 adult participants

Inclusion criteria: with diagnosis of cirrhosis and chronic HCV (genotype 1) infection with a VL greater than 50,000 copies mRNA/ml serum.

Country: Germany

Interventions

Experimental group: oral 200 mg twice daily for 2 days.

Control group: placebo.

Outcomes

Efficacy assessment, safety assessment

Notes

Unpublished data only

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Low risk

There were no dropouts

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

High risk

The trial was funded by Boehringer Ingelheim

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Bronowicki 2013a1

Methods

Randomised clinical trial

Participants

48 adult participants

Sex: 35 men, 12 women

Mean age: 48.5 years

Inclusion criteria: 18‐70 years old with Chronic HCV genotype 1 infection and HCV RNA above 100,000 IU/mL. Participants had no prior treatment or < 4 weeks of total exposure to RBV or peg‐IFN‐based therapy. Participants had to be non‐cirrhotic, documented by liver biopsy obtained within 24 months before randomisation.

Exclusion criteria: advanced liver disease, co‐infection with HBC or HIV, hepatocellular carcinoma, other clinical relevant comorbidity. ALT > 5 x the ULN, total bilirubin > 2 mL/dL, albumin < 3.5 g/dL, international normalised ratio > 1.7, platelets < 90x10^9, haemoglobin < 12 g/dL (women) or < 13 g/dL (men).

Interventions

Experimental group 1: oral asunaprevir (200 mg) twice daily for 48 weeks.

Experimental group 2: oral asunaprevir (600 mg) twice daily for 48 weeks.

Experimental group 3: oral asunaprevir (600 mg) once daily for 48 weeks.

Control group: placebo 48 weeks.

Co‐intervention: peg‐IFN (subcutaneously at 180 µg/week) and RBV orally twice daily dosed according to body weight.

Outcomes

Proportion of participants with undetectable HCV RNA at week 4 and 12, SAE, AE, mortality, sustained virological response.

Notes

Experimental group 1 vs control. We contacted trial authors on 20 April 2016 for additional information on allocation concealment, specifics of the blinding, what SAE were experienced, and how they dealt with missing data, reached required sample size but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random allocation sequence

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

It was stated that investigators and participants were blinded to treatment assignment throughout the study but it was not stated how the blinding was maintained.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

The sponsor was blinded to treatment assignment until the primary end point analysis which was at 12 weeks and we used data at week 24.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was unclear how many participants had missing data and how the trial handled participants with missing data

Selective reporting (reporting bias)

Low risk

All outcomes stated in the pre published protocol (NCT01030432) were reported

Vested‐interest bias

High risk

The study was funded by Bristol‐Myers Squibb

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Bronowicki 2013a2

Methods

For characteristics see Bronowicki 2013a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random allocation sequence

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

It was stated that "Investigators and participants were blinded to treatment assignment throughout the study" but it was not stated how the blinding was maintained.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

The sponsor was blinded to treatment assignment until the primary end point analysis which was at 12 weeks and we used data at week 24.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was unclear how many participants had missing data and how the trial handled participants with missing data

Selective reporting (reporting bias)

Low risk

All outcomes stated in the pre published protocol (NCT01030432) were reported

Vested‐interest bias

High risk

The study was funded by Bristol‐Myers Squibb

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Bronowicki 2013a3

Methods

For characteristics see Bronowicki 2013a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random allocation sequence

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

It was stated that investigators and participants were blinded to treatment assignment throughout the study but it was not stated how the blinding was maintained.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

The sponsor was blinded to treatment assignment until the primary end point analysis which was at 12 weeks and we used data at week 24.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was unclear how many participants had missing data and how the trial handled participants with missing data

Selective reporting (reporting bias)

Low risk

All outcomes stated in the pre published protocol (NCT01030432) were reported

Vested‐interest bias

High risk

The study was funded by Bristol Myers Squibb

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Bronowicki 2014

Methods

Randomised clinical trial

Participants

238 adult participants

Sex: 153 men, 85 women

Mean age: 47.7 years

Inclusion criteria: 18–70 years, with chronic HCV genotype 1 or 4 infection and HCV RNA P100,000 IU/mL. Participants had to have ALT < 5 ULN and no history or evidence of hepatic decompensation. Compensated cirrhotic participants (genotype 1 only) were eligible with a liver biopsy documenting cirrhosis from any period prior to randomisation. For non‐cirrhotic participants, absence of cirrhosis had to be documented by a liver biopsy obtained within 24 months pre‐randomisation.

Exclusion criteria: prior exposure to anti‐HCV agents, co‐infection with HBV or HIV, and chronic liver disease other than HCV.

Interventions

Experimental group: 200 mg asunaprevir twice a day for 12 or 24 weeks.

Control intervention: placebo twice a day for 12 weeks.

Co‐intervention: peg‐IFNa‐2a administered subcutaneously at 180 lg per week, and oral RBV twice a day dosed by body weight (< 75 kg, 1000 mg daily; P75 kg, 1200 mg daily).

Outcomes

SAE, AEs, discontinuations due to AEs, eRVR at week 4 and 12, SVR24, RVR at week 4, complete eRVR, SVR 12, resistant variants associated with virologic failure.

Notes

At week 12, asunaprevir‐treated participants who achieved a protocol‐defined response (HCV RNA < LLOQ at week 4 and undetectable at week 10 were re‐randomised (1:1) to continue triple therapy with asunaprevir plus peg‐IFNα/RBV for a total of 24 weeks (24‐Triple) or to receive placebo plus peg‐IFNα/RBV for an additional 12 weeks (12‐Triple + 12; Fig. 1). Asunaprevir‐treated participants without PDR and those initially assigned to placebo received placebo plus peg‐IFNα/RBV from week 13 to 24. At week 24, PDR‐positive participants who received 24‐Triple or 12‐Triple + 12 stopped treatment and were followed through week 48. PDR‐negative participants and those initially assigned to placebo were switched to open‐label peg‐IFNα/RBV through week 48 and followed through week 72.

We report re‐randomisation in Bronowicki 2014a.

We contacted the trial authors on 25 February 2016 by email jp.bronowicki@chu‐nancy.fr about allocation concealment, SAE at maximum follow‐up, specific SAE at maximum follow‐up, how the authors accounted for data of missing participants.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random allocation sequence

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Investigators and participants were blinded to treatment assignment through week 24; the sponsor was blinded

through week 12. It was unknown how the blinding was maintained. Additionally, some of the participants had open‐label peg‐IFNα/RBV

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Investigators and participants were blinded to treatment assignment through week 24; the sponsor was blinded through week 12. It was unknown how the blinding was maintained. Additionally, some of the participants had open‐label peg‐IFNα/RBV

Incomplete outcome data (attrition bias)
All outcomes

Low risk

There were under 5% dropouts

Selective reporting (reporting bias)

Low risk

The outcomes stated in the pre published protocol (ClinicalTrials.gov:NCT01030432) were reported on

Vested‐interest bias

High risk

Funded by Bristol‐Myers Squibb

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

C‐EDGE CO STAR 2015

Methods

Randomised clinical trial

Participants

301 participants

Sex: 228 men and 73 women

Mean age: 47 years

Inclusion criteria: documented chronic HCV genotype 1 (genotype1), genotype4, or genotype6 infection with no evidence of genotype2 or genotype3 or non‐typeable genotypes and HCV RNA confirmed by screening lab results prior to randomisation on opiate substitution therapy (methadone, levamethadone, buprenorphine, naloxone, naltrexone) for at least 3 months prior to screening, treatment‐naive to all HCV therapies. HIV‐infected participants enrolled in this study had to meet following criteria: Documented HIV infection, naive to treatment with any antiretroviral therapy or on HIV antiretroviral therapy for at least 8 weeks prior to study entry using a dual nucleoside reverse transcriptase inhibitor backbone of tenofovir or abacavir and either emtricitabine or lamivudine plus raltegravir (or dolutegravir or rilpivirine). Dose modifications or changes in antiretroviral therapy during the 4 weeks prior to study entry (Day 1) were not permitted. Cluster of differentiation 4 (CD4+) T‐cell count > 200 cells/mm^3 if on antiretroviral therapy or > 500 cell/mm^3 if antiretroviral therapy treatment‐naive undetectable plasma HIV‐1 RNA at least 8 weeks prior to screening if on antiretroviral therapy or < 50,000 copies/mL if antiretroviral therapy treatment‐naive. Participants with HIV‐1 infection and on antiretroviral therapy must have at least 1 viable antiretroviral regimen alternative beyond their current regimen in the event of HIV virologic failure or the development of anti‐retroviral drug resistance Women who are of reproductive potential had to agree to avoid becoming pregnant while receiving study drug and for 14 days after the last dose of study drug by complying with 1 of the following: (1) practice abstinence from heterosexual activity OR (2) use (or have her partner use) acceptable contraception during heterosexual activity.

Exclusion criteria: evidence of decompensated liver disease. For participants with cirrhosis, participants who are Child‐Pugh Class B or C or who have a Pugh‐Turcotte score > 6 Is co‐infected with HBV. Has cirrhosis and liver imaging within 6 months of Day 1 showing evidence of HCC or is under evaluation for HCC. Currently using or intends to use barbiturates during the treatment period of this study. Is a female and is pregnant or breast‐feeding, or expecting to conceive or donate eggs from Day 1 or anytime during treatment, and 14 days after the last dose of study medication, or longer if dictated by local regulations. Any medical condition requiring or likely to require chronic systemic administration of corticosteroids, Tumor Necrosis Factor‐antagonists, or other immunosuppressant drugs during the course of the trial. Evidence or history of chronic hepatitis not caused by HCV.

Interventions

Experimental group: oral 100 mg of grazoprevir and 50 mg of elbasvir for 12 weeks.

Control group: placebo.

Outcomes

Safety assessment, HCV RNA (virological failure).

Notes

Abstract only (still ongoing). Only data for the first 12 weeks could be used, since the control group received the same DAA in the following 12 weeks. (NCT02105688).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No description of dropouts after 12 weeks for the control group.

Selective reporting (reporting bias)

Unclear risk

Only an abstract could be found, and no data on SVR12 and SVR24 were presented. However the trial was still stated as ongoing. (NCT02105688)

Vested‐interest bias

High risk

The trial was funded by Merck

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

C‐EDGE TN 2015

Methods

Randomised clinical trial

Participants

421 participants

Sex: 227 men, 194 women

Mean age: 52.6 years

Countries: Australia, Czech Republic, France, Germany, Israel, Puerto Rico, South Korea, Sweden, Taiwan, and USA

Inclusion criteria: treatment‐naive cirrhotic and non‐cirrhotic adults (aged > 18 years) with HCV RNA levels > 104 IU/mL were eligible. Hepatic fibrosis was staged by biopsy or noninvasive assessment.

Exclusion criteria: decompensated liver disease, HCC, HIV or HBV co‐infection, uncontrolled diabetes mellitus (haemoglobin A1c level > 10%), elevated prothrombin time unrelated to anticoagulation, creatinine clearance < 50 mL/min, haemoglobin level < 95 g/L, thrombocytopenia (platelet count < 50 × 109 cells/L), aminotransferase levels more than 10 times the ULN, or hypoalbuminaemia (albumin level < 30 g/L).

Interventions

Experimental group: oral 100 mg of grazoprevir and 50 mg of elbasvir for 12 weeks.

Control group: placebo.

Outcomes

HCV RNA, safety assessment.

Notes

Only data for the first 12 weeks could be used, since the control group received the same DAA in the following 12 weeks. We emailed Zeuzem and colleagues on 27 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The trial used a "computer‐generated random allocation schedule"

Allocation concealment (selection bias)

Low risk

The trial used a "central interactive voice‐response system"

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The participants and personnel were blinded to treatment assignment for the first 12 weeks (and we used the data from this time point)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The sponsors performing the analyses were blinded to treatment assignment for the first 12 weeks (and we used the data from this time point)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

< 5% dropped out after 12 weeks

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol were assessed (NC02105467)

Vested‐interest bias

High risk

The trial was funded by Merck‐Sharp which performed the analyses

Other bias

Low risk

Trial seems to be free of other potential sources of bias

Chandra 2006a

Methods

Randomised clinical trial

Participants

An unknown amount of participants

Sex: unknown

Mean age: unknown

Inclusion criteria: chronic HCV infection (> 6 months) and were treatment‐naive. Participants aged 18‐64 years with ≥ 104 IU/mL HCV RNA levels were enrolled in sequential, ascending dose cohorts of up to 16 participants (12 active, 4 placebo) per cohort.

Interventions

Experimental group: participants received 50, 100, 250, 500, 1000, or 1500 mg oral doses of HCV‐796 or placebo given as monotherapy twice daily.

Control group: placebo twice a day.

Co‐intervention: none.

Outcomes

Most frequent AE, dose‐limiting toxicities or serious treatment‐emergent AEs, PK parameters, maximal antiviral effects.

Notes

The authors were contacted on VIROPHARMA all bias domains, mortality, SAE, SVR24. mean age, male:female, number of participants, final publication.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

The number of participants with incomplete data was not described

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

Unclear risk

It was unclear how the trial was funded

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

COMMAND‐1 2015a1

Methods

Randomised clinical phase IIb trial

Participants

395 participants

Sex: 262 men, 133 women

Mean age: 50.8 years

Countries: North and Central America, Australia, North Africa, and Europe.

Inclusion criteria: treatment‐naive, aged 18–70 years who had chronic HCV genotype 1 or 4 infection. Compensated cirrhotic, infected with HCV genotype 1, and HCV genotype 4, were each capped at 10% of randomised participants. Cirrhosis was confirmed by biopsy at any time prior to randomisation. For non‐cirrhotic participants, a liver biopsy must have been obtained within 24 months prior to randomisation. Additional inclusion criteria included HCV RNA ≥ 100,000 IU/mL and ALT levels < 5×ULN.

Exclusion criteria: history or evidence of hepatic decompensation, prior exposure to any agent with potential anti‐HCV activity, co‐infection with HBV or HIV, or evidence of chronic liver disease other than HCV.

Interventions

Initally the trial was randomised into 3 groups (2 experimental groups, and 1 control group). After week 12, the participants who received a protocol‐defined response, were re‐randomised to placebo or additional 12 weeks of therapy. The participants without a protocol‐defined response were treated with placebo and co‐intervention.

Experimental group: oral 20 mg of daclatasvir once a day for 12 weeks (after week 12, the participants with a protocol‐defined response were re‐randomised).

Experimental group: oral 60 mg of daclatasvir once a day for 12 weeks (after week 12, the participants with a protocol‐defined response were re‐randomised).

Control group: placebo.

Co‐intervention: peg‐IFN α‐2a administered subcutaneously at a dose of 180 mg per week and twice a day RBV dosed orally according to body weight (< 75 kg, 1000 mg daily; > 75 kg, 1200 mg daily). After week 24, all participants received standard care (peg‐IFN‐α‐2a and RBV)

Outcomes

Safety assessment, efficacy assessment

Notes

We emailedWe emailed Hezode and colleagues on 21 April 2016 for additional information on sequence generation, missing data, additional data, death but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Low risk

The trial used interactive voice‐response system

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The participants were only blinded until week 24

Blinding of outcome assessment (detection bias)
All outcomes

High risk

The sponsors, who performed the analyses, were only blinded until week 12

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol were assessed

Vested‐interest bias

High risk

The trial was funded by Bristol‐Myers Squibb.

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

COMMAND‐1 2015a2

Methods

For characteristics see COMMAND‐1 2015a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Low risk

The trial used interactive voice‐response system

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The participants were only blinded until week 24

Blinding of outcome assessment (detection bias)
All outcomes

High risk

The sponsors, who performed the analyses, were only blinded until week 12

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol were assessed

Vested‐interest bias

High risk

The trial was funded by Bristol‐Myers Squibb.

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

CONCERTO‐1 2015

Methods

Randomised phase III clinical trial

Participants

188 participants

Inclusion criteria: HCV genotype 1 infection, treatment‐naive male and female participants aged 20–70 years with documented chronic genotype 1 HCV infection and plasma HCV RNA P5.0 log10 IU/mL at screening.

Exclusion criteria: liver cirrhosis, hepatic failure, any other liver disease of non‐HCV etiology and co‐infection with HIV‐1, HIV‐2, hepatitis B, or non‐genotype 1 HCV.

Interventions

Experimental group: simeprevir 100 mg once a day plus peg‐IFNa‐2a/RBV for 12 weeks followed by response‐guided therapy with peg‐IFNa‐2a/RBV alone for 12 or 36 weeks.

Control group: placebo with peg‐IFNa‐2a/RBV for 12 weeks followed by peg‐IFNa‐2a/RBV for 36 weeks. Peg‐IFNα‐2a (Pegasys®, Chugai, Japan) was administered as a subcutaneous injection (180 μg once weekly) and RBV (Copegus®, Chugai) as oral tablets (600‐1000 mg total daily dose, depending on body weight).

Co‐intervention: peg‐IFN (subcutaneously at 180 µg/week) and RBV orally twice daily dosed according to body weight.

Outcomes

HCV RNA, safety assessment, ALT/AST elevations.

Notes

We emailedWe emailed Hayashi and colleagues on 21 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as double‐blinded but it was unclear how the blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as double‐blinded but it was unclear how the blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol were assessed (NCT01292239)

Vested‐interest bias

High risk

The trial was funded by Bristol‐Myers Squibb

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Cooper 2009

Methods

Randomised clinical trial

Participants

34 participants

Sex: 21 men, 10 women (analysed only)

Mean age: 42.9 years (analysed only)

Inclusion criteria: treatment‐naive genotype 1‐infected male or female participants between 18 and 60 years of age with a BMI 633 kg/m2 were recruited. Baseline plasma HCV RNA greater than 100,000 IU/mL, ALT values < 5 times the ULN and a Metavir liver fibrosis stage between 0 and 3 were required.

Exclusion criteria: none specified.

Interventions

Experimental group: VCH‐759 doses (400 mg three times a day, 800 mg twice a day and 800 mg three times a day). VCH‐759 was supplied as an oral solution formulation in individual 120 mL clear glass bottles. The oral solution was reconstituted by combining the appropriate VCH‐759 powder‐in‐bottle dose in a 30% polyethylene glycol 400/15% Solutol HS15 aqueous reconstitution vehicle (20 mL for the 400 mg dose and 40 mL for the 800 mg dose).

Control group: placebo.

Co‐intervention: none.

Outcomes

Absolute change in plasma HCV RNA levels between baseline to nadir, blood samples for evaluation of the plasma HCV RNA viral load, blood samples for NS5B polymerase, the complete PK profile.

Notes

We contacted the trial authors on 26 February 2016 by email [email protected] about random sequence generation, allocation concealment, blinding of participants, personnel and outcome assessment, did the trial account for the missing data, which group the the 2 participants dropped out from and was if there was a prepublished protocol, mortality, SAE, SVR24.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The study was described as double‐blinded but it was unclear how the blinding was maintained.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The study was described as double‐blinded but it was unclear how the blinding was maintained and who performed the outcome assessment.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There were above 5% drop outs and it was unclear how the trial handled participants with missing data

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

High risk

The authors have declared that this study was funded by ViroChem Pharma Inc. JB, NC, RT, IB, ON, and LP are employees of ViroChem Pharma Inc. The other authors have also declared a relationship with the manufacturers of the drugs involved.

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Dauphine 2015a1

Methods

Randomised, active‐controlled phase IIb trial

Participants

Sex: 260 men, 157 women

Mean age: 48 years

Inclusion criteria: eligible participants were treatment‐naive adults aged > 18 years with chronic HCV genotype 1 or 4 infection and HCV RNA above 50,000 IU/mL. Participants had to have evidence of chronic hepatitis C, documented by liver biopsy obtained within 24 months before randomisation.

Exclusion criteria: participants with cirrhosis or incomplete/transition to cirrhosis (Knodell, Metavir, or Batts and Ludwig ≥ 3 or Ishak modified HAI ≥ 4); BMI < 18 or ≥ 36 kg/m2, other forms of liver disease; HIV infection; HCC; severe cardiac disease; severe depression or other psychiatric disease; renal disease; uncontrolled seizure disorders; severe retinopathy; haemoglobin < 12 g/dL for women or < 13 g/dL for men; neutrophil count < 90 cells/nL; serum creatinine > 1.5 times the ULN.

Interventions

Participants were randomised (2:2:2:2:1) to 1 of 5 treatment arms

Experimental group 1: ritonavir boosted danoprevir (danoprevir/r) 200/100 mg twice a day for 24 weeks

Experimental group 2: ritonavir boosted danoprevir (danoprevir/r) 100/100 mg twice a day for 24 weeks

Experimental group 3: ritonavir boosted danoprevir (danoprevir/r) 50/100 mg twice a day for 24 weeks

Experimental group 4: ritonavir boosted danoprevir (danoprevir/r) 100/100 mg twice a day for 12 weeks or 24 weeks (participants achieving undetectable HCV RNA from Weeks 2 to 10 (eRVR2) stopped treatment at Week 12)

Control group: participants in Arm E with detectable HCV RNA at Week 12 had the option to roll over to treatment with danoprevir/r 200/100 mg twice a day

Co‐intervention: peg‐IFN α‐2a (40KD) 180 lg/week and RBV 1000 mg/day (bodyweight < 75 kg) or 1200 mg/day (bodyweight ≥ 75 kg)

Outcomes

Proportion of participants with SVR24, with SAE, AEs, mortality.

Notes

Experimental group 1 vs Control.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Unblinded

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Unblinded

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There were above 5% dropouts and it was unclear how the trial handled the missing participants

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol were reported on: NCT01220947

Vested‐interest bias

High risk

The trial was funded by F. Hoffmann‐La Roche Ltd

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Dauphine 2015a2

Methods

For characteristics see Dauphine 2015a2

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Unblinded

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Unblinded

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There were above 5% dropouts and it was unclear how the trial handled the missing participants

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol were reported on: NCT01220947

Vested‐interest bias

High risk

The trial was funded by F. Hoffmann‐La Roche Ltd

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Dauphine 2015a3

Methods

For characteristics see Dauphine 2015a2

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

computer‐generated randomisation

Allocation concealment (selection bias)

Unclear risk

not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

unblinded

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

unblinded

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There were above 5 % drop outs and it was unclear how the trial handLed the missing participants

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol were reported on: NCT01220947

Vested‐interest bias

High risk

the trial was funded by F. Hoffmann‐La Roche Ltd. Support

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Dauphine 2015a4

Methods

For characteristics see Dauphine 2015a2

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Unblinded

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Unblinded

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There were above 5 % drop outs and it was unclear how the trial handLed the missing participants

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol were reported on: NCT01220947

Vested‐interest bias

High risk

The trial was funded by F. Hoffmann‐La Roche Ltd

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

De Bruijne 2010a1

Methods

Randomised clinical trial

Participants

41 participants

Sex: 31 men, 9 women

Mean age: 48.8 years

Inclusion criteria: 18‐65 years with BMI of 18‐40 kg/m2, HCV genotype 1 (any subtype), and HCV RNA level > 1 105 copies/mL (or equivalent international units). Chronic hepatitis C participants were naive, nonresponders or relapsers to previous IFN‐based treatment. Relapse was defined as undetectable HCV RNA upon completion of a previous IFN‐based treatment, but positive HCV RNA during follow‐up. Nonresponse was defined as positive HCV RNA at the end of a previous IFN‐based treatment or < 2‐log decline in HCV RNA levels at 12 weeks and discontinued treatment.

Exclusion criteria: key exclusion criteria included decompensated liver disease, findings consistent with Child‐Pugh class B or C liver cirrhosis, and co‐infection with HIV or HBV.

Interventions

Experimental group: participants received either 800 mg narlaprevir 3 times daily or 400 mg narlaprevir as an oral suspension in combination with for 7 days in the first period and for 14 days in the second period.

Control group: placebo.

Co‐intervention: 200 mg ritonavir in cohort 3 and 4, a wash‐out period after 1 week of treatment, 1.5 lg/kg/week peg‐IFN‐α‐2b (in period 2) and standard care for 24 weeks after period 2.

Outcomes

Safety assessment, pharmacokinetic assessment, viral assessments.

Notes

Cohort 1 and 3 each included 10 participants naive to HCV treatment; cohorts 2 and 4 each included 10 HCV treatment‐experienced participants. We report here the treatment‐naive participants

We contacted the trial authors on 26 February 2016 by email [email protected] about allocation concealment, how the blinding was maintained and who performed the outcome assessment; number of deaths, SAE, which group was the missing participants randomised to.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random code

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The study was described as double‐blinded but it was unclear how the blinding was maintained

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The study was described as double‐blinded but it was unclear how the blinding was maintained and who performed the outcome assessment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 1 participant dropped out

Selective reporting (reporting bias)

Low risk

A protocol was found (NCT01081158) and the outcomes stated in the protocol were reported on

Vested‐interest bias

High risk

Sponsored by Schering‐Plough and designed by Schering‐Plough employees and HW Reesink

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias.

De Bruijne 2010a2

Methods

For characteristics see De Bruijne 2010a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random code

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The study was described as double‐blinded and but it was unclear how the blinding was maintained

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The study was described as double‐blinded but it was unclear how the blinding was maintained and who performed the outcome assessment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 1 participant dropped out

Selective reporting (reporting bias)

Low risk

A protocol was found (NCT01081158) and the outcomes stated in the protocol are reported on

Vested‐interest bias

High risk

Sponsored by Schering‐Plough and designed by Schering‐Plough employees and HW Reesink

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of other bias.

Detishin 2011

Methods

Randomised clinical trial

Participants

18 participants (only number of experimental group)

Country: Moldova

Inclusion criteria: healthy, treatment‐naive or experienced HCV genotype 1 participants.

Interventions

Experimental group: oral 400 mg or 600 mg of ACH‐1625 in fasted state for 5 days, or 600 mg of ACH‐1625 once daily following a medium‐fat meal for 5 days.

Control group: placebo.

Outcomes

PK, safety, tolerability, effects on viral kinetics.

Notes

It was unclear whether the included participants included healthy participants, or healthy HCV genotype 1 participants.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as being placebo blinded, but it was unclear how the blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as being placebo blinded, but it was unclear how the blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

High risk

Several authors were sponsored by Achillion Pharmaceuticals

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Dore 2015a1

Methods

Randomised clinical trial

Participants

152 adult participants

Sex: 96 men, 55 women (analysed)

Mean age: 47.9 years

Countries: USA, Australia, Canada, Denmark, France, and Italy.

Inclusion criteria: men and women aged 18–70 years, with chronic HCV genotype 2 or 3 infection and no prior exposure to HCV therapeutic agents including DAA, IFN preparations, or RBV. Participants were stratified by HCV genotype (2 or 3) before randomisation. Plasma HCV RNA levels at screening were required to be ≥100,000 IU/mL. Liver disease staging was conducted by liver biopsy within 2 years of screening (biopsies confirming cirrhosis), or by FibroScan (Echosens, Paris, France) within 1 year of screening (14.6 kPa was considered consistent with cirrhosis); participants with compensated cirrhosis were capped at approximately 10% of the study population. Women of childbearing potential and men who were sexually active partners of women of childbearing potential were required to use 2 forms of contraception, including at least 1 barrier method.

Exclusion criteria: history or evidence of HCC, decompensated cirrhosis, or chronic liver disease other than hepatitis C; history of cancer within 5 years of enrolment; chronic HBV or HIV infection; presence of any other medical, psychiatric, and/or social reason that would render the patient inappropriate for study participation; gastrointestinal disease or surgical procedure that may impact absorption of the study drug; medical conditions prohibiting use of peg‐α‐2a or RBV, based on their respective product labels; or a history of hypersensitivity to compounds related to NS5A inhibitors. Exclusionary laboratory parameters included ALT level of 5 or more times the ULN; total bilirubin level of ≥ 2 mg/dL; international normalizsed ratio of ≥ 1.7; albumin level of ≤ 3.5 g/dL; haemoglobin level of ≤ 12 g/dL (for women) or ≤ 13 g/dL (for men); absolute neutrophil count of ≤ 1.5 109 cells/L (1.2 109 cells/L for black participants); platelet count of ≤ 90 109 cells/L; creatinine clearance of ≤ 50 mL/min; a fetoprotein level > 100 ng/mL; and corrected QT interval (QTcF) > 450 ms (for men) or > 470 ms (for women). Prohibited concomitant medications included inducers or strong or moderate inhibitors of CYP3A4; P‐glycoprotein substrates with a narrow therapeutic index; strong P‐glycoprotein inhibitors; nonstudy medications with known or potential anti‐ HCV activity; or any prescription or herbal product not prescribed for the treatment of a specific clinical condition. Doses of concomitant medications were required to be stable for 4 weeks or longer before the first dose of study drug.

Interventions

Experimental group: oral 60 mg of daclatasvir for 12 or 16 weeks.

Control group: placebo for 24 weeks.

Co‐intervention: all participants received antiviral combination therapy with peg‐α‐2a 180 mg weekly, RBV 400 mg twice daily (800 mg/day).

Outcomes

Virological response, safety assessment.

Notes

NCT01257204

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Interactive voice‐response system

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The participants were only blinded during treatment period

Blinding of outcome assessment (detection bias)
All outcomes

High risk

The study sponsor, who performed the analyses, were only blinded for the first 16 weeks

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out

Selective reporting (reporting bias)

High risk

The trial changed the secondary outcomes

Vested‐interest bias

High risk

The trial was funded by Bristol‐Myers Squibb

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Dore 2015a2

Methods

For characteristics see Dore 2015a2

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Interactive voice‐response system

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The participants were only blinded during treatment period

Blinding of outcome assessment (detection bias)
All outcomes

High risk

The study sponsor, who performed the analyses, were only blinded for the first 16 weeks

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out

Selective reporting (reporting bias)

High risk

The trial changed the secondary outcomes

Vested‐interest bias

High risk

The trial was funded by Bristol‐Myers Squibb

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

DRAGON 2014a1

Methods

Multicenter, randomised, open‐label, parallel‐group comparison trial (ClinicalTrials.gov number, NCT00996476)

Participants

93 were randomised to treatment groups, of whom 92 received at least 1 dose of the study drug

Mean age: 54 years

Sex: 43 men, 49 women

Inclusion criteria: eligible participants were treatment‐naive, chronically infected with genotype 1 HCV, aged 20–70 years and had plasma levels of HCV RNA > 5.0 log 10 IU/mL at screening

Exclusion criteria:

  1. presence of liver cirrhosis or hepatic failure, or other liver disease

  2. infection/co‐infection with HIV‐1, HIV‐2, hepatitis B or nongenotype 1 HCV

  3. malignant tumor within 5 years prior to study

  4. HCC

  5. meeting conditions that required caution with peg‐IFN α‐2a or RBV treatment

  6. any clinically significant disease

  7. organ transplant

  8. defined laboratory abnormalities during screening.

Interventions

Eligible participants were randomised to 1 of 5 treatment groups in a 2:1:2:1:1 ratio.
Experimental group 1: simeprevir 50 mg once a day for 12 weeks.
Experimental group 2: simeprevir 50 mg once a day for 24 weeks.
Experimental group 3: simeprevir 100 mg once a day for 12 weeks.
Experimental group 4: simeprevir 100 mg once a day for 24 weeks.
NOTE: In these 4 groups, at week 24, participants either stopped or continued treatment with peg‐IFN α‐2a/RBV up to week 48, according to response‐guided therapy criteria (stop treatment if plasma HCV RNA \1.4 log10 IU/mL at week 4 and undetectable at weeks 12, 16 and 20, otherwise continuing peg‐IFN α‐2a/RBV to week 48). In the PR48 group, criteria were not applied; participants received peg‐IFNα‐2a/RBV for 48 weeks.
Control group: peg‐IFN α‐2a/RBV for additional 24 weeks (48 weeks PR treatment in total).
Co‐intervention: peg‐IFN α‐2a/RBV for 24 weeks.

Outcomes

Proportion of participants with undetectable plasma HCV RNA 24 weeks after the end of treatment (SVR24), with SAE, AEs, mortality

Notes

According to predefined virologic stopping rules, participants in the simeprevir groups discontinued simeprevir and continued peg‐IFN α‐2a/RBV if viral breakthrough occurred during the first 24 weeks, and stopped all treatment if the decrease in plasma HCV RNA from baseline to week 12 was < 2 log10 IU/mL, or plasma HCV RNA level at week 24 was > = 1.2 log10 IU/mL.

In this review SVR24 rates in the experimental group were analysed only from participants who did not continue treatment after 24 weeks.

This is Group 1 vs control.

We emailed Hayashi and colleagues on 21 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not reported

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Unblinded study

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Unblinded study

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was unclear how many participants dropped out

Selective reporting (reporting bias)

Low risk

The outcomes stated in the protocol were reported on

Vested‐interest bias

High risk

Janssen Pharmaceutical KK

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

DRAGON 2014a2

Methods

For characteristics see DRAGON 2014a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not reported

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Unblinded study

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Unblinded study

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was unclear how many participants dropped out

Selective reporting (reporting bias)

Low risk

The outcomes stated in the protocol were reported on

Vested‐interest bias

High risk

Janssen Pharmaceutical K.K

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

DRAGON 2014a3

Methods

For characteristics see DRAGON 2014a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not reported

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Unblinded study

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Unblinded study

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was unclear how many participants dropped out

Selective reporting (reporting bias)

Low risk

The outcomes stated in the protocol were reported on

Vested‐interest bias

High risk

Janssen Pharmaceutical KK

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

DRAGON 2014a4

Methods

For characteristics see DRAGON 2014a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not reported

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Unblinded study

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Unblinded study

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was unclear how many participants dropped out

Selective reporting (reporting bias)

Low risk

The outcomes stated in the protocol were reported on

Vested‐interest bias

High risk

Janssen Pharmaceutical KK

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Erhardt 2009

Methods

Randomised clinical trial

Participants

96 adult men

Sex: 96 men

Mean age: 44.6 years

Country: Germany, Spain, and France

Inclusion criteria: chronic HCV genotype 1 with minimal to mild liver fibrosis (Ishak score or Metavir grade < 2, confirmed by liver biopsy within the past 24 months) and HCV RNA viral load > 100.000 IU/mL at screening. No restriction was on the basis of prior IFN treatment experience.

Exclusion criteria: laboratory measurements, HIV, HBV, any other additional cause for chronic liver disease, concurrent disease requiring treatment, any use of co‐medication, treatment with IFN and/or RBV within 6 months prior to screening and use of any investigational drug 30 days prior to screening or 5 periods of drug plasma half life.

Interventions

Trial was divided into 8 cohorts and randomised in these cohorts.
Experimental group: oral 10, 20, 40, 60, 80, 100, 150, 200, 300, 450 mg BILB‐1941 three times a day for 4 days, plus a morning dose on 5th day.

Control group: placebo.

Outcomes

Antiviral response, pharmacokinetics, safety assessment.

Notes

We emailed Erhardt and colleagues on 20 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

High risk

The trial was funded by Boehringer‐Ingelheim

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Feld 2014

Methods

Multicenter, randomised, double‐blind, placebo‐controlled, parallel‐design trial (SAPPHIRE‐I) (NCT01716585)

Participants

631 participants

Location: USA, Europe and Australia

Inclusion criteria: age 18‐70 years, chronic hepatitis C infection, genotype 1, HCV RNA level > 10,000 IU/mL, treatment‐naive, no evidence of liver cirrhosis, women had to be post‐menopausal for at least 2 years or surgically sterile or practicing specific forms of birth control.

Exclusion criteria: hepatitis B or HIV co‐infection, positive screen for drugs or alcohol, significant sensitivity to any drug, use of contraindicated medications within 2 weeks of dosing, certain predefined abnormal laboratory tests.

Group A: 473 participants

Sex: 217 men, 256 women

Mean age, years (range): 49.4(18.0‐70.0)

Race, n(%): white: 428(90.5), black: 26(5.5), other: 19(4.0)

Fibrosis score ≥ 2, n(%): 110(23.3), IL28B CC genotype, n(%): 144(30.4), HCV genotype, n(%): 1a: 322(68.1), 1b: 151(31.9)

Group B: 158 participants

Sex: 73 men, 85 women

Mean age, years (range): 51.2(21.0‐70.0)

Race, n(%): white: 144(91.1), black: 8(5.1), other: 6(3.8)

Fibrosis score ≥ 2, n(%): 42(26.6), IL28B CC genotype, n(%): 50(31.6), HCV genotype, n(%): 1a: 105(66.5), 1b: 53(33.5).

Interventions

Experimental group: ABT‐450 orally at once‐daily dose of 150 mg with ritonavir 100 mg once daily and ombitasvir orally 25 mg once daily for 12 weeks. Dasabuvir orally at a dose of 250 mg twice daily for 12 weeks.

Control group: matching placebos for 12 weeks, followed by an open‐label period of 12 weeks' administration of the active treatment.

Co‐interventions: weight‐based oral RBV 1000 to 1200 mg in 2 divided doses (1000 mg daily if body weight was < 75 kg, and 1200 mg daily if body weight was ≥ 75 kg).

Outcomes

Primary outcomes: percentage of participants achieving SVR 12 weeks after treatment. Safety of ABT‐450/r/ombitasvir and dasabuvir co‐administered with RBV for 12 weeks.

Secondary outcomes: percentage of participants achieving RVR. Percentage of participants achieving end of treatment response. Percentage of participants with ALT normalisation at end of treatment.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated schedules

Allocation concealment (selection bias)

Low risk

All participants assigned a unique participant number through the use of interactive response system in order to receive a unique study drug bottle/kit numbers and a unique randomisation number

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Matching placebos were used

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

All data were blinded to participants, study personnel, and sponsor. An independent data and safety monitoring committee reviewed safety data.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Number and reasons for withdrawal and discontinuation were clearly stated.

Selective reporting (reporting bias)

Low risk

A protocol was published before randomisation and all pre‐specified outcomes were reported on.

Vested‐interest bias

High risk

The sponsor (AbbVie) was directly involved in trial design, data analysis, drafting the manuscript and publication.

Other bias

Low risk

Seems free of other potential sources of bias.

Feld 2015

Methods

Randomised clinical trial

Participants

701 adult participants

Sex: 442 men, 298 women (including genotype 5 participants)

Mean age: 53.8 years (including genotype 5 participants)

Inclusion criteria: chronic infection with HCV genotype 1, 2, 4, or 6, willing and able to provide written informed consent, HCV RNA ≥ 10^4 IU/mL at screening, classification as treatment‐naive or treatment‐experienced.

Exclusion criteria: current or prior history of clinically‐significant illness (other than HCV) or any other major medical disorder that may interfere with treatment, assessment, or compliance with the protocol; individuals currently under evaluation for a potentially clinically‐significant illness (other than HCV) are also excluded, screening ECG with clinically significant abnormalities, laboratory results outside of acceptable ranges at screening, prior exposure to sofosbuvir or other nucleotide analogue HCV NS5B inhibitor or any HCV NS5A inhibitor, infection with HBV or HIV.

Interventions

Experimental group: 400 mg of sofosbuvir and 100 mg of velpatasvir administered orally once daily for 12 weeks.

Control group: placebo.

Outcomes

SVR12, SAE, AE, viral resistance.

Notes

Participants in the placebo group were eligible for deferred treatment with 12 weeks of sofosbuvir–velpatasvir. Genotype 5 participants were not eligible for randomisation.

We contacted the trial authors on health‐related quality of life (HRQoL), allocation sequence generation, if they reported their SVR24 anywhere, at email [email protected].

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Low risk

An interactive web response system

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The trial was described as double‐blind (participant, caregiver, investigator, outcomes assessor), and the placebo was described in the protocol

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The trial was described as double‐blind (participant, caregiver, investigator, outcomes assessor), and the placebo was described in the protocol

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 5 participants had missing data

Selective reporting (reporting bias)

High risk

The trial did not report SVR24 as stated as a secondary objective in the protocol (NCT02201940 and supplementary material at NEJM.org)

Vested‐interest bias

High risk

"The study was designed and conducted by the sponsor (Gilead Sciences) in collaboration with the principal investigators."

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

FISSION 2013

Methods

Randomised, open label, active‐control study

Participants

527 participants were randomised and 499 participants were treated

Sex: 327 men, 172 women

Mean age: 48 years

Inclusion criteria: eligible participants were treatment‐naive adults aged 18 years or older with chronic hepatitis C genotype 2 or 3 infection and HCV RNA above 10,000 IU/mL. Participants with Childs A cirrhosis were included.

Exclusion criteria: BMI < 18 kg/m2; positive HbS‐Ab, positive HbC‐Ag, positive immunoglobulin‐M antibody, positive anti‐HIV antibody, history of other liver disease, current evidence of psychiatric illness, immunologic disorder, haemoglobinopathy, pulmonary disease (including pneumonia or pneumonitis), cardiac disease, seizure disorder or anticonvulsant use, poorly controlled diabetes, cancer, or history of malignancy, clinical signs and symptoms of acute pancreatitis with elevated lipase, clinically significant ECG findings at screening, history of major organ transplantation with an existing functional graft, active substance abuse, history of uncontrolled thyroid disease, haemoglobin < 11 g/dL for women or < 12 g/dL for men; neutrophil count < 1500 cells/nL, serum creatinine > 1.5 times the ULN, ALT or AST ≥ 10 x ULN, albumin ≤ 3.2 g/dL, total bilirubin 1.5 x ULN (except participants with Gilbert's syndrome).

Interventions

Experimental group 1: oral sofosbuvir 400 mg once daily for 12 weeks.

Control group: peg‐IFN α‐2a subcutaneous once weekly 180 µg for 24 weeks.

Co‐intervention: RBV 1000 mg/day (bodyweight < 75 kg) or 1200 mg/day (bodyweight ≥ 75 kg) for 12 or 24 weeks.

Outcomes

Proportion of participants with undetctable HCV RNA‐level at week 2 and week 4 under treatment, with SVR12, with SAE, AEs, mortality.

Notes

We emailed Lawitz and colleagues on 26 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Centralised system

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Unblinded

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Unblinded

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was unclear how the trial handled participants with missing data

Selective reporting (reporting bias)

Low risk

All outcomes in the protocol were reported on

Vested‐interest bias

Unclear risk

The sponsor (Gilead) collected the data, monitored the conduct of the study, and performed the statistical analysis

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Flamm 2013

Methods

Parallel group, double‐blind, placebo‐controlled, randomised trial

Participants

201 participants (134 in experimental group, 67 in control group)

Sex: 141 men (70%), 60 women (30%)

Race: 20 black (10%), 181 non‐black (90%)

Cirrhosis, n(%): 32(16%)

Location: USA

Inclusion criteria: chronic hepatitis C infection genotype 1 HCV RNA ≥ 10,000 IU/mL, demonstrated responsiveness to IFN (minimum duration of therapy 12 weeks); non‐response defined as a decrease in HCV RNA of at least 2 log10 IU/mL by week 12, but with detectable HCV RNA during the therapy period; relapse defined as an undetectable HCV RNA at end of treatment, but without subsequent attainment of SVR. A liver biopsy with histology consistent with chronic hepatitis C, age ≥ 18 years, weight between 40‐125 kg, signed informed consent, acceptable method of contraception for the participant and participant's partner(s) for at least 2 week before day 1 and continue until at least 6 months after treatment termination

Exclusion criteria: hepatitis B infection, HIV infection, other causes of liver disease, decompensated liver disease, uncontrolled diabetes mellitus, a severe psychiatric disorder, active substance abuse, active or suspected malignancy, or a history of malignancy within last 5 years, pregnant or nursing women, severe AE during prior treatment, seizure disorder, cerebrovascular diseases, cardiovascular disease, autoimmune diseases, prior organ transplantation, haemoglobinopathies, coagulopathies, abnormal levels of serum bilirubin, albumin, and creatinine, haemoglobin < 120 g/L (women) and < 130 g/L (men), neutrophil count < 1500/mm3, platelet count < 100,000/mm3.

Interventions

Experimental group: oral boceprevir 800 mg thrice daily for 44 weeks, beginning at week 5.

Control group: placebo for 44 weeks, beginning at week 5.

Co‐interventions: peg‐IFNα‐2a 180 μg subcutaneously once weekly and oral weight‐base RBV 1000 to 1200 mg daily in divided doses for 48 weeks.

Outcomes

Primary outcome: SVR 24 weeks post‐therapy.

Notes

We emailed Flamm and colleagues on 20 April 2016 for additional information (on: random sequence generation; method of allocation concealment; description of blinding procedure; blinding of outcome assessors; potential number and reasons for dropouts; pre‐defined outcomes; sponsorship and its role; type of SAE) but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Stated that trial was randomised, but the method of sequence generation was not described

Allocation concealment (selection bias)

Low risk

The trial used an interactive voice‐response system.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Trial defined as double‐blind and placebo was used in the control group. However, method of blinding was not adequately described.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It was not mentioned if the outcome assessors were blinded

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out

Selective reporting (reporting bias)

High risk

The trial authors changed their primary outcomes according to the protocol (NCT00845065)

Vested‐interest bias

High risk

The trial was funded by Schering‐Plough

Other bias

Low risk

The trial appeared to be free of other bias domains that could put it at risk of bias.

Forestier 2007

Methods

Randomised clinical trial

Participants

20 participants.

Sex: 12 men, 8 women

Mean age: 45.5 years

Inclusion criteria: men and women of non‐childbearing potential aged between 18 and 60 years. Participants satisfied the following criteria for inclusion in the study: genotype 1 chronic hepatitis C; had not received any prior therapy for hepatitis C, including approved treatments or participation in studies of investigational treatments; HCV RNA level > 1 × 105 IU/mL ALT concentration < 4.0 times the ULN, no clinically significant deviations from the normal range for haematology or clinical chemistry values; willing to refrain from the concomitant use of herbal dietary supplements or vitamins during the study drug‐dosing period; and willing to initiate standard‐of‐care treatment (peg‐IFNα and RBV) at the conclusion of the study drug‐dosing period.

Exclusion criteria: contraindications to peg‐IFNα‐2a or RBV; decompensated liver disease; alcohol‐related cirrhosis or primary biliary cirrhosis; positive screening for hepatitis B surface antigen or HIV co‐infection; donation of blood (500 mL) within 60 days before the first dose of study drug; concurrent antiviral therapy (except for antiviral agents approved for treatment of herpes viruses) within 3 months preceding study entry; regular treatment with nontopical medications or with topical medications with known systemic absorption within 4 weeks before study drug administration (with the exception of oestrogen replacement therapy for women); regular consumption of more than 24 units of alcoholic drinks per week or more than 8 cups of coffee per day; history of drug abuse within 6 months of study entry; history of methadone use within 3 months of study entry; positive urine screen for drugs of abuse; participation in an investigational drug study within 90 days before study drug administration or participation in more than 2 drug studies in the last 12 months (excluding the present study); or participation in a prior clinical study of telaprevir unless it was documented that the participant had been randomised to placebo treatment. Participants were also excluded if they had a history of any illness that, in the opinion of the investigator or the participant’s general practitioner, may have confounded the results of the study or posed an additional risk in administering study drug to the participant. This included but was not limited to a history of relevant drug or food allergies; cardiovascular or central nervous system disease; clinically significant illness; or mental illness that may have affected compliance with study requirements.

Interventions

Experimental group: telaprevir was given as 750 mg oral doses every 8 h. Telaprevir alone every 8 h orally for 14 days (8 participants); or telaprevir every 8 h orally for 14
days and peg‐IFNα‐2a once weekly for 2 weeks (8 participants)

Control group: placebo every 8 h orally for 14 days and peg‐IFNα‐2a via subcutaneous injection once weekly for 2 weeks (4 participants)

Co‐intervention: peg‐IFNα‐2a was given as weekly 180 mg subcutaneous injections

After completing study drug dosing, participants were offered the opportunity to begin standard therapy for chronic hepatitis C (180 g/week peg‐IFNα‐2a and 1000 or 1200 mg/day RBV, depending on body weight

Outcomes

Safety assessment, pharmacokinetic assessment, viral assessments

Notes

We contacted trial authors for additional information on allocation concealment, blinding of participants and personal, blinding of outcome assessment, SVR data protocol

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computerised random‐number generator

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The study was placebo‐controlled for telaprevir; peg‐IFNα‐2a treatment was open‐label. Investigators and participants were blinded to HCV RNA
results during the study drug‐dosing period

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It was unclear how (and if there was any blinding at all) the blinding was maintained and who performed the outcome assessment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No dropouts

Selective reporting (reporting bias)

Unclear risk

No protocol was found

Vested‐interest bias

High risk

Supported by Vertex Pharmaceuticals Incorporated

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Forestier 2011a1

Methods

Randomised clinical trial

Participants

50 participants

Sex: 40 men, 10 women

Mean age: 48 years

Inclusion criteria: men and women between 18 and 65 years of age with a history of chronic HCV genotype 1 infection and detectable plasma HCV RNA (> 1 104 IU/mL) at the study screening visit. Additional enrolment criteria included a BMI between 18 and 30, minimum body weight of 45 kg, and a liver biopsy or non‐invasive procedure (liver scan) within the previous 2 years showing no evidence of cirrhosis. In addition, participants in Part A were required to have no history of prior therapy with IFN‐based regimens; participants in Part B were required to have had failed previous IFN‐alfa and RBV‐based therapy as defined above.

Exclusion criteria: participants were excluded from the study if they met any of the following criteria: decompensated liver disease; impaired liver function; clinical or histopathologic evidence of cirrhosis; history of non‐hepatitis C chronic liver disease; positive screening for hepatitis B surface antigen or HIV infection; history of active malignancy within the preceding 5 years; history of clinically significant cardiovascular or cerebrovascular disease; treatment with peg‐IFN‐α and RBV (Part A) or treatment with peg‐IFN‐α and RBV within 3 months before screening (Part B); treatment with growth factors within 3 months before screening; history of drug abuse within the previous year; regular consumption of more than 1 glass of alcohol per day for women or 2 glasses of alcohol per day for men; participation in an investigational drug study within 3 months of screening or any prior participation in a study of an experimental HCV therapy; and selected laboratory abnormalities, including serum ALT > 5 times the upper limit of the reference range, creatinine clearance < 30 mL/min, or total bilirubin P26 lmol/L. Pregnant or lactating women, women of childbearing potential, and male partners of pregnant or lactating women were excluded from enrolment. Additionally, anyone who, in the opinion of the investigator, was not a suitable candidate for enrolment or was unlikely to comply with the requirements of the study was also excluded from enrolment.

Interventions

Experimental group:

Group 1: danoprevir was administered orally in soft gelatin capsule form in total daily doses of 200, 300, 400 and 600 mg in treatment‐naive participants.

Group 2: a single dose level of danoprevir (600 mg daily) was explored in a cohort of non‐responders (NR)

Control intervention: placebo

Outcomes

Safety assessments, pharmacokinetics, viral kinetics

Notes

4 cohorts of 10 participants each were randomised (8:2) to treatment with danoprevir or placebo equivalent. In Part A, treatment‐naive (Cohorts 1–5) were permitted but not required to begin standard of care (SOC) treatment with peg‐IFN‐α/RBV anytime after 24 h following the last dose of the study drug. 3 treatment‐naive participants in the 200 mg every‐12‐h cohort who were mis‐dosed at a single study site were excluded from the efficacy analysis. We sent an email was sent to Forestier and colleagues on 20 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were randomised using an interactive voice‐response system that assigned a participant identification number that corresponded to treatment assignment (danoprevir or placebo) according to the randomisation code.

Allocation concealment (selection bias)

Low risk

Participants were randomised using an interactive voice‐response system that assigned a participant identification number that corresponded to treatment assignment (danoprevir or placebo) according to the randomisation code.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The study was described as double‐blinded, but it was unclear how the blinding was maintained

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The study was described as double‐blinded but it was unclear how the blinding was maintained and who performed the outcome assessment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

3 participants excluded. Clearly stated reason

Selective reporting (reporting bias)

Unclear risk

No protocol was found

Vested‐interest bias

High risk

The study was sponsored by InterMune, Inc. and Roche

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Forestier 2011a2

Methods

For characteristics see Forestier 2011a2

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were randomised using an interactive voice‐response system that assigned a participant identification number that corresponded to treatment assignment (danoprevir or placebo) according to the randomisation code.

Allocation concealment (selection bias)

Low risk

Participants were randomised using an interactive voice‐response system that assigned a participant identification number that corresponded to treatment assignment (danoprevir or placebo) according to the randomisation code.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The study was described as double‐blinded, but it was unclear how the blinding was maintained

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The study was described as double‐blinded but it was unclear how the blinding was maintained and who performed the outcome assessment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

3 participants excluded. Clearly stated reason

Selective reporting (reporting bias)

Unclear risk

No protocol was found

Vested‐interest bias

High risk

The study was sponsored by InterMune, Inc. and Roche

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Forestier 2011b

Methods

Randomised clinical trial

Participants

59 participants

Sex: 46 men, 13 women

Mean age: 45.8 years

Inclusion criteria: genotype 1 chronic HCV infection with detectable plasma HCV RNA levels (> 1 x 104 IU/mL), no previous treatment for HCV infection, an age of 18–65 years, a BMI (defined as the weight in kilograms divided by the square of the height in meters) of 18–30, and no evidence of cirrhosis during the previous 2 years in a liver biopsy or noninvasive procedure (e.g. elastography).

Exclusion criteria: decompensated liver disease; impaired liver function; clinical or histopathologic evidence of cirrhosis; history of non–hepatitis C chronic liver disease; screening positive for hepatitis B surface antigen or HIV infection; history of active malignancy during the preceding 5 years; history of clinically significant cardiovascular or cerebrovascular disease; previous treatment with peg‐IFN‐a and RBV; treatment with growth factors within 3 months before screening; history of drug use within the previous year; regular consumption of > 1 glass of alcohol per day for women or > 2 glasses of alcohol per day for men; participation in an investigational drug study within 3 months before screening or any prior participation in a study of an experimental HCV therapy; and selected laboratory abnormalities, including ALT level .5 times the upper limit of the reference range, creatinine clearance < 30 mL/min, or total bilirubin level >26 mmol/L. Pregnant or lactating women, women of childbearing potential, and male partners of pregnant or lactating women were excluded from enrolment. In addition, anyone who, in the opinion of the investigator, was not a suitable candidate for enrolment or was unlikely to comply with the requirements of the study was also excluded from enrolment.

Interventions

Experimental group: danoprevir was administered orally in soft gelatin capsule form in the following dose regimens: 100 mg 3 times daily, 200 mg 3 times daily, 300 mg 3 times daily, 400 mg twice daily, 600 mg twice daily, and 900 mg twice daily. The 5 lowest dose cohorts consisted of 10 participants randomised (8:2) to receive treatment with danoprevir or placebo equivalent. The highest dose cohort consisted of 9 participants randomised (7:2) to receive treatment with danoprevir or placebo equivalent. 6 dose cohorts (400 mg, 600 mg, and 900 mg twice daily and 100 mg, 200 mg, and 300 mg 3 times daily). Participants also received peg‐IFN a‐2a (180 lg once weekly) and RBV (1000–1200 mg/day) on day 0 and 15.

Control group: placebo plus peg‐IFN a‐2a (180 lg once weekly) and RBV (1000–1200 mg/day)

Co‐intervention: peg‐IFN‐a 2 a(180 lg once weekly) and RBV (1000–1200 mg/day)

Outcomes

Safety assessments and viral kinetics

Notes

We sent an email to Forestier and colleagues on 20 April 2016 for additional information (missing blinding during assessment of allocation concealment, missing SVR and mortality data ‐ is it investigated) but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Participants were randomised using an interactive voice‐response system, which assigned a participant identification number corresponding with treatment assignment (danoprevir or placebo), according to the randomisation code.

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The study was described as double‐blinded but it was unclear how the blinding was maintained

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The study was described as double‐blinded but it was unclear how the blinding was maintained and who performed the outcome assessment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

One participant withdrew because of a family emergency after 1 dose of study drug, and 1 participant withdrew because of poor venous access after 4 doses of study drug. A third participant (administered 100 mg 3 times daily) missed 6 danoprevir doses during days 12–14 but was included in efficacy analyses, because 0.90% of danoprevir doses were administered

Selective reporting (reporting bias)

Unclear risk

No protocol was found

Vested‐interest bias

High risk

This study was supported by InterMune and Roche.

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Forns 2014

Methods

Randomised, multicenter, double‐blind, parallel‐group, placebo‐controlled, phase III clinical trial (PROMISE)(NCT01281839)

Participants

393 participants (260 in experimental group and 133 in control group)

Sex: 258 men, 135 women

Mean age: 52 years (range 20‐70 years)

Location: Europe, North America, Australia, and New Zealand.

Inclusion criteria: age ≥ 18 years. Confirmed chronic genotype 1 HCV infection. Screening plasma HCV RNA levels > 10,000 IU/mL. Treatment‐experienced participants who had relapsed after 24 weeks or more of IFN‐based therapy (undetectable HCV RNA at end of treatment or within 2 months after end of treatment, with documented relapse within 1 year after therapy). A liver biopsy specimen obtained within 3 years of screening showing histology consistent with chronic HCV infection (participants with bridging fibrosis (F3) or cirrhosis (F4) were eligible if they had an ultrasound performed within 6 months before screening (or between the screening and baseline visit) with no findings suspicious for HCC)

Exclusion criteria: hepatic decompensation. Non–HCV‐related liver disease. HBV, HIV, or non‐genotype 1 HCV co‐infection. Defined laboratory abnormalities: platelets < 90,000/mm3, white blood cell count < 3000/μL, haemoglobin level < 12 g/dL for women and < 13 g/dL for men, creatinine level > 1.5 mg/dL, ALT and/or AST level > 10 times the upper limit and normal, total serum bilirubin level 1.5 times or more the ULN, and α‐fetoprotein level > 50 ng/mL in participants with cirrhosis. Any other active disease. Pregnant women or planning pregnancy were excluded

Interventions

Experimental group: oral simeprevir 150 mg once daily for 12 weeks

Control group: placebo for 12 weeks

Co‐interventions:

Experimental group: peg‐IFN α‐2a 180 μg subcutaneously once weekly for 24 weeks (if HCV RNA < 25 IU/mL at week 4 and undetectable at week 12) or 48 weeks if not meeting these criteria. Oral weight‐based RBV 1000 to 1200 mg daily for 24 weeks (if HCV RNA < 25 IU/mL at week 4 and undetectable at week 12) or 48 weeks if not meeting these criteria

Control group: peg‐IFN α‐2a 180 μg subcutaneously once weekly for 48 weeks. Oral weight‐based RBV 1000 to 1200 mg daily for 48 weeks

Outcomes

Primary outcome: proportion of participants achieving SVR 12 weeks after planned end of treatment (SVR12)

Secondary outcomes: comparison of other virologic response rates at other time points. Rate of RVR. Proportion of simeprevir‐treated participants meeting response‐guided treatment criteria to complete treatment at week 24. Incidence of viral breakthrough. Incidence of on‐treatment failure. Incidence of viral relapse. Incidence of AEs. Laboratory abnormalities. Quality‐of‐life measures.

Notes

We sent an email to Forns and colleagues on 20 April 2016 for the following additional information. Reply received on 27 April 2016 with data on baseline number of participants with elevated AST/ALT and randomisation details.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information given

Allocation concealment (selection bias)

Unclear risk

Insufficient information given

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Stated that "participants, study personnel, and the sponsor were blinded to the treatment assignments"

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Stated that "participants, study personnel, and the sponsor were blinded to the treatment assignments"

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: "The proportion of patients who discontinued simeprevir/placebo intake early was 3.5% and 72.2% in the simeprevir/PR and placebo/PR groups, respectively. The main reason for discontinuation was meeting the week 4 virologic stopping rule for simeprevir or placebo in both arms, with a large proportion of patients in the placebo group (69.9%) stopping placebo at week 4. The proportion of patients who completed PR treatment was 93.5% in the simeprevir/PR group (24 or 48 weeks) and 72.2% in the placebo/PR group (48 weeks)"

Selective reporting (reporting bias)

Low risk

A protocol was published before randomisation. Outcomes specified in the protocol are similar, but not completely equal to the ones stated in the article. Not all outcomes stated in the protocol were reported in the article, but results of all outcomes were reported and available on www.ClinicalTrials.gov.

Vested‐interest bias

High risk

Trial sponsored by Janssen

Other bias

Low risk

The trial appeared to be free of other bias domains that could put it at risk of bias.

Foster 2011a1

Methods

Multicenter randomised clinical trial

Participants

52 participants

Sex: 35 men, 17 women

Mean age: 44 years

Countries: France, UK, Italy, and Sweden

Inclusion criteria: 18–65 years; chronic infection with either genotype 2 or genotype 3 HCV (serum HCV RNA > 10,000 IU/mL); absolute neutrophil count > 1500 mm3 and platelet count > 100,000 mm3; no prior treatment for HCV

Exclusion criteria: relevant concomitant medical condition; decompensated liver disease or cirrhosis, or other significant liver disease; HIV or HBV co‐infection; peg‐IFN or RBV contraindication; a history of alcohol or illicit drug use; pregnancy/breast feeding

Interventions

The participants were randomised according to genotype 2 and 3

Experimental group 1: oral 750 mg telaprevir every 8th hour for 2 weeks

Experimental group 2: oral 750 mg telaprevir every 8th hour + peg‐IFN‐α‐2a 180 µg once weekly plus RBV 400 mg twice daily for 2 weeks

Control group: telaprevir placebo (every 8 h) plus peg‐IFN‐α‐2a 180 µg once weekly plus RBV 400 mg twice daily for 2 weeks

Co‐intervention: The peg‐IFN‐α‐2a and RBV were a co‐intervention between control group and experimental group 2 during treatment period, and all participants received peg‐IFN‐α‐2a 180 g once weekly plus RBV 400 mg twice daily for 24 weeks after treatment.

Outcomes

Viral kinetics, efficacy and safety assessment

Notes

We emailed Foster and colleagues on 21 April 2016 for additional information (randomisation, blinding, death, missing data) but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described (central randomisation system)

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The monotherapy group was not blinded

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% percent dropped out (7 participants)

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol were assessed

Vested‐interest bias

Unclear risk

The trial was funded by Janssen Pharmaceuticals and Vertex Pharmaceuticals)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Foster 2011a2

Methods

For characteristics see Foster 2011a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described (central randomisation system)

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The monotherapy group was not blinded

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% percent dropped out (7 participants)

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol were assessed

Vested‐interest bias

High risk

The trial was funded by Janssen Pharmaceuticals and Vertex Pharmaceuticals)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Foster 2015a1

Methods

Randomised clinical trial

Participants

558 adult participants

Sex: 374 men, 178 women

Mean age: 49.5 years

Inclusion criteria: chronic hepatitis C genotype 3 who were treatment‐naive or treatment‐experienced, and were required to have liver imaging within 6 months of baseline/Day 1; adults with cirrhosis to exclude HCC , women of childbearing potential (as defined in Appendix 4 must have a negative serum pregnancy test at screening and a negative urine pregnancy test on Baseline/Day 1 prior to randomisation, male participants and female participants of childbearing potential who engage in heterosexual intercourse had to agree to use protocol‐specified method(s) of contraception, lactating women had to agree to discontinue nursing before the study drug was administered, participant had to be of generally good health, with the exception of chronic HCV infection, as determined by the Investigator, participant had to be able to comply with the dosing instructions for study drug administration and able to complete the study schedule of assessments

Exclusion criteria: current or prior history of clinically‐significant illness (other than HCV that may interfere with treatment, assessment or compliance with the protocol, screening ECG with clinically significant abnormalities, laboratory results outside of acceptable ranges at screening, pregnant or nursing female or male with pregnant female partner, chronic liver disease of a non‐HCV aetiology (e.g. haemochromatosis, Wilson's disease, alfa‐1 antitrypsin deficiency, cholangitis), infection with HBV or HIV

Interventions

Experimental group: 100 mg of velpatasvir once a day and 400 mg of sofosbuvir once a day for 12 weeks

Control group: 400 mg of sofosbuvir plus RBV 1000 or 1200 mg (weight‐based) both for 24 weeks

Outcomes

SVR12, SAE, death, viral resistance

Notes

We could only use data reported at 12 weeks meaning no data were available. We contacted the trial authors for additional information on allocation sequence generation, how many had incomplete outcome data at 12 weeks, SAE, death, health‐related quality of life) at 12 weeks at [email protected] on 21 April 2016 but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

An Interactive Web Response System was used

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as open‐label

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Described as open‐label

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There were above 5% dropouts and it was unclear how the trial handled missing participants. It was unclear how many dropouts there were at 12 weeks.

Selective reporting (reporting bias)

Unclear risk

SVR 24 was not reported as described in the prepublished protocol NCT02201953 and supplementary material at NEJM.org

Vested‐interest bias

High risk

The trial was funded by a company that might have an interest in a given result (Gilead Sciences)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Foster 2015a2

Methods

For characteristics see Foster 2015a2

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Low risk

An Interactive Web Response System was used

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Described as open‐label

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Described as open‐label

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 5 participants dropped out

Selective reporting (reporting bias)

High risk

SVR 24 was not reported as described in the prepublished protocol (NCT02220998 and supplementary material at NEJM.org)

Vested‐interest bias

High risk

The trial was funded by a company that might have an interest in a given result (Gilead Sciences)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Fried 2013

Methods

Phase IIb, double‐blind, placebo‐controlled, parallel‐group trial (PILLAR)(NCT00882908)

Participants

386 participants

Sex: 213 men, 173 women

Location: 13 countries in North America, Europe, and Asia‐Pacific regions

Inclusion criteria: adult participants with chronic hepatitis C, plasma HCV RNA > 100,000 IU/mL, genotype 1, treatment‐naive, eligible to be treated with peg‐IFN‐based regimens according to standard criteria

Exclusion criteria: cirrhosis on liver biopsy (required within 24 months of enrolment), HIV or HBV co‐infection, platelet count < 90,000/mm3, haemoglobin< 12 g/dL for women and 13 g/dL for men

Group 1:

78 participants

Sex: 40 men (51.3%), 38 women (48.7%)

Median age: 47 years (range 19‐66)

Group 2:

75 participants

Sex: 47 men(62,7%), 28 women (37.3%)

Median age: 46 years (range 18‐67)

Group 3:

77 participants

Sex: 43 men (55.8%), 34 women (44.2%)

Median age: 47 years (range 18‐69)

Group 4:

79 participants

Sex: 44 men (55.7%), 35 women (44.3%)

Median age: 47 years (range 19‐69)

Group 5:

77 participants

Sex: 39 men (50.6%), 38 women (49.4%)

Median age: 45 years (range 21‐67).

Interventions

Experimental group:

Group 1: oral simeprevir 75 mg once daily for 12 weeks, followed by placebo for 12 weeks.

Group 2: oral simeprevir 75 mg once daily for 24 weeks.

Group 3: oral simeprevir 150 mg once daily for 12 weeks, followed by placebo for 12 weeks.

Group 4: oral simeprevir 75 mg once daily for 24 weeks.

Control group:

Group 5: matched placebo for 24 weeks.

Co‐intervention for all groups: peg‐IFN‐α‐2a 180 μg subcutaneously once weekly. Oral RBV 1000‐1200 mg daily.

Outcomes

Primary outcome: proportion of participants with HCV RNA< 25 IU/mL undetectable at week 72 (SVR W72).

Secondary outcome: SVR at 12 and 24 weeks after planned end of treatment (SVR12 and SVR24, respectively). Adverse events. Quality‐of‐life measures. Assessment of HCV‐NS3 sequence in participants not achieving SVR. Assessment of simeprevir pharmacokinetics. The influence of interleukin‐28 (IL28)B genotype on efficacy was explored in a subset of participants for whom genomic DNA was available. Influence of IL28B genotype on treatment efficacy.

Notes

We emailed Fried and colleagues on 21 April 2016 for additional information (baseline number of participants with elevated AST/ALT and method of sequence generation but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The method of sequence generation was not described

Allocation concealment (selection bias)

Low risk

Participants were randomly assigned in equal proportions, using a centralised, interactive voice/web response randomisation system

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Stated that "participants and personnel were blinded to the experimental intervention. A simeprevir‐matched placebo was used."

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Stated as blinded. An external physician monitored individual HCV RNA results and informed investigators regarding protocol‐directed treatment discontinuation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Withdrawals reported with reasons given. Treatment discontinuation rate 7.5%

Selective reporting (reporting bias)

Unclear risk

A protocol was published before randomisation began and all outcome results were reported adequately (NCT00882908)

Vested‐interest bias

Unclear risk

This study was funded by Janssen Research & Development, LLC. Editorial support was provided by Dr Bethan Hahn, on behalf of Complete Medical Communications, funded by Janssen Research & Development, LLC.

Other bias

Low risk

The trial appeared to be free of other bias domains that could put it at risk of bias

Fundamental 2014a1

Methods

Prospective, double‐blind, multinational, randomised, placebo controlled phase II trial (CDEB025A2210; ClinicalTrials.gov NCT01183169) conducted between 30 August 2010 and 9 May 2013

Participants

459 eligible participants

Sex: 278 men, 181 women

Mean age: 50.6 years

Countries: Europe, North America, Asia‐Pacific region

Inclusion criteria: 9‐69 years with chronic hepatitis C genotype 1 infection and HCV RNA > = 1000 IU/mL and had failed to respond to or had relapse after prior P/R therapy; all participants had to have a liver biopsy within 3 years or transient elastography within 6 months of enrolment. Participants with compensated cirrhosis were eligible.

Exclusion criteria: nongenotype 1 infection, presence or history of hepatic decompensation and haematological abnormalities, and recent treatment with any anti‐HCV drug, concomitant treatment with known substrates or inhibitors of cytochrome P450 3A, P‐gp, OATPs, MRP2 or BSEP was not permitted within 2 weeks of study entry.

459 participants randomised, 77% white, 25% compensated cirrhosis/transition to cirrhosis, 57% prior P/R‐non responders, 79% genotype IL28B

457 treated.

Interventions

Participants were randomised (1:1:1:1)

Experimental group 1: alisporivir 600 mg once a day for 48 weeks.

Experimental group 2: alisporivir 800 mg once a day for 48 weeks.

Experimental group 3: alisporivir 400 mg twice a day for 48 weeks.

Control group: placebo for 48 weeks.

Co‐intervention: peg‐IFN‐α‐2a 180 lg/week plus RBV 1000 or 1200 mg/day based on body weight for 48 weeks.

Outcomes

eEVR (weeks 12 on treatment), SVR12, SVR24, all‐cause mortality, AEs.

Notes

Following a partial clinical hold imposed by FDA, alisporivir/placebo was discontinued in all participants; at that time, all active participants had received at least 31 weeks of triple therapy out of a total of 48 weeks.

Analysis group 1 vs control.

In the placebo arm, 57% of participants were switched in a blinded manner to alisporivir plus P/R after Week 16 due to failure to achieve the efficacy criterion (HCV RNA < limit of quantification) at Week 12. We could therefore not use the results from this trial.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not reported

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not reported

Incomplete outcome data (attrition bias)
All outcomes

Low risk

There was only missing data for 2 participants

Selective reporting (reporting bias)

High risk

The secondary outcomes were changed from the original secondary outcomes

Vested‐interest bias

High risk

The study was funded by Novartis

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Fundamental 2014a2

Methods

For characteristics see Fundamental 2014a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not reported

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not reported

Incomplete outcome data (attrition bias)
All outcomes

Low risk

There was only missing data for 2 participants

Selective reporting (reporting bias)

High risk

The secondary outcomes were changed from the original secondary outcomes

Vested‐interest bias

High risk

The study was funded by Novartis.

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Fundamental 2014a3

Methods

For characteristics see Fundamental 2014a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not reported

Allocation concealment (selection bias)

Unclear risk

Not reported

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not reported

Incomplete outcome data (attrition bias)
All outcomes

Low risk

There was only missing data for 2 participants

Selective reporting (reporting bias)

High risk

The secondary outcomes were changed from the original secondary outcomes

Vested‐interest bias

High risk

The study was funded by Novartis

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Gane 2008

Methods

Randomised clinical trial

Participants

25 adult participants

Country: New Zealand

Inclusion criteria: Non responders for RBV and IFN, infected with genotype 2 or 3. All participants were non‐cirrhotics, and treated with at least 12 weeks of IFN prior to randomisation.

Interventions

Experimental group: 1500 mg R7128 twice daily for 28 days.

Control group: placebo twice daily for 28 days.

Co‐intervention: 180μg peg‐IFN and 1000‐1200mg RBV.

Outcomes

HCV RNA, SAE, AEs

Notes

We emailed Gane and colleagues on 21 April 2016 for additional information regarding randomisation, blinding, missing data, death, additional data, separate data from Genotype 2 and 3.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There were missing data from 7 participants (above 5%)

Selective reporting (reporting bias)

Unclear risk

A clinicalTrials.gov number was found, but it was unclear which outcome was supposed to be assessed in each part of the trial

Vested‐interest bias

High risk

The main author was consulting in pharmaceutical companies

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Gane 2010

Methods

Randomised clinical trial

Participants

71 participants

Sex: 54 men, 17 women

Mean age: 47.6 years

Inclusion criteria: treatment‐naive and treatment‐experienced adults aged 18–65 years, who were chronically infected with HCV genotype 1 but did not have cirrhosis, and who had a minimum HCV RNA of 10⁵ IU/mL. Participants were required to have normal renal and hepatic function and no clinically significant comorbidities.

Exclusion criteria: co‐infection with hepatitis B or HIV, concurrent medical or psychiatric disorder (or history of such), history of any neoplastic disease, history of clinically significant cardiovascular or cerebrovascular disease, use of growth factors, or anticipated use or need for significant concomitant medical treatment.

Interventions

Experimental group:

Arm B: 500 mg RG7128 twice daily and 100 mg danoprevir every 8 h (treatment‐naive)

Arm C1: 500 mg RG7128 twice daily and 200 mg danoprevir every 8 h (treatment‐naive)

Arm C2 1000 mg RG7128 twice daily and 100 mg danoprevir every 8 h (treatment‐naive)

Arm D: 1000 mg RG7128 twice daily and 200 mg danoprevir every 8 h (treatment‐naive)

Arm E: 1000 mg RG7128 twice daily and 600 mg danoprevir twice a day (non‐null responders)

Arm F: 1000 mg RG7128 twice daily and 900 mg danoprevir twice a day (null responders)

Arm G: 1000 mg RG7128 twice daily and 900 mg danoprevir twice a day (treatment‐naive)

Control group: placebo RG7128 and Placebo Danoprevir

Co‐intervention: standard of care treatment (180 μg/week peg‐IFN α‐2a, and RBV at 1000 mg/day for participants weighing < 75 kg or 1200 mg/day for those weighing ≥ 75 kg)

Outcomes

Safety, pharmacokinetics, antiviral activity

Notes

We emailed Gane and colleagues on 06 June 2016 for additional information on SVR24 but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The random allocation sequence was computer‐generated

Allocation concealment (selection bias)

Low risk

Randomly assigned by interactive voice or web response system to active treatment or placebo

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Investigators, personnel at the study centre, and participants were masked to treatment allocation. Study drugs and placebo were identical in colour, size, shape, and taste but "(...) apart from patients in cohort F, who were unmasked after the last assessment was completed"

Blinding of outcome assessment (detection bias)
All outcomes

High risk

The pharmacist who prepared the doses, personnel involved in pharmacokinetic sample analyses, statisticians who prepared data summaries, and the clinical pharmacologists who reviewed the data before deciding to initiate dosing in the next cohort were not masked to treatment allocation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

There was under 5% dropouts (only 2 dropouts)

Selective reporting (reporting bias)

Low risk

The outcomes stated in the protocol were reported on (NCT00801255)

Vested‐interest bias

High risk

The trial was funded by a company that might have an interest in a given result (Hoffmann‐La Roche)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Gane 2011

Methods

Randomised clinical trial

Participants

30 adult participants

Sex: 21 men, 9 women

Mean age: 44.5 years

Countries: New Zealand, France, Poland

Inclusion criteria: 18‐65 years and with chronic treatment‐naive hepatitis C genotype 1 infection, an HCV RNA level > x105 IU/mL, a BMI between 18 and 35 kg/m2 and without evidence of liver cirrhosis on a liver biopsy or non‐invasive procedure (e.g. Fibroscan) obtained within the preceding 24 months were eligible for the trial.

Exclusion criteria: decompensated liver disease; impaired liver function (indicated by a history of ascites, hepatic encephalopathy, HCC or bleeding oesophageal varices); chronic liver disease attributed to a cause other than HCV; or serological evidence of HBV or HIV infection. Increased risk of anaemia; a clinically significant medical condition such as cardiovascular or cerebrovascular disease, chronic pulmonary disease, poorly controlled thyroid function, diabetes mellitus requiring medication, ophthalmic disorders related to diabetes or hypertension, or diseases associated with alterations in immune function; or a history of clinically significant psychiatric disease, a history of excessive alcohol consumption (defined as more than 2 standard drinks per day within the previous 3 months), or a history of drug abuse within the last year, pregnant and lactating women and male partners of pregnant women, any recent use or anticipated need for drugs, herbal preparations or nutrients known to inhibit or induce CYP enzymes, or were substrates of CYP3A or CYP2C9 with a narrow therapeutic index (including oral contraceptives, steroids, antacids, H‐2 blockers or proton‐pump inhibitors). Systemic immunosuppressive drugs, cytotoxic or chemotherapeutic agents, radiation therapy, oral or inhaled corticosteroids, or topical class 1 and 2 steroids. ALT level > 5 times the ULN, creatinine clearance < 50 mL/min, haemoglobin < 120 g/L (if female) or < 130 g/L (if male), an absolute neutrophil count < 1.5 109/L, platelet count < 100 x 109/L, or serum albumin level < 35 g/L

Interventions

The study consisted of 3 cohorts. The randomisation was within each cohort.

Experimental group: participants received 100 mg oral danoprevir twice a day, 200 mg oral danoprevir once a day, or 200 mg oral danoprevir twice a day for 15 days.

Control group: placebo in same numbers as above.

Co‐intervention: both groups received equal amounts of ritonavir (100 mg) pr pill. subcutaneous peg‐IFN α‐2a (40KD) (Pegasys, Roche, Basel, Switzerland) 180 μg once weekly plus oral RBV 1000 mg/day (bodyweight < 75 kg) or 1200 mg/day (bodyweight > 75 kg).

After the 15 days, both groups received peg‐IFN α‐2a (40KD) plus RBV for a total of 48 weeks.

Outcomes

Pharmacokinetic parameters (plasma concentration, AUC), HCV RNA level, safety assessment (laboratory test, AEs).

Notes

We emailed Gane and colleagues on 06 June 2016 for additional information on blinding, other outcomes, protocol but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computerised randomisation

Allocation concealment (selection bias)

Low risk

Randomisation was managed through a centralised interactive voice and web response system through a 3rd party

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The study was described as "partially" double‐blinded but it was unclear how the blinding was maintained

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The study was described as "partially" double‐blinded but it was unclear how the blinding was maintained and who performed the outcome assessment

Incomplete outcome data (attrition bias)
All outcomes

Low risk

There were no dropouts

Selective reporting (reporting bias)

High risk

The protocol stated "Virological response in prior null‐responders" as a secondary outcome. This outcome was not assessed in any study

Vested‐interest bias

High risk

The trial was sponsored by F. Hoffmann‐La Roche Ltd

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Gane 2015

Methods

Randomised clinical trial

Participants

30 adults with chronic hepatitis C infection

Sex: 17 men, 13 women

Mean age: 45 years

Countries: New Zealand and USA

Interventions

Experimental group 1: 12 participants randomised to 50 mg ACH‐3102 (odalasavir) and 400 mg sofosbuvir once a day for 8 weeks
Control group 1: 6 participants randomised to observation for 8 weeks.

Experimental group 2: 6 participants randomised to 50 mg ACH‐3102 (odalasavir) and 400 mg sofosbuvir once a day for 6 weeks

Control group 2: 6 participants randomised to observation for 6 weeks.

Outcomes

SVR, SAE.

Notes

Abstract only. After 4 weeks of treatment, group 1 (both experimental and control group) were merged, and received active treatment, therefore data can not be used after week 4. We emailed Gane and colleagues on 21 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

"Observation group" not placebo controlled trial

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

High risk

Sponsored by Achillion Pharmaceuticals

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Gardner 2014a

Methods

Randomised clinical trial

Participants

16 participants

Sex: 14 men, 1 women (analysed)

Mean age: 53 years

Countries: USA and Puerto Rico

Inclusion criteria: treatment‐naive participants men and women 18‐70 years of age with HCV genotype 1 or 4 infection for at least 6 months and HCV RNA ≥ 1,00,000 IU/mL at screening. Eligible participants had no evidence of cirrhosis documented by liver biopsy within 3 years. Fertile men or women were required to use 2 forms of effective contraception between them and their partner during treatment and for 24 weeks afterwards

Exclusion criteria: co‐infection with hepatitis B, HIV, clinically significant chronic liver disease, conditions consistent with decompensated liver disease, drug or alcohol abuse, significant ECG findings, history of suicide attempt, major depression or current severe or poorly controlled psychiatric disorder. Abnormal haematological and biochemical parameters that excluded participation were: Neutrophil count (< 1500 cells/mm3 ((or < 1250 cells/mm3 for African American/Black participants)); haemoglobin (< 11 g/dL in women or 12 g/dL in men); creatinine > 1.5 x ULN (ULN); ALT, AST, or alkaline phosphatase > 5 x ULN; total bilirubin > 2.0 x ULN ((except in participants with Gilbert's) syndrome; albumin < 3.0 g/dL and platelet count < 90,000/mm3. Participants were excluded if they received herbal/natural remedies with anti‐HCV activity within 30 days of the baseline visit. The use of systemic antineoplastic or immunomodulatory treatments within 6 months of the baseline visit excluded participation and was not allowed during this study. The use of growth factors was not allowed during this study. In the absence of clinical drug interaction study data, medications that modulate stomach acid and known inhibitors or inducers of the cytochrome P450 3A enzyme and P‐glycoprotein transporter systems were prohibited.

Interventions

Experimental group: oral 60 mg of GSK2336805 for 28 days.

Control group: placebo for 28 days.

Co‐intervention: peg‐IFN α‐2a (180 μg per week) and RBV (1000–1200 mg daily) from day 2 and for 27 days in total.

Outcomes

Safety assessment, HCV RNA, pharmacokinetics.

Notes

NCT01439373. The trial had 2 parts. Part 1: 1‐day therapy with GSK2336805 versus placebo. Part 2: 27 days of GSK2336805 versus placebo with RBV and peg‐IFN as co‐intervention. We emailed Gardner and colleagues on 21 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out (1 person)

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol (NCT01439373) were assessed

Vested‐interest bias

High risk

GlaxoSmithKline, LLC

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

GlaxoSmithKline 2014

Methods

Randomised clinical trial

Participants

37 adult participants (18‐60 years) chronically infected with HCV (genotype 1 (1a or 1b), genotype 2 or genotype 4.

Interventions

Experimental group: oral GSK2878175 10 mg, 30 mg or 60 mg for 2 days.
Control group: placebo.

Outcomes

Safety, pharmacokinetics, HCV RNA.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The participants and personnel were blinded

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as being blinded but it was unclear how the blinding of outcome assessors was performed

Incomplete outcome data (attrition bias)
All outcomes

Low risk

< 5% dropped out

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

High risk

The trial was sponsored by Glaxo Smith Kline

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Goldwater 2010

Methods

Randomised clinical trial

Participants

32 adult treatment‐naive participants with HCV genotype1

Country: USA

Interventions

Experimental group: oral 150 mg, 300 mg, 450 mg of GS‐9256 as a single dose.

Control group: placebo.

Outcomes

HCV RNA, pharmacokinetics.

Notes

The trial also had groups with healthy volunteers.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as being placebo blinded, but it was unclear how the blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as being placebo blinded, but it was unclear how the blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No data

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

High risk

The trial was funded by Gilead Sciences

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

HALLMARK‐DUAL 2014

Methods

Randomised clinical trial

Participants

307 adult participants

Sex: 155 men, 152 women

Mean age: 54.5 years

Countries: Argentina, Australia, Austria, Canada, France, Germany, Ireland, Israel, Italy, Republic of Korea, Netherlands, New Zealand, Poland, Russian Federation, Spain, Taiwan, UK and USA.

Inclusion criteria: aged at least 18 years with genotype 1b infection and HCV RNA of 10,000 IU/mL or greater who met inclusion criteria for 1 of 3 cohorts: treatment‐naive, previous non‐responder to peg‐IFNα plus RBV (null or partial response), or ineligible for, intolerant of, or ineligible for and intolerant of peg‐IFN α plus RBV (treatment‐naive and treatment‐experienced). Ineligible or intolerant (or both) participants included those with depression, anaemia or neutropenia, or compensated advanced fibrosis or cirrhosis (F3/F4) with thrombocytopaenia. Anaemia was defined as haemoglobin between 85 g/L and < 120 g/L (women) or < 130 g/L (men), neutropenia as absolute neutrophils between 0.5 x 10⁹ cells per L and < 1.5 x 10⁹ cells per L, and thrombocytopenia as platelets between 50 x 10⁹ cells per L and < 90 x 10⁹ cells per L, at screening or history of these conditions, while receiving peg‐IFN α plus RBV, or both.

Exclusion criteria: people with HIV, ascites, oesophageal varices, or other evidence of hepatic decompensation.

Interventions

Experimental group: oral 60 mg once daily of daclatasvir and oral 100 mg twice daily of asunaprevir for 24 weeks.

Control group: placebo for 12 weeks.

Outcomes

HCV RNA (SVR), safety assessment.

Notes

Only participants in the treatment‐naive group were randomised. The placebo group entered a new study after 12 weeks, therefore only data for the first 12 weeks could be used. We emailed Manns and colleagues on 27 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random allocation sequence

Allocation concealment (selection bias)

Low risk

Interactive voice‐response system

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The participants and personnel were blinded to treatment allocation until week 12, and we used data until week 12

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The sponsors, who performed the analyses, were blinded until week 12, and we used data until week 12

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

The amount of drop‐outs until week 12 were not described

Selective reporting (reporting bias)

High risk

2 outcomes were added to the secondary outcomes in the protocol (NCT01581203)

Vested‐interest bias

High risk

The trial was funded by Bristol‐Myers Squibb

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Han 2014

Methods

Randomised, placebo‐controlled, parallel‐group trial

Participants

107 participants

Ethnicity: Korean

Race: Asian

Country: South Korea, India, Taiwan

Inclusion criteria: chronic hepatitis C infection and genotype 1. Previous treatment failure (relapse, non‐responders, and partial responders)

Interventions

Experimental group: boceprevir for 32 weeks, beginning at week 5.

Control group: placebo for 44 weeks, beginning at week 5.

Co‐interventions:

Experimental group: peg‐IFN and RBV for 36 week (participants with detectable HCV RNA at week 8 received additional 12 weeks of treatment, in total 48 weeks).

Control group: peg‐IFN and RBV for 48 weeks.

Outcomes

Not specified

Notes

This trial was only available as an abstract of an interim‐analysis.

The co‐interventions in both groups (experimental and control) were not completely equal ‐ while all the participants in the control group received Peg‐IFN + RBV for 48 weeks, the experimental group received a response‐guided regimen which implied that some participants received shorter duration of treatment (36 weeks), while others received 48 weeks.

The following Information is required: number of participants randomised per group; method of sequence generation; method of allocation concealment; description of blinding; number and reasons for withdrawal; pre‐specified outcomes; sponsorship and its role

No contact details of authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The method of sequence generation was not specified

Allocation concealment (selection bias)

Unclear risk

Insufficient information provided

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Use of placebo suggests blinding, but method of blinding was not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Insufficient information provided

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Insufficient information provided

Selective reporting (reporting bias)

Unclear risk

Insufficient information provided. No protocol available

Vested‐interest bias

Unclear risk

It was uncertain how the trial was sponsored

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Hezode 2009

Methods

A phase IIb, randomised, partially double‐blind, placebo‐controlled, parallel‐group trial (PROVE‐2) (NCT00372385)

Participants

323 participants

Sex: 192 men, 131 women

Country: France, Germany, the UK, and Austria

Inclusion criteria: age between 18 and 65 years. Chronic hepatitis C infection. HCV genotype 1. Detectable plasma HCV RNA levels. treatment‐naive. No histologic evidence of cirrhosis within 2 years before study Day 1. Seronegative for hepatitis B surface antigen and HIV‐1 and 2. Adequate double method of contraception. Negative pregnancy test for women.

Exclusion criteria: any medical contraindication to peg‐IFN α‐2a or RBV therapy. Any other cause of significant liver disease in addition to hepatitis C. Diagnosed or suspected HCC. Alcohol/drug abuse or excessive use in the last 12 months. Participation in any investigational drug study within 90 days before drug administration.

Group 1: 81 participants: (T12PR24)

Sex: 54 men, 27 women

Median age: 46 years (range 19‐65)

Race: 75 white (93%), 1 black (1%), 3 Asian (4%), 1 Hispanic (1%), 1 other (1%)

HCV RNA ≥ 800,000 IU/mL, n(%): 73(90)

Fibrosis, n(%): none or minimal: 35(43). Portal: 37(46). Bridging: 9(11). Cirrhosis: 0

HCV genotype, n(%): 1a: 31(38). 1b: 50(62). Intermediate: 0

Group 2: 82 participants (T12PR12)

Sex: 49 men, 33 women

Median age: 44 years (range 22‐65)

Race: 76 white (93%), 2 black (2%), 2 Asian (2%), 1 Hispanic (1%), 1 other (1%)

HCV RNA ≥ 800,000 IU/mL, n(%): 67(82)

Fibrosis, n(%): none or minimal: 30(37). Portal: 46(56). Bridging: 6(7). Cirrhosis: 0

HCV genotype, n(%): 1a: 37(45). 1b: 45(55). Intermediate: 0

Group 3: 78 participants (T12P12)

Sex: 43 men, 55 women

Median age: 45 years (range 20‐64)

Race: 77 white (99%), 1 black (1%), 0 Asian, 0 Hispanic, 0 other

HCV RNA ≥800,000 IU/mL, n(%): 63(81)

Fibrosis, n(%): none or minimal: 31(40). Portal: 43(55). Bridging: 3(4). Cirrhosis: 1(1)

HCV genotype, n(%): 1a: 40(51). 1b: 38(49). Intermediate: 0

Group 4: 82 participants (PR48)

Sex: 46 men, 36 women

Median age: 45 years (range 18‐64)

Race: 76 white (93%), 2 black (2%), 4 Asian (5%), 0 Hispanic, 0 other

HCV RNA ≥800,000 IU/mL, n(%): 68(83)

Fibrosis, n(%): none or minimal: 28(34). Portal: 46(56). Bridging: 8(10). Cirrhosis: 0

HCV genotype, n(%): 1a: 35(43). 1b: 45(55). Intermediate: 2(2).

Interventions

Experimental group:

1, 2, and 3: oral telaprevir given as a single dose of 1250 mg on study day 1, followed by a dose of 750 mg every 8 h for 12 weeks.

Control group:

4: placebo for 12 weeks.

Co‐interventions:

1: peg‐IFN α‐2a 180 μg subcutaneously once weekly plus oral weight‐based RBV 1000 to 1200 mg in 2 divided daily doses for 24 weeks

2: peg‐IFN α‐2a 180 μg subcutaneously once weekly plus oral weight‐based RBV 1000 to 1200 mg in 2 divided daily doses for 12 weeks

3: peg‐IFN α‐2a 180 μg subcutaneously once weekly for 12 weeks

4: peg‐IFN α‐2a 180 μg subcutaneously once weekly plus oral weight‐based RBV 1000 to 1200 mg in 2 divided daily doses for 48 weeks.

Outcomes

Primary outcome: proportion of participants who achieved SVR at 24 weeks after end of treatment (HCV RNA undetectable (< 10 IU/mL) 24 weeks after completion of study treatment)

Secondary outcomes: proportion of participants with undetectable HCV RNA at week 12 after end of treatment. Proportion of participants with undetectable HCV RNA at completion of study drug dosing. Number of participants with AEs. Number of participants with viral relapse. Maximum, minimum, and average plasma concentration of telaprevir

Notes

We emailedWe emailed Hezode and colleagues on 21 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No information provided

Allocation concealment (selection bias)

Unclear risk

Randomisation was performed through a central telephone‐based system. No other information was provided

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Group 3 (T12P12) was not blinded. Other treatment groups were blinded to the interventions

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not enough information provided

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Number and reasons for discontinuation were clearly reported on.

Selective reporting (reporting bias)

Low risk

Protocol was available and all pre‐specified outcomes were reported on

Vested‐interest bias

High risk

The sponsor (Vertex Pharmaceuticals) was directly involved in trial design and protocol development

Other bias

Low risk

The trial seems to be free of other potential sources of bias

Hinrichsen 2004

Methods

Randomised clinical trial

Participants

51 adult participants

Sex: 41 men, 10 women

Mean age: 47.8 years

Countries: Germany, France, and Spain.

Inclusion criteria: women or men aged 18 years or older with chronic genotype 1 HCV infection. The line probe assay was used to determine the genotype of the viral infection. A liver biopsy specimen showing changes consistent with chronic HCV infection had to have been performed within the previous 12 months. At screening, the HCV load had to be 50,000 copies/mL serum.

Exclusion criteria: women were excluded if they were breast‐feeding or at risk of pregnancy; men had to use an adequate form of contraception if their partner was of childbearing potential. They were not enrolled if there were other or additional reasons for chronic liver disease, including the presence of other hepatitis‐causing viruses and/or a history of alcohol abuse within the previous 12 months and/or evidence of Child’s B or C liver disease at screening. No other antiviral or antimicrobial or investigational therapies were allowed during the study (screening, pretreatment, and treatment phases). Patients were excluded if, at screening, their baseline ALT/AST) plasma levels exceeded the ULN by more than 5‐fold (5 times the ULN) or their total bilirubin or alkaline phosphatase levels were 1.5 times the ULN. Other exclusion criteria included co‐infection with HIV, a platelet count 100,000/mm3, a white blood cell count 2000 cells/mm3, any clinically significant laboratory abnormalities, and a positive test result for illicit or nonprescription drugs.

Interventions

The trial was divided into 3 different cohorts, according to grade of liver disease (Ishak score, Metavir score).

Experimental group: 2 days of oral 25 mg, 200 mg or 500 mg of BILN‐2061 in participants with Ishak score 0‐2. Oral 200 mg of BILN 2061 in participants with Ishak score 3‐4. Oral 200 mg of BILN 2061 in participants with Ishak score 5‐6.

Control group: placebo.

Outcomes

Virologic efficacy, pharmacokinetics, safety assessment.

Notes

We emailedWe emailed Hinrichsen and colleagues on 21 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Low risk

0 participants dropped out

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained for all 3 stages, and the clinicalTrials.gov information was added after completion

Vested‐interest bias

High risk

The trial was funded by Boehringer Ingelheim

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Hoeben 2015a1

Methods

Phase III, randomised, double‐blind, placebo‐controlled, parallel‐group trial (TIGER)(NCT01725529)

Participants

457 participants

Median age: 48 years (range 18‐68)

Sex: 236 men, 221 women

Country: China, Korea

Ethnicity (%): Chinese (80.3%), Korean (19.7%)

HCV genotype (%): 1a (1.1%), 1b (98.9%)

Inclusion criteria: treatment‐naive East Asian participants with chronic hepatitis C. A liver biopsy within 3 years prior to the screening visit (or between screening and day of randomisation) with histology consistent with chronic Hepatitis C virus (HCV) infection (presence of contraindications for a liver biopsy in participants who are otherwise deemed eligible for participation does not exclude the patient from participation). Genotype 1 HCV infection (confirmed at screening). Plasma HCV RNA of > 10,000 IU/mL at screening. Age between 18‐70 years.

Exclusion criteria: prior treatment with any approved or investigational drug for the treatment of hepatitis C. Co‐infection with HBV or HIV.

Interventions

Experimental group:

Group 1: Simeprevir 150 mg orally once daily for 12 weeks.

Group 2: Simeprevir 100 mg orally once daily for 12 weeks.

Control group:

Group 3: matching placebo capsules taken orally with food once‐daily for 48 weeks.

Co‐interventions:

Group 1 and 2: peg‐IFN α‐2a μg once weekly administered as weekly subcutaneous injections of 0.5 mL for 24 or 48 weeks. RBV 1000 or 1200 mg/day (taken as 100 mg or 200 mg tablets) depending on body weight for 24 or 48 weeks (If body weight is < 75 kg the total daily dose of RBV will be 1000 mg, administered as 400 mg intake with food in the morning and 600 mg intake with food in the evening. If body weight is > or = 75 kg the total daily dose will be 1200 mg, administered as 2 x 600 mg per intake with food, morning and evening).

Group 3: peg‐IFN α‐2a μg once weekly administered as weekly subcutaneous injections of 0.5 mL for 48 weeks. RBV 1000 or 1200 mg/day (taken as 100 mg or 200 mg tablets) depending on body weight for 48 weeks (If body weight is < 75 kg the total daily dose of RBV will be 1000 mg, administered as 400 mg intake with food in the morning and 600 mg intake with food in the evening. If body weight is ≥ 75 kg the total daily dose will be 1200 mg, administered as 2 x 600 mg per intake with food, morning and evening).

Outcomes

Primary outcome measures: percentage of participants with SVR 12 weeks after end of study drug treatment (participants considered to have achieved SVR12 if both conditions are met: 1. HCV RNA < 25 IU/mL or undetectable at end of treatment and; 2. HCV RNA is < 25 IU/mL or undetectable at 12 weeks after the planned end of study drug treatment).

Secondary outcome measures: percentage of participants with SVR 24 weeks after end of study drug treatment (participants considered to have achieved SVR24 if both conditions are met: 1. HCV RNA < 25 IU/mL or undetectable at end of treatment; 2. HCV RNA < 25 IU/mL or undetectable at 24 weeks after the planned end of study drug treatment). Percentage of participants with SVR at week 72. Percentage of participants with on‐treatment failure (refers to a participant with confirmed detectable HCV RNA at the end of treatment). Percentage of participants with viral breakthrough (defined as a confirmed increase of > 1 log10 IU/mL in HCV RNA level from the lowest level reached, or a confirmed HCV RNA level of > 100 IU/mL in participants whose HCV RNA levels had previously been below the limit of quantification (< 25 IU/mL detectable) or undetectable (< 25 IU/mL undetectable) while on study treatment). Percentage of participants with viral relapse (defined as undetectable HCV RNA at the actual end of treatment and last HCV RNA measurement during follow‐up ≥ 25 IU/mL). Percentage of participants with on‐treatment normalisation of ALT level.

Notes

Abstract. Interim analysis. We emailedWe emailed Hoeben and colleagues on 21 April 2016 for additional information (on method of sequence generation and method of allocation concealment) but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The method of sequence generation was not specified

Allocation concealment (selection bias)

Unclear risk

The method of allocation concealment was not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

A simeprevir‐matched placebo was used

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The protocol stated that outcomes assessors were blinded to the intervention

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Number and reasons for withdrawal were stated at www.ClinicalTrials.gov (NCT01725529)

Selective reporting (reporting bias)

Low risk

A protocol was published before randomisation began and all outcome results were reported adequately

Vested‐interest bias

High risk

The trial was sponsored by a pharmaceutical company (Janssen)

Other bias

Low risk

The trial appeared to be free of other bias domains that could put it at risk of bias

Hoeben 2015a2

Methods

For characteristics see Hoeben 2015a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The method of sequence generation was not specified

Allocation concealment (selection bias)

Unclear risk

The method of allocation concealment was not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

A simeprevir‐matched placebo was used

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The protocol stated that outcomes assessors were blinded to the intervention

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Number and reasons for withdrawal were stated at www.ClinicalTrials.gov (NCT01725529)

Selective reporting (reporting bias)

Low risk

A protocol was published before randomisation began and all outcome results were reported adequately

Vested‐interest bias

High risk

The trial was sponsored by a pharmaceutical company (Janssen)

Other bias

Low risk

The trial appeared to be free of other bias domains that could put it at risk of bias

Hotho 2012

Methods

Randomised clinical trial

Participants

13 participants

Sex: 12 men, 1 woman

Mean age: 49 years

Countries: Netherlands and USA.

Inclusion criteria: chronic hepatitis C participants, both treatment‐naive or treatment‐experienced, aged 18‐65 with a BMI 18‐32.

Exclusion criteria: decompensated liver disease, uncontrolled or active major systemic disease and co‐infection with HIV or HBV. Participants with chronic stable haemophilia or on stable methadone substitution treatment.

Interventions

The trial was divided into single and multi ascending cohorts (only cohort 4, 5 and 11, 12 were HCV‐infected participants)

Experimental group 1: single ascending dose: 100 mg, 500 mg once daily, or 250 mg twice daily PHX1766

Experimental group 2: multi ascending dose: 400 mg twice daily, 800 mg twice daily PHX1766

Control group: placebo, only in the multi ascending dose

Outcomes

Pharmacokinetics, safety assessment, pharmacodynamics.

Notes

We emailed Hotho and colleagues on 21 April 2016 for additional information but reply not received yet.

The trial also included healthy volunteers.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as being placebo‐controlled, but method was not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as being placebo‐controlled, but method was not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

High risk

The trial was funded by Phenomix Corporation

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Isakov 2016

Methods

Randomised clinical trial

Participants

Treatment‐naive and treatment‐ experienced participants (prior treatment with PR for ≥ 12 weeks had failed) with chronic HCV genotype 1 infection.

Interventions

All participants initially received PR for 4 weeks. Participants randomised to control treatment then received PR for an additional 44 weeks. Treatment‐naive participants randomised to triple therapy received boceprevir (800 mg 3 times daily) plus PR for 24 weeks and then further therapy according to treatment week 8 HCV RNA levels. Treatment‐experienced participants received boceprevir plus PR for 32 wk and then further therapy according to treatment week 8 HCV RNA levels.

Outcomes

SVR defined as undetectable HCV RNA 24 weeks after completing all study therapy.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

< 5% missing data

Selective reporting (reporting bias)

Unclear risk

No protocol

Vested‐interest bias

High risk

"Supported by Merck and Co., Inc. Kenilworth, NJ, US."; "Medical writing and editorial assistance were provided by Tim Ibbotson, Ph.D. of ApotheCom, Yardley,PA, United States."

Izumi 2014a1

Methods

Randomised clinical trial

Participants

42 adult participants

Sex: 20 men, 22 women

Mean age: 55 years

Country: Japan
Inclusion criteria: Japanese men and women 20‐70 years of age chronically infected with HCV genotype 1 (HCV RNA > 105 IU/mL) who were treatment‐naive (with alfa‐2a or 2b/RBV or DAA), or those who were non‐responders to previous therapy. Women of childbearing potential were required to use effective methods of contraception.

Exclusion criteria: history of HCC, co‐infection with HBV or HIV, other chronic liver disease, or evidence of hepatic decompensation. Liver cirrhosis, liver biopsy within 24 months, elevated ALT, bilirubin, albumin, decreased haemoglobin, white blood cells, neutrophil count, platelets, creatinine, participants exposed any investigational HCV therapeutic agent 4 weeks prior to dosing.

Interventions

Experimental group: oral 10 mg or 60 mg of daclatasvir once daily.

Control group: placebo.

Co‐intervention: weight‐based RBV twice daily, once weekly subcutaneous alfa‐2a IFN.

Participants receiving protocol‐defined response were treated for 24 weeks. Participants not receiving protocol‐defined response were treated for 48 weeks.

Outcomes

Efficacy assessment, safety assessment, virological response.

Notes

NCT01017575 ‐ only data from the treatment‐naive group could be used, since the non‐responders couldn't be randomised to placebo.

We emailed Izumi and colleagues on 21 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

not described

Allocation concealment (selection bias)

Low risk

Central randomisation centre

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Unblinded after week 24

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Unblinded after week 24

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 1 person dropped out

Selective reporting (reporting bias)

High risk

The trial changed the primary outcomes

Vested‐interest bias

High risk

The trial was funded by Bristol‐Myers Squibb

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Izumi 2014a2

Methods

For characteristics see Izumi 2014a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Low risk

Central randomisation centre

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Unblinded after week 24

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Unblinded after week 24

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 1 person dropped out

Selective reporting (reporting bias)

High risk

The trial changed the primary outcomes

Vested‐interest bias

High risk

The trial was funded by Bristol‐Myers Squibb

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Jacobson 2010

Methods

Randomised clinical trial

Participants

35 adult participants

Sex: 18 men, 17 women

Mean age: not reported

Country: USA and Puerto Rico.

Inclusion criteria: 18‐65 years of age treatment‐naive (no prior treatment with IFN‐a +/‐ RBV regimens, discontinued IFN‐a containing regimens after < 2 weeks of therapy due to tolerability issues were considered treatment‐naive, HCV RNA > 100,000 IU/mL at screening, genotype 1, a diagnosis of chronic HCV infection for at least 6 months.

Exclusion criteria: evidence of acute or chronic infection with HIV or HBV, exposure within the previous 3 months to an investigational anti‐HCV agent, evidence of severe or decompensated liver disease, participants with liver disease unrelated to HCV infection.

Interventions

Experimental group: oral 200 mg, 300 mg, 500 mg twice daily for 4 weeks.

Control group: placebo.

Co‐intervention: standard care as per investigator's discretion up to Week 48, then off‐treatment up to Week 72 in open‐label period. Standard of care included peg‐IFN α‐2a 180 μg subcutaneously once weekly starting from day 1 and RBV 1000 mg/day tablet orally in 2 divided doses for participants weighing ≤ 75 kg; 1200 mg/day orally in 2 divided doses for participants weighing > 75 kg.

Outcomes

Plasma HCV, pharmacokinetics, ALT levels, safety assessment.

Notes

NCT00720434

We emailed Jacobson and colleagues on 21 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label after week 4

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Open‐label after week 4

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out (Jacobson 2010, described 2 dropping out)

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol were assessed

Vested‐interest bias

High risk

The trial was funded by Pfizer

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Jacobson 2014

Methods

A phase III, multicenter, randomised, double‐blind, parallel‐group trial (QUEST‐1) (NCT01289782)

Participants

394 participants

Country: Australia, Canada, Germany, Italy, Mexico, New Zealand, Puerto Rico, Romania, Russia, Spain, Ukraine, UK, and USA

Inclusion criteria: age ≥ 18 years with chronic hepatitis C infection and HCV genotype 1. Screening HCV RNA level > 10,000 IU/mL, treatment‐naive, an ultrasound performed within 6 months of enrolment showing no signs of HCC in participants with cirrhosis.

Exclusion criteria: hepatic decompensation, any non‐HCV‐related liver disease, HIV or HBV co‐infection, non‐genotype 1 HCV infection, significant laboratory abnormalities, any other active disease, male or female participants who had, or were planning to conceive

Simeprevir group: 264 participants:

Sex: 148 men, 116 women

Median age: 48 years (range 39‐54)

Race: 227 white (86%), 27 black or African‐American (10%), 5 Asian (2%)

HCV genotype 1a: 147 (56%). HCV genotype1b: 117 (44%)

Interleukin (IL) 28B genotype CC: 77 (29%). IL28B genotype CT: 150 (57%). IL28B genotype TT: 37(14)

HCV RNA > 800,000 IU/mL, n(%): 218(83)

Placebo group: 130 participants:

Sex: 74 men, 56 women

Median age: 48 years (range 36‐54)

Race: 122 white (94%), 4 black or African‐American (3%), 3 Asian (2%)

HCV genotype, n(%): 1a: 74(57). 1b: 56(43).

IL28B genotype, n(%): CC: 37(28). CT: 76(58). TT: 17(13)

METAVIR score, n(%): F0‐F1: 50(38). F2: 40(31). F3: 23(18). F4: 17(13).

HCV RNA > 800,000 IU/mL, n(%): 96(74)

Interventions

Experimental group: oral simeprevir 150 mg once daily for 12 weeks.

Control group: oral placebo 150 mg once daily for 12 weeks.

Co‐interventions:

Experimental group: peg‐IFN alfa‐2a 180 μg subcutaneously once weekly and oral weight‐based RBV 1000‐1200 mg in 2 divided daily doses for 24‐48 weeks

Control group: pegIFN α‐2a 180 μg subcutaneously once weekly and oral weight‐based RBV 1000‐1200 mg in 2 divided daily doses (1000 mg if body weight < 75 kg; 1200 mg if body‐weight ≥ 75 kg) for 48 weeks.

Outcomes

Primary outcome: proportion of participants achieving SVR12 (HCV RNA < 25 IU/mL undetectable at end of treatment and < 25 IU/mL detectable or undetectable 12 weeks after planned end of treatment)

Secondary outcomes: comparison of SVR 24 weeks after planned end of treatment. Percentage of participants meeting criteria for response‐guided therapy to complete treatment at week 24. Rapid virological response (HCV RNA < 25 IU/mL undetectable at week 4). On‐treatment failure (detectable HCV RNA at end of treatment). Incidence of viral breakthrough (HCV RNA increase of more than 1 log10 from the lowest level noted or an HCV RNA ≥ 25 IU/mL during follow‐up or at time of SVR assessments after achieving undetectable levels at end of treatment). Incidence of AEs. Incidence of laboratory abnormalities. Patient‐reported symptoms and functioning. Effect of baseline characteristics on treatment response. Assessment of depression severity. Assessment of health status.

Notes

We emailed Jacobson and colleagues on 21 April 2016 for additional information (on blinding of outcomes assessors) but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

A computer‐generated schedule prepared by or under the supervision of the sponsor was used

Allocation concealment (selection bias)

Low risk

Allocation concealment was performed by "using an interactive voice‐response system (IVRS) which assigned a unique code that dictated the treatment assignment and matching study drug kit for each patient". Randomisation codes were maintained within the IVRS

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Authors stated that "patients, study personnel, and the sponsor were masked to the treatment group assignment", the blinding method was not adequately described. A matched placebo was used

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Although RNA levels were monitored by an unmasked independent external person who informed the sponsor of any required changes to treatment, the blinding method for other outcome assessors was not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Number and reasons for discontinuation were clearly reported on

Selective reporting (reporting bias)

Low risk

Protocol is available. All pre‐specified study outcomes were reported on

Vested‐interest bias

High risk

The sponsor (Janssen Infectious Diseases‐Diagnostics) was directly involved in trial design, analyses and interpretation of data, writing and reviewing the manuscript

Other bias

Low risk

The trial seems free of other potential sources of bias

JUMP‐C 2013

Methods

Phase IIb, randomised, double‐blind, parallel‐group study in treatment‐naive participants with HCV genotype 1 or 4 infection (ClinicalTrials.gov NCT01057667)

Participants

168 participants were randomised

Sex: 118 men, 48 women

Mean age: experimental group: 49.7 years/control group: 48.2 years

Countries: 25 sites in the USA and Canada.

Inclusion criteria: eligible participants were treatment‐niıve adults 18‐70 years of age with chronic hepatitis C of at least 6 months’ duration, a serum HCV RNA titer of at least 50,000 IU/mL (COBAS AmpliPrep/ COBAS TaqMan HCV Test; lower limit of detection ¼ 15 IU/mL), and HCV genotype 1 or 4 infection were eligible for the study. Participants were required to have had a liver biopsy within the previous 24 months (36 months in participants with cirrhosis/bridging fibrosis). Participants with compensated cirrhosis (Child‐Pugh grade A) or transition to cirrhosis were required to have had an abdominal ultrasound, computerised tomography scan, magnetic resonance imaging scan demonstrating the absence of evidence of HCC (within 2 months before randomisation), and a serum alpha‐fetoprotein level < 100 ng/mL.

Exclusion criteria: infection with hepatitis A or B viruses or HIV; previous treatment with IFN‐based therapy or any investigational anti‐HCV agent; systemic antiviral therapy within the previous 3 months; history or evidence of medical condition associated with chronic liver disease other than HCV; absolute neutrophil count < 1.5 x 109 cells/L; platelet count < 90 x 109 cells/L; haemoglobin concentration < 12 g/dL in women (< 13 g/dL in men); history of renal disease, serum creatinine > 1.5 times the ULN, an estimated creatinine clearance ≤ 70 mL/min or microproteinuria.

Interventions

Participants were randomised in a 1:1 ratio. 166 participants received at least 1 dose.

Experimental group: oral mericitabine (Genentech, San Francisco, CA) 1000 mg twice a day for 24 weeks in participants with eRVR (defined as undetectable HCV RNA from week 4 through 22) or for 48 in participants without eRVR.

Control group: placebo twice a day.

Co‐intervention: peg‐IFN α‐2a (40 kD) (Pegasys; Roche, Basel,Switzerland) 180 lg subcutaneously once‐weekly and oral RBV (Copegus; Roche) at a dosage of 1000 (body weight: < 75 kg) or 1200 mg/day (body weight: > 75 kg) in 2 divided doses for 24 or 48 weeks.

Outcomes

Proportion of participants with undetectable plasma HCV RNA 24 weeks after the end of treatment (SVR24), with SAE, AEs, mortality.

Notes

We emailed Pockros and colleagues on 06 June 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

A computer‐generated randomisation list was maintained by the sponsor, and neither study personnel nor investigators had access to the list

Allocation concealment (selection bias)

Low risk

Participants were randomised by an interactive voice‐response system. A computer‐generated randomisation list was maintained by the sponsor, and neither study personnel nor investigators had access to the list

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blinding was achieved through the use of matching placebo tablets. Investigators were advised byinteractive voice‐response system at week 24 as to whether a participant was to stop treatment (mericitabine‐treated participants with an eRVR) or continue to week 48 (mericitabine‐treated participants without an eRVR and all placebo‐treated participants). JF: "I guess that all participants were not blinded to maximum‐follow up then? Since it would be obvious that the ones who stopped treatment after 24 weeks, received the study drug?"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

The trial authors reported that only 55 participants in the experimental group completed 24 weeks of follow‐up. It seems like there are 81 participants in the included analysis of SVR24. The trial authors do not account for how they imputed the participants with missing data

Selective reporting (reporting bias)

Low risk

All outcomes in the protocol were reported on

Vested‐interest bias

Unclear risk

This research was funded by F. Hoffmann‐La Roche Ltd.

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Kwo 2010a1

Methods

An open‐label, randomised, multicenter, parallel group, phase II trial (SPRINT‐1) (NCT00423670)

Participants

520 participants

Country: USA, Canada, and Europe

Inclusion criteria: chronic hepatitis C infection genotype 1 treatment‐naive, 18‐60 years. Liver biopsy consistent with chronic HCV infection within 5 years of enrolment, haemoglobin ≥ 130 g/L (men), ≥ 120 g/L (women), neutrophil count ≥ 1.5 x 109/L, platelet count ≥ 100 x 109/L. Bilirubin, albumin, and creatinine within normal limits.

Exclusion criteria: decompensated liver cirrhosis, HIV infection, previous organ transplantation, other causes of liver disease, pre‐existing psychiatric disease, seizure disorder, cardiovascular disease, haemoglobinopathies, haemophilia, poorly controlled diabetes, autoimmune diseases

Group 1: 104 participants

Sex: 70 men (67%), 34 women (33%)

Mean age ± SD: 48.3 ± 6.9 years

Race: 83 white (80%), 2 American Indian or Alaskan (2%), 3 Asian (3%), 16 black (15%), 0 multiracial

Weight, mean ± SD (kg): 83.4 ± 16.2

HCV genotype, n(%): 1a: 53(51), 1b: 42(40), 1, no subtype: 9(9)

Baseline HCV RNA: log10 of geometric mean: 6.53. > 600,000 IU/mL, n(%): 94(90). Cirrhosis, n(%): 8(8)

Group 2: 103 participants

Sex: 51 men (50%), 52 women (50%)

Mean age ± SD: 47.7 ± 7.4

Race: 85 white (83%), 1 American Indian or Alaskan (1%), 1 Asian (1%), 15 black (15%), 1 multiracial (1%)

Weight, mean ± SD (kg): 79.9 ± 14.2

HCV genotype, n(%): 1a: 53(51), 1b: 37(36), 1, no subtype: 13(13)

Baseline HCV RNA: log10 of geometric mean: 6.53. > 600,000 IU/mL, n(%): 90(87), cirrhosis, n(%): 7(7)

Group 3: 103 participants

Sex: 58 men (56%), 45 women (44%)

Mean age ± SD: 47.6 ± 8.3 years

Race: 85 white (83%), 1 American Indian or Alaskan (1%), 2 Asian (2%), 15 black (15%), 0 multiracial

Weight, mean ± SD (kg): 78.4 ± 16.5

HCV genotype, n(%): 1a: 60(58). 1b: 35(34). 1, no subtype: 8(8)

Baseline HCV RNA: log10 of geometric mean: 6.53. > 600,000 IU/mL, n(%): 93(90), cirrhosis, n(%): 6(6)

Group 4: 107 participants

Sex: 63 men (59%), 44 women (41%)

Mean age ± SD: 46.4 ± 8.0 years

Race: 86 white (80%), 0 American Indian or Alaskan, 2 Asian (2%), 18 black (17%), 1 multiracial (1%)

Weight, mean ± SD (kg): 83.4 ± 17.3

HCV genotype, n(%): 1a: 67(63), 1b: 30(28), 1, no subtype: 10(9)

Baseline HCV RNA: log10 of geometric mean: 6.64. > 600,000 IU/mL, n(%): 98(92), cirrhosis, n(%): 7(7)

Group 5: 103 participants

Sex: 63 men (61%), 40 women (39%)

Mean age ± SD: 46.7 ± 8.8

Race: 87 white (84%), 0 American Indian or Alaskan, 1 Asian (1%), 14 black (14%), 1 multiracial (1%)

Weight, mean ± SD (kg): 80.0 ± 19.4

HCV genotype, n(%): 1a: 55(53), 1b: 36(35), 1, no subtype: 12(12)

Baseline HCV RNA: log10 of geometric mean: 6.54. > 600,000 IU/mL, n(%): 94(91), cirrhosis, n(%): 9(9).

Interventions

Experimental group:

2: oral boceprevir 800 mg 3 times per day, starting at week 5 for a total of 24 weeks.

3: oral boceprevir 800 mg 3 times per day, starting at week 5 for a total of 44 weeks.

4: oral boceprevir 800 mg 3 times per day for a total of 28 weeks.

5: oral boceprevir 800 mg 3 times per day for a total of 48 weeks.

Control group:

1: no intervention.

Co‐interventions:

1‐5: peg‐IFN α‐2b 1.5 μg/kg body weight subcutaneously once weekly

‐ weight‐based oral RBV from 800‐1400 mg daily (if body weight ≤ 65 kg dosage is 800 mg (400 mg twice daily); if body weight is 66‐80 kg dosage is 1000 mg daily (400 mg in the morning and 600 mg in the evening); if body weight is 81‐105 kg dosage is 1200 mg daily (600 mg twice daily); and if body weight is > 105 kg dosage is 1400 mg daily (600 mg in the morning and 800 mg in the evening)).

Outcomes

Primary outcome: SVR, defined as the proportion of participants with undetectable HCV RNA 24 weeks after discontinuation of treatment.

Secondary outcomes:

  1. number of participants with SVR based on a 4‐week lead‐in treatment with peg‐IFN and RBV

  2. number of participants with SVR based on duration of boceprevir treatment

  3. number of participants negative for HCV RNA at week 12

  4. number of participants negative for HCV RNA at 72 weeks post randomisation

  5. number of participants with an EVR that achieved SVR

  6. number of participants with a virologic response at week 12 that achieved SVR

  7. number of participants with a virologic response at 72 weeks post randomisation that achieved SVR.

Notes

2 additional groups were present in the trial (Groups 6 and 7), which were randomised separately, but did not satisfy inclusion criteria, therefore were not included.

We emailed Kwo and colleagues on 26 April 2016 for further explanation on difference between number of SAE stated in published article compared to results published on www.ClinicalTrials.gov but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random code

Allocation concealment (selection bias)

Low risk

Allocation performed by an external randomisation centre through interactive voice‐response system in 1:1:1:1:1 ratio. Randomisation was stratified according to race (black vs non‐black) and cirrhosis status (cirrhosis vs no cirrhosis).

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Trial described as open‐label

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Trial described as open‐label

Incomplete outcome data (attrition bias)
All outcomes

High risk

Although number and reasons for withdrawal were clearly stated, the proportion of participants who discontinued treatment was high, from 26% to 50%, mostly due to AEs or treatment inefficiency.

Selective reporting (reporting bias)

High risk

Although a protocol was available and published before randomisation began, number of SAE were differently stated in the published article compared to data presented on www.ClinicalTrials.gov. Data presented in the latter were somewhat higher. Data reported are from www.ClinicalTrials.gov.

Vested‐interest bias

High risk

The sponsor of the study contributed to patient recruitment, trial management, data collection, statistical analyses, and the writing and review of the report.

Other bias

Low risk

The trial appeared to be free of other bias domains that could put it at risk of bias.

Kwo 2010a2

Methods

For characteristics see Kwo 2010a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random code

Allocation concealment (selection bias)

Low risk

Allocation performed by an external randomisation centre through interactive voice‐response system in 1:1:1:1:1 ratio. Randomisation was stratified according to race (black vs non‐black) and cirrhosis status (cirrhosis vs no cirrhosis)

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Trial described as open‐label

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Trial described as open‐label

Incomplete outcome data (attrition bias)
All outcomes

High risk

Although number and reasons for withdrawal were clearly stated, the proportion of participants who discontinued treatment was high, from 26% to 50%, mostly due to AEs or treatment inefficiency.

Selective reporting (reporting bias)

High risk

Although a protocol was available and published before randomisation began, number of SAE were differently stated in the published article compared to data presented on www.ClinicalTrials.gov. Data presented in the latter were somewhat higher. Data reported are from www.ClinicalTrials.gov

Vested‐interest bias

High risk

The sponsor of the study contributed to patient recruitment, trial management, data collection, statistical analyses, and the writing and review of the report

Other bias

Low risk

The trial appeared to be free of other bias domains that could put it at risk of bias

Kwo 2010a3

Methods

For characteristics see Kwo 2010a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random code

Allocation concealment (selection bias)

Low risk

Allocation performed by an external randomisation centre through interactive voice‐response system in 1:1:1:1:1 ratio. Randomisation was stratified according to race (black vs non‐black) and cirrhosis status (cirrhosis vs no cirrhosis)

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Trial described as open‐label

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Trial described as open‐label

Incomplete outcome data (attrition bias)
All outcomes

High risk

Although number and reasons for withdrawal were clearly stated, the proportion of participants who discontinued treatment was high, from 26% to 50%, mostly due to AEs or treatment inefficiency

Selective reporting (reporting bias)

High risk

Although a protocol was available and published before randomisation began, number of SAE were differently stated in the published article compared to data presented on www.ClinicalTrials.gov. Data presented in the latter were somewhat higher. Data reported are from www.ClinicalTrials.gov

Vested‐interest bias

High risk

The sponsor of the study contributed to patient recruitment, trial management, data collection, statistical analyses, and the writing and review of the report

Other bias

Low risk

The trial appeared to be free of other bias domains that could put it at risk of bias

Kwo 2010a4

Methods

For characteristics see Kwo 2010a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random code

Allocation concealment (selection bias)

Low risk

Allocation performed by an external randomisation centre through interactive voice‐response system in 1:1:1:1:1 ratio. Randomisation was stratified according to race (black vs. non‐black) and cirrhosis status (cirrhosis vs. no cirrhosis)

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Trial described as open‐label

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Trial described as open‐label

Incomplete outcome data (attrition bias)
All outcomes

High risk

Although number and reasons for withdrawal were clearly stated, the proportion of participants who discontinued treatment was high, from 26% to 50%, mostly due to AEs or treatment inefficiency

Selective reporting (reporting bias)

High risk

Although a protocol was available and published before randomisation began, number of SAE were differently stated in the published article compared to data presented on www.ClinicalTrials.gov. Data presented in the latter were somewhat higher. Data reported are from www.ClinicalTrials.gov

Vested‐interest bias

High risk

The sponsor of the study contributed to patient recruitment, trial management, data collection, statistical analyses, and the writing and review of the report

Other bias

Low risk

The trial appeared to be free of other bias domains that could put it at risk of bias

Lalezari 2011

Methods

Randomised clinical trial

Participants

64 participants

Mean age: 50 years

Country: USA

Inclusion criteria: treatment‐naive adult participants with chronic hepatitis C.

Interventions

Experimental group: oral 200 mg, 400 mg, 800 mg of ACH‐1625 for 28 days.

Control group: placebo.

Co‐intervention: peg‐IFN‐α 2a/RBV.

Outcomes

Pharmacokinetics, HCV RNA, safety assessment.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as placebo‐blinded but it was unclear how the blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as placebo‐blinded but it was unclear how the blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

Unclear risk

Not described

Other bias

Low risk

The trial appeared to be free of other bias domains that could put it at risk of bias

Lalezari 2012

Methods

Randomised clinical trial

Participants

41 adult participants

Sex: 29 men, 12 women

Mean age: 48 years

Country: USA

Inclusion criteria: male or female adults 18‐65 years of age, inclusive; a documented clinical history compatible with chronic hepatitis C, including the presence of HCV RNA in the plasma for least 6 months and a liver biopsy sample within 24 months with histology consistent with chronic HCV infection; HCV genotype 1, plasma HCV RNA > 5 log10 IU/ml, and anti‐HCV antibody positive at screening; and agreement by participants to use a double‐barrier method of birth.

Sex: 29 men, 12 women.

Exclusion criteria: BMI > 32 kg/m2; pregnancy or breastfeeding; co‐infection with HBV or HIV; history or evidence of decompensated liver disease; history of HCC or findings suggestive of possible HCC; other causes of liver disease; previous antiviral treatment for HCV infection; current abuse of alcohol or illicit drugs or treatment for opioid addiction; use of any known inhibitor and/or inducer of CYP 3A4 or any other investigational drugs within 30 days of dosing; abnormal laboratory values at screening (a hemoglobin level < 12.0 g/dl for males or < 11.0 g/dl for females; an absolute neutrophil count < 1.5 × 109/liter; a platelet count < 130 × 109/liter; an alanine aminotransferase [ALT] or aspartate aminotransferase [AST] level > 2.5 × upper limit of normal [ULN]; an alkaline phosphatase level > 1.25 × ULN; an albumin level < 3.5 g/dl; total bilirubin, amylase, lipase, or international normalized ratio [INR] > ULN; a serum creatinine or blood urea nitrogen value > ULN; creatinine clearance < 80 ml/min as estimated by the Cockcroft‐Gault formula; or any other laboratory abnormality > grade 1, except for asymptomatic cholesterol or triglycerides); or other clinically significant diseases that, in the opinion of the investigator, would jeopardize the safety of the patient or impact the validity of the study results.

Interventions

Experimental group: oral 25 mg, 50 mg, 75 mg, 100 mg of IDX184 for 3 days.

Control group: placebo.

Co‐intervention: 14 days after treatment the participants were offered extended therapy with peg‐IFN/RBV.

Outcomes

Safety assessment, antiviral activity.

Notes

We emailed Lalezari and colleagues on 26 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Only some outcomes were blinded for outcome assessors

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No dropouts

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

High risk

The trial was funded by Idenix pharmaceuticals Inc

Other bias

Low risk

The trial appeared to be free of other bias domains that could put it at risk of bias

Lalezari 2013

Methods

Randomised clinical trial

Participants

81 adult participants

Sex: 56 men, 25 women

Mean age: 48 years

Country: USA

Inclusion criteria: male or female participants 18‐65 years old; documented clinical history compatible with chronic hepatitis C, including positive anti‐HCV antibody or presence of HCV RNA in the plasma for at least 6 months and liver biopsy within 24 months with histology consistent with chronic hepatitis C infection; HCV‐genotype 1, plasma HCV RNA> 5 log10 IU/mL; all participants agreed to use double‐barrier birth control (such as condom plus spermicide) from screening through at least 6 months after the last dose of the study drug.

Exclusion criteria: pregnancy or breastfeeding; BMI > 35 kg/m2; co‐infection with HBV or HIV; history or evidence of decompensated liver disease; prior clinical or histological evidence of cirrhosis; ALT or AST level > 3 ULN; histology of HCC or findings suggestive of possible HCC; 1 or more additional known primary or secondary causes of liver disease, other than hepatitis C, previous antiviral treatment for HCV; current abuse of alcohol or illicit drugs; current use of any major inhibitor or inducer of cytochrome P450 3A4 or any other investigational drugs within 30 days of dosing, or other clinically significant diseases that, in the opinion of the investigator, would jeopardise the safety of the participants or affect the validity of the study results.

Interventions

Experimental groups: oral rising daily doses of 50, 100, 150 or 200 mg of IDX184 for 2 weeks.

Control group: placebo.

Co‐intervention: peg‐IFN‐ α 2a and RBV for 2 weeks. All participants received additional 2 weeks of peg‐IFN and RBV.

Outcomes

HCV RNA, Safety, pharmacokinetics.

Notes

We emailed Lalezari and colleagues on 26 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Despite being a double‐blinded study, there were different doses, syringes plus capsules, different administrations – once vs twice

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

The number of dropouts was unclear

Selective reporting (reporting bias)

High risk

Not all outcomes stated in the protocol were assessed (NCT01011166 )

Vested‐interest bias

High risk

The trial was funded by Idenix Pharmaceuticals Inc.

Other bias

Low risk

The trial appeared to be free of other bias domains that could put it at risk of bias

Larrey 2012

Methods

Randomised phase I clinical trial

Participants

27 participants

Sex: 21 men, 6 women
Mean age: 46 years

Countries: France, Germany, and Switzerland.

Inclusion criteria: treatment‐naive participants male or female (with documented hysterectomy or postmenopausal), 18–70 years of age, had chronic hepatitis C infection of genotype‐1, with a HCV viral load > 100,000 IU/mL at screening.

Exclusion criteria: cirrhosis was ruled out by biopsy or elastometry (FibroScan; cut‐off used by investigators ranged from 12.5 to 16.0 kPa) performed within 24 months prior to study enrolment. Participants with HBV or HIV co‐infection, concurrent liver disease other than HCV, past treatment with any experimental polymerase inhibitor, or hyperbilirubinaemia (> 1.5 ULN not due to Gilbert’s polymorphism).

Interventions

Experimental group: oral 400 mg, 600 mg, or 800 mg 3 times daily of BI 207127 for 28 days.

Control group: placebo.

Co‐intervention: peg‐IFN α‐2a was administered subcutaneously at a dose of 180 lg per week, and RBV was given orally at a dose of 1000 mg per day (body weight < 75 kg) or 1200 mg per day (body weight > 75 kg) in 2 divided doses. Participants were advised to use sun protection. After 4 weeks, participants were given the opportunity to continue peg‐IFN α‐2a or 2b and RBV up to week 48 at the investigators' discretion.

Outcomes

Efficacy assessment, safety assessment, drug resistance monitoring, HCV RNA, PK assessment.

Notes

NCT00905632 Only treatment‐naive participants received placebo, and could be used in the analyses.

We emailed Larrey and colleagues on 26 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as being double‐blinded but it was unclear how the blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Described as being double‐blinded but it was unclear how the blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No participants in the treatment‐naive group were lost to follow‐up

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol (NCT00905632) were assessed

Vested‐interest bias

High risk

The trial was funded by Boehringer Ingelheim

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Larrey 2013

Methods

Randomised clinical trial

Participants

60 participants

Sex: 48 men, 12 women

Mean age: 50.2 years

Inclusion criteria: treatment‐naive or treatment‐experienced participants without cirrhosis or treatment‐experienced participants with compensated cirrhosis female, aged 18‐70 years, with confirmed chronic HCV genotype 1 infection. Deleobuvir had shown activity against HCV genotype 1a and 1b in vitro; therefore, participants with either subgenotype were eligible. All participants had an HCV RNA level > 100,000 IU/mL at screening. The treatment‐experienced group included previous null responders, partial responders, and relapsers. The presence or absence of cirrhosis was confirmed by liver biopsy or transient elastography (Fibroscan 12.5 kPa).

Exclusion criteria: hepatitis B or HIV co‐infection, concurrent liver disease other than HCV, past treatment with any experimental polymerase inhibitor, planned or concurrent use of any other approved or investigational pharmacological therapy, or current drug or alcohol abuse. Participants were also excluded if they had hyperbilirubinaemia, abnormal hematologic or laboratory values at screening, or concurrent disease considered clinically significant by the investigator.

Interventions

Experimental group: rising doses of 100 mg, 200 mg, 400 mg, 800 mg, and 1200 mg every 8 h of deleobuvir (BI 207127).

Control group: placebo.

Outcomes

N25B variants, safety assessment, pharmacokinetics.

Notes

We emailed Larrey and colleagues on 26 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as double‐blinded but method was not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Described as double‐blinded but method was not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described (it was described that 3 participants dropped out due to AEs)

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol were assessed

Vested‐interest bias

High risk

The trial was funded by Boehringer Ingelheim

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Lawitz 2008

Methods

Randomised clinical trial

Participants

33 participants

Sex: 28 men, 5 women

Mean age: not described.

Inclusion criteria: treatment‐naive and treatment‐experienced noncirrhotic participants,18‐55 years old, with high viral load, genotype 1, chronic HCV infection.

Interventions

Experimental group: oral 125 mg, 600 mg of MK‐7009 once daily for 8 days or 25 mg, 75 mg, 250 mg, or 500 mg of MK‐7009 twice daily for 8 days.

Control group: placebo.

Outcomes

HCV RNA, safety assessment.

Notes

We emailed Lawitz and colleagues on 26 April 2016 for additional information on random, blinding, missing data, protocol, data, participants' characteristics, funding, number of participants in placebo/exp group but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as placebo‐blinded, but it was not described how blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Described as placebo‐blinded, but it was not described how blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

High risk

Several authors worked for several pharmaceutical companies

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Lawitz 2009

Methods

Randomised clinical trial

Participants

40 participants

Sex: not reported

Mean age: not reported

Country: USA

Inclusion criteria: participants both treatment‐naive and treatment‐experienced with chronic HCV 1

Interventions

The trial was divided into 4 cohorts, with different experimental intervention.

Experimental group: oral 100 mg or 200 mg of VCH‐916 3 times daily for 14 days. Oral 300 or 400 mg of VCH‐916 twice daily for 3 days.

Control group: placebo.

Outcomes

Safesty assessment, HCV RNA level, pharmacokinetics

Notes

We emailed Lawitz and colleagues on 26 April 2016 for additional information on random, blinding, missing data, protocol, data, participants characteristics, funding, number of participants in placebo/exp group but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as double‐blinded, but it was not described how blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Described as double‐blinded, but it was not described how blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All participants completed the study

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

Unclear risk

Not described

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Lawitz 2010a

Methods

Randomised clinical trial

Participants

54 participants

Country: USA

Inclusion criteria: Adult treatment‐naive participants in genotype 1 HCV participants.

Interventions

Experimental group: oral 25 mg, 75 mg, or 200 mg of GS‐9256 twice daily, or 300 mg of GS‐9256 once daily for 3 days.

Control group: placebo.

Outcomes

Safesty assessment, HCV RNA level, pharmacokinetics.

Notes

We emailed Lawitz and colleagues on 26 April 2016 for additional information on random, blinding, missing data, protocol, data, participants characteristics, funding, number of participants in placebo/exp group but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as double‐blinded, but it was not described how blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Described as double‐blinded, but it was not described how blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

High risk

Several authors worked for Gilead Sciences

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Lawitz 2010b

Methods

Randomised clinical trial

Participants

63 participants

Inclusion criteria: non‐cirrhotic treatment‐naive adult participants with genotype 1 HCV participants.

Exclusion criteria: not described.

Interventions

The trial used 3 cohorts

Experimental group: oral 100 mg, 200 mg, or 400 mg of PSI‐7977 once daily for 28 days.

Control group: placebo.

Co‐intervention: epg‐IFN/RBV.

Outcomes

HCV RNA level, pharmacokinetics.

Notes

We emailed Lawitz and colleagues on 26 April 2016 for additional information on random, blinding, missing data, protocol, data, participants characteristics, funding, number of participants in placebo/exp group but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as double‐blinded, but it was not described how blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Described as double‐blinded, but it was not described how blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

High risk

Main author worked for several pharmaceutical companies

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Lawitz 2010c

Methods

Randomised clinical trial

Participants

63 participants

Inclusion criteria: participants received at least 1 dose of the drug and in cohort 200 mg twice a day adults with hepatitis C genotype 1.

Interventions

Experimental group: 200 mg or 400 mg of ANA598 twice a day.

Control group: placebo.

Co‐intervention: 12 weeks of standard of care treatment.

Outcomes

Safety, antiviral activity, pharmacokinetics.

Notes

Only the cohort with 200 mg is reported here. We emailed Lawitz and colleagues on 26 April 2016 for additional information on sequence generation, blinding, incomplete outcome data, number of deaths, SVR24 but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as double‐blind and placebo controlled, but the placebo was not further described.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was unclear how many participants were actually randomised to the experimental and control group and therefore, it is unclear how many participants are with missing data

Selective reporting (reporting bias)

High risk

The trial did not report on the level of RBV and peg‐IFN in the blood as is stated in the protocol (NCT00978497)

Vested‐interest bias

High risk

The trial was supported by a company with an interest in a given result Hoffmann‐La Roche

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of other bias

Lawitz 2011a

Methods

Randomised clinical trial

Participants

35 participants

Sex: 25 men, 10 women

Mean age: 50 years

Country: USA

Inclusion criteria: treatment‐naive adults diagnosed with hepatitis C genotype 1.

Exclusion criteria: not described.

Interventions

The trial used different experimental groups, with different doses of ABT‐450.

Experimental group: 50 mg ABT‐450 + 100 mg RBV, 100 mg ABT‐450 + 100 mg RBV, 200 mg ABT‐450 + 100 mg RBV once daily for 3 days.

Control group: placebo.

Co‐intervention: peg‐IFN α‐2a 180 mg/week + weight‐based RBV 1000–1200 mg/day (standard of care) for 12 weeks. After week 12, participants received standard of care treatment alone for 36 weeks.

Outcomes

Safety assessment, HCV RNA level, pharmacokinetics.

Notes

We emailed Lawitz and colleagues on 26 April 2016 for additional information on randomisation, blinding, missing data, protocol, data, funding, IL28b data but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as placebo‐controlled, but it was not described how blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Described as placebo‐controlled, but it was not described how blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out of the placebo group

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

High risk

Several authors worked for Gilead Sciences

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Lawitz 2011b

Methods

Randomised clinical trial

Participants

252 participants

Sex: 151 men, 101 women

Countries: USA and Europe.

Inclusion criteria: non cirrhotic treatment‐naive adult participants with chronic hepatitis C genotype 1.

Exclusion criteria: not described.

Interventions

Experimental group 1: oral tegobuvir 40 mg twice daily for 48 weeks.
Experimental group 2: oral tegobuvir 40 mg response‐guided for 24‐48 weeks.

Control group: placebo.

Co‐intervention: peg/RBV.

Outcomes

Safety assessment, pharmacokinetics, HCV RNA.

Notes

We emailed Lawitz and colleagues on 26 April 2016 for additional information on randomisation, blinding, missing data, protocol, complete trial, data, funding but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as placebo‐controlled, but it was not described how blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Described as placebo‐controlled, but it was not described how blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% percent dropped out

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

High risk

Several authors worked for Gilead Sciences

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Lawitz 2012a

Methods

Randomised clinical trial

Participants

72 adult participants

Sex: 52 men, 20 women
Mean age: 48 years

Country: USA

Inclusion criteria: 18–65 years of age, with chronic infection with genotype 1a or 1b HCV virus and plasma HCV RNA > 5 log10 IU/mL at screening. Participants were HCV treatment‐naive and had a BMI of 19–35 kg/m2 inclusive, creatinine clearance > 70 mL/min, and a QTcF interval < 450 ms.

Exclusion criteria: known cirrhosis, hepatic decompensation, excessive ongoing alcohol intake, Gilbert’s syndrome, evidence of HCC, co‐infection with HIV or HBV, prothrombin time > 1.5 ULN, albumin < 3 g/dL, ALT and AST levels > 5 ULN, total bilirubin > ULN, hemoglobin < 11 g/dL, platelets < 90,000/mm3, or absolute neutrophil count < 1000 cells/mm3 (< 900 cells/mm3 for African Americans). Concomitant prescription or non‐prescription medications were prohibited during the study unless prior approval was received from the medical monitor. The only exception was the use of hormonal contraception; additional double barrier method contraception was mandated for all women of childbearing potential.

Interventions

The trial was divided into 6 different cohorts, and randomised to experimental intervention or placebo.
Experimental group: oral 1 mg, 3 mg, 10 mg (genotype 1a), 10 mg (genotype 1b), 30 mg, or 90 mg of GS‐5885 for 3 days.
Control group: placebo.

Outcomes

Safety assessment, pharmacokinetics, HCV RNA, viral sequencing.

Notes

We emailed Lawitz and colleagues on 26 April 2016 for additional information on allocation, blinding, protocol, separate data from IL28b but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Centralised randomisation schedule generated via computer by the sponsor’s Biometrics group

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as double‐blinded, but it was not described how blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Described as double‐blinded, but it was not described how blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 1 person dropped out

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

High risk

This trial was supported by Gilead Sciences

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Lawitz 2012b

Methods

Randomised clinical trial

Participants

90 participants

Country: USA

Inclusion criteria: treatment‐naive adult participants with chronic hepatitis C genotype 1.

Exclusion criteria: not described.

Interventions

The trial was divided into 9 cohorts

Experimental group: oral 50 mg, 100 mg, or 300 mg of GS‐6620 once daily administered for 5 days. Oral 100 mg, 300 mg, or 900 mg of GS‐6620 once daily administered for 5 days. Oral 450 mg or 900 mg of GS‐6620 twice daily administered for 5 days.

Control group: placebo.

Outcomes

Safety assessment, pharmacokinetics, HCV RNA

Notes

We emailed Lawitz and colleagues on 26 April 2016 for additional information on randomisation, blinding, missing data, protocol, data, funding, SAE (non‐treatment related) but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as placebo‐controlled, but it was not described how blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Described as placebo‐controlled, but it was not described how blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

High risk

Several authors worked for Gilead Sciences

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Lawitz 2013a1

Methods

Randomised clinical trial

Participants

122 participants

Sex: 73 men, 49 women

Mean age: 49.4 years

Country: USA

Inclusion criteria: treatment‐naive participants HCV genotypes 1 had to have an HCV RNA concentration of 50,000 IU/mL or greater. HCV genotypes participants had a liver biopsy within 36 months before enrolment. Inclusion criteria also included the following haematological and biochemical laboratory variables: a neutrophil count of 1.5 × 10⁹/L (or ≥ 1.25 × 10⁹/L for black participants), a haemoglobin concentration of 11 g/dL or higher in women or 12 g/dL or higher in men, a platelet count of greater than 90 × 10⁹/L, total bilirubin within 2 times the ULN (21 μmol/L), and an albumin concentration of 30 g/L or lower.

Exclusion criteria: cirrhosis, HBV or HIV, psychiatric illness, pulmonary or cardiac disease, seizure disorder, or other serious comorbid disorders.

Interventions

Experimental group: oral 200 mg, or 400 mg of sofosbuvir once daily for 12 weeks.

Control group: placebo.

Co‐intervention: 48 weeks of peg‐IFN 180 μg per week subcutaneously; RBV was dosed according to weight (ie, participants < 75 kg received 1000 mg and those > 75 kg received 1200 mg; RBV was given in 2 daily doses. 400 mg in the morning and 600 mg in the evening for participants receiving 1000 mg a day, or 600 mg in the morning and 600 mg in the evening for participants receiving 1200 mg a day).

Outcomes

Virological response, pharmacokinetics, AEs.

Notes

We emailed Lawitz and colleagues on 26 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random code

Allocation concealment (selection bias)

Low risk

Interactive online response system

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out

Selective reporting (reporting bias)

High risk

The trial added additional secondary outcomes

Vested‐interest bias

High risk

The trial was funded by Gilead Sciences

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Lawitz 2013a2

Methods

For characteristics see Lawitz 2013a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random code

Allocation concealment (selection bias)

Low risk

Interactive online response system

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out

Selective reporting (reporting bias)

High risk

The trial added additional secondary outcomes

Vested‐interest bias

High risk

The trial was funded by Gilead Sciences

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Lawitz 2013b

Methods

Randomised clinical phase I, multicentre trial

Participants

44 participants

Sex: 32 men, 9 women

Median age: 49 years

Country: USA

Inclusion criteria: 18‐65 years of age and had chronic HCV 1a or 1b and plasma HCV RNA > 5 log10 IU/mL at screening. Participants were HCV treatment‐naive and had a BMI of 19‐35 kg/m2 inclusive, creatinine clearance > 60 mL/min and a QTcF interval < 450 ms.

Exclusion criteria: cirrhosis, hepatic decompensation, excessive ongoing alcohol intake, Gilbert's syndrome, evidence of HCC, co‐infection with HIV or HBV, ALT or AST levels > 5 x ULN, total bilirubin > ULN, haemoglobin < 11 g/dL, or absolute neutrophil count 1000 cells/mm2 (750 cells/mm2). Concomitant prescription during the study unless prior approval was received from the medical monitor. Participants using hormonal contraception were required to employ 2 additional barrier methods of contraception.

Interventions

The trial divided into 4 cohorts, and randomised to experimental group or control group

Experimental group: oral 60 mg, 200 mg (genotype 1a), 200 mg (genotype 1b), or 400 mg of GS‐9451 once daily for 3 days.

Control group: placebo.

Outcomes

Antiviral response, sequence analyses, pharmacokinetics, safety assessment.

Notes

We emailed Lawitz and colleagues on 26 April 2016 for additional information on allocation, blinding (placebo pill), protocol but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

It was described that all were blinded, however it was not stated if there were any similarities between the placebo pill and intervention

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It was described that all were blinded, however it was not stated if there were any similarities between the placebo pill and intervention

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There were above 5% dropouts. 3 participants were not included in the efficacy analyses. In addition, 3 participants were withdrawn after enrolment and not included in any analysis due to unknown reasons. It was unclear how the trial handled missing data.

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

High risk

The trial was funded by Gilead Sciences

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Lawitz 2013c

Methods

Randomised phase IIb clinical trial

Participants

211 participants

Sex: 131 men, 80 women

Mean age: 49.5 years

Countries: Australia, Austria, Belgium, Canada, Chile, Czech Republic, France, Germany, Israel, Korea, Lithuania, New Zealand, Poland, South Korea, Sweden, Taiwan, Thailand, UK, and USA

Inclusion criteria: treatment‐experienced non‐cirrhotic adults chronic genotype 1 HCV‐infected participants whose previous treatments with P/R had failed, a minimum of 25% of participants prior null responders, men and women 18–65 years of age, and baseline HCV RNA > 4 x 105 IU/mL.

Exclusion criteria: non‐HCV‐related chronic hepatitis, HIV co‐infection, evidence of cirrhosis on liver biopsy or approved non‐invasive imaging, or any other condition contraindicated for treatment with P/R

Interventions

4 different experimental arms

Experimental group 1: oral MK‐7009 600 mg twice daily for 24 weeks.
Experimental group 2: oral MK‐7009 600 mg twice daily for 24 weeks and 24 weeks of placebo for 24 weeks.

Experimental group 3: oral MK‐7009 300 mg twice daily for 48 weeks.

Experimental group 4: oral MK‐7009 600 mg twice daily for 48 weeks.

Control group: placebo for 48 weeks.

Co‐intervention: peg‐IFN 180 μg weekly and RBV 1000–1200 mg/day for 24–48 weeks.

Outcomes

Safety assessment, SVR.

Notes

We emailed Lawitz and colleagues on 26 April 2016 for additional information on randomisation, blinding, dealing with missing data, baseline characteristics for IL28B genotype but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Above 5% dropouts, and it was unclear how the trial handled missing data

Selective reporting (reporting bias)

Low risk

All outcomes in the protocol were reported on. NCT00704405

Vested‐interest bias

High risk

The trial was funded by Merck, Sharp & Dohme Corp

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Lawitz 2013d

Methods

Randomised clinical trial

Participants

40 participants

Sex: 36 men, 4 women

Mean age: 43 years

Country: USA

Inclusion criteria: treatment‐naive ages between 18 and 65 years, non‐cirrhotic chronic HCV genotype 1 infection and HCV RNA levels of 50,000 IU/mL ages with BMIs ranging from 18‐36 kg/m2

Exclusion criteria: women were to be surgically sterile, postmenopausal for at least 12 months at screening, or taking protocol‐specified contraceptive measures. Positive for anti‐hepatitis A virus immunoglobulin M (IgM) antibodies, hepatitis B surface antigen, anti‐hepatitis B core protein IgM antibodies, or anti‐HIV antibodies. No medication associated with QT interval prolongation was permitted within 30 days prior to dosing or during the study, and any other concurrent medication required approval by the investigator and the sponsor. Participants who had received any systemic antineoplastic or immunomodulatory treatment within 6 months prior to the first dose of study drug or who might have needed such treatments at any time.

Interventions

Participants were randomised in 4 cohorts with different doses of GS‐9851

Experimental group: 3 days of either 50 mg, 100 mg, 200 mg, or 400 mg as oral intake of GS‐9851.

Control group: placebo.

Outcomes

Pharmacokinetics, clinical virology assessment, safety and tolerability assessment.

Notes

We emailed Lawitz and colleagues on 26 April 2016 for additional information on protocol, randomisation, blinding but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as double‐blinded, however it was not stated how the blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Described as double‐blinded, however it was not stated how the blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All completed the study

Selective reporting (reporting bias)

Unclear risk

It was stated that there was a protocol, however the protocol could not be found.

Vested‐interest bias

High risk

The trial was funded by Pharmasset, Inc. Severina Moreira and Justin Cook of Niche Science and Technology Ltd.

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Lawitz 2013e

Methods

Randomised clinical trial

Participants

40 participants

Sex: 33 men, 7 women
Mean age: 46 years

Country: USA

Inclusion criteria: participants aged 18–55 years with a BMI 18.5 to 636 kg/m2 and chronic, compensated, genotype1 HCV infection. All participants had a baseline HCV RNA > 106 IU/mL and no evidence of cirrhosis or bridging fibrosis (according to biopsy within 3 years of screening). Participants also had laboratory values within pre‐specified criteria at study entry.

Exclusion criteria: participants previously treated with approved HCV therapy or with a DAA for HCV, or with chronic HBV or HIV infection were excluded.

Interventions

Experimental group: received different doses of vaniprevir orally, for 8 days twice daily (25 mg, 75 mg, 250 mg, 500 mg, 700 mg) or 8 days once daily (125 mg, 600 mg).
Control group: matching placebo.

Outcomes

Safety, tolerability and efficacy, pharmacokinetics, medication adherence.

Notes

We emailed Lawitz and colleagues on 26 April 2016 for additional information on allocation concealment, blinding of outcome assessors, sample size and protocol for trial 1 and 2 but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated centralised randomisation

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Matching placebo delivered in equal amounts

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only on person dropped out

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol were assessed

Vested‐interest bias

High risk

This study was funded by Merck Sharp & Dohme Corp

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Lawitz 2013f

Methods

Randomised clinical trial

Participants

38 adult participants

Country: USA

Inclusion criteria: chronic hepatitis C genotype 1 either with cirrhosis, or without cirrhosis

Interventions

Experimental group: oral 100 mg or 400 mg of ACH‐2684 once daily for 3 days. Oral 400 mg of ACH‐2684 twice daily.

Control group: placebo.

Outcomes

Safety assessment, pharmacokinetics, HCV RNA.

Notes

We emailed Lawitz and colleagues on 26 April 2016 for additional information on randomisation, blinding, missing data, protocol, data, funding, SAE, participants in each group but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as placebo‐controlled, but it was not described how blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Described as placebo‐controlled, but it was not described how blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

High risk

Several authors worked for Achillion pharmaceuticals

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Lawitz 2014a

Methods

Randomised clinical trial

Participants

100 participants

Sex: 65 men, 35 women

Country: USA

Inclusion criteria: compensated cirrhotic adults with chronic HCV genotype 1 infection.

Exclusion criteria: not described.

Interventions

Experimenatal group: oral 250 mg of GS‐9669 once daily for 8 weeks or oral 500 mg of GS‐9669 once daily for 8 weeks.

Control group: RBV.
Co‐intervention: ledipasvir and sofosbuvir.

Outcomes

Adverse events, HCV RNA SVR12

Notes

We emailed Lawitz and colleagues on 26 April 2016 for additional information on random, blinding, missing data, protocol, data separate from the groups, participants characteristics, funding, IL28b‐databut reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

High risk

Several authors worked for several pharmaceutical companies

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Lawitz 2015

Methods

Randomised clinical trial

Participants

85 adult participants

Sex: 68 men, 19 women

Mean age: 47 years

Countries: USA and Puerto Rico

Inclusion criteria: 18–65 years, with treatment‐naive chronic genotype 1–6 HCV infection and HCV RNA levels ≥5 log10 IU/mL at screening. Participants were required to have a BMI of 19–34 kg/m2 inclusive, creatinine clearance > 70 mL/min and QTcF ≤450 ms for men and ≤470 ms for women.

Exclusion criteria: co‐infected with HBV or HIV, had prior treatment with a HCV NS5Ainhibitor, evidence of cirrhosis or HCC, history of clinical hepatic decompensation (e.g. ascites, jaundice, encephalopathy or variceal haemorrhage) or any other clinically significant condition other than chronic HCV infection.

Interventions

The trial was divided into 11 dosing cohorts: 5 cohorts of participants with genotype 1a infection; 1 cohort of participants with genotype 1b infection, 1 cohort of participants with genotype 2 infection, 3 cohorts of participants with genotype 3 HCV infection and 1 cohort of participants with genotype 4 HCV infection.

Experimental group: oral GS‐5816 (5 mg, 25 mg, 50 mg, 100 mg, 150 mg).
Control group: matching placebo.

Outcomes

Safety assessment, efficacy analysis, pharmacokinetic analysis.

Notes

ClinicalTrials.gov number: NCT01740791. The trial reported that 87 participants were randomised, however it was also stated that those with genotype 4 (n = 2) were not randomised. Therefore we could not use data from the combined 150 mg group, as the non‐randomised genotype 4 participants were included in this group. We emailed Lawitz and colleagues on 26 April 2016 for additional information on allocation, blinding, how the trial handled missing data but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Central computer‐generated randomisation scheme, by the sponsor

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Matching placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There were more than 5% dropouts and it was unclear how the trial handled missing data.

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol were reported on

Vested‐interest bias

High risk

The study was funded by Gilead Sciences

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Liu 2015a

Methods

Randomised clinical phase Ib trial

Participants

48 participants

Sex: 48 men

Country: USA

Inclusion criteria: non‐cirrhotic participants aged 18‐60 years (up to 65 years old at the discretion of the investigator) with HCV RNA levels of > 100,000 IU/mL

Interventions

The trial was divided into cohorts, in which randomisation was performed

Experimental group: 5 mg, 10 mg, and 50 mg once daily of MK‐8742 for participants infected with genotype 1a or 1b, and 10 mg, 50 mg, and 100 mg once daily of MK‐8742 for participants infected with genotype 3.

Control group: placebo.

Outcomes

Activity, pharmacokinetics, safety

Notes

We emailed Liu and colleagues on 26 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described

Selective reporting (reporting bias)

High risk

The trial did not assess safety (NCT01532973)

Vested‐interest bias

High risk

The trial was funded by Merck Sharp & Dohme Corp

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Mallalieu 2014

Methods

Randomised clinical trial

Participants

35 participants

Sex: 24 men, 11 women

Mean age: 47.6 years

Inclusion criteria: treatment‐naive male or female participants with chronic hepatitis C aged 18‐55 years, with a BMI of 18‐35 were eligible for a multicenter, double‐blind, randomised, placebo‐controlled study. Participants were required to have HCV genotype 1a or 1b infection, a serum HCV RNA concentration greater than 75,000 IU/mL, a serum ALT concentration under 5 times the ULN, and compensated liver disease.

Exclusion criteria: participants with evidence of cirrhosis or decompensated liver disease were excluded, as were participants with a history of or current alcohol abuse, poorly controlled insulin‐dependent diabetes, unstable or poorly controlled asthma, congestive heart failure, unstable cardiopulmonary disease, renal disease, or seizure disorder. Eligible participants in all studies were required to have a negative urine drug screen, serum pregnancy test (if female), and to have a negative hepatitis B surface antigen test and anti‐HIV antibody test. Pregnant and breast feeding female participants were ineligible. Other exclusion criteria included donation of 4500 mL of blood within 30 days (participants with chronic hepatitis C).

Interventions

Experimental group: sequential cohorts of participants were randomly assigned to receive setrobuvir 200 mg, 400 mg, or 800 mg twice a day for 3 days.

Control group: received placebo for 3 days.

Outcomes

Safety, kinetics, antiviral activity.

Notes

5 participants originally enrolled in cohort 2 (400 mg twice a day) were dosed incorrectly. These participants received setrobuvir 200 mg twice a day and were thus included with cohort 1 in the analysis. We emailed Mallalieu and colleagues on 26 April 2016 for additional information random sequence generation + allocation, participants completing the study, blinding but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as double‐blinded but there was no further description of the placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It is unclear if any participants dropped out

Selective reporting (reporting bias)

Low risk

The trial reports all outcomes stated in the protocol (NCT00782353)

Vested‐interest bias

High risk

The trial was sponsored by a company that might have an interest in a given outcome (Hoffmann‐La Roche)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Manns 2011

Methods

Randomised clinical trial

Participants

53 participants were randomised

Sex: 27 men, 7 women

Mean age: 48.9 years

Inclusion criteria: participants with chronic HCV infection of genotype‐1 were recruited to the study, if they were treatment‐naive (no prior therapy with IFN, peg‐IFN, or RBV) or treatment‐experienced (virologic failure during or after treatment with an approved dose of peg‐IFN combined with RBV), had HCV RNA P100,000 IU/mL and were aged 18 years or older.

Exclusion criteria: participants with liver cirrhosis, hyperbilirubinaemia (> 1.5x ULN; participants with Gilbert’s disease were accepted), HIV, or HBV co‐infection were excluded. Furthermore, participants who had previously received any treatment with a protease inhibitor and women of child‐bearing potential not agreeing or able to use medically accepted contraception throughout the study were excluded.

Interventions

Experimental group: treatment‐naive participants: BI201335 monotherapy (20 mg, 48 mg, 120 mg, and 240 mg once a day) for 14 days, participants with a HCV RNA decrease P1 log10 from baseline (on Day 10), BI201335 treatment was combined with peg‐IFN α‐2a (180 lg/week) and RBV (1000 mg or 1200 mg/day) from Days 14 to 28.

Control group: placebo combined with peg‐IFN α‐2a and RBV. All participants were offered to extend standard of care to Week 48, with an additional 24 weeks of follow‐up.

Co‐intervention: peg‐IFN α‐2a (180 lg/week) and RBV (1000 mg or 1200 mg/day).

Outcomes

Primary: virologic response, AEs, SAE, laboratory test abnormalities.

Secondary: viral load reduction, change from baseline in viral load, rapid virological response, early virological response, complete early virological response 1+2, end of treatment response and SVR

Notes

We emailed Manns and colleagues on 26 April 2016 for additional information on allocation concealment, random sequence generation, unpublished data, dealing with missing data, SVR data and AE, il28b and blinding in general. Data on SAEs and non‐SAEs distinguishing between treatment‐naive and treatment‐experienced but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The study was described as double‐blinded and but it was unclear how the blinding was maintained

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The study was described as double‐blinded but it was unclear how the blinding was maintained and who performed the outcome assessment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 1 participant dropped out (The reason for the discontinuation of 1 participant was the diagnosis of an unexpected pregnancy of his partner representing an exclusion criterion for treatment with RBV)

Selective reporting (reporting bias)

Low risk

A protocol was found (NCT00793793) and all outcomes were reported on

Vested‐interest bias

High risk

"Michael Manns has received grant support, contributed to clinical trials, and is a member of a speaker bureau and/or consulted for Schering Plough, Roche, Merck, Bristol‐Myers Squibb, Vertex, Tibotec, Astra/Arrows, Novartis, Human Genome Sciences, Boehringer Ingelheim, and Valeant. Peter W. White, Jerry Stern, Gerhard Steinmann, Chan‐Loi Yong, George Kukolj, Joe Scherer and Wulf O. Boecher are employees of Boehringer Ingelheim."

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Manns 2012a1

Methods

Randomised clinical trial

Participants

95 participants were randomised

Sex: 55 men, 39 women

Mean age: 46.2 years

Inclusion criteria: adult, treatment‐naive participants with chronic, compensated, HCV genotype 1 infection, defined as HCV RNA levels ≥ 4 × 105 IU/mL at screening (i.e. within 75 days preceding the first dose of vaniprevir or placebo), were enrolled. All participants had positive serology for HCV or detectable HCV RNA ≥ 6 months before study initiation.

Exclusion criteria: Participants with evidence of cirrhosis by histology, imaging, or physical findings were excluded.

Interventions

Experimental group:

  1. 300 mg twice a day plus open‐label peg‐IFN α‐2a and RBV 180 μg/week + 1000 mg‐1200 mg/day for 28 days.

  2. 600 mg twice a day plus open‐label peg‐IFN α‐2a and RBV 180 μg/week + 1000 mg‐1200 mg/day for 28 days.

  3. 600 mg once a day plus open‐label peg‐IFN α‐2a and RBV 180 μg/week + 1000 mg‐1200 mg/day for 28 days.

  4. 800 mg once a day plus open‐label peg‐IFN α‐2a and RBV 180 μg/week + 1000‐1200 mg/day for 28 days.

Control group: placebo plus open‐label peg‐IFN α‐2a and RBV 180 μg/week + 1000 mg‐1200 mg/day for 28 days.

Co‐intervention: peg‐IFN α‐2a and RBV 180 μg/week + 1000 mg‐1200 mg/day.

Outcomes

Primary: proportion of participants achieving RVR. AEs and participants that discontinued due to AEs.

Exploratory: proportion of participants achieving EVR, proportion of participants achieving SVR.

Notes

We emailed Manns and colleagues on 26 April 2016 for additional information on allocation concealment, unpublished data, correlation of il28b genotype data and SVR but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Central randomisation procedure by an interactive voice‐response system

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The study was described as double‐blinded to investigator and participant

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The study was described as double‐blinded but it was unclear how the blinding was maintained and who performed the outcome assessment

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

15 participants dropped out

Selective reporting (reporting bias)

Unclear risk

A protocol was found (NCT00704184), primary objectives were reported correctly, secondary outcomes changed and new exploratory outcomes were reported in the paper

Vested‐interest bias

High risk

This study was funded by Merck Scharp and Dohme Corp.

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Manns 2012a2

Methods

For characteristics see Manns 2012a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Central randomisation procedure by an interactive voice‐response system

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The study was described as double‐blinded to investigator and participant

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The study was described as double‐blinded but it was unclear how the blinding was maintained and who performed the outcome assessment.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

15 participants dropped out

Selective reporting (reporting bias)

Unclear risk

A protocol was found (NCT00704184), primary objectives were reported correctly, secondary outcomes changed and new exploratory outcomes were reported in the paper

Vested‐interest bias

High risk

This study was funded by Merck Scharp and Dohme Corp.

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Manns 2012a3

Methods

For characteristics see Manns 2012a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Central randomisation procedure by an interactive voice‐response system

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The study was described as double‐blinded to investigator and participant

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The study was described as double‐blinded but it was unclear how the blinding was maintained and who performed the outcome assessment.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

15 participants dropped out

Selective reporting (reporting bias)

Unclear risk

A protocol was found (NCT00704184), primary objectives were reported correctly, secondary outcomes changed and new exploratory outcomes were reported in the paper.

Vested‐interest bias

High risk

This study was funded by Merck Scharp and Dohme Corp.

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Manns 2012a4

Methods

For characteristics see Manns 2012a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Central randomisation procedure by an interactive voice‐response system

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The study was described as double‐blinded to investigator and participant

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The study was described as double‐blinded but it was unclear how the blinding was maintained and who performed the outcome assessment.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

15 participants dropped out

Selective reporting (reporting bias)

Unclear risk

A protocol was found (NCT00704184), primary objectives were reported correctly, secondary outcomes changed and new exploratory outcomes were reported in the paper.

Vested‐interest bias

High risk

This study was funded by Merck Scharp and Dohme Corp.

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Manns 2014a

Methods

A phase III, randomised, double‐blind, placebo‐controlled, parallel‐design trial (QUEST‐2)(NCT01290679)

Participants

391 participants

Location: 14 countries in Europe, North America, and South America

Inclusion criteria: age ≥ 18 years. Chronic hepatitis C infection. HCV genotype 1. HCV RNA level at screening > 100,000 IU/mL. Treatment‐naive. An ultrasound performed within 6 months of enrolment showing no signs of HCC in participants with cirrhosis.

Exclusion criteria: hepatic decompensation. Any non‐HCV‐related liver disease. HIV or HBV co‐infection. Non‐genotype 1 HCV infection. Significant laboratory abnormalities. Any other active disease. Male or female participants who had, or were planning to conceive.

Simeprevir group: 257 participants

Sex: 140 men, 117 women

Median age: 46 years (range 18‐73)

Race: 237 white (92%), 16 black or African American (6%), 2 Asian (< 1%), and 2 other (< 1%)

HCV genotype 1a: 105 (41%), HCV genotype 1b: 150 (58%), other HCV genotype: 2 (< 1%)

IL28B genotype CC: 75 (29%), IL28B genotype CT: 142 (55%), IL28B genotype TT: 40 (16%)

METAVIR score F0‐F1: 130 (52%), METAVIR score F2: 65 (26%), METAVIR score F3: 36 (15%), METAVIR score F4: 17 (7%)

HCV RNA > 800,000 IU/mL, n(%): 199(77).

Placebo group: 134 participants

Sex: 77 men, 57 women

Median age: 47 years (range 18‐73)

Race: 123 white (92%), 10 black or African‐American (10%), 1 Asian (< 1%), and 0 other

HCV genotype 1a: 54 (41%), HCV genotype 1b: 77 (58%), other HCV genotype: 2 (2%)

IL28B genotype CC: 42 (31%), IL28B genotype CT: 71 (53%), IL28B genotype TT: 21 (16%)

METAVIR score, n(%): METAVIR score F0‐F1: 60 (45%), METAVIR score F2: 42 (31%), METAVIR score F3: 17 (13%), METAVIR score F4: 15 (11%)

HCV RNA > 800,000 IU/mL, n(%): 98(73).

Interventions

Experimental group: oral simeprevir 150 mg once daily for 12 weeks.

Control group: oral placebo 150 mg once daily for 12 weeks.

Co‐interventions:

Experimental group: peg‐IFN α‐2a 180 μg subcutaneously once weekly or peg‐IFN α‐2b 1.5 μg/kg body weight subcutaneously once weekly and oral weight‐based RBV 1000 mg to 1200 mg in 2 divided daily doses (1000 mg if body weight < 75 kg; 1200 mg if body weight ≥ 75 kg) for 24‐48 weeks

Control group: peg‐IFN α‐2a 180 μg subcutaneously once weekly or peg‐IFN α‐2b 1.5 μg/kg body weight subcutaneously once weekly and oral weight‐based RBV 1000 to 1200 mg in 2 divided daily doses (1000 mg if body weight < 75 kg; 1200 mg if body weight ≥75 kg) for 48 weeks.

Outcomes

Primary outcome: proportion of participants achieving SVR12 (HCV RNA < 25 IU/mL undetectable at end of treatment and < 25 IU/mL detectable or undetectable 12 weeks after the planned end of treatment).

Secondary outcomes: proportion of participants meeting criteria for response‐guided therapy to complete treatment at week 24. RVA (HCV RNA < 25 IU/mL undetectable at week 4). Activity, safety, and tolerability of simeprevir in the 2 subpopulations of participants who were given peg‐IFN α‐2a or 2b. On‐treatment failure (detectable HCV RNA at end of treatment). Incidence of viral relapse (HCV RNA ≥ 25 IU/mL during follow‐up or at the time of SVR assessments in participants with undetectable levels at end of treatment). Incidence of AEs. Incidence of laboratory abnormalities. Quality‐of‐life measures. SVR at 24 weeks after the planned end of treatment. Assessment of depression severity. Assessment of health status. Assessment of polymorphisms (HCV NS3 protease domain) at baseline and their correlation with efficacy of simeprevir plus peg‐IFN and RBV.

Notes

We emailed Manns and colleagues on 26 April 2016 for additional information blinding of outcome assessors but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"A computer‐generated randomisation schedule that was prepared by or under the supervision of the sponsor before the study was used"

Allocation concealment (selection bias)

Low risk

Concealment of allocation was obtained by using an interactive web‐based or voice‐response system

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Authors stated that "patients, study personnel, and the sponsor were masked to the treatment group assignment", the blinding method was not adequately described. A matched placebo was used

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It was not mentioned if the outcome assessors were blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Number and reasons for discontinuation were clearly reported on

Selective reporting (reporting bias)

Low risk

Protocol was available. All pre‐specified study outcomes were reported on

Vested‐interest bias

Unclear risk

"The sponsor (Janssen Infectious Diseases‐Diagnostics) was directly involved in trial design, data analyses and interpretation, and writing and reviewing the manuscript."

Other bias

Low risk

The trial seems to be free of other potential sources of bias

Marcellin 2013a

Methods

Randomised clinical trial

Participants

20 participants

Inclusion criteria: treatment‐naive for chronic hepatitis C

Countries: France, Moldova, Romania, USA

Interventions

Experimental group: oral ALS‐2200 200 mg once daily for 7 days

Control group: placebo for 7 days

Outcomes

Safety assessment, HCV RNA

Notes

We emailed Marcellin and colleagues on 27 April 2016 for additional information but reply not received yet.

Ongoing study

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described

Selective reporting (reporting bias)

Unclear risk

Safety assessment was not properly described (NCT01356160)

Vested‐interest bias

Unclear risk

Not described

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Marcellin 2013b

Methods

Randomised clinical trial

Participants

351 participants

Countries: France, Germany, Poland, and USA

Inclusion criteria: treatment‐naive non‐cirrhotic genotype 1 infected HCV participants

Exclusion criteria: not described

Interventions

Experimental group: GS‐9451 (200 mg) once daily (those who achieved an extended very rapid virological response (defined as HCV RNA < LLOQ at Weeks 2 and 4 that remained undetectable through week 8) were randomised to stop treatment at either Week 12 or Week 24)

Control group: no intervention

Co‐intervention: GS‐5885 (30mg once a day) + peg (180 mg/week) + RBV (1000 mg–1200 mg/day)

Outcomes

Adverse events, SVR

Notes

We emailed Marcellin and colleagues on 27 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described

Selective reporting (reporting bias)

Unclear risk

Safety assessment was not properly described (NCT01356160)

Vested‐interest bias

Unclear risk

Not described

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

MATTERHORN 2015a1

Methods

Randomised, open‐label, parallel‐group trial (ClinicalTrials.gov: NCT01331850)

Participants

381 participants, randomised: 152 prior partial responders (Cohort A) and 229 prior null responders (cohort B)

Sex: 111 men, 40 women (Cohort A)

Mean age: 49.4 years

Countries: Australia, Austria, Brazil, Canada, France, Germany, Italy, Mexico, Poland, Spain, UK, and USA.

Inclusion criteria: non‐cirrhotic adults with HCV genotype 1a or 1b infection, a baseline HCV RNA level P50,000 IU/mL and evidence of prior peg‐IFN α‐2a/RBV treatment failure. The prior course of treatment must have been discontinued > 12 weeks prior to enrolment, must have comprised at least 12 weeks of therapy with approved doses of peg‐IFNα/RBV and participants must have taken a minimum of approximately 80% of the prescribed doses. Prior treatment failure must have been due to either a partial response (> log10 reduction in HCV RNA at week 12, without achieving an undetectable HCV RNA level by the end of treatment), or a null response (< 2 log10 reduction in HCV RNA at week 12). Absence of cirrhosis must have been documented within 24 months of receiving the first dose of study drug either by liver biopsy (Knodell, METAVIR, Batts & Ludwig fibrosis score 63, or Ishak score 64) or, alternatively, by transient elastography (< 14.5 kPa). Participants with a previous liver biopsy were required to have a platelet count > 90 /nL and those with a transient elastography result were required to have a platelet count of 140–400 /nL

Exclusion criteria: participants were excluded if they were co‐infected with HBV or HIV, had liver disease attributed to a cause other than HCV infection, had previously received a DAA agent or had a serious concomitant chronic illness.

Interventions

Participants were grouped according to their prior treatment response (A: partial responders; B: null responders) and were randomised (1:1:1) within each cohort to 1 of 3 treatment arms, stratified by HCV genotype 1 subtype and host IL28B genotype. Participants who received at least 1 dose of study medication: 151 prior partial responders (Cohort A) and 228 prior null responders (cohort B).

Experimental group A1: oral mericitabine 1000 mg twice a day for 24 weeks.

Control group A2: peg‐IFNα‐2a 180 µg once weekly for 24 weeks.

Experimental group A3: oral mericitabine 1000 mg twice a day for 24 weeks + peg‐IFN α‐2a 180 µg once weekly for 24 weeks.

24 weeks of peg‐IFNα‐2a/RBV.

Co‐intervention: oral danoprevir/r 100/100 mg twice daily (twice a day) for 24 weeks + oral RBV 1000 mg (body weight < 75 kg) or 1200 mg (P75 kg) daily for 24 weeks (group A1,A2,A3,)

Outcomes

Proportion of participants with sustained virological response (SVR24), with SAE, AEs, mortality.

Notes

Due to the parallel design only group A1 and group A3 had an adequate control group (A2), Group B1, B2 and B3 were excluded from the analysis

This analysis A1 vs. control.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomization was centralised and the computer‐generated randomisation list was maintained

Allocation concealment (selection bias)

Low risk

Randomisation list was maintained by Perceptive Informatics (Waltham, MA, USA). "Study sites were informed of participant treatment assignments by an interactive voice/web response system."

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Unblinded study

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Unblinded study

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 2 participants had incomplete data.

Selective reporting (reporting bias)

High risk

The authors did not report on "Change in danoprevir plasma concentration" as was prespecified in their protocol

Vested‐interest bias

High risk

This study was funded by F Hoffmann‐La Roche Ltd

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

MATTERHORN 2015a2

Methods

For characteristics see MATTERHORN 2015a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was centralised and the computer‐generated randomisation list was maintained

Allocation concealment (selection bias)

Unclear risk

Randomisation list was maintained by Perceptive Informatics (Waltham, MA,
USA). Study sites were informed of participant treatment assignments by an interactive voice/web response system

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Unblinded study

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Unblinded study

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Only 2 participants had incomplete data.

Selective reporting (reporting bias)

High risk

The authors did not report on "Change in danoprevir plasma concentration" as was prespecified in their protocol

Vested‐interest bias

High risk

This study was funded by F Hoffmann‐La Roche Ltd

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

McHutchison 2009

Methods

A phase IIb, randomised, double‐blind, multicenter, parallel‐group trial (PROVE‐1)(NCT00336479)

Participants

250 participants

Sex: 157 men, 93 women

Country: USA

Inclusion criteria: age between 18 and 65 years. Chronic hepatitis C infection. HCV genotype 1. Treatment‐naive. Seronegative for hepatitis B surface. antigen and antibodies against HIV‐1 and HIV‐2. Absolute neutrophil count ≥ 1500 cells/mm3. Platelet count ≥ 90,000 cells/mm3. Normal haemoglobin level

Exclusion criteria: decompensated liver disease. Another cause of clinically significant liver disease. HCC. Histologic evidence of cirrhosis (on liver biopsy, which was required within 2 years before the study).

Group 1: 79 participants (T12PR24)

Median age: 49 years (range 21‐61)

Sex: 54 men, 25 women

Race: 60 white (76%), 7 black (9%), 1 Asian (1%), 9 Hispanic (11%), and 2 other (3%)

HCV genotype, n(%): 1a: 53(67), 1b: 17(22), intermediate: 9(11)

HCV RNA ≥ 800,000 IU/mL, n(%): 66(84)

Fibrosis, n(%): none or minimal: 24(30), portal: 41(52), bridging: 14(18)

Group 2: 79 participants (T12PR48)

Median age: 50 years (range 26‐61)

Sex: 48 men, 31 women

Race: 60 white (76%), 8 black (10%), 3 Asian (4%), 7 Hispanic (9%), and 1 other (1%)

HCV genotype, n(%): 1a: 48(61), 1b: 27(34), intermediate: 4(5)

HCV RNA ≥ 800,000 IU/mL, n(%): 68(86)

Fibrosis, n(%): none or minimal: 34(43), portal: 31(39), bridging: 14(18)

Group 3: 17 participants (T12PR12)

Median age: 49 years (range 34‐63)

Sex: 12 men, 5 women

Race: 13 white (76%), 3 black (18%), 0 Asian, 1 Hispanic (6%), and 0 other

HCV genotype, n(%): 1a: 9(53), 1b: 6(35), intermediate: 2(12)

HCV RNA ≥800,000 IU/mL, n(%): 15(88)

Fibrosis, n(%): none or minimal: 4(24), portal: 9(53), bridging: 4(24)

Group 4: 75 participants (PR48)

Median age: 49 years (range 24‐59)

Sex: 43 men, 32 women

Race: 59 white (79%), 9 black (12%), 0 Asian, 6 Hispanic (8%), and 1 other (1%)

HCV genotype, n(%): 1a: 50(67), 1b: 20(27), intermediate: 5(7)

HCV RNA ≥ 800,000 IU/mL, n(%): 69(92)

Fibrosis, n(%): none or minimal: 19(25), portal: 37(49), bridging: 19(25).

Interventions

Experimental group:

1, 2, and 3: oral telaprevir given as a single initial dose of 1250 mg, followed by 750 mg every 8 h for 12 weeks (T12).

Control group:

4: Placebo for 12 weeks.

Co‐interventions:

1: peg‐IFN α‐2a 180 μg subcutaneously once weekly plus oral weight‐based RBV 1000 mg‐1200 mg daily in 2 divided doses for 24 weeks (PR24).

2 and 4: peg‐IFN α‐2a 180 μg subcutaneously once weekly plus oral weight‐based RBV 1000 mg‐1200 mg daily in 2 divided doses for 48 weeks (PR48).

3: peg‐IFN α‐2a 180 μg subcutaneously once weekly plus oral weight‐based RBV 1000 mg‐1200 mg daily in 2 divided doses for 12 weeks (PR12).

Outcomes

Primary outcome: proportion of participants with undetectable HCV RNA at 24 weeks after completion of study drug dosing (SVR24).

Secondary outcomes: proportion of participants with SVR at 12 weeks after completion of study drug dosing. Number of participants with AEs and SAE. Number of participants with viral relapse. Maximum, minimum, and average plasma concentration of telaprevir.

Notes

We emailed McHutchinson and colleagues on 27 April 2016 for additional information on random sequence generation, allocation concealment and SAE but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The method of random sequence generation was not described

Allocation concealment (selection bias)

Unclear risk

Not enough information was provided

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

A telaprevir‐matched placebo given in the same manner was used

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"Data management and interim analyses were performed by the Duke Clinical Research Institute. An independent data‐monitoring committee reviewed the results of all interim analyses"

Incomplete outcome data (attrition bias)
All outcomes

High risk

The number of participants who discontinued treatment was clearly stated, but reasons were not mentioned. Up to 36% of participants in a group discontinued study treatment

Selective reporting (reporting bias)

Low risk

The protocol was available and all pre‐specified outcomes were reported on

Vested‐interest bias

High risk

The sponsor (Vertex Pharmaceuticals) was directly involved in trial design and protocol development

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

McHutchison 2010

Methods

A phase II, randomised, partially placebo‐controlled, partially double‐blind, parallel‐group trial (PROVE‐3)(NCT00420784)

Participants

453 participants

Sex: 306 men, 147 women

Mean age: 51 years

Inclusion criteria: age between 18 and 70 years, chronic hepatitis C infection, HCV genotype 1, previously treated, but without achieving SVR. Seronegative for hepatitis B surface antigen and antibodies against HIV‐1 and HIV‐2, absolute neutrophil count ≥ 1500 cells/mm3, platelet count ≥ 100,000 cell/mm3, normal bilirubin values.

Exclusion criteria: decompensated liver disease, HCC, other clinically significant liver disease.

Country: Canada, Germany, the Netherlands, Puerto Rico and USA.

Group 1: 115 participants (T12PR24)

Sex: 78 men, 37 women

Median age: 51 years (range 22‐65)

Race, n(%): white: 103(90), black: 9(8), Asian: 2(2), other: 1(1)

HCV genotype, n(%): 1a: 69(60), 1b: 33(29), unknown: 13(11)

HCV RNA ≥ 800,000 IU/mL, n(%): 106(92)

Stage of fibrosis or cirrhosis, n(%): none or minimal: 26(23), portal fibrosis: 44(38), bridging fibrosis: 26(23), cirrhosis. 19(17)

Group 2: 113 participants (T24PR48)

Sex: 80 men, 33 women

Median age: 52 years (range 31‐66)

Race, n(%): white: 99(88), black: 11(10), Asian: 0, other: 3(3)

HCV genotype, n(%): 1a: 61(54), 1b: 42(37), unknown: 10(9)

HCV RNA ≥ 800,000 IU/mL, n(%): 104(92)

Stage of fibrosis or cirrhosis, n(%): None or minimal: 20(18), portal fibrosis: 40(35), bridging fibrosis: 33(29), cirrhosis. 20(18)

Group 3: 111 participants (T24PR24)

Sex: 72 men, 39 women

Median age: 53 years (range 19‐69)

Race, n(%): white: 100(90), black: 10(9), Asian: 1(1), other: 0.

HCV genotype, n(%): 1a: 64(58), 1b: 36(32), unknown: 11(10)

HCV RNA ≥ 800,000 IU/mL, n(%): 104(94)

Stage of fibrosis or cirrhosis, n(%): none or minimal: 17(15), portal fibrosis: 40(36), bridging fibrosis: 32(29), cirrhosis. 22(20)

Group 4: 114 participants (PR48)

Sex: 76 men, 38 women

Median age: 50 years (range 18‐65)

Race, n(%): white: 100(88), black: 10(9), Asian: 2(2), other: 2(2)

HCV genotype, n(%): 1a: 71(62), 1b: 34(30), unknown: 9(8)

HCV RNA ≥ 800,000 IU/mL, n(%): 104(91)

Stage of fibrosis or cirrhosis, n(%): none or minimal: 33(29), portal fibrosis: 37(32), bridging fibrosis: 31(27), cirrhosis 13(11).

Interventions

Experimental group:

1: oral telaprevir given in a single initial dose of 1125 mg, followed by 750 mg every 8 h for 12 weeks (T12).

2 and 3: oral telaprevir given in a single initial dose of 1125 mg, followed by 750 mg every 8 h for 24 weeks (T24).

Control group:

1: placebo from Week 13 to Week 24.

4: placebo for 24 weeks.

Co‐intervention:

1 and 3: peg‐IFN α‐2a 180 μg subcutaneously once weekly plus oral weight‐based RBV 1000 mg to 1200 mg daily in 2 divided doses for 24 weeks (PR24).

2 and 4: peg‐IFN α‐2a 180 μg subcutaneously once weekly plus oral weight‐based RBV 1000 mg to 1200 mg daily in 2 divided doses for 48 weeks (PR48).

Outcomes

Primary outcome: SVR defined as undetectable HCV RNA level 24 weeks after the last dose of study drugs.

Secondary outcome measures: proportion of participants with undetectable HCV RNA at completion of study drug dosing. Number of participants with AEs and SAE. Number of participants with viral relapse. Maximum, minimum, and average plasma concentration of telaprevir.

Notes

We emailed McHutchinson and colleagues on 27 April 2016 for additional information on generation of random sequence, allocation concealment, description of blinding but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The method of sequence generation was not specified

Allocation concealment (selection bias)

Unclear risk

Insufficient information was provided on allocation concealment

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The method of blinding was insufficiently described

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"Data management and interim analyses were conducted by the Duke Clinical Research Institute, without revealing the unblinded data"

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Number and reasons for discontinuation of treatment were clearly reported. Most participants discontinued treatment due to meeting pre‐specified stopping rules

Selective reporting (reporting bias)

Low risk

Protocol was available and all pre‐specified outcomes were reported on

Vested‐interest bias

High risk

The sponsor (Vertex Pharmaceuticals) was directly involved in trial design, protocol development, study co‐ordination, drafting and reviewing the manuscript

Other bias

Low risk

The trial appeared to be free of other potential sources of bias

Mostafa 2015

Methods

Randomised clinical trial

Participants

40 participants

Inclusion criteria: previously untreated adults with chronic hepatitis C genotype 4 infection.

Country: Egypt

Interventions

Experimental group: 44 weeks of boceprevir 800 mg 3 times daily.

Control group: no intervention.

Co‐intervention: peg α‐2b 1.5 lg/kg once per week subcutaneously plus weight‐based dosing RBV 15 mg/kg/day (800 mg‐1400 mg/day) for 48 weeks.

Outcomes

Proportion of participants who achieved early response

Notes

We emailed Mostafa and colleagues on 27 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open label study

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Open label study

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described

Selective reporting (reporting bias)

Unclear risk

The trial is not finished according to ClinicalTrials.gov, therefore not all data might have been collected yet

Vested‐interest bias

Low risk

Trial was funded by a non‐profit organisation (Theodor Bilharz Research Institute)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Muir 2014

Methods

Randomised clinical trial

Participants

30 participants

Sex: 18 men, 12 women

Mean age: 51.7 years

Inclusion criteria: adults with chronic hepatitis C, HCV RNA > 10,000 IU/mL at screening, treatment‐naive participants defined as participants who have never received peg‐IFN, RBV, or a DAA agent for the treatment of chronic HCV infection and a liver biopsy within the last 3 years without evidence of cirrhosis.

Exclusion criteria: BMI > 36.0, pregnant or nursing (lactating) women, confirmed by a positive human chorionic gonadotropin laboratory test or women contemplating pregnancy, participation in any interventional clinical trial within 35 days prior to first study medication dose administration on Day 1, known HIV‐1 or HIV‐2 infection/serology and/or positive Hepatitis B surface antigen, use of dietary supplements, grapefruit juice, herbal supplements, cytochrome P2C8 substrates, cytochrome P3A4 inducers and inhibitors, P‐glycoprotein inducers and substrates, organic anion transporting polypeptides inhibitors and substrates, and potent inducers of other cytochrome P enzymes within 14 days prior to dosing through 7 days following completion of study meds. Clinically significant laboratory abnormality at screening (specified in protocol), other forms of liver disease, history of severe or uncontrolled psychiatric disease, history of malignancy of any organ system, treated or untreated within the past 5 years, history of major organ transplantation, use of bone marrow colony‐stimulating factor agents within 3 months prior to baseline, history of seizure disorder requiring ongoing medical therapy, history of known coagulopathy including haemophilia, history of haemoglobinopathy, including sickle cell anemia and thalassaemia, history of immunologically‐mediated disease (specified in protocol), history of clinical evidence of significant chronic cardiac disease (specified in protocol), ECG with any clinically significant abnormality, structural or functional cardiac abnormalities (specified in protocol), history of chronic obstructive pulmonary disease, emphysema, or other chronic lung disease, participants currently abusing amphetamines, cocaine or opiates, or with ongoing alcohol abuse in the judgement of the investigator.

Interventions

Experimental group:

Arm 1: sovaprevir 200 mg once a day + ACH‐3102 150 mg loading dose on Day 1 followed by 50 mg once a day + RBV weight‐based 1000 mg‐1200 mg once a day for 12 weeks.

Arm 2: sovaprevir 400 mg once a day + ACH‐3102 150 mg loading dose on Day 1 followed by 50 mg once a day + RBV weight‐based 1000 mg‐1200 mg once a day for 12 weeks.

Control group: placebo for sovaprevir capsule once a day + placebo for ACH‐3102 150 mg loading dose on Day 1 followed by 50 mg capsule once a day + placebo for weight‐based RBV once a day for 12 weeks.

Outcomes

Safety, SVR4 (only experimental group).

Notes

We contacted the trial authors about random sequence generation, allocation, participants completing the study, blinding, number of deaths, SVR24.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as double‐blinded but there was no description of the placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Described as double‐blinded but there was no description of the placebo

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There were above 5% dropouts (2/30) and it was unclear how the trial dealt with missing data

Selective reporting (reporting bias)

High risk

The original secondary outcomes were later removed (NCT01849562)

Vested‐interest bias

High risk

The trial was sponsored by a company with a given interest in a result (Achillion Pharmaceuticals)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Nelson 2011

Methods

Phase IIb, randomised, dose‐ranging, parallel‐design trial (PROTON)

Participants

121 participants

Country: not stated

Inclusion criteria: chronic hepatitis C, genotype 1, treatment‐naive participants.

Exclusion criteria: cirrhosis.

Interventions

Experimental group:

Group 1: 95 participants: PSI‐7977 200 or 400 mg daily for 12 weeks.

Control group:

Group 2: 26 participants: placebo for 12 weeks.

Co‐intervention in both groups: peg‐IFN α‐2a for 24‐48 weeks in a response‐guided regimen. RBV for 24‐48 weeks in a response‐guided regimen.

Outcomes

Not clearly stated.

Notes

We contacted the trial authors about whole risk of bias assessment, male:female ratio, SVR results and AEs.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Insufficient information

Allocation concealment (selection bias)

Unclear risk

Insufficient information

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Use of placebo suggests blinding, but method not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There was insufficient information to assess whether missing data were likely to induce bias on the results

Selective reporting (reporting bias)

Unclear risk

No protocol available. Not enough information given

Vested‐interest bias

Unclear risk

It was uncertain how the trial was sponsored

Other bias

Low risk

The trial may or may not have been free of other domains that could put it at risk of bias

Nelson 2012a1

Methods

Randomised clinical trial

Participants

516 adult participants

Sex: 311 men, 193 women (analysed only)

Mean age: 46.5 years

Inclusion criteria: participants aged 18‐65 years with HCV genotype l infection who had never received treatment for chronic hepatitis C were eligible for the trial. Chronic hepatitis C was defined as the presence of anti‐HCV antibodies and an HCV RNA titer ≥ 50,000 IU/mL in serum (COBAS® Ampliprep/COBAS® TaqMan® HCV test; detection limit 15 IU/mL, Roche Diagnostics, Indianapolis, USA) with a liver biopsy obtained within the previous 24 months (36 months in participants with cirrhosis or incomplete/transition to cirrhosis) consistent with chronic hepatitis C. HCV genotype 1 infection was confirmed by a molecular assay (Versant HCV Genotyping 2.0 Assay (LiPA), Bayer Diagnostics And Innogenetics, NY, USA). Participants with advanced fibrosis according to a biopsy obtained within the previous 36 months were required to have compensated liver disease (Child–Pugh grade A), a serum α‐ fetoprotein level < 100 ng/mL, and no evidence of HCC on an ultrasound, computerised tomography, or magnetic resonance imaging scan performed within the previous 2 months.

Exclusion criteria: participants were not eligible if they were infected with any HCV genotype other than genotype 1 or had serological evidence of infection with HBV or HIV. Participants were also excluded if they had a BMI < 18 kg/ m2 or ≥ 36 kg/m2, an absolute neutrophil count < 2 × 109 cells/L, a platelet count < 90 × 109 cells/L, a hemoglobin concentration < 120 g/L in women or < 130 g/L in men (or in participants with risk factors for anemia or in whom anemia would be medically problematic), or a serum creatinine level > 1.5 times the ULN. Use of erythropoietin‐stimulating agents or colony‐stimulating factors to elevate haematology parameters to facilitate entry into the study was prohibited. Participants who had previously received any IFN preparation, RBV (or RBV analog), or any investigational HCV protease or polymerase inhibitor were excluded, as were those with a history or evidence of a chronic liver disease other than chronic hepatitis C, a current or past history of chronic disease (including severe psychiatric or pulmonary disease), or a history or evidence of a clinically relevant ophthalmological disorder (e.g. cytomegalovirus infection or macular degeneration). Pregnant or breast‐feeding women and male partners of pregnant women were ineligible for the trial. Female participants of childbearing potential and male participants with partners of childbearing potential were required to use 2 forms of effective contraception during treatment and after the last dose of RBV in accordance with the locally approved label for RBV.

Interventions

Expertimental group:

  1. RO4588161 1000 mg orally twice a day for 24 weeks

  2. RO4588161 500 mg orally twice a day for 24 weeks

  3. RO4588161 500 mg orally twice a day for 24 weeks. Those participants with undetectable HCV RNA in serum (< 15 IU/mL) at week 4 and who remained HCV RNA undetectable through week 22 were to stop all treatment at week 24; those participants who did not meet this criterion were to continue the 3‐drug combination for a further 24 weeks to complete a total treatment duration of 48 weeks.

  4. RO4588161 1500 mg orally twice a day for 24 weeks

  5. RO4588161 1000 mg orally twice a day for 24 weeks

  6. RO4588161 500 mg orally twice a day for 24 weeks

Control group: placebo.

Co‐interventions: Copegus 1000 mg/1200 mg orally daily for 48 weeks. Peg 180 μgsubcutaneously weekly for 24 weeks (groups 1‐3 + control). Copegus 1000 mg/1200 mg orally daily for 48 weeks. Peg 90 μg subcutaneously weekly for 24 weeks (groups 4‐6 + control).

Outcomes

Safety, antiviral activity, SVR12, relapse.

Notes

The planned treatment duration with balapiravir was reduced from 24 to 12 weeks due to safety concerns. We emailed Nelson and colleagues on 06 June 2016 for additional information on incomplete outcome data and SVR but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The computerised randomisation list was generated by the sponsor, maintained in a central repository accessible only to the randomisation list managers, and incorporated in double‐blind labelling of medication containers

Allocation concealment (selection bias)

Low risk

The computerised randomisation list was generated by the sponsor, maintained in a central repository accessible only to the randomisation list managers, and incorporated in double‐blind labelling of medication containers

Blinding of participants and personnel (performance bias)
All outcomes

High risk

"All patiants were unblinded to their treatment assignment and, among those who received balapiravir and who had a CD4+ count < 200 cells/mm3, treatment with peg‐IFN α‐2a (40KD) and RBV was permanently discontinued"

Blinding of outcome assessment (detection bias)
All outcomes

High risk

"All patients were unblinded to their treatment assignment and, among those who received balapiravir and who had a CD4+ count < 200 cells/mm3, treatment with peg‐IFN α‐2a (40KD) and RBV was permanently discontinued"

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There was more than 5% dropouts and it was unclear how the trial accounted for missing data

Selective reporting (reporting bias)

High risk

Relapse rate was not reported on despite being stated as an outcome in the protocol (NCT 00517439).

Vested‐interest bias

High risk

The trial was supported by a company with an interest in a given result (Hoffmann‐La Roche)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Nelson 2012a2

Methods

For characteristics see Nelson 2012a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The computerised randomisation list was generated by the sponsor, maintained in a central repository accessible only to the randomisation list managers, and incorporated in double‐blind labelling of medication containers

Allocation concealment (selection bias)

Low risk

The computerised randomisation list was generated by the sponsor, maintained in a central repository accessible only to the randomisation list managers, and incorporated in double‐blind labelling of medication containers

Blinding of participants and personnel (performance bias)
All outcomes

High risk

All participants were unblinded to their treatment assignment and, among those who received balapiravir and who had a CD4+ count < 200 cells/mm3, treatment with peg‐IFN alfa‐2a (40KD) and RBV was permanently discontinued

Blinding of outcome assessment (detection bias)
All outcomes

High risk

All participants were unblinded to their treatment assignment and, among those who received balapiravir and who had a CD4+ count < 200 cells/mm3, treatment with peg‐IFN alfa‐2a (40KD) and RBV was permanently discontinued

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There was more than 5% dropouts and it was unclear how the trial accounted for missing data

Selective reporting (reporting bias)

High risk

Relapse rate was not reported on despite being stated as an outcome in the protocol (NCT 00517439).

Vested‐interest bias

High risk

The trial was supported by a company with an interest in a given result (Hoffmann‐La Roche)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Nelson 2012a3

Methods

For characteristics see Nelson 2012a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The computerised randomisation list was generated by the sponsor, maintained in a central repository accessible only to the randomisation list managers, and incorporated in double‐blind labelling of medication containers

Allocation concealment (selection bias)

Low risk

The computerised randomisation list was generated by the sponsor, maintained in a central repository accessible only to the randomisation list managers, and incorporated in double‐blind labelling of medication containers

Blinding of participants and personnel (performance bias)
All outcomes

High risk

All participants were unblinded to their treatment assignment and, among those who received balapiravir and who had a CD4+ count < 200 cells/mm3, treatment with peg‐IFN alfa‐2a (40KD) and RBV was permanently discontinued.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

All participants were unblinded to their treatment assignment and, among those who received balapiravir and who had a CD4+ count < 200 cells/mm3, treatment with peg‐IFN alfa‐2a (40KD) and RBV was permanently discontinued

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There was more than 5% dropouts and it was unclear how the trial accounted for missing data

Selective reporting (reporting bias)

High risk

Relapse rate was not reported on despite being stated as an outcome in the protocol (NCT 00517439)

Vested‐interest bias

High risk

The trial was supported by a company with an interest in a given result (Hoffmann‐La Roche)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Nelson 2012a4

Methods

For characteristics see Nelson 2012a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The computerised randomisation list was generated by the sponsor, maintained in a central repository accessible only to the randomisation list managers, and incorporated in double‐blind labelling of medication containers

Allocation concealment (selection bias)

Low risk

The computerised randomisation list was generated by the sponsor, maintained in a central repository accessible only to the randomisation list managers, and incorporated in double‐blind labelling of medication containers

Blinding of participants and personnel (performance bias)
All outcomes

High risk

All participants were unblinded to their treatment assignment and, among those who received balapiravir and who had a CD4+ count < 200 cells/mm3, treatment with peg‐IFN alfa‐2a (40KD) and RBV was permanently discontinued

Blinding of outcome assessment (detection bias)
All outcomes

High risk

All participants were unblinded to their treatment assignment and, among those who received balapiravir and who had a CD4+ count < 200 cells/mm3, treatment with peg‐IFN alfa‐2a (40KD) and RBV was permanently discontinued

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There was more than 5% dropouts and it was unclear how the trial accounted for missing data

Selective reporting (reporting bias)

High risk

Relapse rate was not reported on despite being stated as an outcome in the protocol (NCT 00517439).

Vested‐interest bias

High risk

The trial was supported by a company with an interest in a given result (Hoffmann‐La Roche)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Nelson 2012a5

Methods

For characteristics see Nelson 2012a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The computerised randomisation list was generated by the sponsor, maintained in a central repository accessible only to the randomisation list managers, and incorporated in double‐blind labelling of medication containers

Allocation concealment (selection bias)

Low risk

The computerized randomizsation list was generated by the sponsor, maintained in a central repository accessible only to the randomizsation list managers, and incorporated in double‐blind labelling of medication containers

Blinding of participants and personnel (performance bias)
All outcomes

High risk

All participants were unblinded to their treatment assignment and, among those who received balapiravir and who had a CD4+ count < 200 cells/mm3, treatment with peg‐IFN alfa‐2a (40KD) and RBV was permanently discontinued.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

All participants were unblinded to their treatment assignment and, among those who received balapiravir and who had a CD4+ count < 200 cells/mm3, treatment with peg‐IFN alfa‐2a (40KD) and RBV was permanently discontinued

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There was more than 5% dropouts and it was unclear how the trial accounted for missing data

Selective reporting (reporting bias)

High risk

Relapse rate was not reported on despite being stated as an outcome in the protocol (NCT 00517439)

Vested‐interest bias

High risk

The trial was supported by a company with an interest in a given result (Hoffmann‐La Roche)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Nelson 2012a6

Methods

For characteristics see Nelson 2012a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The computerised randomisation list was generated by the sponsor, maintained in a central repository accessible only to the randomisation list managers, and incorporated in double‐blind labelling of medication containers

Allocation concealment (selection bias)

Low risk

The computerised randomisation list was generated by the sponsor, maintained in a central repository accessible only to the randomisation list managers, and incorporated in double‐blind labelling of medication containers

Blinding of participants and personnel (performance bias)
All outcomes

High risk

All participants were unblinded to their treatment assignment and, among those who received balapiravir and who had a CD4+ count < 200 cells/mm3, treatment with peg‐IFN alfa‐2a (40KD) and RBV was permanently discontinued.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

All participants were unblinded to their treatment assignment and, among those who received balapiravir and who had a CD4+ count < 200 cells/mm3, treatment with peg‐IFN alfa‐2a (40KD) and RBV was permanently discontinued.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There was more than 5% dropouts and it was unclear how the trial accounted for missing data

Selective reporting (reporting bias)

High risk

Relapse rate was not reported on despite being stated as an outcome in the protocol (NCT 00517439).

Vested‐interest bias

High risk

The trial was supported by a company with an interest in a given result (Hoffmann‐La Roche)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Nelson 2012b

Methods

Randomised clinical trial

Participants

323 adult participants

Inclusion criteria: chronic HCV infection for at least 6 months prior to baseline (Day 1), liver biopsy results (performed no more than 2 years prior to screening) indicating the absence of cirrhosis, mono‐infection with HCV genotype 1a or 1b, HCV treatment‐naive, BMI between 18 and 36 kg/m2, creatinine clearance >/= 50 mL/min, participant agreed to use highly effective contraception methods if female of childbearing potential or sexually active male, screening laboratory values within defined thresholds for ALT, AST, leukopenia, neutropenia, anaemia, thrombocytopenia, thyroid stimulating hormone, potassium, magnesium.

Exclusion criteria: autoimmune disease, decompensated liver disease or cirrhosis, poorly controlled diabetes mellitus, severe psychiatric illness, severe chronic obstructive pulmonary disease, serological evidence of co‐infection with HIV, HBV, or another HCV genotype, suspicion of HCC or other malignancy (with exception of certain skin cancers), history of haemoglobinopathy, known retinal disease. participants who were immunosuppressed, participants with known, current use of amphetamines, cocaine, opiates (i.e. morphine, heroin), methadone, or ongoing alcohol abuse, participants who were on or are expected to be on a potent cytochrome P450 (CYP) 3A4 or Pgp inhibitor, or a QT prolonging medication within 2 weeks of baseline (Day 1) or during the study, participants must have had no history of clinically significant cardiac disease, including a family history of Long QT syndrome, and no relevant ECG abnormalities at screening.

Interventions

Experimental group 1: tegobuvir (20 mg twice a day) + GS‐9256 (150 mg twice a day).

Experimental group 2: GS‐9256 (150 mg twice a day).

Control group: placebo.

Co‐intervention: Peg (180 mg/week) + RBV (1000–1400 mg/day).

Outcomes

Safety, SVR12 (not fully reported so could not be used).

Notes

Participants receiving the 4‐drug therapy who achieved an extended vRVR were randomised to stop treatment at either Week 16 or Week 24. We contacted the trial authors on 06 June 2016 for additional information allocation sequence generation, blinding, dropouts and how this was handled, primary publication, SAE, death, SVR24, number of participants randomised to each group.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as double blind but the placebo was not further described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was unclear how many participants dropped out

Selective reporting (reporting bias)

High risk

Not all predefined outcomes in the protocol were reported on (viral resistance, SVR24)

Vested‐interest bias

High risk

The trial was supported by a company that might have an interest in a given result (Gilead Sciences)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Nettles 2010

Methods

Randomised clinical trial

Participants

18 participants

Sex: 10 men, 8 women

Mean age: 44 years

Inclusion criteria: participants chronically infected with hepatitis C virus genotype 1, treatment‐naive or treatment non‐responders or treatment intolerant; and not co‐infected with HIV or HBV, HCV‐RNA viral load of ≥ 10*5* IU/mL and had a BMI 18‐35 kg/m²

Exclusion criteria: any significant acute or chronic medical illness which was not stable or not controlled with medication and not consistent with HCV infection and major surgery within 4 weeks of study drug administration and any gastrointestinal surgery that could impact the absorption of study drug

Interventions

Experimental group:

  1. daclatasvir 1 mg

  2. daclatasvir 10 mg

  3. daclatasvir 100 mg

Control group: placebo

Outcomes

Pharmacokinetics, antiviral activity, safety.

Notes

We contacted trial authors for additional information on allocation sequence generation and concealment, how was blinding maintained, whether HIV participants included.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as double‐blinded but the placebo was not described in detail

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% of participants dropped out and it was unclear how the trial handled missing data

Selective reporting (reporting bias)

Low risk

All the outcomes stated in the protocol were reported on NCT00546715

Vested‐interest bias

Unclear risk

The trial was funded by a company that might have an interest in a given result (Bristol‐Myers Squibb)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Nettles 2011a1

Methods

Randomised clinical trial

Participants

Sex: 25 men, 5 women

Mean age: 44.3 years

Inclusion criteria: eligible participants for this study were men and women, ages 18‐60 years inclusive, with a BMI of 18‐35 kg/m2, who were chronically infected (longer than 6 months) with HCV genotype 1, and who were treatment‐naive to IFN and RBV. Additional inclusion criteria were: plasma HCV RNA 100,000 IU/mL; documented FibroTest score of 0.72 and APRI 2, or the absence of cirrhosis based on liver biopsy within 12 months; women of childbearing potential were not to be nursing or pregnant and had to be willing to agree to use double barrier contraception for at least 1 month before dosing, during dosing, and at least 12 weeks after the last dose of study medication.

Exclusion criteria: participants with prior documented cirrhosis on liver biopsy; previous exposure to a NS5A replication cofactor inhibitor; co‐infection with HIV; co‐infection with HBV.

Interventions

Experimental group:

  1. daclatasvir (1 mg) once a day.

  2. daclatasvir (10 mg) once a day.

  3. daclatasvir (30 mg) once a day.

  4. daclatasvir (60 mg) once a day.

  5. daclatasvir (100 mg) once a day.

  6. daclatasvir (30 mg) twice a day.

Control group: placebo.

Outcomes

Pharmacokinetics, mortality, SAE, antiviral efficacy

Notes

We contacted the trial authors on 06 June 2016 for additional information on blinding of participants, personnel and outcome assessors, SVR24

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Compute‐ generated randomisation scheme

Allocation concealment (selection bias)

Low risk

Interactive voice‐response system

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as double‐blinded but the placebo was not described in detail

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 1 participant did not complete the study until day 28

Selective reporting (reporting bias)

High risk

There were added multiple secondary outcomes to the original protocol after the trial was conducted (NCT00663208)

Vested‐interest bias

High risk

The trial was supported by a company that might have an interest in a given result (Bristol‐Myers Squibb)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Nettles 2011a2

Methods

For characteristics see Nettles 2011a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation scheme

Allocation concealment (selection bias)

Low risk

Interactive voice‐response system

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as double‐blinded but the placebo was not described in detail

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 1 participant did not complete the study until day 28

Selective reporting (reporting bias)

High risk

There were added multiple secondary outcomes to the original protocol after the trial was conducted (NCT00663208)

Vested‐interest bias

High risk

The trial was supported by a company that might have an interest in a given result (Bristol‐Myers Squibb)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Nettles 2011a3

Methods

For characteristics see Nettles 2011a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation scheme

Allocation concealment (selection bias)

Low risk

Interactive voice‐response system

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as double‐blinded but the placebo was not described in detail

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 1 participant did not complete the study until day 28

Selective reporting (reporting bias)

High risk

There were added multiple secondary outcomes to the original protocol after the trial was conducted (NCT00663208)

Vested‐interest bias

High risk

The trial was supported by a company that might have an interest in a given result (Bristol‐Myers Squibb)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Nettles 2011a4

Methods

For characteristics see Nettles 2011a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation scheme

Allocation concealment (selection bias)

Low risk

Interactive voice‐response system

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as double‐blinded but the placebo was not described in detail

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 1 participant did not complete the study until day 28

Selective reporting (reporting bias)

High risk

There were added multiple secondary outcomes to the original protocol after the trial was conducted (NCT00663208)

Vested‐interest bias

High risk

The trial was supported by a company that might have an interest in a given result (Bristol‐Myers Squibb)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Nettles 2011a5

Methods

For characteristics see Nettles 2011a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation scheme

Allocation concealment (selection bias)

Low risk

Interactive voice‐response system

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as double‐blinded but the placebo was not described in detail

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 1 participant did not complete the study until day 28

Selective reporting (reporting bias)

High risk

There were added multiple secondary outcomes to the original protocol after the trial was conducted (NCT00663208)

Vested‐interest bias

High risk

The trial was supported by a company that might have an interest in a given result (Bristol‐Myers Squibb)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Nettles 2011a6

Methods

For characteristics see Nettles 2011a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation scheme

Allocation concealment (selection bias)

Low risk

Interactive voice‐response system

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as double‐blinded but the placebo was not described in detail

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 1 participant did not complete the study until day 28

Selective reporting (reporting bias)

High risk

There were added multiple secondary outcomes to the original protocol after the trial was conducted (NCT00663208)

Vested‐interest bias

High risk

The trial was supported by a company that might have an interest in a given result (Bristol‐Myers Squibb)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Nishiguchi 2014a1

Methods

Randomised clinical trial

Participants

Sex: 13 men, 9 women

Mean age: 53.9 years

Inclusion criteria: treatment‐naive adults aged 20–70 years, with chronic genotype‐1 HCV infection and HCV RNA viral load at screening ≥ 100,000 IU/mL.

Exclusion criteria: cirrhosis.

Interventions

Experimental group:

1: faldaprevir 120 mg once a day (treatment‐naive).

2: faldaprevir 240 mg once a day (treatment‐naive).

Control group: placebo.

Co‐intervention: peg‐IFN α‐2a 180 μg and RBV 600 mg/day (≤ 60 kg), 800 mg/day (> 60 to ≤ 80 kg) or 1000 mg/day (> 80 kg). Both peg‐IFN and RBV were for 44 weeks.

Outcomes

Safety, SVR24.

Notes

We emailed Nishiguchi and colleagues on 24 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The trial used a "pseudo‐random number generator and supplied seed number" to generate the allocation sequence

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The trial was only blinded up to week 8

Blinding of outcome assessment (detection bias)
All outcomes

High risk

The trial was only blinded up to week 8

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There was above 5% dropouts and it was unclear how the trial accounted for missing data

Selective reporting (reporting bias)

High risk

The secondary outcomes were changed after the trial was completed (NCT00947349)

Vested‐interest bias

High risk

The trial was supported by a company that might have an interest in a given result (Boehringer Ingelheim)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Nishiguchi 2014a2

Methods

For characteristics see Nishiguchi 2014a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The trial used a "pseudo‐random number generator and supplied seed number" to generate the allocation sequence

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The trial was only blinded up to week 8

Blinding of outcome assessment (detection bias)
All outcomes

High risk

The trial was only blinded up to week 8

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There was above 5% dropouts and it was unclear how the trial accounted for missing data

Selective reporting (reporting bias)

High risk

The secondary outcomes were changed after the trial was completed (NCT00947349)

Vested‐interest bias

High risk

The trial was supported by a company that might have an interest in a given result (Boehringer Ingelheim)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

OPERA 2011a1

Methods

Phase IIa, randomised, placebo‐controlled study, parallel‐group design (NCT00561353)

Participants

77 participants (Cohort 1 and 2) and 39 participants (Cohort 4)

Countries: 26 centres in Belgium, France, Germany, the Netherlands, Poland, and the UK

Inclusion criteria: eligible participants were aged 18–70 years with documented chronic HCV infection (genotype 1; diagnosis > 6 months prior to screening), a plasma HCV RNA ≥ 10,000 IU/mL (COBAS® TaqMan HCV/HPS assay v2.0 (Roche Molecular Systems, Pleasanton, CA, USA)) and a BMI 18–32 kg/m2. Participants were either treatment‐naive, or were non‐responders or relapsers to prior IFN/RBV or peg‐IFN/RBV therapy who did not discontinue anti‐HCV therapy due to AEs. Participants with compensated cirrhosis (up to Child–Pugh A according to standard criteria) were included. Treatment‐experienced participants were defined as non‐responders or relapsers who had virologically failed prior IFN/RBV or peg‐IFN/RBV therapy. Prior non‐responders were those who had not achieved a 2 log10 IU/ mL decrease in HCV RNA from baseline after 12 weeks of prior IFN‐based therapy. Prior relapsers were those who had detectable HCV RNA during follow‐up after achieving undetectable HCV RNA at the end of previous treatment.

Exclusion criteria: other causes of significant liver disease, decompensated cirrhosis, HCC, prolonged Qtc value, platelet count < 90/nl, neutrophile count < 2/nl, bilirubin > 1.5 x ULN, AST or ALT level > 5 x ULN, excessive use of alcohol, positive urinary drug screening, HIV, Hepatitis B, contraindication for treatment with peg‐IFN or RBV.

Interventions

The trial included multiple treatment cohorts. Cohort 1 and 2 included treatment‐naive participants. Participants in Cohort 4 were treatment‐experienced.

Cohort 1, Panel A: participants were randomised 3:3:2

Experimental group 1A_1: simeprevir 25 mg once daily for 4 weeks.

Experimental group 1A_2: simeprevir 75 mg once daily for 4 weeks.

Control group 1A: placebo.

Co‐intervention 1A: peg‐IFN α‐2a + RBV in week 2‐4.

Cohort 1, Panel B: Participants were randomised 3:3:2

Experimental group 1B_1: simeprevir 25 mg once daily for 4 weeks.

Experimental group 1B_2: simeprevir 75 mg once daily for 4 weeks.

Control group 1B: placebo.

Co‐intervention 1B: peg‐IFN α‐2a + RBV for 4 weeks.

Cohort 2, Panel A: participants were randomised 3:1

Experimental group 2A: simeprevir 200 mg once daily for 4 weeks.

Control group 2A: placebo.

Co‐intervention 2A: peg‐IFN α‐2a + RBV in week 2‐4.

Cohort 2, Panel B: participants were randomised 3:1.

Experimental group 2B: simeprevir 200 mg once daily for 4 weeks.

Control group 2B: placebo.

Co‐intervention 2B: peg‐IFN α‐2a + RBV for 4 weeks.

Cohort 4: participants randomised 1:1:1:1

Experimental group 4_1: simeprevir 75 mg once daily for 4 weeks.

Experimental group 4_1: simeprevir 150 mg once daily for 4 weeks.

Experimental group 4_1: simeprevir 250 mg once daily for 4 weeks.

Control group 4: placebo.

Co‐intervention 4: peg‐IFN α‐2a + RBV for 4 weeks.

Participants in all cohorts 1, 2 and 4 could receive P/R up to week 48 following the initial 28‐day TMC435 treatment period.

Outcomes

AE, SAE, change from baseline in HCV RNA level at day 7, percentage of participants with undetectable HCV RNA at week 4.

Notes

A planned cohort 3 should have investigated simeprevir 400 mg once daily, but was cancelled before participant enrolment.

This is cohort 125 mg vs control. We emailed Manns and colleagues on 26 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Low risk

"Randomisation was achieved using the central interactive web response system, managed by ClinPhone Group Ltd"

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blinded and placebo described as identical

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 5 participants were not included in the intention‐to‐treat analysis resulting in under 5% with missing data

Selective reporting (reporting bias)

Low risk

All outcomes in the protocol were reported on

Vested‐interest bias

High risk

This study was sponsored by Tibotec Pharmaceuticals

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

OPERA 2011a2

Methods

For characteristics see OPERA 2011a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Low risk

Randomisation was achieved using the central interactive web response system, managed by ClinPhone Group Ltd

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blinded and placebo described as identical

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 5 participants were not included in the intention‐to‐treat analysis resulting in under 5% with missing data.

Selective reporting (reporting bias)

Low risk

All outcomes in the protocol were reported on

Vested‐interest bias

High risk

This study was sponsored by Tibotec Pharmaceuticals

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

OPERA 2011a3

Methods

For characteristics see OPERA 2011a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Low risk

Randomisation was achieved using the central interactive web response system, managed by ClinPhone Group Ltd

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blinded and placebo described as identical

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 5 participants were not included in the intention‐to‐treat analysis resulting in under 5% with missing data.

Selective reporting (reporting bias)

Low risk

All outcomes in the protocol were reported on

Vested‐interest bias

High risk

This study was sponsored by Tibotec Pharmaceuticals

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

OPERA 2011a4

Methods

For characteristics see OPERA 2011a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Low risk

Randomisation was achieved using the central interactive web response system, managed by ClinPhone Group Ltd

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blinded and placebo described as identical

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 5 participants were not included in the intention‐to‐treat analysis resulting in under 5% with missing data.

Selective reporting (reporting bias)

Low risk

All outcomes in the protocol were reported on

Vested‐interest bias

High risk

This study was sponsored by Tibotec Pharmaceuticals

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

OPERA 2011a5

Methods

For characteristics see OPERA 2011a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Low risk

Randomisation was achieved using the central interactive web response system, managed by ClinPhone Group Ltd

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blinded and placebo described as identical

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 5 participants were not included in the intention‐to‐treat analysis resulting in under 5% with missing data

Selective reporting (reporting bias)

Low risk

All outcomes in the protocol were reported on

Vested‐interest bias

High risk

This study was sponsored by Tibotec Pharmaceuticals

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

OPERA 2011a6

Methods

For characteristics see OPERA 2011a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Low risk

Randomisation was achieved using the central interactive web response system, managed by ClinPhone Group Ltd

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Described as double‐blinded and placebo described as identical

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 5 participants were not included in the intention‐to‐treat analysis resulting in under 5% with missing data.

Selective reporting (reporting bias)

Low risk

All outcomes in the protocol were reported on

Vested‐interest bias

High risk

This study was sponsored by Tibotec Pharmaceuticals

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Pasquinelli 2012a1

Methods

Randomised clinical trial

Participants

Sex: 18 men, 6 women

Mean age: 48 years

Inclusion criteria: eligible participants with chronic HCV infection were men or women aged 18‐60 years with a BMI of 18‐35 kg/m2 and chronic infection with HCV genotype 1, either treatment‐naive, treatment nonresponders (including relapsers), or treatment intolerant. Additional inclusion criteria were plasma HCV RNA levels of 100,000 IU/mL, a documented FibroTest score of 0.72 or 0.59, and an AST platelet ratio index of 2 or the absence of cirrhosis based on liver biopsy within 12 months

Exclusion criteria: main exclusion criteria included previous exposure to another NS3 protease inhibitor, co‐infection with HIV or HBV, or being women of childbearing potential

Interventions

Experimental group:

  1. 10 mg single dose

  2. 50 mg single dose

  3. 200 mg single dose

  4. 600 mg single dose

Control group: placebo every 12 h

Outcomes

Antiviral activity, safety, pharmacokinetics

Notes

We emailed Pasquinelli and colleagues on 06 June 2016 for additional information on description of the placebo, were outcome assessors blinded, who experienced a SAE, how was missing data handled, SVR24 data.but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation scheme

Allocation concealment (selection bias)

Low risk

An interactive voice‐response system was used to assign a unique participant number

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial is described as double‐blinded but the placebo was not described in detail

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Low risk

There were no dropouts

Selective reporting (reporting bias)

Low risk

The outcomes reported in the protocol are reported (NCT00559247)

Vested‐interest bias

High risk

The trial was funded by a company that might have an interest in a given result (Bristol‐Myers Squibb)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Pasquinelli 2012a2

Methods

For characteristics see Pasquinelli 2012a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation scheme

Allocation concealment (selection bias)

Low risk

An interactive voice‐response system was used to assign a unique participant number

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial is described as double‐blinded but the placebo was not described in detail

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There were above 5% dropouts and it was unclear how the trial handled missing data

Selective reporting (reporting bias)

Low risk

The outcomes reported in the protocol are reported (NCT00722358)

Vested‐interest bias

High risk

The trial was funded by a company that might have an interest in a given result (Bristol‐Myers Squibb)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Pearlman 2014

Methods

Randomised clinical trial

Participants

101 participants were randomised to either triple (n = 49) or to double therapy (n = 52)

Sex: 63 men, 38 women

Mean age: 53 years

Inclusion criteria: treatment‐naive, infected with genotype 1 HCV, and had low viral load at baseline (< 600,000 IU/mL). Participants were 18 years of age or older and had a liver biopsy in the past 2 years consistent with chronic hepatitis. Before randomisation, participants had been rapid virologic responders to 4 weeks of peg‐IFN α‐2b.

Exclusion criteria: cirrhosis participants. HCV/HIV co‐infection; HCV genotype other than 1; biopsy‐proven or strongly suspected clinical cirrhosis; other causes of liver disease, including co‐infection with hepatitis B; creatinine clearance < 50 mL/min (modification of diet in renal disease equation); platelet count < 80 3 109/L; neutrophil count < 1.5 3 109/L; haemoglobin concentration < 13 g/dL and 12 g/dL in men and women, respectively; coexisting uncontrolled psychiatric or cardiopulmonary disorders; haemoglobinopathy; sarcoidosis; malignant neoplasm; receipt of immunosuppressive or immunomodulatory therapy in the previous 6 months; pregnancy; and men whose partners were pregnant or unwilling to use contraception during the study period. Female participants of childbearing age also agreed to avoid systemic contraception if ultimately randomised into the protease inhibitor‐containing arm. Participants were also excluded if they imbibed significant amounts of alcohol (> 30 g/day), or if they were active substance abusers in the past 6 months.

Interventions

Experimental group: 24 weeks of peg/RBV/BOC (boceprevir 800 mg three times a day) (Group A).

Co‐intervention: 20 weeks of peg/RBV only (Group B).

Outcomes

Side effects, viral response.

Notes

We contacted trial authors for additional information on unpublished results, randomisation, blinding of outcome assessment, allocation concealment, SAEs and AEs.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label, no blinding

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

4% in group A and 6% in group B, a total of 10% discontinuations

Selective reporting (reporting bias)

Unclear risk

Protocol not found

Vested‐interest bias

High risk

Dr. Pearlman consults, advises, and is on the speakers’ bureau for Merck

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Pearlman 2015

Methods

Randomised clinical trial

Participants

93 participants

Sex: 53 men, 29 women (analysed)

Mean age: 56.5 (analysed)

Country: USA

Inclusion criteria: chronic HCV infection. Participants 18 years or older were eligible for enrolment if they had genotype 1a infection and a plasma HCV RNA level greater than 10,000 IU/mL. African American ethnicity was self‐identified by participants at screening. All participants either were previously untreated or had shown a prior null response to peg‐IFN/RBV as defined by < a 2‐log10 decrease at 12 weeks of therapy compared with a baseline value and as verified by laboratory records. Other eligibility criteria included documentation of cirrhosis by means of a liver biopsy (METAVIR stage 4) or a FibroTest (Lab Corp, Burlington, NC) score > 0.75 and an AST:platelet ratio index > 2, with a Child‐Turcotte‐Pugh score of <

7 at screening (class A). Participants needed to have had an ultrasound performed within 6 months before screening, or by the time of the baseline visit, with no findings suspicious for HCC, and to have an international normalised ratio of ≤ 2.3, a total bilirubin level of < 3 mg/dL, a platelet count of ≥ 50,000 per mL3, and a serum albumin level > 2.7 g/dL. There were no upper age or BMI limits. Participants with stable, medicated psychiatric disease and methadone maintenance participants also were eligible.

Exclusion criteria: non–genotype 1a, including genotype 1 infection that could not be subtyped; prior treatment with telaprevir or boceprevir; a history of decompensation or history of Child‐Turcotte‐Pugh class B or C; co‐infection with HIV or HBV; a creatinine clearance of < 50 mL/min (modification of diet in renal disease equation); a haemoglobin concentration < 12 g/dL in men and < 11 g/dL in women; co‐existing uncontrolled psychiatric or cardiopulmonary disorders; haemoglobinopathy; sarcoidosis; malignant neoplasm in the past 5 years except localised nonmelanoma skin cancer; receipt of immunosuppressive or immunomodulatory therapy within the previous 6 months; or participants who were either pregnant or planning to be pregnant or were men whose partners were pregnant or unwilling to use contraception during the study period. Participants who had discontinued prior therapy because of an AE were not eligible.

Interventions

Experimental group: oral simeprevir (150 mg) once daily for 12 weeks.

Control group: peg‐IFN α‐2b (1.5 μg/kg/wk) (Merck, Whitehouse Station, NJ), oral RBV (1000 mg–1200 mg/day, based on body weight < 75 kg or ≥ 75 kg, respectively) for 12 weeks.

Co‐intervention: sofosbuvir (400 mg) once daily for 12 weeks.

Outcomes

Efficacy, quality of life, safety assessment, virological response

Notes

The trial reported it was linked to (NCT021683615) however the NCT number could not be identified on ClinicalTrials.gov. Seperate data from African‐American/white was presented

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The trial was open‐label

Blinding of outcome assessment (detection bias)
All outcomes

High risk

The trial was open‐label

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

High risk

The trial was funded by Gilead Sciences

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Petry 2011

Methods

Randomised clinical trial

Participants

84 participants

Sex: 84 men

Inclusion criteria: 18‐65 years old with HCV RNA > 105 IU/L, and genotype‐1 or ‐3 chronic HCV infection without clinical evidence of cirrhosis.

Interventions

Experimental group: doses of 50 mg (genotype‐1) or 100 mg (genotype‐3) to 800 mg MK‐5172) for 7 days.
Control group: placebo.

Outcomes

Plasma HCV RNA, pharmacokinetics.

Notes

NCT00998985

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as being placebo‐blinded, but it was unclear how the blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as being placebo‐blinded, but it was unclear how the blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not described

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

Unclear risk

Not described

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Pockros 2008a1

Methods

Randomised clinical trial

Participants

107 adult participants

Sex: 67 men, 37 women

Mean age: 47.08 years

Inclusion criteria: participants were eligible for inclusion if they were aged 18‐65 years and had chronic HCV genotype 1 infection with HCV RNA levels 50,000 IU/mL. Only treatment‐naive participants were enrolled in the study. Other inclusion criteria included chronic liver disease consistent with chronic HCV infection on biopsy, and compensated liver disease (Child‐Turcotte‐Pugh grade A). Women of childbearing potential were required to have a negative blood pregnancy test within the 24‐h period prior to the first dose of study medication. All fertile participants, male and female, were required to use 2 forms of effective contraception during treatment and for 6 months afterward.

Exclusion criteria: participants were excluded from the study if they had infection with any HCV genotype other than genotype 1, or an indeterminate or mixed genotype; hepatic cirrhosis (Knodell score of 4, Metavir score of 4, or Ishak modified histological activity index score of 5 or 6) or incomplete/ transition to cirrhosis (Knodell score of 3, Metavir score of 3, or an Ishak modified histological activity index score of 4 with nodules or 3 bridges); a low absolute neutrophil count (1500 cells/mm3); a low platelet count (120,000 cells/mm3); or a low haemoglobin concentration (13 g/dL in women or 14 g/dL in men), HIV, Hepatitis A, Hepatitis B infection.

Interventions

Experimental group:

  1. RO5024048 1500 mg orally twice a day for 4 weeks.

  2. RO5024048 3000 mg orally twice a day for 4 weeks.

  3. RO5024048 1500 mg orally twice a day for 4 weeks and Copegus 1000 mg/1200 mg orally daily.

Control group: placebo + Copegus 1000 mg/1200 mg orally daily.

Co‐intervention: Pegasys 180 μg subcutaneously weekly for 4 weeks and 44 weeks of standard of care (peg‐IFN α‐2a (180 μg subcutaneously), RBV (1000 mg orally once a day for those weighing < 75 kg; 1200 mg orally once a day if ≥ 75 kg) for 4 weeks).

Outcomes

Safety, pharmacokinetics, antiviral efficacy.

Notes

We emailed Pockros and colleagues on 06 June 2016 for additional information on allocation sequence generation, allocation concealment, blinding of outcome assessment, how many dropped out but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The participants and care providers were blinded up until week 8. Outcomes were only reported till week 8 and therefore results were blinded.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was unclear how many dropped out and how the trial dealt with missing data

Selective reporting (reporting bias)

Unclear risk

All outcomes stated in the protocol were reported on (NCT00377182)

Vested‐interest bias

High risk

The trial was funded by a company that might have an interest in a given result (Hoffmann‐La Roche)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Pockros 2008a2

Methods

For characteristics see Pockros 2008a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The participants and care providers were blinded up until week 8. Outcomes were only reported till week 8 and therefore results were blinded.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was unclear how many dropped out and how the trial dealt with missing data

Selective reporting (reporting bias)

Unclear risk

All outcomes stated in the protocol were reported on (NCT00377182)

Vested‐interest bias

High risk

The trial was funded by a company that might have an interest in a given result (Hoffmann‐La Roche)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Pockros 2008a3

Methods

For characteristics see Pockros 2008a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The participants and care providers were blinded up until week 8. Outcomes were only reported till week 8 and therefore results were blinded.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was unclear how many dropped out and how the trial dealt with missing data

Selective reporting (reporting bias)

Unclear risk

All outcomes stated in the protocol were reported on (NCT00377182)

Vested‐interest bias

High risk

The trial was funded by a company that might have an interest in a given result (Hoffmann‐La Roche)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Pockros 2009

Methods

Randomised clinical trial

Participants

244 participants

Mean age: 50 years

Inclusion criteria: treatment‐naive or prior non‐responders.

Exclusion criteria: women who were pregnant or breastfeeding, ALT >/ or = 5 x the ULN, AST >/ or = 5 x the ULN.

Interventions

Experimental group:

  1. HCV 796 capsules, 500 mg, every 12 h. daily, 48 weeks (treatment‐naive).

  2. HCV 796 capsules, 500 mg, every 12 h daily, 48 weeks (non‐responders).

Control group: placebo.

Co‐intervention: Peg‐Intron subcutaneous injection, weight‐based dosing, weekly and Rebetol capsules, weight‐based dosing, every 12 h daily for 48 weeks.

Outcomes

Primary outcome complete early virologic response. Secondary outcome rapid virological response.

Notes

We contacted trial authors for addition information on whether HIV participants included, allocation sequence generation and concealment, how was blinding maintained, who was blinded, maximum follow‐up, how many participants dropped out, how was missing data handled, SAE, death, SVR24 but reply not received.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial is described as double‐blinded but the placebo was not described in detail

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

The number of dropouts was not described

Selective reporting (reporting bias)

Unclear risk

The outcome called upon in the protocol was reported (NCT00367887)

Vested‐interest bias

High risk

The trial was funded by a company that might have an interest in a given result (PfizerViroPharma)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Pol 2012

Methods

Randomised clinical trial

Participants

48 participants

Sex: 32 men, 16 women

Mean age: 51.3 years

Countries: USA and France.

Inclusion criteria: chronic HCV genotype 1 infection and were treatment‐naive or had < 4 weeks of exposure to RBV or IFN‐based therapy. Participants needed to have an HCV RNA concentration of ≥ 105 IU/mL and be aged 18‐70 years.

Exclusion criteria: cirrhosis, by liver biopsy within 24 months of baseline, clinically significant comorbidities, and HIV or hepatitis B co‐infection.

Interventions

Experimental group: oral 3 mg, 10 mg, 60 mg once daily for 48 weeks.

Control group: placebo.

Co‐intervention: peg‐IFN α‐2a (180 μg per week) and RBV (1000 mg–1200 mg daily).

Outcomes

HCV RNA, safety assessment, virological response.

Notes

We emailed Pol and colleagues on 27 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random allocation sequence

Allocation concealment (selection bias)

Low risk

Interactive voice‐response system

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The participants and personnel were only blinded until week 12

Blinding of outcome assessment (detection bias)
All outcomes

High risk

The sponsors, who performed the analyses, were only blinded until week 12

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out

Selective reporting (reporting bias)

High risk

The trial changed outcomes from the protocol

Vested‐interest bias

High risk

The trial was funded by Bristol‐Myers Squibb

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Pol 2013

Methods

Randomised clinical trial

Participants

239 participants non‐cirrhotic genotype 1 HCV participants

Sex: unknown

Mean age: unknown

Exclusion criteria: none specified.

Interventions

Experimental group: GS‐9451 (200 mg once a day) alone for 16 or 24 weeks (arm 1) or GS‐9451 (200 mg once a day) and tegobuvir (30 mg twice a day) 24 weeks (arm 2).

Control group: placebo.

Co‐intervention: peg (180 mg/week) + RBV (1000 mg–1200 mg/day) up to 48 weeks based on response to therapy.

Outcomes

Very rapid virological response, rapid virological response, SVR, serious adverse events

Notes

The authors were contacted on 06 June 2016 for additional information on allocation sequence generation, blinding, missing data, SVR24, safety, deaths, full publication

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was unclear how many participants had missing data

Selective reporting (reporting bias)

High risk

SVR24 was not reported but was stated in the protocol (NCT01271790)

Vested‐interest bias

High risk

The trial was sponsored by a company with an interest in a given outcome (Gilead Sciences)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Poordad 2007

Methods

Randomised clinical trial

Participants

117 treatment‐naive participants with chronic hepatitis C

Exclusion criteria: pregnant, breastfeeding, or co‐infected with HBV and/or HIV.

Interventions

Experimental group: valopicitabine 200 mg once a day.

Control group: RBV 1000 mg‐1200 mg daily + valopicitabine placebo once a day.

Co‐intervention: peg‐IFNα‐2a 180 μg weekly.

Outcomes

Pharmacokinetics, antiviral activity, SAE (not reported fully, so we could not use the data).

Notes

We contacted the trial authors on 06 June 2016 for additional information on allocation sequence generation and concealment, maximum follow‐up, how many participants dropped out, how was missing data handled, SAE, Death, SVR24, number randomised in each group.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Only described as single blind

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Only described as single blind

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was unclear how many dropped out

Selective reporting (reporting bias)

Unclear risk

No outcomes were reported in the protocol (NCT00395421)

Vested‐interest bias

High risk

The trial was funded by a company that might have an interest in a given result: Merck Sharp & Dohme Corp.

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Poordad 2011a1

Methods

A phase III, international, randomised, placebo‐controlled, parallel‐group study (SPRINT‐2)(NCT00705432)

Participants

1097 participants

Country: France, Germany, Italy and USA

Inclusion criteria: treatment‐naive participants, age ≥ 18 years, weight of 40‐125 kg, chronic infection with HCV genotype 1, plasma HCV RNA level ≥ 10,000 IU/mL.

Exclusion criteria: liver disease of other cause, decompensated cirrhosis, renal insufficiency, HIV or hepatitis B infection, pregnancy, current breastfeeding, active cancer.

Group 1: 363 participants

Sex: 206 men, 157 women

Mean age ± SD: 49 ± 10 years

Race, n (%): white: 296 (82), black: 52 (14), Asian: 9 (2), other: 6 (2)

Location, n (%): North America: 254 (70), Europe: 99 (27), Latin America: 10 (3)

Weight, mean ± SD (kg): 80 ± 16

HCV subtype, n (%): 1a: 227 (63), 1b: 121 (33), missing data: 15 (4)

HCV RNA level, n (%): > 400,000 IU/mL: 337 (93), > 800,000 IU/mL: 308 (85)

METAVIR fibrosis score, n(%): 0, 1, or 2: 328 (90), 3 or 4: 24 (7), missing data: 11 (3)

Group 2: 368 participants

Sex: 229 men, 139 women

Mean age ± SD: 50 ± 9 years

Race, n (%): white: 304 (83), black: 52 (14), Asian: 4 (1), other: 8 (2)

Location, n (%): North America: 277 (75), Europe: 79 (21), Latin America: 12 (3)

Weight, mean ± SD (kg): 82 ± 17

HCV subtype, n (%): 1a: 234 (64), 1b: 124 (34), missing data: 10 (3)

HCV RNA level, n (%): > 400,000 IU/mL: 336 (91), > 800,000 IU/mL: 314 (85)

METAVIR fibrosis score, n(%): 0, 1, or 2: 319 (87), 3 or 4: 34 (9), missing data: 15 (4)

Group 3: 366 participants

Sex: 221 men, 145 women

Mean ± SD: 49 ± 9 years

Race, n (%): white: 295 (81), black: 55 (15), Asian: 8 (2), other: 8 (2)

Location, n (%): North America: 270 (74), Europe: 86 (23), Latin America: 10 (3)

Weight, mean ± SD (kg) = 82 ± 17

HCV subtype, n (%): 1a: 237 (65), 1b: 117 (32), missing data: 12 (3)

HCV RNA level, n (%): > 400,000 IU/mL: 341 (93), > 800,000 IU/mL: 313 (86)

METAVIR fibrosis score, n (%): 0, 1, or 2: 313 (86), 3 or 4: 42 (11), missing data: 11 (3)

Interventions

Experimental group:

Group 2: oral boceprevir 800 mg thrice‐daily in 4 capsules of 200 mg each (to be taken with food and at an interval of 7‐9 h between doses) beginning at week 5, for a total of 24 weeks; if HCV RNA levels were undetectable from week 8‐24, treatment was considered complete; if HCV RNA levels were detectable between week 8‐24 (not including week 24), boceprevir was continued for additional 20 weeks (total of 44 weeks).

Group 3: oral boceprevir 800 mg thrice‐daily in 4 capsules of 200 mg each (to be taken with food and at an interval of 7‐9 h between doses) beginning at week 5, for a total of 44 weeks.

Control group:

1: a matched placebo thrice‐daily beginning at week 5 for 44 weeks.

Co‐intervention:

All groups: peg‐IFN α‐2b 1.5 μg/kg body weight subcutaneously once weekly and weight‐based oral RBV at a total dose of 600 mg‐1400 mg daily in divided doses for 4 weeks (lead‐in period).

Groups 1 and 3: peg‐IFN α‐2b 1.5 μg/kg body weight subcutaneously once weekly and weight‐based oral RBV at a total dose of 600mg‐1400 mg daily in divided doses for additional 44 weeks (total of 48 weeks).

Group 2: peg‐IFN α‐2b 1.5 μg/kg body weight subcutaneously once weekly and weight‐based oral RBV at a total dose of 600 mg‐1400 mg daily in divided doses for additional 24 weeks (total of 28 weeks), and those with a detectable HCV RNA level between weeks 8‐24 received the same therapy for an additional 20 weeks (total of 48 weeks).

Outcomes

Primary outcomes: achievement of SVR, defined as undetectable plasma HCV RNA at week 24

(if a participant was missing follow‐up week 24 and had undetectable HCV RNA level at week 12, the participant was considered an SVR).

Secondary outcomes: achievement of SVR defined as undetectable HCV RNA at week 24 in non‐black/African American randomised participants who received at least 1 dose of experimental study drug or placebo. The proportion of participants with EVR (e.g. undetectable HCV RNA at weeks 2, 4, 8, or 12) who achieved SVR. The proportion of participants with undetectable HCV RNA at week 12. The proportion of participants with undetectable HCV RNA at 72 weeks after randomisation.

Notes

Co‐intervention in Group 2 was different from Groups 1 and 3.

We emailed Poordad and colleagues on 27 April 2016 for additional information about blinding outcome assessors and number of participants experiencing non‐serious AEs but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random code

Allocation concealment (selection bias)

Low risk

Allocation concealment was done through interactive voice‐response system

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

In the trial's protocol it is described that placebo would be matched to boceprevir and would be given in the same manner

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It was not mentioned if the outcome assessors were blinded, or the extent of blinding was insufficiently described

Incomplete outcome data (attrition bias)
All outcomes

High risk

49/1099 (4.5%) participants discontinued the peg‐IFN/RBV therapy during the lead‐in period. No specific reasons were given. Due to futility at week 24 another 108, 33, and 36 participants in groups 1, 2, and 3, respectively, discontinued treatment. In total 226/1099 (20,5%) of participants discontinued treatment. No other dropouts were stated

Selective reporting (reporting bias)

Low risk

A protocol was published before randomisation began and all outcome results were reported adequately

Vested‐interest bias

High risk

The sponsor (Merck) was directly involved in trial's design, managing, analyses, as well as, writing, decision of submission for publication, reviewing and drafting the manuscript

Other bias

Low risk

The trial appeared to be free of other bias domains that could put it at risk of bias

Poordad 2011a2

Methods

For characteristics see Poordad 2011a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random code

Allocation concealment (selection bias)

Low risk

Allocation concealment was done through interactive voice‐response system

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

In the trial's protocol it is described that placebo would be matched to boceprevir and would be given in the same manner

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It was not mentioned if the outcome assessors were blinded, or the extent of blinding was insufficiently described

Incomplete outcome data (attrition bias)
All outcomes

High risk

49/1099 (4.5%) participants discontinued the peg‐IFN+RBV therapy during the lead‐in period. No specific reasons were given. Due to futility at week 24 another 108, 33, and 36 participants in groups 1, 2, and 3, respectively, discontinued treatment. In total 226/1099(20,5%) of participants discontinued treatment. No other drop‐outs were stated

Selective reporting (reporting bias)

Low risk

A protocol was published before randomisation began and all outcome results were reported adequately

Vested‐interest bias

High risk

The sponsor (Merck) was directly involved in trial's design, managing, analyses, as well as, writing, decision of submission for publication, reviewing and drafting the manuscript

Other bias

Low risk

The trial appeared to be free of other bias domains that could put it at risk of bias

POSITRON 2013

Methods

Blinded placebo‐controlled trial (NCT01542788)

Participants

Randomised: 280 underwent randomisation, and 278 began treatment
Experimental group: 209 randomised, 207 treated
Control group: 71 randomised, 71 treated

Sex: 151 men, 127 women
Mean age: 52 years

Countries: 63 sites in the USA, Canada, Australia, and New Zealand from March 2012‐May 2012.

Inclusion criteria: eligible participants were cirrhotic or non‐cirrhotic adults with HCV genotype 2 or 3 infection, a baseline HCV RNA level > 10,000 IU/mL unwilling or uneligible or intolerant for IFN‐treatment. Participants had chronic hepatitis C infection (documented by positive anti‐HCV antibody test or positive HCV RNA, or positive HCV genotyping test ≥ 6 months prior to the Baseline/Day 1 visit; or documented by liver biopsy performed prior to the Baseline/Day 1 visit with evidence of chronic HCV). Participants had a BMI > = 18 kg/m2, a screening ECG without clinically significant abnormalities, no evidence of HCC, no Chronic liver disease of a non‐HCV aetiology (e.g. hemochromatosis, Wilson’s disease, α1‐antitrypsin deficiency, and cholangitis) and no co‐infection with HBV or HIV. Participants had no history of significant pulmonary or cardiac disease, or porphyria; no current or prior history of clinical hepatic decompensation (e.g. ascites, jaundice, encephalopathy, or variceal haemorrhage).

Interventions

Randomisation was performed centrally in a 3:1 ratio with stratification according to the presence or absence of cirrhosis.

Experimental group: oral sofosbuvir 400 mg once daily + RBV (1000 mg daily in participants with a body weight < 75 kg, and 1200 mg daily in participants with a body weight ≥ 75 kg) for 12 weeks.

Control group: placebo.

Outcomes

Proportion of participants with end‐of‐treatment response (week12),

SVR12, SAE, AEs, mortality.

Notes

We emailed Jacobson and colleagues on 21 April 2016 for additional information on generation of allocation sequence, how many participants dropped out and how the trial handled missing data but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not reported

Allocation concealment (selection bias)

Low risk

"An Interactive Web Response System (IWRS) will be employed to manage participant randomization and study drug assignment."

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The participant, caregiver, investigator, outcomes assessor were described as being blinded and the placebo was identical in appearance

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The participant, caregiver, investigator, outcomes assessor were described as being blinded and the placebo was identical in appearance

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was unclear how many participants dropped out and how the trial handled missing data

Selective reporting (reporting bias)

Low risk

The outcomes stated in the protocol were reported on

Vested‐interest bias

High risk

The sponsor collected the data, monitored study conduct, and performed the statistical analysis

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Reddy 2007

Methods

Randomised clinical trial

Participants

40 adult participants

Inclusion criteria: chronic hepatitis C genotype 1 whose alpha‐IFN treatment had failed.

Exclusion criteria: non‐cirrhotic.

Interventions

Experimental group:

  1. 750 mg once a day R7128

  2. 1500 mg once a day R7128

  3. 750 mg twice a day R7128

  4. 1500 mg twice a day R7128

Control group: placebo

Outcomes

SAE, antiviral activity, safety.

Notes

We contacted the trial authors on allocation sequence generation and concealment, maximum follow‐up, how many participants dropped out, how was missing data handled, death, SVR24, and number randomised in each group.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

There was a placebo but it was unclear how well matched the placebo was and who was blinded to it

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was unclear how many participants had missing data

Selective reporting (reporting bias)

Unclear risk

No prepublished protocol could be found

Vested‐interest bias

Unclear risk

It was unclear how the trial was funded

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Reesink 2006

Methods

Randomised phase I clinical trial

Participants

37 adult participants

Sex: 22 men, 12 women (analysed)

Mean age: 47 years

Countries: Germany, the Netherlands.

Inclusion criteria: men or women between the ages of 18 and 65 years, with BMI between 18.5 and 29.0 kg/m2 (men) or 18.5 and 32.5 (women). Entry criteria included an HCV RNA level 1 105 IU/mL as measured using the Roche COBAS TaqMan HCV assay (Roche Molecular Diagnostics, Pleasanton, CA) (confirmed by repeat measure of 2 separate samples taken during the screening period), HCV genotype 1 (any subtype), and an ALT concentration 4 times the ULN.

Exclusion criteria: decompensated liver disease, cirrhosis, and positive screening for hepatitis B surface antigen or anti‐HIV 1/2.

Interventions

Experimental group: oral 450 mg or 750 mg of VX‐950 3 times daily, or 1250 mg twice daily for 14 days.

Control group: placebo.

Outcomes

Pharmacokinetics, safety assessment, antiviral assessment.

Notes

We emailed Reesink and colleagues on 27 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as double‐blinded with matching placebo, but it was unclear if the participants and investigators were blinded to results (except HCV RNA)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as double‐blinded with matching placebo, but it was unclear if the participants and investigators were blinded to results (except HCV RNA)

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% did not complete the trial (3 participants were not included in the analyses)

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

High risk

The trial was funded by Vertex Pharmaceuticals Incorporated

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Reiser 2005

Methods

Randomised clinical trial

Participants

10 adult participants

Sex: 8 men, 2 women

Mean age: 34.5 years

Inclusion criteria: women or men aged 18 years or older with chronic genotype 2 or 3 HCV infection. The line probe assay was used to determine the genotype of the viral infection. A liver biopsy specimen showing changes consistent with chronic HCV infection had to have been performed within the previous 12 months. At screening, the HCV load had to be 50,000 copies/mL serum.

Exclusion criteria: women were excluded if they were breast‐feeding or at risk of pregnancy; men had to use an adequate form of contraception if their partner was of childbearing potential. They were not enrolled if there were other or additional reasons for chronic liver disease, including the presence of other hepatitis‐causing viruses and/or a history of alcohol abuse within the previous 12 months and/or evidence of Child’s B or C liver disease at screening. No other antiviral or antimicrobial or investigational therapies were allowed during the study (screening, pretreatment, and treatment phases). Participants were excluded if, at screening, their baseline ALT/AST plasma levels exceeded the ULN by more than 5‐fold (5 times the ULN) or their total bilirubin or alkaline phosphatase levels were 1.5 times the ULN. Other exclusion criteria included co‐infection with HIV, a platelet count 100,000/mm3, a white blood cell count 2000 cells/mm3, any clinically significant laboratory abnormalities, and a positive test result for illicit or nonprescription drugs.

Interventions

Experimental group: oral 500 mg of BILN‐2061 for 2 days.

Control group: placebo.

Outcomes

Virological efficacy, pharmacokinetics, safety.

Notes

We emailed Reiser and colleagues on 27 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Low risk

0 participants dropped out

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained for all 3 stages, and the ClinicalTrials.gov information was added after completion

Vested‐interest bias

High risk

The trial was funded by Boehringer Ingelheim

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Rodriguez‐Torres 2008

Methods

Randomised clinical trial

Participants

50 adult participants

Inclusion criteria: chronic hepatitis C genotype 1 who were treatment‐naive.

Exclusion criteria: none reported.

Interventions

Experimental group:

  1. 500 mg twice a day R7128 for 28 days.

  2. 1500 mg twice a day R7128 for 28 days.

Control group: placebo.

Co‐intervention: 180 μg peg‐IFN α‐2a and 1000 mg‐1200 mg RBV.

Outcomes

Antiviral activity (RVR), SAE, AE.

Notes

We emailed Rodriguez‐Torres and colleagues on 06 June 2016 for additional information on allocation sequence generation and concealment, blinding, incomplete outcome data including which groups the 2 participants who were omitted from the analyses were from, how the trial was funded, prepublished protocol, death, SVR but reply not received.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

A placebo was mentioned but it was unclear who was blinded to the intervention and how well matched the placebo was

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was unclear how many participants had missing data

Selective reporting (reporting bias)

Unclear risk

No prepublished protocol could be found

Vested‐interest bias

Unclear risk

It was unclear how the trial was funded

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Rodriguez‐Torres 2010

Methods

Randomised clinical trial

Participants

24 participants (first 3 cohorts)

Inclusion criteria: participants who were 18‐65 years of age, had laboratory evidence of HCV infection for 6 months, defined by 1. presence of anti‐HCV antibody (genotype 1a and 1b infection), or 2. documented HCV RNA presence by a sensitive and specific assay and 3. histologic evidence of CHC (Fibrosis on a standardised histological grading system), plasma HCV RNA of 100,000 IU/mL, were HIV 1 and HIV2 ab seronegative, BMI ≤ 35 kg/m2 BMI and treatment‐naive.

Exclusion criteria: contraindications to peg‐IFN or RBV therapy, have evidence of liver cirrhosis, decompensated liver disease, and Child‐Pugh score > 5, have haemoglobinopathies, unstable cardiac disease, history of organ transplant, active malignant disease or uncontrolled Type I or II diabetes.

Interventions

Experimental group:

  1. 250 mg twice a day for 3 days.

  2. 500 mg twice a day for 3 days.

  3. 750 mg twice a day for 3 days.

  4. 1500 mg once a day for 3 days.

Control group: placebo

Co‐intervention: peg‐IFN α‐2a plus RBV were offered from day 4 for up to 48 weeks.

Outcomes

Pharmacokinetics, antiviral activity, AEs.

Notes

We emailed Rodriguez‐Torres and colleagues on 06 June 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as double‐blind (participant, caregiver, investigator, outcomes assessor) at ClinicalTrials.gov, but it is not clear how well the placebo was matched

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as double‐blind (participant, caregiver, investigator, outcomes assessor) at ClinicalTrials.gov, but it was not clear how well the placebo was matched

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was unclear how many participants had missing data

Selective reporting (reporting bias)

Unclear risk

No prepublished protocol could be found. The outcomes stated at ClinicalTrials.gov were submitted after the start of the trial (NCT00911963)

Vested‐interest bias

High risk

The trial was funded by companies that might have an interest in a given result (Vertex Pharmaceuticals Incorporated and ViroChem Pharma)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Rodriguez‐Torres 2011a1

Methods

Randomised clinical trial

Participants

70 adult participants

Inclusion criteria: chronic hepatitis C genotype 1 who were men and women, 18‐65 years of age inclusive (BMI of at least 18kg/m2 not exceeding 36kg/m2), had a diagnosis of chronic HCV by 1 previous PCR result prior to screening, with a positive HCV viral load of at least 100,000 IU/mL at screening measured by quantitative PCR, HCV genotype 1 per central lab testing report, HCV treatment‐naive (defined as no prior treatment with IFN, peg‐IFN, RBV, or any HCV DAA drugs), liver biopsy consistent with chronic HCV infection but non‐cirrhotic as judged by a pathologist (Knodell < 3, Metavir < 2, Ishak < 4, or Batts & Ludwig < 2) within the last 2 years and before Visit 2 (biopsy can be done within screening period), negative urine drug screen for drugs of abuse at screening and Study Day ‐1 (methadone use allowed), women would have a negative serum βHCG pregnancy test at screening & negative urine dipstick pregnancy test upon entry to clinical unit on Study Day ‐1, agreement by both women of childbearing potential and men(who have not been surgically sterilised) to practice an acceptable method of birth control. Surgical sterilisation of either female or male partner must have occurred at least 6 months prior to first dose and women must be post‐menopausal for 2 years to be considered of non‐child‐bearing potential. Acceptable contraceptive methods include 1 of the following: oral and implantable hormonal contraceptives by woman at least 3 months prior to the 1st dose of Study Drug, IUD in place at least 6 months prior to first dose, barrier methods either diaphragm or condom with spermicide. (Abstinence is not an acceptable method of birth control, participants who indicate sexual inactivity must agree to utilise birth control in the event of sexual activity), willing and able to complete all study visits and procedures, and able to communicate with the investigator and other personnel, signed informed consent form executed prior to protocol screening assessments.

Exclusion criteria: advanced liver disease, cirrhosis, or with signs of decompensated liver disease such as variceal bleeding, ascites, hepatic encephalopathy, active jaundice (total bilirubin > 2, or other evidence of decompensated liver disease, co‐infection with HBV or HIV (positive test for HBsAg or anti‐HIV Ab), acute cardiac ischaemias, unstable heart disease or clinically symptomatic cardiac abnormalities apparent on ECG & PE, or a QTcB interval at Visit 1 of ≥ to 450 ms by Bazette's correction, or personal or family history of Torsades de pointes, use of the following medications concurrently or within the 30 days prior to screening associated with QT prolongation: macrolides, antiarrhythmic agents, azoles, fluoroquinolones, and tricyclic anti‐depressants (methadone use allowed), use of immunosuppressive or immune‐modulating agents (including corticosteroids and immunosuppressive agents) or presence of an immunologically‐mediated autoimmune disease (other than asthma) or history of organ transplantation (inhaled steroids for asthma and topical steroid for minor skin conditions allowed), use of strong CYP3A4‐inhibiting protease inhibitors (specifically atazanavir, indinavir, nelfinavir, saquinavir, and ritonavir), strong CYP3A4 inhibitors (specifically clarithromycin, itraconazole, ketoconazole, nefazodone, telithromycin), or strong CYP3A4 inducers (specifically rifampin, efavirenz, etravirine, phenobarbital, phenytoin, and carbamazepine); absolute NEUT count of < 1800 cells/mm3 (or < 1500 cells/mm3 for African Americans), or platelet count < 130,000 cells/mm3, or haemoglobin < 11g/dL for women and < 13g/dL for men, a history of abnormal thyroid function not adequately controlled (defined as TSH levels < 0.8 x LLN or > 1.2 x the ULN), serum creatinine concentration > 1.5 times the ULN, or albumin < 3g/dL, presence or history of severe, or uncontrolled, or hospitalisation‐requiring psychiatric disease including severe depression, suicide attempts or any severity of psychosis, any malignancy within the last 5 years other than treated cervical carcinoma in situ or treated basal cell carcinoma with no more than 20% risk of recurrence within 2 years, alcohol abuse (investigator assessment) within the past 2 years or an alcohol use pattern that will interfere with the study conduct, drug abuse (investigator assessment) within the last 6 months with exception of methadone, current lactation or breastfeeding, major surgery within 30 days prior Visit 1, participation in another clinical trial of an investigational drug or device within 6 months prior to visit donation of blood or plasma within 30 days prior to Visit 1.

Interventions

Experimental group:

  1. 9 mg INX‐08189 once a day for 7 days.

  2. 25 mg INX‐08189 once a day for 7 days.

  3. 50 mg + 9 mg INX‐08189 once a day for 7 days.

  4. 50 mg + 9 mg INX‐08189 once a day for 7 days.

  5. 9 mg INX‐08189 once a day + RBV for 7 days.

  6. 25 mg INX‐08189 once a day + RBV for 7 days.

  7. 100 mg INX‐08189 once a day.

Control group:

Control for arm 1‐3: placebo.

Control for arm 4‐6: placebo + RBV.

Outcomes

Adverse events, antiviral activity

Notes

We emailed Rodriguez‐Torres and colleagues on 06 June 2016 for additional information on allocation sequence generation and concealment, how blinding was maintained, if outcome assessors were blinded, how many participants dropped out, SAE, death, SVR, male:female, mean age but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as double‐blind but it was unclear how the blinding was maintained

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was unclear how many participants dropped out

Selective reporting (reporting bias)

Low risk

The outcomes stated in the protocol were reported on (NCT01250366)

Vested‐interest bias

High risk

The trial was supported by a company that might have an interest in a given result (Bristol‐Myers Squibb)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Rodriguez‐Torres 2011a2

Methods

Randomised clinical trial

Participants

40 adults with chronic hepatitis C genotype 1 who were treatment‐naive.

Interventions

Experimental group:

  1. 100 mg once a day PSI‐322938.

  2. 200 mg once a day PSI‐322938.

  3. 300 mg once a day PSI‐322938.

  4. 100 mg twice a day PSI‐322938.

Control group: placebo

Outcomes

SAE, AE, HCV RNA, HCV mutations.

Notes

We emailed Rodriguez‐Torres and colleagues on 06 June 2016 for additional information on allocation sequence generation and concealment, blinding, incomplete outcome data, how the trial was funded, prepublished protocol, death, SVR but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

A placebo was mentioned but it was unclear who was blinded to the intervention and how well matched the placebo was

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was unclear how many participants had missing data

Selective reporting (reporting bias)

Unclear risk

No prepublished protocol could be found

Vested‐interest bias

Unclear risk

It was unclear how the trial was funded

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Rodriguez‐Torres 2013

Methods

Randomised clinical trial

Participants

64 participants

Sex: 43 men, 20 women

Mean age: 45.1 years

Inclusion criteria: 64 treatment‐naive participants with chronic HCV genotype 1 infection were enrolled (HCV RNA levels P100,000 IU/mL at screening), 18–65 years of age with a BMI of 18–36 kg/m2. Women of childbearing potential were required to use a protocol‐approved method of contraception. 1 participant in the sofosbuvir 200 mg arm withdrew consent before receiving the first dose of study medication.

Exclusion criteria: a liver biopsy within 3 years of dosing was required to exclude cirrhosis. Participants were otherwise in good health, with no significant co‐morbidities. Other key exclusion criteria included positive test for hepatitis B surface antigen, anti‐hepatitis B core protein IgM antibodies and anti‐HIV antibodies.

Randomization was stratified by interleukin(IL) 28B status (rs12979860) for CC or CT/TT allele.

Interventions

Participants were randomised in a ratio of active:placebo of 1:1:1:1

Experimental group: participants received 1 of 3 once‐daily doses of sofosbuvir (100 mg, 200 mg, or 400 mg).

Control group: placebo plus peg‐IFN α‐2a/RBV for 28 days.

Co‐intervention: peg‐IFN α‐2a and RBV were administered according to the package insert for participants with genotype 1 infection.
After end of treatment, participants continued treatment with peg‐IFN α‐2a/RBV alone for a further 44 weeks.

Outcomes

Primary outcome: AEs.

Secondary outcomes: change in circulating HCV RNA at Week 4, percentage of participants with RVR at Week 4, percentage of participants with SVR at 12 and 24 weeks after last dose of peg+RBV following completion of 48 weeks of treatment, pharmacokinetics, percentage of participants who developed resistance to sofosbuvir.

Notes

We emailed Rodriguez‐Torres and colleagues on 27 April 2016 for additional information on blinding during assessment, unpublished data, (mortality data) but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The randomisation schedule was provided by PharStat, Inc. (NC, USA)

Allocation concealment (selection bias)

Low risk

Participants were randomised by a central web‐based system using permutated blocks

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Both investigators and participants were blinded to the treatment assignment

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The study was described as double‐blinded but it was unclear how the blinding was maintained and who performed the outcome assessment

Incomplete outcome data (attrition bias)
All outcomes

Low risk

2 participants dropped out during study

Selective reporting (reporting bias)

Low risk

A protocol was found (NCT01054729) and all outcomes reported on

Vested‐interest bias

High risk

This study was funded by Gilead Sciences, Inc.

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Rodriguez‐Torres 2014a1

Methods

Randomised clinical trial

Participants

74 participants were randomised

Sex: 49 men, 25 women

Mean age: 54.3 years

Inclusion criteria: participants 18‐65 years of age with hepatitis C genotype 1 infection who had had unsuccessful prior treatment with standard P/R therapy and their screening HCV RNA level was 4 x 105 IU/mL or greater. Participants with cirrhosis by liver biopsy or noninvasive assessment (such as Fibroscan ultrasound and other approved methods according to the local standard of care) were enrolled in a separate cohort. The diagnosis of cirrhosis was based on the interpretation provided by the enrolling investigator.

Exclusion criteria: complicated cirrhosis (defined per protocol as ascites, bleeding oesophageal varices, hepatic encephalopathy, or other signs or symptoms of decompensated cirrhosis), evidence of HCC, HIV co‐infection, or any condition contraindicating re‐treatment with P/R. Participants also were ineligible if recent laboratory tests showed hyperbilirubinaemia (total, > 2.4 mg/dL; or direct, > 1.0 mg/dL), hypoalbuminaemia (< 3.3 g/dL), anemia (< 13 g/dL for men or < 12 g/dL for women), thrombocytopenia (< 100 ‐103/mL), coagulopathy (international normalised ratio, > 1.2), or renal insufficiency (estimated creatinine clearance < 60 mL/min by the Cockcroft–Gault equation).

Interventions

Experimental group:

  1. 600mg vaniprevir twice a day for 24 weeks with P/R for 24 weeks.

  2. 600mg vaniprevir twice a day for 24 weeks with P/R for 48 weeks.

  3. 600mg vaniprevir twice a day for 48 weeks with P/R for 48 weeks.

  4. 300mg vaniprevir twice a day for 48 weeks with P/R for 48 weeks.

Control group: P/R plus placebo for 48 weeks.

Co‐intervention: P/R.

Outcomes

Primary: SVR rate, AEs, discontinuations due to AEs.

Secondary: cEVR, SVR24 for 300 mg vaniprevir, and SVR24 for 600 mg vaniprevir 24 weeks.

Notes

We emailed Rodriguez‐Torres and colleagues on 27 April 2016 for additional information on allocation concealment, randomisation, blinding of participants and personnel as well as outcome assessment, specification of il28b genotypes and the SVR rates for these. Missing data, number of participants analysed for HCV‐related morbidity, sample size calculation, SAEs, but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The study was described as double‐blinded, but it was unclear how the blinding was maintained

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The study was described as double‐blinded but it was unclear how the blinding was maintained and who performed the outcome assessment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

4 participants dropped out (5.4%) due to administrative discontinuations

Selective reporting (reporting bias)

Low risk

A protocol was found (NCT00704405) and all outcomes reported on

Vested‐interest bias

High risk

This study was sponsored and funded by Merck

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Rodriguez‐Torres 2014a2

Methods

For characteristics see Rodriguez‐Torres 2014a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The study was described as double‐blinded, but it was unclear how the blinding was maintained

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The study was described as double‐blinded but it was unclear how the blinding was maintained and who performed the outcome assessment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

4 participants dropped out (5.4%) due to administrative discontinuations

Selective reporting (reporting bias)

Low risk

A protocol was found (NCT00704405) and all outcomes reported on

Vested‐interest bias

High risk

This study was sponsored and funded by Merck

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Rodriguez‐Torres 2014a3

Methods

For characteristics see Rodriguez‐Torres 2014a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The study was described as double‐blinded, but it was unclear how the blinding was maintained

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The study was described as double‐blinded but it was unclear how the blinding was maintained and who performed the outcome assessment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

4 participants dropped out (5.4%) due to administrative discontinuations

Selective reporting (reporting bias)

Low risk

A protocol was found (NCT00704405) and all outcomes reported on

Vested‐interest bias

High risk

This study was sponsored and funded by Merck

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Rodriguez‐Torres 2014a4

Methods

For characteristics see Rodriguez‐Torres 2014a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The study was described as double‐blinded, but it was unclear how the blinding was maintained

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The studywas described as double‐blinded but it was unclear how the blinding was maintained and who performed the outcome assessment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

4 participants dropped out (5.4%) due to administrative discontinuations

Selective reporting (reporting bias)

Low risk

A protocol was found (NCT00704405) and all outcomes reported on

Vested‐interest bias

High risk

This study was sponsored and funded by Merck

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Rodriguez‐Torres 2014b1

Methods

Randomised clinical trial

Participants

288 participants were randomised.

Sex: 153 men, 135 women

Mean age: 47.8 years

Inclusion criteria: treatment‐naive (no prior treatment with IFN ± RBV or investigational anti‐HCV agents). Male and female participants aged ≥ 18 years were eligible for inclusion in the study. All participants were required to be HCV seropositive, infected with a genotype 1 strain, and have plasma HCV RNA levels ≥ 10,000 IU/mL at screening. In addition, a non‐cirrhotic fibrosis classification (i.e. Ishak score ≤ 4 or equivalent) from a liver biopsy obtained within 24 months of screening was required for enrolment.

Exclusion criteria: co‐infected with either HIV or hepatitis B, had evidence of severe or decompensated liver disease or liver disease unrelated to HCV infection, or had any pre‐existing medical condition or laboratory abnormality that made them unsuitable for treatment with peg‐IFN/RBV. Additional exclusion criteria included an abnormal ECG suggestive of clinically significant cardiac disease or QTc > 450 ms at screening, and history of solid organ transplant, or active alcohol or substance abuse sufficient to prevent adherence to study medication and/or follow‐up. Lastly, female participants who were pregnant or nursing and male participants whose female partner was pregnant were excluded.

Interventions

Experimental group:

  1. FLV dosed at 300 mg twice a day in combination with peg‐IFN/RBV for 24 weeks

  2. 600 mg twice a day in combination with peg‐IFN/RBV for 24 weeks.

Control group: placebo in combination with peg‐IFN/RBV for 24 weeks peg‐IFN (Pegasys) was administered at a dose of 180 μg subcutaneously once weekly. RBV (Copegus) was administered at 1000 mg twice a day for participants weighing ≤ 75 kg or 1200 mg twice a day for participants weighing > 75 kg.

Co‐intervention: peg‐IFN/RBV.

Outcomes

Primary: proportion of participants who achieved SVR.

Secondary: the proportion of participants with RVR, complete EVR, end of treatment response (ETR); the proportion of participants with relapsed viraemia; and patterns of AEs and safety measures.

Notes

We emailed Rodriguez‐Torres and colleagues on 27 April 2016 for additional information on randomisation, allocation concealment, blinding of outcome assessment, unpublished data, overview of SAEs and the nature of the SAE but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

All sponsor personnel responsible for the conduct of the trial, with the exception of the sponsor study programmer, remained blinded to the results provided to the data monitoring committee. (Participants and investigators were unblinded to treatment assignment at week 24 to determine eligibility to discontinue therapy)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

67 participants dropped out

Selective reporting (reporting bias)

Low risk

A protocol was found (NCT00987337) and the outcomes reported on

Vested‐interest bias

High risk

This study was sponsored by Pfizer Inc.

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Rodriguez‐Torres 2014b2

Methods

For characteristics see Rodriguez‐Torres 2014b1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

All sponsor personnel responsible for the conduct of the trial, with the exception of the sponsor study programmer, remained blinded to the results provided to the data monitoring committee. (Participants and investigators were unblinded to treatment assignment at week 24 to determine eligibility to discontinue therapy)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

High risk

67 participants dropped out

Selective reporting (reporting bias)

Low risk

A protocol was found (NCT00987337) and the outcomes reported

Vested‐interest bias

High risk

This study was sponsored by Pfizer Inc.

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Rodriguez‐Torres 2015

Methods

Randomised clinical trial

Participants

69 adult participants

Sex: 49 men, 20 women

Mean age: 50 years

Inclusion criteria: chronic genotype 1‐4 HCV infection, for cohorts 1‐9, HCV RNA ≥ 100,000 IU/mL at screening (no HCV RNA restriction for cohort 10), screening laboratory values within defined thresholds and use of 2 effective contraception methods if female of childbearing potential or sexually active male

Exclusion criteria: pregnant or nursing woman or man with pregnant female partner, presence of cirrhosis, prior exposure to approved or experimental HCV protease inhibitors, co‐infection with HIV or HBV, current or prior history of clinical hepatic decompensation, chronic use of systemic immunosuppressive agents, history of clinically significant illness or any other medical disorder that may interfere with participant treatment, assessment or compliance with the protocol.

Interventions

Experimental group:

1: GS‐9857 up to 300 mg (genotype 1a) for 3 days.

2: GS‐9857 up to 300 mg (genotype 3) for 3 days.

3: GS‐9857 up to 300 mg (genotype 2) for 3 days.

4‐9: GS‐9857 up to 600 mg (genotype 1a, 1b, 2, 3, or 4) for 3 days.

10: GS‐9857 100 mg on Day 1 and GS‐9857 100 mg plus SOF/GS‐5816 on Days 2 and 3.

Control group: placebo.

Outcomes

Safety, antiviral activity.

Notes

We contacted the trial authors about allocation sequence generation and concealment, how blinding was maintained, if outcome assessors were blinded, how many participants dropped out, SAE, death, SVR.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described that the control group received placebo but the similarity of the placebo with the study drug was not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was unclear how many participants dropped out

Selective reporting (reporting bias)

Unclear risk

No prepublished protocol could be found (NCT02185794 was published after the start of the trial)

Vested‐interest bias

High risk

The trial was funded by a company that might have an interest in a given result (Gilead Sciences)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Sarrazin 2007

Methods

Randomised clinical trial

Participants

26 adult participants

Inclusion criteria: participants who could be of either sex and any race could be included in this study if they were 18‐60 years of age, were willing to give written informed consent, and were willing to undergo multiple inpatient periods and outpatient visits during the study. Female participants had to be surgically sterile or of non‐childbearing potential, and men had to practice acceptable methods of contraception. Female partners of male enrollees also had to practice acceptable methods of contraception, and all contraception had to have been practiced for 30 days before the dosing period during all dosing periods, and for 30 days after discontinuation of dosing. Participants had to be serum positive for HCV RNA by quantitative polymerase chain reaction assay, with 100,000 IU/mL RNA and be genotype 1a or 1b nonresponders to peg‐IFN‐2b with or without RBV. Nonresponse was defined as achieving < a 2‐log10 decline in HCV RNA levels after at least 12 weeks of dosing with peg‐IFN‐2b at 1.5 g/kg/week. Participants had to have ALT and AST 5 times ULN, ‐fetoprotein values within normal levels, negative screen for drugs with high potential for abuse, normal or clinically acceptable ECG (QTc
value, 450 milliseconds (ms) for women and 430 ms for men), and evidence of compensated liver disease. Participants were required to meet the following criteria: haemoglobin 11 g/dL for women and 12 g/dL for men, white blood cells 4000/mm3, neutrophil count 1500/mm3, and platelets 100,000/mm3 and the following parameters within normal limits: direct bilirubin, indirect bilirubin, albumin, prothrombin time, activated partial thromboplastin time, and serum creatinine.

Exclusion criteria: participants were excluded from the study if they met any of the following criteria: haemophilia or use of anticoagulant therapy; evidence of advanced liver disease (e.g. known cirrhosis, history or presence of ascites, bleeding varices, encephalopathy); presence of organ transplant; known HIV or HBV positivity based on recent tests for anti‐HIV antibodies and hepatitis B surface antigen; or liver disease with a cause other than chronic hepatitis C. The significance of antinuclear antibodies, if present, was to be evaluated by investigators for individual participants to determine whether any interference with the protocol that would warrant exclusion from the study could be expected.

Interventions

Experimental group:

  1. SCH 503034 monotherapy for 1 week of either 200 mg or 400 mg three times a day.

  2. administration of combination SCH 503034 plus peg‐IFN‐2b for 2 weeks. The SCH 503034 could be 200 mg or 400 mg three times a day.

Control group: peg‐IFN‐2b monotherapy administered at 1.5 g/kg once per week.

Outcomes

Antiviral activity, safety, pharmacokinetics.

Notes

We emailed Sarrazin and colleagues on 27 April 2016 for additional information on prepublished protocol, data on SAE, death, SVR24 before the second phase began, allocation concealment but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The trial was described as an open‐label trial

Blinding of outcome assessment (detection bias)
All outcomes

High risk

The trial was described as an open‐label trial

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was unclear how many participants completed the first phase of the trial

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

High risk

The trial was conducted at the Schering‐Plough Research Institute

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Schiff 2008

Methods

Randomised clinical trial

Participants

357 participants

Inclusion criteria: prior null responders with chronic hepatitis C genotype 1, with no evidence of cirrhosis on liver biopsy, results of physical examination and laboratory tests within specified ranges and abstinence from use of abused substances.

Exclusion criteria: women who were pregnant or nursing a child, participants with cirrhosis, co‐infection with Hepatitis B or HIV, and African‐American participants, previous treatment with any HCV polymerase or protease inhibitor, participants who relapsed following response to previous treatment, evidence of advanced liver disease, or liver disease from a cause other than chronic hepatitis C, pre‐existing psychiatric condition.

Interventions

Experimental group:

2: boceprevir 100 mg orally three times a day for 48 weeks.

3: boceprevir 200 mg orally three times a day for 48 weeks.

4: boceprevir 400 mg orally three times a day for 24 weeks

5: boceprevir 400 mg orally three times a day + RBV.

6: boceprevir 400 mg orally three times a day for 48 weeks.

7: boceprevir 800 mg orally three times a day.

8 (added as an amendment): boceprevir 800 mg + RBV.

Control group: (arm 1): placebo + a single dose of peg was given first, followed 1 week later by peg + RBV for 12 weeks. If participant was HCV RNA negative, peg + RBV was continued for another 36 weeks.

Co‐intervention: peg‐IFN alfa‐2b (1.5 mg/kg/wk).

Outcomes

Pharmacokinetics, antiviral activity, safety.

Notes

Control group crossed over at week 17 if with detectable HCV RNA at week 12. Data needed to be available prior to week 12 before we could report the data. We contacted the trial authors on 06 June 2016 for additional information on allocation sequence generation and concealment, maximum follow‐up, how many participants dropped out, how was missing data handled, was there a prepublished protocol other than ClinicalTrials.gov, SAE, death, SVR24, data at week 12, and how much RBV was given but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as double‐blinded but the placebo was not described in detail

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There was above 5% dropouts and it was unclear how the trial handled missing data

Selective reporting (reporting bias)

Unclear risk

Secondary outcomes were first added after the trial was completed

Vested‐interest bias

High risk

The trial was funded by a company that might have an interest in a given result (Merck Sharp and Dohme Corp.)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Silva 2013a1

Methods

Randomised clinical trial

Participants

39 participants were randomised to treatment

Sex: 32 men, 7 women

Mean age: 41.5 years

Inclusion criteria: male and female participants aged 18–60 years, with a BMI between 18 and 29 kg/m2 were enrolled. All participants were serum positive for HCV RNA by quantitative PCR assay, classified as G2/3, and naive to treatment for HCV infection. They were required to have ALT and AST 65 times ULN, no evidence of HCC (per ultrasound and serum alfa‐fetoprotein levels), and haematologic and biochemical evidence of compensated liver disease.

Exclusion criteria: participants with a history of substance abuse within 1 year of study participation, or any clinically significant medical disorder, such as HIV or HBV infection, haemophilia, or evidence of other liver disease not caused by chronic hepatitis C were excluded.

Interventions

Experimental group

  1. boceprevir 200 mg twice a day or placebo.

  2. boceprevir 400 mg twice a day or placebo.

  3. boceprevir 400 mg three times a day or placebo for 14 days.

Control group: placebo.

Co‐intervention: none.

Outcomes

Primary: to evaluate the safety and tolerability of boceprevir.

Secondary: pharmacokinetics and changes in HCV RNA viral load.

Notes

We emailed Silva and colleagues on 27 April 2016 for additional information on allocation concealment, unpublished data, SVR data, (AEs and non serious AEs listed) plus published protocols but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random code provided by the sponsor (Schering‐Plough Research Institute)

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The study was described as double‐blinded, (active drug and matched placebo capsules were used to maintain third‐party blind dispensing)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The study was described as double‐blinded but it was unclear how the blinding was maintained and who performed the outcome assessment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 1 participant dropped out due to AE

Selective reporting (reporting bias)

Unclear risk

Protocol not found

Vested‐interest bias

High risk

This study was supported by Merck & Co. Inc.

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Silva 2013a2

Methods

For characteristics see Silva 2013a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random code provided by the sponsor (Schering‐Plough Research Institute)

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The study was described as double‐blinded, (active drug and matched placebo capsules were used to maintain third‐party blind dispensing)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The study was described as double‐blinded but it was unclear how the blinding was maintained and who performed the outcome assessment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 1 participant dropped out due to AE

Selective reporting (reporting bias)

Unclear risk

Protocol not found

Vested‐interest bias

High risk

This study was supported by Merck & Co., Inc.

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Silva 2013a3

Methods

For characteristics see Silva 2013a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

computer‐generated random code provided by the sponsor (Schering‐Plough Research Institute)

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The study was described as double‐blinded, (Active drug and matched placebo cap‐sules were used to maintain third‐party blind dispensing)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The study was described as double‐blinded but it was unclear how the blinding was maintained and who performed the outcome assessment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 1 participant dropped out due to AE

Selective reporting (reporting bias)

Unclear risk

Protocol not found

Vested‐interest bias

High risk

This study was supported by Merck & Co. Inc.

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Sims 2014

Methods

Randomised clinical trial

Participants

24 participants

Sex: 18 men, 6 women

Mean age: 45.8 years

Country: USA

Inclusion criteria: men and women aged 18‐60 years with chronic HCV genotype 1 infection, a screening plasma HCV RNA level of at least 100,000 IU/mL, and a BMI between 18 and 35 kg/m2. Participants were noncirrhotic (screening FibroTest score of 0.59 with an aminotransferase/platelet ratio index of 2 or with absence of cirrhosis documented by biopsy within the previous 12 months) and could be either treatment‐naive or have previously received and discontinued alfa IFN, with or without RBV, at least 6 months before enrolment.

Exclusion criteria: previous exposure to HCV NS5A or NS5B inhibitors, co‐infected with HIV or HBV or infected with other HCV genotypes. Pregnant or nursing women were also excluded, as were women of childbearing age unwilling to use contraception from 1 month predose through 8 weeks postdose. Men were excluded if unwilling to practice barrier contraception with female partners for at least 12 weeks postdose.

Interventions

The trial was divided into 4 different cohorts comprising

Experimental group: oral 100 mg, 300 mg, 600 mg, and 900 mg of BMS‐791325 for 5 days.

Control group: placebo.

Outcomes

Safety assessment, HCV RNA assessment, pharmacokinetics

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated scheme

Allocation concealment (selection bias)

Low risk

Interactive voice‐response telephone system

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded, but it was unclear how the blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded, but it was unclear how the blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All participants completed the study

Selective reporting (reporting bias)

High risk

The trial added extra primary outcomes in ClinicalTrials.gov (NCT00664625)

Vested‐interest bias

High risk

The trial was funded by Bristol‐Myers Squibb

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

STARTVerso‐1 2015a1

Methods

Randomised multicenter phase III clinical trial

Participants

656 participants

Sex: 342 men, 314 women

Mean age: 47.6 years

Countries: 10 European countries and Japan

Inclusion criteria: treatment‐naïve, aged 18–70 years (Europe), or 20–70 years (Japan), with chronic HCV genotype 1 infection diagnosed by positive anti‐HCV antibodies and HCV RNA > 1000 IU/mL at screening plus a positive antibody or HCV RNA test more than 6 months before screening, or a liver biopsy consistent with chronic HCV infection. Participants with compensated liver disease, including cirrhosis, were eligible for inclusion. All participants had a liver biopsy within 3 years or had a FibroScan within 6 months of randomisation to determine fibrosis stage. For participants without a liver biopsy, fibrosis stage was determined by FibroScan results using a cut‐off value of 9.5 kPa to indicate fibrosis stage > F3 (< 9.5 kPa F0–F2; > 9.5 kPa F3–F4), consistent with evaluations of the use of FibroScan in chronic HCV however, there are no reliable cut‐offs in the literature for distinguishing < F3 from > F3. The FibroScan threshold for cirrhosis was > 13 kPa.

Exclusion criteria: HCV infection of mixed genotype (1/2, 1/3, and 1/4) diagnosed by genotypic testing at screening, evidence of acute or chronic liver disease due to causes other than chronic HCV infection, HIV co‐infection, HBV infection based on presence of HBs‐Ag, active malignancy, or history of malignancy within the last 5 years prior to screening (with an exception of appropriately treated basal cell carcinoma of the skin or in situ carcinoma of the uterine cervix), active or, history of alcohol or illicit drug abuse other than cannabis within the past 12 months, a condition that is defined as one which in the opinion of investigator may put the patient at risk because of participation in this study, may influence the results of this study, or limit the patient's ability to participate in this study, usage of any investigational drugs within 28 days prior to screening, or planned usage of an investigational drug during the course of this study, received concomitant systemic antiviral, hematopoietic growth factor, or immunomodulatory treatment within 28 days prior to screening. Participants being treated with oral antivirals such as acyclovir, famciclovir or valacyclovir for recurrent herpes simplex infection; or with oseltamivir or zanamivir for influenza A infection, may be screened, received silymarin (milk thistle), glycyrrhizin, or Sho‐saiko‐to (SST) within 28 days prior to screening and throughout the treatment phase, known hypersensitivity to any ingredient of the study drugs, alpha fetoprotein value > 100 ng/mL at screening; if > 20 ng/mL and = 100 ng/mL, participants may be included if there is no evidence of liver cancer in an appropriate imaging study (e.g. ultrasound, CT scan, or MRI) within last 6 months prior to randomisation (Visit 2), decompensated liver disease, or history of decompensated liver disease, as defined by the presence of: hepatic encephalopathy, ascites, or oesophageal variceal bleeding and/or laboratory results of any of the following: international normalized ratio = 1.7; serum albumin = 3.5 g/dL; serum total bilirubin = 2.0 mg/dL (except when the increase is predominately due to unconjugated bilirubin and related to Gilbert's syndrome), pre‐existing psychiatric condition that could interfere with the participant's participation in and completion of the study including but not limited to prior suicidal attempt, schizophrenia, major depression syndrome, severe anxiety, severe personality disorder, a period of disability or impairment due to a psychiatric disease within the past 5 years.

Interventions

Experimental group 1: faldaprevir 120 mg once daily. Those with early treatment success (ETS, HCV RNA < 25 IU/mL target detected() or target not detected() at week 4 and < 25 IU/mL TND at week 8) stopped faldaprevir at week 12 and received placebo plus peg‐IFN and RBV for a further 12 weeks. Participants without ETS received faldaprevir plus peg‐IFN and RBV for 24 weeks.

Experimental group 2: faldaprevir 240 mg once daily plus peg‐IFN and RBV for 12 weeks followed by placebo plus peg‐IFN and RBV to week 24, and either stopped treatment (early treatment success) or continued peg‐IFN and RBV to week 48 (no early treatment success).

Control group: placebo.

Co‐intervention: all participants received peg‐IFNα‐2a administered subcutaneously at 180 lg once weekly. RBV administered orally at a total dose of 1000 or 1200 mg (for bodyweight < 75 kg or P75 kg, respectively) daily in 2 divided doses, except in Japan where the total dose was 600, 800, or 1200 mg (for bodyweight 660 kg, > 60–680 kg, or > 80 kg, respectively) daily in 2 divided doses according to the local label peg‐IFN and RBV for 24 weeks after intervention period. All study medication was stopped in the event of virologic breakthrough at or after week 4 (increase in HCV RNA > 1 log10 from nadir or > 25 IU/mL after an initial decrease to < 25 IU/mL), lack of EVR (decrease in HCV RNA P2 log10 from baseline at week 12), or lack of virologic response (detectable HCV RNA at week 24).

Outcomes

Safety assessment, SVR, AST or ALT normalisation, early treatment success.

Notes

We contacted the trial authors for additional information on sequence generation, blinding, who was blinded for the HCV RNA results, missing data.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Low risk

The trial used interactive voice‐response system

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Investigators, sponsor, and participants were blinded to treatment group allocation through the use of matching placebo capsules

Blinding of outcome assessment (detection bias)
All outcomes

High risk

HCV RNA results were only blinded up to week 8

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out (and 34% dropped out of the placebo‐group) and it was unclear if the trial used proper methodology to account for this

Selective reporting (reporting bias)

High risk

The trial changed the primary outcomes from the original version

Vested‐interest bias

High risk

The trial was funded by Boehringer Ingelheim Pharmaceuticals, GmbH & Co. KG

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

STARTVerso‐1 2015a2

Methods

For characteristics see STARTVerso‐1 2015a2

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Low risk

The trial used interactive voice‐response system

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Investigators, sponsor, and participants were blinded to treatment group allocation through the use of matching placebo capsules

Blinding of outcome assessment (detection bias)
All outcomes

High risk

HCV RNA results were only blinded up to week 8

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out (and 34% dropped out of the placebo‐group) and it was unclear if the trial used proper methodology to account for this

Selective reporting (reporting bias)

High risk

The trial changed the primary outcomes from the original version

Vested‐interest bias

High risk

The trial was funded by Boehringer Ingelheim Pharmaceuticals, GmbH & Co. KG.

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

STARTverso‐2 2014a1

Methods

Randomised multicenter phase III clinical trial (STARTverso‐2)

Participants

658 participants

Sex: 389 men, 268 women

Mean age: 50.3

Inclusion criteria: treatment‐naive, 18–70 years (Europe), or 20–70 years (Japan), with chronic HCV genotype 1 infection diagnosed by positive anti‐HCV antibodies and HCV RNA > 1000 IU/ml at screening plus a positive antibody or HCV RNA test more than 6 months before screening, or a liver biopsy consistent with chronic HCV infection. Patients with compensated liver disease, including cirrhosis, were eligible for inclusion. All participants had a liver biopsy within 3 years or had a FibroScan within 6 months of randomisation to determine fibrosis stage. For participants without a liver biopsy, fibrosis stage was determined by FibroScan results using a cut‐off value of 9.5 kPa to indicate fibrosis stage > F3 (< 9.5 kPa F0–F2; > 9.5 kPa F3–F4), consistent with evaluations of the use of FibroScan in chronic HCV however, there are no reliable cut‐offs in the literature for distinguishing < F3 from > F3. The FibroScan threshold for cirrhosis was > 13 kPa.

Exclusion criteria: mixed genotype HCV; HIV or hepatitis B co‐infection; decompensated liver disease; and contraindications to peg‐IFN or RBV. Asian participants were limited to 20% of the total population.

Interventions

Experimental group 1: faldaprevir (BI 201335) 120 mg once daily (oral), for 24 weeks, with pegylated IFN α‐2a (peg‐IFN/RBV), subcutaneous injection/oral. At week 24, if the participants did not achieve early treatment success they received an additional 24 weeks of peg‐IFN/RBV alone.

Experimental group 2: faldaprevir 240 mg once daily. faldaprevir 240 mg once daily (oral), for 12 weeks, with peg‐IFN/RBV (subcutaneous injection/oral). Followed by an additional 12 weeks of placebo plus peg‐IFN/RBV. At week 24, if the participants did not achieve early treatment success they received an additional 24 weeks of peg‐IFN/RBV alone.

Control group: placebo (oral) once daily combined with peg‐IFN/RBV (subcutaneous injection) for 24 weeks, followed by an additional 24 weeks of peg‐IFN/RBV (oral) alone.

Outcomes

Safety assessment, SVR, AST or ALT normalisation, early treatment success.

Notes

Email was sent to Asselah and colleagues on 20 April 2016 for additional information on primary publication, randomisation, blinding, all bias, death but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as double‐blinded but the placebo was not further described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out and the trial did not report how they dealt with missing data

Selective reporting (reporting bias)

High risk

The trial changed the primary outcomes from the original version

Vested‐interest bias

High risk

The trial was funded by Boehringer Ingelheim

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

STARTverso‐2 2014a2

Methods

For characteristics see STARTverso‐2 2014a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as double‐blinded but the placebo was not further described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out and the trial did not report how they dealt with missing data

Selective reporting (reporting bias)

High risk

The trial changed the primary outcomes from the original version

Vested‐interest bias

High risk

The trial was funded by Boehringer Ingelheim

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

STARTverso‐3 2013a1

Methods

Randomised clinical trial

Participants

678 participants

Sex: 403 men, 274 women

Mean age: 53.4 years

Inclusion criteria: chronic hepatitis C genotype 1 infection, diagnosed at least 6 months prior to screening, confirmed prior virological failure with an approved dose of peg‐IFN/RBV age 18‐70 years, HCV RNA = 1000 IU/mL at screening.

Exclusion criteria: HCV infection of mixed genotype; HBV or HIV co‐infection. Evidence of acute or chronic liver disease due to causes other than chronic HCV infection, decompensated liver disease, or history of decompensated liver disease. Body weight < 40 or > 125 kg, clinical evidence of significant or unstable cardiovascular disease, chronic pulmonary disease, history or evidence of retinopathy or clinically significant ophthalmological disorder. Pre‐existing psychiatric condition that could interfere with the participant's participation in and completion of the study, laboratory parameters disorders (thalassaemia major, sickle cell anaemia or G6PD deficit). Haemoglobin < 12 g/dL for women and < 13 g/dL for men, participants who had been previously treated with at least 1 dose of any antiviral or immunomodulatory drug other than IFN alfa or RBV for acute or chronic HCV infection including and not restricted to protease or polymerase inhibitors.

Interventions

The trial was divided into 3 cohorts according to virological failure (relapse, partial, null response) and randomised to 1 of the following groups:

Experimental group 1: participants received faldaprevir 240 mg once daily, in the form of 2 soft gelatin capsules administered orally, combined with peg‐IFN/RBV, administered by injection, for 12 weeks, followed by placebo once daily combined with peg‐IFN/RBV for 12 weeks

Experimental group 2: participants received faldaprevir 240mg once daily, in the form of 2 soft gelatin capsules administered orally, combined with peg‐IFN/RBV, administered by injection, for 24 weeks.

Control group: received 2 soft gelatin capsules identical to those containing faldaprevir once daily (orally) and peg‐IFN α‐2a/RBV) administered by injection, for 24 weeks.

Co‐intervention: At week 24, if the participants did not achieve early treatment success the participants received an additional 24 weeks of peg‐IFN/RBV alone.

Outcomes

SVR, early treatment success, AST, ALT normalisation, safety.

Notes

We emailed Jacobson and colleagues on 26 April 2016 for additional information on primary publication, randomisation, blinding, all bias, death but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as being blinded but method was not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as being blinded but method was not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out and the trial did not report how they dealt with missing data

Selective reporting (reporting bias)

High risk

The trial changed the primary outcomes from the original version (NCT01358864 )

Vested‐interest bias

High risk

The trial was funded by Boehringer Ingelheim

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

STARTverso‐3 2013a2

Methods

For characteristics see STARTverso‐3 2013a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as being blinded but method was not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as being blinded but method was not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out and the trial did not report how they dealt with missing data

Selective reporting (reporting bias)

High risk

The trial changed the primary outcomes from the original version (NCT01358864 )

Vested‐interest bias

High risk

The trial was funded by Boehringer Ingelheim

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

STARTverso‐3 2013a3

Methods

For characteristics see ADVANCE 2011a2

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as being blinded but method was not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as being blinded but method was not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out and the trial did not report how they dealt with missing data

Selective reporting (reporting bias)

High risk

The trial changed the primary outcomes from the original version (NCT01358864 )

Vested‐interest bias

High risk

The trial was funded by Boehringer Ingelheim

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

STARTverso‐4 2015

Methods

Randomised clinical trial

Participants

308 participants

Sex: 248 men, 60 women

Mean age: 46.9 years

Inclusion criteria: 18–70 years, had chronic HCV genotype 1 infection (positive anti‐HCV antibody and HCV RNA > 1000 IU/mL at screening, and documented positive anti‐HCV antibody or HCV RNA > 1000 IU/ mL > 6 months prior to screening), and chronic HIV infection (HIV‐1 viral load testing or HIV‐1 western blot at screening and documented for > 6 months prior to screening) with a Karnofsky score greater than 70. HCV treatment‐naive individuals and those with prior relapse after completion of an IFN‐based regimen (detectable HCV/RNA < 24weeks after treatment with undetectable HCV/RNA at end of treatment) were eligible. Individuals naive to highly active antiretroviral therapy (HAART) were required to have a CD4þ cell count at least 500 cells/mL and HIV plasma RNA below 100,000 copies/mL at screening; those stabilised on HAART (HIV‐1 plasma RNA < 40 copies/mL at screening and < 50 copies/mL for > 6 months before randomisation) were required to have been on an acceptable combination of antiretrovirals (as defined in the protocol, Supplemental Table S1, http://links.lww.com/QAD/A638) for at least 6 weeks prior to randomisation and to have a CD4þ cell count at least 200 cells/mL. Individuals prescribed an atazanavir/ritonavir‐containing HAART regimen were required to have total bilirubin 2.5 times or less the ULN at screening. Documentation of a liver biopsy < 3 years or liver elastography < 6 months of randomisation was mandatory.

Exclusion criteria: mixed genotype HCV, evidence of non‐HCV‐related liver disease, hepatitis B infection, decompensated liver disease, and hypersensitivity to the study treatments.

Interventions

Experimental group: faldaprevir 240 mg for additional 12 weeks

Control group: no intervention

Co‐intervention: peg‐IFN and RBV + faldaprevir 240 mg for the first 12 weeks

Outcomes

ALT, AST, SVR, SAE, mortality.

Notes

Only the group with faldaprevir 240 mg 12W and faldaprevir 240 mg 24W could be used for analyses.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Interactive voice‐response system

Allocation concealment (selection bias)

Low risk

Interactive voice‐response system

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Open‐label study

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Open‐label study

Incomplete outcome data (attrition bias)
All outcomes

High risk

12 participants from the experimental group dropped out, while none from the control group dropped out

Selective reporting (reporting bias)

Low risk

A protocol were published and the trial reported all outcomes (NCT01399619)

Vested‐interest bias

High risk

The trial was funded by Boehringer Ingelheim Pharma GmbH & Co. KG.

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Sulkowski 2013a

Methods

Randomised clinical trial

Participants

62 participants

Sex: 53 men, 7 women (60 analysed)

Mean age: 44.5 years (60 analysed)

Countries: France, Germany, Spain and USA.

Inclusion criteria: treatment‐naive participants age of 18‐65 years, genotype 1 chronic HCV infection, chronic HIV‐1 infection, no previous HCV treatment, and haemoglobin levels of 120 g/L or greater in women and 130 g/L or greater in men. Participants were required to have stable HIV disease defined as follows: part A (no antiretroviral therapy) participants had CD4 counts of ≥ 0.500 x 10^ cells/L and HIV RNA levels of ≤ 100,000 copies/mL, and part B (antiretroviral therapy for > 12 weeks) participants had CD4 counts of ≥ 0.300 x 10^ cells/L and HIV RNA levels < 50 copies/ mL. For part B, permissible antiretroviral regimens were efavirenz, tenofovir, and emtricitabine, or ritonavir‐boosted atazanavir, tenofovir, and either emtricitabine or lamivudine.

Exclusion criteria: hepatic decompensation; other causes of significant liver disease, cancer within 5 years, significant cardiac dysrhythmia, and active AIDS‐related conditions within 6 months. All participants had liver biopsies within 1 year unless previous biopsies indicated cirrhosis; histologic assessment according to the METAVIR scoring system was done by a local pathologist.

Interventions

Experimental group: oral 750 mg of telaprevir 3 times daily for 12 weeks (when the antiretroviral therapy included efavirenz, telaprevir dosage was 1125 3 times daily for 8 weeks).

Control group: placebo.

Co‐intervention: peg‐IFN 2a (180 μg/wk) and RBV (800 mg/d) for a total of 48 weeks.

Outcomes

Safety assessment, efficacy assessment, SVR, pharmacokinetics.

Notes

NCT00983853 participants were randomised in cohorts according to HIV‐treatment. We emailed Sulkowski and colleagues on 27 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Low risk

The trial used interactive web‐response system

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The trial was only blinded for the first 24 weeks

Blinding of outcome assessment (detection bias)
All outcomes

High risk

The trial was only blinded for the first 24 weeks

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out

Selective reporting (reporting bias)

High risk

The trial changed the primary outcome. Safety assessments were originally a primary outcome, this was changed

Vested‐interest bias

High risk

The trial was funded by Vertex pharmaceuticals

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Sulkowski 2013b

Methods

Randomised clinical trial

Participants

99 participants

Sex: 68 men, 31 women

Mean age: 44 years

Countries: Argentina, Belgium, Canada, France and USA.

Inclusion criteria: aged 18–65 years who were infected with both HIV and HCV at 30 academic and non‐academic study sites. Eligible participants had to have untreated, chronic HCV genotype 1 infection without hepatic decompensation, plasma HCV RNA of more than 10,000 IU/mL at screening, no infection with other HCV genotypes, and a liver biopsy sample with histological findings consistent for chronic hepatitis C (and no other cause), participants with a history of HIV infection for > 6 months and stable HIV disease, with a CD4 cell count of ≥ 200 cells per μL and HIV‐1 RNA viral load of < 50 copies per mL.

Exclusion criteria: HBV surface antigen positive; use of didanosine, zidovudine, efavirenz, or other non‐nucleoside reverse transcriptase inhibitors; a neutrophil count of < 1500 cells per μL; a haemoglobin concentration of < 110 g/L for women and < 120 g/L for men; or a platelet count of < 100,000 platelets per μL.

Interventions

Experimental group: 800 mg of boceprevir (MK‐3034) twice a day for 44 weeks.

Control group: placebo.

Co‐intervention: peg‐IFN–RBV for 4 weeks prior to intervention period. Additional 44 weeks of Peg‐IFN alfa‐2b 1.5 μg/kg administered once weekly by subcutaneous injection. RBV 600 mg–1400 mg per day (weight‐based) was taken orally twice daily with food. Erythropoietin was permitted if haemoglobin concentrations decreased to < 100 g/L.

Outcomes

Pharmacokinetics, safety assessment, laboratory values.

Notes

After 12 weeks of treatment the control group was allowed to cross‐over to the experimental group, therefore no data could be used. (NCT01482767)

We emailed Sulkowski and colleagues on 27 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated sequence

Allocation concealment (selection bias)

Low risk

Interactive voice‐response system

Blinding of participants and personnel (performance bias)
All outcomes

High risk

All study site personnel (including the investigators), the sponsor, and participants were masked to treatment assignment until final database lock. But it was unclear when final database lock was defined. Additionally control group were allowed to crossover.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

All study site personnel (including the investigators), the sponsor, and participants were masked to treatment assignment until final database lock. But it was unclear when final database lock was defined. Additionally control group were allowed to crossover

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol were assessed

Vested‐interest bias

High risk

The trial was funded by Merck

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Sulkowski 2013c

Methods

A phase IIb, multicenter, randomised, double‐blind, placebo‐controlled, parallel‐group trial (SILEN‐C1)(NCT00774397)

Participants

429 participants

Sex: 234 men, 195 women

Mean age ± SD: 46 ± 10.5 years

Country: Argentina, Australia, Austria, Canada, Czech Republic, France, Germany, Republic of Korea, the Netherlands, Portugal, Romania, Spain, Switzerland, UK, and USA.

Inclusion criteria: age between 18 and 65 years, chronic hepatitis C infection genotype 1, treatment‐naive, HCV RNA > 100,000 IU/mL. A liver biopsy within 24 months before enrolment providing histologic evidence of any degree of chronic necroinflammatory activity or the presence of fibrosis, but no evidence of cirrhosis, a normal retinal finding on fundoscopy within 6 months before enrolment.

Exclusion criteria: HCV of mixed genotype, HBV or HIV co‐infection, decompensated liver disease, hyperbilirubinaemia > 1.5 ULN, concomitant treatment with medications that are substrates of P‐gp, UGT1A1, CYP3A4 or 2C9.

Group 1: 71 participants

Sex: 41 men, 30 women

Mean age ± SD: 46 ± 10.9 years

Ethnicity, n(%): Asian: 8(11), black: 4(6), white: 57(80), other: 2(3)

HCV genotype, n(%): 1: 1(1), 1a: 32(45), 1b: 38(54). 3a, 4a, 6e, 6q: 0

IL28B genotype, n(%): CC: 11(15), non‐CC: 29(41), missing: 31(44)

Group 2: 69 participants

Sex: 40 men, 29 women

Mean age ± SD: 46 ± 10.9 years

Ethnicity, n(%): Asian: 9(13), black: 1(1), white: 58(84), other: 1(1)

HCV genotype, n(%): 1: 0, 1a: 19(28), 1b: 50(72). 3a, 4a, 6e, 6q: 0

IL28B genotype, n(%): CC: 8(12), non‐CC: 33(48), missing: 28(41)

Group 3: 143 participants

Sex: 74 men, 69 women

Mean age ± SD: 45 ± 10.2 years

Ethnicity, n(%): Asian: 21(15), black: 1(1), white: 119(83), other: 2(1)

HCV genotype, n(%): 1: 0, 1a: 67(47), 1b: 74(52). 3a, 4a, 6e, 6q: 2(1)

IL28B genotype, n(%): CC: 19(13), non‐CC: 53(37), missing: 71(50)

Group 4: 146 participants

Sex: 79 men, 67 women

Mean age ± SD: 46 ± 10.5 years

Ethnicity, n(%): Asian: 17(12), black: 4(3), white: 122(84), other: 3(2)

HCV genotype, n(%): 1: 0, 1a: 51(35), 1b: 91(62). 3a, 4a, 6e, 6q: 4(3)

IL28B genotype, n(%): CC: 22(15), non‐CC: 48(33), missing: 76(52).

Interventions

Experimental group:

2: faldaprevir 120 mg once daily for 24 weeks,

3: faldaprevir 240 mg once daily for 24 weeks,

4: faldaprevir 240 mg once daily for 24 weeks.

Control group:

1: placebo once daily for 24 weeks.

Co‐interventions:

2 and 3: peg‐IFN alfa‐2a 180 μg once weekly and oral weight‐based RBV 1000 mg‐1200 mg daily in 2 divided doses for 48 weeks with a 3‐day lead in period given with placebo.

1 and 4: peg‐IFN alfa‐2a 180 μg once weekly and oral weight‐based RBV 1000 mg to 1200 mg daily in 2 divided doses for 48 weeks.

Outcomes

Primary outcome: sustained virological response 24 weeks after end of treatment

Secondary outcomes: number of participants with virological rebound (HCV RNA < 1 log10 from nadir, or ≥ 100 IU/mL after previous viral load below the lower limit of detection in 2 consecutive visits at least 2 weeks apart. Number of participants with breakthrough (HCV RNA rebound during treatment). Number of participants with relapse (HCV RNA undetectable at end of treatment, but detectable during the follow‐up period). Number of participants with no response (participants who did not achieve SVR, but did not experience a virological breakthrough or relapse).

Notes

We emailed Sulkowski and colleagues on 27 April 2016 for additional information on random sequence generation, allocation concealment, description of blinding, blinding of outcome assessors but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The method of sequence generation was not specified

Allocation concealment (selection bias)

Unclear risk

Insufficient information provided

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Authors stated that participants and investigators were blinded to treatment groups until 24 weeks after the end of treatment, but the method of blinding was not sufficiently described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It was not mentioned if outcomes assessors were blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Number and reasons for treatment discontinuation and withdrawal were clearly stated. From 23%‐40% in the 3 groups of participants discontinued treatment, mostly due to lack of efficacy

Selective reporting (reporting bias)

Low risk

A protocol was published before randomisation began and all outcome results were reported adequately

Vested‐interest bias

Unclear risk

The study was sponsored by Boehringer Ingelheim

Other bias

Low risk

The trial appeared to be free of other bias domains that could put it at risk of bias

Sullivan 2012

Methods

Randomised clinical trial

Participants

37 adult participants

Sex: 22 men, 15 women

Mean age: 48.3 years

Inclusion criteria: chronic hepatitis C genotype 1, who were treatment‐naive participants, where women had to be either postmenopausal for at least 2 years or surgically sterile and men had to be surgically sterile or practicing specific forms of birth control and had documented FibroTest score in combination with an AST to Platelet Ratio Index, or a liver biopsy within the last 12 months to document absence of cirrhosis.

Exclusion criteria: pregnant or breastfeeding woman, use of any medications contraindicated for use with peg‐IFN or RBV 2 weeks prior to study drug administration or 10 half‐lives, whichever was longer, clinically significant cardiac, respiratory (except mild asthma), renal, gastrointestinal, haematologic, neurologic disease, or any uncontrolled medical illness or psychiatric disease or disorder, current or past clinical evidence of cirrhosis or bridging fibrosis, abnormal screening laboratory results.

Interventions

Experimental group:

  1. 5 mg once a day.

  2. 50 mg once a day.

  3. 2000 mg once a day.

Control group: placebo

Co‐intervention: peg‐IFN α‐2a 180 μg/week + weight‐based RBV 1000 mg‐1200 mg/day for 48 weeks.

Outcomes

Notes

We emailed Sullivan and colleagues on 27 April 2016 for additional information on allocation sequence generation and concealment, description of placebo, and prepublished protocol but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as double‐blinded (participant, caregiver, investigator, outcomes assessor) but it was unclear how well matched the placebo was

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as double‐blinded (participant, caregiver, investigator, outcomes assessor) but it was unclear how well matched the placebo was

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was unclear how the trial handled missing data (many were lost to follow‐up but still 'included' in the analyses)

Selective reporting (reporting bias)

High risk

The primary and secondary outcomes were changed after the trial was completed (NCT01314261)

Vested‐interest bias

Unclear risk

The trial was funded by a company that might have an interest in a given result (AbbVie)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Tanwandee 2012

Methods

Randomised clinical phase II trial

Participants

24 adults with chronic hepatitis C, genotype 1, who were naive to antiviral treatment.

Country: Thailand

Exclusion criteria: not described.

Interventions

Experimental group: oral 200 mg, 400 mg of BIT225 for 28 days.

Control group: placebo.

Co‐intervention: IFN alfa 2b and RBV for a total of 48 weeks.

Outcomes

SVR, safety, pharmacokinetics.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as being placebo‐blinded, but it was unclear how the blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as being placebo‐blinded, but it was unclear how the blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No data

Selective reporting (reporting bias)

Unclear risk

No protocol could be obtained

Vested‐interest bias

High risk

The trial was funded by Bristol‐Myers Squibb

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Tatum 2015a1

Methods

Randomised phase II clinical trial

Participants

39 participants

Country: USA

Inclusion criteria: treatment‐naive adults chronically infected with HCV genotype 1 adult participants. Participants were required to have HCV RNA ≥ 10–5 IU/mL (COBAS TaqMan HCV Test 2.0; Roche Molecular Diagnostics, Pleasanton, California; lower limit of quantitation (LLOQ) 25 IU/mL) at screening, with no evidence of cirrhosis by liver biopsy within 24 months of randomisation.

Exclusion criteria: > 4 weeks of prior treatment with IFN or RBV within 6 months prior to randomisation;ALT > 5 x ULN; total bilirubin > 34 μmol/L (> 2 mg/dL) or direct bilirubin > ULN; international normalisation ratio > 1.7; confirmed creatinine clearance < 50 mL/min; or concurrent diagnosis of chronic hepatitis B infection, HIV infection, HCC or other non‐HCV liver disease.

Interventions

Experimental group: oral 75 mg or 150 mg of beclabuvir twice daily for 48 weeks.

Control group: placebo.

Co‐intervention: once‐weekly subcutaneous peg‐IFN (180 lg) and twice‐daily oral RBV (weight‐based dosing of 1000 mg/day (< 75 kg) or 1200 mg/day (> 75 kg)).

Outcomes

HCV RNA, safety assessment, pharmacokinetics.

Notes

We emailed Tatum and colleagues on 27 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol were assessed

Vested‐interest bias

High risk

Bristol‐Myers Squibb

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Tatum 2015a2

Methods

For characteristics see Tatum 2015a1

Participants

Interventions

Outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was described as being double‐blinded but it was unclear how the blinding was performed

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol were assessed

Vested‐interest bias

High risk

Bristol‐Myers Squibb

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Vierling 2011

Methods

Randomised clinical trial

Participants

111 participants

Sex: 64 men, 47 women

Mean age: 46 years

Inclusion criteria: adults with chronic hepatitis C genotype 1 with no previous treatment for chronic hepatitis C, 18‐55 years of age, weight between 40 kg and 125 kg, liver biopsy within 2 years of screening with histology consistent with chronic hepatitis C and no evidence of bridging fibrosis or cirrhosis, participant and participant's partner(s) must each agree to use acceptable methods of contraception for at least 2 weeks prior to Day 1 and continue until at least 6 months after last dose of study drugs and participants must be willing to give written informed consent.

Exclusion criteria: prior treatment for hepatitis C other than herbal remedies, HIV‐positive or known to be co‐infected with hepatitis B, medically significant gallbladder or hepatobiliary findings on screening ultrasound, use of any known significant inducers or substrates of CYP3A4 2 weeks prior to start of study medications, use of herbal supplements (milk thistle permitted), diabetic and hypertensive participants with clinically significant ocular examination findings, current moderate or severe depression, history of depression associated with any of the following: hospitalisation for depression, electroconvulsive therapy for depression, depression that resulted in a prolonged absence from work and/or significant disruption of daily functions, suicidal or homicidal ideation and/or attempt, history of severe psychiatric disorders, past history or current use of lithium, clinical diagnosis of substance abuse of alcohol, intravenous drugs, inhalational (not including marijuana), psychotropics, narcotics, cocaine use, prescription or over‐the‐counter drugs within 5 years of Day 1, past or current use of opiate agonist substitution therapy, any known pre‐existing medical condition (CNS, cardiac, pulmonary, immune mediated) that could interfere with the participant's participation in and completion of the study, active clinical gout within the last year, haemoglobinopathy or coagulopathy, myelodysplastic syndromes, organ transplants other than cornea and hair, poor venous access that precluded routine peripheral blood sampling or an indwelling venous catheter, participants with a history of gastric surgery (e.g. stapling, banding, bypass) or participants with a history of malabsorption disorders (e.g. celiac sprue disease), evidence of active or suspected malignancy, or a history of malignancy, within the last 5 years (except adequately treated basal cell carcinoma of the skin). Participants under evaluation for malignancy were not eligible, participants who were pregnant or nursing, participants who intended to become pregnant during the study period and male participants with partners who were, or intended to become, pregnant during the study period.

Interventions

Experimental group:

  1. narlaprevir 100 mg twice a day and ritonavir 100 mg.

  2. narlaprevir 200 mg once a day and ritonavir 100 mg.

  3. narlaprevir 400 mg once a day and ritonavir 100 mg.

  4. narlaprevir 200 mg once a day and ritonavir 100 mg. There was a 4‐week run in with peg‐IFN and RBV.

  5. narlaprevir 400 mg once a day and ritonavir 100 mg. There was a 4‐week run in with peg‐IFN and RBV.

Control group: no intervention.

Co‐intervention: peg‐IFN α‐2b (1.5 μg/kg subcutaneously, weekly) and RBV (600 mg‐1400 mg/d based on weight) for 48 weeks.

Outcomes

Antiviral effects, pharmacokinetics, safety.

Notes

Participants from the control group were allowed to cross over to the experimental group after 12 weeks of treatment. We could therefore only use results from the first 12 weeks. We contacted trial authors about allocation sequence generation and concealment, how was missing data accounted for, SAE, number randomised to each group.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Described as open‐label

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Described as open‐label

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Above 5% dropouts in the control group and it was unclear how the trial handled missing data

Selective reporting (reporting bias)

Low risk

All outcomes stated in the protocol were reported on (NCT00797745)

Vested‐interest bias

High risk

The trial was funded by a company that might have an interest in a given result (Merck Sharp & Dohme Corp.)

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Villano 2007

Methods

Randomised clinical trial

Participants

Adults with chronic hepatitis C who were naive to treatment

Interventions

Experimental group:

  1. HCV‐796 every 12 h for 14 days + peg‐IFN 2b 1.5 μg/kg/week.

  2. HCV‐796 + peg‐IFN 2a 180 μg/week.

Control group:

Control 1: placebo HCV‐796 + peg‐IFN 2b.

Control 2: placebo HCV‐796 + peg‐IFN 2a.

Outcomes

Antiviral activity

Notes

We contacted trial authors for additional information on allocation sequence generation and concealment, how was blinding maintained, were outcome assessment blinded, how many dropped out, how many were randomised to each group, SVR, death, SAE, prepublished protocol, how was the trial funded.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Described as double‐blind but it was unclear how the blinding was maintained

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Not described

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was unclear how many participants dropped out

Selective reporting (reporting bias)

Unclear risk

No protocol could be found

Vested‐interest bias

High risk

Multiple authors were employees of Wyers

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Vince 2014

Methods

Randomised clinical trial

Participants

64 adult participants

Sex: 36 men, 28 women

Mean age: 45 years

Country: USA

Inclusion criteria: male or female participants 18–65 years old inclusive, with a BMI of 18–35 kg/m2; documented clinical history compatible with chronic HCV, including positive anti‐HCV antibody, presence of HCV RNA in the plasma for at least 6 months or liver biopsy within 24 months with histology consistent with chronic HCV infection; HCV genotype 1, 2, 3 or 4; plasma HCV RNA P5 log10 IU/mL; all participants agreed to use double‐barrier birth control (such as a condom plus spermicide) from screening through at least 90 days following the last dose of the study drug.

Exclusion criteria: pregnancy or breastfeeding; co‐infection with HBV or HIV; history or evidence of decompensated liver disease; prior clinical or histological evidence of cirrhosis; ALT or AST level > 3.0 ULN; history of HCC or findings suggestive of possible HCC; 1 or more additional known primary or secondary causes of liver disease, other than HCV; previous antiviral treatment for HCV; current abuse of alcohol or illicit drugs; or other clinically significant diseases that, in the opinion of the investigator, would jeopardise the safety of the participant or impact the validity of the study results.

Interventions

Experimental group: oral 25 mg, 50 mg, 100 mg of samatasvir once a day for 3 days, or 50 mg of samatasvir twice a day for 3 days.

Control group: placebo.

Outcomes

Safety assessment, pharmacokinetics, antiviral activity, NS5A sequence analysis.

Notes

We emailed Vince and colleagues on 27 April 2016 for additional information but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation sequence

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Randomisation code were "kept blinded to participants and clinical investigators" and matching placebo was used

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Randomisation code were "kept blinded to participants and clinical investigators" and matching placebo was used

Incomplete outcome data (attrition bias)
All outcomes

Low risk

There were no dropouts

Selective reporting (reporting bias)

High risk

The primary outcomes were changed (NCT01508156)

Vested‐interest bias

High risk

Idenix Pharmaceuticals, Inc

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Wedemeyer 2013

Methods

A phase IIb, randomised, double‐blind, active‐controlled, parallel‐group trial (PROPEL) (NCT00869661)

Participants

424 participants

Sex: 255 men (60.1%), 169 women (39.9%)

Location: North America, Europe, and Australia.

Inclusion criteria: participants with chronic hepatitis C infection genotype 1 or 4, age 18‐65 years, treatment‐naive, serum HCV RNA level of at least 50,000 IU/mL, liver biopsy consistent with chronic hepatitis C obtained within 24 calendar months before first dose of study drug (36 months for participants with cirrhosis or incomplete/transition to cirrhosis, fibrosis score 3‐4). Participants with fibrosis score 3‐4 were required to have had an abdominal ultrasound, computerised tomography scan, or magnetic resonance imaging scan without evidence of HCC (within 2 months prior to randomisation) and a serum alfa‐fetoprotein < 100 ng/mL.

Exclusion criteria: hepatitis A or B co‐infection, HIV co‐infection, history or evidence of other chronic liver disease other than HCV, history or evidence of decompensated liver disease, absolute neutrophil count < 1.5 x 109 cells/L, haemoglobin concentration < 12 g/dL in women and < 13 g/dL in men. Platelet count < 90 x 109 cells/L, history of renal disease, serum creatinine > 1.5 times the ULN, BMI < 18 or ≥ 36 kg/m2. Pregnant or breastfeeding women and male partners of pregnant women, inadequate forms of contraception in women of childbearing age and men with female partners of childbearing age (2 forms of contraception required).

Group A: 80 participants

Mean age: 47 years (range 18‐62)

Race, n(%): white: 70(88), black: 8(10), other: 2(3)

HCV genotype, n(%): 1a: 44(55), 1b: 28(35), 4: 8(10)

Cirrhosis, n(%): 17(21)

Group B: 81 participants

Mean age: 47 years (range 23‐62)

Race, n(%): white: 69(85), black: 9(11), other: 3(4)

HCV genotype, n(%): 1a: 51(63), 1b: 26(32), 4: 4(5)

Cirrhosis, n(%): 18(22)

Group C: 82 participants

Mean age: 47 years (range 21‐65)

Race, n(%): white: 70(85), black: 9(11), other: 3(4)

HCV genotype, n(%): 1a. 50(61), 1b: 26(32), 4: 6(7)

Cirrhosis, n(%): 18(22)

Group D: 81 participants

Mean age: 48 years (range 23‐60)

Race, n(%): white: 71(88), black: 6(7), other: 4(5)

HCV genotype, n(%): 1a: 56(69), 1b: 22(27), 4: 3(4)

Cirrhosis, n(%): 23(28)

Group E: 84 participants

Mean age: 48 years (range 22‐65)

Race, n(%): white: 75(89), black: 3(4), other: 6(7)

HCV genotype, n(%): 1a: 52(62), 1b: 25(30), 4: 7(8)

Cirrhosis, n(%): 19(23).

Interventions

Experimental group:

Group A: oral mericitabine 500 mg twice daily for 12 weeks.

Group B: oral mericitabine 1000 mg twice daily for 8 weeks.

Group C: oral mericitabine 1000 mg twice daily for 12 weeks.

Group D: oral mericitabine 1000 mg twice daily for 12 weeks.

Control group:

Group E: matched placebo orally twice daily for 12 weeks.

Co‐interventions:

Groups A, B, and C: peg‐IFN alfa‐2a 180 μg subcutaneously once weekly for 24 weeks ifeRVR achieved, or for 48 weeks if eRVR not achieved. Weight‐based oral RBV 1000 mg1‐200 mg daily in 2 divided doses for 24 weeks if eRVR achieved, or for 48 weeks if eRVR not achieved (eRVR was defined as undetectable HCV RNA (< 15 IU/mL) by week 4 and maintained through week 22).

Groups D and E: peg‐IFN α‐2a 180 μg subcutaneously once weekly for 48 weeks. Weight‐based oral RBV 1000 mg‐1200 mg daily in 2 divided doses for 48 weeks.

Outcomes

Primary outcome: SVR at week 24 after the last dose of study medication.

Secondary outcomes: viral responses at clinic visits (HCV RNA was determined at baseline and at weeks 1, 2, 4, 6, 8, 10, 12, 14, 18, 24, 30, 36, 42, 48 of treatment and at weeks 4, 12, and 24 of follow‐up). Proportion of participants with relapse.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Randomization was stratified by geographical region and the randomization sequence was generated centrally by the sponsor...The randomization list was not available to personnel at the study centers or to the sponsor’s monitors during the study."

Allocation concealment (selection bias)

Low risk

Quote: "...were randomized in enrollment order by central interactive voice‐response system or interactive web response system."

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

A mericitabine‐matched placebo was used. Quote: "Patients and investigators remained blinded to individual treatment assignments during 24/48 weeks of study treatment and 24 weeks of study follow‐up."

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "The randomization list was made available to selected individuals from the sponsor at the time of Data Monitoring Committee review of ˜50% of patients in Cohort 2 at week 12, an independent statistician at the sponsor for analysis of ongoing safety data and an independent medical officer to review interim analysis data."

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Number and reasons for withdrawal have been clearly stated.

Selective reporting (reporting bias)

Low risk

The protocol was published prior to randomisation and all pre‐specified outcomes were reported on.

Vested‐interest bias

High risk

Trial funded by Hoffmann‐LaRoche Ltd.

Other bias

Low risk

The trial appeared to be free of other bias domains that could put it at risk of bias.

Wilfret 2013

Methods

Randomised clinical trial

Participants

23 adult participants

Sex: 20 men, 3 women

Mean age: 51.5 years

Country: USA

Inclusion criteria: chronic HCV (for 6 months) were eligible if they were treatment‐naive and noncirrhotic with HCV RNA levels of > 100,000 IU/mL

Exclusion criteria: infected with HIV, HBV.

Interventions

The trial was divided into 5 cohorts

Experimental group: oral 1 mg, 10 mg, 30 mg, 60 mg, 120 mg in a single dose of GSK2336805.

Control group: placebo.

Outcomes

Safety analysis, pharmacokinetics, metabolite identification, clinical virology assessment.

Notes

We emailed Wilfret and colleagues on 27 April 2016 for additional information but reply not received yet.

The study included healthy volunteers.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

It was described that the trial was double‐blinded but it was unclear how the blinding of participants and personnel was performed

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Those performing the outcome assessment were not blinded

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

More than 5% dropped out

Selective reporting (reporting bias)

High risk

The order of the primary outcomes were changed

Vested‐interest bias

High risk

The trial was funded by GlaxoSmithKline

Other bias

Low risk

The trial appeared to be free of other components that could put it at risk of bias

Younossi 2015

Methods

Parallel‐group, randomised, placebo‐controlled (SIRIUS)

Participants

154 participants

Sex: 114 men, 40 women

Mean age: 56.5 (SD 9.2) years

Country: USA

Inclusion criteria: treatment‐experienced chronic hepatitis C participants with genotype 1. Compensated cirrhosis.

Interventions

Experimental group: ledipasvir and sofosbuvir for 24 weeks

Control group: placebo for 12 weeks, followed by ledipasvir, sofosbuvir, and RBV for 12 weeks.

Outcomes

Not stated.

Notes

Published only as abstract.

We emailed Younossi and colleagues on 27 April 2016 for additional information number of participants randomised per group, random sequence generation, method of allocation concealment, description of blinding procedure, blinding of outcome assessors, potential number and reasons for drop‐outs, pre‐defined outcomes, sponsorship and its role, race and ethnicity of participants, full text or at least the figure published in the abstract, and data from quality‐of‐life assessment but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Stated that trial was randomised, but method of sequence generation was not specified

Allocation concealment (selection bias)

Unclear risk

Method of allocation concealment was not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

It was unclear if participants and treatment providers were blinded

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

It was not mentioned if the outcome assessors were blinded

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Insufficient information provided

Selective reporting (reporting bias)

Unclear risk

Insufficient information provided

Vested‐interest bias

Unclear risk

It was uncertain how the trial was sponsored

Other bias

Low risk

The trial may or may not have been free of other domains that could put it at risk of bias

Zeuzem 2011a

Methods

A randomised, double‐blind, placebo‐controlled, parallel‐group, phase III trial (REALIZE) (NCT00703118)

Participants

662 participants

Location: Europe, South America, and North America

Inclusion criteria: age between 18 and 70 years, chronic hepatitis C infection, HCV genotype 1, HCV RNA level ≥ 1000 IU/mL, previously treated, but not achieving SVR, a liver biopsy within 18 months before screening, absolute neutrophil count ≥ 1200 cells/mm3, platelet count ≥ 90,000 cells/mm3, haemoglobin level ≥ 12 g/dL for women, and ≥ 13 g/dL for men

Exclusion criteria: decompensated liver disease, other causes of significant liver disease, other severe active diseases

Group 1: 266 participants (T12PR48)

Sex: 183 men 83 women

Mean age: 51 years (range 23‐69)

Race, n(%): white: 246(92), black: 11(4), Asian or other: 9(3)

HCV genotype, n(%): 1a: 118(44), 1b: 121(45), 1c: 0, unknown: 27(10)

HCV RNA ≥ 800,000 IU/mL, n(%): 238(89)

Stage of fibrosis or cirrhosis, n(%): no or minimal fibrosis: 51(19), portal fibrosis: 83(31), bridging fibrosis: 60(23), cirrhosis: 72(27).

Group 2: 264 participants (lead‐in T12PR48)

Sex: 189 men, 75 women

Mean age: 51 years (range 24‐70)

Race, n(%): white: 252(95), black: 8(3), Asian or other: 4(2)

HCV genotype, n(%): 1a: 121(46), 1b: 115(44), 1c: 0, unknown: 28(11)

HCV RNA ≥ 800,000 IU/mL, n(%): 234(89)

Stage of fibrosis or cirrhosis, n(%): no or minimal fibrosis: 68(26), portal fibrosis: 71(27), bridging fibrosis: 58(22), cirrhosis: 67(25).

Control group: 132 participants (PR48)

Sex: 88 men, 44 women

Mean age: 50 years (range 21‐69)

Race, n(%): white: 117(89), black: 11(8), Asian or other: 4(3)

HCV genotype, n(%): 1a: 59(45), 1b: 59(45), 1c: 1(1), unknown: 13(10)

HCV RNA ≥ 800,000 IU/mL, n(%): 114(86)

Stage of fibrosis or cirrhosis, n(%): no or minimal fibrosis: 35(27), portal fibrosis: 38(29), bridging fibrosis: 29(22), cirrhosis: 30(23).

Interventions

Experimental group:

  1. oral telaprevir 750 mg every 8 h for 12 weeks.

  2. oral telaprevir 750 mg every 8 h for 12 weeks, beginning at week 5.

Control group: placebo.

Co‐interventions: peg‐IFN α‐2a 180 μg subcutaneously once weekly and oral weight‐based RBV 1000 mg‐1200 mg in 2 divided daily doses for 48 weeks.

Outcomes

Primary outcome: proportion of participants with SVR at week 24 (undetectable HCV RNA 24 weeks after end of treatment).

Secondary outcomes: effect of lead‐in treatment.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Randomization was performed with the use of a centralized system according to a predefined randomization list, constructed through random permuted blocks..."

Allocation concealment (selection bias)

Low risk

Allocation concealment was obtained by use of an interactive voice‐response/web‐response system (IVRS/IWRS). Treatment codes were assigned by the system to the participants, and all codes were kept by IVRS/IWRS and could only be broken by contacting the IVRS/IWRS

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

A telaprevir‐matching placebo was used. All participants and study personnel and sponsor were unaware of treatment assignment

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "Results of HCV RNA tests up to week 24 were masked and were monitored by an independent reviewer to assess whether participants had met a predefined stopping rule..."

Incomplete outcome data (attrition bias)
All outcomes

High risk

Number and reasons for discontinuation were clearly stated. However, the discontinuation rate was very high, from 30%‐38% in the experimental groups, up to 62% in the control group. A majority of participants in the experimental groups discontinued treatment due to AEs, while the main reason for discontinuation in the control group was reaching the virologic stopping rule

Selective reporting (reporting bias)

Low risk

The study protocol was available and all pre‐specified outcomes were reported on

Vested‐interest bias

High risk

The sponsor (Janssen) was directly involved in trial design and protocol development, as well as editorial assistance in the preparation of the manuscript

Other bias

Low risk

Trial seems to be free of other potential sources of bias

Zeuzem 2014a

Methods

A phase III, randomised, placebo‐controlled, double‐blind, parallel‐group trial (SAPPHIRE‐II) (NCT01715415)

Participants

394 participants

Sex: 227 men, 167 women

Location: Australia, North America, and Europe

Inclusion criteria: age between 18 and 70 years, prior null‐responder, partial responder, or relapser to peg‐IFN/RBV treatment. Chronic hepatitis C HCV genotype 1, no cirrhosis, HCV RNA level > 10,000 IU/mL

Exclusion criteria: recent history of drug or alcohol abuse (within 6 months prior to study drug administration), HVB or HIV co‐infection, history of uncontrolled seizures, history of uncontrolled diabetes, active malignancy or history of malignancy, ALT > 5 x ULN, AST > 5 x ULN, calculated creatinine clearance < 60 mL/min, albumin < lower limit of normal (LLN), prothrombin time/international normalised ratio > 1.5, haemoglobin < LLN, platelets < 120,000 cells per mm3, absolute neutrophil count < 1500 cells/μL, indirect bilirubin > 1.5 ULN and direct bilirubin > ULN

Group 1: 297 participants

Sex: 167 men, 130 women

Mean age: 51.7 years (range: 19.0‐71.0)

Race, n(%): white: 269(90.6), black: 22(7.4), Asian: 6(2.0)

Fibrosis score F2‐F3, n(%): 95(32.0)

IL28B genotype CC, n(%): 34(11.4)

HCV genotype, n(%): 1a: 173(58.2), 1b: 123(41.4)

Group 2: 97 participants

Sex: 60 men, 37 women

Mean age: 54.9 years (range 30.0‐69.0)

Race, n(%): white: 86(88.7), black: 10(10.3), Asian: 0

Fibrosis score F2‐F3, n(%): 32(33.0)

IL28B genotype CC, n(%): 7(7.2)

HCV genotype, n(%): 1a: 57(58.8), 1b: 40(41.2)

Interventions

Experimental group:

Group 1: ABT‐450 orally 150 mg once daily with ritonavir 100 mg once daily and ombitasvir 25 mg once daily for 12 weeks. Dasabuvir orally 250 mg twice daily for 12 weeks

Control group:

Group 2: matching placebos for 12 weeks, followed by an open‐label period of 12 weeks' administration of the active treatment

Co‐intervention: oral weight‐based RBV 1000 mg‐1200 mg in 2 divided daily doses (1000 mg daily if body weight was < 75 kg and 1200 mg daily if body weight was ≥ 75 kg)

Outcomes

Primary outcome: SVR 12 weeks after the end of study treatment. AEs

Secondary outcomes: virological failure during treatment. Post‐treatment relapse. Percentage of participants with ALT normalisation at the final treatment visit among participants with ALT > ULN at baseline

Notes

We emailed Zeuzem and colleagues on 27 April 2016 for additional information on SVR for placebo group, normalisation of ALT level after treatment but reply not received yet.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated schedule

Allocation concealment (selection bias)

Low risk

Allocation was performed "through IRT (interactive response technology) system in order to receive unique study bottle/kit numbers and a unique randomisation number", which was used only by the sponsor for loading treatment assignments into the database.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Matching placebos were used identical to study drugs.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

An independent DMC received safety data and provided recommendations. All data were blinded to study all study personnel.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Number and reasons for discontinuation were clearly stated.

Selective reporting (reporting bias)

Low risk

Study protocol was published and available before randomisation. All pre‐specified outcomes were reported on.

Vested‐interest bias

High risk

The sponsor (AbbVie) was directly involved in study design, data analyses, drafting the manuscript, and submission for publication.

Other bias

Low risk

Trial seems to be free of other potential sources of bias.

AE: adverse events; ALT: alanine aminotransferase;AST: aspartate aminotransferase; BMI: body mass index; DAA: direct‐acting antiviral(s); ECG: electrocardiogram; EVR: early virological response;eRVR: extended rapid virological response; FDA: Food and Drug Administration; h: hour(s); HBV: hepatitis B virus; HCC: hepatocellular carcinoma; HCV: hepatitis C virus; HCV VL (viral load); LLOQ: lower limit of quantification; mRNA: messenger RNA; IFN: interferon; PK: protein kinase; P/R: peg‐interferon/RBV; RBV: ribavirin; RNA: ribonucleic acid; RVR: rapid virologic response; SAE: serious adverse events; SVR: sustained viral response; vs: versus; ULN: upper limit of normal

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

AGATE‐I 2015

All arms were treated with DAAs

ALLY 2015

All participants were treated with DAAs

ANNAPURNA 2013

All participants were treated with DAAs

APRICOT 2004

Participants were not treated with DAAs

ATOMIC 2013

All participants were treated with DAAs

ATTAIN 2015

All participants were treated with DAAs

AVIATOR 2015

Not a randomised clinical trial. All participants were treated with DAAs

Basu 2014b

The trial compared same treatment regimens (simeprevir 150 mg and sofosbuvir 400 mg) with concomitant different dosages of RBV (modified doses vs 1000 mg) and different treatment duration (24 weeks vs 16 weeks)

Bathgate 2011

Short review written as 'Clinical opinion' for RESPOND‐2 and SPRINT‐2 trials

Bognar 2011

A Markov model simulation

Bourgeois 2015

Wrong control (different doses of simeprevir)

C‐SURFER 2015

All participants were treated with DAAs

C‐WORTHY 2015

All participants were treated with DAAs

Chandra 2006b

Participants were healthy

CONCISE 2013

All participants were treated with DAAs

COSMOS 2014

All participants were treated with DAAs

Di Bisceglie 2014

The trial compared the same treatment in equal or different dosages (telaprevir and VX‐222) combined with or without peg‐IFN and RBV

Dore 2014

Pooled analysis from two different trials

Dusheiko 2015

Was an analyses of multiple trials. It was not clear which trials the study looked at.

Ferenci 2014

Wrong intervention (trial does not actually compare DAA with placebo/other medical intervention)

Ferrante 2011a

A Markov model projection

Ferrante 2011b

A Markov model projection

Ferrante 2013

A Markov model projection

Foster 2010

Pooled analysis of data from different trials

FOURward 2014

Parallel‐group design, no control arm

FUSION 2013

No control arm

Gardner 2014b

Participants were healthy

HCVerso 1 2014

No control group

HCVerso 2 2014

No control group

ION‐3 2014

Parallel‐group design, no control arm

Jacobson 2013

Pooled analysis from two different trials

Kawada 2015

Wrong control

Liu 2015b

RBV was assessed as active treatment

Lok 2010

Wrong control (different doses of DAA)

Lok 2011

Wrong control (different doses of DAA)

Lok 2012a

Wrong control (different doses of DAA)

Lok 2012b

Wrong control (different doses of DAA)

Lok 2014

Wrong control (different doses of DAA)

MALACHITE‐I 2016

Wrong control group (control group received another DAA)

MALACHITE‐II 2016

Wrong control group (control group received another DAA)

Manns 2014b

Combined analysis of 3 trials

Manns 2015

Compared the same treatment (ledipasvir/sofosbuvir + RBV) of different duration (12 weeks vs 24 weeks)

Mendez 2014

Not a randomised clinical trial (compared other trials)

Mizokami 2015

Wrong intervention/control (compared RBV vs no RBV)

Molina 2015

Not randomised

Muir 2011

Not randomised

Muir 2015

Not randomised

NEUTRINO 2013

Single‐group, open label study

Nishiguchi 2014b

No control group

Nomura 2014

Not randomised

NUCLEAR 2013

Parallel‐group design, no control arm

OPTIMIST‐1 2015

Parallel‐group design, no control group

OPTIMIZE 2013

Wrong control (different time points of telaprevir)

Poordad 2014

The trial compared different treatment durations (12 weeks vs 24 weeks) of the same treatment regimen (ABT‐450/r‐ombitasvir, dasabuvir, and RBV)

Proulx 2008

Healthy volunteers

Reddy 2011

Combined analysis of three trials

Serfaty 2012

Wrong control (all groups received DAA)

Sulkowski 2011

Retrospective study

Sulkowski 2012a

Wrong intervention/control (The trial compared ribavirin versus no ribavirin). Same as Sulkowski 2014 (NCT01359644)

Sulkowski 2012b

Wrong control group (no groups could be used as control)

Sulkowski 2013d

Trial comparing different dosages of the same DAA

Sulkowski 2014

Wrong intervention/control (compared RBV versus no RBV)

Zeuzem 2012

Study evaluating 5 arms of participants treated with same drug regimen comparing different dosages, treatment durations, and/or RBV co‐intervention

Zeuzem 2013

Evaluated different dosages of the same treatment regimen

Zeuzem 2014b

The trial was initially designed as a multicenter, phase 3, randomised, placebo‐controlled, double‐blind trial of sofosbuvir + RBV vs placebo + RBV. Based on new published information, the protocol was amended and the study was redefined as a descriptive study in which the groups were unblinded, the placebo group was terminated, and the study assessed sofosbuvir + RBV for 12 weeks vs sofosbuvir + RBV for 24 weeks

DAA: direct‐acting antivirals; HCV: hepatitis C virus; peg‐IFN: pegylated interferon; RBV: ribavirin; vs: versus

Characteristics of ongoing studies [ordered by study ID]

Izumi 2012

Trial name or title

D‐Lite

Methods

Randomised clinical trial

Participants

165 adults with chronic hepatitis C, genotype 1, HCV RNA > 100,000 IU/mL at screening, seronegative for HIV and Hepatitis B surface antigen, liver biopsy within prior 2 years; subjects with compensated cirrhosis can enrol and will be capped at approximately 10%

Interventions

BMS‐790052 or BMS‐650032

Outcomes

Starting date

4 March 2011

Contact information

Notes

NCT01309932

Lawitz 2014b

Trial name or title

A randomised study to evaluate the safety and efficacy of IDX719 in combinations with simeprevir and/or TMC647055/ritonavir with or without ribavirin for 12 weeks in subjects with chronic hepatitis C infection

Methods

Randomised clinical trial

Participants

Treatment‐naïve, genotype 1b, 4 and 6 hepatitis C virus‐infected participants

Interventions

Samatasvir

Outcomes

Starting date

6 May 2013

Contact information

Notes

NCT01852604

Data and analyses

Open in table viewer
Comparison 1. DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Hepatitis C‐related morbidity or all‐cause mortality Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.1

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 1 Hepatitis C‐related morbidity or all‐cause mortality.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 1 Hepatitis C‐related morbidity or all‐cause mortality.

2 Hepatitis C‐related morbidity or all‐cause mortality ‐ bias risk Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.2

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 2 Hepatitis C‐related morbidity or all‐cause mortality ‐ bias risk.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 2 Hepatitis C‐related morbidity or all‐cause mortality ‐ bias risk.

2.1 Trials at high risk of bias

71

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.2 Trials at low risk of bias

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to type of DAA Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.3

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 3 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to type of DAA.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 3 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to type of DAA.

3.1 ABT‐072

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.2 ACH‐2684

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.3 Alisporivir

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.4 ALS‐2200

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.5 Asunaprevir

6

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.6 Balapiravir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.7 Beclabuvir

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.8 BILB‐1941

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.9 BIT‐225

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.10 Boceprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.11 Ciluprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.12 Daclatasvir

14

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.13 Danoprevir

9

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.14 Dasabuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.15 Deleobuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.16 Faldaprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.17 Filibuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.18 Grazoprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.19 GS‐6620

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.20 GS‐9256

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.21 GS‐9451

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.22 GS‐9669

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.23 GS‐9851

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.24 GS‐9857

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.25 GSK2336805

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.26 GSK2878175

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.27 IDX‐184

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.28 INX‐08189

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.29 Ledispasvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.30 Mericitabine

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.31 Narlaprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.32 Nesbuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.33 Odalasavir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.34 Ombitasvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.35 Paritaprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.36 PHX1766

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.37 PPI‐461

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.38 PSI‐352938

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.39 Samatasvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.40 Setrobuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.41 Simeprevir

14

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.42 Sofosbuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.43 Sovaprevir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.44 Tegobuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.45 Telaprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.46 Valopicitabine

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.47 Vaniprevir

9

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.48 VCH‐759

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.49 VCH‐916

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.50 Velpatasvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.51 VX‐222

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.52 Mixed

4

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to group of DAA Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.4

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 4 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to group of DAA.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 4 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to group of DAA.

4.1 Cyclophilin

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.2 NS3/NS4A inhibitors

41

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.3 NS5B inhibitors (NPI)

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.4 NS5B inhibitors (NNPI)

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.5 NS5A inhibitors

18

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.6 VPU‐ion channel inhibitors

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.7 Mixed

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to HIV‐infection Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.5

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 5 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to HIV‐infection.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 5 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to HIV‐infection.

5.1 With HIV‐infection

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.2 Without HIV‐infection

69

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.3 Mixed (with and without HIV‐infection)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.4 Unclear

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to comorbidity Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.6

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 6 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to comorbidity.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 6 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to comorbidity.

6.1 With comorbidity

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6.2 Without comorbidity

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6.3 Unclear

71

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to viral genotype Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.7

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 7 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to viral genotype.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 7 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to viral genotype.

7.1 Genotype 1

57

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.2 Genotype 2

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.3 Genotype 3

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.4 Genotype 4

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.5 Mixed

14

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to human genotype (IL28b) Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.8

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 8 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to human genotype (IL28b).

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 8 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to human genotype (IL28b).

8.1 IL28b (CC)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.2 IL28B (CT)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.3 IL28B (TT)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.4 IL28B (CT + TT)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.5 Mixed

71

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to Asian‐region Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.9

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 9 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to Asian‐region.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 9 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to Asian‐region.

9.1 From Asian region

8

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.2 Not from Asian region

52

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.3 Mixed

11

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.4 Unclear

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to specific ethnicities Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.10

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 10 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to specific ethnicities.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 10 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to specific ethnicities.

10.1 White

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.2 Black

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.3 Hispanic

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.4 Mixed

70

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.5 Unclear

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to reaching planned sample size Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.11

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 11 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to reaching planned sample size.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 11 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to reaching planned sample size.

11.1 Trials reaching planned sample size

10

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.2 Trials not reaching planned sample size

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.3 Unclear

58

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to prior treatment Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.12

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 12 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to prior treatment.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 12 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to prior treatment.

12.1 Treatment‐naive

47

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.2 Treatment‐experienced

16

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.3 Mixed

8

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.4 Unclear

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to interferon Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.13

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 13 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to interferon.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 13 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to interferon.

13.1 Trials where both groups received interferon

52

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13.2 Trials where neither group received interferon

19

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to ribavirin Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.14

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 14 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to ribavirin.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 14 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to ribavirin.

14.1 Trials where both groups received ribavirin

52

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14.2 Trials where neither group received ribavirin

19

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to chronic kidney disease Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.15

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 15 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to chronic kidney disease.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 15 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to chronic kidney disease.

15.1 With chronic kidney disease

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.2 Without chronic kidney disease

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.3 Unclear

71

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to cryoglobulinaemia Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.16

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 16 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to cryoglobulinaemia.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 16 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to cryoglobulinaemia.

16.1 With cryoglobulinaemia

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.2 Without cryoglobulinaemia

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.3 Unclear

71

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

17 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to DAA group as co‐intervention Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.17

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 17 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to DAA group as co‐intervention.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 17 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to DAA group as co‐intervention.

17.1 Trials where DAA were used as co‐intervention

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

17.2 Trials where DAA were not a co‐intervention

69

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

18 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to median dose Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.18

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 18 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to median dose.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 18 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to median dose.

18.1 Over or equal to median dose

41

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

18.2 Under median dose

27

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

18.3 Not available

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 2. DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Serious adverse events Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.1

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 1 Serious adverse events.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 1 Serious adverse events.

2 Serious adverse events ‐ bias risk Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.2

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 2 Serious adverse events ‐ bias risk.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 2 Serious adverse events ‐ bias risk.

2.1 Trials at high risk of bias

101

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.2 Trials at low risk of bias

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3 Serious adverse events ‐ according to type of DAA Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.3

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 3 Serious adverse events ‐ according to type of DAA.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 3 Serious adverse events ‐ according to type of DAA.

3.1 ABT‐072

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.2 ACH‐2684

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.3 Alisporivir

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.4 ALS‐2200

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.5 Asunaprevir

6

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.6 Balapiravir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.7 Beclabuvir

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.8 BILB‐1941

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.9 BIT‐225

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.10 Boceprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.11 Ciluprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.12 Daclatasvir

14

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.13 Danoprevir

9

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.14 Dasabuvir

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.15 Deleobuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.16 Faldaprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.17 Filibuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.18 Grazoprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.19 GS‐6620

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.20 GS‐9256

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.21 GS‐9451

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.22 GS‐9669

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.23 GS‐9851

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.24 GS‐9857

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.25 GSK2336805

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.26 GSK2878175

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.27 IDX‐184

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.28 INX‐08189

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.29 Ledispasvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.30 Mericitabine

7

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.31 Narlaprevir

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.32 Nesbuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.33 Odalasavir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.34 Ombitasvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.35 Paritaprevir

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.36 PHX1766

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.37 PPI‐461

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.38 PSI‐352938

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.39 Samatasvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.40 Setrobuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.41 Simeprevir

18

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.42 Sofosbuvir

4

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.43 Sovaprevir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.44 Tegobuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.45 Telaprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.46 Valopicitabine

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.47 Vaniprevir

10

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.48 VCH‐759

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.49 VCH‐916

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.50 Velpatasvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.51 VX‐222

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.52 Mixed

7

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4 Serious adverse events ‐ according to group of DAA Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.4

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 4 Serious adverse events ‐ according to group of DAA.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 4 Serious adverse events ‐ according to group of DAA.

4.1 Cyclophilin

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.2 NS3/NS4A inhibitors

56

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.3 NS5B inhibitors (NPI)

8

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.4 NS5B inhibitors (NNPI)

5

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.5 NS5A inhibitors

25

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.6 VPU‐ion channel inhibitors

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.7 Mixed

4

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5 Serious adverse events ‐ according to HIV‐infection Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.5

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 5 Serious adverse events ‐ according to HIV‐infection.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 5 Serious adverse events ‐ according to HIV‐infection.

5.1 With HIV‐infection

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.2 Without HIV‐infection

94

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.3 Mixed (with and without HIV‐infection)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.4 Unclear

7

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6 Serious adverse events ‐ according to comorbidity Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.6

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 6 Serious adverse events ‐ according to comorbidity.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 6 Serious adverse events ‐ according to comorbidity.

6.1 With comorbidity

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6.2 Without comorbidity

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6.3 Unclear

101

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7 Serious adverse events ‐ according to viral genotype Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.7

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 7 Serious adverse events ‐ according to viral genotype.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 7 Serious adverse events ‐ according to viral genotype.

7.1 Genotype 1

84

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.2 Genotype 2

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.3 Genotype 3

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.4 Genotype 4

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.5 Mixed

17

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8 Serious adverse events ‐ according to human genotype (IL28b) Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.8

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 8 Serious adverse events ‐ according to human genotype (IL28b).

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 8 Serious adverse events ‐ according to human genotype (IL28b).

8.1 IL28b (CC)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.2 IL28B (CT)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.3 IL28B (TT)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.4 IL28B (CT + TT)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.5 Mixed

101

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9 Serious adverse events ‐ according to Asian‐region Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.9

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 9 Serious adverse events ‐ according to Asian‐region.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 9 Serious adverse events ‐ according to Asian‐region.

9.1 From Asian region

10

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.2 Not from Asian region

76

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.3 Mixed

11

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.4 Unclear

4

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10 Serious adverse events ‐ according to specific ethnicities Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.10

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 10 Serious adverse events ‐ according to specific ethnicities.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 10 Serious adverse events ‐ according to specific ethnicities.

10.1 White

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.2 Black

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.3 Hispanic

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.4 Mixed

101

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.5 Unclear

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11 Serious adverse events ‐ according to reaching planned sample size Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.11

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 11 Serious adverse events ‐ according to reaching planned sample size.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 11 Serious adverse events ‐ according to reaching planned sample size.

11.1 Trials reaching planned sample size

15

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.2 Trials not reaching planned sample size

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.3 Unclear

83

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12 Serious adverse events ‐ according to prior treatment Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.12

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 12 Serious adverse events ‐ according to prior treatment.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 12 Serious adverse events ‐ according to prior treatment.

12.1 Treatment‐naive

72

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.2 Treatment‐experienced

19

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.3 Mixed

9

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.4 Unclear

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13 Serious adverse events ‐ according to interferon Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.13

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 13 Serious adverse events ‐ according to interferon.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 13 Serious adverse events ‐ according to interferon.

13.1 Trials where both groups received interferon

69

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13.2 Trials where neither group received interferon

29

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13.3 Unclear

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14 Serious adverse events ‐ according to ribavirin Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.14

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 14 Serious adverse events ‐ according to ribavirin.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 14 Serious adverse events ‐ according to ribavirin.

14.1 Trials where both groups received ribavirin

73

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14.2 Trials where neither group received ribavirin

27

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14.3 Unclear

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15 Serious adverse events ‐ according to chronic kidney disease Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.15

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 15 Serious adverse events ‐ according to chronic kidney disease.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 15 Serious adverse events ‐ according to chronic kidney disease.

15.1 With chronic kidney disease

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.2 Without chronic kidney disease

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.3 Unclear

101

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16 Serious adverse events ‐ according to cryoglobulinaemia Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.16

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 16 Serious adverse events ‐ according to cryoglobulinaemia.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 16 Serious adverse events ‐ according to cryoglobulinaemia.

16.1 With cryoglobulinaemia

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.2 Without cryoglobulinaemia

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.3 Unclear

101

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

17 Serious adverse events ‐ according to DAA group as co‐intervention Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.17

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 17 Serious adverse events ‐ according to DAA group as co‐intervention.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 17 Serious adverse events ‐ according to DAA group as co‐intervention.

17.1 Trials where DAA were used as co‐intervention

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

17.2 Trials where DAA were not a co‐intervention

99

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

18 Serious adverse events ‐ according to median dose Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.18

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 18 Serious adverse events ‐ according to median dose.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 18 Serious adverse events ‐ according to median dose.

18.1 Over or equal to median dose

58

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

18.2 Under median dose

37

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

18.3 Not available

6

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 3. DAA on or on the way to the market versus placebo/no intervention (sustained virological response)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Without sustained virological response Show forest plot

61

7115

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.37, 0.52]

Analysis 3.1

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 1 Without sustained virological response.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 1 Without sustained virological response.

2 Without sustained virological response ‐ bias risk Show forest plot

61

7115

Odds Ratio (M‐H, Fixed, 95% CI)

0.24 [0.22, 0.27]

Analysis 3.2

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 2 Without sustained virological response ‐ bias risk.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 2 Without sustained virological response ‐ bias risk.

2.1 Trials at high risk of bias

61

7115

Odds Ratio (M‐H, Fixed, 95% CI)

0.24 [0.22, 0.27]

2.2 Trials at low risk of bias

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3 Without sustained virological response ‐ according to type of DAA Show forest plot

61

7115

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.37, 0.52]

Analysis 3.3

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 3 Without sustained virological response ‐ according to type of DAA.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 3 Without sustained virological response ‐ according to type of DAA.

3.1 ABT‐072

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.2 ACH‐2684

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.3 Alisporivir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.4 ALS‐2200

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.5 Asunaprevir

4

285

Risk Ratio (M‐H, Random, 95% CI)

0.49 [0.29, 0.85]

3.6 Balapiravir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.7 Beclabuvir

2

39

Risk Ratio (M‐H, Random, 95% CI)

0.78 [0.43, 1.40]

3.8 BILB‐1941

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.9 BIT‐225

1

23

Risk Ratio (M‐H, Random, 95% CI)

0.27 [0.03, 2.51]

3.10 Boceprevir

1

229

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.29, 0.61]

3.11 Ciluprevir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.12 Daclatasvir

7

619

Risk Ratio (M‐H, Random, 95% CI)

0.60 [0.50, 0.73]

3.13 Danoprevir

5

642

Risk Ratio (M‐H, Random, 95% CI)

0.38 [0.28, 0.51]

3.14 Dasabuvir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.15 Deleobuvir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.16 Faldaprevir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.17 Filibuvir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.18 Grazoprevir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.19 GS‐6620

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.20 GS‐9256

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.21 GS‐9451

1

329

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.26, 0.67]

3.22 GS‐9669

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.23 GS‐9851

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.24 GS‐9857

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.25 GSK2336805

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.26 GSK2878175

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.27 IDX‐184

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.28 INX‐08189

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.29 Ledispasvir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.30 Mericitabine

4

725

Risk Ratio (M‐H, Random, 95% CI)

0.78 [0.49, 1.27]

3.31 Narlaprevir

2

40

Risk Ratio (M‐H, Random, 95% CI)

0.69 [0.43, 1.09]

3.32 Nesbuvir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.33 Odalasavir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.34 Ombitasvir

1

37

Risk Ratio (M‐H, Random, 95% CI)

0.64 [0.39, 1.07]

3.35 Paritaprevir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.36 PHX1766

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.37 PPI‐461

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.38 PSI‐352938

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.39 Samatasvir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.40 Setrobuvir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.41 Simeprevir

19

2898

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.33, 0.46]

3.42 Sofosbuvir

3

181

Risk Ratio (M‐H, Random, 95% CI)

0.34 [0.20, 0.58]

3.43 Sovaprevir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.44 Tegobuvir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.45 Telaprevir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.46 Valopicitabine

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.47 Vaniprevir

9

333

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.25, 0.43]

3.48 VCH‐759

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.49 VCH‐916

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.50 Velpatasvir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.51 VX‐222

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.52 Mixed

2

735

Risk Ratio (M‐H, Random, 95% CI)

0.06 [0.00, 7.05]

4 Without sustained virological response ‐ according to group of DAA Show forest plot

61

7115

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.37, 0.52]

Analysis 3.4

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 4 Without sustained virological response ‐ according to group of DAA.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 4 Without sustained virological response ‐ according to group of DAA.

4.1 Cyclophilin

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

4.2 NS3/NS4A inhibitors

41

4756

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.36, 0.46]

4.3 NS5B inhibitors (NPI)

7

906

Risk Ratio (M‐H, Random, 95% CI)

0.57 [0.36, 0.90]

4.4 NS5B inhibitors (NNPI)

2

39

Risk Ratio (M‐H, Random, 95% CI)

0.78 [0.43, 1.40]

4.5 NS5A inhibitors

9

686

Risk Ratio (M‐H, Random, 95% CI)

0.59 [0.49, 0.69]

4.6 VPU‐ion channel inhibitors

1

23

Risk Ratio (M‐H, Random, 95% CI)

0.27 [0.03, 2.51]

4.7 Mixed

1

705

Risk Ratio (M‐H, Random, 95% CI)

0.01 [0.00, 0.02]

5 Without sustained virological response ‐ according to HIV‐infection Show forest plot

61

7115

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.37, 0.52]

Analysis 3.5

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 5 Without sustained virological response ‐ according to HIV‐infection.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 5 Without sustained virological response ‐ according to HIV‐infection.

5.1 With HIV‐infection

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

5.2 Without HIV‐infection

58

6726

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.37, 0.52]

5.3 Mixed (with and without HIV‐infection)

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

5.4 Unclear

3

389

Risk Ratio (M‐H, Random, 95% CI)

0.50 [0.35, 0.72]

6 Without sustained virological response ‐ according to comorbidity Show forest plot

61

7115

Odds Ratio (M‐H, Fixed, 95% CI)

0.24 [0.22, 0.27]

Analysis 3.6

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 6 Without sustained virological response ‐ according to comorbidity.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 6 Without sustained virological response ‐ according to comorbidity.

6.1 With comorbidity

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6.2 Without comorbidity

61

7115

Odds Ratio (M‐H, Fixed, 95% CI)

0.24 [0.22, 0.27]

6.3 Unclear

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7 Without sustained virological response ‐ according to viral genotype Show forest plot

58

7098

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.36, 0.51]

Analysis 3.7

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 7 Without sustained virological response ‐ according to viral genotype.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 7 Without sustained virological response ‐ according to viral genotype.

7.1 Genotype 1

54

5984

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.37, 0.50]

7.2 Genotype 2

3

185

Risk Ratio (M‐H, Random, 95% CI)

0.14 [0.01, 3.21]

7.3 Genotype 3

2

80

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.43, 1.43]

7.4 Genotype 4

5

226

Risk Ratio (M‐H, Random, 95% CI)

0.10 [0.02, 0.68]

7.5 Genotype 6

1

49

Risk Ratio (M‐H, Random, 95% CI)

0.01 [0.00, 0.20]

7.6 Mixed

2

574

Risk Ratio (M‐H, Random, 95% CI)

0.92 [0.52, 1.62]

8 Without sustained virological response ‐ according to human genotype (IL28b) Show forest plot

58

6745

Risk Ratio (M‐H, Random, 95% CI)

0.46 [0.40, 0.54]

Analysis 3.8

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 8 Without sustained virological response ‐ according to human genotype (IL28b).

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 8 Without sustained virological response ‐ according to human genotype (IL28b).

8.1 IL28b (CC)

25

1444

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.29, 0.61]

8.2 IL28B (CT)

10

1304

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.42, 0.66]

8.3 IL28B (TT)

10

359

Risk Ratio (M‐H, Random, 95% CI)

0.54 [0.44, 0.67]

8.4 IL28B (CT + TT)

14

1798

Risk Ratio (M‐H, Random, 95% CI)

0.37 [0.23, 0.57]

8.5 Unclear

7

147

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.33, 0.68]

8.6 Mixed

26

1693

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.40, 0.63]

9 Without sustained virological response ‐ according to Asian‐region Show forest plot

61

7115

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.37, 0.52]

Analysis 3.9

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 9 Without sustained virological response ‐ according to Asian‐region.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 9 Without sustained virological response ‐ according to Asian‐region.

9.1 From Asian region

10

1128

Risk Ratio (M‐H, Random, 95% CI)

0.34 [0.28, 0.42]

9.2 Not from Asian region

42

4910

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.43, 0.60]

9.3 Mixed

7

1010

Risk Ratio (M‐H, Random, 95% CI)

0.19 [0.03, 1.17]

9.4 Unclear

2

67

Risk Ratio (M‐H, Random, 95% CI)

0.53 [0.35, 0.79]

10 Without sustained virological response ‐ according to specific ethnicities Show forest plot

61

7115

Odds Ratio (M‐H, Fixed, 95% CI)

0.24 [0.22, 0.27]

Analysis 3.10

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 10 Without sustained virological response ‐ according to specific ethnicities.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 10 Without sustained virological response ‐ according to specific ethnicities.

10.1 White

2

412

Odds Ratio (M‐H, Fixed, 95% CI)

0.24 [0.15, 0.38]

10.2 Black

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.3 Hispanic

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.4 Mixed

48

5384

Odds Ratio (M‐H, Fixed, 95% CI)

0.23 [0.20, 0.27]

10.5 Unclear

9

862

Odds Ratio (M‐H, Fixed, 95% CI)

0.28 [0.20, 0.39]

10.6 Asian

2

457

Odds Ratio (M‐H, Fixed, 95% CI)

0.38 [0.23, 0.63]

11 Without sustained virological response ‐ according to reaching planned sample size Show forest plot

61

7115

Odds Ratio (M‐H, Fixed, 95% CI)

0.24 [0.22, 0.27]

Analysis 3.11

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 11 Without sustained virological response ‐ according to reaching planned sample size.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 11 Without sustained virological response ‐ according to reaching planned sample size.

11.1 Trials reaching planned sample size

13

3071

Odds Ratio (M‐H, Fixed, 95% CI)

0.21 [0.18, 0.25]

11.2 Trials not reaching planned sample size

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.3 Unclear

48

4044

Odds Ratio (M‐H, Fixed, 95% CI)

0.28 [0.23, 0.33]

12 Without sustained virological response ‐ according to prior treatment Show forest plot

61

7115

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.37, 0.52]

Analysis 3.12

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 12 Without sustained virological response ‐ according to prior treatment.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 12 Without sustained virological response ‐ according to prior treatment.

12.1 Treatment‐naive

44

4777

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.41, 0.56]

12.2 Treatment‐experienced

13

1274

Risk Ratio (M‐H, Random, 95% CI)

0.50 [0.36, 0.69]

12.3 Mixed

4

1064

Risk Ratio (M‐H, Random, 95% CI)

0.15 [0.02, 0.96]

12.4 Unclear

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

13 Without sustained virological response ‐ according to interferon Show forest plot

61

7115

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.37, 0.52]

Analysis 3.13

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 13 Without sustained virological response ‐ according to interferon.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 13 Without sustained virological response ‐ according to interferon.

13.1 Trials where both groups received interferon

57

6229

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.41, 0.54]

13.2 Trials where neither group received interferon

2

735

Risk Ratio (M‐H, Random, 95% CI)

0.06 [0.00, 7.05]

13.3 Trials where only the experimental group received interferon

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

13.4 Trials where only the control group received interferon

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

13.5 Mixed

2

151

Risk Ratio (M‐H, Random, 95% CI)

0.58 [0.15, 2.30]

14 Without sustained virological response ‐ according to ribavirin Show forest plot

61

7115

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.37, 0.52]

Analysis 3.14

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 14 Without sustained virological response ‐ according to ribavirin.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 14 Without sustained virological response ‐ according to ribavirin.

14.1 Trials where both groups received ribavirin

60

6410

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.41, 0.55]

14.2 Trials where neither group received ribavirin

1

705

Risk Ratio (M‐H, Random, 95% CI)

0.01 [0.00, 0.02]

14.3 Trials where only the experimental group received ribavirin

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

14.4 Trials where only the control group received ribavirin

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

15 Without sustained virological response ‐ according to chronic kidney disease Show forest plot

61

7115

Odds Ratio (M‐H, Fixed, 95% CI)

0.24 [0.22, 0.27]

Analysis 3.15

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 15 Without sustained virological response ‐ according to chronic kidney disease.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 15 Without sustained virological response ‐ according to chronic kidney disease.

15.1 With chronic kidney disease

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.2 Without chronic kidney disease

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.3 Unclear

61

7115

Odds Ratio (M‐H, Fixed, 95% CI)

0.24 [0.22, 0.27]

16 Without sustained virological response ‐ according to cryoglobulinaemia Show forest plot

61

7115

Odds Ratio (M‐H, Fixed, 95% CI)

0.24 [0.22, 0.27]

Analysis 3.16

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 16 Without sustained virological response ‐ according to cryoglobulinaemia.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 16 Without sustained virological response ‐ according to cryoglobulinaemia.

16.1 With cryoglobulinaemia

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.2 Without cryoglobulinaemia

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.3 Unclear

61

7115

Odds Ratio (M‐H, Fixed, 95% CI)

0.24 [0.22, 0.27]

17 Without sustained virological response ‐ according to DAA group as co‐intervention Show forest plot

61

7115

Odds Ratio (M‐H, Fixed, 95% CI)

0.24 [0.22, 0.27]

Analysis 3.17

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 17 Without sustained virological response ‐ according to DAA group as co‐intervention.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 17 Without sustained virological response ‐ according to DAA group as co‐intervention.

17.1 Trials where DAA were used as co‐intervention

3

480

Odds Ratio (M‐H, Fixed, 95% CI)

0.42 [0.27, 0.66]

17.2 Trials where DAA were not a co‐intervention

58

6635

Odds Ratio (M‐H, Fixed, 95% CI)

0.23 [0.21, 0.26]

18 Without sustained virological response ‐ 'Best‐worst case' scenario Show forest plot

61

7294

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.34, 0.49]

Analysis 3.18

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 18 Without sustained virological response ‐ 'Best‐worst case' scenario.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 18 Without sustained virological response ‐ 'Best‐worst case' scenario.

19 Without sustained virological response ‐ 'Worst‐best case' scenario Show forest plot

61

7294

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.43, 0.60]

Analysis 3.19

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 19 Without sustained virological response ‐ 'Worst‐best case' scenario.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 19 Without sustained virological response ‐ 'Worst‐best case' scenario.

20 Without sustained virological response ‐ according to median dose Show forest plot

61

7115

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.37, 0.52]

Analysis 3.20

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 20 Without sustained virological response ‐ according to median dose.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 20 Without sustained virological response ‐ according to median dose.

20.1 Over or equal to median dose

34

4154

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.32, 0.53]

20.2 Under median dose

23

2086

Risk Ratio (M‐H, Random, 95% CI)

0.46 [0.39, 0.55]

20.3 Not available

4

875

Risk Ratio (M‐H, Random, 95% CI)

0.62 [0.26, 1.47]

Open in table viewer
Comparison 4. Danoprevir versus placebo/no intervention

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Hepatitis C‐related morbidity or all‐cause mortality Show forest plot

9

781

Odds Ratio (M‐H, Fixed, 95% CI)

0.56 [0.06, 5.19]

Analysis 4.1

Comparison 4 Danoprevir versus placebo/no intervention, Outcome 1 Hepatitis C‐related morbidity or all‐cause mortality.

Comparison 4 Danoprevir versus placebo/no intervention, Outcome 1 Hepatitis C‐related morbidity or all‐cause mortality.

2 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to dose Show forest plot

9

781

Odds Ratio (M‐H, Fixed, 95% CI)

0.56 [0.06, 5.19]

Analysis 4.2

Comparison 4 Danoprevir versus placebo/no intervention, Outcome 2 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to dose.

Comparison 4 Danoprevir versus placebo/no intervention, Outcome 2 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to dose.

2.1 Over or equal to median dose

6

606

Odds Ratio (M‐H, Fixed, 95% CI)

0.56 [0.06, 5.19]

2.2 Under median dose

3

175

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.3 Not available

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3 Serious adverse events Show forest plot

9

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 4.3

Comparison 4 Danoprevir versus placebo/no intervention, Outcome 3 Serious adverse events.

Comparison 4 Danoprevir versus placebo/no intervention, Outcome 3 Serious adverse events.

4 Serious adverse events ‐ according to median dose Show forest plot

9

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 4.4

Comparison 4 Danoprevir versus placebo/no intervention, Outcome 4 Serious adverse events ‐ according to median dose.

Comparison 4 Danoprevir versus placebo/no intervention, Outcome 4 Serious adverse events ‐ according to median dose.

4.1 Over or equal to median dose

6

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.2 Under median dose

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.3 Not available

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5 Without sustained virological response Show forest plot

5

642

Odds Ratio (M‐H, Fixed, 95% CI)

0.19 [0.12, 0.32]

Analysis 4.5

Comparison 4 Danoprevir versus placebo/no intervention, Outcome 5 Without sustained virological response.

Comparison 4 Danoprevir versus placebo/no intervention, Outcome 5 Without sustained virological response.

6 Without sustained virological response ‐ according to median dose Show forest plot

5

642

Odds Ratio (M‐H, Fixed, 95% CI)

0.19 [0.12, 0.32]

Analysis 4.6

Comparison 4 Danoprevir versus placebo/no intervention, Outcome 6 Without sustained virological response ‐ according to median dose.

Comparison 4 Danoprevir versus placebo/no intervention, Outcome 6 Without sustained virological response ‐ according to median dose.

6.1 Over or equal to median dose

4

537

Odds Ratio (M‐H, Fixed, 95% CI)

0.18 [0.11, 0.32]

6.2 Under median dose

1

105

Odds Ratio (M‐H, Fixed, 95% CI)

0.27 [0.07, 0.99]

6.3 Not available

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 5. All DAA versus placebo/no intervention/other medical intervention (morbidity or all‐cause mortality analyses)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Hepatitis C‐related morbidity or all‐cause mortality Show forest plot

95

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 5.1

Comparison 5 All DAA versus placebo/no intervention/other medical intervention (morbidity or all‐cause mortality analyses), Outcome 1 Hepatitis C‐related morbidity or all‐cause mortality.

Comparison 5 All DAA versus placebo/no intervention/other medical intervention (morbidity or all‐cause mortality analyses), Outcome 1 Hepatitis C‐related morbidity or all‐cause mortality.

1.1 Trials assessing DAAs on or on the way to the market

71

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.2 Trials assessing DAAs withdrawn from market

22

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.3 Trials using other medical intervention as control group

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.4 Trials using other medical intervention as experimental group

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Hepatitis C‐related morbidity or all‐cause mortality ‐ drugs not discontinued

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.1 Trials assessing discontinued drugs

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.2 Trials assessing drugs still used

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3 Hepatitis C‐related morbidity or all‐cause mortality ‐ bias risk

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.1 Trials with a high risk of bias

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.2 Trials with a low risk of bias

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to type of DAA

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.1 ABT‐072

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.2 ACH‐2684

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.3 Alisporivir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.4 ALS‐2200

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.5 Asunaprevir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.6 Balapiravir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.7 Beclabuvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.8 BILB‐1941

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.9 BIT‐225

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.10 Boceprevir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.11 Ciluprevir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.12 Daclatasvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.13 Danoprevir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.14 Dasabuvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.15 Deleobuvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.16 Faldaprevir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.17 Filibuvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.18 Grazoprevir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.19 GS‐6620

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.20 GS‐9256

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.21 GS‐9451

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.22 GS‐9669

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.23 GS‐9851

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.24 GS‐9857

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.25 GSK2336805

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.26 GSK2878175

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.27 IDX‐184

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.28 INX‐08189

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.29 Ledispasvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.30 Mericitabine

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.31 Narlaprevir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.32 Nesbuvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.33 Odalasavir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.34 Ombitasvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.35 Paritaprevir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.36 PHX1766

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.37 PPI‐461

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.38 PSI‐352938

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.39 Samatasvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.40 Setrobuvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.41 Simeprevir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.42 Sofosbuvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.43 Sovaprevir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.44 Tegobuvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.45 Telaprevir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.46 Valopicitabine

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.47 Vaniprevir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.48 VCH‐759

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.49 VCH‐916

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.50 Velpatasvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.51 VX‐222

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.52 Mixed

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to group of DAA

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.1 Cyclophilin

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.2 NS3/NS4A inhibitors

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.3 NS5B inhibitors (NPI)

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.4 NS5B inhibitors (NNPI)

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.5 NS5A inhibitors

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.6 VPU‐ion channel inhibitors

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.7 Mixed

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to HIV‐infection

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6.1 With HIV‐infection

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6.2 Without HIV‐infection

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6.3 Mixed (with and without HIV‐infection)

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6.4 Unclear

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to comorbidity

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.1 With comorbidity

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.2 Without comorbidity

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.3 Unclear

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to viral genotype

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.1 Genotype 1

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.2 Genotype 2

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.3 Genotype 3

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.4 Genotype 4

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.5 Unclear

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to human genotype (IL28b)

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.1 IL28b (CC)

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.2 IL28B (CT)

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.3 IL28B (TT)

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.4 IL28B (CT + TT)

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.5 Unclear

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to Asian‐region

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.1 From Asian region

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.2 Not from Asian region

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.3 Mixed

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.4 Unclear

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to specific ethnicities

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.1 White

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.2 Black

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.3 Hispanic

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.4 Mixed

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.5 Unclear

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to reaching planned sample size

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.1 Trials reaching planned sample size

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.2 Trials not reaching planned sample size

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.3 Unclear

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to prior treatment

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13.1 Treatment‐naive

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13.2 Treatment‐experienced

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13.3 Mixed

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13.4 Unclear

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to interferon

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14.1 Trials where both groups received interferon

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14.2 Trials where neither group received interferon

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14.3 Trials where only the experimental group received interferon

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14.4 Trials where only the control group received interferon

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to ribavirin

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.1 Trials where both groups received ribavirin

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.2 Trials where neither group received ribavirin

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.3 Trials where only the experimental group received ribavirin

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.4 Trials where only the control group received ribavirin

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to chronic kidney disease

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.1 With chronic kidney disease

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.2 Without chronic kidney disease

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.3 Unclear

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

17 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to cryoglobulinaemia

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

17.1 With cryoglobulinaemia

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

17.2 Without cryoglobulinaemia

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

17.3 Unclear

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

18 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to DAA group as co‐intervention

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

18.1 Trials where DAA were used as co‐intervention

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

18.2 Trials where DAA were not a co‐intervention

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 6. All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Serious adverse events Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 6.1

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 1 Serious adverse events.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 1 Serious adverse events.

1.1 Trials assessing DAAs on or on the way to the market

101

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.2 Trials assessing DAAs withdrawn from market

62

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.3 Trials using other medical intervention as control group

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.4 Trials using other medical intervention as experimental group

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Serious adverse events ‐ bias risk Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 6.2

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 2 Serious adverse events ‐ bias risk.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 2 Serious adverse events ‐ bias risk.

2.1 Trials with a high risk of bias

167

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.2 Trials with a low risk of bias

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3 Serious adverse events ‐ according to type of DAA Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 6.3

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 3 Serious adverse events ‐ according to type of DAA.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 3 Serious adverse events ‐ according to type of DAA.

3.1 ABT‐072

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.2 ACH‐2684

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.3 Alisporivir

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.4 ALS‐2200

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.5 Asunaprevir

6

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.6 Balapiravir

9

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.7 Beclabuvir

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.8 BILB‐1941

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.9 BIT‐225

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.10 Boceprevir

13

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.11 Ciluprevir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.12 Daclatasvir

14

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.13 Danoprevir

9

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.14 Dasabuvir

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.15 Deleobuvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.16 Faldaprevir

13

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.17 Filibuvir

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.18 Grazoprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.19 GS‐6620

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.20 GS‐9256

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.21 GS‐9451

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.22 GS‐9669

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.23 GS‐9851

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.24 GS‐9857

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.25 GSK2336805

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.26 GSK2878175

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.27 IDX‐184

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.28 INX‐08189

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.29 Ledispasvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.30 Mericitabine

7

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.31 Narlaprevir

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.32 Nesbuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.33 Odalasavir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.34 Ombitasvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.35 Paritaprevir

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.36 PHX1766

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.37 PPI‐461

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.38 PSI‐352938

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.39 Samatasvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.40 Setrobuvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.41 Simeprevir

19

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.42 Sofosbuvir

6

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.43 Sovaprevir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.44 Tegobuvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.45 Telaprevir

13

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.46 Valopicitabine

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.47 Vaniprevir

10

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.48 VCH‐759

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.49 VCH‐916

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.50 Velpatasvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.51 VX‐222

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.52 Mixed

8

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4 Serious adverse events ‐ according to group of DAA Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 6.4

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 4 Serious adverse events ‐ according to group of DAA.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 4 Serious adverse events ‐ according to group of DAA.

4.1 Cyclophilin

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.2 NS3/NS4A inhibitors

92

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.3 NS5B inhibitors (NPI)

24

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.4 NS5B inhibitors (NNPI)

14

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.5 NS5A inhibitors

27

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.6 VPU‐ion channel inhibitors

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.7 Mixed

7

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5 Serious adverse events ‐ according to HIV‐infection Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 6.5

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 5 Serious adverse events ‐ according to HIV‐infection.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 5 Serious adverse events ‐ according to HIV‐infection.

5.1 With HIV‐infection

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.2 Without HIV‐infection

154

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.3 Mixed (with and without HIV‐infection)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.4 Unclear

11

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6 Serious adverse events ‐ according to comorbidity Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 6.6

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 6 Serious adverse events ‐ according to comorbidity.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 6 Serious adverse events ‐ according to comorbidity.

6.1 With comorbidity

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6.2 Without comorbidity

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6.3 Unclear

167

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7 Serious adverse events ‐ according to viral genotype Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 6.7

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 7 Serious adverse events ‐ according to viral genotype.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 7 Serious adverse events ‐ according to viral genotype.

7.1 Genotype 1

138

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.2 Genotype 2

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.3 Genotype 3

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.4 Genotype 4

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.5 Mixed

26

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8 Serious adverse events ‐ according to human genotype (IL28b) Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 6.8

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 8 Serious adverse events ‐ according to human genotype (IL28b).

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 8 Serious adverse events ‐ according to human genotype (IL28b).

8.1 IL28b (CC)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.2 IL28B (CT)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.3 IL28B (TT)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.4 IL28B (CT + TT)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.5 Unclear

79

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.6 Mixed IL28b

88

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9 Serious adverse events ‐ according to Asian‐region Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 6.9

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 9 Serious adverse events ‐ according to Asian‐region.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 9 Serious adverse events ‐ according to Asian‐region.

9.1 From Asian region

12

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.2 Not from Asian region

119

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.3 Mixed

31

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.4 Unclear

5

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10 Serious adverse events ‐ according to specific ethnicities Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 6.10

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 10 Serious adverse events ‐ according to specific ethnicities.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 10 Serious adverse events ‐ according to specific ethnicities.

10.1 White

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.2 Black

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.3 Hispanic

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.4 Mixed

133

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.5 Unclear

31

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11 Serious adverse events ‐ according to reaching planned sample size

0

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

11.1 Trials reaching planned sample size

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.2 Trials not reaching planned sample size

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.3 Unclear

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12 Serious adverse events ‐ according to prior treatment Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 6.12

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 12 Serious adverse events ‐ according to prior treatment.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 12 Serious adverse events ‐ according to prior treatment.

12.1 Treatment‐naive

122

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.2 Treatment‐experienced

27

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.3 Mixed

18

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.4 Unclear

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13 Serious adverse events ‐ according to interferon Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 6.13

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 13 Serious adverse events ‐ according to interferon.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 13 Serious adverse events ‐ according to interferon.

13.1 Trials where both groups received interferon

126

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13.2 Trials where neither group received interferon

40

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13.3 Trials where only the experimental group received interferon

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13.4 Trials where only the control group received interferon

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14 Serious adverse events ‐ according to ribavirin Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 6.14

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 14 Serious adverse events ‐ according to ribavirin.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 14 Serious adverse events ‐ according to ribavirin.

14.1 Trials where both groups received ribavirin

127

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14.2 Trials where neither group received ribavirin

37

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14.3 Trials where only the experimental group received ribavirin

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14.4 Trials where only the control group received ribavirin

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15 Serious adverse events ‐ according to chronic kidney disease Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 6.15

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 15 Serious adverse events ‐ according to chronic kidney disease.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 15 Serious adverse events ‐ according to chronic kidney disease.

15.1 With chronic kidney disease

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.2 Without chronic kidney disease

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.3 Unclear

167

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16 Serious adverse events ‐ according to cryoglobulinaemia Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 6.16

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 16 Serious adverse events ‐ according to cryoglobulinaemia.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 16 Serious adverse events ‐ according to cryoglobulinaemia.

16.1 With cryoglobulinaemia

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.2 Without cryoglobulinaemia

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.3 Unclear

167

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

17 Serious adverse events ‐ according to DAA group as co‐intervention Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 6.17

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 17 Serious adverse events ‐ according to DAA group as co‐intervention.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 17 Serious adverse events ‐ according to DAA group as co‐intervention.

17.1 Trials where DAA were used as co‐intervention

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

17.2 Trials where DAA were not a co‐intervention

165

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 7. All DAA versus placebo/no intervention/other medical intervention (sustained virological response analyses)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Without sustained virological response Show forest plot

107

17101

Risk Ratio (M‐H, Random, 95% CI)

0.53 [0.48, 0.59]

Analysis 7.1

Comparison 7 All DAA versus placebo/no intervention/other medical intervention (sustained virological response analyses), Outcome 1 Without sustained virological response.

Comparison 7 All DAA versus placebo/no intervention/other medical intervention (sustained virological response analyses), Outcome 1 Without sustained virological response.

1.1 Trials assessing DAAs on or on the way to the market

60

6886

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.37, 0.52]

1.2 Trials assessing DAAs withdrawn from market

43

9075

Risk Ratio (M‐H, Random, 95% CI)

0.61 [0.55, 0.69]

1.3 Trials using other medical intervention as control group

3

862

Risk Ratio (M‐H, Random, 95% CI)

0.81 [0.36, 1.82]

1.4 Trials using other medical intervention as experimental group

1

278

Risk Ratio (M‐H, Random, 95% CI)

0.23 [0.17, 0.29]

Open in table viewer
Comparison 8. All DAA versus placebo/no intervention/other medical intervention (quality of life scores)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 SF‐36 physical score Show forest plot

1

215

Mean Difference (IV, Fixed, 95% CI)

‐1.17 [‐3.65, 1.31]

Analysis 8.1

Comparison 8 All DAA versus placebo/no intervention/other medical intervention (quality of life scores), Outcome 1 SF‐36 physical score.

Comparison 8 All DAA versus placebo/no intervention/other medical intervention (quality of life scores), Outcome 1 SF‐36 physical score.

2 SF‐36 mental score Show forest plot

1

215

Mean Difference (IV, Fixed, 95% CI)

1.36 [‐1.53, 4.25]

Analysis 8.2

Comparison 8 All DAA versus placebo/no intervention/other medical intervention (quality of life scores), Outcome 2 SF‐36 mental score.

Comparison 8 All DAA versus placebo/no intervention/other medical intervention (quality of life scores), Outcome 2 SF‐36 mental score.

Open in table viewer
Comparison 9. Daclatasvir versus placebo/no intervention

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Hepatitis C‐related morbidity or all‐cause mortality Show forest plot

14

666

Odds Ratio (M‐H, Fixed, 95% CI)

1.25 [0.06, 26.65]

Analysis 9.1

Comparison 9 Daclatasvir versus placebo/no intervention, Outcome 1 Hepatitis C‐related morbidity or all‐cause mortality.

Comparison 9 Daclatasvir versus placebo/no intervention, Outcome 1 Hepatitis C‐related morbidity or all‐cause mortality.

2 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to dose Show forest plot

14

666

Odds Ratio (M‐H, Fixed, 95% CI)

1.25 [0.06, 26.65]

Analysis 9.2

Comparison 9 Daclatasvir versus placebo/no intervention, Outcome 2 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to dose.

Comparison 9 Daclatasvir versus placebo/no intervention, Outcome 2 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to dose.

2.1 Over or equal to median dose

7

374

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.2 Under median dose

7

292

Odds Ratio (M‐H, Fixed, 95% CI)

1.25 [0.06, 26.65]

2.3 Not available

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3 Serious adverse events Show forest plot

13

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 9.3

Comparison 9 Daclatasvir versus placebo/no intervention, Outcome 3 Serious adverse events.

Comparison 9 Daclatasvir versus placebo/no intervention, Outcome 3 Serious adverse events.

4 Serious adverse events ‐ according to median dose Show forest plot

14

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 9.4

Comparison 9 Daclatasvir versus placebo/no intervention, Outcome 4 Serious adverse events ‐ according to median dose.

Comparison 9 Daclatasvir versus placebo/no intervention, Outcome 4 Serious adverse events ‐ according to median dose.

4.1 Over or equal to median dose

7

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.2 Under median dose

8

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.3 Not available

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5 Without sustained virological response Show forest plot

7

619

Odds Ratio (M‐H, Fixed, 95% CI)

0.40 [0.27, 0.59]

Analysis 9.5

Comparison 9 Daclatasvir versus placebo/no intervention, Outcome 5 Without sustained virological response.

Comparison 9 Daclatasvir versus placebo/no intervention, Outcome 5 Without sustained virological response.

6 Without sustained virological response ‐ according to median dose Show forest plot

7

619

Odds Ratio (M‐H, Fixed, 95% CI)

0.40 [0.27, 0.59]

Analysis 9.6

Comparison 9 Daclatasvir versus placebo/no intervention, Outcome 6 Without sustained virological response ‐ according to median dose.

Comparison 9 Daclatasvir versus placebo/no intervention, Outcome 6 Without sustained virological response ‐ according to median dose.

6.1 Over or equal to median dose

4

360

Odds Ratio (M‐H, Fixed, 95% CI)

0.43 [0.26, 0.70]

6.2 Under median dose

3

259

Odds Ratio (M‐H, Fixed, 95% CI)

0.36 [0.19, 0.68]

6.3 Not available

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 10. Simeprevir versus placebo/no intervention

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Hepatitis C‐related morbidity or all‐cause mortality Show forest plot

14

1589

Odds Ratio (M‐H, Fixed, 95% CI)

0.49 [0.08, 2.96]

Analysis 10.1

Comparison 10 Simeprevir versus placebo/no intervention, Outcome 1 Hepatitis C‐related morbidity or all‐cause mortality.

Comparison 10 Simeprevir versus placebo/no intervention, Outcome 1 Hepatitis C‐related morbidity or all‐cause mortality.

2 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to dose Show forest plot

14

1589

Odds Ratio (M‐H, Fixed, 95% CI)

0.49 [0.08, 2.96]

Analysis 10.2

Comparison 10 Simeprevir versus placebo/no intervention, Outcome 2 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to dose.

Comparison 10 Simeprevir versus placebo/no intervention, Outcome 2 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to dose.

2.1 Over or equal to median dose

4

441

Odds Ratio (M‐H, Fixed, 95% CI)

0.51 [0.03, 8.21]

2.2 Under median dose

8

705

Odds Ratio (M‐H, Fixed, 95% CI)

0.41 [0.01, 12.22]

2.3 Not available

2

443

Odds Ratio (M‐H, Fixed, 95% CI)

0.55 [0.02, 13.62]

3 Serious adverse events Show forest plot

18

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 10.3

Comparison 10 Simeprevir versus placebo/no intervention, Outcome 3 Serious adverse events.

Comparison 10 Simeprevir versus placebo/no intervention, Outcome 3 Serious adverse events.

4 Serious adverse events ‐ according to median dose Show forest plot

18

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 10.4

Comparison 10 Simeprevir versus placebo/no intervention, Outcome 4 Serious adverse events ‐ according to median dose.

Comparison 10 Simeprevir versus placebo/no intervention, Outcome 4 Serious adverse events ‐ according to median dose.

4.1 Over or equal to median dose

7

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.2 Under median dose

9

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.3 Not available

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5 Without sustained virological response Show forest plot

19

2898

Odds Ratio (M‐H, Fixed, 95% CI)

0.22 [0.19, 0.27]

Analysis 10.5

Comparison 10 Simeprevir versus placebo/no intervention, Outcome 5 Without sustained virological response.

Comparison 10 Simeprevir versus placebo/no intervention, Outcome 5 Without sustained virological response.

6 Without sustained virological response ‐ according to median dose Show forest plot

19

2898

Odds Ratio (M‐H, Fixed, 95% CI)

0.22 [0.19, 0.27]

Analysis 10.6

Comparison 10 Simeprevir versus placebo/no intervention, Outcome 6 Without sustained virological response ‐ according to median dose.

Comparison 10 Simeprevir versus placebo/no intervention, Outcome 6 Without sustained virological response ‐ according to median dose.

6.1 Over or equal to median dose

9

1765

Odds Ratio (M‐H, Fixed, 95% CI)

0.25 [0.20, 0.32]

6.2 Under median dose

8

696

Odds Ratio (M‐H, Fixed, 95% CI)

0.19 [0.13, 0.29]

6.3 Not available

2

437

Odds Ratio (M‐H, Fixed, 95% CI)

0.13 [0.07, 0.24]

Open in table viewer
Comparison 11. Vaniprevir versus placebo/no intervention

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Hepatitis C‐related morbidity or all‐cause mortality Show forest plot

9

379

Odds Ratio (M‐H, Fixed, 95% CI)

0.76 [0.03, 18.90]

Analysis 11.1

Comparison 11 Vaniprevir versus placebo/no intervention, Outcome 1 Hepatitis C‐related morbidity or all‐cause mortality.

Comparison 11 Vaniprevir versus placebo/no intervention, Outcome 1 Hepatitis C‐related morbidity or all‐cause mortality.

2 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to dose Show forest plot

9

379

Odds Ratio (M‐H, Fixed, 95% CI)

0.76 [0.03, 18.90]

Analysis 11.2

Comparison 11 Vaniprevir versus placebo/no intervention, Outcome 2 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to dose.

Comparison 11 Vaniprevir versus placebo/no intervention, Outcome 2 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to dose.

2.1 Over or equal to median dose

6

313

Odds Ratio (M‐H, Fixed, 95% CI)

0.76 [0.03, 18.90]

2.2 Under median dose

3

66

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.3 Not available

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3 Serious adverse events Show forest plot

10

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 11.3

Comparison 11 Vaniprevir versus placebo/no intervention, Outcome 3 Serious adverse events.

Comparison 11 Vaniprevir versus placebo/no intervention, Outcome 3 Serious adverse events.

4 Serious adverse events ‐ according to median dose Show forest plot

10

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 11.4

Comparison 11 Vaniprevir versus placebo/no intervention, Outcome 4 Serious adverse events ‐ according to median dose.

Comparison 11 Vaniprevir versus placebo/no intervention, Outcome 4 Serious adverse events ‐ according to median dose.

4.1 Over or equal to median dose

6

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.2 Under median dose

4

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.3 Not available

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5 Without sustained virological response Show forest plot

9

333

Odds Ratio (M‐H, Fixed, 95% CI)

0.12 [0.06, 0.22]

Analysis 11.5

Comparison 11 Vaniprevir versus placebo/no intervention, Outcome 5 Without sustained virological response.

Comparison 11 Vaniprevir versus placebo/no intervention, Outcome 5 Without sustained virological response.

6 Without sustained virological response ‐ according to median dose Show forest plot

9

333

Odds Ratio (M‐H, Fixed, 95% CI)

0.12 [0.06, 0.22]

Analysis 11.6

Comparison 11 Vaniprevir versus placebo/no intervention, Outcome 6 Without sustained virological response ‐ according to median dose.

Comparison 11 Vaniprevir versus placebo/no intervention, Outcome 6 Without sustained virological response ‐ according to median dose.

6.1 Over or equal to median dose

6

280

Odds Ratio (M‐H, Fixed, 95% CI)

0.10 [0.05, 0.20]

6.2 Under median dose

3

53

Odds Ratio (M‐H, Fixed, 95% CI)

0.26 [0.06, 1.04]

6.3 Not available

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Open in table viewer
Comparison 12. All DAA versus placebo/no intervention

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Without significant reductions in ALT/AST serum levels Show forest plot

11

2099

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.68, 0.92]

Analysis 12.1

Comparison 12 All DAA versus placebo/no intervention, Outcome 1 Without significant reductions in ALT/AST serum levels.

Comparison 12 All DAA versus placebo/no intervention, Outcome 1 Without significant reductions in ALT/AST serum levels.

2 Without significant reductions in ALT/AST serum levels ‐ according to DAA status Show forest plot

11

2099

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.68, 0.92]

Analysis 12.2

Comparison 12 All DAA versus placebo/no intervention, Outcome 2 Without significant reductions in ALT/AST serum levels ‐ according to DAA status.

Comparison 12 All DAA versus placebo/no intervention, Outcome 2 Without significant reductions in ALT/AST serum levels ‐ according to DAA status.

2.1 Trials assessing DAAs on or on the way to the market

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Trials assessing DAAs withdrawn from market

11

2099

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.68, 0.92]

3 Without significant reductions in ALT/AST serum levels ‐ according to type of drug Show forest plot

11

2099

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.68, 0.92]

Analysis 12.3

Comparison 12 All DAA versus placebo/no intervention, Outcome 3 Without significant reductions in ALT/AST serum levels ‐ according to type of drug.

Comparison 12 All DAA versus placebo/no intervention, Outcome 3 Without significant reductions in ALT/AST serum levels ‐ according to type of drug.

3.1 Faldaprevir

8

2019

Risk Ratio (M‐H, Random, 95% CI)

0.81 [0.69, 0.96]

3.2 Balaparavir

3

80

Risk Ratio (M‐H, Random, 95% CI)

0.61 [0.41, 0.92]

original image
Figuras y tablas -
Figure 1

Study flow diagram
Figuras y tablas -
Figure 2

Study flow diagram

Trial Sequential Analysis of the effects of direct‐acting antivirals on the market or under development versus placebo or no intervention on risk of serious adverse events. The analysis was based on a proportion in the control group (Pc) of 4.5%, a relative risk reduction (RRR) of 20%, and alfa of 2.5%, a beta of 20%, and a diversity of 0%. The cumulative Z‐curve enters the futility area after the randomisation of about 6000 participants.
Figuras y tablas -
Figure 3

Trial Sequential Analysis of the effects of direct‐acting antivirals on the market or under development versus placebo or no intervention on risk of serious adverse events. The analysis was based on a proportion in the control group (Pc) of 4.5%, a relative risk reduction (RRR) of 20%, and alfa of 2.5%, a beta of 20%, and a diversity of 0%. The cumulative Z‐curve enters the futility area after the randomisation of about 6000 participants.

Trial Sequential Analysis of the effects of simeprevir versus placebo or no intervention on risk of serious adverse events. The analysis was based on a proportion in the control group (Pc) of 8.4%, a relative risk reduction (RRR) of 20%, and alfa of 2.5%, a beta of 20%, and a diversity of 0%. The cumulative Z‐curve crosses the naive type I error level of 5%, but it does not cross the trial monitoring boundary for benefit.
Figuras y tablas -
Figure 4

Trial Sequential Analysis of the effects of simeprevir versus placebo or no intervention on risk of serious adverse events. The analysis was based on a proportion in the control group (Pc) of 8.4%, a relative risk reduction (RRR) of 20%, and alfa of 2.5%, a beta of 20%, and a diversity of 0%. The cumulative Z‐curve crosses the naive type I error level of 5%, but it does not cross the trial monitoring boundary for benefit.

Trial Sequential Analysis of the effects of direct‐acting antivirals on the market or under development versus placebo or no intervention on risk of no sustained virological response. The analysis was based on a proportion in the control group (Pc) of 60.2%, a RRR of 20%, and alfa of 2.5%, a beta of 20%, and a diversity of 83%. After randomisation of about 1000 participants, the cumulative Z‐curve crosses the trial sequential monitoring boundary for benefit.
Figuras y tablas -
Figure 5

Trial Sequential Analysis of the effects of direct‐acting antivirals on the market or under development versus placebo or no intervention on risk of no sustained virological response. The analysis was based on a proportion in the control group (Pc) of 60.2%, a RRR of 20%, and alfa of 2.5%, a beta of 20%, and a diversity of 83%. After randomisation of about 1000 participants, the cumulative Z‐curve crosses the trial sequential monitoring boundary for benefit.

Trial Sequential Analysis of the effects of withdrawn direct‐acting antivirals versus placebo or no intervention on risk of serious adverse events. The analysis was based on a proportion in the control group (Pc) of 7.5%, a RRR of 20%, and alfa of 2.5%, a beta of 20%, and a diversity of 0%. After randomisation of about 5000 participants, the cumulative Z‐curve crosses the trial sequential monitoring boundary for harm.
Figuras y tablas -
Figure 6

Trial Sequential Analysis of the effects of withdrawn direct‐acting antivirals versus placebo or no intervention on risk of serious adverse events. The analysis was based on a proportion in the control group (Pc) of 7.5%, a RRR of 20%, and alfa of 2.5%, a beta of 20%, and a diversity of 0%. After randomisation of about 5000 participants, the cumulative Z‐curve crosses the trial sequential monitoring boundary for harm.

Funnel plot of comparison: 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), outcome: 3.1 Without sustained virological response.
Figuras y tablas -
Figure 7

Funnel plot of comparison: 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), outcome: 3.1 Without sustained virological response.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 1 Hepatitis C‐related morbidity or all‐cause mortality.
Figuras y tablas -
Analysis 1.1

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 1 Hepatitis C‐related morbidity or all‐cause mortality.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 2 Hepatitis C‐related morbidity or all‐cause mortality ‐ bias risk.
Figuras y tablas -
Analysis 1.2

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 2 Hepatitis C‐related morbidity or all‐cause mortality ‐ bias risk.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 3 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to type of DAA.
Figuras y tablas -
Analysis 1.3

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 3 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to type of DAA.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 4 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to group of DAA.
Figuras y tablas -
Analysis 1.4

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 4 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to group of DAA.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 5 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to HIV‐infection.
Figuras y tablas -
Analysis 1.5

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 5 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to HIV‐infection.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 6 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to comorbidity.
Figuras y tablas -
Analysis 1.6

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 6 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to comorbidity.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 7 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to viral genotype.
Figuras y tablas -
Analysis 1.7

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 7 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to viral genotype.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 8 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to human genotype (IL28b).
Figuras y tablas -
Analysis 1.8

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 8 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to human genotype (IL28b).

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 9 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to Asian‐region.
Figuras y tablas -
Analysis 1.9

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 9 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to Asian‐region.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 10 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to specific ethnicities.
Figuras y tablas -
Analysis 1.10

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 10 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to specific ethnicities.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 11 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to reaching planned sample size.
Figuras y tablas -
Analysis 1.11

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 11 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to reaching planned sample size.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 12 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to prior treatment.
Figuras y tablas -
Analysis 1.12

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 12 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to prior treatment.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 13 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to interferon.
Figuras y tablas -
Analysis 1.13

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 13 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to interferon.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 14 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to ribavirin.
Figuras y tablas -
Analysis 1.14

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 14 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to ribavirin.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 15 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to chronic kidney disease.
Figuras y tablas -
Analysis 1.15

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 15 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to chronic kidney disease.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 16 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to cryoglobulinaemia.
Figuras y tablas -
Analysis 1.16

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 16 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to cryoglobulinaemia.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 17 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to DAA group as co‐intervention.
Figuras y tablas -
Analysis 1.17

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 17 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to DAA group as co‐intervention.

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 18 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to median dose.
Figuras y tablas -
Analysis 1.18

Comparison 1 DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses), Outcome 18 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to median dose.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 1 Serious adverse events.
Figuras y tablas -
Analysis 2.1

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 1 Serious adverse events.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 2 Serious adverse events ‐ bias risk.
Figuras y tablas -
Analysis 2.2

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 2 Serious adverse events ‐ bias risk.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 3 Serious adverse events ‐ according to type of DAA.
Figuras y tablas -
Analysis 2.3

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 3 Serious adverse events ‐ according to type of DAA.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 4 Serious adverse events ‐ according to group of DAA.
Figuras y tablas -
Analysis 2.4

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 4 Serious adverse events ‐ according to group of DAA.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 5 Serious adverse events ‐ according to HIV‐infection.
Figuras y tablas -
Analysis 2.5

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 5 Serious adverse events ‐ according to HIV‐infection.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 6 Serious adverse events ‐ according to comorbidity.
Figuras y tablas -
Analysis 2.6

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 6 Serious adverse events ‐ according to comorbidity.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 7 Serious adverse events ‐ according to viral genotype.
Figuras y tablas -
Analysis 2.7

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 7 Serious adverse events ‐ according to viral genotype.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 8 Serious adverse events ‐ according to human genotype (IL28b).
Figuras y tablas -
Analysis 2.8

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 8 Serious adverse events ‐ according to human genotype (IL28b).

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 9 Serious adverse events ‐ according to Asian‐region.
Figuras y tablas -
Analysis 2.9

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 9 Serious adverse events ‐ according to Asian‐region.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 10 Serious adverse events ‐ according to specific ethnicities.
Figuras y tablas -
Analysis 2.10

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 10 Serious adverse events ‐ according to specific ethnicities.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 11 Serious adverse events ‐ according to reaching planned sample size.
Figuras y tablas -
Analysis 2.11

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 11 Serious adverse events ‐ according to reaching planned sample size.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 12 Serious adverse events ‐ according to prior treatment.
Figuras y tablas -
Analysis 2.12

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 12 Serious adverse events ‐ according to prior treatment.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 13 Serious adverse events ‐ according to interferon.
Figuras y tablas -
Analysis 2.13

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 13 Serious adverse events ‐ according to interferon.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 14 Serious adverse events ‐ according to ribavirin.
Figuras y tablas -
Analysis 2.14

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 14 Serious adverse events ‐ according to ribavirin.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 15 Serious adverse events ‐ according to chronic kidney disease.
Figuras y tablas -
Analysis 2.15

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 15 Serious adverse events ‐ according to chronic kidney disease.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 16 Serious adverse events ‐ according to cryoglobulinaemia.
Figuras y tablas -
Analysis 2.16

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 16 Serious adverse events ‐ according to cryoglobulinaemia.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 17 Serious adverse events ‐ according to DAA group as co‐intervention.
Figuras y tablas -
Analysis 2.17

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 17 Serious adverse events ‐ according to DAA group as co‐intervention.

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 18 Serious adverse events ‐ according to median dose.
Figuras y tablas -
Analysis 2.18

Comparison 2 DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses), Outcome 18 Serious adverse events ‐ according to median dose.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 1 Without sustained virological response.
Figuras y tablas -
Analysis 3.1

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 1 Without sustained virological response.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 2 Without sustained virological response ‐ bias risk.
Figuras y tablas -
Analysis 3.2

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 2 Without sustained virological response ‐ bias risk.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 3 Without sustained virological response ‐ according to type of DAA.
Figuras y tablas -
Analysis 3.3

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 3 Without sustained virological response ‐ according to type of DAA.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 4 Without sustained virological response ‐ according to group of DAA.
Figuras y tablas -
Analysis 3.4

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 4 Without sustained virological response ‐ according to group of DAA.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 5 Without sustained virological response ‐ according to HIV‐infection.
Figuras y tablas -
Analysis 3.5

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 5 Without sustained virological response ‐ according to HIV‐infection.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 6 Without sustained virological response ‐ according to comorbidity.
Figuras y tablas -
Analysis 3.6

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 6 Without sustained virological response ‐ according to comorbidity.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 7 Without sustained virological response ‐ according to viral genotype.
Figuras y tablas -
Analysis 3.7

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 7 Without sustained virological response ‐ according to viral genotype.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 8 Without sustained virological response ‐ according to human genotype (IL28b).
Figuras y tablas -
Analysis 3.8

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 8 Without sustained virological response ‐ according to human genotype (IL28b).

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 9 Without sustained virological response ‐ according to Asian‐region.
Figuras y tablas -
Analysis 3.9

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 9 Without sustained virological response ‐ according to Asian‐region.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 10 Without sustained virological response ‐ according to specific ethnicities.
Figuras y tablas -
Analysis 3.10

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 10 Without sustained virological response ‐ according to specific ethnicities.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 11 Without sustained virological response ‐ according to reaching planned sample size.
Figuras y tablas -
Analysis 3.11

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 11 Without sustained virological response ‐ according to reaching planned sample size.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 12 Without sustained virological response ‐ according to prior treatment.
Figuras y tablas -
Analysis 3.12

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 12 Without sustained virological response ‐ according to prior treatment.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 13 Without sustained virological response ‐ according to interferon.
Figuras y tablas -
Analysis 3.13

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 13 Without sustained virological response ‐ according to interferon.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 14 Without sustained virological response ‐ according to ribavirin.
Figuras y tablas -
Analysis 3.14

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 14 Without sustained virological response ‐ according to ribavirin.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 15 Without sustained virological response ‐ according to chronic kidney disease.
Figuras y tablas -
Analysis 3.15

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 15 Without sustained virological response ‐ according to chronic kidney disease.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 16 Without sustained virological response ‐ according to cryoglobulinaemia.
Figuras y tablas -
Analysis 3.16

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 16 Without sustained virological response ‐ according to cryoglobulinaemia.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 17 Without sustained virological response ‐ according to DAA group as co‐intervention.
Figuras y tablas -
Analysis 3.17

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 17 Without sustained virological response ‐ according to DAA group as co‐intervention.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 18 Without sustained virological response ‐ 'Best‐worst case' scenario.
Figuras y tablas -
Analysis 3.18

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 18 Without sustained virological response ‐ 'Best‐worst case' scenario.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 19 Without sustained virological response ‐ 'Worst‐best case' scenario.
Figuras y tablas -
Analysis 3.19

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 19 Without sustained virological response ‐ 'Worst‐best case' scenario.

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 20 Without sustained virological response ‐ according to median dose.
Figuras y tablas -
Analysis 3.20

Comparison 3 DAA on or on the way to the market versus placebo/no intervention (sustained virological response), Outcome 20 Without sustained virological response ‐ according to median dose.

Comparison 4 Danoprevir versus placebo/no intervention, Outcome 1 Hepatitis C‐related morbidity or all‐cause mortality.
Figuras y tablas -
Analysis 4.1

Comparison 4 Danoprevir versus placebo/no intervention, Outcome 1 Hepatitis C‐related morbidity or all‐cause mortality.

Comparison 4 Danoprevir versus placebo/no intervention, Outcome 2 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to dose.
Figuras y tablas -
Analysis 4.2

Comparison 4 Danoprevir versus placebo/no intervention, Outcome 2 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to dose.

Comparison 4 Danoprevir versus placebo/no intervention, Outcome 3 Serious adverse events.
Figuras y tablas -
Analysis 4.3

Comparison 4 Danoprevir versus placebo/no intervention, Outcome 3 Serious adverse events.

Comparison 4 Danoprevir versus placebo/no intervention, Outcome 4 Serious adverse events ‐ according to median dose.
Figuras y tablas -
Analysis 4.4

Comparison 4 Danoprevir versus placebo/no intervention, Outcome 4 Serious adverse events ‐ according to median dose.

Comparison 4 Danoprevir versus placebo/no intervention, Outcome 5 Without sustained virological response.
Figuras y tablas -
Analysis 4.5

Comparison 4 Danoprevir versus placebo/no intervention, Outcome 5 Without sustained virological response.

Comparison 4 Danoprevir versus placebo/no intervention, Outcome 6 Without sustained virological response ‐ according to median dose.
Figuras y tablas -
Analysis 4.6

Comparison 4 Danoprevir versus placebo/no intervention, Outcome 6 Without sustained virological response ‐ according to median dose.

Comparison 5 All DAA versus placebo/no intervention/other medical intervention (morbidity or all‐cause mortality analyses), Outcome 1 Hepatitis C‐related morbidity or all‐cause mortality.
Figuras y tablas -
Analysis 5.1

Comparison 5 All DAA versus placebo/no intervention/other medical intervention (morbidity or all‐cause mortality analyses), Outcome 1 Hepatitis C‐related morbidity or all‐cause mortality.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 1 Serious adverse events.
Figuras y tablas -
Analysis 6.1

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 1 Serious adverse events.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 2 Serious adverse events ‐ bias risk.
Figuras y tablas -
Analysis 6.2

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 2 Serious adverse events ‐ bias risk.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 3 Serious adverse events ‐ according to type of DAA.
Figuras y tablas -
Analysis 6.3

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 3 Serious adverse events ‐ according to type of DAA.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 4 Serious adverse events ‐ according to group of DAA.
Figuras y tablas -
Analysis 6.4

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 4 Serious adverse events ‐ according to group of DAA.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 5 Serious adverse events ‐ according to HIV‐infection.
Figuras y tablas -
Analysis 6.5

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 5 Serious adverse events ‐ according to HIV‐infection.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 6 Serious adverse events ‐ according to comorbidity.
Figuras y tablas -
Analysis 6.6

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 6 Serious adverse events ‐ according to comorbidity.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 7 Serious adverse events ‐ according to viral genotype.
Figuras y tablas -
Analysis 6.7

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 7 Serious adverse events ‐ according to viral genotype.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 8 Serious adverse events ‐ according to human genotype (IL28b).
Figuras y tablas -
Analysis 6.8

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 8 Serious adverse events ‐ according to human genotype (IL28b).

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 9 Serious adverse events ‐ according to Asian‐region.
Figuras y tablas -
Analysis 6.9

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 9 Serious adverse events ‐ according to Asian‐region.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 10 Serious adverse events ‐ according to specific ethnicities.
Figuras y tablas -
Analysis 6.10

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 10 Serious adverse events ‐ according to specific ethnicities.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 12 Serious adverse events ‐ according to prior treatment.
Figuras y tablas -
Analysis 6.12

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 12 Serious adverse events ‐ according to prior treatment.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 13 Serious adverse events ‐ according to interferon.
Figuras y tablas -
Analysis 6.13

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 13 Serious adverse events ‐ according to interferon.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 14 Serious adverse events ‐ according to ribavirin.
Figuras y tablas -
Analysis 6.14

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 14 Serious adverse events ‐ according to ribavirin.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 15 Serious adverse events ‐ according to chronic kidney disease.
Figuras y tablas -
Analysis 6.15

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 15 Serious adverse events ‐ according to chronic kidney disease.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 16 Serious adverse events ‐ according to cryoglobulinaemia.
Figuras y tablas -
Analysis 6.16

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 16 Serious adverse events ‐ according to cryoglobulinaemia.

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 17 Serious adverse events ‐ according to DAA group as co‐intervention.
Figuras y tablas -
Analysis 6.17

Comparison 6 All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses), Outcome 17 Serious adverse events ‐ according to DAA group as co‐intervention.

Comparison 7 All DAA versus placebo/no intervention/other medical intervention (sustained virological response analyses), Outcome 1 Without sustained virological response.
Figuras y tablas -
Analysis 7.1

Comparison 7 All DAA versus placebo/no intervention/other medical intervention (sustained virological response analyses), Outcome 1 Without sustained virological response.

Comparison 8 All DAA versus placebo/no intervention/other medical intervention (quality of life scores), Outcome 1 SF‐36 physical score.
Figuras y tablas -
Analysis 8.1

Comparison 8 All DAA versus placebo/no intervention/other medical intervention (quality of life scores), Outcome 1 SF‐36 physical score.

Comparison 8 All DAA versus placebo/no intervention/other medical intervention (quality of life scores), Outcome 2 SF‐36 mental score.
Figuras y tablas -
Analysis 8.2

Comparison 8 All DAA versus placebo/no intervention/other medical intervention (quality of life scores), Outcome 2 SF‐36 mental score.

Comparison 9 Daclatasvir versus placebo/no intervention, Outcome 1 Hepatitis C‐related morbidity or all‐cause mortality.
Figuras y tablas -
Analysis 9.1

Comparison 9 Daclatasvir versus placebo/no intervention, Outcome 1 Hepatitis C‐related morbidity or all‐cause mortality.

Comparison 9 Daclatasvir versus placebo/no intervention, Outcome 2 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to dose.
Figuras y tablas -
Analysis 9.2

Comparison 9 Daclatasvir versus placebo/no intervention, Outcome 2 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to dose.

Comparison 9 Daclatasvir versus placebo/no intervention, Outcome 3 Serious adverse events.
Figuras y tablas -
Analysis 9.3

Comparison 9 Daclatasvir versus placebo/no intervention, Outcome 3 Serious adverse events.

Comparison 9 Daclatasvir versus placebo/no intervention, Outcome 4 Serious adverse events ‐ according to median dose.
Figuras y tablas -
Analysis 9.4

Comparison 9 Daclatasvir versus placebo/no intervention, Outcome 4 Serious adverse events ‐ according to median dose.

Comparison 9 Daclatasvir versus placebo/no intervention, Outcome 5 Without sustained virological response.
Figuras y tablas -
Analysis 9.5

Comparison 9 Daclatasvir versus placebo/no intervention, Outcome 5 Without sustained virological response.

Comparison 9 Daclatasvir versus placebo/no intervention, Outcome 6 Without sustained virological response ‐ according to median dose.
Figuras y tablas -
Analysis 9.6

Comparison 9 Daclatasvir versus placebo/no intervention, Outcome 6 Without sustained virological response ‐ according to median dose.

Comparison 10 Simeprevir versus placebo/no intervention, Outcome 1 Hepatitis C‐related morbidity or all‐cause mortality.
Figuras y tablas -
Analysis 10.1

Comparison 10 Simeprevir versus placebo/no intervention, Outcome 1 Hepatitis C‐related morbidity or all‐cause mortality.

Comparison 10 Simeprevir versus placebo/no intervention, Outcome 2 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to dose.
Figuras y tablas -
Analysis 10.2

Comparison 10 Simeprevir versus placebo/no intervention, Outcome 2 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to dose.

Comparison 10 Simeprevir versus placebo/no intervention, Outcome 3 Serious adverse events.
Figuras y tablas -
Analysis 10.3

Comparison 10 Simeprevir versus placebo/no intervention, Outcome 3 Serious adverse events.

Comparison 10 Simeprevir versus placebo/no intervention, Outcome 4 Serious adverse events ‐ according to median dose.
Figuras y tablas -
Analysis 10.4

Comparison 10 Simeprevir versus placebo/no intervention, Outcome 4 Serious adverse events ‐ according to median dose.

Comparison 10 Simeprevir versus placebo/no intervention, Outcome 5 Without sustained virological response.
Figuras y tablas -
Analysis 10.5

Comparison 10 Simeprevir versus placebo/no intervention, Outcome 5 Without sustained virological response.

Comparison 10 Simeprevir versus placebo/no intervention, Outcome 6 Without sustained virological response ‐ according to median dose.
Figuras y tablas -
Analysis 10.6

Comparison 10 Simeprevir versus placebo/no intervention, Outcome 6 Without sustained virological response ‐ according to median dose.

Comparison 11 Vaniprevir versus placebo/no intervention, Outcome 1 Hepatitis C‐related morbidity or all‐cause mortality.
Figuras y tablas -
Analysis 11.1

Comparison 11 Vaniprevir versus placebo/no intervention, Outcome 1 Hepatitis C‐related morbidity or all‐cause mortality.

Comparison 11 Vaniprevir versus placebo/no intervention, Outcome 2 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to dose.
Figuras y tablas -
Analysis 11.2

Comparison 11 Vaniprevir versus placebo/no intervention, Outcome 2 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to dose.

Comparison 11 Vaniprevir versus placebo/no intervention, Outcome 3 Serious adverse events.
Figuras y tablas -
Analysis 11.3

Comparison 11 Vaniprevir versus placebo/no intervention, Outcome 3 Serious adverse events.

Comparison 11 Vaniprevir versus placebo/no intervention, Outcome 4 Serious adverse events ‐ according to median dose.
Figuras y tablas -
Analysis 11.4

Comparison 11 Vaniprevir versus placebo/no intervention, Outcome 4 Serious adverse events ‐ according to median dose.

Comparison 11 Vaniprevir versus placebo/no intervention, Outcome 5 Without sustained virological response.
Figuras y tablas -
Analysis 11.5

Comparison 11 Vaniprevir versus placebo/no intervention, Outcome 5 Without sustained virological response.

Comparison 11 Vaniprevir versus placebo/no intervention, Outcome 6 Without sustained virological response ‐ according to median dose.
Figuras y tablas -
Analysis 11.6

Comparison 11 Vaniprevir versus placebo/no intervention, Outcome 6 Without sustained virological response ‐ according to median dose.

Comparison 12 All DAA versus placebo/no intervention, Outcome 1 Without significant reductions in ALT/AST serum levels.
Figuras y tablas -
Analysis 12.1

Comparison 12 All DAA versus placebo/no intervention, Outcome 1 Without significant reductions in ALT/AST serum levels.

Comparison 12 All DAA versus placebo/no intervention, Outcome 2 Without significant reductions in ALT/AST serum levels ‐ according to DAA status.
Figuras y tablas -
Analysis 12.2

Comparison 12 All DAA versus placebo/no intervention, Outcome 2 Without significant reductions in ALT/AST serum levels ‐ according to DAA status.

Comparison 12 All DAA versus placebo/no intervention, Outcome 3 Without significant reductions in ALT/AST serum levels ‐ according to type of drug.
Figuras y tablas -
Analysis 12.3

Comparison 12 All DAA versus placebo/no intervention, Outcome 3 Without significant reductions in ALT/AST serum levels ‐ according to type of drug.

Summary of findings for the main comparison. Direct‐acting antivirals versus control

Direct‐acting antivirals versus control

Patient or population: adults with chronic hepatitis C
Setting: any setting
Intervention: direct‐acting antivirals on the market or under development
Comparison: placebo or no intervention

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

(TSA‐adjusted CI)

№ of participants
(trials)

Quality of the evidence
(GRADE)

Comments

Risk with placebo or no intervention

Risk with direct‐acting antivirals

All‐cause mortality at maximum follow‐up

2 per 1000

7 per 1000
(1 to 42)

OR 3.72
(0.53 to 26.18)

(‐)

2996
(11 RCTs)

⊕⊝⊝⊝
Very low1

It was not possible to perform Trial Sequential Analysis because of limited data and too few events

Proportion of participants with one or more serious adverse event at maximum follow‐up

56 per 1000

52 per 1000
(49 to 55)

OR 0.93
(0.75 to 1.15)

(TSA CI 0.71 to 1.33)

15,817
(43 RCTs)

⊕⊝⊝⊝
Very low2

Trial Sequential Analysis showed that the boundary for futility was crossed. This leads us to conclude that any possible intervention effect, if any, is less than 20%

Proportion of participants with no sustained virological response at maximum follow‐up

541 per 1000

238 per 1000
(200 to 281)

RR 0.44
(0.37 to 0.52)

(TSA CI 0.42 to 0.55)

6886
(32 RCTs)

⊕⊕⊝⊝

Low3

Trial Sequential Analysis showed that the boundary for benefit was crossed. This indicates that DAAs seem to decrease the risk of no sustained virological response by at least 20% if risk of bias and other threats to the validity can be disregarded

*The risk in the intervention group (and its 95% confidence interval) is based on the observed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; DAA: direct‐acting antivirals; OR: odds ratio; RCTs: randomised clinical trials; RR: risk ratio; TSA: Trial Sequential Analysis

GRADE Working Group grades of evidence
High quality: we are very confident that the true effect lies close to that of the estimate of the effect
Moderate quality: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low quality: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect
Very low quality: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect

1Downgraded two levels because of very serious risk of bias in the included trials (see Figure 1) and two levels due to very serious imprecision (none of the TSA boundaries are crossed, so the information size is too low).
2Downgraded two levels due to very serious risk of bias in the included trials (see Figure 1) and one level due to serious indirectness (the components of this composite outcome consisted of events with very different degrees of severity, which limits the interpretability of this outcome result).
3Downgraded two levels because of very serious risk of bias in the included trials (Figure 1).

Figuras y tablas -
Summary of findings for the main comparison. Direct‐acting antivirals versus control
Summary of findings 2. Direct‐acting antivirals withdrawn from the market versus control

Direct‐acting antivirals withdrawn from the market versus control

Patient or population: adults with chronic hepatitis C
Setting: any setting
Intervention: direct‐acting antivirals withdrawn from the market
Comparison: placebo or no intervention

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

(TSA‐adjusted CI)

№ of participants
(trials)

Quality of the evidence
(GRADE)

Comments

Risk with placebo or no intervention

Risk with direct‐acting antivirals

All‐cause mortality

at maximum follow‐up

7 per 1000

5 per 1000
(2 to 12)

OR 0.64
(0.23 to 1.79)

(‐)

3045
(5 RCTs)

⊕⊝⊝⊝
Very low1

It was not possible to perform Trial Sequential Analysis because of limited data and too few events

Proportion of participants with one or more serious adverse event at maximum follow‐up

75 per 1000

108 per 1000
(91 to 129)

OR 1.45
(1.22 to 1.73)

(TSA 1.16 to 1.82)

9229
(29 RCTs)

⊕⊝⊝⊝
Very low2

Trial Sequential Analysis showed that the boundary for harm was crossed. This shows that there is firm evidence that withdrawn DAAs increase the risk of a serious adverse event by at least 20%

Proportion of participants with no sustained virological response at maximum follow‐up

586 per 1000

356 per 1000
(322 to 404)

RR 0.61 (0.55, 0.69)

(TSA CI 0.42 to 0.55)

9075
(21 RCTs)

⊕⊕⊝⊝

Low3

Trial Sequential Analysis not performed

*The risk in the intervention group (and its 95% confidence interval) is based on the observed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; DAA: direct‐acting antivirals; OR: odds ratio; RCTs: randomised clinical trials; RR: risk ratio; TSA: Trial Sequential Analysis

GRADE Working Group grades of evidence
High quality: we are very confident that the true effect lies close to that of the estimate of the effect
Moderate quality: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low quality: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect
Very low quality: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect

1Downgraded two levels because of very serious risk of bias in the included trials (see Figure 1) and two levels due to very serious imprecision (none of the TSA boundaries are crossed so the information size is too low).
2Downgraded two levels because of very serious risk of bias in the included trials (see Figure 1) and one level due to serious indirectness (the components of this composite outcome consisted of events with very different degrees of severity which limits the interpretability of this outcome result).
3Downgraded two levels because of very serious risk of bias in the included trials (Figure 1).

Figuras y tablas -
Summary of findings 2. Direct‐acting antivirals withdrawn from the market versus control
Table 1. List of direct‐acting antivirals

Direct‐acting antiviral agents (DAAs)

NS3/NS4A inhibitors

NS5B inhibitors

NS5A inhibitors

NPI

NNPI

ACH‐2684

ALS2200/VX135

ABT‐072

ACH‐2928

Asunaprevir

BILB1941

Beclabuvir

Daclatasvir

Boceprevir

GS0938/PSI352938

BI201127

Elbasvir

Celuprevir

GS6620

Dasabuvir

GSK2336805

Danoprevir

GS9851(PSI7851)

Deleobuvir

Ledipasvir

Faldaprevir

IDX184

Filibuvir

MK‐8408

Grazoprevir

INX189/BMS986094

GSK2878175/GSK175

Odalasvir

GS9256

Mericitabine

IDX375

Ombitasvir

GS9857

MK‐3682

MK‐3281

PPI461

IDX320

Sofosbuvir

Nesbuvir

Ravidasvir

Narlaprevir

VX‐135

Radalbuvir

Samatasvir

Paritaprevir

Setrobuvir

Velpatasvir

PHX1766

Tegobuvir

Simperevir

TMC‐647055

Sovaprevir

VCH‐759

Telaprevir

VCH‐916

Vaniprevir

VX222

Vedroprevir

The table presents a list of 58 direct‐acting antiviral agents (DAAs). We have listed the DAAs according to the DAA class they belong to (see Background section). When a DAA has not been assigned a generic or brand name, we have presented it with its experimental compound number prefix.

Figuras y tablas -
Table 1. List of direct‐acting antivirals
Table 2. Serious adverse events

Trial

Experimental intervention

Type and number of serious adverse events (experimental group)

Proportion of participants with a serious adverse event (experimental group)

Type and number of serious adverse events (control group)

Proportion of participants with a serious adverse event (control group)

Bronowicki 2013a1

Asunaprevir

1 abdominal pain, 1 lung neoplasm malignant, 1 cytolytic hepatitis, and 2 unspecified events

5 out of 36

None reported

0 out of 11

Bronowicki 2014

Asunaprevir

2 deaths and 14 unspecified events

16 out of 177

3 unspecified events

3 out of 61

Nelson 2012a1

Balapiravir

Many events but only a few were specified: 3 deaths, 10 haematological, 10 infection, 8 eye disorders

49 out of 432

Many events but not all were specified: 2 infections, 1 death

9 out of 72

Tatum 2015a1

Beclabuvir

1 anaemia, 1 constipation, 1 febrile neutropenia, 1 leukopenia

1 out of 26

1 serotonin syndrome

1 out of 13

Bacon 2011a1

Boceprevir

5 anaemia, 1 angina pectoris, 1 atrial fibrillation, 1 coronary artery disease, 1 myocardial infarction, 1 myopericarditis, 2 abdominal pain, 1 constipation, 1 diarrhoea, 1 gastritis, 1 irritable bowel syndrome, 1 oesophageal varices haemorrhage, 1 pancreatitis acute, 1 pancreatitis necrotising, 1 peptic ulcer, 1 asthenia, 3 chest pain, 1 oedema peripheral, 1 pyrexia, 1 cholecystitis, 3 appendicitis, 1 bronchopneumonia, 1 catheter site infection, 1 gastroenteritis viral, 1 pneumonia, 1 lower limb fracture, 1 overdose, 1 decreased appetite, 1 dehydration, 1 hyperglycaemia, 1 back pain, 2 intervertebral disc protrusion, 1 pain in extremity, 1 hepatic neoplasm malignant, 1 hepatic encephalopathy, 1 sciatica, 1 syncope, 1 bipolar disorder, 1 completed suicide, 4 depression, 2 homicidal ideation, 5 suicidal ideation, 2 dyspnoea, 1 pleuritic pain, 1 pneumothorax, 1 abdominal hernia repair, 1 deep vein thrombosis, 1 phlebitis

39 out of 323

2 chest pain, 1 cholelithiasis, 1 gastroenteritis

4 out of 80

Flamm 2013

Boceprevir

1 coronary artery disease, 1 diarrhoea, 1 asthenia, 1 pyrexia, 2 pneumonia, 2 syncope, 1 suicidal ideation, 1 deep vein thrombosis, 1 neutropenia, 1 thrombocytopenia, 1 cardiac failure, 1 upper gastrointestinal haemorrhage , 1 multi‐organ failure, 1 bronchitis, 1 cellulitis, 1 chlamydia infection, 1 influenza, 1 pneumonia staphylococcal, 1 staphylococcal bacteraemia, 1 staphylococcal infection, 1 urosepsis, 1 gun shot wound, 2 hyponatraemia, 1 lethargy, 1 subarachnoid haemorrhage, 1 mental status changes

18 out of 134

1 chest pain, 1 intervertebral disc protrusion, 1 abnormal behaviour, 1 irritability, 1 osteotomy, 1 foreign body, 1 neuralgia, 1 anxiety, 1 renal colic

7 out of 67

Isakov 2016

Boceprevir

14 neutropenia, 1 intestinal obstruction, 1 osteomyelitis chronic, 1 pneumonia, 1 diabetic ketoacidosis, 1 intervertebral disc protrusion, 1 transient ischaemic attack

17 out of 159

4 neutropenia, 1 general disorders, 1 accidental overdose, 1 prostatitis, 2 hypertension

9 out of 78

Kwo 2010a1

Boceprevir

1 anaemia, 1 abdominal pain, 2 asthenia, 2 pyrexia, 2 pneumonia, 1 decreased appetite, 1 dehydration, 2 depression, 2 homicidal ideation, 3 suicidal ideation, 1 dyspnoea, 1 deep vein thrombosis, 3 nausea, 1 vomiting, 3 neutropenia, 1 multi‐organ failure, 2 cellulitis, 2 abdominal pain upper, 1 headache, 1 suicide attempt, 1 accidental overdose, 1 fall, 1 pulmonary embolism, 1 gastroenteritis, 1 erysipelas, 1 panic attack, 1 fatigue, 1 supraventricular tachycardia, 3 pancreatitis, 1 cerebrovascular accident, 1 hypoaesthesia, 1 anxiety, 1 retinal ischaemia, 1 neuropathy peripheral, 1 aggression, 1 scotoma, 1 hypovolaemia, 1 vulval abscess, 1 retinopathy, 1 inguinal hernia, 1 cervix carcinoma, 1 pericarditis, 1 paranoia, 1 neutrophil count decreased, 1 paraesthesia, 1 peritoneal haemorrhage, 1 deafness unilateral, 1 periodontal disease, 1 corneal infection, 1 pneumonia streptococcal, 1 drug toxicity, 1 blood amylase increased, 1 lipase increased, 1 basal cell carcinoma, 1 renal cell carcinoma

40 out of 527

1 suicidal ideation, 1 breast cancer, 1 parathyroid tumour benign, 1 muscle spasms, 1 rib fracture, 1 contusion, 1 inguinal hernia, 1 diplopia, 1 staphylococcal sepsis, 1 animal bite, 1 hand fracture, 1 third nerve paralysis, 1 alcoholism, 1 dependence

8 out of 104

Pearlman 2014

Boceprevir

1 anaemia

1 out of 49

1 anaemia

1 out of 52

Poordad 2011a1

Boceprevir

7 anaemia, 1 atrial fibrillation, 1 coronary artery disease, 2 abdominal pain, 1 gastritis, 1 pancreatitis acute, 5 chest pain, 4 pyrexia, 1 cholecystitis, 4 pneumonia, 1 overdose, 1 dehydration, 1 back pain, 1 intervertebral disc protrusion, 5 syncope, 1 completed suicide, 2 depression, 4 suicidal ideation, 1 dyspnoea, 1 nausea, 2 vomiting, 3 neutropenia, 3 thrombocytopenia, 2 bronchitis, 3 cellulitis, 1 staphylococcal infection, 1 hyponatraemia, 1 pancytopenia, 1 breast cancer, 1 malaise, 1 pneumonia pneumococcal, 1 haemoptysis, 1 road traffic accident, 1 suicide attempt, 1 pruritus, 1 rash erythematous, 1 dizziness, 2 pulmonary embolism, 1 haemorrhoids, 4 gastroenteritis, 1 general physical health deterioration, 1 hypertensive crisis, 1 colon cancer, 1 drug abuse, 2 hypokalaemia, 2 chest discomfort, 1 fatigue, 1 perirectal abscess, 1 acute myocardial infarction, 1 gastrointestinal haemorrhage, 1 aplasia pure red cell, 2 leukopenia, 1 atrial flutter, 1 cardiac arrest, 1 hypertrophic cardiomyopathy, 1 tachycardia, 1 deafness, 1 conjunctivitis, 1 optic neuropathy, 1 papilledema, 1 abdominal pain lower, 1 colonic polyp, 1 gastroesophageal reflux disease, 1 hematemesis, 1 haemorrhoidal haemorrhage, 1 Mallory‐weiss syndrome, 1 umbilical hernia, 1 sarcoidosis, 1 abscess, 1 abscess limb, 1 bacteraemia, 1 epiglottitis, 1 infected bites, 1 injection site infection, 1 scrotal abscess, 1 tracheobronchitis, 1 post procedural complication, 1 transfusion reaction, 1 vascular pseudoaneurysm, 1 wound dehiscence, 1 flank pain, 1 groin pain, 1 musculoskeletal chest pain, 1 bladder cancer, 1 pancreatic carcinoma, 1 prostate cancer, 1 carotid artery stenosis, 1 cerebral ischaemia, 1 motor neurone disease, 1 muscle spasticity, 1 affective disorder, 1 alcohol abuse, 1 anxiety, 1 psychiatric decompensation, 1 scrotal pain, 2 cough, 1 pleural fibrosis, 1 alcohol use, 1 laryngeal operation, 1 accelerated hypertension, 1 arterial thrombosis limb, 2 hypotension

87 out of 734

1 anaemia, 1 myocardial infarction, 1 abdominal pain, 2 pyrexia, 1 cholecystitis, 1 appendicitis, 1 pneumonia, 1 hepatic neoplasm malignant, 1 completed suicide, 1 depression, 1 suicidal ideation, 1 pneumothorax, 2 cholelithiasis, 1 nausea, 1 vomiting, 1 cellulitis, 1 breast cancer, 1 colitis, 1 upper respiratory tract infection, 1 suicide attempt, 2 death, 1 accidental overdose, 1 dizziness, 1 loss of consciousness, 1 cholecystitis acute, 1 sinusitis, 2 pancreatitis, 1 leukocytosis, 1 cardiac arrest, 1 cardio‐respiratory arrest, 1 hypothyroidism, 1 cholelithiasis obstructive, 1 atypical mycobacterial infection, 1 diverticulitis, 1 enterocolitis infectious, 1 alcohol poisoning, 1 spinal fracture, 1 white blood cell count decreased, 1 lung adenocarcinoma, 1 prostate cancer, 1 hypoaesthesia, 1 affective disorder, 1 bipolar disorder, 1 drug dependence, 1 intentional self‐injury, 1 personality disorder, 1 glomerulonephritis minimal lesion, 1 renal tubular necrosis, 1 physical assault, 1 cholecystectomy, 1 skin neoplasm excision

31 out of 363

Silva 2013a1

Boceprevir

None reported

0 out of 28

1 atrial fibrillation

1 out of 10

Sulkowski 2013a

Boceprevir

3 anaemia, 2 pneumonia, 1 syncope, 1 depression, 1 deep vein thrombosis, 1 lymphadenopathy, 1 renal failure acute, 2 pulmonary embolism, 1 arthralgia, 1 sinusitis, 1 urinary tract infection, 1 lung infection pseudomonal, 1 pelvic inflammatory disease, 1 pulmonary hypertension, 1 suicide attempt

11 out of 64

2 anaemia, 1 overdose, 1 cholelithiasis, 1 abdominal pain upper, 1 meniscus lesion, 1 pancreatitis, 1 post procedural infection, 1 renal failure, 1 cholecystectomy, 1 vulval abscess, 1 ventricular fibrillation, 1 ligament rupture, 1 lactic acidosis, 1 respiratory failure

7 out of 34

Dore 2015a1

Daclatasvir

1 hepatic neoplasm malignant, 1 rectal ulcer haemorrhage, 1 gastrointestinal inflammation, 1 adhesion, 1 biliary colic, 1 hyperbilirubinaemia, 1 appendiceal abscess, 1 tonsil cancer

6 out of 196

1 abdominal pain upper, 1 epicondylitis, 1 conversion disorder

3 out of 100

COMMAND‐1 2015a1

Daclatasvir

1 anaemia, 1 abdominal pain, 1 gastritis, 1 chest pain, 2 pneumonia, 1 overdose, 1 syncope, 2 depression, 2 suicidal ideation, 1 dyspnoea, 1 bronchitis, 1 peritonitis, 1 rash generalised, 1 febrile neutropenia, 1 aplastic anaemia, 1 auricular perichondritis, 2 gastric ulcer haemorrhage, 1 death, 1 bile duct stone, 1 clostridium difficile, 1 furuncle, 1 carbuncle, 1 oral herpes, 1 accidental overdose, 2 falls, 1 bursitis, 1 rhabdomyolysis, 1 muscle spasms, 1 costochondritis, 1 dizziness, 1 loss of consciousness, 1 adjustment disorder, 1 hypomania, 1 mental disorder, 1 substance‐induced psychotic disorder, 1 schizophrenia, paranoid type

25 out of 317

2 anaemia, 1 atrial fibrillation, 1 pneumonia, 1 pyelonephritis, 1 haemoglobin decreased, 1 epistaxis, 1 electrocardiogram change, 1 neutrophil count decreased, 1 myalgia, 1 aphasia, 1 paraesthesia

6 out of 78

Izumi 2014a1

Daclatasvir

1 pancreatitis acute, 1 back pain

2 out of 34

None reported

0 out of 8

Pol 2012

Daclatasvir

1 anaemia, 1 chest pain, 2 syncope, 1 bronchitis, 1 epistaxis

3 out of 36

None reported

0 out of 12

Dauphine 2015a1

Danoprevir

28 unspecified SAEs and 2 deaths

29 out of 373

1 unspecified SAE

1 out of 44

Forestier 2011a1

Danoprevir

1 benign paroxysmal vertigo

1 out of 40

None reported

0 out of 8

Forestier 2011b

Danoprevir

1 gastroenteritis viral

1 out of 47

None reported

0 out of 12

Gane 2011

Danoprevir

1 altered mood

1 out of 25

None reported

0 out of 5

ATLAS 2013

Danoprevir

14 SAEs but not specified, 1 death

15 out of 194

6 SAEs but not specified

6 out of 31

Larrey 2013

Deleobuvir

1 drug eruption

1 out of 46

None reported

0 out of 14

Larrey 2012

Deleobuvir

1 syncope, 1 rash maculo‐papular, 1 umbilical hernia

3 out of 49

None reported

0 out of 8

STARTverso‐2 2014a1

Faldaprevir

2 anaemia, 1 angina pectoris, 2 diarrhoea, 1 oesophageal varices haemorrhage, 1 cholecystitis, 2 pneumonia, 1 dehydration, 1 back pain, 1 intervertebral disc protrusion, 1 bipolar disorder, 1 depression, 1 suicidal ideation, 1 dyspnoea, 2 nausea, 3 vomiting, 2 neutropenia, 1 thrombocytopenia, 1 cellulitis, 1 mental status changes, 1 pancytopenia, 1 breast cancer, 1 malaise, 2 rash, 2 sepsis, 1 suicide attempt, 1 renal failure acute, 1 rash maculo‐papular, 1 accidental overdose, 1 muscle spasm, 1 tibia fracture, 1 contusion, 1 pulmonary embolism, 2 abortion spontaneous, 1 hypokalaemia, 1 subcutaneous abscess, 1 acute myocardial infarction, 1 pancreatitis, 1 umbilical hernia, 1 diverticulitis, 1 cerebral ischaemia, 1 drug dependence, 1 personality disorder, 1 epidermolysis, 1 ascites, 1 duodenal ulcer haemorrhage, 1 large intestine perforation, 1 hepatic cirrhosis, 2 hepatic failure, 1 hypersensitivity, 1 infective chondritis, 1 vulval abscess, 1 fibula fracture, 1 jaw fracture, 1 ligament sprain, 1 hypocalcaemia, 1 hyponatraemia, 1 hepatocellular carcinoma, 1 papillary thyroid cancer

47 out of 525

1 anaemia, 2 depression, 1 suicidal ideation, 1 bile duct stone, 1 subcutaneous abscess, 1 optic ischaemic neuropathy, 1 laceration, 1 mental status change

8 out of 132

STARTVerso‐1 2015a1

Faldaprevir

3 anaemia, 1 atrial fibrillation, 1 myocardial infarction, 1 asthenia, 1 chest pain, 1 pyrexia, 1 bronchopneumonia, 1 pneumonia, 1 sciatica, 2 vomiting, 1 thrombocytopenia, 1 pancytopenia, 1 headache, 2 rash, 1 drug eruption, 1 dizziness, 1 haemorrhoids, 1 psychotic disorder, 1 urinary tract infection, 1 diabetes mellitus, 1 parapsoriasis, 1 pancreatitis, 1 histiocytosis haematophagic, 1 cerebrovascular accident, 1 muscular weakness, 1 epistaxis, 1 leukopenia, 1 sarcoidosis, 1 hypotension, 1 idiopathic thrombocytopenic purpura, 1 optic ischaemic neuropathy, 1 hypersensitivity, 1 hypoparathyroidism, 1 retinopathy, 1 subdural hematoma, 1 cervix carcinoma, 1 cubital tunnel syndrome, 1 dyspnoea exertional

34 out of 520

1 anaemia, 1 cholecystitis, 1 gun shot wound, 1 rash maculo‐papular, 1 diverticulitis, 1 inguinal hernia, 1 hepatic lesion, 1 polymyositis, 1 blister

8 out of 132

STARTverso‐3 2013a1

Faldaprevir

5 anaemia, 1 atrial fibrillation, 1 abdominal pain, 5 diarrhoea, 1 pancreatitis acute, 1 asthenia, 1 chest pain, 8 pyrexia, 2 appendicitis, 1 gastroenteritis viral, 2 pneumonia, 1 decreased appetite, 1 dehydration, 1 back pain, 1 hepatic neoplasm malignant, 2 cholelithiasis, 1 biliary colic, 2 hyperbilirubinaemia, 3 nausea, 2 vomiting, 1 thrombocytopenia, 1 cellulitis, 1 bradycardia, 2 presyncope, 2 malaise, 2 headache, 2 sepsis, 1 rash erythematous, 1 rash generalised, 1 fall, 1 multiple injuries, 1 haematochezia, 1 peritonitis bacterial, 1 congestive cardiac failure, 1 gastroenteritis, 1 hypertensive crisis, 1 hypokalaemia, 1 fatigue, 1 pancreatitis, 1 coma, 1 renal colic, 1 leukopenia, 1 cardio‐respiratory arrest, 1 anxiety, 1 psychiatric decompensation, 2 hypotension, 1 viral infection, 2 ascites, 1 hepatic failure, 1 hypoglycaemia, 1 haemolytic anaemia, 1 keratosis follicular, 1 oral lichen planus, 1 peritoneal haemorrhage, 1 salivary gland calculus, 1 hepatorenal failure, 2 jaundice, 1 streptococcal infection, 1 blood lactate dehydrogenase increased, 1 international normalised ratio abnormal, 1 metabolic acidosis, 1 fasciitis, 1 joint instability, 1 musculoskeletal discomfort, 1 haemothorax, 1 venous thrombosis

54 out of 599

1 depression, 1 pleural effusion

1 out of 78

Nishiguchi 2014a1

Faldaprevir

1 abdominal pain upper

1 out of 35

1 abdominal pain

1 out of 8

Manns 2011

Faldaprevir

1 asthenia, 1 cataract , 1 hypoalbuminaemia, 1 metabolic disorder, 1 ascites

4 out of 88

None reported

0 out of 8

Sulkowski 2013a

Faldeprevir

4 anaemia, 1 angina pectoris, 1 myocardial infarction, 1 diarrhoea, 1 asthenia, 1 chest pain, 1 oedema peripheral, 4 pyrexia, 1 cholecystitis, 1 pneumonia, 2 dehydration, 1 intervertebral disc protrusion, 2 syncope, 1 depression, 1 nausea, 2 vomiting, 1 thrombocytopenia, 1 upper gastrointestinal haemorrhage, 1 influenza, 1 lower respiratory tract infection, 2 photosensitivity reaction, 1 upper respiratory tract infection, 2 headache, 1 rash, 1 road traffic accident, 2 suicide attempt, 3 drug eruption, 2 rash maculo‐papular, 1 rash erythematous, 2 febrile neutropenia, 1 oral herpes, 1 pulmonary embolism, 1 pyelonephritis, 1 cataract, 1 anaemia haemolytic autoimmune, 1 lymphopenia, 1 microvascular angina, 1 prinzmetal angina, 1 anal fistula, 1 haemorrhoids, 1 mouth ulceration, 1 rectal haemorrhage, 1 chest discomfort, 1 fatigue, 1 mucosal inflammation, 1 gallbladder polyp, 1 cryoglobulinaemia, 1 anal abscess, 1 ear infection, 2 H1N1 influenza, 1 infected skin ulcer, 1 lymphangitis, 1 perirectal abscess, 1 pharyngitis, 1 subcutaneous abscess, 1 superinfection bacterial, 1 urinary tract infection, 1 diabetes mellitus, 1 ischaemic stroke, 1 acute psychosis, 1 depressed mood, 1 calculus ureteric, 1 endometrial hyperplasia, 1 dermatitis atopic, 1 eczema, 1 erythema multiforme, 1 lichen planus, 1 palmar‐plantar erythrodysaesthesia syndrome, 1 parapsoriasis, 1 pruritus allergic, 1 rash pruritic, 1 appendicectomy

61 out of 641

1 headache, 1 photophobia, 1 cyst, 1 benign salivary gland neoplasm, 1 migraine

2 out of 71

Jacobson 2010

Filibuvir

1 blood creatinine increased, 1 chronic obstructive pulmonary disease, 1 pulmonary embolism

3 out of 27

1 thyroiditis, 1 gait disturbance

2 out of 8

Rodriguez‐Torres 2014b1

Filibuvir

1 anaemia, 1 appendicitis, 1 rectal ulcer haemorrhage, 1 craniocerebral injury, 1 vertigo, 1 vestibular disorder, 1 haematochezia, 1 peritonitis bacterial, 1 lymph node tuberculosis, 1 scapula fracture, 1 blood urea nitrogen/creatinine increased, 1 gastric cancer, 1 rectal cancer, 1 abortion spontaneous, 1 cardiac necrosis, 1 pyoderma gangrenosum, 1 depression, 1 breast cancer, 1 chronic obstructive pulmonary disease, 1 lung neoplasm malignant, 1 fall, 1 loss of consciousness, 1 bacterial abscess CNS, 1 actinomyces test positive, 1 pulmonary calcification

20 out of 192

1 neutropenia, 1 sepsis, 1 pulmonary embolism, 1 cerebral haemorrhage, 1 ecchymosis, 1 Appendicitis perforated

6 out of 96

Lawitz 2013b

GS‐9451

1 death, 1 heroin overdose

1 out of 33

None reported

0 out of 8

Gardner 2014a

GSK2336805

1 pneumonia, 1 upper lobe cavitary lesion

1 out of 11

None reported

0 out of 4

Lalezari 2013

IDX‐184

1 pancreatitis, 1 acute cholecystitis

2 out of 65

1 agitation

1 out of 16

Gane 2010

Mericitabine/danoprevir

1 multiple drug overdose, 1 ankle fracture

2 out of 73

None reported

0 out of 14

Feld 2015

Mericitabine

1 nephrolithiasis, 1 porphyria non‐acute

2 out of 102

1 arthritis infective

1 out of 49

JUMP‐C 2013

Mericitabine

6 SAEs but not specified

5 out of 81

4 SAEs but not specified

3 out of 85

De Bruijne 2010a1

Narlaprevir

1 pyrexia, 1 elevated CRP

1 out of 32

None reported

0 out of 8

Muir 2014

Odalasvir (ACH‐3102) and sovaprevir

1 non‐cardiac chest pain

1 out of 20

None reported

0 out of 10

Zeuzem 2014a

Paritaprevir (ABT‐450)/r–ombitasvir

1 pneumonia, 1 nausea, 1 vomiting, 1 bradycardia, 1 chronic obstructive pulmonary disease, 1 renal failure acute,
1 dizziness, 1 intestinal obstruction, 1 cerebrovascular accident, 1 bile duct stone, 1 calculus ureteric, 1 angioedema

9 out of 393

1 atrial fibrillation

1 out of 97

Anderson 2014a1

Paritaprevir/ABT‐072/dasabuvir

1 haemorrhoids, 1 malignant melanoma

2 out of 63

None reported

0 out of 11

Feld 2014

Paritaprevir/ombitasvir

1 anaemia, 1 abdominal pain, 1 diarrhoea, 1 cholecystitis, 1 appendicitis, 1 overdose, 1 sinus tachycardia, 1 ventricular extrasystoles, 1 nausea, 1 vomiting, 1 chills, 1 non‐cardiac chest pain, 1 lobar pneumonia, 1 postoperative wound infection, 1 lumbar vertebral fracture, 1 non‐small cell lung cancer, 1 encephalopathy, 1 acute respiratory failure, 1 hypoxia, 1 mediastinal mass, 1 aortic stenosis, 1 biliary colic, 1 subcutaneous abscess

12 out of 630

None reported

0 out of 158

Pockros 2008a1

R1626

6 SAEs but not specified

6 out of 84

1 SAE but not specified

1 out of 20

Forns 2014

Simeprevir

1 abdominal pain, 1 pyrexia, 1 appendicitis, 2 pneumonia, 1 depression, 1 dyspnoea, 1 cholelithiasis, 1 anaemia haemolytic autoimmune, 1 pancytopenia, 1 angina pectoris, 1 bradycardia, 1 myocardial ischaemia, 1 hepatitis, 1 endocarditis, 1 lower respiratory tract infection, 1 septic shock, 1 breast cancer, 1 Guillain‐Barre syndrome, 1 presyncope, 1 confusional state, 1 vaginal haemorrhage, 1 chronic obstructive pulmonary disease, 1 respiratory acidosis, 1 photosensitivity reaction

14 out of 260

1 atrial fibrillation, 1 depression, 1 bronchitis, 1 hypercalcaemia, 1 head injury, 1 bacterial prostatitis, 1 pericarditis, 1 infection, 1 inguinal hernia, 1 neuropathy peripheral, 1 arthritis infective, 1 headache

11 out of 133

Fried 2013

Simeprevir

1 cholecystitis, 1 intervertebral disc protrusion, 1 depression, 1 nausea, 1 breast cancer, 1 hyperthyroidism, 1 ocular vasculitis, 1 abdominal pain upper, 1 colitis, 1 small intestinal obstruction, 1 malaise, 1 incision site cellulitis, 1 necrotising fasciitis, 1 perihepatic abscess, 1 pneumonia pneumococcal, 1 upper respiratory tract infection, 1 post‐procedural bile leak, 1 malnutrition, 1 type 1 diabetes mellitus, 1 spinal disorder, 1 parathyroid tumour benign, 1 headache, 1 haemoptysis, 1 cutaneous vasculitis, 1 hypertension

20 out of 309

1 myocardial infarction, 1 myopericarditis, 1 asthenia, 1 appendicitis, 1 vomiting, 1 chronic obstructive pulmonary disease, 1 headache, 1 subcutaneous abscess, 1 vulval abscess, 1 myositis, 1 ovarian neoplasm

10 out of 77

DRAGON 2014a1

Simeprevir

1 subarachnoid haemorrhage, 1 malaise, 1 cerebral infarction, 1 vulvar erosion, 1 rash, 1 incorrect dose administered

5 out of 79

None reported

0 out of 13

Hoeben 2015a1

Simeprevir

1 depression, 1 non‐cardiac chest pain, 1 angina unstable, 1 nephrolithiasis, 1 ureteric stenosis, 1 colitis ischaemic, 1 incision site infection, 1 craniocerebral injury, 1 foot fracture, 1 meniscus lesion, 1 multiple injuries, 1 rib fracture, 1 tibia fracture, 1 traumatic lung injury, 1 wound, 1 cholesterosis, 1 type 2 diabetes mellitus, 1 shock haemorrhagic

10 out of 305

1 anaemia, 1 decreased appetite, 1 cholelithiasis, 1 contusion, 1 supraventricular tachycardia, 1 ligament sprain, 1 pain, 1 atypical pneumonia, 1 chronic hepatitis C, 1 pulmonary tuberculosis, 1 undifferentiated connective tissue disease, 1 brain neoplasm

9 out of 152

OPERA 2011a1

Simeprevir

1 sinus arrest, 1 erysipelas, 1 type 1 diabetes mellitus, 1 psychotic disorder, 1 drug abuse, 1 bronchitis, 1 exostosis, 1 toe deformity, 1 hyperthyroidism, 1 Bowen's disease, 1 neutropenia, 1 thrombocytopenia, 1 breast cancer, 1 sepsis, 1 cupulolithiasis, 1 pneumonia escherichia, 1 panic reaction

13 out of 88

1 pneumonia, 1 sinusitis, 1 panic attack, 1 social stay hospitalisation, 1 pneumonia escherichia

3 out of 28

Manns 2014a

Simeprevir

2 anaemia, 1 back pain, 1 syncope, 1 hyperthyroidism, 1 death, 1 muscle spasms, 1 colon cancer, 1 anal abscess, 1 urinary tract infection, 1 mixed deafness, 1 hyphaema, 1 visual impairment, 1 enterocutaneous fistula, 1 autoimmune hepatitis, 1 lymphadenitis bacteria, 1 fluid overload, 1 epilepsy, 1 memory impairment, 1 aggression

16 out of 257

1 anaemia, 1 pancreatitis acute, 1 dehydration, 1 vomiting, 1 pancytopenia, 1 loss of consciousness, 1 angina unstable, 1 meniscus lesion, 1 pulmonary embolism, 1 cholecystitis acute, 1 drug abuse, 1 retinal ischaemia, 1 respiratory tract infection viral, 1 viral infection, 1 neuropathy peripheral, 1 thoracic outlet syndrome

10 out of 134

Pearlman 2015

Simeprevir

None reported

0 out of 58

1 liver decompensation

1 out of 24

ASPIRE 2014

Simeprevir

1 anaemia, 1 abdominal pain, 1 diarrhoea, 1 oedema peripheral, 2 cholecystitis, 1 pneumonia, 1 overdose, 2
dehydration, 1 intervertebral disc protrusion, 1 hepatic neoplasm malignant, 2 vomiting, 1 non‐cardiac chest pain, 1 neutropenia, 2 cellulitis, 1 pancytopenia, 1 headache, 1 hypertension, 1 suicide attempt, 1 drug eruption, 2 clostridium difficile colitis, 1 nephrolithiasis, 1 pulmonary embolism, 1 rectal cancer, 1 sinusitis, 3 urinary tract infection, 1 diabetes mellitus, 1 migraine, 1 coma, 1 epistaxis, 1 alcohol abuse, 1 haemorrhagic anaemia, 1 cervix carcinoma, 1 periodontal disease, 1 enteritis, 1 gastro intestinal pain, 1 gingival infection, 1 lung infection, 1 meningitis bacterial, 1 pneumonia bordetella, 1 salpingitis, 1 thermal burn, 1 neurilemmoma benign, 1 brain injury, 1 cerebral haemorrhage, 1 vii nerve paralysis, 1 metrorrhagia, 1 pelvic adhesions

31 out of 396

1 sciatica, 1 nausea, 1 vomiting, 1 lower respiratory tract infection, 1 haemorrhoids, 1 weight decreased, 1 histiocytosis haematophagic, 1 tuberculosis

4 out of 66

POSITRON 2013

Sofosbuvir

1 drug withdrawal syndrome, 1 non‐cardiac chest pain, 1 oedema peripheral, 1 pyrexia, 1 hypersensitivity, 1 abdominal abscess, 1 cellulitis, 2 overdose, 1 injury, 1 road traffic accident, 1 spinal compression fracture, 1 hypoglycaemia, 1 hepatic neoplasm malignant, 1 abnormal behaviour, 1 eczema

11 out of 207

1 pancreatitis, 1 bile duct stone, 1 bronchitis

2 out of 71

Lawitz 2013a1

Sofosbuvir

1 retinal vein occlusion, 1 depression, 1 suicidal ideation, 1 lymphangitis, 1 acute myocardial infarction

4 out of 95

1 chest pain, 1 electrocardiogram ST segment elevation

1 out of 26

FISSION 2013

Sofosbuvir

1 anaemia, 1 chest pain, 1 cellulitis, 1 chronic obstructive pulmonary disease, 1 urinary tract infection, 1 allergy to anthropod sting, 1 osteomyelitis chronic , 1 toxicity to various agents

7 out of 256

1 pneumothorax, 1 breast cancer, 1 infection, 1 atrioventricular shock, 1 clavicle fracture, 1 rib fracture

3 out of 243

Rodriguez‐Torres 2013

Sofosbuvir

1 anaemia, 1 depression, 1 peripheral ischaemia, 1 pancreatitis acute

4 out of 49

1 abdominal pain

1 out of 14

Feld 2015

Sofosbuvir/velpatasvir

1 bronchitis, 1 cellulitis, 1 influenza, 1 chronic obstructive pulmonary disease, 1 death, 1 gastroenteritis, 1 acute myocardial infarct, 1 ligament sprain, 1 foot abscess, 1 foot necrosis, 1 recurring appendicitis, 1 epileptic seizure, 1 rotator cuff syndrome, 1 lung cancer, 1 mania, 1 palpitations, 1 small bowel obstruction, 1 upper limb fracture, 1 vestibular neuronitis

15 out of 624

None reported

0 out of 116

Benhamou 2013a1

Telaprevir

1 cholelithiasis

1 out of 16

None reported

0 out of 8

Hezode 2009

Telaprevir

5 anaemia, 2 abdominal pain, 1 asthenia, 1 pyrexia, 1 back pain, 3 syncope, 3 depression, 2 dyspnoea, 1 nausea, 1 chills, 1 pancytopenia, 5 rash, 1 lymphadenopathy, 1 hydrocele, 1 retinal haemorrhage, 1 catheter‐related complication, 1 bacterial sepsis, 1 pneumonia, 1 herpes viral, 1 sepsis, 1 road traffic accident, 1 tendon rupture, 1 lung neoplasm malignant, 1 speech disorder, 1 disorientation, 1 emotional distress, 1 suicide attempt, 1 renal failure, 1 acute testicular swelling, 3 pruritis, 2 drug eruption, 2 rash maculo‐papular, 1 rash erythematous, 1 rash generalised, 1 toxic skin eruption, 1 urticaria, 1 splenectomy

36 out of 241

2 anaemia, 1 angina pectoris, 1 syncope, 1 hyperthyroidism, 1 gastroenteritis, 1 haemorrhagic anaemia, 1 alcoholic pancreatitis, 1 paranoia, 1 uterine polyp

8 out of 82

ADVANCE 2011a1

Telaprevir

18 anaemia, 3 pneumonia, 3 syncope, 2 cellulitis, 5 rash, 3 psychiatric disorder, 2 musculoskeletal disorder, 2 cardiac disorder, 2 eye disorder, 3 hepatobiliary disorders, 2 vascular disorder

64 out of 727

4 anaemia, 1 cellulitis, 3 psychiatric disorder, 3 musculoskeletal disorder, 2 cardiac disorder, 4 renal and urinary disorder, 1 eye disorder, 1 vascular disorder

24 out of 361

McHutchison 2009

Telaprevir

2 anaemia, 1 gastroenteritis viral, 1 dehydration, 2 depression, 1 non‐cardiac chest pain, 1 chronic obstructive pulmonary disease, 1 rash, 1 rash generalised, 1 furuncle, 1 colitis ischaemic, 1 acute myocardial infarction, 1 adrenal disorder, 2 scotoma, 1 retinal exudates, 1 retinal infarction, 1 bronchitis bacterial, 1 incisional hernia, 1 lumbar radiculopathy, 1 exfoliative rash

18 out of 175

1 lobar pneumonia, 1 pancytopenia, 1 anxiety, 1 lymphadenitis bacteria, 1 deafness neurosensory

4 out of 75

McHutchison 2010

Telaprevir

6 anaemia, 1 pancreatitis acute, 1 gastroenteritis viral, 1 pneumonia, 1 dehydration, 1 back pain, 1 suicidal ideation, 2 cholelithiasis, 1 postoperative wound infection, 1 upper gastrointestinal haemorrhage, 1 confusional state, 2 small intestinal obstruction, 1 necrotising fasciitis, 1 pneumonia pneumococcal, 1 post‐procedural bile leak, 1 rash, 1 renal failure acute, 1 retinal detachment, 9 gastroenteritis, 1 cholecystitis acute, 1 sinusitis, 1 hypokalaemia, 1 eczema, 2 pancreatitis, 1 dermatitis, 1 diverticulitis, 1 alcohol abuse, 1 hypotension, 1 haemorrhagic anaemia, 1 idiopathic thrombocytopenic purpura, 1 cardiomyopathy, 1 diverticular perforation, 1 gastritis erosive, 1 abscess intestinal, 1 cholecystitis infective, 1 infected insect bite, 1 sepsis syndrome, 1 hypovolaemia, 1 B‐cell unclassifiable lymphoma low grade, 1 migraine with aura, 1 ruptured cerebral aneurysm, 1 neurogenic bladder, 1 lichenoid keratosis, 1 rash macular

28 out of 339

1 anaemia, 1 pneumonia, 2 dehydration, 1 syncope, 1 depression, 1 non‐small cell lung cancer, 1 headache, 2 rash, 1 renal failure acute, 2 gastroenteritis, 1 renal tubular acidosis, 1 decubitus ulcer, 1 hematoma

9 out of 114

Sulkowski 2013a

Telaprevir

1 myocardial infarction, 1 staphylococcal infection, 1 pyelonephritis acute, 1 haemolytic anaemia, 1 groin infection, 1 cellulitis staphylococcal, 1 staphylococcal abscess, 1 hypokalaemia, 1 hyponatraemia, 1 epididymitis, 1 non‐ cardiac chest pain

7 out of 38

1 anaemia, 1 appendicitis, 1 peritonitis

2 out of 22

Zeuzem 2011a

Telaprevir

13 anaemia, 1 febrile neutropenia, 2 pancytopenia, 1 thrombocytopenia , 3 acute myocardial infarction, 2 atrial fibrillation, 1 cardiac valve disease, 1 myocardial infarction, 1 supraventricular tachycardia, 1 sudden hearing loss, 1 Basedow's disease, 1 retinal detachment, 1 abdominal pain, 1 anal fissure, 1 caecitis, 1 gastrointestinal haemorrhage, 1 pancreatitis, 2 pancreatitis acute, 1 general physical health deterioration, 1 pyrexia, 1 appendicitis, 2 bronchitis, 1 erysipelas, 1 folliculitis, 1 Helicobacter gastritis, 1 pneumonia, 1 post‐procedural infection, 1 rectal abscess, 2 sepsis, 1 sinusitis, 1 tooth abscess, 2 urinary tract infection, 1 injection site reaction, 1 animal scratch, 1 ankle fracture, 1 femoral neck fracture, 1 multiple drug overdose, 1 blood corticotrophin decreased, 1 weight decreased, 1 anorexia, 1 diabetes mellitus, 1 bronchial carcinoma, 2 gastric cancer, 2 hepatic neoplasm malignant, 1 histiocytosis haematophagic, 1 lung neoplasm malignant, 1 lethargy, 1 subarachnoid haemorrhage, 2 syncope, 1 delirium, 1 depression, 1 insomnia, 1 substance abuse, 1 renal cyst, 1 renal failure, 1 urinary bladder polyp, 1 prostatitis, 1 pulmonary embolism, 1 dermatitis, 1 eczema , 1 erythema multiforme, 1 pruritus 1 pustular psoriasis, 1 rash, 2 toxic skin eruption, 1 orthostatic hypotension, 1 peripheral artery aneurysm

65 out of 530

1 anaemia, 1 atrial fibrillation, 1 abdominal pain, 1 pneumonia, 1 colitis, 1 pyelonephritis, 1 cerebral thrombosis, 1 coma

7 out of 132

Manns 2012a1

Vaniprevir

1 appendicitis, 1 lobar pneumonia, 1 septic shock, 1 confusional state, 1 gastroenteritis, 1 cholecystitis acute, 1 empyema, 1 haemoglobin decreased, 1 myopathy

8 out of 75

1 colon cancer

1 out of 19

Lawitz 2013c

Vaniprevir

1 anaemia, 1 pneumonia, 1 syncope, 1 upper gastrointestinal haemorrhage, 1 cellulitis, 1 confusional state, 1 dizziness, 1 nephrolithiasis, 1 malignant melanoma, 3 retinal detachment, 1 joint dislocation, 1 congestive cardiac failure, 2 gastroenteritis, 1 femur fracture, 1 hyperglycaemia, 1 dermatomyositis, 1 retinal vascular thrombosis, 1 general physical health deterioration, 1 anaphylactic reaction, 1 pyelonephritis, 1 pyelonephritis acute, 1 carbon monoxide poisoning, 1 arthralgia, 1 arthritis infective, 1 completed suicide

22 out of 229

1 hypertensive crisis

1 out of 56

SAE: serious adverse events.

Figuras y tablas -
Table 2. Serious adverse events
Table 3. Non‐serious adverse events

Trial

Experimental intervention

Type and number of participants with a non‐serious adverse events (experimental group)

Proportion of participants with a non‐serious adverse event (experimental group)

Type and number of participants with a non‐serious adverse events (control group)

Proportion of participants with a non‐serious adverse event (control group)

Bronowicki 2013a1

Asunaprevir

18 diarrhoea, 13 nausea, 10 asthenia, 21 fatigue, 14 influenza‐like illness, 7 irritability, 12 decreased appetite, 4 arthralgia, 9 myalgia, 13 headache, 9 depression, 10 insomnia, 7 cough, 10 dyspnoea, 7 alopecia, 11 dry skin, 10 pruritus, 6 rash

36 out of 36

1 diarrhoea, 2 nausea, 4 asthenia, 5 fatigue, 5 influenza‐like illness, 4 irritability, 3 decreased appetite, 4 arthralgia, 1 myalgia, 6 headache, 1 depression, 1 insomnia, 3 cough, 3 dyspnoea, 3 alopecia, 1 dry skin, 2 pruritus, 3 rash

11 out of 11

Bronowicki 2014

Asunaprevir

8 anaemia, 63 asthenia, 62 fatigue, 37 influenza‐like illness, 43 decreased appetite, 66 headache, 41 pruritus

173 out of 177

3 anaemia, 1 asthenia, 62 fatigue, 23 influenza‐like illness, 22 decreased appetite, 26 headache, 16 pruritus

57 out of 61

Pasquinelli 2012a1

Asunaprevir

1 nausea, 3 headache, 1 flatulence

Not specified out of 20

None reported

Not specified out of 4

Pasquinelli 2012a2

Asunaprevir

1 nausea, 3 headache

Not specified out of 12

1 nausea, 1 flatulence

Not specified out of 3

Nelson 2012a1

Balapiravir

120 anaemia, 41 neutropenia, 115 diarrhoea, 162 nausea, 138 chills, 231 fatigue, 117 pyrexia, 100 arthralgia, 156 myalgia, 82 dizziness, 241 headache, 90 depression, 174 insomnia, 86 cough, 88 alopecia, 60 dry skin, 108 pruritus, 90 rash

Not specified out of 432

6 anaemia, 3 neutropenia, 16 diarrhoea, 25 nausea, 30 chills, 43 fatigue, 19 pyrexia, 16 arthralgia, 33 myalgia, 15 dizziness, 42 headache, 17 depression, 24 insomnia, 11 cough, 11 alopecia, 16 dry skin, 15 pruritus, 13 rash

Not specified out of 72

Tatum 2015a1

Beclabuvir

5 anaemia, 5 neutropenia, 5 diarrhoea, 9 nausea, 3 chills, 12 fatigue, 6 influenza‐like illness, 7 irritability, 3 pyrexia, 7 decreased appetite, 4 arthralgia, 5 myalgia, 12 headache, 6 depression, 9 insomnia, 6 cough, 5 pruritus, 2 rash

Not specified out of 26

5 anaemia, 1 neutropenia, 1 diarrhoea, 2 nausea, 5 fatigue, 7 influenza‐like illness, 3 irritability, 1 pyrexia, 2 decreased appetite, 3 headache, 3 depression, 3 insomnia, 3 cough, 4 pruritus, 4 rash

Not specified out of 13

Erhardt 2009

BILB‐1941

25 diarrhoea, 7 nausea, 2 vomiting

30 out of 77

2 diarrhoea

3 out of 19

Sims 2014

BMS‐791325

1 diarrhoea, 1 nausea, 1 vomiting, 1 headache, 1 pruritus

9 out of 20

1 nausea, 1 vomiting, 1 headache

1 out of 4

Bacon 2011a1

Boceprevir

145 anaemia, 46 neutropenia, 78 diarrhoea, 140 nausea, 47 vomiting, 68 asthenia, 106 chills, 179 fatigue, 79 influenza‐like illness, 67 irritability, 93 pyrexia, 83 decreased appetite, 73 arthralgia, 81 myalgia, 52 dizziness, 142 dysgeusia, 133 headache, 46 depression, 97 insomnia, 70 cough, 69 dyspnoea, 71 alopecia, 72 dry skin, 62 pruritus, 51 rash

319 out of 323

16 anaemia, 8 neutropenia, 13 diarrhoea, 30 nausea, 6 vomiting, 13 asthenia, 24 chills, 40 fatigue, 20 influenza‐like illness, 10 irritability, 20 pyrexia, 13 decreased appetite, 13 arthralgia, 19 myalgia, 8 dizziness, 9 dysgeusia, 39 headache, 12 depression, 19 insomnia, 14 cough, 14 dyspnoea, 13 alopecia, 7 dry skin, 14 pruritus, 5 rash

77 out of 80

Flamm 2013

Boceprevir

67 anaemia, 41 neutropenia, 33 diarrhoea, 52 nausea, 16 vomiting, 29 asthenia, 14 chills, 67 fatigue, 35 influenza‐like illness, 29 irritability, 18 pyrexia, 27 decreased appetite, 16 arthralgia, 25 myalgia, 17 dizziness, 52 dysgeusia, 37 headache, 22 depression, 32 insomnia, 26 cough, 26 dyspnoea, 22 alopecia, 20 dry skin, 18 pruritus, 31 rash

133 out of 134

22 anaemia, 12 neutropenia, 5 diarrhoea, 18 nausea, 12 asthenia, 8 chills, 36 fatigue, 18 influenza‐like illness, 16 irritability, 8 pyrexia, 12 decreased appetite, 12 arthralgia, 5 myalgia, 10 dizziness, 10 dysgeusia, 21 headache, 6 depression, 20 insomnia, 14 cough, 17 dyspnoea, 5 alopecia, 11 dry skin, 8 pruritus, 5 rash

67 out of 67

Isakov 2016

Boceprevir

105 anaemia, 103 leukopenia, 141 neutropenia, 16 thrombocytopenia, 22 diarrhoea, 13 dry mouth, 59 nausea,

12 vomiting, 63 asthenia, 24 chills, 40 fatigue, 51 hyperthermia,

356 influenza‐like illness, 9 injection site erythema, 18 irritability, 217 pyrexia, 14 body temperature increased, 33 weight decreased, 10 decreased appetite, 16 arthralgia, 33 myalgia, 9 dizziness, 69 dysgeusia, 89 headache, 2 sleep disorder, 38 cough, 7 dyspnoea, 33 alopecia, 12 dry skin, 20 pruritus, 17 rash

153 out of 159

31 anaemia, 35 leukopenia, 45 neutropenia, 7 thrombocytopenia, 3 diarrhoea, 5 dry mouth, 15 nausea,

2 vomiting, 23 asthenia, 2 chills, 18 fatigue, 12 hyperthermia,

72 influenza‐like illness, 2 injection site erythema, 11 irritability,

124 pyrexia, 2 body temperature increased, 9 weight decreased, 7 decreased appetite, 4 arthralgia, 8 myalgia, 7 dizziness, 5 dysgeusia, 51 headache, 4 sleep disorder, 14 cough, 7 dyspnoea, 16 alopecia, 3 dry skin, 6 pruritus, 2 rash

71 out of 78

Kwo 2010a1

Boceprevir

226 anaemia, 96 neutropenia, 109 diarrhoea, 186 nausea, 81 vomiting, 53 asthenia, 130 chills, 259 fatigue, 79 influenza‐like illness, 91 irritability, 129 pyrexia, 49 decreased appetite, 76 arthralgia, 99 myalgia, 70 dizziness, 111 dysgeusia, 190 headache, 91 depression, 146 insomnia, 76 cough, 66 dyspnoea, 131 alopecia, 60 dry skin, 80 pruritus, 27 rash

413 out of 416

35 anaemia, 12 neutropenia, 23 diarrhoea, 45 nausea, 5 vomiting, 14 asthenia, 35 chills, 57 fatigue, 25 influenza‐like illness, 23 irritability, 35 pyrexia, 12 decreased appetite, 21 arthralgia, 17 myalgia, 16 dizziness, 9 dysgeusia, 45 headache, 22 depression, 40 insomnia, 20 cough, 15 dyspnoea, 27 alopecia, 17 dry skin, 16 pruritus, 6 rash

102 out of 104

Poordad 2011a1

Boceprevir

361 anaemia, 184 neutropenia, 180 diarrhoea, 334 nausea, 145 vomiting, 125 asthenia, 255 chills, 405 fatigue, 174 influenza‐like illness, 164 irritability, 240 pyrexia, 186 decreased appetite, 141 arthralgia, 170 myalgia, 146 dizziness, 293 dysgeusia, 335 headache, 151 depression, 239 insomnia, 130 cough, 152 dyspnoea, 179 alopecia, 153 dry skin, 181 pruritus, 181 rash

728 out of 734

107 anaemia, 77 neutropenia, 79 diarrhoea, 153 nausea, 57 vomiting, 70 asthenia, 102 chills, 217 fatigue, 93 influenza‐like illness, 86 irritability, 120 pyrexia, 90 decreased appetite, 66 arthralgia, 94 myalgia, 59 dizziness, 64 dysgeusia, 153 headache, 78 depression, 118 insomnia, 76 cough, 59 dyspnoea, 99 alopecia, 66 dry skin, 98 pruritus, 83 rash

353 out of 363

Sulkowski 2013a

Boceprevir

26 anaemia, 12 neutropenia, 21 diarrhoea, 26 nausea, 18 vomiting, 22 asthenia, 5 chills, 25 fatigue, 16 influenza‐like illness, 10 irritability, 24 pyrexia, 22 decreased appetite, 7 arthralgia, 9 myalgia, 8 dizziness, 18 dysgeusia, 18 headache, 11 depression, 15 insomnia, 9 cough, 5 dyspnoea, 12 alopecia, 8 dry skin, 13 pruritus, 5 rash

62 out of 64

8 anaemia, 2 neutropenia, 6 diarrhoea, 11 nausea, 5 vomiting, 9 asthenia, 5 chills, 12 fatigue, 13 influenza‐like illness, 5 irritability, 7 pyrexia, 6 decreased appetite, 2 arthralgia, 6 myalgia, 2 dizziness, 5 dysgeusia, 6 headache, 4 depression, 9 insomnia, 6 cough, 2 dyspnoea, 6 alopecia, 3 dry skin, 3 pruritus

33 out of 34

Dore 2015a1

Daclatasvir

7 anaemia, 9 neutropenia, 10 diarrhoea, 22 nausea, 4 vomiting, 13 asthenia, 4 chills, 35 fatigue, 19 influenza‐like illness, 17 irritability, 7 pyrexia, 12 decreased appetite, 11 arthralgia, 14 myalgia, 6 dizziness, 5 dysgeusia, 30 headache, 11 depression, 19 insomnia, 8 cough, 12 dyspnoea, 12 alopecia, 13 dry skin, 27 pruritus, 25 rash

98 out of 100

5 anaemia, 8 neutropenia, 3 diarrhoea, 8 nausea, 4 vomiting, 7 asthenia, 19 fatigue, 7 influenza‐like illness, 6 irritability, 2 pyrexia, 8 decreased appetite, 9 arthralgia, 11 myalgia, 6 dizziness, 3 dysgeusia, 9 headache, 9 depression, 17 insomnia, 8 cough, 6 dyspnoea, 5 alopecia, 6 dry skin, 14 pruritus, 12 rash

48 out of 51

COMMAND‐1 2015a1

Daclatasvir

53 anaemia, 43 neutropenia, 73 diarrhoea, 109 nausea, 34 vomiting, 32 asthenia, 49 chills, 174 fatigue, 94 influenza‐like illness, 72 irritability, 48 pyrexia, 67 decreased appetite, 55 arthralgia, 88 myalgia, 46 dizziness, 25 dysgeusia, 136 headache, 45 depression, 102 insomnia, 54 cough, 58 dyspnoea, 80 alopecia, 88 dry skin, 119 pruritus, 94 rash

311 out of 317

9 anaemia, 9 neutropenia, 14 diarrhoea, 20 nausea, 11 vomiting, 7 asthenia, 16 chills, 46 fatigue, 16 influenza‐like illness, 22 irritability, 15 pyrexia, 17 decreased appetite, 19 arthralgia, 24 myalgia, 9 dizziness, 4 dysgeusia, 36 headache, 10 depression, 30 insomnia, 18 cough, 11 dyspnoea, 13 alopecia, 15 dry skin, 26 pruritus, 25 rash

76 out of 78

Izumi 2014a1

Daclatasvir

11 anaemia, 7 neutropenia, 2 diarrhoea, 4 nausea, 6 fatigue, 1 irritability, 11 pyrexia, 7 decreased appetite, 4 arthralgia, 1 myalgia, 1 dizziness, 4 dysgeusia, 3 headache, 7 insomnia, 4 cough, 2 dyspnoea, 8 alopecia, 1 dry skin, 6 pruritus, 7 rash

34 out of 34

5 anaemia, 4 neutropenia, 3 diarrhoea, 2 nausea, 3 vomiting, 4 chills, 4 fatigue, 2 influenza‐like illness, 5 pyrexia, 5 decreased appetite, 2 arthralgia, 2 myalgia, 1 dizziness, 1 dysgeusia, 4 headache, 2 insomnia, 1 cough, 6 alopecia, 1 dry skin, 3 pruritus, 3 rash

8 out of 8

Pol 2012

Daclatasvir

14 anaemia, 9 neutropenia, 5 diarrhoea, 13 nausea, 7 vomiting, 9 asthenia, 4 chills, 19 fatigue, 11 influenza‐like illness, 12 irritability, 7 pyrexia, 9 decreased appetite, 2 arthralgia, 8 myalgia, 5 dizziness, 2 dysgeusia, 19 headache, 7 depression, 9 insomnia, 9 cough, 6 dyspnoea, 8 alopecia, 2 dry skin, 12 pruritus, 10 rash

36 out of 36

5 anaemia, 5 neutropenia, 3 diarrhoea, 6 nausea, 1 asthenia, 2 chills, 9 fatigue, 4 influenza‐like illness, 2 irritability, 3 pyrexia, 3 decreased appetite, 3 myalgia, 1 dizziness, 1 dysgeusia, 3 headache, 3 depression, 6 insomnia, 3 cough, 2 dyspnoea, 2 alopecia, 1 dry skin, 3 pruritus, 3 rash

12 out of 12

Nettles 2010

Daclatasvir

1 diarrhoea, 1 nausea, 4 headache

7 out of 16

None reported

0 out of 2

Nettles 2011a1

Daclatasvir

2 diarrhoea, 3 fatigue, 1 arthralgia, 1 dizziness, 5 headache, 2 insomnia, 1 dry skin

16 out of 24

1 nausea, 1 vomiting, 2 headache

4 out of 6

Dauphine 2015a1

Danoprevir

115 diarrhoea, 106 nausea, 77 asthenia, 89 chills, 158 fatigue, 125 pyrexia, 88 decreased appetite, 72 arthralgia, 93 myalgia,158 headache, 102 insomnia, 62 cough, 54 alopecia, 83 pruritus, 78 rash

364 out of 373

5 diarrhoea, 13 nausea, 9 asthenia, 8 chills, 17 fatigue, 15 pyrexia, 6 decreased appetite, 9 arthralgia, 13 myalgia, 24 headache, 16 insomnia, 12 cough, 4 alopecia, 14 pruritus, 6 rash

42 out of 44

Forestier 2011a1

Danoprevir

2 diarrhoea, 3 myalgia, 5 headache

21 out of 40

1 diarrhoea, 2 headache

3 out of 10

Forestier 2011b

Danoprevir

6 neutropenia, 5 diarrhoea, 4 nausea, 3 asthenia, 5 chills, 8 fatigue, 4 influenza‐like illness, 2 pyrexia, 2 arthralgia, 17 myalgia, 6 dizziness, 23 headache, 2 depression, 6 insomnia, 3 pruritus

42 out of 47

2 neutropenia, 2 diarrhoea, 1 nausea, 1 chills, 3 fatigue, 1 influenza‐like illness, 1 arthralgia, 5 myalgia, 1 dizziness, 4 headache, 1 depression, 2 insomnia

12 out of 12

Gane 2011

Danoprevir

4 diarrhoea, 5 nausea, 5 fatigue, 3 influenza‐like illness, 5 irritability, 5 arthralgia, 5 myalgia, 11 headache, 4 insomnia, 6 rash

24 out of 25

1 diarrhoea, 3 nausea, 1 fatigue, 2 myalgia, 4 headache, 2 insomnia

5 out of 5

Marcellin 2013a

Danoprevir

53 anaemia, 70 neutropenia, 56 diarrhoea, 85 nausea, 29 vomiting, 57 chills, 109 fatigue, 38 irritability, 51 pyrexia, 34 decreased appetite, 29 arthralgia, 60 myalgia, 92 headache, 42 depression, 69 insomnia, 31 alopecia, 46 pruritus, 42 rash

Not specified out of 194

13 anaemia, 11 neutropenia, 7 diarrhoea, 10 nausea, 4 vomiting, 13 chills, 14 fatigue, 7 irritability, 5 pyrexia, 4 decreased appetite, 9 arthralgia, 11 myalgia, 18 headache, 4 depression, 9 insomnia, 5 alopecia, 6 pruritus, 8 rash

Not specified out of 31

Larrey 2012

Deleobuvir

1 anaemia, 19 diarrhoea, 19 nausea, 11 vomiting, 16 asthenia, 3 chills, 12 fatigue, 14 influenza‐like illness, 7 irritability, 6 pyrexia, 14 decreased appetite, 4 arthralgia, 4 myalgia, 6 dizziness, 4 dysgeusia, 20 headache, 15 insomnia, 6 cough, 3 dyspnoea, 1 alopecia, 6 dry skin, 5 pruritus, 8 rash

49 out of 49

1 nausea, 1 asthenia, 1 chills, 2 fatigue, 1 influenza‐like illness, 2 irritability, 1 decreased appetite, 2 headache, 1 dry skin, 1 pruritus, 2 rash

7 out of 8

STARTverso‐2 2014a1

Faldaprevir

114 anaemia, 59 neutropenia, 160 diarrhoea, 249 nausea, 110 vomiting, 29 asthenia, 78 chills, 246 fatigue, 39 influenza‐like illness, 67 irritability, 79 pyrexia, 117 decreased appetite, 72 arthralgia, 100 myalgia, 80 dizziness, 31 dysgeusia, 165 headache, 68 depression, 137 insomnia, 89 cough, 53 dyspnoea, 96 alopecia, 67 dry skin, 164 pruritus, 298 rash

513 out of 525

27 anaemia, 14 neutropenia, 23 diarrhoea, 52 nausea, 11 vomiting, 6 asthenia, 25 chills, 70 fatigue, 15 influenza‐like illness, 27 irritability, 20 pyrexia, 26 decreased appetite, 22 arthralgia, 37 myalgia, 25 dizziness, 3 dysgeusia, 45 headache, 11 depression, 38 insomnia, 21 cough, 20 dyspnoea, 22 alopecia, 15 dry skin, 37 pruritus, 41 rash

130 out of 132

STARTVerso‐1 2015a1

Faldaprevir

89 anaemia, 57 neutropenia, 121 diarrhoea, 168 nausea, 79 vomiting, 96 asthenia, 143 fatigue, 92 influenza‐like illness, 37 irritability, 110 pyrexia, 87 decreased appetite, 39 arthralgia, 41 myalgia, 38 dizziness, 23 dysgeusia, 146 headache, 32 depression, 70 insomnia, 58 cough, 36 dyspnoea, 49 alopecia, 80 dry skin, 160 pruritus, 139 rash

496 out of 520

26 anaemia, 18 neutropenia, 17 diarrhoea, 19 nausea, 6 vomiting, 27 asthenia, 35 fatigue, 21 influenza‐like illness, 9 irritability, 32 pyrexia, 22 decreased appetite, 14 arthralgia, 20 myalgia, 13 dizziness, 5 dysgeusia, 40 headache, 8 depression, 22 insomnia, 20 cough, 16 dyspnoea, 15 alopecia, 17 dry skin, 41 pruritus, 25 rash

120 out of 132

STARTverso‐3 2013a1

Faldaprevir

98 anaemia, 65 neutropenia, 190 diarrhoea, 318 nausea, 171 vomiting, 108 asthenia, 32 chills, 204 fatigue, 107 influenza‐like illness, 49 irritability, 113 pyrexia, 128 decreased appetite, 60 arthralgia, 72 myalgia, 37 dizziness, 39 dysgeusia, 182 headache, 52 depression, 118 insomnia, 99 cough, 47 dyspnoea, 53 alopecia, 108 dry skin, 225 pruritus, 160 rash

585 out of 599

8 anaemia, 12 neutropenia, 10 diarrhoea, 18 nausea, 5 vomiting, 21 asthenia, 5 chills, 16 fatigue, 15 influenza‐like illness, 11 irritability, 14 pyrexia, 10 decreased appetite, 7 arthralgia, 8 myalgia, 6 dizziness, 4 dysgeusia, 22 headache, 10 depression, 13 insomnia, 16 cough, 7 dyspnoea, 4 alopecia, 12 dry skin, 23 pruritus, 16 rash

74 out of 78

Manns 2011

Faldaprevir

2 anaemia, 1 neutropenia, 4 diarrhoea, 7 nausea, 10 asthenia, 2 chills, 2 fatigue, 4 influenza‐like illness, 4 irritability, 2 pyrexia, 1 decreased appetite, 7 myalgia, 1 dizziness, 6 headache, 2 depression, 5 insomnia, 2 cough, 1 alopecia, 5 dry skin, 3 pruritis, 1 rash

32 out of 26

1 diarrhoea, 2 nausea, 2 asthenia, 2 headache, 1 depression, 1 insomnia, 1 cough

5 out of 8

Nishiguchi 2014a1

Faldaprevir

1 neutropenia, 3 diarrhoea, 3 nausea, 3 vomiting, 2 influenza‐like illness, 8 pyrexia, 2 decreased appetite, 2 arthralgia, 4 dizziness, 1 dysgeusia, 7 headache, 1 depression, 5 insomnia, 1 cough, 3 alopecia, 1 dry skin, 6 pruritus, 6 rash

33 out of 35

2 nausea, 2 vomiting, 1 influenza‐like illness, 1 pyrexia, 1 decreased appetite, 1 headache, 1 insomnia, 1 dyspnoea, 2 dry skin, 3 pruritus, 1 rash

6 out of 8

Sulkowski 2013a

Faldeprevir

85 anaemia, 49 neutropenia, 188 diarrhoea, 239 nausea, 105 vomiting, 122 asthenia, 54 chills, 194 fatigue, 217 influenza‐like illness, 82 irritability, 86 pyrexia, 133 decreased appetite, 72 arthralgia, 133 myalgia, 48 dizziness, 25 dysgeusia, 243 headache, 68 depression, 107 insomnia, 100 cough, 73 dyspnoea, 106 alopecia, 122 dry skin, 227 pruritus, 163 rash

620 out of 641

12 anaemia, 8 neutropenia, 13 diarrhoea, 14 nausea, 4 vomiting, 15 asthenia, 8 chills, 24 fatigue, 34 influenza‐like illness, 10 irritability, 11 pyrexia, 11 decreased appetite, 5 arthralgia, 12 myalgia, 5 dizziness, 27 headache, 7 depression, 17 insomnia, 9 cough, 11 dyspnoea, 8 alopecia, 10 dry skin, 12 pruritus, 12 rash

65 out of 71

Jacobson 2010

Filibuvir

13 anaemia,4 neutropenia ,6 diarrhoea, 13 nausea, 3 vomiting, 4 chills, 13 fatigue, 3 influenza‐like illness, 2 irritability, 2 pyrexia, 3 decreased appetite, 2 arthralgia, 4 myalgia, 4 dizziness, 14 headache, 5 depression, 11 insomnia, 5 cough, 2 dyspnoea, 2 alopecia, 3 dry skin, 2 pruritus, 3 rash

27 out of 27

3 anaemia, 3 diarrhoea, 4 nausea, 1 vomiting, 1 chills, 5 fatigue, 2 influenza‐like illness, 2 decreased appetite, 2 arthralgia, 2 headache, 3 depression, 2 insomnia, 2 cough, 2 dyspnoea, 1 alopecia, 2 dry skin, 3 pruritus, 1 rash

8 out of 8

Rodriguez‐Torres 2014a1

Filibuvir

26 anaemia, 26 neutropenia, 24 diarrhoea, 55 nausea, 20 vomiting, 35 asthenia, 25 chills, 73 fatigue, 29 influenza‐like illness, 33 irritability, 28 pyrexia, 36 decreased appetite, 30 arthralgia, 37 myalgia, 20 dizziness, 40 dysgeusia, 61 headache, 32 depression, 55 insomnia, 33 cough, 18 dyspnoea, 34 alopecia, 33 dry skin, 56 pruritus, 34 rash

174 out of 192

anaemia, neutropenia, diarrhoea, nausea, vomiting, asthenia, chills, fatigue, influenza‐like illness, irritability, pyrexia, decreased appetite, arthralgia, myalgia, dizziness, dysgeusia, headache, depression, insomnia, cough, dyspnoea, alopecia, dry skin, pruritus, rash.

The authors did not report number of adverse events in the control group

90 out of 96

Petry 2011

Grazoprevir

9 diarrhoea, 2 nausea, 1 vomiting, 7 fatigue, 1 dysgeusia, 16 headache, 1 insomnia, 2 pruritis

34 out of 76

1 headache

2 out of 15

Gardner 2014a

GSK2336805

3 anaemia, 3 neutropenia, 4 nausea, 2 vomiting, 2 chills, 5 fatigue, 2 cough

11 out of 11

1 fatigue

4 out of 4

Lalezari 2012

IDX‐184

1 diarrhoea, 2 fatigue, 1 dizziness, 4 headache

8 out of 33

1 diarrhoea, 1 fatigue, 1 dizziness, 1 headache

4 out of 8

Lalezari 2013

IDX‐184

6 neutropenia, 7 diarrhoea, 22 nausea, 5 vomiting, 15 chills, 36 fatigue, 10 irritability, 9 pyrexia, 5 decreased appetite, 19 myalgia, 27 headache, 4 depression, 10 insomnia, 6 pruritus

59 out of 65

4 neutropenia, 2 diarrhoea, 3 nausea, 5 chills, 9 fatigue, 4 irritability, 3 decreased appetite, 5 myalgia, 7 headache, 3 depression, 5 insomnia, 2 pruritus

12 out of 16

De Bruijne 2010a1

IDX320

1 diarrhoea, 1 myalgia, 4 headache

Not specified out of 30

None reported

0 out of 8

Lawitz 2012a

Ledispasvir

2 nausea, 6 headache, 2 rash

18 out of 59

1 nausea

4 out of 11

Gane 2010

Mericitabine/danoprevir

7 diarrhoea, 9 nausea, 36 headache, 9 rash

Not specified out of 73

1 diarrhoea, 2 nausea, 8 headache, 1 rash

Not specified out of 14

Feld 2015

Mericitabine

25 diarrhoea, 16 nausea, 6 chills, 47 fatigue, 14 irritability, 16 pyrexia, 12 arthralgia, 19 myalgia, 43 headache, 27 insomnia, 21 cough, 15 pruritus

Not specified out of 102

12 diarrhoea, 14 nausea, 12 chills, 25 fatigue, 10 irritability, 14 pyrexia, 11 arthralgia, 18 myalgia, 28 headache, 11 insomnia, 11 cough, 11 pruritus

Not specified out of 49

JUMP‐C 2013

Mericitabine

18 diarrhoea, 33 nausea, 31 chills, 58 fatigue, 21 irritability, 20 pyrexia, 25 decreased appetite, 18 arthralgia, 24 myalgia, 19 dizziness, 42 headache, 31 insomnia, 17 cough, 14 alopecia, 15 pruritus, 17 rash

Not specified out of 81

20 diarrhoea, 34 nausea, 33 chills, 58 fatigue, 25 irritability, 27 pyrexia, 22 decreased appetite, 21 arthralgia, 24 myalgia, 20 dizziness, 38 headache, 28 insomnia, 22 cough, 17 alopecia, 28 pruritus, 28 rash

Not specified out of 85

De Bruijne 2010a2

Narlaprevir

10 diarrhoea, 8 nausea, 30 influenza‐like illness, 6 dizziness, 11 headache

32 out of 32

1 nausea, 6 influenza‐like illness, 1 dizziness

7 out of 8

Vierling 2011

Narlaprevir

87 anaemia, 84 diarrhoea, 131 nausea, 54 vomiting, 44 chills, 123 fatigue, 106 influenza‐like illness, 58 irritability, 58 pyrexia, 61 decreased appetite, 60 arthralgia, 39 myalgia, 58 dizziness, 83 headache, 33 depression, 80 insomnia, 28 pruritus, 31 rash

Not specified out of 93

6 anaemia, 17 diarrhoea, 50 nausea, 28 vomiting, 17 chills, 56 fatigue, 44 influenza‐like illness, 28 irritability, 17 pyrexia, 44 decreased appetite, 28 arthralgia, 6 dizziness, 39 headache, 22 depression, 39 insomnia, 33 pruritus, 11 rash

Not specified out of 18

Muir 2014

Odalasvir/sovaprevir

5 anaemia, 2 diarrhoea, 5 fatigue, 2 influenza‐like illness, 2 irritability, 1 decreased appetite, 2 arthralgia, 3 myalgia, 1 dizziness, 1 dysgeusia, 6 headache, 2 insomnia, 4 cough, 1 dyspnoea, 1 pruritus, 1 rash

20 out of 20

2 diarrhoea, 4 fatigue, 1 myalgia, 1 dizziness, 1 headache, 1 cough, pruritus

10 out of 10

Sullivan 2012

Ombitasvir

6 anaemia, 5 neutropenia, 4 diarrhoea, 9 nausea, 7 vomiting, 6 chills, 18 fatigue, 1 influenza‐like illness, 1 irritability, 3 pyrexia, 5 decreased appetite, 2 arthralgia, 2 myalgia, 2 dizziness, 9 headache, 4 depression, 4 insomnia, 3 cough, 2 dyspnoea, 5 dry skin, 3 pruritus, 6 rash

26 out of 28

1 neutropenia, 1 diarrhoea, 3 nausea, 2 vomiting, 6 fatigue, 2 influenza‐like illness, 1 irritability, 1 decreased appetite, 1 myalgia, 2 headache, 1 depression, 2 insomnia, 1 cough, dyspnoea, 1 dry skin, 2 rash

9 out of 9

Anderson 2014a1

Paritaprevir/ABT‐072/dasabuvir

12 anaemia, 14 neutropenia, 16 diarrhoea, 15 nausea, 6 vomiting, 4 asthenia, 11 chills, 29 fatigue, 14 influenza‐like illness, 8 irritability, 12 pyrexia, 4 decreased appetite, 9 arthralgia, 14 myalgia, 9 dizziness, 6 dysgeusia, 38 headache, 15 depression, 14 insomnia, 6 cough, 6 dyspnoea, 5 alopecia, 2 dry skin, 8 pruritus, 12 rash

63 out of 63

1 anaemia, 2 neutropenia, 4 diarrhoea, 4 nausea, 1 vomiting, 1 asthenia, 6 fatigue, 1 influenza‐like illness, 1 irritability, 1 pyrexia, 2 decreased appetite, 1 arthralgia, 3 myalgia, 4 dizziness, 2 dysgeusia, 4 headache, 2 insomnia, 2 dyspnoea, 1 alopecia, 2 pruritus, 2 rash

10 out of 11

Feld 2014

Paritaprevir/ombitasvir

24 anaemia, 65 diarrhoea, 112 nausea, 23 vomiting, 59 asthenia, 164 fatigue, 26 irritability, 37 decreased appetite, 23 arthralgia, 21 myalgia, 38 dizziness, 156 headache, 67 insomnia, 34 cough, 38 dyspnoea, 27 dry skin, 80 pruritus, 51 rash

391 out of 473

11 diarrhoea, 22 nausea, 6 vomiting, 6 asthenia, 45 fatigue, 4 irritability, 5 decreased appetite, 9 arthralgia, 8 myalgia, 6 dizziness, 42 headache, 12 insomnia, 8 cough, 4 dyspnoea, 2 dry skin, 7 pruritus, 9 rash

108 out of 158

Zeuzem 2014a

Paritaprevir/ombitasvir

19 anaemia, 47 diarrhoea, 72 nausea, 22 vomiting, 60 asthenia, 115 fatigue, 22 irritability, 24 decreased appetite, 21 arthralgia, 28 myalgia, 30 dizziness, 13 dysgeusia, 126 headache, 52 insomnia, 43 cough, 50 dyspnoea, 27 dry skin, 53 pruritus, 34 rash

328 out of 394

12 diarrhoea, 17 nausea, 11 asthenia, 22 fatigue, 8 irritability, 2 decreased appetite, 7 arthralgia, 10 myalgia, 5 dizziness, 5 dysgeusia, 34 headache, 7 insomnia, 5 cough, 10 dyspnoea, 3 dry skin, 5 pruritus, 6 rash

74 out of 97

Hotho 2012

PHX1766

1 nausea, 2 fatigue, 1 dizziness

Not specified

None reported

Not specified

Pockros 2008a1

R1626

43 neutropenia, 42 diarrhoea, 49 nausea, 26 vomiting, 39 chills, 45 fatigue, 20 irritability, 29 pyrexia, 21 arthralgia, 23 myalgia, 15 dizziness, 47 headache, 27 insomnia, 15 cough, 11 pruritus, 20 rash

Not specified out of 84

5 diarrhoea, 10 nausea, 2 vomiting, 9 chills, 10 fatigue, 1 irritability, 8 pyrexia, 5 arthralgia, 11 myalgia, 3 dizziness, 11 headache, 6 insomnia, 4 cough, 6 pruritus, 2 rash

Not specified out of 20

Vince 2014

Samatasvir

4 nausea, 1 decreased appetite, 6 headache, 1 insomnia

20 out of 48

1 nausea, 1 decreased appetite, 1 headache, 1 insomnia

6 out of 12

Forns 2014

Simeprevir

40 anaemia, 37 neutropenia, 36 diarrhoea, 59 nausea, 18 vomiting, 57 asthenia, 17 chills, 87 fatigue, 78 influenza‐like illness, 63 pyrexia, 35 decreased appetite, 26 arthralgia, 39 myalgia, 14 dizziness, 12 dysgeusia, 87 headache, 22 depression, 49 insomnia, 34 cough, 26 dyspnoea, 26 alopecia, 24 dry skin, 16 pruritus, 33 rash

245 out of 260

24 anaemia, 26 neutropenia, 22 diarrhoea, 26 nausea, 9 vomiting, 25 asthenia, 11 chills, 58 fatigue, 27 influenza‐like illness, 30 pyrexia, 24 decreased appetite, 12 arthralgia, 17 myalgia, 6 dizziness, 7 dysgeusia, 48 headache, 10 depression, 33 insomnia, 21 cough, 5 dyspnoea, 17 alopecia, 18 dry skin, 37 pruritus, 19 rash

123 out of 133

Fried 2013

Simeprevir

63 anaemia, 75 neutropenia, 47 diarrhoea, 86 nausea, 22 vomiting, 63 asthenia, 25 chills, 107 fatigue, 98 influenza‐like illness, 11 irritability, 64 pyrexia, 17 decreased appetite, 53 arthralgia, 55 myalgia, 29 dizziness, 16 dysgeusia, 142 headache, 32 depression, 69 insomnia, 52 cough, 33 dyspnoea, 53 alopecia, 63 dry skin, 173 pruritus, 65 rash

302 out of 309

16 anaemia, 16 neutropenia, 12 diarrhoea, 21 nausea, 5 vomiting, 16 asthenia, 8 chills, 37 fatigue, 29 influenza‐like illness, 8 irritability, 13 pyrexia, 6 decreased appetite, 11 arthralgia, 17 myalgia, 6 dizziness, 5 dysgeusia, 40 headache, 14 depression, 23 insomnia, 15 cough, 6 dyspnoea, 16 alopecia, 14 dry skin, 35 pruritus, 18 rash

75 out of 77

DRAGON 2014a1

Simeprevir

24 anaemia, 13 diarrhoea,13 nausea, 6 vomiting, 4 chills, 2 fatigue, 42 pyrexia, 15 decreased appetite, 27 arthralgia, 15 myalgia, 3 dizziness, 6 dysgeusia, 41 headache, 2 depression, 23 insomnia, 8 cough, 25 alopecia, 5 dry skin, 15 pruritus, 47 rash

79 out of 79

5 anaemia, 5 diarrhoea, 2 vomiting, 7 pyrexia, 3 decreased appetite, 2 arthralgia, 2 myalgia, 8 headache, 2 insomnia, 2 cough, 6 alopecia, 6 rash

13 out of 13

CONCERTO‐1 2015

Simeprevir

70 anaemia, 8 neutropenia, 20 diarrhoea, 16 nausea, 6 vomiting, 13 fatigue, 75 pyrexia, 28 decreased appetite, 30 arthralgia, 9 myalgia, 4 dizziness, 20 dysgeusia, 54 headache, 27 insomnia, 11 cough, 44 alopecia, 8 dry skin, 35 pruritus, 57 rash

123 out of 123

36 anaemia, 1 neutropenia, 20 diarrhoea, 16 nausea, 6 vomiting, 7 fatigue, 31 pyrexia, 20 decreased appetite, 14 arthralgia, 11 myalgia, 4 dizziness, 8 dysgeusia, 26 headache, 3 depression, 25 insomnia, 8 cough, 28 alopecia, 9 dry skin, 18 pruritus, 37 rash

60 out of 60

Hoeben 2015a1

Simeprevir

82 anaemia, 59 neutropenia, 14 diarrhoea, 16 nausea, 15 asthenia, 63 fatigue, 39 influenza‐like illness, 67 pyrexia, 28 decreased appetite, 13 arthralgia, 36 myalgia, 39 headache, 17 insomnia, 16 cough, 47 alopecia, 40 pruritus, 57 rash

298 out of 305

53 anaemia, 32 neutropenia, 7 diarrhoea, 10 nausea, 7 asthenia, 36 fatigue, 19 influenza‐like illness, 46 pyrexia, 16 decreased appetite, 4 arthralgia, 22 myalgia, 28 headache, 18 insomnia, 17 cough, 26 alopecia, 17 pruritus, 27 rash

149 out of 152

Jacobson 2014

Simeprevir

44 anaemia, 54 neutropenia, 35 diarrhoea, 65 nausea, 23 vomiting, 25 asthenia, 33 chills, 111 fatigue, 62 influenza‐like illness, 51 pyrexia, 47 decreased appetite, 34 arthralgia, 39 myalgia, 23 dizziness, 16 dysgeusia, 88 headache, 23 depression, 56 insomnia, 25 cough, 23 dyspnoea, 30 alopecia, 33 dry skin, 68 pruritus, 60 rash

252 out of 264

24 anaemia, 15 neutropenia, 19 diarrhoea, 32 nausea, 9 vomiting, 21 asthenia, 18 chills, 53 fatigue, 26 influenza‐like illness, 28 pyrexia, 19 decreased appetite, 21 arthralgia, 18 myalgia, 9 dizziness, 4 dysgeusia, 51 headache, 16 depression, 31 insomnia, 20 cough, 9 dyspnoea, 16 alopecia, 11 dry skin, 20 pruritus, 30 rash

124 out of 130

OPERA 2011a1

Simeprevir

16 anaemia, 26 neutropenia, 20 diarrhoea, 31 nausea, 9 vomiting, 26 asthenia, 6 chills, 35 fatigue, 24 influenza‐like illness, 10 irritability, 21 pyrexia, 7 decreased appetite, 20 arthralgia, 14 myalgia, 4 dizziness, 5 dysgeusia, 42 headache, 14 depression, 12 insomnia, 19 cough, 18 dyspnoea, 16 alopecia, 19 dry skin, 18 pruritus, 9 rash

82 out of 83

4 anaemia, 4 neutropenia, 3 diarrhoea, 4 nausea, 3 vomiting, 7 asthenia, 5 chills, 13 fatigue, 5 influenza‐like illness, 4 irritability, 4 pyrexia, 2 decreased appetite, 3 arthralgia, 8 myalgia, 3 dizziness, 3 dysgeusia, 16 headache, 2 depression, 7 insomnia, 10 cough, 4 dyspnoea, 3 alopecia, 5 dry skin, 7 pruritus, 5 rash

28 out of 28

Manns 2014a

Simeprevir

46 anaemia, 49 neutropenia, 34 diarrhoea, 63 nausea, 17 vomiting, 59 asthenia, 21 chills, 95 fatigue, 66 influenza‐like illness, 80 pyrexia, 46 decreased appetite, 32 arthralgia, 58 myalgia, 21 dizziness, 101 headache, 29 depression, 51 insomnia, 32 cough, 23 dyspnoea, 43 alopecia, 28 dry skin, 65 pruritus, 46 rash

243 out of 257

33 anaemia, 29 neutropenia, 12 diarrhoea, 24 nausea, 7 vomiting, 38 asthenia, 12 chills, 56 fatigue, 35 influenza‐like illness, 53 pyrexia, 21 decreased appetite, 14 arthralgia, 28 myalgia, 9 dizziness, 49 headache, 19 depression, 21 insomnia, 22 cough, 11 dyspnoea, 27 alopecia, 18 dry skin, 34 pruritus, 15 rash

131 out of 134

Pearlman 2015

Simeprevir

1 anaemia, 1 diarrhoea, 6 nausea, 8 fatigue, 1 irritability, 2 myalgia, 7 headache, 3 insomnia, 6 pruritus, 10 rash

46 out of 58

9 anaemia, 5 neutropenia, 2 diarrhoea, 7 nausea, 4 asthenia, 17 fatigue, 6 influenza‐like illness, 3 irritability, 4 myalgia, 8 headache, 6 insomnia, 4 pruritus, 3 rash

22 out of 24

ASPIRE 2014

Simeprevir

76 anaemia, 101 neutropenia, 59 diarrhoea, 95 nausea, 21 vomiting, 84 asthenia, 34 chills, 174 fatigue, 116 influenza‐like illness, 53 irritability, 69 pyrexia, 69 decreased appetite, 50 arthralgia, 64 myalgia, 29 dizziness, 22 dysgeusia, 138 headache, 45 depression, 79 insomnia, 76 cough, 49 dyspnoea, 31 alopecia, 72 dry skin, 135 pruritus, 61 rash

380 out of 396

13 anaemia, 11 neutropenia, 13 diarrhoea, 14 nausea, 5 vomiting, 7 asthenia, 6 chills, 174 fatigue, 13 influenza‐like illness, 7 irritability, 9 pyrexia, 9 decreased appetite, 9 arthralgia, 12 myalgia, 6 dizziness, 3 dysgeusia, 24 headache, 6 depression, 9 insomnia, 8 cough, 4 dyspnoea, 5 alopecia, 10 dry skin, 11 pruritus, 9 rash

63 out of 66

Jacobson 2014

Sofosbuvir

19 diarrhoea, 46 nausea, 12 vomiting, 91 fatigue, 19 irritability, 7 decreased appetite, 16 arthralgia, 19 dizziness, 43 headache, 15 depression, 39 insomnia, 11 cough, 19 dyspnoea, 23 pruritus, 18 rash

184 out of 207

4 diarrhoea, 13 nausea, 5 vomiting, 17 fatigue, 1 irritability, 7 decreased appetite, 1 arthralgia, 5 dizziness, 14 headache, 1 depression, 3 insomnia, 2 cough, 1 dyspnoea, 6 pruritus, 6 rash

55 out of 71

Lawitz 2013a1

Sofosbuvir

19 anaemia, 23 neutropenia, 18 diarrhoea, 38 nausea, 12 vomiting, 2 asthenia, 37 chills, 64 fatigue, 15 irritability, 22 pyrexia, 11 decreased appetite, 10 arthralgia, 17 myalgia, 11 dizziness, 9 dysgeusia, 37 headache, 12 depression, 24 insomnia, 14 cough, 11 dyspnoea, 10 alopecia, 12 dry skin, 13 pruritus, 29 rash

117 out of 120

7 anaemia, 5 neutropenia, 2 diarrhoea, 9 nausea, 2 vomiting, 1 asthenia, 10 chills, 16 fatigue, 5 irritability, 2 pyrexia, 4 decreased appetite, 5 arthralgia, 6 myalgia, 3 dizziness, 15 headache, 3 depression, 9 insomnia, 3 cough, 4 dyspnoea, 2 alopecia, 3 dry skin, 3 pruritus, 4 rash

26 out of 26

)FISSION 2013

Sofosbuvir

20 anaemia, 23 diarrhoea, 46 nausea, 17 vomiting, 7 chills, 92 fatigue, 7 influenza‐like illness, 25 irritability, 6 pyrexia, 17 decreased appetite, 15 arthralgia, 21 myalgia, 27 dizziness, 64 headache, 14 depression, 31 insomnia, 19 cough, 18 dyspnoea, 12 alopecia, 11 dry skin, 19 pruritus, 23 rash

219 out of 256

28 anaemia, 30 neutropenia, 45 diarrhoea, 70 nausea, 23 vomiting, 44 chills, 134 fatigue, 44 influenza‐like illness, 40 irritability, 33 pyrexia, 44 decreased appetite, 35 arthralgia, 40 myalgia, 33 dizziness, 108 headache, 34 depression, 71 insomnia, 21 cough, 20 dyspnoea, 24 alopecia, 23 dry skin, 42 pruritus, 43 rash

233 out of 243

Rodriguez‐Torres 2013

Sofosbuvir

7 anaemia, 17 nausea, 3 vomiting, 22 fatigue, 4 pyrexia, 7 decreased appetite, 12 arthralgia, 7 myalgia, 6 dizziness, 15 headache, 4 depression, 7 insomnia, 9 pruritus

45 out of 49

1 anaemia, 5 nausea, 2 chills, 6 fatigue, 1 pyrexia, 1 decreased appetite, 1 myalgia, 2 dizziness, 2 headache, 2 insomnia,1 pruritus

13 out of 14

Feld 2015

Sofosbuvir/velpatasvir

48 diarrhoea, 75 nausea, 41 asthenia, 126 fatigue, 40 arthralgia, 25 myalgia, 182 headache, 50 insomnia, 39 cough

485 out of 624

8 diarrhoea, 13 nausea, 9 asthenia, 23 fatigue, 9 arthralgia, 6 myalgia, 33 headache, 11 insomnia, 4 cough

89 out of 116

3Benhamou 2013a1

Telaprevir

1 diarrhoea, 4 nausea, 1 vomiting, 6 asthenia, 4 fatigue, 10 influenza‐like illness, 3 headache, 2 insomnia, 1 dyspnoea, 1 dry skin, 3 pruritus

16 out of 16

1 anaemia, 1 neutropenia, 3 asthenia, 4 influenza‐like illness, 1 decreased appetite, 1 headache, 1 insomnia, 1 cough, 1 dry skin, 2 pruritus, 2 rash

8 out of 8

Forestier 2007

Telaprevir

2 diarrhoea, 3 nausea, 1 chills, 5 myalgia, 2 dizziness, 5 headache, 3 dry skin, 3 rash

14 out of 16

1 diarrhoea, 1 nausea, 1 asthenia, 2 chills, 2 myalgia, 1 dizziness, 2 headache, 1 dry skin

4 out of 4

1Foster 2011a1

Telaprevir

1 anaemia, 4 diarrhoea, 8 nausea, 5 vomiting, 9 asthenia, 1 chills, 5 fatigue, 11 influenza‐like illness, 1 irritability, 2 pyrexia, 1 arthralgia, 4 myalgia, 3 dizziness, 5 headache, 1 depression, 2 insomnia, 1 cough, 2 dyspnoea, 1 alopecia, 3 dry skin, 11 pruritus, 5 rash

26 out of 31

1 neutropenia, 1 diarrhoea, 1 nausea, 5 asthenia, 3 chills, 2 fatigue, 7 influenza‐like illness, 5 pyrexia, 3 myalgia, 1 dysgeusia, 6 headache, 1 depression, 2 insomnia, 2 cough, 2 dyspnoea, 1 dry skin, 2 pruritus

16 out of 18

Hezode 2009

Telaprevir

44 anaemia, 11 neutropenia, 66 diarrhoea, 102 nausea, 22 vomiting, 110 asthenia, 12 chills, 70 fatigue, 92 influenza‐like illness, 22 irritability, 44 pyrexia, 30 decreased appetite, 36 arthralgia, 35 myalgia, 12 dizziness, 20 dysgeusia, 105 headache, 51 depression, 62 insomnia, 37 cough, 50 dyspnoea, 29 alopecia, 64 dry skin, 139 pruritus, 71 rash

240 out of 241

14 anaemia, 14 neutropenia, 23 diarrhoea, 33 nausea, 12 vomiting, 26 asthenia, 10 chills, 30 fatigue, 43 influenza‐like illness, 11 irritability, 19 pyrexia, 16 decreased appetite, 14 arthralgia, 17 myalgia, 8 dizziness, 3 dysgeusia, 37 headache, 19 depression, 32 insomnia, 21 cough, 13 dyspnoea, 17 alopecia, 29 dry skin, 29 pruritus, 22 rash

81 out of 82

Jacobson 2010

Telaprevir

276 anaemia, 217 diarrhoea, 302 nausea, 418 fatigue, 203 pyrexia, 304 headache, 233 insomnia, 346 pruritus, 262 rash

723 out of 727

70 anaemia, 80 diarrhoea, 112 nausea, 206 fatigue, 87 pyrexia, 142 headache, 111 insomnia, 131 pruritus, 88 rash

354 out of 361

McHutchison 2009

Telaprevir

58 anaemia, 30 neutropenia, 64 diarrhoea, 93 nausea, 38 vomiting, 29 chills, 127 fatigue, 75 influenza‐like illness, 23 irritability, 33 pyrexia, 22 decreased appetite, 34 arthralgia, 27 myalgia, 41 dizziness, 16 dysgeusia, 80 headache, 34 depression, 68 insomnia, 36 cough, 25 dyspnoea, 21 alopecia, 28 dry skin, 74 pruritus, 62 rash

175 out of 175

20 anaemia, 18 neutropenia, 21 diarrhoea, 22 nausea, 9 vomiting, 14 chills, 57 fatigue, 32 influenza‐like illness, 22 irritability, 22 pyrexia, 9 decreased appetite, 16 arthralgia, 18 myalgia, 14 dizziness, 8 dysgeusia, 45 headache, 13 depression, 29 insomnia, 14 cough, 11 dyspnoea, 8 alopecia, 19 dry skin, 17 pruritus, 20 rash

75 out of 75

McHutchison 2010

Telaprevir

69 anaemia, 31 neutropenia, 115 diarrhoea, 122 nausea, 37 vomiting, 57 chills, 197 fatigue, 93 influenza‐like illness, 63 irritability, 59 pyrexia, 20 decreased appetite, 51 arthralgia, 60 myalgia, 47 dizziness, 130 headache, 43 depression, 83 insomnia, 46 cough, 27 dyspnoea, 54 alopecia, 32 dry skin, 129 pruritus, 126 rash

329 out of 339

9 anaemia, 7 neutropenia, 22 diarrhoea, 39 nausea, 13 vomiting, 15 chills, 64 fatigue, 36 influenza‐like illness, 25 irritability, 14 pyrexia, 12 decreased appetite, 21 arthralgia, 21 myalgia, 18 dizziness, 41 headache, 19 depression, 19 insomnia, 20 cough, 9 dyspnoea, 13 alopecia, 7 dry skin, 17 pruritus, 20 rash

111 out of 114

Sulkowski 2013a

Telaprevir

35 anaemia, 26 neutropenia, 34 diarrhoea, 44 nausea, 27 vomiting, 22 asthenia, 15 chills, 50 fatigue, 24 influenza‐like illness, 18 irritability, 34 pyrexia, 30 decreased appetite, 9 arthralgia, 20 myalgia, 19 dizziness, 18 dysgeusia, 38 headache, 11 depression, 25 insomnia, 15 cough, 5 dyspnoea, 18 alopecia, 11 dry skin, 30 pruritus, 12 rash

38 out of 38

8 anaemia, 2 neutropenia, 6 diarrhoea, 11 nausea, 5 vomiting, 9 asthenia, 5 chills, 12 fatigue, 13 influenza‐like illness, 5 irritability, 7 pyrexia, 6 decreased appetite, 2 arthralgia, 6 myalgia, 2 dizziness, 5 dysgeusia, 6 headache, 4 depression, 9 insomnia, 6 cough, 2 dyspnoea, 6 alopecia, 3 dry skin, 3 pruritus

22 out of 22

Zeuzem 2011a

Telaprevir

171 anaemia, 73 neutropenia, 135 diarrhoea, 181 nausea, 68 vomiting, 111 asthenia, 73 chills, 276 fatigue, 179 influenza‐like illness, 74 irritability, 130 pyrexia, 40 decreased appetite, 67 arthralgia, 87 myalgia, 47 dizziness, 65 dysgeusia, 221 headache, 59 depression, 152 insomnia, 128 cough, 82 dyspnoea, 78 alopecia, 97 dry skin, 270 pruritus, 194 rash

517 out of 530

19 anaemia, 14 neutropenia, 18 diarrhoea, 31 nausea, 11 vomiting, 38 asthenia, 73 chills, 53 fatigue, 33 influenza‐like illness, 21 irritability, 36 pyrexia, 9 decreased appetite, 20 arthralgia, 24 myalgia, 7 dizziness, 8 dysgeusia, 49 headache, 19 depression, 34 insomnia, 26 cough, 17 dyspnoea, 17 alopecia, 21 dry skin, 36 pruritus, 25 rash

126 out of 132

Lawitz 2013a1

Vaniprevir

6 anaemia, 7 neutropenia, 16 diarrhoea, 26 nausea, 16 vomiting, 13 asthenia, 5 chills, 17 fatigue, 17influenza‐like illness, 4 irritability, 8 pyrexia, 13 decreased appetite, 8 arthralgia, 3 myalgia, 5 dizziness, 2 dysgeusia, 26 headache, 8 depression, 15 insomnia, 8 cough, 7 dyspnoea, 6 alopecia, 6 dry skin, 9 pruritus, 10 rash

71 out of 75

3 anaemia, 2 neutropenia, 4 diarrhoea, 6 nausea, 4 asthenia, 2 chills, 7 fatigue, 4 influenza‐like illness, 3 irritability, 2 pyrexia, 2 decreased appetite, 2 arthralgia, 3 myalgia, 7 headache, 3 depression, 2 insomnia, 3 cough, 5 dyspnoea, 3 alopecia, 6 dry skin, 4 pruritus, 4 rash

19 out of 19

Manns 2012a1

Vaniprevir

6 anaemia, 7 neutropenia, 16 diarrhoea, 26 nausea, 16 vomiting, 13 asthenia, 5 chills, 17 fatigue, 17 influenza‐like illness, 4 irritability, 8 pyrexia, 13 decreased appetite, 8 arthralgia, 3 myalgia, 5 dizziness, 2 dysgeusia, 26 headache, 8 depression, 15 insomnia, 8 cough, 7 dyspnoea, 6 alopecia, 6 dry skin, 9 pruritus, 10 rash

71 out of 75

3 anaemia, 2 neutropenia, 4 diarrhoea, 6 nausea, 4 asthenia, 2 chills, 7 fatigue, 4 influenza‐like illness, 3 irritability, 2 pyrexia, 2 decreased appetite, 2 arthralgia, 3 myalgia, 7 headache, 3 depression, 2 insomnia, 3 cough, 5 dyspnoea, 3 alopecia, 6 dry skin, 4 pruritus, 4 rash

19 out of 19

Rodriguez‐Torres 2014a1

Vaniprevir

43 anaemia, 34 neutropenia, 97 diarrhoea, 110 nausea, 59 vomiting, 50 asthenia, 16 chills, 92 fatigue, 54 influenza‐like illness, 24 irritability, 37 pyrexia, 40 decreased appetite, 37 arthralgia, 38 myalgia, 23 dizziness, 16 dysgeusia, 92 headache, 32 depression, 40 insomnia, 54 cough, 30 dyspnoea, 35 alopecia, 37 dry skin, 75 pruritus, 43 rash

225 out of 229

8 anaemia, 3 neutropenia, 9 diarrhoea, 10 nausea, 3 vomiting, 11 asthenia, 1 chills, 18 fatigue, 12 influenza‐like illness, 8 irritability, 14 pyrexia, 5 decreased appetite, 11 arthralgia, 12 myalgia, 6 dizziness, 3 dysgeusia, 20 headache, 3 depression, 14 insomnia, 14 cough, 8 dyspnoea, 5 alopecia, 10 dry skin, 14 pruritus, 10 rash

55 out of 56

Lawitz 2013c

Vaniprevir

43 anaemia, 34 neutropenia, 97 diarrhoea, 110 nausea, 59 vomiting, 50 asthenia, 16 chills, 92 fatigue, 54 influenza‐like illness, 24 irritability, 37 pyrexia, 40 decreased appetite, 37 arthralgia, 38 myalgia, 23 dizziness, 16 dysgeusia, 92 headache, 32 depression, 40 insomnia, 54 cough, 30 dyspnoea, 35 alopecia, 37 dry skin, 75 pruritus, 43 rash

225 out of 229

8 anaemia, 3 neutropenia, 9 diarrhoea, 10 nausea, 3 vomiting, 11 asthenia, 1 chills, 18 fatigue, 12 influenza‐like illness, 8 irritability, 14 pyrexia, 5 decreased appetite, 11 arthralgia, 12 myalgia, 6 dizziness, 3 dysgeusia, 20 headache, 3 depression, 14 insomnia, 14 cough, 8 dyspnoea, 5 alopecia, 10 dry skin, 14 pruritus, 10 rash

55 out of 56

Cooper 2009

VCH‐759

18 diarrhoea, 3 nausea, 4 vomiting, 1 chills, 5 fatigue, 8 headache

20 out of 23

5 diarrhoea, 1 nausea, 2 headache

6 out of 9

Lawitz 2015

Velpatasvir

2 diarrhoea, 3 nausea, 4 vomiting, 6 headache, 2 cough

18 out of 70

None reported

3 out of 17

Figuras y tablas -
Table 3. Non‐serious adverse events
Comparison 1. DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Hepatitis C‐related morbidity or all‐cause mortality Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

2 Hepatitis C‐related morbidity or all‐cause mortality ‐ bias risk Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

2.1 Trials at high risk of bias

71

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.2 Trials at low risk of bias

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to type of DAA Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

3.1 ABT‐072

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.2 ACH‐2684

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.3 Alisporivir

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.4 ALS‐2200

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.5 Asunaprevir

6

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.6 Balapiravir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.7 Beclabuvir

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.8 BILB‐1941

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.9 BIT‐225

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.10 Boceprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.11 Ciluprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.12 Daclatasvir

14

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.13 Danoprevir

9

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.14 Dasabuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.15 Deleobuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.16 Faldaprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.17 Filibuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.18 Grazoprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.19 GS‐6620

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.20 GS‐9256

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.21 GS‐9451

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.22 GS‐9669

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.23 GS‐9851

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.24 GS‐9857

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.25 GSK2336805

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.26 GSK2878175

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.27 IDX‐184

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.28 INX‐08189

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.29 Ledispasvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.30 Mericitabine

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.31 Narlaprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.32 Nesbuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.33 Odalasavir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.34 Ombitasvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.35 Paritaprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.36 PHX1766

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.37 PPI‐461

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.38 PSI‐352938

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.39 Samatasvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.40 Setrobuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.41 Simeprevir

14

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.42 Sofosbuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.43 Sovaprevir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.44 Tegobuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.45 Telaprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.46 Valopicitabine

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.47 Vaniprevir

9

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.48 VCH‐759

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.49 VCH‐916

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.50 Velpatasvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.51 VX‐222

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.52 Mixed

4

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to group of DAA Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

4.1 Cyclophilin

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.2 NS3/NS4A inhibitors

41

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.3 NS5B inhibitors (NPI)

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.4 NS5B inhibitors (NNPI)

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.5 NS5A inhibitors

18

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.6 VPU‐ion channel inhibitors

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.7 Mixed

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to HIV‐infection Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

5.1 With HIV‐infection

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.2 Without HIV‐infection

69

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.3 Mixed (with and without HIV‐infection)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.4 Unclear

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to comorbidity Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

6.1 With comorbidity

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6.2 Without comorbidity

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6.3 Unclear

71

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to viral genotype Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

7.1 Genotype 1

57

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.2 Genotype 2

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.3 Genotype 3

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.4 Genotype 4

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.5 Mixed

14

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to human genotype (IL28b) Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

8.1 IL28b (CC)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.2 IL28B (CT)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.3 IL28B (TT)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.4 IL28B (CT + TT)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.5 Mixed

71

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to Asian‐region Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

9.1 From Asian region

8

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.2 Not from Asian region

52

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.3 Mixed

11

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.4 Unclear

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to specific ethnicities Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

10.1 White

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.2 Black

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.3 Hispanic

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.4 Mixed

70

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.5 Unclear

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to reaching planned sample size Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

11.1 Trials reaching planned sample size

10

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.2 Trials not reaching planned sample size

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.3 Unclear

58

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to prior treatment Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

12.1 Treatment‐naive

47

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.2 Treatment‐experienced

16

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.3 Mixed

8

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.4 Unclear

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to interferon Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

13.1 Trials where both groups received interferon

52

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13.2 Trials where neither group received interferon

19

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to ribavirin Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

14.1 Trials where both groups received ribavirin

52

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14.2 Trials where neither group received ribavirin

19

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to chronic kidney disease Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

15.1 With chronic kidney disease

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.2 Without chronic kidney disease

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.3 Unclear

71

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to cryoglobulinaemia Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

16.1 With cryoglobulinaemia

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.2 Without cryoglobulinaemia

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.3 Unclear

71

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

17 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to DAA group as co‐intervention Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

17.1 Trials where DAA were used as co‐intervention

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

17.2 Trials where DAA were not a co‐intervention

69

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

18 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to median dose Show forest plot

71

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

18.1 Over or equal to median dose

41

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

18.2 Under median dose

27

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

18.3 Not available

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 1. DAA on or on the way to the market versus placebo/no intervention (morbidity or all cause mortality analyses)
Comparison 2. DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Serious adverse events Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

2 Serious adverse events ‐ bias risk Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

2.1 Trials at high risk of bias

101

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.2 Trials at low risk of bias

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3 Serious adverse events ‐ according to type of DAA Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

3.1 ABT‐072

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.2 ACH‐2684

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.3 Alisporivir

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.4 ALS‐2200

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.5 Asunaprevir

6

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.6 Balapiravir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.7 Beclabuvir

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.8 BILB‐1941

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.9 BIT‐225

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.10 Boceprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.11 Ciluprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.12 Daclatasvir

14

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.13 Danoprevir

9

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.14 Dasabuvir

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.15 Deleobuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.16 Faldaprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.17 Filibuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.18 Grazoprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.19 GS‐6620

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.20 GS‐9256

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.21 GS‐9451

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.22 GS‐9669

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.23 GS‐9851

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.24 GS‐9857

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.25 GSK2336805

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.26 GSK2878175

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.27 IDX‐184

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.28 INX‐08189

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.29 Ledispasvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.30 Mericitabine

7

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.31 Narlaprevir

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.32 Nesbuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.33 Odalasavir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.34 Ombitasvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.35 Paritaprevir

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.36 PHX1766

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.37 PPI‐461

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.38 PSI‐352938

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.39 Samatasvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.40 Setrobuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.41 Simeprevir

18

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.42 Sofosbuvir

4

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.43 Sovaprevir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.44 Tegobuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.45 Telaprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.46 Valopicitabine

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.47 Vaniprevir

10

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.48 VCH‐759

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.49 VCH‐916

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.50 Velpatasvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.51 VX‐222

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.52 Mixed

7

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4 Serious adverse events ‐ according to group of DAA Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

4.1 Cyclophilin

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.2 NS3/NS4A inhibitors

56

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.3 NS5B inhibitors (NPI)

8

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.4 NS5B inhibitors (NNPI)

5

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.5 NS5A inhibitors

25

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.6 VPU‐ion channel inhibitors

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.7 Mixed

4

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5 Serious adverse events ‐ according to HIV‐infection Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

5.1 With HIV‐infection

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.2 Without HIV‐infection

94

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.3 Mixed (with and without HIV‐infection)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.4 Unclear

7

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6 Serious adverse events ‐ according to comorbidity Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

6.1 With comorbidity

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6.2 Without comorbidity

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6.3 Unclear

101

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7 Serious adverse events ‐ according to viral genotype Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

7.1 Genotype 1

84

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.2 Genotype 2

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.3 Genotype 3

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.4 Genotype 4

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.5 Mixed

17

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8 Serious adverse events ‐ according to human genotype (IL28b) Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

8.1 IL28b (CC)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.2 IL28B (CT)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.3 IL28B (TT)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.4 IL28B (CT + TT)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.5 Mixed

101

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9 Serious adverse events ‐ according to Asian‐region Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

9.1 From Asian region

10

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.2 Not from Asian region

76

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.3 Mixed

11

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.4 Unclear

4

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10 Serious adverse events ‐ according to specific ethnicities Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

10.1 White

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.2 Black

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.3 Hispanic

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.4 Mixed

101

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.5 Unclear

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11 Serious adverse events ‐ according to reaching planned sample size Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

11.1 Trials reaching planned sample size

15

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.2 Trials not reaching planned sample size

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.3 Unclear

83

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12 Serious adverse events ‐ according to prior treatment Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

12.1 Treatment‐naive

72

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.2 Treatment‐experienced

19

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.3 Mixed

9

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.4 Unclear

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13 Serious adverse events ‐ according to interferon Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

13.1 Trials where both groups received interferon

69

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13.2 Trials where neither group received interferon

29

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13.3 Unclear

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14 Serious adverse events ‐ according to ribavirin Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

14.1 Trials where both groups received ribavirin

73

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14.2 Trials where neither group received ribavirin

27

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14.3 Unclear

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15 Serious adverse events ‐ according to chronic kidney disease Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

15.1 With chronic kidney disease

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.2 Without chronic kidney disease

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.3 Unclear

101

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16 Serious adverse events ‐ according to cryoglobulinaemia Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

16.1 With cryoglobulinaemia

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.2 Without cryoglobulinaemia

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.3 Unclear

101

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

17 Serious adverse events ‐ according to DAA group as co‐intervention Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

17.1 Trials where DAA were used as co‐intervention

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

17.2 Trials where DAA were not a co‐intervention

99

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

18 Serious adverse events ‐ according to median dose Show forest plot

101

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

18.1 Over or equal to median dose

58

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

18.2 Under median dose

37

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

18.3 Not available

6

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 2. DAA on or on the way to the market versus placebo/no intervention (serious adverse events analyses)
Comparison 3. DAA on or on the way to the market versus placebo/no intervention (sustained virological response)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Without sustained virological response Show forest plot

61

7115

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.37, 0.52]

2 Without sustained virological response ‐ bias risk Show forest plot

61

7115

Odds Ratio (M‐H, Fixed, 95% CI)

0.24 [0.22, 0.27]

2.1 Trials at high risk of bias

61

7115

Odds Ratio (M‐H, Fixed, 95% CI)

0.24 [0.22, 0.27]

2.2 Trials at low risk of bias

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3 Without sustained virological response ‐ according to type of DAA Show forest plot

61

7115

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.37, 0.52]

3.1 ABT‐072

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.2 ACH‐2684

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.3 Alisporivir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.4 ALS‐2200

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.5 Asunaprevir

4

285

Risk Ratio (M‐H, Random, 95% CI)

0.49 [0.29, 0.85]

3.6 Balapiravir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.7 Beclabuvir

2

39

Risk Ratio (M‐H, Random, 95% CI)

0.78 [0.43, 1.40]

3.8 BILB‐1941

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.9 BIT‐225

1

23

Risk Ratio (M‐H, Random, 95% CI)

0.27 [0.03, 2.51]

3.10 Boceprevir

1

229

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.29, 0.61]

3.11 Ciluprevir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.12 Daclatasvir

7

619

Risk Ratio (M‐H, Random, 95% CI)

0.60 [0.50, 0.73]

3.13 Danoprevir

5

642

Risk Ratio (M‐H, Random, 95% CI)

0.38 [0.28, 0.51]

3.14 Dasabuvir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.15 Deleobuvir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.16 Faldaprevir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.17 Filibuvir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.18 Grazoprevir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.19 GS‐6620

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.20 GS‐9256

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.21 GS‐9451

1

329

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.26, 0.67]

3.22 GS‐9669

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.23 GS‐9851

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.24 GS‐9857

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.25 GSK2336805

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.26 GSK2878175

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.27 IDX‐184

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.28 INX‐08189

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.29 Ledispasvir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.30 Mericitabine

4

725

Risk Ratio (M‐H, Random, 95% CI)

0.78 [0.49, 1.27]

3.31 Narlaprevir

2

40

Risk Ratio (M‐H, Random, 95% CI)

0.69 [0.43, 1.09]

3.32 Nesbuvir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.33 Odalasavir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.34 Ombitasvir

1

37

Risk Ratio (M‐H, Random, 95% CI)

0.64 [0.39, 1.07]

3.35 Paritaprevir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.36 PHX1766

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.37 PPI‐461

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.38 PSI‐352938

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.39 Samatasvir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.40 Setrobuvir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.41 Simeprevir

19

2898

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.33, 0.46]

3.42 Sofosbuvir

3

181

Risk Ratio (M‐H, Random, 95% CI)

0.34 [0.20, 0.58]

3.43 Sovaprevir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.44 Tegobuvir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.45 Telaprevir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.46 Valopicitabine

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.47 Vaniprevir

9

333

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.25, 0.43]

3.48 VCH‐759

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.49 VCH‐916

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.50 Velpatasvir

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.51 VX‐222

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3.52 Mixed

2

735

Risk Ratio (M‐H, Random, 95% CI)

0.06 [0.00, 7.05]

4 Without sustained virological response ‐ according to group of DAA Show forest plot

61

7115

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.37, 0.52]

4.1 Cyclophilin

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

4.2 NS3/NS4A inhibitors

41

4756

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.36, 0.46]

4.3 NS5B inhibitors (NPI)

7

906

Risk Ratio (M‐H, Random, 95% CI)

0.57 [0.36, 0.90]

4.4 NS5B inhibitors (NNPI)

2

39

Risk Ratio (M‐H, Random, 95% CI)

0.78 [0.43, 1.40]

4.5 NS5A inhibitors

9

686

Risk Ratio (M‐H, Random, 95% CI)

0.59 [0.49, 0.69]

4.6 VPU‐ion channel inhibitors

1

23

Risk Ratio (M‐H, Random, 95% CI)

0.27 [0.03, 2.51]

4.7 Mixed

1

705

Risk Ratio (M‐H, Random, 95% CI)

0.01 [0.00, 0.02]

5 Without sustained virological response ‐ according to HIV‐infection Show forest plot

61

7115

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.37, 0.52]

5.1 With HIV‐infection

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

5.2 Without HIV‐infection

58

6726

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.37, 0.52]

5.3 Mixed (with and without HIV‐infection)

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

5.4 Unclear

3

389

Risk Ratio (M‐H, Random, 95% CI)

0.50 [0.35, 0.72]

6 Without sustained virological response ‐ according to comorbidity Show forest plot

61

7115

Odds Ratio (M‐H, Fixed, 95% CI)

0.24 [0.22, 0.27]

6.1 With comorbidity

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6.2 Without comorbidity

61

7115

Odds Ratio (M‐H, Fixed, 95% CI)

0.24 [0.22, 0.27]

6.3 Unclear

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7 Without sustained virological response ‐ according to viral genotype Show forest plot

58

7098

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.36, 0.51]

7.1 Genotype 1

54

5984

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.37, 0.50]

7.2 Genotype 2

3

185

Risk Ratio (M‐H, Random, 95% CI)

0.14 [0.01, 3.21]

7.3 Genotype 3

2

80

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.43, 1.43]

7.4 Genotype 4

5

226

Risk Ratio (M‐H, Random, 95% CI)

0.10 [0.02, 0.68]

7.5 Genotype 6

1

49

Risk Ratio (M‐H, Random, 95% CI)

0.01 [0.00, 0.20]

7.6 Mixed

2

574

Risk Ratio (M‐H, Random, 95% CI)

0.92 [0.52, 1.62]

8 Without sustained virological response ‐ according to human genotype (IL28b) Show forest plot

58

6745

Risk Ratio (M‐H, Random, 95% CI)

0.46 [0.40, 0.54]

8.1 IL28b (CC)

25

1444

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.29, 0.61]

8.2 IL28B (CT)

10

1304

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.42, 0.66]

8.3 IL28B (TT)

10

359

Risk Ratio (M‐H, Random, 95% CI)

0.54 [0.44, 0.67]

8.4 IL28B (CT + TT)

14

1798

Risk Ratio (M‐H, Random, 95% CI)

0.37 [0.23, 0.57]

8.5 Unclear

7

147

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.33, 0.68]

8.6 Mixed

26

1693

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.40, 0.63]

9 Without sustained virological response ‐ according to Asian‐region Show forest plot

61

7115

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.37, 0.52]

9.1 From Asian region

10

1128

Risk Ratio (M‐H, Random, 95% CI)

0.34 [0.28, 0.42]

9.2 Not from Asian region

42

4910

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.43, 0.60]

9.3 Mixed

7

1010

Risk Ratio (M‐H, Random, 95% CI)

0.19 [0.03, 1.17]

9.4 Unclear

2

67

Risk Ratio (M‐H, Random, 95% CI)

0.53 [0.35, 0.79]

10 Without sustained virological response ‐ according to specific ethnicities Show forest plot

61

7115

Odds Ratio (M‐H, Fixed, 95% CI)

0.24 [0.22, 0.27]

10.1 White

2

412

Odds Ratio (M‐H, Fixed, 95% CI)

0.24 [0.15, 0.38]

10.2 Black

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.3 Hispanic

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.4 Mixed

48

5384

Odds Ratio (M‐H, Fixed, 95% CI)

0.23 [0.20, 0.27]

10.5 Unclear

9

862

Odds Ratio (M‐H, Fixed, 95% CI)

0.28 [0.20, 0.39]

10.6 Asian

2

457

Odds Ratio (M‐H, Fixed, 95% CI)

0.38 [0.23, 0.63]

11 Without sustained virological response ‐ according to reaching planned sample size Show forest plot

61

7115

Odds Ratio (M‐H, Fixed, 95% CI)

0.24 [0.22, 0.27]

11.1 Trials reaching planned sample size

13

3071

Odds Ratio (M‐H, Fixed, 95% CI)

0.21 [0.18, 0.25]

11.2 Trials not reaching planned sample size

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.3 Unclear

48

4044

Odds Ratio (M‐H, Fixed, 95% CI)

0.28 [0.23, 0.33]

12 Without sustained virological response ‐ according to prior treatment Show forest plot

61

7115

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.37, 0.52]

12.1 Treatment‐naive

44

4777

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.41, 0.56]

12.2 Treatment‐experienced

13

1274

Risk Ratio (M‐H, Random, 95% CI)

0.50 [0.36, 0.69]

12.3 Mixed

4

1064

Risk Ratio (M‐H, Random, 95% CI)

0.15 [0.02, 0.96]

12.4 Unclear

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

13 Without sustained virological response ‐ according to interferon Show forest plot

61

7115

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.37, 0.52]

13.1 Trials where both groups received interferon

57

6229

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.41, 0.54]

13.2 Trials where neither group received interferon

2

735

Risk Ratio (M‐H, Random, 95% CI)

0.06 [0.00, 7.05]

13.3 Trials where only the experimental group received interferon

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

13.4 Trials where only the control group received interferon

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

13.5 Mixed

2

151

Risk Ratio (M‐H, Random, 95% CI)

0.58 [0.15, 2.30]

14 Without sustained virological response ‐ according to ribavirin Show forest plot

61

7115

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.37, 0.52]

14.1 Trials where both groups received ribavirin

60

6410

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.41, 0.55]

14.2 Trials where neither group received ribavirin

1

705

Risk Ratio (M‐H, Random, 95% CI)

0.01 [0.00, 0.02]

14.3 Trials where only the experimental group received ribavirin

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

14.4 Trials where only the control group received ribavirin

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

15 Without sustained virological response ‐ according to chronic kidney disease Show forest plot

61

7115

Odds Ratio (M‐H, Fixed, 95% CI)

0.24 [0.22, 0.27]

15.1 With chronic kidney disease

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.2 Without chronic kidney disease

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.3 Unclear

61

7115

Odds Ratio (M‐H, Fixed, 95% CI)

0.24 [0.22, 0.27]

16 Without sustained virological response ‐ according to cryoglobulinaemia Show forest plot

61

7115

Odds Ratio (M‐H, Fixed, 95% CI)

0.24 [0.22, 0.27]

16.1 With cryoglobulinaemia

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.2 Without cryoglobulinaemia

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.3 Unclear

61

7115

Odds Ratio (M‐H, Fixed, 95% CI)

0.24 [0.22, 0.27]

17 Without sustained virological response ‐ according to DAA group as co‐intervention Show forest plot

61

7115

Odds Ratio (M‐H, Fixed, 95% CI)

0.24 [0.22, 0.27]

17.1 Trials where DAA were used as co‐intervention

3

480

Odds Ratio (M‐H, Fixed, 95% CI)

0.42 [0.27, 0.66]

17.2 Trials where DAA were not a co‐intervention

58

6635

Odds Ratio (M‐H, Fixed, 95% CI)

0.23 [0.21, 0.26]

18 Without sustained virological response ‐ 'Best‐worst case' scenario Show forest plot

61

7294

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.34, 0.49]

19 Without sustained virological response ‐ 'Worst‐best case' scenario Show forest plot

61

7294

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.43, 0.60]

20 Without sustained virological response ‐ according to median dose Show forest plot

61

7115

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.37, 0.52]

20.1 Over or equal to median dose

34

4154

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.32, 0.53]

20.2 Under median dose

23

2086

Risk Ratio (M‐H, Random, 95% CI)

0.46 [0.39, 0.55]

20.3 Not available

4

875

Risk Ratio (M‐H, Random, 95% CI)

0.62 [0.26, 1.47]

Figuras y tablas -
Comparison 3. DAA on or on the way to the market versus placebo/no intervention (sustained virological response)
Comparison 4. Danoprevir versus placebo/no intervention

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Hepatitis C‐related morbidity or all‐cause mortality Show forest plot

9

781

Odds Ratio (M‐H, Fixed, 95% CI)

0.56 [0.06, 5.19]

2 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to dose Show forest plot

9

781

Odds Ratio (M‐H, Fixed, 95% CI)

0.56 [0.06, 5.19]

2.1 Over or equal to median dose

6

606

Odds Ratio (M‐H, Fixed, 95% CI)

0.56 [0.06, 5.19]

2.2 Under median dose

3

175

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.3 Not available

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3 Serious adverse events Show forest plot

9

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

4 Serious adverse events ‐ according to median dose Show forest plot

9

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

4.1 Over or equal to median dose

6

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.2 Under median dose

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.3 Not available

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5 Without sustained virological response Show forest plot

5

642

Odds Ratio (M‐H, Fixed, 95% CI)

0.19 [0.12, 0.32]

6 Without sustained virological response ‐ according to median dose Show forest plot

5

642

Odds Ratio (M‐H, Fixed, 95% CI)

0.19 [0.12, 0.32]

6.1 Over or equal to median dose

4

537

Odds Ratio (M‐H, Fixed, 95% CI)

0.18 [0.11, 0.32]

6.2 Under median dose

1

105

Odds Ratio (M‐H, Fixed, 95% CI)

0.27 [0.07, 0.99]

6.3 Not available

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 4. Danoprevir versus placebo/no intervention
Comparison 5. All DAA versus placebo/no intervention/other medical intervention (morbidity or all‐cause mortality analyses)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Hepatitis C‐related morbidity or all‐cause mortality Show forest plot

95

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

1.1 Trials assessing DAAs on or on the way to the market

71

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.2 Trials assessing DAAs withdrawn from market

22

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.3 Trials using other medical intervention as control group

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.4 Trials using other medical intervention as experimental group

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Hepatitis C‐related morbidity or all‐cause mortality ‐ drugs not discontinued

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.1 Trials assessing discontinued drugs

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.2 Trials assessing drugs still used

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3 Hepatitis C‐related morbidity or all‐cause mortality ‐ bias risk

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.1 Trials with a high risk of bias

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.2 Trials with a low risk of bias

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to type of DAA

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.1 ABT‐072

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.2 ACH‐2684

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.3 Alisporivir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.4 ALS‐2200

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.5 Asunaprevir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.6 Balapiravir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.7 Beclabuvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.8 BILB‐1941

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.9 BIT‐225

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.10 Boceprevir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.11 Ciluprevir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.12 Daclatasvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.13 Danoprevir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.14 Dasabuvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.15 Deleobuvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.16 Faldaprevir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.17 Filibuvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.18 Grazoprevir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.19 GS‐6620

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.20 GS‐9256

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.21 GS‐9451

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.22 GS‐9669

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.23 GS‐9851

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.24 GS‐9857

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.25 GSK2336805

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.26 GSK2878175

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.27 IDX‐184

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.28 INX‐08189

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.29 Ledispasvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.30 Mericitabine

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.31 Narlaprevir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.32 Nesbuvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.33 Odalasavir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.34 Ombitasvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.35 Paritaprevir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.36 PHX1766

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.37 PPI‐461

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.38 PSI‐352938

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.39 Samatasvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.40 Setrobuvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.41 Simeprevir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.42 Sofosbuvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.43 Sovaprevir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.44 Tegobuvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.45 Telaprevir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.46 Valopicitabine

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.47 Vaniprevir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.48 VCH‐759

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.49 VCH‐916

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.50 Velpatasvir

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.51 VX‐222

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.52 Mixed

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to group of DAA

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.1 Cyclophilin

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.2 NS3/NS4A inhibitors

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.3 NS5B inhibitors (NPI)

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.4 NS5B inhibitors (NNPI)

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.5 NS5A inhibitors

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.6 VPU‐ion channel inhibitors

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.7 Mixed

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to HIV‐infection

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6.1 With HIV‐infection

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6.2 Without HIV‐infection

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6.3 Mixed (with and without HIV‐infection)

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6.4 Unclear

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to comorbidity

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.1 With comorbidity

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.2 Without comorbidity

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.3 Unclear

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to viral genotype

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.1 Genotype 1

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.2 Genotype 2

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.3 Genotype 3

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.4 Genotype 4

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.5 Unclear

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to human genotype (IL28b)

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.1 IL28b (CC)

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.2 IL28B (CT)

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.3 IL28B (TT)

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.4 IL28B (CT + TT)

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.5 Unclear

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to Asian‐region

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.1 From Asian region

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.2 Not from Asian region

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.3 Mixed

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.4 Unclear

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to specific ethnicities

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.1 White

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.2 Black

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.3 Hispanic

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.4 Mixed

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.5 Unclear

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to reaching planned sample size

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.1 Trials reaching planned sample size

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.2 Trials not reaching planned sample size

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.3 Unclear

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to prior treatment

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13.1 Treatment‐naive

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13.2 Treatment‐experienced

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13.3 Mixed

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13.4 Unclear

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to interferon

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14.1 Trials where both groups received interferon

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14.2 Trials where neither group received interferon

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14.3 Trials where only the experimental group received interferon

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14.4 Trials where only the control group received interferon

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to ribavirin

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.1 Trials where both groups received ribavirin

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.2 Trials where neither group received ribavirin

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.3 Trials where only the experimental group received ribavirin

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.4 Trials where only the control group received ribavirin

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to chronic kidney disease

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.1 With chronic kidney disease

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.2 Without chronic kidney disease

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.3 Unclear

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

17 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to cryoglobulinaemia

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

17.1 With cryoglobulinaemia

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

17.2 Without cryoglobulinaemia

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

17.3 Unclear

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

18 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to DAA group as co‐intervention

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

18.1 Trials where DAA were used as co‐intervention

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

18.2 Trials where DAA were not a co‐intervention

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 5. All DAA versus placebo/no intervention/other medical intervention (morbidity or all‐cause mortality analyses)
Comparison 6. All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Serious adverse events Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

1.1 Trials assessing DAAs on or on the way to the market

101

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.2 Trials assessing DAAs withdrawn from market

62

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.3 Trials using other medical intervention as control group

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.4 Trials using other medical intervention as experimental group

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Serious adverse events ‐ bias risk Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

2.1 Trials with a high risk of bias

167

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.2 Trials with a low risk of bias

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3 Serious adverse events ‐ according to type of DAA Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

3.1 ABT‐072

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.2 ACH‐2684

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.3 Alisporivir

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.4 ALS‐2200

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.5 Asunaprevir

6

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.6 Balapiravir

9

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.7 Beclabuvir

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.8 BILB‐1941

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.9 BIT‐225

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.10 Boceprevir

13

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.11 Ciluprevir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.12 Daclatasvir

14

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.13 Danoprevir

9

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.14 Dasabuvir

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.15 Deleobuvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.16 Faldaprevir

13

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.17 Filibuvir

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.18 Grazoprevir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.19 GS‐6620

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.20 GS‐9256

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.21 GS‐9451

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.22 GS‐9669

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.23 GS‐9851

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.24 GS‐9857

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.25 GSK2336805

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.26 GSK2878175

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.27 IDX‐184

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.28 INX‐08189

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.29 Ledispasvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.30 Mericitabine

7

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.31 Narlaprevir

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.32 Nesbuvir

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.33 Odalasavir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.34 Ombitasvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.35 Paritaprevir

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.36 PHX1766

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.37 PPI‐461

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.38 PSI‐352938

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.39 Samatasvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.40 Setrobuvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.41 Simeprevir

19

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.42 Sofosbuvir

6

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.43 Sovaprevir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.44 Tegobuvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.45 Telaprevir

13

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.46 Valopicitabine

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.47 Vaniprevir

10

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.48 VCH‐759

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.49 VCH‐916

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.50 Velpatasvir

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.51 VX‐222

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.52 Mixed

8

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4 Serious adverse events ‐ according to group of DAA Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

4.1 Cyclophilin

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.2 NS3/NS4A inhibitors

92

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.3 NS5B inhibitors (NPI)

24

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.4 NS5B inhibitors (NNPI)

14

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.5 NS5A inhibitors

27

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.6 VPU‐ion channel inhibitors

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.7 Mixed

7

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5 Serious adverse events ‐ according to HIV‐infection Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

5.1 With HIV‐infection

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.2 Without HIV‐infection

154

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.3 Mixed (with and without HIV‐infection)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.4 Unclear

11

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6 Serious adverse events ‐ according to comorbidity Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

6.1 With comorbidity

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6.2 Without comorbidity

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6.3 Unclear

167

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7 Serious adverse events ‐ according to viral genotype Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

7.1 Genotype 1

138

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.2 Genotype 2

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.3 Genotype 3

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.4 Genotype 4

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7.5 Mixed

26

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8 Serious adverse events ‐ according to human genotype (IL28b) Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

8.1 IL28b (CC)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.2 IL28B (CT)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.3 IL28B (TT)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.4 IL28B (CT + TT)

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.5 Unclear

79

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.6 Mixed IL28b

88

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9 Serious adverse events ‐ according to Asian‐region Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

9.1 From Asian region

12

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.2 Not from Asian region

119

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.3 Mixed

31

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9.4 Unclear

5

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10 Serious adverse events ‐ according to specific ethnicities Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

10.1 White

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.2 Black

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.3 Hispanic

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.4 Mixed

133

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.5 Unclear

31

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11 Serious adverse events ‐ according to reaching planned sample size

0

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

11.1 Trials reaching planned sample size

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.2 Trials not reaching planned sample size

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11.3 Unclear

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12 Serious adverse events ‐ according to prior treatment Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

12.1 Treatment‐naive

122

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.2 Treatment‐experienced

27

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.3 Mixed

18

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.4 Unclear

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13 Serious adverse events ‐ according to interferon Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

13.1 Trials where both groups received interferon

126

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13.2 Trials where neither group received interferon

40

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13.3 Trials where only the experimental group received interferon

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13.4 Trials where only the control group received interferon

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14 Serious adverse events ‐ according to ribavirin Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

14.1 Trials where both groups received ribavirin

127

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14.2 Trials where neither group received ribavirin

37

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14.3 Trials where only the experimental group received ribavirin

1

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14.4 Trials where only the control group received ribavirin

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15 Serious adverse events ‐ according to chronic kidney disease Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

15.1 With chronic kidney disease

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.2 Without chronic kidney disease

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.3 Unclear

167

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16 Serious adverse events ‐ according to cryoglobulinaemia Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

16.1 With cryoglobulinaemia

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.2 Without cryoglobulinaemia

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.3 Unclear

167

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

17 Serious adverse events ‐ according to DAA group as co‐intervention Show forest plot

167

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

17.1 Trials where DAA were used as co‐intervention

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

17.2 Trials where DAA were not a co‐intervention

165

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 6. All DAA versus placebo/no intervention/other medical intervention (serious adverse events analyses)
Comparison 7. All DAA versus placebo/no intervention/other medical intervention (sustained virological response analyses)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Without sustained virological response Show forest plot

107

17101

Risk Ratio (M‐H, Random, 95% CI)

0.53 [0.48, 0.59]

1.1 Trials assessing DAAs on or on the way to the market

60

6886

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.37, 0.52]

1.2 Trials assessing DAAs withdrawn from market

43

9075

Risk Ratio (M‐H, Random, 95% CI)

0.61 [0.55, 0.69]

1.3 Trials using other medical intervention as control group

3

862

Risk Ratio (M‐H, Random, 95% CI)

0.81 [0.36, 1.82]

1.4 Trials using other medical intervention as experimental group

1

278

Risk Ratio (M‐H, Random, 95% CI)

0.23 [0.17, 0.29]

Figuras y tablas -
Comparison 7. All DAA versus placebo/no intervention/other medical intervention (sustained virological response analyses)
Comparison 8. All DAA versus placebo/no intervention/other medical intervention (quality of life scores)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 SF‐36 physical score Show forest plot

1

215

Mean Difference (IV, Fixed, 95% CI)

‐1.17 [‐3.65, 1.31]

2 SF‐36 mental score Show forest plot

1

215

Mean Difference (IV, Fixed, 95% CI)

1.36 [‐1.53, 4.25]

Figuras y tablas -
Comparison 8. All DAA versus placebo/no intervention/other medical intervention (quality of life scores)
Comparison 9. Daclatasvir versus placebo/no intervention

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Hepatitis C‐related morbidity or all‐cause mortality Show forest plot

14

666

Odds Ratio (M‐H, Fixed, 95% CI)

1.25 [0.06, 26.65]

2 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to dose Show forest plot

14

666

Odds Ratio (M‐H, Fixed, 95% CI)

1.25 [0.06, 26.65]

2.1 Over or equal to median dose

7

374

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.2 Under median dose

7

292

Odds Ratio (M‐H, Fixed, 95% CI)

1.25 [0.06, 26.65]

2.3 Not available

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3 Serious adverse events Show forest plot

13

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

4 Serious adverse events ‐ according to median dose Show forest plot

14

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

4.1 Over or equal to median dose

7

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.2 Under median dose

8

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.3 Not available

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5 Without sustained virological response Show forest plot

7

619

Odds Ratio (M‐H, Fixed, 95% CI)

0.40 [0.27, 0.59]

6 Without sustained virological response ‐ according to median dose Show forest plot

7

619

Odds Ratio (M‐H, Fixed, 95% CI)

0.40 [0.27, 0.59]

6.1 Over or equal to median dose

4

360

Odds Ratio (M‐H, Fixed, 95% CI)

0.43 [0.26, 0.70]

6.2 Under median dose

3

259

Odds Ratio (M‐H, Fixed, 95% CI)

0.36 [0.19, 0.68]

6.3 Not available

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 9. Daclatasvir versus placebo/no intervention
Comparison 10. Simeprevir versus placebo/no intervention

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Hepatitis C‐related morbidity or all‐cause mortality Show forest plot

14

1589

Odds Ratio (M‐H, Fixed, 95% CI)

0.49 [0.08, 2.96]

2 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to dose Show forest plot

14

1589

Odds Ratio (M‐H, Fixed, 95% CI)

0.49 [0.08, 2.96]

2.1 Over or equal to median dose

4

441

Odds Ratio (M‐H, Fixed, 95% CI)

0.51 [0.03, 8.21]

2.2 Under median dose

8

705

Odds Ratio (M‐H, Fixed, 95% CI)

0.41 [0.01, 12.22]

2.3 Not available

2

443

Odds Ratio (M‐H, Fixed, 95% CI)

0.55 [0.02, 13.62]

3 Serious adverse events Show forest plot

18

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

4 Serious adverse events ‐ according to median dose Show forest plot

18

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

4.1 Over or equal to median dose

7

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.2 Under median dose

9

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.3 Not available

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5 Without sustained virological response Show forest plot

19

2898

Odds Ratio (M‐H, Fixed, 95% CI)

0.22 [0.19, 0.27]

6 Without sustained virological response ‐ according to median dose Show forest plot

19

2898

Odds Ratio (M‐H, Fixed, 95% CI)

0.22 [0.19, 0.27]

6.1 Over or equal to median dose

9

1765

Odds Ratio (M‐H, Fixed, 95% CI)

0.25 [0.20, 0.32]

6.2 Under median dose

8

696

Odds Ratio (M‐H, Fixed, 95% CI)

0.19 [0.13, 0.29]

6.3 Not available

2

437

Odds Ratio (M‐H, Fixed, 95% CI)

0.13 [0.07, 0.24]

Figuras y tablas -
Comparison 10. Simeprevir versus placebo/no intervention
Comparison 11. Vaniprevir versus placebo/no intervention

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Hepatitis C‐related morbidity or all‐cause mortality Show forest plot

9

379

Odds Ratio (M‐H, Fixed, 95% CI)

0.76 [0.03, 18.90]

2 Hepatitis C‐related morbidity or all‐cause mortality ‐ according to dose Show forest plot

9

379

Odds Ratio (M‐H, Fixed, 95% CI)

0.76 [0.03, 18.90]

2.1 Over or equal to median dose

6

313

Odds Ratio (M‐H, Fixed, 95% CI)

0.76 [0.03, 18.90]

2.2 Under median dose

3

66

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.3 Not available

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3 Serious adverse events Show forest plot

10

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

4 Serious adverse events ‐ according to median dose Show forest plot

10

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

4.1 Over or equal to median dose

6

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.2 Under median dose

4

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.3 Not available

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5 Without sustained virological response Show forest plot

9

333

Odds Ratio (M‐H, Fixed, 95% CI)

0.12 [0.06, 0.22]

6 Without sustained virological response ‐ according to median dose Show forest plot

9

333

Odds Ratio (M‐H, Fixed, 95% CI)

0.12 [0.06, 0.22]

6.1 Over or equal to median dose

6

280

Odds Ratio (M‐H, Fixed, 95% CI)

0.10 [0.05, 0.20]

6.2 Under median dose

3

53

Odds Ratio (M‐H, Fixed, 95% CI)

0.26 [0.06, 1.04]

6.3 Not available

0

0

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Figuras y tablas -
Comparison 11. Vaniprevir versus placebo/no intervention
Comparison 12. All DAA versus placebo/no intervention

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Without significant reductions in ALT/AST serum levels Show forest plot

11

2099

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.68, 0.92]

2 Without significant reductions in ALT/AST serum levels ‐ according to DAA status Show forest plot

11

2099

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.68, 0.92]

2.1 Trials assessing DAAs on or on the way to the market

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Trials assessing DAAs withdrawn from market

11

2099

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.68, 0.92]

3 Without significant reductions in ALT/AST serum levels ‐ according to type of drug Show forest plot

11

2099

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.68, 0.92]

3.1 Faldaprevir

8

2019

Risk Ratio (M‐H, Random, 95% CI)

0.81 [0.69, 0.96]

3.2 Balaparavir

3

80

Risk Ratio (M‐H, Random, 95% CI)

0.61 [0.41, 0.92]

Figuras y tablas -
Comparison 12. All DAA versus placebo/no intervention